학술논문

HDAC3 Activity within the Nucleus Accumbens Regulates Cocaine-induced Plasticity and Behavior in a Cell-Type Specific Manner
Document Type
article
Source
Journal of Neuroscience. 41(13)
Subject
Drug Abuse (NIDA only)
Basic Behavioral and Social Science
Neurosciences
Behavioral and Social Science
Genetics
Substance Misuse
Aetiology
2.1 Biological and endogenous factors
Good Health and Well Being
Animals
Cocaine
Conditioning
Psychological
Dopamine Uptake Inhibitors
Drug-Seeking Behavior
Enzyme Activation
Epigenesis
Genetic
Histone Deacetylases
Male
Mice
Mice
Inbred C57BL
Mice
Transgenic
Neuronal Plasticity
Nucleus Accumbens
Self Administration
addiction
epigenetics
NAc
Medical and Health Sciences
Psychology and Cognitive Sciences
Neurology & Neurosurgery
Language
Abstract
Epigenetic mechanisms regulate processes of neuroplasticity critical to cocaine-induced behaviors. This includes the Class I histone deacetylase (HDAC) HDAC3, known to act as a negative regulator of cocaine-associated memory formation within the nucleus accumbens (NAc). Despite this, it remains unknown how cocaine alters HDAC3-dependent mechanisms. Here, we profiled HDAC3 expression and activity in total NAc mouse tissue following cocaine exposure. Although chronic cocaine did not affect expression of Hdac3 within the NAc, chronic cocaine did affect promoter-specific changes in HDAC3 and H4K8Ac occupancy. These changes in promoter occupancy correlated with cocaine-induced changes in expression of plasticity-related genes. To causally determine whether cocaine-induced plasticity is mediated by HDAC3's deacetylase activity, we overexpressed a deacetylase-dead HDAC3 point mutant (HDAC3-Y298H-v5) within the NAc of adult male mice. We found that disrupting HDAC3's enzymatic activity altered selective changes in gene expression and synaptic plasticity following cocaine exposure, despite having no effects on cocaine-induced behaviors. In further assessing HDAC3's role within the NAc, we observed that chronic cocaine increases Hdac3 expression in Drd1 but not Drd2-cells of the NAc. Moreover, we discovered that HDAC3 acts selectively within D1R cell-types to regulate cocaine-associated memory formation and cocaine-seeking. Overall, these results suggest that cocaine induces cell-type-specific changes in epigenetic mechanisms to promote plasticity important for driving cocaine-related behaviors.SIGNIFICANCE STATEMENT Drugs of abuse alter molecular mechanisms throughout the reward circuitry that can lead to persistent drug-associated behaviors. Epigenetic regulators are critical drivers of drug-induced changes in gene expression. Here, we demonstrate that the activity of an epigenetic enzyme promotes neuroplasticity within the nucleus accumbens (NAc) critical to cocaine action. In addition, we demonstrate that these changes in epigenetic activity drive cocaine-seeking behaviors in a cell-type-specific manner. These findings are key in understanding and targeting cocaine's impact of neural circuitry and behavior.