17.11.2012 Views

Candida Infection Biology – fungal armoury, battlefields ... - FINSysB

Candida Infection Biology – fungal armoury, battlefields ... - FINSysB

Candida Infection Biology – fungal armoury, battlefields ... - FINSysB

SHOW MORE
SHOW LESS

Create successful ePaper yourself

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

<strong>FINSysB</strong> Conference<br />

<strong>Candida</strong> <strong>Infection</strong><br />

<strong>Biology</strong> –<br />

<strong>fungal</strong> <strong>armoury</strong>,<br />

<strong>battlefields</strong><br />

and host defences<br />

10-14 October, 2011<br />

Acquafredda di Maratea, ITALY<br />

1<br />

The <strong>FINSysB</strong> Marie Curie<br />

Initial Training Network is<br />

supported by the European<br />

Commission.<br />

(grant PITN-GA-2008-214004).


Index<br />

Programme at a glance..................... Pages 4-5<br />

Full Programme.................................<br />

Speaker Abstracts<br />

Pages 7-11<br />

Tuesday, October 11......................... Pages 13-48<br />

Wednesday, October 12.................... Pages 51-70<br />

Thursday, October 13........................<br />

Poster Abstracts<br />

Pages 73-106<br />

The Pathogenic Armoury................... Page 112<br />

The Key Battlefields........................... Page 159<br />

The Defensive Shields....................... Page 177<br />

Defeating the Enemy......................... Page 191<br />

List of Participants............................ Page 203<br />

3


MONDAY OCTOBER 10<br />

<strong>FINSysB</strong> Programme at a glance<br />

Arrivals<br />

TUESDAY OCTOBER 11<br />

08:30 Welcome - Al Brown<br />

08:40 Session I: The Pathogenic Armoury [Fungal virulence factors]<br />

Chair: Bernhard. Hube<br />

Speakers: Malcolm Whiteway, Derek Sullivan, Pedro Miramón<br />

10:15 Coffee and Posters<br />

10:45 Session II: The Pathogenic Armoury [Fungal infection associated genes]<br />

Chair: Joachim Ernst<br />

Speakers: Scott Filler, Joachim Morschhäuser,<br />

Francesco Citiulo, Malcolm McLean,<br />

Ilse Jacobsen<br />

12:30 Lunch and Posters<br />

14:30 Session III: The Pathogenic Armoury [Fungal fitness attributes]<br />

Chair: Mathias Lavie-Richard<br />

Speakers: Janet Quinn, Mike Gustin, Christoph Schüller,<br />

Michael Lorenz, Iryna Bohovych<br />

16:15 Coffee and Posters<br />

16:55 Session IV: The Pathogenic Armoury [Modelling <strong>fungal</strong> responses]<br />

Chair: Edda Klipp<br />

Speakers: Francesc Posas, Duccio Cavalieri, Katarzyna Tyc,<br />

Iuliana Ene, Quentin Lagadec<br />

18:40 Close<br />

WEDNESDAY OCTOBER 12<br />

08:30 Session V: The Key Battlefields [<strong>Candida</strong> biofilms]<br />

Chair: Christophe d’Enfert<br />

Speakers: Aaron Mitchell, Carol Kumamoto, Vitor Cabral,<br />

Alice Sorgo<br />

4


10:20 Coffee and Posters<br />

11:00 Session VI: The Key Battlefields [<strong>Candida</strong> cell wall]<br />

Chair: Frans Klis<br />

Speakers: Carol Munro, Peter Lipke, Iaroslava Kos,<br />

Isabel Miranda, Jane Usher, Craig Murdoch<br />

13:00 Lunch and Posters<br />

14:30 Afternoon for networking and sunshine<br />

THURSDAY OCTOBER 13<br />

08:30 Session VII: The Defensive Shields [Host immune responses]<br />

Chair: Mihai Netea<br />

Speakers: Alberto Mantovani, Gordon Brown, Diana Rosentul<br />

10:05 Coffee and Posters<br />

10:45 Session VIII: The Defensive Shields [Host immune responses]<br />

Chair: Anna Vecchiarelli<br />

Speakers: Jürgen Ruland, Julian Naglik, Neelam Pandey,<br />

Shih-Chin Cheng, Eliška Svobodová<br />

12:30 Lunch and Posters<br />

14:30 Session IX: Defeating The Enemy [Novel anti<strong>fungal</strong> and diagnostic targets]<br />

Chair: Andy Porter<br />

Speakers: Antonio Cassone Deborah O’Neil,<br />

Clemens Heilmann, Abhishek Saxena, Kate Dobb<br />

16:15 Coffee and Posters<br />

16:55 Session X: Defeating The Enemy [Novel anti<strong>fungal</strong> and diagnostic targets]<br />

Chair: Jason Oliver<br />

Speakers: Haoping Liu, Inês Correia, Saif Hameed,<br />

Antresh Kumar<br />

18:10 Close<br />

19:30 Conference Dinner<br />

FRIDAY OCTOBER 14<br />

Departures<br />

5


Full Programme<br />

<strong>Candida</strong> <strong>Infection</strong> <strong>Biology</strong> – <strong>fungal</strong> <strong>armoury</strong>,<br />

<strong>battlefields</strong> and host defences<br />

Acquafredda di Maratea, Italy<br />

10-14 October 2011<br />

MONDAY OCTOBER 10 Arrivals<br />

TUESDAY OCTOBER 11<br />

08:30 Welcome - Al Brown<br />

08:40 Session I: The Pathogenic Armoury [Fungal virulence factors]<br />

Chair – Bernhard Hube<br />

8:40-9:20 Keynote Lecture<br />

Malcolm Whiteway [Montreal, Canada]<br />

Transcriptional rewiring of <strong>fungal</strong> ribosomal gene<br />

9:30-9:55 Derek Sullivan [Dublin, Ireland]<br />

What part of the Pathogenic Armoury is <strong>Candida</strong> dubliniensis<br />

missing?<br />

10:00-10:10 Pedro Miramón [Jena, Germany]<br />

Dissecting the response of <strong>Candida</strong> albicans to the attack by<br />

neutrophils<br />

10:15 Coffee and Posters<br />

10:45 Session II: The Pathogenic Armoury [Fungal infection associated<br />

genes]<br />

Chair – Joachim Ernst<br />

10:45-11:10 Scott Filler [Los Angeles, USA]<br />

EGFR and HER2 signalling mediate epithelial cell invasion by<br />

<strong>Candida</strong> albicans during oropharyngeal infection<br />

11:15-11:40 Joachim Morschhäuser [Würzburg, Germany]<br />

Transcriptional control of white-opaque switching in <strong>Candida</strong><br />

albicans and its role in host-pathogen interactions<br />

11:45-11:55 Francesco Citiulo [Jena, Germany]<br />

Characterization of the molecular mechanism used by C. albicans to<br />

acquire zinc from host cells.<br />

12:00-12:10 Malcolm McLean [Revohot, Israel]<br />

Strand asymmetry in <strong>Candida</strong> genes<br />

7


12:15-12:25 Ilse Jacobsen [Jena, Germany]<br />

Functional analysis of C. albicans infection-associated genes with<br />

unknown function during interaction with host cells and in complex<br />

infection models<br />

12:30 Lunch and Posters<br />

14:30 Session III: The Pathogenic Armoury [Fungal fitness attributes]<br />

Chair - Mathias L. Richard<br />

14:30-14:55 Janet Quinn [Newcastle, UK]<br />

Survival in the host –mechanisms underlying responses to oxidative<br />

stress in the human pathogen <strong>Candida</strong> albicans<br />

15:00-15:25 Mike Gustin [Houston, USA]<br />

Anticipation in the nitrosative stress response of <strong>Candida</strong> albicans<br />

15:30-15.40 Christoph Schüller [Vienna, Austria]<br />

Revisiting the Hog pathway of C. glabrata<br />

15:45-15:55 Michael Lorenz [Houston, USA]<br />

<strong>Candida</strong> albicans alters extracellular pH under host-relevant<br />

conditions of carbon starvation<br />

16:00-16:10 Iryna Bohovych [Aberdeen, UK]<br />

Glucose promotes oxidative stress resistance in <strong>Candida</strong> albicans via<br />

specific signalling pathways<br />

16:15 Coffee and Posters<br />

16:55 Session IV: The Pathogenic Armoury [Modelling <strong>fungal</strong> responses]<br />

Chair – Edda Klipp<br />

16:55-17:20 Francesc Posas [Barcelona, Spain]<br />

Stress-responses controlled by the Hog1 stress-activated protein<br />

kinase.<br />

17:25-17:50 Duccio Cavalieri [Florence, Italy]<br />

Friend or Foe: Systems biology approaches to elucidate the<br />

interaction between fungi and their hosts<br />

17:55-18:05 Katarzyna Tyc [Berlin, Germany]<br />

Agent based modelling and host pathogen interactions<br />

18:10-18:20 Iuliana Ene [Aberdeen, UK]<br />

Impact of non-fermentative growth on the C. albicans cell wall and<br />

dynamic responses to osmotic stress<br />

18:25-18:35 Quentin Lagadec [Düsseldorf, Germany]<br />

Target specificity of the Efg1 regulator in <strong>Candida</strong> albicans<br />

18:40 Close<br />

8


WEDNESDAY OCTOBER 12<br />

08:30 Session V: The Key Battlefields [<strong>Candida</strong> biofilms]<br />

Chair – Christophe d’Enfert<br />

8:30-9:10 Keynote Lecture<br />

Aaron Mitchell [Pittsburgh, USA]<br />

Genetic control of <strong>Candida</strong> albicans biofilm formation<br />

9:20-9:45 Carol Kumamoto [Boston, USA]<br />

<strong>Candida</strong> albicans: sensing the host environment<br />

9:50-10:00 Vitor Cabral [Paris, France]<br />

Identification of <strong>Candida</strong> albicans genes involved in biofilm formation<br />

by an over-expression approach<br />

10:05-10:15 Alice Sorgo [Amsterdam, The Netherlands]<br />

Cell wall stress elicits a conserved response in <strong>Candida</strong> albicans<br />

10:20 Coffee and Posters<br />

11:00 Session VI: The Key Battlefields [<strong>Candida</strong> cell wall]<br />

Chair – Frans Klis<br />

11:00-11:25 Carol Munro [Aberdeen, UK]<br />

<strong>Candida</strong> albicans cell wall dynamics<br />

11:30-11:55 Peter Lipke [New York, USA]<br />

Force-dependent activation of functional amyloid domains increases<br />

cell adhesion in C. albicans<br />

12:00-12:10 Iaroslava Kos [Thiverval Grignon, France]<br />

Iff2 is a cell surface protein involved in adhesion<br />

12:15-12:25 Isabel Miranda [Porto, Portugal]<br />

Deciphering the role of CUG mistranslation on <strong>Candida</strong> albicans<br />

adhesion and cell surface variation<br />

12:30-12:40 Jane Usher [Exeter, UK]<br />

Interaction of a <strong>Candida</strong> glabrata transcription factor knock-out<br />

library with the Drosophila melanogaster innate immune system<br />

12:45-12:55 Craig Murdoch [Sheffield, UK]<br />

Using zebrafish to study <strong>Candida</strong> albicans-mediated systemic<br />

candidiasis in vivo<br />

13:00 Lunch and Posters<br />

14:30 Afternoon for networking and sunshine<br />

9


THURSDAY OCTOBER 13<br />

08:30 Session VII: The Defensive Shields [Host immune responses]<br />

Chair – Mihai Netea<br />

8:30-9:10 Keynote Lecture<br />

Alberto Mantovani [Milan, Italy]<br />

The long pentraxin PTX3 as a paradigm of humoral pattern<br />

recognition molecules in antimicrobial resistance<br />

9:20-9:45 Gordon Brown [Aberdeen, UK]<br />

Pattern recognition and anti-<strong>fungal</strong> immunity: The role of C-type<br />

lectins<br />

9:50-10:00 Diana Rosentul [Nijmegen, The Netherlands]<br />

The Tyr238X dectin-1 polymorphism is a predisposing factor for<br />

mucosal <strong>Candida</strong> albicans infections but not candidemia<br />

10:05 Coffee and Posters<br />

10:45 Session VIII: The Defensive Shields [Host immune responses]<br />

Chair – Anna Vecchiarelli<br />

10:45-11:10 Jürgen Ruland [Munich, Germany]<br />

C-type lectin receptor signaling in innate anti-<strong>fungal</strong> immunity<br />

11:15-11:40 Julian Naglik [London, UK]<br />

<strong>Candida</strong> - epithelial interactions and innate immunity<br />

11:45-11:55 Neelam Pandey [Perugia, Italy]<br />

<strong>Candida</strong> albicans secreted aspartic proteases cause inflammatory<br />

response irrespective of their proteolytic activity<br />

12:00-12:10 Shih-Chin Cheng [Nijmegen, The Netherlands]<br />

A central role of complement for the <strong>Candida</strong> albicans-induced<br />

cytokine production by human cells<br />

12:15-12:25 Eliška Svobodová [Jena, Germany]<br />

The role of pH-regulated antigen 1 of <strong>Candida</strong> albicans in the<br />

interaction of the fungus with human neutrophils<br />

12:30 Lunch and Posters<br />

14:30 Session IX: Defeating The Enemy [Novel anti<strong>fungal</strong> and diagnostic<br />

targets]<br />

Chair – Andy Porter<br />

14:30-14:55 Antonio Cassone [Rome, Italy]<br />

Vaccines against opportunistic <strong>fungal</strong> infections<br />

10


15:00-15:25 Deborah O’Neil [Novabiotics, UK]<br />

Tackling the <strong>Candida</strong> spp infection spectrum with first-in-class<br />

antimicrobial peptide technology<br />

15:30-15:40 Clemens Heilmann [Amsterdam, The Netherlands]<br />

Wall proteins, absolute quantification and concatenated vaccines:<br />

A new approach for <strong>Candida</strong> albicans therapy?<br />

15:45-15:55 Abhishek Saxena [Aberdeen, UK]<br />

scFv Phage Display Library against Complex Immunomodulatory<br />

Glycans<br />

16:00-16:10 Kate Dobb [Manchester, UK]<br />

Essential Genes as Broad-Spectrum Anti<strong>fungal</strong> Targets<br />

16:15 Coffee and Posters<br />

16:55 Session X: Defeating The Enemy [Novel anti<strong>fungal</strong> and diagnostic<br />

targets]<br />

Chair – Jason Oliver<br />

16:55-17:20 Haoping Liu [Irvine, USA]<br />

Hyphal development in <strong>Candida</strong> albicans requires two temporally<br />

linked changes in promoter chromatin for initiation and maintenance<br />

17:25-17:35 Inês Correia [Madrid, Spain]<br />

The transcription factor Sko1 represses the yeast-to-hypha transition<br />

and mediates the oxidative stress response in <strong>Candida</strong> albicans.<br />

17:40-17:50 Saif Hameed [New Delhi, India]<br />

Calcineurin signaling and membrane lipid homeostasis regulates iron<br />

mediated multidrug resistance mechanisms in <strong>Candida</strong> albicans<br />

17:55-18:05 Antresh Kumar [New Delhi, India]<br />

Functional analysis of H-loop residues of Cdr1p, an ABC transporter<br />

of human <strong>fungal</strong> pathogen <strong>Candida</strong> albicans.<br />

18:10 Close<br />

19:30 Conference Dinner<br />

FRIDAY OCTOBER 14 Departures<br />

11


SPEAKER ABSTRACTS<br />

(In order of presentation)<br />

Tuesday, October 11<br />

13


Transcriptional rewiring of <strong>fungal</strong> ribosomal gene<br />

M. Whiteway, H. Lavoie, H. Hogues<br />

NRC-BRI 6100 Royalmount Ave, Montreal, and McGill <strong>Biology</strong>, 1205 Penfield, Montreal<br />

Examination of transcriptional regulatory circuits in fungi have uncovered many<br />

examples of what is termed rewiring - transcription factors dedicated to controlling<br />

a particular regulon in one species are found to control distinct regulons in other<br />

species. A dramatic example of this phenomenon has been observed for the control<br />

of the expression of the genes encoding the proteins that make up the ribosome.<br />

The Tbf1 and Cbf1 proteins are key regulators of ribosomal gene expression in<br />

C. albicans, but these proteins have been replaced in ribosomal control in the<br />

S. cerevisiae lineage by Rap1p, and serve in yeast as telomere binding and<br />

centromere binding proteins respectively. We can trace the evolutionary progression<br />

of this process and speculate on mechanisms driving this exchange of controlling<br />

elements.<br />

14


What part of the Pathogenic Armoury is <strong>Candida</strong><br />

dubliniensis missing?<br />

Derek Sullivan and Gary Moran<br />

Division of Oral Biosciences, Dublin Dental University Hospital, Trinity College, Dublin 2,<br />

Ireland.<br />

The species most closely related to <strong>Candida</strong> albicans is <strong>Candida</strong> dubliniensis,<br />

which, despite sharing many phenotypic attributes is a far less common cause of<br />

human infection and is significantly less virulent in a wide range of infection models.<br />

One of the most important virulence factors associated with C. albicans is the ability<br />

to produce hyphae. Although both species have the capacity to produce filaments,<br />

C. albicans is able to do so under a wider range of environmental conditions. We<br />

have shown that the presence of nutrients specifically inhibits filamentation by C.<br />

dubliniensis and that this can be alleviated by rapamycin, suggesting that<br />

differences in the Tor1 signalling pathway may play a role in the different ability of<br />

C. albicans and C. dubliniensis to produce hyphae.<br />

Comparison of the genomes of C. albicans and C. dubliniensis has confirmed that<br />

the two species are very closely related, with high levels of synteny and 96% of<br />

genes exhibiting >80% identity. Interestingly, one of the most notable differences<br />

between the two genomes is the absence of hypha-specific virulence genes such<br />

as ALS3, SAP4,5 and HYR1. Comparative transcriptomic analysis has also<br />

identified differences in gene expression in the two species that have led to the<br />

identification of Sfl2, a putative heat shock transcription factor that is divergent in<br />

the two species and which appears to play a role in the control of filamentation in<br />

C. albicans.<br />

These data suggest that C. dubliniensis may be losing the ability to produce hyphae,<br />

and while this may reduce it’s capacity to cause invasive infection it may reflect<br />

adaptation to commensal growth in a specific anatomic niche which has yet to be<br />

identified.<br />

16


Dissecting the response of <strong>Candida</strong> albicans to the attack<br />

by neutrophils<br />

Pedro Miramón 1 , Iryna Bohovych 4 , Alistair J. P. Brown 4 , Oliver Kurzai 2,3 ,<br />

Bernhard Hube 1,3<br />

1 Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product<br />

Research and <strong>Infection</strong> <strong>Biology</strong> - Hans Knoell Institute (HKI), Jena, Germany; 2 Department<br />

of Fungal Septomics, Leibniz Institute for Natural Product Research and <strong>Infection</strong> <strong>Biology</strong> -<br />

Hans Knoell Institute (HKI), Jena, Germany; 3 Friedrich-Schiller Universität, Jena, Germany;<br />

4 Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, UK<br />

<strong>Candida</strong> albicans successfully colonises diverse niches in the human host without causing<br />

any harm thanks to a balance with the microflora and possibly due to a constant surveillance<br />

of patrolling immune cells. However, under certain conditions, this fungus is able to cause<br />

life-threatening deep-seated infections. Although it is know that neutrophils are key players<br />

in controlling and eliminating C. albicans, it is unclear how C. albicans is killed by neutrophils<br />

and how the fungus counteracts these phagocytes.<br />

By means of single-cell expression profiling using GFP reporter strains, we characterised<br />

different responses of C. albicans to three main neutrophil-imposed stresses: nutrient<br />

starvation, oxidative stress and nitrosative stress.<br />

We observed that phagocytosed <strong>fungal</strong> cells responded to carbohydrate starvation by<br />

upregulating the glyoxylate cycle, but not the glycolytic pathway. Moreover, a mutant lacking<br />

a key gene of the glyoxylate cycle (icl1 ) exhibited decreased resistance in the presence of<br />

neutrophils.<br />

Next, we investigated the contribution of oxidative stress to killing by neutrophil. The catalase<br />

gene CTA1 was induced upon phagocytosis. However, the surface-associated superoxide<br />

dismutase gene SOD5 was induced in yeast cells even before phagocytosis took place,<br />

suggesting that C. albicans can sense and encounter an extracellular oxidative attack. In a<br />

similar fashion, the thioredoxin gene TRX1 was also upregulated upon extracellular contact<br />

with neutrophils. Strikingly, neither cta1 nor trx1 mutants exhibited decreased resistance<br />

to neutrophils, suggesting that they are not essential for normal resistance of C. albicans.<br />

However, sod5 was more sensitive to neutrophils, indicating that Sod5 is an important<br />

detoxifying enzyme to cope with neutrophil-generated oxidative stress. C. albicans also<br />

responded to nitrosative stress generated in the intracellular milieu and this response was<br />

essential for full resistance as a yhb1 mutant, defective in nitric oxide dioxygenase, exhibited<br />

decreased survival.<br />

In summary, our study provides evidence that C. albicans displays specific responses to<br />

overcome either extracellular or intracellular attack by neutrophils.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial Training Network (PITN-<br />

GA-2008-214004).<br />

18


EGFR and HER2 signalling mediate epithelial cell invasion<br />

by <strong>Candida</strong> albicans during oropharyngeal infection<br />

Weidong Zhu, 1 Quynh T. Phan, 1 Pinmanee Boontheung, 2 Norma V. Solis, 1<br />

Joseph A. Loo, 2,3 Scott G. Filler 1,4<br />

1 Department of Medicine, Division of Infectious Diseases, Los Angeles Biomedical Research<br />

Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA; 2 Department of<br />

Chemistry and Biochemistry, University of California-Los Angeles, Los Angeles, CA 90095,<br />

USA; 3 Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los<br />

Angeles, CA 90095, USA; 4 Department of Medicine, David Geffen School of Medicine at<br />

UCLA, Los Angeles, CA 90024, USA<br />

A key feature of oropharyngeal candidiasis is <strong>fungal</strong> invasion of oral epithelial cells,<br />

a process that can occur by several mechanisms, including <strong>fungal</strong>-induced<br />

endocytosis. Two invasins, Als3 and Ssa1, induce epithelial cell endocytosis of C.<br />

albicans, in part by binding to E-cadherin. However, inhibition of E-cadherin function<br />

only partially reduces C. albicans endocytosis, suggesting that there are addition<br />

epithelial cell receptors for this organism. Here, we demonstrate that the epidermal<br />

growth factor receptor (EGFR) and a related receptor, HER2 function cooperatively<br />

to induce the endocytosis of C. albicans hyphae. EGFR and HER2 are bound by<br />

the C. albicans in an Als3- and Ssa1-dependent manner, and this binding induces<br />

receptor autophosphorylation. Signalling through both EGFR and HER2 is required<br />

for maximal epithelial cell endocytosis of C. albicans in vitro. However, heterologous<br />

expression studies in NIH-3T3 cells indicate that HER2 can mediate endocytosis<br />

of C. albicans by itself, whereas EGFR cannot. In the mouse model of oropharyngeal<br />

infection, C. albicans induces phosphorylation of epithelial cell EGFR and HER2.<br />

Importantly, oral treatment of mice with a dual EGFR and HER2 kinase inhibitor<br />

significantly reduces EGFR and HER2 phosphorylation and attenuates the severity<br />

of oropharyngeal candidiasis. These results demonstrate the importance of EGFR<br />

and HER2 signalling in the pathogenesis of oropharyngeal candidiasis and indicate<br />

the feasibility of treating candidal infections by targeting the host cell receptors to<br />

which the fungus binds.<br />

20


Transcriptional control of white-opaque switching in<br />

<strong>Candida</strong> albicans and its role in host-pathogen<br />

interactions<br />

Michael Weyler, 1 Christoph Sasse, 1 Mike Hasenberg, 2 Matthias Gunzer, 2<br />

Joachim Morschhäuser 1<br />

1 Institut für Molekulare Infektionsbiologie, Universität Würzburg, Germany; 2 Institut für<br />

Molekulare und Klinische Immunologie, Universität Magdeburg, Germany<br />

<strong>Candida</strong> albicans is a morphologically versatile microorganism that can undergo<br />

various developmental programmes, which in turn determine its interactions with<br />

the mammalian host. The transition from the yeast form to filamentous growth,<br />

which is induced by environmental signals, is associated with tissue invasion during<br />

an infection. In addition, C. albicans strains that are homozygous at the mating type<br />

locus can spontaneously and reversibly switch from the normal yeast morphology<br />

(white) to an elongated cell type (opaque), which is the mating competent form of<br />

the fungus. White-opaque switching also influences the ability of C. albicans to<br />

colonize and proliferate in specific host niches and its susceptibility to host defense<br />

mechanisms. We used live imaging to observe the interaction of white and opaque<br />

cells with host phagocytic cells. When mixed populations of differentially labelled<br />

white and opaque cells were incubated with human polymorphonuclear leucocytes<br />

(PMNs) on a surface, the neutrophils selectively phagocytosed and killed white cells,<br />

despite frequent encounters with opaque cells. White cells were usually attacked<br />

only after they started to form a germ tube, indicating that the suppression of<br />

filamentation in opaque cells saved them from recognition by the PMNs. In contrast<br />

to neutrophils, dendritic cells internalized white and opaque cells without apparent<br />

difference. However, when embedded in a collagen matrix, the PMNs also<br />

phagocytosed both white and opaque cells with equal efficiency. These results<br />

suggest that, depending on the environment, white-opaque switching enables C.<br />

albicans to escape from specific host defense mechanisms. We created a genomewide<br />

inducible transcription factor expression library, which was used to identify<br />

novel transcriptional regulators of the developmental programmes of C. albicans.<br />

These studies provide insight into the regulation of cell fate decisions in C. albicans<br />

and their consequences for the host-pathogen interaction.<br />

22


Characterization of the molecular mechanism used by<br />

C. albicans to acquire zinc from host cells.<br />

Francesco Citiulo1 , Duncan Wilson1 1, 2<br />

and Bernhard Hube<br />

1 Department of Microbial Pathogenicity Mechanisms, Hans-Knöll-Institute (HKI) - Leibniz<br />

Institute for Natural Product Research and <strong>Infection</strong> <strong>Biology</strong>, Beutenbergstr. 11a, D-07745<br />

Jena, and 2 Friedrich Schiller University Jena, Germany<br />

Zinc is an essential trace element for several cellular processes of many organisms<br />

ranging from microbes to multicellular animals. Similar to iron up-take systems,<br />

successful microbial pathogens must have specialized zinc acquisition and uptake<br />

systems to gain this essential metal from the host during infection. In this study, we<br />

investigated the molecular mechanisms used by C. albicans to acquire zinc from<br />

the host.<br />

In silico analyses of the C. albicans secretome identified multiple zinc binding<br />

domains in Pra1 (pH-regulated antigen 1), a 35 kDa cell wall associated and<br />

secreted protein, which is expressed by hyphal cells and which has previously been<br />

shown to interact with complement-regulatory proteins and immune cell receptors.<br />

A mutant lacking functional Pra1 (pra1 ) showed growth defects when cultured in<br />

zinc limited conditions suggesting a role for Pra1 in zinc homeostasis.<br />

During interaction with Human Umbilical Vein Endothelial Cells (HUVEC) under zinc<br />

limited conditions, hyphae of the C. albicans pra1 mutant had significantly<br />

decreased abilities to sequester zinc from host cells as compared to wild type cells.<br />

Although pra1 was capable of endothelial invasion, the mutant caused significantly<br />

reduced damage of these cells and this effect was rescued by the addition of<br />

exogenous zinc.<br />

These data suggest that Pra1 plays a role in zinc binding during infection and that<br />

successful acquisition of this metal is critical for C. albicans pathogenicity.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial Training Network<br />

(PITN-GA-2008-214004).<br />

24


Strand asymmetry in <strong>Candida</strong> genes<br />

Malcolm McLean<br />

Department of Molecular Genetics, Weizmann Institute of Science, Revohot, Israel<br />

Genes of virtually all organisms have more A than T on the coding strand.<br />

Additionally <strong>Candida</strong> albicans, in common with many unicellular fungi, shows a<br />

distinct pattern of GC skew. The start of the gene, including the promoter region,<br />

is enriched in C, the end of the gene is enriched in G. However the pattern is much<br />

weaker in the model organisms, Saccharomyces cerevisiae and<br />

Schizosaccharomyces pombe, which is probably why it has not been observed until<br />

now. However similar AT and GC skews have been found in other organisms, and<br />

the standard explanation is mutational pressure. Roughly, the strands are in different<br />

environments when being duplicated or transcribed, and thus accumulate different<br />

mutations.<br />

However it is hard to account for a change of sign of GC skew with the mutational<br />

pressure hypothesis. Furthermore, analysis of recent substitution and indel<br />

mutations, obtained by aligning homologous genes from related species, does not<br />

support mutational pressure as the force maintaining the GC skew. We instead<br />

propose a selective hypothesis. CTP is the nucleotide tri-phosphate with the<br />

smallest pool. We propose that CTP loading is a rate-limiting step in RNA pol II<br />

elongation (the biochemical evidence is ambiguous), and that a gradient of<br />

decreasing C and increasing G in the mRNA transcript reduces the possibility of<br />

RNA pol II traffic jams in highly transcribed genes.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial Training Network<br />

(PITN-GA-2008-214004).<br />

26


Functional analysis of C. albicans infection-associated<br />

genes with unknown function during interaction with host<br />

cells and in complex infection models<br />

Ilse D. Jacobsen 1 , Duncan Wilson 1 , Francois Mayer 1 , Bernhard Hube 1,2<br />

1 Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product<br />

Research and <strong>Infection</strong> <strong>Biology</strong> - Hans-Knöll-Institute, Jena, Germany; 2 Friedrich-Schiller<br />

University, Jena<br />

<strong>Candida</strong> albicans is able to cause a variety of clinical infections. We hypothesized<br />

that C. albicans unknown function genes, which were transcriptionally upregulated<br />

during oral infections, liver invasion or incubation in human blood (“infection<br />

associated genes”) might contribute to the infection process. We selected a subset<br />

of infection-associated genes with unknown function for further analysis.<br />

Twentyfour isogenic deletion mutants were constructed and subsequently analyzed<br />

both in vitro and in infection models of increasing complexity. Sixteen mutants were<br />

impaired in their ability to damage monolayers of human endothelial and/or<br />

epithelial cells. Within this subset, seven mutants additionally displayed decreased<br />

stress resistance in vitro. Only two mutants showed filamentation defects. To<br />

determine whether the deleted genes influenced virulence in more complex<br />

infections models, all 24 mutants were tested in ovo for their ability to kill chicken<br />

embryos infected on the chorio-allantoic membrane. We recently showed that<br />

mortality in this model depends on the <strong>fungal</strong> ability to invade the membrane and<br />

that the pro-inflammatory host response likely contributes to pathogenesis.<br />

Surprisingly, only seven mutants were attenuated in this model, of which five were<br />

also attenuated in damaging epithelial cells in vitro. Three of the mutants, which<br />

were attenuated in ovo, and one mutant, which was attenuated in damaging<br />

endothelial and epithelial cells but fully virulent in chicken embryos, were<br />

subsequently analyzed in a systemic mouse model. Virulence in mice mirrored the<br />

results obtained in ovo.<br />

Our results demonstrate that genes essential for causing full damage of epithelial<br />

or endothelial cells in vitro are not necessarily crucial for full virulence in complex<br />

infection models. We conclude that either such damage potential is not critical for<br />

full virulence in the complex host situation or that the greater complexity of the<br />

environment in animal models stimulates additional <strong>fungal</strong> regulatory networks<br />

leading to compensation of the defect.<br />

28


Survival in the host –mechanisms underlying responses to<br />

oxidative stress in the human pathogen <strong>Candida</strong> albicans<br />

Alessandra da Silva Dantas 1 , Miranda Patterson 1 , Deborah Smith 1 ,<br />

Catherine Bruce 1 , Donna MacCallum 2 , Lars Erwig 3 , Brian Morgan 1 &<br />

Jan Quinn 1<br />

1Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle<br />

University, Newcastle upon Tyne, NE2 4HH, United Kingdom; 2Aberdeen Fungal Group,<br />

Institute of Medical Sciences, University of Aberdeen, Aberdeen, AB25 2ZD, United Kingdom;<br />

3Division of Applied Medicine, Institute of Medical Sciences, University of Aberdeen,<br />

Aberdeen, AB25 2ZD, United Kingdom.<br />

The ability of the major systemic <strong>fungal</strong> pathogen of humans, <strong>Candida</strong> albicans, to<br />

sense and respond to reactive oxygen species (ROS) such as H 2 O 2 , generated by<br />

the host immune system, is essential for survival in the host. This oxidative stress<br />

response at the transcriptional level is regulated largely by the AP-1-like<br />

transcription factor Cap1 and, to a lesser extent, the Hog1 stress-activated protein<br />

kinase (SAPK). In addition, H 2 O 2 treatment of C. albicans stimulates the formation<br />

of hyperpolarized buds in a mechanism that requires activation of the Rad53<br />

checkpoint kinase. However, despite strong links between filamentous growth,<br />

responses to ROS, and virulence, remarkably little is known about the intracellular<br />

signalling mechanisms that regulate the activation of such H 2 O 2 -responsive<br />

signalling pathways in C. albicans. During this talk I will present recent work from<br />

my group which has investigated the mechanisms underlying oxidative stressmediated<br />

activation of both the Cap1 transcription factor, and the Hog1 and Rad53<br />

stress responsive kinases. Whilst the Saccharomyces cerevisiae paradigm has<br />

provided important insight into oxidative stress signalling mechanisms, our studies<br />

have revealed new circuitry, novel players and unexpected mechanisms in the<br />

medically relevant pathogen C. albicans.<br />

30


Anticipation in the Nitrosative Stress Response of <strong>Candida</strong><br />

albicans<br />

Jed Lynn, Erin Stashi, Lisa Sun, Michael Gustin<br />

Dept Biochemistry and Cell <strong>Biology</strong>, Rice University, Houston, TX 77005<br />

The human-resident fungus <strong>Candida</strong> albicans is regularly exposed to nitric oxide<br />

(NO) released by immune cells or non-enzymatically generated from nitrite (NO 2 - )<br />

produced by other microbes in the mouth and skin. One focus of our research is<br />

the molecular mechanisms by which C. albicans senses and defends itself against<br />

NO-related nitrosative stress. C. albicans defense against fungistatic NO requires<br />

the YHB1-encoded nitric oxide dioxygenase which converts NO to nontoxic nitrate<br />

(NO 3 ). NO strongly induces expression of YHB1 and this induction plus NO<br />

resistance requires the zinc cluster transcription factor Cta4, bound to the YHB1<br />

promoter. In Saccharomyces cerevisiae, NO induces ScYHB1, but this induction<br />

requires a different class of transcription factor, the zinc finger protein Fzf1. To test<br />

whether Cta4 is sufficient for NO induction of CaYHB1, a S. cerevisiae strain was<br />

constructed containing a CaYHB1 promoter-lacZ reporter gene and a DNA cassette<br />

allowing tetracycline-regulated expression of CaCTA4. Expression of the latter<br />

induced high level activity of the reporter gene, but both basal and Cta4-induced<br />

lacZ expression were unaffected by NO released from nitrite. In contrast, nitrite<br />

strongly induced expression of a ScYHB1 promoter-lacZ reporter gene in S.<br />

cerevisiae. These data show that Cta4 is not sufficient for NO regulation of CaYHB1<br />

expression and suggest NO sensing involves a negative regulator of Cta4 function.<br />

Unexpectedly, C. albicans NO resistance is induced by not just NO or nitrite, but<br />

also by the nontoxic anions nitrate (NO 3 - ) and thiocyanate (SCN - ). Most other<br />

inorganic anions so far tested, e.g., Cl - , HCO 3 - , are without effect. Microarray<br />

analysis of genome-wide mRNA levels show that NO, nitrite, nitrate, and<br />

thiocyanate each induce nearly identical sets of genes. Nitrate and thiocyanate<br />

induction of CaYHB1 each require CTA4. In contrast, the native YHB1 gene in S.<br />

cerevisiae is induced by NO or nitrite, but not by nitrate or thiocyanate. These results<br />

suggest a model in which the non-toxic saliva chemicals nitrate or thiocyanate are<br />

sensed by C. albicans, allowing anticipation and defense against NO-caused<br />

nitrosative stress .<br />

32


Revisiting the Hog Pathway of C. glabrata<br />

Zeljkica Jandric, Wolfgang Reiter, Gustav Ammerer, Christoph Schüller<br />

Max F. Perutz Laboratories, Department of Biochemistry University of Vienna, Dr.Bohr-Gasse<br />

9/5, A-1030 Vienna, Austria<br />

Signal transduction networks mediated by mitogen – activated protein kinases<br />

(MAPK) play essential roles in eukaryotic cells. Yeast cells have developed<br />

osmoadaptation pathways to cope with osmotic changes in the microenvironment.<br />

In Saccharomyces cerevisiae five MAPK pathways are well characterized. One of<br />

these, especially associated with stress response, is the High Osmolarity Glycerol<br />

(HOG) pathway . Stress induced phosphorylation triggers the activation of the key<br />

MAPK, Hog1. After being phosphorylated by the upstream MAPKK Pbs2, Hog1<br />

enters the cell nucleus and modulates the transcription of its target genes.<br />

Orthologues of the all HOG pathway compounds found in Saccharomyces<br />

cerevisiae have been identified in fungi and animals.<br />

The human <strong>fungal</strong> pathogen <strong>Candida</strong> glabrata is closely related to Saccharomyces<br />

cerevisiae. It is a common commensal but in immunocompromised cancer and<br />

transplant patients or elder persons it can turn into an opportunistic pathogen.<br />

Depending on the host environment, <strong>Candida</strong> glabrata has to adapt to<br />

environmental changes during invasion and adaptation steps. We investigated the<br />

role of Hog1 in <strong>Candida</strong> glabrata. Several stress sources were used to identify HOGspecific<br />

phenotypes and to analyze the role of Hog1 in <strong>Candida</strong> glabrata. As<br />

expected, the deletion of Hog1 caused osmostress hypersensitivity. Physiological<br />

stress conditions, such as nutrients starvation, were simulated by infection of<br />

macrophages with <strong>Candida</strong> glabrata and determination of the number of surviving<br />

<strong>Candida</strong> cells. Cghog1∆ cells showed a much higher sensitivity to macrophages<br />

compared to wild type cells. Further investigations focus on stress induced gene<br />

transcription in Cghog1∆ cells and comparison of the transcription pattern to cells<br />

without Hog1 deletion.<br />

34


<strong>Candida</strong> albicans alters extracellular pH under<br />

host-relevant conditions of carbon starvation<br />

Slavena Vylkova, Heather Danhof, and Michael C. Lorenz<br />

Dept. of Microbiology and Molecular Genetics, The University of Texas Health Science<br />

Center, 6431 Fannin St., Houston, TX 77030, USA<br />

Phagocytosis by immune cells, such as macrophages, induces a remarkable<br />

change in transcription and physiology in <strong>Candida</strong> albicans, with ~10% of the<br />

genome affected. A primary aspect of this response is metabolic shift reflecting<br />

deprivation for carbon, and this response is required for full virulence. We developed<br />

an in vitro system to mimic this response, using media that is glucose-poor and<br />

amino acid-rich; surprisingly, in these conditions, we observed a striking change in<br />

the media pH when C. albicans was grown at an initial pH of 4. Over 12-24 hours,<br />

the pH rose to 7-7.5, and this alkalinization was associated with the induction of<br />

hyphal growth – a key virulence trait in this species. This extracellular pH change<br />

was not dependent on factors previously known to regulate pH responses, such as<br />

RIM101 and MNL1. Using a combination of genetic and genomic screens, we have<br />

linked this process to amino acid catabolism: mutation of genes required for amino<br />

acid permease function and breakdown of amino acids as a carbon source are<br />

impaired in pH changes. Transcript profiles confirm that this pH alteration is the<br />

result of a metabolic change to the use of amino acids as a carbon source, similar<br />

to the profile of phagocytosed cells. Alkalinizing cultures release measurable<br />

quantities of ammonia, a strong base, and we propose that catabolism of amino<br />

acids as a carbon source renders the amine group in excess, and this is excreted<br />

to change the pH. This occurs in conditions that mimic those our data suggest exist<br />

after phagocytosis, and we are now examining the intracellular fate of ingested C.<br />

albicans cells. Published reports are contradictory regarding this fate, but the weight<br />

of evidence indicates intracellular trafficking is aberrant. We propose that C.<br />

albicans alters phagolysosomal pH to induce hyphal growth and impair the<br />

fungicidal effects of the immune cell.<br />

36


Glucose promotes oxidative stress resistance in <strong>Candida</strong><br />

albicans via specific signalling pathways<br />

Iryna Bohovych 1 , Pedro Miramón 2 , Aaron Mitchell 3 , Bernhard Hube 2 ,<br />

Alistair JP Brown 1<br />

1 Aberdeen Fungal Group, School of Medical Sciences, University of Aberdeen, Foresterhill,<br />

Aberdeen AB25 2ZD, UK; 2 Microbial Pathogenicity, Hans Knoell Institute, Beutenbergstraße<br />

11a, Jena 07745, Germany; 3 Department of Biological Sciences, Carnegie Mellon University,<br />

Pittsburgh, Pennsylvania 15213, USA<br />

<strong>Candida</strong> albicans, a successful human pathogen, displays the phenomenon of<br />

glucose-enhanced oxidative stress resistance (Rodaki et al., 2009), which is not<br />

observed in other yeast species tested. The molecular mechanisms that mediate<br />

glucose-enhanced oxidative stress resistance in C. albicans are not clear. We<br />

reasoned that glucose signalling might play a major role. Therefore we tested the<br />

impact of specific C. albicans mutations to determine which of known signalling<br />

pathways are required for glucose-enhanced oxidative stress resistance.<br />

The Sugar Receptor-Repressor (Hgt4-Rgt1) pathway, homologous to the Rgt2/Snf3<br />

pathway in S. cerevisiae, is not required for the phenotype. In contrast, a snf1<br />

mutant, which lacks a key component of the Glucose Repression Pathway,<br />

displayed high oxidative stress resistance on lactate, not altered by glucose. A kis1<br />

mutant, lacking one of the subunits of Snf1 complex, displayed a related phenotype.<br />

The data indicate that in the absence of glucose the Glucose Repression Pathway<br />

represses glucose-enhanced oxidative stress resistance.<br />

cAMP signalling also appears to play a role in glucose-enhanced oxidative stress<br />

resistance. This phenotype was repressed by exogenous dibutyryl cAMP and by<br />

a pde2 mutation that is thought to increase intracellular cAMP levels. Also the<br />

phenotype was enhanced by inactivation of adenylyl cyclase (cyr1).<br />

Having shown that both cAMP signalling and the Glucose Repression Pathway play<br />

important roles in mediating glucose-enhanced stress resistance, the next step was<br />

to identify targets of these pathways that might contribute to the phenotype.<br />

Comparative analyses of transcriptomic profiles of C. albicans glucose- and lactategrown<br />

cells in response to oxidative stress and glucose treatment correspondingly<br />

revealed a small set of commonly up-regu lated genes (Enjalbert et al., 2006; Rodaki<br />

et al., 2009). The impact of the cAMP and Glucose Repression pathways on these<br />

genes is being determined.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial Training Network<br />

(PITN-GA-2008-214004).<br />

38


Stress-responses controlled by the Hog1 stress-activated<br />

protein kinase<br />

Sergi Regot, Núria Conde, Mariona Nadal, Carme Solé, Alba Duch, Javier<br />

Jimenez, Alberto Gonzalez, Eulàlia de Nadal and Francesc Posas<br />

Cell Signaling Unit, Departament de Ciències Experimentals i de la Salut. Universitat<br />

Pompeu Fabra (UPF). PRBB. C/ Doctor Aiguader 88. Barcelona E-08003 (Spain).<br />

Eulalia.Nadal@upf.edu; francesc.posas@upf.edu<br />

Exposure of yeast cells to increases in extracellular osmolarity results in the<br />

activation of the Hog1 MAP kinase. Activation of this MAP kinase is required to<br />

generate a set of osmoadaptive responses essential to survive under high<br />

osmolarity conditions. Fast and transient activation of the Hog1 signaling pathway<br />

is required for proper adaptation. Adaptation to osmostress requires the induction<br />

of a large number of genes, which indicates the necessity to regulate several<br />

aspects of the cell physiology. Induction of gene expression is highly dependent<br />

on the presence of the MAP kinase, which suggests a key role for the HOG signaling<br />

pathway in the regulation of gene expression in response to osmostress. In<br />

response to stress, the MAPK controls several mechanisms related to transcription<br />

initiation and elongation as well as chromatin organization. The MAPK also controls<br />

cell cycle. Here, the MAPK is able to modulate cell cycle delay in different phases<br />

which highlight the relevance of cell cycle control in response to stress.<br />

40


Friend or Foe: Systems biology approaches to elucidate<br />

the interaction between fungi and their hosts<br />

Duccio Cavalieri<br />

Department of Preclinic and Clinic Pharmacology, University of Florence, viale Pieraccini 6,<br />

50139 Firenze. duccio.cavalieri@unifi.it, telephone number: +39-055-4271327, fax number:<br />

+39-055-4271280<br />

Modelling the networks subtending the fruitful coexistence of fungi and their<br />

mammalian host is becoming increasingly important to control emerging <strong>fungal</strong><br />

pathogens. The goal of Systems <strong>Biology</strong> is to obtain a superior level of knowledge<br />

of how function arises in dynamic interactions. Systems <strong>Biology</strong> requires the<br />

acquisition of information on the different levels of regulation of a biological system<br />

and its integration to unravel the complexity and the dynamic nature of the hostfungus<br />

interaction. Immunity is not simply the product of a series of discrete linear<br />

signaling pathways; rather it is comprised of a complex set of integrated responses<br />

arising from a dynamic network of thousands of molecules subject to multiple<br />

influences. In this perspective the <strong>fungal</strong> microbiota and the host should be<br />

analyzed as an ecosystem, and disease considered as an alteration of the<br />

equilibrium. Our work addresses host-fungus interaction with a special focus on<br />

systems biology approaches to investigate the mechanisms developed by <strong>Candida</strong><br />

albicans and Aspergillus fumigatus to circumvent host immune responses during<br />

<strong>fungal</strong> infections and the phenotypic traits that potentially make S.cerevisiae a<br />

microorganism generally recognized as safe. Such genome wide integrative<br />

approaches hold the promise to significantly improve our ability to understand<br />

which <strong>fungal</strong> traits can be considered potential threats and the regulatory networks<br />

involved in immune subversion.<br />

42


Agent based modelling and host pathogen interactions<br />

Katarzyna M. Tyc 1 , Clemens Kühn 1 , Edda Klipp 1<br />

1 Theoretische Biophysik, Humboldt-Universität zu Berlin, Berlin, Germany<br />

Mathematical models of pathogen and host interactions provide a tool for medical<br />

applications and potential for pharmaceutical companies for testing novel drug<br />

treatments and consequences of their application. Host pathogen interactions are<br />

highly complex and experimental techniques even when combined with different<br />

computational methods allow only for partial investigation of their mutual<br />

dependences.<br />

Based on the available literature we reconstruct a computational model of a human<br />

oral epithelium invasion by the pathogenic fungus <strong>Candida</strong> albicans. We investigate<br />

the system’s dynamics using the agent based modelling approach (ABM) where<br />

cells (<strong>Candida</strong> cells, neutrophils (PMNs)) are represented as individual agents. These<br />

agents interact in different ways: <strong>fungal</strong> cells form hyphal colonies on the epithelium<br />

and then epithelial cells secrete cytokines upon contact with the hyphae form of<br />

fungi, indicating inflammation. Cytokine secretion is one of the signals stimulating<br />

PMNs recruitment to the site of infection. Mix of different gradients such as lactate<br />

dehydrogenase (LDH) release from damaged epithelium, cytokines, and secreted<br />

hyphal particles influences PMN activity. In our model, we investigate the essential<br />

parameters that establish the system’s dynamics. We also look at the<br />

consequences of an anti<strong>fungal</strong> drug treatment on the <strong>Candida</strong> albicans survival rate<br />

and its dose dependence. We test the optimal intervals and doses for new<br />

application.<br />

With agent based models, we can investigate the effect of individual dynamics on<br />

global patterns, enabling an intuitive view on host-pathogen interactions that can<br />

provide relevant predictions for further experiments.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial Training Network<br />

(PITN-GA-2008-214004).<br />

44


Impact of non-fermentative growth on the C. albicans cell<br />

wall and dynamic responses to osmotic stress<br />

Iuliana V. Ene, Alistair J. P. Brown<br />

Aberdeen Fungal Group, School of Medical Sciences, University of Aberdeen, Foresterhill,<br />

Aberdeen AB25 2ZD, UK<br />

<strong>Candida</strong> albicans displays considerable metabolic flexibility and robust stress<br />

responses in vivo. Both contribute to the virulence of this pathogen. C. albicans<br />

assimilates both fermentative and non-fermentative carbon sources in vivo, often<br />

occupying poor glucose niches in its mammalian host. We reasoned that changes<br />

in carbon source might influence stress adaptation, but stress responses are<br />

generally studied during growth on glucose.<br />

We found that the C. albicans cell wall is extremely dynamic and susceptible to<br />

variations in carbon source. Growth on lactate rather than glucose has a major<br />

impact on cell wall structure and flexibility. Although the relative proportions of<br />

chitin, -glucan and mannan are similar within the wall of lactate-grown cells, they<br />

display increased porosity and decreased cell wall biomass. TEM confirmed major<br />

differences in cell wall architecture, underlying the importance of the carbon source<br />

in the cell biology of this pathogen. This in turn influences the responses of this<br />

pathogen to stresses as these differences were reflected in the cell wall-related<br />

phenotypes. Lactate-grown cells displayed higher resistance to Congo Red and<br />

Calcofluor White and showed significantly increased resistance to various doses<br />

of osmotic stress. This change was independent of Hog1 and Mkc1, but correlated<br />

with large changes in cell volume following exposure to NaCl. Growth on lactate<br />

also increased resistance to anti<strong>fungal</strong>s such as Caspofungin, Tunicamycin and<br />

Amphotericin B, which is likely to have a major impact upon the behavior of C.<br />

albicans during infection.<br />

The impact of carbon source on stress and drug resistance was confirmed in other<br />

pathogenic <strong>Candida</strong> species and for other carbon sources, resulting in variations<br />

in resistance when compared to a glucose-grown control. Clearly carbon source<br />

strongly influences cell wall architecture and the resistance of C. albicans to certain<br />

stresses, which is likely to have a major impact on its in vivo behavior.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial<br />

Training Network (PITN-GA-2008-214004).<br />

46


Target specificity of the Efg1 regulator in <strong>Candida</strong> albicans<br />

Lassak, T., Lagadec, Q., Kurtz, D., Bussmann, M. and Ernst, J. F.<br />

Heinrich-Heine-Universität Düsseldorf, Institut für Mikrobiologie, Molekulare Mykologie<br />

Efg1 is a general transcription factor, which regulates numerous morphogenetic<br />

and metabolic processes in C. albicans. We previously found that overexpression<br />

of EFG1 and activation of Efg1 during hypha formation leads to downregulation of<br />

EFG1 promoter activity and that EFG1 downregulation is necessary to allow the<br />

formation of true hyphae and prevent pseudohyphal growth.<br />

To explore the Efg1 binding specificity its binding to the EFG1 promoter was<br />

analyzed by EMSA and footprint analyses. These experiments revealed binding of<br />

Efg1 to a promoter sequence close to the transcript start site, which contained a<br />

repeated 8-mer motif (referred to as APSES response element, ARE). Surprisingly,<br />

promoter fusions to the RLUC reporter lacking the ARE motif were still able to<br />

downregulate the EFG1 promoter during hypha formation in an Efg1-dependent<br />

manner. Furthermore, extensive deletions of the 10 kb-large 5 untranslated region<br />

of EFG1 still did not prevent regulation suggesting multiple binding sites for Efg1<br />

within the EFG1 promoter. We subsequently analyzed genome-wide occupation of<br />

chromosomal sequences by Efg1 using ChIP chip analysis. These results revealed<br />

the entire 10 kb EFG1 upstream region as a major site of Efg1 occupancy.<br />

Unexpectedly, Efg1 binding to this region was rapidly lost during hypha induction<br />

suggesting a regulatory model, in which Efg1 acts as transcriptional activator<br />

specifically during yeast growth. In further experiments we attempted to reconstitute<br />

the EFG1 autoregulatory circuit in the yeast S. cerevisiae. In this system the<br />

presence of Efg1 was shown to downregulate EFG1 promoter activity, although not<br />

to the same extent as was observed in C. albicans. This finding hints at coregulators<br />

that together with Efg1 mediate autoregulation in C. albicans.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial Training Network<br />

(PITN-GA-2008-214004).<br />

48


Wednesday, October 12<br />

51


Genetic Control of <strong>Candida</strong> albicans Biofilm Formation<br />

J.S. Finkel 1 , F. Lanni 1 , J. Nett 2 , D. Huang 1 , J. Suhan 1 , A. Nantel 3 , D. Andes 2 ,<br />

A.P. Mitchell 1<br />

1 Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA; 2 Department<br />

of Medicine, University of Wisconsin, Madison, WI; 3 Biotechnology Research Institute,<br />

National Research Council of Canada, Montreal, QC<br />

Implanted medical devices, such as venous catheters and artificial heart valves,<br />

are associated with a significant risk of microbial infection. These infections arise<br />

because device surfaces support biofilm formation. We have worked to define the<br />

genetic basis for C. albicans biofilm formation as a means to understand the<br />

underlying mechanisms. We have focused on transcription factors because they<br />

often govern expression of functionally related genes. We have recently identified<br />

transcription factors that are required for adherence to a silicone substrate in vitro.<br />

In vitro assays indicate that many of these transcription factors are not required for<br />

biofilm formation per se, but are required for normal biofilm morphology.<br />

Importantly, several of these newly defined biofilm transcription factors are required<br />

for biofilm formation in vivo, as assayed in a rat venous catheter model. We will<br />

present target gene analysis that points toward the mechanistic basis for these<br />

biofilm defects.<br />

52


<strong>Candida</strong> albicans: Sensing the Host Environment<br />

Carol A. Kumamoto<br />

Tufts University, Boston, MA, USA<br />

<strong>Candida</strong> albicans, an opportunistic pathogen and a human commensal, is found<br />

almost exclusively in association with a host. Intestinal tract colonization by this<br />

organism is common in humans and disease is thought to arise due to overgrowth<br />

or escape of organisms from the gut. In the host, the organism’s ability to sense<br />

cues from the environment would be expected to enhance its growth and survival.<br />

C. albicans responds to numerous environmental cues that could be encountered<br />

in the host such as temperature, pH, presence of O 2 , CO 2 , nutrients or antimicrobial<br />

compounds, and stress conditions. Our studies focus on the ability of C. albicans<br />

to sense contact with a surface and to sense the environment within the intestinal<br />

tract. These types of sensing mechanisms promote the organism’s ability to sense<br />

its location within the host, the nature of the tissue that it is encountering and the<br />

status of the host’s immune response. As a result of these sensing mechanisms,<br />

the organism controls its physiology so that it maintains benign colonization in a<br />

healthy host but becomes a destructive pathogen that invades host tissue in a<br />

compromised host.<br />

54


Identification of <strong>Candida</strong> albicans genes involved in biofilm<br />

formation by an over-expression approach<br />

Vitor Cabral 1,2 , Sadri Znaidi 1,2 , Mélanie Legrand 1,2 , Keunsook Lee 3 , Sophie<br />

Bachelier-Bassi 1,2 , Murielle Chauvel 1,2 , Tristan Rossignol 1,2 , Carol Munro 3 ,<br />

Christophe d’Enfert 1,2<br />

1 Institut Pasteur, Unité Biologie et Pathogénicité Fongiques, Département Génomes et<br />

Génétique, F-75015 Paris, France; 2 INRA, USC2019, F-75015 Paris, France; 3 Aberdeen<br />

Fungal Group, University of Aberdeen, School of Medical Sciences, Institute of Medical<br />

Sciences, Aberdeen, AB25 2ZD, United Kingdom<br />

<strong>Candida</strong> albicans is the major <strong>fungal</strong> pathogen of humans, being responsible for<br />

benign superficial infections and devastating systemic infections. These latter<br />

infections are often associated with the formation of biofilms on medical devices.<br />

Biofilms are polymicrobial communities adherent to biotic or abiotic surfaces and<br />

are especially resistant to anti<strong>fungal</strong> agents making them difficult to treat. Despite<br />

recent progress, the molecular mechanisms that underlie the formation of biofilms<br />

and the basis for their tolerance to anti<strong>fungal</strong>s remain to be fully understood.<br />

Here, we have undertaken to identify and characterize C. albicans genes whose<br />

over-expression alters biofilm formation. To this aim we have developed through<br />

the Gateway cloning methodology a partial C. albicans ORFeome that includes<br />

531 ORFs and a corresponding collection of C. albicans over-expression strains,<br />

each having a unique C. albicans gene controlled by a doxycycline-inducible<br />

promotor and a unique molecular barcode. This collection has been used to<br />

evaluate the fitness of each strain in competition experiments, taking advantage of<br />

their molecular barcoding for individual microarray-based quantification. First, the<br />

fitness of the strains in planktonic cultures with or without over-expression of the<br />

genes has been studied, revealing 8 genes whose over-expression reduces fitness<br />

(6 related to cell cycle, DNA damage and check-point; 2 involved in filamentation).<br />

Second, the fitness of the strains in biofilms with or without over-expression of the<br />

genes has been investigated. None of the genes whose over-expression altered<br />

fitness in planktonic cultures were identified in this analysis. Strikingly, we observed<br />

a strong enrichment for genes encoding cell wall proteins and genes involved in<br />

budding regulation among those genes whose over-expression altered fitness in<br />

biofilms. Current experiments are aimed at confirming the results of these pool<br />

experiments through analysis of individual over-expression mutants and further<br />

understanding the role of the identified genes in biofilm formation.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial Training Network<br />

(PITN-GA-2008-214004).<br />

56


Cell wall stress elicits a conserved response in <strong>Candida</strong><br />

albicans<br />

Alice G. Sorgo, Clemens J. Heilmann, Sepehr Mohammady, Henk L.<br />

Dekker, Stanley Brul, Chris de Koster, Leo J. de Koning and Frans M. Klis<br />

Swammerdam Institute for Life Sciences, Universiteit van Amsterdam, Science Park 904,<br />

1098 XH Amsterdam, The Netherlands<br />

Both cell wall proteins and secreted proteins of C. albicans are critical for fitness,<br />

virulence and adaptation to environmental challenges. We have recently shown that<br />

fluconazole, an anti<strong>fungal</strong> drug that affects ergosterol synthesis and increases<br />

membrane fluidity, also causes cell wall stress as evidenced by decreased<br />

resistance to wall-perturbing compounds and higher chitin levels in the wall (1). This<br />

is accompanied by substantial changes in the wall proteome and secretome. We<br />

found that the abundance of proteins mainly involved in wall biosynthesis and<br />

integrity increased (Crh11, Pga4, Pir1, Phr1, Phr2, Sap9). However, the abundance<br />

of hypha-associated proteins (Als3, Hwp1, Plb5) decreased in agreement with<br />

reduced hyphal growth. Finally, fluconazole induced cell clustering. As membrane<br />

fluidity also tends to increase at higher temperatures, we wanted to investigate<br />

whether the response to thermal stress is similar to that of fluconazole and could<br />

also result in cell wall stress. When C. albicans was cultured at 42°C, which resulted<br />

in substantial growth inhibition, the cells showed indeed decreased resistance to<br />

wall-perturbing agents, and chitin levels in the wall were elevated, both suggesting<br />

cell wall stress. Likewise, the wall proteome response was similar to that of<br />

fluconazole-treated cells, including increased incorporation levels of proteins<br />

important for wall maintenance and integrity. Additionally, cell separation was<br />

impaired, correlating with a severe reduction of the chitinases Cht2 and Cht3 and<br />

the endoglucanase Eng1 in the medium. In summary, our data indicate that both<br />

thermal stress and fluconazole-induced stress induce membrane stress, which<br />

leads to cell wall stress and a conserved wall stress response.<br />

References<br />

(1) The effects of fluconazole on the secretome, the wall proteome and wall integrity of the clinical fungus<br />

<strong>Candida</strong> albicans. Sorgo AG, Heilmann CJ, Dekker HL, Bekker M, Brul S, de Koster CG, de Koning LJ,<br />

Klis FM. Eukaryot Cell. 2011. doi:10.1128/EC.05011-11.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial Training Network<br />

(PITN-GA-2008-214004).<br />

58


<strong>Candida</strong> albicans cell wall dynamics<br />

Carol A. Munro, Louise A. Walker, Keunsook Lee, David Stead, Laura<br />

Selway, Alistair J.P. Brown and Neil A.R. Gow<br />

Institute of Medical Sciences, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland<br />

The outer surface of <strong>Candida</strong> albicans is composed of chitin and glucan<br />

polysaccharides and an exposed fibrillar layer, rich in highly mannosylated proteins.<br />

The co-ordinated assembly of the cell wall is regulated by a network of signalling<br />

pathways that ensure cellular integrity is maintained in response to intracellular and<br />

extracellular signals. Treatment with sub-inhibitory concentrations of the<br />

echinocandin anti<strong>fungal</strong> drugs, which inhibit beta-1,3-glucan synthesis, results in<br />

significant changes in the cell wall architecture due to activation of compensatory<br />

chitin synthesis and alterations in the cell wall proteome. A number of proteins that<br />

are covalently attached to the cell wall are carbohydrate- active enzymes involved<br />

in modulating and forming cross-links between chitin and glucan. These proteins,<br />

such as Phr1 and Crh11 are positively regulated in response to applied cell wall<br />

and membrane stresses and mutations in cell wall related genes. Changes in the<br />

<strong>fungal</strong> cell wall can in turn alter susceptibility to anti<strong>fungal</strong> drugs in vitro as well as<br />

in vivo.<br />

The cell surface is the interface between host and fungus and a number of virulence<br />

attributes are cell wall localised. Alterations in cell wall composition and architecture<br />

are likely to impinge on pathogenesis and recognition by host’s immune cells. Using<br />

transcript profiling, biochemical assays, glycoproteomics and SILAC (Stable Isotope<br />

Labelling with Amino Acids in Cell Culture) we have started to catalogue quantitative<br />

and qualitative changes in the cell wall in response to pharmacological and genetic<br />

blockade of cell wall biosynthesis. In addition we are studying the phenotypes of a<br />

library of strains that are overexpressing cell wall related genes in order to better<br />

understand the cell wall phenome.<br />

60


Force-dependent activation of functional amyloid domains<br />

increases cell adhesion in C. albicans<br />

Peter N. Lipke 1 , David Alsteens 2 , Cho Tan 1 , Desmond N. Jackson 1 , Caleen<br />

B. Ramsook 1 , Yves F. Dufrêne 2 , and Melissa C. Garcia 1<br />

1 Department of <strong>Biology</strong>, Brooklyn College of the City University of New York; 2 Institute of<br />

Condensed Matter, Université catholique de Louvain, Louvain-le-Neuve, Belgium<br />

Yeast cell adhesion molecules encoded by diverse gene families have conserved<br />

amyloid-forming sequences, and amyloid formation potentiates cell-cell interactions<br />

(Ramsook et al., 2010 Euk. Cell 9:393-404 ; Alsteens et al., 2010 PNAS 107:20744-<br />

9; Garcia et al., 2011 PLoS ONE 6: e17632. doi:10.1371/journal.pone.0017632 ).<br />

Amyloid formation clusters the cell adhesion molecules, and increases cell-cell<br />

binding by clustering of active sites in the same way that multivalence of antibodies<br />

increases avidity relative to affinity of each site.<br />

We show that amyloid formation is initiated by extension force, and results in<br />

formation of nm-scale amyloid domains on the surface of the cell, with different<br />

protein domains playing distinct roles to facilitate amyloid formation. Atomic force<br />

microscopy of single molecules in situ unfolds successive domains in the <strong>Candida</strong><br />

albicans Als adhesins. This unfolding triggers local amyloid formation and<br />

subsequent propagation of amyloid patches around the entire cell surface. Amyloid<br />

formation is accompanied by activation of strong cell-cell and cell-substrate<br />

adhesion. Both amyloid formation and activation of adhesion are inhibited by antiamyloid<br />

dyes and peptides. In addition, a single site mutation in the amyloid core<br />

sequence of Als5p abrogates amyloid formation and activation of adhesion,<br />

although the mutant protein is well-folded and maintains in vitro binding activity.<br />

Amyloid formation is also triggered by forces developed between cells during<br />

assays or by prolonged vortex mixing of cells. The various protein domains in Als<br />

proteins synergize to promote this force-induced amyloid formation. The<br />

consequence is formation of nanoscale patches of amyloid on the cell surface to<br />

potentiate cell interactions by formation of multiple intercellular bonds.<br />

Supported by NIH SCORE grant SC1 GM083756<br />

62


Iff2 is a cell surface protein involved in adhesion<br />

Iaroslava Kos 1 , Grégory Da Costa 1 , Céline Monniot 1 , Anita Boisramé 1<br />

Marie-Noëlle Bellon Fontaine 2 , Murielle Chauvel 3 , Christophe d’Enfert 3 and<br />

Mathias L. Richard 1<br />

1 Team “Virulence and Fungal <strong>Infection</strong>” Institut Micalis, UMR1319 INRA AgroParisTech,<br />

78850 Thiverval-Grignon, FRANCE; 2 Team “Bioadhésion et Hygiène des Matériaux” Institut<br />

Micalis, UMR1319 INRA AgroParisTech, 91744 Massy, FRANCE; 3 Team “Fungal <strong>Biology</strong> and<br />

Pathogenicity”, Pasteur Institute, 75015 Paris, FRANCE.<br />

In <strong>Candida</strong> albicans, the cell wall and especially cell wall proteins are known to play<br />

a key role in the relationship between the <strong>fungal</strong> cell, the host and most indwelling<br />

device, contributing to adhesion, immune response modulation and deep seated<br />

infections. A specific class of cell wall proteins has been particularly under the<br />

scope in the past decade: the glycosylphosphatidylinositol anchored proteins<br />

(GpiPs). The largest family of GpiPs is a family of 12 proteins sharing a high similarity<br />

with Hyr1. One of its members, Iff2 has been the subject of a large scale study in<br />

our laboratory in order to decipher his putative function in C. albicans. The cell<br />

localization of Iff2 was assessed using a V5-tagged protein and clearly showed that<br />

Iff2 is a cell surface protein tethered to the cell wall proteins through alkali-Pir-like<br />

links as well as di-sulfide bonds. We also demonstrated that Iff2 protrude from the<br />

cell wall glucan network at the interface with the cellular environment. At the gene<br />

level, we showed that IFF2 is a gene with an expression level stronger than most of<br />

the member of Hyr/Iff family in laboratory conditions and that IFF2 is induced during<br />

the stationary phase in the same media. An overexpressing strain with IFF2 under<br />

the control of TEF1p was hyper adherent to different plastic but not to silicon,<br />

resulting in the formation of stronger biofilm at the early stage of biofilm formation<br />

compared to the reference strain. The future work is aimed to understand which<br />

part of the protein is important for adhesion and if it is specific to Iff2 functional<br />

domain. Additional experiments are also planned to monitor the consequences of<br />

this overexpression in in vitro and in vivo models.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial Training Network<br />

(PITN-GA-2008-214004).<br />

64


Deciphering the role of CUG mistranslation on <strong>Candida</strong><br />

albicans adhesion and cell surface variation<br />

Isabel Miranda 1,2 , Ana Silva-Dias 1,2 , Rita Rocha 1 , Teresa Gonçalves 3 , Cidália<br />

Pina-Vaz 1,2,4 , Scott G. Filller 5,6 and Acácio G. Rodrigues 1,2,7<br />

1 Department of Microbiology, Faculty of Medicine, University of Porto, 2 Cardiovascular<br />

Research & Development Unit, Faculty of Medicine, University of Porto, Portugal, 3 Center<br />

for Neurosciences and Cell <strong>Biology</strong>, Institute of Microbiology, Faculty of Medicine, University<br />

of Coimbra, Portugal; 4 Department of Microbiology, Hospital S. João; 5 Department.of<br />

Medicine, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center,<br />

Torrance, California, USA; 6 David Geffen School of Medicine at UCLA, Los Angeles,<br />

California, USA; 7 Burn Unit and Department of Plastic and Reconstructive Surgery, Hospital<br />

S. João, Portugal.<br />

Interactions with the surrounding environment represent a continuous stressing<br />

challenge for microorganisms. Until now, the mechanisms regulating changes in<br />

cell-surface in the most common human <strong>fungal</strong> pathogen still remain unclear. In<br />

<strong>Candida</strong> albicans, the CUG codon is naturally mistranslated as two biochemically<br />

distinct residues, serine and leucine. Interestingly, genes encoding adhesins such<br />

as the ALS family are particularly enriched in CUG codons. In order to evaluate the<br />

role of CUG mistranslation in C. albicans adhesion, we compared the behavior of<br />

two strains: a wild-type, in which 3% of CUG codons are mistranslated as leucine<br />

and a highly CUG mistranslating strain wherein 28% of CUG codons are<br />

mistranslated as leucine. We found that the increase in leucine misincorporation<br />

enhances cell wall hydrophobicity and the capacity of yeast cells to adhere to<br />

biomaterials. This adhesion phenotype was further confirmed in Saccharomyces<br />

cerevisiae, through expression of C. albicans ALS3 gene in its two isoforms: Als3p-<br />

Leu and Als3p-Ser. Als3p-Leu yeasts prompted a strong adhesion phenotype,<br />

resulting in flocculation in liquid media and two-to-fivefold more adhesion than<br />

Als3p-Ser. The cell wall variability generated by CUG mistranslation may not be<br />

confined to adhesion and could be relevant for immune system evasion. Indeed,<br />

highly CUG mistranslating cells were less susceptible to phagocytosis by murine<br />

macrophages than wild-type, probably due to the more efficient glucan masking<br />

caused by CUG mistranslation. We propose that CUG mistranslation was<br />

maintained during C. albicans evolution due to its potential to generate cell surface<br />

variability.<br />

A Silva-Dias is supported by a FCT (Fundação Ciência e Tecnologia) doctoral grant<br />

SFRH/BD/44896/2008; I Miranda is supported by Ciência 2008 (FCT) and European Social Fund.<br />

66


Interaction of a <strong>Candida</strong> glabrata transcription factor<br />

knock-out library with the Drosophila melanogaster innate<br />

immune system<br />

Jane Usher 3 , Ilias Kounatidis 1 , Biao Ma 2 , Petros Ligoxygakis 1 &<br />

Ken Haynes 3<br />

University of Oxford 1 , Imperial College London 2 , University of Exeter 3<br />

At least in part <strong>Candida</strong> species are thought to invade following initial<br />

gastrointestinal colonisation. The mechanisms underpinning this translocation<br />

depend on many factors including host intestinal flora, mucosal damage and<br />

deficient host immunity. In addition <strong>fungal</strong> attributes eg the ability to acquire<br />

nutrients and to adapt to host environmental insults are essential to disease<br />

initiation and progression.<br />

To analyse this interaction on a holistic level we have created a library of <strong>Candida</strong><br />

glabrata mutants and developed a Drosophila melanogaster model of<br />

gastrointestinal candidosis. A library of ~200 bar coded mutant strains, each lacking<br />

a specific DNA binding protein, was constructed in a derivative of C. glabrata ATCC<br />

2001. Each of these was screened in a D. melanogaster larval GI infection model,<br />

in which third instar larvae, containing a Drs-GFP allele, were fed with C. glabrata<br />

infected food. Drosomycin (Drs) is activated in the larval fat body in response to<br />

wild-type C. glabrata colonization. Of the 200 knock-out mutants 12 failed to<br />

activate Drs-GFP indicating reduced activation of host innate immunity. We are<br />

currently investigating the basis of this reduced activation.<br />

68


Using zebrafish to study <strong>Candida</strong> albicans-mediated<br />

systemic candidiasis in vivo<br />

Thomas Evans, Simon Tazzyman, Timothy J.A. Chico, Martin H. Thornhill,<br />

Craig Murdoch.<br />

University of Sheffield, Sheffield, UK<br />

<strong>Candida</strong> albicans is a common oral opportunistic pathogen that causes<br />

mucocutaneous infections and rarely systemic candidiasis, a nosocomial infection<br />

which involves the haematogenous spread of <strong>Candida</strong> to multiple organs and is<br />

associated with high mortality. To cause tissue damage C.albicans must bind to the<br />

endothelium of blood vessels, leave the circulation and invade tissues. However,<br />

little is known about the mechanisms involved in these processes. This study aimed<br />

to use transparent embryonic zebrafish (Danio rerio) as an in vivo infection model<br />

in conjunction with an in vitro flow adhesion assay to study how C.albicans bind to<br />

the endothelium, leave the circulation and invade tissues. Suspensions of viable or<br />

treated C.ablicans were flowed over monolayers of endothelial cells and adherent<br />

<strong>Candida</strong> counted. Zebrafish were injected with C.albicans and mortality determined<br />

after 18h. Time-lapse confocal microscopy using fluorescent C.albicans and blood<br />

vessels was performed to image dissemination. Viable C.albicans bound to<br />

endothelium significantly (p


Thursday, October 13<br />

73


The long pentraxin PTX3 as a paradigm of humoral pattern<br />

recognition molecules in antimicrobial resistance<br />

Alberto Mantovani<br />

Laboratory of Immunology and Inflammation , Istituto Clinico Humanitas, Rozzano (Milan),<br />

Italy; and Department of Translational Medicine, University of Milan, Italy<br />

PTX3 is a multifunctional soluble pattern recognition receptor characterized by a<br />

C-terminal domain highly homologous to C-reactive protein and serum amyloid P<br />

component, associated to a N-terminal domain unrelated to other known proteins.<br />

PTX3 is produced upon stimulation with proinflammatory cytokines and Toll-like<br />

receptor engagement by different cell types, including endothelial cells,<br />

monocytes/macrophages, dendritic cells, fibroblasts and epithelial cells. The<br />

molecule binds with high affinity complement component, microbial moieties,<br />

growth factors and apoptotic cells. PTX3 plays non-redundant functions including<br />

innate immunity against selected microorganisms to regulation of inflammation.<br />

PTX3 plays a nonredundant role in resistance against selected fungi, A. fumigatus<br />

in particular. Moreover, levels of PTX3 and genetic polymorphisms are associated<br />

with infections in humans. In addition PTX3 has a regulatory function. PTX3 serves<br />

as a paradigm for the interplay between the cellular and the humoral arm of innate<br />

immunity.<br />

Bottazzi B, Doni A, Garlanda C, Mantovani A. An Integrated View of Humoral Innate Immunity: Pentraxins<br />

as a Paradigm. Annu Rev Immunol. 2010, 28:157-83.<br />

Deban L, Russo RC, Sironi M, Moalli F, Scanziani M, Zambelli V, Cuccovillo I, Bastone A, Gobbi M,<br />

Valentino S, Doni A, Garlanda C, Danese S, Salvatori G, Sassano M, Evangelista V, Rossi B, Zenaro E,<br />

Constantin G, Laudanna C, Bottazzi B, Mantovani A. Regulation of leukocyte recruitment by the long<br />

pentraxin PTX3. Nat Immunol. 2010 Apr;11:328-34.<br />

74


Pattern recognition and anti-<strong>fungal</strong> immunity: The role of<br />

C-type lectins<br />

Gordon D. Brown<br />

Aberdeen Fungal Group, University of Aberdeen, UK<br />

The innate ability to detect pathogens is essential for multicellular existence, and<br />

has been achieved through the evolution of germ-line encoded receptors which<br />

can recognise non-self structures, the so-called “pattern recognition receptors”<br />

(PRR). One such receptor is Dectin-1, a type II transmembrane glycoprotein with a<br />

single extracellular non-classical C-type carbohydrate recognition domain (CRD)<br />

and a cytoplasmic tail possessing an immunoreceptor tyrosine-based activationlike<br />

(ITAM) motif. Dectin-1 is predominantly expressed on myeloid cells and<br />

recognises (1 3)-linked glucans. We and others have demonstrated that this<br />

receptor mediates a variety of cellular responses to -glucans, including<br />

phagocytosis, endocytosis and the oxidative burst and can induce the production<br />

of arachidonic acid and numerous cytokines and chemokines. These responses<br />

are triggered through the cytoplasmic ITAM-like motif of this receptor, utilising novel<br />

signalling pathways involving a unique interaction with Syk kinase and collaborative<br />

signalling with the TLRs. Dectin-1 is the first example of a signalling non-Toll-like<br />

pattern recognition receptor being involved in the induction of protective immune<br />

responses, and through these activities Dectin-1 plays a fundamental role in anti<strong>fungal</strong><br />

immunity in mice and humans. Here, we will review new insights into the role<br />

of Dectin-1 as well as other PRRs during <strong>fungal</strong> infections.<br />

76


The Tyr238X dectin-1 polymorphism is a predisposing<br />

factor for mucosal <strong>Candida</strong> albicans infections but not<br />

candidemia<br />

Diana C. Rosentul 1,2 , Theo S. Plantinga 1,2 , Marije Oosting 1,2 , William K.<br />

Scott 3 , Digna R. Velez Edwards 4 , P. Brian Smith 5 , Barbara D. Alexander 5 ,<br />

John C. Yang 6 , Gregory M. Laird 5 , Walter J. F. M. van der Velden 2,7 , Bart<br />

Ferwerda 1,2 , Annemiek B. van Spriel 2,8 , Gosse Adema 2,8 , Ton Feuth 2,9 , J.<br />

Peter Donnelly 1,2 , Gordon D. Brown 10 , Nicole M. A. Blijlevens 2,7 , Leo A.B.<br />

Joosten 1,2 , Jos W. M. van der Meer 1,2 , John R. Perfect 5 , Bart-Jan<br />

Kullberg 1,2 , Melissa D. Johnson 5,11 , Mihai G. Netea 1,2<br />

1 Department of Internal Medicine, Radboud University Nijmegen Medical Center, Nijmegen,<br />

The Netherlands; 2 Nijmegen Institute for <strong>Infection</strong>, Inflammation and Immunity (N4i), Radboud<br />

University Nijmegen Medical Center, Nijmegen, The Netherlands; 3 Dr. John T. Macdonald<br />

Foundation Department of Human Genetics and John P. Hussman Institute for Human<br />

Genomics, University of Miami, Miller School of Medicine, Miami, FL, USA; 4 Department of<br />

Obstetrics & Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA; 5 Duke<br />

University Medical Center, Durham, NC, USA; 6 National Jewish Health, Denver, CO, USA;<br />

7 Department of Haematology, Radboud University Nijmegen Medical Center, Nijmegen, The<br />

Netherlands; 8 Department of Tumor Immunology, Radboud University Nijmegen Medical<br />

Center, Nijmegen, The Netherlands; 9 Department of Epidemiology, Biostatistics and Health<br />

Technology Assessment, Radboud University Nijmegen Medical Center, Nijmegen, The<br />

Netherlands; 10 Institute of Infectious Disease and Molecular Medicine, University of Cape<br />

Town, Cape Town, South Africa; 11 Department of Clinical Research, Campbell University<br />

School of Pharmacy, Buies Creek, NC, USA.<br />

The genetic make up of innate immune genes is believed to play an important role<br />

for susceptibility to <strong>fungal</strong> infections. We recently described the Tyr238X dectin-1<br />

single nucleotide polymorphism (SNP) as an important factor influencing<br />

susceptibility to vulvo-vaginal candidiasis and onychomycosis in a Dutch family<br />

with recurrent <strong>fungal</strong> infections.<br />

The aim of our studies was to assess the role of this polymorphism in the<br />

susceptibility to mucosal and systemic <strong>Candida</strong> albicans infections. To achieve this<br />

aim we have investigated the effect of this SNP in several cohorts of patients:<br />

-142 Dutch patients undergoing hematopoietic stem cells transplantation (HSCT).<br />

-331 Dutch and American candidemia patients and 351 non infected matched<br />

controls.<br />

Functional studies have shown that cells isolated from individuals heterozygous for<br />

the 238X allele had a lower cytokine production upon stimulation with <strong>Candida</strong>. The<br />

HSCT patients bearing the early stop variant of the dectin-1 gene presented a<br />

higher C. albicans oral an gastrointestinal colonization compared to those who were<br />

78


wild type for the polymorphism and required higher amounts of fluconazole in order<br />

to avoid a disseminated invasion of the fungus.<br />

In contrast, no effect if the Tyr238X dectin-1 SNP on the susceptibility to candidemia<br />

and/or the clinical course of the infection was observed.<br />

In conclusion, the dectin-1 signaling pathway is non-redundant in mucosal immunity<br />

to C. albicans. The 238X allele results in a defective IL-6 and IL-17 response, but<br />

normal IFN-gamma, upon stimulation with C. albicans.<br />

A genetic deficiency of beta-glucan recognition has clear effect on the occurrence<br />

of mucosal <strong>Candida</strong> infections, but a minor impact on susceptibility to candidemia.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial Training Network<br />

(PITN-GA-2008-214004).<br />

79


C-type lectin receptor signaling in innate anti-<strong>fungal</strong><br />

immunity<br />

Jürgen Ruland<br />

Institute for Molecular Immunology, Technical University Munich, Germany<br />

Dectin-1 was the first C-type lectin receptor (CLRs) identified to be important in<br />

innate anti-<strong>fungal</strong> immunity. This receptor couples to the tyrosine kinase Syk and<br />

thereby triggers its own intra cellular signaling pathways for inflammatory responses<br />

and host defence. Futher CLRs that can activate Syk and which mediate <strong>fungal</strong><br />

recognition are Dectin-2 and Mincle. Here we will discuss new insights into the<br />

molecular mechanisms of CLR signaling and how they relate to anti-<strong>fungal</strong> defence.<br />

80


<strong>Candida</strong> - epithelial interactions and innate immunity<br />

David L Moyes 1 , Celia Murciano 1 , Oliver Bader 2 , Michael Weig 2 , Gary<br />

Moran 3 , Bernhard Hube 4 and Julian R Naglik 1<br />

1 King’s College London Dental Institute, King’s College London, London, UK; 2 University<br />

Medical Centre Göttingen, Kreuzbergring 57, Germany; 3 Trinity College,Dublin 2, Ireland;<br />

4 Hans Knoell Institute, Jena, Germany<br />

Host mechanisms enabling discrimination between commensal and pathogenic<br />

organisms are critical in mucosal immune defense and homeostasis. Of particular<br />

interest are opportunistic pathogens such <strong>Candida</strong> albicans, which can act as both<br />

commensal and pathogen. One of the key virulence factors contributing to C.<br />

albicans pathogenicity at mucosal surfaces is hypha formation and recently we<br />

reported a novel mechanism by which oral epithelial cells discriminate between the<br />

yeast and hyphal forms of C. albicans. Discrimination is mediated via the MAPK<br />

pathway and constitutes activation of the MAPK phosphatase 1 (MKP1) and c-Fos<br />

transcription factor and is targeted against the hyphal form of C. albicans.<br />

MAPK/MKP1/c-Fos activation correlates with proinflammatory responses and cell<br />

damage in a dose-dependent manner, but is independent of TLR2, TLR4 and<br />

Dectin-1 and is not triggered by cell wall polysaccharides (mannan, -glucan, chitin).<br />

However, two fundamentally important issues remain unknown: the identity of the<br />

hyphal moiety targeted by epithelial cells and the epithelial receptor that activates<br />

the discriminatory pathway. To identify the hyphal moiety we screened a panel of<br />

cell wall- and morphology-related C. albicans mutants and assayed for MKP1/c-<br />

Fos activation, induction of cytokines and cell damage. This screen identified a<br />

potential candidate hypha-specific protein that activates the epithelial response.<br />

The respective null mutant remained in the hyphal phase but was unable to activate<br />

MKP1/c-Fos, cytokines or cell damage. To verify this, the hyphal protein was<br />

purified and was shown to directly activate c-Fos, cytokines and cell damage in a<br />

dose-dependent manner. Our data indicate that epithelial cells target this hyphal<br />

protein to initiate innate immunity against C. albicans and we propose that this<br />

epithelial MAPK/c-Fos pathway represents a ‘danger response’ mechanism<br />

informing the host of potentially dangerous levels of C. albicans hyphae, which may<br />

be critical in identifying when this normally opportunistic fungus has become<br />

pathogenic.<br />

82


<strong>Candida</strong> albicans Secreted Aspartic Proteases Cause<br />

Inflammatory Response Irrespective of Their Proteolytic<br />

Activity<br />

Neelam Pandey 1 , Donatella Pietrella 1 , Lydia Schild 2 , Francesco Bistoni 1 ,<br />

Bernhard Hube 2 and Anna Vecchiarelli 1<br />

1 Department of Experimental Medicine and Biochemical Science, University of Perugia,<br />

Perugia, Italy; 2 Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for<br />

Natural Product Research and <strong>Infection</strong> <strong>Biology</strong>, Hans Knoell Institute, Jena, Germany<br />

<strong>Candida</strong> albicans normally inhabits humans as a commensal, but causes fatal infections<br />

when the immune system of patients is suppressed. It has a strong armory of virulence<br />

attributes which facilitates bypassing the immune system and causes mucosal,<br />

cutaneous and systemic infections. These attributes are the ability to colonize, invade<br />

the host tissue and secrete aspartic proteases. We here report that recombinant secreted<br />

aspartic proteases (rSaps) including rSap1, rSap2, rSap3 and rSap6 induce inflammatory<br />

cytokine production by human monocytes to different degrees. rSap1, rSap2 and rSap6<br />

induced the secretion of IL-1beta, TNF-alpha and IL-6 significantly, while rSap3<br />

stimulated the secretion of IL-1beta and TNF-alpha. All these rSaps induced Ca 2+ influx<br />

in monocytes. Pepstatin A, a potential inhibitor of aspartic proteases, has no effect on<br />

the cytokine secretion induced by these rSaps. This suggests that the inflammatory<br />

response due to rSaps is not because of their proteolytic activity, a hypothesis further<br />

supported by the observation that the ability of rSaps to induce inflammatory cytokine<br />

secretion was independent of protease-activated receptor (PAR) activation, and of the<br />

optimal pH for individual rSap activity. Moreover, rSaps stimulated Akt activation and<br />

thus induced IkB-alpha phosphorylation which mediated translocation of NFkB into the<br />

nucleus in human monocytes. These findings give evidence that rSap1, rSap2 and rSap3<br />

cause an inflammatory response irrespective of their proteolytic activity and mediate the<br />

inflammatory response via activation of Akt/NF-kB. The inflammatory response<br />

terminates with inflammasome activation. This occurs through a process requiring Sap<br />

internalization via a clathrin dependent mechanism, and caspase-1 activation, and it is<br />

mediated by K + efflux and ROS production. By demonstrating that the recognized<br />

virulence factors of C. albicans such as Sap2 and Sap6 are potent inducers of<br />

inflammasome activation, our results represent a step forward in the comprehension of<br />

the close relationship between fungus virulence and host response mechanisms.<br />

We are extending our study by using an in vivo experimental model of murine candidiasis<br />

to analyze the role of Sap2 and Sap6 in the inflammatory response in the vaginal<br />

candidiasis, and to investigate whether specific mAbs are indeed able to affect this<br />

process.<br />

This study was funded by the Eurpoean Commission through the <strong>FINSysB</strong> Marie Curie Initial Training<br />

Network (PITN-GA-2008-214004).<br />

84


A central role of complement for the <strong>Candida</strong> albicansinduced<br />

cytokine production by human cells<br />

Shih-Chin Cheng 1,2 , Tom Sprong 1,2 , Leo A.B. Joosten 1,2 , Jos W.M. van der<br />

Meer 1,2 , Bart-Jan Kullberg 1,2 , Bernhard Hube 3,4 , Lone Schejbel 5 , Peter<br />

Garred 5 Marcel van Deuren 1,2 , Mihai G. Netea 1,2<br />

1 Departments of Medicine, Radboud University Nijmegen Medical Center, Nijmegen, the<br />

Netherlands; 2 Nijmegen Institute for <strong>Infection</strong>, Inflammation and Immunity (N4i), Nijmegen,<br />

the Netherlands; 3 Department of Microbial Pathogenicity Mechanism, Leibniz Institute for<br />

Natural Product Research and <strong>Infection</strong> <strong>Biology</strong> - Hans Knoell Institute, Jena, Germany;<br />

4 Friedrich Schiller University, Jena, Germany; 5 Department of Clinical Immunology, Laboratory<br />

of Molecular Medicine, Rigshospitalet, Copenhagen, Denmark.<br />

In experimental studies, the role of complement for anti<strong>fungal</strong> host defense has<br />

been attributed to its ability of opsonization. In this study we report that in humans<br />

an activated complement system mainly augments <strong>Candida</strong> albicans-induced host<br />

pro-inflammatory cytokine production via C5a-C5a receptor signaling, while<br />

<strong>Candida</strong> phagocytosis and intracellular killing is not influenced. By blocking the<br />

C5a-C5aR signaling pathway either with anti-C5a antagonist antibodies or with the<br />

C5aR antagonist W-54001, C. albicans-induced IL-6 and IL-1 levels were<br />

significantly reduced. Recombinant C5a augmented cytokine production. In<br />

addition, using serum from patients with various complement deficiencies, we<br />

demonstrate a crucial role of C5, but not C6 or the membrane attack complex, for<br />

C. albicans-induced IL-6 and IL-1 in monocytes. These findings reveal a central<br />

role of anaphylatoxin C5a in augmenting host pro-inflammatory cytokine production<br />

upon contact with C. albicans, and defines the role of the complement system for<br />

anti-<strong>Candida</strong> host defense in humans.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial Training Network<br />

(PITN-GA-2008-214004).<br />

86


The role of pH-regulated antigen 1 of <strong>Candida</strong> albicans in<br />

the interaction of the fungus with human neutrophils<br />

Eliška Svobodová 1 , Josephine Losse 1 , Antje Heyken 2 , Bernhard Hube 2,4 ,<br />

Peter F. Zipfel 3,4 and Mihály Józsi 1<br />

1 Junior Research Group Cellular Immunobiology, 2 Department of Microbial Pathogenicity<br />

Mechanisms and 3 Department of <strong>Infection</strong> <strong>Biology</strong>, Leibniz Institute for Natural Product<br />

Research and <strong>Infection</strong> <strong>Biology</strong> - Hans Knöll Institute, Jena, Germany; 4 Friedrich Schiller<br />

University, Jena, Germany<br />

<strong>Candida</strong> albicans is an opportunistic human-pathogenic yeast and a leading cause<br />

of life-threatening systemic <strong>fungal</strong> infections in immunocompromised individuals.<br />

The virulence of the fungus is associated with its capability to filamentous growth.<br />

The pH-regulated antigen 1 (Pra1) of C. albicans is a surface-associated and<br />

secreted protein whose expression is upregulated in the hyphal form. Pra1 binds<br />

to complement receptor 3 (CD11b/CD18) and can mediate adhesion to and<br />

migration of human phagocytes. Here, we investigated the role of Pra1 in the<br />

activation of human neutrophils. A C. albicans mutant strain lacking Pra1 (pra1 )<br />

supported neutrophil migration to a lower extent than did the parental wild-type<br />

strain. A Pra1-overexpressing C. albicans strain enhanced neutrophil migration and<br />

adherence. While inactivated hyphae of the Pra1-overexpressing mutant enhanced<br />

the production and release of reactive oxygen species, myeloperoxidase,<br />

lactoferrin, and interleukin 8 by neutrophils, such responses were reduced when<br />

stimulated with the pra1 strain. Pra1-overexpressing living hyphae also caused a<br />

reduced neutrophil activation, indicating that <strong>fungal</strong> cells which express and release<br />

more Pra1 can more efficiently inhibit the activation of these innate immune cells.<br />

Fungal cells lacking Pra1 were more efficiently killed by neutrophils. The lack or the<br />

overexpression of Pra1 did not significantly affect the generation of neutrophil<br />

extracellular traps. In conclusion, surface-exposed Pra1 plays a role in the<br />

recognition of C. albicans, especially hyphal cells, by human neutrophils and<br />

enhances neutrophil antimicrobial responses. However, the fungus can counteract<br />

some of these defense mechanisms likely by releasing Pra1.<br />

88


Vaccines against opportunistic <strong>fungal</strong> infections<br />

Antonio Cassone<br />

Department of Infectious, Parasitic and Immuno-mediated Diseases, Istituto Superiore di<br />

Sanità, Rome, Italy.<br />

The rising threat represented by opportunistic <strong>fungal</strong> infections, the scarcity/high<br />

cost of effective anti<strong>fungal</strong> treatments in the setting of the immunocompromised<br />

host, the advances in the knowledge of <strong>fungal</strong> pathogenicity and immune responses<br />

are the main driving factors to recent studies aimed at generating active and passive<br />

vaccination tools against human pathogenic fungi. Two vaccines, one specifically<br />

designed to fight chronic recurrent mucosal candidiasis and another one for more<br />

general anti-<strong>Candida</strong> prevention/therapy, are in clinical trials in Europe and US.<br />

A third vaccine, made up by an algal -glucan (laminarin) conjugated with a protein<br />

component (Lam-CRM vaccine) has been formulated to enter clinical investigation.<br />

This vaccine has been shown to protect against experimental infections by the three<br />

major opportunistic fungi such as <strong>Candida</strong> albicans, Aspergillus fumigatus and<br />

Cryptococcus neoformans and has been therefore designated as pan-<strong>fungal</strong> or<br />

“universal” vaccine. Protection appears to be largely if not exclusively mediated by<br />

anti- -glucan antibodies with restricted specificity to 1-3 linked epitope and<br />

capable to inhibit growth and virulence-associated <strong>fungal</strong> factors. Protective anti-<br />

1-3 glucan monoclonal murine and human-chimeric antibodies have also been<br />

generated thus opening perspectives for passive vaccination in<br />

immunocompromized subjects.<br />

Our research demonstrates the possibility of conveying into a single immunological<br />

tool the potential to protect against multiple <strong>fungal</strong> infections. The above approach<br />

could be theoretically extended to non-<strong>fungal</strong> infections by selecting the<br />

appropriate molecular pattern shared by a given microbial group (e.g. peptidoglycan<br />

for Gram-positive bacteria). Noteworthy, the molecular patterns are those highly<br />

conserved pathogen-associated molecules which foster innate immunity through<br />

their binding to the pattern-recognition structures and intracellular signalling, hence<br />

shaping adaptive immune responses. Single-component, molecular pattern-based<br />

vaccines would merge the broad target range typical of innate immunity with the<br />

highly focussed specificity of the adaptive immunity.<br />

90


Tackling the <strong>Candida</strong> spp infection spectrum with first-inclass<br />

antimicrobial peptide technology<br />

Dr Deborah A O’Neil<br />

NovaBiotics Ltd, Cruickshank Building, Aberdeen, AB21 9TR, UK<br />

Applying a biology-led, rational drug design approach, NovaBiotics has developed<br />

patented antimicrobial peptide technology from which Novamycin ® , a potently<br />

fungicidal API has been derived. Novamycin ® has a novel, membranolytic mode of<br />

action against which acquired resistance cannot develop in target <strong>fungal</strong> pathogens<br />

and is equally active against metabolically active and non-metabolising fungi.<br />

Novamycin ® is active against all <strong>Candida</strong> spp, Cryptococcus spp and limited<br />

Aspergillus spp. Novamycin ® demonstrates efficacy and is well tolerated in wellestablished,<br />

clinically relevant in vivo models of <strong>Candida</strong> spp sepsis (neutropenic<br />

and immunocompetant models), oral pharyngeal candidiasis and vaginal<br />

candidiasis. Novamycin ® will enter the clinical phase of development in the very<br />

near future for topical and mucosal indications whilst in parallel, formulation and<br />

delivery for systemic application in sepsis is further optimised. Novamycin ® offers<br />

a novel first-in-class API solution not only to the whole spectrum of <strong>Candida</strong> spp.<br />

infections, but to those caused by other clinically relevant yeasts and moulds.<br />

92


Wall proteins, absolute quantification and concatenated<br />

vaccines: A new approach for <strong>Candida</strong> albicans therapy?<br />

Clemens J. Heilmann, Alice G. Sorgo, Henk L. Dekker, Stanley Brul,<br />

Chris G. de Koster, Leo J. de Koning and Frans M. Klis<br />

Swammerdam Institute for Life Sciences, Universiteit van Amsterdam, Science Park 904,<br />

1098XH Amsterdam<br />

The cell wall and especially the proteins that are covalently anchored to it are the<br />

first site of host-pathogen interaction. The wall proteins are promising targets for<br />

the development of diagnostic biomarkers and vaccines. Previously, we described<br />

the relative quantitative changes in up to 21 wall proteins during the yeast-to-hypha<br />

transition (1) and upon exposure of cells to fluconazole (2). We identified hyphaassociated<br />

wall proteins (Als3, Hwp2, Hyr1, Plb5, Sod5) conferring increased<br />

adhesion and resistance to host defenses and yeast-associated proteins (Rhd3,<br />

Sod4, Ywp1), which probably play a role in host dispersal and immune evasion.<br />

Morphotype-independent proteins (Cht2, Crh11, Ecm33, Mp65) are mainly involved<br />

in wall structure and remodeling.<br />

However, relative quantification can only provide indications of how protein<br />

abundance changes in response to an environmental change. To compare the<br />

abundances of different proteins, absolute quantification is required. We are<br />

developing a strategy for absolute quantification based on 15 N metabolic labeling<br />

and concatenated proteins (Q-CON-CAT). By using the same 15 N reference culture<br />

that was also used for relative quantification and determining the absolute amount<br />

of a particular protein, we will be able to recalculate the relative quantification results<br />

into absolute values.<br />

These absolute values will be informative for the construction of peptide-based<br />

vaccines. We propose that by concatenating peptides that are predicted to be<br />

highly immunogenic and originate from abundant wall proteins an effective Q-CON-<br />

CAT vaccine protein can be generated.<br />

(1) Hyphal Induction in the human <strong>fungal</strong> pathogen <strong>Candida</strong> albicans reveals a characteristic wall protein<br />

profile. Heilmann CJ, Sorgo AG, Siliakus AR, Dekker HL, Brul S, de Koster CG, de Koning LJ, Klis FM.<br />

Microbiology. 2011 May 20.<br />

(2) The effects of fluconazole on the secretome, the wall proteome and wall integrity of the clinical fungus<br />

<strong>Candida</strong> albicans. Sorgo AG, Heilmann CJ, Dekker HL, Bekker M, Brul S, de Koster CG, de Koning LJ,<br />

Klis FM.Eukaryot Cell. 2011 May 27.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial Training Network<br />

(PITN-GA-2008-214004).<br />

94


scFv Phage Display Library against Complex<br />

Immunomodulatory Glycans<br />

Abhishek Saxena 1 , Soumya Palliyil 1,2 , Keith A. Charlton 3 , Alistair J.P.<br />

Brown 1 & Andrew J.R. Porter 1,2,3<br />

1 Department of Molecular and Cell <strong>Biology</strong>, Institute of Medical Sciences, Aberdeen Medical<br />

School, University of Aberdeen, Aberdeen AB25 2ZD, UK; 2 Scottish Biologics Facility,<br />

University of Aberdeen, Aberdeen AB25 2ZD, UK; 3 ImmunoSolv Ltd, Liberty Research centre,<br />

University of Aberdeen, Aberdeen AB25 2ZP, UK<br />

The concept of Antibody therapy against infectious diseases is still in its infancy.<br />

Numerous studies in last two decades have focused on anti-glycan antibodies that<br />

recognize <strong>fungal</strong> cell surface. The presence of these antibodies in blood has been<br />

used to diagnose <strong>fungal</strong> infection. <strong>Candida</strong> albicans cell wall -glucan and -mannan<br />

are very well established diagnostic and therapeutic targets because they represent<br />

the most abundant cell surface epitopes responsible for generation of unique<br />

antibodies against fungus. We have constructed a scFv M13 phage display library<br />

against complex glycans of <strong>Candida</strong> albicans. To generate an antibody response<br />

and construct a library, a Welch breed/Suffolk sheep was hyper-immunized (IgG<br />

titres > 60,000) against a <strong>Candida</strong> albicans hyphal cell wall preparation. Binding<br />

analysis of crude/purified polyclonal serum IgG indicates that cell wall glycans are<br />

indeed primary targets because they dominate the immune response of the sheep.<br />

We further demonstrated that polyclonal IgG has in vitro hyphae neutralization<br />

potential in growth inhibition assay. A scFv library has been constructed by cloning<br />

the immunoglobulin variable gene repertoire of peripheral blood lymphocytes<br />

isolated from this hyper-immune animal. Variable heavy and variable light gene<br />

segments of immunoglobulins were PCR amplified and linked through a cellulase<br />

linker. Linked scFv segments were cloned into the phagemid vector pHEN2a. The<br />

size of the VH-V and VH-V library in E.coli TG1 cells was 3.8 x 10 10 and 1.1 x 10 8<br />

respectively. Repertoire diversity was confirmed by sequencing random clones<br />

(approximately 45 clones) and CDRs of VH and V /V genes were found to be 100%<br />

different. We have a panel of early monoclonal hits against both CA -glucan and<br />

-mannan antigens. We are currently interested in pulling monoclonal phages from<br />

library against variety of immunomodulatory glycans.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial Training Network<br />

(PITN-GA-2008-214004).<br />

96


Essential Genes as Broad-Spectrum Anti<strong>fungal</strong> Targets<br />

K S Dobb 1 , M Birch 1 , M J Bromley 2 , S Kaye 1 , L Stateva 3 , J D Oliver 1<br />

1 F2G Ltd, Lankro Way, Manchester, UK. 2 Manchester Academic Health Centre,<br />

The University of Manchester UK. 3 Faculty of Life Sciences, The University of<br />

Manchester, UK<br />

Aspergillus fumigatus and <strong>Candida</strong> albicans both cause serious, systemic disease<br />

in vulnerable patients. Current anti<strong>fungal</strong> drugs have many limitations and there is<br />

a need for new classes of anti<strong>fungal</strong> drugs. This study aims to investigate genes<br />

essential for the growth of A. fumigatus and C. albicans as anti<strong>fungal</strong> targets.<br />

In silico analysis of a set of essential genes from A. fumigatus revealed those with<br />

C. albicans homologs. To assess selectivity those with human homologs were also<br />

identified. A subset of the A. fumigatus essential gene set was chosen for<br />

essentiality studies in C. albicans.<br />

One gene found to be essential in both A. fumigatus and C. albicans was<br />

Phosphopantetheinyl transferase b (PPTb). This gene is involved in mitochondrial<br />

fatty acid synthesis and catalyses the transfer of a phosphopantetheine group from<br />

coenzyme A (CoA) to acyl carrier protein (ACP). To identify inhibitors of Pptb a<br />

fluorescence polarisation (FP) assay was developed. The transfer of fluorophore<br />

labelled phosphopantetheine group from CoA to ACP can be measured by a<br />

change in FP value. As Pptb catalyses this transfer inhibition of this enzyme can<br />

also be detected by a change in FP values. A statistically robust FP assay was<br />

achieved using C. albicans ACP and proteins and fluorescently labelled CoA. Single<br />

concentration and IC50 screening of a small molecule compound library has<br />

identified inhibitors of Pptb. Other work on these “hits” has included anti<strong>fungal</strong><br />

susceptibility testing (minimum inhibitory concentration, MIC) and mammalian cell<br />

toxicology testing (MCTs). PPTb has so far shown good potential as a broadspectrum<br />

anti<strong>fungal</strong> target and future work will aim to further characterise these<br />

small molecule inhibitors for their potential as drug leads.<br />

98


Hyphal development in <strong>Candida</strong> albicans requires two<br />

temporally linked changes in promoter chromatin for<br />

initiation and maintenance<br />

Yang Lu, Chang Su, Allen Wang and Haoping Liu<br />

Department of Biological Chemistry, University of California, Irvine, CA 92697, USA.<br />

Phenotypic plasticity is common in development. For <strong>Candida</strong> albicans, the most<br />

common cause of invasive <strong>fungal</strong> infections in humans, morphological plasticity is<br />

its defining feature and is critical for its pathogenesis. Unlike other <strong>fungal</strong> pathogens<br />

that exist primarily in either yeast or hyphal forms, C. albicans is able to switch<br />

reversibly between yeast and hyphal growth forms in response to environmental<br />

cues. Although many regulators have been found involved in hyphal development,<br />

the mechanisms of regulating hyphal development and plasticity of dimorphism<br />

remain unclear. Here we show that hyphal development involves two sequential<br />

regulations of the promoter chromatin of hypha-specific genes. Initiation requires a<br />

rapid but temporary disappearance of the Nrg1 transcriptional repressor of hyphal<br />

morphogenesis via activation of the cAMP-PKA pathway. Maintenance requires<br />

promoter recruitment of Hda1 histone deacetylase under reduced Tor1 (target of<br />

rapamycin) signalling. Hda1 deacetylates a subunit of the NuA4 histone<br />

acetyltransferase module, leading to eviction of the NuA4 acetyltransferase module<br />

and blockage of Nrg1 access to promoters of hypha-specific genes. Promoter<br />

recruitment of Hda1 for hyphal maintenance happens only during the period when<br />

Nrg1 is gone. The sequential regulation of hyphal development by the activation of<br />

the cAMP-PKA pathway and reduced Tor1 signalling provides a molecular<br />

mechanism for plasticity of dimorphism and how C. albicans adapts to the varied<br />

host environments in pathogenesis. Such temporally linked regulation of promoter<br />

chromatin by different signalling pathways provides a unique mechanism for<br />

integrating multiple signals during development and cell fate specification.<br />

100


101


The transcription factor Sko1 represses the yeast-tohypha<br />

transition and mediates the oxidative stress<br />

response in <strong>Candida</strong> albicans.<br />

Inês Correia, Elvira Román, David M. Arana, Daniel Prieto, Verónica<br />

Urrialde, César Nombela, Rebeca Alonso-Monge and Jesús Pla. §<br />

Departamento de Microbiología II. Facultad de Farmacia. Universidad Complutense de<br />

Madrid. Plaza de Ramón y Cajal s/n. E-28040 MADRID. SPAIN<br />

Cells adapt to external changes by triggering transcriptional responses. These<br />

responses are complex and are mediated by different transcription factors. Mutants<br />

lacking the transcription factor Sko1 were generated in <strong>Candida</strong> albicans and its<br />

roles on the physiology of this opportunistic pathogen are reported. Genome-wide<br />

transcriptional analysis revealed that Sko1 acts as a transcriptional repressor of<br />

genes involved in pathogenesis and hyphal formation. The sko1 mutant displayed<br />

an increased expression of the hyphal related genes ECE1 and HWP1 but showed<br />

no drastic alterations in the mouse model of systemic infection. Deletion of SKO1<br />

rendered mutants derepressed in the yeast-to-hypha transition. Moreover, Sko1<br />

was shown to be involved in the response to oxidative stress and sko1 mutants<br />

increased the sensitivity of hog1 mutants to the myelomonocytic cell line HL-60.<br />

Alterations in the phosphorylation pattern of MAP kinase mutants in response to<br />

oxidative challenge were also observed. Genome-wide transcriptional analysis after<br />

hydrogen peroxide treatment revealed that sko1 mutants were able to generate an<br />

adaptive response similar to wild type strains, although important differences were<br />

detected in the magnitude of such transcriptional response. Collectively, these<br />

results implicate Sko1 as a key mediator of the oxidative stress response and<br />

morphological transition in C. albicans.<br />

102


103


Calcineurin Signaling and Membrane Lipid Homeostasis<br />

Regulates Iron Mediated MultiDrug Resistance Mechanisms<br />

in <strong>Candida</strong> albicans<br />

Saif Hameed, Sanjiveeni Dhamgaye, Ashutosh Singh, Shyamal K. Goswami<br />

and Rajendra Prasad<br />

School of Life Sciences, Jawaharlal Nehru University, New Delhi-110067, India<br />

We previously demonstrated that iron deprivation enhances drug susceptibility of<br />

<strong>Candida</strong> albicans by increasing membrane fluidity which correlated with the lower<br />

expression of ERG11 transcript and ergosterol levels. The iron restriction dependent<br />

membrane perturbations led to an increase in passive diffusion and drug<br />

susceptibility. The mechanisms underlying iron homeostasis and multidrug<br />

resistance (MDR), however, are not yet resolved. To evaluate the potential<br />

mechanisms, we used whole genome transcriptome and electrospray ionization<br />

tandem mass spectrometry (ESI-MS/MS) based lipidome analyses of iron deprived<br />

<strong>Candida</strong> cells to examine the new cellular circuitry of the MDR of this pathogen.<br />

Our transcriptome data revealed a link between calcineurin signaling and iron<br />

homeostasis. Among the several categories of iron deprivation responsive genes,<br />

the down regulation of calcineurin signaling genes including HSP90, CMP1 and<br />

CRZ1 was noteworthy. Interestingly, iron deprived <strong>Candida</strong> cells as well as iron<br />

acquisition defective mutants phenocopied molecular chaperone HSP90 and<br />

calcineurin mutants and thus were sensitive to alkaline pH, salinity and membrane<br />

perturbations. In contrast, sensitivity to above stresses did not change in iron<br />

deprived DSY2146 strain with a hyperactive allele of calcineurin. Although, iron<br />

deprivation phenocopied compromised HSP90 and calcineurin, it was independent<br />

of protein kinase C signaling cascade. Notably, the phenotypes associated with<br />

iron deprivation in genetically impaired calcineurin and HSP90 could be reversed<br />

with iron supplementation. The observed down regulation of ergosterol (ERG1,<br />

ERG2, ERG11 and ERG25) and sphingolipid biosynthesis (AUR1 and SCS7) genes<br />

followed by lipidome analysis confirmed that iron deprivation not only disrupted<br />

ergosterol biosynthesis, but it also affected sphingolipid homeostasis in <strong>Candida</strong><br />

cells. These lipid compositional changes suggested extensive remodeling of the<br />

membranes in iron deprived <strong>Candida</strong> cells. Taken together, our data provide the<br />

first novel insight into the intricate relationship between cellular iron, calcineurin<br />

signaling, membrane lipid homeostasis and drug susceptibility of <strong>Candida</strong> cells.<br />

104


105


Functional analysis of H-loop residues of Cdr1p, an ABC<br />

transporter of human <strong>fungal</strong> pathogen <strong>Candida</strong> albicans<br />

Antresh Kumar 1 and Rajendra Prasad 1<br />

1 School of Life Sciences, Jawaharlal Nehru University, New Delhi, INDIA<br />

Nucleotide Binding Domains (NBDs) of multidrug transporter of <strong>Candida</strong> albicans,<br />

Cdr1p possess unique divergent amino acids in their conserved motifs. For<br />

example, NBD1 possesses divergent H-loop (IYQ) while the same motif is<br />

conserved (IHQ) in NBD2.<br />

We report here the contribution of these conserved and divergent H-loop motifs of<br />

Cdr1p in ATP catalysis and drug transport. For this, we mutagenized two residues<br />

of divergent (IYQ) and conserved H-loop region (IHQ) either by replacing them with<br />

alanines or replacing residues with equiposition residues of another H-loop. These<br />

mutations were introduced into GFP-tagged Cdr1p which was stably overexpressed<br />

at the PDR5 locus in a heterologous host S. cerevisiae mutant strain, AD1-8u - which<br />

lacks seven major ABC transporters.<br />

All the Cdr1p mutant variants of H-loop region were properly expressed and<br />

localized to the cell surface similar to wild-type protein. Alanine mutants of H-loop<br />

region, Y361A in NBD1 has no effect on Cdr1p function and showed normal R6G<br />

transport and ATPase activity whereas corresponding mutation (H1059A) in NBD2<br />

abolished R6G transport without any significant loss of ATPase activity. Moreover,<br />

cells expressing mutation like Q362A severely abrogated Cdr1p function while<br />

Q1060A mutation in NBD2 showed no effect on Cdr1p function and measured<br />

normal transport and ATPase activity similar to wild type. Though, Arginine<br />

substitution of glutamine residue present in H-loop region of Cdr1p (Q362R and<br />

Q1060R) displayed deferential effect on Cdr1 transport. These results suggest that<br />

not only the conserved residue like H1059 but divergent Q362 is also crucial for<br />

Cdr1p function.<br />

106


107


108


POSTER ABSTRACTS<br />

109


110


The Pathogenic Armoury<br />

111


Poster number: 01<br />

Glucose promotes oxidative stress resistance in <strong>Candida</strong><br />

albicans via specific signalling pathways<br />

Iryna Bohovych 1 , Pedro Miramón 2 , Aaron Mitchell 3 , Bernhard Hube 2 ,<br />

Alistair JP Brown 1<br />

1 Aberdeen Fungal Group, School of Medical Sciences, University of Aberdeen, Foresterhill,<br />

Aberdeen AB25 2ZD, UK; 2 Microbial Pathogenicity, Hans Knoell Institute, Beutenbergstraße<br />

11a, Jena 07745, Germany; 3 Department of Biological Sciences, Carnegie Mellon University,<br />

Pittsburgh, Pennsylvania 15213, USA<br />

<strong>Candida</strong> albicans, a successful human pathogen, displays the phenomenon of<br />

glucose-enhanced oxidative stress resistance (Rodaki et al., 2009), which is not<br />

observed in other yeast species tested. The molecular mechanisms that mediate<br />

glucose-enhanced oxidative stress resistance in C. albicans are not clear. We<br />

reasoned that glucose signalling might play a major role. Therefore we tested the<br />

impact of specific C. albicans mutations to determine which of known signalling<br />

pathways are required for glucose-enhanced oxidative stress resistance.<br />

The Sugar Receptor-Repressor (Hgt4-Rgt1) pathway, homologous to the Rgt2/Snf3<br />

pathway in S. cerevisiae, is not required for the phenotype. In contrast, a snf1<br />

mutant, which lacks a key component of the Glucose Repression Pathway,<br />

displayed high oxidative stress resistance on lactate, not altered by glucose. A kis1<br />

mutant, lacking one of the subunits of Snf1 complex, displayed a related phenotype.<br />

The data indicate that in the absence of glucose the Glucose Repression Pathway<br />

represses glucose-enhanced oxidative stress resistance.<br />

cAMP signalling also appears to play a role in glucose-enhanced oxidative stress<br />

resistance. This phenotype was repressed by exogenous dibutyryl cAMP and by a<br />

pde2 mutation that is thought to increase intracellular cAMP levels. Also the<br />

phenotype was enhanced by inactivation of adenylyl cyclase (cyr1).<br />

Having shown that both cAMP signalling and the Glucose Repression Pathway play<br />

important roles in mediating glucose-enhanced stress resistance, the next step was<br />

to identify targets of these pathways that might contribute to the phenotype.<br />

Comparative analyses of transcriptomic profiles of C. albicans glucose- and lactategrown<br />

cells in response to oxidative stress and glucose treatment correspondingly<br />

revealed a small set of commonly up-regu lated genes (Enjalbert et al., 2006; Rodaki<br />

et al., 2009). The impact of the cAMP and Glucose Repression pathways on these<br />

genes is being determined.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial Training Network<br />

(PITN-GA-2008-214004).<br />

112


113


Poster number: 02<br />

The Osmotic Stress Response of <strong>Candida</strong> glabrata<br />

Emily Cook, Ken Haynes<br />

Exeter University, Biosciences- Geoffrey Pope Building, Stocker Rd. Exeter, EX4 4QD<br />

The osmotic stress response has been characterized in great detail in the model<br />

yeast Saccharomyces cerevisiae however less is known in the opportunistic<br />

pathogen <strong>Candida</strong> glabrata. We have approached our goal of better defining the<br />

osmotic stress response in C. glabrata by using the current understanding in S.<br />

cerevisiae as a framework.<br />

Multiple differences between the described S. cerevisiae response and C. glabrata<br />

were observed. The overall osmotic stress tolerance of C. glabrata was higher than<br />

that of S. cerevisiae, as demonstrated by growth curves and plating assays. Quite<br />

interestingly, we have observed that while C. glabrata did not grow under the highest<br />

concentration of osmotic stress tested (2M NaCl) most cells remained viable when<br />

transferred to normal growth media. We have measured change in cell size of C.<br />

glabrata and S. cerevisiae and found that while S. cerevisiae decreased in size under<br />

increasing concentrations of NaCl, C. glabrata increased in size by ≈10% under all<br />

tested osmotic stress conditions. We are currently investigating biophysical<br />

differences that could explain these results and their contribution to overall osmotic<br />

stress tolerance. Glycerol production was found to be Hog1 independent as<br />

comparable glycerol levels were measured by enzymatic assay in both wild-type<br />

C. glabrata and a hog1 mutant. Differences in osmo-tolerance of single null mutants<br />

in the Hog1 pathway suggest that a Hog1 independent response may be more<br />

important in driving osmotic stress adaptation.<br />

The osmotic stress response in C. glabrata is markedly different than S. cerevisiae,<br />

highlighting the importance of further characterization of stress responses in<br />

pathogenic species, even when closely related to model organisms. We are further<br />

examining the novel mechanisms of osmotic stress adaptation in C. glabrata and<br />

their potential contributions to pathogenicity.<br />

114


115


Poster number: 03<br />

The transcription factor Sko1 represses the yeast-tohypha<br />

transition and mediates the oxidative stress<br />

response in <strong>Candida</strong> albicans.<br />

Inês Correia, Elvira Román, David M. Arana, Daniel Prieto, Verónica<br />

Urrialde, César Nombela, Rebeca Alonso-Monge and Jesús Pla. §<br />

Departamento de Microbiología II. Facultad de Farmacia. Universidad Complutense de<br />

Madrid. Plaza de Ramón y Cajal s/n. E-28040 MADRID. SPAIN<br />

Cells adapt to external changes by triggering transcriptional responses. These<br />

responses are complex and are mediated by different transcription factors. Mutants<br />

lacking the transcription factor Sko1 were generated in <strong>Candida</strong> albicans and its<br />

roles on the physiology of this opportunistic pathogen are reported. Genome-wide<br />

transcriptional analysis revealed that Sko1 acts as a transcriptional repressor of<br />

genes involved in pathogenesis and hyphal formation. The sko1 mutant displayed<br />

an increased expression of the hyphal related genes ECE1 and HWP1 but showed<br />

no drastic alterations in the mouse model of systemic infection. Deletion of SKO1<br />

rendered mutants derepressed in the yeast-to-hypha transition. Moreover, Sko1<br />

was shown to be involved in the response to oxidative stress and sko1 mutants<br />

increased the sensitivity of hog1 mutants to the myelomonocytic cell line HL-60.<br />

Alterations in the phosphorylation pattern of MAP kinase mutants in response to<br />

oxidative challenge were also observed. Genome-wide transcriptional analysis after<br />

hydrogen peroxide treatment revealed that sko1 mutants were able to generate an<br />

adaptive response similar to wild type strains, although important differences were<br />

detected in the magnitude of such transcriptional response. Collectively, these<br />

results implicate Sko1 as a key mediator of the oxidative stress response and<br />

morphological transition in C. albicans.<br />

116


117


Poster number: 04<br />

Co-infection of an in vitro oral epithelium by <strong>Candida</strong><br />

glabrata and <strong>Candida</strong> albicans<br />

Sónia Silva 1 , Mariana Henriques 1 , Rosário Oliveira 1 , David W Williams 2 ,<br />

Joana Azeredo 1<br />

1 Institute for Biotechnology and Bioengineering, Universidade do Minho, Campus de Gualtar<br />

4710-057, Braga, Portugal; 2 Tissue Engineering and Reparative Dentistry, School of Dentistry,<br />

Cardiff University, Heath Park, Cardiff, CF14 4XY, UK<br />

<strong>Candida</strong> albicans is regarded as the leading of cause of candidosis. However,<br />

<strong>Candida</strong> glabrata has emerged as an important pathogen of oral mucosa, occurring<br />

both singly or in mixed species infections, often with C. albicans. Compared with<br />

C. albicans, little is known about the role of C. glabrata in oral infection. Thus, this<br />

work aimed to examine single and mixed species infection of oral epithelium<br />

involving C. glabrata, and to establish its ability to colonize, invade and damage an<br />

oral epithelium.<br />

A reconstituted human oral epithelium (RHOE) was infected with C. glabrata (n=7),<br />

or with each C. glabrata plus one C. albicans oral isolate. The ability of both species<br />

to colonize and invade an oral epithelium was examined by confocal laser scanning<br />

electron microscopy (CLSM) using species-specific peptide nucleic acid (PNA)<br />

probe for distinguishing C. glabrata of C. albicans cells. Qualitative oral epithelial<br />

damage was assessed by determination of lactate dehydrogenase (LDH) released<br />

by the oral epithelium cells.<br />

This work revealed that all C. glabrata strains were able to colonise the RHOE,<br />

however, in a strain dependent manner. <strong>Candida</strong> glabrata single infection after 12<br />

h, revealed generally no invasion of the RHOE, which contrasted with extensive<br />

tissue invasion demonstrated by C. albicans. Mixed infection showed that C.<br />

albicans enhanced the invasiveness of C. glabrata, and led to increased LDH release<br />

by the RHOE, which confirms the results observed by histological studies.<br />

Summarizing, the results suggest that there is an enhanced invasion and increased<br />

tissue damage caused by mixed C. glabrata and C. albicans infections, which has<br />

important clinical significance and highlights the need to identify <strong>Candida</strong> species<br />

involved in oral candidosis.<br />

118


119


Poster number: 05<br />

The N-terminal part of Als1 protein from <strong>Candida</strong> albicans<br />

specifically binds fucose-containing glycans<br />

Dagmara S. Donohue, Francesco S. Ielasi, Katty V. Y. Goossens and<br />

Ronnie G. Willaert<br />

Laboratory of Structural <strong>Biology</strong>, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels,<br />

Belgium.<br />

The opportunistic pathogen <strong>Candida</strong> albicans expresses on its surface Als<br />

(Agglutinin like sequence) proteins, which play an important role in the adhesion to<br />

host cells and in the development of candidiasis. The binding specificity of these<br />

proteins is broad, as they can bind to various mammalian proteins,such as<br />

extracellular matrix proteins, and N- and E-cadherins. The N-terminal part of Als<br />

proteins constitutes the substrate-specific binding domain and is responsible for<br />

attachment to epithelial and endothelial cells. We have used glycan array screening<br />

to identify possible glycan receptors for the binding domain of Als1p-N. Under those<br />

conditions, Als1p-N binds specifically to fucose-containing glycans, which adds a<br />

lectin function to the functional diversity of the Als1 protein. The binding between<br />

Als1p-N and BSA-fucose glycoconjugate was quantitatively characterized using<br />

surface plasmon resonance, which demonstrated a weak millimolar affinity between<br />

Als1p-N and fucose. Furthermore, we have also quantified the affinity of Als1p-N<br />

to the extracellular matrix proteins proteins fibronectin and laminin, which is situated<br />

in the micromolar range. Surface plasmon resonance characterization of Als1p-N–<br />

Als1p-N interaction was in the micromolar affinity range.<br />

120


121


Poster number: 06<br />

Impact of non-fermentative growth on the C. albicans cell<br />

wall and dynamic responses to osmotic stress<br />

Iuliana V. Ene, Alistair J. P. Brown<br />

Aberdeen Fungal Group, School of Medical Sciences, University of Aberdeen, Foresterhill,<br />

Aberdeen AB25 2ZD, UK<br />

<strong>Candida</strong> albicans displays considerable metabolic flexibility and robust stress<br />

responses in vivo. Both contribute to the virulence of this pathogen. C. albicans<br />

assimilates both fermentative and non-fermentative carbon sources in vivo, often<br />

occupying poor glucose niches in its mammalian host. We reasoned that changes<br />

in carbon source might influence stress adaptation, but stress responses are<br />

generally studied during growth on glucose.<br />

We found that the C. albicans cell wall is extremely dynamic and susceptible to<br />

variations in carbon source. Growth on lactate rather than glucose has a major<br />

impact on cell wall structure and flexibility. Although the relative proportions of<br />

chitin, -glucan and mannan are similar within the wall of lactate-grown cells, they<br />

display increased porosity and decreased cell wall biomass. TEM confirmed major<br />

differences in cell wall architecture, underlying the importance of the carbon source<br />

in the cell biology of this pathogen. This in turn influences the responses of this<br />

pathogen to stresses as these differences were reflected in the cell wall-related<br />

phenotypes. Lactate-grown cells displayed higher resistance to Congo Red and<br />

Calcofluor White and showed significantly increased resistance to various doses<br />

of osmotic stress. This change was independent of Hog1 and Mkc1, but correlated<br />

with large changes in cell volume following exposure to NaCl. Growth on lactate<br />

also increased resistance to anti<strong>fungal</strong>s such as Caspofungin, Tunicamycin and<br />

Amphotericin B, which is likely to have a major impact upon the behavior of C.<br />

albicans during infection.<br />

The impact of carbon source on stress and drug resistance was confirmed in other<br />

pathogenic <strong>Candida</strong> species and for other carbon sources, resulting in variations<br />

in resistance when compared to a glucose-grown control. Clearly carbon source<br />

strongly influences cell wall architecture and the resistance of C. albicans to certain<br />

stresses, which is likely to have a major impact on its in vivo behavior.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial<br />

Training Network (PITN-GA-2008-214004).<br />

122


123


Poster number: 07<br />

<strong>Candida</strong> albicans apoptotic markers during the interaction<br />

with macrophages<br />

A. Gil-Bona 1 , L. Monteoliva 1 , V. Cabezón 2 , M. Ramsdale 2 , C. Gil 1 .<br />

1 2 Facultad de Farmacia. Universidad Complutense de Madrid. School of Biosciences,<br />

University of Exeter, UK<br />

<strong>Candida</strong> albicans is a dimorphic fungus member of the human microbiota that<br />

causes a range of opportunistic infections from superficial to systemic. The last one<br />

remains as a high rate of morbidity-mortality in patients from intensive care units,<br />

surgery or cancer treatment. This is related with the problems of the few available<br />

drugs, as the appearance of anti<strong>fungal</strong>-drug resistance, and with the diagnostic<br />

limitations.<br />

The host-pathogen interaction is being studied using an in vitro interaction model<br />

between <strong>Candida</strong> albicans and murine macrophages to analyze the fungus answer.<br />

Previous proteomics and genomics studies of C. albicans upon interaction with<br />

macrophages allowed us to detect the fungus response to adapt to the new<br />

environment. This response includes the expression of several genes and proteins<br />

related to the regulation of apoptosis (Fernández-Arenas et al., 2007).<br />

In order to verify the apoptotic death of C. albicans cells in the interaction with<br />

macrophages, several apoptotic markers have been analyzed after host-pathogen<br />

interaction; namely, caspase-like enzymatic activity, generation of intracellular<br />

reactive oxygen species (ROS), phosphatidyl-serine externalization and cell viability.<br />

Also, TUNEL (TdT-mediated X-Dutp Nick end labelling) assays and transmission<br />

electron microscopy (TEM) studies have been made.<br />

These analyses have been carried out at different incubation times: 3h, 6h, 8h, 12h,<br />

14h, 16h, 18h, 20h and 24 hours. The results showed that after 6 hours of C.<br />

albicans-macrophage interaction, ROS and active caspase are present in the cell,<br />

positive TUNEL cells were detected after 8 hours of interaction and the viability of<br />

the yeast at these times is not compromised. Furthermore, chromatin condensation<br />

and nuclear fragmentation were observed by TEM after 12 hours of interaction.<br />

Data obtained in this work confirm a percentage up to 30% of apoptotic death of<br />

C. albicans upon interaction with macrophages in ratio 1:1.<br />

124


125


Poster number: 08<br />

Histidine kinase Sln1 interferes with mitochondrial<br />

functions of <strong>Candida</strong> albicans<br />

Rabeay Y.A. Hassan, Anna Buschart, and Ursula Bilitewski*<br />

Helmholtz Centre for <strong>Infection</strong> Research, Biological Systems Analysis, Inhoffenstr. 7, 38124<br />

Braunschweig, Germany. rabeay.hassan@helmholtz-hzi.de<br />

In the pathogenic fungus <strong>Candida</strong> albicans, three histidine kinases (HK) have been<br />

identified: Sln1p, Cos1p (also known as Nik1p) and Chk1p. They are involved in the<br />

adaptation to stress, growth, morphogenesis and sensitivity to several anti<strong>fungal</strong><br />

compounds. Deletion of SLN1 does not affect cell viability in rich media and only<br />

slightly affects the tolerance to osmotic stress.<br />

However, we show for the first time that deletion of the SLN1 perturbs mitochondrial<br />

functions. The oxygen uptake rates of the three HKs deletion mutants, ∆sln1, ∆chk1<br />

and ∆cos1 were measured and a faster rate of oxygen consumption was observed<br />

in sln1 than in the wild-type. This can be explained by the up-regulated expression<br />

of the alternative oxidase AOX2 in sln1, indicating that the alternative respiratory<br />

pathway is activated in addition to the presence of the classical respiratory chain<br />

(CRC). The alternative oxidative pathway is usually activated in response to<br />

inhibitors of the CRC.<br />

Rotenone-treatment of the wild type strain stimulated the production of reactive<br />

oxygen species (ROS) and inhibited the reduction of dichlorophenolindophenol<br />

(DCIP-complex I activity assay). However, rotenone had no or only a small effect<br />

on the ROS production and the inhibition of the DCIP reaction in sln1. These<br />

findings point to a low activity of complex I in sln1. This conclusion was supported<br />

by whole-genome gene expression analysis, which showed a down-regulation of<br />

genes encoding components of the mitochondrial NADH:ubiquinone<br />

oxidoreductase complex (Complex I), namely NAD3, NAD4 and NAD5 in addition<br />

to subunits of cytochrome c oxidase (complex IV) (COX2 and COX5) in sln1.<br />

126


127


Poster number: 09<br />

Phenotypic investigation of virulence factors in <strong>Candida</strong><br />

species isolated from different clinical specimens<br />

Alina Maria Holban 1 , Crina Maria Saviuc 1,5 , Alexandru Mihai Grumezescu 2 ,<br />

Coralia Bleotu 1,4 , Otilia Banu 3 , Dan Mihaiescu 2 , Paul Balaure 2 , Mariana<br />

Carmen Chifiriuc 1 , Veronica Lazar 1<br />

1 University of Bucharest, Faculty of <strong>Biology</strong>, Microbiology University Department; 2 University<br />

Politechnica of Bucharest, Faculty of Applied Chemistry and Materials Science, Organic<br />

Chemistry Department; 3 Institute of Cardiovascular Diseases Prof. C.C. Iliescu, Bucharest;<br />

4 S. Nicolau Institute of Virology, Bucharest; 5 S.C. Biotehnos S.A.<br />

Introduction. The purpose of this study was to investigate the phenotypic expression<br />

of the cell associated and soluble virulence factors in 30 <strong>fungal</strong> strains recently isolated<br />

from different clinical specimens.<br />

Materials and methods. Fungal strains belonging to <strong>Candida</strong> genus were isolated<br />

from different clinical specimens (urinary tract isolates, plague secretions, upper airway<br />

secretions) and identified using Vitek II automatic system. Susceptibility to currently<br />

used anti<strong>fungal</strong>s was assessed by E-test. The phenotypic study of soluble virulence<br />

factors was performed by specific enzymatic assays of haemolysins, lecithinase,<br />

gelatinase, lipase, DNA-se, amylase and iron chelating agents production. Microbial<br />

adherence capacity to the inert (plastic, catheter sections) and cellular substrata was<br />

performed by microtiter method, Brun Buisson method and modified Cravioto method,<br />

using HeLa cells. Fungal dimorphism was studied by filamentation assay. Statistics—<br />

Data were compiled and analyzed using Microsoft Excel and GraphPad In Stat<br />

software, using One-way Analysis of Variance ANOVA.<br />

Results. All tested strains were susceptible to voriconazole, itraconazole, caspofungin<br />

B, fluconazole, flucytosin and amphotericin B. The tested strains exhibited different<br />

abilities to colonize the plastic/catheter sections as well as the cellular substrate, being<br />

divided in non-adherent, moderately and highly adherent. Concerning the filamentation<br />

assay, the strains were classified in the following descendant order: C. albicans, C.<br />

tropicalis, C. glabrata, C. krusei, C. Famata. The profiles of the soluble virulence factors<br />

have been statistically different, related to the species and clinical origin of the tested<br />

strains. The tested strains abundantly secreted haemolysins, DNA-se, amylase and iron<br />

chelating agents.<br />

Conclusion. The phenotypic investigation of virulence factors production in <strong>Candida</strong><br />

strains revealed specific virulence profiles depending on the source of isolation and the<br />

taxonomic affiliation of the tested strains, proving the usefulness of these assays in<br />

predicting the clinical outcome of the <strong>fungal</strong> infections and in the adjustment of the<br />

anti<strong>fungal</strong> treatment.<br />

128


129


Poster number: 10<br />

The Role of Hyphae and Hypha-specific genes in the<br />

pathogenesis of <strong>Candida</strong> dubliniensis and <strong>Candida</strong><br />

albicans<br />

Peter Hyde, Derek Sullivan, Gary Moran, David Coleman.<br />

Oral Biosciences Division, Dublin Dental School and Hospital, Trinity College Dublin, Lincoln<br />

Place, Dublin 2, Ireland<br />

The ability of <strong>Candida</strong> albicans to cause invasive disease has been well<br />

documented. <strong>Candida</strong> dubliniensis, the most closely related phylogenetic neighbour<br />

of C. albicans, causes life-threatening infections at a much lower rate,<br />

predominantly in immunocompromised individuals. Both species can grow as true<br />

hyphae, pseudohyphae and yeast cells. Hyphae are typically associated with host<br />

tissue adhesion and invasion. This study aims to characterise the contribution of<br />

general hyphal morphology, in comparison to the expression of hypha-specific cell<br />

wall proteins, to host tissue adhesion and invasion using molecular methods and<br />

top-down “holistic” approaches. By expressing C. albicans-specific hyphaassociated<br />

cell wall proteins in a C. dubliniensis background under the control of a<br />

tetracycline-inducible promoter, the ability of proteins CaALS3p, CaHWP1p, and<br />

CaHWP1p, to cause tissue adhesion and invasion is being measured, independent<br />

of cell morphology. In addition, by priming the C. dubliniensis mutant strains to<br />

produce hyphae at different rates the combined effect of hypha-associated cell wall<br />

proteins and hyphal morphology is being investigated. The ability of a panel of C.<br />

dubliniensis strains, including systemic isolates, to produce hyphae has also been<br />

examined and has shown high levels of inter-species variation. Interestingly, the<br />

highest rates of true hyphae formation have been associated with strains isolated<br />

from systemic infections, suggesting a significant contribution from the filamentous<br />

morphology in the virulence of C. dubliniensis. This study is exploring the basis<br />

behind the relative lack of pathogenicity of C. dubliniensis and further elaborating<br />

on the role of hyphae and hypha-specific proteins as virulence factors in the<br />

pathogenesis of C. albicans.<br />

130


131


Poster number: 11<br />

Mini-chromosomes in <strong>Candida</strong> glabrata clinical isolates<br />

originate through two different mechanisms<br />

Olena P. Ishchuk 1 , Khadija Mohamed Ahmad 1 , Linda Hellborg 1 , Gloria<br />

Jørgensen 2 , Miha Škvar 1 , Jørgen Stenderup 3 , Dorte Jørck-Ramberg 2 ,<br />

Silvia Polakova 1 and Jure Piškur 1<br />

1 Department of <strong>Biology</strong>, Lund University, Sölvegatan 35, Lund SE-223 62, Sweden,<br />

Olena.Ishchuk@biol.lu.se; 2 BioCentrum-DTU, Technical University of Denmark, DK-2800<br />

Lyngby, Denmark; 3 Department of Clinical Microbiology, Regionshospitalet Herning,<br />

DK-7400 Herning, Denmark<br />

We analyzed around two hundred strains of the pathogenic yeast <strong>Candida</strong> glabrata,<br />

collected from immuno-compromised patients in Danish hospitals during 1985 -<br />

1999. In this report we showed that these strains were closely related but exhibited<br />

large karyotype polymorphism. Nine strains contained mini-chromosomes, which<br />

were smaller than 0.4 Mb. These strains had an independent origin, regarding the<br />

year, patient and hospital, and the analyzed mini-chromosomes were structurally<br />

not related to each other (i.e., they contained different sets of genes). We inferred<br />

two mechanisms involved in their origin: (i) through a segmental duplication which<br />

covered the centromeric region, and (ii) by a translocation event moving a larger<br />

chromosome arm to another chromosome thereby leaving the centromere part with<br />

the shorter arm. The first type of mini-chromosomes carrying duplicated genes<br />

exhibited mitotic instability, while the second type, which contained the<br />

corresponding genes in only one copy in the genome, was mitotically stable.<br />

Apparently, in patients C. glabrata chromosomes are frequently reshuffled resulting<br />

in various genetic configurations, including appearance of mini-chromosomes,<br />

which could increase the fitness in certain “environments”.<br />

132


133


Poster number: 12<br />

Functional analysis of C. albicans infection-associated<br />

genes with unknown function during interaction with host<br />

cells and in complex infection models<br />

Ilse D. Jacobsen 1 , Duncan Wilson 1 , Francois Mayer 1 , Bernhard Hube 1,2<br />

1 Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product<br />

Research and <strong>Infection</strong> <strong>Biology</strong> - Hans-Knöll-Institute, Jena, Germany; 2 Friedrich-Schiller<br />

University, Jena<br />

<strong>Candida</strong> albicans is able to cause a variety of clinical infections. We hypothesized<br />

that C. albicans unknown function genes, which were transcriptionally upregulated<br />

during oral infections, liver invasion or incubation in human blood (“infection<br />

associated genes”) might contribute to the infection process. We selected a subset<br />

of infection-associated genes with unknown function for further analysis.<br />

Twentyfour isogenic deletion mutants were constructed and subsequently analyzed<br />

both in vitro and in infection models of increasing complexity. Sixteen mutants were<br />

impaired in their ability to damage monolayers of human endothelial and/or<br />

epithelial cells. Within this subset, seven mutants additionally displayed decreased<br />

stress resistance in vitro. Only two mutants showed filamentation defects. To<br />

determine whether the deleted genes influenced virulence in more complex<br />

infections models, all 24 mutants were tested in ovo for their ability to kill chicken<br />

embryos infected on the chorio-allantoic membrane. We recently showed that<br />

mortality in this model depends on the <strong>fungal</strong> ability to invade the membrane and<br />

that the pro-inflammatory host response likely contributes to pathogenesis.<br />

Surprisingly, only seven mutants were attenuated in this model, of which five were<br />

also attenuated in damaging epithelial cells in vitro. Three of the mutants, which<br />

were attenuated in ovo, and one mutant, which was attenuated in damaging<br />

endothelial and epithelial cells but fully virulent in chicken embryos, were<br />

subsequently analyzed in a systemic mouse model. Virulence in mice mirrored the<br />

results obtained in ovo.<br />

Our results demonstrate that genes essential for causing full damage of epithelial<br />

or endothelial cells in vitro are not necessarily crucial for full virulence in complex<br />

infection models. We conclude that either such damage potential is not critical for<br />

full virulence in the complex host situation or that the greater complexity of the<br />

environment in animal models stimulates additional <strong>fungal</strong> regulatory networks<br />

leading to compensation of the defect.<br />

134


135


Revisiting the Hog Pathway of C. glabrata<br />

136<br />

Poster number: 13<br />

Zeljkica Jandric, Wolfgang Reiter, Gustav Ammerer, Christoph Schüller<br />

Max F. Perutz Laboratories, Department of Biochemistry University of Vienna, Dr.Bohr-<br />

Gasse 9/5, A-1030 Vienna, Austria<br />

Signal transduction networks mediated by mitogen – activated protein kinases<br />

(MAPK) play essential roles in eukaryotic cells. Yeast cells have developed<br />

osmoadaptation pathways to cope with osmotic changes in the microenvironment.<br />

In Saccharomyces cerevisiae five MAPK pathways are well characterized. One of<br />

these, especially associated with stress response, is the High Osmolarity Glycerol<br />

(HOG) pathway . Stress induced phosphorylation triggers the activation of the key<br />

MAPK, Hog1. After being phosphorylated by the upstream MAPKK Pbs2, Hog1<br />

enters the cell nucleus and modulates the transcription of its target genes.<br />

Orthologues of the all HOG pathway compounds found in Saccharomyces<br />

cerevisiae have been identified in fungi and animals.<br />

The human <strong>fungal</strong> pathogen <strong>Candida</strong> glabrata is closely related to Saccharomyces<br />

cerevisiae. It is a common commensal but in immunocompromised cancer and<br />

transplant patients or elder persons it can turn into an opportunistic pathogen.<br />

Depending on the host environment, <strong>Candida</strong> glabrata has to adapt to<br />

environmental changes during invasion and adaptation steps. We investigated the<br />

role of Hog1 in <strong>Candida</strong> glabrata. Several stress sources were used to identify HOGspecific<br />

phenotypes and to analyze the role of Hog1 in <strong>Candida</strong> glabrata. As<br />

expected, the deletion of Hog1 caused osmostress hypersensitivity. Physiological<br />

stress conditions, such as nutrients starvation, were simulated by infection of<br />

macrophages with <strong>Candida</strong> glabrata and determination of the number of surviving<br />

<strong>Candida</strong> cells. Cghog1∆ cells showed a much higher sensitivity to macrophages<br />

compared to wild type cells. Further investigations focus on stress induced gene<br />

transcription in Cghog1∆ cells and comparison of the transcription pattern to cells<br />

without Hog1 deletion.


137


Poster number: 14<br />

Functional analysis of H-loop residues of Cdr1p, an ABC<br />

transporter of human <strong>fungal</strong> pathogen <strong>Candida</strong> albicans.<br />

Antresh Kumar 1 and Rajendra Prasad 1<br />

1 School of Life Sciences, Jawaharlal Nehru University, New Delhi, INDIA<br />

Nucleotide Binding Domains (NBDs) of multidrug transporter of <strong>Candida</strong> albicans,<br />

Cdr1p possess unique divergent amino acids in their conserved motifs. For<br />

example, NBD1 possesses divergent H-loop (IYQ) while the same motif is<br />

conserved (IHQ) in NBD2.<br />

We report here the contribution of these conserved and divergent H-loop motifs of<br />

Cdr1p in ATP catalysis and drug transport. For this, we mutagenized two residues<br />

of divergent (IYQ) and conserved H-loop region (IHQ) either by replacing them with<br />

alanines or replacing residues with equiposition residues of another H-loop. These<br />

mutations were introduced into GFP-tagged Cdr1p which was stably overexpressed<br />

at the PDR5 locus in a heterologous host S. cerevisiae mutant strain, AD1-8u - which<br />

lacks seven major ABC transporters.<br />

All the Cdr1p mutant variants of H-loop region were properly expressed and<br />

localized to the cell surface similar to wild-type protein. Alanine mutants of H-loop<br />

region, Y361A in NBD1 has no effect on Cdr1p function and showed normal R6G<br />

transport and ATPase activity whereas corresponding mutation (H1059A) in NBD2<br />

abolished R6G transport without any significant loss of ATPase activity. Moreover,<br />

cells expressing mutation like Q362A severely abrogated Cdr1p function while<br />

Q1060A mutation in NBD2 showed no effect on Cdr1p function and measured<br />

normal transport and ATPase activity similar to wild type. Though, Arginine<br />

substitution of glutamine residue present in H-loop region of Cdr1p (Q362R and<br />

Q1060R) displayed deferential effect on Cdr1 transport. These results suggest that<br />

not only the conserved residue like H1059 but divergent Q362 is also crucial for<br />

Cdr1p function.<br />

138


139


Poster number: 15<br />

Target specificity of the Efg1 regulator in <strong>Candida</strong> albicans<br />

Lassak, T., Lagadec, Q., Kurtz, D., Bussmann, M. and Ernst, J. F.<br />

Heinrich-Heine-Universität Düsseldorf, Institut für Mikrobiologie, Molekulare Mykologie<br />

Efg1 is a general transcription factor, which regulates numerous morphogenetic<br />

and metabolic processes in C. albicans. We previously found that overexpression<br />

of EFG1 and activation of Efg1 during hypha formation leads to downregulation of<br />

EFG1 promoter activity and that EFG1 downregulation is necessary to allow the<br />

formation of true hyphae and prevent pseudohyphal growth.<br />

To explore the Efg1 binding specificity its binding to the EFG1 promoter was<br />

analyzed by EMSA and footprint analyses. These experiments revealed binding of<br />

Efg1 to a promoter sequence close to the transcript start site, which contained a<br />

repeated 8-mer motif (referred to as APSES response element, ARE). Surprisingly,<br />

promoter fusions to the RLUC reporter lacking the ARE motif were still able to<br />

downregulate the EFG1 promoter during hypha formation in an Efg1-dependent<br />

manner. Furthermore, extensive deletions of the 10 kb-large 5 untranslated region<br />

of EFG1 still did not prevent regulation suggesting multiple binding sites for Efg1<br />

within the EFG1 promoter. We subsequently analyzed genome-wide occupation of<br />

chromosomal sequences by Efg1 using ChIP chip analysis. These results revealed<br />

the entire 10 kb EFG1 upstream region as a major site of Efg1 occupancy.<br />

Unexpectedly, Efg1 binding to this region was rapidly lost during hypha induction<br />

suggesting a regulatory model, in which Efg1 acts as transcriptional activator<br />

specifically during yeast growth. In further experiments we attempted to reconstitute<br />

the EFG1 autoregulatory circuit in the yeast S. cerevisiae. In this system the<br />

presence of Efg1 was shown to downregulate EFG1 promoter activity, although<br />

not to the same extent as was observed in C. albicans. This finding hints at<br />

co-regulators that together with Efg1 mediate autoregulation in C. albicans.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial Training Network<br />

(PITN-GA-2008-214004).<br />

140


141


Poster number: 16<br />

The contributions of polyubiquitin to stress resistance,<br />

metabolic adaptation and virulence in <strong>Candida</strong> albicans<br />

Michelle D. Leach, Doblin Sandai, David Stead, Evelyn Argo, Donna<br />

MacCallum and Alistair J.P. Brown<br />

Aberdeen Fungal Group, School of Medical Sciences, Aberdeen University, Institute of<br />

Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK<br />

Microorganisms exist in complex and dynamic environments. To survive these<br />

challenges they must continuously monitor environmental change and adapt<br />

appropriately. The major <strong>fungal</strong> pathogen of humans, <strong>Candida</strong> albicans can be<br />

subjected to diverse stresses during commensalism, colonisation of mucosa and<br />

systemic infection. These include chemical and enzymic insults that damage<br />

external and internal structures. Not surprisingly, robust stress adaptation is vital<br />

for the virulence of C. albicans. In eukaryotic cells post-translational modifications<br />

such as ubiquitination are known to play crucial regulatory roles in many cellular<br />

processes including stress adaptation. Indeed, the highly conserved 76 amino acid<br />

protein, ubiquitin, is induced by stresses, mediating protein turnover via the<br />

proteasome in stressed cells. We have combined molecular, cellular and proteomic<br />

approaches to examine the roles of ubiquitination in C. albicans, as little is known<br />

about ubiquitination in this major <strong>fungal</strong> pathogen of humans. Independent null<br />

(ubi4/ubi4) and conditional (MET3p-UBI4/ubi4) mutations were constructed at the<br />

C. albicans polyubiquitin-encoding locus. These mutants displayed morphological<br />

and cell cycle defects, as well as sensitivity to thermal, oxidative and cell wall<br />

stresses. Furthermore ubi4/ubi4 cells rapidly lost viability under starvation<br />

conditions. Consistent with these phenotypes, proteins with roles in stress<br />

responses (Gnd1, Pst2, Ssb1), metabolism (Acs2, Eno1, Fba1, Gpd2, Pdx3, Pgk1,<br />

Tkl1) and ubiquitination (Ubi4, Ubi3, Pre1, Pre3, Rpt5) were amongst the<br />

ubiquitination targets we identified, further indicating that ubiquitination plays key<br />

roles in growth, stress responses and metabolic adaptation in C. albicans. Clearly<br />

ubiquitination plays key roles in the regulation of fundamental cellular processes<br />

that underpin the pathogenicity of this medically important fungus. This was<br />

confirmed by the observation that the virulence of C. albicans ubi4/ubi4 cells is<br />

significantly attenuated.<br />

142


143


Poster number: 17<br />

Dissecting the response of <strong>Candida</strong> albicans to the attack<br />

by neutrophils<br />

Pedro Miramón 1 , Iryna Bohovych 4 , Alistair J. P. Brown 4 , Oliver Kurzai 2,3 ,<br />

Bernhard Hube 1,3<br />

1 Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product<br />

Research and <strong>Infection</strong> <strong>Biology</strong> - Hans Knoell Institute (HKI), Jena, Germany; 2 Department of<br />

Fungal Septomics, Leibniz Institute for Natural Product Research and <strong>Infection</strong> <strong>Biology</strong> - Hans<br />

Knoell Institute (HKI), Jena, Germany; 3 Friedrich-Schiller Universität, Jena, Germany; 4 Aberdeen<br />

Fungal Group, Institute of Medical Sciences, University of Aberdeen, UK<br />

<strong>Candida</strong> albicans successfully colonises diverse niches in the human host without causing any<br />

harm thanks to a balance with the microflora and possibly due to a constant surveillance of<br />

patrolling immune cells. However, under certain conditions, this fungus is able to cause lifethreatening<br />

deep-seated infections. Although it is know that neutrophils are key players in<br />

controlling and eliminating C. albicans, it is unclear how C. albicans is killed by neutrophils and<br />

how the fungus counteracts these phagocytes.<br />

By means of single-cell expression profiling using GFP reporter strains, we characterised<br />

different responses of C. albicans to three main neutrophil-imposed stresses: nutrient starvation,<br />

oxidative stress and nitrosative stress.<br />

We observed that phagocytosed <strong>fungal</strong> cells responded to carbohydrate starvation by<br />

upregulating the glyoxylate cycle, but not the glycolytic pathway. Moreover, a mutant lacking a<br />

key gene of the glyoxylate cycle (icl1 ) exhibited decreased resistance in the presence of<br />

neutrophils.<br />

Next, we investigated the contribution of oxidative stress to killing by neutrophil. The catalase<br />

gene CTA1 was induced upon phagocytosis. However, the surface-associated superoxide<br />

dismutase gene SOD5 was induced in yeast cells even before phagocytosis took place,<br />

suggesting that C. albicans can sense and encounter an extracellular oxidative attack. In a<br />

similar fashion, the thioredoxin gene TRX1 was also upregulated upon extracellular contact with<br />

neutrophils. Strikingly, neither cta1 nor trx1 mutants exhibited decreased resistance to<br />

neutrophils, suggesting that they are not essential for normal resistance of C. albicans. However,<br />

sod5 was more sensitive to neutrophils, indicating that Sod5 is an important detoxifying enzyme<br />

to cope with neutrophil-generated oxidative stress. C. albicans also responded to nitrosative<br />

stress generated in the intracellular milieu and this response was essential for full resistance as<br />

a yhb1 mutant, defective in nitric oxide dioxygenase, exhibited decreased survival.<br />

In summary, our study provides evidence that C. albicans displays specific responses to<br />

overcome either extracellular or intracellular attack by neutrophils.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial Training Network (PITN-GA-<br />

2008-214004).<br />

144


145


Poster number: 18<br />

A C-terminal hyphal activation motif of the transcription<br />

factor Tec1p is essential for biofilm formation and<br />

nematode infection in <strong>Candida</strong> albicans<br />

Schröppel K., Singh A., Dietsche T., Eickhoff H., Wiesmüller K.-H., Fischer<br />

K., and Abu Rayyan W.<br />

Institute of Medical Microbiology and Hygiene, University Hospital and Clinics, University of<br />

Tübingen, Tübingen, Germany<br />

Hyphal formation of <strong>Candida</strong> albicans is a prerequisite for penetration of host<br />

tissues. The formation of biofilms allows this pathogen to persist and to increase<br />

resistance against anti-<strong>fungal</strong> compounds. In order to define a structure function<br />

relationship for the important transcription factor Tec1p, we integrated modified<br />

open reading frames (ORFs) of the wild type TEC1 under the control of the C.<br />

albicans adapted reverse Tet-dependent transactivator (rtTA) into the tec1/tec1<br />

mutant. We screened a library of such C. albicans mutants for their ability of in vitro<br />

biofilm formation and morphogenetic development, and examined their virulence<br />

in a nematode host using C. elegans as an infection model. Additionally, we<br />

characterized the anti<strong>fungal</strong> activity of EMC120B12, a novel (S)-2-aminoalkyl<br />

benzimidazole derivative, against the library of C. albicans TEC1 mutants in vitro<br />

and in vivo. In comparison to a full length TEC1 ORF, the ORF constructs either with<br />

a deletion of the C-terminus from position 637 aa to the stop codon at 744 aa or a<br />

deletion of the TEA domain from 216 to 244 aa showed abrogation in restoring<br />

hyphal growth and biofilm formation in the tec1/tec1 mutant. Interestingly, the<br />

tec1/tec1 phenotype was rescued in a mutant containing an ORF with an additional<br />

72 aa that spanned the region from 1 to 709 aa. The deletion of the proposed Cterminal<br />

hyphal activation motif CHAM of Tec1p did not affect DNA binding to a<br />

TEA consensus sequence TCS probe.<br />

In the C. elegans infection model we found that the C. albicans mutant lacking<br />

CHAM showed less virulence in terms of hyphal invasion and killing of the nematode<br />

host. During in vivo infection studies, we could demonstrate that EMC120B12 can<br />

inhibit filamentation of C. albicans and prolong survival of the C. albicans infected<br />

nematodes.<br />

146


147


Poster number: 19<br />

Resistance of <strong>Candida</strong> albicans biofilms formed on<br />

catheters and probes and the discovery of a<br />

chlamydospore in the biofilm<br />

S.M.L. Seddiki a, , Z. Boucherit-Atmani a , K. Boucherit a ,<br />

L. Sari-Belkharoubi a , D. Kunkel b<br />

a Laboratoire antibiotique antifongique, physico- chimique synthèse et activité biologique.<br />

Département de biologie, Faculté des sciences de la nature et de la vie et des sciences de<br />

la terre et de l’univers, Université Abou bekr belkaid. BP 119, Imama 13000 Tlemcen.<br />

ALGERIE; b Dennis Kunkel Microscopy, Inc. P.O. Box 2008 Kailua, HI 96734, USA<br />

In Algeria, many studies on bacterial biofilms were clarified but those of <strong>fungal</strong><br />

origin, particularly caused by the yeast <strong>Candida</strong> albicans remained unidentified.<br />

This work was performed at the hospital Chabane Hamdoune of Maghnia (Algeria),<br />

where 51 strains of C. albicans representing 16.94% of all samples taken were<br />

isolated. They were collected from catheters and probes from different hospital<br />

services with variable rates, and the most concerned service was the ICU (40.74%)<br />

followed by the gynecology department (17.39 %), while that of general surgery<br />

came in third (15.79 %). Testing the anti<strong>fungal</strong> amphotericin B (AmB) showed clearly<br />

that the sessile cells of C. albicans were much more resistant than their planktonic<br />

counterparts (suspended cells), especially at the resistance increases during the<br />

different phases of biofilm formation until it reaches its threshold at the ripening<br />

stage (48 hrs). Furthermore, scanning electron microscopy of the isolated strains<br />

in the laboratory proved the formation of biofilms on catheters by <strong>Candida</strong> albicans.<br />

Surprisingly, observations revealed a new structure in these biofilms: The<br />

chlamydospore.<br />

keywords: <strong>Candida</strong> albicans; nosocomials infections; catheters; biofilms;<br />

chlamydospore.<br />

148


149


Poster number: 20<br />

Investigation of combinatorial stress responses in C.<br />

albicans<br />

Anna Theresa Tillmann 1 , Elahe Radmaneshfar 2 , Tao You 2 , Despoina<br />

Kaloriti 1 , Mette Jacobsen 1 , Zhikang Yin 1 , Marco Thiel 2 , Celso Grebogi 2 ,<br />

Neil A.R. Gow 1 , Mike Gustin 3 , Alistair J.P.Brown 1<br />

1 School of Medical Sciences, University of Aberdeen, UK; 2 Institute of Complex Systems<br />

and Mathematical <strong>Biology</strong>, University of Aberdeen, UK; 3 Department of Cell Biochemistry<br />

and Cell <strong>Biology</strong>, Rice University, USA.<br />

<strong>Candida</strong> albicans is a major human <strong>fungal</strong> pathogen that causes mucosal and<br />

systemic infections. Immune cells combat <strong>Candida</strong> infections with reactive nitrogen<br />

and reactive oxygen species. Meanwhile <strong>Candida</strong> has evolved strategies to<br />

minimise the damage caused by these environmental stresses, and the success of<br />

this pathogen depends partly on its robust adaptation to these stresses. We<br />

examined three infection-associated stresses: nitrosative, oxidative and osmotic<br />

stress. First we examined responses to each individual stress. Then we analysed<br />

responses to stress combinations because C. albicans cells are exposed to such<br />

combinatorial stresses in vivo. The Hog1 SAPK, the cell wall integrity and the Cta4-<br />

Yhb1 pathways contribute to these combinatorial stress responses. The<br />

involvement of these pathways was confirmed by sensitivity testing of null mutants<br />

to NO, H 2 O 2 and NaCl, and by testing the activation of the corresponding MAP<br />

kinases by Western blotting. Furthermore transcription factor localisation and the<br />

expression of downstream target genes provided further evidence for this activation.<br />

The relative contributions of these nitrosative, oxidative and/or osmotic stress<br />

resistance pathways during C. albicans infections are discussed.<br />

150


151


Poster number: 21<br />

Agent based modelling and host pathogen interactions<br />

Katarzyna M. Tyc 1 , Clemens Kühn 1 , Edda Klipp 1<br />

1 Theoretische Biophysik, Humboldt-Universität zu Berlin, Berlin, Germany<br />

Mathematical models of pathogen and host interactions provide a tool for medical<br />

applications and potential for pharmaceutical companies for testing novel drug<br />

treatments and consequences of their application. Host pathogen interactions are<br />

highly complex and experimental techniques even when combined with different<br />

computational methods allow only for partial investigation of their mutual<br />

dependences.<br />

Based on the available literature we reconstruct a computational model of a human<br />

oral epithelium invasion by the pathogenic fungus <strong>Candida</strong> albicans. We investigate<br />

the system’s dynamics using the agent based modelling approach (ABM) where<br />

cells (<strong>Candida</strong> cells, neutrophils (PMNs)) are represented as individual agents. These<br />

agents interact in different ways: <strong>fungal</strong> cells form hyphal colonies on the epithelium<br />

and then epithelial cells secrete cytokines upon contact with the hyphae form of<br />

fungi, indicating inflammation. Cytokine secretion is one of the signals stimulating<br />

PMNs recruitment to the site of infection. Mix of different gradients such as lactate<br />

dehydrogenase (LDH) release from damaged epithelium, cytokines, and secreted<br />

hyphal particles influences PMN activity. In our model, we investigate the essential<br />

parameters that establish the system’s dynamics. We also look at the<br />

consequences of an anti<strong>fungal</strong> drug treatment on the <strong>Candida</strong> albicans survival rate<br />

and its dose dependence. We test the optimal intervals and doses for new<br />

application.<br />

With agent based models, we can investigate the effect of individual dynamics on<br />

global patterns, enabling an intuitive view on host-pathogen interactions that can<br />

provide relevant predictions for further experiments.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial Training Network<br />

(PITN-GA-2008-214004).<br />

152


153


Poster number: 22<br />

<strong>Candida</strong> albicans unique genes: specific roles in host cell<br />

damage<br />

Duncan Wilson 1 , Francois Mayer 1 , Ilse Jacobsen 1 , Bernhard Hube 1,2<br />

1 Hans Knoell Institute, Jena, Germany; 2 Friedrich Schiller University, Jena, Germany<br />

<strong>Candida</strong> albicans is amongst the most pathogenic species of fungi and causes both<br />

superficial (high prevalence) and systemic (high mortality) infections. Because the<br />

major human pathogenic <strong>fungal</strong> species are distantly related, we hypothesized that<br />

they may possess unique genes which account for their high pathogenic potential.<br />

Using C. albicans as a model pathogen, we performed in silico genome subtraction<br />

and transcriptional analysis to identify [C. albicans] species-unique genes<br />

expressed during infection. Gene deletion combined with a series of infection<br />

models (including biochemical analysis of cellular damage and fluorescence<br />

microscopy-based morphological, adhesion and invasion assays for in vitro<br />

epithelial, endothelial and macrophage infection models, as well as an in vivo mouse<br />

virulence model) were used to determine the role of C. albicans unique genes during<br />

infection.<br />

A total of 65 C. albicans unique genes, expressed during infection, were identified<br />

and six were deleted. Five of the resultant homozygous mutants exhibited<br />

significantly altered phenotypes in at least one infection model. One gene, PGA16,<br />

was found to be required both for epithelial destruction and virulence in a mouse<br />

model of hematogenously disseminated candidiasis. Furthermore, this gene was<br />

required for hyphal ramification and optimal dissemination within epithelia.<br />

This study therefore provides molecular evidence that filament branching of a <strong>fungal</strong><br />

pathogen contributes to virulence.<br />

154


155


Poster number: 23<br />

Fitness Profiling in <strong>Candida</strong> albicans by Systematic Gene<br />

Overexpression<br />

Sadri Znaidi 1,2 , Vitor Cabral 1,2 , Melanie Legrand 1,2 , Arnaud Firon 1 , Murielle<br />

Chauvel 1 , Carol. A. Munro 3 and Christophe d’Enfert 1,2 .<br />

1 Institut Pasteur, Unité Biologie et Pathogénicité Fongiques, Département Génomes et<br />

Génétique, F-75015 Paris, France; 2 INRA, USC2019, F-75015 Paris, France; 3 School of<br />

Medical Sciences, University of Aberdeen, Aberdeen, AB25 2ZD, UK<br />

Background: Gene overexpression is a powerful strategy for mapping pathways<br />

and phenotypes. It mimics gain-of-function mutations, complements loss-offunction<br />

phenotypes and allows to study the function of both essential and non<br />

essential genes. Because lack of a complete sexual cycle and aneuploidy hamper<br />

the use of classical genetics in C. albicans, we hypothesized that combining gene<br />

overexpression and strain barcoding would help in understanding gene function in<br />

C. albicans.<br />

Methods: Pools of 531 barcoded C. albicans strains, each overexpressing a unique<br />

ORF under the control of a doxycycline (dox)-inducible promoter, were grown i) for<br />

18 generations in rich medium in the absence or presence of dox or ii) in doxsupplemented<br />

medium in the presence or absence of caspofungin (~IC 90% ). The<br />

relative abundances of the strains were determined using barcode arrays. To<br />

validate the microarray data, growth rates of the strains were measured individually,<br />

using spot or liquid growth assays.<br />

Results: We found 8 genes whose overexpression decreased C. albicans fitness in<br />

rich medium by at least 1.5-fold (p


157


158


The Key Battlefields<br />

159


Poster number: 24<br />

Characterization of the <strong>Candida</strong> albicans PKH1 and YPK1<br />

genes for their roles in biofilm formation and cell wall<br />

integrity<br />

Vitor Cabral 1,2 , Arnaud Firon 1,2 , Tobias Schwarzmüller 3 , Karl Kuchler 3 ,<br />

Christophe d’Enfert 1,2 and Sophie Bachellier-Bassi 1,2<br />

1 Institut Pasteur, Unité Biologie et Pathogénicité Fongiques, Département Génomes et<br />

Génétique, F-75015 Paris, France; 2 INRA, USC2019, F-75015 Paris, France; 3 Medical<br />

University Vienna, Max F. Perutz Laboratories, Christian Doppler Laboratory for <strong>Infection</strong><br />

<strong>Biology</strong>, Department of Medical Biochemistry, A-1030 Vienna, Austria<br />

In Saccharomyces cerevisiae, the Pkh1/Pkh2 and Ypk1/Ypk2 protein kinases are<br />

part of a signaling cascade important for maintenance of cell wall integrity. Our<br />

analysis of a large collection of <strong>Candida</strong> glabrata knock-out mutants for their ability<br />

to form biofilms has revealed that the C. glabrata Pkh2 protein kinase<br />

(CAGL0I07513g) is necessary for growth on solid surfaces and efficient biofilm<br />

formation while Pkh1/CAGL0G04609g is not (Schwarzmüller et al., in preparation).<br />

<strong>Candida</strong> albicans has only one gene for the Pkh protein kinase and one gene for<br />

the Ypk protein kinase that we have named PKH1 and YPK1, respectively. In order<br />

to test whether the Pkh1 and Ypk1 protein kinases also play a role in biofilm<br />

formation in C. albicans, heterozygous and homozygous knockout mutants for the<br />

YPK1 and PKH1 genes have been constructed. Preliminary data show that the<br />

ypk1∆/ypk1∆ knock-out mutant has altered sensitivity to the cell wall perturbing<br />

agents Congo Red and Calcofluor White suggesting that Ypk1 might perform a<br />

function similar to that of Ypk1 and Ypk2 in S. cerevisiae. Additional studies of these<br />

mutants and complemented strains are ongoing and will be presented.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial Training Network<br />

(PITN-GA-2008-214004).<br />

160


161


Poster number: 25<br />

Insights into <strong>Candida</strong> tropicalis virulence factors<br />

Melyssa Negri 1 , Sónia Silva 1 , Mariana Henriques 1 , Terezinha Svidzinski 2 ,<br />

Joana Azeredo 1 , Rosário Oliveira 1<br />

1 Institute for Biotechnology and Bioengineering, Centre of Biological Engineering,<br />

Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal; 2 Teaching and<br />

Research in Clinical Analysis Laboratory, Division of Medical Mycology, Universidade<br />

Estadual de Maringá, Av. Colombo, 5790, postal code 87020-900, Maringá, Paraná, Brazil.<br />

<strong>Candida</strong> tropicalis is a common nosocomial species related to candidemia and<br />

candiduria. Several virulence factors seem to be responsible for C. tropicalis<br />

infections, which lead to high mortality. Adhesion to surfaces (medical devices and<br />

host cells) and biofilm formation are considered important factors that contribute<br />

to the development of candidosis. Therefore, adhesion to urinary catheters and<br />

biofilm formation were assessed in an optimized in vitro flow model, using silicone<br />

and latex urinary catheters and artificial urine (AU). Moreover, biofilm matrices were<br />

also evaluated in terms of proteins and carbohydrates. Regarding adhesion to biotic<br />

surfaces, the interaction of C. tropicalis with host cells was determined using three<br />

different human epithelial cell lines: TCC-SUP (urinary bladder); HeLa (cervical<br />

carcinoma) and Caco-2 (colorectal adenocarcinoma). Specifically, the degree of<br />

human cells damage and activity reduction induced by C. tropicalis adhesion and<br />

the role of <strong>Candida</strong> tropicalis aspartyl proteinases (SAPT) genes expression were<br />

assessed. Additionally, the influence of C. tropicalis biofilm cells with different ages<br />

(24 - 120 h) on TCC-SUP cells integrity was also studied. Another important<br />

<strong>Candida</strong> factor is its resistance to anti<strong>fungal</strong> agents, which was also assessed and<br />

related with the expression of enzymes and hyphae formation.<br />

In summary, C. tropicalis strains were able to form biofilms in AU, in static or<br />

dynamic mode, although, with differences among strains. It is important to<br />

emphasize that human cells response to C. tropicalis adhesion, as well as SAPs<br />

production, is strain and cell line dependent. Additionally, it should be highlighted<br />

that C. tropicalis cells detached from biofilms are able to colonize human cells and<br />

cause injury and reduction of metabolic activity. In addition SAPT3 was highly<br />

expressed compared to other SAPT genes. Therefore, it should be pointed out that<br />

C. tropicalis presented a set of different virulence factors that might be responsible<br />

for its high degree of infection.<br />

162


163


Poster number: 26<br />

Iff2 is a cell surface protein involved in adhesion<br />

Iaroslava Kos 1 , Grégory Da Costa 1 , Céline Monniot 1 , Anita Boisramé 1<br />

Marie-Noëlle Bellon Fontaine 2 , Murielle Chauvel 3 , Christophe d’Enfert 3 and<br />

Mathias L. Richard 1<br />

1 Team “Virulence and Fungal <strong>Infection</strong>” Institut Micalis, UMR1319 INRA AgroParisTech,<br />

78850 Thiverval-Grignon, FRANCE. 2 Team “Bioadhésion et Hygiène des Matériaux” Institut<br />

Micalis, UMR1319 INRA AgroParisTech, 91744 Massy, FRANCE. 3 Team “Fungal <strong>Biology</strong> and<br />

Pathogenicity”, Pasteur Institute, 75015 Paris, FRANCE.<br />

In <strong>Candida</strong> albicans, the cell wall and especially cell wall proteins are known to play<br />

a key role in the relationship between the <strong>fungal</strong> cell, the host and most indwelling<br />

device, contributing to adhesion, immune response modulation and deep seated<br />

infections. A specific class of cell wall proteins has been particularly under the<br />

scope in the past decade: the glycosylphosphatidylinositol anchored proteins<br />

(GpiPs). The largest family of GpiPs is a family of 12 proteins sharing a high similarity<br />

with Hyr1. One of its members, Iff2 has been the subject of a large scale study in<br />

our laboratory in order to decipher his putative function in C. albicans. The cell<br />

localization of Iff2 was assessed using a V5-tagged protein and clearly showed that<br />

Iff2 is a cell surface protein tethered to the cell wall proteins through alkali-Pir-like<br />

links as well as di-sulfide bonds. We also demonstrated that Iff2 protrude from the<br />

cell wall glucan network at the interface with the cellular environment. At the gene<br />

level, we showed that IFF2 is a gene with an expression level stronger than most of<br />

the member of Hyr/Iff family in laboratory conditions and that IFF2 is induced during<br />

the stationary phase in the same media. An overexpressing strain with IFF2 under<br />

the control of TEF1p was hyper adherent to different plastic but not to silicon,<br />

resulting in the formation of stronger biofilm at the early stage of biofilm formation<br />

compared to the reference strain. The future work is aimed to understand which<br />

part of the protein is important for adhesion and if it is specific to Iff2 functional<br />

domain. Additional experiments are also planned to monitor the consequences of<br />

this overexpression in in vitro and in vivo models.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial<br />

Training Network (PITN-GA-2008-214004).<br />

164


165


Poster number: 27<br />

C. glabrata cell wall attached and intracellular proteinases<br />

Pirjo Pärnänen 1,2 , Pirjo Nikula-Ijäs 1<br />

1 Department of Biosciences, Division of Biochemistry, University of Helsinki, Finland;<br />

2 Institute of Dentistry, University of Helsinki, Finland<br />

Background: C. glabrata is the most common non-albicans <strong>Candida</strong> species<br />

causing oral mucosal infections and the third most common microbe that causes<br />

invasive mortal hospital infections. C. glabrata has an an innate immunity to the<br />

most common anti<strong>fungal</strong> group azoles. Proteinases cause tissue protein<br />

degradation and are involved in <strong>Candida</strong> virulence and invasion. Compared to the<br />

most studied C. albicans, C. glabrata proteinases are mostly unfound or<br />

uncharacterized. Against this background we studied clinical C. glabrata species<br />

to locate and characterize these proteinases.<br />

Materials and methods: Eight C. glabrata clinical strains, both mucosal and blood<br />

isolates from hospitalized patients,were included. Gelatin zymography was used<br />

for preliminary scanning of potential active proteinases and their inhibition by<br />

proteinase inhibitors. The most gelatinolytic blood isolate (T-1639) was used for<br />

further studies. Cell wall bound proteinases were enzymatically released and yeast<br />

cells were sonicated to isolate plasma membrane and intracellular fractions in a<br />

native state. Further activity testing was performed by fluorometric detection of<br />

degraded synthetic L-Arg-AMC proteinase substrate.<br />

Results: All the strains showed variable gelatinolytic activity by zymography. The<br />

strain T-1639 showed enzymatic activity of approximate 25 kDa size released from<br />

the 10x concentrated cell wall fraction, and more pronounced intracellular activity<br />

of approximately 75 kDa size, which were both verified with fluorometry. The cell<br />

wall bound proteinase was inhibited by a serine proteinase inhibitor (PMSF),and<br />

the intracellular proteinase by a metalloproteinase inhibitor (EDTA).<br />

Conclusions: The found proteinases are potentially new to our knowledge and<br />

merrit further studies. The found proteinases belong to two different proteinase<br />

classes and most probably have distinct roles in protein degradation and<br />

pathogenic properties of C. glabrata.<br />

166


167


Poster number: 28<br />

Dolichol-dependent glycosylation; a prerequiste of<br />

<strong>Candida</strong> albicans morphogenesis<br />

Mateusz Juchimiuk 1 , Jacek Orłowski 1 , Joachim Ernst 2 Katarzyna<br />

Gawarecka 1 , Ewa Kula Świeżewska 1 and Grażyna Palamarczyk 1<br />

1 Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawi skiego 5a,<br />

02 106 Warsaw, Poland; 2 Department of <strong>Biology</strong>, Molecular Mycology, Heinrich-Heine-<br />

Universitaet, Duesseldorf, Germany<br />

An interesting feature of C. albicans biology is its ability to grow as unicellular<br />

budding yeast, pseudohyphae, true hyphae and chlamydospore. The yeast to<br />

hyphae transition is considered as a prerequisite of C. albicans virulence. On the<br />

other hand, recent studies on the role of mannoproteins in C. albicans virulence<br />

have demonstrated the importance of protein glycosylation for the <strong>fungal</strong><br />

pathogenesis. In the present work we characterized <strong>Candida</strong> albicans RER2 gene,<br />

[cis-prenyltransferase (Rer2p) encoding]. Rer2p is the first enzyme of the<br />

mevalonate pathway committed to the biosynthesis of dolichol, the isoprenoid lipid,<br />

involved in protein glycosylation. A conditional mutant, expressing the C. albicans<br />

ortholog of RER2 from the regulatable MET3 promoter, contained only 4 % of cisprenyltransferase<br />

activity and markedly diminished amount of dolichols, when<br />

cultivated in the repressive condition. Furthermore, at 28 o C the growth of the strain<br />

was retarded and the strain was unable to grow at the restrictive temperature of 37<br />

°C. Particularly, decreased dolichol level prevented morphological differentiation of<br />

the strain leading to hyphae formation. Biochemical analysis confirmed<br />

glycosylation disorder and compromised cell wall integrity. Our results suggest that<br />

biosynthesis and level of dolichol and its ability to enter glycosylation pathway upon<br />

phosphorylation, are essential not only for protein glycosylation and the cell wall<br />

integrity but also for the growth and morphological differentiation of C. albicans<br />

leading to virulence.<br />

168


169


Poster number: 29<br />

Localization of the Phr1p, a glucan remodeling enzyme<br />

crucial for morphogenesis and virulence in <strong>Candida</strong><br />

albicans<br />

G. Degani and L.Popolo<br />

Università degli Studi di Milano, Dipartimento di Scienze Biomolecolari e Biotecnologie, Via<br />

Celoria 26, Milano, Italy<br />

PHR1 is a pH-regulated gene belonging to a family of five genes of C. albicans.<br />

PHR1 expression is triggered at pH values of the growth medium > 5.5. Loss of<br />

PHR1 induces morphological defects at neutral-alkaline ambient pH, inability to<br />

support hyphal growth, to adhere and invade human epithelia. PHR1 mutants are<br />

avirulent in mouse models of systemic infection. At the restrictive pH, cells lacking<br />

Phr1p have an irregular surface and higher and delocalized content of chitin in the<br />

abnormal germ tubes which the mutant produces. Phr1p is a glycoprotein endowed<br />

with beta(1,3)-glucan elongase activity (GH72) similarly to Gas1p of S. cerevisiae<br />

and Gel proteins of A. fumigatus. We obtained a GFP fusion to determine its cellular<br />

localization. During growth in yeast form Phr1p-GFP was predominantly detected<br />

at the bud periphery, bud-neck and septum. During hyphal growth Phr1p-GFP<br />

concentrated at the tip of the germ tubes and gradually distributed along the lateral<br />

side of the hyphae. Phr1p also localized to the septa of the hyphae. By exploiting<br />

the conditional nature of PHR1 and the intrinsic instability of the fluorescent hybrid,<br />

we performed pH shift-down experiments. Differently from the cell wall-bound form<br />

of ScGas1p which is stabilized by cross-links to the chitin ring and remains in the<br />

bud scars after generations, Phr1p-GFP did not persist. Further studies were<br />

performed using mutants lacking CHS3 or members of the CRH family whose<br />

homologues are known to be required for Gas1p immobilization to the bud scar.<br />

Our results suggest that the activity of Phr1p is required at the sites of wall formation<br />

where the incorporation of newly synthesized beta(1,3)-glucan is strictly coupled<br />

to morphogenic events. Moreover, our studies revealed that the mechanisms active<br />

in tethering Gas1p to the bud scars in S. cerevisiae are not operative in C. albicans.<br />

170


171


Poster number: 30<br />

A Novel Flow Cytometric Protocol for Assessment of Yeast<br />

Cell Adhesion<br />

A. Silva-Dias 1,2 , I.M. Miranda 1,2 , R. Rocha 1 , M. Monteiro-Soares 3 ,<br />

A. G. Rodrigues 1,2,4 , C. Pina-Vaz 1,2,5<br />

1 2 Department of Microbiology, Faculty of Medicine, University of Porto, Cardiovascular<br />

Research & Development Unit, Faculty of Medicine, University of Porto; 3CINTESIS, Department of Biostatistics and Medical Informatics, Faculty of Medicine, University of Porto,<br />

4 5<br />

Burn Unit and Department of Plastic and Reconstructive Surgery, Hospital S. João,<br />

Department of Microbiology, Hospital S. João, Porto, Portugal.<br />

The capacity of microorganisms to adhere to each other, to biomaterials, to biotic<br />

substrates and tissues is an important trait at environmental, industrial or medical<br />

level. Pathogenic yeasts belonging to genus <strong>Candida</strong> are frequently involved in<br />

systemic infections, mainly due to its ability to adhere and colonize tissues and<br />

medical indwelling devices. In order to quantify this attribute, several methods have<br />

been used, with advantages and drawbacks. Some of the major limitations found<br />

within these methodologies concern the incubation time, the small number of cells<br />

analyzed and the operator`s subjectivity. In order to overcome these aspects we<br />

have developed a quantitative method to measure yeast cells` adhesion through<br />

flow cytometry. The adhesion assay is based upon a simple principle: yeast cells<br />

became fluorescent when attached to highly green fluorescent microspheres.<br />

Therefore by flow cytometry a quantitative distinction between non-adherent yeast<br />

cells (non fluorescent) and adherent cells (fluorescent) is achieved. By measuring<br />

the intensity of fluorescence emitted by each cell it is possible to evaluate the<br />

number of beads that are attached to each cell. Additionally, fluorescent beads<br />

could be coated with different substrates to which yeast adhesion ability can be<br />

tested.<br />

A suspension of yeast cells (1x10 6 cell.ml -1 ) is mixed with green fluorescent<br />

polystyrene microspheres, uncoated or coated with host proteins, and incubated<br />

for 30 minutes. Samples (50.000 cells) are analyzed in the FL3 fluorescence channel<br />

(FACSCalibur BD Biosciences). Within two hours an adhesion profile is obtained<br />

based on three parameters: percentage of adherent cells, mean of fluorescence<br />

intensity (MFI) and cells-microsphere population`s distribution pattern. Comparing<br />

to the classical assays, flow cytometry protocol represents a useful tool to quantify<br />

yeast adhesion to different substrata in a large scale, providing manifold data in a<br />

speedy and informative manner.<br />

A Silva-Dias is supported by a FCT (Fundação Ciência e Tecnologia) doctoral grant<br />

SFRH/BD/44896/2008; I Miranda is supported by Ciência 2008 (FCT) and European Social Fund.<br />

172


173


Poster number: 31<br />

Cell wall stress elicits a conserved response in <strong>Candida</strong><br />

albicans<br />

Alice G. Sorgo, Clemens J. Heilmann, Sepehr Mohammady, Henk L.<br />

Dekker, Stanley Brul, Chris de Koster, Leo J. de Koning and Frans M. Klis<br />

Swammerdam Institute for Life Sciences, Universiteit van Amsterdam, Science Park 904,<br />

1098 XH Amsterdam, The Netherlands<br />

Both cell wall proteins and secreted proteins of C. albicans are critical for fitness,<br />

virulence and adaptation to environmental challenges. We have recently shown that<br />

fluconazole, an anti<strong>fungal</strong> drug that affects ergosterol synthesis and increases<br />

membrane fluidity, also causes cell wall stress as evidenced by decreased<br />

resistance to wall-perturbing compounds and higher chitin levels in the wall (1). This<br />

is accompanied by substantial changes in the wall proteome and secretome. We<br />

found that the abundance of proteins mainly involved in wall biosynthesis and<br />

integrity increased (Crh11, Pga4, Pir1, Phr1, Phr2, Sap9). However, the abundance<br />

of hypha-associated proteins (Als3, Hwp1, Plb5) decreased in agreement with<br />

reduced hyphal growth. Finally, fluconazole induced cell clustering. As membrane<br />

fluidity also tends to increase at higher temperatures, we wanted to investigate<br />

whether the response to thermal stress is similar to that of fluconazole and could<br />

also result in cell wall stress. When C. albicans was cultured at 42°C, which resulted<br />

in substantial growth inhibition, the cells showed indeed decreased resistance to<br />

wall-perturbing agents, and chitin levels in the wall were elevated, both suggesting<br />

cell wall stress. Likewise, the wall proteome response was similar to that of<br />

fluconazole-treated cells, including increased incorporation levels of proteins<br />

important for wall maintenance and integrity. Additionally, cell separation was<br />

impaired, correlating with a severe reduction of the chitinases Cht2 and Cht3 and<br />

the endoglucanase Eng1 in the medium. In summary, our data indicate that both<br />

thermal stress and fluconazole-induced stress induce membrane stress, which<br />

leads to cell wall stress and a conserved wall stress response.<br />

References<br />

(1) The effects of fluconazole on the secretome, the wall proteome and wall integrity of the clinical fungus<br />

<strong>Candida</strong> albicans. Sorgo AG, Heilmann CJ, Dekker HL, Bekker M, Brul S, de Koster CG, de Koning LJ,<br />

Klis FM. Eukaryot Cell. 2011. doi:10.1128/EC.05011-11.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial Training Network<br />

(PITN-GA-2008-214004).<br />

174


175


176


The Defensive Shields<br />

177


Poster number: 32<br />

Human cells response to Non- <strong>Candida</strong> albicans <strong>Candida</strong><br />

infection<br />

Cláudia Botelho 1 , Melyssa Negri 1 , Catarina Seabra 1 , Mariana Henriques 1 ,<br />

Joana Azeredo 1 , Rosário Oliveira 1<br />

1 Institute for Biotechnology and Bioengineering, Centre of Biological Engineering,<br />

Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal<br />

This study evaluated the influence of adhered <strong>Candida</strong> spp (C.tropicalis ATCC 750,<br />

C. parapsilosis ATCC 2201, C. glabrata ATCC 2001 and C. albicans SC5413) on the<br />

biological behavior of human cells (HeLa and TCC-SUP). A yeast suspension was<br />

added to a confluent layer of human cells and after 2 and 24h of incubation yeast<br />

cells were detached for CFU counts. All <strong>Candida</strong> spp assayed adhered to epithelial<br />

cells, although C. glabrata was the species adhering in higher extent to both<br />

epithelial cell lines. From 2 to 24h there is a significant increase in the number of C.<br />

glabrata adhered to TCC-SUP cells, while no increase in the number of C. glabrata<br />

colonizing HeLa cells was observed. An opposite result was observed for C.<br />

tropicalis; there was no increase in the number of adherent cells to TCC-SUP cells,<br />

but there was an increase for HeLa cells. The metabolic activity of HeLa cells<br />

decreased after a period of 24h of <strong>Candida</strong> infection, especially in the presence C.<br />

tropicalis. The biological response of TCC-SUP cells was different from the one<br />

observed with HeLa cells, the presence of C. albicans and C. parapsilosis induced<br />

an increase in the metabolic activity of TCC-SUP cells, while C. tropicalis and C.<br />

glabrata promoted a decrease.. After 24h of C. albicans adhesion, the highest<br />

human cells damage was detected in HeLa cells (measured by LDH release), while<br />

C.tropicalis seems to induce higher LDH release after 24h in contact with TCC-SUP<br />

cells. The biological response of human cells showed to be dependent on their<br />

origin and on the colonizing yeast species/strain, and no direct relation between<br />

the number of adherent cells and the biological response was observed.<br />

178


179


Poster number: 33<br />

Using zebrafish to study <strong>Candida</strong> albicans-mediated<br />

systemic candidiasis in vivo<br />

Thomas Evans, Simon Tazzyman, Timothy J.A. Chico, Martin H. Thornhill,<br />

Craig Murdoch.<br />

University of Sheffield, Sheffield, UK<br />

<strong>Candida</strong> albicans is a common oral opportunistic pathogen that causes<br />

mucocutaneous infections and rarely systemic candidiasis, a nosocomial infection<br />

which involves the haematogenous spread of <strong>Candida</strong> to multiple organs and is<br />

associated with high mortality. To cause tissue damage C.albicans must bind to the<br />

endothelium of blood vessels, leave the circulation and invade tissues. However,<br />

little is known about the mechanisms involved in these processes. This study aimed<br />

to use transparent embryonic zebrafish (Danio rerio) as an in vivo infection model<br />

in conjunction with an in vitro flow adhesion assay to study how C.albicans bind to<br />

the endothelium, leave the circulation and invade tissues. Suspensions of viable or<br />

treated C.ablicans were flowed over monolayers of endothelial cells and adherent<br />

<strong>Candida</strong> counted. Zebrafish were injected with C.albicans and mortality determined<br />

after 18h. Time-lapse confocal microscopy using fluorescent C.albicans and blood<br />

vessels was performed to image dissemination. Viable C.albicans bound to<br />

endothelium significantly (p


181


Poster number: 34<br />

<strong>Candida</strong> albicans Secreted Aspartic Proteases Cause<br />

Inflammatory Response Irrespective of Their Proteolytic<br />

Activity<br />

Neelam Pandey 1 , Donatella Pietrella 1 , Lydia Schild 2 , Francesco Bistoni 1 ,<br />

Bernhard Hube 2 and Anna Vecchiarelli 1<br />

1 Department of Experimental Medicine and Biochemical Science, University of Perugia,<br />

Perugia, Italy; 2 Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for<br />

Natural Product Research and <strong>Infection</strong> <strong>Biology</strong>, Hans Knoell Institute, Jena, Germany2;<br />

<strong>Candida</strong> albicans normally inhabits humans as a commensal, but causes fatal infections<br />

when the immune system of patients is suppressed. It has a strong armory of virulence<br />

attributes which facilitates bypassing the immune system and causes mucosal, cutaneous<br />

and systemic infections. These attributes are the ability to colonize, invade the host tissue<br />

and secrete aspartic proteases. We here report that recombinant secreted aspartic<br />

proteases (rSaps) including rSap1, rSap2, rSap3 and rSap6 induce inflammatory cytokine<br />

production by human monocytes to different degrees. rSap1, rSap2 and rSap6 induced<br />

the secretion of IL-1beta, TNF-alpha and IL-6 significantly, while rSap3 stimulated the<br />

secretion of IL-1beta and TNF-alpha. All these rSaps induced Ca 2+ influx in monocytes.<br />

Pepstatin A, a potential inhibitor of aspartic proteases, has no effect on the cytokine<br />

secretion induced by these rSaps. This suggests that the inflammatory response due to<br />

rSaps is not because of their proteolytic activity, a hypothesis further supported by the<br />

observation that the ability of rSaps to induce inflammatory cytokine secretion was<br />

independent of protease-activated receptor (PAR) activation, and of the optimal pH for<br />

individual rSap activity. Moreover, rSaps stimulated Akt activation and thus induced IkBalpha<br />

phosphorylation which mediated translocation of NFkB into the nucleus in human<br />

monocytes. These findings give evidence that rSap1, rSap2 and rSap3 cause an<br />

inflammatory response irrespective of their proteolytic activity and mediate the inflammatory<br />

response via activation of Akt/NF-kB. The inflammatory response terminates with<br />

inflammasome activation. This occurs through a process requiring Sap internalization via a<br />

clathrin dependent mechanism, and caspase-1 activation, and it is mediated by K + efflux<br />

and ROS production. By demonstrating that the recognized virulence factors of C. albicans<br />

such as Sap2 and Sap6 are potent inducers of inflammasome activation, our results<br />

represent a step forward in the comprehension of the close relationship between fungus<br />

virulence and host response mechanisms.<br />

We are extending our study by using an in vivo experimental model of murine candidiasis<br />

to analyze the role of Sap2 and Sap6 in the inflammatory response in the vaginal candidiasis,<br />

and to investigate whether specific mAbs are indeed able to affect this process.<br />

This study was funded by the European Commission (<strong>FINSysB</strong> Marie Curie Initial Training Network grant PITN-<br />

GA-2008-214004).<br />

182


183


Poster number: 35<br />

The Tyr238X dectin-1 polymorphism is a predisposing<br />

factor for mucosal <strong>Candida</strong> albicans infections but not<br />

candidemia<br />

Diana C. Rosentul 1,2 , Theo S. Plantinga 1,2 , Marije Oosting 1,2 , William K.<br />

Scott 3 , Digna R. Velez Edwards 4 , P. Brian Smith 5 , Barbara D. Alexander 5 ,<br />

John C. Yang 6 , Gregory M. Laird 5 , Walter J. F. M. van der Velden 2,7 , Bart<br />

Ferwerda 1,2 , Annemiek B. van Spriel 2,8 , Gosse Adema 2,8 , Ton Feuth 2,9 , J.<br />

Peter Donnelly 1,2 , Gordon D. Brown 10 , Nicole M. A. Blijlevens 2,7 , Leo A.B.<br />

Joosten 1,2 , Jos W. M. van der Meer 1,2 , John R. Perfect 5 , Bart-Jan Kullberg 1,2 ,<br />

Melissa D. Johnson 5,11 , Mihai G. Netea 1,2<br />

1 Department of Internal Medicine, Radboud University Nijmegen Medical Center, Nijmegen,<br />

The Netherlands; 2 Nijmegen Institute for <strong>Infection</strong>, Inflammation and Immunity (N4i), Radboud<br />

University Nijmegen Medical Center, Nijmegen, The Netherlands; 3 Dr. John T. Macdonald<br />

Foundation Department of Human Genetics and John P. Hussman Institute for Human<br />

Genomics, University of Miami, Miller School of Medicine, Miami, FL, USA; 4 Department of<br />

Obstetrics & Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA; 5 Duke<br />

University Medical Center, Durham, NC, USA; 6 National Jewish Health, Denver, CO, USA;<br />

7 Department of Haematology, Radboud University Nijmegen Medical Center, Nijmegen, The<br />

Netherlands; 8 Department of Tumor Immunology, Radboud University Nijmegen Medical<br />

Center, Nijmegen, The Netherlands; 9 Department of Epidemiology, Biostatistics and Health<br />

Technology Assessment, Radboud University Nijmegen Medical Center, Nijmegen, The<br />

Netherlands; 10 Institute of Infectious Disease and Molecular Medicine, University of Cape<br />

Town, Cape Town, South Africa; 11 Department of Clinical Research, Campbell University<br />

School of Pharmacy, Buies Creek, NC, USA.<br />

The genetic make up of innate immune genes is believed to play an important role<br />

for susceptibility to <strong>fungal</strong> infections. We recently described the Tyr238X dectin-1<br />

single nucleotide polymorphism (SNP) as an important factor influencing<br />

susceptibility to vulvo-vaginal candidiasis and onychomycosis in a Dutch family<br />

with recurrent <strong>fungal</strong> infections.<br />

The aim of our studies was to assess the role of this polymorphism in the<br />

susceptibility to mucosal and systemic <strong>Candida</strong> albicans infections. To achieve this<br />

aim we have investigated the effect of this SNP in several cohorts of patients:<br />

-142 Dutch patients undergoing hematopoietic stem cells transplantation (HSCT).<br />

-331 Dutch and American candidemia patients and 351 non infected matched<br />

controls.<br />

Functional studies have shown that cells isolated from individuals heterozygous for<br />

the 238X allele had a lower cytokine production upon stimulation with <strong>Candida</strong>. The<br />

HSCT patients bearing the early stop variant of the dectin-1 gene presented a<br />

184


higher C. albicans oral an gastrointestinal colonization compared to those who were<br />

wild type for the polymorphism and required higher amounts of fluconazole in order<br />

to avoid a disseminated invasion of the fungus.<br />

In contrast, no effect if the Tyr238X dectin-1 SNP on the susceptibility to candidemia<br />

and/or the clinical course of the infection was observed.<br />

In conclusion, the dectin-1 signaling pathway is non-redundant in mucosal immunity<br />

to C. albicans. The 238X allele results in a defective IL-6 and IL-17 response, but<br />

normal IFN-gamma, upon stimulation with C. albicans.<br />

A genetic deficiency of beta-glucan recognition has clear effect on the occurrence<br />

of mucosal <strong>Candida</strong> infections, but a minor impact on susceptibility to candidemia.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial Training Network<br />

(PITN-GA-2008-214004).<br />

185


Poster number: 36<br />

Survival of <strong>Candida</strong> glabrata within phagocytes<br />

Lydia Schild, Katja Seider, Pedro Miramón, Sascha Brunke and Bernhard<br />

Hube<br />

Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product<br />

Research and <strong>Infection</strong> <strong>Biology</strong> – Hans Knoell Institute (HKI), Jena, Germany<br />

<strong>Candida</strong> glabrata is both a human commensal and a pathogenic fungus. Immune<br />

evasion strategies likely play a key role during infection since C. glabrata elicits little<br />

inflammation in mice. To investigate immune evasion as a pathogenic strategy, we<br />

characterize the interaction of C. glabrata with human phagocytes such as<br />

macrophages and neutrophils.<br />

C. glabrata is able to survive and even replicate within human macrophages in vitro.<br />

Low pro-inflammatory response to <strong>fungal</strong> infection and inhibition of macrophage<br />

ROS production by C. glabrata support the view that this fungus can persist in<br />

macrophages as an immune evasion strategy. Our data suggest that C. glabrata<br />

modifies phagosome maturation, residing in a non-acidic phagosome. While the<br />

inhibition of ROS production is affected by viability of <strong>fungal</strong> cells, inhibition of<br />

phagosome maturation does not require metabolic activity of the fungus. Instead,<br />

an UV-indestructible <strong>fungal</strong> attribute seems to be involved this process.<br />

To elucidate which C. glabrata factors are responsible for modification of<br />

phagosome maturation and persistence in phagocytes, we analyzed a set of<br />

deletion mutants for survival in macrophages and identified 24 genes that are crucial<br />

for intracellular survival of C. glabrata within macrophages. Phenotypic analyses of<br />

these mutants point to an important role of cell wall integrity and stress resistance<br />

for macrophage survival of C. glabrata. Furthermore, nutrient acquisition, in<br />

particular iron uptake has been shown to play a role during intra-phagosomal<br />

persistence.<br />

Similar attributes seem to be important for resistance to neutrophils, as mutants<br />

lacking genes with suggested functions in cell wall integrity and nutritional sensing<br />

are attenuated in survival in both, neutrophils and macrophages.<br />

186


187


Poster number: 37<br />

The role of pH-regulated antigen 1 of <strong>Candida</strong> albicans in<br />

the interaction of the fungus with human neutrophils<br />

Eliška Svobodová 1 , Josephine Losse 1 , Antje Heyken 2 , Bernhard Hube 2,4 ,<br />

Peter F. Zipfel 3,4 and Mihály Józsi 1<br />

1 Junior Research Group Cellular Immunobiology, 2 Department of Microbial Pathogenicity<br />

Mechanisms and 3 Department of <strong>Infection</strong> <strong>Biology</strong>, Leibniz Institute for Natural Product<br />

Research and <strong>Infection</strong> <strong>Biology</strong> - Hans Knöll Institute, Jena, Germany; 4 Friedrich Schiller<br />

University, Jena, Germany<br />

<strong>Candida</strong> albicans is an opportunistic human-pathogenic yeast and a leading cause<br />

of life-threatening systemic <strong>fungal</strong> infections in immunocompromised individuals.<br />

The virulence of the fungus is associated with its capability to filamentous growth.<br />

The pH-regulated antigen 1 (Pra1) of C. albicans is a surface-associated and<br />

secreted protein whose expression is upregulated in the hyphal form. Pra1 binds<br />

to complement receptor 3 (CD11b/CD18) and can mediate adhesion to and<br />

migration of human phagocytes. Here, we investigated the role of Pra1 in the<br />

activation of human neutrophils. A C. albicans mutant strain lacking Pra1 (pra1 )<br />

supported neutrophil migration to a lower extent than did the parental wild-type<br />

strain. A Pra1-overexpressing C. albicans strain enhanced neutrophil migration and<br />

adherence. While inactivated hyphae of the Pra1-overexpressing mutant enhanced<br />

the production and release of reactive oxygen species, myeloperoxidase,<br />

lactoferrin, and interleukin 8 by neutrophils, such responses were reduced when<br />

stimulated with the pra1 strain. Pra1-overexpressing living hyphae also caused a<br />

reduced neutrophil activation, indicating that <strong>fungal</strong> cells which express and release<br />

more Pra1 can more efficiently inhibit the activation of these innate immune cells.<br />

Fungal cells lacking Pra1 were more efficiently killed by neutrophils. The lack or the<br />

overexpression of Pra1 did not significantly affect the generation of neutrophil<br />

extracellular traps. In conclusion, surface-exposed Pra1 plays a role in the<br />

recognition of C. albicans, especially hyphal cells, by human neutrophils and<br />

enhances neutrophil antimicrobial responses. However, the fungus can counteract<br />

some of these defense mechanisms likely by releasing Pra1.<br />

188


189


190


Defeating The Enemy<br />

191


Poster number: 38<br />

Alkali-metal-cation homeostasis of <strong>Candida</strong> species is<br />

affected by subinhibitory concentrations of azoles<br />

Hana Elicharová, Hana Sychrová<br />

Department of Membrane Transport, Institute of Physiology AS CR, v. v. i., Videnska 1083,<br />

14220 Prague, Czech Republic, elicharova@biomed.cas.cz<br />

Homeostasis of alkali-metal cations is essential for all living cells including the<br />

pathogenic <strong>Candida</strong> species. <strong>Candida</strong> cells sustain high internal concentration of<br />

potassium cations that are involved in osmotic and pH regulations, enzymatic<br />

function and other processes. In contrast, <strong>Candida</strong> cells keep the internal<br />

concentration of sodium low due to its intracellular toxicity. Plasma membrane and<br />

transporters play a key role in maintenance of cation homeostasis. Azoles form the<br />

most important group of drugs with anti<strong>fungal</strong> activity. They destabilize plasma<br />

membrane of <strong>Candida</strong> cells. The combination of subinhibitory concentrations of<br />

fluconazole and NaCl has been shown to inhibit the growth of both fluconazolesensitive<br />

and fluconazole-resistant C. albicans strains (1).<br />

We have tested the influence of subinhibitory concentrations of azoles on the alkalimetal-cation<br />

homeostasis of four <strong>Candida</strong> species (C. albicans, C. dubliniensis,<br />

C. parapsilosis and C. glabrata).We have found that all tested <strong>Candida</strong> species can<br />

generally tolerate high concentrations of azoles or salts separately, but the<br />

combination of both compounds inhibits their growth effectively. The level of<br />

inhibition of single <strong>Candida</strong> species differs strongly. Presence of subinhibitory<br />

concentration of fluconazole increases the intracellular content of toxic Na + in all<br />

four <strong>Candida</strong> species. Estimation of relative membrane potential together with the<br />

use of <strong>Candida</strong> mutants lacking plasma-membrane transporters of alkali metal<br />

cations should explain how alkali-metal-cation homeostasis is affected by presence<br />

of fluconazole.<br />

(1) Kolecka et al., Can J Microbiol 2009 55(5):605-10<br />

Supported by MSMT LC531<br />

192


193


Poster number: 39<br />

Calcineurin Signaling and Membrane Lipid Homeostasis<br />

Regulates Iron Mediated MultiDrug Resistance<br />

Mechanisms in <strong>Candida</strong> albicans<br />

Saif Hameed, Sanjiveeni Dhamgaye, Ashutosh Singh, Shyamal K. Goswami<br />

and Rajendra Prasad<br />

School of Life Sciences, Jawaharlal Nehru University, New Delhi-110067, India<br />

We previously demonstrated that iron deprivation enhances drug susceptibility of<br />

<strong>Candida</strong> albicans by increasing membrane fluidity which correlated with the lower<br />

expression of ERG11 transcript and ergosterol levels. The iron restriction dependent<br />

membrane perturbations led to an increase in passive diffusion and drug<br />

susceptibility. The mechanisms underlying iron homeostasis and multidrug<br />

resistance (MDR), however, are not yet resolved. To evaluate the potential<br />

mechanisms, we used whole genome transcriptome and electrospray ionization<br />

tandem mass spectrometry (ESI-MS/MS) based lipidome analyses of iron deprived<br />

<strong>Candida</strong> cells to examine the new cellular circuitry of the MDR of this pathogen.<br />

Our transcriptome data revealed a link between calcineurin signaling and iron<br />

homeostasis. Among the several categories of iron deprivation responsive genes,<br />

the down regulation of calcineurin signaling genes including HSP90, CMP1 and<br />

CRZ1 was noteworthy. Interestingly, iron deprived <strong>Candida</strong> cells as well as iron<br />

acquisition defective mutants phenocopied molecular chaperone HSP90 and<br />

calcineurin mutants and thus were sensitive to alkaline pH, salinity and membrane<br />

perturbations. In contrast, sensitivity to above stresses did not change in iron<br />

deprived DSY2146 strain with a hyperactive allele of calcineurin. Although, iron<br />

deprivation phenocopied compromised HSP90 and calcineurin, it was independent<br />

of protein kinase C signaling cascade. Notably, the phenotypes associated with<br />

iron deprivation in genetically impaired calcineurin and HSP90 could be reversed<br />

with iron supplementation. The observed down regulation of ergosterol (ERG1,<br />

ERG2, ERG11 and ERG25) and sphingolipid biosynthesis (AUR1 and SCS7) genes<br />

followed by lipidome analysis confirmed that iron deprivation not only disrupted<br />

ergosterol biosynthesis, but it also affected sphingolipid homeostasis in <strong>Candida</strong><br />

cells. These lipid compositional changes suggested extensive remodeling of the<br />

membranes in iron deprived <strong>Candida</strong> cells. Taken together, our data provide the<br />

first novel insight into the intricate relationship between cellular iron, calcineurin<br />

signaling, membrane lipid homeostasis and drug susceptibility of <strong>Candida</strong> cells.<br />

194


195


Poster number: 40<br />

Wall proteins, absolute quantification and concatenated<br />

vaccines: A new approach for <strong>Candida</strong> albicans therapy?<br />

Clemens J. Heilmann, Alice G. Sorgo, Henk L. Dekker, Stanley Brul, Chris<br />

G. de Koster, Leo J. de Koning and Frans M. Klis<br />

Swammerdam Institute for Life Sciences, Universiteit van Amsterdam, Science Park 904,<br />

1098XH Amsterdam<br />

The cell wall and especially the proteins that are covalently anchored to it are the<br />

first site of host-pathogen interaction. The wall proteins are promising targets for<br />

the development of diagnostic biomarkers and vaccines. Previously, we described<br />

the relative quantitative changes in up to 21 wall proteins during the yeast-to-hypha<br />

transition (1) and upon exposure of cells to fluconazole (2). We identified hyphaassociated<br />

wall proteins (Als3, Hwp2, Hyr1, Plb5, Sod5) conferring increased<br />

adhesion and resistance to host defenses and yeast-associated proteins (Rhd3,<br />

Sod4, Ywp1), which probably play a role in host dispersal and immune evasion.<br />

Morphotype-independent proteins (Cht2, Crh11, Ecm33, Mp65) are mainly involved<br />

in wall structure and remodeling.<br />

However, relative quantification can only provide indications of how protein<br />

abundance changes in response to an environmental change. To compare the<br />

abundances of different proteins, absolute quantification is required. We are<br />

developing a strategy for absolute quantification based on 15 N metabolic labeling<br />

and concatenated proteins (Q-CON-CAT). By using the same 15 N reference culture<br />

that was also used for relative quantification and determining the absolute amount<br />

of a particular protein, we will be able to recalculate the relative quantification results<br />

into absolute values.<br />

These absolute values will be informative for the construction of peptide-based<br />

vaccines. We propose that by concatenating peptides that are predicted to be<br />

highly immunogenic and originate from abundant wall proteins an effective Q-CON-<br />

CAT vaccine protein can be generated.<br />

(1) Hyphal Induction in the human <strong>fungal</strong> pathogen <strong>Candida</strong> albicans reveals a characteristic wall protein<br />

profile. Heilmann CJ, Sorgo AG, Siliakus AR, Dekker HL, Brul S, de Koster CG, de Koning LJ, Klis FM.<br />

Microbiology. 2011 May 20.<br />

(2) The effects of fluconazole on the secretome, the wall proteome and wall integrity of the clinical fungus<br />

<strong>Candida</strong> albicans. Sorgo AG, Heilmann CJ, Dekker HL, Bekker M, Brul S, de Koster CG, de Koning LJ,<br />

Klis FM.Eukaryot Cell. 2011 May 27.<br />

We are grateful to the European Commission for funding the <strong>FINSysB</strong> Marie Curie Initial Training Network<br />

(PITN-GA-2008-214004).<br />

196


197


Poster number: 41<br />

Dynamic assessment of the anti<strong>fungal</strong> effect of different<br />

plant essential oils by flow cytometry<br />

Crina Maria Saviuc 1,5 , Alexandru Mihai Grumezescu 2 , Coralia Bleotu 1,4 ,<br />

Alina Maria Holban 1 , Otilia Banu 3 , Dan Mihaiescu 2 , Paul Balaure 2 , Mariana<br />

Carmen Chifiriuc 1 , Veronica Lazar 1<br />

1 University of Bucharest, Faculty of <strong>Biology</strong>, Microbiology University Department; 2 University<br />

Politechnica of Bucharest, Faculty of Applied Chemistry and Materials Science, Organic<br />

Chemistry Department; 3 Institute of Cardiovascular Diseases Prof. C.C. Iliescu, Bucharest;<br />

4 S. Nicolau Institute of Virology, Bucharest; 5 S.C. Biotehnos S.A.<br />

Introduction: The in vitro and in vivo models already demonstrated the microbicidal<br />

as well as the influence of the essential oils on the coordinated expression of the<br />

virulence factors in <strong>fungal</strong> strains. In the context of incomplete standardization of<br />

susceptibility testing assays, our aim was to evaluate a rapid tool for the<br />

assessment of the anti<strong>fungal</strong> activity of the essential oils using the flow citometry.<br />

Materials and methods: Four essential oils were extracted by microwave assisted<br />

hydrodistilation from different vegetative and reproduction organs of Eugenia<br />

caryophyllata, Anethum graveolens, Mentha piperita and Rosmarinus officinalis.<br />

Chemical composition of the essential oils was established by GC-MS analysis.<br />

The bioassays were performed on five <strong>Candida</strong> spp. strains recently isolated from<br />

clinical specimens. The qualitative screening of the antimicrobial effect and its<br />

evolution on time was assessed after 1’, 3’, 5’, 15’ and 30’ of contact by viable cell<br />

counts. Minimum inhibitory concentrations (MIC) was assessed by the classical<br />

twofold micro-dilution technique in 96 multiwell plates and also by flow cytometry<br />

after 15’ and 24h of contact.<br />

Results. The essential oils proved to be reach in terpenes and terpenoids in a<br />

balanced composition wich explained the antimicrobial activity. The essential oil<br />

exhibited an early microbicidal effect, with MIC ranging from 0.75 to 7.5µL/mL,<br />

values also confirmed by flow cytometry.<br />

Conclusion. Our results are proving that the tested essential oils exhibited a strong<br />

anti<strong>fungal</strong> effect, probably due to their good diffusion rates and easy contact with<br />

the target structures. Essential oils complex composition and their volatility raise<br />

specific problems in susceptibility testing, due to their poor solubility, unknown<br />

diffusion pattern in solid media and vapor phase effect. In this context flow<br />

cytometry technique could be considered a reliable tool for evaluating the anti<strong>fungal</strong><br />

activity of this type of complex mixtures.<br />

198


199


Poster number: 42<br />

The role of <strong>fungal</strong> cell wall composition and remodelling in<br />

echinocandin drug tolerance<br />

Louise Walker, Kerstin Nather, Laura Selway, David Stead, Keunsook Lee,<br />

Alistair Brown, Carol Munro<br />

Institute of Medical Sciences, School of Medical Sciences, University of Aberdeen, Aberdeen, UK<br />

Echinocandins are anti<strong>fungal</strong> agents, which affect <strong>fungal</strong> cell wall integrity by<br />

inhibiting (1,3)-glucan synthesis. Assembly of the <strong>fungal</strong> cell wall is a dynamic<br />

process. Echinocandin treatment below the MIC activates compensatory<br />

mechanisms that upregulate chitin biosynthesis and result in altered cross-linking<br />

and changes in the cell wall proteome, in an attempt to reinforce the cell wall.<br />

Proteomic analysis and transcript profiling have identified cell wall proteins (CWP)<br />

and genes that are more abundant and have elevated expression in response to<br />

treatment with sub-MIC caspofungin. These include transglycosidases and crosslinking<br />

proteins involved in modifying cell wall polysaccharides (Utr2, Crh11, Phr1,<br />

Cht2), as well as novel proteins (Pga31). Changes in cell wall architecture and<br />

composition can alter a cell’s sensitivity to the echinocandins. Using a tetracycline<br />

inducible promoter, genes of CWPs which were upregulated in response to<br />

caspofungin treatment, were overexpressed to determine whether this altered<br />

susceptibility to caspofungin. Overexpression of CHT2 lead to reduced<br />

susceptibility whereas overexpression of UTR2 and PHR1 resulted in increased<br />

sensitivity to caspofungin. To determine whether altered susceptibility to<br />

caspofungin was observed in vivo, larvae of Galleria mellonella were used as a<br />

preliminary screening model. Overexpression of UTR2 lead to reduced susceptibility<br />

to caspofungin in G. mellonella. Treatment with caspofungin increased the survival<br />

time of larvae infected with the UTR2 overexpression strain compared to larvae<br />

infected with the wild-type. Fungal burdens were significantly reduced in infected<br />

larvae treated with caspofungin and this reduction was greater in larvae infected<br />

with the UTR2 overexpression strain. The number of immune cells also determines<br />

the fitness of G. mellonella. Larvae infected with wild-type and UTR2 overexpression<br />

strain had more immune cells after treatment with caspofungin, indicating an<br />

increase in fitness. Therefore, overexpression of <strong>fungal</strong> cell surface proteins can<br />

affect the susceptibility of C. albicans to caspofungin in vitro and in vivo.<br />

200


201


Poster number: 43<br />

Genetic code ambiguity accelerates evolution of drug<br />

resistance in <strong>Candida</strong> albicans<br />

Tobias Weil, Ana R. Bezerra, João Simões and Manuel A. S. Santos<br />

Department of <strong>Biology</strong> – CESAM, University of Aveiro, 3810-193 Aveiro, Portugal.<br />

The opportunistic <strong>fungal</strong> pathogen <strong>Candida</strong> albicans has evolved to survive within<br />

human hosts where it can cause serious diseases in immunocompromised patients.<br />

The emergence of drug-resistant C. albicans strains has become a severe clinical<br />

problem and the mechanisms of <strong>fungal</strong> drug resistance are still poorly understood.<br />

In the present study we are testing the hypothesis that mRNA mistranslation is<br />

relevant for the evolution of drug resistance. A characteristic feature of C. albicans<br />

biology is the reassignment of the leucine CUG codon to serine. Remarkably, the<br />

serine tRNA that decodes the CUG codons is aminoacylated in vivo with both serine<br />

(3%) and leucine (97%). Such tRNA ambiguity is incorporated into proteins and<br />

creates a statistical proteome. It is not yet clear whether proteome variation is<br />

relevant for C. albicans parthogenesis, however, the potential of CUG ambiguity to<br />

alter cell wall proteins and remodel surface antigens may help C. albicans evading<br />

the immune system and gain advantages in terms of drug resistance. To determine<br />

whether increasing mistranslation levels have important implications for C. albicans<br />

drug resistance we have constructed a series of C. albicans strains that mistranslate<br />

leucine CUG codons as serine at a rate up to 65% and exposed them to azoles,<br />

which are commonly used to treat patients with <strong>fungal</strong> infections. Interestingly,<br />

these strains developed drug resistance quite rapidly confirming our hypothesis<br />

that mistranslation is indeed relevant for drug resistance. We are now applying in<br />

vivo approaches combined with systems biology techniques based on gene<br />

expression profiling with microarrays and deep genome sequencing in order to<br />

identify novel putative drug target genes and pathways.<br />

Acknowledgements: TW is supported by the European FP7 framework programme<br />

SYBARIS.<br />

202


203


204


Barkay, Naama<br />

Weizmann Institute<br />

Meyer Building 404<br />

Rehovot 76100<br />

Israel<br />

naama.barkai@weizmann.ac.il<br />

Bohovych, Iryna<br />

University of Aberdeen<br />

Institute of Medical Science<br />

Aberdeen, AB25 2ZD, UK<br />

i.bohovych@abdn.ac.uk<br />

Botelho, Claudia<br />

University of Minho<br />

Centre of Biological Engineering<br />

4710-057 Braga<br />

Portugal<br />

claudiabotelho@deb.uminho.pt<br />

Brown, Alistair<br />

University of Aberdeen<br />

Institute of Medical Science<br />

Aberdeen<br />

AB25 2ZD, UK<br />

al.brown@abdn.ac.uk<br />

Brown, Gordon<br />

University of Aberdeen<br />

Institute of Medical Science<br />

Aberdeen<br />

AB25 2ZD, UK<br />

gordon.brown@abdn.ac.uk<br />

Cabral, Vitor<br />

Pasteur Institute<br />

25 rue du Docteur Roux<br />

75015, Paris<br />

France<br />

vitor.cabral@pasteur.fr<br />

Cassone, Antonio<br />

Istituto Superiore di Sanità<br />

Viale Regina Elena 299<br />

00161 Rome<br />

Italy<br />

antonio.cassone@iss.it<br />

List of Participants:<br />

205<br />

Cavalieri, Duccio<br />

Università di Firenze<br />

Viale Pieraccini 6<br />

50139, Firenze<br />

Italy<br />

duccio.cavalieri@unifi.it<br />

Chen, Shin-Chin<br />

Radboud University Nijmegen Medical<br />

Center<br />

Geert Grooteplein-Zuid 10<br />

6525 GA Nijmegen<br />

The Netherlands<br />

S.Cheng@AIG.umcn.nl<br />

Citulio, Francesco<br />

Hans Knoell Institute (HKI)<br />

Beutenbergstrasse 11a<br />

07745 Jena<br />

Germany<br />

francesco.citiulo@hki-jena.de<br />

Cook, Emily<br />

University of Exeter<br />

Geoffrey Pope Building<br />

Exeter, EX4 4QD, UK<br />

E.Cook@exeter.ac.uk<br />

Correia, Inês<br />

University of Madrid<br />

Plaza de ramón y Cajal s/n<br />

28040 Madrid<br />

Spain<br />

inescorreia351@gmail.com<br />

D’Enfert, Christophe<br />

Pasteur Institute<br />

25 rue du Docteur Roux<br />

75015, Paris<br />

France<br />

denfert@pasteur.fr<br />

Dobb, Kate<br />

F2G Ltd<br />

PO Box 1, Lankro Way<br />

Eccles<br />

Manchester, M30 0BH, UK<br />

KDobb@f2g.com


Donohue, Dagmara<br />

Vrije Universiteit Brussel<br />

Pleinlaan 2<br />

1050 Brussels<br />

Belgium<br />

dszestak@vub.ac.be<br />

Elicharová, Hana<br />

Academy of Sciences CR<br />

Videnska 1083<br />

14220 Prague 4<br />

Czech Republic<br />

elicharova@biomed.cas.cz<br />

Ene, Iuliana<br />

University of Aberdeen<br />

Institute of Medical Science<br />

Aberdeen<br />

AB25 2ZD, UK<br />

iuliana.ene@abdn.ac.uk<br />

Ernst, Joachim<br />

University of Düsseldorf<br />

Universitaetsstr. 1, Geg 26.12<br />

40225 Düsseldorf<br />

Germany<br />

joachim.ernst@uni-duesseldorf.de<br />

Filler, Scott<br />

Harbor-UCLA Medl Cntr-Inf Dis<br />

1124 West Carson Street<br />

Torrance, CA 90509<br />

USA<br />

sfiller@ucla.edu<br />

Gil de Bona, Ana<br />

University of Madrid<br />

Plaza de ramón y Cajal s/n<br />

28040 Madrid<br />

Spain<br />

anagildebona@farm.ucm.es<br />

Gustin, Mike<br />

Rice University<br />

W100 George R. Brown Hall<br />

P.O. Box 1892, MS-140,<br />

Houston, Texas 77251-1892<br />

USA<br />

gustin@rice.edu<br />

206<br />

Hameed, Saif<br />

School of Life Sciences,J.N.U<br />

Membrane <strong>Biology</strong> Lab #101<br />

New Delhi-110067<br />

India<br />

saifhameed@yahoo.co.in<br />

Hassan, Rabeay<br />

Helmholtz Centre for <strong>Infection</strong> Research<br />

Inhoffenstr. 7<br />

38124 Braunschweig<br />

Germany<br />

Rabeay.Hassan@helmholtz-hzi.de<br />

Heilmann, Clemens<br />

University of Amsterdam<br />

Nieuwe Achtergracht 166<br />

1018 WV Amsterdam<br />

The Netherlands<br />

C.J.Heilmann@uva.nl<br />

Henriques, Mariana<br />

University of Minho<br />

Centre of Biological Engineering<br />

4710-057 Braga<br />

Portugal<br />

mcrh@deb.uminho.pt<br />

Holban, Alina<br />

University of Bucharest<br />

Faculty of <strong>Biology</strong>,<br />

Portocalelor Street, nr 1-3<br />

Bucharest<br />

Romania<br />

alina_m_h@yahoo.com<br />

Hube, Bernhard<br />

Hans Knoell Institute (HKI)<br />

Beutenbergstrasse 11a<br />

07745 Jena<br />

Germany<br />

Bernhard.Hube@hki-jena.de<br />

Hyde, Peter<br />

Trinity College Dublin<br />

Lincoln Place, Dublin 2,<br />

Ireland<br />

Peter.Hyde@dental.tcd.ie


Ishchuk, Olena<br />

Lund University<br />

Sölvegatan 35<br />

Lund SE-223 62<br />

Sweden<br />

olenkaishchuk@gmail.com<br />

Jacobsen, Ilse<br />

Hans Knoell Institute (HKI)<br />

Beutenbergstrasse 11a<br />

07745 Jena<br />

Germany<br />

Ilse.Jacobsen@hki-jena.de<br />

Jandric, Zeljkica<br />

University of Vienna<br />

Dr.Bohr-Gasse 9<br />

A-1030 Vienna<br />

Austria<br />

zeljkica.jandric@univie.ac.at<br />

Juchimiuk, Mateusz<br />

Polish Academy of Sciences<br />

Pawinskiego 5a<br />

02-106 Warszawa<br />

Poland<br />

mjuchimiuk@ibb.waw.pl<br />

Klipp, Edda<br />

Humboldt University<br />

Theoretical Biophysics<br />

Invalidenstraße 42<br />

D-10115 Berlin, Germany<br />

edda.klipp@rz.hu-berlin.de<br />

Klis, Frans<br />

University of Amsterdam<br />

Nieuwe Achtergracht 166<br />

1018 WV Amsterdam<br />

The Netherlands<br />

F.M.Klis@uva.nl<br />

Kos, Iaroslava<br />

INRA/AGROPARITECH<br />

UMR Micalis 1319<br />

CBAI<br />

78850 Thiverval Grignon, France<br />

ikos@grignon.inra.fr<br />

207<br />

Kumamoto, Carol<br />

Tufts University<br />

136 Harrison Ave.,<br />

Boston, MA,<br />

USA<br />

carol.kumamoto@tufts.edu<br />

Kumar, Antresh<br />

School of Life Sciences, J.N.U<br />

Membrane <strong>Biology</strong> Lab #101<br />

New Delhi-110067<br />

India<br />

antreshkumar@gmail.com<br />

Lagadec, Quentin<br />

University of Düsseldorf<br />

Universitaetsstr. 1, Geg 26.12<br />

40225 Düsseldorf<br />

Germany<br />

quentin.lagadec@uni-duesseldorf.de<br />

Lavie-Richard, Mathias<br />

INRA/AGROPARITECH<br />

UMR Micalis 1319<br />

CBAI<br />

78850 Thiverval Grignon, France<br />

mathias.lavie-richard@grignon.inra.fr<br />

Leach, Michelle<br />

University of Aberdeen<br />

Institute of Medical Science<br />

Aberdeen<br />

AB25 2ZD, UK<br />

michelle.leach@abdn.ac.uk<br />

Lipke, Peter<br />

Brooklyn College<br />

City University New York<br />

2900 Bedford Avenue, Brooklyn,<br />

NY 11210, USA<br />

plipke@brooklyn.cuny.edu<br />

Liu, Haoping<br />

University of California, Irvine<br />

D250, D288 MED SCI I<br />

Irvine, CA 92697<br />

USA<br />

h4liu@uci.edu


Lorenz, Michael<br />

University of Texas<br />

6431 Fannin, Houston<br />

TX 77030<br />

USA<br />

Michael.Lorenz@uth.tmc.edu<br />

Mantovani, Alberto<br />

University of Milan<br />

MANZONI 56,<br />

20089 ROZZANO<br />

ITALY<br />

Alberto.Mantovani@humanitasresearch.it<br />

McLean, Malcolm<br />

Weizmann Institute<br />

Meyer Building 404<br />

Rehovot 76100<br />

Israel<br />

malcolm.mclean@weizmann.ac.il<br />

Miramón, Pedro<br />

Hans Knoell Institute (HKI)<br />

Beutenbergstrasse 11a<br />

07745 Jena<br />

Germany<br />

pedro.miramon@hki-jena.de<br />

Miranda, Isabel<br />

University of Porto<br />

Alameda Prof. Hernani Monteiro<br />

4200-319 Porto<br />

PORTUGAL<br />

imiranda@med.up.pt<br />

Mitchell, Aaron<br />

Carnegie Mellon University<br />

4400 Fifth Avenue,<br />

Pittsburgh, PA,<br />

USA, 15213<br />

apm1@cmu.edu<br />

Morschhäuser, Joachim<br />

Universität Würzburg<br />

Josef-Schneider-Str. 2, Bau D15<br />

97080 Würzburg<br />

Germany<br />

joachim.morschhaeuser@uni-wuerzburg.de<br />

208<br />

Munro, Carol<br />

University of Aberdeen<br />

Institute of Medical Science<br />

Aberdeen<br />

AB25 2ZD, UK<br />

c.a.munro@abdn.ac.uk<br />

Murdoch, Craig<br />

University of Sheffield<br />

Claremont Crescent<br />

Sheffield<br />

S10 2TA, UK<br />

c.murdoch@sheffield.ac.uk<br />

Naglik, Julian<br />

King's College London<br />

St Thomas Street<br />

London<br />

SE1 9RT, UK<br />

julian.naglik@kcl.ac.uk<br />

Netea, Mihai<br />

Radboud University Nijmegen Medical<br />

Center<br />

Geert Grooteplein-Zuid 10<br />

6525 GA Nijmegen<br />

The Netherlands<br />

M.Netea@aig.umcn.nl<br />

Oliver, Jason<br />

F2G Ltd<br />

PO Box 1, Lankro Way<br />

Eccles<br />

Manchester, M30 0BH, UK<br />

O’Neil, Deborah<br />

NovaBiotics Ltd<br />

Cruickshank Building, Craibstone<br />

Aberdeen<br />

AB21 9TR, UK<br />

deborah@novabiotics.co.uk<br />

Pandey, Neelam<br />

University of Perugia<br />

Via del Giochetto<br />

06122 Perugia<br />

ITALY<br />

neelambiotech@yahoo.co.in


Pärnänen, Pirjo<br />

University of Helsinki<br />

P.O.Box 56 (Viikinkaari 5D)<br />

FIN-00014<br />

Finland<br />

pirjo.parnanen@helsinki.fi<br />

Popolo, Laura<br />

Università degli Studi di Milano<br />

Via Celoria 26<br />

Milano<br />

Italy<br />

laura.popolo@unimi.it<br />

Porter, Andy<br />

University of Aberdeen<br />

Institute of Medical Science<br />

Aberdeen<br />

AB25 2ZD, UK<br />

a.porter@abdn.ac.uk<br />

Posas, Francesc<br />

Universitat Pompeu Fabra<br />

Parc de Recerca Biomèdica de Barcelona<br />

Doctor Aiguader, 88 ; E-08003 Barcelona<br />

Spain<br />

francesc.posas@upf.edu<br />

Quinn, Janet<br />

Newcastle University<br />

I Framlington Place<br />

Newcastle upon Tyne<br />

NE2 4HH<br />

janet.quinn@ncl.ac.uk<br />

Rosentul, Diana<br />

Radboud University Nijmegen Medical<br />

Center<br />

Geert Grooteplein-Zuid 10<br />

6525 GA Nijmegen<br />

The Netherlands<br />

D.RosentulAmram@aig.umcn.nl<br />

Ruland, Jürgen<br />

Technical University Munich<br />

Ismaningerstr. 22,<br />

81675 München,<br />

Germany<br />

jruland@lrz.tum.de<br />

209<br />

Saviuc, Crina<br />

University of Bucharest<br />

Faculty of <strong>Biology</strong>,<br />

Portocalelor Street, nr 1-3<br />

Bucharest<br />

Romania<br />

crina.saviuc@yahoo.com<br />

Saxena, Abishek<br />

University of Aberdeen<br />

Institute of Medical Science<br />

Aberdeen<br />

AB25 2ZD, UK<br />

abhisheksaxena@abdn.ac.uk<br />

Schild, Lydia<br />

Hans Knoell Institute (HKI)<br />

Beutenbergstrasse 11a<br />

07745 Jena<br />

Germany<br />

Lydia.Schild@hki-jena.de<br />

Schröppel, Klaus<br />

University of Tübingen,<br />

Elfriede-Aulhorn-Straße 6<br />

Tübingen<br />

Germany<br />

Klaus.Schroeppel@med.uni-tuebingen.de<br />

Christoph Schüller<br />

University of Vienna<br />

Dr.Bohr-Gasse 9<br />

A-1030 Vienna<br />

Austria<br />

Christoph.Schueller@univie.ac.at<br />

Sedikki, Sidi Mohammed Lahbib<br />

Université Abou bekr belkaid,<br />

BP 119, Imama<br />

13000 Tlemcen<br />

Algeria<br />

sdadek@hotmail.com<br />

Silva-Dias, Ana<br />

University of Porto<br />

Alameda Prof. Hernani Monteiro<br />

4200-319 Porto<br />

PORTUGAL<br />

asilvadias@med.up.pt


Silva, Sónia<br />

University of Minho<br />

Centre of Biological Engineering<br />

4710-057 Braga<br />

Portugal<br />

soniasilva@deb.uminho.pt<br />

Sorgo, Alice<br />

University of Amsterdam<br />

Nieuwe Achtergracht 166<br />

1018 WV Amsterdam<br />

The Netherlands<br />

a.g.sorgo@uva.nl<br />

Sullivan, Derek<br />

Trinity College Dublin<br />

Lincoln Place<br />

Dublin 2<br />

Ireland<br />

derek.sullivan@dental.tcd.ie<br />

Svobodová, Eliška<br />

Hans Knoell Institute (HKI)<br />

Beutenbergstrasse 11a<br />

07745 Jena<br />

Germany<br />

Eliska.Svobodova@hki-jena.de<br />

Tillmann, Anna<br />

University of Aberdeen<br />

Institute of Medical Science<br />

Aberdeen<br />

AB25 2ZD, UK<br />

anna.tillmann@abdn.ac.uk<br />

Tyc, Katarzyna<br />

Humboldt-University<br />

Theoretical Biophysics<br />

Invalidenstraße 42<br />

D-10115 Berlin, Germany<br />

katarzyna.tyc@staff.hu-berlin.de<br />

Usher, Jane<br />

University of Exeter<br />

Geoffrey Pope Building<br />

Exeter<br />

EX4 4QD, UK<br />

J.Usher@exeter.ac.uk<br />

210<br />

Vecchiarelli, Anna<br />

University of Perugia<br />

Via del Giochetto<br />

06122 Perugia<br />

ITALY<br />

vecchiar@unipg.it<br />

Walker, Louise<br />

University of Aberdeen<br />

Institute of Medical Science<br />

Aberdeen<br />

AB25 2ZD, UK<br />

louise.walker@abdn.ac.uk<br />

Weil, Tobias<br />

University of Aveiro<br />

Campus Universitário de Santiago<br />

3810-193 Aveiro<br />

Portugal<br />

tobias@ua.pt<br />

Whiteway, Malcolm<br />

NRC/BRI<br />

6100 Royalmount Ave,<br />

Montreal, Quebec<br />

Canada H4P 2R2<br />

malcolm.whiteway@nrc-cnrc.gc.ca<br />

Wilson, Duncan<br />

Hans Knoell Institute (HKI)<br />

Beutenbergstrasse 11a<br />

07745 Jena<br />

Germany<br />

Duncan.Wilson@hki-jena.de<br />

Zabke, Claudia<br />

University of Aberdeen<br />

Institute of Medical Science<br />

Aberdeen<br />

AB25 2ZD, UK<br />

c.zabke@abdn.ac.uk<br />

Znaidi, Sadri<br />

Pasteur Institute<br />

25 rue du Docteur Roux<br />

75015, Paris<br />

France<br />

sadri_znaidi@hotmail.com


211


212

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!