Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Hepatol ; 78(2): 343-355, 2023 02.
Article in English | MEDLINE | ID: mdl-36309131

ABSTRACT

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.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Animals , Mice , Carcinoma, Hepatocellular/genetics , Claudin-1/genetics , Liver Neoplasms/genetics , Carcinogens , Tumor Microenvironment , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Cell Line, Tumor
2.
Sci Transl Med ; 14(676): eabj4221, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36542691

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal , Cell Plasticity , Animals , Mice , Humans , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Claudin-1 , Liver Cirrhosis/drug therapy
3.
Int J Mol Sci ; 23(20)2022 Oct 14.
Article in English | MEDLINE | ID: mdl-36293168

ABSTRACT

OCTN2 (SLC22A5) is a carnitine transporter whose main function is the active transport of carnitine into cells. In skeletal muscle and other organs, the regulation of the SLC22A5 gene transcription has been shown to depend on the nuclear transcription factor PPAR-α. Due to the observation that the muscle OCTN2 mRNA level is maintained in PPAR-α knock-out mice and that PGC-1α overexpression in C2C12 myoblasts increases OCTN2 mRNA expression, we suspected additional regulatory pathways for SLC22A5 gene transcription. Indeed, we detected several binding sites of the myocyte-enhancing factor MEF2 in the upstream region of the SLC22A5 gene, and MEF2C/MEF2D stimulated the activity of the OCTN2 promoter in gene reporter assays. This stimulation was increased by PGC-1α and was blunted for a SLC22A5 promoter fragment with a mutated MEF2 binding site. Further, we demonstrated the specific binding of MEF2 to the SLC22A5 gene promoter, and a supershift of the MEF2/DNA complex in electrophoretic mobility shift assays. In immunoprecipitation experiments, we could demonstrate the interaction between PGC-1α and MEF2. In addition, SB203580, a specific inhibitor of p38 MAPK, blocked and interferon-γ stimulated the transcriptional activity of the SLC22A5 gene promoter. Finally, mice with muscle-specific overexpression of OCTN2 showed an increase in OCTN2 mRNA and protein expression in skeletal muscle. In conclusion, we detected and characterized a second stimulatory pathway of SLC22A5 gene transcription in skeletal muscle, which involves the nuclear transcription factor MEF2 and co-stimulation by PGC-1α and which is controlled by the p38 MAPK signaling cascade.


Subject(s)
Carnitine , Peroxisome Proliferator-Activated Receptors , Mice , Animals , Carnitine/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Interferon-gamma/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Muscle, Skeletal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism
4.
Nat Commun ; 12(1): 5525, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34535664

ABSTRACT

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.


Subject(s)
Drug Discovery , Liver/pathology , Models, Biological , Animals , Carcinogenesis/pathology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Chemoprevention , Cohort Studies , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Hepacivirus/physiology , Hepatitis C/genetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Immunologic Surveillance/drug effects , Inflammation/pathology , Liver/drug effects , Liver/metabolism , Liver Cirrhosis/pathology , Liver Neoplasms/pathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Knockout , Nizatidine/pharmacology , Prognosis , Signal Transduction/drug effects , Transcriptome/genetics
5.
Gastroenterology ; 156(8): 2313-2329.e7, 2019 06.
Article in English | MEDLINE | ID: mdl-30836093

ABSTRACT

BACKGROUND & AIMS: Chronic hepatitis C virus (HCV) infection is an important risk factor for hepatocellular carcinoma (HCC). Despite effective antiviral therapies, the risk for HCC is decreased but not eliminated after a sustained virologic response (SVR) to direct-acting antiviral (DAA) agents, and the risk is higher in patients with advanced fibrosis. We investigated HCV-induced epigenetic alterations that might affect risk for HCC after DAA treatment in patients and mice with humanized livers. METHODS: We performed genome-wide ChIPmentation-based ChIP-Seq and RNA-seq analyses of liver tissues from 6 patients without HCV infection (controls), 18 patients with chronic HCV infection, 8 patients with chronic HCV infection cured by DAA treatment, 13 patients with chronic HCV infection cured by interferon therapy, 4 patients with chronic hepatitis B virus infection, and 7 patients with nonalcoholic steatohepatitis in Europe and Japan. HCV-induced epigenetic modifications were mapped by comparative analyses with modifications associated with other liver disease etiologies. uPA/SCID mice were engrafted with human hepatocytes to create mice with humanized livers and given injections of HCV-infected serum samples from patients; mice were given DAAs to eradicate the virus. Pathways associated with HCC risk were identified by integrative pathway analyses and validated in analyses of paired HCC tissues from 8 patients with an SVR to DAA treatment of HCV infection. RESULTS: We found chronic HCV infection to induce specific genome-wide changes in H3K27ac, which correlated with changes in expression of mRNAs and proteins. These changes persisted after an SVR to DAAs or interferon-based therapies. Integrative pathway analyses of liver tissues from patients and mice with humanized livers demonstrated that HCV-induced epigenetic alterations were associated with liver cancer risk. Computational analyses associated increased expression of SPHK1 with HCC risk. We validated these findings in an independent cohort of patients with HCV-related cirrhosis (n = 216), a subset of which (n = 21) achieved viral clearance. CONCLUSIONS: In an analysis of liver tissues from patients with and without an SVR to DAA therapy, we identified epigenetic and gene expression alterations associated with risk for HCC. These alterations might be targeted to prevent liver cancer in patients treated for HCV infection.


Subject(s)
Antiviral Agents/therapeutic use , Carcinoma, Hepatocellular/virology , Hepatitis C, Chronic/pathology , Liver Neoplasms/genetics , Liver Neoplasms/virology , Adult , Animals , Carcinoma, Hepatocellular/genetics , Case-Control Studies , Cohort Studies , Disease Models, Animal , Epigenesis, Genetic , Europe , Female , Gene Expression Regulation, Neoplastic , Hepatitis C, Chronic/complications , Hepatitis C, Chronic/drug therapy , Humans , Japan , Liver Neoplasms/pathology , Male , Mice , Mice, SCID , Random Allocation , Sustained Virologic Response
6.
Gut ; 67(5): 953-962, 2018 05.
Article in English | MEDLINE | ID: mdl-28159835

ABSTRACT

BACKGROUND AND AIMS: HCV infection is a leading risk factor of hepatocellular carcinoma (HCC). However, even after viral clearance, HCC risk remains elevated. HCV perturbs host cell signalling to maintain infection, and derailed signalling circuitry is a key driver of carcinogenesis. Since protein phosphatases are regulators of signalling events, we aimed to identify phosphatases that respond to HCV infection with relevance for hepatocarcinogenesis. METHODS: We assessed mRNA and microRNA (miRNA) expression profiles in primary human hepatocytes, liver biopsies and resections of patients with HCC, and analysed microarray and RNA-seq data from paired liver biopsies of patients with HCC. We revealed changes in transcriptional networks through gene set enrichment analysis and correlated phosphatase expression levels to patient survival and tumour recurrence. RESULTS: We demonstrate that tumour suppressor protein tyrosine phosphatase receptor delta (PTPRD) is impaired by HCV infection in vivo and in HCC lesions of paired liver biopsies independent from tissue inflammation or fibrosis. In liver tissue adjacent to tumour, high PTPRD levels are associated with a dampened transcriptional activity of STAT3, an increase of patient survival from HCC and reduced tumour recurrence after surgical resection. We identified miR-135a-5p as a mechanistic regulator of hepatic PTPRD expression in patients with HCV. CONCLUSIONS: We previously demonstrated that STAT3 is required for HCV infection. We conclude that HCV promotes a STAT3 transcriptional programme in the liver of patients by suppressing its regulator PTPRD via upregulation of miR-135a-5p. Our results show the existence of a perturbed PTPRD-STAT3 axis potentially driving malignant progression of HCV-associated liver disease.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Hepacivirus/pathogenicity , Hepatitis C/complications , Liver Neoplasms/metabolism , MicroRNAs/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Adult , Aged , Aged, 80 and over , Blotting, Western , Carcinogenesis/metabolism , Carcinoma, Hepatocellular/virology , Down-Regulation , Female , Hepatocytes/metabolism , Humans , In Situ Hybridization, Fluorescence , Liver/pathology , Liver Neoplasms/virology , Male , Middle Aged , RNA, Messenger/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction
7.
J Biol Chem ; 292(43): 17928-17938, 2017 10 27.
Article in English | MEDLINE | ID: mdl-28900038

ABSTRACT

Type I (α and ß) and type III (λ) IFNs are induced upon viral infection through host sensory pathways that activate IFN regulatory factors (IRFs) and nuclear factor κB. Secreted IFNs induce autocrine and paracrine signaling through the JAK-STAT pathway, leading to the transcriptional induction of hundreds of IFN-stimulated genes, among them sensory pathway components such as cGAS, STING, RIG-I, MDA5, and the transcription factor IRF7, which enhance the induction of IFN-αs and IFN-λs. This positive feedback loop enables a very rapid and strong host response that, at some point, has to be controlled by negative regulators to maintain tissue homeostasis. Type I IFN signaling is controlled by the inducible negative regulators suppressor of cytokine signaling 1 (SOCS1), SOCS3, and ubiquitin-specific peptidase 18 (USP18). The physiological role of these proteins in IFN-γ signaling has not been clarified. Here we used knockout cell lines and mice to show that IFN-λ signaling is regulated by SOCS1 but not by SOCS3 or USP18. These differences were the basis for the distinct kinetic properties of type I and III IFNs. We found that IFN-α signaling is transient and becomes refractory after hours, whereas IFN-λ provides a long-lasting IFN-stimulated gene induction.


Subject(s)
Gene Expression Regulation/physiology , Interferons/metabolism , Signal Transduction/physiology , Suppressor of Cytokine Signaling 1 Protein/metabolism , Animals , Cell Line, Tumor , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Endopeptidases/genetics , Endopeptidases/metabolism , Humans , Interferon-Induced Helicase, IFIH1/genetics , Interferon-Induced Helicase, IFIH1/metabolism , Interferons/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Receptors, Immunologic , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
8.
Lab Invest ; 96(9): 972-80, 2016 09.
Article in English | MEDLINE | ID: mdl-27428080

ABSTRACT

Notch signaling pathways have recently been implicated in the pathogenesis of metabolic diseases. However, the role of hepatic Notch signaling in glucose and lipid metabolism remains unclear and needs further investigation as it might be a candidate therapeutic target in metabolic diseases such as nonalcoholic steatohepatitis (NASH) and nonalcoholic fatty liver disease (NAFLD). We used hepatocyte-specific Notch1 knockout (KO) mice and liver biopsies from NASH and NAFLD patients to analyze the role of Notch1 in glucose and lipid metabolism. Hepatocyte-specific Notch1 KO mice were fed with a high fat diet (HFD) or a regular diet (RD). We assessed the metabolic phenotype, glucose and insulin tolerance tests, and liver histology. Hepatic mRNA expression was profiled by Affymetrix Mouse Gene arrays and validated by quantitative reverse transcription PCR (qPCR). Akt phosphorylation was visualized by immunoblotting. Gene expression was analyzed in liver biopsies from NASH, NAFLD, and control patients by qPCR. We found that Notch1 KO mice had elevated fasting glucose. Gene expression analysis showed an upregulation of glucose-6-phosphatase, involved in the final step of gluconeogenesis and glucose release from glycogenolysis, and perilipin-5, a regulator of hepatic lipid accumulation. When fed with an HFD KO mice developed overt diabetes and hepatic steatosis. Akt was highly phosphorylated in KO animals and the Foxo1 target gene expression was altered. Accordingly, a reduction in Notch1 and increase in glucose-6-phosphatase and perilipin-5 expression was observed in liver biopsies from NAFLD/NASH compared with controls. Notch1 is a regulator of hepatic glucose and lipid homeostasis. Hepatic impairment of Notch1 expression may be involved in the pathogenesis of human NAFLD/NASH.


Subject(s)
Diabetes Mellitus/genetics , Fatty Liver/genetics , Genetic Predisposition to Disease/genetics , Glucose-6-Phosphatase/genetics , Perilipin-1/genetics , Receptor, Notch1/genetics , Animals , Diabetes Mellitus/etiology , Diet, High-Fat/adverse effects , Fatty Liver/etiology , Gene Expression Profiling/methods , Hepatocytes/metabolism , Humans , Immunoblotting , Liver/metabolism , Liver/pathology , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Notch1/deficiency , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
10.
Nat Biotechnol ; 33(5): 549-554, 2015 May.
Article in English | MEDLINE | ID: mdl-25798937

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Claudin-1/immunology , Hepatitis C/therapy , Liver Cirrhosis/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/immunology , Claudin-1/therapeutic use , Hepacivirus/immunology , Hepacivirus/pathogenicity , Hepatitis C/immunology , Hepatitis C/virology , Hepatocytes/immunology , Humans , Liver Cirrhosis/therapy , Liver Cirrhosis/virology , Mice
11.
Nat Commun ; 5: 5699, 2014 Dec 23.
Article in English | MEDLINE | ID: mdl-25534433

ABSTRACT

Hepatitis C virus (HCV) infections are the major cause of chronic liver disease, cirrhosis and hepatocellular carcinoma worldwide. Both spontaneous and treatment-induced clearance of HCV depend on genetic variation within the interferon-lambda locus, but until now no clear causal relationship has been established. Here we demonstrate that an amino-acid substitution in the IFNλ4 protein changing a proline at position 70 to a serine (P70S) substantially alters its antiviral activity. Patients harbouring the impaired IFNλ4-S70 variant display lower interferon-stimulated gene (ISG) expression levels, better treatment response rates and better spontaneous clearance rates, compared with patients coding for the fully active IFNλ4-P70 variant. Altogether, these data provide evidence supporting a role for the active IFNλ4 protein as the driver of high hepatic ISG expression as well as the cause of poor HCV clearance.


Subject(s)
Hepacivirus/physiology , Hepatitis C/immunology , Interleukins/immunology , Acetyltransferases/genetics , Acetyltransferases/immunology , Cell Line , Cytokines/genetics , Cytokines/immunology , Hepacivirus/genetics , Hepatitis C/genetics , Humans , Interleukins/genetics , Membrane Proteins/genetics , Membrane Proteins/immunology , Oxidoreductases Acting on CH-CH Group Donors , Proteins/genetics , Proteins/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Ubiquitins/genetics , Ubiquitins/immunology
12.
J Immunol ; 192(12): 6037-44, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24821973

ABSTRACT

Recent clinical research suggests a role for vitamin D in the response to IFN-α-based therapy of chronic hepatitis C. Therefore, we aimed to explore the underlying mechanisms in vitro. Huh-7.5 cells harboring subgenomic hepatitis C virus (HCV) replicons or infected with cell culture-derived HCV were exposed to bioactive 1,25-dihydroxyvitamin D3 (calcitriol) with or without IFN-α. In these experiments, calcitriol alone had no effect on the HCV life cycle. However, calcitriol enhanced the inhibitory effect of IFN-α on HCV replication. This effect was based on a calcitriol-mediated increase of IFN-α-induced gene expression. Further mechanistic studies revealed a constitutive inhibitory interaction between the inactive vitamin D receptor (VDR) and Stat1, which was released upon stimulation with calcitriol and IFN-α. As a consequence, IFN-α-induced binding of phosphorylated Stat1 to its DNA target sequences was enhanced by calcitriol. Importantly, and in line with these observations, silencing of the VDR resulted in an enhanced hepatocellular response to IFN-α. Our findings identify the VDR as a novel suppressor of IFN-α-induced signaling through the Jak-STAT pathway.


Subject(s)
Hepacivirus/physiology , Hepatitis C/immunology , Interferon-alpha/pharmacology , Janus Kinases/immunology , Receptors, Calcitriol/immunology , STAT1 Transcription Factor/immunology , Signal Transduction/immunology , Virus Replication/immunology , Calcitriol/pharmacology , Cell Line , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Hepatitis C/drug therapy , Hepatitis C/genetics , Hepatitis C/pathology , Humans , Janus Kinases/genetics , Phosphorylation/drug effects , Phosphorylation/genetics , Phosphorylation/immunology , Receptors, Calcitriol/genetics , STAT1 Transcription Factor/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Virus Replication/drug effects , Vitamins/pharmacology
13.
J Exp Med ; 211(5): 857-68, 2014 May 05.
Article in English | MEDLINE | ID: mdl-24752298

ABSTRACT

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.


Subject(s)
Hepatitis C, Chronic/metabolism , Interferons/metabolism , Liver/metabolism , Receptors, Interferon/metabolism , Biopsy , Blotting, Western , Case-Control Studies , DNA Primers/genetics , Fluorescent Antibody Technique , Genotype , Humans , In Situ Hybridization, Fluorescence , Interferon-alpha/therapeutic use , Microscopy, Confocal , Polymorphism, Single Nucleotide/genetics , Real-Time Polymerase Chain Reaction , Switzerland
14.
Carcinogenesis ; 35(1): 114-22, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23901063

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide. Most HCCs develop in cirrhotic livers. Alcoholic liver disease, chronic hepatitis B and chronic hepatitis C are the most common underlying liver diseases. Hepatitis C virus (HCV)-specific mechanisms that contribute to HCC are presently unknown. Transgenic expression of HCV proteins in the mouse liver induces an overexpression of the protein phosphatase 2A catalytic subunit (PP2Ac). We have previously reported that HCV-induced PP2Ac overexpression modulates histone methylation and acetylation and inhibits DNA damage repair. In this study, we analyze tumor formation and gene expression using HCV transgenic mice that overexpress PP2Ac and liver tissues from patients with HCC. We demonstrate that PP2Ac overexpression interferes with p53-induced apoptosis. Injection of the carcinogen, diethylnitrosamine, induced significantly more and larger liver tumors in HCV transgenic mice that overexpress PP2Ac compared with control mice. In human liver biopsies from patients with HCC, PP2Ac expression was significantly higher in HCC tissue compared with non-tumorous liver tissue from the same patients. Our findings demonstrate an important role of PP2Ac overexpression in liver carcinogenesis and provide insights into the molecular pathogenesis of HCV-induced HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Protein Phosphatase 2/metabolism , Animals , Biopsy , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/enzymology , Diethylnitrosamine/toxicity , Disease Models, Animal , Etoposide/analogs & derivatives , Etoposide/pharmacology , Gene Expression Regulation, Enzymologic , Hepacivirus/genetics , Hepacivirus/pathogenicity , Hepatitis B, Chronic/enzymology , Hepatitis B, Chronic/pathology , Humans , Liver Neoplasms/chemically induced , Liver Neoplasms/enzymology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organophosphorus Compounds/pharmacology , Phosphorylation/drug effects , Protein Phosphatase 2/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
15.
J Exp Med ; 210(6): 1109-16, 2013 Jun 03.
Article in English | MEDLINE | ID: mdl-23712427

ABSTRACT

Approximately 3% of the world population is chronically infected with the hepatitis C virus (HCV), with potential development of cirrhosis and hepatocellular carcinoma. Despite the availability of new antiviral agents, treatment remains suboptimal. Genome-wide association studies (GWAS) identified rs12979860, a polymorphism nearby IL28B, as an important predictor of HCV clearance. We report the identification of a novel TT/-G polymorphism in the CpG region upstream of IL28B, which is a better predictor of HCV clearance than rs12979860. By using peripheral blood mononuclear cells (PBMCs) from individuals carrying different allelic combinations of the TT/-G and rs12979860 polymorphisms, we show that induction of IL28B and IFN-γ-inducible protein 10 (IP-10) mRNA relies on TT/-G, but not rs12979860, making TT/-G the only functional variant identified so far. This novel step in understanding the genetic regulation of IL28B may have important implications for clinical practice, as the use of TT/G genotyping instead of rs12979860 would improve patient management.


Subject(s)
Hepacivirus/physiology , Hepatitis C, Chronic/genetics , Hepatitis C, Chronic/virology , Interleukins/biosynthesis , Alleles , Chemokine CXCL10/biosynthesis , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Genotype , Hepatitis C, Chronic/immunology , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interferon-gamma/immunology , Interferons , Interleukins/genetics , Interleukins/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Polymorphism, Genetic , RNA, Messenger/genetics
16.
Cell Host Microbe ; 13(3): 302-13, 2013 Mar 13.
Article in English | MEDLINE | ID: mdl-23498955

ABSTRACT

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.


Subject(s)
Claudin-1/metabolism , Hepacivirus/physiology , Hepatitis C/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , Tetraspanin 28/metabolism , Virus Internalization , Claudin-1/chemistry , ErbB Receptors/genetics , ErbB Receptors/metabolism , Hepatitis C/genetics , Hepatitis C/virology , Humans , Protein Binding , Protein Multimerization , Proto-Oncogene Proteins p21(ras)/genetics , Tetraspanin 28/chemistry , Tetraspanins/genetics , Tetraspanins/metabolism
17.
Hepatology ; 58(4): 1225-35, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23519785

ABSTRACT

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.


Subject(s)
Antiviral Agents/pharmacology , ErbB Receptors/physiology , Hepacivirus/drug effects , Hepatitis C/pathology , Hepatocytes/drug effects , Interferon-alpha/pharmacology , Signal Transduction/physiology , Antiviral Agents/therapeutic use , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Line , Cells, Cultured , Drug Synergism , Drug Therapy, Combination , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/drug effects , Erlotinib Hydrochloride , Hepatitis C/drug therapy , Hepatitis C/metabolism , Hepatocytes/pathology , Hepatocytes/virology , Humans , Interferon-alpha/therapeutic use , Liver Neoplasms/pathology , Liver Neoplasms/virology , Quinazolines/pharmacology , Quinazolines/therapeutic use , Receptor Protein-Tyrosine Kinases/physiology , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , Treatment Outcome
18.
J Hepatol ; 58(4): 743-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23207140

ABSTRACT

BACKGROUND & AIMS: The toll-like receptor 9 (TLR9) agonist IMO-2125 is currently evaluated in clinical trials for chronic hepatitis C therapy. The aim of this study was to investigate the in vivo mode of action of a closely related compound, referred to as immunomodulatory oligonucleotide (IMO). METHODS: We analyzed the Jak-STAT pathway activation and induction of interferon-stimulated genes in the liver of wild type, interferon-α/ß receptor-deficient and interferon-γ-deficient mice, after administration of IMO. RESULTS: IMO induced a prolonged activation of the Jak-STAT pathway and upregulation of interferon-stimulated genes in the mouse liver. Contrary to the response observed after interferon-α injection, the signalling induced by IMO was not abrogated following repeated administration. At early time points after IMO injection, STAT1 phosphorylation and interferon-stimulated gene induction required a functional interferon-α/ß receptor, whereas at the later time points, the activation was type I interferon-independent. Microarray analysis revealed that IMO induced a broad transcriptional response in the mouse liver. This included upregulation of cytokine and chemokine genes responsible for recruitment of IFN-γ producers, such as T cells and natural killer cells. Interferon-γ-deficient mice showed a transient response to IMO, demonstrating the central role of interferon-γ in sustained activation of Jak-STAT pathway by IMO. CONCLUSIONS: The bimodal kinetics of response to IMO in the mouse liver are driven by the sequential endogenous production of type I and II interferons. The lack of refractoriness to IMO, combined with the long-lasting induction of interferon-stimulated genes, reveals a favourable pharmacodynamics profile of this novel TLR9 agonist for the treatment of chronic viral hepatitis.


Subject(s)
Interferon Type I/biosynthesis , Interferon-gamma/biosynthesis , Liver/drug effects , Liver/immunology , Toll-Like Receptor 9/agonists , Animals , Chemokines/genetics , Cytokines/genetics , Immunologic Factors/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligodeoxyribonucleotides/pharmacology , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 9/deficiency , Toll-Like Receptor 9/genetics , Transcriptional Activation/drug effects , Up-Regulation/drug effects
19.
Gastroenterology ; 143(3): 777-786.e6, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22677194

ABSTRACT

BACKGROUND & AIMS: Approximately 50% of patients with chronic hepatitis C (CHC) have a sustained virologic response to treatment with pegylated interferon (pegIFN)-α and ribavirin. Nonresponse to treatment is associated with constitutively increased expression of IFN-stimulated genes (ISGs) in the liver. Treatment of patients with acute hepatitis C (AHC) is more effective, with sustained virologic response rates greater than 90%. We investigated mechanisms of the different responses of patients with CHC and AHC to pegIFN-α therapy. METHODS: We analyzed IFN signaling and ISG expression in liver samples from patients with AHC, patients with CHC, and individuals without hepatitis C (controls) using microarray, immunohistochemical, and protein analyses. Findings were compared with those from primary human hepatocytes stimulated with IFN-α or IFN-γ, as reference sets. RESULTS: Expression levels of hundreds of genes, primarily those regulated by IFN-γ, were altered in liver samples from patients with AHC compared with controls. Expression of IFN-γ-stimulated genes was induced in liver samples from patients with AHC, whereas expression of IFN-α-stimulated genes was induced in samples from patients with CHC. In an expression analysis of negative regulators of IFN-α signaling, we did not observe differences in expression of suppresor of cytokine signaling 1 or SOCS3 between liver samples from patients with AHC and those with CHC. However, USP18 (another negative regulator of IFN-α signaling), was up-regulated in liver samples of patients with CHC that did not respond to therapy, but not in AHC. CONCLUSIONS: Differences in expression of ISGs might account for the greater response of patients with AHC, compared with those with CHC, to treatment with pegIFN-α and ribavirin. Specifically, USP18 is up-regulated in liver samples of patients with CHC that did not respond to therapy, but not in patients with AHC.


Subject(s)
Antiviral Agents/therapeutic use , Endopeptidases/genetics , Hepatitis C, Chronic/drug therapy , Hepatitis C/drug therapy , Interferon-alpha/therapeutic use , Interferon-gamma/metabolism , Liver/drug effects , Polyethylene Glycols/therapeutic use , Ribavirin/therapeutic use , Acute Disease , Adolescent , Adult , Biopsy , Blotting, Western , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Drug Resistance, Viral/genetics , Drug Therapy, Combination , Endopeptidases/metabolism , Female , Gene Expression Profiling/methods , Gene Expression Regulation , Hepacivirus/pathogenicity , Hepatitis C/diagnosis , Hepatitis C/genetics , Hepatitis C/metabolism , Hepatitis C, Chronic/diagnosis , Hepatitis C, Chronic/genetics , Hepatitis C, Chronic/metabolism , Host-Pathogen Interactions , Humans , Immunohistochemistry , Liver/metabolism , Liver/pathology , Liver/virology , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Phosphorylation , Recombinant Proteins/therapeutic use , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Switzerland , Time Factors , Transcription, Genetic , Treatment Outcome , Ubiquitin Thiolesterase
20.
Hepatology ; 53(4): 1154-63, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21480323

ABSTRACT

UNLABELLED: Therapy of chronic hepatitis C with pegylated interferon α (pegIFN-α) and ribavirin achieves sustained virological responses in approximately half of the patients. Nonresponse to treatment is associated with constitutively increased expression of IFN-stimulated genes in the liver already before therapy. This activation of the endogenous IFN system could prevent cells from responding to therapeutically injected (peg)IFN-α, because prolonged stimulation of cells with IFN-α induces desensitization of the IFN signal transduction pathway. Whether all types of IFNs induce refractoriness in the liver is presently unknown. We therefore treated mice with multiple injections and different combinations of IFN-α, IFN-ß, IFN-γ, and IFN-λ. Pretreatment of mice with IFN-α, IFN-ß, and IFN-λ induced a strong expression of the negative regulator ubiquitin-specific peptidase 18 in the liver and gut. As a result, IFN-α signaling was significantly reduced when mice where reinjected 16 hours after the first injection. Surprisingly, both IFN-ß and IFN-λ could activate the Janus kinase-signal transducer and activator of transcription (STAT) pathway and the expression of IFN-stimulated genes despite high levels of ubiquitin-specific peptidase 18. IFN-λ treatment of human liver biopsies ex vivo resulted in strong and maintained phosphorylation of STAT1, whereas IFN-α-induced STAT1 activation was transient. CONCLUSION: Contrary to the action of IFN-α, IFN-ß, and IFN-λ signaling in the liver does not become refractory during repeated stimulation of the IFN signal transduction pathway. The sustained efficacy of IFN-ß and IFN-λ could be an important advantage for the treatment patients who are nonresponders to pegIFN-α, through a preactivated endogenous IFN system.


Subject(s)
Cytokines/physiology , Interferon-alpha/pharmacology , Interferon-beta/pharmacology , Interferon-gamma/pharmacology , Liver/drug effects , Signal Transduction/physiology , Animals , Cell Line, Tumor , Endopeptidases/metabolism , Hepatitis C, Chronic/physiopathology , Humans , Interferon Inducers/pharmacology , Intestine, Small/drug effects , Janus Kinases/metabolism , Liver/physiology , Male , Mice , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , Ubiquitin Thiolesterase
SELECTION OF CITATIONS
SEARCH DETAIL
...