A common finding from studies of drug abuse is that exposure to the context in which drug use occurs can trigger relapse, resulting in an increase in drug-seeking behavior and subsequent drug use. A major goal of interventions designed to eliminate drug seeking therefore must be to eliminate the ability of contextual cues to elicit memories that result in relapse. Behavioral and neurobiological approaches to memory have identified extinction, in which the relation between the context and the drug is severed, as a way to eliminate conditioned behavior. A major challenge for extinction-based therapies, however, is that extinguished behavior often returns with time or after re-exposure to the drug. Thus, extinction-based behavioral interventions must focus on ways to enhance the development of extinction, as well as methods to make the extinction memory long- lasting, causing persistent suppression of the original context-drug association. At a molecular level, studies of memory and extinction have demonstrated the necessity of gene transcription for long-term memory storage. Our research has found that regulation of gene transcription necessary for long-term memory formation involves the concerted action of multiple transcription factors and cofactors that interact with chromatin, a protein complex that packages DNA. Chromatin modification is a main mechanism of epigenetic gene regulation, which is emerging as a major molecular pathway involved in the transcriptional regulation of gene expression required for synaptic plasticity and memory storage. Epigenetic gene regulation has been shown to underlie persistent long-term changes at the cellular level as well as the behavioral level. Importantly, in animal models of addiction, chronic drug exposure induces stable chromatin modification resulting in maintained gene expression, which is thought to drive persistent changes in behavior. Considering the substantial overlap in the circuitry involved in drug addiction and learning and memory pathways, the focus of this grant application is to modulate learning and memory pathways in order to extinguish context-drug associated memories. We will use a combined behavioral, pharmacological, and genetic strategy to examine ways to enhance the development of extinction of cocaine-induced conditioned place preferences to cause persistent suppression of the original context-drug association.
In Specific Aim 1, we will determine the best temporal pattern of context presentations to extinguish context-cocaine associations using the conditioned place preference (CPP) procedure.
In Specific Aim 2, we will examine the behavioral and molecular consequences of administration of systemic or intra-hippocampal drugs that relax chromatin structure, thereby enhancing gene transcription during extinction of cocaine-induced CPP.
In Specific Aim 3, we will examine the effects of inhibiting a chromatin modifying enzyme called CREB-binding protein (CBP), which relaxes chromatin structure, through genetic mutations and focal deletions of CBP, thereby inhibiting transcription during extinction of cocaine-induced CPP. This novel approach promises to elucidate behavioral and epigenetic mechanisms of extinction and will allow us to identify further molecular targets and brain systems for pharmacological interventions. ? ?

Public Health Relevance

Cocaine addiction is a major public health problem in the United States. In this project, we examine behavioral and pharmacological interventions in a mouse model of cocaine seeking that may help reverse cocaine seeking and reduce relapse. ? ? ?

Agency
National Institute of Health (NIH)
Institute
National Institute on Drug Abuse (NIDA)
Type
Research Project (R01)
Project #
1R01DA025922-01
Application #
7578155
Study Section
Special Emphasis Panel (ZDA1-RXL-E (08))
Program Officer
Satterlee, John S
Project Start
2008-09-15
Project End
2013-05-31
Budget Start
2008-09-15
Budget End
2009-05-31
Support Year
1
Fiscal Year
2008
Total Cost
$400,479
Indirect Cost
Name
Oregon Health and Science University
Department
Other Basic Sciences
Type
Schools of Medicine
DUNS #
096997515
City
Portland
State
OR
Country
United States
Zip Code
97239
Malvaez, Melissa; Greenfield, Venuz Y; Matheos, Dina P et al. (2018) Habits Are Negatively Regulated by Histone Deacetylase 3 in the Dorsal Striatum. Biol Psychiatry 84:383-392
Alaghband, Yasaman; Kramár, Enikö; Kwapis, Janine L et al. (2018) CREST in the Nucleus Accumbens Core Regulates Cocaine Conditioned Place Preference, Cocaine-Seeking Behavior, and Synaptic Plasticity. J Neurosci 38:9514-9526
Williams, Amy R; Lattal, K Matthew (2018) Rapid reacquisition of contextual fear following extinction in mice: effects of amount of extinction, acute ethanol withdrawal, and ethanol intoxication. Psychopharmacology (Berl) :
López, Alberto J; Jia, Yousheng; White, André O et al. (2018) Medial habenula cholinergic signaling regulates cocaine-associated relapse-like behavior. Addict Biol :
Hitchcock, Leah N; Lattal, K Matthew (2018) Involvement of the dorsal hippocampus in expression and extinction of cocaine-induced conditioned place preference. Hippocampus 28:226-238
Pizzimenti, Christie L; Navis, Tom M; Lattal, K Matthew (2017) Persistent effects of acute stress on fear and drug-seeking in a novel model of the comorbidity between post-traumatic stress disorder and addiction. Learn Mem 24:422-431
Alaghband, Yasaman; Kwapis, Janine L; López, Alberto J et al. (2017) Distinct roles for the deacetylase domain of HDAC3 in the hippocampus and medial prefrontal cortex in the formation and extinction of memory. Neurobiol Learn Mem 145:94-104
Kwapis, Janine L; Alaghband, Yasaman; López, Alberto J et al. (2017) Context and Auditory Fear are Differentially Regulated by HDAC3 Activity in the Lateral and Basal Subnuclei of the Amygdala. Neuropsychopharmacology 42:1284-1294
López, Alberto J; Kramár, Enikö; Matheos, Dina P et al. (2016) Promoter-Specific Effects of DREADD Modulation on Hippocampal Synaptic Plasticity and Memory Formation. J Neurosci 36:3588-99
Abraham, Antony D; Neve, Kim A; Lattal, K Matthew (2016) Activation of D1/5 Dopamine Receptors: A Common Mechanism for Enhancing Extinction of Fear and Reward-Seeking Behaviors. Neuropsychopharmacology 41:2072-81

Showing the most recent 10 out of 52 publications