Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Brain Res ; 1565: 74-81, 2014 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-24746498

RESUMO

Bone marrow derived mononuclear cell (MNC) transplantation is a potential therapy for ischemic stroke. Here, we hypothesized that valproic acid (VPA) would modulate transplantation effects of MNCs in a rat ischemic stroke model. Male Sprague-Dawley rats were subjected to transient 90min middle cerebral artery occlusion. Infarct volume, neurological outcome, and immunohistological assessments were performed 7 days after ischemia. MNCs injected 6 or 24h but not 48 or 72h after ischemia significantly reduced infarct volume and improved neurological deficits. We then tested whether the therapeutic window of MNC transplantation could be expanded through combination therapy with VPA. MNC transplantation at 48h combined with VPA injection three times at 47, 53, and 72h after ischemia significantly ameliorated infarct volume and neurological deficits compared to a vehicle group. Combination therapy reduced the number of myeloperoxidase-positive cells, ionized calcium binding adapter molecule 1-positive cells, tumor necrosis factor-α-positive cells, and von Willebrand factor-positive cells in the ischemic boundary zone. The number of engrafted MNCs that were fluorescently labeled with PKH 26, on day 7, was significantly higher after combination therapy than after that MNC transplantation alone. Our results demonstrated that combination therapy with VPA enhanced the anti-inflammatory and vasculo-protective effects against endothelial damage following ischemia, and increased the survival of transplanted cells, leading to expansion of the therapeutic time window for MNC transplantation. Together, these findings suggest that VPA may be an appropriate partner for cell-based treatment of ischemic stroke.


Assuntos
Transplante de Medula Óssea , Isquemia Encefálica/terapia , Acidente Vascular Cerebral/terapia , Ácido Valproico/uso terapêutico , Animais , Células da Medula Óssea/efeitos dos fármacos , Isquemia Encefálica/tratamento farmacológico , Terapia Combinada , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média , Masculino , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/tratamento farmacológico
2.
Gene Ther ; 16(3): 383-91, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18818668

RESUMO

Interleukin-10 (IL-10) ameliorates various T-helper type 1 cell-mediated chronic inflammatory diseases. Although the therapeutic benefits of IL-10 include antiatherosclerotic effects, pathophysiological effects of IL-10 on vascular remodeling in hypertension have not yet been elucidated. These studies were designed to determine whether sustained IL-10 expression, mediated by an adeno-associated virus (AAV) vector, prevents vascular remodeling and target-organ damage in the stroke-prone spontaneously hypertensive rat (SHR-SP)-an animal model of malignant hypertension. A single intramuscular injection of an AAV1 vector encoding rat IL-10 introduced long-term IL-10 expression. These IL-10-transduced rats had decreased stroke episodes and proteinuria, resulting in improved survival. Histological examination revealed a reduced level of deleterious vascular remodeling of resistance vessels in the brain and kidney of these rats. Immunohistochemical analysis indicated that IL-10 inhibited the enhanced renal transforming growth factor-beta expression and perivascular infiltration of monocytes/macrophages and nuclear factor-kappaB-positive cells normally observed in the SHR-SP. Four weeks after IL-10 vector injection, systolic blood pressure significantly decreased and this effect persisted for several months. Overall, AAV vector-mediated systemic IL-10 expression prevented vascular remodeling and inflammatory lesions of target organs in the SHR-SP. This approach provides significant insights into the prevention strategy of disease onset with unknown genetic predisposition or intractable polygenic disorders.


Assuntos
Terapia Genética/métodos , Hipertensão/complicações , Interleucina-10/biossíntese , Acidente Vascular Cerebral/prevenção & controle , Animais , Pressão Sanguínea/fisiologia , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Artérias Carótidas/patologia , Dependovirus/genética , Vetores Genéticos , Hipertensão/metabolismo , Hipertensão/patologia , Interleucina-10/genética , Nefropatias/etiologia , Nefropatias/patologia , Nefropatias/prevenção & controle , Masculino , Ratos , Ratos Endogâmicos SHR , Acidente Vascular Cerebral/etiologia , Acidente Vascular Cerebral/patologia , Análise de Sobrevida , Transdução Genética
5.
Acta Neurochir Suppl ; 86: 113-6, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14753417

RESUMO

To explore effects of Immunosuppressant FK506 on signal transduction pathway. we studied changes in subcellular distribution of protein kinase Cgamma (PKCgamma), CaM kinase II (CaMKII), as well as changes of tyrosine phosphorylation levels after ischemia. Male Mongolian gerbils were divided into 3 groups; FK506 (1 mg/kg, 3 mg/kg) and vehicle. FK506 was administered intravenously after 5 min ischemia. At the designated time points (0 time, 5 min ischemia, 1 hour, or 24 hour recovery), heads were frozen and samples were taken from CAI subfield of hippocampus. Western blot analysis was carried out with specific antibodies for PKCgamma, CaMKII, and phosphotyrosine. FK506 administration significantly decreased translocation of PKCgamma and CaMKII at 24 h of recovery (p < 0.05, ANOVA followed by Student-Newman Keuls' test) in P2 fraction. The levels of tyrosine phosphorylated p160, p140, p100, p90, and p80 in P2 fraction were also significantly decreased with FK506 treatment at 24 h of recovery. The persistently elevated PKCgamma and CaMKII level in P2 fraction which may be related to cell death, are attenuated with FK506 treatment. FK506 may contribute to recover calcium homeostasis in the post ischemic phase and promote cell survival.


Assuntos
Isquemia Encefálica/enzimologia , Hipocampo/metabolismo , Imunossupressores/farmacologia , Proteínas Quinases/metabolismo , Tacrolimo/farmacologia , Tirosina/metabolismo , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Gerbillinae , Hipocampo/enzimologia , Masculino , Fosforilação/efeitos dos fármacos , Fosfotirosina/metabolismo , Proteína Quinase C/metabolismo , Distribuição Tecidual
6.
Neurol Res ; 23(7): 751-4, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11680516

RESUMO

Brief ischemic episode, which in itself is not lethal, confers tolerance to subsequent ischemic insults. Since intracellular signal transduction system has been implicated in ischemic cell death, we studied the effect of pre-conditioning on the changes in the subcellular distribution of protein kinase Cgamma (PKCgamma) as well as CaM kinase II (CaMKII). Gerbils were pre-conditioned by a sublethal 2 min cerebral ischemia 24 h prior to lethal 5 min ischemia. The pre-conditioning generally downregulated PKCgamma and CaMKII in the CA1 hippocampus. Especially at the starting point of the second lethal ischemia, the cytosolic PKCgamma level was about 40% lower in the pre-conditioned group. Also, the crude synaptosomal CaMKII level at 24 h reperfusion following the second ischemia was significantly lower in the pre-conditioned group, showing enhanced recovery of CaMKII translocation. Present results suggest that ischemic pre-conditioning may downregulate calcium-mediated cell signaling system, enhancing normalization of calcium homeostasis, perturbed by the second ischemia of lethal duration.


Assuntos
Isquemia Encefálica/enzimologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Compartimento Celular/fisiologia , Hipocampo/enzimologia , Precondicionamento Isquêmico , Neurônios/enzimologia , Proteína Quinase C/metabolismo , Animais , Isquemia Encefálica/patologia , Isquemia Encefálica/fisiopatologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Morte Celular/fisiologia , Membrana Celular/metabolismo , Citosol/metabolismo , Regulação para Baixo/fisiologia , Gerbillinae , Hipocampo/patologia , Hipocampo/fisiopatologia , Immunoblotting , Masculino , Neurônios/patologia , Transporte Proteico/fisiologia , Transdução de Sinais/fisiologia , Sinaptossomos/metabolismo
7.
Neurol Res ; 23(7): 755-60, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11680517

RESUMO

Tacrolimus (FK506), an immunosuppressant currently used in clinic, is known to have neuroprotective properties. However, effects in focal ischemia are shown only in a endothelin induced middle cerebral artery (MCA) occlusion model or with filament technique at a relatively high dose. We have previously shown that FK506 had significant protective effects at a low dose of 0.3 mg kg(-1) when administered immediately after ischemia. In this study, we explored the therapeutic time window of FK506 at this low dose, in a transient focal ischemia model using filament technique. Male Sprague-Dawley rats were subjected to 2 h MCA occlusion and subsequent reperfusion. They received FK506 or vehicle (0.3 mg kg(-1)) i.v. at 30, 60 or 120 min after induction of ischemia, and were decapitated 24 h after ischemia. FK506 injected at 30 and 60 min significantly reduced cortical infarction volume (FK506 vs. vehicle; 30 min: 95 +/- 33 mm3 vs. 170 +/- 62 mm3, p < 0.05; 60 min: 93 +/- 45 mm3, vs. 168 +/- 35 mm3, p < 0.05, respectively). FK506 was ineffective when given at 120 min after ischemia. FK506 had no effect on edema formation, nor on the infarct volume in striatum. The therapeutic time window for this low dose of FK506 given i.v. is between 60 and 120 min in this model.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Imunossupressores/farmacologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Traumatismo por Reperfusão/tratamento farmacológico , Tacrolimo/farmacologia , Animais , Glicemia/efeitos dos fármacos , Glicemia/fisiologia , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Edema Encefálico/tratamento farmacológico , Edema Encefálico/metabolismo , Edema Encefálico/fisiopatologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatologia , Fenômenos Fisiológicos Cardiovasculares/efeitos dos fármacos , Infarto Cerebral/tratamento farmacológico , Infarto Cerebral/metabolismo , Infarto Cerebral/fisiopatologia , Esquema de Medicação , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/fisiopatologia , Masculino , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/fisiopatologia , Resultado do Tratamento
8.
J Neurol Sci ; 137(1): 7-13, 1996 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9120490

RESUMO

The primary objective of this study was to attempt to induce excessive intraglial acidosis during ischemia by subjecting rats to an initial insult which leads to post insult accumulation of glycogen, presumed to accumulate primarily in astrocytes. The initial insults were 15 min of transient forebrain ischemia, 30 min of hypoglycemic coma, and intraperitonial injection of methionine-sulphoximine (MSO). In the first two of these insults, glycogen content in neocortex increased to 6-7 mM kg(-1) after 6 h of recovery, and in MSO-treated animals even higher values were measured 24 h after administration ( 12 mM kg(-1)). In spite of this glycogen loading, the amount of lactate formed during a subsequent ischemic insult (of 5-30 min duration) did not exceed values usually obtained during complete ischemia in animals with normal glycogen contents (tissue lactate contents of 15 mM kg(-1)) This was because appreciable amounts of glycogen (3-7 mM kg(-1)) remained undegraded even after 30 min of ischemia. The undigested part largely reflected the extra amount of glycogen accumulated after the initial insults. It is discussed whether this part is unavailable to degradation by phosphorylase.


Assuntos
Isquemia Encefálica/metabolismo , Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Glicogênio/metabolismo , Traumatismo por Reperfusão/metabolismo , Acidose/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Encéfalo/citologia , Coma/etiologia , Coma/metabolismo , Metabolismo Energético/fisiologia , Hipoglicemia/complicações , Hipoglicemia/metabolismo , Ataque Isquêmico Transitório/metabolismo , Masculino , Metionina Sulfoximina , Fosforilação , Ratos , Ratos Wistar , Convulsões/induzido quimicamente , Convulsões/metabolismo
10.
Acta Neurochir Suppl ; 66: 8-14, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8780790

RESUMO

The present article is concerned with mechanisms which are responsible for the exaggerated brain damage observed in hyperglycemic animals subjected to transient global or forebrain ischemia. Since hyperglycemia enchances the production of lactate plus H+ during ischemia, it seems likely that aggravation of damage is due to exaggerated intra- and extracellular acidosis. This contention is supported by results showing a detrimental effect of extreme hypercapnia in normoglycemic rats subjected to transient ischemia or to hypoglycemic coma. Enhanced acidosis may exaggerate ischemic damage by one of three mechanisms: (i) accelerating free radical production via H(+)-dependent reactions, some of which are catalyzed by iron released from protein bindings by a lowering of pH, (ii) by perturbing the intracellular signal transduction pathway, leading to changes in gene expression or protein synthesis, or (iii) by activating endonucleases which cause DNA fragmentation. While activation of endonucleases must affect the nucleus, the targets of free radical attack are not known. Microvessels are considered likely targets of such attack in sustained ischemia and in trauma; however, enhanced acidosis is not known to aggravate microvascular dysfunction, or to induce inflammatory responses at the endothelial-blood interface. A more likely target is the mitochondrion. Thus, if the ischemia is of long duration (30 min) hyperglycemia triggers rapidly developing mitochondrial failure. It is speculated that this is because free radicals damage components of the respiratory chain, leading to a secondary deterioration of oxidative phosphorylation.


Assuntos
Equilíbrio Ácido-Base/genética , Dano Encefálico Crônico/genética , Isquemia Encefálica/genética , Cetoacidose Diabética/genética , Prosencéfalo/irrigação sanguínea , Animais , Barreira Hematoencefálica/genética , Fragmentação do DNA/genética , Transporte de Elétrons/genética , Radicais Livres , Mitocôndrias/genética , Ratos , Transdução de Sinais/genética
11.
Neurobiol Dis ; 2(2): 97-108, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8980013

RESUMO

The objective of the present experiments was to characterize conditions under which pre-ischaemic hyperglycaemia aggravates brain damage following transient forebrain ischaemia. Specifically, we wished to explore whether accentuated damage is a threshold function of plasma glucose concentration or pH, as assessed by measurements of extracellular pH (pHe). Forebrain ischaemia of 10 min duration was induced in rats at varying degrees of hyperglycaemia, with continuous measurements of pHe, and the animals were allowed to survive for 7 days before histopathological evaluation of the density and distribution of brain damage. Ischaemic brain damage appeared as a threshold function of plasma glucose concentration. At values of 4-6 mM virtually no damage was observed in any other structure than the CA1 sector of the hippocampus and, even in that structure, damage was variable. At glucose concentrations of 8-12 mM moderate damage was observed infrequently in caudoputamen, parietal cortex, and thalamus. At values above 12 mM, damage increased dramatically in these areas, and additional structures were recruited in the damage process (cingulate cortex, the CA3 sector of the hippocampus, and substantia nigra). Measurements of pHe in parietal cortex showed a threshold for seizure induction at values of 6.4-6.5, probably corresponding to intracellular pH values of 6.2-6.3. The threshold for aggravation of histopathological damage was similar. It is concluded that a moderate increase in plasma glucose in the threshold range predisposes the tissue to aggravated damage, probably by activating biochemical reactions or pathophysiological events with a steep pH dependence.


Assuntos
Glicemia/metabolismo , Hiperglicemia/sangue , Ataque Isquêmico Transitório/sangue , Prosencéfalo/irrigação sanguínea , Animais , Concentração de Íons de Hidrogênio , Hiperglicemia/patologia , Incidência , Ataque Isquêmico Transitório/patologia , Masculino , Necrose , Neurônios/patologia , Ratos , Ratos Wistar , Convulsões/sangue , Convulsões/epidemiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...