Your browser doesn't support javascript.
loading
PI3K couples long-term synaptic potentiation with cofilin recruitment and actin polymerization in dendritic spines via its regulatory subunit p85α.
López-García, Sergio; López-Merino, Esperanza; Fernández-Rodrigo, Alba; Zamorano-González, Pablo; Gutiérrez-Eisman, Silvia; Jiménez-Sánchez, Raquel; Esteban, José A.
Affiliation
  • López-García S; Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.
  • López-Merino E; Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.
  • Fernández-Rodrigo A; Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.
  • Zamorano-González P; Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.
  • Gutiérrez-Eisman S; Current address: Universidad de Málaga, Málaga, Spain.
  • Jiménez-Sánchez R; Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.
  • Esteban JA; Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.
Cell Mol Life Sci ; 81(1): 358, 2024 Aug 19.
Article in En | MEDLINE | ID: mdl-39158722
ABSTRACT
Long-term synaptic plasticity is typically associated with morphological changes in synaptic connections. However, the molecular mechanisms coupling functional and structural aspects of synaptic plasticity are still poorly defined. The catalytic activity of type I phosphoinositide-3-kinase (PI3K) is required for specific forms of synaptic plasticity, such as NMDA receptor-dependent long-term potentiation (LTP) and mGluR-dependent long-term depression (LTD). On the other hand, PI3K signaling has been linked to neuronal growth and synapse formation. Consequently, PI3Ks are promising candidates to coordinate changes in synaptic strength with structural remodeling of synapses. To investigate this issue, we targeted individual regulatory subunits of type I PI3Ks in hippocampal neurons and employed a combination of electrophysiological, biochemical and imaging techniques to assess their role in synaptic plasticity. We found that a particular regulatory isoform, p85α, is selectively required for LTP. This specificity is based on its BH domain, which engages the small GTPases Rac1 and Cdc42, critical regulators of the actin cytoskeleton. Moreover, cofilin, a key regulator of actin dynamics that accumulates in dendritic spines after LTP induction, failed to do so in the absence of p85α or when its BH domain was overexpressed as a dominant negative construct. Finally, in agreement with this convergence on actin regulatory mechanisms, the presence of p85α in the PI3K complex determined the extent of actin polymerization in dendritic spines during LTP. Therefore, this study reveals a molecular mechanism linking structural and functional synaptic plasticity through the coordinate action of PI3K catalytic activity and a specific isoform of the regulatory subunits.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Actins / Long-Term Potentiation / Dendritic Spines / Actin Depolymerizing Factors / Hippocampus Limits: Animals Language: En Journal: Cell Mol Life Sci Journal subject: BIOLOGIA MOLECULAR Year: 2024 Document type: Article Affiliation country: España

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Actins / Long-Term Potentiation / Dendritic Spines / Actin Depolymerizing Factors / Hippocampus Limits: Animals Language: En Journal: Cell Mol Life Sci Journal subject: BIOLOGIA MOLECULAR Year: 2024 Document type: Article Affiliation country: España