1
|
Sahebi K, Arianejad M, Azadi S, Hosseinpour-Soleimani F, Kazemi R, Tajbakhsh A, Negahdaripour M. The interplay between gut microbiome, epigenetics, and substance use disorders: from molecular to clinical perspectives. Eur J Pharmacol 2025; 998:177630. [PMID: 40252900 DOI: 10.1016/j.ejphar.2025.177630] [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: 12/26/2024] [Revised: 03/27/2025] [Accepted: 04/15/2025] [Indexed: 04/21/2025]
Abstract
Substance use disorders (SUDs) involve a complex series of central and peripheral pathologies, leading to impairments in cognitive, behavioral, and physiological processes. Emerging evidence indicates a more significant role for the microbiome-gut-brain axis (MGBA) in SUDs than previously recognized. The MGBA is interconnected with various body systems by producing numerous metabolites, most importantly short-chain fatty acids (SCFAs), cytokines, and neurotransmitters. These mediators influence the human body's epigenome and transcriptome. While numerous epigenetic alterations in different brain regions have been reported in SUD models, the intricate relationship between SUDs and the MGBA suggests that the gut microbiome may partially contribute to the underlying mechanisms of SUDs. Promising results have been observed with gut microbiome-directed interventions in patients with SUDs, including prebiotics, probiotics, antibiotics, and fecal microbiota transplantation. Nonetheless, the long-term epigenetic effects of these interventions remain unexplored. Moreover, various confounding factors and study limitations have hindered the identification of molecular mechanisms and clinical applications of gut microbiome interventions in SUDs. In the present review, we will (i) provide a comprehensive discussion on how the gut microbiome influences SUDs, with an emphasis on epigenetic alterations; (ii) discuss the current evidence on the bidirectional relationship of gut microbiome and SUDs, highlighting potential targets for intervention; and (iii) review recent advances in gut microbiome-directed therapies, along with their limitations and future directions.
Collapse
Affiliation(s)
- Keivan Sahebi
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mona Arianejad
- Department of Molecular Medicine, School of Advanced Technologies of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Soha Azadi
- Department of Clinical Pharmacy, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Hosseinpour-Soleimani
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Applied Cell Sciences and Tissue Engineering, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Radmehr Kazemi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Manica Negahdaripour
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
2
|
Yunusa S, Müller CP, Hassan Z. Mitragynine (Kratom)-Withdrawal behaviour and cognitive impairments can be ameliorated by an epigenetic mechanism. Br J Pharmacol 2024; 181:2070-2084. [PMID: 38523471 DOI: 10.1111/bph.16352] [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: 08/20/2023] [Revised: 12/11/2023] [Accepted: 01/02/2024] [Indexed: 03/26/2024] Open
Abstract
BACKGROUND AND PURPOSE Kratom is a preparation from Mitragyna speciosa, which is used as a natural drug preparation for many purposes around the world. However, an overdose of Kratom may cause addiction-like problems including aversive withdrawal states resulting in cognitive impairments via unknown mechanisms. Its main psychoactive alkaloid is mitragynine, showing opioid-like properties. EXPERIMENTAL APPROACH Here, we analysed the neuropharmacological effects of mitragynine compared with morphine withdrawal in rats and searched for a pharmacological treatment option that may reverse the occurring cognitive deficits that usually aggravate withdrawal. KEY RESULTS We found that withdrawal from 14-day mitragynine (1-10 mg·kg-1·day-1) treatment caused dose-dependent behavioural withdrawal signs resembling those of morphine (5 mg·kg-1·day-1) withdrawal. However, mitragynine (5 and 10 mg·kg-1·day-1) withdrawal also induced impairments in a passive avoidance task. Mitragynine withdrawal not only reduced hippocampal field excitatory postsynaptic potential (fEPSP) amplitudes in basal synaptic transmission and long-term potentiation (LTP) but also reduced epigenetic markers, such as histone H3K9 and H4K12 expression. At the same time, it up-regulates HDAC2 expression. Targeting the epigenetic adaptations with the HDAC inhibitor, SAHA, reversed the effects of mitragynine withdrawal on epigenetic dysregulation, hippocampal input/output curves, paired-pulse facilitation, LTP and attenuated the cognitive deficit. However, SAHA amplified the effects of morphine withdrawal. CONCLUSION AND IMPLICATIONS The data from this work show that changes in histone expression and downstream hippocampal plasticity may explain mitragynine, but not morphine, withdrawal behaviours and cognitive impairments. Thus, it may provide a new treatment approach for aversive Kratom/mitragynine withdrawal and addiction.
Collapse
Affiliation(s)
- Suleiman Yunusa
- Centre for Drug Research, Universiti Sains Malaysia, Penang, Malaysia
- Department of Pharmacology, Bauchi State University Gadau, Bauchi State, Nigeria
| | - Christian P Müller
- Centre for Drug Research, Universiti Sains Malaysia, Penang, Malaysia
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
- Institute of Psychopharmacology, Central Institute of Mental Health, Faculty of Medicine Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Zurina Hassan
- Centre for Drug Research, Universiti Sains Malaysia, Penang, Malaysia
| |
Collapse
|
3
|
Jammoul M, Jammoul D, Wang KK, Kobeissy F, Depalma RG. Traumatic Brain Injury and Opioids: Twin Plagues of the Twenty-First Century. Biol Psychiatry 2024; 95:6-14. [PMID: 37217015 DOI: 10.1016/j.biopsych.2023.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 04/22/2023] [Accepted: 05/12/2023] [Indexed: 05/24/2023]
Abstract
Traumatic brain injury (TBI) and opioid use disorder (OUD) comprise twin plagues causing considerable morbidity and mortality worldwide. As interactions between TBI and OUD are to our knowledge uncharted, we review the possible mechanisms by which TBI may stimulate the development of OUD and discuss the interaction or crosstalk between these two processes. Central nervous system damage due to TBI appears to drive adverse effects of subsequent OUD and opioid use/misuse affecting several molecular pathways. Pain, a neurological consequence of TBI, is a risk factor that increases the likelihood of opioid use/misuse after TBI. Other comorbidities including depression, anxiety, posttraumatic stress disorder, and sleep disturbances are also associated with deleterious outcomes. We examine the hypothesis that a TBI "first hit" induces a neuroinflammatory process involving microglial priming, which, on a second hit related to opioid exposure, exacerbates neuroinflammation, modifies synaptic plasticity, and spreads tau aggregates to promote neurodegeneration. As TBI also impairs myelin repair by oligodendrocytes, it may reduce or degrade white matter integrity in the reward circuit resulting in behavioral changes. Along with approaches focused on specific patient symptoms, understanding the CNS effects following TBI offers a promise of improved management for individuals with OUD.
Collapse
Affiliation(s)
- Maya Jammoul
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Dareen Jammoul
- Anesthesiology Department, Lebanese American University Medical Center-Rizk Hospital, Beirut, Lebanon
| | - Kevin K Wang
- Center for Neurotrauma, MultiOmics & Biomarkers, Department of Neurobiology, Morehouse School of Medicine, Atlanta, Georgia; Department of Emergency Medicine, University of Florida, Gainesville, Florida.
| | - Firas Kobeissy
- Center for Neurotrauma, MultiOmics & Biomarkers, Department of Neurobiology, Morehouse School of Medicine, Atlanta, Georgia; Department of Emergency Medicine, University of Florida, Gainesville, Florida; Faculty of Medicine, Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon.
| | - Ralph G Depalma
- Office of Research and Development, Department of Veterans Affairs, Washington, DC; Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
4
|
King'uyu DN, Nti-Kyemereh L, Bonin JL, Feustel PJ, Tram M, MacNamara KC, Kopec AM. The effect of morphine on rat microglial phagocytic activity: An in vitro study of brain region-, plating density-, sex-, morphine concentration-, and receptor-dependency. J Neuroimmunol 2023; 384:578204. [PMID: 37774553 DOI: 10.1016/j.jneuroim.2023.578204] [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: 04/07/2023] [Revised: 07/24/2023] [Accepted: 09/19/2023] [Indexed: 10/01/2023]
Abstract
Opioids have long been used for clinical pain management, but also have addictive properties that have contributed to the ongoing opioid epidemic. While opioid activation of opioid receptors is well known to contribute to reward and reinforcement, data now also suggest that opioid activation of immune signaling via toll-like receptor 4 (TLR4) may also play a role in addiction-like processes. TLR4 expression is enriched in immune cells, and in the nervous system is primarily expressed in microglia. Microglial phagocytosis is important for developmental, homeostatic, and pathological processes. To examine how morphine impacts microglial phagocytosis, we isolated microglia from adult male and female rat cortex and striatum and plated them in vitro at 10,000 (10K) or 50,000 cells/well densities. Microglia were incubated with neutral fluorescent microbeads to stimulate phagocytosis in the presence of one of four morphine concentrations. We found that the brain region from which microglia are isolated and plating density, but not morphine concentration, impacts cell survival in vitro. We found that 10-12 M morphine, but not higher concentrations, increases phagocytosis in striatal microglia in vitro independent of sex and plating density, while 10-12 M morphine increased phagocytosis in cortical microglia in vitro independent of sex, but contingent on a plating density. Finally, we demonstrate that the effect of 10-12 M morphine in striatal microglia plated at 10 K density is mediated via TLR4, and not μORs. Overall, our data suggest that in rats, a morphine-TLR4 signaling pathway increases phagocytic activity in microglia independent of sex. This may is useful information for better understanding the possible neural outcomes associated with morphine exposures.
Collapse
Affiliation(s)
- David N King'uyu
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY 12208, United States of America.
| | - Lily Nti-Kyemereh
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY 12208, United States of America; Siena College, Loudonville, NY 12211, United States of America
| | - Jesse L Bonin
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, United States of America
| | - Paul J Feustel
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY 12208, United States of America
| | - Michelle Tram
- Siena College, Loudonville, NY 12211, United States of America
| | - Katherine C MacNamara
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, United States of America
| | - Ashley M Kopec
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY 12208, United States of America
| |
Collapse
|
5
|
Kolli U, Roy S. The role of the gut microbiome and microbial metabolism in mediating opioid-induced changes in the epigenome. Front Microbiol 2023; 14:1233194. [PMID: 37670983 PMCID: PMC10475585 DOI: 10.3389/fmicb.2023.1233194] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 07/26/2023] [Indexed: 09/07/2023] Open
Abstract
The current opioid pandemic is a major public health crisis in the United States, affecting millions of people and imposing significant health and socioeconomic burdens. Preclinical and clinical research over the past few decades has delineated certain molecular mechanisms and identified various genetic, epigenetic, and environmental factors responsible for the pathophysiology and comorbidities associated with opioid use. Opioid use-induced epigenetic modifications have been identified as one of the important factors that mediate genetic changes in brain regions that control reward and drug-seeking behavior and are also implicated in the development of tolerance. Recently, it has been shown that opioid use results in microbial dysbiosis, leading to gut barrier disruption, which drives systemic inflammation, impacting the perception of pain, the development of analgesic tolerance, and behavioral outcomes. In this review, we highlight the potential role of microbiota and microbial metabolites in mediating the epigenetic modifications induced by opioid use.
Collapse
Affiliation(s)
| | - Sabita Roy
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
6
|
Kalamarides DJ, Singh A, Wolfman SL, Dani JA. Sex differences in VTA GABA transmission and plasticity during opioid withdrawal. Sci Rep 2023; 13:8460. [PMID: 37231124 DOI: 10.1038/s41598-023-35673-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 05/19/2023] [Indexed: 05/27/2023] Open
Abstract
The effectiveness of current treatments for opioid use disorder (OUD) varies by sex. Our understanding of the neurobiological mechanisms mediating negative states during withdrawal is lacking, particularly with regard to sex differences. Based on preclinical research in male subjects, opioid withdrawal is accompanied by increased gamma-aminobutyric acid (GABA) release probability at synapses onto dopamine neurons in the ventral tegmental area (VTA). It is unclear, however, if the physiological consequences of morphine that were originally elucidated in male rodents extend to females. The effects of morphine on the induction of future synaptic plasticity are also unknown. Here, we show that inhibitory synaptic long-term potentiation (LTPGABA) is occluded in the VTA in male mice after repeated morphine injections and 1 day of withdrawal, while morphine-treated female mice maintain the ability to evoke LTPGABA and have basal GABA activity similar to controls. Our observation of this physiological difference between male and female mice connects previous reports of sex differences in areas upstream and downstream of the GABA-dopamine synapse in the VTA during opioid withdrawal. The sex differences highlight the mechanistic distinctions between males and females that can be targeted when designing and implementing treatments for OUD.
Collapse
Affiliation(s)
- Daniel J Kalamarides
- Department of Neuroscience, Perelman School of Medicine, Mahoney Institute for Neurosciences, University of Pennsylvania, 415 Curie Blvd, Philadelphia, PA, 19104, USA
| | - Aditi Singh
- Department of Neuroscience, Perelman School of Medicine, Mahoney Institute for Neurosciences, University of Pennsylvania, 415 Curie Blvd, Philadelphia, PA, 19104, USA
| | - Shannon L Wolfman
- Department of Neuroscience, Perelman School of Medicine, Mahoney Institute for Neurosciences, University of Pennsylvania, 415 Curie Blvd, Philadelphia, PA, 19104, USA
| | - John A Dani
- Department of Neuroscience, Perelman School of Medicine, Mahoney Institute for Neurosciences, University of Pennsylvania, 415 Curie Blvd, Philadelphia, PA, 19104, USA.
| |
Collapse
|
7
|
Nikbakhtzadeh M, Ashabi G, Saadatyar R, Doostmohammadi J, Nekoonam S, Keshavarz M, Riahi E. Restoring the firing activity of ventral tegmental area neurons by lateral hypothalamic deep brain stimulation following morphine administration in rats: LH DBS and the spiking activity of VTA neurons. Physiol Behav 2023; 267:114209. [PMID: 37105347 DOI: 10.1016/j.physbeh.2023.114209] [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: 03/06/2023] [Revised: 04/12/2023] [Accepted: 04/24/2023] [Indexed: 04/29/2023]
Abstract
We have previously shown that high-frequency deep brain stimulation (DBS) of the lateral hypothalamus (LH) compromises morphine-induced addiction-like behavior in rats. The exact mechanism underlying this effect is not known. Here, we investigated the assumption that DBS in the LH influences the firing activity of neurons in the ventral tegmental area (VTA). To that end, male Wistar rats received morphine (5 mg/kg; s.c.) for three days and underwent extracellular single unit recording under general anesthesia one day later. During the recording, the rats received an intraoperative injection of morphine (5 mg/kg; s.c.) plus DBS in the LH (130 Hz pulse frequency, 150 μA amplitude, and 100 μs pulse width). One group of animals also received preoperative DBS after each morphine injection before the recording. The spiking frequency of VTA neurons was measured at three successive phases: (1) baseline (5-15 min); (2) DBS-on (morphine + DBS for 30 min); and (3) After-DBS (over 30 min after termination of DBS). Results showed that morphine suppressed the firing activity of a large population of non-DA neurons, whereas it activated most DA neurons. Intraoperative DBS reversed morphine suppression of non-DA firing, but did not alter the excitatory effect of morphine on DA neurons firing. With repeated preoperative application of DBS, non-DA neurons returned to the morphine-induced suppressive state, but DA neurons released from the excitatory effect of morphine. It is concluded that the development of morphine reward is associated with a hypoactivity of VTA non-DA neurons and a hyperactivity of DA neurons, and that DBS modulation of the spiking activity may contribute to the blockade of morphine addiction-like behavior.
Collapse
Affiliation(s)
- Marjan Nikbakhtzadeh
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghorbangol Ashabi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Saadatyar
- Department of Biomedical Engineering, School of Electrical Engineering, Iran University of Science and Technology, Tehran, Iran
| | - Jafar Doostmohammadi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saied Nekoonam
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mansoor Keshavarz
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Esmail Riahi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Ostlund I, Von Gunten M, Smith C, Edwards JG. Chronic Δ9-tetrahydrocannabinol impact on plasticity, and differential activation requirement for CB1-dependent long-term depression in ventral tegmental area GABA neurons in adult versus young mice. Front Neurosci 2023; 16:1067493. [PMID: 36699526 PMCID: PMC9869137 DOI: 10.3389/fnins.2022.1067493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/12/2022] [Indexed: 01/11/2023] Open
Abstract
The ventral tegmental area (VTA) mediates incentive salience and reward prediction error through dopamine (DA) neurons that are regulated by local VTA GABA neurons. In young mice, VTA GABA cells exhibit a form of synaptic plasticity known as long-term depression (LTD) that is dependent on cannabinoid 1 (CB1) receptors preceded by metabotropic glutamate receptor 5 (mGluR5) signaling to induce endocannabinoid production. This LTD was eliminated following chronic (7-10 consecutive days) exposure to the marijuana derived cannabinoid Δ9 -tetrahydrocannabinol (THC). We now examine the mechanism behind THC-induced elimination of LTD in adolescents as well as plasticity induction ability in adult versus young male and female mice using whole-cell electrophysiology experiments of VTA GABA cells. Chronic THC injections in adolescents resulted in a loss of CB1 agonist-mediated depression, illustrating chronic THC likely desensitizes or removes synaptic CB1. We noted that seven days withdrawal from chronic THC restored LTD and CB1 agonist-induced depression, suggesting reversibility of THC-induced changes. Adult mice continue to express functional mGluR5 and CB1, but require a doubling of the synaptic stimulation compared to young mice to induce LTD, suggesting a quantitative difference in CB1-dependent plasticity between young and adult mice. One potential rationale for this difference is changes in AMPA and NMDA glutamate receptors. Indeed, AMPA/NMDA ratios were increased in in adults compared to young mice. Lastly, we performed quantitative reverse-transcription PCR and identified that CB1, DAGLα, and GluA1 levels increased following chronic THC exposure. Collectively, our data demonstrate the first age-dependent GABA neuron plasticity in the VTA, which could have implications for decreased THC dependence capacity in adults, as well as the mechanism behind chronic THC-induced synaptic alterations in young mice.
Collapse
Affiliation(s)
- Isaac Ostlund
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT, United States
| | | | - Calvin Smith
- Neuroscience Center, Brigham Young University, Provo, UT, United States
| | - Jeffrey G. Edwards
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT, United States
- Neuroscience Center, Brigham Young University, Provo, UT, United States
| |
Collapse
|
9
|
Simmons SC, Grecco GG, Atwood BK, Nugent FS. Effects of prenatal opioid exposure on synaptic adaptations and behaviors across development. Neuropharmacology 2023; 222:109312. [PMID: 36334764 PMCID: PMC10314127 DOI: 10.1016/j.neuropharm.2022.109312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
Abstract
In this review, we focus on prenatal opioid exposure (POE) given the significant concern for the mental health outcomes of children with parents affected by opioid use disorder (OUD) in the view of the current opioid crisis. We highlight some of the less explored interactions between developmental age and sex on synaptic plasticity and associated behavioral outcomes in preclinical POE research. We begin with an overview of the rich literature on hippocampal related behaviors and plasticity across POE exposure paradigms. We then discuss recent work on reward circuit dysregulation following POE. Additional risk factors such as early life stress (ELS) could further influence synaptic and behavioral outcomes of POE. Therefore, we include an overview on the use of preclinical ELS models where ELS exposure during key critical developmental periods confers considerable vulnerability to addiction and stress psychopathology. Here, we hope to highlight the similarity between POE and ELS on development and maintenance of opioid-induced plasticity and altered opioid-related behaviors where similar enduring plasticity in reward circuits may occur. We conclude the review with some of the limitations that should be considered in future investigations. This article is part of the Special Issue on 'Opioid-induced addiction'.
Collapse
Affiliation(s)
- Sarah C Simmons
- Department of Pharmacology and Molecular Therapeutics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Greg G Grecco
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA; Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Brady K Atwood
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Fereshteh S Nugent
- Department of Pharmacology and Molecular Therapeutics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
10
|
Chen Y, Wang X, Xiao M, Kang N, Zeng W, Zhang J. Prenatal morphine exposure increases gamma oscillation and theta coherence in the rat reward system. Neurotoxicology 2022; 90:246-255. [DOI: 10.1016/j.neuro.2022.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/13/2022] [Accepted: 04/16/2022] [Indexed: 10/18/2022]
|
11
|
Zhang W, Zheng X, Gong Y, Jiang T, Qiu J, Wu X, Lu F, Wang Z, Hong Z. VX-11e protects articular cartilage and subchondral bone in osteoarthritis by inhibiting the RIP1/RIP3/MLKL and MAPK signaling pathways. Bioorg Chem 2022; 120:105632. [DOI: 10.1016/j.bioorg.2022.105632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/16/2022] [Indexed: 12/18/2022]
|
12
|
Addiction-induced plasticity in underlying neural circuits. Neurol Sci 2022; 43:1605-1615. [DOI: 10.1007/s10072-021-05778-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/20/2021] [Indexed: 10/19/2022]
|
13
|
Fan X, Shi G, He X, Li X, Wan Y, Jian L. Oxytocin prevents cue-induced reinstatement of oxycodone seeking: Involvement of DNA methylation in the hippocampus. Addict Biol 2021; 26:e13025. [PMID: 33609013 DOI: 10.1111/adb.13025] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/26/2021] [Accepted: 02/05/2021] [Indexed: 12/20/2022]
Abstract
Oxycodone is one of the most commonly used analgesics in the clinic. However, long-term use can contribute to drug dependence. Accumulating evidence of changes in DNA methylation after opioid relapse has provided insight into mechanisms underlying drug-associated memory. The neuropeptide oxytocin is reported to be a potential treatment for addiction. The present study sought to identify changes in global and synaptic gene methylation after cue-induced reinstatement of oxycodone conditioned place preference (CPP) and the effect of oxytocin. We analyzed hippocampal mRNA of synaptic genes and also synaptic density in response to oxycodone CPP. We determined the mRNA levels of DNA methyltransferases (Dnmts) and ten-eleven translocations (Tets), observed global 5-methylcytosine (5-mC) and 5-hydroxymethylcytosine (5-hmC) levels, and measured DNA methylation status of four synaptic genes implicated in learning and memory (Arc, Dlg1, Dlg4, and Syn1). Both synaptic density and the transcription of 15 hippocampal synaptic genes significantly increased following cue-induced reinstatement of oxycodone CPP. Oxycodone relapse was also related to markedly decreased 5-mC levels and decreased transcription of Dnmt1, Dnmt3a, and Dnmt3b; in contrast, 5-hmC levels and the transcription of Tet1 and Tet3 were increased. Oxycodone exposure induced DNA hypomethylation at the exons of the Arc, Dlg1, Dlg4, and Syn1 genes. Intracerebroventricular (ICV) administration of oxytocin (2.5 μg/μl) specifically blocked oxycodone relapse, possibly by inhibition of Arc, Dlg1, Dlg4, and Syn1 hypomethylation in oxycodone-treated rats. Together, these data indicate the occurrence of epigenetic changes in the hippocampus following oxycodone relapse and the potential role of oxytocin in oxycodone addiction.
Collapse
Affiliation(s)
- Xin‐Yu Fan
- Department of Pharmacy Shengjing Hospital of China Medical University Shenyang China
| | - Guang Shi
- Department of Neurology, People's Hospital of Liaoning Province Shenyang China
| | - Xiao‐Jing He
- Department of Pharmacy Shengjing Hospital of China Medical University Shenyang China
| | - Xin‐Yang Li
- Department of Pharmacy Shengjing Hospital of China Medical University Shenyang China
| | - Yu‐Xiao Wan
- Department of Anesthesiology Shengjing Hospital of China Medical University Shenyang China
| | - Ling‐Yan Jian
- Department of Pharmacy Shengjing Hospital of China Medical University Shenyang China
| |
Collapse
|
14
|
Abstract
Neuroepigenetics, a new branch of epigenetics, plays an important role in the regulation of gene expression. Neuroepigenetics is associated with holistic neuronal function and helps in formation and maintenance of memory and learning processes. This includes neurodevelopment and neurodegenerative defects in which histone modification enzymes appear to play a crucial role. These modifications, carried out by acetyltransferases and deacetylases, regulate biologic and cellular processes such as apoptosis and autophagy, inflammatory response, mitochondrial dysfunction, cell-cycle progression and oxidative stress. Alterations in acetylation status of histone as well as non-histone substrates lead to transcriptional deregulation. Histone deacetylase decreases acetylation status and causes transcriptional repression of regulatory genes involved in neural plasticity, synaptogenesis, synaptic and neural plasticity, cognition and memory, and neural differentiation. Transcriptional deactivation in the brain results in development of neurodevelopmental and neurodegenerative disorders. Mounting evidence implicates histone deacetylase inhibitors as potential therapeutic targets to combat neurologic disorders. Recent studies have targeted naturally-occurring biomolecules and micro-RNAs to improve cognitive defects and memory. Multi-target drug ligands targeting HDAC have been developed and used in cell-culture and animal-models of neurologic disorders to ameliorate synaptic and cognitive dysfunction. Herein, we focus on the implications of histone deacetylase enzymes in neuropathology, their regulation of brain function and plausible involvement in the pathogenesis of neurologic defects.
Collapse
|
15
|
Cheron J, Kerchove d'Exaerde AD. Drug addiction: from bench to bedside. Transl Psychiatry 2021; 11:424. [PMID: 34385417 PMCID: PMC8361217 DOI: 10.1038/s41398-021-01542-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/14/2021] [Accepted: 07/23/2021] [Indexed: 02/06/2023] Open
Abstract
Drug addiction is responsible for millions of deaths per year around the world. Still, its management as a chronic disease is shadowed by misconceptions from the general public. Indeed, drug consumers are often labelled as "weak", "immoral" or "depraved". Consequently, drug addiction is often perceived as an individual problem and not societal. In technical terms, drug addiction is defined as a chronic, relapsing disease resulting from sustained effects of drugs on the brain. Through a better characterisation of the cerebral circuits involved, and the long-term modifications of the brain induced by addictive drugs administrations, first, we might be able to change the way the general public see the patient who is suffering from drug addiction, and second, we might be able to find new treatments to normalise the altered brain homeostasis. In this review, we synthetise the contribution of fundamental research to the understanding drug addiction and its contribution to potential novel therapeutics. Mostly based on drug-induced modifications of synaptic plasticity and epigenetic mechanisms (and their behavioural correlates) and after demonstration of their reversibility, we tried to highlight promising therapeutics. We also underline the specific temporal dynamics and psychosocial aspects of this complex psychiatric disease adding parameters to be considered in clinical trials and paving the way to test new therapeutic venues.
Collapse
Affiliation(s)
- Julian Cheron
- Laboratory of Neurophysiology, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), Brussels, B-1070, Belgium
| | - Alban de Kerchove d'Exaerde
- Laboratory of Neurophysiology, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), Brussels, B-1070, Belgium.
| |
Collapse
|
16
|
Effect of histone acetylation on maintenance and reinstatement of morphine-induced conditioned place preference and ΔFosB expression in the nucleus accumbens and prefrontal cortex of male rats. Behav Brain Res 2021; 414:113477. [PMID: 34302880 DOI: 10.1016/j.bbr.2021.113477] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/10/2021] [Accepted: 07/16/2021] [Indexed: 01/14/2023]
Abstract
Recently, epigenetic mechanisms are considered as the new potential targets for addiction treatment. This research was designed to explore the effect of histone acetylation on ΔFosB gene expression in morphine-induced conditioned place preference (CPP) in male rats. CPP was induced via morphine injection (5 mg/kg) for three consecutive days. Animals received low-dose theophylline (LDT) or Suberoylanilide Hydroxamic acid (SAHA), as an histone deacetylase (HDAC) activator or inhibitor, respectively, and a combination of both in subsequent extinction days. Following extinction, a priming dose of morphine (1 mg/kg) was administered to induce reinstatement. H4 acetylation and ΔFosB expression in the nucleus accumbens (NAc) and medial prefrontal cortex (mPFC) were assessed on the last day of extinction and the following CPP reinstatement. Our results demonstrated that daily administration of SAHA (25 mg/kg; i.p.), facilitated morphine-extinction and decreased CPP score in reinstatement of place preference. Conversely, injections of LDT (20 mg/kg; i.p.) prolonged extinction in animals. Co-administration of LDT and SAHA on extinction days counterbalanced each other, such that maintenance and reinstatement were no different than the control group. The gene expression of ΔFosB was increased by SAHA in NAc and mPFC compared to the control group. Administration of SAHA during extinction days, also altered histone acetylation in the NAc and mPFC on the last day of extinction, but not on reinstatement day. Collectively, administration of SAHA facilitated extinction and reduced reinstatement of morphine-induced CPP in rats. This study confirms the essential role of epigenetic mechanisms, specifically histone acetylation, in regulating drug-induced plasticity and seeking behaviors.
Collapse
|
17
|
Ahmadi S, Zobeiri M, Mohammadi Talvar S, Masoudi K, Khanizad A, Fotouhi S, Bradburn S. Differential expression of H19, BC1, MIAT1, and MALAT1 long non-coding RNAs within key brain reward regions after repeated morphine treatment. Behav Brain Res 2021; 414:113478. [PMID: 34302875 DOI: 10.1016/j.bbr.2021.113478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/22/2021] [Accepted: 07/19/2021] [Indexed: 12/14/2022]
Abstract
Morphine-induced analgesic tolerance and dependence are significant limits of pain control; however, the exact molecular mechanisms underlying morphine tolerance and dependence have remained unclear. The role of long non-coding RNAs (lncRNAs) in morphine tolerance and dependence is yet to be determined. We aimed to explore the association of specific lncRNAs expression in key brain reward regions after repeated injection of morphine. Male Wistar rats received subcutaneous injections of twice-daily morphine (10 mg/kg) or saline (1 mL/kg) for eight days. On day 8 of the repeated injections, induction of morphine analgesic tolerance and dependence was confirmed through a hotplate test and a naloxone-precipitated withdrawal analysis, respectively. Expression of H19, BC1, MIAT1, and MALAT1 lncRNAs was determined from the midbrain, striatum, hypothalamus, prefrontal cortex (PFC), and hippocampus by real-time PCR on day 8 of the repeated injections. The H19 expression was significantly different between morphine-treated and control saline-treated rats in all investigated areas except for the hippocampus. The BC1 expression significantly altered in the midbrain, hypothalamus, and hippocampus, but not in the striatum and PFC after repeated morphine treatment. The MIAT1 and MALAT1 expression site-specifically altered in the midbrain, hypothalamus, and striatum; however, no significant changes were detected in their expression in the PFC and hippocampus after repeated morphine treatment. We conclude that alterations in the expression of these lncRNAs in the brain reward regions especially in the midbrain, striatum and hypothalamus may have critical roles in the development of morphine dependence and tolerance, which need to be considered in future researches.
Collapse
Affiliation(s)
- Shamseddin Ahmadi
- Department of Biological Science, Faculty of Science, University of Kurdistan, Sanandaj, Iran.
| | - Mohammad Zobeiri
- Department of Biological Science, Faculty of Science, University of Kurdistan, Sanandaj, Iran
| | - Shiva Mohammadi Talvar
- Department of Biological Science, Faculty of Science, University of Kurdistan, Sanandaj, Iran
| | - Kayvan Masoudi
- Department of Biological Science, Faculty of Science, University of Kurdistan, Sanandaj, Iran
| | - Amir Khanizad
- Department of Biological Science, Faculty of Science, University of Kurdistan, Sanandaj, Iran
| | - Shima Fotouhi
- Department of Biological Science, Faculty of Science, University of Kurdistan, Sanandaj, Iran
| | - Steven Bradburn
- Department of Life Sciences, Bioscience Research Centre, Manchester Metropolitan University, Manchester, UK
| |
Collapse
|
18
|
Carpenter MD, Manners MT, Heller EA, Blendy JA. Adolescent oxycodone exposure inhibits withdrawal-induced expression of genes associated with the dopamine transmission. Addict Biol 2021; 26:e12994. [PMID: 33325096 DOI: 10.1111/adb.12994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 10/09/2020] [Accepted: 11/12/2020] [Indexed: 12/21/2022]
Abstract
Prescription opioid misuse is a major public health concern among children and adolescents in the United States. Opioids are the most commonly abused drugs and are the fastest growing drug problem among adolescents. In humans and animals, adolescence is a particularly sensitive period associated with an increased response to drugs of abuse. Our previous studies indicate that oxycodone exposure during adolescence increases morphine reward in adulthood. How early drug exposure mediates long-term changes in the brain and behavior is not known, but epigenetic regulation is a likely mechanism. To address this question, we exposed mice to oxycodone or saline during adolescence and examined epigenetic modifications at genes associated with dopamine activity during adulthood at early and late withdrawal, in the ventral tegmental area (VTA). We then compared these with alterations in the VTA of adult-treated mice following an equivalent duration of exposure and withdrawal to determine if the effects of oxycodone are age dependent. We observed persistence of adolescent-like gene expression following adolescent oxycodone exposure relative to age-matched saline exposed controls, although dopamine-related gene expression was transiently activated at 1 day of withdrawal. Following prolonged withdrawal enrichment of the repressive histone mark, H3K27me3, was maintained, consistent with inhibition of gene regulation following adolescent exposure. By contrast, mice exposed to oxycodone as adults showed loss of the repressive mark and increased gene expression following 28 days of withdrawal following oxycodone exposure. Together, our findings provide evidence that adolescent oxycodone exposure has long-term epigenetic consequences in VTA of the developing brain.
Collapse
Affiliation(s)
- Marco D. Carpenter
- Department of Systems Pharmacology and Translational Therapeutics University of Pennsylvania Philadelphia Pennsylvania USA
- Institute for Translational Medicine and Therapeutics University of Pennsylvania Philadelphia Pennsylvania USA
- Penn Epigenetics Institute, Perelman School of Medicine University of Pennsylvania Philadelphia Pennsylvania USA
| | - Melissa T. Manners
- Department of Systems Pharmacology and Translational Therapeutics University of Pennsylvania Philadelphia Pennsylvania USA
- Department of Biological Sciences University of the Sciences Philadelphia Pennsylvania USA
| | - Elizabeth A. Heller
- Department of Systems Pharmacology and Translational Therapeutics University of Pennsylvania Philadelphia Pennsylvania USA
- Institute for Translational Medicine and Therapeutics University of Pennsylvania Philadelphia Pennsylvania USA
- Penn Epigenetics Institute, Perelman School of Medicine University of Pennsylvania Philadelphia Pennsylvania USA
| | - Julie A. Blendy
- Department of Systems Pharmacology and Translational Therapeutics University of Pennsylvania Philadelphia Pennsylvania USA
- Institute for Translational Medicine and Therapeutics University of Pennsylvania Philadelphia Pennsylvania USA
| |
Collapse
|
19
|
Akbarabadi A, Sadat-Shirazi MS, Kabbaj M, Nouri Zadeh-Tehrani S, Khalifeh S, Pirri F, Zarrindast MR. Effects of Morphine and Maternal Care on Behaviors and Protein Expression of Male Offspring. Neuroscience 2021; 466:58-76. [PMID: 33915201 DOI: 10.1016/j.neuroscience.2021.04.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 01/26/2023]
Abstract
Genes and environment interact during development to alter gene expression and behavior. Parental morphine exposure before conception has devastating effects on the offspring. In the present study, we evaluated the role of maternal care in the intergenerational effect of maternal morphine exposure. Female rats received morphine or saline for ten days and were drugfree for another ten days. Thereafter, they were allowed to mate with drug-naïve male rats. When pups were born, they were cross-fostered to assess the contribution of maternal care versus morphine effects on the offspring. Adult male offspring were examined for anxiety-like behavior, spatial memory, and obsessive-compulsive-like behavior. To determine the mechanisms underlying the observed behavioral changes, protein levels of acetylated histone H3, BDNF, Trk-B, NMDA subunits, p-CREB, and 5-HT3R were measured in the brain. Our results indicate that maternal caregiving is impaired in morphine-abstinent mothers. Interestingly, maternal care behaviors were also affected in drug-naïve mothers that raised offspring of morphine-exposed mothers. In addition, the offspring of morphine abstinent and non-drug dependent mothers, when raised by morphine abstinent mothers, exhibited more anxiety, obsessive-compulsive behaviors and impaired spatial memory. These altered behaviors were associated with alterations in the levels of the above-mentioned proteins. These data illustrate the intergenerational effects of maternal morphine exposure on offspring behaviors. Moreover, exposure to morphine before gestation not only affects maternal care and offspring behavior, but also has negative consequences on behaviors and protein expression in adoptive mothers of affected offspring.
Collapse
Affiliation(s)
- Ardeshir Akbarabadi
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mohamed Kabbaj
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306-1270, United States; Program of Neuroscience, Florida State University, Tallahassee, FL 32306-1270, United States
| | | | - Solmaz Khalifeh
- Cognitive and Neuroscience Research Center (CNRC), Tehran Medical Sciences, Amir-Almomenin Hospital, Islamic Azad University, Tehran, Iran
| | - Fardad Pirri
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Reza Zarrindast
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran; Cognitive and Neuroscience Research Center (CNRC), Tehran Medical Sciences, Amir-Almomenin Hospital, Islamic Azad University, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Endocrinology and Metabolism Research Institute, Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
20
|
Doyle MA, Mazei-Robison MS. Opioid-Induced Molecular and Cellular Plasticity of Ventral Tegmental Area Dopamine Neurons. Cold Spring Harb Perspect Med 2021; 11:cshperspect.a039362. [PMID: 31964652 PMCID: PMC7371531 DOI: 10.1101/cshperspect.a039362] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Opioid drugs are highly valued as potent analgesics; however, there are significant risks associated with long-term use because of their abuse liability. Opioids cause changes in ventral tegmental area (VTA) gene expression and cell activity that have been linked to addiction-related behaviors in rodent models. Here, we focus on VTA dopamine (DA) neurons and review the cellular, structural, and synaptic plasticity changes induced by acute and chronic opioid exposure. We also discuss many avenues for future research including determination of whether opioid neuroadaptations are specific for subpopulations of VTA DA neurons. A better understanding of the molecular adaptations within the cells and circuits that drive opioid abuse is crucial for the development of better treatments for substance use disorders and to create novel, safer pain-relieving therapeutics.
Collapse
|
21
|
Shepard RD, Nugent FS. Early Life Stress- and Drug-Induced Histone Modifications Within the Ventral Tegmental Area. Front Cell Dev Biol 2020; 8:588476. [PMID: 33102491 PMCID: PMC7554626 DOI: 10.3389/fcell.2020.588476] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/14/2020] [Indexed: 12/11/2022] Open
Abstract
Psychiatric illnesses are a major public health concern due to their prevalence and heterogeneity of symptom presentation resulting from a lack of efficacious treatments. Although dysregulated dopamine (DA) signaling has been observed in a myriad of psychiatric conditions, different pathophysiological mechanisms have been implicated which impede the development of adequate treatments that work across all patient populations. The ventral tegmental area (VTA), a major source of DA neurons in the brain reward pathway, has been shown to have altered activity that contributes to reward dysregulation in mental illnesses and drug addiction. It has now become better appreciated that epigenetic mechanisms contribute to VTA DA dysfunction, such as through histone modifications, which dynamically regulate transcription rates of critical genes important in synaptic plasticity underlying learning and memory. Here, we provide a focused review on differential histone modifications within the VTA observed in both humans and animal models, as well as their relevance to disease-based phenotypes, specifically focusing on epigenetic dysregulation of histones in the VTA associated with early life stress (ELS) and drugs of abuse. Locus- and cell-type-specific targeting of individual histone modifications at specific genes within the VTA presents novel therapeutic targets which can result in greater efficacy and better long-term health outcomes in susceptible individuals that are at increased risk for substance use and psychiatric disorders.
Collapse
Affiliation(s)
- Ryan D Shepard
- Department of Pharmacology, Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Fereshteh S Nugent
- Department of Pharmacology, Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
22
|
Hood LE, Leyrer-Jackson JM, Olive MF. Pharmacotherapeutic management of co-morbid alcohol and opioid use. Expert Opin Pharmacother 2020; 21:823-839. [PMID: 32103695 PMCID: PMC7239727 DOI: 10.1080/14656566.2020.1732349] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 02/17/2020] [Indexed: 12/12/2022]
Abstract
Opioid use disorder (OUD) and alcohol use disorder (AUD) are two highly prevalent substance-related disorders worldwide. Co-use of the substances is also quite prevalent, yet there are no pharmacological treatment approaches specifically designed to treat co-morbid OUD and AUD. Here, the authors critically summarize OUD, AUD and opioid/alcohol co-use and their current pharmacotherapies for treatment. They also review the mechanisms of action of opioids and alcohol within the brain reward circuitry and discuss potential combined mechanisms of action and resulting neuroadaptations. Pharmacotherapies that aim to treat AUD or OUD that may be beneficial in the treatment of co-use are also highlighted. Preclinical models assessing alcohol and opioid co-use remain sparse. Lasting neuroadaptations in brain reward circuits caused by co-use of alcohol and opioids remains largely understudied. In order to fully understand the neurobiological underpinnings of alcohol and opioid co-use and develop efficacious pharmacotherapies, the preclinical field must expand its current experimental paradigms of 'single drug' use to encompass polysubstance use. Such studies will provide insights on the neural alterations induced by opioid and alcohol co-use, and may help develop novel pharmacotherapies for individuals with co-occurring alcohol and opioid use disorders.
Collapse
Affiliation(s)
- Lauren E. Hood
- Department of Psychology, Arizona State University, Tempe, Arizona, USA
| | | | - M. Foster Olive
- Department of Psychology, Arizona State University, Tempe, Arizona, USA
| |
Collapse
|
23
|
Shepard RD, Langlois LD, Authement ME, Nugent FS. Histone deacetylase inhibition reduces ventral tegmental area dopamine neuronal hyperexcitability involving AKAP150 signaling following maternal deprivation in juvenile male rats. J Neurosci Res 2020; 98:1457-1467. [PMID: 32162391 DOI: 10.1002/jnr.24613] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/19/2020] [Accepted: 02/27/2020] [Indexed: 12/27/2022]
Abstract
Traumatic early life stress (ELS) is linked to dopamine (DA) dysregulation which increases the probability of developing psychiatric disorders in adolescence and adulthood. Our prior studies demonstrated that a severe early life stressor, a 24-hr maternal deprivation (MD) in juvenile male rats, could lead to altered DA signaling from the ventral tegmental area (VTA) due to impairment of GABAergic synaptic plasticity (promoting GABAergic long-term depression, LTD) with concomitant changes in the abundance of synaptic regulators including A-kinase anchoring protein (AKAP150). Importantly, these MD-induced synaptic changes in the VTA were accompanied by upregulation of histone deacetylase 2, histone hypoacetylation, and were reversible by HDAC inhibition. Using cell-attached and whole-cell patch clamp recordings, we found that MD stress also increased spontaneous VTA DA neuronal activity and excitability in juvenile male rats without affecting intrinsic excitability. Postsynaptic chemical disruption of AKAP150 and protein kinase A interaction increased VTA DA neuronal excitability in control non-MD rats mimicking the effects of MD on DA cell excitability with similar changes in membrane properties. Interestingly, this disruption decreased MD-induced VTA DA hyperexcitability. This MD-induced DA neuronal hyperexcitability could also be normalized at 24 hr after injection of the class 1 HDAC inhibitor, CI-994. Altogether, our data suggest that AKAP150 plays a critical role in the regulation of VTA DA neuronal excitability and that HDAC-mediated targeting of AKAP150 signaling could normalize VTA DA dysfunction following ELS thereby providing novel therapeutic targets for prevention of later life psychopathology.
Collapse
Affiliation(s)
- Ryan D Shepard
- Edward Hebert School of Medicine, Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Ludovic D Langlois
- Edward Hebert School of Medicine, Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Michael E Authement
- Edward Hebert School of Medicine, Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Fereshteh S Nugent
- Edward Hebert School of Medicine, Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
24
|
Sadat-Shirazi MS, Asgari P, Mahboubi S, Nouri Zadeh-Tehrani S, Ashabi G, Rohbani K, Sabzevari S, Soltani H, Khalifeh S, Zarrindast MR. Effect of morphine exposure on novel object memory of the offspring: The role of histone H3 and ΔFosB. Brain Res Bull 2020; 156:141-149. [PMID: 31958477 DOI: 10.1016/j.brainresbull.2020.01.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 12/19/2019] [Accepted: 01/14/2020] [Indexed: 01/09/2023]
Abstract
It has been demonstrated that alteration in histone acetylation in the regions of the brain involved in the reward which may have an important role in morphine addiction. It is well established that epigenetic changes prior to birth influence the function and development of the brain. The current study was designed to evaluate changes in novel object memory, histone acetylation and ΔFosB in the brain of the offspring of morphine-withdrawn parents. Male and female Wistar rats received morphine orally for 21 following days. After ten days of abstinent, they were prepared for mating. The male offspring of the first parturition were euthanized on postnatal days 5, 21, 30 and 60. The novel object recognition (NOR) test was performed on adult male offspring. The amount of acetylated histone H3 and ΔFosB were evaluated in the prefrontal cortex (PFC) and hippocampus using western blotting. Obtained results indicated that the discrimination index in the NOR test was decreased in the offspring of morphine-withdrawn parents as compared with morphine-naïve offspring. In addition, the level of acetylated histone H3 was decreased in the PFC and hippocampus in the offspring of morphine-withdrawn parents during lifetime (postnatal days 5, 21, 30 and 60). In the case of ΔFosB, it also decreased in these regions in the morphine-withdrawn offspring. These results demonstrated that parental morphine exposure affects NOR memory, and decreased the level of histone H3 acetylation and ΔFosB in the PFC and hippocampus. Taken together, the effect of morphine might be transmitted to the next generation even after stop consuming morphine.
Collapse
Affiliation(s)
| | - Pardis Asgari
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran
| | - Sarah Mahboubi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Ghorbangol Ashabi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kiyana Rohbani
- Department of Molecular and Cellular Sciences, Faculty of Advanced Sciences and Technology, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Saba Sabzevari
- Department of Molecular and Cellular Sciences, Faculty of Advanced Sciences and Technology, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Haniyeh Soltani
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran
| | - Solmaz Khalifeh
- Cognitive and Neuroscience Research Center (CNRC), Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad-Reza Zarrindast
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran; Cognitive and Neuroscience Research Center (CNRC), Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Endocrinology and Metabolism Research Institute, Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
25
|
Guo D, Hong D, Wang P, Wang J, Chen L, Zhao W, Zhang L, Yao C, Chu B, Chen S, Li Z, Chen H. Histone deacetylase inhibitor CI-994 inhibits osteoclastogenesis via suppressing NF-κB and the downstream c-Fos/NFATc1 signaling pathways. Eur J Pharmacol 2019; 848:96-104. [PMID: 30682334 DOI: 10.1016/j.ejphar.2019.01.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 01/16/2019] [Accepted: 01/22/2019] [Indexed: 01/09/2023]
Abstract
[4-(acetylamino)-N-(2-amino-phenyl) benzamide] (CI-994) is a histone deacetylase 1-3 specific inhibitor that has been shown to indirectly increase the production of Dickkopf-1, which is an inhibitor of osteoclastic development. However, whether CI-994 has an influence on receptor activator of nuclear factor-kappa B ligand (RANKL)-induced osteoclastogenesis is still unclear; in our study, this mechanism was investigated. In an in vitro study, using a tartrate-resistant acid phosphatase (TRAP) stain, an F-actin ring, bone absorption test, quantitative PCR and Western blotting, the role of CI-994 in osteoclastogenesis and the expression of related genes and proteins were investigated. In an in vivo study, the effect of CI-994 on osteolysis was evaluated using a titanium particle-induced murine calvarial osteolysis model. Our results indicated that CI-994 inhibited osteoclast differentiation and the function of bone resorption without cytotoxic effects. Moreover, CI-994 inhibited the expression of osteoclast-related genes, including ACP5, CTSK, NFATc1, c-Fos, DC-STAMP and V-ATPase-d2. Furthermore, CI-994 suppressed the phosphorylation of IκBα and p65 and the expression of downstream c-Fos and NFATc1. Consistent with the in vitro results described above, our in vivo experiment indicated that CI-994 inhibited Ti-induced osteolysis. In conclusion, CI-994 inhibited osteoclastogenesis by suppressing NF-κB and the downstream c-Fos/NFATc1 signaling pathway. Thus, this study showed the possibility of using CI-994 for the treatment of exorbitant osteoclastic bone resorption.
Collapse
Affiliation(s)
- Di Guo
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Bone development and metabolism research center of Taizhou Hospital, Linhai 317000, China
| | - Dun Hong
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Bone development and metabolism research center of Taizhou Hospital, Linhai 317000, China
| | - Peng Wang
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Bone development and metabolism research center of Taizhou Hospital, Linhai 317000, China
| | - Jiacheng Wang
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Bone development and metabolism research center of Taizhou Hospital, Linhai 317000, China
| | - Lihua Chen
- Enze Medical Research Center, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Bone development and metabolism research center of Taizhou Hospital, Linhai 317000, China
| | - Weibo Zhao
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Bone development and metabolism research center of Taizhou Hospital, Linhai 317000, China
| | - Liwei Zhang
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Orthopedic Institute, Soochow University of Medicine, Soochow 215008, China
| | - Can Yao
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Bone development and metabolism research center of Taizhou Hospital, Linhai 317000, China
| | - Binxiang Chu
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Bone development and metabolism research center of Taizhou Hospital, Linhai 317000, China
| | - Shenao Chen
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Bone development and metabolism research center of Taizhou Hospital, Linhai 317000, China
| | - Zhiyan Li
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Bone development and metabolism research center of Taizhou Hospital, Linhai 317000, China
| | - Haixiao Chen
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai 317000, China; Bone development and metabolism research center of Taizhou Hospital, Linhai 317000, China.
| |
Collapse
|
26
|
You C, Vandegrift BJ, Zhang H, Lasek AW, Pandey SC, Brodie MS. Histone Deacetylase Inhibitor Suberanilohydroxamic Acid Treatment Reverses Hyposensitivity to γ-Aminobutyric Acid in the Ventral Tegmental Area During Ethanol Withdrawal. Alcohol Clin Exp Res 2018; 42:2160-2171. [PMID: 30103280 PMCID: PMC6214766 DOI: 10.1111/acer.13870] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 08/08/2018] [Indexed: 12/24/2022]
Abstract
Background The ventral tegmental area (VTA) is important for alcohol‐related reward and reinforcement. Mouse VTA neurons are hyposensitive to γ‐aminobutyric acid (GABA) during ethanol (EtOH) withdrawal, and GABA responsiveness is normalized by in vitro treatment with histone deacetylase inhibitors (HDACi). The present study examined the effect of a systemically administered HDACi, suberanilohydroxamic acid (SAHA) on GABA sensitivity, and related molecular changes in VTA neurons during withdrawal after chronic EtOH intake in rats. Methods Sprague Dawley male adult rats were fed with Lieber‐DeCarli diet (9% EtOH or control diet) for 16 days. Experimental groups included control diet‐fed and EtOH diet‐fed (0‐ or 24‐hour withdrawal) rats treated with either SAHA or vehicle injection. Single‐unit recordings were used to measure the response of VTA neurons to GABA. Immunohistochemistry was performed to examine levels of HDAC2, acetylated histone H3 lysine 9 (acH3K9), and GABAA receptor α1 and α5 subunits in the VTA; quantitative polymerase chain reaction was performed to examine the mRNA levels of HDAC2 and GABAA receptor subunits. Results VTA neurons from the withdrawal group exhibited GABA hyposensitivity. In vivo SAHA treatment 2 hours before sacrifice normalized the sensitivity of VTA neurons to GABA. EtOH withdrawal was associated with increased HDAC2 and decreased acH3K9 protein levels; SAHA treatment normalized acH3K9 levels. Interestingly, no significant change was observed in the mRNA levels of HDAC2. The mRNA levels, but not protein levels, of GABAA receptor α1 and α5 subunits were increased during withdrawal. Conclusions Withdrawal from chronic EtOH exposure results in a decrease in GABA‐mediated inhibition, and this GABA hyposensitivity is normalized by in vivo SAHA treatment. Disruption of signaling in the VTA produced by alteration of GABA neurotransmission could be 1 neuroadaptive physiological process leading to craving and relapse. These results suggest that HDACi pharmacotherapy with agents like SAHA might be an effective treatment for alcoholism.
Collapse
Affiliation(s)
- Chang You
- Department of Physiology and Biophysics , University of Illinois at Chicago, Chicago, Illinois.,Center for Alcohol Research in Epigenetics , Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois
| | - Bertha J Vandegrift
- Department of Physiology and Biophysics , University of Illinois at Chicago, Chicago, Illinois.,Center for Alcohol Research in Epigenetics , Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois
| | - Huaibo Zhang
- Center for Alcohol Research in Epigenetics , Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois.,Jesse Brown VA Medical Center , Chicago, Illinois
| | - Amy W Lasek
- Center for Alcohol Research in Epigenetics , Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois
| | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics , Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois.,Jesse Brown VA Medical Center , Chicago, Illinois
| | - Mark S Brodie
- Department of Physiology and Biophysics , University of Illinois at Chicago, Chicago, Illinois.,Center for Alcohol Research in Epigenetics , Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
27
|
Shepard RD, Gouty S, Kassis H, Berenji A, Zhu W, Cox BM, Nugent FS. Targeting histone deacetylation for recovery of maternal deprivation-induced changes in BDNF and AKAP150 expression in the VTA. Exp Neurol 2018; 309:160-168. [PMID: 30102916 DOI: 10.1016/j.expneurol.2018.08.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/17/2018] [Accepted: 08/09/2018] [Indexed: 12/21/2022]
Abstract
Severe early life stressors increase the probability of developing psychiatric disorders later in life through modifications in neuronal circuits controlling brain monoaminergic signaling. Our previous work demonstrated that 24 h maternal deprivation (MD) in male Sprague Dawley rats modifies dopamine (DA) signaling from the ventral tegmental area (VTA) through changes at GABAergic synapses that were reversible by in vitro histone deacetylase (HDAC) inhibition which led to restoration of the scaffold A-kinase anchoring protein (AKAP150) signaling and subsequently recovered GABAergic plasticity (Authement et al., 2015). Using a combination of in situ hybridization, Western blots and immunohistochemistry, we confirmed that MD-induced epigenetic modifications at the level of histone acetylation were associated with an upregulation of HDAC2. MD also increased Akap5 mRNA levels in the VTA. Western blot analysis of AKAP150 protein expression showed an increase in synaptic levels of AKAP150 protein in the VTA with an accompanying decrease in synaptic levels of protein kinase A (PKA). Moreover, the abundance of mature brain-derived neurotrophic factor (BDNF) protein of VTA tissues from MD rats was significantly lower than in control groups. In vivo systemic injection with a selective class I HDAC inhibitor (CI-994) was sufficient to reverse MD-induced histone hypoacetylation in the VTA for 24 h after the injection. Furthermore, HDAC inhibition normalized the levels of mBDNF and AKAP150 proteins at 24 h. Our data suggest that HDAC-mediated targeting of BDNF and AKAP-dependent local signaling within VTA could provide novel therapeutics for prevention of later-life psychopathology.
Collapse
Affiliation(s)
- Ryan D Shepard
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Shawn Gouty
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Haifa Kassis
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Aylar Berenji
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - William Zhu
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Brian M Cox
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Fereshteh S Nugent
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA.
| |
Collapse
|
28
|
You C, Vandegrift B, Brodie MS. Ethanol actions on the ventral tegmental area: novel potential targets on reward pathway neurons. Psychopharmacology (Berl) 2018; 235:1711-1726. [PMID: 29549390 PMCID: PMC5949141 DOI: 10.1007/s00213-018-4875-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 03/06/2018] [Indexed: 12/14/2022]
Abstract
The ventral tegmental area (VTA) evaluates salience of environmental stimuli and provides dopaminergic innervation to many brain areas affected by acute and chronic ethanol exposure. While primarily associated with rewarding and reinforcing stimuli, recent evidence indicates a role for the VTA in aversion as well. Ethanol actions in the VTA may trigger neuroadaptation resulting in reduction of the aversive responses to alcohol and a relative increase in the rewarding responses. In searching for effective pharmacotherapies for the treatment of alcohol abuse and alcoholism, recognition of this imbalance may reveal novel strategies. In addition to conventional receptor/ion channel pharmacotherapies, epigenetic factors that control neuroadaptation to chronic ethanol treatment can be targeted as an avenue for development of therapeutic approaches to restore the balance. Furthermore, when exploring therapies to address reward/aversion imbalance in the action of alcohol in the VTA, sex differences have to be taken into account to ensure effective treatment for both men and women. These principles apply to a VTA-centric approach to therapies, but should hold true when thinking about the overall approach in the development of neuroactive drugs to treat alcohol use disorders. Although the functions of the VTA itself are complex, it is a useful model system to evaluate the reward/aversion imbalance that occurs with ethanol exposure and could be used to provide new leads in the efforts to develop novel drugs to treat alcoholism.
Collapse
Affiliation(s)
- Chang You
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 S. Wolcott Ave, Room E-202, M/C 901, Chicago, IL, 60612, USA
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Bertha Vandegrift
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 S. Wolcott Ave, Room E-202, M/C 901, Chicago, IL, 60612, USA
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Mark S Brodie
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 S. Wolcott Ave, Room E-202, M/C 901, Chicago, IL, 60612, USA.
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
29
|
Ostroumov A, Dani JA. Convergent Neuronal Plasticity and Metaplasticity Mechanisms of Stress, Nicotine, and Alcohol. Annu Rev Pharmacol Toxicol 2017; 58:547-566. [PMID: 28977763 DOI: 10.1146/annurev-pharmtox-010617-052735] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Stress and tobacco smoking are risk factors for alcoholism, but the underlying neural mechanisms are not well understood. Although stress, nicotine, and alcohol have broad, individual effects in the brain, some of their actions converge onto the same mechanisms and circuits. Stress and nicotine augment alcohol-related behaviors, in part via modulation of alcohol-evoked neuronal plasticity and metaplasticity mechanisms. Stress modulates alcohol-evoked plasticity via the release of signaling molecules that influence synaptic transmission. Nicotine also activates some of the same signaling molecules, cells, and circuits, producing a convergence of both stress and nicotine onto common plasticity mechanisms that influence alcohol self-administration. We describe several forms of alcohol-induced plasticity, including classic Hebbian plasticity at glutamatergic synapses, and we highlight less appreciated forms, such as non-Hebbian and GABAergic synaptic plasticity. Risk factors such as stress and nicotine initiate lasting neural changes that modify subsequent alcohol-induced synaptic plasticity and increase the vulnerability to alcohol addiction.
Collapse
Affiliation(s)
- Alexey Ostroumov
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School for Medicine, Philadelphia, Pennsylvania 19104, USA; ,
| | - John A Dani
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School for Medicine, Philadelphia, Pennsylvania 19104, USA; ,
| |
Collapse
|
30
|
Abstract
Opioids are among the most effective pain relievers; however, their abuse has been on the rise worldwide evident from an alarming increase in accidental opioid overdoses. This demands for an urgent increase in scientific endeavors for better understanding of main cellular mechanisms and circuits involved in opiate addiction. Preclinical studies strongly suggest that memories associated with positive and negative opioid experiences are critical in promoting compulsive opiate-seeking and opiate-taking behaviors, and relapse. Particular focus on synaptic plasticity as the cellular correlate of learning and memory has rapidly evolved in drug addiction field over the past two decades. Several critical addiction-related brain areas are identified, one of which is the ventral tegmental area (VTA), an area intensively studied as the initial locus for drug reward. Here, we provide an update to our previous review on "Opiates and Plasticity" highlighting the most recent discoveries of synaptic plasticity associated with opiates in the VTA. Electrophysiological studies of plasticity of addiction to date have been invaluable in addressing learning processes and mechanisms that underlie motivated and addictive behaviors, and now with the availability of powerful technologies of transgenic approaches and optogenetics, circuit-based studies hold high promise in fostering synaptic studies of opiate addiction.
Collapse
Affiliation(s)
- Ludovic D. Langlois
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, United States
| | - Fereshteh S. Nugent
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, United States
| |
Collapse
|