1
|
Pohořalá V, Kuchař M, Spanagel R, Bernardi RE. Psilocybin administered following extinction sessions does not affect subsequent cocaine cue reinstatement in male and female rats and mice. Neuroscience 2024; 559:156-165. [PMID: 39236802 DOI: 10.1016/j.neuroscience.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/19/2024] [Accepted: 09/01/2024] [Indexed: 09/07/2024]
Abstract
There are currently no pharmacological treatments for cocaine use disorder. Recently there has been a great deal of interest in the potential of psychedelic drugs such as psilocybin to treat psychiatric disorders. Human studies have indicated that a single administration of psilocybin can have long-lasting effects. Few preclinical studies have examined a role for psilocybin in addiction models. The goal of the current study was to determine whether psilocybin would enhance extinction following cocaine self-administration in male and female mice and rats and thus result in an attenuation of cue-induced drug-seeking. In experiments in mice, 16 female and 19 male mice underwent 8d of cocaine self-administration (0.5 mg/kg/infusion) and extinction training. Immediately following extinction trials, mice were injected with vehicle or 1.0 mg/kg psilocybin. Following the conclusion of extinction training, mice were tested for cue-induced reinstatement. In experiments in rats, 24 female and 23 male rats underwent 15d of cocaine self-administration (0.8 mg/kg/infusion) and extinction training. Immediately following extinction trials, rats were injected with vehicle, 1.0 mg/kg psilocybin, or 2.5 mg/kg psilocybin. Following the conclusion of extinction training, rats were tested for cue-induced reinstatement. Psilocybin administered following extinction trials had no effect, as both female and male mice and rats demonstrated significant cue-induced reinstatement. These data suggest that psilocybin is ineffective at altering cocaine-seeking behavior in the paradigm and doses used in the current study. It remains to be seen whether treatment with psilocybin under different conditions may be useful in the long-standing goal of finding pharmacotherapies to treat CUD.
Collapse
Affiliation(s)
- Veronika Pohořalá
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Martin Kuchař
- Forensic Laboratory of Biologically Active Substances, Department of Chemistry of Natural Compounds, University of Chemistry and Technology Prague, Prague, Czech Republic; Psychedelics Research Centre, National Institute of Mental Health, Klecany, Czech Republic
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Rick E Bernardi
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
2
|
Withey SL, Deshpande HU, Cao L, Bergman J, Kohut SJ. Effects of chronic naltrexone treatment on relapse-related behavior and neural responses to fentanyl in awake nonhuman primates. Psychopharmacology (Berl) 2024; 241:2289-2302. [PMID: 39122918 DOI: 10.1007/s00213-024-06633-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 06/03/2024] [Indexed: 08/12/2024]
Abstract
Naltrexone, an opioid antagonist that blocks the reinforcing properties of opioid agonists, is often prescribed to preclude relapse to opioid use disorder (OUD) following detoxification. However, few laboratory studies have directly investigated the ability of naltrexone to alter relapse-inducing effects of opioid agonists, including their priming strength in reinstatement studies and their impact in brain regions known to be involved in drug-induced reinforcement in MRI studies. Here we directly address this issue by investigating the effects of continuous exposure to naltrexone on 1) fentanyl-induced reinstatement of drug-seeking behavior, 2) fentanyl-induced patterns of blood oxygenation level dependent (BOLD) activation in the nucleus accumbens (NAcc), and 3) fentanyl-induced changes in NAcc functional connectivity (FC) in awake non-human primates that are engaged in ongoing opioid self-administration studies. We found that naltrexone antagonizes the priming strength of fentanyl as shown by a rightward shift in its reinstatement dose-effect curve and that naltrexone surmountably antagonizes the BOLD response induced by fentanyl. However, while naltrexone also countered fentanyl's effects on NAcc FC, the effects were not surmounted by a higher dose of fentanyl. Together, these data suggest that, in contrast to naltrexone's modulation of fentanyl's effects on behavior and BOLD responses, their interactive effects on FC between multiple brain regions do not reflect their receptor-mediated activity. Additionally, we demonstrated opposing effects in the absence and presence of naltrexone on NAcc FC at baseline (i.e., in the absence of any fentanyl prime) suggesting that naltrexone alters FC at baseline, even though naltrexone appears behaviorally silent in the absence of an agonist prime. Together these data provide additional insight into ways in which naltrexone interacts with opioid agonists, both behaviorally and in the brain. Further understanding the effects of opioid agonists on patterns of FC could help elucidate our understanding of the neural processes that contribute to the initiation of and relapse to opioid-seeking behavior in OUD.
Collapse
Affiliation(s)
- Sarah L Withey
- Behavioral Biology Program, McLean Hospital/Harvard Medical School, 115 Mill St., Belmont, MA, 02478, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Harshawardhan U Deshpande
- Behavioral Biology Program, McLean Hospital/Harvard Medical School, 115 Mill St., Belmont, MA, 02478, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Behavioral Neuroimaging Laboratory, McLean Hospital, Belmont, MA, USA
| | - Lei Cao
- Behavioral Biology Program, McLean Hospital/Harvard Medical School, 115 Mill St., Belmont, MA, 02478, USA
- Behavioral Neuroimaging Laboratory, McLean Hospital, Belmont, MA, USA
| | - Jack Bergman
- Behavioral Biology Program, McLean Hospital/Harvard Medical School, 115 Mill St., Belmont, MA, 02478, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Stephen J Kohut
- Behavioral Biology Program, McLean Hospital/Harvard Medical School, 115 Mill St., Belmont, MA, 02478, USA.
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
- Behavioral Neuroimaging Laboratory, McLean Hospital, Belmont, MA, USA.
- McLean Imaging Center, McLean Hospital, Belmont, MA, USA.
| |
Collapse
|
3
|
Bakhtazad A, Kabbaj M, Garmabi B, Joghataei MT. The role of CART peptide in learning and memory: A potential therapeutic target in memory-related disorders. Peptides 2024; 181:171298. [PMID: 39317295 DOI: 10.1016/j.peptides.2024.171298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 08/19/2024] [Accepted: 09/20/2024] [Indexed: 09/26/2024]
Abstract
Cocaine and amphetamine-regulated transcript (CART) mRNA and peptide are vastly expressed in both cortical and subcortical brain areas and are involved in critical cognitive functions. CART peptide (CARTp), described in reward-related brain structures, regulates drug-induced learning and memory, and its role appears specific to psychostimulants. However, many other drugs of abuse, such as alcohol, opiates, nicotine, and caffeine, have been shown to alter the expression levels of CART mRNA and peptides in brain structures directly or indirectly associated with learning and memory processes. However, the number of studies demonstrating the contribution of CARTp in learning and memory is still minimal. Notably, the exact cellular and molecular mechanisms underlying CARTp effects are still unknown. The discoveries that CARTp effects are mediated through a putative G-protein coupled receptor and activation of cellular signaling cascades via NMDA receptor-coupled ERK have enhanced our knowledge about the action of this neuropeptide and allowed us to comprehend better CARTp exact cellular/molecular mechanisms that could mediate drug-induced changes in learning and memory functions. Unfortunately, these efforts have been impeded by the lack of suitable and specific CARTp receptor antagonists. In this review, following a short introduction about CARTp, we report on current knowledge about CART's roles in learning and memory processes and its recently described role in memory-related neurological disorders. We will also discuss the importance of further investigating how CARTp interacts with its receptor(s) and other neurotransmitter systems to influence learning and memory functions. This topic is sure to intrigue and motivate further exploration in the field of neuroscience.
Collapse
Affiliation(s)
- Atefeh Bakhtazad
- Cellular and Molecular Research Center, Deputy of Research and Technology, Iran University of Medical Sciences, Tehran, Iran; Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Mohamed Kabbaj
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306-1270, United States; Program of Neuroscience, Florida State University, Tallahassee, FL 32306-1270, United States
| | - Behzad Garmabi
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mohammad Taghi Joghataei
- Cellular and Molecular Research Center, Deputy of Research and Technology, Iran University of Medical Sciences, Tehran, Iran; Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Song SH, Kim S, Jang WJ, Ryu IS, Jeong CH, Lee S. Exploring the progression of drug dependence in a methamphetamine self-administration rat model through targeted and non-targeted metabolomics analyses. Sci Rep 2024; 14:22543. [PMID: 39343795 PMCID: PMC11439939 DOI: 10.1038/s41598-024-73247-5] [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: 06/14/2024] [Accepted: 09/16/2024] [Indexed: 10/01/2024] Open
Abstract
Persistent neurochemical and biological disturbances resulting from repeated cycles of drug reward, withdrawal, and relapse contribute to drug dependence. Methamphetamine (MA) is a psychostimulant with substantial abuse potential and neurotoxic effects, primarily affecting monoamine neurotransmitter systems in the brain. In this study, we aimed to explore the progression of drug dependence in rat models of MA self-administration, extinction, and reinstatement through targeted and non-targeted metabolomics analyses. Metabolic profiles were examined in rat plasma during the following phases: after 16 days of MA self-administration (Group M); after 16 days of self-administration followed by 14 days of extinction (Group MS); and after self-administration and extinction followed by a reinstatement injection of MA (Group MSM). Each group of MA self-administration, extinction, and reinstatement induces distinct changes in the metabolic pathways, particularly those related to the TCA cycle, arginine and proline metabolism, and arginine biosynthesis. Additionally, the downregulation of glycerophospholipids and sphingomyelins in Group MSM suggests their potential role in MA reinstatement. These alterations may signify the progressive deterioration of these metabolic pathways, possibly contributing to drug dependence following repeated cycles of drug reward, withdrawal, and relapse. These results provide valuable insights into the metabolic changes associated with MA use at various stages, potentially facilitating the discovery of early diagnostic biomarkers and therapeutic targets for MA use disorders.
Collapse
Affiliation(s)
- Sang-Hoon Song
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Dalseo-gu, Daegu, 42601, Republic of Korea
| | - Suji Kim
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Dalseo-gu, Daegu, 42601, Republic of Korea
| | - Won-Jun Jang
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Dalseo-gu, Daegu, 42601, Republic of Korea
| | - In Soo Ryu
- Biorchestra Co., Ltd, Techno4-ro 17, Daejeon, 34013, Republic of Korea
| | - Chul-Ho Jeong
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Dalseo-gu, Daegu, 42601, Republic of Korea.
| | - Sooyeun Lee
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Dalseo-gu, Daegu, 42601, Republic of Korea.
| |
Collapse
|
5
|
Barry SM, Huebschman J, Devries DM, McCue LM, Tsvetkov E, Anderson EM, Siemsen BM, Berto S, Scofield MD, Taniguchi M, Penrod RD, Cowan CW. Histone deacetylase 5 in prelimbic prefrontal cortex limits context-associated cocaine seeking. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.21.614125. [PMID: 39345428 PMCID: PMC11429996 DOI: 10.1101/2024.09.21.614125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Background Repeated cocaine use produces neuroadaptations that support drug craving and relapse in substance use disorders (SUDs). Powerful associations formed with drug-use environments can promote a return to active drug use in SUD patients, but the molecular mechanisms that control the formation of these prepotent drug-context associations remain unclear. Methods In the rat intravenous cocaine self-administration (SA) model, we examined the role and regulation of histone deacetylase 5 (HDAC5) in the prelimbic (PrL) and infralimbic (IL) cortices in context-associated drug seeking. To this end, we employed viral molecular tools, chemogenetics, RNA-sequencing, electrophysiology, and immunohistochemistry. Results In the PrL, reduction of endogenous HDAC5 augmented context-associated, but not cue-or drug prime-reinstated cocaine seeking, whereas overexpression of HDAC5 in PrL, but not IL, reduced context-associated cocaine seeking, but it had no effects on sucrose seeking. In contrast, PrL HDAC5 overexpression following acquisition of cocaine SA had no effects on future cocaine seeking. We found that HDAC5 and cocaine SA altered the expression of numerous PrL genes, including many synapse-associated genes. HDAC5 significantly increased inhibitory synaptic transmission onto PrL deep-layer pyramidal neurons, and it reduced the induction of FOS-positive neurons in the cocaine SA environment. Conclusions Our findings reveal an essential and selective role for PrL HDAC5 to limit associations formed in cocaine, but not sucrose, SA environments, and that it alters the PrL excitatory/inhibitory balance, possibly through epigenetic regulation of synaptic genes. These results further position HDAC5 as a key factor regulating reward-circuit neuroadaptations that underlie common relapse triggers in SUD.
Collapse
|
6
|
Xu Y, Lin Y, Yu M, Zhou K. The nucleus accumbens in reward and aversion processing: insights and implications. Front Behav Neurosci 2024; 18:1420028. [PMID: 39184934 PMCID: PMC11341389 DOI: 10.3389/fnbeh.2024.1420028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/26/2024] [Indexed: 08/27/2024] Open
Abstract
The nucleus accumbens (NAc), a central component of the brain's reward circuitry, has been implicated in a wide range of behaviors and emotional states. Emerging evidence, primarily drawing from recent rodent studies, suggests that the function of the NAc in reward and aversion processing is multifaceted. Prolonged stress or drug use induces maladaptive neuronal function in the NAc circuitry, which results in pathological conditions. This review aims to provide comprehensive and up-to-date insights on the role of the NAc in motivated behavior regulation and highlights areas that demand further in-depth analysis. It synthesizes the latest findings on how distinct NAc neuronal populations and pathways contribute to the processing of opposite valences. The review examines how a range of neuromodulators, especially monoamines, influence the NAc's control over various motivational states. Furthermore, it delves into the complex underlying mechanisms of psychiatric disorders such as addiction and depression and evaluates prospective interventions to restore NAc functionality.
Collapse
Affiliation(s)
| | | | | | - Kuikui Zhou
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| |
Collapse
|
7
|
Lenoir M, Engeln M, Navailles S, Girardeau P, Ahmed SH. A large-scale c-Fos brain mapping study on extinction of cocaine-primed reinstatement. Neuropsychopharmacology 2024; 49:1459-1467. [PMID: 38664549 PMCID: PMC11251268 DOI: 10.1038/s41386-024-01867-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/29/2024] [Accepted: 04/11/2024] [Indexed: 07/17/2024]
Abstract
Individuals with cocaine addiction can experience many craving episodes and subsequent relapses, which represents the main obstacle to recovery. Craving is often favored when abstinent individuals ingest a small dose of cocaine, encounter cues associated with drug use or are exposed to stressors. Using a cocaine-primed reinstatement model in rat, we recently showed that cocaine-conditioned interoceptive cues can be extinguished with repeated cocaine priming in the absence of drug reinforcement, a phenomenon we called extinction of cocaine priming. Here, we applied a large-scale c-Fos brain mapping approach following extinction of cocaine priming in male rats to identify brain regions implicated in processing the conditioned interoceptive stimuli of cocaine priming. We found that cocaine-primed reinstatement is associated with increased c-Fos expression in key brain regions (e.g., dorsal and ventral striatum, several prefrontal areas and insular cortex), while its extinction mostly disengages them. Moreover, while reinstatement behavior was correlated with insular and accumbal activation, extinction of cocaine priming implicated parts of the ventral pallidum, the mediodorsal thalamus and the median raphe. These brain patterns of activation and inhibition suggest that after repeated priming, interoceptive signals lose their conditioned discriminative properties and that action-outcome associations systems are mobilized in search for new contingencies, a brain state that may predispose to rapid relapse.
Collapse
Affiliation(s)
- Magalie Lenoir
- Univ. Bordeaux, CNRS, INCIA, UMR 5287, F-33000, Bordeaux, France.
| | - Michel Engeln
- Univ. Bordeaux, CNRS, INCIA, UMR 5287, F-33000, Bordeaux, France.
| | | | - Paul Girardeau
- Univ. Bordeaux, UFR des Sciences Odontologiques, Bordeaux, France
| | - Serge H Ahmed
- Univ. Bordeaux, CNRS, INCIA, UMR 5287, F-33000, Bordeaux, France
| |
Collapse
|
8
|
Carter JS, Costa CC, Lewandowski SI, Nelson KH, Goldsmith ST, Scofield MD, Reichel CM. Estrogen receptor beta signaling enhances extinction memory recall for heroin-conditioned cues in a sex- and region-specific manner. Transl Psychiatry 2024; 14:283. [PMID: 38997258 PMCID: PMC11245532 DOI: 10.1038/s41398-024-03001-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/14/2024] Open
Abstract
Return to use, or relapse, is a major challenge in the treatment of opioid use disorder (OUD). Relapse can be precipitated by several factors, including exposure to drug-conditioned cues. Identifying successful treatments to mitigate cue-induced relapse has been challenging, perhaps due to extinction memory recall (EMR) deficits. Previously, inhibition of estradiol (E2) signaling in the basolateral amygdala (BLA) impaired heroin-cue EMR. This effect was recapitulated by antagonism of BLA estrogen receptors (ER) in a sex-specific manner such that blocking ERα in males, but ERβ in females, impaired EMR. However, it is unclear whether increased E2 signaling, in the BLA or systemically, enhances heroin-cue EMR. We hypothesized that ERβ agonism would enhance heroin-cue EMR in a sex- and region-specific manner. To determine the capacity of E2 signaling to improve EMR, we pharmacologically manipulated ERβ across several translationally designed experiments. First, male and female rats acquired heroin or sucrose self-administration. Next, during a cued extinction session, we administered diarylpropionitrile (DPN, an ERβ agonist) and tested anxiety-like behavior on an open field. Subsequently, we assessed EMR in a cue-induced reinstatement test and, finally, measured ERβ expression in several brain regions. Across all experiments, females took more heroin and sucrose than males and had greater responses during heroin-cued extinction. Administration of DPN in the BLA enhanced EMR in females only, driven by ERβ's impacts on memory consolidation. Interestingly, however, systemic DPN administration improved EMR for heroin cues in both sexes across several different tests, but did not impact sucrose-cue EMR. Immunohistochemical analysis of ERβ expression across several different brain regions showed that females only had greater expression of ERβ in the basal nucleus of the BLA. Here, in several preclinical experiments, we demonstrated that ERβ agonism enhances heroin-cue EMR and has potential utility in combatting cue-induced relapse.
Collapse
Affiliation(s)
- Jordan S Carter
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA.
| | - Caitlyn C Costa
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Stacia I Lewandowski
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Katharine H Nelson
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Sarah T Goldsmith
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Michael D Scofield
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Carmela M Reichel
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
9
|
Cristina Bianchi P, Palombo P, Antonagi Engi S, Eduardo Carneiro de Oliveira P, Emily Boaventura Tavares G, Anjos-Santos A, Suemi Yokoyama T, da Silva Planeta C, Cardoso Cruz F, Molini Leão R. Involvement of Pre-limbic Cortex-Nucleus accumbens projections in Context-Induced alcohol seeking. Brain Res 2024; 1841:149086. [PMID: 38876319 DOI: 10.1016/j.brainres.2024.149086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 06/16/2024]
Abstract
Alcohol use disorder (AUD) remains a critical public health issue worldwide, characterized by high relapse rates often triggered by contextual cues. This research investigates the neural mechanisms behind context-induced reinstatement of alcohol-seeking behavior, focusing on the nucleus accumbens and its interactions with the prelimbic cortex, employing Male Long-Evans rats in an ABA renewal model. In our experimental setup, rats were trained to self-administer 10 % ethanol in Context A, followed by extinction of lever pressing in the presence of discrete cues in Context B. The context-induced reinstatement of ethanol-seeking was then assessed by re-exposing rats to Context A or B under extinction conditions, aiming to simulate the environmental cues' influence on relapse behaviors. Three experiments were conducted: Experiment 1 utilized Fos-immunohistochemistry to examine neuronal activation in the nucleus accumbens; Experiment 2 applied the baclofen + muscimol inactivation technique to probe the functional importance of the nucleus accumbens core; Experiment 3 used Fos-immunofluorescence along with Retrobeads injection to investigate activation of neurons projecting from the prelimbic cortex to the nucleus accumbens core. Our findings revealed significant increases in Fos-immunoreactive nuclei within the nucleus accumbens core and shell during the reinstatement phase in Context A, underscoring the environment's potent effect on ethanol-seeking behavior. Additionally, inactivation of the nucleus accumbens core markedly reduced reinstatement, and there was a notable activation of neurons from the prelimbic cortex to the nucleus accumbens core in the ethanol-associated context. These results highlight the critical role of the nucleus accumbens core and its corticostriatal projections in the neural circuitry underlying context-driven ethanol seeking.
Collapse
Affiliation(s)
- Paula Cristina Bianchi
- Laboratory of Behavioral Neuroscience, Paulista Medicine School, Universidade Federal de São Paulo-UNIFESP, São Paulo, SP, Brazil
| | - Paola Palombo
- Laboratory of Behavioral Neuroscience, Paulista Medicine School, Universidade Federal de São Paulo-UNIFESP, São Paulo, SP, Brazil
| | - Sheila Antonagi Engi
- Laboratory of Behavioral Neuroscience, Paulista Medicine School, Universidade Federal de São Paulo-UNIFESP, São Paulo, SP, Brazil
| | | | | | - Alexia Anjos-Santos
- Laboratory of Behavioral Neuroscience, Paulista Medicine School, Universidade Federal de São Paulo-UNIFESP, São Paulo, SP, Brazil
| | - Thais Suemi Yokoyama
- Laboratory of Behavioral Neuroscience, Paulista Medicine School, Universidade Federal de São Paulo-UNIFESP, São Paulo, SP, Brazil
| | - Cleopatra da Silva Planeta
- Laboratory of Neuropsychopharmacology, School of Pharmaceutical Sciences, São Paulo State University, UNESP, Araraquara, SP, Brazil
| | - Fabio Cardoso Cruz
- Laboratory of Behavioral Neuroscience, Paulista Medicine School, Universidade Federal de São Paulo-UNIFESP, São Paulo, SP, Brazil
| | - Rodrigo Molini Leão
- Laboratory of Pharmacology, Biomedical Sciences Institute, Department of Pharmacology, Federal University of Uberlândia, Uberlândia, MG, Brazil; Graduate Program in Genetics and Biochemistry, Institute of Biotechnology, Federal University of Uberlândia/MG, Brazil.
| |
Collapse
|
10
|
Negishi K, Fredriksson I, Bossert JM, Zangen A, Shaham Y. Relapse after electric barrier-induced voluntary abstinence: A review. Curr Opin Neurobiol 2024; 86:102856. [PMID: 38508102 PMCID: PMC11162942 DOI: 10.1016/j.conb.2024.102856] [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: 01/02/2024] [Revised: 02/23/2024] [Accepted: 02/25/2024] [Indexed: 03/22/2024]
Abstract
Relapse to drug use during abstinence is a defining feature of addiction. To date, however, results from studies using rat relapse/reinstatement models have yet to result in FDA-approved medications for relapse prevention. To address this translational gap, we and others have developed rat models of relapse after voluntary abstinence from drug self-administration. One of these models is the electric barrier conflict model. Here, we introduce the model, and then review studies on behavioral and neuropharmacological mechanisms of cue-induced relapse and incubation of drug seeking (time-dependent increase in drug seeking during abstinence) after electric barrier-induced abstinence. We also briefly discuss future directions and potential clinical implications. One major conclusion of our review is that the brain mechanisms controlling drug relapse after electrical barrier-induced voluntary abstinence are likely distinct from those controlling relapse after homecage forced abstinence.
Collapse
Affiliation(s)
| | - Ida Fredriksson
- Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
| | | | - Abraham Zangen
- Department of Life Science and the Zelman Neuroscience Center, Ben-Gurion University, Beer Sheba, Israel
| | - Yavin Shaham
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, MD, USA.
| |
Collapse
|
11
|
Nevado B, Nelson JB. Perceived stress and renewal: The effects of long-term stress on the renewal effect. Neurobiol Learn Mem 2024; 211:107927. [PMID: 38582295 DOI: 10.1016/j.nlm.2024.107927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 03/19/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024]
Abstract
Two online experiments evaluated the relationship between long-term stress, as measured with the Perceived Stress Scale-10, and the Renewal Effect. In the first experiment renewal was assessed with a behavioral suppression task in a science-fiction based video game. Participants learned to suppress mouse clicking during a signal for an upcoming attack to avoid losing points. The signal was first paired with an attack in Context A and extinguished in Context B and tested back in Context A. The contexts were different space galaxies where the gameplay took place. Experiment 2 used a food/illness predictive-learning paradigm. Two food items were paired with stomachache in one restaurant (A) and extinguished in Context B prior to testing in both contexts without feedback. Positive correlations were obtained between renewal and stress in each experiment. Unlike acute stress (Drexler et al., 2017), long term stress was associated with greater renewal. The effects of stress, both chronic and punctual, on renewal are discussed.
Collapse
Affiliation(s)
- Borja Nevado
- University of the Basque Country (UPV/EHU), Spain.
| | | |
Collapse
|
12
|
Markovic T, Higginbotham J, Ruyle B, Massaly N, Yoon HJ, Kuo CC, Kim JR, Yi J, Garcia JJ, Sze E, Abt J, Teich RH, Dearman JJ, McCall JG, Morón JA. A locus coeruleus to dorsal hippocampus pathway mediates cue-induced reinstatement of opioid self-administration in male and female rats. Neuropsychopharmacology 2024; 49:915-923. [PMID: 38374364 PMCID: PMC11039689 DOI: 10.1038/s41386-024-01828-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 02/21/2024]
Abstract
Opioid use disorder is a chronic relapsing disorder encompassing misuse, dependence, and addiction to opioid drugs. Long term maintenance of associations between the reinforcing effects of the drug and the cues associated with its intake are a leading cause of relapse. Indeed, exposure to the salient drug-associated cues can lead to drug cravings and drug seeking behavior. The dorsal hippocampus (dHPC) and locus coeruleus (LC) have emerged as important structures for linking the subjective rewarding effects of opioids with environmental cues. However, their role in cue-induced reinstatement of opioid use remains to be further elucidated. In this study, we showed that chemogenetic inhibition of excitatory dHPC neurons during re-exposure to drug-associated cues significantly attenuates cue-induced reinstatement of morphine-seeking behavior. In addition, the same manipulation reduced reinstatement of sucrose-seeking behavior but failed to alter memory recall in the object location task. Finally, intact activity of tyrosine hydroxylase (TH) LC-dHPCTh afferents is necessary to drive cue induced reinstatement of morphine-seeking as inhibition of this pathway blunts cue-induced drug-seeking behavior. Altogether, these studies show an important role of the dHPC and LC-dHPCTh pathway in mediating cue-induced reinstatement of opioid seeking.
Collapse
Affiliation(s)
- Tamara Markovic
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Jessica Higginbotham
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Brian Ruyle
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Nicolas Massaly
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Hye Jean Yoon
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Chao-Cheng Kuo
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, St. Louis, MO, USA
- Center for Clinical Pharmacology, St. Louis College of Pharmacy and Washington University in St. Louis, St. Louis, MO, USA
| | - Jenny R Kim
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, St. Louis, MO, USA
- Center for Clinical Pharmacology, St. Louis College of Pharmacy and Washington University in St. Louis, St. Louis, MO, USA
| | - Jiwon Yi
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Jeniffer J Garcia
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Eric Sze
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Julian Abt
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Rachel H Teich
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Joanna J Dearman
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Jordan G McCall
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, St. Louis, MO, USA
- Center for Clinical Pharmacology, St. Louis College of Pharmacy and Washington University in St. Louis, St. Louis, MO, USA
| | - Jose A Morón
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA.
- Pain Center, Washington University in St Louis, St. Louis, MO, USA.
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA.
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA.
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
13
|
Badshah I, Qazi NG, Anwar M, Shaukat B, Khan MI, Murtaza B. Carveol mitigates the development of the morphine anti-nociceptive tolerance, physical dependence, and conditioned place preference in mice. Heliyon 2024; 10:e27809. [PMID: 38496833 PMCID: PMC10944274 DOI: 10.1016/j.heliyon.2024.e27809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/28/2024] [Accepted: 03/06/2024] [Indexed: 03/19/2024] Open
Abstract
Emergence of analgesic tolerance and dependence to morphine is frequently the limiting factor in the use of this agent in the management of pain. Hence, this study aimed to investigate the beneficial effects of the natural compound carveol (CV) against morphine antinociceptive tolerance, dependence and conditioned place preference (CPP) in mice. Behavioural paradigms included hot plate and tail-flick (for tolerance), observation of withdrawal signs (for dependence) while biochemical tests involved the assays for mRNA expression, nitrite levels, antioxidants, and immunohistochemistry studies. Behavioural tests indicated that treatment with CV significantly attenuated the morphine analgesic tolerance, physical dependence and CPP in mice. It was observed during biochemical analysis that CV-treated animals exhibited reduced mRNA expression of inducible nitric oxide synthase (iNOS) and NR2B (an NMDA subtype). In addition, decreased levels of nitrite were observed in mouse hippocampus following CV treatment than morphine administration only. Further, CV enhanced the neuronal innate antioxidants including Glutathione-S-Transferase (GST), glutathione (GSH) and catalase (CAT), while curtailed lipid peroxidase (LPO) levels in mice brain tissues. Moreover, CV exerted significant anti-inflammatory effects as evidenced by reduced expression of TNF-α and p-NF-κB in these animals than with morphine treatment only. Together, anti-inflammatory and antioxidant effects might confer needed neuro-protection following morphine administration. These observations warrant further investigations of the beneficial role of CV as a novel agent in overcoming the development of tolerance and physical dependence following morphine use.
Collapse
Affiliation(s)
- Ismail Badshah
- Riphah Institute of Pharmaceutical Sciences, Islamabad, Pakistan
| | - Neelum Gul Qazi
- Department of Pharmacy, Iqra University, Islamabad, Pakistan
| | - Maira Anwar
- Riphah Institute of Pharmaceutical Sciences, Islamabad, Pakistan
| | - Bushra Shaukat
- Department of Pharmacy, Government College University, Faisalabad, Pakistan
| | - Muhammad Imran Khan
- Department of Biomedical Sciences, Pak-Austria Fachhochschule Institute of Applied Sciences and Technology, Haripur, Pakistan
| | - Babar Murtaza
- Riphah Institute of Pharmaceutical Sciences, Islamabad, Pakistan
| |
Collapse
|
14
|
Chow JJ, Pitts KM, Schoenbaum A, Costa KM, Schoenbaum G, Shaham Y. Different Effects of Peer Sex on Operant Responding for Social Interaction and Striatal Dopamine Activity. J Neurosci 2024; 44:e1887232024. [PMID: 38346894 PMCID: PMC10919252 DOI: 10.1523/jneurosci.1887-23.2024] [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: 10/05/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 03/08/2024] Open
Abstract
When rats are given discrete choices between social interactions with a peer and opioid or psychostimulant drugs, they choose social interaction, even after extensive drug self-administration experience. Studies show that like drug and nondrug food reinforcers, social interaction is an operant reinforcer and induces dopamine release. However, these studies were conducted with same-sex peers. We examined if peer sex influences operant social interaction and the role of estrous cycle and striatal dopamine in same- versus opposite-sex social interaction. We trained male and female rats (n = 13 responders/12 peers) to lever-press (fixed-ratio 1 [FR1] schedule) for 15 s access to a same- or opposite-sex peer for 16 d (8 d/sex) while tracking females' estrous cycle. Next, we transfected GRAB-DA2m and implanted optic fibers into nucleus accumbens (NAc) core and dorsomedial striatum (DMS). We then retrained the rats for 15 s social interaction (FR1 schedule) for 16 d (8 d/sex) and recorded striatal dopamine during operant responding for a peer for 8 d (4 d/sex). Finally, we assessed economic demand by manipulating FR requirements for a peer (10 d/sex). In male, but not female rats, operant responding was higher for the opposite-sex peer. Female's estrous cycle fluctuations had no effect on operant social interaction. Striatal dopamine signals for operant social interaction were dependent on the peer's sex and striatal region (NAc core vs DMS). Results indicate that estrous cycle fluctuations did not influence operant social interaction and that NAc core and DMS dopamine activity reflect sex-dependent features of volitional social interaction.
Collapse
Affiliation(s)
- Jonathan J Chow
- Intramural Research Program, NIDA, NIH, Baltimore, Maryland 21230
| | - Kayla M Pitts
- Intramural Research Program, NIDA, NIH, Baltimore, Maryland 21230
| | - Ansel Schoenbaum
- Intramural Research Program, NIDA, NIH, Baltimore, Maryland 21230
| | - Kauê M Costa
- Intramural Research Program, NIDA, NIH, Baltimore, Maryland 21230
| | | | - Yavin Shaham
- Intramural Research Program, NIDA, NIH, Baltimore, Maryland 21230
| |
Collapse
|
15
|
Lee JH, Kang S, Maier SU, Lee SA, Goldfarb EV, Ahn WY. Acute Stress Enhances Memory and Preference for Smoking-Related Associations in Smokers. Nicotine Tob Res 2024; 26:333-341. [PMID: 37589502 DOI: 10.1093/ntr/ntad152] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 08/08/2023] [Accepted: 08/13/2023] [Indexed: 08/18/2023]
Abstract
INTRODUCTION Nicotine dependence follows a chronic course that is characterized by repeated relapse, often driven by acute stress and rewarding memories of smoking retrieved from related contexts. These two triggers can also interact, with stress influencing retrieval of contextual memories. However, the roles of these processes in nicotine dependence remain unknown. AIMS AND METHODS We investigated how acute stress biases memory for smoking-associated contexts among smokers (N = 65) using a novel laboratory paradigm. On day 1, participants formed associations between visual stimuli of items (either neutral or related to smoking) and places (background scenes). On day 2 (24 hours later), participants were exposed to an acute laboratory-based stressor (socially evaluated cold pressor test; N = 32) or a matched control condition (N = 33) prior to being tested on their memory recognition and preferences for each item and place. We distinguished the accuracy of memory into specific (ie, precisely correct) or gist (ie, lure items with similar content) categories. RESULTS Results demonstrated that the stressor significantly induced physiological and subjective perceived stress responses, and that stressed smokers exhibited a memory bias in favor of smoking-related items. In addition, the stressed group displayed greater preference for both smoking-related items and places that had been paired with the smoking-related items. We also found suggestive evidence that stronger smoking-related memory biases were associated with more severe nicotine dependence (ie, years of smoking). CONCLUSIONS These results highlight the role of stress in biasing smokers toward remembering contexts associated with smoking, and amplifying their preference for these contexts. IMPLICATIONS The current study elucidates the role of acute stress in promoting memory biases favoring smoking-related associations among smokers. The results suggest that the retrieval of smoking-biased associative memory could be a crucial factor in stress-related nicotine seeking. This may lead to a potential intervention targeting the extinction of smoking-related context memories as a preventive strategy for stress-induced relapse.
Collapse
Affiliation(s)
- Jeung-Hyun Lee
- Department of Psychology, Seoul National University, Seoul, South Korea
| | - Sanghoon Kang
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Silvia U Maier
- Translational Neuromodeling Unit, University of Zurich, Zürich, Switzerland
| | - Sang Ah Lee
- Department of Brain and Cognitive Sciences, Seoul National University, Seoul, Korea
| | | | - Woo-Young Ahn
- Department of Psychology, Seoul National University, Seoul, South Korea
- Department of Brain and Cognitive Sciences, Seoul National University, Seoul, Korea
| |
Collapse
|
16
|
Craig AR. Resistance to change, of behavior and of theory. J Exp Anal Behav 2023; 120:440-456. [PMID: 37526100 DOI: 10.1002/jeab.875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/10/2023] [Indexed: 08/02/2023]
Abstract
The persistence of operant behavior when disrupted tends to be positively related to how often reinforcers were delivered in the past. Behavioral momentum theory describes this finding as the outcome of Pavlovian processes. That is, the relation between discriminative stimuli and reinforcers that were delivered in their presence strengthens behavior, thereby making it more likely to persist. If only the story were that simple. A growing number of findings challenge the basic tenets of behavioral momentum theory. Some even call into question whether Pavlovian relations contribute to persistence in the first place. In this paper, I will review behavioral momentum theory and some of the data that have been problematic for the theory. I will argue that despite these very real challenges, the theory provides important utility not only to basic analyses of response persistence but also to clinical interventions directed at long-term reductions in problem behavior. It, for example, has set the stage for the development of alternative conceptual analyses of resistance to change, two of which will be highlighted for readers. Moreover, behavioral momentum theory may tell us something important about the reasons it continues to have an influence on the field, despite the challenging data that deter it.
Collapse
|
17
|
Carter JS, Costa CC, Kearns AM, Reichel CM. Inhibition of Estradiol Signaling in the Basolateral Amygdala Impairs Extinction Memory Recall for Heroin-Conditioned Cues in a Sex-Specific Manner. Neuroendocrinology 2023; 114:207-222. [PMID: 37848008 PMCID: PMC10922099 DOI: 10.1159/000534647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 10/10/2023] [Indexed: 10/19/2023]
Abstract
INTRODUCTION Relapse is a major treatment barrier for opioid use disorder. Environmental cues become associated with the rewarding effects of opioids and can precipitate relapse, even after numerous unreinforced cue presentations, due to deficits in extinction memory recall (EMR). Estradiol (E2) modulates EMR of fear-related cues, but it is unknown whether E2 impacts EMR of reward cues and what brain region(s) are responsible for E2s effects. Here, we hypothesize that inhibition of E2 signaling in the basolateral amygdala (BLA) will impair EMR of a heroin-associated cue in both male and female rats. METHODS We pharmacologically manipulated E2 signaling to characterize the role of E2 in the BLA on heroin-cue EMR. Following heroin self-administration, during which a light/tone cue was co-presented with each heroin infusion, rats underwent cued extinction to extinguish the conditioned association between the light/tone and heroin. During extinction, E2 signaling in the BLA was blocked by an aromatase inhibitor or specific estrogen receptor (ER) antagonists. The next day, subjects underwent a cued test to assess heroin-cue EMR. RESULTS In both experiments, females took more heroin than males (mg/kg) and had higher operant responding during cued extinction. Inhibition of E2 synthesis in the BLA impaired heroin-cue EMR in both sexes. Notably, E2s actions are mediated by different ER mechanisms, ERα in males but ERβ in females. CONCLUSIONS This study is the first to demonstrate a behavioral role for centrally-produced E2 in the BLA and that E2 also impacts EMR of reward-associated stimuli in both sexes.
Collapse
Affiliation(s)
- Jordan S. Carter
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Caitlyn C. Costa
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Angela M. Kearns
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Carmela M. Reichel
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
18
|
Japarin RA, Harun N, Hassan Z, Müller CP. The dopamine D1 receptor antagonist SCH-23390 blocks the acquisition, but not expression of mitragynine-induced conditioned place preference in rats. Behav Brain Res 2023; 453:114638. [PMID: 37619769 DOI: 10.1016/j.bbr.2023.114638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/21/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
Mitragynine (MG) is the primary active constituent of Mitragyna speciosa Korth (kratom), a psychoactive Southeast Asian plant with potential therapeutic use. Numerous studies support roles of dopaminergic system in drug reward. However, the involvement of the dopaminergic system in mediating MG reward and drug-seeking is poorly understood. Using conditioned place preference (CPP) paradigm, the present study aims to evaluate the roles of the dopamine (DA) D1 receptor in the acquisition and expression of MG-induced CPP in rats. The effects of SCH-23390, a selective DA D1 receptor antagonist, on the acquisition of MG-induced CPP were first investigated. Rats were pre-treated systemically with SCH-23390 (0, 0.1 and 0.3 mg/kg, i.p.) prior to MG (10 mg/kg) conditioning sessions. Next, we tested the effects of the DA D1 receptor antagonist on the expression of MG-induced CPP. Furthermore, the effects of a MG-priming dose (5 mg/kg) on the reinstatement of extinguished CPP were tested. The results showed that SCH-23390 dose-dependently suppressed the acquisition of a MG-induced CPP. In contrast, SCH-23390 had no effect on the expression of a MG-induced CPP. The findings of this study suggested a crucial role of the DA D1 receptor in the acquisition, but not the expression of the rewarding effects of MG in a CPP test. Furthermore, blockade of the D1-like receptor during conditioning did not prevent MG priming effects on CPP reinstatement test, suggesting no role for the DA D1 receptor in reinstatement sensitivity.
Collapse
Affiliation(s)
- Rima Atria Japarin
- Centre for Drug Research, Universiti Sains Malaysia, 11800 Gelugor, Penang, Malaysia
| | - Norsyifa Harun
- Centre for Drug Research, Universiti Sains Malaysia, 11800 Gelugor, Penang, Malaysia.
| | - Zurina Hassan
- Centre for Drug Research, Universiti Sains Malaysia, 11800 Gelugor, Penang, Malaysia
| | - Christian P Müller
- Centre for Drug Research, Universiti Sains Malaysia, 11800 Gelugor, Penang, Malaysia; Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University Erlangen-Nuremberg, Schwabachanlage 6, 91054 Erlangen, Germany
| |
Collapse
|
19
|
Charpentier ANH, Olekanma DI, Valade CT, Reeves CA, Cho BR, Arguello AA. Influence of reconsolidation in maintenance of cocaine-associated contextual memories formed during adolescence or adulthood. Sci Rep 2023; 13:13936. [PMID: 37626103 PMCID: PMC10457301 DOI: 10.1038/s41598-023-39949-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Adolescents are at increased risk to develop substance use disorders and suffer from relapse throughout life. Targeted weakening of drug-associated memories has been shown to reduce relapse-like behavior in adult rats, however this process has been understudied in adolescents. We aimed to examine whether adolescent-formed, cocaine-associated memories could be manipulated via reconsolidation mechanisms. To accomplish this objective, we used an abbreviated operant cocaine self-administration paradigm (ABRV Coc-SA). Adult and adolescent rats received jugular catheterization surgery followed by ABRV Coc-SA in a distinct context for 2 h, 2×/day over 5 days. Extinction training (EXT) occurred in a second context for 2 h, 2×/day over 4 days. To retrieve cocaine-context memories, rats were exposed to the cocaine-paired context for 15 min, followed by subcutaneous injection of vehicle or the protein synthesis inhibitor cycloheximide (2.5 mg/kg). Two additional EXT sessions were conducted before a 2 h reinstatement test in the cocaine-paired context to assess cocaine-seeking behavior. We find that both adult and adolescent cocaine-exposed rats show similar levels of cocaine-seeking behavior regardless of post-reactivation treatment. Our results suggest that systemic treatment with the protein synthesis inhibitor cycloheximide does not impair reconsolidation of cocaine-context memories and subsequent relapse during adulthood or adolescence.
Collapse
Affiliation(s)
- André N Herrera Charpentier
- Department of Psychology, Behavioral Neuroscience, Michigan State University (MSU), Interdisciplinary Science and Technology Building, West Rm. 4010, 766 Service Rd., East Lansing, MI, 48824, USA
| | - Doris I Olekanma
- Department of Psychology, Behavioral Neuroscience, Michigan State University (MSU), Interdisciplinary Science and Technology Building, West Rm. 4010, 766 Service Rd., East Lansing, MI, 48824, USA
| | - Christian T Valade
- Department of Psychology, Behavioral Neuroscience, Michigan State University (MSU), Interdisciplinary Science and Technology Building, West Rm. 4010, 766 Service Rd., East Lansing, MI, 48824, USA
| | - Christopher A Reeves
- Department of Psychology, Behavioral Neuroscience, Michigan State University (MSU), Interdisciplinary Science and Technology Building, West Rm. 4010, 766 Service Rd., East Lansing, MI, 48824, USA
| | - Bo Ram Cho
- Department of Psychology, Behavioral Neuroscience, Michigan State University (MSU), Interdisciplinary Science and Technology Building, West Rm. 4010, 766 Service Rd., East Lansing, MI, 48824, USA
| | - Amy A Arguello
- Department of Psychology, Behavioral Neuroscience, Michigan State University (MSU), Interdisciplinary Science and Technology Building, West Rm. 4010, 766 Service Rd., East Lansing, MI, 48824, USA.
| |
Collapse
|
20
|
Humburg BA, Bardo MT. Renewal of cocaine seeking using social and nonsocial contextual stimuli. Psychopharmacology (Berl) 2023:10.1007/s00213-023-06414-7. [PMID: 37391496 PMCID: PMC10806405 DOI: 10.1007/s00213-023-06414-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/20/2023] [Indexed: 07/02/2023]
Abstract
RATIONALE Various nonsocial cues have been used as stimuli to examine the contextual control of drug seeking behavior, but little is known about the role of social stimuli. OBJECTIVES This study determined if renewal of cocaine seeking is differentially controlled using a context consisting of either a social peer and/or house light illumination. METHODS In Experiment 1, male and female rats trained to self-administer cocaine in the presence of a same-sex social peer and house light illumination (context A). Following self-administration, rats were randomly assigned to either an AAA (control) or ABA (renewal) group for extinction. For AAA rats, extinction consisted of the same context A as self-administration; for ABA rats, extinction occurred without the peer or house light (context B). Following extinction, renewal of cocaine seeking occurred by testing the peer alone, house light alone, and the peer + house light combination. Experiment 2 was conducted to ensure that the house light alone was sufficiently salient to produce renewal. RESULTS Both experiments showed that rats acquired cocaine self-administration and extinguished lever pressing. In Experiment 1, the ABA group renewed cocaine seeking to the peer and peer + house light, but not to the house light alone. In Experiment 2, ABA rats renewed cocaine seeking to the house light alone, indicating it was sufficiently salient to produce renewal. The AAA group did not show renewal in either experiment. CONCLUSION Social peers serve as powerful stimuli that can overshadow nonsocial visual stimuli in the renewal of cocaine seeking.
Collapse
Affiliation(s)
- Bree A Humburg
- Department of Psychology, University of Kentucky, Biomedical Biological Science Research Building, Room 447, 741 S. Limestone, Lexington, KY, 40536-0509, USA
| | - Michael T Bardo
- Department of Psychology, University of Kentucky, Biomedical Biological Science Research Building, Room 447, 741 S. Limestone, Lexington, KY, 40536-0509, USA.
| |
Collapse
|
21
|
Luján MÁ, Oliver BL, Young-Morrison R, Engi SA, Zhang LY, Wenzel JM, Li Y, Zlebnik NE, Cheer JF. A multivariate regressor of patterned dopamine release predicts relapse to cocaine. Cell Rep 2023; 42:112553. [PMID: 37224011 DOI: 10.1016/j.celrep.2023.112553] [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: 11/07/2022] [Revised: 02/28/2023] [Accepted: 05/05/2023] [Indexed: 05/26/2023] Open
Abstract
Understanding mesolimbic dopamine adaptations underlying vulnerability to drug relapse is essential to inform prognostic tools for effective treatment strategies. However, technical limitations have hindered the direct measurement of sub-second dopamine release in vivo for prolonged periods of time, making it difficult to gauge the weight that these dopamine abnormalities have in determining future relapse incidence. Here, we use the fluorescent sensor GrabDA to record, with millisecond resolution, every single cocaine-evoked dopamine transient in the nucleus accumbens (NAc) of freely moving mice during self-administration. We reveal low-dimensional features of patterned dopamine release that are strong predictors of cue-induced reinstatement of cocaine seeking. Additionally, we report sex-specific differences in cocaine-related dopamine responses related to a greater resistance to extinction in males compared with females. These findings provide important insights into the sufficiency of NAc dopamine signaling dynamics-in interaction with sex-for recapitulating persistent cocaine seeking and future relapse vulnerability.
Collapse
Affiliation(s)
- Miguel Á Luján
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Brandon L Oliver
- Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, CA, USA
| | - Reana Young-Morrison
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sheila A Engi
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Lan-Yuan Zhang
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jennifer M Wenzel
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China; PKU-IDG/McGovern Institute for Brain Research, Beijing, China; Peking-Tsinghua Center for Life Sciences, Beijing, China
| | - Natalie E Zlebnik
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA; Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, CA, USA.
| | - Joseph F Cheer
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
22
|
Emerson SD, Chevée M, Mews P, Calipari ES. The transcriptional response to acute cocaine is inverted in male mice with a history of cocaine self-administration and withdrawal throughout the mesocorticolimbic system. Mol Cell Neurosci 2023; 125:103823. [PMID: 36868542 PMCID: PMC10247534 DOI: 10.1016/j.mcn.2023.103823] [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: 11/06/2022] [Revised: 01/30/2023] [Accepted: 02/14/2023] [Indexed: 03/05/2023] Open
Abstract
A large body of work has demonstrated that cocaine-induced changes in transcriptional regulation play a central role in the onset and maintenance of cocaine use disorder. An underappreciated aspect of this area of research, however, is that the pharmacodynamic properties of cocaine can change depending on an organism's previous drug-exposure history. In this study, we utilized RNA sequencing to characterize how the transcriptome-wide effects of acute cocaine exposure were altered by a history of cocaine self-administration and long-term withdrawal (30 days) in the ventral tegmental area (VTA), nucleus accumbens (NAc), and prefrontal cortex (PFC) in male mice. First, we found that the gene expression patterns induced by a single cocaine injection (10 mg/kg) were discordant between cocaine-naïve mice and mice in withdrawal from cocaine self-administration. Specifically, the same genes that were upregulated by acute cocaine in cocaine-naïve mice were downregulated by the same dose of cocaine in mice undergoing long-term withdrawal; the same pattern of opposite regulation was observed for the genes downregulated by initial acute cocaine exposure. When we analyzed this dataset further, we found that the gene expression patterns that were induced by long-term withdrawal from cocaine self-administration showed a high degree of overlap with the gene expression patterns of acute cocaine exposure - even though animals had not consumed cocaine in 30 days. Interestingly, cocaine re-exposure at this withdrawal time point reversed this expression pattern. Finally, we found that this pattern was similar across the VTA, PFC, NAc, and within each brain region the same genes were induced by acute cocaine, re-induced during long-term withdrawal, and reversed by cocaine re-exposure. Together, we identified a longitudinal pattern of gene regulation that is conserved across the VTA, PFC, and NAc, and characterized the genes constituting this pattern in each brain region.
Collapse
Affiliation(s)
- Soren D Emerson
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
| | - Maxime Chevée
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
| | - Philipp Mews
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Erin S Calipari
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; Department of Psychiatry and Behavioral Sciences, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
23
|
Daws SE, Gillespie A. Circular RNA regulation and function in drug seeking phenotypes. Mol Cell Neurosci 2023; 125:103841. [PMID: 36935046 PMCID: PMC10247439 DOI: 10.1016/j.mcn.2023.103841] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/27/2023] [Accepted: 03/06/2023] [Indexed: 03/19/2023] Open
Abstract
Drug overdoses have increased dramatically in the United States over the last decade where they are now the leading cause of accidental death. To develop efficient therapeutic options for decreasing drug consumption and overdose risk, it is critical to understand the neurobiological changes induced by drug exposure. Chronic systemic exposure to all drug classes, including opioids, psychostimulants, nicotine, cannabis, and alcohol, induces profound molecular neuroadaptations within the central nervous system that may reveal crucial information about the lasting effects that these substances impart on brain cells. Transcriptome analyses of messenger RNAs (mRNAs) have identified gene patterns in the brain that result from exposure to various classes of drugs. However, mRNAs represent only a small fraction of the RNA within the cell, and drug exposure also impacts other classes of RNA that are largely understudied, especially circular RNAs. Circular RNAs (circRNAs) are a naturally occurring RNA species formed from back-splicing events during mRNA processing and are enriched in the nervous system. circRNAs are a pleiotropic class of RNAs and have a diverse impact on cellular function, with putative functions including regulation of mRNA transcription, protein translation, microRNA sponging, and sequestration of RNA-binding proteins. Recent studies have demonstrated that circRNAs can modulate cognition and are regulated in the brain in response to drug exposure, yet very few studies have explored the contribution of circRNAs to drug seeking phenotypes. In this review, we will provide an overview of the mechanisms of circRNA function in the cell to highlight how drug-induced circRNA dysregulation may impact the molecular substrates that mediate drug seeking behavior and the current studies that have reported drug-induced dysregulation of circRNAs in the brain. Furthermore, we will discuss how principles of circRNA biology can be adapted to study circRNAs in models of drug exposure and seek to provide further insight into the neurobiology of addiction.
Collapse
Affiliation(s)
- Stephanie E Daws
- Center for Substance Abuse Research, Temple University, Philadelphia, PA, USA; Department of Neural Sciences, Temple University, Philadelphia, PA, USA.
| | - Aria Gillespie
- Center for Substance Abuse Research, Temple University, Philadelphia, PA, USA; Department of Neural Sciences, Temple University, Philadelphia, PA, USA
| |
Collapse
|
24
|
Claypool SM, Reiner DJ, Behdin S, Orihuel J, Batista A, Caldwell KE, Chow JJ, Bossert JM, Rubio FJ, Hope BT, Shaham Y. Role of Piriform Cortex and Its Afferent Projections in Relapse to Fentanyl Seeking after Food Choice-Induced Voluntary Abstinence. J Neurosci 2023; 43:2597-2614. [PMID: 36898838 PMCID: PMC10082459 DOI: 10.1523/jneurosci.0034-23.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/15/2023] [Accepted: 02/20/2023] [Indexed: 03/12/2023] Open
Abstract
We previously demonstrated a role of piriform cortex (Pir) in relapse to fentanyl seeking after food choice-induced voluntary abstinence. Here, we used this model to further study the role of Pir and its afferent projections in fentanyl relapse. We trained male and female rats to self-administer palatable food pellets for 6 d (6 h/day) and fentanyl (2.5 µg/kg/infusion, i.v.) for 12 d (6 h/day). We assessed relapse to fentanyl seeking after 12 voluntary abstinence sessions, achieved through a discrete choice procedure between fentanyl and palatable food (20 trials/session). We determined projection-specific activation of Pir afferents during fentanyl relapse with Fos plus the retrograde tracer cholera toxin B (injected into Pir). Fentanyl relapse was associated with increased Fos expression in anterior insular cortex (AI) and prelimbic cortex (PL) neurons projecting to Pir. We next used an anatomical disconnection procedure to determine the causal role of these two projections (AI→Pir and PL→Pir) in fentanyl relapse. Contralateral but not ipsilateral disconnection of AI→Pir projections decreased fentanyl relapse but not reacquisition of fentanyl self-administration. In contrast, contralateral but not ipsilateral disconnection of PL→Pir projections modestly decreased reacquisition but not relapse. Fluorescence-activated cell sorting and quantitative PCR data showed molecular changes within Pir Fos-expressing neurons associated with fentanyl relapse. Finally, we found minimal or no sex differences in fentanyl self-administration, fentanyl versus food choice, and fentanyl relapse. Our results indicate that AI→Pir and PL→Pir projections play dissociable roles in nonreinforced relapse to fentanyl seeking versus reacquisition of fentanyl self-administration after food choice-induced voluntary abstinence.SIGNIFICANCE STATEMENT We previously showed a role of Pir in fentanyl relapse after food choice-induced voluntary abstinence in rats, a procedure mimicking human abstinence or a significant reduction in drug self-administration because of the availability of alternative nondrug rewards. Here, we aimed to further characterize the role of Pir in fentanyl relapse by investigating the role of Pir afferent projections and analyzing molecular changes in relapse-activated Pir neurons. We identified dissociable roles of two Pir afferent projections (AI→Pir and PL→Pir) in relapse to fentanyl seeking versus reacquisition of fentanyl self-administration after voluntary abstinence. We also characterized molecular changes within Pir Fos-expressing neurons associated with fentanyl relapse.
Collapse
Affiliation(s)
- Sarah M Claypool
- Behavioral Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224
| | - David J Reiner
- Behavioral Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224
| | - Sana Behdin
- Behavioral Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224
| | - Javier Orihuel
- Behavioral Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224
| | - Ashley Batista
- Behavioral Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224
| | - Kiera E Caldwell
- Behavioral Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224
| | - Jonathan J Chow
- Behavioral Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224
| | - Jennifer M Bossert
- Behavioral Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224
| | - F Javier Rubio
- Behavioral Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224
| | - Bruce T Hope
- Behavioral Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224
| | - Yavin Shaham
- Behavioral Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224
| |
Collapse
|
25
|
Park A, Jacob AD, Hsiang HLL, Frankland PW, Howland JG, Josselyn SA. Formation and fate of an engram in the lateral amygdala supporting a rewarding memory in mice. Neuropsychopharmacology 2023; 48:724-733. [PMID: 36261624 PMCID: PMC10066178 DOI: 10.1038/s41386-022-01472-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/27/2022] [Accepted: 10/01/2022] [Indexed: 11/08/2022]
Abstract
Memories allow past experiences to guide future decision making and behavior. Sparse ensembles of neurons, known as engrams, are thought to store memories in the brain. Most previous research has focused on engrams supporting threatening or fearful memories where results show that neurons involved in a particular engram ("engram neurons") are both necessary and sufficient for memory expression. Far less is understood about engrams supporting appetitive or rewarding memories. As circumstances and environments are dynamic, the fate of a previously acquired engram with changing circumstances is unknown. Here we examined how engrams supporting a rewarding cue-cocaine memory are formed and whether this original engram is important in reinstatement of memory-guided behavior following extinction. Using a variety of techniques, we show that neurons in the lateral amygdala are allocated to an engram based on relative neuronal excitability at training. Furthermore, once allocated, these neurons become both necessary and sufficient for behavior consistent with recall of that rewarding memory. Allocated neurons are also critical for cocaine-primed reinstatement of memory-guided behavior following extinction. Moreover, artificial reactivation of initially allocated neurons supports reinstatement-like behavior following extinction even in the absence of cocaine-priming. Together, these findings suggest that cocaine priming after extinction reactivates the original engram, and that memory-guided reinstatement behavior does not occur in the absence of this reactivation. Although we focused on neurons in one brain region only, our findings that manipulations of lateral amygdala engram neurons alone were sufficient to impact memory-guided behavior indicate that the lateral amygdala is a critical hub region in what may be a larger brain-wide engram.
Collapse
Affiliation(s)
- Albert Park
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON, M5G 1X8, Canada
| | - Alexander D Jacob
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON, M5G 1X8, Canada
- Department of Psychology, University of Toronto, Toronto, ON, M5G 1X8, Canada
| | - Hwa-Lin Liz Hsiang
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON, M5G 1X8, Canada
| | - Paul W Frankland
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON, M5G 1X8, Canada
- Department of Psychology, University of Toronto, Toronto, ON, M5G 1X8, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5G 1X8, Canada
- Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, ON, M5G 1M1, Canada
| | - John G Howland
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada.
| | - Sheena A Josselyn
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON, M5G 1X8, Canada.
- Department of Psychology, University of Toronto, Toronto, ON, M5G 1X8, Canada.
- Department of Physiology, University of Toronto, Toronto, ON, M5G 1X8, Canada.
| |
Collapse
|
26
|
Japarin RA, Harun N, Hassan Z, Shoaib M. Mitragynine, a primary constituent of kratom reinstates morphine-seeking behaviour in rats. Behav Pharmacol 2023; 34:123-130. [PMID: 36752325 DOI: 10.1097/fbp.0000000000000715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Mitragynine (MG) is a pharmacologically active alkaloid derived from the leaves of Mitragyna speciosa Korth (Kratom). This plant has sparked significant interest as a potential alternative treatment for managing opioid dependence and withdrawal due to its opioid-like pharmacological effects. However, whether MG exposure would trigger opioid-seeking behaviour following abstinence has not been investigated. The present study examined the effects of MG priming on morphine-seeking behaviour in rats. Male Sprague-Dawley rats were initially trained to intravenously self-administer morphine (0.5 mg/kg/infusion) under a fixed ratio-3 schedule of reinforcement. Removal of both morphine infusions and drug-associated cues led to the subsequent extinction of the drug-seeking behaviour. Tests of reinstatement were made following exposure to a randomised order of intraperitoneal injections of MG (3, 10 and 30 mg/kg), morphine (5 mg/kg) and vehicle. Significant levels of drug-seeking behaviour were observed following extended access to morphine self-administration, which was extinguished following removal of morphine and cues indicative of morphine-seeking behaviour, supporting the relapse model. The present finding demonstrated that MG priming in a dose of 10 mg/kg resulted in the reinstatement of morphine-seeking behaviour, whereas the higher MG dose (30 mg/kg) tested suppressed the seeking response. This study indicated that exposure to a low MG dose may increase the likelihood of relapsing to opioids, suggesting that the potential of MG as a treatment for opioid management merits further scientific assessment of its ability to trigger relapse to opioid abuse.
Collapse
Affiliation(s)
| | - Norsyifa Harun
- Centre for Drug Research, Universiti Sains Malaysia, Gelugor, Malaysia
| | - Zurina Hassan
- Centre for Drug Research, Universiti Sains Malaysia, Gelugor, Malaysia
| | - Mohammed Shoaib
- Institute of Neuroscience, Medical School, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
27
|
Horton AL, Campbell EJ, Aumann TD, O'Brien KR, Lawrence AJ, Brown RM. Addiction-like behaviour towards high-fat high-sugar food predicts relapse propensity in both obesity prone and obesity resistant C57BL/6 J mice. Prog Neuropsychopharmacol Biol Psychiatry 2023; 121:110654. [PMID: 36209772 DOI: 10.1016/j.pnpbp.2022.110654] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 07/11/2022] [Accepted: 10/02/2022] [Indexed: 11/07/2022]
Abstract
Compulsive overeating of palatable food is thought to underlie some forms of obesity. Similarities are often observed in the behavioural symptomology and the neuropathophysiology underlying substance use disorder and compulsive overeating. As such, preclinical animal models which assess addiction-like behaviour towards food may assist the understanding of the neurobiology underlying overeating behaviour. Further, the relationship between these behaviours and the propensity for diet-induced obesity warrants examination. In this study we investigated the relationship between the propensity for diet-induced obesity (DIO) and addiction-like behaviour towards highly palatable food in C57BL/6 J mice as measured by a 3-criteria model. We also examined the extent to which performance on this 3-criteria model predicted two key hallmark features of addiction - resistance to extinction and relapse propensity (as measured by reinstatement of lever pressing). C57BL/6 J mice were allowed free access to a palatable diet for 8 weeks then separated by weight gain into DIO-prone and DIO-resistant subgroups. Access to palatable food was then restricted to daily operant self-administration sessions whereby addiction-like behaviour towards a high-fat high-sugar food reward was assessed using a 3-criteria model similar to that used to assess addiction-like behaviour towards drugs of abuse. In contrast to findings in rats, no difference in addiction-like behaviour towards food was observed between obesity prone (OP) and obesity resistant (OR) mice. Similarly, principal components analysis found no distinct patterns in the relationship between addiction-like behaviours across treatment groups. This suggests that the strain and species of rodent may be critical for studying the mechanisms underlying pathological overconsumption. Further analysis revealed that the extent of performance on the 3-criteria model correlated with the propensity for C57BL/6 J mice to both extinguish food seeking behaviour and "relapse" after a period of withdrawal. This finding was evident across all groups, regardless of DIO. Collectively, these data validate the 3-criteria model as a robust model to comprehensively assess food addiction-like behaviour in mice, regardless of prior food intake history.
Collapse
Affiliation(s)
- Anna L Horton
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, VIC, Australia; Department of Biochemistry and Pharmacology, University of Melbourne, VIC, Australia; Florey Department of Neuroscience & Mental Health, University of Melbourne, VIC, Australia
| | - Erin J Campbell
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, VIC, Australia; Florey Department of Neuroscience & Mental Health, University of Melbourne, VIC, Australia; School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Timothy D Aumann
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, VIC, Australia
| | - Katrina R O'Brien
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, VIC, Australia
| | - Andrew J Lawrence
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, VIC, Australia; Florey Department of Neuroscience & Mental Health, University of Melbourne, VIC, Australia
| | - Robyn M Brown
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, VIC, Australia; Department of Biochemistry and Pharmacology, University of Melbourne, VIC, Australia; Florey Department of Neuroscience & Mental Health, University of Melbourne, VIC, Australia.
| |
Collapse
|
28
|
Bossert JM, Mejias-Aponte CA, Saunders T, Altidor L, Emery M, Fredriksson I, Batista A, Claypool SM, Caldwell KE, Reiner DJ, Chow JJ, Foltz M, Kumar V, Seasholtz A, Hughes E, Filipiak W, Harvey BK, Richie CT, Vautier F, Gomez JL, Michaelides M, Kieffer BL, Watson SJ, Akil H, Shaham Y. Effect of Selective Lesions of Nucleus Accumbens µ-Opioid Receptor-Expressing Cells on Heroin Self-Administration in Male and Female Rats: A Study with Novel Oprm1-Cre Knock-in Rats. J Neurosci 2023; 43:1692-1713. [PMID: 36717230 PMCID: PMC10010456 DOI: 10.1523/jneurosci.2049-22.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/29/2022] [Accepted: 01/18/2023] [Indexed: 02/01/2023] Open
Abstract
The brain µ-opioid receptor (MOR) is critical for the analgesic, rewarding, and addictive effects of opioid drugs. However, in rat models of opioid-related behaviors, the circuit mechanisms of MOR-expressing cells are less known because of a lack of genetic tools to selectively manipulate them. We introduce a CRISPR-based Oprm1-Cre knock-in transgenic rat that provides cell type-specific genetic access to MOR-expressing cells. After performing anatomic and behavioral validation experiments, we used the Oprm1-Cre knock-in rats to study the involvement of NAc MOR-expressing cells in heroin self-administration in male and female rats. Using RNAscope, autoradiography, and FISH chain reaction (HCR-FISH), we found no differences in Oprm1 expression in NAc, dorsal striatum, and dorsal hippocampus, or MOR receptor density (except dorsal striatum) or function between Oprm1-Cre knock-in rats and wildtype littermates. HCR-FISH assay showed that iCre is highly coexpressed with Oprm1 (95%-98%). There were no genotype differences in pain responses, morphine analgesia and tolerance, heroin self-administration, and relapse-related behaviors. We used the Cre-dependent vector AAV1-EF1a-Flex-taCasp3-TEVP to lesion NAc MOR-expressing cells. We found that the lesions decreased acquisition of heroin self-administration in male Oprm1-Cre rats and had a stronger inhibitory effect on the effort to self-administer heroin in female Oprm1-Cre rats. The validation of an Oprm1-Cre knock-in rat enables new strategies for understanding the role of MOR-expressing cells in rat models of opioid addiction, pain-related behaviors, and other opioid-mediated functions. Our initial mechanistic study indicates that lesioning NAc MOR-expressing cells had different effects on heroin self-administration in male and female rats.SIGNIFICANCE STATEMENT The brain µ-opioid receptor (MOR) is critical for the analgesic, rewarding, and addictive effects of opioid drugs. However, in rat models of opioid-related behaviors, the circuit mechanisms of MOR-expressing cells are less known because of a lack of genetic tools to selectively manipulate them. We introduce a CRISPR-based Oprm1-Cre knock-in transgenic rat that provides cell type-specific genetic access to brain MOR-expressing cells. After performing anatomical and behavioral validation experiments, we used the Oprm1-Cre knock-in rats to show that lesioning NAc MOR-expressing cells had different effects on heroin self-administration in males and females. The new Oprm1-Cre rats can be used to study the role of brain MOR-expressing cells in animal models of opioid addiction, pain-related behaviors, and other opioid-mediated functions.
Collapse
Affiliation(s)
- Jennifer M Bossert
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | - Carlos A Mejias-Aponte
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | | | - Lindsay Altidor
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | | | - Ida Fredriksson
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | - Ashley Batista
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | - Sarah M Claypool
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | - Kiera E Caldwell
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | - David J Reiner
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | - Jonathan J Chow
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | | | - Vivek Kumar
- University of Michigan, Ann Arbor, Michigan, 48104
| | | | | | | | - Brandon K Harvey
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | - Christopher T Richie
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | - Francois Vautier
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | - Juan L Gomez
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | - Michael Michaelides
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| | - Brigitte L Kieffer
- University of Strasbourg-Institut National de la Santé et de la Recherche Médicale U1114, Strasbourg, France, 67084
| | | | - Huda Akil
- University of Michigan, Ann Arbor, Michigan, 48104
| | - Yavin Shaham
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, Baltimore, Maryland, 21224
| |
Collapse
|
29
|
Moorman DE, Aston-Jones G. Prelimbic and infralimbic medial prefrontal cortex neuron activity signals cocaine seeking variables across multiple timescales. Psychopharmacology (Berl) 2023; 240:575-594. [PMID: 36464693 PMCID: PMC10406502 DOI: 10.1007/s00213-022-06287-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022]
Abstract
RATIONALE AND OBJECTIVES The prefrontal cortex is critical for execution and inhibition of reward seeking. Neural manipulation of rodent medial prefrontal cortex (mPFC) subregions differentially impacts execution and inhibition of cocaine seeking. Dorsal, or prelimbic (PL), and ventral, or infralimbic (IL) mPFC are implicated in cocaine seeking or extinction of cocaine seeking, respectively. This differentiation is not seen across all studies, indicating that further research is needed to understand specific mPFC contributions to drug seeking. METHODS We recorded neuronal activity in mPFC subregions during cocaine self-administration, extinction, and cue- and cocaine-induced reinstatement of cocaine seeking. RESULTS Both PL and IL neurons were phasically responsive around lever presses during cocaine self-administration, and activity in both areas was reduced during extinction. During both cue- and, to a greater extent, cocaine-induced reinstatement, PL neurons exhibited significantly elevated responses, in line with previous studies demonstrating a role for the region in relapse. The enhanced PL signaling in cocaine-induced reinstatement was driven by strong excitation and inhibition in different groups of neurons. Both of these response types were stronger in PL vs. IL neurons. Finally, we observed tonic changes in activity in all tasks phases, reflecting both session-long contextual modulation as well as minute-to-minute activity changes that were highly correlated with brain cocaine levels and motivation associated with cocaine seeking. CONCLUSIONS Although some differences were observed between PL and IL neuron activity across sessions, we found no evidence of a go/stop dichotomy in PL/IL function. Instead, our results demonstrate temporally heterogeneous prefrontal signaling during cocaine seeking and extinction in both PL and IL, revealing novel and complex functions for both regions during these behaviors. This combination of findings argues that mPFC neurons, in both PL and IL, provide multifaceted contributions to the regulation of drug seeking and addiction.
Collapse
Affiliation(s)
- David E Moorman
- Department of Psychological and Brain Sciences & Neuroscience and Behavior Graduate Program, University of Massachusetts Amherst, Amherst, MA, 01003, USA.
| | - Gary Aston-Jones
- Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, NJ, 08854, USA
| |
Collapse
|
30
|
Olekanma DI, Reeves CA, Cho BR, Herrera Charpentier AN, Gerena J, Bal A, Arguello AA. Context-drug-associations and reinstatement of drug-seeking behavior in male rats: Adolescent and adult time-dependent effects. Neurobiol Learn Mem 2023; 199:107722. [PMID: 36639018 DOI: 10.1016/j.nlm.2023.107722] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/27/2022] [Accepted: 01/02/2023] [Indexed: 01/11/2023]
Abstract
RATIONALE Drug use during adolescence results in a life-long risk to develop substance-use disorders. Adolescent rats are sensitive to different drug-associated cues, compared to adults; however, the contribution of adolescent-formed context-drug-associations to elicit relapse-like behavior is underexplored. OBJECTIVES The present study compared the effect of adolescent vs adult-formed context-drug associations to elicit time-dependent increases in cocaine-seeking behavior. This objective was accomplished using an abbreviated (ABRV) operant cocaine self-administration (Coc-SA), Extinction (EXT) paradigm, with cocaine-seeking tests occurring 1 day after training (T1, early relapse) or following 15 days of abstinence (T15, late relapse). METHODS Adolescent and adult rats received ABRV Coc-SA in a distinct context (2 hr, 2x/day over 5 days) then EXT in a second context (2 hr, 2x/day over 4 days). Adolescent or adult cocaine-exposed rats were then tested (2 hr, non-rewarded) in either the previous EXT or Coc-paired contexts during early or late relapse. RESULTS & CONCLUSIONS As previously reported, both adolescent and adult cocaine-exposed rats displayed similar magnitudes of cocaine intake and lever presses during Coc-SA, EXT, and early relapse. Independent analysis of adolescent and adult groups revealed differences in lever responding, specifically rats with cocaine exposure during adolescence showed time-dependent increases in lever responding during late relapse. These data suggest that cocaine-context associations formed during adolescence can elicit craving during adulthood and that these age-specific differences in contextual sensitivity may not be immediately observed at early relapse periods.
Collapse
Affiliation(s)
- Doris I Olekanma
- Psychology Dept., Michigan State University, Interdisciplinary Science and Technology Building, West Rm. 4010, 766 Service Rd., East Lansing, MI 48824, USA
| | - Christopher A Reeves
- Psychology Dept., Michigan State University, Interdisciplinary Science and Technology Building, West Rm. 4010, 766 Service Rd., East Lansing, MI 48824, USA
| | - Bo Ram Cho
- Psychology Dept., Michigan State University, Interdisciplinary Science and Technology Building, West Rm. 4010, 766 Service Rd., East Lansing, MI 48824, USA
| | - André N Herrera Charpentier
- Psychology Dept., Michigan State University, Interdisciplinary Science and Technology Building, West Rm. 4010, 766 Service Rd., East Lansing, MI 48824, USA
| | - Jennifer Gerena
- Psychology Dept., Michigan State University, Interdisciplinary Science and Technology Building, West Rm. 4010, 766 Service Rd., East Lansing, MI 48824, USA
| | - Aneesh Bal
- Psychology Dept., Michigan State University, Interdisciplinary Science and Technology Building, West Rm. 4010, 766 Service Rd., East Lansing, MI 48824, USA
| | - Amy A Arguello
- Psychology Dept., Michigan State University, Interdisciplinary Science and Technology Building, West Rm. 4010, 766 Service Rd., East Lansing, MI 48824, USA.
| |
Collapse
|
31
|
Beck A, Ebrahimi C, Rosenthal A, Charlet K, Heinz A. The Dopamine System in Mediating Alcohol Effects in Humans. Curr Top Behav Neurosci 2023. [PMID: 36705911 DOI: 10.1007/7854_2022_415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Brain-imaging studies show that the development and maintenance of alcohol use disorder (AUD) is determined by a complex interaction of different neurotransmitter systems and multiple psychological factors. In this context, the dopaminergic reinforcement system appears to be of fundamental importance. We focus on the excitatory and depressant effects of acute versus chronic alcohol intake and its impact on dopaminergic neurotransmission. Furthermore, we describe alterations in dopaminergic neurotransmission as associated with symptoms of alcohol dependence. We specifically focus on neuroadaptations to chronic alcohol consumption and their effect on central processing of alcohol-associated and reward-related stimuli. Altered reward processing, complex conditioning processes, impaired reinforcement learning, and increased salience attribution to alcohol-associated stimuli enable alcohol cues to drive alcohol seeking and consumption. Finally, we will discuss how the neurobiological and neurochemical mechanisms of alcohol-associated alterations in reward processing and learning can interact with stress, cognition, and emotion processing.
Collapse
Affiliation(s)
- Anne Beck
- Faculty of Health, Health and Medical University, Potsdam, Germany
| | - Claudia Ebrahimi
- Department of Psychiatry and Neurosciences, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt- Universität zu Berlin, Berlin, Germany
| | - Annika Rosenthal
- Department of Psychiatry and Neurosciences, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt- Universität zu Berlin, Berlin, Germany
| | - Katrin Charlet
- Department of Psychiatry and Neurosciences, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt- Universität zu Berlin, Berlin, Germany
| | - Andreas Heinz
- Department of Psychiatry and Neurosciences, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt- Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
32
|
Leonardo M, Brunty S, Huffman J, Kastigar A, Dickson PE. Intravenous fentanyl self-administration in male and female C57BL/6J and DBA/2J mice. Sci Rep 2023; 13:799. [PMID: 36646781 PMCID: PMC9842734 DOI: 10.1038/s41598-023-27992-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
The genetic mechanisms underlying fentanyl addiction, a highly heritable disease, are unknown. Identifying these mechanisms will lead to better risk assessment, early diagnosis, and improved intervention. To this end, we used intravenous fentanyl self-administration to quantify classical self-administration phenotypes and addiction-like fentanyl seeking in male and female mice from the two founder strains of the BXD recombinant inbred mouse panel (C57BL/6J and DBA/2J). We reached three primary conclusions from these experiments. First, mice from all groups rapidly acquired intravenous fentanyl self-administration and exhibited a dose-response curve, extinction burst, and extinction of the learned self-administration response. Second, fentanyl intake (during acquisition and dose response) and fentanyl seeking (during extinction) were equivalent among groups. Third, strain effects, sex effects, or both were identified for several addiction-like behaviors (cue-induced reinstatement, stress-induced reinstatement, escalation of intravenous fentanyl self-administration). Collectively, these data indicate that C57BL/6J and DBA/2J mice of both sexes were able to acquire, regulate, and extinguish intravenous fentanyl self-administration. Moreover, these data reveal novel strain and sex effects on addiction-like behaviors in the context of intravenous fentanyl self-administration in mice and indicate that the full BXD panel can be used to identify and dissect the genetic mechanisms underlying these effects.
Collapse
Affiliation(s)
- Michael Leonardo
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1700 3rd Ave., Huntington, WV, 25703, USA
| | - Sarah Brunty
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1700 3rd Ave., Huntington, WV, 25703, USA
| | - Jessica Huffman
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1700 3rd Ave., Huntington, WV, 25703, USA
| | - Alexis Kastigar
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1700 3rd Ave., Huntington, WV, 25703, USA
| | - Price E Dickson
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1700 3rd Ave., Huntington, WV, 25703, USA.
| |
Collapse
|
33
|
Kavaliotis E, Boardman JM, Clark JW, Ogeil RP, Verdejo-García A, Drummond SPA. The relationship between sleep and appetitive conditioning: A systematic review and meta-analysis. Neurosci Biobehav Rev 2023; 144:105001. [PMID: 36529310 DOI: 10.1016/j.neubiorev.2022.105001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/24/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
This systematic review and meta-analysis (PROSPERO registration animal/human studies: CRD42021234793/CRD42021234790) examined the relationship between sleep and appetitive conditioning. Inclusion criteria included: a) appetitive conditioning paradigm; b) measure of conditioning; c) sleep measurement and/or sleep loss; d) human and/etor non-human animal samples; and e) written in English. Searches of seven databases returned 3777 publications. The final sample consisted of 42 studies using primarily animal samples and involving food- and drug-related conditioning tasks. We found sleep loss disrupted appetitive conditioning of food rewards (p < 0.001) but potentiated appetitive conditioning of drug rewards (p < 0.001). Furthermore, sleep loss negatively impacted extinction learning irrespective of the reward type. Post-learning sleep was associated with increases in REM sleep (p = 0.02). Findings suggest sleep loss potentiates the impact of psychoactive substances in a manner likely to produce an increased risk of problematic substance use. In obese/overweight populations, sleep loss may be associated with deficits in the conditioning and extinction of reward-related behaviours. Further research should assess the relationship between sleep and appetitive conditioning in humans.
Collapse
Affiliation(s)
- Eleni Kavaliotis
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Victoria 3800, Australia
| | - Johanna M Boardman
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Victoria 3800, Australia
| | - Jacob W Clark
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Victoria 3800, Australia
| | - Rowan P Ogeil
- Eastern Health Clinical School and Monash Addiction Research Centre, Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria 3800, Australia; Turning Point, Eastern Health, Victoria 3121, Australia
| | - Antonio Verdejo-García
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Victoria 3800, Australia
| | - Sean P A Drummond
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Victoria 3800, Australia.
| |
Collapse
|
34
|
Slosky LM, Pires A, Bai Y, Clark NB, Hauser ER, Gross JD, Porkka F, Zhou Y, Chen X, Pogorelov VM, Toth K, Wetsel WC, Barak LS, Caron MG. Establishment of multi-stage intravenous self-administration paradigms in mice. Sci Rep 2022; 12:21422. [PMID: 36503898 PMCID: PMC9742147 DOI: 10.1038/s41598-022-24740-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/18/2022] [Indexed: 12/14/2022] Open
Abstract
Genetically tractable animal models provide needed strategies to resolve the biological basis of drug addiction. Intravenous self-administration (IVSA) is the gold standard for modeling psychostimulant and opioid addiction in animals, but technical limitations have precluded the widespread use of IVSA in mice. Here, we describe IVSA paradigms for mice that capture the multi-stage nature of the disorder and permit predictive modeling. In these paradigms, C57BL/6J mice with long-standing indwelling jugular catheters engaged in cocaine- or remifentanil-associated lever responding that was fixed ratio-dependent, dose-dependent, extinguished by withholding the drug, and reinstated by the presentation of drug-paired cues. The application of multivariate analysis suggested that drug taking in both paradigms was a function of two latent variables we termed incentive motivation and discriminative control. Machine learning revealed that vulnerability to drug seeking and relapse were predicted by a mouse's a priori response to novelty, sensitivity to drug-induced locomotion, and drug-taking behavior. The application of these behavioral and statistical-analysis approaches to genetically-engineered mice will facilitate the identification of neural circuits driving addiction susceptibility and relapse and focused therapeutic development.
Collapse
Affiliation(s)
- Lauren M Slosky
- Department of Cell Biology, Duke University, Durham, NC, USA.
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA.
| | - Andrea Pires
- Department of Cell Biology, Duke University, Durham, NC, USA
| | - Yushi Bai
- Department of Cell Biology, Duke University, Durham, NC, USA
| | | | - Elizabeth R Hauser
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Joshua D Gross
- Department of Cell Biology, Duke University, Durham, NC, USA
| | - Fiona Porkka
- Department of Cell Biology, Duke University, Durham, NC, USA
| | - Yang Zhou
- Department of Cell Biology, Duke University, Durham, NC, USA
| | - Xiaoxiao Chen
- School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Vladimir M Pogorelov
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| | - Krisztian Toth
- Department of Pharmaceutical Sciences, Campbell University, Buies Creek, NC, USA
| | - William C Wetsel
- Department of Cell Biology, Duke University, Durham, NC, USA
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
- Department of Neurobiology, Duke University, Durham, NC, USA
- Mouse Behavioral and Neuroendocrine Analysis Core Facility, Duke University, Durham, NC, USA
| | | | - Marc G Caron
- Department of Cell Biology, Duke University, Durham, NC, USA
- Department of Neurobiology, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
| |
Collapse
|
35
|
Mesa JR, Wesson DW, Schwendt M, Knackstedt LA. The roles of rat medial prefrontal and orbitofrontal cortices in relapse to cocaine-seeking: A comparison across methods for identifying neurocircuits. ADDICTION NEUROSCIENCE 2022; 4:100031. [PMID: 36277334 PMCID: PMC9583858 DOI: 10.1016/j.addicn.2022.100031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
A large body of research supports the notion that regions of the rodent frontal cortex regulate reinstatement of cocaine seeking after cessation of intravenous cocaine self-administration. However, earlier studies identifying the roles of medial (mPFC) and orbital prefrontal cortices (OFC) in reinstatement relied on pharmacological inactivation methods, which indiscriminately inhibited cells within a target region. Here, we first review the anatomical borders and pathways of the rat mPFC and OFC. Next, we compare and contrast findings from more recent cocaine seeking and reinstatement studies that used chemogenetics, optogenetics, or advanced tracing to manipulate specific local cell types or input/output projections of the mPFC and OFC subregions. We found that these studies largely corroborated the roles for mPFC subregions as ascribed by pharmacological inactivation studies. Namely, the prelimbic cortex generally drives cocaine seeking behaviors while the infralimbic cortex is recruited to inhibit cocaine seeking by extinction training but may contribute to seeking after prolonged abstinence. While the OFC remains understudied, we suggest it should not be overlooked, and, as with prelimbic and infralimbic cortices, we identify specific pathways of interest for future studies.
Collapse
Affiliation(s)
- Javier R. Mesa
- Department of Psychology, University of Florida, 114 Psychology, 945 Center Dr., Gainesville, FL 32611, USA,Center for Addiction Research and Education, University of Florida, Gainesville, FL, USA,Corresponding author at: Department of Psychology, University of Florida, 114 Psychology, 945 Center Dr., Gainesville, FL 32611, USA. (J.R. Mesa)
| | - Daniel W. Wesson
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA,Center for Addiction Research and Education, University of Florida, Gainesville, FL, USA
| | - Marek Schwendt
- Department of Psychology, University of Florida, 114 Psychology, 945 Center Dr., Gainesville, FL 32611, USA,Center for Addiction Research and Education, University of Florida, Gainesville, FL, USA
| | - Lori A. Knackstedt
- Department of Psychology, University of Florida, 114 Psychology, 945 Center Dr., Gainesville, FL 32611, USA,Center for Addiction Research and Education, University of Florida, Gainesville, FL, USA
| |
Collapse
|
36
|
Craig AR, Shahan TA. Nondrug reinforcers contingent on alternative behavior or abstinence increase resistance to extinction and reinstatement of ethanol-maintained behavior. J Exp Anal Behav 2022; 118:353-375. [PMID: 36149379 PMCID: PMC9643639 DOI: 10.1002/jeab.801] [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: 01/22/2022] [Revised: 07/28/2022] [Accepted: 09/07/2022] [Indexed: 11/08/2022]
Abstract
The effects of delivering nondrug alternative reinforcement on resistance to extinction and reinstatement of rats' ethanol-maintained lever pressing were evaluated in two experiments. In both, rats self-administered ethanol by lever pressing in a two-component multiple schedule during baseline. In the Rich component, alternative food reinforcement was made available for performing an alternative response (Experiment 1) or according to a differential-reinforcement-of-other-behavior schedule for lever pressing (Experiment 2). In the Lean component, only ethanol was available. Moreover, the frequency of alternative reinforcement was manipulated across conditions in Experiment 1. Following baseline, lever pressing was extinguished in both components by suspending ethanol reinforcement, and alternative food reinforcers were discontinued. Finally, to test for reinstatement, ethanol reinforcers were delivered independently of lever pressing in both components. In both experiments, proportion-of-baseline response rates were higher during extinction and reinstatement testing in the Rich component than in the Lean component (although differentiation was not observed at the lowest frequency of alternative reinforcement in Experiment 1). Thus, alternative nondrug reinforcers increased resistance to extinction and reinstatement of rats' ethanol-maintained lever pressing, even when those reinforcers were delivered contingently on an alternative response or on abstinence from lever pressing.
Collapse
|
37
|
Vengeliene V, Spanagel R. mGlu2 mechanism-based interventions to treat alcohol relapse. Front Pharmacol 2022; 13:985954. [PMID: 36188569 PMCID: PMC9520163 DOI: 10.3389/fphar.2022.985954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Recently we identified a deficiency in metabotropic glutamate receptor 2 (mGlu2) function in the corticoaccumbal pathway, as a common pathological mechanism underlying alcohol-seeking and relapse behavior. Based on this mechanism, we hypothesized that mGlu2/3 agonists and mGlu2 positive allosteric modulators (PAMs) may be effective in reducing relapse-like behavior. Two mGlu2/3 agonists, LY379268 and LY354740 (a structural analog of LY379268 six-fold more potent in activating mGlu2 over mGluR3), were tested in a well-established rat model of relapse, the alcohol deprivation effect (ADE) with repeated deprivation phases. Since these agonists do not readily discriminate between contributions of mGlu2 and mGluR3, we also tested LY487379, a highly specific PAM that potentiates the effect of glutamate on the mGlu2 with less specificity on other mGlu receptor subtypes. Both LY379268 and LY354740 significantly and dose-dependently reduced the expression of the ADE. No significant changes in water intake, body weight and locomotor activity were observed. Importantly, repeated administration of mGlu2/3 agonist did not lead to tolerance development. mGlu2 PAM LY487379 treatment significantly reduced expression of the ADE in both male and female rats. Combination treatment of mGlu2/3 agonist and PAM had similar effect on relapse-like drinking to that seen in mGlu2/3 agonist treatment alone. Together with other preclinical data showing that PAMs can reduce alcohol-seeking behavior we conclude that mGlu2 PAMs should be considered for clinical trials in alcohol-dependent patients.
Collapse
Affiliation(s)
- Valentina Vengeliene
- Institute of Psychopharmacology, Central Institute of Mental Health, Faculty of Medicine Mannheim, University of Heidelberg, Heidelberg, Germany
- Department of Neurobiology and Biophysics, Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Faculty of Medicine Mannheim, University of Heidelberg, Heidelberg, Germany
- *Correspondence: Rainer Spanagel,
| |
Collapse
|
38
|
Webb SM, Sacramento AD, McCloskey MA, Wroten MG, Ploense KL, Kippin TE, Ben-Shahar O, Szumlinski KK. The incubation of cocaine craving is dissociated from changes in glial cell markers within prefrontal cortex and nucleus accumbens of rats. ADDICTION NEUROSCIENCE 2022; 3:100030. [PMID: 36034166 PMCID: PMC9410194 DOI: 10.1016/j.addicn.2022.100030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Affiliation(s)
- Sierra M. Webb
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA 93106-9660, USA
| | - Arianne D. Sacramento
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA 93106-9660, USA
| | - Megan A. McCloskey
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA 93106-9660, USA
| | - Melissa G. Wroten
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA 93106-9660, USA
| | - Kyle L. Ploense
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA 93106-9660, USA
| | - Tod E. Kippin
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA 93106-9660, USA
- Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106-9625, USA
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA 93106-9625, USA
| | - Osnat Ben-Shahar
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA 93106-9660, USA
| | - Karen K. Szumlinski
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA 93106-9660, USA
- Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106-9625, USA
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA 93106-9625, USA
| |
Collapse
|
39
|
Farrell MR, Ye Q, Xie Y, Esteban JSD, Mahler SV. Ventral pallidum GABA neurons bidirectionally control opioid relapse across rat behavioral models. ADDICTION NEUROSCIENCE 2022; 3:100026. [PMID: 36156918 PMCID: PMC9494709 DOI: 10.1016/j.addicn.2022.100026] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Opioid addiction is a chronic, relapsing disorder. Whether addicted individuals are forced to abstain or they decide themselves to quit using drugs, relapse rates are high-especially upon encountering contexts and stimuli associated with prior opioid use. Rodents similarly show context- and cue-induced reinstatement of drug seeking following abstinence, and intriguingly, the neural circuits underlying these relapse-like behaviors differ when abstinence is involuntarily imposed, responding is extinguished, or animals decide themselves to cease taking drug. Here, we employ two complementary rat behavioral models of relapse-like behavior for the highly reinforcing opioid drug remifentanil, and asked whether GABAergic neurons in the ventral pallidum (VPGABA) control opioid seeking under these behavioral conditions. Specifically, we asked how chemogenetically stimulating VPGABA neurons with clozapine-N-oxide (CNO) influences the ability of contextual or discrete remifentanil-paired cues to reinstate drug seeking following either voluntary abstinence (punishment-induced; GroupPunish), or extinction training (GroupExt). In GroupPunish rats, we also chemogenetically inhibited VPGABA neurons, and examined spontaneous VP activity (Fos) during cued reinstatement. In both GroupPunish and GroupExt rats, stimulating Gq-signaling in VPGABA neurons augmented remifentanil reinstatement in a cue- and context-dependent manner. Conversely, engaging inhibitory Gi-signaling in VPGABA neurons in GroupPunish suppressed cue-induced reinstatement, and cue-triggered seeking was correlated with Fos expression in rostral, but not caudal VP. Neither stimulating nor inhibiting VPGABA neurons influenced unpunished remifentanil self-administration. We conclude that VPGABA neurons bidirectionally control opioid seeking regardless of the specific relapse model employed, highlighting their fundamental role in opioid relapse-like behavior across behavioral models, and potentially across species.
Collapse
Affiliation(s)
- Mitchell R. Farrell
- University of California, Irvine Department of Neurobiology and Behavior, 1203 McGaugh Hall Irvine, CA, 92697, USA
| | - Qiying Ye
- University of California, Irvine Department of Neurobiology and Behavior, 1203 McGaugh Hall Irvine, CA, 92697, USA
| | - Yiyan Xie
- University of California, Irvine Department of Neurobiology and Behavior, 1203 McGaugh Hall Irvine, CA, 92697, USA
| | - Jeanine Sandra D. Esteban
- University of California, Irvine Department of Neurobiology and Behavior, 1203 McGaugh Hall Irvine, CA, 92697, USA
| | - Stephen V. Mahler
- University of California, Irvine Department of Neurobiology and Behavior, 1203 McGaugh Hall Irvine, CA, 92697, USA
| |
Collapse
|
40
|
Visser E, Matos MR, Mitrić MM, Kramvis I, van der Loo RJ, Mansvelder HD, Smit AB, van den Oever MC. Extinction of Cocaine Memory Depends on a Feed-Forward Inhibition Circuit Within the Medial Prefrontal Cortex. Biol Psychiatry 2022; 91:1029-1038. [PMID: 34715992 DOI: 10.1016/j.biopsych.2021.08.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 08/04/2021] [Accepted: 08/04/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Cocaine-associated environments (i.e., contexts) evoke persistent memories of cocaine reward and thereby contribute to the maintenance of addictive behavior in cocaine users. From a therapeutic perspective, enhancing inhibitory control over cocaine-conditioned responses is of pivotal importance but requires a more detailed understanding of the neural circuitry that can suppress context-evoked cocaine memories, e.g., through extinction learning. The ventral medial prefrontal cortex (vmPFC) and dorsal medial prefrontal cortex (dmPFC) are thought to bidirectionally regulate responding to cocaine cues through their projections to other brain regions. However, whether these mPFC subregions interact to enable adaptive responding to cocaine-associated contextual stimuli has remained elusive. METHODS We used antero- and retrograde tracing combined with chemogenetic intervention to examine the role of vmPFC-to-dmPFC projections in extinction of cocaine-induced place preference in mice. In addition, electrophysiological recordings and optogenetics were used to determine whether parvalbumin-expressing inhibitory interneurons and pyramidal neurons in the dmPFC are innervated by vmPFC projections. RESULTS We found that vmPFC-to-dmPFC projecting neurons are activated during unreinforced re-exposure to a cocaine-associated context, and selective suppression of these cells impairs extinction learning. Parvalbumin-expressing inhibitory interneurons in the dmPFC receive stronger monosynaptic excitatory input from vmPFC projections than local dmPFC pyramidal neurons, consequently resulting in disynaptic inhibition of pyramidal neurons. In line with this, we show that chemogenetic suppression of dmPFC parvalbumin-expressing inhibitory interneurons impairs extinction learning. CONCLUSIONS Our data reveal that vmPFC projections mediate extinction of a cocaine-associated contextual memory through recruitment of feed-forward inhibition in the dmPFC, thereby providing a novel neuronal substrate that promotes extinction-induced inhibitory control.
Collapse
Affiliation(s)
- Esther Visser
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Mariana R Matos
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Miodrag M Mitrić
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Ioannis Kramvis
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Rolinka J van der Loo
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Huibert D Mansvelder
- Department of Integrated Neurophysiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Michel C van den Oever
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
41
|
Strickland JC, Stoops WW, Banks ML, Gipson CD. Logical fallacies and misinterpretations that hinder progress in translational addiction neuroscience. J Exp Anal Behav 2022; 117:384-403. [PMID: 35362559 PMCID: PMC9090969 DOI: 10.1002/jeab.757] [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: 09/23/2021] [Revised: 03/08/2022] [Accepted: 03/11/2022] [Indexed: 11/11/2022]
Abstract
Substance use disorders (SUDs) are heterogeneous and complex, making the development of translationally predictive rodent and nonhuman primate models to uncover their neurobehavioral underpinnings difficult. Neuroscience-focused outcomes have become highly prevalent, and with this, the notion that SUDs are disorders of the brain embraced as a dominant theoretical orientation to understand SUD etiology and treatment. These efforts, however, have led to few efficacious pharmacotherapies, and in some cases (as with cocaine or methamphetamine), no pharmacotherapies have translated from preclinical models for clinical use. In this theoretical commentary, we first describe the development of animal models of substance use behaviors from a historical perspective. We then define and discuss three logical fallacies including 1) circular explanation, 2) affirming the consequent, and 3) reification that can apply to developed models. We then provide three case examples in which conceptual or logical issues exist in common methods (i.e., behavioral economic demand, escalation, and reinstatement). Alternative strategies to refocus behavioral models are suggested for the field to better bridge the translational divide between animal models, the clinical condition of SUDs, and current and future regulatory pathways for intervention development.
Collapse
Affiliation(s)
- Justin C. Strickland
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine
| | | | - Matthew L. Banks
- Department of Pharmacology and Toxicology, Virginia Commonwealth University
| | - Cassandra D. Gipson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington
| |
Collapse
|
42
|
Nall RW, Chalhoub RM, Kalivas PW. Drug versus non-drug behaviors: A dual-reward model of sex differences and neurobiological mechanisms in rats. J Exp Anal Behav 2022; 117:457-471. [PMID: 35297047 PMCID: PMC10775707 DOI: 10.1002/jeab.752] [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: 10/15/2021] [Revised: 02/02/2022] [Accepted: 02/09/2022] [Indexed: 11/05/2022]
Abstract
Substance Use Disorders (SUDs) are an impactful problem characterized by chronic relapse and engagement in drug-related behaviors at the expense of non-drug behaviors. Brain regions implicated in drug and non-drug-related behaviors often overlap, complicating investigations of neurobiological mechanisms underlying SUDs. Here we presented a within-subject model for studying self-administration, reinforcer competition, extinction, and cued reinstatement of cocaine- and food-seeking in rats. Due to differences in cocaine- and food-reinforced behavior, we transformed data to proportions of baseline, revealing increased resistance to extinction and disproportionately greater cued reinstatement of cocaine seeking relative to food seeking. Consistent with previous reports, females showed greater preference for cocaine reinforcement than males, though these findings failed to reach statistical significance. To demonstrate the model's utility for investigating neurobiological mechanisms, we included proof-of-concept calcium imaging data demonstrating the utility of the behavioral model for detecting cellular activity patterns associated with cocaine- and food-seeking behaviors. Future studies utilizing this model should improve understanding of the development and expression of pathological behaviors characteristic of SUDs in humans, sex differences in these behaviors, and their neurobiological correlates. Thus, the model has utility for improving understanding of SUDs, leading to novel treatments to reduce the pathological behaviors associated with SUDs.
Collapse
Affiliation(s)
- Rusty W. Nall
- Medical University of South Carolina
- Jacksonville State University
| | | | | |
Collapse
|
43
|
Wu R, Liu J, Vu J, Huang Y, Dietz DM, Li JX. Interleukin-1 receptor-associated kinase 4 (IRAK4) in the nucleus accumbens regulates opioid-seeking behavior in male rats. Brain Behav Immun 2022; 101:37-48. [PMID: 34958862 PMCID: PMC8885906 DOI: 10.1016/j.bbi.2021.12.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 11/09/2021] [Accepted: 12/18/2021] [Indexed: 12/12/2022] Open
Abstract
Opioid addiction remains a severe health problem. While substantial insights underlying opioid addiction have been yielded from neuron-centric studies, the contribution of non-neuronal mechanisms to opioid-related behavioral adaptations has begun to be recognized. Toll-like receptor 4 (TLR4), a pattern recognition receptor, has been widely suggested in opioid-related behaviors. Interleukin-1 receptor-associated kinase 4 (IRAK4) is a kinase essential for TLR4 responses, However, the potential role of IRAK4 in opioid-related responses has not been examined. Here, we explored the role of IRAK4 in cue-induced opioid-seeking behavior in male rats. We found that morphine self-administration increased the phosphorylation level of IRAK4 in the nucleus accumbens (NAc) in rats; the IRAK4 signaling remained activated after morphine extinction and cue-induced reinstatement test. Both systemic and local inhibition of IRAK4 in the NAc core attenuated cue-induced morphine-seeking behavior without affecting the locomotor activity and cue-induced sucrose-seeking. In addition, inhibition of IRAK4 also reduced the cue-induced reinstatement of fentanyl-seeking. Our findings suggest an important role of IRAK4 in opioid relapse-like behaviors and provide novel evidence in the association between innate immunity and drug addiction.
Collapse
Affiliation(s)
- Ruyan Wu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY,Medical College of Yangzhou University, Yangzhou, China
| | - Jianfeng Liu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY
| | - Jimmy Vu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY
| | - Yufei Huang
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY
| | - David M. Dietz
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States.
| |
Collapse
|
44
|
Peart DR, Andrade AK, Logan CN, Knackstedt LA, Murray JE. Regulation of Cocaine-related Behaviors by Estrogen and Progesterone. Neurosci Biobehav Rev 2022; 135:104584. [DOI: 10.1016/j.neubiorev.2022.104584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/30/2022] [Accepted: 02/12/2022] [Indexed: 10/19/2022]
|
45
|
Transcranial direct current stimulation (tDCS) reduces motivation to drink ethanol and reacquisition of ethanol self-administration in female mice. Sci Rep 2022; 12:198. [PMID: 34997004 PMCID: PMC8741977 DOI: 10.1038/s41598-021-03940-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 11/24/2021] [Indexed: 12/02/2022] Open
Abstract
Transcranial direct current stimulation (tDCS) is an emerging noninvasive brain neuromodulation technique aimed at relieving symptoms associated with psychiatric disorders, including addiction. The goal of the present study was to better identify which phase of alcohol-related behavior (hedonic effect, behavioral sensitization, self-administration, or motivation to obtain the drug) might be modulated by repeated anodal tDCS over the frontal cortex (0.2 mA, 20 min, twice a day for 5 consecutive days), using female mice as a model. Our data showed that tDCS did not modulate the hedonic effects of ethanol as assessed by a conditioned place preference test (CPP) or the expression of ethanol-induced behavioral sensitization. Interestingly, tDCS robustly reduced reacquisition of ethanol consumption (50% decrease) following extinction of self-administration in an operant paradigm. Furthermore, tDCS significantly decreased motivation to drink ethanol on a progressive ratio schedule (30% decrease). Taken together, our results show a dissociation between the effects of tDCS on “liking” (hedonic aspect; no effect in the CPP) and “wanting” (motivation; decreased consumption on a progressive ratio schedule). Our tDCS procedure in rodents will allow us to better understand its mechanisms of action in order to accelerate its use as a complementary and innovative tool to help alcohol-dependent patients maintain abstinence or reduce ethanol intake.
Collapse
|
46
|
Nicolas C, Zlebnik NE, Farokhnia M, Leggio L, Ikemoto S, Shaham Y. Sex Differences in Opioid and Psychostimulant Craving and Relapse: A Critical Review. Pharmacol Rev 2022; 74:119-140. [PMID: 34987089 PMCID: PMC11060335 DOI: 10.1124/pharmrev.121.000367] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 08/15/2021] [Indexed: 01/11/2023] Open
Abstract
A widely held dogma in the preclinical addiction field is that females are more vulnerable than males to drug craving and relapse. Here, we first review clinical studies on sex differences in psychostimulant and opioid craving and relapse. Next, we review preclinical studies on sex differences in psychostimulant and opioid reinstatement of drug seeking after extinction of drug self-administration, and incubation of drug craving (time-dependent increase in drug seeking during abstinence). We also discuss ovarian hormones' role in relapse and craving in humans and animal models and speculate on brain mechanisms underlying their role in cocaine craving and relapse in rodent models. Finally, we discuss imaging studies on brain responses to cocaine cues and stress in men and women.The results of the clinical studies reviewed do not appear to support the notion that women are more vulnerable to psychostimulant and opioid craving and relapse. However, this conclusion is tentative because most of the studies reviewed were correlational, not sufficiently powered, and not a priori designed to detect sex differences. Additionally, imaging studies suggest sex differences in brain responses to cocaine cues and stress. The results of the preclinical studies reviewed provide evidence for sex differences in stress-induced reinstatement and incubation of cocaine craving but not cue- or cocaine-induced reinstatement of cocaine seeking. These sex differences are modulated in part by ovarian hormones. In contrast, the available data do not support the notion of sex differences in craving and relapse/reinstatement for methamphetamine or opioids in rodent models. SIGNIFICANCE STATEMENT: This systematic review summarizes clinical and preclinical studies on sex differences in psychostimulant and opioid craving and relapse. Results of the clinical studies reviewed do not appear to support the notion that women are more vulnerable to psychostimulant and opioid craving and relapse. Results of preclinical studies reviewed provide evidence for sex differences in reinstatement and incubation of cocaine seeking but not for reinstatement or incubation of methamphetamine or opioid seeking.
Collapse
Affiliation(s)
- Céline Nicolas
- Neurocentre Magendie, University of Bordeaux, Bordeaux, France (C.N.); Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, Present address: Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, CA (N.E.Z.); Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD (M.F., L.L., S.I., Y.S.); and Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (M.F., L.L.)
| | - Natalie E Zlebnik
- Neurocentre Magendie, University of Bordeaux, Bordeaux, France (C.N.); Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, Present address: Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, CA (N.E.Z.); Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD (M.F., L.L., S.I., Y.S.); and Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (M.F., L.L.)
| | - Mehdi Farokhnia
- Neurocentre Magendie, University of Bordeaux, Bordeaux, France (C.N.); Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, Present address: Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, CA (N.E.Z.); Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD (M.F., L.L., S.I., Y.S.); and Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (M.F., L.L.)
| | - Lorenzo Leggio
- Neurocentre Magendie, University of Bordeaux, Bordeaux, France (C.N.); Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, Present address: Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, CA (N.E.Z.); Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD (M.F., L.L., S.I., Y.S.); and Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (M.F., L.L.)
| | - Satoshi Ikemoto
- Neurocentre Magendie, University of Bordeaux, Bordeaux, France (C.N.); Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, Present address: Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, CA (N.E.Z.); Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD (M.F., L.L., S.I., Y.S.); and Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (M.F., L.L.)
| | - Yavin Shaham
- Neurocentre Magendie, University of Bordeaux, Bordeaux, France (C.N.); Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, Present address: Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, CA (N.E.Z.); Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD (M.F., L.L., S.I., Y.S.); and Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (M.F., L.L.)
| |
Collapse
|
47
|
Rabin RA, Mackey S, Parvaz MA, Cousijn J, Li C, Pearlson G, Schmaal L, Sinha R, Stein E, Veltman D, Thompson PM, Conrod P, Garavan H, Alia‐Klein N, Goldstein RZ. Common and gender-specific associations with cocaine use on gray matter volume: Data from the ENIGMA addiction working group. Hum Brain Mapp 2022; 43:543-554. [PMID: 32857473 PMCID: PMC8675419 DOI: 10.1002/hbm.25141] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 06/26/2020] [Accepted: 07/10/2020] [Indexed: 11/30/2022] Open
Abstract
Gray matter volume (GMV) in frontal cortical and limbic regions is susceptible to cocaine-associated reductions in cocaine-dependent individuals (CD) and is negatively associated with duration of cocaine use. Gender differences in CD individuals have been reported clinically and in the context of neural responses to cue-induced craving and stress reactivity. The variability of GMV in select brain areas between men and women (e.g., limbic regions) underscores the importance of exploring interaction effects between gender and cocaine dependence on brain structure. Therefore, voxel-based morphometry data derived from the ENIGMA Addiction Consortium were used to investigate potential gender differences in GMV in CD individuals compared to matched controls (CTL). T1-weighted MRI scans and clinical data were pooled from seven sites yielding 420 gender- and age-matched participants: CD men (CDM, n = 140); CD women (CDW, n = 70); control men (CTLM, n = 140); and control women (CTLW, n = 70). Differences in GMV were assessed using a 2 × 2 ANCOVA, and voxelwise whole-brain linear regressions were conducted to explore relationships between GMV and duration of cocaine use. All analyses were corrected for age, total intracranial volume, and site. Diagnostic differences were predominantly found in frontal regions (CD < CTL). Interestingly, gender × diagnosis interactions in the left anterior insula and left lingual gyrus were also documented, driven by differences in women (CDW < CTLW). Further, lower right hippocampal GMV was associated with greater cocaine duration in CDM. Given the importance of the anterior insula to interoception and the hippocampus to learning contextual associations, results may point to gender-specific mechanisms in cocaine addiction.
Collapse
Affiliation(s)
- Rachel A. Rabin
- Department of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Scott Mackey
- Departments of Psychiatry and PsychologyUniversity of VermontBurlingtonVermontUSA
| | - Muhammad A. Parvaz
- Department of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Janna Cousijn
- Department of PsychologyUniversity of AmsterdamAmsterdamThe Netherlands
| | - Chiang‐shan Li
- Department of PsychiatryYale University School of MedicineNew HavenConnecticutUSA
| | - Godfrey Pearlson
- Department of PsychiatryYale University School of MedicineNew HavenConnecticutUSA
| | - Lianne Schmaal
- Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, Australia and Centre for Youth Mental HealthThe University of MelbourneMelbourneAustralia
| | - Rajita Sinha
- Department of PsychiatryYale University School of MedicineNew HavenConnecticutUSA
| | - Elliot Stein
- Intramural Research Program—Neuroimaging Research BranchNational Institute on Drug AbuseBaltimoreMarylandUSA
| | - Dick Veltman
- Department of PsychiatryVU University Medical CenterAmsterdamThe Netherlands
| | - Paul M. Thompson
- Imaging Genetics Center, Department of Neurology Keck School of MedicineUniversity of Southern CaliforniaMarina del ReyCaliforniaUSA
| | - Patricia Conrod
- Department of PsychiatryUniversité de Montreal, CHU Ste Justine HospitalMontrealQuebecCanada
| | - Hugh Garavan
- Departments of Psychiatry and PsychologyUniversity of VermontBurlingtonVermontUSA
| | - Nelly Alia‐Klein
- Department of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Rita Z. Goldstein
- Department of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| |
Collapse
|
48
|
Gómez-Bujedo J, Lorca-Marín JA, Pérez-Moreno PJ, Díaz Batanero C, Fernández-Calderón F, Moraleda-Barreno E. Changes in Drug-Related Implicit Associations during Substance Use Disorder Treatment: The Role of the Therapeutic Context. Subst Use Misuse 2022; 57:185-192. [PMID: 34738506 DOI: 10.1080/10826084.2021.1995755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Implicit cognition has been linked to relapse in substance use disorder (SUD). Studies on attentional bias have found different outcomes related to the therapeutic context, finding an association with relapse in inpatients but not in outpatients. There are no similar studies that use associations in semantic memory as a measure of implicit cognition. OBJECTIVES (i) to analyze the relationship between a measure of associations in semantic memory and relapse in inpatients and outpatients; (ii) to compare the evolution of these associations between inpatients and outpatients after 3 months of treatment. METHODS Eighty nine outpatients and 94 inpatients with SUD for cocaine and alcohol participated in this study. We employed a longitudinal design with a baseline evaluation and follow-up after three months, using the Word Association Task for Drug Use Disorder (WAT-DUD). RESULTS The choice of drug-related words predicted relapse in cocaine (odds ratio = 1.97, z = 2.01, p = .045) and alcohol-cocaine (odds ratio = 2.39, z = 2.55, p = .011) use. Follow-up at 3 months revealed a reduction in the choice of drug-related words in inpatients (Z = 2.031, p = .042). CONCLUSIONS A greater choice of drug-associated words in the presence of ambiguous images was related to relapse in inpatients but not in outpatients. The inpatients group showed a reduction in the semantic association with drugs during the first three months of treatment.
Collapse
Affiliation(s)
- Jesús Gómez-Bujedo
- Department of Clinical and Experimental Psychology, Facultad de Educación, Psicología y Ciencias del Deporte, University of Huelva, Huelva, Spain, EU
- Research Center for Natural Resources, Health and the Environment, University of Huelva, Spain
| | - José Andrés Lorca-Marín
- Department of Clinical and Experimental Psychology, Facultad de Educación, Psicología y Ciencias del Deporte, University of Huelva, Huelva, Spain, EU
- Research Center for Natural Resources, Health and the Environment, University of Huelva, Spain
| | - Pedro Juan Pérez-Moreno
- Department of Clinical and Experimental Psychology, Facultad de Educación, Psicología y Ciencias del Deporte, University of Huelva, Huelva, Spain, EU
- Research Center for Natural Resources, Health and the Environment, University of Huelva, Spain
| | - Carmen Díaz Batanero
- Department of Clinical and Experimental Psychology, Facultad de Educación, Psicología y Ciencias del Deporte, University of Huelva, Huelva, Spain, EU
- Research Center for Natural Resources, Health and the Environment, University of Huelva, Spain
| | - Fermín Fernández-Calderón
- Department of Clinical and Experimental Psychology, Facultad de Educación, Psicología y Ciencias del Deporte, University of Huelva, Huelva, Spain, EU
- Research Center for Natural Resources, Health and the Environment, University of Huelva, Spain
| | - Enrique Moraleda-Barreno
- Department of Clinical and Experimental Psychology, Facultad de Educación, Psicología y Ciencias del Deporte, University of Huelva, Huelva, Spain, EU
- Research Center for Natural Resources, Health and the Environment, University of Huelva, Spain
| |
Collapse
|
49
|
Singh PK, Lutfy K. The Role of Beta-Endorphin in Cocaine-Induced Conditioned Place Preference, Its Extinction, and Reinstatement in Male and Female Mice. Front Behav Neurosci 2021; 15:763336. [PMID: 34955777 PMCID: PMC8702804 DOI: 10.3389/fnbeh.2021.763336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/16/2021] [Indexed: 11/21/2022] Open
Abstract
Endogenous opioids have been implicated in cocaine reward. However, the role of each opioid peptide in this regard is unknown. Notably, the role of each peptide in extinction and reinstatement is not fully characterized. Thus, we assessed whether cocaine-induced conditioned place preference (CPP) and its extinction and reinstatement would be altered in the absence of beta-endorphin. We also examined if sex-related differences would exist in these processes. Male and female mice lacking beta-endorphin and their respective controls were tested for baseline place preference on day 1. On day 2, mice were treated with saline/cocaine (15 mg/kg) and confined to the vehicle- or drug-paired chamber for 30 min, respectively. In the afternoon, mice were treated with the alternate treatment and confined to the opposite chamber. Mice were then tested for CPP on day 3. Mice then received additional conditioning on this day as well as on day 4. Mice were then tested for CPP on day 5. Mice then received extinction training on day 9. On day 10, mice were tested for extinction and then reinstatement of CPP following a priming dose of cocaine (7.5 mg/kg). Male and female mice lacking beta-endorphin did not exhibit CPP following single conditioning with cocaine. On the other hand, only male mice lacking beta-endorphin failed to show CPP after repeated conditioning. Nonetheless, reinstatement of CPP was blunted in both male and female mice lacking beta-endorphin compared to controls. The present results suggest that beta-endorphin plays a functional role in cocaine-induced CPP and its reinstatement, and sex-related differences exist in the regulatory action of beta-endorphin on the acquisition but not reinstatement of cocaine CPP.
Collapse
Affiliation(s)
- Prableen K Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| | - Kabirullah Lutfy
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| |
Collapse
|
50
|
Gong S, Fayette N, Heinsbroek JA, Ford CP. Cocaine shifts dopamine D2 receptor sensitivity to gate conditioned behaviors. Neuron 2021; 109:3421-3435.e5. [PMID: 34506723 PMCID: PMC8571051 DOI: 10.1016/j.neuron.2021.08.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 07/16/2021] [Accepted: 08/11/2021] [Indexed: 12/21/2022]
Abstract
Cocaine addiction is a chronic, relapsing disorder characterized by maladaptation in the brain mesolimbic and nigrostriatal dopamine system. Although changes in the properties of D2-receptor-expressing medium spiny neurons (D2-MSNs) and connected striatal circuits following cocaine treatment are known, the contributions of altered D2-receptor (D2R) function in mediating the rewarding properties of cocaine remain unclear. Here, we describe how a 7-day exposure to cocaine alters dopamine signaling by selectively reducing the sensitivity, but not the expression, of nucleus accumbens D2-MSN D2Rs via an alteration in the relative expression and coupling of G protein subunits. This cocaine-induced reduction of D2R sensitivity facilitated the development of the rewarding effects of cocaine as blocking the reduction in G protein expression was sufficient to prevent cocaine-induced behavioral adaptations. These findings identify an initial maladaptive change in sensitivity by which mesolimbic dopamine signals are encoded by D2Rs following cocaine exposure.
Collapse
Affiliation(s)
- Sheng Gong
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Nicholas Fayette
- Department of Anesthesiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jasper A Heinsbroek
- Department of Anesthesiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Christopher P Ford
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|