Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
FASEB J ; 31(8): 3383-3392, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28420695

RESUMO

Cyclin-dependent kinase 5 (Cdk5), which binds to and is activated by p35, phosphorylates multiple substrates and plays an essential role in the development and function of the CNS; however, proteolytic production of p25 from p35 under stress conditions leads to the inappropriate activation of Cdk5 and contributes to hyperphosphorylation of τ and other substrates that are related to the pathogenesis of Alzheimer's disease. Selective inhibition of aberrant Cdk5 activity via genetic overexpression of Cdk5 inhibitory peptide (CIP) reduces pathologic changes and prevents brain atrophy and memory loss in p25-transgenic mice. In the present study, we delivered adeno-associated virus 9 carrying green fluorescent protein-CIP (AAV9-GFP-CIP) to brain cells via intracerebroventricular infusion in amyloid precursor protein/presenilin 1 (APP/PS1) double-transgenic 3-mo-old mice after the occurrence of ß-amyloid (Aß) aggregation and the hyperphosphorylation of τ. Three months of treatment of AAV9-GFP-CIP reduced pathologic changes, including τ hyperphosphorylation, (Aß) deposit, astrocytosis, and microgliosis, which were correlated with the reversal of memory loss and anxiety-like behavior observed in APP/PS1 mice. The neuroprotection effect of AAV9-GFP-CIP lasted an additional 7 mo-the end point of the study. These findings provide a novel strategy to selectively target Cdk5 for the treatment of Alzheimer's disease.-He, Y., Pan, S., Xu, M., He, R., Huang, W., Song, P., Huang, J., Zhang, H.-T., Hu, Y. Adeno-associated virus 9-mediated Cdk5 inhibitory peptide reverses pathologic changes and behavioral deficits in the Alzheimer's disease mouse model.


Assuntos
Doença de Alzheimer/terapia , Proteínas do Tecido Nervoso/genética , Fragmentos de Peptídeos/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides , Animais , Apoptose , Comportamento Animal , Encéfalo/patologia , Dependovirus/fisiologia , Regulação da Expressão Gênica , Terapia Genética , Vetores Genéticos , Proteínas de Fluorescência Verde , Células HEK293 , Humanos , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Fragmentos de Peptídeos/metabolismo , Fosforilação , Presenilina-1/genética , Presenilina-1/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
2.
J Alzheimers Dis ; 53(3): 843-55, 2016 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-27258419

RESUMO

BACKGROUND: The transgenic mice models overexpressing human p25 contribute greatly to the in vivo neurotoxic mechanism of p25 in neurodegenerative diseases. However, it is time-consuming to manipulate existing transgenic mice models. OBJECTIVE: Here we aim to establish a novel mouse model of neurodegeneration by overexpressing p25 mediated by recombinant adeno-associated virus serotype 9 (rAAV9). METHODS: AAV9-GFP-p25 encoding GFP-fused p25 driven by synapsin promoter, and the control, AAV9-GFP, were delivered in mice by tail-vein injection. Assessments of p25 expression, neurodegenerative pathology, and behavioral changes were performed. RESULTS: GFP expression was detected by in vivo imaging as early as one week after virus injection. Notably, widespread expression of p25 was obviously found in cortex, hippocampus, and cerebellum in AAV9-GFP-p25 mice. Moreover, decreased hippocampus volumes in AAV9-GFP-p25 mice were detected by 7T MRI examination about one month after injection. Further, these AAV9-GFP-p25 mice exhibited progressive memory impairment from three-month to six-month after virus injection. At last, hyperphosphorylated tau, neurofibrillary tangles, activated astrocytes and microglia cells were elevated in these p25 mice at about six months after virus delivery. However, amyloid-ß plaques, overt neuronal loss, and apoptosis in the hippocampus and cortex were not significantly induced by AAV9-mediated p25 overexpression. CONCLUSION: The AAV9-mediated p25 overexpression mouse model, which is a practical model exhibiting neurodegeneration-like pathological and behavioral changes, provides an easier and time-saving method to explore the functions of p25 in vivo, as well as an alternative tool for development of drugs against neurotoxic of p25.


Assuntos
Encéfalo/patologia , Modelos Animais de Doenças , Transtornos Mentais/etiologia , Proteínas do Tecido Nervoso/metabolismo , Doenças Neurodegenerativas , Animais , Encéfalo/metabolismo , Dependovirus/genética , Comportamento Exploratório/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Marcação In Situ das Extremidades Cortadas , Imageamento por Ressonância Magnética , Aprendizagem em Labirinto , Transtornos Mentais/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora , Proteínas do Tecido Nervoso/genética , Doenças Neurodegenerativas/complicações , Doenças Neurodegenerativas/diagnóstico por imagem , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Estatísticas não Paramétricas , Sinapsinas/genética , Sinapsinas/metabolismo
3.
Neurosci Lett ; 632: 1-7, 2016 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-27542341

RESUMO

Parkinson's disease (PD) is pathologically characterized by progressively loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) and the formation of Lewy bodies. In 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) induced PD mice models, the calpain- cyclin-dependent kinase 5 (Cdk5)-myocyte enhancer factor 2 (MEF2) signaling has been proven in governing dopaminergic neuronal death. Under MPTP insult, p35 is cleaved by calpain into p25, which binds to Cdk5 and exhibits hyperactivity of Cdk5/p25. Cdk5/p25 inactivates MEF2, a survivor factor, which is critical for DA neuronal death. In this study, neuroprotective effect of the Cdk5/p25 specific peptide, TFP5, was evaluated in sub-acute MPTP induced PD mouse model by intraperitoneal (i.p.) injection of MPTP for five consecutive days. The results indicated that the levels of p35 and p25, and p25/p35 ratio increased in the sub-acute MPTP mice. TFP5 broadly reached cortex neuron, hippocampus and SNpc areas after i.p. injections. Pretreatment with 45mg/kg/day TFP5, as well as 10mgkg/day Cdk5 inhibitor roscovitine, for three days significantly rescued DA neuronal loss up to 9.8% or 9.7% respectively compared to the saline treated group. Treatment of TFP5 and roscovitine reduced the levels of inactive form of MEF2 and cleaved caspase 3, thus protected apoptosis of DA neurons against MPTP insult. Our results propose that TFP5 might be a potential therapeutic candidate for PD.


Assuntos
Morte Celular/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Quinase 5 Dependente de Ciclina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Transtornos Parkinsonianos/patologia , Animais , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Hipocampo/metabolismo , Hipocampo/patologia , Camundongos , Fármacos Neuroprotetores/farmacologia , Transtornos Parkinsonianos/metabolismo
4.
Sci Rep ; 4: 7091, 2014 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-25404538

RESUMO

Co-treatment of neuroprotective reagents may improve the therapeutic efficacy of hypothermia in protecting neurons during ischemic stroke. This study aimed to find promising drugs that enhance the neuroprotective effect of mild hypothermia (MH). 26 candidate drugs were selected based on different targets. Primary cultured cortical neurons were exposed to oxygen-glucose deprivation and reoxygenation (OGD/R) to induce neuronal damage, followed by either single treatment (a drug or MH) or a combination of a drug and MH. Results showed that, compared with single treatment, combination of MH with brain derived neurotrophic factor, glibenclamide, dizocilpine, human urinary kallidinogenase or neuroglobin displayed higher proportion of neuronal cell viability. The latter three drugs also caused less apoptosis rate in combined treatment. Furthermore, co-treatment of those three drugs and MH decreased the level of reactive oxygen species (ROS) and intracellular calcium accumulation, as well as stabilized mitochondrial membrane potential (MMP), indicating the combined neuroprotective effects are probably via inhibiting mitochondrial apoptosis pathway. Taken together, the study suggests that combined treatment with hypothermia and certain neuroprotective reagents provide a better protection against OGD/R-induced neuronal injury.


Assuntos
Maleato de Dizocilpina/farmacologia , Globinas/farmacologia , Calicreínas/farmacologia , Proteínas do Tecido Nervoso/farmacologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Temperatura Baixa , Terapia Combinada , Meios de Cultura/química , Glucose/deficiência , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neuroglobina , Neurônios/metabolismo , Neurônios/patologia , Oxigênio/farmacologia , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/terapia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa