Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
PLoS Pathog ; 20(3): e1012060, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38442126

ABSTRACT

The recent discovery of Hepatitis D (HDV)-like viruses across a wide range of taxa led to the establishment of the Kolmioviridae family. Recent studies suggest that kolmiovirids can be satellites of viruses other than Hepatitis B virus (HBV), challenging the strict HBV/HDV-association dogma. Studying whether kolmiovirids are able to replicate in any animal cell they enter is essential to assess their zoonotic potential. Here, we compared replication of three kolmiovirids: HDV, rodent (RDeV) and snake (SDeV) deltavirus in vitro and in vivo. We show that SDeV has the narrowest and RDeV the broadest host cell range. High resolution imaging of cells persistently replicating these viruses revealed nuclear viral hubs with a peculiar RNA-protein organization. Finally, in vivo hydrodynamic delivery of viral replicons showed that both HDV and RDeV, but not SDeV, efficiently replicate in mouse liver, forming massive nuclear viral hubs. Our comparative analysis lays the foundation for the discovery of specific host factors controlling Kolmioviridae host-shifting.


Subject(s)
Hepatitis D , Hepatitis Delta Virus , Mice , Animals , Humans , Rodentia , Hepatitis B virus/genetics , Snakes , Virus Replication , RNA, Viral/genetics
2.
BMC Cancer ; 22(1): 428, 2022 Apr 20.
Article in English | MEDLINE | ID: mdl-35443621

ABSTRACT

BACKGROUND: To evaluate the effect of regorafenib on soluble MHC class I polypeptide-related sequence A (MICA) (sMICA) level in vitro. In addition, we clinically examined whether its plasma levels were associated with regorafenib activity in terms of progression-free survival (PFS) in patients with CRC. METHODS: Human CRC cell line HCT116 and HT29 cells were treated with regorafenib and its pharmacologically active metabolites, M2 or M5 at the same concentrations as those in sera of patients. We also examined the sMICA levels and the area under the plasma concentration-time curve of regorafenib, M2 and M5. RESULTS: Regorafenib, M2, and M5 significantly suppressed shedding of MICA in human CRC cells without toxicity. This resulted in the reduced production of sMICA. In the clinical examination, patients with CRC who showed long median PFS (3.7 months) had significantly lower sMICA levels than those with shorter median PFS (1.2 months) (p = 0.045). CONCLUSIONS: MICA is an attractive agent for manipulating the immunological control of CRC and baseline sMICA levels could be a predictive biomarker for the efficacy of regorafenib treatment.


Subject(s)
Colorectal Neoplasms , Histocompatibility Antigens Class I , Biomarkers , Colorectal Neoplasms/drug therapy , Humans , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Pyridines
3.
Gut ; 70(1): 157-169, 2021 01.
Article in English | MEDLINE | ID: mdl-32217639

ABSTRACT

OBJECTIVE: Hepatocellular carcinoma (HCC) is the fastest-growing cause of cancer-related mortality with chronic viral hepatitis and non-alcoholic steatohepatitis (NASH) as major aetiologies. Treatment options for HCC are unsatisfactory and chemopreventive approaches are absent. Chronic hepatitis C (CHC) results in epigenetic alterations driving HCC risk and persisting following cure. Here, we aimed to investigate epigenetic modifications as targets for liver cancer chemoprevention. DESIGN: Liver tissues from patients with NASH and CHC were analysed by ChIP-Seq (H3K27ac) and RNA-Seq. The liver disease-specific epigenetic and transcriptional reprogramming in patients was modelled in a liver cell culture system. Perturbation studies combined with a targeted small molecule screen followed by in vivo and ex vivo validation were used to identify chromatin modifiers and readers for HCC chemoprevention. RESULTS: In patients, CHC and NASH share similar epigenetic and transcriptomic modifications driving cancer risk. Using a cell-based system modelling epigenetic modifications in patients, we identified chromatin readers as targets to revert liver gene transcription driving clinical HCC risk. Proof-of-concept studies in a NASH-HCC mouse model showed that the pharmacological inhibition of chromatin reader bromodomain 4 inhibited liver disease progression and hepatocarcinogenesis by restoring transcriptional reprogramming of the genes that were epigenetically altered in patients. CONCLUSION: Our results unravel the functional relevance of metabolic and virus-induced epigenetic alterations for pathogenesis of HCC development and identify chromatin readers as targets for chemoprevention in patients with chronic liver diseases.


Subject(s)
Carcinoma, Hepatocellular/prevention & control , Epigenesis, Genetic , Hepatitis C, Chronic/complications , Liver Neoplasms/prevention & control , Non-alcoholic Fatty Liver Disease/complications , Animals , Carcinoma, Hepatocellular/etiology , Disease Models, Animal , Hepatitis C, Chronic/genetics , Hepatitis C, Chronic/pathology , Humans , Liver Neoplasms/etiology , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology
4.
Cancer Immunol Immunother ; 70(1): 203-213, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32683508

ABSTRACT

In our previous genome-wide association study, we demonstrated the association between MHC class I-related chain A (MICA) and hepatocellular carcinoma (HCC) development in patients with chronic hepatitis C. Increasing membrane-bound MICA (mMICA) in cancer cells by reducing MICA sheddases facilitates natural killer (NK) cell-mediated cytotoxicity. Our recent study clarified that A disintegrin and metalloproteases (ADAM), including ADAM9, are MICA sheddases in HCC, and that the suppression of ADAMs increases mMICA, demonstrating the rationality of mMICA-NK targeted therapy. Furthermore, we showed that regorafenib suppresses ADAM9 transcriptionally and translationally. A library of FDA-approved drugs was screened for more efficient inhibitors of ADAM9. Flow cytometry evaluation of the expression of mMICA after treatment with various candidate drugs identified leukotriene receptor antagonists as potential ADAM9 inhibitors. Furthermore, leukotriene receptor antagonists alone or in combination with regorafenib upregulated mMICA, which was in turn downregulated by leukotriene C4 and D4 via ADAM9 function. Our study demonstrates that leukotriene receptor antagonists could be developed as novel drugs for immunological control and suppression of ADAM9 in HCC. Further, leukotriene receptor antagonists should be explored as combination therapy partners with conventional multi-kinase inhibitors for developing therapeutic strategies with enhanced efficacies for HCC management and treatment.


Subject(s)
ADAM Proteins/metabolism , Carcinoma, Hepatocellular/drug therapy , Histocompatibility Antigens Class I/metabolism , Leukotriene Antagonists/pharmacology , Liver Neoplasms/drug therapy , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genome-Wide Association Study/methods , Hep G2 Cells , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Liver Neoplasms/metabolism , Membrane Proteins/metabolism , Phenylurea Compounds/pharmacology , Pyridines/pharmacology , Treatment Outcome , Up-Regulation/drug effects
5.
Int J Mol Sci ; 21(16)2020 Aug 07.
Article in English | MEDLINE | ID: mdl-32784555

ABSTRACT

Hepatitis B virus (HBV), a highly persistent pathogen causing hepatocellular carcinoma (HCC), takes full advantage of host machinery, presenting therapeutic targets. Here we aimed to identify novel druggable host cellular factors using the reporter HBV we have recently generated. In an RNAi screen of G protein-coupled receptors (GPCRs), GPCR39 (GPR39) appeared as the top hit to facilitate HBV proliferation. Lentiviral overexpression of active GPR39 proteins and an agonist enhanced HBV replication and transcriptional activities of viral promoters, inducing the expression of CCAAT/enhancer binding protein (CEBP)-ß (CEBPB). Meanwhile, GPR39 was uncovered to activate the heat shock response, upregulating the expression of proviral heat shock proteins (HSPs). In addition, glioma-associated oncogene homologue signaling, a recently reported target of GPR39, was suggested to inhibit HBV replication and eventually suppress expression of CEBPB and HSPs. Thus, GPR39 provirally governed intracellular circuits simultaneously affecting the carcinopathogenetic gene functions. GPR39 and the regulated signaling networks would serve as antiviral targets, and strategies with selective inhibitors of GPR39 functions can develop host-targeted antiviral therapies preventing HCC.


Subject(s)
Hepatitis B virus/physiology , Receptors, G-Protein-Coupled/metabolism , Virus Replication , CCAAT-Enhancer-Binding Protein-beta/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Response , Hedgehog Proteins/metabolism , Hep G2 Cells , Humans , Models, Biological , RNA Interference , Signal Transduction
6.
Int J Mol Sci ; 21(9)2020 Apr 26.
Article in English | MEDLINE | ID: mdl-32357520

ABSTRACT

Chronic infection with hepatitis C virus (HCV) is a major cause of hepatocellular carcinoma (HCC). Novel treatments with direct-acting antivirals achieve high rates of sustained virologic response; however, the HCC risk remains elevated in cured patients, especially those with advanced liver disease. Long-term HCV infection causes a persistent and accumulating damage of the liver due to a combination of direct and indirect pro-oncogenic mechanisms. This review describes the processes involved in virus-induced disease progression by viral proteins, derailed signaling, immunity, and persistent epigenetic deregulation, which may be instrumental to develop urgently needed prognostic biomarkers and as targets for novel chemopreventive therapies.


Subject(s)
Carcinoma, Hepatocellular/virology , Hepacivirus/metabolism , Hepatitis C, Chronic/complications , Liver Neoplasms/virology , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Carcinoma, Hepatocellular/genetics , Disease Progression , Epigenesis, Genetic , Hepacivirus/drug effects , Hepatitis C, Chronic/drug therapy , Humans , Liver Neoplasms/genetics , Tumor Escape
7.
Int J Cancer ; 143(10): 2575-2583, 2018 11 15.
Article in English | MEDLINE | ID: mdl-29873070

ABSTRACT

In our previous study on hepatocellular carcinoma (HCC) susceptibility genes in chronic hepatitis patients, we identified the MHC class I polypeptide-related sequence A (MICA). Natural killer cells eliminate various cancer cells, including HCC, by suppressing MICA shedding. Therefore, we investigated MICA sheddases and inhibitors for HCC immunotherapy. In this study, HepG2, PLC/PRF/5, and Hep3B were treated with the siRNA of a disintegrin and metalloproteases (ADAMs) and matrix metalloproteases to measure the concentration of soluble MICA (sMICA) by ELISA to detect the therapeutic target. Furthermore, an FDA-approved drug library was tested for the enzymatic inhibition of the targeted enzyme in an in vitro drug screening assay system. ADAM17 knockdown reduced sMICA levels and increased membrane-bound MICA (mMICA) expression in HCC cells. In an in vitro drug screen using an FDA-approved drug library, lomofungin, an antifungal drug, was found to strongly decrease ADAM17 activity. In HCC cells, mMICA expression was induced and sMICA production was inhibited in a dose-dependent manner. These effects were cancelled upon ADAM17 knockdown, suggesting that lomofungin targeted ADAM17. Analysis of lomofungin analogs revealed the responsible functional groups. In summary, we suggest lomofungin to be an attractive agent for the immunological control of HCC, via the suppression of ADAM17.


Subject(s)
ADAM17 Protein/antagonists & inhibitors , Carcinoma, Hepatocellular/drug therapy , Histocompatibility Antigens Class I/metabolism , Liver Neoplasms/drug therapy , Phenazines/pharmacology , ADAM17 Protein/immunology , ADAM17 Protein/metabolism , ADAM17 Protein/pharmacology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Hep G2 Cells , Histocompatibility Antigens Class I/immunology , Humans , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Recombinant Proteins/pharmacology
8.
J Gastroenterol Hepatol ; 33(5): 1075-1081, 2018 May.
Article in English | MEDLINE | ID: mdl-29055152

ABSTRACT

BACKGROUND AND AIM: The multi-kinase inhibitor regorafenib (REG) was recently demonstrated to be effective in patients with sorafenib (SOR)-resistant hepatocellular carcinoma (HCC). Interestingly, SOR is known to enhance the accumulation of membrane-bound MHC class I polypeptide-related sequence A (mMICA) in HCC cells and to block the production of soluble MICA (sMICA), an immunological decoy. In addition, MICA is associated with HCC in patients with chronic hepatitis C. We have now compared the impact of REG and SOR on MICA in HCC cells, as well as the immunotherapeutic implications thereof. METHODS: HepG2 and PLC/PRF/5 cells were exposed to REG and SOR, and levels of sMICA and mMICA were measured by ELISA and flow cytometry, respectively. The drugs were also tested in vitro for inhibitory activity against recombinant human A disintegrin and metalloprotease 9 (ADAM9), a sheddase that releases MICA from the membrane. RESULTS: To a greater extent than SOR, but without marked difference in cytotoxicity, REG significantly suppressed mRNA and protein expression of ADAM9 and ADAM10, thereby decreasing production of sMICA and boosting accumulation of mMICA. Accumulation of mMICA in response to REG was reversed by siRNA against ADAM9. However, the drugs did not inhibit the enzymatic activity of ADAM9 in vitro. CONCLUSIONS: The clinical superiority of REG over SOR is partially attributable to reduced MICA shedding via transcriptional suppression of ADAM9 and ADAM10.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Histocompatibility Antigens Class I/metabolism , Liver Neoplasms/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Pyridines/pharmacology , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Carcinoma, Hepatocellular/complications , Depression, Chemical , Gene Expression/drug effects , Hep G2 Cells , Hepatitis C, Chronic/complications , Humans , Liver Neoplasms/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Niacinamide/pharmacology , RNA, Messenger/metabolism , Solubility , Sorafenib
9.
J Hepatol ; 66(6): 1223-1230, 2017 06.
Article in English | MEDLINE | ID: mdl-28192189

ABSTRACT

BACKGROUND & AIMS: Primary biliary cholangitis (PBC) is an autoimmune liver disease of unknown pathogenesis. Consequently, therapeutic targets for PBC have yet to be identified. CD4+ T cells play a pivotal role in immunological dysfunction observed in PBC, and therefore, microRNA (miRNA) and mRNA expression were analysed in CD4+ T cells, to investigate PBC pathogenesis and identify novel therapeutic targets. METHODS: Integral miRNA and mRNA analysis of 14 PBC patients and ten healthy controls was carried out using microarray and quantitative real-time polymerase chain reaction (qRT-PCR), with gene set enrichment analysis. The functional analyses of miRNA were then assessed using reporter and miRNA-overexpression assays. RESULTS: The integral analysis of miRNA and mRNA identified four significantly downregulated miRNAs (miR-181a, -181b, -374b, and -425) related to the T cell receptor (TCR) signalling pathway in CD4+ T cells of PBC. N-Ras, a regulator of the TCR signalling pathway, was found to be targeted by all four identified miRNAs. In addition, in vitro assays confirmed that decreased miR-425 strongly induced inflammatory cytokines (interleukin [IL]-2 and interferon [IFN]-γ) via N-Ras upregulation in the TCR signalling pathway. CONCLUSION: The decreased expression of four miRNAs that dysregulate TCR signalling in PBC CD4+ T cells was identified. miR-425 was demonstrated as an inflammatory regulator of PBC via N-Ras upregulation. Therefore, the restoration of decreased miR-425 or the suppression of N-Ras may be a promising immunotherapeutic strategy against PBC. LAY SUMMARY: Primary biliary cholangitis (PBC) is an autoimmune liver disease, but the causes are unknown. MicroRNAs are molecules known to regulate biological signals. In this study, four microRNAs were identified as being decreased in PBC patients, leading to activation of T cell receptor signalling pathways, involved in inflammation. One particular target, N-Ras, could be an attractive and novel immunotherapeutic option for PBC. TRANSCRIPT PROFILING: Microarray data are deposited in GEO (GEO accession: GSE93172).


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Genes, ras , Liver Cirrhosis, Biliary/genetics , Liver Cirrhosis, Biliary/immunology , MicroRNAs/genetics , Aged , CD4-Positive T-Lymphocytes/metabolism , Case-Control Studies , Cytokines/genetics , Cytokines/metabolism , Farnesol/analogs & derivatives , Farnesol/pharmacology , Gene Expression Profiling , Humans , Inflammation Mediators/metabolism , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interleukin-2/biosynthesis , Interleukin-2/genetics , Jurkat Cells , Liver Cirrhosis, Biliary/metabolism , MicroRNAs/metabolism , Middle Aged , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell/metabolism , Salicylates/pharmacology , Signal Transduction/genetics , Signal Transduction/immunology , Up-Regulation
10.
Hepatology ; 59(4): 1250-61, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23913866

ABSTRACT

UNLABELLED: Several genome-wide association studies (GWAS) have identified a genetic polymorphism associated with the gene locus for interleukin 28B (IL28B), a type III interferon (IFN), as a major predictor of clinical outcome in hepatitis C. Antiviral effects of the type III IFN family have previously been shown against several viruses, including hepatitis C virus (HCV), and resemble the function of type I IFN including utilization of the intracellular Janus kinase signal transducer and activator of transcription (JAK-STAT) pathway. Effects unique to IL28B that would distinguish it from IFN-α are not well defined. By analyzing the transcriptomes of primary human hepatocytes (PHH) treated with IFN-α or IL28B, we sought to identify functional differences between IFN-α and IL28B to better understand the roles of these cytokines in the innate immune response. Although our data did not reveal distinct gene signatures, we detected striking kinetic differences between IFN-α and IL28B stimulation for interferon stimulated genes (ISGs). While gene induction was rapid and peaked at 8 hours of stimulation with IFN-α in PHH, IL28B produced a slower, but more sustained increase in gene expression. We confirmed these findings in the human hepatoma cell line Huh7.5.1. Interestingly, in HCV-infected cells the rapid response after stimulation with IFN-α was blunted, and the induction pattern resembled that caused by IL28B. CONCLUSION: The kinetics of gene induction are fundamentally different for stimulations with either IFN-α or IL28B in hepatocytes, suggesting distinct roles of these cytokines within the immune response. Furthermore, the observed differences are substantially altered by infection with HCV.


Subject(s)
Carcinoma, Hepatocellular/epidemiology , Gene Expression Regulation, Neoplastic/drug effects , Hepatitis C/epidemiology , Hepatocytes/metabolism , Interferon-alpha/pharmacology , Interleukins/pharmacology , Liver Neoplasms/epidemiology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Comorbidity , Dose-Response Relationship, Drug , Hepatitis C/metabolism , Hepatitis C/pathology , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Interferons , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Phosphorylation , STAT1 Transcription Factor/metabolism , Time Factors , Transcriptome/drug effects
11.
Nihon Rinsho ; 73(2): 333-8, 2015 Feb.
Article in Japanese | MEDLINE | ID: mdl-25764692

ABSTRACT

Hepatitis C virus (HCV) infection is a major risk factor for developing hepatocellular carcinoma (HCC). The host genetic factors involved in the development of HCC in patients with HCV infection were investigated. To identify the genetic susceptibility factors for HCV-induced HCC, genome wide association studies (GWAS) were conducted in HCV-induced HCC cases and controls of Japanese origin. Single nucleotide polymorphisms (SNPs) which showed possible association in the GWAS were further genotyped using different cohorts. By these analyses, MICA and DEPDC5 SNPs were found to be strongly associated with HCV-induced HCC. These results highlight the importance of MICA and DEPDC5 genetic variations not only as predictive biomarkers for HCV-induced HCC but also as therapeutic targets against hepatocarcinogenesis or HCC.


Subject(s)
Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/genetics , Hepatitis C/complications , Liver Neoplasms/etiology , Liver Neoplasms/genetics , Asian People , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Humans , Polymorphism, Single Nucleotide
12.
Hepatol Res ; 44(10): E137-44, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24125181

ABSTRACT

AIM: The PNPLA3 rs738409 C>G polymorphism (encoding for I148M) has recently been identified as a susceptibility factor for steatosis-mediated liver damage. We evaluated the influence of this polymorphism on hepatocarcinogenesis in patients with chronic hepatitis C (CHC) virus infection. METHODS: We genotyped the rs738409 single nucleotide polymorphism in 358 hepatitis C-associated hepatocellular carcinoma (HCC) patients and correlated the age at onset of HCC and the interval between hepatitis C virus (HCV) infection and the development of HCC in patients with each genotype. RESULTS: The frequencies of CC, CG and GG genotypes were 27.9% (100/358), 49.2% (176/358) and 22.9% (82/358), respectively, and were in Hardy-Weinberg equilibrium. The median age at onset of HCC for the GG genotype was significantly younger compared to for non-GG genotypes (67.81 vs 69.87 years, P < 0.001), and the median interval between HCV infection and the development of HCC was significantly shorter in patients with the GG genotype (39.96 vs 40.85 years, P = 0.008). PNPLA3 GG genotype was also associated with a higher aspartate aminotransferase level (69.5 vs 59.0 IU/L, P = 0.02), lower prothrombin time (73.0% vs 78.0%, P = 0.008) and a higher prevalence of histological steatosis (40.0% vs. 22.2%, P = 0.01) at the time of HCC onset. CONCLUSION: The PNPLA3 genotype GG may be associated with accelerated hepatocarcinogenesis in CHC patients through increased steatosis in the liver.

13.
J Hepatol ; 59(5): 942-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23831117

ABSTRACT

BACKGROUND & AIMS: Hepatitis C virus (HCV) is a major cause of chronic liver disease worldwide. The biological and therapeutic importance of host cellular cofactors for viral replication has been recently appreciated. Here we examined the roles of SNF1/AMP kinase-related kinase (SNARK) in HCV replication and pathogenesis. METHODS: The JFH1 infection system and the full-length HCV replicon OR6 cell line were used. Gene expression was knocked down by siRNAs. SNARK mutants were created by site-directed mutagenesis. Intracellular mRNA levels were measured by qRT-PCR. Endogenous and overexpressed proteins were detected by Western blot analysis and immunofluorescence. Transforming growth factor (TGF)-ß signaling was monitored by a luciferase reporter construct. Liver biopsy samples from HCV-infected patients were analyzed for SNARK expression. RESULTS: Knockdown of SNARK impaired viral replication, which was rescued by wild type SNARK but not by unphosphorylated or kinase-deficient mutants. Knockdown and overexpression studies demonstrated that SNARK promoted TGF-ß signaling in a manner dependent on both its phosphorylation and kinase activity. In turn, chronic HCV replication upregulated the expression of SNARK in patients. Further, the SNARK kinase inhibitor metformin suppressed both HCV replication and SNARK-mediated enhancement of TGF-ß signaling. CONCLUSIONS: Thus reciprocal regulation between HCV and SNARK promotes TGF-ß signaling, a major driver of hepatic fibrogenesis. These findings suggest that SNARK will be an attractive target for the design of novel host-directed antiviral and antifibrotic drugs.


Subject(s)
Hepacivirus/physiology , Hepatitis C/etiology , Protein Serine-Threonine Kinases/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Virus Replication/physiology , Biopsy , Cell Line , Gene Expression Regulation/drug effects , Hepatitis C/physiopathology , Humans , Liver/pathology , Liver/virology , Metformin/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/pharmacology
14.
J Virol ; 86(16): 8581-91, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22674974

ABSTRACT

Responses to alpha interferon (IFN-α)-based treatment are dependent on both host and viral factors and vary markedly among patients infected with different hepatitis C virus (HCV) genotypes (GTs). Patients infected with GT3 viruses consistently respond better to IFN treatment than do patients infected with GT1 viruses. The mechanisms underlying this difference are not well understood. In this study, we sought to determine the effects of HCV NS5A proteins from different genotypes on IFN signaling. We found that the overexpression of either GT1 or GT3 NS5A proteins significantly inhibited IFN-induced IFN-stimulated response element (ISRE) signaling, phosphorylated STAT1 (P-STAT1) levels, and IFN-stimulated gene (ISG) expression compared to controls. GT1 NS5A protein expression exhibited stronger inhibitory effects on IFN signaling than did GT3 NS5A protein expression. Furthermore, GT1 NS5A bound to STAT1 with a higher affinity than did GT3 NS5A. Domain mapping revealed that the C-terminal region of NS5A conferred these inhibitory effects on IFN signaling. The overexpression of HCV NS5A increased HCV replication levels in JFH1-infected cells through the further reduction of levels of P-STAT1, ISRE signaling, and downstream ISG responses. We demonstrated that the overexpression of GT1 NS5A proteins resulted in less IFN responsiveness than did the expression of GT3 NS5A proteins through stronger binding to STAT1. We confirmed that GT1 NS5A proteins exerted stronger IFN signaling inhibition than did GT3 NS5A proteins in an infectious recombinant JFH1 virus. The potent antiviral NS5A inhibitor BMS-790052 did not block NS5A-mediated IFN signaling suppression in an overexpression model, suggesting that NS5A's contributions to replication are independent of its subversive action on IFN. We propose a model in which the binding of the C-terminal region of NS5A to STAT1 leads to decreased levels of P-STAT1, ISRE signaling, and ISG transcription and, ultimately, to preferential GT1 resistance to IFN treatment.


Subject(s)
Hepacivirus/pathogenicity , Host-Pathogen Interactions , Interferon Type I/immunology , STAT1 Transcription Factor/metabolism , Signal Transduction , Viral Nonstructural Proteins/metabolism , Humans , Models, Biological , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping
15.
Antiviral Res ; 209: 105477, 2023 01.
Article in English | MEDLINE | ID: mdl-36511319

ABSTRACT

Chronic hepatitis D is the most aggressive form of chronic viral hepatitis. It is caused by super-infection of hepatitis B virus (HBV)-infected hepatocytes with hepatitis D virus (HDV). While the recent conditional approval of bulevirtide for HDV treatment offers a new therapeutic modality in Europe, there is an unmet medical need to further improve therapy. A more detailed characterization of virus-host interactions is needed for the identification of novel therapeutic targets. Addressing this need, we engineered a new stably-transformed cell line, named HuH7-2C8D, producing high titer recombinant HDV and allowing the study of viral particles morphogenesis and infectivity. Using this culture system, where viral propagation by re-infection is limited, we observed an increased accumulation of edited version of the viral genomes within secreted HDV viral particles over time that is accompanied with a decrease in viral particle infectivity. We confirmed the interaction of HDV proteins with a previously described host factor in HuH7-2C8D cells and additionally showed that these cells are suitable for co-culture assays with other cell types such as macrophages. Finally, the use of HuH7-2C8D cells allowed to confirm the dual antiviral activity of farnesyl transferase inhibitors, including the clinical candidate lonafarnib, against HDV. In conclusion, we have established an easy-to-handle cell culture model to investigate HDV replication, morphogenesis, and host interactions. HuH7-2C8D cells are also suitable for high-throughput antiviral screening assays for the development of new therapeutic strategies.


Subject(s)
Hepatitis Delta Virus , Virus Replication , Hepatitis Delta Virus/genetics , Cell Line , Hepatitis B virus , Antiviral Agents/pharmacology , Drug Discovery
16.
J Biol Chem ; 286(4): 2665-74, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21098019

ABSTRACT

HIV/HCV coinfection leads to accelerated hepatic fibrosis progression, with higher rates of cirrhosis, liver failure, and liver death than does HCV mono-infection. However, the profibrogenic role of HIV on hepatocytes and hepatic stellate cells (HSC) has not been fully clarified. We hypothesized that HIV, HCV induce liver fibrosis through altered regulation of the production of extracellular matrix and matrix metalloproteinases. We examined the fibrogenesis- and fibrolysis-related gene activity in LX2 HSC and Huh7.5.1 cells in the presence of inactivated CXCR4 and CCR5 HIV, as well as HCV JFH1 virus. The role of reactive oxygen species (ROS) upon fibrosis gene expression was assessed using the ROS inhibitor. Fibrosis-related transcripts including procollagen α1(I) (CoL1A), TIMP1, and MMP3 mRNA were measured by qPCR. TIMP1 and MMP3 protein expression were assessed by ELISA. We found that inactivated CXCR4 HIV and CCR5 HIV increased CoL1A, and TIMP1 expression in both HSC and Huh7.5.1 cells; the addition of JFH1 HCV further increased CoL1A and TIMP1 expression. CXCR4 HIV and CCR5 HIV induced ROS production in HSC and Huh7.5.1 cells which was further enhanced by JFH1 HCV. The ROS inhibitor DPI abrogated HIV-and HCV-induced CoL1A and TIMP1 expression. HIV and HCV-induced CoL1A and TIMP1 expression were also blocked by NFκB siRNA. Our data provide further evidence that HIV and HCV independently regulate hepatic fibrosis progression through the generation of ROS; this regulation occurs in an NFκB-dependent fashion. Strategies to limit the viral induction of oxidative stress are warranted to inhibit fibrogenesis.


Subject(s)
HIV Infections , HIV/metabolism , Hepacivirus/metabolism , Hepatitis C , Liver Cirrhosis , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Gene Expression Regulation , HIV Infections/complications , HIV Infections/metabolism , HIV Infections/pathology , Hepatitis C/complications , Hepatitis C/metabolism , Hepatitis C/pathology , Humans , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/virology , Oxidative Stress
18.
J Hepatol ; 56(2): 326-33, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21888876

ABSTRACT

BACKGROUND & AIMS: The precise mechanisms by which IFN exerts its antiviral effect against HCV have not yet been elucidated. We sought to identify host genes that mediate the antiviral effect of IFN-α by conducting a whole-genome siRNA library screen. METHODS: High throughput screening was performed using an HCV genotype 1b replicon, pRep-Feo. Those pools with replicate robust Z scores ≥2.0 entered secondary validation in full-length OR6 replicon cells. Huh7.5.1 cells infected with JFH1 were then used to validate the rescue efficacy of selected genes for HCV replication under IFN-α treatment. RESULTS: We identified and confirmed 93 human genes involved in the IFN-α anti-HCV effect using a whole-genome siRNA library. Gene ontology analysis revealed that mRNA processing (23 genes, p=2.756e-22), translation initiation (nine genes, p=2.42e-6), and IFN signaling (five genes, p=1.00e-3) were the most enriched functional groups. Nine genes were components of U4/U6.U5 tri-snRNP. We confirmed that silencing squamous cell carcinoma antigen recognized by T cells (SART1), a specific factor of tri-snRNP, abrogates IFN-α's suppressive effects against HCV in both replicon cells and JFH1 infectious cells. We further found that SART1 was not IFN-α inducible, and its anti-HCV effector in the JFH1 infectious model was through regulation of interferon stimulated genes (ISGs) with or without IFN-α. CONCLUSIONS: We identified 93 genes that mediate the anti-HCV effect of IFN-α through genome-wide siRNA screening; 23 and nine genes were involved in mRNA processing and translation initiation, respectively. These findings reveal an unexpected role for mRNA processing in generation of the antiviral state, and suggest a new avenue for therapeutic development in HCV.


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus/drug effects , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/genetics , Host-Pathogen Interactions/genetics , Interferon-alpha/pharmacology , Antigens, Neoplasm/genetics , Cell Line , Computational Biology , Genome, Human , Genomics , Hepacivirus/genetics , Hepacivirus/physiology , Hepatitis C, Chronic/virology , High-Throughput Screening Assays , Humans , Pharmacogenetics , RNA, Small Interfering/genetics , Receptor, Interferon alpha-beta/genetics , Replicon , Ribonucleoproteins, Small Nuclear/genetics
19.
Nihon Rinsho ; 70(4): 664-70, 2012 Apr.
Article in Japanese | MEDLINE | ID: mdl-22568151

ABSTRACT

Recently, first direct acting antiviral (DAA) against hepatitis C virus (HCV) has just approved in Japan. It is a first generation protease inhibitor, telaprevir. Telaprevir inhibits HCV NS3 & 4A serine protease, and combination with pegylated-interferon and ribavirin has now become a standard of care (SOC) for patients with genotype 1 high viral load hepatitis C. Fortunately, more than 50 new antivirals against HCV are under development including antivirals in preclinical trials. New antivirals are classified into several categories; (1) NS3 & 4A protease inhibitor, (2) NS5B polymerase inhibitor, (3) NS5A inhibitor, (4) host factor targeting antivirals, (5) interferon-related antivirals, and others. Combination of different classes of antivirals without interferon is expected to become a future SOC for hepatitis C.


Subject(s)
Hepacivirus/drug effects , DNA-Directed RNA Polymerases/antagonists & inhibitors , Enzyme Precursors/antagonists & inhibitors , Hepatitis C/drug therapy , Humans , Pancreatic Elastase/antagonists & inhibitors
20.
J Hepatol ; 55(2): 289-98, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21147189

ABSTRACT

BACKGROUND & AIMS: The combination of pegylated interferon (IFN) α and ribavirin (RBV) is the standard therapy for patients with chronic HCV infection. However, it produces a sustained virologic response (SVR) in only half of the treated individuals and is associated with significant side effects. Recently, several single-nucleotide polymorphisms (SNPs) near the IL28B locus, also known as IFNλ3, were identified to be strong predictors of SVR in patients receiving PEG-IFN and RBV. We sought to determine whether IL28B was capable of inhibiting HCV replication and to determine the pathway by which IL28B exhibits anti-HCV activity. METHODS: Using the full-length HCV replicon OR6 and the infectious HCV clones JFH1 and Jc1, we assessed the anti-HCV effect of IL28B on HCV and characterized the key steps of the JAK-STAT pathway by real time PCR, luciferase assay, and Western blot. Finally, we evaluated the anti-HCV effect of IL28B in the presence of JAK-STAT pathway inhibitors such as blocking antibodies, a pharmacological inhibitor, and siRNAs. RESULTS: We found that IL28B inhibits HCV replication in a dose- and time-dependent manner. Like IFNα, IL28B induces the phosphorylation of STAT1 and STAT2, ISRE-driven transcription, and expression of known ISGs. The anti-HCV effects of IL28A, IL28B, and IL29 were abrogated by an IL10R2 blocking antibody, a pharmacological inhibitor of JAK1/TYK2, and by siRNA against IL28R1, STAT1, STAT2, and IRF9. CONCLUSIONS: Our data demonstrate that IL28A, IL28B, and IL29 signal through the JAK-STAT pathway to inhibit HCV. These data suggest possible applications of new approaches in HCV treatment.


Subject(s)
Hepacivirus/drug effects , Hepacivirus/physiology , Interleukins/pharmacology , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Antiviral Agents/pharmacology , Base Sequence , Cell Line , DNA Primers/genetics , Humans , Interferon-Stimulated Gene Factor 3, gamma Subunit/antagonists & inhibitors , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Interferons , Interleukins/genetics , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Janus Kinases/antagonists & inhibitors , Phosphorylation/drug effects , Polymorphism, Single Nucleotide , RNA, Small Interfering/genetics , Receptors, Interferon/metabolism , Recombinant Proteins/pharmacology , STAT Transcription Factors/antagonists & inhibitors , STAT1 Transcription Factor/antagonists & inhibitors , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/antagonists & inhibitors , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism , Signal Transduction/drug effects , TYK2 Kinase/antagonists & inhibitors , TYK2 Kinase/metabolism , Virus Replication/drug effects , Virus Replication/physiology
SELECTION OF CITATIONS
SEARCH DETAIL