Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
Front Neurol ; 12: 663793, 2021.
Article in English | MEDLINE | ID: mdl-34367046

ABSTRACT

The HIV-1 pandemic is a significant challenge to the field of medicine. Despite advancements in antiretroviral (ART) development, 38 million people worldwide still live with this disease without a cure. A significant barrier to the eradication of HIV-1 lies in the persistently latent pool that establishes early in the infection. The "shock and kill" strategy relies on the discovery of a latency-reversing agent (LRA) that can robustly reactivate the latent pool and not limit immune clearance. We have found that a benzodiazepine (BDZ), that is commonly prescribed for panic and anxiety disorder, to be an ideal candidate for latency reversal. The BDZ Alprazolam functions as an inhibitor of the transcription factor RUNX1, which negatively regulates HIV-1 transcription. In addition to the displacement of RUNX1 from the HIV-1 5'LTR, Alprazolam potentiates the activation of STAT5 and its recruitment to the viral promoter. The activation of STAT5 in cytotoxic T cells may enable immune activation which is independent of the IL-2 receptor. These findings have significance for the potential use of Alprazolam in a curative strategy and to addressing the neuroinflammation associated with neuroHIV-1.

2.
Front Microbiol ; 12: 670016, 2021.
Article in English | MEDLINE | ID: mdl-34122382

ABSTRACT

A unique population of HIV-1 infected individuals can control infection without antiretroviral therapy. These individuals fall into a myriad of categories based on the degree of control (low or undetectable viral load), the durability of control over time and the underlying mechanism (i.e., possession of protective HLA alleles or the absence of critical cell surface receptors). In this study, we examine a cohort of HIV-1 infected individuals with a documented history of sustained low viral loads in the absence of therapy. Through in vitro analyses of cells from these individuals, we have determined that infected individuals with naturally low viral loads are capable of controlling spreading infection in vitro in a CD8+ T-cell dependent manner. This control is lost when viral load is suppressed by antiretroviral therapy and correlates with a clinical CD4:CD8 ratio of <1. Our results support the conclusion that HIV-1 controllers with low, but detectable viral loads may be controlling the virus due to an effective CD8+ T-cell response. Understanding the mechanisms of control in these subjects may provide valuable understanding that could be applied to induce a functional cure in standard progressors.

3.
Methods Mol Biol ; 2175: 23-31, 2020.
Article in English | MEDLINE | ID: mdl-32681481

ABSTRACT

Modifications in chromatin structure are traditionally monitored by biochemical assays that provide average measurements of static events in a population of cells. Microscopy provides a method by which single cells or nuclei can be observed. Traditionally, microscopy has been used to image the nucleus by the application of immunostaining to chemically fixed samples or the use of exogenously expressed fluorescent proteins. This method represents an approach to observe changes in endogenous proteins relating to chromatin structure in real time. Here we describe a method for isolating transcriptionally and enzymatically active nuclei from live cells and visualizing events using fluorescently labeled antibodies. This method allows the observation of real time changes in chromatin architecture and can be used to observe the effects of drugs on nuclei while under microscopic observation.


Subject(s)
Cell Nucleus/metabolism , Chromatin/metabolism , Immunohistochemistry/methods , Microscopy, Confocal/methods , Acetylation , HeLa Cells , Histones/metabolism , Humans , Lamin Type B/metabolism , Nuclear Proteins
4.
PLoS One ; 15(3): e0230563, 2020.
Article in English | MEDLINE | ID: mdl-32210470

ABSTRACT

Despite antiretroviral therapy human immunodeficiency virus type-1 (HIV-1) infection results in neuroinflammation of the central nervous system that can cause HIV-associated neurocognitive disorders (HAND). The molecular mechanisms involved in the development of HAND are unclear, however, they are likely due to both direct and indirect consequences of HIV-1 infection and inflammation of the central nervous system. Additionally, opioid abuse in infected individuals has the potential to exacerbate HIV-comorbidities, such as HAND. Although restricted for productive HIV replication, astrocytes (comprising 40-70% of all brain cells) likely play a significant role in neuropathogenesis in infected individuals due to the production and response of viral proteins. The HIV-1 protein Tat is critical for viral transcription, causes neuroinflammation, and can be secreted from infected cells to affect uninfected bystander cells. The Wnt/ß-catenin signaling cascade plays an integral role in restricting HIV-1 infection in part by negatively regulating HIV-1 Tat function. Conversely, Tat can overcome this negative regulation and inhibit ß-catenin signaling by sequestering the critical transcription factor TCF-4 from binding to ß-catenin. Here, we aimed to explore how opiate exposure affects Tat-mediated suppression of ß-catenin in astrocytes and the downstream modulation of neuroinflammatory genes. We observed that morphine can potentiate Tat suppression of ß-catenin activity in human astrocytes. In contrast, Tat mutants deficient in secretion, and lacking neurotoxic effects, do not affect ß-catenin activity in the presence or absence of morphine. Finally, morphine treatment of astrocytes was sufficient to reduce the expression of genes involved in neuroinflammation. Examining the molecular mechanisms of how HIV-1 infection and opiate exposure exacerbate neuroinflammation may help us inform or predict disease progression prior to HAND development.


Subject(s)
Analgesics, Opioid/adverse effects , HIV Infections/complications , HIV-1/drug effects , Morphine/adverse effects , Neurocognitive Disorders/etiology , Substance-Related Disorders/complications , tat Gene Products, Human Immunodeficiency Virus/immunology , Astrocytes/drug effects , Astrocytes/immunology , Astrocytes/virology , Cell Line , Cells, Cultured , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , Humans , Neurocognitive Disorders/immunology , Neurocognitive Disorders/virology , Substance-Related Disorders/immunology , beta Catenin/immunology
5.
Viruses ; 12(2)2020 02 09.
Article in English | MEDLINE | ID: mdl-32050449

ABSTRACT

Antiretroviral therapy (ART) lowers human immunodeficiency virus type 1 (HIV-1) viral load to undetectable levels, but does not eliminate the latent reservoir. One of the factors controlling the latent reservoir is transcriptional silencing of the integrated HIV-1 long terminal repeat (LTR). The molecular mechanisms that control HIV-1 transcription are not completely understood. We have previously shown that RUNX1, a host transcription factor, may play a role in the establishment and maintenance of HIV-1 latency. Prior work has demonstrated that inhibition of RUNX1 by the benzodiazepine (BDZ) Ro5-3335 synergizes with suberanilohydroxamic acid (SAHA) to activate HIV-1 transcription. In this current work, we examine the effect of RUNX1 inhibition on the chromatin state of the integrated HIV-1 LTR. Using chromatin immunoprecipitation (ChIP), we found that Ro5-3335 significantly increased the occupancy of STAT5 at the HIV-1 LTR. We also screened other BDZs for their ability to regulate HIV-1 transcription and demonstrate their ability to increase transcription and alter chromatin at the LTR without negatively affecting Tat activity. These findings shed further light on the mechanism by which RUNX proteins control HIV-1 transcription and suggest that BDZ compounds might be useful in activating HIV-1 transcription through STAT5 recruitment to the HIV-1 LTR.


Subject(s)
Benzodiazepines/pharmacology , Chromatin/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , HIV Long Terminal Repeat/genetics , Transcription, Genetic/drug effects , Virus Integration , Chromatin Immunoprecipitation , Core Binding Factor Alpha 2 Subunit/antagonists & inhibitors , Gene Expression Regulation , HIV-1 , Humans , Leukocytes, Mononuclear/virology , STAT5 Transcription Factor/genetics
6.
Sci Rep ; 10(1): 1821, 2020 02 04.
Article in English | MEDLINE | ID: mdl-32020017

ABSTRACT

Chemoresistance is one of the leading causes of mortality in breast cancer (BC). Understanding the molecules regulating chemoresistance is critical in order to combat chemoresistant BC. Drug efflux pump ABCB1 is overexpressed in chemoresistant neoplasms where it effluxes various chemotherapeutic agents from cells. Because it is expressed in normal and cancerous cells alike, attempts at targeting ABCB1 directly have failed due to low specificity and disruption of normal tissue. A proposed method to inhibit ABCB1 is to target its cancer-specific, upstream regulators, mitigating damage to normal tissue. Few such cancer-specific upstream regulators have been described. Here we characterize ROR1 as an upstream regulator of ABCB1. ROR1 is highly expressed during development but not expressed in normal adult tissue. It is however highly expressed in several cancers. ROR1 is overexpressed in chemoresistant BC where it correlates with poor therapy response and tumor recurrence. Our data suggests, ROR1 inhibition sensitizes BC cells to chemo drugs. We also show ROR1 regulates ABCB1 stability and transcription via MAPK/ERK and p53. Validating our overall findings, inhibition of ROR1 directly correlated with decreased efflux of chemo-drugs from cells. Overall, our results highlight ROR1's potential as a therapeutic target for multidrug resistant malignancies.


Subject(s)
Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Breast Neoplasms/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation , Doxorubicin/metabolism , Doxorubicin/therapeutic use , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , MAP Kinase Signaling System , Real-Time Polymerase Chain Reaction
7.
PLoS One ; 14(5): e0217789, 2019.
Article in English | MEDLINE | ID: mdl-31150511

ABSTRACT

Triple Negative Breast Cancer (TNBC), the most aggressive subtype of breast cancer, is characterized by the absence of hormone receptors usually targeted by hormone therapies like Tamoxifen. Because therapy success and survival rates for TNBC lag far behind other breast cancer subtypes, there is significant interest in developing novel anti-TNBC agents that can target TNBC specifically, with minimal effects on non-malignant tissue. To this aim, our study describes the anti-TNBC effect of strictinin, an ellagitanin previously isolated from Myrothamnus flabellifolius. Using various in silico and molecular techniques, we characterized the mechanism of action of strictinin in TNBC. Our results suggest strictinin interacts strongly with Receptor Tyrosine Kinase Orphan like 1 (ROR1). ROR1 is an oncofetal receptor highly expressed during development but not in normal adult tissue. It is highly expressed in several human malignancies however, owing to its numerous pro-tumor functions. Via its interaction and inhibition of ROR1, strictinin reduced AKT phosphorylation on ser-473, inhibiting downstream phosphorylation and inhibition of GSK3ß. The reduction in AKT phosphorylation also correlated with decreased cell survival and activation of the caspase-mediated intrinsic apoptotic cascade. Strictinin treatment also repressed cell migration and invasion in a beta-catenin independent manner, presumably via the reactivated GSK3ß's repressing effect on microtubule polymerization and focal adhesion turnover. This could be of potential therapeutic interest considering heightened interest in ROR1 and other receptor tyrosine kinases as targets for development of anti-cancer agents. Further studies are needed to validate these findings in other ROR1-expressing malignancies but also in more systemic models of TNBC. Our findings do however underline the potential of strictinin and other ROR1-targeting agents as therapeutic tools to reduce TNBC proliferation, survival and motility.


Subject(s)
Cell Proliferation/drug effects , Phenols/pharmacology , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Triple Negative Breast Neoplasms/drug therapy , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Glycogen Synthase Kinase 3 beta/genetics , Humans , Oncogene Protein v-akt/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation/drug effects , Receptor Tyrosine Kinase-like Orphan Receptors/antagonists & inhibitors , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
8.
Traffic ; 2018 Apr 30.
Article in English | MEDLINE | ID: mdl-29708629

ABSTRACT

The human immunodeficiency virus type 1 (HIV-1) transactivator of transcription (Tat) protein functions both intracellularly and extracellularly. Intracellularly, the main function is to enhance transcription of the viral promoter. However, this process only requires a small amount of intracellular Tat. The majority of Tat is secreted through an unconventional mechanism by binding to phosphatidylinositol-4,5-bisphosphate (PtdIns(4,5)P2 ), a phospholipid in the inner leaflet of the plasma membrane that is required for secretion. This interaction is mediated by the basic domain of Tat (residues 48-57) and a conserved tryptophan (residue 11). After binding to PtdIns(4,5)P2 , Tat secretion diverges into multiple pathways, which we categorized as oligomerization-mediated pore formation, spontaneous translocation and incorporation into exosomes. Extracellular Tat has been shown to be neurotoxic and toxic to other cells of the central nervous system (CNS) and periphery, able to recruit immune cells to the CNS and cerebrospinal fluid, and alter the gene expression and morphology of uninfected cells. The effects of extracellular Tat have been examined in HIV-1-associated neurocognitive disorders (HAND); however, only a small number of studies have focused on the mechanisms underlying Tat secretion. In this review, the molecular mechanisms of Tat secretion will be examined in a variety of biologically relevant cell types.

9.
Sci Rep ; 8(1): 5916, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29651043

ABSTRACT

Despite numerous recent advances in imaging technologies, one continuing challenge for cell biologists and microscopists is the visualization and measurement of endogenous proteins as they function within living cells. Achieving this goal will provide a tool that investigators can use to associate cellular outcomes with the behavior and activity of many well-studied target proteins. Here, we describe the development of a plasmid-based fluorescent biosensor engineered to measure the location and activity of matrix metalloprotease-14 (MMP14). The biosensor design uses fluorogen-activating protein technology coupled with a MMP14-selective protease sequence to generate a binary, "switch-on" fluorescence reporter capable of measuring MMP14 location, activity, and temporal dynamics. The MMP14-fluorogen activating protein biosensor approach is applicable to both short and long-term imaging modalities and contains an adaptable module that can be used to study many membrane-bound proteases. This MMP14 biosensor promises to serve as a tool for the advancement of a broad range of investigations targeting MMP14 activity during cell migration in health and disease.


Subject(s)
Biosensing Techniques , Cell Membrane/genetics , Matrix Metalloproteinase 14/isolation & purification , Cell Membrane/chemistry , Cell Movement/genetics , Fluorescence , Humans , Matrix Metalloproteinase 14/chemistry , Protein Binding/genetics , Surface Properties
10.
Am J Primatol ; 80(1)2018 01.
Article in English | MEDLINE | ID: mdl-26676710

ABSTRACT

Enteric dysbiosis is a characteristic feature of progressive human immunodeficiency virus type 1 (HIV-1) infection but has not been observed in simian immunodeficiency virus (SIVmac)-infected macaques, including in animals with end-stage disease. This has raised questions concerning the mechanisms underlying the HIV-1 associated enteropathy, with factors other than virus infection, such as lifestyle and antibiotic use, implicated as playing possible causal roles. Simian immunodeficiency virus of chimpanzees (SIVcpz) is also associated with increased mortality in wild-living communities, and like HIV-1 and SIVmac, can cause CD4+ T cell depletion and immunodeficiency in infected individuals. Given the central role of the intestinal microbiome in mammalian health, we asked whether gut microbial constituents could be identified that are indicative of SIVcpz status and/or disease progression. Here, we characterized the gut microbiome of SIVcpz-infected and -uninfected chimpanzees in Gombe National Park, Tanzania. Subjecting a small number of fecal samples (N = 9) to metagenomic (shotgun) sequencing, we found bacteria of the family Prevotellaceae to be enriched in SIVcpz-infected chimpanzees. However, 16S rRNA gene sequencing of a larger number of samples (N = 123) failed to show significant differences in both the composition and diversity (alpha and beta) of gut bacterial communities between infected (N = 24) and uninfected (N = 26) chimpanzees. Similarly, chimpanzee stool-associated circular virus (Chi-SCV) and chimpanzee adenovirus (ChAdV) identified by metagenomic sequencing were neither more prevalent nor more abundant in SIVcpz-infected individuals. However, fecal samples collected from SIVcpz-infected chimpanzees within 5 months before their AIDS-related death exhibited significant compositional changes in their gut bacteriome. These data indicate that SIVcpz-infected chimpanzees retain a stable gut microbiome throughout much of their natural infection course, with a significant destabilization of bacterial (but not viral) communities observed only in individuals with known immunodeficiency within the last several months before their death. Am. J. Primatol. 80:e22515, 2018. © 2015 Wiley Periodicals, Inc.


Subject(s)
Ape Diseases/microbiology , Bacteria/classification , Gastrointestinal Microbiome , Pan troglodytes , Simian Acquired Immunodeficiency Syndrome/microbiology , Adenoviruses, Simian/genetics , Animals , Ape Diseases/virology , Bacteria/genetics , DNA Viruses/genetics , Feces/microbiology , Feces/virology , Female , Male , Metagenome , RNA, Ribosomal, 16S , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus , Tanzania
11.
J Cell Sci ; 130(17): 2926-2940, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28743737

ABSTRACT

Chromatin modification is traditionally assessed in biochemical assays that provide average measurements of static events given that the analysis requires components from many cells. Microscopy can visualize single cells, but the cell body and organelles can hamper staining and visualization of the nucleus. Normally, chromatin is visualized by immunostaining a fixed sample or by expressing exogenous fluorescently tagged proteins in a live cell. Alternative microscopy tools to observe changes of endogenous chromatin in real-time are needed. Here, we isolated transcriptionally competent nuclei from cells and used antibody staining without fixation to visualize changes in endogenous chromatin. This method allows the real-time addition of drugs and fluorescent probes to one or more nuclei while under microscopy observation. A high-resolution map of 11 endogenous nuclear markers of the histone code, transcription machinery and architecture was obtained in transcriptionally active nuclei by performing confocal and structured illumination microscopy. We detected changes in chromatin modification and localization at the single-nucleus level after inhibition of histone deacetylation. Applications in the study of RNA transcription, viral protein function and nuclear architecture are presented. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Chromatin/metabolism , Acetylation , Computer Systems , HeLa Cells , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Imaging, Three-Dimensional , Lysine/metabolism , Microscopy , Nuclear Lamina/metabolism , Nuclear Proteins/metabolism , Nucleic Acids/metabolism , RNA/genetics , RNA/metabolism , Time-Lapse Imaging , Transcription, Genetic , tat Gene Products, Human Immunodeficiency Virus/metabolism
12.
PLoS Negl Trop Dis ; 10(8): e0004877, 2016 08.
Article in English | MEDLINE | ID: mdl-27560129

ABSTRACT

The ongoing Zika virus epidemic in the Americas and the observed association with both fetal abnormalities (primary microcephaly) and adult autoimmune pathology (Guillain-Barré syndrome) has brought attention to this neglected pathogen. While initial case studies generated significant interest in the Zika virus outbreak, larger prospective epidemiology and basic virology studies examining the mechanisms of Zika viral infection and associated pathophysiology are only now starting to be published. In this review, we analyze Zika fetal neuropathogenesis from a comparative pathology perspective, using the historic metaphor of "TORCH" viral pathogenesis to provide context. By drawing parallels to other viral infections of the fetus, we identify common themes and mechanisms that may illuminate the observed pathology. The existing data on the susceptibility of various cells to both Zika and other flavivirus infections are summarized. Finally, we highlight relevant aspects of the known molecular mechanisms of flavivirus replication.


Subject(s)
Brain/virology , Fetal Diseases/virology , Fetus/virology , Microcephaly/virology , Zika Virus Infection/virology , Zika Virus/pathogenicity , Adult , Americas/epidemiology , Brain/pathology , Disease Outbreaks , Epidemics , Fetal Diseases/pathology , Flavivirus Infections/epidemiology , Flavivirus Infections/virology , Genome, Viral , Guillain-Barre Syndrome/epidemiology , Guillain-Barre Syndrome/virology , Humans , Microcephaly/economics , Virus Replication , Zika Virus/genetics , Zika Virus/physiology , Zika Virus Infection/complications , Zika Virus Infection/physiopathology
13.
Retrovirology ; 13: 25, 2016 Apr 08.
Article in English | MEDLINE | ID: mdl-27060080

ABSTRACT

BACKGROUND: Long term infection with HIV-1, even in the context of therapy, leads to chronic health problems including an array of neurocognitive dysfunctions. The viral Tat protein has previously been implicated in neuropathogenesis through its effect on astrocytes. Tat has also been shown to inhibit the biogenesis of miRNAs by inhibiting the activity of the cellular Dicer protein in an RNA dependent fashion. Whether there is a mechanistic connection between the ability of HIV-1 Tat to alter miRNAs and its observed effects on cells of the central nervous system has not been well examined. RESULTS: Here, we examined the ability of HIV-1 Tat to bind to and inhibit the production of over 300 cellular miRNAs. We found that the Tat protein only binds to and inhibits a fraction of the total cellular miRNAs. By mapping the downstream targets of these miRNAs we have determined a possible role for Tat alterations of miRNAs in the development of neuropathogenesis. Specifically, this work points to suppression of miRNAs function as the mechanism for Tat suppression of ß-catenin activity. CONCLUSIONS: The discovery that HIV-1 Tat inhibits only a fraction of miRNAs opens new areas of research regarding changes in cellular pathways through suppression of RNA interference. Our initial analysis strongly suggests that these pathways may contribute to HIV-1 disruption of the central nervous system.


Subject(s)
Astrocytes/virology , HIV-1/physiology , Host-Pathogen Interactions , MicroRNAs/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , tat Gene Products, Human Immunodeficiency Virus/metabolism , Cells, Cultured , Humans , Protein Binding , RNA-Binding Proteins/metabolism
14.
Mol Ther ; 24(3): 416-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26952919
15.
J Biol Chem ; 291(3): 1251-66, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26553869

ABSTRACT

HIV-1 infection results in a chronic illness because long-term highly active antiretroviral therapy can lower viral titers to an undetectable level. However, discontinuation of therapy rapidly increases virus burden. Moreover, patients under highly active antiretroviral therapy frequently develop various metabolic disorders, neurocognitive abnormalities, and cardiovascular diseases. We have previously shown that exosomes containing trans-activating response (TAR) element RNA enhance susceptibility of undifferentiated naive cells to HIV-1 infection. This study indicates that exosomes from HIV-1-infected primary cells are highly abundant with TAR RNA as detected by RT-real time PCR. Interestingly, up to a million copies of TAR RNA/µl were also detected in the serum from HIV-1-infected humanized mice suggesting that TAR RNA may be stable in vivo. Incubation of exosomes from HIV-1-infected cells with primary macrophages resulted in a dramatic increase of proinflammatory cytokines, IL-6 and TNF-ß, indicating that exosomes containing TAR RNA could play a direct role in control of cytokine gene expression. The intact TAR molecule was able to bind to PKR and TLR3 effectively, whereas the 5' and 3' stems (TAR microRNAs) bound best to TLR7 and -8 and none to PKR. Binding of TAR to PKR did not result in its phosphorylation, and therefore, TAR may be a dominant negative decoy molecule in cells. The TLR binding through either TAR RNA or TAR microRNA potentially can activate the NF-κB pathway and regulate cytokine expression. Collectively, these results imply that exosomes containing TAR RNA could directly affect the proinflammatory cytokine gene expression and may explain a possible mechanism of inflammation observed in HIV-1-infected patients under cART.


Subject(s)
Activating Transcription Factors/metabolism , Cytokines/metabolism , Exosomes/metabolism , HIV-1/immunology , Leukocytes/metabolism , MicroRNAs/metabolism , Active Transport, Cell Nucleus , Animals , Cell Line , Cell Line, Transformed , Cell Transformation, Viral , Cells, Cultured , Exosomes/immunology , Exosomes/virology , HIV Infections/blood , HIV Infections/immunology , HIV Infections/virology , Humans , Interleukin-6/metabolism , Leukocytes/immunology , Leukocytes/virology , Lymphotoxin-alpha/metabolism , Mice, Inbred NOD , Mice, Transgenic , MicroRNAs/blood , Toll-Like Receptor 3/antagonists & inhibitors , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
17.
Immunity ; 41(3): 493-502, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25238099

ABSTRACT

The viral accessory protein Vpx, expressed by certain simian and human immunodeficiency viruses (SIVs and HIVs), is thought to improve viral infectivity of myeloid cells. We infected 35 Asian macaques and African green monkeys with viruses that do or do not express Vpx and examined viral targeting of cells in vivo. While lack of Vpx expression affected viral dynamics in vivo, with decreased viral loads and infection of CD4⁺ T cells, Vpx expression had no detectable effect on infectivity of myeloid cells. Moreover, viral DNA was observed only within myeloid cells in tissues not massively depleted of CD4⁺ T cells. Myeloid cells containing viral DNA also showed evidence of T cell phagocytosis in vivo, suggesting that their viral DNA may be attributed to phagocytosis of SIV-infected T cells. These data suggest that myeloid cells are not a major source of SIV in vivo, irrespective of Vpx expression.


Subject(s)
CD4-Positive T-Lymphocytes/virology , DNA, Viral/analysis , Myeloid Cells/virology , Simian Immunodeficiency Virus/genetics , Viral Regulatory and Accessory Proteins/biosynthesis , Animals , CD4-Positive T-Lymphocytes/immunology , Chlorocebus aethiops , Lymphocyte Depletion , Macaca , Monomeric GTP-Binding Proteins/biosynthesis , Phagocytosis , Simian Acquired Immunodeficiency Syndrome , Viral Load , Viral Regulatory and Accessory Proteins/genetics
18.
PLoS Pathog ; 10(3): e1003997, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24651404

ABSTRACT

A major barrier to the elimination of HIV-1 infection is the presence of a pool of long-lived, latently infected CD4+ memory T-cells. The search for treatments to re-activate latent HIV to aid in clearance is hindered by the incomplete understanding of the mechanisms that lead to transcriptional silencing of viral gene expression in host cells. Here we identify a previously unknown role for RUNX1 in HIV-1 transcriptional latency. The RUNX proteins, in combination with the co-factor CBF-ß, are critical transcriptional regulators in T-cells. RUNX1 strongly modulates CD4 expression and contributes to CD4+ T-cell function. We show that RUNX1 can bind DNA sequences within the HIV-1 LTR and that this binding represses transcription. Using patient samples we show a negative correlation between RUNX1 expression and viral load. Furthermore, we find that pharmacologic inhibition of RUNX1 by a small molecule inhibitor, Ro5-3335, synergizes with the histone deacetylase (HDAC) inhibitor SAHA (Vorinostat) to enhance the activation of latent HIV-1 in both cell lines and PBMCs from patients. Our findings indicate that RUNX1 and CBF-ß cooperate in cells to modulate HIV-1 replication, identifying for the first time RUNX1 as a cellular factor involved in HIV-1 latency. This work highlights the therapeutic potential of inhibitors of RUNX1 to re-activate virus and aid in clearance of HIV-1.


Subject(s)
Core Binding Factor Alpha 2 Subunit/antagonists & inhibitors , HIV Infections/virology , Virus Activation/physiology , Virus Latency/physiology , CCAAT-Binding Factor/metabolism , Chromatin Immunoprecipitation , Drug Synergism , Flow Cytometry , HIV-1/physiology , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Microscopy, Confocal , Mutagenesis, Site-Directed , Reverse Transcriptase Polymerase Chain Reaction , Viral Load , Vorinostat
19.
Methods Mol Biol ; 1087: 285-96, 2014.
Article in English | MEDLINE | ID: mdl-24158831

ABSTRACT

Quantitative PCR (qPCR) provides a robust method for quantifying DNA species. By combining modern qPCR techniques with the isolation of small RNA, the polyadenylation of the RNA, and the use of reverse transcriptase to create miRNA derived cDNA, it is now possible to use qPCR to quantify miRNA. This method is scalable and provides a useful addition to the retrovirologists' toolbox. Here, we also describe the use of one-LTR infectious molecular clones to verify miRNA target sites within the retroviral LTR.


Subject(s)
HIV-1/genetics , HIV-1/physiology , MicroRNAs/analysis , MicroRNAs/genetics , Poly A/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Terminal Repeat Sequences/genetics , Cloning, Molecular , HEK293 Cells , Humans , MicroRNAs/isolation & purification , Reproducibility of Results
20.
Virology ; 443(1): 106-12, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23747197

ABSTRACT

HIV-1 and HTLV-1 can infect CD4+ T cells and can co-infect the same individual. In principle, it is possible that both viruses can infect the same CD4+ T cells in dually infected persons. Currently, how efficiently HTLV-1 and HIV-1 co-infects the same cell and the full extent of their biological interactions are not well-understood. Here, we report evidence confirming that both viruses can infect the same cells and that HTLV-1 envelope (Env) can pseudotype HIV-1 viral particles and HIV-1 envelope (Env) can pseudotype HTLV-1 virions to mediate subsequent infections of substrate cells. We also show that the construction of a chimeric HTLV-1 molecular clone carrying the HIV-1 Env in place of its HTLV-1 counterpart results in a replication competent moiety. These findings raise new implications of viral complementation and assortment between HIV-1 and HTLV-1 in dually infected persons.


Subject(s)
HIV-1/physiology , Human T-lymphotropic virus 1/physiology , Viral Envelope Proteins/metabolism , Virus Replication , Cell Line , HIV-1/genetics , Human T-lymphotropic virus 1/genetics , Humans , T-Lymphocytes/virology , Viral Envelope Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...