Synaptic plasticity is thought to shape the connectivity of neural circuits, and to maintain the stability of neural circuits for optimal information processing. Connectivity is shaped, at least in part, by Hebbian-type plasticity, which adjusts the strength of a subset of synapses in a circuit in an input-specific fashion. In contrast, network stability is maintained via homeostatic synaptic plasticity, which modifies all synapses of a neuron concurrently in a multiplicative fashion (referred to as synaptic scaling). We recently demonstrated that all-trans retinoic acid (RA) directly acts on synapses to increase synaptic strength by inducing the synaptic insertion of GluR1-type AMPA receptors, and that the action of RA is essential for at least some forms of homeostatic plasticity. Moreover, we showed that RA may act by binding to dendritically localized RAR1 receptors in synapses, and that RA-signaling may intersect with Hebbian-type synaptic plasticity. These findings lead us to the overall hypothesis that RA constitutes a novel diffusible messenger in synapses that serves to regulate synaptic strength in response to external stimuli by altering postsynaptic GluR1 receptor levels. To test this overall hypothesis, we propose three specific aims that focus on key questions emerging from this hypothesis, and will primarily employ physiological measurements in hippocampal neurons.
Aim 1 will examine the role of RA in various forms of homeostatic plasticity and short-term synaptic plasticity to explore the generality of its action. Results from this aim will define the physiological framework under which RA operates.
Aim 2 will test the role of RA-binding to RAR1 in regulating hippocampal synaptic strength using conditional deletion of RAR1 in vitro in cultured neurons and slices, and employ rescue experiments to explore the relative contribution of various RAR1 protein domains to its synaptic function.
Aim 3 will conditionally delete RAR1 in mice in vivo (as opposed to culture preparations) to investigate the functional interplay between RA-mediated synaptic scaling and Hebbian plasticity in vivo. Together, these experiments will thus utilize innovative approaches to explore the unexpected role of RA and RAR1 in hippocampal synaptic transmission, and provide initial insights into their mechanism of action. The significance of these experiments not only rests on their implications for understanding of synaptic transmission, but also on their potential relevance for retinoids-related neurological disorders such as depression and memory loses.

Public Health Relevance

RA signaling has been implicated in learning and memory as well as mood disorders. However, we are at the beginning of our understanding of the molecular mechanisms involved in RA signaling and synaptic function. Work proposed here will increase our understanding of the fundamental mechanisms involved in RA-mediated synaptic plasticity, which will broaden our view of brain function and may help in developing novel therapies for neurological disorders.

Agency
National Institute of Health (NIH)
Institute
National Institute of Mental Health (NIMH)
Type
Research Project (R01)
Project #
5R01MH091193-03
Application #
8394935
Study Section
Synapses, Cytoskeleton and Trafficking Study Section (SYN)
Program Officer
Asanuma, Chiiko
Project Start
2011-02-01
Project End
2015-12-31
Budget Start
2013-01-01
Budget End
2013-12-31
Support Year
3
Fiscal Year
2013
Total Cost
$449,778
Indirect Cost
$168,580
Name
Stanford University
Department
Neurosciences
Type
Schools of Medicine
DUNS #
009214214
City
Stanford
State
CA
Country
United States
Zip Code
94305
Zhang, Zhenjie; Marro, Samuele G; Zhang, Yingsha et al. (2018) The fragile X mutation impairs homeostatic plasticity in human neurons by blocking synaptic retinoic acid signaling. Sci Transl Med 10:
Li, Jie; Park, Esther; Zhong, Lei R et al. (2018) Homeostatic synaptic plasticity as a metaplasticity mechanism?-?a molecular and cellular perspective. Curr Opin Neurobiol 54:44-53
Wu, Dick; Bacaj, Taulant; Morishita, Wade et al. (2017) Postsynaptic synaptotagmins mediate AMPA receptor exocytosis during LTP. Nature 544:316-321
Yee, Ada X; Chen, Lu (2016) Differential regulation of spontaneous and evoked inhibitory synaptic transmission in somatosensory cortex by retinoic acid. Synapse 70:445-52
Arendt, Kristin L; Zhang, Yingsha; Jurado, Sandra et al. (2015) Retinoic Acid and LTP Recruit Postsynaptic AMPA Receptors Using Distinct SNARE-Dependent Mechanisms. Neuron 86:442-56
Liang, J; Xu, W; Hsu, Y-T et al. (2015) Conditional neuroligin-2 knockout in adult medial prefrontal cortex links chronic changes in synaptic inhibition to cognitive impairments. Mol Psychiatry 20:850-9
Kim, Jae-Ick; Ganesan, Subhashree; Luo, Sarah X et al. (2015) Aldehyde dehydrogenase 1a1 mediates a GABA synthesis pathway in midbrain dopaminergic neurons. Science 350:102-6
Arendt, Kristin L; Zhang, Zhenjie; Ganesan, Subhashree et al. (2015) Calcineurin mediates homeostatic synaptic plasticity by regulating retinoic acid synthesis. Proc Natl Acad Sci U S A 112:E5744-52
Chen, Lu; Lau, Anthony G; Sarti, Federica (2014) Synaptic retinoic acid signaling and homeostatic synaptic plasticity. Neuropharmacology 78:3-12
Hastie, Peter; Ulbrich, Maximilian H; Wang, Hui-Li et al. (2013) AMPA receptor/TARP stoichiometry visualized by single-molecule subunit counting. Proc Natl Acad Sci U S A 110:5163-8

Showing the most recent 10 out of 15 publications