1
|
Saeedi N, Pourabdolhossein F, Dadashi M, Suha A, Janahmadi M, Behzadi G, Hosseinmardi N. Faecal Microbiota Transplantation Modulates Morphine Addictive-Like Behaviours Through Hippocampal Metaplasticity. Addict Biol 2025; 30:e70034. [PMID: 40237231 PMCID: PMC12000926 DOI: 10.1111/adb.70034] [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: 08/03/2024] [Revised: 02/11/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025]
Abstract
The microbiota-gut-brain axis has been implicated in the pathology of substance use disorders (SUDs). In light of the brain's capability to reorganize itself in response to intrinsic and extrinsic stimuli, opioid-induced dysbiosis is likely to contribute to addictive behaviour through modulating neuroplasticity. In this study, a faecal microbiota transplantation (FMT) from a saline-donor was performed on morphine-treated rats to evaluate the effects of gut microbiota on morphine-induced metaplasticity and addictive behaviours. Male Wistar rats were treated with subcutaneous injections of 10 mg/kg morphine sulphate every 12 h for 9 days in an effort to induce dependence. The withdrawal syndrome was precipitated by injecting naloxone (1.5 mg/kg, ip) after the final dose of morphine. The tolerance was induced by repeated morphine injections over a period of 7 days (10 mg/kg, once a day, ip). FMT was applied daily through gavage of processed faeces 1 week before and during the morphine treatment. Field potential recordings (i.e., fEPSP) were carried out to assess short-term and long-term synaptic plasticity in the CA1 area of the hippocampus following Schaffer-collateral stimulation. Animals subjected to FMT exhibited significant reductions in naloxone-precipitated withdrawal syndrome (one-way ANOVA, p < 0.05). Tolerance to the analgesic effects of morphine was not affected by FMT (two-way ANOVA, p > 0.05). Following high-frequency stimulation (HFS) to induce long-term potentiation (LTP), a greater fEPSP slope was observed in morphine-treated animals (unpaired t test, p < 0.05). FMT from saline-donor rats diminished morphine-induced augmented LTP (unpaired t test, p < 0.05). These results highlighted the alleviating effects of FMT from saline-donors on morphine-induced metaplasticity and dependence potentially by modulating the dysbiosis of gut microbiota.
Collapse
Affiliation(s)
- Negin Saeedi
- Department of Physiology, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | | | - Masoud Dadashi
- Department of Microbiology, School of MedicineAlborz University of Medical SciencesKarajIran
| | - Ali Jaafari Suha
- Department of Physiology, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Mahyar Janahmadi
- Neurophysiology Research Center, Department of Physiology, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Gila Behzadi
- Neurophysiology Research Center, Department of Physiology, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Narges Hosseinmardi
- Neurophysiology Research Center, Department of Physiology, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
2
|
Looschen K, Khatri SN, Maulik M, Salisbury C, Carman AF, Corriveau K, Smith C, Manetti D, Romanelli MN, Arias HR, Gipson CD, Mitra S. Novel psychoplastogen DM506 reduces cue-induced heroin-seeking and inhibits tonic GABA currents in the Prelimbic Cortex. Neurochem Int 2024; 178:105785. [PMID: 38838988 DOI: 10.1016/j.neuint.2024.105785] [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: 04/04/2024] [Revised: 05/10/2024] [Accepted: 06/01/2024] [Indexed: 06/07/2024]
Abstract
Opioid use disorder is a major public health crisis that is manifested by persistent drug-seeking behavior and high relapse frequency. Most of the available treatments rely on targeting opioid receptors using small molecules that do not provide sustained symptom alleviation. Psychoplastogens are a novel class of non-opioid drugs that produce rapid and sustained effects on neuronal plasticity, intended to produce therapeutic benefits. Ibogalogs are synthetic derivatives of iboga alkaloids that lack hallucinogenic or adverse side effects. In the current study, we examine the therapeutic potential of DM506, a novel ibogalog lacking any cardiotoxic or hallucinogenic effects, in cue-induced seeking behavior following heroin self-administration. At a single systemic dose of 40 mg/kg, DM506 significantly decreased cue-induced seeking in both male and female rats at abstinence day 1 (AD1) following heroin self-administration. Upon re-testing for cue-induced seeking at AD14, we found that males receiving DM506 continued to show decreased cue-induced seeking, an effect not observed in females. Since there is evidence of psychedelics influencing tonic GABA currents, and opioid and psychoplastogen-mediated neuroadaptations in the medial prefrontal cortex (PrL) underlying its functional effects, we performed patch-clamp recordings on PrL slices of drug-naïve rats with an acute application or chronic incubation with DM506. Tonic GABA current was decreased in slices incubated with DM506 for 2 h. qPCR analysis did not reveal any differences in the mRNA levels of GABAA receptor α and δ subunits at AD14 in heroin and saline self-administered animals that received vehicle or DM506 at AD1. Overall, our data indicate that DM506 attenuates cue-induced heroin seeking and inhibits tonic GABA current in the prelimbic cortex.
Collapse
Affiliation(s)
- Kassandra Looschen
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Shailesh Narayan Khatri
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Malabika Maulik
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA; Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA
| | - Colin Salisbury
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Alaina F Carman
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Katilyn Corriveau
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Colton Smith
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Dina Manetti
- Department of Neurosciences, Psychology, Drug Research and Child Health Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Italy
| | - Maria Novella Romanelli
- Department of Neurosciences, Psychology, Drug Research and Child Health Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Italy
| | - Hugo R Arias
- Department of Pharmacology and Physiology, Oklahoma State University Center for Health Sciences, Tulsa, USA; Department of Pharmacology and Physiology, Oklahoma State University Center for Health Sciences, Tahlequah, USA
| | - Cassandra D Gipson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Swarup Mitra
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA; Department of Pharmacology and Physiology, Oklahoma State University Center for Health Sciences, Tulsa, USA.
| |
Collapse
|
3
|
Soares-Cardoso C, Leal S, Sá SI, Dantas-Barros R, Dinis-Oliveira RJ, Faria J, Barbosa J. Unraveling the Hippocampal Molecular and Cellular Alterations behind Tramadol and Tapentadol Neurobehavioral Toxicity. Pharmaceuticals (Basel) 2024; 17:796. [PMID: 38931463 PMCID: PMC11206790 DOI: 10.3390/ph17060796] [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: 05/27/2024] [Revised: 06/11/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Tramadol and tapentadol are chemically related opioids prescribed for the analgesia of moderate to severe pain. Although safer than classical opioids, they are associated with neurotoxicity and behavioral dysfunction, which arise as a concern, considering their central action and growing misuse and abuse. The hippocampal formation is known to participate in memory and learning processes and has been documented to contribute to opioid dependence. Accordingly, the present study assessed molecular and cellular alterations in the hippocampal formation of Wistar rats intraperitoneally administered with 50 mg/kg tramadol or tapentadol for eight alternate days. Alterations were found in serum hydrogen peroxide, cysteine, homocysteine, and dopamine concentrations upon exposure to one or both opioids, as well as in hippocampal 8-hydroxydeoxyguanosine and gene expression levels of a panel of neurotoxicity, neuroinflammation, and neuromodulation biomarkers, assessed through quantitative real-time polymerase chain reaction (qRT-PCR). Immunohistochemical analysis of hippocampal formation sections showed increased glial fibrillary acidic protein (GFAP) and decreased cluster of differentiation 11b (CD11b) protein expression, suggesting opioid-induced astrogliosis and microgliosis. Collectively, the results emphasize the hippocampal neuromodulator effects of tramadol and tapentadol, with potential behavioral implications, underlining the need to prescribe and use both opioids cautiously.
Collapse
Affiliation(s)
- Cristiana Soares-Cardoso
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal; (C.S.-C.); (S.L.); (R.D.-B.); or (R.J.D.-O.)
- UCIBIO—Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal
| | - Sandra Leal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal; (C.S.-C.); (S.L.); (R.D.-B.); or (R.J.D.-O.)
- UCIBIO—Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal
| | - Susana I. Sá
- RISE-HEALTH, Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal;
| | - Rita Dantas-Barros
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal; (C.S.-C.); (S.L.); (R.D.-B.); or (R.J.D.-O.)
- UCIBIO—Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal
| | - Ricardo Jorge Dinis-Oliveira
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal; (C.S.-C.); (S.L.); (R.D.-B.); or (R.J.D.-O.)
- UCIBIO—Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- FOREN-Forensic Science Experts, Av. Dr. Mário Moutinho 33-A, 1400-136 Lisboa, Portugal
| | - Juliana Faria
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal; (C.S.-C.); (S.L.); (R.D.-B.); or (R.J.D.-O.)
- UCIBIO—Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal
| | - Joana Barbosa
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal; (C.S.-C.); (S.L.); (R.D.-B.); or (R.J.D.-O.)
- UCIBIO—Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal
| |
Collapse
|
4
|
Ahmadianmoghadam MA, Nematollahi MH, Mehrabani M, Fatemi I, Rostamzadeh F, Dell'Agli M, Mehrabani M, Abolhassani M, Mehrbani M. Effect of an herbal formulation containing Peganum harmala L. and Fraxinus excelsior L. on oxidative stress, memory impairment and withdrawal syndrome induced by morphine. Int J Neurosci 2024; 134:570-583. [PMID: 36168934 DOI: 10.1080/00207454.2022.2130293] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 09/05/2022] [Accepted: 09/15/2022] [Indexed: 10/17/2022]
Abstract
Background: Traditional Persian medicine has introduced effective remedies in opioid dependence care. One of the most widely used remedies is an herbal formulation containing Peganum harmala L. and Fraxinus excelsior L. (HF). This study investigated the effects of HF to attenuate the withdrawal signs and rewarding effects in morphine-dependent rats. Methods: Forty-nine male Wistar rats were randomly divided into seven groups. The control and vehicle groups received normal saline and sodium carboxymethyl cellulose, respectively. The morphine group received morphine for one week. The single and daily dose of HF groups received morphine similar to the morphine group, and HF (1.4 and 2.8 g/kg) once a day in the daily dose group and only on the last day of the experiment in the single dose of HF group. Finally, the withdrawal signs as well biochemical tests were evaluated. The behavioral parameters were assessed by conditioned place preference (CPP), elevated plus-maze and Y-maze tests. The antioxidant activity of HF was evaluated by measurement of serum contents of malondialdehyde, stable nitric oxide metabolites and total antioxidant capacity (TAC). Moreover, the protein expression of c-fos was assessed by western blotting. Results: Daily treatment with HF significantly reduced the score of CPP behavioral test, all of the withdrawal signs, TAC and the c-fos protein level. Conclusions: The results indicated that HF might be a promising complementary treatment in reducing morphine-induced physical and psychological dependence probably through modulation of c-fos protein expression.
Collapse
Affiliation(s)
- Mohammad Ali Ahmadianmoghadam
- Herbal and Traditional Medicines Research Center, Kerman University of Medical Sciences, Kerman, Iran
- Department of Traditional Medicine, Faculty of Persian Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Hadi Nematollahi
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
- Department of Clinical Biochemistry, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mitra Mehrabani
- Herbal and Traditional Medicines Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Iman Fatemi
- Research Center of Tropical and Infectious Diseases, Kerman University of Medical Sciences, Kerman, Iran
| | - Farzaneh Rostamzadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mario Dell'Agli
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Mehrnaz Mehrabani
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Moslem Abolhassani
- Department of Clinical Biochemistry, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mehrzad Mehrbani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
5
|
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
|
6
|
Zhang J, Liu Z, Liu X, Wang X, Yu L. Intravenous Injection of GluR2-3Y Inhibits Repeated Morphine-Primed Reinstatement of Drug Seeking in Rats. Brain Sci 2023; 13:brainsci13040590. [PMID: 37190555 DOI: 10.3390/brainsci13040590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/25/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Studies have demonstrated that the α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptor is essential to drug addiction. In this study, we explored the influence of GluR2-3Y, an interfering peptide to prevent the endocytosis of AMPA receptors containing the GluR2 subunit, on morphine-seeking behavior in the rat self-administration model. After self-administration was established, the rats received intravenous injections of GluR2-3Y during the extinction sessions. There were no significant differences in both active and inactive pokes compared to the control group of rats that received GluR2-3S, indicating that GluR2-3Y has no significant influences on the extinction of morphine self-administration. The other two groups of rats were trained, extinguished, and reinstated by repeated morphine priming (respectively, called Prime 1, Prime 2, and Prime 3). Only one intravenous injection of GluR2-3Y was performed before Prime 1. Compared to the control group, GluR2-3Y did not affect Prime 1, but significantly attenuated the morphine-seeking behavior during repeated morphine-primed reinstatement, indicating an inhibitory after effect of GluR2-3Y on morphine-seeking behavior in rats. The long-term depression (LTD) in the nucleus accumbens (NAc) shell was also assessed. Pretreatment with GluR2-3Y altered the ability of LTD induction to the level of that in the naive group, while pretreatment with GluR2-3S had no effects on LTD. Our results demonstrated that the intravenous injection of GluR2-3Y, to block the endocytosis of AMPA receptors, inhibited the reinstatement of morphine-seeking behavior, which may be induced by modulating the neuronal plasticity in the NAc shell of rats.
Collapse
Affiliation(s)
- Jianjun Zhang
- College of Basic Medical, Shanxi University of Chinese Medicine, Jinzhong 030619, China
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong 030619, China
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China
| | - Zhuo Liu
- School of Crime Investigation, People’s Public Security University of China, Beijing 100038, China
| | - Xiaodong Liu
- Beijing Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiaoqian Wang
- College of Basic Medical, Shanxi University of Chinese Medicine, Jinzhong 030619, China
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong 030619, China
| | - Longchuan Yu
- School of Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
7
|
Plasil SL, Collins VJ, Baratta AM, Farris SP, Homanics GE. Hippocampal ceRNA networks from chronic intermittent ethanol vapor-exposed male mice and functional analysis of top-ranked lncRNA genes for ethanol drinking phenotypes. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2022; 2:10831. [PMID: 36908580 PMCID: PMC10004261 DOI: 10.3389/adar.2022.10831] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The molecular mechanisms regulating the development and progression of alcohol use disorder (AUD) are largely unknown. While noncoding RNAs have previously been implicated as playing key roles in AUD, long-noncoding RNA (lncRNA) remains understudied in relation to AUD. In this study, we first identified ethanol-responsive lncRNAs in the mouse hippocampus that are transcriptional network hub genes. Microarray analysis of lncRNA, miRNA, circular RNA, and protein coding gene expression in the hippocampus from chronic intermittent ethanol vapor- or air- (control) exposed mice was used to identify ethanol-responsive competing endogenous RNA (ceRNA) networks. Highly interconnected lncRNAs (genes that had the strongest overall correlation to all other dysregulated genes identified) were ranked. The top four lncRNAs were novel, previously uncharacterized genes named Gm42575, 4930413E15Rik, Gm15767, and Gm33447, hereafter referred to as Pitt1, Pitt2, Pitt3, and Pitt4, respectively. We subsequently tested the hypothesis that CRISPR/Cas9 mutagenesis of the putative promoter and first exon of these lncRNAs in C57BL/6J mice would alter ethanol drinking behavior. The Drinking in the Dark (DID) assay was used to examine binge-like drinking behavior, and the Every-Other-Day Two-Bottle Choice (EOD-2BC) assay was used to examine intermittent ethanol consumption and preference. No significant differences between control and mutant mice were observed in the DID assay. Female-specific reductions in ethanol consumption were observed in the EOD-2BC assay for Pitt1, Pitt3, and Pitt4 mutant mice compared to controls. Male-specific alterations in ethanol preference were observed for Pitt1 and Pitt2. Female-specific increases in ethanol preference were observed for Pitt3 and Pitt4. Total fluid consumption was reduced in Pitt1 and Pitt2 mutants at 15% v/v ethanol and in Pitt3 and Pitt4 at 20% v/v ethanol in females only. We conclude that all lncRNAs targeted altered ethanol drinking behavior, and that lncRNAs Pitt1, Pitt3, and Pitt4 influenced ethanol consumption in a sex-specific manner. Further research is necessary to elucidate the biological mechanisms for these effects. These findings add to the literature implicating noncoding RNAs in AUD and suggest lncRNAs also play an important regulatory role in the disease.
Collapse
Affiliation(s)
- SL Plasil
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - VJ Collins
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - AM Baratta
- Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - SP Farris
- Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - GE Homanics
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
8
|
High Morphine Use Disorder Susceptibility Is Predicted by Impaired Learning Ability in Mice. Brain Sci 2022; 12:brainsci12121650. [PMID: 36552110 PMCID: PMC9776386 DOI: 10.3390/brainsci12121650] [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: 09/24/2022] [Revised: 10/24/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
An obvious reason for substance uses disorders (SUDs) is drug craving and seeking behavior induced by conditioned context, which is an abnormal solid context memory. The relationship between susceptibility to SUD and learning ability remains unclear in humans and animal models. In this study, we found that susceptibility to morphine use disorder (MUD) was negatively correlated with learning ability in conditioned place preference (CPP) in C57 mice. By using behavioral tests, we identified the FVB mouse as learning impaired. In addition, we discovered that learning-relevant proteins, such as the glutamate receptor subunits GluA1, NR1, and NR2A, were decreased in FVB mice. Finally, we assessed the context learning ability of FVB mice using the CPP test and priming. We found that FVB mice had lower learning performance with respect to normal memory but higher performance of morphine-reinstatement memory. Compared to C57 mice, FVB mice are highly sensitive to MUDs. Our results suggest that SUD susceptibility is predicted by impaired learning ability in mice; therefore, learning ability can play a simple and practical role in identifying high-risk SUD groups.
Collapse
|
9
|
A Novel CaMKII Inhibitory Peptide Blocks Relapse to Morphine Seeking by Influencing Synaptic Plasticity in the Nucleus Accumbens Shell. Brain Sci 2022; 12:brainsci12080985. [PMID: 35892425 PMCID: PMC9394410 DOI: 10.3390/brainsci12080985] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/12/2022] [Accepted: 07/23/2022] [Indexed: 12/04/2022] Open
Abstract
Drugs of abuse cause enduring functional disorders in the brain reward circuits, leading to cravings and compulsive behavior. Although people may rehabilitate by detoxification, there is a high risk of relapse. Therefore, it is crucial to illuminate the mechanisms of relapse and explore the therapeutic strategies for prevention. In this research, by using an animal model of morphine self-administration in rats and a whole-cell patch–clamp in brain slices, we found changes in synaptic plasticity in the nucleus accumbens (NAc) shell were involved in the relapse to morphine-seeking behavior. Compared to the controls, the amplitude of long-term depression (LTD) induced in the medium spiny neurons increased after morphine self-administration was established, recovered after the behavior was extinguished, and increased again during the relapse induced by morphine priming. Intravenous injection of MA, a new peptide obtained by modifying Ca2+/calmodulin-dependent protein kinase II (CaMKII) inhibitor “myr-AIP”, decreased CaMKII activity in the NAc shell and blocked the reinstatement of morphine-seeking behavior without influence on the locomotor activity. Moreover, LTD was absent in the NAc shell of the MA-pretreated rats, whereas it was robust in the saline controls in which morphine-seeking behavior was reinstated. These results indicate that CaMKII regulates morphine-seeking behavior through its involvement in the change of synaptic plasticity in the NAc shell during the relapse, and MA may be of great value in the clinical treatment of relapse to opioid seeking.
Collapse
|
10
|
Coffey KR, Lesiak AJ, Marx RE, Vo EK, Garden GA, Neumaier JF. A cAMP-Related Gene Network in Microglia Is Inversely Regulated by Morphine Tolerance and Withdrawal. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2022; 2:180-189. [PMID: 35441155 PMCID: PMC9015218 DOI: 10.1016/j.bpsgos.2021.07.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 06/29/2021] [Accepted: 07/27/2021] [Indexed: 11/12/2022] Open
Abstract
Background Microglia have recently been implicated in opioid dependence and withdrawal. Mu Opioid (MOR) receptors are expressed in microglia, and microglia form intimate connections with nearby neurons. Accordingly, opioids have both direct (MOR mediated) and indirect (neuron-interaction mediated) effects on microglia function. Methods To investigate this directly, we used RNA sequencing of ribosome-associated RNAs from striatal microglia (RiboTag-Seq) after the induction of morphine tolerance and followed by naloxone precipitated withdrawal (n=16). We validated the RNA-Seq data by combining fluorescent in-situ hybridization with immunohistochemistry for microglia (n=18). Finally, we expressed and activated the Gi/o-coupled hM4Di DREADD receptor in CX3CR1-expressing cells during morphine withdrawal (n=18). Results We detected large, inverse changes in RNA translation following opioid tolerance and withdrawal. WGCNA analysis revealed an intriguing network of cAMP-associated genes that are known to be involved in microglial motility, morphology, and interactions with neurons that were downregulated with morphine tolerance and upregulated rapidly by withdrawal. Three-dimensional histological reconstruction of microglia allowed for volumetric, visual colocalization of mRNA within individual microglia that validated our bioinformatics results. Direct activation of Gi/o-coupled DREADD receptors in CX3CR1-expressing cells exacerbated signs of opioid withdrawal rather than mimicking the effects of morphine. Conclusions These results indicate that Gi-signaling and cAMP-associated gene networks are inversely engaged during opioid tolerance and early withdrawal, perhaps revealing a role of microglia in mitigating the consequences of opioids.
Collapse
Affiliation(s)
- Kevin R. Coffey
- Puget Sound VA Health Care System, University of Washington School of Medicine, Seattle, Washington
- Department of Psychiatry & Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington
| | - Atom J. Lesiak
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, Washington
| | - Ruby E. Marx
- Puget Sound VA Health Care System, University of Washington School of Medicine, Seattle, Washington
- Department of Psychiatry & Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington
| | - Emily K. Vo
- Puget Sound VA Health Care System, University of Washington School of Medicine, Seattle, Washington
- Department of Psychiatry & Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington
| | - Gwenn A. Garden
- Department of Neurology, University of North Carolina, Chapel Hill, North Carolina
| | - John F. Neumaier
- Puget Sound VA Health Care System, University of Washington School of Medicine, Seattle, Washington
- Department of Psychiatry & Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington
| |
Collapse
|
11
|
Li J, Shao D, Jiang D, Huang Q, Guan Y, Lai B, Zhao J, Hua F, Xie F. Alteration of neuroinflammation detected by 18F-GE180 PET imaging in place-conditioned rats with morphine withdrawal. EJNMMI Res 2021; 11:103. [PMID: 34637020 PMCID: PMC8511235 DOI: 10.1186/s13550-021-00849-9] [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: 06/08/2021] [Accepted: 09/28/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Accumulating evidence indicates that neuroinflammation (NI) significantly contributes to drug addiction, but the conversion of NI after drug withdrawal is not clear. Here, we conducted 18F-flutriciclamide (GE180) positron emission tomography (PET) imaging to investigate the conversion of NI during drug withdrawal and conditioning-induced aversion by measuring the change in microglial activation with 18F-GE180. METHODS Twelve male adult Sprague-Dawley rats were subjected to morphine withdrawal by the administration of naloxone, and six of them were used to model conditioned place aversion (CPA). 18F-GE180 PET imaging was performed for 11 rats on the last day of the morphine treatment phase and for 10 rats on the response assessment phase of the behavior conditioning procedure. A 18F-GE180 template was established for spatial normalization of each individual image, and the differential 18F-GE180 uptakes between the drug withdrawal (DW) group and the drug addiction (DA) group, the CPA group and the DA group, and the CPA group and the DW group were compared by a voxel-wise two-sample t test using SPM8. RESULTS Both the DW group and the CPA group spent less time in the conditioning cage during the post-test phase compared with the pretest phase, but only the difference in the CPA group was significant (63.2 ± 34.6 vs. - 159.53 ± 22.02, P < 0.005). Compared with the DA group, the uptake of 18F-GE180 increased mainly in the hippocampus, visual cortex, thalamus and midbrain regions and decreased mainly in the sensory-related cortices after the administration of naloxone in both the DW and CPA groups. Increased 18F-GE180 uptake was only observed in the mesolimbic regions after conditioned aversion compared with the DW group. CONCLUSION In morphine-dependent rats, Neuroinflammation (NI) became more severe in the addiction-involved brain regions but remitted in the sensory-related brain regions after the administration of naloxone, and this NI induced by withdrawal was further aggravated after conditioned aversion formation thus may help to consolidate the withdrawal memory.
Collapse
Affiliation(s)
- Junpeng Li
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Da Shao
- Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, Shanghai, 200032, China.,Research Center of Translation Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Donglang Jiang
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Qi Huang
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Bin Lai
- Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Jun Zhao
- Department of Nuclear Medicine, Dongfang Hospital, Tongji University, Shanghai, 200120, China.
| | - Fengchun Hua
- Department of Nuclear Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Fang Xie
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
12
|
Proteomic Analysis Unveils Expressional Changes in Cytoskeleton- and Synaptic Plasticity-Associated Proteins in Rat Brain Six Months after Withdrawal from Morphine. Life (Basel) 2021; 11:life11070683. [PMID: 34357055 PMCID: PMC8304287 DOI: 10.3390/life11070683] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/28/2021] [Accepted: 07/10/2021] [Indexed: 11/17/2022] Open
Abstract
Drug withdrawal is associated with abstinence symptoms including deficits in cognitive functions that may persist even after prolonged discontinuation of drug intake. Cognitive deficits are, at least partially, caused by alterations in synaptic plasticity but the precise molecular mechanisms have not yet been fully identified. In the present study, changes in proteomic and phosphoproteomic profiles of selected brain regions (cortex, hippocampus, striatum, and cerebellum) from rats abstaining for six months after cessation of chronic treatment with morphine were determined by label-free quantitative (LFQ) proteomic analysis. Interestingly, prolonged morphine withdrawal was found to be associated especially with alterations in protein phosphorylation and to a lesser extent in protein expression. Gene ontology (GO) term analysis revealed enrichment in biological processes related to synaptic plasticity, cytoskeleton organization, and GTPase activity. More specifically, significant changes were observed in proteins localized in synaptic vesicles (e.g., synapsin-1, SV2a, Rab3a), in the active zone of the presynaptic nerve terminal (e.g., Bassoon, Piccolo, Rims1), and in the postsynaptic density (e.g., cadherin 13, catenins, Arhgap35, Shank3, Arhgef7). Other differentially phosphorylated proteins were associated with microtubule dynamics (microtubule-associated proteins, Tppp, collapsin response mediator proteins) and the actin–spectrin network (e.g., spectrins, adducins, band 4.1-like protein 1). Taken together, a six-month morphine withdrawal was manifested by significant alterations in the phosphorylation of synaptic proteins. The altered phosphorylation patterns modulating the function of synaptic proteins may contribute to long-term neuroadaptations induced by drug use and withdrawal.
Collapse
|
13
|
Asuni GP, Speidell A, Mocchetti I. Neuronal apoptosis induced by morphine withdrawal is mediated by the p75 neurotrophin receptor. J Neurochem 2021; 158:169-181. [PMID: 33742683 PMCID: PMC10176599 DOI: 10.1111/jnc.15355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/22/2021] [Accepted: 03/15/2021] [Indexed: 01/01/2023]
Abstract
Morphine withdrawal evokes neuronal apoptosis through mechanisms that are still under investigation. We have previously shown that morphine withdrawal increases the levels of pro-brain-derived neurotrophic factor (BDNF), a proneurotrophin that promotes neuronal apoptosis through the binding and activation of the pan-neurotrophin receptor p75 (p75NTR). In this work, we sought to examine whether morphine withdrawal increases p75NTR-driven signaling events. We employed a repeated morphine treatment-withdrawal paradigm in order to investigate biochemical and histological indicators of p75NTR-mediated neuronal apoptosis in mice. We found that repeated cycles of spontaneous morphine withdrawal promote an accumulation of p75NTR in hippocampal synapses. At the same time, TrkB, the receptor that is crucial for BDNF-mediated synaptic plasticity in the hippocampus, was decreased, suggesting that withdrawal alters the neurotrophin receptor environment to favor synaptic remodeling and apoptosis. Indeed, we observed evidence of neuronal apoptosis in the hippocampus, including activation of c-Jun N-terminal kinase (JNK) and increased active caspase-3. These effects were not seen in saline or morphine-treated mice which had not undergone withdrawal. To determine whether p75NTR was necessary in promoting these outcomes, we repeated these experiments in p75NTR heterozygous mice. The lack of one p75NTR allele was sufficient to prevent the increases in phosphorylated JNK and active caspase-3. Our results suggest that p75NTR participates in the neurotoxic and proinflammatory state evoked by morphine withdrawal. Because p75NTR activation negatively influences synaptic repair and promotes cell death, preventing opioid withdrawal is crucial for reducing neurotoxic mechanisms accompanying opioid use disorders.
Collapse
Affiliation(s)
- Gino P. Asuni
- Laboratory of Preclinical Neurobiology, Georgetown University Medical Center, Washington DC, USA
| | - Andrew Speidell
- Laboratory of Preclinical Neurobiology, Georgetown University Medical Center, Washington DC, USA
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington DC, USA
| | - Italo Mocchetti
- Laboratory of Preclinical Neurobiology, Georgetown University Medical Center, Washington DC, USA
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington DC, USA
| |
Collapse
|
14
|
Wang S, Zeng M, Ren Y, Han S, Li J, Cui W. In vivo reduction of hippocampal Caveolin-1 by RNA interference alters morphine addiction and neuroplasticity changes in male mice. Neurosci Lett 2021; 749:135742. [PMID: 33607203 DOI: 10.1016/j.neulet.2021.135742] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/29/2021] [Accepted: 02/12/2021] [Indexed: 11/29/2022]
Abstract
Prescription opioids are powerful pain-controlling medications that have both benefits and potentially serious risks. Morphine is one of the preferred analgesics that are widely used to treat chronic pain. However, chronic morphine exposure has been found to cause both functional and structural changes in several brain regions, including the medial prefrontal cortex (mPFC), ventral tegmental area (VTA), and hippocampus (HPC), which lead to addictive behavior. Caveolin-1 (Cav-1), a scaffolding protein of membrane lipid rafts (MLRs), has been shown to organize GPCRs and multiple synaptic signaling proteins within the MLRs to regulate synaptic signaling and neuroplasticity. Previously, we showed that in vitro morphine treatment significantly elevates Cav-1 expression and causes neuroplasticity changes. In this study, we confirmed that chronic morphine exposure can significantly increase Cav-1 expression (P < 0.05) and microtubule-associated protein (MAP-2)-positive neuronal dendritic growth in the hippocampus. Moreover, the rewarding effect and dendritic growth in the HPC induced by chronic morphine exposure were significantly inhibited by hippocampal Cav-1 knockdown. Together, these data suggest that Cav-1 in the hippocampus plays an essential role in the neuroplasticity changes that underlie morphine addiction behaviors.
Collapse
Affiliation(s)
- Shanshan Wang
- Department of Anesthesiology, Beijing Tian Tan Hospital, Capital Medical University, #119 Nan Si Huan Xi Lu, Beijing, 100050, PR China; Department of Anesthesiology, University of California San Diego, 3350 La Jolla Village Dr., San Diego, CA, 92161, USA
| | - Min Zeng
- Department of Anesthesiology, Beijing Tian Tan Hospital, Capital Medical University, #119 Nan Si Huan Xi Lu, Beijing, 100050, PR China
| | - Yi Ren
- Department of Anesthesiology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, #56 Nan Li Shi Lu, Beijing, 100045, PR China
| | - Song Han
- Department of Neurobiology and Beijing Institute for Neuroscience, Capital Medical University, #10 You An Men Wai Xi TouTiao, Beijing, 100069, PR China
| | - Junfa Li
- Department of Neurobiology and Beijing Institute for Neuroscience, Capital Medical University, #10 You An Men Wai Xi TouTiao, Beijing, 100069, PR China
| | - Weihua Cui
- Department of Anesthesiology, Beijing Tian Tan Hospital, Capital Medical University, #119 Nan Si Huan Xi Lu, Beijing, 100050, PR China.
| |
Collapse
|
15
|
Fatahi Z, Zeinaddini-Meymand A, Karimi-Haghighi S, Haghparast A, Khodagholi F, Haghparast A. BDNF and p-GSK3β in the hippocampus mediate the impairment of delay-based decision making in morphine-dependent rats. Neuroreport 2020; 31:1208-1214. [PMID: 33075004 DOI: 10.1097/wnr.0000000000001535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
It has been shown that morphine addiction impairs cognitive brain functions. However, there is no document to consider the effect of morphine dependency and its withdrawal on cost-benefit decision making and its molecular pathways. The present study aimed to evaluate the influences of morphine dependency and its withdrawal on delay-based decision making and the BDNF, p-GSK3β, and p-CREB levels during the decision making in the hippocampus. Different groups of rats were trained in a T-maze with the delay-based cost-benefit decision-making paradigm. After that, the animals were dependent on morphine, and the percentage of the high reward preference was evaluated. After behavioral tests, BDNF level, p-GSK3β/GSK3β ratio, and p-CREB/CREB ratio in the hippocampus measured by Western blot analysis. The gathered data showed that level of BDNF enhanced while p-GSK3β/GSK3β ratio and p-CREB/CREB ratio in the hippocampus did not change during delay-based decision making. In morphine-dependent rats, the p-GSK3β/GSK3β ratio increased, the BDNF level and p-CREB/CREB ratio did not change in the decision making procedure. After withdrawal from morphine, the BDNF level raised while p-GSK3β/GSK3β ratio and p-CREB/CREB ratio did not change compared to the addiction group. The data declared that BDNF in the hippocampus has a critical role in delay-based decision making. Change in p-CREB in the hippocampus is not related to decision making in normal and morphine-dependent rats. P-GSK3 in the hippocampus is not involved in the decision making in normal rats, but during decision making in morphine-dependent rats, its level increased.
Collapse
Affiliation(s)
- Zahra Fatahi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences
| | - Arman Zeinaddini-Meymand
- Neurobiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences
| | - Saeideh Karimi-Haghighi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences
| | - Amir Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences
| | - Fariba Khodagholi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences
| |
Collapse
|
16
|
Guo KF, Dai M, Liu YM, Zhang JC, Chen YM, Ye H, Li MB, Mao RR, Cao J. Acute Administration of Methyleugenol Impairs Hippocampus-Dependent Contextual Fear Memory and Increases Anxiety-like Behavior in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:7490-7497. [PMID: 32551566 DOI: 10.1021/acs.jafc.0c01863] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Methyleugenol (ME) as a natural essential oil in many plant species is widely used in human food and beverage for its fragrance and possible beneficial health effects. Previous chronic or subacute studies in rodents show that ME mainly causes liver toxicity. However, whether and how acute ME affects the central nervous system still remain elusive. Here, we found that ME administrated into the hippocampus impaired the acquisition of hippocampus-dependent contextual fear memory in mice (ME vs control: repeated-measures two-way ANOVA, F(5,70) = 2.937, p < 0.05; Fisher test, p < 0.05, respectively, 53 ± 5.2% vs 73 ± 7.6% during trial 4 and 46.8 ± 6% vs 74.5 ± 9.3% during trial 5). Meanwhile, acute ME impaired hippocampal CA1 long-term potentiation (LTP; ME vs control: independent t-test, p < 0.01, 110.6 ± 1.8% vs 133.3 ± 5.6%) while facilitated long-term depression (LTD; p < 0.01, 75.7 ± 3.4% vs 88.6 ± 1.7%) in mice brain slices and inducing a decrease in learning-dependent phosphorylation of Ser831 (ME vs control: independent t-test, p < 0.001, 0.87 ± 0.03 vs 1.23 ± 0.03) and Ser845 (p < 0.01, 0.42 ± 0.07 vs 0.97 ± 0.14) sites of excitatory glutamate AMPA receptor subunit 1 (GluA1) in the hippocampus, which may be the underlying mechanisms of impairment of hippocampus-dependent learning. In addition, intrahippocampal infusion of ME also increased anxiety-like behaviors in mice. These results suggested that acute ME impaired the hippocampus function at behavioral, cellular, and molecular levels, indicating the potential risks of ME on the central nervous system.
Collapse
Affiliation(s)
- Kai-Fei Guo
- Yunnan Key Laboratory of Plant Reproductive Adaption and Evolutionary Ecology and School of Ecology and Environmental Science, Yunnan University, Kunming, Yunnan 650504, China
- School of Agriculture, Yunnan University, Kunming, Yunnan 650504, China
| | - Man Dai
- Yunnan Key Laboratory of Plant Reproductive Adaption and Evolutionary Ecology and School of Ecology and Environmental Science, Yunnan University, Kunming, Yunnan 650504, China
| | - Yi-Miao Liu
- Yunnan Key Laboratory of Plant Reproductive Adaption and Evolutionary Ecology and School of Ecology and Environmental Science, Yunnan University, Kunming, Yunnan 650504, China
| | - Ji-Chuan Zhang
- Department of Basic Medicine, Medical School, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Yan-Mei Chen
- Department of Basic Medicine, Medical School, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Hui Ye
- School of Agriculture, Yunnan University, Kunming, Yunnan 650504, China
| | - Man-Bi Li
- Yunnan Institute of Environmental Sciences, Kunming, Yunnan 650034, China
| | - Rong-Rong Mao
- Department of Pathology and Pathophysiology, School of Basic Medical Science, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Jun Cao
- Yunnan Key Laboratory of Plant Reproductive Adaption and Evolutionary Ecology and School of Ecology and Environmental Science, Yunnan University, Kunming, Yunnan 650504, China
| |
Collapse
|
17
|
Liu LT, Song YQ, Chen XS, Liu Y, Zhu JJ, Zhou LM, Xu SJ, Wan LH. Morphine-induced RACK1-dependent autophagy in immortalized neuronal cell lines. Br J Pharmacol 2020; 177:1609-1621. [PMID: 31747048 DOI: 10.1111/bph.14922] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 10/08/2019] [Accepted: 10/21/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Autophagy is a critical cellular catabolic process in cell homoeostasis and brain function. Recent studies indicate that receptor for activated C kinase 1 (RACK1) is involved in autophagosome formation in Drosophila and mice, and that it plays an essential role in morphine-associated memory. However, the exact mechanism of the role of RACK1 in morphine-induced autophagy is not fully understood. EXPERIMENTAL APPROACH SH-SY5Y cells were cultured and morphine, rapamycin, 3-methyladenine and RACK1 siRNA were used to evaluate the regulation of RACK1 protein in autophagy. Western blotting and immunofluorescence were used to assess protein expression. KEY RESULTS Activation of autophagy (i.e. autophagosome accumulation and an increase in the LC3-II/LC3-I ratio) induced by morphine contributes to the maintenance of conditioned place preference (CPP) memory in mice. Moreover, morphine treatment significantly increased Beclin-1 expression and decreased the p-mTOR/mTOR and SQSTM1/p62 levels, whereas knockdown of RACK1 prevented morphine-induced autophagy in vitro. Furthermore, we found that in the mouse hippocampus, knockdown of RACK1 also markedly suppressed morphine-induced autophagy (decreased LC3-II/LC3-I ratio and increased p-mTOR/mTOR ratio). Importantly, morphine-induced autophagy in a RACK1-dependent manner. Conversely, morphine-induced RACK1 upregulation in vitro is partially inhibited by autophagy feedback. CONCLUSIONS AND IMPLICATIONS Our findings revealed a critical role for RACK1-dependent autophagy in morphine-promoted maintenance of CPP memory in mice and supported the notion that control of RACK1-dependent autophagic pathways may become an important target for novel therapeutics for morphine-associated memory.
Collapse
Affiliation(s)
- Li-Tao Liu
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Ying-Qi Song
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Xue-Shen Chen
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Yin Liu
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China.,Department of Anesthesiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Jie-Jun Zhu
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Li-Ming Zhou
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Shi-Jun Xu
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li-Hong Wan
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Aghighi F, Mohammadifar M, Banafsheh H, Salami M, Talaei SA. Behavioral and electrophysiological aspects of cognition in neonate rats lactated by morphine addicted mothers. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019; 22:1059-1064. [PMID: 31807250 PMCID: PMC6880524 DOI: 10.22038/ijbms.2019.36892.8789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Objectives In addition to genetic factors, environmental phenomena during postnatal age highly affect development and, in turn, function of the brain. The present work evaluates if morphine consumption during lactation period influences the spatial performances and synaptic plasticity in rats at neonatal period of age. Materials and Methods Three groups of mothers were subcutaneously administered by 5 (M5), 10 (M10) or 20 (M20) mg/kg morphine every 12 hours during the lactation period. At 45 days old, their offspring were introduced to Morris water maze for assessment of spatial learning and memory. Basic field excitatory post-synaptic potentials (fEPSPs) were recorded in the CA1 area of hippocampus and, then, long term potentiation (LTP) was induced by tetanic stimulation. Results We found that the M10 and M20 rats spent more time and traveled longer distance to find the hidden platform of maze when compared to the control animals (P<0.05 for all comparisons). Similarly, these two morphine-exposed groups were inferior in the memory consolidation compared to their control counterparts. Comparing control and M20 rats revealed that morphine exposure decreases the mean amplitude and slope 10-90% of fEPSPs about 30 percent (P<0.001 for both comparisons) and inhibits the LTP induction in the CA1 area circuits. Conclusion The present study provides behavioral and electrophysiological proofs for negative effect of morphine on the hippocampal-related function in the neonatally morphine-exposed rats.
Collapse
Affiliation(s)
- Fatemeh Aghighi
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mojgan Mohammadifar
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamidreza Banafsheh
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mahmoud Salami
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Sayyed Alireza Talaei
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
19
|
Zhang Q, Zhang M, Li J, Xiao H, Wu D, Guo Q, Zhang Y, Wang H, Li S, Liao S. Selection and Validation of Reference Genes for RT-PCR Expression Analysis of Candidate Genes Involved in Morphine-Induced Conditioned Place Preference Mice. J Mol Neurosci 2018; 66:587-594. [PMID: 30386959 DOI: 10.1007/s12031-018-1198-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 10/15/2018] [Indexed: 12/18/2022]
Abstract
The expression of reference genes should be constitutively stable under the experimental conditions, so determining stable reference genes is critical for obtaining reliable results in gene expression studies. Morphine addiction persistently influences neurotransmitters and signal transduction systems, which may negatively alter behavioral responses at the cellular levels and interfere the expression of reference genes. In order to research morphine dependence, animal models are commonly used in physiology, pathology, and therapeutics field since human trials have many limitations. Therefore, it is necessary to select stable reference genes in standardized animal model. The objective of this study is to find out a set of optimal reference genes to standardize the gene expression of morphine-induced conditioned place preference (CPP) mice. During the process, eight reference genes were chosen. Then, the stability of their expression in two different brain tissues (Caudate Putamen and Hippocampus) was tested in two developmental stages (puberty and adult) under two treatments (physiological saline as control and morphine). Based on two algorithm-based methods (geNorm and NormFinder), which can rank and assess the stability of expression of eight reference genes, thereby quantifying the transcriptional levels of these genes by high sensitive, specific, and accurate real-time quantitative reverse transcription PCR (RT-qPCR) assays.
Collapse
Affiliation(s)
- Qian Zhang
- Henan Provincial Key Medical Laboratory of Genetics, Institute of Medical Genetics, Henan Provincial People's Hospital, Zhengzhou, Henan, China.,Zhengzhou University People's Hospital, Zhengzhou, Henan, China.,Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Mengting Zhang
- Henan Provincial Key Medical Laboratory of Genetics, Institute of Medical Genetics, Henan Provincial People's Hospital, Zhengzhou, Henan, China.,Zhengzhou University People's Hospital, Zhengzhou, Henan, China
| | - Jiaqi Li
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, China.,College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hai Xiao
- Henan Provincial Key Medical Laboratory of Genetics, Institute of Medical Genetics, Henan Provincial People's Hospital, Zhengzhou, Henan, China.,Zhengzhou University People's Hospital, Zhengzhou, Henan, China
| | - Dong Wu
- Henan Provincial Key Medical Laboratory of Genetics, Institute of Medical Genetics, Henan Provincial People's Hospital, Zhengzhou, Henan, China.,Zhengzhou University People's Hospital, Zhengzhou, Henan, China
| | - Qiannan Guo
- Henan Provincial Key Medical Laboratory of Genetics, Institute of Medical Genetics, Henan Provincial People's Hospital, Zhengzhou, Henan, China.,Zhengzhou University People's Hospital, Zhengzhou, Henan, China
| | - Yuwei Zhang
- Henan Provincial Key Medical Laboratory of Genetics, Institute of Medical Genetics, Henan Provincial People's Hospital, Zhengzhou, Henan, China.,Zhengzhou University People's Hospital, Zhengzhou, Henan, China
| | - Hongdan Wang
- Henan Provincial Key Medical Laboratory of Genetics, Institute of Medical Genetics, Henan Provincial People's Hospital, Zhengzhou, Henan, China. .,Zhengzhou University People's Hospital, Zhengzhou, Henan, China. .,College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Shengbin Li
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, China. .,College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Shixiu Liao
- Henan Provincial Key Medical Laboratory of Genetics, Institute of Medical Genetics, Henan Provincial People's Hospital, Zhengzhou, Henan, China. .,Zhengzhou University People's Hospital, Zhengzhou, Henan, China.
| |
Collapse
|
20
|
Abstract
This paper is the thirty-ninth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2016 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior, and the roles of these opioid peptides and receptors in pain and analgesia, stress and social status, tolerance and dependence, learning and memory, eating and drinking, drug abuse and alcohol, sexual activity and hormones, pregnancy, development and endocrinology, mental illness and mood, seizures and neurologic disorders, electrical-related activity and neurophysiology, general activity and locomotion, gastrointestinal, renal and hepatic functions, cardiovascular responses, respiration and thermoregulation, and immunological responses.
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and CUNY Neuroscience Collaborative, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
21
|
Chen C, Nong Z, Liang X, Meng M, Xuan F, Xie Q, He J, Huang R. Effect of Yulangsan Polysaccharide on the Reinstatement of Morphine-Induced Conditioned Place Preference in Sprague-Dawley Rats. Neurochem Res 2018; 43:918-929. [PMID: 29455417 DOI: 10.1007/s11064-018-2497-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 02/04/2018] [Accepted: 02/07/2018] [Indexed: 02/07/2023]
Abstract
We previously reported that Yulangsan polysaccharide (YLSP), which was isolated from the root of Millettia pulchra Kurz, attenuates withdrawal symptoms of morphine dependence by regulating the nitric oxide pathway and modulating monoaminergic neurotransmitters. In this study, we investigated the effects and mechanism of YLSP on the reinstatement of morphine-induced conditioned place preference (CPP) in rats. A CPP procedure was employed to assess the behavior of rats, and indicators of serum and four brain regions (nucleus accumbens, ventral tegmental area, hippocampus and prefrontal cortex) were determined to explore its underlying mechanism. YLSP inhibited priming morphine-induced reinstatement of CPP in a dose-dependent manner. YLSP markedly reduced nitric oxide and nitric oxide synthase levels in the brain. Moreover, YLSP significantly decreased the dopamine and norepinephrine levels in the serum and brain. Furthermore, YLSP significantly decreased cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) concentrations, inhibited the expression of dopamine D1 receptors and cAMP response element binding protein mRNA, and improved the expression of dopamine D2 receptor mRNA in the four brain regions. Our findings indicated that YLSP could inhibit the reinstatement of morphine-induced CPP possibly by modulating the NO-cGMP and D1R-cAMP signaling pathways.
Collapse
Affiliation(s)
- Chunxia Chen
- Department of Pharmacology, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, People's Republic of China.,Department of Hyperbaric Oxygen, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, People's Republic of China
| | - Zhihuan Nong
- Department of Pharmacology, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, People's Republic of China
| | - Xingmei Liang
- Department of Pharmacology, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, People's Republic of China
| | - Mingyu Meng
- Department of Pharmacology, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, People's Republic of China
| | - Feifei Xuan
- Department of Pharmacology, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, People's Republic of China
| | - Qiuqiao Xie
- Department of Pharmacology, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, People's Republic of China
| | - Junhui He
- Department of Pharmacology, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, People's Republic of China
| | - Renbin Huang
- Department of Pharmacology, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, People's Republic of China.
| |
Collapse
|
22
|
Kim J, Ham S, Hong H, Moon C, Im HI. Brain Reward Circuits in Morphine Addiction. Mol Cells 2016; 39:645-53. [PMID: 27506251 PMCID: PMC5050528 DOI: 10.14348/molcells.2016.0137] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 07/18/2016] [Accepted: 07/20/2016] [Indexed: 12/30/2022] Open
Abstract
Morphine is the most potent analgesic for chronic pain, but its clinical use has been limited by the opiate's innate tendency to produce tolerance, severe withdrawal symptoms and rewarding properties with a high risk of relapse. To understand the addictive properties of morphine, past studies have focused on relevant molecular and cellular changes in the brain, highlighting the functional roles of reward-related brain regions. Given the accumulated findings, a recent, emerging trend in morphine research is that of examining the dynamics of neuronal interactions in brain reward circuits under the influence of morphine action. In this review, we highlight recent findings on the roles of several reward circuits involved in morphine addiction based on pharmacological, molecular and physiological evidences.
Collapse
Affiliation(s)
- Juhwan Kim
- Center for Neuroscience, Brain Science Institute, Seoul 02792,
Korea
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul 02792,
Korea
- Department of Veterinary Anatomy, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 61186,
Korea
| | - Suji Ham
- Center for Neuroscience, Brain Science Institute, Seoul 02792,
Korea
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul 02792,
Korea
- Department of Neuroscience, Korea University of Science and Technology (UST), Daejeon 34113,
Korea
| | - Heeok Hong
- Department of Medical Science, Konkuk University School of Medicine, Seoul 05029,
Korea
| | - Changjong Moon
- Department of Veterinary Anatomy, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 61186,
Korea
| | - Heh-In Im
- Center for Neuroscience, Brain Science Institute, Seoul 02792,
Korea
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul 02792,
Korea
- Department of Neuroscience, Korea University of Science and Technology (UST), Daejeon 34113,
Korea
| |
Collapse
|