Your browser doesn't support javascript.
loading
MicroRNA miR124 is required for the expression of homeostatic synaptic plasticity.
Hou, Qingming; Ruan, Hongyu; Gilbert, James; Wang, Guan; Ma, Qi; Yao, Wei-Dong; Man, Heng-Ye.
Affiliation
  • Hou Q; Department of Biology, Boston University, 5 Cummington Mall, Boston, Massachusetts 02215, USA.
  • Ruan H; Division of Neuroscience, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts 01772, USA.
  • Gilbert J; Department of Biology, Boston University, 5 Cummington Mall, Boston, Massachusetts 02215, USA.
  • Wang G; Department of Biology, Boston University, 5 Cummington Mall, Boston, Massachusetts 02215, USA.
  • Ma Q; Division of Neuroscience, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts 01772, USA.
  • Yao WD; Division of Neuroscience, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts 01772, USA.
  • Man HY; Department of Biology, Boston University, 5 Cummington Mall, Boston, Massachusetts 02215, USA.
Nat Commun ; 6: 10045, 2015 Dec 01.
Article in En | MEDLINE | ID: mdl-26620774
ABSTRACT
Homeostatic synaptic plasticity is a compensatory response to alterations in neuronal activity. Chronic deprivation of neuronal activity results in an increase in synaptic AMPA receptors (AMPARs) and postsynaptic currents. The biogenesis of GluA2-lacking, calcium-permeable AMPARs (CP-AMPARs) plays a crucial role in the homeostatic response; however, the mechanisms leading to CP-AMPAR formation remain unclear. Here we show that the microRNA, miR124, is required for the generation of CP-AMPARs and homeostatic plasticity. miR124 suppresses GluA2 expression via targeting its 3'-UTR, leading to the formation of CP-AMPARs. Blockade of miR124 function abolishes the homeostatic response, whereas miR124 overexpression leads to earlier induction of homeostatic plasticity. miR124 transcription is controlled by an inhibitory transcription factor EVI1, acting by association with the deacetylase HDAC1. Our data support a cellular cascade in which inactivity relieves EVI1/HDAC-mediated inhibition of miR124 gene transcription, resulting in enhanced miR124 expression, formation of CP-AMPARs and subsequent induction of homeostatic synaptic plasticity.
Subject(s)

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: MicroRNAs / Neuronal Plasticity / Neurons Limits: Animals / Humans Language: En Journal: Nat Commun Journal subject: BIOLOGIA / CIENCIA Year: 2015 Document type: Article Affiliation country: Estados Unidos

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: MicroRNAs / Neuronal Plasticity / Neurons Limits: Animals / Humans Language: En Journal: Nat Commun Journal subject: BIOLOGIA / CIENCIA Year: 2015 Document type: Article Affiliation country: Estados Unidos