Cargando…

α2δ-1 switches the phenotype of synaptic AMPA receptors by physically disrupting heteromeric subunit assembly

Many neurological disorders show an increased prevalence of GluA2-lacking, Ca(2+)-permeable AMPA receptors (CP-AMPARs), which dramatically alters synaptic function. However, the molecular mechanism underlying this distinct synaptic plasticity remains enigmatic. Here, we show that nerve injury potent...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Lingyong, Chen, Shao-Rui, Zhou, Meng-Hua, Wang, Li, Li, De-Pei, Chen, Hong, Lee, Garam, Jayaraman, Vasanthi, Pan, Hui-Lin
Formato: Online Artículo Texto
Lenguaje:English
Publicado: 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8353586/
https://www.ncbi.nlm.nih.gov/pubmed/34289359
http://dx.doi.org/10.1016/j.celrep.2021.109396
Descripción
Sumario:Many neurological disorders show an increased prevalence of GluA2-lacking, Ca(2+)-permeable AMPA receptors (CP-AMPARs), which dramatically alters synaptic function. However, the molecular mechanism underlying this distinct synaptic plasticity remains enigmatic. Here, we show that nerve injury potentiates postsynaptic, but not presynaptic, CP-AMPARs in the spinal dorsal horn via α2δ-1. Overexpressing α2δ-1, previously regarded as a Ca(2+) channel subunit, augments CP-AMPAR levels at the cell surface and synapse. Mechanistically, α2δ-1 physically interacts with both GluA1 and GluA2 via its C terminus, inhibits the GluA1/GluA2 heteromeric assembly, and increases GluA2 retention in the endoplasmic reticulum. Consequently, α2δ-1 diminishes the availability and synaptic expression of GluA1/GluA2 heterotetramers in the spinal cord in neuropathic pain. Inhibiting α2δ-1 with gabapentin or disrupting the α2δ-1-AMPAR complex fully restores the intracellular assembly and synaptic dominance of heteromeric GluA1/GluA2 receptors. Thus, α2δ-1 is a pivotal AMPAR-interacting protein that controls the subunit composition and Ca(2+) permeability of postsynaptic AMPARs.