Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Pharmacol Res Perspect ; 11(3): e01094, 2023 06.
Article in English | MEDLINE | ID: mdl-37204099

ABSTRACT

Ceramide is a central molecule of sphingolipid metabolism and is involved in the development of non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). It has already been reported that the inhibition of serine palmitoyltransferase (SPT), the rate-limiting enzyme in the sphingolipid biosynthetic pathway, has an inhibitory effect on hepatic lipidosis, but its effect on severe hepatic fibrosis is not clear. In this study, we examined whether a SPT inhibitor could suppress the activation of hepatic stellate cells (HSC) and ameliorate the progression of NASH. Effects on sphingolipid metabolism and HSC activation marker genes by NA808, a SPT inhibitor, were evaluated in an immortalized HSC cell line (E14C12). NA808 decreased sphingolipid synthesis and the expression of α-smooth muscle actin (α-SMA) and collagen 1A1 mRNA in HSC. We identified a novel oral SPT inhibitor, CH5169356, which is a prodrug of NA808. CH5169356 was administered in the Ath+HF model, a NASH mouse model with liver fibrosis induced by atherogenic and high-fat content diets. CH5169356 showed a significant decrease in the expression of α-SMA and collagen 1A1 mRNA in the liver and an inhibition of liver fibrosis progression. CH5169356 was also evaluated in a Stelic animal model (STAM), a NASH mouse model induced through a different mechanism than that of the Ath+HF model, and showed a significant anti-fibrotic effect. In conclusion, CH5169356 could inhibit the progression of hepatic fibrosis in the pathogenesis of NASH by suppressing HSC activation, suggesting that CH5169356 would be a potential oral NASH therapeutic agent.


Subject(s)
Non-alcoholic Fatty Liver Disease , Mice , Animals , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Serine C-Palmitoyltransferase/metabolism , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Liver Cirrhosis/etiology , Disease Models, Animal , RNA, Messenger/metabolism
2.
Cell Death Discov ; 7(1): 130, 2021 Jun 02.
Article in English | MEDLINE | ID: mdl-34078875

ABSTRACT

Chronic hepatitis B virus (HBV) infections remain a health burden affecting ~250 million people worldwide. Thus far, available interferon-alpha (IFNα)-based therapies have shown unsatisfactory cure rates, and alternative therapeutic molecules are still required. However, their development has been hampered because accessible cell models supporting relevant HBV replication and appropriate antiviral activity are lacking. Strategies that reverse epigenetic alterations offer a unique opportunity for cell reprogramming, which is valuable for restoring altered cellular functions in human cell lines. This work aimed to investigate the feasibility of converting HepG2 cells that stably overexpress the HBV entry receptor (sodium/taurocholate cotransporting polypeptide, NTCP) toward IFNα-responsive cells using epigenetic reprogramming. Herein, we showed that an epigenetic regimen with non-cytotoxic doses of the demethylating compound 5-azacytidine restored the anti-HBV action of IFNα in epigenetically reprogrammed HepG2-NTCP-C4 cells, named REP-HepG2-NTCP cells. Thus, a significant inhibition in HBV DNA levels was measured in REP-HepG2-NTCP cells after IFNα treatment. This inhibitory effect was associated with the enhancement of IFNα-mediated induction of critical interferon-stimulated genes (ISGs), which was limited in non-reprogrammed cells. In particular, our data indicated that re-expression of 2'-5'-oligoadenylate synthetase 1 (OAS1) and interferon regulatory factor 9 (IRF9) was the result of an epigenetically driven unmasking of these genes in reprogrammed cells. At last, we evaluated the therapeutic potential of the IFN analog CDM-3008 in REP-HepG2-NTCP cells and demonstrated the efficiency of this chemical compound in triggering ISG induction and HBV inhibition. In summary, this study shows that epigenetic reprogramming promotes the IFNα response in HBV-infected cells and is potentially attractive for cell-based experimental screening of IFN-like compounds.

3.
PLoS One ; 14(6): e0216139, 2019.
Article in English | MEDLINE | ID: mdl-31188831

ABSTRACT

Oral administration of nucleotide analogues and injection of interferon-α (IFNα) are used to achieve immediate suppression in replication of hepatitis B virus (HBV). Nucleotide analogs and IFNα inhibit viral polymerase activity and cause long-term eradication of the virus at least in part through removing covalently closed circular DNA (cccDNA) via induction of the APOBEC3 deaminases family of molecules, respectively. This study aimed to explore whether the orally administrable low molecular weight agent CDM-3008 (RO8191), which mimics IFNα through the binding to IFNα/ß receptor 2 (IFNAR2) and the activation of the JAK/STAT pathway, can suppress HBV replication and reduce cccDNA levels. In primary cultured human hepatocytes, HBV DNA levels were decreased after CDM-3008-treatment in a dose-dependent manner with a half-maximal inhibitory concentration (IC50) value of 0.1 µM, and this was accompanied by significant reductions in cellular cccDNA levels, both HBeAg and HBsAg levels in the cell culture medium. Using a microarray we comprehensively analyzed and compared changes in gene (mRNA) expression in CDM-3008- and IFNα-treated primary cultured human hepatocytes. As reported previously, CDM-3008 mimicked the induction of genes that participate in the interferon signaling pathway. OAS1 and ISG20 mRNA expression was similarly enhanced by both CDM-3008 and IFNα. Thus, CDM-3008 could suppress pgRNA expression to show anti-HBV activity. APOBEC3F and 3G mRNA expression was also induced by CDM-3008 and IFNα treatments, suggesting that cccDNA could be degraded through induced APOBEC3 family proteins. We identified the genes whose expression was specifically enhanced in CDM-3008-treated cells compared to IFNα-treated cells. The expression of SOCS1, SOCS2, SOCS3, and CISH, which inhibit STAT activation, was enhanced in CDM-3008-treated cells suggesting that a feedback inhibition of the JAK/STAT pathway was enhanced in CDM-3008-treated cells compared to IFNα-treated cells. In addition, CDM-3008 showed an additive effect with a clinically-used nucleoside entecavir on inhibition of HBV replication. In summary, CDM-3008 showed anti-HBV activity through activation of the JAK/STAT pathway, inducing the expression of interferon-stimulated genes (ISGs), with greater feedback inhibition than IFNα.


Subject(s)
Gene Expression Regulation/drug effects , Hepatitis B virus/drug effects , Interferon-alpha/pharmacology , Naphthyridines/pharmacology , Oxadiazoles/pharmacology , Antiviral Agents/pharmacology , Cells, Cultured , DNA, Viral/drug effects , Hepatitis B virus/genetics , Hepatocytes/cytology , Hepatocytes/virology , Humans , Molecular Mimicry , Protein-Tyrosine Kinases/metabolism , STAT Transcription Factors/metabolism , Virus Replication/drug effects
4.
J Biol Chem ; 294(15): 5759-5773, 2019 04 12.
Article in English | MEDLINE | ID: mdl-30755480

ABSTRACT

Hepatitis C virus (HCV) establishes a chronic infection that can lead to cirrhosis and hepatocellular carcinoma. The HCV life cycle is closely associated with host factors that promote or restrict viral replication, the characterization of which could help to identify potential therapeutic targets. To this end, here we performed a genome-wide microarray analysis and identified ribonucleotide reductase M2 (RRM2) as a cellular factor essential for HCV replication. We found that RRM2 is up-regulated in response to HCV infection in quiescent hepatocytes from humanized chimeric mouse livers. To elucidate the molecular basis of RRM2 expression in HCV-infected cells, we used HCV-infected hepatocytes from chimeric mice and hepatoma cells infected with the HCV strain JFH1. Both models exhibited increased RRM2 mRNA and protein expression levels. Moreover, siRNA-mediated silencing of RRM2 suppressed HCV replication and infection. Of note, RRM2 and RNA polymerase nonstructural protein 5B (NS5B) partially co-localized in cells and co-immunoprecipitated, suggesting that they might interact. RRM2 knockdown reduced NS5B expression, which depended on the protein degradation pathway, as NS5B RNA levels did not decrease and NS5B protein stability correlated with RRM2 protein levels. We also found that RRM2 silencing decreased levels of hPLIC1 (human homolog 1 of protein linking integrin-associated protein and cytoskeleton), a ubiquitin-like protein that interacts with NS5B and promotes its degradation. This finding suggests that there is a dynamic interplay between RRM2 and the NS5B-hPLIC1 complex that has an important function in HCV replication. Together, these results identify a role of host RRM2 in viral RNA replication.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Hepacivirus/physiology , Hepatitis C, Chronic/metabolism , Proteasome Endopeptidase Complex/metabolism , Ribonucleoside Diphosphate Reductase/biosynthesis , Viral Nonstructural Proteins/metabolism , Virus Replication/physiology , Adaptor Proteins, Signal Transducing , Animals , Autophagy-Related Proteins , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Gene Expression Regulation, Enzymologic , Hepatitis C, Chronic/genetics , Hepatitis C, Chronic/pathology , Humans , Liver/metabolism , Liver/pathology , Liver/virology , Mice , Mice, SCID , Mice, Transgenic , Proteasome Endopeptidase Complex/genetics , Protein Stability , Proteolysis , Ribonucleoside Diphosphate Reductase/genetics , Ubiquitination/genetics , Viral Nonstructural Proteins/genetics
5.
Bioorg Med Chem ; 27(3): 470-478, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30552008

ABSTRACT

Hepatitis B, a viral infectious disease caused by hepatitis B virus (HBV), is a life-threatening disease that leads liver cirrhosis and liver cancer. Because the current treatments for HBV, such as an interferon (IFN) formulation or nucleoside/nucleotide analogues, are not sufficient, the development of a more effective agent for HBV is urgent required. CDM-3008 (1, 2-(2,4-bis(trifluoromethyl)imidazo[1,2-a][1,8]naphthyridin-8-yl)-1,3,4-oxadiazole) (RO8191)) is a small molecule with an imidazo[1,2-a][1,8]naphthyridine scaffold that shows anti-HCV activity with an IFN-like effect. Here, we report that 1 was also effective for HBV, although the solubility and metabolic stability were insufficient for clinical use. Through the structure-activity relationship (SAR), we discovered that CDM-3032 (11, N-(piperidine-4-yl)-2,4-bis(trifluoromethyl)imidazo[1,2-a][1,8]naphthyridine-8-carboxamide hydrochloride) was more soluble than 1 (>30 mg/mL for 11 versus 0.92 mg/mL for 1). In addition, the half-life period of 11 was dramatically improved in both mouse and human hepatic microsomes (T1/2, >120 min versus 58.2 min in mouse, and >120 min versus 34.1 min in human, for 11 and 1, respectively).


Subject(s)
Antiviral Agents/pharmacology , Hepatitis B virus/drug effects , Naphthyridines/pharmacology , Oxadiazoles/pharmacology , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Cells, Cultured , Dose-Response Relationship, Drug , Drug Development , Humans , Mice , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Molecular Structure , Naphthyridines/chemical synthesis , Naphthyridines/chemistry , Oxadiazoles/chemical synthesis , Oxadiazoles/chemistry , Structure-Activity Relationship
6.
PLoS One ; 10(11): e0142145, 2015.
Article in English | MEDLINE | ID: mdl-26536627

ABSTRACT

We have used homozygous albumin enhancer/promoter-driven urokinase-type plasminogen activator/severe combined immunodeficient (uPA/SCID) mice as hosts for chimeric mice with humanized livers. However, uPA/SCID mice show four disadvantages: the human hepatocytes (h-heps) replacement index in mouse liver is decreased due to deletion of uPA transgene by homologous recombination, kidney disorders are likely to develop, body size is small, and hemizygotes cannot be used as hosts as more frequent homologous recombination than homozygotes. To solve these disadvantages, we have established a novel host strain that has a transgene containing albumin promoter/enhancer and urokinase-type plasminogen activator cDNA and has a SCID background (cDNA-uPA/SCID). We applied the embryonic stem cell technique to simultaneously generate a number of transgenic lines, and found the line with the most appropriate levels of uPA expression-not detrimental but with a sufficiently damaged liver. We transplanted h-heps into homozygous and hemizygous cDNA-uPA/SCID mice via the spleen, and monitored their human albumin (h-alb) levels and body weight. Blood h-alb levels and body weight gradually increased in the hemizygous cDNA-uPA/SCID mice and were maintained until they were approximately 30 weeks old. By contrast, blood h-alb levels and body weight in uPA/SCID chimeric mice decreased from 16 weeks of age onwards. A similar decrease in body weight was observed in the homozygous cDNA-uPA/SCID genotype, but h-alb levels were maintained until they were approximately 30 weeks old. Microarray analyses revealed identical h-heps gene expression profiles in homozygous and hemizygous cDNA-uPA/SCID mice were identical to that observed in the uPA/SCID mice. Furthermore, like uPA/SCID chimeric mice, homozygous and hemizygous cDNA-uPA/SCID chimeric mice were successfully infected with hepatitis B virus and C virus. These results indicate that hemizygous cDNA-uPA/SCID mice may be novel and useful hosts for producing chimeric mice for use in future long-term studies, including hepatitis virus infection analysis or drug toxicity studies.


Subject(s)
Chimerism , Disease Models, Animal , Hepatitis, Viral, Human , Liver/metabolism , Mice, Inbred Strains/genetics , Animals , Breeding , Child , Child, Preschool , Female , Hemizygote , Hepatitis Viruses/pathogenicity , Hepatocytes/transplantation , Humans , Liver/cytology , Male , Mice, Inbred Strains/virology , Mice, SCID
8.
Sci Rep ; 4: 4750, 2014 Apr 23.
Article in English | MEDLINE | ID: mdl-24756133

ABSTRACT

The development of RNA interference (RNAi)-based therapy faces two major obstacles: selecting small interfering RNA (siRNA) sequences with strong activity, and identifying a carrier that allows efficient delivery to target organs. Additionally, conservative region at nucleotide level must be targeted for RNAi in applying to virus because hepatitis C virus (HCV) could escape from therapeutic pressure with genome mutations. In vitro preparation of Dicer-generated siRNAs targeting a conserved, highly ordered HCV 5' untranslated region are capable of inducing strong RNAi activity. By dissecting the 5'-end of an RNAi-mediated cleavage site in the HCV genome, we identified potent siRNA sequences, which we designate as Dicer-hunting siRNAs (dh-siRNAs). Furthermore, formulation of the dh-siRNAs in an optimized multifunctional envelope-type nano device inhibited ongoing infectious HCV replication in human hepatocytes in vivo. Our efforts using both identification of optimal siRNA sequences and delivery to human hepatocytes suggest therapeutic potential of siRNA for a virus.


Subject(s)
Hepacivirus/genetics , Hepatitis C/metabolism , Hepatitis C/virology , RNA, Small Interfering/genetics , Ribonuclease III/metabolism , 5' Untranslated Regions , Animals , Cell Line , Disease Models, Animal , Gene Silencing , Genome, Viral , Hepatitis C/therapy , Hepatocytes/metabolism , Humans , Hydrogen-Ion Concentration , Male , Mice , Mice, Transgenic , RNA Interference , RNA, Viral , Virus Replication
9.
PLoS One ; 8(12): e82094, 2013.
Article in English | MEDLINE | ID: mdl-24349192

ABSTRACT

BACKGROUND: It has been hypothesized that persistent hepatitis C virus (HCV) infection is mediated in part by viral proteins that abrogate the host immune response, including the complement system, but the precise mechanisms are not well understood. We investigated whether HCV proteins are involved in the fragmentation of complement component 4 (C4), composed of subunits C4α, C4ß, and C4γ, and the role of HCV proteins in complement activation. METHODS: Human C4 was incubated with HCV nonstructural (NS) 3/4A protease, core, or NS5. Samples were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and then subjected to peptide sequencing. The activity of the classical complement pathway was examined using an erythrocyte hemolysis assay. The cleavage pattern of C4 in NS3/4A-expressing and HCV-infected cells, respectively, was also examined. RESULTS: HCV NS3/4A protease cleaved C4γ in a concentration-dependent manner, but viral core and NS5 did not. A specific inhibitor of NS3/4A protease reduced C4γ cleavage. NS3/4A protease-mediated cleavage of C4 inhibited classical pathway activation, which was abrogated by a NS3/4A protease inhibitor. In addition, co-transfection of cells with C4 and wild-type NS3/4A, but not a catalytic-site mutant of NS3/4A, produced cleaved C4γ fragments. Such C4 processing, with a concomitant reduction in levels of full-length C4γ, was also observed in HCV-infected cells expressing C4. CONCLUSIONS: C4 is a novel cellular substrate of the HCV NS3/4A protease. Understanding disturbances in the complement system mediated by NS3/4A protease may provide new insights into the mechanisms underlying persistent HCV infection.


Subject(s)
Carrier Proteins/metabolism , Complement Activation , Complement C4/metabolism , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Complement Activation/drug effects , Complement C4/chemistry , Hemolysis/drug effects , Hepatitis C/virology , Humans , Intracellular Signaling Peptides and Proteins , Molecular Sequence Data , Oligopeptides/pharmacology , Protease Inhibitors/pharmacology , Sheep
10.
Gastroenterology ; 145(4): 865-73, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23791700

ABSTRACT

BACKGROUND & AIMS: Host cell lipid rafts form a scaffold required for replication of hepatitis C virus (HCV). Serine palmitoyltransferases (SPTs) produce sphingolipids, which are essential components of the lipid rafts that associate with HCV nonstructural proteins. Prevention of the de novo synthesis of sphingolipids by an SPT inhibitor disrupts the HCV replication complex and thereby inhibits HCV replication. We investigated the ability of the SPT inhibitor NA808 to prevent HCV replication in cells and mice. METHODS: We tested the ability of NA808 to inhibit SPT's enzymatic activity in FLR3-1 replicon cells. We used a replicon system to select for HCV variants that became resistant to NA808 at concentrations 4- to 6-fold the 50% inhibitory concentration, after 14 rounds of cell passage. We assessed the ability of NA808 or telaprevir to inhibit replication of HCV genotypes 1a, 1b, 2a, 3a, and 4a in mice with humanized livers (transplanted with human hepatocytes). NA808 was injected intravenously, with or without pegylated interferon alfa-2a and HCV polymerase and/or protease inhibitors. RESULTS: NA808 prevented HCV replication via noncompetitive inhibition of SPT; no resistance mutations developed. NA808 prevented replication of all HCV genotypes tested in mice with humanized livers. Intravenous NA808 significantly reduced viral load in the mice and had synergistic effects with pegylated interferon alfa-2a and HCV polymerase and protease inhibitors. CONCLUSIONS: The SPT inhibitor NA808 prevents replication of HCV genotypes 1a, 1b, 2a, 3a, and 4a in cultured hepatocytes and in mice with humanized livers. It might be developed for treatment of HCV infection or used in combination with pegylated interferon alfa-2a or HCV polymerase or protease inhibitors.


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus/drug effects , Hepatocytes/virology , Serine C-Palmitoyltransferase/antagonists & inhibitors , Virus Replication/drug effects , Animals , Hepacivirus/classification , Hepacivirus/genetics , Humans , Mice , RNA, Viral/analysis
11.
Int J Exp Pathol ; 94(2): 93-103, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23305254

ABSTRACT

Non-alcoholic steatohepatitis (NASH) is a progressive fibrotic disease, the pathogenesis of which has not been fully elucidated. One of the most common models used in NASH research is a nutritional model where NASH is induced by feeding a diet deficient in both methionine and choline. However, the dietary methionine-/choline-deficient model in mice can cause severe weight loss and liver atrophy, which are not characteristics of NASH seen in human patients. Exclusive, long-term feeding with a high-fat diet (HFD) produced fatty liver and obesity in mice, but the HFD for several months did not affect fibrosis. We aimed to establish a mouse model of NASH with fibrosis by optimizing the methionine content in the HFD. Male mice were fed a choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD) consisting of 60 kcal% fat and 0.1% methionine by weight. After 1-14 weeks of being fed CDAHFD, the mice were killed. C57BL/6J mice maintained or gained weight when fed CDAHFD, while A/J mice showed a steady decline in body weight (of up to 20% of initial weight). In both strains of mice, plasma levels of alanine aminotransferase increased from week 1, when hepatic steatosis was also observed. By week 6, C57BL/6J mice had developed enlarged fatty liver with fibrosis as assessed by Masson's trichrome staining and by hydroxyproline assay. Therefore, this improved CDAHFD model may be a mouse model of rapidly progressive liver fibrosis and be potentially useful for better understanding human NASH disease and in the development of efficient therapies for this condition.


Subject(s)
Disease Models, Animal , Fatty Liver/pathology , Liver Cirrhosis/pathology , Mice, Inbred C57BL , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Biomarkers/blood , Choline Deficiency , Diet, Fat-Restricted , Diet, High-Fat/adverse effects , Fatty Liver/metabolism , Humans , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Male , Methionine/deficiency , Mice , Mice, Inbred A , Non-alcoholic Fatty Liver Disease , Obesity , Specific Pathogen-Free Organisms , Time Factors
12.
J Med Virol ; 85(2): 241-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23192857

ABSTRACT

The influence of the intracellular redox state on the hepatitis C virus (HCV) life cycle is poorly understood. This study demonstrated the anti-HCV activity of 2,3-dihydro-5-hydroxy-2,2-dipentyl-4,6-di-tert-butylbenzofuran (BO-653), a synthetic lipophilic antioxidant, and examined whether BO-653's antioxidant activity is integral to its anti-HCV activity. The anti-HCV activity of BO-653 was investigated in HuH-7 cells bearing an HCV subgenomic replicon (FLR3-1 cells) and in HuH-7 cells infected persistently with HCV (RMT-tri cells). BO-653 inhibition of HCV replication was also compared with that of several hydrophilic and lipophilic antioxidants. BO-653 suppressed HCV replication in FLR3-1 and RMT-tri cells in a concentration-dependent manner. The lipophilic antioxidants had stronger anti-HCV activities than the hydrophilic antioxidants, and BO-653 displayed the strongest anti-HCV activity of all the antioxidants examined. Therefore, the anti-HCV activity of BO-653 was examined in chimeric mice harboring human hepatocytes infected with HCV. The combination treatment of BO-653 and polyethylene glycol-conjugated interferon-α (PEG-IFN) decreased serum HCV RNA titer more than that seen with PEG-IFN alone. These findings suggest that both the lipophilic property and the antioxidant activity of BO-653 play an important role in the inhibition of HCV replication.


Subject(s)
Antioxidants/pharmacology , Antiviral Agents/pharmacology , Benzofurans/pharmacology , Hepacivirus/drug effects , Virus Replication/drug effects , Animals , Antioxidants/administration & dosage , Antiviral Agents/administration & dosage , Benzofurans/administration & dosage , Cell Line , Disease Models, Animal , Drug Therapy, Combination/methods , Hepacivirus/physiology , Hepatitis C/drug therapy , Hepatocytes/virology , Humans , Interferon-alpha/administration & dosage , Mice , RNA, Viral/blood , Treatment Outcome , Viral Load
13.
Bioorg Med Chem Lett ; 23(1): 336-9, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23164713

ABSTRACT

Hepatitis C virus (HCV) infection represents a serious health-care problem. Previously we reported the identification of NA255 from our natural products library using a HCV sub-genomic replicon cell culture system. Herein, we report how the absolute stereochemistry of NA255 was determined and an enantioselective synthetic method for NA255 derivatives was developed. The structure-activity relationship of the NA255 derivatives and rat pharmacokinetic profiles of the representative compounds are disclosed.


Subject(s)
Antiviral Agents/chemical synthesis , Citrates/chemistry , Hepacivirus/growth & development , Phenylpropionates/chemistry , Animals , Antiviral Agents/pharmacokinetics , Antiviral Agents/toxicity , Cell Line , Cell Survival/drug effects , Citrates/pharmacokinetics , Citrates/toxicity , Half-Life , Hepacivirus/drug effects , Humans , Phenylpropionates/pharmacokinetics , Phenylpropionates/toxicity , Rats , Stereoisomerism , Structure-Activity Relationship , Virus Replication/drug effects
14.
J Hepatol ; 57(6): 1186-92, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22889955

ABSTRACT

BACKGROUND & AIMS: Early menopause in women with chronic hepatitis C virus (HCV) infection is associated with a low likelihood of a sustained virological response (SVR) in conjunction with their antiviral treatment. This is potentially related to their reduced estrogen secretion. The study was done to determine whether selective estrogen receptor modulator administration might improve the efficacy of the current standard of care (SOC) treatment, pegylated interferon (PegIFN) α2a plus ribavirin (RBV), for postmenopausal women. METHODS: One hundred and twenty-three postmenopausal women with genotype 1b chronic hepatitis C were randomly assigned to one of two treatment groups: raloxifene hydrochloride (RLX) (60 mg/day) plus SOC (PegIFNα2a 180 µg/week and RBV 600-1,000 mg/day) (n=62) or SOC only (n=61). Genotyping was performed of the polymorphism in the interleukin-28B (IL28B) gene region (rs8099917) of DNA collected from each patient. RESULTS: One RLX-treated patient discontinued RLX because of a systemic rash following 2 weeks of treatment. Twenty-four weeks after treatment, the SVR rate was significantly higher for RLX plus SOC patients (61.3%) than for SOC only patients (34.4%) (p=0.0051). Further, the SVR rate was significantly higher for RLX plus SOC patients with IL28B TT (72.5%) than for SOC only patients with IL28B TT (39.2%) (p=0.0014), but no such relationship was observed in patients carrying the minor IL28B allele. CONCLUSIONS: RLX improved the efficacy of SOC in the treatment of postmenopausal women with chronic hepatitis C. RLX shows promise as an adjuvant to the standard antiviral treatment of such patients.


Subject(s)
Adjuvants, Pharmaceutic/therapeutic use , Antiviral Agents/therapeutic use , Hepatitis C, Chronic/drug therapy , Postmenopause , Raloxifene Hydrochloride/therapeutic use , Selective Estrogen Receptor Modulators/therapeutic use , Aged , Female , Genotype , Hepatitis C, Chronic/virology , Humans , Interferon-alpha/therapeutic use , Interferons , Interleukins/genetics , Middle Aged , Polyethylene Glycols/therapeutic use , Prospective Studies , RNA, Viral/analysis , Recombinant Proteins/therapeutic use , Ribavirin/therapeutic use
15.
PLoS Pathog ; 8(8): e1002860, 2012.
Article in English | MEDLINE | ID: mdl-22916015

ABSTRACT

Lipids are key components in the viral life cycle that affect host-pathogen interactions. In this study, we investigated the effect of HCV infection on sphingolipid metabolism, especially on endogenous SM levels, and the relationship between HCV replication and endogenous SM molecular species. We demonstrated that HCV induces the expression of the genes (SGMS1 and 2) encoding human SM synthases 1 and 2. We observed associated increases of both total and individual sphingolipid molecular species, as assessed in human hepatocytes and in the detergent-resistant membrane (DRM) fraction in which HCV replicates. SGMS1 expression had a correlation with HCV replication. Inhibition of sphingolipid biosynthesis with a hepatotropic serine palmitoyltransferase (SPT) inhibitor, NA808, suppressed HCV-RNA production while also interfering with sphingolipid metabolism. Further, we identified the SM molecular species that comprise the DRM fraction and demonstrated that these endogenous SM species interacted with HCV nonstructural 5B polymerase to enhance viral replication. Our results reveal that HCV alters sphingolipid metabolism to promote viral replication, providing new insights into the formation of the HCV replication complex and the involvement of host lipids in the HCV life cycle.


Subject(s)
Hepacivirus/physiology , Hepatitis C/metabolism , Sphingolipids/biosynthesis , Virus Replication/physiology , Animals , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Hepatitis C/genetics , Humans , Membrane Proteins/biosynthesis , Mice , Nerve Tissue Proteins/biosynthesis , Serine C-Palmitoyltransferase/antagonists & inhibitors , Serine C-Palmitoyltransferase/genetics , Serine C-Palmitoyltransferase/metabolism , Sphingolipids/genetics , Transferases (Other Substituted Phosphate Groups)/biosynthesis , Virus Replication/drug effects
16.
Sci Rep ; 2: 259, 2012.
Article in English | MEDLINE | ID: mdl-22355771

ABSTRACT

Most acute hepatitis C virus (HCV) infections become chronic and some progress to liver cirrhosis or hepatocellular carcinoma. Standard therapy involves an interferon (IFN)-α-based regimen, and efficacy of therapy has been significantly improved by the development of protease inhibitors. However, several issues remain concerning the injectable form and the side effects of IFN. Here, we report an orally available, small-molecule type I IFN receptor agonist that directly transduces the IFN signal cascade and stimulates antiviral gene expression. Like type I IFN, the small-molecule compound induces IFN-stimulated gene (ISG) expression for antiviral activity in vitro and in vivo in mice, and the ISG induction mechanism is attributed to a direct interaction between the compound and IFN-α receptor 2, a key molecule of IFN-signaling on the cell surface. Our study highlights the importance of an orally active IFN-like agent, both as a therapy for antiviral infections and as a potential IFN substitute.


Subject(s)
Hepacivirus/drug effects , Interferon Type I/pharmacology , Virus Replication/drug effects , Administration, Oral , Animals , Blotting, Western , Hepacivirus/physiology , Interferon Type I/administration & dosage , Mice , Phosphorylation , Real-Time Polymerase Chain Reaction , Signal Transduction , Surface Plasmon Resonance
17.
J Infect Dis ; 204(8): 1172-80, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21917889

ABSTRACT

BACKGROUND: We recently established a monoclonal antibody (2-152a MAb) that binds to 3ß-hydroxysterol-Δ24-reductase (DHCR24) by immunizing mice with cells (RzM6-LC) persistently expressing hepatitis C virus (HCV). Here, we aimed to analyze the activity of 2-152a MAb against HCV replication and explore the molecular mechanism underlying the antiviral activity. METHODS: We characterized the effects of 2-152a MAb on HCV replication and performed a microarray analysis of antibody-treated HCV replicon cells. The molecules showing a significant change after the antibody treatment were screened to examine their relationship with HCV replication. RESULTS: The antibody had antiviral activity both in vitro and in vivo (chimeric mice). In the microarray analysis, 2-152a MAb significantly suppressed the expression of betaine/GABA transporter-1 (BGT-1) in 2 HCV replicon cell lines but not in HCV-cured cells. Silencing of BGT-1 expression by small interfering RNA (siRNA) revealed significant suppression of HCV replication and infection without cytotoxicity. Further, BGT-1 expression was significantly increased in the presence of HCV (P < .05). CONCLUSIONS: Our results suggest that 2-152a MAb suppresses HCV replication and infection through BGT-1. These findings highlight important roles of BGT-1 in HCV replication and reveal a possible target for anti-HCV therapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Carrier Proteins/metabolism , GABA Plasma Membrane Transport Proteins/metabolism , Hepacivirus/physiology , Hepatitis C, Chronic/drug therapy , Virus Replication/drug effects , Animals , Antibodies, Monoclonal/immunology , Cell Line, Tumor , GABA Plasma Membrane Transport Proteins/genetics , Hep G2 Cells , Hepatitis C, Chronic/metabolism , Hepatitis C, Chronic/virology , Humans , Mice , Mice, Inbred BALB C , Mice, Transgenic , Nerve Tissue Proteins/immunology , Oligonucleotide Array Sequence Analysis , Oxidoreductases Acting on CH-CH Group Donors/immunology , RNA/chemistry , RNA/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
18.
J Med Virol ; 83(4): 587-93, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21328371

ABSTRACT

Serine palmitoyltransferase (SPT) catalyzes the first step in the sphingolipid biosynthetic pathway. Myriocin inhibits SPT and was shown to suppress the replication of hepatitis C virus (HCV) in vitro and in vivo. However, its effect on hepatitis B virus (HBV) replication is unknown. In this study, the HBV DNA levels in HuH7 cell culture supernatants were lowered successfully by using myriocin and it was found that the 50% inhibitory concentration of myriocin is approximately 5 µM. Myriocin and/or pegylated interferon (PEG-IFN) were also administered to chimeric mice for 2 weeks and the effects of these compounds on HBV DNA levels were determined. Myriocin alone did not reduce effectively the HBV DNA levels, whereas PEG-IFN alone reduced the DNA levels to 1/10th of the control levels. The combination of myriocin with PEG-IFN reduced the HBV levels to about 1/1,000 th of the control levels and induced a 1.0 log reduction in the levels of the HBV surface antigen and core protein. This latter effect was not observed in the other treatment groups. In conclusion, the combination of myriocin with PEG-IFN represses synergistically HBV replication in vivo without inducing hepatotoxicity.


Subject(s)
Antiviral Agents/pharmacology , Fatty Acids, Monounsaturated/pharmacology , Hepatitis B virus/physiology , Sphingolipids/antagonists & inhibitors , Virus Replication , Animals , Antiviral Agents/therapeutic use , Cell Line , DNA, Viral/analysis , Disease Models, Animal , Drug Therapy, Combination , Fatty Acids, Monounsaturated/therapeutic use , Hepatocytes/drug effects , Hepatocytes/virology , Humans , Inhibitory Concentration 50 , Interferons/therapeutic use , Mice , Treatment Outcome
19.
Arch Virol ; 156(2): 295-304, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21246385

ABSTRACT

Hepatitis C virus (HCV) replicon systems enable in-depth analysis of the life cycle of HCV. However, the previously reported full-genome replicon system is unable to produce authentic virions. On the basis of these results, we constructed newly designed full-genomic replicon RNA, which is composed of the intact 5'-terminal-half RNA extending to the NS2 region flanked by an extra selection marker gene. Huh-7 cells harboring this full-genomic RNA proliferated well under G418 selection and secreted virion-like particles into the supernatant. These particles, which were round and 50 nm in diameter when analyzed by electron microscopy, had a buoyant density of 1.08 g/mL that shifted to 1.19 g/mL after NP-40 treatment; these figures match the putative densities of intact virions and nucleocapsids without envelope. The particles also showed infectivity in a colony-forming assay. This system may offer another option for investigating the life cycle of HCV.


Subject(s)
Hepacivirus/genetics , Hepacivirus/physiology , RNA, Viral/genetics , Replicon , Cell Line , Genome, Viral , Hepacivirus/ultrastructure , Humans , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Open Reading Frames , Virion/genetics , Virion/physiology , Virion/ultrastructure , Virus Replication/genetics
20.
J Hepatol ; 55(3): 512-521, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21184787

ABSTRACT

BACKGROUND & AIMS: We characterized the role of 24-dehydrocholesterol reductase (DHCR24) in hepatitis C virus infection (HCV). DHCR24 is a cholesterol biosynthetic enzyme and cholesterol is a major component of lipid rafts, which is reported to play an important role in HCV replication. Therefore, we examined the potential of DHCR24 as a target for novel HCV therapeutic agents. METHODS: We examined DHCR24 expression in human hepatocytes in both the livers of HCV-infected patients and those of chimeric mice with human hepatocytes. We targeted DHCR24 with siRNA and U18666A which is an inhibitor of both DHCR24 and cholesterol synthesis. We measured the level of HCV replication in these HCV replicon cell lines and HCV infected cells. U18666A was administrated into chimeric mice with humanized liver, and anti-viral effects were assessed. RESULTS: Expression of DHCR24 was induced by HCV infection in human hepatocytes in vitro, and in human hepatocytes of chimeric mouse liver. Silencing of DHCR24 by siRNA decreased HCV replication in replicon cell lines and HCV JFH-1 strain-infected cells. Treatment with U18666A suppressed HCV replication in the replicon cell lines. Moreover, to evaluate the anti-viral effect of U18666A in vivo, we administrated U18666A with or without pegylated interferon to chimeric mice and observed an inhibitory effect of U18666A on HCV infection and a synergistic effect with interferon. CONCLUSIONS: DHCR24 is an essential host factor which augmented its expression by HCV infection, and plays a significant role in HCV replication. DHCR24 may serve as a novel anti-HCV drug target.


Subject(s)
Hepacivirus/physiology , Hepatitis C/virology , Nerve Tissue Proteins/metabolism , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Virus Replication/physiology , Androstenes/pharmacology , Androstenes/therapeutic use , Animals , Cholesterol/metabolism , Hep G2 Cells , Hepatitis C/drug therapy , Hepatitis C/metabolism , Humans , Interferon-alpha/therapeutic use , Mice , Mice, Inbred BALB C , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/physiology , Oxidoreductases Acting on CH-CH Group Donors/drug effects , Oxidoreductases Acting on CH-CH Group Donors/physiology , Polyethylene Glycols/therapeutic use , RNA, Small Interfering/pharmacology , Recombinant Proteins/therapeutic use , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...