Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters











Type of study
Publication year range
1.
Microbiol Spectr ; 10(1): e0134221, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35138119

ABSTRACT

Human phospholipid scramblase 1 (PLSCR1) is strongly expressed in response to interferon (IFN) treatment and viral infection, and it has been suggested to play an important role in IFN-dependent antiviral responses. In this study, we showed that the levels of human cytomegalovirus (HCMV) plaque formation in OUMS-36T-3 (36T-3) cells with high basal expression of PLSCR1 were significantly lower than those in human embryonic lung (HEL) cells with low basal expression of PLSCR1. In addition, the levels of HCMV plaque formation and replication in PLSCR1-knockout (KO) 36T-3 cells were significantly higher than those in parental 36T-3 cells and were comparable to those in HEL cells. Furthermore, compared to that in PLSCR1-KO cells, the expression of HCMV major immediate early (MIE) proteins was repressed and/or delayed in parental 36T-3 cells after HCMV infection. We also showed that PLSCR1 expression decreased the levels of the cAMP-responsive element (CRE)-binding protein (CREB)•HCMV immediate early protein 2 (IE2) and CREB-binding protein (CBP)•IE2 complexes, which have been suggested to play important roles in the IE2-mediated transactivation of the viral early promoter through interactions with CREB, CBP, and IE2. Interestingly, PLSCR1 expression repressed CRE- and HCMV MIE promoter-regulated reporter gene activities. These observations reveal, for the first time, that PLSCR1 negatively regulates HCMV replication by repressing the transcription from viral MIE and early promoters, and that PLSCR1 expression may contribute to the IFN-mediated suppression of HCMV infection. IMPORTANCE Because several IFN-stimulated genes (ISGs) have been reported to suppress HCMV replication, HCMV replication is thought to be regulated by an IFN-mediated host defense mechanism, but the mechanism remains unclear. PLSCR1 expression is induced in response to viral infection and IFN treatment, and PLSCR1 has been reported to play an important role in IFN-dependent antiviral responses. Here, we demonstrate that HCMV plaque formation and major immediate early (MIE) gene expression are significantly increased in PLSCR1-KO human fibroblast cells. PLSCR1 reduces levels of the CREB•IE2 and CBP•IE2 complexes, which have been suggested to play important roles in HCMV replication through its interactions with CREB, CBP, and IE2. In addition, PLSCR1 expression represses transcription from the HCMV MIE promoter. Our results indicate that PLSCR1 plays important roles in the suppression of HCMV replication in the IFN-mediated host defense system.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus/physiology , Interferons/immunology , Phospholipid Transfer Proteins/immunology , Antigens, Viral/genetics , Antigens, Viral/metabolism , CREB-Binding Protein/genetics , CREB-Binding Protein/immunology , Cytomegalovirus/genetics , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/virology , Gene Expression Regulation, Viral , Host-Pathogen Interactions , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Interferons/genetics , Phospholipid Transfer Proteins/genetics , Virus Replication
2.
FEBS Open Bio ; 8(4): 646-654, 2018 04.
Article in English | MEDLINE | ID: mdl-29632816

ABSTRACT

4',5,7-trihydroxy-3',5'-dimethoxyflavone (tricin), derived from Sasa albo-marginata, has been reported to suppress significantly human cytomegalovirus (HCMV) replication in human embryonic lung (HEL) fibroblast cells. However, the target protein of tricin remains unclear. This study focused on the anti-HCMV activity of tricin in terms of its binding affinity to cyclin-dependent kinase 9 (CDK9). A molecular docking study predicted that tricin binds well to the ATP-binding site of CDK9. Experimental measurements then revealed that tricin inhibits the kinase activity of CDK9 and affects the phosphorylation of the carboxy-terminal domain of RNA polymerase II. Based on these results, we conclude that CDK9 is one of the target proteins of tricin. We also found that tricin possesses anti-HCMV activity with no cytotoxicity against HEL cells.

3.
Microbiol Immunol ; 62(5): 341-347, 2018 May.
Article in English | MEDLINE | ID: mdl-29603339

ABSTRACT

Treatment of human embryonic lung fibroblast (HEL) cells with tricin (4', 5, 7-trihydroxy-3', 5'-dimethoxyflavone) following infection with human cytomegalovirus (HCMV) reportedly significantly suppresses HCMV replication. In the present work, the mechanisms for the anti-HCMV effects of tricin in HEL cells were examined. It was found that exposure of HEL cells to tricin inhibited HCMV replication, with concomitant decreases in amounts of transcripts of the CC chemokine RANTES (CCL5)-encoding gene and in expression of the CCL5 protein. It was also found that transcripts of HCMV immediate early 1 (IE1), and HCMV UL54 (encoding DNA polymerase) and replication of HCMV was significantly lower in CCL5 gene-knockdown cells. These results suggest that the anti-HCMV activity of tricin differs from that of ganciclovir and that CCL5 is one of the chemokines involved in HCMV replication. In addition, it is possible that chemokine CCL5 is one of the targets of tricin.


Subject(s)
Antiviral Agents/antagonists & inhibitors , Chemokine CCL5/genetics , Cytomegalovirus Infections/virology , Cytomegalovirus/growth & development , Flavonoids/antagonists & inhibitors , Gene Expression/drug effects , Virus Replication/drug effects , Cell Line , Cytomegalovirus/drug effects , Cytomegalovirus/genetics , DNA Replication/drug effects , DNA-Directed DNA Polymerase , Fibroblasts/drug effects , Ganciclovir/antagonists & inhibitors , Gene Knockdown Techniques , Gene Silencing , Humans , Immediate-Early Proteins , RNA, Small Interfering , Transfection , Viral Proteins/genetics
5.
Antiviral Res ; 154: 10-16, 2018 06.
Article in English | MEDLINE | ID: mdl-29559264

ABSTRACT

A novel type of antiviral agent for human cytomegalovirus (HCMV) is required, because the appearance of ganciclovir (GCV) resistant viruses has been reported. Tricin (4',5,7-trihydroxy-3',5'-dimethoxyflavone) has been shown to suppress significantly HCMV replication in human embryonic lung (HEL) fibroblast cells. Recently, we revealed that the action of tricin is different from that of GCV and cyclin-dependent kinase 9 (CDK9) is one of the target proteins of tricin. These results suggested that tricin is considered as a novel type of anti-HCMV agent. However, its anti-HCMV potency is not greater than that of GCV. This study tried to develop novel compounds with much greater anti-HCMV activity than GCV. We first made modifications to tricin by introducing fluorine atom, and then performed molecular docking simulations using the designed compounds and CDK9. The calculated binding energies showed that 6F-tricin (6-fluoro-4'-hydroxy-3',5'-dimetoxyflavone) binds to CDK9 much stronger than tricin. Based on these results, 6F-tricin was synthesized, and then its anti-HCMV effect was analyzed in HEL cell cultures. As a result, 6F-tricin strongly suppressed HCMV replication in a dose-dependent manner. The anti-HCMV activity with a 50% effective concentration (EC50) was 0.126 nM, corresponding to about 1/200 and 1/400 of EC50 of GCV (27.5 nM) and tricin (54.3 nM), respectively. Moreover, 6F-tricin had no cytotoxicity against HEL cells at concentrations up to 10 µM. We further performed detailed analysis on the amino acid contributions to the binding energies and found that the strong binding affinity for 6F-tricin to CDK9 is attributed to the specific binding orientation of 6F-tricin in the ATP-binding site. These results suggest that 6F-tricin is a promising candidate for anti-HCMV drug development.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cytomegalovirus/drug effects , Fibroblasts/virology , Flavones/chemistry , Flavones/pharmacology , Ganciclovir/pharmacology , Virus Replication/drug effects , Cell Line , Cytomegalovirus/physiology , DNA Replication , Drug Design , Humans , Molecular Docking Simulation
6.
Antiviral Res ; 148: 15-19, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28965916

ABSTRACT

We previously reported that treatment with tricin (4',5,7-trihydroxy-3',5'-dimethoxyflavone) after human cytomegalovirus (HCMV) infection significantly suppressed both infectious virion production and HCMV replication in human embryonic lung fibroblast (HEL) cells. Moreover, we recently demonstrated that HCMV infection can increase the expression of CC-motif ligand 2 (CCL2/MCP-1) and of CCR2, a CCL2-specific receptor, effects that can in turn enhance HCMV infection and replication. Hence, we here examined whether the CCL2-CCR2 axis is involved in the anti-HCMV effects of tricin in HEL cells. Tricin exposure yielded dose-dependent decreases in the accumulation of transcripts for the HCMV immediate early gene and the DNA polymerase gene in HCMV-infected cells, along with decreased production of infectious HCMV. Concomitantly, tricin caused dose-dependent attenuation of HCMV infection-induced up-regulation of expression of CCL2 and CCR2 mRNAs and of CCL2 protein. Moreover, CCL2 reversed tricin-mediated inhibition of HCMV virion production in a dose-dependent manner. Thus, tricin appears to exert anti-HCMV activity by depressing CCL2 expression.


Subject(s)
Antiviral Agents/pharmacology , Chemokine CCL2/genetics , Cytomegalovirus/drug effects , Flavonoids/pharmacology , Gene Expression Regulation, Viral/drug effects , Virus Replication/drug effects , Cell Line , Chemokine CCL2/metabolism , Cytomegalovirus/growth & development , Cytomegalovirus/physiology , Cytomegalovirus Infections/virology , DNA-Directed DNA Polymerase/genetics , Fibroblasts , Host-Pathogen Interactions/drug effects , Humans , Immediate-Early Proteins/genetics , Receptors, CCR2/genetics , Viral Proteins/genetics
7.
Biol Pharm Bull ; 39(12): 1912-1921, 2016.
Article in English | MEDLINE | ID: mdl-27904034

ABSTRACT

In order to understand a possible etiology of adverse pregnancy outcomes associated with intrauterine influenza virus infection, we examined the effect of influenza virus infection on gene expression of matrix metalloproteinases (MMPs) in cultured amnion epithelial, amnion mesenchymal and chorion trophoblast cells prepared from human fetal membrane tissues by gelatin zymography, Western blotting and reverse transcriptase-PCR. The cells were infected with influenza A (H1N1) virus. The levels of pro-MMP-9 activity in culture supernatants of three types of cells were increased during the period of 24-48 h after the virus infection as compared to those of mock infection. Chorion trophoblast cells spontaneously released a much greater level of pro-MMP-2 activity than amnion epithelial and amnion mesenchymal cells. The cleavage of pro-MMP-2 into an active intermediate form was enhanced in chorion trophoblast cells by the virus infection. The activity levels of MMP-2 and MMP-9 in culture supernatants were consistent with their protein levels. The virus infection induced the mRNA expression of MMP-9, but not MMP-2, in three types of cells. These results suggest that influenza virus infection induces the gene expression of MMP-9 and the cleavage of pro-MMP-2 into an active intermediate form in human fetal membrane cells, resulting in weakening of the membranes through extracellular matrix degradation. Therefore, it is possible that the regulation of MMPs gene expression in fetal membrane cells by influenza virus infection is implicated in a part of the etiology of adverse pregnancy outcomes associated with intrauterine infection with the virus.


Subject(s)
Extraembryonic Membranes/cytology , Influenza A Virus, H1N1 Subtype , Influenza, Human/genetics , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Cells, Cultured , Cytoskeleton/metabolism , Epithelial Cells/metabolism , Epithelial Cells/virology , Extraembryonic Membranes/metabolism , Extraembryonic Membranes/virology , Female , Gene Expression Regulation , Humans , Influenza, Human/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/virology , Pregnancy , Trophoblasts/metabolism , Trophoblasts/virology
8.
Antiviral Res ; 125: 79-83, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26640224

ABSTRACT

It has been demonstrated as the first report that combination treatment with ganciclovir (GCV) and tricin (4',5,7-trihydroxy-3',5' -dimethoxyflavone), a derivative of Sasa albo-marginata, after human cytomegalovirus (HCMV) infection has synergistic effects on both infectious virus production and HCMV DNA synthesis in the human embryonic fibroblast cell line MRC-5. In this paper, we examined the anti-HCMV effects of GCV plus various concentrations of tricin, and tricin plus various concentrations of GCV in MRC-5 cells. We found that expression of the HCMV UL54 gene was significantly inhibited by combination of GCV with tricin when compared with GCV mono-treatment. These results suggest that tricin is a novel compound for combination therapy with GCV against HCMV replication. In addition, reduced-dose combination therapy may provide a direction for treatment in patients with HCMV infection while reducing drug toxicity.


Subject(s)
Antiviral Agents/pharmacology , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/virology , Cytomegalovirus/physiology , Flavonoids/pharmacology , Ganciclovir/pharmacology , Virus Replication/drug effects , Cell Line , Cytomegalovirus/drug effects , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , DNA Replication/drug effects , DNA-Directed DNA Polymerase/biosynthesis , DNA-Directed DNA Polymerase/genetics , Drug Synergism , Drug Therapy, Combination , Fibroblasts/virology , Gene Expression , Humans , Male , Sasa/chemistry , Viral Proteins/biosynthesis , Viral Proteins/genetics
9.
Folia Microbiol (Praha) ; 60(6): 505-14, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25906999

ABSTRACT

Full-length tlyA gene and its adjacent genetic loci from the urease-positive thermophilic Campylobacter (UPTC) CF89-12 [approximately 15,000 base pairs (bp) in length], as well as a reference strain Campylobacter lari RM2100 (approximately 9,000 bp), were analyzed. The possible open-reading frame of tlyA from UPTC CF89-12 was shown to have 720 bp with a calculated molecular mass of approximately 26.7 kDa. Using a primer pair designed in silico, a total of approximately 1.1 kbp consisting of putative promoter region, structural gene for tlyA, and its adjacent genetic loci were identified in all 17 C. lari isolates [n = 13 for UPTC; n = 4 for urease-negative (UN) C. lari]. Although sequence differences were demonstrated at approximately 20 loci within the 90 bp non-coding (NC) region, including the putative promoter structure candidates immediately upstream of the tlyA gene among the 18 isolates including C. lari RM2100, no sequence differences were identified within the NC region among the five UN C. lari isolates examined. A start codon ATG and a probable ribosome-binding site, AGGC(T)GG(A), for the tlyA gene were identified in all 18 isolates, including C. lari RM2100. The putative intrinsic ρ-independent transcriptional terminator structure candidate was also identified for the tlyA gene in both UPTC CF89-12 and C. lari RM2100. Additionally, the hemolysis assay was performed with some of the C. lari isolates. The tlyA gene nucleotide sequence data may possibly be useful for discrimination between UN C. lari and UPTC organisms, as well as for the differentiation among the four thermophilic Campylobacter species.


Subject(s)
Bacterial Proteins/genetics , Campylobacter Infections/microbiology , Campylobacter Infections/veterinary , Campylobacter lari/genetics , Hemolysin Proteins/genetics , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , Base Sequence , Birds/microbiology , Bivalvia/microbiology , Campylobacter lari/chemistry , Campylobacter lari/classification , Campylobacter lari/isolation & purification , Chickens/microbiology , Environmental Microbiology , Hemolysin Proteins/chemistry , Molecular Sequence Data , Open Reading Frames , Phylogeny , Promoter Regions, Genetic , Sequence Alignment
10.
Biochem Biophys Res Commun ; 453(3): 321-5, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25261725

ABSTRACT

We previously revealed that human cytomegalovirus (HCMV) infection can cause aberrant expression of the chemokine IL-8/CXCL8. We first examined the effects of HCMV infection on the expression of another chemokine, CCL2. HCMV infection induced CCL2 expression at the mRNA and protein levels in human embryonic lung fibroblasts cells (HEL). Moreover, HCMV induced the mRNA expression of CCR2, a specific receptor for CCL2. CCL2 siRNA treatment reduced HCMV virion production, and this reduction was reversed by the addition of CCL2. We further observed that CCL2 siRNA, but not control siRNA, reduced the expression of HCMV immediate early gene (IE1) and HCMV UL54 gene (DNA polymerase) in a dose-dependent manner. Thus, HCMV infection is able to activate the CCL2-CCR2 interactions to further enhance HCMV infection and/or replication.


Subject(s)
Chemokine CCL2/physiology , Cytomegalovirus/physiology , Receptors, CCR2/physiology , Virus Replication/physiology , Base Sequence , Cells, Cultured , DNA Primers , Humans , Polymerase Chain Reaction
11.
Plant Biotechnol J ; 11(6): 734-46, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23551455

ABSTRACT

Flavonoids possess diverse health-promoting benefits but are nearly absent from rice, because most of the genes encoding enzymes for flavonoid biosynthesis are not expressed in rice seeds. In the present study, a transgenic rice plant producing several classes of flavonoids in seeds was developed by introducing multiple genes encoding enzymes involved in flavonoid synthesis, from phenylalanine to the target flavonoids, into rice. Rice accumulating naringenin was developed by introducing phenylalanine ammonia lyase (PAL) and chalcone synthase (CHS) genes. Rice producing other classes of flavonoids, kaempferol, genistein, and apigenin, was developed by introducing, together with PAL and CHS, genes encoding flavonol synthase/flavanone-3-hydroxylase, isoflavone synthase, and flavone synthases, respectively. The endosperm-specific GluB-1 promoter or embryo- and aleurone-specific 18-kDa oleosin promoters were used to express these biosynthetic genes in seed. The target flavonoids of naringenin, kaempferol, genistein, and apigenin were highly accumulated in each transgenic rice, respectively. Furthermore, tricin was accumulated by introducing hydroxylase and methyltransferase, demonstrating that modification to flavonoid backbones can be also well manipulated in rice seeds. The flavonoids accumulated as both aglycones and several types of glycosides, and flavonoids in the endosperm were deposited into PB-II-type protein bodies. Therefore, these rice seeds provide an ideal platform for the production of particular flavonoids due to efficient glycosylation, the presence of appropriate organelles for flavonoid accumulation, and the small effect of endogenous enzymes on the production of flavonoids by exogenous enzymes.


Subject(s)
Flavonoids/biosynthesis , Health , Oryza/genetics , Oryza/metabolism , Seeds/metabolism , Apigenin/biosynthesis , Apigenin/chemistry , Biosynthetic Pathways/genetics , Flavanones/biosynthesis , Flavanones/chemistry , Flavonoids/chemistry , Gene Expression Regulation, Plant , Genes, Plant , Genistein/chemistry , Genistein/metabolism , Humans , Kaempferols/biosynthesis , Kaempferols/chemistry , Microscopy, Fluorescence , Plants, Genetically Modified , Seeds/genetics
12.
Microbes Infect ; 14(12): 1086-92, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22683667

ABSTRACT

It has been reported that treatment with tricin (4',5,7-trihydroxy-3',5'-dimethoxyflavone), a derivative of Sasa albo-marginata, after human cytomegalovirus (HCMV) infection significantly suppressed both infectious virus production and HCMV replication in the human embryonic fibroblast cell line MRC-5. In this paper, we examined the mechanisms for the anti-HCMV effects of tricin in MRC-5 cells. Exposure of fibroblasts to tricin inhibited infectious HCMV production, with concomitant decreases in levels of transcripts of the CXC chemokine IFN-inducible T cell alpha chemoattractant (I-TAC or CXCL11) gene. We also found that the transcripts of the HCMV immediate early (IE) gene and replication of HCMV were lower in CXCL11 gene-knockdown cells. These results suggest that tricin is a novel compound with potential anti-HCMV activity and that CXCL11 is one of the chemokines involved in HCMV replication. In addition, it is possible that CXCL11 is the one of the targets of tricin.


Subject(s)
Antiviral Agents/pharmacology , Chemokine CXCL11/metabolism , Cytomegalovirus/drug effects , Flavonoids/pharmacology , Antiviral Agents/isolation & purification , Cell Line , Chemokine CXCL11/genetics , Cytomegalovirus/physiology , Fibroblasts/drug effects , Fibroblasts/virology , Flavonoids/isolation & purification , Gene Expression/drug effects , Gene Expression Profiling , Gene Knockout Techniques , Humans , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Sasa/chemistry , Virus Replication/drug effects
13.
J Virol ; 86(1): 492-503, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22013039

ABSTRACT

Tankyrase 1 is a poly(ADP-ribose) polymerase (PARP) which localizes to multiple subcellular sites, including telomeres and mitotic centrosomes. Poly(ADP-ribosyl)ation of the nuclear mitotic apparatus (NuMA) protein by tankyrase 1 during mitosis is essential for sister telomere resolution and mitotic spindle pole formation. In interphase cells, tankyrase 1 resides in the cytoplasm, and its role therein is not well understood. In this study, we found that herpes simplex virus (HSV) infection induced extensive modification of tankyrase 1 but not tankyrase 2. This modification was dependent on extracellular signal-regulated kinase (ERK) activity triggered by HSV infection. Following HSV-1 infection, tankyrase 1 was recruited to the nucleus. In the early phase of infection, tankyrase 1 colocalized with ICP0 and thereafter localized within the HSV replication compartment, which was blocked in cells infected with the HSV-1 ICP0-null mutant R7910. In the absence of infection, ICP0 interacted with tankyrase 1 and efficiently promoted its nuclear localization. HSV did not replicate efficiently in cells depleted of both tankyrases 1 and 2. Moreover, XAV939, an inhibitor of tankyrase PARP activity, decreased viral titers to 2 to 5% of control values. We concluded that HSV targets tankyrase 1 in an ICP0- and ERK-dependent manner to facilitate its replication.


Subject(s)
Cell Nucleus/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , Herpes Simplex/enzymology , Herpesvirus 1, Human/physiology , Immediate-Early Proteins/metabolism , Tankyrases/metabolism , Ubiquitin-Protein Ligases/metabolism , Virus Replication , Animals , Cell Line , Cell Nucleus/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Herpes Simplex/genetics , Herpes Simplex/virology , Herpesvirus 1, Human/enzymology , Herpesvirus 1, Human/genetics , Humans , Immediate-Early Proteins/genetics , Phosphorylation , Protein Binding , Protein Transport , Tankyrases/genetics , Ubiquitin-Protein Ligases/genetics
14.
Antivir Chem Chemother ; 22(1): 1-11, 2011 Aug 23.
Article in English | MEDLINE | ID: mdl-21860068

ABSTRACT

BACKGROUND: We examined the anti-influenza virus activity of tricin, 4',5,7-trihydroxy-3',5'-dimethoxyflavone, against five viruses: A/Solomon islands/3/2006 (H1N1), A/Hiroshima/52/2005 (H3N2), A/California/07/2009 (H1N1pdm), A/Narita/1/2009 (H1N1pdm) and B/Malaysia/2506/2004 strains in vitro and against A/PR/8/34 virus in vivo. METHODS: The effect of tricin was studied by an infectious virus yield reduction assay. The anti-influenza virus mechanism of the tricin was examined by western blot analysis, real-time reverse transcriptase PCR analysis, haemagglutination inhibition (HI) assay and neuraminidase (NA) inhibition assay. The anti-influenza virus efficacy of tricin was further examined in a murine influenza virus infection model. RESULTS: Tricin of 3.3 to 30 µM significantly reduced seasonal A (H1N1), (H3N2) viruses, novel A (H1N1pdm) virus, as well as B virus in a dose-dependent manner. The 50% effective concentrations of tricin were 3.4 µM for seasonal A (H3N2) virus, 4.9 µM for B virus and 8.2 µM for A/Narita (H1N1pdm) virus. Tricin decreased the expression of haemagglutinin (HA) protein and matrix (M) protein, and messenger RNA expression of HA and M of influenza virus in the infected cells. Tricin exhibited little or no effects on influenza virus HI and NA activities. In the mouse infection model, tricin was significantly effective in reducing body weight loss, and also effective in prolonging survival times of infected mice. CONCLUSIONS: Tricin was indicated to possess anti-influenza virus activity and to ameliorate body weight loss and survival rate of influenza-A-virus-infected mice. Tricin is a novel compound with potential anti-influenza virus activity in vitro and in vivo.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , Flavones/chemistry , Flavones/therapeutic use , Flavonoids/chemistry , Flavonoids/therapeutic use , Influenza A virus/drug effects , Orthomyxoviridae Infections/drug therapy , Animals , Antiviral Agents/pharmacology , Cell Line , Flavones/pharmacology , Flavonoids/pharmacology , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/drug effects , Mice
15.
Antiviral Res ; 91(3): 296-303, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21745500

ABSTRACT

The anti-human cytomegalovirus (HCMV) activity of tricin (4',5,7-trihydroxy-3',5'-dimethoxyflavone), a derivative from Sasa albo-marginata, was studied in the human embryonic fibroblast cell line MRC-5. In a plaque assay, tricin and ganciclovir (GCV) showed concentration-dependent inhibitory properties from 0.05 to 3.6 µM and 0.01 to 1.0 µM, respectively. Tricin had no virucidal effects on cell-free HCMV. Treatment with tricin 1h before, or 1h or 3h after viral infection significantly suppressed HCMV replication. Moreover, tricin inhibited the expression of immediate early (IE) 2 mRNA and DNA polymerase (UL54) mRNA in HCMV-infected cells. Western blot analysis also demonstrated that tricin decreased the expression of IE antigen (especially IE2) and cyclooxygenase 2 (COX-2) expression in HCMV-infected cells. In the presence of tricin, prostaglandin E2 (PGE2) accumulation by HCMV infection was completely inhibited. These results suggest that tricin is a novel compound with potential COX inhibitor-dependent anti-HCMV activity.


Subject(s)
Cytomegalovirus Infections/drug therapy , Cytomegalovirus/drug effects , DNA, Viral/antagonists & inhibitors , Fibroblasts/drug effects , Flavonoids/pharmacology , Ganciclovir/pharmacology , Virus Replication/drug effects , Antiviral Agents/pharmacology , Blotting, Western , Cell Line , Cyclooxygenase 2/biosynthesis , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Cytomegalovirus Infections/virology , DNA, Viral/biosynthesis , DNA-Directed DNA Polymerase/biosynthesis , Dinoprostone/antagonists & inhibitors , Dinoprostone/biosynthesis , Fibroblasts/cytology , Fibroblasts/virology , Humans , Immediate-Early Proteins/antagonists & inhibitors , Immediate-Early Proteins/biosynthesis , Nucleic Acid Synthesis Inhibitors , Reverse Transcriptase Polymerase Chain Reaction , Sasa/chemistry , Virus Replication/genetics
16.
Biol Res ; 43(4): 475-80, 2010.
Article in English | MEDLINE | ID: mdl-21526275

ABSTRACT

Human cytomegalovirus (HCMV) is a herpesvirus associated with serious diseases in immunocompromised subjects. The region between ORF UL133 and UL151 from HCMV, named ULb' is frequently deleted in attenuated AD169 and in highly passaged laboratory strains. However, this region is conserved in low-passaged and more virulent HCMV, like the Toledo strain. The UL146 gene, which is located in the ULb' region, encodes a CXC-chemokine analogue. The diversity of UL146 gene was evaluated among fifty-six clinical isolates of HCMV from Japan. Results show that UL146 gene was successfully amplified by the polymerase chain reaction (PCR) in only 17/56 strains (30%), while the success rate for UL145/UL147 gene was 18/56 strains (32%). After DNA sequencing, the 35 amplified strains were classified into 8 groups. When compared, variability of UL146 ranged from 25.1% to 52.9% at the DNA level and from 34.5% to 67% at the amino acid level. Seven groups had the interleukin-8 (IL-8) motif ERL (Glu-Leu-Arg) CXC and one group had only the CXC motif, suggesting the absence of the IL-8 function of UL146. In conclusion, we found that UL146 gene of HCMV is hyper-variable in clinical strains from Japan suggesting the possibility of a different function in each sequence group.


Subject(s)
Chemokines, CXC/genetics , Cytomegalovirus Infections/virology , Cytomegalovirus/genetics , Genes, Viral/genetics , Genetic Variation/genetics , Viral Proteins/genetics , Base Sequence , Cytomegalovirus/isolation & purification , Fibroblasts/virology , Genotype , Humans , Japan , Molecular Sequence Data , Polymerase Chain Reaction , Sequence Analysis, DNA
17.
Biol. Res ; 43(4): 475-480, 2010. ilus, tab
Article in English | LILACS | ID: lil-582863

ABSTRACT

Human Cytomegalovirus (HCMV) is a herpesvirus associated with serious diseases in immunocompromised subjects. The region between ORF UL133 and UL151 from HCMV, named ULb' is frequently deleted in attenuated AD169 and in highly passaged laboratory strains. However, this region is conserved in low-passaged and more virulent HCMV, like the Toledo strain. The UL146 gene, which is located in the ULb' region, encodes a CXC-chemokine analogue. The diversity of UL146 gene was evaluated among fifty-six clinical isolates of HCMV from Japan. Results show that UL146 gene was successfully amplified by the polymerase chain reaction (PCR) in only 17/56 strains (30 percent), while the success rate for UL145/UL147 gene was 18/56 strains (32 percent). After DNA sequencing, the 35 amplified strains were classified into 8 groups. When compared, variability of UL146 ranged from 25.1 percent to 52.9 percent at the DNA level and from 34.5 percent to 67 percent at the amino acid level. Seven groups had the interleukin-8 (IL-8) motif ERL (Glu-Leu-Arg) CXC and one group had only the CXC motif, suggesting the absence of the IL-8 function of UL146. In conclusion, we found that UL146 gene of HCMV is hyper-variable in clinical strains from Japan suggesting the possibility of a different function in each sequence group.


Subject(s)
Humans , Chemokines, CXC/genetics , Cytomegalovirus Infections/virology , Cytomegalovirus/genetics , Genes, Viral/genetics , Genetic Variation/genetics , Viral Proteins/genetics , Base Sequence , Cytomegalovirus/isolation & purification , Fibroblasts/virology , Genotype , Japan , Molecular Sequence Data , Polymerase Chain Reaction , Sequence Analysis, DNA
18.
Microbes Infect ; 11(13): 1029-36, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19651232

ABSTRACT

Human antibodies specific for HCMV are currently considered as potential anti-HCMV therapeutic agents. In this study, we used a combinatorial human antibody library to isolate and characterize complete human monoclonal antibodies that effectively neutralize HCMV in a complement-dependent manner. One hundred and six clones were isolated in two independent screens using HCMV virions and recombinant glycoprotein B, gB654, as antigens. All of the clones recognized the same molecule gB and were classified into 14 groups based on the amino acid sequence of the V(H) region. Seven representative clones from these 14 groups had a strong gB654 binding affinity by surface plasmon resonance (SPR). A pairwise binding competition analysis suggested that there were three groups based on differences in the gB recognition sites. Although Fab fragments of the seven groups showed strong affinity for gB, none of the Fab fragments neutralized HCMV infectivity in vitro. In contrast, complete human IgG(1) antibodies of at least three groups neutralized HCMV in a complement-dependent manner. These data suggest that potent therapeutic antibodies can be obtained from a human antibody library, including most of the functional antibodies that mediate humoral immunity to the selected pathogen.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/metabolism , Viral Envelope Proteins/metabolism , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antibodies, Viral/metabolism , Antigens, Viral/metabolism , Binding, Competitive , Cell Line , Complement System Proteins/metabolism , Enzyme-Linked Immunosorbent Assay , Epitopes , Humans , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin G/metabolism , Peptide Library , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Surface Plasmon Resonance , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
19.
Virus Res ; 142(1-2): 68-77, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19201384

ABSTRACT

Proteasome inhibitor, which inhibits NF-kappaB activation, has been reported to activate c-Jun N-terminal kinase (JNK)-c-Jun pathway. In this study, we investigated the effects of proteasome inhibitor on the human cytomegalovirus (HCMV) major immediate early (MIE) gene expression in human central nervous system (CNS)-derived cell lines. Treatment of HCMV-infected 118MGC glioma and U373-MG astrocytoma cells with three proteasome inhibitors, MG132, clasto-lactacystin beta-lactone, and epoxomicin, suppressed MIE protein expression. In contrast, in HCMV-infected IMR-32 neuroblastoma cells, the proteasome inhibitors increased MIE protein expression, even in the presence of NF-kappaB inhibitor SN-50. A luciferase reporter assay demonstrated that MG132 markedly elevated the MIE promoter/enhancer (MIEP) activity in IMR-32 cells, but down-regulated it in 118MGC and U373-MG cells. Mutation in five cAMP response elements (CREs) within the MIEP resulted in a loss of the ability to respond to MG132 in IMR-32 cells. Moreover, Western blotting analysis revealed that MG132 induced c-Jun phosphorylation in all three CNS-derived cell lines, whereas a high level of activating transcription factor-2 (ATF-2) phosphorylation was observed only in IMR-32 cells. Finally, MG132-induced MIE protein expression was suppressed by JNK inhibitor that reduced the phosphorylation levels of both c-Jun and ATF-2. Taken together, these results suggest that the proteasome inhibitors activate CRE binding proteins consisting of c-Jun and ATF-2 through activating the JNK-c-Jun pathway, thereby inducing MIE protein synthesis in IMR-32 cells under the condition where NF-kappaB activity is inhibited.


Subject(s)
Central Nervous System/virology , Cytomegalovirus Infections/virology , Cytomegalovirus/drug effects , Gene Expression Regulation, Viral/drug effects , Immediate-Early Proteins/genetics , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Activating Transcription Factor 2/genetics , Activating Transcription Factor 2/metabolism , Cell Line , Central Nervous System/metabolism , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/metabolism , Humans , Immediate-Early Proteins/metabolism , Lactones/pharmacology , Leupeptins/pharmacology , Oligopeptides/pharmacology , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism
20.
Antivir Chem Chemother ; 18(3): 133-9, 2007.
Article in English | MEDLINE | ID: mdl-17626597

ABSTRACT

We examined the anticytomegalovirus properties of four compounds: pristimerin, the pristimerin analogue, lupeol and 2-acetylphenol-1-beta-D-glucopyranosyl (1 --> 6)-beta-D-xylpyranoside (acetophenol glycoside), isolated from Maytenus heterophylla, a Kenyan medicinal plant. The effects were studied on human cytomegalovirus (HCMV) replication in the human embryonic fibroblast cell line, MRC-5. In a viral plaque-reduction assay, pristimerin showed dose-dependent inhibitory properties with a 50% inhibitory concentration of 0.53 microg/ml (selective index = 27.9). The cells treated with pristimerin inhibited the cytopathic effects in HCMV-infected cells. Moreover, pristimerin suppressed viral replication without affecting the cell growth. Pristimerin inhibited the synthesis of viral DNA but had no virucidal effect on cell-free HCMV. Furthermore, Western blot analysis demonstrated that pristimerin decreased the amount of immediate early (IE) antigen (especially IE2) expression in the infected cells. These results suggest that pristimerin is a unique compound with potential anti-HCMV activity.


Subject(s)
Antiviral Agents/pharmacology , Cytomegalovirus/drug effects , Triterpenes/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/isolation & purification , Blotting, Western , Cell Line , Cytopathogenic Effect, Viral/drug effects , DNA, Viral/biosynthesis , Fibroblasts/drug effects , Humans , Immediate-Early Proteins/biosynthesis , Inhibitory Concentration 50 , Maytenus/chemistry , Pentacyclic Triterpenes , Phenols/isolation & purification , Phenols/pharmacology , Trans-Activators/biosynthesis , Triterpenes/chemistry , Triterpenes/isolation & purification , Triterpenes/toxicity , Viral Plaque Assay , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL