Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Neuroreport ; 22(3): 126-30, 2011 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-21266900

RESUMO

Interleukin-6 (IL-6) is a proinflammatory cytokine known to modulate neurogenesis. We presently evaluated neural progenitor proliferation, survival, and phenotypic maturation in the hippocampal dentate gyrus, subventricular zone, and the posterior periventricle in the brains of IL-6 knockout mice and their wild-type littermates. In all the three neurogenic regions of the IL-6 mice there was a significant decrease in the number of 5-bromo-2-deoxyuridine positive (BrdU) proliferating progenitors compared with the IL-6 mice. The IL-6 mice also showed a significantly lower progenitor cell survival in the dentate gyrus and subventricular zone compared with the IL-6 mice. In conclusion, a complete lack of IL-6 might be detrimental to neurogenesis in the adult brain.


Assuntos
Giro Denteado/fisiologia , Interleucina-6/deficiência , Interleucina-6/genética , Neurogênese/genética , Telencéfalo/fisiologia , Animais , Diferenciação Celular/genética , Proliferação de Células , Sobrevivência Celular/genética , Giro Denteado/citologia , Ventrículos Laterais/citologia , Camundongos , Camundongos Knockout , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Plasticidade Neuronal/genética , Telencéfalo/citologia
2.
J Neurochem ; 116(4): 499-507, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21133923

RESUMO

One of the limiting factors in stroke therapeutic development is the use of animal models that do not well represent the underlying medical conditions of patients. In humans, diabetes increases the risk of stroke incidence as well as post-stroke mortality. To understand the mechanisms that render diabetics to increased brain damage, we evaluated the effect of transient middle cerebral artery occlusion in adult db/db mice. The db/db mouse is a model of type-2 diabetes with four times higher blood sugar than its normoglycemic genetic control(db/+ mouse). Following transient middle cerebral artery occlusion, the db/db mice showed significantly higher mortality, bigger infarcts, increased cerebral edema, worsened neurological status compared to db/+ mice. The db/db mice also showed significantly higher post-ischemic inflammatory markers (ICAM1(+) capillaries, extravasated macrophages/neutrophils and exacerbated proinflammatory gene expression) compared to db/+ mice. In addition, the post-ischemic neuroprotective heat-shock chaperone gene expression was curtailed in the db/db compared to db/+ mice.


Assuntos
Edema Encefálico/patologia , Isquemia Encefálica/patologia , Diabetes Mellitus Tipo 2/patologia , Infarto da Artéria Cerebral Média/patologia , Animais , Glicemia/genética , Glicemia/metabolismo , Peso Corporal/genética , Edema Encefálico/etiologia , Edema Encefálico/genética , Isquemia Encefálica/etiologia , Isquemia Encefálica/genética , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/genética , Progressão da Doença , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/genética , Inflamação/etiologia , Inflamação/genética , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Transgênicos
3.
J Cereb Blood Flow Metab ; 29(4): 675-87, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19142192

RESUMO

MicroRNAs (miRNAs) are approximately 22 nucleotides long, noncoding RNAs that control cellular function by either degrading mRNAs or arresting their translation. To understand their functional significance in ischemic pathophysiology, we profiled miRNAs in adult rat brain as a function of reperfusion time after transient middle cerebral artery occlusion. Of the 238 miRNAs evaluated, 8 showed increased and 12 showed decreased expression at least at 4 out of 5 reperfusion time points studied between 3 h and 3 days compared with sham. Of those, 17 showed >5 fold change. Bioinformatics analysis indicated a correlation between miRNAs altered to several mRNAs known to mediate inflammation, transcription, neuroprotection, receptors function, and ionic homeostasis. Antagomir-mediated prevention of mir-145 expression led to an increased protein expression of its downstream target superoxide dismutase-2 in the postischemic brain. In silico analysis showed sequence complementarity of eight miRNAs induced after focal ischemia to 877 promoters indicating the possibility of noncoding RNA-induced activation of gene expression. The mRNA expression of the RNases Drosha and Dicer, cofactor Pasha, and the pre-miRNA transporter exportin-5, which modulate miRNA biogenesis, were not altered after transient middle cerebral artery occlusion. Thus, the present studies indicate a critical role of miRNAs in controlling mRNA transcription and translation in the postischemic brain.


Assuntos
Córtex Cerebral/metabolismo , Ataque Isquêmico Transitório/genética , MicroRNAs/análise , Animais , Infarto Cerebral , Biologia Computacional , Perfilação da Expressão Gênica , Infarto da Artéria Cerebral Média , Biossíntese de Proteínas , Ratos , Reperfusão , Ribonucleases/genética , Superóxido Dismutase/genética , Fatores de Tempo , Transcrição Gênica
4.
J Neurochem ; 105(4): 1313-24, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18208539

RESUMO

Early growth response-1 (Egr1) is a sequence-specific transcription factor (TF) which is induced under hypoxic conditions. We presently report that transient middle cerebral artery occlusion (MCAO) leads to increased expression of Egr1 in the brains of adult mice and rats between 2 h and 5 days of reperfusion with a peak increase of 8-12-fold at 1 day. When subjected to transient MCAO and 3 days of reperfusion, Egr1-/- mice showed significantly smaller infarcts (by 44.9 +/- 8.4%, p < 0.05) and improved neurological function than Egr1+/+ littermates. Following transient MCAO, brains of Egr1-/- mice showed less water accumulation and decreased neutrophil infiltration (by 42 +/- 8%, p < 0.05) compared to Egr1+/+ mice. The number of activated microglia/macrophages were also significantly lower (OX42+ cells by 53 +/- 9%, p < 0.05 and ED1+ cells by 59 +/- 11%) in the post-ischemic cortex of Egr1-/- mice compared to Egr1+/+ mice. In addition, post-ischemic inflammatory gene expression was less pronounced in the brains of Egr1-/- mice compared to Egr1+/+ mice. Preventing cerebral Egr1 protein induction with small interference RNAs that target Egr1 decreased inflammatory gene expression and led to smaller infarcts (by 40.2 +/- 6.9%, p < 0.05) and reduced neurological deficits in rats subjected to transient MCAO. Conversely, transient MCAO following adenoviral-mediated Egr1 over-expression exacerbated the infarct volume (by 29 +/- 5.3%, p < 0.05) and worsened the neurological deficits in rats. These studies indicate Egr1 as a significant contributor of inflammation and neuronal damage after stroke.


Assuntos
Proteína 1 de Resposta de Crescimento Precoce/biossíntese , Regulação da Expressão Gênica/fisiologia , Hipóxia Encefálica/metabolismo , Mediadores da Inflamação/fisiologia , Ataque Isquêmico Transitório/metabolismo , Animais , Linhagem Celular , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 1 de Resposta de Crescimento Precoce/fisiologia , Humanos , Hipóxia Encefálica/genética , Hipóxia Encefálica/patologia , Ataque Isquêmico Transitório/genética , Ataque Isquêmico Transitório/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos , Ratos Endogâmicos SHR , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia
5.
J Neurochem ; 101(1): 41-56, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17394460

RESUMO

Thiazolidinediones (TZDs) are synthetic agonists of the ligand-activated transcription factor peroxisome proliferator-activated receptor-gamma (PPARgamma). TZDs are known to curtail inflammation associated with peripheral organ ischemia. As inflammation precipitates the neuronal death after stroke, we tested the efficacy of TZDs in preventing brain damage following transient middle cerebral artery occlusion (MCAO) in adult rodents. As hypertension and diabetes complicate the stroke outcome, we also evaluated the efficacy of TZDs in hypertensive rats and type-2 diabetic mice subjected to transient MCAO. Pre-treatment as well as post-treatment with TZDs rosiglitazone and pioglitazone significantly decreased the infarct volume and neurological deficits in normotensive, normoglycemic, hypertensive and hyperglycemic rodents. Rosiglitazone neuroprotection was not enhanced by retinoic acid x receptor agonist 9-cis-retinoic acid, but was prevented by PPARgamma antagonist GW9662. Rosiglitazone significantly decreased the post-ischemic intercellular adhesion molecule-1 expression and extravasation of macrophages and neutrophils into brain. Rosiglitazone treatment curtailed the post-ischemic expression of the pro-inflammatory genes interleukin-1beta, interleukin-6, macrophage inflammatory protein-1alpha, monocyte chemoattractant protein-1, cyclooxygenase-2, inducible nitric oxide synthase, early growth response-1, CCAAT/enhancer binding protein-beta and nuclear factor-kappa B, and increased the expression of the anti-oxidant enzymes catalase and copper/zinc-superoxide dismutase. Rosiglitazone also increased the expression of the anti-inflammatory gene suppressor of cytokine signaling-3 and prevented the phosphorylation of the transcription factor signal transducer and activator of transcription-3 after focal ischemia. Thus, PPARgamma activation with TZDs might be a potent therapeutic option for preventing inflammation and neuronal damage after stroke with promise in diabetic and hypertensive subjects.


Assuntos
Infarto Cerebral/tratamento farmacológico , Diabetes Mellitus Tipo 2/fisiopatologia , Hipertensão/fisiopatologia , Ataque Isquêmico Transitório/tratamento farmacológico , PPAR gama/agonistas , Tiazolidinedionas/farmacologia , Anilidas/farmacologia , Animais , Infarto Cerebral/fisiopatologia , Infarto Cerebral/prevenção & controle , Quimiotaxia de Leucócito/efeitos dos fármacos , Quimiotaxia de Leucócito/fisiologia , Citocinas/genética , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/genética , Modelos Animais de Doenças , Encefalite/tratamento farmacológico , Encefalite/metabolismo , Encefalite/fisiopatologia , Hipertensão/genética , Molécula 1 de Adesão Intercelular/efeitos dos fármacos , Molécula 1 de Adesão Intercelular/metabolismo , Ataque Isquêmico Transitório/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , PPAR gama/antagonistas & inibidores , PPAR gama/metabolismo , Pioglitazona , Ratos , Ratos Endogâmicos SHR , Ratos Sprague-Dawley , Rosiglitazona , Superóxido Dismutase/efeitos dos fármacos , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1 , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/efeitos dos fármacos , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Tiazolidinedionas/química , Tiazolidinedionas/uso terapêutico
6.
J Pharmacol Exp Ther ; 320(3): 1002-12, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17167171

RESUMO

Thiazolidinediones (TZDs) are potent synthetic agonists of the ligand-activated transcription factor peroxisome proliferator-activated receptor-gamma (PPARgamma). TZDs were shown to induce neuroprotection after cerebral ischemia by blocking inflammation. As spinal cord injury (SCI) induces massive inflammation that precipitates secondary neuronal death, we currently analyzed the therapeutic efficacy of TZDs pioglitazone and rosiglitazone after SCI in adult rats. Both pioglitazone and rosiglitazone (1.5 mg/kg i.p.; four doses at 5 min and 12, 24, and 48 h) significantly decreased the lesion size (by 57 to 68%, p < 0.05), motor neuron loss (by 3- to 10-fold, p < 0.05), myelin loss (by 66 to 75%, p < 0.05), astrogliosis (by 46 to 61%, p < 0.05), and microglial activation (by 59 to 78%, p < 0.05) after SCI. TZDs significantly enhanced the motor function recovery (at 7 days after SCI, the motor scores were 37 to 45% higher in the TZD groups over the vehicle group; p < 0.05), but the treatment was effective only when the first injection was given by 2 h after SCI. At 28 days after SCI, chronic thermal hyperalgesia was decreased significantly (by 31 to 39%; p < 0.05) in the pioglitazone group compared with the vehicle group. At 6 h after SCI, the pioglitazone group showed significantly less induction of inflammatory genes [interleukin (IL)-6 by 83%, IL-1beta by 87%, monocyte chemoattractant protein-1 by 75%, intracellular adhesion molecule-1 by 84%, and early growth response-1 by 67%] compared with the vehicle group (p < 0.05 in all cases). Pioglitazone also significantly enhanced the post-SCI induction of neuroprotective heat shock proteins and antioxidant enzymes. Pretreatment with a PPARgamma antagonist, 2-chloro-5-nitro-N-phenyl-benzamide (GW9662), prevented the neuroprotection induced by pioglitazone.


Assuntos
Atividade Motora/efeitos dos fármacos , Neurônios Motores/efeitos dos fármacos , Bainha de Mielina/patologia , Fármacos Neuroprotetores/uso terapêutico , PPAR gama/agonistas , Dor/tratamento farmacológico , Traumatismos da Medula Espinal , Tiazolidinedionas/uso terapêutico , Animais , Sobrevivência Celular/efeitos dos fármacos , Citocinas/genética , Citocinas/imunologia , Modelos Animais de Doenças , Expressão Gênica/efeitos dos fármacos , Gliose/patologia , Gliose/prevenção & controle , Proteínas de Choque Térmico/genética , Mediadores da Inflamação/imunologia , Neurônios Motores/patologia , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Sprague-Dawley , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/patologia , Tiazolidinedionas/administração & dosagem , Tiazolidinedionas/farmacologia
7.
J Neurochem ; 98(5): 1353-68, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16923154

RESUMO

Increased levels of interleukin-6 (IL-6) play a role in post-ischemic cerebral inflammation. IL-6 binding to its receptors induces phosphorylation of the receptor associated janus kinases (JAKs), and the down-stream signal transducer and activator of transcription (STAT) family of transcription factors, which amplify the IL-6 signal transduction. We evaluated the functional significance of JAK2 and STAT3 activation in focal ischemia-induced neuronal damage. Transient middle cerebral artery occlusion in adult rats led to increased JAK2 and STAT3 phosphorylation in the ipsilateral cortex and striatum after 6-72 h of reperfusion. Fluorescent immunohistochemistry with cell specific markers (NeuN for neurons, glial fibrillary acidic protein for reactive astrocytes and ED1/OX42 for activated macrophages/microglia) showed that both pJAK2 and pSTAT3 staining is predominantly localized in the macrophages/microglia in the post-ischemic brain. Intracerebroventricular infusion of rats with AG490 (a JAK2 phosphorylation inhibitor) prevented the post-ischemic JAK2 and STAT3 phosphorylation and significantly decreased the infarct volume, number of apoptotic cells and neurological deficits, compared to vehicle control. Furthermore, intracerebral injection of siRNA specific for STAT3 led to curtailed STAT3 mRNA expression and phosphorylation, decreased infarct volume, fewer apoptotic cells and improved neurological function following transient middle cerebral artery occlusion. These studies show that JAK2-STAT3 activation plays a role in post-ischemic brain damage.


Assuntos
Ataque Isquêmico Transitório/patologia , Neurônios/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fator de Transcrição STAT3/metabolismo , Análise de Variância , Animais , Western Blotting/métodos , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Imuno-Histoquímica/métodos , Marcação In Situ das Extremidades Cortadas/métodos , Janus Quinase 2 , Proteínas do Tecido Nervoso/metabolismo , Exame Neurológico/métodos , Neurônios/efeitos dos fármacos , Neurônios/patologia , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Ratos , Reperfusão/métodos , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Fator de Transcrição STAT3/genética , Fatores de Tempo , Tirfostinas/farmacologia
8.
J Neurochem ; 98(6): 1718-31, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16899075

RESUMO

CCAAT/enhancer binding protein beta (C/EBPbeta) is a leucine-zipper transcription factor that regulates cell growth and differentiation in mammals. Expression of many pro-inflammatory genes including the cytokine interleukin-6 is known to be controlled by C/EBPbeta. We report that focal cerebral ischemia induced by transient middle cerebral artery occlusion (MCAO) significantly increases C/EBPbeta gene expression in mouse brain at between 6 and 72 h of reperfusion. To understand the functional significance of C/EBPbeta in postischemic inflammation and brain damage, we induced transient MCAO in cohorts of adult C/EBPbeta null mice and their wild-type littermates. At 3 days of reperfusion following transient MCAO, C/EBPbeta null mice showed significantly smaller infarcts, reduced neurological deficits, decreased terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling-positive cells, decreased intercellular adhesion molecule 1 (ICAM1) immunopositive vessels, decreased extravasated neutrophils and fewer activated microglia/macrophages, compared with their wild-type littermates. Furthermore, GeneChip analysis showed that postischemic induction of many transcripts known to promote inflammation and neuronal damage was less pronounced in the brains of C/EBPbeta-/- mice compared with C/EBPbeta+/+ mice. These results suggest a significant role for C/EBPbeta in postischemic inflammation and brain damage.


Assuntos
Encéfalo/patologia , Proteína beta Intensificadora de Ligação a CCAAT/deficiência , Encefalite/etiologia , Encefalite/patologia , Ataque Isquêmico Transitório/metabolismo , Ataque Isquêmico Transitório/patologia , Animais , Apoptose , Encéfalo/metabolismo , Proteína beta Intensificadora de Ligação a CCAAT/genética , Infarto Cerebral/metabolismo , Infarto Cerebral/patologia , Encefalite/genética , Expressão Gênica , Marcação In Situ das Extremidades Cortadas , Molécula 1 de Adesão Intercelular/metabolismo , Interleucina-6/genética , Ataque Isquêmico Transitório/complicações , Ataque Isquêmico Transitório/fisiopatologia , Camundongos , Camundongos Knockout , Doenças do Sistema Nervoso/etiologia , Doenças do Sistema Nervoso/prevenção & controle , Neurônios/patologia , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/metabolismo , Transcrição Gênica
9.
Neurochem Int ; 49(2): 127-35, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16759752

RESUMO

A brief ischemic insult induces significant protection against subsequent massive ischemic events. The molecular mechanisms underlying this phenomenon known as preconditioning (PC)-induced ischemic tolerance are not completely understood. Inflammation seen during the acute phase after stroke is known to be detrimental to the neurological outcome. We presently evaluated if the neuroprotective actions of PC involves prevention of post-ischemic inflammation. Cohorts of adult rats were subjected to transient focal ischemia (60 min middle cerebral artery occlusion; MCAO), PC (10 min MCAO) and focal ischemia followed 72 h after PC. Prior PC significantly reduced the post-ischemic increased expression of many inflammatory genes including cytokines, chemokines, adhesion molecules and pro-inflammatory transcription factors, and prevented the infiltration of neutrophils and macrophages in the ipsilateral cortex of rats subjected to focal ischemia. PC also decreased the volume of infarction and neurological dysfunction caused by transient focal ischemia. These studies indicate that prevention of inflammation might be a contributing mechanism by which PC induces protection against focal ischemia.


Assuntos
Isquemia Encefálica/imunologia , Isquemia Encefálica/terapia , Citoproteção/imunologia , Encefalite/imunologia , Encefalite/prevenção & controle , Precondicionamento Isquêmico/métodos , Animais , Encéfalo/irrigação sanguínea , Encéfalo/imunologia , Encéfalo/fisiopatologia , Infarto Encefálico/imunologia , Infarto Encefálico/fisiopatologia , Infarto Encefálico/prevenção & controle , Isquemia Encefálica/fisiopatologia , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Quimiocinas/genética , Quimiocinas/metabolismo , Quimiotaxia de Leucócito/genética , Quimiotaxia de Leucócito/imunologia , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Encefalite/fisiopatologia , Regulação da Expressão Gênica/imunologia , Infarto da Artéria Cerebral Média/imunologia , Infarto da Artéria Cerebral Média/fisiopatologia , Infarto da Artéria Cerebral Média/terapia , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Masculino , Ratos , Ratos Endogâmicos SHR , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
10.
Eur J Neurosci ; 23(5): 1172-88, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16553781

RESUMO

Identification of gene expression changes that promote focal neuronal death and neurological dysfunction can further our understanding of the pathophysiology of these disease states and could lead to new pharmacological and molecular therapies. Impairment of oxidative metabolism is a pathogenetic mechanism underlying neuronal death in many chronic neurodegenerative diseases as well as in Wernicke's encephalopathy (WE), a disorder induced by thiamine deficiency (TD). To identify functional pathways that lead to neuronal damage in this disorder, we have examined gene expression changes in the vulnerable thalamus and inferior colliculus of TD rats using Affymetrix Rat Genome GeneChip analysis in combination with gene ontology and functional categorization assessment utilizing the NetAffx GO Mining Tool. Of the 15 927 transcripts analysed, 125 in thalamus and 141 in inferior colliculus were more abundantly expressed in TD rats compared with control animals. In both regions, the major functional categories of transcripts that were increased in abundance after TD were those associated with inflammation (approximately 33%), stress (approximately 20%), cell death and repair ( approximately 26%), and metabolic perturbation (approximately 19%), together constituting approximately 98% of all transcripts up-regulated. These changes occurred against a background of neuronal cell loss and reactive astro- and microgliosis in both structures. Our results indicate that (i) TD produces changes in gene expression that are consistent with the observed dysfunction and pathology, and (ii) similar alterations in expression occur in thalamus and inferior colliculus, brain regions previously considered to differ in pathology. These findings provide important new insight into processes responsible for lesion development in TD, and possibly WE.


Assuntos
Expressão Gênica , Colículos Inferiores/fisiologia , Inflamação/genética , Estresse Oxidativo , Tálamo/fisiologia , Deficiência de Tiamina , Animais , Análise por Conglomerados , Perfilação da Expressão Gênica , Colículos Inferiores/citologia , Colículos Inferiores/patologia , Masculino , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Ratos Sprague-Dawley , Tálamo/citologia , Tálamo/patologia , Deficiência de Tiamina/imunologia , Deficiência de Tiamina/metabolismo , Deficiência de Tiamina/patologia , Deficiência de Tiamina/fisiopatologia
11.
Neurosurgery ; 57(6): 1254-63; discussion 1254-63, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16331174

RESUMO

OBJECTIVE: Epidermal growth factor (EGF) and fibroblast growth factor-2 (FGF-2) play a critical role in neurogenesis. In the present study, we evaluated the additive effect of administering these two factors on post-ischemic progenitor cell proliferation, survival, and phenotypic maturation in the hippocampal dentate gyrus (DG) and the subventricular zone (SVZ) in the adult rat brain after transient middle cerebral artery occlusion. METHODS: A combination of EGF+FGF-2 (each 1.44 ng/d) was continuously administered into the lateral ventricles for 3 days, 5-bromodeoxyuridine (BrdUrd) was injected (50 mg/Kg) twice daily for 3 days starting on Day 1 of reperfusion, and cohorts of rats were sacrificed on Day 5 and Day 21 of reperfusion. RESULTS: Compared with sham controls, ischemic rats showed a significantly higher number of newly proliferated cells in both the DG (by 766 +/- 37%, P < 0.05) and the SVZ (by 650 +/- 43%, P < 0.05). Of the progenitor cells proliferated on Day 5 after ischemia, 41 +/- 6% in the DG and 28 +/- 5% in the SVZ survived to 3 weeks. Compared with vehicle control, the EGF + FGF-2 infusion significantly increased the post-ischemic progenitor cell proliferation (by 319 +/- 40%, P < 0.05 in the DG and by 366 +/- 32%, P < 0.05 in the SVZ) and survival (by 40 +/- 12%, P < 0.05 in the DG and by 522 +/- 47%, P < 0.05 in the SVZ) studied at 5 and 21 days, respectively. Furthermore, of the newly proliferated cells survived to 3 weeks after ischemia, EGF + FGF-2 infusion caused a significantly higher number of neuronal nuclear protein-BrdUrd double-positive mature neurons in the DG (46 +/- 9%, P < 0.05) compared with vehicle control. Neuronal nuclear protein and BrdUrd double-positive mature neurons were also found in the DG. Glial fibrillary acidic protein-positive astrocytes did not show double-positive staining in either region. CONCLUSION: Specific growth factor infusion enhances post-ischemic progenitor cell proliferation by 5 days of reperfusion and neuronal maturation by 21 days of reperfusion in both the DG and SVZ in the adult rat brain.


Assuntos
Fator de Crescimento Epidérmico/farmacologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Ataque Isquêmico Transitório/patologia , Ataque Isquêmico Transitório/fisiopatologia , Neurônios/patologia , Células-Tronco/patologia , Animais , Arteriopatias Oclusivas/complicações , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Fator de Crescimento Epidérmico/administração & dosagem , Fator 2 de Crescimento de Fibroblastos/administração & dosagem , Injeções Intraventriculares , Ataque Isquêmico Transitório/etiologia , Masculino , Artéria Cerebral Média , Fenótipo , Ratos , Ratos Endogâmicos SHR
12.
Neurochem Int ; 47(8): 565-72, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16154234

RESUMO

Preconditioning (PC) is a phenomenon in which a brief ischemic insult induces tolerance against a subsequent severe ischemic insult. Recent studies showed that cerebral ischemia in adult rat upregulates progenitor cell proliferation in the hippocampal dentate gyrus. We presently evaluated whether PC can also stimulate progenitor cell proliferation in rat brain. Middle cerebral artery was transiently occluded in spontaneously hypertensive rats for 10 min to induce PC and 1h to induce focal ischemia. Progenitor cell proliferation (defined as BrdU(+) cell number) significantly increased after focal ischemia (by 3.9-fold; p<0.05) as well as PC (by 2.7-fold; p<0.05) compared to sham. PC 3 days prior had neither an inhibitory nor an additive effect on focal ischemia-induced progenitor cell proliferation. In both ischemia and PC groups, approximately 45% of the progenitor cells proliferated in week 1 survived to the end of week 3 and approximately 21% of those matured into NeuN(+) neurons. Furthermore, cerebral mRNA expression of the growth factors IGF1, FGF2, TGFbeta1, EGF and PDGF-A was significantly elevated after PC. Thus, we show that the beneficial effects of PC extend beyond providing neuroprotection during the acute phase after ischemia. Induction of growth factor expression and neurogenesis by PC might be a positive adaptation for an efficient repair and plasticity in the event of an ischemic insult.


Assuntos
Isquemia Encefálica/metabolismo , Infarto Cerebral/metabolismo , Substâncias de Crescimento/genética , Hipocampo/metabolismo , Precondicionamento Isquêmico , Plasticidade Neuronal/fisiologia , Animais , Isquemia Encefálica/fisiopatologia , Isquemia Encefálica/terapia , Bromodesoxiuridina , Diferenciação Celular/fisiologia , Proliferação de Células , Infarto Cerebral/fisiopatologia , Infarto Cerebral/prevenção & controle , Proteínas de Ligação a DNA , Modelos Animais de Doenças , Hipocampo/irrigação sanguínea , Hipocampo/fisiopatologia , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/fisiopatologia , Infarto da Artéria Cerebral Média/terapia , Masculino , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos SHR , Células-Tronco/metabolismo
13.
J Neurosurg ; 101(5): 799-805, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15540918

RESUMO

OBJECT: Throughout the life of a mammal, new neurons are produced each day from resident progenitor cells located in the hippocampal dentate gyrus (DG). The availability of transgenic and knockout mice enables the evaluation of specific molecular mediators of this phenomenon. To facilitate such studies the authors characterized the proliferation, survival, and maturation of progenitor cells in the DG of adult mice following transient focal cerebral ischemia. METHODS: Anesthesia was induced in adult C57BL/6 mice by administering halothane. The middle cerebral artery (MCA) was then occluded for 120 minutes by applying an endovascular suture. The marker used to detect the presence of proliferating cells, 5-bromodeoxyuridine (BrdU; 50 mg/kg) was administered intraperitoneally twice daily on Days 2 through 6 after the MCA occlusion. Cohorts of mice were killed on Days 7 and 21, after which their brains were sectioned and BrdU-positive cells were detected using immunohistochemical analysis. The phenotype of the BrdU-positive cells was identified by fluorescent triple labeling by using antibodies specific for neuronal and astroglial markers together with anti-BrdU antibodies. The infarction was confirmed by applying cresyl violet staining. Compared with sham-operated control animals, there was a 4.6-fold (p < 0.05) increase in BrdU-positive cells in the ipsilateral DG at Day 7 postischemia. Twenty-one percent of the newly proliferated cells survived to Day 21 postischemia. At this time, the newly proliferated cells expressed the immature and mature neuron markers doublecortin and NeuN, respectively, but none expressed the astroglial marker glial fibrillary acidic protein. CONCLUSIONS: Focal ischemia induces neurogenesis in the DG of the mouse brain; this may be critical for postischemic brain repair.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células , Giro Denteado/patologia , Ataque Isquêmico Transitório/fisiopatologia , Células-Tronco/fisiologia , Animais , Sobrevivência Celular/fisiologia , Giro Denteado/fisiopatologia , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/fisiopatologia , Ataque Isquêmico Transitório/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/fisiologia
14.
J Neurochem ; 89(1): 73-89, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15030391

RESUMO

In brain, a brief ischemic episode induces protection against a subsequent severe ischemic insult. This phenomenon is known as preconditioning-induced neural ischemic tolerance. An understanding of the molecular mechanisms leading to preconditioning helps in identifying potential therapeutic targets for preventing the post-stroke brain damage. The present study conducted the genomic and proteomic analysis of adult rat brain as a function of time following preconditioning induced by a 10-min transient middle cerebral artery (MCA) occlusion. GeneChip analysis showed induction of 40 putative neuroprotective transcripts between 3 to 72 h after preconditioning. These included heat-shock proteins, heme oxygenases, metallothioneins, signal transduction mediators, transcription factors, ion channels and apoptosis/plasticity-related transcripts. Real-time PCR confirmed the GeneChip data for the transcripts up-regulated after preconditioning. Two-dimensional gel electrophoresis combined with MALDI-TOF analysis showed increased expression of HSP70, HSP27, HSP90, guanylyl cyclase, muskelin, platelet activating factor receptor and beta-actin at 24 h after preconditioning. HSP70 protein induction after preconditioning was localized in the cortical pyramidal neurons. The infarct volume induced by focal ischemia (1-h MCA occlusion) was significantly smaller (by 38 +/- 7%, p < 0.05) in rats subjected to preconditioning 3 days before the insult. Preconditioning also prevented several gene expression changes induced by focal ischemia.


Assuntos
Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Perfilação da Expressão Gênica , Precondicionamento Isquêmico , Animais , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Isquemia Encefálica/patologia , Modelos Animais de Doenças , Regulação da Expressão Gênica/fisiologia , Genômica , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Proteômica , Ratos , Ratos Endogâmicos SHR , Transdução de Sinais/genética , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Fatores de Tempo , Regulação para Cima/genética
15.
Stroke ; 35(1): 179-84, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14657453

RESUMO

BACKGROUND AND PURPOSE: The present study was performed to determine whether antisense inhibition of intercellular adhesion molecule-1 (ICAM-1) protein expression decreases focal ischemic brain damage. METHODS: Male spontaneously hypertensive rats underwent 1-hour middle cerebral artery occlusion (MCAO) and 24-hour reperfusion. Rats were infused with ICAM-1 antisense or control oligodeoxynucleotides (ODNs) (48 nmol/d ICV) or vehicle, starting 24 hours before MCAO and continuing until the time of death. ICAM-1 and vascular cell adhesion molecule-1 (VCAM-1) mRNA levels were measured by real-time polymerase chain reaction. ICAM-1 protein knockdown was confirmed by Western blotting. Infarct volume was quantified by the use of cresyl violet-stained brain sections. Neurological deficits were evaluated. Mean arterial blood pressure was recorded by laser Doppler. Tissue penetration of antisense was confirmed by the use of fluorescent ODNs. RESULTS: Transient MCAO upregulated ICAM-1, but not VCAM-1, mRNA expression in the ipsilateral cortex between 3 and 72 hours of reperfusion. ICAM-1 antisense infusion prevented ischemia-induced ICAM-1 protein expression and reduced total infarct volume (by 53%; P<0.05; 226+/-35 mm3 in control ODN group and 104+/-27 mm3 in antisense ODN group; n=8 each) and mean neurological deficit score (by 44%; P<0.05; 2.4 in control ODN group and 1.3 in antisense ODN group; n=8 each). Neither control nor antisense ODN had any effect on mean arterial blood pressure and the physiological parameters monitored during MCAO. Compared with noninfused control, intracerebroventricular infusion of artificial cerebrospinal fluid or antisense or sense ODN had no significant effect on the regional cerebral blood flow changes that accompanied ischemia and reperfusion. CONCLUSIONS: Increased ICAM-1 expression is implicated in the pathogenesis of focal ischemia since ICAM-1 protein knockdown decreased ischemic brain damage. The mechanism by which ICAM-1 inhibition offers neuroprotection is independent of blood pressure modulation.


Assuntos
Expressão Gênica/efeitos dos fármacos , Infarto da Artéria Cerebral Média/tratamento farmacológico , Molécula 1 de Adesão Intercelular/biossíntese , Fármacos Neuroprotetores/farmacologia , Oligonucleotídeos Antissenso/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Infarto Cerebral/patologia , Infarto Cerebral/prevenção & controle , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Molécula 1 de Adesão Intercelular/efeitos dos fármacos , Molécula 1 de Adesão Intercelular/genética , Masculino , Exame Neurológico/efeitos dos fármacos , Fármacos Neuroprotetores/farmacocinética , Oligonucleotídeos Antissenso/farmacocinética , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos SHR , Reperfusão , Distribuição Tecidual , Resultado do Tratamento
16.
J Neurochem ; 87(3): 586-97, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14535942

RESUMO

Progenitor cells in the dentate gyrus of hippocampus (DG) and the subventricular zone of lateral ventricles (SVZ) generate new neurons throughout the life of mammals. Cerebral ischemia increases this basal progenitor cell proliferation. The present study evaluated the time frame of proliferation, length of survival and the phenotypes of the new cells formed after transient middle cerebral artery occlusion (MCAO) in adult spontaneously hypertensive rats. Compared to sham controls, ischemic rats showed a significantly higher number of newly proliferated cells (as defined by BrdU immunostaining) in both the DG (by fourfold, p < 0.05) and the SVZ (by twofold, p < 0.05). DG showed increased proliferation only in the first week of reperfusion and 49% of the cells formed in this period survived to the end of third week. Whereas, SVZ showed a continuous proliferation up to 3 weeks after MCAO, but the cells formed survived for less than a week. In both DG and SVZ, at the end of the first week of reperfusion, majority of the BrdU-positive (BrdU+) cells were immature neurons (DCX positive). In the DG, 28% of the cells formed in the first week after MCAO mature into neurons (NeuN positive). The ischemic cortex and striatum showed several BrdU+ cells which were ED-1 positive microglia/macrophages. At 1 week of reperfusion, MCAO-induced progenitor cell proliferation in the ipsilateral DG was significantly increased by i.c.v. infusion of IGF-1 (by 127 +/- 14%, p < 0.05) and GDNF (by 91 +/- 5%, p < 0.05), compared to vehicle. In the growth factor treated rats subjected to transient MCAO, several BrdU+ cells formed in the first week survived up to the third week.


Assuntos
Fator de Crescimento Insulin-Like I/farmacologia , Fatores de Crescimento Neural/farmacologia , Células-Tronco/patologia , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/patologia , Animais , Isquemia Encefálica/complicações , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/patologia , Diferenciação Celular , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular , Giro Denteado/efeitos dos fármacos , Giro Denteado/patologia , Modelos Animais de Doenças , Proteína Duplacortina , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Ventrículos Laterais/patologia , Masculino , Neurônios/efeitos dos fármacos , Neurônios/patologia , Ratos , Ratos Endogâmicos SHR , Reperfusão , Células-Tronco/efeitos dos fármacos , Acidente Vascular Cerebral/complicações
17.
J Neurosci Res ; 71(2): 208-19, 2003 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-12503083

RESUMO

Proper CNS function depends on concerted expression of thousands of genes in a controlled and timely manner. Traumatic brain injury (TBI) in mammals results in neuronal death and neurological dysfunction, which might be mediated by altered expression of several genes. By employing a CNS-specific GeneChip and real-time polymerase chain reaction (PCR), the present study analyzed the gene expression changes in adult rat cerebral cortex in the first 24 hr after a controlled cortical impact injury. Many functional families of genes not previously implicated in TBI-induced brain damage are altered in the injured cortex. These include up-regulated transcription factors (SOCS-3, JAK-2, STAT-3, CREM, IRF-1, SMN, silencer factor-B, ANIA-3, ANIA-4, and HES-1) and signal transduction pathways (cpg21, Narp, and CRBP) and down-regulated transmitter release mechanisms (CITRON, synaptojanin II, ras-related rab3, neurexin-1beta, and SNAP25A and -B), kinases (IP-3-kinase, Pak1, Ca(2+)/CaM-dependent protein kinases), and ion channels (K(+) channels TWIK, RK5, X62839, and Na(+) channel I). In addition, several genes previously shown to play a role in TBI pathophysiology, including proinflammatory genes, proapoptotic genes, heat shock proteins, immediate early genes, neuropeptides, and glutamate receptor subunits, were also observed to be altered in the injured cortex. Real-time PCR analysis confirmed the GeneChip data for many of these transcripts. The novel physiologically relevant gene expression changes observed here might explain some of the molecular mechanisms of TBI-induced neuronal damage.


Assuntos
Lesões Encefálicas/genética , Córtex Cerebral/metabolismo , Expressão Gênica , Animais , Lesões Encefálicas/metabolismo , Lesões Encefálicas/fisiopatologia , Córtex Cerebral/lesões , Córtex Cerebral/fisiopatologia , Análise por Conglomerados , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Ataque Isquêmico Transitório/etiologia , Ataque Isquêmico Transitório/metabolismo , Ataque Isquêmico Transitório/fisiopatologia , Masculino , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Regulação para Cima
18.
J Neurochem ; 83(5): 1072-86, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12437578

RESUMO

Identification of novel modulators of ischemic neuronal death helps in developing new strategies to prevent the stroke-induced neurological dysfunction. Hence, the present study evaluated the gene expression changes in rat cerebral cortex at 6 and 24 h of reperfusion following transient middle cerebral artery occlusion (MCAO) by GeneChip analysis. Transient MCAO resulted in selective increased mRNA levels of genes involved in stress, inflammation, transcription and plasticity, and decreased mRNA levels of genes which control neurotransmitter function and ionic balance. In addition to a number of established ischemia-related genes, many genes not previously implicated in transient focal ischemia-induced brain damage [suppressor of cytokine signaling (SOCS)-3, cAMP responsive element modulator (CREM), cytosolic retinol binding protein (CRBP), silencer factor-B, survival motor neuron (SMN), interferon-gamma regulatory factor-1 (IRF-1), galanin, neurotrimin, proteasome subunit RC8, synaptosomal-associated protein (SNAP)-25 A and B, synapsin 1a, neurexin 1-beta, ras-related rab3, vesicular GABA transporter (VGAT), digoxin carrier protein, neuronal calcium sensor-1 and neurodap] were observed to be altered in the ischemic cortex. Real-time PCR confirmed the GeneChip results for several of these transcripts. SOCS-3 is a gene up-regulated after ischemia which modulates inflammation by controlling cytokine levels. Antisense knockdown of ischemia-induced SOCS-3 protein expression exacerbated transient MCAO-induced infarct volume assigning a neuroprotective role to SOCS-3, a gene not heretofore implicated in ischemic neuronal damage.


Assuntos
Córtex Cerebral/metabolismo , Perfilação da Expressão Gênica , Hipertensão/genética , Hipertensão/metabolismo , Ataque Isquêmico Transitório/metabolismo , Proteínas Repressoras , Animais , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Modulador de Elemento de Resposta do AMP Cíclico , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Galanina/genética , Galanina/metabolismo , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/metabolismo , Ataque Isquêmico Transitório/etiologia , Ataque Isquêmico Transitório/patologia , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Oligonucleotídeos Antissenso/farmacologia , Reação em Cadeia da Polimerase , Proteínas/antagonistas & inibidores , Proteínas/genética , Proteínas/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos SHR , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA