A typical neocortical pyramidal neuron integrates information from thousands of excitatory inputs from both ?local? circuits, within a cortical region, and from many ?long-range? circuits from other cortical and non-cortical regions, both ipsi- and contralaterally. These long-range connections develop postnatally, are highly specified, and regulated by sensory experience. Remarkably, >50% of excitatory synapses a typical neocortical pyramidal neuron receives are from long-range connections, but virtually nothing is known of how functional long-range connections develop, are regulated by experience or if and how long-range and local inputs are balanced. In the last grant cycle, we discovered that the activity-dependent transcription factor, Myocyte-Enhancer Factor 2C (MEF2C) functions postnatally and cell autonomously to regulate the balance of local and long-range excitatory connectivity onto layer (L) 2/3 neocortical neurons in primary somatosensory cortex. Specifically, MEF2C promotes connectivity from multiple local excitatory circuits onto L2/3 neurons, while weakening callosal inputs from contralateral somatosensory cortex. Importantly, sensory experience is necessary for MEF2C regulation of both local and callosal circuits suggesting that MEF2C as a key player in experience-dependent, input-specific development of cortical circuits. Our findings have important implications for neurodevelopmental disorders. Brain mapping studies in humans with autism spectrum disorder (ASD) and schizophrenia (SCZ) reveal imbalances in local vs. long-range functional cortical connectivity. Furthermore, loss of function mutations in Mef2c are linked with intellectual disability (ID), ASD, epilepsy and SCZ in humans and mice. We hypothesize that sensory experience-driven neural activity regulates MEF2C-dependent transcriptional control of target genes to mediate input-specific development and plasticity of cortical circuits. Thus, a corollary of this hypothesis predicts that loss of function mutations in Mef2c or its effectors would result in imbalances of local and long-range connectivity, abnormal sensory-related behaviors and neuropsychiatric disease. To test these hypotheses:
In Aim 1, we will use state-of-the-art optogenetic and photostimulation circuit mapping methods to determine if Mef2c deletion generally strengthens long-range inputs onto L2/3 neurons, bidirectionally regulates inputs based on their dendritic location and maintains the input-specificity of mature circuits.
Aim 2 : MEF2C functions both to repress transcription of target genes and stimulate their transcription in response to neural activity. Using MEF2C mutants, we will determine which of these functions mediates experience-dependent, input-specific regulation of cortical circuits.
Aim 3 : FMRP, mGluR5 and Arc are required for MEF2 regulation of synapses in culture neurons and associated with ASD, ID and SCZ. Here, we will test their role in MEF2C-mediated input-specific development of cortical circuits.
Aim 4 : To identify candidate genes that regulate cortical circuit input specificity, we will identify the postnatal MEF2C- and experience-dependent transcriptomes in L2/3 neocortical neurons using fluorescent-activated cell sorting (FACS) and RNA sequencing.

Public Health Relevance

In response to experience, neurons form specific connections locally, within a brain region, and long-range connections, with neurons from diverse brain regions and the balance of local and long-range connections is abnormal in neurodevelopmental disorders. We have identified a gene, MEF2C, which is mutated in autism and schizophrenia and regulated by experience, that differentially affects local and long-range connections; the first of its kind. In this grant application, we will determine how MEF2C and experience determine the specific connectivity of local and long-range connections to understand normal brain development and neurodevelopmental disorders.

Agency
National Institute of Health (NIH)
Institute
Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD)
Type
Research Project (R01)
Project #
5R01HD052731-14
Application #
10102137
Study Section
Neurodifferentiation, Plasticity, and Regeneration Study Section (NDPR)
Program Officer
King, Tracy
Project Start
2008-02-08
Project End
2023-01-31
Budget Start
2021-02-01
Budget End
2022-01-31
Support Year
14
Fiscal Year
2021
Total Cost
Indirect Cost
Name
University of Texas Sw Medical Center Dallas
Department
Neurosciences
Type
Schools of Medicine
DUNS #
800771545
City
Dallas
State
TX
Country
United States
Zip Code
75390
Wilkerson, Julia R; Albanesi, Joseph P; Huber, Kimberly M (2018) Roles for Arc in metabotropic glutamate receptor-dependent LTD and synapse elimination: Implications in health and disease. Semin Cell Dev Biol 77:51-62
Chang, Chia-Wei; Wilkerson, Julia R; Hale, Carly F et al. (2017) Distinct stages of synapse elimination are induced by burst firing of CA1 neurons and differentially require MEF2A/D. Elife 6:
Tsai, Nien-Pei; Huber, Kimberly M (2017) Protocadherins and the Social Brain. Biol Psychiatry 81:173-174
Tsai, Nien-Pei; Wilkerson, Julia R; Guo, Weirui et al. (2017) FMRP-dependent Mdm2 dephosphorylation is required for MEF2-induced synapse elimination. Hum Mol Genet 26:293-304
Rajkovich, Kacey E; Loerwald, Kristofer W; Hale, Carly F et al. (2017) Experience-Dependent and Differential Regulation of Local and Long-Range Excitatory Neocortical Circuits by Postsynaptic Mef2c. Neuron 93:48-56
Harrington, Adam J; Raissi, Aram; Rajkovich, Kacey et al. (2016) MEF2C regulates cortical inhibitory and excitatory synapses and behaviors relevant to neurodevelopmental disorders. Elife 5:
Zhang, Zilai; Cao, Mou; Chang, Chia-Wei et al. (2016) Autism-Associated Chromatin Regulator Brg1/SmarcA4 Is Required for Synapse Development and Myocyte Enhancer Factor 2-Mediated Synapse Remodeling. Mol Cell Biol 36:70-83
Loerwald, Kristofer W; Patel, Ankur B; Huber, Kimberly M et al. (2015) Postsynaptic mGluR5 promotes evoked AMPAR-mediated synaptic transmission onto neocortical layer 2/3 pyramidal neurons during development. J Neurophysiol 113:786-95
Byers, Christopher E; Barylko, Barbara; Ross, Justin A et al. (2015) Enhancement of dynamin polymerization and GTPase activity by Arc/Arg3.1. Biochim Biophys Acta 1850:1310-8
Wilkerson, Julia R; Tsai, Nien-Pei; Maksimova, Marina A et al. (2014) A role for dendritic mGluR5-mediated local translation of Arc/Arg3.1 in MEF2-dependent synapse elimination. Cell Rep 7:1589-1600

Showing the most recent 10 out of 26 publications