Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Hepatology ; 51(4): 1144-57, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20069648

ABSTRACT

UNLABELLED: 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.


Subject(s)
Antibodies/pharmacology , Antigens, CD/immunology , Cell Adhesion Molecules/immunology , Hepatitis C/therapy , Membrane Proteins/physiology , 12E7 Antigen , Antigens, CD/physiology , Claudin-1 , Humans , Immunization , Membrane Proteins/immunology , Neutralization Tests , Scavenger Receptors, Class B/physiology , Tetraspanin 28 , Tight Junctions/physiology , Virus Internalization
2.
Gastroenterology ; 135(5): 1719-1728.e1, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18718838

ABSTRACT

BACKGROUND & AIMS: Hepatitis C virus (HCV) is a leading cause of chronic hepatitis worldwide. Viral attachment and entry, representing the first steps of virus-host cell interactions, are major targets of adaptive host cell defenses. The mechanisms of antibody-mediated neutralization by host neutralizing responses in HCV infection are only poorly understood. Retroviral HCV pseudotypes (HCVpp) and recombinant cell culture-derived HCV (HCVcc) have been successfully used to study viral entry and antibody-mediated neutralization. METHODS: In this study, we used these model systems to investigate the mechanism of antibody-mediated neutralization by monoclonal antienvelope antibodies and polyclonal anti-HCV immunoglobulins purified from HCV-infected patients. RESULTS: Using a panel of monoclonal antienvelope antibodies, we identified an epitope within the E1 glycoprotein targeted by human neutralizing antibodies during postbinding events. Interestingly, we observed that host neutralizing responses in the majority of HCV-infected individuals include antibodies targeting HCV entry after binding of the virus to the target cell membrane. Using a kinetic assay based on HCVpp and HCVcc entry, we demonstrate that purified antiviral immunoglobulins derived from individual HCV-infected patients appear to inhibit HCV infection at an entry step closely linked to CD81 and scavenger receptor BI (SR-BI). CONCLUSIONS: Our results indicate that host neutralizing responses in HCV-infected patients target viral entry after HCV binding most likely related to HCV-CD81, and HCV-SR-BI interactions, as well as membrane fusion. These findings have implications not only for the understanding of the pathogenesis of HCV infection but also for the design of novel immunotherapeutic and preventive strategies.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD/immunology , Hepacivirus/immunology , Hepatitis C Antibodies/immunology , Hepatitis C, Chronic/drug therapy , Membrane Fusion/drug effects , Scavenger Receptors, Class B/immunology , Adult , Aged , Antibodies, Anti-Idiotypic/immunology , Antigens, CD/drug effects , Antigens, CD/metabolism , Cells, Cultured , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/virology , Humans , Immunoenzyme Techniques , Immunoglobulin G/immunology , Middle Aged , Receptors, Virus , Scavenger Receptors, Class B/drug effects , Scavenger Receptors, Class B/metabolism , Tetraspanin 28 , Viral Envelope Proteins/drug effects , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism
3.
J Virol ; 82(7): 3466-79, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18216094

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.


Subject(s)
Cross-Priming , Dendritic Cells/immunology , Dendritic Cells/virology , Hepacivirus/immunology , Receptors, Virus/physiology , Scavenger Receptors, Class B/physiology , Virus Internalization , Animals , Antigens, Surface/analysis , B-Lymphocytes/chemistry , Cell Line , Cells, Cultured , Cricetinae , Dendritic Cells/chemistry , Hepacivirus/physiology , Hepatocytes/chemistry , Humans , Insecta , Monocytes/chemistry , Receptors, Virus/biosynthesis , Scavenger Receptors, Class B/biosynthesis , T-Lymphocytes/chemistry , Virosomes/metabolism , Virus Attachment
4.
Hepatology ; 46(6): 1722-31, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18000990

ABSTRACT

UNLABELLED: Hepatitis C virus (HCV) is a major cause of chronic hepatitis worldwide. Scavenger receptor class B type I (SR-BI) has been shown to bind HCV envelope glycoprotein E2, participate in entry of HCV pseudotype particles, and modulate HCV infection. However, the functional role of SR-BI for productive HCV infection remains unclear. In this study, we investigated the role of SR-BI as an entry factor for infection of human hepatoma cells using cell culture-derived HCV (HCVcc). Anti-SR-BI antibodies directed against epitopes of the human SR-BI extracellular loop specifically inhibited HCVcc infection in a dose-dependent manner. Down-regulation of SR-BI expression by SR-BI-specific short interfering RNAs (siRNAs) markedly reduced the susceptibility of human hepatoma cells to HCVcc infection. Kinetic studies demonstrated that SR-BI acts predominately after binding of HCV at an entry step occurring at a similar time point as CD81-HCV interaction. Although the addition of high-density lipoprotein (HDL) enhanced the efficiency of HCVcc infection, anti-SR-BI antibodies and SR-BI-specific siRNA efficiently inhibited HCV infection independent of lipoprotein. CONCLUSION: Our data suggest that SR-BI (i) represents a key host factor for HCV entry, (ii) is implicated in the same HCV entry pathway as CD81, and (iii) targets an entry step closely linked to HCV-CD81 interaction.


Subject(s)
Antigens, CD/immunology , Hepatitis C/immunology , Scavenger Receptors, Class B/immunology , Carcinoma, Hepatocellular , Cell Line, Tumor , Cells, Cultured , Hepacivirus , Humans , Tetraspanin 28
5.
J Virol ; 80(21): 10579-90, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16928753

ABSTRACT

Cellular binding and entry of hepatitis C virus (HCV) are the first steps of viral infection and represent a major target for antiviral antibodies and novel therapeutic strategies. We have recently demonstrated that heparan sulfate (HS) plays a key role in the binding of HCV envelope glycoprotein E2 to target cells (Barth et al., J. Biol. Chem. 278:41003-41012, 2003). In this study, we characterized the HCV-HS interaction and analyzed its inhibition by antiviral host immune responses. Using recombinant envelope glycoproteins, virus-like particles, and HCV pseudoparticles as model systems for the early steps of viral infection, we mapped viral and cellular determinants of HCV-HS interaction. HCV-HS binding required a specific HS structure that included N-sulfo groups and a minimum of 10 to 14 saccharide subunits. HCV envelope binding to HS was mediated by four viral epitopes overlapping the E2 hypervariable region 1 and E2-CD81 binding domains. In functional studies using HCV pseudoparticles, we demonstrate that HCV binding and entry are specifically inhibited by highly sulfated HS. Finally, HCV-HS binding was markedly inhibited by antiviral antibodies derived from HCV-infected individuals. In conclusion, our results demonstrate that binding of the viral envelope to a specific HS configuration represents an important step for the initiation of viral infection and is a target of antiviral host immune responses in vivo. Mapping of viral and cellular determinants of HCV-HS interaction sets the stage for the development of novel HS-based antiviral strategies targeting viral attachment and entry.


Subject(s)
Hepacivirus/physiology , Hepacivirus/pathogenicity , Heparitin Sulfate/metabolism , Viral Envelope Proteins/physiology , Animals , Antibodies, Viral , Antigens, Viral , Binding Sites , Cell Line , Epitope Mapping , HeLa Cells , Hepacivirus/genetics , Hepacivirus/immunology , Heparitin Sulfate/chemistry , Hepatitis C, Chronic/etiology , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/virology , Humans , Mice , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Surface Plasmon Resonance , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...