Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Banco de datos
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Stroke ; 43(6): 1647-53, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22426312

RESUMEN

BACKGROUND AND PURPOSE: Methamphetamine is a powerful addictive, which has been associated with ischemic stroke and brain hemorrhage in humans. Whether and how methamphetamine influences the expression of tight junctions and adenosine triphosphate-binding cassette transporters, which have previously been shown to be regulated by apolipoprotein-E (ApoE) under conditions of brain ischemia, was unknown. METHODS: C57BL/6J mice received intraperitoneal injections of methamphetamine (3 times 4 mg/kg separated by 3 hours) either alone or in combination with the ApoE receptor-2 inhibitor receptor-associated protein (40 µg/kg) or the inducible nitric oxide synthase inhibitor 1400W (5 mg/kg). Animals were euthanized 3 or 24 hours after methamphetamine exposure. Tissue responses were evaluated with Western blots, immunoprecipitation, and immunohistochemistry using total brain and cerebral microvessel extracts. RESULTS: Methamphetamine induced a transient activation of stress kinases c-Jun N-terminal kinase 1/2 and p38 in the brain parenchyma and increased intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 expression on cerebral microvessels without inducing loss of tight junction proteins and without inducing IgG extravasation. Methamphetamine transiently increased the expression of the luminal adenosine triphosphate-binding cassette transporter ABCB1 on cerebral microvessels and reduced the expression of the abluminal transporter ABCC1. Elevated expression of ApoE was noted in the brain parenchyma by methamphetamine, activating ApoE receptor-2 on brain capillaries, deactivating c-Jun N-terminal kinase 1/2 and c-Jun, and regulating ABCB1 and ABCC1 expression. Indeed, ApoE receptor-2 and inducible nitric oxide synthase inhibition prevented the ABCB1 and ABCC1 expression changes. CONCLUSIONS: Acute exposure to methamphetamine at doses comparable to those consumed in drug addiction does not induce tight junction breakdown but differentially regulates adenosine triphosphate-binding cassette transporters through the ApoE/ApoE receptor-2/c-Jun N-terminal kinase 1/2 pathway.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Apolipoproteínas E/metabolismo , Isquemia Encefálica/metabolismo , Estimulantes del Sistema Nervioso Central/efectos adversos , Cerebelo/metabolismo , Circulación Cerebrovascular/efectos de los fármacos , Metanfetamina/efectos adversos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Animales , Isquemia Encefálica/fisiopatología , Capilares/metabolismo , Capilares/fisiopatología , Estimulantes del Sistema Nervioso Central/farmacología , Cerebelo/irrigación sanguínea , Cerebelo/fisiopatología , Humanos , Iminas/farmacología , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Metanfetamina/farmacología , Ratones , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Trastornos Relacionados con Sustancias/metabolismo , Trastornos Relacionados con Sustancias/patología , Trastornos Relacionados con Sustancias/fisiopatología , Uniones Estrechas/metabolismo , Uniones Estrechas/patología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
2.
J Neuroinflammation ; 8: 165, 2011 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-22114930

RESUMEN

BACKGROUND: Preconditioning is a phenomenon by which tolerance develops to injury by previous exposure to a stressor of mild severity. Previous studies have shown that single or repeated low dose MDMA can attenuate 5-HT transporter loss produced by a subsequent neurotoxic dose of the drug. We have explored the mechanism of delayed preconditioning by low dose MDMA. METHODS: Male Dark Agouti rats were given low dose MDMA (3 mg/kg, i.p.) 96 h before receiving neurotoxic MDMA (12.5 mg/kg, i.p.). IL-1ß and IL1ra levels and 5-HT transporter density in frontal cortex were quantified at 1 h, 3 h or 7 days. IL-1ß, IL-1ra and IL-1RI were determined between 3 h and 96 h after low dose MDMA. sIL-1RI combined with low dose MDMA or IL-1ß were given 96 h before neurotoxic MDMA and toxicity assessed 7 days later. RESULTS: Pretreatment with low dose MDMA attenuated both the 5-HT transporter loss and elevated IL-1ß levels induced by neurotoxic MDMA while producing an increase in IL-1ra levels. Low dose MDMA produced an increase in IL-1ß at 3 h and in IL-1ra at 96 h. sIL-1RI expression was also increased after low dose MDMA. Coadministration of sIL-1RI (3 µg, i.c.v.) prevented the protection against neurotoxic MDMA provided by low dose MDMA. Furthermore, IL-1ß (2.5 pg, intracortical) given 96 h before neurotoxic MDMA protected against the 5-HT neurotoxicity produced by the drug, thus mimicking preconditioning. CONCLUSIONS: These results suggest that IL-1ß plays an important role in the development of delayed preconditioning by low dose MDMA.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Tolerancia a Medicamentos/fisiología , Interleucina-1beta/metabolismo , N-Metil-3,4-metilenodioxianfetamina/toxicidad , Serotonina/metabolismo , Animales , Western Blotting , Temperatura Corporal/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Masculino , N-Metil-3,4-metilenodioxianfetamina/administración & dosificación , Ratas , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo
3.
J Neuroinflammation ; 8: 53, 2011 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-21595923

RESUMEN

BACKGROUND: 3,4-Methylenedioxymethamphetamine (MDMA) produces a neuroinflammatory reaction in rat brain characterized by an increase in interleukin-1 beta (IL-1ß) and microglial activation. The CB2 receptor agonist JWH-015 reduces both these changes and partially protects against MDMA-induced neurotoxicity. We have examined MDMA-induced changes in IL-1 receptor antagonist (IL-1ra) levels and IL-1 receptor type I (IL-1RI) expression and the effects of JWH-015. The cellular location of IL-1ß and IL-1RI was also examined. MDMA-treated animals were given the soluble form of IL-1RI (sIL-1RI) and neurotoxic effects examined. METHODS: Dark Agouti rats received MDMA (12.5 mg/kg, i.p.) and levels of IL-1ra and expression of IL-1RI measured 1 h, 3 h or 6 h later. JWH-015 (2.4 mg/kg, i.p.) was injected 48 h, 24 h and 0.5 h before MDMA and IL-1ra and IL-1RI measured. For localization studies, animals were sacrificed 1 h or 3 h following MDMA and stained for IL-1ß or IL-1RI in combination with neuronal and microglial markers. sIL-1RI (3 µg/animal; i.c.v.) was administered 5 min before MDMA and 3 h later. 5-HT transporter density was determined 7 days after MDMA injection. RESULTS: MDMA produced an increase in IL-ra levels and a decrease in IL-1RI expression in hypothalamus which was prevented by CB2 receptor activation. IL-1RI expression was localized on neuronal cell bodies while IL-1ß expression was observed in microglial cells following MDMA. sIL-1RI potentiated MDMA-induced neurotoxicity. MDMA also increased IgG immunostaining indicating that blood brain-barrier permeability was compromised. CONCLUSIONS: In summary, MDMA produces changes in IL-1 signal modulators which are modified by CB2 receptor activation. These results indicate that IL-1ß may play a partial role in MDMA-induced neurotoxicity.


Asunto(s)
Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Interleucina-1/metabolismo , N-Metil-3,4-metilenodioxianfetamina/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Receptor Cannabinoide CB2/metabolismo , Transducción de Señal/fisiología , Animales , Temperatura Corporal/efectos de los fármacos , Encéfalo/anatomía & histología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Alucinógenos/farmacología , Masculino , Neuronas/citología , Ratas , Receptores de Interleucina-1/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo
4.
Front Mol Neurosci ; 10: 278, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28928633

RESUMEN

Lysophosphatidic acid (LPA) is an extracellular lipid mediator that regulates nervous system development and functions acting through G protein-coupled receptors (GPCRs). Here we explore the crosstalk between LPA1 receptor and glutamatergic transmission by examining expression of glutaminase (GA) isoforms in different brain areas isolated from wild-type (WT) and KOLPA1 mice. Silencing of LPA1 receptor induced a severe down-regulation of Gls-encoded long glutaminase protein variant (KGA) (glutaminase gene encoding the kidney-type isoforms, GLS) protein expression in several brain regions, particularly in brain cortex and hippocampus. Immunohistochemical assessment of protein levels for the second type of glutaminase (GA) isoform, glutaminase gene encoding the liver-type isoforms (GLS2), did not detect substantial differences with regard to WT animals. The regional mRNA levels of GLS were determined by real time RT-PCR and did not show significant variations, except for prefrontal and motor cortex values which clearly diminished in KO mice. Total GA activity was also significantly reduced in prefrontal and motor cortex, but remained essentially unchanged in the hippocampus and rest of brain regions examined, suggesting activation of genetic compensatory mechanisms and/or post-translational modifications to compensate for KGA protein deficit. Remarkably, Golgi staining of hippocampal regions showed an altered morphology of glutamatergic pyramidal cells dendritic spines towards a less mature filopodia-like phenotype, as compared with WT littermates. This structural change correlated with a strong decrease of active matrix-metalloproteinase (MMP) 9 in cerebral cortex and hippocampus of KOLPA1 mice. Taken together, these results demonstrate that LPA signaling through LPA1 influence expression of the main isoenzyme of glutamate biosynthesis with strong repercussions on dendritic spines maturation, which may partially explain the cognitive and learning defects previously reported for this colony of KOLPA1 mice.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA