29.12.2012 Views

11-13 May 2012 Helsingør - Denmark www.networkbio.org

11-13 May 2012 Helsingør - Denmark www.networkbio.org

11-13 May 2012 Helsingør - Denmark www.networkbio.org

SHOW MORE
SHOW LESS

Create successful ePaper yourself

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

Network<br />

Medicine<br />

<strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong><br />

<strong>Helsingør</strong> - <strong>Denmark</strong><br />

<strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong>


Friday <strong>11</strong>th may <strong>2012</strong><br />

09:00 - 15:00 Registration Open<br />

Session I: KINOME BIOLOGY<br />

Chair: Blagoy Blagoev - University of Southern <strong>Denmark</strong> Session<br />

Programme<br />

15:00 Welcome by Rune Linding<br />

15:10 - 16:00 OPENING LETURE: Eng Lim Goh (SGI, USA) - New Developments in Computing for the Life<br />

Sciences Researcher<br />

16:00 - 16:30 Andrea Califano (Columbia University, New York NY) - Dissecting and interrogating signaling<br />

networks in human malignancies<br />

16:30 - 16:45 Coffee Break and Sponsor Exhibition<br />

16:45 - 17:15 Rune Linding (DTU, Lyngby DK) - Modeling Cancer Kinome Networks<br />

17:15 - 17:30 Gianni Cesareni (Tor Vergata, Rome, Italy) - Mapping the human phosphatome on growth<br />

pathways<br />

17:30 - 17:50 Sponsored Talk (SBV Improver) - Julia Hoeng – Verification of System Biology Research in<br />

the age of Collaborative Competition<br />

17:50 - 18:00 Break<br />

18:00 - 18:15 Sol Efroni (Bar Ilan University, Israel) - Network-based metrics reveals a novel role for<br />

hsa-miR-9 and drug control over the p38 network in glioblastoma multiforme progression<br />

18:15 - 18:45 Garry Nolan (Stanford, San Francisco CA)<br />

19:00 - Poster Session, Drinks and Welcome Dinner at Hotel<br />

Saturday 12th may <strong>2012</strong><br />

Session II: DISEASE NETWORKS<br />

Chair: Ramneek Gutpa - Technical Univeristy of <strong>Denmark</strong> Session<br />

09:10 - 10:00 KEYNOTE LECTURE: Norbert Perrimon (Harvard Medical School, Boston USA) - Building<br />

and validating signaling networks in Drosophila<br />

10:00 - 10:30 Dana Pe’er (Columbia,USA) - On the road to personalized therapy, a systems approach<br />

10:30 - <strong>11</strong>:00 Marc Vidal (DFCI, Boston USA) - Interactome Networks and Human Disease<br />

<strong>11</strong>:00 - <strong>11</strong>:30 Coffe Break and Sponsor Exhibition<br />

<strong>11</strong>:30 - <strong>11</strong>:45 Stephan M. Feller (Oxford, UK) - How are complex signal computations in cells accomplished<br />

by multiprotein complexes assembled on ‘intrinsically disordered’ platform proteins?<br />

The N-terminal folding nucleation (NFN)hypothesis<br />

12:45 -12:00 Theo Knijnenburg (NCI,Amsterdam, NL) - Drug sensitivity of cancer cell lines explained as a<br />

logic combination of mutations<br />

12:00 - 12:30 Søren Brunak (DTU, Lyngby DK) - Interfacing disease phenotypes from electronic patient<br />

records to the underlying network biology<br />

12:30 - <strong>13</strong>:30 Lunch with Sponsored Talk (Thermo Fisher) - Christian Kelstrup (NNF-CPR, Copenhagen,<br />

DK) Optimized Fast and Sensitive Acquisition Methods for Shotgun Proteomics on a Quadru<br />

pole Orbitrap Mass Spectrometer<br />

Programme<br />

Session III: NETWORK DRUGS<br />

Chair: Christopher Workman - Technical University of <strong>Denmark</strong><br />

<strong>13</strong>:30 - 14:20 KEYNOTE LECTURE: Michael Yaffe (MIT, Cambridge USA)<br />

14:20 - 14:50 Matt Onsum (Merrimack Pharmaceuticals, USA) - Using systems biology to accelerate onco<br />

logy drug development<br />

14:50 - 15:15 Coffee Break and Sponsor Exhibition<br />

15:15 - 15:30 Ruth Hüttenhain (ETH Zurich, CH) - A mass spectrometric map for reproducible quantification<br />

of cancer associated proteins in body fluids<br />

15:30 - 16:00 Janine Erler (BRIC, Copenhagen DK) - Molecular networks associated with<br />

cancer progression<br />

16:00 - 16:30 Nevan Krogan (UCFS, San Francisco USA) - Functional Insights from Protein-<br />

Protein and Genetic Interaction Maps<br />

16:30 - 18:30 Poster Session and Sponsor Exhibition<br />

19:00 Transfer to Louisiana Museum for Gala Dinner<br />

20:00 Welcome Speech by SGI<br />

23:00 Buses back to Hotels<br />

Sunday <strong>13</strong>th may <strong>2012</strong><br />

Session IV: INTEGRATIVE NETWORK BIOLOGY<br />

Chair: tba<br />

09:00 -09:50 KEYNOTE LECTURE: Ruedi Aebersold (ETH, Switzerland) - Network Driven Protein Biomar<br />

ker Discovery and Validation<br />

09:50 - 10:20 Anne Claude Gavin (EMBL, Heidelberg DE) - Expanding the interaction space;<br />

protein-metabolite networks<br />

10:20 - 10:45 Coffee Break and Sponsor Exhibition<br />

10:45 - <strong>11</strong>:00 Ulrich Stelzl (MPI Molecular Genetics, Berlin, Germany) A Y2H-seq approach to define the<br />

protein ethyltransferase interactome<br />

<strong>11</strong>:00 - <strong>11</strong>:30 Marian Walhout (UMASS-MED, Worcester MA) - Gene regulatory networks<br />

<strong>11</strong>:30 - 12:00 Ben Lehner (CRG, Barcelona, Spain) - The biology of individuals<br />

12:00 - 12:30 Bernhard Pallson (DTU/UCSD, Lyngby, DK / San Diego, CA) - Systems Biology of<br />

Metabolism<br />

2 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 3


ENG LIM GOh, Ph.D.<br />

SVP & ChIEf TEChNOLOGY OffICER AT SGI<br />

abStract For SPeakerS<br />

Dr. Eng Lim Goh joined SGI in 1989, becoming a chief engineer in 1998 and<br />

then chief technology officer in 2001. He oversees technical computing programs<br />

with the goal to develop the next generation computer architecture for<br />

the new many-core era.<br />

In 2005, InfoWorld named Dr. Goh one of the World’s 25 Most Influential<br />

CTOs. That same year he was also included in the HPCwire list of “15 People<br />

to Watch.” In 2007, he was named “Champions 2.0” of the industry by BioIT<br />

World magazine, and received the HPC Community Recognition Award from<br />

HPCwire. Dr. Goh is a frequent industry speaker and he continues to discuss,<br />

in different forums, innovative technologies and their applications.<br />

Before joining SGI, Dr. Goh worked for Intergraph Systems, Schlumberger Wireline and Shell Research.<br />

A Shell Cambridge University Scholar, Dr. Goh completed his Ph.D. research and dissertation on parallel<br />

architectures and computer graphics, and holds a first-class honors degree in mechanical engineering from<br />

Birmingham University in the U.K.<br />

Dr. Goh has been granted four U.S. patents, two of which as the inventor and the others as co-inventor.<br />

New Developments in Computing for the Life Sciences Researcher<br />

abStract For SPeakerS<br />

ANDREA CALIfANO<br />

COLUMBIA UNIVERSITY, NEW YORK NY<br />

Dissecting and interrogating signaling networks in human malignacies<br />

We will discuss novel experimentally validated computational approaches<br />

to the dissection and interrogation of signal transduction networks in human<br />

malignancies. Specifically, we will address the issue of signaling pathway analysis<br />

from gene expression and phospho-proteomic profile data. We will first<br />

present the identification of KLHL9 deletions as key events in the etiology of<br />

the mesenchymal subtype of high-grade, associated with worst prognosis. We<br />

show that KLHL9, a substrate-specific adapter of a Cul3-based E3 ubiquitinprotein<br />

ligase complex, is responsible for the ubiquitination and proteasomal<br />

degradation of two previously identified master regulators of this glioma<br />

subtype, C/EBPb and C/EBPd. We will also discuss the identification of synergistic synthetic lethality in the<br />

tyrosine kinase signaling network that is dysregulated in non-small cell lung tumors, leading to a potential<br />

application of personalized combination therapy, using available kinase inhibitors.<br />

4 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 5


RUNE LINDING<br />

DTU, LYNGBY - DENMARK<br />

abStract For SPeakerS<br />

Dr Rune Linding is Professor and Research Group Leader for the Cellular Signal<br />

Integration Group (C-SIG) at the Technical University of <strong>Denmark</strong> (DTU),<br />

Center for Biological Sequence Analysis (CBS), Department of Systems Biology,<br />

<strong>Denmark</strong>. He performed his graduate work at the EMBL (Germany), where<br />

he pioneered computational analysis of cell signaling by developing popular<br />

tools like ELM, GlobPlot and DisEMBL for analysing post-translational modifications,<br />

intrinsic protein disorder and modularity of signaling proteins. Dr<br />

Linding was Human Frontiers Science Program Postdoctoral Research Fellow<br />

jointly with Profs Tony Pawson and Mike Yaffe at Samuel Lunenfeld Research<br />

Institute (SLRI, Canada) and Massachusetts Institute of Technology (MIT,<br />

USA), respectively. His postdoctoral work on the cellular phosphorylation networks and development of the<br />

NetworKIN algorithm pioneered Integrative Network Biology and led to the discovery of the quantitative<br />

importance of contextual kinase specificity. He started his own lab (the Cellular & Molecular Logic Team)<br />

at The Institute of Cancer Research (ICR) in London in 2007. At ICR his lab unravelled systems-level models<br />

of JNK and EphR kinase networks, demonstrated a link between specificity and oncogenecity of kinases<br />

and introduced the concept of Network Medicine. Dr Linding leads the NetPhorest community resource<br />

and have pioneered comparative phospho-proteomics and evolutionary studies of signalling networks. Dr<br />

Linding founded the Integrative Network Biology initiative (INBi) which aims to block cancer metastasis by<br />

integration of large-scale, high-dimensional quantitative genomic, proteomic and phenotypic data. His lab<br />

moved to DTU/<strong>Denmark</strong> in 20<strong>11</strong> and the long-term focus of his research group is on studying cellular signal<br />

processing and decision making.<br />

Modeling of Cancer Kinome Networks<br />

Abstract: Biological systems are composed of highly dynamic and interconnected molecular networks that<br />

drive biological decision processes. A goal of integrative network biology is to describe, quantify and predict<br />

the information flow and functional behaviour of living systems in a formal language and with an accuracy<br />

that parallels our characterisation of other physical systems such as Jumbo-jets. Decades of targeted<br />

molecular and biological studies have led to numerous pathway models of developmental and disease<br />

related processes. However, so far no global models have been derived from pathways, capable of predicting<br />

cellular trajectories in time, space or disease. The development of high-throughput methodologies has<br />

further enhanced our ability to obtain quantitative genomic, proteomic and phenotypic readouts for many<br />

genes/proteins simultaneously. Here, I will discuss how it is now possible to derive network models through<br />

computational integration of systematic, large-scale, high-dimensional quantitative data sets. I will review<br />

our latest advances in methods for exploring phosphorylation networks. In particular I will discuss how the<br />

combination of quantitative mass-spectrometry, systems-genetics and computational algorithms (NetworKIN<br />

[1] and NetPhorest [4]) made it possible for us to derive systems-level models of JNK and EphR signalling<br />

networks [2,3]. I shall discuss work we have done in comparative phospho-proteomics and network<br />

evolution[5-7]. Finally, I will discuss our most recent work in analysing genomic sequencing data from NGS<br />

studies and how we have developed new powerful algorithms to predict the impact of disease mutations on<br />

cellular signaling networks and applied them to profiling of ovarian cancer cells.<br />

References:<br />

http://<strong>www</strong>.lindinglab.<strong>org</strong><br />

Linding et al., Cell 2007.<br />

Bakal et al., Science 2008.<br />

Jørgensen et al., Science 2009.<br />

Miller et al., Science Signaling 2008.<br />

Tan et al., Science Signaling 2009.<br />

abStract For SPeakerS<br />

GIANNI CESARENI<br />

Mapping the human phosphatome on growth pathways<br />

fRANCESCA SACCO 1<br />

, PIER fEDERICO GhERARDINI 1<br />

, SERENA PAOLUzI 1<br />

, JULIO SAEz-RODRIGUEz 3<br />

,<br />

MANUELA hELMER-CITTERICh 1<br />

, ANTONELLA RAGNINI-WILSON 1,2<br />

, LUISA CASTAGNOLI 1<br />

AND GIANNI<br />

6 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 7<br />

CESARENI 1<br />

1<br />

DEPARTMENT Of BIOLOGY, UNIVERSITY Of ROME “TOR VERGATA”, ITALY 2hIGh-ThROUGhPUT MICROS-<br />

COPY fACILITY; DEPARTMENT Of TRANSLATIONAL AND CELLULARPhARMACOLOGY, CONSORzIO MARIO<br />

NEGRI SUD, SM. IMBARO, ITALY 3EMBL-EBI, hINxTON, UK AND EMBL-GENOME BIOLOGY UNIT, hEIDEL-<br />

BERG, GERMANY<br />

Large-scale siRNA screenings allow linking the function of poorly characterized genes to phenotypic<br />

readouts. According to this strategy, genes are associated to a function of interest if the alteration of their<br />

expression perturbs the phenotypic readouts. However, given the intricacy of the cell regulatory network, the<br />

mapping procedure is low resolution and the resulting models provide little mechanistic insights. We have<br />

developed a new strategy that combines high-content multiparametric analysis of cell perturbation and logic<br />

modeling to achieve a more detailed functional mapping of human genes onto complex pathways. By this


JULIA hOENG<br />

Verification of Systems Biology Research in the Age of Collaborative-Competition<br />

abStract For SPeakerS<br />

JULIA hOENG1, MARJA TALIKKA1, STéPhANIE BOUé1, PABLO MEYER ROJAS2, RAqUEL NOREL2, JOhN J<br />

RICE2, JöRG SPRENGEL3, MANUEL PEITSCh1, GUSTAVO STOLOVITzKY2<br />

1PhILIP MORRIS INTERNATIONAL R&D, NEUChâTEL, SWITzERLAND, 2IBM COMPUTATIONAL BIOLOGY<br />

CENTER, YORKTOWN hEIGhTS, NY, USA, 3IBM GLOBAL BUSINESS SERVICES, SWITzERLAND<br />

Abstract:<br />

Modern society demands greater scrutiny of the potential health risks and benefits of long-term, and sometimes<br />

lifelong, exposure to drugs, chemicals, and substances found in consumer products and the environment.<br />

Organizations such as companies and academic consortia conduct large multi-year scientific studies that<br />

entail the collection and analysis of thousands of data points. The individual experiments are often conducted<br />

over many physical sites and with internal and outsourced components. To extract maximum value,<br />

the interested parties need to verify the accuracy and reproducibility of automated collection and analysis<br />

workflows in systems biology before the initiation of large multi-year studies.<br />

Traditional verification using the peer-review process has shortcomings, such as lack of scalability, which<br />

renders it insufficient for the assessment of high throughout research. A team of researchers at PMI and<br />

IBM, whose aim is to improve the effectiveness of scientific studies and verification of scientific findings<br />

propose a scheme called IMPROVER, for Industrial Methodology for Process Verification of Research. This<br />

methodology evaluates a research program by dividing its workflow into smaller building blocks, whereby the<br />

verification of each building block can be done internally or externally via challenge-based `crowdsourcing`<br />

to a research community.<br />

Scientific challenges will be broadcast to potential stakeholders in the form of an open call for participation<br />

with the intention of providing the community with the opportunity to test their computational methods on<br />

new data as well as to partake in a collaborative effort whose ultimate goal could contribute to solving a<br />

grand scientific problem.<br />

Considering cancer as the leading cause of death worldwide, we formulate the Diagnostics Signature Challenge<br />

to evaluate novel approaches for the identification of robust and predictive signatures for this disease.<br />

The goal of a Diagnostics Signature Challenge is to verify that transcriptomics data contains enough<br />

information for the determination and prognosis of certain human disease states that could profit from better<br />

diagnostics signatures.<br />

Here we will describe the approach, the necessary operational steps, and how we intend to engage the<br />

wider scientific community to assess the applicability of the IMPROVER approach to molecular diagnostics<br />

(i.e., genomic signatures).<br />

abStract For SPeakerS<br />

ROTEM BEN-hAMO, SOL EfRONI<br />

ThE MINA & EVERARD GOODMAN fACULTY Of LIfE SCIENCES, BAR ILAN UNIVERSITY, RAMAT GAN,<br />

ISRAEL<br />

Network-based metrics reveals a novel role for hsa-miR-9 and drug control over the p38 network in glioblastoma<br />

multiforme progression<br />

Contributors:<br />

Background<br />

Glioblastoma multiforme (GBM) is the most common, aggressive and malignant primary tumor of the brain<br />

and is associated with one of the worst 5-year survival rates among all human cancers. Identification of<br />

molecular interactions that associate with disease progression may be key in finding novel treatments.<br />

Using five independent molecular and clinical datasets with a set of computational algorithms we were able<br />

to identify a gene-gene and gene-microRNA network that significantly stratifies patient prognosis. By combining<br />

gene expression microarray data with microRNA expression levels, copy number alterations, drug<br />

response and clinical data, combined with network knowledge, we were able to identify a single pathway at<br />

the core of glioblastoma.<br />

This network, the p38 network, and an associated microRNA, hsa-miR-9, facilitate prognostic stratification.<br />

The microRNA hsa-miR-9 correlated with network behavior and presents binding affinities with network<br />

members in a manner that suggests control over network behavior. A similar control over network behavior<br />

is possible through a set of drugs. These drugs are part of the treatment regimen for a subpopulation of the<br />

patients that participated in the TCGA study and for which the study provides clinical information. Interestingly,<br />

the patients that were treated with these specific sets of drugs, all of which targeted against p38<br />

network members, demonstrate highly significant stratification of prognosis.<br />

Combined, these results call for attention to p38 network targeted treatment and present the p38 networkhsa-miR-9<br />

control mechanism as critical in GBM progression.<br />

8 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 9


GARRY NOLAN<br />

STANfORD, STANfORD, CA<br />

TBA<br />

abStract For SPeakerS<br />

abStract For SPeakerS<br />

NORBERT PERRIMON<br />

hARVARD MEDICAL SChOOL, BOSTON, MA<br />

Dr. Perrimon is the James Stillman Professor of Developmental Biology at<br />

Harvard medical School and an Investigator of the Howard Hughes Medical<br />

Institute. He has 30 years of experience in the fields of developmental<br />

genetics, signal transduction and genomics. He has made a number of contributions<br />

to the fields of Genetics, Developmental Biology, signal transduction<br />

and functional genomics. His group developed many methods that have<br />

significantly improved the Drosophila toolbox. These include: the FLP-FRT<br />

Dominant Female Sterile technique to generate mosaics in the female germline,<br />

the Gal4-UAS method to control gene expression both spatially and<br />

temporally; the “Positively Marked Labeling Method” for lineage analyses;<br />

and thermosensitive inteins to generate conditional alleles. His contributions<br />

include the characterization of: the maternal effects of zygotic lethal mutations; the logic of head patterning;<br />

the identification of Scribble and the <strong>org</strong>anization of the cell polarity complexes; the discovery of adult<br />

gut stem cells; and the mechanisms of muscle growth and aging. His lab has characterized many signaling<br />

components of receptor tyrosine kinases, Wnt and JAK/STAT pathways, in particular. These include: Raf<br />

kinase and demonstration that it acts downstream of Ras; Corkscrew/SHP2 non receptor tyrosine phosphatase<br />

as a positive transducer of RTK signaling; Spitz as a ligand, and Kekkon as a negative regulator, of<br />

EGFR; Porcupine, Dishevelled and GSK3 as components of Wnt/Wg signaling; Unpaired, Hopscotch/JAK and<br />

Marelle/STAT as members of the JAK/STAT pathway; and Heparan Sulfate Proteglycans in Hedgehog, Wnt<br />

and FGF signaling. Regarding large scale functional genomics, his group established high-throughout genome-wide<br />

RNAi screens to systematically interrogate the entire Drosophila genome in various cell-based<br />

assays, demonstrated that long dsRNAs are associated with off target effects, established a cross-species<br />

method for rescue of RNAi phenotypes, developed RNAi methods in primary embryonic cell cultures, and<br />

generated algorithms for automated image analyses. In 2003 he created the Drosophila RNAi Screening<br />

Center (DRSC) at Harvard Medical School to make this technology available to the community. In addition,<br />

his group developed new shRNA vectors for in vivo RNAi and in 2008 established the Transgenic RNAi Project<br />

(TRiP) at Harvard Medical School to build a genome scale resource of transgenic shRNA flies. Currently,<br />

his laboratory is applying large-scale RNAi and proteomic methods to obtain a global understanding of the<br />

structure of a number of signaling pathways and their cross-talks. In addition, he is studying the roles of<br />

signaling pathways in homeostasis and tissue remodeling in Drosophila muscles and gut stem cells. Dr.<br />

Perrimon has trained more than 80 students and postdoctoral fellows, with most of them currently holding<br />

academic positions.<br />

Building and validating signaling networks in Drosophila<br />

Characterizing the extent and logic of signaling networks is essential to understanding developmental<br />

processes, mechanisms of oncogenesis, and resistance to chemotherapy. A major focus of our lab is to<br />

apply “Omics” approaches to describe the <strong>org</strong>anization of signaling networks. We have spent considerable<br />

effort to implement complementary technologies of genome-wide RNAi HTS, tandem affinity purification/<br />

mass spectrometry (TAP/MS), and transcriptome analyses in Drosophila cell lines, to identify core pathway<br />

components, pathway dynamics, and the extent of cross-talk between pathways. Recently, because studying<br />

networks in the fly is hampered by the paucity of phosphoantibodies available, we have used the Tandem<br />

Mass Tags Mass Spec method to measure quantitative changes in the phosphoproteome. Integration of<br />

various Omic approaches is necessary for network studies because much of the noise associated with each<br />

technology can be filtered by the integration of orthogonal data sets.<br />

We have applied these approaches successfully to date to the study of five pathways in established Drosophila<br />

cell lines: Insulin/PI3K, EGF/MAPK, JAK/STAT, JNK and Hippo and have generated high-confidence<br />

protein-protein interaction (PPI) networks that have been validated to various extents by RNAi and transcriptome<br />

analyses. Our networks are of high quality because they identify most previously known interactions,<br />

and by various means we have validated a number of new components and interactions.<br />

10 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / <strong>11</strong>


DANA PE’ER<br />

COLUMBIA UNIVERSITY, NEW YORK, NY<br />

Dana Pe’er is assistant professor at the Department of Biological Sciences at<br />

Columbia and Columbia’s Center for Computational Biology and Bioinformatics<br />

(C2B2). In her Ph.D. (Computer Science) at the Hebrew University of Jerusalem<br />

(with Nir Friedman), Dana pioneered the use of machine learning to uncover<br />

the structure and function of molecular networks from genomics data,<br />

based on Bayesian networks. She subsequently did a postdoc with Ge<strong>org</strong>e<br />

Church at Harvard Medical School and there she began to work towards understanding<br />

of how genetic variation alters the regulatory network between<br />

individuals and subsequently manifests in phenotypic diversity. This is now<br />

the focus of Dana’s lab at Columbia University, where she and her team are<br />

developing methods to infer how variation in sequence modulates signal processing and is manifested in<br />

cellular phenotypes, with applications towards personalized cancer treatment. Dana is recipient of the Burroughs<br />

Wellcome Fund Career Award, NIH Directors New Innovator Award, Stand Up To Cancer Innovative<br />

Research Grant and a Packard Fellow in Science and Engineering.<br />

On the road to personalized therapy, a systems approach<br />

Cancer is an individual disease—unique in how it develops and behaves in every patient. The emergence<br />

of revolutionary technologies has stimulated hope that treatment will improve by becoming more targeted<br />

and individualized in nature. Characterization of cancer genomes has revealed a staggering complexity of<br />

aberrations among individuals, such that the functional importance and physiological impact of most tumor<br />

genetic alterations remains poorly defined. Genomic and proteomic data in tumor samples, using a battery<br />

of using high-throughput, massively parallel technologies is accumulating at a astounding rates. A major<br />

challenge involves the development of analysis methods to integrate this data towards patient-specific tumor<br />

network models. We demonstrate progress on a number of fronts.<br />

• We demonstrate approaches to integrate heterogeneous genomic data types to identify the key<br />

alterations functionally driving the cancer and associate these with their tumorigenic phenotypes (e.g.<br />

proliferation, invasion).<br />

• To understand tumor response to drug and the heterogeneity of this response among patients it is<br />

critical to molecularly interrogate the tumor following drug perturbations. We will demonstrate how such<br />

interrogation of a tumor panel reveals variation in network wiring that connects to drug response.<br />

• Tumors are not only heterogeneous between patients, but there is also pervasive heterogeneity within<br />

a single patient. Mass-cytometry, a novel technology that can measure more than forty signaling mole-<br />

abStract For SPeakerS abStract For SPeakerS<br />

MARC VIDAL<br />

CENTER fOR CANCER SYSTEMS BIOLOGY (CCSB) AND DEPARTMENT Of<br />

CANCER BIOLOGY DANA-fARBER CANCER INSTITUTE & DEPARTMENT Of<br />

GENETICS hARVARD MEDICAL SChOOL BOSTON, MA 02<strong>11</strong>5<br />

Marc Vidal is Professor of Genetics at Harvard Medical School and Director<br />

of the Center for Cancer Systems Biology (CCSB) at the Dana-Farber Cancer<br />

Institute. Dr. Vidal received his PhD in 1991 from Gembloux University (Belgium)<br />

for work performed at Northwestern University (Evanston, IL, USA)<br />

where he identified the yeast genes RPD3 and SIN3, and demonstrated that<br />

they encode global transcriptional regulators. These genes were subsequently<br />

found to encode Histone Deacetylase (HDAC) and its main recruiting<br />

factor, respectively. During postdoctoral training at the Massachusetts General Hospital Cancer Center, he<br />

developed the reverse two-hybrid system, a widely applicable method used to genetically characterize protein-protein<br />

interactions. Having developed interdisciplinary strategies together with collaborators from<br />

the fields of physics, computer science, mathematics, genomics and human genetics, he and his team have<br />

been charting protein-protein and other interactome networks for 15 years and are developing ways to integrate<br />

interactome maps with other large-scale functional genomic and proteomic maps, with the ultimate<br />

objective to discover novel network properties from a systems point-of-view. Dr. Vidal was elected Associate<br />

Member of the Royal Academy for Science and the Arts of Belgium and has received several awards,<br />

including a Chair from the Francqui Foundation (Belgium) and an Abbott Bioresearch Award.<br />

Interactome Networks and Human Disease<br />

For over half a century it has been conjectured that macromolecules form complex networks of functionally<br />

interacting components, and that the molecular mechanisms underlying most biological processes correspond<br />

to particular steady states adopted by such cellular networks. However, until a decade ago, systemslevel<br />

theoretical conjectures remained largely unappreciated, mainly because of lack of supporting experimental<br />

data.<br />

To generate the information necessary to eventually address how complex cellular networks relate to biology,<br />

we initiated, at the scale of the whole proteome, an integrated approach for modeling protein-protein<br />

interaction or “interactome” networks. Our main questions are: How are interactome networks <strong>org</strong>anized at<br />

the scale of the whole cell? How can we uncover local and global features underlying this <strong>org</strong>anization, and<br />

how are interactome networks modified in human disease, such as cancer?<br />

12 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / <strong>13</strong>


STEPhAN M. fELLER, WIMM,<br />

OxfORD UNIVERSITY, OxfORD, UK<br />

abStract For SPeakerS<br />

How are complex signal computations in cells accomplished by multi-protein complexes assembled on<br />

‘intrinsically disordered’ platform proteins? The N terminal folding nucleation (NFN) hypothesis<br />

Proteins with little recognisable secondary structure -according to current prediction programs -comprise<br />

one third of the human proteome. Some of these are large and serve as docking platforms for many proteins<br />

involved in the processing of cell signals, thereby creating large signalosomes in which signal computations<br />

are performed. It was always difficult to conceptualise how an intrinsically disordered, i.e. ‘chaotic’ signal<br />

computation platform protein should be able to mediate effective pathway crosstalk in cells. However, we<br />

have now first evidence that the cancer-relevant Gab family proteins display a previously unrecognised<br />

<strong>org</strong>anisation (Simister et al. 20<strong>11</strong>, PLoS Biol; Simister & Feller <strong>2012</strong>, Mol BioSystems). Gab proteins contain<br />

N terminal PH domains followed by long ‘tail regions’ supposedly lacking any structure. Our data point to<br />

several specific interactions of the Gab1 tail with is structured N-terminus. These generate, around the PH<br />

domain, tail loop structures, in which docking sites for pathway-specific signaling proteins are clustered.<br />

Functionally dedicated, distinct sub-complexes can assembly in these loops. If this model is correct, it is<br />

easy to see how effective signaling pathway crosstalk occurs: simply by interactions of distinct sub-complexes<br />

bound to specialised loops, which are held in place by their interactions with the PH domain. We believe<br />

that proving this hypothesis, which is also relevant to multiple similarly structured signaling protein families<br />

(p<strong>13</strong>0Cas, IRS/Dok, Frs etc.), will have a profound impact on the understanding of molecular signal computing<br />

in cells and we will report on our first results.<br />

abStract For SPeakerS<br />

ThEO KNIJNENBURG<br />

NEThERLANDS CANCER INSTITUTE, DIVISION Of MOLECULAR CARCINOGENESIS<br />

PLESMANLAAN 121, 1066Cx AMSTERDAM, ThE NEThERLANDS<br />

MAThEW J. GARNETT<br />

CANCER GENOME PROJECT , WELLCOME TRUST SANGER INSTITUTE<br />

hINxTON CAMBRIDGE CB10 1SA, UNITED KINGDOM<br />

GUNNAR W. KLAU<br />

LIfE SCIENCES GROUP, CENTRUM WISKUNDE & INfORMATICA<br />

SCIENCE PARK 123, 1098 xG AMSTERDAM, ThE NEThERLANDS<br />

fRANCESCO IORIO<br />

EUROPEAN BIOINfORMATICS INSTITUTE, WELLCOME TRUST GENOME CAMPUS<br />

hINxTON CAMBRIDGE CB10 1SD, UNITED KINGDOM<br />

JULIO SAEz-RODRIGUEz<br />

EUROPEAN BIOINfORMATICS INSTITUTE, WELLCOME TRUST GENOME CAMPUS<br />

hINxTON CAMBRIDGE CB10 1SD, UNITED KINGDOM<br />

ULTAN MCDERMOTT<br />

CANCER GENOME PROJECT , WELLCOME TRUST SANGER INSTITUTE<br />

hINxTON CAMBRIDGE CB10 1SA, UNITED KINGDOM<br />

LODEWYK WESSELS<br />

NEThERLANDS CANCER INSTITUTE, DIVISION Of MOLECULAR CARCINOGENESIS<br />

PLESMANLAAN 121, 1066Cx AMSTERDAM, ThE NEThERLANDS<br />

Drug sensitivity of cancer cell lines explained as a logic combination of mutations<br />

Cancer arises as a result of the acquisition of DNA mutations. It is still unclear which and how combinations<br />

of mutations are involved in tumor initiation and development. Two major challenges need to be addressed in<br />

order to systematically unravel these genetic interactions.<br />

First, large amounts of high quality data are necessary in order to ensure the statistical power required<br />

to uncover these genotype-to-phenotype relationships. Current advances in high-throughput biology are<br />

enabling the generation of very large datasets that should facilitate the detection of higher order genetic<br />

interactions. Here, we report on the analysis of a panel of 1000 cancer cell lines. The mutation status of 66<br />

known cancer genes has been characterized for each cell line in this panel. Additionally, these cell lines<br />

have been screened to model drug response with a large number (250+) of anti-cancer therapeutics. This<br />

dataset is part of the Cancer Genome Project at the Wellcome Trust Sanger Institute.<br />

The second challenge is to design a computational framework that employs a mathematical formalism rich<br />

enough to capture the underlying biological complexity, while limiting the computational complexity that<br />

would otherwise prohibit finding optimal (or even good) solutions in the vast combinatorial search space.<br />

We contribute to addressing this second challenge by proposing a novel computational approach based on<br />

integer programming that infers logic combinations of mutations that predict the observed drug response.<br />

The use of a logic formalism enables the formulation of intelligible models, from which the cancer biologists<br />

can generate a deeper understanding based on domain knowledge and easily formulate novel hypotheses<br />

and experiments.<br />

Our models show that for most drugs, combinations of mutations explain the drug response better than<br />

single mutations. For example, of the 8 BRAF inhibitors in the panel, the drug sensitivity to 3 of them is better<br />

explained using a logic combination of a BRAF mutation and one or more mutations in other genes, such<br />

as TET2. These results immediately suggest putative drug combination therapies.<br />

Additionally, cancer signaling pathways as annotated in e.g. the Pathway Interaction Database can be<br />

employed to dramatically reduce the search space and offer a mechanistic explanation of the uncovered<br />

gene combinations.<br />

14 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 15


abStract For SPeakerS<br />

SøREN BRUNAK,<br />

TEChNICAL UNIVERSITY Of DENMARK & UNIVERSITY Of COPENhAGEN<br />

Søren Brunak, Ph.D., is professor of Bioinformatics at the Technical University<br />

of <strong>Denmark</strong> and professor of Disease Systems Biology at the University of Copenhagen.<br />

Prof. Brunak is the founding Director of the enter for Biological Sequence<br />

Analysis, which was formed in 1993 as a multi-disciplinary research<br />

group of molecular biologists, biochemists, medical doctors, physicists, and<br />

computer scientists. Søren Brunak has been highly active within data integration,<br />

where machine learning techniques often have been used to integrate<br />

predicted or experimentally established functional genome and proteome annotation.<br />

His current research does combine molecular level systems biology<br />

and healthcare sector data such as electronic patient records and biobank<br />

questionnaires. The aim is to group and stratify patients not only from their genotype, but also phenotypically<br />

based on the clinical descriptions in the medical records.<br />

Interfacing disease phenotypes from electronic patient records to the underlying network biology<br />

Interfacing sequencing and network biology data to personal healthcare sector information World-wide the<br />

healthcare sector is confronted with the availability of database information which describe the individual<br />

in great detail. These data range all the way from the molecular level, where they for example reveal the<br />

genetic makup of the patient, to the fine-grained descriptions of disease phenotypes as they are found in<br />

electronic patient records at hospitals. Linking these data is a huge undertaking which soon will represent a<br />

major challenge given that it already has become feasible to sequence the DNA of entire populations at low<br />

cost. Combining molecular level data with clinical information and data on the chemical environment may<br />

add complementary types of knowledge which - together with genotype and metagenomic information from<br />

the individual - can reveal disease mechanisms in novel ways. Electronic patient records remain a rather<br />

unexplored, but potentially rich data source for example for discovering correlations between diseases. We<br />

describe a general approach for gathering phenotypic descriptions of patients from medical records in a systematic<br />

and non-cohort dependent manner. By extracting phenotype information from the free-text in such<br />

records we demonstrate that we can extend the information contained in the structured record data, and use<br />

it for producing fine-grained patient stratification and disease co-occurrence statistics. The approach uses<br />

a dictionary based on the WHO International classification of Disease ontology and is therefore in principle<br />

language independent.<br />

References<br />

Using electronic patient records to discover disease correlations and stratify patient cohorts. Roque FS,<br />

Jensen PB, Schmock H, Dalgaard M, Andreatta M, Hansen T, Søeby K, Bredkjær S, Juul A, Werge T, Jensen<br />

LJ, Brunak S. PLoS Comput Biol. 20<strong>11</strong> Aug;7(8):e1002141.<br />

Knowledge engineering for health: A new discipline required to bridge the “ICT gap” between research and<br />

healthcare. Beck T, Gollapudi S, Brunak S, Graf N, Lemke HU, Dash D, Buchan I, Díaz C, Sanz F, Brookes<br />

AJ. Hum Mutat. <strong>2012</strong> Mar 5. doi: 10.1002/humu.22066<br />

Mining electronic health records: towards better research applications and clinical care Jensen PB, Jensen<br />

LJ, and Brunak S, Nature Reviews Genetics, June <strong>2012</strong>, to appear.<br />

abStract For SPeakerS<br />

ChRISTIAN KELSTRUP<br />

NNf-CPR, COPENhAGEN, DENMARK<br />

Optimized Fast and Sensitive Acquisition Methods for Shotgun Proteomics on a Quadrupole Orbitrap Mass<br />

Spectrometer<br />

16 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 17


SPonSorPage<br />

abStract For SPeakerS<br />

MIChAEL YAffE<br />

MIT, CAMBRIDGE, MA<br />

18 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 19


abStract For SPeakerS<br />

MATThEW ONSUM<br />

Ph.D., MERRIMACK PhARMACEUTICALS, CAMBRIDGE, MA<br />

Dr. Onsum is an Associate Director of Translational Research at Merrimack<br />

Pharmaceuticals. He received his B.S., M.S., and Ph.D. degrees in Mechanical<br />

Engineering from the University of California, Berkeley. His doctoral work,<br />

under the supervision of Adam Arkin and Kameshwar Poolla, used both computational<br />

and wet biology to study how immune cells track and capture invading<br />

microbes. Additionally, he was a member of the Alliance for Cellular<br />

Signaling where he developed mathematical models of GPCR mediated calcium<br />

signaling and model validation software. He spent two years at Astra-<br />

Zeneca R&D Boston, where he used model simulations to help identify new<br />

drug targets. He is currently at Merrimack Pharmaceuticals where he is leading<br />

the translational research program for MM-<strong>11</strong>1, a bi-specific antibody against ErbB3 that uses an ErbB2<br />

targeting arm to enhance avidity and inhibitor potency.<br />

Using systems biology to accelerate oncology drug development<br />

This session will discuss how Merrimack uses mathematical models of cancer signaling pathways to design<br />

novel therapeutics, identify predictive biomarkers, and guide clinical development plans. By combining the<br />

knowledge gained from our biochemical model together with biomarker measurements from a large panel<br />

of archived tumors and clinical data from the literature, we simulated the effect of our lead oncology drug in<br />

a variety of cancer indications and used these simulations to help prioritize our clinical development plans.<br />

We will also discuss how we use our mathematical models to assess other targeted oncology drugs and<br />

determine which of these drugs should be combined with our therapies.<br />

abStract For SPeakerS<br />

RUTh hüTTENhAIN 1<br />

, MARTIN SOSTE 2<br />

, NAThALIE SELEVSEK 1<br />

, hANNES RöST 1<br />

, ATUL SEThI 1<br />

, ChRISTINE<br />

CARAPITO 3<br />

, TERRY fARRAh 4<br />

, ERIC W. DEUTSCh 4<br />

, ULRIKE KUSEBAUCh 4<br />

, ROBERT L. MORITz 4<br />

, EMMA<br />

NIMèUS-MALMSTRöM 5<br />

, OLIVER RINNER 6<br />

AND RUEDI AEBERSOLD 1,7<br />

20 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 21<br />

1<br />

DEPARTMENT Of BIOLOGY, INSTITUTE Of MOLECULAR SYSTEMS BIOLOGY, ETh zURICh, zURICh, SWITzERLAND<br />

2<br />

DEPARTMENT Of BIOLOGY, INSTITUTE Of BIOChEMISTRY, ETh zURICh, zURICh, SWITzER-<br />

LAND 3<br />

LABORATOIRE DE SPECTROMéTRIE DE MASSE BIO-ORGANIqUE, INSTITUT PLURIDISCIPLINAIRE<br />

hUBERT CURIEN, UMR7178 CNRS-UNIVERSITé DE STRASBOURG, STRASBOURG, fRANCE 4<br />

INSTITUTE<br />

fOR SYSTEMS BIOLOGY, SEATTLE, WA, USA 5<br />

DEPARTMENT Of ONCOLOGY, LUND UNIVERSITY, LUND, SWE-<br />

DEN 6<br />

BIOGNOSYS AG, zURICh, SWITzERLAND 7<br />

fACULTY Of SCIENCE, UNIVERSITY Of zURICh, zURICh,<br />

SWITzERLAND<br />

A mass spectrometric map for reproducible quantification of cancer associated proteins in body fluids<br />

A major bottleneck in applying targeted proteomic approaches to protein biomarker research is the limited<br />

availability of accurate, reproducible and sensitive assays for testing hypotheses on cohorts of patient<br />

samples. Therefore, we aimed to generate a high-quality resource of selected reaction monitoring (SRM)<br />

assays for cancer associated proteins (CAPs), from an evidence-based list of <strong>11</strong>72 proteins, that have been<br />

previously documented to be differentially expressed in various types of cancer1. Using a protein functional<br />

network, we demonstrated that these proteins are enriched among the interaction partners of genes mutated<br />

in cancer.<br />

Following the development of the SRM assays we examined their detectability in two types of samples which<br />

are highly relevant for biomarker studies, plasma and urine. The concentrations of the detected CAPs in<br />

plasma span six orders of magnitude demonstrating the high sensitivity of these assays for protein quantification.<br />

The developed SRM assays and detectability information are publicly available via PeptideAtlas<br />

SRM Experiment Library (PASSEL)2 to enable researchers to test hypotheses related to these CAPs in any<br />

sample of interest.<br />

We demonstrated the utility of this resource for biomarker research by measuring CAPs from the FDA-approved<br />

OVA1 biomarker panel across plasma samples collected from women diagnosed with a pelvic mass.<br />

The measurements were able to recapitulate the expected capability of this panel to stratify ovarian cancer<br />

(OC) patients and patients with benign ovarian tumors (BOT). Moreover, using this resource, we explored<br />

the prospect of discovering novel biomarker candidates by in silico prediction. Using a functional protein network,<br />

we derived a set of 21 CAPs, that interact with genes mutated in OC, and are measurable in plasma.<br />

12 of these network derived proteins showed a significant difference in abundance between patients with OC<br />

and patients with BOT.<br />

In sum, by developing a publicly accessible resource of SRM assays and testing their detectability in body<br />

fluids, this study will facilitate applying targeted proteomics to protein biomarker research. Furthermore, we<br />

explored the promise of combining genomic data and protein network analysis for predicting novel biomarker<br />

candidates. We demonstrated that this resource can be used to rapidly test these hypotheses in body fluids.<br />

Polanski, M. & Anderson, N.L. A list of candidate cancer biomarkers for targeted proteomics. Biomarker<br />

Insights 1, 1-48 (2007).<br />

Farrah, T. et al. PASSEL: The PeptideAtlas SRM Experiment Library. Proteomics, 10.1002/pmic.20<strong>11</strong>00515<br />

(<strong>2012</strong>).


DR JANINE T ERLER,<br />

BRIC, UNIVERSITY Of COPENhAGEN<br />

COPENhAGEN, DENMARK<br />

abStract For SPeakerS<br />

Elucidating the molecular networks associated with metastasis<br />

Metastasis is responsible for over 90% of cancer patient deaths due to a<br />

lack of effective therapies against metastatic disease. We require a better<br />

understanding of the underlying molecular processes in order to identify and<br />

develop novel effective therapeutic strategies. We aim to identify the signaling<br />

networks associated with metastasis through several different approaches.<br />

Our goal is to identify key nodes in the network and target these to prevent<br />

disease progression, and translate these findings into the clinic.<br />

One approach we are taking is to identify and compare the signaling networks<br />

and associated genetic mutations in metastatic versus non-metastatic samples derived from patients. In<br />

our model, we have used non-metastatic and metastatic matched human cancer cell lines isolated from the<br />

same patient at different stages. We have performed quantitative mass spectrometry to compare the global<br />

proteome and phosphoproteome, and next generation sequencing to both identify and compare mutations,<br />

and investigate their impact on the signaling networks. We are deploying the NetPhorest and NetworKIN<br />

algorithms to predict phospho-binding modules likely to interact with the identified sites. Additionally, computational<br />

integration of the molecular data with quantitative phenotypic data acquired from high throughput<br />

invasion assays allows us to build predictive models of cancer progression. We are using RNAi to perturb<br />

these networks and refine our models to identify the core network predicted to drive metastasis. We will additionally<br />

integrate data collected from fresh frozen human patient samples. Finally, we will test our predictions<br />

using an in vivo model of metastasis.<br />

The results of this study will aid the development of a network-based therapeutic strategy for the treatment<br />

and prevention of metastatic disease.<br />

abStract For SPeakerS<br />

NEVAN KROGAN<br />

CELLULAR AND MOLECULAR PhARMACOLOGY/CALIfORNIA INSTITUTE fOR<br />

qUANTITATIVE BIOMEDICAL SCIENCES, UNIVERSITY Of CALIfORNIA, SAN<br />

fRANCISCO, CA, 94158<br />

Dr. Krogan is an Associate Professor in the Department of Cellular and Molecular<br />

Pharmacology at the University of California-San Francisco and is<br />

an expert in the fields of functional genomics and systems biology. He was<br />

born and raised in Regina, Saskatchewan, Canada and obtained his undergraduate<br />

degree from the University of Regina. As a graduate student at the<br />

University of Toronto, Dr. Krogan led a project that systematically identified<br />

protein complexes in the model <strong>org</strong>anism, Saccharomyces cerevisiae, through<br />

an affinity tagging-purification/mass spectrometry strategy. This work led to<br />

the characterization of 547 complexes, comprising over 4000 proteins, and represents the most comprehensive<br />

protein-protein interaction map to date in any <strong>org</strong>anism. To complement this physical interaction<br />

data, Dr. Krogan developed an approach, termed E-MAP (or epistatic miniarray profile), which allows for<br />

high-throughput generation and quantitative analysis of genetic interaction data. Dr. Krogan’s lab at UCSF<br />

focuses on applying these global proteomic and genomic approaches to formulate hypotheses about various<br />

biological processes, including transcriptional regulation, DNA repair/ replication and RNA processing.<br />

He is now developing and applying methodologies to create genetic and physical interactions between<br />

pathogenic <strong>org</strong>anisms, including HIV and TB, and their hosts, which is providing insight into the human<br />

pathways and complexes that are being hijacked during the course of infection.<br />

Functional Insights from Protein-Protein and Genetic Interaction Maps<br />

Pathways and complexes can be considered fundamental units of cell biology, but their relationship to each<br />

other is difficult to define. Comprehensive tagging and purification experiments have generated networks<br />

of interactions that represent most stable protein complexes. We describe this work in various <strong>org</strong>anisms,<br />

including budding yeast and in infectious <strong>org</strong>anisms like HIV and TB, and show how the analysis of pairwise<br />

epistatic relationships between genes complements the physical interaction data, and furthermore can be<br />

used to classify gene products into parallel and linear pathways.<br />

22 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 23


abStract For SPeakerS<br />

RUEDI AEBERSOLD<br />

ETh zURICh AND UNIVERSITY Of zURICh, SWITzERLAND<br />

Dr. Ruedi Aebersold is a Professor in the field of proteomics and systems biology<br />

with joint appointments at the ETH (Swiss Federal Institute of Technology)<br />

Zurich, Switzerland and the University of Zurich. He has served on<br />

the faculties of the Universities of Washington and British Columbia. He cofounded<br />

the Seattle Institute for Systems Biology, and participates as a member<br />

of Scientific Advisory Boards for a number of academic and private sector<br />

research <strong>org</strong>anizations. Dr. Aebersold, one of the pioneers in the field of<br />

proteomics and systems biology, is known for developing a series of methods<br />

and technologies for quantitative proteomics that can be applied to enhance<br />

our understanding of the structure, function, and control of complex biological<br />

systems. His group was instrumental in the landmark development of methods and reagents for stable<br />

isotopic labeling of protein samples enabling a quantitative dimension to biological mass spectrometry and<br />

the development of software tools for the statistically supported analysis of proteomics data. Recently, the<br />

group has pioneered the use of targeted mass spectrometry for the generation of consistent quantitative<br />

proteomic datasets on differentially perturbed systems. Dr. Aebersold has published more than 500 peer<br />

reviewed papers that have generated > 50.000 citations. He has reached an h-factor of 107 and is the recipient<br />

of numerous awards for his contribution to the field of protein sciences and proteomics including the<br />

MCP-HUPO lectureship (20<strong>11</strong>), the ASBMB Herbert Sober award (2009) the Otto Naegeli Prize (2009), the<br />

ABRF Award (2008), the FEBS Buchner Medal (2006), the HUPO Award (2005), the ASMS Biemann medal<br />

(2002) the Widmer award (2002), and the 2003 World Technology award. His group is currently focused on<br />

establishing novel label-free methods, leveraging new instrumentation and knowledge of representative<br />

“proteotypic” peptides, to rapidly and quantitatively profile global proteomes for discovery of new diagnostic<br />

markers for disease, and to facilitate a more complete understanding of the biochemical processes that<br />

control and constitute cell physiology.<br />

Network Driven Protein Biomarker Discovery and Validation<br />

A key barrier to the realization of personalized medicine for cancer is the identification of biomarkers. Of all<br />

types of biomarkers, plasma protein biomarkers are particularly attractive because they can be measured<br />

in easily accessible samples. Unfortunately, the search for plasma protein biomarkers has been highly<br />

challenging and met with surprisingly low level of success. Specifically, the comparison of plasma sample<br />

proteomes of control and disease affected individuals has to date not uncovered any new markers.<br />

On the backdrop of the emerging personal genome information and large scale cancer genome projects we<br />

have developed and applied a biomarker strategy that is driven by cancer genetic and genomic information.<br />

In a fist stage we use comparative genomic data to computationally predict which signaling systems might<br />

be perturbed in a particular type of cancer. We use targeted proteomic measurements on human tissue<br />

samples or tissue samples from suitable mouse models to experimentally validate these predictions, i.e. to<br />

determine which proteins are disregulated in the specific disease. We then use then the such validated perturbed<br />

molecular networks to select proteins that are likely to be secreted or otherwise released into plasma<br />

and quantify these proteins in sets of plasma samples by selected reaction monitoring, a highly sensitive<br />

targeted mass spectrometry technique.<br />

In this presentation we will discuss this novel biomarker strategy, its present status and expected directions.<br />

A case study on PTEN dependent prostate cancer will illustrate the concept.<br />

abStract For SPeakerS<br />

ANNE-CLAUDE GAVIN<br />

EMBL, hEIDELBERG, GERMANY<br />

Expanding the interaction space; protein-metabolite networks<br />

Biological function emerges from the concerted action of numerous interacting<br />

biomolecules. Deciphering the molecular mechanisms behind cellular<br />

processes requires the systematic charting of the multitude of interactions<br />

between all cellular components. Since the sequencing of the first eukaryotic<br />

genome, Saccharomyces cerevisiae, more than 10 years ago, explosion of<br />

new analytical tools in the fields of transcriptomics, proteomics and metabolomics<br />

contributes ever-growing molecular repertoires of the building blocks<br />

that make up a cell. Biology does not rely on biomolecules acting in isolation.<br />

Biological function depends on the concerted action of molecules acting<br />

in protein complexes, pathways or networks. Biomolecular interactions are<br />

central to all biological functions. In human, for example, impaired or deregulated protein–protein or protein–<br />

metabolite interaction often leads to disease. Recent strategies have been designed that allow the study of<br />

interactions more globally at the level of entire biological systems. We will discuss the use of these biochemical<br />

approaches to genome-wide screen in model <strong>org</strong>anisms.<br />

While protein–protein and protein– DNA networks have been the subject of many systematic surveys, others<br />

critically important cellular components, such as lipids, have to date rarely been studied in large-scale interaction<br />

screens. The importance of protein–lipid interactions is evident from the variety of protein domains<br />

that have evolved to bind particular lipids and from the large list of disorders, such as cancer and bipolar<br />

disorder, arising from altered protein–lipid interactions. The importance of lipids in biological processes and<br />

their under-representation in current biological networks suggest the need for systematic, unbiased biochemical<br />

screens. Here, we report a screen to catalog protein–lipid interactions in yeast using a lipid arrays. To<br />

illustrate the data set’s biological value, we studied further several novel interactions with sphingolipids, a<br />

class of conserved bioactive lipids with an elusive mode of action. Integration of live-cell imaging suggests<br />

new cellular targets for these molecules, including several with pleckstrin homology (PH) domains. The<br />

dataset presented here represents an excellent resource to enhance the understanding of lipids function in<br />

eukaryotic systems.<br />

24 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 25


abStract For SPeakerS<br />

MAREIKE WEIMANN*, JONAThAN WOODSMITh*, ARNDT GROSSMANN, zIYA …zKAN, PETRA BIRTh,<br />

DAVID MEIERhOfER, SASChA SAUER, ULRICh STELzL OTTO-WARBURG LABORATORY, MAx-PLANCK<br />

INSTITUTE fOR MOLECULAR GENETICS (MPIMG)<br />

A Y2H-seq approach to define the protein methyltransferase interactome<br />

Protein methylation, in particular on arginine and lysine residues, is an important, widespread postranslational<br />

modification. The large number of human methyltransferases, potential demethylases and Merecognition<br />

domain containing proteins, which are not only expressed in a large variety of tissues but also at<br />

different subcellular localizations, indicate roles in many cellular processes other than epigenetic regulation.<br />

However, the transient nature of substrate enzyme recognition, the lack of affinity reagents and appropriate<br />

tools to detect methyltransferase substrate pairs largely hampered progress in defining the global role of<br />

non-histone protein methylation.<br />

Here we present a novel proteome wide Y2H protein interaction screening approach involving a second<br />

generation sequencing readout. The method has significantly improved sensitivity in comparison to our state<br />

of the art Y2H matrix screening protocol. Importantly, 2nd generation sequencing provides a quantitative<br />

readout that correlates very well with the retest success rate indicative of the quality of the PPI information.<br />

Y2H-seq will thus accelerate large scale interactome mapping efforts.<br />

We applied the Y2H-seq method to comprehensively screen proteins involved in methylation and demethylation,<br />

i.e. protein methyl transferases (PMTs) and JMJ-domain containing putative demethylases<br />

(PDeMs) such as LSM1, for interacting proteins. We found more than 500 interactions involving 22 PMTs<br />

and PDeMs and 324 potential methylation substrates. Exemplarily, 7 candidate proteins are characterized<br />

with respect to novel R and K methylation sites using a mass spectrometry approach. The final network is<br />

comprehensively annotated, validated in co-IP experiments and will serve as a major informational resource<br />

to define cellular roles of non-histone protein methylation.<br />

abStract For SPeakerS<br />

A.J. MARIAN WALhOUT<br />

UNIVERSITY Of MASSAChUSETTS MEDICAL SChOOL<br />

WORCESTER, MA<br />

A.J. Marian Walhout obtained her BS and PhD degrees in Biochemistry/Medicine<br />

from Utrecht University in the Netherlands. After a post-doc at Harvard<br />

Medical School, she became a Faculty member at the University of Massachusetts<br />

Medical School, Worcester, USA in 2003. She is currently a Professor<br />

in Molecular Medicine and a Co-Director of the Program in Systems Biology.<br />

Gene regulatory networks<br />

Transcriptional regulation of gene expression is pivotal to all biological processes.<br />

Each of our ~20,000 genes must be expressed at the right place, time<br />

and level, and under the right conditions. As a consequence, improper gene expression is associated with<br />

a myriad of human diseases, including congenital disorders, cancer and obesity. The basic mechanisms<br />

of RNA polymerase II transcription have been studied in great detail for decades. However, little is known<br />

about the gene regulatory networks (GRNs) that are composed of physical and regulatory interactions<br />

between transcription factors and their target genes, and that orchestrate spatiotemporal gene expression<br />

during development or upon physiological stresses and pathological insults. Our long-term goal is to comprehensively<br />

characterize the structure, function and evolution of complex metazoan GRNs. As a model we<br />

use the nematode C. elegans, which is amenable to a variety of genetic and genomic approaches. We have<br />

developed a variety of gene-centered methods for the elucidation of GRNs. Progress will be discussed.<br />

26 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 27


abStract For SPeakerS<br />

BEN LEhNER<br />

EMBL-CRG SYSTEMS BIOLOGY RESEARCh UNIT AND ICREA, CENTRE fOR<br />

GENOMIC REGULATION, UPf, BARCELONA, SPAIN<br />

Ben Lehner is an ICREA Research Professor at the EMBL-CRG Systems Biology<br />

Program in Barcelona. He has a degree (Natural Sciences) and PhD (protein interaction<br />

networks, antisense transcription) from the University of Cambridge<br />

and was a post-doctoral fellow in the Fraser lab at the Wellcome Trust Sanger<br />

Institute (genetic interactions, large-scale integrated networks). Since Dec<br />

2006 he has been at the CRG, funded by the ERC, EMBO YIP program, ERASys-<br />

Bio+, AGAUR, Plan Nacional, and the CRG. The main aim of the lab is to answer<br />

basic questions in genetics, using highly quantitative or systematic experimental<br />

and computational approaches in different model systems, as necessary.<br />

The biology of individuals<br />

To what extent is it possible to predict the phenotypic differences among individuals from their completely<br />

sequenced genomes? We use model <strong>org</strong>anisms (yeast, worms) to understand when you can, and why you<br />

cannot, predict the biology of an individual from their genome sequence.<br />

abStract For SPeakerS<br />

onships are utilized.<br />

BERNhARD PALSSON<br />

UCSD / DTU - SAN DIEGO, CA / LYNGBY<br />

Systems Biology of Metabolism<br />

The full genome sequences that began to appear some 15 years ago enabled<br />

the bottom-up reconstruction of biochemical reaction networks that operate<br />

in a particular target <strong>org</strong>anism. Such reconstructions can be converted into a<br />

mathematical format that represents mechanistic genotype-phenotype relationship.<br />

This relationship is fundamental in biology, and it has a very different<br />

characteristic that the basic physical laws elucidated about a century ago. In<br />

this talk we; 1) put the field of molecular systems biology into a historical<br />

context, 2) review the workflows and procedures that have been developed<br />

over the past decade for network reconstruction, and 3) go through a series<br />

of examples that show how mechanistic metabolic genotype-phenotype relati-<br />

28 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 29


You are<br />

ready<br />

Building on the best of two industry-leading capabilities, our new LC/MS systems identify,<br />

quantitate and confi rm compounds in the most demanding samples. Whether unraveling<br />

protein structures, developing new drugs, or analyzing trace pesticides, we provide new<br />

possibilities to help move your science and productivity forward. And as you drive discovery<br />

and face new unknowns, our advanced systems and software provide unprecedented<br />

confi dence in the fi nal result. Whatever challenges arise in the future, you’ll be ready.<br />

for the toughest<br />

analytical challenges<br />

• see our new line up at <strong>www</strong>.thermoscientifi c.com/ready<br />

© 20<strong>11</strong> Thermo Fisher Scientifi c Inc. All rights reserved.<br />

SPonSorPage<br />

Orbitrap Elite Hybrid LC-MS<br />

Gold standard for accurate mass<br />

and now to 240,000 resolution<br />

Q Exactive Benchtop Orbitrap<br />

High resolution, accurate mass and<br />

simultaneous qual/quan<br />

Velos Pro Ion Trap<br />

Fastest, most sensitive ion trap<br />

with HCD fragmentation<br />

TSQ Quantum XLS Ultra<br />

GC-MS/MS<br />

High sensitivity and selectivity in<br />

GC and LC triple quadrupole MS<br />

abStractS For PoSterS<br />

ThOMAS R COx1 , ERWIN SChOOf2, SARA zANIVAN3, RUNE LINDING2 AND JANINE T ERLER1<br />

1 BIOTECh RESEARCh AND INNOVATION CENTRE, UNIVERSITY Of COPENhAGEN, DENMARK 2 CENTER<br />

fOR BIOLOGICAL SEqUENCE ANALYSIS, TEChNICAL UNIVERSITY Of DENMARK, DENMARK 3 BEATSON<br />

INSTITUTE fOR CANCER RESEARCh, GLASGOW, UK<br />

Remodelling of the ECM as a critical mediator of tumour metastasis<br />

Tumour metastasis is a highly complex, dynamic and inefficient process involving multiple steps, yet accounts<br />

for over 90% of cancer patient deaths. The tumour microenvironment and in particular the extracellular<br />

matrix is a key component in driving this process at multiple stages. Both the biochemical and biomechanical<br />

properties or tumour extracellular matrix (ECM) contribute to progression. Metastatic tumours show<br />

elevated ECM remodelling and increased stiffness in comparison to their non-metastatic counterparts and<br />

these changes in stiffness are known to drive metastatic cell behaviour although the underlying molecular<br />

mechanisms remain elusive. The aim of this project is to utilise multiple molecular approaches and evaluate<br />

both the molecular and behavioural changes occurring in tumour cells in response to ECM remodelling and<br />

in particular changes in ECM stiffness. By computationally integrating molecular and phenotypic data, we<br />

aim to derive a molecular network associated with stiffness and identify key enablers of metastatic progression.<br />

Using breast and colorectal cancer models, we have found that metastatic tumours are stiffer than matched<br />

non-metastatic tumours. We have shown that increasing ECM stiffness can drive the invasive behaviour of<br />

the non-metastatic cancer cells. We observe associated cell signalling events and gene expression changes,<br />

and are further investigating the molecular networks associated with enhanced metastasis in response<br />

to increased stiffness.<br />

The goal of the study is to predict and test novel therapeutic strategies for the treatment and prevention of<br />

metastasis that could then be translated into the clinic.<br />

30 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 31


PETER hUSEN1, KIRILL TARASOV2, ALBERT CASANOVAS1, KIM EKROOS2, ChRISTER S. EJSING1<br />

1DEPARTMENT Of BIOChEMISTRY AND MOLECULAR BIOLOGY, UNIVERSITY Of SOUThERN DENMARK<br />

2zORA BIOSCIENCES OY, ESPOO, fINLAND<br />

A software routine for charting the composition and dynamicsof lipid metabolic networks<br />

Lipids constitute a large part of the metabolism of eukaryotic <strong>org</strong>anisms where they play an important role as<br />

constituents of the membranes separating various cellular compartments from each other and the cell itself<br />

from its surroundings. The specific lipid composition of membranes is believed to play an important functional<br />

role in controlling membrane shape and to affect both affinity and function of membrane-embedded<br />

proteins. The importance of the lipidome and its entanglement in the overall cellular metabolic network call<br />

for efficient and reliable methods to quantify lipids as part of the general efforts in mapping the metabolome.<br />

We have established a software tool and framework for streamlined quantification of cellular lipidomes. The<br />

approach is based on high resolution mass spectrometry using direct infusion and application of internal<br />

standards to allow absolute quantification. The software platform is based on targeted extraction of spectral<br />

data using target lists taylored to the individual experiments from a lipid database. The spectral peak<br />

extraction supports offline calibration of the spectra using lock masses to improve mass accuracy and thus<br />

maintain high specificity even in crowded spectra.<br />

The efficient and automated lipidome-wide quantification facilitates studies of the kinematics and complex<br />

regulation of lipid metabolic pathways, e.g. by flux analysis. Particularly, we have employed this framework<br />

to chart the dynamics of the yeast metabolic network under various growth conditions. A total of ~240 lipid<br />

species were quantified simultaneously in this experiment at various time points for each growth condition.<br />

Using the software toolbox, the evolution of both individual lipid species abundances and overall lipidome<br />

features are now efficiently tracked, which provides a huge amount of information on the lipid metabolic<br />

network of this <strong>org</strong>anism.<br />

abStractS For PoSterS abStractS For PoSterS<br />

JOAN ChANG,ANDREAS hADJIPROCOPIS, MARIE-CLAUDE DJIDJA, JOhN SINCLAIR, CLAUS JORGENSEN,<br />

RUNE LINDING, JANINE ERLER<br />

MoleCular Networks Associated with Hypoxia-regulated Metastasis<br />

Hypoxia has been shown to increase metastasis, however the underlying molecular mechanisms are still<br />

unclear. As of yet, there have been no systematic approaches to investigate the proteomic networks associated<br />

with hypoxia-driven tumour progression. Here, we investigated the molecular networks associated with<br />

hypoxia in both in vitro and in vivo samples, using a proteomics approach.<br />

4T1mouse mammary carcinoma cells were subjected to stable isotope labeling by amino acids in cell culture<br />

(SILAC), incubated for 24 hours in air or hypoxia, followed by quantitative mass spectrometry ( MS) using a<br />

ThermoFisher OrbiTrap-Velos. 4T1 cells were also implanted into mice to form in vivo tumours.<br />

Samples were isolated from regions of normoxia and hypoxia in the tumours using laser capture microdissection<br />

(Leica), and proteins were identified through MS (OrbiTrapVelos). Finally, sections of tumours<br />

were subjected to MALDI-MSI using an ABSciex 5800 to determine the distribution of various proteins in the<br />

tumour.<br />

We identified hypoxia-regulated proteins in cancer cells in vitro, and verified our findings with protein identification<br />

in in vivo samples. Furthermore, MALDI-MSI allowed us to visualize the distribution of proteins in<br />

the tumour sections, and confirm hypoxic localization. We developed a computer programme that analysed<br />

all proteins quantitatively detected in the in vitro SILAC system, and visualized them using Cytoscape. This<br />

enabled the build up of an interaction network of all proteins detected, including those that were not significantly<br />

up-or down-regulated, providing a snapshot of the global state of the protein networks within the cells.<br />

This is the first comprehensive study investigating hypoxia-regulated proteins both in vitro and in vivo. This<br />

presents an exciting prospect that with further optimization, these techniques will provide a powerful tool to<br />

identify specific proteins associated with the microenvironment that drive cancer progression.<br />

32 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 33


KRzYSzTOf fUJAREWICz<br />

SILESIAN UNIVERSITY Of TEChNOLOGY, POLAND<br />

Optimal sampling for parameter estimation of cell signaling pathway models based on multivariate measurements<br />

Mathematical modeling of cell signaling pathways became a very important and challenging problem<br />

in recent years. The importance comes from possible application of obtained models. It may help us to<br />

understand phenomena appearing in single cells and in cell populations. Furthermore, it may help us with<br />

discovering new drug therapies.<br />

Mathematical models of cell signaling pathways take different forms. The most popular way of mathematical<br />

modeling is to use a set of nonlinear ordinary differential equations (ODEs).<br />

Usually there are many hypotheses about the structure of the model (set of variables and set of phenomena).<br />

It leads to several rival models and one of them should be chosen among others. This choice may be<br />

supported by so called T-optimal experiment design.<br />

The next step, estimation of the model’s parameters, is also very complicated because of the nature of<br />

measurements. The blotting technique usually gives only semi-quantitative observations which are very<br />

noisy and they are collected only at limited number of time moments. Once more, the accuracy of parameter<br />

estimation may be significantly improved by proper experiment planning.<br />

We present two-stage experiment plan optimization algorithm. The first step is gradient-based D-optimization<br />

procedure to find all stationary points. The second step is pair-wise replacement for finding optimal<br />

numbers of replicates of measurements. The algorithm is applied to one of cell signaling pathway model,<br />

known from the literature.<br />

For multivariate measurements the presented approach gives, in general, different optimal time points for<br />

different variables. In practice it is better to take measurements at the same time points. We show how this<br />

constraint influences the final result of optimization.<br />

This work has been supported by Polish Ministry of Education and Science under grant N N514 4<strong>11</strong>936.<br />

abStractS For PoSterS abStractS For PoSterS<br />

IA VAN DIJK, MJ OUDhOff, J VAN DE AMEIDE, JGM BOLSChER, ECI VEERMAN.<br />

DEPARTMENT Of ORAL BIOChEMISTRY, ACADEMIC CENTRE fOR DENTISTRY AMSTERDAM (ACTA), UNI-<br />

VERSITY Of AMSTERDAM AND VU UNIVERSITY AMSTERDAM, ThE NEThERLANDS.<br />

Histatin from saliva promotes wound healing: Salivary peptide histatin induces GPCR- and ERK-dependent<br />

cell migration<br />

Wounds in the oral cavity heal faster than at other sites in the human body, e.g. the skin. Several factors<br />

have been implicated in this phenomenon, including the presence of saliva, which in rodents is a reservoir of<br />

many growth factors, such as epidermal growth factor (EGF) and nerve growth factor (NGF). In humans the<br />

identity of the involved compounds has remained elusive, since the saliva concentration of growth factors is<br />

1,000 to 100,000 times lower than in rodent saliva. The present study was aimed at the identification of the<br />

factor(s) responsible for the alleged wound healing power of saliva, and a first characterization of the cellular<br />

processes.<br />

Using an in vitro scratch assay we found that human saliva is able to induce epithelial cell migration, without<br />

involvement of EGF. By testing protein fractions of human saliva obtained by HPLC, we identified salivary<br />

histatins as the main migration-inducing factors in saliva. The ᴅ-enantiomer of histatin is not active, pointing<br />

towards the involvement of a stereospecific receptor. N-to-C terminal cyclization of histatin potentiates the<br />

molar activity approximately 1,000-fold, suggesting that the activation of the putative receptor requires a<br />

specific spatial conformation of the peptide. Histatin activity was abolished both by inhibition of mitogen activated<br />

protein kinases ERK1/2 and MEK, and by pertussis toxin, an inhibitor of G protein coupled receptors,<br />

suggesting the involvement of a GPCR-dependent ERK1/2 signaling pathway. Using a functional phosphokinase<br />

assay, we found, in addition, activation as well as inhibition of a large number of other phosphokinases<br />

upon treating epithelial cells with histatin.<br />

Conclusion: Our results emphasize the importance of histatin in human saliva for tissue protection and recovery,<br />

and establish the experimental basis for the development of synthetic histatins as novel wound-healing<br />

agents.<br />

34 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 35


VEIT SChWÄMMLE AND OLE NøRREGAARD JENSEN DEPARTMENT Of BIOChEMISTRY AND MOLECULAR<br />

BIOLOGY, UNIVERSITY Of SOUThERN DENMARK, CAMPUSVEJ 55, DK-5230 ODENSE M, DENMARK<br />

A simple model of histone mark propagation reproduces dynamic features of chromatin structure and transcriptional<br />

regulation<br />

Heterochromatin, the transcriptionally inactive part of the genome, is densely packed and contains histone<br />

H3 that is methylated at Lys 9 (H3K9me). The propagation of H3K9me in nucleosomes along the DNA in<br />

chromatin is antagonized by methylation of H3 Lysine 4 (H3K4me), which is related to euchromatin and<br />

active genes. Both modification marks are assumed to be initiated within distinct nucleation sites in the<br />

DNA and to propagate bi-directionally. We propose a simple computer model that simulates the distribution<br />

of heterochromatin in human chromosomes. Our model explains how heterochromatin is prevented from<br />

occupying regions of active gene expression through continuous competition between the two marks. The<br />

computa¬tional simulations of heterochromatin distribution in chromosomes are in agreement with previously<br />

reported experimental observations. An extended model considers multiple co-existing histone marks<br />

and reproduces important features of chromatin, including switch-like behavior for activation/inactivation of<br />

chromatin domains, and temporal and spatial regulation of genes/chromatin by histone modification domain<br />

rearrangements. The simulations reproduce not only complex spatial conformations but also temporal<br />

features, such as oscillatory behavior found in cell cycle processes and circadian rhythms. The propagation<br />

rates of co-existing histone marks play a crucial role for the transition between distinct chromatin and gene<br />

regulatory states of the system. Our model demonstrates that the interplay of multiple co-existing histone<br />

modifications can explain different chromatin states, thereby controlling for instance cell growth, diff erentiation<br />

and metabolism in a spatio-temporal manner.<br />

abStractS For PoSterS abStractS For PoSterS<br />

ANNA-MARIA LAhESMAA-KORPINENA, SARI JALKANENB, JUKKA VAKKILAB, KIMMO PORKKAB, SATU<br />

MUSTJOKIB, SAMPSA hAUTANIEMIA<br />

A RESEARCh PROGRAMS UNIT, GENOME-SCALE BIOLOGY AND INSTITUTE Of BIOMEDICINE, BIOChEMIS-<br />

TRY AND DEVELOPMENTAL BIOLOGY, UNIVERSITY Of hELSINKI, fINLAND; B hEMATOLOGY RESEARCh<br />

UNIT, BIOMEDICUM, DIVISION Of MEDICINE, hELSINKI UNIVERSITY CENTRAL hOSPITAL AND UNIVER-<br />

SITY Of hELSINKI, fINLAND<br />

Single cell analysis of phosphoprotein network response to tyrosine kinase inhibitor therapy in chronic<br />

myeloid leukemia patients<br />

Tyrosine kinase inhibitors (TKI) are currently the therapy choice for treatment of chronic myeloid leukemia<br />

(CML) patients. TKIs have been shown to be immunosuppressive in vitro, while on the other hand some<br />

patients experience immunoactivation during dasatinib-therapy. Our previous results show an effect of dasatinib<br />

treatment on the basal activation status of the STAT3 signaling pathway, and the monocyte populations<br />

of CML patients at the diagnosis phase were found to respond poorly to cellular cytokine stimulation, which<br />

was restored to normal levels during TKI therapy. Previous studies, however, focused on protein expression<br />

medians of the cell populations studied, and were not able to utilize the single cell information from flow<br />

cytometry (FCM) measurements.<br />

To study the phenomenon in more detail, we used a computational framework designed for large scale FCM<br />

data analysis to automatically gate the lymphocyte populations and interesting sub-populations for individual<br />

patients and various cytokine stimulations. We studied the individual differences in phosphoprotein networks<br />

of CML patients after TKI therapy of patients at diagnosis (n=10), patients after dasatinib (n=10) or imatinib<br />

(n=10) treatment and control subjects (n=7). We measured the expression of phosphorylated ERK1/2,<br />

STAT1, STAT3, STAT5 and STAT6, and cell surface markers with FCM after various cytokine treatments. We<br />

constructed comprehensive signaling networks for the phosphoproteins using known databases for proteinprotein<br />

interactions and pathway information. Using each distinct cell population, we clustered the phosphoprotein<br />

profiles for the various patient groups and looked for clusters of differential signaling networks within<br />

patients and cell populations. We identified various cell population shifts showing differential phosphoprotein<br />

expression affecting immunological signaling pathways.<br />

36 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 37


BLAKE BORGESON(1), CUIhONG WAN(2), ANDREW EMILI(2), EDWARD MARCOTTE(1)<br />

1: ThE UNIVERSITY Of TExAS AT AUSTIN, CENTER fOR SYSTEMS AND SYNThETIC BIOLOGY<br />

2: ThE UNIVERSITY Of TORONTO, BANTING AND BEST DEPARTMENT Of MEDICAL RESEARCh<br />

Conservation and Divergence of Protein Complexes Across Evolution<br />

Despite agreement that the vast majority of life’s processes at a cellular level are carried out by complexes<br />

of multiple proteins, knowledge of all the complexes formed in a cell and their members is a distant goal. By<br />

using a new approach developed by collaborators Havugimana and Hart, et al, consisting of 1) subjecting<br />

biological samples to many levels of many types of fractionations, 2) using LC-MS/MS to quantify protein<br />

levels in each fraction, and 3) processing the data through a machine learning pipeline, we identify putative<br />

soluble (non¬membrane) complexes using a high-throughput all-by-all approach. By incorporating additional<br />

functional genomic information into our learning process, we are able to reconstruct maps of complexes that<br />

so far seem to rival in quality those generated using previous, more labor-intensive methods. Here, we apply<br />

the approach to biological samples from many <strong>org</strong>anisms, including human, sea urchin, fly, worm, and multicellular<br />

amoebas, in order to rapidly learn soluble complexes in many species. From such maps we identify<br />

interesting conservation and divergence of complexes not previously well-understood or studied.<br />

abStractS For PoSterS abStractS For PoSterS<br />

ChRISTOPhER A. BARNES, ALESSIO MAIOLICA, RUEDI AEBERSOLD<br />

INSTITUTE Of MOLECULAR SYSTEMS BIOLOGY, ETh züRICh, SWITzERLAND<br />

A Global Kinase-Substrate Network in Yeast Mapped Using In Vitro Phosphorylation and Phosphoproteomics<br />

Kinase inhibitor drugs have been a major focus in the pharmaceuticals industry over the past decade.<br />

Despite extensive development of these inhibitors, their effectiveness has been limited. It is thought that<br />

the difficulties associated with kinase inhibitor drug effectiveness relates to network compensation events<br />

in the cell that allow other kinases to replace the lost function of the inhibited enzyme. To date, it has been<br />

difficult to link kinases to substrates at the amino acid level with methodologies suitable to creating a global<br />

kinase-substrate wiring diagram. Identifying direct kinase-substrate relationships for each kinase could help<br />

researchers to design pharmaceutical solutions that take into account network compensations. The aim<br />

of this project is to identify kinase-substrate relationships in yeast for all of the kinases in S. cerevisiae. In<br />

previous studies in the lab1, experiments were performed where kinases were sequentially deleted, but the<br />

identification of direct kinase-substrate relationships with these deletion studies proved difficult. Here, we<br />

have taken a large library of the most regulated phosphosites from these deletion experiments and synthesized<br />

unphosphorylated peptides suitable for in vitro phosphorylation followed by LC-MS/MS identification of<br />

the phosphorylated product peptides. We use endogenously expressed immunopurified kinases followed by<br />

a phosphopeptide enrichment step prior to mass spectrometry analysis. Our phosphorylation assays have<br />

shown good specificity in vitro as evidenced by the identification of specific known consensus motifs and<br />

we have begun to identify subfamily redundancies in the substrate networks as this yeast phosphorylation<br />

wiring diagram is beginning to emerge.<br />

References:<br />

1. Bodenmiller et al. Sci. Signal. 3, rs4 (2010).<br />

38 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 39


PAU CREIxELL1, DORIEN WIJTE1, ERWIN SChOOf1, AGATA WESOLOWSKA2, ThOMAS NORDAhL PETERS-<br />

EN2, RAMNEEK GUPTA2, hIROAKI ITAMOChI3, JANINE ERLER4 & RUNE LINDING1<br />

1 CELLULAR SIGNAL INTEGRATION GROUP (C-SIG), CENTER fOR BIOLOGICAL SEqUENCE ANALYSIS (CBS),<br />

DEPARTMENT Of SYSTEMS BIOLOGY, TEChNICAL UNIVERSITY Of DENMARK (DTU), DK-2800LYNGBY,<br />

DENMARK.2 fUNCTIONAL hUMAN VARIATION, CENTER fOR BIOLOGICAL SEqUENCE ANALYSIS (CBS),<br />

DEPARTMENTOf SYSTEMS BIOLOGY, TEChNICAL UNIVERSITY Of DENMARK (DTU), DK-2800 LYNGBY,<br />

DENMARK.3 DEPARTMENT Of OBSTETRICS AND GYNECOLOGY, TOTTORI UNIVERSITY SChOOL Of MEDI-<br />

CINE, 36-1NIShIChO, YONAGO 683-8504, JAPAN.4 BIOTECh RESEARCh & INNOVATION CENTRE (BRIC),<br />

COPENhAGEN UNIVERSITY (KU), DK-2200COPENhAGEN, DENMARK.<br />

Kinomewide Discovery of Functional Cancer Mutations.<br />

Systematic sequencing and targeted studies of tumors are revealing protein kinases as key drivers of cancer.<br />

More than a thousand distinct cancer mutations (equal to around 3fold enrichment) have been identified<br />

in protein kinases alone. However, pinpointing whichof these mutations are functional and how they dysregulate<br />

signaling networks in cells remains a challenge. We have developed an algorithm, ReKINect, that<br />

identifies functionalmutations and predicts how these loss-and gain-of-function mutations lead to changes<br />

insignaling network dynamics and architecture. By combining genome-wide sequencing datawith proteomics<br />

data we want to show how functional mutations can result in kinaseactivation, kinase inactivation, activation<br />

rewiring or substrate rewiring. We define a superSILAC-type strategy to validate our in silico predictions,<br />

which suggests that theeffect of these different types of mutations can be observed at the (phospho)proteomelevel.<br />

• Creixell et al. Navigating Cancer Network Attractors for Tumor-specific Therapy.Submitted.<br />

• Creixell et al. Kinome-wide Discovery of Functional Cancer Mutations. In preparation.<br />

• Creixell et al. Mutational Properties of Amino Acid Residues - Implications for Evolvabilityof Phosphorylatable<br />

Residues. Accepted in Philosophical Transactions of the RoyalSociety B.<br />

abStractS For PoSterS abStractS For PoSterS<br />

SIMONE DAMINELLI†, V. JOAChIM hAUPT†, MATThIAS REIMANN AND MIChAEL SChROEDER<br />

BIOTEC, TU DRESDEN, GERMANY.<br />

Drug Repositioning through Incomplete Bi-cliques in an Integrated Drug-Target-Disease Network<br />

Recently, there has been much interest in gene-disease networks and polypharmacology as a basis for drug<br />

repositioning. Here, we integrate data from structural and chemical databases to create a drug-target-disease<br />

network for 147 promiscuous drugs, their 553 protein targets, and 44 disease indications. Visualizing<br />

and analyzing such complex networks is still an open problem. We approach it by mining the network for<br />

network motifs of bi-cliques. In our case, a bi-clique is a subnetwork in which every drug is linked to every<br />

target and disease. Since the data is incomplete, we identify incomplete bi-cliques, whose completion introduces<br />

novel, predicted links from drugs to targets and diseases. We demonstrate the power of this approach<br />

by repositioning cardiovascular drugs to parasitic diseases, by predicting the cancer-related kinase PIK3CG<br />

as novel target of resveratrol, and by identifying for five drugs a shared binding site in four serine proteases<br />

and novel links to cancer, cardiovascular, and parasitic diseases.<br />

† These authors contributed equally to this work.<br />

40 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 41


ELENA NIKONOVA DIRK fEY† MIKhAIL TSYGANOV‡ BORIS KhOLODENKO<br />

Temporal dynamics of two layer GTPase cascade with GDI binding<br />

GTPases control intracellular signaling and are responsible for a variety of vital cellular mechanisms such<br />

as cytoskeleton formation, motility and vesicle transport. Functioning of GTPases occurs due to monomeric<br />

G proteins cycling between inactive GDP-bound and active GTP-bound states. The reaction is catalyzed<br />

by guanine nucleotide exchange factors (GEFs) for the transformation from GDP to GTP and by GTPase<br />

activating proteins (GAPs) for the reverse transformation. Malfunction of the GTPases often occurs due<br />

to the deregulated expression and activities of GAP and GEF, which was found to be one of the causes of<br />

tumorogenesis [2]. Guanine nucleotide dissociation inhibitors (GDIs) were also found to regulate the GTPase<br />

cycles by binding to the inactive GDB-bound form and transporting the formed complex away from the membrane<br />

to the cytosol. Previous characterization of spatiotemporal dynamics showed that models comprised<br />

of two GTPases without GDI binding can exhibit 3 distinct regimes: sustained oscillations, bistable switches<br />

and excitable behavior [1]. The current work explores the change in dynamics by allowing GDI to bind to<br />

the active and inactive sites of both GTPases. In particular, our results show that as the dissociation and<br />

association rates of GDI binding are varied, the two layer GTPase model can exhibit transformations within<br />

previously outlined regimes.<br />

References<br />

[1] M.A.Tsyganov, W. Kolch and B.N. Kholodenko Molecular BioSystems, <strong>2012</strong>.<br />

[2] D. Vigil, J. Cherfils, K.L. Rossman and C.J. Der Nat. Rev. Cancer, 10(12), 842-857, 2010.<br />

*Systems Biology Ireland, University College Dublin, Belfield, Dublin 4<br />

†Systems Biology Ireland, University College Dublin, Belfield, Dublin 4<br />

‡Institute of Theoretical and Experimental Biophysics, Pushchino, Moscow Region, Russia §Systems Biology<br />

Ireland, University College Dublin, Belfield, Dublin 4<br />

abStractS For PoSterS abStractS For PoSterS<br />

ALExEY GOLTSOV (A), DANA fARATIAN(B), SIMON LANGDON(B), DAVID hARRISON(B), JAMES BOWN(A)<br />

(A) CENTRE fOR RESEARCh IN INfORMATICS AND SYSTEMS PAThOLOGY, UNIVERSITY Of ABERTAY<br />

DUNDEE, DUNDEE, UK (B) EDINBURGh BREAKThROUGh RESEARCh UNIT AND DIVISION Of PAThOLOGY,<br />

WESTERN GENERAL hOSPITAL, UNIVERSITY Of EDINBURGh, EDINBURGh, UK<br />

Systems biology of drug sensitivity-resistance transition in PI3K/AKT signalling in cancer<br />

Systems biology offers a useful approach to study dependence of drug efficacy on oncogene-driven<br />

transformations in drug target pathways relevant to cancer therapy. Systems approach was developed to<br />

elucidate mechanisms underlying the changes of the efficacy of monoclonal antibody therapy (trastuzumab,<br />

pertuzumab) targeting HER2 receptor at cancer genome transformation in the PI3K/AKT signalling network<br />

(SN) [Goltsov et al Cell. Signalling 20<strong>11</strong>]. In silico experiments showed that HER2 inhibition sensitises the<br />

SN both to external signals and to kinetic characteristics of the proteins and their expression levels. We suggested<br />

that a drug-induced increase in SN sensitivity to internal perturbations, and specifically mutations,<br />

causes SN fragility. In particular, the SN is vulnerable to mutations that compensate for drug action and this<br />

may result in a drug sensitivity-to-resistance transition in SN [Goltsov et al Cell. Signalling <strong>2012</strong>]. Modelling<br />

showed the increase of SN sensitivity to typical aberrations in cancer causing drug resistance: loss of<br />

PTEN activity, PI3K and AKT mutations, HER2 overexpression, and overproduction of GSK3ᴅ controlling<br />

PTEN activity. In particular, the SN is vulnerable to mutations that compensate for drug action. PTEN loss or<br />

PIK3CA mutation was shown to cause resistance to HER2 inhibition and leads to the restoration of maximal<br />

pAKT signal with a consequent decrease in SN sensitivity. The drug-induced sensitivity of SN was tested in<br />

experiments on ovarian cancer cells which demonstrated that HER2 inhibition increased SN sensitivity to<br />

the second inhibitor targeting downstream pathway, in particular PI3K inhibition. The developed method is<br />

proposed to be used in the pointed development of combined treatment of cancer which provides both synergetic<br />

inhibition of SN activated in cancer and prevention of the SN from acquired drug resistance caused<br />

by oncogenic mutations.<br />

42 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 43


TRI hIEU NIM1, LE LUO2, JACOB K. WhITE1,3, MARIE-VéRONIqUE CLéMENT2, LISA<br />

TUCKER-KELLOGG4<br />

1 COMPUTATIONAL SYSTEMS BIOLOGY PROGRAMME, SINGAPORE-MIT ALLIANCE 2 DEPARTMENT Of<br />

BIOChEMISTRY, YONG LOO LIN SChOOL Of MEDICINE, NATIONAL UNIVERSITY Of SINGAPORE 3 DEPART-<br />

MENT Of ELECTRICAL ENGINEERING AND COMPUTER SCIENCE, M.I.T. 4 MEChANOBIOLOGY INSTITUTE,<br />

NATIONAL UNIVERSITY Of SINGAPORE<br />

Mathematical modeling of Akt phosphorylation dynamics in serum stimulated fibroblasts<br />

Pathological over-activation of Akt is responsible for cell survival advantages in cancer, and may arise from<br />

mechanisms present in normal cells. In normal cells, peaks of high Akt phosphorylation (activation) occur<br />

briefly after serum stimulation, before reaching a moderate steady-state. It is not known whether canonical<br />

mechanisms of Akt regulation are sufficient to explain the observed dynamics of Akt phosphorylation.<br />

Methods: Possible mechanisms for the brief extreme of Akt phosphorylation were represented by a null<br />

hypothesis and four alternative hypotheses. Ordinary differential equation models of the hypotheses were<br />

constructed using our previously published measurements of phosphatidylinositol(3,4,5)-trisphosphate<br />

(PIP3), total-Akt, and Akt-phosphoThr308 (Aktp308) in mouse embryonic fibroblasts. Simulations revealed<br />

qualitative differences in the dynamics of membrane-cytosol translocation. Additional immuno-blots<br />

were performed to measure the dynamics of membrane localization. Results: Two of five hypotheses were<br />

incompatible with the observed dynamics of PIP3 and PDK1. Measurements of membrane Aktp308 and<br />

membrane total-Akt peaked at 5 and 30 minutes, while measurements of PIP3 peaked at 2 minutes; this<br />

could be explained by a hypothetical non-PIP3 mechanism that increases Akt membrane recruitment. Two<br />

remaining hypotheses (Aktp308 sequestration at membrane, and inhibited Akt dephosphorylation) were able<br />

to reproduce many but not all observed trends. Conclusion: We conclude that a non-canonical enhancement<br />

of the Akt pathway occurs downstream of PDK1. According to computational simulations analyzing the<br />

implied dynamics of different hypotheses, three mechanisms are possible, but the data are most consistent<br />

with a non-PIP3 mechanism to increase recruitment of Akt to the membrane.<br />

abStractS For PoSterS abStractS For PoSterS<br />

DASSI E, MALOSSINI A, RE A, MAzzA T, TEBALDI T, CAPUTI L, qUATTRONE A.<br />

LABORATORY Of TRANSLATIONAL GENOMICS - CENTRE fOR INTEGRATIVE BIOLOGY, UNIVERSITY Of<br />

TRENTO, VIA DELLE REGOLE, 101, 38123 MATTARELLO (TN), ITALY.<br />

AURA: Atlas of UTR Regulatory Activity.<br />

The Atlas of UTR Regulatory Activity (AURA) is a manually compiled and comprehensive catalog of human<br />

mRNA untranslated regions (UTRs) and UTR regulatory annotations. Through its intuitive web interface,<br />

it provides full access to a wealth of information on UTRs that integrates phylogenetic conservation, RNA<br />

sequence and structure data, single nucleotide variation, gene expression and gene functional descriptions<br />

from literature and specialized databases.<br />

Reference<br />

http://aura.science.unitn.it<br />

44 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 45


ROBERT J WEAThERITT1, NORMAN E DAVEY1, TOBY J GIBSON1<br />

1 STRUCTURAL AND COMPUTATIONAL BIOLOGY UNIT, EUROPEAN MOLECULAR BIOLOGY LABORATORY,<br />

MEYERhOfSTRASSE 1, 69<strong>11</strong>7 hEIDELBERG, GERMANY<br />

Linear Motifs confer functional diversity onto splice variants<br />

The pre-translational modification of mRNAs by alternative promoter usage and alternative splicing is an<br />

important source of pleiotropy. Despite intensive efforts, our understanding of the functional implications<br />

of this dynamically created diversity is still incomplete. Using the recent expansion in our knowledge of the<br />

interaction modules within intrinsically disordered regions, we analysed the occurrences of protein modular<br />

architecture within alternative exons. We find that regions affected by pre-translational variation are enriched<br />

in linear motifs and phosphorylation sites suggesting that the modulating of exons containing these interaction<br />

modules is an important regulatory mechanism. In particular, we observe PDZ, PTB and SH2 binding<br />

motifs are particularly prone to be altered between splice variants. We also determine that regions affected<br />

by alternative promoter usage are enriched in IDRs suggesting that protein isoform diversity is tightly coupled<br />

to the modulation of IDRs. This study therefore demonstrates that short linear motifs, and to a lesser<br />

extent phosphorylation sites, are key components for establishing protein diversity between splice variants.<br />

abStractS For PoSterS abStractS For PoSterS<br />

MISS SOPhIE KERShAW, DR. JAMES OSBORNE, PROf. hELEN BYRNE, PROf. DAVID GAVAGhAN.<br />

DEPARTMENT Of COMPUTER SCIENCE, UNIVERSITY Of OxfORD; COLLABORATION WITh ThE WEAThER-<br />

ALL INSTITUTE fOR MOLECULAR MEDICINE, UNIVERSITY Of OxfORD.<br />

Proliferation and Cell Fate in the Colorectal Epithelium<br />

Endemic across the developed world, the prevalence of colorectal cancer (CRC) has not been matched by<br />

equivalent success in pharmaceutical development. Drug failure in clinical trial patients after early-stage<br />

success in laboratory tests motivates a more detailed consideration of the<br />

fundamental differences between in vitro and in vivo cell behaviour. To what extent does tissue geometry<br />

influence expression of the subcellular biochemistry in CRC?<br />

We therefore present a novel framework for in silico translation experiments, enabling experimental hypotheses<br />

to be explored through simulations of colorectal tissue. Developed in collaboration with biochemists<br />

at the Weatherall Institute of Molecular Medicine, we provide a representation of colorectal tissue that incorporates<br />

mathematical equations to govern the biochemical behavior of its constituent cells. Our focus rests<br />

on models for two key pathways implicated in early-stage CRC, namely Notch (involved in cell fate specification)<br />

and Wnt (a governor of proliferation).<br />

We present our model for Notch and Wnt signalling, based upon existing models in the literature, and incorporate<br />

crosstalk between the two pathways. We will discuss an embedding of our model within cells of both<br />

monolayer and colorectal geometries, discussing the spatial constraints imposed in these two scenarios and<br />

demonstrating the resultant cell patterning and proliferative behaviour.<br />

This work presents a new approach to CRC modelling on several counts. Firstly, via the inclusion of crosstalk<br />

between Notch and Wnt pathways to demonstrate coupled control over proliferation and lineage decisions.<br />

This research also represents a novel development in modelling the<br />

Notch-Wnt interaction in situ, in its application of multiscale frameworks to examine the impact of tissue<br />

configuration.<br />

46 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 47


RIKU LOUhIMO, TATIANA LEPIKhOVA, OUTI MONNI, SAMPSA hAUTANIEMI<br />

RESEARCh PROGRAMS UNIT, fACULTY Of MEDICINE, UNIVERSITY Of hELSINKI, fINLAND<br />

Comparative Analysis of Algorithms for Integration of Copy Number and Expression Data<br />

Genomic instability is a key enabling characteristic of cancer, and genes that display differential expression<br />

in regions with notable copy-number alteration are likely to be drivers ofcancer. Identifyingsuchdrivergenesfromhigh-throughput<br />

and sequencingdata requires computational tools that are capable of integrating data<br />

from several sources. Hence, several algorithms that integrate copy-number and expression data have been<br />

developed. Their performance, however, has not so far been assessed relative to one another.<br />

We have compared ten algorithms that integrate high-throughput copy number and transcriptomicsdata<br />

using simulated,head and neck squamous cell carcinoma(HNSCC) cellline andlung squamous cell<br />

carcinoma(LUSC) primary tumordata. The simulated data enabled us to assess algorithms’ sensitivity and<br />

specificity. For the comparison with the HNSCC and LUSC data we selected 30 genes, whose expression<br />

has been shown to be altered due to underlying copy-number aberrations in squamous cell carcinomas.<br />

Algorithms exhibit clear differences in sensitivity and specificity, and their performance decreases with small<br />

sample sets.<br />

abStractS For PoSterS abStractS For PoSterS<br />

APURV GOEL, MARC WILKINS<br />

SYSTEMS BIOLOGY INITIATIVE, SChOOL Of BIOTEChNOLOGY AND BIOMOLECULAR SCIENCES, UNIVER-<br />

SITY Of NEW SOUTh WALES<br />

Visualising the Dynamics of the Interactome<br />

An important part of network biology is the visualisation of networks. Visualisation can provide many benefits<br />

not only to communicate findings but as part of experiments themselves. Some network elements such<br />

as hubs, just-in-time assembly and network motifs were discovered with the help of networks. Protein-protein<br />

interaction networks are typically built with interactions collated from many experiments. These networks<br />

are thus composite and show all possible interactions that might occur in a cell. However, these are static<br />

and ignore the dynamics of protein-protein interactions.<br />

We have adapted a Java-based, open source software package known as GEOMI to visualise protein interaction<br />

data in 3 dimensions. In addition we have developed the capability for the software to co-visualise<br />

3-dimensional networks with non-traditional forms of interaction data (‘integrated networks’). In particular<br />

the integration of abundance-based information, specifically time course gene expression data, permits the<br />

construction of 4-dimensional visualisations. This allows the ‘dynamic’ range of protein abundance to be<br />

visualised with experimental time, in the context of the protein interactions. The software and demonstration<br />

videos are available at <strong>www</strong>.systemsbiology.<strong>org</strong>.au/downloads_geomi.html.<br />

We will demonstrate the utility of the software in visualising post-translational modifications, transcriptional<br />

regulation and synthetic lethality data in association with pairwise protein interaction data. We will also<br />

present results on other experiments we have conducted. These will include experiments that investigate the<br />

how the cell regulates the interactions of singlish hub proteins (one or two interaction interfaces, but many<br />

interaction partners). This is done in the context of the yeast cell cycle.<br />

48 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 49


MAREIKE WEIMANN*, JONAThAN WOODSMITh*, ARNDT GROSSMANN, zIYA özKAN, PETRA BIRTh,<br />

DAVID MEIERhOfER, SASChA SAUER, ULRICh STELzL<br />

OTTO-WARBURG LABORATORY, MAx-PLANCK INSTITUTE fOR MOLECULAR GENETICS (MPIMG), BERLIN<br />

A Y2H-seq approach to define the protein methyltransferase interactome<br />

Protein methylation, in particular on arginine and lysine residues, is a physiologically important post translational<br />

modification (PTM). Whilst a large number of human methyltransferases (PMTs), potential demethylases<br />

(DeMs) and methyl-recognition domain containing genes are annotated in the human genome,<br />

systematic characterisation of this PTM remains poorly developed. Canonically studied with regards to its<br />

role epigenetic regulation, methylation components or substrates have been shown to have diverse subcellular<br />

localisations and molecular functions, indicating a wider role in cellular physiology. However, the lack<br />

of affinity reagents and appropriate tools to detect methyltransferase substrate pairs has largely hampered<br />

progress in defining the global role of non-histone protein methylation.<br />

Here we present a novel proteome wide Y2H protein interaction screening approach involving a 2nd generation<br />

sequencing readout that has a significantly improved sensitivity in comparison to our state of the art<br />

Y2H matrix screening protocol. Furthermore, the sequencing readout provides a quantitation that correlates<br />

very well with the retest success rate, indicative of the quality of the PPI information. Importantly, the workflow<br />

presented here allows for rapid scalability using advances in sequencing technology to enable Y2H-seq<br />

to accelerate large scale interactome mapping efforts.<br />

We applied the Y2H-seq method to comprehensively screen proteins involved in either methylation or<br />

demethylation. We present a network of >500 interactions involving 22 PMTs or putative DeMs and 324<br />

potential methylation substrates. The network is validated using co-IP experiments and will serve as a major<br />

informational resource to define cellular roles of protein methylation Furthermore, 7 candidate proteins are<br />

characterized with respect to novel R and K methylation sites using a mass spectrometry approach, highlighting<br />

the utility of the network to identify enzyme-substrate pairs.<br />

abStractS For PoSterS abStractS For PoSterS<br />

MOhAMMAD MOBAShIR AND DR. TILO BEYER<br />

INSTITUTE Of MOLECULAR AND CLINICAL IMMUNOLOGY OTTO-VON-GUERICKE UNIVERSITY LEIPzIGER-<br />

STRASSE 44 39120 MAGDEBURG GERMANY CONTACT: MOhAMMAD.MOBAShIR@MED.OVGU.DE AND<br />

TILO.BEYER@MED.OVGU.DE<br />

Unraveling of Network Motifs in Signal Transduction Networks Using Evolutionary Approach<br />

Living <strong>org</strong>anisms control their behavior and cellular function by propagating signals across multiple levels.<br />

In T-cell signaling, the receptor, after receiving the signal activates downstream molecules that transmit the<br />

signal to the nucleus, in order to control cellular functions such as proliferation, differentiation, and apoptosis.<br />

During this process the change in the protein-levels, post-translational modification of the proteins, and<br />

interaction strengths affect the final response of the cell. To investigate possible design priciples we developed<br />

an in-silico model using evolutionary algorithm and ordinary differential equations. From our current<br />

simulations, we found that (i) kinetics of the final response of signaling pathway is predominantly controlled<br />

by the expression levels of proteins, (ii) the variations in protein-levels within the cell plays critical roles in<br />

controlling the kinetic behavior of the cell and consequently the cellular functions, and (iii) an increase in<br />

the interaction strengths up to a certain level leads to strong activation patterns, i.e. in the cells become<br />

sensitive to stimuli. Thus, it appears that the kinetic properties of the signaling network mainly determine<br />

the response threshold of a cell while the quantitative behavior is controlled by the expression levels of the<br />

proteins.<br />

50 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 51


GRzEGORz SLODKOWICz1, ChRISTOPhER T. WORKMAN1, OLGA RIGINA1, hANS-hENRIK STæRfELDT1,<br />

KRISTOffER RAPACKI1, SøREN BRUNAK1, KASPER LAGE hANSEN1, 2<br />

1 CENTER fOR BIOLOGICAL SEqUENCE ANALYSIS, TEChNICAL UNIVERSITY Of DENMARK, 2800 KGS.<br />

LYNGBY, DENMARK<br />

2 PROGRAM IN MEDICAL AND POPULATION GENETICS, BROAD INSTITUTE, CAMBRIDGE, USA<br />

Prioritization of causal disease genes with InWeb, the inferred human interactome<br />

Protein-protein interactions are an important tool for understanding cellular processes, in particular inferring<br />

protein function, disease-gene finding and predicting protein complex co-membership. Several genomewide<br />

studies provide direct evidence for physical interactions in human but far more data is available for<br />

other species, in particular S. cerevisiae and D. melanogaster. In the IntAct database, for instance, out of<br />

292,000 reported interactions, only 77,000 (26%) are measured directly in human.<br />

We present InWeb, the human inferred interactome, a database aggregating interactions from BIND,<br />

ConsensusPathDB, DIP, DOMINO, GRID, HPRD, I2D, InnateDB, IntAct, MINT and others. Interactions<br />

from other model <strong>org</strong>anisms are transferred to human by orthology. The aggregated interactions are then<br />

rigorously filtered, scored and validated against a gold standard of high confidence interactions. In total,<br />

InWeb integrates over 10 million redundant interactions from 21 databases.<br />

Earlier versions of InWeb have been previously used in several studies, including an integrative analysis of<br />

protein complexes implicated in genetic disorders (Lage, Karlberg et al., Nature 2007), a disease chemical<br />

biology database (Taboureau, Nielsen et al., Nucleic Acids Research 20<strong>11</strong>), a novel approach to SNP mapping<br />

(Rossin, Lage et al, PLoS Genetics 20<strong>11</strong>) and others. Here we present InWeb version 5 which provides<br />

a state-of-the-art view of the human interactome.<br />

abStractS For PoSterS abStractS For PoSterS<br />

KATRIN SAMEITh1, MARIAN GROOT KOERKAMP1, MARIEL BROK1, DIK VAN LEENEN1, NAThALIE BRA-<br />

BERS1, ChEUK KO1, TINEKE LENSTRA1, JORIS BENSChOP1, SANDER VAN hOOff1, BEREND SNEL2,<br />

PATRICK KEMMEREN1, fRANK hOLSTEGE1<br />

1MOLECULAR CANCER RESEARCh, UNIVERSITY MEDICAL CENTRE UTREChT, UNIVERSITEITSWEG 100,<br />

3584 CG UTREChT, ThE NEThERLANDS 2DEPARTMENT Of BIOLOGY, UTREChT UNIVERSITY, PADUALAAN<br />

8, 3584 Ch UTREChT, ThE NEThERLANDS.<br />

An atlas of genetic interactions between gene specific transcription factors in<br />

Saccharomyces cerevisiae<br />

Transcription plays a key role in cellular processes and its regulation is paramount for a cell’s homeostasis.<br />

Environmental cues, transmitted through signaling pathways, cause activation or repression of gene specific<br />

transcription factors (GSTFs). How GSTFs regulate each other and how they combine to regulate transcription<br />

across the genome are important research questions. To this end, we systematically measure the effect<br />

of deleting each individual GSTF on gene expression genome-wide. So far, a set of 183 gene expression<br />

profiles has been generated. These profiles show that around half of all GSTF deletions do not result in any<br />

transcriptional changes. This can partly be explained by redundancy relationships between GSTFs, whereby<br />

the loss of one GSTFs is compensated by the presence of another GSTF. To study GSTF redundancy<br />

relationships, we selected 98 GSTF pairs. The results demonstrate that gene expression profiling of double<br />

deletion mutants allow for detailed understanding of combinatorial control through GSTF pairs. Regulatory<br />

circuitries and redundancy relationships between GSTFs are investigated through the determination of genes<br />

that are directly affected in transcription upon the deletion of one GSTF, or upon the combined deletion<br />

of two GSTFs. To support novel relationships found between pairs of GSTFs, we are currently examining<br />

how DNA binding of one GSTF is altered in the deletion of its partner GSTF.<br />

52 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 53


ChRISTOf WINTER1, GLEN KRISTIANSEN2, STEPhAN KERSTING3, JANINE ROY1, DANIELA AUST4,<br />

ThOMAS KNOSEL5, PETRA RüMMELE6, BEATRIx JAhNKE3, VERA hENTRICh3, fELIx RüCKERT3, MARCO<br />

NIEDERGEThMANN7, WILKO WEIChERT8, MARCUS BAhRA9, hANS J. SChLITT10, UTz SETTMAChER<strong>11</strong>,<br />

hELMUT fRIESS12, MARKUS B¨UChLER<strong>13</strong>, hANS-DETLEV SAEGER3, MIChAEL SChROEDER1, ChRISTIAN<br />

PILARSKY3, ROBERT GRüTzMANN3<br />

1 DEPARTMENT Of BIOINfORMATICS, BIOTEChNOLOGY CENTER, TEChNISChE UNIVERSIT ¨ AT DRESDEN,<br />

GERMANY, 2 INSTITUTE Of PAThOLOGY, UNIVERSIT ¨ ATSSPITAL z¨ URICh, zURICh, SWITzERLAND, 3<br />

DEPARTMENT Of VISCERAL, ThORACIC, AND VASCULAR SURGERY, UNIVERSITY hOSPITAL CARL GUSTAV<br />

CARUS, TEChNISChE UNIVERSIT ¨ AT DRESDEN, GERMANY, 4 INSTITUTE Of PAThOLOGY, UNIVERSITY<br />

hOSPITAL CARL GUSTAV CARUS, TEChNISChE UNIVERSIT ¨ AT DRESDEN, GERMANY, 5 INSTITUTE Of PA-<br />

ThOLOGY, UNIVERSITY Of JENA, GERMANY, 6 INSTITUTE Of PAThOLOGY, UNIVERSITY Of REGENSBURG,<br />

GERMANY, 7 DEPARTMENT Of SURGERY, UNIVERSITY hOSPITAL MANNhEIM, GERMANY, 8 DEPARTMENT<br />

Of PAThOLOGY, ChARIT ´ E, BERLIN, GERMANY, 9 DEPARTMENT Of SURGERY, ChARIT ´ E, BERLIN,<br />

GERMANY, 10 DEPARTMENT Of SURGERY, UNIVERSITY hOSPITAL REGENSBURG, GERMANY, <strong>11</strong> DEPART-<br />

MENT Of SURGERY, UNIVERSITY hOSPITAL JENA, GERMANY, 12 DEPARTMENT Of SURGERY, TEChNIS-<br />

ChE UNIVERSIT ¨ AT MUNChEN, GERMANY, <strong>13</strong> DEPARTMENT Of SURGERY, UNIVERSITY Of hEIDELBERG,<br />

GERMANY<br />

Google goes cancer: Improving outcome prediction for cancer patients by network-based ranking of marker<br />

genes<br />

Predicting the clinical outcome of cancer patients based on the expression of marker genes in their tumors<br />

has received increasing interest in the past decade. Accurate predictors of out¬come and response to<br />

therapy could be used to personalize and thereby improve therapy. However, state of the art methods used<br />

so far often found marker genes with limited predic¬tion accuracy, limited reproducibility, and unclear biological<br />

relevance. To address this problem, we developed a novel computational approach to identify genes<br />

prognostic for outcome that couples gene expression measurements from primary tumor samples with a<br />

network of known relationships between the genes. Our approach ranks genes according to their prognostic<br />

rel¬evance using both expression and network information in a manner similar to Google’s Page-Rank. We<br />

applied this method to gene expression profiles which we obtained from 30 patients with pancreatic cancer,<br />

and identified seven candidate marker genes prognostic for outcome. Compared to genes found with state<br />

of the art methods, such as Pearson correlation of gene expression with survival time, we improve the<br />

prediction accuracy by up to 7%. Accuracies were assessed using support vector machine classifiers and<br />

Monte Carlo cross-validation. We then validated the prognostic value of our seven candidate markers using<br />

immunohistochemistry on an independent set of 412 pancreatic cancer samples. Notably, signatures derived<br />

from our candidate markers were independently predictive of outcome and superior to established clin¬ical<br />

prognostic factors such as grade, tumor size, and nodal status. As the amount of genomic data of individual<br />

tumors grows rapidly, our algorithm meets the need for powerful computa¬tional approaches that are key to<br />

exploit these data for personalized cancer therapies in clinical practice.<br />

abStractS For PoSterS abStractS For PoSterS<br />

MARKO LAAKSO, RIKU LOUhIMO, SAMPSA hAUTANIEMI<br />

COMPUTATIONAL SYSTEMS BIOLOGY LABORATORY, RESEARCh PROGRAMS UNIT, GENOME-SCALE BIOL-<br />

OGY, AND INSTITUTE Of BIOMEDICINE, BIOChEMISTRY AND DEVELOPMENTAL BIOLOGY, UNIVERSITY Of<br />

hELSINKI<br />

Predicting drug targets for individual tumour sample<br />

The identifcation of therapeutic targets is a major challenge in cancer as the pattern of mu¬tations varies<br />

between individual tumours and during their progression. We have addressed this question by focusing on<br />

the known hallmarks of cancer and by tracing them back to the driving genes independently for each patient<br />

sample. We have analysed over 580 breast cancer samples from The Cancer Genome Atlas. The gene expression<br />

profle of each sample was frst converted to a vector of diferentially expressed genes (DEG), which<br />

in turn was transformed to a vector of afected biological processes representing the hallmarks. The DEGs<br />

that were now mapped to the afected processes were next estimated for their therapeutic relevance. We<br />

checked if drugs were already available for them and how likely the restoration of the gene function would<br />

restore the target process.<br />

The computational framework that has been used for the analysis is freely available at http: //csbi.ltdk.<br />

helsinki.fi/moksiskaan/. We believe an automated in silica pipeline, which takes in gene expression data<br />

is an efcient way of predicting therapeutic candidates. Our method considers many-to-many relationships<br />

between drugs, their targets and the processes driven by these targets and is readily applicable for the<br />

combinations of drugs.<br />

54 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 55


JAKOBSEN JS, WAAGE JE, PORSE BT<br />

1BRIC, UNIVERSITY Of COPENhAGEN AND fINSEN LABORATORY, RIGShOSPITALET, OLE MAALøES VEJ 5,<br />

DK¬2200 COPENhAGEN, DENMARK<br />

A C/EBP binary switch: Transcriptional networks behind the dynamic balancing of cell proliferation and<br />

metabolism in liver regeneration<br />

Understanding liver regeneration at a fundamental level may open up new opportunities in regenerative medicine<br />

for this crucial <strong>org</strong>an, for which donation falls dramatically short. During the regenerative process, a<br />

complex network of transcriptional regulators ensures the dynamic balancing between compensatory growth<br />

and metabolic functions. Central among the regulators are the bZIP transcription factors (TFs) C/EBPᴅ and<br />

C/EBPᴅ. C/ebpᴅ is essential for hepatocyte differentiation and correct expression of key metabolic enzymes,<br />

while C/ebpᴅ is required for a full mitogenic response to liver injury. However, the precise and global<br />

role of these factors in liver regeneration has not been comprehensively mapped on a genomic scale so far.<br />

Here, we employ a time course of chromatin-immunoprecipitation followed by deep sequencing (ChIP-seq)<br />

to map the binding of C/EBPᴅ, C/EBPᴅ and RNA-POL2 in the mouse model. Noticeably, the protein level<br />

ratios of C/EBPᴅ and C/EBPᴅ are precisely reflected in global binding patterns. Two major ‘timed’ binding<br />

patterns are clearly associated with expression of distinct sets of genes, related to either metabolic function<br />

or cell cycle. Using available databases (Uniprobe, Transfac and Jaspar) we generated an extensive list of<br />

TFs potentially involved in either the metabolic or the proliferative phase of liver regeneration. We identify<br />

distinct panels of feed-forward loops associated with C/EBP patterns involving TFs such as circadian clock<br />

regulators, E2fs, Klf-factors, Lxr/Fxrs, Fox-members. A follow-up ChIP-seq examination of EGR1¬binding<br />

surprisingly showed overlap with the C/EBP pattern under¬represented in EGR1¬binding sequence.<br />

Using mice lacking C/ebpᴅ, we demonstrate two binding modes of EGR1 in vivo; direct to its cognate DNA<br />

sequence or assisted/indirect via C/EBPᴅ. This suggests a global scale model for differential regulation of<br />

EGR1¬target genes: with or without dependency on C/EBPs for metabolic or cell cycle regulated genes,<br />

respectively.<br />

In summary, our study shows how ChIP-seq and genomics analyses can be used on a mammalian in vivo<br />

system to elucidate dynamic transcriptional networks required for appropriate physiological gene regulation,<br />

exemplified by the fine-tuned balancing of cell proliferation and homeostasis in liver regeneration.<br />

abStractS For PoSterS abStractS For PoSterS<br />

ANDREI zINOVYEV123, INNA KUPERSTEIN123, DAVID COhEN123, SIMON fOURqUET123, LAURENCE<br />

CALzONE123, STUART POOK4, PAOLA VERA-LICONA123, ERIC BONNET123, DANIEL ROVERA123, EM-<br />

MANUEL BARILLOT123<br />

(1) INSTITUT CURIE, 26 RUE D’ULM, f-75248 PARIS fRANCE, (2) INSERM, U900, PARIS, f-75248<br />

fRANCE, (3) MINES PARISTECh, fONTAINEBLEAU, f-77300 fRANCE, (4) SYSRA, f-75248 PARIS fRANCE<br />

Towards of Atlas of Cancer Signaling Networks<br />

Basis for the Institut Curie systems biology platform for data analysis and interpretation<br />

Cancer is a complex disorder that can be seen as a systems biology disease. There are numerous cell<br />

signaling mechanisms that are dysregulated in cancer. To understand involvement of different mechanisms<br />

in the disease, systematic representation and analysis of the processes are needed. To achieve the goal,<br />

we are currently in the process of creating the Atlas of Cancer Signaling Networks (ACSN), where signaling<br />

mechanisms are represented as comprehensive maps amenable for computational and mathematical analysis<br />

(http://acsn.curie.fr). Currently ACSN consists of four maps: cell-cycle regulation by RB-E2F, DNA repair,<br />

Cell Cycle and checkpoints, Apoptosis and energy metabolism and Cell Survival signaling networks. We<br />

will include in ACSN additional maps for Epithelia-Mesenchimal Transition (EMT), Telomeres maintenance,<br />

Centrosome maintenance, DNA replication and Inflammatory processes. We have developed a Google<br />

Map-based tool NaviCell (http://navicell.curie.fr) for exploring large signaling networks. The tool is characterized<br />

by the unique combination of three essential features: (1) map navigation based on Google Maps<br />

engine, (2) semantic zooming for viewing different levels of details of the map and (3) integrated web-based<br />

blog for collecting the community curation feedbacks. NaviCell facilitates curation of molecular interactions<br />

maps by the community helping to update and maintain maps in an interactive and user-friendly fashion. We<br />

have developed a series of tools for network analysis (BiNoM, OCSANA, etc), which enable structural analysis,<br />

target identification and integration and analysis of high-throughput data using ACSN maps.<br />

56 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 57


fLORIAN MARTIN1; TY M. ThOMSON2; ALAIN SEWER1; DAVID A. DRUBIN2; CAROLE MAThIS1; DExTER<br />

PRATT2; JULIA hOENG1; MANUEL C. PEITSCh1<br />

1PhILIP MORRIS INTERNATIONAL R&D, PhILIP MORRIS PRODUCTS S.A., NEUChâTEL, SWITzERLAND<br />

2SELVENTA, ONE ALEWIfE CENTER, CAMBRIDGE, MA, USA<br />

Assessment of Network Perturbation Amplitude by Applying High-Throughput Data to Causal Biological<br />

Networks<br />

BACKGROUND: High-throughput technologies have the potential to elucidate the biological impact of<br />

disease, drug treatment, and environmental agents on humans. An ongoing challenge has been the analysis<br />

of the generated data to more accurately characterize the perturbed biological processes at the mechanistic<br />

level. Here, a new approach was taken, which uses prior knowledge of cause and effect relationships<br />

structured into biological network models describing specific processes, such as inflammatory signaling or<br />

cell cycle progression.<br />

RESULTS: Four complementary methods and their companion statistics were devised to quantify treatmentinduced<br />

activity changes in processes described by network models. This approach is called Network<br />

Perturbation Amplitude (NPA) scoring because the amplitudes of treatment-induced perturbations are computed<br />

for biological network models. The NPA methods were tested on two transcriptomic data sets: normal<br />

human bronchial epithelial (NHBE) cells treated with the pro-inflammatory signaling mediator TNFᴅ, and<br />

HCT<strong>11</strong>6 colon cancer cells treated with the CDK cell cycle inhibitor R547. Each data set was scored against<br />

network models representing different aspects of inflammatory signaling and cell cycle progression. The<br />

NPA scores successfully quantified the amplitude of the TNFᴅ-induced perturbations in treated NHBE cells,<br />

as confirmed by NF-ᴅB nuclear localization measurements. In HCT<strong>11</strong>6 cells, the degree and specificity to<br />

which CDK-inhibition affected cell cycle and inflammatory signaling were also meaningfully determined by<br />

the NPA results.<br />

CONCLUSION: The NPA scoring method leverages high-throughput measurements and a priori knowledge<br />

in the form of causal network models to characterize the activity change for a broad collection of biological<br />

processes. Applications of this framework include quantitative assessment of the biological impact caused<br />

by drug treatments, environmental factors, or toxic substances.<br />

abStractS For PoSterS abStractS For PoSterS<br />

ALESSANDRO ORI1, AMPARO ANDRES-PONS1, NICCOLò BANTERLE1, MURAT ISKAR1, CLAUDIA ES-<br />

ChER2, OLIVER RINNER2, PEER BORK1, EDWARD A. LEMKE1 AND MARTIN BECK1<br />

1 STRUCTURAL AND COMPUTATIONAL BIOLOGY UNIT, EUROPEAN MOLECULAR BIOLOGY LABORATORY,<br />

hEIDELBERG, GERMANY. 2 BIOGNOSYS AG, SChLIEREN, SWITzERLAND.<br />

Compositional remodelling of the nuclear pore complex<br />

Structure determination of nuclear pores complexes (NPCs) turned out to be challenging due to their positioning<br />

in a membranous environment, their sheer size and their intricate composition. In order to generate<br />

structural models of the human NPC, data obtained by several different structure determination techniques<br />

have to be integrated into a common framework. To achieve this goal, the knowledge of human nucleoporin<br />

stoichiometry is critical. We have absolutely quantified human nucleoporins using targeted proteomics in<br />

combination with a heavy labeled reference peptide strategy. We used selected reaction monitoring (SRM)<br />

to establish multiple independent measurements per protein across several biological replicates and different<br />

biological states which allowed us to accurately define the absolute composition of fully assembled<br />

NPCs. Our data demonstrate that nucleoporin abundance is structured in discreet steps reflecting the<br />

geometrical <strong>org</strong>anization of subcomplexes into NPC scaffold structure. The integration of these data with<br />

complementary techniques including fluorescence and electron microscopy enable deciphering the compositional<br />

inventory of the human NPC and thus provide crucial information for the generation of structural<br />

models at atomic resolution. We have furthermore absolutely quantified NPC composition in different human<br />

cell lines and identified several nucleoporins that are differentially incorporated into the NPC. Our data imply<br />

that the human nuclear pore is remodeled in a cell-type specific manner.<br />

58 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 59


ALI ALTINTAS1, ChRISTOPhER WORKMAN1<br />

1TEChNICAL UNIVERSITY Of DENMARK (DTU), DEPARTMENT Of SYSTEMS BIOLOGY, CENTER fOR BIO-<br />

LOGICAL SEqUENCE ANALYSIS (CBS)<br />

Growth Physiology and Time Dependent Metabolic Remodelling of Saccharomyces cerevisiae under Oxidative<br />

Stress<br />

Oxidative stress caused by an excess of reactive oxygen species (ROS), is known to damage cellular<br />

components and triggers a number of signalling pathways that contribute to a remodelling of transcription,<br />

translation and metabolism. ROS are produced during normal aerobic physiological processes or when<br />

cells are exposed to oxidizing agents such as hydrogen peroxide (HP). Cells have to sense ROS to trigger<br />

a response against its harmful effects. Protein kinases (PK) and protein phosphatases (PP) regulate this<br />

sense-response switch to create adaptation to oxidative stress.<br />

The objective of the project was to investigate the time-dependent growth effects of oxidizing environments<br />

on PK and PP deletion mutants of budding yeast. To do this, 38 different PK and PP mutants and two<br />

wild-type strains of S. cerevisiae were investigated for oxidative stress responses. These 38 mutants were<br />

selected for their known activities in both general/oxidative stress response and DNA damage response. HP<br />

was used to create an oxidizing environment at a number of different concentrations (0.25, 0.50, 1.0 and 2.0<br />

mM) ranging from mild to moderate stress. Growth physiology and the time dynamics of the stress response<br />

were characterized in a microfermentation system (m2p BioLector) for all deletion strains.<br />

The microfermentation platform allowed us to measure biomass with high time resolution (3 minute sample<br />

rate). Important metabolites of carbon metabolism were measured by HPLC before, during and after the<br />

diauxic shift (DS) of stressed and un¬stressed S. cerevisiae. The metabolic delay of DS after stress provided<br />

a quantitative measure of strain sensitivity. Measured DS delays, due to growth arrest and/or a decrease<br />

in the metabolic activity with increasing HP, allow for the detection of phenotypic variation in mild stress<br />

conditions.<br />

The most important growth physiology findings relate to the opposing mechanism of PK and PP, the central<br />

activity of OCA1 gene among responses of oxidative stress, and also the investigation of slow growing mutants:<br />

bud32∆, vps15∆, ptc1∆ and oca1∆.<br />

abStractS For PoSterS abStractS For PoSterS<br />

AGNIESzKA SzWAJDA1, LEENA KARhINEN1, SAWAN KUMAR JhA1, TEA PEMOVSKA1, BhAGWAN YA-<br />

DAV1, KRISTER WENNERBERG1, TERO AITTOKALLIO1<br />

1 INSTITUTE fOR MOLECULAR MEDICINE fINLAND, fIMM, UNIVERSITY Of hELSINKI, hELSINKI, fINLAND<br />

Identification of molecular drivers in breast cancer using kinome-wide drug-target network and drug<br />

sensitivity screening<br />

To systematically study how kinase inhibitors and their cellular targets function together as networks, we<br />

integrated three recent large-scale studies of kinase inhibitor specificities1-3, along with drug binding<br />

information from the ChEMBL database and individual experiments, into a quantitative data matrix for a<br />

total of 1679 chemical compounds and 445 kinase targets. When using relatively stringent thresholds for<br />

biochemical drug inhibition levels (drug dissociation constant and IC50 34% kinase activity<br />

inhibition at 0.5µM), the resulting drug-target network (26416 interactions between 1464 drugs and 444<br />

kinases) exhibited approximately a scale-free property with a degree distribution ( ) . Drugs with highly<br />

correlated target profiles (Spearman correlation > 0.5) were then linked to construct a drug-drug network.<br />

The interconnected drugs turned out to have similar chemical structure and mode of action, demonstrating<br />

that such global drug networks could be used to reveal mechanisms of drug action or suggest effective drug<br />

combinations.<br />

In a specific case study, we elucidated the signaling pathways driving 10 breast cancer cell lines by correlating<br />

cellular viability inhibition measurements (EC50 values) of 35 kinase inhibitors with the biochemical drug<br />

inhibition information from the drug-target network. The analysis was based on the assumption that kinase<br />

inhibitors targeting the essential kinases should effectively decrease cell numbers. As a result, we obtained<br />

a ranking of kinases according to their likelihood of being the key molecular drivers in each cell line. The<br />

ERBB2/HER2 receptor tyrosine kinase, a well-known oncogenic driver in a subset of breast cancers, served<br />

as a positive control and was found among the top hits in cell lines known to be driven by this kinase, but not<br />

in others. Other breast cancer- related kinases, such as PTK6/BRK or members of the PI3K pathway, were<br />

also highly ranked in relevant cell lines. The significance of the predicted driver kinases for cancer progression<br />

will be experimentally tested with siRNA-mediated knockdowns and other kinase inhibitors or their<br />

combinations.<br />

References:<br />

1. Davis et al., “Comprehensive analysis of kinase inhibitor selectivity”, Nat Biotechnol. 20<strong>11</strong>; 29:1046-1051.<br />

2. Metz et al., “Navigating the kinome”, Nat Chem Biol. 20<strong>11</strong>; 7:200-202.<br />

3. Anastassiadis et al., “Comprehensive assay of kinase catalytic activity reveals features of kinase inhibitor<br />

selectivity”, Nat Biotechnol. 20<strong>11</strong>; 29:1039-1045.<br />

60 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 61


NAThANIEL STANLEY, GIANNI DE fABRITIIS<br />

UNIVERSITAT POMPEU fABRA (UPf), RESEARCh UNIT ON BIOMEDICAL INfORMATICS (GRIB), PARC DE<br />

RECERCA BIOMèDICA DE BARCELONA (PRBB), DR. AIGUADER 88, 08003 BARCELONA, SPAIN<br />

Kinetic, Thermodynamic, and Conformational Characterization of Biological Network Components Using<br />

Molecular Dynamics: Towards better understanding and better drugs<br />

The full description of the kinetics and thermodynamics of interacting components in a biological system are<br />

critically important for us to accurately represent and understand them. With recent advancements in both<br />

methodology and hardware, molecular dynamics simulations are at a point where they can now be used<br />

to accurately calculate such data for those components where traditional experimental techniques prove<br />

inadequate or prohibitive. In addition, conformational changes in the structure of interacting molecules can<br />

be assessed to high degree of precision. Finally, target conformations can be subjected to high throughput<br />

fragment docking in order to build novel inhibitors and allosteric modulators. We present our work in this vein<br />

done thus far on multiple components of the endocannabinoid system.<br />

abStractS For PoSterS abStractS For PoSterS<br />

LUISIER R, LEMPIÄINEN h, MUELLER A, hAhNE f, UNTERBERGER E, BOLOGNANI f, BALWIERz P, BRAE-<br />

UNING A, DUBOST V, GOODMAN J, ChIBOUT SD, ThEIL D, hEARD D, MOULIN P ,GRENET O, SChWARz<br />

M, MOGGS J, VAN NIMWEGEN E, TERRANOVA R<br />

Prediction of early regulatory networks underlying liver non-genotoxic carcinogenesis upon xenobiotic<br />

exposure.<br />

Non-genotoxic carcinogenesis (NGC) is a common drug-induced toxicity that occurs in rodent models, for<br />

which no well-validated short-term assays exist. Here we have used genome-wide transcriptome profiling<br />

and computational modeling to investigate the temporal sequence of events and identify potential gene regulatory<br />

networks involved in NGC using a well-characterized rodent model for liver tumor promotion.<br />

Singular value decomposition was used to identify and quantify early dynamics of gene expression changes<br />

in mouse liver upon exposure to the non-genotoxic carcinogen phenobarbital (PB). Applying Motif Activity<br />

Response Analysis (MARA), which models gene expression dynamics in terms of predicted cis-regulatory<br />

sites, led to the identification of transcription factors perturbed upon PB treatment and potentially responsible<br />

for specific kinetic responses to treatment. Genetic analyses revealed the relation to cancer-relevant<br />

pathways of candidate NGC biomarkers and enabled the disentangling of tumor promotion pathways from<br />

the xenobiotic response.<br />

Altogether our results indicate that in vivo gene expression changes upon PB exposure are not linearly<br />

dependent on time, consistent with possible changes in tissue composition over time. Gene regulatory<br />

networks underlying tumor promotion upon PB are not fully “active” at early stages of tumor promotion,<br />

“inactive” in tumor-resistant mouse models and “active” in promoted end-tumors.<br />

Our data highlight novel potential early mechanisms and pathways for liver tumor promotion, providing new<br />

opportunities for assessing the carcinogenic potential of environmental cues including novel therapeutics.<br />

62 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 63


ROSSUKON ThONGWIChIAN 1*, hONOR ROSE 1*, fRANCOIS-xAVIER ThEILLET 1, JONAS KOSTEN1, AL-<br />

ExANDER DOSE 2, UWE BENARY 3, DIRK SChWARzER 2, JANA WOLf 3, PhILIPP SELENKO 1.<br />

1DEPARTMENT Of NMR-ASSISTED STRUCTURAL BIOLOGY IN-CELL NMR GROUP AND 2DEPARTMENT Of<br />

ChEMICAL BIOLOGY, LEIBNIz INSTITUTE fOR MOLECULAR PhARMACOLOGY (fMP), BERLIN, GERMANY.<br />

3MAThEMATICAL MODELING Of CELLULAR PROCESS, MAx DELBRUECK CENTER fOR MOLECULAR MEDI-<br />

CINE (MDC), BERLIN, GERMANY. ROSE@fMP-BERLIN.DE * EqUAL CONTRIBUTION.<br />

Systems level profiling of cellular kinase activities by multiplexed NMR spectroscopy.<br />

Complex kinase activities lie at the heart of most eukaryotic signaling networks. Here we introduce a novel<br />

concept to directly monitor cellular kinase activities by high-resolution NMR spectroscopy. Using isotopelabeled<br />

Kinase Activity Reporters (KARs) and multiplexed NMR readouts, we follow the activities of up to 15<br />

different, cellular kinases in parallel, and within a single set of time-resolved NMR experiments. Quantitative<br />

in situ monitoring of reversible kinase and phosphatase activities-, in connection with multiplexed kinase<br />

inhibitor screening-, enables high-accuracy mathematical modeling of cellular signal response behaviors,<br />

which, in turn, can be directly applied to network medicine approaches.<br />

abStractS For PoSterS abStractS For PoSterS<br />

Negative Feedback is a Subordinated Control Structure in Signal Transduction<br />

Protein signaling and metabolic systems often contain multiple feedback interactions which are a prominent<br />

motif of regulation. We hypothesized that feedback interactions influence concentration range, timing, signal<br />

transmission strength and the appearance of transients in a predictable and computable manner. In steadystate,<br />

feedback interactions are resolved, i.e. cyclic structures can be eliminated and subsumed by pure<br />

input-output relations. We use this fact in order to systematically analyze the effect of a multiple negative<br />

feedback structure (figure). We employ a tool for steady-state analysis that operates over input ranges and<br />

calculates systemic delays ([1]).<br />

We find that negative feedback universally shortens delays and limits concentration ranges. Furthermore,<br />

it linearizes signal transmission, increases the appearance of transients, but also shortens their decay<br />

times. In other words, the directed path of signal transduction from receptor to cytosolic and nuclear targets<br />

is adorned by negative feedback interactions, such that signal transduction becomes better controlled in<br />

timing, concentration and by reduction of nonlinearity. An additional property, the increased appearance of<br />

transients, shows that below the time resolution of steady-state analysis, feedback has an additional role<br />

in providing for fast interactions. However, in steady-state analysis, negative feedback has a subordinated<br />

control function compared to a primary role of signal transduction along pathways.<br />

[1] Scheler, G.: Transfer Functions in Signal Transduction: Applying a Protein Signaling Function (PSF) to a<br />

Model of Striatal Neural Plasticity. Plos Comp Biol, submitted.<br />

64 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 65


SEBASTIAN STUDENT, MAGDALENA SKONIECzNA, JOANNA RzESzOWSKA<br />

BIOSYSTEMS GROUP, INSTITUTE Of AUTOMATIC CONTROL, SILESIAN UNIVERSITY Of TEChNOLOGY, 44-<br />

100 GLIWICE, POLAND<br />

Osteopontin expression in human colon cancer cells<br />

Osteopontin is a phosphorylated glycoprotein which plays a crucial role in many normal physiological as<br />

well as pathological cellular processes. It was originally identified as a important factor in bone remodeling,<br />

cell adhesion, migration and cell survival, activation, chemotaxis and apoptosis. Osteopontin is an important<br />

factor in the progression of several cancer types and in various aspect of metastasis. Regulation of the<br />

osteopontin gene is only incompletely understood, and cell types may differ in the regulatory mechanisms of<br />

the gene and the roles of osteopontin.<br />

In this study we concentrated on identifying the relationships between osteopontin and the apoptosis signaling<br />

pathway in a model of colon cancer. We used different oligonucleotide microarray datasets to study the<br />

potential role of osteopontin, mainly in the p53-dependent apoptosis signaling pathway. Experiments were<br />

conducted with the Affymetrix platform on HCT <strong>11</strong>6 wild-type and p53-mutated colon cancer cell lines. The<br />

cells were irradiated with 4 Gy of ionizing radiation, and non-irradiated cells were used as a control group.<br />

Ionizing radiation induces the production of reactive oxygen species which play an important role in apoptotic<br />

cell death, and by inducing DNA damage activates the p53-dependent apoptosis signaling pathway which<br />

can cause cell cycle arrest and apoptosis. Not all interactions between apoptosis-related genes that are<br />

transcriptionally regulated by p53 have been identified.<br />

The computational analysis was carried out using the PLS (partial least squares)-based gene selection<br />

method, which enables assignment of biological meanings for the genes with the highest weights in the PLS<br />

model. The PLS method, in contrast to the PCA (principal component analysis) criterion based on maximization<br />

of the variance of a linear combination of genes, extracts components by maximizing the sample<br />

covariance between the class variable and a linear combination of genes. The information for genes included<br />

in components described by PLS can be directly related to the biological meaning by this analysis. The<br />

relationship between the expression level of the osteopontin gene and p53 was confirmed by real time PCR<br />

experiments on the same cell lines. The list of genes involved in apoptosis signaling pathways was prepared<br />

based on KEGG, Biocarta and other published apoptosis signal interaction models.<br />

Our results show that the osteopontin expression level depends strongly on the p53 status of colon cancer<br />

cells, and is lower in the HCT<strong>11</strong>6 p53-mutated cell line. We also observed an inverse correlation with expression<br />

of the PTEN gene, which has a growth suppressive effect through its inhibition of the phosphatidylinositol<br />

3-kinase (PI3K) pathway. It is known that this pathway is involved in promoting cell growth, survival<br />

and tumorigenesis when overstimulated. Our results suggest that osteopontin plays an important role in<br />

PI3K regulation, and in this way can affect the apoptosis pathway. This analysis is the first step in the investigation<br />

of osteopontin’s role in the apoptosis pathway, which will require more detailed investigation.<br />

This work was supported by grants No. NN5144<strong>11</strong>936 and N N518497639 from MNiSW and ZIS ¬BK/ 274<br />

/20<strong>11</strong> t.<strong>13</strong> from Silesian University of Technology in Gliwice.<br />

abStractS For PoSterS abStractS For PoSterS<br />

ANNA hEGELE 1,*, ATANAS KAMBUROV 1,*, ARNDT GROSSMANN 1, ChRYSOVALANTIS SOURLIS 1, SYL-<br />

VIA WOWRO 1, MAREIKE WEIMANN 1, CINDY L. WILL 2, VLAD PENA 2, REINhARD LühRMANN 2, ULRICh<br />

STELzL 1,#<br />

1 MAx-PLANCK INSTITUTE fOR MOLECULAR GENETICS (MPI-MG), OTTO-WARBURG LABORATORY, BER-<br />

LIN, GERMANY. AND 2 MAx-PLANCK INSTITUTE Of BIOPhYSICAL ChEMISTRY (MPI-BPC), DEPARTMENT<br />

Of CELLULAR BIOChEMISTRY, GöTTINGEN, GERMANY.<br />

Dynamic protein-protein interaction wiring of the human spliceosome<br />

Pre-mRNA splicing is catalyzed by the spliceosome, a highly complex, dynamic and protein rich ribonucleoprotein<br />

complex (RNP) that assembles de novo on each intron to be spliced. During spliceosome assembly,<br />

activation, catalysis and disassembly, defined large RNP complexes are formed in an ordered, stepwise<br />

manner. More than 200 proteins copurify at one or more stages with human spliceosomes assembled on<br />

prototype pre-mRNAs in cellular extracts. To better understand protein -protein interactions governing splicing,<br />

we systematically investigated interactions between human spliceosomal proteins. A comprehensive<br />

Y2H interaction matrix screen generated a protein interaction map comprising 632 interactions between 196<br />

proteins. 242 interactions were found between spliceosomal core proteins, and largely validated by co-immunoprecipitation.<br />

To reveal dynamic changes in protein interactions, we integrated spliceosomal complex<br />

purification information with our interaction data and performed link clustering. These data, together with<br />

interaction competition experiments, suggest that during step 1 of splicing, hPRP8 interactions with SF3b<br />

proteins are replaced by hSLU7, positioning this second step factor close to the active site, and that the<br />

DEAH-box helicases hPRP2 and hPRP16 cooperate through ordered interactions with GPKOW. Our data<br />

provide extensive information about the spliceosomal protein interaction network and its dynamics.<br />

Reference:<br />

Hegele et al., Mol Cell. <strong>2012</strong> Feb 24;45(4):567-80.<br />

66 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 67


CAMILLE TERfVE & JULIO SAEz-RODRIGUEz, EMBL-EBI, WELLCOME TRUST GENOME CAMPUS, CAM-<br />

BRIDGE, CB10 1SD, U.K.<br />

Logic modeling of signalling networks with high-content phosphoproteomic data<br />

Cells are continuously exposed to signals, which need to be received, interpreted and propagated in an integrated<br />

manner to produce the appropriate response. This information processing is performed through the<br />

use of highly dynamic and context specific networks whose deregulation is often involved in the development<br />

of diseases. Mathematical modeling is a very useful tool to make sense of this complexity. Many different<br />

formalisms can be applied depending on the type of data available and the question to be answered.<br />

Logic models are a flexible yet computationally simple way to approach this problem. Cause-effect relationships<br />

in biological pathways can often be found in the literature, but rarely include specific gates, nor cell-type<br />

specific information. An approach to provide predictive power and context-specificity models was introduced<br />

in [1], by using perturbation data to train a Boolean model from a generic prior knowledge network. Here we<br />

present CellNOptR, and R/Bioconductor package that implements this method and extends it including a<br />

palette of logic formalisms that handle both quantitative and time-dependent aspects with various levels of<br />

complexity.<br />

We also using this as a framework to investigate the challenges and opportunities [2] associated with using<br />

mass spectrometry proteomics as a primary data collection technology to build models of signalling networks.<br />

References<br />

[1] J. Saez-Rodriguez, L. G. Alexopoulos, J. Epperlein, R. Samaga, D. A. Lauffenburger,<br />

S. Klamt, and P. K. S<strong>org</strong>er. Discrete logic modelling as a means to link protein signalling networks with<br />

functional analysis of mammalian signal transduction. Molecular Systems Biology, 5:331, 2009. PMID:<br />

19953085.<br />

[2] C. Terfve and J. Saez-Rodriguez. Modeling signaling networks using high-throughput phospho-proteomics.<br />

Advances in Experimental Medicine and Biology, 736:19–57, <strong>2012</strong>. PMID: 2216<strong>13</strong>21.<br />

abStractS For PoSterS abStractS For PoSterS<br />

ThOMAS SChLITT1, NIKOLAOS BARKAS1, ChRISTOPhER TEBBE2, fRAUKE SPRENGEL2, VOLKER AhL-<br />

ERS2, BENJAMIN LEhNE1<br />

1DEPARTMENT Of MEDICAL AND MOLECULAR GENETICS, SChOOL Of MEDICINE, KING’S COLLEGE LON-<br />

DON, GUY’S hOSPITAL, LONDON SE1 9RT, UNITED KINGDOM<br />

2DEPARTMENT Of COMPUTER SCIENCE, fACULTY IV, UNIVERSITY Of APPLIED SCIENCES AND ARTS hAN-<br />

NOVER, P.O. BOx 920251, 30441 hANNOVER, GERMANY<br />

De-novo pathway discovery for genes linked to complex diseases<br />

Genome-wide association studies (GWAS) proved successful in the identification of sequence variants<br />

associated with complex diseases. However, the necessity to apply very stringent thresholds to ensure genome-wide<br />

significance suggests many disease associated genes might be missed. We developed a novel<br />

approach to support the discovery of genes and pathways underlying complex diseases that is independent<br />

of previous pathway annotation. Based on data from GWAS we derive gene-specific p-values by mapping<br />

SNPs to genes controlling for confounding effects such as the number of SNPs per gene. We use additional<br />

biological information in the form of protein or gene networks to identify additional genes that might be associated<br />

with disease. Our hypothesis is that genes truly linked to the disease under study (true-positives) are<br />

localised in proximity in gene networks while genes not linked to the disease (false-positives) will be scattered<br />

randomly over the network. Our novel algorithm -Region Growing Analysis (RGA) -maps gene-specific<br />

p-values to molecular gene networks and identifies regions enriched for genes with association signal.<br />

These regions could span several densely connected subnetworks (modules or cliques) and do not have to<br />

be identical to known pathways. We demonstrate the utility of our RGA algorithm by applying it to proteinprotein<br />

interaction networks to identify regions associated with Crohn’s disease and Type-1¬diabetes. We<br />

are able to identify several candidate disease genes in addition to the genes identified in the GWAS studies<br />

we used to obtain the gene-specific p-values; many of these candidate genes have been confirmed by larger<br />

meta-analysis and functional studies, others we are currently validating experimentally.<br />

68 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 69


abStractS For PoSterS<br />

ANDREI zINOVYEV1,2,3*, INNA KUPERSTEIN1,2,3, EMMANUEL BARILLOT1,2,3 , AND WOLf-DIETRICh<br />

hEYER4<br />

1INSTITUT CURIE, 26 RUE D’ULM, f-75248 PARIS fRANCE<br />

2INSERM, U900, PARIS, f-75248 fRANCE<br />

3MINES PARISTECh, fONTAINEBLEAU, f-77300 fRANCE<br />

4DEPARTMENTS Of MICROBIOLOGY AND Of MOLECULAR AND CELLULAR BIOLOGY, UNIVERSITY Of CALIfORNIA,<br />

DAVIS, ONE ShIELDS AVENUE, DAVIS, CA 95616-8665<br />

Synthetic lethality within one pathway and cancer treatment<br />

A synthetic lethal interaction is usually stated when defects in two non-essential genes cause cell death.<br />

Synthetic lethality and synthetic dosage lethality studies in model <strong>org</strong>anisms and human cells give hope<br />

to develop cancer drugs that would kill cancer cells very selectively: if a cancer cell has a characteristic<br />

deletion or amplification of a gene, then inhibiting or overexpressing another nonessential gene forming a<br />

synthetic interaction pair, will lead to specific lethality of cancer cells. For example, some breast cancers are<br />

characterized by loss-of-function mutation in BRCA1 gene involved in DNA repair. The PARP1 gene forms a<br />

synthetic lethal pair with BRCA1 in cellular models and, therefore, inhibitors of PARP1 for treating BRCA1deficient<br />

breast cancer were developed and went to clinical trials.<br />

The genes from a synthetic interaction pair are generally assumed functioning in two parallel and mutually<br />

compensatory pathways (multi-pathway Synthetic Lethality). However, several examples of synthetic lethal<br />

relationships involving genes implicated in the homologous recombination DNA repair pathway extend this<br />

paradigm. In this situation defects in two genes which function in the same pathway lead to cell death (single-pathway<br />

Synthetic Lethality). We explored the inherent system properties of such a genetic relationship<br />

using mathematical modeling. We found that three circumstances are pre-requisites for the single-pathway<br />

Synthetic Lethality scenario: reversibility of pathway steps, presence of a compensatory pathway and toxicity<br />

of at least one pathway intermediate. Further modeling revealed the potential contribution of synthetic dosage<br />

lethal interactions in such a genetic system.<br />

We discuss implications of single-pathway synthetic lethality on cancer treatment modalities acting through<br />

DNA damage.<br />

SPonSorS<br />

PLATINUM LEVEL<br />

PREMIUM LEVEL<br />

PARTNER LEVEL<br />

SUPPORTER LEVEL<br />

PROMOTIONAL LEVEL<br />

70 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 71


Ruedi Aebersold<br />

aebersold@imsb.biol.ethz.ch<br />

ETH Zürich<br />

Zürich<br />

CH<br />

Hassan Ahmed<br />

hahmed@eisbm.<strong>org</strong><br />

EISBM<br />

Massy, France<br />

Ali Altintas<br />

ali@cbs.dtu.dk<br />

Center for Biological Sequence Analysis<br />

(CBS)<br />

Department of Systems Biology<br />

Technical University of <strong>Denmark</strong><br />

(DTU)<br />

Lyngby, <strong>Denmark</strong><br />

Emmanuel BARILLOT<br />

Emmanuel.Barillot@curie.fr<br />

Institut Curie<br />

PARIS, FRANCE<br />

Christopher Barnes<br />

barnes@imsb.biol.ethz.ch<br />

ETH Zürich<br />

Copenhagen, Switzerland<br />

Tilo Beyer<br />

tilo.beyer@med.ovgu.de<br />

Otto-von-Guericke University, Institute<br />

of Molecular and Clinical Immunology<br />

Magdeburg, Germany<br />

Willy Björklund<br />

willy.bjorklund@thermofisher.com<br />

Thermo Fisher Scientific<br />

Kungens Kurva, Sverige<br />

Blagoy Blagoev<br />

bab@bmb.sdu.dk<br />

SDU<br />

Odense, <strong>Denmark</strong><br />

Nikolaj Blom<br />

blom@cbs.dtu.dk<br />

DTU - Technical University of <strong>Denmark</strong><br />

Kongens Lyngby, <strong>Denmark</strong><br />

Blake B<strong>org</strong>eson<br />

b<strong>org</strong>eson@utexas.edu<br />

University of Texas at Austin<br />

Austin, US<br />

Susanne Brix<br />

sbp@cbs.dtu.dk<br />

DTU Systems Biology<br />

Kgs. Lyngby, <strong>Denmark</strong><br />

Søren Brunak<br />

brunak@cbs.dtu.dk<br />

Center for Biological Sequence<br />

Analysis<br />

Kgs. Lyngby, <strong>Denmark</strong><br />

Andrea Califano<br />

califano@c2b2.columbia.edu<br />

Columbia University<br />

New York, MA, USA<br />

Gianni Cesareni<br />

cesareni@uniroma2.it<br />

University of Rome Tor Vergata<br />

Roma, Italy<br />

Joan Chang<br />

joan.chang@icr.ac.uk<br />

Institute of Cancer Research<br />

London, UK<br />

Sara Holm Christiansen<br />

sara@intomics.com<br />

Intomics<br />

Lyngby DK, <strong>Denmark</strong><br />

Morten Colding-Jørgensen<br />

mcj@novonordisk.com<br />

Novo Nordisk A/S<br />

Søb<strong>org</strong> DK, <strong>Denmark</strong><br />

Thomas Cox<br />

coxthomasr@gmail.com<br />

Biotech Research & Innovation Centre<br />

(BRIC)<br />

Copenhagen, <strong>Denmark</strong><br />

Pau Creixell<br />

creixell@cbs.dtu.dk<br />

Cellular Signal Integration Group (C-<br />

SIG) - Center for Biological Sequence<br />

Analysis (CBS) - Department of Systems<br />

Biology - Technical University<br />

of <strong>Denmark</strong> (DTU)<br />

Lyngby, <strong>Denmark</strong><br />

Simone Daminelli<br />

simone.daminelli@biotec.tu-dresden.<br />

de, Biotechnology Center, Technische<br />

Universitet Dresden<br />

Dresden, Germany<br />

Federico De Masi<br />

fdemasi@cbs.dtu.dk<br />

CBS - DTU<br />

Kgs. Lyngby, <strong>Denmark</strong><br />

ParticiPantS ParticiPantS<br />

Sol Efroni<br />

sol.efroni@biu.ac.il<br />

Bar Ilan University<br />

Ramat Gan, Israel<br />

Janine Erler<br />

Janine.erler@bric.ku.dk<br />

BRIC<br />

Copenhagen, <strong>Denmark</strong><br />

Stephan Feller<br />

stephan.feller@imm.ox.ac.uk<br />

WIMM, Oxford University<br />

Oxford, United Kingdom<br />

Mogens Fenger<br />

mogens.fenger@hvh.regionh.dk<br />

Hvidovre Univeristy Hospital<br />

Hvidovre, <strong>Denmark</strong><br />

Robin Friedman<br />

robin.friedman@gmail.com<br />

Institut Pasteur<br />

Paris, France<br />

Krzysztof Fujarewicz<br />

krzysztof.fujarewicz@polsl.pl<br />

Silesian University of Technology<br />

Gliwice, Poland<br />

Adam Galuszka<br />

adam.galuszka@polsl.pl<br />

Silesian University of Technology<br />

Gliwice, Poland<br />

Christian Garde<br />

garde@cbs.dtu.dk<br />

Center for Biologcal Sequence Analysis<br />

(CBS), Department of Systems<br />

Biology, Technical University of<br />

<strong>Denmark</strong> (DTU)<br />

Lyngby, <strong>Denmark</strong><br />

Anne Claude Gavin<br />

gavin@embl.de<br />

EMBL-Heidelberg<br />

Heidelberg, Germany<br />

Pier Federico Gherardini<br />

federico.gherardini@gmail.com<br />

University of Rome Tor Vergata<br />

Rome, Italy<br />

Apurv Goel<br />

a.goel@student.unsw.edu.au<br />

University of New South Wales<br />

Sydney, Australia<br />

Eng Lim Goh<br />

englim@sgi.com<br />

SGI<br />

USA<br />

Alexey Golstov<br />

a.goltsov@abertay.ac.uk<br />

Abertay University<br />

Dundee DD1 1HG, SCOTLAND<br />

Valb<strong>org</strong> Gudmundsdottir<br />

valb<strong>org</strong>g@gmail.com<br />

CBS<br />

Kemitorvet, Building 208<br />

Lyngby, <strong>Denmark</strong><br />

Alejandro Guiliani<br />

guiliani@gmail.com<br />

BRIC<br />

Copenhagen, <strong>Denmark</strong><br />

Ramneek Gupta<br />

ramneek@cbs.dtu.dk<br />

Center for Biological Sequence<br />

Analysis<br />

Lyngby, <strong>Denmark</strong><br />

Frederik Gwinner<br />

frederik.gwinner@pasteur.fr<br />

Institut Pasteur<br />

Paris,France<br />

René Normann Hansen<br />

rnha@novonordisk.com<br />

Novo Nordisk A/S<br />

Søb<strong>org</strong>, <strong>Denmark</strong><br />

V. Joachim Haupt<br />

joachim.haupt@biotec.tu-dresden.de<br />

Biotechnology Center, Technische<br />

Universitet Dresden<br />

Dresden, Germany<br />

Peter Henriksen<br />

peterhe@cpr.ku.dk<br />

Copenhagen University, CPR<br />

København, <strong>Denmark</strong><br />

Markus Herrgard<br />

herrgard@biosustain.dtu.dk<br />

DTU Biosustainability<br />

Hørsholm, <strong>Denmark</strong><br />

Sanna Herrgard<br />

herrgard@cbs.dtu.dk<br />

Danish Technical University<br />

Kongens Lyngby, <strong>Denmark</strong><br />

Carl-Magnus Høgerkorp<br />

cmgh@novonordisk.com<br />

Novo Nordisk<br />

Måløv, <strong>Denmark</strong><br />

Heiko Horn<br />

heiko.horn@cpr.ku.dk<br />

NNF Center for Protein Research,<br />

Faculty of Health Sciences, University<br />

of Copenhagen<br />

København, Danmark<br />

Martin Hornshaw<br />

martin.hornshaw@thermofisher.com<br />

Thermo<br />

Hemel Hempstead, UK<br />

Luisa Hugerth<br />

luisa.hugerth@scilifelab.se<br />

Science for Life Laboratory<br />

Solna, Sweden<br />

Peter Husen<br />

phusen@bmb.sdu.dk<br />

Department of Biochemistry and Molecular<br />

Biology, University of Southern<br />

<strong>Denmark</strong><br />

Odense M, <strong>Denmark</strong><br />

Ruth Hüttenhain<br />

huettenhain@imsb.biol.ethz.ch<br />

Institute for Molecular Systems Biology,<br />

ETH Zurich<br />

Zurich, Switzerland<br />

Tae Hyun Hwang<br />

hwang071@umn.edu<br />

University of Minnesota<br />

Minneapolis, USA<br />

Janus Jakobsen<br />

janus.jakobsen@bric.ku.dk<br />

University of Copenhagen<br />

Copenhagen N, <strong>Denmark</strong><br />

Janette Jones<br />

janette.jones@unilever.com<br />

Unilever<br />

Bebington, Wirral, UK<br />

Haja Kadarmideen<br />

hajak@life.ku.dk<br />

Faculty of Health and Medical<br />

Sciences<br />

Frederiksberg C, Danmark<br />

Kumaran Kandasamy<br />

kkandasamy@cemm.oeaw.ac.at<br />

Center for Molecular Medicine<br />

Vienna, Austria<br />

Sophie Kershaw<br />

sophie.kershaw@keble.ox.ac.uk<br />

University of Oxford<br />

Oxford, United Kingdom<br />

Theo Knijnenburg<br />

t.knijnenburg@nki.nl<br />

Netherlands Cancer Institute<br />

Amsterdam 1066 CX, Netherlands<br />

Lisette Kogelman<br />

lkog@life.ku.dk<br />

Copenhagen University<br />

Frederiksberg C, <strong>Denmark</strong><br />

Alexey Kopylov<br />

kopylov.alex@gmail.com<br />

Chemistry Department of Moscow<br />

State University, Apto-pharm<br />

Moscow, Russian Federation<br />

Nevan Krogan<br />

Nevan.Krogan@ucsf.edu<br />

UCSF<br />

San Francisco, CA, USA<br />

Inna Kuperstein<br />

inna.kuperstein@curie.fr<br />

Institut Curie<br />

Paris, France<br />

Marie Kveib<strong>org</strong><br />

marie.kveib<strong>org</strong>@bric.ku.dk<br />

Copenhagen University<br />

Copenhagen, <strong>Denmark</strong><br />

Anna-Maria Lahesmaa-Korpinen<br />

anna-maria.lahesmaa@helsinki.fi<br />

University of Helsinki<br />

Finland<br />

Janne Marie Laursen<br />

jml@cbs.dtu.dk<br />

Technical University of <strong>Denmark</strong><br />

Kgs. Lyngby, <strong>Denmark</strong><br />

Ben Lehner<br />

lehner.ben@gmail.com<br />

EMBL-CRG<br />

Barcelona, Spain<br />

Rune Linding<br />

linding@cbs.dtu.dk<br />

C-SIG, CBS, DTU<br />

Lyngby, DENMARK<br />

Riku Louhimo<br />

Riku.Louhimo@Helsinki.FI<br />

University of Helsinki<br />

Finland<br />

Raphaëlle Luisier<br />

raphaelle.luisier@unibas.ch<br />

Basel University, Biozenturm<br />

Basel, Switzerland<br />

72 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 73


Kenji Maeda<br />

maeda@embl.de<br />

EMBL-Heidelberg<br />

Heidelberg, Germany<br />

Mohammad Mobashir<br />

mohammad.mobashir@med.ovgu.de<br />

Otto-von-Guericke University<br />

Magdeburg, Germany<br />

Gian Luca Negri<br />

gian.negri@ucd.ie<br />

University College of Dublin<br />

Dublin, Ireland<br />

Elena Nikonova<br />

elena.nikonova@ucdconnect.ie<br />

University College Dublin<br />

Dublin, Ireland<br />

Tri Hieu Nim<br />

nimtrihieu<strong>11</strong><strong>11</strong>@gmail.com<br />

Singapore-MIT Alliance<br />

Singapore, Singapore<br />

Garry Nolan<br />

gnolan@stanford.edu<br />

Stanford University<br />

San Francisco, CA, USA<br />

Matt Onsum<br />

monsum@merrimackpharma.com<br />

Merrimack Pharmaceuticals<br />

Cambridge, MA, USA<br />

Alessandro Ori<br />

alessandro.ori@embl.de<br />

EMBL<br />

Heidelberg, Germany<br />

Bernhard Palsson<br />

bpalsson@ucsd.edu<br />

UCSD/DTU<br />

San Diego/Lyngby, USA/<strong>Denmark</strong><br />

Galina Pavlova<br />

lkorochkin@mail.ru<br />

Institute of Gene Biology, Apto-pharm<br />

Moscow, Russian Federation<br />

Dana Pe’er<br />

dpeer@biology.columbia.edu<br />

Columbia University<br />

New York, MA, USA<br />

Helle Krogh Pedersen<br />

hellekp@cbs.dtu.dk<br />

DTU<br />

Copenhagen, <strong>Denmark</strong><br />

Stine Falsig Pedersen<br />

sfpedersen@bio.ku.dk<br />

University of Copenhagen<br />

Copenhagen 2100, <strong>Denmark</strong><br />

Lars Hagsholm Pedersen<br />

lap@bioneer.dk<br />

Bioneer<br />

Hørsholm, Danmark<br />

Norbert Perrimon<br />

perrimon@receptor.med.harvard.edu<br />

Harvard Medical School<br />

Boston, MA, USA<br />

Ian Prior<br />

iprior@liv.ac.uk<br />

University of Liverpool<br />

Liverpool, UK<br />

Christian Hove Rasmussen<br />

chvr@novonordisk.com<br />

Novo Nordisk A/S<br />

Søb<strong>org</strong>, Søb<strong>org</strong><br />

Angela Re<br />

re@science.unitn.it<br />

Trento University<br />

Trento, Italy<br />

Xavier Robin<br />

Xavier.Robin@unige.ch<br />

University of Geneva<br />

Genève, Switzerland<br />

Honor <strong>May</strong> Rose<br />

rose@fmp-berlin.de<br />

Leibniz Institute of Molecular Pharmacology<br />

(FMP Berlin)<br />

Berlin, Germany<br />

Janine Roy<br />

janine.roy@biotec.tu-dresden.de<br />

Biotechnology Center, Technische<br />

Universitet Dresden<br />

Dresden, Germany<br />

Francesca Sacco<br />

francesca.sacco@uniroma2.it<br />

University of Rome Tor Vergata<br />

Roma, Italia<br />

Katrin Sameith<br />

k.sameith@umcutrecht.nl<br />

Molecular Cancer Research, UMC<br />

Utrecht<br />

Utrecht, The Netherlands<br />

ParticiPantS ParticiPantS<br />

Gabriele Scheler<br />

gscheler@gmail.com<br />

Carl-Correns Foundation for Mathematical<br />

Biology<br />

Mountain View, USA<br />

Thomas Schlitt<br />

thomas.schlitt@kcl.ac.uk<br />

King’s College London<br />

London, UK<br />

Erwin Schoof<br />

schoofe@cbs.dtu.dk<br />

Center for Biological Sequence<br />

Analysis<br />

Lyngby, <strong>Denmark</strong><br />

Veit Schwemmle<br />

veitveit@gmail.com<br />

SDU<br />

Odense, <strong>Denmark</strong><br />

Benno Schwikowski<br />

benno@pasteur.fr<br />

Institut Pasteur<br />

Paris, France<br />

Alain Sewer<br />

Alain.Sewer@pmi.com<br />

PMP SA<br />

Neuchatel, Switzerland<br />

Greg Slodkowicz<br />

greg@cbs.dtu.dk<br />

DTU<br />

Kgs. Lyngby, <strong>Denmark</strong><br />

Nathaniel Stanley<br />

nathaniel.stanley@gmail.com<br />

Universitat Pompeu Fabra<br />

Barcelona, Spain<br />

Ulrich Stelzl<br />

stelzl@molgen.mpg.de<br />

MPI-MG<br />

Berlin, Germany<br />

Sebastian Student<br />

sebastian.student@polsl.pl<br />

Silesian University of Technology<br />

Gliwice, Poland<br />

Anthony Sullivan<br />

anthony.sullivan@absciex.com<br />

AB SCIEX<br />

Warrington WA1 1RX, UK<br />

Damian Szklarczyk<br />

damian.szk@gmail.com<br />

NNF Center for Protein Research<br />

Copenhagen, <strong>Denmark</strong><br />

Agnieszka Szwajda<br />

agnieszka.szwajda@helsinki.fi<br />

Institute for Molecular Medicine Finland<br />

(FIMM)<br />

Helsinki FI-00014, Finland<br />

Camille Terfve<br />

terfve@ebi.ac.uk<br />

EMBL-EBI<br />

Hinxton, United Kingdom<br />

Rossukon Thongwichian<br />

thongwichian@fmp-berlin.de<br />

Leibniz Institute of Molecular Pharmacology<br />

(FMP Berlin)<br />

Berlin, Germany<br />

Ala Trusina<br />

trusina@nbi.dk<br />

Niels Bohr Institute<br />

Copenhagen<br />

<strong>Denmark</strong><br />

Kalliopi Tsafou<br />

ptsafou@gmail.com<br />

NNF Center for Protein Research,<br />

Faculty of Health Sciences, University<br />

of Copenhagen<br />

København, Danmark<br />

Bora Uyar<br />

bora.uyar@embl.de<br />

EMBL<br />

Heidelberg, Germany<br />

Anatoly Uzdensky<br />

auzd@yandex.ru<br />

Southern Federal University<br />

Rostov-on-Don, Russia<br />

Irene van Dijk<br />

I.v.Dijk@acta.nl<br />

Academic Centre Dentistry Research<br />

(ACTA)<br />

Amsterdam, Netherlands<br />

Hanne Varmark<br />

hvarmark@gmail.com<br />

Dept. of systems biology<br />

Lyngby, <strong>Denmark</strong><br />

Jose Velazquez<br />

jvelazqu@mail.nih.gov<br />

National Institute on Aging<br />

Bethesda, USA<br />

Marc Vidal<br />

marc_vidal@dcfi.edu<br />

DCFI, HMS<br />

Boston, USA<br />

Marian Walhout<br />

marian.walhout@cbs.dtu.dk<br />

UMASS Medical School<br />

Worcester, USA<br />

Robert Weatheritt<br />

robert.weatheritt@embl.de<br />

EMBL<br />

Heidelberg , Germany<br />

Rasmus Wernersson<br />

raz@intomics.com<br />

Intomics<br />

Lyngby, <strong>Denmark</strong><br />

Lodewyk Wessels<br />

l.wessels@nki.nl<br />

Netherlands Cancer Institute<br />

Amsterdam, Netherlands<br />

Dorien Wijte<br />

dorien.wijte@gmail.com<br />

DTU<br />

Lyngby, <strong>Denmark</strong><br />

Christof Winter<br />

christof.winter@med.lu.se<br />

Lund University<br />

Lund, Sweden<br />

Jonathan Woodsmith<br />

woodsmit@molgen.mpg.de<br />

MPIMG<br />

Berlin, Germany<br />

Christopher Workman<br />

workman@cbs.dtu.dk<br />

DTU<br />

Kgs. Lyngby, <strong>Denmark</strong><br />

Michael Yaffe<br />

myaffe@mit.edu<br />

MIT<br />

Cambridge, MA, USA<br />

Andrei Zinovyev<br />

andrei.zinovyev@curie.fr<br />

Institut Curie<br />

Paris, France<br />

74 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 75


conFerence venue<br />

Comwell Borupgaard<br />

Nørrevej 80<br />

3070 Snekkersten<br />

<strong>Denmark</strong><br />

+45 4838 0399<br />

WiFi<br />

For wireless internet choose TDC Hotspot and log in with<br />

Password: comwell<br />

Username: comwell<br />

inFo<br />

Production<br />

COVER AND POSTER DESIGN<br />

Studio Grafico L'Asterisco<br />

Rome, Italy<br />

<strong>www</strong>.asterisco.eu<br />

LAYOUT<br />

JS - CEC - GP<br />

PRINTED AT<br />

<strong>www</strong>.GP-Reklame.dk<br />

76 / INB <strong>2012</strong> • <strong>11</strong>-<strong>13</strong> <strong>May</strong> <strong>2012</strong> <strong>www</strong>.<strong>networkbio</strong>.<strong>org</strong> / 77


Network<br />

Medicine

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!