22nd Annual Meeting of the Society of Virology (GfV) Essen, Germany

50
22 nd Annual Meeting of the Society of Virology (GfV) Essen, Germany 14 – 17 March 2012

Transcript of 22nd Annual Meeting of the Society of Virology (GfV) Essen, Germany

22nd Annual Meeting of the

Society of Virology (GfV)

Essen, Germany

14 – 17 March 2012

TABLE OF CONTENTS FluResearchNet at the 22nd Annual Meeting of the GfV ......................... 3

ORAL PRESENTATIONS ...................................................................... 7

POSTER PRESENTATIONS ................................................................. 19

PUBLICATIONS 2011 - 2012 .............................................................. 39

FluResearchNet ANNOUNCEMENTS 2012 ............................................ 47

FluResearchNet at the 22nd Annual Meeting of

the GfV

Dear friends and colleagues,

Dear supporters of the FluResearchNet,

established in 2007 the FluResearchNet has developed a dynamic interdisciplinary network

on influenza research including 17 individual research projects. Actually these projects are

running at 13 different research organizations. Thereby FluResearchNet combines expertises

of different disciplines and organizations in a fruitful team.

Although we have learned much about influenza during the outbreak of 2009 pandemic

influenza A (H1N1), many research questions are still open. This is shown impressively by

the controversial dual-use research debate on H5N1 influenza strains. Regarding the

scientists’ perspective these experiments show that there is indeed a H5N1 pandemic threat.

On the other hand the knowledge about correlation of few mutations and pathogenicity offer

the chance to improve prevention and risk assessment. In our opinion it is absolutely

necessary to discuss the various aspects of this debate with different stakeholders in an

unbiased manner.

The results of the FluResearchNet projects may contribute to a better understanding of

pathogenicity, host susceptibility, and transmission across species barriers regarding

influenza.

Once again the FluResearchNet demonstrate its power by 20 presentations at the

22nd Annual Meeting of the Society of Virology in Essen. With this volume we summarize the

FluResearchNet abstracts representing the most recent research data of the network

members. Please use the opportunity in Essen and visit the FluResearchNet presentations.

Please notice the first FluResearchNet member meeting 2012

taking place in Essen on Friday, 16th March, lunchtime (Room B,

6th floor). Please consider further FluResearchNet

announcements in this volume and on www.fluresearchnet.de.

Meeting you in Essen, sincerely yours

Stephan Ludwig

– FluResearchNet Coordinator –

Page 6 of 49

ORAL PRESENTATIONS

Page 8 of 49

Session

VECTORS, NEW VACCINES

Thursday, March 15, 2012 Venue: Room A1 Time: 04.00 pm – 05.30 pm

Oral presentations

Page 10 of 49

Individual FluResearchNet project no. 6

Principal investigator: Prof. Dr. S. Ludwig

Introduction of silent mutations into the NP gene of influenza A viruses as a novel

strategy to generate a live attenuated influenza vaccine.

Anhlan Darisuren1, Eike-Roman Hrincius1, Christoph Scholtissek2, and Stephan Ludwig1

1Institute of Molecular Virology, Center for Molecular Biology of Inflammation, University of

Münster, Von-Esmarch-Str. 56, 48149 Münster 2Wald-Str. 53, 35440 Linden

The nucleoprotein (NP) of influenza A virus (IAV) is associated with many different functions

including host range restriction. Multiple sequence alignment analyses of about 600 NP gene

sequences from GenBank revealed a highly conserved region of about 80 nucleotides within

the ORF at the 3´ends of the cRNA, in which even silent variations were absent. This

suggests that the RNA structure integrity within this region is crucial for IAV replication. To

explore the impact of these conserved nucleotides for viral replication we created mutant

viruses with one or more silent mutations in the respective region of the NP gene of the IAV

strain A/WSN/33 (H1N1) (WSN). Assessment of viral replication of these WSN mutant viruses

showed significant growth disadvantages when compared to the corresponding parental

strain. On the basis of these findings we tested whether the attenuation of IAV by

introduction of silent mutations into the NP gene may serve as a strategy to create alive

attenuated vaccine. Mice vaccinated with the apathogenic WSN mutant survived a lethal

challenge dose of wild type WSN virus or the mouse adapted pandemic H1N1v strain

A/Hamburg/4/2009. Thus, introduction of silent mutations in the NP of IAV is a feasible

approach that leads to attenuation of the live vaccine but leaves the antigenicity of the gene

product unaltered. This principle is potentially applicable for all viruses with segmented

genomes.

Session

INNATE IMMUNE RESPONSE I

Friday, March 16, 2012 Venue: Room A1 Time: 08.30 am – 10.00 am

Oral presentations

Page 12 of 49

Individual FluResearchNet project no. 6

Principal investigator: Prof. Dr. S. Ludwig

The NS1 protein of influenza A virus suppresses RIG-I mediated activation of the

alternative NF-κB pathway and p52/RelB dependent gene expression in lung

epithelial cells

Andrea Rückle1, Emanuel Haasbach2, Oliver Planz2, and Stephan Ludwig1

1Institute of Molecular Virology, Center for Molecular Biology of Inflammation, University of

Münster, Von-Esmarch-Str. 56, 48149 Münster 2 Interfaculty Institute for Cell Biology, Department of Immunology, University of Tuebingen,

Auf der Morgenstelle 15, 72076 Tuebingen

Activation of NF-κB transcription factors is a major cellular response to virus infections,

including influenza A viruses (IAV). Two major NF-κB activating signalling pathways exist in

cells. The classical pathway involves degradation of IκBα resulting in the nuclear

translocation of p65/p50 dimers. The alternative pathway regulates proteolytic processing of

NF-κB2/p100 to form p52/RelB dimers. It has been previously shown that in IAV infected

cells the classical NF-κB pathway is not only crucial for expression of major antiviral

cytokines but also is required for efficient viral replication via its apoptosis regulating

features. In contrast, nothing is known so far about a potential role of the alternative NF-κB

pathway in IAV infection. Here we show for the first time that wt IAV is a poor activator of

the alternative NF-κB pathway, while a mutant IAV lacking the viral NS1 protein (delNS1)

strongly induces alternative NF-κB signalling, resulting in processing of p100 and nuclear

translocation of p52/RelB. The RNA receptor RIG-I was identified to mediate alternative NF-

κB activation. In clear contrast to the classical NF-κB pathway, disruption of alternative

signalling had no effect on viral replication, however, p52/RelB was shown to regulate IAV

induced expression of the chemokine CCL19. Based on these results we hypothesize that

alternative NF-κB signalling is relevant to mount the systemic immune response to IAV

infection and therefore is strongly antagonized by NS1.

Oral presentations

Page 13 of 49

Individual FluResearchNet project no. 6

Principal investigator: Prof. Dr. S. Ludwig

Phosphatidylinositol-3-kinase (PI3K) isactivated by influenza virus vRNA via the

pathogen pattern receptorRig-I to promote efficient type I interferon production

Rüdiger Dierkes, Eike R. Hrincius, Darisuren Anhlan, Viktor Wixler, Stephan Ludwig, and

Christina Ehrhardt

Institute of Molecular Virology, Center for Molecular Biology of Inflammation, University of

Münster, Von Esmarch-Str. 56, 48149 Muenster

The phosphatidylinositol 3-kinase (PI3K) is activated upon influenza A virus infection in a

biphasic manner. An early and transient induction of PI3K signaling is induced by viral

attachment to cells and promotes virus entry. In later phases of the infection cycle the

kinase is activated by direct interaction with the viral NS1 protein leading to prevention of

premature apoptosis induction. Besides these virus-supporting functions, it was also

suggested that PI3K signaling is essential for full activation of interferon regulatory factor 3

(IRF-3) in response to synthetic dsRNA and IAV infections. However, a direct role of PI3K

signaling in the innate immune response to influenza virus infections was not described yet.

Here we show that accumulation of vRNA in human lung epithelial cells infected with either

influenza A or B viruses results in PI3K activation. Furthermore, expression of the RNA

receptors Rig-I and MDA5 was increased upon stimulation with virion-extracted vRNA or IAV

infection. Using siRNA approaches, Rig-I was identified as the pathogen receptor necessary

for influenza virus vRNA sensing and subsequent PI3K activation in a TRIM25 and MAVS

signaling dependent fashion. Rig-I induced PI3K signaling was further shown to be essential

for full IRF-3 activation resulting in the induction of the type I interferon response. These

data show that PI3K is activated as part of the Rig-I mediated anti-pathogen response to

enhance expression of type I interferons.

Oral presentations

Page 14 of 49

Session

INNATE IMMUNE RESPONSE II

Friday, March 16, 2012 Venue: Room A1 Time: 10.30 am – 12.00 am

Oral presentations

Page 16 of 49

Individual FluResearchNet project no. 6

Principal investigator: Prof. Dr. S. Ludwig

The influenza virus PB1-F2 protein has interferon-antagonistic activity

Sabine E. Dudek, Ludmilla Wixler, Carolin Nordhoff, Alexandra Nordmann, Darisuren Anhlan,

Viktor Wixler, and Stephan Ludwig

Institute of Molecular Virology, Center for Molecular Biology of Inflammation, University of

Münster, Von-Esmarch-Str. 56, 48149 Münster

PB1-F2 is a non-structural protein of influenza viruses encoded by the PB1 gene segment

from a +1 open reading frame. It has been shown that PB1-F2 contributes to viral

pathogenicity even though the underlying mechanisms are still under debate. Induction of

type I interferon (IFN) is a first line of defense against viral infections. Here we show that

influenza A viruses lacking the PB1-F2 protein induced enhanced expression of IFN-β and

interferon-stimulated genes (ISGs) in infected epithelial cells demonstrating a type I IFN-

antagonistic activity of PB1-F2. On a molecular level PB1-F2 interfered with the RIG-I/MAVS

protein complex thereby inhibiting the activation of the downstream transcription factor

IRF-3. These findings were also reflected in in vivo studies demonstrating that infection with

PR8 wild type (wt) virus resulted in higher lung titers and a more severe onset of disease

compared to infection with its PB1-F2 deficient counterpart. Accordingly, a much more

pronounced infiltration of lungs with immune cells was detected in mice infected with the

PB1-F2 wt virus. In summary, we have identified PB1-F2 protein as a second type I IFN-

antagonistic protein of influenza A viruses that acts via interference with the RIG-I/MAVS

complex, thereby contributing to enhanced pathogenicity in vivo.

Oral presentations

Page 17 of 49

Individual FluResearchNet project no. 6

Principal investigator: Prof. Dr. S. Ludwig

MAPKAP kinase 3 inhibits IFNgamma secretion upon influenza A virus infection

Katharina Köther1, Carolin Nordhoff1, Matthias Gaestel2, Viktor Wixler1, Stephan Ludwig1

1Institute of Molecular Virology, Center for Molecular Biology of Inflammation, University of

Münster, Von-Esmarch-Str. 56, 48149 Münster 2Hannover Medical School, Institute of Biochemistry, Carl-Neuberg-Str. 1, 30625 Hannover

The replication of influenza A virus is strongly dependent on the host cell and in part exploits

the cellular signaling machinery for its own purpose. One of the signaling pathways that is

activated by virus infection is the p38 MAPK pathway. While this pathway is activated by the

cell as part of the antiviral response e.g. to stimulate cytokine expression, it also exhibits

virus-supportive activity, e.g. via activation of the p38 downstream substrates MAPKAP

kinases 2 (MK2) and 3 (MK3). In vitro studies showed that absence of MK2 or MK3 led to a

strong reduction of viral titers, most likely due to the lack of PKR inactivation. In in vivo

studies we were able to confirm the influenza A virus supportive function of the MK3 kinase.

The MK3 knockout mice showed a reduction in the viral titer and an enhanced survival

compared to wt mice. Furthermore, comparative analysis of the immune status of infected

mice showed that the MK3 kinase is also involved in regulation of the host innate immune

response. The MK3 knockout mice differed in their expression and/or secretion of IFN

gamma after influenza virus infection. Especially the number of IFN gamma expressing

natural killer (NK) cells was increased in MK3 knockout mice compared to wt mice. This data

suggests that MK3 is involved in regulation of IFN gamma upon influenza A virus infection.

Oral presentations

Page 18 of 49

POSTER

PRESENTATIONS

Page 20 of 49

Poster Viewing 1

Workshop topic:

Negative Strand RNA Viruses

Wednesday, March 14, 2012 Venue: Poster area Time: 07.30 pm – 09.00 pm

Poster presentations

Page 22 of 49

Workshop topic: Negative Strand RNA Viruses

Individual FluResearchNet project no. 1 | Poster no. 123

Principal investigator: PD Dr. T. Wolff

Revisiting the antiviral ISG15 system and its role in the replication of human and

animal influenza A viruses

J. Knepper1, V. K. Weinheimer1, T. Wolff1

1Div. of Influenza/Respiratory Viruses, Robert Koch Institute, Berlin,

Influenza A viruses (IAV) infect a wide range of avian and mammalian species. However,

there is still only incomplete knowledge about the virus- and host-encoded factors involved

in IAV host range restriction. ISG15 is a ubiquitin-like polypeptide that can be covalently

attached to target proteins in a process mediated by the ligases Ube1L, UbcH8 and Herc5.

Previous data of our group showed variations in the upregulation of ISG15 in human cells

upon infections with human, avian and porcine IAV. The underlying mechanisms responsible

for these differences and the impact of ISG15 on IAV propagation as well as species

specificity are subject of our investigation.

In our study we used infections of human lung cells, transfection-based experiments in

HEK 293T cells and Western Blot as well as FACS analyses.

Our initial investigations revealed a strong induction of ISG15 in A549 cell cultures upon

infection with seasonal IAV. Interestingly, we observed on a single cell level that this

induction occurred predominantly in uninfected cells, whereas little ISG15 was detected in

the initially infected cells. In contrast, a population of both infected and ISG15-positive cells

was present after infection with a ΔNS1 mutant virus. A similar effect was observed upon

infections with porcine and low pathogenic avian IAV. Furthermore, transfection-based

experiments elucidated that the sole expression of the viral polymerase in combination with

viral RNA was sufficient to induce ISG15, which was inhibited by co-expressed viral NS1

protein.

In conclusion, our results indicate that the NS1 protein of seasonal IAV suppresses ISG15

induction during infection. Ongoing analyses address the roles of ISG15 and NS1 in human

cells infected with non-human IAV.

Poster presentations

Page 23 of 49

Individual FluResearchNet project no. 2 | Poster no. 118

Principal investigator: Prof. Dr. G. Herrler | Prof. Dr. Schwegmann-Weßels

Adaptation of avian influenza viruses to growth in differentiated porcine airway

epithelial cells

DARSANIYA PUNYADARSANIYA1, Fandan Meng1 HENNIG-PAUKA I.2, WINTER C.1,3, SCHWEGMANN-

WESSELS C.1, HERRLER G.1

1Institute of Virology, University of Veterinary Medicine, Hannover 2Clinic for swine, small ruminants and forensic medicine, University of Veterinary Medicine,

Hannover 3Clinic for Poultry, University of Veterinary Medicine, Hannover

Swine are an important host for the epidemiology and interspecies transmission of influenza

A viruses. The differentiated epithelial cells of the respiratory tract are the primary target

cells for influenza virus infection. To analyze the infection of porcine airway epithelial cells by

influenza viruses, we established precision-cut lung slices as a culture system for

differentiated respiratory epithelial cells. A comparison of the infection by a porcine (H3N2

subtype) and an avian (H9N2 subtype) showed that the avian virus was inferior in the

following parameters: (i) production of infectious virus, (ii) length of replication cycle, (iii)

ciliostatic effect. To analyze the adaptation of avian influenza viruses to the porcine

respiratory epithelium, the H9N2 was passaged three times in precision-cut slices prepared

from the porcine lung. Titration of the infectious virus released into the supernatant revealed

that the virus recovered from the third passage was characterized by a shorter replication

time resembling that of the porcine virus. On the other hand, the amount of infectious virus

and the ciliostatic effect were not affected by the adapation process. These results show that

the replication time is the first parameter that is affected during adaptation of the avian

H9N2 virus to growth in porcine respiratory eptihelial cells. The mutations that are correlated

with these changes are currently determined.

Poster presentations

Page 24 of 49

Individual FluResearchNet project no. 4 | Poster no. 120

Principal investigator: Prof. Dr. M. Schwemmle

Influenza A virus NEP is a potent co-factor of the viral polymerase promoting viral

replication by direct interaction with PB2

Peter Reuther1, 2, Linda Brunotte1, Benjamin Mänz1, Martin Schwemmle1

Department of Virology1, Faculty of Biology2 University of Freiburg, 79104 Freiburg

Zoonotic transmission of avian influenza viruses into the human population requires adaptive

mutations to achieve high-level replication in the new host. We provide evidence that NEP

plays a crucial role in the adaptation of avian H5N1 viruses. Specifically, NEP containing

adaptive mutations overcomes a failure of avian H5N1 polymerases to generate bona fide

cRNA templates, resulting in enhanced viral replication and transcription. By mutational

analysis we could demonstrate that the polymerase-activity enhancing function of NEP

resides in its C terminus while the N terminus negatively regulates this function most likely

by binding to the C terminus. We could further show that the adaptive mutations in NEP

reduce the inhibitory effect exerted by the N-terminus of this protein. Most strikingly, NEP

function is mediated by interaction between NEP C terminus and the polymerase subunit

PB2, which we could show by co-immunoprecipitation studies and functional analysis. A

more detailed study of the NEP-PB2 interaction demonstrated that the C terminus of NEP

targets a 50 amino acid comprising motif of PB2 adjacent to the Cap-binding domain. These

data indicate that NEP is a novel and potent co-factor of the viral polymerase promoting viral

replication by a direct interaction with the PB2 subunit.

Poster presentations

Page 25 of 49

Individual FluResearchNet project no. 10 | Poster no. 100

Principal investigator: Prof. Dr. R. Zell

Genotyping of swine influenza A viruses in Germany

J. Lange1, A. Philipps1, A. Krumbholz1, S. Bergmann1, S. Motzke1, C. Steglich1, P. Wutzler1, M.

Groth2, S. Taudien2, M. Platzer2, R. Dürrwald3, R. Zell1

1Dept. of Virology and Antiviral Therapy, Jena University Hospital; 2Genome Analysis Group, Leipniz Institute for Age Research, Fritz Lipmann Institute (FLI),

Jena; 3IDT Biologika GmbH, Dessau-Rosslau

The emergence of pandemic (H1N1) 2009 virus evidenced that pigs play a special role in the

epidemiology of influenza viruses because they are “mixing vessels” for the reassortment of

gene segments from human, avian and swine influenza virus strains. The segments M and

NA of the H1N1v virus belong to the European lineage. This example demonstrates the

importance of swine influenza surveillance and genetic characterization of virus isolates.

Currently, more than 200 porcine influenza A viruses from a unique collection of 450 German

virus strains have been characterized with conventional and next generation sequencing

(NGS) techniques. The majority of these isolates were collected in the ongoing swine

influenza surveillance of the IDT Biologika since 2003. Sequencing of 60-80 influenza virus

strains per year requires a sequencing capacity of roughly one megabase which necessitates

a NGS sequencer. Two NGS instruments were tested, the FLX genome sequencer of Roche

and the GAII of Illumina. After test runs, the Illumina technique is better suited for our

purpose than the 454 pyrosequencing method. The Illumina technology yielded up to 70

million reads per run with a length of 50 nt and up to 47 million reads that could be assigned

to indexed virus isolates. After optimization, the number of reads mappable to specific virus

isolates increased to 550000-2.1 million. The Illumina genome analyzer GAII allows the

multiplex sequencing of up to 30 virus genomes per lane.

Phylogenetic analysis of these isolates enables to reconstruct the evolution of the European

lineage of swine influenza viruses since their emergence in 1979. There is evidence of

reassortments, antigenetic drift and accidental transmission of human seasonal and

pandemic strains.

Poster presentations

Page 26 of 49

Individual FluResearchNet project no. 11 | Poster no. 107

Principal investigator: Dr. M. Matrosovich

Characterization of the hemadsorption activity of the neuraminidase of H1N1

swine influenza viruses

V. Czudai-Matwich,

J. Uhlendorff, V. Laukemper, T. Matrosovich, H.-D. Klenk, M. Matrosovich

Institute of Virology, Philipps University, Marburg

The neuraminidases (NA) of avian influenza A viruses contain, in addition to the catalytic

site, a second sialic acid-binding site, which displays hemadsorption (HAD) activity. By

contrast, the HAD activity is lost by human influenza viruses soon after their emergence from

the avian precursors. To study evolution of the hemadsorption site in swine virus NAs and to

access its potential role in avian-to-swine and swine-to-human transmission, we compared

HAD activity of phylogenetically related NAs of i) H1N1 avian viruses, ii) their descendant

swine viruses of the Eurasian avian-like lineage and iii) the swine-origin H1N1/09 pandemic

influenza viruses. Our results suggest, that the NAs of swine viruses lost their HAD activity

several years after the virus transmission from birds to pigs and long before the emergence

of the H1N1/09 pandemic viruses, which also lack HAD activity. To specify the time of

disappearance of the HAD activity of the NA of Eurasian avian-like swine viruses, we

performed phylogenetic analysis and identified mutations in the NA which could be

responsible for the HAD-negative phenotype. Study is in progress on phenotypic

characterization of these mutations and the results will be presented.

Poster Viewing 3

Workshop topics:

Innate Immune Response

Antiviral Therapy and Resistance

Viral Pathogenesis

Friday, March 16, 2012 Venue: Poster area Time: 05.30 pm – 07.00 pm

Poster presentations

Page 28 of 49

Workshop topic: Innate Immune Response

Individual FluResearchNet project no. 6 | Poster no. 329

Principal investigator: Prof. Dr. S. Ludwig

Biphasic action of p38 MAPK signaling in the influenza A virus induced primary

and secondary host gene response

Börgeling Yvonne1, Schmolke Mirco1,2, Viemann Dorothee3,4, Roth Johannes3, Ludwig

Stephan

These observations show, that p38 acts on two levels of the antiviral IFN response: Initially

the kinase regulates IFN induction and later p38 controls IFN signaling and thereby

expression of IFN-stimulated genes. Thus, inhibition of p38 maybe an antiviral strategy that

protects mice from lethal influenza via suppression of overshooting cytokine expression.

1

1Institute of Molecular Virology, Center for Molecular Biology of Inflammation, University of

Münster, Von-Esmarch-Str. 56, 48149 Münster 2present address: Mount Sinai School of Medicine, Department of Microbiology, New York 3Institute of Immunology, University of Münster, Röntgenstr. 21, 48149 Münster 4present address: Department of Pediatric Pulmonology, Allergology and Neonatology,

MH Hannover

One characteristic of infections with highly pathogenic avian influenza viruses (HPAIV) is the

cytokine burst that strongly contributes to viral pathogenicity. It has been suggested, that

this hypercytokinemia is an intrinsic feature of infected cells and involves hyperinduction of

p38 MAP kinase. Here we investigate the role of p38 signaling in the antiviral response in

endothelial cells, a primary source for cytokines during systemic infections.

Global gene expression profiling of HPAIV infected endothelial and epithelial cells in presence

of the p38 inhibitor SB202190 revealed, that inhibition of p38 leads to reduced expression of

interferons (IFN) and other cytokines after H5N1 and H7N7 infection. Furthermore, the

expression of IFN stimulated genes (ISGs) after treatment with IFN or conditioned media

from infected cells was decreased when p38 was inhibited in target cells by pretreatment

with SB202190 or expression of dominant negative MKK6. Finally, promoter analysis

confirmed a direct impact of p38 on the IFN- and ISG-promoter activity. In vivo inhibition of

p38 leads to a nearly complete shutdown of virus induced cytokine expression concomitant

with reduced viral titers, thereby protecting mice from lethal infection.

Poster presentations

Page 29 of 49

Individual FluResearchNet project no. 7 | Poster no. 337

Principal investigator: Prof. Dr. K. Schughart

Complex genetics and transcriptome analysis of the host response to influenza A

infections

Claudia Pommerenke, Esther Wilk, Tatiana Nedelko, Heike Kollmus, and Klaus Schughart.

Department of Infection Genetics, Helmholtz Centre for Infection Research & University of

Veterinary Medicine Hannover, Braunschweig, Germany.

To identify genetic determinants of host susceptibility to infectious diseases, our laboratory is

investigating the host response after infection with influenza A virus in various mouse

genetic reference populations, including inbred and BXD recombinant inbred strains. One of

the highly susceptible strains, DBA/2J, succumbed to influenza virus at early times after

infection whereas C57BL/6J mice were more resistant and survived. Subsequently, we

characterized viral replication and the immune responses in these two strains. In addition,

we mapped several QTLs which contribute to susceptibility and resistance using BXD

recombinant inbred strains. Interestingly, both C57BL/6J and DBA/2J genomic regions

increased resistance to influenza infections. In addition, we performed a time-series of global

expression studies in mouse lungs after influenza infections for up to 60 days. Our results

showed that the different phases of the host defense were clearly discernible in the changing

transcriptome profiles: innate immune response, infiltration of T cells and formation of

bronchus-associated lymphoid tissue.

Poster presentations

Page 30 of 49

Individual FluResearchNet project no. 7 | Poster no. 342

Principal investigator: Prof. Dr. K. Schughart

Transcriptome analysis of whole lungs area valuable tool to analyze host

responses during the course of an influenza A virus infection

Esther Wilk, Claudia Pommerenke, Barkha Srivastava, Annika Schulze, Klaus Schughart

Department of Infection Genetics, Helmholtz Centre for Infection Research,

Braunschweigand University of Veterinary Medicine, Hannover

Murine models enable us to study systematically the infection with influenza A and its

consequences for the host.We performed a comprehensive systematic description of all

aspects of the host response within one experimental setting.For these studies, we infected

C57BL/6J mice with the H1N1 PR8 virus and analyzed the transcriptome of whole lungs from

days 1 to 60 after infection using Agilent´s mouse 4x44k microarrays. Our analyzes revealed

that various pathophysiological host responses, like the activation of the innate immune

response including e.g. RIG-I pathway, cytokines and chemokines and NK cells and the

switch to the adaptive immunity is well reflected in the kinetics of gene expression patterns

and were confirmed by flow cytometry and histology. In addition our results demonstrated

that the host response is not finalized 60 days after infection. These results will not only

provide a valuable basis for a systems biology modeling of the normal course of an infection

but also to allow to unravel important alterations of the host response in mutant mice or

deleterious host responses to highly virulent virus subtypes.

Poster presentations

Page 31 of 49

Individual FluResearchNet project no. 8 | Poster no. 330

Principal investigator: Prof. Dr. P. Stäheli

The role of chicken interferon lambda in antiviral defense

A. Reuter1,4, D. Rubbenstroth1, C. Winter2, S. Härtle3, P. Stäheli1

1Department of Virology, University of Freiburg, Freiburg 2Institute for Virology, University of Veterinary Medicine Hannover, Hannover 3Department of Veterinary Sciences, University of Munich, Munich 4International Max Planck Research School for Molecular and Cellular Biology, Freiburg

Interferon-lambda (IFN-λ) contributes to virus resistance of mammals by inducing interferon-

stimulated genes in epithelial cells of mucosal surfaces. Although there is redundancy in the

function of type I and type III interferons, a unique role for IFN-λ in the defense of rotavirus

in the gut of mice has recently been shown. In contrast, little is known about the antiviral

activity of IFN-λ in avian species. In this study we used a retroviral vector to constitutively

express chicken IFN-λ (chIFN-λ) in cultured chicken cells and whole chicken embryos. In

mosaic-transgenic chicken embryos expressing chIFN-λ prominent induction of Mx, an ISG

frequently used as marker of type I and III IFN responsiveness, was detected in tissues

containing large numbers of epithelial cells, such as intestine, lung, trachea and kidney.

Retroviral vector-mediated expression of chIFN-λ strongly reduced viral replication after in

ovo infection of chicken embryos with various influenza A virus strains, Newcastle disease

virus or infectious bronchitis virus. Enhanced virus resistance was also observed in primary

kidney cells and tracheal organ cultures from chicken embryos mosaic-transgenic for chIFN-

λ, but not in chIFN-λ-expressing chicken fibroblasts. These results suggest that, similar to its

mammalian counterpart, chIFN-λ confers antiviral protection mainly to epithelial cells in

chickens. Interestingly, chicks mosaic-transgenic for chIFN-λ hatched less efficiently than

control animals and died within the first week after hatching for yet unclear reasons.

Poster presentations

Page 32 of 49

Individual FluResearchNet project no. 8 | Poster no. 357

Principal investigator: Prof. Dr. P. Stäheli

Identification and preliminary characterization of IFNα-producing cells in virus-

infected chickens

S. Bender1, B. Meyer1, S. Härtle2, B. Kaspers2, P. Staeheli1, D. Rubbenstroth1

1Department of Virology, University of Freiburg, Freiburg 2Department of Veterinary Sciences, University of Munich, Munich

Little is known about the innate antiviral immune response of domestic chickens. How

chickens are able to produce high levels of type I interferon (IFN) in response to infection

with viruses that carry inhibitors of IFN production, such as the non-structural protein 1

(NS1) of influenza A virus, is a question of particular interest. Therefore, the aim of this

study was to identify and characterize cells in tissues of chickens which produce large

amounts of IFNα in response to viral infection. Ex vivo stimulation of primary chicken

splenocytes with Newcastle disease virus or a toll-like-receptor 7 (TLR-7) agonist resulted in

high levels of type I IFN in the culture supernatant. Using a monoclonal anti-chicken-IFNα

antibody and flowcytometric intracellular cytokine staining we were able to detect a small

population of IFN-producing cells in stimulated cultures. Further characterization showed that

the IFN-producing cells were positive for CD45 and MHC-II. Small numbers of chIFNα-

producing cells were also detected in the spleen of chickens infected with the highly

pathogenic avian influenza virus (HPAIV) strain A/Cygnus cygnus/Germany/R65/2006 (H5N1)

or treated with a TLR-7 agonist. Immunohistochemical analysis revealed that the chIFNα-

producing cells in HPAIV-infected spleens were negative for viral antigen but usually located

in close proximity to infected cells. Our data indicate that a small population of immune cells

is responsible for the production of high amounts of chIFNα in virus-infected chickens and

may represent specialized producers of type I IFNs.

Poster presentations

Page 33 of 49

Workshop topic: Antiviral Therapy and Resistance

Individual FluResearchNet project no. 6 | Poster no. 394

Principal investigator: Prof. Dr. O. Planz

Antiviral activity of different MEK-inhibitors against pandemic H1N1v and highly

pathogenic avian influenza virus in vitro and in vivo

Emanuel Haasbacha, Karoline Droebnerb, Carmen Müllera, Stephan Pleschkac, Stephan

Ludwigd and Oliver Planza,

aUniversity of Tuebingen, Interfaculty Institute for Cell Biology, Department of Immunology,

Tuebingen bUniversity Witten/Herdecke cInstitute for Medical Virology, Justus-Liebig-University Giessen dInstitute of Molecular Virology (IMV), University of Muenster

The emergence of the 2009 H1N1 pandemic swine influenza A virus is a good example of

how this viral infection can impact health systems around the world in a very short time. The

continuous zoonotic circulation and reassortment potential of influenza A viruses (IAV) in

nature represents an enormous public health threat to humans. Beside vaccination antivirals

are needed to efficiently control spreading of the disease. In the present work we

investigated whether different MEK-inhibitors (U0126 PD184352 and PD0325901), targeting

the intracellular Raf/MEK/ERK signaling pathway, are able to suppress propagation of the

2009 pandemic H1N1 as well as highly pathogenic avian influenza viruses (HPAIV) in cell

culture and also in vivo in the mouse lung. MEK-inhibitors showed antiviral activity in cell

culture against all tested IAV strains including oseltamivir resistant variants. Furthermore, we

were able to demonstrate that treatment of mice with MEK-inhibitors via the aerosol route

led to (i) inhibition of MEK-activation in the lung (ii) reduction of progeny IAV titers

compared to untreated controls (iii) protection of IAV infected mice against a 100% lethal

viral challenge. Moreover, no adverse effects of U0126, PD184352 and PD0325901 were

found in cell culture or in the mouse. Thus, we conclude that U0126, PD184352 and

PD0325901, by inhibiting the cellular target MEK, have antiviral potential in cell culture and

in the mouse model.

Poster presentations

Page 34 of 49

Individual FluResearchNet project no. 7 | Poster no. 414

Principal investigator: Prof. Dr. K. Schughart

Host proteases and influenza infection: characterization and inhibition

Mahmoud M. Bahgat and Klaus Schughart

Department of Infection Genetics, the Helmholtz Center for Infection Research, and the

University of Veterinary Medicine Hannover, Inhoffenstrasse 7, 38124 Braunschweig

Question

Here we investigated the nature of involved proteases in influenza infection and the capacity

of specific protease inhibitors to interfere with the infection.

Methods

DBA/2J as a highly susceptible and C57Bl/6J as a more resistant inbred mouse strains to

influenza infection were studied. The serine proteases from mice lung homogenates were

investigated by zymography and using the peptide-mimetic substrate Bz-Val-Gly-Arg-p-

nitroanilide. The transcripts encoding the serine proteases were quantified by real-time RT-

PCR. The effects of the specific inhibitors AEBSF and pAB on the measured activities were

investigated. The same inhibitors were used to interfere with the infection of the influenza

A/Puerto Rico/8/34 (H1N1; PR8) and the A/Seal/Massachusetts/1/80 (H7N7; SC35M) viruses

in lung cell lines and in mice.

Results

Up-regulation in the measured serine protease activities was recorded post infection in

comparison to non-infected mice. zymography results indicated up-regulation of several

proteolytically active peptides of a broad range of molecular weights after influenza infection

suggesting involvement of more than one enzyme that was confirmed at the transcriptional

level by real-time RT-PCR. The inhibitory effects of AEBSF and pAB confirmed the serine

protease nature of the quantified activities. Individual and cocktailed AEBSF and pAB blocked

the H1N1-PR8 and the H7N7-SC35M propagation in lung cell culture and in mice.

Conclusions

Multiple serine protease activities are implicated in mediating influenza A infection. Blocking

influenza A virus infection in cultured lung cells and in mice by serine protease inhibitors may

provide an alternative approach for influenza therapy.

Poster presentations

Page 35 of 49

Individual FluResearchNet project no. 10 | Poster no. 409

Principal investigator: PD Dr. M. Schmidtke

Characterization of the oseltamivir susceptibility of the pandemic H1N1 influenza

virus A/Jena/5258/09 in vitro and in vivo

N. Seidel, H. Braun, M. Richter, A. Krumbholz, P. Wutzler, M. Schmidtke

Jena University Hospital, Department of Virology and Antiviral Therapy, Jena

Since the emergence in 2009, the pandemic H1N1 influenza A viruses (H1N1v) became the

dominant seasonal influenza subtype. Because of their genetic background H1N1v are

adamantane resistant and neuraminidase inhibitor (NAI) susceptible. Therefore, only the

latter drugs can be used as positive control compounds in in vitro and in vivo experiments.

In the present study, the oseltamivir susceptibility of the isolate A/Jena/5258/09 was

investigated.

First, the isolate was once passaged in BALB/c mice. Lung homogenates were used to

produce a virus stock in MDCK cells. A chemiluminescence-based enzyme inhibition assay

and a virus yield reduction assay revealed that the original and the mouse-passaged isolate

(mp-Jena/5258) are NAI susceptible in vitro. Sequence analysis demonstrates that the NA

genes of both isolates are identical and possess no known resistance mutation.

Subsequently, BALB/c mice were intranasally infected with 105 or 106 TCID50 of mp-

Jena/5258. Oseltamivir was administered orally at 10 mg/kg/d from day 0 to 4, twice daily.

Survival, body weight and general condition were monitored over 21 days. To determine

lung parameters (weight, score, viral load) 5 animals per group were necropsied on day 4

and 21 p.i. The virus caused in placebo-treated as well as in oseltamivir-treated mice a

severe disease with strong body weight loss, impairment of general condition, and lung

histopathology. Moreover, mortality of animals infected with 105 and 106 TCID50 was ~40%

and 90%, respectively, regardless of treatment.

These results indicate that higher oseltamivir concentrations are necessary for an effective

treatment of H1N1v in mice.

Poster presentations

Page 36 of 49

Workshop topic: Viral Pathogenesis

Individual FluResearchNet project no. 6 | Poster no. 575

Principal investigator: Prof. Dr. S. Ludwig

Three point mutations render the pandemic 2009 H1N1 virus lethal for mice

R. Seyer1, E.R. Hrincius1, D. Ritzel2, M. Abt2, +, H. Marjuki3, J. Kühn4, T. Wolff2, S. Ludwig1,

C. Ehrhardt1

1Institute of Molecular Virology, Center for Molecular Biology of Inflammation, University of Münster,

Von Esmarch-Str. 56, 48149 Muenster; 2Robert Koch Institute, FG17, Nordufer 20, 13353 Berlin; +present address: Institute for Medical Microbiology and Hygiene; University Medical Centre

Mannheim; Theodor-Kutzer-Ufer 1-3, 68167 Mannheim; 3Division of Virology, Department of

Infectious Diseases, St Jude Children`s Research Hospital, Memphis, TN 38105; 4Institute of Medical

Microbiology, University of Münster, Von Stauffenberg Strasse 36, 48151 Muenster

Influenza impressively reflects the paradigm of a viral disease in which continued evolution

of the virus is of paramount importance for annual epidemics and occasional pandemics in

humans. The pandemic outbreak of the H1N1variant (H1N1v) virus in 2009 caused relatively

mild symptoms in the majority of patients. However, the high mutation rate of influenza

viruses may facilitate to the emergence of future H1N1v strains that exhibit higher

pathogenicity and cause more severe outbreaks of respiratory disease. Because of the

continuous threat of novel influenza outbreaks it appears important to gather further

knowledge about viral pathogenicity determinants. Here, we explored the adaptive potential

of the H1N1v isolate A/Hamburg/04/09 (HH/04) by sequential passaging in mice lungs.

Three passages in mice lungs were sufficient to dramatically enhance pathogenicity of

HH/04, as evidenced by increased mortality in mice. Sequence analysis identified four

nonsynonymous mutations in the third passage virus. Using reverse genetics, three

synergistically acting mutations were defined as pathogenicity determinants, comprising two

mutations in the hemagglutinin (D222G and K163E), whereby the HA(D222G) mutation was

shown to determine receptor binding specificity, and the PA(F35L) mutation increasing

polymerase activity. In conclusion, synergistically action of all three mutations results in a

mice lethal pandemic H1N1v virus. Our results highlight the potential of H1N1v to rapidly

adapt to a new mammalian host within a few passages, clearly indicating the potential for

the emergence of highly pathogenic H1N1v variants.

Poster presentations

Page 37 of 49

Individual FluResearchNet project no. 6 | Poster no. 580

Principal investigator: Prof. Dr. S. Ludwig

A single point mutation (Y89F) within the non-structural protein 1 of influenza A

viruses limits epithelial cell tropism and virulence in mice

Eike Roman Hrincius1, Ann-Katrin Hennecke1, Lisa Gensler1, Darisuren Anhlan1, Peter Vogel2,

Jonathan A. McCullers3, Stephan Ludwig1 and Christina Ehrhardt1

1Institute of Molecular Virology, Center for Molecular Biology of Inflammation, University of

Münster, Von Esmarch-Str. 56, 48149 Muenster 2Veterinary Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place,

Memphis, TN 38105-3678 3Department of Infectious Diseases, St. Jude Children's Research Hospital, 262 Danny

Thomas Place, Memphis, TN 38105-3678

The non-structural protein 1 (A/NS1) of influenza A viruses (IAV) harbors several src

homology domain (SH)-binding motifs (bm) (one SH2bm and two SH3bm), which mediate

interaction with cellular proteins. In contrast to the sequence variability of the second

SH3bm, the tyrosine 89 within the SH2bm is highly conserved among different IAV strains.

This prompted us to evaluate the necessity of this SH2bm for IAV virulence. In an in vivo

mouse model, we observed a dramatically reduced mortality upon infection with the A/NS1

Y89F mutant in comparison to wild-type virus. Infectious titers in the lung and

bronchoalveolar-lavage fluid (BALF) were also reduced in comparison to wild-type virus.

Concomitantly, we observed decreased inflammation as well as less severe pathological

changes, reflecting reduced levels of virus-titers. Interestingly the replication of the A/NS1

mutant in mouse lung was overall reduced and strongly restricted to alveoli. In contrast,

wild-type virus infection led to virus antigen positive areas in tracheal, bronchus, bronchiole

and alveolar epithelium. Finally, wild-type virus infection resulted in a dramatic destruction of

the bronchiole epithelium in clear contrast to infection with the A/NS1 mutant.

Taken together, we could show that disruption of the highly conserved SH2bm within the

A/NS1 results in decreased virus distribution in the mouse lung and dramatically reduces

virulence illustrating the necessity of the SH2bm for IAV induced pathogenicity.

Poster presentations

Page 38 of 49

PUBLICATIONS

2011 - 2012

Publications

Page 41 of 49

Please consider that the publication list summarizes FluResearchNet publications

as well as influenza associated publications of FluResearchNet members.

Abt M, de Jonge J, Laue M, Wolff T. Improvement of H5N1 influenza vaccine viruses: influence of internal gene segments of avian and human origin on production and hemagglutinin content. (2011). Vaccine. 29(32):5153-62.

Anhlan D, Grundmann N, Makalowski W, Ludwig S, Scholtissek C. Origin of the 1918 pandemic H1N1 influenza A virus as studied by codon usage patterns and phylogenetic analysis. (2011). RNA 17(1):64-73.

Bahgat MM, Błazejewska P, Schughart K. Inhibition of lung serine proteases in mice: a potentially new approach to control influenza infection. (2011). Virol J. 8:27.

Bauer K, Dürrwald R, Schlegel M, Pfarr K, Topf D, Wiesener N, Dahse HM, Wutzler P, Schmidtke M. Neuraminidase inhibitor susceptibility of swine influenza A viruses isolated in Germany between 1981 and 2008. (2012). Med Microbiol Immunol. 201(1):61-72.

Bel M, Ocaña-Macchi M, Liniger M, McCullough KC, Matrosovich M, Summerfield A. Efficient sensing of avian influenza viruses by porcine plasmacytoid dendritic cells. (2011). Viruses. 3(4):312-30.

Blazejewska P, Koscinski L, Viegas N, Anhlan D, Ludwig S, Schughart K. Pathogenicity of different PR8 influenza A virus variants in mice is determined by both viral and host factors. (2011). Virology. 412(1):36-45.

Bogs J, Kalthoff D, Veits J, Pavlova S, Schwemmle M, Mänz B, Mettenleiter TC, Stech J. Reversion of PB2-627E to -627K during replication of an H5N1 Clade 2.2 virus in mammalian hosts depends on the origin of the nucleoprotein. (2011). J Virol. 85(20):10691-8.

Bortz E, Westera L, Maamary J, Steel J, Albrecht RA, Manicassamy B, Chase G, Martínez-Sobrido L, Schwemmle M, García-Sastre A. Host- and strain-specific regulation of influenza virus polymerase activity by interacting cellular proteins. (2011). MBio. 16;2(4). pii: e00151-11. doi: 10.1128/mBio.00151-11.

Chase G, Wunderlich K, Reuther P, Schwemmle M. Identification of influenza virus inhibitors which disrupt of viral polymerase protein-protein interactions. (2011). Methods. 55(2):188-91.

Chase GP, Rameix-Welti MA, Zvirbliene A, Zvirblis G, Götz V, Wolff T, Naffakh N, Schwemmle M. Influenza Virus Ribonucleoprotein Complexes Gain Preferential Access to Cellular Export Machinery through Chromatin Targeting. (2011). PLoS Pathog. 7(9):e1002187. Epub 2011 Sep 1.

Demirov D, Gabriel G, Schneider C, Hohenberg H, Ludwig S. Interaction of influenza A virus matrix protein with RACK1 is required for virus release. (2012). Cell Microbiol. Jan 31. doi: 10.1111/j.1462-5822.2012.01759.x. [Epub ahead of print]

Deng Q, Wang D, Xiang X, Gao X, Hardwidge PR, Kaushik R, Wolff T, Chakravarty S, Li F. Nuclear localization of influenza B polymerase proteins and their binary complexes. (2011). Virus Res. 156(1-2):49-53.

Publications

Page 42 of 49

Deng Q, Wang D, Xiang X, Gao X, Hardwidge PR, Kaushik RS, Wolff T, Chakravarty S, Li F. Application of a split luciferase complementation assay for the detection of viral protein-protein interactions. (2011). J Virol Methods. 176(1-2):108-11.

Dimitrakopoulou K, Tsimpouris C, Papadopoulos G, Pommerenke C, Wilk E, Sgarbas KN, Schughart K, Bezerianos A. Dynamic gene network reconstruction from gene expression data in mice after influenza A (H1N1) infection. (2011). J Clin Bioinformatics. 1:27 doi:10.1186/2043-9113-1-27

Droebner K, Pleschka S, Ludwig S, Planz O. Antiviral activity of the MEK-inhibitor U0126 against pandemic H1N1v and highly pathogenic avian influenza virus in vitro and in vivo. (2011). Antiviral Res. 92(2):195-203

Dudek SE, Wixler L, Nordhoff C, Nordmann A, Anhlan D, Wixler V, Ludwig S. The influenza virus PB1-F2 protein has interferon antagonistic activity. (2011). Biol Chem. 392(12):1135-44.

Gambaryan AS, Matrosovich TY, Philipp J, Munster VJ, Fouchier RA, Cattoli G, Capua I, Krauss SL, Webster RG, Banks J, Bovin NV, Klenk HD, Matrosovich MN. Receptor-binding Profiles of H7 Subtype Influenza Viruses in Different Host Species. (2012). J Virol. Febr 15 [Epub ahead of print]

Gao S, von der Malsburg A, Dick A, Faelber K, Schröder GF, Haller O, Kochs G, Daumke O. Structure of myxovirus resistance protein a reveals intra- and intermolecular domain interactions required for the antiviral function. (2011). Immunity. 35(4):514-25.

Ge X, Rameix-Welti MA, Gault E, Chase G, dos Santos Afonso E, Picard D, Schwemmle M, Naffakh N. Influenza virus infection induces the nuclear relocalization of the Hsp90 co-chaperone p23 and inhibits the glucocorticoid receptor response. (2011). PLoS One. 6(8):e23368. Epub Aug 10.

Grienke U, Schmidtke M, von Grafenstein S, Kirchmair J, Liedl KR, Rollinger JM. Influenza neuraminidase: a druggable target for natural products. (2012) Nat Prod Rep

Haasbach E, Pauli EK, Spranger R, Mitzner D, Schubert U, Kircheis R, Planz O.

. 29(1):11-36. Review.

Haasbach E, Droebner K, Vogel AB, Planz O. Low-Dose Interferon Type I Treatment Is Effective Against H5N1 and Swine-Origin H1N1 Influenza A Viruses In Vitro and In Vivo. (2011). J Interferon Cytokine Res. 31(6):515-25.

Antiviral activity of the proteasome inhibitor VL-01 against influenza A viruses. (2011). Antiviral Res. 91(3):304-13.

Herold S, Ludwig S, Pleschka S, Wolff T. Apoptosis signaling in influenza virus propagation, innate host defense, and lung injury. (2012). J Leukoc Biol. Feb 17. [Epub ahead of print]

Herold S. Pathogenesis, symptoms and treatment of virus influenza. (2011). Pharm Unserer Zeit. 40(2):115-9. Review. German.

Hrincius ER, Dierkes R, Anhlan D, Wixler V, Ludwig S, Ehrhardt C. Phosphatidylinositol-3-kinase (PI3K) is activated by influenza virus vRNA via the pathogen pattern receptor Rig-I to promote efficient type I interferon production. (2011). Cell Microbiol. 13(12):1907-19.

Publications

Page 43 of 49

Hundt B, Mölle N, Stefaniak S, Dürrwald R, Weyand J. Large pilot scale cultivation process study of adherent MDBK cells for porcine Influenza A virus propagation using a novel disposable stirred-tank bioreactor. (2011). BMC Proc. 5 Suppl 8:P128.

Kaminski MM, Ohnemus A, Cornitescu M, Staeheli P. Plasmacytoid dendritic cells and Toll-like receptor 7-dependent signalling promote efficient protection of mice against highly virulent influenza A virus. (2012). J Gen Virol

Kirchmair J, Rollinger JM, Liedl KR, Seidel N, Krumbholz A, Schmidtke M.

. 93(Pt 3):555-9.

Novel neuraminidase inhibitors: identification, biological evaluation and investigations of the binding mode. (2011). Future Med Chem. 3(4):437-50.

Klenk HD, Garten W, Matrosovich M. Molecular mechanisms of interspecies transmission and pathogenicity of influenza viruses: Lessons from the 2009 pandemic. (2011). Bioessays. 33(3):180-8.

Koerner I, Matrosovich MN, Haller O, Staeheli P, Kochs G. Altered receptor specificity and fusion activity of the hemagglutinin contribute to high virulence of a mouse-adapted influenza A virus. (2012). J Gen Virol. Jan 18. [Epub ahead of print].

Krawitz C, Abu Mraheil M, Stein M, Imirzalioglu C, Domann E, Pleschka S, Hain T. Inhibitory activity of a standardized elderberry liquid extract against clinically-relevant human respiratory bacterial pathogens and influenza A and B viruses. (2011). BMC Complement Altern Med. 11(1):16.

Liu S, Li R, Zhang R, Chan CC, Xi B, Zhu Z, Yang J, Poon VK, Zhou J, Chen M, Münch J, Kirchhoff F, Pleschka S, Haarmann T, Dietrich U, Pan C, Du L, Jiang S, Zheng B. CL-385319 inhibits H5N1 avian influenza A virus infection by blocking viral entry. (2011). Eur J Pharmacol. 660(2-3):460-7.

Ludwig S. Disruption of virus-host cell interactions and cell signaling pathways as an anti-viral approach against influenza virus infections. (2011). Biol Chem. 392(10):837-47.

Mänz B, Götz V, Wunderlich K, Eisel J, Kirchmair J, Stech J, Stech O, Chase G, Frank R, Schwemmle M. Disruption of the viral polymerase complex assembly as a novel approach to attenuate influenza a virus. (2011). J Biol Chem. 286(10):8414-24.

Muhammad S, Haasbach E, Kotchourko M, Strigli A, Krenz A, Ridder DA, Vogel AB, Marti HH, Al-Abed Y, Planz O, Schwaninger M. Influenza virus infection aggravates stroke outcome. (2011). Stroke. 42(3):783-91.

Nordmann A, Wixler L, Boergeling Y, Wixler V, Ludwig S. A new splice variant of the human guanylate-binding protein 3 mediates anti-influenza activity through inhibition of viral transcription and replication. (2011). FASEB J. Nov 21. [Epub ahead of print].

Ott U, Sauerbrei A, Lange J, Schäfler A, Walther M, Wolf G, Wutzler P, Zell R, Krumbholz A. Serological response to influenza A H1N1 vaccine (Pandemrix(®)) and seasonal influenza vaccine 2009/2010 in renal transplant recipients and in hemodialysis patients. (2012). Med Microbiol Immunol. Feb 17. [Epub ahead of print]

Penski N, Härtle S, Rubbenstroth D, Krohmann C, Ruggli N, Schusser B, Pfann M, Reuter A, Gohrbandt S, Hundt J, Veits J, Breithaupt A, Kochs G, Stech J, Summerfield A, Vahlenkamp T, Kaspers B, Staeheli P. Highly pathogenic avian influenza viruses do not inhibit interferon synthesis in infected chickens but can override the interferon-induced antiviral state. (2011). J Virol. 85(15):7730-41.

Publications

Page 44 of 49

Pinto R, Herold S, Cakarova L, Hoegner K, Lohmeyer J, Planz O, Pleschka S. Inhibition of influenza virus-induced NF-kappaB and Raf-MEK-ERK activation can reduce both virus titers and cytokine expression simultaneously in vitro and in vivo. (2011). Antiviral Res. 92(1):45-56.

Punyadarsaniya D, Liang CH, Winter C, Petersen H, Rautenschlein S, Hennig-Pauka I, Schwegmann-Wessels C, Wu CY, Wong CH, Herrler G. Infection of differentiated porcine airway epithelial cells by influenza virus: differential susceptibility to infection by porcine and avian viruses. (2011). PLoS One. 6(12):e28429. Epub Dec 9.

Reuther P, Mänz B, Brunotte L, Schwemmle M, Wunderlich K. Targeting of the influenza A virus polymerase PB1-PB2 interface indicates strain-specific assembly differences.(2011). J Virol. 85(24):13298-309. Epub Sep 28.

Robb NC, Chase G, Bier K, Vreede FT, Shaw PC, Naffakh N, Schwemmle M, Fodor E. The influenza A virus NS1 protein interacts with the nucleoprotein of viral ribonucleoprotein complexes. (2011). J Virol. 85(10):5228-31.

Schusser B, Reuter A, von der Malsburg A, Penski N, Weigend S, Kaspers B, Staeheli P, Härtle S. Mx is dispensable for interferon-mediated resistance of chicken cells against influenza A virus. (2011). J Virol. 85(16):8307-15.

Seliger C, Schaerer B, Kohn M, Pendl H, Weigend S, Kaspers B, Härtle S. A rapid high-precision flow cytometry based technique for total white blood cell counting in chickens. (2012). Vet Immunol Immunopathol. 145(1-2):86-99.

Seyer R, Hrincius ER, Ritzel D, Abt M, Mellmann A, Marjuki H, Kühn J, Wolff T, Ludwig S, Ehrhardt C. Synergistic adaptive mutations in the hemagglutinin and polymerase acidic protein lead to increased virulence of pandemic 2009 H1N1 influenza A virus in mice. (2012). J Infect Dis. 205(2):262-71.

Sipo I, Knauf M, Fechner H, Poller W, Planz O, Kurth R, Norley S. Vaccine protection against lethal homologous and heterologous challenge using recombinant AAV vectors expressing codon-optimized genes from pandemic swine origin influenza virus (SOIV). (2011). Vaccine. 29(8):1690-9.

Stech J, Garn H, Herwig A, Stech O, Dauber B, Wolff T, Mettenleiter TC, Klenk HD. Influenza B virus with modified hemagglutinin cleavage site as a novel attenuated live vaccine. (2011). J Infect Dis. 204(10):1483-90.

Thaa B, Tielesch C, Möller L, Schmitt AO, Wolff T, Bannert N, Herrmann A, Veit M. Growth of influenza A virus is not impeded by simultaneous removal of the cholesterol-binding and acylation sites in the M2 protein. (2012). J Gen Virol. 93(Pt 2):282-92.

Viemann D, Schmolke M, Lueken A, Boergeling Y, Friesenhagen J, Wittkowski H, Ludwig S, Roth J. H5N1 virus activates signaling pathways in human endothelial cells resulting in a specific imbalanced inflammatory response. (2011). J Immunol. 186(1):164-73.

von der Malsburg A, Abutbul-Ionita I, Haller O, Kochs G, Danino D. Stalk domain of the dynamin-like MxA GTPase protein mediates membrane binding and liposome tubulation via the unstructured L4 loop. (2011). J Biol Chem. 286(43):37858-65.

Publications

Page 45 of 49

Wunderlich K, Juozapaitis M, Ranadheera C, Kessler U, Martin A, Eisel J, Beutling U, Frank R, Schwemmle M. Identification of high-affinity PB1-derived peptides with enhanced affinity to the PA protein of influenza A virus polymerase. (2011). Antimicrob Agents Chemother. 55(2):696-702.

Zhirnov OP, Matrosovich TY, Matrosovich MN, Klenk HD. Aprotinin, a protease inhibitor, suppresses proteolytic activation of pandemic H1N1v influenza virus. (2011). Antivir Chem Chemother. 21(4):169-74. Erratum in: (2011). Antivir Chem Chemother. 21(6):245.

Zimmermann P, Mänz B, Haller O, Schwemmle M, Kochs G. The viral nucleoprotein determines Mx sensitivity of influenza A viruses. (2011). J Virol. 85(16):8133-40.

Publications

Page 46 of 49

FluResearchNet

ANNOUNCEMENTS 2012

First FluResearchNet member meeting 2012

Venue 22nd Annual Meeting of the Society for Virology (GfV) HAUS DER TECHNIK

Hollestraße 1 | 45127 Essen

Room B, 6th floor (please follow the instructions in Essen)

Date and time Friday, 16th March 2012

12.00 – 01.30 pm

3rd International Influenza Meeting 2012

Registration and abstract submission are open now!

Deadline for abstract submission: June 15, 2012

Registration via www.fluresearchnet.de

International Conference Innate Immunity of the Lung – Improving Pneumonia Outcome Organized by the Transregional Collaborative Research Center SFB-TR 84 and

German Academy of Science Leopoldina.

Venue Berlin-Brandenburg Academy of Sciences

Markgrafenstraße 10 | Berlin-Gendarmenmarkt

Date September 19 – 22, 2012