04.04.2013 Views

Transcriptional Characterization of Glioma Neural Stem Cells Diva ...

Transcriptional Characterization of Glioma Neural Stem Cells Diva ...

Transcriptional Characterization of Glioma Neural Stem Cells Diva ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

<strong>Transcriptional</strong> <strong>Characterization</strong><br />

<strong>of</strong><br />

<strong>Glioma</strong> <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong><br />

<strong>Diva</strong> Tommei<br />

Clare Hall College<br />

A dissertation submitted to the University <strong>of</strong> Cambridge<br />

for the degree <strong>of</strong> Doctor <strong>of</strong> Philosophy<br />

European Molecular Biology Laboratory<br />

European Bioinformatics Institute<br />

Wellcome Trust Genome Campus<br />

dt322@cam.ac.uk


A Claudia e Daniela.


This dissertation is my own work and includes nothing which is the outcome<br />

<strong>of</strong> work done in collaboration, except when specified in the text. It is not sub-<br />

stantially the same as any other that I have submitted for a degree, diploma<br />

or other qualification and no part has already been, or is currently being sub-<br />

mitted for any degree, diploma or other qualification. This dissertation does<br />

not exceed the specified length limit <strong>of</strong> 60,000 words as defined by The Biology<br />

Degree Committee. This dissertation has been typeset in 12pt Palatino with<br />

one-and-a-half spacing using L ATEX 2ε according to the specifications defined<br />

by the Board <strong>of</strong> Graduate Studies and the Biology Degree Committee.<br />

<strong>Diva</strong> Tommei<br />

March 25, 2013


Abstract<br />

Tumours affecting the glial portion <strong>of</strong> brain parenchyma are termed gliomas and consti-<br />

tute the most frequent and lethal cancers affecting the central nervous system. Glioblastoma<br />

multiforme is the most aggressive glioma in adults and a World Health Organisation clas-<br />

sified grade IV astrocytoma, characterised by widespread intra-tumoural heterogeneity. A<br />

recent advance in the study <strong>of</strong> gliomas has been the establishment <strong>of</strong> glioma-derived neural<br />

stem (GNS) cell lines that may represent the glioma cell <strong>of</strong> origin. While these cell lines<br />

show many similarities to normal neural stem (NS) cells, an important difference is their<br />

capacity to give rise to authentic glioma-like tumours when xenografted into subventricular<br />

strata <strong>of</strong> immunocompromised mice.<br />

Here I describe an in-depth characterisation <strong>of</strong> the transcriptome <strong>of</strong> GNS cells, to identify<br />

differences in the gene expression between normal and glioma-derived cell lines that may<br />

underlie tumorigenesis. Analyses were carried out at the levels <strong>of</strong> gene expression, molecular<br />

signature pr<strong>of</strong>iling, transcript is<strong>of</strong>orm detection and the quantitation <strong>of</strong> small non-coding<br />

RNAs, taking genetic alterations into account at both the karyotype and mutational level.<br />

Importantly, the cell lines studied were established from tumours with differing histology,<br />

allowing us to sample the breadth <strong>of</strong> the disease rather than focus on the differences between<br />

unhealthy versus healthy counterparts.<br />

We identified a large cohort <strong>of</strong> significantly differentially-expressed genes and a smaller<br />

subset <strong>of</strong> strictly up- and down-regulated ones, including several known glioma oncogenes<br />

as well as novel candidates. An extensive glioblastoma pathway was manually curated<br />

to show the expression <strong>of</strong> our dataset on the known and unknown glioblastoma-affected<br />

pathways. Interestingly, gene set enrichment analysis revealed a consistent up-regulation <strong>of</strong><br />

inflammatory genes in the GNS lines belonging to the MHC class II family, suggesting an<br />

immune-evasion phenotype that has been noted in a number <strong>of</strong> early glioma studies.<br />

<strong>Glioma</strong>s have been classified into a small number <strong>of</strong> subtypes on the basis <strong>of</strong> patient<br />

survival and response to therapy. We found that the expression signatures <strong>of</strong> GNS cell lines<br />

closely resembled the mesenchymal and proneural subtypes, as well as reflecting their known<br />

histopathological features. To characterise genes correlating with patient survival time, we<br />

tested for the association between survival time and gene expression in publicly available<br />

glioma and glioblastoma data sets and found four genes to be strongly positively correlated<br />

with patient survival time and patient age. Together these studies provide an in-depth<br />

analysis <strong>of</strong> a model <strong>of</strong> glioma pathology driven by an aberrant population <strong>of</strong> NS cells.<br />

Finally, a package for the performance evaluation <strong>of</strong> eight leading microRNA target pre-<br />

diction algorithms was built using exon array and microRNA array data from the same GNS<br />

cell lines. This data was used to validate experimentally the target prediction algorithms<br />

that were assessed in their performance as single and combinations <strong>of</strong> them. The combi-<br />

natorial weight analysis allowed us to conclude that (i) tissue specificity bears a non-trivial<br />

weight in predicting what set <strong>of</strong> genes a certain microRNA regulates and, therefore, should<br />

be included in future versions <strong>of</strong> these algorithms, and that (ii) the ElMMO prediction<br />

algorithm fares better than any other combination <strong>of</strong> prediction algorithms.


Acknowledgements<br />

I would like to thank on a pr<strong>of</strong>essional note all the members <strong>of</strong> my lab, starting<br />

from my supervisor Paul Bertone, who has given me the opportunity <strong>of</strong> con-<br />

ducting research at the University <strong>of</strong> Cambridge and has taught me over the<br />

years invaluable life lessons that I will always remember. An incredibly special<br />

thank you goes to Pär Engstrom, who supervised my work throughout my time<br />

as a doctoral student and has always been there for me in any matter scientific.<br />

On a more personal note, I would like to thank my parents. Mamma, grazie<br />

dell’appoggio psicologico, culinario, telefonico, automobilistico e soprattutto<br />

affettivo che mi hai dato negli anni passati lontano da casa. Senza le tue cure<br />

ed il tuo affetto non sarei mai arrivata viva a questo giorno. Babbo, grazie<br />

del DNA che condividiamo, grazie del costante supporto mentale, pecuniario<br />

e filos<strong>of</strong>ico che mi dai sempre incondizionatamente, a mo’ di martello pneu-<br />

matico. Grazie degli stimoli incessanti sui quali posso sempre contare. I also<br />

want to thank Sean Cheng for being there for me since March <strong>of</strong> 2009. Thanks<br />

for these past four years <strong>of</strong> exciting PhD life that you lived with me. Who<br />

knows what’s next for us. Thanks for being a great person, thanks for under-<br />

standing what goes on in my mind, thanks for the love and affection that you<br />

give me every day and thanks for ruining movies by always anticipating what<br />

will happen next. I secretly love that.


Contents<br />

Page<br />

Introduction 1<br />

1 Glioblastoma 2<br />

1.1 Glial <strong>Cells</strong> in the Central Nervous System . . . . . . . . . . . . 2<br />

1.2 Glioblastoma Multiforme . . . . . . . . . . . . . . . . . . . . . . 5<br />

1.3 Primary and Secondary Glioblastomas . . . . . . . . . . . . . . 10<br />

1.4 Pathways Involved in Glioblastoma . . . . . . . . . . . . . . . . 21<br />

1.5 Pathway Crosstalk . . . . . . . . . . . . . . . . . . . . . . . . . 30<br />

2 Neurogenesis 33<br />

2.1 Radial Glia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33<br />

2.2 <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> . . . . . . . . . . . . . . . . . . . . . . . . . . 37<br />

3 Brain Cancer <strong>Stem</strong> <strong>Cells</strong> 57<br />

3.1 The Cancer <strong>Stem</strong> Cell Hypothesis . . . . . . . . . . . . . . . . . 57<br />

3.2 Brain Cancer <strong>Stem</strong> <strong>Cells</strong> . . . . . . . . . . . . . . . . . . . . . . 63<br />

3.3 <strong>Glioma</strong> Culture Systems . . . . . . . . . . . . . . . . . . . . . . 72<br />

4 The Non-Coding RNA World 85<br />

4.1 MicroRNA regulation . . . . . . . . . . . . . . . . . . . . . . . . 86<br />

4.2 Target Prediction and Validation . . . . . . . . . . . . . . . . . 90<br />

Methods 93<br />

5 Methods 94<br />

5.1 Tag-sequencing Data Processing . . . . . . . . . . . . . . . . . . 94<br />

5.2 Array Comparative Genomic Hybridization . . . . . . . . . . . . 98<br />

5.3 Differential Gene Expression . . . . . . . . . . . . . . . . . . . . 100<br />

5.4 Quantitative Real Time-PCR Validation . . . . . . . . . . . . . 101<br />

5.5 Literature Mining . . . . . . . . . . . . . . . . . . . . . . . . . . 107<br />

5.6 Differential Is<strong>of</strong>orm Expression . . . . . . . . . . . . . . . . . . . 110<br />

5.7 Differential Long ncRNA Expression . . . . . . . . . . . . . . . 112<br />

5.8 <strong>Glioma</strong> Expression Signatures . . . . . . . . . . . . . . . . . . . 112<br />

i


5.9 External Dataset Expression Correlation . . . . . . . . . . . . . 112<br />

5.10 Glioblastoma Pathway Construction . . . . . . . . . . . . . . . . 114<br />

5.11 MicroRNA Target Prediction Analysis . . . . . . . . . . . . . . 117<br />

Results 119<br />

6 Digital Transcriptome Pr<strong>of</strong>iling 120<br />

6.1 Clinical Data . . . . . . . . . . . . . . . . . . . . . . . . . . . . 120<br />

6.2 Tag mapping . . . . . . . . . . . . . . . . . . . . . . . . . . . . 121<br />

6.3 Copy Number Aberrations . . . . . . . . . . . . . . . . . . . . . 125<br />

6.4 Core Differentially Expressed Genes . . . . . . . . . . . . . . . . 129<br />

6.5 Large-scale qRT-PCR Validation . . . . . . . . . . . . . . . . . 136<br />

6.6 Literature Mining for Differentially Expressed Genes . . . . . . 142<br />

6.7 Is<strong>of</strong>orm Differential Expression . . . . . . . . . . . . . . . . . . . 144<br />

6.8 Long ncRNA Differential Expression . . . . . . . . . . . . . . . 157<br />

7 Dataset Correlation Analyses 161<br />

7.1 Enrichment Analysis . . . . . . . . . . . . . . . . . . . . . . . . 161<br />

7.2 Glioblastoma Expression Signatures . . . . . . . . . . . . . . . . 168<br />

7.3 Tumour Expression Correlation . . . . . . . . . . . . . . . . . . 170<br />

7.4 Survival Analysis . . . . . . . . . . . . . . . . . . . . . . . . . . 182<br />

7.5 Glioblastoma Pathway Analysis . . . . . . . . . . . . . . . . . . 187<br />

8 MicroRNA Target Prediction Ensemble S<strong>of</strong>tware 202<br />

8.1 Principles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 202<br />

8.2 Workflows . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 204<br />

8.3 Databases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 205<br />

8.4 Filters . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 209<br />

8.5 Target Prediction Ensemble Analysis . . . . . . . . . . . . . . . 211<br />

Conclusions 219<br />

9 Discussion 220<br />

9.1 Digital Pr<strong>of</strong>iling <strong>of</strong> GNS Cell Lines . . . . . . . . . . . . . . . . 220<br />

9.2 MicroRNA Target Prediction Analysis . . . . . . . . . . . . . . 231<br />

9.3 Concluding Remarks . . . . . . . . . . . . . . . . . . . . . . . . 232<br />

Appendix A Differentially Expressed Genes 234<br />

A.1 Differential Expression . . . . . . . . . . . . . . . . . . . . . . . 234<br />

A.2 Classified Differential Expression . . . . . . . . . . . . . . . . . 250<br />

A.3 Quantitative RT-PCR . . . . . . . . . . . . . . . . . . . . . . . 256<br />

A.4 Tag-seq vs qRT-PCR Correlation . . . . . . . . . . . . . . . . . 265<br />

Appendix B Literature Mining Script 266<br />

ii


Appendix C Long ncRNAs 273<br />

Appendix D Glioblastoma Pathway 276<br />

D.1 Pathway Interactions . . . . . . . . . . . . . . . . . . . . . . . . 276<br />

D.2 Pathway Images . . . . . . . . . . . . . . . . . . . . . . . . . . . 282<br />

Appendix E Exon Array Data 286<br />

Appendix F MicroRNA Array Data 300<br />

List <strong>of</strong> Abbreviations 306<br />

List <strong>of</strong> Figures 311<br />

List <strong>of</strong> Tables 314<br />

Bibliography 315<br />

iii


Introduction<br />

1


Chapter 1<br />

Glioblastoma<br />

Contents<br />

1.1 Glial <strong>Cells</strong> in the Central Nervous System . . . . . . . . . . 2<br />

1.2 Glioblastoma Multiforme . . . . . . . . . . . . . . . . . . . 5<br />

1.3 Primary and Secondary Glioblastomas . . . . . . . . . . . . 10<br />

1.4 Pathways Involved in Glioblastoma . . . . . . . . . . . . . 21<br />

1.5 Pathway Crosstalk . . . . . . . . . . . . . . . . . . . . . . . 30<br />

1.1 Glial <strong>Cells</strong> in the Central Nervous System<br />

The Central Nervous System (CNS) is built up <strong>of</strong> neurons and special kinds<br />

<strong>of</strong> supporting cells called glial cells. Neurons are responsible for the functions<br />

that are unique to the nervous system, whereas the glial cells primarily serve<br />

the needs <strong>of</strong> the neurons. The functions <strong>of</strong> the glial cells are still not completely<br />

known. It has been established, however, that they are responsible for isolat-<br />

ing neuronal processes and controlling the environment <strong>of</strong> neurons as well as<br />

taking part in repair processes. Importantly, glial cells are fundamental during<br />

the development <strong>of</strong> the nervous system by providing surfaces and scaffoldings<br />

for migrating neurons and outgrowing axons [10,73]. The accepted hypothesis<br />

that glial cells outnumber the 100 billion neurons present in the human brain<br />

with a ratio <strong>of</strong> up to 50 glial cells per neuron [529] has recently been challenged<br />

and scaled down to a ratio that is closer to one glial cell per neuron [33,73].<br />

Independently <strong>of</strong> the neuronal and glial counts, the importance <strong>of</strong> glial cells<br />

remains unquestioned and their functional roles continue to expand away from<br />

the conservative notion <strong>of</strong> a support structure for neurons [73,173,516]. In fact,<br />

although glial cells do not send precise signals over long distances, they can<br />

produce brief electric currents by opening the membrane channels for Ca 2+<br />

2


1.1 Glial <strong>Cells</strong> in the Central Nervous System Introduction<br />

and producing "calcium signals”. These signals can spread rapidly and thus<br />

influence many neurons almost simultaneously. Also, since neurotransmitter<br />

release depends on extracellular Ca 2+ concentrations, glial cells can contribute<br />

to coordination <strong>of</strong> synaptic activity [10,17,73,516]. Finally, glial activation<br />

and inflammation has been implicated in neurodegenerative diseases such as<br />

Alzheimer’s disease, Parkinson’s disease and multiple sclerosis [269]. Glial cells<br />

are usually divided into three categories: astrocytes, oligodendrocytes and mi-<br />

croglial cells, each carrying functional as well as structural differences. Whilst<br />

astrocytes have many processes <strong>of</strong> various shapes and appear to serve a home-<br />

ostatic function, oligodendrocytes tend to have fewer and shorter processes,<br />

their main function being production <strong>of</strong> myelin sheaths for axon insulation.<br />

Microglial cells are much smaller than other glial cells and serve to the brain<br />

similar functions that macrophages 1 serve to the rest <strong>of</strong> the body [73].<br />

In addition to the three main kinds described above, there are other spe-<br />

cialised forms <strong>of</strong> glial cells. Schwann cells are present only in the peripheral<br />

nervous system and form myelin sheaths that influence axonal thickness, ax-<br />

onal transport and neur<strong>of</strong>ilament content [73]. Ependyma cells are epithelial<br />

cylindrical cells that line the surface <strong>of</strong> the ventricles 2 <strong>of</strong> the CNS and are<br />

responsible for the production, transport and absorption <strong>of</strong> cerebrospinal fluid<br />

(CSF). These cells have recently become candidates for the location <strong>of</strong> the<br />

neural stem cell niche [81,328]. Müller cells are glial cells that reside in the<br />

vertebrate retina and have recently been observed to undergo dedifferentiation<br />

in vitro into multi-potent progenitor cells in fish, chicken and mouse, to then<br />

differentiate into a number <strong>of</strong> retinal cell types, although in vivo evidence is<br />

not conclusive [144]. They have also been shown to act as a light collector in<br />

the mammalian eye [53,298]. Bergman cells are astrocytes that reside in the<br />

cerebellum and are responsible for the migration, dendritogenesis, synaptoge-<br />

nesis and maturation <strong>of</strong> Purkinje neurons 3 [534]. Finally, pituicytes are glial<br />

cells that reside in the posterior <strong>of</strong> the pituitary gland, where they participate<br />

in the control <strong>of</strong> secretory events [434].<br />

1 Macrophages are white blood cells that protect the body from harmful bacteria, particles<br />

and dying cells by ingesting them in a process called phagocytosis.<br />

2 The ventricular system <strong>of</strong> the brain is composed <strong>of</strong> four communicating ventricles filled<br />

with cerebrospinal fluid that bathes and cushions the brain and communicates with the<br />

central canal <strong>of</strong> the spinal chord.<br />

3 Purkinje cells are very large neurons in the cerebellum that release the inhibitory neurotransmitter<br />

gamma-aminobutyric acid (GABA) and are responsible for transmission <strong>of</strong> all<br />

motor coordination signals.<br />

3


1.1 Glial <strong>Cells</strong> in the Central Nervous System Introduction<br />

The numerous short and long processes <strong>of</strong> astrocytes amount to a very large<br />

exposed surface that makes these glial cells well suited for efficient exchange<br />

<strong>of</strong> molecules and ions. This fact, put together with the intimate contacts that<br />

astrocytes establish with neurons, capillaries and the CSF, puts them in the<br />

unique position to control the environment in which neurons function [394].<br />

Neuronal homeostasis is crucial due to the extreme sensitivity <strong>of</strong> neurons to<br />

changes in concentrations <strong>of</strong> ions and neurotransmitters and the fact that these<br />

changes can become significant at very low additional amounts <strong>of</strong> substance<br />

given the very limited extracellular space 4 . Extracellular neurotransmitter<br />

concentration must be kept very low at all times in order for the synaptic re-<br />

lease to generate a significant change in neurotransmitter concentration [73].<br />

Astrocytes also contribute to extracellular pH by removing CO2 [136] and<br />

help control extracellular osmotic pressure by maintaining the water balance<br />

<strong>of</strong> the brain [20]. Mechanisms for the control <strong>of</strong> extracellular osmolarity may<br />

involve exchange <strong>of</strong> small neutral molecules like the amino acid taurin [384]<br />

and special channels for transport <strong>of</strong> water called aquaporins that are present<br />

on the membrane <strong>of</strong> astrocytic processes in contact with capillaries [540,545].<br />

By surrounding capillaries with their processes and forming very extensive<br />

tight junctions 5 between the endothelial cells, astrocytes decrease the perme-<br />

ability <strong>of</strong> brain capillaries and help establish a fully functional blood-brain<br />

barrier 6 [172,279,438].<br />

Since their discovery, oligodendrocytes have been classified in many different<br />

ways due to their inherent morphological heterogeneity including variable num-<br />

ber <strong>of</strong> processes, thickness <strong>of</strong> myelin sheath, density <strong>of</strong> cytoplasm and clumping<br />

<strong>of</strong> nuclear chromatin [44]. The function <strong>of</strong> most oligodendrocytes is to pro-<br />

duce myelin sheaths to wrap around axons in order to isolate and quicken<br />

transportation <strong>of</strong> the nerve impulse [73]. This contributes to the much higher<br />

conduction speeds observed in myelinated axons versus unmyelinated ones,<br />

with the thickest myelinated axons conducting at about 120 meters/sec and<br />

the slowest unmyelinated ones at 1 meter/sec [44,73]. A myelin sheath consists<br />

almost exclusively <strong>of</strong> numerous layers <strong>of</strong> oligodendrocyte or Schwann cell mem-<br />

brane (lamellae) that in the process <strong>of</strong> wrapping around the axon squeeze away<br />

4 Less than 20% <strong>of</strong> the total brain volume.<br />

5 Type <strong>of</strong> junctional complexes present only in vertebrates that seal together the membranes<br />

<strong>of</strong> two cells, effectively preventing passage <strong>of</strong> water and ions.<br />

6 Unlike most organs in our body, the composition <strong>of</strong> the extracellular fluid <strong>of</strong> the brain<br />

differs from that in the blood plasma.<br />

4


1.2 Glioblastoma Multiforme Introduction<br />

the cytoplasm, thus allowing the membrane layers to lie closely apposed [73].<br />

Although the structure and function <strong>of</strong> the myelin sheath produced by oligo-<br />

dendrocytes and Schwann cells is identical, one oligodendrocyte can extend its<br />

processes to 50 axons, whilst each Schwann cell can only wrap its processes<br />

around one axon [44,413]. Recent observations have suggested that oligoden-<br />

drocytes and Schwann cells are also responsible for the long-term functional<br />

integrity <strong>of</strong> axons [356]. Furthermore, there exists a pool <strong>of</strong> oligodendrocytes<br />

named "satellite" that are found next to neuronal cell bodies where they can-<br />

not serve an isolating role, but rather it is suggested they regulate neuronal<br />

homeostasis in much a similar way as do astrocytes [44].<br />

1.2 Glioblastoma multiforme 7<br />

Primary brain tumours comprise a wide range <strong>of</strong> histological and pathologi-<br />

cal entities, each with a distinct natural history [77,128]. For simplicity, CNS<br />

tumours are classified as gliomas or non-gliomas. <strong>Glioma</strong>s affect the glial por-<br />

tion <strong>of</strong> brain parenchyma and they are the most frequent and lethal cancers<br />

affecting the CNS [383]. Diagnosis <strong>of</strong> gliomas is heavily based on the predom-<br />

inantly affected cell type, which in turn is indicative <strong>of</strong> prognosis [128] (Table<br />

1.1). <strong>Glioma</strong>s <strong>of</strong> astrocytic, oligodendroglial, oligoastrocytic 8 and ependymal<br />

origin account for more than 70% <strong>of</strong> all brain tumours [383], with glioblas-<br />

toma multiforme (GBM) being the most frequent (65%) and malignant <strong>of</strong> all<br />

adult gliomas [154,300,365]. Grade I astrocytomas are well circumscribed and<br />

curative surgical resections may be attempted. However, most gliomas are<br />

characterised by diffuse infiltration <strong>of</strong> brain parenchyma, making surgical ex-<br />

tirpation impossible [77]. Some lower-grade astrocytic tumours change their<br />

identity over time and turn into more aggressive forms. Depending on the his-<br />

tology <strong>of</strong> the tumour, patients undergo surgery, chemotherapy, radiotherapy<br />

or a combination <strong>of</strong> these treatments, although the chances <strong>of</strong> cure are very<br />

low [128].<br />

In 1979 the World Health Organisation (WHO) published the first edition<br />

<strong>of</strong> Histological Typing <strong>of</strong> Tumours <strong>of</strong> the Central Nervous System with the<br />

7 The term glioblastoma is used synonymously with "glioblastoma multiforme".<br />

8 Oligoastrocytic tumours are composed <strong>of</strong> roughly equivalent amounts <strong>of</strong> astrocyte-like<br />

and oligodendrocyte-like cells.<br />

5


1.2 Glioblastoma Multiforme Introduction<br />

Table 1.1: Histological Types and Prognosis <strong>of</strong> <strong>Glioma</strong>s (y, years). Taken from<br />

Doyle et al 2005 [128].<br />

Tumour Cell Type Tumour WHO<br />

grade<br />

Incidence<br />

Relative Survival<br />

Rates (%)<br />

1y 2y 3y<br />

Astrocytic tumours Pilocytic astrocytoma l 10% <strong>of</strong> cerebral astrocytomas<br />

most common<br />

in children<br />

95.7 94.3 91.3<br />

Astrocytic tumours Diffuse astrocytoma II 10-15% <strong>of</strong> astrocytic<br />

tumours<br />

73.9 61.8 46.9<br />

Astrocytic tumours Anaplastic astrocytoma III - 60.3 44.0 29.4<br />

Astrocytic tumours Glioblastoma multiforme IV 50-60% <strong>of</strong> astrocy- 29.3 8.7 3.3<br />

tomas; 12-15% <strong>of</strong> all<br />

Oligodendroglial<br />

tumours<br />

Oligodendroglioma II<br />

intracranial neoplasms<br />

- 89.7 83.4 70.5<br />

Ependymal<br />

tumours<br />

Ependymoma or<br />

Anaplastic ependymoma<br />

II-III - 87.6 81.4 70.6<br />

aim <strong>of</strong> establishing a comprehensive four-tiered malignancy 9 grading guideline<br />

for the evaluation <strong>of</strong> brain tumour progression. The WHO system is based on<br />

the appearance <strong>of</strong> specific characteristics, such as atypia 10 , mitosis, endothelial<br />

proliferation and necrosis. By reflecting the malignant potential <strong>of</strong> the tumour,<br />

these features help assess the choice <strong>of</strong> therapies [301]. The four grades are<br />

detailed as follows:<br />

· Grade I lesions are considered low grade and include lesions with low<br />

proliferative potential that are <strong>of</strong>ten cured through surgical resection<br />

alone;<br />

· Grade II lesions are also considered low grade, but they are infiltrative<br />

in nature and <strong>of</strong>ten recur despite surgical resection;<br />

· Grade III lesions carry histological evidence <strong>of</strong> malignancy, such as<br />

atypia and fast mitotic activity, and are considered intermediate to high<br />

grade lesions;<br />

· Grade IV lesions are considered high grade and are cytologically ma-<br />

lignant, mitotically active, necrosis-prone neoplasms <strong>of</strong>ten associated<br />

with rapid pre- and post-operative disease evolution and a fatal out-<br />

come [154,300,301].<br />

The WHO classifies glioblastoma as a grade IV astrocytoma, accounting for<br />

approximately 12-15% <strong>of</strong> all intracranial neoplasms and 60-75% <strong>of</strong> astrocytic<br />

9 Medical term used to describe the state <strong>of</strong> tumours that are resistant to therapy, spread<br />

rapidly and have a destructive clinical course.<br />

10 Term that indicates a general cellular abnormality.<br />

6


1.2 Glioblastoma Multiforme Introduction<br />

tumours [154,301,383]. As the moniker "multiforme" implies, GBM is charac-<br />

terised by a widespread intratumoural heterogeneity that makes it extremely<br />

hard to understand and treat [154,383]. The histopathological features <strong>of</strong> GBM<br />

include nuclear atypia, cellular pleomorphism, mitotic activity, vascular throm-<br />

bosis, microvascular proliferation and necrosis [154,301]. This complexity, com-<br />

bined with a putative cancer stem cell subpopulation and an incomplete atlas<br />

<strong>of</strong> epigenetic and genetic lesions, has contributed to make this cancer one <strong>of</strong><br />

the most difficult to understand and to treat [154].<br />

The average life expectancy <strong>of</strong> a patient with glioblastoma lies between several<br />

weeks and several months after postoperative radiotherapy, with a protracted<br />

course when treated with temozolomide in addition to radiotherapy alone (Fig<br />

1.1). Median survival is generally less than one year from the time <strong>of</strong> diag-<br />

nosis and most patients die within two years, with most long-term survivors<br />

being given the wrong histological diagnosis at first [128,472]. Unless the<br />

neoplasm has developed from a lower grade astrocytoma, in more than 50%<br />

<strong>of</strong> cases the clinical history is less than 3 months. In most European and<br />

North American countries, the incidence lies in the range <strong>of</strong> 3-4 new cases per<br />

100,000 population per year, preferentially affecting adults and with a slightly<br />

higher incidence in men than women [128,301]. Although infiltrative spread<br />

Figure 1.1: Estimates <strong>of</strong> survival amongst GBM patients treated with radiotherapy<br />

alone or radiotherapy with the alkylating agent temozolomide. Taken from Stupp et<br />

al 2005 [472].<br />

is a common feature <strong>of</strong> all diffuse astrocytic tumours, glioblastoma is particu-<br />

larly notorious for its rapid invasion <strong>of</strong> the neighboring brain structures [154].<br />

7


1.2 Glioblastoma Multiforme Introduction<br />

Invading cells reside outside the contrast-enhancing rim <strong>of</strong> the tumour thereby<br />

escaping surgical resection and evading radiotherapy. However, the generation<br />

<strong>of</strong> metastases outside <strong>of</strong> the CNS remains very rare because the subarach-<br />

noidal space 11 and CSF tend to remain unaffected [301]. The transforming<br />

growth factor β (TGFβ) and Akt signaling pathways have been reported to<br />

act as molecular mediators for glioblastoma invasion [245,527], as well as the<br />

possibility <strong>of</strong> activation through hypoxia with the hypoxia-inducible factors<br />

HIF1 12 [219]. An important aspect <strong>of</strong> glioblastoma invasion is the production<br />

<strong>of</strong> a thick extracellular matrix to support migration and that <strong>of</strong> proteolytic<br />

enzymes to enhance invasion across this matrix [301]. The current standard<br />

for glioma patients involves resection <strong>of</strong> the tumour followed by extensive radi-<br />

ation therapy and chemotherapy with the temozolomide alkylating agent - or<br />

carmustine, a nitrosourea drug, in the United States - that grants the patient<br />

a median survival <strong>of</strong> 15 months [472].<br />

Methods and Technologies in Cancer Genomics The field <strong>of</strong> cancer<br />

genomics has been evolving over the past decade at a very fast pace, bring-<br />

ing in gargantuan amounts <strong>of</strong> fresh data to analyse from increasingly more<br />

efficient technology platforms. A brief overview <strong>of</strong> the methods and technolo-<br />

gies referred to in the rest <strong>of</strong> this chapter is given below. It should be noted<br />

that this paragraph is by no means a comprehensive representation <strong>of</strong> all the<br />

technologies currently deployed in the field <strong>of</strong> cancer genomics.<br />

· Expression pr<strong>of</strong>iling determines the expression level <strong>of</strong> transcripts within<br />

a cell using platforms that can be distinguished between array and non-<br />

array. In the case <strong>of</strong> microarrays, the expression level is measured through<br />

probe-transcript interaction for those transcripts that are represented on<br />

the array. An a priori knowledge <strong>of</strong> what sequences to measure is nec-<br />

essary in terms <strong>of</strong> the type <strong>of</strong> transcript (i.e. microRNAs, mRNAs), the<br />

specific transcripts <strong>of</strong> interest for the correct probe design, and the re-<br />

gions within that transcript (i.e. exons, introns, UTRs, single nucleotide<br />

polymorphisms (SNPs), etc), making this platform inadequate for novel<br />

gene discovery. Unlike microarrays, RNA sequencing measures expres-<br />

sion levels by averaging the number <strong>of</strong> sequenced "reads", fragments <strong>of</strong><br />

11Interval between two membranes that protect the brain, the arachnoid membrane and<br />

the pia mater.<br />

12Transcription factors that respond to decreased availability <strong>of</strong> oxygen in the cellular<br />

environment and are highly conserved transcriptional complexes <strong>of</strong> heterodimers constituted<br />

by an α and a β subunit.<br />

8


1.2 Glioblastoma Multiforme Introduction<br />

up to several hundred base pairs, depending on the technology used, col-<br />

lected along the entire length <strong>of</strong> the transcript. Potentially any RNA<br />

population can be selected for sequencing but the mRNA and small non-<br />

coding RNA population are the most commonly measured. Due to its<br />

probe-free nature, sequencing is an appropriate platform for novel gene<br />

discovery.<br />

· Epigenetic pr<strong>of</strong>iling measures the amount <strong>of</strong> DNA methylation that oc-<br />

curs at CpG islands in promoters using different types <strong>of</strong> assays on array<br />

and non-array platforms. Many high-throughput pr<strong>of</strong>iling assays have<br />

genomic DNA treated with a bisulfite conversion kit that converts un-<br />

methylated cytosine residues to uracil and leaves methylated cytosine<br />

residues unaffected. This treatment yields single nucleotide resolution<br />

information about the methylation status <strong>of</strong> a segment <strong>of</strong> DNA and var-<br />

ious analyses can be performed that depend on the platform used, to<br />

retrieve this information.<br />

· Exome sequencing is a selective method <strong>of</strong> DNA and RNA sequencing in<br />

which samples are enriched for the coding regions <strong>of</strong> the genome, or the<br />

"exome", and are successively sequenced. Targeted enrichment <strong>of</strong> the<br />

exome may be performed by capture, using hybridization to microarrays<br />

with probe sequences defined, for example, by the National Centre for<br />

Biotechnology Information (NCBI) Consensus Coding Sequence (CCDS)<br />

database [358], or by amplicon-based PCR amplification employing se-<br />

quencing adaptors to amplify specific loci. As coding regions constitute<br />

approximately 1% <strong>of</strong> the human genome [98], exome sequencing is a<br />

potentially efficient strategy for the identification <strong>of</strong> rare functional mu-<br />

tations, thereby detaining clinical relevance in the assessment <strong>of</strong> the role<br />

<strong>of</strong> sequence variation in genetic disorders [56,98,358]. Although meth-<br />

ods <strong>of</strong> enriching targeted genomic segments by hybridization have been<br />

historically limited by the large amounts <strong>of</strong> genomic DNA required and<br />

the modest throughput <strong>of</strong> the coupled sequence platforms, nowadays ad-<br />

vances in hybridization specificity and sequencing technology have suc-<br />

cessfully reduced the costs and increased the coverage <strong>of</strong> the exome se-<br />

quencing process. However, cost effectiveness and completeness <strong>of</strong> the<br />

information obtained are still key considerations [98].<br />

· Array comparative genomic hybridization is a method for the detection<br />

<strong>of</strong> copy number aberrations that uses an array platform containing thou-<br />

9


1.3 Primary and Secondary Glioblastomas Introduction<br />

sands <strong>of</strong> defined DNA probes that are hybridised to a sample mixture<br />

containing DNA from tumour cells and DNA from a normal control, each<br />

labeled with a different fluorescent dye. Abnormal regions in the genome<br />

are then detected by calculating the ratio <strong>of</strong> the fluorescence intensity <strong>of</strong><br />

the hybridised sample to that <strong>of</strong> the reference DNA.<br />

1.3 Primary and Secondary Glioblastomas<br />

High-grade malignant gliomas are uniformly fatal despite the therapeutic ag-<br />

gressiveness with which they are treated. As already mentioned, a classical<br />

feature <strong>of</strong> these tumours is their widespread morphological and lineage het-<br />

erogeneity. This plasticity is especially appreciable in gliomas presenting both<br />

astrocytic and oligodendroglial histopathological features, the basis <strong>of</strong> which,<br />

however, remains unknown [549].<br />

Glioblastomas have been classified into two subtypes, primary or de novo<br />

and secondary glioblastomas [154,301,383,549]. Primary glioblastomas rep-<br />

resent the majority (more than 90%) <strong>of</strong> diagnosed cases and they develop<br />

very rapidly without clinical or histopathological evidence <strong>of</strong> a pre-existing,<br />

less malignant precursor lesion [154,301,334,450]. Secondary glioblastomas, on<br />

the other hand, have a history <strong>of</strong> malignant progression from a lower-grade tu-<br />

mour such as diffuse astrocytoma (WHO grade II) or anaplastic 13 astrocytoma<br />

(WHO grade III), with more than 70% <strong>of</strong> WHO grade II gliomas transform-<br />

ing into WHO grade III/IV diseases within five to 10 years <strong>of</strong> diagnosis [154].<br />

Only 5% <strong>of</strong> glioblastomas are classified as secondary and they tend to pertain<br />

to a younger cohort <strong>of</strong> patients (average age 45 years) compared to primary<br />

glioblastoma patients (average age 60 years) [154,301,383]. Interestingly, de-<br />

spite their distinct clinical histories, primary and secondary glioblastomas are<br />

morphologically and clinically indistinguishable and their prognoses are equally<br />

poor after having performed age-adjusted analysis [154,383,390].<br />

Presently, the tools used to study glioblastoma are: primary tissue, genetically<br />

modified mouse models (GEMMs), and glioma cell lines [308].<br />

Primary Tissue The genetic events involved in the initiation and progres-<br />

sion <strong>of</strong> glioblastoma are still unknown because <strong>of</strong> the limited availability <strong>of</strong><br />

early stage neoplastic tissue [308]. A number <strong>of</strong> studies today have focused<br />

on large-scale sequencing <strong>of</strong> glioblastoma samples from different patients to<br />

13 A cancer that is very poorly differentiated is called anaplastic.<br />

10


1.3 Primary and Secondary Glioblastomas Introduction<br />

attempt focusing on this problem. In the Parsons et al study from 2008 [383],<br />

mutational data obtained from the exome sequencing <strong>of</strong> 22 human glioblas-<br />

tomas was analysed for copy number aberrations (CNAs), and integrated to<br />

identify glioblastoma candidate driver genes, i.e. genes that carry mutations<br />

providing a selective advantage to the tumour cell. Interestingly, in 12% <strong>of</strong> the<br />

glioblastoma patients mutations were found in the active site <strong>of</strong> the Isocitrate<br />

dehydrogenase 1 (IDH1) gene on the long arm <strong>of</strong> chromosome 22, a gene never<br />

previously associated with glioblastoma. IDH1 encodes an isocitrate dehydro-<br />

genase, which catalyses the carboxylation <strong>of</strong> isocitrate to α-ketoglutarate and<br />

nicotinamide adenine dinucleotide phosphate (NADPH), a coenzyme used as a<br />

reducing agent in anabolic 14 biosynthetic reactions [225]. Five isocitrate dehy-<br />

drogenase genes exist in humans and three are localised in the mitochondria,<br />

while IDH1 is localised within the cytoplasm and peroxisomes. The func-<br />

tion <strong>of</strong> IDH1 is to help release cellular stress from oxidative damage through<br />

the generation <strong>of</strong> NADPH. Mutations in IDH1 were observed preferentially<br />

in younger glioblastoma patients, on average 33 years <strong>of</strong> age, as opposed to<br />

wild type carriers, on average 53 years <strong>of</strong> age, and most <strong>of</strong> them were found<br />

in patients with secondary glioblastomas. These patients had a longer median<br />

survival time <strong>of</strong> 3.8 years as compared to 1.1 years for patients with wild-type<br />

IDH1. A similar pattern was also observed in the subgroup <strong>of</strong> young patients<br />

with Tumour protein 53 (TP53) mutations. All mutations <strong>of</strong> IDH1 resulted<br />

in an amino acid activating substitution at an evolutionary conserved residue<br />

located within the enzyme’s binding site, reminiscent <strong>of</strong> known activating al-<br />

terations in oncogenes such as BRAF, KRAS and PIK3CA [383].<br />

The study by Watanabe et al [522] carried these results further by finding a<br />

total <strong>of</strong> 130 IDH1 mutations involving amino acid 132 in 321 gliomas that<br />

specifically affected 88% <strong>of</strong> the low-grade diffuse astrocytomas, 82% <strong>of</strong> the<br />

secondary glioblastomas that developed through progression from low-grade<br />

diffuse or anaplastic astrocytoma, 79% <strong>of</strong> the oligodendrogliomas and 94% <strong>of</strong><br />

the oligoastrocytomas. Interestingly, analyses <strong>of</strong> multiple biopsies from the<br />

same patient showed that an IDH1 mutation never occurred after the acquisi-<br />

tion <strong>of</strong> a TP53 mutation, suggesting that IDH1 mutation is a very early event<br />

in gliomagenesis that may affect a common glial precursor cell population.<br />

IDH1 mutations were co-present with TP53 mutations in 63% <strong>of</strong> low-grade<br />

14 An anabolic reaction, as opposed to a catabolic one, is a metabolic pathway that constructs<br />

molecules from smaller units and requires energy.<br />

11


1.3 Primary and Secondary Glioblastomas Introduction<br />

diffuse astrocytomas, but only 10% <strong>of</strong> pilocytic astrocytomas, 5% <strong>of</strong> primary<br />

glioblastomas and none <strong>of</strong> the ependymomas. The frequent presence <strong>of</strong> IDH1<br />

mutations in secondary glioblastomas and their near complete absence in pri-<br />

mary glioblastomas reinforces the concept that, despite their histological sim-<br />

ilarities, these subtypes are genetically and clinically distinct entities [522].<br />

In an even larger study by Yan et al [535], the sequences <strong>of</strong> the IDH1 and<br />

closely related IDH2 genes were determined in 445 tumours <strong>of</strong> the CNS and<br />

494 tumours that did not affect the CNS. In corroboration <strong>of</strong> the Parsons et<br />

al [383] and Watanabe et al [522] studies, mutations in the IDH1 gene were<br />

found in more than 70% <strong>of</strong> WHO grade II and III astrocytomas and oligoden-<br />

drogliomas, as well as in the glioblastomas that developed from these lower-<br />

grade lesions. Each <strong>of</strong> these mutations affected amino acid 132 and reduced<br />

the enzymatic activity <strong>of</strong> the encoded protein [535]. Interestingly, tumours<br />

that did not carry mutations in the IDH1 gene <strong>of</strong>ten had mutations affect-<br />

ing the analogous amino acid (R172) on the closely related IDH2 gene that<br />

also reduced the enzymatic activity <strong>of</strong> the encoded protein, suggesting a form<br />

<strong>of</strong> functional redundancy between the two genes. Similarly to the results by<br />

Parsons et al [383], the tumours carrying IDH1 or IDH2 mutations showed dis-<br />

tinctive genetic and clinical characteristics that resulted in better outcomes for<br />

those patients with respect to the patients carrying wild-type IDH genes [535].<br />

In a study by Zhao et al [548] the functional impact <strong>of</strong> the IDH1 mutation<br />

was assessed in cultured glioma cells. By using the human cytosolic IDH1<br />

crystal structure reported by Xu et al in 2004 [532], this study showed that<br />

the tumour-derived IDH1 mutation impaired the affinity <strong>of</strong> the enzyme for its<br />

substrate by forming catalytically inactive heterodimers. Interestingly, when<br />

the expression <strong>of</strong> a mutant IDH1 was forced in cultured glioma cells, the for-<br />

mation <strong>of</strong> α-ketoglutarate was greatly reduced but levels <strong>of</strong> the subunit α <strong>of</strong><br />

hypoxia-inducible factor 1 (HIF1A) were greatly increased. In fact, the tran-<br />

scription factor HIF1A is regulated by the product <strong>of</strong> the reaction catalysed by<br />

IDH1, α-ketoglutarate, and as a result, the IDH1 mutated human glioma cul-<br />

tures displayed higher levels <strong>of</strong> HIF1A unlike wild-type IDH1 glioma cultures.<br />

This was indicative that IDH1 may function as a tumour suppressor and, when<br />

inactivated by mutation, may contribute to tumourigenesis through induction<br />

<strong>of</strong> the HIF1 pathway [548]. In summary, the possibility <strong>of</strong> detecting an IDH1<br />

mutation in a patient has the clinical potential, for a subpopulation <strong>of</strong> mostly<br />

12


1.3 Primary and Secondary Glioblastomas Introduction<br />

secondary and few primary glioblastoma patients, to hypothesise a protracted<br />

clinical course [383]. New treatments could be designed to take advantage<br />

<strong>of</strong> IDH1 alterations in these patients, especially since the inhibition <strong>of</strong> the<br />

IDH2 enzyme has recently been shown to increase sensitivity <strong>of</strong> tumour cells<br />

to chemotherapeutic agents [225].<br />

The glioblastoma cancer genome was the first to be characterised in the con-<br />

certed efforts <strong>of</strong> the Cancer Genome Atlas project (TCGA) [326]. The aim<br />

was to "catalogue and discover major cancer-causing genome alterations in<br />

large cohorts <strong>of</strong> human tumours through integrated multi-dimensional analy-<br />

ses". The pilot project screened a total <strong>of</strong> 587 samples down to 206, which<br />

were used to conduct genome-wide analysis <strong>of</strong> DNA copy number and gene<br />

expression, and DNA methylation screening on a total <strong>of</strong> 2,305 assayed genes.<br />

Of the 206 chosen biospecimens, 21 were post-treatment glioblastoma cases<br />

and the remaining 185 represented predominantly primary glioblastomas. A<br />

total <strong>of</strong> 91 samples and 601 genes, inclusive <strong>of</strong> 7932 exons, were chosen from<br />

the 206 sample pool for re-sequencing towards mutational analysis. Although<br />

the method <strong>of</strong> biospecimen selection ensured high-quality data, the stringency<br />

<strong>of</strong> selection criteria may have introduced a degree <strong>of</strong> bias since small samples<br />

and samples with high levels <strong>of</strong> necrosis were excluded [326].<br />

In the TCGA study, upon a statistical analysis <strong>of</strong> mutation significance in the<br />

91 matched glioblastoma-normal pairs selected for detection <strong>of</strong> somatic muta-<br />

tions in 601 selected genes, eight genes were found to be significantly mutated:<br />

TP53, PTEN, NF1, EGFR, ERBB2, RB1, PIK3R1 and PIK3CA. All the mu-<br />

tations involving TP53 were clustered in its DNA-binding domain, a known<br />

hotspot for TP53 mutations in human cancers. TP53 is an important tran-<br />

scription factor and tumour suppressor involved in most cell survival pathways<br />

and for this reason <strong>of</strong>ten referred to as "the guardian <strong>of</strong> the genome" [133].<br />

Given that 27 <strong>of</strong> the 72 untreated samples and 11 <strong>of</strong> the 19 treated samples<br />

harboured TP53 mutations and given that most <strong>of</strong> the 91 samples were pri-<br />

mary glioblastomas, one can conclude that TP53 mutation is a common event<br />

in primary glioblastoma [326]. Neur<strong>of</strong>ibromin 1 (NF1) is a tumour suppressor<br />

that when mutated in humans is responsible for the development <strong>of</strong> neur<strong>of</strong>i-<br />

bromatosis type I and is associated with increased risk <strong>of</strong> optic gliomas, as-<br />

trocytomas and glioblastomas [35]. The protein encoded by NF1 is a negative<br />

regulator <strong>of</strong> the rat sarcoma (RAS) signaling pathway <strong>of</strong> small guanosine-5’-<br />

13


1.3 Primary and Secondary Glioblastomas Introduction<br />

triphosphate hydrolases (GTPase) that activate the mitogen activated protein<br />

kinase (MAPK) signaling cascade amongst other pathways [496]. Overall, at<br />

least 47 <strong>of</strong> the 206 samples harboured somatic NF1 inactivating mutations or<br />

deletions, confirming the relevance <strong>of</strong> this gene in sporadic human glioblas-<br />

toma [326].<br />

Furthermore, within the TCGA dataset it was observed that mutation rates<br />

between untreated and treated glioblastomas were markedly different, aver-<br />

aging at 1.4 and 5.8 somatic silent mutations per sample, respectively. The<br />

higher average in the treated samples was mostly due to the contributions<br />

from a cohort <strong>of</strong> seven hypermutated tumours treated with temozolomide or<br />

lomustine. The hypermutator phenotypes previously described in glioblas-<br />

toma [79,204] were known to carry mutations in MutS homolog 6 (MSH6),<br />

a component <strong>of</strong> the post-replicative DNA mismatch repair system (MMR).<br />

MSH6 heterodimerizes with MSH2 to form MutSα, which binds to DNA mis-<br />

matches thereby initiating DNA repair [6]. An analysis <strong>of</strong> the genes involved<br />

in mismatch repair within the TCGA dataset uncovered that six out <strong>of</strong> the<br />

seven hypermutated samples harbored mutations in at least one <strong>of</strong> the mis-<br />

match repair genes MLH1, MSH2, MSH6 or PMS2 [326].<br />

Recurrent focal alterations found in the TCGA samples that have already<br />

been described and are common in glioblastoma are the amplification <strong>of</strong> Epi-<br />

dermal growth factor receptor (EGFR), cyclin-dependent-kinase (CDK) CDK4<br />

and CDK6, PDGFRA, MDM2, MDM4, MET, MYCN, CCND2 and PIK2CA<br />

[104,255,262,292,374,423,429]. Interestingly, uncommon focal alterations were<br />

also found, such as the amplification <strong>of</strong> the serine/threonine protein kinase<br />

AKT3 and the homozygous deletions <strong>of</strong> NF1 and PARK2. V-akt murine thy-<br />

moma viral oncogene homolog 3 (AKT3) belongs to the AKT family <strong>of</strong> ser-<br />

ine/threonine kinases together with AKT1 and AKT2. The three AKT kinases<br />

are now known to represent central nodes in a variety <strong>of</strong> signaling cascades that<br />

regulate normal cellular process such as cell size and growth, proliferation,<br />

survival, glucose metabolism, genome stability, and neo-vascularization. It is<br />

currently less clear, however, whether AKT1, AKT2, and AKT3 are function-<br />

ally redundant or whether each carries out a specific functional role [45,46].<br />

Parkinson protein 2 (PARK2) encodes for a component <strong>of</strong> an E3 ubiquitin<br />

ligase complex that mediates the targeting <strong>of</strong> substrate proteins for protea-<br />

somal degradation. The functions carried by PARK2 are currently unknown,<br />

14


1.3 Primary and Secondary Glioblastomas Introduction<br />

but mutations in this gene are known to cause a familial form <strong>of</strong> Parkinson’s<br />

disease known as autosomal recessive juvenile Parkinson disease [229]. The<br />

most significant loss-<strong>of</strong>-heterozygosity (LOH 15 ) event identified in the TCGA<br />

dataset was observed on the long arm <strong>of</strong> chromosome 17 where the TP53 gene<br />

resides [326].<br />

Cytosine-phosphate-Guanine (CpG 16 ) islands are regions <strong>of</strong> DNA in which a<br />

cytosine is linked to a guanine via a phopsphodiester bond to form the dinu-<br />

cleotide CpG that is repeated many times along a linear sequence. In formal<br />

definitions, a CpG island must occupy more than 50% <strong>of</strong> a 200bp sequence<br />

and have an expected/observed CpG ratio <strong>of</strong> 0.6 [102,159]. Methylation <strong>of</strong> the<br />

5’ carbon <strong>of</strong> cytosine is a form <strong>of</strong> epigenetic modification that affects regula-<br />

tion <strong>of</strong> gene expression via non-sequence based interactions. Such methylated<br />

cytosines are present in the coding regions <strong>of</strong> mammalian genes and, over evo-<br />

lutionary time, spontaneously deaminate to become thymines [102,287]. In<br />

mammals, 70% to 80% <strong>of</strong> CpG cytosines are methylated [208]. Oppositely,<br />

CpG islands in promoters tend to be unmethylated when genes are expressed,<br />

suggesting that methylation is an inhibiting event for gene expression [142,214].<br />

Methylation <strong>of</strong> CpG sites within promoters has been observed as a mechanism<br />

<strong>of</strong> tumour suppressor gene silencing in a number <strong>of</strong> human cancers. In contrast,<br />

the hypomethylation <strong>of</strong> CpG sites has been associated with the over-expression<br />

<strong>of</strong> oncogenes within cancer cells [214].<br />

Evaluation <strong>of</strong> promoter methylation was conducted in the TCGA project across<br />

91 glioblastoma samples. A pattern emerged between the methylation <strong>of</strong> the<br />

O-6-methylguanine-DNA methyltransferase (MGMT) promoter and the sub-<br />

stitution spectrum <strong>of</strong> treated samples. MGMT is a DNA repair enzyme that<br />

repairs damaged alkylated guanine residues and its promoter methylation sta-<br />

tus has already been associated to sensitivity to the temozolomide alkylating<br />

agent, which is the current standard <strong>of</strong> care for glioblastoma patients [79,204].<br />

Amongst the 13 samples treated with alkylating agent that did not show<br />

MGMT methylation, the validated somatic mutations from GC to AT, caused<br />

by the spontaneous deamination <strong>of</strong> methylated cytosines to thymines, occurred<br />

in comparable amounts between CpG and non-CpG dinucleotides. However,<br />

in the six treated samples with MGMT methylation, the GC to AT transitions<br />

15 The loss <strong>of</strong> normal function <strong>of</strong> one allele <strong>of</strong> a gene in which the other allele was already<br />

inactivated. In the context <strong>of</strong> oncogenesis it refers to when the remaining functional allele<br />

in a somatic cell <strong>of</strong> the <strong>of</strong>fspring becomes inactivated by mutation.<br />

16 The "CpG" notation is used to distinguish CG base-pairing <strong>of</strong> cytosine and guanine.<br />

15


1.3 Primary and Secondary Glioblastomas Introduction<br />

were found mostly in all non-CpG dinucleotides. This pattern is consistent<br />

with the failure to repair alkylated guanine residues that is caused by treat-<br />

ment if MGMT methylation is also shifting the mutation spectrum <strong>of</strong> treated<br />

samples to a preponderance <strong>of</strong> GC to AT transitions at non-CpG sites [326].<br />

The molecular mechanisms lying behind such pattern could find an expla-<br />

nation in the interesting observation that the mutation spectra <strong>of</strong> mismatch<br />

repair genes reflected MGMT promoter methylation as well. In fact, in treated<br />

hypermutated samples with methylated MGMT, mismatch repair genes accu-<br />

mulated GC to AT transitions in non-CpG islands, which was not observed<br />

in any <strong>of</strong> the hypermutated tumours with unmethylated MGMT. Thus, mis-<br />

match repair deficiency and MGMT methylation status together could have<br />

powerful clinical implications in the context <strong>of</strong> treatment, raising the possibil-<br />

ity that patients who initially respond to the frontline therapy in use today may<br />

evolve not only treatment resistance, but also an MMR-defective hypermuta-<br />

tor phenotype. The fact that newly diagnosed glioblastomas with methylated<br />

MGMT respond well to treatment with alkylating agents, is in part due to<br />

the initiation <strong>of</strong> many mismatch repair cycles that attempt to repair the alky-<br />

lated guanines and in doing so lead to cell death, which is consistent with<br />

the observation that the mismatch repair genes themselves are mutated with<br />

CG to AT transitions at non-CpG sites [326]. Therefore, initial methylation<br />

<strong>of</strong> MGMT in this scenario would have an effect on two fronts: shifting the<br />

mutation spectrum that will affect mutations at mismatch repair genes, and<br />

increasing the selective pressure to lose mismatch repair function, resulting<br />

in aggressive recurrent tumours with a hypermutator phenotype [363]. These<br />

findings highlight the importance <strong>of</strong> designing selective strategies that target<br />

mismatch-repair-deficient cells in combination with alkylating agents, in order<br />

to prevent or minimise the emergence <strong>of</strong> treatment resistance [326].<br />

Genetically Engineered Mouse Models Murine gliomas that appear to<br />

develop in the absence <strong>of</strong> lower grade precursors are very important disease<br />

models because they reproduce de novo conditions for the onset <strong>of</strong> glioblas-<br />

toma. GEMMs are useful research tools towards that end because they can<br />

accurately reproduce the initiation and progression stages in the human pathol-<br />

ogy upon introduction <strong>of</strong> few mutations, although it is still debated whether<br />

they can accurately recreate the genomic and expression heterogeneity <strong>of</strong> the<br />

original human disease [308]. In some cases these mouse models helped pre-<br />

dict the importance in human gliomas <strong>of</strong> events such as TP53 and NF1 in-<br />

16


1.3 Primary and Secondary Glioblastomas Introduction<br />

activation [426,549]. Thus, GEMMs have been instrumental so far in the<br />

molecular understanding <strong>of</strong> the causes <strong>of</strong> human gliomagenesis. Mutations in<br />

Phosphatase and tensin homolog (PTEN), TP53 and Retinoblastoma 1 (RB1)<br />

have all been tested in mouse models expressing Cre specifically in the brain<br />

and their phenotypical effects have all been cell-type as well as developmental-<br />

stage specific [100]. The use <strong>of</strong> the Cre-lox system allows specific recombination<br />

events to occur in genomic DNA. The Cre protein is a site-specific DNA re-<br />

combinase that catalyses the recombination <strong>of</strong> DNA between specific loxP sites<br />

that have a directional core sequence between them. When cells express Cre,<br />

the DNA is cut at both loxP sites and the result <strong>of</strong> the recombination depends<br />

on whether the lox sites are located on the same chromosome and whether in<br />

an inverted or direct repeat fashion. Same chromosome inverted lox sites pro-<br />

duce an insertion, while direct repeats cause a deletion. Different chromosome<br />

lox sites may cause translocation events [371].<br />

<strong>Glioma</strong> Cell Lines Cancer cell lines have been the historical standard<br />

both for exploring the biology <strong>of</strong> human tumours and as preclinical models<br />

for screening <strong>of</strong> potential therapeutic agents [261]. Due to the inherent diffi-<br />

culty in establishing and maintaining primary tumour cell cultures, established<br />

cell lines have been traditionally used to characterize the genomic aberrations<br />

identified in primary tumours [275]. However, it is possible that the genetic<br />

aberrations accumulated by repeatedly passaging the cells in vitro may cause<br />

their phenotypic characteristics to bear little resemblance with the primary<br />

human tumour [66,261].<br />

The laboratory <strong>of</strong> Howard Fine has attempted a systematic genomic survey<br />

<strong>of</strong> five <strong>of</strong> the most commonly used glioma cell lines - A172, Hs683, T98G,<br />

U251, and U87 - for the purpose <strong>of</strong> evaluating their similarity to primary<br />

gliomas [275]. Their research, conducted with high-density Single Nucleotide<br />

Polymorphism (SNP) arrays, showed that established glioma cell lines and pri-<br />

mary tumour have significant differences in both genomic alterations and gene<br />

expression, indicating that glioma cell lines may not be an accurate representa-<br />

tion or model system for primary gliomas [275]. The differences observed in the<br />

biological phenotype <strong>of</strong> glioma cell lines compared with primary tumours are<br />

further confounded by the lack <strong>of</strong> molecular hallmarks in serially passaged cell<br />

lines, such as the over-expression <strong>of</strong> EGFR, the silencing <strong>of</strong> cyclin-dependent<br />

kinase inhibitor (CDKN) CDKN2A and the loss <strong>of</strong> PTEN [207,445]. This ex-<br />

plains why in vitro and in vivo cancer cell line-based preclinical therapeutic<br />

17


1.3 Primary and Secondary Glioblastomas Introduction<br />

screening models have been poorly predictive <strong>of</strong> useful therapeutic agents and<br />

may have led to important misinterpretations on the relevance <strong>of</strong> aberrant sig-<br />

naling pathways within cell lines compared to primary tumours [261].<br />

Nonetheless, cell lines don’t contain the typical mixture <strong>of</strong> genetically distinct<br />

cells <strong>of</strong> primary tumours and can therefore be more easily characterised. In<br />

fact, the problematic presence <strong>of</strong> non-tumour cells in primary tumours, makes<br />

it harder to pinpoint the rare mutations that are not spread uniformly through-<br />

out the tumour [275]. On the basis <strong>of</strong> this claim, cancer cell line sequencing<br />

efforts are recently flourishing, including works on the commonly studied grade<br />

IV glioma cell line U87MG [101].<br />

With these pros and cons in mind, in the study by Lee et al [261] they went on<br />

to search for a more biologically relevant model system for exploring glioma bi-<br />

ology and for the screening <strong>of</strong> new therapeutic targets, which they found in neu-<br />

ral stem (NS) cells. These cells have characteristics <strong>of</strong> continuous self-renewal,<br />

extensive migration and infiltration <strong>of</strong> brain parenchyma and the potential for<br />

full or partial differentiation, which are lost in glioma cell lines [261,286,347]<br />

and will be discussed in greater detail in chapter three (see Section 3.3).<br />

Classification Systems<br />

Several decades <strong>of</strong> experimentation on glioblastoma have highlighted that<br />

specific genetic lesions are more commonly observed in certain subclasses <strong>of</strong><br />

glioblastoma. Primary glioblastoma typically harbours mutations in the EGFR<br />

receptor tyrosine kinase gene, tumour suppressor PTEN and cyclin inhibitor<br />

CDKN2A, while secondary glioblastoma harbours mutations in Platelet-derived<br />

growth factor (PDGF) and tumour suppressor TP53. However, the latter as-<br />

sociation is now starting to be considered a historical one, since an increasing<br />

number <strong>of</strong> studies are showing that TP53 mutations occur in a significant<br />

amount <strong>of</strong> primary glioblastomas [383,549]. These alterations can become pre-<br />

dictive <strong>of</strong> glioma subclasses. Glioblastomas with intact expression <strong>of</strong> the PTEN<br />

and EGFR vIII 17 proteins, for example, correlate with increased EGFR kinase<br />

inhibitor response as compared to tumours expressing EGFR vIII but lacking<br />

PTEN [329].<br />

Immunohistochemical markers have been important tools so far in the classifi-<br />

cation and diagnosis <strong>of</strong> malignant gliomas, with Glial fibrillary acidic protein<br />

(GFAP) and Oligodendrocyte lineage transcription factor 2 (OLIG2) being two<br />

17 The vIII mutant <strong>of</strong> EGFR is the most common in glioblastoma and results from a<br />

non-random 801bp in-frame deletion <strong>of</strong> exons 2 to 7 <strong>of</strong> the EGFR gene [371].<br />

18


1.3 Primary and Secondary Glioblastomas Introduction<br />

<strong>of</strong> the most specific ones [154]. GFAP is universally expressed in astrocytic<br />

and ependymal tumours and OLIG2 is an oligodendroglial as well as stem cell<br />

marker expressed at high levels only in diffuse gliomas [281,338,435]. Recently<br />

investigated novel markers are stem and progenitor cell markers. Intensive<br />

research efforts are attempting to uncover agents that may target subpopula-<br />

tions <strong>of</strong> these cells with high tumourigenic potential and increased resistance<br />

to current therapies [154]. The cell surface marker CD133 and other mark-<br />

ers <strong>of</strong> stem cells, such as Nestin, Musashi and Sex determining region y-box<br />

2 (SOX2), have been shown to negatively correlate with outcome parame-<br />

ters [304].<br />

In an attempt to optimise the association <strong>of</strong> different prognoses with differ-<br />

ent therapies, several studies have focused their efforts in building an accurate<br />

classification system [154,383,390]. Elucidating patterns between prognosis<br />

and specific genetic lesions would allow therapies to tailor to the group <strong>of</strong><br />

patients who will most likely respond to them, an approach also known as<br />

"stratification <strong>of</strong> treatment" [383]. Genome-wide pr<strong>of</strong>iling studies such as the<br />

ones conducted by Freije et al in 2004 [148] Phillips et al in 2006 [390] and<br />

Verhaak et al in 2010 [511], have tried to categorise glioblastoma in molecular<br />

subclasses that could be predictive <strong>of</strong> survival outcomes. Thus, microarray<br />

gene expression data for hundreds <strong>of</strong> high-grade glioma samples was analysed<br />

and has shown that most tumours can be classified into a small number <strong>of</strong><br />

subtypes correlated with survival and response to therapy.<br />

The largest such study to date [511] built a dataset from 200 GBM and two<br />

normal brain samples that was used to identify four glioblastoma subtypes<br />

named Proneural, <strong>Neural</strong>, Classical and Mesenchymal, each characterised by<br />

a distinct gene expression signature encompassing a set <strong>of</strong> 210 up-regulated<br />

genes. An independent set <strong>of</strong> 260 GBM expression pr<strong>of</strong>iles was compiled from<br />

the public domain, including TCGA and Phillips et al [390], that successfully<br />

assessed subtype reproducibility. The Proneural subtype was associated with<br />

younger age, Platelet-derived growth factor receptor α (PDGFRA) abnormal-<br />

ities, and IDH1 and TP53 mutations, all <strong>of</strong> which have previously been as-<br />

sociated with secondary GBM and correlate with longer survival times [326].<br />

In confirmation <strong>of</strong> this pattern, the Proneural subtype previously identified<br />

in the study by Phillips et al also included most grade III gliomas and 75%<br />

<strong>of</strong> lower grade gliomas [390]. The Classical subtype was strongly associated<br />

with the astrocytic signature and contained all common genomic aberrations<br />

19


1.3 Primary and Secondary Glioblastomas Introduction<br />

observed in GBM, such as chromosome 7 amplifications, chromosome 10 dele-<br />

tions, EGFR amplification, and deletion <strong>of</strong> the TP53-stabilising is<strong>of</strong>orm <strong>of</strong> the<br />

cyclin-dependent inhibitor CDKN2A:ARF [326]. As already observed in the<br />

study by Phillips et al, the Mesenchymal subtype was characterised by high<br />

expression <strong>of</strong> Chitinase 3-like 1 (CHI3L1) and Met proto-oncogene (MET) and<br />

also a lack <strong>of</strong> association with a specific signature, but rather an equal cor-<br />

relation with the neural, astrocytic, and oligodendrocytic gene signatures. A<br />

striking characteristic <strong>of</strong> this class was the strong association with NF1 dele-<br />

tion, already known to induce GBM in Nf1;p53 double knockout mice [426] and<br />

shown to occur in a variety <strong>of</strong> tumours such as neur<strong>of</strong>ibromas [550], but only<br />

recently observed in human GBMs [69,320]. Since the Proneural subtype was<br />

associated with a trend toward longer survival and the samples did not show a<br />

survival advantage from aggressive treatment protocols, but a clear treatment<br />

effect was observed in the Classical and Mesenchymal samples, the results <strong>of</strong><br />

this pr<strong>of</strong>iling-based classification study may find important roles in suggesting<br />

different therapeutic strategies <strong>of</strong> high clinical impact. For example, extend-<br />

ing the current biomarker assays for GBM to include subtyping tests for key<br />

genetic events, including NF1 and PTEN loss, IDH1 and Phosphoinositide-3-<br />

kinase (PI3K) mutation, PDGFRA and EGFR amplification [511].<br />

Other studies attempting to define distinct subgroups <strong>of</strong> glioma identified CpG<br />

island methylator phenotypes [363] and microRNA pr<strong>of</strong>iles [231] as part <strong>of</strong><br />

the same goal towards a new therapeutic approach. In the former study by<br />

Noushmehr et al, promoter DNA methylation was assessed in 272 glioblas-<br />

tomas from the TCGA dataset and validated in a different set <strong>of</strong> non-TCGA<br />

glioblastomas and low-grade gliomas. Three DNA methylation clusters were<br />

identified on array-based methylation assay platforms and one <strong>of</strong> these formed<br />

a particularly tight cluster with a highly characteristic DNA methylation pro-<br />

file designated as the "glioma CpG island methylator phenotype" or G-CIMP.<br />

The G-CIMP sample cluster was highly enriched for the Proneural expression<br />

pr<strong>of</strong>ile defined by Verhaak et al [511] and the 24 G-CIMP-positive patients<br />

were all significantly associated with IDH1 somatic mutations and a longer<br />

survival time, making G-CIMP status a potential predictor <strong>of</strong> improved pa-<br />

tient survival. The authors claim that if a transacting factor were involved in<br />

the protection from methylation <strong>of</strong> the CpG island promoters in the G-CIMP<br />

cluster, then the loss <strong>of</strong> its function could provide a favourable context for the<br />

acquisition <strong>of</strong> specific genetic lesions such as IDH1 mutation. The two GBM<br />

20


1.4 Pathways Involved in Glioblastoma Introduction<br />

subgroups identified through the G-CIMP status would therefore have impor-<br />

tant implications in the assessment <strong>of</strong> therapeutic strategies for different GBM<br />

patients [363].<br />

The microRNA pr<strong>of</strong>iling study by Kim et al [231] analysed 261 microRNA<br />

expression pr<strong>of</strong>iles from TCGA, identifying five clinically and genetically dis-<br />

tinct subclasses <strong>of</strong> glioblastoma that each related to a different neural precursor<br />

cell type: radial glia, oligoneuronal precursors, neuronal precursors, neuroep-<br />

ithelial/neural crest precursors and astrocyte precursors, suggesting a rela-<br />

tionship between each subclass and a distinct stage <strong>of</strong> neural differentiation.<br />

Interestingly, when compared to the glioblastoma subclasses identified by Ver-<br />

haak et al [511], the microRNA-based oligoneural, radial glial, and astrocytic<br />

subclasses were enriched in tumours from the Proneural, Classical, and Mes-<br />

enchymal subtypes, respectively. MicroRNA-based consensus clustering also<br />

yielded robust survival differences, with oligoneural glioblastoma patients liv-<br />

ing significantly longer, and a distinct pattern <strong>of</strong> somatic mutations, with the<br />

oligoneural subclass enriched for IDH1 and Phosphoinositide-3-kinase receptor<br />

1 (PIK3R1) mutations but lacking NF1 mutations. A very high connectivity,<br />

i.e. the number <strong>of</strong> directly connected mRNAs, was displayed by miR-9 and<br />

miR-222 to the oligoneural and astrocytic precursor subtypes, respectively,<br />

suggesting that these microRNAs might serve as core regulators <strong>of</strong> subclass-<br />

specific gene expression in glioblastoma. Overall, this study provided strong<br />

evidence that glioblastomas can arise from the transformation <strong>of</strong> neural precur-<br />

sors at each <strong>of</strong> the stages represented by a microRNA-identified subclass and<br />

that microRNAs are therefore useful for sub-classifying glioblastomas to gener-<br />

ate accurate prognoses and for the development <strong>of</strong> molecular-based treatment<br />

decisions [231].<br />

1.4 Pathways Involved in Glioblastoma<br />

Glioblastoma <strong>of</strong>ten involves the concurrent deregulation <strong>of</strong> three core path-<br />

ways: RTK/PI3K/PTEN signaling, p53 signaling and Rb-mediated control <strong>of</strong><br />

cell cycle progression (Fig 1.2) [2,286,326,383]. Therefore, important genetic<br />

events in human glioblastoma are the deregulation <strong>of</strong> growth factor signaling<br />

pathways via amplification or mutational activation <strong>of</strong> receptor tyrosine ki-<br />

nases (RTKs), the activation <strong>of</strong> the PI3K pathway and inactivation <strong>of</strong> the p53<br />

and Rb tumour suppressor pathways [286,326]. The Rb and p53 pathways,<br />

21


1.4 Pathways Involved in Glioblastoma Introduction<br />

which regulate cell cycle primarily by governing the G1/S 18 phase transition,<br />

are major targets <strong>of</strong> inactivating mutations in glioblastoma and their absence<br />

renders tumours particularly susceptible to inappropriate cell division driven<br />

by constitutively active mitogenic signaling effectors, such as PI3K and MAP<br />

kinases [154]. The same three pathways were singled out in the TCGA project<br />

when mapping the somatic nucleotide substitutions, homozygous deletions and<br />

focal amplifications, onto the major pathways implicated in glioblastoma. A<br />

statistical tendency was observed in which components within each pathway<br />

were altered in a mutually exclusive fashion, hinting at deregulation <strong>of</strong> one com-<br />

ponent relieving the selective pressure for additional ones in the same pathway.<br />

Also, 74% <strong>of</strong> the samples harbored aberrations in all three pathways, suggest-<br />

ing a functional non-redundancy between the three as a key requirement for<br />

glioblastoma pathogenesis [326].<br />

The study by Parsons et al in 2008 [383] detected critical genes within im-<br />

portant cell signaling pathways in glioma: TP53, Mdm2 p53 binding protein<br />

homolog (MDM2) and Mdm4 p53 binding protein homolog (MDM4) in the<br />

p53 pathway; RB1, CDK4 and CDKN2A in the Rb pathway and PIK3CA,<br />

PIK3R1, PTEN and IRS1 in the RTK/PI3K/PTEN pathway. All but one <strong>of</strong><br />

the cancers bearing mutations in members <strong>of</strong> one <strong>of</strong> these three pathways did<br />

not show alterations in any <strong>of</strong> the other two, suggesting functional redundancy<br />

for these mutations in tumourigenesis [383].<br />

RTK/PI3K/PTEN Pathway<br />

Tumour cells acquire genomic alterations that greatly reduce their dependence<br />

on exogenous growth stimulation via transmembrane receptor contact with dif-<br />

fusible growth factors, cell to cell adhesion, or extracellular matrix. Most <strong>of</strong>ten<br />

these cells enable inappropriate cell division, survival, and motility through the<br />

constitutive activation <strong>of</strong> the PI3K and MAPK pathways. The predominant<br />

mechanism <strong>of</strong> mitogenic signaling activation for gliomas occurs through RTKs,<br />

high-affinity cell surface receptors that bind and transduce the signal from cy-<br />

tokines, growth factors and hormones, and integrins, membrane-bound recep-<br />

tors that mediate the interaction between the extracellular matrix and the cy-<br />

toskeleton [154]. Receptors that belong to the RTK family are EGFR, PDGFR,<br />

MET, FGFR and VEGFR [431]. The epidermal growth factor EGF and the<br />

platelet-derived growth factor PDGF pathways play important roles in both<br />

18 Major checkpoint in the regulation <strong>of</strong> cell cycle beyond which the cell is committed to<br />

dividing.<br />

22


1.4 Pathways Involved in Glioblastoma Introduction<br />

23<br />

Figure 1.2: KEGG <strong>Glioma</strong> Pathway [2]. Oncogenes and tumour suppressors are highlighted in red.


1.4 Pathways Involved in Glioblastoma Introduction<br />

CNS development and gliomagenesis [154]. The EGFR family is composed<br />

<strong>of</strong> four structurally related members: EGFR, ERBB2, ERBB3, and ERBB4.<br />

The importance <strong>of</strong> RTKs is made obvious by the fact that 58 out <strong>of</strong> the 90<br />

unique tyrosine kinase genes in the human genome, encode for receptor tyro-<br />

sine kinase proteins [431]. EGFR gene amplification occurs in roughly 40% <strong>of</strong><br />

all glioblastomas, and the amplified genes are frequently rearranged [154,262].<br />

Alterations in this family were found in 41 <strong>of</strong> the 91 TCGA samples, includ-<br />

ing the vIII mutant, extracellular domain point mutations and cytoplasmic<br />

domain deletions [326]. The vIII EGFR mutant contains deletions <strong>of</strong> exons<br />

two to seven and occurs in 20-30% <strong>of</strong> all human glioblastoma, making it the<br />

most common EGFR mutant [154]. Although ERBB2 mutation was previously<br />

reported in only one glioblastoma, seven samples <strong>of</strong> the 91 TCGA harbored<br />

11 somatic ERBB2 mutations, including mostly missense 19 and one splice-site<br />

mutation. Unlike in breast cancer, however, no amplifications were observed<br />

for ERBB2 [326]. The PDGFR family <strong>of</strong> receptors contains two members,<br />

PDGFRα and PDGFRβ, that homodimerize or heterodimerize depending on<br />

which growth factor is bound to them [191]. PDGFRα and its ligands, PDGFA<br />

and PDGFB, are expressed in gliomas, particularly in high-grade tumours,<br />

while strong expression <strong>of</strong> PDGFRβ occurs in proliferating endothelial cells<br />

in glioblastoma [154]. In contrast to EGFR, amplification or rearrangement<br />

<strong>of</strong> PDGFR is much less common, and a relatively rare oncogenic deletion <strong>of</strong><br />

exons eight and nine, similarly to EGFR vIII, is constitutively active and en-<br />

hances tumourigenicity [154]. Given the tumoural co-expression <strong>of</strong> PDGF and<br />

PDGFR, autocrine and paracrine loops may be the primary means by which<br />

this growth factor axis exerts its effects [154]. Although RTKs are known to<br />

signal through both the MAPK and PI3K pathways, GEMMs showed con-<br />

sistent high activation <strong>of</strong> just the PI3K pathway in high-grade astrocytomas,<br />

hinting at the fact that downstream consequences <strong>of</strong> RTK activation may vary<br />

greatly [100].<br />

The PI3K complex belongs to class I <strong>of</strong> the lipid kinase family <strong>of</strong> phospho-<br />

inositide 3-kinases and is composed <strong>of</strong> a catalytic subunit, PIK3CA, and a<br />

regulatory subunit, PIK3R1. Activating missense mutations in the adaptor<br />

binding and kinase domains <strong>of</strong> the catalytic subunit <strong>of</strong> class I PI3K complexes<br />

are known to occur in different types <strong>of</strong> tumours, including glioblastoma, while<br />

19 A missense mutation is a point mutation that results in a codon coding for a different<br />

amino acid.<br />

24


1.4 Pathways Involved in Glioblastoma Introduction<br />

mutations in the regulatory subunits are less common [35,36,286]. Of the 91<br />

samples from the TCGA study, nine carried somatic mutations in the regu-<br />

latory subunit that clustered around the three amino acids acting as contact<br />

points for the catalytic subunit, suggesting the possibility that these mutations<br />

prevent inhibitory contacts between the two subunits and cause constitutive<br />

PI3K activity [326]. The PI3K family <strong>of</strong> enzymes phosphorylates the inositol<br />

ring <strong>of</strong> phosphatidylinositol 20 on the three, four and five hydroxyl groups in<br />

many different combinations [157,440]. Class I PI3K complexes can be acti-<br />

vated by small GTPases like RAS or RTKs [154].<br />

Upon activation from RTKs, PI3K catalyses the phosphorylation <strong>of</strong> phos-<br />

phatidylinositol (3,4)-bisphosphate to phosphatidylinositol (3,4,5)-trisphosphate<br />

(PIP3) [154,267]. The generation <strong>of</strong> PIP3 in the cytosolic side <strong>of</strong> the cell mem-<br />

brane acts as a docking site for the serine/threonine protein kinases <strong>of</strong> the<br />

Akt family and the 3-phosphoinositide dependent protein kinase-1 (PDPK1).<br />

Upon relocation, PDPK1 and mammalian target <strong>of</strong> rapamycin mTOR activate<br />

AKT1 via phosphorylation <strong>of</strong> two key residues, starting the AKT-mediated sig-<br />

naling cascade that promotes cell survival and proliferation [154]. AKT activa-<br />

tion may be compromised via two other mechanisms in glioblastoma: dephos-<br />

phorylation from the PH domain and leucine rich repeat protein phosphatase 1<br />

(PHLPP) [74] or inhibition <strong>of</strong> phosphorylation through the C-terminal modu-<br />

lator protein (CTMP) inhibitor [307]. One <strong>of</strong> the targets <strong>of</strong> AKT1 is phospho-<br />

rylation <strong>of</strong> the Forkhead box O (FOXO) family <strong>of</strong> transcription factors, which<br />

promotes their exclusion from the nucleus and reduces the expression <strong>of</strong> im-<br />

portant target genes such as the cyclin-dependent kinase inhibitors CDKN1A<br />

and CDKN1B, both also directly targeted by AKT1, and the Rb family mem-<br />

ber p130 [154]. The action <strong>of</strong> class I PI3Ks is directly antagonized by PTEN<br />

through dephosphorylation <strong>of</strong> PIP3 and inhibition <strong>of</strong> AKT1 relocation, which<br />

strongly reduces AKT1-mediated cell cycle promotion [84,99,111].<br />

Furthermore, in 86% <strong>of</strong> the TCGA samples, at least one genetic event was har-<br />

boured in the RTK/PI3K/PTEN pathway as well as frequent deletions <strong>of</strong> the<br />

PTEN lipid phosphatase gene. Within the RTK/PI3K/PTEN pathway, fre-<br />

quent aberrations were shown in EGFR, ERBB2, PDGFRA and MET [326].<br />

Patients with PTEN deletions or activating mutations in the catalytic and<br />

regulatory subunits <strong>of</strong> class I PI3K complexes, might benefit from PI3K or<br />

PDPK1 inhibitors, whereas patients in which the PI3K pathway is altered by<br />

20 Negatively charged phospholipid and a minor component in the cytosolic side <strong>of</strong> eukary-<br />

otic cell membranes.<br />

25


1.4 Pathways Involved in Glioblastoma Introduction<br />

AKT amplification might be refractory. Also, the co-amplification exhibited<br />

by multiple RTKs in the same glioblastoma sample may be tailored with anti-<br />

RTK therapies to specific patterns <strong>of</strong> RTK mutation [326].<br />

PTEN is a major tumour suppressor that is inactivated in 50% <strong>of</strong> high-grade<br />

gliomas by mutations or epigenetic mechanisms, each resulting in uncontrolled<br />

PI3K signaling [154]. PTEN expression is completely extinguished in tumour<br />

cells <strong>of</strong> hGFAP-Cre + ;p53 lox/lox ;Pten lox/+ GEMMs 21 with developed anaplastic<br />

astrocytomas and glioblastomas, but it is present in the surrounding normal<br />

tissue and vessels supplying tumour tissue. Such LOH effect is very frequent in<br />

human high-grade gliomas [549]. PTEN is located in the long arm <strong>of</strong> chromo-<br />

some 10 and acts as the central negative regulator <strong>of</strong> the PI3K/AKT pathway<br />

due to its lipid phosphatase activity that affects RTK signaling [84,99,111].<br />

As a consequence, the RTK/PI3K pathway is commonly affected by the bial-<br />

lelic inactivation <strong>of</strong> PTEN or LOH <strong>of</strong> the long arm <strong>of</strong> chromosome 10. Loss<br />

<strong>of</strong> PTEN most <strong>of</strong>ten results in constitutive activation <strong>of</strong> AKT1 but is not, in<br />

mature astrocytes, sufficient to drive proliferation and initiate gliomagenesis<br />

in the absence <strong>of</strong> other mutations. This suggests that the PI3K/AKT pathway<br />

is not sufficiently stimulated by the absence <strong>of</strong> its main negative regulator to<br />

elevate pathway activity in astrocytes [100]. Furthermore, PTEN may act to<br />

suppress transformation and tumour progression beyond regulation <strong>of</strong> PI3K<br />

signaling. In a study by Shen et al [453], quiescent cells from mouse model<br />

cell systems 22 harboured high levels <strong>of</strong> nuclear PTEN, which appeared to fulfill<br />

important roles in the maintenance <strong>of</strong> genomic integrity, through centromere<br />

stabilisation and promotion <strong>of</strong> DNA repair [453]. In other studies, a number<br />

<strong>of</strong> PTEN point mutations found in familial cancer predisposition syndromes<br />

had no effect on enzyme activity and lied within important sequences for the<br />

localisation <strong>of</strong> PTEN. Analysis <strong>of</strong> such mutants has confirmed that aberrant<br />

sequestration <strong>of</strong> PTEN into either the nucleus or the cytoplasm compromises<br />

its tumour suppressor function [117,154,495].<br />

21 In the study by Zheng et al [549], the hGFAP-Cre transgene was used to delete p53 alone<br />

or in combination with Pten in all CNS lineages using conditional p53 and Pten alleles, with<br />

modelling efforts directed towards the Pten lox/+ genotype since broad CNS deletion <strong>of</strong> Pten<br />

results in lethal hydrocephalus in early mouse postnatal life.<br />

22 This mouse system included mouse embryonic fibroblasts and mouse embryonic stem<br />

cells.<br />

26


1.4 Pathways Involved in Glioblastoma Introduction<br />

p53 Pathway<br />

The tumour suppressor and transcription factor TP53 is the most commonly<br />

mutated gene in human cancers and a master regulator <strong>of</strong> cell survival path-<br />

ways, as the number <strong>of</strong> solely its protein interactors suggests (Fig 1.3). After<br />

activation by cellular stresses, TP53 functions to trans-activate genes that<br />

mediate cell cycle arrest, apoptosis, DNA repair, inhibition <strong>of</strong> angiogenesis<br />

and metastasis, and other p53-dependent activities [185]. In humans, TP53<br />

Figure 1.3: Visualisation generated from list <strong>of</strong> 345 interactors (orange) <strong>of</strong> TP53<br />

(yellow) from the BioGRID 3.1 [62] repository for interaction datasets.<br />

is located on the short arm <strong>of</strong> chromosome 17 and may be inactivated di-<br />

rectly by gene mutations or indirectly by alterations in genes that promote<br />

degradation <strong>of</strong> the TP53 protein [100,491,515]. TP53 signaling is commonly<br />

affected by biallelic inactivation <strong>of</strong> TP53 and sometimes via amplification <strong>of</strong><br />

MDM2 or loss or mutation <strong>of</strong> the cyclin inhibitor CDKN2A:ARF [100]. While<br />

MDM2 is a direct inhibitor <strong>of</strong> TP53 through its ubiquitin ligase activity,<br />

CDKN2A has been identified in at least three alternatively spliced variants,<br />

two <strong>of</strong> which encode is<strong>of</strong>orms inhibitors <strong>of</strong> the CDK4 kinase. The remain-<br />

ing transcript, however, includes an alternate first exon that contains an al-<br />

ternate open reading frame (ARF) specifying a protein that is structurally<br />

unrelated to the products <strong>of</strong> the other variants and stabilises TP53 by se-<br />

questering MDM2 [233,491,515]. CDKN2A is predominantly inactivated by<br />

biallelic loss or hypermethylation in 50% to 70% <strong>of</strong> high-grade gliomas and<br />

roughly 90% <strong>of</strong> cultured glioma cell lines. Concordantly, the chromosomal re-<br />

gion containing MDM2 is amplified in roughly 10% <strong>of</strong> primary glioblastomas,<br />

the majority <strong>of</strong> which contain intact TP53 [154]. Furthermore, the discovery<br />

27


1.4 Pathways Involved in Glioblastoma Introduction<br />

<strong>of</strong> the MDM2-related gene MDM4, which inhibits TP53 and enhances the ac-<br />

tivity <strong>of</strong> MDM2, prompted the finding <strong>of</strong> 4% <strong>of</strong> glioblastomas with MDM4<br />

amplification and no TP53 mutation nor MDM2 amplification [284]. Through<br />

CDKN2A:ARF mediated stabilisation and activation, TP53 is able to acti-<br />

vate a potent cyclin-dependent kinase inhibitor, CDKN1A. By binding and<br />

inhibiting CDK4 and CDK6 complexes, CDKN1A acts as a regulator <strong>of</strong> the<br />

G1 progression <strong>of</strong> cell cycle [160,233]. Another cyclin-dependent inhibitor <strong>of</strong><br />

the same family as CDKN2A is CDKN2C, which has recently been suggested<br />

to drive glioblastoma pathogenesis. CDKN2C inhibits the formation <strong>of</strong> the<br />

CDK4/CDK6 complex with cyclin dependent kinases, needed to keep the cell<br />

cycle from stalling at the G1 phase. Homozygous deletions <strong>of</strong> CDKN2C were<br />

reported in glioblastoma multiforme as well as missense mutations that dis-<br />

turb its binding with CDK6. Suggestions based on these GBM studies are that<br />

CDKN2C is a tumour suppressor that compensates for CDKN2A homozygous<br />

deletion by being up-regulated through the action <strong>of</strong> the E2F1 transcription<br />

factor [467].<br />

In the TCGA project, inactivation <strong>of</strong> the p53 pathway occurred mostly in the<br />

form <strong>of</strong> TP53 mutations, but also <strong>of</strong> CDKN2A:ARF deletions and MDM2 and<br />

MDM4 amplifications. While genetic lesions in TP53 were mutually exclusive<br />

<strong>of</strong> those in MDM2 or MDM4, CDKN2A:ARF deletions were concurrent to<br />

TP53 mutations in 30% <strong>of</strong> the samples [326]. The best-characterised effector<br />

<strong>of</strong> TP53 is the transcriptional target CDKN1A. Although this gene has not<br />

been found to be altered in gliomas, its expression is frequently abrogated<br />

by TP53 functional inactivity as well as by mitogenic signaling through the<br />

PI3K and MAPK pathways [154]. Although TP53 mutation was historically<br />

associated with low-grade gliomas and secondary human glioblastomas, works<br />

done with GEMMs prompted the re-sequencing <strong>of</strong> both TP53 and PTEN to<br />

re-evaluate their combinatorial role in the disease [549]. PTEN had already<br />

been associated with primary glioblastoma [423] and the results <strong>of</strong> these works<br />

showed that 60% <strong>of</strong> the clinically annotated human primary glioblastomas with<br />

TP53 mutations also harboured a PTEN mutation or homozygous deletion,<br />

indicating that TP53, together with PTEN, is also a key player in human<br />

primary glioblastoma, as the TCGA data also reports [326,549].<br />

28


1.4 Pathways Involved in Glioblastoma Introduction<br />

Rb Pathway<br />

The importance <strong>of</strong> the inactivation <strong>of</strong> the Rb pathway in glioma progression is<br />

evidenced by the near-universal and mutually exclusive alteration <strong>of</strong> Rb path-<br />

way effectors and inhibitors in both primary and secondary glioblastoma [154].<br />

RB1 is located on the long arm <strong>of</strong> chromosome 13 and similarly to the p53<br />

pathway, the Rb pathway is also affected by homozygous deletions <strong>of</strong> the<br />

CDKN2A locus. The CDKN2A gene is, with the exception <strong>of</strong> its alternate<br />

reading frame is<strong>of</strong>orm CDKN2A:ARF, a negative regulator <strong>of</strong> p53 signaling<br />

as well as a regulator <strong>of</strong> the G1 checkpoint in the Rb-mediated progression <strong>of</strong><br />

cell cycle [154,398]. The RB1 gene is mutated in roughly 25% <strong>of</strong> high-grade<br />

astrocytomas and the loss <strong>of</strong> the long arm <strong>of</strong> chromosome 13 characterises<br />

the transition from low to intermediate grade gliomas. Amplification <strong>of</strong> the<br />

CDK4 gene accounts for the functional inactivation <strong>of</strong> RB1 in roughly 15% <strong>of</strong><br />

high-grade gliomas, and CDK6 is also amplified but at a lower frequency [154].<br />

Deregulation <strong>of</strong> Rb signaling leading to G1/S progression appears to be a crit-<br />

ical event in gliomagenesis whether or not inactivation <strong>of</strong> RB1 is an initiating<br />

event [100]. Cell cycle progression is regulated by the activities <strong>of</strong> complexes <strong>of</strong><br />

cyclins and CDKs, which phosphorylate RB1 and block its growth-inhibitory<br />

functions [318]. G1 progression is controlled by the D-type cyclins, which<br />

form active complexes with CDK4 or CDK6, and E-type cyclins in associa-<br />

tion with CDK2 (Fig 1.4) [398]. Within the TCGA dataset, 77% <strong>of</strong> samples<br />

showed genetic alterations in the Rb pathway. Among these, the deletion <strong>of</strong> the<br />

CDKN2A/CDKN2B locus on the short arm <strong>of</strong> chromosome nine was the most<br />

common event, followed by amplification <strong>of</strong> the CDK4 locus [326]. CDKN2B<br />

and CDKN2A lie adjacent in the short arm <strong>of</strong> chromosome nine and together<br />

define a region that is frequently mutated and deleted in a wide variety <strong>of</strong><br />

tumours [233]. CDKN2A and CDKN2B both form complexes with CDK4,<br />

CDK6 and cyclin D to block their activation and progression <strong>of</strong> cell cycle into<br />

the G1/S phase [382,454]. Interestingly, all samples with RB1 nucleotide sub-<br />

stitutions lacked CDKN2A/CDKN2B locus deletion, suggesting that this type<br />

<strong>of</strong> RB1 inactivation obviates the genetic pressure for activation <strong>of</strong> upstream<br />

cyclin and cyclin-dependent kinase complexes. Thus, it would be reasonable<br />

to speculate that patients with deletions in CDKN2A or CDKN2B or with<br />

amplifications in CDK4 or CDK6 could benefit from a treatment with CDK<br />

inhibitors, unlikely to affect patients with RB1 mutation [326]. However, nu-<br />

merous in vitro and in vivo assays have demonstrated that the neutralisation<br />

29


1.5 Pathway Crosstalk Introduction<br />

Figure 1.4: The Biocarta pathway for Rb signaling [61]. The cell cycle checkpoints<br />

at the G1/S and G2/M transitions prevent progression when DNA is damaged. The<br />

cyclin-dependent kinase CDK2 targets and phosphorylates RB1 to allow progression<br />

to the G1/S transition. When the cell is in a quiescent state, hypophosphorylated<br />

RB1 blocks proliferation by binding and sequestering the E2F family <strong>of</strong> transcription<br />

factors, which prevents the transactivation <strong>of</strong> genes essential for progression through<br />

the cell cycle [154]. Upon stimulation and activation <strong>of</strong> the MAPK cascade, cyclin D<br />

forms complexes with cyclin-dependent kinases CDK4 and CDK6 and CDK2 is released<br />

from the inhibitory interaction with CDKN1B and binds to cyclin E. Together<br />

these activated complexes phosphorylate RB1, which stops inhibiting E2F transcription<br />

factors so that the cell cycle can proceed through the G1/S checkpoint [68,151].<br />

<strong>of</strong> this pathway alone is insufficient to abrogate cell cycle control to the ex-<br />

tent needed for cellular transformation, suggesting that other important cell<br />

cycle regulation pathways complement its activities in preventing gliomagene-<br />

sis [154].<br />

1.5 Pathway Crosstalk<br />

While the RTK/PI3K/PTEN, p53, and Rb pathways are <strong>of</strong>ten considered as<br />

distinct entities, there is significant crosstalk among them, reinforcing the in-<br />

appropriate regulation <strong>of</strong> single pathway perturbations [154].<br />

While 70% <strong>of</strong> secondary glioblastomas share the common event <strong>of</strong> IDH1 mu-<br />

tation, which initiates them into the development <strong>of</strong> the higher-grade pathol-<br />

ogy, primary glioblastomas arise de novo and lack a similar common initi-<br />

ating event. In trying to assess this, the study by Chow et al looked into<br />

the cooperativity <strong>of</strong> the three most important pathways in glioblastoma using<br />

GEMMs [100]. Mutations in the three tumour suppressors Pten, p53 and Rb1<br />

were introduced in various combinations in astrocytes and neural precursors<br />

30


1.5 Pathway Crosstalk Introduction<br />

and eventually developed into astrocytomas ranging from grade III to grade<br />

IV [100,308]. Interestingly, none <strong>of</strong> the GEMMs carrying a deletion in only one<br />

<strong>of</strong> the three tumour suppressors developed high-grade astrocytomas. Only the<br />

deletion <strong>of</strong> p53 caused a late onset and low frequency <strong>of</strong> astrocytomas. The<br />

earliest tumour onset was observed in triple knockout mice and the highest<br />

frequency <strong>of</strong> astrocytomas in double knockout mice that carried a p53 dele-<br />

tion. This observation supports a role for p53 inactivation in astrocytoma<br />

initiation, alongside other factors such as the low frequency <strong>of</strong> RB1 and PTEN<br />

mutations and high frequency <strong>of</strong> TP53 mutations in grade II human astro-<br />

cytomas [300,526]. Furthermore, the earlier onset <strong>of</strong> high-grade gliomas in<br />

Pten:p53 knockout mice shows that Pten cooperates more efficiently with p53<br />

mutation than Rb1 mutation in double knockout mice. Interestingly, Pten and<br />

Rb1 mutations together fail to cause gliomagenesis in GEMMs, indicating that<br />

in the absence <strong>of</strong> other mutations these two pathways fail to cooperate [100].<br />

Moreover, these pathways can negatively regulate each other by having TP53<br />

inhibiting the activation <strong>of</strong> the FOXO transcription factors via the activation<br />

<strong>of</strong> the Serum/glucocorticoid regulated kinase 1 (SGK1). Such kinase, in fact,<br />

could induce through phosphorylation the translocation <strong>of</strong> FOXO transcription<br />

factors out <strong>of</strong> the nucleus. This is turn would cause the inhibition <strong>of</strong> TP53 tran-<br />

scriptional activity via a FOXO-mediated increase in the association <strong>of</strong> TP53<br />

with the nuclear export receptors translocating it to the cytoplasm [154,541].<br />

In the study by Chow et al, the array comparative genomics hybridization<br />

(CGH) showed a subset <strong>of</strong> focal and large-scale genomic aberrations typical <strong>of</strong><br />

human glioblastoma subclasses. For example, p53:Pten tumours had acquired<br />

the secondary amplifications <strong>of</strong> RTKs such as Pdgfra, Egfr and Met that are<br />

considered hallmarks <strong>of</strong> the human pathology. Also, p53:Pten:Rb1 tumours<br />

seemed to lose the selective pressure for RTK amplification, since only Pdgfra<br />

was found to be amplified. The understanding <strong>of</strong> how tumour suppressor losses<br />

induce secondary genomic alterations is a key to the direction <strong>of</strong> glioma pro-<br />

gression [100,308]. Thus, Rb1 inactivation seems to further cooperate with<br />

Pten and p53 deletions to generate high-grade gliomas with similar histologi-<br />

cal and biochemical signatures as in Pten:p53 double knockout mice, but with<br />

different selective pressure for RTK amplifications. Other genes affected by<br />

CNAs aside from the RTKs mentioned above are Cdk4, Cdk6, Ccnd1, Ccnd2<br />

and Ccnd3, which all directly regulate the G1/S cell cycle checkpoint and thus<br />

Rb1 activity [68,100,382,454].<br />

31


1.5 Pathway Crosstalk Introduction<br />

The complicated interplay among these critical molecules highlights the need<br />

for detailed dissection <strong>of</strong> the pathways that are aberrant in each tumour to<br />

accurately guide the choice <strong>of</strong> combination therapies that can simultaneously<br />

target multiple pathways [154].<br />

32


Chapter 2<br />

Neurogenesis<br />

Contents<br />

2.1 Radial Glia . . . . . . . . . . . . . . . . . . . . . . . . . . . 33<br />

2.2 <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> . . . . . . . . . . . . . . . . . . . . . . . 37<br />

2.1 Radial Glia<br />

Early ultrastructural studies with electron microscopy revealed that, during<br />

the development <strong>of</strong> the mammalian brain, newly born neurons used the ex-<br />

tended bipolar processes <strong>of</strong> radial glia as a structural support and guide to<br />

migrate to a new location [417]. However, the role <strong>of</strong> radial glia has recently<br />

been extended to that <strong>of</strong> a progenitor population that can divide in the devel-<br />

oping cortex and possibly the entire CNS, producing daughter cells including<br />

neurons, astrocytes and glia [400]. Historically, radial glia were believed to co-<br />

exist with neural progenitors in the ventricular zone <strong>of</strong> the brain, but the past<br />

decade has seen an increase in the amount <strong>of</strong> experimental data challenging<br />

this theory, with radial glia isolated in vitro displaying neuronal differentiation<br />

capacity [310], and dividing precursors in the developing cortex displaying a<br />

radial glia phenotype, as revealed by morphology and markers such as Brain<br />

lipid binding protein (BLBP) and Vimentin [187,361]. After cortical neuroge-<br />

nesis is complete, radial glia retract their processes and convert to multi-polar<br />

astrocytes, leaving specialized forms <strong>of</strong> radial glia persisting in the adult CNS<br />

in locations such as the cerebellum, retina and adult hippocampus [343]. In<br />

mammalian species, for example, radial glia persist into adulthood in the form<br />

<strong>of</strong> Bergmann glia in the cerebellum and Müller glia in the retina [400]. In<br />

non-mammalian vertebrates the number <strong>of</strong> radial glia that persist through-<br />

33


2.1 Radial Glia Introduction<br />

out adulthood is much higher and more widely spread across the CNS, which<br />

might account for the considerable regenerative capacity seen in such species<br />

with respect to that in mammalian species [552].<br />

During the development <strong>of</strong> the mammalian CNS, the steps that precede the<br />

appearance <strong>of</strong> the radial glia progenitor population are the following:<br />

1. Commitment <strong>of</strong> an early population <strong>of</strong> cells to the neural lineage [400];<br />

2. Induction <strong>of</strong> neuroectoderm, promoted by the absence <strong>of</strong> Bone morpho-<br />

genetic protein (BMP) and Fibroblast growth factor 2 (FGF2), and com-<br />

posed <strong>of</strong> a progenitor population termed "neuroepithelial progenitors"<br />

(NEP) cells. Although continuous self-renewal has not been demon-<br />

strated for NEP cells, all cells <strong>of</strong> the CNS directly or indirectly derive<br />

from them [400];<br />

3. NEP cells undergo a process <strong>of</strong> interkinetic nuclear migration, in which<br />

the nucleus oscillates between the apical and basal cellular membrane<br />

in synchrony with cell cycle progression, leading to the formation <strong>of</strong> a<br />

pseudo-stratified epithelium <strong>of</strong> which Sox1 is the earliest marker [389];<br />

4. The neural plate defined by the action <strong>of</strong> NEP cells undergoes a mor-<br />

phogenetic movement that results in the formation <strong>of</strong> the neural tube;<br />

5. Signaling molecules (i.e. retinoic acid (RA), BMP, notochord-derived<br />

Sonic hedgehog (SHH)) are secreted from the nearby tissues to establish a<br />

positional gradient that defines sub-regions <strong>of</strong> the CNS, in which distinct<br />

neuronal and glial subtypes are specified [72];<br />

6. At approximately embryonic day 9.5-10.5 in mouse, a second morpho-<br />

logically distinct cell type appears, termed "radial glia" [400].<br />

Structural features <strong>of</strong> radial glia that distinguish them from their earlier NEP<br />

progenitors are the bipolar morphology, with one extension at the luminal sur-<br />

face 23 <strong>of</strong> the ventricular zone (VZ) and a longer process extending in the op-<br />

posite direction through to the basement membrane adjacent to the pia mater<br />

24 [193] (Fig 2.1); the ovoid cell body, where the nucleus is positioned in the<br />

23This term indicates the surface that looks into the space defined by the interior <strong>of</strong> a<br />

tubular structure.<br />

24The CNS is enclosed in three connective tissue membranes called meninges, one <strong>of</strong> them<br />

being the pia mater that follows the surface <strong>of</strong> the brain and spinal cord closely, extending<br />

into all sulci and depressions <strong>of</strong> the surface [73].<br />

34


2.1 Radial Glia Introduction<br />

VZ adjacent to the lumen; electron lucent processes; abundant intermediate<br />

filaments 25 ; numerous glycogen granules condensed at the end <strong>of</strong> the process<br />

closest to the luminal surface. A second relevant NEP-distinguishing feature <strong>of</strong><br />

radial glia is the expression <strong>of</strong> markers characteristic <strong>of</strong> the astrocytic lineage,<br />

such as the Glutamate aspartate transporter (GLAST), BLBP and GFAP, as<br />

well as the consistent immunoreactivity shown towards the Nestin and Vi-<br />

mentin antibodies [122,310,400]. In figure 2.1, dividing NEP cells appear in<br />

blue and populate the VZ and the sub-ventricular zone (SVZ), while mature<br />

migrated neurons are highlighted in yellow and populate the stratum closest to<br />

the pia mater. Radial glia are shown in green and their process extends with<br />

a bipolar morphology across from the VZ to the pia mater to support mature<br />

neuronal migration in the developing cortex. The stratification <strong>of</strong> the NEP<br />

cell population in the VZ is concordant with cell cycle phase (G1, G1/G2, M),<br />

while the more superficial SVZ displays continued mitotic activity but does not<br />

host a similar cellular stratification. Therefore, the mature cortex eventually<br />

displays an inside-out pattern <strong>of</strong> layering, with the early-born neurons residing<br />

in the deeper layers and the late-born ones residing more superficially next to<br />

the pia mater [193].<br />

Figure 2.1: Cross-section through the neural tube with morphological zones indicated<br />

on the left. NEP cells appear in blue, mature migrated neurons are highlighted<br />

in yellow and radial glia are shown in green. Adapted from Herrup et al 2007 [193].<br />

25 Components <strong>of</strong> the cytoskeletal system that are distinguishable from micr<strong>of</strong>ilaments by<br />

the size <strong>of</strong> their diameter, 8-12 nanometers. They function as a tension-bearing element to<br />

help maintain cell shape and rigidity as well as anchor in place several organelles.<br />

35


2.1 Radial Glia Introduction<br />

Radial glia are by no means a uniform cell population. In fact, they are<br />

found within the cortex and also throughout the developing brain and spinal<br />

cord. This spatial and temporal heterogeneity likely generates the diversity <strong>of</strong><br />

cellular phenotypes within the nervous system. For example, region-specific<br />

expression <strong>of</strong> transcription factors in radial glia is likely to determine the fate<br />

<strong>of</strong> progeny towards one <strong>of</strong> the lineages <strong>of</strong> the CNS [249]. Furthermore, there<br />

are circumstances in which the radial glia phenotype is reacquired, such as af-<br />

ter injury, during reprogramming and dedifferentiation in vitro, and following<br />

epigenetic disruptions in tumorigenesis [400].<br />

Anatomically, a ventricular system is present within the cerebrum 26 that is<br />

composed <strong>of</strong> four communicating compartments, or ventricles, filled by CSF<br />

[387]. The SVZ is a paired brain structure situated in the lining <strong>of</strong> the two<br />

lateral ventricles and is one <strong>of</strong> the major germinal layers during embryogene-<br />

sis [18,41] together with the sub-granular zone (SGZ 27 ), as well as the largest<br />

district in which NS cells with the characteristics <strong>of</strong> astrocytes persist after<br />

birth in the mammalian adult brain [18,332]. Several studies have demon-<br />

strated that radial glia not only give rise to multiple classes <strong>of</strong> brain cells, but<br />

also generate adult SVZ stem cells that maintain the neurogenic lineage in the<br />

adult brain [293,332,413], with a similar relationship proposed also between<br />

radial glia and hippocampal progenitors [223,449]. Specifically, these adult<br />

SVZ stem cells have been shown to arise from a subpopulation <strong>of</strong> radial glia<br />

present within the developing striatum and display characteristics intermedi-<br />

ate between normal astrocytes and radial glia, hinting that NEP cells, radial<br />

glia and adult SVZ stem cells are the components <strong>of</strong> a continuous lineage with<br />

multipotent neural differentiation potential [107,332]. Although these in vitro<br />

studies have demonstrated the shared molecular and morphological charac-<br />

teristics between radial glia and adult SVZ stem cells (Fig 2.2) [107], several<br />

aspects <strong>of</strong> in vivo biology cannot be accounted for. For example, the fact<br />

that radial glia exist only transiently during fetal development makes it harder<br />

experimentally to validate whether they function as self-renewing stem cells.<br />

Also, the artificial environment <strong>of</strong> cultures may result in a unique synthetic cell<br />

state, and the combination <strong>of</strong> transcription factors expressed in cultured SVZ<br />

stem cells is not found in vivo. Therefore, it is fairest to term these precursor<br />

26Largest structure in the mammalian and human brain composed <strong>of</strong> the white and grey<br />

matter in the cranial cavity.<br />

27Adult mammalian neural stem cells have also been isolated from the sub-granular zone<br />

(SGZ) <strong>of</strong> the dentate gyrus in the hippocampus, and the subcortical white matter [442].<br />

36


2.2 <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

cells "radial glia-like NS cells", although in the rest <strong>of</strong> this thesis they will<br />

be referred to simply as NS cells [400]. Radial glia can be obtained from the<br />

Figure 2.2: Surface markers <strong>of</strong> radial glia are expressed by NS cell lines, indicating<br />

that these cells may provide the biological context to work with progenitors <strong>of</strong><br />

the CNS. For example, the GFAP is a type III Intermediate filament; GLAST is<br />

an astrocyte-specific glutamate transporter; Prominin, also known as CD133, is a<br />

glycoprotein that is a neural and hematopoietic stem cell marker. Adapted from<br />

Conti et al 2005 [107].<br />

dissociation <strong>of</strong> fetal CNS tissues and the subsequent establishment <strong>of</strong> primary<br />

cultures. The heterogeneity linked to these primary cultures was overcome by<br />

the development <strong>of</strong> cell type specific monoclonal antibodies like RC1, which<br />

reduces the presence <strong>of</strong> multiple immature and differentiated cell types and<br />

distinct radial glia subtypes. Fluorescent activated cell sorting (FACS) can<br />

also be used with cell surface markers like CD15 and CD133, which, however,<br />

enable only the enrichment and not the isolation <strong>of</strong> radial glia subpopula-<br />

tions. Another technique uses reporter mice in which an endogenous radial<br />

glia promoter drives the expression <strong>of</strong> fluorescent reporters, and cells with ac-<br />

tivated reporter expression are then isolated using FACS. Although primary<br />

cell cultures are a useful tool to isolate and characterise radial glia cells iso-<br />

lated directly from neural tissues, the mitogen-driven expansion <strong>of</strong> cells in vitro<br />

leads to the formation <strong>of</strong> NS cell lines that are an invaluable tool for molec-<br />

ular and biochemical studies on nervous system pathological models amongst<br />

others [400].<br />

2.2 <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong><br />

NS cells are defined as clonogenic cells capable <strong>of</strong> self-renewal and multipotent<br />

differentiation into the three main cell types <strong>of</strong> the CNS: neurons, astrocytes<br />

37


2.2 <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

and oligodendrocytes. NS cells don’t express the pluripotent specific transcrip-<br />

tion factors Oct-4 and Nanog, but rather show expression <strong>of</strong> neural genes and<br />

lack the expression <strong>of</strong> mesoderm and endoderm specific genes. As shown in<br />

figure 2.3, pure NS cells can be derived from ES cells taken from the inner cell<br />

mass (ICM) <strong>of</strong> the blastocyst or from the SVZ and germinative area <strong>of</strong> adult<br />

and fetal brain tissue, respectively [400,403]. NS cells can be isolated from the<br />

Figure 2.3: Sources <strong>of</strong> NS cells: (a) cultured indirectly starting from ES cells<br />

derived from the ICM <strong>of</strong> the blastocyst; (b) cultured directly from the dissociation<br />

<strong>of</strong> germinative areas <strong>of</strong> the fetal brain or SVZ <strong>of</strong> the adult brain. Adapted from<br />

Pollard et al 2007 [400].<br />

embryonic or adult mammalian brain <strong>of</strong> mice [297,427,525], primates [171] and<br />

humans [135,145,149,436,443,512], although their precise purification remains<br />

elusive since they cannot yet be unambiguously identified with markers. Until<br />

recently, NS cells were mainly defined by the expression <strong>of</strong> Nestin, a cyto-<br />

plasmic intermediate filament protein discovered by Hockfield and McKay in<br />

1985 [195], although it is now clear that Nestin identifies neural progenitors as<br />

well as stem cells. Nestin expression is lost in vitro with differentiation <strong>of</strong> NS<br />

cells and, in vivo, is retained postnatally only in proliferative zones. Direct iso-<br />

lation <strong>of</strong> NS cells from human fetal brain using flow cytometry for the cell sur-<br />

face marker Prominin-1 (CD133) was reported by Uchida et al. in 2000 [502].<br />

CD133 was originally shown to be a hematopoietic stem cell marker, but it is<br />

also expressed in the SVZ <strong>of</strong> developing mice and humans on the apical mem-<br />

brane <strong>of</strong> cells lining the lateral ventricles, with more restricted expression than<br />

Nestin. In vitro NS cell-like cells with a marked stem cell activity have been<br />

isolated from human fetal brain cells expressing CD133 [122]. In vivo, NS cells<br />

have been identified as radial glial cells, expressing markers BLBP, GLAST<br />

38


2.2 <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

and RC2, an antibody recognising the presence <strong>of</strong> Nestin [310]. Until the late<br />

1990s, the only cell line that could consistently generate human neuronal cells<br />

in vitro was NTERA-2, a teratocarcinoma 28 derived cell line that required the<br />

performance <strong>of</strong> complex manipulations to induce differentiation [22,397]. In<br />

1997, Sah et al. established the first immortalized adherent human fetal neu-<br />

ral precursor cell line using retrovirally expressed avian v-myc [441] that led<br />

to subsequent independent reports using similar strategies [114,145,513], until<br />

the possibility <strong>of</strong> expanding human fetal neural precursors in suspension cul-<br />

tures was explored [83,428,483]). The floating aggregates <strong>of</strong> cells were termed<br />

"neurospheres" [427] and only recently adherent monoculture protocols have<br />

been developed as an alternative.<br />

Neurospheres With the exception <strong>of</strong> ES cells, it has always proven difficult<br />

to obtain homogeneous propagation <strong>of</strong> stem cell cultures ex vivo since they tend<br />

to be accompanied by differentiation. In 1992 Weiss and Reynolds discovered<br />

that cells from fetal mouse CNS could be propagated in suspension culture<br />

with EGF, as a cluster <strong>of</strong> floating cells that they termed "neurospheres" (Fig<br />

2.4). A neurosphere represents a clonal single cell-derived floating cluster <strong>of</strong><br />

Figure 2.4: (a,b) Contrast microscopy images <strong>of</strong> early phase neurosphere formation,<br />

in which individual cells form small clusters. (c,d) Immun<strong>of</strong>luorescence microscopy<br />

images in which (c) EGFR (green) and the Nestin protein (red) are detected on an<br />

intact neurosphere, and (d) nuclei (DAPI staining, blue) and cell mitosis (5-bromo-<br />

2’-deoxyuridine (BrdU) incorporation, green) are detected on a frozen neurosphere<br />

section. Image adapted from [375].<br />

proliferating cells [427] that contains thousands <strong>of</strong> cells and is a mixture <strong>of</strong><br />

stem and progenitor cells, with only up to 5% stem cells [122]. The number <strong>of</strong><br />

stem cells in a neurosphere is evaluated in a clonogenic assay by determining<br />

28 A germ cell tumor.<br />

39


2.2 <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

the number <strong>of</strong> singly dissociated cells from primary spheres that can give rise<br />

to secondary spheres that, in turn, can differentiate down all three lineages <strong>of</strong><br />

the CNS (Fig 2.5) [494]. Therefore, the re-plating efficiency <strong>of</strong> neurospheres<br />

is a measure <strong>of</strong> the number <strong>of</strong> stem cells, and the size <strong>of</strong> the sphere reflects<br />

progenitor proliferative efficiency. This assay also allows for a quantitation <strong>of</strong><br />

self-renewal, which is distinct from proliferation in that self-renewal involves a<br />

cell division with a cell fate decision, so that at least one daughter cell retains<br />

the full stem cell potential <strong>of</strong> the parent cell (see Fig 3.2). A multipotent<br />

secondary sphere can only form from a stem cell. Undifferentiated neurospheres<br />

can be extensively passaged in suspension, but when plated onto an adherent<br />

substrate in serum, they differentiate into the three main neural lineages <strong>of</strong><br />

the CNS: neurons, astrocytes and oligodendrocytes [122].<br />

In their 1992 neurosphere assay Weiss and Reynolds [427] employed a serum-<br />

free culture system, whereby the majority <strong>of</strong> primary differentiated CNS cells<br />

did not survive but a small population <strong>of</strong> EGF-responsive cells were maintained<br />

in an undifferentiated state and proliferated to form clusters, that could then<br />

be dissociated to form numerous secondary spheres or induced to differentiate<br />

into the three major cell types <strong>of</strong> the CNS. After approximately seven days<br />

in growth medium containing EGF, the neurospheres isolated measured 100-<br />

200µm in diameter, were composed <strong>of</strong> 3,000-5,000 cells, and differentiated into<br />

the three primary CNS phenotypes when, as intact clusters or dissociated cells,<br />

they were plated without growth factors on an adhesive substrate (Figure 2.5).<br />

Over the past decade the use <strong>of</strong> the neurosphere assay has demonstrated that<br />

a population <strong>of</strong> cells existed in the fetal through to the adult mammalian CNS<br />

that could be isolated in culture, and exhibited the critical stem cell attributes<br />

<strong>of</strong> proliferation, self-renewal, and the ability to give rise to a number <strong>of</strong> dif-<br />

ferentiated, functional progeny [116]. The neurosphere assay has proven to be<br />

an excellent technique to isolate NS cells and progenitor cells to investigate<br />

the differentiation and potential <strong>of</strong> cell lineages. These spheres can be dissoci-<br />

ated, expanded and pooled in sufficient quantity for scientific inquiry, and lend<br />

themselves easily to sectioning for histology or immunocytochemistry applica-<br />

tions, and being cryopreserved [375]. Moreover, the recent finding that human<br />

brain tumours can be similarly cultured in neurosphere conditions generates<br />

the opportunity <strong>of</strong> understanding the stem cell hierarchy <strong>of</strong> these neoplasms<br />

(see Section 3.2) [122]. Neurospheres contain a mix <strong>of</strong> differently committed<br />

cells including radial glia, committed progenitors and differentiated astrocytes<br />

40


2.2 <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

Figure 2.5: The neurosphere assay used to study neural precursor cells in culture.<br />

<strong>Cells</strong> are first isolated from embryonic or adult brain, then cultured in serum-free<br />

conditions in the presence <strong>of</strong> EGF and FGF2 to generate floating colonies. The<br />

primary neurospheres can then be dissociated and re-plated in EGF and FGF2 to<br />

generate secondary neurospheres that can then be made to differentiate in the three<br />

primary lineages <strong>of</strong> the CNS by subtracting the growth factors in adherent conditions.<br />

Adapted from Dirks et al 2008 [122].<br />

and neurons, that, upon removal <strong>of</strong> EGF, differentiate into the three main<br />

lineages <strong>of</strong> the CNS with a strong preference towards astrocytes [107]. This<br />

heterogeneity likely provides a niche that sustains 3-4% <strong>of</strong> stem cells, raising<br />

the question as to whether it is the multipotent cells or the more differenti-<br />

ated ones within the mixed cellular environment, that give rise to the three<br />

lineages [107,400]. In vivo, the external signals such as secreted factors and<br />

cell-cell interactions mediated by integral membrane proteins and the extracel-<br />

lular matrix, control stem cell fate collectively, defining the stem cell "niche",<br />

which has a powerful effect in maintaining the balance between quiescence,<br />

self-renewal and cell fate commitment [165]. In vitro, the neurosphere prob-<br />

ably provides the right mixture <strong>of</strong> cellular environments that resembles the<br />

complex niche, sustaining NS cells in the mammalian brain. Therefore, the<br />

neurosphere discovery is invaluable in that it demonstrates the potential in<br />

the developing and adult CNS <strong>of</strong> rodents and primates, to give rise to stem<br />

cells [107].<br />

41


2.2 <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

A pro<strong>of</strong>-<strong>of</strong>-principle experiment performed by Conti et al [107] demonstrated<br />

the presence <strong>of</strong> NS cells within neurospheres, and consisted in allowing passage<br />

40 mouse neurospheres derived from fetal forebrain to attach to gelatin-coated<br />

plastic in the presence <strong>of</strong> EGF and FGF2. Since under these conditions Conti et<br />

al had already proven the generation <strong>of</strong> NS cells in adherent monoculture (see<br />

Section 2.2), the appearance <strong>of</strong> bipolar cells that were indistinguishable from<br />

NS cells and could be serially propagated as uniform RC2 + /GFAP - populations<br />

and then induced to differentiate into astrocytes or neurons, concluded that<br />

radial glia-like cells present in neurospheres give rise to NS cells in adherent<br />

culture in the presence <strong>of</strong> FGF2 and EGF. Conversely, they observed that<br />

NS cells <strong>of</strong> either ES cell or fetal brain origin readily formed neurospheres if<br />

detached from the substratum mechanically or due to overgrowth, confirming<br />

that NS cells and thus radial glia are likely the neurosphere forming stem cells,<br />

although in neurospheres they constitute only a fraction <strong>of</strong> the cell population.<br />

Analogously to the embryoid body (EB) differentiation observed in ES cell<br />

aggregates, the differentiation observed within neurospheres is presumably due<br />

to aggregation [125].<br />

An important limitation <strong>of</strong> the neurosphere culture system is that, when used<br />

to screen compounds that affect NS cell expansion, human NS cells expand<br />

more slowly in suspension culture in vitro than do their mouse counterparts,<br />

which makes quantification <strong>of</strong> cell proliferation harder due to variable cell<br />

death. A second important limitation <strong>of</strong> neurosphere assays is that it is dif-<br />

ficult to identify the precise cellular target due to the presence <strong>of</strong> restricted<br />

progenitors and differentiated cell types, and real-time monitoring <strong>of</strong> cellu-<br />

lar responses is not possible in aggregates. Finally, fusion <strong>of</strong> neurospheres is<br />

a common occurrence in suspension, which confounds analyses based solely<br />

on sphere numbers or size [404]. To summarise, the neurosphere paradigm<br />

is invaluable in that it has demonstrated the existence <strong>of</strong> progenitors within<br />

cultured tissues, but it is accompanied by several important shortcomings:<br />

· the cellular complexity created by the mixed environment is a barrier for<br />

dissecting the mechanisms responsible for the self-renewal and commit-<br />

ment processes;<br />

· the heterogeneity <strong>of</strong> the cellular population pollutes global expression<br />

pr<strong>of</strong>iling experiments and makes it hard to identify a precise cellular tar-<br />

get;<br />

42


2.2 <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

· neurospheres differentiate more promptly into astrocytes rather than<br />

neurons both in vitro and when transplanted into mice;<br />

· human NS cells cannot be properly screened for compounds affecting<br />

them because quantification is made difficult by their slow growth in<br />

suspension culture and their variable death rate;<br />

· cellular responses cannot be monitored within aggregates and fusion <strong>of</strong><br />

neurospheres can confound results based on their number and size.<br />

Therefore, a niche-independent environment is better suited for the growth <strong>of</strong><br />

stem cell cultures, in that differentiation towards a specific lineage can always<br />

be traced back to the stem cells themselves [165]. Such an environment was<br />

produced when the presence <strong>of</strong> FGF2 and EGF alone was discovered to be<br />

sufficient for the continuous expansion <strong>of</strong> NS cells in adherent conditions [107].<br />

Niche-independent NS cell derivation<br />

The key to proper usage <strong>of</strong> ES cell-based technologies is the development <strong>of</strong><br />

robust, reproducible and reliable protocols for controlling propagation and dif-<br />

ferentiation <strong>of</strong> cells, and an important goal in embryonic stem cell biology over<br />

the past 10 years has been that <strong>of</strong> developing protocols to enable the conver-<br />

sion <strong>of</strong> mouse and human ES cells to the neural lineage [400].<br />

The default model <strong>of</strong> neural induction proposes that the key event is the re-<br />

moval <strong>of</strong> BMP signaling with no positive induction required [352]. In mouse<br />

ES cells, however, positive induction is necessary for differentiation to take<br />

place, since these cells can be maintained in vitro through the addition <strong>of</strong> the<br />

Leukemia inhibitory factor (LIF) and BMP extrinsic factors, as well as in-<br />

trinsic determinants Sox2, Oct-4 and Nanog transcription factors, but require<br />

replacement <strong>of</strong> LIF and BMP to specify the direction <strong>of</strong> differentiation. It<br />

was initially thought that exposure to RA and serum 29 in suspension culture<br />

was required, provided LIF retraction, to generate neurons [38,471], although<br />

later reports showed RA was unnecessary and neural precursors could be en-<br />

riched in a serum-free basal media [367]. During neural differentiation, ES<br />

cells are believed to undergo progressive lineage restrictions similar to those<br />

observed during normal fetal development, providing a means to isolate dis-<br />

tinct neural precursor populations such as NEP cells and radial glia, as well as<br />

29 Animal derived fluid most commonly drawn from a bovine fetus that contains hormones<br />

and growth factors that allow cells in culture to proliferate.<br />

43


2.2 <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

a platform to study the molecular events involved in the transitions between<br />

precursors [400]. Based on the view that neural differentiation <strong>of</strong> ES cells in<br />

vitro does recapitulate neural development in vivo, several studies isolated an<br />

RC2 immunoreactive radial glia-like cell as the transient neural progenitor in-<br />

volved in the transition from NEP cells to neuronal and glial subtypes (Fig<br />

2.6) [302,399].<br />

Figure 2.6: Diagram to visualise the progressive lineage restriction <strong>of</strong> ES cells<br />

differentiating toward the neural phenotype in neurospheres, showing the transition<br />

<strong>of</strong> NEP cells to RC2 immunoreactive radial glia-like cells. Adapted from Pollard et<br />

al 2007 [400].<br />

Likely due to paracrine LIF signaling, many ES cell neural differentiation pro-<br />

tocols have the drawback <strong>of</strong> fostering the generation <strong>of</strong> non-homogeneous cul-<br />

tures that include contaminating populations <strong>of</strong> non-neural cells and residual<br />

ES cells [400]. This effect can be overcome by adopting a "lineage selection"<br />

strategy, in which a reporter gene or drug resistance gene is expressed as a<br />

transgene 30 under cell type specific promoter elements, such as the Sox1-GFP<br />

reporter construct for the isolation <strong>of</strong> NEP cells [32]. ES cell-derived cultures<br />

engineered to express the Sox1-GFP reporter are initially enriched in Sox1 +<br />

NEP cells but quickly differentiate to neurons and glia due to the niche environ-<br />

ment recreated in neurospheres that mimics in vivo developmental cues [400].<br />

In order to isolate cells capable <strong>of</strong> undergoing symmetrical stem cell divisions<br />

without differentiation, a niche-independent protocol was devised by Conti et<br />

al [107] that uses adherent conditions to ensure homogeneity <strong>of</strong> the stem cell<br />

population bypassing the formation <strong>of</strong> neurospheres. The unique characteris-<br />

tic <strong>of</strong> this protocol is the use <strong>of</strong> EGF in attached monolayer culture, which<br />

30 A gene that does not belong to the wild type genome sequence but can be introduced<br />

from an another organism naturally or by means <strong>of</strong> genetic engineering.<br />

44


2.2 <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

had been previously only used in the culturing <strong>of</strong> neurospheres. The proto-<br />

col was successfully applied to the production <strong>of</strong> non-immortalized NS cells<br />

in adherent monolayer from mouse and human ES cells, as well as mouse and<br />

human fetal brain [107,481], and later adapted for derivation from adult mouse<br />

brain [403], with the most recent optimisation made for drug screening appli-<br />

cations from different regions <strong>of</strong> the fetal human CNS [198]. Other protocols<br />

that describe adherent monolayer culturing <strong>of</strong> NS cells have been explored by<br />

other researchers, but their approaches are all somewhat different in that they<br />

either generate immortalised cell lines [405], are tailored to derivation from the<br />

SGZ [34], or generate non-immortalized cell lines but have limited the charac-<br />

terisation to primary cultures without demonstrating long-term stability and<br />

tripotent differentiation capacity [376,536].<br />

In the protocol described by Conti et al, prior to initiating differentiation,<br />

mouse ES cells are plated at relatively high density and cultured for 24 hours<br />

in standard ES cell medium containing LIF. To start monolayer differenti-<br />

ation, undifferentiated ES cells are dissociated and resuspended directly in<br />

N2B27 medium, a mixed formulation <strong>of</strong> basal media and supplements includ-<br />

ing insulin and lacking LIF. Under this monoculture condition, ES cells lose<br />

pluripotent status and predominantly commit to a neural fate [539]. The cul-<br />

ture is re-plated after seven days in basal medium with FGF2 alone or FGF2<br />

and EGF, during which time residual undifferentiated ES cells are eliminated<br />

because the NS-A component <strong>of</strong> basal medium does not allow for ES cell prop-<br />

agation. However, since in this media neural precursors associated into floating<br />

clusters, a lineage selection strategy was adopted (Fig 2.7). Thus, after neural<br />

commitment was induced in monolayer in Sox1-GFP reporter ES cell lines, the<br />

neural precursors were maintained adherent in N2B27 medium, and the undif-<br />

ferentiated ES cell and non-neural differentiation products were eliminated via<br />

addition <strong>of</strong> puromycin. In these reporter cell lines, Sox1 is linked via an inter-<br />

nal ribosome entry site (IRES) to a gene conferring puromycin resistance (Fig<br />

2.8), so that the transition to NEP cells is marked by GFP fluorescence and the<br />

selection <strong>of</strong> these cells can be completed via the addition <strong>of</strong> puromycin. The<br />

subsequent addition <strong>of</strong> FGF2 and EGF outgrows a population <strong>of</strong> bipolar cells<br />

called LC1, during which process the ES cell-derived NEP cells gradually ex-<br />

tinguish GFP expression and acquire the NS cell phenotype, whereby they stop<br />

expressing pluripotent markers Nanog, Oct-4 and Sox1 and gain the expression<br />

<strong>of</strong> Nestin, BLBP, Sox2 and RC2 immunoreactivity and lack the expression <strong>of</strong><br />

GFAP. These are the same markers expressed by the NS cell lines obtained<br />

45


2.2 <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

Figure 2.7: Protocol describing conversion <strong>of</strong> ES cells into immortalised NS cell<br />

lines. (a) <strong>Neural</strong> precursor differentiation is induced from ES cells in serum-free<br />

adherent monoculture by subtracting LIF and detected via the Sox1-GFP reporter<br />

transgene. Sox1 is one <strong>of</strong> the earliest neural differentiation markers. (b) After seven<br />

days cells are re-plated in basal medium in the presence <strong>of</strong> either FGF2 alone or<br />

FGF2 and EGF. (c) Residual undifferentiated ES cells are eliminated from the culture<br />

through puromycin selection. (d) <strong>Neural</strong> precursors lose GFP expression as they<br />

become Sox1 - and are re-plated in fresh medium in the presence <strong>of</strong> EGF and FGF2.<br />

They also attach and outgrow a population <strong>of</strong> bipolar cells, termed LC1. (e) Clonogenic<br />

NS cell lines are generated by plating single cells from the LC1 population.<br />

Adapted from Conti et al 2005 [107].<br />

Figure 2.8: Representation <strong>of</strong> the Sox1-GFP reporter construct used in the nicheindependent<br />

NS cell protocol.<br />

from mouse fetal and adult tissues using the same protocol. To establish the<br />

presence <strong>of</strong> clonogenic NS cells, single cells were isolated from the LC1 cul-<br />

ture and expanded as adherent cultures to show the same morphology, growth<br />

characteristics and markers <strong>of</strong> the LC1 population. Upon withdrawal <strong>of</strong> EGF<br />

and FGF2 and exposure to serum or BMP4, these NS cells differentiate into<br />

astrocytes. In contrast, removal <strong>of</strong> EGF followed by FGF2 gives rise to cells<br />

46


2.2 <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

with immunochemical and electrophysiological properties <strong>of</strong> mature neurons.<br />

Importantly, even after prolonged expansion, NS cells maintain their potential<br />

to differentiate efficiently into neurons and astrocytes in vitro as well as upon<br />

transplantation into the adult brain [107]. To promote oligodendroglial differ-<br />

entiation cells were cultured on laminin coated dishes in medium containing<br />

N2 supplement plus FGF2, PDGF and forskolin, a growth factor combination<br />

known to enhance oligodendrocyte progenitor proliferation, which efficiently<br />

differentiated NS cells into oligodendrocytes [165]. To summarise the charac-<br />

terisation <strong>of</strong> the mouse NS cell lines derived by Conti et al:<br />

· they express the astrocyte differentiation marker GFAP upon addition<br />

<strong>of</strong> serum or BMP4 and differentiate into astrocytes;<br />

· they express neuronal markers type III β-tubulin (TUBB3), Microtubule-<br />

associated protein 2 (MAP2) and ERBB2 upon removal <strong>of</strong> EGF and<br />

FGF2 (in this order) and differentiate into neurons. All NS cell lines<br />

produced in the study by Conti et al were electrophysiologically active<br />

and exhibited voltage-gated Na + and Ca 2+ conductance, typical <strong>of</strong> ma-<br />

turing nerve cells;<br />

· they show no significant decline after many passages and retain diploid<br />

chromosome content throughout late passages, maintaining intact the<br />

differentiation potential in both the neuronal and glial direction;<br />

· they do not form teratomas 31 , an important step towards the confirma-<br />

tion <strong>of</strong> the identity <strong>of</strong> NS cells. Unlike ES cells, in fact, the differentiation<br />

potential <strong>of</strong> NS cells is incapable <strong>of</strong> teratoma formation and this obser-<br />

vation was reproduced in an experiment by Conti et al [107] in which NS<br />

cells were transplanted in mouse fetal and adult brain and grafted onto<br />

mouse kidney, where they did not proliferate or give rise to teratomas.<br />

The absence <strong>of</strong> any histological evidence <strong>of</strong> unregulated proliferation or<br />

tumor formation was a clear confirmation <strong>of</strong> the identity <strong>of</strong> the NS cells.<br />

Although the NS cell lines generated via the niche-independent NS cell protocol<br />

are capable <strong>of</strong> differentiating into all three lineages <strong>of</strong> the CNS, i.e. astrocytes,<br />

oligodendrocytes and neurons, the neuronal subtypes are limited to the gen-<br />

eration <strong>of</strong> large amounts <strong>of</strong> GABAergic neurons 32 according to the results by<br />

31 Encapsulated germ cell tumor derived from pluripotent cells with tissue or organ components<br />

resembling normal derivatives <strong>of</strong> all three germ layers.<br />

32 Neurons that release the main CNS inhibitory neurotransmitter GABA.<br />

47


2.2 <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

Conti et al [107]. This differs vastly from the neuronal subtypes identified upon<br />

direct differentiation <strong>of</strong> ES cell with no intermediate expansion <strong>of</strong> the neural<br />

progenitors, which are skewed towards the generation <strong>of</strong> large amounts <strong>of</strong> glu-<br />

tamatergic neurons 33 [55]. This inconsistency shows that in vitro expansion<br />

<strong>of</strong> NS cells may be somehow restricting the neuronal subtype differentiation<br />

capacity <strong>of</strong> these cells and further studies will have to address whether cell<br />

culture conditions can be altered for long-term expanded NS cells so that re-<br />

gional identities can be re-established [400].<br />

Previous studies have shown the derivation <strong>of</strong> glial restricted progenitors in ad-<br />

herent culture using FGF2 for survival and expansion <strong>of</strong> the cells [76,212,276,<br />

367]. However, these cultures change their properties over time and should not<br />

be considered equivalent to expanded long-term stem cell lines [403]. In the<br />

NS cell derivation from mouse ES cells described by Conti et al, the absence<br />

<strong>of</strong> EGF causes caspase 3-lead apoptosis and an immature neuronal phenotype.<br />

This phenomenon was found to be avoidable by culturing NS cells on laminin<br />

in the absence <strong>of</strong> EGF, although this addressed NS cells towards neuroblast 34<br />

commitment with differentiation upon mitogen withdrawal [290]. The NS cells<br />

obtained in the protocol by Conti et al exhibit phenotypic similarities to ra-<br />

dial glia in that they show no expression <strong>of</strong> neuronal or astrocyte antigens, but<br />

uniform expression <strong>of</strong> neural precursor markers Nestin, RC2, Vimentin, 3CB2,<br />

Lex1, Paired box gene 6 (Pax6) and Prominin (see Fig 2.2). In addition to<br />

this set <strong>of</strong> markers considered diagnostic for neurogenic radial glia, they show<br />

expression <strong>of</strong> the neural precursor markers Sox2, Sox3, and Emx2, and the<br />

transcription factors Olig2 and Mash1. The absence <strong>of</strong> Sox1 and maintenance<br />

<strong>of</strong> Sox2 is noteworthy <strong>of</strong> NS cells since the former marks all early neuroec-<br />

todermal precursors and its absence in stem cells might indicate that Sox2<br />

is playing the key role. In a study by Gomez-Lopez et al [169] the roles <strong>of</strong><br />

Sox2 and Pax6 were investigated in mouse fetal forebrain-derived NS cells,<br />

to find that conditional deletion <strong>of</strong> either gene reduced the clonogenicity <strong>of</strong><br />

these cells in a gene dosage-dependent manner. <strong>Cells</strong> heterozygous for either<br />

gene displayed moderate proliferative defects, but in the complete absence <strong>of</strong><br />

Sox2, cells exited the cell cycle with concomitant down-regulation <strong>of</strong> neural<br />

progenitor markers Nestin and Blbp, and ablation <strong>of</strong> Pax6 also caused major<br />

33 Neurons that release the main CNS excitatory neurotransmitter glutamate.<br />

34 Neuroblasts are the descendants <strong>of</strong> NS cells in the SVZ that migrate into damaged brain<br />

areas after strokes or other brain injuries to generate regionally appropriate new neurons<br />

[290]<br />

48


2.2 <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

proliferative defects. However, a subpopulation <strong>of</strong> cells was able to expand<br />

continuously without Pax6, retaining the progenitor markers but displaying<br />

an altered capacity to differentiate into astrocytes and oligodendrocytes, high-<br />

lighting the role <strong>of</strong> Pax6 beyond neurogenic competence. The findings by this<br />

study, therefore, suggest that Sox2 and Pax6 are both critical for self-renewal<br />

<strong>of</strong> differentiation-competent radial glia [169].<br />

Time-lapse videomicroscopy <strong>of</strong> the NS cells derived by Conti et al [107] also<br />

demonstrated that cell nuclei <strong>of</strong> those NS cells undergo interkinetic nuclear<br />

migration, a well-characterised feature <strong>of</strong> NEP and radial glia cells in vivo.<br />

Importantly, the mouse fetal brain, human ES cell and human fetal cortex-<br />

derived NS cells, expressed the same radial glia and neurogenic markers as the<br />

mouse ES cell-derived NS cells, although the human cells exhibited moderate<br />

levels <strong>of</strong> GFAP, consistently with the known activity <strong>of</strong> the human GFAP pro-<br />

moter in radial glia and unlike the very feeble expression observed in mouse NS<br />

cells [310,418]. Human ES cell and fetal cortex-derived NS cells also proliferate<br />

more slowly than the mouse-derived ones, like in neurospheres, and after se-<br />

quential withdrawal <strong>of</strong> EGF and FGF2, generate mixed populations <strong>of</strong> TuJ1 +<br />

neuron-like cells and GFAP + cells, with pure populations <strong>of</strong> cells with typical<br />

astrocyte morphology and intense GFAP immunoreactivity readily produced<br />

after exposure to serum [107].<br />

In a separate study, Sun et al [481] report the derivation and characterisa-<br />

tion <strong>of</strong> human NS cell lines from human fetal cortex and spinal cord using<br />

a continuous adherent procedure that is more efficient than allowing primary<br />

cells to form neurospheres and subsequently isolating NS cells, as described<br />

in the protocol by Conti et al. In the protocol developed by Sun et al, pri-<br />

mary cells are seeded onto laminin coated dishes in growth medium containing<br />

both EGF and FGF2. In these conditions cells readily attach and produce<br />

a morphologically heterogeneous population containing both Nestin + neural<br />

precursors and Tuj1 + neurons. In order to enrich for undifferentiated neural<br />

precursors, the cells are temporarily transferred onto gelatin coated dishes, in<br />

which conditions neurons and committed neuronal progenitors fail to survive.<br />

Three weeks after initial plating, the primary human culture is homogeneously<br />

Nestin + and Tuj1 - [481].<br />

Once established, human NS cells can be expanded continuously in monolayer<br />

culture where they homogeneously express immature neural precursor markers<br />

Nestin and Sox2. The long-term expansion <strong>of</strong> these cells can also occur suc-<br />

49


2.2 <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

cessfully in EGF alone, confirming previous findings with mouse NS cells [403],<br />

where addition <strong>of</strong> FGF2 is essential for initial derivation but can be dispensed<br />

with during subsequent propagation, suggesting that EGF is the major mito-<br />

gen for NS cell self-renewal, although a contribution <strong>of</strong> autocrine FGF2 is not<br />

excluded. However, neither human nor mouse primary cells produce stable cell<br />

lines unless they are exposed to FGF2 during the first 2-4 weeks after plating,<br />

suggesting that a possible contributing factor is that FGF2 may induce EGF<br />

responsiveness in NS cells. Furthermore, when human NS cells expanded in<br />

EGF only are exposed to the differentiation conditions described above, they<br />

are able to generate both neurons and glial cells [481].<br />

Immunostaining <strong>of</strong> the derived human fetal NS cells showed the expression <strong>of</strong><br />

the markers that are hallmarks for radial glial cells, including BLBP, 3CB2,<br />

GLAST, Vimentin, and GFAP, as well as neural progenitor markers Nestin,<br />

Sox2, Pax6, Olig2, and CD133, with Sox1 transiently expressed in mouse<br />

and human neural precursor cells, but not maintained in human fetal NS<br />

cells [107,302,481]. It takes, on average, one month to derive an adherent and<br />

morphologically homogeneous human NS cell population with a total num-<br />

ber <strong>of</strong> approximately 2 million cells. To the date the research article by Sun<br />

et al was published, the group had successfully derived five human brain NS<br />

cell lines: CB192, CB516, CB525, CB541, and CB660 [481]. The differentia-<br />

tion potential <strong>of</strong> these human NS cells was assessed using protocols previously<br />

developed for mouse NS cells by Conti et al [107] and Glaser et al [165]:<br />

· Neuronal differentiation was triggered by removing EGF from growth<br />

medium first and FGF2 successively. By the end <strong>of</strong> the fourth week <strong>of</strong><br />

neuronal differentiation, many cells became Tuj1 + and exhibited thin<br />

elongated processes.<br />

· To generate oligodendrocytes, cells were treated with basal medium sup-<br />

plemented with insulin-cotaining N2, forskolin, FGF2, and PDGF. From<br />

the third week, the supplements were changed to N2, PDGF, T3, and<br />

ascorbic acid, with successive withdrawal <strong>of</strong> PDGF inducing the appear-<br />

ance by the fifth week <strong>of</strong> 1-2% Olig-4 + cells bearing branched oligoden-<br />

drocyte morphology.<br />

· In the absence <strong>of</strong> EGF and FGF2 and presence <strong>of</strong> BMP4 or serum, a<br />

morphologically homogeneous astrocyte population was derived. The<br />

sole removal <strong>of</strong> EGF and FGF2 without addition <strong>of</strong> BMP or serum also<br />

led to NS cell differentiation into astrocytes but with significant cell death<br />

50


2.2 <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

ensuing. Recently it was reported that different types <strong>of</strong> human astro-<br />

cytes may express distinct GFAP is<strong>of</strong>orms: adult SVZ astroglial cells<br />

express GFAPδ, while most other astrocytes, including the derivative as-<br />

trocytes <strong>of</strong> the human NS cells developed in the protocol by Sun et al,<br />

express GFAPα [432].<br />

In the protocol described by Sun et al [481], during the first four weeks after<br />

cells are plated, primary human cells attach on laminin substrate within 24<br />

hours and start to proliferate in the presence <strong>of</strong> both EGF and FGF2, with<br />

no extended cell proliferation observed in medium containing only one <strong>of</strong> the<br />

two growth factors and the subsequent impossibility <strong>of</strong> establishing NS cell<br />

lines successfully. In addition to EGF and FGF2, the laminin substrate is<br />

important for efficient human NS cell derivation, since primary cells grown<br />

on gelatin coated or uncoated dishes easily detach and tend to form neuro-<br />

spheres resulting in slower proliferation, as described in the study by Conti<br />

et al [107]. The laminin substrate was also found to be optimal for human<br />

NS cell propagation, indicating that laminin may play important roles in reg-<br />

ulating neural cell behaviour [481]. Finally, although the human fetal-derived<br />

NS cells exhibited some features <strong>of</strong> radial glia, the artificial nature <strong>of</strong> culture<br />

environments may result in unique cell populations in vitro, which may indi-<br />

cate, in turn, that NS cells do not have direct in vivo counterparts. In fact, as<br />

a further consideration, the combination <strong>of</strong> transcription factor expression in<br />

mouse NS cells is not routinely observed during normal development [403,481].<br />

In order to shed the light on the nature <strong>of</strong> NS cells and their relationship<br />

to endogenous cell types, Pollard et al [403] have investigated whether cells<br />

capable <strong>of</strong> giving rise to NS cell cultures are restricted to developmental stages<br />

or may also be present in the mouse adult brain. Although both FGF2 and<br />

EGF were necessary for the derivation in culture <strong>of</strong> NS cells from adult mouse<br />

forebrain (containing the adult SVZ), once established, the stem cell lines could<br />

be maintained in added EGF alone. As already seen in mouse ES cell-derived<br />

NS cell cultures, the absence <strong>of</strong> EGF causes the majority <strong>of</strong> the NS cells to die<br />

<strong>of</strong> caspase 3-activated apoptosis and the rest to start differentiating towards<br />

the neuronal lineage, although never reaching the fully mature phenotype (Fig<br />

2.9). On the contrary, withdrawal <strong>of</strong> FGF2 did not result in any striking<br />

change in NS cell morphology or behaviour, with the exception <strong>of</strong> a slightly<br />

lower doubling time for EGF-only grown colonies possibly due to a higher<br />

cell death, although more detailed analysis are required [403]. For complete<br />

51


2.2 <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

differentiation to neurons, NS cells had to be plated on laminin-coated plates<br />

in basal medium withdrawing first EGF and then FGF2 in this order, since so<br />

long as EGF and FGF2 are supplied together no cell differentiation can occur<br />

and cells continue to self-renew. For differentiation to astrocytes, NS cells were<br />

exposed to 1% serum without EGF and FGF2 or exposed to BMP4 and LIF<br />

[399]. For differentiation to oligodendrocytes, the culture conditions involved<br />

re-culturing on laminin coated dishes in medium containing FGF2, PDGF<br />

and forskolin first, adding T3 and ascorbic acid later, a procedure known to<br />

promote the differentiation and survival <strong>of</strong> oligodendrocytes [165]. Under these<br />

conditions, NS cells efficiently differentiated into oligodendrocytes, astrocytes<br />

and neurons, with similar outcomes seen for NS cells derived from fetal mouse<br />

brain, altogether demonstrating that oligodendrocyte progenitor proliferation<br />

and differentiation seems to be preserved in adult mouse-derived NS cells.<br />

Importantly, the ability for oligodendroglial differentiation is maintained after<br />

transplantation. Therefore, the differentiation spectrum <strong>of</strong> these NS cells is<br />

not restricted to neurons and astrocytes but also extends to oligodendrocytes<br />

[165,399].<br />

Figure 2.9: Roles <strong>of</strong> EGF and FGF2 in the derivation and maintenance <strong>of</strong> NS<br />

cells. Fetal forebrain progenitors or ES cell-derived neural progenitors (green) can<br />

be converted into NS cell lines (yellow) using a combination <strong>of</strong> EGF with FGF2.<br />

Once established, NS cells can be maintained in added EGF alone, whereas in FGF2<br />

alone, they undergo differentiation (blue) and apoptosis. Adapted from Pollard et al<br />

2006 [403] and Conti et al 2005 [107].<br />

52


2.2 <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

In light <strong>of</strong> the successful engraftment into adult mouse brain [107], the ES<br />

cell or primary CNS tissue-derived NS cells (fetal and adult) show the poten-<br />

tial for delivery <strong>of</strong> cell replacement and gene therapies. Furthermore, since<br />

homogeneous expansion <strong>of</strong> any stem cell in defined conditions has been until<br />

now a prerogative <strong>of</strong> ES cells, these NS cells provide an accessible system for<br />

characterisation, manipulation and analysis <strong>of</strong> the stem cell state, as well as a<br />

resource for direct comparison with ES cells [107]. A summary <strong>of</strong> commonali-<br />

ties and differences <strong>of</strong> lineage-restricted and pluripotent stem cells is shown in<br />

table 2.1.<br />

Table 2.1: Summary <strong>of</strong> commonalities and differences between ES cells and NS<br />

cells. Adapted from Pollard et al 2006 [399]<br />

Features ES cells NS cells<br />

Species Rodent and primate Rodent and primate<br />

Source Blastocyst ES cells, germinate areas<br />

<strong>of</strong> fetal brain, SVZ <strong>of</strong><br />

adult brain<br />

Growth factor dependance LIF+BMP (serum free) EGF+FGF2 (serum free)<br />

Expansion in vitro Immortal Immortal<br />

Clonogenic Yes Yes<br />

Doubling time 12 hours 24 hours<br />

<strong>Stem</strong> cell divisions Symmetrical Symmetrical<br />

Karyotype Stable diploid Stable diploid<br />

Niche dependence None None<br />

In vivo counterpart Similarities to ICM Similarities to radial glia<br />

Potency Pluripotent Multipotent<br />

Genetic manipulation Yes Yes<br />

Neurosphere versus adherent culture method Since NS cells <strong>of</strong>fer a po-<br />

tential source for cell and tissue replacement therapy and for drug discovery,<br />

but no specific markers have been identified so far that distinguish them from<br />

their more differentiated progenitor cells, both NS cells and their progenitor<br />

cells have been isolated in a recent study by Sun et al [479] from embryonic<br />

or adult brain and spinal cord, and maintained in suspension culture as neu-<br />

rospheres as well as in adherent substrate-bound culture. The side-by-side<br />

comparison <strong>of</strong> these two culture systems was called for by the necessity to<br />

understand how processes such as cell survival, proliferation, differentiation<br />

and passaging performed in each system for potential use in drug discovery<br />

53


2.2 <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

and cell therapy. For example, the neurosphere culture method has been used<br />

extensively to study molecular and cellular mechanisms that control neuro-<br />

genesis, differentiation and cancer proliferation [116,261], but a recent study<br />

showed that long-term neurosphere cultures induce changed differentiation and<br />

self-renewal capacities, and the occurrence <strong>of</strong> chromosomal instability [518].<br />

Similarly, an earlier study by Pollard et al [401] had established that caution<br />

should be exercised also when extrapolating in vitro adherent monolayer NS<br />

cell culture findings to in vivo development because FGF2 induces a subset <strong>of</strong><br />

cell surface markers that are not found in vivo. In this study, microarray-based<br />

expression pr<strong>of</strong>iling was used to identify a set <strong>of</strong> markers expressed by fetal<br />

mouse NS cells but not ES cells and found the cell surface protein CD44 to<br />

be differentially expressed with higher expression levels in fetal mouse-derived<br />

NS cells. Although CD44 was expressed homogeneously by all NS cell lines<br />

derived in this study, appreciable numbers <strong>of</strong> CD44 + cells could not be found<br />

in the developing brain during neurogenic stages, nor in differentiating ES cell<br />

cultures. Moreover, CD44 expression was found to be induced by FGF2 in<br />

a subset <strong>of</strong> cells in primary culture and this effect did not appear to be re-<br />

stricted to CD44, with many other NS cell markers found to be activated in<br />

vitro, including Adam12, Cadherin20, Cx3cl1, EGFR, Frizzled9, Kitl, Olig1,<br />

Olig2 and Vav3. Therefore, it was speculated that the self-renewing NS cell<br />

state may be generated in vitro following transcriptional resetting induced by<br />

FGF2 and the latter did not act simply to maintain and expand pre-existing<br />

stem cells, but also impart significant changes to the transcriptional and cel-<br />

lular phenotype [401].<br />

Since the ability <strong>of</strong> NS cells to self-renew and differentiate into the three ma-<br />

jor neural cell lineages make them ideal candidates to treat impaired cells<br />

and tissues in neurodegenerative diseases, spinal cord injuries and stroke [544],<br />

methods that can scale up their production need to be evaluated accurately.<br />

Thus, the study by Sun et al [479] aimed at characterising the proliferative,<br />

differentiation and passaging capacities <strong>of</strong> the two culture methods <strong>of</strong> election<br />

for culturing NS cells, i.e. the neurosphere and adherent monolayer culture<br />

methods. The starting material for this study was dissociated from E14 rat<br />

cortical neuroepithelium, early enough in the development <strong>of</strong> the CNS that<br />

proliferating NEP cells and NS cells coexist with their neuronal, neuroglial,<br />

and glial progenitors, as well as newly post-mitotic cells (Fig 3.1). These cells<br />

were expanded in serum-free media in the presence <strong>of</strong> the mitogenic growth fac-<br />

54


2.2 <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

tors FGF2 and EGF as described by several previous studies [107,165,403,481].<br />

In their study, Sun et al [479] have shown that NS cells and their progenitors<br />

grew on adherent substrate significantly faster in the first four passages than<br />

the NS cells in neurospheres, but slowing down and plateauing their growth<br />

rate after the sixth or seventh passage, while in neurospheres their growth kept<br />

increasing slowly but steadily for more than 10 passages.<br />

Immunocytochemical analysis using multiple lineage-specific antibodies in early<br />

primary cultures for both neurosphere and adherent methods showed about<br />

60% MAP2 + neurons and about 40% Nestin + progenitors, indicating that<br />

both cultures were initially composed <strong>of</strong> neural progenitor and mature neu-<br />

ronal populations. However, proliferating neural progenitors became dominat-<br />

ing in adherent culture over the next few days to finish <strong>of</strong>f on the ninth day<br />

with a Nestin + progenitor percentage <strong>of</strong> 93.76%, clearly identifying the type<br />

<strong>of</strong> cells that eventually compose the culture [479]. In order to test for the<br />

ability <strong>of</strong> both culture systems to differentiate into neuronal and glial cells,<br />

growth factor withdrawal was induced for seven days and differentiation was<br />

assessed through the use <strong>of</strong> antibodies against Tuj1, GFAP and Olig-4 for the<br />

identification <strong>of</strong> neurons, astrocytes and oligodendrocytes, respectively. The<br />

data gathered from passages 1, 3 and 5 revealed no significant differences in<br />

the differentiation potential <strong>of</strong> cells cultured with either method [479].<br />

Self-renewal capacity was assessed in the two culture systems through a clono-<br />

genic assay that evaluated the capacity <strong>of</strong> primary neurospheres to form sec-<br />

ondary neurospheres, as well as the capacity <strong>of</strong> plating from a single cell from<br />

the original adherent culture. The clone forming rates at passage 1 were found<br />

to be 63% for neurospheres and 44% for adherent culture, a number signifi-<br />

cantly higher for neurospheres than the 20% reported by Reynolds et al [525].<br />

Interestingly, as discussed in the study by Marten et al [319], in the fetal<br />

mouse forebrain geminal zone there seem to be two differently responding self-<br />

renewing cells that grow neurospheres with greater diameters in the presence<br />

<strong>of</strong> EGF alone in their culture in vitro than FGF. Since in the culture systems<br />

developed by Sun et al both EGF and FGF were added, the two differently<br />

responsive stem cells might have been produced together, increasing the clone<br />

forming rates observed [479].<br />

Finally, neurosphere cultures were found to have a greater passage potential<br />

than adherent cultures, possibly due to the three-dimensional reproduction <strong>of</strong><br />

the niche environment experienced by the cells in vivo. In fact, the regulatory<br />

55


2.2 <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

signals from a niche are provided by soluble factors and the ECM, and neuro-<br />

spheres are spheroid structures that consist <strong>of</strong> cells producing their own ECM<br />

molecules, such as laminins, fibronectin, chondroitin sulphate proteoglycans,<br />

as well as growth factors and beta integrins, epidermal growth factor recep-<br />

tors, and cadherins. Therefore, the cell-cell and cell-matrix interactions in the<br />

three-dimensional structure <strong>of</strong> neurospheres can create an environment that is<br />

more physiologically relevant than the two-dimensional one in adherent culture<br />

systems. Even so, the robustness <strong>of</strong> cell proliferation in adherent cultures is<br />

probably due to the fibronectin and laminin substrates, in that fibronectin is<br />

a ubiquitous component <strong>of</strong> various types <strong>of</strong> ECMs, and laminin was reported<br />

as one <strong>of</strong> the five different substrates regulating neural differentiation <strong>of</strong> hu-<br />

man ES cells, and precisely stimulating their expansion in a dose dependent<br />

manner [303]. In the light <strong>of</strong> the increasing evidence that astrocytes [210] and<br />

endothelial cells [354,452] might also be important components <strong>of</strong> a niche for<br />

NS cells on top <strong>of</strong> the ECM, the adherent culture system needs to be optimised<br />

in this regard.<br />

56


Chapter 3<br />

Brain Cancer <strong>Stem</strong> <strong>Cells</strong><br />

Contents<br />

3.1 The Cancer <strong>Stem</strong> Cell Hypothesis . . . . . . . . . . . . . . 57<br />

3.2 Brain Cancer <strong>Stem</strong> <strong>Cells</strong> . . . . . . . . . . . . . . . . . . . 63<br />

3.3 <strong>Glioma</strong> Culture Systems . . . . . . . . . . . . . . . . . . . . 72<br />

3.1 The Cancer <strong>Stem</strong> Cell Hypothesis<br />

The cancer stem cell hypothesis proposes that organisation <strong>of</strong> cell lineage in<br />

tumours is hierarchical and only a subpopulation <strong>of</strong> cells termed "cancer stem<br />

cells" is responsible for tumour expansion. According to this hypothesis, stem<br />

cells or cells that acquired the ability to self-renew, accumulate genetic changes<br />

over long periods <strong>of</strong> time, escape from the control <strong>of</strong> their environment, and<br />

give rise to cancerous growth. One <strong>of</strong> the postulations <strong>of</strong> the cancer stem cell<br />

hypothesis is that a population <strong>of</strong> cells with stem cell-like features exists in tu-<br />

mours and this population gives rise to the bulk <strong>of</strong> the tumour cells with more<br />

differentiated phenotypes [123,457]. Two types <strong>of</strong> cells could fulfill the role <strong>of</strong><br />

tumour-initiating cell: adult stem cells or their progenitors, which normally<br />

undergo limited numbers <strong>of</strong> cell divisions and aberrantly acquire the capacity<br />

to self-renew by accumulating genetic lesions and subsequently becoming the<br />

long-lived target (Fig 3.1). Regardless <strong>of</strong> the cell <strong>of</strong> origin, the cancer stem<br />

cell is defined by its stem cell-like properties [486]. Normal adult stem cells<br />

are attractive candidates because they are tissue-specific, they can self-renew<br />

and, finally, they can differentiate into all cell types <strong>of</strong> the tissue <strong>of</strong> origin.<br />

Each division produces at least one daughter cell that maintains the indefinite<br />

capacity for cell division and a progenitor cell that has finite division capacity,<br />

ultimately differentiating into the mature cell types that constitute specific<br />

57


3.1 The Cancer <strong>Stem</strong> Cell Hypothesis Introduction<br />

Figure 3.1: <strong>Stem</strong> cell differentiation hierarchy, in which a possible increase in plasticity<br />

is highlighted that may be present within cancer populations. Such plasticity<br />

would be the enabler for the bidirectional interconvertibility between cancer stem<br />

cells and non-cancer stem cells. Adapted from Gupta et al 2009 [182].<br />

tissues. This type <strong>of</strong> cell division is referred to as "asymmetrical division" or<br />

"asymmetrical self-renewal" and is specifically characterised by adult stem cell<br />

divisions that produce a new adult stem cell together with a non-stem cell sis-<br />

ter that becomes the progenitor for short-lived, differentiating, functional cells<br />

that in most cases mature to a terminal division arrest [420]. For the vast ma-<br />

jority <strong>of</strong> adult tissues, it remains unclear how stem cells succeed in maintaining<br />

a precise balance between proliferation and differentiation in steady-state. To<br />

explain their long-term viability, it has been argued that tissue stem cells are<br />

maintained in a long-lived quiescent state, with most divisions supported by<br />

differentiating progenitor cells that ultimately exit the cell cycle and are re-<br />

placed by stem cell progeny, which provides a mechanism for protecting stem<br />

cells from damage and loss throughout adult life (Fig 3.2). Clear evidence<br />

for asymmetric stem cell divisions is found in invertebrates C. elegans and D.<br />

melanogaster, although in recent lineage-tracing studies in mammals it was<br />

shown that stem cells behave as an equipotent population, in which the bal-<br />

ance between proliferation and differentiation is achieved through frequent and<br />

stochastic stem cell loss and replacement [240]. While the asymmetrical self-<br />

renewing properties <strong>of</strong> adult stem cells are particularly important for homeo-<br />

static control in adult tissues that undergo continuous cellular turnover such<br />

as epithelium and blood [457,486], the non-stem cells <strong>of</strong> most adult tissues go<br />

through a rapid cycle in which they are born, mature, expire, and are removed<br />

from the tissue by apoptosis. The fact that the rate <strong>of</strong> adult cellular turnover<br />

is much faster than that <strong>of</strong> tumour development is the basis for the hypothesis<br />

58


3.1 The Cancer <strong>Stem</strong> Cell Hypothesis Introduction<br />

Figure 3.2: (a) Asymmetric cell division, in which each stem cell (orange) generates<br />

one daughter stem cell and one daughter destined to differentiate (green). (b-c)<br />

Population strategies that provide dynamic control over the balance between stem<br />

cells and differentiated cells, a capacity that is necessary for repair after injury or<br />

disease. In this scheme, stem cells are defined by their "potential" to generate both<br />

stem cells and differentiated daughters, rather than their actual production <strong>of</strong> a<br />

stem cell and a differentiated cell at each division. (b) Symmetric cell division: each<br />

stem cell can divide symmetrically to generate either two daughter stem cells or two<br />

differentiated cells. (c) Combination <strong>of</strong> cell divisions: each stem cell can divide either<br />

symmetrically or asymmetrically. Adapted from Morrison et al 2006 [345].<br />

that non-stem cells cannot effectively initiate cancers, a concept that contrasts<br />

with the commonly held idea that cancers may arise from any tissue cell with<br />

equal likelihood [420]. Although adult stem cells are attractive candidates to<br />

fulfill the role <strong>of</strong> tumour-initiating cells, two <strong>of</strong> their properties are also con-<br />

sidered limitations to that end: firstly, asymmetric division potentially limits<br />

the number <strong>of</strong> stem cells and therefore the incidence with which they could<br />

drive tumourigenesis, and secondly, the immortal DNA strand co-segregation 35<br />

process reduces the rate at which they can accumulate mutations. Through<br />

the DNA strand co-segregation molecular manoeuvre, adult stem cells reduce<br />

their mutation rate by more than 1000-fold, avoiding all mutations that arise<br />

from replication errors that are not properly repaired [420].<br />

Since all stem cells alike must self-renew and regulate the relative balance be-<br />

tween self-renewal and differentiation, and cancer can be considered a disease<br />

<strong>of</strong> unregulated self-renewal, understanding the regulation <strong>of</strong> normal stem cell<br />

self-renewal is fundamental to understanding the regulation <strong>of</strong> cancer cell pro-<br />

liferation [458]. By maintaining at least some <strong>of</strong> the properties <strong>of</strong> their tissue<br />

35 Non random segregation <strong>of</strong> the set <strong>of</strong> chromosomes with the oldest template <strong>of</strong> the DNA<br />

strands operated at each cell division.<br />

59


3.1 The Cancer <strong>Stem</strong> Cell Hypothesis Introduction<br />

<strong>of</strong> origin, cancer stem cells give rise to tumours that phenotypically resemble<br />

their origin, either by morphology or by expression <strong>of</strong> tissue-specific genes.<br />

However, what distinguishes cancerous tissue from normal tissue is the loss<br />

<strong>of</strong> homeostatic mechanisms that maintain normal cell numbers, and much <strong>of</strong><br />

this regulation normally occurs at the stem cell level. The cancer stem cell<br />

hypothesis raises the important experimental implication that if a population<br />

<strong>of</strong> biologically unique cancer stem cells exists, then tumour cells lacking stem<br />

cell properties will not be able to initiate self-propagating tumours, regardless<br />

<strong>of</strong> their differentiation status or proliferative capacity, which has an impact<br />

on the experimental definition <strong>of</strong> cancer stem cell. Furthermore, the cancer<br />

stem cell hypothesis raises a clinical implication that curative therapy will re-<br />

quire complete elimination <strong>of</strong> the cancer stem cell population, since patients<br />

who show an initial response to treatment may ultimately relapse if even a<br />

small number <strong>of</strong> cancer stem cells survive. On the other hand, targeted ther-<br />

apies that eliminate the cancer stem cell population <strong>of</strong>fer the potential for a<br />

cure [486].<br />

The concept <strong>of</strong> cancer stem cell initially arose from the observation that cancer<br />

tissues resembled developing tissues and self-renewal mechanisms were com-<br />

mon to cancer cells and stem cells. The definitive demonstration <strong>of</strong> cancer stem<br />

cells in human neoplasia was first made in 1994 in leukemia [122], a non-solid<br />

tumour found harbouring a stem cell hierarchy pattern, in which a minority<br />

<strong>of</strong> cells within the leukemic population possessed extensive proliferation and<br />

self-renewal capacity not found, however, in the rest <strong>of</strong> the leukemic cells.<br />

The putative cancer stem cells in leukemia were isolated and characterised<br />

on the basis <strong>of</strong> their phenotypical similarities to normal hematopoietic stem<br />

cells. The principle <strong>of</strong> uncovering significant similarities between putative can-<br />

cer stem cells and normal stem cells, was then extended to solid tumours, first<br />

in breast cancer then in brain cancer, although normal stem cells, their differ-<br />

entiation hierarchy and markers that identify them, are not well characterized<br />

in most solid organs. Since then, other tissues in which the connection be-<br />

tween stem cells and cancer has been found are mammary gland [15,288,289],<br />

gut [177,395,414], skin [75], bladder [89] and prostate [103] .<br />

So far, cancer stem cells have been defined on the basis <strong>of</strong> their ability to seed<br />

tumours in animal hosts, to self-renew and to generate non-cancer stem cell<br />

differentiated progeny. Accordingly, the number <strong>of</strong> cancer stem cells within<br />

a population <strong>of</strong> cancer cells can be measured by the number <strong>of</strong> cells that are<br />

60


3.1 The Cancer <strong>Stem</strong> Cell Hypothesis Introduction<br />

required, at limiting dilutions, to seed new tumours. The pioneering studies in<br />

leukemia and later solid tumours showed that it is possible to use cell surface<br />

marker pr<strong>of</strong>iles to isolate cancer cell subpopulations that are enriched for or<br />

depleted <strong>of</strong> cancer stem cells. Subsequent reports showed that, after implanta-<br />

tion in vivo, cancer stem cell-enriched populations generate tumours that are<br />

no longer enriched for cancer stem cells, implying that the cancer cells within<br />

a single tumour are naturally found in multiple states <strong>of</strong> differentiation with<br />

distinct tumour-seeding properties. The stemming possibility <strong>of</strong> bidirectional<br />

interconversion between cancer stem cell and non-cancer stem cell populations<br />

does not undermine the cancer stem cell hypothesis, since the two populations<br />

always retain their distinct identities and they can be distinguished phenotyp-<br />

ically and functionally at any moment (Fig 3.1) [182].<br />

One <strong>of</strong> the emerging caveats <strong>of</strong> the cancer stem cell hypothesis is the actual<br />

frequency <strong>of</strong> cancer stem cells. In normal tissues, somatic stem cells are inher-<br />

ently rare, and most <strong>of</strong> this type <strong>of</strong> data in cancer is derived from xenograft<br />

experiments, in which the frequency <strong>of</strong> human cancer stem cells is determined<br />

upon transplantation into a mouse environment. Differences between human<br />

and mouse stromal and support cells, cytokines as well as distinct levels <strong>of</strong><br />

immunological function in different immunodeficient recipient mouse strains,<br />

have led to conflicting frequencies <strong>of</strong> cancer stem cells ranging between 1% and<br />

25%. This discrepancy has highlighted that the stem cell properties <strong>of</strong> cancer<br />

stem cells are inherent but might also be the result <strong>of</strong> the interaction with the<br />

environmental milieu [393]. Thus, as suggested by recent findings, the number<br />

<strong>of</strong> cancer stem cells in a tumour may be a function <strong>of</strong> the cell type <strong>of</strong> origin,<br />

stromal 36 microenvironment, accumulated somatic mutations and stage <strong>of</strong> ma-<br />

lignant progression reached by the tumour. In fact, an early report indicated<br />

that the proportion <strong>of</strong> leukemia stem cells varies up to 500-fold between patient<br />

samples. More recent reports have suggested that relatively undifferentiated<br />

tumours at the histopathological level may contain higher proportions <strong>of</strong> cancer<br />

stem cells than their more differentiated counterparts. Furthermore, the num-<br />

ber <strong>of</strong> cancer stem cells may be different between tumour subtypes that arise<br />

from a single tissue, indicating that the cancer stem cells may be as numerous<br />

as the non-cancer stem cells in certain subtypes. However, the cancer stem cell<br />

hypothesis can be adapted to state that cancer cells can exist in at least two<br />

alternative phenotypic states with different tumour-seeding potentials, with-<br />

36 Connective tissue supporting the parenchymal cells <strong>of</strong> an organ.<br />

61


3.1 The Cancer <strong>Stem</strong> Cell Hypothesis Introduction<br />

out imposing requirements on the number <strong>of</strong> cancer stem and non-stem cells<br />

needed for different tumour subtypes or developmental stages [182]. In vitro<br />

cultures in unattached conditions promote the growth <strong>of</strong> sphere-like cell ag-<br />

gregates that are routinely used for enrichment and propagation <strong>of</strong> stem cells.<br />

When tumours such as glioblastoma were cultured this way, only CD133 + cells<br />

and not CD133 - cells, were found to successfully grow these spheres, renamed<br />

"tumour spheres", which expressed neuronal stem cell markers, showed highly<br />

tumourigenic potential and were resistant to radiation. Despite the success<br />

<strong>of</strong> suspension cultures to enrich for stem cell-like cells in glioblastoma and<br />

other tumour types found to behave similarly, it is unknown how stem cells in<br />

this in vitro simulation find the correct niche for the support <strong>of</strong> normal tissue<br />

stem cells, such as self-renewal, multipotency, proliferation, and differentiation.<br />

Therefore, radiation and drug sensitivity assays performed in sphere cultures<br />

have to be carefully designed and interpreted and comparing cells growing in<br />

adherent and suspension cultures may yield differences irrespective <strong>of</strong> cellular<br />

differentiation status [457].<br />

A major paradox <strong>of</strong> the cancer stem cell model lies in the multi-step tumour<br />

progression model, in which one precursor population <strong>of</strong> pre-malignant cells<br />

evolves via mutation into a successor population that has a phenotypic advan-<br />

tage, such as an increased resistance to apoptosis or growth-inhibitory signals.<br />

The conventional depiction <strong>of</strong> the cancer stem cell model would state that<br />

the only cells within the precursor population that are qualified to evolve into<br />

a successor population are its stem cells, since only these cells are endowed<br />

with the self-renewal capabilities that are required to generate unlimited num-<br />

bers <strong>of</strong> progeny [182]. This view, however, ignores the inherent properties<br />

<strong>of</strong> malignant cells, i.e. genomic instability and the ability to undergo rapid<br />

evolutionary changes [457]. Also, if the percentage <strong>of</strong> stem cells in the precur-<br />

sor cell population is small, then according to the cancer stem cell model the<br />

number <strong>of</strong> cells that can serve as targets for genetic evolution is just as small,<br />

and this postulation does not agree with the observation that the mutation<br />

rate required to complete cancer formation would have to be up to two orders<br />

<strong>of</strong> magnitude above the rates described so far for human tumour cells [182].<br />

Thus, a major problem with this hypothesis is that it assumes stability within<br />

the tumour and does not consider the possibility that the cancer stem cell phe-<br />

notype can be acquired. To this end, several studies have demonstrated that<br />

more differentiated cancer cells can acquire mutation or activate a transcrip-<br />

62


3.2 Brain Cancer <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

tion factor and become cancer stem cells [457]. This paradox may be resolved<br />

if the non-cancer stem cells in a precursor cell population could also serve as<br />

targets <strong>of</strong> mutation, leading to clonal succession and, therefore, tumour pro-<br />

gression [182]. For example, when tumour spheres derived from highly vascular<br />

glioblastomas were transplanted into mice, the initial tumours demonstrated<br />

low-grade glioma phenotype without any sign <strong>of</strong> angiogenesis. However, upon<br />

serial transplantation in vivo, the tumour cells developed a highly malignant<br />

phenotype with extensive angiogenesis and necrosis being present in the tu-<br />

mours. This finding highlights a well-established fact that tumour cells evolve<br />

and if more malignant and less differentiated cancer cells have growth advan-<br />

tage, they will be selected for and expanded in the tumour. Therefore, as<br />

tumours progress, the line between cancer stem cells and the rest <strong>of</strong> tumour<br />

cells might gradually become blurred and can even disappear [457].<br />

Finally, the recent finding that, depending on the tumour analysed, glioblas-<br />

toma cancer stem cells can be CD133 + or CD133 - cells, also emphasises that<br />

either we do not have good markers for cancer stem cells or all tumour cells<br />

are tumourigenic at varying degree, which brings us back to what we already<br />

know about tumours being diverse, genetically unstable, and evolving due to<br />

the intratumoural diversity <strong>of</strong> cellular genotypes and phenotypes. Especially<br />

in light <strong>of</strong> the recent finding that normal human differentiated cells can be<br />

converted into functional pluripotent embryonic stem cells by expressing the<br />

right combination <strong>of</strong> transcription factors [215,226,462], it is a possibility that<br />

more differentiated cancer cells may acquire stem cell phenotypes. This means<br />

that eradication <strong>of</strong> tumours will likely be achieved by the successful target-<br />

ing <strong>of</strong> all cancer cells using a cocktail <strong>of</strong> drugs effective against all cancer cell<br />

sub-populations and not only the stem cell type [457].<br />

3.2 Brain Cancer <strong>Stem</strong> <strong>Cells</strong><br />

For many solid tumours within the CNS evidence in support <strong>of</strong> the cancer<br />

stem cell hypothesis has emerged. In human glioblastoma, two separate stud-<br />

ies from Bao et al [40] and Piccirillo et al [392] have separately tried to discern<br />

the stem cell nature <strong>of</strong> this tumour, identified in a pioneering study by Singh<br />

et al [458]. Glioblastomas are diffuse tumours that invade normal brain tissues<br />

and frequently recur from focal masses after radiation, suggesting that only a<br />

fraction <strong>of</strong> tumour cells is responsible for regrowth, supporting the cancer stem<br />

cell hypothesis in solid tumours. The heterogeneity <strong>of</strong> glioblastoma starts at<br />

63


3.2 Brain Cancer <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

the cellular level thanks to the identification <strong>of</strong> tumour-initiating cells express-<br />

ing or not expressing Prominin as CD133 + stem cells, or more differentiated<br />

CD133 - cells that include glioblastoma progenitor cells, respectively [121,458].<br />

The study conducted by Singh et al [458] reports the identification and pu-<br />

rification <strong>of</strong> a cancer stem cell exclusively isolated within the cell fraction<br />

expressing the neural stem cell surface marker CD133, from different pheno-<br />

types <strong>of</strong> human brain tumours. Higher-grade gliomas showed an increased<br />

self-renewal capacity with respect to lower-grade gliomas and, importantly,<br />

the CD133 + cells could differentiate in culture into tumour cells that pheno-<br />

typically resembled the tumour from the patient. Neurosphere assays were<br />

used to functionally characterize the tumour cell population and identified the<br />

CD133 + cell as representing the minority <strong>of</strong> the tumour cell population. This<br />

cell lacked the expression <strong>of</strong> neural differentiation markers, was necessary for<br />

the proliferation and self-renewal <strong>of</strong> the tumour in culture, and was capable<br />

<strong>of</strong> differentiating in vitro into cell phenotypes identical to the tumour in situ.<br />

Since the identified cancer stem cell markers CD133 and Nestin were also iden-<br />

tified as defining normal NS cells, it was suggested that brain tumours can be<br />

generated from cancer stem cells that share a very similar phenotype as normal<br />

NS cells. Such identification <strong>of</strong> a brain tumour cancer stem cell, set up the<br />

research scene for the following investigations <strong>of</strong> the tumourigenic process in<br />

the CNS.<br />

In the study by Singh et al, cultures from 14 solid primary pediatric brain<br />

tumours were set to favour stem cell growth, resulting in all tumours grow-<br />

ing within the first 48 hours as clonally derived neurosphere-like clusters, the<br />

"tumour spheres", that continued to proliferate and expand the tumour cell<br />

culture over time. All primary tumour spheres were assessed for ability to<br />

form secondary tumour spheres upon re-plating, and all successfully exhibited<br />

self-renewal abilities. The frequency <strong>of</strong> secondary tumour sphere generation<br />

correlated with the tumour’s tumour clinical aggressiveness and varied accord-<br />

ing to tumour pathological subtype. Both primary and secondary tumour<br />

spheres retained the expression <strong>of</strong> the NS cell markers Nestin and CD133,<br />

failing instead to express the neural differentiation marker <strong>of</strong> election for as-<br />

trocytes, GFAP, and neurons, TUBB3 [458]. In neurosphere conditions, in<br />

fact, brain tumour cells express Nestin and CD133 but also markers <strong>of</strong> neural<br />

precursors such as Sox2, Notch and Jagged-1 [122]. By inducing differentiation<br />

in culture, Singh et al observed that the dissociated tumour spheres preferen-<br />

64


3.2 Brain Cancer <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

tially differentiated down the lineage that characterised the original tumour<br />

phenotype <strong>of</strong> the patient, losing the expression <strong>of</strong> Nestin and CD133 and gain-<br />

ing the markers <strong>of</strong> differentiation that reflected the immunophenotype <strong>of</strong> the<br />

original tumour [458]. This is a common finding amongst astrocytoma cells<br />

that are grown as neurospheres, which differentiate into GFAP + astrocytes,<br />

and glioblastoma cells, which instead differentiate into GFAP + astrocytes as<br />

well as TUBB3 neurons, suggesting that the tumours derive from a cell with<br />

multi-lineage differentiation capacity, i.e. a stem cell, and not a dedifferenti-<br />

ated astrocyte as previously thought [122]. When tumour cell cultures from<br />

the 14 pediatric tumours were sorted for CD133 expression, CD133 + tumour<br />

cells showed growth as non-adherent tumour spheres with continuous expan-<br />

sion <strong>of</strong> their cell population, while CD133 - cells adhered to the culture dishes<br />

without showing proliferation and not forming spheres, demonstrating that<br />

the stem cell properties resided in the CD133 + fraction [458]. With this study,<br />

Singh et al demonstrated that the CD133 marker can identify an exclusive<br />

subpopulation <strong>of</strong> brain tumour cells with NS cell activity, with three pieces <strong>of</strong><br />

evidence supporting this view:<br />

1. CD133 + cells generated clusters <strong>of</strong> clonally derived cells that resembled<br />

neurospheres, termed "tumour spheres";<br />

2. CD133 + cells were capable <strong>of</strong> constant self-renewal, as well as prolifera-<br />

tion;<br />

3. CD133 + cells differentiated to recapitulate the phenotype <strong>of</strong> the tumour<br />

from which they were derived.<br />

It must be noted that self-renewal and proliferation differ because the former is<br />

a cell division that must involve a cell fate decision, so that at least one <strong>of</strong> the<br />

two daughter cells retains the full stem cell potential <strong>of</strong> the parent cell (Fig 3.2).<br />

Exploring the molecular mechanisms involved in cell fate decisions <strong>of</strong> brain tu-<br />

mour stem cell divisions could have important implications in understanding<br />

clonal expansion and maintenance <strong>of</strong> these tumours [122]. Although cancer-<br />

initiating abilities clearly reside in the CD133 + fraction, not every CD133 +<br />

cell is capable <strong>of</strong> initiating sphere formation in vitro, demonstrating that not<br />

every CD133 + cell has stem cell properties [122] and implying the existence <strong>of</strong><br />

a hierarchy, which will be functionally elucidated as more surface markers for<br />

NS cells emerge and additional subpopulations are identified [458].<br />

65


3.2 Brain Cancer <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

The study by Bao et al [40] aimed at assessing the resistance <strong>of</strong> the CD133 +<br />

cells to ionization therapies caused by the more efficient and active DNA re-<br />

pair mechanisms present in these cells with respect to the rest constituting the<br />

bulk <strong>of</strong> the tumour. The observations in this study showed that after ionizing-<br />

radiation treatment <strong>of</strong> glioblastoma cells grown in vitro or as grafts in mice,<br />

the surviving fraction was enriched in CD133 + cells. These cells had the ability<br />

to reinitiate heterogeneous tumours when transplanted into other mice, thus<br />

demonstrating retention <strong>of</strong> their stem cell abilities. To assess the biological<br />

relevance <strong>of</strong> the enriched CD133 + cells, xenografts with a constant number <strong>of</strong><br />

total cells but increasing fraction <strong>of</strong> CD133 + cells were generated that showed<br />

a dose-dependent decrease in tumour latency, enhancement <strong>of</strong> tumour growth<br />

and vascularity. The successive irradiation <strong>of</strong> these xenografts demonstrated<br />

that viable tumour cells enriched for CD133 + cells could form secondary tu-<br />

mours with decreased latencies themselves, demonstrating that enrichment <strong>of</strong><br />

CD133 + cells is crucial in glioma recurrence after radiotherapy. The CD133 +<br />

tumour cells showed characteristics consistent with cancer stem cells, i.e. neu-<br />

rosphere formation, expression <strong>of</strong> neural and cancer stem cell markers CD133,<br />

Sox2, Musashi and Nestin, as well as multi-lineage differentiation with mark-<br />

ers for astrocytes, neurons or oligodendrocytes. Furthermore, CD133 + cells<br />

derived from xenografts or biopsy specimens formed neurospheres, whereas<br />

CD133 - cells rarely did. Finally, CD133 + tumour cells were highly tumouri-<br />

genic in brains <strong>of</strong> immunocompromised mice with characteristics <strong>of</strong> glioblas-<br />

toma and CD133 - cells did not form detectable tumours even when implanted<br />

with high doses <strong>of</strong> CD133 - cells, showing that CD133 + subpopulations were<br />

enriched for characteristics <strong>of</strong> cancer stem cells, including tumourigenesis in<br />

vivo [40].<br />

Although ionizing radiation damages tumour cells through several mechanisms,<br />

it kills cancer cells primarily through DNA damage, identifying DNA damage<br />

checkpoint responses as having essential roles in cellular radio-sensitivity [40].<br />

Progression through the mitotic cycle is driven by cyclin-CDK complexes,<br />

which ensure that all phases <strong>of</strong> the cell cycle are executed in the correct order.<br />

Terminally differentiated neurons cannot undergo cell cycle re-entry and CDK<br />

activity is suppressed through interactions with two main families <strong>of</strong> inhibitory<br />

proteins, the INK4 family that exhibits selectivity for CDK4 and CDK6, and<br />

the CIP/KIP family that has a broader range <strong>of</strong> CDK inhibitory activity (Fig<br />

3.3) [115]. As demonstrated in the study by Bao et al, activating phosphoryla-<br />

tion <strong>of</strong> the checkpoint proteins Ataxia telangiectasia mutated (ATM), RAD17,<br />

66


3.2 Brain Cancer <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

CDK1 and CDK2 was significantly higher in CD133 + cells than in CD133 - cells,<br />

indicating that the former show greater checkpoint activation in response to<br />

DNA damage. The fact that CD133 + glioma cells demonstrated to activate<br />

checkpoint responses to a greater extent than CD133 - cells, suggested that the<br />

resistance <strong>of</strong> CD133 + cells to ionizing radiation is due to preferential check-<br />

point activation. The finding that CD133 + cells were made less resistant to<br />

radiation if the checkpoint kinases CDK1 and CDK2, which control the pauses<br />

in cell-cycle progression that are scheduled for DNA repair to occur (Fig 3.3),<br />

were to be pharmacologically inhibited, provided the potential for a cure tar-<br />

geting the resilient stem cell mass. The CD133 + resistant population should be<br />

Figure 3.3: The cell cycle <strong>of</strong> eukaryotic cells can be divided into four successive<br />

phases: M for mitosis, S for DNA synthesis, and two gap phases, G1 and G2. In<br />

the G1 phase extracellular cues may induce either commitment to a further round <strong>of</strong><br />

cell division or withdrawal from the cell cycle into G0 to embark on a differentiation<br />

pathway. The G1 phase is also involved in the control <strong>of</strong> DNA integrity before the<br />

onset <strong>of</strong> DNA replication. During the G2 phase the cell checks the completion <strong>of</strong><br />

DNA replication and the genomic integrity before cell division starts. Adapted from<br />

Dehay et al 2007 [115].<br />

targeted with DNA checkpoint blockers to make these cells radiosensitive and<br />

thus, in one therapy cycle, potentially wipe out the entire tumour mass (Fig<br />

3.4) [123]. Together, the results exposed by Bao et al showed that CD133 +<br />

cancer cells contributed to glioma radioresistance and tumour re-population<br />

through preferential checkpoint response and DNA repair, and targeting <strong>of</strong><br />

checkpoint response in CD133 + cancer cells can overcome glioma radioresis-<br />

tance in vitro and in vivo, which may provide a therapeutic advantage to<br />

reduce brain tumour recurrence [40].<br />

67


3.2 Brain Cancer <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

Figure 3.4: Glioblastoma treatment with ionizing radiation. Following radiation,<br />

the bulk glioblastoma responds and the tumour shrinks. But CD133 + cells activate<br />

checkpoint controls for DNA repair more strongly than CD133 - cells, resisting radiation<br />

and prompting the tumour to regrow. These cells could be targeted with<br />

DNA checkpoint blockers to make them radiosensitive. Adapted from Dirks et al<br />

2006 [121].<br />

The approach taken by Piccirillo et al [392] is based on the role that BMPs<br />

detain as soluble factors that induce mature astrocyte differentiation in brain<br />

tumour-initiating cells in glioblastoma. As already mentioned, these tumour-<br />

initiating cells represent a small fraction <strong>of</strong> glioblastoma cells that belong to the<br />

CD133 pool, display self-renewal in vitro, generate a large number <strong>of</strong> progeny,<br />

are multipotent and can perpetuate across serial transplantation. Given that<br />

BMPs favour the acquisition <strong>of</strong> an astroglial fate (see Section 2.2), the study<br />

aimed to assess their effect on weakening the tumour-forming ability <strong>of</strong> CD133 +<br />

cells by prompting their differentiation into astrocytes both in vitro and in<br />

vivo [392]. This approach implies that tumour populations at least partially<br />

retain a developmental hierarchy based on stem cells and remain able to re-<br />

spond to the normal signals inducing them to mature. Treatment <strong>of</strong> cultured<br />

glioblastoma progenitor cells or CD133 + cells with BMP, reduced the size <strong>of</strong><br />

the tumours grafted into mice and prolonged the survival <strong>of</strong> the animal be-<br />

cause these cells were more mature and less invasive. Furthermore, CD133 +<br />

cells could not be recovered from these small tumours and they were also found<br />

to be incapable <strong>of</strong> serial engraftment, although some mice still died after three<br />

months from treatment, showing that presumably some cancer stem cells es-<br />

caped BMP treatment and were capable <strong>of</strong> re-iterating tumour formation (Fig<br />

3.5) [123].<br />

Amongst all BMPs assessed, BMP4 elicited the strongest effect by triggering<br />

a significant reduction in the stem-like, tumour-initiating precursors <strong>of</strong> hu-<br />

man glioblastomas. In fact, transient in vitro exposure to BMP4 abolished<br />

the capacity <strong>of</strong> glioblastoma cells to establish intracerebral grafts, produc-<br />

68


3.2 Brain Cancer <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

ing a pro-differentiation action predominantly in the astroglial direction and<br />

depleting the pool <strong>of</strong> tumour-initiating cells. Based on these results, it was<br />

inferred that the in vitro reduction in the stem-cell-like tumour-initiating cells<br />

would correspond to a similar decline in the ability <strong>of</strong> BMP4-treated cells to<br />

form tumours in vivo. By transiently exposing glioblastoma cells to BMP4,<br />

in fact, the tumour-initiating stem-like population produced a significant de-<br />

crease in the in vivo tumour-initiating ability <strong>of</strong> glioblastoma cells, since it<br />

effectively blocked the tumour growth and associated mortality in 100% <strong>of</strong> the<br />

mice subjected to intracerebral grafting. It is hypothesised that BMPs acti-<br />

vate their cognate receptors and trigger the Smad signaling cascade, causing a<br />

reduction in proliferation and increased expression <strong>of</strong> markers <strong>of</strong> neural differ-<br />

entiation, with no effect on cell viability. In fact, blocking endogenous BMP4<br />

reduces Smad signaling 37 and increases glioblastoma cell growth, perhaps by<br />

regulating the balance between proliferation and differentiation, and favouring<br />

the production <strong>of</strong> the differentiated astroglial-like cells normally found within<br />

glioblastomas. The authors surmise that BMP4 may reduce the frequency <strong>of</strong><br />

tumour-initiating stem cell-like cells by decreasing symmetric cell cycles that<br />

generate two identical cells on division, triggering differentiation <strong>of</strong> a subpop-<br />

ulation <strong>of</strong> tumour-initiating stem cell-like cells or blocking their proliferation<br />

and progeny, which, although not mutually exclusive events, could all con-<br />

tribute to reducing the tumour-initiating stem cell-like population. Thus, the<br />

signaling system constituted by BMPs and their receptors may also act as a<br />

key inhibitory regulator <strong>of</strong> tumour-initiating, stem-like cells in glioblastomas,<br />

other than controlling the activity <strong>of</strong> normal brain stem cells. Importantly,<br />

the results <strong>of</strong> the study by Piccirillo et al also identified BMP4 as a novel,<br />

non-cytotoxic therapeutic effector, which may be used to prevent growth and<br />

recurrence <strong>of</strong> glioblastomas in humans [392].<br />

Both these studies added depth to the cancer stem cell hypothesis, illustrat-<br />

ing the potential <strong>of</strong> re-examining cancer under this new light and highlighting<br />

the importance in cancer research <strong>of</strong> dissociating solid tumour samples into<br />

single cell suspensions to purify the stem cell fraction and test its response<br />

to treatment. Improved purification <strong>of</strong> the tumour, however, will be required<br />

37 A signaling system that involves the activation by membrane receptor protein kinases<br />

bound by the TGF-β ligand, <strong>of</strong> a family <strong>of</strong> receptor substrates, the Smad proteins, that<br />

assemble into multi-subunit complexes to go into the nucleus and activate transcription.<br />

This signaling pathway regulates a wide variety <strong>of</strong> cell-specific responses, depending on what<br />

is "the cellular context" that the TGFβ family members and multifunctional hormones are<br />

acting in [321].<br />

69


3.2 Brain Cancer <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

Figure 3.5: Glioblastoma treatment with BMPs. BMPs normally cause NS cells<br />

to differentiate into astrocytes. When used to treat isolated glioblastoma CD133 +<br />

cells, they weaken their tumourigenicity both in vitro and, when engrafted into mice,<br />

in vivo. The knowledge that a tumour retains a developmental hierarchy suggests<br />

that targeting different cell populations is a promising therapeutic strategy. Adapted<br />

from Dirks et al 2006 [121].<br />

because the true stem cells are probably a subpopulation <strong>of</strong> the CD133 + frac-<br />

tion, as demonstrated in the study by Bao et al by the death rates <strong>of</strong> mice<br />

after three months from treatment [123]. Another study attempting to eluci-<br />

date the role <strong>of</strong> CD133 was conducted in vivo on mice that were injected with<br />

100 to 1,000 uncultured malignant brain tumour cells purified by bead sorting<br />

for CD133 [459]. These CD133 + cells reproduced a phenotypical copy <strong>of</strong> the<br />

patient’s original tumour and were also heterogeneous, with only a minority<br />

<strong>of</strong> cells expressing CD133. This suggested differentiation in vivo, making dif-<br />

ferentiation therapy look like a real option for brain tumour treatment and<br />

denoting that brain tumours <strong>of</strong> different types are also functionally heteroge-<br />

neous for tumour-initiating ability [122].<br />

Several other studies have since then focused on the isolation <strong>of</strong> functional can-<br />

cer stem cell markers in different types and stages <strong>of</strong> brain tumours. In a study<br />

by Balenci et al [39], the mammalian IQ motif containing GTPase activating<br />

protein 1 (IQGAP1), considered to be a scaffolding protein at the intersection<br />

<strong>of</strong> several signaling pathways such as control <strong>of</strong> cell adhesion, polarization, di-<br />

rectional migration and neuronal motility, was found to be a reliable marker<br />

<strong>of</strong> Nestin + amplifying neural progenitors in rat brain. This protein is highly<br />

70


3.2 Brain Cancer <strong>Stem</strong> <strong>Cells</strong> Introduction<br />

abundant in rat and human glioma cell lines and it specifies a subpopulation<br />

<strong>of</strong> amplifying tumour cells in glioblastoma-like tumours but not in tumours<br />

with oligodendroglioma features, making it a reliable marker to distinguish<br />

oligodendroglioma from glioblastoma. These findings suggest that the ampli-<br />

fying IQGAP1 + cancer cells are closer to a multipotent progenitor cell and<br />

they represent the most aggressive cancer cell population in glioblastoma [39].<br />

In a study by Ma et al [304], the expression <strong>of</strong> the stem cell markers CD133,<br />

Nestin, Sox2 and Musashi-1 amongst others was investigated in 72 astrocy-<br />

tomas <strong>of</strong> different WHO grades to find out that the expression <strong>of</strong> these markers<br />

positively correlated with an increase in the WHO grade <strong>of</strong> the astrocytomas.<br />

Finally, in a very interesting study by Wang et al [520], the stem-cell-like<br />

CD133 + fraction from 14 human glioblastomas was shown to include a subset<br />

<strong>of</strong> vascular endothelial-cadherin (CD144)-expressing cells with characteristics<br />

<strong>of</strong> endothelial progenitors capable <strong>of</strong> maturing into endothelial cells. They<br />

conclude that a subpopulation <strong>of</strong> cells within glioblastoma can give rise to<br />

endothelial cells via a CD133 + /CD144 + endothelial progenitor intermediate<br />

included in the CD133 + cancer stem-cell-like fraction. This discovery opens<br />

up important clinical options since the strong correlation <strong>of</strong> tumour grade and<br />

neoplastic vasculature in human gliomas indicates that agents blocking the en-<br />

dothelial transition <strong>of</strong> tumour cells may provide a novel therapeutic strategy.<br />

Recognition <strong>of</strong> the forebrain SVZ astrocytes as the descendants <strong>of</strong> radial glia in<br />

the adult brain and their capacity to act as NS cells in vitro raises the prospect<br />

that this cell type might be responsible for tumour expansion, identifying it-<br />

self with the cancer stem cell population <strong>of</strong> the cancer stem cell hypothesis.<br />

Therefore, although it is not yet clear whether cancer-initiating events occur<br />

in NS cells, progenitors or differentiated cells, NS cells are attractive candi-<br />

dates. Their self-renewal abilities would allow an oncogene to more easily<br />

initiate uncontrolled proliferation, and their potential for transformation has<br />

been further considered based on the observations that human brain tumours<br />

frequently arise deep in the brain near the SVZ. In p53 -/- mice, more prolifer-<br />

ative activity is found in the SVZ and more neurospheres can be isolated from<br />

this region, suggesting an expansion <strong>of</strong> the NS cell pool, which may make the<br />

area more susceptible to neoplastic transformation [122]. Moreover, normal NS<br />

cells were found in the CD133 + population <strong>of</strong> the normal human fetal brain,<br />

again suggesting that the cell <strong>of</strong> origin <strong>of</strong> brain tumours may be a normal NS<br />

71


3.3 <strong>Glioma</strong> Culture Systems Introduction<br />

cell [458]. Much <strong>of</strong> the insight into brain tumour stem cells comes directly from<br />

NS cell research because the discovery <strong>of</strong> such stem cells in the mammalian<br />

brain has laid the foundations for designing experiments aimed at assessing<br />

whether the hierarchy based on stem cells seen in adult healthy brain also<br />

exists in human brain tumours and future investigations will hopefully clarify<br />

where the brain tumour stem cell sits along the lineage hierarchy <strong>of</strong> cells [122].<br />

In murine models for skin carcinogenesis, the tumour phenotype arising from<br />

overexpression <strong>of</strong> HRAS, a human gene involved in the regulation <strong>of</strong> cell divi-<br />

sion in response to growth factor stimulation, depended on the cell compart-<br />

ment it occurred in, with suprabasal layers yielding benign tumours and hair<br />

follicle bulge regions, the putative location for skin stem cells, yielding inva-<br />

sive carcinomas. Along these lines, different cells <strong>of</strong> origin might give rise to<br />

different types <strong>of</strong> brain tumours, with the more benignant ones arising from<br />

restricted progenitors and the more aggressive ones from stem cells or early<br />

progenitors. The cell <strong>of</strong> origin question is hampered by limited definition <strong>of</strong><br />

the normal NS cell hierarchy, especially by a lack <strong>of</strong> promoters that can spec-<br />

ify gene expression in distinct compartments <strong>of</strong> the stem cell hierarchy and<br />

<strong>of</strong> cell surface markers that can distinguish stem cells from multipotent or<br />

lineage-restricted progenitors. Nestin, Sox2 and CD133 identify NS cells and<br />

progenitors but no definite lineage-restricted progenitor cells have been identi-<br />

fied. GFAP marks a rare NS cell population as well as differentiated astrocytes,<br />

complicating the interpretation <strong>of</strong> these studies [122].<br />

3.3 <strong>Glioma</strong> Culture Systems<br />

<strong>Glioma</strong> Cancer Cell Lines<br />

The historically adopted use <strong>of</strong> cancer cell lines to delineate tumour biology<br />

and do preclinical drug screenings needs to be re-evaluated in light <strong>of</strong> the re-<br />

cently discovered stem cell component <strong>of</strong> solid tumours, in order to assess how<br />

well cancer cell lines reflect this characteristic amongst others with respect<br />

to primary tumour cultures. Phenotypic characteristics and the multitude <strong>of</strong><br />

genetic aberrations found within repeatedly in vitro passaged cancer cell lines<br />

<strong>of</strong>ten bear little resemblance to those found within the corresponding primary<br />

human tumour and to this end, the study by Lee et al [261] attempted to find<br />

a more biologically relevant model system for exploring glioma biology and for<br />

72


3.3 <strong>Glioma</strong> Culture Systems Introduction<br />

the screening <strong>of</strong> new therapeutic agents. In fact, little else is understood on the<br />

similarities <strong>of</strong> glioma stem-like cells, human NS cells and the primary tumours,<br />

besides the morphological similarities and the differentiation capacity <strong>of</strong> these<br />

cells [156,458,543]. Therefore, Lee et al undertook a series <strong>of</strong> experiments to<br />

identify and better characterise glioma tumour stem cells and their relation-<br />

ship with the primary tumour and traditional glioma cell lines [261].<br />

Firstly, most cancer cell lines including glioma, are grown in media containing<br />

serum, unlike NS cell cultures that are serum-free since serum causes irre-<br />

versible differentiation <strong>of</strong> NS cells [107,427]. In order to assess how primary<br />

tumour cells are affected by the presence or absence <strong>of</strong> serum, single cell sus-<br />

pensions <strong>of</strong> freshly resected and dissociated glioblastoma tissues were cultured<br />

under conditions optimal for propagation <strong>of</strong> normal NS cells, termed "NBE"<br />

conditions [107], as well as conditions optimal for growth <strong>of</strong> glioma cancer<br />

cell lines, termed "serum" conditions. Under these two conditions, pr<strong>of</strong>ound<br />

biological differences were found in vitro:<br />

· <strong>Cells</strong> in NBE conditions readily proliferated both as tumour spheres and<br />

as an adherent monolayer, as is seen with normal NS cells. In contrast,<br />

cells cultured in serum conditions formed a morphologically heteroge-<br />

neous monolayer that within a month became homogeneous with a mor-<br />

phology reminiscent <strong>of</strong> fibroblasts or epithelial cells.<br />

· <strong>Cells</strong> cultured in NBE proliferated at a constant rate regardless <strong>of</strong> passage<br />

number, whereas cells cultured in serum showed initial growth followed<br />

by a plateau phase, only to eventually proliferate at a much greater rate<br />

in later passages. To verify the extent <strong>of</strong> this behaviour, NBE cells were<br />

influenced by addition <strong>of</strong> serum and recapitulated the growth pattern<br />

seen in the serum cell population, while serum cells that were influenced<br />

by addition <strong>of</strong> NBE culture media nearly ceased growing. This indicated<br />

that the initial culture <strong>of</strong> primary tumour cells in serum conditions leads<br />

to pr<strong>of</strong>ound biological changes that cannot be subsequently reversed fol-<br />

lowing transition to NBE conditions.<br />

· Upon removal <strong>of</strong> EGF and FGF2 or addition <strong>of</strong> RA and serum, NBE<br />

cells stop expressing NS cell markers Nestin, Sox2, and Stage-specific<br />

embryonic antigen 1 (SSEA1) and start differentiation towards the glial<br />

and neuronal lineages, although 40% <strong>of</strong> cells co-stained for glial and<br />

neuronal markers together, suggesting that differentiation pathways in<br />

these NBE cells are not entirely normal. Serum cells expressed few or no<br />

73


3.3 <strong>Glioma</strong> Culture Systems Introduction<br />

NS cell markers and did not respond to differentiation cues such as RA.<br />

· In clonogenicity assays in which cells were tested for neurosphere forma-<br />

tion upon single cell plating, NBE cells showed clonogenic frequencies<br />

reminiscent <strong>of</strong> NS cell-derived neurospheres, whereas serum cells failed<br />

to form neurosphere-like cells when plated in NBE conditions.<br />

· The telomerase activities <strong>of</strong> NBE and serum-cultured cells differed in<br />

that, although both cells retained telomeres as determined by fluores-<br />

cence in situ hybridization (FISH), NBE cells had consistent telomerase<br />

activity regardless <strong>of</strong> passage number, whereas telomerase activity was<br />

lost when these cells were cultured in serum-containing media, consis-<br />

tent with what occurs in normal NS cells. Likewise, early passage serum<br />

cells did not have appreciable telomerase activity, which, however, they<br />

gained back in later passages <strong>of</strong> exponential growth phase.<br />

Taken together, these data demonstrate that NBE cells, as observed by the<br />

two NBE cell lines followed in the study, contain many <strong>of</strong> the self-renewal<br />

and differentiation characteristics <strong>of</strong> NS cells, whereas serum cultured cells do<br />

not [261]. Other important characteristics tested on these two cell types were:<br />

· Tumourigenic potential in vivo. Upon injection into the brains <strong>of</strong><br />

neonatal immunodeficient mice, NBE cells demonstrated retention <strong>of</strong> a<br />

tumourigenic potential independent <strong>of</strong> passage number with as low as<br />

1,000 cells, whereas serum-cultured cells at early passages did not show<br />

any tumourigenicity. When established NBE cell-derived tumours were<br />

dissociated and cultured under NBE conditions to be injected again into<br />

the brains <strong>of</strong> new recipient mice, there was no loss <strong>of</strong> tumourigenic po-<br />

tential, unlike when the same xenograft-derived cells were grown under<br />

serum conditions and all subsequent tumourigenic potential was lost.<br />

This suggested that the loss <strong>of</strong> tumourigenicity was due to the serum-<br />

culture conditions. Interestingly, although early passage serum-cultured<br />

cells did not form tumours, the late passage, exponentially growing,<br />

telomerase-positive ones did at an increasing rate with progressive pas-<br />

sage number.<br />

· Phenocopy <strong>of</strong> the original human glioblastoma tumour. While<br />

the intracranial tumours generated by NBE cells demonstrated exten-<br />

sive infiltration along white matter tracts as observed in glioblastoma<br />

patients, all the tumours generated from late passage serum cells were<br />

74


3.3 <strong>Glioma</strong> Culture Systems Introduction<br />

well delineated with little tumour cell infiltration, a characteristic pheno-<br />

typically identical to the human tumour xenografts generated from the<br />

standard glioma cell lines, demonstrating that only tumours derived from<br />

NBE cells phenocopy the critically important histopathological features<br />

<strong>of</strong> the original human glioblastoma tumours.<br />

· <strong>Transcriptional</strong> activity. The gene expression pr<strong>of</strong>iles <strong>of</strong> NBE cells,<br />

serum cells, their derived xenograft tumours, and the original glioblas-<br />

toma tumours, show that the transcriptional landscape <strong>of</strong> NBE cells<br />

and their derivative xenograft tumours is more closely related to that<br />

<strong>of</strong> NS cells, and parental tumours, while the transcriptional landscape<br />

<strong>of</strong> serum cells and their derivative xenografts is more closely related to<br />

that <strong>of</strong> glioma cell lines and their derivative tumours. These data demon-<br />

strate that NBE cells are remarkably similar to normal NS cells and their<br />

derivative tumours properly maintain many biological characteristics <strong>of</strong><br />

the parental glioblastomas and other primary glioblastomas, whereas tra-<br />

ditionally grown, serum-cultured cancer cell lines do not.<br />

· Genomic changes. These were evaluated by performing SNP analy-<br />

sis and spectral karyotyping (SKY) on NBE and serum-cultured glioma<br />

cells. Deletion <strong>of</strong> the CDKN2A:ARF locus on chromosome 9, loss <strong>of</strong><br />

chromosome 10q, trisomy <strong>of</strong> chromosome 7, and local amplification <strong>of</strong><br />

the EGFR locus are common genomic features in primary glioblastomas<br />

and they were found in all surgical samples. The serial genomic DNA<br />

pr<strong>of</strong>iles <strong>of</strong> NBE and serum cultured cells at various passage numbers were<br />

analysed by SNP analysis and showed that even after one year <strong>of</strong> main-<br />

tenance (more than 70 passages) in NBE culture condition, NBE cells<br />

largely maintained their parental tumour genotype, while serum cells<br />

underwent significant genomic rearrangements as early as two months<br />

<strong>of</strong> culture (less than 10 passages). Intriguingly, LOH in chromosomes 4<br />

and 17 found in most <strong>of</strong> the late passage serum cells coincided with the<br />

onset <strong>of</strong> increased proliferation, tumourigenicity and aneuploidy typical<br />

<strong>of</strong> these cells, and carried the hCDC4 and p53 genes, respectively. The<br />

fact that the remaining copy <strong>of</strong> hCDC4 was found to be down-regulated<br />

hinted at the possibility that this E3 ubiquitin ligase involved in the<br />

regulation <strong>of</strong> the aurora kinase STK15, up-regulated in these cells and<br />

known to cause aneuploidy through inactivation [313,416], was partially<br />

responsible for the increased proliferation, tumourigenicity, and aneu-<br />

75


3.3 <strong>Glioma</strong> Culture Systems Introduction<br />

ploidy observed in late passage serum cells. In addition, loss <strong>of</strong> the<br />

wild-type p53 allele in late passage serum cells leaves only the mutant<br />

p53 allele found in both the NBE cells and the parental tumours, caus-<br />

ing a well-known genomic instability [130,153]. These genomic changes<br />

further demonstrate the significant differences between serum cells and<br />

their matched parental tumours [261].<br />

In conclusion, by using a model system derived from primary glioblastomas,<br />

Lee et al [261] have demonstrated that NBE-cultured cells derived from pri-<br />

mary glioblastomas bear remarkable similarity to normal NS cells by retaining<br />

the ability to form neurospheres in vitro; an indefinite self-renewal potential;<br />

the ability to differentiate into the glial and neuronal lineages <strong>of</strong> the CNS; hav-<br />

ing gene expression pr<strong>of</strong>iles similar to NS cells; bearing genetic stability over<br />

many passages in vitro; harbour all <strong>of</strong> the genetic aberrations found within<br />

the primary tumour; have gene expression pr<strong>of</strong>iles similar to the glioblastomas<br />

they were derived from; appear to be the principal tumourigenic cell type that<br />

can recapitulate the overall in vivo phenotype <strong>of</strong> the parental glioblastoma.<br />

By contrast, cells derived from the same glioblastoma specimens but grown in<br />

serum-containing media lose all the characteristics <strong>of</strong> primary tumour cultures,<br />

although they ultimately regain the tumourigenic potential in later passages<br />

without being able to recapitulate the tumourigenic phenotype <strong>of</strong> the original<br />

tumour however, but rather matching the phenotypic and genotypic patterns<br />

found in most glioma cell lines.<br />

Since several groups have reported that tumour stem cell-like cells can be<br />

isolated from the established tumour cell lines by culture in serum-free me-<br />

dia with selected growth factors [242,385], experiments need to be carried out<br />

to validate how these cells maintain their tumour stem cell-like properties in<br />

differentiation-inducing conditions and if cells with stem-like properties may<br />

emerge again through epigenetic reprogramming or selection <strong>of</strong> a subpopula-<br />

tion <strong>of</strong> cells with genomic instability.<br />

Based on their findings, Lee et al propose that the inherent tumour stem cell<br />

population within primary glioblastomas is quickly lost in typical glioma cul-<br />

ture conditions, and the cells found following prolonged in vitro passages are<br />

the product <strong>of</strong> an outgrowth <strong>of</strong> a cell clone that has undergone pr<strong>of</strong>ound de<br />

novo genetic and/or epigenetic changes. This indicates that NBE cells may<br />

be an optimal model system for understanding the biology <strong>of</strong> primary human<br />

tumours, for the preclinical screening <strong>of</strong> agents and to guide personalized tu-<br />

mour therapy. The table 3.1 below summarises the findings <strong>of</strong> this study [261].<br />

76


3.3 <strong>Glioma</strong> Culture Systems Introduction<br />

Table 3.1: Summary <strong>of</strong> characteristics <strong>of</strong> NBE and serum-cultured glioblastoma<br />

cells. Adapted from Lee et al 2006.<br />

Proliferation Constant<br />

Clonogenicity,<br />

tumourigenicity<br />

Differentiation<br />

potential<br />

Telomerase activity Positive<br />

tumour histology<br />

Global gene expression<br />

NSC-related genes<br />

Genotype<br />

<strong>Glioma</strong> <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong><br />

NBE-cultured Serum-cultured<br />

Limited growth, plateau, exponential<br />

growth<br />

Yes, regardless <strong>of</strong> passages Not at early passages<br />

Induce to become glial<br />

and neuronal lineages<br />

Extensive migration, phenocopy<br />

<strong>of</strong> primary human<br />

GBMs<br />

Similar to primary human<br />

GBMs<br />

Nestin, Sox2, CD133,<br />

Musashi and Bmi<br />

Same as parental tumour<br />

regardless <strong>of</strong> passages<br />

Do not respond to differentiation<br />

stimuli<br />

Negative initially, but became<br />

positive at late passages<br />

Fail to show infiltration like<br />

glioma lines<br />

Differentiation from primary<br />

tumours but similar<br />

to common glioma lines<br />

-<br />

Additional alterations not<br />

found in parental tumour<br />

The demonstration that the adult human brain maintains areas <strong>of</strong> radial<br />

glia populations that have been shown to give rise to NS cells within the<br />

SVZ, raised the prospect that these multipotent cells could be the alternate<br />

cells responsible for glioma expansion to the differentiated glia in the brain<br />

parenchyma [122,390,399]. As rare populations <strong>of</strong> stem cells are being discov-<br />

ered in different tissues, the cancer stem cell hypothesis reinforces its statement<br />

that cell lineage organization in tumours is hierarchical rather than stochastic<br />

and only the sub-population <strong>of</strong> cancer stem cells is responsible for the expan-<br />

sion <strong>of</strong> the tumour [123,458]. Therefore, in vitro expansion <strong>of</strong> the putative<br />

brain cancer stem cells as stable cell lines would provide a powerful model<br />

system to study the human disease by giving insights into the origin <strong>of</strong> tumour<br />

heterogeneity and enable further analysis <strong>of</strong> the self-renewal, commitment, and<br />

differentiation processes, which will hopefully lead to more targeted therapeu-<br />

tic strategies [404].<br />

77


3.3 <strong>Glioma</strong> Culture Systems Introduction<br />

As mentioned in section 3.1, the neurosphere culture paradigm has been used<br />

successfully for enrichment <strong>of</strong> tumour-initiating cells from brain tumours in-<br />

cluding glioblastoma in serum-free media [156,261], an improvement on "clas-<br />

sic" serum-cultured glioma cell lines that fail to model accurately the human<br />

disease [404]. However, neurosphere culture has several limitations:<br />

· short-lived progenitor cells also proliferate in suspension culture and true<br />

clonal analysis is hampered by sphere aggregation;<br />

· the spontaneous differentiation and cell death accompanying stem cell<br />

divisions in the sphere environment limits rigorous assessment <strong>of</strong> stem<br />

cell behaviour and marker analysis based on bulk populations;<br />

· the true nature <strong>of</strong> the stem cell compartment across the spectrum <strong>of</strong><br />

gliomas and their relationship to in vivo progenitors remains unclear.<br />

Unlike neurosphere culture, adherent culture provides uniform access to growth<br />

factors, suppressing differentiation and enabling expansion <strong>of</strong> highly pure pop-<br />

ulations <strong>of</strong> stem cells. The methodology reported by Conti et al [107], Pol-<br />

lard et al [403] and Sun et al [481] in section 2.2 for deriving and expanding<br />

mouse and human adherent NS cell lines in the presence <strong>of</strong> EGF and FGF2,<br />

was adopted by Pollard et al [404] in an experiment designed to test whether<br />

these same conditions enabled the isolation and expansion <strong>of</strong> stem cells from<br />

gliomas. In these conditions, six cell lines were expanded for at least one year<br />

and more than 20 passages, without any significant alteration in growth rate<br />

or known GBM-related genetic aberrations. Cell lines were established from<br />

histopathologically distinct types <strong>of</strong> tumour:<br />

· three cases <strong>of</strong> glioblastoma multiforme: G144, G166 and GliNS2, with<br />

the glioma sample from patient number 144 established as a biological<br />

replicate in cell line G144ED, derived independently <strong>of</strong> G144 but using<br />

the same initial tumour sample. In all analyses performed, no striking<br />

differences in marker expression or behaviour were found between the<br />

two cell lines;<br />

· one case <strong>of</strong> giant cell glioblastoma: G179;<br />

· one case <strong>of</strong> anaplastic oligoastrocytoma: G174.<br />

These cell lines were phenotypically characterised by immunocytochemistry<br />

to ensure their similarity to the fetal NS cells used as normal counterpart,<br />

78


3.3 <strong>Glioma</strong> Culture Systems Introduction<br />

which confirmed coherent expression <strong>of</strong> NS cell markers and neural progenitor<br />

markers Vimentin, Sox2, Nestin, CD44 and 3CB2, and by time-lapse videomi-<br />

croscopy, which confirmed the dynamic changes in cell shape and the high<br />

motility typical <strong>of</strong> fetal NS cells, with G166 cells being less motile than G144<br />

cells. Given these similarities the cell lines were termed glioma NS cell lines<br />

or GNS cell lines [404]. In comparing the efficiency with which adherent GNS<br />

cell lines could be established compared to suspension culture in neurospheres,<br />

it was found that although most samples generated neurospheres upon initial<br />

plating, only two cell lines could be passaged further, while in adherent condi-<br />

tions the establishment <strong>of</strong> cultures was successful for all cell lines for at least 10<br />

passages, possibly due to the increased differentiation and apoptosis processes<br />

that take place in neurosphere culture [404].<br />

To determine whether the GNS cells displayed genomic alterations charac-<br />

teristic <strong>of</strong> glioblastoma, molecular cytogenetic analyses were performed us-<br />

ing SKY, locus-specific FISH, and comparative genomic hybridization, which<br />

showed G144 cultures exhibiting simple numerical gains <strong>of</strong> chromosomes 7 and<br />

19, together with loss <strong>of</strong> chromosomes 6, 8, and 15. These aberrations, with<br />

the exception <strong>of</strong> chromosome 8 deletion, are all commonly associated with<br />

glioblastoma [326,383]. Furthermore, comparative genomic hybridization for<br />

G144 revealed an amplification <strong>of</strong> the CDK4 locus on chromosome 12 and<br />

deletion <strong>of</strong> PTEN on chromosome 10. However, late passage G144 cultures<br />

did show a more complex and heterogeneous pattern <strong>of</strong> both numerical and<br />

structural chromosomal change. Similarly, G179 exhibited a more complex<br />

chromosomal constitution at later passages, and like in G144, polysomic gain<br />

<strong>of</strong> whole chromosome 7 was evident. Thus, although GNS cells do not display<br />

gross chromosome instability, alterations in whole chromosome copy number<br />

do occur following long-term in vitro expansion, an issue that can be circum-<br />

vented by routinely using cultures that are expanded for no more than 20<br />

passages [404].<br />

In order to test the capacity <strong>of</strong> GNS cells to initiate tumour formation, cells<br />

from all GNS cell lines were injected intracranially into immunocompromised<br />

mice that were sacrificed at 5 and 20 weeks, with the former scenario revealing<br />

large numbers <strong>of</strong> engrafted human Nestin immunoreactive cells and the lat-<br />

ter scenario showing the formation <strong>of</strong> large and highly vascularised tumours<br />

histopathologically similar to human glioblastoma tumours. The cellular het-<br />

erogeneity was highlighted by immunostaining the xenografts for Nestin and<br />

79


3.3 <strong>Glioma</strong> Culture Systems Introduction<br />

GFAP, and performing flow cytometry for CD133. Furthermore, transplanta-<br />

tion <strong>of</strong> these cells after in vitro exposure to differentiation-promoting conditions<br />

delayed tumour formation. In most transplants a striking infiltration <strong>of</strong> the<br />

brain reminiscent <strong>of</strong> the human disease was observable with the exception <strong>of</strong><br />

cell line G166, which generated a more defined tumour mass. To determine<br />

the tumour-initiating potential <strong>of</strong> the GNS cell lines, transplants were carried<br />

using 10-fold dilutions starting with 100 cells, which were sufficient for cell en-<br />

graftment <strong>of</strong> all GNS cells and for G144 and G174 were capable <strong>of</strong> generating<br />

an aggressive tumour mass, which required 1,000 cells in the case <strong>of</strong> G166 and<br />

G179. Unlike GNS cells, normal fetal NS cells never generated tumours at<br />

any dilution. Finally, to determine whether the tumour-initiating cells could<br />

self-renew within the xenograft, serial transplantations from the tumour mass<br />

into secondary and tertiary recipients was carried out using G144, G144ED<br />

and G179, showing in each case the generation <strong>of</strong> a tumour and thus demon-<br />

strating that long-term expanded GNS cell lines remain highly tumourigenic<br />

and are capable <strong>of</strong> forming tumours that appear to recapitulate the human<br />

disease [404].<br />

Since the important defining property <strong>of</strong> stem cells is their ability to gen-<br />

erate differentiated progeny, the differentiation programs <strong>of</strong> the GNS cells<br />

were evaluated keeping in mind that the prevalent form <strong>of</strong> glioma is the<br />

GFAP + astrocyte-like cell-containing astrocytoma, which can also contain<br />

anaplastic cell populations and, in some cases, an oligodendrocyte compo-<br />

nent [301]. For all GNS cell lines, the differentiation to Octamer-binding pro-<br />

tein 4 (Oct4) + oligodendrocytes or TuJ1 + neurons was fully suppressed in the<br />

presence <strong>of</strong> EGF and FGF2, in contrast to what is observed in glioma neuro-<br />

spheres. Upon growth factor withdrawal, NS cells differentiate into neurons<br />

(see Fig 2.9), but in contrast to that, G144 and G179 GNS cells differentiated<br />

into Oct4 + or CNPase +38 oligodendrocyte-like cells, and TuJ1 + cells, respec-<br />

tively. Neuronal-like cells or oligodendrocytes were not apparent in G166 cul-<br />

tures, which continued to proliferate in the absence <strong>of</strong> EGF and FGF2 without<br />

clear differentiation. The tendency <strong>of</strong> G144 cells to differentiate into oligo-<br />

dendrocytes was surprising because efficient oligodendrocyte differentiation <strong>of</strong><br />

mouse and human fetal NS cells requires exposure to thyroid hormone, ascor-<br />

38 CNPase is a 2Õ, 3Õ-cyclic nucleotide 3Õ-phosphodiesterase that catalyses the in vitro<br />

hydrolysis <strong>of</strong> 2Õ, 3Õ-cyclic nucleotides to produce 2Õ-nucleotides and has an in vivo function<br />

that remains to be elucidated. High CNPase expression is seen in oligodendrocytes and<br />

Schwann cells, accounting for roughly 4% <strong>of</strong> the total myelin protein in the CNS [137]<br />

80


3.3 <strong>Glioma</strong> Culture Systems Introduction<br />

bic acid, and PDGF [165]. Thus, the authors suggested that either G144 cells<br />

represented a corrupted three-directional potent state with acquired genetic<br />

changes influencing the lineage choice toward oligodendrocyte commitment,<br />

or that they may have a distinct phenotype more similar to oligodendrocyte<br />

precursor cells than NS cells. To distinguish between these two possibilities,<br />

G144 was tested for the expression <strong>of</strong> established markers <strong>of</strong> oligodendrocyte<br />

precursor cells, i.e. Olig2, Sox10, NG2, and PDGFRα prior to and during<br />

differentiation, to find out that G144 cells stably exhibited an oligodendro-<br />

cyte precursor-like phenotype expressing these markers prior to the beginning<br />

<strong>of</strong> differentiation by growth factor withdrawal. In fact, upon re-examination<br />

based on histopathology and CNPase staining, G144 was shown to have a<br />

significant oligodendrocyte component even though originally diagnosed as a<br />

glioblastoma [404].<br />

In determining whether GNS cells could generate astrocytes, G144 and G179<br />

were observed to undergo a striking change in cell morphology after seven days<br />

from addition <strong>of</strong> BMP4 and removal <strong>of</strong> EGF and FGF2, the protocol used for<br />

astrocyte differentiation <strong>of</strong> NS cells (see Fig 2.9). The majority <strong>of</strong> cells ex-<br />

pressed high levels <strong>of</strong> GFAP, although the frequency was much lower for G166,<br />

and cell lines G144 and G179 expressed low levels <strong>of</strong> the Doublecortin (Dcx) +<br />

neuronal marker. This ensemble <strong>of</strong> events showed that although GNS cells<br />

retain the capacity to differentiate, efficiency and lineage choice vary dramat-<br />

ically between each line [404].<br />

GFAP is expressed in radial progenitors and radial glia in the developing pri-<br />

mate nervous system, as well as putative NS cells within the adult SVZ and,<br />

specifically, at low levels in human fetal NS cell lines. Since an alternatively<br />

spliced form, GFAPδ, was shown to mark human SVZ astrocytes [432], the<br />

behaviour <strong>of</strong> the GNS cell lines with respect to GFAPδ was assessed. The<br />

expression level <strong>of</strong> GFAPδ mRNA was five times greater in G179 than in<br />

G144 and G166, with G179 cells also up-regulating GFAP expression following<br />

BMP treatment and G144 cells remaining, instead, predominantly negative.<br />

Co-expression <strong>of</strong> GFAPδ, Sox2, and Nestin was specific to G179 cells, which,<br />

together with the ability to generate neuronal-like cells in vitro, are all fea-<br />

tures conserved in adult SVZ astrocytes [443]. The fact that G166 cells lacked<br />

the expression <strong>of</strong> GFAPδ and oligodendrocyte precursor markers, but differ-<br />

entiated toward GFAP + astrocytes in vitro, suggested similarity to a more<br />

restricted astrocyte precursor. Thus, despite their shared capacity to prolif-<br />

erate in response to EGF and FGF2 and the widespread expression <strong>of</strong> neural<br />

81


3.3 <strong>Glioma</strong> Culture Systems Introduction<br />

progenitor markers, there are underlying differences between GNS cell lines<br />

that may reflect the existence <strong>of</strong> distinct subtypes <strong>of</strong> regular neural progeni-<br />

tors, and GFAPδ may be <strong>of</strong> use in discriminating astrocyte-like cells that have<br />

stem cell properties [404].<br />

To evaluate the relationship between each GNS cell line and their correspon-<br />

dence to fetal NS cells, mRNA expression pr<strong>of</strong>iling was carried out using mi-<br />

croarrays, and PCA 39 revealed that each GNS cell line had a transcriptional<br />

state that more closely related to fetal NS cells than to adult brain tissue (Fig<br />

3.6). Consistent with what was found about the various GNS cell lines in the<br />

study by Pollard et al [404], the PCA analysis confirmed that G179 and G166<br />

have a distinct expression pr<strong>of</strong>ile, both from one another and to G174, G144,<br />

and GliNS2. The microarray data also specifically confirmed the expression<br />

Figure 3.6: PCA <strong>of</strong> global mRNA expression in each GNS cell line (black, G144,<br />

G144ED, G166, G179, G174, and GliNS2), fetal NS cells (red, hf240, hf286, and<br />

hf289), and normal adult brain tissue (blue). The "a" and "b" are biological replicates<br />

<strong>of</strong> cell line hf240. Taken from Pollard et al 2009 [404].<br />

<strong>of</strong> the oligodendrocyte precursor cell markers Sox8, Sox10, Olig1, Olig2, and<br />

Nkx2.2 in G144, and the lower expression levels <strong>of</strong> the same markers observed<br />

in G179, which, instead, showed higher levels <strong>of</strong> GFAPδ expression. The fact<br />

that GliNS2 clustered closely with G144 and expressed the oligodendrocyte<br />

precursor markers, suggested that the G144 phenotype may not be unique. The<br />

G174 cell line, instead, derived from an oligoastrocytoma, clustered closely to<br />

GliNS2 and G144 because it expressed higher levels <strong>of</strong> Olig2, although it failed<br />

to express Sox10 or the pluripotency markers Oct4 and Nanog. Finally, G166<br />

39 A common technique to find patterns in data <strong>of</strong> high dimension and expressing the data<br />

in such a way as to highlight similarities and differences. Mathematically, it is a procedure<br />

that uses an orthogonal transformation to convert a set <strong>of</strong> observations <strong>of</strong> possibly correlated<br />

variables into a set <strong>of</strong> values <strong>of</strong> uncorrelated variables called principal components that are<br />

less than or equal to the number <strong>of</strong> original variables.<br />

82


3.3 <strong>Glioma</strong> Culture Systems Introduction<br />

showed higher levels <strong>of</strong> EGFR than any other cell line, perhaps explaining its<br />

resistance to differentiation upon EGF withdrawal or BMP treatment [404].<br />

The CD133 and CD15 cell surface markers are expressed on fetal and adult<br />

neural progenitors and brain tumour-initiating cells (see Section 3.2). For<br />

G144 and G179, heterogeneity for both markers was observed, similarly to<br />

fetal NS cells [480], whereas G166 was found to be negative consistently with<br />

the microarray expression data. The fact that CD133 was not present on<br />

all GNS cell lines confirmed that this marker does not universally identify tu-<br />

mourigenic cells in malignant glioma. Unlike CD133 and CD15, the hyaluronic<br />

acid-binding protein CD44, characterised as an astrocyte precursor marker and<br />

recently demonstrated to also mark NS cells in vitro [401], was uniformly ex-<br />

pressed in all GNS cell lines [404].<br />

To demonstrate the pro<strong>of</strong> <strong>of</strong> principle <strong>of</strong> the utility <strong>of</strong> GNS cells with respect to<br />

the shortcomings <strong>of</strong> the neurosphere assay (see Section 2.2), a small-scale chem-<br />

ical screen <strong>of</strong> known pharmaceutical drugs was carried out. Importantly, the<br />

results <strong>of</strong> this screen extended to human brain cancer stem cells the observa-<br />

tion that mouse neurospheres are sensitive to modulation <strong>of</strong> neurotransmitter<br />

pathways, and future more in depth studies will have to determine whether the<br />

drugs found in the screen that modulate the serotonin pathway <strong>of</strong> the adherent<br />

GNS cell lines, can limit growth <strong>of</strong> xenograft tumours in vivo [404].<br />

In their paper, Pollard et al [404] have tackled two critical issues <strong>of</strong> the brain<br />

cancer stem cell hypothesis:<br />

1. How to maintain and expand pure populations <strong>of</strong> cancer stem cells in<br />

vitro by expanding them as cell lines using the adherent culture methods<br />

previously established for fetal and human NS cells [107,481]. Specifi-<br />

cally, Pollard et al have demonstrated that suspension culture is not a<br />

requirement for successful long-term propagation <strong>of</strong> tumour-derived stem<br />

cells, and that expansion in adherent conditions overcomes the limita-<br />

tions <strong>of</strong> the neurosphere culture paradigm, such as increased levels <strong>of</strong><br />

differentiation and apoptosis.<br />

2. Elucidation <strong>of</strong> the phenotypic similarities between GNS cells and the en-<br />

dogenous progenitors within the developing and adult nervous system,<br />

e.g. NEP cells, radial glia, glial progenitors, oligodendrocyte progeni-<br />

tor cells, and SVZ astrocytes. For example, G144 cells strongly express<br />

markers <strong>of</strong> the oligodendrocyte precursor cell lineage and are biased to-<br />

ward oligodendrocyte differentiation, while G179 cells appear to be more<br />

83


3.3 <strong>Glioma</strong> Culture Systems Introduction<br />

similar to adult SVZ astrocytes, including expression <strong>of</strong> GFAPδ, although<br />

more specific markers are needed in order to confirm this. This perhaps<br />

indicates that the histological spectrum <strong>of</strong> different gliomas is dictated<br />

by the phenotype <strong>of</strong> the underlying tumour-initiating cells.<br />

To summarise, when the same culture conditions for NS cells were tested on<br />

glioma tumours, NS-like cells were isolated and propagated as GNS cells. Nor-<br />

mal NS cells and GNS cells feature many commonalities, both in their morphol-<br />

ogy and immunoreactivity, to radial glia markers. However, unlike NS cells,<br />

GNS cells require less exogenous growth factor for stable proliferation and reca-<br />

pitulate the pathology <strong>of</strong> the original gliomas when xenografted into immuno-<br />

compromised mice. This system is highly unusual in that normal and diseased<br />

counterparts are morphologically and immunohistologically indistinguishable<br />

and yet the differentiation behaviour <strong>of</strong> the cancer stem cells is clearly aber-<br />

rant. The data generated using this system described by Pollard et al [404] is<br />

consistent with tumour stem cells arising following transformation <strong>of</strong> oligoden-<br />

droglial precursors or adult SVZ astrocytes, although it is considered equally<br />

plausible that short-lived progenitors or differentiated cells can be converted<br />

to a stem cell state through genetic or epigenetic disruptions. In conclusion,<br />

GNS cells provide a versatile and renewable resource to probe the biology <strong>of</strong><br />

tumour-initiating cells and screen for agents that selectively and directly target<br />

them. tumour stem cell self-renewal, migration, apoptosis, and differentiation<br />

all represent potential therapeutic opportunities that are accessible in GNS<br />

cell cultures [404].<br />

84


Chapter 4<br />

The Non-Coding RNA World<br />

Contents<br />

4.1 MicroRNA regulation . . . . . . . . . . . . . . . . . . . . . 86<br />

4.2 Target Prediction and Validation . . . . . . . . . . . . . . . 90<br />

Non-coding RNAs are a growing subset <strong>of</strong> RNAs whose sizes, regulatory func-<br />

tions, range <strong>of</strong> regulated pathways and differential conservation within species<br />

distinguish them from the remainder <strong>of</strong> the coding and non-coding transcripts<br />

(mRNAs, tRNAs, rRNAs) classified as the traditional repertoire <strong>of</strong> RNAs ex-<br />

pressed within a cell. This new set <strong>of</strong> regulatory molecules in the RNA world<br />

is in continual expansion, as new types - unique in length, tissue specificity<br />

and regulatory mechanisms - are discovered and classified in different species.<br />

A broad distinction within this class <strong>of</strong> regulatory molecules is made in rela-<br />

tion to their length, distinguishing them between long and short non-coding<br />

RNAs (Fig 4.1). As a transcriptional class, long non-coding (nc)RNAs were<br />

first described during the large-scale sequencing <strong>of</strong> full-length cDNA libraries<br />

in the mouse [368], and are today arbitrarily considered longer than 200 nu-<br />

cleotides due to a practical cut-<strong>of</strong>f in RNA purification protocols that excludes<br />

small RNAs. Their specific roles are still in the process <strong>of</strong> being unveiled,<br />

although they already have been implicated in high-order chromosomal dy-<br />

namics, telomere biology and sub-cellular structural organization [331]. While<br />

this class <strong>of</strong> molecules has been most extensively studied in animal species,<br />

studies <strong>of</strong> long non-coding RNAs in plants begin to emerge showing some con-<br />

servation <strong>of</strong> mechanisms [31].<br />

On the other hand, the first small non-coding RNAs discovered were endoge-<br />

nous small-interfering RNAs (siRNAs) in plants [355,505], followed by mi-<br />

croRNAs (microRNAs) in C. elegans [264], repeat-associated small interfering<br />

85


4.1 MicroRNA regulation Introduction<br />

Figure 4.1: Classes <strong>of</strong> non-coding RNAs (ncRNAs) discovered to date: siRNA,<br />

small interfering RNA; microRNAs, microRNAs; rasiRNAs, repeat-associated small<br />

interfering RNAs; piRNAs, piwi-associated RNAs; endosiRNAs, endogenous small<br />

interfering RNAs; scnRNAs, scanRNAs; tasiRNAs, trans-acting RNAs.<br />

RNAs (rasiRNAs) in D. melanogaster [24] and, only recently, piwi-associated<br />

RNAs (piRNAs) [23,164,523] and endogenous small interfering RNAs (en-<br />

dosiRNAs) in mouse [485,524]. Other classes are found in ciliates, such as<br />

scanRNAs (scnRNAs) that ensure the fidelity <strong>of</strong> genome inheritance to the<br />

next generation [336], and plants, such as trans-acting RNAs (tasiRNAs) [509].<br />

Today siRNAs are known to act as gene silencers if exogenously introduced in<br />

mammals and microRNAs are well-established gene regulators in plants and<br />

animals, including the unicellular ciliate C. reinhardtii. Also, piRNAs are un-<br />

derstood to play a role in zebrafish - widening the regulatory horizons <strong>of</strong> such<br />

molecules - and rasiRNAs, now considered a subset <strong>of</strong> the piRNA class, have<br />

also been found in zebrafish [199]. In this thesis the focus will be maintained<br />

on the role <strong>of</strong> microRNA regulation in mammals.<br />

4.1 MicroRNA regulation<br />

The role <strong>of</strong> microRNAs has been studied in many different tissues and path-<br />

ways, as well as through various time points in the development <strong>of</strong> organisms.<br />

The first microRNA, lin-4, was identified in 1993 in a genetic screen for mutants<br />

that disrupted the timing <strong>of</strong> post-embryonic development in C. elegans [129],<br />

and today several hundred microRNAs are known to exist in the mammalian<br />

86


4.1 MicroRNA regulation Introduction<br />

genome regulating up to two thirds <strong>of</strong> protein coding genes [150,256]. The<br />

way that microRNAs regulate gene expression is through an enzymatic pro-<br />

cess that starts in the nucleus and is driven by Drosha. The biogenesis <strong>of</strong><br />

microRNAs in animal models starts when they are transcribed by RNA poly-<br />

merase II as primary transcripts, termed "pri-microRNAs". The initiation<br />

step <strong>of</strong> "cropping" is mediated by the Drosha-DGCR8 complex, also known as<br />

the Microprocessor complex. Drosha and DGCR8 are both located mainly in<br />

the nucleus. The product <strong>of</strong> this nuclear processing step is an approximate 70<br />

nucleotide pre-microRNA, which possesses a short stem plus an approximate<br />

two nucleotide 3' overhang. This structure might serve as a signature mo-<br />

tif that is recognised by the nuclear export factor exportin-5. Pre-microRNA<br />

constitutes a transport complex together with exportin-5 and the GTP-bound<br />

form <strong>of</strong> c<strong>of</strong>actor Ran. Following export outside <strong>of</strong> the nucleus, the cytoplasmic<br />

RNase III Dicer participates in the second processing step termed "dicing" to<br />

produce microRNA duplexes. The duplex is separated and usually one strand<br />

is selected as the mature microRNA, whereas the other strand is degraded.<br />

When the other strand is not degraded it is indicated with the "*" mark in<br />

microRNA target prediction algorithms [232].<br />

<strong>Transcriptional</strong> and Post-transcriptional Regulation<br />

MicroRNA genes are encoded either in independent transcription units, in<br />

polycistronic clusters or within introns <strong>of</strong> protein coding genes. Indepen-<br />

dently <strong>of</strong> Drosha, pre-microRNA hairpins can also be generated from introns<br />

through the combined action <strong>of</strong> the spliceosome and the lariat-debranching en-<br />

zyme [239]. Animal microRNAs have been shown to function differently from<br />

plant microRNAs, with the former class binding to their target 3'UTRs by<br />

imperfect matching. In fact, the RNA-Induced Silencing Complex (RISC) me-<br />

diates target mRNA cleavage by loading siRNAs and mature microRNAs with<br />

perfect sequence complimentarily to their target mRNA. The extent <strong>of</strong> the<br />

mismatch region varies amongst different microRNA-mRNA duplexes in ani-<br />

mals, but it is rarely the case that sequence complementarity spans the entire<br />

microRNA sequence, meaning that transcript cleavage would seem to be just<br />

as rare a mechanism <strong>of</strong> repression. Because the extent <strong>of</strong> this complementarity<br />

is low in animals, there is a region spanning two to eight nucleotides from the<br />

5' end <strong>of</strong> the microRNA that proves to be essential for the correct recognition<br />

<strong>of</strong> the message target. The 5' most nucleotide (t1) <strong>of</strong> the microRNA sequence<br />

does not have a significant role in this recognition process, as even when base<br />

87


4.1 MicroRNA regulation Introduction<br />

pairing occurs between t1, usually an adenine, and m1 on the messenger, usu-<br />

ally a uridine, this feature alone does not contribute to a repressive effect on<br />

the target [364].<br />

Interestingly, structural studies <strong>of</strong> the Argonaute 40 (AGO)-siRNA complex,<br />

demonstrated that the 5' most nucleotide <strong>of</strong> the guide RNA (equivalent to the<br />

m1 base <strong>of</strong> microRNAs) is not base-paired but instead bound by AGO [291].<br />

Due to its biological role, the region two to eight nucleotides from the 5' end<br />

<strong>of</strong> the microRNA sequence is termed "seed" [273], which later proved an excel-<br />

lent name choice in view <strong>of</strong> the conservation found for it across most metazoan<br />

microRNAs [282]. A number <strong>of</strong> single nucleotide mutation studies monitoring<br />

the effect <strong>of</strong> repression <strong>of</strong> microRNAs on their known targets also confirmed<br />

the biological importance <strong>of</strong> the seed region [237,469]. The number and variety<br />

<strong>of</strong> complementary seed sequences on the 3'UTRs <strong>of</strong> target genes determines<br />

the potential for modulating their expression by different types and numbers<br />

<strong>of</strong> microRNAs at various stages <strong>of</strong> the life <strong>of</strong> a cell [239].<br />

Three types <strong>of</strong> seeds have been observed to date displaying a range <strong>of</strong> target-<br />

ing efficiencies: "7mer-A1", lowest in the hierarchy, "7mer-m8", outperforming<br />

it, and "8mer" sites with the strongest repressive effects. The 7mer-A1 type<br />

matches over the entire seed sequence (seven positions) and carries an adenine<br />

at position t1. The 7mer-m8 type matches over the entire seed sequence as<br />

well but has an extra match at position 8 <strong>of</strong> the microRNA sequence, which<br />

accounts for its outperformance <strong>of</strong> the 7mer-A1 type. Finally, the 8mer site has<br />

a complete match over the entire seed sequence with an extra one at position<br />

eight, but outperforms the 7mer-m8 site by carrying an adenine at position<br />

t1 [359,446]. This relatively simplistic model, in which only the seed sequence<br />

and its closest nucleotides play a role in determining the efficacy <strong>of</strong> repression,<br />

has recently been refined to account for other features that seem to determine<br />

the identity <strong>of</strong> a microRNA target. Examples <strong>of</strong> these are the presence <strong>of</strong> spe-<br />

cific secondary structures, specific 3'UTR sequences surrounding the target site<br />

and the extent <strong>of</strong> complementarity between the 3'UTR and the 3' end <strong>of</strong> the<br />

microRNA sequence in order to compensate for the mismatching regions [239].<br />

Another structural feature to account for are the G:U matches or "bulges"<br />

that may form along the recognition sites <strong>of</strong> some microRNAs [70]. In vivo<br />

experiments, in fact, seem to suggest that more than one G:U base pair within<br />

40 Family <strong>of</strong> proteins that act as the catalytic component <strong>of</strong> the RNA-induced silencing<br />

complex by using small RNA guides (microRNAs, siRNAs and piRNAs) to bind and cleave<br />

target mRNAs.<br />

88


4.1 MicroRNA regulation Introduction<br />

the seed region could compromise the efficiency <strong>of</strong> target repression, a finding<br />

recently confirmed by large-scale whole proteome microRNA impact analy-<br />

ses [446,469]. However, extensive complementarity with the 3' end <strong>of</strong> the<br />

microRNA sequence [469,538] and/or additional target pairing to microRNA<br />

nucleotides 13-16 [43,180] seem to counteract the compromising effect <strong>of</strong> G:U<br />

bulges on 7mer seed matches although these 3' compensatory and 13-16nt sup-<br />

plementary sites, unlike the seed sequence, do not appear to be under selection<br />

pressure [71,272]<br />

Of equal importance to the quality <strong>of</strong> the microRNA-target sequence match, is<br />

the overall accessibility <strong>of</strong> the target site [224], which may easily be obstructed<br />

by RNA-binding proteins [222] or the possible un-pairing <strong>of</strong> the seed flank-<br />

ing sequences rendered necessary by the secondary structures took on by the<br />

3'UTR [224].<br />

Various mechanisms have been documented to account for microRNA repres-<br />

sion in animals: translational inhibition during the initiation or elongation <strong>of</strong><br />

protein synthesis, degradation <strong>of</strong> the nascent peptide chain, mRNA seques-<br />

tration to P-bodies 41 and mRNA degradation. Even though understanding<br />

the choice <strong>of</strong> which mechanism is used for different microRNA-mRNA du-<br />

plexes is still a matter <strong>of</strong> controversy, it is a generally accepted concept that<br />

features <strong>of</strong> the microRNA sequences and proteins bound to them play an im-<br />

portant role in determining the method <strong>of</strong> suppression. The data available<br />

today is indicative <strong>of</strong> two main types <strong>of</strong> regulation: indirect or with direct<br />

effects on translation [359]. Amongst the transcriptionally indirect mecha-<br />

nisms are mRNA degradation through deadenylation and decappying. The<br />

direct effects on translation, instead, are divided into inhibition at the level <strong>of</strong><br />

translation initiation, in which few or no ribosomes occupy the mRNA that is<br />

thought to be sequestered/stored in P-bodies, and inhibition post-initiation,<br />

in which the silenced mRNAs sediment in the polyribosome fractions in a su-<br />

crose gradient [239]. This latter type <strong>of</strong> inhibition can be further divided into<br />

three scenarios: premature ribosome drop-<strong>of</strong>f, stalled/slowed elongation and<br />

co-translational protein degradation [359]. Data from many separate studies<br />

support all the listed mechanisms, indicating that the inhibition scenario is a<br />

very complex one. AGO2, for example, has been shown to bind to the m7G<br />

cap <strong>of</strong> mRNAs through a domain similar in structure to the one on translation<br />

initiation factor eIF4E. This observation suggests that the recruitment on the<br />

41 Distinct foci within the cytoplasm <strong>of</strong> the eukaryotic cell consisting <strong>of</strong> many enzymes<br />

involved in mRNA turnover.<br />

89


4.2 Target Prediction and Validation Introduction<br />

mRNA’s 3'UTR <strong>of</strong> AGO2 via microRNA pairing blocks m7G cap recognition<br />

from the translation initiation factor, which is needed to start translation, thus<br />

supporting the inhibition <strong>of</strong> initiation view <strong>of</strong> miR-mediated repression [239].<br />

Even so, mRNAs inhibited by the action <strong>of</strong> miRs have been found to be associ-<br />

ated with actively translating polysomes, reinforcing the idea that, depending<br />

on the mRNA-miR duplex, a different subset <strong>of</strong> cases is selected for miR-<br />

mediated repression [239].<br />

4.2 Target Prediction and Validation<br />

Initial efforts to characterise the properties <strong>of</strong> microRNAs focused on their<br />

comprehensive identification in sequenced eukaryotic genomes, the numbers<br />

<strong>of</strong> mRNAs targeted, and the degree <strong>of</strong> evolutionary conservation among dif-<br />

ferent microRNA species. Whole-transcriptome and proteome analyses have<br />

reinforced the importance <strong>of</strong> target site evolutionary conservation by demon-<br />

strating a stronger down-regulation <strong>of</strong> the mRNAs having conserved sites<br />

[37,139,359,446]. Although the surprising high 3'UTR homology shared among<br />

vertebrates, very few target sites are conserved among the drosophilid and ne-<br />

matode lineages [91] and only nine orthologous targets <strong>of</strong> miR-7 were found<br />

to be shared between Drosophila and human (as an intersection from 97<br />

Drosophila and 581 human predicted targets) [266]. Following the cataloguing<br />

<strong>of</strong> microRNAs in a variety <strong>of</strong> organisms, subsequent work shifted towards the<br />

prediction and characterisation <strong>of</strong> their mRNA targets and the functional con-<br />

sequences <strong>of</strong> these regulatory interactions. In this context, a comprehensive<br />

list <strong>of</strong> empirical rules for microRNA target prediction should not overlook the<br />

importance <strong>of</strong> site accessibility.<br />

A fundamental challenge in the target prediction field has been to successfully<br />

predict the biologically functional targets while excluding false predictions [43].<br />

To date, a number <strong>of</strong> sophisticated algorithms have been developed to address<br />

these questions and at least seven <strong>of</strong> these tools are publicly available, each<br />

one based on distinct criteria with varying false-positive rates. These tools<br />

include: Targetscan [272,273], EMBL [470], PicTar [127,247], EIMMo [155],<br />

miRanda [134,213], miRBase [179], PITA [224] and DIANA-microT [237,316].<br />

Such methods differ considerably in their implementations <strong>of</strong> various predic-<br />

tion criteria; most search for complementary sequences between microRNAs<br />

and putative gene targets, while some consider physical and statistical hy-<br />

bridization properties, cross-conservation <strong>of</strong> regulatory RNAs between related<br />

90


4.2 Target Prediction and Validation Introduction<br />

species, etc. As a result, prediction methods differ widely in their relative de-<br />

grees <strong>of</strong> accuracy and coverage and the results produced <strong>of</strong>ten disagree.<br />

With predictions in the range <strong>of</strong> 300 evolutionarily conserved targets per<br />

mammalian microRNA family, it appears that microRNAs have the poten-<br />

tial to modulate the expression <strong>of</strong> nearly all the mammalian mRNAs [43]. The<br />

strong evolutionary pressure enacting the maintenance <strong>of</strong> the conserved tar-<br />

get sites within most <strong>of</strong> the 3'UTRs [150] is avoided by some housekeeping<br />

genes through the acquisition <strong>of</strong> exceptionally short 3'UTRs that are depleted<br />

<strong>of</strong> target sites [71]. A fundamental step in the microRNA target prediction<br />

pipeline is the experimental target validation <strong>of</strong> the predicted targets in order<br />

to confirm the validity <strong>of</strong> an approach over another. Several validation meth-<br />

ods have been employed, ranging from traditional genetic studies, rescue as-<br />

says [70], reporter-gene constructs [237,273] and mutation studies [71,124,237].<br />

In addition to confirming or discharging hypotheses built on networks <strong>of</strong> pre-<br />

dicted targets, these validation approaches represent the real bottleneck <strong>of</strong><br />

the whole process since they are most time-consuming and expensive. High-<br />

throughput approaches have also been developed, involving over-expression <strong>of</strong><br />

microRNAs in cell lines followed by microarray pr<strong>of</strong>iling to detect downregu-<br />

lated targets [283] and the reverse approach <strong>of</strong> depleting microRNAs to identify<br />

up-regulated targets [425].<br />

Assaying only relative changes in target mRNA levels without measuring the<br />

corresponding protein abundance is not sufficient to characterise all functional<br />

targets [19]. Thus, it is necessary to demonstrate that in addition to medi-<br />

ating the repression <strong>of</strong> gene expression through transcriptional degradation,<br />

microRNAs also directly repress translation [446]. Since proteins have dif-<br />

ferent turnover rates, a microRNA may require more or less time to change<br />

their steady-state levels. With the new version <strong>of</strong> the Stable Isotope Label-<br />

ing by Amino acids in Cell culture (SILAC) protocol developed by Rajwesky<br />

et al [446], called pulse-SILAC (pSILAC), only differences in newly synthe-<br />

sized proteins are detected by assaying the changes in their steady-state levels.<br />

SILAC is a technique based on mass spectrometry that uses isotopic rather<br />

than radioactive labelling <strong>of</strong> amino acids, to assay protein abundance in a cell.<br />

In standard and pSILAC, two cell populations are grown in identical culture<br />

media except for the presence, in one <strong>of</strong> the two cultures, <strong>of</strong> an isotope-labeled<br />

amino acid. In standard SILAC the labeled amino acid is fed to the cell cul-<br />

ture with the growth medium, so that it can be slowly incorporated into all<br />

91


4.2 Target Prediction and Validation Introduction<br />

newly synthesized proteins and the raw protein concentration is monitored.<br />

In pSILAC, instead, the labeled amino acid is fed to the cell culture for a<br />

short period <strong>of</strong> time (a pulse), so that only de novo protein production is<br />

monitored [446]. Specifically, the pSILAC protocol developed by Rajwesky et<br />

al [446] involves labelling two cell cultures (transfected and control) with a<br />

heavy and a medium-heavy version <strong>of</strong> the same amino acid, respectively. Af-<br />

ter 8h from transfection the cultures are pulse-labeled and the samples, united<br />

after 32h post-transfection, are analysed by mass spectrometry. RNA from<br />

the same samples (8h and 32h) was analyzed by Affymetrix arrays. In this<br />

fashion, approximately 5,000 proteins were identified in HeLa cells with high<br />

confidence. The search for six nucleotide motifs within the 3'UTRs <strong>of</strong> those<br />

mRNAs whose protein levels decreased the most (never exceeding 4-fold) re-<br />

vealed that the most significant motifs were exactly the seed sequences <strong>of</strong><br />

each respective microRNA. This showed that out <strong>of</strong> all proteins demonstrat-<br />

ing reduced synthesis, the levels <strong>of</strong> the direct targets <strong>of</strong> microRNAs decreased<br />

the most, and that this reduction is directly linked to the presence <strong>of</strong> the<br />

3'UTR target site. Nevertheless, a number <strong>of</strong> repressed proteins without seed<br />

sequences are still targeted, since their level is decreased, but prediction al-<br />

gorithms cannot identify them because they are non-canonical and seedless.<br />

Amongst the strongly repressed targets, a Gene Ontology search revealed that<br />

these are mainly proteins synthesized on the Endoplasmic Reticulum. A po-<br />

tential reason is that only mRNAs from cytosolic free ribosomes are targeted<br />

to P-bodies for degradation [446]. Other observations were that a nine to 11<br />

nucleotide mismatch along the mRNA-microRNA duplex was necessary for the<br />

protein production to be repressed, being otherwise indistinguishable to that<br />

<strong>of</strong> mRNAs lacking the seed. Although mismatches are deleterious to siRNA-<br />

mediated mRNA cleavage, they seem to correlate with increased repression<br />

<strong>of</strong> protein production by microRNAs. Also, the presence <strong>of</strong> multiple seeds<br />

seemed to have multiplicative repressive effects with a higher impact when<br />

the seeds were proximal rather than distant. On average, the repression was<br />

found more pronounced for conserved rather than non-conserved seed sites,<br />

indicating that there are more determinants other than the seed that mediate<br />

efficient down-regulation <strong>of</strong> protein synthesis. Interestingly, as opposed to the<br />

seed enrichment observed in the down-regulated genes, no seed enrichment was<br />

observed in the up-regulated ones, strongly speaking against the microRNA-<br />

mediated activation observed in other works [446].<br />

92


Methods<br />

93


Chapter 5<br />

Methods<br />

Contents<br />

5.1 Tag-sequencing Data Processing . . . . . . . . . . . . . . . 94<br />

5.2 Array Comparative Genomic Hybridization . . . . . . . . . 98<br />

5.3 Differential Gene Expression . . . . . . . . . . . . . . . . . 100<br />

5.4 Quantitative Real Time-PCR Validation . . . . . . . . . . . 101<br />

5.5 Literature Mining . . . . . . . . . . . . . . . . . . . . . . . 107<br />

5.6 Differential Is<strong>of</strong>orm Expression . . . . . . . . . . . . . . . . 110<br />

5.7 Differential Long ncRNA Expression . . . . . . . . . . . . . 112<br />

5.8 <strong>Glioma</strong> Expression Signatures . . . . . . . . . . . . . . . . 112<br />

5.9 External Dataset Expression Correlation . . . . . . . . . . 112<br />

5.10 Glioblastoma Pathway Construction . . . . . . . . . . . . . 114<br />

5.11 MicroRNA Target Prediction Analysis . . . . . . . . . . . . 117<br />

5.1 Tag-sequencing Data Processing<br />

Instead <strong>of</strong> the traditional microarray expression pr<strong>of</strong>iling, Solexa sequencing<br />

was performed on total RNA from four GNS cell lines: G144ED, G144, G179<br />

and G166, and two NS cell lines from fetal brain: CB541 and CB660 (Table<br />

5.1). Tag-sequencing (Tag-seq) library preparation and sequencing was carried<br />

out using the Illumina DGE Tag Pr<strong>of</strong>iling kit according to the manufacturer’s<br />

instructions. We have submitted the data to ArrayExpress. The libraries<br />

were specifically constructed with the longSAGE protocol, in which first and<br />

second strand cDNA is synthesized with a biotinylated oligo deoxy-thymine<br />

(oligo-dT) onto streptavidin beads. The bound cDNA is cleaved at the 5’ end<br />

by an anchoring enzyme, such as NlaIII, that adds GTAC, the recognition<br />

site for MmeI, to the poly-thymine (poly-T) strand. Sequencing adapters with<br />

94


5.1 Tag-sequencing Data Processing Methods<br />

Table 5.1: Summary <strong>of</strong> cell lines investigated with the Tag-seq platform. (M=Male,<br />

F=Female)<br />

Type <strong>of</strong> cell line Name <strong>of</strong> cell line Tissue Sex<br />

GNS G144 GBM M<br />

GNS G144ED GBM M<br />

GNS G166 GBM F<br />

GNS G179 Giant cell glioblastoma M<br />

NS CB541 Fetal forebrain -<br />

NS CB660 Fetal forebrain -<br />

an MmeI recognition site are then linked to the cDNA and MmeI digestion<br />

removes the 3’ portion <strong>of</strong> the cDNA from the beads, generating tags <strong>of</strong> con-<br />

stant length <strong>of</strong> 17nt that are then ligated to a 3’ adaptor and sequenced, as<br />

shown in figure 5.1 [490]. In the case <strong>of</strong> an Illumina sequencing platform, these<br />

Figure 5.1: Step by step diagram <strong>of</strong> the longSAGE protocol used to generate reads<br />

that contain tag sequences derived from the mRNA pool.<br />

95


5.1 Tag-sequencing Data Processing Methods<br />

constructs are hybridised onto the glass surface <strong>of</strong> a slide coated with comple-<br />

mentary oligonucleotides and "bridge" amplified to then have them sequenced<br />

with fluorescence-labeled nucleotide analogs [47]. Because each tag measures<br />

expression levels that are not based on transcript length, a digit or "count" is<br />

associated to it and the technology is <strong>of</strong>ten referred to as "digital gene expres-<br />

sion" tag pr<strong>of</strong>iling [490].<br />

The Tag-seq technology is based on the principles <strong>of</strong> solid-phase sequencing and<br />

Serial Analysis <strong>of</strong> Gene Expression (SAGE). In this way, Tag-seq imports the<br />

quantitative and unbiased transcriptome pr<strong>of</strong>iling proper <strong>of</strong> SAGE library con-<br />

struction as well as the sequencing depth, dynamic range and cost-effectiveness<br />

<strong>of</strong> the latest high-throughput sequencing platforms. Furthermore, Tag-seq is<br />

not affected by the cross-hybridization and dynamic range limitations typical<br />

<strong>of</strong> microarray technology, since no sequence-specific hybridization is required<br />

for expression detection and the dynamic range is limited in principle only by<br />

the sequencing depth <strong>of</strong> the platform [346,490]. Tag-seq is similar to RNA-seq<br />

in that both technologies don’t require pre-existing knowledge <strong>of</strong> the genome<br />

analysed and Tag-seq has proven to perform comparably in terms <strong>of</strong> gene<br />

discovery and dynamic range [346,507]. Unlike RNA-seq, however, Tag-seq<br />

strictly monitors the 3’ end usage <strong>of</strong> transcripts and carries no information on<br />

their internal structure [194]. Finally, Tag-seq can be used as a strand-specific<br />

gene expression platform, which is not always true <strong>of</strong> RNA-seq, depending on<br />

the type <strong>of</strong> adaptor ligation performed [346].<br />

We extracted tag sequences as the first 17nt <strong>of</strong> each read and counted the<br />

number <strong>of</strong> occurrences <strong>of</strong> each tag in each sample. Due to sequence errors<br />

introduced during library preparation and sequencing, highly expressed tran-<br />

scripts might have caused a significant number <strong>of</strong> tags to differ by a single base<br />

from the expected tag. To compensate for such errors, we used the Recount<br />

program [528], setting the hamming distance parameter to 1. Recount uses an<br />

expectation maximization algorithm to estimate "true" tag counts, i.e. counts<br />

in the absence <strong>of</strong> error, based on observed tag counts and base call quality<br />

scores.<br />

We excluded tags matching adapters or primers used in library construction<br />

and sequencing, as well as tags matching ribosomal RNA (rRNA) or mitochon-<br />

drial sequence. Adapter and primer contamination was identified by running<br />

96


5.1 Tag-sequencing Data Processing Methods<br />

the TagDust program [258] with a target false discovery rate (FDR) <strong>of</strong> 1%.<br />

Tags matching ribosomal sequence were identified by using the Bowtie pro-<br />

gram [257] to align against a database consisting <strong>of</strong> all rRNA genes, including<br />

pseudogenes, from Ensembl 56 [147] and all ribosomal repeats in the UCSC<br />

Genome Browser RepeatMasker track for genome assembly GRCh37 [152];<br />

only perfect matches to the extended 21nt tag sequence, consisting <strong>of</strong> the<br />

NlaIII site CATG followed by the observed 17nt tag, were accepted. Mito-<br />

chondrial tags were similarly excluded by searching for perfect matches to the<br />

mitochondrial chromosome sequence. The parameters used to run the Bowtie<br />

program were the following:<br />

-f parameter indicates that the query input files are FASTA files;<br />

-n parameter set to 0 means that the alignments may have no more than n<br />

mismatches. Since we are only looking for perfect matches, n=0.<br />

-y parameter specifies to the program to try as hard as possible to find valid<br />

alignments when they exist, which makes running this mode much slower;<br />

-k 2 instructs the program to report up to two valid alignments;<br />

-m 1 instructs the program to refrain from reporting any alignments for reads<br />

having more than one reportable alignment. This option is useful when<br />

the user wants to guarantee that reported alignments are unique, which<br />

is our case.<br />

To assign tags to genes, we employed a hierarchical strategy based on the ex-<br />

pectation that tags are most likely to originate from the 3’-most NlaIII site in<br />

known transcripts. Tags were assigned to transcripts using virtual tag data<br />

from the SAGE Genie database [65] and virtual tags extracted from Ensembl<br />

transcript models. The SAGE Genie annotation consisted <strong>of</strong> 105 sets <strong>of</strong> virtual<br />

tags obtained by scanning for NlaIII sites in cDNAs from RefSeq, Mammalian<br />

Gene Collection (MGC) and Genbank, then Expressed Sequence Tags (ESTs),<br />

UniGene consensus sequences and transfrags. The virtual tag sets are further<br />

classified based on the position <strong>of</strong> the tag relative to the 3’ end <strong>of</strong> the transcript<br />

and indicators <strong>of</strong> 3’ end reliability such as polyadenylation signal and poly-A<br />

tail. Since SAGE Genie does not cover Ensembl transcripts, we also extracted<br />

virtual tags from the 3’-most cut site in each Ensembl transcript. We used the<br />

CATG recognition sequence as a "signal" that indicated where to start count-<br />

ing 17nt ahead. This allowed us to extract that 17nt tag and know exactly from<br />

97


5.2 Array Comparative Genomic Hybridization Methods<br />

which transcript it was extracted. If the cut site was less than 17nt from the<br />

end <strong>of</strong> the transcript, we extended the sequence with adenine stretches, rep-<br />

resenting the poly-adenine (poly-A) tail. In these cases, additional upstream<br />

tags were also extracted until one tag fully contained in the Ensembl cDNA<br />

sequence was obtained. We disregarded Ensembl transcripts not belonging<br />

to a gene <strong>of</strong> biotype "protein_coding" or "processed_transcript". Ensembl<br />

annotates genes <strong>of</strong> several other biotypes, e.g. pseudogenes and small RNAs,<br />

but those annotations are not based on full-length transcript sequences, so<br />

we would not expect to find valid virtual tags in those transcripts. For the<br />

majority <strong>of</strong> this study, we used a conservative subset <strong>of</strong> the virtual tags from<br />

SAGE Genie and Ensembl comprising 25,593 unique tags assigned to 15,103<br />

genes (Table 5.2). Specifically, we used SAGE Genie tags extracted from to the<br />

3’-most cut site in RefSeq or MGC cDNAs having a poly-A tail or a polyadeny-<br />

lation signal, and Ensembl tags from transcripts <strong>of</strong> type "protein_coding" or<br />

"non_coding". Any virtual tags that mapped to multiple loci by these criteria<br />

were excluded. For certain analyses, we made use <strong>of</strong> more comprehensive vir-<br />

tual tag sets. In addition, we determined unique, perfect matches for tags to<br />

the genome using Bowtie as described above. We calculated a single expression<br />

value for each gene in each cell line by summing the counts <strong>of</strong> tags assigned to<br />

the gene.<br />

5.2 Array Comparative Genomic Hybridization<br />

We re-analysed the array comparative genomic hybridization (CGH) data de-<br />

scribed by Pollard et al [404] CGH was performed with Human Genome CGH<br />

Microarray 4x44K arrays (Agilent), using genomic DNA from each cell line<br />

hybridised in duplicate (dye swap) and normal human female DNA as ref-<br />

erence (Promega). Log2 ratios were computed from processed Cy3 and Cy5<br />

intensities reported by the s<strong>of</strong>tware CGH Analytics (Agilent). We corrected<br />

for effects related to GC content and restriction fragment size using a modi-<br />

fied version <strong>of</strong> the waves array CGH correction algorithm [271]. Log2 ratios<br />

were adjusted by sequential loess normalization on three factors: fragment GC<br />

content, fragment size, and probe GC content. These were selected after in-<br />

vestigating dependence <strong>of</strong> log ratio on multiple factors, including GC content<br />

in windows <strong>of</strong> up to 500 kilobases centred around each probe. The Biocon-<br />

ductor package CGHnormaliter [506] was then used to correct for intensity<br />

dependence and log2 ratios scaled to be comparable between arrays using the<br />

98


5.2 Array Comparative Genomic Hybridization Methods<br />

Table 5.2: Classification <strong>of</strong> sequenced tags in each cell line.<br />

G144ED G144 G166 G179 CB541 CB660<br />

Sequenced tags 6,383,175 7,133,520 13,415,402 11,610,415 12,103,066 10,043,561<br />

Filtered tags 751,698 11.78% 912,971 12.80% 378,009 2.82% 347,537 2.99% 327,597 2.71% 382,819 3.81%<br />

Adapter 594,513 9.31% 765,484 10.73% 90,407 0.67% 45,318 0.39% 39,992 0.33% 248,799 2.48%<br />

Mitochondrial 156,534 2.45% 147,245 2.06% 285,881 2.13% 302,148 2.60% 287,493 2.38% 133,935 1.33%<br />

Ribosomal RNA 651 0.01% 242 0.00% 1,721 0.01% 71 0.00% 112 0.00% 85 0.00%<br />

Tags assigned to a single locus 2,812,750 44.07% 2,640,558 37.02% 6,271,471 46.75% 5,603,364 48.26% 5,984,114 49.44% 3,708,853 36.93%<br />

Reference tags, unique 1,420,009 22.25% 1,344,879 18.85% 2,970,554 22.14% 2,805,423 24.16% 2,894,539 23.92% 1,712,143 17.05%<br />

Reference tags, best 628,707 9.85% 577,418 8.09% 1,783,685 13.30% 1,267,594 10.92% 1,485,211 12.27% 982,190 9.78%<br />

cDNA tags, unique 146,442 2.29% 142,369 2.00% 295,764 2.20% 284,990 2.45% 261,411 2.16% 171,970 1.71%<br />

99<br />

cDNA tags, best 34,999 0.55% 34,472 0.48% 103,632 0.77% 105,787 0.91% 81,547 0.67% 49,374 0.49%<br />

Other SAGE Genie tags, unique 345,759 5.42% 322,034 4.51% 652,449 4.86% 684,718 5.90% 749,653 6.19% 455,045 4.53%<br />

Other SAGE Genie tags, best 72,658 1.14% 60,725 0.85% 158,601 1.18% 107,955 0.93% 148,854 1.23% 84,542 0.84%<br />

Tags not mapping to known transcrip-<br />

164,176 2.57% 158,661 2.22% 306,786 2.29% 346,897 2.99% 362,899 3.00% 253,589 2.52%<br />

tome but uniquely to genome<br />

Ambiguously mapping tags 205,626 3.22% 185,115 2.60% 608,978 4.54% 418,267 3.60% 437,887 3.62% 388,085 3.86%<br />

Reference tags 165,895 2.60% 148,512 2.08% 516,134 3.85% 337,710 2.91% 366,115 3.02% 288,253 2.87%<br />

cDNA tags 5,996 0.09% 4,464 0.06% 10,566 0.08% 6,455 0.06% 6,708 0.06% 40,908 0.41%<br />

Other SAGE Genie tags 8,581 0.13% 6,717 0.09% 25,914 0.19% 14,668 0.13% 13,553 0.11% 12,508 0.12%<br />

Tags not mapping to known transcrip-<br />

25,154 0.39% 25,422 0.36% 56,364 0.42% 59,434 0.51% 51,511 0.43% 46,416 0.46%<br />

tome but to multiple genomic locations<br />

Unclassified tags 2,613,101 40.94% 3,394,874 47.59% 6,156,944 45.89% 5,241,247 45.14% 5,353,468 44.23% 5,563,803 55.40%


5.3 Differential Gene Expression Methods<br />

"scale" method in the package limma [466]. Replicate arrays were averaged<br />

and the genome (GRCh37) segmented into regions with different copy number<br />

using the circular binary segmentation algorithm in the Bioconductor package<br />

DNAcopy [510], with the option undo.SD set to 1. Aberrations were called<br />

using the package CGHcall [503] [37] with the option nclass set to 4. CGH<br />

data are available from ArrayExpress [28] under accession E-MTAB-972.<br />

5.3 Differential Gene Expression<br />

We called the differentially expressed genes with the Bioconductor package<br />

DESeq that uses pseudo reference-based normalization [21]. DESeq compares<br />

sequencing-derived expression pr<strong>of</strong>iles using a method that is able to account<br />

for large variation between biological replicates, as can be expected with can-<br />

cer samples. The R code that generated the differential expression calls is<br />

reported below, with ## signs indicating a comment:<br />

## Define auxiliary functions<br />

read.gns.counts


5.4 Quantitative Real Time-PCR Validation Methods<br />

resVarB=de$resVarB, counts)<br />

rownames(x)


5.4 Quantitative Real Time-PCR Validation Methods<br />

PCR. This gene set comprises 82 validation targets from Tag-seq analysis, eight<br />

glioma and developmental markers, and three endogenous control genes - 18S<br />

ribosomal RNA, TUBB and NDUFB10. The 18S gene was chosen because<br />

used by ABI as a manufacturing control, while TUBB and NDUFB10 were<br />

selected because they are present in our Tag-seq dataset and show low varia-<br />

tion <strong>of</strong> expression across GNS and NS cell lines in an independent microarray<br />

expression dataset [404]. The 93 genes were interrogated using 96 different<br />

TaqMan assays (three <strong>of</strong> the validation targets required two different primer<br />

and probe sets to cover all known transcript is<strong>of</strong>orms matching differentially<br />

expressed tags). cDNA was generated using SuperScript III (Invitrogen) and<br />

real-time PCR carried out using TaqMan fast universal PCR master mix. The<br />

absence <strong>of</strong> a no-template control (NTC) amplification (horizontal slope) en-<br />

sured that random contamination and reagent contamination were not affect-<br />

ing our samples. In complying with the minimum information for publication<br />

<strong>of</strong> quantitative real-time PCR experiments (MIQE) [78], a full assay list with<br />

raw and normalised threshold cycle (Ct) values is provided in Appendix A.3.<br />

The data analysis was performed using the R package HTqPCR [131], which<br />

handles high-throughput qPCR data with a focus on data from Taqman low<br />

density arrays. Ct values were normalised to the average <strong>of</strong> the three control<br />

genes and potential outliers identified and filtered out using the plotCtBoxes<br />

function (Fig 5.2). Figure 5.3 shows the effect <strong>of</strong> the normalisation method<br />

Figure 5.2: Boxplot <strong>of</strong> normalised Ct values identifies outliers (empty circles).<br />

102


5.4 Quantitative Real Time-PCR Validation Methods<br />

Figure 5.3: Correlation scatter plot showing the effect <strong>of</strong> the normalization method<br />

on the raw Ct values measured for the three endogenous controls.<br />

employed on the endogenous controls rearranging the Ct values to a smaller<br />

dynamic range. To capture biological variability within cell lines, we measured<br />

up to four independent RNA samples per line (indicated with the letters A,<br />

B, C, D in tables A.3 and A.4 in Appendix A.3). Figure 5.4 shows that the<br />

concordance between all our A, B biological replicates is very high and few<br />

outliers can be observed - identified with a gene name and highlighted in red<br />

in the figure. Differentially expressed genes were identified by the Wilcoxon<br />

rank sum test after averaging replicates.<br />

103


5.4 Quantitative Real Time-PCR Validation Methods<br />

104


5.4 Quantitative Real Time-PCR Validation Methods<br />

105


5.4 Quantitative Real Time-PCR Validation Methods<br />

Figure 5.4: Scatter plots <strong>of</strong> each <strong>of</strong> our A and B biological replicates shows a high<br />

degree <strong>of</strong> concordance between samples with very few outliers (highlighted in red).<br />

In order to assess the repeatability, or intra-assay variation, <strong>of</strong> the expression<br />

levels measured with qRT-PCR for each <strong>of</strong> the N=96 genes, standard devia-<br />

tions (σ) were computed for the differences in gene expression levels between<br />

two replicates (Fig 5.5). Standard deviations ranged between 3.5


5.5 Literature Mining Methods<br />

Figure 5.5: Dot plot <strong>of</strong> the standard deviations (blue) <strong>of</strong> the differences between<br />

expression levels in two replicates. Standard deviations were computed for each<br />

combination <strong>of</strong> replicate cell lines.<br />

5.5 Literature Mining<br />

A web script (provided in Appendix B) was implemented to automatically pro-<br />

cess the querying <strong>of</strong> the 748 differentially expressed genes (F DR < 10%) and<br />

therefore establish their role, if any so far had been assigned, in glioblastoma<br />

or other cancers.<br />

The script was modelled against a browser query frame type in the Visual<br />

Basic.NET programming language, which I deemed the best coding choice for<br />

this data-hefty application because <strong>of</strong> its "collection" objects, i.e. extremely<br />

powerful hash tables that make use <strong>of</strong> an ultra-fast key and vector content<br />

look-up. Moreover, the presence <strong>of</strong> the .NET Framework enriches this particu-<br />

lar edition <strong>of</strong> Visual Basic with extensive client-provider and internet browsing<br />

libraries that I expected to need for this particular application.<br />

The code I developed implements a process <strong>of</strong> hierarchical look-up, recapit-<br />

ulated in figure 5.6, that starts with the most glioblastoma-specific database<br />

amongst GBMbase [162], Google Scholar, Google Search, BioGraph [280], in-<br />

formation Hyperlinked Over Proteins (iHOP) [197] and PubMed [410], and<br />

loops all the way to the least-specific database. In this way, the probability <strong>of</strong><br />

finding an association between the queried gene and glioblastoma diminishes<br />

at every iteration and is unlikely to exist by the time the execution arrives<br />

107


5.5 Literature Mining Methods<br />

at the last and least-specific <strong>of</strong> the databases. This algorithm maximises the<br />

probability <strong>of</strong> finding a reported connection between any gene and any disease,<br />

prompted minor disease-specific details are changed such as the identity <strong>of</strong> the<br />

first database, which needs to be the most disease-specific <strong>of</strong> the six.<br />

If we call "case" the code that searches for the association between queried<br />

gene and glioblastoma in one database, and "iteration" the search loop com-<br />

pleted from first to last database, then during each case a weighted-index is<br />

accumulated to become, at the end <strong>of</strong> one iteration, a total weighted index, i.e.<br />

a score <strong>of</strong> how well that particular gene did in the search for its association to<br />

glioblastoma in the six databases. Therefore, the total weighted index repre-<br />

sents those parameters used in the look-ups that yielded the most favourable<br />

results, i.e. which database successfully found an association between the gene<br />

and glioblastoma, how many hits were found, and in which part <strong>of</strong> the paper<br />

the hits were found. In fact, I assigned a greater weight to the associations<br />

found in the title, with respect to those found in the abstract, with respect to<br />

those found in the contents <strong>of</strong> a paper. In designing the weight structure I also<br />

decided to favour the associations found in the same sentence to those found<br />

more than one sentence apart. The latter, in fact, barely carry any relevance<br />

in the value cumulation process <strong>of</strong> the total weighted index.<br />

In this particular application it was interesting to know whether the queried<br />

gene was implicated in any type <strong>of</strong> cancer, especially in case the literature<br />

did not report an association with glioblastoma. Therefore, at every iteration,<br />

the databases are also queried with parameters that identify the answer to<br />

the question "Is this gene implicated in any cancer?" and a separate index<br />

from the total weighted index is determined. To calculate this cancer index<br />

I searched for an association between the gene being queried and any <strong>of</strong> the<br />

cancers listed in a database that I compiled for this particular application. All<br />

the cancers listed in the National Cancer Institute A to Z List <strong>of</strong> Cancers [206]<br />

are present in the cancer database as a unique set <strong>of</strong> names. A search was per-<br />

formed in PubMed for every combination <strong>of</strong> queried gene symbol and cancer<br />

name from the compiled cancer database and, if the two terms appeared in the<br />

same sentence, this was considered an association and the cancer index for that<br />

combination incremented by one. At the end <strong>of</strong> the search, every gene-cancer<br />

combination possessed a cancer-index and the association was reported if the<br />

value <strong>of</strong> such index was above an arbitrarily chosen threshold value.<br />

108


5.5 Literature Mining Methods<br />

How many hits?<br />

I = 0<br />

Category 4<br />

I ≧ 1<br />

Category 3<br />

Which cancers?<br />

Yes<br />

Is this gene associated<br />

with cancers?<br />

No<br />

Is this gene<br />

associated with GBM<br />

in GBMbase?<br />

No<br />

Is this gene associated<br />

with GBM in iHOP?<br />

No<br />

Is this gene associated<br />

with GBM in<br />

PubMed?<br />

No<br />

Is this gene<br />

associated with GBM<br />

in BioGraph?<br />

No<br />

Is this gene<br />

associated with GBM<br />

in Google Scholar?<br />

No<br />

Is this gene<br />

associated with GBM<br />

in Google Search?<br />

Yes<br />

Yes<br />

Yes<br />

Yes<br />

Yes<br />

Yes<br />

How many hits?<br />

1 ≦ I < 10<br />

Category 2<br />

How many hits?<br />

1 ≦ I < 10<br />

Category 2<br />

How many hits?<br />

1 ≦ I < 10<br />

Category 2<br />

How many hits?<br />

1 ≦ I < 10<br />

Category 2<br />

How many hits?<br />

1 ≦ I < 10<br />

Category 2<br />

How many hits?<br />

1 ≦ I < 10<br />

Figure 5.6: In this diagram the "cases" (code that searches for the association<br />

between queried gene and glioblastoma) and "iterations" (search loop completed<br />

from first to last database) structures in the execution code are made obvious. At<br />

every question the answers identify the parameters that are used to assign the queried<br />

gene to one <strong>of</strong> the four categories or skip the assignment for that particular case and<br />

go through the rest <strong>of</strong> the iteration. I = number <strong>of</strong> associations found for the gene<br />

symbol <strong>of</strong> the queried gene and the word "glioblastoma" or "GBM".<br />

At the end <strong>of</strong> an iteration the total weighted index and the cancer index for<br />

every gene-cancer combination were available and, depending on their values,<br />

the queried gene was assigned to one <strong>of</strong> four categories, which answer one <strong>of</strong><br />

the following questions:<br />

Category 1. Does extensive literature implicate the gene in GBM?<br />

Category 2. Does a limited amount <strong>of</strong> literature implicate the gene in GBM?<br />

Category 3. Is there no literature implicating the gene in GBM, but does it<br />

appear to be implicated in other cancers?<br />

109<br />

I ≧ 10<br />

I ≧ 10<br />

I ≧ 10<br />

I ≧ 10<br />

I ≧ 10<br />

I ≧ 10<br />

Category 1<br />

Category 2<br />

Probability <strong>of</strong> finding an associa2on with GBM


5.6 Differential Is<strong>of</strong>orm Expression Methods<br />

Category 4. Is there no literature implicating the gene in any cancer?<br />

If W = total weighted index and C = cancer index, then if W > 10 (internal<br />

parameter, does not appear in fig 5.6), the gene is assigned to category one; if 1<br />

< W < 10, the gene is assigned to category two; if W = 0 and C > 10 (internal<br />

parameter, does not appear in fig 5.6) then the gene is assigned to category<br />

three; finally, if W = 0 and C = 0 then the gene is assigned to category four.<br />

5.6 Differential Is<strong>of</strong>orm Expression<br />

With the aim <strong>of</strong> establishing Tag-seq as a sensitive technique for the iden-<br />

tification <strong>of</strong> 3'UTR transcript is<strong>of</strong>orms that are differentially expressed be-<br />

tween GNS and NS cell lines, we evaluated the differential expression <strong>of</strong> 4,727<br />

genes with multiple expressed tags using two alternative methods: the non-<br />

parametric χ 2 test, and the logarithmic algorithm adapted from Morrissy et<br />

al [346]. Non-parametric methods are applied when the assumptions for para-<br />

metric methods about the underlying distribution <strong>of</strong> the data are not met,<br />

namely that the data is normally distributed or that sample size is large enough<br />

to support the assessment <strong>of</strong> the normality distribution. Whilst the normal-<br />

ity assumption is satisfied when tested against the 28,351 tags mapping over<br />

16,025 genes in the six libraries altogether, sample size drops to a range <strong>of</strong> two<br />

to 14 tags per gene when assessing differential expression over a single tran-<br />

script is<strong>of</strong>orm. With a sample size smaller than 30, the use <strong>of</strong> non-parametric<br />

methods is <strong>of</strong>ten advisable. Due to the nature <strong>of</strong> the SAGE protocol, tags map<br />

at the 3'UTR <strong>of</strong> a gene and identify potential splicing is<strong>of</strong>orms. We conducted<br />

a χ 2 test with the chisq.test function in R on all tags mapping on to the same<br />

gene <strong>of</strong> the 12,794 tags expressed at higher than 10 tpm in at least one GNS<br />

and one NS library.<br />

In the second approach, following the logarithmic ratio method adapted by<br />

Morrissy et al [346], the is<strong>of</strong>orm expression between the GNS and NS cell lines<br />

was evaluated by analyzing the expression <strong>of</strong> tags that mapped either twice on<br />

the same gene or, if more than twice, for all their pair combinations. Again,<br />

only tags with a minimum expression <strong>of</strong> 10 tpm in at least one GNS and one<br />

NS library were considered (Table 5.3). The ratio <strong>of</strong> normalised expression<br />

for each tag pair was calculated in each GNS and NS library, and multiplied<br />

by the natural logarithm <strong>of</strong> the difference between the expression <strong>of</strong> each tag<br />

in the pair, ensuring that the more highly expressed tag pairs would also be<br />

more highly ranked (equations 5.4 and 5.5). For pairs <strong>of</strong> genes with positive<br />

110


5.6 Differential Is<strong>of</strong>orm Expression Methods<br />

Table 5.3: Summary <strong>of</strong> statistics using the χ 2 and logarithmic tests.<br />

Non-parametric<br />

χ 2 test<br />

Logarithmic<br />

ratio<br />

Number <strong>of</strong> tags and tag-pairs mapped (2-14/gene) 12,792 13,599<br />

Number <strong>of</strong> genes identified by tags 4,727 4,541<br />

Number <strong>of</strong> multiple mapping tags at p< 0.01 8,169 3,651<br />

Number <strong>of</strong> genes differentially expressed between is<strong>of</strong>orms<br />

identified by multiple mapping tags at p< 0.01<br />

2,682 2,040<br />

ratio changes, the ratio was higher in GNS cell lines compared to NS cell lines,<br />

while pairs with negative values <strong>of</strong> ratio changes had a ratio that was higher in<br />

NS cell lines rather than GNS cell lines (equation 5.7). Gene pair ratio-change<br />

values ranged from -17.4 to 17.7 and no p-values were produced. The equations<br />

used are shown below, where S1 and S2 stand for any two is<strong>of</strong>orms identified<br />

by tag mapping:<br />

S1<br />

S2 = ln[ln(S1expression − S2expression) × ( S1expression<br />

)] (5.4)<br />

S2expression<br />

If the second is<strong>of</strong>orm is more highly expressed than the first, the ratio is cal-<br />

culated as:<br />

If<br />

S1<br />

S2 = −1 × ln[ln(S2expression − S1expression) × ( S2expression<br />

)] (5.5)<br />

S1expression<br />

0 < (S1expression − S2expression)|(S2expression − S1expression) < 1 (5.6)<br />

then the pair was omitted.<br />

For each tag pair, the average and standard deviation was computed for all<br />

modified means in GNS cell lines, and separately for all means in NS cell lines.<br />

An overall measure <strong>of</strong> the change in the ratio between GNS and NS cell lines<br />

was computed as shown below:<br />

δ GNS<br />

NS<br />

= ln(µGNS ) − ln(<br />

σGNS<br />

µNS<br />

) (5.7)<br />

σNS<br />

Dividing each mean by the standard deviation ensures that tag pairs with<br />

lower variance in their ratios are ranked higher than tag pairs with a higher<br />

variance [346]. The logarithmic algorithm uses the mean and standard devi-<br />

ation statistics to describe the ratios <strong>of</strong> expression change between the GNS<br />

and NS lines. Unlike the non-parametric method, this algorithm produces a<br />

ratio change for each pair <strong>of</strong> tags rather than for the sum over all tags and the<br />

111


5.7 Differential Long ncRNA Expression Methods<br />

absolute value tells the fold change whilst the sign the direction <strong>of</strong> the change.<br />

The 13,599 tag-pairs analysed pertained to a total <strong>of</strong> 4,541 genes, a number<br />

closely matching the 4,727 genes assessed with the non-parametric method.<br />

By ranking the tag-pairs according to magnitude <strong>of</strong> ratio change and setting a<br />

threshold at two-fold, the number <strong>of</strong> tag-pairs decreased to 3,651 but the num-<br />

ber <strong>of</strong> genes being represented was still substantially high at 2,040, a number<br />

comparable to the 2,682 genes identified in the non-parametric method.<br />

5.7 Differential Long ncRNA Expression<br />

We called differential expression with the Bioconductor package DESeq for all<br />

tags that mapped to a unique location in the reference human genome with the<br />

Bowtie program and filtered the results to exclude protein-coding transcripts.<br />

This analysis revealed 25 ncRNAs with strong evidence <strong>of</strong> differential expres-<br />

sion between GNS and NS lines. It is unlikely that these transcripts represent<br />

artifacts <strong>of</strong> the Tag-Seq procedure, because all coincide with published cDNA<br />

sequences or ESTs.<br />

5.8 <strong>Glioma</strong> Expression Signatures<br />

The Tag-seq data described in Parsons et al [383] was kindly provided by<br />

Dr. N. Papadopoulos. Following quality control, we excluded three out <strong>of</strong> 21<br />

sequencing lanes. To treat the expression data in a similar manner as Verhaak<br />

et al [511], Tag-seq expression values were transformed by computing log fold<br />

change relative to the mean expression across the glioma tumour and xenograft<br />

samples. Only those genes identified by Verhaak et al [511] as signature genes<br />

for a subtype were used in the correlation calculations for that subtype. To<br />

obtain robust fold change estimates, the gene sets were further limited to those<br />

genes having a normalized expression level above 25 tags per million in any<br />

sample. This reduced the original set <strong>of</strong> 210 signature genes per subtype to 158,<br />

167, 183 and 164 for "proneural", "neural", "classical" and "mesenchymal"<br />

subtypes, respectively. P-values were computed with the R function cor.test.<br />

5.9 External Dataset Expression Correlation<br />

We used Affymetrix Exon 1.0 ST microarray data from The Cancer Genome<br />

Atlas (TCGA; http://cancergenome.nih.gov; [326]) for 397 glioblastomas<br />

112


5.9 External Dataset Expression Correlation Methods<br />

and 10 non-neoplastic brain samples (dataset GBM). In addition, we used<br />

Affymetrix HG-U133A and HG-U133B data for 24 grade III and 50 grade IV<br />

gliomas from Freije et al [148] and 21 grade III and 55 grade IV gliomas from<br />

Phillips et al [390] (dataset HGG), For the survival analysis the dataset from<br />

Gravendeel et al [176] and that from Murat et al [353] were used (Table 5.4).<br />

All tumour microarray data were from primary glioma samples obtained at ini-<br />

Table 5.4: Public gene expression datasets used in trying to establish tumour<br />

expression correlations with the differentially expressed genes found through Tagseq<br />

and with the clinical data available from TCGA in the survival analysis.<br />

Citation Accession Microarray Number <strong>of</strong> cases<br />

platform GBM Grade Other Grade Wt**<br />

(Affymetrix) III III I-II brain<br />

astrocyt* glioma glioma<br />

TCGA [326] n.a. Exon 1.0 ST 397 0 0 0 10<br />

Phillips et al<br />

[390]<br />

GSE4271 U133A,B 55 21 0 0 0<br />

Freije<br />

[148]<br />

et al GSE4412 U133A,B 50 8 16 0 0<br />

Gravendeel et<br />

al [176]<br />

GSE16011 U133 Plus 2.0 141 16 66 27 0<br />

Murat<br />

[353]<br />

et al GSE7696 U133 Plus 2.0 70 0 0 0 0<br />

Gravendeel et al. described 269 samples obtained at histologic diagnosis, from which<br />

we excluded 15 containing mostly non-neoplastic tissue and four lacking survival<br />

data; n.a.=not applicable; *astrocytoma, **Non-neoplastic.<br />

tial diagnosis. For dataset GBM, we used processed (level 3) data from TCGA,<br />

consisting <strong>of</strong> one expression value per gene and sample. For the survival anal-<br />

ysis datasets and the HGG dataset, the raw microarray data was processed<br />

with the Robust Multi-chip Average (RMA) method as implemented in the<br />

Bioconductor package affy [161] and probe-gene mappings were retrieved from<br />

Ensembl 68 [146]. For genes represented by multiple probesets, expression<br />

values were averaged across probesets for randomisation tests, heatmap vi-<br />

sualisation and GNS signature score calculation. Differential expression was<br />

computed using limma [464]. Randomisation tests were conducted with the<br />

limma function geneSetTest, comparing log 2(F C) for the sets <strong>of</strong> core up- or<br />

down-regulated genes against the distribution <strong>of</strong> log 2(F C) for randomly sam-<br />

pled gene sets <strong>of</strong> the same size.<br />

Survival analysis was carried out with the R library survival. To combine<br />

expression values <strong>of</strong> multiple genes for survival prediction, an approach inspired<br />

113


5.10 Glioblastoma Pathway Construction Methods<br />

by Colman et al [105] was taken. The normalised expression values xij, where i<br />

represents the gene and j the sample, were first standardised to be comparable<br />

between genes by subtracting the mean across samples and dividing by the<br />

standard deviation, thus creating a matrix <strong>of</strong> z-scores:<br />

zij = xij − ¯xi<br />

SD(xi)<br />

(5.8)<br />

Using a set U <strong>of</strong> nU genes up-regulated in GNS lines and a set D <strong>of</strong> nD genes<br />

down-regulated in these cells, we then computed a GNS signature score sj for<br />

each sample j by subtracting the mean expression <strong>of</strong> the down-regulated genes<br />

from the mean expression <strong>of</strong> the up-regulated genes:<br />

sj = <br />

i∈U<br />

zij<br />

nu<br />

− <br />

zij<br />

nD<br />

i∈D<br />

(5.9)<br />

IDH1 mutation calls for TCGA samples were obtained from Firehose data run<br />

version 2012-07-07 [143] and data files from the study by Verhaak et al updated<br />

2011-11-28 [408].<br />

5.10 Glioblastoma Pathway Construction<br />

The pathway map was created in Cytoscape 3.0 [451]. Cytoscape is an open-<br />

source platform for complex network analysis and visualisation <strong>of</strong> network<br />

data, such as molecular interaction networks or biological pathways, that can<br />

be integrated with annotations, gene expression pr<strong>of</strong>iles and any other type<br />

<strong>of</strong> useful data. Cytoscape is available for download at www.cityscape.org. In<br />

a Cytoscape network nodes represent objects, i.e. proteins, and connecting<br />

edges represent relationships between objects, i.e. a protein’s physical inter-<br />

action with another protein. Once this basic network is laid out, attributes<br />

can be assigned to nodes and edges to help the visualisation <strong>of</strong> categories <strong>of</strong><br />

objects and types <strong>of</strong> relationships, respectively (Fig 5.7). Cytoscape networks<br />

can become extremely complex as layers <strong>of</strong> attributes are applied to nodes and<br />

edges in the form <strong>of</strong> different colours, shapes, thicknesses and other graphical<br />

features that end up representing an ever-increasing amount <strong>of</strong> biological infor-<br />

mation pertaining to that network. Finally, cytoscape-generated networks can<br />

be analysed with the use <strong>of</strong> "plugins", pieces <strong>of</strong> independent s<strong>of</strong>tware devel-<br />

oped for specific applications on Cytoscape such as graph analysis, clustering,<br />

ontology analysis, etc. All plugins can be found at aps.cytoscape.org.<br />

114


5.10 Glioblastoma Pathway Construction Methods<br />

Figure 5.7: Schematisation <strong>of</strong> a typical Cytoscape network look, with nodes and<br />

edges representing proteins and the type <strong>of</strong> interaction between them, respectively.<br />

The colour and shape <strong>of</strong> the nodes can be attributed to different characteristics <strong>of</strong> the<br />

proteins, such as their family, enzymatic activity, post-translational modifications,<br />

etc. The colour and shape <strong>of</strong> the edges can be attributed to the type <strong>of</strong> interactions<br />

between proteins, such as activating, inhibiting, coenzymatic, etc. In this example<br />

protein A and B interact to form a complex that activates protein D, also repressed<br />

by protein C, that once active can go ahead and activate protein E.<br />

A series <strong>of</strong> pre-defined "layouts" available in Cytoscape, i.e. algorithms that<br />

automatically lay a network out by generating arrangements <strong>of</strong> nodes and<br />

edges that either make it look a specific way, such as the circular, grid and<br />

hierarchical layouts, or use the attribute information as a guide, such as the<br />

attribute circle, degree sorted circle, force-directed, group attributes layouts.<br />

For our purposes we used the edge-weighed force-directed layout, also known<br />

as "biolayout" that, once generated, was manually re-adapted to obtain a best<br />

fit for image generation. In order to graph the network, the edge-weighed<br />

force-directed layout, or biolayout, uses an algorithm that minimises the en-<br />

ergy <strong>of</strong> the model by starting with an initial layout, where the positions <strong>of</strong> the<br />

nodes are randomly assigned. Then, in every iteration, the algorithm tries to<br />

improve the layout according to the energy model using the first derivation <strong>of</strong><br />

the energy function to compute a direction and a distance for the movement<br />

<strong>of</strong> each node. Since the graphs generated are large, the minimising algorithms<br />

do not carry a high complexity per iteration value. The algorithm <strong>of</strong> Barnes<br />

and Hut [42] is used in Cytoscape’s biolayout for this purpose.<br />

The glioblastoma pathway was constructed manually by integrating the infor-<br />

mation on nodes (genes) and edges (types <strong>of</strong> interactions between genes) from<br />

a variety <strong>of</strong> glioblastoma pathways found in the literature with the purpose <strong>of</strong><br />

115


5.10 Glioblastoma Pathway Construction Methods<br />

generating an all-encompassing network containing all the known interactions<br />

that are important in glioblastoma. A fundamental rule that I followed was to<br />

check that every protein interaction had been experimentally validated, via lit-<br />

erature research and the use <strong>of</strong> the BioGRID protein interaction database [62]<br />

and the IntAct molecule interaction database [205], before adding it to the<br />

pathway. If it was not the case then the interaction (edge) and proteins in-<br />

volved (nodes) were not included in the final glioblastoma pathway.<br />

The information integrated to generate the final glioblastoma pathway was<br />

taken from four different sources:<br />

· The pathway database <strong>of</strong> the Kyoto Encyclopedia <strong>of</strong> Genes and Genomes<br />

(KEGG) [3], a collection <strong>of</strong> manually drawn pathway maps;<br />

· GBMbase [162], a collection <strong>of</strong> signalling pathways in glioblastoma based<br />

on the ones described in the TCGA project [326] including a searchable<br />

archive <strong>of</strong> glioblastoma gene publications;<br />

· "Automated Network Analysis Identifies Core Pathways in Glioblas-<br />

toma" by Cerami et al 2010 [86];<br />

· "Malignant Astrocytic <strong>Glioma</strong>: Genetics, Biology, and Paths to Treat-<br />

ment" by Furnari et al 2007 [154].<br />

I also included the information from important pathways in signal transduction<br />

that are known to be specifically affected in glioma, namely:<br />

· The Mitogen-activated protein (MAP) kinase signalling cascade, taken<br />

from three sources:<br />

– The Cell Signalling Technologies website [1]<br />

– "Mitogen-activated protein (MAP) kinase pathways: regulation and<br />

physiological functions" by Pearson et al 2001 [388]<br />

– The pathway database <strong>of</strong> the Kyoto Encyclopedia <strong>of</strong> Genes and<br />

Genomes [3]<br />

· The p53 pathway, taken from three sources:<br />

– The Panther Pathway repository [5]<br />

– The pathway database <strong>of</strong> the Kyoto Encyclopedia <strong>of</strong> Genes and<br />

Genomes [4]<br />

– The Biocarta pathway archive [58]<br />

116


5.11 MicroRNA Target Prediction Analysis Methods<br />

· The Rb1 pathway "RB tumour suppressor/checkpoint signalling in re-<br />

sponse to DNA damage" from the Biocarta pathway archive [60]<br />

· The Pten pathway "PTEN dependent cell cycle arrest and apoptosis"<br />

from the Biocarta pathway archive [59]<br />

The complete pathway has a total <strong>of</strong> 238 nodes and the colour mapping<br />

is based on fold change absolute values and directions taken from the dif-<br />

ferential gene expression analysis (see Results section 6.4).The colour map-<br />

ping was performed using the VistaClara Cytoscape plug-in [234] that assigns<br />

colours to nodes matching expression data imported with the "Import At-<br />

tribute/Expression Matrix File" function <strong>of</strong> Cytoscape. The assignment is<br />

done through the sigmoid function:<br />

y =<br />

2<br />

− 1 (5.10)<br />

1 + e-sx where the value <strong>of</strong> the constant "s" determines the rate <strong>of</strong> change in the colour<br />

gradient, with smaller values allowing for the colour to ramp very gradually,<br />

and larger values bringing the sigmoid function closer to a step function with a<br />

very abrupt colour change around the values −1 < y < 1. The log 2(F C) values<br />

range between −11 < F C < 11 and a value <strong>of</strong> s=1 was applied, with darker<br />

reds and greens indicating smaller absolute values <strong>of</strong> log 2(F C), and brighter<br />

colours indicating greater ones. A pathway image with the same colour coding<br />

and gradient used in the tumour correlation heatmap (Fig 7.4), has also been<br />

generated (yellow and blue shades) to correlate the expression values <strong>of</strong> the<br />

differentially expressed genes between the two analyses.<br />

5.11 MicroRNA Target Prediction Analysis<br />

In trying to answer the question "Is a subset <strong>of</strong> prediction algorithms better<br />

at predicting than the single?" we used exon array data and microRNA mi-<br />

croarray data from a cohort <strong>of</strong> GNS cell lines that was made available to us.<br />

The candidate did not perform this analysis but rather used the data gath-<br />

ered from it in the ensemble analysis <strong>of</strong> microRNA target predictions. The<br />

exon microarray data was analysed in R using s<strong>of</strong>tware packages from the<br />

Bioconductor project [465]. Background correction, quantile normalization<br />

and calculation <strong>of</strong> probe-set expression values from fluorescence data was per-<br />

formed using the Robust Multi-chip Average (RMA) method as implemented<br />

in the affy package [161]. We used the xmapcore system, based on the earlier<br />

117


5.11 MicroRNA Target Prediction Analysis Methods<br />

exonmap package and x:map database [369], to associate microarray probesets<br />

with protein-coding genes annotated in Ensembl 58. Probeset filtering was<br />

applied such that all probe sequences were required to map to exonic gene<br />

components. We estimated the expression level <strong>of</strong> a gene as the median nor-<br />

malised intensity over its associated probe-sets. Differential gene expression<br />

was computed using the limma package [465], where statistical significance was<br />

determined with a moderated eBayes test and the resulting p-values adjusted<br />

using the FDR method. The microRNA microarray data were background<br />

corrected using the method described by Edwards et al [132] and implemented<br />

in the limma package [465]. Following least-variant set normalization with de-<br />

fault parameters [482], differential expression was computed using the limma<br />

package.<br />

118


Results<br />

119


Chapter 6<br />

Digital Transcriptome Pr<strong>of</strong>iling<br />

Contents<br />

6.1 Clinical Data . . . . . . . . . . . . . . . . . . . . . . . . . . 120<br />

6.2 Tag mapping . . . . . . . . . . . . . . . . . . . . . . . . . . 121<br />

6.3 Copy Number Aberrations . . . . . . . . . . . . . . . . . . 125<br />

6.4 Core Differentially Expressed Genes . . . . . . . . . . . . . 129<br />

6.5 Large-scale qRT-PCR Validation . . . . . . . . . . . . . . . 136<br />

6.6 Literature Mining for Differentially Expressed Genes . . . . 142<br />

6.7 Is<strong>of</strong>orm Differential Expression . . . . . . . . . . . . . . . . 144<br />

6.8 Long ncRNA Differential Expression . . . . . . . . . . . . . 157<br />

6.1 Clinical Data<br />

The clinical data available for our cell lines are somewhat limited due to pa-<br />

tient consent forms prohibiting surgeons from revealing it. However, we do<br />

know that all <strong>of</strong> our GNS cell lines are clinically classified as primary glioblas-<br />

tomas, not secondary. Consistent with this, the PCR experiments performed<br />

by Steven Pollard on our GNS cell lines to establish the presence or absence<br />

<strong>of</strong> IDH1 and IDH2 mutations found no evidence <strong>of</strong> any mutation in their re-<br />

spective loci (oral communication). In the paper by Pollard et al [404] the<br />

genetics <strong>of</strong> our GNS cell lines are described using SNP6.0 arrays, which iden-<br />

tified "classic" mutations, in particular frequent loss <strong>of</strong> CDKN2A:ARF, gains<br />

<strong>of</strong> CDK4 and general glioblastoma-pertinent gene instability. The age <strong>of</strong> the<br />

patients is described in the paper and summarised in table 6.1 below.<br />

120


6.2 Tag mapping Results<br />

Table 6.1: Summary <strong>of</strong> the available clinical data for our GNS cell lines (M=Male,<br />

F=Female, n.a=not applicable).<br />

Type <strong>of</strong> Name <strong>of</strong> Tissue Type Sex IDH1 IDH2 Patient<br />

cell line cell line mutation mutation age (years)<br />

GNS G144 GBM Primary M No No 51<br />

GNS G144ED GBM Primary M No No 51<br />

GNS G166 GBM Primary F No No 74<br />

GNS G179 Giant cell GBM Primary M No No 56<br />

NS CB541 Fetal forebrain n.a. n.a. n.a. n.a. n.a.<br />

NS CB660 Fetal forebrain n.a. n.a. n.a. n.a. n.a.<br />

6.2 Tag mapping<br />

We created one Tag-seq library per cell line and obtained between 6 and 13<br />

million sequence reads from each (Table 6.2). Every read was formed by a first<br />

sequencing primer, the 17nt tag, and a second sequencing primer in this order<br />

(Fig 6.1).<br />

Table 6.2: Summary <strong>of</strong> reads per cell line library.<br />

Type <strong>of</strong> cell line Cell line Number <strong>of</strong> reads<br />

GNS G144 7,133,520<br />

GNS G144ED 6,383,175<br />

GNS G166 13,415,402<br />

GNS G179 11,610,415<br />

NS CB541 12,103,066<br />

NS CB660 10,043,561<br />

Figure 6.1: Diagram <strong>of</strong> the construct generated by the longSAGE protocol sent to<br />

the Illumina sequencer for the final step <strong>of</strong> Tag-seq.<br />

Once the read sequences were received as FASTA files, we first extracted the<br />

17nt tags they contained, then filtered these tags and, finally, aligned them<br />

to the genome. The entire process is summarised as a diagram in figure 6.2.<br />

Firstly, the 17nt tags were extracted out <strong>of</strong> each read, separating the primer<br />

sequences from the tag sequences that actually interested us. Secondly, each<br />

tag sequence was counted and the resulting counts adjusted for sequencing and<br />

library preparation errors that might have caused highly expressed transcripts<br />

to give rise to a significant number <strong>of</strong> tags differing from the expected tag se-<br />

quence by just one base. Secondly, the reads that were not going to map onto<br />

121


6.2 Tag mapping Results<br />

Figure 6.2: Step by step diagram <strong>of</strong> the extraction, filtering and mapping phases<br />

for reads and tags. The extracted tag population is coloured with different shades <strong>of</strong><br />

grey that represent a different origin (adapter, mitochondrial or ribosomal tag). In<br />

the mapping phase the diagram to the right represents the human reference genome<br />

and some <strong>of</strong> the filtered tags (light grey) are mapping onto some known transcripts<br />

(gene A, gene B) and non-coding regions.<br />

the reference genome (adapter tags formed by the ligation <strong>of</strong> two sequencing<br />

primers, and mitochondrial RNA), as well as rRNA sequences, were filtered<br />

out. As shown in figure 6.3, on average more than 90% <strong>of</strong> tags remained un-<br />

filtered, meaning that most <strong>of</strong> the sequenced data were available for us to use<br />

in subsequent analyses, the first one being alignment to the reference genome.<br />

Finally, the tag sequences contained within the recounted and filtered reads<br />

were mapped in two complementary ways. Notice that, in order to maximise<br />

the ability <strong>of</strong> the aligner to effectively map our short tag sequences to the ref-<br />

erence genome, we included the CATG recognition site <strong>of</strong> the MmeI anchoring<br />

enzyme, at the 5’ <strong>of</strong> every tag sequence; this generated 21nt tag sequences that<br />

were used for alignment to the reference genome. For each library, we mapped<br />

the so-generated pool <strong>of</strong> 21nt tags to the human reference genome, as well as<br />

to a virtual tag-to-gene library that we assembled from already existing tag-to-<br />

gene libraries and a complementary one that we generated programmatically.<br />

The tag mapping strategy we adopted was a hierarchical one, as summarised in<br />

figure 6.4. This allowed us to generate sets <strong>of</strong> decreasing stringency - regarding<br />

the number <strong>of</strong> tags mapping to known transcripts - that were best fit for differ-<br />

122


6.2 Tag mapping Results<br />

123<br />

Figure 6.3: Proportion <strong>of</strong> tags after the steps that filter for adapter contamination, mitochondrial or rRNA tags are concluded. Values are<br />

normalised across all cell lines.


6.2 Tag mapping Results<br />

ent analyses. We first mapped all the tags obtained from the process described<br />

in fig 6.2 to known transcripts, then we mapped the remaining tags to cDNA<br />

libraries. Finally, we mapped the remaining tags to the genome for the poten-<br />

tial discovery <strong>of</strong> new unannotated transcripts. As a result <strong>of</strong> this strategy, we<br />

collected in "Ref_uniq" - our most stringent set - all the tags mapping to a<br />

single reference gene and in "Ref_best" - our second most stringent set - all<br />

the tags included in the former with the addition <strong>of</strong> tags mapping to multiple<br />

genes <strong>of</strong> which, however, only one was a reference transcript. The conservative<br />

set <strong>of</strong> tag-to-transcript mappings ("Ref_uniq") comprised 25,593 unique tags<br />

assigned to 15,103 genes, and formed our primary dataset for further anal-<br />

ysis. In addition, we created several more comprehensive tag mapping sets,<br />

by including mappings to other cDNA sequences, expressed sequence tags,<br />

internal NlaIII sites and unannotated genomic regions ("cDNA_uniq" and<br />

"cDNA_best").<br />

Figure 6.4: Diagram <strong>of</strong> the tag mapping strategy where each filtered tag is assigned<br />

to a more or less stringent set used for future analyses.<br />

As shown in figure 6.5, on average more than 55% <strong>of</strong> tags (green) in every cell<br />

line could not be classified as belonging to any <strong>of</strong> the sets identified with the<br />

124


6.3 Copy Number Aberrations Results<br />

hierarchical tag assignment process described above and summarised in fig 6.4.<br />

This showed that most <strong>of</strong> the transcriptional activity in our cell lines could not<br />

be represented by known transcripts or cDNAs, but perhaps that much <strong>of</strong> the<br />

regulation might be happening at the non-coding transcriptional level. On av-<br />

erage, in every cell line, less than 45% <strong>of</strong> tags mapped perfectly to the reference<br />

nuclear genome, i.e. the tags contained within the "Ref_uniq", "Ref_best",<br />

"cDNA_uniq" and "cDNA_best" sets in the non-green portions <strong>of</strong> fig 6.5.<br />

This indicated that almost half <strong>of</strong> the transcriptional activity detected in our<br />

cell lines could be traced back to known gene functions, giving us the possi-<br />

bility to correlate gene expression in our cell lines with, for example, known<br />

cancer pathways or other data from similar experiments. Of these 45% tags,<br />

on average 32% were specifically assigned to high-quality reference transcript<br />

models from the widely used Ensembl, RefSeq and Mammalian Gene Collec-<br />

tion (MGC) databases (i.e., belonged to the "Ref_uniq" and "Ref_best" sets).<br />

Reassuringly, other Tag-seq studies have reported similar figures [194,346].<br />

Given that glioblastoma is a heterogeneous tumour, and that our samples were<br />

taken from three distinct glioblastoma patients, we performed an analysis to<br />

observe whether any correlation existed between the cell lines (Fig 6.6) that<br />

would reinforce or weaken that knowledge. We observed the following, calcu-<br />

lating a single expression value for each gene in each cell line by summing the<br />

counts <strong>of</strong> all tags assigned to that gene: the correlation between the techni-<br />

cal replicates G144 and G144ED was the highest (Pearson R = 0.94), as was<br />

assumable since the two cell lines were derived from the same tumour but in<br />

different laboratories (it should be noted that differences between these repli-<br />

cate measurements can be due to a number <strong>of</strong> factors in the procedure from<br />

cell line establishment to tag sequencing); the correlation between the two NS<br />

cell lines was also high (R = 0.87); any correlation between GNS cell lines<br />

(G166, G179 and G144) showed larger differences in gene expression pr<strong>of</strong>iles<br />

(R ranging from 0.78 to 0.82) than the correlation between the two biological<br />

replicates or the two NS cell lines, as was to be expected since the GNS cell<br />

lines originated from histologically distinct tumours while the NS cell lines<br />

supposedly bore a wild type transcriptional activity.<br />

6.3 Copy Number Aberrations<br />

Since the observed differences in gene expression can be caused by a number<br />

<strong>of</strong> factors such as chromosomal aberrations (including rearrangements, losses<br />

125


6.3 Copy Number Aberrations Results<br />

126<br />

Figure 6.5: Proportion <strong>of</strong> filtered tags that, after alignment to the reference genome, cDNA libraries and virtual tag-to-gene libraries, are assigned<br />

to a set. Values are normalised across all cell lines.


6.3 Copy Number Aberrations Results<br />

Figure 6.6: Correlations for all combinations <strong>of</strong> cell lines: any GNS/GNS and<br />

NS/NS cell lines.<br />

and gains <strong>of</strong> DNA) and alterations in transcriptional regulation and mRNA<br />

degradation, we analysed array comparative genomic hybridization (aCGH)<br />

data for G144, G166 and G179 [404]. The candidate did not perform this<br />

analysis but the results will be shown nonetheless to give the reader a clearer<br />

understanding <strong>of</strong> the genomic identity <strong>of</strong> these cell lines. The aCGH method<br />

reveals net gains and losses in a cell population over the average ploidy. Previ-<br />

ous analysis <strong>of</strong> chromosomal alterations in G144, G179 and G166 by SKY and<br />

CGH detected several alterations, mostly whole-chromosome gains and losses,<br />

and found that the lines do not show gross chromosomal instability when cul-<br />

tured. Specifically, the CGH data for G144 revealed the deletion <strong>of</strong> PTEN on<br />

chromosome 10 [404].<br />

These findings were confirmed by our analysis <strong>of</strong> the aCGH data (Fig 6.7).<br />

We identified aberrations known to be common in glioblastoma, including<br />

gain <strong>of</strong> chromosome 7 (EGFR, which is over-expressed in 40% <strong>of</strong> glioblas-<br />

tomas [154,262], lies on chromosome 7) and losses <strong>of</strong> large parts <strong>of</strong> chromosomes<br />

10, 13, 14 and 19 in more than one GNS line, as well as focal gain <strong>of</strong> CDK4<br />

in G144 (arrow, chromosome 12 in figure 6.7) and focal loss <strong>of</strong> the CDKN2A-<br />

CDKN2B locus in G179 (arrow, chromosome 9 in figure 6.7) [52,326]. We<br />

found that both G166 and G179 had low copy numbers <strong>of</strong> chromosome 10, but<br />

could not establish a focal PTEN loss as we did for CDK4.<br />

127


6.3 Copy Number Aberrations Results<br />

Figure 6.7: The image summarises the results <strong>of</strong> the CGH arrays performed on<br />

cell lines G144, G166 and G179. Dots indicate log2 ratios for array CGH probes<br />

along the genome, comparing each GNS line to normal female DNA. The coloured<br />

segments indicate gain (red) and loss (green) calls, with colour intensity proportional<br />

to mean log2 ratio over the segment. The X chromosome was called as lost in G144<br />

and G179 because these two cell lines are from male patients; sex-linked genes were<br />

excluded from downstream analyses <strong>of</strong> aberration calls. The red segments above the<br />

grey line at zero indicate gains and red segments below it indicate losses over the<br />

entire genome.<br />

On a global level, we found a correlation between aberrations and expression<br />

levels, but this trend was modest (Fig 6.8a). Among the 459 autosomal genes<br />

that we found to be up-regulated in GNS relative to NS lines by Tag-seq,<br />

there was a clear enrichment <strong>of</strong> gains (p = 0.002 with Fisher’s exact test) and<br />

depletion <strong>of</strong> losses (p = 0.002) compared to all autosomal genes expressed in<br />

the GNS or NS lines. Down-regulated genes showed an opposite, but weaker,<br />

trend (Table 6.3, Fig 6.8b). We also observed that many <strong>of</strong> the 29 genes that<br />

were found to distinguish GNS lines from NS lines by qRT-PCR had associ-<br />

ated aberration calls that suggested their expression levels may be dictated<br />

by a loss or a gain in their copy numbers (Fig 6.8c). Overall, these results<br />

Table 6.3: Significance <strong>of</strong> the correlation found between CNAs and expression levels<br />

measured with Fisher’s exact test (p-value).<br />

Gains Losses<br />

Up-regulated 0.002163 0.002286<br />

Down-regulated 0.06997 0.4257<br />

suggest that copy number changes are a significant cause <strong>of</strong> the observed gene<br />

expression changes. However, other factors may be more important, because<br />

only a minority <strong>of</strong> up-regulated genes (21%) showed evidence <strong>of</strong> gains. Thus,<br />

128


6.4 Core Differentially Expressed Genes Results<br />

regulatory changes and other alterations not detectable by aCGH, such as bal-<br />

anced translocations and small mutations, likely play a major role in shaping<br />

the GNS transcriptome.<br />

Figure 6.8: (a) Curves show distributions <strong>of</strong> expression level differences between<br />

GNS and NS lines, stratified by aberration calls. The distributions for genes in segments<br />

without aberrations (neutral) peak near the zero mark, corresponding to an<br />

equal expression level in GNS and NS lines. Conversely, genes in lost and gained<br />

regions tend to be expressed at lower and higher levels, respectively. In each plot,<br />

log2(FC) is computed between the indicated GNS line and the mean <strong>of</strong> the two NS<br />

lines, and capped at (-8, 8) for visualisation purposes. To obtain robust FC distributions,<br />

genes with low expression (< 25 tpm) in both GNS and NS conditions were<br />

excluded; consequently, between 6,014 and 6,133 genes underlie each plot. (b) For<br />

each <strong>of</strong> the three gene sets listed in the legend (inset), bars represent the percentage<br />

<strong>of</strong> genes with the indicated copy number status. (c) Aberration calls for the 29 genes<br />

that were found to distinguish GNS from NS lines by qRT-PCR. Circles indicate<br />

focal (< 10 Mb) aberrations; boxes indicate larger chromosomal segments.<br />

6.4 Core Differentially Expressed Genes<br />

To identify genes with differing expression between GNS and NS cells, we used<br />

the Bioconductor package DESeq on the three GNS lines G144, G166 and<br />

G179, and the two NS lines CB541 and CB660. The DESeq package, unlike<br />

its predecessor edgeR, uses a method whose core assumption is that the mean<br />

is a good predictor <strong>of</strong> the variance, which implies that, for a given distribution<br />

<strong>of</strong> genes with similar expression levels, the variance across replicates will be<br />

similar. Given this assumption, this method developed by Simon Anders [21]<br />

estimates a function that predicts the variance from the mean by calculating<br />

the sample mean and variance for each gene within replicates, and then fitting<br />

129


6.4 Core Differentially Expressed Genes Results<br />

a curve to this data. With the diagnostic plot shown in fig 6.9 we validated that<br />

the estimates <strong>of</strong> the single gene variance functions followed the empirical vari-<br />

ance well enough, as indicated by the red line representing the local regression<br />

fit, even though the spread <strong>of</strong> the single gene variance values is considerable,<br />

as one should expect given that each variance value is estimated from just<br />

four values. Having estimated and verified the variance-to-mean dependance,<br />

Figure 6.9: Plot <strong>of</strong> the estimates for each gene <strong>of</strong> the variance against the base<br />

levels, i.e. the count value for a tag divided by the total number <strong>of</strong> counts. The red<br />

line represents the fit from the local regression.<br />

we then proceeded to look for differentially expressed genes using the DESeq<br />

package function nbinomTest. With this function we generated the following<br />

values for each gene: the mean expression level, as a joint estimate between<br />

conditions "N" (normal) and "T" (tumour) and as a separate estimate for each<br />

condition, the fold change (FC) from condition N to T, the natural logarithm<br />

(Ln) <strong>of</strong> the fold change, and the p-value for the statistical significance <strong>of</strong> this<br />

change. The p-adjusted value is also computed by the nbinomTest function<br />

to adjust the p-value for multiple testing with the Benjamin-Hochberg pro-<br />

cedure, which controls the FDR. We first plotted the computed fold changes<br />

against the mean values and coloured in red the genes that were significant<br />

at 1% FDR (Fig 6.10). Interestingly, these genes seemed to cluster at higher<br />

values <strong>of</strong> the mean and absolute value <strong>of</strong> Ln(FC), indicative <strong>of</strong> gene expression<br />

130


6.4 Core Differentially Expressed Genes Results<br />

levels that were coherently higher across all GNS versus NS cell lines, or all<br />

NS versus GNS cell lines. At an FDR <strong>of</strong> 10%, this analysis revealed 485 genes<br />

Figure 6.10: Plot <strong>of</strong> the fold change computed with the DESeq package function<br />

nbinomTest versus the mean for the contrast <strong>of</strong> the two conditions "N" (normal) and<br />

"T" (tumour). The red genes are significant at 1% FDR.<br />

to be expressed at a higher average level in GNS cells (up-regulated) and 254<br />

genes to be down-regulated (see Appendix A.1). To discern genes that are<br />

consistently up- or down-regulated among the GNS lines compared to the NS<br />

lines, and thus capture major gene expression changes common to G144, G166<br />

and G179, we set strict criteria on fold changes and tag counts requiring that:<br />

1. each GNS line show at least a two-fold change compared to each NS line,<br />

with the direction <strong>of</strong> change being consistent among the cell lines;<br />

2. an absolute expression level above 30 tpm in each GNS line for up-<br />

regulated genes or each NS line for down-regulated genes.<br />

This stringent approach yielded 32 up-regulated and 60 down-regulated genes,<br />

in the following referred to as strictly up- and down-regulated genes, respec-<br />

tively, or "core" differentially expressed genes (Table 6.4).<br />

131


6.4 Core Differentially Expressed Genes Results<br />

Table 6.4: Table <strong>of</strong> genes (alphabetical order) with large expression changes common<br />

to the GNS lines G144, G166 and G179, relative to the normal NS lines CB541<br />

and CB660, resulting from filtering with stringent criteria the differentially expressed<br />

genes at 10% FDR.<br />

Gene Entrez Differential expression results Normalised tag counts<br />

symbol gene ID log 2(F C) FDR G144ED G144 G166 G179 CB541 CB660<br />

ADD2 119 6.32 5.1E-04 66.4 91.9 103.1 288.2 0.0 4.1<br />

ATP1A2 477 -6.16 1.3E-08 39.0 37.5 0.6 0.0 471.0 1346.4<br />

BACE2 25825 7.97 5.5E-11 227.5 423.1 253.4 468.2 0.0 2.7<br />

C10orf11 83938 3.82 1.7E-03 195.7 603.7 175.1 152.8 20.5 23.6<br />

C5orf13 9315 -3.29 4.7E-03 230.2 162.6 55.9 234.9 1985.9 962.7<br />

C9orf125 84302 -3.78 6.1E-04 73.9 59.0 3.6 8.0 508.5 148.1<br />

C9orf64 84267 7.88 5.3E-10 114.7 156.8 164.1 752.5 2.9 0.0<br />

CA12 771 -4.65 9.7E-05 45.9 28.1 22.6 30.3 945.6 415.1<br />

CACNG8 59283 -4.77 5.7E-05 33.1 28.3 3.9 2.6 405.1 220.9<br />

CCND2 894 -3.98 9.3E-05 158.0 125.2 0.6 0.0 827.8 496.4<br />

CD74 972 7.11 7.9E-13 3942.1 2107.8 93.4 1427.2 8.4 9.1<br />

CD9 928 4.87 2.7E-06 1058.3 1082.4 396.5 189.3 4.8 32.9<br />

CEBPB 1051 3.29 5.9E-03 106.7 155.6 552.5 270.9 36.7 30.2<br />

CHCHD10 400916 3.70 9.0E-04 844.2 713.0 523.9 457.9 28.4 58.8<br />

CTSC 1075 3.55 1.0E-03 937.2 731.4 311.7 1499.9 10.3 133.2<br />

CXXC4 80319 -4.84 1.2E-03 23.5 21.1 0.0 0.0 219.5 200.8<br />

DDIT3 1649 4.40 4.6E-05 933.6 1345.3 748.9 250.5 26.9 47.3<br />

DNER 92737 -3.89 1.8E-03 90.5 523.2 9.3 847.4 8865.3 4813.8<br />

DTX4 23220 -5.92 4.6E-05 6.8 7.8 3.2 0.0 128.8 312.5<br />

EDA2R 60401 -5.58 7.6E-04 1.4 6.2 9.7 0.0 306.7 208.8<br />

EPDR1 54749 3.09 5.6E-03 310.9 269.0 739.5 319.0 20.3 82.3<br />

FAM38B 63895 -Inf 5.5E-11 0.0 0.0 0.0 0.0 778.6 166.8<br />

FAM69A 388650 3.68 2.2E-03 513.2 393.1 155.0 439.3 27.9 23.3<br />

FBLN2 2199 -Inf 2.0E-07 0.0 0.0 0.0 0.0 232.1 117.5<br />

FOXG1 2290 3.30 6.8E-03 445.0 505.5 104.5 137.4 0.0 50.8<br />

FOXJ1 2302 -4.19 3.6E-03 4.9 3.1 4.4 27.8 254.8 175.8<br />

FUT8 2530 3.32 4.6E-03 666.0 999.8 609.6 2352.7 95.8 168.6<br />

GABBR2 9568 -9.50 1.2E-13 0.0 0.0 5.6 0.0 2569.7 143.1<br />

GPR158 57512 -4.55 9.9E-06 103.8 117.4 0.0 53.3 2311.0 354.9<br />

GRIA1 2890 -4.42 4.2E-05 33.1 68.7 2.7 73.4 1780.2 292.7<br />

HLA-DRA 3122 5.68 6.6E-07 9744.0 5434.9 138.4 270.5 57.4 19.2<br />

HMGA2 8091 -5.68 3.7E-05 0.0 0.0 0.0 23.7 233.3 576.3<br />

HOXD10 3236 Inf 3.9E-11 77.0 145.9 115.3 578.2 0.0 0.0<br />

IL17RD 54756 -4.38 8.0E-04 25.5 17.5 9.8 15.3 364.6 230.4<br />

IRX2 153572 -4.65 7.2E-04 0.0 0.0 0.0 28.9 370.0 111.1<br />

KALRN 8997 -5.25 1.9E-04 0.0 6.3 2.5 7.1 302.9 106.2<br />

KCTD12 115207 -4.77 4.0E-03 48.7 16.9 10.1 54.5 1349.0 147.8<br />

LAMA2 3908 -5.21 4.4E-06 47.3 25.2 8.0 2.6 730.2 149.0<br />

LGALS3 3958 6.44 6.1E-07 2062.3 3254.0 2538.7 134.8 32.1 14.3<br />

LMO2 4005 -4.82 2.5E-04 20.3 21.2 0.0 4.9 134.6 369.0<br />

LMO3 55885 -Inf 1.0E-08 0.0 0.0 0.0 0.0 136.9 446.0<br />

LMO4 8543 3.80 5.0E-04 553.8 1311.2 455.5 1149.8 16.7 122.5<br />

LPAR6 10161 -3.62 5.6E-03 29.8 33.8 39.6 0.0 395.9 211.3<br />

LUM 4060 -4.26 3.2E-03 0.0 0.0 110.9 0.0 507.4 897.7<br />

LYST 1130 5.73 3.5E-09 126.7 180.8 284.0 548.6 10.3 2.1<br />

MAF 4094 -4.79 9.6E-04 14.9 7.1 16.0 0.0 282.4 150.4<br />

MAN1C1 57134 3.59 1.8E-03 556.5 478.6 107.6 378.3 7.6 45.2<br />

MAP6 4135 -3.25 7.6E-03 2.3 32.9 0.6 111.4 629.4 288.0<br />

MBP 4155 6.47 2.7E-09 118.6 402.5 103.3 303.0 0.0 6.3<br />

MMP17 4326 3.58 1.0E-03 1173.5 864.3 422.6 340.8 5.1 84.8<br />

MMRN1 22915 -8.80 1.6E-06 0.0 0.0 1.3 0.0 279.4 93.2<br />

MN1 4330 -7.23 3.0E-07 21.6 4.2 0.6 0.0 146.8 384.1<br />

MT2A 4502 4.13 1.3E-03 2039.2 1924.4 8308.0 5237.5 285.0 301.4<br />

MYL9 10398 -3.85 4.3E-03 3.8 3.1 110.0 8.5 796.2 370.3<br />

NDN 4692 -3.46 2.3E-03 105.3 84.2 0.0 0.0 214.4 405.4<br />

NELL2 4753 -3.25 1.0E-02 117.3 173.6 1.9 380.9 1073.3 2440.6<br />

NKX2-1 7080 -5.52 1.6E-06 4.1 17.2 0.0 0.0 425.3 103.6<br />

NNMT 4837 3.74 5.0E-04 519.9 172.4 918.1 175.6 15.2 47.4<br />

NPTX2 4885 -6.47 1.8E-06 0.0 0.0 6.9 4.8 422.0 272.4<br />

NTN1 9423 -4.22 1.5E-04 95.8 239.9 22.4 250.6 5458.0 966.2<br />

NTRK2 4915 3.61 2.6E-03 90.7 163.9 174.8 219.5 0.0 30.3<br />

ODZ2 57451 -3.68 6.5E-03 27.0 25.0 2.5 36.0 212.8 329.5<br />

PDE1C 5137 4.13 1.3E-04 578.2 430.3 1117.8 981.4 45.7 50.8<br />

PDZRN3 23024 -4.40 9.4E-04 18.4 22.1 12.6 3.0 278.2 251.3<br />

PEG3 5178 -5.14 6.0E-08 105.7 130.6 8.5 0.0 2681.0 594.6<br />

PI15 51050 -6.45 9.3E-08 20.2 3.3 2.5 5.5 397.8 262.2<br />

PLA2G4A 5321 7.34 1.9E-12 909.7 1226.5 483.7 755.9 0.0 10.1<br />

PLCH1 23007 -3.41 2.4E-03 152.0 134.0 83.3 118.9 2002.9 384.3<br />

132


6.4 Core Differentially Expressed Genes Results<br />

Gene Entrez Differential expression results Normalised tag counts<br />

symbol gene ID log 2(F C) FDR G144ED G144 G166 G179 CB541 CB660<br />

PLS3 5358 3.83 3.1E-04 360.2 776.6 476.6 367.2 0.0 75.6<br />

PMEPA1 56937 4.36 5.4E-05 240.8 123.3 621.8 297.2 11.7 22.1<br />

PRSS12 8492 4.80 2.1E-04 218.6 119.9 151.9 156.0 0.0 10.2<br />

PTEN 5728 -5.00 2.9E-03 2.7 0.0 14.8 0.0 191.7 126.9<br />

RAB6B 51560 -3.51 2.2E-03 71.7 52.1 29.4 38.4 754.8 160.7<br />

RGS5 8490 -4.82 2.9E-04 4.1 13.5 6.9 3.0 343.5 112.5<br />

RTN1 6252 -5.34 5.7E-05 8.1 5.5 0.6 19.3 385.7 286.3<br />

S100A6 6277 6.04 5.1E-14 2259.3 324.2 5709.9 512.7 12.3 53.4<br />

SALL2 6297 -4.13 1.7E-03 72.9 38.6 16.5 11.6 194.2 581.9<br />

SDC2 6383 -3.56 2.9E-03 96.7 41.3 53.0 98.9 1015.1 502.4<br />

SEMA6A 57556 -3.48 3.3E-03 68.1 50.0 3.1 0.0 257.1 140.0<br />

SIX3 6496 -5.50 2.9E-06 2.7 26.1 0.0 0.0 203.9 594.6<br />

SLC4A4 8671 -4.49 6.1E-05 36.0 38.9 13.1 18.8 925.2 145.3<br />

SLCO1C1 53919 -Inf 5.9E-07 7.7 0.0 0.0 0.0 166.2 151.8<br />

SLCO2A1 6578 -5.61 1.0E-05 38.8 15.5 0.0 0.0 213.0 297.0<br />

SLIT2 9353 -7.18 1.5E-03 1.4 0.0 0.7 5.2 178.8 400.8<br />

SPARCL1 8404 -4.03 1.5E-03 44.2 39.0 4.8 282.9 1448.1 2097.6<br />

ST6GALNAC5 81849 -8.25 3.4E-09 8.9 0.0 0.0 5.8 261.3 899.5<br />

SULF2 55959 3.31 5.3E-03 749.0 891.0 253.5 581.6 43.3 72.4<br />

SYNM 23336 -4.23 3.3E-03 1.4 0.0 0.0 36.4 238.8 212.8<br />

TAGLN 6876 -7.06 4.4E-11 1.9 3.1 9.4 4.1 1089.6 384.4<br />

TES 26136 -Inf 2.5E-09 0.0 0.0 0.0 0.0 433.0 184.2<br />

TRAM1L1 133022 -5.13 2.5E-03 0.0 0.0 0.0 11.3 121.7 146.3<br />

TUSC3 7991 -3.74 5.9E-03 0.0 0.0 5.9 75.0 379.0 333.7<br />

In a wide literature search that we performed on each gene <strong>of</strong> the core set,<br />

we found that many <strong>of</strong> both the up-and down-regulated genes appeared in<br />

pathways known to be affected in glioma. In the up-regulated cohort we found:<br />

· S100A6 encodes an EF-hand calcium binding protein linked to the reg-<br />

ulation <strong>of</strong> cell proliferation, cytoskeletal organisation and metastasis.<br />

S100A6 is up-regulated in several malignancies. In gliomas, S100A6<br />

appears to be specifically expressed by cancer stem cells [80,186].<br />

· HLA-DRA and HLA-DRB form one <strong>of</strong> the MHC class II receptors, which<br />

present extracellular antigens to T-helper cells. CD74 or "invariant<br />

chain" takes the place <strong>of</strong> the extracellular peptide before MHC class<br />

II receptors are mature in the lumen <strong>of</strong> the endoplasmic reticulum (ER).<br />

Increased antigen presentation can help the immune system target cancer<br />

cells. HLA-DRA and HLA-DRB are induced by inflammatory cytokines<br />

and up-regulated in many tumours [421].<br />

· MBP encodes myelin basic protein, so it’s expression in GNS lines prob-<br />

ably reflects their glial nature.<br />

· PMEPA1 can act as an oncogene by affecting the PI3K pathway, in-<br />

cluding PTEN down-regulation. PMEPA1 is up-regulated in multiple<br />

tumours, but there are no reports implicating it in glioma [460,531].<br />

· LMO4 encodes a transcriptional regulator involved in the development<br />

<strong>of</strong> multiple organs, including the CNS. LMO4 expression is elevated in<br />

133


6.4 Core Differentially Expressed Genes Results<br />

several cancers. LMO4 is well-characterised as an oncogene in breast<br />

cancer, but has not been studied in glioma. LMO4 is regulated trough<br />

the PI3K pathway, which is known to be affected in glioma [?,340].<br />

· The transcription factor gene CEBPB is a well-characterised glioma<br />

oncogene, recently suggested to be a master regulator <strong>of</strong> a mesenchy-<br />

mal gene expression signature associated with poor prognosis [85].<br />

· CD9 is a membrane protein implicated in invasion [253] and regulation<br />

<strong>of</strong> EGF receptor activation [455]. Its expression in astrocytic tumours<br />

correlates with malignancy [220].<br />

· MT2A encodes a metallothionein found to suppress apoptosis in cancer<br />

cell lines [110], potentially by causing TP53 to misfold [411]. Inversely<br />

correlated expression <strong>of</strong> MT2A and TP53 proteins have been observed<br />

in glioblastoma specimens [306].<br />

· NTRK2 encodes a receptor for brain-derived neurotrophic factor, pro-<br />

moting differentiation, proliferation and survival [118]. In several tumour<br />

types, its expression correlates with poor prognosis and metastasis [118]<br />

and the protein has been detected in a subset <strong>of</strong> cells in astrocytomas [29].<br />

As shown in figure 6.11 the Tag-seq data available for NTRK2 happens<br />

to identify a long and a shorter is<strong>of</strong>orm for this gene, potentially the one<br />

lacking the kinase domain that has also been implicated in the regulation<br />

<strong>of</strong> astrocyte morphology [366].<br />

· FOXG1, a transcription factor gene involved in brain development, is<br />

commonly amplified in the childhood brain tumour medulloblastoma [9].<br />

FOXG1 has been proposed to act as an oncogene in glioblastoma as well,<br />

by suppressing growth-inhibitory effects <strong>of</strong> TGFβ [448].<br />

The set <strong>of</strong> core up-regulated genes also includes multiple genes suggested to<br />

play a role in other neoplasias, but for which we failed to find any studies<br />

implicating them in glioma. For five <strong>of</strong> these, previous studies have pointed to<br />

a role in motility and invasion: PLS3 [26,163], LMO4 [475], BACE2 [243,499],<br />

CTSC [113,530,551] and MMP17 [88,430]. Further examples <strong>of</strong> genes impli-<br />

cated in other cancers include the putative growth factor EPDR1 [8,360], which<br />

is highly expressed in some progenitor cell types [178], and PMEPA1, which can<br />

act as an oncogene by affecting the PI3K pathway and PTEN stability [460].<br />

A subset <strong>of</strong> the core up-regulated genes have to the best <strong>of</strong> our knowledge<br />

134


6.4 Core Differentially Expressed Genes Results<br />

Figure 6.11: NTRK2 is a brain-derived neurotrophic factor detected in the literature<br />

in a subset <strong>of</strong> cells in astrocytomas [29] and found in our Tag-seq dataset<br />

by mappings <strong>of</strong> tags on the shortest is<strong>of</strong>orm. Three tracks are visible in the figure:<br />

at the top, a customised track composed <strong>of</strong> 12 sub-tracks representing the expression<br />

levels <strong>of</strong> our GNS cell lines and NS cell lines in red - for tag mappings on the<br />

minus strand - and blue - for tag mappings on the plus strand. The level <strong>of</strong> tag<br />

expression is indicated by the thickness <strong>of</strong> the line representing the mapped tag; at<br />

the centre a track representing UCSC genes, RefSeq genes and Ensembl genes; at<br />

the bottom a track showing the zoom in <strong>of</strong> the central track region where the tag<br />

mapping takes place. The thicker lines in the bottom track indicate the 3’UTRs<br />

<strong>of</strong> the gene. The tags representing the NTRK2 gene map onto a region that identifies<br />

both the shorter - potentially the one that lacks the kinase domain - and a<br />

longer is<strong>of</strong>orm. Adapted from the UCSC genome browser (our Tag-seq track for<br />

hg19 is added by pasting the following line into the custom track box http://gns:<br />

zed9epre@www.ebi.ac.uk/\~engstrom/gns/tracks/hg19/gns\_tpm.bg.gz).<br />

135


6.5 Large-scale qRT-PCR Validation Results<br />

not been implicated in cancer. One <strong>of</strong> these putative oncogenes is PLA2G4A,<br />

which encodes a cytoplasmic phospholipase (cPLA2α) involved in production<br />

<strong>of</strong> lipid signaling molecules with mitogenic and pro-inflammatory effects [192].<br />

Another example is PDE1C, encoding a cyclic nucleotide phosphodiesterase<br />

that controls cellular levels <strong>of</strong> cAMP and cGMP and may regulate cell prolif-<br />

eration [126]. PDE1C expression has been observed in other glioma cell lines,<br />

but also in non-malignant astrocytes from rat brain [508]. Another putative<br />

novel oncogene is C10orf11. Disruptions <strong>of</strong> this gene have been associated with<br />

mental retardation, consistent with a developmental role [501]. Although the<br />

human gene is poorly characterised, the orthologous gene in Ciona intestinalis<br />

is required for embryogenesis and is a component <strong>of</strong> the Wnt/β-catenin sig-<br />

naling pathway, which is <strong>of</strong>ten activated in cancer [519]. Similar observations<br />

were made for core set <strong>of</strong> down-regulated genes:<br />

· the TES gene (testis derived transcript) is a LIM-domain gene at frag-<br />

ile site found to be implicated in glioblastoma as well as prostate and<br />

head/neck cancer [294,349,444];<br />

· the DNER gene (delta/notch-like EGF repeat containing) is a putative<br />

growth factor receptor found to be implicated in glioblastoma neuro-<br />

sphere formation reduce adipocyte cell proliferation [381,478];<br />

· the PTEN gene is a tumour suppressor <strong>of</strong>ten down-regulated and/or<br />

deleted in gliomas [326];<br />

· the TUSC3 gene (tumour suppressor candidate 3) is implicated in ovarian<br />

cancer [90].<br />

6.5 Large-scale qRT-PCR Validation<br />

To assess the accuracy <strong>of</strong> Tag-seq expression level estimates and investigate<br />

gene activity in a larger panel <strong>of</strong> cell lines, we assayed 82 core differentially ex-<br />

pressed genes in 16 GNS cell lines (derived from independent tumour samples)<br />

and 6 normal NS cell lines (Appendix tables A.3 and A.4), by qRT-PCR, using<br />

custom-designed TaqMan micr<strong>of</strong>luidic arrays. The 82 validation targets were<br />

selected from the 92 core differentially expressed genes based on the availabil-<br />

ity <strong>of</strong> TaqMan probes and considering prior knowledge <strong>of</strong> gene functions. For<br />

the cell lines assayed by both Tag-seq and qRT-PCR, measurements agreed<br />

remarkably well between the two technologies: the median Pearson correlation<br />

136


6.5 Large-scale qRT-PCR Validation Results<br />

for expression pr<strong>of</strong>iles <strong>of</strong> individual genes was 0.91 and the differential expres-<br />

sion calls were corroborated for all 82 genes. Figure 6.12 shows the relationship<br />

between the qRT-PCR and the Tag-seq measurements in three different panels<br />

for four different genes that were picked to highlight the dynamic range <strong>of</strong> the<br />

qRT-PCR and the Tag-seq platforms: the matrix metallopeptidase is a mem-<br />

brane protein involved in the breakdown <strong>of</strong> the ECM and a known oncogene in<br />

glioblastoma primary tumours [183]; the damage-inducible transcript DDIT3<br />

is induced in response to a range <strong>of</strong> compounds that sensitises glioma cells<br />

to apoptosis [216]; MYL9 encodes for one <strong>of</strong> the myosin light chains and its<br />

link with glioblastoma has yet to be established (possibly acting via regulation<br />

<strong>of</strong> the cell migration process); the mannosidase gene MAN1C1 is one <strong>of</strong> the<br />

enzymes that hydrolyse the mannose sugar but has to yet be implicated in<br />

glioblastoma - although its sister gene MAN2C1 has been associated with a<br />

reduction <strong>of</strong> PTEN functionality in prostate cancer [190]. In panel a <strong>of</strong> figure<br />

6.12 the high overall correlation between the two platforms is highlighted by<br />

the four data points that lie nearly on a straight line (R = 0.91); panel b shows<br />

the correlation values computed as the correlation between normalised Ct val-<br />

ues and tag counts across the five GNS cell lines (table <strong>of</strong> correlation values<br />

available in Appendix A.4); panel c shows the mean fold-change between the<br />

genes expressed in the GNS and NS cell lines as measured in each <strong>of</strong> the two<br />

platforms plotted so that the influence <strong>of</strong> outliers can be reduced thanks to<br />

the averaging.<br />

Across the entire panel <strong>of</strong> cell lines, 29 <strong>of</strong> the 82 genes showed statistically<br />

significant differences between GNS and NS lines at an FDR <strong>of</strong> 5% (Fig 6.13).<br />

This set <strong>of</strong> 29 genes distinguishes GNS from NS lines and may, therefore, have<br />

broad relevance for elucidating tumourigenic properties <strong>of</strong> GNS cells. Figure<br />

6.14 shows a histogram for each <strong>of</strong> the 29 genes found to be relevant in distin-<br />

guishing GNS from NS cell lines. The height <strong>of</strong> the histogram bar represents<br />

the expression level measured via qRT-PCR for that gene and its designated<br />

probe (see title <strong>of</strong> each histogram for probe’s name). Each expression level is<br />

calculated as the arithmetic mean across all biological replicates for each cell<br />

line. The histogram bar colours comply to the ones selected for GNS and NS<br />

cell lines in the bar displayed at the top <strong>of</strong> fig 6.14, distinguishing the assayed<br />

GNS and NS cell lines in two separate groups along the x-axis.<br />

137


6.5 Large-scale qRT-PCR Validation Results<br />

Figure 6.12: Expression estimates correlate well between Tag-seq and qRT-PCR.<br />

(a) Expression values for MMP17, DDIT3, MYL9 and MAN1C1 in two GNS and<br />

two NS lines assayed by both technologies. Note the near-perfect agreement between<br />

Tag-seq (x-axis) and qRT-PCR (y-axis) over a wide dynamic range. (b) Histogram <strong>of</strong><br />

correlation between Tag-seq and qRT-PCR for each <strong>of</strong> the 82 genes measured by qRT-<br />

PCR. (c) Fold-change estimates (indicating expression level in GNS lines relative to<br />

NS lines) from Tag-seq and qRT-PCR for the 82 genes. qRT-PCR confirmed greater<br />

than two-fold difference in expression (dashed lines at y = 1) for all genes.<br />

Figure 6.13: Heatmap <strong>of</strong> 29 genes differentially expressed between 16 GNS and 6<br />

NS cell lines. Colours indicate δδCt expression values, i.e. normalised expression<br />

on a log 2 scale where zero corresponds to the average expression between the two<br />

groups (GNS and NS).<br />

138


6.5 Large-scale qRT-PCR Validation Results<br />

139


6.5 Large-scale qRT-PCR Validation Results<br />

140


6.5 Large-scale qRT-PCR Validation Results<br />

141


6.6 Literature Mining for Differentially Expressed Genes Results<br />

Figure 6.14: Expression levels <strong>of</strong> the 29 genes distinguishing GNS from NS lines,<br />

measured by qRT-PCR and presented as percent <strong>of</strong> NS geometric mean. Two histograms<br />

reveal the data for the two probes used to measure the expression levels <strong>of</strong><br />

MYL9.<br />

6.6 Literature Mining for Differentially Expressed<br />

Genes<br />

An important assessment that adds value to our pathway analysis, is the clas-<br />

sification <strong>of</strong> every differentially expressed gene (FDR< 10%) as a gene known<br />

142


6.6 Literature Mining for Differentially Expressed Genes Results<br />

to be associated with glioblastoma, cancer or yet unlinked to any <strong>of</strong> these. I<br />

extracted the information held in several literature navigating resources, such<br />

as BioGraph [280], GBMbase [162] and iHOP [197] using a script described in<br />

detail in the Methods section and reported in Appendix B, and classified each<br />

gene in one <strong>of</strong> the following categories:<br />

· Extensive amount <strong>of</strong> literature implicates the gene in glioblastoma;<br />

· Limited amount <strong>of</strong> literature implicates the gene in glioblastoma;<br />

· Unknown to be implicated in glioblastoma, but known in other cancers;<br />

· Unknown to be implicated in any type <strong>of</strong> cancer.<br />

The information retrieved by the script for each <strong>of</strong> the differentially expressed<br />

genes was assigned to one <strong>of</strong> the four categories described above (Appendix<br />

A.2). The data outputted from this query is summarised in table 6.5.<br />

Table 6.5: Differentially expressed genes (F DR < 10%) assigned to a four-tier<br />

classification system. The unique total refers to the unique list <strong>of</strong> all the genes in<br />

each category.<br />

Category Number<br />

Extensive amount <strong>of</strong> literature implicating the gene in GBM 238<br />

Limited amount <strong>of</strong> literature implicating the gene in GBM 94<br />

Unknown to be implicated in GBM, but known in other cancers 199<br />

Unknown to be implicated in any type <strong>of</strong> cancer 232<br />

Unique Total 748<br />

<strong>of</strong> genes<br />

A stripped down version <strong>of</strong> the complete table available in Appendix A.2,<br />

highlights only the genes with limited evidence or no evidence <strong>of</strong> implication<br />

in glioblastoma that appear in our integrated glioblastoma pathway (Table<br />

6.6). Many <strong>of</strong> these genes had not been directly implicated in glioma, but they<br />

participate in glioma-related pathways and they differ in expression between<br />

GNS and NS lines. Thus, by considering expression changes in a pathway<br />

context, we identified additional candidate glioblastoma genes, such as the<br />

putative cell adhesion gene ITGBL1 [50], the orphan nuclear receptor NR0B1,<br />

which is strongly up-regulated in G179 and is known to be up-regulated and<br />

mediate tumour growth in Ewing’s sarcoma [158], and the genes PARP3 and<br />

PARP12 which belong to the PARP family <strong>of</strong> ADP-ribosyl transferase genes<br />

involved in DNA repair. The up-regulation <strong>of</strong> these PARP genes in GNS cells<br />

143


6.7 Is<strong>of</strong>orm Differential Expression Results<br />

may have therapeutic relevance, as inhibitors <strong>of</strong> their homolog PARP1 are in<br />

clinical trials for brain tumours [270]. Our comparison between GNS and NS<br />

cell lines, thus, highlights genes and pathways that are known to be affected in<br />

glioma as well as novel candidates, suggesting that the GNS vs NS comparison<br />

is a promising approach for further understanding molecular aspects <strong>of</strong> glioma.<br />

6.7 Is<strong>of</strong>orm Differential Expression<br />

To establish Tag-seq as a sensitive technique for the identification <strong>of</strong> tran-<br />

script is<strong>of</strong>orms differentially expressed between our GNS and NS libraries, we<br />

performed a parametric and a non-parametric test on the genes identified by<br />

multiple tag mappings using the "Ref_best" collection (see Fig 6.5).<br />

With the non-parametric χ 2 test, 2,682 is<strong>of</strong>orms were found to be differentially<br />

expressed as detected by the sum over all tags for each is<strong>of</strong>orm. With the para-<br />

metric approach, in which the logarithmic ratio <strong>of</strong> expression method adapted<br />

from Morrissy et al [346] was used, we computed a ratio change for each pair<br />

<strong>of</strong> tags that identified an is<strong>of</strong>orm, so that a total <strong>of</strong> 2,040 differentially ex-<br />

pressed is<strong>of</strong>orms were detected. The two methods share a total <strong>of</strong> 1,454 genes<br />

that have differentially expressed is<strong>of</strong>orms in the GNS cell lines with respect<br />

to the NS cell lines at significant levels (p-value2 for parametric), with 1,228 genes uniquely identified by the logarithmic<br />

method and 586 genes uniquely identified by the non-parametric method.<br />

In the attempt to focus on is<strong>of</strong>orms relevant to the pathways affected in<br />

glioma, we overlaid both lists <strong>of</strong> differentially expressed is<strong>of</strong>orms on the in-<br />

tegrated glioblastoma pathway described in section 7.5 and shown in figure<br />

7.8. This highlighted the presence <strong>of</strong> 48 matching genes from the logarithmic<br />

(non-parametric) method and 57 matching genes from the parametric χ 2 test<br />

method, with 35 genes shared between the two methods. Each <strong>of</strong> the 35 genes,<br />

as well as the 13 genes identified exclusively through the logarithmic method<br />

and the 22 genes identified exclusively through the parametric χ 2 test method,<br />

were manually scrutinised on the University <strong>of</strong> California Santa Cruz (UCSC)<br />

genome browser by adding our tag mappings as a custom track together with<br />

the UCSC gene track and the gene predicted Ensembl track, both in "full"<br />

mode for the NCBI37.2/Ensembl 64 human genome assembly. For the follow-<br />

144


6.7 Is<strong>of</strong>orm Differential Expression Results<br />

Table 6.6: Genes with limited evidence or no evidence <strong>of</strong> implication in glioblastoma<br />

that appear in our main glioblastoma pathway. In "Others" the genes with an<br />

important physiological role in the brain that do not appear in our glioblastoma<br />

pathway are listed.<br />

Gene* Log2(FC)** Association Implications in Citations<br />

with glioma other neoplasms<br />

CACNA1A 7.1 None Prostate cancer (mouse model) [235]<br />

CACNA1C -8.2 None Liver cancer [25]<br />

CACNG7 -2.6 None None -<br />

CACNG8 -2.6 None None -<br />

CAMK1D -2.4 None Breast cancer [51]<br />

CPLX2 6.4 None None -<br />

DDIT3<br />

(CHOP,<br />

GADD153)<br />

4.4 Limited General (cellular stress response) [218,227,330,415]<br />

DUSP16 4.2 None Burkitt’s lymphoma [265]<br />

FGF19 - None Liver, lung and colon cancer [119]<br />

ITGA4<br />

(CD49D)<br />

3.0 Limited Chronic lymphocytic leukaemia,<br />

breast cancer and others<br />

[244,263,309]<br />

ITGBL1 + None None -<br />

MAP3K5<br />

(ASK1)<br />

5.1 Limited Gastric cancer and histiocytoma [94,189,504]<br />

NFATC2<br />

(NFAT1)<br />

+ Limited Breast cancer [64,95,311,348]<br />

NFKBIZ 5.1 None Liposarcoma [170]<br />

NR0B1 + None Lung adenocarcinoma and Ew- [?,236]<br />

(DAX1)<br />

ing’s sarcoma<br />

NR1D1 2.9 None Breast cancer [246]<br />

PARP3, 4.1, 2.9 Homology*** The PARP gene family is in- [312,463]<br />

PARP12<br />

volved in DNA repair and several<br />

other processes related to tumourigenesis<br />

PERP 3.8 None Lung and skin cancer [92,317]<br />

PPEF1 4.4 Limited None [296]<br />

SNAP25 3.3 None Lung cancer [175]<br />

SYT1 -2.5 None None -<br />

TNFRSF14 4.0 None Follicular lymphoma [93]<br />

TNFSF4 4.0 None Generally implicated in immune [424]<br />

(OX40L)<br />

response to tumours<br />

* Aliases are listed in parentheses.** Gene expression log2FC between the GNS and<br />

NS lines compared by Tag-seq. Some genes were detected exclusively in GNS or NS<br />

lines (indicated in column two by + or -, respectively).*** The homolog PARP1 has<br />

been implicated in glioma.<br />

ing list <strong>of</strong> genes an alternative tag mapping-based is<strong>of</strong>orm was observed, three<br />

<strong>of</strong> which are reported as adapted images from the UCSC genome browser in<br />

figures 6.15 and 6.16:<br />

145


6.7 Is<strong>of</strong>orm Differential Expression Results<br />

· AKT2: two reads map on the 3'UTR <strong>of</strong> the longest is<strong>of</strong>orm whilst one<br />

read maps on the 3'UTR <strong>of</strong> a shorter subset <strong>of</strong> is<strong>of</strong>orms;<br />

· AKT3: two reads map on the shortest is<strong>of</strong>orm and one on the 3'UTR<br />

and intron <strong>of</strong> the middle and longest is<strong>of</strong>orms, respectively;<br />

· BMP7: one read maps on the 3'UTR <strong>of</strong> the longest is<strong>of</strong>orm and one read<br />

on the 3'UTR <strong>of</strong> a middle-length set is<strong>of</strong>orms;<br />

· BRCA1: one read maps on the 3'UTR <strong>of</strong> the longest is<strong>of</strong>orm, whilst one<br />

read maps on an alternatively spliced exon, contained in a small subset<br />

<strong>of</strong> is<strong>of</strong>orms and which is a single is<strong>of</strong>orm itself;<br />

· CANX: all six reads map onto the 3'UTR <strong>of</strong> the longest is<strong>of</strong>orm, one on<br />

the 3'UTR <strong>of</strong> a shorter is<strong>of</strong>orm;<br />

· CTSB: three reads identify three different is<strong>of</strong>orms on their 3'UTRs;<br />

· ERBB2: two reads map on the 3'UTRs <strong>of</strong> two different is<strong>of</strong>orms (but<br />

one is not recognised by Ensembl Gene Predictions);<br />

· FGFR1: two reads map on the 3'UTRs <strong>of</strong> two different is<strong>of</strong>orms;<br />

· GRIA2: three reads identify three sets <strong>of</strong> lengths <strong>of</strong> is<strong>of</strong>orms on their<br />

3'UTRs with one identifying two <strong>of</strong> them;<br />

· HLA-A: two reads identify two different is<strong>of</strong>orms on their 3'UTR;<br />

· NBR1: one read maps on the 3'UTR <strong>of</strong> the longest is<strong>of</strong>orm and one read<br />

on a consistently present exon, which identifies also a shorter is<strong>of</strong>orm;<br />

· NFYC: two reads identify 3'UTRS <strong>of</strong> two different is<strong>of</strong>orms with one<br />

mapping between the 3'UTRs and last intron;<br />

· PTEN: two reads map on the 3'UTR <strong>of</strong> the longest is<strong>of</strong>orm and on the<br />

3'UTR <strong>of</strong> a much shorter is<strong>of</strong>orm;<br />

· RFX5: one read maps on the 3'UTR <strong>of</strong> two longer is<strong>of</strong>orms and one<br />

identifies a smaller subset <strong>of</strong> shorter ones on the 3'UTR;<br />

· NTRK2: two reads mapping on alternative 3'UTRs;<br />

· FGFR10P: one read maps on the 3'UTR and one on the fifth exon, which<br />

is displayed as a 3'UTR box on the Ensembl Prediction track;<br />

146


6.7 Is<strong>of</strong>orm Differential Expression Results<br />

· METTL13: two reads mapping on alternative 3'UTRs;<br />

· TSC22D2: two reads mapping on alternative 3'UTRs;<br />

· RSRC2: two reads mapping on alternative 3'UTRs;<br />

· TPM1: one read maps on the longest 3'UTR and the other on an internal<br />

exon that is alternatively spliced in one is<strong>of</strong>orm.<br />

We used the GenemiR package described in chapter 8 to find out how many<br />

genes, out <strong>of</strong> the 2,682 and 2,040 genes with differentially expressed is<strong>of</strong>orms,<br />

were predicted to harbour the same microRNA targeting sites in their 3'UTRs.<br />

The functionalities at the core <strong>of</strong> the GenemiR s<strong>of</strong>tware package are to output<br />

a list <strong>of</strong> microRNAs when a list <strong>of</strong> genes is inputted and, vice versa, to output<br />

a list <strong>of</strong> genes when a list <strong>of</strong> microRNAs is inputted. The database used by<br />

these core functionalities consists <strong>of</strong> all the microRNA to mRNA predictions<br />

made by a maximum <strong>of</strong> eight leading algorithms and it varies in size depend-<br />

ing on which algorithms the user has chosen to select as part <strong>of</strong> a specific<br />

search. The search performed in this instance made use <strong>of</strong> the union <strong>of</strong> the<br />

microRNA predictions from five <strong>of</strong> the most widely accepted target prediction<br />

algorithms: PicTar [247], PITA [224], Targetscan [272], miRanda [213] and<br />

DIANA-microT [315,316]. The choice <strong>of</strong> using the union set <strong>of</strong> the results<br />

from the five prediction algorithms as opposed to the intersection set is justi-<br />

fied by the fact that the intersection set for each <strong>of</strong> the two lists <strong>of</strong> over 2,000<br />

genes is null. In fact, as explained in chapter 8, the algorithms available to<br />

predict microRNA to mRNA interactions generate such different outputs that<br />

it is extremely rare to have all <strong>of</strong> them agree on the predictions (granted, <strong>of</strong><br />

course, that the list <strong>of</strong> genes is not so large that finding an intersection set<br />

becomes statistically possible, or that the number <strong>of</strong> prediction algorithms se-<br />

lected are less than 3). When inputted into GenemiR, the 2,682 genes we found<br />

with the parametric method, yielded a list <strong>of</strong> 5,016 microRNAs. Similarly, the<br />

2,040 genes we found with the logarithmic (non-parametric) method, yielded<br />

a list <strong>of</strong> 4,463 microRNAs. When the two lists <strong>of</strong> microRNAs were intersected<br />

(2,358 microRNAs) and inputed again into the GenemiR package to find the<br />

genes targeted by those microRNAs, we found 765 genes to be regulated by<br />

the intersected list <strong>of</strong> microRNAs (Fig 6.17). Table 6.7 shows the microRNA<br />

predictions resulting from the GenemiR query, stratified by prediction algo-<br />

rithm and origin <strong>of</strong> the gene list - parametric vs. non-parametric method - as<br />

well as the results that are common to both methods and unique to each.<br />

147


6.7 Is<strong>of</strong>orm Differential Expression Results<br />

148<br />

Figure 6.15: BMP7 encodes a member <strong>of</strong> the TGFβ superfamily and is represented in our Tag-seq data by two tags that identify transcripts <strong>of</strong><br />

different lengths. The expression levels across our GNS cell lines (top) are in blue (plus strand) and red (minus strand). BMP7, like other members<br />

<strong>of</strong> the bone morphogenetic protein family, plays a key role in the transformation <strong>of</strong> mesenchymal cells into bone and cartilage. However, BMP7 has<br />

also been recently implicated in the suppression <strong>of</strong> tumourigenicity <strong>of</strong> stem-like GBM cells when released from endogenous neural precursor cells [97].<br />

Image adapted from the UCSC genome browser.


6.7 Is<strong>of</strong>orm Differential Expression Results<br />

149<br />

Figure 6.16: The TPM1 gene, encoding for a member <strong>of</strong> the tropomyosin family <strong>of</strong> actin-binding proteins, is represented by two tags, the rightmost<br />

<strong>of</strong> which shows a very high expression in the antisense strand and identifies the four longest 3'UTRs. The expression levels across our GNS cell lines<br />

(top) are in blue (plus strand) and red (minus strand). Image adapted from the UCSC genome browser.


6.7 Is<strong>of</strong>orm Differential Expression Results<br />

Figure 6.17: This figure summarises the process <strong>of</strong> finding genes with differentially<br />

expressed is<strong>of</strong>orms (common to both the parametric and logarithmic method) that<br />

are predicted to harbour the same microRNA targeting sites. One way to assess this<br />

is to find which genes are predicted to be regulated by the same microRNAs. In blue<br />

we find the numbers <strong>of</strong> microRNAs that are predicted to regulate the 2,682 genes<br />

(green set) and the 2,040 genes (orange set). In black we find the numbers that<br />

refer to genes. The 2,358 microRNAs common to both sets are predicted to regulate<br />

765 genes that are in common to the parametric and the logarithmic methods. The<br />

prediction database used comes from the union <strong>of</strong> five prediction algorithms: PicTar<br />

[247], PITA [224], Targetscan [272], miRanda [213] and DIANA-microT [315,316].<br />

In order to find in more detail which <strong>of</strong> the genes with differentially expressed<br />

is<strong>of</strong>orms that were also predicted to harbour the same microRNA targeting<br />

sites, actually had one or more microRNA seed sequences in their 3'UTRs, we<br />

used the Ensembl gene coordinates and the union <strong>of</strong> all microRNA seed coor-<br />

dinates given by the same five prediction algorithms used earlier (Targetscan,<br />

miRanda, PITA and DIANA-microT) to validate which genes harboured which<br />

microRNA seed sequences. We found that, <strong>of</strong> the 765 genes predicted to target<br />

the same microRNA targeting sites, 226 <strong>of</strong> the 2,682 genes with differentially<br />

expressed is<strong>of</strong>orms (identified with the parametric method) and 340 <strong>of</strong> the<br />

2,040 genes with differentially expressed is<strong>of</strong>orms (identified with the logarith-<br />

mic method) hosted a microRNA seed sequence between at least two tags (Fig<br />

6.18). These amounts clearly show that microRNA is a widely adopted mech-<br />

anism <strong>of</strong> is<strong>of</strong>orm expression modulation in GNS cell lines. Since microRNA<br />

array data was available for four GNS cell lines (G7, G26, G144, G166) and<br />

four NS cell lines (CB660, CB130, CB152, CB171), we used it to cross verify<br />

150


6.7 Is<strong>of</strong>orm Differential Expression Results<br />

Table 6.7: Summary <strong>of</strong> predicted microRNAs targeting differentially expressed<br />

is<strong>of</strong>orms <strong>of</strong> the genes identified with the parametric and logarithmic method. The<br />

predictions from each <strong>of</strong> the five algorithms (PicTar [247], PITA [224], Targetscan<br />

[272], miRanda [213] and DIANA-microT [315,316]) are shown in distinct rows.<br />

Prediction<br />

algorithms<br />

Nonparametric<br />

microRNAs<br />

Parametric<br />

microRNAs<br />

Common<br />

microRNAs<br />

Unique to<br />

nonparametric<br />

Unique to<br />

parametric<br />

PicTar4 164 164 164 - -<br />

PITA 674 673 672 miR-658 miR-937<br />

miR-886-3p<br />

TargetscanS 148 148 147 miR-615-3p miR-450a<br />

miRanda 677 677 677 - -<br />

DIANA 510 508 503 miR-151-1p miR-146b-3p<br />

micro-T miR-324-5p miR-199b-5p<br />

miR-369-5p miR-566<br />

miR-423-3p miR-744<br />

miR-602 miR-877<br />

miR-658<br />

miR-941<br />

Figure 6.18: Schematisation <strong>of</strong> the localisation <strong>of</strong> the microRNA seeds given by<br />

the five prediction algorithms within the 3'UTRs <strong>of</strong> pairs <strong>of</strong> differentially expressed<br />

is<strong>of</strong>orms identified by two tags that map onto each one. Of the 765 genes with<br />

differentially expressed is<strong>of</strong>orms identified by both the parametric and logarithmic<br />

methods, 226/2,682 genes and 340/2,040 genes hosted at least one microRNA seed<br />

sequence between at least two tags.<br />

that the microRNA predictions found for the 765 genes with differentially ex-<br />

pressed is<strong>of</strong>orms were reflected in experimental data (Appendix F). Of the 11<br />

microRNAs that were predicted to regulate the 765 genes with differentially<br />

expressed is<strong>of</strong>orms that were also available in the microRNA array dataset, we<br />

found seven to be implicated in regulatory pathways in GBM (Table 6.8):<br />

151


6.7 Is<strong>of</strong>orm Differential Expression Results<br />

Table 6.8: MicroRNA array results for GNS cell lines with respect to NS cell lines.<br />

Based on a literature survey, these microRNAs would be interesting candidates to<br />

validate experimentally in a TLDA assay.<br />

microRNA log 2(F C) FDR<br />

miR-128 0.9 0.000206443<br />

miR-137 1 0.000121777<br />

miR-34a -1.5 0.000004.43<br />

miR-26a 1.4 0.0000143<br />

miR-10b 2.9 1.03E-08<br />

miR-451 1 3.15E-05<br />

miR-129-3p 1 9.63E-05<br />

· The levels <strong>of</strong> miR-128 have been found to be consistently lower in<br />

glioblastoma compared with normal brain tissue [259,364]. Opposite<br />

findings were observed in our microRNA array, in which miR-128 is up-<br />

regulated in GNS cells with respect to NS cells (Table 6.8). miR-128<br />

directly targets the transcription factor E2F3a, which activates genes<br />

necessary for the progression <strong>of</strong> cell-cycle and can thus inhibit prolifera-<br />

tion <strong>of</strong> brain cells by negatively regulating E2F3a. This microRNA also<br />

directly targets BMI1, a gene that is thought to act as an oncogene in<br />

glioblastoma by regulating tumour suppressors like P53 and CDKN2A.<br />

BMI1 also promotes stem cell renewal by acting as part <strong>of</strong> a Polycomb Si-<br />

lencing Complex to silence the expression <strong>of</strong> genes - including CDKN2A<br />

and CDKN1A tumour suppressors - involved in differentiation and senes-<br />

cence. Low levels <strong>of</strong> miR-128 in glioblastoma may contribute to glioma<br />

growth by allowing the increased expression <strong>of</strong> BMI1 to promote an un-<br />

differentiated self-renewing state. High levels <strong>of</strong> miR-128 in GNS cells<br />

with respect to NS cells may identify a stem cell pool specific regulation<br />

that has yet to be studied in detail. miR-128 is known to be highly<br />

expressed in neurons but its role in the brain is still unknown and is<br />

surmised to be the promotion <strong>of</strong> neuronal differentiation through pre-<br />

vention <strong>of</strong> stem cell self-renewal. Our is<strong>of</strong>orm data analysis with the<br />

non-parametric method showed that miR-128 is predicted to target the<br />

nerve growth factor receptor associated protein 1 NGFRAP1, a p75NTR-<br />

associated cell-death executor mediated by the common neutrophin re-<br />

ceptor p75NTR [350] that also plays a role in NGF-induced apopto-<br />

sis in oligodendrocytes [351]. Analsysis with the logarithmic method<br />

showed that miR-128 is predicted to target the Rab GTPase guanine nu-<br />

152


6.7 Is<strong>of</strong>orm Differential Expression Results<br />

cleotide exchange factor GAPVD1, a gene fundamental for the activation<br />

<strong>of</strong> RAB5A during the engulfment <strong>of</strong> apoptotic cells [238] (Fig 6.19).<br />

· miR-137 is one <strong>of</strong> the most down-regulated microRNAs in glioblas-<br />

toma compared with normal brain tissue. In our microRNA microarray<br />

dataset, miR-137 is up-regulated in GNS cell lines with respect to NS cell<br />

lines. Since miR-137 directly targets CDK6, if over-expressed in glioblas-<br />

toma it would induce cell-cycle arrest. The level <strong>of</strong> miR-137 increases<br />

upon differentiation <strong>of</strong> glioma neurosphere cultures and if over-expressed<br />

in these cells it leads to the expression <strong>of</strong> markers consistent with neu-<br />

ronal differentiation. These data suggest that the lower expression <strong>of</strong><br />

this microRNA in glioblastoma, and the higher expression in GNS cells<br />

with respect to NS cells, reflects the lack <strong>of</strong> tumour cell differentiation<br />

in the former and the presence <strong>of</strong> regulatory mechanisms downstream <strong>of</strong><br />

miR-137 in the latter, since cancer stem cells are the least differentiated<br />

within the lineage hierarchy according to the cancer stem cell hypothe-<br />

sis [364]. In our microRNA prediction analysis, miR-137 was predicted<br />

to target the serine/threonine-protein kinase 40 STK40 gene, a nega-<br />

tive regulator <strong>of</strong> NFKB and p53-mediated gene transcription [200] and<br />

the transcription factor TCF4, which associated with β catenin mediates<br />

Wnt signaling by trans-activating downstream target genes [11].<br />

· miR-34a expression is down-regulated in glioblastoma and in p53-null<br />

mutant gliomas since non p53-null mutants, expressed in many tumours<br />

and that can possess gain-<strong>of</strong>-function activities, do not regulate tran-<br />

scription <strong>of</strong> miR-34a. This suggests miR-34a as a transcriptional target<br />

<strong>of</strong> P53 [278]. In our microRNA microarray dataset miR-34a is found to<br />

be down-regulated as well. Furthermore, miR-34a potently inhibits the<br />

protein expression <strong>of</strong> MET, a hepatocyte growth factor receptor encod-<br />

ing a tyrosine-kinase, as well as MET 3'UTR reporter activity in glioma,<br />

medulloblastoma cells and astrocytes. miR-34a also inhibits Notch-1<br />

and Notch-2 protein expression and their 3'UTR reporter activities, as<br />

well as CDK6 protein expression in glioma cells. Transient transfection<br />

<strong>of</strong> miR-34a into brain tumour cell lines inhibited cell proliferation, cell<br />

cycle progression, cell survival, and cell invasion but did not affect hu-<br />

man astrocyte cell survival and cell cycle. miR-34a transfection, also,<br />

did not affect the protein levels <strong>of</strong> PDGFRA in any tested cell line, al-<br />

though miR-34a has predicted seed matches in the 3'UTR <strong>of</strong> PDGFRA.<br />

153


6.7 Is<strong>of</strong>orm Differential Expression Results<br />

154<br />

Figure 6.19: Is<strong>of</strong>orm detection by multi tag mapping <strong>of</strong> gene GAPVD1, a GTPase guanine nucleotide exchange factor essential during engulfment<br />

<strong>of</strong> apoptotic cells [238] and involved in the degradation <strong>of</strong> EGFR [473]. The expression levels across our GNS cell lines (top) are in blue (plus strand)<br />

and red (minus strand). Image adapted from the UCSC genome browser.


6.7 Is<strong>of</strong>orm Differential Expression Results<br />

miR-34a transfected cells generated xenografts that were statistically<br />

significantly smaller than control miR transfected xenografts, demon-<br />

strating that miR-34a expression inhibits in vivo glioblastoma xenograft<br />

growth [278]. In our microRNA prediction analysis miR-34a is predicted<br />

to target C1orf9, an open reading frame 9 <strong>of</strong> chromosome 1 since no p53<br />

targeting is predicted due to the deletion <strong>of</strong> the p53 genomic location<br />

within the GNS cell lines.<br />

· miR-26a is up-regulated in glioblastoma and targets PTEN through the<br />

direct binding in its 3'UTR <strong>of</strong> the B2 and B3 sites, mediating transla-<br />

tional repression and reduced steady-state levels <strong>of</strong> the protein. West-<br />

ern blotting demonstrated that miR-26a over-expression achieved a 50%<br />

knockdown <strong>of</strong> PTEN protein in two glioblastoma cell lines, accompa-<br />

nied by an enhanced Akt signaling pathway. In our microRNA mi-<br />

croarray dataset we observe up-regulation <strong>of</strong> miR-26a as well. In addi-<br />

tion to enhancing tumourigenesis, miR-26a effectively represses endoge-<br />

nous PTEN protein in a relevant PDGF-driven glioma model system.<br />

The miR-26a-mediated knockdown <strong>of</strong> EZH2, a histone methylatrans-<br />

ferase, and SMAD1, a transcription factor, was also observed in glioblas-<br />

toma [259,364]. In our analysis miR-26a was predicted to target the<br />

SMAD1 gene, <strong>of</strong> which two is<strong>of</strong>orms were detected through tag mapping<br />

(Fig 6.20).<br />

· miR-10b is up-regulated in glioblastomas but its function has not been<br />

described yet. Increased levels <strong>of</strong> miR-10b in breast cancer correlated<br />

with the disease’s progression [259,364]. In our microRNA microarray<br />

dataset miR-10b is also up-regulated.<br />

· miR-451 inhibited the growth <strong>of</strong> transfected glioblastoma cells, as de-<br />

tected by neurosphere formation assays [259,364]. We found miR-451 to<br />

be up-regulated in our microRNA microarray dataset, in line with its<br />

role as a cell cycle breaker and growth inhibitor.<br />

· miR-129-3p is found to be down-regulated in glioblastomas but its func-<br />

tion remains unknown to date [259,364]. Interestingly, we observed miR-<br />

129-3p to be up-regulated in our microRNA microarray dataset, possibly<br />

indicating a different regulation in action at the stem cell level.<br />

155


6.7 Is<strong>of</strong>orm Differential Expression Results<br />

156<br />

Figure 6.20: Is<strong>of</strong>orm detection by multi tag mapping <strong>of</strong> gene SMAD1. The expression levels across our GNS cell lines (top) are in blue (plus<br />

strand) and red (minus strand). Image adapted from the UCSC genome browser.


6.8 Long ncRNA Differential Expression Results<br />

6.8 Long ncRNA Differential Expression<br />

In contrast to microarray expression pr<strong>of</strong>iling, Tag-seq is not limited by pre-<br />

selected probes targeting the known transcriptome and we took advantage <strong>of</strong><br />

this to discover differentially expressed ncRNAs. To this end, we called differ-<br />

ential expression for a combination <strong>of</strong> tags that mapped to the genome, the<br />

transcriptome or virtual tags and filtered the results using coding gene anno-<br />

tations. At an FDR <strong>of</strong> 10%, this analysis revealed 25 differentially expressed<br />

putative non-coding RNAs, 18 <strong>of</strong> which were up-regulated and the remaining 7<br />

down-regulated (Appendix C). Five <strong>of</strong> these are putative long antisense RNAs<br />

that are known to be transcribed from the opposite strand <strong>of</strong> protein-coding<br />

genes CDKN2B, CD27, PAX8, MCF2L2 and TXNRD1. Two, instead, are<br />

known long non-coding RNAs: HOTAIRM1 [547] and NEAT1 [314] (Table<br />

6.9). CDKN2BAS, an antisense transcript to tumour suppressor CDKN2B, is<br />

<strong>of</strong> particular interest because <strong>of</strong> its role in Polycomb-mediated repression <strong>of</strong><br />

CDKN2B and CDKN2A [537]. We detected CDKN2BAS exclusively in G144,<br />

G144ED and G166, consistent with the locus being deleted in G179 according<br />

to aCGH data (see 6.3). The functions <strong>of</strong> the remaining differentially expressed<br />

Table 6.9: Multiple putative long non-coding RNAs differentially expressed between<br />

GNS and NS cells at 10% FDR.<br />

Category Up-regulated Down-regulated<br />

Known antisense transcripts 2 (over CDKN2B, CD27) 1 (over PAX8)<br />

Other known ncRNAs 2 (HOTAIRM1, NEAT1) 0<br />

Intronic RNAs 2 (in CDKN2B, TXNRD1) 1 (in FAM38B)<br />

Intergenics RNA 9 7<br />

non-coding RNAs are unknown, but a unifying feature <strong>of</strong> 15 <strong>of</strong> them is that<br />

they are located in gene deserts (Appendix D). Several <strong>of</strong> these transcripts<br />

display an expression pattern similar to a protein-coding gene near the gene<br />

desert, suggesting that the transcripts may be functional RNAs regulating<br />

nearby genes [372] or indicate transcription from active enhancers [230]. The<br />

coding genes exhibiting correlated expression to these non-cancer RNAs are<br />

cancer-related genes CTSC (Fig 6.21) [113,551] and DKK1 [461] and develop-<br />

mental regulators IRX2, SIX3 and ZNF536 [412].<br />

In the case <strong>of</strong> the Cathepsin C (CTSC) gene we were able to detect two dif-<br />

ferent is<strong>of</strong>orms with Tag-seq, as well as a long non-coding RNA lying within<br />

a 150 kilobase distance from the two is<strong>of</strong>orms. The CTSC gene encodes a<br />

42 Megabase sized genomic segments devoid <strong>of</strong> protein-coding genes in vertebrates.<br />

157


6.8 Long ncRNA Differential Expression Results<br />

lysosomal cysteine protease that is part <strong>of</strong> the peptidase C1 family <strong>of</strong> proteins<br />

and is responsible for activating serine proteases in immune and inflammatory<br />

cells to function in processes <strong>of</strong> bone remodelling, epidermal homeostasis, and<br />

antigen presentation [500]. During cancer progression cathepsins are secreted<br />

into the extracellular matrix where they promote tumour invasion by cleav-<br />

ing components <strong>of</strong> the matrix and the basement membrane, thereby creating<br />

a passageway for the migration <strong>of</strong> cancer cells. The disruption <strong>of</strong> adherens<br />

junctions via cleavage <strong>of</strong> E-cadherin is another example. Cathepsins can also<br />

initiate proteolytic cascades in which they activate other proteases such as<br />

matrix metalloproteinases, which in turn promote invasion [166,167]. Mem-<br />

bers B, L and S <strong>of</strong> the cathepsin family have been identified as regulators<br />

<strong>of</strong> E-cadherin function through cleavage <strong>of</strong> its N-terminus. Knockout mice<br />

mutants for cathepsins B, L or S show obvious defects such as the decrease<br />

<strong>of</strong> tumour cell proliferation, tumour invasion and tumour vascularity, while<br />

CTSC - / - knockout mice have more subtle defects that consist in failing to<br />

activate granzymes A and B in cytotoxic lymphocytes [167].<br />

The hypothetical regulation performed by the long non-coding RNA BC038205<br />

on CTSC is<strong>of</strong>orms could be one <strong>of</strong> maintaining the expression levels <strong>of</strong> this<br />

gene high in GNS cell lines through a form <strong>of</strong> direct regulation which is re-<br />

lieved when the non-coding RNA is absent or expressed at lower levels, such as<br />

in NS cells. In figure 6.21 three panels are shown in which the location (panel<br />

a) <strong>of</strong> the CTSC is<strong>of</strong>orms and that <strong>of</strong> the non-coding RNA is shown along the<br />

human reference genome; the expression levels as detected by Tag-seq (panel<br />

b) are displayed in a histogram to visually highlight the relationship between<br />

them; a correlation plot (panel c) that shows the presence <strong>of</strong> a correlation in<br />

both the expression trends between the first CTSC is<strong>of</strong>orm and the non-coding<br />

RNA BC038205, and the second CTSC is<strong>of</strong>orm and the same non-coding RNA<br />

gene. The correlation was calculated with the cor.test function in the R stats<br />

package that uses the Pearson’s product moment correlation coefficient to test<br />

for association between paired samples.<br />

HOTAIRM1 is known to be strongly up-regulated in human NB4 promyelo-<br />

cytic cell lines and normal hematopoietic cells upon induction <strong>of</strong> granulocytic<br />

differentiation and is found to be up-regulated in our GNS lines as well as in<br />

the gliomas from the Parsons et al [383] Tag-seq data with respect to our fetal<br />

human NS cell lines and the Parsons et al primary brain samples. Interestingly,<br />

its knock-down in NB4 cells causes down-regulation <strong>of</strong> HOXA1 and HOXA4<br />

genes and these genes are up-regulated in our Tag-seq dataset (Fig 6.22).<br />

158


6.8 Long ncRNA Differential Expression Results<br />

Figure 6.21: Correlated expression <strong>of</strong> CTSC and a nearby ncRNA. (a) CTSC<br />

(cathepsin C) is located in a gene desert harboring an uncharacterized ncRNA transcribed<br />

in the opposite direction (cDNA BC038205). Image adapted from the Ensembl<br />

Genome Browser [147]. (b) Both CTSC and the ncRNA have strongly elevated<br />

expression in the GNS lines relative to the NS lines, with highest levels in G179.(c)<br />

Correlation plots for the BC038205 ncRNA and first is<strong>of</strong>orm <strong>of</strong> CTSC (grey) and<br />

second is<strong>of</strong>orm <strong>of</strong> CTSC (blue).<br />

159


6.8 Long ncRNA Differential Expression Results<br />

160<br />

Figure 6.22: Histogram displaying the normalised tag counts found in our Tag-seq dataset and the Parsons et al [383] Tag-seq dataset for the<br />

HOTAIRM1 long ncRNA and the surrounding HOX genes.


Chapter 7<br />

Dataset Correlation Analyses<br />

Contents<br />

7.1 Enrichment Analysis . . . . . . . . . . . . . . . . . . . . . . 161<br />

7.2 Glioblastoma Expression Signatures . . . . . . . . . . . . . 168<br />

7.3 Tumour Expression Correlation . . . . . . . . . . . . . . . . 170<br />

7.4 Survival Analysis . . . . . . . . . . . . . . . . . . . . . . . . 182<br />

7.5 Glioblastoma Pathway Analysis . . . . . . . . . . . . . . . 187<br />

7.1 Enrichment Analysis<br />

The differential expression analysis revealed 485 genes to be up-regulated and<br />

254 genes to be down-regulated between GNS and NS cell lines at an FDR <strong>of</strong><br />

10% (Appendix A.1). We performed Gene Set Enrichment Analysis (GSEA)<br />

to investigate which pathways were most highly represented in our set <strong>of</strong> 739<br />

differentially expressed genes.<br />

In order to evaluate the enrichment <strong>of</strong> a group <strong>of</strong> genes that together define a<br />

pathway, the GSEA method looks for those genes to follow the same trends in<br />

the experimental dataset <strong>of</strong> interest. In fact, if a number <strong>of</strong> genes that belong<br />

to the same pathway change expression level even moderately, it could mean<br />

that in the evaluated experimental setting that pathway is being affected. By<br />

having established an a priori relationship between the genes involved in the<br />

same pathway, the GSEA method detains more statistical power to detect<br />

smaller changes that affect the whole set as compared to a per gene statistic.<br />

In order to achieve its goal the GSEA method first ranks the genes in the<br />

dataset <strong>of</strong> interest according to a per gene statistic such as a p-value and<br />

then uses the complete ranked list to assess how the genes that belong to a<br />

161


7.1 Enrichment Analysis Results<br />

specific pathway distribute across the ranked list, whether they are randomly<br />

distributed throughout the ranked list or primarily found at the top or bottom.<br />

Three key elements define the GSEA method [474]:<br />

1. Calculating an enrichment score that measures the degree to which a set<br />

<strong>of</strong> genes belonging to a pathway is overrepresented at the top or bottom<br />

<strong>of</strong> the entire ranked list <strong>of</strong> differentially expressed genes, for example<br />

(and corresponds to a weighted Kolmogorov-Smirnov like statistic).<br />

2. Estimating the significance <strong>of</strong> the enrichment score by permuting the<br />

phenotype labels and recomputing the enrichment score <strong>of</strong> the genes in<br />

the pathway each time. This generates a null distribution that the p-<br />

value <strong>of</strong> the observed original enrichment score is calculated against.<br />

3. Adjusting the enrichment score to account for multiple hypothesis testing<br />

when entire databases <strong>of</strong> pathways are evaluated at once, like in our case<br />

with the KEGG and Gene Ontology (GO) databases. In this case the<br />

enrichment score is first normalised to account for the size <strong>of</strong> the path-<br />

way, and then the proportion <strong>of</strong> false positives, or FDR, is calculated<br />

for each normalised enrichment score. The FDR associated to each nor-<br />

malised enrichment score corresponds to the estimated probability that<br />

a pathway with a given normalised enrichment score represents a false<br />

positive finding.<br />

The enrichment analysis using GO [106] and the KEGG pathway database [2]<br />

confirmed the set <strong>of</strong> 739 differentially expressed genes to be enriched for path-<br />

ways related to brain development, glioma and cancer (Table 7.2 and 7.1). We<br />

also observed enrichment <strong>of</strong> regulatory and inflammatory genes, such as signal<br />

transduction components, cytokines, growth factors and DNA-binding factors.<br />

Several genes related to antigen presentation on MHC class I and II molecules<br />

were up-regulated in GNS cells, consistent with the documented expression <strong>of</strong><br />

their corresponding proteins in glioma tumours and cell lines [120,174]. In line<br />

with these findings, affected pathways from the KEGG database included Anti-<br />

gen Processing and Presentation, Diabetes Mellitus Type I, Cytokine-cytokine<br />

receptor interaction, Neuroactive ligand-receptor interaction, MAPK signaling<br />

and, expectedly, <strong>Glioma</strong>, a collection <strong>of</strong> genes involved in glioma formation<br />

(Table 7.2). The first two plots from the GSEA run that identified these path-<br />

ways as being significantly altered in our dataset, are shown in figure 7.1. In<br />

the top panels <strong>of</strong> the figure the green distribution represents the trend <strong>of</strong> the<br />

162


7.1 Enrichment Analysis Results<br />

Table 7.1: Selected Gene Ontology terms and InterPro domains enriched among differentially expressed genes.<br />

Differentially Expressed, 739 genes Up-regulated, 485 genes Down-regulated, 254 genes<br />

Genes p-value Genes p-value Genes p-value<br />

A. Biological Process GO terms<br />

Nervous system development 106 2.00E-10 62 0.005 44 2.00E-05<br />

Cell differentiation 128 7.00E-07 74 n.s. 54 2.00E-04<br />

Cell proliferation 86 3.00E-04 59 0.014 27 n.s.<br />

Cell adhesion 74 1.00E-07 56 2.00E-07 18 n.s.<br />

Cell migration 44 3.00E-04 30 0.026 14 n.s.<br />

Immune response 70 2.00E-12 61 3.00E-16 9 n.s.<br />

Antigen processing and presentation 17 4.00E-07 17 5.00E-10 0 n.s.<br />

Cellular ion homeostasis 36 0.014 33 2.00E-05 3 n.s.<br />

B. Molecular Function GO terms<br />

Signal transducer activity 111 3.00E-07 66 0.058 45 0.002<br />

Receptor activity 83 8.00E-07 48 n.s. 35 0.002<br />

Cytokine activity 27 2.00E-08 25 7.00E-11 2 n.s.<br />

Growth factor activity 20 0.011 17 0.002 3 n.s.<br />

MHC class II receptor activity 5 0.008 5 9.00E-04 0 n.s.<br />

Sequence-specific DNA binding 52 3.00E-04 34 0.053 18 n.s.<br />

C. Interpro domains<br />

Immunoglobulin-like 45 3.00E-08 32 6.00E-06 13 n.s.<br />

MHC classes I/II-like antigen recognition protein 14 1.00E-07 14 3.00E-10 0 n.s.<br />

Homeobox 28 8.00E-06 18 0.012 10 n.s.<br />

P-values indicating the statistical significance <strong>of</strong> enrichment <strong>of</strong> these terms were computed with Fisher’s exact test and corrected for multiple testing<br />

using the Bonferroni method; n.s., not significant (p-value>0.1).<br />

163


7.1 Enrichment Analysis Results<br />

Table 7.2: Representative KEGG pathways from signaling pathway impact analysis<br />

<strong>of</strong> gene expression differences between GNS and NS lines.<br />

Pathway Genes p-value Predicted status in<br />

GNS cell lines<br />

Cytokine-cytokine receptor interaction 29 4.00E-12 Activated<br />

Chemokine signaling pathway 15 5.00E-06 Activated<br />

Neuroactive ligand-receptor interaction 21 2.00E-04 Inhibited<br />

Antigen processing and presentation 11 7.00E-04 Activated<br />

MAPK signaling pathway 24 0.011 Activated<br />

<strong>Glioma</strong> 10 0.013 Activated<br />

ECM-receptor interaction 10 0.041 Inhibited<br />

Calcium signaling pathway 15 0.041 Activated<br />

P-values and status predictions were obtained by signaling pathway impact analysis<br />

[26], taking fold-change estimates and pathway topology into account. P-values were<br />

FDR-corrected for multiple testing.<br />

enrichment score - a number that reflects the degree to which a gene set is<br />

overrepresented at the top or bottom <strong>of</strong> a ranked list <strong>of</strong> genes - as the analysis<br />

walks down the ranked list. Both enrichment score distributions who positive<br />

values because they indicate gene set enrichment at the top <strong>of</strong> the ranked list.<br />

The score at the peak <strong>of</strong> the plot (farthest from 0.0) is the enrichment score for<br />

the gene set. The middle portions <strong>of</strong> the two plots show where the members <strong>of</strong><br />

the gene set appear in the ranked list <strong>of</strong> genes. The set <strong>of</strong> genes that appear in<br />

the ranked list prior to the peak enrichment score are the ones that contribute<br />

most to the enrichment score and are commonly referred to as "leading edge<br />

subset". The bottom portions <strong>of</strong> the two plots show the value <strong>of</strong> the ranking<br />

metric, which is by default the signal-to-noise ratio and has been kept such in<br />

our GSEA runs, as you move down the list <strong>of</strong> ranked genes. The ranking met-<br />

ric measures a gene’s correlation with a phenotype: a positive value indicates<br />

correlation with the GNS phenotype and a negative value indicates correla-<br />

tion with the NS phenotype. Additional information from our GSEA runs is<br />

provided in the two plots described in figure 7.2. Surprisingly, via GSEA we<br />

detected an up-regulation in the GNS lines <strong>of</strong> several Major Histocompatabil-<br />

ity Complex (MHC) class II genes, as well as related genes involved in antigen<br />

presentation on MHC class I complexes (Fig 7.3). Several works have shown<br />

that MHC class I and II molecules are involved in aspects <strong>of</strong> human cancer<br />

pathology such as invasion and migration [327,421,546]. These two classes <strong>of</strong><br />

molecules are known to be a fundamental component <strong>of</strong> the adaptive immune<br />

response and their misregulation potentially leads to faulty antigen recognition<br />

and processing.<br />

164


7.1 Enrichment Analysis Results<br />

Figure 7.1: Enrichment plots for the top two pathways revealed through GSEA <strong>of</strong><br />

the KEGG database <strong>of</strong> pathways [2].<br />

Figure 7.2: (a) The plot <strong>of</strong> nominal p-values, which estimates the statistical significance<br />

<strong>of</strong> the enrichment score for a single pathway, vs normalised enrichment (NES)<br />

score provides a quick way to grasp the number <strong>of</strong> enriched pathways that are significant.<br />

The FRD q-value represents the estimated probability that the normalised<br />

enrichment score represents a false positive finding. (b) The line graph <strong>of</strong> the enrichment<br />

scores across pathways provides a quick visual way to grasp the number<br />

<strong>of</strong> enriched gene sets at every enrichment score value. The first two peaks are negative<br />

global enrichment scores that represent the enrichment <strong>of</strong> their corresponding<br />

number <strong>of</strong> pathways in the NS phenotype. The last two peaks are positive global<br />

enrichment scores that represent the enrichment <strong>of</strong> their corresponding number <strong>of</strong><br />

pathways in the GNS phenotype.<br />

The endogenous pathway is used by any nucleated cell in the body to<br />

present endogenous, or cytosolic, fragments <strong>of</strong> proteins to cytotoxic CD8 + T<br />

cells. The receptors that are responsible for such presentation are the Major<br />

Histocompatibility Complex (MHC) class I molecules. By exposing cytosolic<br />

165


7.1 Enrichment Analysis Results<br />

proteins outside the cellular membrane, MHC class I receptors can cause the<br />

activation <strong>of</strong> cytotoxic T cells against exogenous peptides derived from an in-<br />

fection. In fact, healthy cells are ignored by the cytotoxic T cells thanks to the<br />

sensitivity <strong>of</strong> the recognition system between the loaded MHC class I receptor<br />

and the T cell receptor, while cells containing foreign proteins can be recog-<br />

nised and, therefore, killed. MHC class I molecules are heterodimers and they<br />

consist <strong>of</strong> two polypeptide chains, the α and the β2-microglobulin chain, <strong>of</strong><br />

which only the α chain is polymorphic. Loading <strong>of</strong> MHC class I receptors with<br />

peptides occurs inside the lumen <strong>of</strong> the endoplasmic reticulum (ER) [323].<br />

The exogenous pathway is used by a cohort <strong>of</strong> specialised cells termed pro-<br />

fessional Antigen Presenting <strong>Cells</strong>, or APCs. These cells are macrophages,<br />

dendritic cells and B cells and they present fragments <strong>of</strong> extracellular proteins<br />

to helper CD4 + T cells triggering a cell-mediated (Th1) or humoral (Th2) re-<br />

sponse for peptides deriving from extracellular pathogens. The receptors that<br />

are responsible for such presentation are the MHC class II molecules. MHC<br />

class II molecules, like the MHC class I molecules, are heterodimers but the<br />

MHC class II molecules consist <strong>of</strong> two homologous peptides, the α and β chain,<br />

both polymorphic and encoded by the HLA gene. Loading <strong>of</strong> both classes oc-<br />

curs inside the cell, but MHC class II molecules are loaded only when the<br />

vesicle generated at the ER fuses with a lysosome containing digested endo-<br />

cysed extracellular proteins. In the ER vesicle, the MHC class II molecule has<br />

its peptide-binding cleft blocked by CD74, a trimer called "invariant chain"<br />

which prevents the binding <strong>of</strong> cellular peptides. Once this vesicle fuses with<br />

the lysosome, the MHC class II molecules are unloaded with the invariant<br />

chain and reloaded with a peptide from the lysosome via an MHC class II-like<br />

structure called HLA-DM. The stable MHC class II thus formed is presented<br />

on the cell surface. the expression <strong>of</strong> MHC class II molecules is constitutive<br />

only on pr<strong>of</strong>essional APCs but can be induced by cytokines such as IFNG on<br />

other cell types, including cancer cells [492].<br />

All genes involved in the two pathways are listed below (7.3) [327], showing all<br />

alleles <strong>of</strong> the same Human Leukocyte Antigen (HLA) class. In a study on ovar-<br />

ian cancer, the MHC class II receptor α polymorphic chain, encoded by the<br />

HLA-DRA gene, was found to be the most over-expressed gene [421]. Although<br />

also the β chain, HLA-DRB, was over-expressed, the almost complete lack <strong>of</strong><br />

this polypeptide at the protein level via an unknown post-transcriptional or<br />

post-translational mechanism <strong>of</strong> regulation, precluded the formation <strong>of</strong> a ma-<br />

ture HLA-DR receptor and a similar pattern was observed when staining brain<br />

166


7.1 Enrichment Analysis Results<br />

Table 7.3: Summary <strong>of</strong> all MHC class I and II genes and the FC (F DR > 10%) <strong>of</strong><br />

those measured in the Tag-seq dataset.<br />

Cytosolic pathway Exogenous pathway<br />

Gene log 2(F C) Gene log 2(F C)<br />

HLA-A 3.04 Alpha chains<br />

HLA-B HLA-DMA Inf<br />

HLA-C HLA-DQA<br />

HLA-E 2.09 DPA1 5.73<br />

HLA-F DQA1 Inf<br />

HLA-G DRA 5.68<br />

HLA-K Beta chains<br />

HLA-L HLA-DMB<br />

B2M HLA-DOB<br />

PSMB5 HLA-DPB1 3.83<br />

PSMB6 HLA-DQA2 6.56<br />

PSMB7 HLA-DQB1 4.60<br />

PSMB8 HLA-DQB2<br />

PSMB9 HLA-DRB1<br />

PSMB10 HLA-DRB3<br />

TAP1 2.36 HLA-DRB4<br />

TAP2 HLA-DRB5 8.36<br />

CANX Invariant chains<br />

CALR CD74,CLIP 7.11<br />

TAPBPL 2.57 <strong>Transcriptional</strong> co-activator<br />

CIITA<br />

tumour tissue [419]. It seems that compensatory mechanisms such as cyto-<br />

plasm over-expression <strong>of</strong> Ii/CD74 and reduction <strong>of</strong> HLA-DRB are in action to<br />

decrease the tumour’s immunogenicity by decreasing the ability <strong>of</strong> MHC class<br />

II to present tumour-specific antigens to the host immune system. Interest-<br />

ingly, no such mechanism is observed in our study, in which HLA-DRA, HLA-<br />

DRB and CD74 are all over-expressed in the GNS cell lines with a fold-change<br />

much greater than two (Table 7.3). The same magnitude <strong>of</strong> up-regulation is<br />

observed for the MHC class II-like peptide HLA-DM that aids in the unloading<br />

<strong>of</strong> CD74 and loading <strong>of</strong> immature MHC class II receptors with the extracel-<br />

lular peptides present in the lysosome to form the mature molecule. Other<br />

two MHC class II αβ heterodimer receptors are over-expressed according to<br />

our dataset and these are HLA-DP and HLA-DQ. In fact, both homologue<br />

chains encoded by the HLA-DPA1 and HLA-DPB1 loci and HLA-DQA1 and<br />

HLA-DQB1 loci are over-expressed and have the potential to form a mature<br />

MHC class II receptor. Altogether these findings suggest that in the GNS cell<br />

lines the exogenous pathway is not affected by compensatory mechanisms <strong>of</strong><br />

167


7.2 Glioblastoma Expression Signatures Results<br />

regulation and the presentation <strong>of</strong> extracellular peptides can potentially be<br />

completed for recognition from T helper cells. As for MHC class I-mediated<br />

immunogenicity, our study identifies two important players as strongly up-<br />

regulated in GNS cell lines, HLA-A and TAP1. The protein encoded by TAP1<br />

is an ATP binding cassette transporter involved in the pumping <strong>of</strong> degraded<br />

cytosolic peptides across the ER into the vesicles where MHC class I receptors<br />

such as HLA-A assemble. TAP1 has been show to interact with TAPBP and<br />

HLA-A [386]. Similarly to the expression observed for MHC class II molecules,<br />

the over-expression <strong>of</strong> these MHC class I molecules seems to suggest that part<br />

<strong>of</strong> the endogenous pathway might also be activated in immunosurveillance<br />

response mechanisms in GNS cell lines. We find an overall transcriptional<br />

up-regulation <strong>of</strong> MHC class II molecules and a smaller subset <strong>of</strong> MHC class I<br />

molecules, suggestive <strong>of</strong> the absence <strong>of</strong> transcriptionally active compensatory<br />

mechanisms. However, it is impossible with the available data to determine<br />

what happens at the post-transcriptional and post-translational level because<br />

this up-regulation should be checked against protein expression data.<br />

7.2 Glioblastoma Expression Signatures<br />

Analysis <strong>of</strong> microarray gene expression data for hundreds <strong>of</strong> high-grade glioma<br />

samples and a smaller number <strong>of</strong> xenografts have shown that most tumours<br />

can be classified into a small number <strong>of</strong> subtypes correlated with survival and<br />

response to therapy [148,390,511]. The largest such study to date identified<br />

four glioblastoma subtypes, each characterised by a distinct gene expression<br />

signature encompassing 210 genes [511]. The subtypes were named "proneu-<br />

ral", "neural", "classical" and "mesenchymal" based on which genes were up-<br />

regulated in their respective expression signatures. To investigate whether<br />

these subtype signatures could be captured by our Tag-seq data, Tag-seq ex-<br />

pression data for three primary glioblastoma tumours, 11 xenografts and two<br />

normal brain samples was analysed, which had been produced with the same<br />

Tag-seq protocol used on our GNS and NS cell lines by Parsons et al [383].<br />

The correlations were highly significant (p < 0.01) for all tumour and xenograft<br />

samples and both normal brain samples (Fig 7.3), confirming that Tag-seq cap-<br />

tures the subtype expression signatures previously observed in large microarray<br />

datasets. Specifically, <strong>of</strong> the tumour and xenograft samples, three were classi-<br />

fied as proneural, seven as classical and three as mesenchymal. However, both<br />

normal brain samples and none <strong>of</strong> the glioblastoma samples, were classified as<br />

168


7.2 Glioblastoma Expression Signatures Results<br />

neural, consistent with this subtype being the least common and characterised<br />

by markedly better prognosis, expressing genes associated with normal brain<br />

and neurogenesis [390,511]. In comparing the GNS line expression pr<strong>of</strong>iles to<br />

the subtype signatures, we found that both G166 and G179 correlated strongly<br />

with the mesenchymal signature. Mesenchymal subtype markers with elevated<br />

expression in these two lines included MET, CD44, CD68 and CASP1 [511].<br />

G144 did not correlate significantly with any <strong>of</strong> the four signatures, but showed<br />

a slight positive correlation (R = 0.07) with the proneural one. Supporting<br />

such classification <strong>of</strong> G144 were several <strong>of</strong> the hallmarks <strong>of</strong> the proneural sub-<br />

type emphasised by Verhaak et al 2010, such as high expression <strong>of</strong> oligodendro-<br />

cytic development genes PDGFRA, NKX2-2 and OLIG2, as well as ERBB3,<br />

DCX and TCF4 genes, and low levels <strong>of</strong> tumour suppressor CDKN1A. All<br />

in all, the Tag-seq data seems to agree with the results from the microarray<br />

technology, which is very reassuring given these technologies are so different.<br />

When we verified in which <strong>of</strong> the four subtypes defined by the Verhaak et<br />

al study [511] our 29 genes fell - the genes distinguishing GNS from NS cell<br />

lines measured via qRT-PCR - we found that only three <strong>of</strong> them, namely<br />

CEBPB, TES and PLS3, were represented as part <strong>of</strong> the original signature<br />

genes. Interestingly, however, each one <strong>of</strong> the three genes fell within the mes-<br />

enchymal subtype. The mesenchymal phenotype has recently been associated<br />

with neoplastic transformation in the CNS as a state in which cells contrive an<br />

uncontrolled ability to invade and stimulate angiogenesis [390,497]. Since the<br />

defining characteristics <strong>of</strong> the aggressiveness <strong>of</strong> GBM are invasion <strong>of</strong> the local<br />

brain parenchyma and the ability to stimulate novel angiogenesis [154,219],<br />

our findings reflect this GBM identity. Although 26 <strong>of</strong> the 29 genes were not<br />

represented in the original 210 gene signature defined by Verhaak et al, the<br />

three genes that were present belonged to the mesenchymal subtype, conducive<br />

<strong>of</strong> the fact that they are core players in establishing the characteristic aggres-<br />

siveness <strong>of</strong> GBM (as confirmed by two separate platforms).<br />

CEBPB, in fact, which we find to be up-regulated in GNS lines with respect<br />

to NS lines in our qRT-PCR measurements (Fig 6.14), is an important player<br />

in the mesenchymal regulatory module together with STAT3 and is needed<br />

for mesenchymal transformation in human glioma cells [85]. The TES gene,<br />

which we instead find to be expressed only in NS cell lines (Fig 6.14), has been<br />

recently identified as a tumour suppressor that is methylated in tumours and<br />

is responsible for repressing cell growth. It is possibly inactivated by transcrip-<br />

tional silencing resulting from CpG island methylation [493] and is interestingly<br />

169


7.3 Tumour Expression Correlation Results<br />

located on the long arm <strong>of</strong> chromosome 7, which we know is gained in our GNS<br />

cell lines. Thus, very effective transcriptional silencing mechanisms must be<br />

actively suppressing the TES gene. Finally, PLS3 encodes for an actin-binding<br />

protein that has yet to become associated with any form <strong>of</strong> neoplasia and that<br />

we find to be up-regulated in our GNS cell lines with respect to NS cell lines<br />

as measured by qRT-PCR.<br />

Figure 7.3: Correlation with glioblastoma subtype expression signatures for tissue<br />

samples and cell lines interrogated by Tag-seq. (a) Shows the heatmap from<br />

the Verhaak et al [511] paper and in (b) colours indicate the correlation (Pearson<br />

R) between subtype-specific centroid values determined by Verhaak et al [511] and<br />

gene expression in our indicated Tag-seq measured samples. <strong>Cells</strong> showing positive<br />

correlation are labeled with p-values indicating the significance <strong>of</strong> the correlation.<br />

7.3 Tumour Expression Correlation<br />

To investigate whether the identified core set <strong>of</strong> differentially expressed genes<br />

that included 32 up-regulated and 60 down-regulated genes, showed similar<br />

expression patterns in primary tumours as in GNS lines, we made use <strong>of</strong> a<br />

cohort <strong>of</strong> public microarray data (see Methods, table 5.4).<br />

The core set <strong>of</strong> differentially expressed genes included genes with established<br />

roles in glioblastoma, such as PTEN [326] and CEBPB [85], as well as others<br />

not previously implicated in the disease. It was our interest to investigate<br />

170


7.3 Tumour Expression Correlation Results<br />

how each <strong>of</strong> these genes behaved in different settings: stem cell component<br />

<strong>of</strong> the tumour (GNS cells), primary tumour and lower grade gliomas, given<br />

they differ significantly from one another under a biological perspective. In<br />

fact, the tissues in the primary tumours comprise a heterogeneous mixture<br />

<strong>of</strong> cell types, whilst our GNS culture system has been designed to maintain<br />

the very specific stem cell component <strong>of</strong> the tumour in culture. Thus, we did<br />

not expect a perfect agreement between tissue and cell based results. Fur-<br />

thermore, assessing the behaviour <strong>of</strong> these same genes in lower grade gliomas<br />

could give us insights into the key players that determine the severity <strong>of</strong> the<br />

disease. Considering that our GNS cell lines have all been classified as primary<br />

GBMs, the differences observed from the lower grade glioma datasets will be<br />

especially meaningful in telling us how the same genes behave in the two dif-<br />

ferent categories (if we approximate that a significant percentage <strong>of</strong> the lower<br />

grade gliomas transform into higher grade gliomas such as GBMs).<br />

Panels a and c <strong>of</strong> figure 7.4 compare the GBM data in the TCGA dataset to<br />

the non-neoplastic brain tissue data in the TCGA dataset (altogether referred<br />

to as dataset GBM in Methods), for the core up-regulated (a) and core down-<br />

regulated (c) genes. Panels b and d, on the other hand, compare the GBM<br />

(grade IV glioma) data to the grade III glioma data in the combined Phillips<br />

and Freije datasets (altogether referred to as dataset HGG in Methods), for<br />

the core up-regulated (b) and core down-regulated (d) genes. From this figure<br />

we observe that there is a clear trend for the core up-regulated genes to be<br />

more highly expressed in glioblastoma tumours than in non-neoplastic brain<br />

tissue (Fig 7.4a; p = 0.02, randomisation test) and an opposite trend for the<br />

core down-regulated genes (Fig 7.4c; p = 3x10 -5 ). This means that, out <strong>of</strong> all<br />

the core differentially expressed genes that we measured with Tag-seq, a signif-<br />

icant part <strong>of</strong> the ones we found to be more highly expressed in GNS cell lines<br />

with respect to NS cell lines are also more highly expressed in primary GBM<br />

with respect to non-neoplastic brain tissue (histogram bars in the direction <strong>of</strong><br />

positive average log2(FC) identify these genes in panel a <strong>of</strong> figure 7.4).<br />

Similarly, a significant part <strong>of</strong> the core differentially expressed genes that we<br />

found to be down-regulated in our GNS cell lines with respect to our NS cell<br />

lines, are also down-regulated in primary GBM with respect to non-neoplastic<br />

brain tissue (histogram bars in the direction <strong>of</strong> negative average log2(FC) iden-<br />

tify these genes in panel c <strong>of</strong> figure 7.4). The colour <strong>of</strong> the histogram bars<br />

represents the level <strong>of</strong> significance <strong>of</strong> the comparison with black indicating a<br />

p-value0.01.<br />

171


7.3 Tumour Expression Correlation Results<br />

Figure 7.4: Core gene expression changes in GNS lines are mirrored in glioblastoma<br />

tumours. Gene expression in tumours for the core up-regulated (a, b) and downregulated<br />

(c, d) genes. The gene sets were identified by comparison <strong>of</strong> Tag-seq<br />

expression pr<strong>of</strong>iles for GNS and NS cell lines. Bars depict average FC between<br />

glioblastoma and non-neoplastic brain tissue (a, c) and between glioblastoma and<br />

grade III astrocytoma (b, d). Black bars indicate genes with significant differential<br />

expression in the microarray data (p < 0.01). The heatmaps show expression in<br />

individual samples relative to the average level in non-neoplastic brain (a, c) or<br />

grade III astrocytoma (b, d). The side dots point to the genes that belong to the<br />

cohort <strong>of</strong> 29 differentially expressed genes measured via qRT-PCR and have the same<br />

colour gradient adopted in figure 6.13. One gene (CHCHD10) not quantified in the<br />

TCGA dataset is omitted from panel a.<br />

172


7.3 Tumour Expression Correlation Results<br />

The orientation <strong>of</strong> the bar represents the direction <strong>of</strong> FC and the height <strong>of</strong><br />

the bar the magnitude <strong>of</strong> the FC (−3 < F C > 3). We hypothesised that ex-<br />

pression <strong>of</strong> these genes might also differ between glioblastoma and less severe<br />

gliomas and, upon examination <strong>of</strong> the expression patterns for grade III gliomas<br />

and glioblastoma in the dataset HGG, we found the core up-regulated genes to<br />

be more highly expressed in glioblastoma than in grade III glioma (Fig 7.4b;<br />

p < 10 -6 ), while the core down-regulated genes showed the opposite pattern<br />

(Fig 7.4d; p < 10 -5 ). Thus, <strong>of</strong> the core differentially expressed genes we mea-<br />

sured with Tag-seq, a significant portion <strong>of</strong> the highly expressed ones in GNS<br />

cell lines with respect to NS cell lines, were also more highly expressed in grade<br />

IV gliomas (GBMs) with respect to grade III gliomas (histogram bars in the<br />

direction <strong>of</strong> positive average log2(FC) identify these genes in panel b <strong>of</strong> figure<br />

7.4). This trend may be indicative <strong>of</strong> the fact that the core up-regulated genes<br />

in our GNS cell lines that are also up-regulated in GBM primary tumours,<br />

belong to a cohort <strong>of</strong> genes involved in regulatory networks already activated<br />

in grade III gliomas. The very fact that the same genes are similarly over ex-<br />

pressed in our GNS cell lines and in grade III gliomas may be an indication <strong>of</strong> a<br />

specific prognosis, namely the progression to a higher grade glioma. Of course,<br />

at this level <strong>of</strong> analysis this is only a hypothesis and it would be interesting to<br />

test it further and try to establish if it can be used as a pre-diagnostic tool.<br />

Similarly to the matching trends observed for the core up-regulated genes,<br />

a significant portion <strong>of</strong> the differentially expressed genes that we found to<br />

be down-regulated in GNS cell lines with respect to NS cell lines, were also<br />

down-regulated in grade IV gliomas (GBMs) with respect to grade III gliomas<br />

(histogram bars in the direction <strong>of</strong> negative average log2(FC) identify these<br />

genes in panel d <strong>of</strong> figure 7.4). This opposite trend is not surprising consid-<br />

ering the hypothesis mentioned before. If the regulatory networks involved in<br />

the progression <strong>of</strong> the disease from a grade III to a grade IV have already been<br />

activated in grade III gliomas, then a set <strong>of</strong> complementary genes and regu-<br />

latory networks must be silenced and rendered transcriptionally inactive. In<br />

this scenario postulating a diagnostic tool that derives from the combination<br />

<strong>of</strong> the two oppositely moving groups <strong>of</strong> genes may serve a pre-diagnostic tool<br />

purpose well. Of course, a specific study would need to be carried to verify the<br />

validity <strong>of</strong> this hypothesis.<br />

The dots next to the gene names on the sides <strong>of</strong> the heatmaps in figure 7.4 are<br />

there to highlight whether a gene also belongs to the cohort <strong>of</strong> the 29 genes<br />

found via qRT-PCR to distinguish GNS cell lines from NS cell lines. The colour<br />

173


7.3 Tumour Expression Correlation Results<br />

<strong>of</strong> the dot indicates the direction (red or blue) and the magnitude (intensity<br />

<strong>of</strong> red or blue) <strong>of</strong> the FC as measured via qRT-PCR and in doing so adopts<br />

the same colour scheme <strong>of</strong> the heatmap in 7.4. Table 7.4 summarises all the<br />

information gathered in the literature searches for the 29 genes differentially<br />

expressed as measured via qRT-PCR, information that is expanded upon in<br />

the paragraphs below.<br />

Up-regulated in GNS cell lines. The genes HOXD10, CD9, PLA2G4A,<br />

MT2A, SULF2, DDIT3, PLS3, CEBPB, PRSS12 and LYST are all highly ex-<br />

pressed in both GNS cell lines and primary GBMs. On the contrary, genes<br />

FOXG1, LMO4, ADD2 and PDE1C are up-regulated in GNS cell lines but are<br />

down-regulated in primary GBMs.<br />

Interestingly, FOXG1 is proposed to act as an oncogene in GBM by suppress-<br />

ing the growth inhibitory effects <strong>of</strong> TGFβ [448], so that the up-regulation we<br />

observe in GNS cell lines (Fig 7.4a) could be a cell type specific regulation that<br />

especially affects the stem cell component <strong>of</strong> the tumour. This would corrob-<br />

orate the hypothesis that NS cells are candidates for tumour-initiating cells<br />

in GBM. Also this up-regulation is not mirrored in grade III astrocytomas,<br />

indicating different oncogenic factors in action (Fig 7.4b).<br />

The transcriptional regulator LMO4 is <strong>of</strong> particular interest as it is involved in<br />

the development <strong>of</strong> multiple organs, including the CNS, and its expression is<br />

elevated in several cancers [202,339,475,488,542]. LMO4 is especially well stud-<br />

ied as an oncogene in breast cancer and regulated through the phosphoinosi-<br />

tide 3-kinase pathway [340], which is commonly affected in glioblastoma [326].<br />

Similarly to FOXG1, the up-regulation <strong>of</strong> LMO4 in GNS cell lines but not<br />

in primary GBM (Fig 7.4a) could be reflective <strong>of</strong> its oncogenic role in GBM<br />

tumour-initiating cells. Interestingly, LMO4 could be an oncogene early at<br />

work in the progression <strong>of</strong> the disease from a lower to a higher grade, as its<br />

up-regulation is observed in grade III astrocytomas (Fig 7.4b).<br />

The ADD2 gene encodes a cytoskeletal protein that interacts with FYN, a<br />

tyrosine kinase promoting cancer cell migration [456,533]. The up-regulation<br />

<strong>of</strong> this gene in GNS cell lines is reflective <strong>of</strong> the invasiveness that charac-<br />

terises GBM, and is possibly lost through differentiation since ADD2 appears<br />

to be down-regulated in primary GBM data (Fig 7.4a). The small level <strong>of</strong><br />

up-regulation observed for this gene in grade III astrocytomas (Fig 7.4b) is a<br />

reflection <strong>of</strong> its activity early on in the progression <strong>of</strong> the disease.<br />

PDE1C is a cyclic nucleotide phosphodiesterase gene that we observe to be up-<br />

regulated in GNS cell lines and down-regulated in primary GBM (Fig 7.4a).<br />

174


7.3 Tumour Expression Correlation Results<br />

Table 7.4: Literature survey for the 29 genes found to distinguish GNS from NS lines across a panel <strong>of</strong> 21 cell lines. The table details whether each<br />

gene has previously been implicated in glioma or other neoplasias, and includes references to relevant publications.<br />

Gene (aliases) Category Evidence in Evidence Selected references (PubMed IDs)<br />

glioma? other cancers?<br />

ADD2 Up-regulated in GNS No No Shima et al. 2001 (11526103), Pariser et al. 2005 (16105548), Ferrandi et al. 2010 (19838659)<br />

CD9 Up-regulated in GNS Yes Yes Shi et al. 2000 (10662783), Kawashima et al. 2002 (12185197), Zöller 2009 (19078974), Kolesnikova et al. 2009<br />

(19107234), Lafleur and Hemler 2009 (19211836)<br />

CEBPB Up-regulated in GNS Yes Yes Homma et al. 2006 (16465418), Nerlov 2007 (17658261), Zahnov 2009 (19351437), Carro et al. 2010 (20032975)<br />

DDIT3 Up-regulated in GNS Limited Yes Oyadomari and Mori 2004 (14685163), Ragel et al. 2007 (17486380), Kaul and Maltese 2009 (19724676), Meng et<br />

al. 2009 (19549908)<br />

FOXG1 Up-regulated in GNS Yes Yes Seoane et al. 2004 (15084259), Adesina et al. 2007 (17522785)<br />

HMGA2 Down-regulated in GNS Limited Yes Akai et al. 2004 (15497774), Young and Narita 2007 (17473167), Cleynen and Van de Ven 2008 (18202751), Liu et<br />

al. 2010 (20368557)<br />

HOXD10 Up-regulated in GNS Limited Yes Osborne et al. 1998 (9773404), Reddy et al. 2008 (18922890), Ma et al. 2007 (17898713), Baffa et al. 2009<br />

(19593777), Sasayama et al. 2009 (19536818), Sun et al. 2011 (21419107)<br />

IRX2 Down-regulated in GNS No Yes Adamowicz et al. 2006 (16752383)<br />

LMO4 Up-regulated in GNS No Yes Sum et al. 2002 (11751867), Mizunuma et al. 2003 (12771919), Sum et al. 2005 (15897450), Taniwaki et al. 2006<br />

(16865272), Yu et al. 2008 (19099607), Montanez-Wiscovich et al. 2009 (19648968)<br />

LYST Up-regulated in GNS No No Kaplan et al. 2008 (18043242)<br />

MAF Down-regulated in GNS No Yes Rasmussen et al. 2003 (14692531), Hurt et al. 2004 (14998494), Murakami et al. 2007 (18059226), Peng et al. 2007<br />

(17823980), Natkunam et al. 2009 (19687312)<br />

MAP6 Down-regulated in GNS No Yes Vater et al. 2009 (19016712)<br />

MT2A Up-regulated in GNS Yes Yes Maier et al. 1997 (9444362), Cui et al. 2003 (12646258), Yamasaki et al. 2007 (17914565), Krona et al. 2007<br />

(17982672), Lim et al. 2009 (19062161), Puca et al. 2009 (18996371)<br />

MYL9 Down-regulated in GNS No Yes Medjkane et al. 2009 (19198601), Lu et al. 2010 (21139803)<br />

NDN Down-regulated in GNS Limited Yes Aizawa et al. 1992 (1394972), Hu et al. 2003 (12913118), Chapman and Knowles 2009 (19626646), Jörnsten et al.<br />

2011 (21525872)<br />

NELL2 Down-regulated in GNS Limited Yes Kuroda et al. 1999 (10548494), Maeda et al. 2001 (11803583), DiLella et al. 2001 (11304808), Nelson et al. 2004<br />

(15183717), Fassunke et al. 2008 (18819986)<br />

PDE1C Up-regulated in GNS No No Das and Sharma 2005 (16142372), Vatter et al. 2005 (15816855), Dolci et al. 2006 (16455054)<br />

PLA2G4A Up-regulated in GNS Limited Yes Hernandez et al. 2000 (10838595), Moolwaney and Igwe 2005 (15950779), Linkous et al. 2010 (20729478), Jeong et<br />

al. 2010 (20944117), Han et al. 2010 (20683962), Caiazza et al. 2011 (21119660)<br />

PLCH1 Down-regulated in GNS No No Stewart et al. 2007 (17895620), Kim et al. 2011 (21262355)<br />

PLS3 Up-regulated in GNS No Yes Lin et al. 1993 (8428952), Arpin et al. 1994 (7806577), Su et al. 2003 (14612505), Kari et al. 2003 (12782714),<br />

Ikeda et al. 2005 (16142308), Capriotti et al. 2008 (18569641)<br />

PRSS12 Up-regulated in GNS No No Mitsui et al. 2007 (17223089), Matsumoto-Miyai et al. 2009 (19303856)<br />

PTEN Down-regulated in GNS Yes Yes The Cancer Genome Atlas Research Network 2008 (18772890), Hollander et al. 2011 (21430697)<br />

SDC2 Down-regulated in GNS Limited Yes Fears et al. 2006 (16574663), Watanabe et al. 2006 (16132527), Theocaris et al. 2008 (20840587)<br />

ST6GALNAC5 Down-regulated in GNS Yes Yes Bos et al. 2009 (19421193), Kroes et al. 2010 (20616019), Oster et al. 2011 (21400501)<br />

SULF2 Up-regulated in GNS Yes Yes Johansson et al. 2005 (15750623), Morimoto-Tomita et al. 2005 (16331886), Dai et al. 2005 (16192265), Lemjabbar-<br />

Alaoui et al. 2010 (19855436), Lai et al. 2010 (20725905), Phillips et al. 2012 (22293178)<br />

SYNM Down-regulated in GNS Yes Yes Jing et al. 2005 (15657940), Pan et al. 2008 (18509200), Noetzel et al. 2010 (20543860), Sun et al. 2010 (19853601),<br />

Liu et al. 2011 (21144834), Pitre et al. 2012 (22337773)<br />

TAGLN Down-regulated in GNS Limited Yes Gunnersen et al. 2000 (11008214), Shields et al. 2002 (11773051), Yeo et al. 2006 (16402363), Assinder et al. 2009<br />

(18378184), Zhao et al. 2009 (19329940), Kim et al. 2010 (20705054), Yeo et al. 2010 (20336793), Prasad et al.<br />

2010 (20012321)<br />

TES Down-regulated in GNS Yes Yes Tatarelli et al. 2000 (10950921), Tobias et al. 2001 (11420696), Mueller et al. 2007 (16909125), Martinez et al.<br />

2009 (19550145), Gunduz et al. 2009 (19289703), Ma et al. 2010 (20626849), Weeks et al. 2010 (20573277), Qui et<br />

al. 2010 (20180808)<br />

TUSC3 Down-regulated in GNS Limited Yes MacGrogan et al. 1996 (8661104), Bookstein et al. 1997 (9088270), Ahuja et al. 1998 (9850084), Li et al. 1998<br />

(9671399), Bashyam et al. 2005 (16036106), Pils et al. 2005 (16270321), Guervos et al. 2007 (17641416), Cooke et<br />

al. 2008 (18840272), Arasaradnam et al. 2010 (20505342), Bui et al. 2010 (19812376)<br />

175


7.3 Tumour Expression Correlation Results<br />

Up-regulation <strong>of</strong> PDE1C has been associated with proliferation in other cell<br />

types through hydrolysis <strong>of</strong> cAMP and cGMP [126,437]. Up-regulation in<br />

GNS cell lines may foster the proliferation that characterises these cells, which<br />

may be lost in more differentiated progeny belonging to the primary tumour<br />

sample. In figure 7.4b we observe a down-regulation <strong>of</strong> PDE1C in grade III<br />

astrocytomas.<br />

Altogether, FOXG1, LMO4, ADD2 and PDE1C may be part <strong>of</strong> the GNS ex-<br />

pression pr<strong>of</strong>ile that defines the stem cell identity <strong>of</strong> GBM and is therefore lost<br />

through the differentiation pathways undertaken by the rest <strong>of</strong> the tumour<br />

cells that do not retain their stem cell identities, according to the cancer stem<br />

cell hypothesis.<br />

Of the ten genes that are highly expressed in both GNS cell lines and primary<br />

GBMs, HOXD10 encodes a protein with a homeobox DNA-binding domain<br />

that is known to be involved in limb development and differentiation [82].<br />

HOXD10 protein levels are suppressed by a microRNA (miR-10b) which is<br />

highly expressed in gliomas, and it has been suggested that HOXD10 suppres-<br />

sion by miR-10b promotes invasion [476]. Interestingly, the HOXD10 mRNA<br />

up-regulation we observe in GNS cell lines and GBM tumours is not mirrored<br />

in grade III astrocytoma, perhaps reflective <strong>of</strong> the less invasive phenotype. In<br />

fact, miR-10b is present at higher levels in glioblastoma compared to gliomas<br />

<strong>of</strong> lower grade [476].<br />

The CD9 gene encodes a cell-surface glycoprotein, or antigen, that has been<br />

previously implicated in glioma with a role in adhesion and migration. In the<br />

CNS CD9 is expressed in the myelin sheath and is believed to suppress the<br />

metastatic potential <strong>of</strong> some human tumours including gliomas [221]. We find<br />

up-regulation <strong>of</strong> CD9 in GNS cell lines as well as primary GBM (Fig 7.4a),<br />

but a down-regulation in grade III astrocytoma (Fig 7.4b), perhaps reflective<br />

<strong>of</strong> the lower invasive potential <strong>of</strong> the latter.<br />

The PLA2G4A gene encodes a phospholipase enzyme that catalyses the hy-<br />

drolysis <strong>of</strong> membrane phospholipids to lipid-based cellular hormones that then<br />

regulate a variety <strong>of</strong> intracellular pathways. PLA2G4A has not been linked to<br />

GBM but has been implicated in other neoplasias [192,285,341]. We observed<br />

an up-regulation <strong>of</strong> PLA2G4A in GNS cell lines as well as primary GBM (Fig<br />

7.4a), but a down-regulation in grade III astrocytoma (Fig 7.4b).<br />

The MT2A gene encodes a metallothionein that has yet to be implicated in<br />

gliomas, but has been observed to interact with the kinase domain <strong>of</strong> a member<br />

<strong>of</strong> the PKC family in prostate cancer [422] and TP53 in breast cancer epithe-<br />

176


7.3 Tumour Expression Correlation Results<br />

lial cells to possibly regulate apoptosis in the latter [373]. Also in the case <strong>of</strong><br />

MT2A we observed an up-regulation in GNS cell lines as well as primary GBM<br />

(Fig 7.4a), but a down-regulation in grade III astrocytoma (Fig 7.4b).<br />

The SULF2 gene encodes a sulfatase that edits the sulfation status <strong>of</strong> heparan<br />

sulfate proteoglycans on the outside <strong>of</strong> cells and, in this way, regulates criti-<br />

cal signaling pathways [433]. Disregulation <strong>of</strong> SULF2 has been implicated in<br />

non-small cell lung cancer [268], pancreatic cancer [357], hepatocellular car-<br />

cinoma [254], breast cancer [344], and gliomas, in which knock-down <strong>of</strong> the<br />

SULF2 gene resulted in decreased GBM growth in vivo in mice. Molecu-<br />

larly, ablation <strong>of</strong> SULF2 resulted in decreased PDGFRα phosphorylation and<br />

decreased downstream MAPK signaling activity. Interestingly, <strong>of</strong> this obser-<br />

vation on the proneural GBM subtype defined by Verhaak et al [511] that is<br />

characterized by aberrations in PDGFRα, showed the strongest SULF2 expres-<br />

sion [391]. In our observations SULF2 was up-regulated in GNS cell lines and<br />

primary GBM (Fig 7.4a), in line with the observations <strong>of</strong> Phillips et al [391]<br />

that made it a candidate oncogene. We observed SULF2 to also be strongly<br />

up-regulated in grade III astrocytoma (Fig 7.4b) indicating the possibility <strong>of</strong> a<br />

regulatory impact on behalf <strong>of</strong> SULF2 early in the progression <strong>of</strong> the disease.<br />

The DDIT3 gene encodes the pro-apoptotic protein CHOP that is known to<br />

drive the down-regulation <strong>of</strong> the anti-apoptotic mitochondrial protein Bcl-2,<br />

thereby favouring apoptosis through the activation <strong>of</strong> cytochrome c and cas-<br />

pase 3. Studies in hepatoma and pheochromocytoma cell lines have shown that<br />

the transcription factor encoded by CEBPB (C/EBPβ) promotes the expres-<br />

sion <strong>of</strong> DDIT3 [324] and thus <strong>of</strong> CHOP, which in turn can inhibit C/EBPβ by<br />

dimerizing with it and acting as a dominant negative [324]. This interplay be-<br />

tween CEBPB and DDIT3 may be relevant for glioma therapy development, as<br />

DDIT3 induction in response to a range <strong>of</strong> compounds sensitises glioma cells to<br />

apoptosis [217]. In line with the pro-apoptotic role <strong>of</strong> DDIT3 described, we ob-<br />

served up-regulation <strong>of</strong> the gene in GNS cell lines and primary GBM (Fig 7.4a)<br />

and down-regulation in grade III astrocytoma (Fig 7.4b); similarly, we found<br />

CEBPB to be up-regulated in GNS cell lines and primary GBM (Fig 7.4a),<br />

and down-regulated in grade III astrocytoma (Fig 7.4b). PLS3 (T-plastin)<br />

encodes a regulator <strong>of</strong> actin organisation and its over-expression in the CV-1<br />

fibroblast-like cell line resulted in partial loss <strong>of</strong> adherence [27]. The elevated<br />

levels <strong>of</strong> PLS3 expression we observe in GNS cell lines and primary GBM may<br />

thus be relevant to the invasive phenotype. Accordingly, the less invasive lower<br />

grade III astrocytoma shows down-regulation <strong>of</strong> PLS3 (Fig 7.4b).<br />

177


7.3 Tumour Expression Correlation Results<br />

The PRSS12 gene encodes a protease that can activate tissue plasminogen acti-<br />

vator (tPA) [335], an enzyme which is highly expressed by glioma cells and has<br />

been suggested to promote invasion [168]. We observe a slight up-regulation<br />

in the GNS cell lines and primary GBM <strong>of</strong> PRSS12 (Fig 7.4a), and a slight<br />

down-regulation in grade III astrocytomas (Fig 7.4b).<br />

The LYST gene encodes a vesicular transport protein called the "lysosomal<br />

trafficking regulator" that so far has not been implicated in any neoplasia but<br />

is known to be associated with a rare recessive disorder (Chédiak-Higashi syn-<br />

drome) caused by a microtubule polymerisation defect that decreases phago-<br />

cytosis ability [87]. In line with the hypothetical increase <strong>of</strong> cellular activities<br />

and rates, and thus <strong>of</strong> vesicular transport, in GNS cells, we observed LYST to<br />

be up-regulated in GNS cell lines and primary GBM (Fig 7.4a), and slightly<br />

down-regulated in grade III astrocytomas (Fig 7.4b).<br />

Down-regulated in GNS cell lines. The panels c and d <strong>of</strong> figure 7.4 the core<br />

down-regulated genes found through Tag-seq are used to evaluate the compar-<br />

ison <strong>of</strong> the GBM data to the non-neoplastic tissue data (TCGA dataset), and<br />

the comparison <strong>of</strong> the GBM (grade IV) to the grade III astrocytoma (HGG<br />

dataset). In this comparison the genes that were also identified via qRT-PCR<br />

as being differentially expressed between GNS cell lines and NS cell lines, are<br />

highlighted by the blue colour gradient dots. The trends highlighted in figure<br />

7.4 show matching down-regulation for genes NELL2, TUSC3, ST6GALNAC5,<br />

PLCH1, NDN, MAP6 and PTEN in GNS cell lines and primary GBM, and non<br />

matching down-regulation in GNS cell lines and up-regulation in primary GBM<br />

for genes MAF, MYL9, HMGA2, SDC2, SYNM, IRX2, TES and TAGLN.<br />

The MAF gene encodes a transcription factor and oncoprotein that belongs to<br />

the same AP-1 super-family <strong>of</strong> JUN and FOS. MAF has been associated with<br />

multiple myeloma whereby its up-regulation is suggested to enhance myeloma<br />

proliferation and adhesion to the bone marrow [406]. Although not previously<br />

linked to glioma, we find MAF to be down-regulated in GNS cell lines but<br />

slightly up-regulated in primary GBM, perhaps as an indicator <strong>of</strong> its cell type<br />

restricted proliferation enhancing functions (Fig 7.4c). Accordingly, we ob-<br />

served a slight down-regulation <strong>of</strong> MAF in grade III astrocytoma with respect<br />

to primary GBM (Fig 7.4d).<br />

The protein encoded by the MYL9 gene is a myosin light chain that has<br />

previously been associated with the stem cell component <strong>of</strong> lung adenocar-<br />

cinoma [447] and medullary breast cancer [54] in gene expression pr<strong>of</strong>iling<br />

studies. MYL9 has never been implicated in glioma and we observed down-<br />

178


7.3 Tumour Expression Correlation Results<br />

regulation <strong>of</strong> its expression in GNS cell lines opposed to its up-regulation in<br />

primary GBM (Fig 7.4c), and a strong down-regulation in grade III astrocy-<br />

toma (Fig 7.4d).<br />

The HMGA2 gene encodes a transcriptional regulator that belongs to the non-<br />

histone family <strong>of</strong> structural proteins. The members <strong>of</strong> this family act as chro-<br />

matin architectural factors and contain structural DNA-binding domains that<br />

allow them to act as transcriptional factors. We find HMGA2 to be down-<br />

regulated in GNS lines and up-regulated in primary GBM (Fig 7.4c), and<br />

slightly down-regulated in grade III astrocytoma (Fig 7.4d). Low or absent<br />

protein expression <strong>of</strong> HMGA2 has been observed in GBM compared to low<br />

grade gliomas [14] and HMGA2 polymorphisms have been associated with<br />

survival time in GBM [295].<br />

The SDC2 gene encodes a transmembrane heparan sulfate proteoglycan that<br />

participates as an extracellular matrix receptor in the processes <strong>of</strong> cell prolifera-<br />

tion, cell migration and cell-matrix interaction. An altered expression <strong>of</strong> SDC2<br />

has been detected in esophageal carcinoma [201], colon carcinoma [184], fi-<br />

brosarcoma [380], prostate cancer [108,407] and gliomas, where over-expression<br />

<strong>of</strong> SDC2 promotes membrane protrusion, migration, capillary tube formation<br />

and cell-cell interactions in microvascular endothelial cells [141]. We found<br />

SDC2 to be down-regulated in GNS cell lines and grade III astrocytoma, but<br />

slightly up-regulated in primary GBM (Fig 7.4).<br />

The SYNM gene is a type IV intermediate filament that has recently been<br />

shown to interact with the LIM domain protein Zyxin, thereby possibly mod-<br />

ulating cell adhesion and cell motility. Aberrant SYNM promoter methylation<br />

has been associated with early breast cancer relapse [362]. In gliomas SYNM<br />

has been found to promote AKT-dependent GBM cell proliferation by antago-<br />

nising protein phosphatase PP2A, the major regulator <strong>of</strong> Akt dephosphoryla-<br />

tion [396]. We found SYNM to be down-regulated in GNS cell lines and grade<br />

III astrocytoma, but slightly up-regulated in primary GBM, although with a<br />

relatively high p-value (Fig 7.4). The IRX2 gene is a iroquois-class homeobox<br />

genes that has been associated with development <strong>of</strong> the vertebrate embryo<br />

and in humans specifically in the development <strong>of</strong> brain [322] and breast [274].<br />

Over-expression <strong>of</strong> IRX2 has been detected in s<strong>of</strong>t tissue sarcomas [7]. IRX2<br />

has been proposed to enhance antitumour immune responses in that ex vivo<br />

pre-treatment <strong>of</strong> CD8 + T cells with IRX-2 provided protection from tumour-<br />

induced apoptosis [112]. In line with this suggested role <strong>of</strong> IRX2, we observed<br />

the gene to be strongly down-regulated in GNS cell lines and grade III astro-<br />

179


7.3 Tumour Expression Correlation Results<br />

cytoma, but up-regulated in primary GBM (Fig 7.4).<br />

Finally, we found the TAGLN and TES genes to be absent or low in most GNS<br />

lines, but displaying the opposite trend in GBM tissue compared to normal<br />

brain (Fig 7.4c) or grade III astrocytoma (Fig 7.4d). Interestingly, the TES<br />

gene is located on chromosome 7, which is known to be gained in all our GNS<br />

cell lines (see 6.3 section). A strong regulatory effect must be in action in<br />

GNS cell lines to effect the down-regulation <strong>of</strong> TES. Similarly to the SYNM<br />

gene, TES has been shown to interact with the LIM domain protein Zyxin and<br />

therefore is believed to have a role in cell motility and is <strong>of</strong>ten found in focal<br />

adhesions [109]. The TAGLN gene encodes an actin protein found in fibrob-<br />

lasts and smooth muscle, the expression <strong>of</strong> which is down-regulated in many<br />

cell lines and may be an early marker for the onset <strong>of</strong> transformation [260].<br />

Both TAGLN and TES have been characterised as tumour suppressors in ma-<br />

lignancies outside the brain and TES is known to <strong>of</strong>ten be silenced by promoter<br />

hypermethylation in GBM [30,349].<br />

Altogether, MAF, MYL9, HMGA2, SDC2, SYNM, IRX2, TES and TAGLN<br />

may be part <strong>of</strong> a GNS expression pr<strong>of</strong>ile that helps define the stem cell identity<br />

<strong>of</strong> these cells and the tumour that derives from them according to the cancer<br />

stem cell hypothesis. Interestingly, the expression pattern <strong>of</strong> most <strong>of</strong> these<br />

genes is mirrored in grade III astrocytomas.<br />

Of the seven genes that are down-regulated in both GNS cell lines and pri-<br />

mary GBMs, TUSC3 is a candidate tumour suppressor known to be silenced<br />

by promoter methylation in GBM, particularly in patients over 40 years <strong>of</strong><br />

age. Loss or down-regulation <strong>of</strong> TUSC3 has been found in other cancers, such<br />

as colon cancer, where its promoter becomes increasingly methylated with age<br />

in the healthy mucosa [12]. These data suggest that transcriptional changes<br />

in healthy aging tissue, such as TUSC3 silencing, may contribute to the more<br />

severe form <strong>of</strong> glioma in older patients. In line with its role as tumour suppres-<br />

sor, we found TUSC3 to be down-regulated in GNS cell lines, primary GBM<br />

and grade III astrocytoma (Fig 7.4).<br />

The PLCH1 gene is a member <strong>of</strong> the phospholipase C family <strong>of</strong> enzymes that<br />

cleave phosphatidylinositol 4,5-bisphosphate to generate second messengers in-<br />

ositol 1,4,5-trisphosphate and diacylglycerol. PLCH1 is thus involved in phos-<br />

phoinositol signaling [228], just like the frequently mutated phosphoinositide<br />

3-kinase complex [326]. We found PLCH1 to be down-regulated in GNS cell<br />

lines and primary GBM (Fig 7.4c) and slightly up-regulated in grade III as-<br />

trocytoma (Fig 7.4d).<br />

180


7.3 Tumour Expression Correlation Results<br />

The PTEN lipid phosphatase gene is a well known tumour suppressor that<br />

is frequently deleted or mutated in GBM and its down-regulation affects the<br />

correct regulation <strong>of</strong> the RTK/PI3K/PTEN pathway in which it acts as an<br />

instrumental player [326]. PTEN is mutated in a variety <strong>of</strong> other cancers be-<br />

sides gliomas, including prostate, breast, endometrial cancer and melanoma<br />

[377,378,439,514]. We observe the expression <strong>of</strong> PTEN to be down-regulated<br />

in GNS cell lines and primary GBM (Fig 7.4c) but up-regulated in grade III<br />

astrocytoma (Fig 7.4d).<br />

The ST6GALNAC5 gene belongs to the sialyltransferase family <strong>of</strong> proteins<br />

that modify proteins and ceramides on the cell surface to alter cell-cell or cell-<br />

extracellular matrix interactions [498]. ST6GALNAC5 facilitates the transmi-<br />

gration <strong>of</strong> cancer cells through the blood-brain barrier and is known to mediate<br />

breast cancer metastasis to the brain [67]. In GBM ST6GALNAC5 has already<br />

been observed to be down-regulated with respect to normal brain tissue [250],<br />

and we observed it to be down-regulated in GNS cell lines as well as primary<br />

GBM and slightly up-regulated in grade III astrocytoma (Fig 7.4).<br />

The NDN gene is an imprinted gene expressed exclusively from the pater-<br />

nal allele that has no introns and acts as a growth suppressor. Studies in<br />

mice suggest that the Necdin protein suppresses growth in postmitotic neu-<br />

rons [209,487]. Necdin has also been suggested to interact with and negatively<br />

regulate HIF1α, a factor that mediates cellular homeostatic responses like<br />

angiogenesis to reduce O2 availability [342]. The expression <strong>of</strong> NDN is also<br />

known to be down-regulated due to the transcriptional control implemented<br />

by STAT3 in human melanoma, prostate cancer and breast cancer [188]. We<br />

found NDN to be strongly down-regulated in GNS cell lines and primary GBM<br />

but strongly up-regulated in grade III astrocytoma (Fig 7.4).<br />

The MAP6 gene encodes a microtubule-associated protein that is calmodulin-<br />

binding and calmodulin-regulated and is therefore involved in microtubule sta-<br />

bilisation. MAP6 has yet to be associated with any neoplasia and we observed<br />

it to be down-regulated in GNS cell lines and primary GBM, and slightly<br />

up-regulated in grade III astrocytoma (Fig 7.4). Finally, the NELL2 gene en-<br />

codes a glycoprotein containing several EGF-like repeats that is found in the<br />

cytoplasm and is involved in neural cell growth and differentiation as well as<br />

oncogenesis [521]. Together with NELL1, NELL2 is predominantly expressed<br />

in neuroblastoma and other embryonal neuroepithelial tumours [305], but has<br />

yet to be characterised in glioblastoma. We observed its expression to be down-<br />

regulated in GNS cell lines and primary GBM, as well as grade III astrocytoma<br />

181


7.4 Survival Analysis Results<br />

(Fig 7.4). Overall, we can confirm that the set <strong>of</strong> core differentially expressed<br />

genes identified by Tag-seq defines an expression signature characteristic <strong>of</strong><br />

glioblastoma and related to glioma histological grade.<br />

7.4 Survival Analysis<br />

To explore the relevance in glioma <strong>of</strong> the patterns observed for the GNS vs. NS<br />

cell line comparison, we decided to integrate clinical information with tumour<br />

expression data. Although this analysis was not performed by the candidate it<br />

is included in this thesis because it is very relevant to the rest <strong>of</strong> the material<br />

and analyses performed by the candidate and will therefore help paint for the<br />

reader a more complete picture <strong>of</strong> the relevance <strong>of</strong> GNS cell lines in glioblas-<br />

toma cancer research.<br />

We first tested for associations between gene expression and survival time using<br />

the TCGA dataset, consisting <strong>of</strong> 397 glioblastoma cases (see Methods section<br />

5.9 for table 5.4). For each gene, we fitted a Cox proportional hazards model<br />

with gene expression as a continuous explanatory variable and computed a<br />

p-value by the score test (Table 7.5). The set <strong>of</strong> 29 genes found to distinguish<br />

GNS cells from NS cells across the 22 cell lines assayed by qRT-PCR was en-<br />

riched for low p-values compared to the complete set <strong>of</strong> 18,632 genes quantified<br />

in the TCGA dataset (p = 0.02, one-sided Kolmogorov-Smirnov test), demon-<br />

strating that expression analysis <strong>of</strong> GNS and NS lines had enriched for genes<br />

associated with patient survival. Seven <strong>of</strong> the 29 genes had a p-value


7.4 Survival Analysis Results<br />

Table 7.5: Survival tests for the 29 genes found via qRT-PCR to distinguish GNS<br />

cell lines from NS cell lines.<br />

Gene Category TCGA dataset Gravendeel dataset (GBM cases)<br />

Coefficient* p-value Probeset** Coefficient* p-value<br />

ADD2 Up -0.13 0.2858 237336_at -0.17 0.1420<br />

CD9 Up 0.18 0.0731 201005_at 0.17 0.0689<br />

CEBPB Up 0.19 0.1028 212501_at 0.17 0.0651<br />

DDIT3 Up 0.17 0.0128 209383_at 0.09 0.2777<br />

FOXG1 Up 0.13 0.0861 206018_at 0.11 0.0380<br />

HMGA2 Down 0.13 0.1456 1561633_at -0.84 0.2459<br />

HOXD10 Up 0.12 0.0108 229400_at 0.15 0.0021<br />

IRX2 Down -0.19 0.2346 228462_at -0.20 4.4x10 -4<br />

LMO4 Up 0.24 0.1046 209205_s_at 0.20 0.1435<br />

LYST Up 0.05 0.5590 203518_at 0.10 0.4151<br />

MAF Down 0.10 0.5873 209348_s_at 0.38 0.0074<br />

MAP6 Down 0.16 0.3063 235672_at -0.30 0.0087<br />

MT2A Up 0.16 0.1554 212185_x_at 0.27 0.0127<br />

MYL9 Down 0.08 0.3764 201058_s_at 0.15 0.0252<br />

NDN Down -0.04 0.4874 209550_at -0.22 6.0x10 -5<br />

NELL2 Down 0.08 0.1021 203413_at 0.14 0.0215<br />

PDE1C Up 0.20 0.0105 236344_at 0.21 0.0134<br />

PLA2G4A Up -0.06 0.3198 210145_at 0.30 2.9x10 -4<br />

PLCH1 Down 0.10 0.3165 214745_at 0.45 0.0094<br />

PLS3 Up 0.13 0.0381 201215_at 0.30 0.0069<br />

PRSS12 -0.11 0.1865 213802_at 0.20 0.0296<br />

PTEN Down -0.53 0.0047 228006_at -0.40 0.0062<br />

SDC2 Down 0.22 0.0044 212158_at 0.28 5.8x10 -4<br />

ST6GALNAC5 Down 0.01 0.9116 220979_s_at 0.08 0.2416<br />

SULF2 -0.11 0.1525 233555_s_at -l0.15 0.0930<br />

SYNM Down -0.06 0.5620 212730_at 0.08 0.2613<br />

TAGLN Down 0.03 0.5947 205547_s_at 0.17 0.0030<br />

TES Down -0.05 0.5759 202720_at 0.07 0.5499<br />

TUSC3 Down -0.14 0.0079 209227_at -0.18 0.0060<br />

*Fitted coefficient from Cox model; a positive coefficient indicates that higher expression<br />

is associated with poor survival and a negative coefficient indicates the<br />

opposite. ** For the Gravendeel dataset, the result for the most significant probeset<br />

interrogating the gene is shown; Up=Up-regulated; Down=Down-regulated.<br />

To test whether these findings generalise to independent clinical sample groups,<br />

we examined the glioblastoma datasets described by Gravendeel et al [176] and<br />

Murat et al [353], consisting <strong>of</strong> 141 and 70 cases, respectively (see Methods<br />

section 5.9 for table 5.4). The GNS signature score was correlated with pa-<br />

tient survival in both <strong>of</strong> these datasets (p = 3x10 5 and p = 0.006, respectively;<br />

Fig 7.5a). At the level <strong>of</strong> individual GNS signature genes, five genes were<br />

significantly associated with survival (p < 0.05) in both <strong>of</strong> the two largest<br />

183


7.4 Survival Analysis Results<br />

Figure 7.5: Association between GNS signature and other survival predictors. (a)<br />

Scatter plots demonstrate the correlation between GNS signature score and age at<br />

diagnosis for the TCGA (left) and Gravendeel (right) datasets. The regression line,<br />

Pearson correlation coefficient (r) and p-value indicating statistical significance <strong>of</strong><br />

the correlation are shown. (b) GNS signature score for samples in the Gravendeel<br />

dataset, stratified by IDH1 mutation status and histological grade. Blue circles<br />

represent individual samples (independent cases) and grey boxplots summarise their<br />

distribution. Only cases with known IDH1 status are shown (127 mutated, 77 wild<br />

type).<br />

glioblastoma datasets we investigated (TCGA and Gravendeel): HOXD10,<br />

PDE1C, PLS3, PTEN and TUSC3 (Table 7.5). In addition to glioblastoma<br />

(grade IV) tumours, Gravendeel et al also characterised 109 grade I-III glioma<br />

cases (see Methods section 5.9). Inclusion <strong>of</strong> these data in survival analyses<br />

made the association with GNS signature even more apparent (Fig 7.5b). This<br />

is consistent with the observation made in section 7.3 whereby core transcrip-<br />

tional alterations in GNS cells correlated with histological grade <strong>of</strong> primary<br />

tumours. Analysis <strong>of</strong> data from the studies <strong>of</strong> Phillips et al [390] and Freije et<br />

al [148], which pr<strong>of</strong>iled both grade III and IV gliomas (see Methods section 5.9<br />

for table 5.4), further confirmed the correlation between GNS signature and<br />

survival (Figure 7.5b). In summary, the association between GNS signature<br />

and patient survival was reproducible in five independent datasets comprising<br />

184


7.4 Survival Analysis Results<br />

867 glioma cases in total (see Methods section 5.9 for table 5.4).<br />

In trying to investigate the presence <strong>of</strong> a relationship to known predictors <strong>of</strong><br />

survival in glioma, we noted that the GNS signature scores correlated with<br />

patient age at diagnosis, suggesting that the GNS-related expression changes<br />

were associated with the more severe form <strong>of</strong> the disease observed in older pa-<br />

tients (Figure 7.6a). Of the genes contributing to the GNS signature, HOXD10,<br />

PLS3, PTEN and TUSC3 correlated with age both in the TCGA and Graven-<br />

deel datasets.<br />

Figure 7.6: Association between GNS signature score and patient survival. Kaplan-<br />

Meier plots illustrate the association between signature score and survival for (a)<br />

three independent glioblastoma datasets and (b) three datasets that include gliomas<br />

<strong>of</strong> lower grade (see Methods section 5.9). Higher scores indicate greater similarity<br />

to the GNS expression pr<strong>of</strong>ile. Hazard ratios and log-rank p-values were computed<br />

by fitting a Cox proportional hazards model to the data. Percentile thresholds were<br />

chosen for illustration; the association with survival is statistically significant across<br />

a wide range <strong>of</strong> thresholds and the p-values given in the text and Table 7.5 were<br />

computed without thresholding, using the score as a continuous variable.<br />

IDH1 mutation affecting codon 132 <strong>of</strong> the IDH1 gene is present in most grade<br />

III astrocytomas and a minority <strong>of</strong> glioblastomas, resulting in an amino acid<br />

change (R132H, R132S, R132C, R132G, or R132L). The presence <strong>of</strong> this muta-<br />

tion is associated with lower age at disease onset and better prognosis [383,517].<br />

As already mentioned in section 6.1, all 16 GNS lines pr<strong>of</strong>iled in this study<br />

were derived from glioblastoma tumours, and the IDH1 locus was sequenced in<br />

each cell line (data not shown) and none <strong>of</strong> the cell lines appeared to harbour<br />

the mutation. We therefore wanted to investigate whether the GNS signature<br />

was characteristic <strong>of</strong> IDH1 wild-type glioblastomas or not. We could perform<br />

this analysis thanks to the fact that IDH1 status had been determined for most<br />

cases in the TCGA and Gravendeel datasets (Table 7.5) [176,326,511]. As ex-<br />

pected, we found that gliomas with the IDH1 mutation tend to have lower GNS<br />

185


7.4 Survival Analysis Results<br />

Table 7.6: Significance <strong>of</strong> survival association for GNS signature and IDH1 status.<br />

dataset Number<br />

<strong>of</strong> cases<br />

Single covariate Two covariates (GBM cases)<br />

GNS signature IDH1 status GNS signature IDH1 status<br />

TCGA 270 5.3x10-5 0.0015 0.0091 0.1489<br />

Gravendeel,<br />

GBM cases<br />

118 2.7x10-5 0.0031 9.2x10-4 0.0840<br />

Gravendeel<br />

et al, grade<br />

I-III cases<br />

86 6.5x10-4 0.5776 6.3x-4 0.5408<br />

Wald test p-values, indicating association with survival, for each covariate in a Cox<br />

proportional hazards model with one or two covariates (GNS signature, IDH1 status<br />

or both). Cases with unknown IDH1 mutation status were excluded.<br />

signature scores than IDH1 wild-type gliomas <strong>of</strong> the same histological grade<br />

(Fig 7.6b). However, we also found that the GNS signature bore a stronger<br />

survival association than the IDH1 status (Table 7.6). In fact, the signature<br />

remained a significant predictor <strong>of</strong> patient survival when controlling for IDH1<br />

status (Table 7.6), demonstrating that it contributes independent information<br />

to the survival model and does not simply represent a transcriptional state <strong>of</strong><br />

IDH1 wild-type tumours. This was evident in glioblastomas as well as grade<br />

I-III gliomas; the effect is thus not limited to grade IV tumours.<br />

Finally, to investigate whether the correlation between GNS signature and<br />

age could be explained by the higher proportion <strong>of</strong> cases with IDH1 mutation<br />

among younger patients, we repeated the correlation analysis described above<br />

(Fig 7.6a), limiting the data to glioblastoma cases without IDH1 mutation. For<br />

the TCGA dataset, the correlation was decreased somewhat (Pearson R = 0.25<br />

compared to R = 0.36 for the full dataset) but still highly significant (p =<br />

6x10 5 ), demonstrating that the correlation with age is only partially explained<br />

by IDH1 status. This result was confirmed in the Gravendeel dataset, where<br />

the effect <strong>of</strong> controlling for IDH1 status and grade was negligible (R = 0.38<br />

compared to R = 0.39 for the full dataset including grade I-III samples).<br />

Among the individual signature genes, both HOXD10 and TUSC3 remained<br />

correlated with age in both datasets when limiting the analysis to IDH1 wild-<br />

type glioblastoma cases.<br />

186


7.5 Glioblastoma Pathway Analysis Results<br />

7.5 Glioblastoma Pathway Analysis<br />

In order to identify differentially expressed genes within pathways known to be<br />

perturbed in glioma and pathways related to the progenitor cell state, I man-<br />

ually built and curated an integrated pathway map by gathering information<br />

about interactions and interactors from the literature and the information data<br />

bases described in the Methods chapters. The glioblastoma pathways that al-<br />

ready exist in the literature, such as the KEGG "<strong>Glioma</strong>" pathway [3], are not<br />

as comprehensive as the present level <strong>of</strong> understanding <strong>of</strong> the disease requires<br />

<strong>of</strong> them. These existing pathways are shortcoming in four specific ways:<br />

1. they do not take into account the contribution from the stem cell com-<br />

ponent <strong>of</strong> the tumour and, thus, the progenitor cell state remains unrep-<br />

resented;<br />

2. core cell-cycle regulatory pathways that are very relevant in cancer, such<br />

as the MAPK cascade and the Integrin signal transduction network, are<br />

presented as zoomed out blocks in which the regulatory genes involved<br />

are omitted;<br />

3. they do not take into account the contribution from cancer-specific path-<br />

ways such as apoptosis, angiogenesis and invasion, or glioblastoma-specific<br />

phenotypes such as antigen processing and presentation and mesenchy-<br />

mal transformation;<br />

4. they are not available in formats other than poorly editable pdf/jpg/giff<br />

images, so they cannot be used to overlay custom expression data and<br />

highlight, in a very quick way, the relevant gene networks that are turned<br />

on/<strong>of</strong>f.<br />

In order to address these shortcomings and be able to visualise gene expression<br />

differences in a pathway context, I compiled our own glioblastoma integrated<br />

pathway map that tries to gather all the relevant information missing so far<br />

from the literature. Therefore, our map includes the pathways most commonly<br />

affected in glioblastoma: (i) RTK/PI3K/PTEN signaling, (ii) p53 signaling<br />

and (iii) Rb-mediated control <strong>of</strong> cell cycle progression [154,326], as well as core<br />

cell-cycle regulatory pathways: (i) MAPK cascade and (ii) RTK signaling, and<br />

finally cancer-specific and glioblastoma-specific pathways: (i) antigen process-<br />

ing and presentation, (ii) apoptosis, (iii) angiogenesis, invasion, motility and<br />

(iii) mesenchymal transformation [85].<br />

187


7.5 Glioblastoma Pathway Analysis Results<br />

Figure 7.7: The integrated glioblastoma pathway is subdivided into rough sections<br />

contained within the orange and blue boxes identifying the gene networks that participate<br />

in the pathway. In the orange boxes the "classic" glioblastoma pathways<br />

are highlighted: TP53, RB1 and PTEN signaling; in the blue boxes the cancer and<br />

glioblastoma-specific pathways are represented.<br />

188


7.5 Glioblastoma Pathway Analysis Results<br />

Figure 7.7 shows the integrated pathway in its default colours, without any<br />

overlaid expression data that would colour the gene nodes. Sections pertaining<br />

to different pathways are coloured in blue if the pathways are glioblastoma-<br />

specific or cancer-specific and orange if they reflect the cross-talk between the<br />

three well-known pathways commonly affected in glioblastoma. The colour,<br />

line and endpoints <strong>of</strong> the edges represent the type <strong>of</strong> interactions between the<br />

nodes: activation (solid, green, arrow point), transcriptional activation (solid,<br />

green, diamond point), tentative activation (dashed, green, arrow point), inhi-<br />

bition (solid, red, T point), transcriptional inhibition (solid, red, delta point),<br />

includes (solid, black, circle point), becomes (solid, black, top half arrow point),<br />

simple interaction (solid, grey, no endpoint), leading to (dashed, grey, arrow),<br />

lets in (dashed, grey, no endpoint). The shape <strong>of</strong> the node represents the type:<br />

gene (round), complex (hexagon), family (hexagon), molecule (fee), process<br />

(round rectangle). Finally, the beige colour distinguishes genes (grey if not<br />

overlaid with colour-coded expression data) from non genes (beige). All the<br />

interactions described by the pathway can be found in Appendix D.1.<br />

In order to ensure that every single interaction described in the pathway had<br />

been experimentally validated I checked every one <strong>of</strong> them in the interactions<br />

databases described in detail in the Methods section, such as BioGrid [62] and<br />

Intact [205]. Thus, this glioblastoma pathway is a representation <strong>of</strong> physically<br />

interacting proteins at work in the specific disease context. When the interac-<br />

tion needs to occur with chromatin to describe the regulation properly, such as<br />

in the case <strong>of</strong> a transcription factor, either a node named "DNA" is described<br />

as one <strong>of</strong> the two interactors, or a special edge named "activates transcription<br />

<strong>of</strong>" or "inhibits transcription <strong>of</strong>" - that can be identified by the diamond or<br />

delta endpoint, respectively - connects the two interactors. The entire pathway<br />

can be recreated from the list <strong>of</strong> interactions and interactors described in Ap-<br />

pendix D.1, but an editable version - with extension CYS - can be downloaded<br />

from www.ebi.ac.uk/~diva/GBM-pathway.cys, where the latest and previous<br />

versions <strong>of</strong> the pathway are available. The CYS extension allows the pathway<br />

to be viewed and/or edited with any network editing s<strong>of</strong>tware, such as Cy-<br />

toscape [451]. The availability <strong>of</strong> the pathway for download and use by other<br />

researchers, as well as its constant curation, attempts to address one <strong>of</strong> the<br />

four main shortcomings <strong>of</strong> already existing GBM pathways: the unavailability<br />

<strong>of</strong> formats other than images. Hopefully this resource will now be used by all<br />

the researchers who want to overlay expression data on a new comprehensive<br />

integrated GBM pathway to observe changes in the wider disease context.<br />

189


7.5 Glioblastoma Pathway Analysis Results<br />

Naturally, we wanted to make use <strong>of</strong> the resource ourselves and used it to<br />

visualise gene expression changes in the context <strong>of</strong> the molecular networks<br />

represented. The complete pathway has a total <strong>of</strong> 245 nodes, <strong>of</strong> which 57 are<br />

not genes and the remaining 188 are single genes. The nodes representing<br />

genes can be recognised by their distinct circular shapes that are unique to<br />

this category. Of the 57 non gene nodes: 22 are families <strong>of</strong> proteins, rep-<br />

resented by a diamond shape; eight are complexes <strong>of</strong> proteins, represented<br />

by a hexagon shape; six are non-proteic molecules, represented by a vee-like<br />

shape; the remaining 20 define end-processes such as "apoptosis", "cell signal-<br />

ing" and "protein synthesis" and are represented by round rectangular shapes.<br />

The only is<strong>of</strong>orms represented in the pathway are the two is<strong>of</strong>orms <strong>of</strong> gene<br />

CDKN2A labeled as CDKN2A and CDKN2A:ARF (Table 7.7). The dataset<br />

Table 7.7: Node assignment in the glioblastoma pathway. The colour <strong>of</strong> the nodes<br />

representing genes and is<strong>of</strong>orms is only grey at the default pathway level. When<br />

expression data are overlaid on the network the gene nodes will become coloured<br />

according to a customisable colour palette.<br />

Node type Number <strong>of</strong> nodes Node shape Node colour<br />

Gene families 22 Diamond Beige<br />

Cellular Processes 20 Round Rectangle Beige<br />

Non-proteic molecules 6 Vee Beige<br />

Complexes 8 Hexagon Beige<br />

Genes 186 Circle Grey, default<br />

Is<strong>of</strong>orms 2 Circle Grey, default<br />

Total 245<br />

<strong>of</strong> differentially expressed genes from the Tag-seq expression dataset contains<br />

739 genes at an FDR <strong>of</strong> 10%. By overlaying the expression data for the 739<br />

differentially expressed genes over the 188 gene nodes in the pathway, we found<br />

that 70 genes were differentially expressed between GNS and NS cell lines at<br />

10% FDR. Of these 70 genes, 51 were up-regulated in GNS cell lines (red colour<br />

gradient), and 19 were down-regulated in GNS cell lines (blue colour gradient)<br />

(Fig 7.8). The intensity <strong>of</strong> the colour reflects the value <strong>of</strong> the log2(FC) be-<br />

tween GNS and NS cell lines and the values defining the red and blue colour<br />

gradients were specifically taken from the same colour gradients defined in<br />

the qRT-PCR heatmap <strong>of</strong> figure 6.13 and the tumour expression correlation<br />

heatmap <strong>of</strong> figure 7.4.<br />

190


7.5 Glioblastoma Pathway Analysis Results<br />

Figure 7.8: Integrated GBM pathway with Tag-seq GNS dataset overlaid. The<br />

map is composed <strong>of</strong> 245 nodes, <strong>of</strong> which 188 nodes represent individual genes, shown<br />

as circles <strong>of</strong> intensities that correlate with FC expression changes.<br />

191


7.5 Glioblastoma Pathway Analysis Results<br />

Figure 7.9 displays a simplified and condensed version <strong>of</strong> the integrated GBM<br />

pathway <strong>of</strong> figure 7.8 that is focused on the pathways most frequently affected<br />

in glioblastoma, meant to highlight that we did capture some <strong>of</strong> the most com-<br />

mon alterations in glioblastoma with the GNS/NS comparison. Such changes<br />

include up-regulation <strong>of</strong> EGFR and down-regulation <strong>of</strong> the tumour suppres-<br />

sor PTEN [326]. Similarly to the complete pathway in figure 7.8, the colour<br />

gradient <strong>of</strong> the condensed pathway in figure 7.9 is taken from the same colour<br />

gradients defined in the qRT-PCR heatmap <strong>of</strong> figure 6.13 and the tumour<br />

expression correlation heatmap <strong>of</strong> figure 7.4.<br />

Figure 7.9: Affected p53, RB1 and PTEN/PI3K pathways. Genes are represented<br />

by circles coloured according to the expression FC measured between GNS and NS<br />

cell lines by Tag-seq.<br />

Given the high heterogeneity typical <strong>of</strong> glioblastoma tumours, we envisioned<br />

another application for our GBM integrated pathway. Since every cell line<br />

originated from a separate patient (for the exception <strong>of</strong> G144ED and G144)<br />

192


7.5 Glioblastoma Pathway Analysis Results<br />

and the heterogeneity <strong>of</strong> the tumour has recently been emphasised further by<br />

signature microarray studies like those <strong>of</strong> Verhaak et al [511] and Phillips et<br />

al [390], we mapped the Tag-seq measured expression levels for every GNS cell<br />

line on our GBM integrated pathway to try and visualise differences poten-<br />

tially unique to any GNS cell line and, therefore, to the tumour it originated<br />

from (single pathways in Appendix D.2). Figure 7.10 highlights the genes in<br />

our GBM pathway that are expressed in each individual GNS cell line, as mea-<br />

sured via Tag-seq. The green nodes reflect the genes that have been measured<br />

and the intensity <strong>of</strong> the green colour correlates with the level <strong>of</strong> expression ob-<br />

served, where lighter greens signify smaller expression values and darker greens<br />

signify higher expression values (normalised digital tag counts range from 0 to<br />

16,500) (Fig 7.10). For a clearer view <strong>of</strong> each <strong>of</strong> these pathways refer to Ap-<br />

pendix D.2 Pathway Images.<br />

By having the GNS cell line-specific GBM pathways next to each other in fig-<br />

ure 7.10, it is possible to observe differences as well as commonalities in gene<br />

expression patterns. For example, in each GNS cell line, tumour suppressor<br />

PTEN is not expressed (blue shaded boxes) and the antigen presentation-<br />

related genes light up with different intensities, highlighting a more highly<br />

activated antigen presentation process at the transcriptional level in cell line<br />

G144 than in cell lines G166 and G179 (red shaded boxes). Also, since the<br />

CDKN2A-CDKN2B tumour suppressor locus is deleted in cell line G179 but<br />

retained in cell lines G144 and G166 - possibly in response to an oncogenic<br />

signal - the expression <strong>of</strong> its CDKN2A, CDKN2A:ARF and CDKN2B genes is<br />

very low (darker green nodes) in the G179 pathway and much higher (lighter<br />

green nodes) in the G144 and G166 pathways (green shaded boxes). Finally,<br />

we can see that the orphan nuclear receptor NR0B1, which is known to be<br />

up-regulated and drive tumour growth in Ewing’s sarcoma [158], is also highly<br />

expressed (lighter green nodes) in our G179 pathway as opposed to the G144<br />

and G166 pathways (yellow shaded boxes). The PARP3 and PARP12 genes,<br />

located below NR0B1 in the pathways and included within the yellow shaded<br />

box <strong>of</strong> figure 7.10, are highly expressed across all GNS pathways, consistent<br />

with the potential therapeutic role <strong>of</strong> these genes in GNS cells as inhibitors <strong>of</strong><br />

their homolog PARP1, for which characteristic they are now being studied in<br />

brain tumour clinical trials [270]. In order to consider other sources <strong>of</strong> ex-<br />

pression data that could highlight interesting patterns when compared to our<br />

Tag-seq dataset, we overlaid the exon and microarray expression data from the<br />

TCGA and HGG datasets (described in Methods section 5.9; all genes overlaid<br />

193


7.5 Glioblastoma Pathway Analysis Results<br />

194<br />

Figure 7.10: Four integrated GBM pathways overlaid with Tag-seq expression level measures for each GNS cell line. Lighter green nodes indicate<br />

lower expression than darker green nodes. The blue box highlights the location <strong>of</strong> PTEN; the red box <strong>of</strong> the antigen presentation network <strong>of</strong> genes;<br />

the green box <strong>of</strong> the location <strong>of</strong> the CDKN2A-CDKN2B locus genes; the yellow box <strong>of</strong> the location <strong>of</strong> genes NR0B1, PARP3 and PARP12.


7.5 Glioblastoma Pathway Analysis Results<br />

195


7.5 Glioblastoma Pathway Analysis Results<br />

were found to be differentially expressed at FDR


7.5 Glioblastoma Pathway Analysis Results<br />

CDKN2A/CDKN2B locus deletion - observable in the TCGA pathway by the<br />

red-orange colours <strong>of</strong> nodes representing CDKNA genes (Fig 7.11). Amplifi-<br />

cation <strong>of</strong> the CDK4 locus was also a common finding in the TCGA dataset,<br />

CDK4 forming complexes with CDKN2A and CDKN2B to maintain cell cycle<br />

progression, which can be observed in the TCGA pathway as an orange node<br />

(Fig 7.11).<br />

Other interesting patterns that can be observed by the comparison <strong>of</strong> the three<br />

pathways in figure 7.13, are those highlighted with the other coloured boxes.<br />

The HIF1A gene, responsible for the regulation <strong>of</strong> poor-oxygen level responses<br />

in the cell, is up-regulated in the TCGA and HGG pathway but undetected in<br />

the GNS pathway (light blue shaded boxes). The SERPINB2 and SERPINE<br />

genes, responsible for the inhibition <strong>of</strong> angiogenesis and metastasis, seem to be<br />

strongly up-regulated in the GNS pathway but slightly down-regulated in the<br />

HGG pathway and undetected in the TCGA pathway (brown shaded boxes).<br />

The green shaded boxes highlight the regulatory loop for mesenchymal trans-<br />

formation that has been found to be active in GBM tumours. In this loop,<br />

CEBPB activates STAT3 and FOSL2 that together activate RUNX1 - also<br />

activated by DEC1 that is activated itself by FOSL2, which in turn activates<br />

JUN leading to mesenchymal transformation [85]. This regulatory loop is<br />

clearly active and up-regulated in the TCGA and HGG pathways, although<br />

only CEBPB appears as detected and up-regulated in the GNS pathway. The<br />

pink shaded boxes highlight the regulation <strong>of</strong> motility-relevant genes, such<br />

as those belonging to the SNARE complex, CALM1 and calcium channels<br />

<strong>of</strong> the CACN family, that appear to be strongly up-regulated in the GNS<br />

pathway but down-regulated in the TCGA and HGG pathways. Finally, the<br />

purple shaded boxes highlight the behaviour <strong>of</strong> the antigen processing and<br />

presentation-related genes such as HOXA, that are consistently up-regulated<br />

in all three pathways, although at different levels with the GNS pathway de-<br />

taining the highest levels <strong>of</strong> up-regulation. This confirms the importance <strong>of</strong> the<br />

immune-evasion phenotype that is transcriptionally active in all three datasets.<br />

Overall, this pathway approach allowed us to identify differentially expressed<br />

genes that participate in glioma-related pathways and highlight patterns be-<br />

tween different tumour cell types. However, it should be made clear that the<br />

GBM integrated pathway is not a flawless representation <strong>of</strong> the gene networks<br />

and regulations that occur inside GBM tumour cells, but rather the widest<br />

yet representation <strong>of</strong> the known interactions. Since the pathway was manually<br />

built there is always a chance to expand specific sub-networks <strong>of</strong> interest by<br />

197


7.5 Glioblastoma Pathway Analysis Results<br />

writing to the curator at dt322@cam.ac.uk or downloading the latest version<br />

<strong>of</strong> the pathway at www.ebi.ac.uk/~diva/GBM-pathway.cys and editing the<br />

pathway yourself. The manual curation <strong>of</strong> the pathway is nonetheless ongoing<br />

and new additions are released on a regular basis at the same address.<br />

198


7.5 Glioblastoma Pathway Analysis Results<br />

Figure 7.11: Integrated GBM pathway overlaid with TCGA dataset. The map<br />

is composed <strong>of</strong> 245 nodes and contains 188 nodes representing individual genes that<br />

correlate with FC expression changes (http://cancergenome.nih.gov; [326]).<br />

199


7.5 Glioblastoma Pathway Analysis Results<br />

Figure 7.12: Integrated GBM pathway overlaid with HGG dataset. The map<br />

composed <strong>of</strong> 245 nodes and contains 188 nodes representing individual genes that<br />

correlate with FC expression changes [148,390].<br />

200


7.5 Glioblastoma Pathway Analysis Results<br />

Figure 7.13: Three integrated GBM pathways overlaid with the GNS, TCGA<br />

and HGG datasets (FDR


Chapter 8<br />

MicroRNA Target Prediction<br />

Ensemble S<strong>of</strong>tware<br />

Contents<br />

8.1 Principles . . . . . . . . . . . . . . . . . . . . . . . . . . . . 202<br />

8.2 Workflows . . . . . . . . . . . . . . . . . . . . . . . . . . . 204<br />

8.3 Databases . . . . . . . . . . . . . . . . . . . . . . . . . . . . 205<br />

8.4 Filters . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 209<br />

8.5 Target Prediction Ensemble Analysis . . . . . . . . . . . . 211<br />

8.1 Principles<br />

Hundreds <strong>of</strong> microRNA sequences are now annotated throughout the human<br />

genome and are predicted to modulate the expression levels <strong>of</strong> thousands <strong>of</strong><br />

mRNAs. Families <strong>of</strong> microRNA sequences can be identified through evolution-<br />

ary conservation <strong>of</strong> sequence patterns in related species, and reciprocally, genes<br />

targeted by microRNAs are under selection pressure to retain the recognition<br />

sites needed for the annealing <strong>of</strong> microRNA to mRNA duplexes. Numerous<br />

regulatory target prediction algorithms have been developed to exploit these<br />

properties, but they vary widely in terms <strong>of</strong> criteria, accuracy and prediction<br />

coverage. Most prediction methods search for complementary sequences be-<br />

tween microRNAs and putative gene targets, while some consider physical and<br />

statistical hybridization properties, cross-conservation <strong>of</strong> regulatory RNAs be-<br />

tween related species, etc. As a result, there is very little overlap between<br />

the microRNA:mRNA annealing predicted by different algorithms. It is <strong>of</strong>-<br />

ten the case that researchers have asked themselves "which target prediction<br />

algorithm will predict the highest number <strong>of</strong> genes in my list?". Needless to<br />

202


8.1 Principles Results<br />

say, this is a flawed approach triggered by the absence <strong>of</strong> a unifying common<br />

theme in the prediction algorithms that would make them more accurate in<br />

their predictions. Rationally, if several algorithms can be developed that yield<br />

vastly different results, then we are missing an important variable in our equa-<br />

tion. In this perspective, the existing prediction algorithms are not capable <strong>of</strong><br />

simulating accurately what is happening in Nature time after time in the same<br />

reproducible way. Having said this, we were interested in finding out if a single<br />

prediction algorithm or a superset <strong>of</strong> several prediction algorithms was best at<br />

predicting microRNA to mRNA annealing. During this analysis we stumbled<br />

upon an observation that might be a hint towards the identity <strong>of</strong> the missing<br />

variable in the prediction algorithm equation.<br />

In order to determine which set <strong>of</strong> prediction algorithms were best at predict-<br />

ing microRNA:mRNA annealing, we needed experimentally validated data to<br />

help us screen the positives from the false positives, which contribute greatly<br />

to the pool <strong>of</strong> predictions. We used exon array data and microRNA array<br />

data that was available to us from our GNS cell lines (Appendix E and F) to<br />

validate our findings. Since a large number <strong>of</strong> prediction algorithms are com-<br />

monly used in research and they each predict a vast number <strong>of</strong> interactions,<br />

we built a tool called "GenemiR" to allow molecular biologists to generally<br />

access and manage microRNA predictions on a large scale and across differ-<br />

ent algorithms that would also help us address our research question. The<br />

GenemiR s<strong>of</strong>tware exists as a command line binary executable for Unix-based<br />

systems or as a compiled local installation for Windows. Due to its length,<br />

the code <strong>of</strong> the binary executable could not be added to the appendix <strong>of</strong> this<br />

thesis, but it is available for anyone to view or download at www.ebi.ac.<br />

uk/~diva/GenemiR/genemir-code.txt. The executable for Windows can be<br />

downloaded at www.ebi.ac.uk/~diva/GenemiR/GenemiR.zip.<br />

In order to address the need for unified search and presentation <strong>of</strong> microRNA<br />

to mRNA target interactions predicted across multiple algorithms, GenemiR<br />

relates human and mouse microRNAs with their predicted target genes as<br />

reported by eight leading predictors. At the core <strong>of</strong> the program are two<br />

primitives that allow it to be extremely fast in extrapolating lists <strong>of</strong> gene tar-<br />

gets given a microRNA and, performing the reciprocal analysis, extrapolating<br />

lists <strong>of</strong> microRNAs predicted to repress one or more genes <strong>of</strong> interest (Fig 8.1).<br />

Many other auxiliary functions and filters allow the expert user to parametrize<br />

203


8.2 Workflows Results<br />

and optimise the analysis to make it as stringent or as loose as necessary. The<br />

target predictions in GenemiR’s database can be queried through a variety <strong>of</strong><br />

selectable filters, allowing the user to include or exclude any combination <strong>of</strong><br />

algorithms, group multiple microRNAs into the same query, and retrieve vari-<br />

able subsets <strong>of</strong> targeted genes (Fig 8.1). Finally, GenemiR allows the matching<br />

<strong>of</strong> prediction data to any gene expression dataset <strong>of</strong> interest, and provides a fa-<br />

cility for loading external datasets determined by the user. Graphical plotting<br />

functions display target expression levels relative to the conditions <strong>of</strong> the ex-<br />

periment, such as tissue-specific transcriptional pr<strong>of</strong>iling or time course series.<br />

The s<strong>of</strong>tware itself does not contain internal experimental data, but simply<br />

aggregates the output <strong>of</strong> prediction algorithms in a manner suitable to explo-<br />

ration and hypothesis testing, reducing the complexity <strong>of</strong> this approach by<br />

integrating microRNA and gene annotations, genome-wide expression data,<br />

and regulatory target predictions in a common analysis framework (Fig 8.1).<br />

8.2 Workflows<br />

The GenemiR s<strong>of</strong>tware allows for the discovery <strong>of</strong> systemic or tissue-specific<br />

patterns that may be hidden in the microRNA targeting prediction data from<br />

eight leading algorithms. In order to do this, two flows <strong>of</strong> information are estab-<br />

lished that address reciprocal biological questions: "which genes are targeted<br />

by one or more specific microRNAs?" and "which microRNAs are predicted<br />

to target a given set <strong>of</strong> genes?". These issues are intimately related within<br />

the context <strong>of</strong> the same biological system, but are organised as two opposite<br />

flows <strong>of</strong> information in terms <strong>of</strong> program operation and layout (Fig 8.2). De-<br />

pending on the type <strong>of</strong> query executed against the internal databases, a list<br />

<strong>of</strong> genes predicted to be targeted by a number <strong>of</strong> microRNAs according to a<br />

customer-defined selection <strong>of</strong> prediction algorithms, is generated (Workflow 1,<br />

figure 8.3). In the opposite workflow, a list <strong>of</strong> microRNAs is generated start-<br />

ing from a list <strong>of</strong> gene symbols or Genbank, RefSeq, EMBL, ENSG or ENST<br />

identifiers, according to the customer-defined selection <strong>of</strong> prediction algorithms<br />

(Workflow 2, figure 8.3). Conversion from the identifiers contained originally<br />

in the prediction files is constantly active in both workflows to always translate<br />

the queries to a human intelligible list <strong>of</strong> gene symbols.<br />

204


8.3 Databases Results<br />

Figure 8.1: Overview <strong>of</strong> GenemiR. (a) Queries based on various data types (the<br />

input section: microRNAs, expression data, genes, and identifiers) are executed<br />

against internal or external databases to generate other data types (the output section:<br />

genes, microRNAs, graphs and converted identifiers). The shaded boxes distinguish<br />

the core primitive functions from the auxiliary ones. (b) Relationships between<br />

databases used by GenemiR and the functions that operate on them. Solid lines depict<br />

databases <strong>of</strong> fixed size over the lifetime <strong>of</strong> the program’s execution, whereas the<br />

dashed lines indicate a dataset <strong>of</strong> variable dimensions according to the files that are<br />

inputted by the user.<br />

8.3 Databases<br />

All microRNAs and genes that could constitute a query are stored in the form<br />

<strong>of</strong> three internal databases:<br />

1. Identifiers for all known human and mouse microRNAs;<br />

2. Genbank, RefSeq [409], EMBL [252], ENSG, ENST [147] identifiers for<br />

all annotated human and mouse genes;<br />

205


8.3 Databases Results<br />

Figure 8.2: Internal organisation <strong>of</strong> the target prediction database <strong>of</strong> GenemiR.<br />

From top to bottom, the two conceptual workflows are consecutively displayed.<br />

Each <strong>of</strong> eight algorithms (circles) is represented by a different acronym:<br />

Diana-microT=DN-T; miRanda=MR; miRBase=MC;ElMMo=EM; PITA=PA; Pic-<br />

Tar=PT5/6; TargetscanS=TSS. MicroRNAs (diamonds) and target genes (squares)<br />

are assigned random labels for illustrative purposes.<br />

3. Gene symbols as determined by the HUGO Gene Nomenclature Commit-<br />

tee for [203] and the MGI Mouse Genome Informatics database resource<br />

for mouse [333].<br />

All expression datasets that the user wants to upload in the s<strong>of</strong>tware are re-<br />

ferred to as "external databases" because they are not part <strong>of</strong> the internal<br />

databases, they are user-defined and variable in size. Gene annotation data<br />

are derived from the Ensembl database [147], and a utility script is provided<br />

with the program to allow automatic updating <strong>of</strong> files, via the Ensembl Perl<br />

API, to reflect the latest annotation release <strong>of</strong> the human and mouse genomes.<br />

The internal databases <strong>of</strong> human and mouse microRNAs and gene identifiers<br />

for all annotated human and mouse genes are linked in a meaningful way by<br />

the algorithms that predict which <strong>of</strong> the microRNAs regulate which <strong>of</strong> the<br />

gens and, vice versa, which <strong>of</strong> the genes are regulated by which microRNAs.<br />

Thus, these linked databases consists <strong>of</strong> the union <strong>of</strong> prediction results gen-<br />

erated from the eight most widely used algorithms (considering PicTar 5-way<br />

and PicTar 6-way as separate algorithms) (Table 8.1), with the possibility <strong>of</strong><br />

increasing this number as new algorithms are produced with new sets <strong>of</strong> pre-<br />

dictions, thanks to the modular internal structure with which GenemiR was<br />

designed:<br />

1. PicTar [127,247] identifies putative microRNA targets in vertebrates, C.<br />

elegans and Drosophila, using the principle <strong>of</strong> cross-conservation. The<br />

two versions, PicTar 5-way and PicTar 6-way, refer to the number <strong>of</strong><br />

species in the comparison: 5-way includes human, chimp mouse, rat<br />

and dog genomes, and 6-way adds the chicken genome. PicTar employs<br />

206


8.3 Databases Results<br />

Figure 8.3: Workflows at the core <strong>of</strong> the primitive functions <strong>of</strong> the GenemiR s<strong>of</strong>tware.<br />

The first workflow answers the question: "which microRNAs target this list<br />

<strong>of</strong> genes?" and returns a list <strong>of</strong> microRNAs filtered according to the Minimum or<br />

Cumulative filter or both. The second workflow answers the question: "which genes<br />

are targeted by this list <strong>of</strong> microRNAs?" and returns a list <strong>of</strong> genes filtered according<br />

to the settings <strong>of</strong> the Minimum or Cumulative filter or both.<br />

pair-wise alignments to filter seed sequences conserved in 7 species <strong>of</strong><br />

Drosophila and 8 species <strong>of</strong> vertebrates. In addition to sequence conser-<br />

vation, PicTar considers secondary evidence, such as co-expression and<br />

clustering <strong>of</strong> microRNAs, as well as target identification based on onto-<br />

logical parameters, such as expression in the common cell types or devel-<br />

207


8.3 Databases Results<br />

opmental stages. Importantly, it also takes into account the number <strong>of</strong><br />

multiple seeds occurring within the same 3'UTR. The false-positive rate<br />

for this algorithm is estimated to be approximately 30% [48].<br />

2. TargetScanS is the second version <strong>of</strong> the mammalian microRNA target<br />

prediction algorithm developed by Lewis and colleagues following the<br />

publication <strong>of</strong> their original TargetScan approach [272,273]. The earlier<br />

program searched for a 7nt-long seed region starting from the second<br />

nucleotide <strong>of</strong> the 5' end miRNA sequence, calculated the free energy <strong>of</strong><br />

hybridization using RNAFold [57], and computed a score based on the<br />

presence <strong>of</strong> multiple seeds capable <strong>of</strong> annealing to the same microRNA.<br />

The criteria used by TargetScanS are more specific, requiring a 6nt-long<br />

seed sequence preceded by an adenosine, positioned in a conserved region<br />

that, in turn, is surrounded by areas <strong>of</strong> lesser conservation. TargetScanS<br />

omits the use <strong>of</strong> free-energy calculations to predict hybridization affinity,<br />

and is estimated to have a false-positive rate <strong>of</strong> between 22 and 31% [48].<br />

3. DIANA-microT [237,316] version 3.0 recognises only single seed se-<br />

quences having a central stem-loop secondary structure, instead <strong>of</strong> select-<br />

ing for near-perfect complementarity between the 5' seed region <strong>of</strong> the<br />

microRNA and the 3'UTR <strong>of</strong> the target message. In addition to search-<br />

ing for the classical annealing pattern in the 5' seed region, DIANA-<br />

microT also requires complementarity at the 3' end <strong>of</strong> the microRNA<br />

sequence [48].<br />

4. miRanda [134,213] identifies putative targets throughout the human and<br />

Drosophila genomes. It implements a dynamic programming approach<br />

to compute local alignments <strong>of</strong> complementary microRNA and mRNA<br />

sequences. The algorithm assigns scores to putative seed sequences using<br />

a position matrix, giving a higher weight to those closer to the 5' end<br />

<strong>of</strong> the microRNA sequence. However, it is not constrained by the re-<br />

quirement for exact seed matches. The program takes into consideration<br />

free-energy annealing calculations (via RNAfold), and seed-sequence con-<br />

servation. To account for tandem annealing <strong>of</strong> microRNAs to multiple<br />

consensus sequences in the targeted transcript, miRanda assigns a high<br />

score to alignment matches to the same as well as different microRNAs.<br />

5. ElMMo [155] uses a general Bayesian method that scores the conser-<br />

vation <strong>of</strong> microRNA binding sites according to an evolutionary model.<br />

208


8.4 Filters Results<br />

Bayesian methods are based on prior probability <strong>of</strong> an observation and<br />

are updated as new data keeps entering. This model assumes a phyloge-<br />

netic relationship among several species.<br />

6. miRBase [179] uses the miRanda algorithm to identify potential binding<br />

sites <strong>of</strong> a microRNA. Dynamic programming alignment is used to identify<br />

highly complementary sites that also require strict complementarity at<br />

the 5' seed region and thermodynamic stability, which is estimated for<br />

each target site. For inclusion in the database, conservation <strong>of</strong> the target<br />

site at the exact same position in at least two species is needed.<br />

7. PITA [224] takes into consideration the strength <strong>of</strong> microRNA repression<br />

given target site accessibility and for each target site, an energy-based<br />

measure representing the difference between the free energy gained by<br />

the binding <strong>of</strong> the microRNA to the target, and the free energy lost<br />

by un-pairing the nucleotides within the target site, is calculated. The<br />

energy used to un-pair additional nucleotides flanking the target sites is<br />

also taken into account.<br />

Table 8.1: microRNA target prediction algorithms used by GenemiR with number<br />

<strong>of</strong> microRNA:3'UTR interactions predicted. The original target identifiers refer to<br />

the identifiers used by a prediction algorithm to identify the targeted genes. The<br />

final target identifiers refer to the identifiers that are returned by any query <strong>of</strong> any<br />

prediction algorithm database.<br />

Prediction algorithm Number<br />

microRNAs<br />

Number <strong>of</strong><br />

targets<br />

Original target<br />

identifiers<br />

Final target<br />

identifiers<br />

PITA 678 22,974 RefSeq HGNC/MGI<br />

PicTar 5-way 178 9,334 RefSeq HGNC/MGI<br />

PicTar 6-way 130 3,585 RefSeq HGNC/MGI<br />

DIANA-microT 555 18,986 ENSG/ENSMUSG HGNC/MGI<br />

Elmmo 1206 31,303 RefSeq,EMBL HGNC/MGI<br />

Miranda 1100 32,641 EMBL HGNC/MGI<br />

Microcosm 694 34,507 ENST/ENSMUST HGNC/MGI<br />

TargetscanS 967 17,725 HGNC/MGI HGNC/MGI<br />

8.4 Filters<br />

Critical to the successful outcome <strong>of</strong> user queries is the appropriate use <strong>of</strong> the<br />

built-in filtering functions. The purpose <strong>of</strong> the filter auxiliary functions (Fig<br />

209


8.4 Filters Results<br />

8.3) is to allow the user to specify an appropriate range <strong>of</strong> return results from<br />

each query, depending on either the numbers <strong>of</strong> different microRNAs expected<br />

to play a role in gene regulation (adjusted according to the Minimum and<br />

Cumulative filters), or the level <strong>of</strong> concordance the user enforces between the<br />

combination <strong>of</strong> prediction algorithms defined. The user can at all times pref-<br />

erentially select a desired subset <strong>of</strong> algorithms upon which prediction results<br />

will be based and the two types <strong>of</strong> filters available - Minimum and Cumulative<br />

- will be applied on the prediction results <strong>of</strong> those algorithms.<br />

The consequence <strong>of</strong> setting the Minimum filter to a given value depends on<br />

the type <strong>of</strong> query he user makes. If the query starts with a list <strong>of</strong> microRNAs<br />

and therefore asks which genes are target by these microRNAs, according to a<br />

number n <strong>of</strong> prediction algorithms, then setting the Minimum filter to a given<br />

value returns a set <strong>of</strong> genes targeted by at least that number <strong>of</strong> microRNAs,<br />

as reported by all <strong>of</strong> the selected algorithms. For example, if the inputted list<br />

contains m = 10 microRNAs and n = 3 prediction algorithms are selected,<br />

then the Minimum filter will range from one to 10 and setting it on four will<br />

cause it to report only those genes that are targeted by at least eight microR-<br />

NAs as predicted by each <strong>of</strong> the three algorithms alone. If the query were to<br />

start from a list containing g = 30 genes and n = 3 prediction algorithms were<br />

selected, then the Minimum filter would range from one to 30 and setting it<br />

on 24 would cause it to report only those microRNAs that target 24 genes as<br />

predicted by each <strong>of</strong> the prediction algorithms alone (Fig 8.3). The number <strong>of</strong><br />

genes returned from the query will therefore depend greatly on the prediction<br />

algorithms selected.<br />

The Cumulative filter acts in an alternative fashion: a gene is reported if<br />

the total number <strong>of</strong> microRNA predictions, according to all <strong>of</strong> the chosen algo-<br />

rithms, is at least equal to or greater than the minimum value set by the user.<br />

This alleviates the requirement that every algorithm independently predict a<br />

set number <strong>of</strong> microRNA interactions; rather, the aggregate sum <strong>of</strong> all results<br />

in determining which target genes to report, will be considered. Using the<br />

Cumulative filter is a less stringent approach and, all things equal, returns a<br />

higher number <strong>of</strong> outputted results. For example, if the inputted list contains<br />

m = 10 microRNAs and n = 3 prediction algorithms are selected, then the Cu-<br />

mulative filter will range from one to 10x3 = 30 and setting it on 20 will cause<br />

it to report the genes that are targeted by at least 20 microRNAs according<br />

to any combination <strong>of</strong> prediction algorithms selected, without the requirement<br />

that any particular algorithm predict a set threshold <strong>of</strong> microRNAs. If the<br />

210


8.5 Target Prediction Ensemble Analysis Results<br />

query were to start from a list containing g = 30 genes and n = 3 prediction<br />

algorithms were selected, then the Cumulative filter would range from one to<br />

30x3 = 90 and setting it on 56 would cause it to report only those microRNAs<br />

that target 56 genes as predicted by any combination <strong>of</strong> the prediction algo-<br />

rithms (Fig 8.3).<br />

While the Minimum and Cumulative filters operate on microRNAs, the possi-<br />

bility <strong>of</strong> selecting a subset <strong>of</strong> the eight prediction algorithms affords the user<br />

control over the number <strong>of</strong> algorithms required to report a given prediction,<br />

and thus define the criteria by which predictions are deemed accurate. For<br />

example, certain applications - such as experimental validation and cloning <strong>of</strong><br />

microRNAs involved in particular pathways <strong>of</strong> interest - involve a significant<br />

amount <strong>of</strong> effort to perform. If this is the ultimate goal <strong>of</strong> the investigation,<br />

then it is desirable to focus on a small set <strong>of</strong> highly-scoring microRNA targets<br />

unanimously reported by several algorithms. Other searches though, such as<br />

those performed for comparative genomic analyses, are limited only by com-<br />

putational feasibility and can therefore afford to include a greater number <strong>of</strong><br />

putative regulators whose involvement may only be predicted by one or two<br />

algorithms.<br />

Any combination <strong>of</strong> the above filtering methods is possible, keeping in mind<br />

that: (i) they are always applied to the original set <strong>of</strong> data, and that (ii) a<br />

given gene is included in the final results only if it passes the criteria imposed<br />

by each individual filter set by the user. Therefore, the utility <strong>of</strong> applying dif-<br />

ferent filter combinations depends on the relevance <strong>of</strong> the biological question<br />

they help to answer.<br />

8.5 Target Prediction Ensemble Analysis<br />

A question we wanted to ask was whether any prediction algorithm fared bet-<br />

ter than any other combination <strong>of</strong> prediction algorithms. Since the divergence<br />

between prediction results from any algorithm is large, finding whether any<br />

combination <strong>of</strong> a subset <strong>of</strong> these algorithms fares better than any algorithm<br />

alone, is a necessary step. Using our s<strong>of</strong>tware tool GenemiR we tried to eluci-<br />

date how different combinations <strong>of</strong> prediction algorithms fared with respect to<br />

the single algorithm, and thus address the hypothesis suggested by Alexiou et<br />

al [16] that combinations <strong>of</strong> algorithms predict more accurately than the single<br />

algorithm alone. The way we addressed this problem was using relevant ex-<br />

perimental data from exon arrays and microRNA microarrays generated from<br />

211


8.5 Target Prediction Ensemble Analysis Results<br />

the same GNS cell lines <strong>of</strong> which we had digital gene expression pr<strong>of</strong>iles. With<br />

this set <strong>of</strong> data we could link the down-regulation <strong>of</strong> expression observed for<br />

the 2,290 genes at F DR < 1% as measured in the exon arrays, to the up-<br />

regulation <strong>of</strong> expression observed for the 258 microRNAs at F DR < 1% as<br />

measured in the microRNA microarrays.<br />

Our first interesting finding was that the final score <strong>of</strong> our analysis, represent-<br />

ing how well the subset <strong>of</strong> algorithms at stake fared when asked to predict<br />

which microRNAs targeted the list <strong>of</strong> 2,290 down-regulated genes, worsened<br />

considerably if an initial filter was not applied that made our analysis tissue-<br />

specific. When I mentioned earlier that the ensemble analysis gave us a hint as<br />

to the identity <strong>of</strong> the variable missing from prediction algorithms, it was tissue<br />

specificity. So far prediction algorithms have not been designed with tissue-<br />

specificity in mind. However, microRNA regulation is highly tissue specific<br />

and there is no way around having to factor this in when designing an algo-<br />

rithm that wil accurately predict the interaction between a microRNA and an<br />

mRNA. Current prediction algorithms consider different cohorts <strong>of</strong> variables<br />

that all refer to a main concept: sequence complementarity. Secondary con-<br />

siderations are factors such as secondary structure and number <strong>of</strong> microRNA<br />

seeds in a 3'UTR. There is no easy way to implement tissue-specificity variables<br />

within a target prediction algorithm because <strong>of</strong> the empirical nature <strong>of</strong> such<br />

variables. We have yet to understand what exactly triggers tissue-specificity in<br />

microRNA regulation <strong>of</strong> mRNAs, but any tuning implemented by epigenetic<br />

mechanisms or feedback regulatory loops may be too computationally inten-<br />

sive to simulate still.<br />

One way to work around this problem in the present is to filter the target<br />

predictions by a background gene list that filters out most non tissue-specific<br />

predictions. Of course, this strategy will only yield partially accurate results<br />

but it can surely improve the use <strong>of</strong> prediction results at the present time. As a<br />

result <strong>of</strong> this understanding, we used a background gene list ourselves in order<br />

to minimise the number <strong>of</strong> false positives caused by the brain tissue-specificity<br />

<strong>of</strong> the query. The background gene list was retrieved from the exon array<br />

dataset and consisted <strong>of</strong> all 6,254 genes with an expression level that was mea-<br />

sured at F DR < 1%. Running the algorithm without this background gene<br />

list worsened the final score by ten fold.<br />

The method we used to evaluate the performance and accuracy <strong>of</strong> each pre-<br />

diction algorithm in the GenemiR internal database is described below and<br />

212


8.5 Target Prediction Ensemble Analysis Results<br />

summarised in figure 8.4. We first run the method on the predictions from<br />

each algorithm alone and then extended it to all combinations <strong>of</strong> prediction<br />

algorithm results. The score E reflecting the accuracy <strong>of</strong> each prediction or<br />

combination <strong>of</strong> predictions, ranges between 0 and 1 (0 > E > 1), with 1 re-<br />

flecting a perfectly matching set <strong>of</strong> predictions between the algorithm being<br />

evaluated and our experimental data. The score is calculated following these<br />

successive steps:<br />

1. filter out, for each prediction algorithm and for each microRNA exper-<br />

imentally observed to be up-regulated, the genes that are predicted to<br />

be target by that microRNA but are not present in the gene background<br />

list;<br />

2. generate a union list from the genes filtered in step 1 for the entire cohort<br />

<strong>of</strong> up-regulated microRNAs, for each prediction algorithm;<br />

3. iterate over each gene in the union list and increase a cumulative counter<br />

every time the gene is predicted to be targeted by one <strong>of</strong> the up-regulated<br />

microRNAs, for each prediction algorithm. This counter is normalised<br />

to the number <strong>of</strong> microRNAs that the prediction contains from the list<br />

<strong>of</strong> 258 experimentally measured microRNAs;<br />

4. sort the union gene list by the count calculated in step 3 to generate a<br />

"hit" list, representative <strong>of</strong> each prediction algorithm;<br />

5. iterate over the hit list summing in a cumulative counter C1, the counts<br />

associated to all the genes in the list, which have been predicted by the<br />

prediction algorithm ;<br />

6. iterate over the hit list summing in a cumulative counter C2, the counts<br />

associated to the genes that are experimentally observed to be down-<br />

regulated in our exon array data;<br />

7. generate a score value by dividing the cumulative counters C1 and C2,<br />

representing a measure <strong>of</strong> how well a prediction algorithm fared based<br />

on how many genes it correctly predicted to be targeted in our GNS cell<br />

lines, considering the same cell line expression pr<strong>of</strong>ile as a background<br />

for calculations.<br />

Scores for each single prediction algorithm are listed in table 8.2 below. To<br />

evaluate whether combinations <strong>of</strong> different algorithms were more accurate at<br />

213


8.5 Target Prediction Ensemble Analysis Results<br />

Figure 8.4: Step by step diagram <strong>of</strong> the ensemble method adopted to find the<br />

score E (=C2/C1) <strong>of</strong> prediction accuracy for prediction algorithms. The method<br />

was applied to the predictions from all combinations <strong>of</strong> target prediction algorithms<br />

and an E-score was generated for each one. The red set <strong>of</strong> gene predictions varied<br />

in size depending on how many and which algorithms were being considered for a<br />

particular round.<br />

predicting than the single, we performed the same steps 1-7 but with lists that<br />

resulted as the union <strong>of</strong> all the user-defined prediction algorithm genes (vary-<br />

ing size <strong>of</strong> the predicted genes set <strong>of</strong> figure 8.4). The results are listed in table<br />

8.5. The purpose <strong>of</strong> the hit list is to "weight" the importance to the predicted<br />

genes. For example, if gene A has been predicted to be targeted by only nine<br />

<strong>of</strong> the 258 microRNAs, its weight when added to the cumulative score C1 will<br />

be only 9/258th the weight added by gene B, if gene B were predicted to be<br />

214


8.5 Target Prediction Ensemble Analysis Results<br />

Table 8.2: Single prediction algorithm ensemble analysis results. Displayed in<br />

descending order <strong>of</strong> E-score.<br />

Prediction name E-score<br />

ElMMo 0,3181<br />

Diana-microT 0,3122<br />

PITA 0,3088<br />

TargetscanS 0,3073<br />

PicTar 6 0,3020<br />

miRBase 0,3008<br />

PicTar 5 0,2989<br />

miRanda 0,2979<br />

targeted by all 258 microRNAs. This scoring system takes into consideration<br />

the possibility that not all prediction algorithms necessarily predict the target-<br />

ing for all the 258 up-regulated microRNAs, by always measuring the accuracy<br />

<strong>of</strong> a prediction set over the number <strong>of</strong> genes <strong>of</strong> the background gene list that<br />

are predicted by a particular algorithm.<br />

This analysis highlights the fact that very little improvement, in the order <strong>of</strong><br />

the thousandth, is achieved by combining prediction algorithms together. The<br />

highest scoring prediction algorithms as-singles are ElMMo and Diana-microT<br />

and when used in combination with other prediction algorithms the E-score<br />

<strong>of</strong> ElMMo always decreases, while the E-score <strong>of</strong> Diana-microT, in combina-<br />

tion with one or two other prediction algorithms, seems to slightly increase.<br />

This analysis reveals that the best performing combination <strong>of</strong> algorithms does<br />

not outperform the best scoring as-single algorithm ElMMo. However, this<br />

initial analysis should be followed by a series <strong>of</strong> other analysis in which other<br />

approaches for the evaluation <strong>of</strong> the score are taken, and the Minimum and<br />

Cumulative filters are taken into consideration as well. This approach would<br />

increase the number <strong>of</strong> combinations from the current 256 (2 8 ) because each<br />

combination <strong>of</strong> filters would have to be evaluated for all the 256 microRNAs<br />

and would therefore require a global energy combinatorial optimisation algo-<br />

rithm such as a genetic algorithm, whereby a search that mimics the process<br />

<strong>of</strong> natural evolution would be performed on a population <strong>of</strong> candidate solu-<br />

tions that evolve towards the best solution starting from a randomly selected<br />

population.<br />

215


8.5 Target Prediction Ensemble Analysis Results<br />

Table 8.3: All combinations <strong>of</strong> prediction algorithms in descending order <strong>of</strong> E-score. The Escores<br />

for the single algorithms are also present and highlighted along the list in bold characters.<br />

Prediction algorithms involved in the combination set E-score<br />

ElMMo 0.3181<br />

ElMMo PicTar6 0.3173<br />

ElMMo PicTar5 0.3162<br />

ElMMo miRBase 0.3160<br />

Diana-microT ElMMo 0.3159<br />

Diana-microT ElMMo PicTar6 0.3155<br />

ElMMo PicTar5 PicTar6 0.3155<br />

ElMMo miRBase PicTar6 0.3154<br />

Diana-microT ElMMo PicTar5 0.3148<br />

Diana-microT ElMMo miRBase 0.3147<br />

ElMMo miRBase PicTar5 0.3145<br />

Diana-microT ElMMo PicTar5 PicTar6 0.3144<br />

ElMMo TargetscanS 0.3144<br />

Diana-microT ElMMo miRBase PicTar6 0.3143<br />

ElMMo miRBase PicTar5 PicTar6 0.3139<br />

ElMMo PicTar6 TargetscanS 0.3139<br />

Diana-microT ElMMo TargetscanS 0.3138<br />

Diana-microT ElMMo miRBase PicTar5 0.3137<br />

Diana-microT ElMMo PicTar6 TargetscanS 0.3135<br />

Diana-microT ElMMo miRBase PicTar5 PicTar6 0.3134<br />

ElMMo PicTar5 TargetscanS 0.3133<br />

ElMMo miRBase TargetscanS 0.3132<br />

Diana-microT ElMMo miRBase TargetscanS 0.3130<br />

Diana-microT ElMMo PicTar5 TargetscanS 0.3130<br />

ElMMo miRBase PicTar6 TargetscanS 0.3129<br />

ElMMo PicTar5 PicTar6 TargetscanS 0.3129<br />

Diana-microT ElMMo miRBase PicTar6 TargetscanS 0.3127<br />

Diana-microT ElMMo PicTar5 PicTar6 TargetscanS 0.3127<br />

ElMMo miRBase PicTar5 TargetscanS 0.3123<br />

Diana-microT ElMMo miRBase PicTar5 TargetscanS 0.3123<br />

Diana-microT 0.3122<br />

ElMMo PITA 0.3122<br />

Diana-microT ElMMo PITA 0.3122<br />

Diana-microT ElMMo miRBase PicTar5 PicTar6 TargetscanS 0.3121<br />

ElMMo PITA PicTar6 0.3120<br />

Diana-microT ElMMo PITA PicTar6 0.3120<br />

ElMMo miRBase PicTar5 PicTar6 TargetscanS 0.3120<br />

Diana-microT ElMMo miRBase PITA 0.3118<br />

Diana-microT ElMMo PITA PicTar5 0.3118<br />

ElMMo miRBase PITA 0.3117<br />

ElMMo PITA PicTar5 0.3117<br />

Diana-microT ElMMo miRBase PITA PicTar6 0.3116<br />

Diana-microT ElMMo PITA PicTar5 PicTar6 0.3116<br />

ElMMo miRBase PITA PicTar6 0.3115<br />

ElMMo PITA PicTar5 PicTar6 0.3115<br />

Diana-microT ElMMo PITA TargetscanS 0.3115<br />

ElMMo PITA TargetscanS 0.3114<br />

Diana-microT ElMMo PITA PicTar6 TargetscanS 0.3114<br />

Diana-microT ElMMo miRBase PITA PicTar5 0.3113<br />

Diana-microT PicTar6 0.3113<br />

ElMMo PITA PicTar6 TargetscanS 0.3113<br />

ElMMo miRBase PITA PicTar5 0.3112<br />

Diana-microT ElMMo miRBase PITA PicTar5 PicTar6 0.3112<br />

Diana-microT ElMMo miRBase PITA TargetscanS 0.3112<br />

Diana-microT ElMMo PITA PicTar5 TargetscanS 0.3112<br />

ElMMo miRBase PITA PicTar5 PicTar6 0.3110<br />

ElMMo miRBase PITA TargetscanS 0.3110<br />

ElMMo PITA PicTar5 TargetscanS 0.3110<br />

Diana-microT ElMMo miRBase PITA PicTar6 TargetscanS 0.3110<br />

Diana-microT ElMMo PITA PicTar5 PicTar6 TargetscanS 0.3110<br />

ElMMo PITA PicTar5 PicTar6 TargetscanS 0.3109<br />

Diana-microT ElMMo miRBase PITA PicTar5 TargetscanS 0.3108<br />

ElMMo miRBase PITA PicTar6 TargetscanS 0.3108<br />

Diana-microT ElMMo miRBase PITA PicTar5 PicTar6 TargetscanS 0.3107<br />

Diana-microT ElMMo miRanda 0.3106<br />

ElMMo miRBase PITA PicTar5 TargetscanS 0.3106<br />

ElMMo miRBase PITA PicTar5 PicTar6 TargetscanS 0.3105<br />

Diana-microT ElMMo miRanda PicTar6 0.3104<br />

Diana-microT miRBase 0.3100<br />

ElMMo miRanda 0.3100<br />

Diana-microT ElMMo miRBase miRanda 0.3100<br />

Diana-microT PicTar5 0.3100<br />

216


8.5 Target Prediction Ensemble Analysis Results<br />

Prediction algorithms involved in the combination set E-score<br />

Diana-microT ElMMo miRanda PicTar5 0.3100<br />

Diana-microT ElMMo miRanda TargetscanS 0.3099<br />

Diana-microT ElMMo miRanda PITA 0.3098<br />

Diana-microT ElMMo miRBase miRanda PicTar6 0.3098<br />

Diana-microT ElMMo miRanda PicTar5 PicTar6 0.3098<br />

Diana-microT TargetscanS 0.3098<br />

Diana-microT ElMMo miRanda PicTar6 TargetscanS 0.3098<br />

Diana-microT PITA 0.3097<br />

ElMMo miRanda PicTar6 0.3097<br />

Diana-microT ElMMo miRanda PITA PicTar6 0.3097<br />

Diana-microT ElMMo miRBase miRanda PITA 0.3095<br />

Diana-microT ElMMo miRBase miRanda PicTar5 0.3095<br />

Diana-microT ElMMo miRanda PITA PicTar5 0.3095<br />

Diana-microT PITA PicTar6 0.3095<br />

Diana-microT ElMMo miRBase miRanda TargetscanS 0.3095<br />

Diana-microT ElMMo miRanda PITA TargetscanS 0.3095<br />

Diana-microT ElMMo miRanda PicTar5 TargetscanS 0.3095<br />

Diana-microT PicTar6 TargetscanS 0.3095<br />

ElMMo miRanda PITA 0.3094<br />

Diana-microT miRBase PicTar6 0.3094<br />

Diana-microT ElMMo miRBase miRanda PITA PicTar6 0.3094<br />

Diana-microT PicTar5 PicTar6 0.3094<br />

Diana-microT ElMMo miRanda PITA PicTar5 PicTar6 0.3094<br />

Diana-microT ElMMo miRBase miRanda PicTar6 TargetscanS 0.3094<br />

Diana-microT ElMMo miRanda PITA PicTar6 TargetscanS 0.3094<br />

Diana-microT ElMMo miRanda PicTar5 PicTar6 TargetscanS 0.3094<br />

ElMMo miRBase miRanda 0.3093<br />

ElMMo miRanda PicTar5 0.3093<br />

ElMMo miRanda PITA PicTar6 0.3093<br />

Diana-microT ElMMo miRBase miRanda PicTar5 PicTar6 0.3093<br />

ElMMo miRanda TargetscanS 0.3093<br />

Diana-microT miRBase PITA 0.3092<br />

Diana-microT PITA PicTar5 0.3092<br />

Diana-microT ElMMo miRBase miRanda PITA PicTar5 0.3092<br />

Diana-microT PITA TargetscanS 0.3092<br />

Diana-microT ElMMo miRBase miRanda PITA TargetscanS 0.3092<br />

Diana-microT ElMMo miRanda PITA PicTar5 TargetscanS 0.3092<br />

ElMMo miRanda PicTar6 TargetscanS 0.3092<br />

Diana-microT ElMMo miRBase miRanda PITA PicTar6 TargetscanS 0.3092<br />

Diana-microT ElMMo miRanda PITA PicTar5 PicTar6 TargetscanS 0.3092<br />

ElMMo miRBase miRanda PITA 0.3091<br />

ElMMo miRanda PITA PicTar5 0.3091<br />

ElMMo miRBase miRanda PicTar6 0.3091<br />

ElMMo miRanda PicTar5 PicTar6 0.3091<br />

Diana-microT ElMMo miRBase miRanda PITA PicTar5 PicTar6 0.3091<br />

ElMMo miRanda PITA TargetscanS 0.3091<br />

Diana-microT ElMMo miRBase miRanda PicTar5 TargetscanS 0.3091<br />

Diana-microT PITA PicTar6 TargetscanS 0.3091<br />

Diana-microT miRBase PITA PicTar6 0.3090<br />

ElMMo miRBase miRanda PITA PicTar6 0.3090<br />

Diana-microT PITA PicTar5 PicTar6 0.3090<br />

ElMMo miRanda PITA PicTar5 PicTar6 0.3090<br />

Diana-microT ElMMo miRBase miRanda PITA PicTar5 TargetscanS 0.3090<br />

ElMMo miRanda PITA PicTar6 TargetscanS 0.3090<br />

Diana-microT ElMMo miRBase miRanda PicTar5 PicTar6 TargetscanS 0.3090<br />

Diana-microT ElMMo miRBase miRanda PITA PicTar5 PicTar6 TargetscanS 0.3089<br />

PITA 0.3088<br />

ElMMo miRBase miRanda PITA PicTar5 0.3088<br />

Diana-microT miRBase TargetscanS 0.3088<br />

ElMMo miRBase miRanda TargetscanS 0.3088<br />

Diana-microT miRBase PITA TargetscanS 0.3088<br />

ElMMo miRBase miRanda PITA TargetscanS 0.3088<br />

Diana-microT PicTar5 TargetscanS 0.3088<br />

ElMMo miRanda PicTar5 TargetscanS 0.3088<br />

Diana-microT PITA PicTar5 TargetscanS 0.3088<br />

ElMMo miRanda PITA PicTar5 TargetscanS 0.3088<br />

ElMMo miRBase miRanda PicTar5 0.3087<br />

Diana-microT miRBase PITA PicTar5 0.3087<br />

ElMMo miRBase miRanda PITA PicTar5 PicTar6 0.3087<br />

ElMMo miRBase miRanda PicTar6 TargetscanS 0.3087<br />

Diana-microT miRBase PITA PicTar6 TargetscanS 0.3087<br />

ElMMo miRBase miRanda PITA PicTar6 TargetscanS 0.3087<br />

ElMMo miRanda PicTar5 PicTar6 TargetscanS 0.3087<br />

Diana-microT PITA PicTar5 PicTar6 TargetscanS 0.3087<br />

217


8.5 Target Prediction Ensemble Analysis Results<br />

Prediction algorithms involved in the combination set E-score<br />

ElMMo miRanda PITA PicTar5 PicTar6 TargetscanS 0.3087<br />

PITA PicTar6 0.3086<br />

Diana-microT miRBase PITA PicTar5 PicTar6 0.3086<br />

ElMMo miRBase miRanda PITA PicTar5 TargetscanS 0.3086<br />

Diana-microT miRBase PicTar5 0.3085<br />

ElMMo miRBase miRanda PicTar5 PicTar6 0.3085<br />

PITA TargetscanS 0.3085<br />

Diana-microT miRBase PITA PicTar5 TargetscanS 0.3085<br />

Diana-microT miRBase PicTar6 TargetscanS 0.3085<br />

Diana-microT PicTar5 PicTar6 TargetscanS 0.3085<br />

ElMMo miRBase miRanda PITA PicTar5 PicTar6 TargetscanS 0.3085<br />

ElMMo miRBase miRanda PicTar5 TargetscanS 0.3084<br />

PITA PicTar6 TargetscanS 0.3083<br />

Diana-microT miRBase PITA PicTar5 PicTar6 TargetscanS 0.3083<br />

miRBase PITA 0.3082<br />

PITA PicTar5 0.3082<br />

ElMMo miRBase miRanda PicTar5 PicTar6 TargetscanS 0.3082<br />

miRBase PITA PicTar6 0.3081<br />

Diana-microT miRBase PicTar5 PicTar6 0.3080<br />

PITA PicTar5 PicTar6 0.3080<br />

miRBase PITA TargetscanS 0.3080<br />

Diana-microT miRBase PicTar5 TargetscanS 0.3080<br />

PITA PicTar5 TargetscanS 0.3080<br />

miRBase PITA PicTar6 TargetscanS 0.3079<br />

PITA PicTar5 PicTar6 TargetscanS 0.3079<br />

Diana-microT miRBase PicTar5 PicTar6 TargetscanS 0.3078<br />

miRBase PITA PicTar5 0.3077<br />

miRBase PITA PicTar5 TargetscanS 0.3076<br />

miRBase PITA PicTar5 PicTar6 0.3075<br />

miRBase PITA PicTar5 PicTar6 TargetscanS 0.3075<br />

TargetscanS 0.3073<br />

Diana-microT miRanda PITA TargetscanS 0.3071<br />

Diana-microT miRanda PITA 0.3070<br />

Diana-microT miRanda PITA PicTar6 TargetscanS 0.3070<br />

Diana-microT miRanda PITA PicTar6 0.3069<br />

Diana-microT miRBase miRanda PITA TargetscanS 0.3068<br />

Diana-microT miRanda PITA PicTar5 TargetscanS 0.3068<br />

PicTar6 TargetscanS 0.3068<br />

Diana-microT miRBase miRanda PITA 0.3067<br />

Diana-microT miRanda PITA PicTar5 0.3067<br />

Diana-microT miRBase miRanda PITA PicTar6 0.3067<br />

Diana-microT miRBase miRanda PITA PicTar6 TargetscanS 0.3067<br />

Diana-microT miRanda PITA PicTar5 PicTar6 TargetscanS 0.3067<br />

Diana-microT miRanda PITA PicTar5 PicTar6 0.3066<br />

Diana-microT miRBase miRanda PITA PicTar5 TargetscanS 0.3066<br />

Diana-microT miRBase miRanda PITA PicTar5 0.3065<br />

Diana-microT miRBase miRanda PITA PicTar5 PicTar6 TargetscanS 0.3065<br />

Diana-microT miRBase miRanda PITA PicTar5 PicTar6 0.3064<br />

miRanda PITA TargetscanS 0.3060<br />

miRanda PITA PicTar6 TargetscanS 0.3060<br />

miRBase TargetscanS 0.3059<br />

miRanda PITA 0.3058<br />

miRBase miRanda PITA TargetscanS 0.3058<br />

PicTar5 TargetscanS 0.3058<br />

miRanda PITA PicTar5 TargetscanS 0.3058<br />

miRanda PITA PicTar6 0.3057<br />

miRBase miRanda PITA PicTar6 TargetscanS 0.3057<br />

miRanda PITA PicTar5 PicTar6 TargetscanS 0.3057<br />

miRBase miRanda PITA PicTar5 TargetscanS 0.3056<br />

miRBase PicTar6 TargetscanS 0.3056<br />

miRBase miRanda PITA 0.3055<br />

miRanda PITA PicTar5 0.3055<br />

PicTar5 PicTar6 TargetscanS 0.3055<br />

miRBase miRanda PITA PicTar5 PicTar6 TargetscanS 0.3055<br />

miRBase miRanda PITA PicTar6 0.3054<br />

miRanda PITA PicTar5 PicTar6 0.3054<br />

Diana-microT miRanda TargetscanS 0.3053<br />

miRBase miRanda PITA PicTar5 0.3052<br />

miRBase miRanda PITA PicTar5 PicTar6 0.3052<br />

Diana-microT miRanda PicTar6 TargetscanS 0.3052<br />

Diana-microT miRBase miRanda TargetscanS 0.3050<br />

miRBase PicTar5 TargetscanS 0.3049<br />

Diana-microT miRanda PicTar5 TargetscanS 0.3049<br />

Diana-microT miRBase miRanda PicTar6 TargetscanS 0.3049<br />

218


8.5 Target Prediction Ensemble Analysis Results<br />

Prediction algorithms involved in the combination set E-score<br />

Diana-microT miRanda PicTar5 PicTar6 TargetscanS 0.3049<br />

Diana-microT miRBase miRanda PicTar5 TargetscanS 0.3047<br />

miRBase PicTar5 PicTar6 TargetscanS 0.3047<br />

Diana-microT miRBase miRanda PicTar5 PicTar6 TargetscanS 0.3046<br />

Diana-microT miRanda 0.3045<br />

Diana-microT miRanda PicTar6 0.3044<br />

Diana-microT miRBase miRanda 0.3041<br />

Diana-microT miRanda PicTar5 0.3040<br />

Diana-microT miRBase miRanda PicTar6 0.3040<br />

Diana-microT miRanda PicTar5 PicTar6 0.3039<br />

Diana-microT miRBase miRanda PicTar5 0.3037<br />

Diana-microT miRBase miRanda PicTar5 PicTar6 0.3037<br />

PicTar6 0.3020<br />

miRanda TargetscanS 0.3020<br />

miRanda PicTar6 TargetscanS 0.3020<br />

miRBase miRanda TargetscanS 0.3019<br />

miRBase miRanda PicTar6 TargetscanS 0.3019<br />

miRanda PicTar5 TargetscanS 0.3018<br />

miRanda PicTar5 PicTar6 TargetscanS 0.3018<br />

miRBase miRanda PicTar5 TargetscanS 0.3017<br />

miRBase miRanda PicTar5 PicTar6 TargetscanS 0.3017<br />

miRBase PicTar6 0.3011<br />

miRBase 0.3008<br />

miRBase PicTar5 PicTar6 0.3003<br />

miRBase PicTar5 0.3000<br />

PicTar5 PicTar6 0.2999<br />

PicTar5 0.2989<br />

miRBase miRanda PicTar5 PicTar6 0.2987<br />

miRBase miRanda PicTar6 0.2986<br />

miRBase miRanda PicTar5 0.2985<br />

miRBase miRanda 0.2984<br />

miRanda PicTar5 PicTar6 0.2983<br />

miRanda PicTar6 0.2982<br />

miRanda PicTar5 0.2981<br />

miRanda 0.2979<br />

219


Chapter 9<br />

Discussion<br />

9.1 Digital Pr<strong>of</strong>iling <strong>of</strong> GNS Cell Lines<br />

Gliobastoma multiforme is the most common primary brain tumour and the<br />

most aggressive glioma in adults. No effective solution has been embodied<br />

as a treatment yet, causing the prognosis for this disease to be very poor,<br />

with a median survival time <strong>of</strong> 15 months [472]. The extensive cellular het-<br />

erogeneity typical <strong>of</strong> glioblastomas is confirmed by the consistently different<br />

molecular signatures and copy number variations observed in the subclasses<br />

present within the primary and secondary subtypes [154,334,450]. These ob-<br />

servations are very important since they point at the need for approaching<br />

treatment research from a more molecular standpoint that allows for patient<br />

treatment diversification, in which the patient’s genome is specifically tailored<br />

to obtain the most effective results. An important finding within the cancer<br />

stem cell field in the context <strong>of</strong> glioblastomas, was the observation that these<br />

tumours contain a population <strong>of</strong> cells with similarities to NS cells. Before<br />

then, gliomas were studied as cancer cell lines, which hid the underlying stem<br />

cell component <strong>of</strong> the tumour by causing it to differentiate and could therefore<br />

never be specifically targeted. NS cells give rise to both neurons and glia during<br />

the development <strong>of</strong> the nervous system and are present in restricted regions <strong>of</strong><br />

the adult human brain, where they constitute proliferating germinative zones<br />

that are active throughout adulthood [248]. According to the cancer stem cell<br />

hypothesis, such stem cell-like cell populations are responsible for maintaining<br />

cancers, as well as giving rise to the differentiated progeny responsible for the<br />

cellular diversity <strong>of</strong> many neoplasias, including glioblastoma [379]. If this is<br />

the case, isolating and characterizing the glioma cancer stem cells will be key<br />

to developing efficient therapies for glioblastoma multiforme.<br />

220


9.1. Digital Pr<strong>of</strong>iling <strong>of</strong> GNS Cell Lines Discussion<br />

Before Pollard et al [404] adapted the protocol developed by Conti et al [107]<br />

for the derivation <strong>of</strong> NS cells in adherent serum-free culture, to glioma tu-<br />

mours, glioma-related research was conducted on glioma cell lines (see section<br />

1.3) and neurosphere cultures (see section 2.2). Unlike the derivation method<br />

for cancer cell lines, which involves serum-containing media that is selective<br />

for proliferative cells which therefore lose their differentiation identity, the<br />

NS cell line derivation method uses a defined medium with the addition <strong>of</strong><br />

growth hormones EGF and FGF2 that rather supports the self-renewal pro-<br />

cess. While neurosphere cultures have been instrumental in the detection and<br />

quantification <strong>of</strong> the presence <strong>of</strong> a stem cell component in gliomas, the imme-<br />

diate differentiation that is observed when neurospheres start adhering, makes<br />

them a suboptimal tool for the long term characterisation and manipulation<br />

<strong>of</strong> glioma stem cells. When it was discovered that, although adult human NS<br />

cells are difficult to study, fetal human NS cells can be isolated and main-<br />

tained as untransformed cell lines in serum-free medium supplemented with<br />

growth factors, the link was made that the same protocol could be used to ob-<br />

tain NS-like cells from gliomas and maintain them in vitro [404]. These cells,<br />

termed <strong>Glioma</strong> <strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> (GNS), have similar morphology as NS cells<br />

obtained in the same manner, when grown as adherent cultures and feature<br />

other similarities including expression <strong>of</strong> progenitor cell markers and capacity<br />

to differentiate into multiple neural lineages. In contrast to NS cells, however,<br />

GNS cells harbour genetic aberrations characteristic <strong>of</strong> glioblastoma and form<br />

glioma-like tumours when transplanted into immunocompromised mice [404].<br />

Thus, the derivation <strong>of</strong> NS cells from brain tissue forms the basis for culturing<br />

GNS cells from gliomas and by using the same protocol for the isolation <strong>of</strong><br />

fetal human NS cells, the isolation <strong>of</strong> NS-like cells in adherent culture [404] set<br />

the best research platform for conducting research that lends itself better to<br />

the achievement <strong>of</strong> patient tailored therapies.<br />

In this thesis I show several lines <strong>of</strong> support for GNS lines being suitable models<br />

for the understanding <strong>of</strong> the molecular basis <strong>of</strong> glioma:<br />

1. GNS/NS differential expression analysis highlights pathways in glioma;<br />

2. GNS expression pr<strong>of</strong>iles capture molecular signatures <strong>of</strong> glioblastoma<br />

subtypes established by large microarray studies;<br />

3. many core DE genes identified in the GNS/NS comparison show similar<br />

changes in expression data from primary glioblastomas and xenografts.<br />

221


9.1. Digital Pr<strong>of</strong>iling <strong>of</strong> GNS Cell Lines Discussion<br />

In order to reveal transcriptional changes that underlie glioblastoma, I per-<br />

formed an in-depth analysis <strong>of</strong> gene expression in malignant stem cells derived<br />

from patient tumours in relation to untransformed, karyotypically normal neu-<br />

ral stem cells. These cell types are closely related and it has been hypothe-<br />

sised that gliomas arise by mutations in NS cells or in glial cells that have<br />

reacquired stem cell features [404]. We measured gene expression by high-<br />

throughput RNA tag sequencing (Tag-seq), a method which features high sen-<br />

sitivity and reproducibility compared to microarrays [490]. qRT-PCR valida-<br />

tion further demonstrates that Tag-seq expression values are highly accurate.<br />

Other cancer samples and cell lines have recently been pr<strong>of</strong>iled with the same<br />

method [346,383], which should make these samples comparable to ours and<br />

the analyses presented in this thesis. Through Tag-seq expression pr<strong>of</strong>iling<br />

<strong>of</strong> normal and cancer stem cells followed by qRT-PCR validation in a wider<br />

panel <strong>of</strong> 22 cell lines, we identified 29 genes strongly discriminating GNS from<br />

NS cells. Some <strong>of</strong> these genes have previously been implicated in glioma, in-<br />

cluding four with a role in adhesion and/or migration, CD9, ST6GALNAC5,<br />

SYNM and TES [63,241,251,477], and two transcriptional regulators, FOXG1<br />

and CEBPB. This observation is in line with the gene ontology analysis, which<br />

revealed "Cell adhesion” and "Cell migration” as relevant biological processes<br />

amongst our set <strong>of</strong> differentially expressed genes (Fig 7.1), and the fact that,<br />

although infiltrative spread is a common feature <strong>of</strong> all diffuse astrocytic tu-<br />

mours, glioblastoma is particularly notorious for its rapid invasion <strong>of</strong> neigh-<br />

boring brain structures [301]. Activation <strong>of</strong> the TGFβ and AKT pathways<br />

have been described as possible molecular mediators <strong>of</strong> this invasion [245,527]<br />

and a number <strong>of</strong> other expression pr<strong>of</strong>iling studies have identified a subset <strong>of</strong><br />

tumours with elevated expression <strong>of</strong> ECM components as well as intracellular<br />

proteins associated with cell motility [148,390]. FOXG1, which has been pro-<br />

posed to act as an oncogene in glioblastoma by suppressing growth-inhibitory<br />

effects <strong>of</strong> TGFβ [448], showed remarkably strong expression in all 16 GNS lines<br />

assayed by qRT-PCR. CEBPB was recently identified as a master regulator<br />

<strong>of</strong> a mesenchymal gene expression signature associated with poor prognosis in<br />

glioblastoma [85]. Studies in hepatoma and pheochromocytoma cell lines have<br />

shown that the transcription factor encoded by CEBPB (C/EBPβ) promotes<br />

expression <strong>of</strong> DDIT3 [140], another transcriptional regulator that we found<br />

to be up-regulated in GNS cells. DDIT3 encodes the protein CHOP, which<br />

in turn can inhibit C/EBPβ by dimerizing with it and acting as a dominant<br />

negative [85]. This interplay between CEBPB and DDIT3 may be relevant for<br />

222


9.1. Digital Pr<strong>of</strong>iling <strong>of</strong> GNS Cell Lines Discussion<br />

glioma therapy development, as DDIT3 induction in response to a range <strong>of</strong><br />

compounds sensitises glioma cells to apoptosis (see e.g. [216]).<br />

Our results also corroborate a role in glioma for several other genes with limited<br />

prior links to the disease. This list includes PLA2G4A, HMGA2, TAGLN and<br />

TUSC3, all <strong>of</strong> which have been implicated in other neoplasias (Appendix A.2).<br />

PLA2G4A encodes a phospholipase that functions in the production <strong>of</strong> lipid<br />

signaling molecules with mitogenic and pro-inflammatory effects. In a subcu-<br />

taneous xenograft model <strong>of</strong> glioblastoma, expression <strong>of</strong> PLA2G4A by the host<br />

mice was required for tumour growth [285]. For HMGA2, a transcriptional<br />

regulator down-regulated in most GNS lines, low or absent protein expression<br />

has been observed in glioblastoma compared to low-grade gliomas [285], and<br />

HMGA2 polymorphisms have been associated with survival time in glioblas-<br />

toma [295]. TAGLN, another gene down-regulated in most GNS lines, encodes<br />

the actin-binding protein transgelin. TAGLN has been characterised as a tu-<br />

mour suppressor with lost expression in prostate, breast and colon cancers [30],<br />

but we only found one prior study on TAGLN in glioma, showing low expression<br />

in a glioma cell line from rats [181]. TUSC3 is commonly silenced by promoter<br />

methylation in glioblastoma, in particular in patients above 40 years <strong>of</strong> age.<br />

Loss or down-regulation <strong>of</strong> TUSC3 has been found in several other cancers,<br />

e.g. <strong>of</strong> the colon where TUSC3 becomes hypermethylated with age [13]. The<br />

function <strong>of</strong> TUSC3 is unknown, but may relate to protein glycosylation [337].<br />

The set <strong>of</strong> 29 genes found to generally distinguish GNS from NS cells also in-<br />

cludes multiple genes implicated in other neoplasias, but without direct links<br />

to glioma, such as SULF2, NNMT and LMO4 (Appendix A.2). Of these, the<br />

transcriptional regulator LMO4 may be <strong>of</strong> particular interest, as it is well stud-<br />

ied as an oncogene in breast cancer and regulated through the phosphoinosi-<br />

tide 3-kinase pathway [340], which is commonly affected in glioblastoma [326].<br />

SULF2 encodes an extracellular sulfatase that modulates interactions between<br />

growth factors and their receptors, with effects on multiple signaling pathways.<br />

It is up-regulated in several cancers, including a mouse model <strong>of</strong> glioma [211]<br />

and, as shown in our differential expression analysis, in many GNS lines.<br />

Five <strong>of</strong> these 29 genes have not been directly implicated in cancer. This<br />

list comprises one gene down-regulated in GNS lines (PLCH1) and four up-<br />

regulated (ADD2, LYST, PLA2G4A, PDE1C and PRSS12). PLCH1 is in-<br />

volved in phosphoinositol signaling [228], like the frequently mutated phospho-<br />

223


9.1. Digital Pr<strong>of</strong>iling <strong>of</strong> GNS Cell Lines Discussion<br />

inositide 3-kinase complex [326]. Interestingly, both PDE1C and PLCH1 are<br />

activated by intracellular Ca 2+ [126,228], in line with these genes being involved<br />

in PI3K and cAMP regulation, which are both Ca 2+ regulated pathways (see<br />

pathway figure 7.8). PLA2G4A encodes a cytoplasmic phospholipase involved<br />

in production <strong>of</strong> lipid signaling molecules with mitogenic and pro-inflammatory<br />

effects [192]. ADD2 encodes a cytoskeletal protein that interacts with FYN,<br />

a tyrosine kinase promoting cancer cell migration [456,533]. For PDE1C, a<br />

cyclic nucleotide phosphodiesterase gene, we found higher expression to cor-<br />

relate with shorter survival after surgery. Up-regulation <strong>of</strong> PDE1C has been<br />

associated with proliferation in other cell types through hydrolysis <strong>of</strong> cAMP<br />

and cGMP [126,437]. PRSS12 encodes a protease that can activate tissue plas-<br />

minogen activator (tPA) [335], an enzyme which is highly expressed by glioma<br />

cells and has been suggested to promote invasion [168]. Indeed, our Tag-seq<br />

data shows that the tissue plasminogen activator gene PLAT is expressed in all<br />

the assayed GNS lines and strongly up-regulated in all except one (Appendix<br />

A.1). LYST, which encodes a cytoplasmic protein involved in lysosomal traf-<br />

ficking, has no clear role in cancer-related processes, although there is evidence<br />

<strong>of</strong> altered protein kinase C levels in LYST-deficient cells [168]. The GNS versus<br />

NS transcriptome comparison thus identified multiple genes known to play a<br />

role in glioma as well as several other genes likely to do so.<br />

I have compared gene expression and non-coding RNA expression between can-<br />

cer stem cells from glioblastoma and NS cells, as well as evaluated correlations<br />

amongst sets <strong>of</strong> highly significant differentially expressed genes found in our<br />

dataset and other glioblastoma cell lines established by The Cancer Genome<br />

Atlas project [326] and other lower grade gliomas from studies by Freije et<br />

al [148] and Phillips et al [390]. I have also verified how gene expression might<br />

be affected by the aberrations known to exist in glioblastoma cell genomes to<br />

conclude that there was only a modest trend between the presence <strong>of</strong> aberra-<br />

tions and the gene expression observed and no adjustment needed to be made<br />

to our differential expression calls. Interestingly, the GNS cell lines tend to<br />

have very low genomic instability outside the one carried in lieu <strong>of</strong> them being<br />

cancer-derived cell lines, which typically increases over a short period <strong>of</strong> time<br />

to then stabilise later on in classic cancer cell lines. GNS cell lines start accu-<br />

mulating chromosomal and genomic aberrations very late, after having passed<br />

the one hundred passage mark [404]. The four GNS lines that we pr<strong>of</strong>iled have<br />

been thoroughly phenotypically characterised and all give rise to glioma-like<br />

224


9.1. Digital Pr<strong>of</strong>iling <strong>of</strong> GNS Cell Lines Discussion<br />

tumours when transplanted into immunocompromised mice [404]. In addition,<br />

the lines were established from tumours with differing histology, allowing us<br />

to sample the breadth <strong>of</strong> the disease. In fact, our correlation analysis (Fig<br />

6.6) showed that when compared with each other the GNS cell lines fared<br />

the worst with respect to the high scoring correlations observed, instead, be-<br />

tween our biological replicates (the two cell lines established from the same<br />

parental tumour, but in different laboratories) and the two NS cell lines. This<br />

is expected, as G144, G166 and G179 originate from different tumours with<br />

histologically distinct properties and further confirms the presence <strong>of</strong> molecular<br />

variability amongst different glioblastoma patients: G144 was derived from a<br />

glioblastoma with a significant oligodendrocyte component, G166 from a case<br />

<strong>of</strong> glioblastoma multiforme and G179 from a giant cell glioblastoma.<br />

Furthermore, by considering expression changes in a pathway context (Fig<br />

7.8), we identified several genes that have not been directly implicated in<br />

glioma, but participate in glioma-related pathways, such as the putative cell<br />

adhesion gene ITGBL1 [49], the orphan nuclear receptor NR0B1, which is<br />

strongly up-regulated in G179 and is known to be up-regulated and mediate<br />

tumour growth in Ewing’s sarcoma [158], and the genes PARP3 and PARP12<br />

(Table 6.6). Other examples include three down-regulated calcium channel<br />

genes, CACNA1C, CACNG7 and CACNG8, and one up-regulated, CACNA1A.<br />

Deregulation <strong>of</strong> these genes could affect cellular calcium levels, which influence<br />

the activation status <strong>of</strong> protein kinase C and indirectly the MAPK pathway.<br />

Another example is ITGBL1, an integrin-related gene that is expressed by<br />

G166 and G179 and may play a role in cell adhesion [49]. The pathway analy-<br />

sis also highlighted the up-regulated genes PARP3 and PARP12, which belong<br />

to the PARP family <strong>of</strong> ADP-ribosyl transferase genes involved in DNA repair.<br />

The up-regulation <strong>of</strong> these genes may have relevance for therapy, as PARP<br />

inhibitors currently are in clinical trials for several cancers and there is evi-<br />

dence that PTEN loss, which is common in glioblastoma, may sensitise cells<br />

to PARP inhibitors [325].<br />

Transcriptome analysis thus identified multiple genes <strong>of</strong> known significance<br />

in glioma pathology as well as several novel candidate genes and pathways.<br />

These results are further corroborated by survival analysis, which revealed a<br />

GNS expression signature associated with patient survival time in five inde-<br />

pendent datasets. This finding is compatible with the notion that gliomas<br />

225


9.1. Digital Pr<strong>of</strong>iling <strong>of</strong> GNS Cell Lines Discussion<br />

contain a GNS component <strong>of</strong> relevance for prognosis. Five individual GNS<br />

signature genes were significantly associated with survival <strong>of</strong> glioblastoma pa-<br />

tients in both <strong>of</strong> the two largest data sets: PLS3, HOXD10, TUSC3, PDE1C<br />

and the well-studied tumour suppressor PTEN. PLS3 (T-plastin) regulates<br />

actin organisation and its over-expression in the CV-1 cell line resulted in<br />

partial loss <strong>of</strong> adherence [26]. Elevated PLS3 expression in GNS cells may<br />

thus be relevant for the invasive phenotype. The association between tran-<br />

scriptional up-regulation <strong>of</strong> HOXD10 and poor survival is surprising, because<br />

HOXD10 protein levels are suppressed by a microRNA (miR-10b), which is<br />

highly expressed in gliomas, and it has been suggested that HOXD10 suppres-<br />

sion by miR-10b promotes invasion [476]. Notably, in our microRNA microar-<br />

ray dataset microRNA miR-10b is up-regulated three-fold in GNS cell lines<br />

with respect to NS cell lines (Table 6.8; Appendix F) and the HOXD10 mRNA<br />

up-regulation we observe in GNS lines also occurs in glioblastoma tumours, as<br />

shown by comparison with grade III astrocytoma (Fig 7.4b). Similarly to<br />

our findings, miR-10b is present at higher levels in glioblastoma compared to<br />

gliomas <strong>of</strong> lower grade [476]. It is conceivable that HOXD10 transcriptional<br />

up-regulation and post-transcriptional suppression is indicative <strong>of</strong> a regulatory<br />

program associated with poor prognosis in glioma.<br />

Also, from the survival analysis it was clear that tumours from older patients<br />

featured an expression pattern more similar to the GNS signature. One <strong>of</strong> the<br />

genes contributing to this trend, TUSC3, is known to be silenced by promoter<br />

methylation in glioblastoma, particularly in patients over 40 years <strong>of</strong> age [277].<br />

Loss or down-regulation <strong>of</strong> TUSC3 has been found in other cancers, e.g. <strong>of</strong><br />

the colon, where its promoter becomes increasingly methylated with age in the<br />

healthy mucosa [13]. Taken together, these data suggest that transcriptional<br />

changes in healthy aging tissue, such as TUSC3 silencing, may contribute to the<br />

more severe form <strong>of</strong> glioma in older patients. Thus, the molecular mechanisms<br />

underlying the expression changes described here are likely to be complex and<br />

varied. To capture these effects and elucidate their causes, transcriptome anal-<br />

ysis <strong>of</strong> cancer samples will benefit from integration <strong>of</strong> diverse genomic data,<br />

including structural and nucleotide-level genetic alterations, as well as DNA<br />

methylation and other chromatin modifications.<br />

To identify expression alterations common to most glioblastoma cases, other<br />

studies have pr<strong>of</strong>iled tumour biopsies in relation to non-neoplastic brain tis-<br />

sue [196,383,497]. While such comparisons have been revealing, their power<br />

226


9.1. Digital Pr<strong>of</strong>iling <strong>of</strong> GNS Cell Lines Discussion<br />

is constrained by discrepancies between reference and tumour samples; for in-<br />

stance the higher neuronal content <strong>of</strong> normal brain tissue compared to tumours.<br />

Gene expression pr<strong>of</strong>iling <strong>of</strong> tumour biopsies further suffers from mixed signal<br />

due to a stromal cell component and heterogeneous populations <strong>of</strong> cancer cells,<br />

only some <strong>of</strong> which contribute to tumour progression and maintenance [379].<br />

Part <strong>of</strong> a recent study bearing a closer relationship to our analysis examined<br />

gene expression in another panel <strong>of</strong> glioma-derived and normal NS cells [299],<br />

but included neurosphere cultures which <strong>of</strong>ten contain a heterogeneous mix-<br />

ture <strong>of</strong> self-renewing and differentiating cells. We have managed to circumvent<br />

these issues by pr<strong>of</strong>iling uniform cultures <strong>of</strong> primary malignant stem cell lines<br />

that can reconstitute the tumour in vivo [402], in direct comparison to normal<br />

counterparts <strong>of</strong> the same cell type [107,481]. While the resulting expression<br />

patterns largely agree with those obtained from glioblastoma tissues, there are<br />

notable differences. For example, we found the breast cancer oncogene LMO4<br />

(discussed above) to be up-regulated in most GNS lines, although its average<br />

expression in glioblastoma tumours is low relative to normal brain tissue (Fig<br />

7.4 3a). Similarly, TAGLN and TES were absent or low in most GNS lines, but<br />

displayed the opposite trend in glioblastoma tissue compared to normal brain<br />

(Fig 7.4c) or grade III astrocytoma (Fig 7.4d). Importantly, both TAGLN and<br />

TES have been characterised as tumour suppressors in malignancies outside<br />

the brain and the latter is <strong>of</strong>ten silenced by promoter hypermethylation in<br />

glioblastoma [30,349]. A very interesting hypothesis about the TES gene that<br />

we could explore further with specific biochemical assays, is that, assuming the<br />

methylation mark observed in glioblastoma literature is maintained in our GNS<br />

cell lines, there must be a very robust mechanism to keep this gene silenced.<br />

In fact, TES is found on chromosome 7, which is nearly always present in very<br />

high copy number, for example in cell line G144, which carries more than 10<br />

copies at late passages (Fig 6.7). If we could verify that the methylation mark<br />

in our GNS cell lines was maintained, this would be consistent with TES being<br />

silenced very early on in the disease, before the chromosomal gains and ane-<br />

uploidy, which may make it a good candidate as a glioblastoma initiating event.<br />

In assigning each GNS cell line to one <strong>of</strong> the four expression signature pr<strong>of</strong>iles<br />

as defined by the latest study by Verhaak et al [511], each match was reflective<br />

<strong>of</strong> their known histopathological features and this further supported the need<br />

for a patient stratification approach in assigning therapies. G166 and G179,<br />

both glioblastoma cell lines, were assigned to the mesenchymal signature, asso-<br />

227


9.1. Digital Pr<strong>of</strong>iling <strong>of</strong> GNS Cell Lines Discussion<br />

ciated with the poorest prognosis. G144, instead, with its high oligodendrocyte<br />

component known to positively correlate with patient survival rates in glioblas-<br />

toma, was assigned to the proneural signature, linked with neural markers and<br />

therefore the best prognosis <strong>of</strong> all. In comparing the GNS line expression pro-<br />

files to the subtype signatures, we found that both G166 and G179 correlated<br />

strongly with the mesenchymal signature with worst prognosis. Mesenchymal<br />

subtype markers with elevated expression in these two lines included MET,<br />

CD44, CD68 and CASP1 [511]. G144 did not correlate significantly with any<br />

<strong>of</strong> the four signatures, but showed a slight positive correlation with the proneu-<br />

ral one. Supporting such classification <strong>of</strong> G144 were several <strong>of</strong> the hallmarks<br />

<strong>of</strong> the proneural subtype emphasised by Verhaak et al 2010 [511]: high expres-<br />

sion <strong>of</strong> oligodendrocytic development genes PDGFRA, NKX2-2 and OLIG2,<br />

as well as ERBB3, DCX and TCF4 genes and low levels <strong>of</strong> tumour suppressor<br />

CDKN1A. These expression signature pr<strong>of</strong>ile studies should be performed on<br />

an always greater number <strong>of</strong> glioblastoma samples in order for them to be able<br />

to capture even finer differences than the ones proposed by the study by Ver-<br />

haak et al, which have already proven the existence <strong>of</strong> different glioblastoma<br />

classes and different prognoses associated with each class. If each class were<br />

to be targeted by a tailored molecular therapy, this class would find it most<br />

beneficial as a treatment and patients would obtain better results.<br />

A Gene Ontology term analysis confirmed that the sets <strong>of</strong> differentially ex-<br />

pressed genes were enriched for genes involved in processes related to brain de-<br />

velopment and cancer biology. We also observed enrichment <strong>of</strong> genes encoding<br />

regulatory and inflammatory proteins, such as signal transduction components,<br />

cytokines, growth factors and DNA binding proteins. In line with these find-<br />

ings, affected pathways from the KEGG database included Cytokine-cytokine<br />

receptor interaction, Neuroactive ligand-receptor interaction, MAPK signaling<br />

and, expectedly, <strong>Glioma</strong>, a collection <strong>of</strong> genes involved in glioma formation.<br />

GSEA analysis revealed a consistent up-regulation <strong>of</strong> inflammatory genes in<br />

the GNS lines belonging especially to the MHC class II family, suggesting an<br />

immune-evasion phenotype that has already been called upon by a small num-<br />

ber <strong>of</strong> early glioma studies [468,484]. The up-regulation in the GNS lines was<br />

seen in several MHC class II genes, as well as related genes involved in antigen<br />

presentation on MHC class I complexes. Several works have already shown<br />

that MHC class I and II molecules are involved in aspects <strong>of</strong> human cancer<br />

pathology such as invasion and migration [327,421,546]. We find an overall<br />

228


9.1. Digital Pr<strong>of</strong>iling <strong>of</strong> GNS Cell Lines Discussion<br />

transcriptional up-regulation <strong>of</strong> MHC class II molecules and a smaller subset<br />

<strong>of</strong> MHC class I molecules, suggestive <strong>of</strong> the absence <strong>of</strong> transcriptionally active<br />

compensatory mechanisms. Follow-up proteomic studies could be carried out<br />

by us to further explore the dynamics <strong>of</strong> this immunoevasion mechanism in ac-<br />

tion. The identification <strong>of</strong> specific protein expression levels, in fact, would help<br />

us understand the level at which this regulation, which is bound to exist given<br />

the up-regulation observed especially in MHC class II molecules, takes place.<br />

It would help us answer questions such as whether an excess <strong>of</strong> MHC class<br />

II molecules is also present on the surface <strong>of</strong> GNS cells, and if there are any<br />

correlations with the expression signature pr<strong>of</strong>iles and the diagnosis associated<br />

with them. In fact, it is possible that in the proneural signature would fall<br />

those glioblastoma samples that carry higher protein expression levels <strong>of</strong> MHC<br />

class II molecules, which by exposing aberrant extracellular antigens, would<br />

be responsible for a more efficient immunological activation against those cells,<br />

in line with the kinder prognosis associated with this signature. On the other<br />

hand, it is also possible that little or no sample-dependent variation exists in<br />

the up-regulation <strong>of</strong> MHC class molecules and therefore no correlation between<br />

prognosis and MHC member protein levels.<br />

In the attempt to observe whether non-coding RNA regulation plays an impor-<br />

tant role in the levels <strong>of</strong> gene expression observed for GNS cell lines, differential<br />

microRNA-targeted is<strong>of</strong>orm expression and long non-coding RNA expression<br />

were evaluated. Assuming that the differential is<strong>of</strong>orm expression is due to<br />

microRNA regulation exerted on a specific is<strong>of</strong>orm, after having identified<br />

approximately 2,000 is<strong>of</strong>orm candidates for differential expression detected by<br />

multiple tag mappings, we verified which predicted microRNA sequences (from<br />

five <strong>of</strong> the leading algorithms) aligned on their 3'UTRs. An interesting candi-<br />

date for is<strong>of</strong>orm microRNA regulation is gene NTRK2, which encodes a recep-<br />

tor for brain-derived neurotrophic factor, promoting differentiation, prolifera-<br />

tion and survival [118]. In several tumour types, NTRK2 expression correlates<br />

with poor prognosis and metastasis [118] and the protein has been detected<br />

in a subset <strong>of</strong> cells in astrocytomas [29]. The Tag-seq data demonstrates that<br />

GNS lines express a short NTRK2 is<strong>of</strong>orm, potentially the one that lacks the<br />

kinase domain (Fig 6.11), which has been implicated in regulation <strong>of</strong> astrocyte<br />

morphology [366]. Other interesting candidate genes for microRNA regulation<br />

<strong>of</strong> is<strong>of</strong>orm expression, are the tumour suppressor gene BRCA1, involved in p53<br />

signaling [370], the genes AKT2 and AKT3, involved in glioblastoma tumour<br />

evasion phenotypes [245], BMP7, a member <strong>of</strong> the TGF-β superfamily <strong>of</strong> bone<br />

229


9.1. Digital Pr<strong>of</strong>iling <strong>of</strong> GNS Cell Lines Discussion<br />

morphogenetic proteins that plays a key role in the transformation <strong>of</strong> mes-<br />

enchymal cells into bone and cartilage [245], and ERBB2, HLA-A and PTEN.<br />

We found that 226 <strong>of</strong> the approx. 2,000 differentially expressed is<strong>of</strong>orms hosted<br />

at least one microRNA seed sequence between at least two tags, which identi-<br />

fied microRNA regulation as a widely adopted mechanism <strong>of</strong> regulation <strong>of</strong> gene<br />

and is<strong>of</strong>orm expression in GNS cell lines. An important follow-up experiment<br />

for which data came in only recently in our laboratory, is the measurement <strong>of</strong><br />

microRNA expression levels in our GNS cell lines on a microRNA microarray<br />

platform. Analysis <strong>of</strong> this data revealed that, <strong>of</strong> the 226 microRNAs identified<br />

on a prediction basis as regulators <strong>of</strong> is<strong>of</strong>orms within our GNS study, miR-<br />

10b (see discussion above) and miR-26a were consistently up-regulated in our<br />

GNS cell lines, and miR-137, miR 128, miR-34a, miR-129-3p, and miR-451<br />

consistently down-regulated, in line with the glioblastoma microRNA litera-<br />

ture [96,278,364].<br />

In the assessment <strong>of</strong> the regulation potentially performed by long non-coding<br />

RNAs, we found 18 up-regulated and 7 down-regulated putative ncRNAs, three<br />

<strong>of</strong> which are known to be long antisense RNAs: CDKN2BAS, HOTAIRM1<br />

and NEAT1. Although long-non coding RNA regulation still needs to be<br />

thoroughly elucidated, an interesting pattern was observed with the levels <strong>of</strong><br />

expression <strong>of</strong> HOTAIRM1 in our GNS cell lines and the expression trend found<br />

in literature in human NB4 promyelocytic cell lines. In fact, upon induction<br />

<strong>of</strong> granulocytic differentiation, HOTAIRM1 becomes strongly up-regulated in<br />

human NB4 promyelocytic cell lines and normal hematopoietic cells, and its<br />

knock-down in NB4 cells causes down-regulation <strong>of</strong> the HOXA1 and HOXA4<br />

genes [547]. In line with these observations, HOTAIRM1 is found to be up-<br />

regulated in our GNS cell lines compared to our NS lines, and the HOXA1 and<br />

HOXA4 genes are also up-regulated (Fig 6.22), which potentially identifies<br />

them as conserved targets <strong>of</strong> HOTAIRM1, although an immuno-precipitation<br />

assay should be performed in order to conclude that.<br />

Overall, in this thesis I demonstrate that our results support GNS lines as<br />

suitable model for understanding the molecular basis <strong>of</strong> glioblastoma, and use<br />

<strong>of</strong> NS cell lines as controls in this setting. By this approach, we have identified<br />

several likely oncogenes and tumour suppressors that have not previously been<br />

associated with glioblastoma. With the advent <strong>of</strong> GNS cell lines, the glioblas-<br />

toma research field is bound to be enriched by experiments that will represent<br />

230


9.2. MicroRNA Target Prediction Analysis Discussion<br />

in their results also the stem cell component <strong>of</strong> the cancer and therefore enable<br />

more accurate targeting therapeutic strategies. Outside <strong>of</strong> the glioblastoma<br />

research field, similar concepts will be applied for the adherent culturing <strong>of</strong><br />

other solid cancers with a preponderant stem cell component, such as other<br />

brain cancers and breast as well as colon, pancreatic and lymphoma cancers.<br />

The ability to work in an adherent culture that maintains intact the stem cell<br />

component <strong>of</strong> the cancer for long periods <strong>of</strong> time is an enabling feature for the<br />

entire cancer research field that will enable researchers to move forward faster<br />

towards the achievement <strong>of</strong> effective cancer therapeutic strategies.<br />

9.2 MicroRNA Target Prediction Analysis<br />

In this thesis I have also constructed a microRNA target prediction analysis<br />

tool for the manipulation <strong>of</strong> microRNA target prediction data and combined<br />

the exon array expression data (Appendix E) with the microRNA array ex-<br />

pression data (Appendix E) to produce, using the GenemiR s<strong>of</strong>tware tool, an<br />

ensemble analysis that would help me answer the question whether any com-<br />

bination <strong>of</strong> prediction algorithms together were more effective and accurate at<br />

predicting mRNA targets that any prediction algorithm alone.<br />

microRNA target prediction is the result <strong>of</strong> the factoring <strong>of</strong> several different<br />

variables that are each treated differently, depending on the algorithm at work,<br />

in terms <strong>of</strong> the importance that is assigned to each within the algorithm itself.<br />

Although several robust prediction algorithms have been developed, they all<br />

have the problem <strong>of</strong> predicting with a very high percentage <strong>of</strong> false positives.<br />

Furthermore, the poor agreement that is so <strong>of</strong>ten observed between sets <strong>of</strong><br />

results originating from exactly the same input list but processed by different<br />

algorithms, tells us that there needs to be more to be then account <strong>of</strong>. Thus,<br />

the characterisation <strong>of</strong> microRNA function and regulation <strong>of</strong> mRNAs is de-<br />

pendent on a robust strategy for managing disparate results. Some prediction<br />

algorithms generate a large number <strong>of</strong> putative regulatory targets, many <strong>of</strong><br />

which are exclusive to a particular method. Moreover, those results that do<br />

agree will not necessarily exhibit higher prediction accuracy. Even in the best<br />

case where the degree <strong>of</strong> overlap is approximately 70% (between TargetScanS<br />

and PicTar), it is not obvious which algorithm produces the optimal set <strong>of</strong><br />

target predictions. When predictions are made, they must then be viewed<br />

231


9.3. Concluding Remarks Discussion<br />

in context with other genomic information in order to elucidate regulatory<br />

function. In an attempt to elucidate the answer to the research question <strong>of</strong><br />

interest, the hypothesis put forward by Alexiou et al [16] that combinations <strong>of</strong><br />

algorithms predict more accurately than the single algorithm alone was chal-<br />

lenged. Through the analysis described in this thesis it is clear that the target<br />

prediction algorithm ElMMo [155]is at the head <strong>of</strong> the list sorted by accuracy<br />

score out <strong>of</strong> the 258 combinations tested for. Although this analysis shows<br />

that ElMMo in particular has the most accurate target prediction algorithm<br />

within the pool <strong>of</strong> eight tested prediction algorithms, it must be noted that<br />

there is a substantial discrepancy between the score achieved by ElMMo and<br />

the next best score for a single target prediction algorithm. Before Diana-<br />

microT [237,316], the second highest solo score, many combinations <strong>of</strong> two to<br />

three target prediction algorithms appear earlier in the list, demonstrating the<br />

potential for better accuracy than another solo prediction algorithm. Although<br />

it is refreshing to see that one target prediction algorithm can fare better than<br />

the rest, the question remains as to whether the variables and factors that are<br />

taken into consideration, and the ones that are not, are fairly judged in the<br />

algorithm so as to simulate the intricate regulations happening within the cell.<br />

In fact, during this analysis the accuracy score <strong>of</strong> each prediction algorithm<br />

decreased greatly when a background gene list was not used to filter out all<br />

the genes that were predicted but were not tissue specific.<br />

While the results <strong>of</strong> this analysis indicate that some algorithms alone and some<br />

group <strong>of</strong> algorithms are better than others, it highlights at the same time the<br />

importance <strong>of</strong> re-assessing the factors that are taken into consideration to<br />

make the microRNA target predictions. If we have reached the limit <strong>of</strong> what<br />

sequence-based algorithms can achieve, we ought to start thinking <strong>of</strong> adding<br />

another dimension to the field <strong>of</strong> microRNA target prediction algorithms that<br />

takes factors in components <strong>of</strong> the tissue-specific regulatory actions <strong>of</strong> microR-<br />

NAs.<br />

9.3 Concluding Remarks<br />

In this thesis I aimed to characterise the transcriptional landscape <strong>of</strong> <strong>Glioma</strong><br />

<strong>Neural</strong> <strong>Stem</strong> <strong>Cells</strong> (GNS) in the most comprehensive way possible. Several<br />

approaches were taken to examine the expression pr<strong>of</strong>iling data that gave in-<br />

sights into the stem cell component <strong>of</strong> the biology <strong>of</strong> these cells, which had<br />

232


9.3. Concluding Remarks Appendix<br />

previously been overlooked due to the suboptimal culturing systems for the<br />

maintenance <strong>of</strong> pluripotency.<br />

To this end I have approached the analysis from the coding transcriptome and<br />

the non-coding transcriptome point <strong>of</strong> view, trying to draw as many parallels<br />

as possible and as many comparisons as possible with other external datasets<br />

that could give insights in the characteristics the are unique to the GNS sys-<br />

tem.<br />

Pairing the available expression data with new DNA methylation data would<br />

allow us to gain insights into the epigenetic mechanisms at work. It is, in<br />

fact, important that the future works in this field and with these cell lines<br />

are directed towards a deeper understanding <strong>of</strong> the mechanisms that elect<br />

and maintain them as tumour-initiating agents <strong>of</strong> the glioblastoma primary<br />

tumour.<br />

233


Appendix A<br />

Differentially Expressed Genes<br />

A.1 Differential Expression<br />

The differentially expressed genes generated with the DESeq Bioconductor<br />

package are listed below in alphabetical order. The values for each gene are<br />

reported in each cell line in order to give the reader an idea on the directionality<br />

<strong>of</strong> the fold change between diseased and normal counterpart. Rows with mul-<br />

tiple gene Ensembl and/or Entrez gene IDs separated by comma correspond<br />

to cases where there was not a one-to-one correspondence between Entrez and<br />

Ensembl genes.<br />

234


A.1 Differential Expression Appendix<br />

Table A.1: Classification <strong>of</strong> differentially expressed genes at 10% FDR.<br />

Gene symbol(s) Entrez gene ID(s) Ensembl 56 gene ID(s) Differential expression results Normalised tag counts<br />

log 2(F C) p-value FDR G144ED G144 G166 G179 CB541 CB660<br />

ABAT 18 ENSG00000183044 -2.61 2.69E-04 1.07E-02 769.4 1682.2 48.1 223.8 6470.4 1513.8<br />

AC007405.8,LOC285141 285141 ENSG00000204334 3.05 1.26E-03 3.56E-02 16.2 23.0 120.1 184.0 0.0 25.9<br />

AC012215.1,UNC5D 137970 ENSG00000156687,ENSG00000233863 -Inf 1.06E-08 1.71E-06 0.0 0.0 0.0 0.0 206.3 57.2<br />

AC012354.2 ENSG00000225301 -Inf 4.53E-07 4.98E-05 0.0 0.0 0.0 0.0 26.6 170.3<br />

AC026410.6 ENSG00000227087 3.52 2.06E-03 5.09E-02 56.7 96.6 93.1 19.3 0.0 11.9<br />

AC034102.1 ENSG00000237493 8.09 9.42E-10 1.95E-07 79.2 227.3 187.2 0.0 0.0 1.0<br />

AC067930.1 ENSG00000228381 4.86 1.80E-03 4.69E-02 28.4 50.3 71.3 0.0 0.7 1.9<br />

AC068399.1 ENSG00000204089 4.25 1.80E-05 1.16E-03 1.4 7.4 346.5 22.9 0.0 12.7<br />

AC092296.1 ENSG00000229449 4.90 2.63E-04 1.05E-02 140.1 87.2 79.9 13.6 0.0 4.2<br />

ACIN1 22985 ENSG00000100813 5.44 4.58E-04 1.65E-02 65.1 0.0 133.1 0.0 0.0 2.0<br />

ACTA2 59 ENSG00000107796 -2.67 3.27E-03 7.27E-02 0.0 0.0 114.4 146.7 618.4 486.3<br />

ACTC1 70 ENSG00000159251 -Inf 1.14E-09 2.33E-07 1.4 0.0 0.0 0.0 0.0 422.8<br />

ADAMTS1 9510 ENSG00000154734 2.88 1.30E-03 3.62E-02 103.3 405.7 57.5 107.6 0.0 51.1<br />

ADAMTS10 81794 ENSG00000142303 -3.00 7.32E-04 2.32E-02 87.9 58.1 12.5 1.7 165.5 218.6<br />

ADAMTS4 9507 ENSG00000158859 Inf 4.45E-04 1.62E-02 31.1 40.2 3.1 54.1 0.0 0.0<br />

ADCYAP1R1 117 ENSG00000078549 -3.35 1.56E-03 4.18E-02 6.1 16.8 0.0 0.0 108.9 8.8<br />

ADD2 119 ENSG00000075340 5.18 1.62E-03 4.29E-02 27.6 18.5 30.6 60.5 0.0 2.0<br />

ADRA1B 147 ENSG00000170214 5.86 2.94E-03 6.68E-02 0.0 0.0 87.6 1.2 0.0 1.0<br />

ADRA2A 150 ENSG00000150594 Inf 7.29E-05 3.65E-03 65.0 130.0 0.0 0.0 0.0 0.0<br />

AFAP1L2 84632 ENSG00000169129 3.56 4.23E-03 8.93E-02 211.6 214.7 1.3 0.0 0.0 12.3<br />

AGPAT9 84803 ENSG00000138678 Inf 3.58E-15 3.32E-12 2.6 0.0 484.9 582.3 0.0 0.0<br />

AGT 183 ENSG00000135744 10.27 3.91E-16 5.28E-13 1585.7 1784.3 4.5 85.8 0.0 1.0<br />

AIDA 64853 ENSG00000186063 2.25 2.85E-03 6.53E-02 349.3 401.7 533.4 334.1 3.2 173.7<br />

AKR1B10 57016 ENSG00000198074 Inf 8.11E-08 1.07E-05 0.0 0.0 27.5 219.6 0.0 0.0<br />

ALDH1A3 220 ENSG00000184254 4.77 1.59E-03 4.23E-02 0.0 0.0 9.9 114.8 0.0 3.0<br />

AMMECR1 9949 ENSG00000101935 3.30 3.46E-03 7.58E-02 42.1 33.4 65.7 96.5 0.0 13.0<br />

ANGPTL1 9068 ENSG00000116194 -7.69 9.68E-06 6.95E-04 5.1 0.0 1.3 0.0 88.4 84.2<br />

ANGPTL2 23452 ENSG00000136859 3.74 2.26E-03 5.48E-02 55.6 128.1 8.0 81.8 3.8 6.7<br />

AP000280.1,C21orf62 56245 ENSG00000205929,ENSG00000239565 -4.76 4.65E-06 3.71E-04 0.0 16.4 0.0 0.0 236.6 47.3<br />

APLN 8862 ENSG00000171388 -7.41 1.52E-06 1.43E-04 1.4 1.6 0.0 0.0 133.9 44.7<br />

APOD 347 ENSG00000189058 3.18 3.52E-04 1.34E-02 558.9 1217.7 12.5 6.6 0.0 90.9<br />

AQP4 361 ENSG00000171885 -Inf 2.09E-03 5.14E-02 5.4 0.0 0.0 0.0 7.8 39.4<br />

ARC 23237 ENSG00000198576 -4.03 1.22E-05 8.48E-04 31.7 27.8 6.8 5.0 417.6 19.0<br />

ARHGAP20 57569 ENSG00000137727 Inf 6.40E-04 2.11E-02 156.7 74.6 0.8 18.0 0.0 0.0<br />

ARHGAP8,PRR5,PRR5-<br />

23779,553158,55615 ENSG00000186654,ENSG00000241484 -5.71 1.48E-03 4.03E-02 0.0 0.0 3.1 0.0 5.1 103.5<br />

235<br />

ARHGAP8<br />

ARHGEF7 8874 ENSG00000102606 -3.05 9.53E-05 4.54E-03 120.8 94.7 18.6 28.3 713.6 79.4<br />

ASNS 440 ENSG00000070669 2.33 4.47E-03 9.28E-02 619.4 1064.7 1962.9 478.9 207.0 256.1<br />

ASPN 54829 ENSG00000106819 -Inf 4.57E-03 9.45E-02 0.0 0.0 0.0 0.0 0.0 39.5<br />

ATAD3C 219293 ENSG00000215915 6.90 1.30E-04 5.85E-03 0.0 13.8 118.7 3.0 1.0 0.0<br />

ATOH8 84913 ENSG00000168874 -3.96 3.00E-05 1.74E-03 129.9 36.2 0.0 18.8 226.6 339.9<br />

ATP10B 23120 ENSG00000118322 Inf 7.54E-13 2.95E-10 669.9 823.1 0.0 8.1 0.0 0.0<br />

ATP1A2 477 ENSG00000018625 -6.16 4.66E-11 1.33E-08 39.0 37.5 0.6 0.0 471.0 1346.4<br />

ATP1B2 482 ENSG00000129244 -2.46 6.07E-04 2.02E-02 337.6 303.2 6.3 69.3 816.5 581.6<br />

AZGP1 563 ENSG00000160862 Inf 1.37E-14 8.49E-12 1174.8 1213.2 23.8 0.0 0.0 0.0<br />

B3GNT9 84752 ENSG00000237172 2.96 5.06E-04 1.77E-02 84.4 78.1 85.8 504.6 10.7 46.6<br />

B4GALNT1 2583 ENSG00000135454 3.56 1.99E-04 8.35E-03 1771.8 1329.5 54.1 93.5 62.7 21.1<br />

BACE2 25825 ENSG00000182240 7.97 1.22E-13 5.48E-11 227.5 423.1 253.4 468.2 0.0 2.7<br />

BAMBI 25805 ENSG00000095739 5.49 4.12E-09 7.20E-07 1880.4 3430.1 25.3 173.3 23.2 30.4<br />

BATF3 55509 ENSG00000123685 5.12 9.08E-05 4.34E-03 212.0 123.3 5.0 83.6 0.0 3.8<br />

BC008001 4.94 1.32E-03 3.67E-02 25.7 171.5 2.7 0.0 3.8 0.0


A.1 Differential Expression Appendix<br />

Gene symbol(s) Entrez gene ID(s) Ensembl 56 gene ID(s) Differential expression results Normalised tag counts<br />

log 2(F C) p-value FDR G144ED G144 G166 G179 CB541 CB660<br />

BEX5 340542 ENSG00000184515 4.39 1.64E-04 7.07E-03 0.0 0.0 159.3 65.7 0.0 6.8<br />

BGN 633 ENSG00000182492 -5.65 8.87E-05 4.27E-03 20.3 4.5 0.0 0.0 0.0 156.2<br />

BID 637 ENSG00000015475 2.53 4.78E-03 9.74E-02 103.7 66.6 117.1 429.8 42.6 28.3<br />

BIRC3 330 ENSG00000023445 Inf 1.20E-13 5.48E-11 0.0 0.0 48.2 739.4 0.0 0.0<br />

BMP7 655 ENSG00000101144 -1.95 4.86E-03 9.82E-02 301.5 145.7 0.0 3.3 197.9 185.3<br />

BMP8B 656 ENSG00000116985 7.34 3.19E-06 2.74E-04 198.7 244.2 2.0 0.0 0.0 1.0<br />

BMPER 168667 ENSG00000164619 Inf 2.97E-03 6.69E-02 81.9 65.6 0.0 6.6 0.0 0.0<br />

BST2 684 ENSG00000130303 7.02 5.19E-10 1.12E-07 0.0 1.6 263.7 229.0 1.1 0.9<br />

BTBD11 121551 ENSG00000151136 -6.75 7.12E-04 2.27E-02 0.0 0.0 1.3 0.0 45.1 44.3<br />

BTC 685 ENSG00000174808 Inf 7.52E-07 7.76E-05 5.4 35.9 37.9 126.8 0.1 0.0<br />

BTG1 694 ENSG00000133639 5.34 7.29E-08 9.85E-06 223.8 302.7 228.7 24.6 0.0 8.6<br />

C10orf11 83938 ENSG00000148655 3.82 2.80E-05 1.65E-03 195.7 603.7 175.1 152.8 20.5 23.6<br />

C10orf116 10974 ENSG00000148671 6.23 4.44E-06 3.59E-04 0.0 0.0 220.6 8.3 0.0 2.0<br />

C10orf81 79949 ENSG00000148735 Inf 2.49E-06 2.19E-04 67.8 57.9 9.7 115.1 0.0 0.0<br />

C10orf90 118611 ENSG00000154493 Inf 2.12E-04 8.75E-03 53.9 106.7 9.4 0.0 0.0 0.0<br />

C14orf143 90141 ENSG00000140025 Inf 3.76E-06 3.10E-04 3.6 0.0 81.2 93.4 0.0 0.0<br />

C1S 716 ENSG00000182326 3.25 4.97E-04 1.75E-02 10.2 27.1 25.1 440.9 15.2 19.1<br />

C1orf133 574036 ENSG00000203706 2.57 2.45E-03 5.81E-02 8.2 35.9 68.2 358.3 0.0 51.7<br />

C1orf187 374946 ENSG00000162490 3.07 3.73E-03 8.08E-02 152.5 112.4 1.4 142.9 0.0 20.2<br />

C1orf94 84970 ENSG00000142698 Inf 3.01E-04 1.18E-02 9.6 53.0 0.0 46.0 0.0 0.0<br />

C20orf103 24141 ENSG00000125869 5.00 3.62E-03 7.87E-02 8.2 7.9 0.0 89.5 0.0 2.3<br />

C2orf80 389073 ENSG00000188674 4.69 1.55E-03 4.18E-02 264.9 154.4 0.0 1.6 0.0 3.8<br />

C3 718 ENSG00000125730 Inf 2.15E-12 7.99E-10 0.0 0.0 24.4 585.6 0.0 0.0<br />

C3orf58 205428 ENSG00000181744 -2.70 1.58E-03 4.22E-02 41.9 53.1 39.3 95.7 489.1 327.6<br />

C4orf32 132720 ENSG00000174749 5.37 6.36E-04 2.11E-02 0.0 0.0 125.8 0.0 0.0 1.7<br />

C5orf13 9315 ENSG00000134986 -3.29 1.01E-04 4.75E-03 230.2 162.6 55.9 234.9 1985.9 962.7<br />

C5orf38 153571 ENSG00000186493 -5.52 9.66E-04 2.88E-02 2.7 1.5 0.0 0.5 54.6 18.2<br />

C5orf41 153222 ENSG00000164463 4.56 3.81E-03 8.23E-02 29.7 25.6 82.8 0.0 0.0 3.0<br />

C6orf138 442213 ENSG00000178729,ENSG00000244694 -Inf 7.73E-04 2.40E-02 1.4 0.0 0.0 0.0 40.5 13.0<br />

C6orf15 29113 ENSG00000204542 Inf 2.61E-18 1.24E-14 0.0 0.0 2446.3 0.0 0.0 0.0<br />

C7orf16 10842 ENSG00000106341 -Inf 1.64E-03 4.35E-02 0.0 0.0 0.0 0.0 2.3 48.4<br />

C7orf40 285958 ENSG00000232956 4.02 7.73E-04 2.40E-02 39.2 63.9 133.2 0.0 0.0 8.1<br />

C8orf4 56892 ENSG00000176907 4.75 2.23E-08 3.31E-06 12.6 20.3 613.3 2647.7 27.4 53.3<br />

C9orf125 84302 ENSG00000165152 -3.78 8.38E-06 6.10E-04 73.9 59.0 3.6 8.0 508.5 148.1<br />

C9orf64 84267 ENSG00000165118 7.88 1.40E-12 5.34E-10 114.7 156.8 164.1 752.5 2.9 0.0<br />

C9orf95 54981 ENSG00000106733 4.40 3.54E-03 7.75E-02 15.9 15.6 98.1 14.8 3.0 1.0<br />

CA12 771 ENSG00000074410 -4.65 9.71E-07 9.69E-05 45.9 28.1 22.6 30.3 945.6 415.1<br />

CABP7 164633 ENSG00000100314 Inf 1.51E-05 1.02E-03 0.0 6.1 6.5 135.5 0.0 0.0<br />

CACNA1A 773 ENSG00000141837 7.13 1.84E-14 1.10E-11 1.3 4.7 22.1 1252.5 0.0 5.7<br />

CACNA1C 775 ENSG00000151067 -8.15 2.57E-05 1.54E-03 1.4 0.0 0.6 0.0 103.0 15.9<br />

CACNG7 59284 ENSG00000105605 -2.58 4.27E-03 8.99E-02 89.2 41.8 12.6 2.5 149.8 78.7<br />

CACNG8 59283 ENSG00000142408 -4.77 5.28E-07 5.68E-05 33.1 28.3 3.9 2.6 405.1 220.9<br />

CALM1 801 ENSG00000198668 -2.30 3.99E-03 8.49E-02 2159.3 2745.1 824.4 1635.9 14162.8 2936.0<br />

CAMK2B 816 ENSG00000058404 Inf 3.26E-06 2.79E-04 124.2 187.0 5.5 0.0 0.0 0.0<br />

CAPN6 827 ENSG00000077274 -Inf 5.42E-07 5.75E-05 0.0 0.0 0.0 0.0 0.0 199.0<br />

CARD17,CASP1,CARD16 114769,440068,834 ENSG00000137752,ENSG00000204397 5.04 2.68E-05 1.60E-03 4.1 10.9 121.7 166.1 6.0 0.0<br />

CBLC 23624 ENSG00000142273 Inf 1.79E-03 4.67E-02 0.0 0.0 76.2 0.0 0.0 0.0<br />

CCDC129 223075 ENSG00000180347 -Inf 3.70E-07 4.29E-05 0.0 0.0 0.0 0.0 169.0 2.0<br />

CCDC48 79825 ENSG00000114654 -4.17 1.39E-03 3.81E-02 4.1 9.0 0.0 0.0 32.7 79.7<br />

CCDC64 92558 ENSG00000135127 6.38 2.45E-04 9.86E-03 0.0 0.0 38.2 88.5 0.0 1.0<br />

CCKBR 887 ENSG00000110148 4.37 3.25E-03 7.22E-02 50.3 187.2 0.6 0.0 6.1 0.0<br />

CCL2 6347 ENSG00000108691 8.81 4.58E-19 6.80E-15 13.3 0.0 826.6 13567.4 0.0 21.1<br />

236


A.1 Differential Expression Appendix<br />

Gene symbol(s) Entrez gene ID(s) Ensembl 56 gene ID(s) Differential expression results Normalised tag counts<br />

log 2(F C) p-value FDR G144ED G144 G166 G179 CB541 CB660<br />

CCL26 10344 ENSG00000006606 Inf 8.09E-11 2.07E-08 0.0 0.0 355.9 78.5 0.0 0.0<br />

CCL7 6354 ENSG00000108688 Inf 1.80E-05 1.16E-03 0.0 0.0 6.9 136.2 0.0 0.0<br />

CCND2 894 ENSG00000118971 -3.98 9.23E-07 9.27E-05 158.0 125.2 0.6 0.0 827.8 496.4<br />

CCNO 10309 ENSG00000152669 Inf 2.09E-06 1.88E-04 2.7 17.2 143.8 22.1 0.0 0.0<br />

CCNY 219771 ENSG00000108100 -2.29 3.93E-03 8.44E-02 265.6 136.3 189.6 199.7 1270.9 448.1<br />

CCR1 1230 ENSG00000163823 Inf 1.75E-03 4.58E-02 33.8 81.5 1.3 0.0 0.0 0.0<br />

CD248 57124 ENSG00000174807 -Inf 1.69E-03 4.45E-02 2.4 0.0 0.0 0.0 4.6 45.7<br />

CD55 1604 ENSG00000196352 6.82 1.95E-05 1.24E-03 0.0 0.0 101.3 71.7 0.0 1.0<br />

CD58 965 ENSG00000116815 3.26 2.05E-03 5.09E-02 40.4 36.8 120.4 165.3 17.5 5.0<br />

CD68 968 ENSG00000129226 3.41 1.53E-03 4.12E-02 4.1 2.4 107.8 180.8 11.8 6.5<br />

CD70 970 ENSG00000125726 Inf 1.78E-16 2.78E-13 0.0 0.0 1301.8 114.4 0.0 0.0<br />

CD74 972 ENSG00000019582 7.11 6.35E-16 7.87E-13 3942.1 2107.8 93.4 1427.2 8.4 9.1<br />

CD9 928 ENSG00000010278 4.87 1.74E-08 2.74E-06 1058.3 1082.4 396.5 189.3 4.8 32.9<br />

CD97 976 ENSG00000123146 3.56 5.05E-04 1.77E-02 78.3 70.3 116.2 101.6 0.0 16.4<br />

CDH13 1012 ENSG00000140945 Inf 2.65E-11 8.17E-09 847.0 494.6 4.8 73.0 0.0 0.0<br />

CDH19 28513 ENSG00000071991 Inf 1.50E-03 4.05E-02 143.8 86.4 0.0 0.0 0.0 0.0<br />

CDH6 1004 ENSG00000113361 -3.05 2.29E-03 5.53E-02 74.7 22.5 3.1 1.0 139.1 4.3<br />

CDHR1 92211 ENSG00000148600 6.68 5.61E-05 2.90E-03 23.9 48.4 46.3 61.5 0.0 1.5<br />

CDKN2A 1029 ENSG00000147889 7.23 1.35E-14 8.49E-12 815.1 848.2 1206.3 0.0 3.0 6.6<br />

CDKN2C 1031 ENSG00000123080 2.55 2.50E-03 5.90E-02 345.8 242.3 258.6 826.0 83.7 67.2<br />

CEBPB 1051 ENSG00000172216 3.29 1.33E-04 5.94E-03 106.7 155.6 552.5 270.9 36.7 30.2<br />

CGREF1 10669 ENSG00000138028 4.77 4.80E-03 9.75E-02 34.5 26.9 20.7 56.6 1.5 1.0<br />

CHCHD10 400916 ENSG00000241579,ENSG00000242131 3.70 1.31E-05 9.02E-04 844.2 713.0 523.9 457.9 28.4 58.8<br />

CHODL 140578 ENSG00000154645 -Inf 4.70E-03 9.64E-02 0.0 0.0 0.0 0.0 24.3 10.4<br />

CHRDL1 91851 ENSG00000101938 3.00 2.10E-03 5.16E-02 1410.3 1413.1 0.7 42.9 75.0 46.4<br />

CILP 8483 ENSG00000138615 -Inf 3.58E-04 1.36E-02 0.0 0.0 0.0 0.0 0.0 71.5<br />

CITED4 163732 ENSG00000179862 6.87 1.58E-05 1.05E-03 0.0 0.0 176.1 2.7 0.0 1.0<br />

CLDN10 9071 ENSG00000134873 Inf 2.43E-04 9.79E-03 25.7 15.6 57.0 27.9 0.0 0.0<br />

CLDN11 5010 ENSG00000013297 5.73 7.86E-11 2.05E-08 2597.6 1809.3 60.7 948.0 0.0 35.3<br />

CLDN3 1365 ENSG00000165215 Inf 4.40E-03 9.18E-02 0.0 0.0 55.8 9.0 0.0 0.0<br />

CMAH 8418 ENSG00000168405 5.82 2.97E-03 6.69E-02 0.0 0.0 3.1 83.2 0.0 1.0<br />

CMPK2 129607 ENSG00000134326 4.21 6.11E-05 3.13E-03 8.1 27.9 53.9 228.5 0.0 11.1<br />

CMTM5 116173 ENSG00000166091 5.35 1.50E-03 4.07E-02 70.1 116.5 0.0 6.6 0.0 2.0<br />

CMTM8 152189 ENSG00000170293 2.76 4.87E-03 9.83E-02 6.8 10.9 217.8 19.4 0.0 24.5<br />

CNKSR2 22866 ENSG00000149970 4.52 1.66E-04 7.15E-03 129.7 154.6 6.8 108.9 3.8 4.4<br />

CNTN1 1272 ENSG00000018236 Inf 5.11E-04 1.77E-02 0.0 0.0 10.5 81.1 0.0 0.0<br />

CNTN6 27255 ENSG00000134115 -Inf 6.87E-06 5.15E-04 0.0 0.0 0.0 0.0 78.3 46.6<br />

COL1A2 1278 ENSG00000164692 -3.11 4.70E-03 9.64E-02 95.9 3.0 642.9 200.5 0.0 4846.9<br />

COL21A1 81578 ENSG00000124749 4.23 8.43E-07 8.58E-05 0.0 0.0 6.7 880.4 0.0 31.3<br />

COL3A1 1281 ENSG00000168542 -6.48 1.64E-09 3.25E-07 7.7 6.5 0.0 33.9 0.8 2365.8<br />

COL4A6 1288 ENSG00000197565 -3.00 2.27E-03 5.50E-02 0.0 21.9 45.6 8.2 232.0 170.8<br />

COL8A1 1295 ENSG00000144810 -3.07 4.73E-04 1.69E-02 249.9 79.2 227.1 1177.6 6989.8 1335.1<br />

CPAMD8 27151 ENSG00000160111 -Inf 1.24E-03 3.53E-02 1.4 0.0 0.0 0.0 32.8 16.2<br />

CPLX2 10814 ENSG00000145920 6.38 2.39E-04 9.74E-03 13.1 18.7 0.0 108.2 0.0 1.1<br />

CPNE2 221184 ENSG00000140848 -2.19 2.61E-03 6.09E-02 487.9 433.9 77.3 138.2 1114.3 863.3<br />

CPNE5 57699 ENSG00000124772 -4.49 4.42E-04 1.61E-02 0.0 0.0 0.0 10.7 156.7 3.3<br />

CRB2 286204 ENSG00000148204 -4.11 2.21E-03 5.38E-02 8.1 3.7 0.6 7.5 64.3 63.5<br />

CREB3L1 90993 ENSG00000157613 -1.99 3.99E-03 8.49E-02 225.2 267.1 119.2 51.6 989.8 173.2<br />

CRIP2 1397 ENSG00000182809 -2.56 1.91E-03 4.86E-02 362.8 95.2 16.9 45.8 277.6 345.0<br />

CRYBB1 1414 ENSG00000100122 -Inf 9.38E-04 2.82E-02 0.0 0.0 0.0 0.0 42.3 8.4<br />

CRYBB2 1415 ENSG00000244752 Inf 1.85E-04 7.86E-03 1.4 0.0 106.5 0.0 0.0 0.0<br />

CRYM 1428 ENSG00000103316 -7.77 5.80E-10 1.23E-07 0.0 0.0 0.6 4.1 19.2 665.5<br />

237


A.1 Differential Expression Appendix<br />

Gene symbol(s) Entrez gene ID(s) Ensembl 56 gene ID(s) Differential expression results Normalised tag counts<br />

log 2(F C) p-value FDR G144ED G144 G166 G179 CB541 CB660<br />

CSGALNACT1 55790 ENSG00000147408 7.68 2.19E-07 2.65E-05 359.0 256.3 0.6 54.1 0.0 1.3<br />

CTLA4 1493 ENSG00000163599 -Inf 1.89E-03 4.84E-02 0.0 0.0 0.0 0.0 0.0 49.8<br />

CTNNA2 1496 ENSG00000066032 6.39 2.30E-06 2.05E-04 59.1 95.6 0.0 159.1 0.0 2.2<br />

CTSC 1075 ENSG00000109861 3.55 1.55E-05 1.03E-03 937.2 731.4 311.7 1499.9 10.3 133.2<br />

CXCL1 2919 ENSG00000163739 7.23 2.34E-13 9.96E-11 0.0 0.0 1.2 912.1 0.0 3.8<br />

CXCL12 6387 ENSG00000107562 2.86 3.24E-03 7.22E-02 2.7 0.0 62.2 215.3 0.0 24.7<br />

CXCL14 9547 ENSG00000145824 2.59 7.14E-04 2.27E-02 0.0 0.0 7.0 2853.2 0.0 313.6<br />

CXCL2 2920 ENSG00000081041 Inf 5.32E-11 1.44E-08 0.0 0.0 17.3 446.4 0.0 0.0<br />

CXCL3 2921 ENSG00000163734 7.86 1.87E-16 2.78E-13 0.0 0.0 29.7 2452.6 0.0 7.0<br />

CXCL6 6372 ENSG00000124875 6.13 3.56E-09 6.46E-07 0.0 0.0 5.6 529.8 0.0 5.1<br />

CXXC4 80319 ENSG00000168772 -5.50 1.94E-05 1.24E-03 18.1 5.8 0.0 0.0 78.3 110.7<br />

CXorf38 159013 ENSG00000185753 5.08 2.32E-03 5.57E-02 1.4 10.9 72.7 19.6 0.0 2.0<br />

CYB5R2 51700 ENSG00000166394 6.33 2.99E-04 1.17E-02 9.5 15.6 91.0 16.5 0.0 1.0<br />

DCBLD2 131566 ENSG00000057019 2.43 3.57E-03 7.77E-02 145.0 145.2 612.9 5664.5 263.6 529.0<br />

DCHS1 8642 ENSG00000166341 -3.37 1.38E-03 3.80E-02 91.8 21.1 3.5 10.7 73.8 176.8<br />

DCN 1634 ENSG00000011465 -Inf 1.87E-09 3.65E-07 0.0 0.0 0.0 0.0 0.0 401.7<br />

DDAH2 23564 ENSG00000213722 -3.46 1.51E-03 4.09E-02 128.6 23.3 49.4 2.7 37.0 510.7<br />

DDIT3 1649 ENSG00000175197 4.40 4.13E-07 4.59E-05 933.6 1345.3 748.9 250.5 26.9 47.3<br />

DDIT4L 115265 ENSG00000145358 -6.41 1.15E-03 3.31E-02 1.4 0.0 1.3 0.0 51.5 18.9<br />

DDO 8528 ENSG00000203797 Inf 2.79E-05 1.65E-03 0.0 17.1 29.5 88.4 0.0 0.0<br />

DHRS3 9249 ENSG00000162496 5.20 2.00E-09 3.87E-07 44.5 20.3 592.3 173.7 0.0 14.1<br />

DIAPH2 1730 ENSG00000147202 Inf 1.10E-04 5.09E-03 31.0 57.8 33.9 27.6 0.0 0.0<br />

DKFZp434H1419,AC012513.4 150967 ENSG00000226052 -2.12 4.68E-03 9.64E-02 34.6 71.8 0.0 28.0 239.5 50.3<br />

DKK1 22943 ENSG00000107984 Inf 1.16E-04 5.31E-03 0.0 0.0 46.5 64.9 0.0 0.0<br />

DLK1 8788 ENSG00000185559 -7.95 2.69E-11 8.17E-09 0.0 6.3 0.0 0.0 11.1 1017.5<br />

DNER 92737 ENSG00000187957 -3.89 3.03E-05 1.75E-03 90.5 523.2 9.3 847.4 8865.3 4813.8<br />

DNM3 26052 ENSG00000197959 4.45 4.19E-05 2.28E-03 115.8 128.5 4.6 233.7 6.1 5.0<br />

DOCK10 55619 ENSG00000135905 3.79 2.43E-05 1.48E-03 513.1 652.2 2.2 257.8 4.6 39.3<br />

DOCK5 80005 ENSG00000147459 -4.03 3.76E-04 1.42E-02 0.0 0.0 0.0 21.1 191.6 40.5<br />

DPY19L1 23333 ENSG00000173852 6.17 9.15E-06 6.64E-04 129.0 142.9 86.9 16.3 2.3 0.0<br />

DRD2 1813 ENSG00000149295 8.58 2.39E-10 5.55E-08 0.0 0.0 20.1 414.9 1.1 0.0<br />

DTX4 23220 ENSG00000110042 -5.92 4.13E-07 4.59E-05 6.8 7.8 3.2 0.0 128.8 312.5<br />

DUSP16 80824 ENSG00000111266 4.16 3.36E-03 7.40E-02 62.7 95.2 40.7 0.0 0.0 5.2<br />

DUSP5 1847 ENSG00000138166 3.87 4.66E-05 2.48E-03 130.7 410.4 181.5 18.5 10.4 16.9<br />

DYNC1I1 1780 ENSG00000158560 3.40 1.83E-03 4.73E-02 14.9 60.9 39.5 167.7 6.9 9.9<br />

ECHDC2 55268 ENSG00000121310 Inf 1.20E-05 8.39E-04 29.2 48.6 78.6 24.4 0.0 0.0<br />

EDA2R 60401 ENSG00000131080 -4.02 6.46E-04 2.12E-02 6.8 9.1 4.8 0.0 134.7 20.7<br />

EEF1A2 1917 ENSG00000101210 2.74 3.81E-03 8.23E-02 53.7 53.1 166.2 157.1 15.5 22.4<br />

EEF1D 1936 ENSG00000104529 5.12 3.08E-05 1.77E-03 669.4 175.8 0.0 87.7 0.0 4.9<br />

EFEMP1 2202 ENSG00000115380 2.85 2.27E-04 9.29E-03 4.1 3.1 551.2 613.9 2.4 104.6<br />

EFHD2 79180 ENSG00000142634 2.54 2.77E-03 6.38E-02 275.7 251.5 676.0 84.9 75.7 40.3<br />

EFNA5 1946 ENSG00000184349 -4.73 2.34E-03 5.61E-02 1.4 0.0 4.5 2.5 45.6 75.1<br />

EGFR 1956 ENSG00000146648 2.94 4.40E-03 9.18E-02 49.9 132.3 90.2 48.6 2.3 21.3<br />

ELMO1 9844 ENSG00000155849 4.43 4.57E-06 3.67E-04 642.9 731.1 0.6 128.0 11.4 15.1<br />

ELMO2 63916 ENSG00000062598 -4.56 1.78E-04 7.62E-03 5.5 0.0 11.4 3.3 224.0 4.7<br />

ELMOD1 55531 ENSG00000110675 3.24 2.81E-03 6.45E-02 273.5 272.5 1.9 124.8 21.9 6.1<br />

ELN 2006 ENSG00000049540 4.26 2.36E-06 2.09E-04 915.0 1024.5 2.9 136.1 0.0 40.7<br />

ELOVL2 54898 ENSG00000197977 -2.68 2.04E-04 8.54E-03 176.2 207.2 7.2 89.1 1134.0 167.8<br />

ELTD1 64123 ENSG00000162618 -Inf 1.52E-07 1.92E-05 0.0 0.0 0.0 0.0 184.9 5.0<br />

EML2 24139 ENSG00000125746 4.53 8.68E-05 4.20E-03 137.2 161.5 138.8 7.4 6.9 2.1<br />

EPAS1 2034 ENSG00000116016 -2.84 8.37E-05 4.09E-03 184.1 372.0 51.3 68.8 2186.6 166.7<br />

EPB41L3 23136 ENSG00000082397 2.93 4.86E-04 1.72E-02 243.2 268.9 0.0 487.4 2.3 63.5<br />

238


A.1 Differential Expression Appendix<br />

Gene symbol(s) Entrez gene ID(s) Ensembl 56 gene ID(s) Differential expression results Normalised tag counts<br />

log 2(F C) p-value FDR G144ED G144 G166 G179 CB541 CB660<br />

EPDR1 54749 ENSG00000086289 3.09 1.22E-04 5.55E-03 310.9 269.0 739.5 319.0 20.3 82.3<br />

EPHB3 2049 ENSG00000182580 -2.78 1.26E-04 5.69E-03 204.6 268.5 14.4 14.6 902.5 469.4<br />

ERBB4 2066 ENSG00000178568 -4.00 8.10E-05 3.97E-03 8.1 29.7 0.7 5.2 59.4 314.9<br />

ESRRG 2104 ENSG00000196482 Inf 4.35E-05 2.35E-03 36.2 39.4 1.4 86.1 0.0 0.0<br />

ETS1 2113 ENSG00000134954 5.23 1.13E-05 7.98E-04 121.4 201.8 19.4 92.2 1.3 4.0<br />

ETS2 2114 ENSG00000157557 2.83 7.06E-04 2.27E-02 169.8 124.6 235.9 202.8 0.0 52.4<br />

EVC2 132884 ENSG00000173040 Inf 2.87E-03 6.55E-02 8.1 3.1 39.5 28.7 0.0 0.0<br />

F12 2161 ENSG00000131187 4.30 3.11E-05 1.78E-03 181.6 247.2 84.6 66.2 2.3 10.9<br />

F2RL1 2150 ENSG00000164251 4.11 5.83E-04 1.95E-02 0.0 0.0 74.7 129.5 0.8 7.1<br />

FAM126A 84668 ENSG00000122591 2.84 1.16E-03 3.33E-02 1061.8 1338.9 374.2 276.1 82.2 102.7<br />

FAM129A 116496 ENSG00000135842 2.81 1.72E-03 4.52E-02 43.9 71.2 250.9 489.4 64.0 13.1<br />

FAM134B 54463 ENSG00000154153 9.61 1.37E-14 8.49E-12 886.3 1073.7 39.0 78.7 0.0 1.0<br />

FAM150B 285016 ENSG00000189292 -5.14 3.09E-04 1.19E-02 0.0 4.7 0.6 0.0 55.4 69.6<br />

FAM176A 84141 ENSG00000115363 3.37 2.00E-03 5.01E-02 6.0 34.8 47.6 165.8 3.8 12.4<br />

FAM181A 90050 ENSG00000140067 -Inf 2.48E-03 5.86E-02 4.1 0.0 0.0 0.0 0.8 45.6<br />

FAM184B 27146 ENSG00000047662 Inf 1.05E-03 3.11E-02 93.6 82.5 5.5 0.0 0.0 0.0<br />

FAM189A1 23359 ENSG00000104059 -Inf 3.09E-05 1.77E-03 0.0 0.0 0.0 0.0 78.2 18.2<br />

FAM196A,C10orf141 642938 ENSG00000188916 4.89 6.80E-06 5.13E-04 602.4 580.3 1.3 13.8 11.3 2.3<br />

FAM38B2,FAM38B,C18orf58 63895 ENSG00000154864,ENSG00000168738,ENSG00000175388 -Inf 1.16E-13 5.48E-11 0.0 0.0 0.0 0.0 778.6 166.8<br />

FAM55C,NFKBIZ 64332,91775 ENSG00000144802,ENSG00000144815 3.08 4.25E-04 1.58E-02 36.5 91.7 90.7 783.8 48.0 28.1<br />

FAM5B 57795 ENSG00000198797 2.65 2.97E-03 6.69E-02 13.7 34.4 0.6 356.7 0.0 41.3<br />

FAM69A 388650 ENSG00000154511 3.68 3.96E-05 2.18E-03 513.2 393.1 155.0 439.3 27.9 23.3<br />

FAM70A 55026 ENSG00000125355 6.01 1.99E-06 1.84E-04 202.7 114.1 15.7 163.8 0.0 3.5<br />

FAM84A,LOC653602 151354,653602 ENSG00000162981 5.14 4.32E-04 1.59E-02 74.7 110.8 5.4 44.3 0.0 2.7<br />

FBLN2 2199 ENSG00000163520 -Inf 9.47E-10 1.95E-07 0.0 0.0 0.0 0.0 232.1 117.5<br />

FBN2 2201 ENSG00000138829 -4.63 2.08E-04 8.65E-03 2.7 0.0 16.3 8.2 12.2 389.9<br />

FBXO27 126433 ENSG00000161243 5.09 1.64E-05 1.07E-03 0.0 0.0 64.5 195.6 0.0 4.9<br />

FBXO32 114907 ENSG00000156804 5.32 1.07E-10 2.71E-08 30.4 88.9 572.3 1235.4 17.5 14.1<br />

FCGR2B,FCGR2C,FCGR2A 2212,2213,9103 ENSG00000072694,ENSG00000143226,ENSG00000244682 6.26 1.33E-05 9.12E-04 338.7 219.8 13.2 0.0 0.0 2.2<br />

FCRLA 84824 ENSG00000132185 Inf 2.46E-15 2.61E-12 2134.8 1514.3 6.1 0.0 0.0 0.0<br />

FERMT3 83706 ENSG00000149781 4.18 2.53E-03 5.95E-02 2.5 5.2 13.9 119.9 0.0 5.0<br />

FEZF2 55079 ENSG00000153266 -Inf 1.45E-07 1.86E-05 0.0 0.0 0.0 0.0 29.7 196.7<br />

FGF19 9965 ENSG00000162344 -Inf 3.78E-07 4.32E-05 26.0 0.0 0.0 0.0 0.0 208.3<br />

FGFBP2 83888 ENSG00000137441 8.40 6.75E-14 3.72E-11 1699.4 939.1 0.6 84.3 0.0 2.0<br />

FGFR1 2260 ENSG00000077782 -1.90 4.51E-03 9.34E-02 1177.0 1288.2 147.2 311.0 3067.1 1294.6<br />

FMNL1 752 ENSG00000184922 3.74 4.24E-03 8.94E-02 0.0 0.0 47.7 95.0 0.0 6.6<br />

FOXG1 2290 ENSG00000176165 3.30 1.57E-04 6.83E-03 445.0 505.5 104.5 137.4 0.0 50.8<br />

FOXJ1 2302 ENSG00000129654 -4.19 7.16E-05 3.60E-03 4.9 3.1 4.4 27.8 254.8 175.8<br />

FOXQ1 94234 ENSG00000164379 -Inf 4.65E-04 1.66E-02 0.0 0.0 0.0 0.0 49.7 9.1<br />

FUT8 2530 ENSG00000033170 3.32 9.74E-05 4.63E-03 666.0 999.8 609.6 2352.7 95.8 168.6<br />

FXYD1 5348 ENSG00000221857 7.79 8.28E-12 2.86E-09 269.6 673.5 0.0 0.0 0.0 2.0<br />

FXYD3 5349 ENSG00000089356 Inf 6.70E-06 5.08E-04 110.9 180.1 0.0 0.0 0.0 0.0<br />

FXYD5 53827 ENSG00000089327 3.06 3.46E-04 1.32E-02 45.6 1.6 489.3 0.0 1.9 37.7<br />

FXYD7 53822 ENSG00000221946 Inf 1.11E-16 2.06E-13 1650.8 1205.4 6.9 705.9 0.0 0.0<br />

FZD3 7976 ENSG00000104290 -2.63 2.29E-03 5.53E-02 98.9 57.5 11.2 52.7 240.7 262.9<br />

GABBR2 9568 ENSG00000136928 -9.50 5.68E-17 1.21E-13 0.0 0.0 5.6 0.0 2569.7 143.1<br />

GABRA5 2558 ENSG00000186297 -6.00 1.45E-03 3.99E-02 8.1 1.7 0.0 0.0 0.9 65.5<br />

GABRQ 55879 ENSG00000147402 Inf 8.54E-05 4.15E-03 87.7 126.9 0.0 0.0 0.0 0.0<br />

GAL 51083 ENSG00000069482 Inf 1.01E-03 3.00E-02 0.0 0.0 0.0 82.6 0.0 0.0<br />

GALNT5 11227 ENSG00000136542 4.71 5.17E-04 1.78E-02 8.1 32.5 121.1 5.7 0.0 3.8<br />

GALR1 2587 ENSG00000166573 Inf 7.97E-04 2.45E-02 86.2 96.5 0.0 0.0 0.0 0.0<br />

GAS7 8522 ENSG00000007237 Inf 6.98E-08 9.52E-06 294.5 288.3 0.0 3.6 0.0 0.0<br />

239


A.1 Differential Expression Appendix<br />

Gene symbol(s) Entrez gene ID(s) Ensembl 56 gene ID(s) Differential expression results Normalised tag counts<br />

log 2(F C) p-value FDR G144ED G144 G166 G179 CB541 CB660<br />

GBP1 2633 ENSG00000117228 3.90 2.24E-04 9.22E-03 1.5 0.0 64.1 266.1 7.6 6.9<br />

GBP2 2634 ENSG00000162645 4.06 6.94E-06 5.18E-04 0.0 4.7 468.3 654.3 41.3 4.0<br />

GBP3 2635 ENSG00000117226 3.47 1.53E-04 6.69E-03 0.0 0.0 49.3 720.9 37.5 9.1<br />

GBP4 115361 ENSG00000162654 Inf 3.54E-04 1.35E-02 7.5 0.0 15.3 82.2 0.0 0.0<br />

GBP5 115362 ENSG00000154451 Inf 3.88E-09 6.87E-07 0.0 0.0 52.6 276.2 0.0 0.0<br />

GCNT1 2650 ENSG00000187210 Inf 4.77E-03 9.74E-02 5.5 1.6 15.0 46.2 0.0 0.0<br />

GDF15 9518 ENSG00000130513 4.37 3.15E-07 3.72E-05 288.9 1402.4 1358.2 108.3 22.8 69.9<br />

GDPD2 54857 ENSG00000130055 -3.12 7.12E-04 2.27E-02 32.7 37.2 0.0 4.2 133.1 108.5<br />

GEM 2669 ENSG00000164949 4.64 1.68E-06 1.57E-04 57.7 61.0 37.7 610.9 15.7 3.0<br />

GFAP 2670 ENSG00000131095 5.34 1.43E-06 1.37E-04 468.5 336.4 0.0 94.7 0.0 7.2<br />

GFPT2 9945 ENSG00000131459 2.25 4.84E-03 9.82E-02 24.3 15.6 247.6 243.2 0.0 70.6<br />

GGH 8836 ENSG00000137563 2.45 4.70E-03 9.64E-02 899.6 886.6 7220.3 4880.1 475.4 1096.6<br />

GJA1 2697 ENSG00000152661 -3.49 1.89E-04 8.00E-03 334.7 130.2 764.6 116.4 6363.9 1206.7<br />

GJB2 2706 ENSG00000165474 2.80 1.20E-03 3.42E-02 1.4 0.0 18.4 448.1 0.0 44.0<br />

GJC3 349149 ENSG00000176402 Inf 1.50E-03 4.05E-02 31.1 86.2 0.0 0.0 0.0 0.0<br />

GLYATL2 219970 ENSG00000156689 -3.99 2.64E-03 6.15E-02 39.2 9.5 0.0 0.0 25.8 73.6<br />

GMPR 2766 ENSG00000137198 3.72 2.86E-03 6.54E-02 108.1 140.1 33.5 35.5 4.5 6.3<br />

GNG11 2791 ENSG00000127920 -2.33 3.49E-03 7.64E-02 393.2 316.9 345.3 1039.6 4802.2 908.6<br />

GPC3 2719 ENSG00000147257 -5.79 1.24E-06 1.19E-04 0.0 0.0 35.1 0.0 11.6 1268.7<br />

GPNMB 10457 ENSG00000136235 6.74 1.68E-11 5.42E-09 7688.5 16473.1 127.8 224.6 82.2 23.2<br />

GPR158 57512 ENSG00000151025 -4.55 7.39E-08 9.89E-06 103.8 117.4 0.0 53.3 2311.0 354.9<br />

GPR98 84059 ENSG00000164199 -3.99 2.40E-04 9.74E-03 30.5 20.0 4.4 0.0 76.9 183.8<br />

GRB14 2888 ENSG00000115290 5.96 1.66E-03 4.39E-02 31.2 30.3 4.6 60.7 0.0 1.2<br />

GRIA1 2890 ENSG00000155511 -4.42 3.62E-07 4.24E-05 33.1 68.7 2.7 73.4 1780.2 292.7<br />

GRIA3 2892 ENSG00000125675 7.17 7.45E-05 3.71E-03 85.8 129.9 1.4 32.7 0.8 0.0<br />

GRIK3 2899 ENSG00000163873 5.24 3.84E-07 4.35E-05 580.1 631.6 0.0 0.0 0.0 11.1<br />

GRM3 2913 ENSG00000198822 -Inf 4.70E-08 6.47E-06 0.0 0.0 0.0 0.0 162.4 66.0<br />

GYG2 8908 ENSG00000056998 -2.89 5.71E-04 1.93E-02 52.6 51.3 3.8 0.8 223.4 55.4<br />

H1F0 3005 ENSG00000189060 3.59 9.61E-04 2.87E-02 93.7 46.8 89.0 103.7 0.0 13.5<br />

HAND2 9464 ENSG00000164107 Inf 1.11E-03 3.22E-02 75.0 84.1 1.0 2.9 0.0 0.0<br />

HAPLN1 1404 ENSG00000145681 6.40 4.55E-07 4.98E-05 521.8 383.7 0.0 0.0 0.0 3.0<br />

HCP5 10866 ENSG00000206337 Inf 8.81E-09 1.49E-06 0.0 0.0 180.4 114.3 0.0 0.0<br />

HEPACAM 220296 ENSG00000165478 Inf 3.91E-03 8.39E-02 82.3 65.6 0.0 3.0 0.0 0.0<br />

HIF3A 64344 ENSG00000124440 -Inf 1.27E-03 3.56E-02 0.0 0.0 0.0 0.0 15.2 36.3<br />

HLA-A 3105 ENSG00000206503 3.04 3.02E-04 1.18E-02 294.8 469.0 840.5 535.7 83.8 65.7<br />

HLA-DMA 3108 ENSG00000204257 Inf 8.98E-07 9.08E-05 383.9 196.7 13.7 7.4 0.0 0.0<br />

HLA-DPA1 3113 ENSG00000231389 5.73 5.76E-11 1.53E-08 1554.1 1033.9 49.5 243.3 4.6 12.4<br />

HLA-DPB1 3115 ENSG00000223865 3.83 4.38E-04 1.61E-02 1007.2 359.8 0.6 12.3 2.4 15.1<br />

HLA-DQA1 3117 ENSG00000196735 Inf 1.25E-03 3.53E-02 198.2 89.6 0.0 0.0 0.0 0.0<br />

HLA-DQA2 3118 ENSG00000237541 6.56 5.45E-07 5.75E-05 0.0 0.0 8.1 280.8 0.0 2.4<br />

HLA-DQB1 3119 ENSG00000179344 4.60 3.26E-05 1.84E-03 348.2 347.0 16.8 31.7 3.4 7.4<br />

HLA-DRA 3122 ENSG00000204287 5.68 3.67E-09 6.58E-07 9744.0 5434.9 138.4 270.5 57.4 19.2<br />

HLA-DRB3,HLA-DRB1,HLA- 3123,3125,3126 ENSG00000196126 Inf 2.04E-10 4.89E-08 1105.3 381.8 3.1 87.8 0.0 0.0<br />

240<br />

DRB4<br />

HLA-DRB5 3127 ENSG00000198502 8.36 1.38E-13 6.04E-11 2885.9 995.1 0.0 0.0 0.0 2.0<br />

HMGA2 8091 ENSG00000149948 -5.68 3.13E-07 3.72E-05 0.0 0.0 0.0 23.7 233.3 576.3<br />

HOMER1 9456 ENSG00000152413 6.06 4.09E-05 2.24E-03 94.6 173.2 28.9 0.0 0.0 2.0<br />

HOXA1 3198 ENSG00000105991 Inf 2.21E-03 5.39E-02 13.5 10.9 12.5 48.3 0.0 0.0<br />

HOXA10 3206 ENSG00000153807 Inf 8.63E-14 4.42E-11 479.4 631.7 19.5 273.5 0.0 0.0<br />

HOXA4 3201 ENSG00000197576 Inf 4.31E-11 1.26E-08 63.1 100.0 2.5 382.0 0.0 0.0<br />

HOXA5 3202 ENSG00000106004 Inf 3.20E-03 7.17E-02 22.7 33.6 2.5 33.1 0.0 0.0<br />

HOXA7 3204 ENSG00000122592 Inf 5.79E-04 1.94E-02 51.7 102.1 0.7 0.0 0.0 0.0


A.1 Differential Expression Appendix<br />

Gene symbol(s) Entrez gene ID(s) Ensembl 56 gene ID(s) Differential expression results Normalised tag counts<br />

log 2(F C) p-value FDR G144ED G144 G166 G179 CB541 CB660<br />

HOXB6 3216 ENSG00000108511 Inf 1.08E-03 3.16E-02 0.0 0.0 23.8 59.8 0.0 0.0<br />

HOXB7 3217 ENSG00000120087 Inf 2.51E-14 1.43E-11 0.0 0.0 795.3 109.6 0.0 0.0<br />

HOXB9 3219 ENSG00000170689 Inf 2.50E-10 5.72E-08 0.0 0.0 338.7 55.6 0.0 0.0<br />

HOXC10 3226 ENSG00000180818 Inf 1.74E-06 1.62E-04 90.5 160.7 47.1 0.0 0.0 0.0<br />

HOXC13 3229 ENSG00000123364 Inf 2.48E-03 5.86E-02 3.5 76.4 0.5 0.0 0.0 0.0<br />

HOXD10 3236 ENSG00000128710 Inf 7.25E-14 3.85E-11 77.0 145.9 115.3 578.2 0.0 0.0<br />

HOXD13 3239 ENSG00000128714 Inf 2.33E-05 1.44E-03 27.2 45.4 0.0 93.3 0.0 0.0<br />

HOXD3 3232 ENSG00000128652 Inf 4.05E-03 8.61E-02 49.4 40.5 1.5 25.7 0.0 0.0<br />

HOXD9 3235 ENSG00000128709 Inf 7.80E-04 2.42E-02 49.7 61.6 23.2 7.4 0.0 0.0<br />

HPRT1 3251 ENSG00000165704 2.62 6.73E-04 2.18E-02 28.4 82.7 614.7 246.9 4.3 96.9<br />

HPSE 10855 ENSG00000173083 6.28 4.17E-04 1.56E-02 10.8 16.0 18.2 84.8 0.0 1.0<br />

HRCT1 646962 ENSG00000196196 6.40 1.67E-03 4.42E-02 0.0 0.0 89.8 6.6 0.8 0.0<br />

HTATIP2 10553 ENSG00000109854 7.51 1.09E-14 7.73E-12 38.0 50.0 3.8 1331.9 0.0 4.7<br />

ICAM1 3383 ENSG00000090339 2.75 7.47E-04 2.35E-02 138.3 272.9 100.5 848.2 28.1 92.5<br />

ID4 3400 ENSG00000172201 -2.33 4.89E-04 1.73E-02 1342.6 1447.2 17.6 161.5 4504.8 958.5<br />

IFI27 3429 ENSG00000165949 3.86 2.55E-05 1.54E-03 11.8 7.4 446.9 38.2 2.3 20.5<br />

IFI30 10437 ENSG00000216490 4.27 2.84E-06 2.47E-04 25.7 40.6 470.2 277.2 16.3 11.1<br />

IFI6 2537 ENSG00000126709 3.36 2.69E-05 1.60E-03 110.0 57.7 2294.7 801.8 29.0 175.0<br />

IFITM2 10581 ENSG00000185201 -4.89 3.84E-05 2.13E-03 0.0 0.0 31.2 8.3 0.0 775.8<br />

IFITM8P ENSG00000215096 -Inf 2.27E-03 5.50E-02 0.0 0.0 0.0 0.0 0.6 46.3<br />

IGF2,INS,INS-<br />

3481,3630,723961 ENSG00000129965,ENSG00000167244,ENSG00000240801 -7.34 3.37E-08 4.77E-06 0.0 0.0 0.6 4.5 2.8 505.9<br />

241<br />

IGF2,AC132217.2<br />

IGFBP5 3488 ENSG00000115461 -2.23 4.78E-03 9.74E-02 840.8 936.7 30.1 1081.5 3234.4 3148.3<br />

IGLON5 402665 ENSG00000142549 5.38 1.58E-07 1.98E-05 750.3 630.9 0.0 0.0 0.0 10.1<br />

IGSF11 152404 ENSG00000144847 3.49 7.56E-04 2.37E-02 453.7 549.5 0.0 0.0 14.7 18.3<br />

IGSF3 3321 ENSG00000143061 3.78 5.11E-04 1.77E-02 301.4 260.7 41.9 45.7 6.9 10.0<br />

IKBKE 9641 ENSG00000143466 -4.25 2.18E-03 5.35E-02 7.4 2.3 2.1 3.3 82.9 22.2<br />

IL13RA2 3598 ENSG00000123496 5.66 2.22E-11 7.02E-09 0.0 0.0 451.0 628.7 0.0 14.3<br />

IL17RD 54756 ENSG00000144730 -4.38 1.14E-05 8.03E-04 25.5 17.5 9.8 15.3 364.6 230.4<br />

IL1B 3553 ENSG00000125538 Inf 1.93E-08 2.96E-06 0.0 0.0 115.5 175.1 0.0 0.0<br />

IL1R1 3554 ENSG00000115594 2.49 1.56E-03 4.18E-02 25.7 15.0 67.6 977.0 19.4 104.7<br />

IL1RAPL1 11141 ENSG00000169306 5.30 1.20E-03 3.42E-02 81.1 75.0 1.4 43.5 0.0 2.3<br />

IL33 90865 ENSG00000137033 -Inf 1.60E-03 4.26E-02 0.0 0.0 0.0 0.0 0.0 51.2<br />

IL4I1 259307 ENSG00000104951 6.01 1.36E-03 3.77E-02 2.7 2.1 12.6 84.2 0.0 1.0<br />

IL6 3569 ENSG00000136244 Inf 7.89E-07 8.09E-05 44.6 25.3 110.4 64.6 0.0 0.0<br />

IL8 3576 ENSG00000169429 7.68 4.39E-08 6.10E-06 0.0 0.0 26.4 286.9 0.0 1.5<br />

INHBA 3624 ENSG00000122641 7.38 1.07E-06 1.05E-04 86.5 172.1 66.0 16.5 0.0 1.0<br />

INHBB 3625 ENSG00000163083 5.89 2.05E-06 1.87E-04 240.6 361.5 0.0 0.0 0.0 4.1<br />

INPP5D 3635 ENSG00000168918 -3.50 4.59E-03 9.47E-02 6.8 10.9 3.8 0.0 61.6 49.5<br />

INSIG1 3638 ENSG00000186480 2.68 1.89E-03 4.84E-02 780.8 1475.9 1531.6 3387.9 282.2 383.3<br />

IRAK1 3654 ENSG00000184216 2.95 4.63E-04 1.66E-02 292.6 279.4 764.0 375.0 75.1 46.1<br />

IRX1 79192 ENSG00000170549 -3.75 4.11E-03 8.72E-02 30.5 11.4 0.0 0.0 5.1 92.7<br />

IRX2 153572 ENSG00000170561 -4.65 1.01E-05 7.21E-04 0.0 0.0 0.0 28.9 370.0 111.1<br />

ISYNA1 51477 ENSG00000105655 -2.54 3.24E-03 7.22E-02 308.0 123.3 127.2 149.7 668.8 883.1<br />

ITGA4 3676 ENSG00000115232 2.99 9.76E-04 2.91E-02 70.2 63.0 11.9 594.6 35.7 20.2<br />

ITGBL1 9358 ENSG00000198542 Inf 1.12E-03 3.24E-02 0.0 0.0 33.4 48.5 0.0 0.0<br />

ITIH5 80760 ENSG00000123243 5.87 3.70E-10 8.21E-08 1336.6 1302.0 2.8 1.4 3.6 10.7<br />

ITM2A 9452 ENSG00000078596 -Inf 4.47E-05 2.40E-03 0.0 0.0 0.0 0.0 0.0 105.8<br />

JAM2 58494 ENSG00000154721 -2.11 3.94E-03 8.44E-02 196.8 310.6 12.5 199.1 780.0 726.7<br />

JPH1 56704 ENSG00000104369 6.70 5.47E-05 2.84E-03 36.3 77.3 31.3 50.7 0.0 1.0<br />

KALRN 8997 ENSG00000160145 -5.25 2.09E-06 1.88E-04 0.0 6.3 2.5 7.1 302.9 106.2<br />

KANK1 23189 ENSG00000107104 2.97 8.79E-04 2.67E-02 374.9 583.2 79.6 135.3 14.5 53.9


A.1 Differential Expression Appendix<br />

Gene symbol(s) Entrez gene ID(s) Ensembl 56 gene ID(s) Differential expression results Normalised tag counts<br />

log 2(F C) p-value FDR G144ED G144 G166 G179 CB541 CB660<br />

KCNA2 3737 ENSG00000177301 Inf 4.29E-04 1.58E-02 149.2 103.9 0.0 2.4 0.0 0.0<br />

KCNJ12 3768 ENSG00000184185 Inf 4.57E-05 2.44E-03 60.5 139.1 0.0 0.0 0.0 0.0<br />

KCNK1 3775 ENSG00000135750 5.95 1.82E-03 4.72E-02 0.0 0.0 90.8 3.3 0.0 1.0<br />

KCNK12 56660 ENSG00000184261 -Inf 1.37E-03 3.79E-02 0.0 0.0 0.0 0.0 39.5 6.8<br />

KCNMB2 10242 ENSG00000197584 5.32 1.85E-05 1.19E-03 2.6 0.0 38.9 204.5 0.0 4.5<br />

KCTD12 115207 ENSG00000178695 -4.06 2.20E-06 1.97E-04 105.3 71.6 25.5 58.7 1598.8 150.0<br />

KIAA1217 56243 ENSG00000120549 4.49 1.62E-03 4.29E-02 35.7 52.3 81.5 4.0 0.0 3.6<br />

KLF6 1316 ENSG00000067082 4.56 5.25E-04 1.80E-02 58.2 113.0 67.2 0.0 0.0 5.4<br />

L3MBTL4 91133 ENSG00000154655 Inf 2.11E-10 4.97E-08 58.0 67.9 11.6 341.6 0.0 0.0<br />

LAMA2 3908 ENSG00000196569 -5.21 3.06E-08 4.42E-06 47.3 25.2 8.0 2.6 730.2 149.0<br />

LAMA4 3910 ENSG00000112769 2.81 2.23E-03 5.43E-02 885.8 984.7 31.7 116.1 38.0 69.6<br />

LEMD1 93273 ENSG00000186007 2.85 3.79E-04 1.43E-02 0.0 6.3 630.6 4.1 0.0 58.9<br />

LFNG 3955 ENSG00000106003 -2.21 2.21E-03 5.38E-02 66.1 96.7 6.9 49.2 429.5 42.4<br />

LGALS3 3958 ENSG00000131981 2.80 1.27E-03 3.56E-02 2898.0 3802.5 3936.6 643.9 219.8 578.3<br />

LHFPL3 375612 ENSG00000187416 Inf 2.25E-09 4.29E-07 485.5 402.6 0.0 0.0 0.0 0.0<br />

LIF 3976 ENSG00000128342 3.97 5.11E-06 3.99E-04 25.7 28.0 98.9 891.5 15.9 27.3<br />

LIFR 3977 ENSG00000113594 -2.95 1.06E-03 3.15E-02 181.0 124.8 38.0 727.7 1803.6 2775.9<br />

LINGO1 84894 ENSG00000169783 5.52 9.07E-04 2.74E-02 113.3 139.7 0.0 0.0 0.0 2.0<br />

LMO2 4005 ENSG00000135363 -4.82 2.89E-06 2.50E-04 20.3 21.2 0.0 4.9 134.6 369.0<br />

LMO3 55885 ENSG00000048540 -Inf 3.43E-11 1.02E-08 0.0 0.0 0.0 0.0 136.9 446.0<br />

LMO4 8543 ENSG00000143013 3.80 6.59E-06 5.03E-04 553.8 1311.2 455.5 1149.8 16.7 122.5<br />

LNX1 84708 ENSG00000072201 4.67 2.34E-03 5.61E-02 2.7 3.1 12.6 101.0 0.0 3.0<br />

LOC283070,CAMK1D 283070,57118 ENSG00000183049 -2.40 5.40E-04 1.84E-02 109.8 142.2 0.6 35.0 578.0 52.7<br />

LOXL4 84171 ENSG00000138131 4.94 1.10E-04 5.09E-03 186.4 184.3 32.9 15.6 0.0 4.7<br />

LPAR6 10161 ENSG00000139679 -3.62 1.24E-04 5.61E-03 29.8 33.8 39.6 0.0 395.9 211.3<br />

LPHN2 23266 ENSG00000117114 -2.05 2.04E-03 5.09E-02 268.8 205.3 10.0 12.7 446.7 183.5<br />

LRAT 9227 ENSG00000121207 5.42 6.40E-06 4.93E-04 0.0 0.0 1.9 290.9 1.5 3.0<br />

LRP4 4038 ENSG00000134569 -2.68 2.41E-03 5.73E-02 29.6 38.9 5.6 18.6 187.5 86.0<br />

LRRC2 79442 ENSG00000163827 Inf 4.14E-06 3.38E-04 0.0 0.0 169.1 0.0 0.0 0.0<br />

LRRC55 219527 ENSG00000183908 -Inf 1.89E-03 4.84E-02 0.0 0.0 0.0 0.0 0.0 49.6<br />

LRRN2 10446 ENSG00000170382 -4.91 1.54E-04 6.71E-03 7.8 5.5 0.0 0.0 97.9 27.3<br />

LTF 4057 ENSG00000012223 Inf 2.03E-07 2.47E-05 0.0 0.0 222.4 0.0 0.0 0.0<br />

LUM 4060 ENSG00000139329 -4.26 6.32E-05 3.23E-03 0.0 0.0 110.9 0.0 507.4 897.7<br />

LXN 56925 ENSG00000079257 -5.76 8.55E-05 4.15E-03 0.0 0.0 0.0 3.7 139.4 9.0<br />

LY96 23643 ENSG00000154589 7.53 1.62E-07 2.01E-05 3.4 59.4 106.5 115.6 0.0 1.0<br />

LYST 1130 ENSG00000143669 5.22 1.71E-07 2.10E-05 85.4 117.1 79.3 454.5 10.3 1.0<br />

MACROD2 140733 ENSG00000172264 -3.07 2.85E-03 6.53E-02 20.6 21.8 0.0 0.0 90.0 33.4<br />

MAF 4094 ENSG00000178573 -4.79 1.41E-05 9.55E-04 14.9 7.1 16.0 0.0 282.4 150.4<br />

MAL 4118 ENSG00000172005 6.24 2.59E-10 5.83E-08 22.8 23.3 11.9 655.2 0.0 6.0<br />

MALL 7851 ENSG00000144063 4.75 3.34E-08 4.77E-06 0.0 0.0 0.0 1117.4 5.4 22.2<br />

MAN1C1 57134 ENSG00000117643 3.59 3.15E-05 1.79E-03 556.5 478.6 107.6 378.3 7.6 45.2<br />

MAP3K5 4217 ENSG00000197442 5.06 1.46E-05 9.83E-04 43.3 28.1 110.6 178.0 5.0 1.0<br />

MAP6 4135 ENSG00000171533 -3.25 1.78E-04 7.62E-03 2.3 32.9 0.6 111.4 629.4 288.0<br />

MAP7 9053 ENSG00000135525 4.45 6.64E-04 2.17E-02 0.0 1.6 25.8 139.8 0.0 5.0<br />

MAPT 4137 ENSG00000186868 4.55 8.34E-06 6.10E-04 353.1 572.9 3.3 34.4 5.3 12.2<br />

MARCH1 55016 ENSG00000145416 6.53 1.27E-04 5.70E-03 18.9 37.1 61.6 41.7 0.0 1.1<br />

MARS 4141 ENSG00000166986 2.80 1.53E-03 4.12E-02 1181.8 771.8 272.0 1.3 39.5 60.2<br />

MARVELD3 91862 ENSG00000140832 -Inf 3.77E-04 1.42E-02 0.3 0.0 0.0 0.0 57.1 3.3<br />

MATN2 4147 ENSG00000132561 3.44 4.46E-04 1.62E-02 326.2 720.9 35.9 6.6 22.8 24.1<br />

MBP 4155 ENSG00000197971 6.47 7.66E-12 2.71E-09 118.6 402.5 103.3 303.0 0.0 6.3<br />

MEIS2 4212 ENSG00000134138 3.05 6.37E-04 2.11E-02 69.3 85.9 36.6 331.3 0.0 36.3<br />

MEST 4232 ENSG00000106484 -2.13 3.82E-03 8.25E-02 1932.6 895.3 66.5 334.9 1665.0 2122.4<br />

242


A.1 Differential Expression Appendix<br />

Gene symbol(s) Entrez gene ID(s) Ensembl 56 gene ID(s) Differential expression results Normalised tag counts<br />

log 2(F C) p-value FDR G144ED G144 G166 G179 CB541 CB660<br />

MET 4233 ENSG00000105976 9.09 1.36E-18 1.01E-14 1.4 0.0 43.8 9851.7 12.2 0.0<br />

MFAP2 4237 ENSG00000117122 -3.26 2.73E-03 6.32E-02 12.0 6.2 124.4 55.3 3.4 1174.2<br />

MGC87042 256227 ENSG00000105889 Inf 4.14E-03 8.76E-02 65.1 67.1 1.3 0.0 0.0 0.0<br />

MGLL 11343 ENSG00000074416 5.05 1.96E-04 8.25E-03 191.4 165.8 30.8 17.4 2.6 2.0<br />

MIA 8190 ENSG00000213054 5.64 5.51E-04 1.87E-02 21.5 151.9 0.0 0.0 0.0 2.0<br />

MICA 4276 ENSG00000204520 3.69 1.56E-05 1.03E-03 0.0 3.1 1713.4 98.0 65.6 28.8<br />

MICB 4277 ENSG00000204516 4.20 4.73E-03 9.68E-02 0.0 0.0 27.2 85.6 0.0 4.0<br />

MID1IP1 58526 ENSG00000165175 6.08 8.98E-04 2.72E-02 12.1 1.6 101.3 0.0 0.0 1.0<br />

MIPOL1 145282 ENSG00000151338 -Inf 7.14E-04 2.27E-02 0.0 0.0 0.0 0.0 6.3 54.5<br />

MKI67 4288 ENSG00000148773 -Inf 2.65E-04 1.06E-02 0.0 0.0 0.0 0.0 63.7 0.0<br />

MLC1 23209 ENSG00000100427 -2.52 6.39E-04 2.11E-02 207.8 109.7 22.5 12.3 495.8 59.5<br />

MLPH 79083 ENSG00000115648 2.92 2.65E-03 6.15E-02 0.0 0.0 27.7 267.6 1.5 24.7<br />

MMP17 4326 ENSG00000198598 3.58 1.55E-05 1.03E-03 1173.5 864.3 422.6 340.8 5.1 84.8<br />

MMP7 4316 ENSG00000137673 5.74 1.01E-04 4.75E-03 0.0 0.0 17.5 146.3 0.0 2.3<br />

MMRN1 22915 ENSG00000138722 -8.80 1.01E-08 1.65E-06 0.0 0.0 1.3 0.0 279.4 93.2<br />

MN1 4330 ENSG00000169184 -7.23 1.51E-09 3.03E-07 21.6 4.2 0.6 0.0 146.8 384.1<br />

MOCOS 55034 ENSG00000075643 4.47 3.85E-06 3.16E-04 14.1 75.8 201.7 195.0 0.0 14.0<br />

MOSC2 54996 ENSG00000117791 4.40 1.96E-04 8.25E-03 39.5 69.7 77.7 77.9 0.0 7.1<br />

MT1A 4489 ENSG00000205362 Inf 7.71E-04 2.40E-02 1.8 17.4 41.7 25.4 0.0 0.0<br />

MT2A 4502 ENSG00000125148 4.13 2.03E-05 1.28E-03 2039.2 1924.4 8308.0 5237.5 285.0 301.4<br />

MTTP 4547 ENSG00000138823 -3.36 8.32E-04 2.54E-02 6.5 6.3 7.2 27.0 249.5 25.2<br />

MX1 4599 ENSG00000157601 7.05 5.50E-06 4.28E-04 45.1 82.6 86.3 31.8 0.0 0.9<br />

MXRA5 25878 ENSG00000101825 -3.18 1.27E-03 3.56E-02 46.1 23.4 0.0 23.0 123.2 158.0<br />

MYBL1 4603 ENSG00000185697 3.44 4.68E-05 2.49E-03 87.8 81.2 162.7 765.9 17.4 44.4<br />

MYC 4609 ENSG00000136997 3.95 7.75E-06 5.72E-04 155.1 341.8 478.0 32.0 11.4 24.8<br />

MYH3 4621 ENSG00000109063 -4.54 2.06E-05 1.30E-03 18.9 17.2 16.5 51.9 11.8 1298.9<br />

MYL1 4632 ENSG00000168530 -Inf 1.75E-08 2.74E-06 0.0 0.0 0.0 0.0 6.6 310.5<br />

MYL9 10398 ENSG00000101335 -3.85 8.93E-05 4.28E-03 3.8 3.1 110.0 8.5 796.2 370.3<br />

MYO1B 4430 ENSG00000128641 -3.03 1.10E-03 3.19E-02 13.5 0.0 55.3 111.5 514.9 387.9<br />

NAMPT 10135 ENSG00000105835 2.92 5.09E-04 1.77E-02 499.3 678.3 484.4 1970.5 117.5 158.3<br />

NBL1 4681 ENSG00000158747 2.84 7.32E-04 2.32E-02 584.5 491.1 763.1 2068.2 156.4 151.9<br />

NCALD 83988 ENSG00000104490 -2.50 4.45E-03 9.25E-02 53.2 51.5 0.0 4.9 86.7 126.4<br />

NCAM1 4684 ENSG00000149294 3.13 8.63E-04 2.63E-02 3171.6 3747.4 36.2 1733.8 232.8 187.5<br />

NDE1 54820 ENSG00000072864 3.42 4.84E-04 1.72E-02 195.9 200.2 200.6 102.0 24.5 6.9<br />

NDN 4692 ENSG00000182636 -3.46 4.28E-05 2.32E-03 105.3 84.2 0.0 0.0 214.4 405.4<br />

NEFM 4741 ENSG00000104722 -Inf 1.35E-10 3.34E-08 0.0 0.0 0.0 0.0 0.0 536.3<br />

NELL2 4753 ENSG00000184613 -3.24 2.99E-04 1.17E-02 117.2 174.9 0.7 378.4 1070.5 2418.9<br />

NFASC 23114 ENSG00000163531 5.94 2.04E-03 5.08E-02 16.7 25.2 11.9 56.8 0.0 1.0<br />

NFATC2 4773 ENSG00000101096 Inf 1.18E-03 3.37E-02 74.3 83.5 4.4 0.0 0.0 0.0<br />

NFE2L3 9603 ENSG00000050344 3.89 1.43E-04 6.37E-03 43.2 51.1 49.9 215.7 0.0 14.3<br />

NFIA 4774 ENSG00000162599 2.95 3.06E-03 6.87E-02 220.0 429.2 10.7 9.8 1.5 37.4<br />

NID1 4811 ENSG00000116962 2.90 4.46E-04 1.62E-02 954.4 470.8 328.9 2577.4 96.8 204.4<br />

NKX2-1 7080 ENSG00000136352 -5.52 1.01E-08 1.65E-06 4.1 17.2 0.0 0.0 425.3 103.6<br />

NKX6-2 84504 ENSG00000148826 -Inf 1.47E-03 4.02E-02 0.0 0.0 0.0 0.0 0.0 52.2<br />

NLGN4X 57502 ENSG00000146938 3.12 4.96E-04 1.75E-02 570.9 700.9 8.1 162.0 8.4 58.2<br />

NMU 10874 ENSG00000109255 4.32 5.32E-04 1.82E-02 8.1 20.7 153.8 8.9 0.0 6.1<br />

NNAT 4826 ENSG00000053438 -Inf 4.86E-09 8.40E-07 0.0 0.0 0.0 0.0 0.0 363.2<br />

NNMT 4837 ENSG00000166741 3.74 6.59E-06 5.03E-04 519.9 172.4 918.1 175.6 15.2 47.4<br />

NOP16 51491 ENSG00000048162 2.60 1.91E-03 4.86E-02 305.8 448.6 493.2 198.3 57.0 67.7<br />

NOTCH3 4854 ENSG00000074181 -4.24 5.14E-04 1.78E-02 0.0 0.0 0.0 25.1 0.0 317.9<br />

NOV 4856 ENSG00000136999 Inf 4.28E-06 3.47E-04 265.6 174.9 9.8 0.0 0.0 0.0<br />

NPFFR2 10886 ENSG00000056291 -Inf 9.86E-09 1.65E-06 0.0 0.0 0.0 0.0 250.3 5.0<br />

243


A.1 Differential Expression Appendix<br />

Gene symbol(s) Entrez gene ID(s) Ensembl 56 gene ID(s) Differential expression results Normalised tag counts<br />

log 2(F C) p-value FDR G144ED G144 G166 G179 CB541 CB660<br />

NPTX2 4885 ENSG00000106236 -6.47 1.11E-08 1.78E-06 0.0 0.0 6.9 4.8 422.0 272.4<br />

NR0B1 190 ENSG00000169297 Inf 6.79E-15 5.60E-12 0.0 0.0 0.0 1039.8 0.0 0.0<br />

NR1D1 9572 ENSG00000126368 2.86 1.07E-03 3.15E-02 80.9 136.8 469.2 220.4 51.7 24.2<br />

NR4A2 4929 ENSG00000153234 Inf 4.54E-04 1.64E-02 86.9 79.7 4.4 14.6 0.0 0.0<br />

NRBF2 29982 ENSG00000148572 -Inf 2.85E-04 1.12E-02 0.0 0.0 0.0 0.0 12.9 59.5<br />

NRG1 3084 ENSG00000157168 Inf 2.27E-04 9.29E-03 2.7 9.7 60.4 30.0 0.0 0.0<br />

NRN1 51299 ENSG00000124785 Inf 6.59E-09 1.13E-06 317.7 235.8 49.5 48.5 0.0 0.0<br />

NRP2 8828 ENSG00000118257 2.86 4.26E-04 1.58E-02 33.7 60.1 169.0 901.9 26.6 76.3<br />

NTN1 9423 ENSG00000065320 -4.22 1.58E-06 1.48E-04 95.8 239.9 22.4 250.6 5458.0 966.2<br />

NTRK2 4915 ENSG00000148053 3.61 4.89E-05 2.57E-03 90.7 163.9 174.8 219.5 0.0 30.3<br />

NXPH1 30010 ENSG00000122584 4.57 4.70E-05 2.49E-03 325.0 414.1 0.0 26.6 8.4 4.0<br />

OAS1 4938 ENSG00000089127 6.47 1.46E-04 6.41E-03 0.0 0.0 53.9 81.2 0.0 1.4<br />

OAS2 4939 ENSG00000111335 Inf 2.18E-05 1.36E-03 0.0 0.0 73.4 65.7 0.0 0.0<br />

OAS3 4940 ENSG00000111331 3.88 1.48E-03 4.03E-02 2.7 6.5 55.6 162.4 9.1 1.0<br />

OASL 8638 ENSG00000135114 Inf 2.20E-08 3.30E-06 1.2 7.8 102.4 180.2 0.0 0.0<br />

ODZ2 57451 ENSG00000145934 -3.68 1.49E-04 6.51E-03 27.0 25.0 2.5 36.0 212.8 329.5<br />

OGN 4969 ENSG00000106809 -Inf 3.58E-04 1.36E-02 0.0 0.0 0.0 0.0 0.0 71.6<br />

OLFM1 10439 ENSG00000130558 4.72 2.31E-04 9.44E-03 200.9 227.7 11.3 0.0 0.0 6.0<br />

OTOR 56914 ENSG00000125879 9.92 3.31E-15 3.28E-12 1169.3 1474.1 0.0 0.0 0.0 1.0<br />

OTX2 5015 ENSG00000165588 -Inf 6.59E-04 2.16E-02 0.0 0.0 0.0 0.0 40.1 14.8<br />

OXTR 5021 ENSG00000180914 3.67 1.99E-03 5.00E-02 28.0 111.2 36.0 94.6 7.6 4.6<br />

P2RX7 5027 ENSG00000089041 7.72 1.34E-11 4.42E-09 691.5 611.8 25.5 2.2 0.0 2.0<br />

PARP12 64761 ENSG00000059378 2.92 1.36E-03 3.77E-02 18.2 21.9 196.2 300.2 24.5 21.2<br />

PARP3 10039 ENSG00000041880 4.12 3.46E-05 1.93E-03 5.4 25.0 145.0 267.2 7.7 9.3<br />

PAX8 7849 ENSG00000125618 -2.80 1.78E-03 4.66E-02 0.0 0.0 40.1 66.0 438.3 53.5<br />

PCDH20 64881 ENSG00000197991 6.16 1.08E-05 7.67E-04 65.1 7.8 1.3 208.6 0.0 2.0<br />

PCDHB12 56124 ENSG00000120328 -7.05 2.05E-03 5.09E-02 2.7 0.0 0.7 0.0 31.9 23.1<br />

PCDHB3 56132 ENSG00000113205 5.39 1.08E-03 3.16E-02 90.5 107.6 0.0 35.2 2.3 0.0<br />

PCDHB4 56131 ENSG00000081818 5.88 2.01E-06 1.85E-04 337.8 351.5 5.6 0.0 0.0 4.0<br />

PCOLCE2 26577 ENSG00000163710 5.77 4.31E-03 9.05E-02 0.0 0.0 3.7 79.4 0.0 1.3<br />

PDE1C 5137 ENSG00000154678 4.13 1.38E-06 1.32E-04 578.2 430.3 1117.8 981.4 45.7 50.8<br />

PDE4B 5142 ENSG00000184588 -5.35 2.42E-03 5.74E-02 9.5 1.6 0.6 0.0 42.5 17.2<br />

PDGFA 5154 ENSG00000197461 3.58 1.02E-04 4.80E-03 1540.7 1178.9 41.4 32.8 20.8 49.3<br />

PDGFRA 5156 ENSG00000134853 7.42 1.03E-14 7.67E-12 2309.4 2706.9 66.4 355.6 0.0 12.1<br />

PDGFRB 5159 ENSG00000113721 -3.44 1.87E-03 4.81E-02 34.3 18.8 29.9 3.3 58.0 316.0<br />

PDPN 10630 ENSG00000162493 4.68 4.08E-08 5.72E-06 1470.9 1283.2 107.6 2041.3 12.9 75.3<br />

PDZRN3 23024 ENSG00000121440 -4.40 1.38E-05 9.38E-04 18.4 22.1 12.6 3.0 278.2 251.3<br />

PEA15 8682 ENSG00000162734 -2.44 2.01E-03 5.04E-02 1189.8 1377.8 590.8 1060.2 9727.1 1297.6<br />

PERP 64065 ENSG00000112378 3.83 4.28E-04 1.58E-02 1.4 0.0 200.3 108.2 11.4 3.3<br />

PHACTR3 116154 ENSG00000087495 Inf 2.70E-04 1.07E-02 44.0 117.1 0.0 0.0 0.0 0.0<br />

PHLPP1 23239 ENSG00000081913 -2.64 8.54E-04 2.61E-02 469.3 331.2 166.3 100.8 1557.1 924.7<br />

PI15 51050 ENSG00000137558 -6.45 4.25E-10 9.29E-08 20.2 3.3 2.5 5.5 397.8 262.2<br />

PIGA 5277 ENSG00000165195 3.43 1.79E-03 4.67E-02 13.9 10.3 182.7 97.2 16.1 2.0<br />

PION 54103 ENSG00000186088 Inf 7.59E-05 3.75E-03 7.3 18.7 13.2 87.4 0.0 0.0<br />

PITPNC1 26207 ENSG00000154217 3.72 5.25E-04 1.80E-02 293.0 215.2 39.5 54.9 1.7 13.7<br />

PITX1 5307 ENSG00000069011 6.80 7.33E-13 2.94E-10 1.4 3.1 480.6 531.7 6.2 0.0<br />

PKNOX2 63876 ENSG00000165495 -2.77 3.36E-03 7.40E-02 62.9 32.3 1.5 1.9 109.0 53.4<br />

PLA2G2A 5320 ENSG00000188257 Inf 3.34E-18 1.24E-14 1227.8 4419.1 0.6 6.3 0.0 0.0<br />

PLA2G4A 5321 ENSG00000116711 7.34 1.68E-15 1.92E-12 909.7 1226.5 483.7 755.9 0.0 10.1<br />

PLAC9 219348 ENSG00000189129 2.81 2.96E-03 6.69E-02 182.5 578.2 0.6 18.7 0.0 56.6<br />

PLCB1 23236 ENSG00000182621 -2.27 2.87E-03 6.55E-02 103.0 73.1 8.4 53.8 366.7 70.9<br />

PLCH1 23007 ENSG00000114805 -3.41 4.41E-05 2.38E-03 152.0 134.0 83.3 118.9 2002.9 384.3<br />

244


A.1 Differential Expression Appendix<br />

Gene symbol(s) Entrez gene ID(s) Ensembl 56 gene ID(s) Differential expression results Normalised tag counts<br />

log 2(F C) p-value FDR G144ED G144 G166 G179 CB541 CB660<br />

PLD3 23646 ENSG00000105223 -3.31 2.42E-04 9.76E-03 228.5 33.1 87.5 9.8 700.5 162.0<br />

PLEKHA6 22874 ENSG00000143850 -5.40 3.21E-03 7.18E-02 6.8 0.0 0.6 1.6 39.9 24.7<br />

PLS1 5357 ENSG00000120756 2.94 9.39E-04 2.82E-02 67.5 149.4 218.6 344.8 36.5 25.3<br />

PLS3 5358 ENSG00000102024 3.83 3.67E-06 3.06E-04 360.2 776.6 476.6 367.2 0.0 75.6<br />

PLSCR1 5359 ENSG00000188313 2.45 4.58E-03 9.47E-02 37.0 137.7 307.6 533.6 81.2 38.1<br />

PMEPA1 56937 ENSG00000124225 4.36 4.99E-07 5.41E-05 240.8 123.3 621.8 297.2 11.7 22.1<br />

PMP2 5375 ENSG00000147588 Inf 9.08E-15 7.10E-12 948.1 1312.7 0.0 0.0 0.0 0.0<br />

PNMA2 10687 ENSG00000240694 -2.28 1.93E-03 4.90E-02 155.7 183.7 8.8 368.1 1517.6 305.5<br />

PNP 4860 ENSG00000198805 3.92 3.87E-03 8.32E-02 36.5 20.7 115.6 8.3 2.4 3.7<br />

PPAP2C 8612 ENSG00000141934 7.37 4.02E-07 4.52E-05 21.6 6.3 242.9 3.1 0.0 1.2<br />

PPCS 79717 ENSG00000127125 2.94 3.03E-04 1.18E-02 57.1 106.2 771.9 214.9 32.7 61.6<br />

PPEF1 5475 ENSG00000086717 4.44 6.87E-04 2.21E-02 12.2 165.1 9.6 23.0 0.0 6.4<br />

PPHLN1 51535 ENSG00000134283 3.56 7.59E-04 2.37E-02 98.5 165.8 119.1 6.6 2.6 14.1<br />

PPL 5493 ENSG00000118898 2.45 4.84E-03 9.82E-02 2.7 1.6 90.7 358.0 11.2 43.8<br />

PPM1K 152926 ENSG00000163644 5.15 1.93E-03 4.90E-02 16.2 28.1 56.3 23.6 0.0 2.4<br />

PPP4R1 9989 ENSG00000154845 -2.16 2.30E-03 5.54E-02 168.6 215.1 42.9 218.4 1239.4 180.3<br />

PRIMA1 145270 ENSG00000175785 6.84 2.97E-09 5.51E-07 865.4 465.5 0.0 54.9 0.0 3.0<br />

PROCR 10544 ENSG00000101000 4.09 1.87E-03 4.81E-02 5.5 0.0 181.8 0.0 6.3 1.5<br />

PRSS12 8492 ENSG00000164099 3.66 1.95E-03 4.92E-02 128.0 49.8 55.6 87.1 0.0 10.1<br />

PSORS1C1 170679 ENSG00000204540 Inf 1.11E-07 1.43E-05 0.0 0.0 212.3 29.0 0.0 0.0<br />

PSPH 5723 ENSG00000146733 3.45 1.87E-03 4.81E-02 40.0 60.0 168.5 1.9 4.0 10.2<br />

PTEN 5728 ENSG00000171862 -5.00 5.70E-05 2.94E-03 2.7 0.0 14.8 0.0 191.7 126.9<br />

PTGS1 5742 ENSG00000095303 -3.91 4.31E-03 9.05E-02 12.2 6.8 1.6 0.0 61.9 20.2<br />

PTPRD 5789 ENSG00000153707 -3.18 2.97E-03 6.69E-02 2.8 20.1 7.0 0.0 101.0 63.7<br />

PTPRH 5794 ENSG00000080031 2.67 3.36E-03 7.40E-02 13.5 2.0 162.7 371.3 35.8 19.7<br />

PTPRR 5801 ENSG00000153233 5.05 2.78E-03 6.42E-02 0.0 0.0 12.8 88.6 0.0 2.0<br />

PXDN 7837 ENSG00000130508 3.01 6.69E-04 2.18E-02 1025.9 1801.8 163.8 146.9 41.9 132.8<br />

PYGL 5836 ENSG00000100504 3.71 6.76E-05 3.41E-03 569.0 837.9 179.4 74.3 36.2 19.4<br />

RAB11FIP1 80223 ENSG00000156675 -3.57 4.79E-03 9.75E-02 19.1 6.9 19.0 0.0 56.8 140.1<br />

RAB38 23682 ENSG00000123892 3.17 2.79E-04 1.10E-02 137.3 221.7 53.7 963.8 58.6 33.4<br />

RAB6B 51560 ENSG00000154917 -3.75 8.86E-05 4.27E-03 36.5 22.8 10.7 8.2 303.4 76.3<br />

RAB7L1 8934 ENSG00000117280 6.23 5.37E-04 1.83E-02 82.1 62.9 52.1 0.0 0.0 1.0<br />

RAD50 10111 ENSG00000113522 3.49 3.57E-03 7.77E-02 25.7 49.4 70.3 84.2 6.3 5.7<br />

RADIL 55698 ENSG00000157927 3.00 4.70E-03 9.64E-02 113.9 92.3 13.5 137.8 0.0 20.3<br />

RARRES1 5918 ENSG00000118849 6.00 5.06E-11 1.42E-08 5.6 0.0 16.6 1153.4 6.1 6.3<br />

RARRES3 5920 ENSG00000133321 3.53 2.08E-04 8.65E-03 64.8 26.5 64.7 471.8 24.3 8.1<br />

RASGEF1C 255426 ENSG00000146090 4.10 3.55E-03 7.75E-02 471.5 143.6 2.8 9.8 0.0 6.0<br />

RASGRP3 25780 ENSG00000152689 Inf 2.81E-03 6.45E-02 0.0 5.2 26.1 38.5 0.0 0.0<br />

REC8 9985 ENSG00000100918 -2.64 1.11E-03 3.22E-02 99.8 91.8 23.9 125.9 633.6 373.3<br />

RGAG4 340526 ENSG00000242732 -3.26 7.97E-04 2.45E-02 21.3 18.6 28.2 19.8 267.2 157.6<br />

RGL3 57139 ENSG00000205517 Inf 3.90E-04 1.47E-02 10.2 4.7 61.0 30.3 0.0 0.0<br />

RGS17 26575 ENSG00000091844 4.76 7.40E-04 2.33E-02 84.3 107.6 5.6 50.7 0.0 4.2<br />

RGS20 8601 ENSG00000147509 -3.16 4.95E-03 9.97E-02 0.0 0.0 7.1 25.6 136.0 56.5<br />

RGS5 8490 ENSG00000143248 -4.82 3.40E-06 2.87E-04 4.1 13.5 6.9 3.0 343.5 112.5<br />

RHBDF2 79651 ENSG00000129667 3.41 3.12E-05 1.78E-03 9.5 18.8 214.9 956.3 10.7 63.6<br />

RHOBTB3 22836 ENSG00000164292 2.77 2.60E-03 6.09E-02 3292.5 2727.3 146.6 130.6 90.0 203.9<br />

RHPN1 114822 ENSG00000158106 -4.18 2.88E-03 6.56E-02 21.7 5.9 0.0 0.0 40.3 35.3<br />

RIPK4 54101 ENSG00000183421 6.16 6.76E-04 2.19E-02 2.4 0.0 50.8 58.0 0.0 1.0<br />

RNASEH1 246243 ENSG00000171865 2.43 4.37E-03 9.14E-02 68.9 165.9 213.0 440.5 48.5 52.6<br />

RNF175 285533 ENSG00000145428 6.81 5.25E-05 2.74E-03 225.4 165.0 5.1 0.0 0.0 1.1<br />

RP11-473I1.1 ENSG00000220793 2.89 5.13E-04 1.78E-02 73.0 177.2 882.0 3.2 48.9 46.0<br />

RP11-93B14.2,hCG_2018279 100127888 ENSG00000232803 Inf 3.11E-03 6.99E-02 41.3 72.1 1.3 0.0 0.0 0.0<br />

245


A.1 Differential Expression Appendix<br />

Gene symbol(s) Entrez gene ID(s) Ensembl 56 gene ID(s) Differential expression results Normalised tag counts<br />

log 2(F C) p-value FDR G144ED G144 G166 G179 CB541 CB660<br />

RP5-955M13.1,KCNG1 3755 ENSG00000026559,ENSG00000242964 3.17 4.85E-03 9.82E-02 31.5 43.7 73.1 75.4 0.0 14.1<br />

RP9 6100 ENSG00000164610 2.59 3.34E-03 7.40E-02 569.1 650.0 335.5 82.3 64.7 53.8<br />

RPSAP52 204010 ENSG00000241749 4.40 2.37E-03 5.66E-02 0.0 0.0 12.2 116.6 0.0 4.2<br />

RRAD 6236 ENSG00000166592 Inf 2.54E-09 4.79E-07 0.0 0.0 84.0 253.0 0.0 0.0<br />

RRP7A 27341 ENSG00000189306 3.00 2.73E-03 6.32E-02 78.1 35.6 263.9 2.0 9.9 15.1<br />

RRS1 23212 ENSG00000179041 4.41 8.14E-04 2.50E-02 72.2 64.0 98.5 0.0 0.0 5.5<br />

RTN1 6252 ENSG00000139970 -5.34 5.31E-07 5.68E-05 8.1 5.5 0.6 19.3 385.7 286.3<br />

RTP4 64108 ENSG00000136514 Inf 7.77E-06 5.72E-04 0.0 0.7 98.1 58.2 0.0 0.0<br />

S100A6 6277 ENSG00000197956 6.04 2.05E-17 5.08E-14 2259.3 324.2 5709.9 512.7 12.3 53.4<br />

S100B 6285 ENSG00000160307 -2.18 5.35E-04 1.83E-02 3679.1 1302.1 6.3 66.5 3663.5 496.5<br />

S1PR3 1903 ENSG00000213694 -4.14 4.96E-03 9.98E-02 29.7 6.3 0.0 0.0 4.6 69.1<br />

SALL1 6299 ENSG00000103449 -2.39 2.66E-03 6.19E-02 126.2 81.2 0.0 112.8 409.7 267.7<br />

SALL2 6297 ENSG00000165821 -4.13 2.94E-05 1.71E-03 72.9 38.6 16.5 11.6 194.2 581.9<br />

SCN11A 11280 ENSG00000168356 -Inf 3.20E-03 7.17E-02 0.0 0.0 0.0 0.0 0.0 43.4<br />

SCN1B 6324 ENSG00000105711 4.36 3.42E-06 2.87E-04 654.7 1438.9 141.0 25.4 31.9 20.2<br />

SCN5A 6331 ENSG00000183873 -Inf 4.18E-03 8.84E-02 0.0 0.0 0.0 0.0 0.0 40.4<br />

SDC2 6383 ENSG00000169439 -3.28 7.59E-04 2.37E-02 18.2 11.0 14.3 47.7 253.9 216.2<br />

SDK2 54549 ENSG00000069188 -3.29 3.28E-03 7.27E-02 44.4 23.9 0.0 4.1 0.0 178.5<br />

SELENBP1 8991 ENSG00000143416 -3.91 7.83E-04 2.42E-02 25.5 5.2 20.1 6.7 110.1 203.2<br />

SEMA3D 223117 ENSG00000153993 -6.30 5.75E-04 1.94E-02 0.0 2.3 0.0 0.0 0.0 82.0<br />

SEMA4G 57715 ENSG00000095539 -3.43 4.43E-03 9.22E-02 29.7 13.2 4.0 0.0 69.2 47.4<br />

SEMA6A 57556 ENSG00000092421 -3.48 6.56E-05 3.32E-03 68.1 50.0 3.1 0.0 257.1 140.0<br />

SERPINE2 5270 ENSG00000135919 3.55 1.21E-04 5.51E-03 1392.0 2436.4 127.5 513.6 92.8 83.1<br />

SEZ6L 23544 ENSG00000100095 6.22 1.07E-03 3.15E-02 103.7 109.6 0.0 4.0 0.0 1.0<br />

SFRP1 6422 ENSG00000104332 -2.10 7.59E-04 2.37E-02 91.7 535.5 0.0 12.3 1178.7 396.2<br />

SFTA3 253970 ENSG00000229415 -Inf 9.82E-07 9.73E-05 0.0 0.0 0.0 0.0 125.0 32.3<br />

SGCD 6444 ENSG00000170624 4.73 1.20E-05 8.39E-04 172.6 443.4 0.0 0.0 0.0 11.0<br />

SH3BGR 6450 ENSG00000185437 3.88 4.80E-06 3.79E-04 47.5 40.0 989.7 672.0 39.6 37.6<br />

SHC4 399694 ENSG00000185634 2.94 4.92E-03 9.93E-02 50.1 30.0 184.2 63.3 12.9 11.1<br />

SHISA2 387914 ENSG00000180730 -Inf 1.15E-03 3.31E-02 0.0 0.0 0.0 0.0 0.0 55.9<br />

SHOX2 6474 ENSG00000168779 Inf 8.30E-04 2.54E-02 41.9 23.4 64.0 0.0 0.0 0.0<br />

SHROOM3 57619 ENSG00000138771 -3.30 3.19E-04 1.23E-02 95.0 29.6 72.1 132.7 807.8 726.4<br />

SIX3 6496 ENSG00000138083 -5.50 1.88E-08 2.91E-06 2.7 26.1 0.0 0.0 203.9 594.6<br />

SIX6 4990 ENSG00000184302 -10.35 4.28E-15 3.74E-12 1.4 1.6 0.0 0.0 1346.6 17.9<br />

SKAP2 8935 ENSG00000005020 4.09 4.19E-05 2.28E-03 52.5 58.8 26.6 360.6 8.4 8.9<br />

SLC15A2 6565 ENSG00000163406 -6.36 1.23E-05 8.53E-04 1.4 1.2 1.9 0.0 150.4 38.3<br />

SLC16A3 9123 ENSG00000141526 Inf 1.99E-03 5.01E-02 0.0 0.0 21.9 52.7 0.0 0.0<br />

SLC26A2 1836 ENSG00000155850 2.94 2.54E-03 5.96E-02 590.4 1552.1 12.7 95.7 96.6 48.5<br />

SLC2A5 6518 ENSG00000142583 Inf 7.62E-05 3.75E-03 1.7 3.1 4.5 109.7 0.0 0.0<br />

SLC38A1 81539 ENSG00000111371 3.22 3.45E-04 1.32E-02 150.8 124.7 320.3 98.7 14.5 24.5<br />

SLC38A5 92745 ENSG00000017483 Inf 8.90E-04 2.70E-02 63.4 20.6 61.7 4.0 0.0 0.0<br />

SLC46A1 113235 ENSG00000076351 Inf 3.44E-03 7.54E-02 12.1 25.0 10.0 34.8 0.0 0.0<br />

SLC4A11 83959 ENSG00000088836 Inf 1.75E-05 1.14E-03 15.5 41.5 87.7 11.6 0.0 0.0<br />

SLC4A4 8671 ENSG00000080493 -4.49 5.85E-07 6.12E-05 36.0 38.9 13.1 18.8 925.2 145.3<br />

SLCO1C1 53919 ENSG00000139155 -Inf 3.19E-09 5.86E-07 7.7 0.0 0.0 0.0 166.2 151.8<br />

SLCO2A1 6578 ENSG00000174640 -5.61 7.54E-08 1.00E-05 38.8 15.5 0.0 0.0 213.0 297.0<br />

SLITRK5 26050 ENSG00000165300 -4.56 7.21E-04 2.29E-02 13.7 0.0 16.3 0.0 46.4 207.6<br />

SMAGP 57228 ENSG00000170545 3.32 4.27E-04 1.58E-02 17.6 11.6 176.0 226.3 8.5 19.2<br />

SMOC2 64094 ENSG00000112562 -Inf 2.12E-04 8.75E-03 2.7 0.0 0.0 0.0 0.0 79.9<br />

SNAP25 6616 ENSG00000132639 3.34 1.14E-04 5.26E-03 306.2 396.7 79.9 513.8 20.5 44.6<br />

SNCAIP 9627 ENSG00000064692 -5.66 3.64E-03 7.90E-02 4.4 1.6 0.0 0.0 2.8 49.4<br />

SNHG12 85028 ENSG00000197989 2.75 1.36E-03 3.77E-02 29.7 29.7 357.1 235.4 25.1 36.6<br />

246


A.1 Differential Expression Appendix<br />

Gene symbol(s) Entrez gene ID(s) Ensembl 56 gene ID(s) Differential expression results Normalised tag counts<br />

log 2(F C) p-value FDR G144ED G144 G166 G179 CB541 CB660<br />

SNX10 29887 ENSG00000086300 4.39 2.53E-03 5.95E-02 82.6 39.4 24.4 63.7 0.0 4.3<br />

SNX22 79856 ENSG00000157734 7.52 1.08E-06 1.05E-04 162.4 276.1 1.9 0.0 0.0 1.0<br />

SOD2 6648 ENSG00000112096 3.20 1.06E-04 4.94E-03 45.8 51.4 187.6 1755.6 53.2 90.9<br />

SOD3 6649 ENSG00000109610 Inf 1.66E-10 4.05E-08 112.8 3.1 401.2 0.8 0.0 0.0<br />

SORCS2 57537 ENSG00000184985 -9.04 6.02E-13 2.49E-10 1.5 1.6 0.6 0.0 690.7 77.4<br />

SORCS3 22986 ENSG00000156395 Inf 1.63E-04 7.07E-03 82.4 81.1 2.6 30.0 0.0 0.0<br />

SORL1 6653 ENSG00000137642 3.93 3.16E-04 1.22E-02 586.9 293.8 1.4 52.7 3.0 12.1<br />

SOX10 6663 ENSG00000100146 Inf 1.63E-17 4.85E-14 2350.8 3216.5 0.0 0.0 0.0 0.0<br />

SOX3 6658 ENSG00000134595 3.74 1.67E-04 7.18E-03 2.5 3.1 256.8 75.8 1.8 15.2<br />

SPAG4 6676 ENSG00000061656 5.25 1.07E-03 3.15E-02 6.8 4.7 81.5 30.5 0.0 2.4<br />

SPARCL1 8404 ENSG00000152583 -4.03 2.40E-05 1.48E-03 44.2 39.0 4.8 282.9 1448.1 2097.6<br />

SPINK2 6691 ENSG00000128040 Inf 9.14E-04 2.76E-02 0.0 3.8 79.9 0.0 0.0 0.0<br />

SPINT1 6692 ENSG00000166145 4.31 1.01E-06 9.90E-05 0.0 0.0 1009.1 7.2 19.9 13.7<br />

SPP1 6696 ENSG00000118785 3.58 1.10E-04 5.10E-03 75.6 194.5 2549.4 7850.3 335.4 254.9<br />

SPRED1 161742 ENSG00000166068 -3.32 6.80E-04 2.20E-02 15.4 17.7 6.9 24.1 217.2 101.8<br />

SPTBN5 51332 ENSG00000137877 -5.93 3.39E-05 1.90E-03 0.0 0.0 5.0 0.0 151.8 52.0<br />

SQRDL 58472 ENSG00000137767 3.58 1.07E-03 3.15E-02 31.7 90.8 183.0 52.3 18.4 0.0<br />

SRGAP3 9901 ENSG00000196220 -3.35 5.76E-04 1.94E-02 21.5 14.4 0.0 38.0 214.2 138.7<br />

SRP9 6726 ENSG00000143742 -2.29 2.21E-03 5.38E-02 443.5 726.1 195.4 161.4 1982.7 1557.9<br />

SRPX 8406 ENSG00000101955 2.86 1.23E-03 3.49E-02 1070.0 1473.9 771.8 4757.1 321.8 319.2<br />

ST6GAL1 6480 ENSG00000073849 4.93 4.81E-04 1.71E-02 184.6 171.7 0.7 12.0 0.0 4.0<br />

ST6GALNAC3 256435 ENSG00000184005 -4.73 1.69E-03 4.46E-02 0.0 4.7 0.0 0.0 23.6 59.7<br />

ST6GALNAC5 81849 ENSG00000117069 -8.25 1.01E-11 3.42E-09 8.9 0.0 0.0 5.8 261.3 899.5<br />

STAMBPL1 57559 ENSG00000138134 3.81 7.34E-05 3.66E-03 0.0 0.0 86.7 334.9 3.5 15.9<br />

STC2 8614 ENSG00000113739 3.31 1.18E-03 3.37E-02 0.0 0.0 230.2 26.8 0.0 17.2<br />

STEAP1 26872 ENSG00000164647 6.19 2.40E-07 2.87E-05 6.2 35.9 219.6 104.7 2.5 1.4<br />

STEAP2 261729 ENSG00000157214 3.94 2.25E-05 1.40E-03 0.0 0.0 319.9 414.3 25.9 6.1<br />

STRA6 64220 ENSG00000137868 -5.09 6.26E-07 6.51E-05 0.0 38.8 7.5 9.8 16.7 1255.8<br />

STX3 6809 ENSG00000166900 Inf 5.56E-04 1.88E-02 4.1 0.0 72.6 19.5 0.0 0.0<br />

STXBP5L 9515 ENSG00000145087 -Inf 1.60E-05 1.05E-03 0.0 0.0 0.0 0.0 103.4 0.0<br />

SULF1 23213 ENSG00000137573 -3.79 3.40E-03 7.47E-02 0.0 11.3 0.0 0.0 13.7 86.9<br />

SULF2 55959 ENSG00000196562 3.31 1.15E-04 5.28E-03 749.0 891.0 253.5 581.6 43.3 72.4<br />

SYNGR1 9145 ENSG00000100321 2.48 2.40E-03 5.73E-02 127.2 130.6 487.6 414.2 52.8 71.1<br />

SYNM 23336 ENSG00000182253 -4.23 6.56E-05 3.32E-03 1.4 0.0 0.0 36.4 238.8 212.8<br />

SYT1 6857 ENSG00000067715 -2.46 6.89E-04 2.22E-02 87.5 110.3 0.0 16.1 374.6 93.7<br />

SYTL4 94121 ENSG00000102362 3.14 2.93E-03 6.67E-02 37.0 54.7 71.1 142.0 5.9 14.1<br />

SYTL5 94122 ENSG00000147041 3.34 1.88E-03 4.82E-02 0.0 0.0 14.8 277.9 12.2 7.3<br />

TAGLN 6876 ENSG00000149591 -7.06 8.97E-14 4.44E-11 1.9 3.1 9.4 4.1 1089.6 384.4<br />

TAPBPL 55080 ENSG00000139192 2.57 3.86E-03 8.32E-02 123.3 118.8 114.3 780.6 93.5 20.3<br />

TBC1D8 11138 ENSG00000204634 5.89 5.01E-05 2.62E-03 13.8 15.7 73.7 90.9 0.0 2.4<br />

TCEAL2 140597 ENSG00000184905 Inf 2.51E-08 3.69E-06 1.4 3.3 27.2 248.9 0.0 0.0<br />

TCF7 6932 ENSG00000081059 3.00 6.69E-04 2.18E-02 59.5 70.8 87.7 368.1 8.4 35.1<br />

TCF7L2 6934 ENSG00000148737 -2.60 3.98E-03 8.49E-02 33.8 34.3 69.4 42.3 343.9 248.4<br />

TECRL 253017 ENSG00000205678 Inf 3.34E-06 2.83E-04 256.3 195.2 0.0 0.0 0.0 0.0<br />

TES 26136 ENSG00000135269 -Inf 6.77E-12 2.45E-09 0.0 0.0 0.0 0.0 433.0 184.2<br />

TESC 54997 ENSG00000088992 -3.42 2.19E-03 5.38E-02 21.6 13.6 0.0 0.0 92.8 8.6<br />

TF 7018 ENSG00000091513 3.70 4.33E-03 9.07E-02 140.3 205.9 0.0 14.5 5.5 6.1<br />

TFAP2A 7020 ENSG00000137203 3.71 4.55E-04 1.64E-02 476.7 357.8 42.5 104.2 26.0 0.0<br />

TFCP2 7024 ENSG00000135457 5.47 6.46E-04 2.12E-02 47.7 99.6 20.3 14.7 0.0 2.0<br />

TGFA 7039 ENSG00000163235 4.55 3.44E-05 1.92E-03 216.7 272.7 99.7 54.3 12.1 0.0<br />

TGM2 7052 ENSG00000198959 3.93 9.46E-06 6.83E-04 0.0 62.7 290.3 418.6 11.5 22.5<br />

THBS2 7058 ENSG00000186340 3.55 2.42E-05 1.48E-03 237.0 73.4 39.0 1655.3 38.9 61.5<br />

247


A.1 Differential Expression Appendix<br />

Gene symbol(s) Entrez gene ID(s) Ensembl 56 gene ID(s) Differential expression results Normalised tag counts<br />

log 2(F C) p-value FDR G144ED G144 G166 G179 CB541 CB660<br />

THY1 7070 ENSG00000154096 3.29 2.41E-04 9.76E-03 2994.5 2506.5 150.3 2389.9 150.0 195.0<br />

TM4SF1 4071 ENSG00000169908 5.56 2.08E-08 3.16E-06 117.3 242.3 308.7 4.9 1.0 7.4<br />

TMCO4 255104 ENSG00000162542 5.51 1.45E-04 6.41E-03 2.7 1.6 42.3 129.9 1.2 1.0<br />

TMEM100 55273 ENSG00000166292 4.02 1.48E-03 4.03E-02 186.9 150.7 0.0 47.0 0.0 8.1<br />

TMEM132D 121256 ENSG00000151952 -Inf 1.01E-04 4.75E-03 0.0 0.0 0.0 0.0 57.0 23.2<br />

TMEM158 25907 2.67 1.85E-03 4.77E-02 535.5 466.7 154.3 1586.1 140.7 91.0<br />

TMEM176B 28959 ENSG00000106565 6.03 2.07E-03 5.11E-02 20.3 93.6 0.0 5.5 0.0 1.0<br />

TMEM200B 399474 ENSG00000187975 4.13 9.61E-04 2.87E-02 2.7 1.6 58.5 119.9 0.9 6.5<br />

TMEM38A 79041 ENSG00000072954 3.09 1.80E-03 4.67E-02 205.9 608.9 70.7 5.1 33.3 20.2<br />

TMEM71 137835 ENSG00000165071 -3.20 2.42E-03 5.74E-02 1.4 12.6 20.2 1.0 194.7 10.1<br />

TMSB15A 11013 ENSG00000158164 -2.98 1.29E-03 3.62E-02 0.0 0.0 171.6 265.9 1121.9 1173.9<br />

TMSL1 ENSG00000223551 -2.64 4.07E-03 8.65E-02 25.6 9.8 177.7 36.9 595.0 336.6<br />

TNC 3371 ENSG00000041982 2.99 3.08E-04 1.19E-02 215.9 188.2 31.9 1597.5 59.8 91.8<br />

TNFAIP2 7127 ENSG00000185215 6.89 5.19E-11 1.43E-08 0.0 0.0 28.1 513.3 0.0 3.0<br />

TNFAIP3 7128 ENSG00000118503 2.86 3.34E-03 7.40E-02 16.2 17.2 83.3 177.3 0.0 25.7<br />

TNFAIP6 7130 ENSG00000123610 4.95 8.74E-08 1.14E-05 621.2 458.0 6.8 288.5 0.0 15.8<br />

TNFRSF14 8764 ENSG00000157873 4.01 2.55E-04 1.02E-02 12.7 7.9 67.4 260.7 12.9 1.0<br />

TNFSF4 7292 ENSG00000117586 3.98 3.69E-06 3.06E-04 35.1 21.4 55.2 2851.5 58.6 64.6<br />

TNNC2 7125 ENSG00000101470 -Inf 1.46E-04 6.41E-03 0.0 0.0 0.0 0.0 0.0 85.9<br />

TNNI1 7135 ENSG00000159173 -Inf 2.55E-05 1.54E-03 0.0 0.0 0.0 0.0 0.0 116.0<br />

TNNI2 7136 ENSG00000130598 -Inf 2.84E-05 1.67E-03 0.0 0.0 0.0 0.0 0.0 114.0<br />

TOX 9760 ENSG00000198846 3.31 1.07E-03 3.15E-02 197.4 221.1 5.6 134.9 3.2 21.2<br />

TPM1 7168 ENSG00000140416 -2.94 1.95E-03 4.92E-02 85.7 78.5 269.4 215.4 955.6 1911.5<br />

TPMT 7172 ENSG00000137364 2.85 1.09E-03 3.17E-02 179.6 367.0 563.7 1733.5 198.9 47.9<br />

TRAF1 7185 ENSG00000056558 3.92 2.00E-03 5.01E-02 17.9 36.3 19.5 145.7 6.8 2.0<br />

TRAM1L1 133022 ENSG00000174599 -5.13 4.75E-05 2.50E-03 0.0 0.0 0.0 11.3 121.7 146.3<br />

TRIB2 28951 ENSG00000071575 -2.05 1.95E-03 4.92E-02 434.9 352.2 34.9 55.8 919.1 307.4<br />

TRIB3 57761 ENSG00000101255 3.42 5.71E-05 2.94E-03 83.9 109.6 4203.7 837.1 158.2 161.8<br />

TRIM14 9830 ENSG00000106785 3.38 2.02E-03 5.05E-02 54.4 63.7 73.3 155.4 13.5 5.3<br />

TRIM47 91107 ENSG00000132481 3.88 7.17E-06 5.33E-04 106.8 49.3 265.0 527.1 7.6 29.9<br />

TRIM48 79097 ENSG00000150244 Inf 1.84E-04 7.84E-03 21.6 124.2 0.0 0.0 0.0 0.0<br />

TRPM8 79054 ENSG00000144481 4.14 2.35E-05 1.45E-03 156.8 802.9 10.6 75.3 20.5 13.1<br />

TSHZ3 57616 ENSG00000121297 4.45 2.11E-05 1.32E-03 136.6 142.5 38.5 184.1 0.0 11.1<br />

TSLP 85480 ENSG00000145777 5.71 1.24E-04 5.63E-03 4.1 4.6 155.0 0.0 0.0 2.0<br />

TSPAN13 27075 ENSG00000106537 2.62 1.25E-03 3.53E-02 126.1 234.5 127.9 748.4 28.1 91.9<br />

TSPAN7 7102 ENSG00000156298 3.70 6.53E-05 3.32E-03 737.7 849.2 1.3 1.7 0.0 43.7<br />

TSPAN9 10867 ENSG00000011105 2.72 2.51E-03 5.92E-02 78.4 90.4 99.0 4588.1 416.6 68.8<br />

TSTD1 100131187 ENSG00000215845 4.79 1.38E-03 3.80E-02 0.0 0.0 127.5 0.0 0.0 2.7<br />

TTF2 8458 ENSG00000116830 3.05 6.65E-04 2.17E-02 55.3 112.4 65.1 472.7 25.1 27.3<br />

TTN 7273 ENSG00000155657 -Inf 2.68E-03 6.22E-02 0.0 0.0 0.0 0.0 0.0 45.5<br />

TTYH1 57348 ENSG00000167614 -1.91 2.34E-03 5.61E-02 1960.4 834.0 0.0 21.2 1183.5 958.4<br />

TUBA4A 7277 ENSG00000127824 4.85 2.91E-05 1.70E-03 54.0 47.9 185.7 29.1 0.0 6.1<br />

TUBB4 10382 ENSG00000104833 4.59 1.43E-03 3.94E-02 150.5 113.6 32.7 0.0 0.0 3.6<br />

TUSC3 7991 ENSG00000104723 -3.74 1.32E-04 5.89E-03 0.0 0.0 5.9 75.0 379.0 333.7<br />

UGT8 7368 ENSG00000174607 4.64 4.89E-06 3.85E-04 155.7 274.5 169.2 38.9 3.8 8.6<br />

UNC80 285175 ENSG00000144406 -1.98 4.34E-03 9.08E-02 113.5 162.2 0.0 38.7 322.5 207.8<br />

VAX1 11023 ENSG00000148704 -Inf 2.74E-06 2.39E-04 0.0 0.0 0.0 0.0 32.7 121.6<br />

VCAM1 7412 ENSG00000162692 4.34 1.49E-07 1.89E-05 0.0 0.0 46.0 2398.4 8.4 71.4<br />

VIPR1 7433 ENSG00000114812 -4.43 4.74E-06 3.77E-04 6.5 3.1 66.7 6.8 1096.8 1.0<br />

VIPR2 7434 ENSG00000106018 -Inf 2.65E-08 3.86E-06 0.0 0.0 0.0 0.0 216.4 15.9<br />

VIT 5212 ENSG00000205221 6.07 1.83E-05 1.18E-03 14.9 12.4 0.0 193.0 0.0 2.1<br />

VSNL1 7447 ENSG00000163032 3.76 1.35E-04 5.99E-03 0.0 1.6 16.2 332.9 0.0 16.7<br />

248


A.1 Differential Expression Appendix<br />

Gene symbol(s) Entrez gene ID(s) Ensembl 56 gene ID(s) Differential expression results Normalised tag counts<br />

log 2(F C) p-value FDR G144ED G144 G166 G179 CB541 CB660<br />

WBSCR17 64409 ENSG00000185274 -Inf 3.18E-05 1.80E-03 0.0 0.0 0.0 0.0 17.6 90.9<br />

XXbac-BPG116M5.1,C2,CFB 629,717 ENSG00000166278,ENSG00000243649,ENSG00000244255 3.95 4.11E-04 1.54E-02 5.4 4.0 98.3 132.4 0.0 10.6<br />

XYLT1 64131 ENSG00000103489 4.78 1.10E-03 3.20E-02 174.3 164.9 0.0 0.8 0.0 4.0<br />

ZEB1 6935 ENSG00000148516 -2.40 2.63E-03 6.14E-02 504.6 290.3 85.0 57.0 508.8 1006.3<br />

ZIC2 7546 ENSG00000043355 -5.61 2.65E-05 1.59E-03 1.4 6.3 0.8 0.0 0.0 223.2<br />

ZIC3 7547 ENSG00000156925 -Inf 2.12E-04 8.75E-03 0.0 0.0 0.0 0.0 0.0 79.8<br />

ZIC4 84107 ENSG00000174963 -7.51 5.84E-04 1.95E-02 0.0 0.0 0.6 0.0 28.5 47.4<br />

ZIC5 85416 ENSG00000139800 -7.19 6.13E-06 4.74E-04 0.0 1.6 0.6 0.0 0.0 211.7<br />

ZNF281 23528 ENSG00000162702 5.45 4.39E-04 1.61E-02 60.6 51.6 81.7 0.0 0.0 2.0<br />

ZNF423 23090 ENSG00000102935 -7.23 3.72E-07 4.29E-05 0.0 0.0 0.0 3.0 80.2 247.2<br />

ZNF454 285676 ENSG00000178187 4.41 7.62E-05 3.75E-03 24.3 118.3 19.6 152.8 0.0 9.2<br />

ZNF536 9745 ENSG00000198597 Inf 3.95E-05 2.18E-03 41.7 100.5 3.8 37.9 0.0 0.0<br />

ZNF710 374655 ENSG00000140548 -4.55 3.99E-03 8.49E-02 4.1 2.1 5.3 0.0 33.8 68.7<br />

ZNF714 148206 ENSG00000160352 Inf 2.07E-03 5.12E-02 86.5 59.1 18.8 0.0 0.0 0.0<br />

ZNF747 65988 ENSG00000169955 Inf 4.62E-04 1.66E-02 12.2 9.5 14.6 71.1 0.0 0.0<br />

249


A.2 Classified Differential Expression Appendix<br />

A.2 Classified Differential Expression<br />

The differentially expressed genes generated by the Bioconductor R package<br />

DESeq at a FDR


A.2 Classified Differential Expression Appendix<br />

Table A.2: Classification <strong>of</strong> differentially expressed genes based on literature mining analysis.<br />

Implicated in glioma Limited evidence in glioma Not implicated in glioma but other cancers Cancer type Not implicated in cancers<br />

ACIN1 ABAT ADAMTS1 non-small lung carcinoma AC012354.2<br />

ACTC1 ADCYAP1R1 ADAMTS10 glaucoma AC026410.6<br />

ADAMTS4 ADD2 AKR1B10 cervical, endometrial AC034102.1<br />

AGT ADRA1B AMMECR1 renal cell carcinoma AC067930.1<br />

ALDH1A3 BGN ANGPTL2 ovarian cancer AC068399.1<br />

APLN BST2 ARC breast cancer AC092296.1<br />

APOD CNTN6 ARHGAP20 breast, BCLL ACTA2<br />

AQP4 DDAH2 ARHGAP8 colorectal ADRA2A<br />

ASNS DDIT3 ARHGEF7 gastric AFAP1L2<br />

ASPN FCGR2B ATAD3C lung adenocarcinoma AGPAT9<br />

ATOH8 FUT8 AZGP1 breast, pancreatic AIDA<br />

ATP10B FXYD1 BACE2 breast ANGPTL1<br />

ATP1B2 GABRA5 BEX5 breast ATP1A2<br />

BAMBI GBP1 BTBD11 neuroblastoma B3GNT9<br />

BID GBP2 BTG1 ovarian B4GALNT1<br />

BIRC3 GFPT2 C5orf13 esophageal BATF3<br />

BMP7 GJB2 C6orf15 blood BMP8B<br />

BTC HAND2 C8orf4 thyroid BMPER<br />

C1S HAPLN1 CAPN6 HeLa C10orf11<br />

C2 HLA-DMA CARD17 head, neck C10orf116<br />

C3 HLA-DPA1 CDH19 head, neck C10orf81<br />

CA12 HLA-DRB5 CDH6 renal cell carcinoma C10orf90<br />

CALM1 HOXA5 CILP breast C14orf143<br />

CAMK2B HOXA7 CLDN10 hepatocelluar carcinoma C1orf133<br />

CASP1 HOXB6 CLDN11 gastric cancer C1orf187<br />

CBLC HOXB9 CMPK2 chronic myelogenous leukemia and CLL C1orf94<br />

CCKBR HOXC10 CMTM5 prostate,pancreas,carcinoma C20orf103<br />

CCL2 HOXC13 CMTM8 HeLa C21orf62<br />

CCL26 IFITM2 CNKSR2 HL60 leukemia cells C2orf80<br />

CCL7 IGSF3 COL8A1 hepatocarcinoma C3orf58<br />

CCND2 IL17RD CRIP2 nasopharyngeal epithelial cell line C4orf32<br />

CCNY IL1RAPL1 CRYBB2 rhabdoid tumour (kidney) C5orf38<br />

CCR1 INSIG1 CRYM prostate C5orf41<br />

CD248 IRX1 CTNNA2 endometrial C6orf138<br />

CD55 ITGA4 CTSC esophageal cancer C7orf16<br />

CD58 KCNMB2 CXXC4 renal carcinoma C7orf40<br />

CD68 LAMA2 DCBLD2 gastric,lung C9orf125<br />

CD70 LFNG DDIT4L melanoma C9orf64<br />

CD74 LHFPL3 DHRS3 neuroblastoma C9orf95<br />

CD9 MAP3K5 DOCK10 melanoma CABP7<br />

CD97 MEST DOCK5 osteosarcoma CACNA1A<br />

CDH13 MFAP2 DUSP16 Burkitt’s lymphoma CACNA1C<br />

CDKN2A MMP17 DYNC1I1 myeloma CACNG7<br />

CDKN2C MOSC2 EDA2R colorectal CACNG8<br />

CEBPB MX1 EEF1A2 ovarian, pancreatic, lung CAMK1D<br />

CFB NCALD EPDR1 colorectal CARD16<br />

CITED4 NEFM EPHB3 non small cell lung cancer, colon CCDC129<br />

CLDN3 NELL2 ETS2 prostate,colon,breast CCDC48<br />

CNTN1 NFATC2 F12 small cell lung CCDC64<br />

COL1A2 NFKBIZ FAM84A colon CCNO<br />

COL3A1 NR0B1 FBLN2 breast CDHR1<br />

COL4A6 NRN1 FBN2 squamous cell carcinoma CGREF1<br />

251


A.2 Classified Differential Expression Appendix<br />

Implicated in glioma Limited evidence in glioma Not implicated in glioma but other cancers Cancer type Not implicated in cancers<br />

CPAMD8 NXPH1 FCGR2B follicular lymphoma CHCHD10<br />

CREB3L1 OAS3 FCRLA B cell lymphoma CHODL<br />

CTLA4 OLFM1 FERMT3 lung CHRDL1<br />

CXCL1 OXTR FGF19 liver,lung,colon CMAH<br />

CXCL12 PCDHB3 FOXJ1 pancreatic COL21A1<br />

CXCL14 PDE4B FOXQ1 colorectal,breast CPLX2<br />

CXCL2 PERP FXYD5 gastric,pancreatic,esophageal,cervical CPNE2<br />

CXCL3 PIGA GCNT1 prostate CPNE5<br />

CXCL6 PITPNC1 GJB2 breast,head,neck,pancreatic,endometrial CRB2<br />

DCN PKNOX2 GJC3 HeLa CRYBB1<br />

DKK1 PMP2 GMPR Human promyelocytic leukemia-cells CSGALNACT1<br />

DLK1 PNP GPC3 hepatocelluar carcinoma,liver CXorf38<br />

DNER PPEF1 GRB14 breast,prostate CYB5R2<br />

DRD2 PSPH HAND2 neuroblastoma DCHS1<br />

DUSP5 PTPRR HLA-DPA1 pilocytic astrocytoma DDO<br />

EEF1D PXDN HLA-DQA1 breast, gastric,lung adenocarcinoma DIAPH2<br />

EFEMP1 PYGL HLA-DRB4 childhood ALL DKFZp434H1419<br />

EFNA5 RAD50 HOXA4 ovarian,AML DNM3<br />

EGFR RARRES3 HTATIP2 medulloblastoma,neuroblastoma DPY19L1<br />

ELMO1 RASGEF1C IFI27 skin,epithelial,breast DTX4<br />

EPAS1 RRAD IFI6 hepatocellular,gastric,fibrosarcoma ECHDC2<br />

EPB41L3 SALL1 IGSF11 hepatocellular,gastrointestinal EFHD2<br />

ERBB4 SCN1B IL4I1 B-cell lymphoma ELMO2<br />

ESRRG SCN5A INS colon,breast, ELMOD1<br />

ETS1 SIX6 IRX2 s<strong>of</strong>t tissue sarcoma ELN<br />

F2RL1 SLC4A4 ITIH5 breast cancer ELOVL2<br />

FAM38B SLITRK5 ITM2A alveolar rhabdomyosarcoma ELTD1<br />

FCGR2A SNAP25 JPH1 colorectal carcinoma EML2<br />

FGFBP2 SNX10 KANK1 renal cell carcinoma EPDR1<br />

FGFR1 SORL1 KCNJ12 prostate,stomach,breast EVC2<br />

FMNL1 SQRDL KIAA1217 non-small cell lung cancer FAM126A<br />

FOXG1 SRGAP3 L3MBTL4 breast cancer FAM129A<br />

FXYD3 SRPX LEMD1 prostate,colorectal FAM134B<br />

GAL STAMBPL1 LMO2 gastrointestinal,hepatocellular,BLL,T-ALL FAM150B<br />

GALR1 SULF2 LMO4 squamous cell carcinoma,breast,pancreatic FAM176A<br />

GAS7 SYT1 LOXL4 head/neck carcinoma,bladder FAM181A<br />

GDF15 TAGLN LPHN2 breast cancer FAM184B<br />

GFAP TM4SF1 LRAT colon,colorectal,renal,mammary FAM189A1<br />

GGH TMEM71 LRP4 CLL FAM196A<br />

GJA1 TNFAIP6 LRRC55 pancreatic FAM5B<br />

GPNMB TUSC3 LUM cervical,colorectal FAM69A<br />

GRIA1 LXN prostate,gastric,colon,melanoma,medulloblastoma FAM70A<br />

GRIA3 MAF cervical squamous FBXO27<br />

GRM3 MALL oligodendrogliomas FBXO32<br />

HEPACAM MAN1C1 malignant fibrous histiocytomas FCGR2C<br />

HIF3A MAP7 colon FEZF2<br />

HLA-A MARVELD3 pancreatic FXYD7<br />

HLA-DPB1 MEIS2 ovarian,embryonal carcinoma,myeloid leukemia FZD3<br />

HLA-DQB1 MGLL breast GABBR2<br />

HLA-DRA MICB cervical,osteosarcoma,squamous cell carcinoma GABRQ<br />

HLA-DRB1 MIPOL1 nasopharyngeal carcinoma,prostate GALNT5<br />

HLA-DRB3 MLPH lung,breast GBP3<br />

HMGA2 MMRN1 non-small cell lung cancer GBP4<br />

252


A.2 Classified Differential Expression Appendix<br />

Implicated in glioma Limited evidence in glioma Not implicated in glioma but other cancers Cancer type Not implicated in cancers<br />

HOMER1 MN1 meningioma,ALL GBP5<br />

HOXA1 MT1A breast GDPD2<br />

HOXA10 MXRA5 ovarian,colon GEM<br />

HOXB7 MYBL1 leukemias GLYATL2<br />

HOXD10 MYH3 adenocarcinoma GNG11<br />

HOXD13 NBL1 pancreatic,colon GPR158<br />

HOXD3 NDE1 AML GPR98<br />

HOXD9 NFE2L3 pancreatic,T-cell lymphoblastic lymphoma GRIK3<br />

HPRT1 NID1 colon,gastric GYG2<br />

HPSE NOP16 murine breast H1F0<br />

ICAM1 NPFFR2 Primary CNS lymphoma HCP5<br />

ID4 NR0B1 lung adenocarcinoma,sarcoma HLA-DQA2<br />

IFI30 NR1D1 breast HRCT1<br />

IGF2 NTN1 colorectal,neuroblastoma IFITM8P<br />

IGFBP5 OAS2 prostate,breast IGLON5<br />

IKBKE ODZ2 lymphoma IL33<br />

IL13RA2 OGN hepatocarcinoma INS-IGF2<br />

IL1B PARP3 breast cancer ISYNA1<br />

IL1R1 PARP12 breast cancer ITGBL1<br />

IL6 PCDH20 non-small-cell lung cancer KALRN<br />

IL8 PHACTR3 non-small-cell lung cancer KCNG1<br />

INHBA PITX1 breast,lung,adenocarcinoma KCNK12<br />

INHBB PKI55 cancer KCTD12<br />

INPP5D PLD3 breast LINGO1<br />

IRAK1 PLS1 lung adenocarcinoma LOC100127888<br />

JAM2 PLS3 HeLa LOC285141<br />

KCNA2 PLSCR1 ovarian,colorectal LRRC2<br />

KCNK1 PMEPA1 renal cell carcinoma LYST<br />

KLF6 PNMA2 gastrointestinal neuroendocrine carcinomas MACROD2<br />

LAMA4 PPAP2C MAP6<br />

LGALS3 PPL esophageal MARCH1<br />

LIF PPP4R1 breast,gastric MID1IP1<br />

LIFR PRIMA1 thyroid,non small cell lung cancer cells,prostate MOCOS<br />

LMO3 PROCR breast MYL1<br />

LNX1 PTPRD gastrointestinal MYL9<br />

LPAR6 PTPRH stomach,colorectal MYO1B<br />

LRRN2 RAB11FIP1 breast NKX6-2<br />

LTF RAB38 melanoma NRBF2<br />

LY96 RAB6B breast,neuroendocrine OASL<br />

MAL RARRES1 melanoma,nasopharyngeal carcinoma,prostate OTOR<br />

MARS RARRES3 ovarian,breast PCDHB12<br />

MATN2 REC8 gastrointestinal stromal tumour,lymphoma PCDHB4<br />

MBP RGS17 lung,prostate,ovarian PCOLCE2<br />

MET RGS5 gastric,renal cell carcinoma PDE1C<br />

MIA RHOBTB3 ganglioglioma PDE1C<br />

MICA RNASEH1 prostate PDZRN3<br />

MKI67 RNF175 pediatric AML,non-Hodgkinslymphoma PION<br />

MLC1 RTN1 neuroblastoma PLA2G4A<br />

MMP7 SALL2 synovial sarcoma PLAC9<br />

MT2A SEMA4G colorectal PLCH1<br />

MTTP SEZ6L lung PLCH1<br />

MYC SHOX2 lung PLEKHA6<br />

NAMPT SIX3 Chondrosarcomas PPCS<br />

253


A.2 Classified Differential Expression Appendix<br />

Implicated in glioma Limited evidence in glioma Not implicated in glioma but other cancers Cancer type Not implicated in cancers<br />

NCAM1 SKAP2 pancreatic PPHLN1<br />

NDN SLC16A3 bladder cancer PPM1K<br />

NFASC SLC26A2 gastric,colon carcinoma PRSS12<br />

NFIA SLC2A5 breast,adenocarcinoma,colon,liver,lymphomas PRSS12<br />

NKX2-1 SLCO2A1 colorectal PSORS1C1<br />

NKX6-2 SMAGP metastatic cell lines RAB7L1<br />

NMU SPAG4 various cancers RADIL<br />

NNAT SPARCL1 colorectal,pancreatic,lung RGAG4<br />

NNMT SPINK2 lymphomas RGL3<br />

NOTCH3 SPINT1 colorectal,gastric,breast,stomach,ovarian RGS20<br />

NOV SPRED1 juvenile myelomonocytic leukemia, RHBDF2<br />

NPTX2 SRP9 colorectal RHPN1<br />

NR4A2 ST6GALNAC3 renal cancer RIPK4<br />

NRG1 ST6GALNAC5 breast RP11-473I1.1<br />

NRP2 STC2 esophageal,squamous-cell carcinoma,ovarian RP9<br />

NTRK2 STEAP1 breast,prostate RPSAP52<br />

OAS1 STEAP2 prostate RRS1<br />

OTX2 STRA6 colorectal RTP4<br />

P2RX7 SULF1 colorectal,ovarian,hepatocellular,gastric SCN11A<br />

PAX8 SULF2 lung adenocarcinoma,squamous cell carcinoma SDK2<br />

PDGFA SYTL4 breast SEMA6A<br />

PDGFRA SYTL5 breast SFTA3<br />

PDGFRB TBC1D8 pancreatic SGCD<br />

PDPN TCEAL2 ovarian SH3BGR<br />

PEA15 TCF7 colon cancer,acanthoma SHISA2<br />

PHLPP1 TM4SF1 pancreatic SHROOM3<br />

PI15 TNFAIP2 nasopharyngeal carcinoma SLC38A5<br />

PLA2G2A TNFRSF14 pancreatic SLC46A1<br />

PLCB1 TNFSF4 Hodgkin’s lymphoma,breast SLC4A11<br />

PTEN TPMT acute and lymphoblastic leukemia SMOC2<br />

PTGS1 TRIB2 lung,AML,melanoma SNHG12<br />

PTPRD TRIB3 breast,colorectal cancer SNX22<br />

RASGRP3 TSHZ3 breast,prostate SORCS2<br />

S100A6 TSPAN13 prostate,breast SORCS3<br />

S100B TSPAN7 neuroblastoma,ALL SPTBN5<br />

S1PR3 TSPAN9 ovarian STEAP1B<br />

SDC2 TTN melanoma STX3<br />

SELENBP1 ZIC3 meningioma STXBP5L<br />

SEMA3D ZIC5 meningioma SYNGR1<br />

SERPINE2 ZNF423 neuroblastoma,CML TAPBPL<br />

SFRP1 TBC1D8<br />

SHC4 TECRL<br />

SLC15A2 TESC<br />

SLC38A1 TMCO4<br />

SLCO1C1 TMEM100<br />

SLITRK5 TMEM132D<br />

SNCAIP TMEM158<br />

SOD2 TMEM176B<br />

SOD3 TMEM200B<br />

SOX10 TMEM38A<br />

SOX3 TMSB15A<br />

SPP1 TMSL1<br />

ST6GAL1 TNNC2<br />

254


A.2 Classified Differential Expression Appendix<br />

Implicated in glioma Limited evidence in glioma Not implicated in glioma but other cancers Cancer type Not implicated in cancers<br />

ST6GALNAC5 TNNI1<br />

SYNM TNNI2<br />

TCF7L2 TOX<br />

TES TRAM1L1<br />

TF TRIM14<br />

TFAP2A TRIM48<br />

TFCP2 TSTD1<br />

TGFA TTF2<br />

TGM2 TUBB4<br />

THBS2 TUSC3<br />

THY1 UNC5D<br />

TNC UNC80<br />

TNFAIP3 VAX1<br />

TPM1 VIPR2<br />

TRAF1 VIT<br />

TRIM47 WBSCR17<br />

TRPM8 XYLT1<br />

TSLP ZNF281<br />

TTYH1 ZNF454<br />

TUBA4A ZNF536<br />

UGT8 ZNF710<br />

VCAM1 ZNF714<br />

VIPR1 ZNF747<br />

255<br />

VSNL1<br />

ZEB1<br />

ZIC2<br />

ZIC4


A.3 Quantitative RT-PCR Appendix<br />

A.3 Quantitative RT-PCR<br />

Ct values were normalised to the mean <strong>of</strong> three endogenous control genes (18S<br />

rRNA, TUBB and NDUFB10). Values were capped at 37 prior to normaliza-<br />

tion. Each sample was then normalised by subtracting the mean Ct for the<br />

sample, <strong>of</strong> the three control assays, and adding the global mean Ct <strong>of</strong> the con-<br />

trol assays. The subtraction corresponds to a standard deltaCt normalisation<br />

to adjust for differences in the amount <strong>of</strong> RNA among samples. The addition<br />

ensures that normalised values occupy approximately the same range as the<br />

original values; this is only done for convenience and does not affect the statis-<br />

tical tests since all Ct values are incremented by the same amount. Replicated<br />

samples have suffix (A) - (D). Raw and normalised Ct values are listed in the<br />

tables below.<br />

256


A.3 Quantitative RT-PCR Appendix<br />

Table A.3: Raw Ct values. Abbreviations: "down" for down-regulated, "up" for up-regulated, and "Norm" for Normalisation control.<br />

Well Applied Biosystems assay ID Gene CB130 CB130 CB130 CB152 CB152 CB152 CB152 CB171 CB171 CB171 CB192 CB541 CB660 CB660 G2 G2 G7 G7 G7 G7 G9 G9 G14 G14<br />

Category A B C A B C D A B C A B A B A B C D A B A B<br />

1 MYL9-Hs00382913_m1 Core down 27.9 27.5 28.0 22.9 25.4 26.2 25.1 28.9 31.9 29.6 26.8 24.4 27.1 25.8 37.0 35.9 31.4 36.2 31.2 28.7 26.7 27.4 34.1 34.8<br />

2 CHCHD10-Hs01369775_g1 Core up 26.7 25.3 25.9 25.8 25.5 25.3 24.3 26.7 28.1 26.2 28.8 29.2 29.1 28.6 25.7 25.6 25.8 25.9 26.4 24.0 25.8 27.1 26.2 26.6<br />

3 RGS5-Hs00186212_m1 Core down 30.9 30.0 30.2 29.0 31.9 31.0 29.0 32.5 31.1 30.9 32.6 27.0 26.3 26.7 31.4 32.0 31.1 31.1 29.2 26.9 30.9 32.6 31.6 31.8<br />

4 ST6GALNAC5-<br />

Core down 28.5 28.3 28.5 33.4 34.0 33.0 31.7 29.4 32.2 29.5 25.6 26.2 23.3 24.0 36.1 35.9 36.0 40.0 35.7 34.0 28.9 29.7 30.2 29.5<br />

Hs00229612_m1<br />

5 CEBPB-Hs00270923_s1 Core up 32.8 30.3 30.5 30.2 32.0 31.8 31.1 32.4 32.0 31.4 30.7 31.5 32.4 30.4 32.6 35.2 28.6 30.0 30.5 28.9 29.9 31.0 29.8 29.7<br />

6 C5orf13-Hs00854282_g1 Core down 28.1 27.3 28.0 25.4 27.6 27.0 25.3 28.0 26.8 26.7 29.0 24.3 25.0 25.3 40.0 27.8 29.9 29.2 23.5 26.2 27.7 40.0 26.9 31.1<br />

7 PDGFRA-Hs00998026_m1 Marker 40.0 37.0 34.7 33.1 40.0 28.6 28.2 40.0 40.0 40.0 32.4 35.2 29.7 31.3 25.2 26.9 22.8 22.4 22.2 21.8 24.3 24.9 25.0 25.8<br />

8 CCND2-Hs00922419_g1 Core down 32.0 30.4 30.9 31.5 33.9 29.0 28.3 31.9 32.7 30.9 25.1 23.7 23.9 23.9 23.5 23.8 22.3 22.2 24.7 22.5 30.7 30.7 33.2 36.9<br />

9 NKX2-1-Hs00163037_m1 Core down 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 28.3 28.8 29.0 40.0 40.0 32.3 34.8 33.0 31.7 40.0 40.0 30.4 30.9<br />

10 CTSC-Hs00175188_m1 Core up 23.1 24.2 24.5 24.4 24.2 25.3 24.1 23.1 25.9 24.0 23.9 24.6 25.3 22.9 21.5 22.5 24.9 24.6 27.2 24.4 23.3 24.3 22.5 22.8<br />

11 18S-Hs99999901_s1 Norm 13.3 11.6 11.4 12.1 14.0 11.5 11.1 13.0 12.4 11.3 12.5 12.3 12.0 11.2 14.9 18.9 12.1 13.0 11.1 10.9 12.9 14.8 11.9 12.2<br />

12 DNER-Hs00294564_m1 Core down 31.9 31.9 32.7 32.6 31.8 28.8 27.3 31.7 31.5 32.1 30.4 21.8 22.3 21.9 26.4 25.9 27.4 25.4 24.3 24.3 26.8 24.8 31.7 31.1<br />

13 LMO4-Hs01086790_m1 Core up 26.6 26.3 26.8 26.6 27.7 27.2 25.2 26.4 27.9 27.2 25.6 25.6 25.3 25.9 23.5 23.9 21.8 22.6 24.7 23.0 24.7 26.5 24.3 24.7<br />

14 PLCH1-Hs00392783_m1 Core down 26.1 25.9 26.1 26.3 27.0 27.9 26.1 25.7 28.1 27.6 26.8 26.5 27.7 27.9 33.6 34.8 29.1 30.1 29.9 28.2 28.2 29.9 26.3 26.5<br />

15 SPARCL1-Hs00949886_m1 Core down 33.3 30.8 30.9 26.9 29.8 24.8 22.0 35.2 29.9 30.4 28.4 24.8 22.6 24.8 25.5 25.6 24.0 27.8 29.8 29.0 22.7 26.2 28.1 27.7<br />

16 SYNM-Hs00322391_m1 Core down 28.9 27.8 28.0 26.7 27.6 27.9 25.5 28.2 28.9 28.7 27.0 27.1 26.9 26.4 29.5 30.2 28.5 29.7 29.4 27.9 25.5 28.6 32.8 34.1<br />

17 FBLN2-Hs00157482_m1 Core down 40.0 35.7 40.0 35.9 40.0 40.0 34.9 40.0 35.2 35.5 33.8 28.6 27.3 28.3 34.4 36.0 35.0 32.9 36.9 34.6 33.5 40.0 36.0 35.8<br />

18 SALL2-Hs00413788_m1 Core down 40.0 33.0 33.4 30.9 37.0 30.7 27.0 37.0 30.3 33.2 32.7 26.0 24.4 27.0 31.4 33.8 27.7 28.7 29.6 29.9 34.9 40.0 30.8 29.9<br />

19 TUSC3-Hs00185147_m1 Core down 24.0 22.4 22.9 23.2 23.8 23.3 21.6 24.0 25.2 23.3 24.3 22.9 23.3 23.0 40.0 40.0 35.3 33.9 37.0 33.5 23.7 23.7 23.1 23.5<br />

20 GPR158-Hs00393109_m1 Core down 34.4 34.1 34.8 36.9 36.5 32.1 32.4 35.6 36.1 33.4 26.9 25.1 27.0 25.7 27.2 29.0 27.7 26.8 28.7 25.9 34.9 35.6 31.7 31.9<br />

21 PEG3-Hs00377844_m1 Core down 30.7 30.9 31.1 29.9 30.9 31.0 29.2 29.9 31.2 31.0 29.8 27.6 25.0 27.0 28.2 29.6 26.1 24.8 26.3 25.6 33.6 36.6 28.1 28.7<br />

22 FUT8-Hs00189535_m1 Core up 26.3 26.3 26.7 26.1 27.0 28.0 26.2 26.5 28.7 27.2 26.3 27.4 28.2 26.2 26.1 26.6 27.5 27.5 27.3 26.6 26.0 27.3 27.6 28.0<br />

23 HLA-DRA-Hs00219575_m1 Core up 27.5 26.1 26.3 28.2 30.1 26.0 22.1 28.6 27.0 27.4 22.3 26.7 30.7 25.5 21.4 23.1 24.3 24.2 24.7 22.9 21.4 26.0 26.8 27.6<br />

24 ASCL1-Hs00269932_m1 Marker 34.0 32.0 31.8 32.7 36.0 28.1 26.1 34.5 32.1 35.1 32.7 28.9 27.0 33.6 24.4 24.9 25.5 24.3 24.4 25.1 40.0 40.0 33.4 40.0<br />

25 PI15-Hs00210658_m1 Core down 31.6 30.3 30.1 23.0 28.6 27.1 24.4 31.8 28.6 31.0 29.6 24.5 24.4 23.9 26.3 28.6 31.8 27.6 29.8 29.3 27.0 33.7 31.1 30.1<br />

26 EPDR1-Hs00378148_m1 Core up 28.6 27.9 28.6 26.5 27.5 29.4 26.7 29.0 30.5 29.1 30.8 28.5 29.8 29.1 27.0 27.8 28.2 29.0 29.5 27.5 26.9 28.9 27.9 28.4<br />

27 DTX4-Hs00392288_m1 Core down 32.9 30.6 31.2 28.2 31.6 29.0 27.0 33.2 29.1 30.3 32.9 26.0 24.9 25.9 34.2 37.0 30.3 30.8 32.0 32.6 29.8 33.1 27.5 27.6<br />

28 CXXC4-Hs00228693_m1 Core down 30.9 30.8 30.7 34.1 40.0 30.8 29.4 31.6 31.4 33.2 28.2 25.5 25.6 27.1 28.6 27.4 27.0 26.7 26.3 26.8 40.0 40.0 28.5 29.9<br />

29 IRX2-Hs01383002_m1 Core down 30.1 30.5 30.6 29.1 29.2 31.4 29.9 29.2 31.9 31.1 30.6 26.1 26.4 25.5 29.8 29.8 40.0 40.0 40.0 40.0 30.3 30.3 40.0 40.0<br />

30 MAP6-Hs01023152_s1 Core down 27.6 24.6 25.3 25.8 27.6 24.7 21.8 27.0 25.8 24.7 28.0 26.4 25.7 27.2 26.8 26.6 27.0 26.9 27.5 26.5 28.6 30.6 28.4 28.7<br />

31 SIX3-Hs00193667_m1 Core down 40.0 40.0 40.0 40.0 40.0 40.0 34.6 40.0 40.0 40.0 40.0 25.0 23.8 24.1 40.0 35.2 32.0 33.8 30.5 31.6 32.6 33.7 34.2 34.3<br />

32 NKX2-2-Hs00159616_m1 Marker 40.0 40.0 40.0 37.0 40.0 33.3 33.6 40.0 40.0 40.0 40.0 28.1 30.7 28.7 25.9 26.2 26.7 26.3 29.0 29.3 26.8 27.2 28.0 27.8<br />

33 S100A6-Hs00170953_m1 Core up 19.7 20.0 20.6 20.8 19.2 20.7 20.6 19.0 23.3 20.6 21.9 20.6 25.6 22.1 19.7 19.8 21.3 22.9 22.4 19.1 19.8 18.6 18.8 18.8<br />

34 MMP17-Hs01108847_m1 Core up 27.9 28.2 28.5 28.2 27.7 30.3 28.9 27.5 31.0 30.1 28.6 31.8 30.2 29.0 26.6 30.0 30.0 30.0 29.8 30.1 27.7 27.3 28.0 28.1<br />

35 FAM38B-Hs00926225_m1 Core down 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 23.8 26.8 25.2 40.0 40.0 40.0 40.0 40.0 40.0 24.7 25.3 28.4 28.7<br />

36 OLIG2-Hs00300164_s1 Marker 29.8 32.6 33.1 29.9 30.2 28.9 28.2 32.1 36.0 35.5 29.2 25.6 28.0 26.9 24.3 26.5 21.8 22.2 22.3 22.7 30.5 32.8 30.5 36.0<br />

37 PMEPA1-Hs00375306_m1 Core up 25.7 23.6 23.9 24.7 25.1 25.8 25.8 25.6 28.1 26.0 27.6 26.0 29.0 26.3 26.5 27.8 27.3 27.9 28.3 26.6 26.2 25.1 23.1 24.2<br />

38 HOXD10-Hs00157974_m1 Core up 40.0 37.0 40.0 40.0 40.0 33.1 32.2 40.0 40.0 40.0 40.0 40.0 40.0 40.0 30.1 29.9 27.0 27.9 28.9 26.7 28.3 28.9 27.8 28.3<br />

39 NDN-Hs00267349_s1 Core down 24.8 24.5 25.1 24.0 24.4 25.4 23.1 24.5 26.9 25.2 25.4 23.3 23.7 23.8 25.9 25.6 24.6 25.6 26.6 25.4 23.7 24.9 23.8 23.9<br />

40 FOXG1-Hs01850784_s1 Core up 40.0 40.0 40.0 33.6 34.9 25.9 25.1 40.0 32.7 35.9 35.3 27.3 27.4 27.9 25.9 26.1 23.5 24.1 22.1 20.7 24.3 25.2 23.7 24.0<br />

41 MN1-Hs00159202_m1 Core down 32.0 31.3 31.4 29.3 30.6 31.8 27.6 33.2 33.4 34.1 31.0 25.8 25.8 26.1 29.6 31.0 29.7 28.4 29.6 27.8 34.3 33.1 32.6 32.9<br />

42 NDUFB10-Hs00605903_m1 Norm 24.8 25.0 25.9 25.5 25.2 26.5 24.8 25.1 27.6 25.9 25.3 25.1 25.8 25.2 25.5 25.0 25.0 25.7 27.2 25.5 25.5 25.2 24.2 25.0<br />

43 HMGA2-Hs00171569_m1 Core down 31.5 27.8 28.4 33.5 30.5 29.7 29.5 29.9 29.9 28.9 28.9 26.9 27.0 26.0 32.0 32.0 30.9 30.5 29.6 28.2 34.4 31.3 29.5 30.7<br />

44 MMRN1-Hs00201182_m1 Core down 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 26.0 27.2 27.7 40.0 40.0 36.6 36.4 40.0 36.0 40.0 36.9 32.4 33.9<br />

45 RAB6B-Hs00981572_m1 Core down 34.4 32.0 32.2 32.8 35.3 32.4 29.5 34.1 32.8 32.5 30.4 27.8 28.5 28.1 29.5 30.6 27.2 27.9 28.1 27.7 29.7 30.2 29.3 28.9<br />

46 NTRK2-Hs00178811_m1 Core up 28.9 30.5 31.0 23.2 24.9 26.2 22.9 29.2 30.1 31.0 29.0 28.5 26.7 27.8 22.4 23.3 29.1 25.0 27.6 26.8 22.7 25.0 24.3 23.6<br />

47 KALRN-Hs00610179_m1 Core down 29.5 29.1 30.0 28.0 29.7 31.0 28.8 29.5 30.0 30.1 28.3 26.0 25.7 25.7 28.6 31.0 28.3 28.0 28.1 27.5 27.1 29.7 29.9 30.2<br />

48 CD74-Hs00269961_m1 Core up 27.3 27.3 27.4 27.9 28.9 28.2 24.0 27.4 27.4 28.9 22.1 26.9 30.1 25.5 21.8 24.1 25.2 23.9 26.5 24.8 21.9 27.1 26.9 27.9<br />

257


A.3 Quantitative RT-PCR Appendix<br />

Well Applied Biosystems assay ID Gene CB130 CB130 CB130 CB152 CB152 CB152 CB152 CB171 CB171 CB171 CB192 CB541 CB660 CB660 G2 G2 G7 G7 G7 G7 G9 G9 G14 G14<br />

Category A B C A B C D A B C A B A B A B C D A B A B<br />

49 LUM-Hs00158940_m1 Core down 29.4 28.9 29.2 30.3 31.9 36.9 32.0 31.6 33.4 32.8 29.9 25.5 24.6 24.6 40.0 40.0 32.5 29.0 33.0 31.0 23.0 27.3 29.6 29.7<br />

50 NELL2-Hs00196254_m1 Core down 24.5 24.3 24.7 28.0 29.3 29.1 24.9 24.3 25.6 25.3 25.3 24.6 23.8 25.0 30.5 30.9 30.2 30.7 31.7 30.7 28.7 30.0 34.2 34.0<br />

51 MAP6-Hs01929835_s1 Core down 27.0 25.3 26.1 25.6 26.6 25.0 22.4 26.5 26.0 25.1 27.4 26.1 25.6 27.9 25.8 25.9 26.8 26.4 28.5 27.7 27.3 29.3 26.3 26.5<br />

52 PDE1C-Hs01095694_m1 Core up 29.9 32.4 32.9 27.0 27.9 29.0 28.0 29.2 31.4 30.8 30.4 26.2 27.9 25.9 26.4 28.0 24.3 24.6 25.5 23.3 23.8 27.1 24.2 24.4<br />

53 SULF2-Hs00378697_m1 Core up 28.5 29.7 30.1 25.9 26.8 29.1 25.4 27.9 30.9 31.3 26.8 26.4 27.8 26.7 23.0 23.9 23.3 23.1 25.3 24.4 24.0 23.7 23.9 23.8<br />

54 TUBB-Hs00962420_g1 Norm 20.0 21.3 21.5 21.2 20.6 23.1 22.7 19.8 24.0 22.2 21.3 20.4 21.0 20.2 21.5 23.0 20.8 21.1 23.1 20.6 21.7 21.5 20.5 20.8<br />

55 LMO2-Hs00277106_m1 Core down 36.8 37.1 33.7 35.3 34.1 30.0 29.1 36.0 34.9 33.1 30.4 26.1 25.7 26.3 26.8 27.2 25.4 26.6 27.1 26.8 28.1 30.8 31.7 31.9<br />

56 MAN1C1-Hs00220595_m1 Core up 27.8 26.9 27.0 25.9 29.0 27.8 25.7 27.2 27.2 27.0 27.1 31.1 29.0 28.9 26.8 29.0 26.0 26.8 27.5 26.4 25.2 27.4 27.6 27.4<br />

57 TAGLN-Hs00162558_m1 Core down 21.2 18.8 18.5 20.5 21.9 20.2 21.9 22.0 20.7 20.0 21.8 21.2 23.6 21.8 27.4 29.7 40.0 36.9 37.2 29.3 21.6 22.1 28.6 28.9<br />

58 RTN1-Hs00382515_m1 Core down 26.4 23.0 23.2 29.7 28.8 28.9 25.9 27.2 27.6 26.9 23.8 23.8 25.5 25.3 26.1 26.1 23.4 24.4 26.3 24.2 26.1 27.3 24.7 25.2<br />

59 MAF-Hs00193519_m1 Core down 26.5 25.9 26.7 26.4 25.8 27.4 26.0 26.4 29.7 27.6 27.1 25.2 26.4 26.0 27.4 27.0 27.8 29.4 27.9 25.2 30.1 30.5 27.9 28.2<br />

60 NPTX2-Hs00383983_m1 Core down 35.2 30.6 30.5 30.5 26.8 32.8 29.8 40.0 36.9 35.3 33.6 22.9 25.5 23.6 40.0 40.0 26.2 29.4 31.4 28.5 31.1 26.8 29.6 29.2<br />

61 EDA2R-Hs00939736_m1 Core down 27.9 28.0 28.0 27.6 27.2 28.9 26.8 27.5 29.6 28.2 28.5 26.6 28.4 27.0 27.8 29.2 30.3 34.3 33.5 30.9 28.2 28.9 26.2 26.7<br />

62 SOX10-Hs00366918_m1 Marker 40.0 40.0 40.0 40.0 40.0 35.2 32.8 40.0 40.0 40.0 40.0 40.0 35.1 40.0 27.9 28.0 40.0 37.0 36.0 36.0 40.0 40.0 40.0 40.0<br />

63 SEMA6A-Hs00221174_m1 Core down 25.0 25.9 25.9 24.3 25.2 26.2 24.3 24.6 26.8 26.4 22.7 22.9 23.0 23.3 25.6 26.4 24.9 25.2 26.7 26.3 26.7 30.2 26.2 27.3<br />

64 DDIT3-Hs00358796_g1 Core up 28.0 29.0 29.2 28.4 27.6 27.9 27.3 28.5 30.3 28.6 28.7 27.6 27.9 27.5 27.4 27.2 25.9 25.9 28.8 26.2 26.6 27.1 26.4 26.9<br />

65 CA12-Hs01080909_m1 Core down 30.1 32.2 32.6 30.9 28.3 29.5 31.0 30.9 32.6 31.5 21.5 22.1 23.4 22.5 25.4 26.1 23.4 23.1 24.9 22.3 24.1 26.4 30.4 30.8<br />

66 C9orf125-Hs00260558_m1 Core down 32.9 32.2 32.8 36.7 40.0 34.6 34.8 33.7 35.4 36.3 29.4 28.5 28.1 29.0 28.0 30.2 29.0 28.5 30.2 29.4 28.3 28.2 30.4 31.0<br />

67 SDC2-Hs00299807_m1 Core down 24.7 24.5 24.7 24.1 24.5 25.0 23.3 25.3 26.4 25.2 25.5 25.1 25.8 25.5 27.4 27.0 26.6 28.4 28.3 26.7 26.4 27.9 26.1 26.5<br />

68 SLIT2-Hs00191193_m1 Core down 29.3 33.1 32.9 28.9 28.7 31.4 29.3 29.8 33.0 32.9 31.8 24.0 24.2 24.4 40.0 36.4 27.8 27.6 31.9 30.7 26.4 28.3 25.1 25.5<br />

69 MT2A-Hs02379661_g1 Core up 25.2 23.6 24.2 22.9 22.4 20.9 20.4 25.5 25.3 23.9 23.9 23.3 24.4 24.2 22.5 22.2 20.3 21.4 22.4 20.0 19.1 19.0 22.5 22.3<br />

70 IL17RD-Hs00296982_m1 Core down 26.8 25.7 25.7 25.8 26.9 27.1 26.2 25.8 28.3 26.9 26.3 23.9 24.4 23.7 27.4 28.7 27.7 27.0 27.6 26.9 24.3 24.8 25.9 26.0<br />

71 PDZRN3-Hs00392900_m1 Core down 29.0 28.9 29.0 27.2 28.8 29.6 26.6 28.4 27.7 28.8 28.4 26.4 26.7 26.8 29.8 32.4 27.1 29.1 29.3 29.6 27.9 30.9 29.3 29.6<br />

72 LGALS3-Hs00173587_m1 Core up 25.7 24.5 25.4 24.8 24.0 24.5 23.0 25.6 26.6 25.1 26.5 25.8 25.6 25.0 25.0 24.0 26.4 27.7 26.2 25.1 22.7 23.5 25.5 25.3<br />

73 FAM69A-Hs00961685_m1 Core up 25.3 25.0 25.2 23.8 24.6 26.6 23.5 24.8 26.5 26.3 26.4 27.6 29.7 28.0 25.6 26.0 25.7 26.5 27.3 25.9 24.9 25.9 25.3 25.3<br />

74 TERT-Hs00972656_m1 Marker 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 36.7 36.5 35.5 33.6 34.4 32.4 32.2 32.7 32.0 33.4 33.4 31.0 34.0<br />

75 PLS3-Hs00418605_g1 Core up 24.6 27.6 27.9 25.0 24.1 26.7 26.4 24.4 29.7 28.9 22.9 26.1 25.6 24.1 23.4 24.4 25.3 26.6 26.9 24.2 22.5 23.1 21.8 22.0<br />

76 PTEN-Hs02621230_s1 Core down 30.0 27.4 27.6 29.4 30.5 28.1 26.4 28.8 28.3 27.5 29.5 27.6 27.6 27.3 40.0 40.0 29.9 30.9 27.9 27.3 40.0 40.0 27.8 28.0<br />

77 SALL2-Hs00826674_m1 Core down 34.4 31.7 31.9 32.2 34.0 33.1 29.7 34.8 32.6 33.0 34.8 32.5 32.4 32.6 31.5 32.2 30.8 31.4 33.2 28.7 33.1 34.9 34.1 33.9<br />

78 KCTD12-Hs00540818_s1 Core down 30.6 30.2 30.3 33.3 33.9 32.7 33.2 29.7 29.7 29.8 26.5 25.3 27.3 26.2 27.8 30.0 30.8 32.4 35.5 32.1 27.2 29.7 33.6 32.4<br />

79 TES-Hs00210319_m1 Core down 25.7 25.6 26.1 27.7 27.3 28.9 28.4 25.7 29.1 26.9 32.6 25.1 26.1 25.6 40.0 36.6 40.0 40.0 40.0 37.0 33.4 40.0 35.9 40.0<br />

80 SOX2-Hs01053049_s1 Marker 25.2 21.3 21.7 24.1 26.2 22.9 20.4 24.3 22.5 22.0 24.4 23.1 23.4 23.5 24.7 24.9 23.0 23.4 22.6 21.7 25.3 27.9 23.7 23.8<br />

81 LPAR6-Hs00271758_s1 Core down 30.9 29.0 29.4 34.3 34.3 32.2 30.7 31.2 30.8 30.1 29.8 27.9 28.5 28.5 31.9 31.9 28.1 29.8 29.4 27.8 27.7 29.6 29.2 29.8<br />

82 ODZ2-Hs00393060_m1 Core down 28.3 27.3 27.5 26.7 28.0 28.1 26.8 28.8 29.9 29.4 25.7 26.9 25.0 24.7 30.8 31.6 24.7 26.4 27.8 26.1 27.8 30.1 25.4 25.8<br />

83 NNMT-Hs00196287_m1 Core up 31.0 27.5 27.9 28.8 32.2 30.1 29.6 32.0 30.9 28.5 29.9 27.7 29.4 27.9 30.9 32.6 23.7 26.1 24.5 22.2 22.3 23.7 28.9 27.0<br />

84 CACNG8-Hs01100182_m1 Core down 40.0 33.0 32.7 32.6 40.0 33.7 34.5 33.6 34.8 35.3 28.7 27.3 26.1 27.3 29.7 30.4 26.8 28.7 30.1 28.8 29.6 32.4 32.0 32.4<br />

85 PRSS12-Hs00186221_m1 Core up 29.9 34.6 34.8 32.1 30.8 34.2 33.4 29.7 33.4 33.8 40.0 29.9 29.6 28.0 27.4 29.4 28.5 30.2 34.2 33.1 25.6 26.8 23.8 24.3<br />

86 FOXJ1-Hs00230964_m1 Core down 35.7 32.9 32.7 26.7 33.7 30.5 26.3 34.8 30.2 33.4 35.5 30.5 29.4 32.2 30.0 31.3 31.2 32.5 31.3 31.0 37.0 40.0 33.0 33.1<br />

87 NTN1-Hs00180355_m1 Core down 30.2 29.3 29.3 26.4 30.4 27.7 25.9 29.5 28.6 29.0 26.9 25.2 25.8 25.5 29.6 30.9 28.9 28.9 28.7 29.5 33.4 32.9 27.5 29.1<br />

88 LMO3-Hs00375237_m1 Core down 29.3 28.5 29.0 34.5 40.0 35.4 30.7 29.6 29.0 30.4 33.9 28.7 25.5 28.4 40.0 40.0 31.2 35.2 28.0 27.9 35.6 36.9 40.0 40.0<br />

89 CHI3L1-Hs00609691_m1 Marker 34.1 31.7 32.4 34.1 35.5 31.4 33.3 40.0 36.0 34.1 32.0 31.8 30.8 29.8 28.7 32.2 22.4 27.3 29.4 26.4 23.0 25.4 30.0 31.1<br />

90 CD9-Hs01124025_g1 Core up 27.5 30.4 30.9 24.9 24.3 26.1 24.1 27.7 29.8 29.9 27.1 27.3 28.0 26.7 21.1 22.2 22.4 22.4 22.8 21.2 24.1 23.8 22.9 23.4<br />

91 ADD2-Hs00242289_m1 Core up 25.9 27.4 27.8 27.1 27.3 29.2 27.8 24.9 29.4 28.8 28.1 37.0 30.3 31.8 27.2 28.9 24.6 25.0 27.7 26.7 28.2 27.9 26.3 26.6<br />

92 MYL9-Hs00697086_m1 Core down 25.4 24.2 24.8 20.9 22.6 24.2 23.6 26.5 30.1 28.8 24.1 22.5 25.5 23.7 31.7 35.8 28.4 33.2 28.6 25.9 22.3 24.2 32.4 33.0<br />

93 LYST-Hs00179814_m1 Core up 31.0 30.1 30.3 30.6 30.6 31.4 29.3 30.3 31.7 31.5 31.1 30.7 32.7 31.5 28.2 28.7 28.4 28.7 30.5 28.5 28.9 30.0 29.1 29.9<br />

94 LAMA2-Hs00166308_m1 Core down 35.3 40.0 37.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 31.1 28.6 26.7 28.7 32.0 35.2 27.7 32.8 27.9 25.2 25.8 29.3 30.6 30.4<br />

95 PLA2G4A-Hs00233352_m1 Core up 29.2 33.2 33.1 36.0 40.0 33.0 33.1 29.2 33.6 31.8 29.9 34.9 32.1 32.4 29.0 30.2 28.6 29.6 31.0 30.7 29.3 28.9 28.4 28.5<br />

96 BACE2-Hs00273238_m1 Core up 26.5 26.7 27.1 26.7 26.2 28.3 26.8 26.2 29.1 28.2 34.0 31.7 31.1 30.5 26.0 26.5 26.6 26.7 28.8 26.3 26.3 25.5 24.5 25.0<br />

258


A.3 Quantitative RT-PCR Appendix<br />

Well Applied Biosystems assay ID Gene G19 G19 G21 G21 G23 G23 G24 G24 G25 G25 G26 G26 G30 G30 G31 G31 G32 G32 G144 G144 G166 G166 G179 G179<br />

Category A B A B A B A B A B A B A B A B A B A B A B A B<br />

1 MYL9-Hs00382913_m1 Core down 34.0 36.1 33.8 33.1 29.1 31.9 27.7 27.4 30.7 29.9 28.8 29.9 34.1 33.6 35.1 34.8 31.1 32.2 34.2 33.9 28.3 28.6 31.9 31.4<br />

2 CHCHD10-Hs01369775_g1 Core up 26.2 27.2 26.9 27.3 25.8 25.9 26.1 26.0 26.5 25.6 25.2 26.3 26.6 26.7 26.5 26.5 27.0 26.6 27.6 26.7 25.3 24.8 24.6 24.0<br />

3 RGS5-Hs00186212_m1 Core down 31.0 31.6 32.1 32.9 30.0 29.9 31.3 31.5 30.3 30.0 30.2 32.3 30.0 30.1 30.0 30.5 30.1 30.6 36.2 33.0 30.4 31.4 31.1 30.1<br />

4 ST6GALNAC5-<br />

Core down 29.7 31.1 25.5 26.1 32.9 30.3 33.1 40.0 35.9 34.3 29.9 31.4 33.5 33.7 27.1 27.0 28.8 28.6 36.0 35.9 30.9 33.5 34.5 33.2<br />

Hs00229612_m1<br />

5 CEBPB-Hs00270923_s1 Core up 30.6 30.4 30.0 30.7 28.2 27.8 29.9 30.2 29.8 29.5 29.7 30.5 29.9 29.7 29.1 29.2 29.7 29.9 32.2 31.5 28.4 28.7 28.1 28.2<br />

6 C5orf13-Hs00854282_g1 Core down 29.1 28.1 27.2 27.8 28.2 25.2 21.6 24.4 26.3 26.7 40.0 29.4 26.4 25.0 40.0 26.3 40.0 27.3 26.9 40.0 26.1 28.0 28.7 28.2<br />

7 PDGFRA-Hs00998026_m1 Marker 24.7 25.0 22.5 22.3 25.3 27.3 24.9 24.4 26.5 26.0 23.8 26.9 23.6 23.6 27.5 27.8 27.3 28.9 26.7 26.2 25.2 28.6 26.5 25.9<br />

8 CCND2-Hs00922419_g1 Core down 35.0 30.5 21.6 22.0 27.9 27.4 30.0 32.4 27.4 26.8 24.5 26.0 22.1 22.0 20.7 20.9 20.6 21.2 25.1 25.3 32.4 32.0 32.4 31.6<br />

9 NKX2-1-Hs00163037_m1 Core down 31.1 31.5 35.3 35.3 40.0 34.8 35.8 40.0 40.0 40.0 40.0 40.0 40.0 40.0 31.2 31.7 32.2 31.0 34.7 36.9 40.0 40.0 40.0 40.0<br />

10 CTSC-Hs00175188_m1 Core up 21.9 23.2 24.2 25.5 21.6 20.7 26.0 25.5 22.5 22.2 20.5 22.1 24.0 23.6 24.4 24.5 24.3 24.5 23.9 22.8 21.2 22.8 24.3 23.1<br />

11 18S-Hs99999901_s1 Norm 12.9 13.1 12.0 12.4 11.9 12.2 11.7 13.0 12.3 11.9 11.7 12.2 12.1 11.9 11.4 11.9 11.6 12.0 14.5 13.8 12.0 13.8 11.8 11.4<br />

12 DNER-Hs00294564_m1 Core down 31.4 28.4 27.3 26.4 32.7 32.4 27.7 27.2 26.6 26.7 25.3 27.9 24.2 24.0 23.9 24.5 25.0 26.1 31.3 28.6 27.0 30.3 27.9 27.0<br />

13 LMO4-Hs01086790_m1 Core up 24.5 25.3 21.9 22.3 23.4 23.6 24.4 24.1 24.4 23.8 23.3 25.4 22.8 22.5 22.4 22.6 22.9 22.9 25.7 24.7 23.7 24.1 24.4 23.2<br />

14 PLCH1-Hs00392783_m1 Core down 26.0 26.3 27.1 27.7 27.0 28.7 28.1 28.5 31.6 30.5 29.4 29.9 29.6 29.3 27.3 27.4 27.5 27.0 32.5 31.5 28.8 29.6 31.2 31.0<br />

15 SPARCL1-Hs00949886_m1 Core down 26.9 25.6 26.4 27.4 26.2 28.9 25.1 24.0 27.5 27.3 24.8 29.3 24.3 24.0 26.5 26.4 27.0 28.9 31.2 31.3 27.9 30.5 26.3 25.2<br />

16 SYNM-Hs00322391_m1 Core down 34.0 33.4 27.9 28.2 27.7 27.6 27.2 28.3 36.0 40.0 29.4 29.3 40.0 36.1 31.9 33.3 31.7 32.0 40.0 40.0 36.8 40.0 29.6 29.0<br />

17 FBLN2-Hs00157482_m1 Core down 40.0 35.5 31.0 31.9 30.1 30.7 36.7 35.9 32.8 33.9 34.3 36.2 34.1 34.6 34.3 34.5 33.9 35.4 34.3 35.4 36.5 34.7 28.7 28.6<br />

18 SALL2-Hs00413788_m1 Core down 29.9 28.1 26.6 26.3 34.3 36.9 35.1 33.2 32.0 31.0 29.5 33.1 26.3 26.2 26.6 26.5 26.8 27.2 30.2 28.6 40.0 40.0 33.9 33.9<br />

19 TUSC3-Hs00185147_m1 Core down 23.2 23.5 40.0 40.0 24.4 25.0 34.1 32.9 37.0 35.0 40.0 40.0 24.9 24.9 40.0 40.0 30.0 30.2 40.0 40.0 32.0 28.8 26.5 25.3<br />

20 GPR158-Hs00393109_m1 Core down 30.6 30.0 27.9 28.2 40.0 40.0 32.3 32.7 31.1 31.6 28.2 30.4 30.5 31.2 28.7 29.3 29.1 29.2 28.2 28.9 31.4 33.3 31.0 30.9<br />

21 PEG3-Hs00377844_m1 Core down 28.9 28.9 23.6 24.9 32.9 33.9 27.8 28.5 27.3 27.3 27.6 30.1 25.2 25.6 40.0 40.0 29.1 32.0 29.5 28.7 29.8 31.4 40.0 40.0<br />

22 FUT8-Hs00189535_m1 Core up 27.4 28.2 25.1 25.1 27.0 27.2 27.1 27.3 26.0 26.0 27.3 28.5 26.2 26.3 26.5 26.2 26.1 26.5 28.3 27.4 25.0 26.0 27.3 26.2<br />

23 HLA-DRA-Hs00219575_m1 Core up 27.0 26.1 33.4 35.0 35.3 40.0 27.9 27.7 22.4 22.9 23.7 24.5 24.0 23.9 33.7 32.0 28.0 30.5 24.4 22.8 28.3 26.6 22.2 21.1<br />

24 ASCL1-Hs00269932_m1 Marker 36.9 30.7 23.1 23.4 28.7 30.4 29.4 33.9 35.4 40.0 36.4 40.0 23.4 23.4 21.9 22.5 22.2 22.6 28.1 26.5 35.1 40.0 31.6 31.3<br />

25 PI15-Hs00210658_m1 Core down 28.9 30.8 32.3 28.9 30.6 32.2 28.9 26.6 27.5 27.6 27.8 26.9 31.1 30.6 36.0 37.0 27.7 34.6 32.6 34.1 27.4 30.0 28.2 26.7<br />

26 EPDR1-Hs00378148_m1 Core up 27.6 29.2 30.2 30.8 28.5 29.2 27.6 28.2 26.9 25.8 26.9 28.0 27.9 27.8 29.2 29.1 29.1 28.9 30.8 29.8 27.1 26.8 29.5 28.6<br />

27 DTX4-Hs00392288_m1 Core down 27.8 27.7 28.5 28.0 32.8 33.8 30.4 30.5 30.0 29.4 34.5 34.2 29.8 29.5 26.4 26.4 26.5 27.6 30.4 30.7 31.0 31.7 28.5 28.1<br />

28 CXXC4-Hs00228693_m1 Core down 29.7 28.2 24.3 24.7 29.1 30.9 28.2 29.9 36.9 40.0 34.9 40.0 26.9 26.7 25.4 26.1 25.4 26.9 30.7 29.3 30.4 32.8 36.9 35.0<br />

29 IRX2-Hs01383002_m1 Core down 40.0 40.0 40.0 40.0 28.4 28.7 36.4 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 37.1 34.9 40.0 40.0 40.0 40.0 30.5 30.4<br />

30 MAP6-Hs01023152_s1 Core down 27.9 28.3 26.0 26.7 28.3 29.6 30.9 29.7 30.4 30.8 32.4 32.6 28.4 28.2 29.1 29.4 28.6 29.8 33.7 31.8 30.7 33.7 27.5 26.5<br />

31 SIX3-Hs00193667_m1 Core down 33.0 34.0 40.0 40.0 29.9 30.5 34.5 34.8 36.0 35.9 40.0 40.0 35.1 36.4 34.7 36.1 40.0 34.9 31.6 30.8 33.7 33.1 40.0 40.0<br />

32 NKX2-2-Hs00159616_m1 Marker 28.7 28.4 26.5 27.8 27.5 27.9 26.4 26.8 25.5 24.8 25.8 27.4 24.9 24.6 25.0 25.3 24.8 24.5 27.1 25.9 28.4 28.8 31.0 31.0<br />

33 S100A6-Hs00170953_m1 Core up 18.5 20.1 29.2 27.3 18.3 17.8 19.9 20.0 20.3 19.2 21.4 21.2 21.1 20.9 23.0 22.9 22.7 21.8 24.0 23.8 17.5 17.9 19.3 18.3<br />

34 MMP17-Hs01108847_m1 Core up 28.4 29.6 30.3 30.1 28.6 27.7 28.9 28.8 25.1 24.9 30.1 31.3 26.2 25.9 29.4 28.8 29.2 29.2 29.2 26.4 26.6 26.4 28.6 28.4<br />

35 FAM38B-Hs00926225_m1 Core down 29.0 29.4 40.0 40.0 36.9 36.4 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 32.7 31.6<br />

36 OLIG2-Hs00300164_s1 Marker 34.9 29.1 22.1 22.8 26.8 27.5 25.4 26.1 23.1 22.9 23.1 24.9 21.8 21.2 22.6 22.8 22.6 22.7 24.8 23.6 31.0 33.2 30.5 30.5<br />

37 PMEPA1-Hs00375306_m1 Core up 24.1 25.1 27.9 27.6 25.5 26.4 22.4 23.0 25.4 24.3 26.7 27.3 25.2 24.6 27.1 26.9 27.4 26.9 27.3 27.7 22.6 25.0 26.0 25.7<br />

38 HOXD10-Hs00157974_m1 Core up 28.5 28.5 40.0 40.0 27.7 28.6 29.4 29.5 29.5 28.9 29.5 31.2 28.9 28.6 28.8 28.9 28.8 29.0 30.7 28.8 26.5 27.0 27.9 27.2<br />

39 NDN-Hs00267349_s1 Core down 23.8 24.2 29.4 30.1 40.0 40.0 25.9 25.8 25.9 25.7 27.1 27.1 36.9 40.0 25.4 25.1 25.5 25.0 28.6 27.0 35.7 40.0 40.0 35.2<br />

40 FOXG1-Hs01850784_s1 Core up 24.4 24.2 22.9 23.0 24.6 25.3 24.3 25.3 25.5 24.8 26.3 26.3 23.5 23.5 21.4 21.6 21.4 21.8 26.3 24.7 23.8 25.5 21.8 21.3<br />

41 MN1-Hs00159202_m1 Core down 32.2 34.9 28.0 27.9 31.8 33.4 26.4 27.3 30.5 28.9 33.7 35.5 28.0 27.1 26.5 27.2 26.8 26.4 31.6 30.1 34.7 35.0 31.5 30.8<br />

42 NDUFB10-Hs00605903_m1 Norm 25.0 25.6 24.5 25.2 25.3 25.3 25.1 25.7 26.0 25.4 24.7 25.9 25.3 24.9 25.2 25.5 25.4 25.2 28.5 26.6 24.5 25.0 26.4 25.3<br />

43 HMGA2-Hs00171569_m1 Core down 29.9 32.0 33.0 33.2 27.5 27.2 35.4 35.9 32.4 31.8 32.1 33.6 32.4 31.5 33.1 32.8 32.6 32.3 35.8 33.8 31.3 32.1 29.4 28.7<br />

44 MMRN1-Hs00201182_m1 Core down 33.7 31.6 40.0 40.0 33.0 34.6 40.0 40.0 40.0 40.0 32.0 31.5 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 33.7 35.0 33.6 32.2<br />

45 RAB6B-Hs00981572_m1 Core down 28.9 30.7 29.8 29.3 33.2 33.0 27.9 28.0 29.5 29.0 30.7 32.4 28.0 27.4 26.6 26.8 26.7 27.1 31.7 30.2 29.6 30.5 31.0 29.9<br />

46 NTRK2-Hs00178811_m1 Core up 24.0 24.6 29.5 29.7 31.6 32.6 33.2 33.1 23.4 23.4 23.3 26.3 24.3 24.0 34.6 37.0 27.5 31.8 26.4 25.1 22.5 24.8 28.1 27.4<br />

47 KALRN-Hs00610179_m1 Core down 30.5 30.6 27.5 27.4 30.3 31.8 28.5 28.3 30.7 30.1 30.6 31.1 27.2 27.5 27.4 27.5 26.7 27.4 32.2 30.8 27.9 30.9 31.2 31.0<br />

48 CD74-Hs00269961_m1 Core up 27.5 26.4 32.1 35.1 32.7 34.2 27.9 28.2 22.2 23.0 24.2 25.6 24.7 24.4 36.5 33.5 28.3 30.3 24.9 24.7 28.7 26.6 22.3 22.0<br />

49 LUM-Hs00158940_m1 Core down 30.1 31.9 40.0 40.0 23.4 26.0 32.8 27.5 40.0 40.0 31.4 31.2 40.0 40.0 32.3 31.2 34.9 40.0 40.0 40.0 28.8 27.5 30.4 29.1<br />

50 NELL2-Hs00196254_m1 Core down 33.1 33.2 33.2 29.0 31.6 32.1 29.8 29.6 36.9 37.0 32.5 34.4 30.8 31.4 27.0 27.4 28.2 29.1 30.1 28.0 29.1 31.8 32.2 31.1<br />

259


A.3 Quantitative RT-PCR Appendix<br />

Well Applied Biosystems assay ID Gene G19 G19 G21 G21 G23 G23 G24 G24 G25 G25 G26 G26 G30 G30 G31 G31 G32 G32 G144 G144 G166 G166 G179 G179<br />

Category A B A B A B A B A B A B A B A B A B A B A B A B<br />

51 MAP6-Hs01929835_s1 Core down 25.7 26.5 25.0 25.4 28.9 30.1 29.7 28.7 29.5 30.3 31.4 31.9 26.9 27.0 29.7 29.8 28.3 30.0 34.8 31.0 30.9 33.9 27.4 26.4<br />

52 PDE1C-Hs01095694_m1 Core up 24.4 25.4 27.3 28.5 24.9 24.1 23.6 23.6 24.4 23.6 25.5 25.9 26.5 25.8 28.0 27.9 28.0 27.8 27.5 26.4 22.9 23.5 26.3 25.7<br />

53 SULF2-Hs00378697_m1 Core up 23.6 24.8 26.1 25.7 24.0 26.3 22.6 22.4 24.6 23.8 23.6 25.7 23.2 22.5 24.9 24.4 24.9 25.1 27.5 24.7 25.2 25.4 26.8 26.2<br />

54 TUBB-Hs00962420_g1 Norm 20.4 21.8 20.0 20.5 21.2 21.3 21.0 21.7 21.8 20.8 20.9 22.1 20.7 20.5 19.7 20.0 19.9 19.8 23.8 22.4 19.7 20.0 23.2 22.8<br />

55 LMO2-Hs00277106_m1 Core down 30.3 30.5 26.3 25.9 31.1 35.5 30.8 30.3 34.3 34.0 27.3 30.5 25.9 26.8 26.0 25.9 25.8 25.4 32.6 32.6 33.3 35.4 29.8 29.0<br />

56 MAN1C1-Hs00220595_m1 Core up 27.6 27.3 24.9 24.9 31.8 34.1 29.3 29.2 27.2 26.3 24.2 26.2 25.3 25.2 24.9 24.3 25.3 24.8 28.9 27.4 27.1 27.3 26.9 26.7<br />

57 TAGLN-Hs00162558_m1 Core down 28.1 30.1 40.0 33.0 24.7 27.2 21.0 21.5 26.5 25.0 29.1 29.3 25.7 24.4 32.7 30.4 28.5 28.7 29.9 31.4 30.4 29.7 27.7 26.8<br />

58 RTN1-Hs00382515_m1 Core down 24.8 25.3 32.7 32.5 27.5 28.6 29.3 29.1 32.6 31.9 24.9 26.5 25.5 25.2 28.0 27.1 29.0 29.6 29.5 29.3 28.1 30.7 31.6 30.7<br />

59 MAF-Hs00193519_m1 Core down 27.9 28.0 27.8 28.6 27.2 30.1 28.0 28.7 31.4 31.5 26.7 28.8 27.5 27.3 26.9 27.4 27.7 26.8 33.9 33.3 27.1 28.5 35.5 34.6<br />

60 NPTX2-Hs00383983_m1 Core down 29.8 28.3 28.1 31.8 28.7 29.2 34.1 37.0 40.0 34.7 27.2 26.8 31.9 31.2 30.3 28.7 33.4 29.5 34.2 33.3 27.4 29.2 32.6 32.4<br />

61 EDA2R-Hs00939736_m1 Core down 27.2 27.8 27.9 28.2 28.9 26.7 28.0 28.1 35.5 34.8 32.0 32.5 28.5 28.2 34.5 34.4 29.8 31.5 37.0 34.0 31.0 30.1 35.3 34.5<br />

62 SOX10-Hs00366918_m1 Marker 40.0 40.0 40.0 40.0 40.0 40.0 36.5 40.0 24.7 24.9 40.0 40.0 35.1 35.0 40.0 40.0 40.0 40.0 27.1 25.7 40.0 40.0 40.0 40.0<br />

63 SEMA6A-Hs00221174_m1 Core down 26.6 26.3 22.4 23.3 25.6 26.1 23.6 24.8 27.7 27.2 24.6 26.8 23.9 23.8 22.4 22.9 22.8 23.0 27.2 26.2 25.3 27.1 28.0 27.7<br />

64 DDIT3-Hs00358796_g1 Core up 26.7 27.2 27.1 27.7 24.9 22.3 27.1 27.2 27.1 25.9 27.6 29.0 27.0 26.6 21.0 21.1 21.0 21.1 25.0 24.4 24.2 23.7 27.2 26.1<br />

65 CA12-Hs01080909_m1 Core down 30.1 28.6 22.4 23.3 25.5 26.5 31.3 30.4 27.9 26.9 23.5 26.3 26.7 27.8 23.9 23.8 24.0 23.2 28.3 26.3 25.9 26.8 26.3 26.1<br />

66 C9orf125-Hs00260558_m1 Core down 30.7 31.6 40.0 40.0 34.5 37.0 28.7 28.9 32.1 31.9 29.8 31.6 27.0 27.3 40.0 40.0 36.5 36.6 32.0 30.4 31.2 33.9 35.8 34.9<br />

67 SDC2-Hs00299807_m1 Core down 26.0 27.0 35.1 36.0 25.9 26.2 25.2 25.6 25.9 24.9 25.8 26.7 29.0 28.5 28.0 27.6 28.8 28.9 29.2 29.0 26.9 28.5 26.1 25.4<br />

68 SLIT2-Hs00191193_m1 Core down 25.1 24.2 33.3 31.9 25.9 26.1 32.3 30.9 33.8 32.3 31.7 32.2 33.3 34.2 28.9 29.0 26.8 27.2 32.9 33.3 32.1 33.3 31.4 30.7<br />

69 MT2A-Hs02379661_g1 Core up 22.6 23.0 21.7 21.5 20.9 21.9 22.8 23.1 24.0 22.5 20.0 21.9 21.8 22.0 21.4 21.4 21.4 21.7 24.3 22.9 18.8 18.5 20.0 18.9<br />

70 IL17RD-Hs00296982_m1 Core down 26.3 27.0 25.5 26.6 23.7 24.4 26.3 27.0 28.0 27.0 26.6 28.5 26.1 25.9 26.2 26.4 26.2 26.6 29.0 27.8 28.0 28.1 28.0 27.6<br />

71 PDZRN3-Hs00392900_m1 Core down 29.0 28.3 26.4 26.7 33.9 40.0 35.8 40.0 26.7 26.0 27.7 28.6 28.6 28.9 26.3 26.3 26.1 26.0 33.1 32.9 30.8 31.6 29.0 29.0<br />

72 LGALS3-Hs00173587_m1 Core up 24.8 24.8 24.3 24.6 23.2 23.3 26.6 25.6 24.7 24.6 22.5 24.1 24.4 24.3 25.4 24.8 24.4 24.6 27.8 25.9 24.1 23.1 24.5 23.3<br />

73 FAM69A-Hs00961685_m1 Core up 25.0 24.8 23.7 25.2 24.9 27.7 25.3 24.0 25.7 25.5 24.3 25.9 26.4 26.3 25.2 25.4 25.0 25.0 28.1 26.5 26.1 26.0 27.4 26.0<br />

74 TERT-Hs00972656_m1 Marker 31.5 33.0 28.1 26.8 30.4 30.2 31.4 31.7 31.4 30.4 30.5 32.0 29.8 29.3 29.8 29.6 29.5 29.2 31.9 29.4 29.2 29.8 34.7 34.8<br />

75 PLS3-Hs00418605_g1 Core up 21.6 22.7 24.4 25.4 22.8 22.9 21.6 22.8 24.2 23.3 22.5 23.9 24.9 24.4 24.4 24.8 24.8 24.3 26.5 24.6 21.0 22.4 26.3 25.1<br />

76 PTEN-Hs02621230_s1 Core down 28.9 29.7 32.9 30.3 27.9 28.6 28.6 28.8 29.9 29.9 30.3 31.4 28.1 28.0 30.4 30.9 30.4 30.7 40.0 40.0 29.1 31.1 37.0 34.9<br />

77 SALL2-Hs00826674_m1 Core down 33.8 32.6 31.7 33.1 33.6 37.0 33.4 32.5 33.1 32.7 32.8 34.6 32.5 31.7 31.0 31.5 31.1 31.1 35.3 34.6 32.4 31.7 31.4 30.8<br />

78 KCTD12-Hs00540818_s1 Core down 31.9 32.0 29.5 32.0 30.2 31.8 25.8 26.3 27.9 28.1 28.2 29.3 27.7 27.6 29.9 31.1 29.4 30.8 30.8 31.0 29.4 30.0 26.7 26.6<br />

79 TES-Hs00210319_m1 Core down 40.0 40.0 40.0 40.0 34.2 36.5 34.1 36.8 34.1 34.2 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 40.0 36.2 36.1 35.8 33.5<br />

80 SOX2-Hs01053049_s1 Marker 23.7 23.9 22.0 22.7 24.5 24.5 24.2 24.7 23.5 23.5 22.8 24.9 22.7 22.5 22.2 22.4 22.0 22.5 25.9 25.1 24.5 26.9 22.7 21.9<br />

81 LPAR6-Hs00271758_s1 Core down 29.6 29.9 28.9 29.3 29.7 31.2 28.7 29.1 33.3 32.9 30.5 32.3 28.2 28.0 29.0 28.7 29.0 28.7 34.1 32.9 28.8 30.2 32.0 31.4<br />

82 ODZ2-Hs00393060_m1 Core down 25.6 26.4 26.7 28.6 24.8 25.9 28.1 26.9 31.9 31.0 24.8 26.9 30.6 30.3 27.1 26.3 27.9 27.1 29.6 29.4 30.0 32.2 28.0 27.6<br />

83 NNMT-Hs00196287_m1 Core up 27.9 28.3 30.0 30.6 23.8 27.1 32.7 30.3 28.2 26.9 23.7 24.6 27.3 27.1 29.0 28.1 29.9 28.3 27.5 28.0 23.0 23.9 24.3 23.5<br />

84 CACNG8-Hs01100182_m1 Core down 31.7 30.9 27.3 28.6 30.2 29.8 30.5 37.0 33.6 32.9 33.7 34.4 27.0 26.9 30.9 32.6 29.6 29.9 30.3 28.9 30.4 31.8 32.8 33.4<br />

85 PRSS12-Hs00186221_m1 Core up 24.2 25.7 30.3 33.5 24.9 24.5 31.0 31.8 27.8 26.6 25.3 26.9 40.0 37.0 27.4 27.8 26.9 26.8 27.3 25.8 23.3 24.3 31.5 31.3<br />

86 FOXJ1-Hs00230964_m1 Core down 32.5 30.5 30.9 30.9 33.5 36.4 30.6 30.9 30.2 30.8 29.6 31.3 27.9 27.6 30.5 30.7 29.9 30.4 32.9 32.1 35.2 40.0 31.9 31.9<br />

87 NTN1-Hs00180355_m1 Core down 29.2 29.0 26.6 26.2 34.2 35.6 25.4 25.0 28.0 28.0 28.1 30.5 27.1 26.8 25.5 25.9 25.7 25.6 28.0 27.9 30.5 35.0 29.4 30.0<br />

88 LMO3-Hs00375237_m1 Core down 40.0 40.0 32.1 34.8 31.7 33.6 40.0 40.0 37.0 40.0 28.1 28.9 32.1 30.6 27.0 27.9 27.4 28.6 40.0 40.0 40.0 40.0 35.9 34.3<br />

89 CHI3L1-Hs00609691_m1 Marker 31.2 25.9 29.9 27.8 33.1 36.9 24.3 24.0 29.3 30.7 23.8 27.4 21.8 22.8 27.6 27.6 28.3 29.2 40.0 37.0 23.0 28.2 22.3 21.8<br />

90 CD9-Hs01124025_g1 Core up 23.1 24.0 22.4 23.5 24.4 24.3 25.6 25.0 22.4 22.5 22.9 24.6 22.5 22.3 22.6 22.9 22.6 23.0 25.2 23.9 22.3 23.0 24.2 23.0<br />

91 ADD2-Hs00242289_m1 Core up 26.5 27.7 24.0 25.2 26.5 25.2 26.7 27.8 28.6 27.9 30.3 32.1 25.8 25.8 24.9 24.9 24.8 24.8 28.9 27.7 25.6 25.4 29.0 28.8<br />

92 MYL9-Hs00697086_m1 Core down 32.6 33.0 31.4 31.3 27.3 29.7 22.8 23.3 28.6 27.7 26.2 28.0 28.4 27.3 33.8 32.2 30.4 30.8 33.0 32.8 25.7 26.1 27.3 26.9<br />

93 LYST-Hs00179814_m1 Core up 29.8 30.3 29.8 30.6 29.6 28.4 29.3 29.0 28.9 29.2 29.3 31.6 28.3 28.7 28.8 29.2 29.2 29.9 30.1 29.4 28.1 28.0 28.9 27.9<br />

94 LAMA2-Hs00166308_m1 Core down 29.9 29.4 36.9 36.9 28.6 29.7 40.0 36.0 31.5 33.0 32.2 32.5 30.6 31.2 29.6 28.8 35.3 34.7 29.4 28.2 32.7 34.0 34.0 35.0<br />

95 PLA2G4A-Hs00233352_m1 Core up 28.1 29.2 29.3 28.9 29.4 29.4 30.1 29.8 27.1 27.0 31.1 32.1 26.5 26.1 26.5 26.2 26.4 26.0 29.0 28.4 28.6 27.3 29.7 28.9<br />

96 BACE2-Hs00273238_m1 Core up 24.8 25.6 34.7 36.9 29.4 30.4 26.4 26.5 26.0 25.5 26.6 29.2 27.4 27.7 29.9 29.8 29.6 30.0 28.9 27.5 25.8 25.6 27.7 27.1<br />

260


A.3 Quantitative RT-PCR Appendix<br />

Table A.4: Normalised Ct values. Abbreviations: "down" for down-regulated, "up" for up-regulated, and "Norm" for Normalisation control.<br />

Well Applied Biosystems assay ID Gene CB130 CB130 CB130 CB152 CB152 CB152 CB152 CB171 CB171 CB171 CB192 CB541 CB660 CB660 G2 G2 G7 G7 G7 G7 G9 G9 G14 G14<br />

Category A B C A B C D A B C A B A B A B C D A B A B<br />

1 MYL9-Hs00382913_m1 Core down 28.2 27.9 28.2 23.0 25.2 25.6 25.3 29.4 30.2 29.5 26.8 24.9 27.2 26.7 36.1 33.4 31.9 36.0 30.5 29.4 26.3 26.6 35.0 35.2<br />

2 CHCHD10-Hs01369775_g1 Core up 27.1 25.7 26.1 25.9 25.3 24.6 24.5 27.2 26.5 26.1 28.8 29.7 29.3 29.5 24.7 23.1 26.2 25.7 25.6 24.7 25.5 26.3 27.1 26.9<br />

3 RGS5-Hs00186212_m1 Core down 31.3 30.4 30.4 29.2 31.7 30.4 29.3 32.9 29.5 30.8 32.6 27.4 26.5 27.6 30.4 29.4 31.5 30.9 28.4 27.6 30.5 31.8 32.4 32.1<br />

4 ST6GALNAC5-<br />

Core down 28.9 28.8 28.6 33.5 33.7 32.4 31.9 29.8 30.5 29.5 25.6 26.7 23.4 24.9 35.2 33.3 36.4 36.8 34.9 34.7 28.6 28.9 31.1 29.9<br />

Hs00229612_m1<br />

5 CEBPB-Hs00270923_s1 Core up 33.1 30.7 30.6 30.3 31.8 31.2 31.3 32.9 30.3 31.3 30.7 32.0 32.6 31.3 31.6 32.7 29.1 29.8 29.8 29.6 29.6 30.2 30.7 30.1<br />

6 C5orf13-Hs00854282_g1 Core down 28.5 27.7 28.1 25.5 27.3 26.4 25.6 28.5 25.2 26.6 29.1 24.8 25.2 26.2 36.1 25.2 30.3 29.0 22.8 26.9 27.3 36.2 27.8 31.5<br />

7 PDGFRA-Hs00998026_m1 Marker 37.4 37.4 34.8 33.2 36.8 27.9 28.4 37.5 35.4 36.9 32.4 35.7 29.9 32.1 24.2 24.3 23.2 22.2 21.5 22.5 24.0 24.1 25.9 26.2<br />

8 CCND2-Hs00922419_g1 Core down 32.3 30.8 31.0 31.6 33.7 28.4 28.5 32.4 31.1 30.9 25.1 24.1 24.1 24.7 22.5 21.3 22.7 22.0 23.9 23.2 30.4 29.9 34.1 37.3<br />

9 NKX2-1-Hs00163037_m1 Core down 37.4 37.4 37.1 37.1 36.8 36.4 37.2 37.5 35.4 36.9 37.0 28.8 29.0 29.8 36.1 34.4 32.8 34.6 32.2 32.4 36.7 36.2 31.2 31.3<br />

10 CTSC-Hs00175188_m1 Core up 23.5 24.6 24.6 24.6 24.0 24.7 24.3 23.5 24.3 24.0 23.9 25.1 25.5 23.7 20.6 20.0 25.3 24.4 26.5 25.1 23.0 23.5 23.4 23.2<br />

11 18S-Hs99999901_s1 Norm 13.7 12.0 11.5 12.2 13.8 10.9 11.3 13.4 10.8 11.2 12.5 12.8 12.1 12.1 14.0 16.4 12.5 12.8 10.4 11.6 12.6 14.0 12.7 12.6<br />

12 DNER-Hs00294564_m1 Core down 32.3 32.3 32.8 32.7 31.5 28.1 27.5 32.2 29.9 32.0 30.4 22.3 22.4 22.7 25.5 23.3 27.8 25.2 23.5 25.0 26.5 24.0 32.5 31.5<br />

13 LMO4-Hs01086790_m1 Core up 27.0 26.8 26.9 26.7 27.5 26.6 25.4 26.9 26.3 27.1 25.7 26.1 25.5 26.7 22.5 21.3 22.2 22.4 24.0 23.7 24.4 25.7 25.1 25.0<br />

14 PLCH1-Hs00392783_m1 Core down 26.5 26.3 26.2 26.4 26.8 27.3 26.3 26.2 26.4 27.5 26.8 27.0 27.8 28.7 32.6 32.2 29.5 29.9 29.1 28.9 27.9 29.1 27.2 26.8<br />

15 SPARCL1-Hs00949886_m1 Core down 33.6 31.2 31.0 27.0 29.5 24.1 22.2 35.6 28.3 30.3 28.5 25.3 22.8 25.6 24.5 23.1 24.4 27.6 29.1 29.7 22.4 25.4 28.9 28.1<br />

16 SYNM-Hs00322391_m1 Core down 29.3 28.2 28.1 26.8 27.3 27.3 25.7 28.6 27.3 28.6 27.1 27.6 27.1 27.2 28.6 27.7 29.0 29.5 28.7 28.6 25.2 27.8 33.7 34.5<br />

17 FBLN2-Hs00157482_m1 Core down 37.4 36.2 37.1 36.0 36.8 36.4 35.1 37.5 33.6 35.5 33.8 29.1 27.5 29.2 33.5 33.4 35.4 32.7 36.2 35.3 33.2 36.2 36.8 36.2<br />

18 SALL2-Hs00413788_m1 Core down 37.4 33.4 33.5 31.0 36.7 30.1 27.2 37.4 28.7 33.1 32.8 26.5 24.5 27.9 30.5 31.2 28.1 28.5 28.9 30.6 34.6 36.2 31.6 30.3<br />

19 TUSC3-Hs00185147_m1 Core down 24.4 22.8 23.0 23.3 23.6 22.7 21.8 24.4 23.5 23.3 24.4 23.4 23.4 23.9 36.1 34.4 35.7 33.7 36.3 34.2 23.3 22.9 24.0 23.8<br />

20 GPR158-Hs00393109_m1 Core down 34.8 34.5 34.9 37.0 36.3 31.4 32.6 36.0 34.5 33.3 26.9 25.6 27.1 26.6 26.3 26.4 28.2 26.6 27.9 26.6 34.6 34.8 32.6 32.3<br />

21 PEG3-Hs00377844_m1 Core down 31.0 31.4 31.2 30.0 30.7 30.3 29.4 30.4 29.6 30.9 29.8 28.1 25.2 27.9 27.2 27.0 26.5 24.6 25.6 26.3 33.3 35.8 29.0 29.0<br />

22 FUT8-Hs00189535_m1 Core up 26.6 26.7 26.8 26.3 26.7 27.4 26.4 27.0 27.1 27.1 26.4 27.9 28.4 27.1 25.2 24.0 27.9 27.3 26.5 27.3 25.6 26.5 28.5 28.3<br />

23 HLA-DRA-Hs00219575_m1 Core up 27.8 26.5 26.4 28.3 29.8 25.3 22.3 29.1 25.4 27.4 22.3 27.2 30.8 26.4 20.5 20.6 24.7 24.0 23.9 23.6 21.1 25.2 27.6 28.0<br />

24 ASCL1-Hs00269932_m1 Marker 34.3 32.4 31.9 32.8 35.7 27.5 26.3 35.0 30.5 35.0 32.7 29.4 27.1 34.4 23.5 22.4 25.9 24.1 23.7 25.8 36.7 36.2 34.3 37.4<br />

25 PI15-Hs00210658_m1 Core down 32.0 30.7 30.3 23.1 28.3 26.5 24.6 32.3 27.0 30.9 29.6 25.0 24.5 24.8 25.4 26.0 32.3 27.4 29.1 30.0 26.6 32.9 32.0 30.5<br />

26 EPDR1-Hs00378148_m1 Core up 29.0 28.3 28.8 26.6 27.3 28.8 26.9 29.5 28.9 29.0 30.8 29.0 30.0 30.0 26.0 25.2 28.6 28.8 28.7 28.2 26.6 28.1 28.8 28.8<br />

27 DTX4-Hs00392288_m1 Core down 33.2 31.1 31.3 28.3 31.4 28.3 27.2 33.6 27.4 30.2 32.9 26.5 25.0 26.7 33.3 34.4 30.8 30.6 31.3 33.3 29.5 32.3 28.4 28.0<br />

28 CXXC4-Hs00228693_m1 Core down 31.3 31.2 30.8 34.2 36.8 30.2 29.6 32.1 29.7 33.1 28.3 25.9 25.7 27.9 27.7 24.9 27.4 26.5 25.5 27.5 36.7 36.2 29.3 30.3<br />

29 IRX2-Hs01383002_m1 Core down 30.4 30.9 30.7 29.3 28.9 30.8 30.1 29.7 30.3 31.1 30.7 26.6 26.6 26.3 28.9 27.3 37.4 36.8 36.3 37.7 29.9 29.5 37.9 37.4<br />

30 MAP6-Hs01023152_s1 Core down 28.0 25.1 25.4 25.9 27.4 24.1 22.0 27.5 24.1 24.7 28.1 26.9 25.8 28.0 25.9 24.1 27.4 26.7 26.8 27.2 28.3 29.8 29.2 29.1<br />

31 SIX3-Hs00193667_m1 Core down 37.4 37.4 37.1 37.1 36.8 36.4 34.8 37.5 35.4 36.9 37.0 25.5 24.0 25.0 36.1 32.6 32.5 33.6 29.8 32.3 32.3 32.9 35.1 34.7<br />

32 NKX2-2-Hs00159616_m1 Marker 37.4 37.4 37.1 37.1 36.8 32.6 33.8 37.5 35.4 36.9 37.0 28.6 30.8 29.6 25.0 23.7 27.2 26.1 28.2 30.0 26.5 26.4 28.9 28.2<br />

33 S100A6-Hs00170953_m1 Core up 20.0 20.4 20.8 20.9 18.9 20.0 20.8 19.5 21.7 20.5 21.9 21.1 25.7 23.0 18.8 17.3 21.7 22.7 21.6 19.8 19.5 17.8 19.7 19.2<br />

34 MMP17-Hs01108847_m1 Core up 28.3 28.7 28.6 28.4 27.5 29.7 29.1 28.0 29.3 30.0 28.6 32.3 30.3 29.9 25.7 27.4 30.4 29.8 29.1 30.8 27.4 26.4 28.9 28.5<br />

35 FAM38B-Hs00926225_m1 Core down 37.4 37.4 37.1 37.1 36.8 36.4 37.2 37.5 35.4 36.9 37.0 24.3 27.0 26.1 36.1 34.4 37.4 36.8 36.3 37.7 24.4 24.5 29.3 29.1<br />

36 OLIG2-Hs00300164_s1 Marker 30.2 33.1 33.2 30.1 30.0 28.3 28.4 32.5 34.3 35.4 29.2 26.1 28.1 27.8 23.4 23.9 22.2 22.0 21.5 23.4 30.2 32.0 31.4 36.4<br />

37 PMEPA1-Hs00375306_m1 Core up 26.0 24.0 24.1 24.8 24.9 25.2 26.0 26.1 26.5 25.9 27.6 26.5 29.1 27.2 25.5 25.3 27.7 27.7 27.5 27.3 25.9 24.3 24.0 24.6<br />

38 HOXD10-Hs00157974_m1 Core up 37.4 37.4 37.1 37.1 36.8 32.5 32.4 37.5 35.4 36.9 37.0 37.5 37.1 37.9 29.1 27.4 27.4 27.7 28.1 27.4 28.0 28.1 28.7 28.7<br />

39 NDN-Hs00267349_s1 Core down 25.1 25.0 25.2 24.2 24.1 24.7 23.4 25.0 25.3 25.1 25.4 23.8 23.9 24.7 25.0 23.1 25.0 25.4 25.9 26.1 23.4 24.1 24.7 24.3<br />

40 FOXG1-Hs01850784_s1 Core up 37.4 37.4 37.1 33.7 34.7 25.3 25.3 37.5 31.1 35.8 35.3 27.8 27.6 28.8 25.0 23.5 23.9 23.9 21.4 21.4 24.0 24.4 24.6 24.4<br />

41 MN1-Hs00159202_m1 Core down 32.3 31.8 31.5 29.4 30.3 31.2 27.8 33.6 31.8 34.1 31.0 26.3 25.9 27.0 28.7 28.5 30.1 28.2 28.8 28.5 34.0 32.3 33.4 33.3<br />

42 NDUFB10-Hs00605903_m1 Norm 25.1 25.5 26.0 25.6 25.0 25.8 25.0 25.5 26.0 25.8 25.4 25.6 26.0 26.0 24.6 22.4 25.5 25.5 26.4 26.2 25.2 24.4 25.0 25.4<br />

43 HMGA2-Hs00171569_m1 Core down 31.8 28.3 28.5 33.6 30.3 29.1 29.7 30.3 28.2 28.8 29.0 27.3 27.1 26.8 31.0 29.5 31.3 30.3 28.9 28.9 34.1 30.5 30.4 31.0<br />

44 MMRN1-Hs00201182_m1 Core down 37.4 37.4 37.1 37.1 36.8 36.4 37.2 37.5 35.4 36.9 37.0 26.4 27.3 28.6 36.1 34.4 37.0 36.2 36.3 36.7 36.7 36.1 33.2 34.3<br />

45 RAB6B-Hs00981572_m1 Core down 34.8 32.4 32.3 32.9 35.1 31.7 29.7 34.6 31.1 32.4 30.4 28.3 28.6 29.0 28.5 28.1 27.6 27.7 27.3 28.4 29.3 29.4 30.2 29.3<br />

46 NTRK2-Hs00178811_m1 Core up 29.3 30.9 31.1 23.4 24.7 25.5 23.1 29.7 28.5 30.9 29.0 29.0 26.9 28.6 21.5 20.8 29.5 24.8 26.9 27.5 22.4 24.2 25.2 24.0<br />

47 KALRN-Hs00610179_m1 Core down 29.9 29.5 30.1 28.1 29.5 30.3 29.0 30.0 28.4 30.0 28.3 26.5 25.9 26.5 27.7 28.5 28.7 27.8 27.4 28.2 26.8 28.9 30.8 30.6<br />

48 CD74-Hs00269961_m1 Core up 27.6 27.7 27.5 28.0 28.7 27.6 24.2 27.8 25.7 28.8 22.2 27.4 30.3 26.3 20.8 21.6 25.7 23.7 25.7 25.5 21.6 26.3 27.8 28.3<br />

261


A.3 Quantitative RT-PCR Appendix<br />

Well Applied Biosystems assay ID Gene CB130 CB130 CB130 CB152 CB152 CB152 CB152 CB171 CB171 CB171 CB192 CB541 CB660 CB660 G2 G2 G7 G7 G7 G7 G9 G9 G14 G14<br />

Category A B C A B C D A B C A B A B A B C D A B A B<br />

49 LUM-Hs00158940_m1 Core down 29.7 29.3 29.3 30.4 31.7 36.3 32.2 32.1 31.8 32.7 29.9 26.0 24.7 25.5 36.1 34.4 32.9 28.8 32.3 31.7 22.7 26.5 30.4 30.0<br />

50 NELL2-Hs00196254_m1 Core down 24.8 24.8 24.8 28.1 29.1 28.5 25.1 24.8 24.0 25.3 25.4 25.0 23.9 25.8 29.6 28.4 30.6 30.5 31.0 31.4 28.4 29.2 35.1 34.4<br />

51 MAP6-Hs01929835_s1 Core down 27.4 25.7 26.2 25.7 26.4 24.4 22.6 27.0 24.4 25.0 27.5 26.6 25.7 28.7 24.9 23.4 27.2 26.2 27.8 28.4 27.0 28.5 27.2 26.9<br />

52 PDE1C-Hs01095694_m1 Core up 30.3 32.8 33.1 27.1 27.6 28.4 28.2 29.7 29.8 30.7 30.4 26.7 28.0 26.7 25.4 25.5 24.7 24.4 24.7 24.0 23.5 26.3 25.1 24.8<br />

53 SULF2-Hs00378697_m1 Core up 28.9 30.1 30.3 26.0 26.6 28.5 25.6 28.4 29.3 31.2 26.9 26.9 27.9 27.5 22.0 21.3 23.7 22.9 24.6 25.1 23.7 22.9 24.8 24.2<br />

54 TUBB-Hs00962420_g1 Norm 20.3 21.7 21.7 21.3 20.4 22.5 22.9 20.2 22.4 22.2 21.3 20.8 21.1 21.1 20.6 20.4 21.2 20.9 22.3 21.3 21.4 20.7 21.4 21.2<br />

55 LMO2-Hs00277106_m1 Core down 37.2 37.4 33.8 35.5 33.8 29.3 29.3 36.4 33.3 33.1 30.5 26.6 25.9 27.2 25.8 24.6 25.8 26.4 26.4 27.5 27.7 30.0 32.6 32.3<br />

56 MAN1C1-Hs00220595_m1 Core up 28.1 27.4 27.2 26.0 28.7 27.1 25.9 27.7 25.6 26.9 27.2 31.6 29.1 29.8 25.9 26.4 26.4 26.6 26.8 27.1 24.9 26.6 28.4 27.8<br />

57 TAGLN-Hs00162558_m1 Core down 21.5 19.3 18.6 20.6 21.7 19.5 22.1 22.4 19.1 19.9 21.9 21.6 23.7 22.6 26.4 27.1 37.4 36.7 36.3 30.0 21.3 21.3 29.5 29.3<br />

58 RTN1-Hs00382515_m1 Core down 26.7 23.4 23.3 29.8 28.6 28.3 26.2 27.7 26.0 26.8 23.8 24.3 25.6 26.2 25.2 23.5 23.8 24.2 25.6 24.9 25.8 26.5 25.6 25.6<br />

59 MAF-Hs00193519_m1 Core down 26.9 26.3 26.8 26.5 25.6 26.8 26.2 26.9 28.1 27.6 27.1 25.7 26.6 26.9 26.5 24.4 28.2 29.1 27.2 25.9 29.7 29.7 28.8 28.6<br />

60 NPTX2-Hs00383983_m1 Core down 35.6 31.0 30.6 30.6 26.6 32.1 30.0 37.5 35.3 35.3 33.6 23.4 25.7 24.5 36.1 34.4 26.6 29.2 30.7 29.2 30.7 26.0 30.5 29.6<br />

61 EDA2R-Hs00939736_m1 Core down 28.2 28.4 28.1 27.7 27.0 28.3 27.0 28.0 28.0 28.1 28.6 27.1 28.5 27.8 26.9 26.6 30.7 34.1 32.8 31.6 27.9 28.1 27.1 27.1<br />

62 SOX10-Hs00366918_m1 Marker 37.4 37.4 37.1 37.1 36.8 34.6 33.0 37.5 35.4 36.9 37.0 37.5 35.2 37.9 27.0 25.4 37.4 36.8 35.2 36.7 36.7 36.2 37.9 37.4<br />

63 SEMA6A-Hs00221174_m1 Core down 25.4 26.4 26.1 24.4 25.0 25.6 24.5 25.1 25.2 26.4 22.7 23.3 23.1 24.2 24.7 23.8 25.4 25.0 25.9 27.0 26.3 29.4 27.1 27.7<br />

64 DDIT3-Hs00358796_g1 Core up 28.3 29.4 29.3 28.5 27.4 27.3 27.5 29.0 28.7 28.5 28.7 28.1 28.1 28.3 26.4 24.6 26.3 25.7 28.1 26.9 26.3 26.3 27.3 27.3<br />

65 CA12-Hs01080909_m1 Core down 30.5 32.7 32.7 31.0 28.1 28.9 31.2 31.4 31.0 31.4 21.5 22.6 23.6 23.4 24.5 23.5 23.8 22.9 24.2 23.0 23.8 25.6 31.3 31.2<br />

66 C9orf125-Hs00260558_m1 Core down 33.2 32.7 32.9 36.8 36.8 33.9 35.0 34.2 33.7 36.3 29.4 29.0 28.3 29.9 27.0 27.6 29.4 28.3 29.4 30.1 28.0 27.4 31.3 31.4<br />

67 SDC2-Hs00299807_m1 Core down 25.1 24.9 24.8 24.3 24.3 24.4 23.5 25.7 24.8 25.1 25.5 25.5 25.9 26.3 26.5 24.4 27.0 28.2 27.5 27.4 26.1 27.1 27.0 26.9<br />

68 SLIT2-Hs00191193_m1 Core down 29.7 33.5 33.0 29.0 28.5 30.8 29.6 30.3 31.4 32.8 31.8 24.5 24.3 25.2 36.1 33.9 28.3 27.4 31.2 31.4 26.1 27.5 26.0 25.9<br />

69 MT2A-Hs02379661_g1 Core up 25.5 24.0 24.4 23.1 22.2 20.2 20.6 26.0 23.7 23.9 23.9 23.8 24.6 25.0 21.6 19.7 20.8 21.2 21.7 20.7 18.8 18.2 23.4 22.7<br />

70 IL17RD-Hs00296982_m1 Core down 27.1 26.1 25.8 25.9 26.7 26.5 26.4 26.3 26.7 26.8 26.3 24.3 24.5 24.6 26.5 26.2 28.1 26.8 26.8 27.6 24.0 24.0 26.8 26.4<br />

71 PDZRN3-Hs00392900_m1 Core down 29.3 29.3 29.1 27.3 28.6 28.9 26.8 28.8 26.1 28.7 28.4 26.9 26.8 27.6 28.8 29.9 27.5 28.9 28.6 30.2 27.6 30.1 30.2 30.0<br />

72 LGALS3-Hs00173587_m1 Core up 26.1 24.9 25.5 24.9 23.8 23.8 23.2 26.1 25.0 25.1 26.6 26.3 25.7 25.8 24.1 21.4 26.8 27.5 25.5 25.8 22.4 22.7 26.4 25.7<br />

73 FAM69A-Hs00961685_m1 Core up 25.7 25.5 25.3 23.9 24.4 25.9 23.7 25.2 24.8 26.2 26.4 28.1 29.8 28.8 24.7 23.4 26.2 26.3 26.6 26.6 24.6 25.1 26.2 25.7<br />

74 TERT-Hs00972656_m1 Marker 37.4 37.4 37.1 37.1 36.8 36.4 37.2 37.5 35.4 36.9 37.0 37.2 36.6 36.3 32.6 31.8 32.8 32.0 32.0 32.6 33.1 32.6 31.9 34.4<br />

75 PLS3-Hs00418605_g1 Core up 25.0 28.0 28.0 25.1 23.9 26.1 26.6 24.9 28.1 28.8 22.9 26.6 25.7 25.0 22.5 21.8 25.7 26.4 26.2 24.9 22.2 22.3 22.7 22.4<br />

76 PTEN-Hs02621230_s1 Core down 30.4 27.9 27.7 29.5 30.3 27.4 26.6 29.3 26.7 27.5 29.6 28.1 27.7 28.1 36.1 34.4 30.3 30.7 27.1 28.0 36.7 36.2 28.7 28.4<br />

77 SALL2-Hs00826674_m1 Core down 34.7 32.2 32.0 32.3 33.7 32.4 29.9 35.3 31.0 33.0 34.9 33.0 32.5 33.4 30.6 29.6 31.2 31.2 32.5 29.4 32.8 34.1 35.0 34.3<br />

78 KCTD12-Hs00540818_s1 Core down 31.0 30.7 30.4 33.4 33.7 32.1 33.4 30.1 28.1 29.7 26.5 25.8 27.4 27.0 26.8 27.4 31.2 32.1 34.8 32.8 26.9 28.9 34.4 32.7<br />

79 TES-Hs00210319_m1 Core down 26.0 26.0 26.2 27.8 27.1 28.2 28.6 26.2 27.5 26.8 32.6 25.6 26.2 26.5 36.1 34.0 37.4 36.8 36.3 37.7 33.0 36.2 36.7 37.4<br />

80 SOX2-Hs01053049_s1 Marker 25.6 21.7 21.8 24.3 26.0 22.2 20.6 24.8 20.8 22.0 24.4 23.6 23.6 24.3 23.7 22.4 23.4 23.2 21.9 22.4 25.0 27.1 24.6 24.1<br />

81 LPAR6-Hs00271758_s1 Core down 31.3 29.4 29.6 34.5 34.0 31.5 30.9 31.6 29.2 30.0 29.9 28.4 28.6 29.4 31.0 29.3 28.6 29.6 28.7 28.5 27.4 28.8 30.1 30.2<br />

82 ODZ2-Hs00393060_m1 Core down 28.7 27.8 27.6 26.8 27.7 27.4 27.0 29.2 28.3 29.4 25.7 27.4 25.1 25.6 29.8 29.0 25.1 26.2 27.1 26.8 27.5 29.3 26.3 26.2<br />

83 NNMT-Hs00196287_m1 Core up 31.3 27.9 28.0 29.0 31.9 29.5 29.8 32.5 29.3 28.4 30.0 28.2 29.6 28.8 30.0 30.0 24.1 25.9 23.7 22.9 21.9 22.9 29.7 27.4<br />

84 CACNG8-Hs01100182_m1 Core down 37.4 33.4 32.8 32.8 36.8 33.1 34.7 34.0 33.1 35.3 28.7 27.8 26.3 28.2 28.8 27.8 27.2 28.5 29.3 29.5 29.2 31.6 32.9 32.7<br />

85 PRSS12-Hs00186221_m1 Core up 30.2 35.0 35.0 32.2 30.6 33.6 33.6 30.2 31.8 33.7 37.0 30.4 29.7 28.9 26.5 26.8 28.9 30.0 33.5 33.8 25.3 26.0 24.7 24.7<br />

86 FOXJ1-Hs00230964_m1 Core down 36.1 33.3 32.8 26.8 33.4 29.8 26.5 35.3 28.5 33.4 35.6 31.0 29.5 33.0 29.1 28.7 31.6 32.3 30.5 31.7 36.7 36.2 33.9 33.5<br />

87 NTN1-Hs00180355_m1 Core down 30.6 29.7 29.4 26.5 30.2 27.1 26.1 30.0 27.0 28.9 27.0 25.7 25.9 26.4 28.7 28.3 29.3 28.7 27.9 30.2 33.1 32.1 28.4 29.5<br />

88 LMO3-Hs00375237_m1 Core down 29.6 28.9 29.1 34.7 36.8 34.8 30.9 30.1 27.4 30.3 33.9 29.2 25.7 29.2 36.1 34.4 31.6 35.0 27.3 28.6 35.3 36.1 37.9 37.4<br />

89 CHI3L1-Hs00609691_m1 Marker 34.4 32.1 32.5 34.2 35.3 30.8 33.5 37.5 34.4 34.0 32.0 32.3 30.9 30.7 27.7 29.6 22.8 27.1 28.6 27.1 22.7 24.6 30.9 31.5<br />

90 CD9-Hs01124025_g1 Core up 27.8 30.8 31.1 25.0 24.1 25.5 24.4 28.2 28.2 29.8 27.1 27.8 28.1 27.6 20.1 19.6 22.8 22.2 22.1 21.9 23.8 23.0 23.8 23.8<br />

91 ADD2-Hs00242289_m1 Core up 26.2 27.8 27.9 27.2 27.1 28.6 28.0 25.4 27.8 28.8 28.1 37.5 30.4 32.6 26.3 26.3 25.0 24.8 26.9 27.4 27.8 27.1 27.2 27.0<br />

92 MYL9-Hs00697086_m1 Core down 25.8 24.7 24.9 21.1 22.4 23.5 23.8 27.0 28.4 28.7 24.1 23.0 25.6 24.5 30.7 33.3 28.8 33.0 27.9 26.6 22.0 23.4 33.3 33.4<br />

93 LYST-Hs00179814_m1 Core up 31.4 30.5 30.4 30.8 30.3 30.8 29.5 30.8 30.1 31.4 31.1 31.1 32.8 32.4 27.3 26.1 28.8 28.5 29.7 29.2 28.5 29.2 29.9 30.3<br />

94 LAMA2-Hs00166308_m1 Core down 35.6 37.4 37.1 37.1 36.8 36.4 37.2 37.5 35.4 36.9 31.1 29.1 26.9 29.5 31.1 32.6 28.1 32.6 27.1 25.9 25.5 28.5 31.4 30.7<br />

95 PLA2G4A-Hs00233352_m1 Core up 29.6 33.6 33.2 36.1 36.8 32.3 33.3 29.7 32.0 31.7 29.9 35.4 32.3 33.3 28.1 27.7 29.0 29.4 30.2 31.4 28.9 28.1 29.3 28.9<br />

96 BACE2-Hs00273238_m1 Core up 26.9 27.1 27.2 26.9 26.0 27.7 27.1 26.6 27.5 28.1 34.1 32.2 31.3 31.3 25.0 23.9 27.0 26.5 28.0 27.0 26.0 24.7 25.4 25.4<br />

262


A.3 Quantitative RT-PCR Appendix<br />

Well Applied Biosystems assay ID Gene G19 G19 G21 G21 G23 G23 G24 G24 G25 G25 G26 G26 G30 G30 G31 G31 G32 G32 G144 G144 G166 G166 G179 G179<br />

Category A B A B A B A B A B A B A B A B A B A B A B A B<br />

1 MYL9-Hs00382913_m1 Core down 34.3 35.7 34.7 33.4 29.4 32.1 28.2 27.1 30.4 30.2 29.5 29.6 34.4 34.2 36.0 35.4 31.9 32.9 31.7 32.8 29.3 28.7 31.1 31.3<br />

2 CHCHD10-Hs01369775_g1 Core up 26.6 26.7 27.8 27.6 26.0 26.1 26.5 25.6 26.2 26.0 25.8 26.0 27.0 27.4 27.4 27.1 27.8 27.3 25.1 25.5 26.3 24.9 23.9 23.9<br />

3 RGS5-Hs00186212_m1 Core down 31.3 31.2 33.0 33.2 30.3 30.0 31.8 31.1 30.0 30.4 30.8 32.0 30.3 30.7 31.0 31.1 30.9 31.3 33.6 31.8 31.4 31.5 30.3 30.0<br />

4 ST6GALNAC5-<br />

Core down 30.0 30.7 26.4 26.5 33.2 30.4 33.6 36.6 35.6 34.7 30.6 31.0 33.9 34.3 28.1 27.6 29.5 29.3 33.5 34.7 31.9 33.7 33.7 33.1<br />

Hs00229612_m1<br />

5 CEBPB-Hs00270923_s1 Core up 30.9 30.0 30.9 31.1 28.5 27.9 30.4 29.9 29.5 29.8 30.3 30.2 30.3 30.3 30.0 29.8 30.5 30.6 29.6 30.4 29.4 28.8 27.3 28.0<br />

6 C5orf13-Hs00854282_g1 Core down 29.4 27.6 28.1 28.1 28.5 25.3 22.1 24.0 26.0 27.1 37.6 29.1 26.8 25.6 37.9 26.9 37.8 28.1 24.4 35.8 27.1 28.1 27.9 28.0<br />

7 PDGFRA-Hs00998026_m1 Marker 25.0 24.6 23.4 22.7 25.5 27.4 25.4 24.0 26.2 26.4 24.4 26.6 23.9 24.2 28.4 28.3 28.1 29.6 24.2 25.0 26.2 28.7 25.8 25.8<br />

8 CCND2-Hs00922419_g1 Core down 35.3 30.0 22.5 22.4 28.2 27.5 30.4 32.0 27.1 27.2 25.1 25.6 22.5 22.7 21.7 21.5 21.4 21.9 22.5 24.2 33.4 32.2 31.7 31.4<br />

9 NKX2-1-Hs00163037_m1 Core down 31.4 31.0 36.2 35.7 37.3 35.0 36.3 36.6 36.7 37.4 37.6 36.7 37.4 37.6 32.1 32.3 33.0 31.8 32.2 35.7 38.0 37.1 36.2 36.9<br />

10 CTSC-Hs00175188_m1 Core up 22.2 22.8 25.1 25.8 21.9 20.9 26.5 25.1 22.2 22.5 21.2 21.8 24.3 24.2 25.3 25.1 25.1 25.2 21.4 21.6 22.2 22.9 23.5 23.0<br />

11 18S-Hs99999901_s1 Norm 13.2 12.7 12.9 12.7 12.1 12.3 12.2 12.6 12.0 12.2 12.3 11.8 12.4 12.5 12.3 12.5 12.4 12.7 12.0 12.6 13.0 13.9 11.0 11.2<br />

12 DNER-Hs00294564_m1 Core down 31.7 27.9 28.2 26.7 33.0 32.5 28.2 26.8 26.3 27.1 25.9 27.6 24.5 24.6 24.9 25.1 25.8 26.8 28.7 27.4 28.0 30.4 27.1 26.8<br />

13 LMO4-Hs01086790_m1 Core up 24.8 24.8 22.8 22.6 23.7 23.7 24.9 23.8 24.1 24.2 23.9 25.1 23.2 23.2 23.3 23.2 23.7 23.7 23.2 23.5 24.7 24.2 23.6 23.1<br />

14 PLCH1-Hs00392783_m1 Core down 26.3 25.9 28.0 28.0 27.2 28.8 28.5 28.1 31.3 30.9 30.0 29.6 29.9 29.9 28.2 28.0 28.3 27.7 30.0 30.3 29.8 29.7 30.5 30.8<br />

15 SPARCL1-Hs00949886_m1 Core down 27.3 25.2 27.3 27.7 26.5 29.0 25.5 23.6 27.2 27.7 25.4 28.9 24.6 24.6 27.5 27.0 27.7 29.7 28.7 30.2 29.0 30.6 25.5 25.1<br />

16 SYNM-Hs00322391_m1 Core down 34.3 33.0 28.8 28.6 28.0 27.7 27.7 27.9 35.7 37.4 30.0 29.0 37.4 36.7 32.9 33.9 32.5 32.7 34.5 35.8 37.8 37.1 28.8 28.9<br />

17 FBLN2-Hs00157482_m1 Core down 37.3 35.0 31.9 32.2 30.4 30.8 37.2 35.5 32.5 34.3 34.9 35.8 34.5 35.2 35.2 35.1 34.7 36.2 31.7 34.2 37.5 34.8 27.9 28.5<br />

18 SALL2-Hs00413788_m1 Core down 30.2 27.7 27.5 26.6 34.6 37.1 35.5 32.8 31.7 31.4 30.1 32.7 26.6 26.8 27.5 27.1 27.6 27.9 27.7 27.5 38.0 37.1 33.1 33.7<br />

19 TUSC3-Hs00185147_m1 Core down 23.5 23.0 37.9 37.3 24.7 25.1 34.6 32.5 36.6 35.4 37.6 36.7 25.3 25.5 37.9 37.6 30.8 30.9 34.5 35.8 33.1 28.9 25.7 25.1<br />

20 GPR158-Hs00393109_m1 Core down 30.9 29.5 28.8 28.5 37.3 37.1 32.8 32.3 30.8 32.0 28.8 30.1 30.8 31.9 29.6 29.9 29.9 29.9 25.7 27.7 32.4 33.4 30.2 30.8<br />

21 PEG3-Hs00377844_m1 Core down 29.3 28.5 24.5 25.2 33.2 34.0 28.3 28.1 27.0 27.6 28.2 29.8 25.6 26.2 37.9 37.6 29.8 32.7 27.0 27.6 30.8 31.6 36.2 36.9<br />

22 FUT8-Hs00189535_m1 Core up 27.7 27.8 26.0 25.4 27.3 27.3 27.6 27.0 25.6 26.3 28.0 28.1 26.6 27.0 27.5 26.7 26.8 27.3 25.8 26.3 26.0 26.1 26.5 26.1<br />

23 HLA-DRA-Hs00219575_m1 Core up 27.3 25.7 34.3 35.4 35.6 37.1 28.4 27.4 22.1 23.3 24.4 24.2 24.3 24.5 34.6 32.6 28.8 31.2 21.9 21.6 29.3 26.7 21.4 21.0<br />

24 ASCL1-Hs00269932_m1 Marker 37.2 30.3 24.0 23.7 29.0 30.6 29.9 33.5 35.1 37.4 37.0 36.7 23.8 24.0 22.8 23.1 22.9 23.4 25.6 25.3 36.1 37.1 30.9 31.2<br />

25 PI15-Hs00210658_m1 Core down 29.2 30.4 33.2 29.3 30.9 32.3 29.3 26.2 27.2 27.9 28.4 26.5 31.5 31.3 36.9 37.6 28.5 35.4 30.1 32.9 28.4 30.1 27.5 26.6<br />

26 EPDR1-Hs00378148_m1 Core up 28.0 28.7 31.1 31.2 28.7 29.3 28.1 27.8 26.6 26.2 27.6 27.7 28.3 28.4 30.2 29.7 29.9 29.6 28.3 28.6 28.1 26.9 28.8 28.4<br />

27 DTX4-Hs00392288_m1 Core down 28.1 27.3 29.4 28.4 33.1 33.9 30.9 30.2 29.7 29.7 35.1 33.8 30.2 30.1 27.4 27.0 27.3 28.3 27.8 29.5 32.0 31.8 27.7 27.9<br />

28 CXXC4-Hs00228693_m1 Core down 30.0 27.8 25.2 25.1 29.4 31.0 28.7 29.5 36.6 37.4 35.5 36.7 27.3 27.4 26.3 26.7 26.2 27.7 28.2 28.1 31.4 32.9 36.1 34.9<br />

29 IRX2-Hs01383002_m1 Core down 37.3 36.6 37.9 37.3 28.6 28.8 36.9 36.6 36.7 37.4 37.6 36.7 37.4 37.6 37.9 37.6 37.8 35.6 34.5 35.8 38.0 37.1 29.8 30.2<br />

30 MAP6-Hs01023152_s1 Core down 28.2 27.9 26.9 27.0 28.6 29.7 31.4 29.3 30.1 31.2 33.1 32.3 28.8 28.8 30.1 30.0 29.4 30.5 31.2 30.6 31.7 33.8 26.7 26.4<br />

31 SIX3-Hs00193667_m1 Core down 33.4 33.6 37.9 37.3 30.2 30.6 35.0 34.5 35.7 36.3 37.6 36.7 35.4 37.1 35.6 36.7 37.8 35.6 29.1 29.7 34.7 33.2 36.2 36.9<br />

32 NKX2-2-Hs00159616_m1 Marker 29.1 28.0 27.4 28.1 27.8 28.0 26.8 26.4 25.2 25.2 26.4 27.0 25.3 25.3 25.9 25.9 25.5 25.2 24.6 24.8 29.4 28.9 30.3 30.9<br />

33 S100A6-Hs00170953_m1 Core up 18.8 19.7 30.1 27.6 18.6 17.9 20.4 19.6 20.0 19.6 22.0 20.9 21.4 21.5 23.9 23.5 23.5 22.5 21.4 22.6 18.5 18.0 18.5 18.1<br />

34 MMP17-Hs01108847_m1 Core up 28.7 29.1 31.2 30.5 28.9 27.8 29.3 28.5 24.8 25.3 30.7 31.0 26.5 26.6 30.3 29.4 30.0 29.9 26.7 25.2 27.6 26.6 27.8 28.3<br />

35 FAM38B-Hs00926225_m1 Core down 29.3 28.9 37.9 37.3 37.2 36.5 37.5 36.6 36.7 37.4 37.6 36.7 37.4 37.6 37.9 37.6 37.8 37.7 34.5 35.8 38.0 37.1 31.9 31.5<br />

36 OLIG2-Hs00300164_s1 Marker 35.2 28.7 23.0 23.2 27.1 27.6 25.8 25.8 22.8 23.3 23.7 24.6 22.1 21.8 23.5 23.4 23.4 23.4 22.3 22.4 32.1 33.3 29.7 30.4<br />

37 PMEPA1-Hs00375306_m1 Core up 24.4 24.7 28.8 28.0 25.8 26.5 22.8 22.6 25.1 24.7 27.4 27.0 25.6 25.3 28.0 27.5 28.1 27.7 24.8 26.5 23.6 25.1 25.3 25.6<br />

38 HOXD10-Hs00157974_m1 Core up 28.8 28.0 37.9 37.3 28.0 28.7 29.8 29.1 29.2 29.3 30.1 30.9 29.3 29.2 29.7 29.5 29.6 29.8 28.1 27.6 27.5 27.1 27.1 27.1<br />

39 NDN-Hs00267349_s1 Core down 24.1 23.7 30.3 30.5 37.3 37.1 26.4 25.4 25.6 26.1 27.8 26.8 37.3 37.6 26.4 25.7 26.3 25.7 26.1 25.8 36.8 37.1 36.2 35.0<br />

40 FOXG1-Hs01850784_s1 Core up 24.7 23.8 23.8 23.3 24.8 25.5 24.8 24.9 25.1 25.1 26.9 25.9 23.8 24.1 22.3 22.2 22.2 22.5 23.8 23.6 24.8 25.7 21.0 21.2<br />

41 MN1-Hs00159202_m1 Core down 32.5 34.5 28.9 28.3 32.1 33.6 26.9 26.9 30.2 29.3 34.3 35.1 28.4 27.7 27.4 27.8 27.6 27.1 29.1 28.9 35.7 35.1 30.7 30.6<br />

42 NDUFB10-Hs00605903_m1 Norm 25.3 25.2 25.4 25.6 25.6 25.4 25.6 25.3 25.7 25.7 25.3 25.6 25.7 25.5 26.2 26.1 26.2 25.9 26.0 25.4 25.5 25.1 25.7 25.2<br />

43 HMGA2-Hs00171569_m1 Core down 30.2 31.6 33.9 33.5 27.7 27.3 35.8 35.5 32.1 32.2 32.7 33.3 32.8 32.1 34.0 33.4 33.3 33.1 33.3 32.6 32.3 32.2 28.6 28.5<br />

44 MMRN1-Hs00201182_m1 Core down 34.0 31.1 37.9 37.3 33.3 34.7 37.5 36.6 36.7 37.4 32.6 31.2 37.4 37.6 37.9 37.6 37.8 37.7 34.5 35.8 34.8 35.1 32.8 32.1<br />

45 RAB6B-Hs00981572_m1 Core down 29.2 30.3 30.7 29.6 33.5 33.1 28.4 27.6 29.2 29.4 31.4 32.1 28.3 28.1 27.6 27.3 27.4 27.8 29.1 29.0 30.7 30.7 30.2 29.7<br />

46 NTRK2-Hs00178811_m1 Core up 24.3 24.1 30.4 30.0 31.9 32.7 33.6 32.7 23.1 23.8 23.9 26.0 24.7 24.6 35.6 37.5 28.2 32.5 23.8 23.9 23.5 24.9 27.3 27.3<br />

47 KALRN-Hs00610179_m1 Core down 30.8 30.2 28.4 27.7 30.6 31.9 29.0 27.9 30.4 30.5 31.2 30.8 27.6 28.1 28.3 28.1 27.4 28.2 29.6 29.6 28.9 31.1 30.5 30.8<br />

48 CD74-Hs00269961_m1 Core up 27.9 25.9 33.0 35.4 33.0 34.3 28.4 27.8 21.9 23.3 24.8 25.2 25.1 25.0 37.5 34.1 29.0 31.0 22.4 23.5 29.7 26.7 21.5 21.8<br />

49 LUM-Hs00158940_m1 Core down 30.4 31.4 37.9 37.3 23.6 26.1 33.3 27.1 36.7 37.4 32.0 30.9 37.4 37.6 33.3 31.8 35.7 37.7 34.5 35.8 29.8 27.6 29.6 29.0<br />

50 NELL2-Hs00196254_m1 Core down 33.4 32.8 34.1 29.3 31.8 32.2 30.3 29.2 36.6 37.4 33.1 34.1 31.2 32.0 27.9 28.0 29.0 29.8 27.5 26.8 30.1 32.0 31.4 31.0<br />

263


A.3 Quantitative RT-PCR Appendix<br />

Well Applied Biosystems assay ID Gene G19 G19 G21 G21 G23 G23 G24 G24 G25 G25 G26 G26 G30 G30 G31 G31 G32 G32 G144 G144 G166 G166 G179 G179<br />

Category A B A B A B A B A B A B A B A B A B A B A B A B<br />

51 MAP6-Hs01929835_s1 Core down 26.0 26.1 25.9 25.7 29.1 30.2 30.2 28.3 29.2 30.7 32.0 31.6 27.3 27.6 30.6 30.4 29.1 30.7 32.2 29.8 31.9 34.0 26.6 26.3<br />

52 PDE1C-Hs01095694_m1 Core up 24.8 25.0 28.2 28.9 25.2 24.2 24.1 23.3 24.1 24.0 26.1 25.5 26.9 26.5 28.9 28.5 28.7 28.5 25.0 25.2 23.9 23.6 25.5 25.5<br />

53 SULF2-Hs00378697_m1 Core up 23.9 24.3 27.0 26.1 24.3 26.4 23.1 22.0 24.3 24.1 24.3 25.4 23.5 23.2 25.9 25.0 25.7 25.8 25.0 23.5 26.2 25.5 26.0 26.0<br />

54 TUBB-Hs00962420_g1 Norm 20.7 21.3 20.9 20.9 21.5 21.4 21.4 21.3 21.5 21.2 21.5 21.8 21.1 21.2 20.7 20.6 20.6 20.5 21.2 21.2 20.7 20.1 22.5 22.7<br />

55 LMO2-Hs00277106_m1 Core down 30.6 30.1 27.2 26.2 31.3 35.6 31.3 29.9 34.0 34.4 27.9 30.2 26.3 27.4 26.9 26.5 26.6 26.1 30.0 31.4 34.3 35.6 29.0 28.9<br />

56 MAN1C1-Hs00220595_m1 Core up 27.9 26.9 25.8 25.2 32.1 34.2 29.7 28.8 26.9 26.7 24.8 25.9 25.6 25.9 25.8 24.9 26.0 25.5 26.3 26.3 28.1 27.4 26.2 26.6<br />

57 TAGLN-Hs00162558_m1 Core down 28.4 29.7 37.9 33.3 24.9 27.3 21.4 21.1 26.2 25.4 29.7 29.0 26.1 25.0 33.7 31.0 29.3 29.4 27.4 30.2 31.4 29.8 26.9 26.7<br />

58 RTN1-Hs00382515_m1 Core down 25.1 24.8 33.6 32.9 27.8 28.8 29.8 28.7 32.2 32.3 25.5 26.2 25.8 25.8 28.9 27.7 29.8 30.3 27.0 28.1 29.1 30.8 30.8 30.6<br />

59 MAF-Hs00193519_m1 Core down 28.2 27.5 28.7 28.9 27.5 30.2 28.5 28.3 31.1 31.9 27.3 28.4 27.8 28.0 27.8 28.0 28.4 27.5 31.4 32.1 28.1 28.7 34.7 34.5<br />

60 NPTX2-Hs00383983_m1 Core down 30.1 27.8 29.0 32.2 28.9 29.3 34.5 36.6 36.7 35.0 27.8 26.5 32.3 31.8 31.2 29.3 34.2 30.2 31.7 32.1 28.4 29.3 31.8 32.3<br />

61 EDA2R-Hs00939736_m1 Core down 27.5 27.4 28.8 28.6 29.2 26.9 28.4 27.8 35.2 35.2 32.7 32.2 28.8 28.8 35.4 35.0 30.6 32.2 34.4 32.8 32.0 30.2 34.6 34.3<br />

62 SOX10-Hs00366918_m1 Marker 37.3 36.6 37.9 37.3 37.3 37.1 37.0 36.6 24.4 25.2 37.6 36.7 35.4 35.6 37.9 37.6 37.8 37.7 24.5 24.5 38.0 37.1 36.2 36.9<br />

63 SEMA6A-Hs00221174_m1 Core down 26.9 25.9 23.3 23.6 25.9 26.2 24.1 24.5 27.4 27.6 25.2 26.4 24.3 24.4 23.4 23.4 23.6 23.7 24.7 25.0 26.3 27.2 27.2 27.6<br />

64 DDIT3-Hs00358796_g1 Core up 27.0 26.8 28.0 28.1 25.2 22.4 27.6 26.8 26.8 26.3 28.3 28.7 27.3 27.2 22.0 21.7 21.8 21.8 22.5 23.2 25.3 23.8 26.4 25.9<br />

65 CA12-Hs01080909_m1 Core down 30.4 28.1 23.3 23.6 25.8 26.6 31.8 30.0 27.6 27.3 24.1 25.9 27.1 28.4 24.8 24.4 24.8 23.9 25.8 25.1 26.9 26.9 25.6 25.9<br />

66 C9orf125-Hs00260558_m1 Core down 31.0 31.1 37.9 37.3 34.8 37.1 29.1 28.5 31.8 32.3 30.4 31.3 27.3 27.9 37.9 37.6 37.3 37.3 29.4 29.2 32.3 34.1 35.1 34.8<br />

67 SDC2-Hs00299807_m1 Core down 26.3 26.5 36.0 36.3 26.2 26.4 25.6 25.2 25.6 25.3 26.4 26.3 29.4 29.2 29.0 28.2 29.6 29.6 26.7 27.8 27.9 28.6 25.4 25.3<br />

68 SLIT2-Hs00191193_m1 Core down 25.4 23.8 34.2 32.3 26.2 26.3 32.8 30.6 33.5 32.7 32.3 31.8 33.7 34.8 29.8 29.6 27.6 27.9 30.4 32.1 33.1 33.5 30.7 30.6<br />

69 MT2A-Hs02379661_g1 Core up 22.9 22.6 22.6 21.8 21.2 22.0 23.3 22.7 23.7 22.9 20.6 21.5 22.2 22.7 22.3 22.0 22.1 22.4 21.8 21.7 19.8 18.6 19.2 18.8<br />

70 IL17RD-Hs00296982_m1 Core down 26.6 26.5 26.4 26.9 24.0 24.5 26.8 26.6 27.7 27.4 27.2 28.2 26.5 26.6 27.1 27.0 26.9 27.4 26.4 26.6 29.0 28.2 27.2 27.5<br />

71 PDZRN3-Hs00392900_m1 Core down 29.3 27.9 27.3 27.0 34.2 37.1 36.2 36.6 26.4 26.4 28.3 28.3 29.0 29.5 27.3 26.9 26.9 26.7 30.5 31.7 31.8 31.8 28.2 28.9<br />

72 LGALS3-Hs00173587_m1 Core up 25.1 24.3 25.2 25.0 23.4 23.4 27.1 25.2 24.4 25.0 23.1 23.8 24.7 24.9 26.3 25.4 25.1 25.3 25.2 24.8 25.1 23.2 23.8 23.2<br />

73 FAM69A-Hs00961685_m1 Core up 25.3 24.4 24.6 25.6 25.1 27.8 25.7 23.6 25.4 25.9 24.9 25.6 26.8 26.9 26.1 26.0 25.7 25.8 25.6 25.3 27.1 26.1 26.6 25.9<br />

74 TERT-Hs00972656_m1 Marker 31.8 32.5 29.0 27.2 30.7 30.3 31.9 31.3 31.0 30.8 31.2 31.7 30.2 29.9 30.7 30.2 30.3 29.9 29.4 28.2 30.2 29.9 33.9 34.7<br />

75 PLS3-Hs00418605_g1 Core up 21.9 22.3 25.3 25.8 23.1 23.1 22.1 22.4 23.9 23.7 23.1 23.6 25.3 25.1 25.3 25.4 25.6 25.0 24.0 23.4 22.1 22.5 25.5 24.9<br />

76 PTEN-Hs02621230_s1 Core down 29.2 29.2 33.8 30.6 28.1 28.7 29.1 28.5 29.6 30.3 31.0 31.1 28.5 28.7 31.3 31.5 31.1 31.4 34.5 35.8 30.1 31.2 36.2 34.8<br />

77 SALL2-Hs00826674_m1 Core down 34.2 32.2 32.6 33.4 33.8 37.1 33.9 32.1 32.8 33.1 33.5 34.2 32.8 32.4 32.0 32.1 31.8 31.8 32.8 33.4 33.4 31.9 30.6 30.7<br />

78 KCTD12-Hs00540818_s1 Core down 32.2 31.6 30.4 32.3 30.4 31.9 26.3 25.9 27.6 28.5 28.9 29.0 28.0 28.2 30.9 31.7 30.2 31.5 28.3 29.8 30.4 30.1 25.9 26.5<br />

79 TES-Hs00210319_m1 Core down 37.3 36.6 37.9 37.3 34.5 36.6 34.5 36.4 33.8 34.6 37.6 36.7 37.4 37.6 37.9 37.6 37.8 37.7 34.5 35.8 37.2 36.2 35.0 33.3<br />

80 SOX2-Hs01053049_s1 Marker 24.0 23.5 22.9 23.0 24.8 24.6 24.6 24.4 23.2 23.9 23.4 24.5 23.0 23.2 23.2 23.0 22.8 23.3 23.4 23.9 25.5 27.0 21.9 21.8<br />

81 LPAR6-Hs00271758_s1 Core down 29.9 29.5 29.7 29.7 30.0 31.4 29.2 28.7 33.0 33.2 31.1 31.9 28.6 28.6 29.9 29.3 29.8 29.4 31.6 31.7 29.8 30.3 31.3 31.2<br />

82 ODZ2-Hs00393060_m1 Core down 25.9 26.0 27.6 28.9 25.0 26.1 28.6 26.5 31.6 31.4 25.5 26.5 31.0 31.0 28.0 26.9 28.7 27.9 27.1 28.2 31.0 32.3 27.2 27.4<br />

83 NNMT-Hs00196287_m1 Core up 28.3 27.9 30.9 30.9 24.1 27.2 33.2 29.9 27.9 27.3 24.3 24.2 27.6 27.8 29.9 28.7 30.6 29.0 25.0 26.8 24.0 24.1 23.5 23.4<br />

84 CACNG8-Hs01100182_m1 Core down 32.0 30.5 28.1 28.9 30.4 29.9 31.0 36.6 33.3 33.3 34.3 34.0 27.3 27.6 31.8 33.2 30.4 30.7 27.7 27.7 31.4 31.9 32.0 33.3<br />

85 PRSS12-Hs00186221_m1 Core up 24.5 25.3 31.2 33.8 25.1 24.6 31.5 31.5 27.4 27.0 25.9 26.6 37.4 37.6 28.4 28.3 27.7 27.5 24.8 24.6 24.3 24.4 30.7 31.1<br />

86 FOXJ1-Hs00230964_m1 Core down 32.8 30.1 31.8 31.2 33.8 36.5 31.1 30.5 29.9 31.2 30.3 30.9 28.2 28.3 31.4 31.3 30.6 31.2 30.3 30.9 36.2 37.1 31.1 31.8<br />

87 NTN1-Hs00180355_m1 Core down 29.5 28.5 27.5 26.6 34.5 35.7 25.9 24.6 27.7 28.4 28.7 30.2 27.4 27.4 26.4 26.5 26.5 26.3 25.5 26.8 31.5 35.1 28.6 29.8<br />

88 LMO3-Hs00375237_m1 Core down 37.3 36.6 33.0 35.2 31.9 33.7 37.5 36.6 36.7 37.4 28.7 28.6 32.4 31.2 28.0 28.5 28.2 29.3 34.5 35.8 38.0 37.1 35.1 34.2<br />

89 CHI3L1-Hs00609691_m1 Marker 31.6 25.5 30.7 28.1 33.4 37.1 24.8 23.7 29.0 31.1 24.4 27.1 22.1 23.4 28.5 28.2 29.0 29.9 34.5 35.8 24.0 28.4 21.5 21.7<br />

90 CD9-Hs01124025_g1 Core up 23.4 23.6 23.3 23.9 24.7 24.4 26.0 24.6 22.1 22.9 23.5 24.2 22.8 23.0 23.5 23.5 23.4 23.8 22.6 22.7 23.4 23.2 23.4 22.9<br />

91 ADD2-Hs00242289_m1 Core up 26.8 27.3 24.9 25.5 26.8 25.3 27.2 27.4 28.3 28.3 30.9 31.7 26.2 26.5 25.8 25.5 25.6 25.6 26.4 26.5 26.6 25.5 28.2 28.7<br />

92 MYL9-Hs00697086_m1 Core down 33.0 32.5 32.3 31.6 27.6 29.8 23.3 22.9 28.3 28.1 26.8 27.6 28.7 27.9 34.7 32.8 31.1 31.5 30.5 31.6 26.7 26.2 26.6 26.8<br />

93 LYST-Hs00179814_m1 Core up 30.1 29.8 30.7 31.0 29.9 28.5 29.7 28.6 28.6 29.6 29.9 31.3 28.7 29.3 29.8 29.8 29.9 30.6 27.6 28.2 29.1 28.1 28.1 27.7<br />

94 LAMA2-Hs00166308_m1 Core down 30.2 28.9 37.8 37.3 28.8 29.9 37.5 35.6 31.2 33.4 32.8 32.2 31.0 31.8 30.5 29.4 36.0 35.4 26.8 27.0 33.7 34.1 33.2 34.9<br />

95 PLA2G4A-Hs00233352_m1 Core up 28.4 28.7 30.2 29.3 29.6 29.5 30.6 29.4 26.8 27.4 31.8 31.8 26.9 26.7 27.4 26.8 27.1 26.8 26.5 27.2 29.6 27.5 29.0 28.7<br />

96 BACE2-Hs00273238_m1 Core up 25.1 25.2 35.6 37.3 29.7 30.5 26.9 26.1 25.7 25.9 27.2 28.9 27.8 28.3 30.8 30.4 30.3 30.8 26.3 26.3 26.8 25.7 27.0 26.9<br />

264


A.4 Tag-seq vs qRT-PCR Correlation Appendix<br />

A.4 Tag-seq vs qRT-PCR Correlation<br />

The correlation between the qRT-PCR measurements from the 21 cell lines<br />

(16 GNS and 5 NS cell lines) and the Tag-seq measurements from the five cell<br />

lines (three GNS and two NS cell lines) was found by taking the normalised<br />

Ct values and tag counts for each <strong>of</strong> the 82 core differentially expressed genes<br />

- as determined by Tag-seq on the three GNS cell lines and two NS cell lines -<br />

and applying the cor function from the stats R package.<br />

Table A.5: Pearson correlation values between the normalised Ct values measured<br />

through qRT-PCR and the tag counts measured across the five GNS and NS cell<br />

lines assayed via Tag-seq.<br />

Gene name Pearson Gene name Pearson<br />

correlation correlation<br />

PTEN 0.9973 MAN1C1 0.9953<br />

NDN 0.9942 HMGA2 0.9932<br />

HOXD10 0.9925 TUSC3 0.9888<br />

TES 0.9848 SYNM 0.9823<br />

SIX3 0.9800 LYST 0.9797<br />

MYL9 0.9777 PLA2G4A 0.9757<br />

IRX2 0.9749 DDIT3 0.9748<br />

MT2A 0.9741 MMP17 0.9735<br />

BACE2 0.9684 MAF 0.9672<br />

CA12 0.9653 LMO3 0.9652<br />

NKX2-1 0.9625 IL17RD 0.9571<br />

SEMA6A 0.9560 CXXC4 0.9520<br />

FAM69A 0.9475 ADD2 0.9451<br />

SPARCL1 0.9416 LMO2 0.9391<br />

CHCHD10 0.9388 PLCH1 0.9362<br />

PEG3 0.9345 CCND2 0.9344<br />

KALRN 0.9310 FAM38B 0.9278<br />

EDA2R 0.9270 GPR158 0.9222<br />

LMO4 0.9203 CD9 0.9187<br />

ODZ2 0.9168 ST6GALNAC5 0.9139<br />

MAP6 0.9116 NPTX2 0.9047<br />

CACNG8 0.9045 SLIT2 0.9016<br />

MMRN1 0.8906 CD74 0.8906<br />

TAGLN 0.8874 PDE1C 0.8832<br />

CEBPB 0.8813 DNER 0.8811<br />

FUT8 0.8804 C9orf125 0.8772<br />

RAB6B 0.8709 PI15 0.8654<br />

NTN1 0.8620 SULF2 0.8602<br />

KCTD12 0.8384 PMEPA1 0.8322<br />

LUM 0.8263 SALL2 0.8078<br />

NNMT 0.8034 HLA-DRA 0.7884<br />

RTN1 0.7777 MN1 0.7714<br />

RGS5 0.7679 CTSC 0.7661<br />

C5orf13 0.7595 PLS3 0.7439<br />

NELL2 0.7146 NTRK2 0.7115<br />

LPAR6 0.7090 EPDR1 0.7053<br />

LAMA2 0.6889 SDC2 0.6595<br />

S100A6 0.6580 FOXG1 0.6051<br />

PDZRN3 0.6032 FBLN2 0.5795<br />

PRSS12 0.5211 LGALS3 0.5204<br />

DTX4 0.4964 FOXJ1 0.4454<br />

265


Appendix B<br />

Literature Mining Script<br />

It should be noted that the text preceded by the single quotation mark sign<br />

(’) that appears in italic typeface is a comment and not code that should be<br />

executed.<br />

Public Class Form1 ’GBMbase iHOP PubMed BioGraph Google Scholar Google Search<br />

Dim genes() As String = {"SFTA3", "SLC4A4", "STRA6", "IL6", "SNX22", "COL21A1",<br />

"CA12", "CCND2", "SPINT1", "INHBA", "GFAP", "GPC3", "HOXC10", "PDE1C",<br />

"FAM70A", "GEM", "KALRN", "NTN1", "INHBB", "CCNO", "C10orf81", "CTNNA2",<br />

"APLN", "VAX1", "PCDHB4", "KCTD12", "ELN", "IFI30", "BMP8B", "CAMK2B",<br />

"LMO2", "TECRL", "RGS5", "NOV", "MOCOS", "SCN1B", "TNFSF4", "C14orf143",<br />

"LRRC2", "PLS3", "AP000280.1", "C21orf62", "C10orf116", "VIPR1", "ELMO1", "UGT8",<br />

"SH3BGR", "LIF", "MX1", "LRAT", "FXYD3", "CNTN6", "NNMT", "ZIC5", "GBP2",<br />

"TRIM47", "FAM196A", "C10orf141", "LMO4", "MYC", "RTP4", "C9orf125", "DPY19L1",<br />

"FCGR2B", "FCGR2C", "FCGR2A", "MAPT", "SLC15A2", "TGM2", "IRX2", "PCDH20",<br />

"ANGPTL1", "IL17RD", "ARC", "CHCHD10", "MAF", "CD55", "FBXO27", "PDZRN3",<br />

"ETS1", "SGCD", "CITED4", "MAP3K5", "STXBP5L", "ECHDC2", "CTSC", "MICA",<br />

"MMP17", "AC068399.1", "VIT", "TSHZ3", "CXXC4", "CABP7", "KCNMB2", "MT2A",<br />

"MYH3", "HOXD13", "STEAP2", "SLC4A11", "CCL7", "IFI27", "SPARCL1", "TNNI1",<br />

"OAS2", "TRPM8", "THBS2", "DOCK10", "ZIC2", "IFI6", "TNNI2", "CARD17", "CASP1",<br />

"CARD16", "ATOH8", "DNER", "FAM189A1", "SALL2", "C10orf11", "EEF1D", "WB-<br />

SCR17", "TUBA4A", "RHBDF2", "PARP3", "MAN1C1", "DDO", "F12", "SPTBN5",<br />

"TGFA", "ZNF536", "HLA-DQB1", "IFITM2", "DNM3", "JPH1", "FAM69A", "CACNA1C",<br />

"ESRRG", "ITM2A", "SKAP2", "TRAM1L1", "NDN", "PLCH1", "KCNJ12", "NXPH1",<br />

"MYBL1", "DUSP5", "NTRK2", "RNF175", "HOMER1", "PTEN", "CMPK2", "TBC1D8",<br />

"TRIB3", "CDHR1", "SYNM", "LUM", "SEMA6A", "ADRA2A", "STAMBPL1", "TSPAN7",<br />

"PYGL", "FOXJ1", "ZNF454", "EML2", "GABRQ", "EPAS1", "ERBB4", "RAB6B",<br />

"LXN", "MYL9", "BGN", "FUT8", "GRIA3", "ARHGEF7", "SLC2A5", "MMP7", "TMEM132D",<br />

"PION", "C5orf13", "SOD2", "PDGFA", "LOXL4", "SPP1", "BATF3", "SNAP25", "SULF2",<br />

"LPAR6", "EPDR1", "EPHB3", "TSLP", "TMCO4", "SERPINE2", "TUSC3", "VSNL1",<br />

"ATAD3C", "MARCH1", "DKK1", "CEBPB", "NFE2L3", "TNNC2", "ODZ2", "OAS1",<br />

266


Appendix<br />

"BEX5", "DIAPH2", "GBP3", "SORCS3", "SOX3", "FOXG1", "LRRN2", "ELMO2",<br />

"MAP6", "TRIM48", "CNKSR2", "MOSC2", "CRYBB2", "GJA1", "ELOVL2", "B4GALNT1",<br />

"C10orf90", "FBN2", "SMOC2", "ZIC3", "RARRES3", "GBP1", "EFEMP1", "GPR98",<br />

"PLD3", "OLFM1", "CCDC64", "MGLL", "THY1", "CPLX2", "FAM150B", "MKI67",<br />

"AC092296.1", "PHACTR3", "TNFRSF14", "NRG1", "RAB38", "ABAT", "NRBF2", "CLDN10",<br />

"C1orf94", "NELL2", "CYB5R2", "TNC", "HLA-A", "PPCS", "SORL1", "SHROOM3",<br />

"SLC38A1", "FXYD5", "CILP", "OGN", "MARVELD3", "APOD", "LEMD1", "DOCK5",<br />

"GBP4", "KCNA2", "FAM55C", "NFKBIZ", "RGL3", "HLA-DPB1", "XXbac-BPG116M5.1",<br />

"C2", "CFB", "NRP2", "HPSE", "CPNE5", "SMAGP", "FAM84A", "LOC653602", "NR4A2",<br />

"PERP", "ZNF281", "NID1", "ACIN1", "FOXQ1", "MATN2", "IRAK1", "NDE1", "COL8A1",<br />

"TFAP2A", "C1S", "ST6GAL1", "CD97", "ID4", "IGSF3", "NAMPT", "RP11-473I1.1",<br />

"EPB41L3", "NOTCH3", "GALNT5", "NLGN4X", "PITPNC1", "KLF6", "B3GNT9",<br />

"RIPK4", "S100B", "GYG2", "LOC283070", "CAMK1D", "ZNF747", "HOXA7", "NMU",<br />

"RAB7L1", "STX3", "SRGAP3", "CNTN1", "ATP1B2", "MAP7", "ADAMTS4", "F2RL1",<br />

"MIA", "MLC1", "ARHGAP20", "C4orf32", "EDA2R", "OTX2", "MEIS2", "SPRED1",<br />

"PPEF1", "TCF7", "TFCP2", "HPRT1", "TTF2", "SLITRK5", "PXDN", "MIPOL1",<br />

"SYT1", "CXCL14", "ETS2", "PPHLN1", "NBL1", "GDPD2", "SDC2", "ADAMTS10",<br />

"C6orf138", "ICAM1", "SELENBP1", "C7orf40", "GALR1", "SFRP1", "IGSF11", "MTTP",<br />

"RRS1", "RGAG4", "BTBD11", "PHLPP1", "RGS17", "NCAM1", "MID1IP1", "TMEM200B",<br />

"CRYBB1", "KANK1", "MT1A", "SHOX2", "PLS1", "H1F0", "SEMA3D", "ITGA4",<br />

"ZIC4", "SPINK2", "SLC38A5", "LINGO1", "FAM184B", "NR1D1", "SQRDL", "SPAG4",<br />

"HOXD9", "HAND2", "LIFR", "MYO1B", "TPMT", "PCDHB3", "SHISA2", "XYLT1",<br />

"REC8", "NFATC2", "TOX", "STC2", "FAM126A", "GAL", "CPAMD8", "HLA-DQA1",<br />

"SRPX", "GJB2", "HOXB6", "HIF3A", "MXRA5", "ITGBL1", "LGALS3", "C5orf38",<br />

"IL1RAPL1", "TSPAN13", "AC007405.8", "LOC285141", "TMSB15A", "ADAMTS1", "KCNK12",<br />

"PARP12", "DCHS1", "TSTD1", "SEZ6L", "SNHG12", "IL4I1", "CCDC48", "NKX6-2",<br />

"OAS3", "TUBB4", "CDH19", "GJC3", "TMEM100", "DDIT4L", "ARHGAP8", "PRR5",<br />

"PRR5-ARHGAP8", "DDAH2", "ADCYAP1R1", "C3orf58", "IL33", "IL1R1", "CD68",<br />

"MARS", "ALDH1A3", "C2orf80", "C7orf16", "KIAA1217", "AC067930.1", "CD248", "CMTM5",<br />

"HRCT1", "CCR1", "FAM129A", "PIGA", "PAX8", "GRB14", "ADD2", "CTLA4", "DYNC1I1",<br />

"KCNK1", "LRRC55", "PDGFRB", "SYTL5", "TMEM158", "PROCR", "PSPH", "CRIP2",<br />

"CBLC", "TMEM38A", "INSIG1", "ST6GALNAC3", "NOP16", "TPM1", "FAM176A",<br />

"PPM1K", "PNMA2", "OXTR", "TRAF1", "TRIB2", "TRIM14", "AQP4", "PEA15",<br />

"PRSS12", "LPHN2", "CD58", "NFASC", "CHRDL1", "AC026410.6", "IKBKE", "CRB2",<br />

"SRP9", "IFITM8P", "SLC16A3", "ANGPTL2", "COL4A6", "LFNG", "FZD3", "CDH6",<br />

"PPP4R1", "LAMA4", "LNX1", "EFNA5", "TMEM71", "RPSAP52", "SYNGR1", "GABRA5",<br />

"TESC", "TTYH1", "FAM181A", "HOXC13", "C1orf133", "LRP4", "FERMT3", "CDKN2C",<br />

"TSPAN9", "CXorf38", "HOXA1", "TTN", "TMEM176B", "CPNE2", "SALL1", "SLC26A2",<br />

"ZEB1", "GLYATL2", "RHOBTB3", "EFHD2", "MLPH", "MFAP2", "PTPRR", "RRP7A",<br />

"SNX10", "ZNF714", "MACROD2", "GMPR", "BMPER", "AIDA", "FAM5B", "PLCB1",<br />

"ADRA1B", "ELMOD1", "RP11-93B14.2", "hCG_2018279", "SYTL4", "NFIA", "CMAH",<br />

"PLAC9", "EVC2", "SCN11A", "RASGRP3", "RHPN1", "PTPRD", "PDE4B", "CXCL12",<br />

"CCKBR", "ISYNA1", "SDK2", "TNFAIP3", "ACTA2", "PCDHB12", "AMMECR1",<br />

"PKNOX2", "PTPRH", "DUSP16", "SULF1", "RP9", "GNG11", "C5orf41", "C9orf95",<br />

"DCBLD2", "RAD50", "RASGEF1C", "PCOLCE2", "PNP", "PLEKHA6", "HEPACAM",<br />

267


Appendix<br />

"TAPBPL", "MEST", "EEF1A2", "C1orf187", "CALM1", "CREB3L1", "CCNY", "MGC87042",<br />

"TCF7L2", "ZNF710", "JAM2", "SCN5A", "SLC46A1", "TMSL1", "C20orf103", "HOXA5",<br />

"FMNL1", "CACNG7", "TF", "RNASEH1", "UNC80", "EGFR", "PTGS1", "ASPN",<br />

"SEMA4G", "ASNS", "FGFR1", "CHODL", "IRX1", "INPP5D", "PLSCR1", "NCALD",<br />

"CLDN3", "COL1A2", "GGH", "MICB", "FNDC5", "RAB11FIP1", "DKFZp434H1419",<br />

"AC012513.4", "CGREF1", "RP5-955M13.1", "KCNG1", "GFPT2", "IGFBP5", "CCDC8",<br />

"RADIL", "BID", "PPL", "RGS20", "BMP7"}<br />

Dim dbUrls() As String = {"www.gbmbase.org", "www.ncbi.nlm.nih.gov/pubmed", "www.ihop-<br />

net.org/UniPub/iHOP/", "biograph.be/", "scholar.google.com/scholar", "www.google.com"}<br />

Dim pageLoadTimes, nDB, ngene As Integer<br />

Public Event nextGene()<br />

Public Event nextDB()<br />

Private Sub ButtonStart_Click(sender As Object, e As EventArgs) Handles ButtonStart.Click<br />

ngene = -1<br />

RaiseEvent nextGene()<br />

End Sub<br />

Private Sub nextGene_() Handles Me.nextGene<br />

ngene += 1<br />

If ngene < genes.Count Then<br />

nDB = 4 ’0 ’-1+<br />

TextBox3.Text = genes(ngene)<br />

RaiseEvent nextDB()<br />

Else<br />

’ End data collection<br />

End If<br />

End Sub<br />

Private Sub nextDB_() Handles Me.nextDB<br />

nDB += 1<br />

If nDB < dbUrls.Count Then<br />

pageLoadTimes = 0<br />

Select Case nDB<br />

Case 0, 3 : WebBrowser1.Navigate(dbUrls(nDB))<br />

Case 1 : WebBrowser1.Navigate(dbUrls(nDB) & "?term=" & genes(ngene) & " glioblas-<br />

toma")<br />

Case 2 : WebBrowser1.Navigate(dbUrls(nDB) & "?search=" & genes(ngene) & " &field=all<br />

&ncbi_tax_id=9606&organism_syn=")<br />

Case 4 : WebBrowser1.Navigate(dbUrls(nDB) & "?as_vis=0&q=" & genes(ngene) & "<br />

+glioblastoma&hl=en&as_sdt=0,5")<br />

Case 5 : WebBrowser1.Navigate(dbUrls(nDB) & "#hl=en&gs_nf=1&cp=10&gs_id=r&xhr=t<br />

&q=" & genes(ngene) & "+NEAR+glioblastoma&fp=1")<br />

268


End Select<br />

End If<br />

End Sub<br />

Appendix<br />

Private Sub WebBrowser1_DocumentCompleted(sender As Object, e As WebBrowserDoc-<br />

umentCompletedEventArgs) Handles WebBrowser1.DocumentCompleted<br />

If nDB = 0 Then ’ GMBbase<br />

Select Case pageLoadTimes<br />

Case 0 ’ Search and fill the "search textbox"<br />

For Each elem As HtmlElement In WebBrowser1.Document.All<br />

Dim NameStr As String = elem.GetAttribute("name")<br />

If ((NameStr IsNot Nothing) AndAlso (NameStr.Length 0)) Then<br />

If NameStr.ToLower().Equals("search") Then<br />

elem.InnerText = genes(ngene)<br />

End If<br />

End If<br />

Next<br />

WebBrowser1.Document.GetElementById("title_submit").InvokeMember("click")<br />

Case 1 ’ Search the link to Gene name<br />

For Each elem As HtmlElement In WebBrowser1.Document.Links<br />

Dim NameStr As String = elem.InnerText<br />

If ((NameStr IsNot Nothing) AndAlso (NameStr.Length 0)) Then<br />

If Trim(elem.InnerText) = genes(ngene) Then<br />

elem.InvokeMember("Click")<br />

Exit Select<br />

End If<br />

End If<br />

Next<br />

Case 2 ’ Get the results from page<br />

Dim st As String = WebBrowser1.DocumentText.ToString<br />

Dim st1 As Integer = st.IndexOf("Glioblastoma multiforme Gene Publications:")<br />

If st1 > 0 Then<br />

TextBox3.Text = Convert.ToInt32(Trim(st.Substring(st1 + 49, 5)))<br />

If TextBox3.Text "0" Then RaiseEvent nextGene() Else RaiseEvent nextDB()<br />

Exit Sub<br />

End If<br />

End Select<br />

ElseIf nDB = 1 Then ’ PubMed<br />

269


Select Case pageLoadTimes<br />

Case 0 ’ Get the results from page<br />

Dim st As String = WebBrowser1.DocumentText.ToString<br />

Dim stp As Integer = st.IndexOf("class=""result_count"">Results: ")<br />

If stp > 0 Then<br />

Appendix<br />

Dim stR As String = st.Substring(stp + 28, 20) ’ ex. : 1 to 20 <strong>of</strong> 87


If NameStr.ToLower().Equals("query") Then<br />

elem.InnerText = genes(ngene) : Exit For<br />

End If<br />

End If<br />

Next<br />

Appendix<br />

For Each elem As HtmlElement In WebBrowser1.Document.All ’ Search and click submit<br />

button<br />

Dim NameStr As String = elem.GetAttribute("name")<br />

If ((NameStr IsNot Nothing) AndAlso (NameStr.Length 0)) Then<br />

If NameStr.ToLower().Equals("commit") Then<br />

elem.InvokeMember("click") : Exit For<br />

End If<br />

End If<br />

Next<br />

Case 1 ’ Get the result page<br />

Dim st As String = WebBrowser1.DocumentText.ToString<br />

End Select<br />

ElseIf nDB = 4 Then ’ Google Scholar<br />

Select Case pageLoadTimes<br />

Case 0 ’ Get the results from page<br />

Dim st As String = WebBrowser1.DocumentText.ToString<br />

Dim stp As Integer = st.IndexOf("About ")<br />

If stp > 0 Then<br />

Dim stR As String = st.Substring(stp + 24, 20) ’ ex. 18,700 results (0.03 sec)...<br />

stp = stR.IndexOf("results")<br />

st = stR.Substring(0, stp).Replace(",", "") ’ ex. 18,700 –>18700<br />

TextBox3.Text = Convert.ToInt32(Trim(st))<br />

If TextBox3.Text "0" Then RaiseEvent nextGene() Else RaiseEvent nextDB()<br />

Exit Sub<br />

End If<br />

End Select<br />

ElseIf nDB = 5 Then ’ Google Search<br />

Select Case pageLoadTimes<br />

Case 0 ’ Get the results from page<br />

Dim st As String = WebBrowser1.DocumentText.ToString<br />

Dim stp As Integer = st.IndexOf("About ")<br />

If stp > 0 Then<br />

Dim stR As String = st.Substring(stp + 24, 20) ’ex. 18,700 results (0.03 sec)...<br />

271


stp = stR.IndexOf("results")<br />

st = stR.Substring(0, stp).Replace(",", "") ’ex. 18,700 –>18700<br />

TextBox3.Text = Convert.ToInt32(Trim(st))<br />

If TextBox3.Text "0" Then RaiseEvent nextGene()<br />

Exit Sub<br />

End If<br />

End Select<br />

End If<br />

pageLoadTimes += 1<br />

End Sub<br />

End Class<br />

272<br />

Appendix


Appendix C<br />

Long ncRNAs<br />

We detected 25 differentially expressed long non-coding RNAs. Several <strong>of</strong> these<br />

display an expression pattern similar to a neighbouring protein-coding gene,<br />

including cancer-associated genes DKK1 and CTSC [113,461,551] and devel-<br />

opmental regulators IRX2, SIX3 and ZNF536 [412], suggesting that they may<br />

be functional RNAs regulating nearby genes [372] or represent transcription<br />

from active enhancers [230].<br />

273


C. Long ncRNAs Appendix<br />

Table C.1: Differentially expressed non-coding RNAs.<br />

Tag Accession Description In gene desert? PubMedID(s) Differential expression results Normalised tag counts<br />

log 2(F C) p-value FDR G144ED G144 G166 G179 CB541 CB660<br />

HOTAIRM1, a ncRNA from the<br />

HOXA locus. There is evidence<br />

CCGCCTTAATAAATGTA BC031342<br />

No 19144990 Inf 7.9E-10 4.3E-06 174.7 158.7 129.8 206.3 0.0 0.0<br />

that it might regulate HOXA gene<br />

expression.<br />

NEAT1, a ncRNA involved in<br />

TACATAATTACTAATCA NR_028272 paraspeckle formation and regu- Yes 20137068; 21170033 4.32 1.6E-06 2.2E-03 123.4 627.4 775.4 1927.1 18.5 91.8<br />

lated during NS cell differentiation.<br />

CDKN2BAS, an antisense transcript<br />

to CDKN2B that appears<br />

TGGATAAACAAAATGAA NR_003529 to function in Polycomb-mediated No 18185590; 20541999 Inf 1.4E-05 1.3E-02 18.0 33.6 125.8 0.0 0.0 0.0<br />

repression <strong>of</strong> tumor suppressors<br />

CDKN2A and CDKN2B.<br />

Transcript from the opposite strand<br />

TATCCCCAAATAAACAA NR_015382<br />

No None 4.42 3.2E-05 2.4E-02 151.4 123.2 332.4 135.3 9.3 9.4<br />

<strong>of</strong> CD27.<br />

Transcript from the opposite strand<br />

ATGGCACCATATTGTGT NR_015377<br />

No None -7.08 2.5E-05 2.0E-02 0.0 0.0 2.2 0.0 136.9 58.3<br />

<strong>of</strong> PAX8.<br />

Transcript from the opposite strand<br />

CCTGACCCTGCATCCCT BC013229<br />

No None Inf 1.4E-04 7.5E-02 1.7 0.0 36.9 68.3 0.0 0.0<br />

<strong>of</strong> MCF2L2.<br />

Transcript from the opposite strand<br />

TAGAACGGTGTTCCTCC BM977209<br />

No None Inf 1.7E-05 1.5E-02 0.0 0.0 121.6 24.7 0.0 0.0<br />

<strong>of</strong> TXNRD1.<br />

Transcript from CDKN2B intron<br />

ACAAACAAAAGCCTTCC DB099927<br />

No None Inf 1.4E-05 1.3E-02 41.9 105.5 51.8 0.0 0.0 0.0<br />

(sense strand).<br />

Transcript from gene desert near<br />

ATTACATTTATGTCCTT BC015429 DKK1. Correlated with DKK1 ex- Yes None Inf 2.8E-09 1.1E-05 0.0 0.0 389.5 18.0 0.0 0.0<br />

pression.<br />

TAACTGATCCTTAGATA BQ957425 Transcript from gene desert. Yes None Inf 2.0E-04 9.9E-02 0.0 0.0 66.8 30.8 0.0 0.0<br />

GAAACATTCCAAACCTA AK094154 Transcript from gene desert. Yes None Inf 1.3E-10 9.9E-07 0.0 0.0 328.5 243.9 0.0 0.0<br />

CAAATAAACTTTATACC BC063641 Transcript downstream <strong>of</strong> PRRG4. No None 7.53 2.4E-11 2.3E-07 4.2 68.0 267.3 1381.8 0.0 5.9<br />

Transcript between CTSC and<br />

GCTTTATTTTTTCTGCT BC038205 GRM5. Correlated with CTSC Yes None 7.46 5.4E-07 8.7E-04 54.0 96.2 49.6 126.3 0.0 1.0<br />

expression.<br />

GAGCCCAGACTAGATGG BF031226 Transcript from gene desert. Yes None Inf 3.9E-05 2.8E-02 1.4 0.0 30.7 99.7 0.0 0.0<br />

274<br />

Yes 12082533; 15872005 -Inf 3.5E-07 6.3E-04 0.0 0.0 0.0 0.0 21.8 283.9<br />

The tag is in last intron <strong>of</strong><br />

ncRNA NCRMS, perhaps detecting<br />

an NCRMS is<strong>of</strong>orm. NCRMS might<br />

be a host transcript for mir-1251<br />

and mir-135a-2. The tag also coincides<br />

with a SINE repeat.<br />

AAATATTAGTTTTTCTT CX868766


C. Long ncRNAs Appendix<br />

Tag Accession Description In gene desert? PubMedID(s) Differential expression results Normalised tag counts<br />

log 2(F C) p-value FDR G144ED G144 G166 G179 CB541 CB660<br />

Yes None 5.95 5.4E-05 3.7E-02 4.2 0.0 96.3 95.6 0.0 2.1<br />

AAATATGGATAAATGTA CA425887<br />

Yes None Inf 9.3E-10 4.9E-06 263.9 566.0 8.8 3.7 0.0 0.0<br />

AAATTGGTGCTGTTGCT BM679519<br />

No None -4.65 1.6E-05 1.4E-02 34.0 27.7 4.5 2.4 354.0 224.1<br />

GAAGGTCCCCCAGGGGT AK131287<br />

Yes None Inf 7.3E-06 7.5E-03 12.5 126.1 0.0 50.3 0.0 0.0<br />

Transcript from gene desert around<br />

FOXG1. Overlaps an EST similar<br />

to a region <strong>of</strong> downstream <strong>of</strong> the<br />

pseudoautosomal gene SPRY3.<br />

Transcript from gene desert harboring<br />

ZNF536 and TSHZ3. Correlated<br />

with ZNF536 expression.<br />

Intergenic transcript, or possibly a<br />

very long CACNG8 3’-UTR extension.<br />

Might be a host transcript for<br />

mir-935.<br />

Transcript from gene desert around<br />

KCNF1.<br />

Transcript from gene desert upstream<br />

<strong>of</strong> SIX3. Correlated with<br />

SIX3 expression.<br />

Transcript from gene desert downstream<br />

<strong>of</strong> SIX3. Correlated with<br />

SIX3 expression.<br />

Transcript from gene desert downstream<br />

<strong>of</strong> SIX3. Correlated with<br />

SIX3 expression.<br />

Transcript from gene desert near<br />

DCBLD2.<br />

Transcript from gene desert near<br />

IRX2. Correlated with IRX2 ex-<br />

GAATACAGATTAATCCT BG201257<br />

Yes 17084678 -Inf 6.9E-08 1.8E-04 0.0 0.0 0.0 0.0 162.1 173.2<br />

ATCATCACGTGAGAGAT AK126832<br />

Yes 17084678 -Inf 4.2E-07 7.2E-04 0.0 0.0 0.0 0.0 123.7 142.3<br />

TATAATAATAATGCTTA GD259214<br />

275<br />

Yes 17084678 -Inf 1.3E-05 1.2E-02 0.0 0.0 0.0 0.0 44.3 125.7<br />

GAGAGTGAATGTTTAAA CR623536<br />

Yes None 6.14 1.3E-06 1.9E-03 0.0 0.0 103.5 225.2 0.0 3.1<br />

TACACAATAAATATTTA BG166405<br />

Yes None -Inf 2.9E-05 2.2E-02 0.0 0.0 0.0 0.0 80.4 35.2<br />

ATAATAAAAGTATTTTT AA993778<br />

pression.<br />

No (1688605) Inf 1.2E-04 6.9E-02 4.2 6.1 73.4 27.6 0.0 0.0<br />

Transcript downstream <strong>of</strong> HLA-<br />

F, on opposite strand. Overlaps<br />

an antisense transcript to HLA-F<br />

(NR_026972).<br />

TTTTTCATCAAGAGGAA DB349183


Appendix D<br />

Glioblastoma Pathway<br />

D.1 Pathway Interactions<br />

Table D.1: Network interaction data for the integrated glioblastoma pathway. The<br />

first column identifies the first interactor; the second column identifies the interaction<br />

type; the third column identifies the second interactor. Sorted in alphabetical order<br />

on the first column interactor.<br />

First interactor Interaction Second interactor<br />

AKT1 activates MAP2K7<br />

AKT1 activates MDM2<br />

AKT1 activates TSC2<br />

AKT1 inhibits CDKN1A<br />

AKT1 inhibits CDKN1B<br />

AKT1 inhibits FOXO3<br />

AKT1 inhibits TSC-complex<br />

APAF1 leads-to APOPTOSIS<br />

ATM activates CHEK1<br />

ATM activates PRKDC<br />

ATM activates TP53<br />

BASC-complex includes BRCA1<br />

BASC-complex includes MSH6<br />

BASC-complex leads-to APOPTOSIS<br />

BASC-complex leads-to DNA-REPAIR<br />

BAX inhibits BCL2<br />

BCL2 inhibits APAF1<br />

BDNF activates NTRK2<br />

BID activates BAX<br />

BID activates CYCS<br />

BRCA1 interacts MSH6<br />

BTRC inhibits NFKB<br />

BTRC inhibits PHLPP1<br />

BUB1B inhibits IRS1<br />

BUB1B leads-to CELL-GROWTH<br />

BUB1B leads-to PROTEIN-SYNTHESIS<br />

CACN activates PKC<br />

CACN includes CACNA1A<br />

CACN includes CACNA1C<br />

CACN includes CACNG7<br />

CACN includes CACNG8<br />

CACN lets-in Calcium<br />

CALM1 activates CAMK1D<br />

CALM1 activates CAMK2B<br />

CALM1 activates PPEF1<br />

CALM1 interacts MAPT<br />

CALM1 interacts SNCA<br />

CALM1 interacts SYT1<br />

CAMK1D activates AKT1<br />

CAMK2A activates RAS<br />

CAMK2B interacts CAMK2A<br />

276


D.1 Pathway Interactions Appendix<br />

First interactor Interaction Second interactor<br />

CASP1 activates CASP3<br />

CASP1 activates IL1B<br />

CASP3 activates PARP<br />

CASP8 activates BID<br />

CBL inhibits RTK<br />

CBL interacts CRK<br />

CCND-CDK4/6-complex activates RB1<br />

CCND-CDK4/6-complex includes CCND1<br />

CCND-CDK4/6-complex includes CCND2<br />

CCND-CDK4/6-complex includes CDK4<br />

CCND-CDK4/6-complex includes CDK6<br />

CCNE-CDK2-complex activates RB1<br />

CCNE-CDK2-complex includes CCNE1<br />

CCNE-CDK2-complex includes CDK2<br />

CDC25C activates CDK1<br />

CDK1 leads-to G2-M-PROGRESSION<br />

CDKN1A inhibits CCND-CDK4/6-complex<br />

CDKN1A inhibits CCNE-CDK2-complex<br />

CDKN1B inhibits CCNE-CDK2-complex<br />

CDKN2A inhibits CCND-CDK4/6-complex<br />

CDKN2A:ARF inhibits MDM2<br />

CDKN2C inhibits CCND-CDK4/6-complex<br />

CEBPB activates DEC1<br />

CEBPB activates FOSL2<br />

CEBPB activates STAT3<br />

CEBPB activates_transcription CEBPB<br />

CHEK1 inhibits CDC25C<br />

CPLX2 inhibits SNARE-complex<br />

CPLX2 interacts STX1A<br />

CREBBP inhibits TP53<br />

CREBBP interacts CREB1<br />

CREBBP interacts JUN<br />

CREBBP interacts MYC<br />

CREBBP interacts NFATC2<br />

CRK interacts GAB1<br />

CYCS activates CASP9<br />

Calcium activates CALM1<br />

Calcium activates SYT1<br />

Cytosolic-antigen activates HLA-A<br />

DAG activates PKC<br />

DDIT3 leads-to APOPTOSIS<br />

DEC1 activates RUNX1<br />

DNA-DAMAGE activates ATM<br />

DNA-DAMAGE activates GADD45<br />

DOCK1 interacts CBL<br />

DOCK1 interacts CRK<br />

DOCK1 interacts ERBB2<br />

DOCK1 interacts PIK3R1<br />

DUSP16 inhibits MAPK14<br />

DUSP16 inhibits MAPK8<br />

DUSP5 inhibits MAPK14<br />

DUSP5 inhibits MAPK8<br />

E2F1 leads-to G1-S-PROGRESSION<br />

EGF interacts EGFR<br />

EGFR activates CASP1<br />

EIF4E leads-to CELL-GROWTH<br />

EIF4E leads-to PROTEIN-SYNTHESIS<br />

EIF4EBP1 inhibits EIF4E<br />

ELK1 interacts ELK4<br />

ELK1 interacts SRF<br />

ELK4 interacts MYC<br />

ELK4 interacts SRF<br />

EP300 activates TP53<br />

EPHB2 activates CCND-CDK4/6-complex<br />

EPHB2 inhibits TSC-complex<br />

EPHB2 leads-to CELL-CYCLE-PROGRESSION<br />

ERBB2 interacts EGFR<br />

ERBB2 interacts PIK3R1<br />

ERRFI1 inhibits EGFR<br />

Endocytosed-antigen activates MHC-classII<br />

FAS leads-to APOPTOSIS<br />

FASLG activates FAS<br />

FGF activates FGFR<br />

FGF includes FGF19<br />

FGF includes FGF23<br />

FGF23 interacts CBL<br />

277


D.1 Pathway Interactions Appendix<br />

First interactor Interaction Second interactor<br />

FGFR includes FGFR1<br />

FOSL2 activates DEC1<br />

FOSL2 activates RUNX1<br />

FOXO includes FOXO3<br />

FOXO leads-to APOPTOSIS<br />

FOXO3 activates-transcription CDKN1B<br />

FOXO3 activates-transcription FASLG<br />

GAB1 activates PI3K-class1a<br />

GADD45 activates CCND-CDK4/6-complex<br />

GADD45 activates MAP3K4<br />

GADD45 interacts PCNA<br />

GRB2 activates GAB1<br />

GRB2 activates SOS1<br />

HIF1A leads-to HYPOXIA<br />

HLA-A leads-to CD8-T-CELL-ACTIVATION<br />

HLA-DM activates MHC-classII<br />

HLA-DM includes HLA-DMA<br />

HLA-DM includes HLA-DMB<br />

HLA-DM inhibits CD74<br />

HLA-DRA interacts CBL<br />

IFI30 activates Endocytosed-antigen<br />

IFNG activates IFI30<br />

IFNG activates Immunoproteasome<br />

IGF1 activates IGF1R<br />

IGF2 leads-to APOPTOSIS<br />

IGFBP5 inhibits IGF2<br />

IKBKE activates NFKB<br />

IKBKE interacts MAP3K14<br />

IL1B activates IL1R1<br />

IL1B leads-to APOPTOSIS<br />

IL1B leads-to DIFFERENTIATION<br />

IL1B leads-to INFLAMMATION<br />

IL1B leads-to PROLIFERATION<br />

IL1R1 activates PIK3R1<br />

IL1R1 activates TRAF2<br />

IL1R1 activates TRAF6<br />

ILK activates AKT1<br />

IP3 activates ITPR1<br />

IRAK1 activates MAP3K14<br />

IRAK1 activates MAP3K1<br />

IRS1 activates PI3K-class1a<br />

ITGB activates ILK<br />

ITGB activates PTK2<br />

ITGB activates SHC<br />

ITGB includes ITGA4<br />

ITGB includes ITGBL1<br />

ITPR1 lets-in Calcium<br />

Immunoproteasome activates Cytosolic-antigen<br />

JUN leads-to DIFFERENTIATION<br />

JUN leads-to INFLAMMATION<br />

JUN leads-to PROLIFERATION<br />

MAP2K1/2 activates MAPK1/2<br />

MAP2K4 activates MAPK14<br />

MAP2K6 activates MAPK14<br />

MAP2K7 activates MAPK8<br />

MAP3K14 tentatively-activates NFKB<br />

MAP3K4 activates MAP2K4<br />

MAP3K4 interacts TRAF4<br />

MAP3K5 activates AKT1<br />

MAP3K7 activates MAP3K14<br />

MAPK1 activates ELK1<br />

MAPK1 activates MAPT<br />

MAPK1/2 activates RPS6KA3<br />

MAPK1/2 inhibits MAP2K1/2<br />

MAPK1/2 inhibits SOS1<br />

MAPK1/2 inhibits TSC2<br />

MAPK14 activates DDIT3<br />

MAPK8 activates JUN<br />

MAPK8 inhibits NFATC2<br />

MAPT interacts S100B<br />

MAPT interacts SNCA<br />

MAPT leads-to MICROTUBULE-DISASSEMBLY<br />

MDM2 inhibits RB1<br />

MDM2 inhibits TP53<br />

MDM4 inhibits TP53<br />

MHC-classI includes HLA-A<br />

278


D.1 Pathway Interactions Appendix<br />

First interactor Interaction Second interactor<br />

MHC-classII includes HLA-DPA1<br />

MHC-classII includes HLA-DPB1<br />

MHC-classII includes HLA-DQA1<br />

MHC-classII includes HLA-DQA2<br />

MHC-classII includes HLA-DQB1<br />

MHC-classII includes HLA-DRA<br />

MHC-classII includes HLA-DRB5<br />

MHC-classII interacts CD74<br />

MHC-classII leads-to CD4-T-CELL-ACTIVATION<br />

NFATC2 interacts EP300<br />

NFKB leads-to ANTI-APOPTOSIS<br />

NFKB leads-to INFLAMMATION<br />

NFKB leads-to PROLIFERATION<br />

NFKBIZ inhibits NFKB<br />

NGF activates NTRK1<br />

NR activates MAP2K6<br />

NR includes NR0B1<br />

NR includes NR1D1<br />

NR includes NR4A2<br />

NTRK1 activates TRAF6<br />

NTRK1 interacts GRB2<br />

NTRK1 interacts SHC1<br />

NTRK2 activates TRAF6<br />

NTRK2 interacts SHC1<br />

PARP includes PARP12<br />

PARP includes PARP3<br />

PARP leads-to APOPTOSIS<br />

PDGF activates PDGFR<br />

PDGFR includes PDGFRA<br />

PDGFR includes PDGFRB<br />

PDGFR interacts CBL<br />

PDGFR interacts CRK<br />

PDGFR interacts PI3K-class1a<br />

PDK1 activates AKT1<br />

PDPK1 activates AKT1<br />

PEA15 interacts CASP8<br />

PEA15 interacts MAPK1/2<br />

PERP activates CYCS<br />

PHLPP1 inhibits AKT1<br />

PI3K includes PI3K-class1a<br />

PI3K includes PI3K-class3<br />

PI3K-class1a activates PIP2<br />

PI3K-class1a interacts RB1<br />

PI3K-class3 activates TORC1-complex<br />

PIP2 becomes DAG<br />

PIP2 becomes IP3<br />

PIP2 becomes PIP3<br />

PIP3 activates ILK<br />

PIP3 activates PDK1<br />

PIP3 becomes PIP2<br />

PIP3 interacts AKT1<br />

PKC activates MAP2K1/2<br />

PKC activates RAF1<br />

PKC activates RAS<br />

PLC activates PIP2<br />

PLCG activates PIP2<br />

PLCG activates PKC<br />

PRKAB1 activates TSC-complex<br />

PRKDC activates TP53<br />

PTEN inhibits AKT1<br />

PTEN inhibits PIP3<br />

PTEN inhibits PTK2<br />

PTEN inhibits SHC<br />

PTEN inhibits TP53<br />

PTEN interacts GAB1<br />

PTEN interacts PIK3CA<br />

PTEN interacts PIK3R1<br />

PTK2 activates MAP3K4<br />

PTK2 activates PI3K-class1a<br />

PTK2 inhibits TSC-complex<br />

PTK2 interacts GRB2<br />

PTK2 leads-to ACTIN-ORGANIZATION<br />

RAF1 activates MAP2K1/2<br />

RAS activates PI3K<br />

RAS activates RAF1<br />

RAS includes HRAS<br />

279


D.1 Pathway Interactions Appendix<br />

First interactor Interaction Second interactor<br />

RAS includes KRAS<br />

RAS includes NRAS<br />

RB1 inhibits E2F1<br />

RHEB activates TORC1-complex<br />

RPS6KA3 activates BUB1B<br />

RPS6KA3 activates STK11<br />

RPS6KA3 inhibits TSC2<br />

RPS6KA3 interacts CREBBP<br />

RPS6KA3 interacts PEA15<br />

RTK activates IRS1<br />

RTK activates PI3K<br />

RTK activates PLCG<br />

RTK activates SHC<br />

RTK activates SRC<br />

RTK includes EGFR<br />

RTK includes FGFR<br />

RTK includes IGF1R<br />

RTK includes MET<br />

RTK includes PDGFR<br />

RTK interacts CRK<br />

RUNX1 activates JUN<br />

RUNX1 leads-to MESENCHYMAL-TRANSFORMATION<br />

S100B inhibits TP53<br />

SERPINB2 leads-to INHIBITION-ANGIOGENESIS<br />

SERPINB2 leads-to INHIBITION-METASTASIS<br />

SERPINE2 interacts SERPINB2<br />

SHC activates GRB2<br />

SHC includes SHC1<br />

SHC includes SHC4<br />

SHC4 interacts NTRK2<br />

SHISA5 interacts PERP<br />

SNARE-complex includes SNAP25<br />

SNARE-complex includes STX1A<br />

SNCA interacts CAMK2B<br />

SNCA interacts SNCAIP<br />

SOS1 activates RAS<br />

SPRY2 inhibits CBL<br />

SPRY2 inhibits RAS<br />

SRC activates GRB2<br />

SRC activates PI3K-class1a<br />

SRC inhibits PTEN<br />

SRF interacts DNA<br />

STAT3 activates FOSL2<br />

STAT3 activates RUNX1<br />

STAT3 activates_transcription STAT3<br />

STK11 activates PRKAB1<br />

SYT1 activates SNARE-complex<br />

TAB2 activates MAP3K7<br />

TGFA activates EGFR<br />

TGFB activates TGFBR<br />

TGFBR activates NR<br />

TLR4 activates TRAF6<br />

TNF activates TNFR<br />

TNF includes TNFSF4<br />

TNFR activates TRAF2<br />

TNFR includes TNFRSF14<br />

TORC1-complex activates BUB1B<br />

TORC1-complex includes MLST8<br />

TORC1-complex includes MTOR<br />

TORC1-complex includes RPTOR<br />

TORC1-complex inhibits EIF4EBP1<br />

TORC2-complex activates AKT1<br />

TORC2-complex includes MAPKAP1<br />

TORC2-complex includes MLST8<br />

TORC2-complex includes MTOR<br />

TORC2-complex includes RICTOR<br />

TORC2-complex leads-to ACTIN-ORGANIZATION<br />

TP53 activates BRCA1<br />

TP53 activates HIF1A<br />

TP53 activates MDM2<br />

TP53 activates S100B<br />

TP53 activates-transcription BAX<br />

TP53 activates-transcription CDKN1A<br />

TP53 activates-transcription FAS<br />

TP53 activates-transcription GADD45<br />

TP53 activates-transcription IGFBP5<br />

280


D.1 Pathway Interactions Appendix<br />

First interactor Interaction Second interactor<br />

TP53 activates-transcription PTEN<br />

TP53 activates-transcription SERPINE2<br />

TP53 activates-transcription SHISA5<br />

TP53 inhibits CDK1<br />

TP53 inhibits CDK2<br />

TP53 inhibits-transcription BCL2<br />

TP53 inhibits-transcription TIMP3<br />

TP53 interacts CDKN2C<br />

TP53 leads-to APOPTOSIS<br />

TRAF2 activates MAP3K5<br />

TRAF4 activates MAPK8<br />

TRAF6 activates IRAK1<br />

TRAF6 interacts TAB2<br />

TSC-complex includes TSC1<br />

TSC-complex includes TSC2<br />

TSC2 inhibits RHEB<br />

281


D.2 Pathway Images Appendix<br />

D.2 Pathway Images<br />

Figure D.1: Integrated GBM pathway overlaid with Tag-seq G144 expression data.<br />

The colour intensity <strong>of</strong> the nodes (green) indicates the magnitude <strong>of</strong> the expression.<br />

282


D.2 Pathway Images Appendix<br />

Figure D.2: Integrated GBM pathway overlaid with Tag-seq G144ED expression<br />

data. The colour intensity <strong>of</strong> the nodes (green) indicates the magnitude <strong>of</strong> the<br />

expression.<br />

283


D.2 Pathway Images Appendix<br />

Figure D.3: Integrated GBM pathway with Tag-seq G166 expression data. The<br />

colour intensity <strong>of</strong> the nodes (green) indicates the magnitude <strong>of</strong> the expression.<br />

284


D.2 Pathway Images Appendix<br />

Figure D.4: Integrated GBM pathway with Tag-seq G179 expression data. The<br />

colour intensity <strong>of</strong> the nodes (green) indicates the magnitude <strong>of</strong> the expression.<br />

285


Appendix E<br />

Exon Array Data<br />

Table E.1: Fold-changes measured by exon array and filtered at FDR


E. Exon Array Data Appendix<br />

Average expression across samples Fold-change p-value FDR Ensembl Gene ID Symbol<br />

4.92 -2.647 0.000 0.000000 ENSG00000141682 PMAIP1<br />

7.47 -1.782 0.000 0.000000 ENSG00000144730 IL17RD<br />

7.37 -1.397 0.000 0.000000 ENSG00000145012 LPP<br />

7.8 -1.172 0.000 0.000000 ENSG00000145431 PDGFC<br />

4.45 -2.471 0.000 0.000000 ENSG00000145708 CRHBP<br />

5.27 -1.439 0.000 0.000000 ENSG00000147234 FRMPD3<br />

7.75 -4.124 0.000 0.000000 ENSG00000149591 TAGLN<br />

5.48 -3.438 0.000 0.000000 ENSG00000151150 ANK3<br />

6.55 -3.912 0.000 0.000000 ENSG00000151388 ADAMTS12<br />

5.76 -4.527 0.000 0.000000 ENSG00000151572 ANO4<br />

7.52 -5.064 0.000 0.000000 ENSG00000151892 GFRA1<br />

8.11 -2.063 0.000 0.000000 ENSG00000152154 TMEM178A<br />

3.22 -1.658 0.000 0.000000 ENSG00000153993 SEMA3D<br />

5.1 -2.672 0.000 0.000000 ENSG00000155966 AFF2<br />

5.84 -2.395 0.000 0.000000 ENSG00000156675 RAB11FIP1<br />

5.63 -1.037 0.000 0.000000 ENSG00000157110 RBPMS<br />

5.49 -2.472 0.000 0.000000 ENSG00000159217 IGF2BP1<br />

5.97 -1.422 0.000 0.000000 ENSG00000159433 STARD9<br />

6.62 -1.718 0.000 0.000000 ENSG00000162849 KIF26B<br />

4.79 -3.556 0.000 0.000000 ENSG00000163071 SPATA18<br />

7.66 -1.821 0.000 0.000000 ENSG00000163110 PDLIM5<br />

6.5 -1.878 0.000 0.000000 ENSG00000163297 ANTXR2<br />

7.09 -3.748 0.000 0.000000 ENSG00000163814 CDCP1<br />

4.72 -1.731 0.000 0.000000 ENSG00000164002 DEM1<br />

5.01 -1.766 0.000 0.000000 ENSG00000164932 CTHRC1<br />

8.12 -1.339 0.000 0.000000 ENSG00000164938 TP53INP1<br />

6.06 -5.065 0.000 0.000000 ENSG00000165588 OTX2<br />

7.09 -2.589 0.000 0.000000 ENSG00000167081 PBX3<br />

7.26 -1.998 0.000 0.000000 ENSG00000167693 NXN<br />

8.27 -1.438 0.000 0.000000 ENSG00000169439 SDC2<br />

6.61 -1.004 0.000 0.000000 ENSG00000170561 IRX2<br />

4.48 -2.288 0.000 0.000000 ENSG00000172123 SLFN12<br />

5.14 -4.167 0.000 0.000000 ENSG00000172260 NEGR1<br />

7.6 -1.24 0.000 0.000000 ENSG00000172667 ZMAT3<br />

5.4 -1.874 0.000 0.000000 ENSG00000173068 BNC2<br />

6.66 -3.789 0.000 0.000000 ENSG00000173530 TNFRSF10D<br />

4.85 -1.134 0.000 0.000000 ENSG00000173535 TNFRSF10C<br />

6.29 -1.907 0.000 0.000000 ENSG00000174099 MSRB3<br />

8.45 -1.552 0.000 0.000000 ENSG00000174136 RGMB<br />

3.84 -1.408 0.000 0.000000 ENSG00000176040 TMPRSS7<br />

7.2 -1.425 0.000 0.000000 ENSG00000176720 BOK<br />

8.22 -1.108 0.000 0.000000 ENSG00000177119 ANO6<br />

6.03 -1.552 0.000 0.000000 ENSG00000178573 MAF<br />

6.78 -4.433 0.000 0.000000 ENSG00000184613 NELL2<br />

4.67 -1.519 0.000 0.000000 ENSG00000184809 C21orf88<br />

6.28 -1.561 0.000 0.000000 ENSG00000184985 SORCS2<br />

5.2 -2.054 0.000 0.000000 ENSG00000185046 ANKS1B<br />

6.45 -4.02 0.000 0.000000 ENSG00000185274 WBSCR17<br />

8.51 -2.037 0.000 0.000000 ENSG00000185567 AHNAK2<br />

7.01 -2.956 0.000 0.000000 ENSG00000189184 PCDH18<br />

6.47 -2.842 0.000 0.000000 ENSG00000196730 DAPK1<br />

7.77 -1.504 0.000 0.000000 ENSG00000196923 PDLIM7<br />

5.87 -3.559 0.000 0.000000 ENSG00000198796 ALPK2<br />

3.59 -2.326 0.000 0.000000 ENSG00000204764 RANBP17<br />

5.82 -2.638 0.000 0.000000 ENSG00000204767 FAM196B<br />

8.15 -1.501 0.000 0.000000 ENSG00000205213 LGR4<br />

5.06 -1.915 0.000 0.000000 ENSG00000206538 VGLL3<br />

4.48 -1.527 0.000 0.000000 ENSG00000213186 TRIM59<br />

5.66 -2.628 0.000 0.000000 ENSG00000244694 PTCHD4<br />

9.77 -1.446 0.000 0.000010 ENSG00000026025 VIM<br />

8.93 -1.491 0.000 0.000010 ENSG00000035403 VCL<br />

7.97 -3.845 0.000 0.000010 ENSG00000079931 MOXD1<br />

5.7 -1.554 0.000 0.000010 ENSG00000080546 SESN1<br />

11.03 -1.357 0.000 0.000010 ENSG00000099194 SCD<br />

9.2 -1.406 0.000 0.000010 ENSG00000100345 MYH9<br />

7.34 -1.425 0.000 0.000010 ENSG00000100918 REC8<br />

4.65 -1.337 0.000 0.000010 ENSG00000107614 TRDMT1<br />

7.08 -1.026 0.000 0.000010 ENSG00000109654 TRIM2<br />

7.28 -1.459 0.000 0.000010 ENSG00000112144 ICK<br />

5.52 -0.778 0.000 0.000010 ENSG00000115648 MLPH<br />

8.11 -3.461 0.000 0.000010 ENSG00000117114 LPHN2<br />

6.19 -1.353 0.000 0.000010 ENSG00000125257 ABCC4<br />

8.36 -2.308 0.000 0.000010 ENSG00000129038 LOXL1<br />

7.12 -2.728 0.000 0.000010 ENSG00000131378 RFTN1<br />

4.4 -2.19 0.000 0.000010 ENSG00000134516 DOCK2<br />

8.18 -1.64 0.000 0.000010 ENSG00000134871 COL4A2<br />

287


E. Exon Array Data Appendix<br />

Average expression across samples Fold-change p-value FDR Ensembl Gene ID Symbol<br />

5.66 -1.108 0.000 0.000010 ENSG00000135299 ANKRD6<br />

6.65 -2.293 0.000 0.000010 ENSG00000139211 AMIGO2<br />

6.01 -0.834 0.000 0.000010 ENSG00000143466 IKBKE<br />

6.94 -1.227 0.000 0.000010 ENSG00000143772 ITPKB<br />

5.93 -3.525 0.000 0.000010 ENSG00000147459 DOCK5<br />

7.08 -1.588 0.000 0.000010 ENSG00000149269 PAK1<br />

4.75 -1.397 0.000 0.000010 ENSG00000149948 HMGA2<br />

8.76 -2.143 0.000 0.000010 ENSG00000150551 LYPD1<br />

5.98 -1.944 0.000 0.000010 ENSG00000150687 PRSS23<br />

8.52 -1.26 0.000 0.000010 ENSG00000150938 CRIM1<br />

7.07 -1.701 0.000 0.000010 ENSG00000151474 FRMD4A<br />

7.81 -1.827 0.000 0.000010 ENSG00000152104 PTPN14<br />

7.53 -1.628 0.000 0.000010 ENSG00000153707 PTPRD<br />

6.11 -1.021 0.000 0.000010 ENSG00000166016 ABTB2<br />

6.79 -1.103 0.000 0.000010 ENSG00000166444 ST5<br />

6.42 -0.938 0.000 0.000010 ENSG00000168140 VASN<br />

6.31 -1.634 0.000 0.000010 ENSG00000171533 MAP6<br />

6.1 -1.588 0.000 0.000010 ENSG00000171843 MLLT3<br />

7.3 -1.553 0.000 0.000010 ENSG00000172175 MALT1<br />

7.77 -1.621 0.000 0.000010 ENSG00000172638 EFEMP2<br />

7.19 -1.756 0.000 0.000010 ENSG00000173848 NET1<br />

6 -1.068 0.000 0.000010 ENSG00000180592 SKIDA1<br />

6.47 -1.659 0.000 0.000010 ENSG00000182985 CADM1<br />

6.33 -2.532 0.000 0.000010 ENSG00000187720 THSD4<br />

9.32 -1.343 0.000 0.000010 ENSG00000188042 ARL4C<br />

7.93 -1.344 0.000 0.000010 ENSG00000197702 PARVA<br />

9.34 -1.438 0.000 0.000020 ENSG00000002586 CD99<br />

7.95 -1.593 0.000 0.000020 ENSG00000065923 SLC9A7<br />

5.65 -1.606 0.000 0.000020 ENSG00000071205 ARHGAP10<br />

7.14 -1.517 0.000 0.000020 ENSG00000072401 UBE2D1<br />

7.46 -1.853 0.000 0.000020 ENSG00000085377 PREP<br />

6.82 -1.943 0.000 0.000020 ENSG00000103460 TOX3<br />

9.2 -1.742 0.000 0.000020 ENSG00000112972 HMGCS1<br />

5.82 -1.582 0.000 0.000020 ENSG00000114861 FOXP1<br />

7.28 -0.983 0.000 0.000020 ENSG00000128739 SNRPN<br />

7.58 -1.104 0.000 0.000020 ENSG00000129474 AJUBA<br />

5.5 -1.192 0.000 0.000020 ENSG00000129682 FGF13<br />

7.08 -1.836 0.000 0.000020 ENSG00000137942 FNBP1L<br />

8.55 -1.31 0.000 0.000020 ENSG00000154380 ENAH<br />

5.64 -2.757 0.000 0.000020 ENSG00000165659 DACH1<br />

9.13 -1.323 0.000 0.000020 ENSG00000166033 HTRA1<br />

7.58 -2.2 0.000 0.000020 ENSG00000168672 FAM84B<br />

7.44 -1.906 0.000 0.000020 ENSG00000170175 CHRNB1<br />

7.08 -0.917 0.000 0.000020 ENSG00000182197 EXT1<br />

4.37 -1.48 0.000 0.000020 ENSG00000183778 B3GALT5<br />

7.62 -3.42 0.000 0.000020 ENSG00000187955 COL14A1<br />

10.42 -1.09 0.000 0.000020 ENSG00000196924 FLNA<br />

3.77 -1.626 0.000 0.000020 ENSG00000203995 ZYG11A<br />

8.82 -1.245 0.000 0.000020 ENSG00000213625 LEPROT<br />

7.54 -1.127 0.000 0.000020 ENSG00000250588 IQCJ-SCHIP1<br />

4.94 -1.757 0.000 0.000020 ENSG00000255994 C14orf162<br />

8.77 -1.205 0.000 0.000030 ENSG00000065150 IPO5<br />

7.74 -1.274 0.000 0.000030 ENSG00000073712 FERMT2<br />

6.31 -1.635 0.000 0.000030 ENSG00000100592 DAAM1<br />

8.63 -1.933 0.000 0.000030 ENSG00000125266 EFNB2<br />

6.64 -3.548 0.000 0.000030 ENSG00000126010 GRPR<br />

7.09 -4.473 0.000 0.000030 ENSG00000132429 POPDC3<br />

8.83 -0.845 0.000 0.000030 ENSG00000137076 TLN1<br />

5.45 -1.938 0.000 0.000030 ENSG00000137831 UACA<br />

9.29 -1.364 0.000 0.000030 ENSG00000138448 ITGAV<br />

7.34 -2.1 0.000 0.000030 ENSG00000139687 RB1<br />

7.03 -2.562 0.000 0.000030 ENSG00000145536 ADAMTS16<br />

8.41 -1.569 0.000 0.000030 ENSG00000148484 RSU1<br />

7.15 -0.853 0.000 0.000030 ENSG00000150457 LATS2<br />

7.54 -1.712 0.000 0.000030 ENSG00000153976 HS3ST3A1<br />

6.68 -2.064 0.000 0.000030 ENSG00000154556 SORBS2<br />

5.6 -1.892 0.000 0.000030 ENSG00000166450 PRTG<br />

7 -1.416 0.000 0.000030 ENSG00000169047 IRS1<br />

5.16 -1.289 0.000 0.000030 ENSG00000183840 GPR39<br />

6.2 -1.112 0.000 0.000030 ENSG00000203772 SPRN<br />

6.92 -0.86 0.000 0.000040 ENSG00000013364 MVP<br />

6.65 -1.951 0.000 0.000040 ENSG00000069869 NEDD4<br />

5.28 -1.552 0.000 0.000040 ENSG00000079102 RUNX1T1<br />

6.07 -1.116 0.000 0.000040 ENSG00000085741 WNT11<br />

6.6 -1.686 0.000 0.000040 ENSG00000099284 H2AFY2<br />

7.93 -1.989 0.000 0.000040 ENSG00000101670 LIPG<br />

288


E. Exon Array Data Appendix<br />

Average expression across samples Fold-change p-value FDR Ensembl Gene ID Symbol<br />

6.88 -0.837 0.000 0.000040 ENSG00000107758 PPP3CB<br />

7.91 -1.277 0.000 0.000040 ENSG00000128829 EIF2AK4<br />

8.37 -0.912 0.000 0.000040 ENSG00000136478 TEX2<br />

6.12 -1.235 0.000 0.000040 ENSG00000137502 RAB30<br />

7.56 -1.128 0.000 0.000040 ENSG00000139668 WDFY2<br />

3.8 -1.027 0.000 0.000040 ENSG00000150672 DLG2<br />

7.69 -0.971 0.000 0.000040 ENSG00000163625 WDFY3<br />

10.31 -1.446 0.000 0.000040 ENSG00000168615 ADAM9<br />

7.77 -1.029 0.000 0.000040 ENSG00000181827 RFX7<br />

8.07 -0.637 0.000 0.000040 ENSG00000198752 CDC42BPB<br />

7.51 -0.797 0.000 0.000040 ENSG00000214717 ZBED1<br />

6.19 -1.808 0.000 0.000050 ENSG00000066468 FGFR2<br />

5.66 -2.079 0.000 0.000050 ENSG00000081803 CADPS2<br />

7.49 -0.96 0.000 0.000050 ENSG00000087088 BAX<br />

6.32 -1.122 0.000 0.000050 ENSG00000100968 NFATC4<br />

8.22 -1.297 0.000 0.000050 ENSG00000102893 PHKB<br />

5.64 -1.108 0.000 0.000050 ENSG00000108239 TBC1D12<br />

9 -2.672 0.000 0.000050 ENSG00000112276 BVES<br />

5.95 -2.589 0.000 0.000050 ENSG00000115232 ITGA4<br />

4.59 -1.69 0.000 0.000050 ENSG00000124134 KCNS1<br />

7.48 -2.329 0.000 0.000050 ENSG00000136928 GABBR2<br />

8.18 -1.075 0.000 0.000050 ENSG00000150347 ARID5B<br />

6.01 -2.228 0.000 0.000050 ENSG00000162745 OLFML2B<br />

6.77 -1.239 0.000 0.000050 ENSG00000166073 GPR176<br />

4.66 -3.976 0.000 0.000050 ENSG00000166342 NETO1<br />

4.69 -1.97 0.000 0.000050 ENSG00000172403 SYNPO2<br />

8.1 -0.895 0.000 0.000050 ENSG00000176014 TUBB6<br />

8.3 -1.04 0.000 0.000060 ENSG00000100403 ZC3H7B<br />

7.95 -1.065 0.000 0.000060 ENSG00000143344 RGL1<br />

7.39 -1.509 0.000 0.000060 ENSG00000171862 PTEN<br />

7.99 -0.895 0.000 0.000060 ENSG00000179820 MYADM<br />

8.3 -0.826 0.000 0.000060 ENSG00000187079 TEAD1<br />

5.29 -2.689 0.000 0.000070 ENSG00000101311 FERMT1<br />

9.82 -0.993 0.000 0.000070 ENSG00000104549 SQLE<br />

7.35 -0.867 0.000 0.000070 ENSG00000112655 PTK7<br />

7.33 -0.735 0.000 0.000070 ENSG00000130338 TULP4<br />

10.17 -0.996 0.000 0.000070 ENSG00000131236 CAP1<br />

8.78 -1.538 0.000 0.000070 ENSG00000134824 FADS2<br />

6.55 -0.792 0.000 0.000070 ENSG00000137936 BCAR3<br />

9.79 -0.973 0.000 0.000070 ENSG00000150093 ITGB1<br />

6.93 -1.288 0.000 0.000070 ENSG00000168502 SOGA2<br />

8.38 -1.506 0.000 0.000070 ENSG00000179431 FJX1<br />

8.32 -1.283 0.000 0.000070 ENSG00000197043 ANXA6<br />

5.8 -1.297 0.000 0.000070 ENSG00000198113 TOR4A<br />

7.19 -1.449 0.000 0.000070 ENSG00000205269 TMEM170B<br />

9.7 -1.29 0.000 0.000080 ENSG00000071127 WDR1<br />

7.75 -1.188 0.000 0.000080 ENSG00000073792 IGF2BP2<br />

7.37 -0.762 0.000 0.000080 ENSG00000100139 MICALL1<br />

6.25 -1.627 0.000 0.000080 ENSG00000140557 ST8SIA2<br />

4.93 -2.465 0.000 0.000080 ENSG00000148798 INA<br />

7.75 -1.245 0.000 0.000080 ENSG00000151612 ZNF827<br />

6.56 -1.118 0.000 0.000080 ENSG00000151718 WWC2<br />

7.26 -1.995 0.000 0.000080 ENSG00000174721 FGFBP3<br />

7.34 -1.183 0.000 0.000080 ENSG00000175115 PACS1<br />

7.06 -1.147 0.000 0.000080 ENSG00000182287 AP1S2<br />

7.79 -1.122 0.000 0.000080 ENSG00000187239 FNBP1<br />

6.27 -0.958 0.000 0.000080 ENSG00000229619 AC106722.1<br />

10.55 -1.573 0.000 0.000090 ENSG00000101608 MYL12A<br />

5.59 -1.066 0.000 0.000090 ENSG00000101665 SMAD7<br />

6.15 -1.436 0.000 0.000090 ENSG00000107518 ATRNL1<br />

5.53 -3.929 0.000 0.000090 ENSG00000144227 NXPH2<br />

5.63 -0.965 0.000 0.000090 ENSG00000158246 FAM46B<br />

6.62 -1.761 0.000 0.000090 ENSG00000165323 FAT3<br />

7.74 -1.946 0.000 0.000100 ENSG00000102996 MMP15<br />

8.65 -1.862 0.000 0.000100 ENSG00000104290 FZD3<br />

6.36 -2.28 0.000 0.000100 ENSG00000151136 BTBD11<br />

7.57 -2.996 0.000 0.000100 ENSG00000152661 GJA1<br />

7.28 -0.967 0.000 0.000100 ENSG00000166912 MTMR10<br />

7.44 -1.18 0.000 0.000110 ENSG00000058091 CDK14<br />

8.62 -0.924 0.000 0.000110 ENSG00000073921 PICALM<br />

5.74 -1.788 0.000 0.000110 ENSG00000152128 TMEM163<br />

7.49 -1.413 0.000 0.000110 ENSG00000158966 CACHD1<br />

5.56 -1.457 0.000 0.000110 ENSG00000165449 SLC16A9<br />

3.3 -2.026 0.000 0.000110 ENSG00000170571 EMB<br />

6.66 -0.922 0.000 0.000110 ENSG00000174307 PHLDA3<br />

5.19 -1.959 0.000 0.000110 ENSG00000183691 NOG<br />

289


E. Exon Array Data Appendix<br />

Average expression across samples Fold-change p-value FDR Ensembl Gene ID Symbol<br />

6.73 -0.823 0.000 0.000120 ENSG00000011114 BTBD7<br />

6.86 -0.624 0.000 0.000120 ENSG00000037757 MRI1<br />

4.78 -1.277 0.000 0.000120 ENSG00000068781 STON1-GTF2A1L<br />

9.49 -1.337 0.000 0.000120 ENSG00000075213 SEMA3A<br />

4.78 -1.205 0.000 0.000120 ENSG00000108187 PBLD<br />

7.01 -1.596 0.000 0.000120 ENSG00000109586 GALNT7<br />

6.73 -0.668 0.000 0.000120 ENSG00000110237 ARHGEF17<br />

6.37 -1.075 0.000 0.000120 ENSG00000126790 L3HYPDH<br />

6.21 -1.532 0.000 0.000120 ENSG00000165996 PTPLA<br />

6.16 -1.903 0.000 0.000120 ENSG00000180914 OXTR<br />

9.41 -1.204 0.000 0.000120 ENSG00000196576 PLXNB2<br />

5.41 -1.323 0.000 0.000120 ENSG00000197646 PDCD1LG2<br />

6.14 -0.846 0.000 0.000130 ENSG00000090975 PITPNM2<br />

6.87 -0.981 0.000 0.000130 ENSG00000148737 TCF7L2<br />

7.63 -1.404 0.000 0.000130 ENSG00000177508 IRX3<br />

6.23 -2.345 0.000 0.000140 ENSG00000104332 SFRP1<br />

8.2 -1.368 0.000 0.000140 ENSG00000140682 TGFB1I1<br />

7.55 -0.981 0.000 0.000140 ENSG00000148468 FAM171A1<br />

6.12 -0.812 0.000 0.000140 ENSG00000162804 SNED1<br />

7.73 -1.008 0.000 0.000140 ENSG00000196865 NHLRC2<br />

7.36 -1.145 0.000 0.000150 ENSG00000055163 CYFIP2<br />

6.64 -0.859 0.000 0.000150 ENSG00000072422 RHOBTB1<br />

6.33 -1.765 0.000 0.000150 ENSG00000130176 CNN1<br />

8.66 -0.711 0.000 0.000150 ENSG00000130638 ATXN10<br />

8.32 -0.937 0.000 0.000150 ENSG00000139793 MBNL2<br />

5.8 -1.718 0.000 0.000150 ENSG00000173320 STOX2<br />

7.41 -1.382 0.000 0.000150 ENSG00000176788 BASP1<br />

5.41 -4.49 0.000 0.000150 ENSG00000187714 SLC18A3<br />

6.97 -0.57 0.000 0.000160 ENSG00000061273 HDAC7<br />

8.41 -1.415 0.000 0.000160 ENSG00000163430 FSTL1<br />

6.58 -1.178 0.000 0.000160 ENSG00000169436 COL22A1<br />

8.98 -1.848 0.000 0.000170 ENSG00000135048 TMEM2<br />

8.41 -1.185 0.000 0.000170 ENSG00000137710 RDX<br />

4.5 -1.657 0.000 0.000170 ENSG00000154027 AK5<br />

8.87 -0.936 0.000 0.000170 ENSG00000167460 TPM4<br />

7.64 -1.238 0.000 0.000170 ENSG00000182319 PRAGMIN<br />

8.22 -0.609 0.000 0.000170 ENSG00000182534 MXRA7<br />

8.7 -1.291 0.000 0.000170 ENSG00000186575 NF2<br />

7.62 -2.017 0.000 0.000180 ENSG00000084710 EFR3B<br />

7.84 -0.796 0.000 0.000180 ENSG00000108091 CCDC6<br />

8.51 -1.519 0.000 0.000180 ENSG00000148848 ADAM12<br />

9.53 -0.645 0.000 0.000180 ENSG00000168175 MAPK1IP1L<br />

7.32 -1.041 0.000 0.000180 ENSG00000170525 PFKFB3<br />

7.31 -1.108 0.000 0.000180 ENSG00000178764 ZHX2<br />

6.15 -0.992 0.000 0.000190 ENSG00000116675 DNAJC6<br />

6.52 -1.174 0.000 0.000190 ENSG00000131370 SH3BP5<br />

6.08 -0.978 0.000 0.000190 ENSG00000143816 WNT9A<br />

6.26 -1.511 0.000 0.000190 ENSG00000255103 KIAA0754<br />

6.71 -1.362 0.000 0.000200 ENSG00000122335 SERAC1<br />

5.2 -3.22 0.000 0.000200 ENSG00000123119 NECAB1<br />

5.45 -1.76 0.000 0.000200 ENSG00000138696 BMPR1B<br />

6.28 -2.18 0.000 0.000200 ENSG00000146426 TIAM2<br />

8.62 -0.877 0.000 0.000200 ENSG00000152558 TMEM123<br />

6.89 -0.815 0.000 0.000210 ENSG00000068383 INPP5A<br />

7.22 -1.283 0.000 0.000210 ENSG00000105974 CAV1<br />

9.12 -1.876 0.000 0.000210 ENSG00000106484 MEST<br />

7.38 -1.482 0.000 0.000210 ENSG00000113328 CCNG1<br />

6.45 -1.103 0.000 0.000210 ENSG00000119681 LTBP2<br />

6.12 -0.72 0.000 0.000210 ENSG00000132334 PTPRE<br />

6.68 -1.403 0.000 0.000210 ENSG00000188158 NHS<br />

7.53 -0.946 0.000 0.000230 ENSG00000107819 SFXN3<br />

7.73 -0.772 0.000 0.000230 ENSG00000120254 MTHFD1L<br />

7.79 -0.638 0.000 0.000230 ENSG00000122863 CHST3<br />

3.92 -2.068 0.000 0.000230 ENSG00000158164 TMSB15A<br />

7.94 -0.812 0.000 0.000230 ENSG00000185950 IRS2<br />

7.35 -1.124 0.000 0.000240 ENSG00000067064 IDI1<br />

7.4 -0.902 0.000 0.000240 ENSG00000124788 ATXN1<br />

5.98 -0.83 0.000 0.000240 ENSG00000138131 LOXL4<br />

7.87 -1.371 0.000 0.000240 ENSG00000187498 COL4A1<br />

7.87 -0.797 0.000 0.000240 ENSG00000198951 NAGA<br />

4.77 -1.178 0.000 0.000250 ENSG00000100285 NEFH<br />

7.33 -0.837 0.000 0.000250 ENSG00000103335 PIEZO1<br />

8.26 -2.268 0.000 0.000250 ENSG00000132470 ITGB4<br />

8.65 -1.241 0.000 0.000250 ENSG00000181019 NQO1<br />

5.67 -0.561 0.000 0.000250 ENSG00000198832 RP3-412A9.11<br />

6.86 -0.871 0.000 0.000260 ENSG00000070269 C14orf101<br />

290


E. Exon Array Data Appendix<br />

Average expression across samples Fold-change p-value FDR Ensembl Gene ID Symbol<br />

5.37 -1.251 0.000 0.000260 ENSG00000113448 PDE4D<br />

7.86 -0.688 0.000 0.000260 ENSG00000139645 ANKRD52<br />

4.6 -1.255 0.000 0.000260 ENSG00000165626 BEND7<br />

6.96 -0.585 0.000 0.000260 ENSG00000182541 LIMK2<br />

6.14 -0.788 0.000 0.000270 ENSG00000014914 MTMR11<br />

8.85 -1.086 0.000 0.000270 ENSG00000064666 CNN2<br />

7.38 -0.682 0.000 0.000270 ENSG00000128283 CDC42EP1<br />

8.39 -1.216 0.000 0.000270 ENSG00000162909 CAPN2<br />

8.41 -0.967 0.000 0.000270 ENSG00000176903 PNMA1<br />

5.62 -1.998 0.000 0.000270 ENSG00000182732 RGS6<br />

6.79 -0.841 0.000 0.000280 ENSG00000151553 FAM160B1<br />

7.41 -1.128 0.000 0.000280 ENSG00000170365 SMAD1<br />

8.41 -1.273 0.000 0.000280 ENSG00000183722 LHFP<br />

3.46 -1.447 0.000 0.000290 ENSG00000022556 NLRP2<br />

2.61 -0.607 0.000 0.000290 ENSG00000133640 LRRIQ1<br />

7.3 -1.36 0.000 0.000300 ENSG00000185339 TCN2<br />

5.04 -1.653 0.000 0.000300 ENSG00000215475 SIAH3<br />

9 -1.175 0.000 0.000310 ENSG00000107779 BMPR1A<br />

8.88 -1.175 0.000 0.000310 ENSG00000117298 ECE1<br />

8.39 -0.598 0.000 0.000310 ENSG00000198954 KIAA1279<br />

5.41 -2.252 0.000 0.000320 ENSG00000124610 HIST1H1A<br />

7.1 -1.379 0.000 0.000320 ENSG00000205726 ITSN1<br />

7.71 -0.927 0.000 0.000330 ENSG00000102753 KPNA3<br />

7 -0.851 0.000 0.000330 ENSG00000143553 SNAPIN<br />

9.78 -0.783 0.000 0.000330 ENSG00000147416 ATP6V1B2<br />

5.61 -2.025 0.000 0.000330 ENSG00000188517 COL25A1<br />

6.51 -2.126 0.000 0.000340 ENSG00000152977 ZIC1<br />

7.7 -0.908 0.000 0.000340 ENSG00000154001 PPP2R5E<br />

4.97 -0.768 0.000 0.000340 ENSG00000203877 RIPPLY2<br />

6.95 -1.345 0.000 0.000350 ENSG00000080298 RFX3<br />

8.79 -1.528 0.000 0.000350 ENSG00000082781 ITGB5<br />

8.19 -0.882 0.000 0.000350 ENSG00000175662 TOM1L2<br />

4.95 -2.667 0.000 0.000360 ENSG00000089472 HEPH<br />

9.11 -1.105 0.000 0.000360 ENSG00000102898 NUTF2<br />

7.58 -1.796 0.000 0.000360 ENSG00000125430 HS3ST3B1<br />

9.2 -1.289 0.000 0.000360 ENSG00000172380 GNG12<br />

6.85 -1.643 0.000 0.000370 ENSG00000139263 LRIG3<br />

8.35 -0.864 0.000 0.000370 ENSG00000165476 REEP3<br />

6.9 -1.305 0.000 0.000380 ENSG00000040199 PHLPP2<br />

8.79 -0.701 0.000 0.000380 ENSG00000120733 KDM3B<br />

5 -1.693 0.000 0.000380 ENSG00000143473 KCNH1<br />

6.27 -2.298 0.000 0.000380 ENSG00000173917 HOXB2<br />

6.76 -0.673 0.000 0.000380 ENSG00000205011 AC073082.1<br />

6.92 -1.127 0.000 0.000390 ENSG00000042493 CAPG<br />

7.91 -0.553 0.000 0.000390 ENSG00000204138 PHACTR4<br />

9.42 -0.581 0.000 0.000400 ENSG00000143742 SRP9<br />

8.05 -1.162 0.000 0.000410 ENSG00000107249 GLIS3<br />

4.6 -0.65 0.000 0.000410 ENSG00000172238 ATOH1<br />

8.33 -1.572 0.000 0.000420 ENSG00000152767 FARP1<br />

4.7 -2.758 0.000 0.000420 ENSG00000163762 TM4SF18<br />

7.29 -1.321 0.000 0.000420 ENSG00000179242 CDH4<br />

4.32 -1.336 0.000 0.000420 ENSG00000188316 ENO4<br />

7.53 -0.977 0.000 0.000430 ENSG00000039560 RAI14<br />

8.64 -1.21 0.000 0.000430 ENSG00000128791 TWSG1<br />

7.73 -1.201 0.000 0.000430 ENSG00000130147 SH3BP4<br />

6.59 -1.229 0.000 0.000440 ENSG00000113721 PDGFRB<br />

7.62 -0.694 0.000 0.000440 ENSG00000150760 DOCK1<br />

7.79 -0.655 0.000 0.000450 ENSG00000148411 NACC2<br />

4.02 -1.254 0.000 0.000450 ENSG00000154760 SLFN13<br />

6.46 -1.117 0.000 0.000450 ENSG00000188153 COL4A5<br />

4.84 -1.145 0.000 0.000460 ENSG00000109339 MAPK10<br />

9.11 -2.027 0.000 0.000460 ENSG00000118523 CTGF<br />

7.55 -1.017 0.000 0.000460 ENSG00000137693 YAP1<br />

7.01 -0.558 0.000 0.000460 ENSG00000186174 BCL9L<br />

6.04 -1.369 0.000 0.000460 ENSG00000197565 COL4A6<br />

7.85 -0.791 0.000 0.000460 ENSG00000198856 OSTC<br />

3.62 -0.988 0.000 0.000470 ENSG00000155974 GRIP1<br />

6.5 -0.997 0.000 0.000470 ENSG00000171055 FEZ2<br />

6.12 -0.539 0.000 0.000470 ENSG00000171680 PLEKHG5<br />

10.02 -1.536 0.000 0.000490 ENSG00000041982 TNC<br />

7.6 -0.459 0.000 0.000490 ENSG00000166454 ATMIN<br />

6.43 -0.794 0.001 0.000500 ENSG00000197261 C6orf141<br />

8.15 -1.104 0.001 0.000510 ENSG00000119655 NPC2<br />

5.83 -1.663 0.001 0.000510 ENSG00000146373 RNF217<br />

3.77 -1.114 0.001 0.000510 ENSG00000150540 HNMT<br />

7.94 -0.855 0.001 0.000510 ENSG00000159842 ABR<br />

291


E. Exon Array Data Appendix<br />

Average expression across samples Fold-change p-value FDR Ensembl Gene ID Symbol<br />

7.84 -0.72 0.001 0.000510 ENSG00000214265 SNURF<br />

3.66 -1.512 0.001 0.000510 ENSG00000214919 AC104472.1<br />

5.04 -1.875 0.001 0.000520 ENSG00000102935 ZNF423<br />

6.22 -0.928 0.001 0.000530 ENSG00000106991 ENG<br />

7.3 -0.798 0.001 0.000530 ENSG00000119977 TCTN3<br />

8.03 -1.528 0.001 0.000530 ENSG00000120437 ACAT2<br />

6.41 -1.877 0.001 0.000530 ENSG00000138829 FBN2<br />

3.03 -0.791 0.001 0.000530 ENSG00000162614 NEXN<br />

6.94 -2.214 0.001 0.000540 ENSG00000121005 CRISPLD1<br />

6.77 -1.284 0.001 0.000540 ENSG00000164465 DCBLD1<br />

4.6 -1.496 0.001 0.000540 ENSG00000172671 ZFAND4<br />

5.97 -0.955 0.001 0.000550 ENSG00000162520 SYNC<br />

6.25 -0.89 0.001 0.000550 ENSG00000163485 ADORA1<br />

4.05 -1.925 0.001 0.000550 ENSG00000165983 PTER<br />

7.78 -1.38 0.001 0.000550 ENSG00000180537 RNF182<br />

7.99 -0.599 0.001 0.000560 ENSG00000166333 ILK<br />

5.29 -0.835 0.001 0.000570 ENSG00000139832 RAB20<br />

8.27 -1.102 0.001 0.000580 ENSG00000122026 RPL21<br />

6.95 -0.781 0.001 0.000580 ENSG00000167123 CERCAM<br />

5.84 -2.436 0.001 0.000580 ENSG00000205664 RP11-706O15.1<br />

7.91 -1.209 0.001 0.000590 ENSG00000122884 P4HA1<br />

4.18 -1.509 0.001 0.000590 ENSG00000172716 SLFN11<br />

8.52 -0.9 0.001 0.000600 ENSG00000149485 FADS1<br />

6.61 -0.601 0.001 0.000610 ENSG00000087903 RFX2<br />

5.67 -0.674 0.001 0.000610 ENSG00000095539 SEMA4G<br />

7.5 -0.85 0.001 0.000610 ENSG00000160584 SIK3<br />

7.03 -0.509 0.001 0.000610 ENSG00000166507 NDST2<br />

7.78 -0.812 0.001 0.000620 ENSG00000074181 NOTCH3<br />

6.24 -0.729 0.001 0.000630 ENSG00000157322 CLEC18A<br />

9.2 -1.574 0.001 0.000640 ENSG00000112186 CAP2<br />

7.07 -2.615 0.001 0.000650 ENSG00000087510 TFAP2C<br />

7.91 -0.832 0.001 0.000650 ENSG00000142173 COL6A2<br />

8.58 -0.86 0.001 0.000650 ENSG00000197965 MPZL1<br />

5.92 -0.73 0.001 0.000660 ENSG00000124831 LRRFIP1<br />

6.9 -1.51 0.001 0.000670 ENSG00000104783 KCNN4<br />

3.47 -0.881 0.001 0.000670 ENSG00000146038 DCDC2<br />

5.8 -0.887 0.001 0.000670 ENSG00000198768 APCDD1L<br />

6.88 -0.976 0.001 0.000680 ENSG00000148429 USP6NL<br />

8.6 -1.479 0.001 0.000680 ENSG00000169855 ROBO1<br />

6.76 -0.914 0.001 0.000680 ENSG00000181649 PHLDA2<br />

7.55 -0.953 0.001 0.000690 ENSG00000014216 CAPN1<br />

5.39 -0.948 0.001 0.000690 ENSG00000112183 RBM24<br />

7.14 -0.869 0.001 0.000690 ENSG00000118263 KLF7<br />

7.46 -0.671 0.001 0.000700 ENSG00000148498 PARD3<br />

5.7 -1.198 0.001 0.000710 ENSG00000135824 RGS8<br />

6.54 -1.323 0.001 0.000720 ENSG00000161958 FGF11<br />

6.12 -1.585 0.001 0.000720 ENSG00000172059 KLF11<br />

8.33 -0.669 0.001 0.000720 ENSG00000172081 MOB3A<br />

6.15 -0.95 0.001 0.000720 ENSG00000239887 C1orf226<br />

6.85 -1.301 0.001 0.000730 ENSG00000123096 SSPN<br />

6.8 -0.95 0.001 0.000740 ENSG00000196935 SRGAP1<br />

6.28 -0.714 0.001 0.000750 ENSG00000157335 CLEC18C<br />

6.9 -2.327 0.001 0.000760 ENSG00000146411 SLC2A12<br />

7.01 -1.299 0.001 0.000760 ENSG00000197093 GAL3ST4<br />

7.62 -0.757 0.001 0.000770 ENSG00000137216 TMEM63B<br />

5.99 -1.143 0.001 0.000770 ENSG00000144218 AFF3<br />

8.19 -1.003 0.001 0.000780 ENSG00000173457 PPP1R14B<br />

6.95 -1.347 0.001 0.000790 ENSG00000065534 MYLK<br />

9.56 -1.273 0.001 0.000790 ENSG00000124942 AHNAK<br />

8.75 -1.094 0.001 0.000790 ENSG00000160285 LSS<br />

6.52 -1.65 0.001 0.000790 ENSG00000172469 MANEA<br />

6.01 -0.921 0.001 0.000790 ENSG00000173715 C11orf80<br />

4.28 -1.027 0.001 0.000790 ENSG00000197140 ADAM32<br />

4.65 -1.509 0.001 0.000800 ENSG00000169946 ZFPM2<br />

7.97 -1.128 0.001 0.000800 ENSG00000183098 GPC6<br />

5.92 -1.632 0.001 0.000810 ENSG00000003436 TFPI<br />

7.2 -0.498 0.001 0.000810 ENSG00000070366 SMG6<br />

6.43 -0.515 0.001 0.000830 ENSG00000172346 CSDC2<br />

4.78 -0.836 0.001 0.000830 ENSG00000228120 AP001631.10<br />

8.58 -0.967 0.001 0.000840 ENSG00000145349 CAMK2D<br />

8.74 -0.995 0.001 0.000850 ENSG00000068793 CYFIP1<br />

7.38 -0.478 0.001 0.000850 ENSG00000197324 LRP10<br />

6.39 -0.685 0.001 0.000860 ENSG00000065665 SEC61A2<br />

6.27 -0.754 0.001 0.000860 ENSG00000140839 CLEC18B<br />

4.96 -1.366 0.001 0.000870 ENSG00000150630 VEGFC<br />

7.31 -0.864 0.001 0.000870 ENSG00000151929 BAG3<br />

292


E. Exon Array Data Appendix<br />

Average expression across samples Fold-change p-value FDR Ensembl Gene ID Symbol<br />

5.94 -1.001 0.001 0.000870 ENSG00000165406 Mar-08<br />

7.37 -0.827 0.001 0.000880 ENSG00000126773 PCNXL4<br />

8.75 -0.652 0.001 0.000890 ENSG00000138107 ACTR1A<br />

8.13 -0.645 0.001 0.000900 ENSG00000132142 ACACA<br />

2.65 -0.596 0.001 0.000900 ENSG00000151338 MIPOL1<br />

7.65 -2.89 0.001 0.000910 ENSG00000145623 OSMR<br />

4.78 -1.089 0.001 0.000930 ENSG00000164484 TMEM200A<br />

8.17 -0.961 0.001 0.000930 ENSG00000180776 ZDHHC20<br />

6.74 -1.118 0.001 0.000940 ENSG00000165512 ZNF22<br />

3.35 -0.987 0.001 0.000950 ENSG00000174792 C4orf26<br />

6.79 -1.011 0.001 0.000950 ENSG00000183580 FBXL7<br />

7.28 -0.97 0.001 0.000960 ENSG00000109536 FRG1<br />

5.53 -0.609 0.001 0.000960 ENSG00000123191 ATP7B<br />

8.79 -0.678 0.001 0.000960 ENSG00000152022 LIX1L<br />

6.78 -1.182 0.001 0.000960 ENSG00000164251 F2RL1<br />

7.8 -0.836 0.001 0.000970 ENSG00000147100 SLC16A2<br />

7.29 -0.674 0.001 0.000990 ENSG00000130643 CALY<br />

6.22 -0.585 0.001 0.000990 ENSG00000198546 ZNF511<br />

4.79 2.559 0.000 0.000000 ENSG00000005020 SKAP2<br />

5.52 1.043 0.000 0.000000 ENSG00000008056 SYN1<br />

8.25 3.134 0.000 0.000000 ENSG00000010278 CD9<br />

4.2 2.503 0.000 0.000000 ENSG00000064763 FAR2<br />

6.63 0.942 0.000 0.000000 ENSG00000085117 CD82<br />

4.32 2.793 0.000 0.000000 ENSG00000086300 SNX10<br />

4.71 2.29 0.000 0.000000 ENSG00000087303 NID2<br />

7.32 0.863 0.000 0.000000 ENSG00000105379 ETFB<br />

3.96 1.434 0.000 0.000000 ENSG00000105499 PLA2G4C<br />

3.11 1.41 0.000 0.000000 ENSG00000105889 STEAP1B<br />

5.57 1.454 0.000 0.000000 ENSG00000106789 CORO2A<br />

5.47 1.651 0.000 0.000000 ENSG00000114948 ADAM23<br />

5.72 0.898 0.000 0.000000 ENSG00000119431 HDHD3<br />

7.21 4.479 0.000 0.000000 ENSG00000124785 NRN1<br />

7.81 0.974 0.000 0.000000 ENSG00000128563 PRKRIP1<br />

5.96 1.277 0.000 0.000000 ENSG00000128709 HOXD9<br />

6.11 3.085 0.000 0.000000 ENSG00000128710 HOXD10<br />

5.82 1.922 0.000 0.000000 ENSG00000128713 HOXD11<br />

6.64 1.409 0.000 0.000000 ENSG00000130203 APOE<br />

5.62 4.15 0.000 0.000000 ENSG00000134853 PDGFRA<br />

3.79 1.54 0.000 0.000000 ENSG00000137727 ARHGAP20<br />

5.02 1.678 0.000 0.000000 ENSG00000140022 STON2<br />

5.67 2.034 0.000 0.000000 ENSG00000142494 SLC47A1<br />

7.69 1.182 0.000 0.000000 ENSG00000147883 CDKN2B<br />

4.8 2.142 0.000 0.000000 ENSG00000153029 MR1<br />

3.98 1.624 0.000 0.000000 ENSG00000154493 C10orf90<br />

7.05 1.693 0.000 0.000000 ENSG00000154537 FAM27C<br />

4.8 1.635 0.000 0.000000 ENSG00000156395 SORCS3<br />

5.16 1.341 0.000 0.000000 ENSG00000160179 ABCG1<br />

5.79 2.664 0.000 0.000000 ENSG00000163235 TGFA<br />

3.18 1.87 0.000 0.000000 ENSG00000164647 STEAP1<br />

4.75 1.82 0.000 0.000000 ENSG00000167216 KATNAL2<br />

5.26 2.322 0.000 0.000000 ENSG00000167306 MYO5B<br />

5.67 2.245 0.000 0.000000 ENSG00000168234 TTC39C<br />

5.37 1.902 0.000 0.000000 ENSG00000168779 SHOX2<br />

8.11 1.311 0.000 0.000000 ENSG00000169919 GUSB<br />

7.18 1.878 0.000 0.000000 ENSG00000170215 FAM27B<br />

5.26 1.276 0.000 0.000000 ENSG00000171729 TMEM51<br />

4.33 2.548 0.000 0.000000 ENSG00000173083 HPSE<br />

6.17 1.425 0.000 0.000000 ENSG00000173805 HAP1<br />

6.57 3.093 0.000 0.000000 ENSG00000175197 DDIT3<br />

6.62 3.734 0.000 0.000000 ENSG00000176165 FOXG1<br />

7.24 1.431 0.000 0.000000 ENSG00000178381 ZFAND2A<br />

3.53 1.663 0.000 0.000000 ENSG00000178538 CA8<br />

7.2 1.913 0.000 0.000000 ENSG00000182368 FAM27A<br />

4.39 2.726 0.000 0.000000 ENSG00000186960 C14orf23<br />

7.12 1.148 0.000 0.000000 ENSG00000187244 BCAM<br />

5.4 1.328 0.000 0.000000 ENSG00000187764 SEMA4D<br />

6.42 4.099 0.000 0.000000 ENSG00000189058 APOD<br />

3.76 1.495 0.000 0.000000 ENSG00000196954 CASP4<br />

5.08 1.747 0.000 0.000000 ENSG00000204634 TBC1D8<br />

5.43 1.793 0.000 0.000000 ENSG00000214575 CPEB1<br />

4.75 2.361 0.000 0.000000 ENSG00000223572 CKMT1A<br />

4.81 2.497 0.000 0.000000 ENSG00000237289 CKMT1B<br />

5.34 2.004 0.000 0.000000 ENSG00000258518 AC112502.1<br />

6.52 1.423 0.000 0.000010 ENSG00000003096 KLHL13<br />

5.29 2.135 0.000 0.000010 ENSG00000019549 SNAI2<br />

4.56 1.944 0.000 0.000010 ENSG00000056736 IL17RB<br />

293


E. Exon Array Data Appendix<br />

Average expression across samples Fold-change p-value FDR Ensembl Gene ID Symbol<br />

8.68 0.866 0.000 0.000010 ENSG00000073578 SDHA<br />

6.03 3.092 0.000 0.000010 ENSG00000089127 OAS1<br />

3.57 1.6 0.000 0.000010 ENSG00000092607 TBX15<br />

4.82 1.835 0.000 0.000010 ENSG00000102445 KIAA0226L<br />

4.8 1.261 0.000 0.000010 ENSG00000103723 AP3B2<br />

7.97 0.683 0.000 0.000010 ENSG00000106263 EIF3B<br />

7.77 1.071 0.000 0.000010 ENSG00000121083 DYNLL2<br />

6.83 1.603 0.000 0.000010 ENSG00000123146 CD97<br />

6.29 0.672 0.000 0.000010 ENSG00000125746 EML2<br />

7.19 0.883 0.000 0.000010 ENSG00000130305 NSUN5<br />

7.03 2.957 0.000 0.000010 ENSG00000146072 TNFRSF21<br />

4.7 1.327 0.000 0.000010 ENSG00000151117 TMEM86A<br />

4.81 1.072 0.000 0.000010 ENSG00000153790 C7orf31<br />

5.74 2.749 0.000 0.000010 ENSG00000166741 NNMT<br />

5.11 1.937 0.000 0.000010 ENSG00000186088 PION<br />

4.71 1.77 0.000 0.000010 ENSG00000187098 MITF<br />

3.72 1.906 0.000 0.000010 ENSG00000204397 CARD16<br />

7.31 1.009 0.000 0.000010 ENSG00000232098 AC012313.1<br />

6.94 1.111 0.000 0.000020 ENSG00000072071 LPHN1<br />

5.36 2.843 0.000 0.000020 ENSG00000089041 P2RX7<br />

7.79 0.858 0.000 0.000020 ENSG00000089057 SLC23A2<br />

5.99 1.358 0.000 0.000020 ENSG00000096433 ITPR3<br />

8.6 0.812 0.000 0.000020 ENSG00000101474 APMAP<br />

6.01 0.937 0.000 0.000020 ENSG00000103241 FOXF1<br />

7.4 0.773 0.000 0.000020 ENSG00000105518 TMEM205<br />

4.7 3.12 0.000 0.000020 ENSG00000106688 SLC1A1<br />

4.11 1.188 0.000 0.000020 ENSG00000107864 CPEB3<br />

6.82 1.511 0.000 0.000020 ENSG00000110841 PPFIBP1<br />

6.66 1.511 0.000 0.000020 ENSG00000117280 RAB7L1<br />

8.48 1.567 0.000 0.000020 ENSG00000125148 MT2A<br />

5.93 2.858 0.000 0.000020 ENSG00000125820 NKX2-2<br />

6.31 2.012 0.000 0.000020 ENSG00000134202 GSTM3<br />

3.83 1.725 0.000 0.000020 ENSG00000134716 CYP2J2<br />

4.06 1.958 0.000 0.000020 ENSG00000136237 RAPGEF5<br />

7.69 2.759 0.000 0.000020 ENSG00000136842 TMOD1<br />

6.32 0.922 0.000 0.000020 ENSG00000141026 MED9<br />

6.37 1.279 0.000 0.000020 ENSG00000147889 CDKN2A<br />

5.91 1.256 0.000 0.000020 ENSG00000158856 EPB49<br />

7.98 1.855 0.000 0.000020 ENSG00000162873 KLHDC8A<br />

6.97 0.685 0.000 0.000020 ENSG00000167106 FAM102A<br />

6.31 3.628 0.000 0.000020 ENSG00000170689 HOXB9<br />

5.8 1.171 0.000 0.000020 ENSG00000172183 ISG20<br />

4.23 1.273 0.000 0.000020 ENSG00000172345 STARD5<br />

7.02 1.316 0.000 0.000020 ENSG00000176490 DIRAS1<br />

6.55 0.867 0.000 0.000020 ENSG00000177556 ATOX1<br />

5.87 2.507 0.000 0.000020 ENSG00000196361 ELAVL3<br />

5.22 0.873 0.000 0.000020 ENSG00000196476 C20orf96<br />

5.9 1.137 0.000 0.000020 ENSG00000196683 TOMM7<br />

8.22 1.421 0.000 0.000020 ENSG00000205362 MT1A<br />

4.9 1.465 0.000 0.000030 ENSG00000018869 ZNF582<br />

6.66 0.818 0.000 0.000030 ENSG00000101181 GTPBP5<br />

5.69 1.972 0.000 0.000030 ENSG00000113296 THBS4<br />

5.74 1.581 0.000 0.000030 ENSG00000119922 IFIT2<br />

6.83 0.596 0.000 0.000030 ENSG00000130813 C19orf66<br />

6.99 1.327 0.000 0.000030 ENSG00000145216 FIP1L1<br />

5.26 1.11 0.000 0.000030 ENSG00000149927 DOC2A<br />

4.84 1.528 0.000 0.000030 ENSG00000151090 THRB<br />

7.77 1.094 0.000 0.000030 ENSG00000157259 GATAD1<br />

3.99 0.896 0.000 0.000030 ENSG00000164398 ACSL6<br />

4.64 2.353 0.000 0.000030 ENSG00000165443 PHYHIPL<br />

5.03 1.62 0.000 0.000030 ENSG00000170941 AC135352.1<br />

4.99 2.142 0.000 0.000030 ENSG00000182326 C1S<br />

2.77 0.976 0.000 0.000030 ENSG00000188906 LRRK2<br />

7.76 1.085 0.000 0.000030 ENSG00000198874 TYW1<br />

4.54 1.754 0.000 0.000040 ENSG00000073910 FRY<br />

6.26 2.104 0.000 0.000040 ENSG00000104419 NDRG1<br />

7.3 1.434 0.000 0.000040 ENSG00000130066 SAT1<br />

4.96 1.985 0.000 0.000040 ENSG00000132436 FIGNL1<br />

3.52 0.689 0.000 0.000040 ENSG00000143226 FCGR2A<br />

4.62 1.31 0.000 0.000040 ENSG00000143320 CRABP2<br />

5.93 4.882 0.000 0.000040 ENSG00000156298 TSPAN7<br />

5.53 2.923 0.000 0.000040 ENSG00000160862 AZGP1<br />

6.28 2.132 0.000 0.000040 ENSG00000163638 ADAMTS9<br />

4.94 1.336 0.000 0.000040 ENSG00000189164 ZNF527<br />

6.19 1.071 0.000 0.000050 ENSG00000105649 RAB3A<br />

5.66 1.003 0.000 0.000050 ENSG00000136002 ARHGEF4<br />

294


E. Exon Array Data Appendix<br />

Average expression across samples Fold-change p-value FDR Ensembl Gene ID Symbol<br />

6.21 1.475 0.000 0.000050 ENSG00000136243 NUPL2<br />

5.64 0.992 0.000 0.000050 ENSG00000151093 OXSM<br />

4.85 1.082 0.000 0.000050 ENSG00000155629 PIK3AP1<br />

7.07 0.716 0.000 0.000050 ENSG00000157778 PSMG3<br />

8.48 1.368 0.000 0.000050 ENSG00000162545 CAMK2N1<br />

4.72 1.871 0.000 0.000050 ENSG00000164741 DLC1<br />

5.24 1.051 0.000 0.000060 ENSG00000058866 DGKG<br />

5.61 1.415 0.000 0.000060 ENSG00000073464 CLCN4<br />

9.27 0.763 0.000 0.000060 ENSG00000086232 EIF2AK1<br />

3.97 2.144 0.000 0.000060 ENSG00000108176 DNAJC12<br />

7.3 1.402 0.000 0.000060 ENSG00000112852 PCDHB2<br />

4.68 1.736 0.000 0.000060 ENSG00000137393 RNF144B<br />

6.16 1.387 0.000 0.000070 ENSG00000116574 RHOU<br />

8.42 1.11 0.000 0.000070 ENSG00000124226 RNF114<br />

5.72 0.79 0.000 0.000070 ENSG00000129282 MRM1<br />

6 1.143 0.000 0.000070 ENSG00000159228 CBR1<br />

7.25 1.234 0.000 0.000070 ENSG00000166770 AC004696.2<br />

6.42 0.938 0.000 0.000070 ENSG00000172840 PDP2<br />

6.38 0.76 0.000 0.000070 ENSG00000174672 BRSK2<br />

7.22 0.934 0.000 0.000070 ENSG00000179304 FAM156B<br />

6.01 2.099 0.000 0.000070 ENSG00000182217 HIST2H4B<br />

6.01 2.099 0.000 0.000070 ENSG00000183941 HIST2H4A<br />

7.93 2.197 0.000 0.000070 ENSG00000196532 HIST1H3C<br />

6.17 0.703 0.000 0.000070 ENSG00000196741 CXorf24<br />

4.98 1.47 0.000 0.000070 ENSG00000214106 AC093726.6<br />

5.49 0.968 0.000 0.000080 ENSG00000036672 USP2<br />

5.53 1.341 0.000 0.000080 ENSG00000088035 ALG6<br />

5.15 1.343 0.000 0.000080 ENSG00000156500 FAM122C<br />

5.74 1.177 0.000 0.000080 ENSG00000163884 KLF15<br />

6.04 1.258 0.000 0.000080 ENSG00000166402 TUB<br />

7.17 0.939 0.000 0.000080 ENSG00000182646 FAM156A<br />

6.64 4.408 0.000 0.000080 ENSG00000250349 RP5-972B16.2<br />

6.97 0.713 0.000 0.000090 ENSG00000101189 C20orf20<br />

3.92 1.212 0.000 0.000090 ENSG00000119514 GALNT12<br />

3.71 0.824 0.000 0.000090 ENSG00000125804 FAM182A<br />

7.55 0.989 0.000 0.000090 ENSG00000163156 SCNM1<br />

3.42 1.18 0.000 0.000090 ENSG00000181016 C7orf53<br />

6.35 0.891 0.000 0.000090 ENSG00000184205 TSPYL2<br />

4.35 1.373 0.000 0.000090 ENSG00000189283 FHIT<br />

5.51 1.475 0.000 0.000090 ENSG00000198270 TMEM116<br />

7.84 1.309 0.000 0.000100 ENSG00000101224 CDC25B<br />

5.52 0.686 0.000 0.000100 ENSG00000126950 TMEM35<br />

4.4 2.366 0.000 0.000100 ENSG00000148735 PLEKHS1<br />

5.04 1.951 0.000 0.000100 ENSG00000163644 PPM1K<br />

7.83 1.683 0.000 0.000100 ENSG00000172115 CYCS<br />

5.24 1.512 0.000 0.000100 ENSG00000204807 FAM27E2<br />

6.23 1.594 0.000 0.000100 ENSG00000251369 AC003682.1<br />

6.76 1.991 0.000 0.000100 ENSG00000255986 MT1JP<br />

5.41 1.122 0.000 0.000110 ENSG00000131242 RAB11FIP4<br />

7.78 0.733 0.000 0.000110 ENSG00000146676 PURB<br />

6.69 2.507 0.000 0.000110 ENSG00000149294 NCAM1<br />

4.24 0.751 0.000 0.000110 ENSG00000175697 GPR156<br />

4.15 0.854 0.000 0.000110 ENSG00000214290 C11orf93<br />

4.27 1.558 0.000 0.000120 ENSG00000082482 KCNK2<br />

4.89 0.887 0.000 0.000120 ENSG00000100307 CBX7<br />

2.71 0.772 0.000 0.000120 ENSG00000146856 AGBL3<br />

5.69 0.606 0.000 0.000120 ENSG00000169136 ATF5<br />

4.36 0.938 0.000 0.000120 ENSG00000175170 FAM182B<br />

6.37 0.774 0.000 0.000120 ENSG00000255098 RP11-481A20.11<br />

3.52 1.432 0.000 0.000130 ENSG00000003147 ICA1<br />

4.87 1.498 0.000 0.000130 ENSG00000007944 MYLIP<br />

5.81 0.772 0.000 0.000130 ENSG00000101104 PABPC1L<br />

5 1.218 0.000 0.000130 ENSG00000102362 SYTL4<br />

6.13 1.354 0.000 0.000130 ENSG00000158186 MRAS<br />

4.22 0.902 0.000 0.000130 ENSG00000176273 SLC35G1<br />

6.5 1.029 0.000 0.000130 ENSG00000256073 C21orf119<br />

5.73 2.686 0.000 0.000140 ENSG00000117602 RCAN3<br />

7.14 1.232 0.000 0.000140 ENSG00000141753 IGFBP4<br />

5.52 0.559 0.000 0.000140 ENSG00000149599 DUSP15<br />

4.73 1.741 0.000 0.000140 ENSG00000154734 ADAMTS1<br />

3.69 1.648 0.000 0.000150 ENSG00000064225 ST3GAL6<br />

5.21 1.771 0.000 0.000150 ENSG00000106004 HOXA5<br />

6.07 1.193 0.000 0.000150 ENSG00000111364 DDX55<br />

6.02 1.5 0.000 0.000150 ENSG00000120318 ARAP3<br />

5.89 0.804 0.000 0.000150 ENSG00000178809 TRIM73<br />

7.59 0.945 0.000 0.000150 ENSG00000184371 CSF1<br />

295


E. Exon Array Data Appendix<br />

Average expression across samples Fold-change p-value FDR Ensembl Gene ID Symbol<br />

6.4 0.769 0.000 0.000150 ENSG00000184857 TMEM186<br />

5.64 1.271 0.000 0.000150 ENSG00000198814 GK<br />

4.72 1.253 0.000 0.000150 ENSG00000213903 LTB4R<br />

5.17 1.876 0.000 0.000160 ENSG00000008277 ADAM22<br />

3.51 1.09 0.000 0.000160 ENSG00000118777 ABCG2<br />

5.85 1.024 0.000 0.000160 ENSG00000164048 ZNF589<br />

6.16 0.956 0.000 0.000160 ENSG00000175175 PPM1E<br />

6.99 1.096 0.000 0.000160 ENSG00000232838 PET117<br />

6.64 0.794 0.000 0.000170 ENSG00000102934 PLLP<br />

6.78 0.622 0.000 0.000170 ENSG00000141582 CBX4<br />

8.37 0.573 0.000 0.000170 ENSG00000143543 JTB<br />

3.96 1.576 0.000 0.000170 ENSG00000158315 RHBDL2<br />

6.45 1.095 0.000 0.000170 ENSG00000204524 ZNF805<br />

6.43 1.7 0.000 0.000180 ENSG00000006652 IFRD1<br />

5.02 1.218 0.000 0.000180 ENSG00000092068 SLC7A8<br />

4.53 2.247 0.000 0.000180 ENSG00000152527 PLEKHH2<br />

6.11 1.012 0.000 0.000180 ENSG00000164620 RELL2<br />

5.74 1.411 0.000 0.000180 ENSG00000165795 NDRG2<br />

6.38 2.236 0.000 0.000190 ENSG00000105409 ATP1A3<br />

8.02 1.715 0.000 0.000190 ENSG00000106537 TSPAN13<br />

6.58 1.894 0.000 0.000190 ENSG00000125144 MT1G<br />

5.6 1.454 0.000 0.000190 ENSG00000164684 ZNF704<br />

6.47 0.985 0.000 0.000200 ENSG00000065717 TLE2<br />

3.93 1.031 0.000 0.000200 ENSG00000122862 SRGN<br />

4.01 1.439 0.000 0.000200 ENSG00000137203 TFAP2A<br />

6.21 1.126 0.000 0.000200 ENSG00000178878 APOLD1<br />

5.99 0.934 0.000 0.000200 ENSG00000180953 ST20<br />

8.5 1.354 0.000 0.000200 ENSG00000198743 SLC5A3<br />

5.6 0.559 0.000 0.000200 ENSG00000251357 AP000350.10<br />

7.33 0.633 0.000 0.000210 ENSG00000130733 YIPF2<br />

5.89 0.896 0.000 0.000210 ENSG00000142528 ZNF473<br />

5.73 1.626 0.000 0.000210 ENSG00000143162 CREG1<br />

6.19 0.828 0.000 0.000210 ENSG00000146909 NOM1<br />

5.34 0.95 0.000 0.000210 ENSG00000149150 SLC43A1<br />

3.29 0.995 0.000 0.000210 ENSG00000165923 AGBL2<br />

6.6 0.893 0.000 0.000210 ENSG00000175898 S1PR2<br />

4.07 0.944 0.000 0.000210 ENSG00000188732 FAM221A<br />

7.44 1.401 0.000 0.000210 ENSG00000231997 FAM27D1<br />

5.25 0.581 0.000 0.000220 ENSG00000126583 PRKCG<br />

6.21 1.077 0.000 0.000220 ENSG00000173875 ZNF791<br />

3.79 1.662 0.000 0.000230 ENSG00000071073 MGAT4A<br />

6.82 0.743 0.000 0.000230 ENSG00000106638 TBL2<br />

7.47 1.573 0.000 0.000230 ENSG00000189060 H1F0<br />

6.2 0.673 0.000 0.000240 ENSG00000106404 CLDN15<br />

6.5 0.814 0.000 0.000240 ENSG00000106479 ZNF862<br />

5.81 0.871 0.000 0.000240 ENSG00000106608 URGCP<br />

5.85 0.819 0.000 0.000240 ENSG00000108852 MPP2<br />

6.65 2.284 0.000 0.000240 ENSG00000154096 THY1<br />

9.19 1.685 0.000 0.000240 ENSG00000166986 MARS<br />

6.7 2.826 0.000 0.000240 ENSG00000175445 LPL<br />

7.67 0.515 0.000 0.000240 ENSG00000184787 UBE2G2<br />

5.33 1.325 0.000 0.000240 ENSG00000237198 FAM27E1<br />

3.64 1.397 0.000 0.000240 ENSG00000250305 KIAA1456<br />

7.42 0.921 0.000 0.000250 ENSG00000130254 SAFB2<br />

4.71 0.878 0.000 0.000250 ENSG00000152049 KCNE4<br />

4.05 1.314 0.000 0.000250 ENSG00000175906 ARL4D<br />

3.91 0.779 0.000 0.000250 ENSG00000204086 RPA4<br />

5.35 2.281 0.000 0.000250 ENSG00000205364 MT1M<br />

4.05 1.053 0.000 0.000260 ENSG00000076554 TPD52<br />

8.37 1.379 0.000 0.000260 ENSG00000085662 AKR1B1<br />

8.61 0.814 0.000 0.000260 ENSG00000104687 GSR<br />

6.29 1.082 0.000 0.000260 ENSG00000105926 MPP6<br />

9.84 0.823 0.000 0.000260 ENSG00000146701 MDH2<br />

5.59 0.957 0.000 0.000260 ENSG00000168301 KCTD6<br />

5.9 0.865 0.000 0.000260 ENSG00000174939 ASPHD1<br />

5.92 0.645 0.000 0.000260 ENSG00000204536 CCHCR1<br />

6.69 0.769 0.000 0.000270 ENSG00000106785 TRIM14<br />

6.99 0.808 0.000 0.000270 ENSG00000125875 TBC1D20<br />

5.82 1.073 0.000 0.000270 ENSG00000164742 ADCY1<br />

4.02 1.271 0.000 0.000270 ENSG00000196932 TMEM26<br />

4.66 0.86 0.000 0.000270 ENSG00000197933 ZNF823<br />

6 1.557 0.000 0.000270 ENSG00000198053 SIRPA<br />

5.43 0.934 0.000 0.000270 ENSG00000215897 ZBTB8B<br />

4.79 0.711 0.000 0.000280 ENSG00000124613 ZNF391<br />

6.98 1.247 0.000 0.000280 ENSG00000126368 NR1D1<br />

4.95 0.642 0.000 0.000290 ENSG00000125510 OPRL1<br />

296


E. Exon Array Data Appendix<br />

Average expression across samples Fold-change p-value FDR Ensembl Gene ID Symbol<br />

4.21 1.238 0.000 0.000290 ENSG00000166387 PPFIBP2<br />

5.58 0.709 0.000 0.000290 ENSG00000186951 PPARA<br />

7.3 1.284 0.000 0.000290 ENSG00000187372 PCDHB13<br />

3.61 0.734 0.000 0.000290 ENSG00000196167 C11orf92<br />

5.69 1.412 0.000 0.000300 ENSG00000068650 ATP11A<br />

7.51 0.631 0.000 0.000300 ENSG00000106070 GRB10<br />

5.19 0.553 0.000 0.000300 ENSG00000170379 FAM115C<br />

5.03 0.896 0.000 0.000300 ENSG00000257755 orphan<br />

4.33 1.526 0.000 0.000310 ENSG00000104324 CPQ<br />

5.18 0.844 0.000 0.000310 ENSG00000107957 SH3PXD2A<br />

6.49 0.752 0.000 0.000310 ENSG00000114859 CLCN2<br />

7.06 1.16 0.000 0.000310 ENSG00000135457 TFCP2<br />

5.35 1.852 0.000 0.000310 ENSG00000153714 LURAP1L<br />

10.2 0.876 0.000 0.000310 ENSG00000171867 PRNP<br />

3.1 1.481 0.000 0.000320 ENSG00000106560 GIMAP2<br />

7.45 1.354 0.000 0.000320 ENSG00000150893 FREM2<br />

5.65 0.632 0.000 0.000320 ENSG00000163132 MSX1<br />

8 0.759 0.000 0.000320 ENSG00000178741 COX5A<br />

5.45 1.098 0.000 0.000320 ENSG00000211445 GPX3<br />

6.16 0.599 0.000 0.000330 ENSG00000083812 ZNF324<br />

7.47 0.735 0.000 0.000330 ENSG00000145882 PCYOX1L<br />

5.9 0.732 0.000 0.000330 ENSG00000155428 TRIM74<br />

6.82 0.92 0.000 0.000340 ENSG00000106415 GLCCI1<br />

5.56 0.658 0.000 0.000340 ENSG00000116039 ATP6V1B1<br />

4.71 0.686 0.000 0.000340 ENSG00000132801 ZSWIM3<br />

6.32 0.688 0.000 0.000340 ENSG00000134086 VHL<br />

7.86 0.575 0.000 0.000350 ENSG00000113269 RNF130<br />

5.23 1.823 0.000 0.000350 ENSG00000131015 ULBP2<br />

4.27 1.325 0.000 0.000350 ENSG00000197757 HOXC6<br />

7.01 1.393 0.000 0.000350 ENSG00000231360 AL592284.2<br />

3.41 0.946 0.000 0.000360 ENSG00000025423 HSD17B6<br />

5.67 1.773 0.000 0.000360 ENSG00000165118 C9orf64<br />

4.77 0.903 0.000 0.000360 ENSG00000175911 AC127496.1<br />

4.45 1.145 0.000 0.000360 ENSG00000232956 SNHG15<br />

5.73 1.061 0.000 0.000370 ENSG00000135245 HILPDA<br />

6.24 1.297 0.000 0.000370 ENSG00000135472 FAIM2<br />

6.44 0.912 0.000 0.000370 ENSG00000154930 ACSS1<br />

7.65 1.07 0.000 0.000370 ENSG00000168303 MPLKIP<br />

5.48 0.809 0.000 0.000370 ENSG00000205358 MT1H<br />

4.75 1.489 0.000 0.000380 ENSG00000112379 KIAA1244<br />

4.21 1.081 0.000 0.000380 ENSG00000119547 ONECUT2<br />

3.38 0.996 0.000 0.000380 ENSG00000144290 SLC4A10<br />

5.43 0.988 0.000 0.000380 ENSG00000146833 TRIM4<br />

7.01 1.023 0.000 0.000380 ENSG00000173276 ZNF295<br />

8.3 0.683 0.000 0.000380 ENSG00000175193 PARL<br />

4.84 0.881 0.000 0.000390 ENSG00000204856 FAM216A<br />

6.52 1.323 0.000 0.000400 ENSG00000107829 FBXW4<br />

6.84 0.91 0.000 0.000400 ENSG00000124228 DDX27<br />

7.24 0.665 0.000 0.000410 ENSG00000101407 TTI1<br />

6.79 0.747 0.000 0.000410 ENSG00000106245 BUD31<br />

6.3 0.592 0.000 0.000410 ENSG00000142700 DMRTA2<br />

6.5 0.915 0.000 0.000410 ENSG00000189046 ALKBH2<br />

5.51 2.199 0.000 0.000410 ENSG00000215247<br />

5.9 0.698 0.000 0.000420 ENSG00000006194 ZNF263<br />

7.11 1.686 0.000 0.000420 ENSG00000120328 PCDHB12<br />

7.42 0.68 0.000 0.000420 ENSG00000136213 CHST12<br />

5.42 1.536 0.000 0.000420 ENSG00000151364 KCTD14<br />

7.23 0.928 0.000 0.000420 ENSG00000198171 DDRGK1<br />

6.44 0.8 0.000 0.000430 ENSG00000131368 MRPS25<br />

8.34 0.747 0.000 0.000440 ENSG00000111678 C12orf57<br />

6.57 0.838 0.000 0.000440 ENSG00000165661 QSOX2<br />

5.88 1.608 0.000 0.000440 ENSG00000186868 MAPT<br />

5.39 0.894 0.000 0.000440 ENSG00000253293 HOXA10<br />

7.35 0.627 0.000 0.000450 ENSG00000156928 MALSU1<br />

5.28 1.124 0.000 0.000450 ENSG00000172006 ZNF554<br />

4.69 0.646 0.000 0.000450 ENSG00000177335 C8orf31<br />

5.68 1.476 0.000 0.000450 ENSG00000206535 LNP1<br />

6.92 0.94 0.000 0.000460 ENSG00000197608 ZNF841<br />

3.25 1.454 0.000 0.000470 ENSG00000168811 IL12A<br />

4.52 0.897 0.000 0.000470 ENSG00000176593 CTD-2368P22.1<br />

3.63 1.342 0.000 0.000480 ENSG00000113389 NPR3<br />

3.35 1.071 0.000 0.000480 ENSG00000176907 C8orf4<br />

5.37 0.494 0.000 0.000480 ENSG00000177380 PPFIA3<br />

7.44 0.526 0.000 0.000480 ENSG00000229212 RP11-561C5.4<br />

4.52 1.355 0.000 0.000490 ENSG00000120915 EPHX2<br />

4.77 1.002 0.000 0.000490 ENSG00000125388 GRK4<br />

297


E. Exon Array Data Appendix<br />

Average expression across samples Fold-change p-value FDR Ensembl Gene ID Symbol<br />

6.05 0.737 0.001 0.000500 ENSG00000103024 NME3<br />

7.42 0.557 0.001 0.000500 ENSG00000105583 WDR83OS<br />

4.38 0.804 0.001 0.000500 ENSG00000133401 PDZD2<br />

6.69 0.801 0.001 0.000500 ENSG00000196652 ZKSCAN5<br />

7.71 1.671 0.001 0.000510 ENSG00000070669 ASNS<br />

6.84 0.567 0.001 0.000510 ENSG00000096070 BRPF3<br />

5.69 1.106 0.001 0.000510 ENSG00000101187 SLCO4A1<br />

5.48 0.832 0.001 0.000510 ENSG00000120158 RCL1<br />

9.84 0.767 0.001 0.000510 ENSG00000163399 ATP1A1<br />

5.71 1.081 0.001 0.000510 ENSG00000196981 WDR5B<br />

4.19 1.925 0.001 0.000520 ENSG00000136514 RTP4<br />

6.56 2.056 0.001 0.000520 ENSG00000156011 PSD3<br />

4.5 0.937 0.001 0.000520 ENSG00000159882 ZNF230<br />

5.22 0.836 0.001 0.000520 ENSG00000163362 C1orf106<br />

5.09 2.391 0.001 0.000530 ENSG00000106511 MEOX2<br />

5.05 1.189 0.001 0.000530 ENSG00000128604 IRF5<br />

6.86 1.183 0.001 0.000530 ENSG00000163938 GNL3<br />

6.95 0.971 0.001 0.000540 ENSG00000127952 STYXL1<br />

7.12 1.905 0.001 0.000540 ENSG00000164649 CDCA7L<br />

9.33 0.772 0.001 0.000540 ENSG00000187145 MRPS21<br />

7.6 0.708 0.001 0.000550 ENSG00000068305 MEF2A<br />

5.5 0.907 0.001 0.000550 ENSG00000124772 CPNE5<br />

6.17 0.721 0.001 0.000550 ENSG00000129347 KRI1<br />

4.03 0.802 0.001 0.000550 ENSG00000196482 ESRRG<br />

5.88 0.673 0.001 0.000560 ENSG00000123358 NR4A1<br />

4.08 0.7 0.001 0.000560 ENSG00000203815 AL358813.2<br />

4.84 0.817 0.001 0.000570 ENSG00000110031 LPXN<br />

6.18 0.467 0.001 0.000580 ENSG00000104731 KLHDC4<br />

5.1 0.815 0.001 0.000580 ENSG00000144115 THNSL2<br />

5 1.87 0.001 0.000590 ENSG00000104611 SH2D4A<br />

5.53 0.752 0.001 0.000590 ENSG00000117266 CDK18<br />

7.58 1.016 0.001 0.000590 ENSG00000171223 JUNB<br />

6.64 0.746 0.001 0.000600 ENSG00000068400 GRIPAP1<br />

5.05 0.96 0.001 0.000600 ENSG00000184208 C22orf46<br />

8 0.613 0.001 0.000610 ENSG00000130717 UCK1<br />

3.53 1.117 0.001 0.000610 ENSG00000135678 CPM<br />

6.86 1.135 0.001 0.000610 ENSG00000257921 RP11-571M6.15<br />

6.36 0.507 0.001 0.000610 ENSG00000258720<br />

3.41 0.725 0.001 0.000620 ENSG00000140009 ESR2<br />

5.09 1.82 0.001 0.000620 ENSG00000144847 IGSF11<br />

3.89 2.44 0.001 0.000620 ENSG00000157214 STEAP2<br />

7.27 0.911 0.001 0.000620 ENSG00000174243 DDX23<br />

5.36 0.76 0.001 0.000620 ENSG00000213983 AP1G2<br />

4.77 0.842 0.001 0.000630 ENSG00000095380 NANS<br />

4.07 2.206 0.001 0.000630 ENSG00000152078 TMEM56<br />

5.1 0.773 0.001 0.000640 ENSG00000188818 ZDHHC11<br />

6.82 1.812 0.001 0.000650 ENSG00000120324 PCDHB10<br />

5.77 1.023 0.001 0.000650 ENSG00000122877 EGR2<br />

6.72 1.068 0.001 0.000650 ENSG00000203814 HIST2H2BF<br />

4.4 1.535 0.001 0.000660 ENSG00000111846 GCNT2<br />

4.62 1.74 0.001 0.000660 ENSG00000174007 CEP19<br />

5.05 1.321 0.001 0.000660 ENSG00000180818 HOXC10<br />

8.34 0.553 0.001 0.000660 ENSG00000214331 RP11-252A24.2<br />

4.6 1.204 0.001 0.000670 ENSG00000185015 CA13<br />

6.4 0.578 0.001 0.000680 ENSG00000148290 SURF1<br />

6.12 1.141 0.001 0.000680 ENSG00000214357 NEURL1B<br />

5.77 3.086 0.001 0.000690 ENSG00000154529 CNTNAP3B<br />

6.46 0.53 0.001 0.000690 ENSG00000160299 PCNT<br />

7.31 0.729 0.001 0.000700 ENSG00000087250 MT3<br />

6.03 2.89 0.001 0.000700 ENSG00000106714 CNTNAP3<br />

6.23 0.712 0.001 0.000700 ENSG00000181191 PJA1<br />

8.21 0.906 0.001 0.000710 ENSG00000101210 EEF1A2<br />

5.14 0.814 0.001 0.000710 ENSG00000160207 HSF2BP<br />

5.97 0.818 0.001 0.000710 ENSG00000160908 ZNF394<br />

5.69 0.548 0.001 0.000710 ENSG00000165655 ZNF503<br />

5.24 2.121 0.001 0.000710 ENSG00000174607 UGT8<br />

7.63 0.754 0.001 0.000720 ENSG00000104980 TIMM44<br />

4.83 0.918 0.001 0.000720 ENSG00000162757 C1orf74<br />

5.19 0.796 0.001 0.000730 ENSG00000062282 DGAT2<br />

6.04 1.083 0.001 0.000730 ENSG00000064300 NGFR<br />

3.4 1.382 0.001 0.000730 ENSG00000165071 TMEM71<br />

3.53 1.144 0.001 0.000730 ENSG00000227471 AKR1B15<br />

6.81 1.233 0.001 0.000740 ENSG00000111276 CDKN1B<br />

6.24 0.731 0.001 0.000740 ENSG00000172366 FAM195A<br />

6.03 1.004 0.001 0.000740 ENSG00000188283 ZNF383<br />

5.05 0.755 0.001 0.000750 ENSG00000111199 TRPV4<br />

298


E. Exon Array Data Appendix<br />

Average expression across samples Fold-change p-value FDR Ensembl Gene ID Symbol<br />

7.26 1.393 0.001 0.000750 ENSG00000116991 SIPA1L2<br />

8.23 0.568 0.001 0.000780 ENSG00000196367 TRRAP<br />

6.49 1.202 0.001 0.000780 ENSG00000198729 PPP1R14C<br />

4.34 0.843 0.001 0.000790 ENSG00000135127 CCDC64<br />

6.54 1.345 0.001 0.000800 ENSG00000106689 LHX2<br />

4.92 0.897 0.001 0.000800 ENSG00000221994 ZNF630<br />

8.31 0.722 0.001 0.000810 ENSG00000101365 IDH3B<br />

7.15 0.669 0.001 0.000810 ENSG00000114767 RRP9<br />

6.95 0.503 0.001 0.000810 ENSG00000198258 UBL5<br />

7.8 0.771 0.001 0.000820 ENSG00000078043 PIAS2<br />

7.41 0.87 0.001 0.000820 ENSG00000111266 DUSP16<br />

6.56 0.92 0.001 0.000820 ENSG00000112715 VEGFA<br />

6.95 1.222 0.001 0.000830 ENSG00000123080 CDKN2C<br />

5.81 0.756 0.001 0.000830 ENSG00000130158 DOCK6<br />

5 0.858 0.001 0.000840 ENSG00000125352 RNF113A<br />

4.69 2.173 0.001 0.000840 ENSG00000138642 HERC6<br />

4.04 2.745 0.001 0.000860 ENSG00000138646 HERC5<br />

6.46 0.684 0.001 0.000860 ENSG00000197037 ZNF498<br />

8.15 0.797 0.001 0.000870 ENSG00000071462 WBSCR22<br />

5.13 1.572 0.001 0.000870 ENSG00000133321 RARRES3<br />

7.79 0.901 0.001 0.000870 ENSG00000160633 SAFB<br />

7.56 0.741 0.001 0.000870 ENSG00000164933 SLC25A32<br />

7.65 0.65 0.001 0.000880 ENSG00000125844 RRBP1<br />

7.59 0.595 0.001 0.000880 ENSG00000164896 FASTK<br />

4.47 1.076 0.001 0.000880 ENSG00000197857 ZNF44<br />

7.31 1.007 0.001 0.000890 ENSG00000077348 EXOSC5<br />

7.23 0.664 0.001 0.000900 ENSG00000101040 ZMYND8<br />

3.75 1.724 0.001 0.000900 ENSG00000137752 CASP1<br />

4.91 0.534 0.001 0.000900 ENSG00000143578 CREB3L4<br />

6.86 1.846 0.001 0.000900 ENSG00000154856 APCDD1<br />

4.11 0.919 0.001 0.000910 ENSG00000105708 ZNF14<br />

4.74 1.522 0.001 0.000910 ENSG00000116761 CTH<br />

7.33 0.778 0.001 0.000910 ENSG00000196365 LONP1<br />

7.82 1.045 0.001 0.000920 ENSG00000013374 NUB1<br />

6.08 0.799 0.001 0.000920 ENSG00000077713 SLC25A43<br />

3.54 0.864 0.001 0.000930 ENSG00000157224 CLDN12<br />

7.3 0.662 0.001 0.000930 ENSG00000160200 CBS<br />

5.12 3.1 0.001 0.000930 ENSG00000227921 AL353791.1<br />

4.02 1.202 0.001 0.000940 ENSG00000152433 ZNF547<br />

6.71 0.572 0.001 0.000950 ENSG00000105559 PLEKHA4<br />

6.45 1.93 0.001 0.000960 ENSG00000113205 PCDHB3<br />

3.5 0.926 0.001 0.000970 ENSG00000136167 LCP1<br />

2.97 0.557 0.001 0.000970 ENSG00000136839 OR13C9<br />

3.02 0.567 0.001 0.000980 ENSG00000130783 CCDC62<br />

4.96 1.806 0.001 0.000980 ENSG00000132498 ANKRD20A3<br />

7.62 0.717 0.001 0.000980 ENSG00000146834 MEPCE<br />

7.66 2.08 0.001 0.000980 ENSG00000196963 PCDHB16<br />

4.6 1.779 0.001 0.000990 ENSG00000110436 SLC1A2<br />

8.16 0.75 0.001 0.000990 ENSG00000137817 PARP6<br />

7.33 1.172 0.001 0.000990 ENSG00000141576 RNF157<br />

4.53 2.338 0.001 0.001000 ENSG00000018236 CNTN1<br />

5.8 2.56 0.001 0.001000 ENSG00000100095 SEZ6L<br />

7.36 0.859 0.001 0.001000 ENSG00000126947 ARMCX1<br />

3.88 1.307 0.001 0.001000 ENSG00000138741 TRPC3<br />

5.29 0.642 0.001 0.001000 ENSG00000163013 FBXO41<br />

299


Appendix F<br />

MicroRNA Array Data<br />

The data generated with the Agilent microRNA array platform is reported be-<br />

low, with the name <strong>of</strong> each microRNA that was probed listed in first column -<br />

the microRNAs with an asterisk in their name represent sequences that origi-<br />

nated from the opposite strand in the pri-microRNA secondary structure [71].<br />

The expression value measured is displayed in each GNS and NS cell line, and<br />

a fold change measurement is calculated - with an FDR associated - to rep-<br />

resent the differential expression for each microRNA in GNS versus NS cell<br />

lines. Negative fold change values identify greater average expression across<br />

the NS cell lines with respect to GNS cell lines, whilst positive fold change<br />

values identify greater average expression across GNS cell lines with respect<br />

to NS cell lines. The FDR values are reported in scientific notation.<br />

300


F. MicroRNA Array Data Appendix<br />

Table F.1: Differentially expressed microRNAs in GNS vs NS cell lines at FDR


F. MicroRNA Array Data Appendix<br />

GNS NS<br />

microRNAs G7A G7B G26A G26B G144A G144B G166A G166B CB660A CB660B CB130A CB130B CB152A CB152B CB171A CB171B LogFC p-value FDR<br />

hsa-miR-514b-5p 2.54 2.61 2.74 3.48 1.98 1.98 1.33 1.73 2.5 2.45 1.41 0.81 0.77 1.57 1.22 1.1 0.8 3.91E-04 1.50E-03<br />

hsa-miR-130b* 3.91 3.92 2.5 2.23 2.52 2.8 -2.89 -2.71 -2.58 -2.93 -1.7 0.06 2.14 0.86 0.08 2.66 1.7 3.75E-04 1.45E-03<br />

hsa-miR-187* 1.14 1.44 0.17 1.1 -0.68 0.37 0.72 -0.49 0.08 -1.87 -0.83 -0.97 -0.7 -1.62 -1.08 -1.28 1.5 2.99E-04 1.19E-03<br />

hsa-miR-365 8.58 8.42 9.03 9.18 8.76 8.66 6.75 7.27 7.8 7.45 7.42 7.34 7.8 7.17 8 7.92 0.7 2.85E-04 1.15E-03<br />

hsa-miR-340* 6.71 6.84 6.21 6.37 7.33 7.43 4.33 4.6 5.66 5.54 5.43 4.89 6.15 6.03 5.52 4.92 0.7 2.71E-04 1.10E-03<br />

hsa-miR-4323 2.11 2.23 2.44 2.22 1.64 1.69 0.23 0.18 1.15 0.91 0.28 0.4 0.98 1.38 1.32 1.33 0.6 2.61E-04 1.07E-03<br />

hsa-miR-374c 5.06 5.32 4.4 4.56 3.8 4 -1.71 1.04 3.15 1.86 -1.03 -1.74 2.12 0.16 0.04 2.78 2.4 2.26E-04 9.49E-04<br />

hsa-miR-214 -1.04 0.09 -0.39 -0.36 2.27 2.83 -2.15 -1.93 -1.62 -1.05 -1.96 -0.91 -1.52 -0.71 -0.94 -2.02 1.3 2.24E-04 9.47E-04<br />

hsa-miR-4299 6.17 5.65 7.74 7.81 6.77 6.66 6.79 6.93 6.83 6.81 5.95 5.56 5.96 5.84 6.06 6.14 0.7 2.22E-04 9.42E-04<br />

hsa-miR-3176 0.34 0.36 0.87 0.83 0.1 -0.73 -1.08 -1.83 -0.36 -0.72 -1.29 -1.13 -0.9 -1.25 -1.06 -1.27 0.9 2.09E-04 9.01E-04<br />

hsa-miR-129* 3.47 3.71 3.6 3.42 4.69 4.23 3.06 3.14 2.99 2.93 2.56 2.75 3.4 3.24 3.17 2.95 0.7 2.09E-04 9.01E-04<br />

hsa-miR-1290 3.42 3.61 2.68 3.42 3.9 3.81 2.38 3.01 3.18 3.13 2.31 2.25 2.43 2.23 2.32 1.98 0.8 2.04E-04 8.87E-04<br />

hsa-miR-185 6.96 7.11 7.17 7.07 7.77 7.55 6.38 6.66 6.15 5.53 6.3 6.36 6.91 6.52 6.59 6.44 0.7 1.81E-04 7.99E-04<br />

hsa-miR-671-5p 3.05 3.16 2.93 3.26 2.9 3.24 3.2 2.62 2.51 2.41 0.83 2.12 2.24 1.32 2.18 1.98 1.1 1.68E-04 7.51E-04<br />

hsa-miR-3607-5p 3.6 3.93 2.58 3.88 2.29 3.57 1.86 3 2.55 2.79 1.44 2.11 -0.08 1.35 1.2 1.06 1.5 1.57E-04 7.07E-04<br />

hsa-miR-545 2.15 2.84 1.45 2.48 4.14 4.37 0.92 1.48 -1.76 -3.67 0.37 1.57 3.6 2.97 1.87 2.26 1.6 1.43E-04 6.56E-04<br />

hsa-miR-590-5p 9.23 9.6 8.57 8.77 8.83 8.57 7.55 8.01 6.99 5.48 8.03 7.8 8.21 7.89 8.13 7.87 1.1 1.43E-04 6.56E-04<br />

hsa-miR-1973 4.94 5 3.38 3.62 2.97 3.63 2.15 3.3 2.61 1.92 -1.6 1.02 1.96 -0.3 1.43 3.28 2.3 1.31E-04 6.14E-04<br />

hsa-miR-135b 11.25 11.09 8.22 8.68 7.12 6.78 10.79 11.45 7.9 7.18 9.73 8.29 6.56 5.95 9.92 8.53 1.4 1.32E-04 6.14E-04<br />

hsa-miR-149* 0.51 0.77 0.41 0.2 -0.8 -0.51 -1.02 -1.06 -1.07 -1.1 -1.22 -1.54 -1.21 -0.83 -1.93 -0.6 1 1.32E-04 6.14E-04<br />

hsa-miR-20b* -0.99 -1.17 -1.15 -0.77 -1.02 -1.3 -1.97 -2.21 -2.03 -2.19 -2.19 -1.81 -2.14 -2.06 -1.86 -2.25 0.7 1.31E-04 6.14E-04<br />

hsa-miR-3162 8.59 9.17 8.29 9.92 8.68 8.85 9.9 9.76 8.57 8.32 7.31 7.26 8.34 8.28 7.45 8.13 1.2 1.26E-04 6.00E-04<br />

hsa-miR-935 1.9 1.58 -0.41 -0.58 0.41 -0.67 -1.31 -0.43 -0.44 0.85 -1.77 -1.11 -1.78 -2.33 -2.34 -1.53 1.4 1.20E-04 5.74E-04<br />

hsa-miR-3200-5p 0.94 0.05 -0.24 -0.25 -0.45 -0.25 -1.06 -0.71 -1.57 -1.74 -0.74 -0.99 -0.74 -0.77 -1.09 -1.01 0.8 1.11E-04 5.42E-04<br />

hsa-miR-3065-3p 2.39 2.87 3.33 3.4 3.79 3.87 -0.19 0.64 1.39 1.69 1.46 -0.23 1.94 2.28 0.79 0.69 1.3 1.06E-04 5.21E-04<br />

hsa-miR-3621 0.96 0.43 -0.05 0.36 -0.18 0.31 -0.96 -1.11 -0.17 -0.82 -1.15 -1.1 -1.35 -0.5 -1.47 -1.47 1 1.04E-04 5.10E-04<br />

hsa-miR-3937 3.09 3.31 3.12 3.56 2.58 2.94 3.03 3.19 2.71 1.95 1.74 1.33 0.88 2.58 1.57 1.81 1.3 9.89E-05 4.90E-04<br />

hsa-miR-34b 3.17 2.7 4.35 4.14 -0.19 -0.4 4.32 4.6 2.73 2.11 -0.76 0.49 3.5 3.43 0.2 1.29 1.2 9.87E-05 4.90E-04<br />

hsa-miR-3188 3.36 3.59 3.17 4.1 2.97 3.21 2.98 3.16 3.13 3.15 1.64 1.26 3.07 3.01 1.76 2.14 0.9 6.69E-05 3.47E-04<br />

hsa-miR-760 2.29 2.4 1.89 2.48 2.11 2.25 1.66 1.4 0.22 0.2 1.93 1.27 1.08 0.7 1.99 1.27 1 6.02E-05 3.16E-04<br />

hsa-miR-423-3p 3.85 3.45 2.93 2.74 2.6 2.22 1.71 2.04 1.28 0.64 1.71 0.82 2.27 1.58 2.39 1.97 1.1 5.54E-05 2.93E-04<br />

hsa-miR-3679-5p 5.72 5.81 5.62 6.16 5.16 5.83 5.01 5.18 6.06 5.87 3.78 3.89 4.28 4.18 4.08 4.76 1 5.21E-05 2.77E-04<br />

hsa-miR-4322 3.44 3.36 2.44 2.81 1.97 2.07 1.69 1.89 2.49 2.23 1.17 1.28 0.8 1.53 1.23 0.06 1.1 4.72E-05 2.54E-04<br />

hsa-miR-26b 10.61 10.75 9.79 10.17 9.35 9.39 8.08 8.74 9.27 8.81 8.44 7.89 8.83 8.86 8.77 8.42 1 4.44E-05 2.41E-04<br />

hsa-miR-769-5p 7.58 7.53 7.21 7.09 6.89 6.74 5.68 5.75 6.1 6.03 6.02 5.56 6.33 6.28 6.09 5.59 0.8 4.33E-05 2.36E-04<br />

hsa-miR-340 8.66 9.06 7.59 7.91 8.32 8.26 5.66 5.97 6.74 5.9 6.7 6.17 7.36 7.27 6.78 6.59 1 4.24E-05 2.34E-04<br />

hsa-miR-125a-3p 5.18 5.19 4.76 4.88 4.33 4.19 4.16 4.06 4.44 3.98 3.52 3.51 4.04 3.86 3.71 3.61 0.8 4.25E-05 2.34E-04<br />

hsa-miR-208b 3.99 4.4 1.91 2.17 4.5 4.53 1.47 1.46 1.23 1.65 2.66 2.36 2.76 2.54 2.37 2.32 0.8 4.01E-05 2.22E-04<br />

hsa-miR-128 8.13 8.18 7.63 7.59 6.6 6.88 5.28 5.67 6.19 5.84 5.67 6.1 6.56 6.22 6.1 6.35 0.9 3.68E-05 2.06E-04<br />

hsa-miR-4304 1.79 2.27 1.8 1.95 1.17 1.24 0.32 0.41 1.4 1.37 0.08 -0.35 -0.02 0.2 0.17 0.81 0.9 3.67E-05 2.06E-04<br />

hsa-miR-29b 12.87 12.94 13.11 13.16 13.07 13.08 13.02 13.56 10.64 8.35 11.83 12.41 11.41 11.23 12.47 13 1.7 3.59E-05 2.04E-04<br />

hsa-miR-95 7.72 8.24 1.52 2.55 4.85 5.34 -2.35 -2.21 4.95 4.86 -0.66 -1.64 4.17 3.78 0.06 -0.82 1.4 2.89E-05 1.69E-04<br />

hsa-miR-124* 3.06 3.15 -0.19 -0.76 2.86 2.95 0.61 0.16 -1.08 -0.02 -1.12 -0.05 -0.32 -0.14 1.1 -0.9 1.8 2.80E-05 1.65E-04<br />

hsa-miR-101 8.43 8.68 6.61 6.9 7.96 8.21 6.77 7.29 6.6 4.83 6.18 5.9 6.75 6.51 6.31 6.35 1.4 2.48E-05 1.49E-04<br />

hsa-miR-570 1.5 1.49 2.86 2.31 2.67 2.39 1.92 2.3 0.92 -0.62 0.44 0.24 1.42 1.16 1.17 1.75 1.4 2.48E-05 1.49E-04<br />

hsa-miR-3196 6.87 7.28 6.59 7.25 6.68 7.05 6.58 6.66 6.54 6.45 5.48 5.42 5.89 5.78 5.76 6.08 0.9 2.27E-05 1.40E-04<br />

hsa-miR-4324 5.64 5.7 4.11 4.73 4.92 5.24 -3.79 -3.54 4.33 3.5 -1.02 -3 2.44 1.73 0.63 0.69 1.7 2.03E-05 1.27E-04<br />

hsa-miR-484 7.33 7.51 7.34 7.01 6.8 6.76 6.04 6 6.17 6.11 5.88 5.77 6.22 6.15 5.96 6.17 0.8 2.00E-05 1.26E-04<br />

hsa-miR-629* 4.88 4.91 4.07 4.08 3.87 4.03 4.31 4.1 4.12 3.91 3.36 3.21 3.52 3.48 3.25 2.88 0.8 2.00E-05 1.26E-04<br />

hsa-miR-371-5p 5.66 5.85 5.93 6.68 5.06 4.99 5.64 5.9 4.75 4.57 4.7 3.9 4.74 4.83 4.8 4 1.2 1.97E-05 1.25E-04<br />

hsa-miR-137 4.51 4.44 6.22 6.54 6.65 6.75 4.03 4.46 7.1 6.31 3.72 3.56 3.75 3.94 3.71 3.3 1 1.91E-05 1.22E-04<br />

hsa-miR-4271 6.58 6.84 6.29 6.94 5.5 5.27 5.83 5.95 5.6 5.34 4.84 3.64 4.65 5.29 4.92 4.63 1.3 1.89E-05 1.22E-04<br />

302


F. MicroRNA Array Data Appendix<br />

GNS NS<br />

microRNAs G7A G7B G26A G26B G144A G144B G166A G166B CB660A CB660B CB130A CB130B CB152A CB152B CB171A CB171B LogFC p-value FDR<br />

hsa-miR-378* 4.12 3.99 0.27 2.1 3.84 3.32 -0.89 -0.46 -0.84 0.35 -1.52 0.85 -0.35 -0.52 -0.93 0.12 2.4 1.86E-05 1.21E-04<br />

hsa-miR-450b-5p 3.48 3.73 2.31 2.74 3.18 3.66 2.32 1.99 2.26 1.63 1.67 2.03 2.66 2.41 1.08 1.3 1 1.73E-05 1.13E-04<br />

hsa-miR-30e 9 9.36 9.93 9.9 8.07 8.21 7.51 8.09 8.44 7.28 7.03 6.87 8.07 8.07 7.49 7.41 1.2 1.61E-05 1.06E-04<br />

hsa-miR-3665 8.04 8.35 8.04 8.83 7.64 7.58 8.09 8.32 7.6 7.72 6.54 5.59 6.66 7.18 6.85 5.91 1.4 1.57E-05 1.04E-04<br />

hsa-miR-129-3p 4.48 4.46 3.52 3.2 4.47 4.63 2.27 2.44 2.93 2.99 2.59 2.7 2.77 2.19 2.9 2.16 1 1.44E-05 9.63E-05<br />

hsa-miR-23a 12.81 12.59 12.67 12.85 11.48 11.41 10.52 11.15 11.61 11.2 10.54 10.26 11.44 11.2 10.78 10.67 1 1.40E-05 9.44E-05<br />

hsa-miR-320b 9.63 9.55 9.06 8.96 8.92 8.72 8.56 8.86 8.22 7.98 8.12 7.78 8.4 8.44 8.25 8.22 0.9 1.39E-05 9.40E-05<br />

hsa-miR-4317 6.09 5.79 5.62 5.85 4.06 4.09 0.37 2.73 4.18 3 -0.34 -1.82 3.32 2.32 0.9 2.49 2.6 1.25E-05 8.61E-05<br />

hsa-miR-629 1.44 2.01 0.67 1.35 0.16 1.05 -0.81 0.29 0.67 0.68 -1.01 -1.9 -1.54 -1.47 -0.77 -1.56 1.6 1.03E-05 7.24E-05<br />

hsa-miR-148a* 0.91 0.65 0.76 0.78 -1.05 -1.08 -1.04 -0.51 -1.2 -1.59 -2.04 -0.8 -1.7 -1.71 -1.61 -0.86 1.4 1.04E-05 7.24E-05<br />

hsa-miR-140-5p 10.97 10.71 9.25 9.58 12.19 12.22 8.34 8.5 9.39 8.74 9.18 8.81 9.47 9.48 9.36 8.85 1.1 9.76E-06 6.96E-05<br />

hsa-miR-146b-5p 7.41 7.49 4.98 5.54 5.51 5.33 4.87 4.97 4.96 4.76 4.87 4.87 4.75 4.99 4.82 4.58 0.9 9.75E-06 6.96E-05<br />

hsa-miR-711 1.63 1.83 1.27 1.41 0.34 1.27 1 0.47 -0.96 -0.43 -0.59 0.19 -0.23 0.26 -0.28 0.22 1.4 9.61E-06 6.95E-05<br />

hsa-miR-660 7.54 7.79 7.55 7.76 8.26 7.84 6.92 7.3 7.06 6.71 6.54 6.43 6.8 6.78 6.51 6.4 1 8.62E-06 6.31E-05<br />

hsa-miR-1207-5p 7.02 7.09 7.22 7.8 7.18 7.59 7.01 7.05 7.41 7.1 5.67 5.15 5.93 6.11 6.06 6.1 1.1 8.14E-06 5.99E-05<br />

hsa-miR-222 6.83 6.41 7.62 7.68 3.66 3.63 6.95 7.09 5.92 5.76 3.8 4.21 4.92 4.73 4.63 5.82 1.3 8.05E-06 5.96E-05<br />

hsa-miR-598 8 8 7.43 7.39 7.68 7.5 6.84 7.01 6.63 6.33 6.69 6.55 6.66 6.52 6.82 6.69 0.9 8.00E-06 5.95E-05<br />

hsa-miR-181a 11.59 11.76 9.88 9.97 11.25 11.66 7.97 8.27 10.01 9.73 8.93 8.21 9.32 9.43 9.08 8.42 1.2 7.90E-06 5.91E-05<br />

hsa-miR-105 1.46 1.97 -0.62 -0.72 2.53 2.71 0.13 -0.56 -0.43 -0.95 -0.2 0.17 -0.44 -0.88 -0.01 -0.84 1.3 7.68E-06 5.81E-05<br />

hsa-miR-449a 3.88 3.85 3.6 3.79 2.56 2.38 1.74 3.52 2.17 1.65 1.24 0.91 0.57 -0.1 1.75 -0.19 2.2 7.41E-06 5.65E-05<br />

hsa-miR-139-3p 2.16 2.02 1.21 1.61 3.89 3.47 -1.75 -0.13 -0.59 -0.68 -0.01 -1.51 -1.15 0.1 -0.31 0.11 2.1 7.43E-06 5.65E-05<br />

hsa-miR-1225-5p 6.66 6.74 6.51 7.16 6.54 6.89 7 7.24 6.43 6.13 5.21 4.39 5.78 6.13 5.55 4.98 1.3 7.35E-06 5.65E-05<br />

hsa-miR-490-5p 0.87 0.79 -0.74 0.86 0.19 -0.25 -1.61 -1.55 -1.29 -1.67 -1.87 -1.55 -1.41 -1.83 -2.25 -1.97 1.6 6.47E-06 5.04E-05<br />

hsa-miR-3663-3p 6.36 6.41 6.14 6.45 5.8 5.79 5.6 5.91 5.37 5.01 4.09 3.42 5.5 5.59 4.65 3.3 1.4 6.11E-06 4.78E-05<br />

hsa-miR-200a 4.01 4.31 3.1 3.61 1.92 0.89 0.47 0.35 0.91 0.9 0.97 0.98 1.26 -0.34 -0.37 0.31 1.8 5.99E-06 4.72E-05<br />

hsa-miR-15b* 4.2 4.12 3.42 3.24 2.24 2.3 2.1 2.34 -0.58 0.13 0.97 1.38 1.06 -0.61 0.61 2.79 2.3 5.80E-06 4.59E-05<br />

hsa-miR-195 11.3 11.31 8.79 9.23 8.48 8.53 7.06 7.67 7.76 6.7 7.62 6.93 8.43 8.19 7.82 7.09 1.5 5.71E-06 4.55E-05<br />

hsa-miR-16 13.44 13.4 12.5 12.61 11.98 11.81 11.64 12.16 11.44 11 11.41 11.21 11.68 11.4 11.67 11.51 1 5.70E-06 4.55E-05<br />

hsa-miR-15b 14.51 14.17 13.91 13.8 12.97 12.7 13.2 13.72 12.27 12.01 12.75 12.46 12.82 12.52 12.91 12.52 1.1 5.20E-06 4.27E-05<br />

hsa-miR-29a* 2.3 2.61 2.6 2.96 3.57 3.69 2.13 2.61 1.55 -0.4 -0.63 0.57 1.11 1 1.5 1.79 2 4.73E-06 3.95E-05<br />

hsa-miR-3652 2.86 3.37 2.52 3.89 2.72 2.61 3.38 3.42 2.19 1.93 0.68 -0.49 2.13 2.23 1.35 1.39 1.7 4.28E-06 3.72E-05<br />

hsa-miR-1226* 4.44 4.62 4.5 4.7 3.6 3.23 3.38 3.58 3.38 3.03 3 2.57 2.7 2.87 2.41 3 1.1 3.94E-06 3.49E-05<br />

hsa-miR-4306 7.84 7.92 7.91 7.87 8.3 8.21 6.92 7.2 6.9 6.34 6.51 6.56 7.11 6.82 6.77 6.95 1 3.90E-06 3.48E-05<br />

hsa-miR-193b* 1.98 1.82 2.36 2.7 2.56 2.42 -0.83 -0.33 0.49 -0.14 -1.62 0.09 -0.56 -1.35 -0.21 0.6 1.9 3.82E-06 3.45E-05<br />

hsa-let-7f 15.52 15.6 15.18 15.47 14.52 14.37 14.19 14.86 14.18 13.51 13.43 13.35 14.1 13.87 13.85 13.99 1.2 3.64E-06 3.33E-05<br />

hsa-miR-3174 4.63 4.83 4.02 3.74 2.41 1.77 0.74 1.71 1.76 1.53 0.86 1.65 1.19 1.67 1.46 1.8 1.5 3.60E-06 3.32E-05<br />

hsa-miR-642b 6.32 6.37 6.02 6.89 6.03 6.16 6.55 6.56 5.25 4.85 4.7 4.22 5.18 5.72 4.94 3.86 1.5 3.53E-06 3.31E-05<br />

hsa-miR-451 4.5 4.32 2.86 2.74 3.32 3.32 2.81 2.63 2.55 2.46 2.39 2.15 2.32 2.46 1.79 2.28 1 3.34E-06 3.15E-05<br />

hsa-miR-7 6.39 6.54 3.65 4.32 6.93 7.22 4.68 4.49 3.24 3.51 3.18 4.38 3.55 3.01 3.35 4.85 1.9 3.20E-06 3.03E-05<br />

hsa-miR-146a 4.58 4.84 2.46 2.99 6.05 5.67 1.66 1.47 2.06 2.47 2.51 2.52 2.05 2.04 3.03 1.94 1.4 3.15E-06 3.03E-05<br />

hsa-let-7a 14.87 14.84 14.45 14.5 13.97 13.82 13.33 14.02 13.03 12.59 12.99 12.68 13.41 13.24 13.38 12.86 1.2 3.17E-06 3.03E-05<br />

hsa-miR-3610 3.31 3.33 3.33 4.91 2.71 3.31 3.68 3.61 3.08 2.2 0.48 0.53 1.4 2.48 1.35 1.22 1.9 3.03E-06 2.94E-05<br />

hsa-miR-4281 8.13 8.21 7.77 8.42 8.14 8.31 8.42 8.49 7.72 7.49 6.41 6.25 7.37 7.64 6.72 7.18 1.1 2.92E-06 2.85E-05<br />

hsa-miR-3651 6.93 6.57 5.82 5.87 5.95 5.51 6.65 7.31 5.59 4.77 4.89 4.52 5.2 5.04 5.04 4.77 1.3 2.88E-06 2.83E-05<br />

hsa-miR-3141 5.22 5.53 5.11 5.48 4.37 4.46 4.41 4.62 4.23 4.11 3.37 3.44 3.53 3.48 3.3 4.15 1.2 2.76E-06 2.73E-05<br />

hsa-miR-29a 12.2 12.39 12.19 12.19 11.55 11.41 11.25 11.85 10.56 9.41 10.62 10.7 10.38 10.24 11.11 10.96 1.4 2.65E-06 2.64E-05<br />

hsa-miR-193b 6.4 6.32 7.28 7.15 6.82 6.6 4.68 5.07 5.12 4.65 5.11 5.12 4.91 4.11 5.82 5.53 1.2 2.45E-06 2.46E-05<br />

hsa-let-7g 12.74 12.74 12.26 12.47 11.31 11.13 10.74 11.34 10.71 10.23 10.52 10.16 11.05 10.94 10.88 10.29 1.2 2.38E-06 2.41E-05<br />

hsa-miR-425 9.31 9 9.46 9.46 8.7 8.54 9.16 9.76 7.76 7.61 8.1 7.92 8.39 8.3 8.33 8.31 1.1 2.28E-06 2.32E-05<br />

hsa-miR-3934 2.46 2.51 2.25 1.83 0.98 1.39 0.4 1.41 0.94 0.51 -0.87 -0.59 0.03 -0.05 0.4 1.06 1.5 2.20E-06 2.27E-05<br />

hsa-miR-503 5.77 5.65 4.71 5.13 4.72 5.49 3.27 3.87 4.78 2.96 1.69 0.98 5.46 4.69 0.18 0.64 2.2 2.05E-06 2.14E-05<br />

hsa-miR-4270 4.8 4.88 4.8 5.28 4.32 4.93 4.27 4.35 4.36 4.11 3.07 2.61 3.82 3.95 2.98 3.05 1.2 1.88E-06 1.98E-05<br />

303


F. MicroRNA Array Data Appendix<br />

GNS NS<br />

microRNAs G7A G7B G26A G26B G144A G144B G166A G166B CB660A CB660B CB130A CB130B CB152A CB152B CB171A CB171B LogFC p-value FDR<br />

hsa-miR-762 5.94 6.4 5.5 6.29 5.68 5.74 5.03 5 5.45 5.68 3.69 2.78 4.47 4.76 3.59 3.62 1.4 1.79E-06 1.90E-05<br />

hsa-miR-106b 13.58 13.74 12.99 12.97 12.23 12.14 10.91 11.37 10.44 9.66 11.5 11.39 11.85 11.57 11.84 11.7 1.2 1.51E-06 1.61E-05<br />

hsa-miR-93 12.56 12.52 12.49 12.25 11.35 11.13 10.08 10.31 9.69 9.46 10.7 10.63 10.81 10.43 10.8 11.19 1.1 1.48E-06 1.59E-05<br />

hsa-miR-1285 3.13 3.55 2.55 2.9 1.58 1.7 -0.07 0.87 -1.02 -1.13 -1.59 0.15 0.09 -0.66 -1.37 0.56 2.6 1.31E-06 1.44E-05<br />

hsa-miR-630 7.37 7.13 7.81 7.71 6.67 8.1 4.32 4.61 7.43 7.17 3.79 3.73 5.18 4.9 3.9 4.38 1.7 1.30E-06 1.44E-05<br />

hsa-miR-26a 11.67 11.8 9.99 10.19 12.63 12.51 8.54 9.09 9.77 9.52 9.48 8.59 9.23 9.48 9.69 9.14 1.4 1.27E-06 1.43E-05<br />

hsa-miR-29c 10.44 10.67 10.87 10.79 9.45 9.3 10 10.6 8.67 7.33 8.65 8.65 8.85 8.65 9.13 9.17 1.6 1.04E-06 1.18E-05<br />

hsa-miR-452 6.57 6.02 -0.35 0.35 0.28 1.01 0.14 -0.12 -0.7 -0.55 0.92 0.77 -0.59 0.3 -0.62 0.17 1.8 9.05E-07 1.05E-05<br />

hsa-miR-188-5p 4.27 4.18 4.62 4.98 4.06 5.02 3.98 4.17 3.55 3.16 2.58 2.03 3.31 3.26 2.57 2.86 1.5 9.02E-07 1.05E-05<br />

hsa-miR-483-5p 4.39 4.58 4.16 4.36 4.23 4.97 2.39 2.68 3.86 3.67 2.09 1.4 1.53 1.61 1.81 2.75 1.6 7.87E-07 9.49E-06<br />

hsa-miR-1915* 1.48 1.38 0.81 0.77 0.11 0.1 0.13 0.61 -0.07 -0.07 -1.02 0.05 -1.03 -1.16 -1.43 -1.8 1.5 7.89E-07 9.49E-06<br />

hsa-miR-589* 0 -0.11 -0.26 0.13 -1.23 -1.25 -0.61 -0.39 -2.07 -1.86 -1.75 -2.53 -1.32 -1.88 -2.12 -1.7 1.4 7.64E-07 9.35E-06<br />

hsa-miR-664 6.32 6.25 5.29 5.7 5.05 4.98 3.68 3.76 4.76 4.64 3.65 3.45 3.98 4.14 3.8 3.6 1.1 7.06E-07 8.72E-06<br />

hsa-miR-224 8.65 7.99 -0.01 -0.12 2.47 2.55 1.1 0.3 0.65 0.62 2.73 1.99 1.29 1.37 0.68 1.05 1.6 6.73E-07 8.47E-06<br />

hsa-miR-550a* 6.74 6.7 5.88 5.58 4.63 4.43 5.17 5.4 3.48 3.74 4.4 4.88 4.28 4.04 4.64 4.97 1.3 6.09E-07 7.81E-06<br />

hsa-miR-345 2.07 2.14 2.47 2.11 2.27 1.98 -0.24 0.64 -0.08 -0.89 -1.35 -1.19 0.16 -0.87 -0.42 0.85 2.2 5.78E-07 7.55E-06<br />

hsa-miR-30b 9.44 9.35 10.26 10.06 8.15 7.81 8.54 9.23 7.63 7.22 7.53 6.77 8.09 7.83 7.8 7.21 1.6 5.83E-07 7.55E-06<br />

hsa-miR-584 4.79 4.62 2.79 2.95 4.2 4.17 3.25 3.31 3.72 3.59 2.26 2.47 2.51 2.58 2.11 1.69 1.1 5.77E-07 7.55E-06<br />

hsa-miR-4318 3.83 3.59 3.01 2.77 0.81 1.43 0.29 0.45 0.85 0.59 0.08 1.01 0.05 0.55 0.25 -0.01 1.6 5.25E-07 7.14E-06<br />

hsa-miR-30d 10.35 10.36 11.76 11.52 9.9 9.71 9.85 10.28 9.65 9.67 8.76 8.04 9.35 9.52 9.01 8.57 1.4 5.13E-07 7.05E-06<br />

hsa-miR-3607-3p 3.84 4.19 2.65 2.8 2.99 3.24 1.81 2.52 2.57 2.37 1.09 0.24 0.76 1.21 1.35 0.59 1.7 4.78E-07 6.64E-06<br />

hsa-miR-3615 1.41 1.66 0.49 2.03 -1 -0.69 -1.13 -0.99 -0.88 -1.04 -2.26 -2.16 -1.5 -1.11 -2.12 -2.15 1.9 4.03E-07 5.78E-06<br />

hsa-miR-30b* 3.86 4.01 4.77 4.68 3.11 2.8 4.27 4.48 1.68 1.65 2.31 2.07 3.26 2.19 2.91 2.03 1.7 4.12E-07 5.78E-06<br />

hsa-miR-20b 11.79 11.98 12.66 12.56 11.47 11 9.71 10.31 9.53 8.79 10.12 10.32 9.38 9.01 10.32 10.94 1.6 4.06E-07 5.78E-06<br />

hsa-miR-3194 2.54 3.02 2.44 3.35 2.56 2.62 2.27 2.32 1.4 1.44 0.36 0.19 1.39 1.95 0.56 1.04 1.6 4.12E-07 5.78E-06<br />

hsa-miR-4298 4.97 5.17 4.64 5.02 3.79 4.53 3.44 3.42 3.58 3.61 2.05 1.99 2.48 2.56 2.37 3.35 1.6 4.10E-07 5.78E-06<br />

hsa-miR-150* 5.95 6.28 5.47 6.35 5.13 5.06 5.64 5.59 5.05 5 3.62 3.41 4 4.31 4.05 3.43 1.6 3.34E-07 5.00E-06<br />

hsa-miR-124 10.04 10.47 0.4 0.44 10.54 10.53 -0.55 -2.15 -0.75 -0.49 3 0.55 3.87 4.27 -1.24 0.93 3.7 3.25E-07 4.92E-06<br />

hsa-miR-1915 8.53 8.9 7.81 8.78 8.27 8.24 9.02 8.95 7.8 7.7 6.14 5.3 6.86 7.44 6.35 5.59 1.9 3.21E-07 4.92E-06<br />

hsa-miR-30d* 2.17 2.45 3.97 3.75 1.08 -0.17 2.72 3.96 -1.92 -3.21 -1.89 -2.27 1.98 -0.18 0.55 -0.8 3.5 2.97E-07 4.60E-06<br />

hsa-miR-1273e 2.2 2.46 1.67 2.65 0.12 0.58 1.9 1.91 -1.25 -0.76 -0.47 -1 -0.08 -0.63 -0.9 0.54 2.3 2.89E-07 4.53E-06<br />

hsa-miR-362-3p 6.21 6.42 5.81 5.88 6.59 6.13 3.96 4.66 4.6 2.9 2.81 1.56 4.23 3.44 2.04 2.72 2.7 2.75E-07 4.40E-06<br />

hsa-miR-454 7.64 7.96 6.42 7.02 7.19 7.37 4.42 5.09 5.78 5.08 4.02 3.17 5.47 5.74 4.58 4.35 1.9 2.30E-07 3.73E-06<br />

hsa-miR-877 1.92 2.27 1.23 2.56 0.53 1.25 1.31 1.16 -0.47 -0.81 -0.68 -1.04 -0.64 0.01 -0.78 -0.33 2.1 2.22E-07 3.65E-06<br />

hsa-miR-500a* 5.75 5.85 5.83 5.87 6.44 5.94 4.86 5.05 5.08 5.16 3.73 3.81 4.6 4.62 4.07 4.32 1.3 2.22E-07 3.65E-06<br />

hsa-miR-320c 8.03 7.86 7.77 7.7 7.12 7.2 6.9 7.17 6.81 6.46 5.75 5.26 6.16 6.24 5.92 6.25 1.4 2.11E-07 3.60E-06<br />

hsa-miR-199a-3p 8.68 8.85 2.18 2.55 8.3 7.86 -0.76 -0.23 3.58 3.37 2.11 1.17 0.26 0.91 2.71 0.5 2.9 2.04E-07 3.52E-06<br />

hsa-miR-423-5p 6.41 6.33 5.65 5.44 4.11 4.01 3.62 4.26 3.69 3.28 2.63 1.54 3.78 3.59 3.59 3.5 1.8 1.88E-07 3.29E-06<br />

hsa-let-7b 14.64 14.61 14.02 13.92 13.3 13.16 12.82 13.47 11.75 11.16 12.1 11.67 12.29 12.13 12.49 11.41 1.9 1.84E-07 3.27E-06<br />

hsa-miR-488* 2.77 3 3.28 3.89 3.88 4.41 -1.71 -1.63 -1.74 -1.57 1.4 0.02 -1.87 -1.09 1.77 0.39 2.6 1.69E-07 3.03E-06<br />

hsa-miR-3065-5p 3.53 3.62 4.44 4.66 4.88 4.88 0.87 2 2.21 1.68 0.63 0.67 3.15 3.29 1.5 0.82 1.9 1.69E-07 3.03E-06<br />

hsa-miR-32 4.12 3.99 0.34 1.75 4.14 4.43 -0.52 2.51 -3.73 -4.55 -0.98 -2.44 -1.46 -2.5 1.72 1.61 4.1 1.60E-07 2.96E-06<br />

hsa-miR-320d 10.53 10.41 10.04 9.87 9.59 9.44 9.25 9.58 8.89 8.55 8.3 7.81 8.55 8.7 8.44 8.33 1.4 1.48E-07 2.80E-06<br />

hsa-miR-4327 5.09 5.34 4.84 5.45 4.16 4.07 4.5 4.57 4.13 4 2.18 1.28 2.58 3.18 2.97 2.2 1.9 1.30E-07 2.53E-06<br />

hsa-miR-361-3p 6.09 5.96 6.01 6.41 4.37 4.46 3.56 3.78 4.17 3.96 3.11 2.32 3.88 3.71 3.59 3.33 1.6 1.29E-07 2.53E-06<br />

hsa-miR-664* 3.63 3.52 2.54 3.26 2.81 2.82 -0.19 0.11 2.11 2.34 -1.16 -1.32 0.55 0.33 0.37 -0.94 2 1.18E-07 2.38E-06<br />

hsa-miR-492 3.09 3.1 1.97 2.97 2.41 2.41 2.91 2.83 1.16 0.44 0.52 -0.52 1.01 1.17 0.66 0.25 2.1 1.11E-07 2.26E-06<br />

hsa-miR-424 12.87 12.79 11.31 11.95 12.07 12.87 9.77 10.36 11.39 9.36 7.33 6.82 11.52 11.17 6.66 6.88 2.9 1.01E-07 2.10E-06<br />

hsa-miR-200b 5.36 5.23 4.31 4.7 3.76 2.92 1.85 2.79 1.95 2.39 1.72 0.64 1.96 2 0.63 1.19 2.3 1.01E-07 2.10E-06<br />

hsa-miR-1181 7.14 7.72 6.28 6.94 5.95 5.79 6.61 6.53 5.36 5.38 4.6 4.5 4.75 5.29 4.81 4.28 1.7 9.04E-08 1.98E-06<br />

hsa-miR-320e 9.82 9.74 9.31 9.2 8.9 8.77 8.46 8.79 8.17 7.83 7.46 6.98 7.7 7.89 7.61 7.75 1.5 9.16E-08 1.98E-06<br />

hsa-miR-542-3p 8.8 9.12 6.99 8.06 8.24 8.95 6.1 6.4 7.57 6.34 3.77 2.8 8.08 7.74 2.97 2.63 2.6 8.39E-08 1.91E-06<br />

304


F. MicroRNA Array Data Appendix<br />

GNS NS<br />

microRNAs G7A G7B G26A G26B G144A G144B G166A G166B CB660A CB660B CB130A CB130B CB152A CB152B CB171A CB171B LogFC p-value FDR<br />

hsa-miR-34c-3p 0.51 0.34 0.45 0.72 -1.75 -2.1 -1.58 -1.28 -1.98 -2.25 -2.4 -2.82 -1.66 -2.5 -2.5 -2.38 1.7 8.50E-08 1.91E-06<br />

hsa-miR-3609 3.73 4.15 3.19 3.56 2.37 2.59 0.16 0.8 2.05 1.95 -0.71 -0.03 0.07 0.31 1.01 0.21 2 7.33E-08 1.70E-06<br />

hsa-miR-25 13.1 13.05 12.06 12.09 11.86 11.63 9.99 10.44 9.88 9.54 10.44 10.44 10.6 10.29 10.65 10.74 1.5 7.07E-08 1.67E-06<br />

hsa-miR-885-5p 6.59 6.58 1.25 1.99 5.06 4.58 -0.48 -0.72 1.21 0.84 0.33 -0.77 -0.51 -1 -0.12 1.66 2.9 6.18E-08 1.51E-06<br />

hsa-miR-140-3p 9.2 8.7 7.21 7.27 9.37 9.37 5.29 5.58 6.43 6.14 6.02 5.74 6.37 6.17 6.38 6.2 1.6 3.99E-08 9.94E-07<br />

hsa-miR-2276 5.71 6.43 3.34 4.75 4.85 4.87 4.99 4.7 3.26 3.2 -0.45 1.43 1.39 2.05 1.39 1.29 3.3 3.62E-08 9.29E-07<br />

hsa-miR-338-3p 8.47 8.76 3.96 4.84 9.63 10.78 2.73 3.02 3.11 2.81 4.8 3.77 2.74 2.67 5.22 3.99 2.9 3.65E-08 9.29E-07<br />

hsa-miR-502-5p 3.6 3.83 3.81 3.93 4.25 3.73 2.27 3.19 2.15 1.96 0.38 -0.44 2 0.92 0.16 0.91 2.6 3.54E-08 9.29E-07<br />

hsa-miR-155 7.97 7.44 6.73 6.98 5.48 5.2 6.99 7.33 5.33 5.83 5.89 6.46 1.16 0.73 6.64 6.88 1.9 3.18E-08 8.56E-07<br />

hsa-miR-542-5p 7.26 7.37 6.24 6.43 6.74 7.36 4.02 4.35 5.96 4.95 1.97 0.8 6.44 6.21 1.9 1.3 2.5 3.00E-08 8.25E-07<br />

hsa-miR-362-5p 6.34 6.36 6.14 6.38 6.79 6.31 5.45 6.04 5.02 5.1 3.75 2.71 4.6 4.72 3.7 3.41 2.1 2.66E-08 7.45E-07<br />

hsa-miR-4257 4.55 5.02 4.38 5.85 3.7 4.39 5.3 5.43 3.78 3 -0.77 -2.4 1.95 3.18 -0.18 -1.54 4 2.33E-08 6.82E-07<br />

hsa-miR-186 7.64 7.91 7.63 8.02 6.22 6.45 5 5.23 6.1 5.84 4.05 4.45 5.53 5.39 4.59 4.81 1.7 2.21E-08 6.61E-07<br />

hsa-miR-532-5p 5.94 6.13 5.81 6.05 6.54 6 5.01 5.69 4.49 4.39 3.08 2.29 4.39 4.37 3.6 2.98 2.2 1.65E-08 5.18E-07<br />

hsa-miR-139-5p 4.27 4.37 1.34 1.36 6.36 6.21 -0.11 -0.38 -0.55 -0.17 -1.54 -0.77 -0.79 1.06 -1.36 -0.03 3.4 1.22E-08 4.00E-07<br />

hsa-miR-192 7.99 8.26 6.54 6.97 6.23 6.13 5.47 5.69 5.43 5.24 5.13 4.95 4.99 4.91 4.67 4.68 1.7 1.13E-08 3.81E-07<br />

hsa-miR-663 6.22 6.99 4.14 5.28 5.31 5.39 5.5 5.43 3.95 4.15 2.59 1.41 2.59 3.02 1.97 1.9 2.8 9.73E-09 3.36E-07<br />

hsa-miR-29c* 4.32 4.33 5.6 5.47 2.58 2.59 4.21 4.51 2.57 1.53 0.55 1.21 3.03 3 1.9 2.01 2.2 9.57E-09 3.36E-07<br />

hsa-miR-1972 2.46 3.57 0.98 2.45 0.4 0.91 1.29 2.52 -0.6 -1.88 -2.86 -2.53 -2.49 -2.18 -2.29 -2.09 3.9 5.28E-09 1.98E-07<br />

hsa-miR-1208 4.58 4.72 3.55 4.49 4 3.75 4.5 4.67 2.59 1.82 1.53 1.46 2.53 2.85 1.21 1.35 2.4 5.32E-09 1.98E-07<br />

hsa-miR-191 2.92 2.6 3.34 3.53 0.08 0.48 1.23 2.24 -0.52 -0.12 -1.04 -2.35 -1.68 -1.06 -1.58 -0.82 3.2 4.89E-09 1.95E-07<br />

hsa-miR-339-5p 1.41 0.96 0.95 1.04 -1.15 -0.41 -1.43 -1.43 -2.38 -2.36 -2.41 -2.22 -1.76 -2 -2.24 -1.72 2.1 4.93E-09 1.95E-07<br />

hsa-miR-501-3p 2.7 3.11 2.6 3.31 3.52 3.02 -1.01 -0.5 -0.65 0.1 -1.72 -1.98 0.06 -1.43 -1.86 -1.55 3.2 4.37E-09 1.84E-07<br />

hsa-miR-424* 4.62 4.34 3.36 3.73 1.93 2.94 -0.62 -0.14 1.82 0.15 -2.63 -2.35 -0.24 -0.77 -2.08 -1.78 3.5 2.91E-09 1.27E-07<br />

hsa-miR-488 6.31 6.56 6.22 6.64 6.88 7.3 -2.47 -2.52 -1.39 -1.59 5.08 4.09 -2.39 -1.8 5.27 4.24 2.9 2.65E-09 1.19E-07<br />

hsa-miR-718 2.78 3.08 2.24 2.9 1.66 2.12 0.39 0.96 0.23 -0.8 -2.01 -2.28 -0.07 -1.32 -1.89 -2.09 3.3 2.26E-09 1.05E-07<br />

hsa-miR-199b-5p 8.7 8.77 1.52 1.24 8.02 7.27 1.96 2.09 3.25 2.81 2.32 2.27 1.95 2.01 1.27 2.23 2.7 1.30E-09 6.50E-08<br />

hsa-miR-10b* 7.24 7.18 0.71 0.53 3.88 4.1 2.21 2.49 -0.46 0.52 0.79 0.72 0.67 0.98 0.28 1.25 2.9 8.25E-10 4.27E-08<br />

hsa-miR-532-3p 4.65 4.71 4.82 4.91 5.51 4.88 2.66 3.6 3.17 3.32 -0.71 -2.7 2.23 1.83 -2.24 -2.56 4.2 7.74E-10 4.17E-08<br />

hsa-miR-135a* 3.87 4.15 3.08 3.76 2.86 2.68 3.38 3.03 2.8 1.96 -1.5 -2.16 -0.79 0.02 -0.38 -1.38 3.5 6.72E-10 3.77E-08<br />

hsa-miR-215 6.72 6.99 5.17 5.69 4.99 4.88 3.79 4.02 3.52 3.18 2.87 2.57 2.53 2.89 2.82 2.86 2.4 4.79E-10 2.80E-08<br />

hsa-miR-339-3p 1.65 1.69 2.03 2.15 0.97 0.42 -1.98 -0.75 -2.39 -2.82 -2.42 -2.9 -2.36 -2.83 -2.06 -2.14 3.3 4.13E-10 2.53E-08<br />

hsa-miR-874 6.65 6.82 4.79 5.75 4.59 4.57 4.6 4.5 4.86 4.73 -1.2 -2.4 2.94 3.55 1.05 0.76 3.5 3.30E-10 2.12E-08<br />

hsa-miR-10b 12.01 11.95 4.74 4.94 8.18 8.22 6.15 6.92 1.92 1.97 5.53 5.61 6.13 6.14 5.94 6.7 2.9 1.53E-10 1.03E-08<br />

hsa-miR-625 5.69 5.92 4.53 4.94 3.78 3.84 4.83 5.15 -3.28 -3.06 3.82 4.1 3.8 3.84 3.99 4.53 2.6 1.46E-10 1.03E-08<br />

hsa-miR-96 9.43 8.84 11.22 10.47 9.1 9.79 10.24 10.87 6.63 4.82 4.42 4.56 5.98 6.15 4.81 4.78 4.7 1.11E-10 8.33E-09<br />

hsa-miR-450a 8.77 8.89 6.75 7.14 6.89 7.59 3.98 4.43 6.05 4.53 -1.22 -2.24 6.31 5.94 -1.08 -0.88 4.6 7.33E-11 5.81E-09<br />

hsa-miR-1469 2.29 2.37 2.47 2.57 0.59 1.31 -1.04 0.19 -1.91 -2.45 -2.93 -2.7 -2.33 -2.22 -2.5 -2.79 3.8 5.98E-11 5.13E-09<br />

hsa-miR-129-5p 3.05 3.21 1.45 1.72 4.88 4.14 -1.13 -0.49 -0.88 -0.41 -1.51 -1.32 -1.06 -0.83 -1.43 -1.42 3.2 6.09E-11 5.13E-09<br />

hsa-miR-363 10.05 10.11 11.75 11.63 10.9 10.67 7.46 7.96 7.92 7.03 7.71 8.06 3.92 3.98 7.9 8.26 3.2 5.76E-11 5.13E-09<br />

hsa-miR-378 6.65 6.4 3.26 4.84 7.16 6.8 -0.15 0.94 -2.19 -2.12 -2.12 -2.27 -1.57 -1.33 -2.01 -0.22 6.2 2.18E-11 2.49E-09<br />

hsa-miR-183* 0.43 -0.16 3.65 2.92 1.27 2.35 3.42 4.08 -1.7 -2.32 -2.33 -2.41 -2.21 -2.35 -2.23 -1.97 4.4 2.00E-11 2.49E-09<br />

hsa-miR-183 3.63 2.03 6.54 5.61 1.16 3.84 3.46 4.62 -4.24 -4.58 -4.79 -4.79 -4.44 -4.41 -4.63 -4.54 8.4 8.49E-12 1.27E-09<br />

hsa-miR-194 5.86 6.18 4.32 4.75 3.78 3.57 0.69 1.93 0.94 0.47 -1.84 -3.02 -1.25 -1.49 -1.94 -2.22 5.2 7.16E-12 1.20E-09<br />

hsa-miR-196a 8.87 9.09 5.05 4.64 5.86 5.37 6.27 6.68 0.42 0.32 -0.65 1.03 1.71 2.32 1.46 1.51 5.5 4.81E-12 9.25E-10<br />

hsa-miR-138 4.97 5.22 5.48 4.16 5.2 4.91 -2.52 -2.22 -2.63 -2.61 -3.95 -3.78 4.31 3.81 -3.78 -3.46 4.7 4.18E-12 9.25E-10<br />

hsa-miR-3648 4.88 4.92 4.35 4.93 4.06 4.87 4.18 4.35 3.87 3.07 -2.62 -2.85 -1.67 -0.53 -2.01 -2.07 5.2 3.29E-12 8.86E-10<br />

hsa-miR-502-3p 4.53 4.56 4.46 4.67 4.9 4.3 1.96 2.49 2.78 2.4 -2.36 -2.05 0.39 0.87 -2.42 -1.88 4.3 1.98E-12 6.66E-10<br />

hsa-miR-182 4.95 4.03 6.91 6.15 3.72 4.84 4.96 5.48 -0.98 -2 -3.14 -2.42 -2.19 -2.12 -2.96 -2.53 7.4 2.74E-13 1.23E-10<br />

hsa-miR-148a 10.36 10.32 10.3 10.87 9.02 8.9 8.89 9.5 6.6 5.95 0 0.66 0.39 1.51 0.36 0.63 7.8 1.63E-14 1.10E-11<br />

hsa-miR-196b 9.79 9.81 3.26 3.39 6.5 6.32 2.45 3.53 -3.08 -3.1 -3.77 -3.41 -2.81 -2.23 -3.57 -3.06 8.8 7.07E-16 9.52E-13<br />

305


List <strong>of</strong> Abbreviations<br />

5-bromo-2’-deoxyuridine BrdU<br />

AGO Argonaute<br />

AKT v-akt murine thymoma viral oncogene<br />

APC Antigen Presenting Cell<br />

ARF Alternate Reading Frame<br />

ASR Age Standardized Rate<br />

ATM Ataxia telangiectasia mutated<br />

BLBP Brain lipid binding protein<br />

BMP Bone Morphogenetic Protein<br />

Ca 2+ Calcium<br />

CCDS Consensus Coding Sequence<br />

CD133 Prominin<br />

CD144 Vascular endothelial-cadherin<br />

CDK Cyclyn-Dependent Kinase<br />

CDKN Cyclyn-Dependent Kinase Inhibitor<br />

CGH Comparative genomics hybridization<br />

CHI3L1 Chitinase 3-like 1<br />

CNA Copy Number Aberrations<br />

CNS Central Nervous System<br />

CO2<br />

Carbon dioxide<br />

CSF Cerebro Spinal Fluid<br />

Ct Cycle threshold<br />

CTL Cytotoxic Lymphocyte<br />

CTMP C-terminal modulator protein<br />

CpG Cytosine-phosphate-Guanine<br />

DNA Deoxyribonucleic Acid<br />

Dcx Doublecortin<br />

ECM Extracellular matrix<br />

EGF Epidermal Growth Factor<br />

EGFR Epidermal Growth Factor Receptor<br />

ES Embryonic <strong>Stem</strong><br />

EST Expressed Sequence Tags<br />

FACS Fluorescent Activated Cell Sorting<br />

FC Fold change<br />

FDR False Discovery Rate<br />

FGF2 Fibroblast Growth Factor 2<br />

FGF2 Fibroblast growth factor 2<br />

FISH Fluorescence In Situ Hybridization<br />

FOXO Forkhead box O<br />

G-CIMP <strong>Glioma</strong> CpG Island Methylator Phenotype<br />

GABA Gamma-aminobutyric acid<br />

GBM Glioblastoma Multiforme<br />

GEMM Genetically Engineered Mouse Model<br />

GFAP Glial Fibrillary Acidic Protein<br />

GFAP Glial Fibrillary Acidic Protein<br />

GFP Green Fluorescent Protein<br />

GLAST Glutamate Aspartate Transporter<br />

GO Gene Ontology<br />

GSEA Gene Set Enrichment Analysis<br />

GTP Guanosine-5’-triphosphate<br />

GTPase Guanosine-5’-triphosphate hydrolase<br />

HGNC HUGO gene nomenclature committee<br />

HIF1 Hypoxia Inducible Factor 1<br />

HIF1A Hypoxia Inducible Factor 1, subunit α<br />

ICM Inner Cell Mass<br />

IDH1 Isocitrate Dehydrogenase 1<br />

IQGAP1 IQ motif containing GTPase activating protein 1<br />

IRES Internal Ribosome Entry Site<br />

KEGG Kyoto Encyclopedia <strong>of</strong> Genes and Genomes<br />

LIF Leukemia Inhibitory Factor<br />

306


Ln Natural logarithm<br />

LOH Loss <strong>of</strong> Heterozygosity<br />

MAP2 Microtubule-associated protein 2<br />

MAPK Mitogen Activated Protein Kinase<br />

MDM2 Mdm2 p53 binding protein homolog<br />

MDM4 Mdm4 p53 binding protein homolog<br />

MGC Mammalian Gene Collection<br />

MGI Mouse Genome Informatics<br />

MGMT O-6-methylguanine-DNA methyltransferase<br />

MIQE Minimum Information for publication <strong>of</strong> Quantitative real-time PCR Experiments<br />

MMR Mismatch Repair<br />

MSH6 MutS homolog 6<br />

NADPH Nicotinamide Adenine Dinucleotide Phosphate<br />

NCBI National Center for Biotechnology Information<br />

NEP Neuroepithelial progenitor<br />

NF1 Neur<strong>of</strong>ibromin 1<br />

NS <strong>Neural</strong> <strong>Stem</strong><br />

OCT4 Octamer-binding protein 4<br />

OLIG2 Oligodendrocyte lineage transcription factor 2<br />

ORF Open Reading Frame<br />

PCA Principal Component Analysis<br />

PDGF Platelet-Derived Growth Factor<br />

PDGFR Platelet-Derived Growth Factor Receptor<br />

PDPK1 3-phosphoinositide dependent protein kinase-1<br />

PHLPP PH domain and leucine rich repeat protein phosphatase 1<br />

PI3K Phosphoinositide-3-Kinase<br />

PI3KR Phosphoinositide-3-Kinase Receptor<br />

PIP3<br />

phosphatidylinositol (3,4,5)-trisphosphate<br />

PTEN Phosphatase and tensin homolog<br />

Pax6 Paired box gene 6<br />

RA Retinoic Acid<br />

RAS Rat Sarcoma<br />

RB1 Retinoblastoma 1<br />

RISC RNA Induced Silencing Complex<br />

RNA Rybonucleic Acid<br />

RTK Receptor Tyrosine Kinase<br />

SAGE Serial Analysis <strong>of</strong> Gene Expression<br />

SCV Squared Coefficient <strong>of</strong> Variation<br />

SGZ Subgranular Zone<br />

SHH Sonic hedge hog<br />

SILAC Stable Isotope Labeling by Amino acids in Cell culture<br />

SKY Spectral Karyotyping<br />

SNP Single Nucleotide Polymorphism<br />

SOX1 Sex determining region Y-box 1<br />

SOX2 Sex determining region Y-box 2<br />

SSEA1 Stage-specific embryonic antigen 1<br />

SVZ Subventricular Zone<br />

TCGA The Cancer Genome Atlas<br />

TGFβ Tumour Growth Factor β<br />

TP53 Tumor Protein 53<br />

TUBB type III β-tubulin<br />

Tag-seq Tag sequencing<br />

UCSC University California Santa Cruz<br />

UTR Untranslated Region<br />

VZ Ventricular Zone<br />

WHO World Health Organization<br />

aCGH array comparative genomics hybridization<br />

bp base pair<br />

iHOP information Hyperlinked Over Proteins<br />

mRNA messenger RNA<br />

mm millimeter<br />

ncRNAs non-coding RNAs<br />

nt nucleotide<br />

oligo-dT oligo deoxy-thymine<br />

poly-A poly-adenine<br />

poly-T poly-thymine<br />

qRT-PCR quantitative Real-Time PCR<br />

rRNA ribosomal RNA<br />

tpm tags per million<br />

UCSC University <strong>of</strong> California Santa Cruz


List <strong>of</strong> Figures<br />

1.1 Estimates <strong>of</strong> survival amongst GBM patients treated with radio-<br />

therapy alone or radiotherapy with the alkylating agent temo-<br />

zolomide. Taken from Stupp et al 2005 [472]. . . . . . . . . . . . 7<br />

1.2 KEGG <strong>Glioma</strong> Pathway. . . . . . . . . . . . . . . . . . . . . . . 23<br />

1.3 Visualisation generated from list <strong>of</strong> 345 interactors (orange) <strong>of</strong><br />

TP53 (yellow) from the BioGRID 3.1 [62] repository for inter-<br />

action datasets. . . . . . . . . . . . . . . . . . . . . . . . . . . . 27<br />

1.4 The Biocarta pathway for Rb signaling. . . . . . . . . . . . . . . 30<br />

2.1 Cross-section through the neural tube. . . . . . . . . . . . . . . 35<br />

2.2 Surface markers <strong>of</strong> radial glia are expressed by NS cell lines,<br />

indicating that these cells may provide the biological context to<br />

work with progenitors <strong>of</strong> the CNS. . . . . . . . . . . . . . . . . . 37<br />

2.3 Sources <strong>of</strong> NS cells: (a) ICM; (b) SVZ. . . . . . . . . . . . . . . 38<br />

2.4 (a,b) Contrast microscopy images <strong>of</strong> early phase neurosphere<br />

formation. (c,d) Immun<strong>of</strong>luorescence microscopy images <strong>of</strong> EGFR<br />

and Nestin detected on an intact neurosphere. . . . . . . . . . . 39<br />

2.5 Schematisation <strong>of</strong> the neurosphere assay used to study neural<br />

precursor cells in culture. . . . . . . . . . . . . . . . . . . . . . . 41<br />

2.6 Diagram <strong>of</strong> the progressive lineage restriction <strong>of</strong> ES cells differ-<br />

entiating toward the neural phenotype. . . . . . . . . . . . . . . 44<br />

2.7 Protocol describing conversion <strong>of</strong> ES cells into immortalised NS<br />

cell lines. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 46<br />

2.8 Representation <strong>of</strong> the Sox1-GFP reporter construct used in the<br />

niche-independent NS cell protocol. . . . . . . . . . . . . . . . . 46<br />

2.9 Roles <strong>of</strong> EGF and FGF2 in the derivation and maintenance <strong>of</strong><br />

NS cells. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 52<br />

3.1 <strong>Stem</strong> cell differentiation hierarchy. . . . . . . . . . . . . . . . . . 58<br />

3.2 Diagram <strong>of</strong> asymmetric and symmetric cell division. . . . . . . . 59<br />

308


3.3 Schematisation <strong>of</strong> cell cycle phases. . . . . . . . . . . . . . . . . 67<br />

3.4 Glioblastoma treatment with ionizing radiation. . . . . . . . . . 68<br />

3.5 Glioblastoma treatment with BMPs. . . . . . . . . . . . . . . . 70<br />

3.6 PCA diagram <strong>of</strong> global mRNA expression in GNS cell lines. . . 82<br />

4.1 Classes <strong>of</strong> non-coding RNAs discovered to date. . . . . . . . . . 86<br />

5.1 Schematisation <strong>of</strong> the longSAGE protocol. . . . . . . . . . . . . 95<br />

5.2 Boxplot <strong>of</strong> normalised Ct values. . . . . . . . . . . . . . . . . . . 102<br />

5.3 Correlation scatter plot <strong>of</strong> the raw Ct values vs endogenous<br />

controls. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 103<br />

5.4 Scatter plots <strong>of</strong> our A and B biological replicates. . . . . . . . . 106<br />

5.5 Dot plot <strong>of</strong> the standard deviations <strong>of</strong> the differences between<br />

expression levels in two replicates. . . . . . . . . . . . . . . . . . 107<br />

5.6 Literature mining diagram <strong>of</strong> code functions. . . . . . . . . . . . 109<br />

5.7 Schematisation <strong>of</strong> a typical Cytoscape network. . . . . . . . . . 115<br />

6.1 Sequencing construct schematisation. . . . . . . . . . . . . . . . 121<br />

6.2 Diagram <strong>of</strong> the extraction, filtering and mapping phases for<br />

reads and tags. . . . . . . . . . . . . . . . . . . . . . . . . . . . 122<br />

6.3 Pie charts for the proportion <strong>of</strong> filtered tags. . . . . . . . . . . . 123<br />

6.4 Diagram <strong>of</strong> the tag mapping strategy. . . . . . . . . . . . . . . . 124<br />

6.5 Pie charts for the assignment <strong>of</strong> tags to genes. . . . . . . . . . . 126<br />

6.6 Correlations for all combinations <strong>of</strong> cell lines. . . . . . . . . . . . 127<br />

6.7 CGH array analysis. . . . . . . . . . . . . . . . . . . . . . . . . 128<br />

6.8 Curves show distributions <strong>of</strong> expression level differences between<br />

GNS and NS lines. . . . . . . . . . . . . . . . . . . . . . . . . . 129<br />

6.9 Plot <strong>of</strong> the estimates <strong>of</strong> the variance against the base levels for<br />

each gene. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 130<br />

6.10 Plot <strong>of</strong> the fold change versus the mean for normal vs tumour<br />

samples. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 131<br />

6.11 Tag mapping <strong>of</strong> NTRK2 on UCSC genome browser. . . . . . . . 135<br />

6.12 Expression estimates correlate well between Tag-seq and qRT-<br />

PCR. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 138<br />

6.13 Heatmap <strong>of</strong> 29 genes differentially expressed between 16 GNS<br />

and 6 NS cell lines . . . . . . . . . . . . . . . . . . . . . . . . . 138<br />

6.14 Expression levels <strong>of</strong> the 29 genes distinguishing GNS from NS<br />

lines as percent <strong>of</strong> NS geometric mean. . . . . . . . . . . . . . . 142<br />

6.15 Tag mapping <strong>of</strong> BMP7 on UCSC genome browser. . . . . . . . . 148


6.16 Tag mapping <strong>of</strong> TPM1 on UCSC genome browser. . . . . . . . . 149<br />

6.17 Schematisation <strong>of</strong> the process <strong>of</strong> finding genes with differentially<br />

expressed is<strong>of</strong>orms. . . . . . . . . . . . . . . . . . . . . . . . . . 150<br />

6.18 Schematisation <strong>of</strong> the localisation <strong>of</strong> the microRNA seeds on the<br />

mapped is<strong>of</strong>orms. . . . . . . . . . . . . . . . . . . . . . . . . . . 151<br />

6.19 Is<strong>of</strong>orm detection by multi tag mapping <strong>of</strong> gene GAPVD1 on<br />

UCSC genome browser. . . . . . . . . . . . . . . . . . . . . . . . 154<br />

6.20 Is<strong>of</strong>orm detection by multi tag mapping <strong>of</strong> gene SMAD1 on<br />

UCSC genome browser. . . . . . . . . . . . . . . . . . . . . . . . 156<br />

6.21 Correlated expression <strong>of</strong> CTSC and a nearby ncRNA. . . . . . . 159<br />

6.22 Histogram <strong>of</strong> expression levels <strong>of</strong> HOTAIRM1 and surrounding<br />

HOX genes. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 160<br />

7.1 Enrichment plots for the top two pathways revealed through<br />

GSEA <strong>of</strong> the KEGG database <strong>of</strong> pathways [2]. . . . . . . . . . . 165<br />

7.2 GSEA plots <strong>of</strong> (a) nominal p-values vs normalised enrichment<br />

score and (b) line graph <strong>of</strong> the enrichment scores across pathways.165<br />

7.3 Correlation <strong>of</strong> Tag-seq interrogated cell lines with glioblastoma<br />

subtype expression signatures. . . . . . . . . . . . . . . . . . . . 170<br />

7.4 Core gene expression changes in GNS lines are mirrored in<br />

glioblastoma tumours. . . . . . . . . . . . . . . . . . . . . . . . 172<br />

7.5 Association between GNS signature and other survival predictors.184<br />

7.6 Association between GNS signature score and patient survival. . 185<br />

7.7 The integrated glioblastoma pathway subdivided into sections<br />

identifying the gene networks that participate in the pathway. . 188<br />

7.8 Integrated GBM pathway used to overlay the Tag-seq GNS<br />

dataset. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 191<br />

7.9 Affected p53, RB1 and PTEN/PI3K pathways. . . . . . . . . . 192<br />

7.10 Four integrated GBM pathways overlaid with Tag-seq expres-<br />

sion level measures for each GNS cell line. . . . . . . . . . . . . 194<br />

7.11 Integrated GBM pathway with the TCGA dataset overlaid. . . . 199<br />

7.12 Integrated GBM pathway with the HGG dataset overlaid. . . . 200<br />

7.13 Three integrated GBM pathways overlaid with the GNS, TCGA<br />

and HGG datasets. . . . . . . . . . . . . . . . . . . . . . . . . . 201<br />

8.1 Overview <strong>of</strong> GenemiR s<strong>of</strong>tware. . . . . . . . . . . . . . . . . . . 205<br />

8.2 Internal organisation <strong>of</strong> the target prediction database <strong>of</strong> Gen-<br />

emiR. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 206


8.3 Workflows at the core <strong>of</strong> the primitive functions <strong>of</strong> the GenemiR<br />

s<strong>of</strong>tware. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 207<br />

8.4 Step by step diagram <strong>of</strong> the ensemble method adopted to find<br />

the score E (=C2/C1) <strong>of</strong> prediction accuracy for prediction al-<br />

gorithms. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 214<br />

D.1 Integrated GBM pathway with G144 cell line Tag-seq expres-<br />

sion data. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 282<br />

D.2 Integrated GBM pathway with G144ED cell line Tag-seq ex-<br />

pression data. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 283<br />

D.3 Integrated GBM pathway with G166 cell line Tag-seq expres-<br />

sion data. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 284<br />

D.4 Integrated GBM pathway with G179 cell line Tag-seq expres-<br />

sion data. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 285


List <strong>of</strong> Tables<br />

1.1 Histological Types and Prognosis <strong>of</strong> <strong>Glioma</strong>s (y, years). Taken<br />

from Doyle et al 2005 [128]. . . . . . . . . . . . . . . . . . . . . 6<br />

2.1 Summary <strong>of</strong> commonalities and differences between ES cells and<br />

NS cells. Adapted from Pollard et al 2006 [399] . . . . . . . . . 53<br />

3.1 Summary <strong>of</strong> characteristics <strong>of</strong> NBE and serum-cultured glioblas-<br />

toma cells. Adapted from Lee et al 2006. . . . . . . . . . . . . . 77<br />

5.1 Summary <strong>of</strong> cell lines investigated with Tag-seq. . . . . . . . . . 95<br />

5.2 Classification <strong>of</strong> sequenced tags in each cell line. . . . . . . . . . 99<br />

5.3 Summary <strong>of</strong> statistics using the χ 2 and logarithmic tests. . . . . 111<br />

5.4 Public gene expression datasets used in thesis. . . . . . . . . . . 113<br />

6.1 Summary <strong>of</strong> the available clinical data for our GNS cell lines. . . 121<br />

6.2 Summary <strong>of</strong> reads per cell line library. . . . . . . . . . . . . . . 121<br />

6.3 Significance <strong>of</strong> the correlation found between CNAs and expres-<br />

sion levels measured with Fisher’s exact test (p-value). . . . . . 128<br />

6.4 Table <strong>of</strong> genes with large expression changes common to the<br />

GNS cell lines. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 132<br />

6.5 Differentially expressed genes assigned to a four-tier classifica-<br />

tion system. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 143<br />

6.6 Genes with limited or no evidence <strong>of</strong> implication in glioblastoma<br />

that appear in our pathway. . . . . . . . . . . . . . . . . . . . . 145<br />

6.7 Summary <strong>of</strong> predicted microRNAs targeting differentially ex-<br />

pressed is<strong>of</strong>orms. . . . . . . . . . . . . . . . . . . . . . . . . . . 151<br />

6.8 MicroRNA array results for GNS cell lines with respect to NS<br />

cell lines. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 152<br />

6.9 Differentially expressed ncRNAs. . . . . . . . . . . . . . . . . . 157<br />

312


7.1 Selected Gene Ontology terms and InterPro domains enriched<br />

among differentially expressed genes. . . . . . . . . . . . . . . . 163<br />

7.2 Representative KEGG pathways from signaling pathway impact<br />

analysis <strong>of</strong> gene expression differences between GNS and NS lines.164<br />

7.3 Summary <strong>of</strong> all MHC class I and II genes. . . . . . . . . . . . . 167<br />

7.4 Literature survey for the 29 genes found to distinguish GNS<br />

from NS lines across a panel <strong>of</strong> 21 cell lines . . . . . . . . . . . . 175<br />

7.5 Survival tests for the 29 genes found via qRT-PCR to distinguish<br />

GNS cell lines from NS cell lines. . . . . . . . . . . . . . . . . . 183<br />

7.6 Significance <strong>of</strong> survival association for GNS signature and IDH1<br />

status. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 186<br />

7.7 Node assignment in the glioblastoma pathway. . . . . . . . . . . 190<br />

8.1 microRNA target prediction algorithms used by GenemiR with<br />

number <strong>of</strong> microRNA:3'UTR interactions predicted. The origi-<br />

nal target identifiers refer to the identifiers used by a prediction<br />

algorithm to identify the targeted genes. The final target iden-<br />

tifiers refer to the identifiers that are returned by any query <strong>of</strong><br />

any prediction algorithm database. . . . . . . . . . . . . . . . . 209<br />

8.2 Single prediction algorithm ensemble analysis results. Displayed<br />

in descending order <strong>of</strong> E-score. . . . . . . . . . . . . . . . . . . . 215<br />

8.3 All combinations <strong>of</strong> prediction algorithms in descending order<br />

<strong>of</strong> E-score. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 216<br />

A.1 Classification <strong>of</strong> differentially expressed genes at 10% FDR. . . . 235<br />

A.2 Classification <strong>of</strong> differentially expressed genes based on litera-<br />

ture mining analysis. . . . . . . . . . . . . . . . . . . . . . . . . 251<br />

A.3 Raw Ct values. Abbreviations: "down" for down-regulated,<br />

"up" for up-regulated, and "Norm" for Normalisation control. . 257<br />

A.4 Normalised Ct values. Abbreviations: "down" for down-regulated,<br />

"up" for up-regulated, and "Norm" for Normalisation control. . 261<br />

A.5 Pearson correlation values between the normalised Ct values<br />

measured through qRT-PCR and the tag counts measured across<br />

the five GNS and NS cell lines assayed via Tag-seq. . . . . . . . 265<br />

C.1 Differentially expressed non-coding RNAs. . . . . . . . . . . . . 274<br />

D.1 GBM pathway interaction data. . . . . . . . . . . . . . . . . . . 276


E.1 Fold-changes measured by exon array for GNS cell lines. . . . . 286<br />

F.1 Differentially expressed microRNAs in GNS vs NS cell lines at<br />

FDR


Bibliography<br />

[1] Cell signaling technology. MAPK-ERK signaling cascade.<br />

http://www.cellsignal.com/reference/pathway/mapk_erk_growth.html.<br />

[2] Kegg pathways. http://www.genome.jp/kegg/pathway.html.<br />

[3] Kyoto encyclopedia <strong>of</strong> genes and genomes. <strong>Glioma</strong> pathway.<br />

http://www.genome.jp/kegg-bin/_pathway?hsa05214.<br />

[4] Kyoto encyclopedia <strong>of</strong> genes and genomes. P53 pathway.<br />

http://www.genome.jp/kegg/pathway/hsa/hsa04115.html.<br />

[5] Panter pathway. P53 pathway. http://www.pantherdb.org/pathway/pathwaydiagram.<br />

[6] S. Acharya, T. Wilson, S. Gradia, M. F. Kane, S. Guerrette, G. T. Marsischky,<br />

R. Kolodner, and R. Fishel. hMSH2 forms specific mispair-binding complexes with<br />

hMSH3 and hMSH6. Proceedings <strong>of</strong> the National Academy <strong>of</strong> Sciences <strong>of</strong> the United<br />

States <strong>of</strong> America, 93(24):13629–13634, Nov. 1996.<br />

[7] M. Adamowicz, B. Radlwimmer, R. Rieker, D. Mertens, M. Schwarzbach, P. Schraml,<br />

A. Benner, P. Lichter, G. Mechtersheimer, and S. Joos. Frequent amplifications and<br />

abundant expression <strong>of</strong> trio, nkd2, and irx2 in s<strong>of</strong>t tissue sarcomas. Genes, Chromosomes<br />

and Cancer, 45(9):829–838, 2006.<br />

[8] D. Adams, B. Hasson, A. Boyer-Boiteau, A. El-Khishin, and V. Shashoua. A peptide<br />

fragment <strong>of</strong> ependymin neurotrophic factor uses protein kinase c and the mitogenactivated<br />

protein kinase pathway to activate c-jun n-terminal kinase and a functional<br />

ap-1 containing c-jun and c-fos proteins in mouse nb2a cells. Journal <strong>of</strong> neuroscience<br />

research, 72(3):405–416, 2003.<br />

[9] A. Adesina, Y. Nguyen, V. Mehta, H. Takei, P. Stangeby, S. Crabtree, M. Chintagumpala,<br />

and M. Gumerlock. Foxg1 dysregulation is a frequent event in medulloblastoma.<br />

Journal <strong>of</strong> neuro-oncology, 85(2):111–122, 2007.<br />

[10] C. Agulhon, J. Petravicz, A. McMullen, E. Sweger, S. Minton, S. Taves, K. Casper,<br />

T. Fiacco, and K. McCarthy. What is the role <strong>of</strong> astrocyte calcium in neurophysiology?<br />

Neuron, 59(6):932–946, 2008.<br />

[11] E. Ah Cho and G. Dressler. Tcf-4 binds [beta]-catenin and is expressed in distinct<br />

regions <strong>of</strong> the embryonic brain and limbs. Mechanisms <strong>of</strong> development, 77(1):9–18,<br />

1998.<br />

[12] N. Ahuja, Q. Li, A. Mohan, S. Baylin, and J. Issa. Aging and dna methylation in<br />

colorectal mucosa and cancer. Cancer research, 58(23):5489–5494, 1998.<br />

[13] N. Ahuja, Q. Li, A. Mohan, S. Baylin, and J. Issa. Aging and dna methylation in<br />

colorectal mucosa and cancer. Cancer research, 58(23):5489, 1998.<br />

[14] T. Akai, Y. Ueda, Y. Sasagawa, T. Hamada, T. Date, S. Katsuda, H. Iizuka, Y. Okada,<br />

and K. Chada. High mobility group ic protein in astrocytoma and glioblastoma.<br />

Pathology-Research and Practice, 200(9):619–624, 2004.<br />

[15] M. Al-Hajj, M. Wicha, A. Benito-Hernandez, S. Morrison, and M. Clarke. Prospective<br />

identification <strong>of</strong> tumorigenic breast cancer cells. Proceedings <strong>of</strong> the National Academy<br />

<strong>of</strong> Sciences, 100(7):3983, 2003.<br />

[16] P. Alexiou, M. Maragkakis, G. Papadopoulos, M. Reczko, and A. Hatzigeorgiou. Lost<br />

in translation: an assessment and perspective for computational microrna target identification.<br />

Bioinformatics, 25(23):3049, 2009.<br />

315


[17] N. Allen and B. Barres. Signaling between glia and neurons: focus on synaptic plasticity.<br />

Current opinion in neurobiology, 15(5):542–548, 2005.<br />

[18] A. Alvarez-Buylla and D. Lim. For the Long Run: Maintaining Germinal Niches in<br />

the Adult Brain. Neuron, 41(5):683–686, 2004.<br />

[19] V. Ambros and X. Chen. The regulation <strong>of</strong> genes and genomes by small RNAs.<br />

Development, 134(9):1635–41, 2007.<br />

[20] M. Amiry-Moghaddam and O. Ottersen. The molecular basis <strong>of</strong> water transport in<br />

the brain. Nature Reviews Neuroscience, 4(12):991–1001, 2003.<br />

[21] S. Anders and W. Huber. Differential expression analysis for sequence count data.<br />

Genome Biol, 11(10):R106, 2010.<br />

[22] P. Andrews. Retinoic acid induces neuronal differentiation <strong>of</strong> a cloned human embryonal<br />

carcinoma cell line in vitro* 1. Developmental biology, 103(2):285–293, 1984.<br />

[23] A. Aravin, D. Gaidatzis, S. Pfeffer’t, M. Lagos-Quintana, P. Landgraf, and T. Tuschl.<br />

A novel class <strong>of</strong> small rnas bind to mili protein in mouse testes. Nature, 442:203–207,<br />

2006.<br />

[24] A. Aravin, N. Naumova, A. Tulin, V. Vagin, Y. Rozovsky, and V. Gvozdev. Doublestranded<br />

rna-mediated silencing <strong>of</strong> genomic tandem repeats and transposable elements<br />

in the d. melanogaster germline. Current Biology, 11(13):1017–1027, 2001.<br />

[25] K. Archer, V. Mas, K. David, D. Maluf, K. Bornstein, and R. Fisher. Identifying genes<br />

for establishing a multigenic test for hepatocellular carcinoma surveillance in hepatitis<br />

c virus-positive cirrhotic patients. Cancer Epidemiology Biomarkers & Prevention,<br />

18(11):2929–2932, 2009.<br />

[26] M. Arpin, E. Friederich, M. Algrain, F. Vernel, and D. Louvard. Functional differences<br />

between l-and t-plastin is<strong>of</strong>orms. The Journal <strong>of</strong> cell biology, 127(6):1995, 1994.<br />

[27] M. Arpin, E. Friederich, M. Algrain, F. Vernel, and D. Louvard. Functional differences<br />

between l-and t-plastin is<strong>of</strong>orms. The Journal <strong>of</strong> cell biology, 127(6):1995–2008, 1994.<br />

[28] ArrayExpress. www.ebi.ac.uk/arrayexpress.<br />

[29] M. Assimakopoulou, M. Kondyli, G. Gatzounis, T. Maraziotis, and J. Varakis. Neurotrophin<br />

receptors expression and jnk pathway activation in human astrocytomas.<br />

BMC cancer, 7(1):202, 2007.<br />

[30] S. Assinder, J. Stanton, and P. Prasad. Transgelin: an actin-binding protein and tumour<br />

suppressor. The international journal <strong>of</strong> biochemistry & cell biology, 41(3):482–<br />

486, 2009.<br />

[31] P. Au, Q. Zhu, E. Dennis, and M. Wang. Long non-coding rna-mediated mechanisms<br />

independent <strong>of</strong> the rnai pathway in animals and plants. RNA biology, 8(3), 2011.<br />

[32] J. Aubert, M. Stavridis, S. Tweedie, M. O’Reilly, K. Vierlinger, M. Li, P. Ghazal,<br />

T. Pratt, J. Mason, D. Roy, et al. Screening for mammalian neural genes via<br />

fluorescence-activated cell sorter purification <strong>of</strong> neural precursors from sox1-gfp knockin<br />

mice. Proceedings <strong>of</strong> the National Academy <strong>of</strong> Sciences <strong>of</strong> the United States <strong>of</strong><br />

America, 100(Suppl 1):11836, 2003.<br />

[33] F. Azevedo, L. Carvalho, L. Grinberg, J. Farfel, R. El Ferreti, R. Leite, W. Jacob filho,<br />

R. lent, and S. herculano houzel. Equal numbers <strong>of</strong> neuronal and non-neuronal cells<br />

make the human brain an isometrically scaled-up primate brain. brain, 513:532–541,<br />

2009.<br />

[34] H. Babu, G. Cheung, H. Kettenmann, T. Palmer, and G. Kempermann. Enriched<br />

monolayer precursor cell cultures from micro-dissected adult mouse dentate gyrus<br />

yield functional granule cell-like neurons. PLoS One, 2(4):e388, 2007.<br />

[35] A. Bader, S. Kang, and P. Vogt. Cancer-specific mutations in PIK3CA are oncogenic<br />

in vivo. Proceedings <strong>of</strong> the National Academy <strong>of</strong> Sciences <strong>of</strong> the United States <strong>of</strong><br />

America, 103(5):1475, 2006.<br />

[36] A. Bader, S. Kang, L. Zhao, and P. Vogt. Oncogenic PI3K deregulates transcription<br />

and translation. Nature reviews cancer, 5(12):921–929, 2005.<br />

[37] D. Baek, J. Villen, C. Shin, F. D. Camargo, S. P. Gygi, and D. P. Bartel. The impact<br />

<strong>of</strong> microRNAs on protein output. Nature, 455(7209):64–71, 2008.


[38] G. Bain, D. Kitchens, M. Yao, J. Huettner, and D. Gottlieb. Embryonic stem cells<br />

express neuronal properties in vitro. Developmental biology, 168(2):342–357, 1995.<br />

[39] L. Balenci. IQGAP1 Protein Specifies Amplifying Cancer <strong>Cells</strong> in Glioblastoma Multiforme.<br />

Cancer Research, 66(18):9074–9082, Sept. 2006.<br />

[40] S. Bao, Q. Wu, R. McLendon, Y. Hao, Q. Shi, A. Hjelmeland, M. Dewhirst, D. Bigner,<br />

and J. Rich. <strong>Glioma</strong> stem cells promote radioresistance by preferential activation <strong>of</strong><br />

the DNA damage response. Nature, 444(7120):756–760, 2006.<br />

[41] I. Barani, S. Benedict, and P. Lin. <strong>Neural</strong> stem cells: implications for the conventional<br />

radiotherapy <strong>of</strong> central nervous system malignancies. International Journal <strong>of</strong><br />

Radiation Oncology* Biology* Physics, 68(2):324–333, 2007.<br />

[42] J. Barnes and P. Hut. A hierarchical 0 (n log iv) force-calculation algorithm. nature,<br />

324:4, 1986.<br />

[43] D. P. Bartel. MicroRNAs: target recognition and regulatory functions. Cell,<br />

136(2):215–33, 2009.<br />

[44] N. Baumann. Biology <strong>of</strong> oligodendrocyte and myelin in the mammalian central nervous<br />

system. Physiological Reviews, 2001.<br />

[45] A. Bellacosa, C. C. Kumar, A. Di Crist<strong>of</strong>ano, and J. R. Testa. Activation <strong>of</strong> AKT<br />

kinases in cancer: implications for therapeutic targeting. Advances in cancer research,<br />

94:29–86, 2005.<br />

[46] A. Bellacosa, J. R. Testa, R. Moore, and L. Larue. A portrait <strong>of</strong> AKT kinases: human<br />

cancer and animal models depict a family with strong individualities. Cancer biology<br />

& therapy, 3(3):268–275, Mar. 2004.<br />

[47] D. R. Bentley. Whole-genome re-sequencing. Current opinion in genetics and development,<br />

16(6):545–52, 2006.<br />

[48] I. Bentwich. Prediction and validation <strong>of</strong> micrornas and their targets. FEBS letters,<br />

579(26):5904–5910, 2005.<br />

[49] R. Berg, E. Leung, S. Gough, C. Morris, W. Yao, S. Wang, J. Ni, and G. Krissansen.<br />

Cloning and characterization <strong>of</strong> a novel-beta integrin-related cdna coding for the protein<br />

tied. Genomics, 56(2):169–178, 1999.<br />

[50] R. Berg, E. Leung, S. Gough, C. Morris, W. Yao, S. Wang, J. Ni, and G. Krissansen.<br />

Cloning and characterization <strong>of</strong> a novel β integrin-related cdna coding for the protein<br />

tied (ÂŞten β integrin egf-like repeat domainsÂŤ) that maps to chromosome band<br />

13q33: a divergent stand-alone integrin stalk structure. Genomics, 56(2):169–178,<br />

1999.<br />

[51] A. Bergamaschi, Y. Kim, K. Kwei, Y. La Choi, M. Bocanegra, A. Langerød, W. Han,<br />

D. Noh, D. Huntsman, S. Jeffrey, et al. Camk1d amplification implicated in epithelialmesenchymal<br />

transition in basal-like breast cancer. Molecular oncology, 2(4):327–339,<br />

2008.<br />

[52] R. Beroukhim, G. Getz, L. Nghiemphu, J. Barretina, T. Hsueh, D. Linhart, I. Vivanco,<br />

J. Lee, J. Huang, S. Alexander, et al. Assessing the significance <strong>of</strong> chromosomal<br />

aberrations in cancer: methodology and application to glioma. Proceedings <strong>of</strong> the<br />

National Academy <strong>of</strong> Sciences, 104(50):20007–20012, 2007.<br />

[53] M. Berry, Z. Ahmed, B. Lorber, M. Douglas, and A. Logan. Regeneration <strong>of</strong> axons in<br />

the visual system. Restorative neurology and neuroscience, 26(2):147–174, 2008.<br />

[54] F. Bertucci, P. Finetti, N. Cervera, E. Charafe-Jauffret, E. Mamessier, J. Adélaïde,<br />

S. Debono, G. Houvenaeghel, D. Maraninchi, P. Viens, et al. Gene expression pr<strong>of</strong>iling<br />

shows medullary breast cancer is a subgroup <strong>of</strong> basal breast cancers. Cancer Research,<br />

66(9):4636–4644, 2006.<br />

[55] M. Bibel, J. Richter, K. Schrenk, K. Tucker, V. Staiger, M. Korte, M. Goetz, and<br />

Y. Barde. Differentiation <strong>of</strong> mouse embryonic stem cells into a defined neuronal lineage.<br />

Nature neuroscience, 7(9):1003–1009, 2004.<br />

[56] L. Biesecker. Exome sequencing makes medical genomics a reality. Nature genetics,<br />

42(1):13, 2010.


[57] E. Bindewald and B. Shapiro. Rna secondary structure prediction from sequence<br />

alignments using a network <strong>of</strong> k-nearest neighbor classifiers. Rna, 12(3):342–352,<br />

2006.<br />

[58] Biocarta. P53 signaling pathway. http://www.biocarta.com/pathfiles/h_p53pathway.asp.<br />

[59] Biocarta. Pten dependent cell cycle arrest and apoptosis.<br />

http://www.biocarta.com/pathfiles/h_ptenpathway.asp.<br />

[60] Biocarta. Rb tumor suppressor/checkpoint signaling in response to dna damage.<br />

http://www.biocarta.com/pathfiles/h_rbpathway.asp.<br />

[61] Biocarta. www.biocarta.com.<br />

[62] BioGRID. Database <strong>of</strong> protein and genetic interactions. www.thebiogrid.org.<br />

[63] B. Boëda, D. Briggs, T. Higgins, B. Garvalov, A. Fadden, N. McDonald, and M. Way.<br />

Tes, a specific mena interacting partner, breaks the rules for evh1 binding. Molecular<br />

cell, 28(6):1071–1082, 2007.<br />

[64] S. Bonnet, S. Archer, J. Allalunis-Turner, A. Haromy, C. Beaulieu, R. Thompson,<br />

C. Lee, G. Lopaschuk, L. Puttagunta, S. Bonnet, et al. A mitochondria-k+ channel<br />

axis is suppressed in cancer and its normalization promotes apoptosis and inhibits<br />

cancer growth. Cancer cell, 11(1):37–51, 2007.<br />

[65] K. Boon, E. Osório, S. Greenhut, C. Schaefer, J. Shoemaker, K. Polyak, P. Morin,<br />

K. Buetow, R. Strausberg, S. De Souza, et al. An anatomy <strong>of</strong> normal and malignant<br />

gene expression. Proceedings <strong>of</strong> the National Academy <strong>of</strong> Sciences, 99(17):11287, 2002.<br />

[66] B. Borrell. How accurate are cancer cell lines? Nature, 463(7283):858, Feb. 2010.<br />

[67] P. Bos, X. Zhang, C. Nadal, W. Shu, R. Gomis, D. Nguyen, A. Minn, M. van de<br />

Vijver, W. Gerald, J. Foekens, et al. Genes that mediate breast cancer metastasis to<br />

the brain. Nature, 459(7249):1005–1009, 2009.<br />

[68] R. Bourgo, U. Ehmer, J. Sage, and E. Knudsen. RB deletion disrupts coordination<br />

between dna replication licensing and mitotic entry in vivo. Molecular biology <strong>of</strong> the<br />

cell, 22(7):931, 2011.<br />

[69] C. Brennan, H. Momota, D. Hambardzumyan, T. Ozawa, A. Tandon, A. Pedraza,<br />

and E. Holland. Glioblastoma subclasses can be defined by activity among signal<br />

transduction pathways and associated genomic alterations. PLoS ONE, 4(11):e7752,<br />

2009.<br />

[70] J. Brennecke, D. R. Hipfner, A. Stark, R. B. Russell, and S. M. Cohen. bantam<br />

encodes a developmentally regulated microRNA that controls cell proliferation and<br />

regulates the proapoptotic gene hid in Drosophila. Cell, 113(1):25–36, 2003.<br />

[71] J. Brennecke, A. Stark, R. B. Russell, and S. M. Cohen. Principles <strong>of</strong> microRNA-target<br />

recognition. PLoS Biol, 3(3):e85, 2005.<br />

[72] J. Briscoe and J. Ericson. Specification <strong>of</strong> neuronal fates in the ventral neural tube.<br />

Current opinion in neurobiology, 11(1):43–49, 2001.<br />

[73] P. Brodal. The Central Nervous System: Structure and Function. Oxford Univ Pr,<br />

2010.<br />

[74] J. Brognard, E. Sierecki, T. Gao, and A. C. Newton. PHLPP and a second is<strong>of</strong>orm,<br />

PHLPP2, differentially attenuate the amplitude <strong>of</strong> Akt signaling by regulating distinct<br />

Akt is<strong>of</strong>orms. Molecular cell, 25(6):917–931, Mar. 2007.<br />

[75] K. Brown, D. Strathdee, S. Bryson, W. Lambie, and A. Balmain. The malignant<br />

capacity <strong>of</strong> skin tumours induced by expression <strong>of</strong> a mutant h-ras transgene depends<br />

on the cell type targeted. Current biology, 8(9):516–524, 1998.<br />

[76] O. Brustle, K. Jones, R. Learish, K. Karram, K. Choudhary, O. Wiestler, I. Duncan,<br />

and R. McKay. Embryonic stem cell-derived glial precursors: A source <strong>of</strong> myelinating<br />

transplants. Science, 285(5428):754–756, 1999.<br />

[77] J. Buckner, P. Brown, and B. O’Neill. Central nervous system tumors. Mayo Clinic<br />

Proceedings, 2007.


[78] S. Bustin, V. Benes, J. Garson, J. Hellemans, J. Huggett, M. Kubista, R. Mueller,<br />

T. Nolan, M. Pfaffl, G. Shipley, et al. The miqe guidelines: minimum information for<br />

publication <strong>of</strong> quantitative real-time pcr experiments. Clinical chemistry, 55(4):611–<br />

622, 2009.<br />

[79] D. Cahill, K. Levine, R. Betensky, P. Codd, C. Romany, L. Reavie, T. Batchelor,<br />

P. Futreal, M. Stratton, W. Curry, et al. Loss <strong>of</strong> the mismatch repair protein MSH6<br />

in human glioblastomas is associated with tumor progression during temozolomide<br />

treatment. Clinical cancer research, 13(7):2038, 2007.<br />

[80] I. Camby, N. Nagy, M. Lopes, B. Schäfer, C. Maurage, M. Ruchoux, P. Murmann,<br />

R. Pochet, C. Heizmann, J. Brotchi, et al. Supratentorial pilocytic astrocytomas,<br />

astrocytomas, anaplastic astrocytomas and glioblastomas are characterized by a differential<br />

expression <strong>of</strong> s100 proteins. Brain pathology, 9(1):1–19, 1999.<br />

[81] M. Carlén, K. Meletis, C. Göritz, V. Darsalia, E. Evergren, K. Tanigaki, M. Amendola,<br />

F. Barnabé-Heider, M. S. Y. Yeung, L. Naldini, T. Honjo, Z. Kokaia, O. Shupliakov,<br />

R. M. Cassidy, O. Lindvall, and J. Frisén. Forebrain ependymal cells are Notchdependent<br />

and generate neuroblasts and astrocytes after stroke. Nature neuroscience,<br />

12(3):259–267, Mar. 2009.<br />

[82] E. Carpenter, J. Goddard, A. Davis, T. Nguyen, and M. Capecchi. Targeted disruption<br />

<strong>of</strong> hoxd-10 affects mouse hindlimb development. Development, 124(22):4505–4514,<br />

1997.<br />

[83] M. Carpenter, X. Cui, Z. Hu, J. Jackson, S. Sherman, Å. Seiger, and L. Wahlberg. In<br />

vitro expansion <strong>of</strong> a multipotent population <strong>of</strong> human neural progenitor cells. Experimental<br />

neurology, 158(2):265–278, 1999.<br />

[84] A. Carracedo, A. Alimonti, and P. P. Pandolfi. PTEN Level in Tumor Suppression:<br />

How Much Is Too Little? Cancer Research, 71(3):629–633, Feb. 2011.<br />

[85] M. Carro, W. Lim, M. Alvarez, R. Bollo, X. Zhao, E. Snyder, E. Sulman, S. Anne,<br />

F. Doetsch, H. Colman, et al. The transcriptional network for mesenchymal transformation<br />

<strong>of</strong> brain tumours. Nature, 463(7279):318–325, 2009.<br />

[86] E. Cerami, E. Demir, N. Schultz, and B. Taylor. Automated network analysis identifies<br />

core pathways in glioblastoma. PLoS ONE, 2010.<br />

[87] S. Certain, F. Barrat, E. Pastural, F. Le Deist, J. Goyo-Rivas, N. Jabado, M. Benkerrou,<br />

R. Seger, E. Vilmer, G. Beullier, et al. Protein truncation test <strong>of</strong> lyst reveals<br />

heterogenous mutations in patients with chediak-higashi syndrome. Blood, 95(3):979–<br />

983, 2000.<br />

[88] V. Chabottaux, S. Ricaud, L. Host, S. Blacher, A. Paye, M. Thiry, A. Gar<strong>of</strong>alakis,<br />

C. Pestourie, K. Gombert, F. Bruyere, et al. Membrane-type 4 matrix metalloproteinase<br />

(mt4-mmp) induces lung metastasis by alteration <strong>of</strong> primary breast tumour<br />

vascular architecture. Journal <strong>of</strong> cellular and molecular medicine, 13(9b):4002–4013,<br />

2009.<br />

[89] K. Chan, I. Espinosa, M. Chao, D. Wong, L. Ailles, M. Diehn, H. Gill, J. Presti,<br />

H. Chang, M. Van De Rijn, et al. Identification, molecular characterization, clinical<br />

prognosis, and therapeutic targeting <strong>of</strong> human bladder tumor-initiating cells. Proceedings<br />

<strong>of</strong> the National Academy <strong>of</strong> Sciences, 106(33):14016–14021, 2009.<br />

[90] C. Cheadle, M. Nesterova, T. Watkins, K. Barnes, J. Hall, A. Rosen, K. Becker,<br />

and Y. Cho-Chung. Regulatory subunits <strong>of</strong> pka define an axis <strong>of</strong> cellular proliferation/differentiation<br />

in ovarian cancer cells. BMC Medical Genomics, 1(1):43, 2008.<br />

[91] K. Chen and N. Rajewsky. Deep conservation <strong>of</strong> microRNA-target relationships and<br />

3’UTR motifs in vertebrates, flies, and nematodes. Cold Spring Harb Symp Quant<br />

Biol, 71:149–56, 2006.<br />

[92] L. Z. L. Y. Z. Q. Chen K, Luo Z. Perp gene therapy attenuates lung cancer xenograft<br />

via inducing apoptosis and suppressing vegf. Cancer biology and therapy, 12, 2011.<br />

[93] A. J. S. T. S. C. B.-N. S. S. J. M. R. M. R. S. S. Q. H. P. J. T. A. R. T. L. W. S.<br />

K. C. J. B. C. M. M. G. R. H. D. Cheung KâĂŘJJ, Johnson NA. Acquired tnfrsf14<br />

mutations in follicular lymphoma are associated with worse prognosis. Cancer Res,<br />

70:9166–9174, 2010.


[94] F. Chibon, O. Mariani, J. Derré, A. Mairal, J. Coindre, L. Guillou, X. Sastre, F. Pédeutour,<br />

and A. Aurias. Ask1 (map3k5) as a potential therapeutic target in malignant<br />

fibrous histiocytomas with 12q14–q15 and 6q23 amplifications. Genes, Chromosomes<br />

and Cancer, 40(1):32–37, 2004.<br />

[95] S. Chigurupati, R. Venkataraman, D. Barrera, A. Naganathan, M. Madan, L. Paul,<br />

J. Pattisapu, G. Kyriazis, K. Sugaya, S. Bushnev, et al. Receptor channel trpc6 is a<br />

key mediator <strong>of</strong> notch-driven glioblastoma growth and invasiveness. Cancer research,<br />

70(1):418–427, 2010.<br />

[96] E. Chiocca. The many functions <strong>of</strong> microRNAs in glioblastoma. World neurosurgery,<br />

2010.<br />

[97] S. Chirasani, A. Sternjak, P. Wend, S. Momma, B. Campos, I. Herrmann, D. Graf,<br />

T. Mitsiadis, C. Herold-Mende, D. Besser, et al. Bone morphogenetic protein-7 release<br />

from endogenous neural precursor cells suppresses the tumourigenicity <strong>of</strong> stem-like<br />

glioblastoma cells. Brain, 133(7):1961–1972, 2010.<br />

[98] M. Choi, U. Scholl, W. Ji, T. Liu, I. Tikhonova, P. Zumbo, A. Nayir, A. Bakkaloğlu,<br />

S. Özen, S. Sanjad, et al. Genetic diagnosis by whole exome capture and massively parallel<br />

dna sequencing. Proceedings <strong>of</strong> the National Academy <strong>of</strong> Sciences, 106(45):19096–<br />

19101, 2009.<br />

[99] L. M. Chow and S. J. Baker. PTEN function in normal and neoplastic growth. Cancer<br />

letters, 241(2):184–196, Sept. 2006.<br />

[100] L. M. Chow, R. Endersby, X. Zhu, S. Rankin, C. Qu, J. Zhang, A. Broniscer, D. W.<br />

Ellison, and S. J. Baker. Cooperativity within and among Pten, p53, and Rb pathways<br />

induces high-grade astrocytoma in adult brain. Cancer Cell, 19(3):305–16, 2011.<br />

[101] M. J. Clark, N. Homer, B. D. O’Connor, Z. Chen, A. Eskin, H. Lee, B. Merriman, and<br />

S. F. Nelson. U87MG decoded: the genomic sequence <strong>of</strong> a cytogenetically aberrant<br />

human cancer cell line. PLoS genetics, 6(1):e1000832, 2010.<br />

[102] N. M. Cohen, E. Kenigsberg, and A. Tanay. Primate CpG islands are maintained by<br />

heterogeneous evolutionary regimes involving minimal selection. Cell, 145(5):773–786,<br />

May 2011.<br />

[103] A. Collins, P. Berry, C. Hyde, M. Stower, and N. Maitland. Prospective identification<br />

<strong>of</strong> tumorigenic prostate cancer stem cells. Cancer research, 65(23):10946, 2005.<br />

[104] V. Collins. Amplified genes in human gliomas. In Seminars in cancer biology, volume 4,<br />

page 27, 1993.<br />

[105] H. Colman, L. Zhang, E. Sulman, J. McDonald, N. Shooshtari, A. Rivera, S. Pop<strong>of</strong>f,<br />

C. Nutt, D. Louis, J. Cairncross, et al. A multigene predictor <strong>of</strong> outcome in glioblastoma.<br />

Neuro-oncology, 12(1):49–57, 2010.<br />

[106] G. O. Consortium et al. Gene ontology: tool for the unification <strong>of</strong> biology. Nature<br />

genetics, 25(1):25–29, 2000.<br />

[107] L. Conti, S. M. Pollard, T. Gorba, E. Reitano, M. Toselli, G. Biella, Y. Sun, S. Sanzone,<br />

Q.-L. Ying, E. Cattaneo, and A. Smith. Niche-Independent Symmetrical Self-Renewal<br />

<strong>of</strong> a Mammalian Tissue <strong>Stem</strong> Cell. PLoS Biology, 3(9):e283, 2005.<br />

[108] H. Contreras, R. Ledezma, J. Vergara, F. Cifuentes, C. Barra, P. Cabello, I. Gallegos,<br />

B. Morales, C. Huidobro, and E. Castellón. The expression <strong>of</strong> syndecan-1 and-2<br />

is associated with gleason score and epithelial-mesenchymal transition markers, ecadherin<br />

and β-catenin, in prostate cancer. In Urologic Oncology: Seminars and<br />

Original Investigations, volume 28, pages 534–540. Elsevier, 2010.<br />

[109] A. Coutts, E. MacKenzie, E. Griffith, and D. Black. Tes is a novel focal adhesion<br />

protein with a role in cell spreading. Journal <strong>of</strong> cell science, 116(5):897–906, 2003.<br />

[110] Y. Cui, J. Wang, X. Zhang, R. Lang, M. Bi, L. Guo, and S. Lu. Ecrg2, a novel<br />

candidate <strong>of</strong> tumor suppressor gene in the esophageal carcinoma, interacts directly<br />

with metallothionein 2a and links to apoptosis* 1,* 2,* 3. Biochemical and biophysical<br />

research communications, 302(4):904–915, 2003.<br />

[111] M. Cully, H. You, and A. Levine. Beyond PTEN mutations: the PI3K pathway as an<br />

integrator <strong>of</strong> multiple inputs during tumorigenesis. Nature Reviews Cancer, 2006.


[112] M. Czystowska, J. Han, M. Szczepanski, M. Szajnik, K. Quadrini, H. Brandwein,<br />

J. Hadden, K. Signorelli, and T. Whiteside. Irx-2, a novel immunotherapeutic, protects<br />

human t cells from tumor-induced cell death. Cell Death & Differentiation, 16(5):708–<br />

718, 2009.<br />

[113] C. Dang, M. Gottschling, K. Manning, E. O’Currain, S. Schneider, W. Sterry,<br />

E. Stockfleth, and I. Nindl. Identification <strong>of</strong> dysregulated genes in cutaneous squamous<br />

cell carcinoma. Oncology reports, 16(3):513–519, 2006.<br />

[114] L. De Filippis, G. Lamorte, E. Snyder, A. Malgaroli, and A. Vescovi. A novel, immortal,<br />

and multipotent human neural stem cell line generating functional neurons and<br />

oligodendrocytes. <strong>Stem</strong> cells, 25(9):2312–2321, 2007.<br />

[115] C. Dehay and H. Kennedy. Cell-cycle control and cortical development. Nature Reviews<br />

Neuroscience, 8(6):438–450, 2007.<br />

[116] L. Deleyrolle and B. Reynolds. Isolation, expansion, and differentiation <strong>of</strong> adult mammalian<br />

neural stem and progenitor cells using the neurosphere assay. Methods Mol Biol,<br />

549:91–101, 2009.<br />

[117] G. Denning, B. Jean-Joseph, C. Prince, D. Durden, and P. Vogt. A short n-terminal<br />

sequence <strong>of</strong> pten controls cytoplasmic localization and is required for suppression <strong>of</strong><br />

cell growth. Oncogene, 26(27):3930–3940, 2007.<br />

[118] C. Desmet and D. Peeper. The neurotrophic receptor trkb: a drug target in anti-cancer<br />

therapy? Cellular and molecular life sciences, 63(7):755–759, 2006.<br />

[119] L. Desnoyers, R. Pai, R. Ferrando, K. Hötzel, T. Le, J. Ross, R. Carano, A. D’Souza,<br />

J. Qing, I. Mohtashemi, et al. Targeting fgf19 inhibits tumor growth in colon cancer<br />

xenograft and fgf19 transgenic hepatocellular carcinoma models. Oncogene, 27(1):85–<br />

97, 2007.<br />

[120] T. Di Tomaso, S. Mazzoleni, E. Wang, G. Sovena, D. Clavenna, A. Franzin, P. Mortini,<br />

S. Ferrone, C. Doglioni, F. Marincola, et al. Immunobiological characterization<br />

<strong>of</strong> cancer stem cells isolated from glioblastoma patients. Clinical Cancer Research,<br />

16(3):800–813, 2010.<br />

[121] P. Dirks. <strong>Stem</strong> cells and brain tumours. Nature, 444(7120):687–688, 2006.<br />

[122] P. Dirks. Brain tumour stem cells: the undercurrents <strong>of</strong> human brain cancer and their<br />

relationship to neural stem cells. Philosophical Transactions <strong>of</strong> the Royal Society B:<br />

Biological Sciences, 363(1489):139, 2008.<br />

[123] P. B. Dirks. Cancer: stem cells and brain tumours. Nature, 444(7120):687–8, 2006.<br />

[124] J. G. Doench and P. A. Sharp. Specificity <strong>of</strong> microRNA target selection in translational<br />

repression. Genes Dev, 18(5):504–11, 2004.<br />

[125] T. Doetschman, H. Eistetter, M. Katz, W. Schmidt, and R. Kemler. The in vitro<br />

development <strong>of</strong> blastocyst-derived embryonic stem cell lines: formation <strong>of</strong> visceral<br />

yolk sac, blood islands and myocardium. Journal <strong>of</strong> embryology and experimental<br />

morphology, 87(1):27, 1985.<br />

[126] S. Dolci, A. Belmonte, R. Santone, M. Giorgi, M. Pellegrini, E. Carosa, E. Piccione,<br />

A. Lenzi, and E. Jannini. Subcellular localization and regulation <strong>of</strong> type-1c<br />

and type-5 phosphodiesterases. Biochemical and biophysical research communications,<br />

341(3):837–846, 2006.<br />

[127] G. Dominic, W. Yi-Lu, L. David, and R. Nikolaus. microrna target predictions across<br />

seven drosophila species and comparison to mammalian targets. 2005.<br />

[128] D. Doyle, G. Hanks, and N. Cherny. Oxford textbook <strong>of</strong> palliative medicine. Oxford<br />

University Press, USA, 2005.<br />

[129] T. Du. microPrimer: the biogenesis and function <strong>of</strong> microRNA. Development,<br />

132(21):4645–4652, Sept. 2005.<br />

[130] A. Duensing and S. Duensing. Guilt by association? p53 and the development <strong>of</strong> aneuploidy<br />

in cancer. Biochemical and biophysical research communications, 331(3):694–<br />

700, 2005.<br />

[131] H. Dvinge and P. Bertone. Htqpcr: high-throughput analysis and visualization <strong>of</strong><br />

quantitative real-time pcr data in r. Bioinformatics, 25(24):3325–3326, 2009.


[132] D. Edwards. Non-linear normalization and background correction in one-channel cdna<br />

microarray studies. Bioinformatics, 19(7):825, 2003.<br />

[133] A. Efeyan and M. Serrano. p53: guardian <strong>of</strong> the genome and policeman <strong>of</strong> the oncogenes.<br />

Cell Cycle, 6(9):1006–1010, 2007.<br />

[134] A. J. Enright, B. John, U. Gaul, T. Tuschl, C. Sander, and D. S. Marks. MicroRNA<br />

targets in Drosophila. Genome Biol, 5(1):R1, 2003.<br />

[135] P. Eriksson, E. Perfilieva, T. Björk-Eriksson, A. Alborn, C. Nordborg, D. Peterson,<br />

and F. Gage. Neurogenesis in the adult human hippocampus. Nature medicine,<br />

4(11):1313–1317, 1998.<br />

[136] J. Erlichman and J. Leiter. Glia modulation <strong>of</strong> the extracellular milieu as a factor in<br />

central CO2 chemosensitivity and respiratory control. Journal <strong>of</strong> Applied Physiology,<br />

108(6):1803, 2010.<br />

[137] C. Esposito, M. Scrima, A. Carotenuto, A. Tedeschi, P. Rovero, G. D’Errico, A. Malfitano,<br />

M. Bifulco, and D. Anna Maria. Structures and micelle locations <strong>of</strong> the<br />

nonlipidated and lipidated c-terminal membrane anchor <strong>of</strong> 2’, 3’-cyclic nucleotide-<br />

3’-phosphodiesterase. Biochemistry, 47(1):308–319, 2008.<br />

[138] S. Falcon and R. Gentleman. Using gostats to test gene lists for go term association.<br />

Bioinformatics, 23(2):257, 2007.<br />

[139] K. K. Farh, A. Grimson, C. Jan, B. P. Lewis, W. K. Johnston, L. P. Lim, C. B.<br />

Burge, and D. P. Bartel. The widespread impact <strong>of</strong> mammalian MicroRNAs on mRNA<br />

repression and evolution. Science, 310(5755):1817–21, 2005.<br />

[140] T. Fawcett, H. Eastman, J. Martindale, and N. Holbrook. Physical and functional<br />

association between gadd153 and ccaat/enhancer-binding protein beta during cellular<br />

stress. Journal <strong>of</strong> Biological Chemistry, 271(24):14285–14289, 1996.<br />

[141] C. Fears, C. Gladson, and A. Woods. Syndecan-2 is expressed in the microvasculature<br />

<strong>of</strong> gliomas and regulates angiogenic processes in microvascular endothelial cells.<br />

Journal <strong>of</strong> Biological Chemistry, 281(21):14533–14536, 2006.<br />

[142] R. Feil and F. Berger. Convergent evolution <strong>of</strong> genomic imprinting in plants and<br />

mammals. Trends in Genetics, 23(4):192–199, 2007.<br />

[143] B. G. Firehose. Broad gdac firehose.<br />

[144] A. Fischer and R. Bongini. Turning müller glia into neural progenitors in the retina.<br />

Molecular neurobiology, pages 1–11, 2010.<br />

[145] J. Flax, S. Aurora, C. Yang, C. Simonin, A. Wills, L. Billinghurst, M. Jendoubi,<br />

R. Sidman, J. Wolfe, S. Kim, et al. Engraftable human neural stem cells respond to<br />

developmental cues, replace neurons, and express foreign genes. Nature biotechnology,<br />

16:1033–1039, 1998.<br />

[146] P. Flicek, M. Amode, D. Barrell, K. Beal, S. Brent, D. Carvalho-Silva, P. Clapham,<br />

G. Coates, S. Fairley, S. Fitzgerald, et al. Ensembl 2012. Nucleic acids research,<br />

40(D1):D84–D90, 2012.<br />

[147] P. Flicek, M. Amode, D. Barrell, K. Beal, S. Brent, Y. Chen, P. Clapham, G. Coates,<br />

S. Fairley, S. Fitzgerald, et al. Ensembl 2011. Nucleic acids research, 39(suppl 1):D800,<br />

2011.<br />

[148] W. Freije, F. Castro-Vargas, Z. Fang, S. Horvath, T. Cloughesy, L. Liau, P. Mischel,<br />

and S. Nelson. Gene expression pr<strong>of</strong>iling <strong>of</strong> gliomas strongly predicts survival. Cancer<br />

research, 64(18):6503, 2004.<br />

[149] R. Fricker, M. Carpenter, C. Winkler, C. Greco, M. Gates, and A. Björklund. Sitespecific<br />

migration and neuronal differentiation <strong>of</strong> human neural progenitor cells after<br />

transplantation in the adult rat brain. The Journal <strong>of</strong> neuroscience, 19(14):5990, 1999.<br />

[150] R. Friedman, K. Farh, C. Burge, and D. Bartel. Most mammalian mrnas are conserved<br />

targets <strong>of</strong> micrornas. Genome research, 19(1):92–105, 2009.<br />

[151] M. Frolov and N. Dyson. Molecular mechanisms <strong>of</strong> E2F-dependent activation and<br />

pRB-mediated repression. Journal <strong>of</strong> cell science, 117(11):2173, 2004.<br />

[152] P. Fujita, B. Rhead, A. Zweig, A. Hinrichs, D. Karolchik, M. Cline, M. Goldman,<br />

G. Barber, H. Clawson, A. Coelho, et al. The ucsc genome browser database: update<br />

2011. Nucleic acids research, 39(suppl 1):D876, 2011.


[153] T. Fujiwara, M. Bandi, M. Nitta, E. Ivanova, R. Bronson, and D. Pellman. Cytokinesis<br />

failure generating tetraploids promotes tumorigenesis in p53-null cells. Nature,<br />

437(7061):1043, 2005.<br />

[154] F. B. Furnari, T. Fenton, R. M. Bachoo, A. Mukasa, J. M. Stommel, A. Stegh, W. C.<br />

Hahn, K. L. Ligon, D. N. Louis, C. Brennan, L. Chin, R. A. DePinho, and W. K.<br />

Cavenee. Malignant astrocytic glioma: genetics, biology, and paths to treatment.<br />

Genes and development, 21(21):2683–710, 2007.<br />

[155] D. Gaidatzis, E. Van Nimwegen, J. Hausser, and M. Zavolan. Inference <strong>of</strong> mirna<br />

targets using evolutionary conservation and pathway analysis. BMC bioinformatics,<br />

8(1):69, 2007.<br />

[156] R. Galli, E. Binda, U. Orfanelli, B. Cipelletti, A. Gritti, S. De Vitis, R. Fiocco,<br />

C. Foroni, F. Dimeco, and A. Vescovi. Isolation and characterization <strong>of</strong> tumorigenic,<br />

stem-like neural precursors from human glioblastoma. Cancer research, 64(19):7011,<br />

2004.<br />

[157] G. Gallia, V. Rand, I. Siu, et al. PIK3CA gene mutations in pediatric and adult<br />

glioblastoma multiforme. Molecular cancer research, 4(10):709, 2006.<br />

[158] E. Garcia-Aragoncillo, J. Carrillo, E. Lalli, N. Agra, G. Gomez-Lopez, A. Pestana,<br />

and J. Alonso. Dax1, a direct target <strong>of</strong> ews/fli1 oncoprotein, is a principal regulator<br />

<strong>of</strong> cell-cycle progression in ewing’s tumor cells. Oncogene, 27(46):6034–6043, 2008.<br />

[159] M. Gardiner-Garden and M. Frommer. CpG islands in vertebrate genomes. Journal<br />

<strong>of</strong> molecular biology, 196(2):261–282, July 1987.<br />

[160] A. Gartel and S. Radhakrishnan. Lost in transcription: p21 repression, mechanisms,<br />

and consequences. Cancer research, 65(10):3980, 2005.<br />

[161] L. Gautier, L. Cope, B. Bolstad, and R. Irizarry. affyÂŮanalysis <strong>of</strong> affymetrix genechip<br />

data at the probe level. Bioinformatics, 20(3):307–315, 2004.<br />

[162] GBMbase. A bioinformatics resource for glioblastoma multiforme.<br />

http://beta.gbmbase.org/page/welcome/display.<br />

[163] A. Giganti, J. Plastino, B. Janji, M. Van Troys, D. Lentz, C. Ampe, C. Sykes, and<br />

E. Friederich. Actin-filament cross-linking protein t-plastin increases arp2/3-mediated<br />

actin-based movement. Journal <strong>of</strong> cell science, 118(6):1255, 2005.<br />

[164] A. Girard, R. Sachidanandam, G. J. Hannon, and M. A. Carmell. A germline-specific<br />

class <strong>of</strong> small RNAs binds mammalian Piwi proteins. Nature, 442(7099):199–202,<br />

2006.<br />

[165] T. Glaser, S. M. Pollard, A. Smith, and O. Brüstle. Tripotential Differentiation <strong>of</strong><br />

Adherently Expandable <strong>Neural</strong> <strong>Stem</strong> (NS) <strong>Cells</strong>. PLoS ONE, 2(3):e298, Mar. 2007.<br />

[166] V. Gocheva and J. Joyce. Cysteine cathepsins and the cutting edge <strong>of</strong> cancer invasion.<br />

Cell cycle, 6(1):60–64, 2007.<br />

[167] V. Gocheva, W. Zeng, D. Ke, D. Klimstra, T. Reinheckel, C. Peters, D. Hanahan,<br />

and J. Joyce. Distinct roles for cysteine cathepsin genes in multistage tumorigenesis.<br />

Genes & development, 20(5):543–556, 2006.<br />

[168] K. Goh, W. Poon, D. Chan, and C. Ip. Tissue plasminogen activator expression in<br />

meningiomas and glioblastomas. Clinical neurology and neurosurgery, 107(4):296–300,<br />

2005.<br />

[169] S. Gomez-Lopez, O. Wiskow, R. Favaro, S. Nicolis, D. Price, S. Pollard, and S. A.<br />

Sox2 and pax6 maintain the proliferative and developmental potential <strong>of</strong> gliogenic<br />

neural stem cells in vitro. Glia, 59:1588–1599, 2011.<br />

[170] M. Göransson, M. Andersson, C. Forni, A. Ståhlberg, C. Andersson, A. Ol<strong>of</strong>sson,<br />

R. Mantovani, and P. Åman. The myxoid liposarcoma fus-ddit3 fusion oncoprotein<br />

deregulates nf-κb target genes by interaction with nfkbiz. Oncogene, 28(2):270–278,<br />

2008.<br />

[171] E. Gould, A. Reeves, M. Graziano, and C. Gross. Neurogenesis in the neocortex <strong>of</strong><br />

adult primates. Science, 286(5439):548, 1999.<br />

[172] A. Gourine and S. Kasparov. Astrocytes as brain interoceptors. Experimental Physiology,<br />

96(4):411, 2011.


[173] A. Gourine, V. Kasymov, N. Marina, F. Tang, M. Figueiredo, S. Lane,<br />

A. Teschemacher, K. Spyer, K. Deisseroth, and S. Kasparov. Astrocytes control<br />

breathing through pH-dependent release <strong>of</strong> ATP. Science, 329(5991):571, 2010.<br />

[174] M. Graeber, C. Tran, P. Wolz, R. Egensperger, S. Kösel, Y. Imai, K. Bise, S. Kohsaka,<br />

and P. Mehraein. Differential expression <strong>of</strong> mhc class ii molecules by microglia and<br />

neoplastic astroglia: relevance for the escape <strong>of</strong> astrocytoma cells from immune surveillance.<br />

Neuropathology and applied neurobiology, 24:293–301, 1998.<br />

[175] B. M. H. H. G. M. Graff L, Castrop F. Expression <strong>of</strong> vesicular monoamine transporters,<br />

synaptosomalâĂŘassociated protein 25 and syntaxin1: a signature <strong>of</strong> human small cell<br />

lung carcinoma. Cancer Res, 61:2138–2144, 2001.<br />

[176] L. Gravendeel, M. Kouwenhoven, O. Gevaert, J. de Rooi, A. Stubbs, J. Duijm, A. Daemen,<br />

F. Bleeker, L. Bralten, N. Kloosterh<strong>of</strong>, et al. Intrinsic gene expression pr<strong>of</strong>iles <strong>of</strong><br />

gliomas are a better predictor <strong>of</strong> survival than histology. Cancer research, 69(23):9065–<br />

9072, 2009.<br />

[177] A. Gregorieff and H. Clevers. Wnt signaling in the intestinal epithelium: from endoderm<br />

to cancer. Genes & development, 19(8):877–890, 2005.<br />

[178] C. Gregorio-King, J. McLeod, F. Collier, G. Collier, K. Bolton, G. Van Der Meer,<br />

J. Apostolopoulos, and M. Kirkland. Merp1: a mammalian ependymin-related protein<br />

gene differentially expressed in hematopoietic cells. Gene, 286(2):249–257, 2002.<br />

[179] S. Griffiths-Jones, H. K. Saini, S. van Dongen, and A. J. Enright. MiRBase: tools for<br />

microRNA genomics. Nucleic Acids Res, 36(Database issue):D154–8, 2008.<br />

[180] A. Grimson, K. K. Farh, W. K. Johnston, P. Garrett-Engele, L. P. Lim, and D. P. Bartel.<br />

MicroRNA targeting specificity in mammals: determinants beyond seed pairing.<br />

Mol Cell, 27(1):91–105, 2007.<br />

[181] J. Gunnersen, V. Spirkoska, P. Smith, R. Danks, and S. Tan. Growth and migration<br />

markers <strong>of</strong> rat c6 glioma cells identified by serial analysis <strong>of</strong> gene expression. Glia,<br />

32(2):146–154, 2000.<br />

[182] P. Gupta, C. Chaffer, and R. Weinberg. Cancer stem cells: mirage or reality? Nature<br />

medicine, 15(9):1010–1012, 2009.<br />

[183] C. Hagemann, J. Anacker, S. Haas, D. Riesner, B. Schömig, R. Ernestus, and G. Vince.<br />

Comparative expression pattern <strong>of</strong> matrix-metalloproteinases in human glioblastoma<br />

cell-lines and primary cultures. BMC research notes, 3(1):293, 2010.<br />

[184] I. Han, H. Park, and E. Oh. New insights into syndecan-2 expression and tumourigenic<br />

activity in colon carcinoma cells. Journal <strong>of</strong> Molecular Histology, 35(3):319–326, 2004.<br />

[185] K. L. Harms and X. Chen. The C terminus <strong>of</strong> p53 family proteins is a cell fate<br />

determinant. Molecular and cellular biology, 25(5):2014–2030, Mar. 2005.<br />

[186] M. Harris, H. Yang, B. Low, J. Mukherje, A. Guha, R. Bronson, L. Shultz, M. Israel,<br />

and K. Yun. Cancer stem cells are enriched in the side population cells in a mouse<br />

model <strong>of</strong> glioma. Cancer research, 68(24):10051–10059, 2008.<br />

[187] E. Hartfuss, R. Galli, N. Heins, and M. Götz. <strong>Characterization</strong> <strong>of</strong> cns precursor<br />

subtypes and radial glia. Developmental biology, 229(1):15–30, 2001.<br />

[188] R. Haviland, S. Eschrich, G. Bloom, Y. Ma, S. Minton, R. Jove, and W. Cress.<br />

Necdin, a negative growth regulator, is a novel stat3 target gene down-regulated in<br />

human cancer. PloS one, 6(10):e24923, 2011.<br />

[189] Y. Hayakawa, Y. Hirata, H. Nakagawa, K. Sakamoto, Y. Hikiba, H. Kinoshita,<br />

W. Nakata, R. Takahashi, K. Tateishi, M. Tada, et al. Apoptosis signal-regulating<br />

kinase 1 and cyclin d1 compose a positive feedback loop contributing to tumor growth<br />

in gastric cancer. Proceedings <strong>of</strong> the National Academy <strong>of</strong> Sciences, 108(2):780–785,<br />

2011.<br />

[190] L. He, C. Fan, A. Kapoor, A. Ingram, A. Rybak, R. Austin, J. Dickhout, J. Cutz,<br />

J. Scholey, and D. Tang. [alpha]-mannosidase 2c1 attenuates pten function in prostate<br />

cancer cells. Nature Communications, 2:307, 2011.<br />

[191] C. Heldin, A. Ostman, A. Eriksson, A. Siegbahn, L. Claesson-Welsh, and B. Westermark.<br />

Platelet-derived growth factor: Is<strong>of</strong>orm-specific signalling via heterodimeric or<br />

homodimeric receptor complexes. Kidney Int, 41(3):571–574, 1992.


[192] M. Hernandez, M. Nieto, and M. Sanchez Crespo. Cytosolic phospholipase a2 and<br />

the distinct transcriptional programs <strong>of</strong> astrocytoma cells. Trends in neurosciences,<br />

23(6):259–264, 2000.<br />

[193] K. Herrup and Y. Yang. Cell cycle regulation in the postmitotic neuron: oxymoron<br />

or new biology? Nature Reviews Neuroscience, 8(5):368–378, 2007.<br />

[194] M. S. Hestand, A. Klingenh<strong>of</strong>f, M. Scherf, Y. Ariyurek, Y. Ramos, W. van Workum,<br />

M. Suzuki, T. Werner, G. J. van Ommen, J. T. den Dunnen, M. Harbers, and P. A.<br />

t Hoen. Tissue-specific transcript annotation and expression pr<strong>of</strong>iling with complementary<br />

next-generation sequencing technologies. Nucleic acids research, 38(16):e165,<br />

2010.<br />

[195] S. Hockfield and R. McKay. Identification <strong>of</strong> major cell classes in the developing<br />

mammalian nervous system. The Journal <strong>of</strong> neuroscience, 5(12):3310, 1985.<br />

[196] J. Hodgson, R. Yeh, A. Ray, N. Wang, I. Smirnov, M. Yu, S. Hariono, J. Silber,<br />

H. Feiler, J. Gray, et al. Comparative analyses <strong>of</strong> gene copy number and mrna expression<br />

in glioblastoma multiforme tumors and xenografts. Neuro-oncology, 11(5):477–<br />

487, 2009.<br />

[197] R. H<strong>of</strong>fmann and A. Valencia. A gene network for navigating the literature. Nature<br />

genetics, 36(7):664–664, 2004.<br />

[198] L. Hook, J. Vives, N. Fulton, M. Leveridge, S. Lingard, M. Bootman, A. Falk, S. Pollard,<br />

T. Allsopp, D. Dalma-Weiszhausz, et al. Non-immortalized human neural stem<br />

(ns) cells as a scalable platform for cellular assays. Neurochemistry international, 2011.<br />

[199] S. Houwing, L. Kamminga, E. Berezikov, D. Cronembold, A. Girard, H. van den Elst,<br />

D. Filippov, H. Blaser, E. Raz, C. Moens, et al. A role for piwi and pirnas in germ<br />

cell maintenance and transposon silencing in zebrafish. Cell, 129(1):69–82, 2007.<br />

[200] S. Huang, L. Shu, M. Dilling, J. Easton, F. Harwood, H. Ichijo, and P. Houghton. Sustained<br />

activation <strong>of</strong> the jnk cascade and rapamycin-induced apoptosis are suppressed<br />

by p53/p21cip1. Molecular cell, 11(6):1491–1501, 2003.<br />

[201] X. Huang, D. Xiao, L. Xu, H. Zhong, L. Liao, Z. Xie, and E. Li. Prognostic significance<br />

<strong>of</strong> altered expression <strong>of</strong> sdc2 and cyr61 in esophageal squamous cell carcinoma.<br />

Oncology reports, 21(4):1123, 2009.<br />

[202] Z. Huang, Y. Kawase-Koga, S. Zhang, J. Visvader, M. Toth, C. Walsh, and T. Sun.<br />

Transcription factor lmo4 defines the shape <strong>of</strong> functional areas in developing cortices<br />

and regulates sensorimotor control. Developmental biology, 327(1):132–142, 2009.<br />

[203] HUGO. Gene nomenclature committee. http://www.genenames.org/.<br />

[204] C. Hunter, R. Smith, D. Cahill, P. Stephens, C. Stevens, J. Teague, C. Greenman,<br />

S. Edkins, G. Bignell, H. Davies, et al. A hypermutation phenotype and somatic MSH6<br />

mutations in recurrent human malignant gliomas after alkylator chemotherapy. Cancer<br />

research, 66(8):3987, 2006.<br />

[205] E. B. Institute. Intact database. http://www.ebi.ac.uk/intact/main.xhtml.<br />

[206] N. C. Institute. A to z list <strong>of</strong> cancers.<br />

[207] N. Ishii, D. Maier, A. Merlo, M. Tada, Y. Sawamura, A. Diserens, and E. Meir.<br />

Frequent co-alterations <strong>of</strong> TP53, p16/CDKN2A, p14ARF, PTEN tumor suppressor<br />

genes in human glioma cell lines. Brain pathology, 9(3):469–479, 1999.<br />

[208] K. Jabbari and G. Bernardi. Cytosine methylation and CpG, TpG, (CpA) and TpA<br />

frequencies. Gene, 333:143–149, 2004.<br />

[209] P. Jay, C. Rougeulle, A. Massacrier, A. Moncla, M. Mattel, P. Malzac, N. Roëckel,<br />

S. Taviaux, J. Lefranc, P. Cau, et al. The human necdin gene, ndn, is maternally imprinted<br />

and located in the prader-willi syndrome chromosomal region. Nature genetics,<br />

17(3):357–361, 1997.<br />

[210] J. Jiao and D. Chen. Induction <strong>of</strong> neurogenesis in non-conventional neurogenic regions<br />

<strong>of</strong> the adult cns by niche astrocyte-produced signals. <strong>Stem</strong> <strong>Cells</strong>, pages 2007–0513v1,<br />

2008.<br />

[211] F. Johansson, H. Göransson, and B. Westermark. Expression analysis <strong>of</strong> genes involved<br />

in brain tumor progression driven by retroviral insertional mutagenesis in mice.<br />

Oncogene, 24(24):3896–3905, 2005.


[212] K. Johe, T. Hazel, T. Muller, M. Dugich-Djordjevic, and R. McKay. Single factors<br />

direct the differentiation <strong>of</strong> stem cells from the fetal and adult central nervous system.<br />

Genes & development, 10(24):3129, 1996.<br />

[213] B. John, A. J. Enright, A. Aravin, T. Tuschl, C. Sander, and D. S. Marks. Human<br />

MicroRNA targets. PLoS Biol, 2(11):e363, 2004.<br />

[214] P. Jones and P. Laird. Cancer-epigenetics comes <strong>of</strong> age. Nature genetics, 21(2):163–<br />

167, 1999.<br />

[215] A. Kanawaty and J. Henderson. Genomic analysis <strong>of</strong> induced pluripotent stem (ips)<br />

cells: routes to reprogramming. Bioessays, 31(2):134–138, 2009.<br />

[216] Y. Kang, I. Kim, E. Kim, M. Yoon, S. Kim, T. Kwon, and K. Choi. Paxilline enhances<br />

trail-mediated apoptosis <strong>of</strong> glioma cells via modulation <strong>of</strong> c-flip, survivin and dr5.<br />

Experimental & molecular medicine, 43(1):24, 2011.<br />

[217] Y. Kang, I. Kim, E. Kim, M. Yoon, S. Kim, T. Kwon, and K. Choi. Paxilline enhances<br />

trail-mediated apoptosis <strong>of</strong> glioma cells via modulation <strong>of</strong> c-flip, survivin and dr5.<br />

Experimental & molecular medicine, 43(1):24–34, 2011.<br />

[218] A. Kaul and W. Maltese. Killing <strong>of</strong> cancer cells by the photoactivatable protein kinase<br />

c inhibitor, calphostin c, involves induction <strong>of</strong> endoplasmic reticulum stress. Neoplasia<br />

(New York, NY), 11(9):823, 2009.<br />

[219] B. Kaur, F. Khwaja, E. Severson, S. Matheny, D. Brat, and E. Van Meir. Hypoxia<br />

and the hypoxia-inducible-factor pathway in glioma growth and angiogenesis. Neurooncology,<br />

7(2):134, 2005.<br />

[220] M. Kawashima, K. Doh-ura, E. Mekada, M. Fukui, and T. Iwaki. Cd9 expression<br />

in solid non-neuroepithelial tumors and infiltrative astrocytic tumors. Journal <strong>of</strong><br />

Histochemistry & Cytochemistry, 50(9):1195, 2002.<br />

[221] M. Kawashima, K. Doh-ura, E. Mekada, M. Fukui, and T. Iwaki. Cd9 expression<br />

in solid non-neuroepithelial tumors and infiltrative astrocytic tumors. Journal <strong>of</strong><br />

Histochemistry & Cytochemistry, 50(9):1195–1203, 2002.<br />

[222] M. Kedde, M. J. Strasser, B. Boldajipour, J. A. Oude Vrielink, K. Slanchev, C. le Sage,<br />

R. Nagel, P. M. Voorhoeve, J. van Duijse, U. A. Orom, A. H. Lund, A. Perrakis, E. Raz,<br />

and R. Agami. RNA-binding protein Dnd1 inhibits microRNA access to target mRNA.<br />

Cell, 131(7):1273–86, 2007.<br />

[223] G. Kempermann, S. Jessberger, B. Steiner, and G. Kronenberg. Milestones <strong>of</strong> neuronal<br />

development in the adult hippocampus. Trends in Neurosciences, 27(8):447–452, 2004.<br />

[224] M. Kertesz, N. Iovino, U. Unnerstall, U. Gaul, and E. Segal. The role <strong>of</strong> site accessibility<br />

in microrna target recognition. Nature genetics, 39(10):1278–1284, 2007.<br />

[225] I. Kil, S. Kim, S. Lee, and J. Park. Small interfering RNA-mediated silencing <strong>of</strong><br />

mitochondrial NADP + -dependent isocitrate dehydrogenase enhances the sensitivity<br />

<strong>of</strong> HeLa cells toward tumor necrosis factor-α and anticancer drugs. Free Radical<br />

Biology and Medicine, 43(8):1197–1207, 2007.<br />

[226] D. Kim, C. H. Kim, J. I. Moon, Y. G. Chung, M. Y. Chang, B. S. Han, S. Ko, E. Yang,<br />

K. Y. Cha, R. Lanza, and K. S. Kim. Generation <strong>of</strong> human induced pluripotent stem<br />

cells by direct delivery <strong>of</strong> reprogramming proteins. Cell <strong>Stem</strong> Cell, 4(6):472–6, 2009.<br />

[227] I. Kim, Y. Kang, M. Yoon, E. Kim, S. Kim, T. Kwon, I. Kim, and K. Choi. Amiodarone<br />

sensitizes human glioma cells but not astrocytes to trail-induced apoptosis via chopmediated<br />

dr5 upregulation. Neuro-oncology, 13(3):267–279, 2011.<br />

[228] J. Kim, J. Choi, S. Lim, O. Kwon, J. Seo, S. Ryu, and P. Suh. Phospholipase c-eta 1 is<br />

activated by intracellular ca2+ mobilization and enhances gpcrs/plc/ca2+ signaling.<br />

Cellular Signalling, 2011.<br />

[229] S. Kim, M. Seong, B. Jeon, H. Ko, J. Kim, and S. Park. Phase analysis identifies<br />

compound heterozygous deletions <strong>of</strong> the PARK2 gene in patients with early-onset<br />

Parkinson disease. Clinical Genetics.<br />

[230] T. Kim, M. Hemberg, J. Gray, A. Costa, D. Bear, J. Wu, D. Harmin, M. Laptewicz,<br />

K. Barbara-Haley, S. Kuersten, et al. Widespread transcription at neuronal activityregulated<br />

enhancers. Nature, 465(7295):182–187, 2010.


[231] T.-M. Kim, W. Huang, R. Park, P. J. Park, and M. D. Johnson. A developmental<br />

taxonomy <strong>of</strong> glioblastoma defined and maintained by MicroRNAs. Cancer Research,<br />

71(9):3387–3399, May 2011.<br />

[232] V. N. Kim. MicroRNA biogenesis: coordinated cropping and dicing. Nature Reviews<br />

Molecular Cell Biology, 6(5):376–385, May 2005.<br />

[233] W. Kim and N. Sharpless. The regulation <strong>of</strong> INK4/ARF in cancer and aging. Cell,<br />

127(2):265–275, 2006.<br />

[234] R. Kincaid, A. Kuchinsky, and M. Creech. Vistaclara: an expression browser plug-in<br />

for cytoscape. Bioinformatics, 24(18):2112–2114, 2008.<br />

[235] S. Kinney, D. Smiraglia, S. James, M. Moser, B. Foster, and A. Karpf. Stage-specific<br />

alterations <strong>of</strong> dna methyltransferase expression, dna hypermethylation, and dna hypomethylation<br />

during prostate cancer progression in the transgenic adenocarcinoma<br />

<strong>of</strong> mouse prostate model. Molecular Cancer Research, 6(8):1365–1374, 2008.<br />

[236] L. S. Kinsey M, Smith R. Nr0b1 is required for the oncogenic phenotype mediated by<br />

ews/fli in ewing’s sarcoma. Mol Cancer Res, 4:851–859, 2006.<br />

[237] M. Kiriakidou, P. T. Nelson, A. Kouranov, P. Fitziev, C. Bouyioukos, Z. Mourelatos,<br />

and A. Hatzigeorgiou. A combined computational-experimental approach predicts<br />

human microRNA targets. Genes Dev, 18(10):1165–78, 2004.<br />

[238] M. Kitano, M. Nakaya, T. Nakamura, S. Nagata, and M. Matsuda. Imaging <strong>of</strong> rab5 activity<br />

identifies essential regulators for phagosome maturation. Nature, 453(7192):241–<br />

245, 2008.<br />

[239] C. Klattenh<strong>of</strong>f and W. Theurkauf. Biogenesis and germline functions <strong>of</strong> piRNAs.<br />

Development, 135(1):3–9, 2008.<br />

[240] A. Klein and B. Simons. Universal patterns <strong>of</strong> stem cell fate in cycling adult tissues.<br />

Development, 138(15):3103, 2011.<br />

[241] T. Kolesnikova, A. Kazarov, M. Lemieux, M. Lafleur, S. Kesari, A. Kung, and M. Hemler.<br />

Glioblastoma inhibition by cell surface immunoglobulin protein ewi-2, in vitro and<br />

in vivo. Neoplasia (New York, NY), 11(1):77, 2009.<br />

[242] T. Kondo, T. Setoguchi, and T. Taga. Persistence <strong>of</strong> a small subpopulation <strong>of</strong> cancer<br />

stem-like cells in the c6 glioma cell line. Proceedings <strong>of</strong> the National Academy <strong>of</strong><br />

Sciences <strong>of</strong> the United States <strong>of</strong> America, 101(3):781, 2004.<br />

[243] K. Kondoh, N. Tsuji, C. Kamagata, M. Sasaki, D. Kobayashi, A. Yagihashi, and<br />

N. Watanabe. A novel aspartic protease gene, alp56, is up-regulated in human breast<br />

cancer independently from the cathepsin d gene. Breast cancer research and treatment,<br />

78(1):37–44, 2003.<br />

[244] A. Korshunov, R. Sycheva, and A. Golanov. Genetically distinct and clinically relevant<br />

subtypes <strong>of</strong> glioblastoma defined by array-based comparative genomic hybridization<br />

(array-cgh). Acta neuropathologica, 111(5):465–474, 2006.<br />

[245] D. Koul, R. Shen, S. Bergh, Y. Lu, J. de Groot, T. Liu, G. Mills, and W. Yung.<br />

Targeting integrin-linked kinase inhibits Akt signaling pathways and decreases tumor<br />

progression <strong>of</strong> human glioblastoma. Molecular cancer therapeutics, 4(11):1681, 2005.<br />

[246] C. R. E. C. B. M. C. D. Kourtidis A, Jain R. An rna interference screen identifies<br />

metabolic regulators nr1d1 and pbp as novel survival factors for breast cancer cells<br />

with the erbb2 signature. Cancer Res, 70:1783–1792, 2010.<br />

[247] A. Krek, D. Grün, M. Poy, R. Wolf, L. Rosenberg, E. Epstein, P. MacMenamin,<br />

I. Da Piedade, K. Gunsalus, M. St<strong>of</strong>fel, et al. Combinatorial microrna target predictions.<br />

Nature genetics, 37(5):495–500, 2005.<br />

[248] A. Kriegstein and A. Alvarez-Buylla. The glial nature <strong>of</strong> embryonic and adult neural<br />

stem cells. Annual review <strong>of</strong> neuroscience, 32:149, 2009.<br />

[249] A. Kriegstein and M. Götz. Radial glia diversity: a matter <strong>of</strong> cell fate. Glia, 43(1):37–<br />

43, 2003.<br />

[250] R. Kroes, G. Dawson, and J. Moskal. Focused microarray analysis <strong>of</strong> glyco-gene<br />

expression in human glioblastomas. Journal <strong>of</strong> neurochemistry, 103(s1):14–24, 2007.


[251] R. Kroes, H. He, M. Emmett, C. Nilsson, F. Leach, I. Amster, A. Marshall,<br />

and J. Moskal. Overexpression <strong>of</strong> st6galnacv, a ganglioside-specific alpha 2, 6sialyltransferase,<br />

inhibits glioma growth in vivo. Proceedings <strong>of</strong> the National Academy<br />

<strong>of</strong> Sciences, 107(28):12646, 2010.<br />

[252] T. Kulikova, P. Aldebert, N. Althorpe, W. Baker, K. Bates, P. Browne, A. Van<br />

Den Broek, G. Cochrane, K. Duggan, R. Eberhardt, et al. The embl nucleotide<br />

sequence database. Nucleic Acids Research, 32(suppl 1):D27–D30, 2004.<br />

[253] M. Lafleur, D. Xu, and M. Hemler. Tetraspanin proteins regulate membrane type-1<br />

matrix metalloproteinase-dependent pericellular proteolysis. Molecular Biology <strong>of</strong> the<br />

Cell, 20(7):2030–2040, 2009.<br />

[254] J. Lai, D. Sandhu, C. Yu, T. Han, C. Moser, K. Jackson, R. Guerrero, I. Aderca,<br />

H. Isomoto, M. Garrity-Park, et al. Sulfatase 2 up-regulates glypican 3, promotes<br />

fibroblast growth factor signaling, and decreases survival in hepatocellular carcinoma.<br />

Hepatology, 47(4):1211–1222, 2008.<br />

[255] Y. Lam, E. di Tomaso, H.-K. Ng, J. Pang, M. Roussel, and N. Hjelm. Expression <strong>of</strong><br />

p19 ink4d, cdk4, cdk6 in glioblastoma multiforme. British journal <strong>of</strong> neurosurgery,<br />

14(1):28–32, 2000.<br />

[256] P. Landgraf, M. Rusu, R. Sheridan, A. Sewer, N. Iovino, A. Aravin, S. Pfeffer, A. Rice,<br />

A. Kamphorst, M. Landthaler, et al. A mammalian microrna expression atlas based<br />

on small rna library sequencing. Cell, 129(7):1401–1414, 2007.<br />

[257] B. Langmead, C. Trapnell, M. Pop, and S. Salzberg. Ultrafast and memory-efficient<br />

alignment <strong>of</strong> short dna sequences to the human genome. Genome Biol, 10(3):R25,<br />

2009.<br />

[258] T. Lassmann, Y. Hayashizaki, and C. Daub. TagdustÂŮa program to eliminate artifacts<br />

from next generation sequencing data. Bioinformatics, 25(21):2839, 2009.<br />

[259] S. Lawler and E. Chiocca. Emerging functions <strong>of</strong> micrornas in glioblastoma. Journal<br />

<strong>of</strong> neuro-oncology, 92(3):297–306, 2009.<br />

[260] D. Lawson, M. Harrison, and C. Shapland. Fibroblast transgelin and smooth muscle<br />

sm22α are the same protein, the expression <strong>of</strong> which is down-regulated in many cell<br />

lines. Cell motility and the cytoskeleton, 38(3):250–257, 1998.<br />

[261] J. Lee, S. Kotliarova, Y. Kotliarov, A. Li, Q. Su, N. M. Donin, S. Pastorino, B. W.<br />

Purow, N. Christopher, W. Zhang, J. K. Park, and H. A. Fine. Tumor stem cells<br />

derived from glioblastomas cultured in bFGF and EGF more closely mirror the phenotype<br />

and genotype <strong>of</strong> primary tumors than do serum-cultured cell lines. Cancer<br />

Cell, 9(5):391–403, May 2006.<br />

[262] J. Lee, I. Vivanco, R. Beroukhim, J. Huang, W. Feng, R. DeBiasi, K. Yoshimoto,<br />

J. King, P. Nghiemphu, Y. Yuza, et al. Epidermal growth factor receptor activation<br />

in glioblastoma through novel missense mutations in the extracellular domain. PLoS<br />

medicine, 3(12):e485, 2006.<br />

[263] K. Lee, W. Han, J. Kim, I. Shin, E. Ko, I. Park, D. Lee, K. Oh, and D. Noh. The<br />

cd49d+/high subpopulation from isolated human breast sarcoma spheres possesses<br />

tumor-initiating ability. International journal <strong>of</strong> oncology, 40(3):665, 2012.<br />

[264] R. Lee, R. Feinbaum, and V. Ambros. The c. elegans heterochronic gene lin-4 encodes<br />

small rnas with antisense complementarity to lin-14. Cell, 75(5):843–854, 1993.<br />

[265] S. Lee, N. Syed, J. Taylor, P. Smith, B. Griffin, M. Baens, M. Bai, K. Bourantas,<br />

J. Stebbing, K. Naresh, et al. Dusp16 is an epigenetically regulated determinant <strong>of</strong><br />

jnk signalling in burkitt’s lymphoma. British journal <strong>of</strong> cancer, 103(2):265–274, 2010.<br />

[266] Y. S. Lee, S. Pressman, A. P. Andress, K. Kim, J. L. White, J. J. Cassidy, X. Li,<br />

K. Lubell, H. Lim do, I. S. Cho, K. Nakahara, J. B. Preall, P. Bellare, E. J. Sontheimer,<br />

and R. W. Carthew. Silencing by small RNAs is linked to endosomal trafficking. Nat<br />

Cell Biol, 11(9):1150–6, 2009.<br />

[267] S. J. Leevers, B. Vanhaesebroeck, and M. D. Waterfield. Signalling through phosphoinositide<br />

3-kinases: the lipids take centre stage. Current opinion in cell biology,<br />

11(2):219–225, Apr. 1999.


[268] H. Lemjabbar-Alaoui, A. van Zante, M. Singer, Q. Xue, Y. Wang, D. Tsay, B. He,<br />

D. Jablons, and S. Rosen. Sulf-2, a heparan sulfate endosulfatase, promotes human<br />

lung carcinogenesis. Oncogene, 29(5):635–646, 2009.<br />

[269] G. Lemke. Glial control <strong>of</strong> neuronal development. Annual review <strong>of</strong> neuroscience,<br />

24:87–105, 2001.<br />

[270] C. Leonetti, A. Biroccio, G. Graziani, and L. Tentori. Targeted therapy for brain<br />

tumours: Role <strong>of</strong> parp inhibitors. Current cancer drug targets, 12(3):218, 2012.<br />

[271] F. Leprêtre, C. Villenet, S. Quief, O. Nibourel, C. Jacquemin, X. Troussard, F. Jardin,<br />

F. Gibson, J. Kerckaert, C. Roumier, et al. Waved acgh: to smooth or not to smooth.<br />

Nucleic acids research, 38(7):e94–e94, 2010.<br />

[272] B. Lewis, C. Burge, and D. Bartel. Conserved seed pairing, <strong>of</strong>ten flanked by adenosines,<br />

indicates that thousands <strong>of</strong> human genes are microrna targets. Cell, 120(1):15–20,<br />

2005.<br />

[273] B. P. Lewis, I. H. Shih, M. W. Jones-Rhoades, D. P. Bartel, and C. B. Burge. Prediction<br />

<strong>of</strong> mammalian microRNA targets. Cell, 115(7):787–98, 2003.<br />

[274] M. Lewis, S. Ross, P. Strickland, C. Snyder, and C. Daniel. Regulated expression<br />

patterns <strong>of</strong> irx-2, an iroquois-class homeobox gene, in the human breast. Cell and<br />

tissue research, 296(3):549–554, 1999.<br />

[275] A. Li, J. Walling, Y. Kotliarov, A. Center, M. E. Steed, S. J. Ahn, M. Rosenblum,<br />

T. Mikkelsen, J. C. Zenklusen, and H. A. Fine. Genomic changes and gene expression<br />

pr<strong>of</strong>iles reveal that established glioma cell lines are poorly representative <strong>of</strong> primary<br />

human gliomas. Molecular cancer research : MCR, 6(1):21–30, 2008.<br />

[276] M. Li, L. Pevny, R. Lovell-Badge, and A. Smith. Generation <strong>of</strong> purified neural precursors<br />

from embryonic stem cells by lineage selection. Current biology, 8(17):971–974,<br />

1998.<br />

[277] Q. Li, A. Jedlicka, N. Ahuja, M. Gibbons, S. Baylin, P. Burger, J. Issa, et al. Concordant<br />

methylation <strong>of</strong> the er and n33 genes in glioblastoma multiforme. Oncogene,<br />

16(24):3197, 1998.<br />

[278] Y. Li, F. Guessous, Y. Zhang, C. DiPierro, B. Kefas, E. Johnson, L. Marcinkiewicz,<br />

J. Jiang, Y. Yang, T. Schmittgen, et al. Microrna-34a inhibits glioblastoma growth<br />

by targeting multiple oncogenes. Cancer research, 69(19):7569, 2009.<br />

[279] S. Liebner, C. Czupalla, and H. Wolburg. current concepts <strong>of</strong> blood-brain barrier<br />

development. The International journal <strong>of</strong> developmental biology, 2011.<br />

[280] A. Liekens, J. De Knijf, W. Daelemans, B. Goethals, P. De Rijk, and J. Del-Favero.<br />

Biograph: unsupervised biomedical knowledge discovery via automated hypothesis<br />

generation. Genome biology, 12(6):R57, 2011.<br />

[281] K. Ligon, J. Alberta, A. Kho, J. Weiss, M. Kwaan, C. Nutt, D. Louis, C. Stiles,<br />

and D. Rowitch. The oligodendroglial lineage marker OLIG2 is universally expressed<br />

in diffuse gliomas. Journal <strong>of</strong> Neuropathology & Experimental Neurology, 63(5):499,<br />

2004.<br />

[282] L. P. Lim, M. E. Glasner, S. Yekta, C. B. Burge, and D. P. Bartel. Vertebrate<br />

microRNA genes. Science, 299(5612):1540, 2003.<br />

[283] L. P. Lim, N. C. Lau, P. Garrett-Engele, A. Grimson, J. M. Schelter, J. Castle,<br />

D. P. Bartel, P. S. Linsley, and J. M. Johnson. Microarray analysis shows that some<br />

microRNAs downregulate large numbers <strong>of</strong> target mRNAs. Nature, 433(7027):769–73,<br />

2005.<br />

[284] L. Linares, A. Hengstermann, A. Ciechanover, S. Müller, and M. Scheffner. Hdmx<br />

stimulates Hdm2-mediated ubiquitination and degradation <strong>of</strong> p53. Proceedings <strong>of</strong> the<br />

National Academy <strong>of</strong> Sciences, 100(21):12009, 2003.<br />

[285] A. Linkous, E. Yazlovitskaya, and D. Hallahan. Cytosolic phospholipase a2 and<br />

lysophospholipids in tumor angiogenesis. Journal <strong>of</strong> the National Cancer Institute,<br />

102(18):1398–1412, 2010.<br />

[286] M. M. Lino and A. Merlo. PI3Kinase signaling in glioblastoma. Journal <strong>of</strong> neurooncology,<br />

103(3):417–427, July 2011.


[287] R. Lister, M. Pelizzola, R. Dowen, R. Hawkins, G. Hon, J. Tonti-Filippini, J. Nery,<br />

L. Lee, Z. Ye, Q. Ngo, et al. Human DNA methylomes at base resolution show<br />

widespread epigenomic differences. Nature, 462(7271):315–322, 2009.<br />

[288] B. Liu, Y. Kim, V. Leatherberry, P. Cowin, and C. Alexander. Mammary gland development<br />

requires syndecan-1 to create a β-catenin/tcf-responsive mammary epithelial<br />

subpopulation. Oncogene, 22(58):9243–9253, 2003.<br />

[289] B. Liu, S. McDermott, S. Khwaja, and C. Alexander. The transforming activity <strong>of</strong><br />

wnt effectors correlates with their ability to induce the accumulation <strong>of</strong> mammary<br />

progenitor cells. Proceedings <strong>of</strong> the National Academy <strong>of</strong> Sciences <strong>of</strong> the United States<br />

<strong>of</strong> America, 101(12):4158, 2004.<br />

[290] F. Liu, Y. You, X. Li, T. Ma, Y. Nie, B. Wei, T. Li, H. Lin, and Z. Yang. Brain injury<br />

does not alter the intrinsic differentiation potential <strong>of</strong> adult neuroblasts. The Journal<br />

<strong>of</strong> Neuroscience, 29(16):5075, 2009.<br />

[291] J. Liu, M. A. Valencia-Sanchez, G. J. Hannon, and R. Parker. MicroRNA-dependent<br />

localization <strong>of</strong> targeted mRNAs to mammalian P-bodies. Nat Cell Biol, 7(7):719–23,<br />

2005.<br />

[292] W. Liu, Y. Fu, S. Xu, F. Ding, G. Zhao, K. Zhang, C. Du, B. Pang, and Q. Pang.<br />

c-Met expression is associated with time to recurrence in patients with glioblastoma<br />

multiforme. Journal <strong>of</strong> clinical neuroscience : <strong>of</strong>ficial journal <strong>of</strong> the Neurosurgical<br />

Society <strong>of</strong> Australasia, 18(1):119–121, 2011.<br />

[293] X. Liu, A. Bolteus, D. Balkin, O. Henschel, and A. Bordey. Gfap-expressing cells<br />

in the postnatal subventricular zone display a unique glial phenotype intermediate<br />

between radial glia and astrocytes. Glia, 54(5):394–410, 2006.<br />

[294] X. Liu, M. Cicek, S. Plummer, E. Jorgenson, G. Casey, and J. Witte. Association<br />

<strong>of</strong> testis derived transcript gene variants and prostate cancer risk. The Journal <strong>of</strong><br />

urology, 177(3):894–898, 2007.<br />

[295] Y. Liu, S. Shete, C. Etzel, M. Scheurer, G. Alexiou, G. Armstrong, S. Tsavachidis,<br />

F. Liang, M. Gilbert, K. Aldape, et al. Polymorphisms <strong>of</strong> lig4, btbd2, hmga2, and<br />

rtel1 genes involved in the double-strand break repair pathway predict glioblastoma<br />

survival. Journal <strong>of</strong> Clinical Oncology, 28(14):2467–2474, 2010.<br />

[296] D. Q. S. P. C. K. L. O. T. J. L. J. K. V. C. T. M. P. L. T. L. L. N. S. T. C.-L.<br />

Liu Q, Nguyen DH. Molecular properties <strong>of</strong> cd133+ glioblastoma stem cells derived<br />

from treatment-refractory recurrent brain tumors. J Neurooncol, 94:1–19, 2009.<br />

[297] C. Lois and A. Alvarez-Buylla. Proliferating subventricular zone cells in the adult<br />

mammalian forebrain can differentiate into neurons and glia. Proceedings <strong>of</strong> the National<br />

Academy <strong>of</strong> Sciences, 90(5):2074, 1993.<br />

[298] B. Lorber, A. Guidi, J. Fawcett, and K. Martin. Activated retinal glia mediated axon<br />

regeneration in experimental glaucoma. Neurobiology <strong>of</strong> Disease, 2011.<br />

[299] C. Lottaz, D. Beier, K. Meyer, P. Kumar, A. Hermann, J. Schwarz, M. Junker,<br />

P. Oefner, U. Bogdahn, J. Wischhusen, et al. <strong>Transcriptional</strong> pr<strong>of</strong>iles <strong>of</strong> cd133+<br />

and cd133- glioblastoma-derived cancer stem cell lines suggest different cells <strong>of</strong> origin.<br />

Cancer research, 70(5):2030–2040, 2010.<br />

[300] D. Louis, H. Ohgaki, and O. Wiestler. The 2007 WHO classification <strong>of</strong> tumours <strong>of</strong><br />

the central nervous system. Acta Neuropathologica, 2007.<br />

[301] N. Louis. WHO Classification <strong>of</strong> Tumours <strong>of</strong> the Central Nervous System. International<br />

Agency for Research on Cancer, 2007.<br />

[302] S. Lowell, A. Benchoua, B. Heavey, and A. Smith. Notch promotes neural lineage<br />

entry by pluripotent embryonic stem cells. PLoS biology, 4(5):e121, 2006.<br />

[303] W. Ma, T. Tavakoli, E. Derby, Y. Serebryakova, M. Rao, and M. Mattson. Cellextracellular<br />

matrix interactions regulate neural differentiation <strong>of</strong> human embryonic<br />

stem cells. BMC developmental biology, 8(1):90, 2008.<br />

[304] Y.-H. Ma, R. Mentlein, F. Knerlich, M.-L. Kruse, H. M. Mehdorn, and J. Held-<br />

Feindt. Expression <strong>of</strong> stem cell markers in human astrocytomas <strong>of</strong> different WHO<br />

grades. Journal <strong>of</strong> neuro-oncology, 86(1):31–45, 2008.


[305] K. MAEDA, S. MATSUHASHI, K. TABUCHI, T. WATANABE, T. KATAGIRI,<br />

M. OYASU, N. SAITO, and S. KURODA. Brain specific human genes, nell1 and<br />

nell2, are predominantly expressed in neuroblastoma and other embryonal neuroepithelial<br />

tumors. Neurologia medico-chirurgica, 41(12):582–589, 2001.<br />

[306] H. Maier, C. Jones, B. Jasani, D. Öfner, B. Zelger, K. Schmid, and H. Budka. Metallothionein<br />

overexpression in human brain tumours. Acta neuropathologica, 94(6):599–<br />

604, 1997.<br />

[307] S. Maira, I. Galetic, D. Brazil, S. Kaech, E. Ingley, M. Thelen, and B. Hemmings.<br />

Carboxyl-terminal modulator protein (ctmp), a negative regulator <strong>of</strong> pkb/akt and<br />

v-akt at the plasma membrane. Science, 294(5541):374, 2001.<br />

[308] C. L. Maire and K. L. Ligon. <strong>Glioma</strong> Models: New GEMMs Add “Class” with Genomic<br />

and Expression Correlations. Cancer Cell, 19(3):295–297, Mar. 2011.<br />

[309] A. Majid, T. Lin, G. Best, K. Fishlock, S. Hewamana, G. Pratt, D. Yallop, A. Buggins,<br />

S. Wagner, B. Kennedy, et al. Cd49d is an independent prognostic marker that is<br />

associated with cxcr4 expression in cll. Leukemia research, 35(6):750–756, 2011.<br />

[310] P. Malatesta, E. Hartfuss, and M. Gotz. Isolation <strong>of</strong> radial glial cells by fluorescentactivated<br />

cell sorting reveals a neuronal lineage. Development, 127(24):5253, 2000.<br />

[311] M. Mancini and A. Toker. Nfat proteins: emerging roles in cancer progression. Nature<br />

Reviews Cancer, 9(11):810–820, 2009.<br />

[312] B. A. Mangerich A. How to kill tumor cells with inhibitors <strong>of</strong> poly(adp-ribosyl)ation.<br />

Int J Cancer, 128:251–265, 2011,.<br />

[313] J. Mao, J. Perez-losada, D. Wu, R. DelRosario, R. Tsunematsu, K. Nakayama,<br />

K. Brown, S. Bryson, and A. Balmain. Fbxw7/cdc4 is a p53-dependent, haploinsufficient<br />

tumour suppressor gene. Nature, 432(7018):775–779, 2004.<br />

[314] Y. Mao, H. Sunwoo, B. Zhang, and D. Spector. Direct visualization <strong>of</strong> the cotranscriptional<br />

assembly <strong>of</strong> a nuclear body by noncoding rnas. Nature cell biology,<br />

13(1):95–101, 2010.<br />

[315] M. Maragkakis, P. Alexiou, G. Papadopoulos, M. Reczko, T. Dalamagas, G. Giannopoulos,<br />

G. Goumas, E. Koukis, K. Kourtis, V. Simossis, et al. Accurate microrna<br />

target prediction correlates with protein repression levels. BMC bioinformatics,<br />

10(1):295, 2009.<br />

[316] M. Maragkakis, M. Reczko, V. Simossis, P. Alexiou, G. Papadopoulos, T. Dalamagas,<br />

G. Giannopoulos, G. Goumas, E. Koukis, K. Kourtis, et al. Diana-microt web<br />

server: elucidating microrna functions through target prediction. Nucleic acids research,<br />

37(suppl 2):W273–W276, 2009.<br />

[317] I. R. B. R. A. L. Marques MR, Horner JS. Mice lacking the p53/p63 target gene perp<br />

are resistant to papilloma development. Cancer Res, 65:6551–6556, 2005.<br />

[318] C. Marshall. RAS and RHO GTPases in G1-phase cell-cycle regulation. Nature<br />

Reviews Molecular Cell Biology, 5(5):355–366, 2004.<br />

[319] D. Martens, V. Tropepe, and D. van der Kooy. Separate proliferation kinetics <strong>of</strong> fibroblast<br />

growth factor-responsive and epidermal growth factor-responsive neural stem<br />

cells within the embryonic forebrain germinal zone. The Journal <strong>of</strong> Neuroscience,<br />

20(3):1085, 2000.<br />

[320] D. L. Masica and R. Karchin. Correlation <strong>of</strong> somatic mutation and expression identifies<br />

genes important in human glioblastoma progression and survival. Cancer Research,<br />

71(13):4550–4561, July 2011.<br />

[321] J. Massagué and D. Wotton. <strong>Transcriptional</strong> control by the tgf-beta/smad signaling<br />

system. The EMBO Journal, 19:1745–1754, 2000.<br />

[322] K. Matsumoto, S. Nishihara, M. Kamimura, T. Shiraishi, T. Otoguro, M. Uehara,<br />

Y. Maeda, K. Ogura, A. Lumsden, and T. Ogura. The prepattern transcription factor<br />

irx2, a target <strong>of</strong> the fgf8/map kinase cascade, is involved in cerebellum formation.<br />

Nature neuroscience, 7(6):605–612, 2004.<br />

[323] M. Matsumura, D. Fremont, P. Peterson, I. Wilson, et al. Emerging principles for the<br />

recognition <strong>of</strong> peptide antigens by mhc class i molecules. Science (New York, NY),<br />

257(5072):927, 1992.


[324] K. McCullough, J. Martindale, L. Klotz, T. Aw, and N. Holbrook. Gadd153 sensitizes<br />

cells to endoplasmic reticulum stress by down-regulating bcl2 and perturbing the<br />

cellular redox state. Science Signalling, 21(4):1249, 2001.<br />

[325] B. McEllin, C. Camacho, B. Mukherjee, B. Hahm, N. Tomimatsu, R. Bachoo, and<br />

S. Burma. Pten loss compromises homologous recombination repair in astrocytes:<br />

implications for glioblastoma therapy with temozolomide or poly (adp-ribose) polymerase<br />

inhibitors. Cancer research, 70(13):5457–5464, 2010.<br />

[326] R. McLendon, A. Friedman, D. Bigner, and E. Van Meir. Comprehensive genomic<br />

characterization defines human glioblastoma genes and core pathways. Nature, 2008.<br />

[327] M. Mehling, P. Simon, M. Mittelbronn, R. Meyermann, S. Ferrone, M. Weller, and<br />

H. Wiendl. WHO grade associated downregulation <strong>of</strong> MHC class I antigen-processing<br />

machinery components in human astrocytomas: does it reflect a potential immune<br />

escape mechanism? Acta neuropathologica, 114(2):111–9, 2007.<br />

[328] K. Meletis, F. Barnabé-Heider, M. Carlén, E. Evergren, N. Tomilin, O. Shupliakov,<br />

and J. Frisén. Spinal cord injury reveals multilineage differentiation <strong>of</strong> ependymal<br />

cells. PLoS biology, 6(7):e182, 2008.<br />

[329] I. Mellingh<strong>of</strong>f, M. Wang, I. Vivanco, D. Haas-Kogan, S. Zhu, E. Dia, K. Lu, K. Yoshimoto,<br />

J. Huang, D. Chute, et al. Molecular determinants <strong>of</strong> the response <strong>of</strong> glioblastomas<br />

to egfr kinase inhibitors. New England Journal <strong>of</strong> Medicine, 353(19):2012–2024,<br />

2005.<br />

[330] X. Meng, M. Leyva, M. Jenny, I. Gross, S. Benosman, B. Fricker, S. Harlepp,<br />

P. Hébraud, A. Boos, P. Wlosik, et al. A ruthenium-containing organometallic compound<br />

reduces tumor growth through induction <strong>of</strong> the endoplasmic reticulum stress<br />

gene chop. Cancer research, 69(13):5458–5466, 2009.<br />

[331] T. Mercer, M. Dinger, and J. Mattick. Long non-coding rnas: insights into functions.<br />

Nature Reviews Genetics, 10(3):155–159, 2009.<br />

[332] F. Merkle, A. Tramontin, J. García-Verdugo, and A. Alvarez-Buylla. Radial glia give<br />

rise to adult neural stem cells in the subventricular zone. Proceedings <strong>of</strong> the National<br />

Academy <strong>of</strong> Sciences <strong>of</strong> the United States <strong>of</strong> America, 101(50):17528, 2004.<br />

[333] MGI. Mouse genome informatics database. http://www.informatics.jax.org/.<br />

[334] P. S. Mischel, S. F. Nelson, and T. F. Cloughesy. Molecular analysis <strong>of</strong> glioblastoma:<br />

pathway pr<strong>of</strong>iling and its implications for patient therapy. Cancer biology and therapy,<br />

2(3):242–7, 2003.<br />

[335] S. Mitsui, N. Yamaguchi, Y. Osako, and K. Yuri. Enzymatic properties and localization<br />

<strong>of</strong> motopsin (prss12), a protease whose absence causes mental retardation. Brain<br />

research, 1136:1–12, 2007.<br />

[336] K. Mochizuki, N. Fine, T. Fujisawa, and M. Gorovsky. Analysis <strong>of</strong> a piwi-related gene<br />

implicates small rnas in genome rearrangement in tetrahymena. Cell, 110(6):689–699,<br />

2002.<br />

[337] E. Mohorko, R. Glockshuber, and M. Aebi. Oligosaccharyltransferase: the central<br />

enzyme <strong>of</strong> n-linked protein glycosylation. Journal <strong>of</strong> inherited metabolic disease, pages<br />

1–10, 2011.<br />

[338] K. Mokhtari, S. Paris, L. Aguirre-Cruz, N. Privat, E. Criniere, Y. Marie, J. Hauw,<br />

M. Kujas, D. Rowitch, K. Hoang-Xuan, et al. Olig2 expression, gfap, p53 and 1p loss<br />

analysis contribute to glioma subclassification. Neuropathology and applied neurobiology,<br />

31(1):62–69, 2005.<br />

[339] M. Montanez-Wiscovich, D. Seachrist, M. Landis, J. Visvader, B. Andersen, and<br />

R. Keri. Lmo4 is an essential mediator <strong>of</strong> erbb2/her2/neu-induced breast cancer cell<br />

cycle progression. Oncogene, 28(41):3608–3618, 2009.<br />

[340] M. E. Montanez-Wiscovich, D. D. Seachrist, M. D. Landis, J. Visvader, B. Andersen,<br />

and R. A. Keri. Lmo4 is an essential mediator <strong>of</strong> erbb2/her2/neu-induced breast<br />

cancer cell cycle progression. Oncogene, 28:3608–3618, 2009.<br />

[341] A. Moolwaney and O. Igwe. Regulation <strong>of</strong> the cyclooxygenase-2 system by interleukin-<br />

1β through mitogen-activated protein kinase signaling pathways: A comparative study<br />

<strong>of</strong> human neuroglioma and neuroblastoma cells. Molecular brain research, 137(1):202–<br />

212, 2005.


[342] H. Moon, M. Ahn, J. Park, K. Min, Y. Kwon, and K. Kim. Negative regulation <strong>of</strong><br />

hypoxia inducible factor-1α by necdin. FEBS letters, 579(17):3797–3801, 2005.<br />

[343] T. Mori, A. Buffo, and M. Götz. The novel roles <strong>of</strong> glial cells revisited: the contribution<br />

<strong>of</strong> radial glia and astrocytes to neurogenesis. Current topics in developmental biology,<br />

69:67–99, 2005.<br />

[344] M. Morimoto-Tomita, K. Uchimura, A. Bistrup, D. Lum, M. Egeblad, N. Boudreau,<br />

Z. Werb, and S. Rosen. Sulf-2, a proangiogenic heparan sulfate endosulfatase, is<br />

upregulated in breast cancer. Neoplasia (New York, NY), 7(11):1001, 2005.<br />

[345] S. Morrison and J. Kimble. Asymmetric and symmetric stem-cell divisions in development<br />

and cancer. Nature, 441(7097):1068–1074, 2006.<br />

[346] A. S. Morrissy, R. D. Morin, A. Delaney, T. Zeng, H. Mcdonald, S. Jones, Y. Zhao,<br />

M. Hirst, and M. A. Marra. Next-generation tag sequencing for cancer gene expression<br />

pr<strong>of</strong>iling. Genome Research, 19(10):1825–1835, Oct. 2009.<br />

[347] C. M. Morshead and D. van der Kooy. Disguising adult neural stem cells. Current<br />

opinion in neurobiology, 14(1):125–131, Feb. 2004.<br />

[348] G. Mosieniak, B. Pyrzynska, and B. Kaminska. Nuclear factor <strong>of</strong> activated t cells<br />

(nfat) as a new component <strong>of</strong> the signal transduction pathway in glioma cells. Journal<br />

<strong>of</strong> neurochemistry, 71(1):134–141, 1998.<br />

[349] W. Mueller, C. Nutt, M. Ehrich, M. Riemenschneider, A. Von Deimling, D. Van<br />

Den Boom, and D. Louis. Downregulation <strong>of</strong> runx3 and tes by hypermethylation in<br />

glioblastoma. Oncogene, 26(4):583–593, 2006.<br />

[350] J. Mukai, T. Hachiya, S. Shoji-Hoshino, M. Kimura, D. Nadano, P. Suvanto,<br />

T. Hanaoka, Y. Li, S. Irie, L. Greene, et al. Nade, a p75ntr-associated cell death<br />

executor, is involved in signal transduction mediated by the common neurotrophin<br />

receptor p75ntr. Journal <strong>of</strong> Biological Chemistry, 275(23):17566, 2000.<br />

[351] J. Mukai, P. Suvant, and T. Sato. Nerve growth factor-dependent regulation <strong>of</strong> nadeinduced<br />

apoptosis. Vitamins & Hormones, 66:385–402, 2003.<br />

[352] I. Muñoz-Sanjuán and A. H. Brivanlou. <strong>Neural</strong> induction, the default model and<br />

embryonic stem cells. Nature Reviews Neuroscience, 3(4):271–280, apr 2002.<br />

[353] A. Murat, E. Migliavacca, T. Gorlia, W. Lambiv, T. Shay, M. Hamou, N. De Tribolet,<br />

L. Regli, W. Wick, M. Kouwenhoven, et al. <strong>Stem</strong> cell-related self-renewal signature<br />

and high epidermal growth factor receptor expression associated with resistance<br />

to concomitant chemoradiotherapy in glioblastoma. Journal <strong>of</strong> Clinical Oncology,<br />

26(18):3015–3024, 2008.<br />

[354] N. Nakagomi, T. Nakagomi, S. Kubo, A. Nakano-Doi, O. Saino, M. Takata,<br />

H. Yoshikawa, D. Stern, T. Matsuyama, and A. Taguchi. Endothelial cells support<br />

survival, proliferation, and neuronal differentiation <strong>of</strong> transplanted adult ischemiainduced<br />

neural stem/progenitor cells after cerebral infarction. <strong>Stem</strong> <strong>Cells</strong>, 27(9):2185–<br />

2195, 2009.<br />

[355] C. Napoli, C. Lemieux, and R. Jorgensen. Introduction <strong>of</strong> a chimeric chalcone synthase<br />

gene into petunia results in reversible co-suppression <strong>of</strong> homologous genes in trans.<br />

The Plant Cell Online, 2(4):279, 1990.<br />

[356] K. Nave. Axon-glial signaling and the glial support <strong>of</strong> axon function. Annual review<br />

<strong>of</strong> neuroscience, 2008.<br />

[357] R. Nawroth, A. Van Zante, S. Cervantes, M. McManus, M. Hebrok, and S. Rosen.<br />

Extracellular sulfatases, elements <strong>of</strong> the wnt signaling pathway, positively regulate<br />

growth and tumorigenicity <strong>of</strong> human pancreatic cancer cells. PLoS One, 2(4):e392,<br />

2007.<br />

[358] S. Ng, K. Buckingham, C. Lee, A. Bigham, H. Tabor, K. Dent, C. Huff, P. Shannon,<br />

E. Jabs, D. Nickerson, et al. Exome sequencing identifies the cause <strong>of</strong> a mendelian<br />

disorder. Nature Genetics, 42(1):30–35, 2009.<br />

[359] C. B. Nielsen, N. Shomron, R. Sandberg, E. Hornstein, J. Kitzman, and C. B. Burge.<br />

Determinants <strong>of</strong> targeting by endogenous and exogenous microRNAs and siRNAs.<br />

RNA, 13(11):1894–910, 2007.


[360] I. Nimmrich, S. Erdmann, U. Melchers, S. Chtarbova, U. Finke, S. Hentsch, I. H<strong>of</strong>fmann,<br />

M. Oertel, W. H<strong>of</strong>fmann, and O. Muller. The novel ependymin related gene<br />

ucc1 is highly expressed in colorectal tumor cells. Cancer letters, 165(1):71–79, 2001.<br />

[361] S. Noctor, A. Flint, T. Weissman, W. Wong, B. Clinton, and A. Kriegstein. Dividing<br />

precursor cells <strong>of</strong> the embryonic cortical ventricular zone have morphological and<br />

molecular characteristics <strong>of</strong> radial glia. The Journal <strong>of</strong> neuroscience, 22(8):3161, 2002.<br />

[362] E. Noetzel, M. Rose, E. Sevinc, R. Hilgers, A. Hartmann, A. Naami, R. Knüchel,<br />

and E. Dahl. Intermediate filament dynamics and breast cancer: Aberrant promoter<br />

methylation <strong>of</strong> the synemin gene is associated with early tumor relapse. Oncogene,<br />

29(34):4814–4825, 2010.<br />

[363] H. Noushmehr, D. Weisenberger, and K. Diefes. Identification <strong>of</strong> a CpG island methylator<br />

phenotype that defines a distinct subgroup <strong>of</strong> glioma. Cancer Cell, 2010.<br />

[364] J. Novakova, O. Slaby, and R. Vyzula. Microrna involvement in glioblastoma pathogenesis.<br />

Biochemical and biophysical research communications, 2009.<br />

[365] H. Ohgaki and P. Kleihues. Epidemiology and etiology <strong>of</strong> gliomas. Acta neuropathologica,<br />

109(1):93–108, 2005.<br />

[366] K. Ohira, N. Funatsu, K. Homma, Y. Sahara, M. Hayashi, T. Kaneko, and S. Nakamura.<br />

Truncated trkb-t1 regulates the morphology <strong>of</strong> neocortical layer i astrocytes in<br />

adult rat brain slices. European Journal <strong>of</strong> Neuroscience, 25(2):406–416, 2007.<br />

[367] S. Okabe, K. Forsberg-Nilsson, A. Spiro, M. Segal, and R. McKay. Development <strong>of</strong><br />

neuronal precursor cells and functional postmitotic neurons from embryonic stem cells<br />

in vitro. Mechanisms <strong>of</strong> development, 59(1):89–102, 1996.<br />

[368] Y. Okazaki, M. Furuno, T. Kasukawa, J. Adachi, H. Bono, S. Kondo, I. Nikaido,<br />

N. Osato, R. Saito, H. Suzuki, et al. Analysis <strong>of</strong> the mouse transcriptome based on<br />

functional annotation <strong>of</strong> 60,770 full-length cdnas. Nature, 420(6915):563–573, 2002.<br />

[369] M. Okoniewski, T. Yates, S. Dibben, and C. Miller. An annotation infrastructure<br />

for the analysis and interpretation <strong>of</strong> affymetrix exon array data. Genome biology,<br />

8(5):R79, 2007.<br />

[370] P. Ongusaha, T. Ouchi, K. Kim, E. Nytko, J. Kwak, R. Duda, C. Deng, and S. Lee.<br />

Brca1 shifts p53-mediated cellular outcomes towards irreversible growth arrest. Oncogene,<br />

22(24):3749–3758, 2003.<br />

[371] P. C. Orban, D. Chui, and J. D. Marth. Tissue- and site-specific DNA recombination<br />

in transgenic mice. Proceedings <strong>of</strong> the National Academy <strong>of</strong> Sciences <strong>of</strong> the United<br />

States <strong>of</strong> America, 89(15):6861–6865, Aug. 1992.<br />

[372] U. Ørom, T. Derrien, M. Beringer, K. Gumireddy, A. Gardini, G. Bussotti, F. Lai,<br />

M. Zytnicki, C. Notredame, Q. Huang, et al. Long noncoding rnas with enhancer-like<br />

function in human cells. Cell, 143(1):46–58, 2010.<br />

[373] E. Ostrakhovitch, P. Olsson, S. Jiang, and M. Cherian. Interaction <strong>of</strong> metallothionein<br />

with tumor suppressor p53 protein. FEBS letters, 580(5):1235–1238, 2006.<br />

[374] T. Ozawa, C. W. Brennan, L. Wang, M. Squatrito, T. Sasayama, M. Nakada, J. T.<br />

Huse, A. Pedraza, S. Utsuki, Y. Yasui, A. Tandon, E. I. Fomchenko, H. Oka, R. L.<br />

Levine, K. Fujii, M. Ladanyi, and E. C. Holland. PDGFRA gene rearrangements are<br />

frequent genetic events in PDGFRA-amplified glioblastomas. Genes & Development,<br />

24(19):2205–2218, Oct. 2010.<br />

[375] L. Pacey, J. Stead, A. Gleave, K. Tomczyk, and L. Doering. <strong>Neural</strong> stem cell culture:<br />

neurosphere generation, microscopical analysis and cryopreservation. Nat. Protoc,<br />

215:1–14, 2006.<br />

[376] T. Palmer, P. Schwartz, P. Taupin, B. Kaspar, S. Stein, and F. Gage. Cell culture:<br />

Progenitor cells from human brain after death. Nature, 411(6833):42–43, 2001.<br />

[377] P. Pandolfi. Breast cancerÂŮloss <strong>of</strong> pten predicts resistance to treatment. New England<br />

Journal <strong>of</strong> Medicine, 351(22):2337–2338, 2004.<br />

[378] K. Paraiso, Y. Xiang, V. Rebecca, E. Abel, Y. Chen, A. Munko, E. Wood, I. Fedorenko,<br />

V. Sondak, A. Anderson, et al. Pten loss confers braf inhibitor resistance<br />

to melanoma cells through the suppression <strong>of</strong> bim expression. Cancer research,<br />

71(7):2750–2760, 2011.


[379] D. Park and J. Rich. Biology <strong>of</strong> glioma cancer stem cells. Molecules and cells, 28(1):7–<br />

12, 2009.<br />

[380] H. Park, I. Han, H. Kwon, and E. Oh. Focal adhesion kinase regulates syndecan-<br />

2–mediated tumorigenic activity <strong>of</strong> ht1080 fibrosarcoma cells. Cancer research,<br />

65(21):9899–9905, 2005.<br />

[381] J. Park, J. Jung, M. Seo, S. Kang, Y. Lee, and K. Kang. Dner modulates adipogenesis<br />

<strong>of</strong> human adipose tissue-derived mesenchymal stem cells via regulation <strong>of</strong> cell<br />

proliferation. Cell proliferation, 43(1):19–28, 2010.<br />

[382] D. Parry, D. Mahony, K. Wills, and E. Lees. Cyclin d-cdk subunit arrangement is<br />

dependent on the availability <strong>of</strong> competing ink4 and p21 class inhibitors. Molecular<br />

and cellular biology, 19(3):1775, 1999.<br />

[383] D. Parsons, S. Jones, X. Zhang, J. Lin, and R. Leary. An integrated genomic analysis<br />

<strong>of</strong> human glioblastoma multiforme. Science, 2008.<br />

[384] H. Pasantes-Morales and A. Schousboe. Role <strong>of</strong> taurine in osmoregulation in brain<br />

cells: mechanisms and functional implications. Amino Acids, 12(3):281–292, 1997.<br />

[385] L. Patrawala, T. Calhoun, R. Schneider-Broussard, J. Zhou, K. Claypool, and D. Tang.<br />

Side population is enriched in tumorigenic, stem-like cancer cells, whereas abcg2+ and<br />

abcg2- cancer cells are similarly tumorigenic. Cancer research, 65(14):6207, 2005.<br />

[386] K. Paulsson, M. Kleijmeer, J. Griffith, M. Jevon, S. Chen, P. Anderson, H. Sjögren,<br />

S. Li, and P. Wang. Association <strong>of</strong> tapasin and copi provides a mechanism for the<br />

retrograde transport <strong>of</strong> major histocompatibility complex (mhc) class i molecules from<br />

the golgi complex to the endoplasmic reticulum. Journal <strong>of</strong> Biological Chemistry,<br />

277(21):18266, 2002.<br />

[387] G. Paxinos, J. Mai, and S. O. service). The Human Nervous System. Elsevier Academic<br />

Press London, 2004.<br />

[388] G. Pearson, F. Robinson, T. Gibson, B. Xu, M. Karandikar, K. Berman, and M. Cobb.<br />

Mitogen-activated protein (map) kinase pathways: regulation and physiological functions.<br />

Endocrine reviews, 22(2):153–183, 2001.<br />

[389] L. Pevny and M. Placzek. Sox genes and neural progenitor identity. Current opinion<br />

in neurobiology, 15(1):7–13, 2005.<br />

[390] H. S. Phillips, S. Kharbanda, R. Chen, W. F. Forrest, R. H. Soriano, T. D. Wu,<br />

A. Misra, J. M. Nigro, H. Colman, L. Soroceanu, P. M. Williams, Z. Modrusan, B. G.<br />

Feuerstein, and K. Aldape. Molecular subclasses <strong>of</strong> high-grade glioma predict prognosis,<br />

delineate a pattern <strong>of</strong> disease progression, and resemble stages in neurogenesis.<br />

Cancer Cell, 9(3):157–173, Mar. 2006.<br />

[391] J. Phillips, E. Huillard, A. Robinson, A. Ward, D. Lum, M. Polley, S. Rosen, D. Rowitch,<br />

and Z. Werb. Heparan sulfate sulfatase sulf2 regulates pdgfrα signaling and<br />

growth in human and mouse malignant glioma. The Journal <strong>of</strong> clinical investigation,<br />

122(3):911, 2012.<br />

[392] S. Piccirillo, B. Reynolds, N. Zanetti, G. Lamorte, E. Binda, G. Broggi, H. Brem,<br />

A. Olivi, F. Dimeco, A. Vescovi, et al. Bone morphogenetic proteins inhibit the<br />

tumorigenic potential <strong>of</strong> human brain tumour-initiating cells. Nature, 444(7120):761,<br />

2006.<br />

[393] S. G. Piccirillo, E. Binda, R. Fiocco, A. L. Vescovi, and K. Shah. Brain cancer stem<br />

cells. Journal <strong>of</strong> molecular medicine, 87(11):1087–95, 2009.<br />

[394] R. Piet, L. Vargová, E. Syková, D. Poulain, and S. Oliet. Physiological contribution<br />

<strong>of</strong> the astrocytic environment <strong>of</strong> neurons to intersynaptic crosstalk. Proceedings <strong>of</strong> the<br />

National Academy <strong>of</strong> Sciences <strong>of</strong> the United States <strong>of</strong> America, 101(7):2151, 2004.<br />

[395] D. Pinto and H. Clevers. Wnt control <strong>of</strong> stem cells and differentiation in the intestinal<br />

epithelium. Experimental cell research, 306(2):357–363, 2005.<br />

[396] A. Pitre, N. Davis, M. Paul, A. Orr, and O. Skalli. Synemin promotes akt-dependent<br />

glioblastoma cell proliferation by antagonizing pp2a. Molecular biology <strong>of</strong> the cell,<br />

23(7):1243–1253, 2012.<br />

[397] S. Pleasure, C. Page, and V. Lee. Pure, postmitotic, polarized human neurons derived<br />

from ntera 2 cells provide a system for expressing exogenous proteins in terminally<br />

differentiated neurons. The Journal <strong>of</strong> neuroscience, 12(5):1802, 1992.


[398] E. Poch, R. Miñambres, E. Mocholí, C. Ivorra, A. Pérez-Aragó, C. Guerri, I. Pérez-<br />

Roger, and R. M. Guasch. RhoE interferes with Rb inactivation and regulates the<br />

proliferation and survival <strong>of</strong> the U87 human glioblastoma cell line. Experimental cell<br />

research, 313(4):719–731, Feb. 2007.<br />

[399] S. Pollard, A. Benchoua, and S. Lowell. <strong>Neural</strong> stem cells, neurons, and glia. Methods<br />

in enzymology, 418:151–169, 2006.<br />

[400] S. Pollard and L. Conti. Investigating radial glia in vitro. Progress in neurobiology,<br />

83(1):53–67, 2007.<br />

[401] S. Pollard, R. Wallbank, S. Tomlinson, L. Grotewold, and A. Smith. Fibroblast growth<br />

factor induces a neural stem cell phenotype in foetal forebrain progenitors and during<br />

embryonic stem cell differentiation. Molecular and Cellular Neuroscience, 38(3):393–<br />

403, 2008.<br />

[402] S. Pollard, K. Yoshikawa, I. Clarke, D. Danovi, S. Stricker, R. Russell, J. Bayani,<br />

R. Head, M. Lee, M. Bernstein, et al. <strong>Glioma</strong> stem cell lines expanded in adherent<br />

culture have tumor-specific phenotypes and are suitable for chemical and genetic<br />

screens. Cell <strong>Stem</strong> Cell, 4(6):568–580, 2009.<br />

[403] S. M. Pollard. Adherent <strong>Neural</strong> <strong>Stem</strong> (NS) <strong>Cells</strong> from Fetal and Adult Forebrain.<br />

Cerebral Cortex, 16(Supplement 1):i112–i120, July 2006.<br />

[404] S. M. Pollard, K. Yoshikawa, I. D. Clarke, D. Danovi, S. Stricker, R. Russell, J. Bayani,<br />

R. Head, M. Lee, M. Bernstein, J. A. Squire, A. Smith, and P. Dirks. <strong>Glioma</strong> stem cell<br />

lines expanded in adherent culture have tumor-specific phenotypes and are suitable<br />

for chemical and genetic screens. Cell <strong>Stem</strong> Cell, 4(6):568–80, 2009.<br />

[405] K. Pollock, P. Stroemer, S. Patel, L. Stevanato, A. Hope, E. Miljan, Z. Dong,<br />

H. Hodges, J. Price, and J. Sinden. A conditionally immortal clonal stem cell line<br />

from human cortical neuroepithelium for the treatment <strong>of</strong> ischemic stroke. Experimental<br />

neurology, 199(1):143–155, 2006.<br />

[406] R. Popovic and J. Licht. Mek and maf in myeloma therapy. Blood, 117(8):2300–2302,<br />

2011.<br />

[407] A. Popovi&cacute, A. Demirovi&cacute, B. Spaji&cacute, G. Štimac, and<br />

D. B Krušlin. Expression and prognostic role <strong>of</strong> syndecan-2 in prostate cancer. Prostate<br />

cancer and prostatic diseases, 13(1):78–82, 2009.<br />

[408] T. D. Portal. Tcga data portal: An integrated genomic analysis identifies clinically<br />

relevant subtypes <strong>of</strong> glioblastoma characterized by abnormalities in pdgfra, idh1, egfr<br />

and nf1.<br />

[409] K. Pruitt, T. Tatusova, and D. Maglott. Ncbi reference sequences (refseq): a curated<br />

non-redundant sequence database <strong>of</strong> genomes, transcripts and proteins. Nucleic acids<br />

research, 35(suppl 1):D61–D65, 2007.<br />

[410] PubMed. Pubmed. http://www.ncbi.nlm.nih.gov/pubmed.<br />

[411] R. Puca, L. Nardinocchi, G. Bossi, A. Sacchi, G. Rechavi, D. Givol, and G. D’Orazi.<br />

Restoring wtp53 activity in hipk2 depleted mcf7 cells by modulating metallothionein<br />

and zinc. Experimental cell research, 315(1):67–75, 2009.<br />

[412] Z. Qin, F. Ren, X. Xu, Y. Ren, H. Li, Y. Wang, Y. Zhai, and Z. Chang. Znf536, a<br />

novel zinc finger protein specifically expressed in the brain, negatively regulates neuron<br />

differentiation by repressing retinoic acid-induced gene transcription. Molecular and<br />

cellular biology, 29(13):3633, 2009.<br />

[413] A. Quiñones Hinojosa and N. Sanai. Cellular composition and cytoarchitecture <strong>of</strong><br />

the adult human subventricular zone: a niche <strong>of</strong> neural stem cells. The Journal <strong>of</strong><br />

comparative neurology, 2006.<br />

[414] F. Radtke and H. Clevers. Self-renewal and cancer <strong>of</strong> the gut: two sides <strong>of</strong> a coin.<br />

Science, 307(5717):1904, 2005.<br />

[415] B. Ragel, W. Couldwell, D. Gillespie, and R. Jensen. Identification <strong>of</strong> hypoxia-induced<br />

genes in a malignant glioma cell line (u-251) by cdna microarray analysis. Neurosurgical<br />

review, 30(3):181–187, 2007.<br />

[416] H. Rajagopalan, P. Jallepalli, C. Rago, V. Velculescu, K. Kinzler, B. Vogelstein,<br />

and C. Lengauer. Inactivation <strong>of</strong> hcdc4 can cause chromosomal instability. Nature,<br />

428(6978):77–81, 2004.


[417] P. Rakic. Guidance <strong>of</strong> neurons migrating to the fetal monkey neocortex. Brain research,<br />

33(2):471, 1971.<br />

[418] P. Rakic. Elusive radial glial cells: historical and evolutionary perspective. Glia,<br />

43(1):19–32, 2003.<br />

[419] S. Ramaswamy, K. Ross, E. Lander, and T. Golub. A molecular signature <strong>of</strong> metastasis<br />

in primary solid tumors. Nature genetics, 33(1):49–54, 2002.<br />

[420] L. Rambhatla, S. Ram-Mohan, J. Cheng, and J. Sherley. Immortal dna strand cosegregation<br />

requires p53/impdh–dependent asymmetric self-renewal associated with adult<br />

stem cells. Cancer research, 65(8):3155, 2005.<br />

[421] L. B. Rangel, R. Agarwal, C. A. Sherman-Baust, V. Mello-Coelho, E. S. Pizer, H. Ji,<br />

D. D. Taub, and P. J. Morin. Anomalous expression <strong>of</strong> the HLA-DR alpha and beta<br />

chains in ovarian and other cancers. Cancer biology and therapy, 3(10):1021–7, 2004.<br />

[422] P. Rao, M. Jaggi, D. Smith, G. Hemstreet, and K. Balaji. Metallothionein 2a interacts<br />

with the kinase domain <strong>of</strong> pkcµ in prostate cancer. Biochemical and biophysical<br />

research communications, 310(3):1032–1038, 2003.<br />

[423] S. K. Rao, J. Edwards, A. D. Joshi, I.-M. Siu, and G. J. Riggins. A survey <strong>of</strong> glioblastoma<br />

genomic amplifications and deletions. Journal <strong>of</strong> neuro-oncology, 96(2):169–179,<br />

2010.<br />

[424] W. A. Redmond WL, Ruby CE. The role <strong>of</strong> ox40-mediated co-stimulation in t-cell<br />

activation and survival. Crit Rev Immunol, 29:187–201, 2009.<br />

[425] J. Rehwinkel, J. Raes, and E. Izaurralde. Nonsense-mediated mRNA decay: Target<br />

genes and functional diversification <strong>of</strong> effectors. Trends Biochem Sci, 31(11):639–46,<br />

2006.<br />

[426] K. Reilly, D. Loisel, and R. Bronson. Nf1; Trp53 mutant mice develop glioblastoma<br />

with evidence <strong>of</strong> strain-specific effects. Nature Genetics, 2000.<br />

[427] B. Reynolds and S. Weiss. Generation <strong>of</strong> neurons and astrocytes from isolated cells<br />

<strong>of</strong> the adult mammalian central nervous system. Science, 255(5052):1707, 1992.<br />

[428] S. Riaz, E. Jauniaux, G. Stern, and H. Bradford. The controlled conversion <strong>of</strong> human<br />

neural progenitor cells derived from foetal ventral mesencephalon into dopaminergic<br />

neurons in vitro. Developmental brain research, 136(1):27–34, 2002.<br />

[429] M. J. Riemenschneider, R. Büschges, M. Wolter, J. Reifenberger, J. Boström, J. A.<br />

Kraus, U. Schlegel, and G. Reifenberger. Amplification and overexpression <strong>of</strong> the<br />

MDM4 (MDMX) gene from 1q32 in a subset <strong>of</strong> malignant gliomas without TP53<br />

mutation or MDM2 amplification. Cancer Research, 59(24):6091–6096, Dec. 1999.<br />

[430] A. Rizki, V. Weaver, S. Lee, G. Rozenberg, K. Chin, C. Myers, J. Bascom, J. Mott,<br />

J. Semeiks, L. Grate, et al. A human breast cell model <strong>of</strong> preinvasive to invasive<br />

transition. Cancer research, 68(5):1378, 2008.<br />

[431] D. Robinson, Y. Wu, and S. Lin. The protein tyrosine kinase family <strong>of</strong> the human<br />

genome. biomedical science, 19:5548–5557, 2000.<br />

[432] R. Roel<strong>of</strong>s, D. Fischer, S. Houtman, J. Sluijs, W. Van Haren, F. Van Leeuwen, and<br />

E. Hol. Adult human subventricular, subgranular, and subpial zones contain astrocytes<br />

with a specialized intermediate filament cytoskeleton. Glia, 52(4):289–300, 2005.<br />

[433] S. Rosen and H. Lemjabbar-Alaoui. Sulf-2: an extracellular modulator <strong>of</strong> cell signaling<br />

and a cancer target candidate. Expert opinion on therapeutic targets, 14(9):935–949,<br />

2010.<br />

[434] L. Rosso and J. Mienville. Pituicyte modulation <strong>of</strong> neurohormone output. Glia,<br />

57(3):235–243, 2009.<br />

[435] A. Rousseau, C. Nutt, R. Betensky, A. Iafrate, M. Han, K. Ligon, D. Rowitch, and<br />

D. Louis. Expression <strong>of</strong> oligodendroglial and astrocytic lineage markers in diffuse<br />

gliomas: use <strong>of</strong> ykl-40, apoe, ascl1, and nkx2-2. Journal <strong>of</strong> Neuropathology & Experimental<br />

Neurology, 65(12):1149, 2006.<br />

[436] N. Roy, A. Benraiss, S. Wang, R. Fraser, R. Goodman, W. Couldwell, M. Nedergaard,<br />

A. Kawaguchi, H. Okano, and S. Goldman. Promoter-targeted selection and<br />

isolation <strong>of</strong> neural progenitor cells from the adult human ventricular zone. Journal <strong>of</strong><br />

neuroscience research, 59(3):321–331, 2000.


[437] S. Rybalkin, C. Yan, K. Bornfeldt, and J. Beavo. Cyclic gmp phosphodiesterases and<br />

regulation <strong>of</strong> smooth muscle function. Circulation research, 93(4):280–291, 2003.<br />

[438] T. Saito, K. Shibasaki, M. Kurachi, S. Puentes, M. Mikuni, and Y. Ishizaki. Cerebral<br />

capillary endothelial cells are covered by the VEGF-expressing foot processes <strong>of</strong><br />

astrocytes. Neuroscience Letters, 2011.<br />

[439] A. Sakurada, H. Hamada, S. Fukushige, T. Yokoyama, K. Yoshinaga, T. Furukawa,<br />

S. Sato, A. Yajima, M. Sato, S. Fujimura, et al. Adenovirus-mediated delivery <strong>of</strong><br />

the pten gene inhibits cell growth by induction <strong>of</strong> apoptosis in endometrial cancer.<br />

International journal <strong>of</strong> oncology, 15(6):1069–1074, 1999.<br />

[440] Y. Samuels, Z. Wang, A. Bardelli, N. Silliman, J. Ptak, S. Szabo, H. Yan, A. Gazdar,<br />

S. Powell, G. Riggins, et al. High frequency <strong>of</strong> mutations <strong>of</strong> the PIK3CA gene in<br />

human cancers. Science, 304(5670):554, 2004.<br />

[441] D. San, J. Ray, and F. Gage. Bipotent progenitor cell lines from the human cns.<br />

Nature biotechnology, 15(6):574–580, 1997.<br />

[442] N. Sanai, A. Alvarez-Buylla, and M. Berger. <strong>Neural</strong> stem cells and the origin <strong>of</strong><br />

gliomas. New England Journal <strong>of</strong> Medicine, 353(8):811–822, 2005.<br />

[443] N. Sanai, A. Tramontin, A. Quiñones-Hinojosa, N. Barbaro, N. Gupta, S. Kunwar,<br />

M. Lawton, M. McDermott, A. Parsa, J. Verdugo, et al. Unique astrocyte ribbon<br />

in adult human brain contains neural stem cells but lacks chain migration. Nature,<br />

427(6976):740–744, 2004.<br />

[444] M. Sarti, C. Sevignani, G. Calin, R. Aqeilan, M. Shimizu, F. Pentimalli, M. Picchio,<br />

A. Godwin, A. Rosenberg, A. Drusco, et al. Adenoviral transduction <strong>of</strong> testin gene<br />

into breast and uterine cancer cell lines promotes apoptosis and tumor reduction in<br />

vivo. Clinical cancer research, 11(2):806–813, 2005.<br />

[445] E. Schmidt, K. Ichimura, H. Goike, A. Moshref, L. Liu, and V. Collins. Mutational<br />

pr<strong>of</strong>ile <strong>of</strong> the pten gene in primary human astrocytic tumors and cultivated xenografts.<br />

Journal <strong>of</strong> Neuropathology & Experimental Neurology, 58(11):1170, 1999.<br />

[446] M. Selbach, B. Schwanhausser, N. Thierfelder, Z. Fang, R. Khanin, and N. Rajewsky.<br />

Widespread changes in protein synthesis induced by microRNAs. Nature,<br />

455(7209):58–63, 2008.<br />

[447] D. Seo, J. Sung, H. Cho, H. Yi, K. Seo, I. Choi, D. Kim, J. Kim, A. El-Aty, H. Shin,<br />

et al. Gene expression pr<strong>of</strong>iling <strong>of</strong> cancer stem cell in human lung adenocarcinoma<br />

a549 cells. Mol Cancer, 6(1):75, 2007.<br />

[448] J. Seoane, H. Le, L. Shen, S. Anderson, and J. Massagué. Integration <strong>of</strong> smad and<br />

forkhead pathways in the control <strong>of</strong> neuroepithelial and glioblastoma cell proliferation.<br />

Cell, 117(2):211–223, 2004.<br />

[449] B. Seri, J. Garcı a Verdugo, B. McEwen, and A. Alvarez-Buylla. Astrocytes give rise<br />

to new neurons in the adult mammalian hippocampus. The Journal <strong>of</strong> Neuroscience,<br />

21(18):7153, 2001.<br />

[450] R. Shai, T. Shi, T. J. Kremen, S. Horvath, L. M. Liau, T. F. Cloughesy, P. S. Mischel,<br />

and S. F. Nelson. Gene expression pr<strong>of</strong>iling identifies molecular subtypes <strong>of</strong> gliomas.<br />

Oncogene, 22(31):4918–4923, July 2003.<br />

[451] P. Shannon, A. Markiel, O. Ozier, N. Baliga, J. Wang, D. Ramage, N. Amin,<br />

B. Schwikowski, and T. Ideker. Cytoscape: a s<strong>of</strong>tware environment for integrated<br />

models <strong>of</strong> biomolecular interaction networks. Genome research, 13(11):2498–2504,<br />

2003.<br />

[452] Q. Shen, Y. Wang, E. Kokovay, G. Lin, S. Chuang, S. Goderie, B. Roysam, and<br />

S. Temple. Adult svz stem cells lie in a vascular niche: a quantitative analysis <strong>of</strong> niche<br />

cell-cell interactions. Cell <strong>Stem</strong> Cell, 3(3):289–300, 2008.<br />

[453] W. Shen, A. Balajee, J. Wang, H. Wu, C. Eng, P. Pandolfi, and Y. Yin. Essential role<br />

for nuclear pten in maintaining chromosomal integrity. Cell, 128(1):157–170, 2007.<br />

[454] C. Sherr and J. Roberts. CDK inhibitors: positive and negative regulators <strong>of</strong> G1-phase<br />

progression. Genes & development, 13(12):1501, 1999.<br />

[455] S. L. Shi W, Fan H and D. R. The tetraspanin cd9 associates with transmembrane<br />

tgf-alpha and regulates tgf-alpha-induced egf receptor activation and cell proliferation.<br />

J. Cell Biol, (148):591–602, 2000.


[456] T. Shima, N. Okumura, T. Takao, Y. Satomi, T. Yagi, M. Okada, and K. Nagai.<br />

Interaction <strong>of</strong> the sh2 domain <strong>of</strong> fyn with a cytoskeletal protein, β-adducin. Journal<br />

<strong>of</strong> Biological Chemistry, 276(45):42233–42240, 2001.<br />

[457] M. Shipitsin and K. Polyak. The cancer stem cell hypothesis: in search <strong>of</strong> definitions,<br />

markers, and relevance. Laboratory Investigation, 88(5):459–463, 2008.<br />

[458] S. Singh, I. Clarke, M. Terasaki, V. Bonn, C. Hawkins, J. Squire, and P. Dirks. Identification<br />

<strong>of</strong> a cancer stem cell in human brain tumors. Cancer Research, 63(18):5821,<br />

2003.<br />

[459] S. Singh, C. Hawkins, I. Clarke, and J. Squire. Identification <strong>of</strong> human brain tumour<br />

initiating cells. Nature, 2004.<br />

[460] P. Singha, I. Yeh, M. Venkatachalam, P. Saikumar, et al. Transforming growth factor<br />

beta-inducible gene tmepai converts tgf-beta from a tumor suppressor to a tumor<br />

promoter in breast cancer. Cancer research, 70(15):6377–6383, 2010.<br />

[461] D. Smadja, C. d’Audigier, L. Weiswald, C. Badoual, V. Dangles-Marie, L. Mauge,<br />

S. Evrard, I. Laurendeau, F. Lallemand, S. Germain, et al. The wnt antagonist<br />

dickkopf-1 increases endothelial progenitor cell angiogenic potential. Arteriosclerosis,<br />

thrombosis, and vascular biology, 30(12):2544–2552, 2010.<br />

[462] K. Smith, M. Luong, and G. Stein. Pluripotency: toward a gold standard for human<br />

es and ips cells. Journal <strong>of</strong> cellular physiology, 220(1):21–29, 2009.<br />

[463] B. J. M. D. C. B. G. R. Smith SJ, Long A. Pediatric high-grade glioma: identification<br />

<strong>of</strong> poly(adp-ribose) polymerase as a potential therapeutic target. Neuro-oncology,<br />

13:1171–1177, 2011.<br />

[464] G. Smyth. Linear models and empirical bayes methods for assessing differential expression<br />

in microarray experiments. Statistical applications in genetics and molecular<br />

biology, 3(1):3, 2004.<br />

[465] G. Smyth. Limma: linear models for microarray data. Bioinformatics and computational<br />

biology solutions using R and Bioconductor, pages 397–420, 2005.<br />

[466] G. Smyth and T. Speed. Normalization <strong>of</strong> cdna microarray data. Methods, 31(4):265–<br />

273, 2003.<br />

[467] D. A. Solomon, J.-S. Kim, W. Jean, and T. Waldman. Conspirators in a capital<br />

crime: co-deletion <strong>of</strong> p18INK4c and p16INK4a/p14ARF/p15INK4b in glioblastoma<br />

multiforme. Cancer Research, 68(21):8657–8660, Nov. 2008.<br />

[468] J. Soos, J. Krieger, O. Stüve, C. King, J. Patarroyo, K. Aldape, K. Wosik, A. Slavin,<br />

P. Nelson, J. Antel, et al. Malignant glioma cells use mhc class ii transactivator<br />

(ciita) promoters iii and iv to direct ifn-γ-inducible ciita expression and can function<br />

as nonpr<strong>of</strong>essional antigen presenting cells in endocytic processing and cd4+ t-cell<br />

activation. Glia, 36(3):391–405, 2001.<br />

[469] A. Stark, J. Brennecke, N. Bushati, R. B. Russell, and S. M. Cohen. Animal MicroR-<br />

NAs confer robustness to gene expression and have a significant impact on 3’UTR<br />

evolution. Cell, 123(6):1133–46, 2005.<br />

[470] A. Stark, J. Brennecke, R. B. Russell, and S. M. Cohen. Identification <strong>of</strong> Drosophila<br />

MicroRNA targets. PLoS Biol, 1(3):E60, 2003.<br />

[471] C. StrÃÂijbing, G. Ahnert-Hilger, J. Shan, B. Wiedenmann, J. Hescheler, and A. M.<br />

Wobus. Differentiation <strong>of</strong> pluripotent embryonic stem cells into the neuronal lineage<br />

in vitro gives rise to mature inhibitory and excitatory neurons. Mechanisms <strong>of</strong> Development,<br />

53(2):275–287, 1995.<br />

[472] R. Stupp, W. P. Mason, M. J. van den Bent, M. Weller, B. Fisher, M. J. B. Taphoorn,<br />

K. Belanger, A. A. Brandes, C. Marosi, U. Bogdahn, J. Curschmann, R. C. Janzer,<br />

S. K. Ludwin, T. Gorlia, A. Allgeier, D. Lacombe, J. G. Cairncross, E. Eisenhauer,<br />

R. O. Miriman<strong>of</strong>f, E. O. for Research, T. <strong>of</strong> Cancer Brain Tumor, R. Groups, and<br />

N. C. I. <strong>of</strong> Canada Clinical Trials Group. Radiotherapy plus concomitant and adjuvant<br />

temozolomide for glioblastoma. The New England journal <strong>of</strong> medicine, 352(10):987–<br />

996, Mar. 2005.<br />

[473] X. Su, C. Kong, and P. Stahl. Gapex-5 mediates ubiquitination, trafficking, and<br />

degradation <strong>of</strong> epidermal growth factor receptor. Journal <strong>of</strong> Biological Chemistry,<br />

282(29):21278–21284, 2007.


[474] A. Subramanian, P. Tamayo, V. Mootha, S. Mukherjee, B. Ebert, M. Gillette,<br />

A. Paulovich, S. Pomeroy, T. Golub, E. Lander, et al. Gene set enrichment analysis:<br />

a knowledge-based approach for interpreting genome-wide expression pr<strong>of</strong>iles.<br />

Proceedings <strong>of</strong> the National Academy <strong>of</strong> Sciences <strong>of</strong> the United States <strong>of</strong> America,<br />

102(43):15545–15550, 2005.<br />

[475] E. Sum, D. Segara, B. Duscio, M. Bath, A. Field, R. Sutherland, G. Lindeman, and<br />

J. Visvader. Overexpression <strong>of</strong> lmo4 induces mammary hyperplasia, promotes cell<br />

invasion, and is a predictor <strong>of</strong> poor outcome in breast cancer. Proceedings <strong>of</strong> the<br />

National Academy <strong>of</strong> Sciences <strong>of</strong> the United States <strong>of</strong> America, 102(21):7659–7664,<br />

2005.<br />

[476] L. Sun, W. Yan, Y. Wang, G. Sun, H. Luo, J. Zhang, X. Wang, Y. You, Z. Yang, and<br />

N. Liu. Microrna-10b induces glioma cell invasion by modulating mmp-14 and upar<br />

expression via hoxd10. Brain research, 1389:9–18, 2011.<br />

[477] N. Sun, T. Huiatt, D. Paulin, Z. Li, and R. Robson. Synemin interacts with the lim<br />

domain protein zyxin and is essential for cell adhesion and migration. Experimental<br />

cell research, 316(3):491–505, 2010.<br />

[478] P. Sun, S. Xia, B. Lal, C. Eberhart, A. Quinones-Hinojosa, J. Maciaczyk, W. Matsui,<br />

F. DiMeco, S. Piccirillo, A. Vescovi, et al. Dner, an epigenetically modulated gene,<br />

regulates glioblastoma-derived neurosphere cell differentiation and tumor propagation.<br />

<strong>Stem</strong> <strong>Cells</strong>, 27(7):1473–1486, 2009.<br />

[479] T. Sun, X. Wang, S. Xie, D. Zhang, X. Wang, B. Li, W. Ma, and H. Xin. A comparison<br />

<strong>of</strong> proliferative capacity and passaging potential between neural stem and progenitor<br />

cells in adherent and neurosphere cultures. International Journal <strong>of</strong> Developmental<br />

Neuroscience, 2011.<br />

[480] Y. Sun, W. Kong, A. Falk, J. Hu, L. Zhou, S. Pollard, and A. Smith. Cd133 (prominin)<br />

negative human neural stem cells are clonogenic and tripotent. PloS one, 4(5):e5498,<br />

2009.<br />

[481] Y. Sun, S. Pollard, L. Conti, M. Toselli, G. Biella, G. Parkin, L. Willatt, A. Falk,<br />

E. Cattaneo, and A. Smith. Long-term tripotent differentiation capacity <strong>of</strong> human<br />

neural stem (ns) cells in adherent culture. Molecular and Cellular Neuroscience,<br />

38(2):245–258, 2008.<br />

[482] C. Suo, A. Salim, K. S. Chia, Y. Pawitan, and S. Calza. Modified least-variant set<br />

normalization for miRNA microarray. RNA, 16(12):2293–303, 2010.<br />

[483] C. Svendsen, M. ter Borg, R. Armstrong, A. Rosser, S. Chandran, T. Ostenfeld, and<br />

M. Caldwell. A new method for the rapid and long term growth <strong>of</strong> human neural<br />

precursor cells. Journal <strong>of</strong> neuroscience methods, 85(2):141–152, 1998.<br />

[484] Y. Takamura, H. Ikeda, T. Kanaseki, M. Toyota, T. Tokino, K. Imai, K. Houkin, and<br />

N. Sato. Regulation <strong>of</strong> mhc class ii expression in glioma cells by class ii transactivator<br />

(ciita). Glia, 45(4):392–405, 2004.<br />

[485] O. Tam, A. Aravin, P. Stein, A. Girard, E. Murchison, S. Cheloufi, E. Hodges,<br />

M. Anger, R. Sachidanandam, R. Schultz, et al. Pseudogene-derived small interfering<br />

rnas regulate gene expression in mouse oocytes. Nature, 453(7194):534, 2008.<br />

[486] B. Tan, C. Park, L. Ailles, and I. Weissman. The cancer stem cell hypothesis: a work<br />

in progress. Laboratory investigation, 86(12):1203–1207, 2006.<br />

[487] N. Taniguchi, H. Taniura, M. Niinobe, C. Takayama, K. Tominaga-Yoshino, A. Ogura,<br />

and K. Yoshikawa. The postmitotic growth suppressor necdin interacts with a<br />

calcium-binding protein (nefa) in neuronal cytoplasm. Journal <strong>of</strong> Biological Chemistry,<br />

275(41):31674–31681, 2000.<br />

[488] M. Taniwaki, Y. Daigo, N. Ishikawa, A. Takano, T. Tsunoda, W. Yasui, K. Inai,<br />

N. Kohno, and Y. Nakamura. Gene expression pr<strong>of</strong>iles <strong>of</strong> small-cell lung cancers:<br />

molecular signatures <strong>of</strong> lung cancer. International journal <strong>of</strong> oncology, 29(3):567–576,<br />

2006.<br />

[489] A. Tarca, S. Draghici, P. Khatri, S. Hassan, P. Mittal, J. Kim, C. Kim, J. Kusanovic,<br />

and R. Romero. A novel signaling pathway impact analysis. Bioinformatics, 25(1):75,<br />

2009.


[490] P. A. C. t’Hoen, Y. Ariyurek, H. H. Thygesen, E. Vreugdenhil, R. H. A. M. Vossen,<br />

R. X. De Menezes, J. M. Boer, G.-J. B. Van Ommen, and J. T. Den Dunnen. Deep<br />

sequencing-based expression analysis shows major advances in robustness, resolution<br />

and inter-lab portability over five microarray platforms. Nucleic Acids Research,<br />

36(21):e141–e141, Oct. 2008.<br />

[491] C. Thomas, G. Ely, C. D. James, R. Jenkins, M. Kastan, A. Jedlicka, P. Burger, and<br />

R. Wharen. Glioblastoma-related gene mutations and over-expression <strong>of</strong> functional<br />

epidermal growth factor receptors in SKMG-3 glioma cells. Acta neuropathologica,<br />

101(6):605–615, June 2001.<br />

[492] J. Ting and J. Trowsdale. Genetic control <strong>of</strong> mhc class ii expression. Cell, 109(2):S21–<br />

S33, 2002.<br />

[493] E. Tobias, A. Hurlstone, E. MacKenzie, R. McFarlane, D. Black, et al. The tes gene at<br />

7q31. 1 is methylated in tumours and encodes a novel growth-suppressing lim domain<br />

protein. Oncogene, 20(22):2844, 2001.<br />

[494] V. Tropepe, M. Sibilia, B. Ciruna, J. Rossant, E. Wagner, and D. Kooy. Distinct neural<br />

stem cells proliferate in response to egf and fgf in the developing mouse telencephalon.<br />

Developmental biology, 208(1):166–188, 1999.<br />

[495] L. Trotman, X. Wang, A. Alimonti, Z. Chen, J. Teruya-Feldstein, H. Yang,<br />

N. Pavletich, B. Carver, C. Cordon-Cardo, H. Erdjument-Bromage, et al. Ubiquitination<br />

regulates pten nuclear import and tumor suppression. Cell, 128(1):141–156,<br />

2007.<br />

[496] A. B. Trovó-Marqui and E. H. Tajara. Neur<strong>of</strong>ibromin: a general outlook. Clinical<br />

genetics, 70(1):1–13, July 2006.<br />

[497] C. Tso, P. Shintaku, J. Chen, Q. Liu, J. Liu, Z. Chen, K. Yoshimoto, P. Mischel,<br />

T. Cloughesy, L. Liau, et al. Primary glioblastomas express mesenchymal stem-like<br />

properties. Molecular cancer research, 4(9):607–619, 2006.<br />

[498] A. Tsuchida, T. Okajima, K. Furukawa, T. Ando, H. Ishida, A. Yoshida, Y. Nakamura,<br />

R. Kannagi, M. Kiso, and K. Furukawa. Synthesis <strong>of</strong> disialyl lewis a (lea) structure in<br />

colon cancer cell lines by a sialyltransferase, st6galnac vi, responsible for the synthesis<br />

<strong>of</strong> α-series gangliosides. Journal <strong>of</strong> Biological Chemistry, 278(25):22787–22794, 2003.<br />

[499] N. Tsuji, K. Kondoh, M. Furuya, D. Kobayashi, A. Yagihashi, Y. Inoue, T. Meguro,<br />

S. Horita, H. Takahashi, and N. Watanabe. A novel aspartate protease gene, alp56,<br />

is related to morphological features <strong>of</strong> colorectal adenomas. International journal <strong>of</strong><br />

colorectal disease, 19(1):43–48, 2004.<br />

[500] V. Turk, B. Turk, G. Guncar, D. Turk, and J. Kos. Lysosomal cathepsins: structure,<br />

role in antigen processing and presentation, and cancer. Advances in enzyme<br />

regulation, 42:285, 2002.<br />

[501] A. Tzschach, A. Bisgaard, M. Kirchh<strong>of</strong>f, L. Graul-Neumann, H. Neitzel, S. Page,<br />

A. Ahmed, I. Müller, F. Erdogan, H. Ropers, et al. Chromosome aberrations involving<br />

10q22: report <strong>of</strong> three overlapping interstitial deletions and a balanced translocation<br />

disrupting c10orf11. European Journal <strong>of</strong> Human Genetics, 18(3):291–295, 2009.<br />

[502] N. Uchida, D. Buck, D. He, M. Reitsma, M. Masek, T. Phan, A. Tsukamoto, F. Gage,<br />

and I. Weissman. Direct isolation <strong>of</strong> human central nervous system stem cells. Proceedings<br />

<strong>of</strong> the National Academy <strong>of</strong> Sciences, 97(26):14720, 2000.<br />

[503] M. Van De Wiel, K. Kim, S. Vosse, W. Van Wieringen, S. Wilting, and B. Ylstra.<br />

Cghcall: calling aberrations for array cgh tumor pr<strong>of</strong>iles. Bioinformatics, 23(7):892–<br />

894, 2007.<br />

[504] J. Van Den Boom, M. Wolter, R. Kuick, D. Misek, A. Youkilis, D. Wechsler, C. Sommer,<br />

G. Reifenberger, and S. Hanash. <strong>Characterization</strong> <strong>of</strong> gene expression pr<strong>of</strong>iles<br />

associated with glioma progression using oligonucleotide-based microarray analysis<br />

and real-time reverse transcription-polymerase chain reaction. The American journal<br />

<strong>of</strong> pathology, 163(3):1033–1043, 2003.<br />

[505] A. Van der Krol, L. Mur, M. Beld, J. Mol, and A. Stuitje. Flavonoid genes in petunia:<br />

addition <strong>of</strong> a limited number <strong>of</strong> gene copies may lead to a suppression <strong>of</strong> gene<br />

expression. The Plant Cell Online, 2(4):291, 1990.


[506] B. van Houte, T. Binsl, H. Hettling, and J. Heringa. Cghnormaliter: a bioconductor<br />

package for normalization <strong>of</strong> array cgh data with many cnas. Bioinformatics,<br />

26(10):1366–1367, 2010.<br />

[507] F. van Ruissen and F. Baas. Serial analysis <strong>of</strong> gene expression (SAGE). Methods in<br />

molecular biology, 383:41–66, 2007.<br />

[508] S. Vatter, G. Pahlke, J. Deitmer, and G. Eisenbrand. Differential phosphodiesterase<br />

expression and cytosolic ca2+ in human cns tumour cells and in non-malignant and<br />

malignant cells <strong>of</strong> rat origin. Journal <strong>of</strong> neurochemistry, 93(2):321–329, 2005.<br />

[509] F. Vazquez, H. Vaucheret, R. Rajagopalan, C. Lepers, V. Gasciolli, A. Mallory,<br />

J. Hilbert, D. Bartel, and P. Crété. Endogenous trans-acting sirnas regulate the<br />

accumulation <strong>of</strong> arabidopsis mrnas. Molecular Cell, 16(1):69–79, 2004.<br />

[510] E. Venkatraman and A. Olshen. A faster circular binary segmentation algorithm for<br />

the analysis <strong>of</strong> array cgh data. Bioinformatics, 23(6):657–663, 2007.<br />

[511] R. G. W. Verhaak, K. A. Hoadley, E. Purdom, V. Wang, Y. Qi, M. D. Wilkerson,<br />

C. R. Miller, L. Ding, T. Golub, J. P. Mesirov, G. Alexe, M. Lawrence, M. O’Kelly,<br />

P. Tamayo, B. A. Weir, S. Gabriel, W. Winckler, S. Gupta, L. Jakkula, H. S. Feiler,<br />

J. G. Hodgson, C. D. James, J. N. Sarkaria, C. Brennan, A. Kahn, P. T. Spellman,<br />

R. K. Wilson, T. P. Speed, J. W. Gray, M. Meyerson, G. Getz, C. M. Perou, D. N.<br />

Hayes, and Cancer Genome Atlas Research Network. Integrated genomic analysis<br />

identifies clinically relevant subtypes <strong>of</strong> glioblastoma characterized by abnormalities<br />

in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell, 17(1):98–110, Jan. 2010.<br />

[512] A. Vescovi, E. Parati, A. Gritti, P. Poulin, M. Ferrario, E. Wanke, P. Frölichsthal-<br />

Schoeller, L. Cova, M. Arcellana-Panlilio, A. Colombo, et al. Isolation and cloning <strong>of</strong><br />

multipotential stem cells from the embryonic human cns and establishment <strong>of</strong> transplantable<br />

human neural stem cell lines by epigenetic stimulation. Experimental neurology,<br />

156(1):71–83, 1999.<br />

[513] A. Villa, E. Snyder, A. Vescovi, and A. Martínez-Serrano. Establishment and properties<br />

<strong>of</strong> a growth factor-dependent, perpetual neural stem cell line from the human<br />

cns. Experimental neurology, 161(1):67–84, 2000.<br />

[514] R. Vlietstra, D. van Alewijk, K. Hermans, G. van Steenbrugge, and J. Trapman.<br />

Frequent inactivation <strong>of</strong> pten in prostate cancer cell lines and xenografts. Cancer<br />

Research, 58(13):2720–2723, 1998.<br />

[515] B. Vogelstein, D. Lane, A. Levine, et al. Surfing the p53 network. Nature,<br />

408(6810):307–310, 2000.<br />

[516] A. Volterra. Astrocytes, from brain glue to communication elements: the revolution<br />

continues. Nature Reviews Neuroscience, 2005.<br />

[517] A. Von Deimling, A. Korshunov, and C. Hartmann. The next generation <strong>of</strong> glioma<br />

biomarkers: Mgmt methylation, braf fusions and idh1 mutations. Brain Pathology,<br />

21(1):74–87, 2011.<br />

[518] V. Vukicevic, A. Jauch, T. Dinger, L. Gebauer, V. Hornich, S. Bornstein, M. Ehrhart-<br />

Bornstein, and A. Müller. Genetic instability and diminished differentiation capacity in<br />

long-term cultured mouse neurosphere cells. Mechanisms <strong>of</strong> ageing and development,<br />

131(2):124–132, 2010.<br />

[519] S. Wada, M. Hamada, K. Kobayashi, and N. Satoh. Novel genes involved in canonical<br />

wnt beta-catenin signaling pathway in early ciona intestinalis embryos. Development,<br />

growth & differentiation, 50(4):215–227, 2008.<br />

[520] R. Wang, K. Chadalavada, J. Wilshire, U. Kowalik, K. Hovinga, A. Geber, B. Fligelman,<br />

M. Leversha, C. Brennan, and V. Tabar. Glioblastoma stem-like cells give rise<br />

to tumour endothelium. Nature, 468(7325):829–833, 2010.<br />

[521] T. Watanabe, T. Katagiri, M. Suzuki, F. Shimizu, T. Fujiwara, N. Kanemoto, Y. Nakamura,<br />

Y. Hirai, H. Maekawa, and E. Takahashi. Cloning and characterization <strong>of</strong> two<br />

novel human cdnas (nell1 and nell2) encoding proteins with six egf-like repeats. Genomics,<br />

38(3):273–276, 1996.<br />

[522] T. Watanabe, S. Nobusawa, P. Kleihues, and H. Ohgaki. Idh1 mutations are early<br />

events in the development <strong>of</strong> astrocytomas and oligodendrogliomas. The American<br />

journal <strong>of</strong> pathology, 174(4):1149, 2009.


[523] T. Watanabe, A. Takeda, T. Tsukiyama, K. Mise, T. Okuno, H. Sasaki, N. Minami,<br />

and H. Imai. Identification and characterization <strong>of</strong> two novel classes <strong>of</strong> small rnas<br />

in the mouse germline: retrotransposon-derived sirnas in oocytes and germline small<br />

rnas in testes. Genes & development, 20(13):1732, 2006.<br />

[524] T. Watanabe, Y. Totoki, A. Toyoda, M. Kaneda, S. Kuramochi-Miyagawa, Y. Obata,<br />

H. Chiba, Y. Kohara, T. Kono, T. Nakano, et al. Endogenous sirnas from naturally<br />

formed dsrnas regulate transcripts in mouse oocytes. Nature, 453(7194):539–543, 2008.<br />

[525] S. Weiss, C. Dunne, J. Hewson, C. Wohl, M. Wheatley, A. Peterson, and B. Reynolds.<br />

Multipotent cns stem cells are present in the adult mammalian spinal cord and ventricular<br />

neuroaxis. The Journal <strong>of</strong> neuroscience, 16(23):7599, 1996.<br />

[526] P. Wen and S. Kesari. Malignant gliomas in adults. New England Journal <strong>of</strong> Medicine,<br />

359(5):492–507, 2008.<br />

[527] W. Wick, U. Naumann, and M. Weller. Transforming growth factor-beta: A molecular<br />

target for the future therapy <strong>of</strong> glioblastoma. Current pharmaceutical design,<br />

12(3):341–349, 2006.<br />

[528] E. Wijaya, M. Frith, Y. Suzuki, and P. Horton. Recount: expectation maximization<br />

based error correction tool for next generation sequencing data. In Genome Inform,<br />

volume 23, pages 189–201, 2009.<br />

[529] R. Williams and K. Herrup. The Control <strong>of</strong> Neuron Number. Annual Review <strong>of</strong><br />

Neuroscience, 11(1):423–453, 1988.<br />

[530] P. Wolters, M. Laig-Webster, and G. Caughey. Dipeptidyl peptidase i cleaves matrixassociated<br />

proteins and is expressed mainly by mast cells in normal dog airways.<br />

American journal <strong>of</strong> respiratory cell and molecular biology, 22(2):183, 2000.<br />

[531] L. Xu, Y. Shi, G. Petrovics, C. Sun, M. Makarem, W. Zhang, I. Sesterhenn,<br />

D. McLeod, L. Sun, J. Moul, et al. Pmepa1, an androgen-regulated nedd4-binding protein,<br />

exhibits cell growth inhibitory function and decreased expression during prostate<br />

cancer progression. Cancer research, 63(15):4299, 2003.<br />

[532] X. Xu, J. Zhao, Z. Xu, B. Peng, Q. Huang, E. Arnold, and J. Ding. Structures <strong>of</strong> human<br />

cytosolic nadp-dependent isocitrate dehydrogenase reveal a novel self-regulatory<br />

mechanism <strong>of</strong> activity. Journal <strong>of</strong> Biological Chemistry, 279(32):33946–33957, 2004.<br />

[533] V. Yadav and M. Denning. Fyn is induced by ras/pi3k/akt signaling and is required<br />

for enhanced invasion/migration. Molecular carcinogenesis, 50(5):346–352, 2011.<br />

[534] K. Yamada and M. Watanabe. Cytodifferentiation <strong>of</strong> Bergmann glia and its relationship<br />

with Purkinje cells. Anatomical science international, 77(2):94–108, 2002.<br />

[535] H. Yan, D. Parsons, G. Jin, R. McLendon, B. Rasheed, W. Yuan, I. Kos, I. Batinic-<br />

Haberle, S. Jones, G. Riggins, et al. Idh1 and idh2 mutations in gliomas. New England<br />

Journal <strong>of</strong> Medicine, 360(8):765–773, 2009.<br />

[536] J. Yan, L. Xu, A. Welsh, G. Hatfield, T. Hazel, K. Johe, and V. Koliatsos. Extensive<br />

neuronal differentiation <strong>of</strong> human neural stem cell grafts in adult rat spinal cord. PLoS<br />

medicine, 4(2):e39, 2007.<br />

[537] K. Yap, S. Li, A. Muñoz-Cabello, S. Raguz, L. Zeng, S. Mujtaba, J. Gil, M. Walsh,<br />

and M. Zhou. Molecular interplay <strong>of</strong> the noncoding rna anril and methylated histone<br />

h3 lysine 27 by polycomb cbx7 in transcriptional silencing <strong>of</strong> ink4a. Molecular cell,<br />

38(5):662–674, 2010.<br />

[538] S. Yekta, I. Shih, et al. Microrna-directed cleavage <strong>of</strong> hoxb8 mrna. Science,<br />

304(5670):594, 2004.<br />

[539] Q. Ying and A. Smith. Defined conditions for neural commitment and differentiation.<br />

Methods in enzymology, 365:327–341, 2003.<br />

[540] A. Yool. Aquaporins: multiple roles in the central nervous system. The Neuroscientist,<br />

13(5):470, 2007.<br />

[541] H. You, K. Yamamoto, and T. Mak. Regulation <strong>of</strong> transactivation-independent<br />

proapoptotic activity <strong>of</strong> p53 by foxo3a. Proceedings <strong>of</strong> the National Academy <strong>of</strong> Sciences,<br />

103(24):9051, 2006.


[542] J. Yu, K. Ohuchida, K. Nakata, K. Mizumoto, L. Cui, H. Fujita, H. Yamaguchi,<br />

T. Egami, H. Kitada, M. Tanaka, et al. Lim only 4 is overexpressed in late stage<br />

pancreas cancer. Mol Cancer, 7:93, 2008.<br />

[543] X. Yuan, J. Curtin, Y. Xiong, G. Liu, S. Waschsmann-Hogiu, D. Farkas, K. Black, and<br />

J. Yu. Isolation <strong>of</strong> cancer stem cells from adult glioblastoma multiforme. Oncogene,<br />

23(58):9392–9400, 2004.<br />

[544] S. Yun, K. Byun, J. Bhin, J. Oh, L. Nhung, D. Hwang, and B. Lee. <strong>Transcriptional</strong><br />

regulatory networks associated with self-renewal and differentiation <strong>of</strong> neural stem<br />

cells. Journal <strong>of</strong> cellular physiology, 225(2):337–347, 2010.<br />

[545] Z. Zador, O. Bloch, X. Yao, and G. Manley. Aquaporins: role in cerebral edema and<br />

brain water balance. Progress in brain research, 161:185–194, 2007.<br />

[546] D. Zagzag, K. Salnikow, L. Chiriboga, H. Yee, L. Lan, M. A. Ali, R. Garcia, S. Demaria,<br />

and E. W. Newcomb. Downregulation <strong>of</strong> major histocompatibility complex<br />

antigens in invading glioma cells: stealth invasion <strong>of</strong> the brain. Laboratory investigation;<br />

a journal <strong>of</strong> technical methods and pathology, (3):328–41.<br />

[547] X. Zhang, Z. Lian, C. Padden, M. Gerstein, J. Rozowsky, M. Snyder, T. Gingeras,<br />

P. Kapranov, S. Weissman, and P. Newburger. A myelopoiesis-associated regulatory<br />

intergenic noncoding rna transcript within the human hoxa cluster. Blood,<br />

113(11):2526–2534, 2009.<br />

[548] S. Zhao, Y. Lin, W. Xu, W. Jiang, Z. Zha, P. Wang, W. Yu, Z. Li, L. Gong, Y. Peng,<br />

et al. <strong>Glioma</strong>-derived mutations in idh1 dominantly inhibit idh1 catalytic activity and<br />

induce hif-1{alpha}. Science Signalling, 324(5924):261, 2009.<br />

[549] H. Zheng, H. Ying, H. Yan, A. Kimmelman, and D. Hiller. p53 and Pten control<br />

neural and glioma stem/progenitor cell renewal and differentiation. Nature, 2008.<br />

[550] Y. Zhu, P. Ghosh, P. Charnay, D. Burns, and L. Parada. Neur<strong>of</strong>ibromas in nf1:<br />

Schwann cell origin and role <strong>of</strong> tumor environment. Science, 296(5569):920, 2002.<br />

[551] Z. Zhuang, P. Jian, L. Longjiang, H. Bo, and X. Wenlin. Oral cancer cells with<br />

different potential <strong>of</strong> lymphatic metastasis displayed distinct biologic behaviors and<br />

gene expression pr<strong>of</strong>iles. Journal <strong>of</strong> Oral Pathology & Medicine, 39(2):168–175, 2010.<br />

[552] G. Zupanc and S. Clint. Potential role <strong>of</strong> radial glia in adult neurogenesis <strong>of</strong> teleost<br />

fish. Glia, 43(1):77–86, 2003.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!