Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 337
Filter
1.
Clin Lab ; 67(2)2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33616332

ABSTRACT

BACKGROUND AND METHODS: 2019 Corona Virus Disease (COVID-19) caused by SARS-CoV-2 is still pandemic now. RT-qPCR detection was the most common method for the diagnosis of SARS-CoV-2 infection, facilitated by amounts of nucleic acid testing kits. However, the accuracy of nucleic acid detection is affected by various factors such as specimen collection, specimen preparation, reagents deficiency, and personnel quality. RESULTS: In this study, we found that unmatched virus preservation solution will inhibit N gene and OFR-1ab gene (two independent genes of SARS-CoV-2) amplification in one-step detection reagent. CONCLUSIONS: Despite just being a particular phenomenon we found in our work to fight 2019-nCoV, we concluded that unmatched virus preservation solution may have an inhibitory effect on SARS-CoV-2 nucleic acid detection which may lead to incorrect clinical diagnosis.


Subject(s)
COVID-19 Nucleic Acid Testing/methods , COVID-19 , Genes, Viral/drug effects , Organ Preservation Solutions/pharmacology , SARS-CoV-2 , Specimen Handling , COVID-19/diagnosis , COVID-19/virology , Diagnostic Errors/prevention & control , Humans , Reagent Kits, Diagnostic/standards , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , Specimen Handling/adverse effects , Specimen Handling/methods
2.
Drug Dev Ind Pharm ; 46(8): 1345-1353, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32643448

ABSTRACT

PURPOSE: Huashi Baidu formula (HSBDF) was developed to treat the patients with severe COVID-19 in China. The purpose of this study was to explore its active compounds and demonstrate its mechanisms against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) through network pharmacology and molecular docking. METHODS: All the components of HSBDF were retrieved from the pharmacology database of TCM system. The genes corresponding to the targets were retrieved using UniProt and GeneCards database. The herb-compound-target network was constructed by Cytoscape. The target protein-protein interaction network was built using STRING database. The core targets of HSBDF were analyzed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). The main active compounds of HSBDF were docked with SARS-CoV-2 and angiotensin converting enzyme II (ACE2). RESULTS: Compound-target network mainly contained 178 compounds and 272 corresponding targets. Key targets contained MAPK3, MAPK8, TP53, CASP3, IL6, TNF, MAPK1, CCL2, PTGS2, etc. There were 522 GO items in GO enrichment analysis (p < .05) and 168 signaling pathways (p < .05) in KEGG, mainly including TNF signaling pathway, PI3K-Akt signaling pathway, NOD-like receptor signaling pathway, MAPK signaling pathway, and HIF-1 signaling pathway. The results of molecular docking showed that baicalein and quercetin were the top two compounds of HSBDF, which had high affinity with ACE2. CONCLUSION: Baicalein and quercetin in HSBDF may regulate multiple signaling pathways through ACE2, which might play a therapeutic role on COVID-19.


Subject(s)
Betacoronavirus/drug effects , Coronavirus Infections/drug therapy , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Molecular Docking Simulation/methods , Pharmacology, Clinical/methods , Pneumonia, Viral/drug therapy , Angiotensin-Converting Enzyme 2 , Betacoronavirus/chemistry , Betacoronavirus/genetics , COVID-19 , China , Databases, Factual , Gene Ontology , Gene Targeting , Genes, Viral/drug effects , Genes, Viral/genetics , Humans , Medicine, Chinese Traditional , Pandemics , Peptidyl-Dipeptidase A/drug effects , Peptidyl-Dipeptidase A/genetics , SARS-CoV-2 , Signal Transduction/drug effects , Signal Transduction/genetics , COVID-19 Drug Treatment
3.
Mar Biotechnol (NY) ; 21(4): 503-514, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31111339

ABSTRACT

Although alginate is known as an immunostimulant in shrimp, the comprehensive and simultaneous study on its activity to resolve the relationship of the hematological parameters, upregulation of immune-related gene expression, and resistance to pathogen has not been found in shrimp. We performed experiments to evaluate the effect and mechanism of alginate from S. siliquosum on Pacific white shrimp immune system. Hematological parameters were examined after oral administration of Na alginate in the shrimp. White spot syndrome virus (WSSV) was injected to the shrimp at 14 days, and its copy number was examined quantitatively (qRT-PCR). Immune-related gene expression was evaluated by qRT-PCR. Alginate increased some hematological immune parameters of shrimp. Before WSSV infection, expression levels of Toll and lectin genes were upregulated. The lectin gene were upregulated post infection, and the Toll gene in all the treatments were downregulated, except the shrimps fed with alginate at 6.0 g kg-1 at 48 h post infection (hpi). The shrimps fed with alginate at 6.0 g kg-1 were the most resistant and gave the least WSSV copy number at 48 hpi. Resistance of shrimps fed the alginate-supplemented diets against WSSV was significantly higher compared to that of the control treatment with 56% and 10% of survival rates, respectively. Oral administration of alginate did not affect the growth and total protein plasma. At 120 h post challenge, alginate treatment at 6.0 g kg-1 exhibited the highest survival rate. It is concluded that oral administration of alginate enhanced the innate immunity by upregulating immune-related gene expression. Consequently, the enhancement of the shrimp innate immunity improves the resistance against WSSV infection.


Subject(s)
Alginates/administration & dosage , Disease Resistance/drug effects , Immunity, Innate/drug effects , Penaeidae/drug effects , Sargassum/chemistry , White spot syndrome virus 1/drug effects , Administration, Oral , Alginates/isolation & purification , Animals , Arthropod Proteins/genetics , Arthropod Proteins/immunology , Disease Resistance/genetics , Gene Dosage , Gene Expression Regulation , Genes, Viral/drug effects , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Lectins/genetics , Lectins/immunology , Penaeidae/genetics , Penaeidae/immunology , Penaeidae/virology , Signal Transduction , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , White spot syndrome virus 1/genetics , White spot syndrome virus 1/metabolism
4.
Antiviral Res ; 140: 142-150, 2017 04.
Article in English | MEDLINE | ID: mdl-28161581

ABSTRACT

Previously we established reporter cell lines for human cytomegalovirus (HCMV) and varicella zoster virus (VZV) and identified several antiviral compounds against these viruses using the reporter cells. In this study, we found that one of the identified anti-HCMV compounds, a thienylcarboxamide derivative (coded as 133G4), was effective against not only HCMV but also VZV. The following findings indicate that 133G4 inhibits the activation of early gene promoters by HCMV IE2 and VZV IE62: i) 133G4 decreased the expression of HCMV early and late genes but not that of HCMV IE1/IE2 in HCMV-infected cells, ii) 133G4 inhibited the activation of several HCMV early gene promoters of transiently-transfected plasmids in HCMV-infected cells, and iii) in transient transfection assays, 133G4 decreased the activation of HCMV (or VZV) early gene promoters by HCMV IE2 (or VZV IE62) in the absence of other viral protein expression. The inhibition of early gene activation was observed in the human and African green monkey cell lines but not in the rodent cell lines, and the compound was not effective against murine CMV. In addition, VZV IE62 activated HCMV early promoters, and 133G4 still inhibited such promoter activation. Therefore, we hypothesized that 133G4 targets a cellular factor used commonly in activation of human herpesvirus promoters and examined whether 133G4 affects the functions of cellular proteins USF1, TBP, Med25 and EAP, the involvement of which in VZV IE62-dependent viral gene activation has been well characterized. Our experimental results using one-hybrid and bimolecular fluorescence complementation assays demonstrated that 133G4 did not inhibit the recruitment of USF1 or TBP to their binding sites, nor inhibited the direct interactions of VZV IE62 with Med25 and EAP. Thus, 133G4 is a unique anti-VZV and -HCMV compound, which warrants further studies to find out its inhibitory mechanism.


Subject(s)
Anilides/pharmacology , Antiviral Agents/pharmacology , Cytomegalovirus/genetics , Herpesvirus 3, Human/genetics , Thiophenes/pharmacology , Transcriptional Activation/drug effects , Anilides/chemistry , Animals , Antiviral Agents/chemistry , Antiviral Agents/isolation & purification , Binding Sites , Cell Line , Chlorocebus aethiops , Cytomegalovirus/drug effects , Cytomegalovirus/physiology , Genes, Immediate-Early/drug effects , Genes, Viral/drug effects , Herpesvirus 3, Human/drug effects , Herpesvirus 3, Human/physiology , Humans , Mice , NIH 3T3 Cells , Promoter Regions, Genetic/drug effects , Protein Binding , Thiophenes/chemistry , Viral Proteins/drug effects , Viral Proteins/genetics
5.
J Virol ; 90(23): 10715-10727, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27654292

ABSTRACT

Cytomegalovirus (CMV) is a ubiquitous human pathogen that increases the morbidity and mortality of immunocompromised individuals. The current FDA-approved treatments for CMV infection are intended to be virus specific, yet they have significant adverse side effects, including nephrotoxicity and hematological toxicity. Thus, there is a medical need for safer and more effective CMV therapeutics. Using a high-content screen, we identified the cardiac glycoside convallatoxin as an effective compound that inhibits CMV infection. Using a panel of cardiac glycoside variants, we assessed the structural elements critical for anti-CMV activity by both experimental and in silico methods. Analysis of the antiviral effects, toxicities, and pharmacodynamics of different variants of cardiac glycosides identified the mechanism of inhibition as reduction of methionine import, leading to decreased immediate-early gene translation without significant toxicity. Also, convallatoxin was found to dramatically reduce the proliferation of clinical CMV strains, implying that its mechanism of action is an effective strategy to block CMV dissemination. Our study has uncovered the mechanism and structural elements of convallatoxin, which are important for effectively inhibiting CMV infection by targeting the expression of immediate-early genes. IMPORTANCE: Cytomegalovirus is a highly prevalent virus capable of causing severe disease in certain populations. The current FDA-approved therapeutics all target the same stage of the viral life cycle and induce toxicity and viral resistance. We identified convallatoxin, a novel cell-targeting antiviral that inhibits CMV infection by decreasing the synthesis of viral proteins. At doses low enough for cells to tolerate, convallatoxin was able to inhibit primary isolates of CMV, including those resistant to the anti-CMV drug ganciclovir. In addition to identifying convallatoxin as a novel antiviral, limiting mRNA translation has a dramatic impact on CMV infection and proliferation.


Subject(s)
Antiviral Agents/pharmacology , Cytomegalovirus Infections/prevention & control , Cytomegalovirus/drug effects , Methionine/metabolism , Strophanthins/pharmacology , Antiviral Agents/chemistry , Biological Transport, Active/drug effects , Cardiac Glycosides/chemistry , Cardiac Glycosides/pharmacology , Cell Line , Cytomegalovirus/genetics , Cytomegalovirus/physiology , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/virology , Genes, Immediate-Early/drug effects , Genes, Viral/drug effects , Humans , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Strophanthins/chemistry , Structure-Activity Relationship , Virus Replication/drug effects
6.
Proc Natl Acad Sci U S A ; 113(13): 3609-14, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26976570

ABSTRACT

Clinically available drugs active against Epstein-Barr virus (EBV) and other human herpesviruses are limited to those targeting viral DNA replication. To identify compounds directed against other steps in the viral life cycle, we searched for drugs active against the EBV SM protein, which is essential for infectious virus production. SM has a highly gene-specific mode of action and preferentially enhances expression of several late lytic cycle EBV genes. Here we demonstrate that spironolactone, a mineralocorticoid receptor antagonist approved for clinical use, inhibits SM function and infectious EBV production. Expression of EBV viral capsid antigen is highly SM dependent, and spironolactone inhibits viral capsid antigen synthesis and capsid formation, blocking EBV virion production at a step subsequent to viral DNA replication. In addition, spironolactone inhibits expression of other SM-dependent genes necessary for infectious virion formation. We further demonstrate that molecules structurally related to spironolactone with similar antimineralocorticoid blocking activity do not inhibit EBV production. These findings pave the way for development of antiherpesvirus drugs with new mechanisms of action directed against SM and homologous essential proteins in other herpesviruses.


Subject(s)
Antiviral Agents/pharmacology , Herpesvirus 4, Human/drug effects , Herpesvirus 4, Human/physiology , Immediate-Early Proteins/antagonists & inhibitors , Spironolactone/pharmacology , Trans-Activators/antagonists & inhibitors , Capsid Proteins/antagonists & inhibitors , Capsid Proteins/genetics , Capsid Proteins/physiology , Cell Line , DNA Replication/drug effects , Gene Deletion , Gene Expression Regulation, Viral/drug effects , Gene Knockout Techniques , Genes, Viral/drug effects , HEK293 Cells , Herpesvirus 4, Human/genetics , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/physiology , Mineralocorticoid Receptor Antagonists/pharmacology , Spironolactone/analogs & derivatives , Spironolactone/chemistry , Structure-Activity Relationship , Trans-Activators/genetics , Trans-Activators/physiology , Transcriptome/drug effects , Virus Release/drug effects , Virus Replication/drug effects , Virus Replication/genetics , Virus Replication/physiology
7.
Cell Physiol Biochem ; 37(3): 853-65, 2015.
Article in English | MEDLINE | ID: mdl-26383101

ABSTRACT

BACKGROUND/AIMS: The ability of human immunodeficiency virus-1(HIV-1) to establish latent infection and its re-activation is considered critical for progression of HIV-1 infection. We previously reported that a bacterial metabolite butyric acid, acting as a potent inhibitor of histone deacetylases (HDACs), could lead to induction of HIV-1 transcription; however, the molecular mechanism remains unclear. The aim of this study was to investigate the effect of butyric acid on HIV-1 gene expression. METHODS: Butyric acid-mediated HIV-1 gene expression was determined by luciferase assay and Chromatin immunoprecipitation assay. Western blot analysis and ELISA were used for the detection of HIV-1. RESULTS: We found that Sp1 binding sites within the HIV-1 promoter are primarily involved in butyric acid-mediated HIV-1 activation. In fact, Sp1 knockdown by small interfering RNA and the Sp1 inhibitor mithramycin A abolished the effect of butyric acid. We also observed that cAMP response element-binding-binding protein (CBP) was required for butyric acid-induced HIV-1 activation. CONCLUSIONS: These results suggest that butyric acid stimulates HIV-1 promoter through inhibition of the Sp1-associated HDAC activity and recruitment of CBP to the HIV-1 LTR. Our findings suggest that Sp1 should be considered as one of therapeutic targets in anti-viral therapy against HIV-1 infection aggravated by butyric acid-producing bacteria.


Subject(s)
Butyric Acid/pharmacology , Genes, Viral/drug effects , HIV-1/genetics , Sp1 Transcription Factor/metabolism , Cell Line , Gene Expression Regulation, Viral/drug effects , HIV-1/drug effects , HeLa Cells , Histone Deacetylase Inhibitors/pharmacology , Humans , Plicamycin/analogs & derivatives , Plicamycin/pharmacology , Promoter Regions, Genetic/drug effects , RNA, Viral/genetics , RNA, Viral/metabolism , Transcriptional Activation/drug effects
8.
Comput Math Methods Med ; 2015: 395826, 2015.
Article in English | MEDLINE | ID: mdl-26170895

ABSTRACT

A variety of enzyme inhibitors have been developed in combating HIV-1, however the fast evolutionary rate of this virus commonly leads to the emergence of resistance mutations that finally allows the mutant virus to survive. This review explores the main genetic consequences of HIV-1 molecular evolution during antiviral therapies, including the viral genetic diversity and molecular adaptation. The role of recombination in the generation of drug resistance is also analyzed. Besides the investigation and discussion of published works, an evolutionary analysis of protease-coding genes collected from patients before and after treatment with different protease inhibitors was included to validate previous studies. Finally, the review discusses the importance of considering genetic consequences of antiviral therapies in models of HIV-1 evolution that could improve current genotypic resistance testing and treatments design.


Subject(s)
Acquired Immunodeficiency Syndrome/drug therapy , Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV-1/drug effects , HIV-1/genetics , Mutation , Antiretroviral Therapy, Highly Active , Computational Biology , Computer Simulation , DNA, Viral/genetics , Drug Resistance, Viral , Evolution, Molecular , Genes, Viral/drug effects , Genetic Variation , Genotype , Humans , Peptide Hydrolases/genetics , Recombination, Genetic , Virus Replication
9.
Exp Biol Med (Maywood) ; 240(12): 1630-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26080461

ABSTRACT

MicroRNAs (miRNAs) play an important role in the regulation of gene expression and are involved in many cellular processes including inhibition of viral replication in infected cells. In this study, three subtypes of influenza A viruses (pH1N1, H5N1 and H3N2) were analyzed to identify candidate human miRNAs targeting and silencing viral genes expression. Candidate human miRNAs were predicted by miRBase and RNAhybrid based on minimum free energy (MFE) and hybridization patterns between human miRNAs and viral target genes. In silico analysis presented 76 miRNAs targeting influenza A viruses, including 70 miRNAs that targeted specific subtypes (21 for pH1N1, 27 for H5N1 and 22 for H3N2) and 6 miRNAs (miR-216b, miR-3145, miR-3682, miR-4513, miR-4753 and miR-5693) that targeted multiple subtypes of influenza A viruses. Interestingly, miR-3145 is the only candidate miRNA targeting all three subtypes of influenza A viruses. The miR-3145 targets to PB1 encoding polymerase basic protein 1, which is the main component of the viral polymerase complex. The silencing effect of miR-3145 was validated by 3'-UTR reporter assay and inhibition of influenza viral replication in A549 cells. In 3'-UTR reporter assay, results revealed that miR-3145 triggered significant reduction of the luciferase activity. Moreover, expression of viral PB1 genes was also inhibited considerably (P value < 0.05) in viral infected cells expressing mimic miR-3145. In conclusion, this study demonstrated that human miR-3145 triggered silencing of viral PB1 genes and lead to inhibition of multiple subtypes of influenza viral replication. Therefore, hsa-miR-3145 might be useful for alternative treatment of influenza A viruses in the future.


Subject(s)
Alphainfluenzavirus/drug effects , Antiviral Agents/pharmacology , MicroRNAs/pharmacology , Viral Proteins/antagonists & inhibitors , Virus Replication/drug effects , Gene Silencing/drug effects , Genes, Viral/drug effects , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/physiology , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/physiology , Alphainfluenzavirus/genetics , Alphainfluenzavirus/physiology , Real-Time Polymerase Chain Reaction , Viral Proteins/genetics , Virus Replication/genetics
10.
Nature ; 499(7457): 219-22, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23748443

ABSTRACT

The mammalian gut ecosystem has considerable influence on host physiology, but the mechanisms that sustain this complex environment in the face of different stresses remain obscure. Perturbations to the gut ecosystem, such as through antibiotic treatment or diet, are at present interpreted at the level of bacterial phylogeny. Less is known about the contributions of the abundant population of phages to this ecological network. Here we explore the phageome as a potential genetic reservoir for bacterial adaptation by sequencing murine faecal phage populations following antibiotic perturbation. We show that antibiotic treatment leads to the enrichment of phage-encoded genes that confer resistance via disparate mechanisms to the administered drug, as well as genes that confer resistance to antibiotics unrelated to the administered drug, and we demonstrate experimentally that phages from treated mice provide aerobically cultured naive microbiota with increased resistance. Systems-wide analyses uncovered post-treatment phage-encoded processes related to host colonization and growth adaptation, indicating that the phageome becomes broadly enriched for functionally beneficial genes under stress-related conditions. We also show that antibiotic treatment expands the interactions between phage and bacterial species, leading to a more highly connected phage-bacterial network for gene exchange. Our work implicates the phageome in the emergence of multidrug resistance, and indicates that the adaptive capacity of the phageome may represent a community-based mechanism for protecting the gut microflora, preserving its functional robustness during antibiotic stress.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteriophages/drug effects , Drug Resistance, Microbial/drug effects , Feces/microbiology , Feces/virology , Genome, Viral/genetics , Metagenome/genetics , Aerobiosis , Ampicillin/pharmacology , Animals , Bacteriophages/genetics , Bacteriophages/isolation & purification , Ciprofloxacin/pharmacology , Drug Resistance, Microbial/genetics , Female , Gene Transfer, Horizontal/drug effects , Gene Transfer, Horizontal/genetics , Genes, Viral/drug effects , Genes, Viral/genetics , Host Specificity/drug effects , Metagenome/drug effects , Mice , Symbiosis/drug effects , Symbiosis/genetics
11.
Cancer Invest ; 30(4): 268-74, 2012 May.
Article in English | MEDLINE | ID: mdl-22480174

ABSTRACT

Human herpesvirus 8 (HHV8) is the etiologic agent for primary effusion lymphoma (PEL). The aim of this study is to investigate the effects of cisplatin on the PEL cells. Cisplatin treatment induced apoptosis and inhibited the growth of PEL cells, and the effect was more profound in the HHV8-positive lymphoma cells compared with the EBV-positive lymphoma cells. Cisplatin treatment decreased the expression of HHV8 latent genes and activated p53 at serine 15 in PEL cells. Our results indicate that cisplatin can disrupt HHV8 latency and induce reactivation of p53 and highly selective treatment modality for this virally induced lymphoma.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Herpesvirus 8, Human/drug effects , Lymphoma, Primary Effusion/virology , Virus Latency/drug effects , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Separation , Flow Cytometry , Gene Expression/drug effects , Genes, Viral/drug effects , Herpesvirus 8, Human/physiology , Humans , Lymphoma, Primary Effusion/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/metabolism
13.
PLoS Pathog ; 7(8): e1002184, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21876670

ABSTRACT

The essential transactivator function of the HIV Tat protein is regulated by multiple posttranslational modifications. Although individual modifications are well characterized, their crosstalk and dynamics of occurrence during the HIV transcription cycle remain unclear.We examine interactions between two critical modifications within the RNA-binding domain of Tat: monomethylation of lysine 51 (K51) mediated by Set7/9/KMT7, an early event in the Tat transactivation cycle that strengthens the interaction of Tat with TAR RNA, and acetylation of lysine 50 (K50) mediated by p300/KAT3B, a later process that dissociates the complex formed by Tat, TAR RNA and the cyclin T1 subunit of the positive transcription elongation factor b (P-TEFb). We find K51 monomethylation inhibited in synthetic Tat peptides carrying an acetyl group at K50 while acetylation can occur in methylated peptides, albeit at a reduced rate. To examine whether Tat is subject to sequential monomethylation and acetylation in cells, we performed mass spectrometry on immunoprecipitated Tat proteins and generated new modification-specific Tat antibodies against monomethylated/acetylated Tat. No bimodified Tat protein was detected in cells pointing to a demethylation step during the Tat transactivation cycle. We identify lysine-specific demethylase 1 (LSD1/KDM1) as a Tat K51-specific demethylase, which is required for the activation of HIV transcription in latently infected T cells. LSD1/KDM1 and its cofactor CoREST associates with the HIV promoter in vivo and activate Tat transcriptional activity in a K51-dependent manner. In addition, small hairpin RNAs directed against LSD1/KDM1 or inhibition of its activity with the monoamine oxidase inhibitor phenelzine suppresses the activation of HIV transcription in latently infected T cells.Our data support the model that a LSD1/KDM1/CoREST complex, normally known as a transcriptional suppressor, acts as a novel activator of HIV transcription through demethylation of K51 in Tat. Small molecule inhibitors of LSD1/KDM1 show therapeutic promise by enforcing HIV latency in infected T cells.


Subject(s)
Histone Demethylases/metabolism , Transcription, Genetic/drug effects , tat Gene Products, Human Immunodeficiency Virus/metabolism , Acetylation , Animals , Epigenesis, Genetic/physiology , Genes, Viral/drug effects , Histone Demethylases/antagonists & inhibitors , Methylation , Phenelzine/pharmacology , Positive Transcriptional Elongation Factor B/metabolism , Rabbits , tat Gene Products, Human Immunodeficiency Virus/immunology
14.
Mol Pharm ; 8(5): 1652-61, 2011 Oct 03.
Article in English | MEDLINE | ID: mdl-21780831

ABSTRACT

Adjuvants modulate protective CD8(+) T cell responses generated by cancer vaccines. We have previously shown that immunostimulatory cytosine-phosphodiester-guanine (CpG) oligodeoxynucleotide (ODN) significantly augments tumor protection in mice given adenovirus cancer vaccines. Here, we examined the impact of chitosan, another candidate vaccine adjuvant, on protection conferred by adenovirus cancer vaccines. Unexpectedly, immunization of mice with adenovirus cancer vaccines in combination with chitosan provided little protection against tumor challenge. This directly correlated with the reduced detection of Ag-specific CD8(+) T cells, interferon-γ (IFN-γ) production, and cytotoxic T cell activity. We ruled out immunosuppressive regulatory T cells since the frequency did not change regardless of whether chitosan was delivered. In mammalian cell lines, chitosan did not interfere with adenovirus transgene expression. However, infection of primary murine bone marrow-derived dendritic cells with adenovirus complexed with chitosan significantly reduced viability, transgene expression, and upregulation of major histocompatability (MHC) class I and CD86. Our in vitro observations indicate that chitosan dramatically inhibits adenovirus-mediated transgene expression and antigen presenting cell activation, which could prevent CD8(+) T cell activation from occurring in vivo. These surprising data demonstrate for the first time that chitosan vaccine formulations can negatively impact the induction of CD8(+) T cell responses via its effect on dendritic cells, which is clinically important since consideration of chitosan as an adjuvant for vaccine formulations is growing.


Subject(s)
Adenoviridae/immunology , Cancer Vaccines/antagonists & inhibitors , Chitosan/toxicity , Down-Regulation/drug effects , Immunologic Factors/toxicity , T-Lymphocytes, Cytotoxic/drug effects , Adenoviridae/genetics , Animals , Antigen Presentation/drug effects , B7-2 Antigen/metabolism , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/virology , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/virology , Genes, Viral/drug effects , Histocompatibility Antigens Class I/metabolism , Interferon-gamma Release Tests , Lymphocyte Activation/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology , Transgenes/drug effects
15.
Antivir Ther ; 16(1): 17-26, 2011.
Article in English | MEDLINE | ID: mdl-21311105

ABSTRACT

BACKGROUND: Alloferon, an immunomodulatory peptide, has antiviral capability against herpesvirus. In this research, we aimed to investigate the effect of alloferon on the regulation of the life cycle of Kaposi's sarcoma-associated herpesvirus (KSHV), and its mechanisms. We also assessed the antiviral activity of alloferon on natural killer (NK) cells as an early antiviral immune responder. METHODS: We first examined the change in cell proliferation and the expression of the viral genes in a KSHV-infected cell line, body-cavity-based B lymphoma (BCBL)-1, under the lytic cycle by 12-O-tetradecanoyl-phorbol-13-acetate (TPA) treatment. To elucidate the antiviral mechanism of alloferon, we tested calcium influx and the activation of the extracellular signal-regulated kinase (ERK) pathway. Furthermore, we evaluated the cytotoxicity of NK cells against BCBL-1 by alloferon. RESULTS: Alloferon effectively recovered the suppressed proliferation of BCBL-1 by TPA, which was achieved by the down-regulation of lytic-cycle-related viral genes, RTA, K8 and vIRF2. To clarify the signal transduction pathways related to the regulation of the viral genes by alloferon, we confirmed that the calcium influx into BCBL-1 was apparently inhibited by alloferon, which preceded the suppression of the phosphorylation of ERK and the activation of AP-1 by TPA. Moreover, when NK cells were exposed to alloferon, their cytolytic activity was improved, and this was mediated by the enhancement of perforin/granzyme secretion. CONCLUSIONS: The results of this study suggest that alloferon can be used as an effective antiviral agent for the regulation of the KSHV life cycle by the down-regulation of AP-1 activity and for the the enhancement of antiviral immunity by up-regulation of NK cell cytotoxicity.


Subject(s)
Antiviral Agents/pharmacology , Herpesviridae Infections/metabolism , Herpesvirus 8, Human/metabolism , Peptides/pharmacology , Antiviral Agents/immunology , Calcium/antagonists & inhibitors , Calcium/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression/drug effects , Genes, Viral/drug effects , Genes, Viral/immunology , Granzymes/metabolism , Herpesviridae Infections/genetics , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/immunology , Humans , Immunomodulation/drug effects , Immunomodulation/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Peptides/immunology , Perforin/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects , Signal Transduction/immunology , Tetradecanoylphorbol Acetate/immunology , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factor AP-1/antagonists & inhibitors , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Up-Regulation , Virus Activation/genetics
16.
Molecules ; 15(9): 6127-39, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20877211

ABSTRACT

Epstein-Barr virus (EBV)-encoded latent membrane protein 1 (LMP1) has been known to have oncogenic properties during latent infection in nasopharyngeal carcinoma (NPC). Genetic manipulation of LMP1 expression may provide a novel strategy for the treatment of NPC. DNAzymes are synthetic, single-stranded DNA catalysts that can be engineered to bind and cleave the target mRNA of a disease-causing gene. By targeting the LMP1 mRNA, we successfully obtained a phosphorothioate-modified ''10-23'' DNAzyme namely DZ1, through screening a series of DNAzymes. DZ1 could significantly down-regulate the expression of LMP1 in NPC cells, inhibit cell proliferation, metastasis, promote apoptosis and enhance radiosensitivity of NPC through interfering signal pathways which are abnormally activated by LMP1, including NF-κB, AP-1 and STAT3 signal pathways. Together, interfering LMP1 signaling pathway could be a promising strategy to target the malignant phenotypes of NPC.


Subject(s)
DNA, Catalytic/pharmacology , Genes, Viral/drug effects , Herpesvirus 4, Human/drug effects , Nasopharyngeal Neoplasms/drug therapy , Viral Matrix Proteins/antagonists & inhibitors , DNA, Catalytic/chemistry , DNA, Catalytic/therapeutic use , Nasopharyngeal Neoplasms/virology , Signal Transduction/drug effects , Viral Matrix Proteins/drug effects , Viral Matrix Proteins/genetics
17.
J Acquir Immune Defic Syndr ; 54(2): 160-6, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20130473

ABSTRACT

OBJECTIVES: To determine the frequency of mutations in the connection domain (CD) of HIV reverse transcriptase in treatment-experienced patients in the Options in Management with Antiretrovirals trial, their impact on susceptibility to antiretroviral (ARV) drugs, and their impact on virologic outcomes. METHODS: Baseline plasma ARV genotypes and inferred resistance phenotypes were obtained. Frequencies of E312Q, Y318F, G333D, G333E, G335C, G335D, N348I, A360I, A360V, V365I, A371V, A376S, and E399G were compared with a treatment-naive population. The association of CD mutations with inferred IC50 fold changes to nucleos(t)ide reverse transcriptase inhibitors was evaluated. Univariate and multivariate analyses examined the association of CD mutations with a >1 log10 per milliliter decrease in HIV viral load after 24 weeks on a new ARV regimen. RESULTS: Higher CD mutation rates were seen in Options in Management with Antiretrovirals patients (n = 345) compared with a treatment-naive population. CD mutations were associated with increased inferred IC50 fold changes to abacavir, stavudine, tenofovir, and zidovudine. On univariate analysis, A371V was associated with lack of virologic response, as was having any CD mutation on multivariate analysis. CONCLUSIONS: CD mutations are frequent in treatment-experienced populations. They are associated with reduced susceptibility to some nucleos(t)ide reverse transcriptase inhibitors and with a diminished response to ARV therapy.


Subject(s)
HIV Infections/virology , HIV Reverse Transcriptase/genetics , HIV-1/genetics , Mutation/drug effects , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , Female , Genes, Viral/drug effects , Genes, Viral/genetics , Genotype , HIV Infections/drug therapy , HIV Reverse Transcriptase/drug effects , HIV-1/drug effects , Humans , Male , Middle Aged , Phenotype
18.
Expert Opin Biol Ther ; 10(2): 201-13, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20088715

ABSTRACT

IMPORTANCE OF THE FIELD: Despite the clinical benefits of highly active antiretroviral therapy (HAART), the prospect of life-long antiretroviral treatment poses significant problems, which has spurred interest in developing new drugs and strategies to treat HIV infection and eliminate persistent viral reservoirs. RNAi has emerged as a therapeutic possibility for HIV. AREAS COVERED IN THIS REVIEW: We discuss progress in overcoming hurdles to translating transient and stable RNAi enabling technologies to clinical application for HIV; covering the past 2 - 3 years. WHAT THE READER WILL GAIN: HIV inhibition can be achieved by transfection of chemically or enzymatically synthesized siRNAs or by DNA-based vector systems expressing short hairpin RNAs (shRNAs) that are processed intracellularly into siRNA. We compare these approaches, focusing on technical and safety issues that will guide the choice of strategy for clinical use. TAKE HOME MESSAGE: Introduction of synthetic siRNA into cells or its stable endogenous production using vector-driven shRNA have been shown to suppress HIV replication in vitro and, in some instances, in vivo. Each method has advantages and limitations in terms of ease of delivery, duration of silencing, emergence of escape mutants and potential toxicity. Both appear to have potential as future therapeutics for HIV, once the technical and safety issues of each approach are overcome.


Subject(s)
HIV Infections/therapy , HIV-1 , RNA Interference , RNA, Small Interfering/therapeutic use , Animals , Drug Design , Genes, Viral/drug effects , Genetic Vectors , HIV Infections/genetics , HIV-1/genetics , Humans , RNA, Small Interfering/adverse effects , RNA, Small Interfering/chemical synthesis
19.
FEBS J ; 276(16): 4372-80, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19614744

ABSTRACT

We report here a new method for inhibition of RNA viruses induced by dsDNA. We demonstrated that both long dsDNA molecules and short interfering DNA with a sequence complementary to that of viral RNA inhibited tobacco mosaic virus expression and prevented virus spread. Also, the expression of the HIV-1 gp41 gene in HeLa cells was inhibited by complementary short interfering DNA. We showed that Dicer processed dsDNA, which suggests activation of the cellular machinery involved in silencing of RNA. For the silencing of viral RNA effected with dsDNA, we coined the term DNA interference technology.


Subject(s)
DNA, Antisense/pharmacology , Gene Silencing/drug effects , Genes, Viral/drug effects , RNA Viruses/drug effects , RNA, Viral/antagonists & inhibitors , DEAD-box RNA Helicases/physiology , DNA, Antisense/genetics , HIV Envelope Protein gp41/antagonists & inhibitors , HIV Envelope Protein gp41/genetics , HeLa Cells , Humans , Methods , Nucleic Acid Hybridization/methods , Oligodeoxyribonucleotides, Antisense/genetics , Oligodeoxyribonucleotides, Antisense/pharmacology , RNA Viruses/genetics , Ribonuclease III/physiology , Tobacco Mosaic Virus/drug effects , Tobacco Mosaic Virus/genetics
20.
Cancer Res ; 69(11): 4878-84, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19487298

ABSTRACT

High-risk human papillomavirus (HPV) infections are necessary but insufficient causes of cervical cancers. Other risk factors for cervical cancer (e.g., pregnancy, smoking, infections causing inflammation) can lead to high and sustained nitric oxide (NO) concentrations in the cervix, and high NO levels are related to carcinogenesis through DNA damage and mutation. However, the effects of NO exposure in HPV-infected cells have not been investigated. In this study, we used the NO donor DETA-NO to model NO exposure to cervical epithelium. In cell culture media, 24-hour exposure to 0.25 to 0.5 mmol/L DETA-NO yielded a pathologically relevant NO concentration. Exposure of cells maintaining episomal high-risk HPV genomes to NO increased HPV early transcript levels 2- to 4-fold but did not increase viral DNA replication. Accompanying increased E6 and E7 mRNA levels were significant decreases in p53 and pRb protein levels, lower apoptotic indices, increased DNA double-strand breaks, and higher mutation frequencies when compared with HPV-negative cells. We propose that NO is a molecular cofactor with HPV infection in cervical carcinogenesis, and that modifying local NO cervical concentrations may constitute a strategy whereby HPV-related cancer can be reduced.


Subject(s)
DNA Damage , Genes, Viral/drug effects , Mutation/drug effects , Nitric Oxide/pharmacology , Papillomavirus Infections/genetics , Transcription, Genetic/drug effects , 3T3 Cells , Animals , Cells, Cultured , Cervix Uteri/pathology , Cervix Uteri/virology , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Epithelial Cells/virology , Female , Gene Expression Regulation, Viral/drug effects , Genome, Viral/drug effects , Humans , Mice , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...