Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
J Gen Virol ; 94(Pt 11): 2424-2428, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23994832

ABSTRACT

Apoptosis has been shown to be induced and downregulated by the Hantaan virus (HTNV) nucleocapsid (N) protein. To address these conflicting data, expression of the p53 protein, one of the key molecules involved in apoptosis, was assessed in the presence of the N protein in A549 and HeLa cells. The amount of p53, increased by drug treatment, was reduced when cells were infected with HTNV or transfected with an expression vector of the HTNV N protein. When cells were treated with a proteasome inhibitor (MG132) or an MDM2 antagonist (Nutlin-3), p53 expression was not reduced in N protein-overexpressed cells. We concluded that the HTNV N protein ubiquitinates and degrades p53 MDM2-dependently. Here we report downregulation of p53 expression through a post-translational mechanism: MDM2-dependent ubiquitination and degradation by the HTNV N protein. These results indicate that N protein-dependent p53 degradation through the ubiquitin proteasome system is one of the anti-apoptotic mechanisms employed by HTNV.


Subject(s)
Capsid Proteins/metabolism , Gene Expression Regulation , Hantaan virus/pathogenicity , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Viral Core Proteins/metabolism , Apoptosis , Cell Line , Down-Regulation , Genes, p53 , Hantaan virus/metabolism , HeLa Cells , Humans , Proteasome Endopeptidase Complex , Proto-Oncogene Proteins c-mdm2/genetics , Tumor Suppressor Protein p53/genetics , Ubiquitin/metabolism , Ubiquitination
2.
J Biol Chem ; 286(26): 23093-101, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21536672

ABSTRACT

We previously demonstrated that the receptor for the complement component C1q (gC1qR) is a lipid raft protein that is indispensable for adipogenesis and insulin signaling. Here, we provide the first report that gC1qR is an essential component of lamellipodia in human lung carcinoma A549 cells. Cell-surface gC1qR was concentrated in the lamellipodia along with CD44, monosialoganglioside, actin, and phosphorylated focal adhesion kinase in cells stimulated with insulin, IGF-1, EGF, or serum. The growth factor-induced lamellipodia formation and cell migration were significantly decreased in gC1qR-depleted cells, with a concomitant blunt activation of the focal adhesion kinase and the respective receptor tyrosine kinases. Moreover, the gC1qR-depleted cells exhibited a reduced proliferation rate in culture as well as diminished tumorigenic and metastatic activities in grafted mice. We therefore conclude that cell-surface gC1qR regulates lamellipodia formation and metastasis via receptor tyrosine kinase activation.


Subject(s)
Adenocarcinoma/metabolism , Carrier Proteins/metabolism , Lung Neoplasms/metabolism , Mitochondrial Proteins/metabolism , Neoplasm Proteins/metabolism , Pseudopodia/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Carrier Proteins/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Enzyme Activation/genetics , Female , Focal Adhesion Protein-Tyrosine Kinases/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondrial Proteins/genetics , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasm Transplantation , Pseudopodia/genetics , Pseudopodia/pathology , Transplantation, Heterologous
3.
Biochem Biophys Res Commun ; 407(1): 248-53, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21385567

ABSTRACT

The replication of viral nucleic acids triggers cellular antiviral responses. The double-stranded RNA (dsRNA)-activated protein kinase (PKR) plays a key role in this antiviral response. We have recently reported that JFH-1 HCV replication in Huh-7 cells triggers PKR activation. Here we show that the HCV-induced PKR activation is further stimulated by the mitogen- and stress-activated protein kinase 2 (MSK2), a member of the 90kDa ribosomal S6 kinase (RSK) family that has emerged as an important downstream effector of ERK and p38 MAPK signaling pathways. We show that MSK2 binds PKR and stimulates PKR phosphorylation, whereas the closely related MSK1 and RSK2 have no effect. Our data further indicate that MSK2 functions as an adaptor in mediating PKR activation, apparently independent of its catalytic activity. These results suggest that, in addition to viral dsRNA, stress signaling contributes to the regulation of cellular antiviral response.


Subject(s)
Hepacivirus , Hepatitis C/enzymology , Hepatitis C/virology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , eIF-2 Kinase/biosynthesis , Cell Line, Tumor , Enzyme Activation , Humans , Ribosomal Protein S6 Kinases, 90-kDa/genetics
4.
PLoS One ; 15(8): e0236704, 2020.
Article in English | MEDLINE | ID: mdl-32790777

ABSTRACT

The hepatitis B virus (HBV) envelope is composed of a lipid bilayer and three glycoproteins, referred to as the large (L), middle (M), and small (S) hepatitis B virus surface antigens (HBsAg). S protein constitutes the major portion of the viral envelope and an even greater proportion of subviral particles (SVP) that circulate in the blood. Recombinant S proteins are currently used as a preventive vaccine, while plasma fractions isolated from vaccinated people, referred to as hepatitis B immune globulin (HBIG), are used for short-term prophylaxis. Here, we characterized a recombinant human IgG1 type anti-S antibody named Lenvervimab regarding its binding property to a variety of cloned S antigens. Immunochemical data showed an overall consistent avidity of the antibody to S antigens of most viral genotypes distributed worldwide. Further, antibody binding was not affected by the mutations in the antigenic 'a' determinant found in many clinical variants, including the immune escape mutant G145R. In addition, mutations in the S gene sequence that confer drug resistance to the viral polymerase did not interfere with the antibody binding. These results support for a preventive use of the antibody against HBV infection.


Subject(s)
Hepatitis B Antibodies/immunology , Hepatitis B Surface Antigens/genetics , Hepatitis B virus/metabolism , Immunoglobulins/immunology , Amino Acid Sequence , Antigen-Antibody Reactions , Cell Line , Drug Resistance, Viral , Genotype , Hep G2 Cells , Hepatitis B/pathology , Hepatitis B/virology , Hepatitis B Antibodies/metabolism , Hepatitis B Surface Antigens/chemistry , Hepatitis B Surface Antigens/immunology , Hepatitis B virus/genetics , Hepatitis B virus/isolation & purification , Humans , Immunoglobulins/genetics , Immunoglobulins/metabolism , Polymorphism, Single Nucleotide , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification
5.
Proteomics ; 9(9): 2373-82, 2009 May.
Article in English | MEDLINE | ID: mdl-19402044

ABSTRACT

Since insulin receptors and their downstream signaling molecules are organized in lipid rafts, proteomic analysis of adipocyte lipid rafts may provide new insights into the function of lipid rafts in adipogenesis and insulin signaling. To search for proteins involved in adipocyte differentiation and insulin signaling, we analyzed detergent-resistant lipid raft proteins from 3T3-L1 preadipocytes and adipocytes by 2-DE. Eleven raft proteins were identified from adipocytes. One of the adipocyte-specific proteins was globular C1q receptor (gC1qR), an acidic 32 kDa protein known as the receptor for the globular domain of complement C1q. The targeting of gC1qR into lipid rafts was significantly increased during adipogenesis, as determined by immunoblotting and immunofluorescence. Since the silencing of gC1qR by small RNA interference abolished adipogenesis and blocked insulin-induced activation of insulin receptor, insulin receptor substrate-1 (IRS-1), Akt, and Erk1/2, we can conclude that gC1qR is an essential molecule involved in adipogenesis and insulin signaling.


Subject(s)
Adipocytes/chemistry , Adipogenesis , Hyaluronan Receptors/metabolism , Insulin/metabolism , Membrane Microdomains/chemistry , Proteome/analysis , Adipocytes/metabolism , Amino Acid Sequence , Animals , Cell Line , Down-Regulation , Electrophoresis, Gel, Two-Dimensional , Gene Expression Regulation , Hyaluronan Receptors/analysis , Membrane Microdomains/metabolism , Mice , Mitochondrial Proteins , Molecular Sequence Data , RNA Interference , Signal Transduction
6.
J Hepatol ; 50(3): 479-88, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19155084

ABSTRACT

BACKGROUND/AIMS: Hepatitis C virus (HCV) is one of the major human hepatic RNA viruses. Recently, we developed a liver-specific siRNA delivery technology using DTC-Apo composed of cationic liposomes (DTC) and apolipoprotein A-I (apo A-I). Here, we investigated whether DTC-Apo nanoparticles can systemically deliver siRNA into mouse hepatocytes expressing HCV proteins and inhibit their expression efficiently. METHODS: A transient HCV model was constructed by hydrodynamic injection of plasmid DNA expressing viral structural proteins under hepatic control region and alpha1-antitrypsin promoter elements. Using this model, DTC-Apo containing HCV-core-specific siRNA was intravenously injected to assess antiviral activity as well as the duration of silencing. RESULTS: Post-administration of DTC-Apo/HCV-specific siRNA at a dose of 2mg siRNA/kg inhibited viral gene expression by 65-75% in the liver on day 2. Improved activity (95% knockdown on day 2) without immunotoxicity was obtained by 2'-OMe-modification at two U sequences on its sense strand. Notably, the gene silencing effect of the modified siRNA was still maintained at day 6, while the unmodified one lost RNAi activity after day 4. CONCLUSIONS: Our results suggest that DTC-Apo liposome is a highly potential delivery vehicle to transfer therapeutic siRNA especially targeting HCV to the liver.


Subject(s)
Apolipoprotein A-I/pharmacology , Hepacivirus/genetics , Liposomes/pharmacology , RNA, Small Interfering/pharmacology , Animals , Cell Line , Drug Delivery Systems , Drug Stability , Female , Hepacivirus/drug effects , Hepatitis C/drug therapy , Humans , Mice , Mice, Inbred C57BL , Microarray Analysis/methods , Plasmids , RNA, Small Interfering/chemistry , Transfection
7.
J Hepatol ; 51(5): 853-64, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19726098

ABSTRACT

BACKGROUND/AIMS: The nonstructural 5A (NS5A) protein of hepatitis C virus (HCV) has been implicated in HCV-induced liver pathogenesis. Wnt/beta-catenin signaling has also been involved in tumorigenesis. To elucidate the molecular mechanism of HCV pathogenesis, we examined the potential effects of HCV NS5A protein on Wnt/beta-catenin signal transduction cascades. METHODS: The effects of NS5A protein on beta-catenin signaling cascades in hepatic cells were investigated by luciferase reporter gene assay, confocal microscopy, immunoprecipitation assay, and immunoblot analysis. RESULTS: beta-Catenin-mediated transcriptional activity is elevated by NS5A protein, in the context of HCV replication, and by infection of cell culture-produced HCV. NS5A protein directly interacts with endogenous beta-catenin and colocalizes with beta-catenin in the cytoplasm. NS5A protein inactivates glycogen synthase kinase 3beta and increases subsequent accumulation of beta-catenin in HepG2 cells. beta-Catenin was also accumulated in HCV patients' liver tissues. In addition, the accumulation of beta-catenin in HCV replicon cells requires both activation of phosphatidylinositol 3-kinase and inactivation of GSK3beta. CONCLUSIONS: NS5A activates beta-catenin signaling cascades through increasing the stability of beta-catenin. This modulation is accomplished by the protein interplay between viral and cellular signaling transducer. These data suggest that NS5A protein may directly be involved in Wnt/beta-catenin-mediated liver pathogenesis.


Subject(s)
Hepacivirus/physiology , Hepacivirus/pathogenicity , Liver Neoplasms/etiology , Viral Nonstructural Proteins/metabolism , beta Catenin/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , COS Cells , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/virology , Cell Line , Cell Nucleus/metabolism , Chlorocebus aethiops , Cytosol/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hepacivirus/genetics , Humans , In Vitro Techniques , Liver Neoplasms/metabolism , Liver Neoplasms/virology , Phosphatidylinositol 3-Kinases/metabolism , Protein Interaction Domains and Motifs , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Replicon , Sequence Deletion , Signal Transduction , Transcription Factor 4 , Transcription Factors/metabolism , Transfection , Viral Nonstructural Proteins/genetics , Virus Replication , Wnt Proteins/metabolism , beta Catenin/chemistry , beta Catenin/genetics
8.
Virus Res ; 142(1-2): 51-6, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19189853

ABSTRACT

Hepatitis C virus (HCV) infection is currently treated with IFNalpha-based therapy but little is known how IFNalpha inhibits HCV replication. We show here that HCV JFH1 infection of human hepatoma Huh-7 cells leads to the activation of IFN-inducible protein kinase PKR and phosphorylation of the translation initiation factor eIF2alpha. Compared to a control cell HCV replication was significantly elevated in a PKR-knockdown cell, giving rise to a 10-fold higher viral titer, and was less sensitive to IFNalpha treatment. Conversely, transient expression of PKR inhibited HCV replication in a kinase-dependent manner with concomitant increase of eIF2alpha phosphorylation. Further, expression of a phospho-mimetic eIF2alpha mutant moderately inhibited HCV replication. Together, these results demonstrate that PKR is activated by HCV infection and plays a critical antiviral role through inhibition of viral protein translation.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Hepacivirus/physiology , Hepatitis C/enzymology , Protein Biosynthesis , Transcriptional Activation , Virus Replication , eIF-2 Kinase/genetics , Cell Line , Eukaryotic Initiation Factor-2/genetics , Hepacivirus/genetics , Hepatitis C/genetics , Hepatitis C/virology , Humans , Phosphorylation , Viral Proteins/genetics , Viral Proteins/metabolism , eIF-2 Kinase/metabolism
9.
Cell Microbiol ; 10(3): 583-92, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17877750

ABSTRACT

The vaccinia virus (VV) replicates robustly and alters the progression of the cell cycle via an unknown mechanism. Herein, we provide evidence for the existence of a unique VV infection-induced cell cycle control mechanism. The regulation is correlated with the inactivation of p53 and Rb, which are associated with the RNA polymerase III transcription factor B (TFIIIB) subunits, TBP and Brf1 respectively. VV infection induced the expression of Mdm2 and its translocation into the nucleus, thereby resulting in a disruption of p53. VV also stimulated the expression of TFIIIB and TFIIIC, and consequently induced tRNA synthesis. On the other hand, the total level of Rb was not significantly influenced, but the level of hypo-phosphorylated Rb was enhanced, partially due to the VV-induced downregulation of cyclin-dependent kinases 4 and 6. However, the hypo-phosphorylated Rb appeared to be largely sequestered into a complex with Brf1, which resulted in the blockage of Rb function to repress E2F1 transactivation, thereby leading to a moderately higher proportion of cells in the S and G(2) phases. Conversely, the enforced expression of exogenous Rb restored the normally observed cell cycle patterns. Overall, these controls may contribute to the efficient replication of the virus in rapidly growing cells.


Subject(s)
Cell Cycle , TATA-Binding Protein Associated Factors/metabolism , TATA-Box Binding Protein/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Vaccinia virus/physiology , Cell Line , Cell Nucleus/chemistry , Cyclin-Dependent Kinase 4/biosynthesis , Cyclin-Dependent Kinase 6/biosynthesis , Down-Regulation , E2F1 Transcription Factor/metabolism , Humans , Phosphorylation , Proto-Oncogene Proteins c-mdm2/metabolism , RNA Polymerase III/metabolism , RNA, Transfer/biosynthesis , Transcription Factor TFIIIB/biosynthesis , Transcription Factors, TFIII/biosynthesis , Up-Regulation
10.
Mol Cell Biol ; 26(8): 3048-59, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16581780

ABSTRACT

Hepatitis C virus (HCV) NS5B protein is a membrane-associated phosphoprotein that possesses an RNA-dependent RNA polymerase activity. We recently reported that NS5A protein interacts with TRAF2 and modulates tumor necrosis factor alpha (TNF-alpha)-induced NF-kappaB and Jun N-terminal protein kinase (JNK). Since NS5A and NS5B are the essential components of the HCV replication complex, we examined whether NS5B could modulate TNF-alpha-induced NF-kappaB and JNK activation. In this study, we have demonstrated that TNF-alpha-induced NF-kappaB activation is inhibited by NS5B protein in HEK293 and hepatic cells. Furthermore, NS5B protein inhibited both TRAF2- and IKK-induced NF-kappaB activation. Using coimmunoprecipitation assays, we show that NS5B interacts with IKKalpha. Most importantly, NS5B protein in HCV subgenomic replicon cells interacted with endogenous IKKalpha, and then TNF-alpha-mediated IKKalpha kinase activation was significantly decreased by NS5B. Using in vitro kinase assay, we have further found that NS5B protein synergistically activated TNF-alpha-mediated JNK activity in HEK293 and hepatic cells. These data suggest that NS5B protein modulates TNF-alpha signaling pathways and may contribute to HCV pathogenesis.


Subject(s)
Hepacivirus/metabolism , I-kappa B Kinase/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Viral Nonstructural Proteins/physiology , Animals , COS Cells , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Enzyme Activation/drug effects , Genes, Reporter , Hepacivirus/genetics , Humans , I-kappa B Kinase/analysis , JNK Mitogen-Activated Protein Kinases/analysis , JNK Mitogen-Activated Protein Kinases/metabolism , Liver Neoplasms/pathology , Luciferases/metabolism , MAP Kinase Kinase 4/metabolism , Precipitin Tests , Protein Binding , Tumor Necrosis Factor-alpha/pharmacology , Viral Nonstructural Proteins/genetics
11.
Mol Biol Cell ; 17(2): 824-33, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16314389

ABSTRACT

Heat-shock protein 90 (Hsp90) is a molecular chaperone that plays a key role in the conformational maturation of various transcription factors and protein kinases in signal transduction. Multifunctional ribosomal protein S3 (rpS3), a component of the ribosomal small subunit, is involved in DNA repair and apoptosis. Our data show that Hsp90 binds directly to rpS3 and the functional consequence of Hsp90-rpS3 interaction results in the prevention of the ubiquitination and the proteasome-dependent degradation of rpS3, subsequently retaining the function and the biogenesis of the ribosome. Interference of Hsp90 activity by Hsp90 inhibitors appears to dissociate rpS3 from Hsp90, associate the protein with Hsp70, and induce the degradation of free forms of rpS3. Furthermore, ribosomal protein S6 (rpS6) also interacted with Hsp90 and exhibited a similar effect upon treatment with Hsp90 inhibitors. Therefore, we conclude that Hsp90 regulates the function of ribosomes by maintaining the stability of 40S ribosomal proteins such as rpS3 and rpS6.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Proteasome Endopeptidase Complex/physiology , Ribosomal Proteins/metabolism , Ubiquitin/metabolism , Benzoquinones , Cell Line , Cytosol/metabolism , HSP70 Heat-Shock Proteins/physiology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/physiology , Lactams, Macrocyclic , Microscopy, Confocal , Protein Binding , Quinones/pharmacology , Recombinant Fusion Proteins/metabolism , Ribosomal Proteins/genetics , Ribosomes/physiology
12.
Antiviral Res ; 170: 104572, 2019 10.
Article in English | MEDLINE | ID: mdl-31376425

ABSTRACT

Cellular protein kinases play critical roles in various steps of the hepatitis B virus life cycle. We found that viral replication in infected or transfected hepatoma cell was markedly inhibited by treatment with A-443654, a specific inhibitor of Akt. The antiviral mechanism of the drug mainly depended on the downregulation of Aurora A, a protein kinase that plays an essential role in mitosis but has not been implicated in the viral life cycle. Our data indicated that Aurora kinase A enhances viral replication and expression independently of its kinase activity required for mitotic function. Our findings suggest that mitotic kinases, considered to be an attractive target of antitumor agents, also provide a novel target for the development of antiviral therapy.


Subject(s)
Antiviral Agents/pharmacology , Aurora Kinase A/genetics , Indazoles/pharmacology , Indoles/pharmacology , Virus Replication/drug effects , Aurora Kinase A/antagonists & inhibitors , Down-Regulation , Enzyme Inhibitors/pharmacology , HeLa Cells , Hep G2 Cells , Hepatitis B virus/drug effects , Hepatitis B virus/physiology , Humans
13.
Biochem Biophys Res Commun ; 366(4): 1036-42, 2008 Feb 22.
Article in English | MEDLINE | ID: mdl-18155658

ABSTRACT

The hepatitis B virus X protein (HBX) plays key regulatory roles in viral replication and the development of hepatocellular carcinoma. HBX is an unstable protein; its instability is attributed to rapid degradation through the ubiquitin-proteasome pathway. Here, we show that the middle and carboxyl-terminal domains of HBX, independently fused to GFP, render the recombinant proteins susceptible to proteasomal degradation, while the amino-terminal domain has little effect on the ubiquitination or stability of HBX. Mutation of any single or combination of up to five of six lysine residues, all located in the middle and carboxyl-terminal domain, did not prevent HBX from being ubiquitinated, ruling out any specific lysine as the sole site of ubiquitination. Surprisingly, HBX in which all six lysines were mutated and showed no evidence of ubiquitination, was still susceptible to proteasomal degradation. These results suggest that both ubiquitin-dependent and -independent proteasomal degradation processes are operative in HBX turnover.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , Trans-Activators/metabolism , Ubiquitin/metabolism , Humans , Lysine , Mutant Proteins/metabolism , Protein Structure, Tertiary , Structure-Activity Relationship , Trans-Activators/chemistry , Ubiquitination , Viral Regulatory and Accessory Proteins
14.
Mol Cells ; 24(2): 194-9, 2007 Oct 31.
Article in English | MEDLINE | ID: mdl-17978571

ABSTRACT

GCN4 is a typical eukaryotic transcriptional activator that is implicated in the expression of many genes involved in amino acids and purine biosyntheses under stress conditions. It is degraded by 26S proteasomes following ubiquitination. However, the immediate receptor for ubiquitinated Gcn4p has not yet been identified. We investigated whether ubiquitinated Gcn4p binds directly to Rpn10p as the ubiquitinated substrate receptor of the 26S proteasome. We found that the level of Gcn4p increased in cells deleted for Rpn10p but not in cells deleted for RAD23 and DSK2, the other ubiquitinated substrate receptors and, unlike Rpn10p, neither of these proteins recognized ubiquitinated Gcn4p. These results suggest that Rpn10p is the receptor that binds the polyubiquitin chain during ubiquitin-dependent proteolysis of Gcn4p.


Subject(s)
Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , Receptors, Cell Surface/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism , Ubiquitination , Basic-Leucine Zipper Transcription Factors , Cell Cycle Proteins/metabolism , Gene Deletion , Polyubiquitin/metabolism , Protein Binding , Saccharomyces cerevisiae/enzymology , Thermodynamics , Ubiquitins/metabolism
15.
Mol Cells ; 21(1): 21-8, 2006 Feb 28.
Article in English | MEDLINE | ID: mdl-16511343

ABSTRACT

The interferon-induced, double-stranded RNA (dsRNA)-dependent protein kinase PKR plays a key role in interferon-mediated host defense against viral infection, and is implicated in cellular transformation and apoptosis. We have isolated a cDNA of simian PKR encoding a product with 83% amino acid identity to the human homolog and showed that PKR expression is significantly attenuated in the Vero E6 African green monkey kidney cells devoid of type I interferon genes. A variant form of PKR lacking the exon 12 in the kinase domain is produced in these cells, presumably from an alternatively spliced transcript. Unlike wild type PKR, the variant protein named PKR-DE12 is incapable of auto-phosphorylation and phosphorylation of eIF2-a, indicating that the kinase sub-domains III and IV embedded in exon 12 are indispensable for catalytic function. PKR-DE12 had no dominant negative effect but was weakly phosphorylated in trans by wild type PKR.


Subject(s)
Adenoviruses, Simian/metabolism , Gene Expression Regulation/drug effects , Interferon Type I/deficiency , Interferon-alpha/pharmacology , eIF-2 Kinase/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Chlorocebus aethiops , DNA, Complementary/genetics , Exons/genetics , Humans , Interferon alpha-2 , Molecular Sequence Data , Phosphorylation , Phosphotransferases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins , Sequence Alignment , Sequence Deletion/genetics , Tumor Cells, Cultured , Vero Cells , eIF-2 Kinase/genetics
16.
J Virol Methods ; 137(2): 280-6, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16890998

ABSTRACT

Nucleic acid amplification techniques are used frequently for rapid diagnosis of viral diseases. In this study, a real-time polymerase chain reaction protocol that uses primers specific for the viral VP4 gene and the commercial SYBR Green reagent were evaluated for the quantitative measurement of human rotavirus (HRV) RNA in human stool specimens. SYBR Green I detection involved analysis of the melting temperature of the PCR product and measurement of fluorescence at the optimum temperature. The assay resulted in a sensitive and reproducible detection of targets ranging from low (<10(2)rotavirus cDNA copies/reaction) to high numbers (>10(6)rotavirus cDNA copies/reaction). No cross-reaction was found with crude cell culture stocks of coxsackievirus, echovirus, poliovirus, hepatitis A virus and adenovirus. Analysis with the HRV cDNA standard demonstrated high reproducibility with a coefficient of variation (CV) of 0.2-0.9%. Daily performance among three different laboratories showed a CV no greater than 8%, indicating an intermediate level of variation. These results demonstrate the feasibility of this method for quantitative analysis of human rotavirus in clinical samples.


Subject(s)
RNA, Viral/analysis , Reverse Transcriptase Polymerase Chain Reaction , Rotavirus Infections/virology , Rotavirus/isolation & purification , Analysis of Variance , Benzothiazoles , Capsid Proteins/genetics , Child , Child, Preschool , Diamines , Feces/virology , Humans , Infant , Observer Variation , Organic Chemicals/metabolism , Quinolines , RNA, Viral/genetics , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction/methods , Reverse Transcriptase Polymerase Chain Reaction/standards , Rotavirus/genetics , Rotavirus Infections/diagnosis , Sensitivity and Specificity , Transition Temperature
17.
Virology ; 495: 71-8, 2016 08.
Article in English | MEDLINE | ID: mdl-27179347

ABSTRACT

SAMHD1 is a cellular protein that possesses dNTPase activity and inhibits retroviruses and DNA viruses through the depletion of cellular dNTPs. However, recent evidence suggests the existence of alternative or additional mechanisms that involve novel nuclease activities. Hepatitis B virus is a DNA virus but resembles retroviruses in that its DNA genome is synthesized via reverse transcription of an RNA transcript. SAMHD1 was shown to inhibit the expression and replication of a transfected HBV DNA. We further investigated the antiviral mechanisms in a newly developed infection assay. Our data indicated that SAMHD1 exerts a profound antiviral effect. In addition, unlike previous findings, our results demonstrate the essential role of SAMHD1 dNTPase. SAMHD1 did not affect virion-derived cccDNA and gene expression but specifically inhibited viral DNA synthesis. These results indicate that SAMHD1 inhibits HBV replication at the reverse transcription step, most likely through the depletion of cellular dNTPs.


Subject(s)
Hepatitis B virus/physiology , Hepatitis B/metabolism , Hepatitis B/virology , Host-Pathogen Interactions , Monomeric GTP-Binding Proteins/metabolism , Virus Replication , Cell Line , DNA Replication , Humans , Reverse Transcription , SAM Domain and HD Domain-Containing Protein 1
18.
Reprod Toxicol ; 60: 156-66, 2016 04.
Article in English | MEDLINE | ID: mdl-26867867

ABSTRACT

The endocrine-disrupting effects of androgenic signaling play crucial roles in several androgen-related diseases. In attempting to develop an in vitro cell line to be used in androgen receptor (AR)-mediated reporter gene assays, we developed a stable 22Rv1/MMTV cell line, which is a human prostate cancer cell line that endogenously expresses functional AR, to evaluate AR-mediated transcriptional activation (TA). Using 22Rv1/MMTV cells, we established and optimized a test protocol for the AR-TA assay and validated the proposed assay using 20 compounds recommended by the Interagency Coordinating Committee on the Validation of Alternative Methods (ICCVAM). All the performance parameters for agonist and antagonist assays were 91-100% comparable between the 22Rv1/MMTV assay and the ICCVAM report. In conclusion, the AR-TA assay using 22Rv1/MMTV cells might be a quick and relatively inexpensive method for screening large numbers of chemicals for their potential to activate or inhibit AR-mediated gene transcription.


Subject(s)
Androgen Antagonists/pharmacology , Androgens/pharmacology , Receptors, Androgen/genetics , Transcriptional Activation/drug effects , Biological Assay , Cell Line, Tumor , Genes, Reporter , Humans , Luciferases/metabolism
19.
PLoS One ; 10(7): e0132839, 2015.
Article in English | MEDLINE | ID: mdl-26185986

ABSTRACT

Hepatitis C virus (HCV) usurps host cellular lipid metabolism for production of infectious virus particles. Recently, we have screened a siRNA library targeting host factors that control lipid metabolism and lipid droplet (LD) formation in cell culture grown HCV (HCVcc)-infected cells. Of 10 final candidates, we selected the gene encoding AAM-B for further characterization. We showed that siRNA-mediated knockdown of AAM-B impaired HCV propagation in Jc1-infected cells. More precisely, knockdown of AAM-B abrogated production of infectious HCV particles in both Jc1 RNA electroporated cells and Jc1-infected cells. It is worth noting that knockdown of AAM-B exerted no effect on lipid droplet formation. Moreover, AAM-B interacted with nonstructural 4B (NS4B) through the C-terminal region of NS4B. Protein interplay between AAM-B and NS4B was verified in the context of HCV replication. Using either transient or stable expression of AAM-B, we verified that AAM-B colocalized with NS4B in the cytoplasm. Immunofluorescence data further showed that AAM-B might be involved in recruitment of NS4B to sites in close proximity to LDs to facilitate HCV propagation. Collectively, this study provides new insight into how HCV utilizes cellular AAM-B to facilitate viral propagation.


Subject(s)
Hepacivirus/metabolism , Hepatocytes/metabolism , Lipid Droplets/metabolism , Membrane Proteins/metabolism , Methyltransferases/metabolism , Viral Nonstructural Proteins/metabolism , Cell Line, Tumor , Gene Expression Regulation , Hepacivirus/genetics , Hepacivirus/growth & development , Hepatocytes/ultrastructure , Hepatocytes/virology , Host-Pathogen Interactions , Humans , Lipid Droplets/ultrastructure , Lipid Droplets/virology , Lipid Metabolism/genetics , Membrane Proteins/genetics , Methyltransferases/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Viral Nonstructural Proteins/genetics , Virus Replication/genetics
20.
Antiviral Res ; 104: 118-27, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24509240

ABSTRACT

RNase L is a cellular endoribonuclease that is activated by 2',5'-linked oligoadenylates (2-5A), which are unique and specific ligands synthesized by a family of interferon-inducible, dsRNA-activated enzymes named oligoadenylate synthetases. In the typical antiviral pathway, activated RNase L degrades viral and cellular RNAs, thus limiting viral replication and spread. Although the antiviral activity of RNase L has been demonstrated for several RNA viruses, there is little evidence regarding its role against DNA viruses. In the present study, the potential antiviral activity of RNase L against hepatitis B virus (HBV) was explored utilizing the recently reported infection protocol based on human hepatoma HepG2 cells stably complemented with the virus entry factor NTCP. Viral replication and expression in this cell type was markedly inhibited by poly(I:C)- or 2-5A-mediated activation of RNase L; however, the inhibition was significantly reversed by RNase L knockdown. Further analysis in HBV1.2-transfected Huh-7 hepatoma cells indicated that the antiviral activity of RNase L depends on its ribonuclease function. We also provide evidence for the specific roles of OAS family members in this process. These results suggest that HBV replication can be regulated through interferon-mediated RNA decay pathways and that activation of these host antiviral factors may represent a novel therapeutic strategy for HBV infection.


Subject(s)
Endoribonucleases/metabolism , Hepatitis B virus/physiology , Virus Replication , Adenine Nucleotides/pharmacology , Cell Line , Endoribonucleases/genetics , Enzyme Activation/drug effects , Gene Expression , Gene Knockdown Techniques , Hepatitis B virus/drug effects , Humans , Isoenzymes , Ligands , Oligoribonucleotides/pharmacology , Poly I-C/pharmacology , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL