Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Front Immunol ; 12: 775098, 2021.
Article in English | MEDLINE | ID: mdl-34975862

ABSTRACT

Hepatitis C virus (HCV) is highly variable and transmits through infected blood to establish a chronic liver infection in the majority of patients. Our knowledge on the infectivity of clinical HCV strains is hampered by the lack of in vitro cell culture systems that support efficient viral replication. We and others have reported that HCV can associate with and infect immune cells and may thereby evade host immune surveillance and elimination. To evaluate whether B cells play a role in HCV transmission, we assessed the ability of B cells and sera from recent (<2 years) or chronic (≥ 2 years) HCV patients to infect humanized liver chimeric mice. HCV was transmitted by B cells from chronic infected patients whereas the sera were non-infectious. In contrast, B cells from recently infected patients failed to transmit HCV to the mice, whereas all serum samples were infectious. We observed an association between circulating anti-glycoprotein E1E2 antibodies and B cell HCV transmission. Taken together, our studies provide evidence for HCV transmission by B cells, findings that have clinical implications for prophylactic and therapeutic antibody-based vaccine design.


Subject(s)
B-Lymphocytes/virology , Hepacivirus/pathogenicity , Hepatitis C/transmission , Adult , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Broadly Neutralizing Antibodies/blood , Broadly Neutralizing Antibodies/immunology , Disease Models, Animal , Female , Hepacivirus/immunology , Hepacivirus/isolation & purification , Hepatitis C/blood , Hepatitis C/prevention & control , Hepatitis C/virology , Humans , Liver/pathology , Liver/virology , Liver Transplantation , Male , Mice , Middle Aged , Serum/virology , Transplantation Chimera , Vaccine Development/methods , Viral Envelope Proteins/immunology , Viral Hepatitis Vaccines/therapeutic use , Young Adult
2.
Virology ; 514: 30-41, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29128754

ABSTRACT

Monoclonal antibodies (mAbs) targeting the hepatitis C virus (HCV) envelope have been raised mainly against envelope protein 2 (E2), while the antigenic epitopes of envelope protein 1 (E1) are not fully identified. Here we describe the detailed characterization of a human mAb, designated A6, generated from an HCV genotype 1b infected patient. ELISA results showed reactivity of mAb A6 to full-length HCV E1E2 of genotypes 1a, 1b and 2a. Epitope mapping identified a region spanning amino acids 230-239 within the N-terminal region of E1 as critical for binding. Antibody binding to this epitope was not conformation dependent. Neutralization assays showed that mAb A6 lacks neutralizing capacity and does not interfere with the activity of known neutralizing antibodies. In summary, mAb A6 is an important tool to study the structure and function of E1 within the viral envelope, a crucial step in the development of an effective prophylactic HCV vaccine.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Hepacivirus/immunology , Hepatitis C/immunology , Viral Envelope Proteins/immunology , Antibodies, Monoclonal/analysis , Antibodies, Neutralizing/analysis , Epitope Mapping , Genotype , Hepacivirus/chemistry , Hepacivirus/genetics , Hepatitis C/genetics , Humans , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
3.
Antiviral Res ; 148: 53-64, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29074219

ABSTRACT

Infections with hepatitis C virus (HCV) represent a worldwide health burden and a prophylactic vaccine is still not available. Liver transplantation (LT) is often the only option for patients with HCV-induced end-stage liver disease. However, immediately after transplantation, the liver graft becomes infected by circulating virus, resulting in accelerated progression of liver disease. Although the efficacy of HCV treatment using direct-acting antivirals has improved significantly, immune compromised LT-patients and patients with advanced liver disease remain difficult to treat. As an alternative approach, interfering with viral entry could prevent infection of the donor liver. We generated a human monoclonal antibody (mAb), designated 2A5, which targets the HCV envelope. The neutralizing activity of mAb 2A5 was assessed using multiple prototype and patient-derived HCV pseudoparticles (HCVpp), cell culture produced HCV (HCVcc), and a human-liver chimeric mouse model. Neutralization levels observed for mAb 2A5 were generally high and mostly superior to those obtained with AP33, a well-characterized HCV-neutralizing monoclonal antibody. Using humanized mice, complete protection was observed after genotype 1a and 4a HCV challenge, while only partial protection was achieved using gt1b and 6a isolates. Epitope mapping revealed that mAb 2A5 binding is conformation-dependent and identified the E2-region spanning amino acids 434 to 446 (epitope II) as the predominant contact domain. CONCLUSION: mAb 2A5 shows potent anti-HCV neutralizing activity both in vitro and in vivo and could hence represent a valuable candidate to prevent HCV recurrence in LT-patients. In addition, the detailed identification of the neutralizing epitope can be applied for the design of prophylactic HCV vaccines.


Subject(s)
Antibodies, Neutralizing/pharmacology , Antibodies, Neutralizing/therapeutic use , Hepacivirus/drug effects , Hepatitis C Antibodies/pharmacology , Hepatitis C Antibodies/therapeutic use , Hepatitis C/drug therapy , Viral Envelope Proteins/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/immunology , Disease Models, Animal , Epitope Mapping , Epitopes/genetics , Epitopes/immunology , Genotype , Hepacivirus/genetics , Hepacivirus/immunology , Hepatitis C/immunology , Hepatitis C/prevention & control , Hepatitis C/virology , Hepatitis C Antibodies/immunology , Humans , Liver Transplantation , Mice , Mice, SCID , Mutation , Neutralization Tests , Structure-Activity Relationship , Viral Envelope Proteins/genetics , Virus Internalization/drug effects
4.
Antiviral Res ; 141: 150-154, 2017 05.
Article in English | MEDLINE | ID: mdl-28232247

ABSTRACT

Hepatitis E virus (HEV) is considered as an important pathogen in developing countries but there is growing evidence of its increasing significance and prevalence in the Western world. Although most acute HEV infections resolve spontaneously, chronicity has been observed in immunocompromised patients. The study of HEV has been hampered by the absence of practical animal models. Because the in vivo study of HEV was essentially limited to primates and pigs we recently established the human-liver chimeric uPA-SCID mouse model as a useful tool to study HEV infection. Because the humanized FRG mouse model, another type of mouse with humanized liver, is more easily accessible to the scientific community, we investigated its susceptibility to HEV infection. FRG mice were transplanted with human hepatocytes and challenged with different HEV genotypes using different routes of exposure. Our data clearly shows that the humanized FRG mouse is an alternative animal model for the study HEV infection. As observed in the uPA-SCID model, controlled oral inoculation did not lead to active infection. However, intrasplenic injection of genotype 3-infected patient plasma did result into persistent infection. Although the efficiency of transmission was low, this observation corroborates previously published case reports of blood transfusion-associated HEV transmission.


Subject(s)
Disease Models, Animal , Hepatitis E/transmission , Hepatocytes/virology , Liver/virology , Plasma/virology , RNA, Viral/blood , Animals , Blood Transfusion , DNA-Binding Proteins/deficiency , Genotype , Hepatitis E/virology , Hepatitis E virus/genetics , Hepatitis E virus/isolation & purification , Hepatitis E virus/physiology , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Mice , Mice, SCID
5.
Antiviral Res ; 139: 129-137, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28062191

ABSTRACT

Due to the highly restricted species-tropism of Hepatitis C virus (HCV) a limited number of animal models exist for pre-clinical evaluation of vaccines and antiviral compounds. The human-liver chimeric mouse model allows heterologous challenge with clinically relevant strains derived from patients. However, to date, the transmission and longitudinal evolution of founder viral populations in this model have not been characterized in-depth using state-of-the-art sequencing technologies. Focusing on NS3 protease encoding region of the viral genome, mutant spectra in a donor inoculum and individual recipient mice were determined via Illumina sequencing and compared, to determine the effects of transmission on founder viral population complexity. In all transmissions, a genetic bottleneck was observed, although diverse viral populations were transmitted in each case. A low frequency cloud of mutations (<1%) was detectable in the donor inoculum and recipient mice, with single nucleotide variants (SNVs) > 1% restricted to a subset of nucleotides. The population of SNVs >1% was reduced upon transmission while the low frequency SNV cloud remained stable. Fixation of multiple identical synonymous substitutions was apparent in independent transmissions, and no evidence for reversion of T-cell epitopes was observed. In addition, susceptibility of founder populations to antiviral therapy was assessed. Animals were treated with protease inhibitor (PI) monotherapy to track resistance associated substitution (RAS) emergence. Longitudinal analyses revealed a decline in population diversity under therapy, with no detectable RAS >1% prior to therapy commencement. Despite inoculation from a common source and identical therapeutic regimens, unique RAS emergence profiles were identified in different hosts prior to and during therapeutic failure, with complex mutational signatures at protease residues 155, 156 and 168 detected. Together these analyses track viral population complexity at high-resolution in the human-liver chimeric mouse model post-transmission and under therapeutic intervention, revealing novel insights into the evolutionary processes which shape viral protease population composition at various critical stages of the viral life-cycle.


Subject(s)
Hepacivirus/enzymology , Hepacivirus/genetics , Hepatitis C/transmission , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Animals , Antiviral Agents/therapeutic use , Disease Models, Animal , Evolution, Molecular , Genetic Variation , Genome, Viral , Genotype , Hepacivirus/drug effects , Hepatitis C/drug therapy , Hepatitis C/virology , Humans , Mice , Mutation , Protease Inhibitors/administration & dosage , Protease Inhibitors/therapeutic use , Sequence Analysis, DNA , Serine Endopeptidases/chemistry , Viral Nonstructural Proteins/chemistry
6.
Gut ; 66(5): 920-929, 2017 05.
Article in English | MEDLINE | ID: mdl-27006186

ABSTRACT

OBJECTIVE: The hepatitis E virus (HEV) is responsible for approximately 20 million infections per year worldwide. Although most infected people can spontaneously clear an HEV infection, immune-compromised individuals may evolve towards chronicity. Chronic HEV infection can be cured using ribavirin, but viral isolates with low ribavirin sensitivity have recently been identified. Although some HEV isolates can be cultured in vitro, in vivo studies are essentially limited to primates and pigs. Since the use of these animals is hampered by financial, practical and/or ethical concerns, we evaluated if human liver chimeric mice could serve as an alternative. DESIGN: Humanised mice were inoculated with different HEV-containing preparations. RESULTS: Chronic HEV infection was observed after intrasplenic injection of cell culture-derived HEV, a filtered chimpanzee stool suspension and a patient-derived stool suspension. The viral load was significantly higher in the stool compared with the plasma. Overall, the viral titre in genotype 3-infected mice was lower than that in genotype 1-infected mice. Analysis of liver tissue of infected mice showed the presence of viral RNA and protein, and alterations in host gene expression. Intrasplenic injection of HEV-positive patient plasma and oral inoculation of filtered stool suspensions did not result in robust infection. Finally, we validated our model for the evaluation of novel antiviral compounds against HEV using ribavirin. CONCLUSIONS: Human liver chimeric mice can be infected with HEV of different genotypes. This small animal model will be a valuable tool for the in vivo study of HEV infection and the evaluation of novel antiviral molecules.


Subject(s)
Disease Models, Animal , Hepatitis E virus/genetics , Hepatitis E/virology , Liver/chemistry , RNA, Viral/analysis , Viral Proteins/analysis , Animals , Antiviral Agents/therapeutic use , Gene Expression , Genotype , Hepatitis E/drug therapy , Hepatitis E/genetics , Hepatocytes/transplantation , Hepatocytes/virology , Host-Pathogen Interactions , Humans , Mice , Ribavirin/therapeutic use , Transplantation Chimera , Viral Load
7.
Gut ; 65(12): 1988-1997, 2016 12.
Article in English | MEDLINE | ID: mdl-26589670

ABSTRACT

OBJECTIVE: HCV is a major cause of chronic liver disease worldwide, but the role of neutralising antibodies (nAbs) in its natural history remains poorly defined. We analysed the in vivo role of hypervariable region 1 (HVR1) for HCV virion properties, including nAb susceptibility. DESIGN: Analysis of HCV from human liver chimeric mice infected with cell-culture-derived prototype genotype 2a recombinant J6/JFH1 or HVR1-deleted variant J6/JFH1ΔHVR1 identified adaptive mutations, which were analysed by reverse genetics in Huh7.5 and CD81-deficient S29 cells. The increased in vivo genomic stability of the adapted viruses facilitated ex vivo density analysis by ultracentrifugation and in vivo neutralisation experiments addressing the role of HVR1. RESULTS: In vivo, J6/JFH1 and J6/JFH1ΔHVR1 depended on single substitutions within amino acids 867-876 in non-structural protein, NS2. The identified A876P-substitution resulted in a 4.7-fold increase in genomic stability. In vitro, NS2 substitutions enhanced infectivity 5-10-fold by increasing virus assembly. Mouse-derived mJ6/JFH1A876P and mJ6/JFH1ΔHVR1/A876P viruses displayed similar heterogeneous densities of 1.02-1.1 g/mL. Human liver chimeric mice loaded with heterologous patient H (genotype 1a) immunoglobulin had partial protection against mJ6/JFH1A876P and complete protection against mJ6/JFH1ΔHVR1/A876P. Interestingly, we identified a putative escape mutation, D476G, in mJ6/JFH1A876P. This mutation in hypervariable region 2 conferred 6.6-fold resistance against H06 IgG in vitro. CONCLUSIONS: The A876P-substitution bridges in vitro and in vivo studies using J6/JFH1-based recombinants. We provide the first in vivo evidence that HVR1 protects cross-genotype conserved HCV neutralisation epitopes, which advocates the possibility of using HVR1-deleted viruses as vaccine antigens to boost broadly reactive protective nAb responses.


Subject(s)
Hepacivirus/genetics , Mutation , Viral Proteins/genetics , Amino Acid Substitution/genetics , Animals , Antibodies, Neutralizing/blood , Disease Models, Animal , Genotype , Hepacivirus/pathogenicity , Hepatitis C/genetics , Hepatitis C/immunology , Hepatitis C/prevention & control , Hepatitis C/virology , In Vitro Techniques , Mice , Viral Hepatitis Vaccines/genetics , Viral Nonstructural Proteins/genetics
8.
Gut ; 65(12): 2029-2034, 2016 12.
Article in English | MEDLINE | ID: mdl-26306759

ABSTRACT

OBJECTIVE: Direct-acting antivirals (DAAs) inhibit hepatitis C virus (HCV) infection by targeting viral proteins that play essential roles in the replication process. However, selection of resistance-associated variants (RAVs) during DAA therapy has been a cause of therapeutic failure. In this study, we wished to address whether such RAVs could be controlled by the co-administration of host-targeting entry inhibitors that prevent intrahepatic viral spread. DESIGN: We investigated the effect of adding an entry inhibitor (the anti-scavenger receptor class B type I mAb1671) to a DAA monotherapy (the protease inhibitor ciluprevir) in human-liver mice chronically infected with HCV of genotype 1b. Clinically relevant non-laboratory strains were used to achieve viraemia consisting of a cloud of related viral variants (quasispecies) and the emergence of RAVs was monitored at high resolution using next-generation sequencing. RESULTS: HCV-infected human-liver mice receiving DAA monotherapy rapidly experienced on-therapy viral breakthrough. Deep sequencing of the HCV protease domain confirmed the manifestation of drug-resistant mutants upon viral rebound. In contrast, none of the mice treated with a combination of the DAA and the entry inhibitor experienced on-therapy viral breakthrough, despite detection of RAV emergence in some animals. CONCLUSIONS: This study provides preclinical in vivo evidence that addition of an entry inhibitor to an anti-HCV DAA regimen restricts the breakthrough of DAA-resistant viruses. Our approach is an excellent strategy to prevent therapeutic failure caused by on-therapy rebound of DAA-RAVs. Inclusion of an entry inhibitor to the newest DAA combination therapies may further increase response rates, especially in difficult-to-treat patient populations.


Subject(s)
Antiviral Agents/pharmacology , Drug Resistance, Viral/genetics , Hepacivirus/genetics , Protease Inhibitors/pharmacology , Amino Acid Substitution , Animals , Disease Models, Animal , Genotype , Hepacivirus/drug effects , Hepatitis C, Chronic/drug therapy , Liver/drug effects , Mice , Mutation, Missense , Viral Nonstructural Proteins/genetics
9.
Hepatology ; 63(4): 1120-34, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26710081

ABSTRACT

UNLABELLED: End-stage liver disease (ESLD) caused by hepatitis C virus (HCV) infection is a major indication for liver transplantation. However, immediately after transplantation, the liver graft of viremic patients universally becomes infected by circulating virus, resulting in accelerated liver disease progression. Currently available direct-acting antiviral therapies have reduced efficacy in patients with ESLD and prophylactic strategies to prevent HCV recurrence are still highly needed. In this study, we compared the ability of two broadly reactive monoclonal antibodies (mAbs), designated 3/11 and AP33, recognizing a distinct, but overlapping, epitope in the viral E2 glycoprotein to protect humanized mice from a patient-derived HCV challenge. Their neutralizing activity was assessed using the HCV pseudoparticles and cell-culture-derived HCV systems expressing multiple patient-derived envelopes and a human-liver chimeric mouse model. HCV RNA was readily detected in all control mice challenged with a patient-derived HCV genotype 1b isolate, whereas 3 of 4 AP33-treated mice were completely protected. In contrast, only one of four 3/11-treated mice remained HCV-RNA negative throughout the observation period, whereas the other 3 had a viral load that was indistinguishable from that in the control group. The increased in vivo efficacy of AP33 was in line with its higher affinity and neutralizing capacity observed in vitro. CONCLUSIONS: Although mAbs AP33 and 3/11 target the same region in E2, only mAb AP33 can efficiently protect from challenge with a heterologous HCV population in vivo. Given that mAb AP33 efficiently neutralizes viral variants that escaped the humoral immune response and reinfected the liver graft of transplant patients, it may be a valuable candidate to prevent HCV recurrence. In addition, our data are valuable for the design of a prophylactic vaccine.


Subject(s)
Antibodies, Monoclonal/pharmacology , Hepacivirus/immunology , Hepatitis C Antibodies/immunology , Hepatitis C/drug therapy , Hepatitis C/immunology , Viral Envelope Proteins/immunology , Animals , Cells, Cultured , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Epitopes , Hepatitis C Antibodies/metabolism , Humans , Mice , Mice, Transgenic , Statistics, Nonparametric
10.
Hepatology ; 55(2): 364-72, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21953761

ABSTRACT

UNLABELLED: Endstage liver disease caused by chronic hepatitis C virus (HCV) infection is the leading indication for liver transplantation in the Western world. However, immediate reinfection of the grafted donor liver by circulating virus is inevitable and liver disease progresses much faster than the original disease. Standard antiviral therapy is not well tolerated and usually ineffective in liver transplant patients, whereas anti-HCV immunotherapy is hampered by the extreme genetic diversity of the virus and its ability to spread by way of cell-cell contacts. We generated a human monoclonal antibody against scavenger receptor class B type I (SR-BI), monoclonal antibody (mAb)16-71, which can efficiently prevent infection of Huh-7.5 hepatoma cells and primary hepatocytes by cell-culture-derived HCV (HCVcc). Using an Huh7.5 coculture system we demonstrated that mAb16-71 interferes with direct cell-to-cell transmission of HCV. Finally we evaluated the in vivo efficacy of mAb16-71 in "human liver urokinase-type plasminogen activator, severe combined immune deficiency (uPA-SCID) mice" (chimeric mice). A 2-week anti-SR-BI therapy that was initiated 1 day before viral inoculation completely protected all chimeric mice from infection with serum-derived HCV of different genotypes. Moreover, a 9-day postexposure therapy that was initiated 3 days after viral inoculation (when viremia was already observed in the animals) suppressed the rapid viral spread observed in untreated control animals. After cessation of anti-SR-BI-specific antibody therapy, a rise of the viral load was observed. CONCLUSION: Using in vitro cell culture and human liver-chimeric mouse models, we show that a human mAb targeting the HCV coreceptor SR-BI completely prevents infection and intrahepatic spread of multiple HCV genotypes. This strategy may be an efficacious way to prevent infection of allografts following liver transplantation in chronic HCV patients, and may even hold promise for the prevention of virus rebound during or following antiviral therapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD36 Antigens/immunology , Hepatitis C/prevention & control , Animals , Cell Line, Tumor , Chimera , Genotype , Humans , Liver Transplantation , Mice , Mice, SCID , Secondary Prevention
11.
Hepatology ; 53(3): 755-62, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21319203

ABSTRACT

UNLABELLED: Control of hepatitis C virus (HCV) infection remains a huge challenge of global medical importance. Using a variety of in vitro approaches, neutralizing antibodies (nAbs) have been identified in patients with acute and chronic hepatitis C. The exact role these nAbs play in the resolution of acute HCV infection still remains elusive. We have previously shown that purified polyclonal antibodies isolated from plasma obtained in 2003 from a chronic HCV patient (Patient H) can protect human liver chimeric mice from a subsequent challenge with the autologous HCV strain isolated from Patient H in 1977 (H77). In this study we investigated whether polyclonal antibodies isolated from Patient H in 2006 (H06), which display high cross-genotype neutralizing activity in both the HCV pseudoparticle (HCVpp) and HCV cell culture (HCVcc) systems, were also able to prevent HCV infection of different genotypes (gt) in vivo. Following passive immunization with H06-antibodies, chimeric mice were challenged with the consensus strains H77C (gt1a), ED43 (gt4a), or HK6a (gt6a). In accordance with previous results, H06-antibodies prevented infection of chimeric mice with the autologous virus. However, the outcome of a homologous challenge is highly influenced by the amount of challenge virus injected. Depending on the viral genotype used, H06-antibodies were able to protect up to 50% of chimeric mice from a heterologous challenge. Animals in which the antibody pretreatment failed displayed a clear delay in the kinetics of viral infection. Sequence analysis of the recovered viruses did not suggest antibody-induced viral escape. CONCLUSION: Polyclonal anti-HCV antibodies isolated from a chronic HCV patient can protect against an in vivo challenge with different HCV genotypes. However, the in vivo protective efficacy of cross-genotype neutralizing antibodies was less than predicted by cell culture experiments.


Subject(s)
Antibodies, Neutralizing/immunology , Hepatitis C Antibodies/blood , Amino Acid Sequence , Animals , Hepacivirus/immunology , Hepatitis C, Chronic/immunology , Humans , Mice , Mice, SCID , Transplantation Chimera/immunology , Viral Envelope Proteins/genetics , Viral Vaccines/immunology
12.
J Gen Virol ; 83(Pt 9): 2279-2289, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12185283

ABSTRACT

It was observed recently that recombinant yeast-derived hepatitis B surface antigen (rHBsAg) particles, which contain the S protein only, bind almost exclusively to monocytes. It is shown here that binding requires the presence of the LPS receptor CD14. Furthermore, evidence is presented that a domain on CD14 that is identical to or largely overlaps with the LPS-binding pocket is instrumental for the attachment of rHBsAg. Additionally, it is shown that the heat-labile LPS-binding protein (LBP) catalyses the binding of rHBsAg to the cells. Remarkably, natural plasma-derived HBsAg (pHBsAg) does not have this property. pHBsAg devoid of its lipids and reconstituted with phosphatidylserine or phosphatidylglycerol acquires the characteristic of yeast-derived HBsAg. Clearly, the interaction of rHBsAg with the cell membrane is determined by the presence of charged phospholipids that are absent in pHBsAg. Although a lipid-receptor interaction is suggested, antibody-inhibition experiments suggest a possible involvement of the C-terminal region of the S protein in the interaction with monocytes. The possible implications of these observations for hepatitis B virus (HBV) infection and HBV vaccine efficiency are discussed.


Subject(s)
Acute-Phase Proteins , Carrier Proteins/metabolism , Hepatitis B Surface Antigens/metabolism , Lipopolysaccharide Receptors/metabolism , Membrane Glycoproteins , Monocytes/metabolism , Phospholipids/metabolism , Animals , CHO Cells , Cell Membrane/immunology , Cell Membrane/virology , Cells, Cultured , Cricetinae , Hepatitis B Surface Antigens/chemistry , Humans , Lipopolysaccharide Receptors/genetics , Protein Binding , Recombinant Proteins/metabolism , Transfection
13.
J Gen Virol ; 83(Pt 6): 1281-1289, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12029142

ABSTRACT

During hepatitis B virus (HBV) infection, high numbers of non-infectious HBV surface antigen (HBsAg) particles are present in circulation. It is shown here that recombinant HBsAg (rHBsAg) particles, which contain the S protein only, bind almost exclusively to monocytes. Attachment of rHBsAg to the THP-1 pre-monocytic cell line occurs upon 1,25-dihydroxyvitamin D3-induced differentiation. Binding to monocytes is enhanced by a heat-labile serum protein and is inhibited by Ca(2+)/Mg(2+), low pH and an HBsAg-specific monoclonal antibody. Furthermore, it is shown that rHBsAg suppresses lipopolysaccharide- and IL-2-induced production of cytokines. These results suggest the existence of a monocyte-specific receptor, the engagement of which by HBsAg suppresses the activity of these cells.


Subject(s)
Hepatitis B Surface Antigens/immunology , Monocytes/immunology , Antibodies, Monoclonal/pharmacology , Blood Proteins/pharmacology , Calcitriol , Calcium/pharmacology , Cell Differentiation , Cells, Cultured , Cytokines/analysis , Dose-Response Relationship, Drug , Hot Temperature , Humans , Hydrogen-Ion Concentration , Interleukin-2 , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides , Magnesium/pharmacology , Monocytes/metabolism , Protein Binding/drug effects , Recombinant Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...