Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stroke ; 32(10): 2356-61, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11588326

RESUMO

BACKGROUND AND PURPOSE: Reactive oxygen species produced during reperfusion may play a detrimental role in focal cerebral ischemia (FCI). We examined the protein expression of caspase-8, which plays a major role in both Fas-dependent and cytochrome c-dependent apoptotic pathways after FCI with or without reperfusion. Caspase-8 expression after transient FCI was compared between wild-type and transgenic mice that overexpress the cytosolic antioxidant copper/zinc superoxide dismutase (SOD1). METHODS: Adult male CD-1 mice were subjected to 1 hour of FCI and reperfusion or to permanent FCI by intraluminal blockade of the middle cerebral artery. DNA fragmentation was evaluated by genomic DNA gel electrophoresis. Caspase-8 expression was analyzed by Western blot. RESULTS: Caspase-8 was significantly induced 4 hours after transient FCI and remained at an increased level until 24 hours, whereas it was not modified after permanent FCI. Genomic DNA gel electrophoresis showed DNA laddering in a pattern similar to that seen in apoptosis, with a small amount of background smear 24 hours after transient FCI, whereas 25 hours of permanent FCI resulted in less DNA laddering with a strong background smear. Caspase-8 induction was significantly reduced in SOD1 transgenic mice compared with wild-type mice 4 hours after transient FCI. CONCLUSIONS: The results suggest that increased reactive oxygen species production during reperfusion may contribute to the induction of caspase-8, thereby exacerbating apoptosis after FCI.


Assuntos
Apoptose , Isquemia Encefálica/metabolismo , Caspases/biossíntese , Acidente Vascular Cerebral/metabolismo , Superóxidos/metabolismo , Animais , Western Blotting , Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Isquemia Encefálica/complicações , Caspase 8 , Caspase 9 , Grupo dos Citocromos c/metabolismo , Fragmentação do DNA , Progressão da Doença , Indução Enzimática/efeitos dos fármacos , Ataque Isquêmico Transitório/complicações , Ataque Isquêmico Transitório/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Mitocôndrias/enzimologia , Reperfusão , Acidente Vascular Cerebral/complicações , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1 , Fatores de Tempo
2.
J Cereb Blood Flow Metab ; 21(5): 557-67, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11333366

RESUMO

Release of cytochrome c from mitochondria to cytosol is a critical step in the mitochondrial-dependent signaling pathways of apoptosis. The authors have reported that manganese superoxide dismutase (Mn-SOD) attenuated cytochrome c release and apoptotic cell death after focal cerebral ischemia (FCI). To investigate downstream to the cytochrome c-dependent pathway, the authors examined caspase-9 activation after transient FCI by immunohistochemistry and Western blotting in both wild-type and Sod2 -/+ mice. Mice were subjected to 60 minutes of middle cerebral artery occlusion followed by 1, 2, 4, or 24 hours of reperfusion. Two hours after reperfusion, cytochrome c and caspase-9 were observed in the cytosol and significantly increased in Sod2 -/+ mutants compared with wild-type mice as shown by Western blotting. Immunofluorescent double labeling for cytochrome c and caspase-9 showed cytosolic cytochrome c 1 hour after transient FCI. Cleaved caspase-9 first appeared in the cytosol at 2 hours and colocalized with cytochrome c. Terminal deoxynucleotidyl transferase-mediated uridine 5;-triphosphate-biotin nick and labeling (TUNEL) showed significant increase of positive cells in Sod2 -/+ mice compared with the wild-type in the cortex, but not in the caudate putamen. The current study revealed Mn-SOD might affect cytochrome c translocation and downstream caspase activation in the mitochondrial-dependent cell death pathway after transient FCI.


Assuntos
Encéfalo/enzimologia , Caspases/metabolismo , Grupo dos Citocromos c/metabolismo , Ataque Isquêmico Transitório/enzimologia , Superóxido Dismutase/fisiologia , Animais , Apoptose , Western Blotting , Encéfalo/ultraestrutura , Caspase 9 , Citosol/metabolismo , Fragmentação do DNA , Ativação Enzimática , Imunofluorescência , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Ataque Isquêmico Transitório/patologia , Camundongos , Camundongos Knockout , Mitocôndrias/metabolismo , Superóxido Dismutase/deficiência , Superóxido Dismutase/genética
3.
Stroke ; 32(2): 506-15, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11157190

RESUMO

BACKGROUND AND PURPOSE: We sought to investigate the mechanisms for oxidative injury caused by subarachnoid hemolysate, a pro-oxidant. METHODS: Injection of 50 microL of subarachnoid hemolysate or saline was performed in CD1 mice (n=75), mutant mice deficient in Mn-superoxide dismutase (Sod2+/-; n=23), and their wild-type littermates (n=23). Subcellular location of cytochrome c was studied by immunocytochemistry, immunofluorescence, and immunoblotting of cellular fractions. DNA fragmentation was assessed though DNA laddering and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling (TUNEL). Cell death was examined through basic histology. RESULTS: Cytochrome c immunoreactivity was present in the cytosol of neurons at 2 hours after hemolysate injection and increased by 4 hours compared with saline-injected animals (P:<0.02). Cytosolic cytochrome c was more abundant in Sod2+/- mutants. DNA fragmentation was evident at 24 hours, but not 4 hours, after hemolysate injection as determined by DNA laddering and TUNEL staining (P:<0.02). DNA fragmentation colocalized to cells with cytosolic cytochrome c and iron. In Sod2+/- mutants, the extent of fragmentation was increased as determined by TUNEL staining (52% increase; P:<0.02) and DNA laddering (optical density=0.819 versus 0.391; P:<0.01). Cell death was evident on basic histology as early as 4 hours after hemolysate injection. No cell death was evident in controls. In Sod2+/- mutants, cell death was increased by 51% compared with wild-type littermates (P:<0.05). CONCLUSIONS: These results demonstrate that subarachnoid blood products are associated with the presence of cytochrome c in the cytosol and subsequent cell death in neurons. It appears that Mn-superoxide dismutase plays a role in preventing cell death after exposure to subarachnoid blood products.


Assuntos
Morte Celular , Grupo dos Citocromos c/metabolismo , Fragmentação do DNA/genética , Hemorragia Subaracnóidea/metabolismo , Superóxido Dismutase/deficiência , Animais , Citosol/enzimologia , Fragmentação do DNA/efeitos dos fármacos , Modelos Animais de Doenças , Imunofluorescência , Hemoglobinas/metabolismo , Hemoglobinas/farmacologia , Heterozigoto , Immunoblotting , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Knockout , Neocórtex/efeitos dos fármacos , Neocórtex/patologia , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Neurônios/patologia , Estresse Oxidativo/genética , Hemorragia Subaracnóidea/patologia , Superóxido Dismutase/genética , Taxa de Sobrevida
4.
Brain Res ; 889(1-2): 208-13, 2001 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-11166705

RESUMO

Copper,zinc-superoxide dismutase (SOD1) was shown to be highly protective against ischemia/reperfusion injury in the brain. We have recently reported that SOD1 prevents the release of mitochondrial cytochrome c and subsequent apoptosis after ischemia/reperfusion in mice. To investigate its dose dependent effect on permanent focal cerebral ischemia, we examined neurological deficit scores, infarction volume, and the amount of hemisphere enlargement after 24 h of focal cerebral ischemia in both knockout mutants of SOD1 (Sod1 -/+ and Sod1 -/-) and wild-type littermates. We also examined the release of cytochrome c and subsequent DNA fragmentation after ischemia. There were no differences in the neurological deficit scores, infarction volumes and edema formation. There was also no difference of the amount cytosolic cytochrome c at 2 h and of the amount of DNA fragmentation at 24 h after focal cerebral ischemia. The results indicate that the SOD1 enzyme does not appear to affect cerebral infarction, cerebral edema nor the mitochondrial signaling pathway for apoptosis following permanent focal cerebral ischemia where there is no reperfusion injury.


Assuntos
Edema Encefálico/patologia , Isquemia Encefálica/patologia , Infarto Cerebral/patologia , Grupo dos Citocromos c/metabolismo , Mitocôndrias/enzimologia , Superóxido Dismutase/deficiência , Animais , Comportamento Animal/fisiologia , Western Blotting , Edema Encefálico/enzimologia , Edema Encefálico/genética , Isquemia Encefálica/enzimologia , Isquemia Encefálica/genética , Infarto Cerebral/enzimologia , Infarto Cerebral/genética , Fragmentação do DNA/efeitos dos fármacos , Heterozigoto , Homozigoto , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Knockout , Artéria Cerebral Média/fisiologia , Superóxido Dismutase/genética
5.
Brain Res ; 869(1-2): 105-11, 2000 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-10865064

RESUMO

The X-ray repair cross-complementing group 1 (XRCC1) protein plays a central role in the DNA base excision repair pathway by interacting with DNA ligase III and DNA polymerase beta. The present study examined the protein expression of XRCC1 and DNA fragmentation before and after cold injury-induced brain trauma (CIBT) in mice, in which apoptosis is assumed to participate. Immunohistochemistry showed the nuclear expression of XRCC1 in the entire region of the control brains. Fifteen minutes after CIBT, nuclear immunoreactivity was predominantly decreased in the inner boundary of the lesion, followed by a significant reduction of XRCC1 in the entire lesion 4 h after CIBT. A characteristic 70-kDa band was detected in the non-traumatic area, and was markedly decreased after CIBT as shown by Western blot analysis. DNA fragmentation was also observed after CIBT, and double staining with XRCC1 immunohistochemistry and terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate-biotin nick end labeling showed a spatial relationship between XRCC1 loss and DNA fragmentation 24 h after CIBT. These data indicate that early decrease of XRCC1 and failure of the DNA repair mechanism may contribute to DNA-damaged neuronal cell death after CIBT.


Assuntos
Apoptose/fisiologia , Lesões Encefálicas/fisiopatologia , Fragmentação do DNA/fisiologia , Reparo do DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Animais , Córtex Cerebral/lesões , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Infarto Cerebral/patologia , Infarto Cerebral/fisiopatologia , Temperatura Baixa/efeitos adversos , Edema/patologia , Edema/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos , Proteína 1 Complementadora Cruzada de Reparo de Raio-X
6.
J Cereb Blood Flow Metab ; 20(1): 130-8, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10616801

RESUMO

Matrix metalloproteinases (MMPs), a family of proteolytic enzymes which degrade the extracellular matrix, are implicated in blood-brain barrier disruption, which is a critical event leading to vasogenic edema. To investigate the role of reactive oxygen species (ROS) in the expression of MMPs in vasogenic edema, the authors measured gelatinase activities before and after cold injury (CI) using transgenic mice that overexpress superoxide dismutase-l. A marked induction of pro-gelatinase B (pro-MMP-9) was seen 2 hours after CI and was maximized at 12 hours in wild-type mice. The pro-MMP-9 level was significantly lower in transgenic mice 4 hours (P < 0.001) and 12 hours (P < 0.05) after CI compared to wild-type mice. The activated MMP-9 was detected from 6 to 24 hours after injury. A mild induction of pro-gelatinase A (pro-MMP-2) was seen at 6 hours and was sustained until 7 days. In contrast. the activated form of MMP-2 appeared at 24 hours, was maximized at 7 days, and was absent in transgenic mice. Western blot analysis showed that the tissue inhibitors of metalloproteinases were not modified after CI. The results suggest that ROS production after CI may contribute to the induction and/or activation of MMPs and could thereby exacerbate endothelial cell injury and the development of vasogenic edema after injury. Key Words: Metalloproteinases-Brain-Vasogenic edema-Reactive oxygen species-Superoxide dismutase.


Assuntos
Lesões Encefálicas/enzimologia , Temperatura Baixa , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Superóxido Dismutase/metabolismo , Animais , Ativação Enzimática , Indução Enzimática , Precursores Enzimáticos/metabolismo , Gelatinases/metabolismo , Masculino , Metaloproteinase 1 da Matriz/metabolismo , Metaloendopeptidases/metabolismo , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos/genética , Camundongos Transgênicos/metabolismo , Superóxido Dismutase/genética , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Regulação para Cima
7.
J Neurosci ; 20(8): 2817-24, 2000 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-10751433

RESUMO

Release of mitochondrial cytochrome c into the cytosol is a critical step in apoptosis. We have reported that early release of cytochrome c in vivo occurs after permanent focal cerebral ischemia (FCI) and is mediated by the mitochondrial antioxidant manganese superoxide dismutase (SOD). However, the role of reactive oxygen species produced after ischemia-reperfusion in the mitochondrial apoptosis process is still unknown, although overexpression of copper/zinc-SOD (SOD1), a cytosolic isoenzyme, protects against ischemia-reperfusion. We now hypothesize that the overexpression of SOD1 also prevents apoptosis after FCI. To address this issue, we examined the subcellular distribution of the cytochrome c protein in both wild-type mice and in SOD1 transgenic (Tg) mice after transient FCI. Cytosolic cytochrome c was detected as early as 2 hr after reperfusion, and correspondingly, mitochondrial cytochrome c was significantly reduced after FCI. Cytosolic cytochrome c was significantly lower in the SOD1 Tg mice compared with wild types 2 (p < 0.0001) and 4 (p < 0.05) hr after FCI. Apaf-1, which interacts with cytochrome c and activates caspases, was constitutively expressed in both groups of animals, with no alteration after FCI. Double staining with cytochrome c immunohistochemistry and terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate-biotin nick end labeling showed a spatial relationship between cytosolic cytochrome c expression and DNA fragmentation. A significant amount of DNA laddering was detected 24 hr after ischemia and was reduced in SOD1 Tg mice. These data suggest that SOD1 blocks cytosolic release of cytochrome c and could thereby reduce apoptosis after transient FCI.


Assuntos
Grupo dos Citocromos c/metabolismo , Ataque Isquêmico Transitório/enzimologia , Proteínas/metabolismo , Superóxido Dismutase/metabolismo , Animais , Fator Apoptótico 1 Ativador de Proteases , Western Blotting , Encéfalo/enzimologia , Citosol/enzimologia , Fragmentação do DNA/fisiologia , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Mitocôndrias/enzimologia , Prostaglandina-Endoperóxido Sintases/metabolismo , Superóxido Dismutase/imunologia , Superóxido Dismutase-1
8.
Stroke ; 30(11): 2408-15, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10548678

RESUMO

BACKGROUND AND PURPOSE: DNA damage and its repair mechanism are thought to be involved in ischemia/reperfusion injury in the brain. We have previously shown that apurinic/apyrimidinic endonuclease (APE/Ref-1), a multifunctional protein in the DNA base excision repair pathway, rapidly decreased after transient focal cerebral ischemia (FCI) before the peak of DNA fragmentation. To further investigate the role of reactive oxygen species in APE/Ref-1 expression in vivo, we examined the expression of APE/Ref-1 and DNA damage after FCI in wild-type and transgenic mice overexpressing copper-zinc superoxide dismutase. METHODS: Transgenic mice overexpressing copper-zinc superoxide dismutase and wild-type littermates were subjected to 60 minutes of transient FCI by intraluminal blockade of the middle cerebral artery. APE/Ref-1 protein expression was analyzed by immunohistochemistry and Western blot analysis. DNA damage was evaluated by gel electrophoresis and terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate-biotin nick end-labeling (TUNEL). RESULTS: A similar level of APE/Ref-1 was detected in the control brains from both groups. APE/Ref-1 was significantly reduced 1 hour after transient FCI in both groups, whereas the transgenic mice had less reduction than that seen in wild-type mice 1 and 4 hours after FCI. DNA laddering was detected 24 hours after FCI and was decreased in transgenic mice. Double staining with APE/Ref-1 and TUNEL showed that the neurons that lost APE/Ref-1 immunoreactivity became TUNEL positive. CONCLUSIONS: These results suggest that reactive oxygen species contribute to the early decrease of APE/Ref-1 and thereby exacerbate DNA fragmentation after transient FCI in mice.


Assuntos
Carbono-Oxigênio Liases/fisiologia , Fragmentação do DNA/fisiologia , Ataque Isquêmico Transitório/enzimologia , Superóxido Dismutase/fisiologia , Animais , Western Blotting , Corantes , Dano ao DNA/genética , Dano ao DNA/fisiologia , Fragmentação do DNA/genética , Reparo do DNA/genética , Reparo do DNA/fisiologia , DNA Liase (Sítios Apurínicos ou Apirimidínicos) , Desoxirribonuclease IV (Fago T4-Induzido) , Eletroforese em Gel de Ágar , Regulação Enzimológica da Expressão Gênica , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Transgênicos , Artéria Cerebral Média/fisiopatologia , Neurônios/enzimologia , Neurônios/patologia , Espécies Reativas de Oxigênio/fisiologia , Superóxido Dismutase/genética , Fatores de Tempo
9.
Stroke ; 30(11): 2456-62; discussion 2463, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10548684

RESUMO

BACKGROUND AND PURPOSE: DNA damage and the DNA repair mechanism are known to be involved in ischemia/reperfusion injury in the brain. The x-ray repair cross-complementing group 1 (XRCC1) protein plays a central role in the DNA base excision repair pathway by interacting with DNA ligase III and DNA polymerase beta. The present study examined the protein expression of XRCC1 and DNA fragmentation before and after transient focal cerebral ischemia (FCI). METHODS: Adult male CD-1 mice were subjected to 60 minutes of FCI by intraluminal blockade of the middle cerebral artery. XRCC1 protein expression was analyzed by immunohistochemistry and Western blot analysis. DNA damage was evaluated by gel electrophoresis and terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate-biotin nick end-labeling (TUNEL). The spatial relationship between XRCC1 expression and DNA damage was examined by double staining with XRCC1 and TUNEL after FCI. RESULTS: Immunohistochemistry showed the nuclear expression of XRCC1 in all regions of the control brains and that it was predominant in the hippocampus. The XRCC1 level was markedly reduced in the caudate putamen at 10 minutes, further decreased in the entire middle cerebral artery territory at 1 hour, and remained reduced until 4 and 24 hours after FCI. Western blot analysis of the normal control brain showed a characteristic band of 70 kDa, which decreased after FCI. A significant amount of DNA fragmentation was detected by DNA gel electrophoresis 24 hours but not 4 hours after FCI. Double staining showed that the neurons that lost XRCC1 immunoreactivity became TUNEL positive. CONCLUSIONS: These results suggest that the early decrease of XRCC1 and the failure of the DNA repair mechanism may contribute, at least in part, to DNA fragmentation after FCI.


Assuntos
Fragmentação do DNA/genética , Reparo do DNA/genética , Proteínas de Ligação a DNA/genética , Ataque Isquêmico Transitório/genética , Animais , Western Blotting , Corantes , Dano ao DNA/genética , DNA Ligase Dependente de ATP , DNA Ligases/genética , DNA Polimerase beta/genética , Eletroforese em Gel de Ágar , Regulação da Expressão Gênica , Hipocampo/metabolismo , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Infarto da Artéria Cerebral Média/genética , Masculino , Camundongos , Camundongos Endogâmicos , Artéria Cerebral Média/fisiopatologia , Proteínas de Ligação a Poli-ADP-Ribose , Putamen/metabolismo , Traumatismo por Reperfusão/genética , Proteína 1 Complementadora Cruzada de Reparo de Raio-X , Proteínas de Xenopus
10.
J Neurosci ; 19(22): RC39, 1999 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-10559429

RESUMO

Release of cytochrome c from mitochondria to the cytosol is a critical step in apoptotic cell death after focal cerebral ischemia. The relationship among cytochrome c release, selective vulnerability, and delayed death of hippocampal CA1 neurons after transient global ischemia was examined. Global ischemia was induced by 10 min of bilateral common carotid artery occlusion and hypotension in rats. Cytosolic expression of cytochrome c was evaluated by immunohistochemistry and Western blotting. Apoptosis after global ischemia was also characterized by terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate-biotin nick end-labeling (TUNEL) staining and DNA gel electrophoresis. Immunohistochemistry showed cytosolic cytochrome c-positive cells exclusively in the CA1 subregion of the hippocampus as early as 2 hr after ischemia. Double fluorescent immunostaining confirmed that CA1 neurons and a small number of astrocytes expressed cytochrome c. Western blot analysis revealed a band (15 kDa) of cytochrome c in the cytosolic fraction and a corresponding decrease in the mitochondrial fraction. A significant number of TUNEL-positive cells appeared only in the CA1 pyramidal cell layer of the hippocampus, and DNA gel electrophoresis showed a significant amount of DNA fragmentation 3-5 d after ischemia. Our data provide the first evidence that cytochrome c was released to the cytosol from mitochondria in CA1 neurons after global ischemia and that the release preceded DNA fragmentation. These findings suggest cytochrome c involvement in the delayed death of hippocampal CA1 neurons in rats after transient global ischemia.


Assuntos
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Grupo dos Citocromos c/metabolismo , Hipocampo/metabolismo , Hipocampo/patologia , Mitocôndrias/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Animais , Western Blotting , Fragmentação do DNA , Feminino , Imuno-Histoquímica , Ratos , Ratos Sprague-Dawley
11.
Brain Res ; 842(1): 92-100, 1999 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-10526099

RESUMO

Blood-brain barrier (BBB) disruption is thought to play a critical role in the pathophysiology of ischemia/reperfusion. Matrix metalloproteinases (MMPs) are a family of proteolytic enzymes that can degrade all the components of the extracellular matrix when they are activated. Gelatinase A (MMP-2) and gelatinase B (MMP-9) are able to digest the endothelial basal lamina, which plays a major role in maintaining BBB impermeability. The present study examined the expression and activation of gelatinases before and after transient focal cerebral ischemia (FCI) in mice. Adult male CD1 mice were subjected to 60 min FCI and reperfusion. Zymography was performed from 1 to 23 h after reperfusion using the protein extraction method with detergent extraction and affinity-support purification. MMP-9 expression was also examined by both immunohistochemistry and Western blot analysis, and tissue inhibitors to metalloproteinase-1 was measured by reverse zymography. The BBB opening was evaluated by the Evans blue extravasation method. The 88-kDa activated MMP-9 was absent from the control specimens, while it appeared 3 h after transient ischemia by zymography. At this time point, the BBB permeability alteration was detected in the ischemic brain. Both pro-MMP-9 (96 kDa) and pro-MMP-2 (72 kDa) were seen in the control specimens, and were markedly increased after FCI. A significant induction of MMP-9 was confirmed by both immunohistochemistry and Western blot analysis. The early appearance of activated MMP-9, associated with evidence of BBB permeability alteration, suggests that activation of MMP-9 contributes to the early formation of vasogenic edema after transient FCI.


Assuntos
Barreira Hematoencefálica/fisiologia , Ataque Isquêmico Transitório/enzimologia , Ataque Isquêmico Transitório/patologia , Metaloproteinase 9 da Matriz/metabolismo , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/patologia , Animais , Compostos de Benzil , Western Blotting , Colágeno/metabolismo , Colagenases/metabolismo , Corantes , Dexametasona/farmacologia , Combinação de Medicamentos , Inibidores Enzimáticos/farmacologia , Azul Evans , Gelatinases/antagonistas & inibidores , Gelatinases/metabolismo , Imuno-Histoquímica , Infarto da Artéria Cerebral Média/patologia , Masculino , Inibidores de Metaloproteinases de Matriz , Camundongos , Pentoxifilina/farmacologia , Succinatos , Inibidor Tecidual de Metaloproteinase-1/biossíntese
12.
Neuroscience ; 93(4): 1465-73, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10501471

RESUMO

Apurinic/apyrimidinic endonuclease, a multifunctional protein in the DNA base excision repair pathway, plays a central role in repairing DNA damage caused by reactive oxygen species. We examined protein expression of apurinic/apyrimidinic endonuclease before and after cold injury-induced brain trauma in mice, where we have previously shown reactive oxygen species to participate. Immunohistochemistry showed the nuclear expression of apurinic/apyrimidinic endonuclease in the entire region of control brains. One hour after cold injury-induced brain trauma, nuclear immunoreactivity was predominantly decreased in the inner boundary of the lesion, whereas there was a slight increase in the outer boundary area. Four hours after cold injury-induced brain trauma, nuclear immunoreactivity was almost absent in the entire lesion, and remained so until 24 h. At this time, a marked increase in apurinic/apyrimidinic endonuclease immunoreactivity was seen in the outer boundary zone. Western blot analysis of the sample from the non-ischemic area showed a characteristic 37,000 mol. wt band, which decreased markedly 24 h after cold injury-induced brain trauma. A time-dependent increase in DNA fragmentation was also observed after cold injury-induced brain trauma. Our data provide the first evidence that apurinic/apyrimidinic endonuclease decreased rapidly in the lesion after cold injury-induced brain trauma, whereas it was significantly increased at the outer boundary zone. Although further examination is necessary to elucidate the direct relationship between apurinic/apyrimidinic endonuclease alteration and the pathogenesis of cold injury-induced brain trauma, our results suggest the possibility that an early decrease in apurinic/apyrimidinic endonuclease and failure of the DNA repair mechanism may contribute to DNA-damaged neuronal cell death after cold injury-induced brain trauma.


Assuntos
Apoptose/fisiologia , Lesões Encefálicas/enzimologia , Carbono-Oxigênio Liases/metabolismo , Temperatura Baixa/efeitos adversos , Fragmentação do DNA , Animais , Western Blotting , Edema Encefálico/enzimologia , Carbono-Oxigênio Liases/análise , Carbono-Oxigênio Liases/biossíntese , Infarto Cerebral/enzimologia , Circulação Cerebrovascular , Reparo do DNA , DNA Liase (Sítios Apurínicos ou Apirimidínicos) , Desoxirribonuclease IV (Fago T4-Induzido) , Eletroforese , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Endogâmicos , Proteínas do Tecido Nervoso/análise , Neurônios/química , Neurônios/citologia , Neurônios/enzimologia
13.
J Cereb Blood Flow Metab ; 19(9): 1020-8, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10478654

RESUMO

During cerebral ischemia blood-brain barrier (BBB) disruption is a critical event leading to vasogenic edema and secondary brain injury. Gelatinases A and B are matrix metalloproteinases (MMP) able to open the BBB. The current study analyzes by zymography the early gelatinases expression and activation during permanent ischemia in mice (n = 15). ProMMP-9 expression was significantly (P < 0.001) increased in ischemic regions compared with corresponding contralateral regions after 2 hours of ischemia (mean 694.7 arbitrary units [AU], SD +/- 238.4 versus mean 107.6 AU, SD +/- 15.6) and remained elevated until 24 hours (mean 745.7 AU, SD +/- 157.4). Moreover, activated MMP-9 was observed 4 hours after the initiation of ischemia. At the same time as the appearance of activated MMP-9, we detected by the Evan's blue extravasation method a clear increase of BBB permeability. Tissue inhibitor of metalloproteinase-1 was not modified during permanent ischemia at any time. The ProMMP-2 was significantly (P < 0.05) increased only after 24 hours of permanent ischemia (mean 213.2 AU, SD +/- 60.6 versus mean 94.6 AU, SD +/- 13.3), and no activated form was observed. The appearance of activated MMP-9 after 4 hours of ischemia in correlation with BBB permeability alterations suggests that MMP-9 may play an active role in early vasogenic edema development after stroke.


Assuntos
Barreira Hematoencefálica , Isquemia Encefálica/enzimologia , Colagenases/metabolismo , Animais , Ativação Enzimática , Imuno-Histoquímica , Masculino , Metaloproteinase 9 da Matriz , Camundongos
14.
Stroke ; 30(9): 1962-8, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10471451

RESUMO

BACKGROUND AND PURPOSE: We have demonstrated that copper-zinc superoxide dismutase (CuZn-SOD), a cytosolic isoenzyme of SODs, has a protective role in the pathogenesis of superoxide radical-mediated brain injury. Using mice bearing a disruption of the CuZn-SOD gene (Sod1), the present study was designed to clarify the role of superoxide anion in the pathogenesis of selective vulnerability after transient global ischemia. METHODS: Sod1 knockout homozygous mutant mice (Sod1 -/-) with a complete absence of endogenous CuZn-SOD activity, heterozygous mutant mice (Sod1 +/-) with a 50% decrease in the activity, and littermate wild-type mice (male, 35 to 45 g) were subjected to global ischemia. Since the plasticity of the posterior communicating artery (PcomA) has been reported to influence the outcome of hippocampal injury, we assessed the relation between the plasticity of PcomAs and the decrease of regional cerebral blood flow in global ischemia. RESULTS: The fluorescence intensity of hydroethidine oxidation, a measurement of ethidium fluorescence for superoxide radicals, was increased in mutant mice 1 day after both 5 and 10 minutes of global ischemia, compared with wild-type mice. Hippocampal injury in the PcomA hypoplastic brains showed significant exacerbation in mutant mice compared with wild-type littermates 3 days after 5 minutes of global ischemia, although a marked difference was not observed at 1 day. CONCLUSIONS: These data suggest that superoxide radicals play an important role in the pathogenesis of delayed injury in the vulnerable hippocampal CA1 subregion after transient global ischemia.


Assuntos
Encéfalo/patologia , Ataque Isquêmico Transitório/patologia , Superóxido Dismutase/fisiologia , Animais , Morte Celular , Hipocampo/patologia , Ataque Isquêmico Transitório/metabolismo , Ataque Isquêmico Transitório/mortalidade , Camundongos , Camundongos Knockout/genética , Superóxido Dismutase/deficiência , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Superóxidos/metabolismo , Fatores de Tempo
15.
Neurosci Lett ; 267(3): 201-5, 1999 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-10381011

RESUMO

Recent studies have shown that release of mitochondrial cytochrome c is a critical step in the apoptosis process. In this study, we examined the subcellular distribution of the cytochrome c protein after cold injury (CI), in which apoptosis is assumed to participate. Western blotting and immunohistochemistry showed cytosolic cytochrome c as early as 1 h after CI, and correspondingly, there was a reduction in mitochondrial cytochrome c after injury. Neuronal distribution of cytosolic cytochrome c was shown by double staining with a neuronal nuclear marker by immunohistochemistry. A significant amount of DNA laddering was detected 4 h after CI, and increased in a time-dependent manner. These data suggest that early cytochrome c release from mitochondria may contribute to apoptosis induction after traumatic brain injury.


Assuntos
Edema Encefálico/fisiopatologia , Grupo dos Citocromos c/metabolismo , Citosol/metabolismo , Fragmentação do DNA/fisiologia , Mitocôndrias/metabolismo , Animais , Apoptose/fisiologia , Temperatura Baixa , Masculino , Camundongos
16.
J Cereb Blood Flow Metab ; 19(6): 634-42, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10366193

RESUMO

The authors examined the effect of z-VAD.FMK, an inhibitor that blocks caspase family proteases, on cold injury-induced brain trauma, in which apoptosis as well as necrosis is assumed to play a role. A vehicle alone or with z-VAD.FMK was administered into the cerebral ventricles of mice 15 minutes before and 24 and 48 hours after cold injury. At 24 hours after cold injury, infarction volumes in the z-VAD.FMK-treated animals were significantly smaller than infarction volumes in the vehicle-treated animals, and were further decreased at 72 hours (0.92 +/- 1.80 mm3, z-VAD.FMK-treated animals; 7.46 +/- 3.53 mm3, vehicle-treated animals; mean +/- SD, n = 7 to 8). The amount of DNA fragmentation was significantly decreased in the z-VAD.FMK-treated animals compared with the vehicle-treated animals, as shown by terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate-biotin nick end labeling staining and DNA gel electrophoresis. By Western blot analysis, both the proform and activated form of interleukin-1beta converting enzyme (caspase 1) were detected in the control brain, and the activated form showed moderate reduction after cold injury-induced brain trauma. These results indicate that caspase inhibitors could reduce cold injury-induced brain trauma by preventing neuronal cell death by DNA damage. The caspase family proteases appear to contribute to the mechanisms of cell death in cold injury-induced brain trauma and to provide therapeutic targets for traumatic brain injury.


Assuntos
Clorometilcetonas de Aminoácidos/farmacologia , Lesões Encefálicas/tratamento farmacológico , Inibidores de Caspase , Temperatura Baixa/efeitos adversos , Inibidores de Cisteína Proteinase/farmacologia , Fragmentação do DNA/efeitos dos fármacos , Animais , Western Blotting , Encéfalo/patologia , Lesões Encefálicas/enzimologia , Lesões Encefálicas/patologia , Eletroforese em Gel de Poliacrilamida , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos
17.
J Cereb Blood Flow Metab ; 19(5): 495-501, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10326716

RESUMO

The authors examined the protein expression of apurinic/apyrimidinic endonuclease (APE/Ref-1), a multifunctional protein in the DNA base excision repair pathway, before and after transient focal ischemia in mice. Immunohistochemistry showed the nuclear expression of APE/Ref-1 in the entire region of the control brains. Nuclear immunoreactivity was decreased as early as 5 minutes after 60 minutes of ischemia in the ischemic core, which was followed by a significant reduction of APE/Ref-1-positive cells in the entire middle cerebral artery territory. Western blot analysis of the sample from the nonischemic brain showed a characteristic 37-kDa band, which was reduced after ischemia. A significant amount of DNA fragmentation was observed at 24 hours, but not at 4 hours, after ischemia. The authors' data provide the first evidence that APE/Ref-1 rapidly decreases after transient focal ischemia, and that this reduction precedes the peak of DNA fragmentation in the brain regions that are destined to show necrosis and apoptosis. Although further examination is necessary to elucidate the direct relationship between the APE/Ref-1 decrease and ischemic necrosis and apoptosis, our results suggest the possibility that rapid decrease of APE/Ref-1 and the failure of the DNA repair mechanism may contribute to necrosis or apoptosis after transient focal ischemia.


Assuntos
Carbono-Oxigênio Liases/biossíntese , Reparo do DNA , Ataque Isquêmico Transitório/metabolismo , Animais , Western Blotting , Morte Celular , Infarto Cerebral/metabolismo , Fragmentação do DNA , DNA Liase (Sítios Apurínicos ou Apirimidínicos) , Desoxirribonuclease IV (Fago T4-Induzido) , Genoma , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos , Neurônios/metabolismo , Neurônios/patologia , Reperfusão
18.
J Neurosci ; 19(9): 3414-22, 1999 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10212301

RESUMO

Recent studies have shown that release of mitochondrial cytochrome c is a critical step in the apoptosis process. We have reported that cytosolic redistribution of cytochrome c in vivo occurred after transient focal cerebral ischemia (FCI) in rats and preceded the peak of DNA fragmentation. Although the involvement of reactive oxygen species in the cytosolic redistribution of cytochrome c in vitro has been suggested, the detailed mechanism by which cytochrome c release is mediated in vivo has not yet been established. Also, the role of mitochondrial oxidative stress in cytochrome c release is unknown. These issues can be addressed using knock-out mutants that are deficient in the level of the mitochondrial antioxidant manganese superoxide dismutase (Mn-SOD). In this study we examined the subcellular distribution of the cytochrome c protein in both wild-type mice and heterozygous knock-outs of the Mn-SOD gene (Sod2 -/+) after permanent FCI, in which apoptosis is assumed to participate. Cytosolic cytochrome c was detected as early as 1 hr after ischemia, and correspondingly, mitochondrial cytochrome c showed a significant reduction 2 hr after ischemia (p < 0.01). Cytosolic accumulation of cytochrome c was significantly higher in Sod2 -/+ mice compared with wild-type animals (p < 0.05). N-benzyloxycarbonyl-val-ala-asp-fluoromethyl ketone (z-VAD.FMK), a nonselective caspase inhibitor, did not affect cytochrome c release after ischemia. A significant amount of DNA laddering was detected 24 hr after ischemia and increased in Sod2 -/+ mice. These data suggest that Mn-SOD blocks cytosolic release of cytochrome c and could thereby reduce apoptosis after permanent FCI.


Assuntos
Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Grupo dos Citocromos c/metabolismo , Fragmentação do DNA , Ataque Isquêmico Transitório/metabolismo , Mitocôndrias/metabolismo , Superóxido Dismutase/metabolismo , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Apoptose , Pressão Sanguínea , Isquemia Encefálica/genética , Isquemia Encefálica/fisiopatologia , Cardiomiopatia Dilatada/genética , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Infarto Cerebral/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Citosol/metabolismo , Heterozigoto , Ataque Isquêmico Transitório/genética , Ataque Isquêmico Transitório/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Estresse Oxidativo , Ratos , Superóxido Dismutase/deficiência , Superóxido Dismutase/genética , Superóxidos/metabolismo
19.
Prog Neurobiol ; 57(3): 289-99, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10096842

RESUMO

Small rodents, mice in particular, have been widely used for genetic manipulation because of the extensive knowledge in development, embryology and other molecular aspects of this species. However, the use of mice for neurobiology research in the area of brain edema and neuronal injury has not been common. Here we summarize the studies of cold injury-induced brain edema and neuronal apoptosis using mice. Blood-brain barrier (BBB) permeability, demonstrated by extravasation of a serum albumin tracer, Evans Blue, was increased immediately after the injury and returned to the control level by 24 hr. Water content was maximized at 24 hr, whereas a secondary lesion gradually progressed up to 72 hr after cold injury. The mechanism of the development of the cold injury-induced edema and the secondary lesion, involving of oxygen radicals in particular, was determined using superoxide dismutase (SOD)-1 transgenic (Tg) mice with overexpressed copper, zinc-SOD. All of the parameters, BBB permeability, water content and secondary lesion, were attenuated in the Tg mice as compared to littermate non-Tg mice. This clearly demonstrates that oxygen radicals, superoxide anion in particular, mediate cold injury. We also studied whether apoptosis contributes to brain injury following cold injury. Staining with terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate-biotin nick end labeling showed the apoptotic cells widespread throughout the entire lesion while still remaining in the margin. DNA laddering was exhibited by gel electrophoresis. These studies indicate that oxidative mediates the development of cold injury-induced edema and the secondary injury, and induces apoptotic cell death. We believe that cold injury in mice provides a simple animal model to study the pathogenesis of brain edema and apoptosis in genetically altered animals.


Assuntos
Apoptose/fisiologia , Edema Encefálico/etiologia , Edema Encefálico/fisiopatologia , Lesões Encefálicas/complicações , Temperatura Baixa , Neurônios/fisiologia , Animais , Edema Encefálico/patologia , Camundongos
20.
Stroke ; 30(2): 441-8; discussion 449, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9933285

RESUMO

BACKGROUND AND PURPOSE: To clarify the relationship between apurinic/apyrimidinic endonuclease (APE/Ref-1), a multifunctional protein in the DNA base excision repair pathway, and delayed neuronal cell death associated with apoptosis, we examined the expression of APE/Ref-1 before and after transient global ischemia in rats. METHODS: Global ischemia was induced by bilateral common carotid artery occlusion and hypotension. Expression of the APE/Ref-1 protein was evaluated by Western blot and immunohistochemical analyses. Apoptosis after global ischemia was observed by DNA electrophoresis and terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate-biotin nick end labeling (TUNEL) staining. RESULTS: Immunohistochemistry showed the nuclear expression of APE/Ref-1 in the control brains. Nuclear immunoreactivity of APE/Ref-1 was significantly decreased 2 days after 10 minutes of ischemia in the hippocampal CA1 subregion. Western blot analysis of a sample from the normal brains showed a characteristic 37-kDa band, which was reduced in the hippocampal CA1 subregion after ischemia. A significant amount of DNA fragmentation was observed at 3 days but not at 1 day after ischemia. Double staining with APE/Ref-1 and TUNEL clearly showed that the neurons that lost APE/Ref-1 immunoreactivity became TUNEL positive. CONCLUSIONS: Our data provide evidence that APE/Ref-1 decreased in hippocampal CA1 neurons after transient global ischemia and that this reduction precedes DNA fragmentation, which is destined to cause apoptosis. Our results suggest the possibility that a decrease of APE/Ref-1 activity and the failure of DNA repair may underlie the mechanism of apoptosis after transient focal ischemia.


Assuntos
Apoptose , Carbono-Oxigênio Liases/metabolismo , Reparo do DNA , DNA Liase (Sítios Apurínicos ou Apirimidínicos) , Endodesoxirribonucleases/metabolismo , Ataque Isquêmico Transitório/enzimologia , Animais , Western Blotting , Carbono-Oxigênio Liases/genética , Sondas de DNA/química , Eletroforese em Gel de Ágar , Endodesoxirribonucleases/genética , Hipocampo/irrigação sanguínea , Hipocampo/enzimologia , Marcação In Situ das Extremidades Cortadas , Ataque Isquêmico Transitório/patologia , Masculino , Neurônios/enzimologia , Neurônios/patologia , RNA Mensageiro/biossíntese , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...