Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunol ; 185(11): 6985-98, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20974981

ABSTRACT

Hepatitis C virus (HCV) infection is associated with the development of hepatocellular carcinoma and putatively also non-Hodgkin's B cell lymphoma. In this study, we demonstrated that PBMCs obtained from HCV-infected patients showed frequent chromosomal aberrations and that HCV infection of B cells in vitro induced enhanced chromosomal breaks and sister chromatid exchanges. HCV infection hypersensitized cells to ionizing radiation and bleomycin and inhibited nonhomologous end-joining repair. The viral core and nonstructural protein 3 proteins were shown to be responsible for the inhibition of DNA repair, mediated by NO and reactive oxygen species. Stable expression of core protein induced frequent chromosome translocations in cultured cells and in transgenic mice. HCV core protein binds to the NBS1 protein and inhibits the formation of the Mre11/NBS1/Rad50 complex, thereby affecting ATM activation and inhibiting DNA binding of repair enzymes. Taken together, these data indicate that HCV infection inhibits multiple DNA repair processes to potentiate chromosome instability in both monocytes and hepatocytes. These effects may explain the oncogenicity and immunological perturbation of HCV infection.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , DNA Damage/immunology , DNA Repair/immunology , DNA-Binding Proteins/antagonists & inhibitors , Hepacivirus/immunology , Hepatocytes/immunology , Monocytes/immunology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Reactive Nitrogen Species/physiology , Reactive Oxygen Species/pharmacology , Tumor Suppressor Proteins/antagonists & inhibitors , Acid Anhydride Hydrolases , Animals , Ataxia Telangiectasia/enzymology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Line, Transformed , Cell Line, Tumor , Cells, Cultured , DNA Repair Enzymes/antagonists & inhibitors , DNA Repair Enzymes/physiology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , HEK293 Cells , Hep G2 Cells , Hepatocytes/metabolism , Hepatocytes/virology , Humans , MRE11 Homologue Protein , Mice , Mice, Transgenic , Monocytes/metabolism , Monocytes/virology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Protein Binding/immunology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/immunology , Tumor Suppressor Proteins/metabolism , Viral Core Proteins/metabolism
2.
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
3.
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
4.
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
5.
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
6.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...