Program Project Grant research has identified several signaling proteins that modulate behavioral responses to drugs of abuse or are themselves modulated by chronic drug exposure. The purpose of the Behavioral Core is to provide a high-throughput, behavior screen to characterize the role played by these proteins in several relatively straightforward drug- related behaviors that are thought to model aspects of drug addiction. In these studies, a protein of interest will be modulated by 1) by intracerebral injection of activators or inhibitors; 2) by direct transfection of neurons with viral vectors over expressing the protein of interest; or 3) by using genetic mutant mice that lack or over express the protein of interest (see Transgenic Core). The effects of modulating the target protein will then be tested in one or more of the following behavioral models: 1) novelty-seeking behavior in drug-naive animals, a trait thought to predict and perhaps contribute to individual differences in drug abuse; 2) locomotor sensitization to cocaine or morphine, a behavioral phenomenon thought to result from neuroplasticity in the mesolimbic dopamine system following repeated exposure to drugs of abuse; 3) opiate physical withdrawal, where specific behaviors are thought to reflect the neuroadaptations of opiate dependence in discrete brain regions, including the locus coeruleus; and 4) place conditioning to cocaine or morphine, which provides an indirect measure of the rewarding effects of initial periods of drug exposure, as well as conditioned place aversion in opiate-dependent animals, which measures the aversive consequence of opiate withdrawal; both behaviors are thought to involve altered neurotransmission in the nucleus accumbens and related limbic structures. The Behavioral Core will thereby provide investigators in the individual Projects of this grant with a repertoire of behavioral assays. The goal is to enable investigators to relate the molecular and cellular phenomena that form the foundation of our program of research to specific and functionally relevant behavioral endpoints related to drug addiction. Ultimately, a smaller number of molecular events will be studied in the more complex behavioral models of drug self-administration, relapse, and conditioned reinforcement that form the basis of Project 4. By consolidating more routine behavioral assays in this Core, we ensure quality control of the assays and the ability to relate behavioral findings in one Project to another, as well as facilitate the integration of molecular, cellular, and behavioral levels of analysis, a major objective of our Program Project Grant.

Agency
National Institute of Health (NIH)
Institute
National Institute on Drug Abuse (NIDA)
Type
Research Program Projects (P01)
Project #
7P01DA008227-09
Application #
6441027
Study Section
Project Start
2001-01-01
Project End
2001-06-30
Budget Start
Budget End
Support Year
9
Fiscal Year
2000
Total Cost
Indirect Cost
Name
Yale University
Department
Type
DUNS #
082359691
City
New Haven
State
CT
Country
United States
Zip Code
06520
Ayata, Pinar; Badimon, Ana; Strasburger, Hayley J et al. (2018) Epigenetic regulation of brain region-specific microglia clearance activity. Nat Neurosci 21:1049-1060
Walker, Deena M; Nestler, Eric J (2018) Neuroepigenetics and addiction. Handb Clin Neurol 148:747-765
Li, Xuan; Carreria, Maria B; Witonsky, Kailyn R et al. (2018) Role of Dorsal Striatum Histone Deacetylase 5 in Incubation of Methamphetamine Craving. Biol Psychiatry 84:213-222
Anderson, Ethan M; Larson, Erin B; Guzman, Daniel et al. (2018) Overexpression of the Histone Dimethyltransferase G9a in Nucleus Accumbens Shell Increases Cocaine Self-Administration, Stress-Induced Reinstatement, and Anxiety. J Neurosci 38:803-813
Walker, Deena M; Cates, Hannah M; Loh, Yong-Hwee E et al. (2018) Cocaine Self-administration Alters Transcriptome-wide Responses in the Brain's Reward Circuitry. Biol Psychiatry 84:867-880
Cahill, Michael E; Browne, Caleb J; Wang, Junshi et al. (2018) Withdrawal from repeated morphine administration augments expression of the RhoA network in the nucleus accumbens to control synaptic structure. J Neurochem 147:84-98
Cates, Hannah M; Li, Xuan; Purushothaman, Immanuel et al. (2018) Genome-wide transcriptional profiling of central amygdala and orbitofrontal cortex during incubation of methamphetamine craving. Neuropsychopharmacology 43:2426-2434
Gaspari, Sevasti; Purushothaman, Immanuel; Cogliani, Valeria et al. (2018) Suppression of RGSz1 function optimizes the actions of opioid analgesics by mechanisms that involve the Wnt/?-catenin pathway. Proc Natl Acad Sci U S A 115:E2085-E2094
Miller, M L; Ren, Y; Szutorisz, H et al. (2018) Ventral striatal regulation of CREM mediates impulsive action and drug addiction vulnerability. Mol Psychiatry 23:1328-1335
Monteggia, Lisa M; Heimer, Hakon; Nestler, Eric J (2018) Meeting Report: Can We Make Animal Models of Human Mental Illness? Biol Psychiatry 84:542-545

Showing the most recent 10 out of 312 publications