Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
1.
Acta Neurochir Suppl ; 106: 147-50, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19812938

RESUMO

Evidence suggests that microglia activation contributes to brain injury after intracerebral hemorrhage (ICH). The present study aimed to determine if minocycline, an inhibitor of microglia activation, can reduce brain edema, brain atrophy and neurological deficits after ICH.Male Sprague-Dawley rats received an infusion of 100-microL autologous whole blood into the right basal ganglia. Rats received minocycline or vehicle treatment. There were two sets of experiments in this study. In the first set of experiments, the effects of minocycline on ICH-induced brain edema were examined at day 3. In the second set, behavioral tests were performed at days 1, 3, 7, 14 and 28. Rats were killed at day 28 for brain atrophy measurement (caudate and lateral ventricle size).Minocycline reduced perihematomal brain edema in the ipsilateral basal ganglia (78.8 +/- 0.4 vs. 80.9 +/- 1.1% in the vehicle-treated group, p < 0.01). Minocycline also improved functional outcome. In addition, minocycline reduced brain tissue loss in the ipsilateral caudate (p < 0.01) and ventricular enlargement (p < 0.05).In conclusion, minocycline attenuates ICH-induced brain edema formation, neurological deficits and brain atrophy in rats suggesting an important role of microglia in ICH-related brain injury.


Assuntos
Atrofia/etiologia , Edema Encefálico/tratamento farmacológico , Edema Encefálico/etiologia , Hemorragia Cerebral/complicações , Minociclina/uso terapêutico , Doenças do Sistema Nervoso/tratamento farmacológico , Doenças do Sistema Nervoso/etiologia , Análise de Variância , Animais , Atrofia/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Núcleo Caudado/patologia , Modelos Animais de Doenças , Ventrículos Laterais/patologia , Masculino , Minociclina/farmacologia , Exame Neurológico , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
2.
Acta Neurochir Suppl ; 106: 159-62, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19812941

RESUMO

Our previous studies found that 17-beta estradiol attenuates edema formation after intracerebral hemorrhage (ICH). As brain iron overload occurs after ICH and contributes to ICH-induced brain injury, the present study examined the effects of estrogen on iron-induced brain injury in vivo and in vitro.There were two sets of experiments in this study. In the first set, male Sprague-Dawley rats were pretreated with 17-beta estradiol or vehicle prior to an intracerebral injection of ferrous iron. Ferrous iron was injected into the right caudate and the rats were killed 24 h later for brain edema measurement. In the second set, primary cultured neurons were pretreated with different doses of 17-beta estradiol or vehicle for 24 h. The cells were then exposed to ferrous iron for 48 h when culture medium was collected for lactate dehydrogenase measurement. Neuronal death was also assessed by live/dead cell assay.Estrogen pretreatment reduced brain water content (p < 0.01) 24 h after iron injection. Estrogen also protected against iron-induced cell death in cultured neurons. Estrogen reduces iron-induced brain edema in vivo and neuronal death in vitro suggesting estrogen could be a potential therapeutic agent for ICH.


Assuntos
Edema Encefálico/prevenção & controle , Estradiol/farmacologia , Estrogênios/farmacologia , Ferro/farmacologia , Neurônios/efeitos dos fármacos , Animais , Edema Encefálico/etiologia , Morte Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Hemorragia Cerebral/complicações , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Interações Medicamentosas , Estradiol/uso terapêutico , Estrogênios/uso terapêutico , Lateralidade Funcional , L-Lactato Desidrogenase/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley
3.
Acta Neurochir Suppl ; 105: 3-6, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19066072

RESUMO

Intracerebral hemorrhage (ICH) is a subtype of stroke with very high mortality. Experiments have indicated that clot lysis and iron play an important role in ICH-induced brain injury. Iron overload occurs in the brain after ICH in rats. Intracerebral infusion of iron causes brain edema and neuronal death. Deferoxamine, an iron chelator, is an FDA-approved drug for the treatment of acute iron intoxication and chronic iron overload due to transfusion-dependent anemia. Deferoxamine can rapidly penetrate the blood-brain barrier and accumulate in the brain tissue in significant concentration after systemic administration. We have demonstrated that deferoxamine reduces ICH-induced brain edema, neuronal death, brain atrophy, and neurological deficits. Iron chelation with deferoxamine could be a new therapy for ICH.


Assuntos
Hemorragia Cerebral/tratamento farmacológico , Desferroxamina/uso terapêutico , Sideróforos/uso terapêutico , Animais , Edema Encefálico/etiologia , Edema Encefálico/patologia , Edema Encefálico/prevenção & controle , Morte Celular , Hemorragia Cerebral/complicações , Hemorragia Cerebral/patologia , Humanos , Neurônios/efeitos dos fármacos , Neurônios/patologia
4.
Acta Neurochir Suppl ; 105: 13-8, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19066074

RESUMO

In this study, we examine the effects of deferoxamine on hemoglobin-induced brain swelling in a newly developed hippocampal model of intracerebral hemorrhage (ICH). There were 2 parts to the experiments in this study. In the first part, male Sprague-Dawley rats received a 10-microL infusion of either packed red blood cells (RBC), lysed RBC, hemoglobin, ferrous iron, or saline, into the hippocampus. In the second part, rats received a 10-microL infusion of hemoglobin and then were treated with either deferoxamine (100 mg/kg, intraperitoneally, given immediately after hemoglobin injection, then every 12h for 24h) or vehicle. Rats were then killed to obtain hippocampus size and DNA damage measurements. We found that lysed RBC induced marked brain swelling in the hippocampus. Compared to saline, hemoglobin or iron injection caused swelling. Systemic use of deferoxamine reduced hemoglobin-induced brain swelling (6.14 +/- 0.45 vs. 7.11 +/- 0.58 mm2 in the vehicle group, p < 0.05). In addition, deferoxamine reduced hemoglobin-induced DNA damage. These results indicate that iron has a key role in hemoglobin-induced brain swelling. Deferoxamine may be a useful treatment for ICH patients.


Assuntos
Edema Encefálico , Hemorragia Cerebral/complicações , Desferroxamina/uso terapêutico , Hipocampo/patologia , Sideróforos/uso terapêutico , Análise de Variância , Animais , Edema Encefálico/tratamento farmacológico , Edema Encefálico/etiologia , Edema Encefálico/patologia , Morte Celular , Hemorragia Cerebral/induzido quimicamente , Hemorragia Cerebral/patologia , Modelos Animais de Doenças , Lateralidade Funcional , Hemoglobinas/efeitos adversos , Hipocampo/efeitos dos fármacos , Marcação In Situ das Extremidades Cortadas/métodos , Ferro/administração & dosagem , Masculino , Ratos , Ratos Sprague-Dawley
5.
Acta Neurochir Suppl ; 105: 29-32, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19066077

RESUMO

Autophagy occurs in the brain after intracerebral hemorrhage (ICH). Iron is an important factor causing neuronal death and brain atrophy after ICH. In this study, we examined whether iron can induce autophagy in the hippocampus and in cultured neurons. For in vivo studies, rats received an infusion of either saline or ferrous iron into the right hippocampus and were killed 1, 3, or 7 days later for Western blot analysis of microtubule-associated protein light chain-3 (LC3). For in vitro studies, primary cultured cortex neurons from rat embryos were exposed to ferrous iron. Cells were used for Western blot analysis of LC3 and monodansylcadaverine (MDC) staining 24h later. Intrahippocampal injection of ferrous iron resulted in an increased conversion of LC3-I to LC3-II. Exposure of primary cultured neurons to ferrous iron also induced an enhanced conversion of LC3-I to LC3-II. MDC labeling showed an accumulation of MDC in cultured neurons exposed to ferrous iron. These results indicate that autophagy is induced by iron in neurons and that iron-induced autophagy may contribute to brain injury after ICH.


Assuntos
Autofagia/efeitos dos fármacos , Compostos Ferrosos/farmacologia , Hipocampo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Análise de Variância , Animais , Cadaverina/análogos & derivados , Cadaverina/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Embrião de Mamíferos , Hipocampo/metabolismo , Masculino , Proteínas Associadas aos Microtúbulos/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
6.
Acta Neurochir Suppl ; 105: 37-40, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19066079

RESUMO

Metallothioneins (MTs) are metal-binding proteins that can be upregulated in the brain after injury and are associated with neuroprotection. A recent genomics study has shown that brain MT-1 and MT-2 mRNA levels are upregulated following intracerebral hemorrhage (ICH) in rats. Our study examines whether brain MT-1 and MT-2 protein levels are increased after ICH. We also investigated the effect of exogenous MT-1 in perihematomal edema formation in vivo and iron-induced cell death in vitro. We found that MT-1/-2 immunoreactivity in ipsilateral basal ganglia was significantly increased after ICH and exogenous MT-1 attenuated perihematomal edema formation. In addition, MT-1 also reduced cell death induced by iron in cultured astrocytes. These results suggest a role for MT in ICH-induced brain injury, and MT could be a therapeutic target for ICH.


Assuntos
Lesões Encefálicas/etiologia , Hemorragia Cerebral/complicações , Hemorragia Cerebral/metabolismo , Metalotioneína/metabolismo , Regulação para Cima/fisiologia , Análise de Variância , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Gânglios da Base/metabolismo , Gânglios da Base/patologia , Células Cultivadas , Córtex Cerebral , Hemorragia Cerebral/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , L-Lactato Desidrogenase/metabolismo , Masculino , Metalotioneína/farmacologia , Ratos , Ratos Sprague-Dawley , Regulação para Cima/efeitos dos fármacos
7.
Acta Neurochir Suppl ; 105: 47-50, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19066081

RESUMO

Thrombin has been shown to play a major role in brain injury after intracerebral hemorrhage (ICH). In this study, we measured thrombin activity in the perihematomal zone and examined the role of thrombin in ICH-induced brain tissue loss. There were 2 experiments in this study. In the first part, adult male Sprague-Dawley rats received 100 microL of either autologous whole blood or saline. The rats were killed at 1 h or 24 h later for thrombin activity measurement. Thrombin activity was measured using the thrombin-specific chromogenic substrate, S2238. In the second part, rats received a 50-microL intracaudate injection of either thrombin or saline, and the rats were killed at days 1, 3, or 28 for determination of neuronal death and brain tissue loss. We found that brain thrombin activity was elevated in ipsilateral basal ganglia 1 h after ICH. Intracerebral injection of thrombin rather than saline caused significant neuronal death at days 1 and 3, and resulted in significant brain tissue loss at day 28. These results suggest that thrombin inhibition in the acute phase may reduce ICH-induced brain damage.


Assuntos
Encéfalo/metabolismo , Hemorragia Cerebral/patologia , Regulação da Expressão Gênica/fisiologia , Trombina/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Hemorragia Cerebral/metabolismo , Dipeptídeos/metabolismo , Modelos Animais de Doenças , Fluoresceínas , Lateralidade Funcional , Regulação da Expressão Gênica/efeitos dos fármacos , Hematoma/metabolismo , Masculino , Compostos Orgânicos , Ratos , Ratos Sprague-Dawley , Trombina/farmacologia , Fatores de Tempo
8.
Acta Neurochir Suppl ; 105: 59-65, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19066084

RESUMO

Microglial activation and thrombin formation contribute to brain injury after intracerebral hemorrhage (ICH). Tumor necrosis factor-alpha (TNF-alpha) and interleukin-1 beta (IL-1beta) are 2 major proinflammatory cytokines. In this study, we investigated whether thrombin stimulates TNF-alpha and IL-1beta secretion in vitro, and whether microglial inhibition reduces ICH-induced brain injury in vivo. There were 2 parts to this study. In the first part, cultured rat microglial cells were treated with vehicle, thrombin (5 and 10U/mL), or thrombin plus tuftsin (0.05 microg/mL), an inhibitor of microglia activation. Levels of TNF-alpha and IL-1beta in culture medium were measured by ELISA at 4, 8, and 24 h after thrombin treatment. In the second part of the study, rats received an intracerebral infusion of 100 microL autologous whole blood with or without 25 microg of tuftsin 1-3 fragment. Rats were killed at day 1 or day 3 for immunohistochemistry and brain water content measurement. We found that thrombin receptors were expressed in cultured microglia cells, and TNF-alpha and IL-1beta levels in the culture medium were increased after thrombin treatment. Tuftsin reduced thrombin-induced upregulation of TNF-alpha and IL-1beta. In vivo, microglia were activated after ICH, and intracerebral injection of tuftsin reduced brain edema in the ipsilateral basal ganglia (81.1 +/- 0.7% vs. 82.7 +/- 1.3% in vehicle-treated group; p < 0.05) after ICH. These results suggest a critical role of microglia activation in ICH-related brain injury.


Assuntos
Lesões Encefálicas/etiologia , Lesões Encefálicas/patologia , Hemorragia Cerebral/complicações , Microglia/fisiologia , Animais , Animais Recém-Nascidos , Edema Encefálico/metabolismo , Edema Encefálico/patologia , Antígeno CD11b/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Modelos Animais de Doenças , Fatores Imunológicos/farmacologia , Interleucina-1beta/metabolismo , Íons/metabolismo , Masculino , Microglia/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Trombina/farmacologia , Fatores de Tempo , Tuftsina/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima/efeitos dos fármacos
9.
Acta Neurochir Suppl ; 105: 67-70, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19066085

RESUMO

Intracerebral hemorrhage (ICH)-induced brain edema and neurological deficits are greater in aged rats than in young rats. Complement activation and neutrophil infiltration contribute to brain injury after ICH. In this study, we investigated the effects of aging on activation of the complement cascade and neutrophil influx following ICH. Male Sprague-Dawley rats (3 or 18 months old) received an infusion of 100 microL autologous blood into right caudate. Rats were killed at 1, 3, 7, and 28 days after ICH and the brains were sampled for immunohistochemistry and Western blot analysis. Levels of complement factor C9 and clusterin were used as markers for complement activation, and myeloperoxidase (MPO) staining was performed to detect neutrophil infiltration. Western blot analysis showed that complement C9 and clusterin levels in ipsilateral basal ganglia after ICH were higher in aged rats than in young rats (p < 0.05). Immunohistochemistry showed there were more C9- and clusterin-positive cells around the hematoma in aged rats. However, MPO-positive cells in ipsilateral basal ganglia were fewer in aged rats (p < 0.05) after ICH. Our results suggest that ICH causes more severe complement activation and less neutrophil infiltration in aged rats. Clarification of the mechanisms of brain injury after ICH in the aging brain should help develop new therapeutic strategies for ICH.


Assuntos
Envelhecimento/fisiologia , Lesões Encefálicas/etiologia , Hemorragia Cerebral/complicações , Ativação do Complemento/fisiologia , Infiltração de Neutrófilos/fisiologia , Análise de Variância , Animais , Gânglios da Base/metabolismo , Gânglios da Base/patologia , Clusterina/metabolismo , Ativação do Complemento/efeitos dos fármacos , Complemento C9/metabolismo , Modelos Animais de Doenças , Lateralidade Funcional , Masculino , Infiltração de Neutrófilos/efeitos dos fármacos , Peroxidase/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
10.
Acta Neurochir Suppl ; 105: 73-7, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19066086

RESUMO

In this paper, we review current knowledge on blood-brain barrier (BBB) dysfunction following intracerebral hemorrhage (ICH). BBB disruption is a hallmark of ICH-induced brain injury. Such disruption contributes to edema formation, the influx of leukocytes, and the entry of potentially neuroactive agents into the perihematomal brain, all of which may contribute to brain injury. A range of factors have been implicated in inducing BBB disruption, including inflammatory mediators (e.g., cytokines and chemokines), thrombin, hemoglobin breakdown products, oxidative stress, complement, and matrix metalloproteinases. While there is interaction between some of these mediators, it is probable that prevention of ICH-induced BBB disruption will involve blocking multiple pathways or blocking a common end pathway (e.g., by stabilizing tight junction structure). While the effects of ICH on BBB passive permeability have been extensively examined, effects on other 'barrier' properties (metabolic and transport functions) have been less well-studied. However, recent data suggests that ICH can affect transport and that this may help protect the BBB and the brain. Indeed, it is possible in small bleeds that BBB disruption may be beneficial, and it is only in the presence of larger bleeds that disruption has detrimental effects.


Assuntos
Barreira Hematoencefálica/fisiopatologia , Hemorragia Cerebral/patologia , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Hemorragia Cerebral/tratamento farmacológico , Humanos
11.
Acta Neurochir Suppl ; 105: 95-7, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19066090

RESUMO

In this study, we examine 3CB2 expression, a marker of radial glia, after intracerebral hemorrhage (ICH). Adult male Sprague-Dawley rats received an intracaudate injection of 100 microL autologous whole blood. Animals were sacrificed, and 3CB2 expression was quantified on Western blot. Single and double labeled immunohistochemistry was used to identify which cells express 3CB2. Neurobehavioral examinations (forelimb placing test) were perfomed as an evaluation of function. By Western blot, 3CB2 was strongly expressed at day 3 and expression persisted for at least 1 month. By immunohistochemistry, 3CB2 immunoreactivity was present in large numbers of astrocytes surrounding the hematoma at day 3 after ICH. At 1 month later, 3CB2 immunoreactivity was co-localized with a neuronal marker (TUC-4). Neurobehavioral function in the 1 month after ICH group was significantly improved compared with that of 3 days after ICH. The ICH-induced 3CB2 expression in astrocytes may reflect an early response of these cells to injury, while the delayed expression in neurons might be a part of the adaptative response to injury, perhaps leading to recovery of neurobehavioral function.


Assuntos
Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/patologia , Regulação da Expressão Gênica/fisiologia , Tubulina (Proteína)/metabolismo , Análise de Variância , Animais , Hemorragia Cerebral/etiologia , Modelos Animais de Doenças , Membro Anterior/fisiopatologia , Proteína Glial Fibrilar Ácida/metabolismo , Masculino , Proteínas do Tecido Nervoso/metabolismo , Exame Neurológico , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica , Fatores de Tempo
12.
Acta Neurochir Suppl ; 105: 113-7, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19066094

RESUMO

Acute brain edema formation contributes to brain injury after intracerebral hemorrhage (ICH). It has been reported that hyperbaric oxygen (HBO) is neuroprotective in cerebral ischemia, subarachnoid hemorrhage, and brain trauma. In this study, we investigated the effects of HBO on brain edema following ICH in rats. Male Sprague-Dawley rats received intracerebral infusion of autologous whole blood, thrombin, or ferrous iron. HBO (100% O2, 3.0 ATA for 1 h) was initiated 1 h after intracerebral injection. Control rats were exposed to air at room pressure. Brains were sampled at 24 or 72 h for water content, ion measurement, and Western blot analysis. We found that 1 session of HBO reduced perihematomal brain edema (p < 0.05) 24 h after ICH. HBO also reduced heat shock protein-32 (HSP-32) levels (p < 0.05) in ipsilateral basal ganglia 24h after ICH. However, HBO failed to attenuate thrombin-induced brain edema and exaggerated ferrous iron-induced brain edema (p < 0.05). Three sessions of HBO also failed to reduce brain edema 72h after ICH. In summary, HBO reduced early perihematomal brain edema and HSP-32 levels in brain. HBO-related brain protection does not occur through reduction in thrombin toxicity because HBO failed to attenuate thrombin-induced brain edema. Our results also indicate that HBO treatment after hematoma lysis for ICH may be harmful, since HBO amplifies iron-induced brain edema.


Assuntos
Hemorragia Cerebral/terapia , Oxigenoterapia Hiperbárica/métodos , Análise de Variância , Animais , Gânglios da Base/metabolismo , Gânglios da Base/patologia , Coagulação Sanguínea/fisiologia , Edema Encefálico/etiologia , Edema Encefálico/prevenção & controle , Hemorragia Cerebral/etiologia , Hemorragia Cerebral/patologia , Modelos Animais de Doenças , Heme Oxigenase (Desciclizante)/metabolismo , Ferro/efeitos adversos , Masculino , Ratos , Ratos Sprague-Dawley , Sódio/metabolismo , Trombina/efeitos adversos , Fatores de Tempo
13.
Acta Neurochir Suppl ; 105: 127-30, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19066097

RESUMO

We examined the time course of neurological deficits in gerbils after an intracerebral hemorrhage (ICH) induced by autologous blood infusion and examined its correlation with the severity of perihematomal edema. Mongolian gerbils (n = 15) were subjected to stereotaxic autologous blood infusion (30 or 60 microL) into the left caudate nucleus. Corner-turn and forelimb-placing tests were performed before, and 1 and 3 days after ICH. Perihematomal water content was measured by tissue gravimetry. Gerbils developed neurological deficits and perihematomal edema at day 1 after ICH. Both neurological deficits and perihematomal edema were significantly greater in animals with 60 microL blood infusion compared to the 30 microL infusion group, and both neurological deficits and edema were also greater at 3 days compared to 1 day after ICH. The severity of neurological deficits paralleled the degree of perihematomal edema. We conclude that the Mongolian gerbil is a suitable model for studies on the behavioral effects of ICH.


Assuntos
Edema Encefálico/etiologia , Hemorragia Cerebral/complicações , Doenças do Sistema Nervoso/etiologia , Animais , Encéfalo/metabolismo , Edema Encefálico/patologia , Hemorragia Cerebral/etiologia , Modelos Animais de Doenças , Membro Anterior/fisiopatologia , Lateralidade Funcional , Gerbillinae , Masculino , Doenças do Sistema Nervoso/patologia , Reação Transfusional , Água/metabolismo
14.
Acta Neurochir Suppl ; 102: 317-20, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19388337

RESUMO

BACKGROUND: New protein synthesis is key to ischemic tolerance induced by preconditioning and ribosomal protein S6 kinases (p70 S6 K) are important enzymes in protein synthesis. Hyperbaric oxygen preconditioning (HBOP) reduces ischemic brain damage. This study investigated if HBOP can activate p70 S6 K and increase new protein synthesis and if HBOP induces brain tolerance against brain swelling after intracerebral hemorrhage (ICH). METHODS: There were two parts of the studies. 1) Rats received five consecutive sessions of HBOP. Twenty-four hours after HBOP, the rats had an ICH and were sacrificed one or three days later for brain edema measurement. 2) Rats received five sessions of HBOP or control pretreatment and were sacrificed for Western blot analysis and immunohistochemistry of activated p70 S6 K and heme oxygenase-1 (HO-1). FINDINGS: Five sessions of HBOP significantly reduced brain edema in the ipsilateral basal ganglia after ICH. Western blot analysis showed that HBOP activated p70 S6 K and increased HO-1 levels in the basal ganglia. Strong activated p70 S6 K immunoreactivity was also found in the basal ganglia. CONCLUSIONS: Our results suggest activation of p70 S6 K may have a role in heat shock protein synthesis after HBOP and may contribute to HBOP-induced brain protection.


Assuntos
Edema Encefálico/prevenção & controle , Hemorragia Cerebral/complicações , Hemorragia Cerebral/enzimologia , Oxigenoterapia Hiperbárica/métodos , Precondicionamento Isquêmico , Proteínas Quinases S6 Ribossômicas/metabolismo , Animais , Gânglios da Base/enzimologia , Edema Encefálico/etiologia , Edema Encefálico/patologia , Hemorragia Cerebral/patologia , Modelos Animais de Doenças , Ativação Enzimática/fisiologia , Heme Oxigenase-1/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley
15.
Acta Neurochir Suppl ; 96: 134-8, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16671441

RESUMO

The purpose of this study was to examine the possibility of neuronal remodeling and repair after cold injury-induced brain edema using immunoassays of nestin, 3CB2, and TUC-4. Male ddN strain mice were subjected to cold-induced cortical injury. Animals were divided into the following 6 groups: 1) 1-day after injury, 2) 1-week after injury, 3) 2-weeks after injury, 4) 1-month after injury, 5) sham, and 6) normal controls. Brain water content measurement, Western blot analysis, histological examination, and neurobehavioral examination were performed. Brain water content was significantly increased in the ipsilateral cortex at 1-day after injury. At 1-day and 1-week after injury, immunoreactivity of nestin, 3CB2, and TUC-4 were absent. Nestin was expressed in 3CB2-positive astrocytes at 1-month after injury, and nestin expression with TUC-4 was present in the hippocampal cell layer. Neurobehavioral function of the 1-month after injury group was significantly improved compared with function 1-day after injury. These results suggest that delayed precursor cell marker expression in glia and neuron-like cells might be part of adaptation to the injury. Although brain injury causes brain edema and neuronal death, there is the possibility of remodeling.


Assuntos
Edema Encefálico/metabolismo , Lesões Encefálicas/metabolismo , Hipocampo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Células-Tronco/metabolismo , Animais , Biomarcadores/metabolismo , Edema Encefálico/etiologia , Edema Encefálico/patologia , Lesões Encefálicas/etiologia , Lesões Encefálicas/patologia , Diferenciação Celular , Células Cultivadas , Temperatura Baixa/efeitos adversos , Hipocampo/patologia , Camundongos , Neuroglia/patologia , Neurônios/patologia , Células-Tronco/patologia
16.
Acta Neurochir Suppl ; 96: 194-8, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16671453

RESUMO

We investigated the occurrence of DNA damage in brain after intracerebral hemorrhage (ICH) and the role of iron in such injury. Male Sprague-Dawley rats received an infusion of 100 microL autologous whole blood or 30 microL FeCl2 into the right basal ganglia and were sacrificed 1, 3, or 7 days later. 8-hydroxyl-2'-deoxyguanosine (8-OHdG) was analyzed by immunohistochemistry, while the number of apurinic/apyrimidinic abasic sites (AP sites) was also quantified. 8-OHdG and AP sites are two hallmarks of DNA oxidation. DNA damage was also examined using PANT and TUNEL labeling. Dinitrophenyl (DNP) was measured by Western blot to compare the time course of protein oxidative damage to that of DNA. DNA repair APE/Ref-1 and Ku-proteins were also measured by Western blot. Bipyridine, a ferrous iron chelator, was used to examine the role of iron in ICH-induced oxidative brain injury. An increase in 8-OHdG, AP sites, and DNP levels, and a decrease in APE/Ref-1 and Ku levels were observed. Abundant PANT-positive cells were also observed in the perihematomal area 3 days after ICH. Bipyridine attenuated ICH-induced changes in PANT and DNP. These results suggest that iron-induced oxidation causes DNA damage in brain after ICH and that iron is a therapeutic target for ICH.


Assuntos
Isquemia Encefálica/induzido quimicamente , Isquemia Encefálica/genética , Hemorragia Cerebral/complicações , Hemorragia Cerebral/genética , Dano ao DNA , DNA/efeitos dos fármacos , Ferro/toxicidade , Animais , Lesões Encefálicas , Modelos Animais de Doenças , Masculino , Ratos , Ratos Sprague-Dawley
17.
Acta Neurochir Suppl ; 96: 199-202, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16671454

RESUMO

Iron overload occurs in brain after intracerebral hemorrhage (ICH). Deferoxamine, an iron chelator, attenuates perihematomal edema and oxidative stress in brain after ICH. We investigated the effects of deferoxamine on cerebrospinal fluid (CSF) free iron and brain total iron following ICH. Rats received an infusion of 100-microL autologous whole blood into the right basal ganglia, then were treated with either deferoxamine (100 mg/kg, i.p., administered 2 hours after ICH and then at 12-hour intervals for up to 7 days) or vehicle. The rats were killed at different time points from 1 to 28 days for measurement of free and total iron. Behavioral tests were also performed. Free iron levels in normal rat CSF were very low (1.1 +/- 0.4 micromol). After ICH, CSF free iron levels were increased at all time points. Levels of brain total iron were also increased after ICH (p < 0.05). Deferoxamine given 2 hours after ICH reduced free iron in CSF at all time points. Deferoxamine also reduced ICH-induced neurological deficits (p < 0.05), but did not reduce total brain iron. In conclusion, CSF free iron levels increase after ICH and do not clear for at least 28 days. Deferoxamine reduces free iron levels and improves functional outcome in the rat, indicating that it may be a potential therapeutic agent for ICH patients.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Hemorragia Cerebral/tratamento farmacológico , Hemorragia Cerebral/metabolismo , Desferroxamina/administração & dosagem , Ferro/líquido cefalorraquidiano , Transtornos Mentais/prevenção & controle , Animais , Comportamento Animal/efeitos dos fármacos , Hemorragia Cerebral/complicações , Quelantes de Ferro/administração & dosagem , Masculino , Transtornos Mentais/etiologia , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento
18.
Acta Neurochir Suppl ; 96: 203-6, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16671455

RESUMO

Pretreatment with low-dose thrombin attenuates brain edema induced by iron or intracerebral hemorrhage (ICH). Ceruloplasmin is involved in iron metabolism by oxidizing ferrous iron to ferric iron. The present study examines whether thrombin modulates brain ceruloplasmin levels and whether exogenous ceruloplasmin reduces brain edema induced by ferrous iron in vivo. In the first set of experiments, rats received intracerebral infusion of saline or 1 U thrombin into the right basal ganglia. Rats were killed 1, 3, or 7 days later for Western blot analysis and RT-PCR analysis. In the second set of experiments, rats received either ferric iron, ferrous iron, or ferrous iron plus ceruloplasmin, then were killed 24 hours later for brain edema measurement. We found that ceruloplasmin protein levels in the ipsilateral basal ganglia increased on the first day after thrombin stimulation and peaked at day 3. Brain ceruloplasmin levels were higher after thrombin infusion than after saline injection. RT-PCR showed that brain ceruloplasmin mRNA levels were also up-regulated after thrombin injection (p < 0.05). We also found ipsilateral brain edema after intracerebral infusion of ferrous iron but not ferric iron at 24 hours. Co-injection of ferrous iron with ceruloplasmin reduced ferrous iron-induced brain edema (p < 0.05). Our results demonstrate that thrombin increases brain ceruloplasmin levels and exogenous ceruloplasmin reduces ferrous iron-induced brain edema, suggesting that ceruloplasmin up-regulation may contribute to thrombin-induced brain tolerance to ICH by limiting the injury caused by ferrous iron released from the hematoma.


Assuntos
Edema Encefálico/tratamento farmacológico , Edema Encefálico/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Ceruloplasmina/metabolismo , Ceruloplasmina/uso terapêutico , Trombina/administração & dosagem , Animais , Edema Encefálico/diagnóstico , Precondicionamento Isquêmico/métodos , Masculino , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento , Regulação para Cima/efeitos dos fármacos
19.
Acta Neurochir Suppl ; 96: 207-11, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16671456

RESUMO

The aims of the current study were 1) to establish an adult rat model of intraventricular hemorrhage (IVH) and post-hemorrhagic ventricular dilatation, and 2) to examine the role of alterations in cerebrospinal fluid (CSF) drainage and parenchymal injury in that dilatation. Rats underwent infusion of 200 microl of autologous blood over 15 minutes. The rats were used to measure hematoma mass, ventricular dilatation, and cortical mantle volume (with T2 imaging), resistance to CSF absorption, and brain edema (as a marker of brain injury). IVH resulted in ventricular dilatation peaking at day 2 but persisting for at least 8 weeks. Although there was an increased resistance to CSF absorption at 3 days, it returned to normal at day 7. Long-term ventricular dilatation was not associated with an alteration in cortical mantle volume, although there was evidence of cortical damage (edema). It is possible that initial ventricular distension (due to the hematoma and the impaired CSF drainage) in combination with periventricular white matter damage results in structural changes that prevent total recoil once the hematoma has resolved and CSF drainage is normalized, leading to long-term hydrocephalus.


Assuntos
Hemorragia Cerebral/líquido cefalorraquidiano , Hemorragia Cerebral/patologia , Ventrículos Cerebrais/metabolismo , Ventrículos Cerebrais/patologia , Modelos Animais de Doenças , Hidrocefalia/líquido cefalorraquidiano , Hidrocefalia/patologia , Animais , Hemorragia Cerebral/complicações , Dilatação Patológica , Hidrocefalia/complicações , Masculino , Ratos , Ratos Sprague-Dawley
20.
Acta Neurochir Suppl ; 96: 218-21, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16671458

RESUMO

The present study examined differences in intracerebral hemorrhage (ICH)-induced brain injury in male and female rats, whether delayed administration of 17beta-estradiol can reduce ICH-induced brain damage, and whether these effects are estrogen receptor (ER)-dependent. Male and female Sprague-Dawley rats received an infusion of 100-microL autologous whole blood into the right basal ganglia. The effects of 1beta-estradiol (5 mg/kg, i.p.) on ICH-induced brain injury were examined by measuring brain edema and neurological deficits 24 hours later. Heme oxygenase-1 (HO-1) was investigated by immuno-analysis. Brain edema was significantly less in female compared to male rats. The ER antagonist ICI182,780 exacerbated ICH-induced brain edema in female but not in male rats, suggesting that ER activation during ICH is protective in female rats. Administration of 17beta-estradiol to male (but not female) rats significantly reduced brain edema, neurological deficits, and ICH-induced increases in brain HO-1 levels when given 2 hours after ICH. This study showed that female rats have less ICH-induced injury than male rats. ER is involved in limiting ICH-induced injury in female rats. ICH-injury in male rats can be reduced by 17beta-estradiol. Since 17beta-estradiol treatment was effective in male rats, it could be a potential therapeutic agent for ICH.


Assuntos
Lesões Encefálicas/etiologia , Lesões Encefálicas/metabolismo , Hemorragia Cerebral/complicações , Hemorragia Cerebral/metabolismo , Modelos Animais de Doenças , Estrogênios/administração & dosagem , Estrogênios/metabolismo , Animais , Lesões Encefálicas/diagnóstico , Lesões Encefálicas/tratamento farmacológico , Hemorragia Cerebral/diagnóstico , Hemorragia Cerebral/tratamento farmacológico , Feminino , Masculino , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/metabolismo , Ratos , Ratos Sprague-Dawley , Distribuição por Sexo , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA