Network


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

Hotspot


Dive into the research topics where Joachim Lupberger is active.

Publication


Featured researches published by Joachim Lupberger.


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.


Hepatology | 2010

Apolipoprotein E interacts with hepatitis C virus nonstructural protein 5A and determines assembly of infectious particles

Wagane J. A. Benga; Sophie E. Krieger; Maria Dimitrova; Mirjam B. Zeisel; Marie Parnot; Joachim Lupberger; Eberhard Hildt; Guangxiang Luo; John McLauchlan; Thomas F. Baumert; Catherine Schuster

Chronic hepatitis C virus (HCV) infection is a major cause of liver disease worldwide. Restriction of HCV infection to human hepatocytes suggests that liver‐specific host factors play a role in the viral life cycle. Using a yeast‐two‐hybrid system, we identified apolipoprotein E (apoE) as a liver‐derived host factor specifically interacting with HCV nonstructural protein 5A (NS5A) but not with other viral proteins. The relevance of apoE–NS5A interaction for viral infection was confirmed by co‐immunoprecipitation and co‐localization studies of apoE and NS5A in an infectious HCV cell culture model system. Silencing apoE expression resulted in marked inhibition of infectious particle production without affecting viral entry and replication. Analysis of particle production in liver‐derived cells with silenced apoE expression showed impairment of infectious particle assembly and release. The functional relevance of the apoE–NS5A interaction for production of viral particles was supported by loss or decrease of apoE–NS5A binding in assembly‐defective viral mutants. Conclusion: These results suggest that recruitment of apoE by NS5A is important for viral assembly and release of infectious viral particles. These findings have important implications for understanding the HCV life cycle and the development of novel antiviral strategies targeting HCV–lipoprotein interaction. (HEPATOLOGY 2010)


Journal of Hepatology | 2013

Host-targeting agents for prevention and treatment of chronic hepatitis C - Perspectives and challenges

Mirjam B. Zeisel; Joachim Lupberger; Isabel Fofana; Thomas F. Baumert

Hepatitis C virus (HCV) infection is a major cause of chronic liver disease and hepatocellular carcinoma worldwide. Furthermore, HCV-induced liver disease is a major indication of liver transplantation. In the past years, direct-acting antivirals (DAAs) targeting HCV enzymes have been developed. DAAs increase the virologic response to anti-HCV therapy but may lead to selection of drug-resistant variants and treatment failure. To date, strategies to prevent HCV infection are still lacking and antiviral therapy in immunocompromised patients, patients with advanced liver disease and HIV/HCV-co-infection remains limited. Alternative or complementary approaches addressing the limitations of current antiviral therapies are to boost the hosts innate immunity or interfere with host factors required for pathogenesis. Host-targeting agents (HTAs) provide an interesting perspective for novel antiviral strategies against viral hepatitis since they have (i) a high genetic barrier to resistance, (ii) a pan-genotypic antiviral activity, and (iii) complementary mechanisms of action to DAAs and might therefore act in a synergistic manner with current standard of care or DAAs in clinical development. This review highlights HTAs against HCV infection that have potential as novel antivirals and are in preclinical or clinical development.


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.


Hepatology | 2013

Epidermal growth factor receptor signaling impairs the antiviral activity of interferon-alpha.

Joachim Lupberger; Francois H.T. Duong; Isabel Fofana; Laetitia Zona; Fei Xiao; Christine Thumann; Sarah C. Durand; Patrick Pessaux; Mirjam B. Zeisel; Markus H. Heim; Thomas Baumert

Interferon‐alpha (IFN‐α) exhibits its antiviral activity through signal transducer and activator of transcription protein (STAT) signaling and the expression of IFN response genes (IRGs). Viral infection has been shown to result in activation of epidermal growth factor receptor (EGFR)—a host cell entry factor used by several viruses, including hepatitis C virus. However, the effect of EGFR activation for cellular antiviral responses is unknown. Here, we uncover cross‐talk between EGFR and IFN‐α signaling that has a therapeutic effect on IFN‐α‐based therapies and functional relevance for viral evasion and IFN resistance. We show that combining IFN‐α with the EGFR inhibitor, erlotinib, potentiates the antiviral effect of each compound in a highly synergistic manner. The extent of the synergy correlated with reduced STAT3 phosphorylation in the presence of erlotinib, whereas STAT1 phosphorylation was not affected. Furthermore, reduced STAT3 phosphorylation correlated with enhanced expression of suppressors of cytokine signaling 3 (SOCS3) in the presence of erlotinib and enhanced expression of the IRGs, radical S‐adenosyl methionine domain containing 2 and myxovirus resistance protein 1. Moreover, EGFR stimulation reduced STAT1 dimerization, but not phosphorylation, indicating that EGFR cross‐talk with IFN signaling acts on the STATs at the level of binding DNA. Conclusions: Our results support a model where inhibition of EGFR signaling impairs STAT3 phosphorylation, leading to enhanced IRG expression and antiviral activity. These data uncover a novel role of EGFR signaling in the antiviral activity of IFN‐α and open new avenues of improving the efficacy of IFN‐α‐based antiviral therapies. (Hepatology 2013;58:1225–1235)


Viruses | 2014

CD81-Receptor Associations — Impact for Hepatitis C Virus Entry and Antiviral Therapies

Laetitia Zona; Rajiv G. Tawar; Mirjam B. Zeisel; Fei Xiao; Catherine Schuster; Joachim Lupberger; Thomas Baumert

Tetraspanins are integral transmembrane proteins organized in microdomains displaying specific and direct interactions with other tetraspanins and molecular partners. Among them, CD81 has been implicated in a variety of physiological and pathological processes. CD81 also plays a crucial role in pathogen entry into host cells, including hepatitis C virus (HCV) entry into hepatocytes. HCV is a major cause of liver cirrhosis and hepatocellular carcinoma. HCV entry into hepatocytes is a complex process that requires the coordinated interaction of viral and host factors for the initiation of infection, including CD81, scavenger receptor BI, claudin-1, occludin, membrane-bound host cell kinases, Niemann-Pick C1 Like 1, Harvey rat sarcoma viral oncogene homolog (HRas), CD63 and transferrin receptor 1. Furthermore, recent data in HCV model systems have demonstrated that targeting critical components of tetraspanins and associated cell membrane proteins open new avenues to prevent and treat viral infection.


Journal of Virology | 2013

Functional analysis of claudin-6 and claudin-9 as entry factors for hepatitis C virus infection of human hepatocytes by using monoclonal antibodies.

Isabel Fofana; Laetitia Zona; Christine Thumann; Laura Heydmann; Sarah C. Durand; Joachim Lupberger; Hubert E. Blum; Patrick Pessaux; Claire Gondeau; Gary M. Reynolds; Jane A. McKeating; Fritz Grunert; John F. Thompson; Mirjam B. Zeisel; Thomas F. Baumert

ABSTRACT The relevance of claudin-6 and claudin-9 in hepatitis C virus (HCV) entry remains elusive. We produced claudin-6- or claudin-9-specific monoclonal antibodies that inhibit HCV entry into nonhepatic cells expressing exogenous claudin-6 or claudin-9. These antibodies had no effect on HCV infection of hepatoma cells or primary hepatocytes. Thus, although claudin-6 and claudin-9 can serve as entry factors in cell lines, HCV infection into human hepatocytes is not dependent on claudin-6 and claudin-9.


Biochimica et Biophysica Acta | 2015

Host cell kinases and the hepatitis C virus life cycle

Che C. Colpitts; Joachim Lupberger; Christian Doerig; Thomas F. Baumert

Hepatitis C virus (HCV) infection relies on virus-host interactions with human hepatocytes, a context in which host cell kinases play critical roles in every step of the HCV life cycle. During viral entry, cellular kinases, including EGFR, EphA2 and PKA, regulate the localization of host HCV entry factors and induce receptor complex assembly. Following virion internalization, viral genomes replicate on endoplasmic reticulum-derived membranous webs. The formation of membranous webs depends on interactions between the HCV NS5a protein and PI4KIIIα. The phosphorylation status of NS5a, regulated by PI4KIIIα, CKI and other kinases, also acts as a molecular switch to virion assembly, which takes place on lipid droplets. The formation of lipid droplets is enhanced by HCV activation of IKKα. In view of the multiple crucial steps in the viral life cycle that are mediated by host cell kinases, these enzymes also represent complementary targets for antiviral therapy. This article is part of a Special Issue entitled: Inhibitors of Protein Kinases.


Gut | 2018

miR-135a-5p-mediated downregulation of protein tyrosine phosphatase receptor delta is a candidate driver of HCV-associated hepatocarcinogenesis

Nicolaas Van Renne; Armando Andres Roca Suarez; Francois H.T. Duong; Claire Gondeau; Diego Calabrese; Nelly Fontaine; Amina Ababsa; Simonetta Bandiera; Tom Croonenborghs; Nathalie Pochet; Vito de Blasi; Patrick Pessaux; Tullio Piardi; Daniele Sommacale; Atsushi Ono; Kazuaki Chayama; Masashi Fujita; Hidewaki Nakagawa; Yujin Hoshida; Mirjam B. Zeisel; Markus H. Heim; Thomas F. Baumert; Joachim Lupberger

Background and aims HCV infection is a leading risk factor of hepatocellular carcinoma (HCC). However, even after viral clearance, HCC risk remains elevated. HCV perturbs host cell signalling to maintain infection, and derailed signalling circuitry is a key driver of carcinogenesis. Since protein phosphatases are regulators of signalling events, we aimed to identify phosphatases that respond to HCV infection with relevance for hepatocarcinogenesis. Methods We assessed mRNA and microRNA (miRNA) expression profiles in primary human hepatocytes, liver biopsies and resections of patients with HCC, and analysed microarray and RNA-seq data from paired liver biopsies of patients with HCC. We revealed changes in transcriptional networks through gene set enrichment analysis and correlated phosphatase expression levels to patient survival and tumour recurrence. Results We demonstrate that tumour suppressor protein tyrosine phosphatase receptor delta (PTPRD) is impaired by HCV infection in vivo and in HCC lesions of paired liver biopsies independent from tissue inflammation or fibrosis. In liver tissue adjacent to tumour, high PTPRD levels are associated with a dampened transcriptional activity of STAT3, an increase of patient survival from HCC and reduced tumour recurrence after surgical resection. We identified miR-135a-5p as a mechanistic regulator of hepatic PTPRD expression in patients with HCV. Conclusions We previously demonstrated that STAT3 is required for HCV infection. We conclude that HCV promotes a STAT3 transcriptional programme in the liver of patients by suppressing its regulator PTPRD via upregulation of miR-135a-5p. Our results show the existence of a perturbed PTPRD–STAT3 axis potentially driving malignant progression of HCV-associated liver disease.


Hepatology | 2013

Interferon‐lambda polymorphisms and hepatitis C virus clearance revisited

Joachim Lupberger; Daniel J. Felmlee; Thomas F. Baumert

Chronic infection with hepatitis C virus (HCV) is a common cause of liver cirrhosis and cancer. We performed RNA sequencing in primary human hepatocytes activated with synthetic double-stranded RNA to mimic HCV infection. Upstream of IFNL3 (IL28B) on chromosome 19q13.13, we discovered a new transiently induced region that harbors a dinucleotide variant ss469415590 (TT or DG), which is in high linkage disequilibrium with rs12979860, a genetic marker strongly associated with HCV clearance. ss469415590[DG] is a frameshift variant that creates a novel gene, designated IFNL4, encoding the interferon-k4 protein (IFNL4), which is moderately similar to IFNL3. Compared to rs12979860, ss469415590 is more strongly associated with HCV clearance in individuals of African ancestry, although it provides comparable information in Europeans and Asians. Transient overexpression of IFNL4 in a hepatoma cell line induced STAT1 and STAT2 phosphorylation and the expression of interferon-stimulated genes. Our findings provide new insights into the genetic regulation of HCV clearance and its clinical management.

Collaboration


Dive into the Joachim Lupberger's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
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

Fei Xiao

University of Strasbourg

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge