Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters











Publication year range
1.
J Neuropathol Exp Neurol ; 79(9): 986-997, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32783052

ABSTRACT

HIV-associated neurocognitive disorders (HAND) remain prevalent despite implementation of antiretroviral therapy (ART). Development of HAND is linked to mitochondrial dysfunction and oxidative stress in the brain; therefore, upregulation of antioxidant defenses is critical to curtail neuronal damage. Superoxide dismutase 2 (SOD2) is a mitochondrial antioxidant enzyme essential for maintaining cellular viability. We hypothesized that SOD2 was upregulated during retroviral infection. Using a simian immunodeficiency virus (SIV)-infected macaque model of HIV, quantitative PCR showed elevated SOD2 mRNA in cortical gray ([GM], 7.6-fold for SIV vs uninfected) and white matter ([WM], 77-fold for SIV vs uninfected) during SIV infection. Further, SOD2 immunostaining was enhanced in GM and WM from SIV-infected animals. Double immunofluorescence labeling illustrated that SOD2 primarily colocalized with astrocyte marker glial fibrillary acidic protein (GFAP) in SIV-infected animals. Interestingly, in ART-treated SIV-infected animals, brain SOD2 RNA levels were similar to uninfected animals. Additionally, using principal component analysis in a transcriptomic approach, SOD2 and GFAP expression separated SIV-infected from uninfected brain tissue. Projection of these data into a HIV dataset revealed similar expression changes, thereby validating the clinical relevance. Together, our findings suggest that novel SOD2-enhancing therapies may reduce neuroinflammation in ART-treated HIV-infected patients.


Subject(s)
AIDS Dementia Complex/enzymology , Astrocytes/enzymology , Simian Acquired Immunodeficiency Syndrome/enzymology , Superoxide Dismutase/metabolism , Animals , Anti-Retroviral Agents/pharmacology , Brain/enzymology , Macaca nemestrina , Male , Microglia/enzymology , Neurons/enzymology , Simian Acquired Immunodeficiency Syndrome/complications , Simian Immunodeficiency Virus , Superoxide Dismutase/drug effects , Up-Regulation
2.
J Neuroimmune Pharmacol ; 15(2): 209-223, 2020 06.
Article in English | MEDLINE | ID: mdl-31802418

ABSTRACT

HIV-1 Associated Neurocognitive Disorder (HAND) is a common and clinically detrimental complication of HIV infection. Viral proteins, including Tat, released from infected cells, cause neuronal toxicity. Substance abuse in HIV-infected patients greatly influences the severity of neuronal damage. To repurpose small molecule inhibitors for anti-HAND therapy, we employed MOLIERE, an AI-based literature mining system that we developed. All human genes were analyzed and prioritized by MOLIERE to find previously unknown targets connected to HAND. From the identified high priority genes, we narrowed the list to those with known small molecule ligands developed for other applications and lacking systemic toxicity in animal models. To validate the AI-based process, the selective small molecule inhibitor of DDX3 helicase activity, RK-33, was chosen and tested for neuroprotective activity. The compound, previously developed for cancer treatment, was tested for the prevention of combined neurotoxicity of HIV Tat and cocaine. Rodent cortical cultures were treated with 6 or 60 ng/ml of HIV Tat and 10 or 25 µM of cocaine, which caused substantial toxicity. RK-33 at doses as low as 1 µM greatly reduced the neurotoxicity of Tat and cocaine. Transcriptome analysis showed that most Tat-activated transcripts are microglia-specific genes and that RK-33 blocks their activation. Treatment with RK-33 inhibits the Tat and cocaine-dependent increase in the number and size of microglia and the proinflammatory cytokines IL-6, TNF-α, MCP-1/CCL2, MIP-2, IL-1α and IL-1ß. These findings reveal that inhibition of DDX3 may have the potential to treat not only HAND but other neurodegenerative diseases. Graphical Abstract RK-33, selective inhibitor of Dead Box RNA helicase 3 (DDX3) protects neurons from combined Tat and cocaine neurotoxicity by inhibition of microglia activation and production of proinflammatory cytokines.


Subject(s)
Azepines/pharmacology , Cocaine/toxicity , DEAD-box RNA Helicases/antagonists & inhibitors , Imidazoles/pharmacology , Microglia/drug effects , tat Gene Products, Human Immunodeficiency Virus/toxicity , AIDS Dementia Complex/drug therapy , AIDS Dementia Complex/enzymology , Animals , Azepines/therapeutic use , Cells, Cultured , DEAD-box RNA Helicases/metabolism , Dopamine Uptake Inhibitors/toxicity , Dose-Response Relationship, Drug , Female , Imidazoles/therapeutic use , Male , Microglia/enzymology , Rats , Rats, Sprague-Dawley
3.
Neurotox Res ; 36(3): 563-582, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31286433

ABSTRACT

Animal models have been used to study cellular processes related to human immunodeficiency virus-1 (HIV-1)-associated neurocognitive disorders (HAND). The HIV-1 transgenic (Tg) rat expresses HIV viral genes except the gag-pol replication genes and exhibits neuropathological features similar to HIV patients receiving combined antiretroviral therapy (cART). Using this rat, alterations in dopaminergic function have been demonstrated; however, the data for neuroinflammation and glial reactivity is conflicting. Differences in behavior, tyrosine hydroxylase (TH) immunoreactivity, neuroinflammation, and glia reactivity were assessed in HIV-1 Tg male rats. At 6 and 12 weeks of age, rotarod performance was diminished, motor activity was not altered, and active avoidance latency performance and memory were diminished in HIV-1 Tg rats. TH+ immunoreactivity in the substantia nigra (SN) was decreased at 8 months but not at 2-5 months. At 5 months, astrocyte and microglia morphology was not altered in the cortex, hippocampus, or SN. In the striatum, astrocytes were unaltered, microglia displayed slightly thickened proximal processes, mRNA levels for Iba1 and Cd11b were elevated, and interleukin (Il)1α,Cxcr3, and cell adhesion molecule, Icam, decreased. In the hippocampus, mRNA levels for Tnfa and Cd11b were slightly elevated. No changes were observed in the cortex or SN. The data support an age-related effect of HIV proteins upon the nigrostriatal dopaminergic system and suggest an early response of microglia in the terminal synaptic region with little evidence of an associated neuroinflammatory response across brain regions.


Subject(s)
AIDS Dementia Complex/pathology , Microglia/pathology , Substantia Nigra/enzymology , Tyrosine 3-Monooxygenase/metabolism , AIDS Dementia Complex/enzymology , AIDS Dementia Complex/metabolism , Aging/metabolism , Aging/physiology , Animals , Avoidance Learning , Disease Models, Animal , HIV-1 , Male , Motor Activity , Rats , Rats, Inbred F344 , Rats, Transgenic , Rotarod Performance Test
4.
J Neurochem ; 142(1): 153-159, 2017 07.
Article in English | MEDLINE | ID: mdl-28397245

ABSTRACT

The neuro-pathogenic mechanism(s) underlying HIV-associated neurocognitive disorders are mostly unknown. HIV-infected macrophages and microglial cells play a crucial role and the metabolic fate of l-arginine may be highly relevant to microglia activation. In this context, arginase (ARG), which uses l-arginine as substrate, can be on the same time a target and source of oxidative stress and inflammation. In this study, we investigated whether integrase strand transfer inhibitors share with the other antiretroviral drugs the ability to inhibit ARG activity. We used the previously validated cell model, namely the human microglia cell line, as well as the computational chemistry approach. Furthermore, here we characterized the activity of purified human ARG in a cell-free in vitro system, and investigated the effects of integrase strand transfer inhibitors in this newly validated model. Overall evidence shows that Dolutegravir, Raltegravir and Elvitegravir inhibit ARG activity.


Subject(s)
Arginase/antagonists & inhibitors , HIV Integrase Inhibitors/pharmacology , AIDS Dementia Complex/enzymology , AIDS Dementia Complex/metabolism , Arginase/metabolism , Cell-Free System , Cells, Cultured , HIV Integrase Inhibitors/metabolism , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Microglia/drug effects , Microglia/enzymology , Molecular Docking Simulation , Oxazines , Piperazines , Pyridones , Quinolones/pharmacology , Raltegravir Potassium/pharmacology
5.
J Biosci ; 42(3): 481-490, 2017 Sep.
Article in English | MEDLINE | ID: mdl-29358561

ABSTRACT

The pathogenesis of HIV-associated neurocognitive disorder (HAND) is modulated by host genetic susceptibility factors such as Matrix metalloproteinases (MMPs). Promoter polymorphism of MMP-1 and MMP-3 may modify the expression of the gene. Hence, we evaluated the association of MMP-1-16072G/1G and MMP-3-1612 5A/6A polymorphisms with development of HAND and the modulation of pathogenesis of HAND. We enrolled a total of 180 individuals, 50 HIVinfected individuals with HAND, 130 without HAND, and 150 healthy controls. Polymorphism of MMP-1 and MMP-3 were genotyped by PCR-RFLP. MMP-1-1607 2G1G, -16071G/2G-1G/1G genotypes and -1607 1G allele were associated with the development of HAND (OR = 1.64, P = 0.05; OR = 1.45, P = 0.04; OR = 1.69, P = 0.05). MMP-1- 16071G1G, MMP-3-16125A5A genotypes increased the risk for the development of HAND (OR = 1.78, P = 0.25; OR = 2.39, P = 0.13). MMP-3-1612 5A5A, -1612 6A/5A-5A/5A genotypes and -1612 5A allele were associated with the reduced risk of HAND (OR = 0.40, P = 0.05; OR = 0.53, P = 0.04; OR = 0.40, P = 0.01). Haplotype 5A1G increased the risk of development of HAND (OR = 1.93, P = 0.05). As observed in advanced HIV disease stage, MMP-1-1607 1G1G genotype enhance the risk for advancement of HIV disease (OR = 1.69, P = 0.89). MMP-3-1612 6A5A genotype showed higher risk for development of HAND in alcohol users (0R = 1.65, P = 0.44). MMP-1 genotype may have an influence on development of HAND whereas MMP3-1612 5A5A genotype may reduce risk for pathogenesis of HAND.


Subject(s)
AIDS Dementia Complex/genetics , Genetic Predisposition to Disease , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 3/genetics , Polymorphism, Single Nucleotide , AIDS Dementia Complex/enzymology , AIDS Dementia Complex/pathology , Adult , Alleles , Case-Control Studies , Disease Progression , Female , Gene Expression , Gene Frequency , Haplotypes , Humans , Male , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 3/metabolism , Middle Aged , Odds Ratio , Promoter Regions, Genetic , Risk
6.
J Infect Dis ; 210(6): 904-12, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24688074

ABSTRACT

We recently demonstrated direct evidence of increased monoamine oxidase (MAO) activity in the brain of a simian immunodeficiency virus (SIV) model of human immunodeficiency virus (HIV)-associated central nervous system (CNS) disease, consistent with previously reported dopamine deficits in both SIV and HIV infection. In this study, we explored potential mechanisms behind this elevated activity. MAO B messenger RNA was highest in macaques with the most severe SIV-associated CNS lesions and was positively correlated with levels of CD68 and GFAP transcripts in the striatum. MAO B messenger RNA also correlated with viral loads in the CNS of SIV-infected macaques and with oxidative stress. Furthermore, in humans, striatal MAO activity was elevated in individuals with HIV encephalitis, compared with activity in HIV-seronegative controls. These data suggest that the neuroinflammation and oxidative stress caused by SIV infection in the CNS may provide the impetus for increased transcription of MAO B and that MAO, and more broadly, oxidative stress, have significant potential as therapeutic targets in CNS disease due to HIV.


Subject(s)
AIDS Dementia Complex/enzymology , Brain/enzymology , Monoamine Oxidase/metabolism , Simian Acquired Immunodeficiency Syndrome/enzymology , Adult , Animals , Brain Chemistry , Corpus Striatum/enzymology , Female , Gene Expression Profiling , Glutathione/analysis , Humans , Macaca nemestrina/virology , Male , Middle Aged , Oxidative Stress , Real-Time Polymerase Chain Reaction , Viral Load
7.
Biochem Biophys Res Commun ; 443(1): 32-6, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24269238

ABSTRACT

Glutaminase plays a critical role in the generation of glutamate, a key excitatory neurotransmitter in the CNS. Excess glutamate release from activated macrophages and microglia correlates with upregulated glutaminase suggesting a pathogenic role for glutaminase. Both glutaminase siRNA and small molecule inhibitors have been shown to decrease excess glutamate and provide neuroprotection in multiple models of disease, including HIV-associated dementia (HAD), multiple sclerosis and ischemia. Consequently, inhibition of glutaminase could be of interest for treatment of these diseases. Bis-2-(5-phenylacetimido-1,2,4-thiadiazol-2-yl)ethyl sulfide (BPTES) and 6-diazo-5-oxo-l-norleucine (DON), two most commonly used glutaminase inhibitors, are either poorly soluble or non-specific. Recently, several new BPTES analogs with improved physicochemical properties were reported. To evaluate these new inhibitors, we established a cell-based microglial activation assay measuring glutamate release. Microglia-mediated glutamate levels were significantly augmented by tumor necrosis factor (TNF)-α, phorbol 12-myristate 13-acetate (PMA) and Toll-like receptor (TLR) ligands coincident with increased glutaminase activity. While several potent glutaminase inhibitors abrogated the increase in glutamate, a structurally related analog devoid of glutaminase activity was unable to block the increase. In the absence of glutamine, glutamate levels were significantly attenuated. These data suggest that the in vitro microglia assay may be a useful tool in developing glutaminase inhibitors of therapeutic interest.


Subject(s)
Glutamic Acid/metabolism , Glutaminase/antagonists & inhibitors , Microglia/drug effects , Neuroprotective Agents/pharmacology , Small Molecule Libraries/pharmacology , AIDS Dementia Complex/enzymology , Animals , Biological Assay , Brain Ischemia/enzymology , Cells, Cultured , Drug Evaluation, Preclinical , Mice , Microglia/enzymology , Microglia/metabolism , Multiple Sclerosis/enzymology , Neuroprotective Agents/chemistry , Neuroprotective Agents/isolation & purification , Small Molecule Libraries/chemistry , Small Molecule Libraries/isolation & purification , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/pharmacology , Toll-Like Receptors/agonists , Tumor Necrosis Factor-alpha/pharmacology
8.
Glia ; 59(6): 997-1007, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21438014

ABSTRACT

Microglia are critical cells in mediating the pathophysiology of neurodegenerative disorders such as HIV-associated neurocognitive disorders. We hypothesize that HIV-1 glycoprotein 120 (gp120) activates microglia by enhancing outward K(+) currents, resulting in microglia secretion of neurotoxins, consequent neuronal dysfunction, and death. To test this hypothesis, we studied the effects of gp120 on outward K(+) current in cultured rat microglia. Application of gp120 enhanced outward K(+) current in a dose-dependent manner, which was blocked by voltage-gated K(+) (K(v) ) channel blockers. Western blot analysis revealed that gp120 produced an elevated expression of K(v) channel proteins. Examination of activation and inactivation of outward K(+) currents showed that gp120 shifted membrane potentials for activation and steady-state inactivation. The gp120-associated enhancement of outward K(+) current was blocked by either a CXCR4 receptor antagonist T140 or a specific protein kinase A (PKA) inhibitor H89, suggesting the involvement of chemokine receptor CXCR4 and PKA in gp120-mediated enhancement of outward K(+) current. Biological significance of gp120-induced enhancement of microglia outward K(+) current was demonstrated by experimental results showing the neurotoxic activity of gp120-stimulated microglia, evaluated by TUNEL staining and MTT assay, significantly attenuated by K(v) channel blockers. Taken together, these results suggest that gp120 induces microglia neurotoxic activity by enhancing microglia outward K(+) current and that microglia K(v) channels may function as a potential target for the development of therapeutic strategies.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , HIV Envelope Protein gp120/physiology , Microglia/metabolism , Microglia/virology , Potassium Channels/metabolism , Receptors, CXCR4/physiology , Signal Transduction/physiology , AIDS Dementia Complex/enzymology , AIDS Dementia Complex/metabolism , AIDS Dementia Complex/virology , Animals , Cells, Cultured , Coculture Techniques , Membrane Potentials/physiology , Microglia/enzymology , Nerve Degeneration/enzymology , Nerve Degeneration/metabolism , Nerve Degeneration/virology , Rats , Rats, Sprague-Dawley
9.
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
10.
J Neuroimmune Pharmacol ; 6(2): 202-15, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21286833

ABSTRACT

Infection with human immunodeficiency virus-1 (HIV-1) often leads to HIV-associated neurocognitive disorders (HAND) prior to the progression to acquired immunodeficiency syndrome (AIDS). At the cellular level, mitogen-activated protein kinases (MAPK) provide a family of signal transducers that regulate many processes in response to extracellular stimuli and environmental stress, such as viral infection. Recently, evidence has accumulated suggesting that p38 MAPK plays crucial roles in various pathological processes associated with HIV infection, ranging from macrophage activation to neurotoxicity and impairment of neurogenesis to lymphocyte apoptosis. Thus, p38 MAPK, which has generally been linked to stress-related signal transduction, may be an important mediator in the development of AIDS and HAND.


Subject(s)
AIDS Dementia Complex/enzymology , Brain Injuries/enzymology , HIV-1 , p38 Mitogen-Activated Protein Kinases/physiology , AIDS Dementia Complex/immunology , AIDS Dementia Complex/psychology , Acquired Immunodeficiency Syndrome/enzymology , Acquired Immunodeficiency Syndrome/virology , Animals , Brain Injuries/immunology , Brain Injuries/psychology , Cognition Disorders/enzymology , HIV Infections/enzymology , HIV Infections/immunology , HIV Infections/psychology , Humans , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Signal Transduction/immunology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
11.
J Neurosci ; 30(42): 14153-64, 2010 Oct 20.
Article in English | MEDLINE | ID: mdl-20962236

ABSTRACT

Human immunodeficiency virus-associated neurological disease (HAND) still causes significant morbidity, despite success reducing viral loads with combination antiretroviral therapy. The dopamine (DA) system is particularly vulnerable in HAND. We hypothesize that early, "reversible" DAergic synaptic dysfunction occurs long before DAergic neuron loss. As such, aging human immunodeficiency virus (HIV)-infected individuals may be vulnerable to other age-related neurodegenerative diseases like Parkinson's disease (PD), underscoring the need to understand shared molecular targets in HAND and PD. Previously, we reported that the neurotoxic HIV-1 transactivating factor (Tat) acutely disrupts mitochondrial and endoplasmic reticulum calcium homeostasis via ryanodine receptor (RyR) activation. Here, we further report that Tat disrupts DA transporter (DAT) activity and function, resulting in increased plasma membrane (PM) DAT and increased DAT V(max), without changes in K(m) or total DAT protein. Tat also increases calpain protease activity at the PM, demonstrated by total internal reflection fluorescence microscopy of a cleavable fluorescent calpain substrate. Tat-increased PM DAT and calpain activity are blocked by the RyR antagonists ryanodine and dantrolene, the calpain inhibitor calpastatin, and by a specific inhibitor of GSK-3ß. We conclude that Tat activates RyRs via a calcium- and calpain-mediated mechanism that upregulates DAT trafficking to the PM, and is independent of DAT protein synthesis, reinforcing the feasibility of RyR and GSK-3ß inhibition as clinical therapeutic approaches for HAND. Finally, we provide key translational relevance for these findings by highlighting published human data of increased DAT levels in striata of HAND patients and by demonstrating similar findings in Tat-expressing transgenic mice.


Subject(s)
Calpain/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Peptide Hydrolases/metabolism , Ryanodine Receptor Calcium Release Channel/drug effects , tat Gene Products, Human Immunodeficiency Virus/pharmacology , AIDS Dementia Complex/enzymology , AIDS Dementia Complex/pathology , Animals , Blotting, Western , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Enzyme Activation/drug effects , Glycogen Synthase Kinase 3/physiology , Glycogen Synthase Kinase 3 beta , Kinetics , Mesencephalon/cytology , Mesencephalon/drug effects , Mesencephalon/metabolism , Mice , Mice, Transgenic , Microscopy, Fluorescence , Neurons/metabolism , PC12 Cells , Protease Inhibitors/pharmacology , Rats , Subcellular Fractions/physiology
12.
J Neurochem ; 109(2): 551-61, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19222703

ABSTRACT

Mononuclear phagocyte (MP, macrophages and microglia) dysfunction plays a significant role in the pathogenesis of HIV-1-associated dementia (HAD) through the production and release of soluble neurotoxic factors including glutamate. Glutamate production is greatly increased following HIV-1 infection of cultured MP, a process dependent upon the glutamate-generating enzyme glutaminase. Glutaminase inhibition was previously found to significantly decrease macrophage-mediated neurotoxicity. Potential mechanisms of glutaminase-mediated excitotoxicity including enzyme up-regulation, increased enzyme activity and glutaminase localization were investigated in this report. RNA and protein analysis of HIV-infected human primary macrophage revealed up-regulation of the glutaminase isoform GAC, yet identified no changes in the kidney-type glutaminase isoform over the course of infection. Glutaminase is a mitochondrial protein, but was found to be released into the cytosol and extracellular space following infection. This released enzyme is capable of rapidly converting the abundant extracellular amino acid glutamine into excitotoxic levels of glutamate in an energetically favorable process. These findings support glutaminase as a potential component of the HAD pathogenic process and identify a possible therapeutic avenue for the treatment of neuroinflammatory states such as HAD.


Subject(s)
Glutamic Acid/biosynthesis , Glutaminase/physiology , HIV Infections/metabolism , HIV-1 , Macrophages/metabolism , Macrophages/virology , AIDS Dementia Complex/enzymology , AIDS Dementia Complex/metabolism , AIDS Dementia Complex/pathology , AIDS Dementia Complex/virology , Cells, Cultured , Glutamic Acid/adverse effects , Glutaminase/metabolism , HIV Infections/enzymology , HIV Infections/pathology , HIV Infections/virology , Humans , Macrophages/enzymology , Macrophages/pathology
13.
J Neuroimmunol ; 206(1-2): 106-11, 2009 Jan 03.
Article in English | MEDLINE | ID: mdl-19101040

ABSTRACT

HIV-associated cognitive neurological disorders (HAND) prevail in the antiretroviral therapy era. Proteomics analysis of CSF revealed expression of Cu/Zn superoxide dismutase (Cu/Zn SOD) in Hispanic women with cognitive impairment (CI). We tested the hypothesis that there is reduced capacity of antioxidant enzymes in CI by measures of expression and activity of Cu/Zn SOD, catalase, and Se-glutathione peroxidase in HAND. Our results showed that the function of these antioxidants was decreased in the CSF and monocytes of women with CI. These findings have important implications regarding their possible contribution to oxidative stress and in the diagnosis and therapy for HAND.


Subject(s)
AIDS Dementia Complex/enzymology , Catalase/metabolism , Glutathione Peroxidase/metabolism , Monocytes/enzymology , Superoxide Dismutase/metabolism , AIDS Dementia Complex/blood , AIDS Dementia Complex/cerebrospinal fluid , AIDS Dementia Complex/virology , Adult , Analysis of Variance , Catalase/blood , Catalase/cerebrospinal fluid , Confidence Intervals , Female , Gene Expression Regulation, Enzymologic , Glutathione Peroxidase/blood , Glutathione Peroxidase/cerebrospinal fluid , Hispanic or Latino , Humans , Longitudinal Studies , Retrospective Studies , Superoxide Dismutase/blood , Superoxide Dismutase/cerebrospinal fluid
14.
J Immunol ; 181(9): 6406-16, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18941231

ABSTRACT

HIV-1 infection has significant effect on the immune system as well as on the nervous system. Breakdown of the blood-brain barrier (BBB) is frequently observed in patients with HIV-associated dementia (HAD) despite lack of productive infection of human brain microvascular endothelial cells (HBMEC). Cellular products and viral proteins secreted by HIV-1 infected cells, such as the HIV-1 Gp120 envelope glycoprotein, play important roles in BBB impairment and HIV-associated dementia development. HBMEC are a major component of the BBB. Using cocultures of HBMEC and human astrocytes as a model system for human BBB as well as in vivo model, we show for the first time that cannabinoid agonists inhibited HIV-1 Gp120-induced calcium influx mediated by substance P and significantly decreased the permeability of HBMEC as well as prevented tight junction protein down-regulation of ZO-1, claudin-5, and JAM-1 in HBMEC. Furthermore, cannabinoid agonists inhibited the transmigration of human monocytes across the BBB and blocked the BBB permeability in vivo. These results demonstrate that cannabinoid agonists are able to restore the integrity of HBMEC and the BBB following insults by HIV-1 Gp120. These studies may lead to better strategies for treatment modalities targeted to the BBB following HIV-1 infection of the brain based on cannabinoid pharmacotherapies.


Subject(s)
Anti-HIV Agents/pharmacology , Arachidonic Acids/pharmacology , Brain/blood supply , Brain/virology , Cannabinoid Receptor Modulators/pharmacology , Endothelium, Vascular/virology , Glycerides/pharmacology , HIV Envelope Protein gp120/antagonists & inhibitors , HIV-1/drug effects , AIDS Dementia Complex/enzymology , AIDS Dementia Complex/pathology , AIDS Dementia Complex/prevention & control , AIDS Dementia Complex/virology , Amidohydrolases/antagonists & inhibitors , Anti-HIV Agents/therapeutic use , Arachidonic Acids/physiology , Benzamides/pharmacology , Benzamides/therapeutic use , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiology , Brain/drug effects , Brain/pathology , Cannabinoid Receptor Modulators/agonists , Cannabinoid Receptor Modulators/physiology , Carbamates/pharmacology , Carbamates/therapeutic use , Cell Line , Coculture Techniques , Endocannabinoids , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiology , Glycerides/physiology , HIV Envelope Protein gp120/physiology , HIV-1/physiology , Humans , Microcirculation/drug effects , Microcirculation/physiology , Receptor, Cannabinoid, CB1/physiology
15.
J Neurochem ; 105(3): 994-1005, 2008 May.
Article in English | MEDLINE | ID: mdl-18088378

ABSTRACT

A significant number of patients infected with human immunodeficiency virus-1 (HIV-1) suffer cognitive impairment ranging from mild to severe HIV-associated dementia (HAD), a result of neuronal degeneration in the basal ganglia, cerebral cortex and hippocampus. Mononuclear phagocyte dysfunction is thought to play an important role in the pathogenesis of HAD. Glutamate neurotoxicity is triggered primarily by massive Ca2+ influx arising from over-stimulation of the NMDA subtype of glutamate receptors. The underlying mechanisms, however, remain elusive. We have tested the hypothesis that mitochondrial glutaminase in HIV-infected macrophages is involved in converting glutamine to glutamate. Our results demonstrate that the concentration of glutamate in HIV-1 infected conditioned media was dependent on glutamine dose, and HIV-1 infected conditioned medium mediated glutamine-dependent neurotoxicity. These results indicate HIV-infection mediates neurotoxicity through glutamate production. In addition, glutamate-mediated neurotoxicity correlated with caspase activation and neuronal cell cycle re-activation. Inhibition of mitochondrial glutaminase diminished the HIV-induced glutamate production, and attenuated NMDA over-stimulation and subsequent neuronal apoptosis. These data implicate mitochondrial glutaminase in the induction of glutamate-mediated neuronal apoptosis during HIV-associated dementia, and provides a possible therapeutic strategy for HAD treatment.


Subject(s)
AIDS Dementia Complex/enzymology , Apoptosis/physiology , Glutamic Acid/biosynthesis , Glutaminase/metabolism , Macrophages/enzymology , Nerve Degeneration/enzymology , AIDS Dementia Complex/physiopathology , Animals , Brain/enzymology , Brain/physiopathology , Brain/virology , Caspases/metabolism , Cell Cycle/drug effects , Cell Cycle/physiology , Cells, Cultured , Culture Media, Conditioned/pharmacology , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Glutamine/metabolism , Glutamine/pharmacology , Humans , Macrophages/drug effects , Macrophages/virology , Mitochondria/enzymology , Monocytes/physiology , Nerve Degeneration/physiopathology , Nerve Degeneration/virology , Neurons/metabolism , Neurons/pathology , Neurotoxins/metabolism , Rats
16.
J Neuroimmune Pharmacol ; 2(1): 93-6, 2007 Mar.
Article in English | MEDLINE | ID: mdl-18040831

ABSTRACT

Highly active antiretroviral therapy (HAART) has made a significant impact on the lives of people living with HIV-1 infection. The incidence of neurologic disease associated with HIV-1 infection of the CNS plummeted between 1996-2000, but unfortunately the number of people currently HIV-1 infected (i.e., prevalence) with associated cognitive impairment has been steadily rising. While the reasons for this may be multifactorial, the implication is clear: there is a pressing need for adjunctive therapy directed at reversing or preventing damage to vulnerable pathways in the central nervous system (CNS) from HIV-1 infection. Using a team of preclinical and clinical investigators, we have focused our efforts on defining how proinflammatory mediators and secretory neurotoxins from HIV-1 disrupt signaling of the survival-regulating enzyme, glycogen synthase kinase 3 beta (GSK-3beta). In a series of studies initiated using in vitro, then in vivo models of HIV-1-associated dementia (HAD), we have demonstrated the ability of the mood stabilizing and anticonvulsant drug, sodium valproate (VPA), that inhibits GSK-3beta activity and other downstream mediators, to reverse HIV-1-induced damage to synaptic pathways in the CNS. Based on these results, we successfully performed pharmacokinetic and safety and tolerability trials with VPA in a cohort of HIV-1-infected patients with neurologic disease. VPA was well tolerated in this population and secondary measures of brain metabolism, as evidenced by an increase in N-acetyl aspartate/creatine (NAA/Cr), further suggested that VPA may improve gray matter integrity in brain regions damaged by HIV-1. These findings highlight the therapeutic potential of GSK-3beta blockade.


Subject(s)
AIDS Dementia Complex/enzymology , Acquired Immunodeficiency Syndrome/enzymology , Drug Delivery Systems/methods , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , AIDS Dementia Complex/drug therapy , Acquired Immunodeficiency Syndrome/drug therapy , Animals , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , Glycogen Synthase Kinase 3 beta , Humans , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Valproic Acid/pharmacology , Valproic Acid/therapeutic use
17.
J Neurosci ; 27(41): 11047-55, 2007 Oct 10.
Article in English | MEDLINE | ID: mdl-17928446

ABSTRACT

Previous work has demonstrated that the surface glycoprotein (gp120) of human immunodeficiency virus-1 (HIV-1) can induce damage and apoptosis of neurons both in vitro and in vivo. In this report, we provide evidence that double-stranded RNA-activated protein kinase (PKR), a stress kinase, is involved in HIV/gp120-associated neurodegeneration. In cultures of mixed cortical cells, HIV/gp120 increased the protein level of PKR. Additionally, PKR was phosphorylated in neurons but not glia after exposure to gp120. The use of two independent pharmacological inhibitors of PKR activity abrogated neuronal cell death induced by gp120. Cortical neurons from PKR knock-out mice were significantly protected from neurotoxicity induced by gp120, further validating the pivotal proapoptotic function of PKR. gp120-induced phosphorylated PKR localized prominently to neuronal nuclei; PKR inhibition or the NMDA receptor antagonist MK-801 [(+)-5-methyl-10,11-dihydro-5H-dibenzo [a,d] cyclohepten-5,10-imine maleate] abrogated this effect. PKR inactivation also inhibited gp120-induced caspase-3 activation, consistent with its neuroprotective effect. Finally, brain tissue from individuals diagnosed with HIV-associated dementia (HAD), but not HIV infection alone, contained the activated form of PKR, which localized predominantly to neuronal nuclei. Together, these results identify PKR as a critical mediator of gp120 neurotoxicity, suggesting that activation of PKR contributes to the neuronal injury and cell death observed in HAD.


Subject(s)
Apoptosis/physiology , HIV Envelope Protein gp120/physiology , HIV-1/physiology , Neurons/enzymology , Neurons/virology , Signal Transduction/physiology , eIF-2 Kinase/physiology , AIDS Dementia Complex/enzymology , AIDS Dementia Complex/pathology , AIDS Dementia Complex/virology , Adult , Animals , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neurons/pathology , Rats , Rats, Sprague-Dawley , eIF-2 Kinase/deficiency , eIF-2 Kinase/genetics
18.
Brain Res ; 1123(1): 216-225, 2006 Dec 06.
Article in English | MEDLINE | ID: mdl-17078933

ABSTRACT

Human immunodeficiency virus (HIV)-1 infection of the central nervous system occurs in the vast majority of HIV-infected patients. HIV-associated dementia (HAD) represents the most severe form of HIV-related neuropsychiatric impairment and is associated with neuropathology involving HIV proteins and activation of proinflammatory cytokine circuits. Interferon-gamma (IFN-gamma) activates the JAK/STAT1 pathway, a key regulator of inflammatory and apoptotic signaling, and is elevated in HIV-1-infected brains progressing to HAD. Recent reports suggest green tea-derived (-)-epigallocatechin-3-gallate (EGCG) can attenuate neuronal damage mediated by this pathway in conditions such as brain ischemia. In order to investigate the therapeutic potential of EGCG to mitigate the neuronal damage characteristic of HAD, IFN-gamma was evaluated for its ability to enhance well-known neurotoxic properties of HIV-1 proteins gp120 and Tat in primary neurons and mice. Indeed, IFN-gamma enhanced the neurotoxicity of gp120 and Tat via increased JAK/STAT signaling. Additionally, primary neurons pretreated with a JAK1 inhibitor, or those derived from STAT1-deficient mice, were largely resistant to the IFN-gamma-enhanced neurotoxicity of gp120 and Tat. Moreover, EGCG treatment of primary neurons from normal mice reduced IFN-gamma-enhanced neurotoxicity of gp120 and Tat by inhibiting JAK/STAT1 pathway activation. EGCG was also found to mitigate the neurotoxic properties of HIV-1 proteins in the presence of IFN-gamma in vivo. Taken together, these data suggest EGCG attenuates the neurotoxicity of IFN-gamma augmented neuronal damage from HIV-1 proteins gp120 and Tat both in vitro and in vivo. Thus EGCG may represent a novel natural copound for the prevention and treatment of HAD.


Subject(s)
AIDS Dementia Complex/drug therapy , Catechin/analogs & derivatives , HIV-1/immunology , Interferon-gamma/metabolism , Janus Kinases/metabolism , Neurons/enzymology , Neuroprotective Agents/therapeutic use , STAT1 Transcription Factor/metabolism , AIDS Dementia Complex/enzymology , AIDS Dementia Complex/immunology , AIDS Dementia Complex/pathology , Analysis of Variance , Animals , Apoptosis/drug effects , Apoptosis/immunology , Catechin/therapeutic use , Cells, Cultured , Female , Gene Products, tat/immunology , Gene Products, tat/toxicity , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp120/toxicity , Interferon-gamma/adverse effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/immunology , Neurons/pathology , Neurotoxins/immunology , Neurotoxins/toxicity , STAT1 Transcription Factor/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , tat Gene Products, Human Immunodeficiency Virus
19.
J Acquir Immune Defic Syndr ; 39(4): 422-5, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16010164

ABSTRACT

Expansion of the lysosomal apparatus occurs in subcortical white matter in brains from persons with AIDS. This study examined whether HIV-associated subcortical dementia (HAD) is significantly related to this lysosomal anomaly. Brain cortex and adjacent white matter from the middle frontal gyrus were obtained from the National NeuroAIDS Tissue Consortium. Lysosomal hydrolase activity was assayed in 57 subjects who underwent neuropsychological testing within 6 months prior to autopsy. Decedents were evaluated from 4 geographical sites in the United States: Galveston/Houston, Texas (n = 36), Los Angeles, California (n = 5), New York, New York (n = 5), and San Diego, California (n = 11). Increased beta-glucuronidase activity, a representative lysosomal glycosidase, was correlated with the amount of neurocognitive impairment. Significant correlation was present in 5 of 7 functional testing domains, including some that draw upon frontal lobe output (r = 0.419; P < 0.002). The biochemical anomaly was negligible in cerebral cortex and cerebrospinal fluid and was not correlated with brain dysfunction in those compartments. Glycosidase activation was associated significantly with increased HIV RNA concentration in brain tissue (r = 0.469; P < 0.021) and possibly with HIV RNA in cerebrospinal fluid (r = 0.266; P < 0.067). HIV RNA in blood plasma was not correlated. These results support the suggestion that abnormal metabolism in white matter glial cells contributes to cognitive slowing in persons with HAD. Because membrane turnover is routed through the endosome-lysosome apparatus, these data are in agreement with brain spectroscopic data that have suggested that there is an increase in membrane turnover in white matter glia.


Subject(s)
AIDS Dementia Complex/physiopathology , Frontal Lobe/physiopathology , Lysosomes/enzymology , AIDS Dementia Complex/enzymology , Antiretroviral Therapy, Highly Active , Cognition Disorders/physiopathology , Frontal Lobe/enzymology , Frontal Lobe/ultrastructure , HIV-1 , Humans , RNA, Viral/cerebrospinal fluid
20.
J Neuroimmunol ; 163(1-2): 190-4, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15885322

ABSTRACT

To evaluate the potential role of the uPAR/uPA/PAI-1 system in HIV-induced blood-brain-barrier (BBB) disruption, CSF uPA-dependent plasminogen activation (PdPA) was analyzed by casein zymography, and CSF protein levels of all three molecules were measured by ELISA. CSF uPAR, but not uPA, PAI-1, or PdPA levels was significantly increased in neurologically compromised HIV+ patients. Only individual patients with severe AIDS dementia complex had increased levels of uPA (but not PAI-1) which fell upon initiation of antiretroviral therapy. The levels of all three molecules did not correlate with the CSF to serum albumin ratio suggesting not an important role in HIV-induced BBB disruption.


Subject(s)
HIV Infections/cerebrospinal fluid , HIV-1 , Plasminogen Activator Inhibitor 1/cerebrospinal fluid , Receptors, Cell Surface/metabolism , Urokinase-Type Plasminogen Activator/cerebrospinal fluid , AIDS Dementia Complex/cerebrospinal fluid , AIDS Dementia Complex/enzymology , AIDS Dementia Complex/immunology , CD4 Lymphocyte Count , HIV Infections/enzymology , HIV Infections/immunology , HIV Infections/virology , Hemagglutination Inhibition Tests , Humans , Receptors, Cell Surface/blood , Receptors, Urokinase Plasminogen Activator , Serum Albumin/cerebrospinal fluid , Solubility , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL