Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Neurobiol Dis ; 54: 24-31, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23473743

RESUMO

Increased brain infiltration of polymorphonuclear neutrophils (PMNs) occurs early after stroke and is important in eliciting brain inflammatory response during stroke recovery. In order to understand the molecular mechanism of PMN entry, we investigated the expression and requirement for Slit1, a chemorepulsive guidance cue, and its cognate receptor, Robo1, in a long-term recovery mouse model of cerebral ischemia. The expression levels of Robo1 were significantly decreased bilaterally at 24h following reperfusion. Robo1 expression levels remained suppressed in the ipsilateral cortex until 28d post MCAO-reperfusion, while the levels of Robo1 in the contralateral cortex recovered to the level of sham-operated mouse by 7d reperfusion. Circulating PMNs express high levels of Slit1, but not Robo1. Influx of PMNs into the ischemic core area occurred early (24h) after cerebral ischemia, when endothelial Robo1 expression was significantly reduced in the ischemic brain, indicating that Robo1 may form a repulsive barrier to PMN entry into the brain parenchyma. Indeed, blocking Slit1 on PMNs in a transwell migration assay in combination with an antibody blocking of Robo1 on human umbilical vein endothelial cells (HUVEC) significantly increased PMN transmigration during oxygen glucose deprivation, an in vitro model of ischemia. Collectively, in the normal brain, the presence of Slit1 on PMNs, and Robo1 on cerebral endothelial cells, generated a repulsive force to prevent the infiltration of PMNs into the brain. During stroke recovery, a transient reduction in Robo1 expression on the cerebral endothelial cells allowed the uncontrolled infiltration of Slit1-expressing PMNs into the brain causing inflammatory reactions.


Assuntos
Encéfalo/metabolismo , Quimiotaxia de Leucócito/fisiologia , Células Endoteliais/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neutrófilos/metabolismo , Receptores Imunológicos/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Western Blotting , Encéfalo/imunologia , Modelos Animais de Doenças , Células HEK293 , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/imunologia , Acidente Vascular Cerebral/imunologia , Transfecção , Proteínas Roundabout
2.
J Neurochem ; 122(4): 764-74, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22681613

RESUMO

Microglia are the 'immune cells' of the brain and their activation plays a vital role in the pathogenesis of many neurodegenerative diseases. Activated microglia produce high levels of pro-inflammatory factors, such as TNFα, causing neurotoxicity. Here we show that vimentin played a key role in controlling microglia activation and neurotoxicity during cerebral ischemia. Deletion of vimentin expression significantly impaired microglia activation in response to LPS in vitro and transient focal cerebral ischemia in vivo. Reintroduction of the functional vimentin gene back into vimentin knockout microglia restored their response to LPS. More importantly, impairment of microglia activation significantly protected brain from cerebral ischemia-induced neurotoxicity. Collectively, we demonstrate a previously unknown function of vimentin in controlling microglia activation.


Assuntos
Isquemia Encefálica/patologia , Ativação de Macrófagos/fisiologia , Microglia/fisiologia , Vimentina/fisiologia , Animais , Western Blotting , Encéfalo/patologia , Separação Celular , Técnica Indireta de Fluorescência para Anticorpo , Processamento de Imagem Assistida por Computador , Marcação In Situ das Extremidades Cortadas , Infarto da Artéria Cerebral Média/patologia , Ataque Isquêmico Transitório/patologia , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/efeitos dos fármacos , Microscopia Confocal , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/fisiopatologia , Plasmídeos/efeitos dos fármacos , Plasmídeos/genética , Traumatismo por Reperfusão/patologia , Tetraciclina/farmacologia , Vimentina/genética
3.
J Biol Chem ; 285(13): 9908-9918, 2010 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-20133938

RESUMO

Neuropilins (NRPs) are receptors for the major chemorepulsive axonal guidance cue semaphorins (Sema). The interaction of Sema3A/NRP1 during development leads to the collapse of growth cones. Here we show that Sema3A also induces death of cultured cortical neurons through NRP1. A specific NRP1 inhibitory peptide ameliorated Sema3A-evoked cortical axonal retraction and neuronal death. Moreover, Sema3A was also involved in cerebral ischemia-induced neuronal death. Expression levels of Sema3A and NRP1, but not NRP2, were significantly increased early during brain reperfusion following transient focal cerebral ischemia. NRP1 inhibitory peptide delivered to the ischemic brain was potently neuroprotective and prevented the loss of motor functions in mice. The integrity of the injected NRP1 inhibitory peptide into the brain remained unchanged, and the intact peptide permeated the ischemic hemisphere of the brain as determined using MALDI-MS-based imaging. Mechanistically, NRP1-mediated axonal collapse and neuronal death is through direct and selective interaction with the cytoplasmic tyrosine kinase Fer. Fer RNA interference effectively attenuated Sema3A-induced neurite retraction and neuronal death in cortical neurons. More importantly, down-regulation of Fer expression using Fer-specific RNA interference attenuated cerebral ischemia-induced brain damage. Together, these studies revealed a previously unknown function of NRP1 in signaling Sema3A-evoked neuronal death through Fer in cortical neurons.


Assuntos
Neuropilina-1/metabolismo , Proteínas Tirosina Quinases/metabolismo , Semaforina-3A/química , Animais , Encéfalo/metabolismo , Isquemia Encefálica/patologia , Morte Celular , Regulação Enzimológica da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Neuropilina-1/química , Peptídeos/química , Ligação Proteica , Interferência de RNA , Transdução de Sinais
4.
Biochem Biophys Res Commun ; 401(3): 435-9, 2010 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-20869945

RESUMO

Brain microglia are resident macrophage-like cells representing the first and main form of active immune response during brain injury. Microglia-mediated inflammatory events in the brain are known to be associated with chronic degenerative diseases such as Multiple Sclerosis, Parkinson's, or Alzheimer's disease. Therefore, identification of mechanisms activating microglia is not only important in the understanding of microglia-mediated brain pathologies, but may also lead to the development of new anti-inflammatory drugs for the treatment of chronic neurodegenerative diseases. Recently, abscisic acid (ABA), a phytohormone regulating important physiological functions in higher plants, has been proposed to activate murine microglial cell line N9 through increased intracellular calcium. In the present study, we determined the response to ABA and its analogues from murine primary microglia and immortalized murine microglial cell line BV-2 and N9 cells. A Fura-2-acetoxymethyl ester (Fura-2AM)-based ratiometric calcium imaging and measurement technique was used to determine the intracellular calcium changes in these cells when treated with (-)-ABA, (+)-ABA, (-)-trans-ABA and (+)-trans-ABA. Both primary microglia and microglial cell lines (BV-2 and N9 cells) showed significant increase in intracellular calcium ([Ca(2+)]i) in response to treatment with ATP and ionomycine. However, ABAs failed to evoke dose- and time-dependent [Ca(2+)]i changes in mouse primary microglia, BV-2 and N9 cells. Together, these surprising findings demonstrate that, contrary to that reported in N9 cells [3], ABAs do not evoke intracellular calcium changes in primary microglia and microglial cell lines. The broad conclusion that ABA evokes [Ca(2+)]i in microglia requires more evidence and further careful examination.


Assuntos
Ácido Abscísico/análogos & derivados , Ácido Abscísico/farmacologia , Cálcio/metabolismo , Microglia/efeitos dos fármacos , Animais , Linhagem Celular Transformada , Camundongos , Microglia/metabolismo
5.
Mol Cell Biol ; 27(5): 1696-705, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17178835

RESUMO

The nuclear transcription factor E2F1 plays an important role in modulating neuronal death in response to excitotoxicity and cerebral ischemia. Here, by comparing gene expression in brain cortices from E2F1(+/+) and E2F1(-/-) mice using a custom high-density DNA microarray, we identified a group of putative E2F1 target genes that might be responsible for ischemia-induced E2F1-dependent neuronal death. Neuropilin 1 (NRP-1), a receptor for semaphorin 3A-mediated axon growth cone collapse and retraction, was confirmed to be a direct target of E2F1 based on (i) the fact that the NRP-1 promoter sequence contains an E2F1 binding site, (ii) reactivation of NRP-1 expression in E2F1(-/-) neurons when the E2F1 gene was replaced, (iii) activation of the NRP-1 promoter by E2F1 in a luciferase reporter assay, (iv) electrophoretic mobility gel shift analysis confirmation of the presence of an E2F binding sequence in the NRP-1 promoter, and (v) the fact that a chromatin immunoprecipitation assay showed that E2F1 binds directly to the endogenous NRP-1 promoter. Interestingly, the temporal induction in cerebral ischemia-induced E2F1 binding to the NRP-1 promoter correlated with the temporal-induction profile of NRP-1 mRNA, confirming that E2F1 positively regulates NRP-1 during cerebral ischemia. Functional analysis also showed that NRP-1 receptor expression was extremely low in E2F1(-/-) neurons, which led to the diminished response to semaphorin 3A-induced axonal shortening and neuronal death. An NRP-1 selective peptide inhibitor provided neuroprotection against oxygen-glucose deprivation. Taken together, these findings support a model in which E2F1 targets NRP-1 to modulate axonal damage and neuronal death in response to cerebral ischemia.


Assuntos
Morte Encefálica/patologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Fator de Transcrição E2F1/genética , Neurônios/metabolismo , Neuropilina-1/metabolismo , Adenoviridae/genética , Animais , Isquemia Encefálica/etiologia , Células Cultivadas , Cerebelo/citologia , Imunoprecipitação da Cromatina , Fator de Transcrição E2F1/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Genes Reporter , Luciferases/análise , Luciferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Modelos Biológicos , Neuroglia/citologia , Neurônios/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Exp Cell Res ; 315(16): 2856-68, 2009 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-19559021

RESUMO

Collapsin response mediator proteins (CRMPs) are key modulators of cytoskeletons during neurite outgrowth in response to chemorepulsive guidance molecules. However, their roles in adult injured neurons are not well understood. We previously demonstrated that CRMP3 underwent calcium-dependent N-terminal protein cleavage during excitotoxicity-induced neurite retraction and neuronal death. Here, we report findings that the full-length CRMP3 inhibits tubulin polymerization and neurite outgrowth in cultured mature cerebellar granule neurons, while the N-terminal truncated CRMP3 underwent nuclear translocation and caused a significant nuclear condensation. The N-terminal truncated CRMP3 underwent nuclear translocation through nuclear pores. Nuclear protein pull-down assay and mass spectrometry analysis showed that the N-terminal truncated CRMP3 was associated with nuclear vimentin. In fact, nuclear-localized CRMP3 co-localized with vimentin during glutamate-induced excitotoxicity. However, the association between the truncated CRMP3 and vimentin was not critical for nuclear condensation and neurite outgrowth since over-expression of truncated CRMP3 in vimentin null neurons did not alleviate nuclear condensation and neurite outgrowth inhibition. Together, these studies showed CRMP3's role in attenuating neurite outgrowth possibility through inhibiting microtubule polymerization, and also revealed its novel association with vimentin during nuclear condensation prior to neuronal death.


Assuntos
Calpaína/metabolismo , Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuritos/metabolismo , Isoformas de Proteínas/metabolismo , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Humanos , Camundongos , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Neurônios/metabolismo , Isoformas de Proteínas/genética , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Vimentina/metabolismo
7.
J Neurochem ; 111(3): 870-81, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19735446

RESUMO

Intracellular calcium influx through NMDA receptors triggers a cascade of deleterious signaling events which lead to neuronal death in neurological conditions such as stroke. However, it is not clear as to the molecular mechanism underlying early damage response from axons and dendrites which are important in maintaining a network essential for the survival of neurons. Here, we examined changes of axons treated with glutamate and showed the appearance of betaIII-tubulin positive varicosities on axons before the appearance of neuronal death. Dizocilpine blocked the occurrence of varicosities on axons suggesting that these microstructures were mediated by NMDA receptor activities. Despite early increased expression of pCaMKII and pMAPK after just 10 min of glutamate treatment, only inhibitors to Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) and calpain prevented the occurrence of axonal varicosities. In contrast, inhibitors to Rho kinase, mitogen-activated protein kinase and phosphoinositide 3-kinase were not effective, nor were they able to rescue neurons from death, suggesting CaMKII and calpain are important in axon survival. Activated CaMKII directly phosphorylates collapsin response mediator protein (CRMP) 2 which is independent of calpain-mediated cleavage of CRMP2. Over-expression of CRMP2, but not the phosphorylation-resistant mutant CRMP2-T555A, increased axonal resistance to glutamate toxicity with reduced numbers of varicosities. The levels of both pCRMP2 and pCaMKII were also increased robustly within early time points in ischemic brains and which correlated with the appearance of axonal varicosities in the ischemic neurons. Collectively, these studies demonstrated an important role for CaMKII in modulating the integrity of axons through CRMP2 during excitotoxicity-induced neuronal death.


Assuntos
Axônios/efeitos dos fármacos , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Ácido Glutâmico/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Morte Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Modelos Animais de Doenças , Maleato de Dizocilpina/farmacologia , Embrião de Mamíferos , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Proteínas de Fluorescência Verde/genética , Infarto da Artéria Cerebral Média/patologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Proteínas do Tecido Nervoso/genética , Neurônios/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Semaforina-3A/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transfecção/métodos , Tubulina (Proteína)/metabolismo
8.
Biochem Biophys Res Commun ; 390(1): 115-20, 2009 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-19782044

RESUMO

Basic fibroblast growth factor (bFGF) is a known neuroprotectant against a number of brain injury conditions such as cerebral ischemia. However, bFGF also regulates a plethora of brain developmental processes and functions as a strong mitogen. Therefore, unregulated long-term expression of bFGF in brain may potentially be tumorigenic, limiting its utility in brain therapy. Here, we report the successful construction of an adenoviral vector (Ad-5HRE-bFGF) expressing bFGF under the regulation of five hypoxia-responsive elements (5HRE) and a minimal cytomegalovirus promoter (CMVmp). Following hypoxia treatment in a hypoxic chamber with less than 1% of oxygen, Ad-5HRE-bFGF induced a significant and time-dependent expression of bFGF protein and the fluorescent tag, humanized GFP (hrGFP) protein, in infected PC12 cells. In contrast, normoxia treatment evoked extremely low level of bFGF and hrGFP expression, demonstrating that the 5HRE-CMVmp cassette was effective in regulating the expression of bFGF gene in response to hypoxia. More importantly, bFGF expressed by the Ad-5HRE-bFGF viral vector under the regulation of hypoxia was significantly neuroprotective against PC12 cell death evoked by serum deprivation. Taken together, these studies demonstrated the feasibility to express bFGF in a hypoxia-regulated fashion to provide neuroprotection. The Ad-5HRE-bFGF can be further developed as an effective tool to provide neuroprotection against hypoxia-induced brain diseases, such as cerebral ischemia.


Assuntos
Citoproteção/genética , Fator 2 de Crescimento de Fibroblastos/biossíntese , Regulação da Expressão Gênica , Vetores Genéticos , Neurônios/metabolismo , Adenoviridae , Animais , Apoptose , Hipóxia Celular/genética , Citomegalovirus , Fator 2 de Crescimento de Fibroblastos/genética , Humanos , Células PC12 , Ratos , Elementos de Resposta
9.
J Clin Invest ; 115(7): 1828-38, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15937550

RESUMO

Inhibitors of HIV protease have been shown to have antiapoptotic effects in vitro, yet whether these effects are seen in vivo remains controversial. In this study, we have evaluated the impact of the HIV protease inhibitor (PI) nelfinavir, boosted with ritonavir, in models of nonviral disease associated with excessive apoptosis. In mice with Fas-induced fatal hepatitis, Staphylococcal enterotoxin B-induced shock, and middle cerebral artery occlusion-induced stroke, we demonstrate that PIs significantly reduce apoptosis and improve histology, function, and/or behavioral recovery in each of these models. Further, we demonstrate that both in vitro and in vivo, PIs block apoptosis through the preservation of mitochondrial integrity and that in vitro PIs act to prevent pore function of the adenine nucleotide translocator (ANT) subunit of the mitochondrial permeability transition pore complex.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores da Protease de HIV/farmacologia , Translocases Mitocondriais de ADP e ATP/antagonistas & inibidores , Animais , Anticorpos/administração & dosagem , Modelos Animais de Doenças , Feminino , Hepatite/tratamento farmacológico , Hepatite/patologia , Humanos , Células Jurkat , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Translocases Mitocondriais de ADP e ATP/química , Modelos Moleculares , Nelfinavir/farmacologia , Ritonavir/farmacologia , Choque Séptico/tratamento farmacológico , Choque Séptico/patologia , Transdução de Sinais/efeitos dos fármacos , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/patologia
10.
Biochem Biophys Res Commun ; 367(1): 109-15, 2008 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-18162177

RESUMO

Strategies to provide neuroprotection and to promote regenerative axonal outgrowth in the injured brain are thwarted by the plethora of axon growth inhibitors and the ligand promiscuity of some of their receptors. Especially, new neurons derived from ischemia-stimulated neurogenesis must integrate this multitude of inhibitory molecular cues, generated as a result of cortical damage, into a functional response. More often than not the response is one of growth cone collapse, axonal retraction and neuronal death. Therefore, characterization of the expression of inhibitory molecules in long-term surviving ischemic brains following stroke is important for designing selective therapeutics. Here, we describe a long-term recovery mouse model for cerebral ischemia in which a brief transient occlusion of the middle cerebral artery (30min) was followed by up to 30 days of long-term reperfusion. Significantly decreased grip strength motor function and increased expression of one of the major repulsive guidance cues, Semaphorin 3A (Sema3A) and its receptor Neuropilin1 (NRP1) occurred in brains of these mice. Interestingly, increased Doublecortin (DCX) expression occurred only in the lateral ventricular wall zone, but not in the dentate gyrus granule cell layer on the ischemic side of the brain. Importantly, no DCX positive cells were detected in the infarct core region after 30d ischemic recovery. Collectively, these studies demonstrated the sustained elevation of Sema3A/NRP1 expression in the ischemic territory, which may contribute to the inhibitory microenvironment responsible for preventing new neurons from entering the infarct area. This model will be of use as a platform for testing anti-inhibitory therapies to stroke.


Assuntos
Isquemia Encefálica , Regulação da Expressão Gênica/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Neuropeptídeos/metabolismo , Neuropilina-1/metabolismo , Semaforina-3A/metabolismo , Animais , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Isquemia Encefálica/terapia , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Regulação da Expressão Gênica/genética , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/genética , Modelos Biológicos , Neuropeptídeos/genética , Neuropilina-1/genética , Semaforina-3A/genética , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/terapia , Fatores de Tempo , Resultado do Tratamento , Regulação para Cima/genética , Regulação para Cima/fisiologia
11.
J Neurosci ; 26(8): 2241-9, 2006 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-16495451

RESUMO

Collapsin response mediator proteins (CRMPs) mediate growth cone collapse during development, but their roles in adult brains are not clear. Here we report the findings that the full-length CRMP-3 (p63) is a direct target of calpain that cleaves CRMP-3 at the N terminus (+76 amino acid). Interestingly, activated calpain in response to excitotoxicity in vitro and cerebral ischemia in vivo also cleaved CRMP-3, and the cleavage product of CRMP-3 (p54) underwent nuclear translocation during neuronal death. The expression of p54 was colocalized with the terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end labeling-positive nuclei in glutamate-treated cerebellar granule neurons (CGNs) and in ischemic neurons located in the infarct core after focal cerebral ischemia, suggesting that p54 might be involved in neuronal death. Overexpression studies showed that p54, but not p63, caused death of human embryonic kidney cells and CGNs, whereas knock-down CRMP-3 expression by selective small interfering RNA protected neurons against glutamate toxicity. Collectively, these results reveal a novel role of CRMP-3 in that calpain cleavage of CRMP-3 and the subsequent nuclear translocation of the truncated CRMP-3 evokes neuronal death in response to excitotoxicity and cerebral ischemia. Our findings also establish a novel route of how calpain signals neuron death.


Assuntos
Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Calpaína/metabolismo , Ácido Glutâmico/toxicidade , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Animais , Apoptose/efeitos dos fármacos , Sítios de Ligação , Encéfalo/efeitos dos fármacos , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/patologia , Ligação Proteica
12.
Neurosci Lett ; 419(1): 23-7, 2007 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-17418946

RESUMO

Selective gene expression targeting neurons is a challenge, which, if successfully overcome, carries an enormous potential for clinical applications in therapeutics against neurodegenerative diseases. We have reported previously the construction of a series of adenoviral vectors capable of selectively expressing a reporter gene luciferase in cultured neurons [D. Huang, A. Desbois, S.T. Hou, A novel adenoviral vector which mediates hypoxia-inducible gene expression selectively in neurons, Gene Ther. 12 (2005) 1369-1376]. A combination of neuron restrictive silencer elements (NRSEs), hypoxia responsive elements (HREs) and CMV minimal promoter (CMVmp) was packaged into replication defective adenovirus to target gene expression selectively in neurons in a hypoxia-regulated manner. In the present study, we injected the adenoviral vectors into the neonatal mouse brain followed by treatment with hypoxia. The expression of the reporter luciferase gene was examined by luciferase assay and fluorescent immunostaining. Neurons and glial cells were identified by staining with antibodies against NeuN and GFAP, respectively. Remarkably, in response to hypoxia, Ad/5HRE-3NRSE showed strong hypoxia-inducible gene expression of the reporter luciferase selectively in neurons but not in glial cells. In contrast, brains infected with the control vector Ad/5HRE showed no selectivity in luciferase expression (in both neurons and glial cells) under the hypoxic condition. Taken together, these studies demonstrated that this vector (Ad/5HRE-3NRSE) can mediate gene expression selectively in neurons both in vitro and in vivo, supporting the suggestion that further refinement of this vector may lead to the development of a useful tool to investigate mechanisms of neuronal damage following cerebral ischemia and a possible effective gene therapy vector to stroke.


Assuntos
Encéfalo/metabolismo , Citomegalovirus/fisiologia , Expressão Gênica/fisiologia , Vetores Genéticos/administração & dosagem , Hipóxia/patologia , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Animais , Animais Recém-Nascidos , Linhagem Celular Transformada , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Hipóxia/fisiopatologia , Luciferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neuroglia/metabolismo , Fosfopiruvato Hidratase/metabolismo
13.
Neurol Res ; 38(6): 553-9, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27320251

RESUMO

OBJECTIVES: To investigate whether curcumin regulates Notch signaling to cause neuroprotection and neurogenesis after focal ischemia reperfusion injury. METHOD: Focal ischemia reperfusion injury was modeled in rats by occluding the middle cerebral artery. These animals were given either curcumin (300 mg/kg) or corn oil (vehicle) by intraperitoneal injection starting 1 h after stroke and continuing for 7 d. In parallel, sham-operated control animals received vehicle. All animals were killed on day 12. The different treatment groups were compared in terms of neurobehavioral deficits, BrdU incorporation, and levels of doublecortin (DCX) and Notch intracellular domain (NICD) using immunohistochemistry, immunofluorescence and Western blotting. RESULTS: Animals treated with curcumin showed significantly smaller neurobehavioral deficits than vehicle-treated animals after 3, 7, and 12 d of reperfusion (all p < 0.05). Tissue sections from curcumin-treated animals contained significantly greater numbers of BrdU-positive cells (p < 0.05) and BrdU/DCX-positive cells (p < 0.01), as well as significantly higher NICD levels (p < 0.01). CONCLUSION: Curcumin may protect from focal cerebral ischemia reperfusion injury as well as stimulate neurogenesis by activating the Notch signaling pathway.


Assuntos
Encéfalo/efeitos dos fármacos , Curcumina/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Receptores Notch/metabolismo , Transdução de Sinais/efeitos dos fármacos , Acidente Vascular Cerebral/tratamento farmacológico , Análise de Variância , Animais , Bromodesoxiuridina/metabolismo , Contagem de Células , Ventrículos Cerebrais/citologia , Ventrículos Cerebrais/efeitos dos fármacos , Modelos Animais de Doenças , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Masculino , Proteínas Associadas aos Microtúbulos/metabolismo , Doenças do Sistema Nervoso/tratamento farmacológico , Doenças do Sistema Nervoso/etiologia , Neurogênese/efeitos dos fármacos , Exame Neurológico , Neuropeptídeos/metabolismo , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/metabolismo , Fatores de Tempo
14.
Int Rev Cytol ; 221: 93-148, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12455747

RESUMO

The mode of neuronal death caused by cerebral ischemia and reperfusion appears on the continuum between the poles of catastrophic necrosis and apoptosis: ischemic neurons exhibit many biochemical hallmarks of apoptosis but remain cytologically necrotic. The position on this continuum may be modulated by the severity of the ischemic insult. The ischemia-induced neuronal death is an active process (energy dependent) and is the result of activation of cascades of detrimental biochemical events that include perturbion of calcium homeostasis leading to increased excitotoxicity, malfunction of endoplasmic reticulum and mitochondria, elevation of oxidative stress causing DNA damage, alteration in proapoptotic gene expression, and activation of the effector cysteine proteases (caspases) and endonucleases leading to the final degradation of the genome. In spite of strong evidence showing that brain infarction can be reduced by inhibiting any one of the above biochemical events, such as targeting excitotoxicity, up-regulation of an antiapoptotic gene, or inhibition of a down-stream effector caspase, it is becoming clear that targeting a single gene or factor is not sufficient for stroke therapeutics. An effective neuroprotective therapy is likely to be a cocktail aimed at all of the above detrimental events evoked by cerebral ischemia and the success of such therapeutic intervention relies upon the complete elucidation of pathways and mechanisms of the cerebral ischemia-induced active neuronal death.


Assuntos
Isquemia Encefálica/metabolismo , Morte Celular/genética , Degeneração Neural/metabolismo , Traumatismo por Reperfusão/metabolismo , Animais , Isquemia Encefálica/genética , Isquemia Encefálica/fisiopatologia , Dano ao DNA/genética , Regulação da Expressão Gênica/genética , Humanos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Degeneração Neural/genética , Degeneração Neural/fisiopatologia , Neurotoxinas/metabolismo , Estresse Oxidativo/genética , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/fisiopatologia
15.
Brain Res ; 1036(1-2): 27-34, 2005 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-15725398

RESUMO

The growth-arrest-specific protein gas7 is required for morphological differentiation of cultured mouse cerebellar neurons and PC12 cells. Moreover, its overexpression in various cell types induces neurite-like outgrowth. The role of gas7 in neuronal differentiation was further characterized by adenovirus-mediated overexpression in PC12 cells and quantification of the expression of various neuronal markers, in the absence and presence of different concentrations of nerve growth factor (NGF). The potential neuroprotective activity of gas7 against various neurotoxic insults was also assessed. In addition to promoting the formation of neurite-like extensions, overexpression of gas7 potentiated NGF-mediated neuronal differentiation of PC12 cells, as shown by the enhanced expression of the neuronal proteins betaIII-tubulin, synaptotagmin, alpha7 subunit of the acetylcholine receptor, and dihydropyrimidinase related protein-3. This effect was exerted independently of cell cycle progression, as gas7 did not affect proliferation of PC12 cells. While some differentiation enhancers protect PC12 cells against lethal insults, gas7 overexpression in PC12 cells did not protect against oxygen-glucose deprivation, the calcium ionophore A23187, or the nitric oxide donor sodium nitroprusside, suggesting that gas7 is not neuroprotective. The ability of gas7 to potentiate neuronal differentiation makes it a potential therapeutic target to promote re-establishment of neuronal connections in the injured or diseased brain, such as following stroke.


Assuntos
Diferenciação Celular/fisiologia , Fator de Crescimento Neural/farmacologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Células-Tronco/metabolismo , Animais , Biomarcadores/metabolismo , Cálcio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Hipóxia Celular/fisiologia , Vetores Genéticos , Ionóforos/farmacologia , Camundongos , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/metabolismo , Neurotoxinas/antagonistas & inibidores , Neurotoxinas/metabolismo , Doadores de Óxido Nítrico/farmacologia , Células PC12 , Ratos , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Transfecção
16.
Neurosci Lett ; 377(2): 97-100, 2005 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-15740844

RESUMO

The present study investigated the role of pyridoxal phosphate-6-azophenyl-2',4'-disulfonic acid tetrasodium salt (PPADS), a P2 receptor antagonist, in protecting mouse cerebellar granule neurons (CGNs) against glutamate/NMDA-induced neuronal death. Neurotoxicity caused by 50 microM glutamate or 200 microM NMDA was significantly reduced in CGNs treated with PPADS. Such neuroprotection was in a time- and dose-dependent manner. The possibility that PPADS may block glutamate/NMDA-mediated intracellular Ca2+ influx to CGNs was investigated using temperature-controlled platereader measurements of fluorescence intensity of CGNs loaded with Ca2+-sensitive fluorescent dye Fluo-4AM. Interestingly, the rapid increase of calcium influx following glutamate/NMDA treatment was not significantly affected by prior treatment with PPADS. In contrast, MK801, a specific NMDA receptor antagonist, completely blocked intracellular Ca2+ influx. Taken together, these data suggest that inhibition of the P2 receptor may directly modulate NMDA receptor-mediated neurotoxicity through a Ca2+-independent mechanism.


Assuntos
Ácido Glutâmico/toxicidade , N-Metilaspartato/toxicidade , Fármacos Neuroprotetores/farmacologia , Antagonistas do Receptor Purinérgico P2 , Fosfato de Piridoxal/análogos & derivados , Fosfato de Piridoxal/farmacologia , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Camundongos , Receptores Purinérgicos P2/fisiologia
17.
J Mol Neurosci ; 24(2): 323-8, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15456946

RESUMO

Expression of therapeutic gene products in differentiated human NT2 neurons (NT2/Ns) is being explored for ex vivo gene therapy of human neurological diseases. In this study we determined the efficiency of adenovirus (Ad)-mediated gene delivery into NT2/Ns and characterized the expression of several key receptors known to be required for efficient Ad-mediated gene delivery. Undifferentiated NT2 cells and NT2/Ns were infected by Ad expressing green fluorescent protein at an efficiency of 33% and 17%, respectively percentages much lower than the 92% infectivity obtained from a human non-neuronal cell line A549 cells. This relatively low infectivity of NT2/Ns might be caused by the extremely low expression of integrin subunit beta3 and the reduced expression of beta5 during differentiation. The expression of coxsackie-Ad receptor (CAR) was relatively high and remained constant during differentiation. Blocking CAR receptor using an antibody specific against CAR reduced Ad infectivity in a dose-dependent manner. These observations suggest that modulating the expression of integrin subunits beta3/5 or the functional heterodimer alphavbeta3/5 in human NT2/Ns may enhance adenoviral infectivity of NT2/Ns.


Assuntos
Técnicas de Transferência de Genes , Cadeias beta de Integrinas/metabolismo , Integrina beta3/metabolismo , Neurônios/metabolismo , Receptores Virais/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Diferenciação Celular , Linhagem Celular , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Humanos , Cadeias beta de Integrinas/genética , Integrina beta3/genética , Neurônios/citologia , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Receptores Virais/genética
18.
Brain Res Mol Brain Res ; 116(1-2): 70-9, 2003 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-12941462

RESUMO

The full mechanisms underlying neuronal death following excitotoxic insult remain unclear, despite many in vivo and in vitro studies. Recent work has focused on various signaling molecules and pathways, normally strictly regulated, that can trigger death if perturbed. The transcription factor, E2F1 is pivotal in controlling cell death under stress situations. The current study aimed to investigate the role of this transcription factor in modulating neuronal death following kainic acid (KA) treatment of cultured mouse cerebellar granule cells (CGCs). KA-induced death of CGCs was attenuated by the selective KA/AMPA receptor antagonist CNQX, but not MK-801. Such neuronal death was caspase-3-independent and did not activate many known death genes, such as Fas receptor, caspase-8 and p38. However, hyperphosphorylation of Rb showed a transient increase which may lead to activation of E2F1. Indeed E2F1 +/+ and -/- CGCs showed a differential response to KA-mediated toxicity, in that E2F1 -/- neurons were significantly less susceptible to KA compared to E2F1 +/+ neurons, albeit both E2F1 +/+ and -/- neurons expressed similar levels of KA receptors and responded similarly to kainate antagonist, CNQX. Using selective inhibitors to CDKs, such as olomoucine, roscovitine and flavopiridol, and the inhibitor SB203580 to p38 MAPK, we ruled out the possibility that Rb inactivation through hyperphosphorylation was due to either upstream kinases. Therefore activation of Rb/E2F1 pathway appears to involve novel interactions yet to be elucidated.


Assuntos
Proteínas de Ciclo Celular , Morte Celular/efeitos dos fármacos , Cerebelo/citologia , Proteínas de Ligação a DNA , Agonistas de Aminoácidos Excitatórios/toxicidade , Ácido Caínico/toxicidade , Neurônios/metabolismo , Proteína do Retinoblastoma/metabolismo , Fatores de Transcrição/metabolismo , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Animais , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Maleato de Dizocilpina/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Fatores de Transcrição E2F , Fator de Transcrição E2F1 , Inibidores Enzimáticos/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Imuno-Histoquímica , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , RNA Mensageiro/biossíntese , Receptores de Ácido Caínico/classificação , Receptores de Ácido Caínico/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo
19.
Neuroreport ; 15(14): 2241-4, 2004 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-15371742

RESUMO

The role of B group vitamins in preventing neuronal death against excitotoxicity was investigated. Neuronal death of cultured mouse cerebellar granule neurons (CGNs) caused by glutamate (50 microM) or NMDA (200 microM) was delayed in CGNs that had been treated with riboflavin (B2), folic acid (B9) or cynocobalamin (B12) for 18 h. Such neuroprotection by B2, B9 and B12 was in a dose- and time-dependent manner. In contrast, application of thiamin (B1), nicotinamide (B3), d-pantothenic acid (B5), pyridoxine (B6) or carnitine (BT) did not ameliorate glutamate or NMDA-mediated excitotoxicity to CGCs. These results are the first indication that certain B group vitamins are not only required for the normal brain function, but can also play a protective role against excitotoxicity to the brain.


Assuntos
Cerebelo/efeitos dos fármacos , Ácido Glutâmico/toxicidade , N-Metilaspartato/toxicidade , Fármacos Neuroprotetores/farmacologia , Complexo Vitamínico B/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Células Cultivadas , Cerebelo/patologia , Relação Dose-Resposta a Droga , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/patologia , Riboflavina/farmacologia , Vitamina B 12/farmacologia
20.
J Neurosci Methods ; 138(1-2): 39-44, 2004 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-15325109

RESUMO

Targeting of postmitotic neurons selectively for gene delivery poses a challenge. One way to achieve such a selective targeting is to link the gene delivery vector with small ligand-binding polypeptides which have selective affinity to intact neurons. In order to identify such novel neuron selective polypeptides, we screened a phage-display library displaying random 12-mer polypeptides and subtractively bio-panned for clones having selectivity towards cultured mouse cerebellar granule neurons. The selected phage clones were amplified and sequenced. Affinities of these clones to neurons were determined by the visible presence or absence of fluorescence of phage particles as detected by immunocytochemistry using an antibody to M-13 phage. This affinity was further qualified by how much phage was bound, and where in or on the cell it tended to accumulate. The selectivity of binding to neurons was determined by the negative binding of these clones to several cultured non-neuronal cells, including, primary glial cells, NT2 cells, human embryonic kidney 293 cells, neuroblastoma cells, and mouse 3T3 cells. Among the 46 clones that we have sequenced and characterized, four clones appeared to have excellent selectivity in binding to neurons. Homology comparison of these polypeptides revealed that three of them contained a consensus D(E)-W(F)-I(N)-D-W motif. This motif was also present in the Bdm1 gene product which was predominantly expressed in postnatal brains. Further characterizations of these polypeptides are required to reveal the utilities of these peptides to function as an effective linker to facilitate gene transfer selectively to neurons.


Assuntos
Cerebelo/citologia , Neurônios/efeitos dos fármacos , Biblioteca de Peptídeos , Peptídeos/farmacologia , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Proteínas de Bactérias/metabolismo , Bacteriófago M13/genética , Bacteriófago M13/imunologia , Bacteriófago M13/metabolismo , Células Cultivadas , Clonagem Molecular , DNA de Cadeia Simples/síntese química , DNA de Cadeia Simples/química , Humanos , Imuno-Histoquímica/métodos , Camundongos , Mimetismo Molecular/fisiologia , Dados de Sequência Molecular , Neurônios/metabolismo , Peptídeos/isolamento & purificação , Ligação Proteica/fisiologia , Proteínas Virais de Fusão/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA