Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nucleic Acids Res ; 46(11): 5776-5791, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29373715

ABSTRACT

More than 140 post-transcriptional modifications (PTMs) are known to decorate cellular RNAs, but their incidence, identity and significance in viral RNA are still largely unknown. We have developed an agnostic analytical approach to comprehensively survey PTMs on viral and cellular RNAs. Specifically, we used mass spectrometry to analyze PTMs on total RNA isolated from cells infected with Zika virus, Dengue virus, hepatitis C virus (HCV), poliovirus and human immunodeficiency virus type 1. All five RNA viruses significantly altered global PTM landscapes. Examination of PTM profiles of individual viral genomes isolated by affinity capture revealed a plethora of PTMs on viral RNAs, which far exceeds the handful of well-characterized modifications. Direct comparison of viral epitranscriptomes identified common and virus-specific PTMs. In particular, specific dimethylcytosine modifications were only present in total RNA from virus-infected cells, and in intracellular HCV RNA, and viral RNA from Zika and HCV virions. Moreover, dimethylcytosine abundance during viral infection was modulated by the cellular DEAD-box RNA helicase DDX6. By opening the Pandora's box on viral PTMs, this report presents numerous questions and hypotheses on PTM function and strongly supports PTMs as a new tier of regulation by which RNA viruses subvert the host and evade cellular surveillance systems.


Subject(s)
RNA Processing, Post-Transcriptional , RNA Viruses/genetics , RNA, Viral/metabolism , Cell Line, Tumor , Cytosine/metabolism , DEAD-box RNA Helicases/physiology , Humans , Proto-Oncogene Proteins/physiology , RNA Viruses/metabolism , RNA, Viral/chemistry , Stress, Physiological/genetics
2.
PLoS One ; 9(3): e93108, 2014.
Article in English | MEDLINE | ID: mdl-24667334

ABSTRACT

AIDS is a global pandemic that has seen the development of novel and effective treatments to improve the quality of life of those infected and reduction of spread of the disease. Palmitic Acid (PA), which we identified and isolated from Sargassum fusiforme, is a naturally occurring fatty acid that specifically inhibits HIV entry by binding to a novel pocket on the CD4 receptor. We also identified a structural analogue, 2-bromopalmitate (2-BP), as a more effective HIV entry inhibitor with a 20-fold increase in efficacy. We have used the structure-activity relationship (SAR) of 2-BP as a platform to identify new small chemical molecules that fit into the various identified active sites in an effort to identify more potent CD4 entry inhibitors. To validate further drug development, we tested the PA and 2-BP scaffold molecules for genotoxic potential. The FDA and International Conference on Harmonisation (ICH) recommends using a standardized 3-test battery for testing compound genotoxicity consisting of the bacterial reverse mutation assay, mouse lymphoma assay, and rat micronucleus assay. PA and 2-BP and their metabolites tested negative in all three genotoxicty tests. 2-BP is the first derivative of PA to undergo pre-clinical screening, which will enable us to now test multiple simultaneous small chemical structures based on activity in scaffold modeling across the dimension of pre-clinical testing to enable transition to human testing.


Subject(s)
Biological Products/chemistry , Biological Products/toxicity , HIV Fusion Inhibitors/chemistry , HIV Fusion Inhibitors/toxicity , HIV/drug effects , HIV/physiology , Virus Internalization/drug effects , Animals , Biological Products/pharmacology , Drug Discovery , Female , HIV Fusion Inhibitors/pharmacology , Lymphoma/pathology , Male , Mice , Micronucleus Tests , Palmitates/chemistry , Palmitates/pharmacology , Palmitates/toxicity , Palmitic Acid/chemistry , Palmitic Acid/pharmacology , Palmitic Acid/toxicity , Rats , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Structure-Activity Relationship
3.
PLoS One ; 6(9): e24803, 2011.
Article in English | MEDLINE | ID: mdl-21949756

ABSTRACT

BACKGROUND: Approximately 80% of all new HIV-1 infections are acquired through sexual contact. Currently, there is no clinically approved microbicide, indicating a clear and urgent therapeutic need. We recently reported that palmitic acid (PA) is a novel and specific inhibitor of HIV-1 fusion and entry. Mechanistically, PA inhibits HIV-1 infection by binding to a novel pocket on the CD4 receptor and blocks efficient gp120-to-CD4 attachment. Here, we wanted to assess the ability of PA to inhibit HIV-1 infection in cervical tissue ex vivo model of human vagina, and determine its effect on Lactobacillus (L) species of probiotic vaginal flora. PRINCIPAL FINDINGS: Our results show that treatment with 100-200 µM PA inhibited HIV-1 infection in cervical tissue by up to 50%, and this treatment was not toxic to the tissue or to L. crispatus and jensenii species of vaginal flora. In vitro, in a cell free system that is independent of in vivo cell associated CD4 receptor; we determined inhibition constant (Ki) to be ∼2.53 µM. SIGNIFICANCE: These results demonstrate utility of PA as a model molecule for further preclinical development of a safe and potent HIV-1 entry microbicide inhibitor.


Subject(s)
Anti-Infective Agents/pharmacology , Cervix Uteri/virology , Drug Discovery , HIV Infections/virology , HIV-1/drug effects , Palmitic Acid/pharmacology , Vagina/virology , CD4 Antigens/metabolism , Cervix Uteri/drug effects , Female , HIV Envelope Protein gp120/metabolism , HIV Infections/drug therapy , Humans , Lactobacillus/drug effects , Microbial Sensitivity Tests , Models, Biological , Palmitic Acid/therapeutic use , Vagina/drug effects
4.
PLoS One ; 5(8): e12168, 2010 Aug 13.
Article in English | MEDLINE | ID: mdl-20730055

ABSTRACT

BACKGROUND: We recently reported that palmitic acid (PA) is a novel and efficient CD4 fusion inhibitor to HIV-1 entry and infection. In the present report, based on in silico modeling of the novel CD4 pocket that binds PA, we describe discovery of highly potent PA analogs with increased CD4 receptor binding affinities (K(d)) and gp120-to-CD4 inhibition constants (K(i)). The PA analogs were selected to satisfy Lipinski's rule of drug-likeness, increased solubility, and to avoid potential cytotoxicity. PRINCIPAL FINDINGS: PA analog 2-bromopalmitate (2-BP) was most efficacious with K(d) approximately 74 nM and K(i) approximately 122 nM, ascorbyl palmitate (6-AP) exhibited slightly higher K(d) approximately 140 nM and K(i) approximately 354 nM, and sucrose palmitate (SP) was least efficacious binding to CD4 with K(d) approximately 364 nM and inhibiting gp120-to-CD4 binding with K(i) approximately 1486 nM. Importantly, PA and its analogs specifically bound to the CD4 receptor with the one to one stoichiometry. SIGNIFICANCE: Considering observed differences between K(i) and K(d) values indicates clear and rational direction for improving inhibition efficacy to HIV-1 entry and infection. Taken together this report introduces a novel class of natural small molecules fusion inhibitors with nanomolar efficacy of CD4 receptor binding and inhibition of HIV-1 entry.


Subject(s)
CD4 Antigens/metabolism , HIV Envelope Protein gp120/metabolism , HIV-1 , Palmitic Acid/metabolism , Palmitic Acid/pharmacology , CD4 Antigens/chemistry , HIV Envelope Protein gp120/chemistry , HIV Fusion Inhibitors/chemistry , HIV Fusion Inhibitors/metabolism , HIV Fusion Inhibitors/pharmacology , Models, Molecular , Palmitic Acid/chemistry , Protein Binding/drug effects , Protein Conformation
5.
AIDS Res Hum Retroviruses ; 25(12): 1231-41, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20001317

ABSTRACT

The high rate of HIV-1 mutation and the frequent sexual transmission highlight the need for novel therapeutic modalities with broad activity against both CXCR4 (X4) and CCR5 (R5)-tropic viruses. We investigated a large number of natural products, and from Sargassum fusiforme we isolated and identified palmitic acid (PA) as a natural small bioactive molecule with activity against HIV-1 infection. Treatment with 100 microM PA inhibited both X4 and R5 independent infection in the T cell line up to 70%. Treatment with 22 microM PA inhibited X4 infection in primary peripheral blood lymphocytes (PBL) up to 95% and 100 microM PA inhibited R5 infection in primary macrophages by over 90%. Inhibition of infection was concentration dependent, and cell viability for all treatments tested remained above 80%, similar to treatment with 10(-6)M nucleoside analogue 2', 3'-dideoxycytidine (ddC). Micromolar PA concentrations also inhibited cell-to-cell fusion and specific virus-to-cell fusion up to 62%. PA treatment did not result in internalization of the cell surface CD4 receptor or lipid raft disruption, and it did not inhibit intracellular virus replication. PA directly inhibited gp120-CD4 complex formation in a dose-dependent manner. We used fluorescence spectroscopy to determine that PA binds to the CD4 receptor with K(d) approximately 1.5 +/- 0.2 microM, and we used one-dimensional saturation transfer difference NMR (STD-NMR) to determined that the PA binding epitope for CD4 consists of the hydrophobic methyl and methelene groups located away from the PA carboxyl terminal, which blocks efficient gp120-CD4 attachment. These findings introduce a novel class of antiviral compound that binds directly to the CD4 receptor, blocking HIV-1 entry and infection. Understanding the structure-affinity relationship (SAR) between PA and CD4 should lead to the development of PA analogs with greater potency against HIV-1 entry.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Enzyme Inhibitors/pharmacology , HIV Fusion Inhibitors/pharmacology , HIV Infections/metabolism , HIV-1/drug effects , Palmitic Acid/pharmacology , CD4 Antigens/drug effects , CD4 Antigens/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Fusion , Cell Line , Cells, Cultured , Enzyme Inhibitors/chemistry , HIV Fusion Inhibitors/chemistry , HIV Infections/virology , HIV-1/metabolism , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/virology , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Palmitic Acid/chemistry , Receptors, CCR5/drug effects , Receptors, CCR5/metabolism , Receptors, CXCR4/drug effects , Receptors, CXCR4/metabolism , Sargassum/chemistry , Virus Internalization/drug effects , Virus Replication/drug effects , Virus Replication/physiology
6.
Virology ; 380(2): 173-81, 2008 Oct 25.
Article in English | MEDLINE | ID: mdl-18768194

ABSTRACT

In this study we provide evidence that the transcription factor BCL11B represses expression from the HIV-1 long terminal repeat (LTR) in T lymphocytes through direct association with the HIV-1 LTR. We also demonstrate that the NuRD corepressor complex mediates BCL11B transcriptional repression of the HIV-1 LTR. In addition, BCL11B and the NuRD complex repressed TAT-mediated transactivation of the HIV-1 LTR in T lymphocytes, pointing to a potential role in initiation of silencing. In support of all the above results, we demonstrate that BCL11B affects HIV-1 replication and virus production, most likely by blocking LTR transcriptional activity. BCL11B showed specific repression for the HIV-1 LTR sequences isolated from seven different HIV-1 subtypes, demonstrating that it is a general transcriptional repressor for all LTRs.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Viral , HIV Long Terminal Repeat , HIV-1/physiology , Histone Deacetylases/metabolism , Repressor Proteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/virology , Tumor Suppressor Proteins/metabolism , Artificial Gene Fusion , Cell Line , Genes, Reporter , Humans , Luciferases/biosynthesis , Luciferases/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Protein Binding , Virus Replication , tat Gene Products, Human Immunodeficiency Virus/antagonists & inhibitors
7.
Virol J ; 5: 8, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18197976

ABSTRACT

Sargassum fusiforme (Harvey) Setchell has been shown to be a highly effective inhibitor of HIV-1 infection. To identify its mechanism of action, we performed bioactivity-guided fractionation on Sargassum fusiforme mixture. Here, we report isolation of a bioactive fraction SP4-2 (S. fusiforme), which at 8 mug/ml inhibited HIV-1 infection by 86.9%, with IC50 value of 3.7 mug. That represents 230-fold enhancement of antiretroviral potency as compared to the whole extract. Inhibition was mediated against both CXCR4 (X4) and CCR5 (R5) tropic HIV-1. Specifically, 10 mug/ml SP4-2 blocked HIV-1 fusion and entry by 53%. This effect was reversed by interaction of SP4-2 with sCD4, suggesting that S. fusiforme inhibits HIV-1 infection by blocking CD4 receptor, which also explained observed inhibition of both X4 and R5-tropic HIV-1. SP4-2 also inhibited HIV-1 replication after virus entry, by directly inhibiting HIV-1 reverse transcriptase (RT) in a dose dependent manner by up to 79%. We conclude that the SP4-2 fraction contains at least two distinct and biologically active molecules, one that inhibits HIV-1 fusion by interacting with CD4 receptor, and another that directly inhibits HIV-1 RT. We propose that S. fusiforme is a lead candidate for anti-HIV-1 drug development.


Subject(s)
Biological Products/pharmacology , HIV Infections/virology , HIV-1/drug effects , Sargassum , Anti-HIV Agents/pharmacology , CD4 Antigens/drug effects , CD4 Antigens/metabolism , Cell Line , Dose-Response Relationship, Drug , Enzyme Inhibitors , HIV Infections/metabolism , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/physiology , Humans , Receptors, CCR5/metabolism , Receptors, CXCR4/metabolism , Receptors, Virus/metabolism , Virus Attachment/drug effects , Virus Internalization/drug effects
8.
AIDS Res Ther ; 3: 15, 2006 May 25.
Article in English | MEDLINE | ID: mdl-16725040

ABSTRACT

BACKGROUND: The high rate of HIV-1 mutation and increasing resistance to currently available antiretroviral (ART) therapies highlight the need for new antiviral agents. Products derived from natural sources have been shown to inhibit HIV-1 replication during various stages of the virus life cycle, and therefore represent a potential source of novel therapeutic agents. To expand our arsenal of therapeutics against HIV-1 infection, we investigated aqueous extract from Sargassum fusiforme (S. fusiforme) for ability to inhibit HIV-1 infection in the periphery, in T cells and human macrophages, and for ability to inhibit in the central nervous system (CNS), in microglia and astrocytes. RESULTS: S. fusiforme extract blocked HIV-1 infection and replication by over 90% in T cells, human macrophages and microglia, and it also inhibited pseudotyped HIV-1 (VSV/NL4-3) infection in human astrocytes by over 70%. Inhibition was mediated against both CXCR4 (X4) and CCR5 (R5)-tropic HIV-1, was dose dependant and long lasting, did not inhibit cell growth or viability, was not toxic to cells, and was comparable to inhibition by the nucleoside analogue 2', 3'-didoxycytidine (ddC). S. fusiforme treatment blocked direct cell-to-cell infection spread. To investigate at which point of the virus life cycle this inhibition occurs, we infected T cells and CD4-negative primary human astrocytes with HIV-1 pseudotyped with envelope glycoprotein of vesicular stomatitis virus (VSV), which bypasses the HIV receptor requirements. Infection by pseudotyped HIV-1 (VSV/NL4-3) was also inhibited in a dose dependant manner, although up to 57% less, as compared to inhibition of native NL4-3, indicating post-entry interferences. CONCLUSION: This is the first report demonstrating S. fusiforme to be a potent inhibitor of highly productive HIV-1 infection and replication in T cells, in primary human macrophages, microglia, and astrocytes. Results with VSV/NL4-3 infection, suggest inhibition of both entry and post-entry events of the virus life cycle. Absence of cytotoxicity and high viability of treated cells also suggest that S. fusiforme is a potential source of novel naturally occurring antiretroviral compounds that inhibit HIV-1 infection and replication at more than one site of the virus life cycle.

9.
J Neurovirol ; 10 Suppl 1: 25-32, 2004.
Article in English | MEDLINE | ID: mdl-14982736

ABSTRACT

Neurodegeneration and dementia caused by human immunodeficiency virus type 1 (HIV-1) infection of the brain are common complications of acquired immunodeficiency syndrome (AIDS). Introduction of highly active antiretroviral therapy (HAART) reduced the incidence of HIV-1-associated dementia, but so far had no effect on the high frequency of milder neurological disorders caused by HIV-1. This indicates that some neuropathogenic processes persist during limited HIV-1 replication in the central nervous system (CNS). The authors are evaluating the hypothesis that interaction of HIV-1 with astrocytes, which bind HIV-1 but support limited productive HIV-1 infection, may contribute to these processes by disrupting astrocyte functions that are important for neuronal activity or survival. Using laser-capture microdissection on brain tissue samples from HIV-1-infected individuals, we found that HIV-1 DNA can be detected in up to 1% of cortical and basal ganglia astrocytes, thus confirming HIV-1 infection in astrocytes from symptomatic patients. Using rapid subtraction hybridization, the authors cloned and identified 25 messenger RNAs in primary human fetal astrocytes either up-regulated or down-regulated by native HIV-1 infection or exposure to gp120 in vitro. Extending this approach to gene microarray analysis using Affymetrix U133A/B gene chips, the authors determined that HIV-1 alters globally and significantly the overall program of gene expression in astrocytes, including changes in transcripts coding for cytokines, G-coupled protein receptors, transcription factors, and others. Focusing on a specific astrocyte function relevant to neuropathogenesis, the authors showed that exposure of astrocytes to HIV-1 or gp120 in vitro impairs the ability of the cells to transport L-glutamate and the authors related this defect to transcriptional inhibition of the EAAT2 glutamate transporter gene. These findings define new pathways through which HIV-1 may contribute to neuropathogenesis under conditions of limited virus replication in the brain.


Subject(s)
AIDS Dementia Complex/etiology , AIDS Dementia Complex/physiopathology , Astrocytes/physiology , Astrocytes/virology , HIV-1 , Humans , Oligonucleotide Array Sequence Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...