Your browser doesn't support javascript.
loading
The Pluripotency Factor Nanog Protects against Neuronal Amyloid ß-Induced Toxicity and Oxidative Stress through Insulin Sensitivity Restoration.
Chang, Ching-Chi; Li, Hsin-Hua; Tsou, Sing-Hua; Hung, Hui-Chih; Liu, Guang-Yaw; Korolenko, Tatiana A; Lai, Te-Jen; Ho, Ying-Jui; Lin, Chih-Li.
Afiliação
  • Chang CC; Institute of Medicine, Chung Shan Medical University, Taichung 402367, Taiwan.
  • Li HH; Department of Psychiatry, Chung Shan Medical University Hospital, Taichung 402367, Taiwan.
  • Tsou SH; Institute of Medicine, Chung Shan Medical University, Taichung 402367, Taiwan.
  • Hung HC; Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402367, Taiwan.
  • Liu GY; Institute of Medicine, Chung Shan Medical University, Taichung 402367, Taiwan.
  • Korolenko TA; Department of Life Sciences and Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402204, Taiwan.
  • Lai TJ; Institute of Medicine, Chung Shan Medical University, Taichung 402367, Taiwan.
  • Ho YJ; Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk 630117, Russia.
  • Lin CL; Institute of Medicine, Chung Shan Medical University, Taichung 402367, Taiwan.
Cells ; 9(6)2020 05 27.
Article em En | MEDLINE | ID: mdl-32471175
ABSTRACT
Amyloid ß (Aß) is a peptide fragment of the amyloid precursor protein that triggers the progression of Alzheimer's Disease (AD). It is believed that Aß contributes to neurodegeneration in several ways, including mitochondria dysfunction, oxidative stress and brain insulin resistance. Therefore, protecting neurons from Aß-induced neurotoxicity is an effective strategy for attenuating AD pathogenesis. Recently, applications of stem cell-based therapies have demonstrated the ability to reduce the progression and outcome of neurodegenerative diseases. Particularly, Nanog is recognized as a stem cell-related pluripotency factor that enhances self-renewing capacities and helps reduce the senescent phenotypes of aged neuronal cells. However, whether the upregulation of Nanog can be an effective approach to alleviate Aß-induced neurotoxicity and senescence is not yet understood. In the present study, we transiently overexpressed Nanog-both in vitro and in vivo-and investigated the protective effects and underlying mechanisms against Aß. We found that overexpression of Nanog is responsible for attenuating Aß-triggered neuronal insulin resistance, which restores cell survival through reducing intracellular mitochondrial superoxide accumulation and cellular senescence. In addition, upregulation of Nanog expression appears to increase secretion of neurotrophic factors through activation of the Nrf2 antioxidant defense pathway. Furthermore, improvement of memory and learning were also observed in rat model of Aß neurotoxicity mediated by upregulation of Nanog in the brain. Taken together, our study suggests a potential role for Nanog in attenuating the neurotoxic effects of Aß, which in turn, suggests that strategies to enhance Nanog expression may be used as a novel intervention for reducing Aß neurotoxicity in the AD brain.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Resistência à Insulina / Peptídeos beta-Amiloides / Estresse Oxidativo / Células-Tronco Pluripotentes / Proteína Homeobox Nanog / Neurônios Idioma: En Ano de publicação: 2020 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Resistência à Insulina / Peptídeos beta-Amiloides / Estresse Oxidativo / Células-Tronco Pluripotentes / Proteína Homeobox Nanog / Neurônios Idioma: En Ano de publicação: 2020 Tipo de documento: Article