Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neurochem Res ; 44(9): 2182-2189, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31410708

RESUMO

Tripartite motif 32 (TRIM32) is a member of TRIM family that plays a potential role in neural regeneration. However, the biological function of TRIM32 in cerebral ischemia reperfusion injury has not been investigated. In the present study, we evaluated the expression level of TRIM32 in hippocampal neurons following oxygen-glucose deprivation/reperfusion (OGD/R). The results showed that TRIM32 expression was significantly elevated in hippocampal neurons subjected to OGD/R as compared to the neurons cultured in the normoxia condition. To further evaluate the role of TRIM32, hippocampal neurons were transfected with TRIM32 small interfering RNA (si-TRIM32) to knock down TRIM32. We found that knockdown of TRIM32 improved cell viability of OGD/R-stimulated hippocampal neurons. Generation of reactive oxygen species was decreased, while contents of superoxide dismutase and glutathione peroxidase were increased after si-TRIM32 transfection. Knockdown of TRIM32 suppressed cell apoptosis, as proved by the increased bcl-2 expression along with decreased bax expression and caspase-3 activity. We also found that TRIM32 knockdown enhanced OGD/R-induced activation of Nrf2 signaling pathway in hippocampal neurons. Furthermore, siRNA-Nrf2 was transfected to knock down Nrf2. SiRNA-Nrf2 transfection reversed the protective effects of TRIM32 knockdown on neurons. These data suggested that knockdown of TRIM32 protected hippocampal neurons from OGD/R-induced oxidative injury through activating Nrf2 signaling pathway.


Assuntos
Hipocampo/metabolismo , Neurônios/metabolismo , Estresse Oxidativo/fisiologia , Fatores de Transcrição/metabolismo , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Apoptose/fisiologia , Feminino , Técnicas de Silenciamento de Genes , Glucose/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Neuroproteção/fisiologia , Oxigênio/metabolismo , Gravidez , Ratos Wistar , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais/fisiologia , Fatores de Transcrição/genética , Proteínas com Motivo Tripartido/genética , Ubiquitina-Proteína Ligases/genética
2.
Cancer Cell Int ; 18: 195, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30524203

RESUMO

BACKGROUND: MicroRNAs play crucial roles in tumorigenesis and tumor progression. miR-770 has been reported to be downregulated in several cancers and affects cancer cell proliferation, apoptosis, metastasis and drug resistance. However, the role and underlying molecular mechanism of miR-770 in human glioma remain unknown and need to be further elucidated. METHODS: The expression of miR-770 in glioma tissues and cell lines was measured by quantitative real-time PCR (qRT-PCR) to explore the association of miR-770 expression with clinicopathological characteristics. The expression of CDK8 was detected by qRT-PCR and Western blotting in glioma tissues. A target prediction program and a dual-luciferase reporter assay were used to confirm that CDK8 is a target gene of miR-770. MTT and cell counting assays were used to assess the effect of miR-770 on glioma cell proliferation. The cell cycle distribution and apoptosis were examined by flow cytometry. CDK8 siRNA and overexpression were used to further confirm the function of the target gene. RESULTS: We demonstrated that miR-770 expression was downregulated in human glioma tissues and cell lines. The overexpression of miR-770 inhibited glioma cell proliferation and cell cycle G1-S transition and induced apoptosis. The inhibition of miR-770 facilitated cell proliferation and G1-S transition and suppressed apoptosis. miR-770 expression was inversely correlated with CDK8 expression in glioma tissues. CDK8 was confirmed to be a direct target of miR-770 by using a luciferase reporter assay. The overexpression of miR-770 decreased CDK8 expression at both the mRNA and protein levels, and the suppression of miR-770 increased CDK8 expression. Importantly, CDK8 silencing recapitulated the cellular and molecular effects observed upon miR-770 overexpression, and CDK8 overexpression eliminated the effects of miR-770 overexpression on glioma cells. Moreover, both exogenous expression of miR-770 and silencing of CDK8 resulted in suppression of the Wnt/ß-catenin signaling pathway. CONCLUSIONS: Our study demonstrates that miR-770 inhibits glioma cell proliferation and G1-S transition and induces apoptosis through suppression of the Wnt/ß-catenin signaling pathway by targeting CDK8. These findings suggest that miR-770 plays a significant role in glioma progression and serves as a potential therapeutic target for glioma.

3.
Epilepsy Res ; 127: 40-49, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27561187

RESUMO

Abnormal hippocampal neurogenesis after acute seizures has been well addressed. However, whether newly generated cells continued to be disturbed even they were born in the chronic stage after pilocarpine-induce status epilepticus has remained elusive. Labeling dividing progenitors and their progeny with retroviral vector expressing green fluorescent protein or proliferation marker 5-bromo-2'-deoxyuridine at 3 months post pilocarpine-induced status epilepticus in mice, a spot of newly born neurons exhibiting hilar ectopic location (4.57±2.3%), aberrant basal dendrites (8.09±1.5%) and abnormal axon sprouting into inner molecular layer of dentate gyrus was identified when examined 6 weeks later. No significant difference on the amount of mossy fiber sprouting was found when cohorts of newborn cells were eliminated by methylazoxymethanol acetate injection initiated at 3 months after SE, suggesting that adult generated neurons in the chronically epileptic hippocampus don't contribute a lot to the mossy fiber sprouting. These results indicated that the aberrant neurogenesis in the chronically epileptic hippocampus occurs only in a small population of newly generated granule cells.


Assuntos
Epilepsia/fisiopatologia , Hipocampo/fisiopatologia , Neurogênese/fisiologia , Animais , Bromodesoxiuridina , Movimento Celular , Doença Crônica , Modelos Animais de Doenças , Epilepsia/patologia , Feminino , Imunofluorescência , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/patologia , Camundongos , Neurônios/patologia , Neurônios/fisiologia , Pilocarpina
4.
J Mol Neurosci ; 58(4): 507-16, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26768135

RESUMO

MicroRNA-25 (miR-25) has been reported to be a major miRNA marker in neural cells and is strongly expressed in ischemic brain tissues. However, the precise mechanism and effect of miR-25 in cerebral ischemia/reperfusion (I/R) injury needs further investigations. In the present study, the oxygen-glucose deprivation (OGD) model was constructed in human SH-SY5Y and IMR-32 cells to mimic I/R injury and to evaluate the role of miR-25 in regulating OGD/reperfusion (OGDR)-induced cell apoptosis. We found that miR-25 was downregulated in the OGDR model. Overexpression of miR-25 via miRNA-mimics transfection remarkably inhibited OGDR-induced cell apoptosis. Moreover, Fas was predicted as a target gene of miR-25 through bioinformatic analysis. The interaction between miR-25 and 3'-untranslated region (UTR) of Fas mRNA was confirmed by dual-luciferase reporter assay. Fas protein expression was downregulated by miR-25 overexpression in OGDR model. Subsequently, the small interfering RNA (siRNA)-mediated knockdown of Fas expression also inhibited cell apoptosis induced by OGDR model; in contrast, Fas overexpression abrogated the protective effects of miR-25 on OGDR-induced cells. Taken together, our results indicate that the upregulation of miR-25 inhibits cerebral I/R injury-induced apoptosis through downregulating Fas/FasL, which will provide a promising therapeutic target.


Assuntos
Apoptose , Glucose/deficiência , MicroRNAs/genética , Neurônios/metabolismo , Oxigênio/metabolismo , Hipóxia Celular , Linhagem Celular Tumoral , Regulação para Baixo , Proteína Ligante Fas/metabolismo , Humanos , MicroRNAs/metabolismo , Transdução de Sinais , Receptor fas/metabolismo
5.
Exp Neurol ; 273: 273-87, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26384773

RESUMO

Emerging evidence has linked chronic temporal lobe epilepsy to dramatically reduced neurogenesis in the dentate gyrus. However, the profile of different components of neurogenesis in the chronically epileptic hippocampus is still unclear, especially the incorporation of newly generated cells. To address the issue, newly generated cells in the sub-granular zone of the dentate gyrus were labeled by the proliferation marker bromodeoxyuridine (BrdU) or retroviral vector expressing green fluorescent protein 2 months after pilocarpine-induced status epilepticus. The newly generated neurons that extended axons to CA3 area or integrated into memory circuits were visualized by cholera toxin B subunit retrograde tracing, and detecting activation of BrdU(+) cells following a recall of spatial memory test at the chronic stage of TLE. We found that the microenvironment was still able to sustain significant neuronal differentiation of newly generated cells at 2 months post-status epilepticus time-point, and newly added neurons into granular cell layer were still able to integrate into neuronal circuitry, both anatomically and functionally. Quantified analyses of BrdU(+) or Ki-67(+) cells demonstrated that there was a reduced proliferation of progenitor cells and diminished survival of newly generated cells in the epileptic hippocampus. Both decreased levels of neurotrophic factors in the surrounding milieu and cell loss in the CA3 area might contribute the decreased production of new cells and their survival following chronic epilepsy. These results suggest that decreased neurogenesis in the chronically epileptic hippocampus 2 months post status epilepticus is not associated with altered integration of newly generated neurons, and that developing strategies to augment hippocampal neurogenesis in chronic epilepsy might be protective.


Assuntos
Hipocampo/patologia , Hipocampo/fisiopatologia , Vias Neurais/fisiologia , Neurogênese/fisiologia , Neurônios/fisiologia , Estado Epiléptico/patologia , Animais , Diferenciação Celular , Movimento Celular , Toxina da Cólera/metabolismo , Modelos Animais de Doenças , Feminino , Seguimentos , Camundongos , Camundongos Endogâmicos , Agonistas Muscarínicos , Rede Nervosa/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Pilocarpina/toxicidade , Retroviridae/genética , Estado Epiléptico/induzido quimicamente , Células-Tronco , Fatores de Tempo , Transdução Genética
6.
Int J Biochem Cell Biol ; 64: 252-64, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25936756

RESUMO

Mounting evidence suggests that the pathological hallmarks of Alzheimer's disease (AD) are caused by the intraneuronal accumulation of beta-amyloid protein (Aß). Reuptake of extracellular Aß is believed to contribute significantly to the intraneuronal Aß pool in the early stages of AD. Published reports have claimed that the low-density lipoprotein receptor-related protein 1 (LRP1) mediates Aß1-42 uptake and lysosomal trafficking in GT1-7 neuronal cells and mouse embryonic fibroblast non-neuronal cells. However, there is no direct evidence supporting the role of LRP1 in Aß internalization in primary neurons. Our recent study indicated that p38 MAPK and ERK1/2 signaling pathways are involved in regulating α7 nicotinic acetylcholine receptor (α7nAChR)-mediated Aß1-42 uptake in SH-SY5Y cells. This study was designed to explore the regulation of MAPK signaling pathways on LRP1-mediated Aß internalization in neurons. We found that extracellular Aß1-42 oligomers could be internalized into endosomes/lysosomes and mitochondria in cortical neurons. Aß1-42 and LRP1 were also found co-localized in neurons during Aß1-42 internalization, and they could form Aß1-42-LRP1 complex. Knockdown of LRP1 expression significantly decreased neuronal Aß1-42 internalization. Finally, we identified that p38 MAPK and ERK1/2 signaling pathways regulated the internalization of Aß1-42 via LRP1. Therefore, these results demonstrated that LRP1, p38 MAPK and ERK1/2 mediated the internalization of Aß1-42 in neurons and provided evidence that blockade of LRP1 or inhibitions of MAPK signaling pathways might be a potential approach to lowering brain Aß levels and served a potential therapeutic target for AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Sistema de Sinalização das MAP Quinases , Fragmentos de Peptídeos/metabolismo , Receptores de LDL/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Linhagem Celular Tumoral , Células Cultivadas , Córtex Cerebral/citologia , Endossomos/metabolismo , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Lisossomos/metabolismo , Camundongos , Mitocôndrias/metabolismo , Neurônios , Cultura Primária de Células , Transporte Proteico
7.
Neurochem Int ; 68: 10-7, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24480781

RESUMO

We investigated localization of Phospholipase C beta (PLCß1 and PLCß4) in laminaes of dorsal hippocampus and different subtypes of hippocampal interneurons in normal Kunming mouse, and their progressive changes during pilocarpine induced status epilepticus (SE) by quantitative immunohistochemistry and real time PCR. PLCß1 was observed in the pyramidal layer of CA1-3 area, hilus of the dentate gyrus, whereas PLCß4 was mainly expressed in calcium binding protein positive interneurons, i.e. calbindin, calretinin, parvalbumin positive interneurons in the strata oriens, radiatum of the CA area and hilus of the dentate gyrus. During pilocarpine induced SE, a temporary down-regulation of PLCß4 in the interneurons of CA area at SE 30min, and a progressive reduction of PLCß1/PLCß4 in dentate hilar cells were demonstrated. These findings confirm and extend the regional specific distribution of PLCß1 and PLCß4 immunoreactivity in mouse hippocampus, and suggest that PLCß1 and PLCß4 may play an important role in maintenance of the status epilepticus.


Assuntos
Hipocampo/efeitos dos fármacos , Interneurônios/enzimologia , Fosfolipase C beta/metabolismo , Pilocarpina/farmacologia , Estado Epiléptico/enzimologia , Animais , Modelos Animais de Doenças , Regulação para Baixo/fisiologia , Feminino , Hipocampo/enzimologia , Imuno-Histoquímica/métodos , Camundongos , Estado Epiléptico/induzido quimicamente
8.
Neurochem Int ; 61(2): 227-35, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22561406

RESUMO

The characteristic pathological change of Alzheimer's disease (AD) include deposits of ß-amyloid protein (Aß) in brain, neurofibrillary tangles (NFTs), as well as a few neuronal loss. Evidence shows that Aß causes calcium influx and induces the cleavage of p35 into p25. Furthermore, the binding of p25 to cyclin-dependent kinase 5 (Cdk5) constitutively activates Cdk5. The p25/Cdk5 complex then hyperphosphorylates tau. Tanshinone IIA (tanIIA), a natural product extracted from Chinese herbal medicine Salvia miltiorrhiza BUNGE, has been reported to exert antioxidative activity. However, its neuroprotective activity remains unclear. The present study determined whether tanIIA protects neurons against Aß(25-35)-induced cytotoxicity and detected the association of this protective effect with calpain and the p35/Cdk5 pathway. The results showed that tanIIA protected neurons against the neurotoxicity of Aß(25-35), increased the viability of neurons, decreased expression of phosphorylated tau in neurons induced by Aß(25-35), improved the impairment of the cell ultrastructure (such as nuclear condensation and fragmentation, and neurofibril collapse). Further more, we found that tanIIA maintained the normal expression of p35 on peripheral membranes, and decreased p25 expression in the cytoplasm. TanIIA also inhibited the translocation of Cdk5 from the nucleus into the cytoplasm of primary neurons induced by Aß(25-35). These data suggested that tanIIA possessed neuroprotective action and the protection may involve in calpain and the p35/Cdk5 pathway.


Assuntos
Abietanos/farmacologia , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/toxicidade , Calpaína/fisiologia , Córtex Cerebral/metabolismo , Neurônios/metabolismo , Fármacos Neuroprotetores , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/toxicidade , Fosfotransferases/fisiologia , Animais , Western Blotting , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/ultraestrutura , Citoplasma/metabolismo , Feminino , Imuno-Histoquímica , Camundongos , Microscopia Eletrônica de Transmissão , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Fosforilação , Gravidez , Transporte Proteico , Transdução de Sinais/efeitos dos fármacos , Sais de Tetrazólio , Tiazóis , Proteínas tau/biossíntese
9.
Lasers Med Sci ; 25(5): 711-7, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20428912

RESUMO

Histamine is a powerful modulator that regulates blood vessels and blood flow. The effect of histamine on the extracortical vessels has been well described, while much less is known about the effect of histamine on intracortical vessels. In this study, we investigated the effect of histamine on regional cerebral blood flow in rat parietal lobe with laser Doppler flowmetry. The pharmacological characteristics of distinct ways (intracerebroventricular injection, intraperitoneal injection, and cranial window infusion) in applying histamine to the brain were also obtained and compared. Histamine applied in three ways all produced a decrease of rCBF in parietal lobe in a concentration-dependent manner. Cranial window infusion was the most effective way and intraperitoneal injection of L-histidine was the most ineffective, although it is a simple and applied way. To determine which type of receptor takes part in the vessel contraction induced by histamine, H1 receptor antagonist, diphenhydramine, and H2 receptor antagonist, cimetidine, were applied, respectively, before histamine administration. When the injection of cimetidine was conducted in advance, histamine still resulted in a decrease of infusion amount; while the injection of diphenhydramine was conducted in advance, the infusion of blood amount wasn't changed. These findings indicated that histamine could result in a reduction of rCBF in the rat parietal lobe and this effect of histamine may attribute partly to its combination with H1 receptor.


Assuntos
Circulação Cerebrovascular/efeitos dos fármacos , Histamina/farmacologia , Lobo Parietal/irrigação sanguínea , Lobo Parietal/efeitos dos fármacos , Animais , Cimetidina/farmacologia , Difenidramina/farmacologia , Histamina/administração & dosagem , Antagonistas dos Receptores Histamínicos H1/farmacologia , Antagonistas dos Receptores H2 da Histamina/farmacologia , Histidina/administração & dosagem , Injeções Intraperitoneais , Injeções Intraventriculares , Fluxometria por Laser-Doppler , Masculino , Ratos , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional/efeitos dos fármacos
10.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 33(2): 99-102, 2008 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-18326902

RESUMO

OBJECTIVE: To prove the existence neurons in the rat corpus callosum, the potential function of these neurons and their connection. METHODS: Immunohistochemistry was used performed to examine the expressions of NeuN, a mature neuron marker,and N-type voltage-dependent valcium channel alpha1-subunit (Cav2.2)in the section of the rat corpus callosum. Horseradish peroxidase (HRP) normal sodium solution (30%), the retrograde tracer,was injected under the frontal forceps of corpus callousm and HRP absorbed by the process of neurons in the brain slices was stained with tetramethyl benzidine. RESULTS: There were some NeuN positive cells in the rat corpus callosum and Cav2.2 was detected in some of these NeuN positive cells.Neurons with positive HRP were found in the rat corpus callosum and some of these neurons connected to the cortex or corpus striatum. CONCLUSION: There are a few neurons in the corpus callosum of adult rats and some of them express Cav2.2. Neurons in the corpus callosum have connections with the brain cortex or corpora striatum.


Assuntos
Canais de Cálcio Tipo N/biossíntese , Corpo Caloso/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Neurônios/citologia , Proteínas Nucleares/biossíntese , Animais , Corpo Caloso/citologia , Proteínas de Ligação a DNA , Masculino , Vias Neurais/fisiologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley
11.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 32(3): 396-400, 2007 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-17611313

RESUMO

OBJECTIVE: To observe the effect of ligustrazine on cell proliferation in subventricular zone (SVZ) in rat brain with focal cerebral ischemia reperfusion injury. METHODS: Male SD rats were randomly divided into a normal group,a sham operation group,a ligustrazine treatment group, and a control group. The ligustrazine treatment group and the control group were further divided into 5 subgroups: 1d, 3d, 7d, 14d, and 21d reperfusion after 2h middle cerebral artery occlusion (MCAO). The focal cerebral ischemia-reperfusion model was made by MCAO. S phase cells were labelled with BrdU. Immunohistochemistry method was conducted to detect the BrdU positive cells. The total number of BrdU positive cells in the SVZ was measured. The expression of neuro nitric oxide synthase (nNOS) was detected with Western blot method. RESULTS: There was a significant increase of BrdU positive cells in SVZ of ligustrazine treatment in the 1d and 3d group compared with that of the control group (P<0.01). The total number of BrdU positive cells reached a peak in 7d group and declined afterwards. Cells proliferated also in SVZ on the contralateral side, and peaked at 7d. The nNOS expression of ligustrazine administration after the focal cerebral ischemia-reperfusion decreased at 1d and 3d after the reperfusion compared with that of the control group (P<0.05), and increased at 7d, but with no significant difference compared with that of the control group. CONCLUSION: Ligustrazine may promote the cell proliferation in SVZ of adult rats with ischemia-reperfusion injury by decreasing the nNOS expression.


Assuntos
Isquemia Encefálica/fisiopatologia , Proliferação de Células/efeitos dos fármacos , Ventrículos Cerebrais/patologia , Pirazinas/farmacologia , Traumatismo por Reperfusão/fisiopatologia , Animais , Western Blotting , Ventrículos Cerebrais/metabolismo , Infarto da Artéria Cerebral Média/fisiopatologia , Masculino , Óxido Nítrico Sintase Tipo I/metabolismo , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
12.
Nan Fang Yi Ke Da Xue Xue Bao ; 27(6): 771-4, 2007 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-17584635

RESUMO

OBJECTIVE: To observe the effect of ligustrazine on cell proliferation in the subventricular zone (SVZ) and dentate gyrus (DG) and nNOS expression in rat brain after cerebral ischemia-reperfusion injury. METHODS: Male SD rats were randomly divided into normal control group, sham operation group, model group and ligustrazine treatment group. The latter two groups were further divided into 5 subgroups for observation at 1, 3, 7, 14 and 21 days after reperfusion following a 2-hour middle cerebral artery occlusion (MCAO). The cells in S phase were labeled with BrdU, and immunohistochemistry was employed to detect BrdU- and nNOS-positive cells. The numbers of BrdU-positive cells in the SVZ and DG were measured. The expression of nNOS was detected by Western blotting. RESULTS: nNOS expression increased significantly in the model group as compared to the sham operation group (P<0.05), and ligustrazine treatment significantly lowered the expression level in comparison with the model group (P<0.05). Compared with the model group, a significant increase in BrdU-positive cells occurred in the SVZ of rats 1 and 3 days after igustrazine treatment (P<0.05), along with an increase of DG BrdU-positive cells. CONCLUSION: Ligustrazine significantly restrains ischemia-reperfusion injury-induced nNOS activity enhancement and promotes cell proliferation in the SVZ and DG of adult rats after ischemia-reperfusion injury.


Assuntos
Regeneração Nervosa/efeitos dos fármacos , Óxido Nítrico Sintase Tipo I/biossíntese , Pirazinas/farmacologia , Traumatismo por Reperfusão/fisiopatologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Western Blotting , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Isquemia Encefálica/complicações , Proliferação de Células/efeitos dos fármacos , Ventrículos Cerebrais/irrigação sanguínea , Ventrículos Cerebrais/efeitos dos fármacos , Ventrículos Cerebrais/patologia , Giro Denteado/irrigação sanguínea , Giro Denteado/efeitos dos fármacos , Giro Denteado/patologia , Imuno-Histoquímica , Masculino , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/etiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...