Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Curr HIV Res ; 12(4): 234-42, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25053368

RESUMEN

According to a survey from the HIV Cost and Services Utilization Study (HCSUS), approximately 53% of HIV-infected patients reported drinking alcohol and 8% were classified as heavy drinkers. The role of alcohol as a risk factor for HIV infection has been widely studied and recent research has found a significant association between heavy alcohol consumption and lower levels of CD4 T cells among HIV-infected alcoholics. Although there is evidence on the role of alcohol as a risk factor for HIV transmission and disease progression, there is a need for population studies to determine the genetic mechanisms that affect alcohol's role in HIV disease progression. One of the mechanisms of interest is the dopaminergic system. To date, the effects of dopamine on HIV neuroimmune pathogenesis are not well understood; however, dopaminergic neural degeneration due to HIV is known to occur by viral invasion into the brain via immune cells, and modulation of dopamine in the CNS may be a common mechanism by which different types of substances of abuse impact HIV disease progression. Although previous studies have shown an association of D(2) dopamine receptor (DRD2) polymorphisms with severity of alcohol dependence, the expression of this allele risk on HIV patients with alcohol dependence has not been systematically explored. In the current study, DRD2 Taq1A and C957T SNP genotyping analyses were performed in 165 HIV-infected alcohol abusers and the results were examined with immune status and CD4 counts.


Asunto(s)
Alcoholismo/complicaciones , Alcoholismo/genética , Infecciones por VIH/complicaciones , Infecciones por VIH/inmunología , Polimorfismo de Nucleótido Simple , Receptores de Dopamina D2/genética , Recuento de Linfocito CD4 , Predisposición Genética a la Enfermedad , Humanos
2.
Neurochem Int ; 61(5): 807-14, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21756955

RESUMEN

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.


Asunto(s)
Complejo SIDA Demencia/metabolismo , Elementos de Respuesta Antioxidante/fisiología , Astrocitos/metabolismo , Regulación Viral de la Expresión Génica , Proteína gp120 de Envoltorio del VIH/fisiología , Factor 2 Relacionado con NF-E2/biosíntesis , Complejo SIDA Demencia/virología , Células Cultivadas , Humanos , Factor 2 Relacionado con NF-E2/fisiología , Estrés Oxidativo/fisiología
3.
Alcohol Clin Exp Res ; 35(8): 1550-6, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21447001

RESUMEN

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.


Asunto(s)
Depresores del Sistema Nervioso Central/toxicidad , Etanol/toxicidad , Histona Desacetilasa 2/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Fármacos Neuroprotectores/farmacología , Línea Celular , Relación Dosis-Respuesta a Droga , Expresión Génica/efectos de los fármacos , Histona Desacetilasa 2/efectos de los fármacos , Histona Desacetilasa 2/genética , Humanos , Ácidos Hidroxámicos/metabolismo , Fármacos Neuroprotectores/metabolismo , Especies Reactivas de Oxígeno/análisis , Especies Reactivas de Oxígeno/metabolismo
4.
Neurochem Int ; 58(6): 656-64, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21315782

RESUMEN

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.


Asunto(s)
Complejo SIDA Demencia/enzimología , Productos del Gen tat/fisiología , VIH-1/metabolismo , Histona Desacetilasa 2/metabolismo , Neuronas/enzimología , Línea Celular Tumoral , Citometría de Flujo , Productos del Gen tat/antagonistas & inhibidores , Humanos , Ácidos Hidroxámicos/farmacología , Reacción en Cadena de la Polimerasa , Regulación hacia Arriba
5.
J Neurovirol ; 16(4): 294-305, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20624003

RESUMEN

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.


Asunto(s)
Complejo SIDA Demencia/fisiopatología , Barrera Hematoencefálica/patología , Cocaína/toxicidad , Inhibidores de Captación de Dopamina/toxicidad , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/farmacología , Complejo SIDA Demencia/patología , Complejo SIDA Demencia/virología , Astrocitos/efectos de los fármacos , Astrocitos/patología , Astrocitos/virología , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/virología , Western Blotting , Permeabilidad Capilar/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Células Endoteliales/virología , Expresión Génica , Perfilación de la Expresión Génica , VIH-1/genética , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/patología , Uniones Estrechas/virología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA