Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cells ; 13(12)2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38920664

ABSTRACT

Hepatitis C virus (HCV) is an oncogenic virus that causes chronic liver disease in more than 80% of patients. During the last decade, efficient direct-acting antivirals were introduced into clinical practice. However, clearance of the virus does not reduce the risk of end-stage liver diseases to the level observed in patients who have never been infected. So, investigation of HCV pathogenesis is still warranted. Virus-induced changes in cell metabolism contribute to the development of HCV-associated liver pathologies. Here, we studied the impact of the virus on the metabolism of polyamines and proline as well as on the urea cycle, which plays a crucial role in liver function. It was found that HCV strongly suppresses the expression of arginase, a key enzyme of the urea cycle, leading to the accumulation of arginine, and up-regulates proline oxidase with a concomitant decrease in proline concentrations. The addition of exogenous proline moderately suppressed viral replication. HCV up-regulated transcription but suppressed protein levels of polyamine-metabolizing enzymes. This resulted in a decrease in polyamine content in infected cells. Finally, compounds targeting polyamine metabolism demonstrated pronounced antiviral activity, pointing to spermine and spermidine as compounds affecting HCV replication. These data expand our understanding of HCV's imprint on cell metabolism.


Subject(s)
Hepacivirus , Polyamines , Proline , Urea , Virus Replication , Proline/metabolism , Humans , Hepacivirus/physiology , Hepacivirus/drug effects , Polyamines/metabolism , Urea/metabolism , Urea/pharmacology , Virus Replication/drug effects , Arginase/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/metabolism , Hepatitis C/metabolism , Hepatitis C/virology , Cell Line, Tumor , Proline Oxidase/metabolism
2.
JHEP Rep ; 5(3): 100647, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36718430

ABSTRACT

Background & Aims: Chronic HCV infection causes cellular stress, fibrosis and predisposes to hepatocarcinogenesis. Mitochondria play key roles in orchestrating stress responses by regulating bioenergetics, inflammation and apoptosis. To better understand the role of mitochondria in the viral life cycle and disease progression of chronic hepatitis C, we studied morphological and functional mitochondrial alterations induced by HCV using productively infected hepatoma cells and patient livers. Methods: Biochemical and imaging assays were used to assess localization of cellular and viral proteins and mitochondrial functions in cell cultures and liver biopsies. Cyclophilin D (CypD) knockout was performed using CRISPR/Cas9 technology. Viral replication was quantified by quantitative reverse-transcription PCR and western blotting. Results: Several HCV proteins were found to associate with mitochondria-associated endoplasmic reticulum (ER) membranes (MAMs), the points of contact between the ER and mitochondria. Downregulation of CypD, which is known to disrupt MAM integrity, reduced viral replication, suggesting that MAMs play an important role in the viral life cycle. This process was rescued by ectopic CypD expression. Furthermore, HCV proteins were found to associate with voltage dependent anion channel 1 (VDAC1) at MAMs and to reduce VDAC1 protein levels at MAMs in vitro and in patient biopsies. This association did not affect MAM-associated functions in glucose homeostasis and Ca2+ signaling. Conclusions: HCV proteins associate specifically with MAMs and MAMs play an important role in viral replication. The association between viral proteins and MAMs did not impact Ca2+ signaling between the ER and mitochondria or glucose homeostasis. Whether additional functions of MAMs and/or VDAC are impacted by HCV and contribute to the associated pathology remains to be assessed. Impact and implications: Hepatitis C virus infects the liver, where it causes inflammation, cell damage and increases the long-term risk of liver cancer. We show that several HCV proteins interact with mitochondria in liver cells and alter the composition of mitochondrial subdomains. Importantly, HCV requires the architecture of these mitochondrial subdomains to remain intact for efficient viral replication.

3.
Sci Rep ; 9(1): 8378, 2019 06 10.
Article in English | MEDLINE | ID: mdl-31182764

ABSTRACT

Active liver diseases are characterized by an infiltration of inflammatory immune cells, which interact locally with hepatocytes. Co-cultures between non- and -activated human peripheral blood mononuclear cells (PBMCs) and human hepatoma HepaRG cells were used to determine the role of these cell interactions in the inflammatory response. At the early stage, PBMC-HepaRG cell interactions increased mRNA expression and/or secretion of IL-6, IL-8, CCL-20 and MCP-1, in part through direct cell contact and the induction was higher in PHA-activated conditions. The pro-inflammatory cytokines IL-17 and/or TNFα contributed to the increase of IL-6 and IL-8 secretion. HepaRG cells modulated T cell polarization by increasing Th1 cell transcription factor expression and by reducing CD3+ CD4+ IL-17+ cell frequency when PBMCs were activated with PHA. At a later stage, the presence of HepaRG cells inhibited PHA-induced HLA-DR expression on PBMCs, and PBMC proliferation. In contrast, the presence of skin fibroblasts had no effect of PBMC proliferation induced by PHA. After a first pro-inflammatory phase, PBMC-HepaRG cell interactions may down-regulate the immune response. The PBMC-hepatocyte interactions can thus participate first to the initiation of hepatitis and later to the maintenance of immune tolerance in liver, possibly contributing to chronicity.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cell Communication/genetics , Inflammation/genetics , Liver Neoplasms/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Chemokine CCL2/genetics , Coculture Techniques , Gene Expression Regulation, Neoplastic , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Inflammation/pathology , Interleukin-17/genetics , Interleukin-6/genetics , Interleukin-8/genetics , Kinetics , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Liver/metabolism , Liver/pathology , Liver Neoplasms/pathology , RNA, Messenger/genetics , Tumor Necrosis Factor-alpha/genetics
4.
J Gen Virol ; 98(7): 1646-1657, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28721844

ABSTRACT

The roles of CD81 in the hepatitis C virus (HCV) life cycle are multiple but remain ill characterized. CD81 is known to interact with the HCV glycoproteins as an attachment factor. It also has an important role in the post-attachment entry process. Its interaction with claudin-1, for example, is vital for viral uptake and trafficking. Furthermore, CD81 and its role in membrane organization and trafficking are thought to play a pivotal role in HCV replication. Some of these functions are particularly limited to human CD81; others can be substituted with CD81 molecules from other species. However, with the exception of the large extracellular loop sequence, the structure-function analysis of CD81 in the HCV infectious cycle remains ill characterized. We describe here the fusion molecules between the large extracellular loops of human or mouse CD81 and lipid-raft-associated or unassociated GPI anchors. These fusion molecules have strong antiviral activity in a dominant negative fashion, independent of membrane raft association. Their expression in the hepatoma cell line Huh7.5 blocks HCV uptake, transmission and replication. These molecules will be useful to decipher the various roles of CD81 in the HCV life cycle and transmission in more detail.


Subject(s)
Hepacivirus/physiology , Hepatitis C/transmission , Membrane Microdomains/metabolism , Tetraspanin 28/metabolism , Viral Envelope Proteins/metabolism , Virus Attachment , Virus Replication/physiology , Animals , Cell Line, Tumor , HEK293 Cells , HIV-1/physiology , HeLa Cells , Humans , Mice , Protein Binding/physiology , Tetraspanin 28/genetics , Virus Internalization
5.
Biochemistry ; 56(24): 3029-3048, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28535337

ABSTRACT

Hepatitis C virus (HCV) nonstructural protein 5A (NS5A) is a RNA-binding phosphoprotein composed of a N-terminal membrane anchor (AH), a structured domain 1 (D1), and two intrinsically disordered domains (D2 and D3). The knowledge of the functional architecture of this multifunctional protein remains limited. We report here that NS5A-D1D2D3 produced in a wheat germ cell-free system is obtained under a highly phosphorylated state. Its NMR analysis revealed that these phosphorylations do not change the disordered nature of D2 and D3 domains but increase the number of conformers due to partial phosphorylations. By combining NMR and small angle X-ray scattering, we performed a comparative structural characterization of unphosphorylated recombinant D2 domains of JFH1 (genotype 2a) and the Con1 (genotype 1b) strains produced in Escherichia coli. These analyses highlighted a higher intrinsic folding of the latter, revealing the variability of intrinsic conformations in HCV genotypes. We also investigated the effect of D2 mutations conferring resistance of HCV replication to cyclophilin A (CypA) inhibitors on the structure of the recombinant D2 Con1 mutants and their binding to CypA. Although resistance mutations D320E and R318W could induce some local and/or global folding perturbation, which could thus affect the kinetics of conformer interconversions, they do not significantly affect the kinetics of CypA/D2 interaction measured by surface plasmon resonance (SPR). The combination of all our data led us to build a model of the overall structure of NS5A, which provides a useful template for further investigations of the structural and functional features of this enigmatic protein.


Subject(s)
Antiviral Agents/pharmacology , Cyclosporine/pharmacology , Drug Resistance, Viral/drug effects , Hepacivirus/drug effects , Mutation , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Virus Replication/drug effects , Hepacivirus/genetics , Hepacivirus/growth & development , Mass Spectrometry , Microbial Sensitivity Tests , Models, Molecular , Protein Conformation
6.
Hepatology ; 65(3): 789-803, 2017 03.
Article in English | MEDLINE | ID: mdl-27863447

ABSTRACT

Chronic infection with hepatitis C virus (HCV) is one of the main causes of hepatocellular carcinoma. However, the molecular mechanisms linking the infection to cancer development remain poorly understood. Here we used HCV-infected cells and liver biopsies to study how HCV modulates the glutaminolysis pathway, which is known to play an important role in cellular energetics, stress defense, and neoplastic transformation. Transcript levels of glutaminolytic factors were quantified in Huh7.5 cells or primary human hepatocytes infected with the Japanese fulminant hepatitis 1 HCV strain as well as in biopsies of chronic HCV patients. Nutrient deprivation, biochemical analysis, and metabolite quantification were performed with HCV-infected Huh7.5 cells. Furthermore, short hairpin RNA vectors and small molecule inhibitors were used to investigate the dependence of HCV replication on metabolic changes. We show that HCV modulates the transcript levels of key enzymes of glutamine metabolism in vitro and in liver biopsies of chronic HCV patients. Consistently, HCV infection increases glutamine use and dependence. We finally show that inhibiting glutamine metabolism attenuates HCV infection and the oxidative stress associated with HCV infection. CONCLUSION: Our data suggest that HCV establishes glutamine dependence, which is required for viral replication, and, importantly, that glutamine addiction is a hallmark of tumor cells. While HCV induces glutaminolysis to create an environment favorable for viral replication, it predisposes the cell to transformation. Glutaminolytic enzymes may be interesting therapeutic targets for prevention of hepatocarcinogenesis in chronic hepatitis C. (Hepatology 2017;65:789-803).


Subject(s)
Glutamine/metabolism , Hepacivirus/pathogenicity , Hepatocytes/metabolism , Hepatocytes/virology , Virus Replication/genetics , Biopsy, Needle , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cells, Cultured , Hepacivirus/genetics , Hepatitis C, Chronic/pathology , Hepatitis C, Chronic/physiopathology , Humans , Immunohistochemistry , Liver Neoplasms/pathology , Liver Neoplasms/virology , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction/methods , Statistics, Nonparametric , Transfection/methods
7.
Methods Mol Biol ; 1432: 119-31, 2016.
Article in English | MEDLINE | ID: mdl-27485333

ABSTRACT

This chapter includes a practical method of membrane protein production in Leishmania tarentolae cells. We routinely use it to express membrane proteins of the ABC (adenosine triphosphate-binding cassette) family, here exemplified with ABCG6 from L. braziliensis, implicated in phospholipid trafficking and drug efflux. The pLEXSY system used here allows membrane protein production with a mammalian-like N-glycosylation pattern, at high levels and at low costs. Also the effects of an N-terminal truncation of the protein are described. The method is described to allow any kind of membrane protein production.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Leishmania/growth & development , Protein Engineering/methods , ATP-Binding Cassette Transporters/genetics , Animals , Gene Expression , Glycosylation , Humans , Leishmania/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protozoan Proteins/genetics
8.
Protein Expr Purif ; 116: 1-6, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26325423

ABSTRACT

Non-structural protein 2 (NS2) of the hepatitis C virus (HCV) is an integral membrane protein that contains a cysteine protease and that plays a central organizing role in assembly of infectious progeny virions. While the crystal structure of the protease domain has been solved, the NS2 full-length form remains biochemically and structurally uncharacterized because recombinant NS2 could not be prepared in sufficient quantities from cell-based systems. We show here that functional NS2 in the context of the NS2-NS3pro precursor protein, ensuring NS2-NS3 cleavage, can be efficiently expressed by using a wheat germ cell-free expression system. In this same system, we subsequently successfully produce and purify milligram amounts of a detergent-solubilized form of full-length NS2 exhibiting the expected secondary structure content. Furthermore, immuno-electron microscopy analyses of reconstituted proteoliposomes demonstrate NS2 association with model membranes.


Subject(s)
Hepacivirus/chemistry , Hepacivirus/genetics , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Amino Acid Sequence , Cell-Free System/metabolism , Chromatography, Gel , Cloning, Molecular , Detergents/chemistry , Gene Expression , Hepatitis C/virology , Liposomes/chemistry , Membrane Lipids/chemistry , Molecular Sequence Data , Plasmids/genetics , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Solubility , Triticum/genetics , Viral Nonstructural Proteins/isolation & purification
9.
J Biol Chem ; 285(33): 25802-11, 2010 Aug 13.
Article in English | MEDLINE | ID: mdl-20551330

ABSTRACT

The density of hepatitis C virus (HCV) particles circulating in the blood of chronically infected patients and of cell-culture produced HCV is heterogeneous. Specific infectivity and fusion of low density particles are higher than those of high density particles. We recently characterized hybrid particles produced by Caco-2 colon or Huh-7.5 liver cells transduced with HCV E1 and E2 envelope glycoproteins. Caco-2-derived particles, called empty lipo-viral particles (eLVP), are composed of triglyceride-rich lipoproteins positive for apolipoproteins B (i.e. apoB100 and apoB48) and contain HCV E1 and E2. Here we aimed at characterizing the morphology and in vitro fusion properties of eLVP using electron microscopy and fluorescence spectroscopy. They displayed the aspect of beta-lipoproteins, and immunogold labeling confirmed the presence of apoB and HCV E1 and E2 at their surface. These particles are able to fuse with lipid bilayers (liposomes) in a fusion process leading to the coalescence of internal contents of triglyceride-rich lipoproteins particles and liposomes. Fusion was pH-dependent and could be inhibited by either Z-fFG, a peptide known to inhibit viral fusion, or by monoclonal antibodies directed against HCV E2 or the apolipoprotein moiety of the hybrid particle. Interestingly, particles derived from Huh-7.5 cells failed to display equivalent efficient fusion. Optimal fusion activity is, thus, observed when HCV envelope proteins are associated to apoB-positive hybrid particles. Our results, therefore, point to a crucial role of the E1 and E2 proteins in HCV fusion with a subtle interplay with the apolipoprotein part of eLVP.


Subject(s)
Hepacivirus/genetics , Lipoproteins/metabolism , Triglycerides/metabolism , Viral Fusion Proteins/metabolism , Caco-2 Cells , Cell Line, Tumor , Cryoelectron Microscopy , Humans , Lipoproteins/ultrastructure , Microscopy, Electron , Transduction, Genetic , Viral Fusion Proteins/genetics , Virion/metabolism , Virion/ultrastructure
10.
J Biol Chem ; 282(44): 32357-69, 2007 Nov 02.
Article in English | MEDLINE | ID: mdl-17761674

ABSTRACT

Cell entry of hepatitis C virus (HCV) is strikingly linked to lipoproteins and their receptors. Particularly, high density lipoprotein (HDL) enhances infectivity of HCV by involving the lipid-transfer function of the scavenger receptor BI, a receptor for both HDL and HCV. Here, we demonstrate that apoC-I, an exchangeable apolipoprotein that predominantly resides in HDL, specifically enhances HCVcc and HCVpp infectivity and increases the fusion rates between viral and target membranes via a direct interaction with the HCV surface. We identify the hypervariable region 1, located at the N terminus of the HCV E2 glycoprotein, as an essential viral component that modulates apoC-I-mediated enhancement of HCV fusion properties. The affinity of apoC-I for the HCV membrane may predispose it for fusion with a target membrane via alterations of its outer phospholipid layer. Conversely, we found that excess apoC-I provided as lipoprotein-free protein induces the disruption of the HCV membrane and subsequent loss of infectivity. Furthermore, our data indicate that HDL neither interacts nor spontaneously exchanges apoC-I with HCV virions. Because apoC-I is not present in serum as a lipoprotein-free form, our results suggest that HDL-embedded apoC-I could be released from the lipoprotein particle through a fine-tuned mechanism regulated via a triple interplay between hypervariable region 1, HDL, and scavenger receptor BI, resulting in optimal apoC-I recruitment on the viral membrane. These results provide the first description of a host serum factor helping the fusion process of an enveloped virus.


Subject(s)
Apolipoprotein C-I/metabolism , Hepacivirus/physiology , Virus Internalization , Cell Line, Tumor , Humans , Lipoproteins, HDL/metabolism , Scavenger Receptors, Class B/metabolism , Viral Proteins/metabolism
11.
J Virol ; 81(16): 8752-65, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17537855

ABSTRACT

Infection of eukaryotic cells by enveloped viruses requires the merging of viral and cellular membranes. Highly specific viral surface glycoproteins, named fusion proteins, catalyze this reaction by overcoming inherent energy barriers. Hepatitis C virus (HCV) is an enveloped virus that belongs to the genus Hepacivirus of the family Flaviviridae. Little is known about the molecular events that mediate cell entry and membrane fusion for HCV, although significant progress has been made due to recent developments in infection assays. Here, using infectious HCV pseudoparticles (HCVpp), we investigated the molecular basis of HCV membrane fusion. By searching for classical features of fusion peptides through the alignment of sequences from various HCV genotypes, we identified six regions of HCV E1 and E2 glycoproteins that present such characteristics. We introduced conserved and nonconserved amino acid substitutions in these regions and analyzed the phenotype of HCVpp generated with mutant E1E2 glycoproteins. This was achieved by (i) quantifying the infectivity of the pseudoparticles, (ii) studying the incorporation of E1E2 and their capacity to mediate receptor binding, and (iii) determining their fusion capacity in cell-cell and liposome/HCVpp fusion assays. We propose that at least three of these regions (i.e., at positions 270 to 284, 416 to 430, and 600 to 620) play a role in the membrane fusion process. These regions may contribute to the merging of viral and cellular membranes either by interacting directly with lipid membranes or by assisting the fusion process through their involvement in the conformational changes of the E1E2 complex at low pH.


Subject(s)
Hepacivirus/physiology , Membrane Fusion , Viral Envelope Proteins/metabolism , Virion/metabolism , Amino Acid Sequence , Hepacivirus/genetics , Humans , Molecular Sequence Data , Mutation , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Virion/chemistry
12.
Biochemistry ; 46(20): 6050-9, 2007 May 22.
Article in English | MEDLINE | ID: mdl-17455911

ABSTRACT

Hepatitis C affects approximately 3% of the world population, yet its current treatment options are limited to interferon-ribavirin drug regimens which achieve a 50-70% cure rate depending on the hepatitis C virus (HCV) genotype. Besides extensive screening for HCV-specific compounds, some well-established medicinal drugs have recently demonstrated an anti-HCV effect in HCV replicon cells. One of these drugs is arbidol (ARB), a Russian-made broad-spectrum antiviral agent, which we have previously shown to inhibit acute and chronic HCV infection. Here we show that ARB inhibits the cell entry of HCV pseudoparticles of genotypes 1a, 1b, and 2a in a dose-dependent fashion. ARB also displayed a dose-dependent inhibition of HCV membrane fusion, as assayed by using HCV pseudoparticles (HCVpp) and fluorescent liposomes. ARB inhibition of HCVpp fusion was found to be more effective on genotype 1a than on genotypes 1b and 2a. In vitro biochemical studies revealed association of ARB with membranelike environments such as detergents and with lipid membranes. This association was particularly prominent at acidic pH which is optimal for HCV-mediated fusion. Our results suggest that the affinity of ARB for lipid membranes could account for its anti-HCV actions, together with a differential level of interaction with key motifs in HCV glycoproteins of different genotypes.


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus/drug effects , Indoles/pharmacology , Antiviral Agents/chemistry , Cell Line, Tumor , Detergents , Hepacivirus/chemistry , Hepacivirus/pathogenicity , Humans , Liposomes , Membrane Fusion/drug effects , Membrane Lipids/antagonists & inhibitors , Membrane Lipids/chemistry , Micelles , Virion/chemistry , Virion/drug effects , Virion/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL
...