1
|
Lloret-Torres ME, Barreto-Estrada JL. LF-DBS of the Ventral Striatum Shortens Persistence for Morphine Place Preference and Modulates BDNF Expression in the Hippocampus. Behav Brain Res 2024; 477:115300. [PMID: 39490421 DOI: 10.1016/j.bbr.2024.115300] [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: 08/02/2024] [Revised: 09/30/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Deep brain stimulation (DBS) of the ventral capsule/ventral striatum (VC/VS) represents a promising therapy for treatment-refractory patients with substance-use disorders. We previously found that low-frequency (LF) DBS aimed to the VC/VS during extinction training strengthens the extinction memory for morphine seeking under a partial extinction protocol. OBJECTIVES/HYPOTHESIS In this study, animals were tested in a full extinction protocol to determine whether LF-DBS applied during extinction facilitates extinction while preventing drug reinstatement, and study the molecular mechanisms underlying the effects of LF-DBS, METHODS/RESULTS: We used a full extinction CPP paradigm combined with LF-DBS to assess behavior. Western blots for the pro-extinction molecule, brain-derived neurotrophic factor (BDNF) were then performed in corticomesolimbic regions of the brain. Lastly, to determine whether changes in BDNF expression elicited by LF-DBS were specific to the VS/NAc afferents from the hippocampus, amygdala, and medial prefrontal cortex, we performed BDNF-like immunohistochemistry, combined with the retrograde tracer cholera toxin B (CtB). RESULTS We showed a significant reduction in the number of days required to fully extinguish morphine CPP in animals exposed to LF-DBS during extinction training accompanied by a significant increase in BDNF expression in the hippocampus. However, LF-DBS applied during extinction did not prevent drug reinstatement. Lastly, no changes in BDNF/CtB double-labeled cells were found in VS/NAc projecting cells after one-day exposure to LF-DBS. CONCLUSION(S) These data suggest that LF-DBS can facilitate extinction of morphine CPP by decreasing drug seeking through potential synaptic plasticity changes in the hippocampus to strengthen extinction memories.
Collapse
Affiliation(s)
- Mario E Lloret-Torres
- Department of Anatomy and Neurobiology, School of Medicine, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico, 00936
| | - Jennifer L Barreto-Estrada
- Department of Anatomy and Neurobiology, School of Medicine, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico, 00936.
| |
Collapse
|
2
|
Huang S, Liu X, Li Z, Si Y, Yang L, Deng J, Luo Y, Xue YX, Lu L. Memory Reconsolidation Updating in Substance Addiction: Applications, Mechanisms, and Future Prospects for Clinical Therapeutics. Neurosci Bull 2024:10.1007/s12264-024-01294-z. [PMID: 39264570 DOI: 10.1007/s12264-024-01294-z] [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: 01/06/2024] [Accepted: 05/09/2024] [Indexed: 09/13/2024] Open
Abstract
Persistent and maladaptive drug-related memories represent a key component in drug addiction. Converging evidence from both preclinical and clinical studies has demonstrated the potential efficacy of the memory reconsolidation updating procedure (MRUP), a non-pharmacological strategy intertwining two distinct memory processes: reconsolidation and extinction-alternatively termed "the memory retrieval-extinction procedure". This procedure presents a promising approach to attenuate, if not erase, entrenched drug memories and prevent relapse. The present review delineates the applications, molecular underpinnings, and operational boundaries of MRUP in the context of various forms of substance dependence. Furthermore, we critically examine the methodological limitations of MRUP, postulating potential refinement to optimize its therapeutic efficacy. In addition, we also look at the potential integration of MRUP and neurostimulation treatments in the domain of substance addiction. Overall, existing studies underscore the significant potential of MRUP, suggesting that interventions predicated on it could herald a promising avenue to enhance clinical outcomes in substance addiction therapy.
Collapse
Affiliation(s)
- Shihao Huang
- Department of Neurobiology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Xiaoxing Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, 100191, China
| | - Zhonghao Li
- Department of Neurobiology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Yue Si
- Department of Neurobiology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Liping Yang
- Department of Neurobiology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Jiahui Deng
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, 100191, China
| | - Yixiao Luo
- Department of Anesthesiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Yan-Xue Xue
- Department of Neurobiology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, 100191, China.
- Chinese Institute for Brain Research, Beijing, 102206, China.
| | - Lin Lu
- Department of Neurobiology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, 100191, China.
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, 100191, China.
- Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.
| |
Collapse
|
3
|
Xie B, Wang Y, Lu Y, Wang M, Hui R, Yu H, Li W, Zhang L, Yu F, Ni Z, Cong B, Ma C, Wen D. A novel intervention of molecular hydrogen on the unbalance of the gut microbiome in opioid addiction: Experimental and human studies. Biomed Pharmacother 2024; 178:117273. [PMID: 39116782 DOI: 10.1016/j.biopha.2024.117273] [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: 04/11/2024] [Revised: 07/04/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024] Open
Abstract
The gut-brain axis mediates the interaction pathway between microbiota and opioid addiction. In recent years, many studies have shown that molecular hydrogen has therapeutic and preventive effects on various diseases. This study aimed to investigate whether molecular hydrogen could serve as pharmacological intervention agent to reduce risks of reinstatement of opioid seeking and explore the mechanism of gut microbiota base on animal experiments and human studies. Morphine-induced conditioned place preference (CPP) was constructed to establish acquisition, extinction, and reinstatement stage, and the potential impact of H2 on the behaviors related to morphine-induced drug extinction was determined using both free accessible and confined CPP extinction paradigms. The effects of morphine on microbial diversity and composition of microbiota, as well as the subsequent changes after H2 intervention, were assessed using 16 S rRNA gene sequencing. Short-Chain Fatty Acids (SCFAs) in mice serum were detected by gas chromatography-mass spectrometry (GC-MS). Meanwhile, we also conducted molecular hydrogen intervention and gut microbiota testing in opioid-addicted individuals. Our results revealed that molecular hydrogen could enhance the extinction of morphine-related behavior, reducing morphine reinstatement. Gut microbes may be a potential mechanism behind the therapeutic effects of molecular hydrogen on morphine addiction. Additionally, molecular hydrogen improved symptoms of depression and anxiety, as well as gut microbial features, in individuals with opioid addiction. This study supports molecular hydrogen as a novel and effective intervention for morphine-induced addiction and reveals the mechanism of gut microbiota.
Collapse
Affiliation(s)
- Bing Xie
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China
| | - Yong Wang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China
| | - Yun Lu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China
| | - Mengmeng Wang
- Affiliated Hospital of Hebei University, College of Clinical Medicine, Hebei University, Collaborative Innovation Center of Tumor Microecological Metabolism Regulation, Baoding, Hebei Province 071000, PR China
| | - Rongji Hui
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China
| | - Hailei Yu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China
| | - Wenbo Li
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China
| | - Ludi Zhang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, Hebei Province 050017, PR China
| | - Feng Yu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China
| | - Zhiyu Ni
- Affiliated Hospital of Hebei University, College of Clinical Medicine, Hebei University, Collaborative Innovation Center of Tumor Microecological Metabolism Regulation, Baoding, Hebei Province 071000, PR China; Clinical Medical College, Hebei University of Engineering, Handan, Hebei Province 056038, PR China
| | - Bin Cong
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China
| | - Chunling Ma
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China.
| | - Di Wen
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China.
| |
Collapse
|
4
|
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
|
5
|
Socha J, Grochecki P, Marszalek-Grabska M, Skrok A, Smaga I, Slowik T, Prazmo W, Kotlinski R, Filip M, Kotlinska JH. Cannabidiol Protects against the Reinstatement of Oxycodone-Induced Conditioned Place Preference in Adolescent Male but Not Female Rats: The Role of MOR and CB1R. Int J Mol Sci 2024; 25:6651. [PMID: 38928357 PMCID: PMC11204276 DOI: 10.3390/ijms25126651] [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: 04/11/2024] [Revised: 06/04/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Cannabidiol (CBD), a phytocannabinoid, appeared to satisfy several criteria for a safe approach to preventing drug-taking behavior, including opioids. However, most successful preclinical and clinical results come from studies in adult males. We examined whether systemic injections of CBD (10 mg/kg, i.p.) during extinction of oxycodone (OXY, 3 mg/kg, i.p.) induced conditioned place preference (CPP) could attenuate the reinstatement of CPP brought about by OXY (1.5 mg/kg, i.p.) priming in adolescent rats of both sexes, and whether this effect is sex dependent. Accordingly, a priming dose of OXY produced reinstatement of the previously extinguished CPP in males and females. In both sexes, this effect was linked to locomotor sensitization that was blunted by CBD pretreatments. However, CBD was able to prevent the reinstatement of OXY-induced CPP only in adolescent males and this outcome was associated with an increased cannabinoid 1 receptor (CB1R) and a decreased mu opioid receptor (MOR) expression in the prefrontal cortex (PFC). The reinstatement of CCP in females was associated with a decreased MOR expression, but no changes were detected in CB1R in the hippocampus (HIP). Moreover, CBD administration during extinction significantly potentialized the reduced MOR expression in the PFC of males and showed a tendency to potentiate the reduced MOR in the HIP of females. Additionally, CBD reversed OXY-induced deficits of recognition memory only in males. These results suggest that CBD could reduce reinstatement to OXY seeking after a period of abstinence in adolescent male but not female rats. However, more investigation is required.
Collapse
Affiliation(s)
- Justyna Socha
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki 4a, 20-093 Lublin, Poland; (J.S.); (P.G.); (A.S.)
| | - Pawel Grochecki
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki 4a, 20-093 Lublin, Poland; (J.S.); (P.G.); (A.S.)
| | - Marta Marszalek-Grabska
- Department of Experimental and Clinical Pharmacology, Medical University, Jaczewskiego 8b, 20-090 Lublin, Poland;
| | - Aleksandra Skrok
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki 4a, 20-093 Lublin, Poland; (J.S.); (P.G.); (A.S.)
| | - Irena Smaga
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Smetna 12, 31-343 Krakow, Poland; (I.S.); (M.F.)
| | - Tymoteusz Slowik
- Experimental Medicine Center, Medical University, Jaczewskiego 8, 20-090 Lublin, Poland;
| | - Wojciech Prazmo
- Breast Surgery Department, Provincial Specialist Hospital, Al. Krasnicka 100, 20-718 Lublin, Poland;
| | - Robert Kotlinski
- Clinical Department of Cardiac Surgery, University of Rzeszow, Lwowska 60, 35-301 Rzeszow, Poland;
| | - Malgorzata Filip
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Smetna 12, 31-343 Krakow, Poland; (I.S.); (M.F.)
| | - Jolanta H. Kotlinska
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki 4a, 20-093 Lublin, Poland; (J.S.); (P.G.); (A.S.)
| |
Collapse
|
6
|
Lin J, Peng Y, Zhang J, Cheng J, Chen Q, Wang B, Liu Y, Niu S, Yan J. Interfering with reconsolidation by rimonabant results in blockade of heroin-associated memory. Front Pharmacol 2024; 15:1361838. [PMID: 38576487 PMCID: PMC10991728 DOI: 10.3389/fphar.2024.1361838] [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: 12/27/2023] [Accepted: 03/13/2024] [Indexed: 04/06/2024] Open
Abstract
Drug-associated pathological memory remains a critical factor contributing to the persistence of substance use disorder. Pharmacological amnestic manipulation to interfere with drug memory reconsolidation has shown promise for the prevention of relapse. In a rat heroin self-administration model, we examined the impact of rimonabant, a selective cannabinoid receptor indirect agonist, on the reconsolidation process of heroin-associated memory. The study showed that immediately administering rimonabant after conditioned stimuli (CS) exposure reduced the cue- and herion + cue-induced heroin-seeking behavior. The inhibitory effects lasted for a minimum of 28 days. The effect of Rimonabant on reduced drug-seeking was not shown when treated without CS exposure or 6 hours after CS exposure. These results demonstrate a disruptive role of rimonabant on the reconsolidation of heroin-associated memory and the therapeutic potential in relapse control concerning substance use disorder.
Collapse
Affiliation(s)
- Jiang Lin
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Yilin Peng
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Jinlong Zhang
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Department of Forensic Science, School of Basic Medical Science, Xinjiang Medical University, Urumqi, China
| | - Junzhe Cheng
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Qianqian Chen
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Binbin Wang
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Yuhang Liu
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Shuliang Niu
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Department of Anatomy, School of Basic Medical Science, Xinjiang Medical University, Urumqi, China
| | - Jie Yan
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| |
Collapse
|
7
|
Hleihil M, Benke D. Restoring GABA B receptor expression in the ventral tegmental area of methamphetamine addicted mice inhibits locomotor sensitization and drug seeking behavior. Front Mol Neurosci 2024; 17:1347228. [PMID: 38384279 PMCID: PMC10879384 DOI: 10.3389/fnmol.2024.1347228] [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: 11/30/2023] [Accepted: 01/15/2024] [Indexed: 02/23/2024] Open
Abstract
Repeated exposure to psychostimulants such as methamphetamine (METH) induces neuronal adaptations in the mesocorticolimbic dopamine system, including the ventral tegmental area (VTA). These changes lead to persistently enhanced neuronal activity causing increased dopamine release and addictive phenotypes. A factor contributing to increased dopaminergic activity in this system appears to be reduced GABAB receptor-mediated neuronal inhibition in the VTA. Dephosphorylation of serine 783 (Ser783) of the GABAB2 subunit by protein phosphatase 2A (PP2A) appears to trigger the downregulation GABAB receptors in psychostimulant-addicted rodents. Therefore, preventing the interaction of GABAB receptors with PP2A using an interfering peptide is a promising strategy to restore GABAB receptor-mediated neuronal inhibition. We have previously developed an interfering peptide (PP2A-Pep) that inhibits the GABAB receptors/PP2A interaction and thereby restores receptor expression under pathological conditions. Here, we tested the hypothesis that restoration of GABAB receptor expression in the VTA of METH addicted mice reduce addictive phenotypes. We found that the expression of GABAB receptors was significantly reduced in the VTA and nucleus accumbens but not in the hippocampus and somatosensory cortex of METH-addicted mice. Infusion of PP2A-Pep into the VTA of METH-addicted mice restored GABAB receptor expression in the VTA and inhibited METH-induced locomotor sensitization as assessed in the open field test. Moreover, administration of PP2A-Pep into the VTA also reduced drug seeking behavior in the conditioned place preference test. These observations underscore the importance of VTA GABAB receptors in controlling addictive phenotypes. Furthermore, this study illustrates the value of interfering peptides targeting diseases-related protein-protein interactions as an alternative approach for a potential development of selective therapeutic interventions.
Collapse
Affiliation(s)
- Mohammad Hleihil
- Institute of Pharmacology and Toxicology, University of Zurich, Zürich, Switzerland
| | - Dietmar Benke
- Institute of Pharmacology and Toxicology, University of Zurich, Zürich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zürich, Switzerland
| |
Collapse
|
8
|
Yang H, Zhang X, Zhang M, Lu Y, Xie B, Sun S, Yu H, Cong B, Luo Y, Ma C, Wen D. Roles of lncLingo2 and its derived miR-876-5p in the acquisition of opioid reinforcement. Addict Biol 2024; 29:e13375. [PMID: 38380802 PMCID: PMC10898844 DOI: 10.1111/adb.13375] [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: 11/14/2023] [Revised: 01/13/2024] [Accepted: 01/15/2024] [Indexed: 02/22/2024]
Abstract
Recent studies found that non-coding RNAs (ncRNAs) played crucial roles in drug addiction through epigenetic regulation of gene expression and underlying drug-induced neuroadaptations. In this study, we characterized lncRNA transcriptome profiles in the nucleus accumbens (NAc) of mice exhibiting morphine-conditioned place preference (CPP) and explored the prospective roles of novel differentially expressed lncRNA, lncLingo2 and its derived miR-876-5p in the acquisition of opioids-associated behaviours. We found that the lncLingo2 was downregulated within the NAc core (NAcC) but not in the NAc shell (NAcS). This downregulation was found to be associated with the development of morphine CPP and heroin intravenous self-administration (IVSA). As Mfold software revealed that the secondary structures of lncLingo2 contained the sequence of pre-miR-876, transfection of LV-lncLingo2 into HEK293 cells significantly upregulated miR-876 expression and the changes of mature miR-876 are positively correlated with lncLingo2 expression in NAcC of morphine CPP trained mice. Delivering miR-876-5p mimics into NAcC also inhibited the acquisition of morphine CPP. Furthermore, bioinformatics analysis and dual-luciferase assay confirmed that miR-876-5p binds to its target gene, Kcnn3, selectively and regulates morphine CPP training-induced alteration of Kcnn3 expression. Lastly, the electrophysiological analysis indicated that the currents of small conductance calcium-activated potassium (SK) channel was increased, which led to low neuronal excitability in NAcC after CPP training, and these changes were reversed by lncLingo2 overexpression. Collectively, lncLingo2 may function as a precursor of miR-876-5p in NAcC, hence modulating the development of opioid-associated behaviours in mice, which may serve as an underlying biomarker and therapeutic target of opioid addiction.
Collapse
Affiliation(s)
- Hongyu Yang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
| | - Xiuning Zhang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
| | - Minglong Zhang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
- Department of GeneticsQiqihar Medical UniversityQiqiharHeilongjiang ProvinceChina
| | - Yun Lu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
| | - Bing Xie
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
| | - Shaoguang Sun
- Department of Biochemistry and Molecular Biology, Key Laboratory of Medical Biotechnology of Hebei ProvinceHebei Medical UniversityShijiazhuangChina
- Key Laboratory of Neural and Vascular BiologyMinistry of EducationShijiazhuangHebei ProvinceChina
| | - Hailei Yu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
| | - Bin Cong
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
| | - Yixiao Luo
- Hunan Province People's HospitalThe First‐Affiliated Hospital of Hunan Normal UniversityChangshaChina
| | - Chunling Ma
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
- Key Laboratory of Neural and Vascular BiologyMinistry of EducationShijiazhuangHebei ProvinceChina
| | - Di Wen
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
- Key Laboratory of Neural and Vascular BiologyMinistry of EducationShijiazhuangHebei ProvinceChina
| |
Collapse
|
9
|
Franco-García A, Gómez-Murcia V, Fernández-Gómez FJ, González-Andreu R, Hidalgo JM, Victoria Milanés M, Núñez C. Morphine-withdrawal aversive memories and their extinction modulate H4K5 acetylation and Brd4 activation in the rat hippocampus and basolateral amygdala. Biomed Pharmacother 2023; 165:115055. [PMID: 37356373 DOI: 10.1016/j.biopha.2023.115055] [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: 03/30/2023] [Revised: 06/13/2023] [Accepted: 06/21/2023] [Indexed: 06/27/2023] Open
Abstract
Chromatin modification is a crucial mechanism in several important phenomena in the brain, including drug addiction. Persistence of drug craving and risk of relapse could be attributed to drug-induced epigenetic mechanisms that seem to be candidates explaining long-lasting drug-induced behaviour and molecular alterations. Histone acetylation has been proposed to regulate drug-seeking behaviours and the extinction of rewarding memory of drug taking. In this work, we studied the epigenetic regulation during conditioned place aversion and after extinction of aversive memory of opiate withdrawal. Through immunofluorescence assays, we assessed some epigenetic marks (H4K5ac and p-Brd4) in crucial areas related to memory retrieval -basolateral amygdala (BLA) and hippocampus-. Additionally, to test the degree of transcriptional activation, we evaluated the immediate early genes (IEGs) response (Arc, Bdnf, Creb, Egr-1, Fos and Nfkb) and Smarcc1 (chromatin remodeler) through RT-qPCR in these nuclei. Our results showed increased p-Brd4 and H4K5ac levels during aversive memory retrieval, suggesting a more open chromatin state. However, transcriptional activation of these IEGs was not found, therefore suggesting that other secondary response may already be happening. Additionally, Smarcc1 levels were reduced due to morphine chronic administration in BLA and dentate gyrus. The activation markers returned to control levels after the retrieval of aversive memories, revealing a more repressed chromatin state. Taken together, our results show a major role of the tandem H4K5ac/p-Brd4 during the retrieval of aversive memories. These results might be useful to elucidate new molecular targets to improve and develop pharmacological treatments to address addiction and to avoid drug relapse.
Collapse
Affiliation(s)
- Aurelio Franco-García
- Group of Cellular and Molecular Pharmacology, Department of Pharmacology, CEIR Campus Mare Nostrum, University of Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB) - Pascual Parrilla, Murcia, Spain
| | - Victoria Gómez-Murcia
- Group of Cellular and Molecular Pharmacology, Department of Pharmacology, CEIR Campus Mare Nostrum, University of Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB) - Pascual Parrilla, Murcia, Spain
| | - Francisco José Fernández-Gómez
- Group of Cellular and Molecular Pharmacology, Department of Pharmacology, CEIR Campus Mare Nostrum, University of Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB) - Pascual Parrilla, Murcia, Spain
| | - Raúl González-Andreu
- Group of Cellular and Molecular Pharmacology, Department of Pharmacology, CEIR Campus Mare Nostrum, University of Murcia, Spain
| | - Juana M Hidalgo
- Group of Cellular and Molecular Pharmacology, Department of Pharmacology, CEIR Campus Mare Nostrum, University of Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB) - Pascual Parrilla, Murcia, Spain
| | - M Victoria Milanés
- Group of Cellular and Molecular Pharmacology, Department of Pharmacology, CEIR Campus Mare Nostrum, University of Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB) - Pascual Parrilla, Murcia, Spain.
| | - Cristina Núñez
- Group of Cellular and Molecular Pharmacology, Department of Pharmacology, CEIR Campus Mare Nostrum, University of Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB) - Pascual Parrilla, Murcia, Spain.
| |
Collapse
|
10
|
Ghrelin/GHS-R1A antagonism in memory test and its effects on central molecular signaling involved in addiction in rats. Pharmacol Biochem Behav 2023; 224:173528. [PMID: 36870422 DOI: 10.1016/j.pbb.2023.173528] [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: 07/23/2022] [Revised: 12/23/2022] [Accepted: 02/12/2023] [Indexed: 03/06/2023]
Abstract
Central ghrelin signaling seems to play important role in addiction as well as memory processing. Antagonism of the growth hormone secretagogue receptor (GHS-R1A) has been recently proposed as a promising tool for the unsatisfactory drug addiction therapy. However, molecular aspects of GHS-R1A involvement in specific brain regions remain unclear. The present study demonstrated for the first time that acute as well as subchronic (4 days) administration of the experimental GHS-R1A antagonist JMV2959 in usual intraperitoneal doses including 3 mg/kg, had no influence on memory functions tested in the Morris Water Maze in rats as well as no significant effects on the molecular markers linked with memory processing in selected brain areas in rats, specifically on the β-actin, c-Fos, two forms of the calcium/calmodulin-dependent protein kinase II (CaMKII, p-CaMKII) and the cAMP-response element binding protein (CREB, p-CREB), within the medial prefrontal cortex (mPFC), nucleus accumbens (NAc), dorsal striatum, and hippocampus (HIPP). Furthermore, following the methamphetamine intravenous self-administration in rats, the 3 mg/kg JMV2959 pretreatment significantly reduced or prevented the methamphetamine-induced significant decrease of hippocampal β-actin and c-Fos as well as it prevented the significant decrease of CREB in the NAC and mPFC. These results imply, that the GHS-R1A antagonist/JMV2959 might reduce/prevent some of the memory-linked molecular changes elicited by methamphetamine addiction within brain structures associated with memory (HIPP), reward (NAc), and motivation (mPFC), which may contribute to the previously observed significant JMV2959-induced reduction of the methamphetamine self-administration and drug-seeking behavior in the same animals. Further research is necessary to corroborate these results.
Collapse
|
11
|
Karimi-Haghighi S, Chavoshinezhad S, Mozafari R, Noorbakhsh F, Borhani-Haghighi A, Haghparast A. Neuroinflammatory Response in Reward-Associated Psychostimulants and Opioids: A Review. Cell Mol Neurobiol 2023; 43:649-682. [PMID: 35461410 DOI: 10.1007/s10571-022-01223-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/26/2022] [Indexed: 11/03/2022]
Abstract
Substance abuse is one of the significant problems in social and public health worldwide. Vast numbers of evidence illustrate that motivational and reinforcing impacts of addictive drugs are primarily attributed to their ability to change dopamine signaling in the reward circuit. However, the roles of classic neurotransmitters, especially dopamine and neuromodulators, monoamines, and neuropeptides, in reinforcing characteristics of abused drugs have been extensively investigated. It has recently been revealed that central immune signaling includes cascades of chemokines and proinflammatory cytokines released by neurons and glia via downstream intracellular signaling pathways that play a crucial role in mediating rewarding behavioral effects of drugs. More interestingly, inflammatory responses in the central nervous system modulate the mesolimbic dopamine signaling and glutamate-dependent currents induced by addictive drugs. This review summarized researches in the alterations of inflammatory responses accompanied by rewarding and reinforcing properties of addictive drugs, including cocaine, methamphetamine, and opioids that were evaluated by conditioned place preference and self-administration procedures as highly common behavioral tests to investigate the motivational and reinforcing impacts of addictive drugs. The neuroinflammatory responses affect the rewarding properties of psychostimulants and opioids.
Collapse
Affiliation(s)
- Saeideh Karimi-Haghighi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, P.O. Box: 19615-1178, Tehran, Iran
| | - Sara Chavoshinezhad
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Roghayeh Mozafari
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, P.O. Box: 19615-1178, Tehran, Iran
| | - Farshid Noorbakhsh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | | | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, P.O. Box: 19615-1178, Tehran, Iran.
| |
Collapse
|
12
|
Osorio-Gómez D, Miranda MI, Guzmán-Ramos K, Bermúdez-Rattoni F. Transforming experiences: Neurobiology of memory updating/editing. Front Syst Neurosci 2023; 17:1103770. [PMID: 36896148 PMCID: PMC9989287 DOI: 10.3389/fnsys.2023.1103770] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 02/07/2023] [Indexed: 02/23/2023] Open
Abstract
Long-term memory is achieved through a consolidation process where structural and molecular changes integrate information into a stable memory. However, environmental conditions constantly change, and organisms must adapt their behavior by updating their memories, providing dynamic flexibility for adaptive responses. Consequently, novel stimulation/experiences can be integrated during memory retrieval; where consolidated memories are updated by a dynamic process after the appearance of a prediction error or by the exposure to new information, generating edited memories. This review will discuss the neurobiological systems involved in memory updating including recognition memory and emotional memories. In this regard, we will review the salient and emotional experiences that promote the gradual shifting from displeasure to pleasure (or vice versa), leading to hedonic or aversive responses, throughout memory updating. Finally, we will discuss evidence regarding memory updating and its potential clinical implication in drug addiction, phobias, and post-traumatic stress disorder.
Collapse
Affiliation(s)
- Daniel Osorio-Gómez
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Maria Isabel Miranda
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Mexico
| | - Kioko Guzmán-Ramos
- División de Ciencias Biológicas y de la Salud, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana, Lerma de Villada, Mexico
| | - Federico Bermúdez-Rattoni
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
13
|
Noël X. A critical perspective on updating drug memories through the integration of memory editing and brain stimulation. Front Psychiatry 2023; 14:1161879. [PMID: 37124256 PMCID: PMC10140428 DOI: 10.3389/fpsyt.2023.1161879] [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: 02/08/2023] [Accepted: 03/27/2023] [Indexed: 05/02/2023] Open
Abstract
Addiction is a persistent, recurring condition characterized by repeated relapses despite the desire to control drug use or maintain sobriety. The attainment of abstinence is hindered by persistent maladaptive drug-associated memories, which drive drug-seeking and use behavior. This article examines the preliminary evidence supporting the combination of non-invasive brain stimulation (NIBS) techniques and memory editing (or reconsolidation) interventions as add-on forms of treatment for individuals with substance-related disorders (SUD). Studies have shown that NIBS can modestly reduce drug use and craving through improved cognitive control or other undetermined reasons. Memory reconsolidation, a process by which a previously consolidated memory trace can be made labile again, can potentially erase or significantly weaken SUD memories underpinning craving and the propensity for relapse. This approach conveys enthusiasm while also emphasizing the importance of managing boundary conditions and null results for interventions found on fear memory reconsolidation. Recent studies, which align with the state-dependency and activity-selectivity hypotheses, have shown that the combination of NIBS and behavioral interventions holds promise for treating SUD by reducing self-reported and physiological aspects of craving. Effective long-term outcomes for this procedure require better identification of critical memories, a deeper understanding of the brain mechanisms underlying SUD and memory reconsolidation and overcoming any boundary conditions of destabilized memories. This will enable the procedure to be personalized to the unique needs of individual patients.
Collapse
Affiliation(s)
- Xavier Noël
- Laboratoire de Psychologie Médicale et d’Addictologie, Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- *Correspondence: Xavier Noël,
| |
Collapse
|
14
|
Rose TR, Marron Fernandez de Velasco E, Mitten EH, Wickman K. GIRK channel activity in prelimbic pyramidal neurons regulates the extinction of cocaine conditioned place preference in male mice. Addict Biol 2023; 28:e13256. [PMID: 36577727 PMCID: PMC10078116 DOI: 10.1111/adb.13256] [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: 02/11/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022]
Abstract
Drug-induced neuroadaptations in the prefrontal cortex (PFC) have been implicated in drug-associated memories that motivate continued drug use. Chronic cocaine exposure increases pyramidal neuron excitability in the prelimbic subregion of the PFC (PL), an adaptation that has been attributed in part to a suppression of inhibitory signalling mediated by the GABAB receptor (GABAB R) and G protein-gated inwardly rectifying K+ (GIRK/Kir3) channels. Although reduced GIRK channel activity in PL pyramidal neurons enhances the motor-stimulatory effect of cocaine in mice, the impact on cocaine reward and associated memories remains unclear. Here, we employed Cre- and CRISPR/Cas9-based viral manipulation strategies to evaluate the impact of GIRK channel or GABAB R ablation in PL pyramidal neurons on cocaine-induced conditioned place preference (CPP) and extinction. Neither ablation of GIRK channels nor GABAB R impacted the acquisition of cocaine CPP. GIRK channel ablation in PL pyramidal neurons, however, impaired extinction of cocaine CPP in male but not female mice. Since ablation of GIRK channels but not GABAB R increased PL pyramidal neuron excitability, we used a chemogenetic approach to determine if acute excitation of PL pyramidal neurons impaired the expression of extinction in male mice. While acute chemogenetic excitation of PL pyramidal neurons induced locomotor hyperactivity, it did not impair the extinction of cocaine CPP. Lastly, we found that persistent enhancement of GIRK channel activity in PL pyramidal neurons accelerated the extinction of cocaine CPP. Collectively, our findings show that the strength of GIRK channel activity in PL pyramidal neurons bi-directionally regulates cocaine CPP extinction in male mice.
Collapse
Affiliation(s)
- Timothy R Rose
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Eric H Mitten
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
15
|
Dai Z, Liu Y, Nie L, Chen W, Xu X, Li Y, Zhang J, Shen F, Sui N, Liang J. Locus coeruleus input-modulated reactivation of dentate gyrus opioid-withdrawal engrams promotes extinction. Neuropsychopharmacology 2023; 48:327-340. [PMID: 36302846 PMCID: PMC9751301 DOI: 10.1038/s41386-022-01477-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/08/2022] [Accepted: 10/10/2022] [Indexed: 12/26/2022]
Abstract
Extinction training during the reconsolidation window following memory recall is an effective behavioral pattern for promoting the extinction of pathological memory. However, promoted extinction by recall-extinction procedure has not been universally replicated in different studies. One potential reason for this may relate to whether initially acquired memory is successfully activated. Thus, the methods for inducing the memory into an active or plastic condition may contribute to promoting its extinction. The aim of this study is to find and demonstrate a manipulatable neural circuit that engages in the memory recall process and where its activation improves the extinction process through recall-extinction procedure. Here, naloxone-precipitated conditioned place aversion (CPA) in morphine-dependent mice was mainly used as a pathological memory model. We found that the locus coeruleus (LC)-dentate gyrus (DG) circuit was necessary for CPA memory recall and that artificial activation of LC inputs to the DG just prior to initiating a recall-extinction procedure significantly promoted extinction learning. We also found that activating this circuit caused an increase in the ensemble size of DG engram cells activated during the extinction, which was confirmed by a cFos targeted strategy to label cells combined with immunohistochemical and in vivo calcium imaging techniques. Collectively, our data uncover that the recall experience is important for updating the memory during the reconsolidation window; they also suggest a promising neural circuit or target based on the recall-extinction procedure for weakening pathological aversion memory, such as opioid withdrawal memory and fear memory.
Collapse
Affiliation(s)
- Zhonghua Dai
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Ying Liu
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Lina Nie
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Weiqi Chen
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
- Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing, China
| | - Xing Xu
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Yonghui Li
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
- Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing, China
| | - Jianjun Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Fang Shen
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Nan Sui
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Jing Liang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China.
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China.
- Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
16
|
Qian S, Shi C, Huang S, Yang C, Luo Y. DNA methyltransferase activity in the basolateral amygdala is critical for reconsolidation of a heroin reward memory. Front Mol Neurosci 2022; 15:1002139. [PMID: 36176958 PMCID: PMC9513049 DOI: 10.3389/fnmol.2022.1002139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 08/18/2022] [Indexed: 11/17/2022] Open
Abstract
The persistence of drug memory contributes to relapse to drug seeking. The association between repeated drug exposure and drug-related cues leads to cravings triggered by drug-paired cues. The erasure of drug memories has been considered a promising way to inhibit cravings and prevent relapse. The re-exposure to drug-related cues destabilizes well-consolidated drug memories, during which a de novo protein synthesis-dependent process termed “reconsolidation” occurs to restabilize the reactivated drug memory. Disrupting reconsolidation of drug memories leads to the attenuation of drug-seeking behavior in both animal models and people with addictions. Additionally, epigenetic mechanisms regulated by DNA methyltransferase (DNMT) are involved in the reconsolidation of fear and cocaine reward memory. In the present study, we investigated the role of DNMT in the reconsolidation of heroin reward memory. In the heroin self-administration model in rats, we tested the effects of DNMT inhibition during the reconsolidation process on cue-induced reinstatement, heroin-priming-induced reinstatement, and spontaneous recovery of heroin-seeking behavior. We found that the bilateral infusion of 5-azacytidine (5-AZA) inhibiting DNMT into the basolateral amygdala (BLA) immediately after heroin reward memory retrieval, but not delayed 6 h after retrieval or without retrieval, decreased subsequent cue-induced and heroin-priming-induced reinstatement of heroin-seeking behavior. These findings demonstrate that inhibiting the activity of DNMT in BLA during the reconsolidation of heroin reward memory attenuates heroin-seeking behavior, which may provide a potential strategy for the therapeutic of heroin addiction.
Collapse
Affiliation(s)
- Shuyi Qian
- Department of Nephrology and Laboratory of Kidney Disease, Hunan Provincial People’s Hospital, Hunan Normal University, Changsha, China
| | - Cuijie Shi
- Hunan Province People’s Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Shihao Huang
- National Institute on Drug Dependence, Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Chang Yang
- Hunan Province People’s Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Yixiao Luo
- Hunan Province People’s Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha, China
- *Correspondence: Yixiao Luo,
| |
Collapse
|
17
|
Xia Y, Gong Y, Wang H, Li S, Mao F. Family Function Impacts Relapse Tendency in Substance Use Disorder: Mediated Through Self-Esteem and Resilience. Front Psychiatry 2022; 13:815118. [PMID: 35237192 PMCID: PMC8882822 DOI: 10.3389/fpsyt.2022.815118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/20/2022] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Substance abuse has been a public health concern, and even after detoxification treatment, the relapse rate is still high. Family function is closely related to substance dependence. However, studies on psychological mechanisms between them are rare. OBJECTIVES We aimed to explore the mediating role of self-esteem and resilience in the pathway that family function impacts the relapse tendency among patients with substance use disorder (SUD). METHODS A total of 282 SUD patients were recruited, and standard questionnaires were administered for each patient. The relapse tendency, family function, self-esteem and resilience were assessed by the family care index questionnaire, the Connor-Davidson resilience scale, the Rosenberg self-esteem scale and the relapse tendency questionnaire. Bootstrap method was conducted for mediation analysis to test the effects of how family function affects relapse tendency mediated through self-esteem and resilience. RESULTS The average score of relapse tendency of the patients was 28.47 (SD = 11.89). Intermediary analysis found that self-esteem played an intermediary role in the relationship between family function and relapse tendency. Resilience plays an intermediary role in the relationship between family function and relapse tendency. Further, the path analysis showed that family function not only had a direct association with relapse tendency, but also indirectly related to relapse tendency through self-esteem and resilience. CONCLUSIONS Self-esteem and resilience are the key factors in the relationship between family function and relapse tendency of SUD patients.
Collapse
Affiliation(s)
- Yuwei Xia
- Department of Psychiatry and Psychology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yu Gong
- Department of Psychiatry and Psychology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hanbin Wang
- Department of Mathematics, Ximou Primary School, Yantai, China
| | - Shen Li
- Department of Psychiatry and Psychology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Fuqiang Mao
- Department of Psychiatry and Psychology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
18
|
Che X, Bai Y, Cai J, Liu Y, Li Y, Yin M, Xu T, Wu C, Yang J. Hippocampal neurogenesis interferes with extinction and reinstatement of methamphetamine-associated reward memory in mice. Neuropharmacology 2021; 196:108717. [PMID: 34273388 DOI: 10.1016/j.neuropharm.2021.108717] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 06/30/2021] [Accepted: 07/11/2021] [Indexed: 12/16/2022]
Abstract
Drugs of abuse, including morphine and cocaine, can reduce hippocampal neurogenesis (HN). Whereas promotion of HN is being increasingly recognized as a promising strategy for treating morphine and cocaine addiction. The present study is focused on exploring the changes of HN during methamphetamine (METH) administration and further clarify if HN is involved in METH-associated reward memory. After successfully establishing the conditioned place preference (CPP) paradigm to simulate the METH-associated reward memory in C57BL/6 mice, we observed that HN was significantly inhibited during METH (2 mg/kg, i. p.) administration and returned to normal after the extinction of METH CPP, as indicated by the immunostaining of bromodeoxyuridine (BrdU) and doublecortin (DCX) in the hippocampus. To promote/inhibit HN levels, 7,8-dihydroxyflavone (DHF), a small tyrosine kinase receptor B (TrkB) agonist and temozolomide (TMZ), an alkylating agent, were administered intraperitoneally (i.p.), respectively. The data showed that either DHF (5 mg/kg, i. p.) or TMZ (25 mg/kg, i. p.) pre-treatment before METH administration could significantly prolong extinction and enhance reinstatement of the reward memory. Notably, DHF treatment after METH administration significantly facilitated extinction and inhibited METH reinstatement, while TMZ treatment resulted in opposite effects. The present study indicated that METH administration could induce a temporal inhibitory effect on HN. More importantly, promotion of HN after the acquisition of METH-associated reward memory, but not inhibition of HN or promotion of HN before the acquisition of reward memory, could facilitate METH extinction and inhibit METH reinstatement, indicating the beneficial effect of HN on METH addiction by erasing the according reward memory.
Collapse
Affiliation(s)
- Xiaohang Che
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Yijun Bai
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Jialing Cai
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Yueyang Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Yuting Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Meixue Yin
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Tianyu Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Chunfu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.
| | - Jingyu Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.
| |
Collapse
|
19
|
Zhang L, Meng S, Chen W, Chen Y, Huang E, Zhang G, Liang Y, Ding Z, Xue Y, Chen Y, Shi J, Shi Y. High-Frequency Deep Brain Stimulation of the Substantia Nigra Pars Reticulata Facilitates Extinction and Prevents Reinstatement of Methamphetamine-Induced Conditioned Place Preference. Front Pharmacol 2021; 12:705813. [PMID: 34276387 PMCID: PMC8277946 DOI: 10.3389/fphar.2021.705813] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/10/2021] [Indexed: 12/21/2022] Open
Abstract
Persistent and stable drug memories lead to a high rate of relapse among addicts. A number of studies have found that intervention in addiction-related memories can effectively prevent relapse. Deep brain stimulation (DBS) exhibits distinct therapeutic effects and advantages in the treatment of neurological and psychiatric disorders. In addition, recent studies have also found that the substantia nigra pars reticulata (SNr) could serve as a promising target in the treatment of addiction. Therefore, the present study aimed to investigate the effect of DBS of the SNr on the reinstatement of drug-seeking behaviors. Electrodes were bilaterally implanted into the SNr of rats before training of methamphetamine-induced conditioned place preference (CPP). High-frequency (HF) or low-frequency (LF) DBS was then applied to the SNr during the drug-free extinction sessions. We found that HF DBS, during the extinction sessions, facilitated extinction of methamphetamine-induced CPP and prevented drug-primed reinstatement, while LF DBS impaired the extinction. Both HF and LF DBS did not affect locomotor activity or induce anxiety-like behaviors of rats. Finally, HF DBS had no effect on the formation of methamphetamine-induced CPP. In conclusion, our results suggest that HF DBS of the SNr could promote extinction and prevent reinstatement of methamphetamine-induced CPP, and the SNr may serve as a potential therapeutic target in the treatment of drug addiction.
Collapse
Affiliation(s)
- Libo Zhang
- Shenzhen Public Service Platform for Clinical Application of Medical Imaging, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China.,National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Shiqiu Meng
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Wenjun Chen
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Yun Chen
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Enze Huang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Guipeng Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Yisen Liang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Zengbo Ding
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Yanxue Xue
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Yun Chen
- Shenzhen Public Service Platform for Clinical Application of Medical Imaging, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jie Shi
- Shenzhen Public Service Platform for Clinical Application of Medical Imaging, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China.,National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Yu Shi
- Shenzhen Public Service Platform for Clinical Application of Medical Imaging, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
20
|
Rulan D, Zhenbang Y, Yipu Z, Yuan G, Galaj E, Xiaorui S, Wenshuya L, Jiaqi L, Yan Z, Chang Y, Xi Y, Li S, Yixiao L, Haishui S. Exogenous SO 2 donor treatment impairs reconsolidation of drug reward memory in mice. Eur J Pharmacol 2021; 896:173911. [PMID: 33503460 DOI: 10.1016/j.ejphar.2021.173911] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/16/2021] [Accepted: 01/21/2021] [Indexed: 12/25/2022]
Abstract
Substance-related and addictive disorders (SRADs) are characterized by compulsive drug use and recurrent relapse. The persistence of pathological drug-related memories indisputably contributes to a high propensity to relapse. Hence, strategies to disrupt reconsolidation of drug reward memory are currently being pursued as potential anti-relapse interventions. Sulfur dioxide (SO2), acting as a potential gaseous molecule, endogenously derives from sulfur amino acid and can exert significant neural regulatory effects. However, the role of SO2 in reconsolidation of drug memory has not been determined. In the present study, we used morphine- or cocaine-induced conditioned place preference (CPP) mouse models with retrieval to investigate the effects of exogenous SO2 donor treatment on reconsolidation of drug reward memory. We found that administration of SO2 donor immediately after the retrieval impaired the expression of morphine or cocaine CPP. Furthermore, the exogenous SO2 donor treatment 6 h post-retrieval or in the absence of retrieval had no effect on drug reward memory and the expression of CPP. SO2 itself did not produce aversive effects nor did it acutely block morphine CPP. Our results indicate that exogenous SO2 impairs reconsolidation of drug reward memory rather than inhibits the expression of drug reward memory. As such, SO2 holds potential for the treatment and prevention of SRADs and should be studied further.
Collapse
Affiliation(s)
- Ding Rulan
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410081, China; Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yang Zhenbang
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Zhang Yipu
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Gao Yuan
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Ewa Galaj
- National Institute on Drug Abuse, Molecular Targets and Medications Discovery Branch, Baltimore, MD, USA
| | - Shi Xiaorui
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Li Wenshuya
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Luo Jiaqi
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Zhang Yan
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yang Chang
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410081, China
| | - Yin Xi
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410081, China; Department of Functional Region of Diagnosis, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Song Li
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Luo Yixiao
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410081, China.
| | - Shi Haishui
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China; Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medicinal University, 050017, China.
| |
Collapse
|
21
|
Yang X, Wen Y, Zhang Y, Gao F, Yang J, Yang Z, Yan C. Dynamic Changes of Cytoskeleton-Related Proteins Within Reward-Related Brain Regions in Morphine-Associated Memory. Front Neurosci 2021; 14:626348. [PMID: 33584180 PMCID: PMC7876246 DOI: 10.3389/fnins.2020.626348] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 12/28/2020] [Indexed: 11/13/2022] Open
Abstract
Drug-induced memory engages complex and dynamic processes and is coordinated at multiple reward-related brain regions. The spatiotemporal molecular mechanisms underlying different addiction phases remain unknown. We investigated the role of β-actin, as well as its potential modulatory protein activity-regulated cytoskeletal-associated protein (Arc/Arg3.1) and extracellular signal-regulated kinase (ERK), in reward-related associative learning and memory using morphine-induced conditioned place preference (CPP) in mice. CPP was established by alternate morphine (10 mg/kg) injections and extinguished after a 10-day extinction training, while the withdrawal group failed to extinguish without training. In the nucleus accumbens (NAc), morphine enhanced the level of β-actin and Arc only during extinction, while p-ERK1/2 was increased during both CPP acquisition and extinction phases. In the dorsal hippocampus, morphine induced an upregulation of p-ERK only during extinction, while p-β-actin was elevated during both CPP establishment and extinction. In the dorsal hippocampus, Arc was elevated during CPP formation and suppressed during extinction. Compared with the NAc and dorsal hippocampus, dynamic changes in the medial prefrontal cortex (mPFC) and caudate putamen (CPu) were not very significant. These results suggested region-specific changes of p-β-actin, Arc/Arg3.1, and p-ERK1/2 protein during establishment and extinction phases of morphine-induced CPP. These findings unveiled a spatiotemporal molecular regulation in opiate-induced plasticity.
Collapse
Affiliation(s)
- Xixi Yang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Bio-Evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, China
| | - Yichong Wen
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yuxiang Zhang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Bio-Evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, China.,NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
| | - Feifei Gao
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Bio-Evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, China
| | - Jingsi Yang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Bio-Evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, China
| | - Zhuojin Yang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Bio-Evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, China
| | - Chunxia Yan
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Bio-Evidence Sciences Academy, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
22
|
Macatee RJ, Carr M, Afshar K, Preston TJ. Development and validation of a cannabis cue stimulus set. Addict Behav 2021; 112:106643. [PMID: 32977269 DOI: 10.1016/j.addbeh.2020.106643] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/26/2020] [Accepted: 08/31/2020] [Indexed: 11/27/2022]
Abstract
Regular cannabis use and cannabis use disorder (CUD) have become increasingly prevalent in the United States over the past two decades. Theory and empirical data suggest that the incentive salience of cannabis cues is important to the development and chronicity of CUD. Cannabis cue incentive salience is often assessed with a cannabis cue reactivity paradigm wherein cannabis-related and neutral images are presented. However, prior cannabis cue reactivity studies have been limited by the use of heterogeneous stimuli that were not properly characterized across motivational/affective characteristics, physical image attributes, or non-cannabis-related salient image features (e.g., human presence, face visibility). In order to increase standardization and flexibility of future cannabis cue reactivity tasks, the aim of the present study was to develop and validate a cannabis cue and matched neutral image database comprised of motivational/affective ratings as well as physical image attributes. 234 regular cannabis users varying in primary use method (i.e., bowl, blunt/joint, bong, vaporizer) made motivational (i.e., urge to smoke cannabis) and affective (i.e., arousal, valence) ratings of cannabis-related and neutral images matched on salient, non-cannabis-related features. Physical features (hue, saturation, value) of each image were also analyzed. Motivational/affective ratings of cannabis-related and neutral images differed as expected, and cannabis use frequency and cannabis craving correlations with cannabis image ratings generally supported stimulus validity. Motivational/affective ratings did not significantly differ across cannabis use method-specific images. This database may be a useful tool for future behavioral and neuroscience research on cannabis cue reactivity.
Collapse
|
23
|
Goldstein RZ, Barrot M, Everitt BJ, Foxe JJ. Addiction in focus: molecular mechanisms, model systems, circuit maps, risk prediction and the quest for effective interventions. Eur J Neurosci 2020; 50:2007-2013. [PMID: 31502353 DOI: 10.1111/ejn.14544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Rita Z Goldstein
- Department of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, The Leon and Norma Hess Center for Science and Medicine, New York, NY, USA
| | - Michel Barrot
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| | - Barry J Everitt
- Behavioural and Clinical Neuroscience Institute and Department of Psychology, University of Cambridge, Cambridge, UK
| | - John J Foxe
- The Cognitive Neurophysiology Laboratory, Department of Neuroscience, The Ernest J. Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
24
|
Shen X, Hui R, Luo Y, Yu H, Feng S, Xie B, Bi H, Galaj E, Cong B, Ma C, Wen D. Berberine Facilitates Extinction of Drug-Associated Behavior and Inhibits Reinstatement of Drug Seeking. Front Pharmacol 2020; 11:476. [PMID: 32390837 PMCID: PMC7194034 DOI: 10.3389/fphar.2020.00476] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 03/26/2020] [Indexed: 01/19/2023] Open
Abstract
A high rate of relapse is a major clinical problem among drug-addicted individuals. Persistent traces of drug-associated reward memories contribute to intense craving and often trigger relapse. A number of interventions on drug-associated memories have shown significant benefits in relapse prevention. Among them are pre- or post-extinction pharmacological manipulations that facilitate the extinction of drug-associated behavior. Berberine, a bioactive isoquinoline alkaloid, has been recently reported to provide therapeutic benefits for a number of central nervous system (CNS) disorders, including morphine addiction. The present study aimed to investigate whether berberine could serve as a post-extinction pharmacological intervention agent to reduce risks of reinstatement of drug seeking. We found that an intragastric administration of berberine at doses of 25 and 50 mg/kg during the critical time window significantly facilitated the extinction of morphine-reward related behavior in free access and confined conditioned place preference (CPP) extinction paradigms, and subsequently, it prevented reinstatement and spontaneous recovery of morphine-induced CPP in mice. Intriguingly, the berberine treatment with or without extinction training altered expression of plasticity-related proteins such as brain-derived neurotrophic factor (BDNF), AMPA receptors (GluA1 and GluA2) in the nucleus accumbens (NAc). Moreover, the post-extinction berberine treatment significantly reduced reinstatement of cocaine-induced CPP and operant intravenous self-administration (IVSA) memories in rats. Altogether, our findings suggest that extinction training combined with the post-extinction berberine treatment can facilitate extinction of drug-associated behavior making it an attractive therapeutic candidate in relapse prevention.
Collapse
Affiliation(s)
- Xi Shen
- College of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Rongji Hui
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang, China
| | - Yixiao Luo
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Hailei Yu
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang, China
| | - Suiyuan Feng
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang, China
| | - Bing Xie
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang, China
| | - Haitao Bi
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang, China
| | - Ewa Galaj
- Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Baltimore, MD, United States
| | - Bin Cong
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang, China
| | - Chunling Ma
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang, China
| | - Di Wen
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
25
|
Meir Drexler S, Merz CJ, Lissek S, Tegenthoff M, Wolf OT. Reactivation of the Unconditioned Stimulus Inhibits the Return of Fear Independent of Cortisol. Front Behav Neurosci 2019; 13:254. [PMID: 31780910 PMCID: PMC6861211 DOI: 10.3389/fnbeh.2019.00254] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/23/2019] [Indexed: 01/03/2023] Open
Abstract
Reconsolidation is the post-retrieval stabilization of memories, a time-limited process during which reactivated (i.e., retrieved) memories can be updated with new information, become stronger or weaker, depending on the specific treatment. We have previously shown that the stress hormone cortisol has an enhancing effect on the reconsolidation of fear memories in men. This effect was specific, i.e., limited to the conditioned stimulus (CS) that was reactivated, and did not generalize to other previously reinforced, but not reactivated CS. Based on these results, we suggested that cortisol plays a critical role in the continuous strengthening of reactivated emotional memories, contributing to their persistence and robustness. In the current study, we aimed to achieve a more generalized reconsolidation enhancement using an alternative reactivation method, i.e., by a low-intensity unconditioned stimulus (UCS) presentation instead of the more common unreinforced CS presentation. In previous studies, UCS reactivation was shown to lead to a more generalized reconsolidation effect. Therefore, we hypothesized that the combination of cortisol treatment and UCS reactivation would lead to an enhanced fear memory reconsolidation, which would generalize from previously reinforced CS to stimuli that resemble it. We tested 75 men in a 3-day fear conditioning paradigm: fear acquisition training on day 1; UCS reactivation/no reactivation and pharmacological treatment (20 mg hydrocortisone/placebo) on day 2; extinction training, reinstatement and test (of original and modified stimuli) on day 3. In contrast to our hypothesis, UCS reactivation prevented the return of fear [observed in skin conductance responses (SCR)] regardless of the pharmacological manipulation: while reinstatement to the original CS was found in the no-reactivation group, both reactivation groups (cortisol and placebo) showed no reinstatement. As the only methodological difference between our previous study and the current one was the reactivation method, we focus on UCS reactivation as the main explanation for these unexpected findings. We suggest that the robust prediction error generated by the UCS reactivation method (as opposed to CS reactivation), combined with the lower UCS intensity, has by itself weakened the emotional value of the UCS, thus preventing the return of fear to the CS that was associated with it. We call for future research to support these findings and to examine the potential of this reactivation method, or variations thereof, as a tool for therapeutic use.
Collapse
Affiliation(s)
- Shira Meir Drexler
- Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Ruhr University Bochum, Bochum, Germany
| | - Christian J. Merz
- Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Ruhr University Bochum, Bochum, Germany
| | - Silke Lissek
- Department of Neurology, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Martin Tegenthoff
- Department of Neurology, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Oliver T. Wolf
- Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
26
|
Drug addiction: a curable mental disorder? Acta Pharmacol Sin 2018; 39:1823-1829. [PMID: 30382181 DOI: 10.1038/s41401-018-0180-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 09/29/2018] [Indexed: 02/06/2023] Open
Abstract
Drug addiction is a chronic, relapsing brain disorder. Multiple neural networks in the brain including the reward system (e.g., the mesocorticolimbic system), the anti-reward/stress system (e.g., the extended amygdala), and the central immune system, are involved in the development of drug addiction and relapse after withdrawal from drugs of abuse. Preclinical and clinical studies have demonstrated that it is promising to control drug addiction by pharmacologically targeting the addiction-related systems in the brain. Here we review the pharmacological targets within the dopamine system, glutamate system, trace amine system, anti-reward system, and central immune system, which are of clinical interests. Furthermore, we discuss other potential therapies, e.g., brain stimulation, behavioral treatments, and therapeutic gene modulation, which could be effective to treat drug addiction. We conclude that, although drug addiction is a complex disorder that involves complicated neural mechanisms and psychological processes, this mental disorder is treatable and may be curable by therapies such as gene modulation in the future.
Collapse
|