Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nat Commun ; 10(1): 1133, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30850604

ABSTRACT

Genome editing for therapeutic applications often requires cleavage within a narrow sequence window. Here, to enable such high-precision targeting with zinc-finger nucleases (ZFNs), we have developed an expanded set of architectures that collectively increase the configurational options available for design by a factor of 64. These new architectures feature the functional attachment of the FokI cleavage domain to the amino terminus of one or both zinc-finger proteins (ZFPs) in the ZFN dimer, as well as the option to skip bases between the target triplets of otherwise adjacent fingers in each zinc-finger array. Using our new architectures, we demonstrate targeting of an arbitrarily chosen 28 bp genomic locus at a density that approaches 1.0 (i.e., efficient ZFNs available for targeting almost every base step). We show that these new architectures may be used for targeting three loci of therapeutic significance with a high degree of precision, efficiency, and specificity.


Subject(s)
Deoxyribonucleases, Type II Site-Specific/genetics , Gene Editing/methods , Genome, Human , Protein Engineering/methods , Zinc Finger Nucleases/genetics , Base Pairing , Base Sequence , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Deoxyribonucleases, Type II Site-Specific/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Genetic Loci , Genomic Library , Humans , INDEL Mutation , K562 Cells , Peptide Library , Plasmids/chemistry , Plasmids/metabolism , Transformation, Genetic , Viral Proteins/genetics , Viral Proteins/metabolism , Zinc Finger Nucleases/metabolism
2.
Mol Ther ; 23(8): 1380-1390, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25939491

ABSTRACT

Programmed cell death-1 (PD-1) is expressed on activated T cells and represents an attractive target for gene-editing of tumor targeted T cells prior to adoptive cell transfer (ACT). We used zinc finger nucleases (ZFNs) directed against the gene encoding human PD-1 (PDCD-1) to gene-edit melanoma tumor infiltrating lymphocytes (TIL). We show that our clinical scale TIL production process yielded efficient modification of the PD-1 gene locus, with an average modification frequency of 74.8% (n = 3, range 69.9-84.1%) of the alleles in a bulk TIL population, which resulted in a 76% reduction in PD-1 surface-expression. Forty to 48% of PD-1 gene-edited cells had biallelic PD-1 modification. Importantly, the PD-1 gene-edited TIL product showed improved in vitro effector function and a significantly increased polyfunctional cytokine profile (TNFα, GM-CSF, and IFNγ) compared to unmodified TIL in two of the three donors tested. In addition, all donor cells displayed an effector memory phenotype and expanded approximately 500-2,000-fold in vitro. Thus, further study to determine the efficiency and safety of adoptive cell transfer using PD-1 gene-edited TIL for the treatment of metastatic melanoma is warranted.


Subject(s)
Endoribonucleases/genetics , Gene Expression Regulation, Neoplastic , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/therapy , Programmed Cell Death 1 Receptor/genetics , Zinc Fingers , Alleles , Animals , Cell Separation , Cytokines/metabolism , Female , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Green Fluorescent Proteins/metabolism , Humans , Immunologic Memory , Immunotherapy, Adoptive , Interferon-gamma/metabolism , Lymphocyte Activation/immunology , Mice , Neoplasm Metastasis , Neoplasm Transplantation , Phenotype , Programmed Cell Death 1 Receptor/metabolism , Tumor Necrosis Factor-alpha/metabolism
3.
J Basic Clin Physiol Pharmacol ; 23(4): 139-46, 2012.
Article in English | MEDLINE | ID: mdl-23072849

ABSTRACT

BACKGROUND: Many plant-derived products exhibit potent chemopreventive activity against animal tumor models as well as rodent and human cancer cell lines. They have low side effects and toxicity and presumably modulate the factors that are critical for cell proliferation, differentiation, senescence and apoptosis. The present study investigates the effects of some medicinal plant extracts from generally recognized as safe plants that may be useful in the prevention and treatment of cancer. METHODS: Clonogenic assays using logarithmically-growing cells were performed to test the effect. The cytotoxic effects of Curcuma longa and Zingiber officinale were studied using sulforhodamine B assay, tetrazolium dye assay, colony morphology and microscopic analysis. RESULTS: Out of the 13 lyophilized plant-derived extracts evaluated for growth-inhibitory effects on the PC-3M prostate cancer cell line, two extracts derived from C. longa and Z. officinale showed significant inhibitory effects on colony-forming ability. The individual and augmentative effects of these two extracts were tested for their narrow range effective lower concentration on PC-3M in clonogenic assays. At relatively lower concentrations, C. longa showed significant inhibition of colony formation in clonogenic assays; whereas at same concentrations Z. officinale showed only moderate inhibitory effects. However, when both the agents were tested together at the same concentrations, the combined effects were much more significant than their individual ones. On normal prostate epithelial cells both C. longa and Z. officinale had similar effects but at a lower magnitude. These observations were confirmed by several cytotoxicity assays involving the morphological appearance of the colonies, microscopic observations, per cent inhibition in comparison to control by sulforhodamine B and tetrazolium dye assay. CONCLUSIONS: From these observations, it was concluded that the combined effects of C. longa and Z. officinale are much greater than their individual effects, suggesting the role of multiple components and their synergistic mode of actions to elicit stronger beneficial effects.


Subject(s)
Curcuma , Phytotherapy , Plant Extracts/pharmacology , Prostatic Neoplasms/drug therapy , Zingiber officinale , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Humans , Male , Plant Extracts/administration & dosage , Prostatic Neoplasms/pathology
4.
Neurochem Int ; 61(5): 807-14, 2012 Oct.
Article in English | MEDLINE | ID: mdl-21756955

ABSTRACT

HIV infection affects the central nervous system resulting in HIV associated neurocognitive disorder (HAND), which is characterized by depression, behavioral and motor dysfunctions. The HIV-1 viral envelope protein gp120 is known to induce the release of neurotoxic factors which lead to apoptotic cell death. Although the exact mechanisms involved in HIV-1 gp120-induced neurotoxicity are not completely understood, oxidative stress is suggested to play a vital role in the neuropathogenesis of HAND. Astrocytes represent major population of the non-neuronal cell type in the brain and play a critical role in the neuropathogenesis of HAND. Increased oxidative stress is known to induce nuclear factor erythroid derived 2-related factor 2 (Nrf2), a basic leucine zipper transcription factor which is known to regulate the antioxidant defensive mechanism. However, the role of Nrf2 in HAND has not been elucidated. We report that gp120 significantly upregulates Nrf2 in human astrocytes and is associated with stimulation of key antioxidant defensive enzymes Hemoxygenase (HO-1) and NAD(P)H dehydrogenase quinone1 (Nqo1). Pretreatment of the astrocytes with antioxidants or a specific calcium chelator BAPTA-AM, significantly blocked the upregulation of Nrf2, HO-1 and Nqo1. These results suggest a possible role of the intracellular calcium and oxidative stress in Nrf2 mediated antioxidant defense mechanism, which may have protective role in promoting cell survival.


Subject(s)
AIDS Dementia Complex/metabolism , Antioxidant Response Elements/physiology , Astrocytes/metabolism , Gene Expression Regulation, Viral , HIV Envelope Protein gp120/physiology , NF-E2-Related Factor 2/biosynthesis , AIDS Dementia Complex/virology , Cells, Cultured , Humans , NF-E2-Related Factor 2/physiology , Oxidative Stress/physiology
5.
J Neurovirol ; 17(3): 230-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21491143

ABSTRACT

HIV-1 clades (subtypes) differentially contribute to the neuropathogenesis of HIV-associated dementia (HAD) in neuroAIDS. HIV-1 envelop protein, gp120, plays a major role in neuronal function. It is not well understood how these HIV-1 clades exert these neuropathogenic differences. The N-methyl-D: -aspartate (NMDA) receptor-reduced glutamine synthesis could lead to secretion of neurotoxins such as arachidonic acid (AA) which plays a significant role in the neuropathogenic mechanisms in neuroAIDS. We hypothesize that clade B and C gp120 proteins exert differential effects on human primary astrocytes by production of the neurotoxin arachidonic acid. Our results indicate that clade B gp120 significantly downregulated NMDA receptor gene and protein expression, and level of glutamine while increasing expression of prostaglandin E2 (PGE(2)) and thromboxane A2 receptor (TBXA(2) R) compared to HIV-1 clade C gp120 protein. Thus, our studies for the first time demonstrate that HIV-1 clade B-gp120 protein appears to induce higher levels of expression of the neuropathogenic molecule cyclooxygenase-2 (COX-2)-mediated arachidonic acid by-products, PGE(2), and TBXA(2) R compared to HIV-1 clade C gp120 protein. These studies suggest that HIV-1 clade B and C gp120 proteins may play a differential role in the neuropathogenesis of HAD in neuroAIDS.


Subject(s)
AIDS Dementia Complex/metabolism , Arachidonic Acid/biosynthesis , Astrocytes/drug effects , HIV Envelope Protein gp120/pharmacology , HIV Infections/metabolism , Neurotoxins/biosynthesis , Protein Isoforms/pharmacology , AIDS Dementia Complex/pathology , Astrocytes/metabolism , Astrocytes/pathology , Astrocytes/virology , Cell Culture Techniques , Cells, Cultured , Cyclooxygenase 2/metabolism , Dinoprostone/biosynthesis , Down-Regulation , Glutamine/biosynthesis , HIV Envelope Protein gp120/metabolism , HIV Infections/pathology , HIV-1/physiology , Humans , Protein Isoforms/metabolism , Receptors, N-Methyl-D-Aspartate/biosynthesis , Receptors, Thromboxane A2, Prostaglandin H2/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
6.
Alcohol Clin Exp Res ; 35(8): 1550-6, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21447001

ABSTRACT

BACKGROUND: Previous studies have implicated histone deacetylases (HDACs) and HDAC inhibitors (HDIs) such as trichostatin A (TSA) in the regulation of gene expression during drug addiction. Furthermore, an increase in HDAC activity has been linked to neurodegeneration. Alcohol has also been shown to promote abundant generation of reactive oxygen species (ROS) resulting in oxidative stress. TSA inhibits HDACs and has been shown to be neuroprotective in other neurodegenerative disease models. Although HDACs and HDIs have been associated with drug addiction, there is no evidence of the neurodegenerative role of HDAC2 and neuroprotective role of TSA in alcohol addiction. Therefore, we hypothesize that alcohol modulates HDAC2 through mechanisms involving oxidative stress. METHODS: To test our hypothesis, the human neuronal cell line, SK-N-MC, was treated with different concentrations of ethanol (EtOH); HDAC2 gene and protein expression were assessed at different time points. Pharmacological inhibition of HDAC2 with TSA was evaluated at the gene level using qRT-PCR and at the protein level using Western blot and flow cytometry. ROS production was measured with a fluorescence microplate reader and fluorescence microscopy. RESULTS: Our results showed a dose-dependent increase in HDAC2 expression with EtOH treatment. Additionally, alcohol significantly induced ROS, and pharmacological inhibition of HDAC2 with TSA was shown to be neuroprotective by significantly inhibiting HDAC2 and ROS. CONCLUSIONS: These results suggest that EtOH can upregulate HDAC2 through mechanisms involving oxidative stress and HDACs may play an important role in alcohol use disorders (AUDs). Moreover, the use of HDIs may be of therapeutic significance for the treatment of neurodegenerative disorders including AUDs.


Subject(s)
Central Nervous System Depressants/toxicity , Ethanol/toxicity , Histone Deacetylase 2/metabolism , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Neuroprotective Agents/pharmacology , Cell Line , Dose-Response Relationship, Drug , Gene Expression/drug effects , Histone Deacetylase 2/drug effects , Histone Deacetylase 2/genetics , Humans , Hydroxamic Acids/metabolism , Neuroprotective Agents/metabolism , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism
7.
Neurochem Int ; 58(6): 656-64, 2011 May.
Article in English | MEDLINE | ID: mdl-21315782

ABSTRACT

Histone deacetylases (HDACs) play a pivotal role in epigenetic regulation of transcription and homeostasis of protein acetylation in histones and other proteins involved in chromatin remodeling. Histone hypoacetylation and transcriptional dysfunction have been shown to be associated with a variety of neurodegenerative diseases. More recently, neuron specific overexpression of HDAC2 has been shown to modulate synaptic plasticity and learning behavior in mice. However, the role of HDAC2 in development of HIV-associated neurocognitive disorders (HAND) is not reported. Herein we report that HIV-1 Tat protein upregulate HDAC2 expression in neuronal cells leading to transcriptional repression of genes involved in synaptic plasticity and neuronal function thereby contributing to the progression of HAND. Our results indicate upregulation of HDAC2 by Tat treatment in dose and time dependant manner by human neuroblastoma SK-N-MC cells and primary human neurons. Further, HDAC2 overexpression was associated with concomitant downregulation in CREB and CaMKIIa genes that are known to regulate neuronal activity. These observed effects were completely blocked by HDAC2 inhibition. These results for the first time suggest the possible role of HDAC2 in development of HAND. Therefore, use of HDAC2 specific inhibitor in combination with HAART may be of therapeutic value in treatment of neurocognitive disorders observed in HIV-1 infected individuals.


Subject(s)
AIDS Dementia Complex/enzymology , Gene Products, tat/physiology , HIV-1/metabolism , Histone Deacetylase 2/metabolism , Neurons/enzymology , Cell Line, Tumor , Flow Cytometry , Gene Products, tat/antagonists & inhibitors , Humans , Hydroxamic Acids/pharmacology , Polymerase Chain Reaction , Up-Regulation
8.
J Neurovirol ; 16(4): 255-63, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20602605

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) is commonly associated with immune dysfunctions and the suppression of antigen-presenting cells. This results in immune alterations, which could lead to impaired neuronal functions, such as neuroAIDS. The neurotoxic factor kynurenine (KYN), the rate-limiting enzyme indoleamine 2,3-dioxygenase (IDO), serotonin (5-HT), and serotonin transporter (5-HTT) may play a role in tryptophan deficiency and serotogenic dysfunction in neuroAIDS. HIV-1 transactivator regulatory protein (Tat) is known to play a major role in immune dysfunction. Previous studies suggest that HIV-1 B and C clades differentially manifest neuronal dysfunctions in the infected host. In the present study we examine the effect of HIV-1 B and C clade-derived Tat on IDO and 5-HTT gene and protein expressions by dendritic cells as studied by quantitative polymerase chain reaction (qPCR) and Western blot. In addition, the intracellular IDO expression, IDO enzyme activity, and the levels of 5-HT and KYN were also measured. Results indicate that HIV-1 clade B Tat up-regulates IDO and down-regulates 5-HTT gene and protein expressions. Further, HIV-1 clade B Tat caused a reduction of 5-HT with simultaneous increase in KYN levels as compared to HIV-1 clade C Tat. These studies suggest that HIV-1 clade B and C Tat proteins may play a differential role in the neuropathogenesis of HIV-associated dementia (HAD) or HIV-associated neurocognitive disorder (HAND).


Subject(s)
AIDS Dementia Complex/metabolism , Dendritic Cells/metabolism , HIV-1/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Serotonin/biosynthesis , tat Gene Products, Human Immunodeficiency Virus/immunology , AIDS Dementia Complex/genetics , Blotting, Western , Cell Separation , Dendritic Cells/virology , Flow Cytometry , Gene Expression , Gene Expression Profiling , HIV Infections/immunology , HIV Infections/metabolism , HIV-1/genetics , Humans , Kynurenine/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Serotonin Plasma Membrane Transport Proteins/metabolism , tat Gene Products, Human Immunodeficiency Virus/genetics
9.
J Neurovirol ; 16(4): 294-305, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20624003

ABSTRACT

In recent years, increasing interest has emerged to assess the human immunodeficiency virus type 1 (HIV-1) clade C viral pathogenesis due to its anticipated spread in the United States and other western countries. Previous studies suggest that clade C is less neuropathogenic than clade B; however, the underlying mechanism is poorly understood. Additionally, the interactive role of drugs of abuse such as cocaine on clade C-associated neuropathogenesis has not been reported. In the current study, we hypothesize that HIV-1 clade-specific Tat proteins exert differential effects on blood-brain barrier (BBB) integrity and cocaine further differentially aggravates the BBB dysfunction. We evaluated the effect of Tat B and Tat C and/or cocaine on the BBB integrity using an in vitro model constructed with primary human brain microvascular endothelial cells (HBMECs) and astrocytes. The BBB membrane integrity was measured by transendothelial electrical resistance (TEER) and paracellular permeability was measured by fluorescein isothiocyanate (FITC)-dextran transport assay and monocytes transmigration across the BBB. Results indicate that Tat B disrupts BBB integrity to a greater extent compared to Tat C and cocaine further differentially exacerbates the BBB dysfunction. This BBB dysfunction was associated with altered expression of tight junction proteins zona occuldens (ZO-1) and junctional adhesion molecule (JAM)-2. Thus, these results for the first time delineate the differential role of Tat B and Tat C and/or cocaine in BBB dysfunction, which may be correlated with the clade-specific differences observed in HIV-1-associated neurological disorders.


Subject(s)
AIDS Dementia Complex/physiopathology , Blood-Brain Barrier/pathology , Cocaine/toxicity , Dopamine Uptake Inhibitors/toxicity , tat Gene Products, Human Immunodeficiency Virus/pharmacology , AIDS Dementia Complex/pathology , AIDS Dementia Complex/virology , Astrocytes/drug effects , Astrocytes/pathology , Astrocytes/virology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/virology , Blotting, Western , Capillary Permeability/drug effects , Cell Movement/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelial Cells/virology , Gene Expression , Gene Expression Profiling , HIV-1/genetics , Humans , Reverse Transcriptase Polymerase Chain Reaction , Tight Junctions/drug effects , Tight Junctions/pathology , Tight Junctions/virology , tat Gene Products, Human Immunodeficiency Virus/genetics
10.
Int J Nanomedicine ; 5: 157-66, 2010 Apr 07.
Article in English | MEDLINE | ID: mdl-20463931

ABSTRACT

Despite significant advances in highly active antiretroviral therapy (HAART), the prevalence of neuroAIDS remains high. This is mainly attributed to inability of antiretroviral therapy (ART) to cross the blood-brain barrier (BBB), thus resulting in insufficient drug concentration within the brain. Therefore, development of an active drug targeting system is an attractive strategy to increase the efficacy and delivery of ART to the brain. We report herein development of magnetic azidothymidine 5'-triphosphate (AZTTP) liposomal nanoformulation and its ability to transmigrate across an in vitro BBB model by application of an external magnetic field. We hypothesize that this magnetically guided nanoformulation can transverse the BBB by direct transport or via monocyte-mediated transport. Magnetic AZTTP liposomes were prepared using a mixture of phosphatidyl choline and cholesterol. The average size of prepared liposomes was about 150 nm with maximum drug and magnetite loading efficiency of 54.5% and 45.3%, respectively. Further, magnetic AZTTP liposomes were checked for transmigration across an in vitro BBB model using direct or monocyte-mediated transport by application of an external magnetic field. The results show that apparent permeability of magnetic AZTTP liposomes was 3-fold higher than free AZTTP. Also, the magnetic AZTTP liposomes were efficiently taken up by monocytes and these magnetic monocytes showed enhanced transendothelial migration compared to normal/non-magnetic monocytes in presence of an external magnetic field. Thus, we anticipate that the developed magnetic nanoformulation can be used for targeting active nucleotide analog reverse transcriptase inhibitors to the brain by application of an external magnetic force and thereby eliminate the brain HIV reservoir and help to treat neuroAIDS.


Subject(s)
Blood-Brain Barrier/chemistry , Dideoxynucleotides/administration & dosage , Dideoxynucleotides/chemistry , Drug Carriers/chemistry , Nanostructures/chemistry , Thymine Nucleotides/administration & dosage , Thymine Nucleotides/chemistry , Zidovudine/analogs & derivatives , Antiviral Agents/administration & dosage , Antiviral Agents/chemistry , Cells, Cultured , Diffusion , Drug Carriers/administration & dosage , Drug Compounding/methods , Electromagnetic Fields , Humans , Materials Testing , Nanomedicine/methods , Nanostructures/administration & dosage , Zidovudine/administration & dosage , Zidovudine/chemistry
11.
J Neurovirol ; 15(4): 343-7, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19575326

ABSTRACT

Inefficient cellular phosphorylation of nucleoside and nucleotide analog reverse transcriptase inhibitors (NRTIs) to their active nucleoside 5'-triphosphate (NTPs) form is one of the limitations for human immunodeficiency virus (HIV) therapy. We report herein direct binding of 3'-azido-3'-deoxythymidine-5'-triphosphate (AZTTP) onto magnetic nanoparticles (Fe(3)O(4); magnetite) due to ionic interaction. This magnetic nanoparticle bound AZTTP (MP-AZTTP) completely retained its biological activity as assessed by suppression of HIV-1 replication in peripheral blood mononuclear cells. The developed MP-AZTTP nanoformulation can be used for targeting active NRTIs to the brain by application of an external magnetic force and thereby eliminate the brain HIV reservoir and help to treat NeuroAIDS.


Subject(s)
Dideoxynucleotides/pharmacology , Drug Compounding/methods , Ferrosoferric Oxide/chemistry , Ferrosoferric Oxide/pharmacology , Thymine Nucleotides/pharmacology , Virus Replication/drug effects , Zidovudine/analogs & derivatives , Anti-HIV Agents/pharmacology , Blood-Brain Barrier , Cells, Cultured , Dideoxynucleotides/chemistry , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Electromagnetic Fields , Ferrosoferric Oxide/administration & dosage , Humans , Leukocytes, Mononuclear/virology , Nanomedicine/methods , Thymine Nucleotides/chemistry , Zidovudine/chemistry , Zidovudine/pharmacology
12.
AIDS Res Hum Retroviruses ; 25(7): 691-9, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19621989

ABSTRACT

The existence of multiple subtypes of HIV-1 worldwide has created new challenges to control HIV-1 infection and associated neuropathogenesis. Previous studies indicate a difference in neuropathogenic manifestations of HIV-1-associated neuroAIDS between clade B- and clade C-infected subjects with clade B being more neuropathogenic than clade C. However, the exact mechanism underlying the differences in the neuropathogenesis by both the subtypes remains elusive. Development of neuroAIDS is associated with a complex interplay between proinflammatory and antiinflammatory cytokines and chemokines. In the current study, we hypothesize that HIV-1 clade B and C Tat protein exert differential effects on human primary monocytes leading to differences in gene and protein expression of cytokines implicated in neuroAIDS. Primary human monocytes were treated with clade B and clade C Tat protein and quantitative real time PCR was performed to determine gene expression of proinflammatory cytokines (IL-6 and TNF-alpha) and antiinflammatory cytokines (IL-4 and IL-10). Further, cytokine secretion was measured in culture supernatants by ELISA, whereas intracellular cytokine expression was detected by flow cytometry. Results indicate that monocytes treated with Tat B showed significant upregulation of proinflammatory cytokines, IL-6 and TNF-alpha, as compared to Tat C-treated cultures. However, expression of antiinflammatory molecules and IL-4 and IL-10 was found to be higher in Tat C-treated compared to Tat B-treated cultures. Thus, our result shows for the first time that Tat B and Tat C differentially modulate expression of neuropathogenic molecules that may be correlated with the differences in neuroAIDS manifestation induced by clade-specific infections.


Subject(s)
Acquired Immunodeficiency Syndrome/metabolism , Cytokines/biosynthesis , HIV-1 , Monocytes/metabolism , tat Gene Products, Human Immunodeficiency Virus/physiology , Acquired Immunodeficiency Syndrome/virology , Cell Culture Techniques , Cells, Cultured , Cytokines/genetics , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation , HIV-1/genetics , HIV-1/pathogenicity , Host-Pathogen Interactions , Humans , Interleukin-10/biosynthesis , Interleukin-10/genetics , Interleukin-4/biosynthesis , Interleukin-4/genetics , Interleukin-6/biosynthesis , Interleukin-6/genetics , Monocytes/virology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Virulence
13.
J Neuroimmune Pharmacol ; 4(1): 129-39, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18958626

ABSTRACT

The US is currently experiencing an epidemic of methamphetamine (Meth) use as a recreational drug. Recent studies also show a high prevalence of HIV-1 infection among Meth users. We report that Meth enhances HIV-1 infectivity of dendritic cells as measured by multinuclear activation of a galactosidase indicator (MAGI) cell assay, p24 assay, and LTR-RU5 amplification. Meth induces increased HIV-1 infection in association with an increase in the HIV-1 coreceptors, CXCR4 and CCR5, and infection is mediated by downregulation of extracellular-regulated kinase (ERK2) and the upregulation of p38 mitogen-activated protein kinase (MAPK). A p38 inhibitor (SB203580) specifically reversed the Meth-induced upregulation of the CCR5 HIV-1 coreceptor. The dopamine D2 receptor antagonist RS +/- sulpiride significantly reversed the Meth-induced upregulation of CCR5, demonstrating that the Meth-induced effect is mediated via the D2 receptor. These studies report for the first time that Meth fosters HIV-1 infection, potentially via upregulating coreceptor gene expression. Further, Meth mediates its regulatory effects via dopamine receptors and via downregulating ERK2 with a reciprocal upregulation of p38 MAPK. Elucidation of the role of Meth in HIV-1 disease susceptibility and the mechanism through which Meth mediates its effects on HIV-1 infection may help to devise novel therapeutic strategies against HIV-1 infection in high-risk Meth-using HIV-1-infected subjects.


Subject(s)
Dendritic Cells/drug effects , Dendritic Cells/virology , Dopamine Uptake Inhibitors/pharmacology , HIV-1/pathogenicity , Methamphetamine/pharmacology , Monocytes/drug effects , Monocytes/virology , Adaptor Proteins, Signal Transducing , Blotting, Western , Cell Adhesion Molecules , Cell Adhesion Molecules, Neuronal/metabolism , Cells, Cultured , Cytosol/drug effects , Cytosol/metabolism , Dopamine/metabolism , Dose-Response Relationship, Drug , Flow Cytometry , Guanylate Kinases , HIV Core Protein p24/biosynthesis , HIV Core Protein p24/genetics , Humans , Kinetics , Oxidation-Reduction , RNA, Viral/biosynthesis , RNA, Viral/genetics , Receptors, Dopamine D2/drug effects , Receptors, Dopamine D2/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL