4.5 Article

Shank and Zinc Mediate an AMPA Receptor Subunit Switch in Developing Neurons

Journal

FRONTIERS IN MOLECULAR NEUROSCIENCE
Volume 11, Issue -, Pages -

Publisher

FRONTIERS MEDIA SA
DOI: 10.3389/fnmol.2018.00405

Keywords

zinc signaling; AMPA receptor (AMPAR); GluA2 (GluR2); GluA1 (GluR1); Shank2; Shank3; synaptic development; subunit switch

Categories

Funding

  1. Vietnam Education Foundation
  2. Stanford Graduate Fellowship
  3. Schlumberger Faculty for the Future Fellow Program
  4. Stanford Neurosciences Graduate Program
  5. JSPS KAKENHI Grant [17H06348]
  6. Marsden Fund (Royal Society of New Zealand) [13-UOA-053]
  7. Federal Government of Germany (DFG) [SFB958]
  8. German Center for Neurodegenerative Diseases (DZNE)
  9. National Institutes of Health [R21MH100717, R33MH100717]
  10. Phelan-McDermid Syndrome Foundation Fellowship
  11. Grants-in-Aid for Scientific Research [17H06348] Funding Source: KAKEN

Ask authors/readers for more resources

During development, pyramidal neurons undergo dynamic regulation of AMPA receptor (AMPAR) subunit composition and density to help drive synaptic plasticity and maturation. These normal developmental changes in AMPARs are particularly vulnerable to risk factors for Autism Spectrum Disorders (ASDs), which include loss or mutations of synaptic proteins and environmental insults, such as dietary zinc deficiency. Here, we show how Shank2 and Shank3 mediate a zinc-dependent regulation of AMPAR function and subunit switch from GluA2-lacking to GluA2-containing AMPARs. Over development, we found a concomitant increase in Shank2 and Shank3 with GluA2 at synapses, implicating these molecules as potential players in AMPAR maturation. Since Shank activation and function require zinc, we next studied whether neuronal activity regulated postsynaptic zinc at glutamatergic synapses. Zinc was found to increase transiently and reversibly with neuronal depolarization at synapses, which could affect Shank and AMPAR localization and activity. Elevated zinc induced multiple functional changes in AMPAR, indicative of a subunit switch. Specifically, zinc lengthened the decay time of AMPAR-mediated synaptic currents and reduced their inward rectification in young hippocampal neurons. Mechanistically, both Shank2 and Shank3 were necessary for the zinc-sensitive enhancement of AMPAR-mediated synaptic transmission and act in concert to promote removal of GluA1 while enhancing recruitment of GluA2 at pre-existing Shank puncta. These findings highlight a cooperative local dynamic regulation of AMPAR subunit switch controlled by zinc signaling through Shank2 and Shank3 to shape the biophysical properties of developing glutamatergic synapses. Given the zinc sensitivity of young neurons and its dependence on Shank2 and Shank3, genetic mutations and/or environmental insults during early development could impair synaptic maturation and circuit formation that underlie ASD etiology.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.5
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available