Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
2.
Biochimie ; 113: 35-46, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25828806

ABSTRACT

ADP-ribosylation reactions consist of mono-ADP-ribosylation, poly-ADP-ribosylation and cyclic ADP-ribosylation. These reactions play essential roles in many important physiological and pathophysiological events. The types of chemical linkages, the evolutionarily conserved motif within the enzymes to determine the target specificity, stereochemistry of the ADP-ribosylated products, and the chemical reactions taking place among the enzymes and substrates are discussed.


Subject(s)
ADP Ribose Transferases/metabolism , Adenosine Diphosphate Ribose/metabolism , Animals , Humans
3.
BMC Biochem ; 15: 26, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25494717

ABSTRACT

BACKGROUND: Cholix toxin is an ADP-ribosyltransferase found in non-O1/non-O139 strains of Vibrio cholera. The catalytic fragment of cholix toxin was characterized as a diphthamide dependent ADP-ribosyltransferase. RESULTS: Our studies on the enzymatic activity of cholix toxin catalytic fragment show that the transfer of ADP-ribose to toxin takes place by a predominantly intramolecular mechanism and results in the preferential alkylation of arginine residues proximal to the NAD+ binding pocket. Multiple arginine residues, located near the catalytic site and at distal sites, can be the ADP-ribose acceptor in the auto-reaction. Kinetic studies of a model enzyme, M8, showed that a diffusible intermediate preferentially reacted with arginine residues in proximity to the NAD+ binding pocket. ADP-ribosylarginine activity of cholix toxin catalytic fragment could also modify exogenous substrates. Auto-ADP-ribosylation of cholix toxin appears to have negatively regulatory effect on ADP-ribosylation of exogenous substrate. However, at the presence of both endogenous and exogenous substrates, ADP-ribosylation of exogenous substrates occurred more efficiently than that of endogenous substrates. CONCLUSIONS: We discovered an ADP-ribosylargininyl activity of cholix toxin catalytic fragment from our studies in auto-ADP-ribosylation, which is mediated through diffusible intermediates. The lifetime of the hypothetical intermediate exceeds recorded and predicted lifetimes for the cognate oxocarbenium ion. Therefore, a diffusible strained form of NAD+ intermediate was proposed to react with arginine residues in a proximity dependent manner.


Subject(s)
Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/metabolism , Cholera Toxin/metabolism , Adenosine Diphosphate Ribose/toxicity , Diffusion
4.
J Virol ; 82(13): 6711-20, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18417597

ABSTRACT

Hepatitis C virus (HCV) often causes persistent infection despite the presence of neutralizing antibodies against the virus in the sera of hepatitis C patients. HCV infects both hepatocytes and B cells through the binding of its envelope glycoprotein E2 to CD81, the putative viral receptor. Previously, we have shown that E2-CD81 interaction induces hypermutation of heavy-chain immunoglobulin (V(H)) in B cells. We hypothesize that if HCV infects antibody-producing B cells, the resultant hypermutation of V(H) may lower the affinity and specificity of the HCV-specific antibodies, enabling HCV to escape from immune surveillance. To test this hypothesis, we infected human hybridoma clones producing either neutralizing or non-neutralizing anti-E2 or anti-E1 antibodies with a lymphotropic HCV (SB strain). All of the hybridoma clones, except for a neutralizing antibody-producing hybridoma, could be infected with HCV and support virus replication for at least 8 weeks after infection. The V(H) sequences in the infected hybridomas had a significantly higher mutation frequency than those in the uninfected hybridomas, with mutations concentrating in complementarity-determining region 3. These mutations lowered the antibody affinity against the targeting protein and also lowered the virus-neutralizing activity of anti-E2 antibodies. Furthermore, antibody-mediated complement-dependent cytotoxicity with the antibodies secreted from the HCV-infected hybridomas was impaired. These results suggest that HCV infection could cause some anti-HCV-antibody-producing hybridoma B cells to make less-protective antibodies.


Subject(s)
Antibodies, Viral/immunology , B-Lymphocytes/virology , Hepacivirus/immunology , Immunoglobulin Heavy Chains/genetics , Viral Envelope Proteins/immunology , Amino Acid Sequence , Antibodies, Viral/genetics , Antigens, CD/metabolism , Base Sequence , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Hepacivirus/genetics , Humans , Hybridomas/immunology , Molecular Sequence Data , Mutation/genetics , Sequence Analysis, DNA , Tetraspanin 28 , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virus Replication/genetics , Virus Replication/physiology
5.
Virology ; 361(1): 161-73, 2007 Apr 25.
Article in English | MEDLINE | ID: mdl-17175001

ABSTRACT

It has been reported that hepatitis C virus (HCV) may infect and replicate in human T cells, particularly in perihepatic lymph nodes, but the extent and consequence of T-cell infection in patients is unclear. This study is conducted to characterize the parameters and functional consequences of HCV infection in T lymphocytes. By using a lymphotropic HCV strain, we showed that HCV could infect T cell lines (Molt-4 and Jurkat cells) in vitro. Both positive- and negative-strand HCV RNA were detected for several weeks after infection. Viral proteins could also be detected by immunofluorescence studies. Moreover, infectious HCV particles were produced from Molt-4 cell cultures, and could be used to infect naïve T cell lines. HCV could also infect human primary CD4+ T cells, particularly naïve (CD45RA+CD45RO-) CD4+ cells, in culture. The amounts of STAT-1 and phosphorylated STAT-1 proteins in the infected Molt-4 cells were significantly less than those in uninfected cultures, suggesting the possibility of defect in interferon-gamma signaling. Indeed, T-bet and STAT-1 mRNA levels after interferon-gamma stimulation in infected Molt-4 were suppressed. In conclusion, HCV could infect and transiently replicate in T cells and that HCV replication suppressed the IFN-gamma/STAT-1/T-bet signaling due to the reduction of STAT-1 and inhibition of its activation (phosphorylation).


Subject(s)
Hepacivirus/growth & development , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/virology , Interferon-gamma/metabolism , Cells, Cultured , Down-Regulation , Humans , Interferon-gamma/pharmacology , Phosphorylation , Recombinant Proteins , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , T-Box Domain Proteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Virus Cultivation
6.
Hepatology ; 44(4): 936-44, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17006910

ABSTRACT

Hepatitis C virus (HCV) infection causes acute and chronic liver disease often leading to liver cirrhosis and hepatocellular carcinoma. Numerous studies have shown that despite induction of virus specific immunity, a curative response is often not attained; this has led to the hypothesis that HCV genes modulate immunity, thereby enabling chronic infections. This study examined the effects on immune-mediated liver injury in transgenic mice expressing core protein throughout the body and bone marrow chimeras expressing core protein in either the lymphoid compartment or liver parenchyma. Presence of core protein in the liver parenchyma but not in lymphoid cells protects from autoimmune hepatitis induced by mitogen concanavalin A (ConA). Consistent with this observation, core transgenic hepatocytes are relatively resistant to death induced by anti-Fas antibody and tumor necrosis factor alpha (TNFalpha). This protective effect is associated with preferential activation of signal transducer and activation of transcription factor 3 (STAT3) versus STAT1 in livers of ConA-injected animals. In agreement with this effect of core protein on the Janus kinase (JAK)-STAT signaling pathway, transgenic mice accelerate liver regeneration after partial hepatectomy but are not protected from hepatocyte death. In conclusion, HCV core inhibits STAT1 and stimulates STAT3 activation, which protects infected hepatocytes from attack by the cell-mediated immune system and promotes their proliferation.


Subject(s)
Apoptosis/drug effects , Hepacivirus/genetics , Hepatitis, Autoimmune/virology , Hepatocytes/metabolism , Liver Regeneration/physiology , Viral Core Proteins/genetics , Alanine Transaminase/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Murine-Derived , Caspase 3 , Caspases/metabolism , Chimera , Concanavalin A , Female , Hepacivirus/immunology , Hepatitis, Autoimmune/etiology , Hepatitis, Autoimmune/physiopathology , Hepatocytes/drug effects , Hepatocytes/virology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/pharmacology , Viral Core Proteins/metabolism
7.
J Virol ; 80(14): 7199-207, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16809325

ABSTRACT

Hepatitis C virus (HCV) infection is frequently associated with the development of hepatocellular carcinomas and non-Hodgkin's B-cell lymphomas. Previously, we reported that HCV infection causes cellular DNA damage and mutations, which are mediated by nitric oxide (NO). NO often damages mitochondria, leading to induction of double-stranded DNA breaks (DSBs) and accumulation of oxidative DNA damage. Here we report that HCV infection causes production of reactive oxygen species (ROS) and lowering of mitochondrial transmembrane potential (DeltaPsi(m)) in in vitro HCV-infected cell cultures. The changes in membrane potential could be inhibited by BCL-2. Furthermore, an inhibitor of ROS production, antioxidant N-acetyl-L-cysteine (NAC), or an inhibitor of NO, 1,400W, prevented the alterations of DeltaPsi(m). The HCV-induced DSB was also abolished by a combination of NO and ROS inhibitors. These results indicated that the mitochondrial damage and DSBs in HCV-infected cells were mediated by both NO and ROS. Among the HCV proteins, core, E1, and NS3 are potent ROS inducers: their expression led to DNA damage and activation of STAT3. Correspondingly, core-protein-transgenic mice showed elevated levels of lipid peroxidation and oxidatively damaged DNA. These HCV studies thus identified ROS, along with the previously identified NO, as the primary inducers of DSBs and mitochondrial damage in HCV-infected cells.


Subject(s)
DNA Damage , Hepacivirus/metabolism , Hepatitis C/metabolism , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/virology , Cell Line , DNA, Mitochondrial/metabolism , Humans , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/virology , Membrane Potentials/drug effects , Mice , Mice, Transgenic , Mitochondria/virology , Nitric Oxide/metabolism , Permeability/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Serine/analogs & derivatives , Serine/pharmacology , Viral Envelope Proteins/metabolism , Viral Nonstructural Proteins/metabolism
8.
J Virol ; 80(2): 866-74, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16378988

ABSTRACT

Hepatitis C virus (HCV) induces inflammatory signals, leading to hepatitis, hepatocellular carcinomas, and lymphomas. The mechanism of HCV involvement in the host's innate immune responses has not been well characterized. In this study, we analyzed expression and regulation of the entire panel of toll-like receptors (TLRs) in human B cells following HCV infection in vitro. Among all of the TLRs (TLRs 1 to 10) examined, only TLR4 showed an altered expression (a three- to sevenfold up-regulation) after HCV infection. Peripheral blood mononuclear cells from HCV-infected individuals also showed a higher expression level of TLR4 compared with those of healthy individuals. HCV infection significantly increased beta interferon (IFN-beta) and interleukin-6 (IL-6) secretion from B cells, particularly after lipopolysaccharide stimulation. The increased IFN-beta and IL-6 production was mediated by TLR4 induction, since the introduction of the small interfering RNA against TLR4 specifically inhibited the HCV-induced cytokine production. Among all of the viral proteins, only NS5A caused TLR4 induction in hepatocytes and B cells. NS5A specifically activated the promoter of the TLR4 gene in both hepatocytes and B cells. In conclusion, HCV infection directly induces TLR4 expression and thereby activates B cells, which may contribute to the host's innate immune responses.


Subject(s)
Hepacivirus/immunology , Hepatitis C/immunology , Hepatitis C/metabolism , Interferon-beta/biosynthesis , Interleukin-6/biosynthesis , Toll-Like Receptor 4/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Cell Line, Tumor , Hepacivirus/chemistry , Hepatitis C/virology , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Leukocytes, Mononuclear , Promoter Regions, Genetic , Toll-Like Receptor 4/genetics , Up-Regulation , Viral Nonstructural Proteins/immunology
9.
J Virol ; 79(13): 8079-89, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15956553

ABSTRACT

Hepatitis C virus (HCV) is one of the leading causes of chronic liver diseases and B-lymphocyte proliferative disorders, including mixed cryoglobulinemia and B-cell lymphoma. It has been suggested that HCV infects human cells through the interaction of its envelope glycoprotein E2 with a tetraspanin molecule CD81, the putative viral receptor. Here, we show that the engagement of B cells by purified E2 induced double-strand DNA breaks specifically in the variable region of immunoglobulin (V(H)) gene locus, leading to hypermutation in the V(H) genes of B cells. Other gene loci were not affected. Preincubation with the anti-CD81 monoclonal antibody blocked this effect. E2-CD81 interaction on B cells triggered the enhanced expression of activation-induced cytidine deaminase (AID) and also stimulated the production of tumor necrosis factor alpha. Knockdown of AID by the specific small interfering RNA blocked the E2-induced double-strand DNA breaks and hypermutation of the V(H) gene. These findings suggest that HCV infection, through E2-CD81 interaction, may modulate host's innate or adaptive immune response by activation of AID and hypermutation of immunoglobulin gene in B cells, leading to HCV-associated B-cell lymphoproliferative diseases.


Subject(s)
Antigens, CD/physiology , B-Lymphocytes/immunology , Genes, Immunoglobulin , Viral Envelope Proteins/physiology , Carcinoma, Hepatocellular , Cell Line, Tumor , Hepacivirus , Humans , Liver Neoplasms , Reverse Transcriptase Polymerase Chain Reaction , Tetraspanin 28
10.
J Virol ; 78(16): 8835-43, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15280491

ABSTRACT

Hepatitis C virus (HCV) infection causes hepatitis, hepatocellular carcinoma, and B-cell lymphomas in a significant number of patients. Previously we have shown that HCV infection causes double-stranded DNA breaks and enhances the mutation frequency of cellular genes, including proto-oncogenes and immunoglobulin genes. To determine the mechanisms, we studied in vitro HCV infection of cell culture. Here we report that HCV infection activated the immunologic (type II) isoform of nitric oxide (NO) synthase (NOS), i.e., inducible NOS (iNOS), thereby inducing NO, which in turn induced DNA breaks and enhanced the mutation frequencies of cellular genes. Treatment of HCV-infected cells with NOS inhibitors or small interfering RNA specific for iNOS abolished most of these effects. Expression of the core protein or nonstructural protein 3 (NS3), but not the other viral proteins, in B cells or hepatocytes induced iNOS and DNA breaks, which could be blocked by NOS inhibitors. The core protein also enhanced the mutation frequency of cellular genes in hepatocytes derived from HCV core transgenic mice compared with that in control mice. The iNOS promoter was activated more than fivefold in HCV-infected cells, as revealed by a luciferase reporter assay driven by the iNOS promoter. Similarly, the core and NS3 proteins also induced the same effects. Therefore, we conclude that HCV infection can stimulate the production of NO through activation of the gene for iNOS by the viral core and NS3 proteins. NO causes DNA breaks and enhances DNA mutation. This sequence of events provides a mechanism for HCV pathogenesis and oncogenesis.


Subject(s)
DNA Damage , Hepacivirus/pathogenicity , Mutation , Nitric Oxide Synthase/metabolism , Nitric Oxide/biosynthesis , Tumor Suppressor Protein p53/metabolism , Animals , B-Lymphocytes/virology , Cell Line , DNA Damage/drug effects , Hepatocytes/virology , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Nitric Oxide/pharmacology , Nitric Oxide Synthase Type II , Tumor Suppressor Protein p53/genetics , Viral Core Proteins/metabolism , Viral Nonstructural Proteins/metabolism
11.
Virology ; 324(2): 450-61, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15207630

ABSTRACT

The mechanism and machinery of hepatitis C virus (HCV) RNA replication are still poorly characterized. Our previous study has shown that HCV RNA synthesis occurs on a lipid raft membrane structure [J. Virol. 77 (2003) 77 4160]. In this study, we further characterized these replication complexes (RCs) in Huh-7 cells that support active RNA replication of a subgenomic HCV replicon. Biochemical analysis showed that these membrane structures were resistant to Nonidet P-40 or Triton X-100 (TX-100) at 4 degrees C while solubilized by beta-octylglucoside at 4 degrees C or Triton TX-100 at 37 degrees C, characteristic of lipid rafts. Cholesterol sequestration assay further demonstrated the association between HCV nonstructural (NS) proteins and cholesterol-rich lipid rafts. The RCs contained both minus- and plus-strand HCV RNA, with the plus-stranded RNA being approximately 10-fold more abundant than the minus-strand. Furthermore, the HCV RNA and NS proteins were resistant to RNase and protease digestion, respectively, but became sensitive after treatment with the raft-disrupting agents. These results suggested that the HCV RCs are protected within lipid rafts. Detergent-resistant membrane (DRM) fractions containing NS proteins and viral RNA were capable of HCV RNA synthesis using the endogenous HCV RNA template. NS proteins were distributed in both the ER and the Golgi, but the majority of the active RCs were detected in the Golgi-derived membrane. Depletion of cellular cholesterol selectively reduced HCV RNA replication. These findings provide further insights into the mechanism of HCV replication in vivo.


Subject(s)
Hepacivirus/physiology , Membrane Microdomains/virology , RNA, Viral/biosynthesis , Cell Line, Tumor , Cholesterol/chemistry , Detergents , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Hepacivirus/genetics , Humans , Membrane Microdomains/chemistry , Membrane Microdomains/metabolism , RNA, Viral/metabolism , Replicon , Viral Nonstructural Proteins/metabolism
12.
J Virol ; 78(13): 7257-63, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15194801

ABSTRACT

Human antibodies elicited in response to hepatitis C virus (HCV) infection are anticipated to react with the native conformation of the viral envelope structure. Isolation of these antibodies as human monoclonal antibodies that block virus binding and entry will be useful in providing potential therapeutic reagents and for vaccine development. H-111, an antibody to HCV envelope 1 protein (E1) that maps to the YEVRNVSGVYH sequence and is located near the N terminus of E1 and is able to immunoprecipitate E1E2 heterodimers, is described. Binding of H-111 to HCV E1 genotypes 1a, 1b, 2b, and 3a indicates that the H-111 epitope is highly conserved. Sequence analysis of antibody V regions showed evidence of somatic and affinity maturation of H-111. Finally, H-111 blocks HCV-like particle binding to and HCV virion infection of target cells, suggesting the involvement of this epitope in virus binding and entry.


Subject(s)
Antibodies, Monoclonal/immunology , Hepacivirus/immunology , Hepacivirus/pathogenicity , Hepatitis C Antibodies/immunology , Viral Envelope Proteins/immunology , Amino Acid Sequence , Antibodies, Monoclonal/biosynthesis , B-Lymphocytes , Cell Line , Epitope Mapping , Hepacivirus/physiology , Hepatitis C Antibodies/biosynthesis , Humans , Neutralization Tests , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
13.
Proc Natl Acad Sci U S A ; 101(12): 4262-7, 2004 Mar 23.
Article in English | MEDLINE | ID: mdl-14999097

ABSTRACT

Hepatitis C virus (HCV) is a nonretroviral oncogenic RNA virus, which is frequently associated with hepatocellular carcinoma (HCC) and B cell lymphoma. We demonstrated here that acute and chronic HCV infection caused a 5- to 10-fold increase in mutation frequency in Ig heavy chain, BCL-6, p53, and beta-catenin genes of in vitro HCV-infected B cell lines and HCV-associated peripheral blood mononuclear cells, lymphomas, and HCCs. The nucleotide-substitution pattern of p53 and beta-catenin was different from that of Ig heavy chain in HCV-infected cells, suggesting two different mechanisms of mutation. In addition, the mutated protooncogenes were amplified in HCV-associated lymphomas and HCCs, but not in lymphomas of nonviral origin or HBV-associated HCC. HCV induced error-prone DNA polymerase zeta, polymerase iota, and activation-induced cytidine deaminase, which together, contributed to the enhancement of mutation frequency, as demonstrated by the RNA interference experiments. These results indicate that HCV induces a mutator phenotype and may transform cells by a hit-and-run mechanism. This finding provides a mechanism of oncogenesis for an RNA virus.


Subject(s)
Hepacivirus/metabolism , Hepatitis C/metabolism , Immunoglobulins/genetics , Proto-Oncogenes , B-Lymphocytes/metabolism , Cytidine Deaminase/metabolism , DNA Damage , DNA-Directed DNA Polymerase/metabolism , Hepatitis C/genetics , Humans , Mutation , RNA, Small Interfering
14.
J Virol ; 77(3): 2134-46, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12525648

ABSTRACT

Hepatitis C virus (HCV) is a major cause of chronic hepatitis, liver cirrhosis, and hepatocellular carcinoma. Studies of HCV replication and pathogenesis have so far been hampered by the lack of an efficient tissue culture system for propagating HCV in vitro. Although HCV is primarily a hepatotropic virus, an increasing body of evidence suggests that HCV also replicates in extrahepatic tissues in natural infection. In this study, we established a B-cell line (SB) from an HCV-infected non-Hodgkin's B-cell lymphoma. HCV RNA and proteins were detectable by RNase protection assay and immunoblotting. The cell line continuously produces infectious HCV virions in culture. The virus particles produced from the culture had a buoyant density of 1.13 to 1.15 g/ml in sucrose and could infect primary human hepatocytes, peripheral blood mononuclear cells (PBMCs), and an established B-cell line (Raji cells) in vitro. The virus from SB cells belongs to genotype 2b. Single-stranded conformational polymorphism and sequence analysis of the viral RNA quasispecies indicated that the virus present in SB cells most likely originated from the patient's spleen and had an HCV RNA quasispecies pattern distinct from that in the serum. The virus production from the infected primary hepatocytes showed cyclic variations. In addition, we have succeeded in establishing several Epstein-Barr virus-immortalized B-cell lines from PBMCs of HCV-positive patients. Two of these cell lines are positive for HCV RNA as detected by reverse transcriptase PCR and for the nonstructural protein NS3 by immunofluorescence staining. These observations unequivocally establish that HCV infects B cells in vivo and in vitro. HCV-infected cell lines show significantly enhanced apoptosis. These B-cell lines provide a reproducible cell culture system for studying the complete replication cycle and biology of HCV infections.


Subject(s)
Apoptosis , Hepacivirus/physiology , Lymphoma, B-Cell/virology , Amino Acid Sequence , Base Sequence , Hepatocytes/virology , Herpesvirus 4, Human/genetics , Humans , Lymphoma, B-Cell/pathology , Molecular Sequence Data , RNA, Viral/blood , Tumor Cells, Cultured , Viral Nonstructural Proteins/analysis , Virion/physiology
15.
J Virol ; 76(2): 912-7, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11752180

ABSTRACT

We have developed a system for producing murine leukemia virus (MLV) pseudotyped with human hepatitis B virus (HBV) large (L) and small (S) surface antigens (HBsAg) for targeting primary human hepatocytes. Using the MLV(HBV) pseudotype virus containing a beta-galactosidase reporter gene, we demonstrated that this pseudotype virus exhibits strict tropism for primary human hepatocytes, similar to the natural target cell specificity of HBV. It does not infect any of the established tissue culture cell lines, including human hepatoma cell lines (HepG2 and Huh-7), or rat primary hepatocytes. The infectivity of MLV(HBV) for human hepatocytes was inhibited by anti-HBs antibody. The L form of HBsAg was both necessary and sufficient for virus infectivity, but the presence of both L and S forms enhanced the surface expression of HBsAg and thus increased virus production. The middle form of HBsAg was not necessary. This pseudotype virus bypasses the requirement for the liver-specific transcription factors for HBV replication, enabling direct study of HBV tissue tropism conferred by the viral envelope proteins. This virus also offers a potential liver-specific targeting system for gene therapy.


Subject(s)
Hepatitis B Surface Antigens/immunology , Hepatitis B virus/immunology , Hepatocytes/virology , Mice/virology , Retroviridae/genetics , Animals , Cells, Cultured , Genetic Vectors/genetics , Hepatitis B Surface Antigens/genetics , Hepatitis B virus/genetics , Hepatocytes/metabolism , Humans , Rats , Retroviridae/chemistry , Retroviridae/physiology , Sensitivity and Specificity , Species Specificity , Transcription Factors/metabolism , Tumor Cells, Cultured , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL