Your browser doesn't support javascript.
loading
Pathogenesis of sporadic Alzheimer's disease by deficiency of NMDA receptor subunit GluN3A.
Zhong, Weiwei; Wu, Anika; Berglund, Ken; Gu, Xiaohuan; Jiang, Michael Qize; Talati, Jay; Zhao, Jingjie; Wei, Ling; Yu, Shan Ping.
Afiliação
  • Zhong W; Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia, USA.
  • Wu A; Center for Visual and Neurocognitive Rehabilitation, Department of Veteran's Affair, Atlanta VA Medical Center, Decatur, Georgia, USA.
  • Berglund K; Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia, USA.
  • Gu X; Center for Visual and Neurocognitive Rehabilitation, Department of Veteran's Affair, Atlanta VA Medical Center, Decatur, Georgia, USA.
  • Jiang MQ; Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia, USA.
  • Talati J; Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia, USA.
  • Zhao J; Center for Visual and Neurocognitive Rehabilitation, Department of Veteran's Affair, Atlanta VA Medical Center, Decatur, Georgia, USA.
  • Wei L; Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia, USA.
  • Yu SP; Center for Visual and Neurocognitive Rehabilitation, Department of Veteran's Affair, Atlanta VA Medical Center, Decatur, Georgia, USA.
Alzheimers Dement ; 18(2): 222-239, 2022 02.
Article em En | MEDLINE | ID: mdl-34151525
ABSTRACT
The Ca2+ hypothesis for Alzheimer's disease (AD) conceives Ca2+ dyshomeostasis as a common mechanism of AD; the cause of Ca2+ dysregulation, however, is obscure. Meanwhile, hyperactivities of N-Methyl-D-aspartate receptors (NMDARs), the primary mediator of Ca2+ influx, are reported in AD. GluN3A (NR3A) is an NMDAR inhibitory subunit. We hypothesize that GluN3A is critical for sustained Ca2+ homeostasis and its deficiency is pathogenic for AD. Cellular, molecular, and functional changes were examined in adult/aging GluN3A knockout (KO) mice. The GluN3A KO mouse brain displayed age-dependent moderate but persistent neuronal hyperactivity, elevated intracellular Ca2+ , neuroinflammation, impaired synaptic integrity/plasticity, and neuronal loss. GluN3A KO mice developed olfactory dysfunction followed by psychological/cognitive deficits prior to amyloid-ß/tau pathology. Memantine at preclinical stage prevented/attenuated AD syndromes. AD patients' brains show reduced GluN3A expression. We propose that chronic "degenerative excitotoxicity" leads to sporadic AD, while GluN3A represents a primary pathogenic factor, an early biomarker, and an amyloid-independent therapeutic target.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Receptores de N-Metil-D-Aspartato / Doença de Alzheimer Tipo de estudo: Etiology_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Receptores de N-Metil-D-Aspartato / Doença de Alzheimer Tipo de estudo: Etiology_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2022 Tipo de documento: Article