Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 143
Filtrar
1.
Mol Cell Biol ; 21(20): 6841-50, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11564868

RESUMO

The growth arrest and DNA damage-inducible protein, GADD34, was identified by its interaction with human inhibitor 1 (I-1), a protein kinase A (PKA)-activated inhibitor of type 1 protein serine/threonine phosphatase (PP1), in a yeast two-hybrid screen of a human brain cDNA library. Recombinant GADD34 (amino acids 233 to 674) bound both PKA-phosphorylated and unphosphorylated I-1(1-171). Serial truncations mapped the C terminus of I-1 (amino acids 142 to 171) as essential for GADD34 binding. In contrast, PKA phosphorylation was required for PP1 binding and inhibition by the N-terminal I-1(1-80) fragment. Pulldowns of GADD34 proteins expressed in HEK293T cells showed that I-1 bound the central domain of GADD34 (amino acids 180 to 483). By comparison, affinity isolation of cellular GADD34/PP1 complexes showed that PP1 bound near the C terminus of GADD34 (amino acids 483 to 619), a region that shows sequence homology with the virulence factors ICP34.5 of herpes simplex virus and NL-S of avian sarcoma virus. While GADD34 inhibited PP1-catalyzed dephosphorylation of phosphorylase a, the GADD34-bound PP1 was an active eIF-2alpha phosphatase. In brain extracts from active ground squirrels, GADD34 bound both I-1 and PP1 and eIF-2alpha was largely dephosphorylated. In contrast, the I-1/GADD34 and PP1/GADD34 interactions were disrupted in brain from hibernating animals, in which eIF-2alpha was highly phosphorylated at serine-51 and protein synthesis was inhibited. These studies suggested that modification of the I-1/GADD34/PP1 signaling complex regulates the initiation of protein translation in mammalian tissues.


Assuntos
Proteínas de Transporte , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Endorribonucleases , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Plantas/metabolismo , Proteínas/química , Proteínas/fisiologia , Proteínas de Ligação a RNA/metabolismo , Animais , Antígenos de Diferenciação , Apoptose , Encéfalo/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Biblioteca Gênica , Humanos , Cinética , Modelos Genéticos , Fosfoproteínas Fosfatases , Fosforilação , Testes de Precipitina , Ligação Proteica , Proteína Fosfatase 1 , Proteínas Recombinantes/metabolismo , Sciuridae , Transdução de Sinais , Técnicas do Sistema de Duplo-Híbrido
2.
Cytokine Growth Factor Rev ; 10(2): 119-30, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10743503

RESUMO

Brain injury (ischemia, trauma) is among the leading cause of mortality and disability in the western world. It induces increased production of tumor necrosis factor (TNF alpha) by brain resident cells. There is conflicting evidence on the role of this response in the injured brain, showing its potential effect in both processes of repair and of damage. This review presents data from clinical and experimental studies on the stimulation of TNF alpha production in brain injury and on the deleterious consequence of this acute response. Its inhibition by pharmacologic agents, neutralizing antibodies or soluble receptors has protective effects. In contrast, there are reports (from in-vitro studies or knock-out mice) on the beneficial effects of TNF alpha. To reconcile these apparently conflicting reports, the exact timing and extent of TNF alpha activation must be taken into account, as well as the presence of other mediators such as reactive oxygen species. It is suggested that the appropriate context of mediators, at any given time after brain injury may well determine whether the effect of TNF alpha is protective or toxic.


Assuntos
Lesões Encefálicas/metabolismo , Isquemia Encefálica/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Animais , Lesões Encefálicas/tratamento farmacológico , Proteínas de Transporte/metabolismo , Proteínas de Transporte/farmacologia , Humanos , Camundongos , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral , Receptores Chamariz do Fator de Necrose Tumoral , Fator de Necrose Tumoral alfa/farmacologia
3.
J Neuropathol Exp Neurol ; 47(4): 475-87, 1988 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-3385440

RESUMO

Histological examination by light and electron microscopy of the spinal cords of four dogs rapidly perfusion-fixed after the onset of decompression sickness revealed the presence of numerous non-staining, space-occupying lesions that were absent in similarly prepared sections of control or ischemic spinal cords. We propose the hypothesis that these lesions are caused by the liberation of a gas phase. The possible significance of these lesions in the evolution of spinal cord dysfunction is discussed with reference to the principal theories of the pathogenesis of spinal cord decompression sickness.


Assuntos
Doença da Descompressão/etiologia , Gases , Doenças da Medula Espinal/etiologia , Animais , Doença da Descompressão/patologia , Doença da Descompressão/fisiopatologia , Mergulho/efeitos adversos , Potenciais Somatossensoriais Evocados , Isquemia/fisiopatologia , Microscopia Eletrônica , Valores de Referência , Fluxo Sanguíneo Regional , Medula Espinal/irrigação sanguínea , Medula Espinal/patologia , Medula Espinal/ultraestrutura , Doenças da Medula Espinal/patologia , Doenças da Medula Espinal/fisiopatologia
4.
Brain Pathol ; 10(1): 95-112, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10668900

RESUMO

Ischemic stroke is a leading cause of death and disability in developed countries. Yet, in spite of substantial research and development efforts, no specific therapy for stroke is available. Several mechanism for neuroprotection have been explored including ion channels, excitatory amino acids and oxygen radicals yet none has culminated in an effective therapeutic effect. The review article on "inflammation and stroke" summarizes key data in support for the possibility that inflammatory cells and mediators are important contributing and confounding factors in ischemic brain injury. In particular, the role of cytokines, endothelial cells and leukocyte adhesion molecules, nitric oxide and cyclooxygenase (COX-2) products are discussed. Furthermore, the potential role for certain cytokines in modulation of brain vulnerability to ischemia is also reviewed. The data suggest that novel therapeutic strategies may evolve from detailed research on some specific inflammatory factors that act in spatial and temporal relationships with traditionally recognized neurotoxic factors. The dual nature of some mediators in reformatting of brain cells for resistance or sensitivity to injury demonstrate the delicate balance needed in interventions based on anti-inflammatory strategies.


Assuntos
Encéfalo/fisiopatologia , Moléculas de Adesão Celular/metabolismo , Citocinas/metabolismo , Hipóxia-Isquemia Encefálica/fisiopatologia , Óxido Nítrico Sintase/metabolismo , Acidente Vascular Cerebral/fisiopatologia , Animais , Humanos , Hipóxia-Isquemia Encefálica/metabolismo , Inflamação/metabolismo , Inflamação/fisiopatologia , Acidente Vascular Cerebral/metabolismo
5.
Stroke ; 32(11): 2665-74, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11692032

RESUMO

BACKGROUND AND PURPOSE: Enlimomab, a murine monoclonal anti-human intercellular adhesion molecule (ICAM)-1 antibody, had a negative outcome in a multicenter acute-stroke trial. We did a bedside-to-bench study in standardized rat stroke models to explore mechanisms for these untoward results. METHODS: After focal brain ischemia in Wistar rats and spontaneously hypertensive rats (SHR), we administered murine anti-rat ICAM-1 antibody (1A29), subclass-matched murine immunoglobulin (IgG1), or vehicle intravenously. To examine whether rat anti-mouse antibodies were generated against the mouse protein and whether these were deleterious, we sensitized Wistar rats with 1A29 or vehicle 7 days before surgery. Infarct volume, tissue myeloperoxidase activity, neutrophil CD11b expression, and microvascular E-selectin, P-selectin, and ICAM-1 expression were examined 48 hours after surgery. Complement activation was serially assessed for 2 hours after a single injection of either 1A29 or vehicle. RESULTS: 1A29 treatment did not significantly reduce infarct size in either strain. 1A29 sensitization augmented infarct size and generated rat anti-mouse antibodies. Although 1A29 inhibited neutrophil trafficking shown by reduction in brain myeloperoxidase activity, circulating neutrophils were activated and displayed CD11b upregulation. Complement was activated in 1A29-sensitized Wistar rats and 1A29-treated SHR. E-selectin (SHR), endothelial P-selectin (Wistar and SHR), and ICAM-1 (SHR) were upregulated in animals treated with 1A29. CONCLUSIONS: Administration to rats of a murine antibody preparation against ICAM-1, 1A29, elicits the production of host antibodies against the protein, activation of circulating neutrophils, complement activation, and sustained microvascular activation. These observations provide several possible mechanisms for central nervous system-related clinical deterioration that occurred when Enlimomab was given in acute ischemic stroke.


Assuntos
Anticorpos Monoclonais/efeitos adversos , Infarto Encefálico/etiologia , Complemento C3a/análogos & derivados , Molécula 1 de Adesão Intercelular/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Peso Corporal , Encéfalo/enzimologia , Infarto Encefálico/imunologia , Infarto Encefálico/patologia , Isquemia Encefálica/etiologia , Isquemia Encefálica/imunologia , Isquemia Encefálica/patologia , Circulação Cerebrovascular , Ensaios Clínicos como Assunto , Complemento C3a/análise , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Isoanticorpos/efeitos adversos , Isoanticorpos/imunologia , Isoanticorpos/uso terapêutico , Fluxometria por Laser-Doppler , Contagem de Leucócitos , Camundongos , Peroxidase/metabolismo , Ratos , Ratos Endogâmicos SHR , Ratos Wistar , Selectinas/análise , Selectinas/imunologia , Acidente Vascular Cerebral/terapia
6.
J Cereb Blood Flow Metab ; 16(1): 170-4, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8530550

RESUMO

The utility of microtubule-associated protein 2 (MAP2) immunostaining as a marker of acute focal ischemic injury was investigated. Permanent middle cerebral artery occlusion (MCAO) elicited a rapid reduction in MAP2 immunostaining that was visible 1 h post-MCAO and that increased in intensity and area encompassed over time. The ischemic lesion borders were well defined by loss of MAP2 immunostaining, but alterations in staining within the lesion were more heterogeneous. Lesion volume increased significantly from 1 to 4 h post-MCAO (from 63.8 +/- 10.8 to 111.3 +/- 19.0 mm3, mean +/- SD). Thus, MAP2 immunostaining is a sensitive, quantifiable indicator of acute brain injury following focal ischemia.


Assuntos
Ataque Isquêmico Transitório/patologia , Proteínas Associadas aos Microtúbulos/análise , Análise de Variância , Animais , Biomarcadores/análise , Córtex Cerebral/química , Imuno-Histoquímica , Masculino , Ratos , Ratos Endogâmicos SHR , Fatores de Tempo
7.
J Cereb Blood Flow Metab ; 18(2): 168-75, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9469159

RESUMO

Hibernation in mammals is associated with a regulated depression of global cellular functions accompanied by reductions of cerebral blood flow that would render the brain profoundly ischemic under normal conditions. Homeostatic control is preserved, however, and brain damage does not occur. We investigated the possibility that hibernation not only confers tolerance to profound hypothermia, but also to hypoxia and aglycemia independent of temperature. Hippocampal slices from ground squirrels Citellus tridecemlineatus in both the active and hibernating states and from rats were subjected to in vitro hypoxia and aglycemia at incubation temperatures of 36 degrees C, 20 degrees C, and 7 degrees C and evaluated histologically. A binary bioassay was used to determine the duration of hypoxia/aglycemia tolerated in each group. At all temperatures, slices from hibernating animals were most tolerant compared with both active squirrels and rats. Slices from active ground squirrels were more tolerant than rat at 20 degrees C and 7 degrees C but not at 36 degrees C indicating a species-specific difference that becomes manifest at lower temperatures. These results indicate that hibernation is associated not only with tolerance to profound hypothermia but also to deprivation of oxygen and glucose. Because tolerance was already demonstrable at the shortest duration of hibernation studied, rapid therapeutic induction of a similar state may be possible. Therefore, identification of the regulatory mechanisms underlying this tolerance may lead to novel neuroprotective strategies.


Assuntos
Adaptação Fisiológica , Glicemia/metabolismo , Hibernação/fisiologia , Hipocampo/fisiologia , Hipóxia , Sciuridae/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Cromatina/ultraestrutura , Citoplasma/ultraestrutura , Glucose/administração & dosagem , Hipocampo/ultraestrutura , Hipotermia , Técnicas In Vitro , Masculino , Oxigênio/administração & dosagem , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie , Temperatura
8.
J Cereb Blood Flow Metab ; 21(1): 34-40, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11149666

RESUMO

Preconditioning brain with tumor necrosis factor alpha (TNF-alpha) can induce tolerance to experimental hypoxia and stroke and ceramide is a downstream messenger in the TNF-alpha signaling pathway. A hypoxic-ischemic (HI) insult in the immature rat injures brain primarily through apoptosis. Apoptosis is regulated by Bcl-2 family proteins. The authors explored whether ceramide protects against HI in the immature rat, and whether Bcl-2 family protein expression is involved. Hypoxia-ischemia was produced in seven-day-old rats by ligating the right carotid artery, followed by 2 hours of 8% oxygen exposure. Thirty minutes after HI, C2-ceramide (150 microg/kg) was injected intraventricularly. Infarct volume was measured 5 days later. C2-ceramide reduced HI-induced brain damage by 45% to 65% compared with HI/dimethyl sulfoxide (DMSO) (vehicle control) or HI only groups. In separate experiments, brains of sham-operated control and HI only animals and animals subjected to HI plus C2-ceramide or DMSO infusion were sampled 6 hours, 24 hours, and 5 days after treatments and analyzed for Bcl-2, Bcl-xl, and Bax expression (Western blotting), and apoptosis (TUNEL assay). Augmented Bcl-2 and Bcl-xl levels in the C2-ceramide treated group were associated with a significant decrease in TUNEL-positive cells. The results support a protective role for ceramide in neonatal HI.


Assuntos
Apoptose , Encéfalo/fisiopatologia , Regulação da Expressão Gênica , Genes bcl-2 , Hipóxia-Isquemia Encefálica/fisiopatologia , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/análise , Esfingosina/análogos & derivados , Esfingosina/uso terapêutico , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Modelos Animais de Doenças , Hipóxia-Isquemia Encefálica/prevenção & controle , Marcação In Situ das Extremidades Cortadas , Infusões Parenterais , Neurônios/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Esfingosina/administração & dosagem , Técnicas Estereotáxicas , Fator de Necrose Tumoral alfa/fisiologia
9.
J Cereb Blood Flow Metab ; 17(2): 229-32, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9040503

RESUMO

Tumor necrosis factor alpha (TNF-alpha) is expressed in the ischemic brain; however, its precise role is not fully understood. We studied the effect of the dimeric form of the type I soluble TNF receptor linked to polyethylene glycol (TNFbp) on focal cerebral ischemia in mice using a permanent middle cerebral arterial occlusion (MCAO) model. TNFbp was applied topically, intravenously, or intraperitoneally. TNFbp binds and inhibits TNF-alpha. The volume of cortical ischemic lesions was measured by means of 2,3,5-triphenyltetrazolium chloride 24 h after MCAO. TNFbp produced a significant reduction in the cortical infarct volume of vehicle-treated animals (p < 0.001). The reduction in the volume of brain damage was 26% in animals that received 3 mg/kg of TNFbp topically. Further analysis of TNF-alpha inhibition following acute brain ischemia is indicated.


Assuntos
Infarto Cerebral/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Receptores do Fator de Necrose Tumoral , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Isquemia Encefálica/complicações , Infarto Cerebral/etiologia , Infarto Cerebral/patologia , Avaliação Pré-Clínica de Medicamentos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/farmacologia , Receptores Tipo I de Fatores de Necrose Tumoral , Receptores Chamariz do Fator de Necrose Tumoral , Fator de Necrose Tumoral alfa/fisiologia
10.
J Cereb Blood Flow Metab ; 20(7): 1040-4, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10908037

RESUMO

Astrocytes perform a variety of functions in the adult central nervous system (CNS) that contribute to the survival of neurons. Thus, it is likely that the activities of astrocytes affect the extent of brain damage after ischemic stroke. The authors tested this hypothesis by using a mouse ischemia model to compare the infarct volume produced in wild-type mice with that produced in mice lacking glial fibrillary acidic protein (GFAP), an astrocyte specific intermediate filament component. Astrocytes lacking GFAP have been shown to have defects in process formation, induction of the blood-brain barrier. and volume regulation; therefore, they might be compromised in their ability to protect the CNS after injury. The authors reported here that 48 hours after combined permanent middle cerebral artery occlusion (MCAO) and 15 minutes transient carotid artery occlusion (CAO) GFAP-null mice had a significantly (P < 0.001) larger cortical infarct volume (16.7 +/- 2.2 mm3) than their wild-type littermates (10.1 +/- 3.9 mm3). Laser-Doppler flowmetry revealed that the GFAP-null mice had a more extensive and profound decrease in cortical cerebral blood flow within 2 minutes after MCAO with CAO. These results indicated a high susceptibility to cerebral ischemia in GFAP-null mice and suggested an important role for astrocytes and GFAP in the progress of ischemic brain damage after focal cerebral ischemia with partial reperfusion.


Assuntos
Isquemia Encefálica/etiologia , Proteína Glial Fibrilar Ácida/deficiência , Animais , Encéfalo/patologia , Isquemia Encefálica/patologia , Isquemia Encefálica/fisiopatologia , Circulação Cerebrovascular , Suscetibilidade a Doenças , Deleção de Genes , Proteína Glial Fibrilar Ácida/genética , Fluxometria por Laser-Doppler , Camundongos , Camundongos Mutantes/genética , Valores de Referência
11.
J Cereb Blood Flow Metab ; 21(3): 226-32, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11295877

RESUMO

Previous work in primary cell culture has shown that TNF-alpha and ceramide are involved in the signaling that induces tolerance to brain ischemia (Ginis et al., 1999; Liu et al., 2000). To validate the in vitro studies, the authors administered cell permeable analogs of ceramides intracisternally or intravenously to examine their effect on neuroprotection after focal cerebral ischemia. Permanent middle cerebral artery occlusion (MCAO) was performed in spontaneously hypertensive rats. Infarct volumes were assessed at 24 hours after surgery. D-erythro-N-acetylsphingosine (C2-ceramide) or its vehicle was infused intracisternally for 1 hour before MCAO. In a second set of studies, D-erythro-N-octanoylsphingosine (C8-ceramide) or its vehicle was injected intravenously 48 or 24 hours before MCAO to mimic preconditioning (PC) and was also injected 5 minutes after MCAO. C2-ceramide infusion significantly reduced infarct volumes by approximately 14% (P < 0.05). C8-ceramide injection reduced infarct volumes approximately 17% compared with controls. This effect was constant and significant compared with controls over the time periods examined (P < 0.01). This work supports findings in primary brain cell cultures that implicate ceramide as a downstream signal that is proximate to development of tolerance to brain ischemia. Because the degree of protection represents approximately 50% of the maximal infarct reduction observed in this model, there are probably additional signaling pathways that subserve tolerance.


Assuntos
Isquemia Encefálica/metabolismo , Infarto da Artéria Cerebral Média/tratamento farmacológico , Precondicionamento Isquêmico , Esfingosina/análogos & derivados , Esfingosina/farmacocinética , Animais , Permeabilidade da Membrana Celular , Ceramidas/farmacologia , Técnicas In Vitro , Infarto da Artéria Cerebral Média/metabolismo , Injeções Intravenosas , Injeções Intraventriculares , Lipopolissacarídeos , Masculino , Ratos , Ratos Endogâmicos SHR , Fator de Necrose Tumoral alfa
12.
J Cereb Blood Flow Metab ; 21(3): 244-52, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11295879

RESUMO

Studies of vascular biology during the past decade have identified an expanding list of agonists and antagonists that regulate local hemostasis, inflammation, and reactivity in blood vessels. Interactions at the blood-endothelial interface are intricate and complex and have been postulated to play a role in the initiation of stroke and the progression of brain injury during early hours of ischemia, particularly in conjunction with reperfusion injury (Hallenbeck, 1996). In the current study of normal and activated vessels in rat brain, immunoreactive tumor necrosis factor-alpha (TNF-alpha), heme oxygenase-1 (HO-1), and manganese superoxide dismutase (MnSOD) exhibit concentric perivascular rings involving vessel wall and surrounding parenchyma that appear to coincide with one another in serial sections. The ring patterns suggest periodic radial expansion of these molecules released through a process of cyclic activation and inactivation of brain vessel segments. In this process, the rings appear randomly scattered instead of affecting all vessels within a high power field (HPF) synchronously. The average number of vessels per HPF (mean +/- SD) with perivascular cuffs of immunoreactive MnSOD increased from 51 +/- 28 in Wistar, 72 +/- 46 in Wistar-Kyoto, and 84 +/- 30 in Sprague Dawley rats (no spontaneous strokes) to 184 +/- 72 in spontaneously hypertensive stroke-prone rats (spontaneous strokes). Perivascular immunoreactive cuffs are also increased in spontaneously hypertensive rats by induction of cytokine expression by lipopolysaccharide (64 +/- 15 vs. 131 +/- 32 /HPF). The patterns of TNF-alpha, HO-1, and MnSOD in naïve animals are interpreted to indicate that focal hemostatic balance normally fluctuates in brain vessels and influences surrounding parenchymal cells. Perivascular immunoreactive cuffs representing this process are more frequent in animals with lipopolysaccharide-induced endothelial activation or genetic stroke proneness.


Assuntos
Artérias Cerebrais/enzimologia , Heme Oxigenase (Desciclizante)/análise , Superóxido Dismutase/análise , Fator de Necrose Tumoral alfa/análise , Aldeídos/análise , Animais , Encéfalo/irrigação sanguínea , Artérias Cerebrais/química , Corantes , Endotélio Vascular/química , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/enzimologia , Amarelo de Eosina-(YS) , Corantes Fluorescentes , Hematoxilina , Heme Oxigenase-1 , Imuno-Histoquímica , Lipopolissacarídeos/farmacologia , Masculino , Malondialdeído/análise , Estresse Oxidativo/fisiologia , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Ratos Sprague-Dawley , Ratos Wistar
13.
J Cereb Blood Flow Metab ; 17(5): 483-90, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9183285

RESUMO

Cytokines are recognized to play an important role in acute stroke. Tumor necrosis factor-alpha (TNF) is one of the pro-inflammatory cytokines and is expressed in ischemic brain. We hypothesized that TNF might play a role in the regulation of tolerance to ischemia when administered prior to the ischemic episode. We studied the effects of pretreatment of TNF administered intravenously, intraperitoneally, or intracisternally in mice that were subjected to middle cerebral artery occlusion (MCAO) 48 h later. MCAO was performed in BALB/C mice by direct cauterization of distal MCA, which resulted in pure cortical infarction. A significant reduction in infarct size was noted in mice pretreated by TNF at the dose of 0.5 microgram/mouse (p < 0.01) intracisternally. At the doses used in this study, administration of TNF by intravenous or intraperitoneal routes was not effective. Immunohistochemical analysis of brains subjected to 24 h of MCAO revealed a significant decrease in CD11b immunoreactivity after TNF pretreatment compared with control MCAO. Preconditioning with TNF affects infarct size in a time- and dose-dependent manner. TNF induces significant protection against ischemic brain injury and is likely to be involved in the signaling pathways that regulate ischemic tolerance.


Assuntos
Isquemia Encefálica/prevenção & controle , Fator de Necrose Tumoral alfa/administração & dosagem , Animais , Isquemia Encefálica/patologia , Relação Dose-Resposta a Droga , Injeções Intraperitoneais , Injeções Intravenosas , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos BALB C
14.
J Cereb Blood Flow Metab ; 14(2): 193-205, 1994 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8113316

RESUMO

The breakdown of cellular homeostasis and progressive neuronal destruction in cerebral ischemia appears to be mediated by a complex network of causes that are intricately interrelated. We have investigated a physiological state existing normally in nature in which mammals appear to tolerate the ordinarily detrimental effects of ischemia with reduced oxygen availability and to resist activation of self-destructive processes, i.e., mammalian hibernation. Ground squirrels (Spermophilus tridecemlineatus) were chronically implanted with arterial and venous catheters and telemetry devices for electroencephalography, electrocardiography, and monitoring of body temperature. The animals were placed in an environmental chamber at an ambient temperature of 5 degrees C. Entrance into hibernation was characterized by a drop in heart rate followed by a gradual decline in body temperature and an isoelectric electroencephalogram. Cold-adapted active animals that were not hibernating served as controls. Cerebral blood flow (CBF) was measured in both groups with the autoradiographic [14C]iodoantipyrine method. Mean (+/- SD) mass-weighted CBF in the brain was 62 +/- 18 ml 100 g(-1) min (-1) (n = 4) in the control group but was reduced to ischemic levels, 7 +/- 4 ml 100 g(-1) min (-1) (n = 4), in the hibernating animals (p < 0.001) [corrected]. No neuropathological changes were found in similarly hibernating animals aroused from hibernation. Hibernation appears to be actively regulated, and hormonal factors may be involved. The identification and characterization of such factors and of the mechanisms used by hibernating species to increase ischemic tolerance and to blunt the destructive effects of ischemia may enable us to prevent or minimize the loss of homeostatic control during and after cerebral ischemia in other species.


Assuntos
Adaptação Fisiológica , Isquemia Encefálica/fisiopatologia , Circulação Cerebrovascular , Hibernação/fisiologia , Animais , Antipirina/análogos & derivados , Antipirina/farmacocinética , Autorradiografia , Comportamento Animal/fisiologia , Células Sanguíneas/fisiologia , Encéfalo/metabolismo , Encéfalo/fisiologia , Eletrocardiografia , Eletroencefalografia , Sciuridae
15.
J Cereb Blood Flow Metab ; 16(6): 1137-42, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8898685

RESUMO

A series of experiments was performed to determine the role of interleukin (IL)-1 in the induction of tolerance to global ischemia in Mongolian gerbils. In Group I, a 2-min "preconditioning" ischemia protected CA1 hippocampal neurons in gerbils subjected to 3.5 min ischemia 3 days later. CA1 neuronal density was: sham, 171 +/- 3/mm; 3.5 min ischemia, 30 +/- 30/mm; 2 and 3.5 min ischemia 162 +/- 6/mm. Experiments in Group II addressed the role of IL-1 in the induction of tolerance by sublethal ischemia. Arterial IL-1 alpha and IL-1 beta became elevated between 1 and 3 days after a 2-min ischemic exposure. IL-1 alpha was: sham, 6.4 +/- 0.6 ng/ml; and 2-day, 10.2 +/- 1.2 ng/ml. IL-1 beta was: sham, 6.4 +/- 0.5 ng/ml; and 2-day, 17.3 +/- 2 ng/ml. Recombinant human IL-1 receptor antagonist (IL-1ra) i.p. blocked ischemic tolerance induction by 2-min preconditioning ischemia: 2-min ischemia + vehicle, 162 +/- 6/mm; and 2-min ischemia + IL-1ra, 67 +/- 17/mm. Experiments in Group III assessed the capacity of IL-1 to induce tolerance to brain ischemia. IL-1 alpha i.p. (0, 10, 20 micrograms/kg) for 3 days prior to 3.5-min forebrain ischemia provided significant CA1 neuroprotection in a dose-dependent manner: 2 +/- 2, 68 +/- 83, and 129 +/- 42/mm, respectively. IL-1 beta (15 micrograms/kg) in combination with either IL-1ra (100 mg/kg) or IL-1ra vehicle i.p. on the same schedule demonstrated a significant CA1 neuroprotection that could be nullified by IL-1ra: IL-1 beta + IL-1ra vehicle, 153 +/- 16/mm; and IL-1 beta + IL-1ra, 67 +/- 36/mm. Recognition that tolerance arises from stimulation of a known receptor (IL-1RI) permits molecular analysis of the intracellular signaling that is critical for production of that state.


Assuntos
Isquemia Encefálica/prevenção & controle , Hipocampo/patologia , Interleucina-1/administração & dosagem , Neurônios/patologia , Animais , Isquemia Encefálica/sangue , Isquemia Encefálica/patologia , Morte Celular/efeitos dos fármacos , Gerbillinae , Humanos , Proteína Antagonista do Receptor de Interleucina 1 , Interleucina-1/sangue , Receptores de Interleucina-1/antagonistas & inibidores , Proteínas Recombinantes/administração & dosagem , Sialoglicoproteínas/administração & dosagem
16.
J Cereb Blood Flow Metab ; 9(6): 902-5, 1989 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-2584280

RESUMO

The mechanisms of secondary brain damage following cerebral ischemia or CNS trauma are still unknown. A variety of mediators, released by the injured brain, are held to be responsible for delayed neuronal cell damage. No technique is available yet for repeated sampling of cerebral venous blood (CVB) on a long-term basis, which allows to assess the neurobiochemical responses to brain injury chronically in conscious laboratory animals. This is a report on the establishment of a method to sample CVB from the superior sagittal sinus (SSS) for at least 7 days in conscious freely moving rats. The torcular was exposed for implantation of a catheter into the caudal SSS. Patency was preserved by perfusion with a nonsystemic dose of heparin by an Alzet osmotic pump and verified daily by aspirating of 0.5 ml CVB for blood analysis. No neuropathology was seen in histological examination. The rats appeared comfortable and without distress, and the transient weight loss was rapidly regained. The present model provides the opportunity to evaluate acute or chronic neurobiochemical responses to stroke, CNS trauma, or any other typ of brain injury in the conscious rat.


Assuntos
Coleta de Amostras Sanguíneas/métodos , Veias Cerebrais , Animais , Coleta de Amostras Sanguíneas/instrumentação , Barreira Hematoencefálica , Cateteres de Demora , Heparina , Masculino , Perfusão , Ratos , Ratos Endogâmicos
17.
J Cereb Blood Flow Metab ; 11(1): 88-98, 1991 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-1984008

RESUMO

To study the causes of spatial and temporal evolution of progressive neuro-injury in focal brain ischemia, models with consistent lesion topography are required. In such models, continuous monitoring of the microcirculation in a penumbral area undergoing progressive damage could be possible. We used a fixed-pulse (1.0 s, 40 W) Nd-YAG laser (NYL) to produced discrete brain lesions in rats and monitored the cerebral blood flow (CBF) with laser-Doppler flowmetry (LDF) in nonirradiated areas directly adjacent to the maturing lesion. We also examined the time evolution of the lesion topography over a 4 day period. The lesion volume determined by histopathological methods increased from 3.1 +/- 0.5 to 4.5 +/- 0.5 mm3 (p less than 0.05) during the first 2 h. Simultaneously, LDF indicated severe hypoperfusion (-60 +/- 21%, p less than 0.01) at a zone (1 mm distance from the laser lesion) where progressive neuronal degeneration and increased tissue water content (80.0 +/- 3.3% versus 76.8 +/- 2.1% in normal tissue, n = 7, p less than 0.05) were also observed. At a 4 mm distance from the lesion, hyperemic CBF responses were observed, but no histopathological signs or edema. Secondary brain damage progressed up to 4 days (lesion volume of 6.0 +/- 0.7 mm3). The NYL-induced brain lesion produced a highly reproducible focal injury and progressive neuronal death in a spatial relationship with microcirculatory failure and edema formation. The model allows prospective study of tissue state at a discrete zone, which is separate from the initial injury, but susceptible to secondary brain damage.


Assuntos
Encefalopatias/etiologia , Isquemia Encefálica/complicações , Córtex Cerebral/irrigação sanguínea , Modelos Animais de Doenças , Lasers , Microcirculação/fisiopatologia , Animais , Encéfalo/patologia , Encefalopatias/patologia , Encefalopatias/fisiopatologia , Edema Encefálico , Isquemia Encefálica/etiologia , Córtex Cerebral/patologia , Masculino , Neurônios/patologia , Ratos , Ratos Endogâmicos
18.
J Cereb Blood Flow Metab ; 16(1): 82-91, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8530560

RESUMO

Cortical brain damage was produced in rats by a focal pulse from a Nd-YAG laser, and evolution of the lesion was evaluated at 30 min, and 2, 8, and 24 h with respect to microvascular perfusion, blood-brain barrier (BBB) permeability, and expression of both the heat-shock/stress protein, hsp72, and the c-fos proto-oncogene transcription factor. A double-labeling fluorescence technique employing intravenously injected Evans blue albumin (EBA) and fluorescein-labeled dextran was used to map and measure BBB damage and microvascular perfusion in fresh frozen brain sections. Hsp72 and c-fos mRNAs were localized by in situ hybridization, and the respective proteins were identified by immunocytochemistry. Parallel sections were stained for glial fibrillary acidic protein and for routine histologic examination. Striking hsp72 mRNA expression was evident by 2 h in an approximately 300 microns wide rim surrounding an area of expanding BBB damage. Increased hsp72 mRNA was observed only in regions of preserved microcirculation, where the hsp72 protein was subsequently localized exclusively in the vasculature at 24 h after the insult. Hsp72-positive endothelial cells spanned the narrow margin between the lesion and histologically normal, glial fibrillary acidic protein (GFAP)-positive cortical tissue. There was no hsp72 expression in the area of subcortically migrating edema fluid. Inductions of c-fos mRNA and Fos protein were not strikingly evident around the focal brain lesion, but were observed transiently throughout the injured hemisphere at 30 min and 2.5 h, respectively, indicating that spreading depression was triggered by the focal injury. These results are in striking contrast to those previously obtained from studies of models of focal ischemic or traumatic brain injury, which are characterized by a complex pattern of glial and neuronal hsp72 expression in the periphery of an infarct, and which suggest that the tightly demarcated lesion produced by the Nd-YAG laser lacks these components of graded injury that are evident following other types of focal brain damage.


Assuntos
Lesões Encefálicas/metabolismo , Córtex Cerebral/lesões , Proteínas de Choque Térmico/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Animais , Sequência de Bases , Barreira Hematoencefálica/fisiologia , Química Encefálica , Córtex Cerebral/irrigação sanguínea , Circulação Cerebrovascular , Endotélio Vascular/química , Proteína Glial Fibrilar Ácida/análise , Proteínas de Choque Térmico HSP72 , Proteínas de Choque Térmico/análise , Imuno-Histoquímica , Hibridização In Situ , Masculino , Microcirculação , Microscopia de Fluorescência , Dados de Sequência Molecular , Proteínas Proto-Oncogênicas c-fos/análise , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley
19.
J Cereb Blood Flow Metab ; 18(9): 1040-5, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9740108

RESUMO

Mammalian hibernation is a state of natural tolerance to severely decreased brain blood flow. As protein tyrosine phosphorylation is believed to be involved in the development of resistance to potentially cell-damaging insults, we used immunoblotting for the phosphotyrosine moiety to analyze extracts from various tissues of hibernating and nonhibernating ground squirrels. A single, hibernation-specific phosphoprotein was detected in the brain, but not in any other tissue tested. This protein, designated pp98 to reflect its apparent molecular weight, is distributed throughout the brain, and is associated with the cellular membrane fraction. The presence of the protein is tightly linked to the hibernation state; it is not present in contemporaneously assayed animals that are exposed to the same cold temperature as the hibernators, is present for the duration of a hibernation bout (tested from 1 to 14 days), and disappears within 1 hour of arousal from hibernation. The close association of pp98 with the hibernation state, its presence in cellular membranes, and the known properties of membrane phosphotyrosine proteins suggest that it may transduce a signal for adaptation to the limited availability of oxygen and glucose and low cellular temperature that characterizes hibernation in the ground squirrel.


Assuntos
Encéfalo/fisiologia , Hibernação/fisiologia , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Tirosina Quinases/metabolismo , Sciuridae/fisiologia , Animais , Encéfalo/metabolismo , Clima Frio , Fosforilação , Transdução de Sinais/fisiologia
20.
J Cereb Blood Flow Metab ; 19(6): 616-23, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10366191

RESUMO

Lipopolysaccharide (LPS), administered 72 hours before middle cerebral artery (MCA) occlusion, confers significant protection against ischemic injury. For example, in the present study, LPS (0.9 mg/kg intravenously) induced a 31% reduction in infarct volume (compared with saline control) assessed 24 hours after permanent MCA occlusion. To determine whether LPS induces true tolerance to ischemia, or merely attenuates initial ischemic severity by augmenting collateral blood flow, local CBF was measured autoradiographically 15 minutes after MCA occlusion. Local CBF did not differ significantly between LPS- and saline-pretreated rats (e.g., 34 +/- 10 and 29 +/- 15 mL x 100 g(-1) x min(-1) for saline and LPS pretreatment in a representative region of ischemic cortex), indicating that the neuroprotective action of LPS is not attributable to an immediate reduction in the degree of ischemia induced by MCA occlusion, and that LPS does indeed induce a state of ischemic tolerance. In contrast to the similarity of the initial ischemic insult between tolerant (LPS-pretreated) and nontolerant (saline-pretreated) rats, microvascular perfusion assessed either 4 hours or 24 hours after MCA occlusion was preserved at significantly higher levels in the LPS-pretreated rats than in controls. Furthermore, the regions of preserved perfusion in tolerant animals were associated with regions of tissue sparing. These results suggest that LPS-induced tolerance to focal ischemia is at least partly dependent on the active maintenance of microvascular patency and hence the prevention of secondary ischemic injury.


Assuntos
Isquemia Encefálica/fisiopatologia , Circulação Cerebrovascular/efeitos dos fármacos , Endotoxinas/farmacologia , Lipopolissacarídeos/farmacologia , Animais , Gasometria , Pressão Sanguínea/efeitos dos fármacos , Artérias Cerebrais/fisiologia , Escherichia coli/metabolismo , Masculino , Microcirculação/efeitos dos fármacos , Óxido Nítrico/sangue , Ratos , Ratos Endogâmicos SHR
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa