Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
2.
Acta Pharm Sin B ; 9(4): 769-781, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31384537

ABSTRACT

Bicyclol is a synthetic drug for hepatoprotection in clinic since 2004. Preliminary clinical observations suggest that bicyclol might be active against hepatitis C virus (HCV) with unknown mechanism. Here, we showed that bicyclol significantly inhibited HCV replication in vitro and in hepatitis C patients. Using bicyclol as a probe, we identified glycolipid transfer protein (GLTP) to be a novel restrictive factor for HCV replication. The GLTP preferentially bound host vesicle-associated membrane protein-associated protein-A (VAP-A) in competition with the HCV NS5A, causing an interruption of the complex formation between VAP-A and HCV NS5A. As the formation of VAP-A/NS5A complex is essential for viral RNA replication, up-regulation of GLTP by bicyclol reduced the level of VAP-A/NS5A complex and thus inhibited HCV replication. Bicyclol also exhibited an inhibition on HCV variants resistant to direct-acting antiviral agents (DAAs) with an efficacy identical to that on wild type HCV. In combination with bicyclol, DAAs inhibited HCV replication in a synergistic fashion. GLTP appears to be a newly discovered host restrictive factor for HCV replication, Up-regulation of GLTP causes spontaneous restriction of HCV replication.

3.
World J Gastroenterol ; 24(47): 5297-5311, 2018 Dec 21.
Article in English | MEDLINE | ID: mdl-30598575

ABSTRACT

Hepatitis C virus (HCV) infection commonly causes progressive liver diseases that deteriorate from chronic inflammation to fibrosis, cirrhosis and even to hepatocellular carcinoma. A long-term, persistent and uncontrolled inflammatory response is a hallmark of these diseases and further leads to hepatic injury and more severe disease progression. The levels of inflammatory cytokines and chemokines change with the states of infection and treatment, and therefore, they may serve as candidate biomarkers for disease progression and therapeutic effects. The mechanisms of HCV-induced inflammation involve classic pathogen pattern recognition, inflammasome activation, intrahepatic inflammatory cascade response, and oxidative and endoplasmic reticulum stress. Direct-acting antivirals (DAAs) are the first-choice therapy for effectively eliminating HCV, but DAAs alone are not sufficient to block the uncontrolled inflammation and severe liver injury in HCV-infected individuals. Some patients who achieve a sustained virologic response after DAA therapy are still at a long-term risk for progression to liver cirrhosis and hepatocellular carcinoma. Therefore, coupling with anti-inflammatory/hepatoprotective agents with anti-HCV effects is a promising therapeutic regimen for these patients during or after treatment with DAAs. In this review, we discuss the relationship between inflammatory mediators and HCV infection, summarize the mechanisms of HCV-induced inflammation, and describe the potential roles of anti-inflammatory/hepatoprotective drugs with anti-HCV activity in the treatment of advanced HCV infection.


Subject(s)
Hepacivirus/drug effects , Hepacivirus/immunology , Hepatitis C/drug therapy , Hepatitis C/immunology , Anti-Inflammatory Agents/therapeutic use , Antiviral Agents/therapeutic use , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/prevention & control , Carcinoma, Hepatocellular/virology , Chemokines/immunology , Cytokines/immunology , Disease Progression , Drug Therapy, Combination/methods , Hepatitis C/pathology , Hepatitis C/virology , Humans , Liver/drug effects , Liver/immunology , Liver/pathology , Liver/virology , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Liver Cirrhosis/prevention & control , Liver Cirrhosis/virology , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Liver Neoplasms/prevention & control , Liver Neoplasms/virology , Protective Agents/therapeutic use , Severity of Illness Index , Sustained Virologic Response
4.
Front Pharmacol ; 9: 1438, 2018.
Article in English | MEDLINE | ID: mdl-30618739

ABSTRACT

Treatment with direct-acting antivirals (DAAs) cures most patients infected with hepatitis C virus (HCV) in the real world. However, some patients, especially those with the underlying advanced liver disease, have a limited reduction of liver injury after achieving a sustained viral response (SVR). Bicyclol was widely used in clinics for the treatment of a variety of liver injuries but with an unknown mechanism for the treatment of hepatitis C. We investigated the anti-inflammatory effects and mechanisms of bicyclol in HCV-infected hepatocytes and further confirmed the putative results in a mouse hepatitis model induced by the coinjection of polyinosinic-polycytidylic acid [poly (I:C)] and D-galactosamine (D-GalN). The results showed that the activation of nuclear factor kappa B (NF-κB) and the subsequent increase of inflammatory factors were directly induced by HCV infection and were persistent after clearance of the virus in Huh7.5 cells. Bicyclol decreased the activation of NF-κB and the levels of inflammatory factors in HCV-infected hepatocytes by inhibiting the activation of the ROS-MAPK-NF-κB pathway, and the effect was synergistic with DAAs in HCV-infected hepatocytes. Bicyclol attenuated the ROS-MAPK-NF-κB axis via recovering mitochondrial function without a dependence on dihydronicotinamide adenine dinucleotide phosphate oxidase and superoxide dismutases. The anti-inflammatory effects and mechanism of bicyclol were verified in mouse hepatitis induced by the coinjection of poly(I:C)/D-GalN. Bicyclol directly ameliorates the chronic inflammation caused by HCV infection and might be used with DAAs or after DAA therapy for ultimately curing chronic hepatitis C.

5.
Int J Mol Med ; 40(6): 1792-1802, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29039494

ABSTRACT

The hepatitis C virus (HCV) infection is associated with various extrahepatic manifestations, which are correlated with poor outcomes, and thus increase the morbidity and mortality of chronic hepatitis C (CHC). Therefore, understanding the internal linkages between systemic manifestations and HCV infection is helpful for treatment of CHC. Yet, the mechanism by which the virus evokes the systemic diseases remains to be elucidated. In the present study, using gene set enrichment analysis (GSEA) and signaling pathway impact analysis (SPIA), a comprehensive analysis of microarray data of mRNAs was conducted in HCV-infected and -uninfected Huh7.5 cells, and signaling pathways (which are significantly activated or inhibited) and certain molecules (which are commonly important in those signaling pathways) were selected. Forty signaling pathways were selected using GSEA, and eight signaling pathways were selected with SPIA. These pathways are associated with cancer, metabolism, environmental information processing and organismal systems, which provide important information for further clarifying the intrinsic associations between syndromes of HCV infection, of which seven pathways were not previously reported, including basal transcription factors, pathogenic Escherichia coli infection, shigellosis, gastric acid secretion, dorso-ventral axis formation, amoebiasis and cholinergic synapse. Ten genes, SOS1, RAF1, IFNA2, IFNG, MTHFR, IGF1, CALM3, UBE2B, TP53 and BMP7 whose expression may be the key internal driving molecules, were selected using the online tool Anni 2.1. Furthermore, the present study demonstrated the internal linkages between systemic manifestations and HCV infection, and presented the potential molecules that are key to those linkages.


Subject(s)
Hepacivirus/physiology , Hepacivirus/pathogenicity , Hepatitis C/metabolism , Hepatitis C/physiopathology , Host-Pathogen Interactions/physiology , Signal Transduction/physiology , Algorithms , Cell Line , Computational Biology , Escherichia coli Infections , Gene Expression Regulation , Humans , Oligonucleotide Array Sequence Analysis , Transcription Factors
6.
Biomed Res Int ; 2017: 1236801, 2017.
Article in English | MEDLINE | ID: mdl-28904942

ABSTRACT

Use of direct-acting antivirals sometimes causes viral drug resistance, resulting in inefficiency in treated patients in real-world practice. Therefore, how to rapidly and accurately evaluate drug resistance is an urgent problem to be solved for rational use and development of antivirals in the future. Here, we aim to develop a new method by which we can evaluate easily but effectively whether a drug will still be efficient in the future treatment in infectious hepatitis C virus cell culture system. HCV-infected Huh7.5 cells were treated with drugs and the culture supernatants were replaced with fresh culture media containing the same drugs at 24 hours. The supernatants were harvested at 48 hours and incubated with naïve Huh7.5 cells. Intracellular HCV RNAs or proteins in the newly infected cells were extracted and analyzed at 48 hours or longer. Results showed that after being treated with telaprevir mutant viruses were easily detected which were resistant to telaprevir, while after being treated with sofosbuvir drug-resistant viruses did not emerge. In conclusion, the new method is simple and quick but accurate to evaluate whether a drug will be still efficient in the forthcoming therapeutic regimen and whether drug resistance will occur after long-term treatment with drugs.


Subject(s)
Drug Resistance, Viral/genetics , Hepacivirus/drug effects , Hepatitis C/drug therapy , Antiviral Agents/pharmacology , Cell Line , Drug Resistance, Viral/drug effects , Genotype , Hepacivirus/pathogenicity , Hepatitis C/virology , Humans , Oligopeptides/pharmacology , Viral Nonstructural Proteins/genetics
7.
J Med Chem ; 59(22): 10268-10284, 2016 11 23.
Article in English | MEDLINE | ID: mdl-27783522

ABSTRACT

There still remains a need to develop new anti-HCV agents with distinct mechanism of action (MOA) due to the occurrence of resistance to direct-acting antiviral agents (DAAs). Cajanine, a stilbenic component isolated from Cajanus cajan L., was identified as a potent HCV inhibitor by phenotypic screening in this work (EC50 = 3.17 ± 0.75 µM). The intensive structure optimization provided significant insights into the structure-activity relationships. Furthermore, the MOA study revealed that cajanine inhibited HCV replications via down-regulating a cellular protein chondroitin sulfate N-acetylgalactosaminyltransferase 1. In consistency with this host-targeting mechanism, cajanine showed the similar magnitude of inhibitory activity against both drug-resistant and wild-type HCV and synergistically inhibited HCV replication with approved DAAs. Taken together, our study not only presented cajanine derivatives as a novel class of anti-HCV agents but also discovered a promising anti-HCV target to combat drug resistance.


Subject(s)
Antiviral Agents/pharmacology , Diethylstilbestrol/analogs & derivatives , Down-Regulation/drug effects , Drug Design , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Hepacivirus/drug effects , N-Acetylgalactosaminyltransferases/biosynthesis , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Diethylstilbestrol/chemical synthesis , Diethylstilbestrol/chemistry , Diethylstilbestrol/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Microbial Sensitivity Tests , Molecular Structure , N-Acetylgalactosaminyltransferases/metabolism , Structure-Activity Relationship
8.
Sci Rep ; 6: 21808, 2016 Feb 22.
Article in English | MEDLINE | ID: mdl-26898231

ABSTRACT

The cluster of differentiation 36 (CD36) is a membrane protein related to lipid metabolism. We show that HCV infection in vitro increased CD36 expression in either surface or soluble form. HCV attachment was facilitated through a direct interaction between CD36 and HCV E1 protein, causing enhanced entry and replication. The HCV co-receptor effect of CD36 was independent of that of SR-BI. CD36 monoclonal antibodies neutralized the effect of CD36 and reduced HCV replication. CD36 inhibitor sulfo-N-succinimidyl oleate (SSO), which directly bound CD36 but not SR-BI, significantly interrupted HCV entry, and therefore inhibited HCV replication. SSO's antiviral effect was seen only in HCV but not in other viruses. SSO in combination with known anti-HCV drugs showed additional inhibition against HCV. SSO was considerably safe in mice. Conclusively, CD36 interacts with HCV E1 and might be a co-receptor specific for HCV entry; thus, CD36 could be a potential drug target against HCV.


Subject(s)
Antiviral Agents/pharmacology , CD36 Antigens/genetics , Hepacivirus/drug effects , Oleic Acids/pharmacology , Receptors, Virus/genetics , Succinimides/pharmacology , Virus Attachment/drug effects , Virus Internalization/drug effects , Animals , Antibodies, Neutralizing/pharmacology , CD36 Antigens/antagonists & inhibitors , CD36 Antigens/metabolism , Cell Line, Tumor , Drug Synergism , Gene Expression Regulation , HEK293 Cells , Hepacivirus/genetics , Hepacivirus/metabolism , Hepatocytes/drug effects , Hepatocytes/pathology , Hepatocytes/virology , Host-Pathogen Interactions , Humans , Mice , Oligopeptides/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Virus/antagonists & inhibitors , Receptors, Virus/metabolism , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism , Signal Transduction , Toxicity Tests, Acute , Transgenes , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virus Replication
9.
Yao Xue Xue Bao ; 51(6): 913-8, 2016 06.
Article in Chinese | MEDLINE | ID: mdl-29879344

ABSTRACT

The level of intracellular keratin 8(KRT-8) is associated with liver diseases, whose expression is increased in hepatitis C virus(HCV)-infected patients with hepatocarcinoma and in cultural cells infected with HCV. However, it is not clear whether KRT-8 will impact HCV replication. In this paper, the HCV replication was analyzed in response to high expression and silence of KRT-8. The inhibitory activities against wild-type and mutant HCV were also analyzed by silence of KRT-8 or combined with known anti-HCV drug telaprevir. Results showed that the protein level of KRT-8 was increased in proportion with the HCV replication. The high expression was found to facilitate HCV replication, while the silence of KRT-8 was able to inhibit HCV replication and enhanced the anti-HCV activity of telaprevir. It also inhibited A156 T and D168 V mutant HCV, which are resistant to protease inhibitors. These results suggest that KRT-8 is a co-factor for HCV replication. Down-regulation of KRT-8 can inhibit wild type and mutant HCV replication to enhance the anti-HCV activity of known anti-HCV drugs. Therefore, KRT-8 may be a new target in the development of anti-HCV agents.


Subject(s)
Hepacivirus/physiology , Keratin-8/metabolism , Virus Replication , Antiviral Agents/pharmacology , Carcinoma, Hepatocellular/virology , Cells, Cultured , DNA Replication , Humans , Oligopeptides/pharmacology , Protease Inhibitors/pharmacology
10.
PLoS One ; 10(3): e0121608, 2015.
Article in English | MEDLINE | ID: mdl-25811715

ABSTRACT

Human APOBEC3G (hA3G) is a cytidine deaminase that restricts replication of certain viruses. We have previously reported that hA3G was a host restriction factor against hepatitis C virus (HCV) replication, and hA3G stabilizers showed a significant inhibitory activity against HCV. However, the molecular mechanism of hA3G against HCV remains unknown. We show in this study that hA3G's C-terminal directly binds HCV non-structural protein NS3 at its C-terminus, which is responsible for NS3's helicase and NTPase activity. Binding of hA3G to the C-terminus of NS3 reduced helicase activity, and therefore inhibited HCV replication. The anti-HCV mechanism of hA3G appeared to be independent of its deamination activity. Although early stage HCV infection resulted in an increase in host hA3G as an intracellular response against HCV replication, hA3G was gradually diminished after a long-term incubation, suggesting an unknown mechanism(s) that protects HCV NS3 from inactivation by hA3G. The process represents, at least partially, a cellular defensive mechanism against HCV and the action is mediated through a direct interaction between host hA3G and HCV NS3. We believe that understanding of the antiviral mechanism of hA3G against HCV might open an interesting avenue to explore hA3G stabilizers as a new class of anti-HCV agents.


Subject(s)
Cytidine Deaminase/metabolism , Hepacivirus/physiology , Hepatitis C/metabolism , Hepatitis C/virology , Host-Pathogen Interactions , Viral Nonstructural Proteins/metabolism , Virus Replication , APOBEC-3G Deaminase , Cell Line , Hepatitis C/pathology , Humans , Intracellular Space/metabolism , Models, Molecular , Protein Binding , Protein Interaction Maps
SELECTION OF CITATIONS
SEARCH DETAIL
...