Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 15 de 15
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
J Hepatol ; 78(2): 343-355, 2023 02.
Article de Anglais | MEDLINE | ID: mdl-36309131

RÉSUMÉ

BACKGROUND & AIMS: Despite recent approvals, the response to treatment and prognosis of patients with advanced hepatocellular carcinoma (HCC) remain poor. Claudin-1 (CLDN1) is a membrane protein that is expressed at tight junctions, but it can also be exposed non-junctionally, such as on the basolateral membrane of the human hepatocyte. While CLDN1 within tight junctions is well characterized, the role of non-junctional CLDN1 and its role as a therapeutic target in HCC remains unexplored. METHODS: Using humanized monoclonal antibodies (mAbs) specifically targeting the extracellular loop of human non-junctional CLDN1 and a large series of patient-derived cell-based and animal model systems we aimed to investigate the role of CLDN1 as a therapeutic target for HCC. RESULTS: Targeting non-junctional CLDN1 markedly suppressed tumor growth and invasion in cell line-based models of HCC and patient-derived 3D ex vivo models. Moreover, the robust effect on tumor growth was confirmed in vivo in a large series of cell line-derived xenograft and patient-derived xenograft mouse models. Mechanistic studies, including single-cell RNA sequencing of multicellular patient HCC tumorspheres, suggested that CLDN1 regulates tumor stemness, metabolism, oncogenic signaling and perturbs the tumor immune microenvironment. CONCLUSIONS: Our results provide the rationale for targeting CLDN1 in HCC and pave the way for the clinical development of CLDN1-specific mAbs for the treatment of advanced HCC. IMPACT AND IMPLICATIONS: Hepatocellular carcinoma (HCC) is associated with high mortality and unsatisfactory treatment options. Herein, we identified the cell surface protein Claudin-1 as a treatment target for advanced HCC. Monoclonal antibodies targeting Claudin-1 inhibit tumor growth in patient-derived ex vivo and in vivo models by modulating signaling, cell stemness and the tumor immune microenvironment. Given the differentiated mechanism of action, the identification of Claudin-1 as a novel therapeutic target for HCC provides an opportunity to break the plateau of limited treatment response. The results of this preclinical study pave the way for the clinical development of Claudin-1-specific antibodies for the treatment of advanced HCC. It is therefore of key impact for physicians, scientists and drug developers in the field of liver cancer and gastrointestinal oncology.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , Humains , Animaux , Souris , Carcinome hépatocellulaire/génétique , Claudine-1/génétique , Tumeurs du foie/génétique , Cancérogènes , Microenvironnement tumoral , Anticorps monoclonaux/pharmacologie , Anticorps monoclonaux/usage thérapeutique , Lignée cellulaire tumorale
2.
Sci Transl Med ; 14(676): eabj4221, 2022 12 21.
Article de Anglais | MEDLINE | ID: mdl-36542691

RÉSUMÉ

Tissue fibrosis is a key driver of end-stage organ failure and cancer, overall accounting for up to 45% of deaths in developed countries. There is a large unmet medical need for antifibrotic therapies. Claudin-1 (CLDN1) is a member of the tight junction protein family. Although the role of CLDN1 incorporated in tight junctions is well established, the function of nonjunctional CLDN1 (njCLDN1) is largely unknown. Using highly specific monoclonal antibodies targeting a conformation-dependent epitope of exposed njCLDN1, we show in patient-derived liver three-dimensional fibrosis and human liver chimeric mouse models that CLDN1 is a mediator and target for liver fibrosis. Targeting CLDN1 reverted inflammation-induced hepatocyte profibrogenic signaling and cell fate and suppressed the myofibroblast differentiation of hepatic stellate cells. Safety studies of a fully humanized antibody in nonhuman primates did not reveal any serious adverse events even at high steady-state concentrations. Our results provide preclinical proof of concept for CLDN1-specific monoclonal antibodies for the treatment of advanced liver fibrosis and cancer prevention. Antifibrotic effects in lung and kidney fibrosis models further indicate a role of CLDN1 as a therapeutic target for tissue fibrosis across organs. In conclusion, our data pave the way for further therapeutic exploration of CLDN1-targeting therapies for fibrotic diseases in patients.


Sujet(s)
Anticorps monoclonaux , Plasticité cellulaire , Animaux , Souris , Humains , Anticorps monoclonaux/pharmacologie , Anticorps monoclonaux/usage thérapeutique , Claudine-1 , Cirrhose du foie/traitement médicamenteux
3.
JCI Insight ; 7(13)2022 07 08.
Article de Anglais | MEDLINE | ID: mdl-35801591

RÉSUMÉ

Hepatocellular carcinoma (HCC) is a leading cause of death among cirrhotic patients, for which chemopreventive strategies are lacking. Recently, we developed a simple human cell-based system modeling a clinical prognostic liver signature (PLS) predicting liver disease progression and HCC risk. In a previous study, we applied our cell-based system for drug discovery and identified captopril, an approved angiotensin converting enzyme (ACE) inhibitor, as a candidate compound for HCC chemoprevention. Here, we explored ACE as a therapeutic target for HCC chemoprevention. Captopril reduced liver fibrosis and effectively prevented liver disease progression toward HCC development in a diethylnitrosamine (DEN) rat cirrhosis model and a diet-based rat model for nonalcoholic steatohepatitis-induced (NASH-induced) hepatocarcinogenesis. RNA-Seq analysis of cirrhotic rat liver tissues uncovered that captopril suppressed the expression of pathways mediating fibrogenesis, inflammation, and carcinogenesis, including epidermal growth factor receptor (EGFR) signaling. Mechanistic data in liver disease models uncovered a cross-activation of the EGFR pathway by angiotensin. Corroborating the clinical translatability of the approach, captopril significantly reversed the HCC high-risk status of the PLS in liver tissues of patients with advanced fibrosis. Captopril effectively prevents fibrotic liver disease progression toward HCC development in preclinical models and is a generic and safe candidate drug for HCC chemoprevention.


Sujet(s)
Captopril , Carcinome hépatocellulaire , Tumeurs du foie , Inhibiteurs de l'enzyme de conversion de l'angiotensine/pharmacologie , Inhibiteurs de l'enzyme de conversion de l'angiotensine/usage thérapeutique , Animaux , Captopril/pharmacologie , Captopril/usage thérapeutique , Carcinogenèse , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/prévention et contrôle , Chimioprévention , Évolution de la maladie , Récepteurs ErbB/métabolisme , Cirrhose du foie/prévention et contrôle , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/prévention et contrôle , Peptidyl-Dipeptidase A/métabolisme , Rats , Activation de la transcription
4.
Nat Commun ; 12(1): 5525, 2021 09 17.
Article de Anglais | MEDLINE | ID: mdl-34535664

RÉSUMÉ

Chronic liver disease and hepatocellular carcinoma (HCC) are life-threatening diseases with limited treatment options. The lack of clinically relevant/tractable experimental models hampers therapeutic discovery. Here, we develop a simple and robust human liver cell-based system modeling a clinical prognostic liver signature (PLS) predicting long-term liver disease progression toward HCC. Using the PLS as a readout, followed by validation in nonalcoholic steatohepatitis/fibrosis/HCC animal models and patient-derived liver spheroids, we identify nizatidine, a histamine receptor H2 (HRH2) blocker, for treatment of advanced liver disease and HCC chemoprevention. Moreover, perturbation studies combined with single cell RNA-Seq analyses of patient liver tissues uncover hepatocytes and HRH2+, CLEC5Ahigh, MARCOlow liver macrophages as potential nizatidine targets. The PLS model combined with single cell RNA-Seq of patient tissues enables discovery of urgently needed targets and therapeutics for treatment of advanced liver disease and cancer prevention.


Sujet(s)
Découverte de médicament , Foie/anatomopathologie , Modèles biologiques , Animaux , Carcinogenèse/anatomopathologie , Carcinome hépatocellulaire/anatomopathologie , Lignée cellulaire tumorale , Chimioprévention , Études de cohortes , AMP cyclique/métabolisme , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Modèles animaux de maladie humaine , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Cellules HEK293 , Hepacivirus/physiologie , Hépatite C/génétique , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Hépatocytes/anatomopathologie , Humains , Surveillance immunologique/effets des médicaments et des substances chimiques , Inflammation/anatomopathologie , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Cirrhose du foie/anatomopathologie , Tumeurs du foie/anatomopathologie , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Macrophages/anatomopathologie , Mâle , Souris knockout , Nizatidine/pharmacologie , Pronostic , Transduction du signal/effets des médicaments et des substances chimiques , Transcriptome/génétique
5.
Life Sci Alliance ; 4(9)2021 09.
Article de Anglais | MEDLINE | ID: mdl-34290079

RÉSUMÉ

Chronic hepatitis B virus (HBV) infection is a major cause of hepatocellular carcinoma (HCC) world-wide. The molecular mechanisms of viral hepatocarcinogenesis are still partially understood. Here, we applied two complementary single-cell RNA-sequencing protocols to investigate HBV-HCC host cell interactions at the single cell level of patient-derived HCC. Computational analyses revealed a marked HCC heterogeneity with a robust and significant correlation between HBV reads and cancer cell differentiation. Viral reads significantly correlated with the expression of HBV-dependency factors such as HLF in different tumor compartments. Analyses of virus-induced host responses identified previously undiscovered pathways mediating viral carcinogenesis, such as E2F- and MYC targets as well as adipogenesis. Mapping of fused HBV-host cell transcripts allowed the characterization of integration sites in individual cancer cells. Collectively, single-cell RNA-Seq unravels heterogeneity and compartmentalization of both, virus and cancer identifying new candidate pathways for viral hepatocarcinogenesis. The perturbation of pro-carcinogenic gene expression even at low HBV levels highlights the need of HBV cure to eliminate HCC risk.


Sujet(s)
Carcinome hépatocellulaire/étiologie , Transformation cellulaire virale , Virus de l'hépatite B/physiologie , Hépatite B/complications , Hépatite B/virologie , Tumeurs du foie/étiologie , Adulte , Sujet âgé , Carcinome hépatocellulaire/mortalité , Carcinome hépatocellulaire/anatomopathologie , Différenciation cellulaire , Lignée cellulaire tumorale , Prédisposition aux maladies , Femelle , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes viraux , Hépatite B chronique/complications , Hépatite B chronique/virologie , Humains , Tumeurs du foie/mortalité , Tumeurs du foie/anatomopathologie , Mâle , Adulte d'âge moyen , Grading des tumeurs , ARN viral , Analyse sur cellule unique/méthodes , Transcriptome , Charge virale
6.
Gut ; 69(2): 380-392, 2020 02.
Article de Anglais | MEDLINE | ID: mdl-31076402

RÉSUMÉ

OBJECTIVE: Infection of human hepatocytes by the hepatitis C virus (HCV) is a multistep process involving both viral and host factors. microRNAs (miRNAs) are small non-coding RNAs that post-transcriptionally regulate gene expression. Given that miRNAs were indicated to regulate between 30% and 75% of all human genes, we aimed to investigate the functional and regulatory role of miRNAs for the HCV life cycle. DESIGN: To systematically reveal human miRNAs affecting the HCV life cycle, we performed a two-step functional high-throughput miRNA mimic screen in Huh7.5.1 cells infected with recombinant cell culture-derived HCV. miRNA targeting was then assessed using a combination of computational and functional approaches. RESULTS: We uncovered miR-501-3p and miR-619-3p as novel modulators of HCV assembly/release. We discovered that these miRNAs regulate O-linked N-acetylglucosamine (O-GlcNAc) transferase (OGT) protein expression and identified OGT and O-GlcNAcylation as regulators of HCV morphogenesis and infectivity. Furthermore, increased OGT expression in patient-derived liver tissue was associated with HCV-induced liver disease and cancer. CONCLUSION: miR-501-3p and miR-619-3p and their target OGT are previously undiscovered regulatory host factors for HCV assembly and infectivity. In addition to its effect on HCV morphogenesis, OGT may play a role in HCV-induced liver disease and hepatocarcinogenesis.


Sujet(s)
Hepacivirus/pathogénicité , Hépatite C chronique/génétique , N-acetylglucosaminyltransferase/physiologie , Régulation de l'expression des gènes/physiologie , Techniques de knock-down de gènes/méthodes , Étude d'association pangénomique/méthodes , Hepacivirus/physiologie , Hépatite C chronique/virologie , Hépatocytes/virologie , Interactions hôte-pathogène/génétique , Humains , Étapes du cycle de vie/génétique , microARN/génétique , Morphogenèse/physiologie , N-acetylglucosaminyltransferase/génétique , Régulation positive , Virulence/génétique
7.
Gastroenterology ; 157(2): 537-551.e9, 2019 08.
Article de Anglais | MEDLINE | ID: mdl-30978357

RÉSUMÉ

BACKGROUND & AIMS: The mechanisms of hepatitis C virus (HCV) infection, liver disease progression, and hepatocarcinogenesis are only partially understood. We performed genomic, proteomic, and metabolomic analyses of HCV-infected cells and chimeric mice to learn more about these processes. METHODS: Huh7.5.1dif (hepatocyte-like cells) were infected with culture-derived HCV and used in RNA sequencing, proteomic, metabolomic, and integrative genomic analyses. uPA/SCID (urokinase-type plasminogen activator/severe combined immunodeficiency) mice were injected with serum from HCV-infected patients; 8 weeks later, liver tissues were collected and analyzed by RNA sequencing and proteomics. Using differential expression, gene set enrichment analyses, and protein interaction mapping, we identified pathways that changed in response to HCV infection. We validated our findings in studies of liver tissues from 216 patients with HCV infection and early-stage cirrhosis and paired biopsy specimens from 99 patients with hepatocellular carcinoma, including 17 patients with histologic features of steatohepatitis. Cirrhotic liver tissues from patients with HCV infection were classified into 2 groups based on relative peroxisome function; outcomes assessed included Child-Pugh class, development of hepatocellular carcinoma, survival, and steatohepatitis. Hepatocellular carcinomas were classified according to steatohepatitis; the outcome was relative peroxisomal function. RESULTS: We quantified 21,950 messenger RNAs (mRNAs) and 8297 proteins in HCV-infected cells. Upon HCV infection of hepatocyte-like cells and chimeric mice, we observed significant changes in levels of mRNAs and proteins involved in metabolism and hepatocarcinogenesis. HCV infection of hepatocyte-like cells significantly increased levels of the mRNAs, but not proteins, that regulate the innate immune response; we believe this was due to the inhibition of translation in these cells. HCV infection of hepatocyte-like cells increased glucose consumption and metabolism and the STAT3 signaling pathway and reduced peroxisome function. Peroxisomes mediate ß-oxidation of very long-chain fatty acids; we found intracellular accumulation of very long-chain fatty acids in HCV-infected cells, which is also observed in patients with fatty liver disease. Cells in livers from HCV-infected mice had significant reductions in levels of the mRNAs and proteins associated with peroxisome function, indicating perturbation of peroxisomes. We found that defects in peroxisome function were associated with outcomes and features of HCV-associated cirrhosis, fatty liver disease, and hepatocellular carcinoma in patients. CONCLUSIONS: We performed combined transcriptome, proteome, and metabolome analyses of liver tissues from HCV-infected hepatocyte-like cells and HCV-infected mice. We found that HCV infection increases glucose metabolism and the STAT3 signaling pathway and thereby reduces peroxisome function; alterations in the expression levels of peroxisome genes were associated with outcomes of patients with liver diseases. These findings provide insights into liver disease pathogenesis and might be used to identify new therapeutic targets.


Sujet(s)
Hepacivirus/pathogénicité , Hépatite C chronique/anatomopathologie , Hépatocytes/anatomopathologie , Foie/anatomopathologie , Animaux , Lignée cellulaire tumorale , Jeux de données comme sujet , Modèles animaux de maladie humaine , Analyse de profil d'expression de gènes , Glucose/métabolisme , Hépatite C chronique/métabolisme , Hépatite C chronique/virologie , Hépatocytes/transplantation , Hépatocytes/virologie , Humains , Foie/cytologie , Foie/virologie , Métabolomique , Souris , Péroxysomes/métabolisme , Péroxysomes/anatomopathologie , Protéomique , Facteur de transcription STAT-3/métabolisme , Chimère obtenue par transplantation
8.
Hepatology ; 68(5): 1695-1709, 2018 11.
Article de Anglais | MEDLINE | ID: mdl-29679386

RÉSUMÉ

Chronic hepatitis B virus (HBV) infection is a major cause of chronic liver disease and cancer worldwide. The mechanisms of viral genome sensing and the evasion of innate immune responses by HBV infection are still poorly understood. Recently, the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) was identified as a DNA sensor. In this study, we investigated the functional role of cGAS in sensing HBV infection and elucidate the mechanisms of viral evasion. We performed functional studies including loss-of-function and gain-of-function experiments combined with cGAS effector gene expression profiling in an infectious cell culture model, primary human hepatocytes, and HBV-infected human liver chimeric mice. Here, we show that cGAS is expressed in the human liver, primary human hepatocytes, and human liver chimeric mice. While naked relaxed-circular HBV DNA is sensed in a cGAS-dependent manner in hepatoma cell lines and primary human hepatocytes, host cell recognition of viral nucleic acids is abolished during HBV infection, suggesting escape from sensing, likely during packaging of the genome into the viral capsid. While the hepatocyte cGAS pathway is functionally active, as shown by reduction of viral covalently closed circular DNA levels in gain-of-function studies, HBV infection suppressed cGAS expression and function in cell culture models and humanized mice. Conclusion: HBV exploits multiple strategies to evade sensing and antiviral activity of cGAS and its effector pathways.


Sujet(s)
Virus de l'hépatite B/pathogénicité , Hépatite B/physiopathologie , Hépatocytes/virologie , Échappement immunitaire/physiologie , Nucléotides cycliques/métabolisme , Animaux , Technique de Western , Techniques de culture cellulaire , ADN viral/immunologie , Analyse de profil d'expression de gènes/méthodes , Hépatite B/immunologie , Hépatocytes/métabolisme , Interactions hôte-pathogène , Humains , Échappement immunitaire/immunologie , Hybridation fluorescente in situ/méthodes , Souris , Réaction de polymérisation en chaine en temps réel
9.
Gut ; 67(4): 736-745, 2018 04.
Article de Anglais | MEDLINE | ID: mdl-28360099

RÉSUMÉ

OBJECTIVE: HCV infection is a leading cause of chronic liver disease and a major indication for liver transplantation. Although direct-acting antivirals (DAAs) have much improved the treatment of chronic HCV infection, alternative strategies are needed for patients with treatment failure. As an essential HCV entry factor, the tight junction protein claudin-1 (CLDN1) is a promising antiviral target. However, genotype-dependent escape via CLDN6 and CLDN9 has been described in some cell lines as a possible limitation facing CLDN1-targeted therapies. Here, we evaluated the clinical potential of therapeutic strategies targeting CLDN1. DESIGN: We generated a humanised anti-CLDN1 monoclonal antibody (mAb) (H3L3) suitable for clinical development and characterised its anti-HCV activity using cell culture models, a large panel of primary human hepatocytes (PHH) from 12 different donors, and human liver chimeric mice. RESULTS: H3L3 pan-genotypically inhibited HCV pseudoparticle entry into PHH, irrespective of donor. Escape was likely precluded by low surface expression of CLDN6 and CLDN9 on PHH. Co-treatment of a panel of PHH with a CLDN6-specific mAb did not enhance the antiviral effect of H3L3, confirming that CLDN6 does not function as an entry factor in PHH from multiple donors. H3L3 also inhibited DAA-resistant strains of HCV and synergised with current DAAs. Finally, H3L3 cured persistent HCV infection in human-liver chimeric uPA-SCID mice in monotherapy. CONCLUSIONS: Overall, these findings underscore the clinical potential of CLDN1-targeted therapies and describe the functional characterisation of a humanised anti-CLDN1 antibody suitable for further clinical development to complement existing therapeutic strategies for HCV.


Sujet(s)
Anticorps monoclonaux/pharmacologie , Antiviraux/pharmacologie , Claudine-1/antagonistes et inhibiteurs , Hepacivirus/effets des médicaments et des substances chimiques , Hépatite C/prévention et contrôle , Hépatocytes/effets des médicaments et des substances chimiques , Facteurs immunologiques/pharmacologie , Animaux , Claudine-1/immunologie , Hépatite C/immunologie , Hépatocytes/immunologie , Hépatocytes/virologie , Humains , Souris , Souris SCID , Résultat thérapeutique
10.
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
11.
Hepatology ; 63(1): 35-48, 2016 Jan.
Article de Anglais | MEDLINE | ID: mdl-26224662

RÉSUMÉ

UNLABELLED: Chronic hepatitis B and D infections are major causes of liver disease and hepatocellular carcinoma worldwide. Efficient therapeutic approaches for cure are absent. Sharing the same envelope proteins, hepatitis B virus and hepatitis delta virus use the sodium/taurocholate cotransporting polypeptide (a bile acid transporter) as a receptor to enter hepatocytes. However, the detailed mechanisms of the viral entry process are still poorly understood. Here, we established a high-throughput infectious cell culture model enabling functional genomics of hepatitis delta virus entry and infection. Using a targeted RNA interference entry screen, we identified glypican 5 as a common host cell entry factor for hepatitis B and delta viruses. CONCLUSION: These findings advance our understanding of virus cell entry and open new avenues for curative therapies. As glypicans have been shown to play a role in the control of cell division and growth regulation, virus-glypican 5 interactions may also play a role in the pathogenesis of virus-induced liver disease and cancer.


Sujet(s)
Glypicanes/physiologie , Virus de l'hépatite B/pathogénicité , Virus de l'hépatite delta/pathogénicité , ARN non traduit/physiologie , Pénétration virale , Cellules cultivées , Humains
12.
J Exp Med ; 211(5): 857-68, 2014 May 05.
Article de Anglais | MEDLINE | ID: mdl-24752298

RÉSUMÉ

The molecular mechanisms that link IFN-λ3 genotypes to differential induction of interferon (IFN)-stimulated genes (ISGs) in the liver of patients with chronic hepatitis C (CHC) are not known. We measured the expression of IFN-λ and of the specific IFN-λ receptor chain (IFN-λR1) in 122 liver biopsies of patients with CHC and 53 control samples. The IFN-λ3 genotype was not associated with differential expression of IFN-λ, but rather IFN-λR1. In a series of 30 primary human hepatocyte (PHH) samples, IFN-λR1 expression was low but could be induced with IFN-α. IFN-α-induced IFN-λR1 expression was significantly stronger in PHHs carrying the minor IFN-λ3 allele. The analysis of liver biopsies of patients with CHC revealed a strong association of high IFN-λR1 expression with elevated ISG expression, with IFN-λ3 minor alleles, and with nonresponse to pegylated IFN-α and ribavirin. The findings provide a missing link between the IFN-λ3 genotype and the associated phenotype of treatment nonresponse.


Sujet(s)
Hépatite C chronique/métabolisme , Interférons/métabolisme , Foie/métabolisme , Récepteur interféron/métabolisme , Biopsie , Technique de Western , Études cas-témoins , Amorces ADN/génétique , Technique d'immunofluorescence , Génotype , Humains , Hybridation fluorescente in situ , Interféron alpha/usage thérapeutique , Microscopie confocale , Polymorphisme de nucléotide simple/génétique , Réaction de polymérisation en chaine en temps réel , Suisse
13.
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
14.
Cell Host Microbe ; 13(3): 302-13, 2013 Mar 13.
Article de Anglais | MEDLINE | ID: mdl-23498955

RÉSUMÉ

Hepatitis C virus (HCV) entry is dependent on coreceptor complex formation between the tetraspanin superfamily member CD81 and the tight junction protein claudin-1 (CLDN1) on the host cell membrane. The receptor tyrosine kinase EGFR acts as a cofactor for HCV entry by promoting CD81-CLDN1 complex formation via unknown mechanisms. We identify the GTPase HRas, activated downstream of EGFR signaling, as a key host signal transducer for EGFR-mediated HCV entry. Proteomic analysis revealed that HRas associates with tetraspanin CD81, CLDN1, and the previously unrecognized HCV entry cofactors integrin ß1 and Ras-related protein Rap2B in hepatocyte membranes. HRas signaling is required for lateral membrane diffusion of CD81, which enables tetraspanin receptor complex assembly. HRas was also found to be relevant for entry of other viruses, including influenza. Our data demonstrate that viruses exploit HRas signaling for cellular entry by compartmentalization of entry factors and receptor trafficking.


Sujet(s)
Claudine-1/métabolisme , Hepacivirus/physiologie , Hépatite C/métabolisme , Protéines proto-oncogènes p21(ras)/métabolisme , Transduction du signal , Antigène CD81/métabolisme , Pénétration virale , Claudine-1/composition chimique , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Hépatite C/génétique , Hépatite C/virologie , Humains , Liaison aux protéines , Multimérisation de protéines , Protéines proto-oncogènes p21(ras)/génétique , Antigène CD81/composition chimique , Tétraspanines/génétique , Tétraspanines/métabolisme
15.
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
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...