Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters











Publication year range
1.
Microbiol Spectr ; 9(1): e0075521, 2021 09 03.
Article in English | MEDLINE | ID: mdl-34431717

ABSTRACT

Chronic hepatitis C virus (HCV) infection induces liver inflammation that can lead to fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). Inflammation is the outcome of the action of proinflammatory cytokines and chemokines, including interleukin-1 beta (IL-1ß) and tumor necrosis factor alpha. Mature IL-1ß production and secretion are facilitated by active inflammasome complexes, including the NACHT-LRR pyrin domain-containing protein 3 (NLRP3) inflammasome. Our study shows that the NLRP3 inflammasome is activated in HCV-infected hepatocytes and that the activation is regulated by posttranslational modifications. NLRP3 is modified by lysine-63 ubiquitin chains in hepatocytes and is deubiquitinated during HCV infection. Inhibition of deubiquitinases (DUBs) with chemical inhibitors or blocking UCHL5 DUB expression with small interfering RNA (siRNA) abrogated NLRP3 inflammasome assembly and activation. Inhibition of inflammasome deubiquitination was correlated with a reduction in IL-1ß maturation, decrease in HCV protein expression, and reduction in release of HCV from the cells. Together, this study suggests that HCV-induced activation of the NLRP3 inflammasome through posttranslational modification is crucial for the HCV life cycle and pathogenesis. IMPORTANCE HCV infection induces inflammation leading to fibrosis, cirrhosis, and cancer. The current study identifies the mechanisms leading to the activation of the NLRP3 inflammasome in hepatocytes, which is an important site of viral replication. Deubiquitination of NLRP3 by UCHL5 is required for inflammasome activation. Inhibition of deubiquitination blocks NLRP3 inflammasome activation and IL-1ß maturation and also decreases HCV replication, suggesting the importance of the NLRP3 inflammasome in inflammation as well as other signaling pathways.


Subject(s)
Hepacivirus/physiology , Hepatitis C, Chronic/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Ubiquitin Thiolesterase/metabolism , Amino Acid Motifs , Cell Line, Tumor , Cytokines/genetics , Cytokines/metabolism , Hepacivirus/genetics , Hepatitis C, Chronic/genetics , Hepatitis C, Chronic/virology , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Inflammasomes/genetics , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/chemistry , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Signal Transduction , Ubiquitin/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitination
6.
J Inorg Biochem ; 206: 111023, 2020 05.
Article in English | MEDLINE | ID: mdl-32163811

ABSTRACT

Molecular gold(I) and platinum(II) species were examined for the inhibition of liver fibrosis and the hepatitis C virus (HCV). Determination of inhibition efficiency was conducted via morphological analysis, cell viability, western blot analysis, and quantitative reverse transcription polymerase chain reaction (RT-PCR). Auranofin and Ph3PAuCl demonstrated the greatest inhibition of liver fibrosis amongst the tested gold species in human hepatic stellate LX-2 cells. Western blot analysis indicated that auranofin and Ph3PAuCl prevent signal transducer and activator of transcription 3 (STAT3) phosphorylation, which may be a key connection to fibrosis and inflammation. Auranofin and Ph3PAuCl also reduced expression of HCV-nonstructural protein 3 (NS3) and HCV-NS5a proteins in a HCV subgenomic replicon system. These results demonstrate significant promise for the use of gold compounds in treating liver diseases such as HCV.


Subject(s)
Liver Cirrhosis/pathology , Organogold Compounds/pharmacology , Organoplatinum Compounds/pharmacology , Platinum Compounds/pharmacology , Auranofin/pharmacology , Cell Line , Cell Survival , Gold/chemistry , Hepacivirus/metabolism , Hepatitis C/drug therapy , Hepatitis C/metabolism , Hepatitis C/pathology , Humans , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Organogold Compounds/chemistry , Organoplatinum Compounds/chemistry , Phosphorylation , Platinum/chemistry , Platinum Compounds/chemistry , STAT3 Transcription Factor/metabolism
10.
J Virol ; 92(13)2018 07 01.
Article in English | MEDLINE | ID: mdl-29669827

ABSTRACT

Hepatitis C virus (HCV) replication and assembly occur at the specialized site of endoplasmic reticulum (ER) membranes and lipid droplets (LDs), respectively. Recently, several host proteins have been shown to be involved in HCV replication and assembly. In the present study, we demonstrated the important relationship among osteopontin (OPN), the ER, and LDs. OPN is a secreted phosphoprotein, and overexpression of OPN in hepatocellular carcinoma (HCC) tissue can lead to invasion and metastasis. OPN expression is also enhanced in HCV-associated HCC. Our recent studies have demonstrated the induction, proteolytic cleavage, and secretion of OPN in response to HCV infection. We also defined the critical role of secreted OPN in human hepatoma cell migration and invasion through binding to receptors integrin αVß3 and CD44. However, the role of HCV-induced OPN in the HCV life cycle has not been elucidated. In this study, we showed a significant reduction in HCV replication, assembly, and infectivity in HCV-infected cells transfected with small interfering RNA (siRNA) against OPN, αVß3, and CD44. We also observed the association of endogenous OPN with HCV proteins (NS3, NS5A, NS4A/B, NS5B, and core). Confocal microscopy revealed the colocalization of OPN with HCV NS5A and core in the ER and LDs, indicating a possible role for OPN in HCV replication and assembly. Interestingly, the secreted OPN activated HCV replication, infectivity, and assembly through binding to αVß3 and CD44. Collectively, these observations provide evidence that HCV-induced OPN is critical for HCV replication and assembly.IMPORTANCE Recently, our studies uncovered the critical role of HCV-induced endogenous and secreted OPN in migration and invasion of hepatocytes. However, the role of OPN in the HCV life cycle has not been elucidated. In this study, we investigated the importance of OPN in HCV replication and assembly. We demonstrated that endogenous OPN associates with HCV NS3, NS5A, NS5B, and core proteins, which are in close proximity to the ER and LDs. Moreover, we showed that the interactions of secreted OPN with cell surface receptors αVß3 and CD44 are critical for HCV replication and assembly. These observations provide evidence that HCV-induced endogenous and secreted OPN play pivotal roles in HCV replication and assembly in HCV-infected cells. Taken together, our findings clearly demonstrate that targeting OPN may provide opportunities for therapeutic intervention of HCV pathogenesis.


Subject(s)
Hepatitis C/virology , Hyaluronan Receptors/metabolism , Integrin alphaVbeta3/metabolism , Lipid Droplets/metabolism , Osteopontin/metabolism , Viral Nonstructural Proteins/metabolism , Virus Assembly , Virus Replication , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , DNA Replication , Hepacivirus/physiology , Hepatitis C/metabolism , Humans , Hyaluronan Receptors/genetics , Integrin alphaVbeta3/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/virology , Osteopontin/genetics , RNA, Viral , Tumor Cells, Cultured , Viral Nonstructural Proteins/genetics
11.
J Biol Chem ; 292(52): 21676-21689, 2017 12 29.
Article in English | MEDLINE | ID: mdl-29079574

ABSTRACT

Hepatitis C virus (HCV) infection is a major risk factor for the development of chronic liver disease. The disease typically progresses from chronic HCV to fibrosis, cirrhosis, hepatocellular carcinoma (HCC), and death. Chronic inflammation associated with HCV infection is implicated in cirrhosis and HCC, but the molecular players and signaling pathways contributing to these processes remain largely unknown. Interferon regulatory factor 5 (IRF5) is a molecule of interest in HCV-associated HCC because it has critical roles in virus-, Toll-like receptor (TLR)-, and IFN-induced signaling pathways. IRF5 is also a tumor suppressor, and its expression is dysregulated in several human cancers. Here, we present first evidence that IRF5 expression and signaling are modulated during HCV infection. Using HCV infection of human hepatocytes and cells with autonomously replicating HCV RNA, we found that levels of IRF5 mRNA and protein expression were down-regulated. Of note, reporter assays indicated that IRF5 re-expression inhibited HCV protein translation and RNA replication. Gene expression analysis revealed significant differences in the expression of cancer pathway mediators and autophagy proteins rather than in cytokines between IRF5- and empty vector-transfected HCV replicon cells. IRF5 re-expression induced apoptosis via loss in mitochondrial membrane potential, down-regulated autophagy, and inhibited hepatocyte cell migration/invasion. Analysis of clinical HCC specimens supports a pathologic role for IRF5 in HCV-induced HCC, as IRF5 expression was down-regulated in livers from HCV-positive versus HCV-negative HCC patients or healthy donor livers. These results identify IRF5 as an important suppressor of HCV replication and HCC pathogenesis.


Subject(s)
Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Hepacivirus/genetics , Hepacivirus/metabolism , Hepatitis C/pathology , Hepatitis C, Chronic/pathology , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver/pathology , Liver Cirrhosis/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Signal Transduction , Virus Replication/genetics , Virus Replication/physiology
12.
Bioorg Med Chem Lett ; 27(5): 1129-1135, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28190633

ABSTRACT

Liver fibrosis is a critical wound healing response to chronic liver injury such as hepatitis C virus (HCV) infection. If persistent, liver fibrosis can lead to cirrhosis and hepatocellular carcinoma (HCC). The development of new therapies for preventing liver fibrosis and its progression to cancer associated with HCV infection remains a critical challenge. Identification of novel anti-fibrotic compounds will provide opportunities for innovative therapeutic intervention of HCV-mediated liver fibrosis. We designed and synthesized a focused set of 5-arylthio-5H-chromenopyridines as a new class of anti-fibrotic agents. Liver fibrosis assays demonstrated that the compounds 3a and 3c show inhibitory activity towards human hepatic stellate cells (LX2) activation at 10µM. The HCV NS3 and NS5A proteins in HCV subgenome-expressing cells were also significantly reduced in cells treated with 3a and 3c, suggesting the possible inhibitory role of the compounds in HCV translation/replication activities. We have also examined the reactivity of these compounds with medicinally-relevant metal compounds such as platinum and gold. The reactivity of these complexes with metals and during Mass Spectrometry suggests that CS bond cleavage is relatively facile.


Subject(s)
Hepatitis C/complications , Liver Cirrhosis/prevention & control , Pyrimidines/pharmacology , Hepacivirus/metabolism , Humans , Liver Cirrhosis/etiology , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
13.
Cancer Res ; 76(16): 4887-96, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27287718

ABSTRACT

Hepatocellular carcinoma (HCC) is the third most common cause of cancer-related deaths worldwide. Chronic hepatitis C virus (HCV) infection causes induction of several tumors/cancer stem cell (CSC) markers and is known to be a major risk factor for development of HCC. Therefore, drugs that simultaneously target viral replication and CSC properties are needed for a risk-free treatment of advanced stage liver diseases, including HCC. Here, we demonstrated that (Z)-3,5,4'-trimethoxystilbene (Z-TMS) exhibits potent antitumor and anti-HCV activities without exhibiting cytotoxicity to human hepatocytes in vitro or in mice livers. Diethylnitrosamine (DEN)/carbon tetrachloride (CCl4) extensively induced expression of DCLK1 (a CSC marker) in the livers of C57BL/6 mice following hepatic injury. Z-TMS exhibited hepatoprotective effects against DEN/CCl4-induced injury by reducing DCLK1 expression and improving histologic outcomes. The drug caused bundling of DCLK1 with microtubules and blocked cell-cycle progression at G2-M phase in hepatoma cells via downregulation of CDK1, induction of p21(cip1/waf1) expression, and inhibition of Akt (Ser(473)) phosphorylation. Z-TMS also inhibited proliferation of erlotinib-resistant lung adenocarcinoma cells (H1975) bearing the T790M EGFR mutation, most likely by promoting autophagy and nuclear fragmentation. In conclusion, Z-TMS appears to be a unique therapeutic agent targeting HCV and concurrently eliminating cells with neoplastic potential during chronic liver diseases, including HCC. It may also be a valuable drug for targeting drug-resistant carcinomas and cancers of the lungs, pancreas, colon, and intestine, in which DCLK1 is involved in tumorigenesis. Cancer Res; 76(16); 4887-96. ©2016 AACR.


Subject(s)
Antiviral Agents/pharmacology , Hepatitis C, Chronic/pathology , Microtubules/drug effects , Neoplastic Stem Cells/drug effects , Stilbenes/pharmacology , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Flow Cytometry , Hepatitis C, Chronic/complications , Humans , Liver Neoplasms/pathology , Liver Neoplasms/virology , Mice , Mice, Inbred C57BL , Xenograft Model Antitumor Assays
14.
J Biol Chem ; 291(7): 3254-67, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26698881

ABSTRACT

Hepatitis C virus (HCV) relies on host lipids and lipid droplets for replication and morphogenesis. The accumulation of lipid droplets in infected hepatocytes manifests as hepatosteatosis, a common pathology observed in chronic hepatitis C patients. One way by which HCV promotes the accumulation of intracellular lipids is through enhancing de novo lipogenesis by activating the sterol regulatory element-binding proteins (SREBPs). In general, activation of SREBPs occurs during cholesterol depletion. Interestingly, during HCV infection, the activation of SREBPs occurs under normal cholesterol levels, but the underlying mechanisms are still elusive. Our previous study has demonstrated the activation of the inflammasome complex in HCV-infected human hepatoma cells. In this study, we elucidate the potential link between chronic hepatitis C-associated inflammation and alteration of lipid homeostasis in infected cells. Our results reveal that the HCV-activated NLRP3 inflammasome is required for the up-regulation of lipogenic genes such as 3-hydroxy-3-methylglutaryl-coenzyme A synthase, fatty acid synthase, and stearoyl-CoA desaturase. Using pharmacological inhibitors and siRNA against the inflammasome components (NLRP3, apoptosis-associated speck-like protein containing a CARD, and caspase-1), we further show that the activation of the NLRP3 inflammasome plays a critical role in lipid droplet formation. NLRP3 inflammasome activation in HCV-infected cells enables caspase-1-mediated degradation of insulin-induced gene proteins. This subsequently leads to the transport of the SREBP cleavage-activating protein·SREBP complex from the endoplasmic reticulum to the Golgi, followed by proteolytic activation of SREBPs by S1P and S2P in the Golgi. Typically, inflammasome activation leads to viral clearance. Paradoxically, here we demonstrate how HCV exploits the NLRP3 inflammasome to activate SREBPs and host lipid metabolism, leading to liver disease pathogenesis associated with chronic HCV.


Subject(s)
Carrier Proteins/metabolism , Hepacivirus/physiology , Hepatocytes/virology , Inflammasomes/metabolism , Lipogenesis , Sterol Regulatory Element Binding Protein 1/agonists , Sterol Regulatory Element Binding Protein 2/agonists , CARD Signaling Adaptor Proteins , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Caspase 1/chemistry , Caspase 1/genetics , Caspase 1/metabolism , Cell Line, Tumor , Cysteine Proteinase Inhibitors/pharmacology , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Endopeptidases/chemistry , Endopeptidases/metabolism , Enzyme Induction/drug effects , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Golgi Apparatus/pathology , Golgi Apparatus/virology , Hepacivirus/drug effects , Hepatitis C, Chronic/metabolism , Hepatitis C, Chronic/pathology , Hepatitis C, Chronic/physiopathology , Hepatitis C, Chronic/virology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Host-Pathogen Interactions/drug effects , Humans , Inflammasomes/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Lipogenesis/drug effects , Membrane Proteins/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Non-alcoholic Fatty Liver Disease/etiology , Proprotein Convertases/chemistry , Proprotein Convertases/metabolism , Protein Transport/drug effects , Proteolysis/drug effects , RNA Interference , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism
15.
World J Gastroenterol ; 21(43): 12361-9, 2015 Nov 21.
Article in English | MEDLINE | ID: mdl-26604643

ABSTRACT

AIM: To investigate the mechanisms of insulin resistance in human hepatoma cells expressing hepatitis C virus (HCV) nonstructural protein 5A (NS5A). METHODS: The human hepatoma cell lines, Huh7 and Huh7.5, were infected with HCV or transiently-transfected with a vector expressing HCV NS5A. The effect of HCV NS5A on the status of the critical players involved in insulin signaling was analyzed using real-time quantitative polymerase chain reaction and Western blot assays. Data were analyzed using Graph Pad Prism version 5.0. RESULTS: To investigate the effect of insulin treatment on the players involved in insulin signaling pathway, we analyzed the status of insulin receptor substrate-1 (IRS-1) phosphorylation in HCV infected cells or Huh7.5 cells transfected with an HCV NS5A expression vector. Our results indicated that there was an increased phosphorylation of IRS-1 (Ser(307)) in HCV infected or NS5A transfected Huh7.5 cells compared to their respective controls. Furthermore, an increased phosphorylation of Akt (Ser(473)) was observed in HCV infected and NS5A transfected cells compared to their mock infected cells. In contrast, we observed decreased phosphorylation of Akt Thr308 phosphorylation in HCV NS5A transfected cells. These results suggest that Huh7.5 cells either infected with HCV or ectopically expressing HCV NS5A alone have the potential to induce insulin resistance by the phosphorylation of IRS-1 at serine residue (Ser(307)) followed by decreased phosphorylation of Akt Thr(308), Fox01 Ser(256) and GSK3ß Ser(9), the downstream players of the insulin signaling pathway. Furthermore, increased expression of PECK and glucose-6-phosphatase, the molecules involved in gluconeogenesis, in HCV NS5A transfected cells was observed. CONCLUSION: Taken together, our results suggest the role of HCV NS5A in the induction of insulin resistance by modulating various cellular targets involved in the insulin signaling pathway.


Subject(s)
Carcinoma, Hepatocellular/virology , Gluconeogenesis , Hepacivirus/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance , Liver Neoplasms/virology , Viral Nonstructural Proteins/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hepacivirus/genetics , Host-Pathogen Interactions , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Serine , Signal Transduction , Transfection , Viral Nonstructural Proteins/genetics
16.
PLoS One ; 9(1): e87464, 2014.
Article in English | MEDLINE | ID: mdl-24498111

ABSTRACT

Osteopontin (OPN) is a secreted phosphoprotein which has been linked to tumor progression and metastasis in a variety of cancers including hepatocellular carcinoma (HCC). Previous studies have shown that OPN is upregulated during liver injury and inflammation. However, the role of OPN in hepatitis C virus (HCV)-induced liver disease pathogenesis is not known. In this study, we determined the induction of OPN, and then investigated the effect of secreted forms of OPN in epithelial to mesenchymal transition (EMT), migration and invasion of hepatocytes. We show the induction of OPN mRNA and protein expression by HCV-infection. Our results also demonstrate the processing of precursor OPN (75 kDa) into 55 kDa, 42 kDa and 36 kDa forms of OPN in HCV-infected cells. Furthermore, we show the binding of secreted OPN to integrin αVß3 and CD44 at the cell surface, leading to the activation of downstream cellular kinases such as focal adhesion kinase (FAK), Src, and Akt. Importantly, our results show the reduced expression of epithelial marker (E-cadherin) and induction of mesenchymal marker (N-cadherin) in HCV-infected cells. We also show the migration and invasion of HCV-infected cells using wound healing assay and matrigel coated Boyden chamber. In addition, we demonstrate the activation of above EMT markers, and the critical players involved in OPN-mediated cell signaling cascade using primary human hepatocytes infected with Japanese fulminant hepatitis (JFH)-1 HCV. Taken together, these studies suggest a potential role of OPN in inducing chronic liver disease and HCC associated with chronic HCV infection.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Hepacivirus/metabolism , Hepatitis C, Chronic/metabolism , Hepatocytes/metabolism , Liver Neoplasms/metabolism , Neoplasm Proteins/biosynthesis , Osteopontin/biosynthesis , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Hep G2 Cells , Hepatitis C, Chronic/pathology , Hepatocytes/pathology , Hepatocytes/virology , Humans , Hyaluronan Receptors , Liver Neoplasms/pathology , Liver Neoplasms/virology , Signal Transduction
17.
Arch Virol ; 159(5): 1017-25, 2014 May.
Article in English | MEDLINE | ID: mdl-24240483

ABSTRACT

Chronic hepatitis C is a lethal blood-borne infection often associated with a number of pathologies such as insulin resistance and other metabolic abnormalities. Insulin is a key hormone that regulates the expression of metabolic pathways and favors homeostasis. In this study, we demonstrated the molecular mechanism of hepatitis C virus (HCV) nonstructural protein 5A (NS5A)-induced metabolic dysregulation. We showed that transient expression of HCV NS5A in human hepatoma cells increased lipid droplet formation through enhanced lipogenesis. We also showed increased transcriptional expression of peroxisome proliferator-activated receptor gamma coactivator (PGC)-1α and diacylglycerol acyltransferase-1 (DGAT-1) in NS5A-expressing cells. On the other hand, there was significantly reduced transcriptional expression of microsomal triglyceride transfer protein (MTP) and peroxisome proliferator-activated receptor γ (PPARγ) in cells expressing HCV NS5A. Furthermore, increased gluconeogenic gene expression was observed in HCV-NS5A-expressing cells. In addition, it was also shown that HCV-NS5A-expressing hepatoma cells show serine phosphorylation of IRS-1, thereby hampering metabolic activity and contributing to insulin resistance. Therefore, this study reveals that HCV NS5A is involved in enhanced gluconeogenic and lipogenic gene expression, which triggers metabolic abnormality and impairs insulin signaling pathway.


Subject(s)
Gluconeogenesis/physiology , Hepacivirus/metabolism , Lipogenesis/physiology , Viral Nonstructural Proteins/physiology , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Gene Expression Regulation , Humans , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance , Liver Neoplasms/metabolism , Signal Transduction
18.
J Biol Chem ; 288(52): 36994-7009, 2013 Dec 27.
Article in English | MEDLINE | ID: mdl-24240095

ABSTRACT

Osteopontin (OPN) is a secreted phosphoprotein, originally characterized in malignant-transformed epithelial cells. OPN is associated with tumor metastasis of several tumors and is overexpressed in hepatocellular carcinoma (HCC) tissue involving HCC invasion and metastasis. Importantly, OPN is significantly up-regulated in liver injury, inflammation, and hepatitis C virus (HCV)-associated HCC. However, the underlying mechanisms of OPN activation and its role in HCV-mediated liver disease pathogenesis are not known. In this study, we investigated the mechanism of OPN activation in HCV-infected cells. We demonstrate that HCV-mediated Ca(2+) signaling, elevation of reactive oxygen species, and activation of cellular kinases such as p38 MAPK, JNK, PI3K, and MEK1/2 are involved in OPN activation. Incubation of HCV-infected cells with the inhibitors of AP-1 and Sp1 and site-directed mutagenesis of AP-1- and Sp1-binding sites on the OPN promoter suggest the critical role of AP-1 and Sp1 in OPN promoter activation. In addition, we show the in vivo interactions of AP-1 and Sp1 with the OPN promoter using chromatin immunoprecipitation assay. We also show the calpain-mediated processing of precursor OPN (∼75 kDa) into ∼55-, ∼42-, and ∼36-kDa forms of OPN in HCV-infected cells. Furthermore, we demonstrate the critical role of HCV-induced OPN in increased phosphorylation of Akt and GSK-3ß followed by the activation of ß-catenin, which can lead to EMT of hepatocytes. Taken together, these studies provide an insight into the mechanisms of OPN activation that is relevant to the metastasis of HCV-associated HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Epithelial-Mesenchymal Transition , Hepatitis C/metabolism , Hepatocytes/metabolism , Liver Neoplasms/metabolism , Osteopontin/metabolism , Calcium Signaling/genetics , Calpain/genetics , Calpain/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hepacivirus , Hepatitis C/genetics , Hepatitis C/pathology , Hepatocytes/pathology , Hepatocytes/virology , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/virology , Neoplasm Metastasis , Osteopontin/genetics , Protein Processing, Post-Translational/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Transcription Factor AP-1 , beta Catenin/genetics , beta Catenin/metabolism
19.
PLoS One ; 8(2): e56367, 2013.
Article in English | MEDLINE | ID: mdl-23437118

ABSTRACT

Our previous studies have shown the induction and maturation of transforming growth factor-beta 1 (TGF-ß1) in HCV-infected human hepatoma cells. In this study, we have investigated the molecular mechanism of TGF-ß1 gene expression in response to HCV infection. We demonstrate that HCV-induced transcription factors AP-1, Sp1, NF-κB and STAT-3 are involved in TGF-ß1 gene expression. Using chromatin immunoprecipitation (ChIP) assay, we further show that AP-1 and Sp1 interact with TGF-b1 promoter in vivo in HCV-infected cells. In addition, we demonstrate that HCV-induced TGF-ß1 gene expression is mediated by the activation of cellular kinases such as p38 MAPK, Src, JNK, and MEK1/2. Next, we determined the role of secreted bioactive TGF-ß1 in human hepatic stellate cells (HSCs) activation and invasion. Using siRNA approach, we show that HCV-induced bioactive TGF-ß1 is critical for the induction of alpha smooth muscle actin (α-SMA) and type 1 collagen, the markers of HSCs activation and proliferation. We further demonstrate the potential role of HCV-induced bioactive TGF-ß1 in HSCs invasion/cell migration using a transwell Boyden chamber. Our results also suggest the role of HCV-induced TGF-ß1 in HCV replication and release. Collectively, these observations provide insight into the mechanism of TGF-ß1 promoter activation, as well as HSCs activation and invasion, which likely manifests in liver fibrosis associated with HCV infection.


Subject(s)
Hepacivirus/physiology , Hepatic Stellate Cells/pathology , Hepatic Stellate Cells/virology , Promoter Regions, Genetic/genetics , Sp1 Transcription Factor/metabolism , Transcription Factor AP-1/metabolism , Transforming Growth Factor beta1/genetics , Animals , Base Sequence , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Line, Tumor , DNA Mutational Analysis , Furin/metabolism , Gene Expression Regulation , Genes, Reporter , Hepatic Stellate Cells/metabolism , Humans , Luciferases/metabolism , Mink , Molecular Sequence Data , Signal Transduction/genetics , Transcriptional Activation/genetics , Transforming Growth Factor beta1/metabolism , Virus Replication
20.
J Gen Virol ; 93(Pt 2): 235-246, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21994322

ABSTRACT

Interleukin-1ß (IL-1ß) is a potent pro-inflammatory cytokine involved in the pathogenesis of HCV, but the sensors and underlying mechanisms that facilitate HCV-induced IL-1ß proteolytic activation and secretion remains unclear. In this study, we have identified a signalling pathway leading to IL-1ß activation and secretion in response to HCV infection. Previous studies have shown the induction and secretion of IL-1ß through the inflammasome complex in macrophages/monocytes. Here, we report for the first time the induction and assembly of the NALP3-inflammasome complex in human hepatoma cells infected with HCV (JFH-1). We demonstrate that activation of IL-1ß in HCV-infected cells involves the proteolytic processing of pro-caspase-1 into mature caspase-1 in a multiprotein inflammasome complex. Next, we demonstrate that HCV is sensed by NALP3 protein, which recruits the adaptor protein ASC for the assembly of the inflammasome complex. Using a small interfering RNA approach, we further show that components of the inflammasome complex are involved in the activation of IL-1ß in HCV-infected cells. Our study also demonstrates the role of reactive oxygen species in HCV-induced IL-1ß secretion. Collectively, these observations provide an insight into the mechanism of IL-1ß processing and secretion, which is likely to provide novel strategies for targeting the viral or cellular determinants to arrest the progression of liver disease associated with chronic HCV infection.


Subject(s)
Caspase 1/metabolism , Hepacivirus/immunology , Inflammasomes/metabolism , Interleukin-1beta/metabolism , CARD Signaling Adaptor Proteins , Carrier Proteins/metabolism , Cell Line, Tumor , Cytoskeletal Proteins/metabolism , Hepatocytes/immunology , Hepatocytes/virology , Humans , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Multimerization , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL