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] [MESH Headings] [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
|
Gazerani P. The neuroplastic brain: current breakthroughs and emerging frontiers. Brain Res 2025; 1858:149643. [PMID: 40280532 DOI: 10.1016/j.brainres.2025.149643] [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: 01/12/2025] [Revised: 03/01/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025]
Abstract
Neuroplasticity, the brain's capacity to reorganize itself by forming new neural connections, is central to modern neuroscience. Once believed to occur only during early development, research now shows that plasticity continues throughout the lifespan, supporting learning, memory, and recovery from injury or disease. Substantial progress has been made in understanding the mechanisms underlying neuroplasticity and their therapeutic applications. This overview article examines synaptic plasticity, structural remodeling, neurogenesis, and functional reorganization, highlighting both adaptive (beneficial) and maladaptive (harmful) processes across different life stages. Recent strategies to harness neuroplasticity, ranging from pharmacological agents and lifestyle interventions to cutting-edge technologies like brain-computer interfaces (BCIs) and targeted neuromodulation are evaluated in light of current empirical evidence. Contradictory findings in the literature are addressed, and methodological limitations that hamper widespread clinical adoption are discussed. The ethical and societal implications of deploying novel neuroplasticity-based interventions, including issues of equitable access, data privacy, and the blurred line between treatment and enhancement, are then explored in a structured manner. By integrating mechanistic insights, empirical data, and ethical considerations, the aim is to provide a comprehensive and balanced perspective for researchers, clinicians, and policymakers working to optimize brain health across diverse populations.
Collapse
Affiliation(s)
- Parisa Gazerani
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, Pilestredet 50, 0167 Oslo, Norway.
| |
Collapse
|
3
|
Simon RC, Fleming WT, Briones BA, Trzeciak M, Senthilkumar P, Ishii KK, Hjort MM, Martin MM, Hashikawa K, Sanders AD, Golden SA, Stuber GD. Opioid-driven disruption of the septum reveals a role for neurotensin-expressing neurons in withdrawal. Neuron 2025:S0896-6273(25)00307-1. [PMID: 40378834 DOI: 10.1016/j.neuron.2025.04.024] [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: 03/14/2024] [Revised: 03/14/2025] [Accepted: 04/25/2025] [Indexed: 05/19/2025]
Abstract
Opioid withdrawal is an intensively aversive experience and often drives relapse. The lateral septum (LS) is a forebrain structure that is important in aversion processing and has been linked to substance use disorders, but which LS cell types contribute to the maladaptive state of withdrawal is unknown. We used single-nucleus RNA sequencing to interrogate cell-type-specific gene expression changes induced by chronic morphine exposure and discovered that morphine globally disrupts LS cell types, but neurotensin-expressing neurons (LS-Nts) are selectively activated by naloxone. Using two-photon calcium imaging and ex vivo electrophysiology, we next demonstrate that LS-Nts neurons receive elevated glutamatergic drive in morphine-dependent mice and remain hyperactivated during withdrawal. Finally, we show that manipulating LS-Nts neurons during opioid withdrawal regulates pain coping and sociability. Together, these results suggest that LS-Nts neurons are a key neural substrate involved in opioid withdrawal and establish the LS as a crucial regulator of adaptive behaviors.
Collapse
Affiliation(s)
- Rhiana C Simon
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Weston T Fleming
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Brandy A Briones
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Marta Trzeciak
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Pranav Senthilkumar
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA
| | - Kentaro K Ishii
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Madelyn M Hjort
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Madison M Martin
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Koichi Hashikawa
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Andrea D Sanders
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA
| | - Sam A Golden
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Neurobiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | - Garret D Stuber
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
4
|
Deng X, Xu W, Liu Y, Jing H, Zhong J, Sun K, Zhou R, Xu L, Wu X, Zhang B, Chen W, Jiang S, Chen G, Zhu Y. Social rank modulates methamphetamine-seeking in dominant and subordinate male rodents via distinct dopaminergic pathways. Nat Neurosci 2025:10.1038/s41593-025-01951-0. [PMID: 40355612 DOI: 10.1038/s41593-025-01951-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 03/14/2025] [Indexed: 05/14/2025]
Abstract
Social status has a profound impact on mental health and propensity towards drug addiction. However, the neural mechanisms underlying the effects of social rank on drug-seeking behavior remain unclear. Here we found that dominant male rodents (based on the tube test) had denser mesocortical dopaminergic projections and were more resistant to methamphetamine (METH)-seeking, whereas subordinates had heightened dopaminergic function in the mesolimbic pathway and were more vulnerable to METH seeking. Optogenetic activation of the mesocortical dopaminergic pathway promoted winning and suppressed METH seeking in subordinates, whereas lesions of the mesocortical pathway increased METH seeking in dominants. Elevation of social rank with forced win training in subordinates led to remodeling of the dopaminergic system and prevented METH-seeking behavior. In females, however, both ranks were susceptible to METH seeking, with mesocorticolimbic pathways comparable to those in subordinate males. These results provide a framework for understanding the neural basis of the impact of social status on drug-seeking.
Collapse
Affiliation(s)
- Xiaofei Deng
- Shenzhen Key Laboratory of Drug Addiction, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, China
- Shenzhen Neher Neural Plasticity Laboratory, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wei Xu
- Shenzhen Key Laboratory of Drug Addiction, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, China
- Shenzhen Neher Neural Plasticity Laboratory, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yutong Liu
- Shenzhen Key Laboratory of Drug Addiction, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, China
- Shenzhen Neher Neural Plasticity Laboratory, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Haiyang Jing
- Shenzhen Key Laboratory of Drug Addiction, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jiafeng Zhong
- Shenzhen Key Laboratory of Drug Addiction, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Neher Neural Plasticity Laboratory, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Kaige Sun
- Shenzhen Key Laboratory of Drug Addiction, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Neher Neural Plasticity Laboratory, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ruiyi Zhou
- Shenzhen Key Laboratory of Drug Addiction, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Liang Xu
- Shenzhen Key Laboratory of Drug Addiction, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaocong Wu
- Shenzhen Key Laboratory of Drug Addiction, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Baofang Zhang
- Shenzhen Key Laboratory of Drug Addiction, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wanqi Chen
- Shenzhen Key Laboratory of Drug Addiction, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Neher Neural Plasticity Laboratory, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Shaolei Jiang
- Shenzhen Key Laboratory of Drug Addiction, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Neher Neural Plasticity Laboratory, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Gaowei Chen
- Shenzhen Key Laboratory of Drug Addiction, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, China
- Shenzhen Neher Neural Plasticity Laboratory, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yingjie Zhu
- Shenzhen Key Laboratory of Drug Addiction, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, China.
- Shenzhen Neher Neural Plasticity Laboratory, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
- University of Chinese Academy of Sciences, Beijing, China.
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
- Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, and State Key Laboratory of Biomedical Imaging Science and System, Shenzhen, China.
| |
Collapse
|
5
|
Rombach N, Fedriani R, Scully D, O'Sullivan J, Mulvany SK, Murphy KJ. Initial exposure to addictive drugs activates polysialylated NCAM-mediated hippocampal memory-associated synaptic plasticity in the rat prior to the emergence of dependence behaviour. Prog Neuropsychopharmacol Biol Psychiatry 2025; 139:111395. [PMID: 40334966 DOI: 10.1016/j.pnpbp.2025.111395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 04/18/2025] [Accepted: 05/02/2025] [Indexed: 05/09/2025]
Abstract
The early neural adaptations to addictive substances are crucial to understanding how drug-associated memories form and contribute to later compulsive drug-seeking behaviour. This study investigated whether initial exposures to drugs of abuse engage hippocampal memory mechanisms, specifically through the activation of polysialylated neural cell adhesion molecule (NCAM PSA), a well-established marker of neuroplasticity. Using a rodent model, we assessed hippocampal NCAM PSA expression and synaptic remodelling in response to acute and repeated administration of heroin, cocaine, and amphetamine. We compared these neuroplastic responses with those observed during normal spatial learning. Additionally, we examined changes in hippocampal NCAM PSA levels during heroin self-administration to evaluate the relationship between memory-related hippocampal activation and the emergence of drug-seeking behaviour. Our findings reveal that addictive drugs acutely induce hippocampal neuroplasticity in a manner that parallels normal memory consolidation, including increased NCAM PSA expression and synaptic reorganisation. Following sufficient daily drug exposure, the memory-associated neuroplastic response is lost. Specifically in a setting of drug self-administration, we show that heroin continues to activate molecular memory mechanisms within the hippocampal dentate until the emergence of drug seeking behaviour based on recall of an association between drug reward and lever press. This pattern of hippocampal activity is shown to mirror precisely the molecular events associated with learning a normal explicit memory task, the water maze. Importantly, following chronic exposure to either heroin or cocaine, non-drug-related hippocampal-dependent learning events are no longer processed effectively for long-term storage in the absence of the addictive drug suggesting dependence on the drug for normal memory function. These results identify hippocampal memory machinery as a key early target of addictive drugs and suggest that drug-associated memories may co-opt normal learning processes, contributing to the persistent and intrusive nature of drug cravings. The study provides novel insights into the mechanisms by which early drug experiences shape enduring behavioural vulnerability to addiction.
Collapse
Affiliation(s)
- Nanette Rombach
- Neurotherapeutics Research Group, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin 4 D04 V1W8, Ireland
| | - Rocio Fedriani
- Neurotherapeutics Research Group, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin 4 D04 V1W8, Ireland.
| | - Darren Scully
- Neurotherapeutics Research Group, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin 4 D04 V1W8, Ireland
| | - Josiah O'Sullivan
- Neurotherapeutics Research Group, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin 4 D04 V1W8, Ireland
| | - Sean K Mulvany
- Neurotherapeutics Research Group, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin 4 D04 V1W8, Ireland
| | - Keith J Murphy
- Neurotherapeutics Research Group, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin 4 D04 V1W8, Ireland.
| |
Collapse
|
6
|
Alberquilla S, Nanclares C, Expósito S, Gall G, Kofuji P, Araque A, Martín ED, Moratalla R. Astrocytes Mediate Psychostimulant-Induced Alterations of Spike-Timing Dependent Synaptic Plasticity. Glia 2025; 73:1051-1067. [PMID: 39801264 PMCID: PMC11920680 DOI: 10.1002/glia.24672] [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: 09/14/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 03/20/2025]
Abstract
At cellular and circuit levels, drug addiction is considered a dysregulation of synaptic plasticity. In addition, dysfunction of the glutamate transporter 1 (GLT-1) in the nucleus accumbens (NAc) has also been proposed as a mechanism underlying drug addiction. However, the cellular and synaptic impact of GLT-1 alterations in the NAc remain unclear. Here we show in the NAc that 10 days withdraw after 5 days treatment with cocaine or amphetamine decreases GLT-1 expression in astrocytes, which results in the prolongation of the excitatory postsynaptic potential (EPSP) decay kinetics in D1 receptor-containing medium spiny neurons (D1R-MSNs). Using the spike timing dependent plasticity (STDP) paradigm, we found that enlargement of EPSP duration results in switching the LTP elicited in control animals to LTD in psychostimulant-treated mice. In contrast to D1-MSNs, D2-MSNs did not display changes in EPSP kinetics and synaptic plasticity. Notably, the psychostimulant-induced synaptic transmission and synaptic plasticity effects were absent in IP3R2-/- mice, which lack astrocyte calcium signal, but were mimicked by the selective astrocytes stimulation with DREADDs. Finally, ceftriaxone, which upregulates GLT-1, restored normal GLT-1 function, EPSP kinetics, and synaptic plasticity in psychostimulant-treated mice. Therefore, we propose that cocaine and amphetamine increase dopaminergic levels in the NAc, which stimulates astrocytes and downregulates the GLT-1. The decreased GLT-1 function prolonged the EPSP kinetics, leading to the modulation of the STDP, transforming the LTP observed in control animals into LTD in psychostimulant-treated mice. Present work reveals a novel mechanism underlying the synaptic plasticity changes induced by these drugs of abuse.
Collapse
Affiliation(s)
- Samuel Alberquilla
- Cajal Institute, CSICMadridSpain
- CIBERNED, Instituto de Salud Carlos IIIMadridSpain
| | - Carmen Nanclares
- Department of NeuroscienceUniversity of MinnesotaMinneapolisMinnesotaUSA
| | | | - Grace Gall
- Department of NeuroscienceUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Paulo Kofuji
- Department of NeuroscienceUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Alfonso Araque
- Department of NeuroscienceUniversity of MinnesotaMinneapolisMinnesotaUSA
| | | | - Rosario Moratalla
- Cajal Institute, CSICMadridSpain
- CIBERNED, Instituto de Salud Carlos IIIMadridSpain
| |
Collapse
|
7
|
Alizo Vera V, Childs JE, Kim J, Matheos DP, Wood MA. Expression of HDAC3-Y298H Point Mutant in Medial Habenula Cholinergic Neurons Has No Effect on Cocaine-Induced Behaviors. eNeuro 2025; 12:ENEURO.0590-24.2025. [PMID: 40368589 PMCID: PMC12077810 DOI: 10.1523/eneuro.0590-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 04/14/2025] [Accepted: 04/21/2025] [Indexed: 05/16/2025] Open
Abstract
Histone deacetylase 3 (HDAC3) is one of the most highly expressed HDACs in the brain shown to be a negative regulator of long-term memory formation. HDAC3 has also been shown to be involved in cocaine-associated behaviors, demonstrated by manipulations in the nucleus accumbens. Previous studies have demonstrated that expression of a dominant negative of a key HDAC3 target gene, nuclear receptor subfamily 4 group A member 2 (NR4A2), in cholinergic neurons of the medial habenula (MHb) blocked reinstatement of cocaine-induced conditioned place preference (CPP) as well as cue-induced intravenous self-administration (IVSA). Together, these findings suggested that HDAC3 would also be important for MHb-dependent reinstatement of CPP and IVSA, which we examined in this study. Contrary to our hypothesis, our results found that expression of an HDAC3 deacetylase dead point mutant within the cholinergic neurons of the mouse MHb had no effect on reinstatement or other cocaine-induced behaviors.
Collapse
Affiliation(s)
- Vanessa Alizo Vera
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, California 92697
- Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, Irvine, California 92697
| | - Jessica E Childs
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, California 92697
- Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, Irvine, California 92697
| | - Jisung Kim
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, California 92697
- Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, Irvine, California 92697
| | - Dina P Matheos
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, California 92697
- Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, Irvine, California 92697
| | - Marcelo A Wood
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, California 92697
- Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, Irvine, California 92697
| |
Collapse
|
8
|
Li B, Zhou Y, Qian Y, Wu J. Impact of exercise on drug cravings: mediating role of cardiorespiratory fitness and inhibitory control. Front Psychol 2025; 16:1540648. [PMID: 40271371 PMCID: PMC12014659 DOI: 10.3389/fpsyg.2025.1540648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 03/27/2025] [Indexed: 04/25/2025] Open
Abstract
Background Contemporary research has consistently demonstrated a link between physical exercise, inhibition, and drug cravings, with several hypotheses proposed to explain how exercise enhances inhibition. However, few studies have explored the mechanisms underlying this effect. This study investigates the pivotal role of cardiorespiratory fitness in mediating the impact of physical exercise on inhibition and drug cravings. Methods In this study, the researchers selected participants who had completed physical detoxification in China's compulsory isolation drug rehabilitation centers. Moreover, we conducted surveys and assessed cardiorespiratory fitness using tools such as the Physical Exercise Rating Scale, Inhibition Scale, Drug Craving Scale, and Queen College Step Test. Additionally, we employed exploratory and confirmatory factor analyses, correlation analysis, regression analysis, and structural equation model (SEM) to analyze the study's data. Results and findings The findings of this study reveal that physical exercise significantly reduces drug craving by improving cardiorespiratory fitness and enhancing inhibition. Furthermore, it showed that men had better physical exercise levels, cardiorespiratory fitness, and inhibition compared to women, while women exhibited higher psychological cravings. Besides, the study also highlighted that longer years of drug use were associated with lower physical exercise, reduced cardiorespiratory fitness, and weaker inhibition, leading to higher drug cravings. In addition, cardiorespiratory fitness and inhibition acted as mediators between physical exercise and drug craving, with the combination of both factors serving as a chain mediator in reducing cravings. These findings suggest that physical exercise, particularly through improving cardiorespiratory fitness, plays a key role in mitigating drug cravings and supporting rehabilitation efforts for drug dependence.
Collapse
Affiliation(s)
- Boya Li
- School of Sport Science, Qufu Normal University, Qufu, Shandong, China
| | - Yuehui Zhou
- School of Sport Science, Qufu Normal University, Qufu, Shandong, China
| | - Youling Qian
- School of Physical Education, Hubei Minzu University, Hubei, Enshi, China
| | - Juhua Wu
- School of Sport, Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| |
Collapse
|
9
|
Browne CJ, Mews P, Estill M, Zhou X, Holt LM, Futamura R, Shen L, Zhang B, Nestler EJ. Cocaine and morphine induce shared and divergent transcriptional regulation in nucleus accumbens D1 and D2 medium spiny neurons. Mol Psychiatry 2025:10.1038/s41380-025-03004-1. [PMID: 40188314 DOI: 10.1038/s41380-025-03004-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 03/07/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
Substance use disorders (SUDs) induce widespread molecular dysregulation in nucleus accumbens (NAc), a brain region pivotal for coordinating motivation and reward, which is linked to neural and behavioral disturbances promoting addiction. Despite the overlapping symptomatology of SUDs, different drug classes exert partly unique influences on neural circuits, cell types, physiology, and gene expression. To better understand common and divergent molecular mechanisms governing SUD pathology, we characterized the cell-type-specific restructuring of the NAc transcriptional landscape after psychostimulant or opioid exposure. We combined fluorescence-activated nuclei sorting and deep RNA sequencing to profile NAc D1 and D2 medium spiny neurons (MSNs) across cocaine and morphine exposure paradigms, including initial exposure, prolonged withdrawal after repeated exposure, and re-exposure post-withdrawal. Our analyses reveal that D1 MSNs display many convergent transcriptional responses between the two drug classes, whereas D2 MSNs manifest highly divergent responses, with morphine causing more adaptations in this cell type. Utilizing multiscale embedded gene co-expression network analysis (MEGENA), we discerned transcriptional regulatory networks subserving biological functions altered by cocaine vs. morphine. We observed largely integrative engagement of overlapping gene networks across drug classes in D1 MSNs, but opposite regulation of key D2 networks, highlighting potential therapeutic gene network targets within MSNs. Analysis of gene regulatory systems at the level of enhancers revealed that morphine engages a unique enhancer landscape in D2 MSNs compared to cocaine. Our findings, and future work leveraging this dataset, will open avenues for the development of targeted therapeutic interventions, addressing the urgent need for more effective treatments for SUDs.
Collapse
Affiliation(s)
- Caleb J Browne
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Philipp Mews
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Dept. of Pharmacology, Physiology & Biophysics, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Molly Estill
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xianxiao Zhou
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Dept. of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Leanne M Holt
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rita Futamura
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Li Shen
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bin Zhang
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Dept. of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Dept. of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
10
|
Brida KL, Jorgensen ET, Phillips RA, Newman CE, Tuscher JJ, Morring EK, Zipperly ME, Ianov L, Montgomery KD, Tippani M, Hyde TM, Maynard KR, Martinowich K, Day JJ. Reelin marks cocaine-activated striatal neurons, promotes neuronal excitability, and regulates cocaine reward. SCIENCE ADVANCES 2025; 11:eads4441. [PMID: 40138397 PMCID: PMC12076537 DOI: 10.1126/sciadv.ads4441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 02/20/2025] [Indexed: 03/29/2025]
Abstract
Drugs of abuse activate defined neuronal populations in reward structures such as the nucleus accumbens (NAc), which promote the enduring synaptic, circuit, and behavioral consequences of drug exposure. While the molecular and cellular effects arising from experience with drugs like cocaine are increasingly well understood, mechanisms that dictate NAc neuronal recruitment remain unknown. Here, we leveraged unbiased single-nucleus transcriptional profiling and targeted in situ detection to identify Reln (encoding the secreted glycoprotein, Reelin) as a marker of cocaine-activated neuronal populations within the rat NAc. A CRISPR interference approach enabling selective Reln knockdown in the adult NAc altered expression of calcium signaling genes, promoted a transcriptional trajectory consistent with loss of cocaine sensitivity, and decreased MSN excitability. Behaviorally, Reln knockdown prevented cocaine locomotor sensitization, abolished cocaine place preference memory, and decreased cocaine self-administration behavior. These results identify Reelin as a critical mechanistic link between neuronal activation and cocaine-induced behavioral adaptations.
Collapse
Affiliation(s)
- Kasey L. Brida
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Emily T. Jorgensen
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Robert A. Phillips
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Catherine E. Newman
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jennifer J. Tuscher
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Emily K. Morring
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Morgan E. Zipperly
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lara Ianov
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
- Civitan International Research Center,
University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kelsey D. Montgomery
- Lieber Institute for Brain Development,
Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Madhavi Tippani
- Lieber Institute for Brain Development,
Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Thomas M. Hyde
- Lieber Institute for Brain Development,
Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral
Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205,
USA
- Department of Neurology, Johns Hopkins
University School of Medicine, Baltimore, MD 21205, USA
| | - Kristen R. Maynard
- Lieber Institute for Brain Development,
Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral
Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205,
USA
- Department of Neuroscience, Johns Hopkins
University School of Medicine, Baltimore, MD 21205, USA
| | - Keri Martinowich
- Lieber Institute for Brain Development,
Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral
Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205,
USA
- Department of Neuroscience, Johns Hopkins
University School of Medicine, Baltimore, MD 21205, USA
- The Kavli Neuroscience Discovery
Institute, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jeremy J. Day
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
11
|
Wang B, Wang J, Beacher NJ, Lin DT, Zhang Y. Cell-type specific epigenetic and transcriptional mechanisms in substance use disorder. Front Cell Neurosci 2025; 19:1552032. [PMID: 40226298 PMCID: PMC11985801 DOI: 10.3389/fncel.2025.1552032] [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: 12/27/2024] [Accepted: 03/13/2025] [Indexed: 04/15/2025] Open
Abstract
Substance use disorder (SUD) is a chronic and relapse-prone neuropsychiatric disease characterized by impaired brain circuitry within multiple cell types and neural circuits. Recent advancements in single-cell transcriptomics, epigenetics, and neural circuit research have unveiled molecular and cellular alterations associated with SUD. These studies have provided valuable insights into the transcriptional and epigenetic regulation of neuronal and non-neuronal cells, particularly in the context of drug exposure. Critical factors influencing the susceptibility of individuals to SUD include the regulation of gene expression during early developmental stages, neuroadaptive responses to psychoactive substances, and gene-environment interactions. Here we briefly review some of these mechanisms underlying SUD, with an emphasis on their crucial roles in in neural plasticity and maintenance of addiction and relapse in neuronal and non-neuronal cell-types. We foresee the possibility of integrating multi-omics technologies to devise targeted and personalized therapeutic strategies aimed at both the prevention and treatment of SUD. By utilizing these advanced methodologies, we can gain a deeper understanding of the fundamental biology of SUD, paving the way for more effective interventions.
Collapse
Affiliation(s)
- Bin Wang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jiale Wang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Nicholas J. Beacher
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, United States
| | - Da-Ting Lin
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, United States
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yan Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China
| |
Collapse
|
12
|
Davis JL, Kennedy C, McMahon CL, Keegan L, Clerkin S, Treacy NJ, Hoban AE, Kelly Y, Brougham DF, Crean J, Murphy KJ. Cocaine perturbs neurodevelopment and increases neuroinflammation in a prenatal cerebral organoid model. Transl Psychiatry 2025; 15:94. [PMID: 40140359 PMCID: PMC11947122 DOI: 10.1038/s41398-025-03315-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 01/17/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Prenatal exposure to cocaine causes abnormalities in foetal brain development, which are linked to later development of anxiety, depression and cognitive dysfunction. Previous studies in rodent models have indicated that prenatal cocaine exposure affects proliferation, differentiation and connectivity of neural cell types. Here, using cerebral organoids derived from the human iPSC cell line HPSI1213i-babk_2, we investigated cocaine-induced changes of the gene expression regulatory landscape at an early developmental time point, leveraging recent advances in single cell RNA-seq and single cell ATAC-seq. iPSC-cerebral organoids replicated well-established cocaine responses observed in vivo and provided additional information about the cell-type specific regulation of gene expression following cocaine exposure. Cocaine altered gene expression patterns, in part through epigenetic landscape remodelling, and revealed disordered neural plasticity mechanisms in the cerebral organoids. Perturbed neurodevelopmental cellular signalling and an inflammatory-like activation of astrocyte populations were also evident following cocaine exposure. The combination of altered neuroplasticity, neurodevelopment and neuroinflammatory signalling suggests cocaine exposure can mediate substantial disruption of normal development and maturation of the brain. These findings offer new insights into the cellular mechanism underlying the adverse effects of cocaine exposure on neurodevelopment and point to the possible pathomechanisms of later neuropsychiatric disturbances.
Collapse
Affiliation(s)
- Jessica L Davis
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Ciaran Kennedy
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Ciara L McMahon
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Louise Keegan
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Shane Clerkin
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Niall J Treacy
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Alan E Hoban
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Yazeed Kelly
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Dermot F Brougham
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - John Crean
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Keith J Murphy
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
13
|
Pratelli M, Spitzer NC. Drugs of abuse drive neurotransmitter plasticity that alters behavior: implications for mental health. Front Behav Neurosci 2025; 19:1551213. [PMID: 40177329 PMCID: PMC11962007 DOI: 10.3389/fnbeh.2025.1551213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
Neurotransmission is a complex process with multiple levels of regulation that, when altered, can significantly impact mental health. Neurons in the adult brain can release more than one transmitter and environmental stimuli can change the type of transmitter neurons express. Changes in the transmitter neurons express can generate changes in animal behavior. The ability of neurons to express multiple transmitters and/or switch them in response to environmental stimuli likely evolved to provide flexibility and complexity to neuronal circuit function in an ever-changing environment. However, this adaptability can become maladaptive when generating behavioral alterations that are unfit for the environment in which the animal lives or the tasks it needs to perform. Repeated exposure to addictive substances induces long-lasting molecular and synaptic changes, driving the appearance of maladaptive behaviors that can result in drug misuse and addiction. Recent findings have shown that one way drugs of abuse alter the brain is by inducing changes in the transmitter neurons express. Here, we review evidence of prolonged exposure to addictive substances inducing changes in the number of neurons expressing the neuropeptide orexin, the neuromodulator dopamine, and the inhibitory transmitter GABA. These findings show that drug-induced transmitter plasticity is conserved across species, that addictive substances belonging to different classes of chemicals can induce the same type of plasticity, and that exposure to only one drug can cause different neuronal types to change the transmitter they express. Importantly, drug-induced transmitter plasticity contributes to the long-term negative effects of drug consumption, and it can, in some cases, be either prevented or reversed to alleviate these outcomes. Regional neuronal hyperactivity appears to modulate the appearance and stabilization of drug-induced changes in transmitter expression, which are no longer observed when activity is normalized. Overall, these findings underscore the importance of continuing to investigate the extent and behavioral significance of drug-induced neurotransmitter plasticity and exploring whether non-invasive strategies can be used to reverse it as a means to mitigate the maladaptive effects of drug use.
Collapse
Affiliation(s)
- Marta Pratelli
- Department of Neurobiology, School of Biological Sciences, Center for Neural Circuits and Behavior, University of California, San Diego, La Jolla, CA, United States
- Kavli Institute for Brain and Mind, University of California, San Diego, La Jolla, CA, United States
| | - Nicholas C. Spitzer
- Department of Neurobiology, School of Biological Sciences, Center for Neural Circuits and Behavior, University of California, San Diego, La Jolla, CA, United States
- Kavli Institute for Brain and Mind, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
14
|
Bornhoft KN, Prohofsky J, O’Neal TJ, Wolff AR, Saunders BT. Striatal dopamine represents valence on dynamic regional scales. J Neurosci 2025; 45:e1551242025. [PMID: 40097183 PMCID: PMC12019117 DOI: 10.1523/jneurosci.1551-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/31/2025] [Accepted: 03/11/2025] [Indexed: 03/19/2025] Open
Abstract
Adaptive decision making relies on dynamic updating of learned associations where environmental cues come to predict valenced stimuli, such as food or threat. Cue-guided behavior depends on a network of brain systems, including dopaminergic projections to the striatum. Critically, it remains unclear how dopamine signaling across the striatum encodes multi-valent, dynamic learning contexts, where positive and negative associations must be rapidly disambiguated. To understand this, we employed a Pavlovian discrimination paradigm, where cues predicting food or threat were intermingled during conditioning sessions, and their meaning was serially reversed across training. We found that male and female rats readily distinguished these cues and updated their behavior rapidly upon valence reversal. Using fiber photometry, we recorded dopamine signaling in three major striatal subregions - the dorsolateral striatum (DLS), the nucleus accumbens (NAc) core, and the nucleus accumbens medial shell - finding that valence was represented uniquely across all three regions, indicative of local signals biased for value and salience. Further, ambiguity introduced by cue reversals reshaped striatal dopamine on different timelines: nucleus accumbens signals updated more readily than those in the DLS. Together, these results indicate that striatal dopamine flexibly encodes stimulus valence according to region-specific rules, and these signals are dynamically modulated by changing contingencies in the resolution of ambiguity about the meaning of environmental cues.Significance Statement Adaptive decision making relies on updating learned associations to disambiguate predictions of reward or threat. This cue-guided behavior depends on striatal dopamine, but it remains unclear how dopamine signaling encodes multi-valent, dynamic learning contexts. Here, we employed a paradigm where cues predicting positive and negative outcomes were intermingled, and their meaning was serially reversed across time. We recorded dopamine signaling, finding heterogeneous patterns of valence encoding across striatal subregions, and cue reversal reshaped subregional signals on different timelines. Our results suggest that dopamine flexibly encodes dynamic learning contexts to resolve ambiguity about the meaning of environmental cues.
Collapse
Affiliation(s)
- Kaisa N. Bornhoft
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
- Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota 55455
| | - Julianna Prohofsky
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
- Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota 55455
| | - Timothy J. O’Neal
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
- Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota 55455
| | - Amy R. Wolff
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
- Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota 55455
| | - Benjamin T. Saunders
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
- Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota 55455
| |
Collapse
|
15
|
Nestler EJ. The biology of addiction. Sci Signal 2025; 18:eadq0031. [PMID: 39903810 DOI: 10.1126/scisignal.adq0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 12/10/2024] [Indexed: 02/06/2025]
Abstract
The tools of modern genetics and neurobiology have propelled a renaissance of research that has advanced our understanding of the pathophysiology of drug addiction. We know that an individual's risk for addiction is determined by interactions between genetics and environment and that only a minute fraction of chemical agents share the ability to act on this vulnerability to induce a state of addiction. Repeated exposure to these drugs causes addiction through repeated activation of dopaminergic transmission (and many other actions) in the brain, inducing changes at the molecular, cellular, and synaptic levels that, over time, rewire the circuitry throughout the limbic system. In this Review, I discuss how we are gaining a clearer picture of this drug-induced plasticity-some of which is shared by all addictive drugs, whereas other aspects are specific to certain drug classes-and of the ways in which these adaptations mediate the range of behavioral abnormalities that define the addicted state. Despite the challenges, there is reason for optimism in translating this rich biological understanding of addiction into improved treatments for the many individuals burdened by this illness around the world.
Collapse
Affiliation(s)
- Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
16
|
Vernovsky S, Herning A, Wachman EM. The role of genetics in neonatal abstinence syndrome. Semin Perinatol 2025; 49:152006. [PMID: 39613584 DOI: 10.1016/j.semperi.2024.152006] [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: 11/04/2024] [Accepted: 11/16/2024] [Indexed: 12/01/2024]
Abstract
Neonatal Abstinence Syndrome (NAS) after in-utero exposure to opioids remains a significant public health concern. NAS is a highly variable condition in which presentation and severity cannot be explained by clinical factors alone. Research in human subjects has identified both genetic and epigenetic associations with prenatal opioid exposure and NAS severity, including single nucleotide polymorphisms, DNA methylation differences, and gene expression modifications. Animal studies have also identified key gene pathways that are likely important contributors to NAS phenotype. The clinical significance of identified genetic associations with NAS are unclear and warrant further study to see how they could impact NAS management.
Collapse
Affiliation(s)
- Sarah Vernovsky
- Department of Pediatrics, Boston Medical Center, Boston, MA, United States
| | - Ana Herning
- Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | - Elisha M Wachman
- Department of Pediatrics, Boston Medical Center, Boston, MA, United States; Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States.
| |
Collapse
|
17
|
Engeln M, Ahmed SH. Remission from addiction: erasing the wrong circuits or making new ones? Nat Rev Neurosci 2025; 26:115-130. [PMID: 39663409 DOI: 10.1038/s41583-024-00886-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2024] [Indexed: 12/13/2024]
Abstract
Chronic relapse is a hallmark of substance-use disorders (SUDs), but many people with SUDs do recover and eventually enter remission. Many preclinical studies in this field aim to identify interventions that can precipitate recovery by reversing or erasing the neuronal circuit changes caused by chronic drug use. A better understanding of remission from SUDs can also come from preclinical studies that model factors known to influence recovery in humans, such as the negative consequences of drug use and positive environmental influences. In this Perspective we discuss human neuroimaging studies that have provided information about recovery from SUDs and highlight mechanisms identified in preclinical studies - such as the reconfiguration of neuronal circuits - that could contribute to remission. We also analyse how studies of memory and forgetting can provide insights into the mechanisms of remission. Overall, we propose that remission can be driven by the introduction of new neuronal changes (which outcompete those induced by drugs) as well as by the erasure of drug-induced changes.
Collapse
Affiliation(s)
- Michel Engeln
- Univ. Bordeaux, CNRS, INCIA, UMR 5287, Bordeaux, France.
| | - Serge H Ahmed
- Univ. Bordeaux, CNRS, INCIA, UMR 5287, Bordeaux, France
| |
Collapse
|
18
|
Saint-Jour E, Allichon MC, Andrianarivelo A, Montalban E, Martin C, Huet L, Heck N, Hagenston AM, Ravenhorst A, Marias M, Gervasi N, Arrivet F, Vilette A, Pinchaud K, Betuing S, Lissek T, Caboche J, Bading H, Vanhoutte P. Nuclear Calcium Signaling in D 1 Receptor-Expressing Neurons of the Nucleus Accumbens Regulates Molecular, Cellular, and Behavioral Adaptations to Cocaine. Biol Psychiatry 2025:S0006-3223(25)00055-1. [PMID: 39864789 DOI: 10.1016/j.biopsych.2025.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 01/28/2025]
Abstract
BACKGROUND The persistence of cocaine-evoked adaptations relies on gene regulations within the reward circuit, especially in the ventral striatum (i.e., nucleus accumbens [NAc]). Notably, activation of the ERK (extracellular signal-regulated kinase) pathway in the striatum is known to trigger a transcriptional program shaping long-term responses to cocaine. Nuclear calcium signaling has also been shown to control multiple forms of transcription-dependent neuroadaptations, but the dynamics and roles of striatal nuclear calcium signaling in preclinical models of addiction remain unknown. METHODS A genetically encoded cell type-specific nuclear calcium probe has been developed to monitor calcium dynamics in the nuclei of striatal neurons, including in freely moving mice. A cell type-specific inhibitor of nuclear calcium signaling combined with 3-dimensional imaging of neuronal morphology, immunostaining, and behavior was used to disentangle the roles of nuclear calcium in NAc medium spiny neurons (MSNs) expressing the dopamine D1 receptor (D1R) or D2 receptor (D2R) on cocaine-evoked responses. RESULTS The D1R-mediated potentiation of calcium influx through glutamate NMDA receptors, which shapes cocaine effects, also drives nuclear calcium transients. Fiber photometry revealed that cocaine-treated mice showed a sustained nuclear calcium increase in NAc D1R-MSNs. Disrupting nuclear calcium in D1R-MSNs, but not D2R-MSNs, blocked cocaine-evoked morphological changes of MSNs and gene expression and blunted cocaine's rewarding effects. CONCLUSIONS Our study unravels the dynamics and roles of cocaine-induced nuclear calcium signaling increases in D1R-MSNs on molecular, cellular, and behavioral adaptations to cocaine and represents a significant breakthrough because it could contribute to the development of innovative strategies with therapeutic potential to alleviate addiction symptoms.
Collapse
Affiliation(s)
- Estefani Saint-Jour
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France
| | - Marie-Charlotte Allichon
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France
| | - Andry Andrianarivelo
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France
| | - Enrica Montalban
- Université Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, Unité Mixte de Recherche 8251, Centre National de la Recherche Scientifique, Paris, France
| | - Claire Martin
- Université Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, Unité Mixte de Recherche 8251, Centre National de la Recherche Scientifique, Paris, France
| | - Lisa Huet
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France
| | - Nicolas Heck
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France
| | - Anna M Hagenston
- Heidelberg University, Interdisciplinary Center for Neurosciences, Institute of Neurobiology, Heidelberg, Germany
| | - Aisha Ravenhorst
- Heidelberg University, Interdisciplinary Center for Neurosciences, Institute of Neurobiology, Heidelberg, Germany
| | - Mélanie Marias
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France
| | - Nicolas Gervasi
- Center for Interdisciplinary Research in Biology, College de France, Centre National de la Recherche Scientifique UMR 7241, Institut National de la Santé et de la Recherche Médicale U1050, Paris Science et Lettre Research University, Paris, France
| | - Faustine Arrivet
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France
| | - Adèle Vilette
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France
| | - Katleen Pinchaud
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France
| | - Sandrine Betuing
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France
| | - Thomas Lissek
- Heidelberg University, Interdisciplinary Center for Neurosciences, Institute of Neurobiology, Heidelberg, Germany
| | - Jocelyne Caboche
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France
| | - Hilmar Bading
- Heidelberg University, Interdisciplinary Center for Neurosciences, Institute of Neurobiology, Heidelberg, Germany
| | - Peter Vanhoutte
- Sorbonne University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut of Biology Paris-Seine, Center for Neuroscience at Sorbonne University, Paris, France.
| |
Collapse
|
19
|
Ngo AL, Ahmad CM, Gharavi Alkhansari N, Nguyen L, Zhang H. Epigenetic Insights into Substance Use Disorder and Associated Psychiatric Conditions. Complex Psychiatry 2025; 11:12-36. [PMID: 40201238 PMCID: PMC11975344 DOI: 10.1159/000544912] [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: 09/11/2024] [Accepted: 02/19/2025] [Indexed: 04/10/2025] Open
Abstract
Background Substance use disorder (SUD) is closely associated with epigenetic modifications that significantly impact mental health outcomes. Alcohol and drug misuse induce widespread changes in the epigenome and transcriptome of the central nervous system, disrupting critical processes such as reward signaling and emotional regulation. These alterations in epigenetic regulation and gene expression often persist even after substance cessation, potentially contributing to the onset or worsening of psychiatric conditions, including schizophrenia, depression, stress, and anxiety. Summary This review delves into key epigenetic mechanisms underlying SUD and its comorbid psychiatric disorders, with a focus on DNA methylation, histone modifications, and noncoding RNA regulation. Additionally, it examines the influence of environmental and biological factors on the epigenome and evaluates emerging epigenetic-based therapeutic strategies aimed at treating SUD and related psychiatric conditions. Key Messages Gaining a deeper understanding of the epigenetic mechanisms driving SUD and its associated psychiatric disorders is crucial for the development of effective therapeutic interventions. This review highlights the potential of epigenetic-based pharmacological strategies to mitigate the societal and personal burdens linked to SUD and its mental health complications.
Collapse
Affiliation(s)
- Ambrose Loc Ngo
- College of Medicine, Kansas City University, Kansas City, MO, USA
| | | | | | - Linda Nguyen
- College of Pharmacy, Western University, Pomona, CA, USA
| | - Huiping Zhang
- Department of Psychiatry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Section of Biomedical Genetics, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| |
Collapse
|
20
|
Bremner JD, Russo SJ, Gallagher R, Simon NM. Acute and long-term effects of COVID-19 on brain and mental health: A narrative review. Brain Behav Immun 2025; 123:928-945. [PMID: 39500417 PMCID: PMC11974614 DOI: 10.1016/j.bbi.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/16/2024] [Accepted: 11/02/2024] [Indexed: 11/09/2024] Open
Abstract
BACKGROUND COVID infection has been associated with long term sequalae (Long COVID) which include neurological and behavioral effects in thousands of patients, but the etiology and scope of symptoms is not well understood. This paper reviews long term sequelae of COVID on brain and mental health in patients with the Long COVID syndrome. METHODS This was a literature review which queried databases for Pubmed, Psychinfo, and Medline for the following topics for January 1, 2020-July 15, 2023: Long COVID, PASC, brain, brain imaging, neurological, neurobiology, mental health, anxiety, depression. RESULTS Tens of thousands of patients have developed Long COVID, with the most common neurobehavioral symptoms anosmia (loss of smell) and fatigue. Anxiety and mood disorders are elevated and seen in about 25% of Long COVID patients. Neuropsychological testing studies show a correlation between symptom severity and cognitive dysfunction, while brain imaging studies show global decreases in gray matter and alterations in olfactory and other brain areas. CONCLUSIONS Studies to date show an increase in neurobehavioral disturbances in patients with Long COVID. Future research is needed to determine mechanisms.
Collapse
Affiliation(s)
- J Douglas Bremner
- Departments of Psychiatry & Behavioral Sciences and Radiology, Emory University School of Medicine, Atlanta Georgia, and the Atlanta VA Medical Center, Decatur, GA, USA; Nash Family Department Neuroscience and Brain-Body Research Center, Icahn School of Medicine at Mt. Sinai, New York, NY, USA; Department of Child and Adolescent Psychiatry, New York University (NYU) Langone Health, New York, NY, USA.
| | - Scott J Russo
- Nash Family Department Neuroscience and Brain-Body Research Center, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Richard Gallagher
- Department of Child and Adolescent Psychiatry, New York University (NYU) Langone Health, New York, NY, USA; Department of Psychiatry, New York University (NYU) Langone Health, New York, NY, USA
| | - Naomi M Simon
- Department of Psychiatry, New York University (NYU) Langone Health, New York, NY, USA
| |
Collapse
|
21
|
Al-Marzooqi N, Al-Suhail H, AlRefai MO, Alhaj HA. Genomic factors associated with substance use disorder relapse: A critical review. Addict Behav Rep 2024; 20:100569. [PMID: 39553284 PMCID: PMC11568783 DOI: 10.1016/j.abrep.2024.100569] [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: 05/09/2024] [Revised: 10/01/2024] [Accepted: 10/27/2024] [Indexed: 11/19/2024] Open
Abstract
Several genetic and epigenetic factors contribute to the elevated substance use disorder (SUD) relapse vulnerability, yet a comprehensive investigation into these factors is lacking. This review aims to delve into current literature to highlight key genomic factors associated with SUD relapse. Focusing on genetic predisposition and epigenetic modifications the review synthesized research findings of several genetic polymorphisms, histone modifications and DNA methylation patterns contributing to the initiation of SUD and the elevated relapse susceptibility. Notably, specific gene polymorphisms, such as Dopamine Receptor D2 gene (DRD2), Gamma-Aminobutyric Acid Receptor Alpha gene (GABRA2), Catechol-O-methyltransferase (COMT) gene, Dopamine Transporter (DAT1) gene and others were identified to be connected to various patterns of SUD relapse. Furthermore, SUD initiation and relapse has been shown to be influenced by epigenetics. Specifically, CpG hypermethylation has been associated with severe alcohol use disorder in the 5' untranslated region of the Bladder Cancer Associated Protein gene (BLCAP) and the upstream region of the Active BCR Related gene (ABR). Co-users of cannabis and tobacco showed notable variations in CpG site methylation, especially at the Aryl Hydrocarbon Receptor Repressor (AHRR), and factor II receptor-like 3 gene sites (F2RL3). In conclusion, there is good evidence of certain associations between genomic factors and relapse to SUD. However, further research is needed to ascertain causality effects of these factors and develop novel interventions for effective treatment and relapse prevention.
Collapse
Affiliation(s)
- Noora Al-Marzooqi
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Hanan Al-Suhail
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Mohammad O. AlRefai
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Hamid A Alhaj
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
22
|
Howard PG, Zou P, Zhang Y, Huang F, Tesic V, Wu CYC, Lee RHC. Serum/glucocorticoid regulated kinase 1 (SGK1) in neurological disorders: pain or gain. Exp Neurol 2024; 382:114973. [PMID: 39326820 PMCID: PMC11536509 DOI: 10.1016/j.expneurol.2024.114973] [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: 08/26/2024] [Revised: 09/18/2024] [Accepted: 09/21/2024] [Indexed: 09/28/2024]
Abstract
Serum/Glucocorticoid Regulated Kinase 1 (SGK1), a serine/threonine kinase, is ubiquitous across a wide range of tissues, orchestrating numerous signaling pathways and associated with various human diseases. SGK1 has been extensively explored in diverse types of immune and inflammatory diseases, cardiovascular disorders, as well as cancer metastasis. These studies link SGK1 to cellular proliferation, survival, metabolism, membrane transport, and drug resistance. Recently, increasing research has focused on SGK1's role in neurological disorders, including a variety of neurodegenerative diseases (e.g., Alzheimer's disease, Huntington's disease and Parkinson's disease), brain injuries (e.g., cerebral ischemia and traumatic brain injury), psychiatric conditions (e.g., depression and drug addiction). SGK1 is emerging as an increasingly compelling therapeutic target across the spectrum of neurological disorders, supported by the availability of several effective agents. However, the conclusions of many studies observing the prevalence and function of SGK1 in neurological disorders are contradictory, necessitating a review of the SGK1 research within neurological disorders. Herein, we review recent literature on SGK1's primary functions within the nervous system and its impacts within different neurological disorders. We summarize significant findings, identify research gaps, and outline possible future research directions based on the current understanding of SGK1 to help further progress the understanding and treatment of neurological disorders.
Collapse
Affiliation(s)
- Peyton Grace Howard
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA
| | - Peibin Zou
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA
| | - Yulan Zhang
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA
| | - Fang Huang
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA
| | - Vesna Tesic
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA
| | - Celeste Yin-Chieh Wu
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA.
| | - Reggie Hui-Chao Lee
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA; Department of Neurology, Shreveport, Louisiana State University Health, LA, USA; Department of Department of Cell Biology & Anatomy, Louisiana State University Health, Shreveport, LA, USA.
| |
Collapse
|
23
|
Xi ZX, Bocarsly ME, Galaj E, Hempel B, Teresi C, Shaw M, Bi GH, Jordan C, Linz E, Alton H, Tanda G, Freyberg Z, Alvarez VA, Newman AH. Presynaptic and Postsynaptic Mesolimbic Dopamine D 3 Receptors Play Distinct Roles in Cocaine Versus Opioid Reward in Mice. Biol Psychiatry 2024; 96:752-765. [PMID: 38838841 PMCID: PMC11446657 DOI: 10.1016/j.biopsych.2024.05.020] [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: 11/26/2023] [Revised: 04/23/2024] [Accepted: 05/08/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Past research has illuminated pivotal roles of dopamine D3 receptors (D3R) in the rewarding effects of cocaine and opioids. However, the cellular and neural circuit mechanisms that underlie these actions remain unclear. METHODS We employed Cre-LoxP techniques to selectively delete D3R from presynaptic dopamine neurons or postsynaptic dopamine D1 receptor (D1R)-expressing neurons in male and female mice. We utilized RNAscope in situ hybridization, immunohistochemistry, real-time polymerase chain reaction, voltammetry, optogenetics, microdialysis, and behavioral assays (n ≥ 8 animals per group) to functionally characterize the roles of presynaptic versus postsynaptic D3R in cocaine and opioid actions. RESULTS Our results revealed D3R expression in ∼25% of midbrain dopamine neurons and ∼70% of D1R-expressing neurons in the nucleus accumbens. While dopamine D2 receptors (D2R) were expressed in ∼80% dopamine neurons, we found no D2R and D3R colocalization among these cells. Selective deletion of D3R from dopamine neurons increased exploratory behavior in novel environments and enhanced pulse-evoked nucleus accumbens dopamine release. Conversely, deletion of D3R from D1R-expressing neurons attenuated locomotor responses to D1-like and D2-like agonists. Strikingly, deletion of D3R from either cell type reduced oxycodone self-administration and oxycodone-enhanced brain-stimulation reward. In contrast, neither of these D3R deletions impacted cocaine self-administration, cocaine-enhanced brain-stimulation reward, or cocaine-induced hyperlocomotion. Furthermore, D3R knockout in dopamine neurons reduced oxycodone-induced hyperactivity and analgesia, while deletion from D1R-expressing neurons potentiated opioid-induced hyperactivity without affecting analgesia. CONCLUSIONS We dissected presynaptic versus postsynaptic D3R function in the mesolimbic dopamine system. D2R and D3R are expressed in different populations of midbrain dopamine neurons, regulating dopamine release. Mesolimbic D3R are critically involved in the actions of opioids but not cocaine.
Collapse
Affiliation(s)
- Zheng-Xiong Xi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland.
| | - Miriam E Bocarsly
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, Bethesda, Maryland
| | - Ewa Galaj
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland
| | - Briana Hempel
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland
| | - Catherine Teresi
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, Bethesda, Maryland
| | - Marlisa Shaw
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, Bethesda, Maryland
| | - Guo-Hua Bi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland; Medication Development Program, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland
| | - Chloe Jordan
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland
| | - Emily Linz
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland; Medication Development Program, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland
| | - Hannah Alton
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland; Medication Development Program, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland
| | - Gianluigi Tanda
- Medication Development Program, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Veronica A Alvarez
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, Bethesda, Maryland; National Institute of Mental Health, Center on Compulsive Behaviors, Intramural Research Program, Bethesda, Maryland
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland.
| |
Collapse
|
24
|
Gulyaeva NV, Peregud DI. An Intricated pas de deux of Addicted Brain and Body Is Orchestrated by Stress and Neuroplasticity. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1863-1867. [PMID: 39647816 DOI: 10.1134/s0006297924110014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 12/10/2024]
Abstract
Dependence on psychoactive substances is a phenomenon that is based on the alterations of common molecular and cellular mechanisms, structures and neuronal networks underlying normal brain functioning and realizing stress response, reinforcement and aversion, learning and memory. As a result, aberrant neuroplasticity states associated with somatic changes are formed, which determine the pathogenesis and symptoms of dependence and at the same time can be considered as targets for the development of therapies for such addictions. An integrative scheme of stress and neuroplastic changes participation in the formation of the vicious circle of substance use disorders based on a holistic approach is presented. This special issue of the journal focuses on the molecular mechanisms of psychoactive substance use disorders.
Collapse
Affiliation(s)
- Natalia V Gulyaeva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia.
- Research and Clinical Center for Neuropsychiatry of Moscow Healthcare Department, Moscow, 115419, Russia
| | - Danil I Peregud
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia
- Federal State Budgetary Institution "V. Serbsky National Medical Research Center for Psychiatry and Drug Addiction" of the Ministry of Health of the Russian Federation, Moscow, 119034, Russia
| |
Collapse
|
25
|
Browne CJ. Leaving an Impression: Morphine-Induced Disruptions to Brain Connectivity Persist Through Abstinence to Prime Future Drug Responses. Biol Psychiatry 2024; 96:689-690. [PMID: 39357967 DOI: 10.1016/j.biopsych.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 08/12/2024] [Indexed: 10/04/2024]
Affiliation(s)
- Caleb J Browne
- Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.
| |
Collapse
|
26
|
Liu QM, Lucas M, Badami F, Wu W, Etkin A, Yuan TF. Cortical plasticity differences in substance use disorders. FUNDAMENTAL RESEARCH 2024; 4:1351-1356. [PMID: 39734552 PMCID: PMC11670691 DOI: 10.1016/j.fmre.2023.02.015] [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: 12/14/2022] [Revised: 02/18/2023] [Accepted: 02/28/2023] [Indexed: 03/09/2023] Open
Abstract
Among substances, opiates and psychostimulants are responsible for the most significant public health problems, yet few studies have characterized their similarities or differences in the cortical plasticity of individuals with these substance related problems. This investigation utilized concurrent transcranial magnetic stimulation and electroencephalography (TMS-EEG) to examine cortical plasticity characteristics of individuals with heroin and methamphetamine related substance use disorder (SUD) relative to healthy controls. TMS-EEG data were collected from healthy control subjects (N = 35), subjects with heroin (N = 72) and methamphetamine (N = 69) use disorder. The data were analyzed using our fully-automated artifact rejection algorithm (ARTIST). Analyses were performed separately for F3, F4 and P3 stimulation sites. Linear mixed effects models were used to examine Group (heroin, methamphetamine, healthy control) x Time (pre, post single-session rTMS) interactions. To evaluate plasticity differences across groups, we observed the changes in single pulse TMS before and after single-session of rTMS. There was no change in alpha power after stimulation of the F3 or F4 sites across groups. The alpha power of the control group was significantly decreased when stimulating the P3 site, while there was no significant change in alpha power for either drug group during the same time window. The beta power of the healthy control group increased significantly when the F3 site was stimulated. In contrast, there was no significant change in either the methamphetamine or heroin group. Following a single-session of rTMS intervention, there was a significant difference in alpha-band power between the healthy control group and the two drug groups. Taking together, the study findings identified differential plasticity effects in the two types of SUD population, and highlighted the network effects of rTMS. The findings point to an exciting future path for using rTMS to test new plasticity-based interventions for treating drug addiction.
Collapse
Affiliation(s)
- Qing-Ming Liu
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
- Center for Brain, Mind and Education, Shaoxing University, Shaoxing 312000, China
- School of Psychology, Nanjing Normal University, Nanjing 210024, China
| | - Molly Lucas
- Department of Psychiatry and Behavioral Science, Stanford University, CA 94394, United States
- Wu Tsai Neuroscience Institute, Stanford University, CA 94305, United States
| | - Faizan Badami
- Department of Psychiatry and Behavioral Science, Stanford University, CA 94394, United States
- Wu Tsai Neuroscience Institute, Stanford University, CA 94305, United States
| | - Wei Wu
- Department of Psychiatry and Behavioral Science, Stanford University, CA 94394, United States
- Alto Neuroscience, Inc., Los Altos, CA 94022, United States
| | - Amit Etkin
- Department of Psychiatry and Behavioral Science, Stanford University, CA 94394, United States
- Wu Tsai Neuroscience Institute, Stanford University, CA 94305, United States
- Alto Neuroscience, Inc., Los Altos, CA 94022, United States
| | - Ti-Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| |
Collapse
|
27
|
Tian G, Bartas K, Hui M, Chen L, Vasquez JJ, Azouz G, Derdeyn P, Manville RW, Ho EL, Fang AS, Li Y, Tyler I, Setola V, Aoto J, Abbott GW, Beier KT. Molecular and circuit determinants in the globus pallidus mediating control of cocaine-induced behavioral plasticity. Neuron 2024; 112:3470-3485.e12. [PMID: 39153478 PMCID: PMC11502257 DOI: 10.1016/j.neuron.2024.07.018] [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: 07/28/2023] [Revised: 04/12/2024] [Accepted: 07/22/2024] [Indexed: 08/19/2024]
Abstract
The globus pallidus externus (GPe) is a central component of the basal ganglia circuit that acts as a gatekeeper of cocaine-induced behavioral plasticity. However, the molecular and circuit mechanisms underlying this function are unknown. Here, we show that GPe parvalbumin-positive (GPePV) cells mediate cocaine responses by selectively modulating ventral tegmental area dopamine (VTADA) cells projecting to the dorsomedial striatum (DMS). Interestingly, GPePV cell activity in cocaine-naive mice is correlated with behavioral responses following cocaine, effectively predicting cocaine sensitivity. Expression of the voltage-gated potassium channels KCNQ3 and KCNQ5 that control intrinsic cellular excitability following cocaine was downregulated, contributing to the elevation in GPePV cell excitability. Acutely activating channels containing KCNQ3 and/or KCNQ5 using the small molecule carnosic acid, a key psychoactive component of Salvia rosmarinus (rosemary) extract, reduced GPePV cell excitability and impaired cocaine reward, sensitization, and volitional cocaine intake, indicating its therapeutic potential to counteract psychostimulant use disorder.
Collapse
Affiliation(s)
- Guilian Tian
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Katrina Bartas
- Program in Mathematical, Computational, and Systems Biology, University of California, Irvine, Irvine, CA, USA
| | - May Hui
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Lingxuan Chen
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Jose J Vasquez
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Ghalia Azouz
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Pieter Derdeyn
- Program in Mathematical, Computational, and Systems Biology, University of California, Irvine, Irvine, CA, USA
| | - Rían W Manville
- Bioelectricity Laboratory, Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Erick L Ho
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Amanda S Fang
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Yuan Li
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Isabella Tyler
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Vincent Setola
- Department of Neuroscience, West Virginia University, Morgantown, WV, USA; Department of Behavioral Medicine and Psychiatry, West Virginia University, Morgantown, WV, USA
| | - Jason Aoto
- University of Colorado Anschutz School of Medicine, Department of Pharmacology, Aurora, CO, USA
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Kevin T Beier
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA; Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
28
|
Schilling L, Singleton SP, Tozlu C, Hédo M, Zhao Q, Pohl KM, Jamison K, Kuceyeski A. Sex-specific differences in brain activity dynamics of youth with a family history of substance use disorder. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.03.610959. [PMID: 39282344 PMCID: PMC11398379 DOI: 10.1101/2024.09.03.610959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
An individual's risk of substance use disorder (SUD) is shaped by a complex interplay of potent biosocial factors. Current neurodevelopmental models posit vulnerability to SUD in youth is due to an overreactive reward system and reduced inhibitory control. Having a family history of SUD is a particularly strong risk factor, yet few studies have explored its impact on brain function and structure prior to substance exposure. Herein, we utilized a network control theory approach to quantify sex-specific differences in brain activity dynamics in youth with and without a family history of SUD, drawn from a large cohort of substance-naïve youth from the Adolescent Brain Cognitive Development Study. We summarize brain dynamics by calculating transition energy, which probes the ease with which a whole brain, region or network drives the brain towards a specific spatial pattern of activation (i.e., brain state). Our findings reveal that a family history of SUD is associated with alterations in the brain's dynamics wherein: i) independent of sex, certain regions' transition energies are higher in those with a family history of SUD and ii) there exist sex-specific differences in SUD family history groups at multiple levels of transition energy (global, network, and regional). Family history-by-sex effects reveal that energetic demand is increased in females with a family history of SUD and decreased in males with a family history of SUD, compared to their same-sex counterparts with no SUD family history. Specifically, we localize these effects to higher energetic demands of the default mode network in females with a family history of SUD and lower energetic demands of attention networks in males with a family history of SUD. These results suggest a family history of SUD may increase reward saliency in males and decrease efficiency of top-down inhibitory control in females. This work could be used to inform personalized intervention strategies that may target differing cognitive mechanisms that predispose individuals to the development of SUD.
Collapse
Affiliation(s)
- Louisa Schilling
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | | | - Ceren Tozlu
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Marie Hédo
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Qingyu Zhao
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Kilian M Pohl
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Keith Jamison
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Amy Kuceyeski
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
29
|
Basso V, Döbrössy MD, Thompson LH, Kirik D, Fuller HR, Gates MA. State of the Art in Sub-Phenotyping Midbrain Dopamine Neurons. BIOLOGY 2024; 13:690. [PMID: 39336117 PMCID: PMC11428604 DOI: 10.3390/biology13090690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024]
Abstract
Dopaminergic neurons in the ventral tegmental area (VTA) and the substantia nigra pars compacta (SNpc) comprise around 75% of all dopaminergic neurons in the human brain. While both groups of dopaminergic neurons are in close proximity in the midbrain and partially overlap, development, function, and impairments in these two classes of neurons are highly diverse. The molecular and cellular mechanisms underlying these differences are not yet fully understood, but research over the past decade has highlighted the need to differentiate between these two classes of dopaminergic neurons during their development and in the mature brain. This differentiation is crucial not only for understanding fundamental circuitry formation in the brain but also for developing therapies targeted to specific dopaminergic neuron classes without affecting others. In this review, we summarize the state of the art in our understanding of the differences between the dopaminergic neurons of the VTA and the SNpc, such as anatomy, structure, morphology, output and input, electrophysiology, development, and disorders, and discuss the current technologies and methods available for studying these two classes of dopaminergic neurons, highlighting their advantages, limitations, and the necessary improvements required to achieve more-precise therapeutic interventions.
Collapse
Affiliation(s)
- Valentina Basso
- School of Medicine, Keele University, Staffordshire ST5 5BG, UK
| | - Máté D Döbrössy
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional, Neurosurgery, Medical Center, University of Freiburg, 79106 Freiburg im Breisgau, Germany
- Department of Stereotactic and Functional Neurosurgery, Medical Center, University of Freiburg, 79106 Freiburg im Breisgau, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg im Breisgau, Germany
| | - Lachlan H Thompson
- Charles Perkins Centre, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Deniz Kirik
- Brain Repair and Imaging in Neural Systems (B.R.A.I.N.S) Unit, Department of Experimental Medical Science, Lund University, BMC D11, 22184 Lund, Sweden
| | - Heidi R Fuller
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, TORCH Building, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| | - Monte A Gates
- School of Medicine, Keele University, Staffordshire ST5 5BG, UK
| |
Collapse
|
30
|
Sardari M, Mohammadpourmir F, Hosseinzadeh Sahafi O, Rezayof A. Neuronal biomarkers as potential therapeutic targets for drug addiction related to sex differences in the brain: Opportunities for personalized treatment approaches. Prog Neuropsychopharmacol Biol Psychiatry 2024; 134:111068. [PMID: 38944334 DOI: 10.1016/j.pnpbp.2024.111068] [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: 11/25/2023] [Revised: 06/19/2024] [Accepted: 06/19/2024] [Indexed: 07/01/2024]
Abstract
Biological sex disparities manifest at various stages of drug addiction, including craving, substance abuse, abstinence, and relapse. These discrepancies are underpinned by notable distinctions in neurobiological substrates, encompassing brain structures, functions, and neurotransmitter systems implicated in drug addiction. Neuronal biomarkers, such as neurotransmitters, signaling proteins, and genes may be associated with the diagnosis, prognosis, and treatment outcomes in both biological sexes afflicted by drug abuse. Sex differences in the neural reward system, mainly through dopaminergic transmission during drug abuse, can be attributed to modifications in neurotransmitter systems and signaling pathways. This results in distinct patterns of neural activation and responsiveness to addictive substances in males and females. Sex hormones, the estrus/menstrual cycle, and cerebral neurochemistry contribute to the progression of psychological and physiological dependence in both male and female individuals grappling with addiction. Moreover, the alteration of sex hormone balance and neurotransmitter release plays a pivotal role in substance use disorders, subsequently modulating cognitive functions pertinent to reward, including memory formation, decision-making, and locomotor activity. Comparative investigations reveal distinctions in brain region volume, gene expression, neuronal firing, and circuitry in substance use disorders affecting individuals of both biological sexes. This review examines prevalent substance use disorders to elucidate the impact of sex hormones as therapeutic biomarkers on the mesocorticolimbic neurotransmitter systems via diverse mechanisms within the addicted brain. We underscore the imperative necessity of considering these variations to gain a deeper comprehension of addiction mechanisms and potentially discern sex-specific neuronal biomarkers for tailored therapeutic interventions.
Collapse
Affiliation(s)
- Maryam Sardari
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Farina Mohammadpourmir
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Oveis Hosseinzadeh Sahafi
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Ameneh Rezayof
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| |
Collapse
|
31
|
Michel L, Molina P, Mameli M. The behavioral relevance of a modular organization in the lateral habenula. Neuron 2024; 112:2669-2685. [PMID: 38772374 DOI: 10.1016/j.neuron.2024.04.026] [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: 02/23/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/23/2024]
Abstract
Behavioral strategies for survival rely on the updates the brain continuously makes based on the surrounding environment. External stimuli-neutral, positive, and negative-relay core information to the brain, where a complex anatomical network rapidly organizes actions, including approach or escape, and regulates emotions. Human neuroimaging and physiology in nonhuman primates, rodents, and teleosts suggest a pivotal role of the lateral habenula in translating external information into survival behaviors. Here, we review the literature describing how discrete habenular modules-reflecting the molecular signatures, anatomical connectivity, and functional components-are recruited by environmental stimuli and cooperate to prompt specific behavioral outcomes. We argue that integration of these findings in the context of valence processing for reinforcing or discouraging behaviors is necessary, offering a compelling model to guide future work.
Collapse
Affiliation(s)
- Leo Michel
- The Department of Fundamental Neuroscience, The University of Lausanne, 1005 Lausanne, Switzerland
| | - Patricia Molina
- The Department of Fundamental Neuroscience, The University of Lausanne, 1005 Lausanne, Switzerland
| | - Manuel Mameli
- The Department of Fundamental Neuroscience, The University of Lausanne, 1005 Lausanne, Switzerland; Inserm, UMR-S 839, 75005 Paris, France.
| |
Collapse
|
32
|
Young EJ, Radnai L, Prikhodko V, Miller CA. Novel therapeutics in development for the treatment of stimulant-use disorder. Curr Opin Neurobiol 2024; 87:102898. [PMID: 39096558 DOI: 10.1016/j.conb.2024.102898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 07/14/2024] [Accepted: 07/14/2024] [Indexed: 08/05/2024]
Abstract
Misuse and accidental overdoses attributed to stimulants are escalating rapidly. These stimulants include methamphetamine, cocaine, amphetamine, ecstasy-type drugs, and prescription stimulants such as methylphenidate. Unlike opioids and alcohol, there are no therapies approved by the US Food and Drug Administration (FDA) to treat stimulant-use disorder. The high rate of relapse among this population highlights the insufficiency of current treatment options, which are limited to abstinence support programs and behavioral modification therapies. Here, we briefly outline recent regulatory actions taken by FDA to help support the development of new stimulant use disorder treatments and highlight several new therapeutics in the clinical development pipeline.
Collapse
Affiliation(s)
- Erica J Young
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA; Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA; Myosin Therapeutics, Jupiter, FL, USA
| | - Laszlo Radnai
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA; Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | | | - Courtney A Miller
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA; Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA.
| |
Collapse
|
33
|
Lan YP, Ding CZ, Xia JX, Yang YZ, Zhao YB. Analysis of the functional role and mRNA expression of GABA B R in the nucleus accumbens of cocaine-addicted rats. J Chin Med Assoc 2024; 87:754-764. [PMID: 38860774 DOI: 10.1097/jcma.0000000000001119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Drug addiction is a social and medical problem that must be urgently addressed. The nucleus accumbens (NAc) is closely related to addiction-related learning memory, and γ-aminobutyric acid type B receptor (GABA B R) is a potential target for the treatment of drug addiction. However, the role of GABA B R activity levels in the NAc in cocaine addiction is unclear. METHODS In this study, we established an animal model of cocaine dependence, modulated the level of GABA B R activity, applied a conditioned place preference assay (CPP) to assess the role of the NAc in reconsolidation of addiction memory, evaluated learning and memory functions by behavioral experiments, examined the expression of GB1, GB2, cyclic adenosine monophosphate response element binding protein (CREB), p-CREB, protein kinase A (PKA), protein kinase (ERK), and Brain-derived neurotrophic factor (BDNF) in the NAc by molecular biology experiments, and screened differentially significantly expressed genes by transcriptome sequencing. RESULTS Our study showed that the GABA B receptor agonist baclofen (BLF) had a significant effect on locomotor distance in rats, promoted an increase in GABA levels and significantly inhibited the PKA and ERK1/2/CREB/BDNF signaling pathways. Moreover, transcriptome sequencing showed that GABA B R antagonist intervention identified a total of 21 upregulated mRNAs and 21 downregulated mRNAs. The differentially expressed (DE) mRNA genes were mainly enriched in tyrosine metabolism; however, further study is needed. CONCLUSION GABA B R activity in the NAc is involved in the regulation of cocaine addiction and may play an important role through key mRNA pathways.
Collapse
Affiliation(s)
- Yan-Ping Lan
- Department of Neurosurgery, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
- Key Laboratory of Craniocerebral Diseases, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Chen-Zhe Ding
- Department of Neurosurgery, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| | - Jian-Xue Xia
- Department of Neurosurgery, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| | - Yun-Zhen Yang
- Department of Neurosurgery, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| | - Yan-Bin Zhao
- Department of Neurosurgery, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| |
Collapse
|
34
|
Martínez-Rivera FJ, Yim YY, Godino A, Minier-Toribio A, Tofani S, Holt LM, Torres-Berrío A, Futamura R, Browne CJ, Markovic T, Hamilton PJ, Neve RL, Nestler EJ. Cell-Type-Specific Regulation of Cocaine Reward by the E2F3a Transcription Factor in Nucleus Accumbens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602609. [PMID: 39026727 PMCID: PMC11257579 DOI: 10.1101/2024.07.08.602609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
The development of drug addiction is characterized by molecular changes in brain reward regions that lead to the transition from recreational to compulsive drug use. These neurobiological processes in brain reward regions, such as the nucleus accumbens (NAc), are orchestrated in large part by transcriptional regulation. Our group recently identified the transcription factor E2F3a as a novel regulator of cocaine's rewarding effects and gene expression regulation in the NAc of male mice. Despite this progress, no information is available about the role of E2F3a in regulating cocaine reward at the sex- and cell-specific levels. Here, we used male and female mice expressing Cre-recombinase in either D1- or D2-type medium spiny neurons (MSNs) combined with viral-mediated gene transfer to bidirectionally control levels of E2F3a in a cell-type-specific manner in the NAc during conditioned place preference (CPP) to cocaine. Our findings show that selective overexpression of E2F3a in D1-MSNs increased cocaine CPP in both male and female mice, whereas opposite effects were observed under knockdown conditions. In contrast, equivalent E2F3a manipulations in D2-MSNs had no significant effects. To further explore the role of E2F3a in sophisticated operant and motivated behaviors, we performed viral manipulations of all NAc neurons in combination with cocaine self-administration and behavioral economics procedures in rats and demonstrated that E2F3a regulates sensitivity aspects of cocaine seeking and taking. These results confirm E2F3a as a central substrate of cocaine reward and demonstrate that this effect is mediated in D1-MSNs, thereby providing increased knowledge of cocaine action at the transcriptional level.
Collapse
|
35
|
Zang D, Yang X, Wang H, Li Z, Ma Y, Liu J, Mei X, Li S, Feng J, Shi X, Tan Z. Study on the antidepressant activity of (2R,6R; 2S,6S)-Hydroxynorketamine (HNK) and its derivatives. Clinics (Sao Paulo) 2024; 79:100435. [PMID: 38996724 PMCID: PMC11301192 DOI: 10.1016/j.clinsp.2024.100435] [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: 03/20/2024] [Revised: 05/23/2024] [Accepted: 06/12/2024] [Indexed: 07/14/2024] Open
Abstract
OBJECTIVE This study mainly explores (2R,6R; 2S,6S)-HNK and its compounds whether there are antidepressant effects. METHODS Four HNK compounds were obtained from 2-(Chlorophenyl) Cyclopentylmethanone. Forced swimming test, locomotor sensitization test, and conditioned location preference test were used to screen the antidepressant activity of the synthesized target compounds. RESULTS In the case of 10 mg HNK treatment, compared with saline, the immobile time of mice in the HNK group, I5 group and I6 group at 1 h and 7 days had statistical significance. In the case of 10 mg HNK treatment, compared with saline, the immobile time of compound C and D groups in the glass cylinder area was significantly different. In the locomotor sensitization test, the movement distance of compound C and D groups on day 15 and day 7 mice increased significantly compared with the first day. In the conditioned place preference experiment, compound C and compound D induced conditioned place preference in mice compared with the Veh group. CONCLUSION The results of the forced swimming test, locomotor sensitization test, and conditioned location preference test showed that compounds C and D may have certain anti-depressant activity. However, HNK exerts a rapid and significant antidepressant effect within 1 week, but the duration is short.
Collapse
Affiliation(s)
- Dongdong Zang
- Department of Neonatology, Shenzhen Children's Hospital, Shenzhen City, Guangdong Province, China
| | - Xuemei Yang
- Health Management Center, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen City, Guangdong Province, China
| | - Hao Wang
- Department of Neurosurgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen City, Guangdong Province, China; Guangdong Engineering Technological Research Center for Nervous anatomy and Related Clinical Applications, Shenzhen City, Guangdong Province, China
| | - Zhenxing Li
- Health Management Center, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen City, Guangdong Province, China
| | - Yanjun Ma
- Shenzhen Ruijian Biotechnology Co., LTD, Shenzhen City, Guangdong Province, China
| | - Jianxi Liu
- Shenzhen Ruijian Biotechnology Co., LTD, Shenzhen City, Guangdong Province, China
| | - Xi Mei
- Zhuhai Pengkun Biomedicine Technology Co. LTD, Zhuhai City, Guangdong Province, China
| | - Shupeng Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University, Shenzhen City, Guangdong Province, China
| | - Jinxing Feng
- Department of Neonatology, Shenzhen Children's Hospital, Shenzhen City, Guangdong Province, China
| | - Xin Shi
- Department of Neurosurgery, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy Centre, Shenzhen University, Shenzhen City, Guangdong Province, China
| | - Zhen Tan
- Department of Neonatology, Shenzhen Children's Hospital, Shenzhen City, Guangdong Province, China; Health Management Center, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen City, Guangdong Province, China.
| |
Collapse
|
36
|
Mai Y, Cheng Z, Wang Z, Hu T, Zhang Y, Yuan X, Xu X, Fan Y, Ge F, Shi P, Wang J, Yang X, Guan X. Pathological polarizations from microglia to astrocyte contributes to spatial memory deficit in methamphetamine abstinence mice. Cereb Cortex 2024; 34:bhae281. [PMID: 38981852 DOI: 10.1093/cercor/bhae281] [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: 02/19/2024] [Revised: 06/17/2024] [Accepted: 06/22/2024] [Indexed: 07/11/2024] Open
Abstract
Previously, we found that dCA1 A1-like polarization of astrocytes contributes a lot to the spatial memory deficit in methamphetamine abstinence mice. However, the underlying mechanism remains unclear, resulting in a lack of promising therapeutic targets. Here, we found that methamphetamine abstinence mice exhibited an increased M1-like microglia and A1-like astrocytes, together with elevated levels of interleukin 1α and tumor necrosis factor α in dCA1. In vitro, the M1-like BV2 microglia cell medium, containing high levels of Interleukin 1α and tumor necrosis factor α, elevated A1-like polarization of astrocytes, which weakened their capacity for glutamate clearance. Locally suppressing dCA1 M1-like microglia activation with minocycline administration attenuated A1-like polarization of astrocytes, ameliorated dCA1 neurotoxicity, and, most importantly, rescued spatial memory in methamphetamine abstinence mice. The effective time window of minocycline treatment on spatial memory is the methamphetamine exposure period, rather than the long-term methamphetamine abstinence.
Collapse
Affiliation(s)
- Yuning Mai
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Zhen Cheng
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Ze Wang
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Tao Hu
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Yuanyuan Zhang
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Xiya Yuan
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Xing Xu
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Yu Fan
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Feifei Ge
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Pengbo Shi
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Jun Wang
- Department of Toxicology, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, China
| | - Xin Yang
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Xiaowei Guan
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| |
Collapse
|
37
|
Blaze J, Browne CJ, Futamura R, Javidfar B, Zachariou V, Nestler EJ, Akbarian S. tRNA epitranscriptomic alterations associated with opioid-induced reward-seeking and long-term opioid withdrawal in male mice. Neuropsychopharmacology 2024; 49:1276-1284. [PMID: 38332016 PMCID: PMC11224224 DOI: 10.1038/s41386-024-01813-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/10/2024]
Abstract
DNA cytosine methylation has been documented as a potential epigenetic mechanism of transcriptional regulation underlying opioid use disorder. However, methylation of RNA cytosine residues, which would drive another level of biological influence as an epitranscriptomic mechanism of gene and protein regulation has not been studied in the context of addiction. Here, we probed whether chronic morphine exposure could alter tRNA cytosine methylation (m5C) and resulting expression levels in the medial prefrontal cortex (mPFC), a brain region crucial for reward processing and executive function that exhibits opioid-induced molecular restructuring. We identified dynamic changes in glycine tRNA (tRNAGlyGCC) cytosine methylation, corresponding to altered expression levels of this tRNA at multiple timepoints following 15 days of daily morphine. Additionally, a robust increase in methylation, coupled with decreased expression, was present after 30 days of withdrawal, suggesting that repeated opioid administration produces changes to the tRNA regulome long after discontinuation. Furthermore, forebrain-wide knockout of neuronal Nsun2, a tRNA methyltransferase, was associated with disruption of opioid conditioned place preference, and this effect was recapitulated by regional mPFC Nsun2 knockout. Taken together, these studies provide a foundational link between the regulation of tRNA cytosine methylation and opioid reward and highlight the tRNA machinery as a potential therapeutic target in addiction.
Collapse
Affiliation(s)
- Jennifer Blaze
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Caleb J Browne
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Rita Futamura
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Behnam Javidfar
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Venetia Zachariou
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pharmacology, Physiology, and Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Eric J Nestler
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Schahram Akbarian
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
38
|
Hayase T. Interrelated involvement of the endocannabinoid/endovanilloid (TRPV1) systems and epigenetic processes in anxiety- and working memory impairment-related behavioural effects of nicotine as a stressor. Addict Biol 2024; 29:10.1111/adb.13421. [PMID: 38963015 PMCID: PMC11222983 DOI: 10.1111/adb.13421] [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: 02/08/2023] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 07/05/2024]
Abstract
The addictive use of nicotine contained in tobacco is associated with stressor-like emotional and cognitive effects such as anxiety and working memory impairment, and the involvement of epigenetic mechanisms such as histone acetylation has recently been reported. Although the precise nature of behavioural plasticity remains unclear, both anxiogenic- and working memory impairment-like effects were observed in the present experimental model of mice treated with repeated subcutaneous nicotine and/or immobilization stress, and these effects were commonly attenuated by the histone deacetylase (HDAC) inhibitors that induce histone acetylation. Such HDAC inhibitor-induced resilience was mimicked by ligands for the endocannabinoid (ECB) system, a neurotransmitter system that is closely associated with nicotine-induced addiction-related behaviours: the anxiogenic-like effects were mitigated by the cannabinoid type 1 (CB1) agonist arachidonylcyclopropylamide (ACPA), whereas the working memory impairment-like effects were mitigated by the CB1 antagonist SR 141716A. Moreover, the effects of the HDAC inhibitors were also mimicked by ligands for the endovanilloid (transient receptor potential vanilloid 1 [TRPV1]) system, a system that shares common characteristics with the ECB system: the anxiogenic-like effects were mitigated by the TRPV1 antagonist capsazepine, whereas the working memory impairment-like effects were mitigated by the TRPV1 agonist olvanil. Notably, the HDAC inhibitor-induced anxiolytic-like effects were attenuated by SR 141716A, which were further counteracted by capsazepine, whereas the working memory improvement-like effects were attenuated by capsazepine, which were further counteracted by SR 141716A. These results suggest the contribution of interrelated control of the ECB/TRPV1 systems and epigenetic processes such as histone acetylation to novel therapeutic approaches.
Collapse
Affiliation(s)
- Tamaki Hayase
- Department of Legal MedicineKyoto UniversityKyotoJapan
| |
Collapse
|
39
|
Zeine F, Jafari N, Baron D, Bowirrat A, Pinhasov A, Norling B, Martinez KC, Nami M, Manavi N, Sunder K, Rabin DM, Bagchi D, Khalsa J, Gold MS, Sipple D, Barzegar M, Bodhanapati J, Khader W, Carney P, Dennen CA, Gupta A, Elman I, Badgaiyan RD, Modestino EJ, Thanos PK, Hanna C, McLaughlin T, Cadet JL, Soni D, Braverman ER, Barh D, Giordano J, Edwards D, Ashford JW, Gondre-Lewis MC, Gilley E, Murphy KT, Lewandrowski KU, Sharafshah A, Makale M, Fuehrlein B, Blum K. Solving the Global Opioid Crisis: Incorporating Genetic Addiction Risk Assessment with Personalized Dopaminergic Homeostatic Therapy and Awareness Integration Therapy. JOURNAL OF ADDICTION PSYCHIATRY 2024; 8:50-95. [PMID: 39635461 PMCID: PMC11615735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Objectives The opioid crisis in the last few decades has mounted to a global level, impacting all areas of socioeconomic, demographic, geographic, and cultural boundaries. Traditional treatments have not been deemed to show the degree of efficacy necessary to address the crisis. The authors of this review paper have set forth an unprecedented and in-depth look into multi-factorial determinants that have contributed to the opioid crisis becoming global and multi-faceted. Methods For this narrative review/opinion article, we searched PsychINFO, PubMed, Google Scholar, and Web of Science databases to identify relevant articles on topics including the "opioid crisis," "opioid mechanisms," "genetics and epigenetics," "neuropharmacology," and "clinical aspects of opioid treatment and prevention." Since this was not a systematic review the articles selected could represent unitential bias. Results Despite some success achieved through Opioid Substitution Therapy (OST) in harm reduction, the annual mortality toll in the US alone surpasses 106,699 individuals, a figure expected to climb to 165,000 by 2025. Data from the Substance Abuse and Mental Health Services Administration's (SAMHSA) National Survey on Drug Abuse and Health (NSDUH) reveals that approximately 21.4% of individuals in the US engaged in illicit drug use in 2020, with 40.3 million individuals aged 12 or older experiencing a Substance Use Disorder (SUD). Provisional figures from the Centers for Disease Control and Prevention (CDC) indicate a troubling 15% increase in overdose deaths in 2021, rising from 93,655 in 2020 to 107,622, with opioids accounting for roughly 80,816 of these deaths. Conclusions We advocate reevaluating the "standard of care" and shifting towards inducing dopamine homeostasis by manipulating key neurotransmitter systems within the brain's reward cascade. We propose a paradigm shift towards a novel "standard of care" that begins with incorporating Genetic Addiction Risk Severity (GARS) testing to assess pre-addiction risk and vulnerability to opioid-induced addiction; emphasis should be placed on inducing dopamine homeostasis through safe and non-addictive alternatives like KB220, and comprehensive treatment approaches that address psychological, spiritual, and societal aspects of addiction through Awareness Integration Therapy (AIT).
Collapse
Affiliation(s)
- Foojan Zeine
- Awareness Integration Institute, San Clemente, USA
- Department of Health Science, California State University, Long Beach, USA
| | - Nicole Jafari
- Department of Applied Clinical Psychology, The Chicago School of Professional Psychology, Los Angeles, USA
- Division of Personalized Medicine, Cross-Cultural Research and Educational Institute, San Clemente, USA
| | - David Baron
- Center for Exercise and Sport Mental Health, Western University Health Sciences, Pomona, USA
| | - Abdalla Bowirrat
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Albert Pinhasov
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Brian Norling
- MEMS Precision Technology, Inc., Santa Barbara, USA
- Acies Biomedical, Inc. Santa Barbara, USA
| | - Kathleen Carter Martinez
- Division of General Education-Berkeley College, Paramus Campus, New Jersey, USA
- Chey-Wind Center for Trauma and Healing, Peru, USA
| | - Mohammad Nami
- Brain, Cognition, and Behavior Unit, Brain Hub Academy, Dubai, UAE
| | - Nima Manavi
- College of Osteopathic Medicine, Western University of Health Sciences, Pomona, USA
| | - Keerthy Sunder
- Department of Psychiatry, University of California, UC Riverside School of Medicine, Riverside, USA
- Division of Neuromodulation Research, Karma Doctors and Karma TMS, Palm Springs, USA
| | | | - Debasis Bagchi
- Division of Nutrigenomics, Victory Nutrition International, LLC, Bonita Springs, USA
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, USA
| | - Jag Khalsa
- Department of Medicine, University of Maryland School of Medicine, Baltimore, USA
| | - Mark S. Gold
- Department of Psychiatry, Washington University, School of Medicine, St. Louis, USA
| | - Daniel Sipple
- Minnesota Institute for Pain Management, Minnesota, USA
| | - Mojtaba Barzegar
- Hamad Medical Corporation, National Center for Cancer Care and Research (NCCCR), Doha, Qatar
| | - Jothsna Bodhanapati
- Division of Neuromodulation Research, Karma Doctors and Karma TMS, Palm Springs, USA
| | - Waseem Khader
- Karma Doctors, Palm Springs, USA
- Global Medical Detox Center, Menifee, CA, USA
| | - Paul Carney
- Division of Pediatric Neurology, University of Missouri, School of Medicine, Columbia, USA
| | - Catherine A. Dennen
- Department of Family Medicine, Jefferson Health Northeast, Philadelphia, USA
| | | | - Igor Elman
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel, Israel
- Department of Psychiatry, Harvard School of Medicine, Cambridge, USA
| | - Rajendra D. Badgaiyan
- Department of Psychiatry, Case Western University School of Medicine, The Metro Health System, Cleveland, USA
- Department of Psychiatry, Mt. Sinai University, Ichan School of Medicine, New York, USA
| | | | - Panayotis K. Thanos
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel, Israel
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Research Institute on Addictions, University at Buffalo, Buffalo, USA
| | - Colin Hanna
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Research Institute on Addictions, University at Buffalo, Buffalo, USA
| | - Thomas McLaughlin
- Division of Primary Care Research, Reward Deficiency Syndrome Clinics of America, Inc. Austin, USA
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIH National Institute on Drug Abuse, Baltimore, USA
| | - Diwanshu Soni
- College of Osteopathic Medicine, Western University of Health Sciences, Pomona, USA
| | - Eric R. Braverman
- Division of Clinical Neurological Research, The Kenneth Blum Neurogenetic and Behavioral Institute, LLC., Austin, USA
| | - Debmalya Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology, Nonakuri, Purba Medinipur, West Bengal, India
| | | | | | - J. Wesson Ashford
- Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, USA
| | | | | | - Kevin T. Murphy
- Department of Radiation Oncology, University of California, San Diego, La Jolla, USA
| | - Kai-Uwe Lewandrowski
- Division of Personalized Pain Therapy Research, Center for Advanced Spine Care of Southern Arizona, Tucson, USA
- Department of Orthopaedics, Fundación Universitaria Sanitas, Bogotá, D.C., Colombia
- Department of Orthopedics, Hospital Universitário Gaffrée Guinle Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alireza Sharafshah
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Milan Makale
- Department of Radiation Oncology, University of California, San Diego, La Jolla, USA
| | - Brian Fuehrlein
- Department of Psychiatry, School of Medicine, Yale University, New Haven, USA
| | - Kenneth Blum
- Center for Exercise and Sport Mental Health, Western University Health Sciences, Pomona, USA
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel, Israel
- Division of Primary Care Research, Reward Deficiency Syndrome Clinics of America, Inc. Austin, USA
- Division of Clinical Neurological Research, The Kenneth Blum Neurogenetic and Behavioral Institute, LLC., Austin, USA
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology, Nonakuri, Purba Medinipur, West Bengal, India
- JC’s Recovery and Counseling Center, Hollywood, USA
- Department of Psychiatry, University of Vermont, Burlington, USA
- Department of Psychiatry, Wright University Boonshoft School of Medicine, Dayton, USA
- Institute of Psychology, ELTE Eötvös Loránd University, Budapest, Hungary
- Center for Advanced Spine Care of Southern Arizona, Tucson, USA
| |
Collapse
|
40
|
Brida KL, Jorgensen ET, Phillips RA, Newman CE, Tuscher JJ, Morring EK, Zipperly ME, Ianov L, Montgomery KD, Tippani M, Hyde TM, Maynard KR, Martinowich K, Day JJ. Reelin marks cocaine-activated striatal ensembles, promotes neuronal excitability, and regulates cocaine reward. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.599348. [PMID: 38948801 PMCID: PMC11212904 DOI: 10.1101/2024.06.17.599348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Drugs of abuse activate defined neuronal ensembles in brain reward structures such as the nucleus accumbens (NAc), which are thought to promote the enduring synaptic, circuit, and behavioral consequences of drug exposure. While the molecular and cellular effects arising from experience with drugs like cocaine are increasingly well understood, the mechanisms that sculpt NAc ensemble participation are largely unknown. Here, we leveraged unbiased single-nucleus transcriptional profiling to identify expression of the secreted glycoprotein Reelin (encoded by the Reln gene) as a marker of cocaine-activated neuronal ensembles within the rat NAc. Multiplexed in situ detection confirmed selective expression of the immediate early gene Fos in Reln+ neurons after cocaine experience, and also revealed enrichment of Reln mRNA in Drd1 + medium spiny neurons (MSNs) in both the rat and human brain. Using a novel CRISPR interference strategy enabling selective Reln knockdown in the adult NAc, we observed altered expression of genes linked to calcium signaling, emergence of a transcriptional trajectory consistent with loss of cocaine sensitivity, and a striking decrease in MSN intrinsic excitability. At the behavioral level, loss of Reln prevented cocaine locomotor sensitization, abolished cocaine place preference memory, and decreased cocaine self-administration behavior. Together, these results identify Reelin as a critical mechanistic link between ensemble participation and cocaine-induced behavioral adaptations.
Collapse
|
41
|
Opioid drugs drive electrical insulation of reward circuitry. Nature 2024:10.1038/d41586-024-01630-3. [PMID: 38840006 DOI: 10.1038/d41586-024-01630-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
|
42
|
Yalçın B, Pomrenze MB, Malacon K, Drexler R, Rogers AE, Shamardani K, Chau IJ, Taylor KR, Ni L, Contreras-Esquivel D, Malenka RC, Monje M. Myelin plasticity in the ventral tegmental area is required for opioid reward. Nature 2024; 630:677-685. [PMID: 38839962 PMCID: PMC11186775 DOI: 10.1038/s41586-024-07525-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/07/2024] [Indexed: 06/07/2024]
Abstract
All drugs of abuse induce long-lasting changes in synaptic transmission and neural circuit function that underlie substance-use disorders1,2. Another recently appreciated mechanism of neural circuit plasticity is mediated through activity-regulated changes in myelin that can tune circuit function and influence cognitive behaviour3-7. Here we explore the role of myelin plasticity in dopaminergic circuitry and reward learning. We demonstrate that dopaminergic neuronal activity-regulated myelin plasticity is a key modulator of dopaminergic circuit function and opioid reward. Oligodendroglial lineage cells respond to dopaminergic neuronal activity evoked by optogenetic stimulation of dopaminergic neurons, optogenetic inhibition of GABAergic neurons, or administration of morphine. These oligodendroglial changes are evident selectively within the ventral tegmental area but not along the axonal projections in the medial forebrain bundle nor within the target nucleus accumbens. Genetic blockade of oligodendrogenesis dampens dopamine release dynamics in nucleus accumbens and impairs behavioural conditioning to morphine. Taken together, these findings underscore a critical role for oligodendrogenesis in reward learning and identify dopaminergic neuronal activity-regulated myelin plasticity as an important circuit modification that is required for opioid reward.
Collapse
Affiliation(s)
- Belgin Yalçın
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Matthew B Pomrenze
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Karen Malacon
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Richard Drexler
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Abigail E Rogers
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Kiarash Shamardani
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Isabelle J Chau
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Kathryn R Taylor
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Lijun Ni
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | | | - Robert C Malenka
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford, CA, USA.
| |
Collapse
|
43
|
Tian G, Bartas K, Hui M, Chen L, Vasquez JJ, Azouz G, Derdeyn P, Manville RW, Ho EL, Fang AS, Li Y, Tyler I, Setola V, Aoto J, Abbott GW, Beier KT. Molecular and circuit determinants in the globus pallidus mediating control of cocaine-induced behavioral plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596557. [PMID: 38853899 PMCID: PMC11160764 DOI: 10.1101/2024.05.29.596557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
The globus pallidus externus (GPe) is a central component of the basal ganglia circuit, receiving strong input from the indirect pathway and regulating a variety of functions, including locomotor output and habit formation. We recently showed that it also acts as a gatekeeper of cocaine-induced behavioral plasticity, as inhibition of parvalbumin-positive cells in the GPe (GPe PV ) prevents the development of cocaine-induced reward and sensitization. However, the molecular and circuit mechanisms underlying this function are unknown. Here we show that GPe PV cells control cocaine reward and sensitization by inhibiting GABAergic neurons in the substantia nigra pars reticulata (SNr GABA ), and ultimately, selectively modulating the activity of ventral tegmental area dopamine (VTA DA ) cells projecting to the lateral shell of the nucleus accumbens (NAcLat). A major input to GPe PV cells is the indirect pathway of the dorsomedial striatum (DMS D 2 ), which receives DAergic innervation from collaterals of VTA DA →NAcLat cells, making this a closed-loop circuit. Cocaine likely facilitates reward and sensitization not directly through actions in the GPe, but rather in the upstream DMS, where the cocaine-induced elevation of DA triggers a depression in DMS D 2 cell activity. This cocaine-induced elevation in DA levels can be blocked by inhibition of GPe PV cells, closing the loop. Interestingly, the level of GPe PV cell activity prior to cocaine administration is correlated with the extent of reward and sensitization that animals experience in response to future administration of cocaine, indicating that GPe PV cell activity is a key predictor of future behavioral responses to cocaine. Single nucleus RNA-sequencing of GPe cells indicated that genes encoding voltage-gated potassium channels KCNQ3 and KCNQ5 that control intrinsic cellular excitability are downregulated in GPe PV cells following a single cocaine exposure, contributing to the elevation in GPe PV cell excitability. Acutely activating channels containing KCNQ3 and/or KCNQ5 using the small molecule carnosic acid, a key psychoactive component of Salvia rosmarinus (rosemary) extract, reduced GPe PV cell excitability and also impaired cocaine reward, sensitization, and volitional cocaine intake, indicating its potential as a therapeutic to counteract psychostimulant use disorder. Our findings illuminate the molecular and circuit mechanisms by which the GPe orchestrates brain-wide changes in response to cocaine that are required for reward, sensitization, and self-administration behaviors.
Collapse
|
44
|
Tizabi Y, Getachew B, Hauser SR, Tsytsarev V, Manhães AC, da Silva VDA. Role of Glial Cells in Neuronal Function, Mood Disorders, and Drug Addiction. Brain Sci 2024; 14:558. [PMID: 38928557 PMCID: PMC11201416 DOI: 10.3390/brainsci14060558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/19/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Mood disorders and substance use disorder (SUD) are of immense medical and social concern. Although significant progress on neuronal involvement in mood and reward circuitries has been achieved, it is only relatively recently that the role of glia in these disorders has attracted attention. Detailed understanding of the glial functions in these devastating diseases could offer novel interventions. Here, following a brief review of circuitries involved in mood regulation and reward perception, the specific contributions of neurotrophic factors, neuroinflammation, and gut microbiota to these diseases are highlighted. In this context, the role of specific glial cells (e.g., microglia, astroglia, oligodendrocytes, and synantocytes) on phenotypic manifestation of mood disorders or SUD are emphasized. In addition, use of this knowledge in the potential development of novel therapeutics is touched upon.
Collapse
Affiliation(s)
- Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, 520 W Street NW, Washington, DC 20059, USA;
| | - Bruk Getachew
- Department of Pharmacology, Howard University College of Medicine, 520 W Street NW, Washington, DC 20059, USA;
| | - Sheketha R. Hauser
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Vassiliy Tsytsarev
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Alex C. Manhães
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, IBRAG, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20550-170, RJ, Brazil
| | - Victor Diogenes Amaral da Silva
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-100, BA, Brazil;
| |
Collapse
|
45
|
Baslam A, Kabdy H, Chait Y, Azraida H, El Yazouli L, Aboufatima R, Chait A, Baslam M. Gut Microbiome-Mediated Mechanisms in Alleviating Opioid Addiction with Aqueous Extract of Anacyclus pyrethrum. Biomedicines 2024; 12:1152. [PMID: 38927359 PMCID: PMC11200529 DOI: 10.3390/biomedicines12061152] [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: 04/12/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
The escalating rates of morbidity and mortality associated with opioid use disorder (OUD) have spurred a critical need for improved treatment outcomes. This study aimed to investigate the impact of prolonged exposure to Fentanyl, a potent opioid, on behavior, biochemical markers, oxidative stress, and the composition of the gut microbiome. Additionally, we sought to explore the therapeutic potential of Anacyclus pyrethrum in mitigating the adverse effects of Fentanyl withdrawal. The study unveiled that chronic Fentanyl administration induced a withdrawal syndrome characterized by elevated cortisol levels (12.09 mg/mL, compared to 6.3 mg/mL for the control group). This was accompanied by heightened anxiety, indicated by a reduction in time spent and entries made into the open arm in the Elevated Plus Maze Test, as well as depressive-like behaviors, manifested through increased immobility time in the Forced Swim Test. Additionally, Fentanyl exposure correlated with decreased gut microbiome density and diversity, coupled with heightened oxidative stress levels, evidenced by elevated malondialdehyde (MDA) and reduced levels of catalase (CAT) and superoxide dismutase (SOD). However, both post- and co-administration of A. pyrethrum exhibited substantial improvements in these adverse effects, effectively alleviating symptoms associated with OUD withdrawal syndrome and eliciting positive influences on gut microbiota. In conclusion, this research underscores the therapeutic potential of A. pyrethrum in managing Fentanyl withdrawal symptoms. The findings indicate promising effects in alleviating behavioral impairments, reducing stress, restoring gut microbiota, and mitigating oxidative stress, offering valuable insights for addressing the challenges of OUD treatment.
Collapse
Affiliation(s)
- Abdelmounaim Baslam
- Laboratory of Pharmacology, Neurobiology, Anthropobiology and Environment, Department of Biology, Faculty of Sciences Semlalia, Cadi Ayyad University, Marrakech 40000, Morocco; (A.B.); (H.A.)
| | - Hamid Kabdy
- Laboratory of Pharmacology, Neurobiology, Anthropobiology and Environment, Department of Biology, Faculty of Sciences Semlalia, Cadi Ayyad University, Marrakech 40000, Morocco; (A.B.); (H.A.)
| | - Yassine Chait
- Agadir Souss Massa University Hospital, Faculty of Medicine and Pharmacy, Ibn Zohr University, Agadir 80000, Morocco
| | - Hajar Azraida
- Laboratory of Pharmacology, Neurobiology, Anthropobiology and Environment, Department of Biology, Faculty of Sciences Semlalia, Cadi Ayyad University, Marrakech 40000, Morocco; (A.B.); (H.A.)
| | - Loubna El Yazouli
- Laboratory of Pharmacology, Neurobiology, Anthropobiology and Environment, Department of Biology, Faculty of Sciences Semlalia, Cadi Ayyad University, Marrakech 40000, Morocco; (A.B.); (H.A.)
| | - Rachida Aboufatima
- Laboratory of Biological Engineering, Faculty of Sciences and Technology, Sultan Moulay Slimane University, Beni Mellal 23000, Morocco
| | - Abderrahman Chait
- Laboratory of Pharmacology, Neurobiology, Anthropobiology and Environment, Department of Biology, Faculty of Sciences Semlalia, Cadi Ayyad University, Marrakech 40000, Morocco; (A.B.); (H.A.)
| | - Marouane Baslam
- Center of Agrobiotechnology and Bioengineering, Research Unit Labelled CNRST (Centre AgroBiotech-URL-7 CNRST-05), Cadi Ayyad University, Marrakech 40000, Morocco
- Laboratory of Agro-Food, Biotechnologies and Valorization of Plant Bioresources (AGROBIOVAL), Department of Biology, Faculty of Science Semlalia, Cadi Ayyad University (UCA), Marrakech 40000, Morocco
- Laboratory of Biochemistry, Department of Applied Biological Chemistry, Faculty of Agriculture, University of Niigata, Niigata 950-2181, Japan
- GrowSmart, Seoul 07516, Republic of Korea
| |
Collapse
|
46
|
Bornhoft KN, Prohofsky J, O'Neal TJ, Wolff AR, Saunders BT. Valence ambiguity dynamically shapes striatal dopamine heterogeneity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.17.594692. [PMID: 38798567 PMCID: PMC11118546 DOI: 10.1101/2024.05.17.594692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Adaptive decision making relies on dynamic updating of learned associations where environmental cues come to predict positive and negatively valenced stimuli, such as food or threat. Flexible cue-guided behaviors depend on a network of brain systems, including dopamine signaling in the striatum, which is critical for learning and maintenance of conditioned behaviors. Critically, it remains unclear how dopamine signaling encodes multi-valent, dynamic learning contexts, where positive and negative associations must be rapidly disambiguated. To understand this, we employed a Pavlovian discrimination paradigm, where cues predicting positive and negative outcomes were intermingled during conditioning sessions, and their meaning was serially reversed across training. We found that rats readily distinguished these cues, and updated their behavior rapidly upon valence reversal. Using fiber photometry, we recorded dopamine signaling in three major striatal subregions -,the dorsolateral striatum (DLS), the nucleus accumbens core, and the nucleus accumbens medial shell - and found heterogeneous responses to positive and negative conditioned cues and their predicted outcomes. Valence ambiguity introduced by cue reversal reshaped striatal dopamine on different timelines: nucleus accumbens core and shell signals updated more readily than those in the DLS. Together, these results suggest that striatal dopamine flexibly encodes multi-valent learning contexts, and these signals are dynamically modulated by changing contingencies to resolve ambiguity about the meaning of environmental cues.
Collapse
|
47
|
Darcq E, Kieffer BL. Neuroscience and addiction research: current advances and perspectives. J Neural Transm (Vienna) 2024; 131:405-408. [PMID: 38492013 DOI: 10.1007/s00702-024-02763-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2024] [Indexed: 03/18/2024]
Affiliation(s)
- Emmanuel Darcq
- Strasbourg Translational Neuroscience and Psychiatry, Université de Strasbourg (UNISTRA), INSERM UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | - Brigitte L Kieffer
- Strasbourg Translational Neuroscience and Psychiatry, Université de Strasbourg (UNISTRA), INSERM UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France.
| |
Collapse
|
48
|
Gerra MC, Dallabona C, Cecchi R. Epigenetic analyses in forensic medicine: future and challenges. Int J Legal Med 2024; 138:701-719. [PMID: 38242965 PMCID: PMC11003920 DOI: 10.1007/s00414-024-03165-8] [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: 04/20/2023] [Accepted: 01/09/2024] [Indexed: 01/21/2024]
Abstract
The possibility of using epigenetics in forensic investigation has gradually risen over the last few years. Epigenetic changes with their dynamic nature can either be inherited or accumulated throughout a lifetime and be reversible, prompting investigation of their use across various fields. In forensic sciences, multiple applications have been proposed, such as the discrimination of monozygotic twins, identifying the source of a biological trace left at a crime scene, age prediction, determination of body fluids and tissues, human behavior association, wound healing progression, and determination of the post-mortem interval (PMI). Despite all these applications, not all the studies considered the impact of PMI and post-sampling effects on the epigenetic modifications and the tissue-specificity of the epigenetic marks.This review aims to highlight the substantial forensic significance that epigenetics could support in various forensic investigations. First, basic concepts in epigenetics, describing the main epigenetic modifications and their functions, in particular, DNA methylation, histone modifications, and non-coding RNA, with a particular focus on forensic applications, were covered. For each epigenetic marker, post-mortem stability and tissue-specificity, factors that should be carefully considered in the study of epigenetic biomarkers in the forensic context, have been discussed. The advantages and limitations of using post-mortem tissues have been also addressed, proposing directions for these innovative strategies to analyze forensic specimens.
Collapse
Affiliation(s)
- Maria Carla Gerra
- Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area Delle Scienze 11a, Viale Delle Scienze 11a, 43124, Parma, PR, Italy
| | - Cristina Dallabona
- Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area Delle Scienze 11a, Viale Delle Scienze 11a, 43124, Parma, PR, Italy.
| | - Rossana Cecchi
- Department of Medicine and Surgery, University of Parma, Via Antonio Gramsci 14, 43126, Parma, PR, Italy
| |
Collapse
|
49
|
Carvalho L, Lasek AW. It is not just about transcription: involvement of brain RNA splicing in substance use disorders. J Neural Transm (Vienna) 2024; 131:495-503. [PMID: 38396082 PMCID: PMC11055753 DOI: 10.1007/s00702-024-02740-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 01/04/2024] [Indexed: 02/25/2024]
Abstract
Alternative splicing is a co-transcriptional process that significantly contributes to the molecular landscape of the cell. It plays a multifaceted role in shaping gene transcription, protein diversity, and functional adaptability in response to environmental cues. Recent studies demonstrate that drugs of abuse have a profound impact on alternative splicing patterns within different brain regions. Drugs like alcohol and cocaine modify the expression of genes responsible for encoding splicing factors, thereby influencing alternative splicing of crucial genes involved in neurotransmission, neurogenesis, and neuroinflammation. Notable examples of these alterations include alcohol-induced changes in splicing factors such as HSPA6 and PCBP1, as well as cocaine's impact on PTBP1 and SRSF11. Beyond the immediate effects of drug exposure, recent research has shed light on the role of alternative splicing in contributing to the risk of substance use disorders (SUDs). This is exemplified by exon skipping events in key genes like ELOVL7, which can elevate the risk of alcohol use disorder. Lastly, drugs of abuse can induce splicing alterations through epigenetic modifications. For example, cocaine exposure leads to alterations in levels of trimethylated lysine 36 of histone H3, which exhibits a robust association with alternative splicing and serves as a reliable predictor for exon exclusion. In summary, alternative splicing has emerged as a critical player in the complex interplay between drugs of abuse and the brain, offering insights into the molecular underpinnings of SUDs.
Collapse
Affiliation(s)
- Luana Carvalho
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 1220 E. Broad ST, Box 980613, Richmond, VA, 23298, USA.
| | - Amy W Lasek
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 1220 E. Broad ST, Box 980613, Richmond, VA, 23298, USA
| |
Collapse
|
50
|
Fang T, Liu MN, Liu MQ, Tian XY, Zhang XJ, Liu F, Hao W, Wu N, Li H, Li J. A preliminary study on the association of single nucleotide polymorphisms and methylation of dopamine system-related genes with psychotic symptoms in patients with methamphetamine use disorder. Eur J Neurosci 2024; 59:1428-1440. [PMID: 38151046 DOI: 10.1111/ejn.16238] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/29/2023]
Abstract
Methamphetamine use disorder (MAUD) can substantially jeopardize public security due to its high-risk social psychology and behaviour. Given that the dopamine reward system is intimately correlated with MAUD, we investigated the association of single nucleotide polymorphisms (SNPs), as well as methylation status of dopamine receptor type 4 (DRD4), catechol-O-methyltransferase (COMT) genes, and paranoid and motor-impulsive symptoms in MAUD patients. A total of 189 MAUD patients participated in our study. Peripheral blood samples were used to detect 3 SNPs and 35 CpG units of methylation in the DRD4 gene promoter region and 5 SNPs and 39 CpG units in the COMT gene. MAUD patients with the DRD4 rs1800955 C allele have a lower percentage of paranoid symptoms than those with the rs1800955 TT allele. Individuals with paranoid symptoms exhibited a reduced methylation degree at a particular DRD4 CpG2.3 unit. The interaction of the DRD4 rs1800955 C allele and the reduced DRD4CpG2.3 methylation degree were associated with a lower occurrence of paranoid symptoms. Meanwhile, those with the COMT rs4818 CC allele had lower motor-impulsivity scores in MAUD patients but greater COMT methylation levels in the promoter region and methylation degree at the COMT CpG 51.52 unit. Therefore, based only on the COMT rs4818 CC polymorphism, there was a negative correlation between COMT methylation and motor-impulsive scores. Our preliminary results provide a clue that the combination of SNP genotype and methylation status of the DRD4 and COMT genes serve as biological indicators for the prevalence of relatively high-risk psychotic symptoms in MAUD patients.
Collapse
Affiliation(s)
- Ting Fang
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Meng-Nan Liu
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Meng-Qi Liu
- National Clinical Research Center for Mental Disorders, and Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiao-Yu Tian
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Xiao-Jie Zhang
- National Clinical Research Center for Mental Disorders, and Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Feng Liu
- Compulsory Detoxification Center of Changsha Public Security Bureau, Changsha, China
| | - Wei Hao
- National Clinical Research Center for Mental Disorders, and Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ning Wu
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Hong Li
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Jin Li
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|