Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 29
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Gut ; 71(12): 2526-2538, 2022 12.
Article de Anglais | MEDLINE | ID: mdl-35058274

RÉSUMÉ

OBJECTIVE: Mucosal-associated invariant T (MAIT) cells are the most abundant T cells in human liver. They respond to bacterial metabolites presented by major histocompatibility complex-like molecule MR1. MAIT cells exert regulatory and antimicrobial functions and are implicated in liver fibrogenesis. It is not well understood which liver cells function as antigen (Ag)-presenting cells for MAIT cells, and under which conditions stimulatory Ags reach the circulation. DESIGN: We used different types of primary human liver cells in Ag-presentation assays to blood-derived and liver-derived MAIT cells. We assessed MAIT cell stimulatory potential of serum from healthy subjects and patients with portal hypertension undergoing transjugular intrahepatic portosystemic shunt stent, and patients with inflammatory bowel disease (IBD). RESULTS: MAIT cells were dispersed throughout healthy human liver and all tested liver cell types stimulated MAIT cells, hepatocytes being most efficient. MAIT cell activation by liver cells occurred in response to bacterial lysate and pure Ag, and was prevented by non-activating MR1 ligands. Serum derived from peripheral and portal blood, and from patients with IBD stimulated MAIT cells in MR1-dependent manner. CONCLUSION: Our findings reveal previously unrecognised roles of liver cells in Ag metabolism and activation of MAIT cells, repression of which creates an opportunity to design antifibrotic therapies. The presence of MAIT cell stimulatory Ags in serum rationalises the observed activated MAIT cell phenotype in liver. Increased serum levels of gut-derived MAIT cell stimulatory ligands in patients with impaired intestinal barrier function indicate that intrahepatic Ag-presentation may represent an important step in the development of liver disease.


Sujet(s)
Maladies inflammatoires intestinales , Cellules T invariantes associées aux muqueuses , Humains , Antigènes mineurs d'histocompatibilité , Antigènes d'histocompatibilité de classe I/génétique , Antigènes d'histocompatibilité de classe I/métabolisme , Foie/métabolisme , Hépatocytes/métabolisme , Maladies inflammatoires intestinales/métabolisme , Activation des lymphocytes
2.
Nat Commun ; 12(1): 4882, 2021 08 12.
Article de Anglais | MEDLINE | ID: mdl-34385466

RÉSUMÉ

Genetic variants of the interferon lambda (IFNL) gene locus are strongly associated with spontaneous and IFN treatment-induced clearance of hepatitis C virus (HCV) infections. Individuals with the ancestral IFNL4-dG allele are not able to clear HCV in the acute phase and have more than a 90% probability to develop chronic hepatitis C (CHC). Paradoxically, the IFNL4-dG allele encodes a fully functional IFNλ4 protein with antiviral activity against HCV. Here we describe an effect of IFNλ4 on HCV antigen presentation. Only minor amounts of IFNλ4 are secreted, because the protein is largely retained in the endoplasmic reticulum (ER) where it induces ER stress. Stressed cells are significantly weaker activators of HCV specific CD8+ T cells than unstressed cells. This is not due to reduced MHC I surface presentation or extracellular IFNλ4 effects, since T cell responses are restored by exogenous loading of MHC with HCV antigens. Rather, IFNλ4 induced ER stress impairs HCV antigen processing and/or loading onto the MHC I complex. Our results provide a potential explanation for the IFNλ4-HCV paradox.


Sujet(s)
Présentation d'antigène/immunologie , Lymphocytes T CD8+/immunologie , Hepacivirus/immunologie , Interleukines/immunologie , Activation des lymphocytes/immunologie , Cellules A549 , Lymphocytes T CD8+/métabolisme , Lymphocytes T CD8+/virologie , Lignée cellulaire tumorale , Régulation de l'expression des gènes/immunologie , Génotype , Cellules HepG2 , Hepacivirus/génétique , Hepacivirus/physiologie , Interactions hôte-pathogène/immunologie , Humains , Interleukines/génétique , Interleukines/métabolisme
3.
PLoS One ; 14(8): e0221762, 2019.
Article de Anglais | MEDLINE | ID: mdl-31465481

RÉSUMÉ

BACKGROUND & AIMS: Hepatocyte-like cells (HLCs) differentiated from induced pluripotent stem cells (iPSCs) have emerged as a promising cell culture model to study metabolism, biotransformation, viral infections and inherited liver diseases. iPSCs provide an unlimited supply for the generation of HLCs, but incomplete HLC differentiation remains a major challenge. iPSC may carry-on a tissue of origin dependent expression memory influencing iPSC differentiation into different cell types. Whether liver derived iPSCs (Li-iPSCs) would allow the generation of more fully differentiated HLCs is not known. METHODS: In the current study, we used primary liver cells (PLCs) expanded from liver needle biopsies and reprogrammed them into Li-iPSCs using a non-integrative Sendai virus-based system. Li-iPSCs were differentiated into HLCs using established differentiation protocols. The HLC phenotype was characterized at the protein, functional and transcriptional level. RNA sequencing data were generated from the originating liver biopsies, the Li-iPSCs, fibroblast derived iPSCs, and differentiated HLCs, and used to characterize and compare their transcriptome profiles. RESULTS: Li-iPSCs indeed retain a liver specific transcriptional footprint. Li-iPSCs can be propagated to provide an unlimited supply of cells for differentiation into Li-HLCs. Similar to HLCs derived from fibroblasts, Li-HLCs could not be fully differentiated into hepatocytes. Relative to the originating liver, Li-HLCs showed lower expression of liver specific transcription factors and increased expression of genes involved in the differentiation of other tissues. CONCLUSIONS: PLCs and Li-iPSCs obtained from small pieces of human needle liver biopsies constitute a novel unlimited source for the production of HLCs. Despite the preservation of a liver specific gene expression footprint in Li-iPSCs, the generation of fully differentiated hepatocytes cannot be achieved with the current differentiation protocols.


Sujet(s)
Hépatocytes/cytologie , Cellules souches pluripotentes induites/cytologie , Foie/anatomopathologie , Animaux , Marqueurs biologiques/métabolisme , Biopsie , Différenciation cellulaire/génétique , Prolifération cellulaire , Cellules cultivées , Reprogrammation cellulaire , Analyse de regroupements , Fibroblastes/cytologie , Régulation de l'expression des gènes , Hépatocytes/métabolisme , Humains , Cellules souches pluripotentes induites/métabolisme , Souris SCID , Analyse en composantes principales , Facteurs de transcription/métabolisme , Transcription génétique
4.
Hepatol Commun ; 3(7): 971-986, 2019 Jul.
Article de Anglais | MEDLINE | ID: mdl-31334445

RÉSUMÉ

Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related deaths worldwide. Treatment options for patients with advanced-stage disease are limited. A major obstacle in drug development is the lack of an in vivo model that accurately reflects the broad spectrum of human HCC. Patient-derived xenograft (PDX) tumor mouse models could overcome the limitations of cancer cell lines. PDX tumors maintain the genetic and histologic heterogeneity of the originating tumors and are used for preclinical drug development in various cancers. Controversy exists about their genetic and molecular stability through serial passaging in mice. We aimed to establish PDX models from human HCC biopsies and to characterize their histologic and molecular stability during serial passaging. A total of 54 human HCC needle biopsies that were derived from patients with various underlying liver diseases and tumor stages were transplanted subcutaneously into immunodeficient, nonobese, diabetic/severe combined immunodeficiency gamma-c mice; 11 successfully engrafted. All successfully transplanted HCCs were Edmondson grade III or IV. HCC PDX tumors retained the histopathologic, transcriptomic, and genomic characteristics of the original HCC biopsies over 6 generations of retransplantation. These characteristics included Edmondson grade, expression of tumor markers, tumor gene signature, tumor-associated mutations, and copy number alterations. Conclusion: PDX mouse models can be established from undifferentiated HCCs, with an overall success rate of approximately 20%. The transplanted tumors represent the entire spectrum of the molecular landscape of HCCs and preserve the characteristics of the originating tumors through serial passaging. HCC PDX models are a promising tool for preclinical personalized drug development.

5.
Cell Rep ; 24(5): 1363-1376, 2018 07 31.
Article de Anglais | MEDLINE | ID: mdl-30067989

RÉSUMÉ

Hepatocellular carcinoma (HCC) is the most common primary liver cancer and the second most frequent cause of cancer-related mortality worldwide. The multikinase inhibitor sorafenib is the only treatment option for advanced HCC. Due to tumor heterogeneity, its efficacy greatly varies between patients and is limited due to adverse effects and drug resistance. Current in vitro models fail to recapitulate key features of HCCs. We report the generation of long-term organoid cultures from tumor needle biopsies of HCC patients with various etiologies and tumor stages. HCC organoids retain the morphology as well as the expression pattern of HCC tumor markers and preserve the genetic heterogeneity of the originating tumors. In a proof-of-principle study, we show that liver cancer organoids can be used to test sensitivity to sorafenib. In conclusion, organoid models can be derived from needle biopsies of liver cancers and provide a tool for developing tailored therapies.


Sujet(s)
Carcinome hépatocellulaire/anatomopathologie , Tumeurs du foie/anatomopathologie , Organoïdes/anatomopathologie , Sujet âgé , Sujet âgé de 80 ans ou plus , Animaux , Cellules cultivées , Femelle , Humains , Mâle , Souris , Adulte d'âge moyen , Techniques de culture de tissus/méthodes
6.
J Virol ; 90(14): 6387-6400, 2016 07 15.
Article de Anglais | MEDLINE | ID: mdl-27147737

RÉSUMÉ

UNLABELLED: Hepatitis C virus (HCV)-induced chronic liver disease is a leading cause of hepatocellular carcinoma (HCC). However, the molecular mechanisms underlying HCC development following chronic HCV infection remain poorly understood. MicroRNAs (miRNAs) play an important role in homeostasis within the liver, and deregulation of miRNAs has been associated with liver disease, including HCC. While host miRNAs are essential for HCV replication, viral infection in turn appears to induce alterations of intrahepatic miRNA networks. Although the cross talk between HCV and liver cell miRNAs most likely contributes to liver disease pathogenesis, the functional involvement of miRNAs in HCV-driven hepatocyte injury and HCC remains elusive. Here we combined a hepatocyte-like cell-based model system, high-throughput small RNA sequencing, computational analysis, and functional studies to investigate HCV-miRNA interactions that may contribute to liver disease and HCC. Profiling analyses indicated that HCV infection differentially regulated the expression of 72 miRNAs by at least 2-fold, including miRNAs that were previously described to target genes associated with inflammation, fibrosis, and cancer development. Further investigation demonstrated that the miR-146a-5p level was consistently increased in HCV-infected hepatocyte-like cells and primary human hepatocytes, as well as in liver tissue from HCV-infected patients. Genome-wide microarray and computational analyses indicated that miR-146a-5p overexpression modulates pathways that are related to liver disease and HCC development. Furthermore, we showed that miR-146a-5p has a positive impact on late steps of the viral replication cycle, thereby increasing HCV infection. Collectively, our data indicate that the HCV-induced increase in miR-146a-5p expression both promotes viral infection and is relevant for pathogenesis of liver disease. IMPORTANCE: HCV is a leading cause of chronic liver disease and cancer. However, how HCV induces liver cancer remains poorly understood. There is accumulating evidence that a viral cure does not eliminate the risk for HCC development. Thus, there is an unmet medical need to develop novel approaches to predict and prevent virus-induced HCC. miRNA expression is known to be deregulated in liver disease and cancer. Furthermore, miRNAs are essential for HCV replication, and HCV infection alters miRNA expression. However, how miRNAs contribute to HCV-driven pathogenesis remains elusive. Here we show that HCV induces miRNAs that may contribute to liver injury and carcinogenesis. The miR-146a-5p level was consistently increased in different cell-based models of HCV infection and in HCV patient-derived liver tissue. Furthermore, miR-146a-5p increased HCV infection. Collectively, our data are relevant to understanding viral pathogenesis and may open perspectives for novel biomarkers and prevention of virus-induced liver disease and HCC.


Sujet(s)
Carcinome hépatocellulaire/virologie , Hepacivirus/pathogénicité , Hépatite C/virologie , Hépatocytes/métabolisme , Tumeurs du foie/virologie , Voies et réseaux métaboliques/génétique , microARN/génétique , Adulte , Sujet âgé , Marqueurs biologiques/analyse , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Femelle , Analyse de profil d'expression de gènes , Hépatite C/génétique , Hépatite C/anatomopathologie , Hépatocytes/cytologie , Hépatocytes/virologie , Séquençage nucléotidique à haut débit , Interactions hôte-pathogène , Humains , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Mâle , Adulte d'âge moyen , Activation de la transcription , Régulation positive
7.
Gastroenterology ; 150(1): 206-217.e4, 2016 Jan.
Article de Anglais | MEDLINE | ID: mdl-26404951

RÉSUMÉ

BACKGROUND & AIMS: Efforts to develop an effective vaccine against hepatitis C virus (HCV) have been hindered by the propensity of the virus to evade host immune responses. HCV particles in serum and in cell culture associate with lipoproteins, which contribute to viral entry. Lipoprotein association has also been proposed to mediate viral evasion of the humoral immune response, though the mechanisms are poorly defined. METHODS: We used small interfering RNAs to reduce levels of apolipoprotein E (apoE) in cell culture-derived HCV-producing Huh7.5-derived hepatoma cells and confirmed its depletion by immunoblot analyses of purified viral particles. Before infection of naïve hepatoma cells, we exposed cell culture-derived HCV strains of different genotypes, subtypes, and variants to serum and polyclonal and monoclonal antibodies isolated from patients with chronic HCV infection. We analyzed the interaction of apoE with viral envelope glycoprotein E2 and HCV virions by immunoprecipitation. RESULTS: Through loss-of-function studies on patient-derived HCV variants of several genotypes and subtypes, we found that the HCV particle apoE allows the virus to avoid neutralization by patient-derived antibodies. Functional studies with human monoclonal antiviral antibodies showed that conformational epitopes of envelope glycoprotein E2 domains B and C were exposed after depletion of apoE. The level and conformation of virion-associated apoE affected the ability of the virus to escape neutralization by antibodies. CONCLUSIONS: In cell-infection studies, we found that HCV-associated apoE helps the virus avoid neutralization by antibodies against HCV isolated from chronically infected patients. This method of immune evasion poses a challenge for the development of HCV vaccines.


Sujet(s)
Anticorps neutralisants/immunologie , Apolipoprotéines E/métabolisme , Hepacivirus/immunologie , Anticorps de l'hépatite C/immunologie , Cellules cultivées , Hepacivirus/génétique , Hépatite C/sang , Hépatocytes/immunologie , Humains , Statistique non paramétrique , Protéines de l'enveloppe virale/immunologie , Protéines de l'enveloppe virale/métabolisme , Pénétration virale
8.
Sci Rep ; 5: 13344, 2015 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-26323588

RÉSUMÉ

Cellular translation is down-regulated by host antiviral responses. Picornaviridae and Flaviviridae including hepatitis C virus (HCV) evade this process using internal ribosomal entry sequences (IRESs). Although HCV IRES translation is a prerequisite for HCV replication, only few host factors critical for IRES activity are known and the global regulator network remains largely unknown. Since signal transduction is an import regulator of viral infections and the host antiviral response we combined a functional RNAi screen targeting the human signaling network with a HCV IRES-specific reporter mRNA assay. We demonstrate that the HCV host cell cofactors PI4K and MKNK1 are positive regulators of HCV IRES translation representing a novel pathway with a functional relevance for the HCV life cycle and IRES-mediated translation of viral RNA.


Sujet(s)
Hepacivirus/génétique , Sites internes d'entrée des ribosomes/génétique , Protéines et peptides de signalisation intracellulaire/métabolisme , Phosphotransferases (Alcohol Group Acceptor)/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Lignée cellulaire , Survie cellulaire , Gènes rapporteurs , Humains , Protéines et peptides de signalisation intracellulaire/antagonistes et inhibiteurs , Protéines et peptides de signalisation intracellulaire/génétique , Antigènes mineurs d'histocompatibilité , Phosphotransferases (Alcohol Group Acceptor)/antagonistes et inhibiteurs , Phosphotransferases (Alcohol Group Acceptor)/génétique , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Protein-Serine-Threonine Kinases/génétique , Interférence par ARN , ARN messager/métabolisme , Petit ARN interférent/métabolisme , ARN viral/génétique
9.
Nat Biotechnol ; 33(5): 549-554, 2015 May.
Article de Anglais | MEDLINE | ID: mdl-25798937

RÉSUMÉ

Hepatitis C virus (HCV) infection is a leading cause of liver cirrhosis and cancer. Cell entry of HCV and other pathogens is mediated by tight junction (TJ) proteins, but successful therapeutic targeting of TJ proteins has not been reported yet. Using a human liver-chimeric mouse model, we show that a monoclonal antibody specific for the TJ protein claudin-1 (ref. 7) eliminates chronic HCV infection without detectable toxicity. This antibody inhibits HCV entry, cell-cell transmission and virus-induced signaling events. Antibody treatment reduces the number of HCV-infected hepatocytes in vivo, highlighting the need for de novo infection by means of host entry factors to maintain chronic infection. In summary, we demonstrate that an antibody targeting a virus receptor can cure chronic viral infection and uncover TJ proteins as targets for antiviral therapy.


Sujet(s)
Anticorps monoclonaux/administration et posologie , Claudine-1/immunologie , Hépatite C/thérapie , Cirrhose du foie/immunologie , Animaux , Anticorps monoclonaux/immunologie , Anticorps monoclonaux humanisés/administration et posologie , Anticorps monoclonaux humanisés/immunologie , Claudine-1/usage thérapeutique , Hepacivirus/immunologie , Hepacivirus/pathogénicité , Hépatite C/immunologie , Hépatite C/virologie , Hépatocytes/immunologie , Humains , Cirrhose du foie/thérapie , Cirrhose du foie/virologie , Souris
10.
Gut ; 64(3): 483-94, 2015 Mar.
Article de Anglais | MEDLINE | ID: mdl-24848265

RÉSUMÉ

OBJECTIVE: Although direct-acting antiviral agents (DAAs) have markedly improved the outcome of treatment in chronic HCV infection, there continues to be an unmet medical need for improved therapies in difficult-to-treat patients as well as liver graft infection. Viral entry is a promising target for antiviral therapy. DESIGN: Aiming to explore the role of entry inhibitors for future clinical development, we investigated the antiviral efficacy and toxicity of entry inhibitors in combination with DAAs or other host-targeting agents (HTAs). Screening a large series of combinations of entry inhibitors with DAAs or other HTAs, we uncovered novel combinations of antivirals for prevention and treatment of HCV infection. RESULTS: Combinations of DAAs or HTAs and entry inhibitors including CD81-, scavenger receptor class B type I (SR-BI)- or claudin-1 (CLDN1)-specific antibodies or small-molecule inhibitors erlotinib and dasatinib were characterised by a marked and synergistic inhibition of HCV infection over a broad range of concentrations with undetectable toxicity in experimental designs for prevention and treatment both in cell culture models and in human liver-chimeric uPA/SCID mice. CONCLUSIONS: Our results provide a rationale for the development of antiviral strategies combining entry inhibitors with DAAs or HTAs by taking advantage of synergy. The uncovered combinations provide perspectives for efficient strategies to prevent liver graft infection and novel interferon-free regimens.


Sujet(s)
Antiviraux/usage thérapeutique , Hepacivirus/effets des médicaments et des substances chimiques , Hépatite C/traitement médicamenteux , Pénétration virale/effets des médicaments et des substances chimiques , Animaux , Antiviraux/administration et posologie , Lignée cellulaire , Chimère , Synergie des médicaments , Association de médicaments , Hépatite C/prévention et contrôle , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/virologie , Humains , Souris , Souris SCID
11.
J Virol ; 88(16): 9418-28, 2014 Aug.
Article de Anglais | MEDLINE | ID: mdl-24920811

RÉSUMÉ

UNLABELLED: At least five New World (NW) arenaviruses cause hemorrhagic fevers in South America. These pathogenic clade B viruses, as well as nonpathogenic arenaviruses of the same clade, use transferrin receptor 1 (TfR1) of their host species to enter cells. Pathogenic viruses are distinguished from closely related nonpathogenic ones by their additional ability to utilize human TfR1 (hTfR1). Here, we investigate the receptor usage of North American arenaviruses, whose entry proteins share greatest similarity with those of the clade B viruses. We show that all six North American arenaviruses investigated utilize host species TfR1 orthologs and present evidence consistent with arenavirus-mediated selection pressure on the TfR1 of the North American arenavirus host species. Notably, one of these viruses, AV96010151, closely related to the prototype Whitewater Arroyo virus (WWAV), entered cells using hTfR1, consistent with a role for a WWAV-like virus in three fatal human infections whose causative agent has not been identified. In addition, modest changes were sufficient to convert hTfR1 into a functional receptor for most of these viruses, suggesting that a minor alteration in virus entry protein may allow these viruses to use hTfR1. Our data establish TfR1 as a cellular receptor for North American arenaviruses, highlight an "arms race" between these viruses and their host species, support the association of North American arenavirus with fatal human infections, and suggest that these viruses have a higher potential to emerge and cause human diseases than has previously been appreciated. IMPORTANCE: hTfR1 use is a key determinant for a NW arenavirus to cause hemorrhagic fevers in humans. All known pathogenic NW arenaviruses are transmitted in South America by their host rodents. North American arenaviruses are generally considered nonpathogenic, but some of these viruses have been tentatively implicated in human fatalities. We show that these North American arenaviruses use the TfR1 orthologs of their rodent host species and identify TfR1 polymorphisms suggesting an ongoing "arms race" between these viruses and their hosts. We also show that a close relative of a North American arenavirus suggested to have caused human fatalities, the Whitewater Arroyo species complex virus AV96010151, uses human TfR1. Moreover, we present data that imply that modest changes in other North American arenaviruses might allow these viruses to infect humans. Collectively, our data suggest that North American arenaviruses have a higher potential to cause human disease than previously assumed.


Sujet(s)
Antigènes CD/métabolisme , Arénavirus du Nouveau Monde/métabolisme , Récepteurs à la transferrine/métabolisme , Lignée cellulaire , Cellules HEK293 , Fièvres hémorragiques virales/métabolisme , Fièvres hémorragiques virales/virologie , Humains , Récepteurs viraux/métabolisme , Protéines virales/métabolisme , Pénétration virale
12.
PLoS Pathog ; 10(5): e1004128, 2014 May.
Article de Anglais | MEDLINE | ID: mdl-24830295

RÉSUMÉ

Hepatitis C virus (HCV) is transmitted between hepatocytes via classical cell entry but also uses direct cell-cell transfer to infect neighboring hepatocytes. Viral cell-cell transmission has been shown to play an important role in viral persistence allowing evasion from neutralizing antibodies. In contrast, the role of HCV cell-cell transmission for antiviral resistance is unknown. Aiming to address this question we investigated the phenotype of HCV strains exhibiting resistance to direct-acting antivirals (DAAs) in state-of-the-art model systems for cell-cell transmission and spread. Using HCV genotype 2 as a model virus, we show that cell-cell transmission is the main route of viral spread of DAA-resistant HCV. Cell-cell transmission of DAA-resistant viruses results in viral persistence and thus hampers viral eradication. We also show that blocking cell-cell transmission using host-targeting entry inhibitors (HTEIs) was highly effective in inhibiting viral dissemination of resistant genotype 2 viruses. Combining HTEIs with DAAs prevented antiviral resistance and led to rapid elimination of the virus in cell culture model. In conclusion, our work provides evidence that cell-cell transmission plays an important role in dissemination and maintenance of resistant variants in cell culture models. Blocking virus cell-cell transmission prevents emergence of drug resistance in persistent viral infection including resistance to HCV DAAs.


Sujet(s)
Antiviraux/pharmacologie , Communication cellulaire , Résistance virale aux médicaments , Hepacivirus/effets des médicaments et des substances chimiques , Hepacivirus/physiologie , Hépatite C/immunologie , Hépatite C/virologie , Pénétration virale , Anticorps neutralisants/métabolisme , Carbamates , Communication cellulaire/immunologie , Cellules cultivées , Résistance virale aux médicaments/immunologie , Hepacivirus/croissance et développement , Hépatite C/anatomopathologie , Humains , Imidazoles/pharmacologie , Oligopeptides/pharmacologie , Proline/analogues et dérivés , Proline/pharmacologie , Pyrrolidines , Valine/analogues et dérivés , Charge virale/immunologie , Pénétration virale/effets des médicaments et des substances chimiques
13.
Antiviral Res ; 104: 136-42, 2014 Apr.
Article de Anglais | MEDLINE | ID: mdl-24525381

RÉSUMÉ

Hepatitis C virus (HCV) is a major cause of liver cirrhosis and hepatocellular carcinoma. Furthermore, HCV-induced liver disease is the leading indication for liver transplantation. The recent introduction of direct-acting antivirals (DAAs) has revolutionized HCV treatment by making possible the cure of the majority of patients. However, their efficacy and safety in difficult-to-treat patients such as patients receiving immunosuppression, those with advanced liver disease, co-morbidity and HIV/HCV-co-infection remain to be determined. Furthermore, prevention of liver graft infection remains a pressing issue. HCV entry inhibitors target the very first step of the HCV life cycle and efficiently inhibit cell-cell transmission - a key prerequisite for viral spread. Because of their unique mechanism of action on cell-cell transmission they may provide a promising and simple perspective for prevention of liver graft infection. A high genetic barrier to resistance and complementary mechanism of action compared to DAAs makes entry inhibitors attractive as a new strategy for treatment of multi-resistant or difficult-to-treat patients. Clinical studies are needed to determine the future role of entry inhibitors in the arsenal of antivirals to combat HCV infection. This article forms part of a symposium in Antiviral Research on "Hepatitis C: next steps toward global eradication."


Sujet(s)
Antiviraux/pharmacologie , Antiviraux/usage thérapeutique , Hepacivirus/effets des médicaments et des substances chimiques , Hepacivirus/physiologie , Hépatite C/traitement médicamenteux , Pénétration virale/effets des médicaments et des substances chimiques , Animaux , Évaluation préclinique de médicament , Humains , Interférons/usage thérapeutique , Transplantation hépatique/effets indésirables
14.
J Virol ; 87(18): 10405-10, 2013 Sep.
Article de Anglais | MEDLINE | ID: mdl-23864633

RÉSUMÉ

The relevance of claudin-6 and claudin-9 in hepatitis C virus (HCV) entry remains elusive. We produced claudin-6- or claudin-9-specific monoclonal antibodies that inhibit HCV entry into nonhepatic cells expressing exogenous claudin-6 or claudin-9. These antibodies had no effect on HCV infection of hepatoma cells or primary hepatocytes. Thus, although claudin-6 and claudin-9 can serve as entry factors in cell lines, HCV infection into human hepatocytes is not dependent on claudin-6 and claudin-9.


Sujet(s)
Claudines/métabolisme , Hepacivirus/physiologie , Hépatocytes/virologie , Pénétration virale , Anticorps monoclonaux/immunologie , Cellules cultivées , Humains
15.
Proc Natl Acad Sci U S A ; 110(32): 13109-13, 2013 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-23878230

RÉSUMÉ

Recent evidence indicates there is a role for small membrane vesicles, including exosomes, as vehicles for intercellular communication. Exosomes secreted by most cell types can mediate transfer of proteins, mRNAs, and microRNAs, but their role in the transmission of infectious agents is less established. Recent studies have shown that hepatocyte-derived exosomes containing hepatitis C virus (HCV) RNA can activate innate immune cells, but the role of exosomes in the transmission of HCV between hepatocytes remains unknown. In this study, we investigated whether exosomes transfer HCV in the presence of neutralizing antibodies. Purified exosomes isolated from HCV-infected human hepatoma Huh7.5.1 cells were shown to contain full-length viral RNA, viral protein, and particles, as determined by RT-PCR, mass spectrometry, and transmission electron microscopy. Exosomes from HCV-infected cells were capable of transmitting infection to naive human hepatoma Huh7.5.1 cells and establishing a productive infection. Even with subgenomic replicons, lacking structural viral proteins, exosome-mediated transmission of HCV RNA was observed. Treatment with patient-derived IgGs showed a variable degree of neutralization of exosome-mediated infection compared with free virus. In conclusion, this study showed that hepatic exosomes can transmit productive HCV infection in vitro and are partially resistant to antibody neutralization. This discovery sheds light on neutralizing antibodies resistant to HCV transmission by exosomes as a potential immune evasion mechanism.


Sujet(s)
Exosomes/virologie , Hepacivirus/génétique , ARN viral/génétique , Virion/génétique , Anticorps neutralisants/immunologie , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/virologie , Lignée cellulaire tumorale , Claudine-1/immunologie , Claudine-1/métabolisme , Exosomes/métabolisme , Exosomes/ultrastructure , Hepacivirus/immunologie , Hepacivirus/physiologie , Hépatite C/immunologie , Hépatite C/virologie , Interactions hôte-pathogène , Humains , Immunoglobuline G/immunologie , Spectrométrie de masse , Microscopie confocale , Microscopie électronique à transmission , ARN viral/métabolisme , RT-PCR , Récepteurs éboueurs de classe B/immunologie , Récepteurs éboueurs de classe B/métabolisme , Antigène CD81/immunologie , Antigène CD81/métabolisme , Virion/physiologie , Virion/ultrastructure
16.
PLoS One ; 8(6): e67254, 2013.
Article de Anglais | MEDLINE | ID: mdl-23840641

RÉSUMÉ

The poor response to the combined antiviral therapy of pegylated alfa-interferon and ribavarin for hepatitis C virus (HCV) infection may be linked to mutations in the viral envelope gene E1E2 (env), which can result in escape from the immune response and higher efficacy of viral entry. Mutations that result in failure of therapy most likely require compensatory mutations to achieve sufficient change in envelope structure and function. Compensatory mutations were investigated by determining positions in the E1E2 gene where amino acids (aa) covaried across groups of individuals. We assessed networks of covarying positions in E1E2 sequences that differentiated sustained virological response (SVR) from non-response (NR) in 43 genotype 1a (17 SVR), and 49 genotype 1b (25 SVR) chronically HCV-infected individuals. Binary integer programming over covariance networks was used to extract aa combinations that differed between response groups. Genotype 1a E1E2 sequences exhibited higher degrees of covariance and clustered into 3 main groups while 1b sequences exhibited no clustering. Between 5 and 9 aa pairs were required to separate SVR from NR in each genotype. aa in hypervariable region 1 were 6 times more likely than chance to occur in the optimal networks. The pair 531-626 (EI) appeared frequently in the optimal networks and was present in 6 of 9 NR in one of the 1a clusters. The most frequent pairs representing SVR were 431-481 (EE), 500-522 (QA) in 1a, and 407-434 (AQ) in 1b. Optimal networks based on covarying aa pairs in HCV envelope can indicate features that are associated with failure or success to antiviral therapy.


Sujet(s)
Antiviraux/usage thérapeutique , Hepacivirus/génétique , Hépatite C/traitement médicamenteux , Interféron alpha/usage thérapeutique , Ribavirine/usage thérapeutique , Génotype , Hépatite C/virologie , Humains , Modèles biologiques , Modèles statistiques , Analyse multifactorielle , Phylogenèse , Résultat thérapeutique , Protéines de l'enveloppe virale/génétique
17.
PLoS One ; 8(5): e64221, 2013.
Article de Anglais | MEDLINE | ID: mdl-23704981

RÉSUMÉ

BACKGROUND AND AIMS: Hepatitis C virus (HCV) infection is a challenge to prevent and treat because of the rapid development of drug resistance and escape. Viral entry is required for initiation, spread, and maintenance of infection, making it an attractive target for antiviral strategies. METHODS: Using genetic immunization, we produced four monoclonal antibodies (mAbs) against the HCV host entry factor CD81. The effects of antibodies on inhibition of HCV infection and dissemination were analyzed in HCV permissive human liver cell lines. RESULTS: The anti-CD81 mAbs efficiently inhibited infection by HCV of different genotypes as well as a HCV escape variant selected during liver transplantation and re-infecting the liver graft. Kinetic studies indicated that anti-CD81 mAbs target a post-binding step during HCV entry. In addition to inhibiting cell-free HCV infection, one antibody was also able to block neutralizing antibody-resistant HCV cell-cell transmission and viral dissemination without displaying any detectable toxicity. CONCLUSION: A novel anti-CD81 mAb generated by genetic immunization efficiently blocks HCV spread and dissemination. This antibody will be useful to further unravel the role of virus-host interactions during HCV entry and cell-cell transmission. Furthermore, this antibody may be of interest for the development of antivirals for prevention and treatment of HCV infection.


Sujet(s)
Anticorps monoclonaux/immunologie , Hepacivirus/immunologie , Hepacivirus/physiologie , Anticorps de l'hépatite C/immunologie , Immunisation , Antigène CD81/immunologie , Pénétration virale , Animaux , Anticorps neutralisants/immunologie , Membrane cellulaire/immunologie , Hépatite C/immunologie , Hépatite C/virologie , Humains , Rats , Rat Wistar , Protéines de l'enveloppe virale/immunologie
18.
Hepatology ; 58(4): 1225-35, 2013 Oct.
Article de Anglais | MEDLINE | ID: mdl-23519785

RÉSUMÉ

UNLABELLED: Interferon-alpha (IFN-α) exhibits its antiviral activity through signal transducer and activator of transcription protein (STAT) signaling and the expression of IFN response genes (IRGs). Viral infection has been shown to result in activation of epidermal growth factor receptor (EGFR)-a host cell entry factor used by several viruses, including hepatitis C virus. However, the effect of EGFR activation for cellular antiviral responses is unknown. Here, we uncover cross-talk between EGFR and IFN-α signaling that has a therapeutic effect on IFN-α-based therapies and functional relevance for viral evasion and IFN resistance. We show that combining IFN-α with the EGFR inhibitor, erlotinib, potentiates the antiviral effect of each compound in a highly synergistic manner. The extent of the synergy correlated with reduced STAT3 phosphorylation in the presence of erlotinib, whereas STAT1 phosphorylation was not affected. Furthermore, reduced STAT3 phosphorylation correlated with enhanced expression of suppressors of cytokine signaling 3 (SOCS3) in the presence of erlotinib and enhanced expression of the IRGs, radical S-adenosyl methionine domain containing 2 and myxovirus resistance protein 1. Moreover, EGFR stimulation reduced STAT1 dimerization, but not phosphorylation, indicating that EGFR cross-talk with IFN signaling acts on the STATs at the level of binding DNA. CONCLUSIONS: Our results support a model where inhibition of EGFR signaling impairs STAT3 phosphorylation, leading to enhanced IRG expression and antiviral activity. These data uncover a novel role of EGFR signaling in the antiviral activity of IFN-α and open new avenues of improving the efficacy of IFN-α-based antiviral therapies.


Sujet(s)
Antiviraux/pharmacologie , Récepteurs ErbB/physiologie , Hepacivirus/effets des médicaments et des substances chimiques , Hépatite C/anatomopathologie , Hépatocytes/effets des médicaments et des substances chimiques , Interféron alpha/pharmacologie , Transduction du signal/physiologie , Antiviraux/usage thérapeutique , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/virologie , Lignée cellulaire , Cellules cultivées , Synergie des médicaments , Association de médicaments , Récepteurs ErbB/antagonistes et inhibiteurs , Récepteurs ErbB/effets des médicaments et des substances chimiques , Chlorhydrate d'erlotinib , Hépatite C/traitement médicamenteux , Hépatite C/métabolisme , Hépatocytes/anatomopathologie , Hépatocytes/virologie , Humains , Interféron alpha/usage thérapeutique , Tumeurs du foie/anatomopathologie , Tumeurs du foie/virologie , Quinazolines/pharmacologie , Quinazolines/usage thérapeutique , Récepteurs à activité tyrosine kinase/physiologie , Facteur de transcription STAT-3/métabolisme , Protéine-3 suppressive de la signalisation des cytokine , Protéines SOCS/métabolisme , Résultat thérapeutique
19.
Microb Pathog ; 58: 66-72, 2013 May.
Article de Anglais | MEDLINE | ID: mdl-23499591

RÉSUMÉ

Approximately 170 million individuals, representing 3% of the global population, are infected with hepatitis C virus (HCV). Whereas strategies for antiviral therapies have markedly improved resulting in clinical licensing of direct-acting antivirals, the development of vaccines has been hampered by the high genetic variability of the virus as well as by the lack of suitable animal models for proof-of-concept studies. Nevertheless, there are several promising vaccine candidates in preclinical and clinical development. After a brief summary of the molecular virology and immunology relevant to vaccine development, this review explains the model systems used for preclinical vaccine development, and highlights examples for most recently developed HCV vaccine candidates.


Sujet(s)
Hepacivirus/immunologie , Hépatite C/prévention et contrôle , Vaccins antiviraux/immunologie , Animaux , Découverte de médicament/tendances , Hepacivirus/génétique , Hepacivirus/physiologie , Hépatite C/immunologie , Vaccins antiviraux/génétique
20.
J Infect Dis ; 207(8): 1306-15, 2013 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-23335805

RÉSUMÉ

BACKGROUND: A major challenge for antiviral treatment of hepatitis C virus (HCV) infection is viral resistance, potentially resulting from the high variability of HCV envelope glycoproteins and subsequent selection of strains with enhanced infectivity and/or immune escape. METHODS: We used a bioinformatics and functional approach to investigate whether E1/E2 envelope glycoprotein structure and function were associated with treatment failure in 92 patients infected with HCV genotype 1. RESULTS: Bioinformatics analysis identified 1 sustain virological response (R)-related residue in E1 (219T) and 2 non-SVR (NR)-related molecular signatures in E2 (431A and 642V) in HCV genotype 1a. Two of these positions also appeared in minimal networks separating NR patients from R patients. HCV pseudoparticles (HCVpp) expressing 431A and 642V resulted in a decrease in antibody-mediated neutralization by pretreatment sera. 431A/HCVpp entry into Huh7.5 cells increased with overexpression of CD81 and SR-BI. Moreover, an association of envelope glycoprotein signatures with treatment failure was confirmed in an independent cohort (Virahep-C). CONCLUSIONS: Combined in silico and functional analyses demonstrate that envelope glycoprotein signatures associated with treatment failure result in an alteration of host cell entry factor use and escape from neutralizing antibodies, suggesting that virus-host interactions during viral entry contribute to treatment failure.


Sujet(s)
Biologie informatique/méthodes , Hépatite C/virologie , Protéines de l'enveloppe virale/génétique , Pénétration virale/effets des médicaments et des substances chimiques , Adulte , Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , Antiviraux/pharmacologie , Femelle , Génotype , Cellules HEK293 , Hepacivirus/classification , Hepacivirus/pathogénicité , Hépatite C/traitement médicamenteux , Hépatite C/immunologie , Humains , Échappement immunitaire , Mâle , Adulte d'âge moyen , Mutagenèse dirigée , Mutation , Tests de neutralisation , Ribavirine/pharmacologie , Relation structure-activité , Échec thérapeutique , Protéines de l'enveloppe virale/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...