1
|
Guo B, Song H, Fan J, Wang B, Chen L, Hu Q, Yin Y. The NR2B-targeted intervention alleviates the neuronal injuries at the sub-acute stage of cerebral ischemia: an exploration of stage-dependent strategy against ischemic insults. Exp Brain Res 2023; 241:2735-2750. [PMID: 37845379 DOI: 10.1007/s00221-023-06717-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 09/30/2023] [Indexed: 10/18/2023]
Abstract
Stroke is reported to be the second leading cause of death worldwide, among which ischemic stroke has fourfold greater incidence than intracerebral hemorrhage. Excitotoxicity induced by NMDAR plays a central role in ischemic stroke-induced neuronal death. However, intervention targeted NMDARs against ischemic stroke has failed, which may result from the complex composition of NMDARs and the dynamic changes of their subunits. In this current study, the levels of NR1, NR2A and NR2B subunits of NMDARs were observed upon different time points during the reperfusion after 1 h ischemia with the western blot assay. It was found that the changes of NR1 subunit were only detected after ischemia 1 h/reperfusion 1 day (1 d). While, the changes of NR2A and NR2B subunits may last to ischemia 1 h/reperfusion 7 day(7 d), indicating that NR2subunits may be a potential target for ischemia-reperfusion injuries at the sub-acute stage of ischemic stroke. Simultaneously, mitochondrial injuries in neurons were investigated with transmission electron microscopy (TEM), and mitochondrial dysfunction was evaluated with mitochondrial membrane proteins oxidative respiratory chain complex and OCR. When the antagonist of NMDARs was used before ischemic exposure, the neuronal mitochondrial dysfunction was alleviated, suggesting that these aberrant deviations of NMDARs from basal levels led to mitochondrial dysfunction. Furthermore, when the antagonist of NR2B was administrated intracerebroventricularly at the sub-acute cerebral ischemia, the volume of cerebral infarct region was decreased and the neural functions were improved. To sum up, the ratio of NR2B-containing NMDARs is vital for mitochondrial homeostasis and then neuronal survival. NR2B-targeted intervention should be chosen at the sub-acute stage of cerebral ischemia.
Collapse
Affiliation(s)
- Bei Guo
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, #10 You An Men Wai Xi Tou Tiao, Beijing, 100069, People's Republic of China
| | - Huimeng Song
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, #10 You An Men Wai Xi Tou Tiao, Beijing, 100069, People's Republic of China
| | - Jiahui Fan
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, #10 You An Men Wai Xi Tou Tiao, Beijing, 100069, People's Republic of China
| | - Bin Wang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, #10 You An Men Wai Xi Tou Tiao, Beijing, 100069, People's Republic of China
| | - Lingyi Chen
- John Bapst Memorial High School, Bangor, CA, USA
| | - Qiandai Hu
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, #10 You An Men Wai Xi Tou Tiao, Beijing, 100069, People's Republic of China
| | - Yanling Yin
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, #10 You An Men Wai Xi Tou Tiao, Beijing, 100069, People's Republic of China.
| |
Collapse
|
2
|
Montemitro C, Angebrandt A, Wang TY, Pettorruso M, Abulseoud OA. Mechanistic insights into the efficacy of memantine in treating certain drug addictions. Prog Neuropsychopharmacol Biol Psychiatry 2021; 111:110409. [PMID: 34324921 DOI: 10.1016/j.pnpbp.2021.110409] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 06/23/2021] [Accepted: 07/22/2021] [Indexed: 01/11/2023]
Abstract
The deleterious effects of the drug addiction epidemic are compounded by treatment strategies that are only marginally efficacious. Memantine is a unique glutamatergic medication with proven ability to attenuate drug addiction in preclinical models. However, clinical translational studies are inconsistent. In this review, we summarize preclinical evidences and clinical trials that investigated the efficacy of memantine in treating patients with alcohol, opiate, cocaine, and nicotine use disorders and discuss the results from a mechanistic point of view. Memantine has shown efficacy in reducing alcohol and opiate craving, consumption, and withdrawal severity. However, in cocaine and nicotine use disorders, memantine did not have significant effect on cravings or consumption. Additionally, memantine was associated with increased subjective effects of alcohol, cocaine, and nicotine. We discuss possible mechanisms behind this variability. Since memantine transiently blocks NMDA receptors and protects neurons from overstimulation by excessive synaptic glutamate, its efficacy should be observed in drug phases that cause hyperglutamatergic states, while hypoglutamatergic drug use states would not resolve with blocking NMDA receptors. Second, memantine pharmacokinetic studies have been done in rodents and healthy volunteers, but not in patients with substance use disorder. Memantine, opiates, cocaine, and nicotine share the same transporter family at the blood brain barrier. This shared transport mechanism could impact brain concentrations of memantine and its effects. In conclusion, memantine remains an intriguing compound in our pharmacopeia with controversial results in treating certain aspects of drug addiction. Further studies are needed to understand the clinical and biological correlates of its efficacy.
Collapse
Affiliation(s)
- Chiara Montemitro
- Neuroimaging Research Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, USA; Department of Neuroscience & Imaging, Università degli Studi G. d'Annunzio Chieti e Pescara, Italy.
| | - Alexandra Angebrandt
- Neuroimaging Research Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Tzu-Yun Wang
- Neuroimaging Research Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, USA; Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Mauro Pettorruso
- Department of Neuroscience & Imaging, Università degli Studi G. d'Annunzio Chieti e Pescara, Italy
| | - Osama A Abulseoud
- Neuroimaging Research Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, USA; Department of Psychiatry and Psychology, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ, USA.
| |
Collapse
|
3
|
GSK3β Activity in Reward Circuit Functioning and Addiction. NEUROSCI 2021. [DOI: 10.3390/neurosci2040033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Glycogen synthase kinase-3β (GSK3β), primarily described as a regulator of glycogen metabolism, is a molecular hub linking numerous signaling pathways and regulates many cellular processes like cytoskeletal rearrangement, cell migration, apoptosis, and proliferation. In neurons, the kinase is engaged in molecular events related to the strengthening and weakening of synapses, which is a subcellular manifestation of neuroplasticity. Dysregulation of GSK3β activity has been reported in many neuropsychiatric conditions, like schizophrenia, major depressive disorder, bipolar disorder, and Alzheimer’s disease. In this review, we describe the kinase action in reward circuit-related structures in health and disease. The effect of pharmaceuticals used in the treatment of addiction in the context of GSK3β activity is also discussed.
Collapse
|
4
|
Smaga I, Wydra K, Piechota M, Caffino L, Fumagalli F, Sanak M, Filip M. Cocaine abstinence modulates NMDA receptor subunit expression: An analysis of the GluN2B subunit in cocaine-seeking behavior. Prog Neuropsychopharmacol Biol Psychiatry 2021; 109:110248. [PMID: 33485963 DOI: 10.1016/j.pnpbp.2021.110248] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 12/28/2020] [Accepted: 01/14/2021] [Indexed: 02/06/2023]
Abstract
Cocaine use disorder develops in part due to the strong associations formed between drugs and the stimuli associated with drug use. Recently, treatment strategies including manipulations of drug-associated memories have been investigated, and the possibility of interfering with N-methyl-d-aspartate (NMDA)-mediated neurotransmission may represent an important option. The aim of this study was to examine the significance of the NMDA receptor subunit GluN2B at the molecular level (the expression of the GluN2B subunit, the Grin2B gene and the association of GluN2B with postsynaptic density protein 95 (PSD95)) in the brain structures of rats with a history of cocaine self-administration after i) cocaine abstinence with extinction training or ii) cocaine abstinence without instrumental tasks, as well as at the pharmacological level (peripheral or intracranial administration of CP 101,606, a GluN2B subunit antagonist during the cocaine- or cue-induced reinstatement). The GluN2B subunit levels and the GluN2B/PSD95 complex levels were either increased in the ventral hippocampus (vHIP) with higher levels of Grin2B gene expression in the HIP or decreased in the dorsal striatum (dSTR) after cocaine abstinence with extinction training. Moreover, CP 101,606, a GluN2B subunit antagonist, administered peripherally, attenuated the reinstatement of active lever presses induced by a priming dose of cocaine or by drug-associated conditioned stimuli, while injection into the vHIP reduced the cocaine- or cue with the subthreshold dose of cocaine-induced reinstatement. In cocaine abstinence without instrumental tasks, an increase in the GluN2B subunit levels and the level of the GluN2B/PSD95 complex in the dSTR was observed in rats that had previously self-administered cocaine. In conclusion, cocaine abstinence with extinction training seems to be associated with the up-regulation of the hippocampal GluN2B subunits, which seems to control cocaine-seeking behavior.
Collapse
Affiliation(s)
- Irena Smaga
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna 12, PL 31-343 Kraków, Poland.
| | - Karolina Wydra
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna 12, PL 31-343 Kraków, Poland
| | - Marcin Piechota
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Molecular Neuropharmacology, Smętna 12, 31-343 Kraków, Poland
| | - Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy
| | - Marek Sanak
- Department of Internal Medicine, Jagiellonian University Medical College, Skawińska 8, PL 31-066 Kraków, Poland
| | - Małgorzata Filip
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna 12, PL 31-343 Kraków, Poland
| |
Collapse
|
5
|
Rulan D, Zhenbang Y, Yipu Z, Yuan G, Galaj E, Xiaorui S, Wenshuya L, Jiaqi L, Yan Z, Chang Y, Xi Y, Li S, Yixiao L, Haishui S. Exogenous SO 2 donor treatment impairs reconsolidation of drug reward memory in mice. Eur J Pharmacol 2021; 896:173911. [PMID: 33503460 DOI: 10.1016/j.ejphar.2021.173911] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/16/2021] [Accepted: 01/21/2021] [Indexed: 12/25/2022]
Abstract
Substance-related and addictive disorders (SRADs) are characterized by compulsive drug use and recurrent relapse. The persistence of pathological drug-related memories indisputably contributes to a high propensity to relapse. Hence, strategies to disrupt reconsolidation of drug reward memory are currently being pursued as potential anti-relapse interventions. Sulfur dioxide (SO2), acting as a potential gaseous molecule, endogenously derives from sulfur amino acid and can exert significant neural regulatory effects. However, the role of SO2 in reconsolidation of drug memory has not been determined. In the present study, we used morphine- or cocaine-induced conditioned place preference (CPP) mouse models with retrieval to investigate the effects of exogenous SO2 donor treatment on reconsolidation of drug reward memory. We found that administration of SO2 donor immediately after the retrieval impaired the expression of morphine or cocaine CPP. Furthermore, the exogenous SO2 donor treatment 6 h post-retrieval or in the absence of retrieval had no effect on drug reward memory and the expression of CPP. SO2 itself did not produce aversive effects nor did it acutely block morphine CPP. Our results indicate that exogenous SO2 impairs reconsolidation of drug reward memory rather than inhibits the expression of drug reward memory. As such, SO2 holds potential for the treatment and prevention of SRADs and should be studied further.
Collapse
Affiliation(s)
- Ding Rulan
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410081, China; Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yang Zhenbang
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Zhang Yipu
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Gao Yuan
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Ewa Galaj
- National Institute on Drug Abuse, Molecular Targets and Medications Discovery Branch, Baltimore, MD, USA
| | - Shi Xiaorui
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Li Wenshuya
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Luo Jiaqi
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Zhang Yan
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yang Chang
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410081, China
| | - Yin Xi
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410081, China; Department of Functional Region of Diagnosis, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Song Li
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Luo Yixiao
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410081, China.
| | - Shi Haishui
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China; Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medicinal University, 050017, China.
| |
Collapse
|
6
|
Wideman CE, Nguyen J, Jeffries SD, Winters BD. Fluctuating NMDA Receptor Subunit Levels in Perirhinal Cortex Relate to Their Dynamic Roles in Object Memory Destabilization and Reconsolidation. Int J Mol Sci 2020; 22:ijms22010067. [PMID: 33374645 PMCID: PMC7793502 DOI: 10.3390/ijms22010067] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/19/2020] [Accepted: 12/20/2020] [Indexed: 01/22/2023] Open
Abstract
Reminder cues can destabilize consolidated memories, rendering them modifiable before they return to a stable state through the process of reconsolidation. Older and stronger memories resist this process and require the presentation of reminders along with salient novel information in order to destabilize. Previously, we demonstrated in rats that novelty-induced object memory destabilization requires acetylcholine (ACh) activity at M1 muscarinic receptors. Other research predominantly has focused on glutamate, which modulates fear memory destabilization and reconsolidation through GluN2B- and GluN2A-containing NMDARs, respectively. In the current study, we demonstrate the same dissociable roles of GluN2B- and N2A-containing NMDARs in perirhinal cortex (PRh) for object memory destabilization and reconsolidation when boundary conditions are absent. However, neither GluN2 receptor subtype was required for novelty-induced destabilization of remote, resistant memories. Furthermore, GluN2B and GluN2A subunit proteins were upregulated selectively in PRh 24 h after learning, but returned to baseline by 48 h, suggesting that NMDARs, unlike muscarinic receptors, have only a temporary role in object memory destabilization. Indeed, activation of M1 receptors in PRh at the time of reactivation effectively destabilized remote memories despite inhibition of GluN2B-containing NMDARs. These findings suggest that cholinergic activity at M1 receptors overrides boundary conditions to destabilize resistant memories when other established mechanisms are insufficient.
Collapse
|
7
|
Bender BN, Torregrossa MM. Molecular and circuit mechanisms regulating cocaine memory. Cell Mol Life Sci 2020; 77:3745-3768. [PMID: 32172301 PMCID: PMC7492456 DOI: 10.1007/s00018-020-03498-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 02/04/2020] [Accepted: 03/02/2020] [Indexed: 01/27/2023]
Abstract
Risk of relapse is a major challenge in the treatment of substance use disorders. Several types of learning and memory mechanisms are involved in substance use and have implications for relapse. Associative memories form between the effects of drugs and the surrounding environmental stimuli, and exposure to these stimuli during abstinence causes stress and triggers drug craving, which can lead to relapse. Understanding the neural underpinnings of how these associations are formed and maintained will inform future advances in treatment practices. A large body of research has expanded our knowledge of how associative memories are acquired and consolidated, how they are updated through reactivation and reconsolidation, and how competing extinction memories are formed. This review will focus on the vast literature examining the mechanisms of cocaine Pavlovian associative memories with an emphasis on the molecular memory mechanisms and circuits involved in the consolidation, reconsolidation, and extinction of these memories. Additional research elucidating the specific signaling pathways, mechanisms of synaptic plasticity, and epigenetic regulation of gene expression in the circuits involved in associative learning will reveal more distinctions between consolidation, reconsolidation, and extinction learning that can be applied to the treatment of substance use disorders.
Collapse
Affiliation(s)
- Brooke N Bender
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, USA
- Center for Neuroscience, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15213, USA
| | - Mary M Torregrossa
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, USA.
- Center for Neuroscience, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
8
|
Drame ML, Balaet M, Lee JLC. Memory reconsolidation impairments in sign-tracking to an audiovisual compound stimulus. Behav Brain Res 2020; 393:112774. [PMID: 32553756 DOI: 10.1016/j.bbr.2020.112774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/11/2020] [Accepted: 05/26/2020] [Indexed: 10/24/2022]
Abstract
Studies of memory reconsolidation of pavlovian memories have typically employed unimodal conditioned stimuli, despite the use of multimodal compound stimuli in other settings. Here we studied sign-tracking behaviour to a compound audiovisual stimulus. First, we observed not unexpectedly that sign-tracking was poorer to the audiovisual compound than to unimodal visual stimuli. Then, we showed that, depending on the parameters of compound stimulus re-exposure at memory reactivation, systemic MK-801 treatment either impaired extinction to improve sign-tracking at test, or disrupted reconsolidation to impair test behaviour. When memory reactivation consisted of re-exposure to only the auditory component of the compound stimulus, we observed sign-tracking impairments following MK-801 treatment, but only under certain test conditions. This was in contrast to the consistent impairment following reactivation with the full audiovisual compound. Moreover, the parameters of auditory stimulus presentation to enable MK-801-induced impairment at test varied depending on whether the stimulus was presented within or outside the training context. These findings suggest that behaviour under the control of appetitive pavlovian compound stimuli can be modulated by targeting both extinction and reconsolidation, and that it is not necessary to re-expose to the full compound stimulus in order to achieve a degree of modulation of behaviour.
Collapse
Affiliation(s)
| | - Maria Balaet
- University of Birmingham, Birmingham, B15 2TT, UK
| | | |
Collapse
|
9
|
Braidy N, Villalva MD, van Eeden S. Sobriety and Satiety: Is NAD+ the Answer? Antioxidants (Basel) 2020; 9:antiox9050425. [PMID: 32423100 PMCID: PMC7278809 DOI: 10.3390/antiox9050425] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/03/2020] [Accepted: 05/05/2020] [Indexed: 02/07/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential pyridine nucleotide that has garnered considerable interest in the last century due to its critical role in cellular processes associated with energy production, cellular protection against stress and longevity. Research in NAD+ has been reinvigorated by recent findings that components of NAD+ metabolism and NAD-dependent enzymes can influence major signalling processes associated with the neurobiology of addiction. These studies implicate raising intracellular NAD+ levels as a potential target for managing and treating addictive behaviour and reducing cravings and withdrawal symptoms in patients with food addiction and/or substance abuse. Since clinical studies showing the use of NAD+ for the treatment of addiction are limited, this review provides literature evidence that NAD+ can influence the neurobiology of addiction and may have benefits as an anti-addiction intervention.
Collapse
Affiliation(s)
- Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW 2052, Australia;
- Correspondence:
| | - Maria D. Villalva
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW 2052, Australia;
| | - Sam van Eeden
- Centre for Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 4NS, UK;
| |
Collapse
|
10
|
Li YC, Panikker P, Xing B, Yang SS, Alexandropoulos C, McEachern EP, Akumuo R, Zhao E, Gulchina Y, Pletnikov MV, Urs NM, Caron MG, Elefant F, Gao WJ. Deletion of Glycogen Synthase Kinase-3β in D 2 Receptor-Positive Neurons Ameliorates Cognitive Impairment via NMDA Receptor-Dependent Synaptic Plasticity. Biol Psychiatry 2020; 87:745-755. [PMID: 31892408 PMCID: PMC7103512 DOI: 10.1016/j.biopsych.2019.10.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Cortical dopaminergic systems are critically involved in prefrontal cortex (PFC) functions, especially in working memory and neurodevelopmental disorders such as schizophrenia. GSK-3β (glycogen synthase kinase-3β) is highly associated with cAMP (cyclic adenosine monophosphate)-independent dopamine D2 receptor (D2R)-mediated signaling to affect dopamine-dependent behaviors. However, the mechanisms underlying the GSK-3β modulation of cognitive function via D2Rs remains unclear. METHODS This study explored how conditional cell-type-specific ablation of GSK-3β in D2R+ neurons (D2R-GSK-3β-/-) in the brain affects synaptic function in the medial PFC (mPFC). Both male and female (postnatal days 60-90) mice, including 140 D2R, 24 D1R, and 38 DISC1 mice, were used. RESULTS This study found that NMDA receptor (NMDAR) function was significantly increased in layer V pyramidal neurons in mPFC of D2R-GSK-3β-/- mice, along with increased dopamine modulation of NMDAR-mediated current. Consistently, NR2A and NR2B protein levels were elevated in mPFC of D2R-GSK-3β-/- mice. This change was accompanied by a significant increase in enrichment of activator histone mark H3K27ac at the promoters of both Grin2a and Grin2b genes. In addition, altered short- and long-term synaptic plasticity, along with an increased spine density in layer V pyramidal neurons, were detected in D2R-GSK-3β-/- mice. Indeed, D2R-GSK-3β-/- mice also exhibited a resistance of working memory impairment induced by injection of NMDAR antagonist MK-801. Notably, either inhibiting GSK-3β or disrupting the D2R-DISC1 complex was able to reverse the mutant DISC1-induced decrease of NMDAR-mediated currents in the mPFC. CONCLUSIONS This study demonstrates that GSK-3β modulates cognition via D2R-DISC1 interaction and epigenetic regulation of NMDAR expression and function.
Collapse
Affiliation(s)
- Yan-Chun Li
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania.
| | - Priyalakshmi Panikker
- Department of Biology, Drexel University College of Medicine, Philadelphia, PA 19104, USA
| | - Bo Xing
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Sha-Sha Yang
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Cassandra Alexandropoulos
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Erin P McEachern
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Rita Akumuo
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Elise Zhao
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Yelena Gulchina
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Mikhail V. Pletnikov
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Nikhil M. Urs
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA
| | - Marc G. Caron
- Departments of Cell Biology, Neurobiology, and Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Felice Elefant
- Department of Biology, Drexel University College of Medicine, Philadelphia, PA 19104, USA
| | - Wen-Jun Gao
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania.
| |
Collapse
|
11
|
Zhu S, Chen P, Chen M, Ruan J, Ren W, Zhang X, Gao Y, Li Y. Alcohol inhibits morphine/cocaine reward memory acquisition and reconsolidation in rats. Psychopharmacology (Berl) 2020; 237:1043-1053. [PMID: 31912191 DOI: 10.1007/s00213-019-05433-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 12/11/2019] [Indexed: 01/05/2023]
Abstract
RATIONALE AND OBJECTIVE Alcohol is a recreational substance that is generally socially acceptable and legal in most areas worldwide. An alcohol overdose will produce an inhibitory effect on the brain and impair cognition and memory. In this study, we examined the effect of alcohol on the acquisition, consolidation, and reconsolidation of drug reward memory induced by morphine and cocaine in rats. METHODS Rats were trained to acquire morphine sulfate (10 mg/kg, s.c.) and cocaine (10 mg/kg, i.p.) conditioned place preference (CPP) via an unbiased CPP paradigm. Vehicle or alcohol (0.25, 0.75, 1.5 g/kg, i.p.) was administered at various time-points, including 30 min before each CPP conditioning session (acquisition), immediately after each CPP conditioning session (consolidation), immediately after the reactivation of CPP (reconsolidation with re-exposure), or without reactivation to the drug-paired context (reconsolidation without re-exposure). Conditioning scores were recorded before or after each conditioning session or memory reactivation. RESULTS Alcohol at a dose of 1.5 g/kg but not 0.25 g/kg or 0.75 g/kg significantly inhibited the acquisition and reconsolidation of morphine- and cocaine-associated memory. In contrast, alcohol had no effect on the consolidation of morphine- or cocaine-induced CPP. CONCLUSIONS The results suggested that pre-exposure alcohol dose-dependently attenuated morphine- or cocaine-induced place preference and prevented drug reinstatement in rats by disrupting memory reconsolidation, which may be explained by the inhibitory effect of alcohol on dopaminergic and glutamatergic neurotransmission.
Collapse
Affiliation(s)
- Shimin Zhu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, and Wuhan University School of Pharmaceutical Sciences, Wuhan, 430071, Hubei, China
| | - Pingping Chen
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, and Wuhan University School of Pharmaceutical Sciences, Wuhan, 430071, Hubei, China
| | - Mingzhu Chen
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, and Wuhan University School of Pharmaceutical Sciences, Wuhan, 430071, Hubei, China
| | - Jiawei Ruan
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, and Wuhan University School of Pharmaceutical Sciences, Wuhan, 430071, Hubei, China
| | - Wanying Ren
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, and Wuhan University School of Pharmaceutical Sciences, Wuhan, 430071, Hubei, China
| | - Xinyu Zhang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, and Wuhan University School of Pharmaceutical Sciences, Wuhan, 430071, Hubei, China
| | - Yang Gao
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, and Wuhan University School of Pharmaceutical Sciences, Wuhan, 430071, Hubei, China
| | - Yanqin Li
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, and Wuhan University School of Pharmaceutical Sciences, Wuhan, 430071, Hubei, China.
| |
Collapse
|
12
|
Correia C, Romieu P, Olmstead MC, Befort K. Can cocaine-induced neuroinflammation explain maladaptive cocaine-associated memories? Neurosci Biobehav Rev 2020; 111:69-83. [PMID: 31935376 DOI: 10.1016/j.neubiorev.2020.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/20/2019] [Accepted: 01/01/2020] [Indexed: 12/19/2022]
Abstract
Persistent and intrusive memories define a number of psychiatric disorders, including posttraumatic stress disorder and substance use disorder. In the latter, memory for drug-paired cues plays a critical role in sustaining compulsive drug use as these are potent triggers of relapse. As with many drugs, cocaine-cue associated memory is strengthened across presentations as cues become reliable predictors of drug availability. Recently, the targeting of cocaine-associated memory through disruption of the reconsolidation process has emerged as a potential therapeutic strategy; reconsolidation reflects the active process by which memory is re-stabilized after retrieval. In addition, a separate line of work reveals that neuroinflammatory markers, regulated by cocaine intake, play a role in memory processes. Our review brings these two literatures together by summarizing recent findings on cocaine-associated reconsolidation and cocaine-induced neuroinflammation. We discuss the interactions between reconsolidation processes and neuroinflammation following cocaine use, concluding with a new perspective on treatment to decrease risk of relapse to cocaine use.
Collapse
Affiliation(s)
- Caroline Correia
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Centre de la Recherche Nationale Scientifique, UMR 7364, Faculté de Psychologie, 12 rue Goethe, F-67000, Strasbourg, France
| | - Pascal Romieu
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Centre de la Recherche Nationale Scientifique, UMR 7364, Faculté de Psychologie, 12 rue Goethe, F-67000, Strasbourg, France
| | - Mary C Olmstead
- Dept. Psychology, Centre for Neuroscience Studies, Queen's University, Kingston ON, K7L 3N6, Canada
| | - Katia Befort
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Centre de la Recherche Nationale Scientifique, UMR 7364, Faculté de Psychologie, 12 rue Goethe, F-67000, Strasbourg, France.
| |
Collapse
|
13
|
Both S, Van Veen RJB, Brom M, Weijenborg PTM. A randomized, placebo-controlled laboratory study of the effects of D-cycloserine on sexual memory consolidation in women. Psychopharmacology (Berl) 2020; 237:1291-1303. [PMID: 31984445 PMCID: PMC7196949 DOI: 10.1007/s00213-020-05457-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 01/10/2020] [Indexed: 11/29/2022]
Abstract
RATIONALE AND OBJECTIVE The aim of this study was to investigate the possible facilitating effect of the partial NMDA receptor agonist D-cycloserine (DCS) on memory consolidation of conditioned sexual responses and to examine the capability of DCS to reduce context-specificity of learning. METHODS In a randomized placebo-controlled double-blind trial, 50 healthy females were exposed to a differential conditioning procedure. Two pictures of a male abdomen were used as conditional stimuli (CSs), of which one (the CS+) was followed by the unconditional stimulus (US), a genital vibrotactile stimulus. After the conditioning session on day 1, participants received either 125 mg of DCS or a placebo. The effects of DCS on affect, sexual arousal and US expectancy in response to the CS+ and CS- were examined 24 h after the conditioning procedure. RESULTS A main effect of DCS was found on affect at the first test trials (p = 0.04, ηp2 = 0.09), and a similar non-significant but trend level effect was found for sexual arousal (p = 0.06, ηp2 = 0.07), which appeared to persist over a longer time (p = 0.07, ηp2 = 0.08). Unexpectedly, ratings of positive affect and sexual arousal in response to both the CS+ and the CS- were higher in the DCS condition compared to the control condition, possibly indicating that DCS administration reduced stimulus specificity. Since the results did not show clear evidence for context learning, we were not able to test effects on context-specificity of learning. CONCLUSION Although largely inconclusive, the results provide tentative support for a facilitating effect of DCS on affect and sexual arousal in response to stimuli that were presented in a sexual conditioning procedure, however, no conclusions can be drawn about effects of DCS on sexual reward learning, since the design and results do not lend themselves to unambiguous interpretation.
Collapse
Affiliation(s)
- S. Both
- Department of Psychosomatic Gynecology and Sexology, Leiden University Medical Center, Poortgebouw-Zuid, 4e etage, Rijnsburgerweg 10, 2333 AA Leiden, The Netherlands
| | - R. J. B. Van Veen
- Department of Psychosomatic Gynecology and Sexology, Leiden University Medical Center, Poortgebouw-Zuid, 4e etage, Rijnsburgerweg 10, 2333 AA Leiden, The Netherlands
| | - M. Brom
- Department of Psychosomatic Gynecology and Sexology, Leiden University Medical Center, Poortgebouw-Zuid, 4e etage, Rijnsburgerweg 10, 2333 AA Leiden, The Netherlands
| | - P. T. M. Weijenborg
- Department of Psychosomatic Gynecology and Sexology, Leiden University Medical Center, Poortgebouw-Zuid, 4e etage, Rijnsburgerweg 10, 2333 AA Leiden, The Netherlands
| |
Collapse
|
14
|
Shi X, von Weltin E, Barr JL, Unterwald EM. Activation of GSK3β induced by recall of cocaine reward memories is dependent on GluN2A/B NMDA receptor signaling. J Neurochem 2019; 151:91-102. [PMID: 31361029 DOI: 10.1111/jnc.14842] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 06/21/2019] [Accepted: 07/26/2019] [Indexed: 12/11/2022]
Abstract
Glycogen synthase kinase-3β (GSK3β) is a critical regulator of the balance between long-term depression and long-term potentiation which is essential for learning and memory. Our previous study demonstrated that GSK3β activity is highly induced during cocaine memory reactivation, and that reconsolidation of cocaine reward memory is attenuated by inhibition of GSK3β. NMDA receptors and protein phosphatase 1 (PP1) are activators of GSK3β. Thus, this study investigated the roles of NMDA receptor subtypes and PP1in the reconsolidation of cocaine contextual reward memory. Cocaine contextual memories were established and evaluated using cocaine conditioned place preference methods. The regulation of GSK3β activity in specific brain areas was assessed by measuring its phosphorylation state using immunoblot assays. Mice underwent cocaine place conditioning for 8 days and were tested for place preference on day 9. Twenty-four hours later, mice were briefly confined to the compartment previous paired with cocaine to reactivate cocaine-associated memories. Administration of the GluN2A- and GluN2B-NMDA receptor antagonists, NVP-AAM077 and ifenprodil, respectively, immediately following recall abrogated an established cocaine place preference, while preventing the activation of GSK3β in the amygdala, nucleus accumbens, and hippocampus during cocaine memory reactivation. PP1 inhibition with okadaic acid also blocked the activation of GSK3β and attenuated a previously established cocaine place preference. These findings suggest that the dephosphorylation of GSK3β that occurred upon activation of cocaine-associated reward memories may be initiated by the activation of PP1 during the induction of NMDA receptor-dependent reconsolidation of cocaine mnemonic traces. Moreover, the importance of NMDA receptors and PP1 in reconsolidation of cocaine memory makes them potential therapeutic targets in treatment of cocaine use disorder and prevention of relapse.
Collapse
Affiliation(s)
- Xiangdang Shi
- Center for Substance Abuse Research and Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Eva von Weltin
- Center for Substance Abuse Research and Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Jeffrey L Barr
- Center for Substance Abuse Research and Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Ellen M Unterwald
- Center for Substance Abuse Research and Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
15
|
Wideman CE, Jardine KH, Winters BD. Involvement of classical neurotransmitter systems in memory reconsolidation: Focus on destabilization. Neurobiol Learn Mem 2018; 156:68-79. [DOI: 10.1016/j.nlm.2018.11.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 10/09/2018] [Accepted: 11/01/2018] [Indexed: 10/27/2022]
|
16
|
Exton-McGuinness MTJ, Milton AL. Reconsolidation blockade for the treatment of addiction: challenges, new targets, and opportunities. Learn Mem 2018; 25:492-500. [PMID: 30115771 PMCID: PMC6097762 DOI: 10.1101/lm.046771.117] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 05/21/2018] [Indexed: 11/25/2022]
Abstract
Addiction is a chronic, relapsing disorder. The progression to pathological drug-seeking is thought to be driven by maladaptive learning processes which store and maintain associative memory, linking drug highs with cues and actions in the environment. These memories can encode Pavlovian associations which link predictive stimuli (e.g., people, places, and paraphernalia) with a hedonic drug high, as well as instrumental learning about the actions required to obtain drug-associated incentives. Learned memories are not permanent however, and much recent interest has been generated in exploiting the process of reconsolidation to erase or significantly weaken maladaptive memories to treat several mental health disorders, including addictions. Normally reconsolidation serves to update and maintain the adaptive relevance of memories, however administration of amnestic agents within the critical "reconsolidation window" can weaken or even erase maladaptive memories. Here we discuss recent advances in the field, including ongoing efforts to translate preclinical reconsolidation research in animal models into clinical practice.
Collapse
Affiliation(s)
| | - Amy L Milton
- Department of Psychology, University of Cambridge, Downing Site, Cambridge CB2 3EB, United Kingdom
| |
Collapse
|
17
|
Kochli DE, Campbell TL, Hollingsworth EW, Lab RS, Postle AF, Perry MM, Mordzinski VM, Quinn JJ. Combined administration of MK-801 and cycloheximide produces a delayed potentiation of fear discrimination memory extinction. Behav Neurosci 2018; 132:99-105. [PMID: 29672107 DOI: 10.1037/bne0000232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Mixed evidence exists regarding the role of N-methyl-D-aspartate (NMDA) receptors in memory reconsolidation. We provide no evidence that NMDA receptors are involved with memory reconsolidation, but instead demonstrate that prereactivation systemic MK-801 injection, combined with postreactivation intrabasolateral amygdala (BLA) cycloheximide infusion, produces a delayed potentiation of extinction learning. These data suggest that an interaction between NMDA antagonism and protein synthesis inhibition may enhance extinction by exerting effects outside of the intended reconsolidation manipulation window. The present work demonstrates a novel pharmacological enhancement of extinction, and underscores the importance of employing proper control procedures in reconsolidation research. (PsycINFO Database Record
Collapse
Affiliation(s)
- Daniel E Kochli
- Department of Psychology and Center for Neuroscience & Behavior, Miami University
| | - Tiffany L Campbell
- Department of Psychology and Center for Neuroscience & Behavior, Miami University
| | | | - Rain S Lab
- Department of Psychology and Center for Neuroscience & Behavior, Miami University
| | - Abagail F Postle
- Department of Psychology and Center for Neuroscience & Behavior, Miami University
| | - Megan M Perry
- Department of Psychology and Center for Neuroscience & Behavior, Miami University
| | | | - Jennifer J Quinn
- Department of Psychology and Center for Neuroscience & Behavior, Miami University
| |
Collapse
|
18
|
Hafenbreidel M, Rafa Todd C, Mueller D. Infralimbic GluN2A-Containing NMDA Receptors Modulate Reconsolidation of Cocaine Self-Administration Memory. Neuropsychopharmacology 2017; 42:1113-1125. [PMID: 28042872 PMCID: PMC5506782 DOI: 10.1038/npp.2016.288] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 12/10/2016] [Accepted: 12/19/2016] [Indexed: 01/01/2023]
Abstract
Addiction is characterized by high relapse susceptibility, and relapse can be triggered by drug-associated cues. Cue presentation induces retrieval of the drug-cue memory, which becomes labile and must be reconsolidated into long-term storage. Repeated unpaired cue presentation, however, promotes extinction. Cue-reactivity can be reduced by blocking reconsolidation or facilitating extinction, which are mediated by NMDA receptors (NMDArs). However, the role of NMDArs in either process following self-administration is unclear. Thus, to determine their role in extinction, rats learned to self-administer cocaine before receiving injections of the NMDAr antagonist CPP immediately after four 45-min extinction sessions. During a subsequent 90-min extinction retention test, CPP-treated rats lever pressed less than saline-treated rats indicating that NMDAr blockade facilitated extinction or disrupted drug-cue memory reconsolidation. In addition, infusing CPP into the infralimbic medial prefrontal cortex (IL-mPFC), a structure implicated in extinction, before four 45-min or immediately after four 30min extinction sessions, had similar results during the extinction retention tests. Next, the GluN2A-selective antagonist NVP or GluN2B-selective antagonist Ro25 was infused into IL-mPFC or nucleus accumbens (NAc) shell, another structure implicated in extinction, after four 45-min extinction sessions. Blocking GluN2A-, but not GluN2B-, containing NMDArs, in IL-mPFC or NAc shell reduced lever pressing during the extinction retention tests. Finally, to dissociate reconsolidation from extinction, NVP was infused into IL-mPFC after four 10-min reactivation sessions, which resulted in reduced lever pressing during the retention test. These results indicate that IL-mPFC GluN2A-containing NMDArs modulate reconsolidation, and suggest a novel treatment strategy, as reducing cue reactivity could limit relapse susceptibility.
Collapse
Affiliation(s)
| | - Carolynn Rafa Todd
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Devin Mueller
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA,Department of Basic Sciences, Ponce Health Sciences University-School of Medicine/Ponce Research Institute, Ponce, Puerto Rico,Department of Basic Sciences, Ponce Health Sciences University-School of Medicine, Ponce Research Institute, PO Box 7004, Ponce 00732-7004, Puerto Rico, Tel: +1 787 840 2575 Ext. 2588, Fax: +1 787 844 1980, E-mail:
| |
Collapse
|
19
|
Nygard SK, Klambatsen A, Balouch B, Quinones-Jenab V, Jenab S. NMDAR dependent intracellular responses associated with cocaine conditioned place preference behavior. Behav Brain Res 2017; 317:218-225. [PMID: 27664298 PMCID: PMC5107324 DOI: 10.1016/j.bbr.2016.09.047] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 09/16/2016] [Accepted: 09/20/2016] [Indexed: 01/16/2023]
Abstract
The aim of this study was to investigate the intracellular responses associated with the acquisition and expression of cocaine-context associations. ERK (extracellular regulated kinase), CREB (cAMP responsive element binding protein), FosB and ΔFosB proteins were of particular interest due to their involvement in cocaine reward and in synaptic plasticity underlying learning and memory. We used the conditioned place preference (CPP) paradigm, which employs a Pavlovian conditioning procedure to establish an association between a drug-paired environment and the drug's rewarding effects, to study the role of these signaling pathways in cocaine-context associations. N-methyl-D-aspartate receptor (NMDAR) antagonism prior to cocaine administration during conditioning blocked the acquisition of cocaine CPP and reduced Nucleus Accumbens (NAc) phosphorylated-ERK (pERK) and phosphorylated CREB (pCREB) levels following the CPP test (drug-free). We also show that cocaine-induced increases in Caudate Putamen (CPu) FosB and ΔFosB levels are decreased after MK-801 pre-treatment during conditioning. In addition, our results provide evidence for the involvement of striatal SIRT (Silent Information Regulator of Transcription) proteins in cocaine-CPP. These results will aid in the advancement of general knowledge about the molecular formation and retrieval of cocaine-associated memories that can be used in the future when designing treatments for cocaine addiction that target both prevention and relapse.
Collapse
Affiliation(s)
- Stephanie K Nygard
- Department of Psychology, Hunter College, CUNY, 695 Park Avenue, New York, NY 10065 USA; Biopsychology and Behavioral Neuroscience Subprogram, Graduate School and University Center, CUNY, 365 Fifth Avenue, New York, NY 10016 USA.
| | - Anthony Klambatsen
- Department of Psychology, Hunter College, CUNY, 695 Park Avenue, New York, NY 10065 USA; Biopsychology and Behavioral Neuroscience Subprogram, Graduate School and University Center, CUNY, 365 Fifth Avenue, New York, NY 10016 USA.
| | - Bailey Balouch
- Department of Psychology, Hunter College, CUNY, 695 Park Avenue, New York, NY 10065 USA.
| | - Vanya Quinones-Jenab
- Department of Psychology, Hunter College, CUNY, 695 Park Avenue, New York, NY 10065 USA; Biopsychology and Behavioral Neuroscience Subprogram, Graduate School and University Center, CUNY, 365 Fifth Avenue, New York, NY 10016 USA.
| | - Shirzad Jenab
- Department of Psychology, Hunter College, CUNY, 695 Park Avenue, New York, NY 10065 USA; Biopsychology and Behavioral Neuroscience Subprogram, Graduate School and University Center, CUNY, 365 Fifth Avenue, New York, NY 10016 USA.
| |
Collapse
|
20
|
Metabolic shift of the kynurenine pathway impairs alcohol and cocaine seeking and relapse. Psychopharmacology (Berl) 2016; 233:3449-59. [PMID: 27475106 DOI: 10.1007/s00213-016-4384-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 07/16/2016] [Indexed: 12/31/2022]
Abstract
RATIONALE The glutamatergic system plays a key role in the maintenance of drug use and development of drug-related conditioned behaviours. In particular, hyper-glutamatergic activity and N-methyl-D-aspartate receptor (NMDAR) activation may drive drug craving and relapse. Inhibition of kynurenine-3-monooxygenase (KMO) shifts the metabolic kynurenine pathway towards production of kynurenic acid, which leads to a reduction of glutamatergic/NMDAR activity via different mechanisms. OBJECTIVES In this study, we investigated whether drug-seeking and relapse behaviour could be modified by the metabolic shift of endogenous kynurenine pathway. METHODS An inhibitor of kynurenine-3-monooxygenase (KMO) Ro61-8048 (4 and 40 mg/kg) and its prodrug JM6 (100 and 200 mg/kg) were tested in two behavioural rat models for drug seeking and relapse-the alcohol deprivation effect (ADE) model in long-term alcohol-drinking rats and the model of cue-induced reinstatement of alcohol- and cocaine-seeking behaviour. RESULTS Our results show that relapse-like alcohol drinking during the ADE was abolished by repeated intraperitoneal administration of Ro61-8048 and significantly reduced by its oral prodrug JM6. Cue-induced reinstatement of both alcohol- and cocaine-seeking behaviour was also abolished by administration of Ro61-8048. CONCLUSIONS Pharmacological enhancement of endogenous kynurenic acid levels provides a novel treatment strategy to interfere with glutamatergic/NMDAR activity as well as with craving and relapse in alcohol-dependent patients and drug addicts.
Collapse
|
21
|
Szumlinski KK, Wroten MG, Miller BW, Sacramento AD, Cohen M, Ben-Shahar O, Kippin TE. Cocaine Self-Administration Elevates GluN2B within dmPFC Mediating Heightened Cue-Elicited Operant Responding. ACTA ACUST UNITED AC 2016; 2. [PMID: 27478879 PMCID: PMC4962921 DOI: 10.21767/2471-853x.100022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Cue-elicited drug-craving correlates with hyperactivity within prefrontal cortex (PFC), which is theorized to result from dysregulated excitatory neurotransmission. The NMDA glutamate receptor is highly implicated in addiction-related neuroplasticity. As NMDA receptor function is regulated critically by its GluN2 subunits, herein, we assayed the relation between incubated cue-elicited cocaine-seeking following extended access to intravenous cocaine (6 h/d; 0.25 mg/infusion for 10 d) and the expression of GluN2A/B receptor subunits within PFC sub regions during early versus late withdrawal (respectively, 3 vs. 30 days). Cocaine-seeking rats exhibited elevated GluN2B expression within the dorsomedial aspect of the PFC (dmPFC); this effect was apparent at both 3 and 30 days withdrawal and occurred in cocaine-experienced rats, regardless of experiencing an extinction test or not. Thus, elevated dmPFC GluN2B expression appears to reflect a pharmacodynamic response to excessive cocaine intake that is independent of the duration of drug withdrawal or re-exposure to drug-taking context. The functional relevance of elevated dmPFC GluN2B expression for drug-seeking was assessed by the local infusion of the prototypical GluN2B-selective antagonist ifenprodil (1.0 µg/side). Ifenprodil did not alter cue-elicited responding in animals with a history of saline self-administration. In contrast, ifenprodil lowered cue-elicited cocaine-seeking, while potentiating cue-elicited sucrose-seeking. Thus, the effects of an intra-dmPFC ifenprodil infusion upon cue reactivity are reinforcer-specific, arguing in favor of targeting GluN2B-containing NMDA receptors as a pharmacological strategy for reducing behavioral reactivity to drug-associated cues with the potential benefit of heightening the reinforcing properties of cues associated with non-drug primary rewards.
Collapse
Affiliation(s)
- Karen K Szumlinski
- Department of Psychological and Brain Sciences & Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Melissa G Wroten
- Department of Psychological and Brain Sciences & Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Bailey W Miller
- Department of Psychological and Brain Sciences & Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Arianne D Sacramento
- Department of Psychological and Brain Sciences & Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Matan Cohen
- Department of Psychological and Brain Sciences & Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Osnat Ben-Shahar
- Department of Psychological and Brain Sciences & Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Tod E Kippin
- Department of Psychological and Brain Sciences & Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
| |
Collapse
|
22
|
Liddie S, Itzhak Y. Variations in the stimulus salience of cocaine reward influences drug-associated contextual memory. Addict Biol 2016; 21:242-54. [PMID: 25351485 DOI: 10.1111/adb.12191] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Drugs of abuse act as reinforcers because they influence learning and memory processes resulting in long-term memory of drug reward. We have previously shown that mice conditioned by fixed daily dose of cocaine (Fix-C) or daily escalating doses of cocaine (Esc-C) resulted in short- and long-term persistence of drug memory, respectively, suggesting different mechanisms in acquisition of cocaine memory. The present study was undertaken to investigate the differential contribution of N-methyl-D-aspartate receptor (NMDAR) subunits in the formation of Fix-C and Esc-C memory in C57BL/6J mice. Training by Esc-C resulted in marked elevation in hippocampal expression of Grin2b mRNA and NR2B protein levels compared with training by Fix-C. The NR2B-containing NMDAR antagonist ifenprodil had similar attenuating effects on acquisition and reconsolidation of Fix-C and Esc-C memory. However, the NMDAR antagonist MK-801 had differential effects: (1) higher doses of MK-801 were required for post-retrieval disruption of reconsolidation of Esc-C memory than Fix-C memory; and (2) pre-retrieval MK-801 inhibited extinction of Fix-C memory but it had no effect on Esc-C memory. In addition, blockade of NMDAR downstream signaling pathways also showed differential regulation of Fix-C and Esc-C memory. Inhibition of neuronal nitric oxide synthase attenuated acquisition and disrupted reconsolidation of Fix-C but not Esc-C memory. In contrast, the mitogen-activating extracellular kinase inhibitor SL327 attenuated reconsolidation of Esc-C but not Fix-C memory. These results suggest that NMDAR downstream signaling molecules associated with consolidation and reconsolidation of cocaine-associated memory may vary upon changes in the salience of cocaine reward during conditioning.
Collapse
Affiliation(s)
- Shervin Liddie
- Division of Neuroscience; University of Miami Miller School of Medicine; Miami FL USA
| | - Yossef Itzhak
- Department of Psychiatry and Behavioral Sciences; University of Miami Miller School of Medicine; Miami FL USA
- Division of Neuroscience; University of Miami Miller School of Medicine; Miami FL USA
| |
Collapse
|
23
|
Liu JF, Thorn DA, Zhang Y, Li JX. Effects of Trace Amine-associated Receptor 1 Agonists on the Expression, Reconsolidation, and Extinction of Cocaine Reward Memory. Int J Neuropsychopharmacol 2016; 19:pyw009. [PMID: 26822713 PMCID: PMC4966273 DOI: 10.1093/ijnp/pyw009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 01/25/2016] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND As a modulator of dopaminergic system, trace amine-associated receptor 1 has been shown to play a critical role in regulating the rewarding properties of additive drugs. It has been demonstrated that activation of trace amine-associated receptor 1 decreased the abuse-related behaviors of cocaine in rats. However, the role of trace amine-associated receptor 1 in specific stages of cocaine reward memory is still unclear. METHODS Here, using a cocaine-induced conditioned place preference model, we tested the effects of a selective trace amine-associated receptor 1 agonist RO5166017 on the expression, reconsolidation, and extinction of cocaine reward memory. RESULTS We found that RO5166017 inhibited the expression but not retention of cocaine-induced conditioned place preference. RO5166017 had no effect on the reconsolidation of cocaine reward memory. Pretreatment with RO5166017 before extinction hindered the formation of extinction long-term memory. RO5166017 did not affect the movement during the conditioned place preference test, indicating the inhibitory effect of RO5166017 on the expression of cocaine-induced conditioned place preference was not caused by locomotion inhibition. Using a cocaine i.v. self-administration model, we found that the combined trace amine-associated receptor 1 partial agonist RO5263397 with extinction had no effect on the following cue- and drug-induced reinstatement of cocaine-seeking behavior. Repeated administration of the trace amine-associated receptor 1 agonist during extinction showed a continually inhibitory effect on the expression of cocaine reward memory both in cocaine-induced conditioned place preference and cocaine self-administration models. CONCLUSIONS Taken together, these results indicate that activation of trace amine-associated receptor 1 specifically inhibited the expression of cocaine reward memory. The inhibitory effect of trace amine-associated receptor 1 agonists on cocaine reward memory suggests that trace amine-associated receptor 1 agonists could be a promising agent to prevent cocaine relapse.
Collapse
Affiliation(s)
| | | | | | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY (Drs Liu, Thorn, and Li); Research Triangle Institute, Research Triangle Park, NC (Dr Zhang).
| |
Collapse
|
24
|
Vengeliene V, Olevska A, Spanagel R. Long-lasting effect of NMDA receptor antagonist memantine on ethanol-cue association and relapse. J Neurochem 2015; 135:1080-5. [PMID: 26342155 DOI: 10.1111/jnc.13350] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 08/20/2015] [Accepted: 08/28/2015] [Indexed: 12/13/2022]
Abstract
It is well known that the glutamatergic system plays a crucial role in alcohol addiction and especially in relapse-like behaviour. However, results of clinical studies on compounds that influence the activity of the glutamatergic system have been disappointing so far. The aim of our study was to establish treatment conditions under which the N-methyl-d-aspartate receptor (NMDAR) antagonist memantine may produce more reliable treatment effect with respect to alcohol relapse-like behaviour. For this purpose, male Wistar rats were trained to associate several discrete stimuli with ethanol delivery. Thereafter, half of the animals received a brief memory reactivation session followed by two administrations of 20 mg/kg of memantine, while the other half received the same treatment without memory reactivation. Afterwards, a cue-induced ethanol-seeking behaviour test was performed followed by repeated extinction sessions and a reacquisition test. Our data show that administration of memantine reduced responding on the ethanol-associated lever in a cue-induced ethanol-seeking test. This reduction did not depend on whether or not a memory reactivation session was introduced prior to memantine administration. Following extinction, however, reacquisition of ethanol self-administration was only impaired in the group where memantine was given after a short memory reactivation session, showing that this schedule of drug administration produced a long-lasting disruption of the association between the conditioned stimuli and the delivery of ethanol. In conclusion, we show that memantine disrupted the drug-cue association, which consequently interfered with relapse-like behaviour supporting the possibility that memantine is a treatment option for alcoholism. Our data supports the possibility that memantine is a treatment option for alcoholism. However, the effectiveness of this drug seems to lie in its ability to disrupt conditioned behaviours and should be given in conjunction with exposure to conditioned drug stimuli.
Collapse
Affiliation(s)
- Valentina Vengeliene
- Institute of Psychopharmacology, Central Institute of Mental Health, Faculty of Medicine Mannheim, Heidelberg University, Mannheim, Germany
| | - Anastasia Olevska
- Institute of Psychopharmacology, Central Institute of Mental Health, Faculty of Medicine Mannheim, Heidelberg University, Mannheim, Germany
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Faculty of Medicine Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
25
|
Dennis TS, Perrotti LI. Erasing Drug Memories Through the Disruption of Memory Reconsolidation: A Review of Glutamatergic Mechanisms. ACTA ACUST UNITED AC 2015. [DOI: 10.1111/jabr.12031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
26
|
Exton-McGuinness MTJ, Lee JLC. Reduction in Responding for Sucrose and Cocaine Reinforcement by Disruption of Memory Reconsolidation. eNeuro 2015; 2:ENEURO.0009-15.2015. [PMID: 26464973 PMCID: PMC4596086 DOI: 10.1523/eneuro.0009-15.2015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 03/03/2015] [Accepted: 03/04/2015] [Indexed: 11/21/2022] Open
Abstract
Stored memories are dynamic and, when reactivated, can undergo a process of destabilization and reconsolidation to update them with new information. Reconsolidation has been shown for a variety of experimental settings; most recently for well-learned instrumental memories, a class of memory previously thought not to undergo reconsolidation. Here we tested, in rats, whether a weakly-trained lever-pressing memory destabilized following a shift in reinforcement contingency. We show that lever-pressing memory for both sucrose and cocaine reinforcement destabilized under appropriate conditions, and that the reconsolidation of this memory was impaired by systemic administration of the NMDA receptor (NMDAR) antagonist [5R,10S]-[+]-5-methyl-10,1-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine (MK-801). We went on to investigate the potential role of the nucleus accumbens (NAc) in the reconsolidation of sucrose-reinforced instrumental memories, showing that co-infusion of the NMDAR antagonist 2-amino-5-phosphonopentanoic acid (AP-5) and the dopamine-1 receptor (D1R) antagonist 7-chloro-3-methyl-1-phenyl-1,2,4,5-tetrahydro-3-benzazepin-8-ol (SCH23390) into the NAc prior to memory reactivation impaired reconsolidation; however, there was no effect when these drugs were infused alone. Further investigation of this effect suggests the combined infusion disrupted the reconsolidation of pavlovian components of memory, and we hypothesize that coactivation of accumbal D1Rs and NMDARs may contribute to both the destabilization and reconsolidation of appetitive memory. Our work demonstrates that weakly-trained instrumental memories undergo reconsolidation under similar parameters to well-trained ones, and also suggests that receptor coactivation in the NAc may contribute to memory destabilization. Furthermore, it provides an important demonstration of the therapeutic potential of reconsolidation-based treatments that target the instrumental components of memory in maladaptive drug seeking.
Collapse
Affiliation(s)
| | - Jonathan L C Lee
- School of Psychology, University of Birmingham , B15 2TT, United Kingdom
| |
Collapse
|
27
|
Abstract
Many psychiatric disorders are characterized by intrusive, distracting, and disturbing memories that either perpetuate the illness or hinder successful treatment. For example, posttraumatic stress disorder (PTSD) involves such strong reemergence of memories associated with a traumatic event that the individual feels like the event is happening again. Furthermore, drug addiction is characterized by compulsive use and repeated relapse that is often driven by internal memories of drug use and/or by exposure to external stimuli that were associated with drug use. Therefore, identifying pharmacological methods to weaken the strength of maladaptive memories is a major goal of research efforts aimed at finding new treatments for these disorders. The primary mechanism by which memories could be pharmacologically disrupted or altered is through manipulation of memory reconsolidation. Reconsolidation occurs when an established memory is remembered or reactivated, reentering a labile state before again being consolidated into long-term memory storage. Memories are subject to disruption during this labile state. In this chapter we will discuss the preclinical and clinical studies identifying potential pharmacological methods for disrupting the integrity of maladaptive memory to treat mental illness.
Collapse
Affiliation(s)
- Jane R Taylor
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | | |
Collapse
|
28
|
Das RK, Hindocha C, Freeman TP, Lazzarino AI, Curran HV, Kamboj SK. Assessing the translational feasibility of pharmacological drug memory reconsolidation blockade with memantine in quitting smokers. Psychopharmacology (Berl) 2015; 232:3363-74. [PMID: 26093656 PMCID: PMC4537501 DOI: 10.1007/s00213-015-3990-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 06/01/2015] [Indexed: 11/28/2022]
Abstract
RATIONALE Preclinical reconsolidation research offers the first realistic opportunity to pharmacologically weaken the maladaptive memory structures that support relapse in drug addicts. N-methyl D-aspartate receptor (NMDAR) antagonism is a highly effective means of blocking drug memory reconsolidation. However, no research using this approach exists in human addicts. OBJECTIVES The objective of this study was to assess the potential and clinical outcomes of blocking the reconsolidation of cue-smoking memories with memantine in quitting smokers. METHODS Fifty-nine dependent and motivated to quit smokers were randomised to one of three groups receiving the following: (1) memantine with or (2) without reactivation of associative cue-smoking memories or (3) reactivation with placebo on their target quit day in a double-blind manner. Participants aimed to abstain from smoking for as long as possible. Levels of smoking and FTND score were assessed prior to intervention and up to a year later. Primary outcome was latency to relapse. Subjective craving measures and attentional bias to smoking cues were assessed in-lab. RESULTS All study groups successfully reduced their smoking up to 3 months. Memantine in combination with smoking memory reactivation did not affect any measure of smoking outcome, reactivity or attention capture to smoking cues. CONCLUSIONS Brief exposure to smoking cues with memantine did not appear to weaken these memory traces. These findings could be due to insufficient reconsolidation blockade by memantine or failure of exposure to smoking stimuli to destabilise smoking memories. Research assessing the treatment potential of reconsolidation blockade in human addicts should focus on identification of tolerable drugs that reliably block reward memory reconsolidation and retrieval procedures that reliably destabilise strongly trained memories.
Collapse
Affiliation(s)
- Ravi K Das
- Clinical Psychopharmacology Unit, University College London, 1-19 Torrington Place, London, WC1E 6BT, UK,
| | | | | | | | | | | |
Collapse
|
29
|
Retrieval-induced NMDA receptor-dependent Arc expression in two models of cocaine-cue memory. Neurobiol Learn Mem 2014; 116:79-89. [PMID: 25225165 DOI: 10.1016/j.nlm.2014.09.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 09/02/2014] [Accepted: 09/04/2014] [Indexed: 01/15/2023]
Abstract
The association of environmental cues with drugs of abuse results in persistent drug-cue memories. These memories contribute significantly to relapse among addicts. While conditioned place preference (CPP) is a well-established paradigm frequently used to examine the modulation of drug-cue memories, very few studies have used the non-preference-based model conditioned activity (CA) for this purpose. Here, we used both experimental approaches to investigate the neural substrates of cocaine-cue memories. First, we directly compared, in a consistent setting, the involvement of cortical and subcortical brain regions in cocaine-cue memory retrieval by quantifying activity-regulated cytoskeletal-associated (Arc) protein expression in both the CPP and CA models. Second, because NMDA receptor activation is required for Arc expression, we investigated the NMDA receptor dependency of memory persistence using the CA model. In both the CPP and CA models, drug-paired animals showed significant increases in Arc immunoreactivity in regions of the frontal cortex and amygdala compared to unpaired controls. Additionally, administration of a NMDA receptor antagonist (MK-801 or memantine) immediately after cocaine-CA memory reactivation impaired the subsequent conditioned locomotion associated with the cocaine-paired environment. The enhanced Arc expression evident in a subset of corticolimbic regions after retrieval of a cocaine-context memory, observed in both the CPP and CA paradigms, likely signifies that these regions: (i) are activated during retrieval of these memories irrespective of preference-based decisions, and (ii) undergo neuroplasticity in order to update information about cues previously associated with cocaine. This study also establishes the involvement of NMDA receptors in maintaining memories established using the CA model, a characteristic previously demonstrated using CPP. Overall, these results demonstrate the utility of the CA model for studies of cocaine-context memory and suggest the involvement of an NMDA receptor-dependent Arc induction pathway in drug-cue memory interference.
Collapse
|
30
|
Shi X, Miller JS, Harper LJ, Poole RL, Gould TJ, Unterwald EM. Reactivation of cocaine reward memory engages the Akt/GSK3/mTOR signaling pathway and can be disrupted by GSK3 inhibition. Psychopharmacology (Berl) 2014; 231:3109-18. [PMID: 24595501 PMCID: PMC4110417 DOI: 10.1007/s00213-014-3491-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 02/04/2014] [Indexed: 11/14/2022]
Abstract
RATIONAL Memories return to a labile state following their retrieval and must undergo a process of reconsolidation to be maintained. Thus, disruption of cocaine reward memories by interference with reconsolidation may be therapeutically beneficial in the treatment of cocaine addiction. OBJECTIVE The objectives were to elucidate the signaling pathway involved in reconsolidation of cocaine reward memory and to test whether targeting this pathway could disrupt cocaine-associated contextual memory. METHODS Using a mouse model of conditioned place preference, regulation of the activity of glycogen synthase kinase-3 (GSK3), mammalian target of Rapamycin complex 1 (mTORC1), P70S6K, β-catenin, and the upstream signaling molecule Akt, was studied in cortico-limbic-striatal circuitry after re-exposure to an environment previously paired with cocaine. RESULT Levels of phosporylated Akt-Thr308, GSK3α-Ser21, GSK3β-Ser9, mTORC1, and P70S6K were reduced in the nucleus accumbens and hippocampus 10 min after the reactivation of cocaine cue memories. Levels of pAkt and pGSK3 were also reduced in the prefrontal cortex. Since reduced phosphorylation of GSK3 indicates heightened enzyme activity, the effect of a selective GSK3 inhibitor, SB216763, on reconsolidation was tested. Administration of SB216763 immediately after exposure to an environment previously paired with cocaine abrogated a previously established place preference, suggesting that GSK3 inhibition interfered with reconsolidation of cocaine-associated reward memories. CONCLUSIONS These findings suggest that the Akt/GSK3/mTORC1 signaling pathway in the nucleus accumbens, hippocampus, and/or prefrontal cortex is critically involved in the reconsolidation of cocaine contextual reward memory. Inhibition of GSK3 activity during memory retrieval can erase an established cocaine place preference.
Collapse
Affiliation(s)
- Xiangdang Shi
- Department of Pharmacology and the Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA 19140 USA
| | - Jonathan S. Miller
- Department of Pharmacology and the Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA 19140 USA
| | - Lauren J. Harper
- Department of Pharmacology and the Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA 19140 USA
| | - Rachel L. Poole
- Department of Psychology, Temple University, Philadelphia, PA USA
| | - Thomas J. Gould
- Department of Psychology, Temple University, Philadelphia, PA USA
| | - Ellen M. Unterwald
- Department of Pharmacology and the Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA 19140 USA
| |
Collapse
|
31
|
Reichelt AC, Lee JLC. Memory reconsolidation in aversive and appetitive settings. Front Behav Neurosci 2013; 7:118. [PMID: 24058336 PMCID: PMC3766793 DOI: 10.3389/fnbeh.2013.00118] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 08/20/2013] [Indexed: 11/16/2022] Open
Abstract
Memory reconsolidation has been observed across species and in a number of behavioral paradigms. The majority of memory reconsolidation studies have been carried out in Pavlovian fear conditioning and other aversive memory settings, with potential implications for the treatment of post-traumatic stress disorder. However, there is a growing literature on memory reconsolidation in appetitive reward-related memory paradigms, including translational models of drug addiction. While there appears to be substantial similarity in the basic phenomenon and underlying mechanisms of memory reconsolidation across unconditioned stimulus valence, there are also notable discrepancies. These arise both when comparing aversive to appetitive paradigms and also across different paradigms within the same valence of memory. We review the demonstration of memory reconsolidation across different aversive and appetitive memory paradigms, the commonalities and differences in underlying mechanisms and the conditions under which each memory undergoes reconsolidation. We focus particularly on whether principles derived from the aversive literature are applicable to appetitive settings, and also whether the expanding literature in appetitive paradigms is informative for fear memory reconsolidation.
Collapse
Affiliation(s)
- Amy C Reichelt
- School of Psychology, University of Birmingham Birmingham, UK
| | | |
Collapse
|
32
|
Tronson NC, Taylor JR. Addiction: a drug-induced disorder of memory reconsolidation. Curr Opin Neurobiol 2013; 23:573-80. [PMID: 23415831 PMCID: PMC3677957 DOI: 10.1016/j.conb.2013.01.022] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Revised: 01/15/2013] [Accepted: 01/16/2013] [Indexed: 12/15/2022]
Abstract
Persistent maladaptive memories that maintain drug seeking and are resistant to extinction are a hallmark of addiction. As such, disruption of memory reconsolidation after retrieval has received attention for its therapeutic potential. Unrestrained reconsolidation may have the opposite effect, leading to reiterative and cumulative strengthening of memory over long periods of time. Here we review the molecular mechanisms underlying reconsolidation of appetitive and drug-rewarded memories, and discuss how these findings contribute to our understanding of the nature of this process. Finally, we suggest that drug-induced alterations to signal transduction might lead to dysregulation of reconsolidation, causing enhancements of drug-related memory after retrieval, and significantly contribute to the compulsive drug seeking that is a core component of addiction.
Collapse
Affiliation(s)
- Natalie C Tronson
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109,
| | - Jane R. Taylor
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06508
- Department of Psychology, Yale University, New Haven, CT 06508,
| |
Collapse
|
33
|
Abstract
Cocaine-induced neuroplasticity mediated by histone acetylating and deacetylating enzymes may contribute to addiction-like behaviors. For example, overexpression of histone deacetylases (HDACs) 4 or 5 in the nucleus accumbens suppresses cocaine-induced conditioned place preference (CPP) acquisition in mice. HDAC4 and HDAC5 are known to interact with HDAC3, but the role of HDAC3 in cocaine-induced behaviors has never been examined. In this study, we address the hypothesis that HDAC3 is a negative regulator of cocaine-context-associated memory formation in mice. We examined the role of HDAC3 during the conditioning phase of CPP, when the mouse has the opportunity to form an associative memory between the cocaine-paired context and the subjective effects of cocaine. To address this hypothesis, Hdac3(flox/flox) and Hdac3(+/+) mice (generated from a C57BL/6 background) were infused into the nucleus accumbens with adeno-associated virus expressing Cre recombinase to create focal, homozygous Hdac3 deletions. Hdac3(flox/flox) mice exhibit significantly enhanced CPP acquisition, which is correlated with increased gene expression during the consolidation phase of acquisition. Increased gene expression of c-Fos and Nr4a2 is correlated with decreased HDAC3 occupancy and increased histone H4 lysine 8 acetylation at their promoters. The results from this study demonstrate that HDAC3 negatively regulates cocaine-induced CPP acquisition.
Collapse
|