Identifying key molecules that mediate brain reward plasticity remains an important goal of current drug abuse research. We recently identified a key role for MEF2 transcription factors as regulators of cocaine-induced synaptic and behavioral plasticity associated with repeated cocaine exposure. We find that cocaine-dependent inhibition of MEF2 in the NAc is required for increased MSN spine density, and that enhanced synaptic connectivity in the NAc after chronic cocaine exposure represents a compensatory mechanism that limits maladaptive behavioral responses associated with addiction, rather than supporting them. In this grant, we will elucidate cocaine- and cAMP-induced signaling events that control MEF2 activity in the striatum, including an exciting new regulatory mechanism for class IIa histone deacetylases that could have important implications for epigenetic regulation of drug addiction. To this end, we propose the following:
Specific Aim 1 : Our preliminary findings suggest that chronic cocaine exposure inhibits MEF2 activity by a cAMP-dependent process involving inhibitory phosphorylation of MEF2 (P-S408/444). In this aim, we will test the hypothesis that cocaine and cAMP signaling regulates MEF2 activity through control of P-S408/444 levels. In doing so, we will also characterize the spatial and temporal regulation of P-MEF2 in vivo after cocaine exposure. To these ends, we will use established experimental approaches, such as immunohistochemistry, RNAi-based protein replacement and transcriptional reporter assays, to test in vivo and in cultured primary striatal neurons the importance of P-S408/444 MEF2 for cocaine and cAMP-dependent regulation of MEF2.
Specific Aim 2 : Our preliminary findings revealed that overexpression of the regulator of calmodulin signaling (RCS) negatively regulates MEF2 activity in cultured striatal neurons in a Ser55 dependent manner (PKA site). In this aim, we will test the hypothesis that P-S55 RCS is required for cAMP-dependent inhibition of basal and calcium-activated MEF2-dependent transcription, and test the hypothesis that RCS functions in vivo to limit sensitized behavioral responses to cocaine. To this end, we will analyze existing RCS knockout mice using established experimental approaches to test in vivo, and in cultured striatal neurons, the importance of RCS as a MEF2 negative regulator and as a cocaine-induced regulator of behavioral plasticity in vivo.
Specific Aim 3 : Our preliminary studies indicate that activation of cAMP/PKA signaling promotes the nuclear import of HDAC5 in striatal neurons via dephosphorylation of HDAC5 at a novel Cdk5 site. In this aim, we will test the hypothesis that chronic cocaine exposure, via PKA-dependent dephosphorylation of HDAC5, promotes enhanced HDAC5 nuclear localization, which serves to reduce transcriptional responses and limit sensitized behavioral responses to repeated cocaine exposure. We will test this idea using novel P-HDAC5 antibodies with NAc tissues of cocaine exposed mice, mechanistic analysis of HDAC5 nucleocytoplasmic shuttling, and behavioral analysis of HDAC5 phospho-site mutant-expressing mice.

Public Health Relevance

Drug addiction is a chronic human diseases characterized by uncontrolled drug use despite severe adverse consequences to the addict. The results of the proposed studies will provide valuable new insights into the genes and brain mechanisms that control long- lasting behavioral responses to chronic drug use, and could generate new therapeutic targets for the treatment of drug addiction, for which there are currently very limited treatment options.

Agency
National Institute of Health (NIH)
Institute
National Institute on Drug Abuse (NIDA)
Type
Research Project (R01)
Project #
7R01DA027664-07
Application #
9318755
Study Section
Molecular Neuropharmacology and Signaling Study Section (MNPS)
Program Officer
Wu, Da-Yu
Project Start
2011-03-01
Project End
2017-02-28
Budget Start
2016-07-19
Budget End
2017-02-28
Support Year
7
Fiscal Year
2015
Total Cost
$4,562
Indirect Cost
$1,510
Name
Medical University of South Carolina
Department
Neurosciences
Type
Schools of Medicine
DUNS #
183710748
City
Charleston
State
SC
Country
United States
Zip Code
29403
Parrilla-Carrero, Jeffrey; Buchta, William C; Goswamee, Priyodarshan et al. (2018) Restoration of Kv7 Channel-Mediated Inhibition Reduces Cued-Reinstatement of Cocaine Seeking. J Neurosci 38:4212-4229
Taniguchi, Makoto; Carreira, Maria B; Cooper, Yonatan A et al. (2017) HDAC5 and Its Target Gene, Npas4, Function in the Nucleus Accumbens to Regulate Cocaine-Conditioned Behaviors. Neuron 96:130-144.e6
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:
Smith, Laura N; Penrod, Rachel D; Taniguchi, Makoto et al. (2016) Assessment of Cocaine-induced Behavioral Sensitization and Conditioned Place Preference in Mice. J Vis Exp :53107
Smith, Laura N; Jedynak, Jakub P; Fontenot, Miles R et al. (2014) Fragile X mental retardation protein regulates synaptic and behavioral plasticity to repeated cocaine administration. Neuron 82:645-58
Smith, Laura N; Cowan, Christopher W (2013) Striking a balance in fragile X. Nat Med 19:1370-1
Zang, Tong; Maksimova, Marina A; Cowan, Christopher W et al. (2013) Postsynaptic FMRP bidirectionally regulates excitatory synapses as a function of developmental age and MEF2 activity. Mol Cell Neurosci 56:39-49
Rothenfluh, Adrian; Cowan, Christopher W (2013) Emerging roles of actin cytoskeleton regulating enzymes in drug addiction: actin or reactin'? Curr Opin Neurobiol 23:507-12
Taniguchi, Makoto; Carreira, Maria B; Smith, Laura N et al. (2012) Histone deacetylase 5 limits cocaine reward through cAMP-induced nuclear import. Neuron 73:108-20
Tsai, Nien-Pei; Wilkerson, Julia R; Guo, Weirui et al. (2012) Multiple autism-linked genes mediate synapse elimination via proteasomal degradation of a synaptic scaffold PSD-95. Cell 151:1581-94