4.8 Article

A key requirement for synaptic Reelin signaling in ketamine-mediated behavioral and synaptic action

Publisher

NATL ACAD SCIENCES
DOI: 10.1073/pnas.2103079118

Keywords

ketamine; antidepressant; Reelin; Apoer2; NMDA receptor

Funding

  1. NIH [MH070727, MH081060, MH066198]
  2. Basic Science Research Program through the National Research Foundation of Korea - Ministry of Education [2016R1A6A3A03008533]
  3. National Heart, Lung, and Blood Institute [R37 HL063762]
  4. National Institute on Aging (NIA) [RF AG053391]
  5. National Institute of Neurological Disorders and Stroke
  6. NIA [R01 NS093382]
  7. BrightFocus [A2016396S]
  8. Bluefield Project to Cure frontotemporal dementia
  9. Harrington Scholar Innovator Award
  10. National Research Foundation of Korea [2016R1A6A3A03008533] Funding Source: Korea Institute of Science & Technology Information (KISTI), National Science & Technology Information Service (NTIS)

Ask authors/readers for more resources

This study investigated whether disruptions in Reelin-mediated synaptic signaling alter ketamine-triggered synaptic plasticity and behavioral effects. The findings suggest that impairments in Reelin-Apoer2-SFK pathway components may in part underlie nonresponsiveness to ketamine's antidepressant action.
Ketamine is a noncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist that produces rapid antidepressant action in some patients with treatment-resistant depression. However, recent data suggest that similar to 50% of patients with treatment-resistant depression do not respond to ketamine. The factors that contribute to the nonresponsiveness to ketamine's antidepressant action remain unclear. Recent studies have reported a role for secreted glycoprotein Reelin in regulating pre- and postsynaptic function, which suggests that Reelin may be involved in ketamine's antidepressant action, although the premise has not been tested. Here, we investigated whether the disruption of Reelin-mediated synaptic signaling alters ketamine-triggered synaptic plasticity and behavioral effects. To this end, we used mouse models with genetic deletion of Reelin or apolipoprotein E receptor 2 (Apoer2), as well as pharmacological inhibition of their downstream effectors, Src family kinases (SFKs) or phosphoinositide 3-kinase. We found that disruption of Reelin, Apoer2, or SFKs blocks ketamine-driven behavioral changes and synaptic plasticity in the hippocampal CA1 region. Although ketamine administration did not affect tyrosine phosphorylation of DAB1, an adaptor protein linked to downstream signaling of Reelin, disruption of Apoer2 or SFKs impaired baseline NMDA receptor-mediated neurotransmission. These results suggest that maintenance of baseline NMDA receptor function by Reelin signaling may be a key permissive factor required for ketamine's antidepressant effects. Taken together, our results suggest that impairments in Reelin-Apoer2-SFK pathway components may in part underlie nonresponsiveness to ketamine's antidepressant action.

Authors

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

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available