Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
2.
Biochem Cell Biol ; 96(4): 391-406, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29370536

RESUMO

Traumatic brain injury (TBI) is a leading cause of morbidity and mortality worldwide. Due to its high incidence rate and often long-term sequelae, TBI contributes significantly to increasing costs of health care expenditures annually. Unfortunately, advances in the field have been stifled by patient and injury heterogeneity that pose a major challenge in TBI prevention, diagnosis, and treatment. In this review, we briefly discuss the causes of TBI, followed by its prevalence, classification, and pathophysiology. The current imaging detection methods and animal models used to study brain injury are examined. We discuss the potential use of molecular markers in detecting and monitoring the progression of TBI, with particular emphasis on microRNAs as a novel class of molecular modulators of injury and its repair in the neural tissue.


Assuntos
Biomarcadores/análise , Lesões Encefálicas Traumáticas , Neuroimagem Funcional , MicroRNAs/uso terapêutico , Animais , Encéfalo/diagnóstico por imagem , Lesões Encefálicas Traumáticas/diagnóstico , Lesões Encefálicas Traumáticas/diagnóstico por imagem , Lesões Encefálicas Traumáticas/terapia , Modelos Animais de Doenças , Humanos
3.
J Alzheimers Dis ; 57(4): 1265-1279, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28372324

RESUMO

Neuroinflammation plays a critical role in neuronal dysfunction and death of Alzheimer's disease (AD). ApoE4 is a major risk factor of AD, while ApoE2 is neuroprotective. Little is known about the roles of ApoE isoforms in the neuroinflammation seen in AD. Their roles and mechanisms in Aß-induced/neuroinflammation were investigated in this study using in vivo and in vitro models. Rat astrocytes were treated with lipid-poor recombinant hApoE and/or Aß42. Mouse astrocyte lines-expressing lipidated hApoE were treated with Aß42 and/or vitamin D receptor (VDR) agonist, 1α,25-dihydroxyvitamin D3. Cells and media were harvested for cytokine ELISA, RNA isolated for qRT-PCR, and nuclear protein for transcription factor (TF) arrays and EMSA. hApoE-transgenic and AD mice were mated to generate hApoE2/AD and hApoE4/AD mice. Mice were euthanized at 6 months of age. Brain tissues were collected for cytokine ELISA array, Aß ELISA, immunoblotting, and immunohistochemistry. hApoE4/AD mice had significantly higher levels of inflammatory cytokines than hApoE2/AD mice. Lipidated hApoE4 significantly promoted inflammatory gene expression induced by Aß42 but not recombinant hApoE4 in astrocytes as compared to controls. Lipidated hApoE3 provided a certain degree of protection against Aß42-induced inflammatory response but not recombinant hApoE3 as compared to controls. Both lipidated and recombinant hApoE2 provided protection against Aß42-induced inflammatory response compared to controls. TF array revealed that ApoE2 strongly activated VDR in Aß42-treated astrocytes. Application of 1α,25-dihydroxyvitamin D3 completely inhibited Aß-induced inflammatory gene expression in hApoE4-expressing astrocytes. The results suggest that ApoE4 promotes, but ApoE2 inhibits, AD/Aß-induced neuroinflammation via VDR signaling. Targeting VDR signaling or active form of VD3 may relieve AD neuroinflammation or/and neurodegeneration.


Assuntos
Doença de Alzheimer/imunologia , Apolipoproteínas E/metabolismo , Astrócitos/imunologia , Encéfalo/imunologia , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/administração & dosagem , Peptídeos beta-Amiloides/metabolismo , Animais , Apolipoproteínas E/administração & dosagem , Apolipoproteínas E/genética , Astrócitos/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Células Cultivadas , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Neuroimunomodulação/efeitos dos fármacos , Neuroimunomodulação/fisiologia , Neuroproteção/efeitos dos fármacos , Neuroproteção/fisiologia , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/metabolismo , Isoformas de Proteínas , Ratos Sprague-Dawley , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/metabolismo , Vitamina D/análogos & derivados , Vitamina D/farmacologia
4.
J Alzheimers Dis ; 53(3): 875-92, 2016 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-27314524

RESUMO

Alzheimer's disease (AD) is characterized by extracellular deposits of amyloid-ß (Aß) in the brain. ABCA7 is highly expressed in the brain and a susceptibility gene for late-onset AD (LOAD). The minor alleles at two ABCA7 single-nucleotide polymorphisms (SNPs), rs3764650 (T>G; intron13) and rs3752246 at a predicted myristoylation site (C>G; exon33; p.Gly1527Ala), are significantly associated with LOAD risk; however, the mechanism of this association is unknown. Functional consequences of both SNPs were examined in HEK293 and CHO cells stably expressing AßPPSwe. Luciferase reporter assays in HEK293 cells suggested that intron13 carrying rs3764650 major T-allele (int13-T) possessed promoter-enhancing capabilities. Co-transfection experiments with hABCA7 and int13-T resulted in significantly increased ABCA7 protein level relative to that with int13-G. Expression of hABCA7 carrying rs3752246 risk allele led to increases in secreted Aß40 and Aß42 and ß-secretase activity in CHO- and HEK-AßPPSwe cells. Hydroxymyristic acid treatment of cells expressing hABCA7 carrying the rs3752246 major G allele resulted in increased ß-secretase activity and levels of Aß, suggesting that lack of myristoylation contributes to the observed cell-phenotypes. Molecular weight determination, by gel-electrophoresis and mass spectrometry, of hABCA7 peptides spanning position 1527 showed loss of post-translational modification in the risk-allele peptide. These results suggest that decreased expression, or impaired function, of ABCA7 may contribute to AD pathology.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Variação Genética/genética , Fragmentos de Peptídeos/metabolismo , Processamento de Proteína Pós-Traducional/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Análise de Variância , Animais , Células CHO , Cricetulus , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Células HEK293 , Humanos , Íntrons/genética , Ácido Mirístico/farmacologia , Polimorfismo de Nucleotídeo Único , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Transfecção
5.
Neurosci Bull ; 32(3): 227-38, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27207326

RESUMO

Alzheimer's disease (AD) is characterized by amyloid-ß (Aß) toxicity, tau pathology, insulin resistance, neuroinflammation, and dysregulation of cholesterol homeostasis, all of which play roles in neurodegeneration. Insulin has polytrophic effects on neurons and may be at the center of these pathophysiological changes. In this study, we investigated possible relationships among insulin signaling and cholesterol biosynthesis, along with the effects of Aß42 on these pathways in vitro. We found that neuroblastoma 2a (N2a) cells transfected with the human gene encoding amyloid-ß protein precursor (AßPP) (N2a-AßPP) produced Aß and exhibited insulin resistance by reduced p-Akt and a suppressed cholesterol-synthesis pathway following insulin treatment, and by increased phosphorylation of insulin receptor subunit-1 at serine 612 (p-IRS-S612) as compared to parental N2a cells. Treatment of human neuroblastoma SH-SY5Y cells with Aß42 also increased p-IRS-S612, suggesting that Aß42 is responsible for insulin resistance. The insulin resistance was alleviated when N2a-AßPP cells were treated with higher insulin concentrations. Insulin increased Aß release from N2a-AßPP cells, by which it may promote Aß clearance. Insulin increased cholesterol-synthesis gene expression in SH-SY5Y and N2a cells, including 24-dehydrocholesterol reductase (DHCR24) and 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGCR) through sterol-regulatory element-binding protein-2 (SREBP2). While Aß42-treated SH-SY5Y cells exhibited increased HMGCR expression and c-Jun phosphorylation as pro-inflammatory responses, they also showed down-regulation of neuro-protective/anti-inflammatory DHCR24. These results suggest that Aß42 may cause insulin resistance, activate JNK for c-Jun phosphorylation, and lead to dysregulation of cholesterol homeostasis, and that enhancing insulin signaling may relieve the insulin-resistant phenotype and the dysregulated cholesterol-synthesis pathway to promote Aß release for clearance from neural cells.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/farmacologia , Colesterol/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Resistência à Insulina , Insulina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Análise de Variância , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Hidroximetilglutaril-CoA Redutases/genética , Hidroximetilglutaril-CoA Redutases/metabolismo , Resistência à Insulina/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuroblastoma/patologia , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/genética , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/genética , RNA Mensageiro , Transdução de Sinais/genética , Proteína de Ligação a Elemento Regulador de Esterol 2/genética , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo , Transfecção
6.
Rev Neurosci ; 25(4): 509-25, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24622783

RESUMO

Alzheimer's disease (AD) is the most common form of dementia. Pathologically, it is characterized by degeneration of neurons and synapses, the deposition of extracellular plaques consisting of aggregated amyloid-ß (Aß) peptides, and intracellular neurofibrillary tangles made up of hyperphosphorylated tau protein. Recently, the spotlights have been centered on two characteristics of AD, neuroinflammation and insulin resistance. Because both of these pathways play roles in synaptic dysfunction and neurodegeneration, they become potential targets for therapeutic intervention that could impede the progression of the disease. Here, we present an overview of the traditional amyloid hypothesis, as well as emerging data on both inflammatory and impaired insulin signaling pathways in AD. It becomes evident that more than one concurrent treatment can be synergistic and various combinations should be discussed as a potential therapeutic strategy to correct the anomalies in AD. Insulin resistance, Aß/tau pathologies, neuroinflammation, and dysregulation of central nervous system homeostasis are intertwined processes that together create the complex pathology of AD and should be considered as a whole picture.


Assuntos
Doença de Alzheimer/metabolismo , Resistência à Insulina , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Humanos , Inflamação/metabolismo , Insulina/metabolismo , Transdução de Sinais
7.
J Alzheimers Dis ; 40(1): 105-22, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24346217

RESUMO

One of the hallmarks of Alzheimer's disease (AD) is the accumulation and deposition of amyloid-ß (Aß) peptides in the brain and cerebral vasculature. Aß evokes neuroinflammation and has been implicated in insulin signaling disruption and JNK-AP1 activation, contributing to AD neuropathologies including oxidative injury and vascular insufficiencies. In this study we aim to better understand the protective mechanisms of insulin signaling and JNK-AP1 inhibition on the adverse effects of Aß. Four-hour treatment of hCMEC/D3, the immortalized human brain endothelial cells (iHBEC), with Aß1-42 resulted in significant c-Jun phosphorylation, oxidative stress, and cell toxicity. Concurrent treatment with Aß1-42 and insulin or Aß1-42 and JNK inhibitor SP600125 significantly improved cell viability. Cytokine array on conditioned media showed that insulin and SP600125 strongly reduced all Aß1-42-induced cytokines. ELISA confirmed the protective effect of insulin and SP600125 on Aß-induced expression of interleukin (IL)-8 and Growth related oncogene-α (Gro-α). qRT-PCR revealed that insulin and SP600125 protected iHBEC from Aß1-42-induced inflammatory gene expression. Transcription factor profiling showed that treatment of iHBEC with Aß1-42, insulin, or SP600125 alone or in combination resulted in profound changes in modulating the activities of multiple transcription factors and relevant pathways, some of which were validated by western blot. Insulin treatment and JNK inhibition in vitro synergistically reduced c-Jun phosphorylation and thus JNK-AP1 signaling activation. The study suggests that activation of insulin and blocking of JNK-AP1 signaling inhibits Aß-induced dysregulation of insulin signaling and inflammatory response.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Insulina/metabolismo , MAP Quinase Quinase 4/metabolismo , Fragmentos de Peptídeos/toxicidade , Transdução de Sinais/fisiologia , Análise de Variância , Antracenos/farmacologia , Encéfalo/citologia , Linhagem Celular Transformada , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Citocinas/genética , Citocinas/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática , Humanos , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA