Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Neurosci Lett ; 754: 135863, 2021 05 29.
Article in English | MEDLINE | ID: mdl-33794296

ABSTRACT

Various research studies that have investigated the association between HIV infection and addiction underpin the role of various drugs of abuse in impairing immunological and non-immunological pathways of the host system, ultimately leading to augmentation of HIV infection and disease progression. These studies have included both in vitro and in vivo animal models wherein investigators have assessed the effects of various drugs on several disease parameters to decipher the impact of drugs on both HIV infection and progression of HIV-associated neurocognitive disorders (HAND). However, given the inherent limitations in the existing animal models of HAND, these investigations only recapitulated specific aspects of the disease but not the complex human syndrome. Despite the inability of HIV to infect rodents over the last 30 years, multiple strategies have been employed to develop several rodent models of HAND. While none of these models can accurately mimic the overall pathophysiology of HAND, they serve the purpose of modeling some unique aspects of HAND. This review provides an overview of various animal models used in the field and a careful evaluation of methodological strengths and limitations inherent in both the model systems and study designs to understand better how the various animal models complement one another.


Subject(s)
Brain/physiopathology , Disease Models, Animal , HIV Infections/complications , Neurocognitive Disorders/epidemiology , Substance-Related Disorders/epidemiology , Animals , Brain/virology , Comorbidity , HIV Infections/psychology , HIV-1/pathogenicity , Humans , Mice , Neurocognitive Disorders/etiology , Neurocognitive Disorders/physiopathology , Neurocognitive Disorders/psychology , Rats , Substance-Related Disorders/physiopathology , Substance-Related Disorders/psychology
3.
Drug Metab Dispos ; 42(12): 2097-108, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25301936

ABSTRACT

Methamphetamine (MA), which remains one of the widely used drugs of abuse, is metabolized by the cytochrome P450 (P450) family of enzymes in humans. However, metabolism of methamphetamine in macaques is poorly understood. Therefore, we first developed and validated a very sensitive liquid chromatography with tandem mass spectrometry (LC-MS/MS) method using solid phase extraction of rhesus plasma with a lower limit of quantitation at 1.09 ng/ml for MA and its metabolites, 4-hydroxy methamphetamine (4-OH MA), amphetamine (AM), 4-OH amphetamine (4-OH AM), and norephedrine. We then analyzed plasma samples of MA-treated rhesus, which showed >10-fold higher concentrations of AM (∼29 ng/ml) and 4-OH AM (∼28 ng/ml) than MA (∼2 ng/ml). Because the plasma levels of MA metabolites in rhesus were much higher than in human samples, we examined MA metabolism in human and rhesus microsomes. Interestingly, the results showed that AM and 4-OH AM were formed more rapidly and that the catalytic efficiency (Vmax/Km) for the formation of AM was ∼8-fold higher in rhesus than in human microsomes. We further examined the differences in these kinetic characteristics using three selective inhibitors of each human CYP2D6 and CYP3A4 enzymes. The results showed that each of these inhibitors inhibited both d- and l-MA metabolism by 20%-60% in human microsomes but not in rhesus microsomes. The differences between human and rhesus CYP2D6 and CYP3A4 enzymes were further assessed by docking studies for both d and l-MA. In conclusion, our results demonstrated an enhanced MA metabolism in rhesus compared with humans, which is likely to be caused by differences in MA-metabolizing P450 enzymes between these species.


Subject(s)
Amphetamine/blood , Amphetamine/metabolism , Methamphetamine/blood , Methamphetamine/metabolism , Phenylpropanolamine/blood , Phenylpropanolamine/metabolism , Animals , Chromatography, Liquid/methods , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 CYP3A/metabolism , Humans , Kinetics , Macaca mulatta/metabolism , Microsomes, Liver/metabolism , Tandem Mass Spectrometry/methods
4.
Curr HIV Res ; 12(4): 282-92, 2014.
Article in English | MEDLINE | ID: mdl-25053363

ABSTRACT

Approximately 25% of the HIV-1 positive population is also infected with HCV. The effects of alcohol on HIV-1 or HCV infection have been a research topic of interest due to the high prevalence of alcohol use in these infected patient populations. Although it has long been known that HIV-1 infects the brain, it has only been a little more than a decade since HCV infection of the CNS has been characterized. Both viruses are capable of infecting and replicating in microglia and increasing the expression of proinflammatory cytokines and chemokines, including IL-6 and IL-8. Investigations focusing on the effects of HIV-1, HCV or alcohol on neuroinflammation have demonstrated that these agents are capable of acting through overlapping signaling pathways, including MAPK signaling molecules. In addition, HIV-1, HCV and alcohol have been demonstrated to increase permeability of the blood-brain barrier. Patients infected with either HIV-1 or HCV, or those who use alcohol, exhibit metabolic abnormalities in the CNS that result in altered levels of n-acetyl aspartate, choline and creatine in various regions of the brain. Treatment of HIV/HCV co-infection in alcohol users is complicated by drug-drug interactions, as well as the effects of alcohol on drug metabolism. The drug-drug interactions between the antiretrovirals and the antivirals, as well as the effects of alcohol on drug metabolism, complicate existing models of CNS penetration, making it difficult to assess the efficacy of treatment on CNS infection.


Subject(s)
Alcoholism/complications , Central Nervous System Infections/immunology , Central Nervous System Infections/pathology , HIV Infections/complications , HIV-1/immunology , Hepacivirus/immunology , Hepatitis C, Chronic/complications , Antiviral Agents/therapeutic use , Central Nervous System Infections/virology , Drug Interactions , HIV Infections/drug therapy , HIV Infections/immunology , HIV Infections/pathology , HIV-1/growth & development , Hepacivirus/growth & development , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/pathology , Humans , Signal Transduction
5.
Sci Rep ; 4: 4450, 2014 Mar 24.
Article in English | MEDLINE | ID: mdl-24658403

ABSTRACT

The prevalence of HIV-associated neurocognitive disorders (HAND) remains high in patients infected with HIV-1. The production of pro-inflammatory cytokines by astrocytes/microglia exposed to viral proteins is thought to be one of the mechanisms leading to HIV-1- mediated neurotoxicity. In the present study we examined the effects of Nef on CCL5 induction in astrocytes. The results demonstrate that CCL5 is significantly induced in Nef-transfected SVGA astrocytes. To determine the mechanisms responsible for the increased CCL5 caused by Nef, we employed siRNA and chemical antagonists. Antagonists of NF-κB, PI3K, and p38 significantly reduced the expression levels of CCL5 induced by Nef transfection. Furthermore, specific siRNAs demonstrated that the Akt, p38MAPK, NF-κB, CEBP, and AP-1 pathways play a role in Nef-mediated CCL5 expression. The results demonstrated that the PI3K/Akt and p38 MAPK pathways, along with the transcription factors NF-κB, CEBP, and AP-1, are involved in Nef-induced CCL5 production in astrocytes.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Chemokine CCL5/genetics , HIV Infections/genetics , HIV-1/genetics , Transcription Factor AP-1/genetics , nef Gene Products, Human Immunodeficiency Virus/genetics , Astrocytes/metabolism , Astrocytes/pathology , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line , Chemokine CCL5/metabolism , Cognition Disorders/complications , Cognition Disorders/genetics , Cognition Disorders/pathology , Cognition Disorders/virology , Elafin/antagonists & inhibitors , Gene Expression Regulation, Viral , HIV Infections/complications , HIV Infections/virology , HIV-1/pathogenicity , NF-kappa B/antagonists & inhibitors , Neurotoxicity Syndromes/complications , Neurotoxicity Syndromes/genetics , Neurotoxicity Syndromes/pathology , Oncogene Protein v-akt/metabolism , Signal Transduction , Transcription Factor AP-1/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
6.
Alcohol Clin Exp Res ; 38(3): 604-10, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24134164

ABSTRACT

The use of alcohol has been associated with both an increased risk of acquisition of HIV-1 infection and an increased rate of disease progression among those already infected by the virus. The potential for alcohol to exacerbate the effects of HIV infection is especially important in the central nervous system (CNS) because this area is vulnerable to the combined effects of alcohol and HIV infection. The effects of alcohol on glial cells are mediated through receptors such as Toll-like receptor 4 and N-methyl-d-aspartate receptor. This causes the activation of signaling molecules such as interleukin-1 receptor-associated kinase and various members of the P38 mitogen-activated protein kinase family and subsequent activation of transcription factors such as nuclear factor-kappa beta and activator protein 1. The eventual outcome is an increase in pro-inflammatory cytokine production by glial cells. Alcohol also induces higher levels of NADPH oxidase in glial cells, which leads to an increased production of reactive oxygen species (ROS). Viral invasion of the CNS occurs early after infection, and HIV proteins have also been demonstrated to increase levels of pro-inflammatory cytokines and ROS in glial cells through activation of some of the same pathways activated by alcohol. Both cell culture systems and animal models have demonstrated that concomitant exposure to alcohol and HIV/HIV proteins results in increased levels of expression of pro-inflammatory cytokines such as interleukin-1 beta and tumor necrosis factor-alpha, along with increased levels of oxidative stress. Clinical studies also suggest that alcohol exacerbates the CNS effects of HIV-1 infection. This review focuses on the mechanisms by which alcohol causes increased CNS damage in HIV-1 infection.


Subject(s)
Alcohol Drinking/physiopathology , Central Nervous System/physiopathology , HIV Infections/physiopathology , HIV-1/physiology , Host-Pathogen Interactions , Animals , Clinical Trials as Topic , Ethanol , Human Immunodeficiency Virus Proteins/physiology , Humans
7.
PLoS One ; 7(12): e52060, 2012.
Article in English | MEDLINE | ID: mdl-23251686

ABSTRACT

HIV-1 envelope protein gp120 has been extensively studied for neurotoxic effects that have been attributed to the increased expression of various proinflammatory cytokines in the CNS. Recently we have shown that methamphetamine (MA) also increases expression of proinflammatory cytokines in astrocytes. However, combined effect of gp120 and MA is not known. The present study was undertaken to determine cumulative effect and the mechanism(s)/pathways involved in the functional interaction between gp120 and MA in SVGA astrocytes. Our results clearly suggest that gp120 and MA affect IL-6 but not IL-8 in a synergistic manner and this synergy was mediated by PI3K/Akt and NF-κB pathways. Inhibition of either of these pathways could abrogate the increased expression of IL-6 due to MA or gp120 alone, as well as the increased expression of IL-6 when the astrocytes were treated with both gp120 and MA. These results were confirmed by both, using chemical inhibitors/siRNA as well as western blotting. This study therefore provides novel information regarding the interaction between MA and gp120 in terms of the expression of IL-6 and the mechanisms underlying potential synergy between MA and gp120 in astrocytes.


Subject(s)
Astrocytes/drug effects , HIV Envelope Protein gp120/metabolism , Interleukin-6/biosynthesis , Interleukin-8/biosynthesis , Methamphetamine/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Astrocytes/metabolism , Cells, Cultured , Humans , MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/metabolism
8.
Expert Opin Drug Metab Toxicol ; 8(11): 1363-75, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22871069

ABSTRACT

INTRODUCTION: Alcohol consumption, which is highly prevalent in HIV-infected individuals, poses serious concerns in terms of rate of acquisition of HIV-1 infection, HIV-1 replication, response to highly active antiretroviral therapy (HAART) and AIDS/neuroAIDS progression. However, little is known about the mechanistic pathways by which alcohol exerts these effects, especially with respect to HIV-1 replication and the patient's response to HAART. AREAS COVERED: In this review, the authors discuss the effects of alcohol consumption on HIV-1 pathogenesis and its effect on HAART. They also describe the role of cytochrome P450 2E1 (CYP2E1) in alcohol-mediated oxidative stress and toxicity, and the role of CYP3A4 in the metabolism of drugs used in HAART (i.e., protease inhibitors (PI) and non-nucleoside reverse transcriptase inhibitors (NNRTI)). Based on the most recent findings the authors discuss the role of CYP2E1 in alcohol-mediated oxidative stress in monocytes/macrophages and astrocytes, as well as the role of CYP3A4 in alcohol-PI interactions leading to altered metabolism of PI in these cells. EXPERT OPINION: The authors propose that alcohol and PI/NNRTI interact synergistically in monocytes/macrophages and astrocytes through the CYP pathway leading to an increase in oxidative stress and a decrease in response to HAART. Ultimately, this exacerbates HIV-1 pathogenesis and neuroAIDS.


Subject(s)
Alcohol Drinking/adverse effects , Antiretroviral Therapy, Highly Active , Cytochrome P-450 CYP2E1/metabolism , Cytochrome P-450 CYP3A/metabolism , HIV-1/pathogenicity , Oxidative Stress/drug effects , Acquired Immunodeficiency Syndrome/drug therapy , Acquired Immunodeficiency Syndrome/metabolism , Drug Tolerance , HIV Infections/drug therapy , HIV Infections/metabolism , HIV Protease Inhibitors/therapeutic use , HIV-1/drug effects , HIV-1/physiology , Humans , Isoenzymes/metabolism , Liver/drug effects , Liver/metabolism , Protease Inhibitors/therapeutic use , Reverse Transcriptase Inhibitors/therapeutic use , Virus Replication
9.
Curr HIV Res ; 10(5): 369-83, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22591361

ABSTRACT

HIV-1 infection is a global public health problem with more than 34 million people living with HIV infection. Although great strides have been made in treating this epidemic with therapeutic agents, the increase in patient life span has been coincident with an increase in the prevalence of HIV-associated neurocognitive disorders (HAND). HAND is thought to result from the neurotoxic effects of viral proteins that are shed from HIV-infected microglial cells. One of the primary neurotoxins responsible for this effect is the HIV-1 glycoprotein gp120. Exposure of neurons to gp120 has been demonstrated to cause apoptosis in neurons, as well as numerous indirect effects such as an increase in inflammatory cytokines, an increase in oxidative stress, and an increase in permeability of the blood-brain barrier. In many patients, the use of drugs of abuse (DOA) exacerbates the neurotoxic effects of gp120. Cocaine, methamphetamine and morphine are three DOAs that are commonly used by those infected with HIV-1. All three of these DOAs have been demonstrated to increase oxidative stress in the CNS as well as to increase permeability of the blood-brain barrier. Numerous model systems have demonstrated that these DOAs have the capability of exacerbating the neurotoxic effects of gp120. This review will summarize the neurotoxic effects of gp120, the deleterious effects of cocaine, methamphetamine and morphine on the CNS, and the combined effects of gp120 in the context of these drugs.


Subject(s)
AIDS Dementia Complex/metabolism , Acquired Immunodeficiency Syndrome/metabolism , Central Nervous System Stimulants/adverse effects , Central Nervous System/metabolism , Cocaine/adverse effects , HIV Envelope Protein gp120/metabolism , Methamphetamine/adverse effects , Morphine/adverse effects , Substance-Related Disorders/metabolism , AIDS Dementia Complex/physiopathology , Acquired Immunodeficiency Syndrome/physiopathology , Animals , Apoptosis/drug effects , Blood-Brain Barrier/drug effects , Central Nervous System/drug effects , Central Nervous System/physiopathology , Central Nervous System Stimulants/pharmacology , Cocaine/pharmacology , HIV Envelope Protein gp120/drug effects , HIV-1/drug effects , Humans , Methamphetamine/pharmacology , Mice , Morphine/pharmacology , Oxidative Stress/drug effects , Rats , Substance-Related Disorders/complications
10.
Drug Alcohol Depend ; 125(1-2): 49-59, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22498344

ABSTRACT

BACKGROUND: Nicotine is known to generate oxidative stress through cytochrome P450 2A6 (CYP2A6)-mediated metabolism in the liver and other organs, including macrophages. This study has been designed to examine the role of CYP2A6 in nicotine metabolism and oxidative stress in SVGA cells, an immortalized human astrocyte cell line. METHODS: SVGA astrocytes were treated with 1 µM nicotine, followed by determination of mRNA and protein levels of several CYPs using quantitative RT-PCR and western blot analyses, respectively. Quantitation of nicotine and the nicotine metabolites, cotinine and nicotine-derived nitrosamine ketones (NNK), was performed using an LC-MS/MS method. The generation of reactive oxygen species (ROS) was measured using flow cytometry. RESULTS: Nicotine significantly upregulated mRNA and protein expression of the most abundantly expressed CYPs in SVGA astrocytes, CYP2A6 and CYP1A1. To characterize the metabolism of nicotine in astrocytes, a highly sensitive LC-MS/MS method was developed which is capable of quantifying very low concentrations of nicotine (0.3 ng/mL), cotinine and NNK (0.11 ng/mL). The LC-MS/MS results showed that nicotine is steadily metabolized to cotinine and NNK from 0.5 to 4h. Finally, we showed that nicotine initially causes an increase in ROS formation which is then gradually decreased, perhaps due to the increase in superoxide dismutase level. Nicotine metabolism and ROS formation by CYP2A6 were further confirmed by using tryptamine, a selective inhibitor of CYP2A6, which significantly lowered the levels of cotinine and NNK and inhibited ROS formation. CONCLUSIONS: CYP2A6 plays a key role in nicotine metabolism and oxidative stress in astrocytes, and this has implications in nicotine-associated brain toxicity.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Astrocytes/metabolism , Nicotine/analogs & derivatives , Nicotine/analysis , Nicotinic Agonists/analysis , Oxidative Stress/physiology , Aryl Hydrocarbon Hydroxylases/biosynthesis , Astrocytes/chemistry , Astrocytes/enzymology , Blotting, Western , Cell Line , Chromatography, High Pressure Liquid , Cotinine/analysis , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP2A6 , Enzyme Induction/drug effects , Humans , Ketones/analysis , Limit of Detection , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Nitrosamines/analysis , RNA/genetics , RNA/isolation & purification , Reactive Oxygen Species/analysis , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Tandem Mass Spectrometry
11.
PLoS One ; 7(3): e33822, 2012.
Article in English | MEDLINE | ID: mdl-22479453

ABSTRACT

The use of methamphetamine (MA) has increased in recent years, and is a major health concern throughout the world. The use of MA has been associated with an increased risk of acquiring HIV-1, along with an increased probability of the acquisition of various sexually transmitted infections. In order to determine the potential effects of MA exposure in the context of an infectious agent, U937 macrophages were exposed to various combinations of MA and bacterial lipopolysaccharide (LPS). Treatment with MA alone caused significant increases in the levels of TNF-α, while treatment with both MA and LPS resulted in significant increases in TNF-α, IL-1ß and the chemokine IL-8. The increases in cytokine or chemokine levels seen when cells were treated with both LPS and MA were generally greater than those increases observed when cells were treated with only LPS. Treatment with chemical inhibitors demonstrated that the signal transduction pathways including NF-kB, MAPK, and PI3-Akt were involved in mediating the increased inflammatory response. As discussed in the paper, these pathways appear to be utilized by both MA and LPS, in the induction of these inflammatory mediators. Since these pathways are involved in the induction of inflammation in response to other pathogens, this suggests that MA-exacerbated inflammation may be a common feature of infectious disease in MA abusers.


Subject(s)
Interleukin-1beta/metabolism , Interleukin-8/metabolism , Macrophages/drug effects , Macrophages/metabolism , Methamphetamine/pharmacology , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Cell Line , Cell Nucleus , Humans , Inflammation Mediators/metabolism , Lipopolysaccharides/immunology , Macrophages/immunology , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B p50 Subunit/antagonists & inhibitors , NF-kappa B p50 Subunit/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , Thiophenes/pharmacology
12.
J Neuroinflammation ; 9: 52, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22420994

ABSTRACT

Methamphetamine (MA) is one of the commonly used illicit drugs and the central nervous system toxicity of MA is well documented. The mechanisms contributing to this toxicity have not been fully elucidated. In this study, we investigated the effect of MA on the expression levels of the proinflammatory cytokines/chemokines, IL-6 and IL-8 in an astrocytic cell line. The IL-6 and IL-8 RNA levels were found to increase by 4.6 ± 0.2 fold and 3.5 ± 0.2 fold, respectively, after exposure to MA for three days. Exposure of astrocytes to MA for 24 hours also caused increased expression of IL-6 and IL-8 at the level of both RNA and protein. The potential involvement of the nuclear factor-Kappa B (NF-κB) pathway was explored as one of the possible mechanism(s) responsible for the increased induction of IL-6 and IL-8 by MA. The MA-mediated increases in IL-6 and IL-8 were significantly abrogated by SC514. We also found that exposure of astrocytes to MA results in activation of NF-κB through the phosphorylation of IκB-α, followed by translocation of active NF-κB from the cytoplasm to the nucleus. In addition, treatment of cells with a specific inhibitor of metabotropic glutamate receptor-5 (mGluR5) revealed that MA-mediated expression levels of IL-6 and IL-8 were abrogated by this treatment by 42.6 ± 5.8% and 65.5 ± 3.5%, respectively. Also, LY294002, an inhibitor of the Akt/PI3K pathway, abrogated the MA-mediated induction of IL-6 and IL-8 by 77.9 ± 6.6% and 81.4 ± 2.6%, respectively. Thus, our study demonstrates the involvement of an NF-κB-mediated signaling mechanism in the induction of IL-6 and IL-8 by MA. Furthermore, we showed that blockade of mGluR5 can protect astrocytes from MA-mediated increases of proinflammatory cytokines/chemokines suggesting mGluR5 as a potential therapeutic target in treating MA-mediated neurotoxicity.


Subject(s)
Astrocytes/drug effects , Astrocytes/metabolism , Dopamine Agents/pharmacology , Interleukin-6/metabolism , Interleukin-8/metabolism , Methamphetamine/pharmacology , NF-kappa B/metabolism , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction/drug effects , Cell Line, Transformed , Cell Survival/drug effects , Cytokines/metabolism , Enzyme Inhibitors/pharmacology , Humans , NF-kappa B/genetics , Oncogene Protein v-akt/metabolism , RNA, Messenger/metabolism , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/genetics
13.
Expert Rev Anti Infect Ther ; 9(11): 975-7, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22029515

ABSTRACT

This meeting was a special symposium sponsored by the American Society for Biochemistry and Molecular Biology. The conference was held in Gangzhou, China on 24-26 July 2011 and shared a venue with the Society of Chinese Bioscientists in America Thirteenth International Symposium. Over 150 participants from the Americas, Europe, Asia and Australia attended the meeting. This article focuses on two areas of research in which there have been exciting developments that have application to the development of antivirals: the regulation of host and viral mRNA by RNAi and NF-κB regulation of viral gene expression.


Subject(s)
DNA Replication/drug effects , MicroRNAs/pharmacology , Molecular Biology/methods , NF-kappa B/metabolism , RNA Interference , Virology/methods , Gene Expression Regulation, Viral/drug effects , Humans , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , RNA, Viral/antagonists & inhibitors , Signal Transduction , Virus Diseases/drug therapy , Virus Diseases/virology , Viruses/drug effects , Viruses/growth & development
14.
J Neurovirol ; 17(5): 401-15, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21786077

ABSTRACT

Over the past two decades methamphetamine (MA) abuse has seen a dramatic increase. The abuse of MA is particularly high in groups that are at higher risk for HIV-1 infection, especially men who have sex with men (MSM). This review is focused on MA toxicity in the CNS as well as in the periphery. In the CNS, MA toxicity is comprised of numerous effects, including, but not limited to, oxidative stress produced by dysregulation of the dopaminergic system, hyperthermia, apoptosis, and neuroinflammation. Multiple lines of evidence demonstrate that these effects exacerbate the neurodegenerative damage caused by CNS infection of HIV perhaps because both MA and HIV target the frontostriatal regions of the brain. MA has also been demonstrated to increase viral load in the CNS of SIV-infected macaques. Using transgenic animal models, as well as cultured cells, the HIV proteins Tat and gp120 have been demonstrated to have neurotoxic properties that are aggravated by MA. In addition, MA has been shown to exhibit detrimental effects on the blood-brain barrier (BBB) that have the potential to increase the probability of CNS infection by HIV. Although the effects of MA in the periphery have not been as extensively studied as have the effects on the CNS, recent reports demonstrate the potential effects of MA on HIV infection in the periphery including increased expression of HIV co-receptors and increased expression of inflammatory cytokines.


Subject(s)
Blood-Brain Barrier/drug effects , Central Nervous System/drug effects , HIV Infections/pathology , Illicit Drugs/toxicity , Methamphetamine/toxicity , Animals , Apoptosis , Brain/drug effects , Brain/virology , Cells, Cultured , Central Nervous System/virology , Dopamine/metabolism , Dopamine Agents , Fever/chemically induced , Fever/metabolism , HIV Envelope Protein gp120/metabolism , HIV Infections/immunology , HIV-1/physiology , Humans , Male , Oxidative Stress/drug effects , Viral Load/drug effects , Virus Replication , tat Gene Products, Human Immunodeficiency Virus/metabolism
15.
PLoS One ; 6(6): e21261, 2011.
Article in English | MEDLINE | ID: mdl-21712995

ABSTRACT

In addition to its role in virus entry, HIV-1 gp120 has also been implicated in HIV-associated neurocognitive disorders. However, the mechanism(s) responsible for gp120-mediated neuroinflammation remain undefined. In view of increased levels of IL-6 in HIV-positive individuals with neurological manifestations, we sought to address whether gp120 is involved in IL-6 over-expression in astrocytes. Transfection of a human astrocyte cell line with a plasmid encoding gp120 resulted in increased expression of IL-6 at the levels of mRNA and protein by 51.3±2.1 and 11.6±2.2 fold respectively; this effect of gp120 on IL-6 expression was also demonstrated using primary human fetal astrocytes. A similar effect on IL-6 expression was observed when primary astrocytes were treated with gp120 protein derived from different strains of X4 and R5 tropic HIV-1. The induction of IL-6 could be abrogated by use of gp120-specific siRNA. Furthermore, this study showed that the NF-κB pathway is involved in gp120-mediated IL-6 over-expression, as IKK-2 and IKKß inhibitors inhibited IL-6 expression by 56.5% and 60.8%, respectively. These results were also confirmed through the use of NF-κB specific siRNA. We also showed that gp120 could increase the phosphorylation of IκBα. Furthermore, gp120 transfection in the SVGA cells increased translocation of NF-κB from cytoplasm to nucleus. These results demonstrate that HIV-1 gp120-mediated over-expression of IL-6 in astrocytes is one mechanism responsible for neuroinflammation in HIV-infected individuals and this is mediated by the NF-κB pathway.


Subject(s)
HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/metabolism , Interleukin-6/metabolism , NF-kappa B/metabolism , RNA, Small Interfering/metabolism , Astrocytes/cytology , Astrocytes/physiology , Cell Line , Fetus/cytology , Humans , I-kappa B Proteins/metabolism , Interleukin-6/genetics , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , RNA, Small Interfering/genetics
16.
Alcohol Clin Exp Res ; 35(1): 132-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21039635

ABSTRACT

BACKGROUND: ATP-binding cassette (ABC) proteins and cytochrome P450 (CYP) enzymes regulate the bioavailability of HIV-1 antiretroviral therapeutic drugs, non-nucleoside reverse transcriptase inhibitors (NNRTIs), and protease inhibitors (PIs). They are also involved in regulating, and responding to, oxidative stress in various tissues and organs including liver. This study is designed to assess the effect of alcohol on the ABCC1 and CYP enzymes involved in the metabolism of NNRTIs and PIs (CYP2B6, CYP2D6, and CYP3A4) and oxidative stress (CYP1A1, CYP2A6, and CYP2E1) in U937 macrophages. The U937 cell line has been utilized as an in vitro model of human macrophages. METHODS: The expression levels of the ABCC1 and CYP enzymes in U937 macrophages were characterized in terms of mRNA quantification, protein analysis, and assays for functional activity. In addition, oxidative stress was monitored by measuring the activities of oxidative stress marker enzymes and production of reactive oxygen species (ROS). RESULTS: The order of mRNA expression in U937 macrophages was ABCC1 ∼ CYP2A6 > CYP3A4 ∼ CYP2E1 ∼ CYP1A1 > CYP2D6 > CYP2B6. Alcohol (100 mM) increased the mRNA levels of ABCC1 and CYP2A6 (200%), CYP2B6 and CYP3A4 (150%), and CYP2E1 (400%) compared with the control. Alcohol caused significant upregulation of ABCC1, CYP2A6, CYP2E1, and CYP3A4 proteins (50 to 85%) and showed >50% increase in the specific activity of CYP2A6 and CYP3A4 in U937 macrophages. Furthermore, alcohol increased the production of ROS and significantly enhanced the activity of oxidative stress marker enzymes, superoxide dismutase, and catalase in U937 macrophages. CONCLUSIONS: Our study showed that alcohol causes increases in the genetic and functional expressions of ABCC1 and CYP enzymes in U937 macrophages. This study has clinical implications in alcoholic HIV-1 individuals, because alcohol consumption is reported to reduce the therapeutic efficacy of NNRTIs and PIs and increases oxidative stress.


Subject(s)
Anti-HIV Agents/metabolism , Central Nervous System Depressants/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Macrophages/enzymology , Multidrug Resistance-Associated Proteins/metabolism , Ethanol/pharmacology , HIV Protease Inhibitors/metabolism , Humans , Macrophages/drug effects , Oxidative Stress , Reactive Oxygen Species/analysis , Reverse Transcriptase Inhibitors/metabolism , U937 Cells
17.
J Neuroimmune Pharmacol ; 5(1): 122-32, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20013315

ABSTRACT

HIV and simian immunodeficiency virus (SIV) have a formidable capacity for mutation and adaptation, a characteristic that has contributed to the extensive genetic variability. Evolutionary pressures imposed within the host and the viral capacity to mutate lead to the generation of such variants. To date, very little information is available regarding the evolution of HIV with drug abuse as a cofounding factor. Using our macaque model of drug dependency and AIDS, we have investigated the dynamics of SIV mutations in the genes tat, vpr, envelope, and nef. The results presented in this review, from our laboratory and others, contribute to the overall understanding of how drugs of abuse might influence immune selective pressure contribution to variation in different SIV genes. Additionally, the studies presented could help enlighten the development of HIV vaccines that take into consideration viral diversity.


Subject(s)
Acquired Immunodeficiency Syndrome/genetics , Disease Progression , Evolution, Molecular , Morphine Dependence/genetics , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Acquired Immunodeficiency Syndrome/complications , Animals , Disease Models, Animal , Genes, Regulator/genetics , Genes, env , Macaca , Morphine Dependence/complications , Morphine Dependence/virology , Mutation , Simian Acquired Immunodeficiency Syndrome/genetics
18.
J Neuroimmune Pharmacol ; 5(4): 516-20, 2010 Dec.
Article in English | MEDLINE | ID: mdl-19894120

ABSTRACT

Multidrug resistance-associated protein 1 (MRP-1) is a ubiquitously expressed member of the ATP-binding cassette transporter family. MRP-1 is one of the primary transporters of glutathione and glutathione conjugates. This protein also transports antiretroviral therapeutics, such as HIV-1 protease inhibitors (PI). We hypothesized that inflammatory mediators that activate macrophages would modify the expression and activity of MRP-1 in macrophages. Real-time PCR assays, western blots, and calcein efflux assays were used to show that exposure of macrophage cell line RAW 264.7 to lipopolysaccharide (LPS) increased expression of MRP-1 at the levels of mRNA, protein, and functional activity. Treatment of macrophages with LPS resulted in 2-fold increases of MRP-1 expression or functional activity. LPS-mediated increases in calcein efflux were repressed by the MRP-specific inhibitor MK-571. These results suggest that the effectiveness of HIV-1 PI therapy may be compromised by the presence of opportunistic infections.


Subject(s)
Lipopolysaccharides/metabolism , Macrophages/metabolism , Multidrug Resistance-Associated Proteins/biosynthesis , Animals , Blotting, Western , Cell Line , Lipopolysaccharides/immunology , Macrophages/immunology , Mice , Multidrug Resistance-Associated Proteins/immunology , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
19.
Fish Shellfish Immunol ; 23(2): 479-84, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17303437

ABSTRACT

Channel catfish virus (CCV) is a herpesvirus that infects channel catfish fry and fingerlings. Previous research has demonstrated that Type I interferons inhibit the expression of immediate-early (IE) genes of some mammalian herpesviruses. However, CCV is distantly related to the mammalian herpesviruses and Type I interferons from higher vertebrates exhibit only 20% similarity to fish interferons. In this work we demonstrate that treatment of channel catfish ovary (CCO) cells, a fibroblast-like cell line, with poly I:C, a known inducer of Type I interferons, results in inhibition of expression of the CCV IE gene ORF 1. Thus, although the genes involved have diverged, the mechanism appears to be conserved. If this paradigm holds true for other CCV IE-Type I interferon interactions, it could have important implications for the impact of CCV on the host immune system.


Subject(s)
Fish Diseases/virology , Genes, Immediate-Early/drug effects , Herpesviridae Infections/veterinary , Ictaluridae/immunology , Ictalurivirus/immunology , Poly I-C/pharmacology , Animals , Aquaculture , Cell Line , Female , Fibroblasts , Fish Diseases/genetics , Fish Diseases/immunology , Herpesviridae Infections/genetics , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Ictaluridae/genetics , Ictalurivirus/genetics , Interferon Type I/immunology , Open Reading Frames/drug effects , RNA, Viral/chemistry , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction/veterinary
20.
Methods Mol Med ; 122: 225-39, 2006.
Article in English | MEDLINE | ID: mdl-16511984

ABSTRACT

In vitro models have proven to be effective in studying the placental transporters that play a role in the exchange of nutrients, waste products, and drugs between the maternal and fetal circulations. Although primary cultures of trophoblast cells can be used to perform uptake, efflux, and metabolism studies, only the rodent HRP-1 and the human BeWo cell lines have been shown to form confluent monolayers when grown on semi-permeable membranes. Protocols for the revival, maintenance, passage, and growth of BeWo cells for transporter expression and transcellular transport studies are provided.


Subject(s)
Cell Culture Techniques/methods , Cell Line, Tumor/metabolism , Trophoblasts/metabolism , Choriocarcinoma , Female , Humans , Membrane Transport Proteins/metabolism , Models, Biological , Permeability , Placenta/metabolism , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...