One strategy for treating human addiction is to target memory processes that underlie addictive behaviors. Repeated drug use establishes drug-related memories. When these drug-related memories are recalled, as occurs when the organism is re-exposed to drug-associated cues, context, or the drug itself, those memories are reconsolidated to maintain or strengthen them. The medial prefrontal cortex (mPFC) is a key contributor to relapse to cocaine-seeking behavior in humans and reinstatement behavior in rodents. Increased excitatory output from mPFC pyramidal neurons to the nucleus accumbens (NAc) is thought to underlie relapse. In rodents, this increased output is found after 5 cocaine injections and after 45 days of withdrawal from cocaine self-administration, implicating a key role for the mPFC in the maintenance of cocaine-associated memories. However, we do not know the mechanisms by which the mPFC contributes to the expression of these memories. Output from the mPFC is powerfully regulated by parvalbumin (PV)-fast-spiking GABAergic interneurons, the majority of which are surrounded by specialized extracellular matrix structures that form perineuronal nets (PNNs). PNNs envelope certain neurons during development and appear to stabilize synapses, reducing plasticity in neurons during adulthood. However, PNNs can be removed during adulthood to re-establish plasticity or to modify plasticity by other imposing stimuli. We have discovered that removal of PNNs within the rat prelimbic mPFC (PL mPFC) decreases the reconsolidation of cocaine-associated memories as tested with conditioned place preference (CPP; hereafter called cocaine CPP memory). Repeated cocaine exposure has the opposite effect: it increases PNN intensity, and this intensity is positively correlated with behavior, suggesting that PNN intensity in the PL mPFC may serve as a predictor of cocaine- induced behavior. A general layout of our experiments is as follows: Establish cocaine CPP memory? reactivate cocaine CPP memory PNNs? reconsolidate cocaine CPP memory? subsequent test for reinstatement of cocaine CPP memory to see if it is maintained or diminished. We propose that removal of PNNs from the PL mPFC prevents the maintenance of cocaine CPP memory via diminished memory reconsolidation. However, we do not know the mechanisms by which PNN removal decreases memory reconsolidation. We hypothesize that removal of PNNs within the PL mPFC modifies cocaine-induced plasticity during the reconsolidation of a cocaine CPP memory. We further hypothesize that reconsolidation is mediated by PNNs through altered activity of inhibitory interneurons and pyramidal neurons in the PL mPFC. We will train rats for cocaine-induced CPP in the presence and absence of PNNs and define the dynamic changes in PNN-surrounded neurons in the PL mPFC 1) just prior to and after memory reactivation; and 2) just prior to and after cocaine-induced reinstatement. We will use behavioral, electrophysiological, morphological, and confocal and electron microscopic approaches to test our hypotheses. PNNs are a highly novel target for dissecting events critical for cocaine-induced plasticity and the maintenance of cocaine-associated memories. Our findings will have potentially far-reaching consequences for understanding how already-formed cocaine memories can be disrupted by targeting PNN-surrounded neurons within the mPFC.

Public Health Relevance

The proposed studies will assess how specific neurons in the frontal cortex of rats regulate memories associated with cocaine. In particular, we wish to determine how these cocaine-associated memories that develop after repeated exposure to cocaine can be diminished by manipulating a subset of neurons in the frontal cortex. These studies have translational potential in humans because disruption of cocaine-related memories could help break the cycle of relapse.

Agency
National Institute of Health (NIH)
Institute
National Institute on Drug Abuse (NIDA)
Type
Research Project (R01)
Project #
5R01DA040965-04
Application #
9636529
Study Section
Biobehavioral Regulation, Learning and Ethology Study Section (BRLE)
Program Officer
Sorensen, Roger
Project Start
2016-04-15
Project End
2019-06-30
Budget Start
2019-02-01
Budget End
2019-06-30
Support Year
4
Fiscal Year
2019
Total Cost
Indirect Cost
Name
Washington State University
Department
Veterinary Sciences
Type
Schools of Veterinary Medicine
DUNS #
041485301
City
Pullman
State
WA
Country
United States
Zip Code
99164
Dingess, P M; Harkness, J H; Slaker, M et al. (2018) Consumption of a High-Fat Diet Alters Perineuronal Nets in the Prefrontal Cortex. Neural Plast 2018:2108373
Su, Weiping; Matsumoto, Steven; Sorg, Barbara et al. (2018) Distinct roles for hyaluronan in neural stem cell niches and perineuronal nets. Matrix Biol :
Dingess, Paige M; Thakar, Amit; Zhang, Zhaojie et al. (2018) High-Salt Exposure During Perinatal Development Enhances Stress Sensitivity. Dev Neurobiol 78:1131-1145
Slaker, Megan L; Jorgensen, Emily T; Hegarty, Deborah M et al. (2018) Cocaine Exposure Modulates Perineuronal Nets and Synaptic Excitability of Fast-Spiking Interneurons in the Medial Prefrontal Cortex. eNeuro 5:
Blacktop, Jordan M; Sorg, Barbara A (2018) Perineuronal nets in the lateral hypothalamus area regulate cue-induced reinstatement of cocaine-seeking behavior. Neuropsychopharmacology :
Harkness, John H; Bushana, Priyanka N; Todd, Ryan P et al. (2018) Sleep disruption elevates oxidative stress in parvalbumin-positive cells of the rat cerebral cortex. Sleep :
Dingess, Paige M; Darling, Rebecca A; Derman, Rifka C et al. (2017) Structural and Functional Plasticity within the Nucleus Accumbens and Prefrontal Cortex Associated with Time-Dependent Increases in Food Cue-Seeking Behavior. Neuropsychopharmacology 42:2354-2364
Blacktop, Jordan M; Todd, Ryan P; Sorg, Barbara A (2017) Role of perineuronal nets in the anterior dorsal lateral hypothalamic area in the acquisition of cocaine-induced conditioned place preference and self-administration. Neuropharmacology 118:124-136
Polter, Abigail M; Barcomb, Kelsey; Chen, Rudy W et al. (2017) Constitutive activation of kappa opioid receptors at ventral tegmental area inhibitory synapses following acute stress. Elife 6:
Sorg, Barbara A; Berretta, Sabina; Blacktop, Jordan M et al. (2016) Casting a Wide Net: Role of Perineuronal Nets in Neural Plasticity. J Neurosci 36:11459-11468

Showing the most recent 10 out of 11 publications