Your browser doesn't support javascript.
loading
Amyloid-ß-induced dendritic spine elimination requires Ca2+-permeable AMPA receptors, AKAP-Calcineurin-NFAT signaling, and the NFAT target gene Mdm2.
Martinez, Tyler P; Larsen, Matthew E; Sullivan, Emily; Woolfrey, Kevin M; Dell'Acqua, Mark L.
Afiliación
  • Martinez TP; Pharmacology PhD Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.
  • Larsen ME; Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.
  • Sullivan E; Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.
  • Woolfrey KM; Neuroscience PhD Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.
  • Dell'Acqua ML; Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.
eNeuro ; 2024 Feb 08.
Article en En | MEDLINE | ID: mdl-38331575
ABSTRACT
Alzheimer's Disease (AD) is associated with brain accumulation of synaptotoxic amyloid-ß (Aß) peptides produced by the proteolytic processing of amyloid precursor protein (APP). Cognitive impairments associated with AD correlate with dendritic spine and excitatory synapse loss, particularly within the hippocampus. In rodents, soluble Aß oligomers impair hippocampus-dependent learning and memory, promote dendritic spine loss, inhibit NMDA-type glutamate receptor (NMDAR)-dependent long-term potentiation (LTP), and promote synaptic depression (LTD), at least in part through activation of the Ca2+-CaM-dependent phosphatase calcineurin (CaN). Yet, questions remain regarding Aß-dependent postsynaptic CaN signaling specifically at the synapse to mediate its synaptotoxicity. Here, we use pharmacologic and genetic approaches to demonstrate a role for postsynaptic signaling via A kinase-anchoring protein 150 (AKAP150)-scaffolded CaN in mediating Aß-induced dendritic spine loss in hippocampal neurons from rats and mice of both sexes. In particular, we found that Ca2+-permeable AMPA-type glutamate receptors (CP-AMPARs), which were previously shown to signal through AKAP-anchored CaN to promote both LTD and Aß-dependent inhibition of LTP, are also required upstream of AKAP-CaN signaling to mediate spine loss via overexpression of APP containing multiple mutations linked to familial, early-onset AD and increased Aß production. In addition, we found that the CaN-dependent nuclear factor of activated T-cells (NFAT) transcription factors are required downstream to promote Aß-mediated dendritic spine loss. Finally, we identified the E3-ubiquitin ligase Mdm2, which was previously linked to LTD and developmental synapse elimination, as a downstream NFAT target gene upregulated by Aß whose enzymatic activity is required for Aß-mediated spine loss.Significance Statement Impaired hippocampal function and synapse loss are hallmarks of AD linked to Aß oligomers. Aß exposure acutely blocks hippocampal LTP and enhances LTD and chronically leads to dendritic spine synapse loss. In particular, Aß hijacks normal plasticity mechanisms, biasing them toward synapse weakening/elimination, with previous studies broadly linking CaN phosphatase signaling to this synaptic dysfunction. However, we do not understand how Aß engages signaling specifically at synapses. Here we elucidate a synapse-to-nucleus signaling pathway coordinated by the postsynaptic scaffold protein AKAP150 that is activated by Ca2+ influx through CP-AMPARs and transduced to nucleus by CaN-NFAT signaling to transcriptionally upregulate the E3-ubiquitin ligase Mdm2 that is required for Aß-mediated spine loss. These findings identify Mdm2 as potential therapeutic target for AD.

Texto completo: 1 Bases de datos: MEDLINE Tipo de estudio: Prognostic_studies Idioma: En Revista: ENeuro Año: 2024 Tipo del documento: Article

Texto completo: 1 Bases de datos: MEDLINE Tipo de estudio: Prognostic_studies Idioma: En Revista: ENeuro Año: 2024 Tipo del documento: Article