Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Int Neuropsychol Soc ; 30(1): 84-93, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37553288

RESUMEN

OBJECTIVE: Methamphetamine and cannabis are two widely used, and frequently co-used, substances with possibly opposing effects on the central nervous system. Evidence of neurocognitive deficits related to use is robust for methamphetamine and mixed for cannabis. Findings regarding their combined use are inconclusive. We aimed to compare neurocognitive performance in people with lifetime cannabis or methamphetamine use disorder diagnoses, or both, relative to people without substance use disorders. METHOD: 423 (71.9% male, aged 44.6 ± 14.2 years) participants, stratified by presence or absence of lifetime methamphetamine (M-/M+) and/or cannabis (C-/C+) DSM-IV abuse/dependence, completed a comprehensive neuropsychological, substance use, and psychiatric assessment. Neurocognitive domain T-scores and impairment rates were examined using multiple linear and binomial regression, respectively, controlling for covariates that may impact cognition. RESULTS: Globally, M+C+ performed worse than M-C- but better than M+C-. M+C+ outperformed M+C- on measures of verbal fluency, information processing speed, learning, memory, and working memory. M-C+ did not display lower performance than M-C- globally or on any domain measures, and M-C+ even performed better than M-C- on measures of learning, memory, and working memory. CONCLUSIONS: Our findings are consistent with prior work showing that methamphetamine use confers risk for worse neurocognitive outcomes, and that cannabis use does not appear to exacerbate and may even reduce this risk. People with a history of cannabis use disorders performed similarly to our nonsubstance using comparison group and outperformed them in some domains. These findings warrant further investigation as to whether cannabis use may ameliorate methamphetamine neurotoxicity.


Asunto(s)
Trastornos Relacionados con Anfetaminas , Cannabis , Trastornos del Conocimiento , Metanfetamina , Humanos , Masculino , Femenino , Metanfetamina/efectos adversos , Cannabis/efectos adversos , Trastornos del Conocimiento/etiología , Trastornos Relacionados con Anfetaminas/complicaciones , Pruebas Neuropsicológicas
2.
J Neuroinflammation ; 14(1): 49, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28279172

RESUMEN

BACKGROUND: Astrocyte activation is one of the earliest findings in the brain of methamphetamine (Meth) abusers. Our goal in this study was to identify the characteristics of the astrocytic acute response to the drug, which may be critical in pathogenic outcomes secondary to the use. METHODS: We developed an integrated analysis of gene expression data to study the acute gene changes caused by the direct exposure to Meth treatment of astrocytes in vitro, and to better understand how astrocytes respond, what are the early molecular markers associated with this response. We examined the literature in search of similar changes in gene signatures that are found in central nervous system disorders. RESULTS: We identified overexpressed gene networks represented by genes of an inflammatory and immune nature and that are implicated in neuroactive ligand-receptor interactions. The overexpressed networks are linked to molecules that were highly upregulated in astrocytes by all doses of methamphetamine tested and that could play a role in the central nervous system. The strongest overexpressed signatures were the upregulation of MAP2K5, GPR65, and CXCL5, and the gene networks individually associated with these molecules. Pathway analysis revealed that these networks are involved both in neuroprotection and in neuropathology. We have validated several targets associated to these genes. CONCLUSIONS: Gene signatures for the astrocytic response to Meth were identified among the upregulated gene pool, using an in vitro system. The identified markers may participate in dysfunctions of the central nervous system but could also provide acute protection to the drug exposure. Further in vivo studies are necessary to establish the role of these gene networks in drug abuse pathogenesis.


Asunto(s)
Astrocitos/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Metanfetamina/farmacología , Animales , Astrocitos/fisiología , Células Cultivadas , Expresión Génica/fisiología , Redes Reguladoras de Genes/fisiología , Ratas , Ratas Sprague-Dawley
3.
Brain Behav Immun ; 65: 210-221, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28495611

RESUMEN

Methamphetamine abuse is common among humans with immunodeficiency virus (HIV). The HIV-1 regulatory protein TAT induces dysfunction of mesolimbic dopaminergic systems which may result in impaired reward processes and contribute to methamphetamine abuse. These studies investigated the impact of TAT expression on methamphetamine-induced locomotor sensitization, underlying changes in dopamine function and adenosine receptors in mesolimbic brain areas and neuroinflammation (microgliosis). Transgenic mice with doxycycline-induced TAT protein expression in the brain were tested for locomotor activity in response to repeated methamphetamine injections and methamphetamine challenge after a 7-day abstinence period. Dopamine function in the nucleus accumbens (Acb) was determined using high performance liquid chromatography. Expression of dopamine and/or adenosine A receptors (ADORA) in the Acb and caudate putamen (CPu) was assessed using RT-PCR and immunohistochemistry analyses. Microarrays with pathway analyses assessed dopamine and adenosine signaling in the CPu. Activity-dependent neurotransmitter switching of a reserve pool of non-dopaminergic neurons to a dopaminergic phenotype in the ventral tegmental area (VTA) was determined by immunohistochemistry and quantified with stereology. TAT expression enhanced methamphetamine-induced sensitization. TAT expression alone decreased striatal dopamine (D1, D2, D4, D5) and ADORA1A receptor expression, while increasing ADORA2A receptors expression. Moreover, TAT expression combined with methamphetamine exposure was associated with increased adenosine A receptors (ADORA1A) expression and increased recruitment of dopamine neurons in the VTA. TAT expression and methamphetamine exposure induced microglia activation with the largest effect after combined exposure. Our findings suggest that dopamine-adenosine receptor interactions and reserve pool neuronal recruitment may represent potential targets to develop new treatments for methamphetamine abuse in individuals with HIV.


Asunto(s)
Metanfetamina/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/fisiología , Animales , Dopamina/metabolismo , Dopaminérgicos/metabolismo , Neuronas Dopaminérgicas/metabolismo , Productos del Gen tat , VIH-1 , Humanos , Locomoción/efectos de los fármacos , Masculino , Metanfetamina/efectos adversos , Metanfetamina/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos/metabolismo , Núcleo Accumbens/efectos de los fármacos , Recompensa , Área Tegmental Ventral/efectos de los fármacos
4.
Mediators Inflamm ; 2017: 7582437, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28811681

RESUMEN

Osteopontin (OPN) is a molecule that is common in central nervous system (CNS) pathologies, which participates in the activation, migration, and survival of inflammatory cells. However, the mechanisms by which OPN modulates inflammatory pathways are not clear. To understand the role of OPN in CNS viral infections, we used a lethal mouse model of West Nile virus (WNV), characterized by the injection of high doses of the Eg101 strain of WNV, causing the increase of OPN levels in the brain since early time points. To measure the impact of OPN in neuropathogenesis and resistance, we compared C57BI/6 WT with mice lacking the OPN gene (OPN KO). OPN KO presented a significantly higher mortality compared to WT mice, detectable since day 5 pi. Our data suggests that OPN expression at early time points may provide protection against viral spread in the CNS by negatively controlling the type I IFN-sensitive, caspase 1-dependent inflammasome, while promoting an alternative caspase 8-associated pathway, to control the apoptosis of infected cells during WNV infection in the CNS. Overall, we conclude that the expression of OPN maintains a critical threshold in the innate immune response that controls apoptosis and lethal viral spread in early CNS infection.


Asunto(s)
Sistema Nervioso Central/metabolismo , Inflamasomas/metabolismo , Osteopontina/metabolismo , Fiebre del Nilo Occidental/metabolismo , Fiebre del Nilo Occidental/patología , Animales , Encéfalo/inmunología , Encéfalo/metabolismo , Muerte Celular/inmunología , Sistema Nervioso Central/inmunología , Inflamasomas/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteopontina/genética , Fiebre del Nilo Occidental/inmunología
5.
J Infect Dis ; 211(1): 40-4, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25035516

RESUMEN

Human immunodeficiency virus (HIV) accesses the brain early in infection and can lead to neurocognitive disorders. The brain can also serve as a viral reservoir, but how virus is controlled in the brain is unknown. To examine this, CD8-depleting monoclonal antibody was injected into the cerebrospinal fluid of rhesus monkeys with chronic simian immunodeficiency virus (SIV) infection. This treatment led to the rapid increase of SIV in the brain. Virus in the brain is maintained by active suppression from the host immune system. This dynamic interaction can be manipulated in efforts to control and eradicate virus from the brain and other reservoirs.


Asunto(s)
Encéfalo/inmunología , Encéfalo/virología , Linfocitos T CD8-positivos/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Macaca mulatta/inmunología , Macaca mulatta/virología , Síndrome de Inmunodeficiencia Adquirida del Simio/líquido cefalorraquídeo
6.
J Biol Chem ; 289(51): 35149-58, 2014 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-25368325

RESUMEN

Sialic acid terminates glycans of glycoproteins and glycolipids that play numerous biological roles in health and disease. Although genetic tools are available for interrogating the effects of decreased or abolished sialoside expression in mice, pharmacological inhibition of the sialyltransferase family has, to date, not been possible. We have recently shown that a sialic acid analog, 2,4,7,8,9-pentaacetyl-3Fax-Neu5Ac-CO2Me (3F-NeuAc), added to the media of cultured cells shuts down sialylation by a mechanism involving its intracellular conversion to CMP-3F-NeuAc, a competitive inhibitor of all sialyltransferases. Here we show that administering 3F-NeuAc to mice dramatically decreases sialylated glycans in cells of all tissues tested, including blood, spleen, liver, brain, lung, heart, kidney, and testes. A single dose results in greatly decreased sialoside expression for over 7 weeks in some tissues. Although blockade of sialylation with 3F-NeuAc does not affect viability of cultured cells, its use in vivo has a deleterious "on target" effect on liver and kidney function. After administration of 3F-NeuAc, liver enzymes in the blood are dramatically altered, and mice develop proteinuria concomitant with dramatic loss of sialic acid in the glomeruli within 4 days, leading to irreversible kidney dysfunction and failure to thrive. These results confirm a critical role for sialosides in liver and kidney function and document the feasibility of pharmacological inhibition of sialyltransferases for in vivo modulation of sialoside expression.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Ácido N-Acetilneuramínico/metabolismo , Sialiltransferasas/antagonistas & inhibidores , Sialiltransferasas/metabolismo , Animales , Western Blotting , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Glucolípidos/metabolismo , Glicoproteínas/metabolismo , Riñón/efectos de los fármacos , Riñón/metabolismo , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Masculino , Espectrometría de Masas , Ratones Endogámicos C57BL , Miocardio/metabolismo , Polisacáridos/metabolismo , Ácidos Siálicos/farmacología , Bazo/efectos de los fármacos , Bazo/metabolismo , Factores de Tiempo
7.
FASEB J ; 27(9): 3720-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23752207

RESUMEN

MicroRNA (miR)-142 is up-regulated in the brain in HIV and SIV encephalitis (SIVE). We identified the cell types where miR-142 is up-regulated and its relevant downstream target. Fluorescent in situ hybridization combined with immunofluorescent labeling revealed that miR-142-3p and -5p are expressed within hippocampal neurons and myeloid cells in SIVE. Sirtuin1 (SIRT1) was predicted as a potential miR-142 target by analysis of its 3'-UTR and bioinformatic analysis of factors linked to altered hippocampal gene expression profile in SIVE. Overexpression of pre-miR-142 in HEK293T cells led to a 3.7-fold decrease in SIRT1 protein level. Examination of the individual effects of miR-142-5p and miR-142-3p through overexpression and inhibition studies revealed that significant effects on SIRT1 occurred only with miR-142-5p. Luciferase reporter assays revealed a 2.3-fold inhibition of expression due to interaction of miR-142 with the SIRT1 3'-UTR, mutation analysis revealed that only the miR-142-5p target site was active. MiR-142 expression in primary human neurons led to a small (1.3-fold) but significant decrease in SIRT1 protein level. Furthermore, qRT-PCR revealed up-regulation of miR-142-3p (6.4-fold) and -5p (3.9-fold) and down-regulation of SIRT1 (33-fold) in macrophages/microglia from animals with SIVE. We have therefore elucidated a miR-mediated mechanism of regulation of SIRT1 expression in SIVE.


Asunto(s)
Encefalitis/genética , Encefalitis/virología , MicroARNs/genética , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Sirtuina 1/metabolismo , Animales , Western Blotting , Línea Celular , Células Cultivadas , Humanos , Sirtuina 1/genética , Regulación hacia Arriba
9.
Viruses ; 15(3)2023 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-36992383

RESUMEN

OBJECTIVE: Methamphetamine and cannabis are two widely used substances among people living with HIV (PLWH). Whereas methamphetamine use has been found to worsen HIV-associated neurocognitive impairment, the effects of combined cannabis and methamphetamine use disorder on neurocognition in PLWH are not understood. In the present study, we aimed to determine the influence of these substance use disorders on neurocognition in PLWH and to explore if methamphetamine-cannabis effects interacted with HIV status. METHOD AND PARTICIPANTS: After completing a comprehensive neurobehavioral assessment, PLWH (n = 472) were stratified by lifetime methamphetamine (M-/M+) and cannabis (C-/C+) DSM-IV abuse/dependence disorder into four groups: M-C- (n = 187), M-C+ (n = 68), M+C-, (n = 82), and M+C+ (n = 135). Group differences in global and domain neurocognitive performances and impairment were examined using multiple linear and logistic regression, respectively, while holding constant other covariates that were associated with study groups and/or cognition. Data from participants without HIV (n = 423) were added, and mixed-effect models were used to examine possible interactions between HIV and substance use disorders on neurocognition. RESULTS: Compared with M+C+, M+C- performed worse on measures of executive functions, learning, memory, and working memory and were more likely to be classified as impaired in those domains. M-C- performed better than M+C+ on measures of learning and memory but worse than M-C+ on measures of executive functions, learning, memory, and working memory. Detectable plasma HIV RNA and nadir CD4 < 200 were associated with lower overall neurocognitive performance, and these effects were greater for M+C+ compared with M-C-. CONCLUSIONS: In PLWH, lifetime methamphetamine use disorder and both current and legacy markers of HIV disease severity are associated with worse neurocognitive outcomes. There was no evidence of an HIV × M+ interaction across groups, but neurocognition was most impacted by HIV among those with polysubstance use disorder (M+C+). Better performance by C+ groups is consistent with findings from preclinical studies that cannabis use may protect against methamphetamine's deleterious effects.


Asunto(s)
Cannabis , Infecciones por VIH , Metanfetamina , Trastornos Relacionados con Sustancias , Humanos , Metanfetamina/efectos adversos , Cannabis/efectos adversos , Trastornos Relacionados con Sustancias/complicaciones , Cognición , Infecciones por VIH/complicaciones , Infecciones por VIH/psicología
10.
J Immunol ; 181(7): 4648-55, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18802067

RESUMEN

Strong evidence supports that CNS-specific CD4(+) T cells are central to the pathogenesis of multiple sclerosis and experimental autoimmune encephalomyelitis (EAE). Using a model of spontaneous EAE, we demonstrated that myelin basic protein (MBP)-specific CD4(+) T cells up-regulate activation markers in the CNS-draining cervical lymph nodes at a time when there is no T cell activation anywhere else, including the CNS, and before the appearance of clinical signs. In spontaneous EAE, the number of MBP-specific T cell numbers does not build up gradually in the CNS; instead, a swift migration of IFN-gamma-producing T cells into the CNS takes place approximately 24 h before the onset of neurological signs of EAE. Surgical excision of the cervical lymph nodes in healthy pre-EAE transgenic mice delayed the onset of EAE and resulted in a less severe disease. In EAE induced by immunization with MBP/CFA, a similar activation of T cells in the draining lymph nodes of the injection site precedes the disease. Taken together, our results suggest that peripheral activation of T cells in draining lymph nodes is an early event in the development of EAE, which paves the way for the initial burst of IFN-gamma-producing CD4(+) T cell into the CNS.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Movimiento Celular/inmunología , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Proteína Básica de Mielina/inmunología , Animales , Biomarcadores/líquido cefalorraquídeo , Linfocitos T CD4-Positivos/metabolismo , Movimiento Celular/genética , Células Cultivadas , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/cirugía , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Escisión del Ganglio Linfático , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Ganglios Linfáticos/cirugía , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Factores de Tiempo , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
11.
Curr HIV Res ; 17(2): 126-133, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31269883

RESUMEN

BACKGROUND: Methamphetamine abuse and human immunodeficiency virus (HIV) are common comorbidities. HIV-associated proteins, such as the regulatory protein TAT, may contribute to brain reward dysfunction, inducing an altered sensitivity to methamphetamine reward and/or withdrawal in this population. OBJECTIVE: These studies examined the combined effects of TAT protein expression and, chronic and binge methamphetamine regimens on brain reward function. METHODS: Transgenic mice with inducible brain expression of the TAT protein were exposed to either saline, a chronic, or a binge methamphetamine regimen. TAT expression was induced via doxycycline treatment during the last week of methamphetamine exposure. Brain reward function was assessed daily throughout the regimens, using the intracranial self-stimulation procedure, and after a subsequent acute methamphetamine challenge. RESULTS: Both methamphetamine regimens induced withdrawal-related decreases in reward function. TAT expression substantially, but not significantly increased the withdrawal associated with exposure to the binge regimen compared to the chronic regimen, but did not alter the response to acute methamphetamine challenge. TAT expression also led to persistent changes in adenosine 2B receptor expression in the caudate putamen, regardless of methamphetamine exposure. These results suggest that TAT expression may differentially affect brain reward function, dependent on the pattern of methamphetamine exposure. CONCLUSION: The subtle effects observed in these studies highlight that longer-term TAT expression, or its induction at earlier stages of methamphetamine exposure, may be more consequential at inducing behavioral and neurochemical effects.


Asunto(s)
Encéfalo/efectos de los fármacos , Metanfetamina/farmacología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Animales , Encéfalo/metabolismo , Masculino , Ratones , Ratones Transgénicos , Receptores Purinérgicos P1/genética , Recompensa , Regulación hacia Arriba/efectos de los fármacos , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo
12.
Cell Immunol ; 254(1): 56-62, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18678363

RESUMEN

In order to assess the role of osteopontin (OPN) in leukocyte accumulation in inflammatory conditions, native OPN and its thrombin cleaved form (OPN+Thr) were studied in vivo using a rodent subcutaneous air pouch model (AP). Both forms of OPN-induced macrophage infiltration into the AP in wild-type mice. In animals lacking CD44, macrophage numbers were significantly reduced within the cavity, but cells still accumulated along the subcutaneous lining. In animals lacking endogenous OPN, no differences were found in exogenous OPN-induced macrophage accumulation, although macrophage exhibited increased alpha4 integrin expression. These studies reveal that both OPN and OPN+Thr attract macrophages in vivo through CD44.


Asunto(s)
Quimiotaxis de Leucocito/inmunología , Receptores de Hialuranos/biosíntesis , Macrófagos/inmunología , Osteopontina/inmunología , Animales , Western Blotting , Adhesión Celular/inmunología , Citometría de Flujo , Inmunohistoquímica , Macrófagos/metabolismo , Ratones , Osteopontina/metabolismo
13.
Alcohol Clin Exp Res ; 32(9): 1583-92, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18616669

RESUMEN

BACKGROUND: Alcohol consumption is a common problem in HIV-infected individuals, and the effects of alcohol may alter the efficiency of the immune response, potentially aggravating the disease as well as affecting end organs, such as the brain. However, the elements of the virus-host interaction that are modulated by ethanol are poorly dissected. METHODS: Ethanol intake was conditioned in rhesus macaques prior to SIV infection, in order to mimic this common human behavior, and allow the evaluation of aspects of the virus-immune system interactions during acute time-points, when important facets of the infection are set up and when virus reproducibly enters the brain. RESULTS: Although ethanol had a limited effect on the acute plasma viral load, it resulted in reduced circulating memory CD4(+) T cells and increased levels of monocytes expressing the viral coreceptor CCR5. In organs, ethanol consumption impacted immune cells in the liver as well as lymphoid and other nonlymphoid tissues, where CD4(+) T cells were predominantly affected. CONCLUSION: Overall, the consumption of alcohol causes immune cell alterations that can contribute to the generation of a disease susceptible environment upon SIV infection.


Asunto(s)
Consumo de Bebidas Alcohólicas/inmunología , Alcoholismo/inmunología , Sistema Inmunológico/fisiopatología , Enfermedades de los Monos/inmunología , Enfermedades de los Monos/virología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Encéfalo/patología , Encéfalo/virología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/inmunología , Células Asesinas Naturales/patología , Hígado/patología , Hígado/virología , Macaca mulatta , Enfermedades de los Monos/epidemiología , Factores de Riesgo , Síndrome de Inmunodeficiencia Adquirida del Simio/epidemiología , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Bazo/patología , Bazo/virología , Linfocitos T/patología , Carga Viral
14.
BMC Res Notes ; 11(1): 275, 2018 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-29728138

RESUMEN

OBJECTIVE: In HIV+ individuals, the virus enters the central nervous system and invades innate immune cells, producing important changes that result in neurological deficits. We aimed to determine whether HIV plays a direct role in neuronal excitability. Of the HIV peptides, Tat is secreted and acts in other cells. In order to examine whether the HIV Tat can modify neuronal excitability, we exposed primary murine hippocampal neurons to that peptide, and tested its effects on the intrinsic membrane properties, 4 and 24 h after exposure. RESULTS: The exposure of hippocampal pyramidal neurons to Tat for 4 h did not alter intrinsic membrane properties. However, we found a strong increase in intrinsic excitability, characterized by increase of the slope (Gain) of the input-output function, in cells treated with Tat for 24 h. Nevertheless, Tat treatment for 24 h did not alter the resting membrane potential, input resistance, rheobase and action potential threshold. Thus, neuronal adaptability to Tat exposure for 24 h is not applicable to basic neuronal properties. A restricted but significant effect on coupling the inputs to the outputs may have implications to our knowledge of Tat biophysical firing capability, and its involvement in neuronal hyperexcitability in neuroHIV.


Asunto(s)
VIH-1/metabolismo , Hipocampo , Potenciales de la Membrana , Células Piramidales , Transmisión Sináptica , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/farmacología , Animales , Hipocampo/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Células Piramidales/efectos de los fármacos , Proteínas Recombinantes , Transmisión Sináptica/efectos de los fármacos
15.
J Neurosci ; 26(17): 4577-85, 2006 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-16641237

RESUMEN

CNS abnormalities can be detected during chronic human immunodeficiency virus (HIV) infection, before the development of opportunistic infections or other sequelae of immunodeficiency. However, although end-stage dementia caused by HIV has been linked to the presence of infected and activated macrophages and microglia in the brain, the nature of the changes resulting in the motor and cognitive disorders in the chronic stage is unknown. Using simian immunodeficiency virus-infected rhesus monkeys, we sought the molecular basis for CNS dysfunction. In the chronic stable stage, nearly 2 years after infection, all animals had verified CNS functional abnormalities. Both virus and infiltrating lymphocytes (CD8+ T-cells) were found in the brain. Molecular analysis revealed that the expression of several immune response genes was increased, including CCL5, which has pleiotropic effects on neurons as well as immune cells. CCL5 was significantly upregulated throughout the course of infection, and in the chronic phase was present in the infiltrating lymphocytes. We have identified an altered state of the CNS at an important stage of the viral-host interaction, likely arising to protect against the virus but in the long term leading to damaging processes.


Asunto(s)
Quimiocinas/inmunología , Encefalitis Viral/diagnóstico , Encefalitis Viral/inmunología , Huésped Inmunocomprometido/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/diagnóstico , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/aislamiento & purificación , Animales , Trastornos del Conocimiento/diagnóstico , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/inmunología , Encefalitis Viral/etiología , Macaca mulatta , Síndrome de Inmunodeficiencia Adquirida del Simio/complicaciones , Síndrome de Inmunodeficiencia Adquirida del Simio/virología
16.
Viral Immunol ; 19(4): 679-89, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17201663

RESUMEN

Facets of the immune response early after human immunodeficiency virus (HIV) infection influence the course of disease. In the simian immunodeficiency virus (SIV)-rhesus monkey system, a global dysfunction of CD4(+) T cell cytokine secretion was reported to develop early after infection [McKay PF, Barouch DH, Schmitz JE, Veazey RS, Gorgone DA, Lifton MA, Williams KC, and Letvin NL: J Virol 2003;77:4695-4702]. Because differences have been found in SIV pathogenesis depending on the origin of the monkeys, we investigated the correlation between animal background, defined by country of origin (India or China), and circulating T cell cytokine secretion as well as cycling ability within the first 3 mo of SIV infection. An early loss of CD4(+) T cells that produce interferon (IFN)-gamma and interleukin (IL)-2, those that produce IFN-gamma but not tumor necrosis factor (TNF)-alpha, as well as those that do not express IFN-gamma but can express IL-2 or TNF-alpha, was observed in animals of Indian, but not of Chinese, origin after SIV infection. After infection CD4(+) T cells in Chinese macaques developed an increased proliferating pool of T cells compared with Indian animals. These data reveal host diversity in the global effects of SIV infection on functional subsets of immune cells, which can add to a better understanding of differences observed in populations from diverse ethnic origins.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Citocinas/biosíntesis , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , China , India , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Activación de Linfocitos , Macaca mulatta , Especificidad de la Especie , Factor de Necrosis Tumoral alfa/biosíntesis
17.
AIDS ; 19(15): 1704-6, 2005 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-16184046

RESUMEN

The use of China-derived monkeys in AIDS research has been limited by reports of reduced susceptibility to SIV. We performed a serial passage of SIV in Chinese macaques, which resulted in a viral stock capable of inducing simian AIDS and high levels of replication in these animals. Similar to HIV in humans, SIV pathogenesis in non-human primates is not limited by geographical origin. Chinese macaques are useful in pathogenesis, vaccine, and therapeutic studies in AIDS.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Animales , Relación CD4-CD8 , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Macaca mulatta , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/fisiología , Carga Viral , Virulencia , Replicación Viral
18.
AIDS ; 17(11): 1605-11, 2003 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-12853742

RESUMEN

OBJECTIVE: Infection with simian immunodeficiency virus (SIV), like HIV, can lead to central nervous system (CNS) abnormalities. One of the alterations observed in the brain is the accumulation of highly activated CD8 lymphocytes that, while fighting the infection, may cause tissue damage. In order to determine whether these CD8 cells in the brain comprise a distinct clonal population the expression of T-cell receptor (TCR) genes of two SIV-infected monkeys with CNS abnormalities were analyzed, comparing brain to periphery. METHODS: RNA from magnetically sorted CD8+ cells obtained from the brain, blood, lymph nodes, and spleen was analyzed for the distribution of 24 Vbeta family genes by reverse transcriptase-polymerase chain reaction followed by Southern blot. The CDR3 region of the most enriched family in each brain was sequenced in all the sites for comparison. RESULTS: The pattern of Vbeta distribution in the brain and the periphery was polyclonal, but an increase in certain Vbeta families was found in the brain, suggesting that regional mechanisms participate in the determination of the local clonal specificities. The sequence of the CDR3 domain of predominant Vbeta families in the brain revealed that approximately one-third of the CD8 cells were not identified in the periphery. CONCLUSION: CD8 cells in the brain exhibit a distinct clonal repertoire. This distinction may have implications for regional immunity, regulation, or selection of site-specific viral mutants.


Asunto(s)
Encéfalo/inmunología , Linfocitos T CD8-positivos/fisiología , Reordenamiento Génico de la Cadena beta de los Receptores de Antígenos de los Linfocitos T , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios , Animales , Linfocitos T CD8-positivos/inmunología , Células Clonales , Macaca mulatta , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Temperature (Austin) ; 1(3): 227-241, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-26346736

RESUMEN

Hyperthermia is a potentially lethal side effect of Methamphetamine (Meth) abuse, which involves the participation of peripheral thermogenic sites such as the Brown Adipose Tissue (BAT). In a previous study we found that the anti-oxidant N-acetyl cysteine (NAC) can prevent the high increase in temperature in a mouse model of Meth-hyperthermia. Here, we have further explored the ability of NAC to modulate Meth-induced hyperthermia in correlation with changes in BAT. We found that NAC treatment in controls causes hypothermia, and, when administered prior or upon the onset of Meth-induced hyperthermia, can ameliorate the temperature increase and preserve mitochondrial numbers and integrity, without affecting locomotor activity. This was different from Dantrolene, which decreased motor activity without affecting temperature. The effects of NAC were seen in spite of its inability to recover the decrease of mitochondrial superoxide induced in BAT by Meth. In addition, NAC did not prevent the Meth-induced decrease of BAT glutathione. Treatment with S-adenosyl-L-methionine, which improves glutathione activity, had an effect in ameliorating Meth-induced hyperthermia, but also modulated motor activity. This suggests a role for the remaining glutathione for controlling temperature. However, the mechanism by which NAC operates is independent of glutathione levels in BAT and specific to temperature. Our results show that, in spite of the absence of a clear mechanism of action, NAC is a pharmacological tool to examine the dissociation between Meth-induced hyperthermia and motor activity, and a drug of potential utility in treating the hyperthermia associated with Meth-abuse.

20.
Science ; 334(6057): 809-13, 2011 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-22021672

RESUMEN

Phospholipase A(2)(PLA(2)) enzymes are considered the primary source of arachidonic acid for cyclooxygenase (COX)-mediated biosynthesis of prostaglandins. Here, we show that a distinct pathway exists in brain, where monoacylglycerol lipase (MAGL) hydrolyzes the endocannabinoid 2-arachidonoylglycerol to generate a major arachidonate precursor pool for neuroinflammatory prostaglandins. MAGL-disrupted animals show neuroprotection in a parkinsonian mouse model. These animals are spared the hemorrhaging caused by COX inhibitors in the gut, where prostaglandins are instead regulated by cytosolic PLA(2). These findings identify MAGL as a distinct metabolic node that couples endocannabinoid to prostaglandin signaling networks in the nervous system and suggest that inhibition of this enzyme may be a new and potentially safer way to suppress the proinflammatory cascades that underlie neurodegenerative disorders.


Asunto(s)
Ácidos Araquidónicos/metabolismo , Encéfalo/metabolismo , Moduladores de Receptores de Cannabinoides/metabolismo , Endocannabinoides , Glicéridos/metabolismo , Inflamación/metabolismo , Monoacilglicerol Lipasas/metabolismo , Prostaglandinas/metabolismo , Animales , Ácido Araquidónico/metabolismo , Benzodioxoles/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Ciclooxigenasa 1/metabolismo , Citocinas/metabolismo , Eicosanoides/metabolismo , Inhibidores Enzimáticos/farmacología , Hidrólisis , Inflamación/patología , Mediadores de Inflamación/farmacología , Lipopolisacáridos/farmacología , Hígado/metabolismo , Pulmón/metabolismo , Metabolómica , Ratones , Ratones Endogámicos C57BL , Monoacilglicerol Lipasas/antagonistas & inhibidores , Monoacilglicerol Lipasas/genética , Fármacos Neuroprotectores/farmacología , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/patología , Fosfolipasas A2/genética , Fosfolipasas A2/metabolismo , Piperidinas/farmacología , Prostaglandinas/biosíntesis , Transducción de Señal
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda