Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
J Neurosci ; 37(23): 5758-5769, 2017 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-28473642

RESUMEN

Despite marked regional differences in HIV susceptibility within the CNS, there has been surprisingly little exploration into the differential vulnerability among neuron types and the circuits they underlie. The dorsal striatum is especially susceptible, harboring high viral loads and displaying marked neuropathology, with motor impairment a frequent manifestation of chronic infection. However, little is known about the response of individual striatal neuron types to HIV or how this disrupts function. Therefore, we investigated the morphological and electrophysiological effects of HIV-1 trans-activator of transcription (Tat) in dopamine subtype 1 (D1) and dopamine subtype 2 (D2) receptor-expressing striatal medium spiny neurons (MSNs) by breeding transgenic Tat-expressing mice to Drd1a-tdTomato- or Drd2-eGFP-reporter mice. An additional goal was to examine neuronal vulnerability early during the degenerative process to gain insight into key events underlying the neuropathogenesis. In D2 MSNs, exposure to HIV-1 Tat reduced dendritic spine density significantly, increased dendritic damage (characterized by swellings/varicosities), and dysregulated neuronal excitability (decreased firing at 200-300 pA and increased firing rates at 450 pA), whereas insignificant morphologic and electrophysiological consequences were observed in Tat-exposed D1 MSNs. These changes were concomitant with an increased anxiety-like behavioral profile (lower latencies to enter a dark chamber in a light-dark transition task, a greater frequency of light-dark transitions, and reduced rearing time in an open field), whereas locomotor behavior was unaffected by 2 weeks of Tat induction. Our findings suggest that D2 MSNs and a specific subset of neural circuits within the dorsal striatum are preferentially vulnerable to HIV-1.SIGNIFICANCE STATEMENT Despite combination antiretroviral therapy (cART), neurocognitive disorders afflict 30-50% of HIV-infected individuals and synaptodendritic injury remains evident in specific brain regions such as the dorsal striatum. A possible explanation for the sustained neuronal injury is that the neurotoxic HIV-1 regulatory protein trans-activator of transcription (Tat) continues to be expressed in virally suppressed patients on cART. Using inducible Tat-expressing transgenic mice, we found that dopamine subtype 2 (D2) receptor-expressing medium spiny neurons (MSNs) are selectively vulnerable to Tat exposure compared with D1 receptor-expressing MSNs. This includes Tat-induced reductions in D2 MSN dendritic spine density, increased dendritic damage, and disruptions in neuronal excitability, which coincide with elevated anxiety-like behavior. These data suggest that D2 MSNs and specific circuits within the basal ganglia are preferentially vulnerable to HIV-1.


Asunto(s)
Conducta Animal/fisiología , Cuerpo Estriado/metabolismo , Locomoción/fisiología , Receptores de Dopamina D1/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Animales , Espinas Dendríticas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Especificidad de Órganos , Receptores de Dopamina D2 , Distribución Tisular , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
2.
J Biol Chem ; 287(42): 35192-35200, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-22918829

RESUMEN

NMDA ionotropic glutamate receptors gate the cytoplasmic influx of calcium, which may, depending on the intensity of the stimulus, subserve either normal synaptic communication or cell death. We demonstrate that when isolated mitochondria are exposed to calcium and NMDA agonists, there is a significant increase in mitochondrial calcium levels. The agonist/antagonist response studies on purified mitochondria suggest the presence of a receptor on mitochondria with features similar to plasma membrane NMDA receptors. Immunogold electron microscopy of hippocampal tissue sections revealed extensive localization of NR2a subunit immunoreactivity on mitochondria. Transient transfection of neuronal GT1-7 cells with an NR1-NR2a NMDA receptor subunit cassette specifically targeting mitochondria resulted in a significant increase in mitochondrial calcium and neuroprotection against glutamate-induced cell death. Mitochondria prepared from GT1-7 cells in which the NR1 subunit of NMDA receptors was silenced demonstrated a decrease in calcium uptake. Our findings are the first to demonstrate that mitochondria express a calcium transport protein that shares characteristics with the NMDA receptor and may play a neuroprotective role.


Asunto(s)
Calcio/metabolismo , Proteínas Portadoras/metabolismo , Hipocampo/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Calcio/farmacología , Proteínas Portadoras/genética , Línea Celular , Hipocampo/ultraestructura , Masculino , Mitocondrias/genética , Mitocondrias/ultraestructura , Proteínas Mitocondriales/genética , Proteínas del Tejido Nervioso/genética , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/genética , Transfección
3.
Mitochondrion ; 59: 76-82, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33894358

RESUMEN

Our laboratory has demonstrated that functional N-methyl-d-aspartate-like receptors are present on neuronal mitochondria (NMDAm). This novel site gates the influx of Ca2+ and causes a several-fold increase in ATP levels. Although elevations in ATP in other cell types have been linked to increases in mitochondrial Ca2+, it has not been established whether the same holds true for calcium uptake via NMDAm. In this study, we have investigated the effect of NMDAm activation on a variety of bioenergetic parameters. Our findings suggest that mitochondrial bioenergetics are not only modulated by NMDAm activation in a Ca2+-dependent but also in a Ca2+-independent manner.


Asunto(s)
Calcio/metabolismo , Mitocondrias/metabolismo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Línea Celular , Complejo I de Transporte de Electrón/metabolismo , Complejo III de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Metabolismo Energético , Regulación de la Expresión Génica , Ratones , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Neuronas/citología
4.
J Neurotrauma ; 24(5): 895-908, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17518543

RESUMEN

Cerebral ischemia followed by reperfusion activates numerous pathways that lead to cell death. One such pathway involves the release of large quantities of the excitatory amino acid glutamate into the synapse and activation of N-methyl-D-aspartate receptors. This causes an increase in mitochondrial calcium levels ([Ca(2+)](m)) and a production of reactive oxygen species (ROS), both of which may induce the mitochondrial permeability transition (MPT). As a consequence, there is eventual mitochondrial failure culminating in either apoptotic or necrotic cell death. Thus, agents that inhibit MPT might prove useful as therapeutic interventions in cerebral ischemia. In this study, we have investigated the neuroprotective efficacy of the novel compound NIM811. Similar in structure of its parent compound cyclosporin A, NIM811 is a potent inhibitor of the MPT. Unlike cyclosporin A, however, it is essentially void of immunosuppressive actions, allowing the role of MPT to be clarified in ischemia/reperfusion injury. The results of these studies demonstrate that NIM811 provides almost 40% protection in a model of transient focal cerebral ischemia. This was associated with a nearly 10% reduction in mitochondrial reactive species formation and 34% and 38% reduction of cytochrome c release in core and penumbra, respectively. Treatment with NIM811 also increased calcium retention capacity by approximately 20%. Interestingly, NIM811 failed to improve ischemia-induced impairment of bioenergetics. The neuroprotective effects of NIM811 were not due to drug-induced alterations in cerebral perfusion after ischemia. Activation of MPT appears to be an important process in ischemia/reperfusion injury and may be a therapeutic target.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Ciclosporina/farmacología , Proteínas de Transporte de Membrana Mitocondrial/efectos de los fármacos , Degeneración Nerviosa/tratamiento farmacológico , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Animales , Infarto Encefálico/tratamiento farmacológico , Infarto Encefálico/metabolismo , Infarto Encefálico/fisiopatología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatología , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Ciclosporina/uso terapéutico , Citocromos c/antagonistas & inhibidores , Citocromos c/metabolismo , Modelos Animales de Enfermedad , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/fisiología , Masculino , Proteínas de Transporte de Membrana Mitocondrial/fisiología , Poro de Transición de la Permeabilidad Mitocondrial , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Neuronas/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Ratas , Ratas Endogámicas SHR , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Resultado del Tratamiento
5.
J Neurotrauma ; 24(5): 798-811, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17518535

RESUMEN

Following experimental traumatic brain injury (TBI), a rapid and significant necrosis occurs at the site of injury which coincides with significant mitochondrial dysfunction. The present study is driven by the hypothesis that TBI-induced glutamate release increases mitochondrial Ca(2+)cycling/overload, ultimately leading to mitochondrial dysfunction. Based on this premise, mitochondrial uncoupling during the acute phases of TBI-induced excitotoxicity should reduce mitochondrial Ca(2+) uptake (cycling) and reactive oxygen species (ROS) production since both are mitochondrial membrane potential dependent. In the present study, we utilized a cortical impact model of TBI to assess the potential use of mitochondrial uncouplers (2,4-DNP, FCCP) as a neuroprotective therapy. Young adult male rats were intraperitoneally administered vehicle (DMSO), 2,4-DNP (5 mg/kg), or FCCP (2.5 mg/kg) at 5 min post-injury. All animals treated with the uncouplers demonstrated a significant reduction in the amount of cortical damage and behavioral improvement following TBI. In addition, mitochondria isolated from the injured cortex at 3 or 6 h post-injury demonstrated that treatment with the uncouplers significantly improved several parameters of mitochondrial bioenergetics. These results demonstrate that post-injury treatment with mitochondrial uncouplers significantly (p < 0.01) increases cortical tissue sparing ( approximately 12%) and significantly (p< 0.01) improves behavioral outcome following TBI. The mechanism of neuroprotection most likely involves the maintenance of mitochondrial homeostasis by reducing mitochondrial Ca(2+) loading and subsequent mitochondrial dysfunction. These results further implicate mitochondrial dysfunction as an early event in the pathophysiology of TBI and that targeting acute mitochondrial events can result in long-term neuroprotection and improve behavioral outcome following brain injury.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Degeneración Nerviosa/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Desacopladores/farmacología , 2,4-Dinitrofenol/farmacología , 2,4-Dinitrofenol/uso terapéutico , Animales , Encéfalo/metabolismo , Encéfalo/fisiopatología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/fisiopatología , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Carbonil Cianuro p-Trifluorometoxifenil Hidrazona/farmacología , Carbonil Cianuro p-Trifluorometoxifenil Hidrazona/uso terapéutico , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Modelos Animales de Enfermedad , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/fisiología , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Masculino , Mitocondrias/metabolismo , Degeneración Nerviosa/etiología , Degeneración Nerviosa/prevención & control , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Resultado del Tratamiento , Desacopladores/uso terapéutico
7.
Ann N Y Acad Sci ; 1074: 178-90, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17105916

RESUMEN

The human immunodeficiency virus-1 (HIV-1) affects the central nervous system (CNS) in approximately 30% of infected individuals and basal ganglia structures seem to be most affected. The HIV-1-transactivating protein, Tat, has been suggested to be pathogenically relevant in HIV-1-induced neuronal injury. The abuse of methamphetamine (METH), which is great among this patient population, also affects the basal ganglia, causing degeneration of dopaminergic terminals. In previous studies, we demonstrated that coexposure to these two toxins caused a synergistic loss of striatal dopamine and binding to the dopamine transporter (DAT), suggesting a loss of dopamine terminals. Because the loss of dopamine and DAT, however, do not necessarily reflect dopamine terminal degeneration, we have used silver staining and TH immunohistochemistry to further examine this issue. We have also examined the glial reaction using GFAP as a marker of astrocyte activation and OX-42 as a marker of activated microglia. Lastly, we have begun to explore the mechanism of synergy by investigating the role that the cytokine TNF-alpha might play in Tat + METH synergy. Our data indicate that the synergistic loss of dopamine is likely the result of dopamine terminal degeneration. This injury is not a direct result of the number of activated glia but does involve TNF-alpha.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Dopamina/metabolismo , Productos del Gen tat/farmacología , Metanfetamina/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Cuerpo Estriado/metabolismo , Sinergismo Farmacológico , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuroglía/efectos de los fármacos , Ratas , Tinción con Nitrato de Plata , Factor de Necrosis Tumoral alfa/genética
8.
Neurosci Lett ; 623: 47-51, 2016 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-27132085

RESUMEN

N-methyl-d-aspartate (NMDA) receptors have long been known to be associated with the plasma membrane, providing a channel for the passage of extracellular Ca(2+) into the cytosol during synaptic transmission. Recent results from our laboratory indicate that in addition to this classic location, an NMDA-sensitive site (NMDAm) may also exist within the inner mitochondrial membrane. We report direct exposure of mitochondrial to NMDA enhances the production of reactive oxygen species and attenuate ROS-induced cytochrome c release, all the while slowing the rate of Ca(2+)-induced mitochondrial swelling. Treatment with NMDA did not alter the mitochondrial membrane potential. The findings of this study lend further support for the existence of NMDAm and suggest that this site may serve to stabilize mitochondrial function.


Asunto(s)
Mitocondrias/fisiología , N-Metilaspartato/metabolismo , Animales , Calcio/metabolismo , Línea Celular , Citocromos c/metabolismo , Potencial de la Membrana Mitocondrial , Ratones , Mitocondrias/efectos de los fármacos , Tamaño Mitocondrial , N-Metilaspartato/farmacología , Especies Reactivas de Oxígeno/metabolismo
9.
J Neurotrauma ; 22(10): 1142-9, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16238490

RESUMEN

It is now generally accepted that excitotoxic cell death involves bioenergetic failure resulting from the cycling of Ca2+ and the generation of reactive oxygen species (ROS) by mitochondria. Both Ca2+ cycling and ROS formation by mitochondria are dependent on the mitochondrial membrane potential (Deltapsi(m)) that results from the proton gradient that is generated across the inner membrane. Mitochondrial uncoupling refers to a condition in which protons cross the inner membrane back into the matrix while bypassing the ATP synthase. As a consequence of this "short-circuit," there is a reduction in Deltapsi(m). We have previously demonstrated that animals treated with the classic uncoupling agent 2,4-dinitrophenol (DNP) show significant protection against brain damage following striatal injections of the NMDA agonist quinolinic acid (QA). In an effort to elucidate the mechanism of neuroprotection, we have assessed the effects of DNP on several parameters of mitochondrial function caused by QA. The results presented herein demonstrate that treatment with DNP attenuates QA-induced increases in mitochondrial Ca2+ levels and ROS formation and also improves mitochondrial respiration. Our findings indicate that DNP may confer protection against acute brain injury involving excitotoxic pathways by mechanisms that maintain mitochondrial function.


Asunto(s)
2,4-Dinitrofenol/farmacología , Cuerpo Estriado/efectos de los fármacos , Homeostasis/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Desacopladores/farmacología , Animales , Respiración de la Célula/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/administración & dosificación , Inyecciones Intraventriculares , Masculino , Mitocondrias/metabolismo , Ácido Quinolínico/administración & dosificación , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo
10.
Biochem Pharmacol ; 63(8): 1499-505, 2002 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-11996892

RESUMEN

Administration of the mitochondrial inhibitors malonate and 3-nitropropionic acid (3-NP) to rats provides useful models of Huntington's disease. Exposure to these inhibitors has been shown to result in increased extracellular concentrations of striatal dopamine (DA), which is neurotoxic at high concentrations. The cause of this increase is unknown. The purpose of this study was to determine whether mitochondrial inhibition alters dopamine transporter (DAT) function. Striatal synaptosomes were incubated in the presence of several structurally unrelated inhibitors of mitochondrial Complexes I, II, and IV, and [(3)H]DA uptake was measured. Although all of the toxins inhibited [(3)H]DA uptake, there was a large variation in their inhibitory potencies, the rank order being rotenone>>cyanide>azide>3-NP>>malonate. Examination of the kinetic parameters of [(3)H]DA uptake revealed that inhibition was due to a reduction in maximum velocity (V(max)), with no change in affinity (K(m)). The addition of either ATP or of ADP plus P(i) to synaptosomes treated with 3-NP, or of the reactive oxygen species spin trap alpha-phenyl-N-tert-butyl nitrone to synaptosomes exposed to either malonate or cyanide failed to prevent mitochondrial toxin-induced inhibition of DAT function. The lack of effect of high energy substrates or of a free radical scavenger suggests that the mechanism by which extracellular DA is increased by several mitochondrial toxins involves factors other than mitochondrial ATP production or oxidative stress. Taken together, the results suggest that one mechanism whereby mitochondrial toxins increase extracellular concentrations of DA is via interaction with the DAT at a site other than the substrate site, i.e. noncompetitive inhibition of the DAT.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Dopamina/metabolismo , Malonatos/farmacología , Mitocondrias/efectos de los fármacos , Propionatos/farmacología , Sinaptosomas/efectos de los fármacos , Animales , Transporte Biológico , Cuerpo Estriado/metabolismo , Masculino , Mitocondrias/metabolismo , Nitrocompuestos , Ratas , Ratas Sprague-Dawley , Sinaptosomas/metabolismo , Tritio
11.
J Neurotrauma ; 21(10): 1396-404, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15672630

RESUMEN

The purpose of this study was to investigate the potential neuroprotective efficacy of the mitochondrial uncoupler 2,4-dinitrophenol (DNP) in rats following a mild to moderate spinal cord contusion injury. Animals received intraperitoneal injections of vehicle (DMSO) or 5 mg/mL of DNP prior to injury. Twenty-four hours following surgery, mitochondrial function was assessed in mitochondria isolated from spinal cord synaptosomes. In addition, synaptosomes were used to measure indicators of reactive oxygen species formation, lipid peroxidation, and protein oxidation. Relative to vehicle-treated animals, pretreatment with DNP maintained mitochondrial bioenergetics and significantly decreased reactive oxygen species levels, lipid peroxidation, and protein carbonyl content following spinal cord injury. Furthermore, pretreatment with DNP significantly increased the amount of remaining white matter at the injury epicenter 6 weeks after injury. These results indicate that treatment with mitochondrial uncoupling agents may provide a novel approach for the treatment of secondary injury following spinal cord contusion.


Asunto(s)
2,4-Dinitrofenol/farmacología , Mitocondrias/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Traumatismos de la Médula Espinal/prevención & control , Desacopladores/farmacología , Animales , Modelos Animales de Enfermedad , Femenino , Peroxidación de Lípido/efectos de los fármacos , Ratas , Ratas Long-Evans , Especies Reactivas de Oxígeno/metabolismo , Traumatismos de la Médula Espinal/patología
12.
Brain Res ; 966(2): 312-6, 2003 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-12618355

RESUMEN

We have examined the neuroprotective efficacy of the mitochondrial uncoupler 2,4-dinitrophenol (DNP) in animals receiving striatal injections of the neurotoxin quinolinic acid. Animals administered DNP either 1 h before or 3 h following QA infusion developed lesions that were 25% smaller than control animals. Animals treated with the DNP analogue 2,4,6-trinitrophenol, which does not possess uncoupling activity in intact mitochondria, showed no neuroprotection. These results indicate that DNP, and other compounds that diminish the mitochondrial membrane potential, might provide a novel approach to the treatment of acute neurological injury.


Asunto(s)
2,4-Dinitrofenol/farmacología , Cuerpo Estriado/efectos de los fármacos , Ácido Quinolínico/toxicidad , Desacopladores/farmacología , 2,4-Dinitrofenol/análogos & derivados , Análisis de Varianza , Animales , Temperatura Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Masculino , Picratos/farmacología , Ratas , Ratas Sprague-Dawley
13.
Brain Res ; 984(1-2): 133-42, 2003 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-12932847

RESUMEN

HIV-1 infection of the brain can lead to the development of clinical syndromes reminiscent of Parkinson's disease, suggesting that HIV infection may damage nigrostriatal dopamine (DA) neurons. Although the responsible mechanisms have not been well defined, neurotoxic viral proteins, such as Tat, released from infected cells may be involved. Drug abuse is a major risk factor for contracting HIV infection. Methamphetamine (METH), a psychostimulant with high abuse potential, may also be toxic to brain DA neurons. Thus, the combination of METH abuse and HIV infection may lead to substantial alterations in DA neuron functioning. The present experiments examined how Tat, alone and with METH, affects DA release in the striatum. Male rats were given an intrastriatal injection of Tat (25 micro g) or vehicle 24 h before treatment with saline or neurotoxic doses of METH. Seven days later microdialysis studies were carried out to measure potassium- and amphetamine-evoked overflow of DA from the striatum. The Tat treatment alone led to no change in potassium-evoked overflow of DA, a 20% decrease in amphetamine-evoked overflow of DA, and a 16% decrease in striatal DA content. The METH alone led to a 37-42% decrease in striatal DA overflow and content. The combined treatment with Tat and METH led to significantly greater 70-78% decreases in striatal DA overflow and content. These results indicate that Tat enhances METH-induced reductions in striatal DA release and content, possibly in a synergistic manner, and suggest that METH abusers infected with HIV may be at increased risk for basal ganglia dysfunction.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Productos del Gen tat/farmacología , Metanfetamina/farmacología , Animales , Sinergismo Farmacológico , Masculino , Ratas , Ratas Endogámicas F344 , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
14.
Curr HIV Res ; 5(3): 301-13, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17504172

RESUMEN

HIV-1 infection with concurrent methamphetamine (MA) abuse results in exacerbated neurodegenerative changes and rapid progression of a form of sub-cortical dementia termed HIV-1 associated dementia (HAD). A notable feature of HAD is the involvement of the dopaminergic system manifested as parkinsonian like movement abnormalities. The HIV-1 transactivator of transcription (Tat) protein is very often used in experimental studies trying to understand neurotoxic consequences of HIV-1 infection, since the pathophysiological changes induced by Tat mirrors, in part, the means by which HIV-1 infection of the nervous system results in neuronal damage. Understanding the interaction of Tat and MA in the basal ganglia and the resultant injury to the dopaminergic system in rodent models as well as cell culture will shed light on the dopaminergic pathology occurring in HIV-1 infected-MA abusers. The aim of this review is to update the reader on the current knowledge of MA and HIV-1 neurotoxicity, specifically Tat, and discuss the progress in understanding how MA synergizes with the HIV-1 transactivator protein Tat to damage the basal ganglia.


Asunto(s)
Complejo SIDA Demencia/etiología , Ganglios Basales/fisiopatología , Dopaminérgicos/farmacología , Infecciones por VIH/complicaciones , VIH-1 , Metanfetamina/farmacología , Complejo SIDA Demencia/fisiopatología , Trastornos Relacionados con Anfetaminas/complicaciones , Ganglios Basales/efectos de los fármacos , Ganglios Basales/virología , Estimulantes del Sistema Nervioso Central/farmacología , Productos del Gen tat/fisiología , Humanos , Enfermedades Neurodegenerativas/etiología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
15.
J Neurochem ; 100(3): 567-86, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17173547

RESUMEN

Neuronal dysfunction and degeneration are ultimately responsible for the neurocognitive impairment and dementia manifest in neuroAIDS. Despite overt neuronal pathology, HIV-1 does not directly infect neurons; rather, neuronal dysfunction or death is largely an indirect consequence of disrupted glial function and the cellular and viral toxins released by infected glia. A role for glia in HIV-1 neuropathogenesis is revealed in experimental and clinical studies examining substance abuse-HIV-1 interactions. Current evidence suggests that glia are direct targets of substance abuse and that glia contribute markedly to the accelerated neurodegeneration seen with substance abuse in HIV-1 infected individuals. Moreover, maladaptive neuroplastic responses to chronic drug abuse might create a latent susceptibility to CNS disorders such as HIV-1. In this review, we consider astroglial and microglial interactions and dysfunction in the pathogenesis of HIV-1 infection and examine how drug actions in glia contribute to neuroAIDS.


Asunto(s)
Complejo SIDA Demencia/fisiopatología , Encéfalo/fisiopatología , VIH-1/metabolismo , Degeneración Nerviosa/fisiopatología , Neuroglía/metabolismo , Trastornos Relacionados con Sustancias/fisiopatología , Complejo SIDA Demencia/complicaciones , Complejo SIDA Demencia/patología , Animales , Encéfalo/patología , Encéfalo/virología , Causalidad , Comunicación Celular/efectos de los fármacos , Comunicación Celular/fisiología , VIH-1/inmunología , Humanos , Degeneración Nerviosa/patología , Degeneración Nerviosa/virología , Neuroglía/patología , Neuroglía/virología , Psicotrópicos/efectos adversos , Trastornos Relacionados con Sustancias/complicaciones , Trastornos Relacionados con Sustancias/patología
16.
Exp Neurol ; 199(2): 490-8, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16510141

RESUMEN

Human immunodeficiency virus-1 (HIV-1) infection of the brain causes elevation in pro-inflammatory cytokines and inflammatory changes in the striatum. HIV-1-infected individuals who also abuse drugs including the psychostimulant methamphetamine (MA) develop more severe encephalitis and neuronal damage compared to HIV-1-infected patients who do not abuse drugs. In previous studies, we demonstrated that the HIV-1 protein Tat and MA interacted to cause enhanced loss of dopamine in the rat striatum via the destruction of dopaminergic terminals. Since both Tat and MA activate glia and induce cytokine production, we investigated the role of cytokines in the synergistic neurotoxicity induced by Tat and MA using cytokine arrays. Significant increases in monocyte chemotactic protein (MCP-1), interleukin-1 alpha (IL-1alpha) and tissue inhibitor of metalloproteinase-1 (TIMP-1) levels were noted 4 h following Tat + MA treatment compared to saline, Tat or MA. MCP-1 and TIMP-1 levels remained elevated 16 h after Tat + MA compared to saline or MA but were not different from the Tat-treated group at this time point. Weak, but significant elevations in cytokine-induced neutrophil chemoattractant-3 (CINC-3), ciliary neurotrophic factor (CNTF) and macrophage inflammatory protein-3 alpha (MIP-3alpha) were also noted with Tat + MA. The interaction of Tat and MA was prevented in mice genetically deficient in MCP-1 with a consequent attenuation of Tat + MA neurotoxicity. Our findings suggest that HIV-1 infection with concurrent drug abuse might profoundly increase chemokine levels in the striatum resulting in enhanced damage to the dopaminergic system.


Asunto(s)
Cuerpo Estriado , Citocinas/metabolismo , Inhibidores de Captación de Dopamina/administración & dosificación , Productos del Gen tat/metabolismo , VIH-1/química , Metanfetamina/administración & dosificación , Análisis de Varianza , Animales , Western Blotting/métodos , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Cuerpo Estriado/virología , Regulación de la Expresión Génica/fisiología , Proteína Ácida Fibrilar de la Glía/metabolismo , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/patogenicidad , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
17.
Neurobiol Dis ; 23(3): 663-8, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16828290

RESUMEN

Our previous studies demonstrated that the psychostimulant methamphetamine (MA) and the human immunodeficiency virus-1 (HIV-1) protein Tat interacted to cause enhanced dopaminergic neurotoxicity. The present study examined whether tumor necrosis factor-alpha (TNF-alpha) mediates the interaction between Tat and MA. In Sprague-Dawley rats, injections of Tat caused a small but significant increase in striatal TNF-alpha level, whereas MA resulted in no change. The increase in TNF-alpha induced by Tat + MA was not significantly different from that induced by Tat alone. Temporal analysis of TNF-alpha levels revealed a 50-fold increase 4 h after Tat administration. In C57BL/6 mice, Tat + MA induced a 50% decline in striatal dopamine levels, which was significantly attenuated in mice lacking both receptors for TNF-alpha. TNF-alpha synthesis inhibitors significantly attenuated Tat + MA neurotoxicity in hippocampal neuronal culture. The results suggest that Tat-induced elevation of TNF-alpha may predispose the dopaminergic terminals to subsequent damage by MA.


Asunto(s)
Encefalitis/inmunología , Productos del Gen tat/toxicidad , Metanfetamina/toxicidad , Estrés Oxidativo/inmunología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Complejo SIDA Demencia/inmunología , Complejo SIDA Demencia/fisiopatología , Complejo SIDA Demencia/virología , Trastornos Relacionados con Anfetaminas/inmunología , Trastornos Relacionados con Anfetaminas/metabolismo , Trastornos Relacionados con Anfetaminas/fisiopatología , Animales , Ganglios Basales/efectos de los fármacos , Ganglios Basales/inmunología , Ganglios Basales/virología , Células Cultivadas , Modelos Animales de Enfermedad , Dopamina/metabolismo , Inhibidores de Captación de Dopamina/toxicidad , Encefalitis/fisiopatología , Encefalitis/virología , Productos del Gen tat/inmunología , Productos del Gen tat/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo/efectos de los fármacos , Terminales Presinápticos/efectos de los fármacos , Terminales Presinápticos/inmunología , Terminales Presinápticos/virología , Inhibidores de la Síntesis de la Proteína/farmacología , Ratas , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
18.
J Neuroimmune Pharmacol ; 1(4): 351-99, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18040811

RESUMEN

Neurological complications of human immunodeficiency virus (HIV) infection are a public health problem despite the availability of active antiretroviral therapies. The neuropathogenesis of HIV infection revolves around a complex cascade of events that include viral infection and glial immune activation, monocyte-macrophage brain infiltration, and secretion of a host of viral and cellular inflammatory and neurotoxic molecules. Although there is evidence that HIV-infected drug abusers experience more severe neurological disease, the biological basis for this finding is unknown. A scientific workshop organized by the National Institute on Drug Abuse (NIDA) was held on March 23-24, 2006 to address this question. The goal of the meeting was to bring together basic science and clinical researchers who are experts in NeuroAIDS, glial immunity, drugs of abuse, and/or pharmacology in order to find new approaches to understanding interactions between drug abuse and neuroAIDS. The format of the meeting was designed to stimulate open discussion and forge new multidisciplinary research collaborations. This report includes transcripts of active discussions and short presentations from invited participants. The presentations were separated into sections that included: Glial Biology, Inflammation, and HIV; Pharmacology, Neurotoxicology, and Neuroprotection; NeuroAIDS and Virology; and Virus-Drug and Immune-Drug Interactions. Research priorities were identified. Additional information about this meeting is available through links from the NIDA AIDS Research Program website ( http://www.nida.nih.gov/about/organization/arp/arp-websites.htm ).


Asunto(s)
Complejo SIDA Demencia/complicaciones , Complejo SIDA Demencia/patología , Investigación Biomédica/métodos , Investigación Biomédica/tendencias , Encéfalo/patología , Conducta Cooperativa , Trastornos Relacionados con Sustancias/complicaciones , Trastornos Relacionados con Sustancias/patología , Complejo SIDA Demencia/inmunología , Complejo SIDA Demencia/terapia , Animales , Humanos , Inflamación/complicaciones , Inflamación/inmunología , Inflamación/terapia , Trastornos Relacionados con Sustancias/inmunología , Trastornos Relacionados con Sustancias/terapia
19.
Neurochem Res ; 30(4): 527-31, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16076022

RESUMEN

Huntington's disease (HD) is a progressive neurodegenerative disorder, of which the pathogenesis is not completely understood. In patients with Huntington's disease, there is a mutation in the gene encoding the protein huntingtin, which results in an expanded polyglutamine sequence leading to degeneration of the basal ganglia. There is mounting evidence that metabolism of the transmitter dopamine by the enzyme monoamine oxidase may contribute to striatal damage in mitochondrial toxin-induced models of HD. In this study, we have examined the role of the catecholamine tyramine in neural SH-SY5Y cells transfected with normal and expanded polyglutamine repeat numbers. Our findings demonstrate that cells containing a pathological number of polyglutamines are more sensitive to tyramine than cells with a non-pathological number. Tyramine-induced cell death was attenuated by MAO inhibitors as well as with catalase and the iron chelator deferoxamine, suggesting that H202 might mediate the observed toxicity. These observations support the notion that the metabolism of dopamine plays a role in neuron death in Huntington's disease.


Asunto(s)
Péptidos/genética , Tiramina/toxicidad , Catalasa/farmacología , Línea Celular , ADN/biosíntesis , ADN/genética , Deferoxamina/farmacología , Dopamina/metabolismo , Dopamina/fisiología , Humanos , Enfermedad de Huntington/patología , Peróxido de Hidrógeno/metabolismo , Quelantes del Hierro/farmacología , Isoenzimas/metabolismo , Monoaminooxidasa/metabolismo , Inhibidores de la Monoaminooxidasa/farmacología , Neuronas/efectos de los fármacos , Transfección
20.
J Neurochem ; 94(6): 1676-84, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16045446

RESUMEN

Ischemic stroke is caused by acute neuronal degeneration provoked by interruption of cerebral blood flow. Although the mechanisms contributing to ischemic neuronal degeneration are myriad, mitochondrial dysfunction is now recognized as a pivotal event that can lead to either necrotic or apoptotic neuronal death. Lack of suitable 'upstream' targets to prevent loss of mitochondrial homeostasis has, so far, restricted the development of mechanistically based interventions to promote neuronal survival. Here, we show that the uncoupling agent 2,4 dinitrophenol (DNP) reduces infarct volume approximately 40% in a model of focal ischemia-reperfusion injury in the rat brain. The mechanism of protection involves an early decrease in mitochondrial reactive oxygen species formation and calcium uptake leading to improved mitochondrial function and a reduction in the release of cytochrome c into the cytoplasm. The observed effects of DNP were not associated with enhanced cerebral perfusion. These findings indicate that compounds with uncoupling properties may confer neuroprotection through a mechanism involving stabilization of mitochondrial function.


Asunto(s)
2,4-Dinitrofenol/farmacología , Infarto Cerebral/tratamiento farmacológico , Ataque Isquémico Transitorio/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Degeneración Nerviosa/tratamiento farmacológico , Neuronas/efectos de los fármacos , Animales , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Infarto Cerebral/fisiopatología , Infarto Cerebral/prevención & control , Citocromos c/metabolismo , Citoprotección/efectos de los fármacos , Citoprotección/fisiología , Modelos Animales de Enfermedad , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Ataque Isquémico Transitorio/metabolismo , Ataque Isquémico Transitorio/fisiopatología , Masculino , Mitocondrias/metabolismo , Degeneración Nerviosa/fisiopatología , Degeneración Nerviosa/prevención & control , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Desacopladores/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA