Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Christine Thumann is active.

Publication


Featured researches published by Christine Thumann.


Nature Medicine | 2011

EGFR and EphA2 are host factors for hepatitis C virus entry and possible targets for antiviral therapy

Joachim Lupberger; Mirjam B. Zeisel; Fei Xiao; Christine Thumann; Isabel Fofana; Laetitia Zona; Christopher Davis; Christopher J. Mee; Marine Turek; Sebastian Gorke; Cathy Royer; Benoit Fischer; Muhammad Zahid; Dimitri Lavillette; Judith Fresquet; François-Loïc Cosset; S Michael Rothenberg; Thomas Pietschmann; Arvind H. Patel; Patrick Pessaux; Michel Doffoel; Wolfgang Raffelsberger; Olivier Poch; Jane A. McKeating; Laurent Brino; Thomas F. Baumert

Hepatitis C virus (HCV) is a major cause of liver disease, but therapeutic options are limited and there are no prevention strategies. Viral entry is the first step of infection and requires the cooperative interaction of several host cell factors. Using a functional RNAi kinase screen, we identified epidermal growth factor receptor and ephrin receptor A2 as host cofactors for HCV entry. Blocking receptor kinase activity by approved inhibitors broadly impaired infection by all major HCV genotypes and viral escape variants in cell culture and in a human liver chimeric mouse model in vivo. The identified receptor tyrosine kinases (RTKs) mediate HCV entry by regulating CD81–claudin-1 co-receptor associations and viral glycoprotein–dependent membrane fusion. These results identify RTKs as previously unknown HCV entry cofactors and show that tyrosine kinase inhibitors have substantial antiviral activity. Inhibition of RTK function may constitute a new approach for prevention and treatment of HCV infection.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Exosome-mediated transmission of hepatitis C virus between human hepatoma Huh7.5 cells

Vedashree Ramakrishnaiah; Christine Thumann; Isabel Fofana; F. Habersetzer; Qiuwei Pan; Petra E. de Ruiter; Rob Willemsen; Jeroen Demmers; Victor Stalin Raj; Guido Jenster; Jaap Kwekkeboom; Hugo W. Tilanus; Bart L. Haagmans; Thomas F. Baumert; Luc J. W. van der Laan

Recent evidence indicates there is a role for small membrane vesicles, including exosomes, as vehicles for intercellular communication. Exosomes secreted by most cell types can mediate transfer of proteins, mRNAs, and microRNAs, but their role in the transmission of infectious agents is less established. Recent studies have shown that hepatocyte-derived exosomes containing hepatitis C virus (HCV) RNA can activate innate immune cells, but the role of exosomes in the transmission of HCV between hepatocytes remains unknown. In this study, we investigated whether exosomes transfer HCV in the presence of neutralizing antibodies. Purified exosomes isolated from HCV-infected human hepatoma Huh7.5.1 cells were shown to contain full-length viral RNA, viral protein, and particles, as determined by RT-PCR, mass spectrometry, and transmission electron microscopy. Exosomes from HCV-infected cells were capable of transmitting infection to naive human hepatoma Huh7.5.1 cells and establishing a productive infection. Even with subgenomic replicons, lacking structural viral proteins, exosome-mediated transmission of HCV RNA was observed. Treatment with patient-derived IgGs showed a variable degree of neutralization of exosome-mediated infection compared with free virus. In conclusion, this study showed that hepatic exosomes can transmit productive HCV infection in vitro and are partially resistant to antibody neutralization. This discovery sheds light on neutralizing antibodies resistant to HCV transmission by exosomes as a potential immune evasion mechanism.


Hepatology | 2010

Inhibition of hepatitis C virus infection by anti-claudin-1 antibodies is mediated by neutralization of E2–CD81–Claudin-1 associations†

Sophie E. Krieger; Mirjam B. Zeisel; Christopher Davis; Christine Thumann; Helen J. Harris; Eva K. Schnober; Christopher J. Mee; Eric Soulier; Cathy Royer; Mélanie Lambotin; Fritz Grunert; Viet Loan Dao Thi; Marlène Dreux; François-Loı̈c Cosset; Jane A. McKeating; Catherine Schuster; Thomas F. Baumert

The tight junction protein claudin‐1 (CLDN1) has been shown to be essential for hepatitis C virus (HCV) entry—the first step of viral infection. Due to the lack of neutralizing anti‐CLDN1 antibodies, the role of CLDN1 in the viral entry process is poorly understood. In this study, we produced antibodies directed against the human CLDN1 extracellular loops by genetic immunization and used these antibodies to investigate the mechanistic role of CLDN1 for HCV entry in an infectious HCV cell culture system and human hepatocytes. Antibodies specific for cell surface–expressed CLDN1 specifically inhibit HCV infection in a dose‐dependent manner. Antibodies specific for CLDN1, scavenger receptor B1, and CD81 show an additive neutralizing capacity compared with either agent used alone. Kinetic studies with anti‐CLDN1 and anti‐CD81 antibodies demonstrate that HCV interactions with both entry factors occur at a similar time in the internalization process. Anti‐CLDN1 antibodies inhibit the binding of envelope glycoprotein E2 to HCV permissive cell lines in the absence of detectable CLDN1‐E2 interaction. Using fluorescent‐labeled entry factors and fluorescence resonance energy transfer methodology, we demonstrate that anti‐CLDN1 antibodies inhibit CD81‐CLDN1 association. In contrast, CLDN1‐CLDN1 and CD81‐CD81 associations were not modulated. Taken together, our results demonstrate that antibodies targeting CLDN1 neutralize HCV infectivity by reducing E2 association with the cell surface and disrupting CD81‐CLDN1 interactions. Conclusion: These results further define the function of CLDN1 in the HCV entry process and highlight new antiviral strategies targeting E2‐CD81‐CLDN1 interactions. (HEPATOLOGY 2010.)


Gastroenterology | 2010

Monoclonal Anti-Claudin 1 Antibodies Prevent Hepatitis C Virus Infection of Primary Human Hepatocytes

Isabel Fofana; Sophie E. Krieger; Fritz Grunert; Sandra Glauben; Fei Xiao; Samira Fafi–Kremer; Eric Soulier; Cathy Royer; Christine Thumann; Christopher J. Mee; Jane A. McKeating; Tatjana Dragic; Patrick Pessaux; Françoise Stoll–Keller; Catherine Schuster; John F. Thompson; Thomas F. Baumert

BACKGROUND & AIMS Hepatitis C virus (HCV) infection is a challenge to prevent and treat because of the rapid development of drug resistance and escape. Viral entry is required for initiation, spread, and maintenance of infection, making it an attractive target for antiviral strategies. The tight junction protein claudin-1 (CLDN1) has been shown to be required for entry of HCV into the cell. METHODS Using genetic immunization, we produced 6 monoclonal antibodies against the host entry factor CLDN1. The effects of antibodies on HCV infection were analyzed in human cell lines and primary human hepatocytes. RESULTS Competition and binding studies demonstrated that antibodies interacted with conformational epitopes of the first extracellular loop of CLDN1; binding of these antibodies required the motif W(30)-GLW(51)-C(54)-C(64) and residues in the N-terminal third of CLDN1. The monoclonal antibodies against CLDN1 efficiently inhibited infection by HCV of all major genotypes as well as highly variable HCV quasispecies isolated from individual patients. Furthermore, antibodies efficiently blocked cell entry of highly infectious escape variants of HCV that were resistant to neutralizing antibodies. CONCLUSIONS Monoclonal antibodies against the HCV entry factor CLDN1 might be used to prevent HCV infection, such as after liver transplantation, and might also restrain virus spread in chronically infected patients.


Journal of Virology | 2008

Scavenger Receptor Class B Is Required for Hepatitis C Virus Uptake and Cross-Presentation by Human Dendritic Cells

Heidi Barth; Eva K. Schnober; Christoph Neumann-Haefelin; Christine Thumann; Mirjam B. Zeisel; Helmut M. Diepolder; Zongyi Hu; T. Jake Liang; Hubert E. Blum; Robert Thimme; Mélanie Lambotin; Thomas F. Baumert

ABSTRACT Class B scavenger receptors (SR-Bs) bind lipoproteins and play an important role in lipid metabolism. Most recently, SR-B type I (SR-BI) and its splicing variant SR-BII have been found to mediate bacterial adhesion and cytosolic bacterial invasion in mammalian cells. In this study, we demonstrate that SR-BI is a key host factor required for hepatitis C virus (HCV) uptake and cross-presentation by human dendritic cells (DCs). Whereas monocytes and T and B cells were characterized by very low or undetectable SR-BI expression levels, human DCs demonstrated a high level of cell surface expression of SR-BI similar to that of primary human hepatocytes. Antibodies targeting the extracellular loop of SR-BI efficiently inhibited HCV-like particle binding, uptake, and cross-presentation by human DCs. Moreover, human high-density lipoprotein specifically modulated HCV-like particle binding to DCs, indicating an interplay of HCV with the lipid transfer function of SR-BI in DCs. Finally, we demonstrate that anti-SR-BI antibodies inhibit the uptake of cell culture-derived HCV (HCVcc) in DCs. In conclusion, these findings identify a novel function of SR-BI for viral antigen uptake and recognition and may have an important impact on the design of HCV vaccines and immunotherapeutic approaches aiming at the induction of efficient antiviral immune responses.


Nature Biotechnology | 2015

Clearance of persistent hepatitis C virus infection in humanized mice using a claudin-1-targeting monoclonal antibody

Laurent Mailly; Fei Xiao; Joachim Lupberger; Garrick K. Wilson; Philippe Aubert; Francois H.T. Duong; Diego Calabrese; Céline Leboeuf; Isabel Fofana; Christine Thumann; Simonetta Bandiera; M. Lütgehetmann; T. Volz; Christopher Davis; Helen J. Harris; Christopher J. Mee; Erika Girardi; Béatrice Chane-Woon-Ming; Maria Ericsson; Nicola F. Fletcher; Ralf Bartenschlager; Patrick Pessaux; Koen Vercauteren; Philip Meuleman; Pascal Villa; Lars Kaderali; Sébastien Pfeffer; Markus H. Heim; Michel Neunlist; Mirjam B. Zeisel

Hepatitis C virus (HCV) infection is a leading cause of liver cirrhosis and cancer. Cell entry of HCV and other pathogens is mediated by tight junction (TJ) proteins, but successful therapeutic targeting of TJ proteins has not been reported yet. Using a human liver–chimeric mouse model, we show that a monoclonal antibody specific for the TJ protein claudin-1 (ref. 7) eliminates chronic HCV infection without detectable toxicity. This antibody inhibits HCV entry, cell-cell transmission and virus-induced signaling events. Antibody treatment reduces the number of HCV-infected hepatocytes in vivo, highlighting the need for de novo infection by means of host entry factors to maintain chronic infection. In summary, we demonstrate that an antibody targeting a virus receptor can cure chronic viral infection and uncover TJ proteins as targets for antiviral therapy.


Gut | 2015

Synergy of entry inhibitors with direct-acting antivirals uncovers novel combinations for prevention and treatment of hepatitis C.

Fei Xiao; Isabel Fofana; Christine Thumann; Laurent Mailly; Roxane Alles; Eric Robinet; Nicolas Meyer; Mickaël Schaeffer; François Habersetzer; Michel Doffoel; Pieter Leyssen; Johan Neyts; Mirjam B. Zeisel; Thomas Baumert

Objective Although direct-acting antiviral agents (DAAs) have markedly improved the outcome of treatment in chronic HCV infection, there continues to be an unmet medical need for improved therapies in difficult-to-treat patients as well as liver graft infection. Viral entry is a promising target for antiviral therapy. Design Aiming to explore the role of entry inhibitors for future clinical development, we investigated the antiviral efficacy and toxicity of entry inhibitors in combination with DAAs or other host-targeting agents (HTAs). Screening a large series of combinations of entry inhibitors with DAAs or other HTAs, we uncovered novel combinations of antivirals for prevention and treatment of HCV infection. Results Combinations of DAAs or HTAs and entry inhibitors including CD81-, scavenger receptor class B type I (SR-BI)- or claudin-1 (CLDN1)-specific antibodies or small-molecule inhibitors erlotinib and dasatinib were characterised by a marked and synergistic inhibition of HCV infection over a broad range of concentrations with undetectable toxicity in experimental designs for prevention and treatment both in cell culture models and in human liver-chimeric uPA/SCID mice. Conclusions Our results provide a rationale for the development of antiviral strategies combining entry inhibitors with DAAs or HTAs by taking advantage of synergy. The uncovered combinations provide perspectives for efficient strategies to prevent liver graft infection and novel interferon-free regimens.


PLOS ONE | 2013

A novel monoclonal anti-CD81 antibody produced by genetic immunization efficiently inhibits Hepatitis C virus cell-cell transmission.

Isabel Fofana; Fei Xiao; Christine Thumann; Marine Turek; Laetitia Zona; Rajiv G. Tawar; Fritz Grunert; John F. Thompson; Mirjam B. Zeisel; Thomas F. Baumert

Background and Aims Hepatitis C virus (HCV) infection is a challenge to prevent and treat because of the rapid development of drug resistance and escape. Viral entry is required for initiation, spread, and maintenance of infection, making it an attractive target for antiviral strategies. Methods Using genetic immunization, we produced four monoclonal antibodies (mAbs) against the HCV host entry factor CD81. The effects of antibodies on inhibition of HCV infection and dissemination were analyzed in HCV permissive human liver cell lines. Results The anti-CD81 mAbs efficiently inhibited infection by HCV of different genotypes as well as a HCV escape variant selected during liver transplantation and re-infecting the liver graft. Kinetic studies indicated that anti-CD81 mAbs target a post-binding step during HCV entry. In addition to inhibiting cell-free HCV infection, one antibody was also able to block neutralizing antibody-resistant HCV cell-cell transmission and viral dissemination without displaying any detectable toxicity. Conclusion A novel anti-CD81 mAb generated by genetic immunization efficiently blocks HCV spread and dissemination. This antibody will be useful to further unravel the role of virus-host interactions during HCV entry and cell-cell transmission. Furthermore, this antibody may be of interest for the development of antivirals for prevention and treatment of HCV infection.


Blood | 2012

HCV glycoprotein E2 is a novel BDCA-2 ligand and acts as an inhibitor of IFN production by plasmacytoid dendritic cells

Jonathan Florentin; Besma Aouar; Clelia Dental; Christine Thumann; Guylène Firaguay; Françoise Gondois-Rey; Vassili Soumelis; Thomas F. Baumert; Jacques A. Nunès; Daniel Olive; Ivan Hirsch; Ruzena Wiersum Stranska

The elimination of hepatitis C virus (HCV) in > 50% of chronically infected patients by treatment with IFN-α suggests that plasmacytoid dendritic cells (pDCs), major producers of IFN-α, play an important role in the control of HCV infection. However, despite large amounts of Toll-like receptor 7-mediated IFN-α, produced by pDCs exposed to HCV-infected hepatocytes, HCV still replicates in infected liver. Here we show that HCV envelope glycoprotein E2 is a novel ligand of pDC C-type lectin immunoreceptors (CLRs), blood DC antigen 2 (BDCA-2) and DC-immunoreceptor (DCIR). HCV particles inhibit, via binding of E2 glycoprotein to CLRs, production of IFN-α and IFN-λ in pDCs exposed to HCV-infected hepatocytes, and induce in pDCs a rapid phosphorylation of Akt and Erk1/2, in a manner similar to the crosslinking of BDCA-2 or DCIR. Blocking of BDCA-2 and DCIR with Fab fragments of monoclonal antibodies preserves the capacity of pDCs to produce type I and III IFNs in the presence of HCV particles. Thus, negative interference of CLR signaling triggered by cell-free HCV particles with Toll-like receptor signaling triggered by cell-associated HCV results in the inhibition of the principal pDC function, production of IFN.


The Journal of Infectious Diseases | 2007

Early Evolution of Hepatitis C Virus (HCV) Quasispecies after Liver Transplant for HCV-Related Disease

Evelyne Schvoerer; Eric Soulier; Cathy Royer; Anne-Catherine Renaudin; Christine Thumann; Samira Fafi-Kremer; Nicolas Brignon; Stéphane Doridot; Nicolas Meyer; Patricia Pinson; Bernard Ellero; Marie-Lorraine Woehl-Jaegle; Carole Meyer; Philippe Wolf; Pierre Zachary; Thomas F. Baumert; Françoise Stoll-Keller

BACKGROUND End-stage liver disease as a result of chronic hepatitis C virus (HCV) infection is the main indication for liver transplant (LT), but allografts are systematically infected with HCV soon after transplant. Viral quasispecies are poorly described during the early posttransplant period. METHODS For 17 patients who received an LT for HCV disease, plasma viral quasispecies evolution was determined by sequence analysis of hypervariable region 1 of the E2 envelope gene before transplant (BT), after 7 days (D7), and after 1 month (M1). T helper (Th)1/Th2 cytokine levels were determined concomitantly. RESULTS HCV quasispecies showed a significant decrease in amino acid diversity at D7 and M1, compared with BT (P<.05). A correlation was observed between low plasma tumor necrosis factor-alpha levels at D7 and decreased quasispecies amino acid complexity at the same date. Nucleic acid diversity was lower for genotype 1 than for genotype 3 infection (P<.05). The complexity and diversity of amino acids were lower in patients with hepatocellular carcinoma (HCC) BT than in those without HCC (P<.05). Conserved amino acid residues within quasispecies were shared by the whole cohort before and after LT. CONCLUSION Viral structural and/or host immunological features could favor the emergence of fitter HCV strains after LT.

Collaboration


Dive into the Christine Thumann's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Thomas F. Baumert

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Fei Xiao

University of Strasbourg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Isabel Fofana

University of Strasbourg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge