Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
J Hepatol ; 79(1): 93-108, 2023 07.
Article in English | MEDLINE | ID: mdl-36868481

ABSTRACT

BACKGROUND & AIMS: Cholangiocarcinoma (CCA), heterogeneous biliary tumours with dismal prognosis, lacks accurate early diagnostic methods especially important for individuals at high-risk (i.e. those with primary sclerosing cholangitis [PSC]). Here, we searched for protein biomarkers in serum extracellular vesicles (EVs). METHODS: EVs from patients with isolated PSC (n = 45), concomitant PSC-CCA (n = 44), PSC who developed CCA during follow-up (PSC to CCA; n = 25), CCAs from non-PSC aetiology (n = 56), and hepatocellular carcinoma (n = 34) and healthy individuals (n = 56) were characterised by mass spectrometry. Diagnostic biomarkers for PSC-CCA, non-PSC CCA, or CCAs regardless of aetiology (Pan-CCAs) were defined and validated by ELISA. Their expression was evaluated in CCA tumours at a single-cell level. Prognostic EV biomarkers for CCA were investigated. RESULTS: High-throughput proteomics of EVs identified diagnostic biomarkers for PSC-CCA, non-PSC CCA, or Pan-CCA, and for the differential diagnosis of intrahepatic CCA and hepatocellular carcinoma, which were cross-validated by ELISA using total serum. Machine learning-based algorithms disclosed CRP/FIBRINOGEN/FRIL for the diagnosis of PSC-CCA (local disease [LD]) vs. isolated PSC (AUC = 0.947; odds ratio [OR] =36.9) and, combined with carbohydrate antigen 19-9, overpowers carbohydrate antigen 19-9 alone. CRP/PIGR/VWF allowed the diagnosis of LD non-PSC CCAs vs. healthy individuals (AUC = 0.992; OR = 387.5). It is noteworthy that CRP/FRIL accurately diagnosed LD Pan-CCA (AUC = 0.941; OR = 89.4). Levels of CRP/FIBRINOGEN/FRIL/PIGR showed predictive capacity for CCA development in PSC before clinical evidence of malignancy. Multi-organ transcriptomic analysis revealed that serum EV biomarkers were mostly expressed in hepatobiliary tissues, and single-cell RNA sequencing and immunofluorescence analysis of CCA tumours showed their presence mainly in malignant cholangiocytes. Multivariable analysis unveiled EV prognostic biomarkers, with COMP/GNAI2/CFAI and ACTN1/MYCT1/PF4V associated negatively and positively with patients' survival, respectively. CONCLUSIONS: Serum EVs contain protein biomarkers for the prediction, early diagnosis, and prognostication of CCA that are detectable using total serum, representing a tumour cell-derived liquid biopsy tool for personalised medicine. IMPACT AND IMPLICATIONS: The accuracy of current imaging tests and circulating tumour biomarkers for cholangiocarcinoma (CCA) diagnosis is far from satisfactory. Most CCAs are considered sporadic, although up to 20% of patients with primary sclerosing cholangitis (PSC) develop CCA during their lifetime, constituting a major cause of PSC-related death. This international study has proposed protein-based and aetiology-related logistic models with predictive, diagnostic, or prognostic capacities by combining two to four circulating protein biomarkers, moving a step forward into personalised medicine. These novel liquid biopsy tools may allow the (i) easy and non-invasive diagnosis of sporadic CCAs, (ii) identification of patients with PSC with higher risk for CCA development, (iii) establishment of cost-effective surveillance programmes for the early detection of CCA in high-risk populations (e.g. PSC), and (iv) prognostic stratification of patients with CCA, which, altogether, may increase the number of cases eligible for potentially curative options or to receive more successful treatments, decreasing CCA-related mortality.


Subject(s)
Bile Duct Neoplasms , Carcinoma, Hepatocellular , Cholangiocarcinoma , Cholangitis, Sclerosing , Liver Neoplasms , Humans , Cholangitis, Sclerosing/complications , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/complications , Bile Duct Neoplasms/pathology , Cholangiocarcinoma/diagnosis , Cholangiocarcinoma/etiology , Cholangiocarcinoma/metabolism , Biomarkers, Tumor , Early Diagnosis , Liquid Biopsy , Bile Ducts, Intrahepatic/pathology , Liver Neoplasms/etiology , Liver Neoplasms/complications , Carbohydrates , Nuclear Proteins
2.
Br J Cancer ; 127(9): 1701-1708, 2022 11.
Article in English | MEDLINE | ID: mdl-35986087

ABSTRACT

BACKGROUND: Advanced cholangiocarcinoma has a poor prognosis. Molecular targeted approaches have been proposed for patients after progression under first-line chemotherapy treatment. Here, molecular profiling of intrahepatic cholangiocarcinoma in combination with a comprehensive umbrella concept was applied in a real-world setting. METHODS: In total, 101 patients received molecular profiling and matched treatment based on interdisciplinary tumour board decisions in a tertiary care setting. Parallel DNA and RNA sequencing of formalin-fixed paraffin-embedded tumour tissue was performed using large panels. RESULTS: Genetic alterations were detected in 77% of patients and included gene fusions in 21 patients. The latter recurrently involved the FGFR2 and the NRG1 gene loci. The most commonly altered genes were BAP1, ARID1A, FGFR2, IDH1, CDKN2A, CDKN2B, PIK3CA, TP53, ATM, IDH2, BRAF, SMARCA4 and FGFR3. Molecular targets were detected in 59% of patients. Of these, 32% received targeted therapy. The most relevant reason for not initiating therapy was the deterioration of performance status. Patients receiving a molecular-matched therapy showed a significantly higher survival probability compared to patients receiving conventional chemotherapy only (HR: 2.059, 95% CI: 0.9817-4.320, P < 0.01). CONCLUSIONS: Molecular profiling can be successfully translated into clinical treatment of intrahepatic cholangiocarcinoma patients and is associated with prolonged survival of patients receiving a molecular-matched treatment.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Humans , Proto-Oncogene Proteins B-raf/genetics , Precision Medicine , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Mutation , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Class I Phosphatidylinositol 3-Kinases/genetics , Formaldehyde/therapeutic use , DNA Helicases/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics
3.
EMBO J ; 37(17)2018 09 03.
Article in English | MEDLINE | ID: mdl-30037824

ABSTRACT

The mammalian Hippo signaling pathway, through its effectors YAP and TAZ, coerces epithelial progenitor cell expansion for appropriate tissue development or regeneration upon damage. Its ability to drive rapid tissue growth explains why many oncogenic events frequently exploit this pathway to promote cancer phenotypes. Indeed, several tumor types including basal cell carcinoma (BCC) show genetic aberrations in the Hippo (or YAP/TAZ) regulators. Here, we uncover that while YAP is dispensable for homeostatic epidermal regeneration, it is required for BCC development. Our clonal analyses further demonstrate that the few emerging Yap-null dysplasia have lower fitness and thus are diminished as they progress to invasive BCC Mechanistically, YAP depletion in BCC tumors leads to effective impairment of the JNK-JUN signaling, a well-established tumor-driving cascade. Importantly, in this context, YAP does not influence canonical Wnt or Hedgehog signaling. Overall, we reveal Hippo signaling as an independent promoter of BCC pathogenesis and thereby a viable target for drug-resistant BCC.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenocarcinoma/metabolism , Drug Resistance, Neoplasm , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Transcription Factor AP-1/metabolism , Wnt Signaling Pathway , Adaptor Proteins, Signal Transducing/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Cell Cycle Proteins , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Mice , Mice, Knockout , Phosphoproteins/genetics , Proto-Oncogene Proteins c-jun/genetics , Transcription Factor AP-1/genetics , YAP-Signaling Proteins
4.
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
5.
Hepatology ; 62(5): 1497-510, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26173433

ABSTRACT

UNLABELLED: The Yes-associated protein (YAP)/Hippo pathway has been implicated in tissue development, regeneration, and tumorigenesis. However, its role in cholangiocarcinoma (CC) is not established. We show that YAP activation is a common feature in CC patient biopsies and human CC cell lines. Using microarray expression profiling of CC cells with overexpressed or down-regulated YAP, we show that YAP regulates genes involved in proliferation, apoptosis, and angiogenesis. YAP activity promotes CC growth in vitro and in vivo by functionally interacting with TEAD transcription factors (TEADs). YAP activity together with TEADs prevents apoptosis induced by cytotoxic drugs, whereas YAP knockdown sensitizes CC cells to drug-induced apoptosis. We further show that the proangiogenic microfibrillar-associated protein 5 (MFAP5) is a direct transcriptional target of YAP/TEAD in CC cells and that secreted MFAP5 promotes tube formation of human microvascular endothelial cells. High YAP activity in human CC xenografts and clinical samples correlates with increased MFAP5 expression and CD31(+) vasculature. CONCLUSIONS: These findings establish YAP as a key regulator of proliferation and antiapoptotic mechanisms in CC and provide first evidence that YAP promotes angiogenesis by regulating the expression of secreted proangiogenic proteins.


Subject(s)
Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic , Cholangiocarcinoma/pathology , DNA-Binding Proteins/physiology , Drug Resistance, Neoplasm , Neovascularization, Pathologic/etiology , Nuclear Proteins/physiology , Transcription Factors/physiology , Animals , Apoptosis , Bile Duct Neoplasms/blood supply , Bile Duct Neoplasms/drug therapy , Cell Cycle Proteins , Cell Proliferation , Cholangiocarcinoma/blood supply , Cholangiocarcinoma/drug therapy , Contractile Proteins/genetics , Female , Gene Expression Regulation, Neoplastic , Glycoproteins/genetics , Humans , Intercellular Signaling Peptides and Proteins , Mice , Oncogenes , TEA Domain Transcription Factors
6.
Lab Invest ; 95(3): 351-62, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25418579

ABSTRACT

Hepatic angiosarcoma (AS) is a rare and highly aggressive tumor of endothelial origin with dismal prognosis. Studies of the molecular biology of AS and treatment options are limited as animal models are rare. We have previously shown that inducible knockout of Notch1 in mice leads to spontaneous formation of hepatic AS. The aims of this study were to: (1) establish and characterize a cell line derived from this murine AS, (2) identify molecular pathways involved in the pathogenesis and potential therapeutic targets, and (3) generate a tumor transplantation model. AS cells retained specific endothelial properties such as tube formation activity, as well as expression of CD31 and Von Willebrand factor. However, electron microscopy analysis revealed signs of dedifferentiation with loss of fenestrae and loss of contact inhibition. Microarray and pathway analysis showed substantial changes in gene expression and revealed activation of the Myc pathway. Exposing the AS cells to sorafenib reduced migration, filopodia dynamics, and cell proliferation but did not induce apoptosis. In addition, sorafenib suppressed ERK phosphorylation and expression of cyclin D2. Injection of AS cells into NOD/SCID mice resulted in formation of undifferentiated tumors, confirming the tumorigenic potential of these cells. In summary, we established and characterized a murine model of spontaneous AS formation and hepatic AS cell lines as a useful in vitro tool. Our data demonstrate antitumor activity of sorafenib in AS cells with potent inhibition of migration, filopodia formation, and cell proliferation, supporting further evaluation of sorafenib as a novel treatment strategy. In addition, AS cell transplantation provides a subcutaneous tumor model useful for in vivo preclinical drug testing.


Subject(s)
Disease Models, Animal , Hemangiosarcoma/pathology , Liver Neoplasms/pathology , Liver/pathology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Gene Expression Regulation, Neoplastic/drug effects , Hemangiosarcoma/genetics , Hemangiosarcoma/metabolism , Immunohistochemistry , Liver/metabolism , Liver/ultrastructure , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Microscopy, Electron , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Oligonucleotide Array Sequence Analysis , Phenylurea Compounds/pharmacology , Receptor, Notch1/deficiency , Receptor, Notch1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sorafenib , Tumor Burden/genetics
7.
Hepatology ; 59(1): 24-34, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23775920

ABSTRACT

UNLABELLED: Hepatitis C Virus (HCV) entry involves at least four cellular factors, including CD81, the scavenger receptor class B type I (SCARB-1), occludin (OCLN), and claudin-1 (CLDN1). In addition, CLDN6 and CLDN9 have been shown to substitute for CLDN1 as HCV entry factors in human nonliver cells. We examined the role of different CLDN proteins during HCV entry by using cell lines expressing either predominantly CLDN1 (Huh-7.5) or CLDN6 (HuH6). Huh-7.5 cells were susceptible to all tested HCV isolates, whereas HuH6 cells were only permissive to some viral strains. Silencing of CLDN6 in HuH6 cells revealed that these cells are infected in a CLDN6-dependent fashion, and ectopic expression of CLDN1 or CLDN6 in 293T cells lacking endogenous CLDN expression confirmed that only some HCV strains efficiently use CLDN6 for infection. CLDN1-specific neutralizing antibodies (Abs) fully abrogated infection of Huh-7.5 cells by isolates that use CLDN1 only, whereas viruses with broad CLDN tropism were only partially inhibited by these Abs. Importantly, infection by these latter strains in the presence of anti-CLDN1 Ab was further reduced by silencing CLDN6, suggesting that viruses with broad CLDN usage escape CLDN1-specific Abs by utilization of CLDN6. Messenger RNA (mRNA) levels of HCV entry factors in liver biopsies of HCV patients infected with different genotype and with variable degree of liver fibrosis were determined. Uniformly high levels of CD81, SCARB-1, OCLN, and CLDN1 mRNA were detected. In contrast, abundance of CLDN6 mRNA was highly variable between patients. CONCLUSION: These findings highlight differential CLDN usage by HCV isolates, which may evolve based on variable expression of CLDN proteins in human liver cells. Broad CLDN tropism may facilitate viral escape from CLDN1-specific therapeutic strategies.


Subject(s)
Claudin-1/metabolism , Claudins/metabolism , Hepacivirus/physiology , Viral Tropism , Virus Internalization , Antibodies , Biopsy , Cell Line, Tumor , Claudin-1/immunology , HEK293 Cells , Humans , Liver/metabolism , Liver/pathology , RNA, Messenger/metabolism
8.
Hepatology ; 59(6): 2121-30, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24122862

ABSTRACT

UNLABELLED: Approximately 50% of patients with chronic hepatitis C (CHC) have ongoing expression of interferon stimulated genes (ISGs) in the liver. It is unclear why this endogenous antiviral response is inefficient in eradicating the infection. Several viral escape strategies have been identified in vitro, including inhibition of interferon (IFN) induction and ISG messenger RNA (mRNA) translation. The in vivo relevance of these mechanisms is unknown, because reliable methods to identify hepatitis C virus (HCV)-infected cells in human liver are lacking. We developed a highly sensitive in situ hybridization (ISH) system capable of HCV RNA and ISG mRNA detection in human liver biopsies and applied it to study the interaction of HCV with the endogenous IFN system. We simultaneously monitored HCV RNA and ISG mRNA using HCV isolate- and ISG mRNA-specific probes in liver biopsy sections from 18 CHC patients. The signals were quantified at the single-cell resolution in a series of random high-power fields. The proportion of infected hepatocytes ranged from 1%-54% and correlated with viral load, but not with HCV genotype or ISG expression. Infected cells occurred in clusters, pointing to cell-to-cell spread as the predominant mode of HCV transmission. ISG mRNAs were readily detected in HCV-infected cells, challenging previously proposed mechanisms of viral interference with the immune system. Conversely, infected cells and neighboring cells showed increased ISG mRNA levels, demonstrating that the stimulus driving ISG expression originates from HCV-infected hepatocytes. CONCLUSION: HCV infection in human hepatocytes during CHC does not efficiently interfere with IFN induction, IFN signaling, or transcription of ISG mRNA.


Subject(s)
Gene Expression Regulation, Viral , Hepatitis C/virology , Interferons/physiology , Liver/virology , Hepatitis C/genetics , Hepatitis C/metabolism , Hepatocytes/virology , Humans , In Situ Hybridization , Liver/metabolism , RNA, Viral/genetics , Viral Load/genetics
9.
Hepatology ; 59(1): 78-88, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23873628

ABSTRACT

UNLABELLED: Only humans and chimpanzees are susceptible to chronic infection by hepatitis C virus (HCV). The restricted species tropism of HCV is determined by distinct host factor requirements at different steps of the viral life cycle. In addition, effective innate immune targeting precludes efficient propagation of HCV in nonhuman cells. Species-specificity of HCV host factor usage for cell entry and virus release has been explored. However, the reason for inefficient HCV RNA replication efficiency in mouse liver cells remains elusive. To address this, we generated novel mouse liver-derived cell lines with specific lesions in mitochondrial antiviral signaling protein (MAVS), interferon regulatory factor 3 (IRF3), or Interferon-α/ß receptor (IFNAR) by in vivo immortalization. Blunted innate immune responses in these cells modestly increased HCV RNA replication. However, ectopic expression of liver-specific human microRNA 122 (miR-122) further boosted RNA replication in all knockout cell lines. Remarkably, MAVS(-/-) miR-122 cells sustained vigorous HCV RNA replication, attaining levels comparable to the highly permissive human hepatoma cell line Huh-7.5. RNA replication was dependent on mouse cyclophilin and phosphatidylinositol-4 kinase III alpha (PI4KIIIα) and was also observed after transfection of full-length viral RNA. Additionally, ectopic expression of either human or mouse apolipoprotein E (ApoE) was sufficient to permit release of infectious particles. Finally, expression of human entry cofactors rendered these cells permissive to HCV infection, thus confirming that all steps of the HCV replication cycle can be reconstituted in mouse liver-derived cells. CONCLUSION: Blunted innate immunity, abundant miR-122, and HCV entry factor expression permits propagation of HCV in mouse liver-derived cell lines.


Subject(s)
Hepacivirus/physiology , Virus Replication , 1-Phosphatidylinositol 4-Kinase/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apolipoproteins E/metabolism , Cell Line, Tumor , Cyclophilins/metabolism , Humans , Immunity, Innate , Liver/virology , Mice , Mice, Knockout , MicroRNAs/metabolism , RNA, Viral/metabolism , Virus Internalization
10.
Hepatology ; 59(2): 423-33, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23929719

ABSTRACT

UNLABELLED: The hepatitis C virus (HCV) NS3-4A protease is not only an essential component of the viral replication complex and a prime target for antiviral intervention but also a key player in the persistence and pathogenesis of HCV. It cleaves and thereby inactivates two crucial adaptor proteins in viral RNA sensing and innate immunity, mitochondrial antiviral signaling protein (MAVS) and TRIF, a phosphatase involved in growth factor signaling, T-cell protein tyrosine phosphatase (TC-PTP), and the E3 ubiquitin ligase component UV-damaged DNA-binding protein 1 (DDB1). Here we explored quantitative proteomics to identify novel cellular substrates of the NS3-4A protease. Cell lines inducibly expressing the NS3-4A protease were analyzed by stable isotopic labeling using amino acids in cell culture (SILAC) coupled with protein separation and mass spectrometry. This approach identified the membrane-associated peroxidase GPx8 as a bona fide cellular substrate of the HCV NS3-4A protease. Cleavage by NS3-4A occurs at Cys 11, removing the cytosolic tip of GPx8, and was observed in different experimental systems as well as in liver biopsies from patients with chronic HCV. Overexpression and RNA silencing studies revealed that GPx8 is involved in viral particle production but not in HCV entry or RNA replication. CONCLUSION: We provide proof-of-concept for the use of quantitative proteomics to identify cellular substrates of a viral protease and describe GPx8 as a novel proviral host factor targeted by the HCV NS3-4A protease.


Subject(s)
Hepatitis C, Chronic/metabolism , Peptide Hydrolases/metabolism , Peroxidases/metabolism , Proteomics/methods , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Biopsy , Cell Line , Hepacivirus/drug effects , Hepatitis C, Chronic/pathology , Humans , Liver/drug effects , Liver/metabolism , Liver/pathology , Molecular Sequence Data , Peptide Hydrolases/chemistry , Peptide Hydrolases/pharmacology , Peroxidases/chemistry , Peroxidases/drug effects , Substrate Specificity , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Viral Nonstructural Proteins/chemistry , Virion/drug effects
11.
Liver Int ; 35(7): 1824-32, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25533046

ABSTRACT

BACKGROUND & AIMS: Differences in intrahepatic gene expression patterns may be associated with therapy response in peginterferon-treated chronic hepatitis B (CHB) patients. METHODS: We employed gene expression profiling in baseline liver biopsies of 40 CHB patients (19 HBeAg-positive; 21 HBeAg-negative) treated with peginterferon and adefovir for 48 weeks, and compared expression patterns of combined responders (HBeAg loss, HBV-DNA <2000 IU/ml, alanine aminotransferase normalization after 1 year of treatment-free follow-up) with non-responders. Genes identified by transcriptome analysis in 15 biopsies were confirmed in 25 additional biopsies by RT-qPCR. RESULTS: Transcriptome analysis demonstrated significant differences in expression of 41 genes between responders and non-responders. In responders, pathway analysis showed specific upregulation of genes related to the immune response, including chemotaxis and antigen processing and presentation. Genes upregulated in responders exhibited strongest similarity with a set of genes induced in livers of chimpanzees with acute Hepatitis B infection. Differential expression was confirmed for eight selected genes. A 2-gene subset (HLA-DPB1, SERPIN-E1) was found to predict response most accurately. Incorporation of these genes in a multivariable model with HBeAg status, HBV genotype and baseline HBsAg level correctly classified 90% of all patients, in which HLA-DPB1 and SERPIN-E1 were independent predictors of response. CONCLUSION: We identified an intrahepatic transcriptional signature associated with enhanced immune activation which predicts therapy response. These novel associations could lead to better understanding of responsiveness to peginterferon in CHB patients, and may assist in selecting possible responders to interferon-based treatment.


Subject(s)
Antiviral Agents/therapeutic use , Gene Expression Profiling/methods , Hepatitis B, Chronic/drug therapy , Interferon-alpha/therapeutic use , Liver/drug effects , Oligonucleotide Array Sequence Analysis , Polyethylene Glycols/therapeutic use , Adenine/analogs & derivatives , Adenine/therapeutic use , Adult , Biopsy , Chi-Square Distribution , Drug Therapy, Combination , Female , Genetic Markers , Hepatitis B, Chronic/diagnosis , Hepatitis B, Chronic/genetics , Hepatitis B, Chronic/immunology , Humans , Liver/immunology , Liver/metabolism , Liver/virology , Logistic Models , Male , Middle Aged , Multivariate Analysis , Organophosphonates/therapeutic use , Patient Selection , Precision Medicine , Predictive Value of Tests , Recombinant Proteins/therapeutic use , Reproducibility of Results , Time Factors , Treatment Outcome
12.
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
13.
Hepatology ; 57(4): 1607-19, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23175466

ABSTRACT

UNLABELLED: Hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCC) are the most common liver tumors and a leading cause for cancer-related death in men. Notch2 regulates cellular differentiation in the developing and adult liver. Although aberrant Notch signaling is implicated in various cancers, it is still unclear whether Notch2 regulates proliferation and differentiation in liver carcinogenesis and thereby contributes to HCC and CCC formation. Here, we investigated the oncogenic potential of constitutive Notch2 signaling in the liver. We show that liver-specific expression of the intracellular domain of Notch2 (N2ICD) in mice is sufficient to induce HCC formation and biliary hyperplasia. Specifically, constitutive N2ICD signaling in the liver leads to up-regulation of pro-proliferative genes and proliferation of hepatocytes and biliary epithelial cells (BECs). Using the diethylnitrosamine (DEN) HCC carcinogenesis model, we further show that constitutive Notch2 signaling accelerates DEN-induced HCC formation. DEN-induced HCCs with constitutive Notch2 signaling (DEN(N2ICD) HCCs) exhibit a marked increase in size, proliferation, and expression of pro-proliferative genes when compared with HCCs from DEN-induced control mice (DEN(ctrl) HCCs). Moreover, DEN(N2ICD) HCCs exhibit increased Sox9 messenger RNA (mRNA) levels and reduced Albumin and Alpha-fetoprotein mRNA levels, indicating that they are less differentiated than DEN(ctrl) HCCs. Additionally, DEN(N2ICD) mice develop large hepatic cysts, dysplasia of the biliary epithelium, and eventually CCC. CCC formation in patients and DEN(N2ICD) mice is accompanied by re-expression of hepatocyte nuclear factor 4α(HNF4α), possibly indicating dedifferentiation of BECs. CONCLUSION: Our data establish an oncogenic role for constitutive Notch2 signaling in liver cancer development.


Subject(s)
Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/physiopathology , Diethylnitrosamine/adverse effects , Liver Neoplasms/chemically induced , Liver Neoplasms/physiopathology , Receptor, Notch2/physiology , Signal Transduction/physiology , Animals , Bile Duct Neoplasms/physiopathology , Bile Ducts, Intrahepatic , Carcinoma, Hepatocellular/metabolism , Cell Differentiation , Cell Proliferation , Cholangiocarcinoma/physiopathology , Disease Models, Animal , Female , Hepatocyte Nuclear Factor 4/metabolism , Humans , Liver Neoplasms/metabolism , Male , Mice , Mice, Transgenic , Receptor, Notch2/genetics
14.
Liver Int ; 34(4): 594-603, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23870033

ABSTRACT

BACKGROUND & AIMS: Nodular regenerative hyperplasia (NRH) is a rare liver disease characterized by small regenerative nodules without fibrosis and can cause portal hypertension. Aetiology and pathogenesis of NRH remain unclear. We have recently shown that Notch1 knockout induces NRH with portal hypertension through vascular remodelling in mice. The aim of this study was to analyse histological and clinical data of NRH patients and to explore if the endothelial pathways identified in our NRH mouse model are also regulated in human NRH. METHODS: Patients were identified retrospectively from the pathology database. Clinical and laboratory patient data were retrieved. mRNA expression was measured in liver biopsies from a subset of NRH patients. RESULTS: Diagnosis of NRH was confirmed in needle biopsies of 51 patients, including 31 patients with grade 1, 12 patients with grade 2 and 8 patients with grade 3 NRH. Grade 3 nodularity significantly correlated with the presence of portal hypertension: 50% of the patients with grade 3 NRH vs. 6.5% with grade 1 (P = 0.0105). mRNA expression analysis in liver biopsies from 14 NRH patients and in primary human sinusoidal endothelial cells revealed downregulation of identical genes as in the murine NRH model, which are implicated in vascular differentiation: Notch1, delta-like 4 (Dll4) and ephrinB2. CONCLUSIONS: In this large NRH needle biopsy cohort, we demonstrated that advanced nodularity correlates with presence of portal hypertension. Downregulation of the endothelial signalling pathways Dll4/Notch1 and ephrinB2/EphB4 supports the hypothesis that human NRH is caused by a sinusoidal injury providing first insights into the molecular pathogenesis of this liver condition.


Subject(s)
Down-Regulation/physiology , Ephrin-B2/metabolism , Focal Nodular Hyperplasia/genetics , Hypertension, Portal/etiology , Liver/metabolism , Receptor, Notch1/metabolism , Signal Transduction/physiology , Biopsy , Cohort Studies , Endothelial Cells/metabolism , Focal Nodular Hyperplasia/complications , Focal Nodular Hyperplasia/metabolism , Humans , Liver/pathology , Retrospective Studies
15.
Target Oncol ; 19(2): 213-221, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38416377

ABSTRACT

BACKGROUND: The combination of gemcitabine and cisplatin (gem/cis) with the anti-PD-L1-antibody durvalumab was recently approved as first line therapy for biliary tract cancer (BTC) based on the results of the TOPAZ-1 trial. OBJECTIVE: We aim to analyse the feasibility and efficacy of the triple combination therapy in patients with BTC in a real-world setting and in correspondence with the genetic alterations of the cancer. METHODS: In this single-centre retrospective analysis, all patients with BTC and treated with durvalumab plus gem/cis from April 2022 to September 2023 were included. Survival and treatment response were investigated, within the context of the inclusion and exclusion criteria of TOPAZ-1 and in correspondence with genetic alterations of the cancer. RESULTS: In total, 35 patients, of which 51% met the inclusion criteria of the TOPAZ-1 trial, were analysed. Patients treated within TOPAZ-1 criteria did not have a significantly different median overall survival and progression free survival than the rest of the patients (10.3 versus 9.7 months and 5.3 versus 5 months, respectively). The disease control rate of patients within the TOPAZ-1 criteria was 61.1%, in comparison to 58.8% in the rest of patients. A total of 51 grade 3 and 4 adverse events were observed without significant differences in the subgroups. No specific correlating patterns of genetic alterations with survival and response were observed. CONCLUSIONS: The treatment of advanced patients with BTC with durvalumab and gem/cis, even beyond the inclusion criteria of the TOPAZ-1 trial, shows promising safety.


Subject(s)
Antibodies, Monoclonal , Bile Duct Neoplasms , Biliary Tract Neoplasms , Humans , Gemcitabine , Cisplatin/pharmacology , Cisplatin/therapeutic use , Retrospective Studies , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Deoxycytidine/adverse effects , Biliary Tract Neoplasms/drug therapy , Biliary Tract Neoplasms/etiology
16.
JHEP Rep ; 6(6): 101063, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38737600

ABSTRACT

Background & Aims: Inoperable hepatocellular carcinoma (HCC) can be treated by stereotactic body radiotherapy. However, carbon ion radiotherapy (CIRT) is more effective for sparing non-tumorous liver. High linear energy transfer could promote therapy efficacy. Japanese and Chinese studies on hypofractionated CIRT have yielded excellent results. Because of different radiobiological models and the different etiological spectrum of HCC, applicability of these results to European cohorts and centers remains questionable. The aim of this prospective study was to assess safety and efficacy and to determine the optimal dose of CIRT with active raster scanning based on the local effect model (LEM) I. Methods: CIRT was performed every other day in four fractions with relative biological effectiveness (RBE)-weighted fraction doses of 8.1-10.5 Gy (total doses 32.4-42.0 Gy [RBE]). Dose escalation was performed in five dose levels with at least three patients each. The primary endpoint was acute toxicity after 4 weeks. Results: Twenty patients received CIRT (median age 74.7 years, n = 16 with liver cirrhosis, Child-Pugh scores [CP] A5 [n = 10], A6 [n = 4], B8 [n = 1], and B9 [n = 1]). Median follow up was 23 months. No dose-limiting toxicities and no toxicities exceeding grade II occurred, except one grade III gamma-glutamyltransferase elevation 12 months after CIRT, synchronous to out-of-field hepatic progression. During 12 months after CIRT, no CP elevation occurred. The highest dose level could be applied safely. No local recurrence developed during follow up. The objective response rate was 80%. Median overall survival was 30.8 months (1/2/3 years: 75%/64%/22%). Median progression-free survival was 20.9 months (1/2/3 years: 59%/43%/43%). Intrahepatic progression outside of the CIRT target volume was the most frequent pattern of progression. Conclusions: CIRT of HCC yields excellent local control without dose-limiting toxicity. Impact and implications: To date, safety and efficacy of carbon ion radiotherapy for hepatocellular carcinoma have only been evaluated prospectively in Japanese and Chinese studies. The optimal dose and fractionation when using the local effect model for radiotherapy planning are unknown. The results are of particular interest for European and American particle therapy centers, but also of relevance for all specialists involved in the treatment and care of patients with hepatocellular carcinoma, as we present the first prospective data on carbon ion radiotherapy in hepatocellular carcinoma outside of Asia. The excellent local control should encourage further use of carbon ion radiotherapy for hepatocellular carcinoma and design of randomized controlled trials. Clinical Trials Registration: The study is registered at ClinicalTrials.gov (NCT01167374).

17.
Cell Stem Cell ; 31(4): 554-569.e17, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38579685

ABSTRACT

The YAP/Hippo pathway is an organ growth and size regulation rheostat safeguarding multiple tissue stem cell compartments. LATS kinases phosphorylate and thereby inactivate YAP, thus representing a potential direct drug target for promoting tissue regeneration. Here, we report the identification and characterization of the selective small-molecule LATS kinase inhibitor NIBR-LTSi. NIBR-LTSi activates YAP signaling, shows good oral bioavailability, and expands organoids derived from several mouse and human tissues. In tissue stem cells, NIBR-LTSi promotes proliferation, maintains stemness, and blocks differentiation in vitro and in vivo. NIBR-LTSi accelerates liver regeneration following extended hepatectomy in mice. However, increased proliferation and cell dedifferentiation in multiple organs prevent prolonged systemic LATS inhibition, thus limiting potential therapeutic benefit. Together, we report a selective LATS kinase inhibitor agonizing YAP signaling and promoting tissue regeneration in vitro and in vivo, enabling future research on the regenerative potential of the YAP/Hippo pathway.


Subject(s)
Protein Kinase Inhibitors , Protein Serine-Threonine Kinases , YAP-Signaling Proteins , Animals , Humans , Mice , Cell Proliferation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Stem Cells/metabolism , Transcription Factors/metabolism , YAP-Signaling Proteins/agonists , YAP-Signaling Proteins/drug effects , YAP-Signaling Proteins/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology
18.
J Hepatol ; 58(4): 669-75, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23195617

ABSTRACT

BACKGROUND & AIMS: In the last decade, pegylated interferon-α (PegIFN-α) plus ribavirin (RBV) was the standard treatment of chronic hepatitis C for genotype 1, and it remains the standard for genotypes 2 and 3. Recent studies reported associations between RBV-induced anemia and genetic polymorphisms of concentrative nucleoside transporters such as CNT3 (encoded by SLC28A3) and inosine triphosphatase (encoded by ITPA). We aimed at studying genetic determinants of RBV kinetics, efficacy and treatment-associated anemia. METHODS: We included 216 patients from two Swiss study cohorts (61% HCV genotype 1, 39% genotypes 2 or 3). Patients were analyzed for SLC28A2 single nucleotide polymorphism (SNP) rs11854484, SLC28A3 rs56350726, and SLC28A3 rs10868138 as well as ITPA SNPs rs1127354 and rs7270101, and followed for treatment-associated hemoglobin changes and sustained virological response (SVR). In 67 patients, RBV serum levels were additionally measured during treatment. RESULTS: Patients with SLC28A2 rs11854484 genotype TT had higher dosage- and body weight-adjusted RBV levels than those with genotypes TC or CC (p=0.02 and p=0.06 at weeks 4 and 8, respectively). ITPA SNP rs1127354 was associated with hemoglobin drop ≥3 g/dl during treatment, in genotype (relative risk (RR)=2.1, 95% CI 1.3-3.5) as well as allelic analyses (RR=2.0, 95%CI 1.2-3.4). SLC28A3 rs56350726 was associated with SVR in genotype (RR=2.2; 95% CI 1.1-4.3) as well as allelic analyses (RR=2.0, 95% CI 1.1-3.4). CONCLUSIONS: The newly identified association between RBV serum levels and SLC28A2 rs11854484 genotype, as well as the replicated association of ITPA and SLC28A3 genetic polymorphisms with RBV-induced anemia and treatment response, may support individualized treatment of chronic hepatitis C and warrant further investigation in larger studies.


Subject(s)
Antiviral Agents/blood , Hepatitis C, Chronic/blood , Hepatitis C, Chronic/genetics , Membrane Transport Proteins/genetics , Pyrophosphatases/genetics , Ribavirin/blood , Anemia/chemically induced , Antiviral Agents/adverse effects , Antiviral Agents/therapeutic use , Cohort Studies , Female , Genetic Association Studies , Hemoglobins/metabolism , Hepatitis C, Chronic/drug therapy , Humans , Male , Polymorphism, Single Nucleotide , Ribavirin/adverse effects , Ribavirin/therapeutic use , Treatment Outcome
19.
Gastroenterology ; 142(4): 967-977.e2, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22245843

ABSTRACT

BACKGROUND & AIMS: Notch signaling mediates embryonic vascular development and normal vascular remodeling; Notch1 knockout mice develop nodular regenerative hyperplasia (NRH). The pathogenesis of NRH is unclear, but has been associated with vascular injury in the liver sinusoids in clinical studies. We investigated the role of Notch1 signaling in liver sinusoidal endothelial cells (LSECs). METHODS: We studied MxCre Notch1(lox/lox) mice (conditional knockout mice without tissue-specific disruption of Notch1); mice with hepatocyte-specific knockout were created by crossing Notch1(lox/lox) with AlbCre(+/-) mice. Portal vein pressure was measured; morphology of the hepatic vasculature was assessed by histologic and scanning electron microscopy analyses. We performed functional and expression analyses of isolated liver cells. RESULTS: MxCre-induced knockout of Notch1 led to NRH, in the absence of fibrosis, with a persistent increase in proliferation of LSECs. Notch1 deletion led to de-differentiation, vascular remodeling of the hepatic sinusoidal microvasculature, intussusceptive angiogenesis, and dysregulation of ephrinB2/EphB4 and endothelial tyrosine kinase. Time-course experiments revealed that vascular changes preceded node transformation. MxCre Notch1(lox/lox) mice had reduced endothelial fenestrae and developed portal hypertension and hepatic angiosarcoma over time. In contrast, mice with hepatocyte-specific disruption of Notch1 had a normal phenotype. CONCLUSIONS: Notch1 signaling is required for vascular homeostasis of hepatic sinusoids; it maintains quiescence and differentiation of LSECs in adult mice. Disruption of Notch1 signaling in LSECs leads to spontaneous formation of angiosarcoma, indicating its role as a tumor suppressor in the liver endothelium.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Endothelial Cells/metabolism , Focal Nodular Hyperplasia/metabolism , Hemangiosarcoma/metabolism , Liver Neoplasms/metabolism , Liver/blood supply , Neovascularization, Pathologic/metabolism , Receptor, Notch1/metabolism , Animals , Cell Dedifferentiation , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Endothelial Cells/pathology , Ephrin-B2/metabolism , Focal Nodular Hyperplasia/genetics , Focal Nodular Hyperplasia/pathology , Genotype , Hemangiosarcoma/genetics , Hemangiosarcoma/pathology , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Hypertension, Portal/genetics , Hypertension, Portal/metabolism , Hypertension, Portal/physiopathology , Liver/metabolism , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Scanning , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Phenotype , Portal Pressure , Receptor, EphB4/metabolism , Receptor, Notch1/deficiency , Receptor, Notch1/genetics , Signal Transduction , Time Factors
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL