,p5hv)xq )lqdo 0hhwlqj 0dulh &xulh ,qlwldo...

51
ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria Introduction 1 ImResFun2017 - Final Meeting Marie Curie Initial Training Network Molecular Mechanisms of Fungal Pathogen- Host Interactions Hotel Grauer Bär**** Innsbruck, Austria; September 3 – September 7, 2017 ImResFun2017 Organizers: Karl Kuchler (Medical University Vienna, Austria) Steffen Rupp (Fraunhofer-Institut IGB, Germany) Members of the ImResFun2017 Scientific Advisory Board Christophe D'Enfert (FR) Patrick van Dijck (BE) Bernhard Hube (DE) ImResFun2017 Office & Technical Support & Homepage & Registration ImResFun2017 Office: Christa Gregori-Schüller & Maria Schwarzmüller Medical University Vienna Department of Medical Biochemistry Campus Vienna Biocenter, Dr. Bohr-Gasse 9/2 A-1030 Vienna, Austria Phone: +43-1-4277-61806 FAX: +43-1-4277-9618 e-mail: [email protected] Web: http://www.imresfun.org/finalmeeting/ At the Venue: Christa Gregori-Schüller & Maria Schwarzmüller Hotel Grauer Bär**** Innsbruck Universitätsstrasse 5-7; A-6020 Innsbruck, Austria Phone: +43-512-5924-0 FAX: +43-512-574535 Registration & Web: Walter Glaser Technical Management, Database & Registration

Upload: lamtuyen

Post on 10-Feb-2019

215 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Introduction 1

ImResFun2017 - Final Meeting Marie Curie Initial Training Network

Molecular Mechanisms of Fungal Pathogen-Host Interactions Hotel Grauer Bär**** Innsbruck, Austria; September 3 – September 7, 2017

ImResFun2017 Organizers: Karl Kuchler (Medical University Vienna, Austria)

Steffen Rupp (Fraunhofer-Institut IGB, Germany)

Members of the ImResFun2017 Scientific Advisory Board

Christophe D'Enfert (FR) Patrick van Dijck (BE) Bernhard Hube (DE)

ImResFun2017 Office & Technical Support & Homepage & Registration

ImResFun2017 Office: Christa Gregori-Schüller & Maria Schwarzmüller Medical University Vienna Department of Medical Biochemistry Campus Vienna Biocenter, Dr. Bohr-Gasse 9/2 A-1030 Vienna, Austria Phone: +43-1-4277-61806 FAX: +43-1-4277-9618 e-mail: [email protected] Web: http://www.imresfun.org/finalmeeting/ At the Venue: Christa Gregori-Schüller & Maria Schwarzmüller Hotel Grauer Bär**** Innsbruck Universitätsstrasse 5-7; A-6020 Innsbruck, Austria Phone: +43-512-5924-0 FAX: +43-512-574535 Registration & Web: Walter Glaser

Technical Management, Database & Registration

Page 2: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Program2

Welcome to ImResFun2017 in Innsbruck!

The Organizers of ImResFun2017 are delighted to welcome you at ImResFun2017, the Final Meeting of the Marie Curie Initial Training Network ImResFun.

ImResFun aimed for high-level training of PhD students and postdoctoral fellows to gain a better understanding of mechanisms driving the interplay of human fungal pathogens with the host immune system. ImResFun also addressed questions how host immunity promotes invasive fungal diseases or clearance of pathogens. Thus, this conference will discuss the state-of-the-art concerning the molecular basis of fungal pathogen-host interactions.

This high-level international scientific meeting concludes the project. Around 80 scientists from all over the world gather at the venue in Innsbruck and discuss with members of the ImResFun consortium key aspects of fungal immunology in an informal atmosphere.

ImResFun2017 will also address the clinical relevance of fungal infections as well as new research avenues for the future, which may become important. Moreover, leaders in the field will discuss strategies how we can increase the awareness and support for the fungal immunology research community in the public as well as in funding agencies.

We are thankful to the great meeting office team with Christa Gregori-Schüller and Maria Schwarzmüller, and the technical support by Walter Glaser, whose efforts have ensured a smooth organization of ImResFun2017.

We are looking forward to an exciting ImResFun2017 conference and to welcoming you in Innsbruck!

K. Kuchler Steffen Rupp

ImResFun2017 Organizers

Main Sponsors of ImResFun2017

Marie-Curie 'Innovative Training Networks' ImResFun grant MC-ITN-606786

Page 3: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Program 3

Scientific Programme of ImResFun2017

Please note that all posters as well as oral presentations must be considered “privileged personal communications”. No data may be cited or used in any kind of verbal or written scientific correspondence with third parties without explicit permission of the presenting author.

Main Symposia & Plenary Lectures

Four symposia dedicated to distinct topics with 13 plenary lectures and 6 chairpersons as discussion leaders constitute the core programme of ImResFun2017.

Keynote Plenary Lectures

The following keynote lectures shall enrich ImResFun2017:

Keynote Opening Lecture (David Denning)

Keynote Plenary Lecture (Gordon Brown)

Poster Sessions

All Posters will be on display during the entire ImResFun2017 meeting. Participants are highly encouraged to take advantage of two evening poster sessions, a vital part of ImResFun2017 to foster interaction and exchange.

Emergencies & Medicare

Please call the front desk for assistance in case of an emergency. Physicians on duty are Dr. Cziep, Wilhelm Greil Strasse 21, A-6020 Innsbruck, Tel. +43 512 57 23 24 or the University Hospital Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Tel. +43 505 04 0.

IMPORTANT – The ImResFun2017 Organizers cannot accept responsibility or liability for personal accidents, injuries, loss or damage to private property of ImResFun2017 participants. Any leisure activities will be entirely at your own risk. Participants are thus strongly advised to make their own arrangements concerning personal insurance and health coverage if necessary or applicable.

Page 4: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Program 4

Detailed Daily Program & Meeting Grid

ImResFun Outline

September 3 Sunday

September 4 Monday

September 5 Tuesday

September 6 Wednesday

September 7 Thursday

Breakfast 7:00 - 8:30 am 7:00 - 8:30 am 7:00 - 8:30 am 7:00 - 8:30 am

Symposia

10:00 am - 6:00 pm Registration & Check-In

8:30 - 12:00 pm Symposium 2 Host Immunology of Fungal Disease Chair: Julian Naglik PL2 – Daniel Kaplan ST1 - Gloria Molero ST2 - Michael Riedelberger PL3 – Matthias Gunzer ST3 - Stefanie Allert ST4 - Gabriela Neves ST5 - Catarina Oliveira Vaz

8:30 - 12:00 pm Symposium 3 Fungal Response / Evasion to Host Defense Chair: Steffen Rupp PL6 – Constantin Urban ST12 - Samir Jawhara ST13 - Filomena Nogueira ST14 - Sabrina Jenull PL7 - James Konopka ST15 - Sarah Sze Wong ST16 - Leila Lopes-Bezerra ST17 - Concha Gil

8:30 - 12:15 pm Symposium 4 From Fungal Biofilms to Intestinal Disorders Chair: Bernhard Hube PL10 – Andrew Koh ST20 - Boualem Sendid ST21 - Francisco Alvarez Botas ST22 - Uwe Groß PL11 – Toni Gabaldon ST23 - Christoph Schüller

EU Project Officer

End of ImResFun

Meeting Check-Out

Lunch & Break 12:00 - 4:00 pm 12:00 - 4:00 pm 12:15 - 4:00 pm

Workshop & Short Talks

5:30 pm Opening 6:00 – 7:00 pm Symposium 1 Clinical Importance of Fungal Diseases Chair: Thomas Lion PL1-Thomas Lehrnbecher

4:00 – 7:30 pm Chair: Karl Kuchler PL4- Tobias Hohl ST6 - Mariana Blagojevic ST7 - Andreas Kühbacher ST8 - Luana Rossato PL5- Frank van de Veerdonk ST9 - Raju Shivarathri ST10 - Jong-Myeong Kim ST11 - Michelle Holtappels

4:00 – 7:30 pm Chair: Attila Gácser PL8- Joachim Morschhäuser ST18 - Fitz Gerald Silao ST19 - Hana Sychrova PL9 - Kaustuv Sanyal 6:30 – 7:30 ImResFun Keynote Plenary Introduced by Karl Kuchler Gordon Brown

4:00 – 7:30 pm Chair: Karl Kuchler PL12 – Clarissa Nobile ST24 - Oliver Bader ST25 - Per Ljungdahl ST26 - Aaron Hernday PL13 – Patrick van Dijck ST27 - Jonathan Richardson ST28 - Iryna Denega

Dinner 7:30 - 8:30 pm 7:30 - 8:30pm

Posters Sessions

7:00 - 8:00 pm ImResFun Opening Plenary Lecture Introduced by Julian Naglik David Denning 8:00 – 11:30 pm Welcome Dinner

8:30 – 11:00 pm Poster Session 1 & PI Round Table Chair: Karl Kuchler & Steffen Rupp

Deciphering Host-Pathogen interaction: Key issues and relevance to clinical practice and drug development.

8:30 -11:00 pm Poster Session 2

7:00 – 7:30 pm Concluding Remarks

7:30 pm – open end Dinner & Farewell

Page 5: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Program 5

Scientific Program of ImResFun2017

1. Sunday September 3

Meeting Registration & Hotel Check-In 10:00 am - 6:00 pm

Opening Ceremony 5:30 pm - 6:00 pm Karl Kuchler & Steffen Rupp

Symposium 1

Clinical Importance of Fungal Diseases

Chair: Thomas Lion 6:00 - 6:20

Thomas Lehrnbecher 6:20 - 7:00 Clinical importance of fungal diseases Keynote Opening Lecture 7:00 pm - 8:00 pm David Denning (Introduced by Julian Naglik) The ’95-95 by 2025’ agenda to reduce morbidity and mortality from fungal diseases Dinner 8:00 - 9:30 pm

Poster Mounting 9:30 pm - 11:30 pm

2. Monday September 4

Breakfast 7:00 - 8:30 am

Symposium 2 8:30 am - 12:00 am

Host Immunology of Fungal Disease

Chair & Introduction: Julian Naglik 8:30 - 8:50

Daniel Kaplan 8:50 - 9:30 Control of cutaneous C. albicans immunity by peripheral nociceptors

Short Talks ST1 – Gloria Molero 9:30 - 9:45 Candida albicans increases the release of proinflammatory extracellular vesicles in THP1 macrophages

ST2 – Michael Riedelberger 9:45 - 10:00 Type I interferons dysregulate the iron and zinc homeostasis in macrophages during C. glabrata infections

Coffee & Refreshment Break 10:00 – 10:30 am

Matthias Gunzer 10:30 - 11:10 Aspergillus fumigatus: antifungal immunity and the quest for better diagnostics

Short Talks

ST3 – Stefanie Allert 11:10- 11:25 Candida albicans induced epithelial damage mediates translocation through intestinal barriers

ST4 - Gabriela Neves 11:25 - 11:40 The human endothelial cell proteome is modulated by a cell wall polysaccharide of an opportunistic fungal pathogen

ST5 – Catarina Oliveira Vaz 11:40 - 11:55 A quantitative proteomic study of human macrophage interaction with Candida albicans using an ATP affinity enrichment approach

Lunch Break 12:00 – 4:00 pm

Page 6: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Program 6

Afternoon Talks 4:00 pm – 7:25 pm

Chair: Karl Kuchler 4:00 - 4:20

Tobias Hohl 4:20 - 5:00 Sterilizing immunity in the lung relies on targeting fungal apoptosis-like programmed cell death Short Talks

ST6 - Mariana Blagojevic 5:00 - 5:15 Epithelial cell death induced by Candidalysin, a cytolytic peptide toxin of Candida albicans

ST7 – Andreas Kühbacher 5:15 – 5:30 Dermal fibroblasts play a central role in skin model protection against C. albicans invasion

ST8 – Luana Rossato 5:30 - 5:45 Investigation of the immune response against the Sporothrix brasiliensis in murine models and the virulence factors using proteomics techniques

Coffee & Refreshment Break 5:45 - 6:00 pm

Frank van de Veerdonk 6:00 - 6:40 TBA Short Talks

ST9 - Raju Shivarathri 6:40 - 6:55 The Gcn5 histone acetyl transferase controls caspofungin tolerance & virulence of the human fungal pathogen Candida albicans

ST10 - Jong-Myeong Kim 6:55 - 7:10 The zinc finger transcription factor Cas5 represses genes for hyphal growth under yeast growth condition in Candida albicans

ST11 - Michelle Holtappels 7:10 - 7:25 Dual localization of Erg11 in Saccharomyces cerevisiae: shedding light on tolerance

Dinner 7:30 - 8:30 pm

Poster Session 1 8:30 pm - 11:00 pm

PI Round Table Discussion 8:30 pm - 11:00 pm Chair: Karl Kuchler & Steffen Rupp Deciphering Host-Pathogen interaction: Key issues and relevance to clinical practice and drug development.

3. Tuesday September 5

Breakfast 7:00 - 8:30 am

Symposium 3 8:30 am - 12:00 am

Fungal Response / Evasion to Host Defense

Chair & Introduction: Steffen Rupp 8:30 - 8:50

Constantin Urban 8:50 - 9:30 Host factor adenosine modulates inflammatory responses in systemic candidiasis Short Talks ST12 - Samir Jawhara 9:30 - 9:45 Soluble short fractions of β-1,3 glucans derived from Candida albicans act as a shield for pathogenic yeasts and modulate the activation of platelets

ST13 - Filomena Nogueira 9:45 - 10:00 Klebsiella prevents sporulation and hyphal development of Aspergillus species

Page 7: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Program 7

ST14 - Sabrina Jenull 10:00 - 10:15 The Candida albicans HIR histone chaperone regulates the yeast-to-hyphae transition by controlling the sensitivity to morphogenesis signals

Coffee & Refreshment Break 10:15 – 10:35 am

James Konopka 10:35 - 11:15 Plasma membrane architecture: form and function in fungal pathogenesis Short Talks

ST15 - Sarah Sze Wong 11:15 - 11:30 Complement evasion of Aspergillus fumigatus by secretion of metalloprotease Mep1p

ST16 - Leila Lopes-Bezerra 11:30 - 11:45 Structural differences on cell walls of the dimorphic fungal pathogens Sporothrix schenckii and Sporothrix brasiliensis impact human macrophage recognition.

ST17 - Concha Gil 11:45 - 12:00 Host-Pathogen Interaction: Insights from Proteomics

Lunch & Afternoon Break 12:00 – 4:00 pm

Afternoon Talks 4:00 pm – 7:30 pm Chair & Introduction: Attila Gácser 4:00 - 4:20

Joachim Morschhäuser 4:20 - 5:00 Evolving antifungal drug resistance - a strategy of Candida albicans to overcome the last host defense Short Talks

ST18 - Fitz Gerald Silao 5:00 - 5:15 Protein quality control at the inner nuclear membrane sets a threshold that determines the basal expression of virulence factors in Candida albicans

ST19 - Hana Sychrova 5:15 - 5:30 Potassium uptake and homeostasis in Candida species

Coffee & Refreshment Break 5:30 - 5:50 pm

Kaustuv Sanyal 5:50 - 6:30 Genome indexing by histone H3 variants in Candida albicans

ImResFun2017 Keynote Plenary Lecture

Gordon Brown (Introduced by Karl Kuchler) 6:30 - 7:30 MelLec: A new player in antifungal immunity

Dinner 7:30 - 8:30 pm

Poster Session 2 8:30 pm - 11:00 pm

4. Wednesday September 6

Breakfast 7:00 - 8:30 am

Symposium 4 8:30 am - 12:15 am

From Fungal Biofilms to Intestinal Disorders

Chair & Introduction: Bernhard Hube 8:30 - 8:50

Andrew Koh 8:50 - 9:30 Candida albicans gastrointestinal colonization and dissemination in the mammalian host

Page 8: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Program 8

Short Talks

ST20 - Boualem Sendid 9:30 - 9:45 A serum disaccharide signature of invasive fungal infections detected by MALDI-MS.

ST21 - Francisco Alvarez Botas 9:45 - 10:00 New perspectives for fungal research in Ecuador

ST22 - Uwe Groß 10:00- 10:15 Candida in the elderly

Coffee & Refreshment Break 10:15 – 10:35 am

Toni Gabaldon 10:35 - 11:15 Recent evolution of C. glabrata

Short Talks

ST23 - Christoph Schüller 11:15 - 11:30 The broad and malleable phenotypic space of clinical Candida glabrata strains. A high throughput mapping of phenotypes reveals highly individualistic adaptations to a fluctuating environment. EU Project Officer Charalampos Moschopoulos 11:30 - 12:15 Marie Skłodowska-Curie actions - Research Fellowship Programme

Lunch & Afternoon Break 12:15 – 4:00 pm

Afternoon Talks 4:00 pm – 7:30 pm

Chair & Introduction: Karl Kuchler 4:00 – 4:20

Clarissa Nobile 4:20 - 5:00 Discovery of a Novel Bacterial Endosymbiont Living within the Human Fungal Pathogen Candida albicans Short Talks

ST24 - Oliver Bader 5:00 - 5:15 Adhesin variability in clinical isolates of Candida glabrata and Candida parapsilosis

ST25 - Per Ljungdahl 5:15 - 5:30 SPS-sensor and metabolic control of amino acid-induced morphogenesis in Candida albicans

ST26 - Aaron Hernday 5:30 - 5:45 Markerless genome editing in Candida albicans

Coffee & Refreshment Break 5:45 - 6:00 pm

Patrick van Dijck 6:00 - 6:40 Investigation of the underlying mechanisms of antimicrobial drug tolerance of single and dual-species Candida albicans and Staphylococcus aureus biofilms using in vitro and in vivo model systems ST27 - Jonathan Richardson 6:40 - 6:55 The Candidalysins are a family of fungal toxins

ST28 - Iryna Denega 6:55 - 7:10 Molecular mechanisms governing the formation of persister cells in Candida albicans biofilms

Concluding Remarks 7:10 – 7:30

Dinner 7:30 - 9:30 pm

5. Thursday September 7

Breakfast 7:00 - 9:30 am

End of ImResFun2017

Check-out

Page 9: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Keynote Lecture 9

ImResFun2017 Keynote Opening Lecture

KN- 01 The ’95-95 by 2025’ agenda to reduce morbidity and mortality from fungal diseases

David W. Denning

The University of Manchester, Southmoor Road, Manchester M23 9LT, United Kingdom of Great Britain and Northern Ireland

Fungal diseases are neglected by social and political communities. However, over a billion people are affected resulting in approximately 11.5 million life-threatening infections and over 1.5 million deaths every year. There have been enormous advances in fungal diagnostics and antifungal development over the last 20 years but most of the world’s population have not seen these benefits yet. In places with developed health systems, fungal diseases are diagnosed and treated, although many are still missed as judged by autopsy. However, in low and middle income countries (LMICs) the absence of diagnostic tools and antifungals plus a lack of training of health staff ensures that the mortality and morbidity of fungal infections remains unacceptably high. Recently, Kneale et al. (JAC 2016), have highlighted many country differences in antifungal availability and price. Thus, amphotericin B is not available to a population of 481 million and where it exists the daily price varied from <$1 to $171. Flucytosine is unavailable to >2,898 million in more than 70% of countries explored. For azole drugs the situation is better. Thus, fluconazole was licensed in 88.6% of the countries surveyed with a daily price (750-800 mg) variation of <$1 to $31. Itraconazole was unavailable to >78 million with a daily price varying from <$1 to $102. Fluconazole has been on the WHO’s Essential Medicine List for many years, amphotericin B and flucytosine were listed in 2013, and this year itraconazole and voriconazole were added.

GAFFI has called for 95% of the world’s population to have access to fungal disease diagnostics and treatments by 2025 (95-95 by 2025). One aspect of this call is provision of reference laboratories in each country providing the critical mass of expertise and a complete portfolio of diagnostic tests. By scaling the model in Guatemala (where GAFFI has a program of diagnostics for fungal diseases in AIDS), and related models across the globe, patients with fungal infections will be properly diagnosed and treated. Involvement of governments’ health care systems, academia, industry and the public are essential to stimulate the economic, political and scientific measures required to develop a network of health professionals trained in fungal infection management as well as equipped laboratories with rapid diagnostic techniques and availability of antifungals to treat at affordable prices in LMICs.

Page 10: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Clinical Importance of Fungal Diseases 10

Symposium 1

Clinical Importance of Fungal Diseases

Chair & Introduction

Thomas Lion

CCRI- Children's Cancer Research Institute, Zimmermannplatz 10, 1090 Vienna, Austria

Page 11: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Clinical Importance of Fungal Diseases 11

PL-01 Clinical importance of fungal diseases

Thomas Lehrnbecher

University of Frankfurt, Theodor Stern Kai 7, Frankfurt 60590, Germany

Whereas in many countries fungal skin disease and recurrent vulvo-vaginal candidosis are the most common fungal infections and affect a significant percentage of the population, severity of fungal infection and infection-related mortality are highest in immunocompromised patients. Similar to adults, children undergoing allogeneic hematopoietic stem cell transplantation or children suffering from acute myeloid leukemia or from relapsed leukemia are at highest risk for invasive fungal disease, which is mostly caused by Aspergillus spp, less often by Candida spp or mucormycetes. As symptoms are often non-specific and biomarkers such as galactomannan have limited usefulness, early detection of invasive fungal disease is difficult, in particular in pediatric patients. In neutropenic patients with fever not responding to broad-spectrum antibiotics, abnormalities in imaging studies of the lung should lead to invasive diagnostic tests such as broncho-alveolar lavage or biopsy, but both of them may be associated with severe and even life-threatening complications. On the other hand, antifungal prophylaxis which is recommended in high-risk populations, may also be associated with adverse effects such as allergic reactions, renal and hepatic problems, may be problematic due to a number of drug-drug interactions, and is a significant cost factor. However, prevention and early recognition of invasive fungal infection is important because patients suffering from invasive fungal infection aspergillosis stay significant longer in the hospital stay and have a higher treatment-related mortality. To this end, better diagnostic tools to detect invasive fungal infection are clearly warranted, and it is hoped that better strategies of antifungal prophylaxis and treatment will result in better outcome.

Page 12: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Host Immunology of Fungal Disease

12

Symposium 2

Host Immunology of Fungal Disease

Chair & Introduction

Julian Naglik

King's College London, St Thomas Street, SE1 1UL London, UK

Page 13: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Host Immunology of Fungal Disease

13

PL-02 Control of cutaneous C. albicans immunity by peripheral nociceptors

Daniel H Kaplan

Dermatology, University of Pittsburgh, 200 Lothrop St, Pittsburgh 15260, United States of America

The skin is a barrier organ that is a frequent site of entry for numerous pathogens. The skin is able to resist infection through a variety of mechanisms including innate and adaptive immune responses. Innate resistance to Candida albicans in the skin requires the production of interleukin-17A (IL- 17A) by tissue-resident cells early during infection. We found in mice that dermal γδ T cells were the dominant source of IL- 17A during C. albicans infection and were required for pathogen resistance. Induction of IL-17A from dermal γδ T cells and resistance to C. albicans required IL-23 production from CD301b+ dermal dendritic cells (dDCs). In addition, we found that RTX-medaited ablation of sensory neurons increased susceptibility to C. albicans infection, which could be rescued by exogenous addition of the neuropeptide CGRP. Unexpectedly, dorsal root ganglion (DRG) sensory neurons fluxed Ca2+ and secreted CGRP when exposed to C. albicans indicating that DRG can directly recognize C. albicans. Cell wall components of yeast and filamentous forms were unable to induced either Ca2+ flux of CGRP release from dorsal root. Rather, ATP secreted from heat killed and live C. albicans was responsible for Ca2+ flux in DRG. These data define a model in which nociceptive pathways in the skin can be directly activated by C. albicans to drive production of IL-23 by CD301b+ dDCs resulting in IL-17A production from gd T cells and resistance to cutaneous candidiasis.

SHORT TALKS

S1-ST01 Candida albicans increases the release of proinflammatory extracellular vesicles in THP1 macrophages

Molero Gloria, Reales-Calderon Jose Antonio, Vaz Catarina, Monteoliva Lucía, Gil Concha

Microbiology Ii, Faculty of Pharmacy, University Complutense of Madrid, Plaza de Ramón y Cajal s/n, Madrid 28040, Spain

The effectiveness of macrophages in the response to systemic candidiasis is crucial to a fungal effective clearance. Communication between immune cells is mediated, among others, by the secretion of proteins, mRNAs, non-coding RNAs and lipids through extracellular vesicles (EVs). EVs change their cargo to mediate different responses and may play a role in the response against infections. Thus, we have boarded a quantitative proteomic analysis on the protein composition of the THP1 macrophages-derived EVs in response to Candida albicans infection that unraveled an increase in the number of EVs secreted in response to C. albicans vs control. Differential proteomic analyses of EVs showed changes in 133 due to the interaction with C. albicans of the 717 proteins identified and quantified. The differentially abundant proteins were involved in functions as immune response, signaling, or cytoskeletal reorganization. Macrophage-derived EVs, both from control and from Candida-infected macrophages, had effector functions on other THP1 monocytes and differentiated macrophages, activating ERK and p38 kinases, increasing the secretion of proinflammatory cytokines and the candidacidal activity. Our findings provide new information on the role of macrophage-derived EVs in candidiasis and on macrophages communication in response to infections.

S1-ST02 Type I interferons dysregulate the iron and zinc homeostasis in macrophages during C. glabrata infections

Michael Riedelberger, Bernhard Scheidl, Karl Kuchler

Department for Medical Biochemistry, Medical University of Vienna, Dr. Bohrgasse 9, Vienna 1030, Austria

During infections with various pathogens, the cytokine family of type I interferons (IFNs) engage a predominant role in the complex regulation of the antimicrobial immune response and the molecular cross-talk between immune cell populations. However, the contribution and molecular function of type I IFNs remains poorly understood or even controversial for infections with bacteria or fungi.

Here, we show that type I IFNs promote the survival of the human fungal pathogen Candida glabrata during host-pathogen interactions by increasing the intracellular replication of C. glabrata in murine macrophages. Interestingly, type I IFNs tremendously dysregulate the iron and zinc homeostasis in macrophages, whereby these trace metals can be exploited by C. glabrata for metabolism and virulence. In detail, type I IFNs suppress the gene expression of key players involved in iron and zinc metabolism during fungal infections and completely abrogate the iron shuttling from the phagolysosome to the cytoplasm. As a consequence, these altered metal pools could represent a more favourable environmental niche for C. glabrata. Therefore, our results suggest a detrimental role of type I IFNs for the host during C. glabrata infections by dysregulating the cellular homeostasis of iron and zinc in macrophages.

Page 14: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Host Immunology of Fungal Disease

14

PL-03 Aspergillus fumigatus: antifungal immunity and the quest for better diagnostics

Matthias Gunzer

Institute for experimental Immunology and Imaging, University Hospital Essen, Hufelandstr. 55, Essen 45147, Germany

Invasive fungal aspergillosis is a life-threatening lung infection, that can affect immune suppressed individuals, while a normally functioning immune defense very effectively controls the pathogenic growth. I will present our findings on the immune response of neutrophil granulocytes and, more recently, alveolar epithelial cells towards this environmental infection. This is based on extensive microscopic observation in a variety of animal models and the application of comprehensive molecular profiling. In addition we have exploited our findings to develop novel diagnostic regimens that allow the unequivocal identification of this infection in vivo using molecular imaging. An earlier and more precise diagnosis is still the ultimate goal for the clinical control of this severe fungal disease and we hope to be able to contribute to this goal with our work.

SHORT TALKS

S1-ST03 Candida albicans induced epithelial damage mediates translocation through intestinal barriers

Stefanie Allert1, Toni Förster1, Marc Juraschitz1, Daniela Schulz1, Betty Hebecker1, Tony Pawlik1, Selene Mogavero1, Julian Naglik2, Lydia Kasper1, Bernhard Hube1 1 Hans-Knoell-Institute, Adolf-Reichwein-Straße 23, Jena 07745, Germany 2 King’s College London, UK

The opportunistic pathogen Candida albicans can cause life-threatening systemic infections, which predominantly originate from the commensal C. albicans population of the intestinal tract. However, C. albicans translocation from the gut into the bloodstream is not well characterized. The aim of this project is to elucidate the molecular mechanisms associated with damage of and translocation through intestinal epithelium and to define potential pathogenicity factors required for intestinal tissue invasion.

Characterization of processes associated with epithelial damage and fungal translocation using an in vitro translocation model showed that C. albicans translocation mainly occurs via a transcellular route, which is associated with fungal-induced epithelial damage. Hence, translocation correlates with necrosis, but not apoptosis of intestinal cells. Screening of C. albicans libraries identified deletion mutants with reduced or increased damage potential, as well as a number of unknown function genes associated with damage of intestinal cells. Selected damage-defective mutants were analyzed for infection-associated attributes, invasive potential and impact on epithelial barrier function in the translocation model. Almost every mutant of this subgroup showed a unique pattern of defects indicating that several biological properties are required for full damage potential.

Among the identified mutants, was a mutant lacking the gene ECE1. The polypeptide Ece1 and the Ece1-derived lesion-forming peptide toxin Candidalysin are dispensable for the ability to produce hyphae, to adhere to or invade into intestinal cells, but are essential for causing intestinal epithelial damage. Our data also show that translocation of C. albicans through intestinal layers is associated with Ece1-mediated cell lysis. However, ece1Δ mutant cells are still able to reduce epithelial integrity and show a reduced translocation rate. This indicates that fungal translocation can occur without damage, e.g. by a tight junction-associated paracellular mechanism, and suggests the existence of fungal factors modulating epithelial integrity independent of damage.

Page 15: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Host Immunology of Fungal Disease

15

S1-ST04 The human endothelial cell proteome is modulated by a cell wall polysaccharide of an opportunistic fungal pathogen

Gabriela Neves1, Nathália Curty1, Thierry Fontaine2, Jean Paul-Latgé2, Leila Lopes-Bezerra1 1 Laboratory of Cellular Mycology and Proteomics, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil 2 Aspergillus Unit, Institut Pasteur, Paris, France

Invasive aspergillosis is an opportunistic fungal infection with high mortality rates for hematologic patients. The main etiologic agent of this angioinvasive fungal infection is Aspergillus fumigatus. Upon cell-cell contact with human umbilical vein endothelial cells (HUVEC) this fungus induces cellular injury, inflammatory response and a pro-thrombotic endothelial phenotype. The HUVEC model we had developed has the goal to discover which cell surface glycoconjugates of A. fumigatus may induce endothelial activation. Moreover, we will discuss the putative endothelial pathways that can be regulated upon HUVEC interaction with A. fumigatus mutant strains and a purified cell wall polysaccharide, the galactosaminogalactan (GG). A label-free proteomic approach was used to compare protein expression levels of HUVECs challenged with an A. fumigatus mutant strain (Δugm1) that overexpress a cell wall GG and a purified galactosaminogalactan soluble fraction (SGG). The ∆ugm1 mutant of A. fumigatus modulated specific endothelial pathways related to the inflammatory and immune response mediated by TNF-α. The purified SGG was able to induce TNF-α secretion and also regulate pathways identified in the interaction assay of ∆ugm1 mutant with HUVECs. This is the first work describing the impact of an A. fumigatus cell surface glycoconjugate on the endothelial response.

Acknowledgements: This work was supported by Fundação Carlos Chagas de Amparo a Pesquisa do Estado do Rio de Janeiro (FAPERJ), Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) and Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES), Brazil.

S1-ST05 A quantitative proteomic study of human macrophage interaction with Candida albicans using an ATP affinity enrichment approach

Catarina Oliveira Vaz1, Aida Pitarch1, Jose Antonio Reales-Calderon1, Maria Luisa Hernaez2, Lucia Monteoliva1, Concha Gil1 1 Microbiologia II, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramon y Cajal s/n, Madrid 28040, Spain 2 Unidad Proteomica, Universidad Complutense de Madrid

Macrophages are involved in the primary human host response to human pathogens such as Candida albicans. After pathogen recognition by specific receptors a cascade of signaling pathways are activated, leading to the production of costimulatory molecules and proinflammatory cytokines, chemokines and antimicrobial peptides. ATP-binding proteins, such as kinases, are crucial for the regulation of signaling pathways involved in the response to C. albicans. A quantitative proteomic approach was carried out for the study of human macrophage ATP-binding proteins after interaction with C. albicans cells. For this, the human monocytic cell-line THP1 was used and cells were labelled by SILAC method with switched labelling. Monocytes were differentiated into macrophages by adding PMA. After incubation of the macrophages with C. albicans cells (MOI of 1) for 3 hours, protein lysates were enriched in ATP-binding proteins using the ActivX desthiobiotin ATP probes kit. Then, proteins were reduced, alkylated and in-gel digested using trypsin and samples analyzed by mass spectrometry. Overall, 547 non-redundant proteins including 137 ATP-binding proteins were quantified. From them, 22 and 37 proteins were more and less abundant, respectively, during macrophage interaction with C. albicans. A gene ontology term enrichment analysis showed that the more abundant proteins during interaction were involved in RNA splicing and protein synthesis whereas the less abundant proteins were related to proteolysis events, apoptotic processes and ion transmembrane transport. Western blotting and selected reaction monitoring were used to validate the abundance of three proteins (MAP2K2, PRDX5 and NDKA). Bioinformatic tools such as String and Ingenuity Pathway Analysis were used for the analysis. Forty five protein-protein associations were found according to known, predicted and other interactions. Moreover, several upstream regulators such as cytokines, micro-RNAs; transcription regulators and transmembrane receptors were predicted to be differentially expressed. This enrichment is useful to identify and quantify new proteins involved in macrophage-fungus interaction. These proteomic approaches could be valuable for the discovery of mechanisms involved in the innate immune response to C. albicans.

This work is supported by FP7-PEOPLE-2013-ITN, IMRESFUN Project.

Page 16: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Host Immunology of Fungal Disease

16

Afternoon Session 2

Chair & Introduction

Karl Kuchler

Medical University Vienna, Dr. Bohr-Gasse 9/2, 1030 Vienna, Austria

Page 17: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Host Immunology of Fungal Disease

17

PL-04 Sterilizing immunity in the lung relies on targeting fungal apoptosis-like programmed cell death

Tobias Hohl

Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue , New York 10065, United States of America

Humans inhale mold conidia daily and typically experience lifelong asymptomatic clearance. Conidial germination into tissue-invasive hyphae can occur in individuals with defects in myeloid function, though the mechanism of myeloid cell-mediated immune surveillance remains unclear. By monitoring fungal physiology in vivo, we demonstrate that lung neutrophils trigger programmed cell death with apoptosis-like features in Aspergillus fumigatus conidia, the most prevalent human mold pathogen. An anti-apoptotic protein, AfBIR1, opposes this process by inhibiting fungal caspase activation and DNA fragmentation in the murine lung. Genetic and pharmacologic studies indicate that AfBIR1 expression and activity underlies conidial susceptibility to NADPH oxidase-dependent killing and, in turn, host susceptibility to invasive aspergillosis. Immune surveillance exploits a fungal apoptosis-like programmed cell death pathway to maintain sterilizing immunity in the lung.

SHORT TALKS

S1-ST06 Epithelial cell death induced by Candidalysin, a cytolytic peptide toxin of Candida albicans

Mariana Blagojevic, Jonathan P Richardson, Julian R Naglik

King's College London, Newcomen Street, London SE1 1UL, United Kingdom of Great Britain and Northern Ireland

C. albicans is an important fungal pathogen of humans causing mucosal and systemic infections in patients with a compromised immune system. C. albicans is capable of morphological switching, forming hyphae that damage the host and induce cell death. Research in our group has demonstrated that the C. albicans hypha specific peptide (termed Candidalysin) acts as a cytolytic toxin, inserting into epithelial cell membranes to induce damage and cell death. Cell death via necrosis and apoptosis are well known processes during microbial infections, but in recent years additional mechanisms of regulated cell death have also been identified, including necroptosis and pyroptosis. Whilst pyroptosis is activated in macrophages following successful phagocytosis of C. albicans, it is unclear which cell death pathway(s) C. albicans and Candidalysin activate in epithelial cells during mucosal infections. Thus, we investigated the ability of Candidalysin to induce apoptosis, necrosis, necroptosis and/or pyroptosis in epithelial cells. Preliminary data suggests that apoptosis is not triggered in oral epithelial cells following Candidalysin treatment, and therefore epithelial cell death likely proceeds through a necrotic mechanism.

Page 18: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Host Immunology of Fungal Disease

18

S1-ST07 Dermal fibroblasts play a central role in skin model protection against C. albicans invasion

Andreas Kühbacher, Helena Henkel, Philip Stevens, Christian Grumaz, Doris Finkelmeier, Anke Burger-Kentischer, Kai Sohn, Steffen Rupp

Fraunhofer IGB, Nobelstraße 12, Stuttgart 70569, Germany

Research efforts in the last decades have shed light on mechanistic interactions of fungal pathogens with host organisms down to the molecular level. Although considerable progress has been made in the last decades, the function of the individual components of epithelial barriers with regard to host-microbial interaction is not fully understood.

The fungal pathogen Candida albicans colonizes basically all human epithelial surfaces including the skin. Under certain conditions, such as immunosuppression, invasion of the normally protected epithelia occurs. In natural skin these infections are only superficial but not much is known about defense mechanisms against C. albicans in subepithelial layers such as the dermis.

Using immune cell-supplemented 3D skin models we could define a new role for fibroblasts in the dermis and identify a minimal set of cell types for skin protection against C. albicans invasion. Dual RNA-Seq of individual host cell populations and C. albicans revealed that dermal invasion is directly impeded by dermal fibroblasts. They are able to integrate signals from the pathogen and CD4+ T cells and shift towards an antimicrobial phenotype with broad specificity that is dependent on TLR2 and IL-1β. Skin model protection could be induced by addition of IL-1β even in the absence of T cells. TLR2 activation in dermal fibroblasts which occurs through C. albicans, results in induction of IL-1β expression, but it is secreted only in the presence of CD4+ T cells. These results highlight a central function of dermal fibroblasts for skin protection opening new possibilities for treatment of infectious diseases.

S1-ST08 Investigation of the immune response against the Sporothrix brasiliensis in murine models and the virulence factors using proteomics techniques.

Luana Rossato1, Suelen Silvana Santos1, Joanna Freeke2, Sybren De Hoog2, Sandro Rogério Almeida1 1 University of São Paulo, Lineu Prestes, 580, São Paulo 05508-000, Brazil 2 Westerdijk Fungal Biodiversity Institute, Utrecht, Netherlands

Sporotrichosis is a subacute or chronic infection caused by fungals thermo-dimorphic Sporothrix complex. Recent molecular studies show that the pathogenic clade of the genus Sporothrix comprises four distinct species, while some more 4 remote species also occur as occasional opportunists. The leading cause of epidemic outbreaks in South America, with proven virulence but a lack of evaluation of immune response and virulence factors is Sporothrix brasiliensis. The aim of this study was to elucidate the role of Toll-4 and Toll-2 in the immune response against S. brasiliensis and the potential proteins related to virulence using proteomics techniques. Animals (Wild-type, Toll-2-/-, Toll-4-/-) were maintained for 7, 14 and 28 days before being euthanized. Liver, spleen, and kidney were collected for determination of fungal burden and cytokine measurements for CBA (IL-10, IFN-γ, IL-4, TNF-α, IL-6, IL-2). For elicited the proteins expressed we used the Bottom-Up proteomics technique comparing the species S. schenckii and S. brasiliensis. Results: In vivo tests showed that after 14 days of infection, the liver, spleen, and kidney from Toll-2-/- and Toll-4-/- presented higher fungal burden when compared with wild-type animals. In respect to cytokines, we observed an inflammatory response. Using the proteomics techniques, we observed 60 proteins exclusively expressed in S.brasiliensis.Conclusions: It is suggested that absence of Toll-2 and Toll-4 interferes with the course of the infection induced by the fungus S. brasiliensis and the proteins specifically expressed in S.brasiliensis should be identified in relation the virulence.

Page 19: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Host Immunology of Fungal Disease

19

PL-05 TBA Frank van de Veerdonk

SHORT TALKS

S2-ST09 The Gcn5 histone acetyl transferase controls caspofungin tolerance & virulence of the human fungal pathogen Candida albicans

Raju Shivarathri1, Michael Tscherner1, Florian Zwolanek1, Nitesh Kumar Singh2, Neeraj Chauhan3, Karl Kuchler1 1 Medical Biochemistry, Medical university of Vienna, Mfpl, Dr. Bohrgasse-9/2, Vienna 1030, Austria 2 CLC bio, a QIAGEN Company, Silkeborgvej 2, 8000 Aarhus C, Denmark 3 Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, USA

The increased incidence of invasive fungal infections has changed the landscape of fungal infections worldwide and greatly impacts healthcare management. Candida albicans is the most prevalent opportunistic human fungal pathogen and the leading cause of Candida blood-stream infections. In addition to known pathogenicity mechanisms, epigenetic reprogramming has recently emerged as a prime mechanism critically determining the outcome of host-pathogen interactions. Thus, chromatin alterations may be one of the key adaptive survival strategies employed by fungal pathogens to escape from host immune surveillance. Acetylation and deacetylation of chromatin constituents by histone acetyltransferases (HATs) and histone deacetylases (HDACs) are the two dynamic post-translational modifications modulating gene expression and pathogenicity. In fact, targeting HATs and HDACs could constitute a novel therapeutic approach to treat fungal infections. However, the molecular mechanisms of HATs and HDACs in fungal pathogenesis remain little understood. Here, we present novel data on the first phenotypic and molecular characterization of GCN5, a prototypic HAT in C. albicans. Lack of GCN5 leads to pronounced antifungal drug resistance, cell wall stress hyper susceptibility, as well as cell morphology defects, all in all affecting both filamentation and virulence. Remarkably, the absence of Gcn5 dramatically debilitates acetylation of H3 and H4 at certain lysine residues, implying that the corresponding target genes are essential in controlling fungal virulence in response to host signals. Surprisingly, gcn5 mutants also display constitutive hyper-activation of the Cek1 and Hog1 kinases, both operating in fungal Mitogen-Activated Protein (MAP) kinase signaling pathways essential for virulence. Hence, we have delineated the transcriptional response of gcn5∆/∆ cells to caspofungin stress in detail. Moreover, to identify targets for Gcn5-mediated regulation, we pursued a global proteomics approach, allowing for the identification of novel Gcn5 interaction partners, as well as the associated acetylome. Taken together, our data suggest that the Gcn5 HAT may act as a molecular switch controlling the cross-talk of different signaling pathways to control fungal virulence.

Page 20: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Host Immunology of Fungal Disease

20

S2-ST10 The zinc finger transcription factor Cas5 represses genes for hyphal growth under yeast growth condition in Candida albicans

Jong-Myeong Kim1, Hye Yun Moon2, Hyun Ah Kang2, Jeong-Yoon Kim1 1 Department of Microbiology and Molecular Biology, College of Bioscience and Biotechnology, Chungnam National University, Daejeon, Republic of Korea 2 Department of Life Science, College of Natural Science, Chung-Ang University, Seoul, Republic of Korea

Candida albicans, an opportunistic pathogen of humans, exists as yeast, hyphal, or pseudohyphal form, depending on pH, nutrient and temperature. The morphological transition from yeast to hypha, which is important to virulence, is controlled by many transcription factors that activate or repress hypha-specific genes (HSGs), including HGC1, ALS3, ECE1 and HWP1. The putative zinc finger transcription factor Cas5 is known to activate expression of many cell wall integrity genes, but it remains unknown whether Cas5 affects hyphal growth in C. albicans. Interestingly, we found that cas5Δ/Δ mutant cells could not maintain yeast form under non-hyphal inducing condition. Analysis of Cas5 expression revealed that CAS5 transcription is significantly downregulated upon hyphal initiation in wild type, which suggests that Cas5 is a kind of transcription factor repressing genes required for hyphal growth. Consistently with the traits, the cas5Δ/Δ mutant highly expressed hypha-specific ALS3, ECE1 and HWP1 genes under non-hyphal inducing condition. In addition, the cas5Δ/Δ mutant showed decreased transcription of several genes involved in ergosterol biosynthesis pathway. Collectively, this study suggests that Cas5 represses transcription of genes responsible for hyphae formation during yeast-form growth in C. albicans.

S2-ST11 Dual localization of Erg11 in Saccharomyces cerevisiae: shedding light on tolerance

Michelle Holtappels1, Winnie Kerstens1, Alessandro Fiori2, Marilyn De Graeve3, Anneke Kremer4, Saskia Lippens4, Inge Van Bogaert3, Patrick Van Dijck1 1 VIB, KU Leuven, Kasteelpark Arenberg 31, Leuven 3001, Belgium 2 Agrosavfe NV, Ghent 9000, Belgium 3 Coupure Links 653, Ghent 9000, Belgium 4 FVMS Building - Technologiepark 927, Zwijnaarde 9052, Belgium

The azoles form a class of antifungals that are the most widely used to fight human, animal and plant fungal pathogens. They target the production of ergosterol, the main sterol of all fungi, by inhibiting the cytochrome P450 protein Cyp51, encoded by the ERG11 gene. In clinical practice, fluconazole is the most abundantly used azole. However, the fungistatic character favoring the development of drug resistance is one of the major disadvantages when using azoles. Resistance to the azoles has been characterized and is a well investigated topic, the tolerance mechanisms, on the other hand, remain largely unknown.

We discovered that a mitochondrial Erg11 isoform (mtErg11), an N-terminal truncation of the whole protein, is induced upon fluconazole treatment, which proved to be responsible for the tolerance observed. Using several bioinformatics tools, we identified an encrypted mitochondrial targeting sequence (MTS) of Erg11 starting approximately 150 amino acids downstream from the N-terminus. We demonstrated mitochondrial localization of this fragment using confocal microscopy and FIB-SEM imaging. We also included genetic evidence by the fusion of the MTS of Erg11 to Arg8, a protein with known mitochondrial localization and function. Further, we are using several lipid analyses to investigate functionality of this mtErg11 fragment by lipidome analysis and LC-MS. Screening of all non-essential genes annotated as proteases showed that mutants carrying deletions in PRB1 or in PEP4, encoding vacuolar proteases A and B respectively, did not display mtErg11, despite the fact that they still contained normal amounts of the full-length Erg11. Finally, we are also investigating the effect of the presence and absence of mtErg11 on the stability of mtDNA, on mitochondrial functionality and morphology.

A similar mechanism has also been observed in Candida albicans and we are currently identifying the protease involved as this may be an interesting antifungal target.

Page 21: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Fungal Response / Evasion to Host Defense

21

Symposium 3

Fungal Response / Evasion to Host Defense

Chair & Introduction

Steffen Rupp

Fraunhofer-Institute IGB, Nobelstrasse 12, 70569 Stuttgart, Germany

Page 22: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Fungal Response / Evasion to Host Defense

22

PL-06 Host factor adenosine modulates inflammatory responses in systemic candidiasis

Ava Hosseinzadeh1, Munender Vodnala2, Madhu Shankar0, Sujan Yellagunda0, Mattias Carlström3, Anders Hofer4, Constantin Felix Urban0 1 Clinical Microbiology, Umeå University, Umeå, Sweden 2 Medical Biochemistry and Biophysics; Umeå University, Umeå, Sweden 3 Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden 4 Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden 0 Clinical Microbiology, Umeå University, Sjukhusområdet 6C, Umeå, 90185, Sweden

Invasive fungal infections may be accompanied by overwhelming inflammatory responses in the host, often at the time of immune recovery, resulting in immune reconstitution inflammatory syndrome (IRIS). In search for host factors which reduce inflammatory responses in systemic mycoses we investigated adenosine. The nucleoside is produced in many tissues, such as for instance by endothelium. Adenosine is considered as anti-inflammatory molecule conveying function via specific receptors, classified in four different subtypes A1, A2A, A2B and A3. The role of adenosine in regulating inflammatory processes during microbial infection is poorly understood. We showed that adenosine levels increase more than ten-fold in plasma of mice systemically infected with Candida albicans. Neutrophils are recruited to sites of C. albicans infection in large numbers and contribute to an inflammatory milieu by production of ROS, secretion of pro-inflammatory cytokines and release of neutrophil extracellular traps (NETs). Interestingly, we found that adenosine-treated neutrophils released less amounts of NETs upon Candida stimulation. Adenosine-triggered regulation was mediated via adenosine receptor A3 expressed on neutrophils. Pharmacological activation of A3 in a murine model of systemic candidiasis reduced inflammation and markers for organ damage in plasma. We are currently investigating A3 knockout mice for confirmation of pharmacological data. In summary, our findings identify adenosine as important modulator of inflammation during systemic candidiasis and suggest that pharmacological targeting of A3 receptor could be a promising adjunct therapy for IRIS during episodes of invasive mycoses.

SHORT TALKS

S2-ST12 Soluble short fractions of β-1,3 glucans derived from Candida albicans act as a shield for pathogenic yeasts and modulate the activation of platelets Hélène Vancraeyneste1, Yann Guerardel2, Laura Choteau0, Anne Bauters3, Meryem Tardivel4, Nadine François0, Laurent Dubuquoy0, Dmitry Soloviev5, Daniel Poulain0, Boualem Sendid0, Samir Jawhara0 1 Inserm U995/Team2 2 Unité de Glycobiologie Structurale et Fonctionnelle, UGSF 3 Laboratoire d’Hémostase, Pôle de Pathologie Génétique 4 Plateforme d’Interaction Moléculaire, IMPRT-IFR114 5 Joseph J. Jacobs Center for Thrombosis and Vascular Biology, Cleveland Clinic 0 Inserm U995-LIRIC/Team2, Faculté de médecine Pole Formation, 1 place Verdun, Lille, 59000, France

Platelets play a crucial role in hemostasis, thrombosis, and pathogen clearance. Many pathogenic fungi can interact with platelets in circulating blood. The pathogenic fungus Candida albicans is the predominant cause of invasive forms of candidiasis. Its cell wall contains β-1,3 glucans that are known to trigger a wide range of host cell activities. Clinically, β-glucans are released in the circulation during infection and their detection allows the early diagnosis of an invasive fungal infection, but the role of β-glucans in the modulation of platelet activities and platelet-neutrophil interactions is unknown. Platelets pretreated with β-glucan fractions were analyzed in terms of activation, receptor expression, aggregation and adhesion to neutrophils and to C. albicans. Our study shows that β-glucan fractions decrease platelet aggregation, and modulate the coagulation process. The biological activities of these oligoglucosides depend on their degree of polymerization and their concentration. They also affect the regulation of platelet receptors (P-selectin and αIIbβ3). Interestingly, these oligoglucosides at a low concentration reduce platelet activation, and both platelet-C. albicans and platelet-neutrophil interactions, which allow C. albicans to be protected from leukocyte activation. Mechanistically, these oligoglucosides block protein kinase-C activation in platelets that affect the regulation of platelet receptors. Bedsides, we showed that the pentaglucosides reduced platelet activities through TLR4 mediated TGF-β1 production and ATP release, and blocking this receptor by an anti-TLR4 antibody abolished the effect of the pentaglucosides on platelets suggesting that TLR4 is involved in the immuno-modulatory effects induced by β-glucans. Our study offers new insights, showing that these fungal-derived oligoglucosides are not involved exclusively in the protection of C. albicans during infection but also in the modulation of platelet activation mediated via TLR4 stimulation.

Page 23: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Fungal Response / Evasion to Host Defense

23

S2-ST13 Klebsiella prevents sporulation and hyphal development of Aspergillus species

Filomena Nogueira1, Leonel Pereira1, Sabrina Jenull2, Karl Kuchler2, Thomas Lion1 1 CCRI, Zimmermannplatz, Vienna 1090, Austria 2 MFPL, Dr Bohr Gasse, 1030 Vienna

Bacteria and fungi often live as commensal organisms as part of the human microbiota. An imbalance of the microbiome is associated with a shift from the commensal to the pathogenic state of a species which, together with a compromised immune system, leads to disease. Several studies have reported interactions between bacteria and fungi which might be either synergistic or antagonistic. Here, we characterize for the first time the interaction between Klebsiella and Aspergillus species which cohabit the lungs. In our studies, we have used microbiological (culture, microscopy) and molecular methods (qPCR) to investigate this interaction in vitro. The results have shown that Klebsiella pneumoniae is able to prevent Aspergillus spp. spore germination and hyphal development. Interestingly, this effect is only inhibitory since Aspergillus spp. recover growth upon elimination of bacteria. This inhibition is dependent on the physical presence and metabolically active Klebsiella cells. Molecular analysis of Klebsiella-Aspergillus interaction has shown upregulation of Aspergillus cell wall-related genes suggesting that Klebsiella induces cell wall stress response mechanisms. Characterization of polymicrobial interactions may be of relevance for the clinical management of infections, such as improved diagnostics and new treatment strategies.

S2-ST14 The Candida albicans HIR histone chaperone regulates the yeast-to-hyphae transition by controlling the sensitivity to morphogenesis signals

Sabrina Jenull1, Michael Tscherner1, Megha Gulati2, Clarissa J. Nobile2, Karl Kuchler1 1 Department of Medical Biochemistry, Medical University of Vienna, Max F. Perutz Laboratories, Dr. Bohr-Gasse 9/2, Vienna 1030, Austria 2 Department of Molecular and Cell Biology, School of Natural Sciences, University of California-Merced, Merced, CA, USA

Morphological plasticity such as the yeast-to-hyphae transition is a key virulence factor of the human fungal pathogen Candida albicans. Hyphal formation is controlled by a multilayer regulatory network composed of environmental sensing, signaling, transcriptional modulators as well as chromatin modifications. Here, we demonstrate a novel role for the replication-independent HIR histone chaperone complex in fungal morphogenesis. HIR operates as a crucial modulator of hyphal development, since genetic ablation of the HIR complex subunit Hir1 decreases sensitivity to morphogenetic stimuli. Strikingly, HIR1-deficient cells display altered transcriptional amplitudes upon hyphal initiation, suggesting that Hir1 affects transcription by establishing transcriptional thresholds required for driving morphogenetic cell-fate decisions. Furthermore, ectopic expression of the transcription factor Ume6, which facilitates hyphal maintenance, rescues filamentation defects of hir1Δ/Δ cells, suggesting that Hir1 impacts the early phase of hyphal initiation. Hence, chromatin-chaperone-mediated fine-tuning of transcription is crucial for driving morphogenetic conversions in the fungal pathogen C. albicans.

Page 24: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Fungal Response / Evasion to Host Defense

24

PL-07 Plasma membrane architecture: form and function in fungal pathogenesis

Lois Douglas1, Hong Wang0, Lifang Li0, Jenna Foderaro0, James Konopka0 1 Molecular Genetics & Microbiology, Stony Brook University, Life Sciences Bldg., Stony Brook 11794-5222, United States of America 0 Molecular Genetics & Microbiology, Stony Brook University, Life Sciences Bldg., Stony Brook, 11794-5222, United States of America

The important role that the plasma membrane plays in promoting the growth of fungal pathogens in the host is underscored by the fact that it is the target of most antifungal drugs and antimicrobial peptides. This protective barrier mediates a wide range of functions that are critical for virulence, including nutrient uptake, secretion, endocytosis, morphogenesis, cell wall synthesis, and stress resistance. To coordinate these diverse functions, the plasma membrane is organized into distinct compartments that vary in size, shape, and stability. One distinctive type of domain discovered in fungi consists of protein-organized domains called MCC/eisosomes that form stable furrows in the plasma membrane. In Candida albicans, mutants that fail to form MCC/eisosomes (pil1Δ lsp1Δ) have broad defects in cell wall synthesis, polarized morphogenesis, and invasive growth. In addition, MCC/eisosome mutants are more susceptible to a variety of stresses encountered in vivo, including elevated temperature, oxidation, copper and some antifungal drugs. Genetic analysis indicates that proteins recruited to the MCC/eisosomes, such as Sur7, Nce102, Pst1, Pst2, Pst3, and Ycp4, promote the proper regulation of cell wall synthesis and stress responses. Consistent with this, mutants lacking these MCC/eisosome proteins are defective in virulence. Thus, studies on MCC/eisosomes are defining novel plasma membrane functions that will promote a better understanding of the mechanism of action for current antifungal drugs and help to identify new targets for antifungal therapy.

SHORT TALKS

S2-ST15 Complement evasion of Aspergillus fumigatus by secretion of metalloprotease Mep1p

Sarah Sze Wah Wong1, Rajashri Shende2, Jean-Paul Latgé1, Arvind Sahu2, Vishukumar Aimanianda1 1 Unité des Aspergillus, Institut Pasteur, Paris, France 2 Complement Biology Laboratory, National Centre for Cell Science, Pune University, India

Aspergillus fumigatus is a ubiquitous opportunistic fungal pathogen that can cause lethal invasive pulmonary aspergillosis among immunocompromised populations. The host complement system, an important constituent of the innate immunity, plays a significant role in the defense against invading pathogens. However, a previous study showed that A. fumigatus mycelia secrete an alkaline protease, Alp1p, which degrades complement components, thereby, avoiding complement attack. The aim of our study was to examine complement system inactivation mechanism(s) associated with dormant A. fumigatus conidia, as conidium is the main morphotype encountering with the host immune system. A metalloprotease, Mep1p, released by conidia upon incubation with collagen, was shown to inactivate all the three complement pathways (classical, lectin and alternative) by degrading different complement components. Firstly, Mep1p indirectly inactivated C3, the central complement component; it cleaved C3 into C3b in the fluid phase, rather than on the pathogen surface, which would subsequently be cleaved by factor H and factor I, thus inactivating the alternative complement pathway. Secondly, Mep1p directly degraded complement components C4, an important component of the classic and lectin pathways as well as C5 involved in the formation of membrane attack complex. Mep1p could also degrade mannose-binding lectin and ficolin-1, the lectin pathway components. Furthermore, C3a, C4a and C5a, the anaphylatoxins/chemoattractants generated during complement activation, were also degraded by Mep1p. Consistently, it was also shown that Mep1p significantly inhibited conidial opsonisation, and thereby, inhibiting the phagocytic rate of macrophages. In conclusion, it was found that Mep1p is a protease released by dormant A. fumigatus conidia, which is responsible for the evasion of host complement attack.

Page 25: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Fungal Response / Evasion to Host Defense

25

S2-ST16 Structural differences on cell walls of the dimorphic fungal pathogens Sporothrix schenckii and Sporothrix brasiliensis impact human macrophage recognition.

Leila M. Lopes-Bezerra1, Hector Mora-Montes2, Louise Walker3, Gustavo Niño-Vega2, Gabriela Neves1, Carol Munto3, Neil Gow3 1 Lab. Cellular Mycology and Proteomics, Universidade do Estado do Rio de Janeiro, Rua São Francisco Xavier 524 PHLC sl 501D, Rio de Janeiro 20550-013, Brazil 2 Universidad de Guanajuato, Mexico 3 AFG group, University of Aberdeen, UK

Sporotrichosis is a subcutaneous mycosis caused by pathogenic species of the Sporothrix schenckii complex. A new emerging species, Sporothrix brasiliensis, is related to cat-transmitted sporotrichosis and to severe clinical manifestations. The cell wall of pathogenic fungi is a unique structure and can impact the host immune response. We reveal and compare the cell wall structures of S. brasiliensis and S. schenckii sensu stricto. High pressure freezing electron microscopy was used to study the cell wall organization of both Sporothrix species. To analyse the components of the cell wall we also used infrared spectroscopy, and 13C and 1H NMR. The sugar composition was determined by quantitative high-performance anion-exchange chromatography. To determine whether differences in the cell wall of Sporothrix spp. affected interactions with innate immune cells, we compared the uptake of both species by human macrophages. Our data revealed a bi-layered cell wall structure for both species, presenting an external microfibrillar layer and an inner electron-dense layer. The inner and outer layers of the S. brasiliensis cell wall are thicker compared to S. schenckii s. str., correlating with an increase of 30% in the chitin content and 100% in the rhamnose content. Moreover, the outer microfibrillar layer of S. brasiliensis cell wall could have longer microfibrills. The structural differences in the cell wall of these Sporothrix species had an impact on its uptake by human macrophages. Our data show a unique cell wall structure of S. brasiliensis and S. schenckii s. str. during the yeast parasitic phase. This model suggests that these fungi moult cell wall layers that may have significant effects on localized and disseminated immunopathology.

S2-ST17 Host-pathogen interaction: insights from proteomics V. Vialás1, J.A. Reales-Calderón1, V. Cabezón1, A. Gil de Bona1, A. Pitarch1, M.L. Hernaez1, M. Ramsdale2, V. Casas3, M. Carrascal3, J. Abián3,Zhi Sun4, Eric W. Deutsch4, Robert L. Moritz4, L. Monteoliva1, G. Molero1, C. Gil1

1Universidad Complutense de Madrid, Departamento de Microbiología II, Plaza de Ramón y Cajal s/n, 28040-Madrid, Spain.

2Biosciences, University of Exeter, UK. 3 CSIC/UAB Proteomics Laboratory, IIBB-CSIC, Barcelona, Spain. 4Institute for Systems Biology, Seattle, WA, USA.

Host-pathogen interaction studies open interesting opportunities in the search of new virulence determinants and new targets for antimicrobial therapies. We are studying the Candida albicans-macrophages interaction, because together with neutrophils are the first line of host immune defenses. We have analyzed protein differential expression of the ingested yeast using gel (DIGE- Differential In-Gel Electrophoresis) and non-gel proteomics (ITRAQ - Isobaric Tagging for Relative and Absolute Quantification). We provide evidence of a rapid protein response of the fungus to adapt to the new environment inside the phagosome by changing the expression of proteins belonging to different pathways. Network analyses allowed us generate a hypothetical model of Candida cell death after macrophage interaction, highlighting the interconnection between actin cytoskeleton, mitochondria and autophagy in the regulation of apoptosis. Different apoptotic markers like DNA fragmentation, ROS production, and caspase activity were assayed and our hypothesis was confirmed. Targeted proteomic assays of 32 proteins related with apoptosis is underway. In addition to provide new and expanded proteome documentation of the opportunistically pathogen C. albicans, we have developed new protein extraction and analysis routines to provide a new, extended and enhanced version of the C. albicans PeptideAtlas. Two new datasets, resulting from experiments consisting of exhaustive subcellular fractionations and different growing conditions, plus two additional datasets from previous experiments on the surface and the secreted proteomes, have been incorporated to increase the coverage of the proteome. High resolution precursor mass spectrometry (MS) and ion trap tandem MS spectra were analyzed with three different search engines using a database containing allele-specific sequences. This novel approach combined with the post-processing and filtering implemented in the Trans Proteomic Pipeline consistently used in the PeptideAtlas project resulted in 49372 additional peptides and 1630 more proteins identified in the new C. albicans PeptideAtlas with respect to the previous build. A total of 71310 peptides and 4174 canonical (minimal non-redundant set) proteins (4115 if one protein per pair of alleles is considered) were identified representing 66% of the 6218 proteins in the predicted proteome. This C. albicans PeptideAtlas will provide a number of useful features, like comprehensive protein and peptide-centered data exploration tools, and it will stand as a valuable resource to assist targeted proteomic experiments aiding in the selection of candidate proteotypic peptides.

Page 26: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Fungal Response / Evasion to Host Defense

26

Afternoon Session 3

Chair & Introduction

Attila Gácser

University of Szeged, Kozep fasor 52; H-6726 Szeged, Hungary

Page 27: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Fungal Response / Evasion to Host Defense

27

PL-08 Evolving antifungal drug resistance - a strategy of Candida albicans to overcome the last host defense

Joachim Morschhäuser

Institute for Molecular Infection Biology, University of Würzburg, Josef-Schneider-Str. 2, Würzburg 97080, Germany

When intrinsic host defenses fail, even usually harmless fungi can cause symptomatic infections, and to combat the pathogens the affected individuals have to employ external weapons - antifungal drugs. Fluconazole, which inhibits ergosterol biosynthesis, is commonly used to treat infections by Candida albicans. However, C. albicans can develop resistance to fluconazole, especially during long-term treatment of immunocompromised patients. In this lecture, I will give an overview on the molecular mechanisms of fluconazole resistance, the genetic changes involved, and their consequences for the fitness of C. albicans. In addition, I will present a new hypothesis on how highly drug-resistant strains with multiple resistance mechanisms may evolve.

SHORT TALKS

S2-ST18 Protein quality control at the inner nuclear membrane sets a threshold that determines the basal expression of virulence factors in Candida albicans

Fitz Gerald Silao, Kicki Ryman, Per Ljungdahl

Department of Molecular Biosciences, Stockholm University, Stockholm, Sweden SE-10691, Sweden

Integral membrane proteins localized to the inner nuclear membrane (INM) of the nuclear envelope participate in a variety of important processes, including the maintenance of nuclear architecture, chromatin organization, signaling and gene expression. In Saccharomyces cerevisiae, the INM Asi1/Asi3 complex, comprised of two integral INM proteins with RING domains, function as an E3-ubiquitin ligase. The Asi1/Asi3-complex plays an essential role in clearing the nuclear compartment from mislocalized soluble and integral membrane proteins; ubiquitylated proteins are targeted to proteasomes for degradation. Consistent with its important role, cells lacking a functional Asi complex exhibit pleiotropic phenotypes. Specifically, mutant cells accumulate many integral membrane proteins that mislocalize to the INM including the ER protein Erg11. Also, mutant cells exhibit constitutive transcription of SPS-sensor regulated genes, indicating that the Asi complex normally functions to prevent unprocessed and mislocalized Stp1 and Stp2 from gaining access to SPS-sensor regulated promoters. The genome of the pathogenic yeast Candida albicans encodes only one of the Asi components, namely CaASI3 (CR_08650C). We postulated that unregulated nuclear access of transcription factors, such as Stp1 and Stp2, could result in inappropriate expression of genes affecting virulence and that the overabundance of critical membrane proteins, i.e., Erg11, could affect ergosterol levels in a manner leading to altered sensitivities to antifungal agents that target sterol biosynthesis. To address these possibilities, we used CRISPR/Cas9 gene editing to create null alleles of CaASI3. Here we show that similar to finding in S. cerevisiae, asi3 null mutant strains of C. albicans exhibit constitutive expression of some SPS-sensor controlled genes, including Stp2 regulated CAN1. Consistent with these findings, C. albicans asi3 mutants exhibit enhanced virulence in a Drosophila model of infection. Also, the mutant strains exhibit tolerance towards the Erg11 inhibitor fluconazole. The emerging insights about the role of INM in both sequestration and clearance of transcription factors provide an exciting field of study to understand C. albicans virulence. Furthermore, the difference in the number of Asi proteins between S. cerevisiae and C. albicans may shed insight into the evolution of virulence between these two species.

Page 28: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Fungal Response / Evasion to Host Defense

28

S2-ST19 Potassium uptake and homeostasis in Candida species

Hana Sychrova, Hana Elicharova, Vicent Llopis-Torregrosa, Barbora Hušeková

Dept. Membrane Transport, Inst. Physiology CAS, Videnska 1083, Prague 4 14220, Czech Republic

The regulation of ion and pH homeostases is an essential process critical for cell viability. The maintenance of high intracellular concentrations of potassium and neutral pH is important for a variety of cellular functions including cell volume, DNA integrity, protein modification and trafficking. It is becoming increasingly evident that the coordination between primary H+-ATPases and transport systems involved in the influx and efflux of potassium allows this pH maintenance to occur. Genes encoding three types of potassium transporters have been found in genomes of pathogenic Candida species. In silico studies revealed that Candida spp. differ in the number and type of potassium transporters. Whereas C. albicans possesses genes for all three known types of transporters (Trk1, Hak1, Acu1), C. glabrata genome contains only one gene encoding a putative Trk1 uniporter. In the present study, we show 1) that all three genes of C. albicans encode efficient potassium uptake systems, and 2) that the C. glabrata Trk1 is crucial not only for potassium uptake but also for cell fitness and virulence. Deletion of the TRK1 gene in C. glabrata has a pleiotropic effect on several physiological parameters, such as intracellular pH, membrane potential, tolerance to cationic drugs, external pH and toxic cations. Preliminary results also show that, compared to the wild type, the mutant cells are less virulent in insect models, have different surface hydrophobicity and ability to form biofilms. Taken together, our results find the sole potassium uptake system in C. glabrata cells to be a promising target in the search for its specific inhibitors and in developing new antifungal drugs.

Acknowledgments

This work was supported by grants from the Czech Science Foundation (GA CR 16-03398S) and from the FP7-PEOPLE-2013-ITN ImresFun (606786).

Page 29: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Fungal Response / Evasion to Host Defense

29

PL-09 Genome indexing by histone H3 variants in Candida albicans

Kaustuv Sanyal

Molecular Biology & Genetics Unit, JNCASR, Bangalore 560064, India

Each cell in a multi-cellular organism carries the same genomic content but not all genes are expressed in every cell of the same organism. Multiple mechanisms have been shown to be involved in converting a genome into several lineage-specific “epigenomes”. One of the provocative hypotheses is many similar forms of histone proteins, popularly known as variant histones, may play critical roles in this process. Among the four core histones, histone H3 variants have been shown to be important in indexing mammalian genomes in transcriptionally active or silenced domains. A centromere-specific histone H3 variant, CENP-A, present in centromeres of most known organisms, is a classic example of how a specialized function can be defined on a eukaryotic chromosome by a histone H3 variant. Most unicellular organisms, such as popular model yeasts, Saccharomyces cerevisiae or Schizosaccharomyces pombe, do not need to undergo cellular differentiation. Thus, these organisms have CENP-A but lack variants of canonical histone H3. Interestingly, we discovered presence of both CENP-A and variants of canonical histone H3 in the genomes of a group of Candida species. Our results suggest that the variant histone H3 regulates expression of a large repertoire of genes to determine the planktonic versus biofilm mode of growth in Candida albicans.

Page 30: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Keynote Lecture

30

ImResFun2017 Keynote Plenary Lecture

KN- 02 MelLec: A new player in antifungal immunity

Gordon Brown

University of Aberdeen, MRC Centre for Medical Mycology, Institute of Medical Sciences, Ashgrove Road West, Aberdeen, UK

.

Page 31: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

From Fungal Biofilms to Intestinal Disorders 31

Symposium 4

From Fungal Biofilms to Intestinal Disorders

Chair & Introduction

Bernhard Hube

Hans Knöll Institute, A.-Reichwein-Str. 23, 7745 Jena, Germany

Page 32: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

From Fungal Biofilms to Intestinal Disorders 32

PL-10 Candida albicans gastrointestinal colonization and dissemination in the mammalian host

Andrew Koh

Pediatrics and Microbiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas 75390-9063, United States of America

The gastrointestinal (GI) tract is a source for invasive bacterial and fungal (e.g. Candida albicans) infections in cancer and stem cell transplant patients. By utilizing a murine model of C. albicans (CA) GI colonization and dissemination, we have been able to dissect critical host factors that prevent CA dissemination after administration of cytotoxic chemotherapy. Since CA colonization is required for invasive disease, more recent work has explored the mechanisms by which the murine gut promotes CA colonization resistance. Specifically, we have demonstrated that commensal anaerobic bacteria are critical for maintaining CA colonization resistance in mice via activation of the transcription factor HIF-1a and the antimicrobial peptide LL-37 (CRAMP in mice). Furthermore, pharmacologic activation of these GI intestinal immune effectors significantly decreases CA gut colonization and more importantly can significantly decrease mortality from disseminated disease. Current work explores the mechanisms by which gut commensal bacteria induce these gut immune effectors and whether anaerobic bacterial metabolic byproducts are important for maintaining CA colonization resistance.

SHORT TALKS

S3-ST20 A serum disaccharide signature of invasive fungal infections detected by MALDI-MS

Boualem Sendid1, Marjorie Cornu2, Alexandre Mery3, Yann Guerardel3, Daniel Poulain2 1 Parasitology Mycology, Inserm, University Lille 2, Place Verdun, Lille 59037, France 2 Parasitology Mycology, Inserm, University Lille2 3 UGSF, CNRS UMR 8576, Lille, France

We recently developed a mass spectrometry (MS) procedure based on the detection of a serum disaccharide (MS-DS) in patients with invasive candidiasis (IC). The aim of this study is to compare the performance of MS-DS for the diagnosis of IC, invasive aspergillosis (IA), and mucormycosis (MM) with those of commercially available antigen detection tests. This retrospective study included 48 patients (23 IC patients [74 serum samples], 15 IA patients [40 serum samples], and 16 MM patients [36 serum samples]) and 49 appropriate controls (102 serum samples). MS-DS, mannan (Mnn), galactomannan (GM), and (1,3)-beta-D-glucan (BDG) were detected by matrix-assisted laser desorption ionization–time of flight (MALDI-TOF) MS, Platelia, and Fungitell assays, respectively. For IC, the sensitivity and specificity of the MS-DS index, BDG detection, and Mnn detection were 62% and 84%, 82% and 60%, and 33% and 94% per serum sample and 83% and 69%, 96% and 31%, and 39% and 86% per patient, respectively. For IA, the corresponding values in comparison to BDG and GM detection were 83% and 81%, 62% and 95%, and 62% and 100% per serum sample and 93% and 76%, 87% and 90%, and 93% and 100% per patient, respectively. For the MM, preliminary results show a different circulation kinetics of DNA and MS-DS. MS-DS gave an early diagnosis in IC (73% positivity before blood culture), IA (positive before GM detection in six patients), and MM (positivity mainly preceded the date of diagnosis and complements qPCR results) patients. For IC, persisting MS-DS was associated with a poor prognosis. The different biomarkers were rarely detected simultaneously, suggesting different kinetics of release and clearance. For IA, MS-DS provided better complementation to GM monitoring than BDG monitoring. MS-DS detects panfungal molecules circulating during invasive fungal infections. MS-DS assay compared favorably with those of biological tests currently recommended for monitoring at-risk patients. Further validation of this test in multicenter prospective studies is required.

Page 33: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

From Fungal Biofilms to Intestinal Disorders 33

S3-ST21 New perspectives for fungal research in Ecuador

Francisco Javier Alvarez Botas

Yachay Tech University, Hacienda San José s/n - Proyecto Yachay, Urcuquí 100115, Ecuador

Latin American countries such as Ecuador have been investing a lot in education and research over the last few years. As a result, several opportunities have emerged for sharing the scientific knowledge and open collaborations with international scientific Institutions. A few research projects in the field of fungal microbiology and host-pathogen interactions could only take place in Ecuador or neighbour countries in Latin America, such as those aiming to look at Candida species in the microflora of isolated indigenous populations, or investigating their traditional remedies in use against fungal infections. This could provide us with new insights on the evolutionary aspects of Candida species and their pathogenicity. At the same time, screening for natural compounds in use by indigenous populations could help us identify new potential antifungal drugs. Accomplishment of such projects would best benefit from collaborations with established institutions from developed countries.

S3-ST22 Candida in the elderly

Uwe Groß

Universitätsmedizin Göttingen, Institut für Medizinische Mikrobiologie, Kreuzbergring 57, 37075 Göttingen, Germany

Page 34: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

From Fungal Biofilms to Intestinal Disorders 34

PL-11 Recent evolution of C. glabrata Toni Gabaldón

SHORT TALKS

S3-ST23 The broad and malleable phenotypic space of clinical Candida glabrata strains. A high throughput mapping of phenotypes reveals highly individualistic adaptations to a fluctuating environment.

Christoph Schüller1, Reinhard Beyer1, Birgit Willinger2, Joseph Strauss1 1 Universität für Bodenkultur, Konrad Lorenz Strasse 24, Tulln a.d. Donau 3430, Austria 2 Medizinische Universität Wien, Klinische Abteilung für Klinische Mikrobiologie Währinger Gürtel 18-20, Wien

Candida glabrata is a successful commensal and able to adapt to a variety of environmental situations and a cause of vulvo-vaginal candidiasis and shows high resistance to azole antifungals. To explore how this flexibility is possible, we investigated the phenotypic variation of several hundred C.glabrata clinical isolates collected by the Vienna General Hospital. Our quantitative high-throughput determination of growth parameters revealed a broad variation of phenotypes. For example temperature influenced growth rate in an unexpected range. We found that many strains tolerated either higher or lower temperatures while all strains had a temperature optimum at around 39°C. The collection was also scored for resistance to low pH, weak acid, osmotic stress, carbon source utilization and resistance to antifungals. Interestingly, the population spread for heat stress resistance while it remained confined for osmitic stress. This argues for the polygenetic basis of heat stress versus only few key processes contribute to osmotic stress. Furthermore, about 15% of these strains are efficiently forming biofilms. We further investigated the stability of some phenotypes. We tried to manipulate fluconazole resistance by adapting sensitive strains to intermediate fluconazole concentrations. This succeeded but lead to a simultaneous post-adaption loss of growth rate on rich medium, indicating that adaptation to fluconazole comes at a cost. Genome sequencing of selected phenotypically eye-catching strains revealed that their genomic profiles differed in up to 105 bases making specific assignments difficult. For stress resistance we analyzed the general role of CgHog1 and found a widely differing spectrum of basal Hog1 phosphorylation and stress response rates. The overall change of CgHog1 phosphorylation correlated with stress resistance of the particular strains. We conclude that these C. glabrata clinical isolates exhibit a broad phenotypic spectrum. Further investigation of the genetic and epigenetic basis of these variations and their stability are required to understand the success of C. glabrata as an opportunistic commensal.

Page 35: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

From Fungal Biofilms to Intestinal Disorders 35

Afternoon Session 4

Chair & Introduction

Karl Kuchler

Medical University Vienna, Dr. Bohr-Gasse 9/2, 1030 Vienna, Austria

Page 36: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

From Fungal Biofilms to Intestinal Disorders 36

PL-12 Discovery of a Novel Bacterial Endosymbiont Living within the Human Fungal Pathogen Candida albicans

Clarissa J. Nobile, Thaddeus Seher, Veronica Craik

Molecular and Cell Biology, University of California Merced, 5200 North Lake Road, Merced 95343, United States of America

An infection is often treated as if it is composed of a single microbial species in isolation, yet in reality, infections are immensely complex ecosystems composed of many interacting microbes. In this talk, we discuss our recent discovery of a bacterial endosymbiont living inside Candida albicans, the most common fungal pathogen of humans. We found that the presence of this endosymbiont influences the ability of C. albicans to form biofilms. Biofilms are resilient and drug-resistant communities of microbial cells tightly attached to mucosal surfaces or implanted medical devices that have significant relevance for human health.

SHORT TALKS

S4-ST24 Adhesin variability in clinical isolates of Candida glabrata and Candida parapsilosis

Emilia Gomez-Molero1, Ana Moreno2, Henk Dekker3, Toni Gabaldon4, Attila Gacser5, Uwe Groß1, Michael Weig1, Piet de Groot2, Oliver Bader1 1 Medical Microbiology, University Medical Center Göttingen, Kreuzbergring 57, Göttingen 37075, Germany 2 Regional Center for Biomedical Research, Albacete Science and Technology Park, University of Castilla-La Mancha, Albacete, Spain 3 Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands 4 Comparative Genomics Group, CRG-Centre for Genomic Regulation, Barcelona, Spain 5 Department of Microbiology, University of Szeged, Szeged, Hungary

Candida glabrata and Candida parapsilosis have gained significant importance over the past decades as one of the principal causes of fungal bloodstream infections. The capacity of both species to adhere to host cells and clinically used abiotic surfaces (e.g. prostheses or catheters) is crucial during colonization processes and the establishment of infections. Both species’ genomes encode an extensive number of potential adhesins whose roles remain uncharacterized. Due to the extensive number, we hypothesized that the presence of the adhesion-encoding genes in both species might be differentially modulated in clinical isolates, potentially conferring different biofilm formation capacities.

We analyzed biofilm formation capacity and differences in morphotypes among several hundred clinical isolates and performed cell-wall proteomic analyses of selected hyperbiofilm-forming isolates.

Approx. 20% of clinical C. glabrata and 24% of C. parapsilosis isolates presented a hyperbiofilm-forming phenotype, but no correlation with clinical parameters (i.e body site of isolateon, presence of indwelling device) was evident. For C. parapsilosis were able to confirm that differences in colony morphology seen on agar plates correlate with biofilm formation capacity and agar invasion.

In addition to a core cell wall proteome consisting of carbohydrate-active enzymes an increased number of different adhesins were detected in the cell walls of hyperbiofilm-forming isolates of both species. Downstream analyses showed distinct correlations of phenotypes such as altered virulence or morphology with adhesin incorporation, as well as distinct genomic alterations in hyper biofilm-forming isolates.

The high prevalence of drug resistance and capacity to form biofilms on abiotic and biotic surfaces makes both C. glabrata and C. parapsilosis important targets for development of specific diagnostics and treatment strategies. Here, differences in C. glabrata and C. parapsilosis cell wall adhesion incorporation among clinical isolates which reflect variations in infection-relevant parameters may yield novel approaches.

Page 37: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

From Fungal Biofilms to Intestinal Disorders 37

S4-ST25 SPS-sensor and metabolic control of amino acid-induced morphogenesis in Candida albicans

Fitz Gerald S. Silao, Kicki Ryman, Per O. Ljungdahl

Molecular Biosciences, The Winner-Gren Institute, Stockholm University, , Stockholm SE-10691, Sweden

Amino acids are well-defined environmental inducers of morphogenesis in the human fungal pathogen Candida albicans. A clear mechanistic understanding of how amino acids initiate yeast-to-filamentous switching is lacking. We have previously shown that several amino acids stimulate filamentation in a manner dependent on the plasma membrane-localized SPS-sensor of extracellular amino acids. Here we show that the apparent SPS-sensor dependency is indirect, being the consequence of SPS-sensor induced expression of a discrete set of amino acid permease genes, and that the filament-inducing effect of amino acids is derived from their intracellular metabolism. Thus, the amino acid inducing signals are initiated from within cells rather than from their being sensed as components of the extracellular environment. Specifically, the strong inducers arginine, ornithine and proline are internalized and exclusively metabolized to glutamate in the mitochondria, generating reduced electron carriers NADH and FADH2 as catabolic by-products. These reduced electron carriers are oxidized by the mitochondrial electron transport chain, a process coupled to the generation of ATP. Our data are inconsistent with the notion that arginine induces morphogenesis primarily through the generation of CO2. Together, our results provide the basis of an alternative unifying model in which the catabolism of arginine (and ornithine) to glutamate via proline in the mitochondria elevates intracellular levels of ATP, which triggers the Ras1/cAMP/PKA pathway to activate the morphogenic effector transcription factor Efg1. The metabolic activation of Egf1 induces the genetic programs that engage the yeast-to-filamentous switch. This model is consistent with the notion that enhanced mitochondrial ATP production stimulates adenylate cyclase (Grahl et al., 2015, PLOS Pathogens).

S4-ST26 Markerless genome editing in Candida albicans

Aaron Hernday, Namkha Nguyen, Morgan Quail

University of California Merced, 5200 N Lake Rd, Merced 95343, United States of America

We report the development of a rapid CRISPR-mediated genome editing system for use with Candida albicans. This efficient system can generate targeted genome modifications without introducing markers at the target locus, and without any molecular cloning. Efficient marker recycling methods ensure that no permanent markers are left in the genome, thus enabling endless iterations of strain engineering. We also demonstrate the ability to delete two different genes in a single transformation, and methods for generating homozygous “addback” strains by restoring wild-type alleles at the native locus. This system is also designed to be compatible with high-throughput strain engineering approaches.

Page 38: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

From Fungal Biofilms to Intestinal Disorders 38

PL-13 Investigation of the underlying mechanisms of antimicrobial drug tolerance of single and dual-species Candida albicans and Staphylococcus aureus biofilms using in vitro and in vivo model systems

Patrick Van Dijck1, Wafi Siala2, Sona Kucharikova1, Erik Kong3, Yves Dufrêne4, Mary Ann Jabra-Rizk3, Françoise Van Bambeke2 1 VIB-KU Leuven Center for Microbiology, VIB, KU Leuven, Kasteelpark Arenberg 31, Leuven, Belgium 2 Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, UCL, Brussels, Belgium 3 University of Baltimore, Dental School, Baltimore, USA 4 Institute of Life Sciences, UCL, Louvain La Neuve, Belgium

In our lab, we are studying human fungal pathogens, such as Candida albicans and Candida glabrata. These species are commensal organisms in most people, but in immuno-compromized patients, these pathogens can become deadly organisms. Each species has a set of virulence factors. The most important virulence factor in C. albicans is the yeast-to-hyphae morphogenetic switch and in C. glabrata, the expression of up to 70 different adhesion proteins. Currently a major problem in hospitals with these species is that they easily form biofilms on different types of implants, such as catheters. A biofilm is a community of cells that protect themselves from the immune system and from antifungal drugs by producing an extracellular matrix and by modifying gene expression. In such a biofilm cells need to attach to the substrate as well as to each other. We are investigating the proteins that are involved in these interactions and how different species (bacterial and fungal) may affect each other’s virulence characteristics. We use various tools to investigate the interaction between fungal and bacterial biofilms. We use molecular biology and cell biology tools but also atomic force microscopy (AFM). In addition, we developed in vivo biofilm model systems to study mixed species biofilms.

In the host, single fungal biofilms are identified but in many cases mixed populations of bacterial and fungal cells are found within a biofilm. Depending on the bacteria, these can be antagonistic (e.g. Pseudomonas aeruginosa kills hyphal Candida cells) or synergistic. Examples for the latter type of interaction will be presented. Using AFM and in vivo biofilm model systems, we show that C. albicans and S. aureus work synergistically against antimicrobial drugs when they are growing in biofiolms. We show that the fungal matrix protects bacterial cells against antibiotics.

SHORT TALKS

S4-ST27 The Candidalysins are a family of fungal toxins

Jonathan Richardson, Shir-Lynn Tan, Giulia Carrano, Julian Naglik

King's College London, Newcomen Street, London SE1 1UL, United Kingdom of Great Britain and Northern Ireland

Candida albicans is a commonly encountered opportunistic fungal pathogen of humans responsible for mucosal and life threatening systemic ailments that contribute to high morbidity and mortality worldwide. During mucosal infection, C. albicans grows as infiltrating pathogenic hyphae that secrete Candidalysin, a 31 amino acid peptide toxin derived from its parent protein, Ece1p. Candidalysin destabilises the structural integrity of epithelial cell plasma membranes, facilitating infection. Epithelial cells respond to Candidalysin through a bi-phasic p38/c-Fos signalling pathway that culminates in a strong inflammatory response at mucosal surfaces. Analysis of Ece1p amino acid sequences from different Candida species revealed the presence of additional putative Candidalysin toxins in C. dubliniensis, C. tropicalis, C. albicans 529L and C. maltosa. We used solid phase peptide synthesis to generate these peptide sequences and quantified their ability to cause damage, activate mucosal inflammatory responses and the p38/c-Fos response pathway in oral (TR146) and vaginal (A431) epithelial cells. All putative toxins caused epithelial damage as evidenced by quantification of lactate dehydrogenase activity and induced epithelial signalling (p-38/c-Fos) and cytokine secretion. Notably, the putative toxins of C. tropicalis and C. dubliniensis exhibited greater potency in vitro when compared with the Candidalysin of C. albicans. These data suggest conservation of function between the different Candidalysins despite differences in amino acid sequence and identify the Candidalysins as a family of functionally related fungal toxins.

Page 39: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

From Fungal Biofilms to Intestinal Disorders 39

S4-ST28 Molecular mechanisms governing the formation of persister cells in Candida albicans biofilms

Iryna Denega, Christophe d'Enfert, Sophie Bachellier-Bassi

Mycology, Institut Pasteur, 25-28 rue du Dr. Roux, Paris 75015, France

Candida albicans, although frequently part of the human commensal flora, turns out to be the most prevalent yeast species involved in fungal infections in human. Candidiasis range from superficial mucocutaneous- to life-threatening systemic infections, associated with a mortality rate of more than 30% in immunocompromised patients.

C. albicans is also known for its ability to form biofilms – complex communities of microorganisms embedded in an extracellular matrix, developing upon attachment of the cells to a surface (living or inert). C. albicans biofilms display an intrinsic tolerance to antifungal agents. Apart from the biofilm matrix serving as an antifungal trap, an additional source of antifungal tolerance results from the occurrence in biofilms of so-called persister cells. These are phenotypic variants of wild-type cells, capable of surviving concentrations of antimicrobial agents well above the MIC.

Little is known about the mechanisms leading to the formation of persister cells in C. albicans, or the properties allowing them to survive high doses of antifungals.

The main objective of this project is to identify genes involved in C. albicans persister cells’ appearance. For this purpose, we will use different approaches, including (1) screening knockout mutants collections (focusing on transcription factors) for their persister-forming capability, (2) comparing genomes and transcript profiles of high-persister forming mutants, (3) developing a reporter system of persister cells’ appearance and (4) identifying persister cells’ specific transcripts, followed by the detailed characterization of the identified genes.

The knowledge gained from this project will uncover mechanisms underlying persister cells appearance in C. albicans, which are so far poorly studied. This will allow to develop approaches to potentiate the activity of existing antifungals towards persister cells and help in the treatment of C. albicans infections, preventing their relapse.

Page 40: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Poster Presentation 40

Poster Presentations

PP-01 Global transcriptome characterization of Candida glabrata biofilms in response to acetate and fluconazole

Rosana Alves1, Stavroula Kastora2, Eva Pinho3, Célia Rodrigues3, Sónia Silva3, Margarida Casal1, Alistair J Brown2, Mariana Henriques3, Sandra Paiva1 1 Centre of Molecular and Environmental Biology, Biology Department, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal 2 MRC Centre for Medical Mycology at the University of Aberdeen, Aberdeen Fungal Group, Institute of Medical Sciences, Aberdeen AB25 2ZD. 3 Centre for Biological Engineering, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal

Candida glabrata is considered a major opportunistic fungal pathogen of humans and has emerged as a leading cause of nosocomial fungal infections. The capacity of this yeast species to cause infections is dependent on the ability to grow within the human host environment and to assimilate the carbon sources available.

Previous studies have suggested that Candida can encounter glucose-poor microenvironments during infection and that the ability to use alternative non-fermentable carbon sources, such as carboxylic acids, contributes to the virulence of these fungi. Our recent study (Mota et al., 2015) supported this view by demonstrating that the host metabolite acetate influences C. glabrata biofilm formation, antifungal drug resistance and phagocytosis; and suggesting a potential role of putative carboxylate transporters on these processes.

In order to extend our studies and provide a comprehensive view of the C. glabrata biofilms’ response to alternative carbon sources and antifungal treatment, we performed comparative transcriptomics analyses using RNA-sequencing. An overview of the most significant results will be presented. Our data support the view that adaptative responses of Candida cells to the different carbon sources present in host niches affects their virulence through multifarious mechanisms.

Bibliography: Mota S, Alves R, Carneiro C, Silva S, Brown AJ, Istel F, Kuchler K, Sampaio P, Casal M, Henriques M and Paiva S (2015) Candida glabrata susceptibility to antifungals and phagocytosis is modulated by acetate. Front. Microbiol. 6:919.

PP- 02 RNAi-based Functional Genomics Identifies Myosin V and Phospholipase D as New Virulence Determinants in Mucormycosis. Francisco E. Nicolas, Maria I. Navarro-Mendoza, Trung A. Trieu, Carlos Perez-Arques, Marta Sanchis, Javier Capilla, Patricia Navarro-Rodriguez, Loida Lopez-Fernandez, Santiago Torres-Martinez, Rosa M. Ruiz-Vazquez, Victoriano Garre

Microbiology And Genetics, Universidad de Murcia, Campus de Espinardo, Murcia 30100, Spain

Mucorales are an emerging group of human pathogens that are responsible for the lethal disease mucormycosis. Unfortunately, functional studies on the genetic factors behind the virulence of these organisms are hampered by their limited genetic tractability, since they are reluctant to classical genetic tools like transposable elements or gene mapping. In this work, we describe an RNAi-based functional genomic platform that allows the identification of new virulence factors through a forward genetic approach firstly described in Mucorales. This platform contains a whole-genome collection of Mucor circinelloides silenced transformants that presented a broad assortment of phenotypes related to the main physiological processes in fungi, including virulence, hyphae morphology, mycelial and yeast growth, carotenogenesis and asexual sporulation. Selection of transformants with reduced virulence allowed the identification of mcplD, which encodes a Phospholipase D, and mcmyo5, encoding a probably essential cargo transporter of the Myosin V family, as required for a fully virulent phenotype of M. circinelloides. Knock-out mutants for those genes showed reduced virulence in both Galleria mellonella and Mus musculus models, probably due to a delayed germination and polarized growth within macrophages. This study provides a robust approach to study virulence in Mucorales and as a proof of concept identified new virulence determinants in M. circinelloides that could represent promising targets for future antifungal therapies.

Page 41: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Poster Presentation 41

PP- 03 Bioluminescent Mucor circinelloides – a promising new tool to study mucormycosis and antifungal drug efficacy

Ulrike Binder1, Maria Isabel Navarro-Mendoza2, Francisco Nicolas2, Cornelia Lass-Flörl1, Victoriano Garre2 1 Medical University Innsbruck, Division of Hygiene and Medical Microbiology, Austria 2 University of Murcia, Fungal Genomics and Biotechnology, Spain

Invasive infections caused by members of the Mucorales (mucormycosis) have increased in the last years, making it the third most common invasive fungal infection after aspergillosis and candidiasis. Despite this increasing clinical relevance, little is known about the establishment of disease, its progression and successful therapy.

New tools to study this disease in more detail are needed; therefore the objective of this work was to construct a luciferase expressing Mucor circinelloides strain, as one representative of mucormycosis causing pathogens. Here, we describe the construction and functional analysis a strain, which will further be used as a reporter system for in vivo and in vitro models of Mucorales infections.

A leucine auxotroph M. circinelloides strain, R7B, was used as recipient strain to allow selection of transformants on selective medium. Firefly luciferase gene without the peroxisomal target sequence was cloned in the pMAT1477 vector under the control of a constitutive promoter. Linear plasmid was used to transfect M. circinelloides protoplasts. The targeted integration of the whole construct in the carRP gene resulted in easy identification of transformants, appearing as white colonies.

Homokaryons were obtained by sequential plating on selective media and checked for light emission under various conditions in in vitro assays.

Expression of firefly luciferase was successful in M. circinelloides at several conditions and light emission was detectable by imaging and with a luminometer. Data so far indicate the strain being suitable for further in vivo and in vitro studies.Phenotype, virulence potential and antifungal susceptibility are currently compared to wild-type strains.

The construction of this first bioluminescent Mucor circinelloides strain will allow for the visualization of temporal and spatial progression of infection by a non-invasive method in insect and murine models, and the testing of antifungal efficacy by other means than survival only. This will give valuable new insights in the pathogenesis of Mucorales infections.

PP- 04 Autophagy contributes to intracellular control of Candida glabrata in macrophages

Nayeli Shantal Castrejón-Jiménez1, Frida Cardozo-Mata1, Itzama Iris Rodríguez-Morua1, Shantal Lizbeth Baltierra-Uribe2, Blanca Estela García-Pérez1 1 Microbiologia, Instituto Politécnico Nacional, Prolongación Carpio No. 538, Cdmx 11340, Mexico 2 Inmunologia, Instituto Politécnico Nacional, Prolongación Carpio No. 538, CDMX 11340, Mexico

Candida glabrata is considered an opportunistic fungus, together with C. krusei are now emerging as serious hospital acquired infections in immunocompromised patients. Both species are intrinsically resistant to fluconazole and other azoles, and frequency of isolations of these two species has significantly increased recently. It is know that C. glabrata survives and persist intracellularly in macrophages. Autophagy is a process that targets cellular cytoplasmic contents for lysosomal degradation and serves myriad roles in eukaryotic cells. Xenophagy is a type of selective macroautophagy that specifically targets intracellular pathogens to lysosomes, restricting their replication and survival. Autophagy is induced by inanition, hypoxia, rapamycin and TLRs ligands between others. The present study was designed to investigate if the induction of autophagy contributes to elimination of C. glabrata by macrophages. For that, monolayers of macrophages derived of monocytes THP1 cell line were infected by reference strain CBS138 C. glabrata. In some cases monolayers were previously treated with autophagy inductors (inanition, rapamycin and peptidoglycan). Autophagy was evidenced by immunofluorescence stain of LC3-II, a characteristic protein of autophagy. The effect of autophagy induced on C. glabrata elimination was followed by determination of UFC. The results demonstrated that C. glabrata induces the LC3-II expression. When cells were previously stimulated with autophagy inductors, LC3-II protein was visualized in the cytoplasm of the cells, in some cases colocalization of LC3-II with yeasts was notorious. The determination of UCF showed that the three inductors caused a decrease of intracellular yeast; rapamycin was the best stimuli to dead the intracellular yeast. Altogether, the results indicate that autophagy contributes to clearance of intracellular yeast in macrophages and enhance the role of autophagy in the control of intracellular pathogens.

Page 42: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Poster Presentation 42

PP- 05 Identification of drugs that enhance the fungicidal action of Amphotericin B or that present antifungal activity against Cryptococcus neoformans and Candida albicans

Haroldo Cesar De Oliveira, Suélen Andreia Rossi, Daniel Agreda Mellon, Oscar Zaragoza

National Centre for Microbiology, Instituto de Salud Carlos III, Carretera Majadahonda-Pozuelo, km 2, Majadahonda 28220, Spain

The treatment of invasive fungal infections is based on three antifungal families: polyenes (amphotericin B), azoles and echinocandins, being amphotericin B (AmB) the one with the strongest fungicidal activity. Nevertheless, the use of these drugs has important limitations as their price, toxicity and action spectrum, since not all antifungals are active against all species. An example is the pathogenic yeast Cryptococcus neoformans, that is intrinsically resistant to echinocandins. The treatment of cryptococcosis is based on an initial administration of AmB. However, this drug is very toxic and cannot be administered for a long time due kidney damage. The AmB action mechanism is not totally known. Classically its has been described that this drug binds to ergosterol causing pores at the plasma membrane, but recently it was demonstrated that AmB also induces an accumulation of reactive oxygen species (ROS), that participate in the fungicidal activity of the drug. Since AmB presents different action mechanisms, we decided to identify new compounds that enhance its activity. For this purpose, we used the Prestwick Chemical Library® that contains 1280 molecules approved by Food and Drug Administration (FDA), European Medicines Agency (EMA) and others regulatory agencies and which are not under patent. We performed a phenotypic screening of compounds that inhibited the growth of C. neoformans at sub-inhibitory concentrations of AmB. In addition, as a second objective, we identified compounds with antifungal activity against C. neoformans and C. albicans. We found nine compounds that did not significantly affect C. neoformans, but inhibited the growth of the yeasts in presence of AmB (0.03 mg/L). We next performed checkerboard assays, and confirmed the synergistic effect with AmB of all the selected compounds (FIC<0.5). We also identified 25 compounds that alone presented antifungal activity against C. neofomans, 18 against C. albicans and other 18 that had activity against both species. Among the identified compounds, we found different types of drugs such as antibiotics, antineoplastic, anti-inflammatory, antidepressant and others. Our results indicate that repurposing of drugs can contribute to develop new antifungal therapies and also to improve the available treatments.

PP- 06 Candida glabrata disrupt endolysosomal maturation in human osteoblasts.

Shantal Lizbeth Baltierra-Uribe1, Alfonso Cervantes-Contreras2, Nayeli Shantal Castrejón-Jiménez2, Blanca Estela Garcia Perez2 1 Inmunologia, Instituto Politécnico Nacional, Prolongación De Carpio Y Plan De Ayala S/N, Ciudad De México 11340, México 2 Microbiología, Instituto Politécnico Nacional, Prolongación De Carpio Y Plan De Ayala S/N, Ciudad De México 11340, México

Candida glabrata is the second most frequent cause of candidiasis after Candida albicans, accounting for approximately 15 to 25% of Candida infections. C. glabrata can invade by endocytosis into host cells.

Rab GTPases are critical for intracellular transport following pathogens endocytosis. Several pathogens deploy virulence factors to inhibit their dead by host cells and establish intracellular niches where their growth takes place. The osteoblasts cells are capable to internalize C. glabrata, and the yeast can survive and replicate, but the mechanisms of intracellular transport that C. glabrata exploit are unknown.

In this study we analyzed the trafficking of C. glabrata into osteoblast cell. For that, we evaluate the Rab GTPases involved in intracellular traffic, like Rab5 (early endosomal marker), Rab7 (late endosomal marker) and cathepsin D (to evaluated lysosomal fusion). As cellular model we used the MG-63 osteoblast cell line. Viable and heath-dead reference strain C. glabrata was employed to stimulated osteoblast cells. The MG-63 monolayers were incubated with live and dead yeast, the endosomal trafficking was monitored during 30, 60 and 120 min by immunofluorescence and confocal analysis. Immunofluorescence assays show that viable C. glabrata is not colocalized with Rab GTPases studied. However the dead yeasts were founded colocalized with Rab5, Rab7 and cathepsin D. In other hand we quantified the ROS production in osteoblasts stimulated with viable and dead yeast; both conditions did not induced significant production of this metabolite. These results indicated that viable yeast disrupt endolysosomal maturation suggesting C. glabrata modulated the trafficking to persist intracellularly into osteoblast and this cells can eliminated the dead yeast through lysosomal pathway independently ROS production.

Page 43: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Poster Presentation 43

PP- 07 Examination of virulence-related genes in the opportunistic human pathogen Candida parapsilosis

Sára Pál1, Tibor M. Németh1, Leszek Pryszcz2, Toni Gabaldon2, Csaba Vágvölgyi1, Attila Gácser1 1 Department of Microbiology, University of Szeged, Hungary 2 Bioinformatics and Genomics, Centre for Genomic Regulation, Barcelona, Spain

The number of Candida infections is increasing, due to numerous predisposing factors such as severe immunodeficiency, congenital immune defects, organ transplantation, prolonged use of implanted medical devices and extended antibiotic treatment. Among the species of this genera, C. albicans causes the majority of diseases, although the number of infections caused by the human commensal C. parapsilosis is also rising. Furthermore, C. parapsilosis infection rates can even exceed the candidaemia episodes caused by C. albicans in premature infants. Therefore, understanding the basis of C. parapsilosis pathogenicity is becoming an urging field to explore.

The aim of our project is to create an over-expression mutant library in order to study the role of fungal ORFs potentially involved in C. parapsilosis virulence. To identify such set of genes, transcriptional analysis of fungi co-incubated with THP-1 human monocytes was performed. Differentially expressed kinases and transcriptional factors and additional orthologous genes of C. albicans virulence regulatory ORFs (adherence, biofilm formation, antifungal resistance) were sentenced to over-expression. We used the Thermo Fisher Scientific Gateway cloning technology to create the mutant strains. Transformants were validated by colony-PCR, Southern blot and real-time PCR. The fitness of the verified mutants was monitored in vitro in the presence of different stressors that can influence the virulence of a species (temperature, pH, oxidative stressors, etc.). In order to confirm the role of the identified genes in host-pathogen interaction, we performed both in vitro and in vivo infections using human and murine macrophages and Galleria mellonella larvae.

With the help of these over-expression mutants we may have the potential to identify factors that can determine the virulence of the fungus and may serve as a target for antifungal therapy.

PP- 08 Bomanin (BomΔ55C) deficient Drosophila melanogaster as a host to assess virulence of human fungal pathogens

Kicki Ryman1, Vicent Llopis-Torregrosa2, Julia Koehler3, Hana Sychrová4, Ylva Engström1, Per Ljungdahl1 1 Dep of Molecular Biosciences, The Wenner-Gren Institute Stockholm University, SE-106 91 Stockholm, Sweden 2 Dep of Membrane Transport, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic 3 Div of Infectious Diseases, Boston Children's Hospital/Harvard Medical School, Boston, Massachusetts, USA 4 Dep of Membrane Transport, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic.

Studying infectious disease in the context of whole organisms requires suitable model host systems. Drosophila melanogaster is a versatile host to assess the virulence properties of human fungal pathogens. Wild-type D. melanogaster (OrR) flies are susceptible to virulent Candida albicans infections. However, although Candida glabrata causes virulent infections in humans, it is avirulent in OrR flies. Recognition of fungal cells in flies activates the Toll signaling pathway, triggering a robust induction of innate immune effectors, including the family of twelve Bomanin genes (Clemmons et al., 2015 PLoS Pathog 11: e1004876). The Bomanins (Boms) encode small secreted peptides that bestow resistance to multiple microbial pathogens. The BomΔ55C allele is 9 kb TALON-induced deletion that removes a cluster of 10 Bom genes on chromosome 2. BomΔ55C flies exhibit decreased survival upon microbial infection, similar to eliminating the essential Toll pathway. We report that BomΔ55C flies are suitable to assay C. glabrata virulence.

Page 44: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Poster Presentation 44

PP- 09 The effects of fungal and bacterial stimuli on the metastatic activity of oral squamous cell carcinoma cell lines

Máté Vadovics1, Nóra Igaz2, Judit Szenzenstein1, Dávid Kovács2, Csaba Vágvölgyi1, Mónika Kiricsi2, Attila Gácser1 1 Department of Microbiology, University of Szeged, Közép fasor 52., Szeged 6726, Hungary 2 Department of Biochemistry and Molecular Biology, University of Szeged, Közép fasor 52., Szeged 6726, Hungary

A large number of commensal microbial species reside in the human body that have co-evolved with the human genome and adopted to the host immune system. Previously it has been shown that defects in regulatory processes or alterations in the composition of microbiome can lead to various diseases, including cancer. Even though the human bacteriome is well-investigated, our knowledge is limited about the composition and function of the residential fungal microflora or mycobiome. The most frequent opportunistic fungal infection in the oral cavity is candidiasis. Previous studies have shown that the number of Candida cells in this particular niche is significantly higher in patients with oral squamous cell carcinoma (OSCC) compared to healthy individuals. It was suggested that tumour-induced immunosuppression could cause fungal overgrowth and contribute to the development of an infection. However, the etiological relationship between oral cancer and Candida infections is still a matter of debate. Our aim is to examine the influence of fungal or bacterial infections on the metastatic activity of cancerous cells. Therefore, cell proliferation and migration activity of HSC-2 and HO-1-N-1 OSCC cells were investigated after fungal and bacterial stimuli. Scratch wound assay was performed to monitor the migration of cancerous cells. The wound-healing capacity of HO-1-N-1 cells was significantly increased after the treatments compared to the untreated samples, however no significant differences could be identified in case of stimulated and unstimulated HSC-2 cells. The proliferation activity of OSCC cells was examined by BrdU assay. Both cell lines showed increased relative BrdU incorporation upon treatments, which clearly indicates that fungal or bacterial infections can accelerate cancer cell proliferation. To investigate the possible molecular mechanisms underlying the increased metastatic activity of OSCC cells after fungal or bacterial exposure, we examined the expression level of various TAM receptors, since they advance cancer cell migration by assisting the epithelial-mesenchymal-transition (EMT). Elevated expression of Tyro3, Axl and Mer in OSCC cells could be verified by RT-QPCR following microbial treatment. These findings suggest that the presence of microbes in the oral cavity can promote cancer cell proliferation, increased motility and metastatic activity of oral squamous cell carcinoma cells at least in part by the induction of TAM receptors.

PP- 10 Human macrophage response to Sporothrix spp. is dependent on thermo-labile human serum factors

Gabriela W.P. Neves1, Caetana Natália1, Janet Willment2, Carol Munro2, Neil Gow2, Gordon Brown2 and Leila Lopes-Bezerra1

1 Lab. Cellular Mycology and Proteomics, Universidade do Estado do Rio de Janeiro, Rua São Francisco Xavier 524 PHLC sl 501D, Rio de Janeiro 20550-013, Brazil 2 AFG group, University of Aberdeen, UK

Sporotrichosis is the most common human deep cutaneous mycosis reported in Latin America. The causative agents of human and animal sporotrichosis are species of the pathogenic clade of the Sporothrix genus: S. schenckii, S. brasiliensis, S. globosa, and S. lurei. Interestingly, cat-transmitted sporotrichosis is only related to S. brasiliensis infection. The innate immune system plays a key role in protection against fungal infections, but the host response against each fungal pathogen is particular and still poorly understood. In the present work a model based on human monocytes-derived macrophages (hMac) was used to study the host primary response to the yeast parasitic phase of two Sporothrix species. These species, S. schenckii and S. brasiliensis, are currently associated, in Brazil, with the human sporotrichosis and the cat-transmitted zoonosis, respectively. We already know that the cell wall structure and organization of these two species are rather distinct. We hereby show that hMac exhibited a distinct ability to uptake S. schenckii and S. brasiliensis. The hMac cytokine profile was also dissimilar as both species could induce TNF-α secretion but only S. brasiliensis had the capacity to induce a significant secretion of an anti-inflammatory cytokine, IL-10. Furthermore, in a parallel work we are showing that aging and/or nutritional stress can modify the cell wall architecture. Accordingly, the recognition of each species by hMAc is also impaired by the yeast cell growth condition. Besides, an important modulation of the Sporothrix spp. uptake was observed when the interaction assay was performed in the presence of heat inactivated human serum. Our results clearly show that a thermo-labile serum factor is important to hMac recognition, uptake and cytokine response to Sporothrix spp. To our knowledge, this is the first report to describe the interaction of primary cell culture human macrophages with the parasitic phase of S. schenckii and S. brasiliensis. Acknowledgements: This work was supported by Fundação Carlos Chagas de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ) and CNPq.

Page 45: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Author Index 45

Author Index

A

Abián, J. 25 Agreda Mellon, Daniel 42 Aimanianda, Vishukumar 24 Allert, Stefanie 14 Almeida, Sandro Rogério 18 Alvarez Botas, Francisco Javier 33 Alves, Rosana 40

B

Bachellier-Bassi, Sophie 39 Bader, Oliver 36 Baltierra-Uribe, Shantal Lizbeth 41, 42 Bauters, Anne 22 Beyer, Reinhard 34 Binder, Ulrike 41 Blagojevic, Mariana 17 Brown, Alistair J 40 Brown, Gordon 30, 44 Burger-Kentischer, Anke 18

C

Cabezón, V. 25 Caetana, Natália 44 Capilla, Javier 40 Cardozo-Mata, Frida 41 Carlström, Mattias 22 Carrano, Giulia 38 Carrascal, M. 25 Casal, Margarida 40 Casas, V. 25 Castrejón-Jiménez, Nayeli Shantal 41, 42 Catarina, Vaz 13 Cervantes-Contreras, Alfonso 42 Chauhan, Neeraj 19 Choteau, Laura 22 Concha, Gil 13 Cornu, Marjorie 32 Craik, Veronica 36 Curty, Nathália 15

D

De Graeve, Marilyn 20 de Groot, Piet 36 De Hoog, Sybren 18 De Oliveira, Haroldo Cesar 42 Dekker, Henk 36 Denega, Iryna 39 d'Enfert, Christophe 39 Denning, David W. 9 Deutsch, Eric W. 25

Douglas, Lois 24 Dubuquoy, Laurent 22 Dufrêne, Yves 38

E

Elicharova, Hana 28 Engström, Ylva 43

F

Finkelmeier, Doris 18 Fiori, Alessandro 20 Foderaro, Jenna 24 Fontaine, Thierry 15 Förster, Toni 14 François, Nadine 22 Freeke, Joanna 18

G

Gabaldón, Toni 34, 36, 43 Gacser, Attila 36 Gácser, Attila 43, 44 Garcia Perez, Blanca Estela 42 García-Pérez, Blanca Estela 41 Garre, Victoriano 41 Garre, Victoriano 40 Gil De Bona, A. 25 Gil, C. 25 Gil, Concha 15 Gloria, Molero 13 Gomez-Molero, Emilia 36 Gow, Neil 25, 44 Groß, Uwe 33, 36 Grumaz, Christian 18 Guerardel, Yann 22 Guerardel, Yann 32 Gulati, Megha 23 Gunzer, Matthias 14

H

Hebecker, Betty 14 Henkel, Helena 18 Henriques, Mariana 40 Hernaez, M.L. 25 Hernaez, Maria Luisa 15 Hernday, Aaron 37 Hofer, Anders 22 Hohl, Tobias 17 Holtappels, Michelle 20 Hosseinzadeh, Ava 22 Hube, Bernhard 14 Hušeková, Barbora 28

Page 46: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Author Index 46

I

Igaz, Nóra 44

J

Jabra-Rizk, Mary Ann 38 Jawhara, Samir 22 Jenull, Sabrina 23 Jose Antonio, Reales-Calderon 13 Juraschitz, Marc 14

K

Kang, Hyun Ah 20 Kaplan, Daniel H 13 Kasper, Lydia 14 Kastora, Stavroula 40 Kerstens, Winnie 20 Kim, Jeong-Yoon 20 Kim, Jong-Myeong 20 Kiricsi, Mónika 44 Koehler, Julia 43 Koh, Andrew 32 Kong, Erik 38 Konopka, James 24 Kovács, Dávid 44 Kremer, Anneke 20 Kucharikova, Sona 38 Kuchler, Karl 13, 19, 23 Kühbacher, Andreas 18

L

Lass-Flörl, Cornelia 41 Latgé, Jean-Paul 24 Lehrnbecher, Thomas 11 Li, Lifang 24 Lion, Thomas 23 Lippens, Saskia 20 Ljungdahl, Per 27, 43 Ljungdahl, Per O. 37 Llopis-Torregrosa, Vicent 28, 43 Lopes-Bezerra, Leila 15, 25, 44 Lopez-Fernandez, Loida 40 Lucía, Monteoliva 13

M

MERY, Alexandre 32 Mogavero, Selene 14 Molero, G. 25 Monteoliva, L. 25 Monteoliva, Lucia 15 Moon, Hye Yun 20 Mora-Montes, Hector 25 Moreno, Ana 36 Moritz, Robert L. 25 Morschhäuser, Joachim 27 Munro, Carol 44 Munto, Carol 25

N

Naglik, Julian 14, 38 Naglik, Julian R 17 Navarro-Mendoza, Maria I. 40, 41 Navarro-Rodriguez, Patricia 40 Németh, Tibor M. 43 Neves, Gabriela 15, 25 Neves, Gabriela W.P 44 Nguyen, Namkha 37 Nicolas, Francisco 41 Nicolas, Francisco E. 40 Niño-Vega, Gustavo 25 Nobile, Clarissa J. 23, 36 Nogueira, Filomena 23

O

Oliveira Vaz, Catarina 15

P

Paiva, Sandra 40 Pál, Sára 43 Paul-Latgé, Jean 15 Pawlik, Tony 14 Pereira, Leonel 23 Perez-Arques, Carlos 40 Pinho, Eva 40 Pitarch, A. 25 Pitarch, Aida 15 Poulain, Daniel 22 Poulain, Daniel 32 Pryszcz, Leszek 43

Q

Quail, Morgan 37

R

Ramsdale, M. 25 Reales-Calderón, J.A. 25 Reales-Calderon, Jose Antonio 15 Richardson, Jonathan 38 Richardson, Jonathan P 17 Riedelberger, Michael 13 Rodrigues, Célia 40 Rodríguez-Morua, Itzama Iris 41 Rossato, Luana 18 Rossi, Suélen Andreia 42 Ruiz-Vazquez, Rosa M. 40 Rupp, Steffen 18 Ryman, Kicki 27, 37, 43

S

Sahu, Arvind 24 Sanchis, Marta 40 Santos, Suelen Silvana 18

Page 47: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Author Index 47

Sanyal, Kaustuv 29 Scheidl, Bernhard 13 Schüller, Christoph 34 Schulz, Daniela 14 Seher, Thaddeus 36 Sendid, Boualem 22 Sendid, Boualem 32 Shankar, Madhu 22 Shende, Rajashri 24 Shivarathri, Raju 19 Siala, Wafi 38 Silao, Fitz Gerald 27 Silao, Fitz Gerald S. 37 Silva, Sónia 40 Singh, Nitesh Kumar 19 Sohn, Kai 18 Soloviev, Dmitry 22 Stevens, Philip 18 Strauss, Joseph 34 Sun, Zhi 25 Sychrova, Hana 28 Sychrová, Hana 43 Szenzenstein, Judit 44

T

Tan, Shir-Lynn 38 Tardivel, Meryem 22 Torres-Martinez, Santiago 40 Trieu, Trung A. 40 Tscherner, Michael 19, 23

U

Urban, Constantin Felix 22

V

Vadovics, Máté 44 Vágvölgyi, Csaba 43, 44 Van Bambeke, Françoise 38 Van Bogaert, Inge 20 van de Veerdonk, Frank 19 Van Dijck, Patrick 20, 38 Vancraeyneste, Hélène 22 Vialás, V. 25 Vodnala, Munender 22

W

Walker, Louise 25 Wang, Hong 24 Weig, Michael 36 Willinger, Birgit 34 Willment, Janet 44 Wong, Sarah Sze Wah 24

Y

Yellagunda, Sujan 22

Z

Zaragoza, Oscar 42 Zwolanek, Florian 19

Page 48: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Participants Addresses 48

Participant Addresses

Allert Stefanie Microbial Pathogenicity Mechanisms Hans-Knoell-Institute Adolf-Reichwein-Straße 23 07745 Jena, Germany P: +4936415321141 E: [email protected]

Alvarez Botas Francisco Javier School of Biological Sciences and Engineering Yachay Tech University Hacienda San José s/n - Proyecto Yachay 100115 Urcuquí, Ecuador P: +593 (0)988455486 E: [email protected]

Alves Rosana Centre of Molecular and Environmental Biology, Biology Dept University of Minho Campus de Gualtar 4710-057 Braga, Portugal P: 00351253601514 E: [email protected]

Bader Oliver Medical Microbiology University Medical Center Göttingen Kreuzbergring 57 37075 Göttingen, Germany P: +49 551 39 22346 E: [email protected]

Binder Ulrike Division of Hygiene and Medical Microbiology Medical University Innsbruck Schöpfstrasse 41 6020 Innsbruck, Austria P: +43 512 9003 70748 E: [email protected]

Blagojevic Mariana King's College London Newcomen Street SE1 1UL London, United Kingdom of Great Britain and Northern Ireland P: +447417380001 E: [email protected]

Brown Gordon D. MRC Centre for Medical Myology University of Aberdeen Ashgrove Road West ab252zd Aberdeen, United Kingdom of Great Britain and Northern Ireland P: 01224437355 E: [email protected]

Burger-Kentischer Anke Molecular Biotechnolgy Fraunhofer-Institute IGB Nobelstrasse 12 70569 Stuttgart, Germany P: +49 711 970 4023 E: [email protected]

Castrejón-Jiménez Nayeli Shantal Microbiologia Instituto Politécnico Nacional Prolongación Carpio No. 538 11340 Ciudad de México, Mexico P: +525557296300 E: [email protected]

d'Enfert Christophe Fungal Biology and Pathogenicity Unit, Dpt of Mycology Institut Pasteur 25 rue du Docteur Roux 75015 Paris, France P: +33140613257 E: [email protected]

Dalle Frederic University of Burgundy Esplanade Erasme 21000 Dijon, France P: +33 6 79244821 E: [email protected]

De Oliveira Haroldo Cesar National Centre for Microbiology Instituto de Salud Carlos III Carretera Majadahonda-Pozuelo, km 2 28220 Majadahonda, Spain P: +34 652 741274 E: [email protected]

Denega Iryna Mycology Institut Pasteur 25-28 rue du Dr. Roux 75015 Paris, France P: +33619882993 E: [email protected]

Denning David W National Aspergillosis Centre The University of Manchester Southmoor Road M23 9LT Manchester, United Kingdom of Great Britain and Northern Ireland P: +441612915811 E: [email protected]

Gabaldon Toni Centre for Genomic Regulation (CRG) Dr. Aiguader, 88 08003 Barcelona, Spain P: +34 616953333 E: [email protected]

Gacser Attila Department of Microbiology University of Szeged Kozep fasor 52 6726 Szeged, Hungary P: +3662544849 E: [email protected]

Garcia Perez Blanca Estela Microbiologia Instituto Politécnico Nacional PROLONGACIÓN DE CARPIO Y PLAN DE AYALA S/N 11340 Ciudad de México, Mexico P: +525557296300 E: [email protected]

Gil Concha Microbiologia II, Facultad de Farmacia Universidad Complutense de Madrid Plaza Ramon y Cajal s/n 28004 Madrid, Spain P: 913941755 E: [email protected]

Glaser Walter Max F. Perutz Laboratories Dr. Bohrgasse 3 1030 Vienna, Austria P: + 43 - 1 - 40160 - 34101 E: [email protected]

Gregori-Schüller Christa Max F. Perutz Laboratories Medical University Vienna Dr. Bohr-Gasse 9/2 1030 Vienna, Austria P: +43-1-4277-61806 E: [email protected]

Groß Uwe Medical Microbiology University Medical Center Göttingen Kreuzbergring 57 37075 Göttingen, Germany P: +49 551 395801 E: [email protected]

Gunzer Matthias Institute for experimental Immunology and Imaging University Hospital Essen Hufelandstr. 55 45147 Essen, Germany P: +49 0201 183-6641 E: [email protected]

Henkel Helena Fraunhofer-Institut für Grenzflächen- und Bioverfahrenstechn Nobelstraße 12 70569 Stuttgart, Germany P: +49 711 970-4135 E: [email protected]

Hernday Aaron University of California Merced 5200 N Lake Rd 95343 Merced, United States of America P: 1-209-228-2450 E: [email protected]

Hohl Tobias Department of Medicine Memorial Sloan Kettering Cancer Center 1275 York Avenue 10065 New York, United States of America P: 646-888-3596 E: [email protected]

Holtappels Michelle Molecular Microbiology VIB, KU Leuven Kasteelpark Arenberg 31 3001 Leuven, Belgium P: +3216374703 E: [email protected]

Hube Bernhard Microbial Pathogenicity Mechanisms Hans Knöll Institute A.-Reichwein-Str. 23 07745 Jena, Germany P: +49 3641 5321401 E: [email protected]

Page 49: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Participants Addresses 49

JAWHARA Samir Inserm U995-LIRIC/Team2 Faculté de médecine Pole Formation, 1 place Verdun 59000 Lille, France P: +33644193655 E: [email protected]

Jenull Sabrina Department of Medical Biochemistry Medical University of Vienna, Max F. Perutz Laboratories Dr. Bohr-Gasse 9/2 1030 Vienna, Austria P: +431-4277-61818 E: [email protected]

Kaplan Daniel H Dermatology University of Pittsburgh 200 Lothrop St 15260 Pittsburgh, United States of America P: 011 412-648-9471 E: [email protected]

Kim Jong-Myeong Chungnam National University 99 Daehak-ro 34134 Daejeon, Korea, Republic of P: +82 10 6625 9116 E: [email protected]

Klein Bruce Pediatrics and Medical Microbiology University of Wisconsin-Madison 1550 Linden Drive 53706 Madison, United States of America P: 608-263-9217 E: [email protected]

Koh Andrew Pediatrics and Microbiology University of Texas Southwestern Medical Center 5323 Harry Hines Boulevard 75390-9063 Dallas, United States of America P: +1 214-648-8802 E: [email protected]

Konopka James Molecular Genetics & Microbiology Stony Brook University Life Sciences Bldg. 11794-5222 Stony Brook, United States of America P: +1 631-632-8715 E: [email protected]

Ksiezopolska Ewa The Centre for Genomic Regulation (CRG) Dr. Aiguader, 88 08003 Barcelona, Spain P: +34 619263262 E: [email protected]

Kuchler Karl Max F. Perutz Laboratories Medical University Vienna Dr. Bohr-Gasse 9/2 1030 VIenna, Austria P: +43-1-4277-61807 E: [email protected]

Kühbacher Andreas Fraunhofer IGB Nobelstraße 12 70569 Stuttgart, Germany P: +49 711 970-4133 E: [email protected]

Lehrnbecher Thomas University of Frankfurt Theodor Stern Kai 7 60590 Frankfurt, Germany P: +49 69 6301 83481 E: [email protected]

Lion Thomas CCRI/LabDia Zimmermannplatz 10 1090 Vienna, Austria P: +4340470 4890 E: [email protected]

Ljungdahl Per O. Molecular Biosciences, The Winner-Gren Institute Stockholm University Svante Arrhenius väg 20C SE-10691 Stockholm, Sweden P: +468164101 E: [email protected]

Lopes-Bezerra Leila M. Lab. Cellular Mycology and Proteomics Universidade do Estado do Rio de Janeiro Rua São Francisco Xavier 524 PHLC sl 501D 20550-013 Rio de Janeiro, Brazil P: +55 21 23340835 E: [email protected]

Molero Gloria Microbiology Ii, Faculty of Pharmacy University Complutense of Madrid Plaza de Ramón y Cajal s/n 28040 Madrid, Spain P: +34 91 3942084 E: [email protected]

Morschhäuser Joachim Institute for Molecular Infection Biology University of Würzburg Josef-Schneider-Str. 2 97080 Würzburg, Germany P: +49-931-3182152 E: [email protected]

Naglik Julian King's College London St Thomas Street SE1 1UL London, United Kingdom of Great Britain and Northern Ireland P: +44 20 7848 6123 E: [email protected]

Neves Gabriela Laboratory of Cellular Mycology and Proteomics Universidade do Estado do Rio de Janeiro Rua São Francisco Xavier, 524 20.550-013 Rio de Janeiro, Brazil P: +552123340835 E: [email protected]

Nicolas Molina Francisco E. Microbiology and Genetics Universidad de Murcia Campus de Espinardo 30100 Murcia, Spain P: +34648104533 E: [email protected]

Nobile Clarissa J. Molecular and Cell Biology University of California Merced 5200 North Lake Road 95343 Merced, United States of America P: 2092282427 E: [email protected]

Nogueira Filomena CCRI Zimmermannplatz 1090 Vienna, Austria P: +43 1 404704491 E: [email protected]

Oliveira Vaz Catarina Microbiologia II, Facultad de Farmacia Universidad Complutense de Madrid Plaza Ramon y Cajal s/n 28040 Madrid, Spain P: 913941755 E: [email protected]

Pál Sára Department of Microbiology University of Szeged Közép fasor 52. 6726 Szeged, Hungary P: +36 30 2194282 E: [email protected]

Petryshyn Andriy Max F. Perutz Laboratories Medical University Vienna Dr. Bohr-Gasse 9/2 1030 Vienna, Austria P: +43-1-4277-61806 E: [email protected]

Richardson Jonathan King's College London Newcomen Street SE1 1UL London, United Kingdom of Great Britain and Northern Ireland P: +44 20 7848 6121 E: [email protected]

Riedelberger Michael Department for Medical Biochemistry Medical University of Vienna Dr. Bohrgasse 9 1030 Vienna, Austria P: +431427761812 E: [email protected]

Rossato Luana University of São Paulo Lineu Prestes, 580 05508-000 São Paulo, Brazil P: +55 11 971224947 E: [email protected]

Rupp Steffen Molecular Biotechnolgie Fraunhofer-Institute IGB Nobelstrasse 12 70569 Stuttgart, Germany P: +49 711 970 4059 E:[email protected]

Ryman Kicki The department of Molecular Biosciences WGI Svante Arrheniusv 20b 10691 Stockholm, Sweden P: +468162836 E: [email protected]

Sanyal Kaustuv Molecular Biology & Genetics Unit JNCASR Jakkur 560064 Bangalore, India P: +918022082878 E: [email protected]

Schwarzmüller Maria Max F. Perutz Laboratories Dr. Bohrgasse 3 1030 Vienna, Austria P: 00436503264614 E: [email protected]

Page 50: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

Participants Addresses 50

Schüller Christoph DAGZ, Department of Applied Genetics and Cell Biology University of Natural Ressources Vienna Konrad Lorenz Stresse 24 3430 Tulln a.d. Donau, Austria P: 014765494188 E: [email protected]

SENDID Boualem Parasitology Mycology Inserm, University Lille 2 Place Verdun 59037 Lille, France P: +33 6 23 54 20 45 E: [email protected]

Shivarathri Raju Medical Biochemistry Medical university of Vienna, Mfpl Dr. Bohrgasse-9/2 1030 Vienna, Austria P: +431427761818 E: [email protected]

Silao Fitz Gerald Department of Molecular Biosciences Stockholm University Svante Arrhenius väg 20C SE-10691 Stockholm, Sweden, Sweden P: +468162836 E: [email protected]

Sychrova Hana Dept. Membrane Transport Inst. Physiology CAS Videnska 1083 14220 Prague 4, Czech Republic P: +420 608 198 989 E: [email protected]

Urban Constantin Felix Clinical Microbiology Umeå University Sjukhusområdet 6C 90185 Umeå, Sweden P: +46907852702 E: [email protected]

Vadovics Máté Department of Microbiology University of Szeged Közép fasor 52. 6726 Szeged, Hungary P: +36 62 544 005 E: [email protected]

van de Veerdonk Frank Department of Internal Medicine Radboud University Medical Center Geert Grooteplein Zuid 10 6525 GA Nijmegen, Netherlands P: +31 24 361 1111 E: [email protected]

Van Dijck Patrick VIB-KU Leuven Center for Microbiology VIB, KU Leuven Kasteelpark Arenberg 31 B-3001 Leuven, Belgium P: +3216321512 E: [email protected]

Wong Sarah Sze Wah Institut Pasteur 25-28 rue du Dr Roux 75015 Paris, France P: +33699162243 E: [email protected]

Page 51: ,P5HV)XQ )LQDO 0HHWLQJ 0DULH &XULH ,QLWLDO …imresfun.cdl.univie.ac.at/finalmeeting/pub/ImResFun2017-Abstract... · ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction,

ImResFun2017 - Molecular Mechanisms of Fungal Pathogen-Host Interaction, Sept. 3 - Sept. 7, 2017, Innsbruck, Austria

51

Organizers & ImResFun2017 National Guard

Organizers Karl Kuchler & Steffen Rupp

ImResFun2017 Meeting Office Christa Gregori-Schüller & Maria Schwarzmüller

Technical Support, Registration, Website & Abstract Book Walter Glaser & Christa Gregori-Schüller & Maria Schwarzmüller

”ImResFun2017 National Team”

Sabrina Jenull

Andriy Petryshyn

Michael Riedelberger

Raju Shivarathri