Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Pflugers Arch ; 471(11-12): 1455-1466, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31664513

RESUMO

Reperfusion therapy is currently the gold standard treatment for acute ischemic stroke. However, reperfusion injuries such as oedema and haemorrhagic transformation largely limit the use of this potent treatment to a narrow time window. Recently, transient receptor potential melastatin 4 (TRPM4) channel has emerged as a potential target for vascular protection in stroke management. Non-specificity and side effects are major concerns for current TRPM4 blockers. The present study was undertaken to develop a novel TRPM4 blocker for stroke management. We report the generation of a TRPM4-specific antibody M4P which binds to a region close to the channel pore. M4P could inhibit TRPM4 current and downregulate TRPM4 surface expression, therefore prevent hypoxia-induced cell swelling. In the rat model of 3-h stroke reperfusion, application of M4P at 2 h after occlusion ameliorated reperfusion injury by improving blood-brain barrier integrity, and enhanced functional recovery. Our results demonstrate that TRPM4 blockade could attenuate reperfusion injury in stroke recanalization. When applied together with reperfusion treatments, TRPM4 blocking antibody has the potential to extend the therapeutic time window for acute ischemic stroke.


Assuntos
Anticorpos Monoclonais/farmacologia , Traumatismo por Reperfusão/tratamento farmacológico , Acidente Vascular Cerebral/tratamento farmacológico , Canais de Cátion TRPM/antagonistas & inibidores , Animais , Barreira Hematoencefálica/metabolismo , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/metabolismo , Modelos Animais de Doenças , Feminino , Masculino , Ratos , Ratos Wistar , Traumatismo por Reperfusão/metabolismo , Acidente Vascular Cerebral/metabolismo , Regulação para Cima/efeitos dos fármacos
3.
Transl Stroke Res ; 10(1): 91-103, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-29569041

RESUMO

The transient receptor potential melastatin 4 (TRPM4) channel has been suggested to play a key role in the treatment of ischemic stroke. However, in vivo evaluation of TRPM4 channel, in particular by direct channel suppression, is lacking. In this study, we used multimodal imaging to assess edema formation and quantify the amount of metabolically functional brain salvaged after a rat model of stroke reperfusion. TRPM4 upregulation in endothelium emerges as early as 2 h post-stroke induction. Expression of TRPM4 channel was suppressed directly in vivo by treatment with siRNA; scrambled siRNA was used as a control. T2-weighted MRI suggests that TRPM4 inhibition successfully reduces edema by 30% and concomitantly salvages functionally active brain, measured by 18F-FDG-PET. These in vivo imaging results correlate well with post-mortem 2,3,5-triphenyltetrazolium chloride (TTC) staining which exhibits a 34.9% reduction in infarct volume after siRNA treatment. Furthermore, in a permanent stroke model, large areas of brain tissue displayed both edema and significant reductions in metabolic activity which was not shown in transient models with or without TRPM4 inhibition, indicating that tissue salvaged by TRPM4 inhibition during stroke reperfusion may survive. Evans Blue extravasation and hemoglobin quantification in the ipsilateral hemisphere were greatly reduced, suggesting that TRPM4 inhibition can improve BBB integrity after ischemic stroke reperfusion. Our results support the use of TRPM4 blocker for early stroke reperfusion.


Assuntos
Regulação da Expressão Gênica/fisiologia , Infarto da Artéria Cerebral Média/tratamento farmacológico , Imagem Multimodal/métodos , Traumatismo por Reperfusão/tratamento farmacológico , Canais de Cátion TRPM/metabolismo , Animais , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/fisiopatologia , Edema Encefálico , Modelos Animais de Doenças , Fluordesoxiglucose F18/farmacocinética , Lateralidade Funcional , Processamento de Imagem Assistida por Computador , Infarto da Artéria Cerebral Média/complicações , Masculino , Análise em Microsséries , Fosfopiruvato Hidratase/metabolismo , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/uso terapêutico , Ratos , Ratos Wistar , Traumatismo por Reperfusão/complicações , Canais de Cátion TRPM/antagonistas & inibidores , Canais de Cátion TRPM/genética , Fator de von Willebrand/metabolismo
4.
Sci Rep ; 6: 23964, 2016 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-27050297

RESUMO

Cerebral hypoperfusion and impaired autophagy are two etiological factors that have been identified as being associated with the development of Alzheimer's disease (AD). Nevertheless, the exact relationships among these pathological processes remain unknown. To elucidate the impact of cerebral hypoperfusion in AD, we created a unilateral common carotid artery occlusion (UCCAO) model by occluding the left common carotid artery in both young and old 3xTg-AD mice. Two months after occlusion, we found that ligation increases phospho-Tau (p-Tau) at Serine 199/202 in the hippocampus of 3-month-old AD mice, compared to sham-operated AD mice; whereas, there is no change in the wild type (WT) mice after ligation. Moreover, cerebral hypoperfusion led to significant increase of p-Tau in both the hippocampus and cortex of 16-month-old AD mice and WT mice. Notably, we did not detect any change in Aß42 level in either young or old AD and WT mice after ligation. Interestingly, we observed a downregulation of LC3-II in the cortex of aged AD mice and WT mice after ligation. Our results suggest that elevated p-Tau and reduced autophagy are major cellular changes that are associated with hypoperfusion in AD. Therefore, targeting p-Tau and autophagy pathways may ameliorate hypoperfusion-induced brain damage in AD.


Assuntos
Doença de Alzheimer/metabolismo , Autofagia , Isquemia Encefálica/metabolismo , Proteínas tau/metabolismo , Fatores Etários , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Isquemia Encefálica/genética , Doença Crônica , Modelos Animais de Doenças , Feminino , Lobo Frontal/metabolismo , Hipocampo/metabolismo , Humanos , Immunoblotting , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Fragmentos de Peptídeos/metabolismo , Fosforilação
5.
J Biol Chem ; 290(14): 9262-72, 2015 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-25694430

RESUMO

L-type Cav1.2 Ca(2+) channel undergoes extensive alternative splicing, generating functionally different channels. Alternatively spliced Cav1.2 Ca(2+) channels have been found to be expressed in a tissue-specific manner or under pathological conditions. To provide a more comprehensive understanding of alternative splicing in Cav1.2 channel, we systematically investigated the splicing patterns in the neonatal and adult rat hearts. The neonatal heart expresses a novel 104-bp exon 33L at the IVS3-4 linker that is generated by the use of an alternative acceptor site. Inclusion of exon 33L causes frameshift and C-terminal truncation. Whole-cell electrophysiological recordings of Cav1.233L channels expressed in HEK 293 cells did not detect any current. However, when co-expressed with wild type Cav1.2 channels, Cav1.233L channels reduced the current density and altered the electrophysiological properties of the wild type Cav1.2 channels. Interestingly, the truncated 3.5-domain Cav1.233L channels also yielded a dominant negative effect on Cav1.3 channels, but not on Cav3.2 channels, suggesting that Cavß subunits is required for Cav1.233L regulation. A biochemical study provided evidence that Cav1.233L channels enhanced protein degradation of wild type channels via the ubiquitin-proteasome system. Although the physiological significance of the Cav1.233L channels in neonatal heart is still unknown, our report demonstrates the ability of this novel truncated channel to modulate the activity of the functional Cav1.2 channels. Moreover, the human Cav1.2 channel also contains exon 33L that is developmentally regulated in heart. Unexpectedly, human exon 33L has a one-nucleotide insertion that allowed in-frame translation of a full Cav1.2 channel. An electrophysiological study showed that human Cav1.233L channel is a functional channel but conducts Ca(2+) ions at a much lower level.


Assuntos
Processamento Alternativo , Canais de Cálcio Tipo L/genética , Miocárdio/metabolismo , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Sequência de Bases , Canais de Cálcio Tipo L/química , DNA , Primers do DNA , Éxons , Masculino , Dados de Sequência Molecular , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Behav Brain Res ; 275: 252-8, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25239606

RESUMO

Cerebral ischemia is one of the leading causes of death and long-term disability in aging populations, due to the frequent occurrence of irreversible brain damage and subsequent loss of neuronal function which lead to cognitive impairment and some motor dysfunction. In the present study, the real time course of motor and cognitive functions were evaluated following the chronic cerebral ischemia induced by permanent, bilateral occlusion of the common carotid arteries (PBOCCA). Male Sprague Dawley rats (200-300g) were subjected to PBOCCA or sham-operated surgery and tested 1, 2, 3 and 4 weeks following the ischemic insult. The results showed that PBOCCA significantly reduced step-through latency in a passive avoidance task at all time points when compared to the sham-operated group. PBOCCA rats also showed significant increase in escape latencies during training in the Morris water maze, as well as a reduction of the percentage of times spend in target quadrant of the maze at all time points following the occlusion. Importantly, there were no significant changes in locomotor activity between PBOCCA and sham-operated groups. The BDNF expression in the hippocampus was 29.3±3.1% and 40.1±2.6% on day 14 and 28 post PBOCCA, respectively compared to sham-operated group. Present data suggest that the PBOCCA procedure effectively induces behavioral, cognitive symptoms associated with cerebral ischemia and, consequently, provides a valuable model to study ischemia and related neurodegenerative disorder such as Alzheimer's disease and vascular dementia.


Assuntos
Isquemia Encefálica/complicações , Transtornos Cognitivos/etiologia , Transtornos dos Movimentos/etiologia , Animais , Isquemia Encefálica/etiologia , Isquemia Encefálica/patologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Estenose das Carótidas/complicações , Modelos Animais de Doenças , Reação de Fuga/fisiologia , Hipocampo/metabolismo , Hipocampo/patologia , Masculino , Aprendizagem em Labirinto/fisiologia , Atividade Motora , Ratos , Ratos Sprague-Dawley , Tempo de Reação/fisiologia , Fatores de Tempo
7.
Pflugers Arch ; 466(3): 563-76, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24043570

RESUMO

Transient receptor potential melastatin 4 (TRPM4) is a voltage-dependent, nonselective cation channel. Under pathological conditions, sustained activation of TRPM4 leads to oncotic cell death. Here, we report the upregulation of TRPM4 in vascular endothelium following hypoxia/ischemia in vitro and in vivo. In human umbilical vein endothelial cells, TRPM4 expression was increased at both the mRNA and protein levels following oxygen-glucose deprivation. Blocking TRPM4 with 9-phenanthrol greatly enhanced tube formation on Matrigel. In a rat permanent middle cerebral artery occlusion model, TRPM4 was upregulated in the vascular endothelium within the penumbra region after stroke. TRPM4 expression peaked 1 day post-occlusion and gradually decreased. In vivo siRNA-mediated TRPM4 silencing enhanced angiogenesis and improved capillary integrity. A twofold reduction in infarct volume and a substantial recovery of motor function were observed in animals receiving the siRNA treatment. Interestingly, the protective effect of TRPM4 suppression disappeared 5 days after stroke induction, indicating that TRPM4 upregulation is critical for cerebral damage during the acute phase of stroke. TRPM4 could be a potential therapeutic target for ischemic stroke.


Assuntos
Infarto da Artéria Cerebral Média/metabolismo , Neovascularização Fisiológica , Canais de Cátion TRPM/metabolismo , Animais , Hipóxia Celular , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Terapia Genética , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Infarto da Artéria Cerebral Média/fisiopatologia , Infarto da Artéria Cerebral Média/terapia , Locomoção , Masculino , Fenantrenos/farmacologia , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Ratos , Ratos Wistar , Canais de Cátion TRPM/antagonistas & inibidores , Canais de Cátion TRPM/genética , Regulação para Cima
8.
Transl Stroke Res ; 4(5): 477-83, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24098313

RESUMO

Immunotherapy represents an active area of biomedical research to treat cancer, autoimmune diseases, and neurodegenerative disorders. In stroke, recanalization therapy is effective in reducing brain tissue damage after acute ischemic stroke. However, the narrow time window restricts its application for the majority of stroke patients. There is an urgent need to develop adjuvant therapies such as immunotherapy, stem cell replacement, and neuroprotective drugs. A number of molecules have been targeted for immunotherapy in stroke management, including myelin-associated proteins and their receptors, N-methyl-d-aspartic acid receptors, cytokines, and cell adhesion molecules. Both active vaccination and passive antibodies were tested in animal models of acute ischemic stroke. However, the mechanisms underlying the efficacy of immunotherapy are different for each target protein. Blocking myelin-associated proteins may enhance neuroplasticity, whereas blocking adhesion molecules may yield neuroprotection by suppressing the immune response after stroke. Although results from animal studies are encouraging, clinical trials using therapeutic antibodies failed to improve stroke outcome due to severe side effects. It remains a challenge to generate specific therapeutic antibodies with minimal side effects on other organs and systems.


Assuntos
Anticorpos/farmacologia , Moléculas de Adesão Celular/antagonistas & inibidores , Imunoterapia/métodos , Proteínas da Mielina/antagonistas & inibidores , Acidente Vascular Cerebral , Animais , Pesquisa Biomédica , Citocinas/metabolismo , Humanos , Fatores Imunológicos/farmacologia , Fatores Imunológicos/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...