Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
PLoS Pathog ; 15(7): e1007869, 2019 07.
Article in English | MEDLINE | ID: mdl-31291371

ABSTRACT

Clonal expansion of HIV infected cells plays an important role in the formation and persistence of the reservoir that allows the virus to persist, in DNA form, despite effective antiretroviral therapy. We used integration site analysis to ask if there is a similar clonal expansion of SIV infected cells in macaques. We show that the distribution of HIV and SIV integration sites in vitro is similar and that both viruses preferentially integrate in many of the same genes. We obtained approximately 8000 integration sites from blood samples taken from SIV-infected macaques prior to the initiation of ART, and from blood, spleen, and lymph node samples taken at necropsy. Seven clones were identified in the pre-ART samples; one persisted for a year on ART. An additional 100 clones were found only in on-ART samples; a number of these clones were found in more than one tissue. The timing and extent of clonal expansion of SIV-infected cells in macaques and HIV-infected cells in humans is quite similar. This suggests that SIV-infected macaques represent a useful model of the clonal expansion of HIV infected cells in humans that can be used to evaluate strategies intended to control or eradicate the viral reservoir.


Subject(s)
HIV Infections/drug therapy , HIV Infections/virology , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/virology , Animals , Anti-Retroviral Agents/therapeutic use , CD4-Positive T-Lymphocytes/virology , Disease Reservoirs/virology , HIV Infections/pathology , Host Microbial Interactions/drug effects , Host Microbial Interactions/genetics , Host Microbial Interactions/physiology , Humans , In Vitro Techniques , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus/pathogenicity , Viral Load/drug effects , Virus Integration/genetics , Virus Integration/physiology , Virus Replication/drug effects
2.
Hum Gene Ther ; 29(10): 1183-1201, 2018 10.
Article in English | MEDLINE | ID: mdl-30160169

ABSTRACT

Genetically modified, autologous hematopoietic stem and progenitor cells (HSPCs) represent a new class of genetic medicine. Following this therapeutic paradigm, we are developing a product candidate, designated CD68-ET3-LV CD34+, for the treatment of the severe bleeding disorder, hemophilia A. The product consists of autologous CD34+ cells transduced with a human immunodeficiency virus 1-based, monocyte lineage-restricted, self-inactivating lentiviral vector (LV), termed CD68-ET3-LV, encoding a bioengineered coagulation factor VIII (fVIII) transgene, termed ET3, designed for enhanced expression. This vector was shown capable of high-titer manufacture under clinical scale and Good Manufacturing Practice. Biochemical and immunogenicity testing of recombinant ET3, as well as safety and efficacy testing of CD68-ET3-LV HSPCs, were utilized to demonstrate overall safety and efficacy in murine models. In the first model, administration of CD68-ET3-LV-transduced stem-cell antigen-1+ cells to hemophilia A mice resulted in sustained plasma fVIII production and hemostatic correction without signs of toxicity. Patient-derived, autologous mobilized peripheral blood (mPB) CD34+ cells are the clinical target cells for ex vivo transduction using CD68-ET3-LV, and the resulting genetically modified cells represent the investigational drug candidate. In the second model, CD68-ET3-LV gene transfer into mPB CD34+ cells isolated from normal human donors was utilized to obtain in vitro and in vivo pharmacology, pharmacokinetic, and toxicology assessment. CD68-ET3-LV demonstrated reproducible and efficient gene transfer into mPB CD34+ cells, with vector copy numbers in the range of 1 copy per diploid genome equivalent without affecting clonogenic potential. Differentiation of human CD34+ cells into monocytes was associated with increased fVIII production, supporting the designed function of the CD68 promoter. To assess in vivo pharmacodynamics, CD68-ET3-LV CD34+ cell product was administered to immunodeficient mice. Treated mice displayed sustained plasma fVIII levels and no signs of product related toxicity. Collectively, the findings of the current study support the preclinical safety and efficacy of CD68-ET3-LV CD34+.


Subject(s)
Factor VIII/genetics , Genetic Engineering , Genetic Vectors/genetics , Hematopoietic Stem Cells/metabolism , Hemophilia A/genetics , Hemophilia A/therapy , Lentivirus/genetics , Animals , Blood Coagulation , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Gene Expression , Gene Order , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Genetic Vectors/adverse effects , Humans , Male , Mice , Mice, Transgenic , Mutagenesis, Insertional , Swine , Transduction, Genetic , Transgenes , Treatment Outcome , Virus Integration
3.
Mol Med Rep ; 18(2): 1682-1691, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29845244

ABSTRACT

The aim of the present study was to explore the effect of Bushen recipe and its disassembled prescriptions on liver injury and chronic hepatitis B. Liver injury was induced in normal and hepatitis B virus (HBV)­transgenic mice through injection of Concanavalin A, followed by treatment with Bushen recipe and its disassembled prescriptions including the Bushen­yang, the Bushen­yin and the QingHua groups as well as the GanYanLing group (positive control). Subsequently, their liver function indexes were investigated by a microplate method and liver sections were blindly evaluated using an optical microscope by a pathologist. Subsequently, the activation state of Toll­like receptor (TLR)3/9 signaling pathway in liver tissues was analyzed by western blotting. Additionally, the inflammatory factors produced following liver injury in peripheral blood were detected via ELISA. Following intervention with the Bushen recipe and its disassembled prescriptions, the liver function indexe alanine aminotransferase had declined, whereas cholinesterase increased. The pathological alterations of liver tissue in HBV transgenic mice were reversed by Bushen recipe and its disassembled prescriptions. In addition, the TLR3/9 signaling pathway in liver tissues of HBV transgenic mice was inhibited and inflammatory factors such as interleukin (IL)­6, IL­1, tumor necrosis factor­α and interferon­Î³ were reduced significantly. In conclusion, the present study demonstrated that Bushen recipe and its disassembled prescriptions repaired liver injury induced by Concanavalin A through inhibition of TLR3/9 signaling pathway.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Chemical and Drug Induced Liver Injury/drug therapy , Drugs, Chinese Herbal/pharmacology , Genome , Toll-Like Receptor 3/genetics , Toll-Like Receptor 9/genetics , Animals , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/pathology , Concanavalin A , Gene Expression Regulation , Hepatitis B virus/genetics , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-1/antagonists & inhibitors , Interleukin-1/genetics , Interleukin-1/immunology , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , Interleukin-6/immunology , Male , Mice , Mice, Transgenic , Signal Transduction , Toll-Like Receptor 3/antagonists & inhibitors , Toll-Like Receptor 3/immunology , Toll-Like Receptor 9/antagonists & inhibitors , Toll-Like Receptor 9/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Virus Integration
4.
BMC Microbiol ; 15: 274, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26630969

ABSTRACT

BACKGROUND: Small-molecule compounds that inhibit human immunodeficiency virus type 1 (HIV-1) infection can be used not only as drug candidates, but also as reagents to dissect the life cycle of the virus. Thus, it is desirable to have an arsenal of such compounds that inhibit HIV-1 infection by various mechanisms. Until now, only a few small-molecule compounds that inhibit nuclear entry of viral DNA have been documented. RESULTS: We identified a novel, small-molecule compound, SJP-L-5, that inhibits HIV-1 infection. SJP-L-5 is a nitrogen-containing, biphenyl compound whose synthesis was based on the dibenzocyclooctadiene lignan gomisin M2, an anti-HIV bioactive compound isolated from Schisandra micrantha A. C. Smith. SJP-L-5 displayed relatively low cytotoxicity (50% cytoxicity concentrations were greater than 200 µg/ml) and high antiviral activity against a variety of HIV strains (50% effective concentrations (EC50)) of HIV-1 laboratory-adapted strains ranged from 0.16-0.97 µg/ml; EC50s of primary isolates ranged from 1.96-5.33 µg/ml). Analyses of the viral DNA synthesis indicated that SJP-L-5 specifically blocks the entry of the HIV-1 pre-integration complex (PIC) into the nucleus. Further results implicated that SJP-L-5 inhibits the disassembly of HIV-1 particulate capsid in the cytoplasm of the infected cells. CONCLUSIONS: SJP-L-5 is a novel small-molecule compound that inhibits HIV-1 nuclear entry by blocking the disassembly of the viral core.


Subject(s)
Anti-HIV Agents/pharmacology , DNA, Viral/metabolism , HIV-1/drug effects , HIV-1/physiology , Virus Integration/drug effects , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/isolation & purification , Anti-HIV Agents/toxicity , Cell Line , Cell Survival/drug effects , Humans , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Schisandra/chemistry
5.
Biochem Pharmacol ; 94(4): 241-56, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25732195

ABSTRACT

HIV-1 infection cannot be cured due to reservoirs formed early after infection. Decreasing the massive CD4+ T cell activation that occurs at the beginning of the disease would delay reservoir seeding, providing a better prognosis for patients. CD4+ T cell activation is mediated by protein kinase C (PKC) theta (θ), which is involved in T-cell proliferation, as well as NF-κB, NF-AT, and AP-1 activation. We found that PKCθ activity increased viral replication, but also that HIV-1 induced higher activation of PKCθ in infected CD4+ T cells, creating a feedback loop. Therefore, specific inhibition of PKCθ activity could contribute to control HIV-1 replication. We tested the efficacy of seven PKCθ specific inhibitors to control HIV-1 replication in CD4+ T cells and selected two of the more potent and safer: CGX1079 and CGX0471. They reduced PKCθ phosphorylation at T538 and its translocation to the plasma membrane, which correlated with decreased HIV-1 retrotranscription through partial inhibition of SAMHD1 antiviral activity, rendering lower proviral integration. CGX1079 and CGX0471 also interfered with viral transcription, which would reduce the production of new virions, as well as the subsequent spread and infection of new targets that would increase the reservoir size. CGX1079 and CGX0471 did not completely abrogate T-cell functions such as proliferation and CD8-mediated release of IFN-γ in PBMCs from HIV-infected patients, thereby avoiding general immunosuppresion. Consequently, using PKCθ inhibitors as adjuvant of antiretroviral therapy in recently infected patients would decrease the pool of activated CD4+ T cells, thwarting proviral integration and reducing the reservoir size.


Subject(s)
Anti-HIV Agents/pharmacology , CD4-Positive T-Lymphocytes/drug effects , HIV-1/drug effects , Isoenzymes/antagonists & inhibitors , Protein Kinase C/antagonists & inhibitors , Retroelements , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/virology , Cell Membrane/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , HIV-1/genetics , HIV-1/physiology , Humans , Jurkat Cells , Monomeric GTP-Binding Proteins/metabolism , Phosphorylation , Protein Kinase C-theta , Protein Transport , SAM Domain and HD Domain-Containing Protein 1 , Transcription, Genetic , Virus Integration/drug effects , Virus Internalization , Virus Replication
6.
Chin J Nat Med ; 12(3): 186-93, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24702804

ABSTRACT

AIM: To evaluate the anti-HIV activity and mechanism of action of wikstroelide M, a daphnane diterpene from Daphne acutiloba Rehder (Thymelaeaceae). METHODS: The anti-HIV activities of wikstroelide M against different HIV strains were evaluated by cytopathic effect assay and p24 quantification assay with ELISA. The inhibitory effect of wikstroelide M on HIV reverse transcription was analyzed by real-time PCR and ELISA. The effect of wikstroelide M on HIV-1 integrase nuclear translocation was observed with a cell-based imaging assay. The effect of wikstroelide M on LEDGF/p75-IN interaction was assayed by molecular docking. RESULTS: Wikstroelide M potently inhibited different HIV-1 strains, including HIV-1IIIB, HIV-1A17, and HIV-19495, induced a cytopathic effect, with EC50 values ranging from 3.81 to 15.65 ng·mL⁻¹. Wikstroelide M also had high inhibitory activities against HIV-2ROD and HIV-2CBL-20-induced cytopathic effects with EC50 values of 18.88 and 31.90 ng·mL⁻¹. The inhibitory activities of wikstroelide M on the three HIV-1 strains were further confirmed by p24 quantification assay, with EC50 values ranging from 15.16 to 35.57 ng·mL⁻¹. Wikstroelide M also potently inhibited HIV-1IIIB induced cytolysis in MT-4 cells, with an EC50 value of 9.60 ng·mL⁻¹. The mechanistic assay showed that wikstroelide M targeted HIV-1 reverse transcriptase and nuclear translocation of integrase through disrupting the interaction between integrase and LEDGF/p75. CONCLUSION: Wikstroelide M may be a potent HIV-1 and HIV-2 inhibitor, the mechanisms of action may include inhibition of reverse trascriptase activity and inhibition of integrase nuclear translocation through disrupting the interaction between integrase and LEDGF/p75.


Subject(s)
Anti-HIV Agents/pharmacology , Daphne/chemistry , Diterpenes/pharmacology , HIV Integrase/metabolism , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , HIV-2/drug effects , Plant Extracts/pharmacology , Anti-HIV Agents/therapeutic use , Cell Line , HIV Infections/drug therapy , HIV Infections/virology , HIV Integrase Inhibitors/pharmacology , HIV Integrase Inhibitors/therapeutic use , HIV-1/enzymology , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Phytotherapy , Plant Extracts/therapeutic use , Virus Integration/drug effects , Virus Replication/drug effects
7.
Article in English | WPRIM | ID: wpr-812288

ABSTRACT

AIM@#To evaluate the anti-HIV activity and mechanism of action of wikstroelide M, a daphnane diterpene from Daphne acutiloba Rehder (Thymelaeaceae).@*METHODS@#The anti-HIV activities of wikstroelide M against different HIV strains were evaluated by cytopathic effect assay and p24 quantification assay with ELISA. The inhibitory effect of wikstroelide M on HIV reverse transcription was analyzed by real-time PCR and ELISA. The effect of wikstroelide M on HIV-1 integrase nuclear translocation was observed with a cell-based imaging assay. The effect of wikstroelide M on LEDGF/p75-IN interaction was assayed by molecular docking.@*RESULTS@#Wikstroelide M potently inhibited different HIV-1 strains, including HIV-1IIIB, HIV-1A17, and HIV-19495, induced a cytopathic effect, with EC50 values ranging from 3.81 to 15.65 ng·mL⁻¹. Wikstroelide M also had high inhibitory activities against HIV-2ROD and HIV-2CBL-20-induced cytopathic effects with EC50 values of 18.88 and 31.90 ng·mL⁻¹. The inhibitory activities of wikstroelide M on the three HIV-1 strains were further confirmed by p24 quantification assay, with EC50 values ranging from 15.16 to 35.57 ng·mL⁻¹. Wikstroelide M also potently inhibited HIV-1IIIB induced cytolysis in MT-4 cells, with an EC50 value of 9.60 ng·mL⁻¹. The mechanistic assay showed that wikstroelide M targeted HIV-1 reverse transcriptase and nuclear translocation of integrase through disrupting the interaction between integrase and LEDGF/p75.@*CONCLUSION@#Wikstroelide M may be a potent HIV-1 and HIV-2 inhibitor, the mechanisms of action may include inhibition of reverse trascriptase activity and inhibition of integrase nuclear translocation through disrupting the interaction between integrase and LEDGF/p75.


Subject(s)
Humans , Anti-HIV Agents , Pharmacology , Therapeutic Uses , Cell Line , Daphne , Chemistry , Diterpenes , Pharmacology , HIV Infections , Drug Therapy , Virology , HIV Integrase , Metabolism , HIV Integrase Inhibitors , Pharmacology , Therapeutic Uses , HIV Reverse Transcriptase , HIV-1 , HIV-2 , Intercellular Signaling Peptides and Proteins , Metabolism , Phytotherapy , Plant Extracts , Pharmacology , Therapeutic Uses , Virus Integration , Virus Replication
8.
Antiviral Res ; 98(3): 394-400, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23588231

ABSTRACT

Once the human immunodeficiency virus (HIV) genome is inserted into the host genome, the virus cannot be removed, which results in latency periods and makes it difficult to eradicate. The majority of strategies to eradicate HIV have been based on preventing virus latency, thereby enabling antiretroviral drugs to act against HIV replication. Another innovative strategy is permanently silencing the integrated virus to prevent the spread of infection. Epigenetic processes are natural mechanisms that can silence viral replication. We describe a new chimeric protein (IN3b) that consists of a HIV-1 integrase domain, which recognises the HIV long terminal repeat (LTR) and the catalytic domain of DNA methyltransferase DNMT3b. Our objective was to silence HIV replication by the specific delivery of the catalytic methyltransferase domain to the LTR promoter to induce its methylation. We found that our IN3b chimeric protein was expressed in the nucleus and decreased LTR-associated HIV genome expression and HIV replication. Therefore, the IN3b chimeric protein may be an effective tool against HIV replication and maybe used in a new line of research to induce or maintain HIV latency.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , Gene Silencing , Genome, Viral , HIV Long Terminal Repeat , Recombinant Fusion Proteins/metabolism , Catalytic Domain , Cell Nucleus , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , Drug Evaluation, Preclinical , Genetic Vectors/genetics , Genetic Vectors/metabolism , HEK293 Cells , HIV Integrase/genetics , HIV Integrase/metabolism , HIV-1/physiology , Humans , Point Mutation , Promoter Regions, Genetic , Recombinant Fusion Proteins/genetics , Transcription, Genetic , Transfection , Virus Integration , Virus Latency , Virus Replication , DNA Methyltransferase 3B
9.
Anticancer Drugs ; 24(4): 337-43, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23388161

ABSTRACT

Globally, hepatitis B virus (HBV) and/or hepatitis C virus (HCV) infection leads to liver fibrosis and cirrhosis, which in turn causes resultant hepatocellular carcinoma (HCC). Frequently, HCC recurs very soon even after a potentially curative treatment such as surgical interference or locoregional ablative therapies. Chronic HBV/HCV infection is often responsible for this recurrence, through secondary carcinogenesis. Antiviral therapy after a curative treatment of HCC plays an important role in preventing or delaying recurrence and improves survival in patients with HBV/HCV infection-related HCC. This article reviews the worldwide epidemiology of HBV/HCV infection, the association of viral infection with HCC, the mechanism of hepatitis virus-related hepatocarcinogenesis, and the paramount importance of antiviral therapy in the management of HCC.


Subject(s)
Antiviral Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Hepatitis B, Chronic/drug therapy , Hepatitis C, Chronic/drug therapy , Liver Neoplasms/drug therapy , Tumor Virus Infections/drug therapy , Antineoplastic Agents/therapeutic use , Apoptosis , Carcinoma, Hepatocellular/epidemiology , Carcinoma, Hepatocellular/surgery , Carcinoma, Hepatocellular/virology , Carrier State/epidemiology , Cell Transformation, Viral , Combined Modality Therapy , Complementary Therapies , Drug Therapy, Combination , Gene Expression Regulation, Viral , Hepacivirus/genetics , Hepacivirus/pathogenicity , Hepatectomy , Hepatitis B virus/genetics , Hepatitis B virus/pathogenicity , Hepatitis B, Chronic/complications , Hepatitis B, Chronic/epidemiology , Hepatitis C, Chronic/complications , Hepatitis C, Chronic/epidemiology , Host-Pathogen Interactions , Humans , Liver Neoplasms/epidemiology , Liver Neoplasms/surgery , Liver Neoplasms/virology , Secondary Prevention , Tumor Virus Infections/complications , Virus Integration
10.
Am J Chin Med ; 40(1): 151-62, 2012.
Article in English | MEDLINE | ID: mdl-22298455

ABSTRACT

Human respiratory syncytial virus (HRSV) causes serious pediatric infection of the lower respiratory tract without effective therapeutic modality. Sheng-Ma-Ge-Gen-Tang (SMGGT; Shoma-kakkon-to) has been proven to be effective at inhibiting HRSV-induced plaque formation, and Cimicifuga foetida is the major constituent of SMGGT. We tested the hypothesis that C. foetida effectively inhibited the cytopathic effects of HRSV by a plaque reduction assay in both human upper (HEp2) and lower (A549) respiratory tract cell lines. Its ability to stimulate anti-viral cytokines was evaluated by an enzyme-linked immunosorbent assay (ELISA). C. foetida dose-dependently inhibited HRSV-induced plaque formation (p < 0.0001) before and after viral inoculation, especially in A549 cells (p < 0.0001). C. foetida dose-dependently inhibited viral attachment (p < 0.0001) and could increase heparins effect on viral attachment. In addition, C. foetida time-dependently and dose-dependently (p < 0.0001) inhibited HRSV internalization. C. foetida could stimulate epithelial cells to secrete IFN-ß to counteract viral infection. However, C. foetida did not stimulate TNF-α secretion. Therefore, C. foetida could be useful in managing HRSV infection. This is the first evidence to support that C. foetida possesses antiviral activity.


Subject(s)
Actaea , Antiviral Agents/therapeutic use , Cimicifuga , Drugs, Chinese Herbal/therapeutic use , Phytotherapy , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus, Human/drug effects , Antiviral Agents/pharmacology , Cell Line , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/pharmacology , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Heparin/pharmacology , Humans , Interferon-beta/metabolism , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/pathogenicity , Respiratory System/drug effects , Respiratory System/virology , Tumor Necrosis Factor-alpha/metabolism , Viral Plaque Assay , Virus Integration/drug effects
11.
Antivir Chem Chemother ; 22(2): 67-74, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21984686

ABSTRACT

BACKGROUND: In addition to activities needed to catalyse integration, retroviral integrases exhibit non-specific endonuclease activity that is enhanced by certain small compounds, suggesting that integrase could be stimulated to damage viral DNA before integration occurs. METHODS: A non-radioactive, plate-based, solution phase, fluorescence assay was used to screen a library of 50,080 drug-like chemicals for stimulation of non-specific DNA nicking by HIV-1 integrase. RESULTS: A semi-automated workflow was established and primary hits were readily identified from a graphic output. Overall, 0.6% of the chemicals caused a large increase in fluorescence (the primary hit rate) without also having visible colour that could have artifactually caused this result. None of the potential stimulators from this moderate-size library, however, passed a secondary test that included an inactive integrase mutant that assessed whether the increased fluorescence depended on the endonuclease activity of integrase. CONCLUSIONS: This first attempt at identifying integrase stimulator compounds establishes the necessary logistics and workflow required. The results from this study should encourage larger scale high-throughput screening to advance the novel antiviral strategy of stimulating integrase to damage retroviral DNA.


Subject(s)
Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Drug Evaluation, Preclinical/methods , HIV Integrase/genetics , Small Molecule Libraries , DNA Breaks, Single-Stranded , Fluorescence , Virus Integration/drug effects
12.
J Biol Chem ; 286(24): 21083-91, 2011 Jun 17.
Article in English | MEDLINE | ID: mdl-21498519

ABSTRACT

Current antiretroviral therapy (ART) provides potent suppression of HIV-1 replication. However, ART does not target latent viral reservoirs, so persistent infection remains a challenge. Small molecules with pharmacological properties that allow them to reach and activate viral reservoirs could potentially be utilized to eliminate the latent arm of the infection when used in combination with ART. Here we describe a cell-based system modeling HIV-1 latency that was utilized in a high-throughput screen to identify small molecule antagonists of HIV-1 latency. A more detailed analysis is provided for one of the hit compounds, antiviral 6 (AV6), which required nuclear factor of activated T cells for early mRNA expression while exhibiting RNA-stabilizing activity. It was found that AV6 reproducibly activated latent provirus from different lymphocyte-based clonal cell lines as well as from latently infected primary resting CD4(+) T cells without causing general T cell proliferation or activation. Moreover, AV6 complemented the latency antagonist activity of a previously described histone deacetylase (HDAC) inhibitor. This is a proof of concept showing that a high-throughput screen employing a cell-based model of HIV-1 latency can be utilized to identify new classes of compounds that can be used in concert with other persistent antagonists with the aim of viral clearance.


Subject(s)
Drug Evaluation, Preclinical/methods , HIV-1/metabolism , Histone Deacetylase Inhibitors/pharmacology , Anti-Retroviral Agents/therapeutic use , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Proliferation , Drug Design , Flow Cytometry/methods , Gene Expression Regulation, Viral , Genome, Viral , Humans , Lentivirus/genetics , Lymphocyte Activation , Virus Integration , Virus Latency
13.
J Ethnopharmacol ; 134(2): 468-74, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21211557

ABSTRACT

AIM OF THE STUDY: Extracts from the aerial parts of the South African resurrection plant Myrothamnus flabellifolia Welw. have been used traditionally against infections of the upper respiratory tract and skin diseases. A polyphenol-enriched extract was investigated for potential antiviral effects against herpes simplex virus type 1 (HSV-1) and adenovirus, and the underlying mode of action was to be studied. MATERIALS AND METHODS: Antiviral effects of an acetone-water extract (MF) from Myrothamnus flabellifolia on HSV-1 and adenovirus type 3 were tested in infected Vero cells by plaque reduction assay, MTT test and immunofluorescence. The influence of the extract on the HSV-1 envelope glycoprotein D was shown by Western blot. Organotypic full thickness skin models consisting of multilayer skin equivalents were used for the investigation of MF effects on HSV-1 replication. RESULTS: MF exhibited strong antiviral activity against HSV-1. The HSV-1-specific inhibitory concentration (IC(50)) was determined as 0.4 µg/mL and the cytotoxic concentration (CC(50)) against Vero cells as 50 µg/mL. A selectivity index (SI) (ratio of CC(50) to IC(50)) of approximately 120 was calculated when MF was added to the virus inoculum for 1h at 37°C prior to infection. The replication of adenovirus 3 was not affected by MF. MF abolished virus entry into the host cell by blocking viral attachment to the cell surface. When added after attachment at a concentration of >6 µg/mL, the extract also inhibited penetration of HSV-1 into the host cell. Polyphenolic compounds from MF directly interacted with viral particles, leading to the oligomerisation of envelope proteins as demonstrated for the essential viral glycoprotein D (gD). Using organotypic full thickness tissue cultures, it was shown that treatment of HSV-1 infected cultures with the MF resulted in reduced viral spread. CONCLUSIONS: A polyphenol-enriched extract from Myrothamnus flabellifolia strongly acts against HSV-1 by blocking viral entry into the cells.


Subject(s)
Antiviral Agents/therapeutic use , Herpes Simplex/drug therapy , Herpesvirus 1, Human/drug effects , Magnoliopsida/chemistry , Phytotherapy , Plant Extracts/therapeutic use , Proanthocyanidins/therapeutic use , Adenoviridae/drug effects , Adenoviridae Infections/microbiology , Animals , Antiviral Agents/pharmacology , Cell Line , Chlorocebus aethiops , Herpes Simplex/microbiology , Herpesvirus 1, Human/chemistry , Herpesvirus 1, Human/pathogenicity , Humans , Inhibitory Concentration 50 , Keratinocytes/drug effects , Keratinocytes/microbiology , Plant Components, Aerial , Plant Extracts/pharmacology , Proanthocyanidins/pharmacology , Skin/drug effects , Skin/microbiology , Vero Cells , Viral Envelope Proteins/chemistry , Virus Integration/drug effects
14.
J Gene Med ; 12(7): 596-603, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20533531

ABSTRACT

BACKGROUND: A recurrent breast cancer patient received high-dose chemotherapy, a transplant of multidrug resistance 1 (MDR1)-transduced cells and four different protocols of post-transplantation chemotherapy. We report the analysis of MDR1-transduced cells in this patient. METHODS: MDR1 transgene levels in the peripheral blood mononuclear cells of the patient were evaluated by polymerase chain reaction (PCR). Retroviral integration sites of the MDR1-transduced cells were identified by linear amplification-mediated (LAM)-PCR. RESULTS: Twelve days after transplantation, approximately 1% of the peripheral blood mononuclear cells were MDR1 transgene-positive. The transgene levels decreased quickly, and were at low levels until day 504. A remarkable increase in MDR1 transgene-positive cells was observed on day 532, during combination chemotherapy with mitomycin C and methotrexate. Using LAM-PCR, 31 MDR1-transduced clones were identified, and eight of these were long-life clones that survived for more than 500 days. Among the 31 clones, ten had a retroviral integration site near genes listed in the Retroviral Tagged Cancer Gene (RTCG) Database. Two long-life clones, N-30 and N-31, had retroviral integration sites within the MDS1-EVI1 locus. Another two long-life clones had integration sites close to PRDM16 or CUEDC1. CONCLUSIONS: These results suggest that MDR1-transduced cells were enriched in vivo by an MDR1 substrate, mitomycin C. The possible activation of EVI1 or other RTCGs by retroviral insertion may have affected the survival and persistence of a proportion of the transduced cells.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Breast Neoplasms/drug therapy , Hematopoietic Stem Cell Transplantation , Methotrexate/therapeutic use , Mitomycin/therapeutic use , Transduction, Genetic , Cell Proliferation/drug effects , Clone Cells , DNA-Binding Proteins/genetics , Female , Genes, Neoplasm/genetics , Humans , MDS1 and EVI1 Complex Locus Protein , Methotrexate/pharmacology , Mitomycin/pharmacology , Polymerase Chain Reaction , Proto-Oncogenes/genetics , Retroviridae/genetics , Transcription Factors/genetics , Transgenes/genetics , Virus Integration/drug effects
15.
J Virol ; 84(16): 8250-61, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20519390

ABSTRACT

Retroviral integrases associate during the early viral life cycle with preintegration complexes that catalyze the integration of reverse-transcribed viral cDNA into the host chromosomes. Several cellular and viral proteins have been reported to be incorporated in the preintegration complex. This study demonstrates that transcription factor Yin Yang 1 binds to Moloney murine leukemia virus, human immunodeficiency virus type 1, and avian sarcoma virus integrases. The results of coimmunoprecipitation and in vitro pulldown assays revealed that Yin Yang 1 interacted with the catalytic core and C-terminal domains of Moloney murine leukemia virus and human immunodeficiency virus type 1 integrases, while the transcriptional repression and DNA-binding domains of the Yin Yang 1 molecule interacted with Moloney murine leukemia virus integrase. Immunoprecipitation of the cytoplasmic fraction of virus-infected cells followed by Southern blotting and chromatin immunoprecipitation demonstrated that Yin Yang 1 associated with Moloney murine leukemia virus cDNA in virus-infected cells. Yin Yang 1 enhanced the in vitro integrase activity of Moloney murine leukemia virus, human immunodeficiency virus type 1, and avian sarcoma virus integrases. Furthermore, knockdown of Yin Yang 1 in host cells by small interfering RNA reduced Moloney murine leukemia virus cDNA integration in vivo, although viral cDNA synthesis was increased, suggesting that Yin Yang 1 facilitates integration events in vivo. Taking these results together, Yin Yang 1 appears to be involved in integration events during the early viral life cycle, possibly as an enhancer of integration.


Subject(s)
Integrases/metabolism , Moloney murine leukemia virus/enzymology , Moloney murine leukemia virus/physiology , Protein Interaction Mapping , Viral Proteins/metabolism , Virus Integration , YY1 Transcription Factor/metabolism , Avian Sarcoma Viruses/enzymology , Cell Fractionation , DNA, Complementary/metabolism , DNA, Viral/metabolism , Gene Knockdown Techniques , HIV-1/enzymology , Humans , Immunoprecipitation , Protein Binding , YY1 Transcription Factor/genetics
16.
Ethn Dis ; 20(1 Suppl 1): S1-45-9, 2010.
Article in English | MEDLINE | ID: mdl-20521384

ABSTRACT

An essential step in the life cycle of human immunodeficiency virus type 1 (HIV-1) is integration of the double-stranded retroviral DNA into the genome of the host cell. HIV-1 integrase, the enzyme that inserts the vital DNA into the host chromosome, is an attractive and rational target for anti-AIDS drug design because it is essential for HIV replication and there are no known counterparts in the host cell. Inhibitors of this enzyme have a great potential to complement the therapeutic use of HIV protease and reverse transcriptase inhibitors. Natural products have provided a source of new drug candidates for anti-AIDS therapy. Dicaffeoylquinic acids, isolated from traditional medicinal plants, are a novel class of integrase inhibitors. These compounds are potent inhibitors of HIV-1 replication in cultured cell lines and catalytic activities of integrase in vitro. They are therefore promising compounds for developing new anti-AIDS drugs. To understand how the inhibitors work and therefore design more potent and specific inhibitors, we have used molecular modeling techniques to investigate the binding modes of 3,4-dicaffeoylquinic acid. Our computational modeling study demonstrated that the inhibitor of this compound on HIV integrase is likely to proceed by two different but equivalent mechanisms with one bound to the active site region of the enzyme and another docked into the binding pocket located on the other side of the catalytic site. Our study will be of help to design new pharmaceuticals for the treatment of AIDS.


Subject(s)
HIV Integrase/physiology , HIV-1/physiology , Integrase Inhibitors/pharmacology , Quinic Acid/analogs & derivatives , Virus Replication/drug effects , Acquired Immunodeficiency Syndrome/drug therapy , Catalytic Domain/drug effects , Catalytic Domain/physiology , Computational Biology , Drug Design , Protein Binding , Quinic Acid/antagonists & inhibitors , Structure-Activity Relationship , Virus Integration/drug effects , Virus Replication/physiology
17.
J Mol Biol ; 400(3): 487-501, 2010 Jul 16.
Article in English | MEDLINE | ID: mdl-20493881

ABSTRACT

The main function of the HIV-1 trans-activator of transcription (Tat protein) is to promote the transcription of the proviral DNA by the host RNA polymerase which leads to the synthesis of large quantities of the full length viral RNA. Tat is also thought to be involved in the reverse transcription (RTion) reaction by a still unknown mechanism. The recently reported nucleic acid annealing activity of Tat might explain, at least in part, its role in RTion. To further investigate this possibility, we carried out a fluorescence study on the mechanism by which the full length Tat protein (Tat(1-86)) and the basic peptide (44-61) direct the annealing of complementary viral DNA sequences representing the HIV-1 transactivation response element TAR, named dTAR and cTAR, essential for the early steps of RTion. Though both Tat(1-86) and the Tat(44-61) peptide were unable to melt the lower half of the cTAR stem, they strongly promoted cTAR/dTAR annealing through non-specific attraction between the peptide-bound oligonucleotides. Using cTAR and dTAR mutants, this Tat promoted-annealing was found to be nucleated through the thermally frayed 3'/5' termini, resulting in an intermediate with 12 intermolecular base pairs, which then converts into the final extended duplex. Moreover, we found that Tat(1-86) was as efficient as the nucleocapsid protein NCp7, a major nucleic acid chaperone of HIV-1, in promoting cTAR/dTAR annealing, and could act cooperatively with NCp7 during the annealing reaction. Taken together, our data are consistent with a role of Tat in the stimulation of the obligatory strand transfers during viral DNA synthesis by reverse transcriptase.


Subject(s)
Base Pairing , HIV-1/physiology , Nucleic Acids/metabolism , Reverse Transcription , Virus Integration , tat Gene Products, Human Immunodeficiency Virus/physiology , Amino Acid Sequence , DNA/metabolism , DNA, Complementary/metabolism , DNA, Viral/metabolism , Models, Molecular , Molecular Sequence Data , Nucleic Acid Conformation , Protein Binding , Zinc/metabolism
18.
Nat Med ; 16(4): 460-5, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20228817

ABSTRACT

Highly active antiretroviral therapy (HAART) results in potent and durable suppression of HIV-1 viremia. However, HIV-1 replication resumes if therapy is interrupted. Although it is generally believed that active replication has been halted in individuals on HAART, immune activation and inflammation continue at abnormal levels, suggesting continued, low-level viral replication. To assess whether active replication might be driving immune activation in HAART, we examined the impact of treatment intensification with the integrase inhibitor raltegravir on viral complementary DNA and immune activation parameters. In the presence of raltegravir, linear HIV-1 cDNA is prevented from integrating into chromatin and is subsequently converted to episomal cDNAs. Raltegravir intensification of a three-drug suppressive HAART regimen resulted in a specific and transient increase in episomal DNAs in a large percentage of HAART-suppressed subjects. Furthermore, in subjects with these episomal DNAs, immune activation was higher at baseline and was subsequently normalized after raltegravir intensification. These results suggest that, despite suppressive HAART, active replication persists in some infected individuals and drives immune activation. The ability of raltegravir intensification to perturb the reservoir that supports active replication has implications for therapeutic strategies aimed at achieving viral eradication.


Subject(s)
Antiretroviral Therapy, Highly Active , HIV Infections/drug therapy , HIV Integrase Inhibitors/therapeutic use , HIV-1/drug effects , Pyrrolidinones/therapeutic use , Virus Replication/drug effects , DNA, Complementary/genetics , DNA, Viral/genetics , HIV Infections/immunology , HIV Infections/virology , HIV Integrase Inhibitors/pharmacology , HIV Long Terminal Repeat/drug effects , HIV Long Terminal Repeat/genetics , HIV-1/immunology , HIV-1/physiology , Humans , Polymerase Chain Reaction , Pyrrolidinones/pharmacology , Raltegravir Potassium , Virus Integration/drug effects , Virus Replication/physiology
19.
Phytochemistry ; 71(2-3): 235-42, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19931103

ABSTRACT

Herpes simplex viruses (HSVs) display affinity for cell-surface heparan sulfate proteoglycans with biological relevance in virus entry. Here, we exploit an approach to inhibiting HSV infection by using a sulfated fucoidan, and a guluronic acid-rich alginate derived from Sargassum tenerrimum, mimicking the active domain of the entry receptor. These macromolecules have apparent molecular masses of 30+/-5 and 26+/-5 kDa, respectively. They and their chemically sulfated derivatives showed activity against herpes simplex virus type 1 (HSV-1). Their inhibitory concentration 50% (IC(50)) values were in the range 0.5-15 microg/ml and they lacked cytotoxicity at concentrations up to 1000 microg/ml. The anti-HSV activity increased with increasing sulfate ester content. Our results suggest the feasibility of inhibiting HSV infection by blocking viral entry with polysaccharide having specific structure.


Subject(s)
Alginates/pharmacology , Antiviral Agents/pharmacology , Herpes Simplex/drug therapy , Herpesvirus 1, Human/drug effects , Plant Extracts/pharmacology , Polysaccharides/pharmacology , Sargassum/chemistry , Alginates/chemistry , Alginates/therapeutic use , Animals , Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , Cells, Cultured , Glucuronic Acid/chemistry , Glucuronic Acid/pharmacology , Glucuronic Acid/therapeutic use , Haplorhini , Herpes Simplex/virology , Hexuronic Acids/chemistry , Hexuronic Acids/pharmacology , Hexuronic Acids/therapeutic use , Inhibitory Concentration 50 , Phytotherapy , Plant Extracts/chemistry , Plant Extracts/therapeutic use , Polysaccharides/chemistry , Polysaccharides/therapeutic use , Structure-Activity Relationship , Virus Integration/drug effects
20.
Phytomedicine ; 17(1): 69-74, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19748255

ABSTRACT

Screening of plants from the Iberian Peninsula for anti-human immunodeficiency virus (-HIV) activity revealed that aqueous extract of Tuberaria lignosa gave positive results. Following an activity-guided procedure, the crude extract was counterextracted, and the subsequent fractions obtained tested for their anti-HIV activity in vitro. The bioassay-guided fractionation of the extract afforded an ellagitannin enriched fraction (EEF) isolated for the first time from this species. This EEF exhibited antiviral activity against HIV in MT-2 infected cells, with an IC(50) value of 2.33mug/ml (selectivity index greater than 21). Inhibition of HIV infection by EEF appears to be mediated by CD4 down-regulation, the main receptor for HIV entry. CXCR4 and CCR5 receptors were not affected by EEF, explaining why EEF is able to inhibit R5 and X4 infections.


Subject(s)
Anti-HIV Agents/therapeutic use , Cistaceae/chemistry , HIV Infections/prevention & control , HIV/drug effects , Hydrolyzable Tannins/therapeutic use , Plant Extracts/therapeutic use , Virus Integration/drug effects , Anti-HIV Agents/isolation & purification , Anti-HIV Agents/pharmacology , CD4-Positive T-Lymphocytes/metabolism , Down-Regulation , Humans , Hydrolyzable Tannins/isolation & purification , Hydrolyzable Tannins/pharmacology , Inhibitory Concentration 50 , Jurkat Cells , Plant Extracts/chemistry , Plant Extracts/pharmacology , Receptors, CCR5 , Receptors, CXCR4
SELECTION OF CITATIONS
SEARCH DETAIL