1
|
Matt SM, Nolan R, Manikandan S, Agarwal Y, Channer B, Oteju O, Daniali M, Canagarajah JA, LuPone T, Mompho K, Runner K, Nickoloff-Bybel E, Li B, Niu M, Schlachetzki JCM, Fox HS, Gaskill PJ. Dopamine-driven increase in IL-1β in myeloid cells is mediated by differential dopamine receptor expression and exacerbated by HIV. J Neuroinflammation 2025; 22:91. [PMID: 40122818 PMCID: PMC11931822 DOI: 10.1186/s12974-025-03403-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 03/01/2025] [Indexed: 03/25/2025] Open
Abstract
The catecholamine neurotransmitter dopamine is classically known for regulation of central nervous system (CNS) functions such as reward, movement, and cognition. Increasing evidence also indicates that dopamine regulates critical functions in peripheral organs and is an important immunoregulatory factor. We have previously shown that dopamine increases NF-κB activity, inflammasome activation, and the production of inflammatory cytokines such as IL-1β in human macrophages. As myeloid lineage cells are central to the initiation and resolution of acute inflammatory responses, dopamine-mediated dysregulation of these functions could both impair the innate immune response and exacerbate chronic inflammation. However, the exact pathways by which dopamine drives myeloid inflammation are not well defined, and studies in both rodent and human systems indicate that dopamine can impact the production of inflammatory mediators through both D1-like dopamine receptors (DRD1, DRD5) and D2-like dopamine receptors (DRD2, DRD3, and DRD4). Therefore, we hypothesized that dopamine-mediated production of IL-1β in myeloid cells is regulated by the ratio of different dopamine receptors that are activated. Our data in primary human monocyte-derived macrophages (hMDM) indicate that DRD1 expression is necessary for dopamine-mediated increases in IL-1β, and that changes in the expression of DRD2 and other dopamine receptors can alter the magnitude of the dopamine-mediated increase in IL-1β. Mature hMDM have a high D1-like to D2-like receptor ratio, which is different relative to monocytes and peripheral blood mononuclear cells (PBMCs). We further confirm in human microglia cell lines that a high ratio of D1-like to D2-like receptors promotes dopamine-induced increases in IL-1β gene and protein expression using pharmacological inhibition or overexpression of dopamine receptors. RNA-sequencing of dopamine-treated microglia shows that genes encoding functions in IL-1β signaling pathways, microglia activation, and neurotransmission increased with dopamine treatment. Finally, using HIV as an example of a chronic inflammatory disease that is substantively worsened by comorbid substance use disorders (SUDs) that impact dopaminergic signaling, we show increased effects of dopamine on inflammasome activation and IL-1β in the presence of HIV in both human macrophages and microglia. These data suggest that use of addictive substances and dopamine-modulating therapeutics could dysregulate the innate inflammatory response and exacerbate chronic neuroimmunological conditions like HIV. Thus, a detailed understanding of dopamine-mediated changes in inflammation, in particular pathways regulating IL-1β, will be critical to effectively tailor medication regimens.
Collapse
Affiliation(s)
- Stephanie M Matt
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Rachel Nolan
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
- Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Samyuktha Manikandan
- Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Yash Agarwal
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
- Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Breana Channer
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
- Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Oluwatofunmi Oteju
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Marzieh Daniali
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Joanna A Canagarajah
- Graduate School of Biomedical Sciences and Professional Studies, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Teresa LuPone
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Krisna Mompho
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Kaitlyn Runner
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Emily Nickoloff-Bybel
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - Benjamin Li
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Meng Niu
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Howard S Fox
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Peter J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA.
| |
Collapse
|
2
|
Worhunsky PD, Mignosa MM, Gallezot JD, Pittman B, Nabulsi NB, Stryjewski A, Jalilian-Khave L, Trinko R, DiLeone RJ, Carson RE, Malison RT, Potenza MN, Angarita GA. Vitamin D's Capacity to Increase Amphetamine-Induced Dopamine Release in Healthy Humans: A Clinical Translational [ 11C]-PHNO Positron Emission Tomography Study. Biol Psychiatry 2025; 97:651-658. [PMID: 39395473 PMCID: PMC11839384 DOI: 10.1016/j.biopsych.2024.09.028] [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: 03/24/2024] [Revised: 09/11/2024] [Accepted: 09/18/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND Dopaminergic tone and phasic release have transdiagnostic relevance. Preclinical research suggests that the active form of vitamin D, calcitriol, increases subcortical tyrosine hydroxylase, D2/D3 receptors, and amphetamine-stimulated dopamine release in rodents. Comparable studies have not been conducted in humans. METHODS Healthy, vitamin D-sufficient adults (N = 18, 32.8 ± 6.6 years; 33% female) participated in a randomized, double-blind, placebo-controlled within-subjects study involving 4 total scans over 2 visits consisting of same-day preamphetamine and postamphetamine (0.3 mg/kg) [11C]-PHNO positron emission tomography scanning to examine D2/D3 receptor availability (nondisplaceable binding potential [BPND]) following active calcitriol (1.5 μg night before experimental day and 1.5 μg morning of experimental day) or placebo at least 6 days apart. Parametric images of [11C]-PHNO positron emission tomography BPND were computed using a simplified reference tissue model with the cerebellum as reference. Blood samples were acquired to measure serum calcitriol, amphetamine, and calcium levels. Regions of interest examined were the dorsal caudate, dorsal putamen, ventral striatum, globus pallidus, and substantia nigra. RESULTS For preamphetamine scans, there was a medication × region of interest interaction (F4,153 = 2.59, p = .039) and a main effect of medication (F1,153 = 4.88, p = .029) on BPND, with higher BPND values on calcitriol in the ventral striatum (t153 = 2.89, p = .004) and dorsal putamen (t153 = 2.15, p = .033). There was a main effect of medication on postamphetamine change in BPND (F4,153 = 5.93, p = .016), with greater decreases in calcitriol in the ventral striatum (t153 = 3.00, p = .003), substantia nigra (t153 = 2.49, p = .014), and dorsal caudate (t153 = 2.29, p = .023). CONCLUSIONS Results provide translational support for vitamin D to target dopaminergic tone, with implications for clinical disorders that involve dysregulated dopamine function.
Collapse
Affiliation(s)
- Patrick D Worhunsky
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Marcella M Mignosa
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut; The Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, Connecticut
| | - Jean-Dominique Gallezot
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Brian Pittman
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Nabeel B Nabulsi
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Adam Stryjewski
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Laya Jalilian-Khave
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Richard Trinko
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut; The Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, Connecticut
| | - Ralph J DiLeone
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut; The Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, Connecticut; Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Robert T Malison
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut; The Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, Connecticut
| | - Marc N Potenza
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut; Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut; Connecticut Council on Problem Gambling, Wethersfield, Connecticut; Child Study Center, Yale University School of Medicine, New Haven, Connecticut; Connecticut Mental Health Center, New Haven, Connecticut
| | - Gustavo A Angarita
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut; The Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, Connecticut.
| |
Collapse
|
3
|
Klausen M, Knudsen G, Vilsbøll T, Fink‐Jensen A. Effects of GLP-1 Receptor Agonists in Alcohol Use Disorder. Basic Clin Pharmacol Toxicol 2025; 136:e70004. [PMID: 39891507 PMCID: PMC11786240 DOI: 10.1111/bcpt.70004] [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: 12/12/2024] [Accepted: 01/20/2025] [Indexed: 02/03/2025]
Abstract
In the search for novel treatment strategies for alcohol use disorder (AUD), glucagon-like peptide-1 (GLP-1) receptor agonists (GLP-1RAs) approved for treating Type 2 diabetes and obesity have caught much attention. GLP-1 is a naturally occurring peptide produced in the small intestines and the brain, regulating plasma glucose levels and satiety. This focused review will report on the preclinical studies, case stories, register-based cohort studies, brain-imaging data and secondary analysis of clinical data supporting the role of GLP-1RAs as a novel treatment of AUD. Several clinical trials are ongoing, examining the potential effects of the GLP-1RA semaglutide in AUD.
Collapse
Affiliation(s)
- Mette Kruse Klausen
- Psychiatric Centre Copenhagen, Mental Health Services in the Capitol Region of DenmarkCopenhagen University Hospital FrederiksbergFrederiksbergDenmark
| | - Gitte Moos Knudsen
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Neurobiology Research UnitCopenhagen University Hospital RigshospitaletCopenhagenDenmark
| | - Tina Vilsbøll
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Clinical ResearchSteno Diabetes Center CopenhagenCopenhagenDenmark
| | - Anders Fink‐Jensen
- Psychiatric Centre Copenhagen, Mental Health Services in the Capitol Region of DenmarkCopenhagen University Hospital FrederiksbergFrederiksbergDenmark
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
4
|
Greydanus DE, Nazeer A, Patel DR. Opioid use and abuse in adolescents and young adults; dealing with science, laws and ethics: Charming the COBRAS. Dis Mon 2025; 71:101853. [PMID: 39809600 DOI: 10.1016/j.disamonth.2025.101853] [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] [Indexed: 01/16/2025]
Abstract
The subject of substance use disorders in the pediatric population remains a disturbing conundrum for clinicians, researchers and society in general. Many of our youth are at risk of being damaged and even killed by drug addictions that result from the collision of rapidly developing as well as vulnerable central nervous systems encountering the current global drug addiction crisis. A major motif of this chemical calamity is opioid use disorder in adolescents and young adults that was stimulated by the 19th century identification of such highly addictive drugs as morphine, heroin and a non-opiate, cocaine. This analysis focuses on the pervasive presence of opioid drugs such as heroin and fentanyl that has become a major tragedy in the 21st century arising from an overall substance use and misuse phenomenon rampant in global society. Themes covered in this article include the history of addictive drugs in humans, diagnostic terms in use, the role of neurobiology in drug addiction, and current psychopharmacologic approaches to opioid overdose as well as addiction. Our youth are continuously confronted by dangers of high-risk behaviors including death and injury from opioid use disorders due to their central nervous system neuroplasticity as well as the widespread availability of these harmful chemicals. Healthcare professionals should actively assist our youth who unknowingly and even innocently encounter this deadly menace in the 21st century.
Collapse
Affiliation(s)
- Donald E Greydanus
- Department of Pediatric and Adolescent Medicine, Western Michigan University, Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States.
| | - Ahsan Nazeer
- Division of Child and Adolescent Psychiatry, Sidra Medicine/Weill Cornell Medicine, Doha, Qatar
| | - Dilip R Patel
- Department of Pediatric and Adolescent Medicine, Western Michigan University, Homer Stryker M.D. School of Medicine, Kalamazoo, MI, USA
| |
Collapse
|
5
|
Jeffery N, Mock PY, Yang K, Tham CL, Israf DA, Li H, Wang X, Lam KW. Therapeutic targeting of neuroinflammation in methamphetamine use disorder. Future Med Chem 2025; 17:237-257. [PMID: 39727147 PMCID: PMC11749361 DOI: 10.1080/17568919.2024.2447226] [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: 10/02/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
Methamphetamine (METH) is a highly addictive illicit psychostimulant with a significant annual fatality rate. Emerging studies highlight its role in neuroinflammation and a range of neurological disorders. This review examines the current landscape of potential drug targets for managing neuroinflammation in METH use disorders (MUDs), with a particular focus on the rationale behind targeting Toll-like receptor 4 (TLR4), the NLR family pyrin domain containing 3 (NLRP3) inflammasome, and other promising targets. Given the multifactorial neurological effects of METH, including cognitive impairment and neurodegeneration, addressing METH-induced neuroinflammation has shown considerable promise in partially mitigating the damaging effects on the central nervous system and improving behavioral outcomes. This article provides an overview of the existing understanding while charting a promising path forward for developing innovative MUD treatments, focusing on neuroinflammation as a therapeutic target. Targeting neuroinflammation in METH-induced neurological disorders shows significant promise in mitigating cognitive impairment and neurodegeneration, offering a potential therapeutic strategy for improving outcomes in MUD. While challenges remain in optimizing treatments, ongoing research into combination therapies, novel drug delivery systems, and neuroprotective agents suggests a positive outlook for more effective interventions.
Collapse
Affiliation(s)
- Natasha Jeffery
- Centre for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Phooi Yan Mock
- Centre for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Kun Yang
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Chau Ling Tham
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
- Natural Medicine and Product Research Laboratory (NaturMeds), Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Daud Ahmad Israf
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
- Natural Medicine and Product Research Laboratory (NaturMeds), Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Hongyuan Li
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, China
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, China
- Department of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, China
| | - Kok Wai Lam
- Centre for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- Structural Biology and Protein Engineering Research Group, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| |
Collapse
|
6
|
Girotti M, Bulin SE, Carreno FR. Effects of chronic stress on cognitive function - From neurobiology to intervention. Neurobiol Stress 2024; 33:100670. [PMID: 39295772 PMCID: PMC11407068 DOI: 10.1016/j.ynstr.2024.100670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 08/30/2024] [Accepted: 09/01/2024] [Indexed: 09/21/2024] Open
Abstract
Exposure to chronic stress contributes considerably to the development of cognitive impairments in psychiatric disorders such as depression, generalized anxiety disorder (GAD), obsessive-compulsive disorder (OCD), post-traumatic stress disorder (PTSD), and addictive behavior. Unfortunately, unlike mood-related symptoms, cognitive impairments are not effectively treated by available therapies, a situation in part resulting from a still incomplete knowledge of the neurobiological substrates that underly cognitive domains and the difficulty in generating interventions that are both efficacious and safe. In this review, we will present an overview of the cognitive domains affected by stress with a specific focus on cognitive flexibility, behavioral inhibition, and working memory. We will then consider the effects of stress on neuronal correlates of cognitive function and the factors which may modulate the interaction of stress and cognition. Finally, we will discuss intervention strategies for treatment of stress-related disorders and gaps in knowledge with emerging new treatments under development. Understanding how cognitive impairment occurs during exposure to chronic stress is crucial to make progress towards the development of new and effective therapeutic approaches.
Collapse
Affiliation(s)
| | - Sarah E. Bulin
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, 7703 Floyd Curl Dr., San Antonio, TX, 78229, USA
| | - Flavia R. Carreno
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, 7703 Floyd Curl Dr., San Antonio, TX, 78229, USA
| |
Collapse
|
7
|
Shen H, Ma Z, Hans E, Duan Y, Bi GH, Chae YC, Bonifazi A, Battiti FO, Newman AH, Xi ZX, Yang Y. Involvement of dopamine D3 receptor in impulsive choice decision-making in male rats. Neuropharmacology 2024; 257:110051. [PMID: 38917939 PMCID: PMC11401648 DOI: 10.1016/j.neuropharm.2024.110051] [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: 04/13/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 06/27/2024]
Abstract
Impulsive decision-making has been linked to impulse control disorders and substance use disorders. However, the neural mechanisms underlying impulsive choice are not fully understood. While previous PET imaging and autoradiography studies have shown involvement of dopamine and D2/3 receptors in impulsive behavior, the roles of distinct D1, D2, and D3 receptors in impulsive decision-making remain unclear. In this study, we used a food reward delay-discounting task (DDT) to identify low- and high-impulsive rats, in which low-impulsive rats exhibited preference for large delayed reward over small immediate rewards, while high-impulsive rats showed the opposite preference. We then examined D1, D2, and D3 receptor gene expression using RNAscope in situ hybridization assays. We found that high-impulsive male rats exhibited lower levels of D2 and D3, and particularly D3, receptor expression in the nucleus accumbens (NAc), with no significant changes in the insular, prelimbic, and infralimbic cortices. Based on these findings, we further explored the role of the D3 receptor in impulsive decision-making. Systemic administration of a selective D3 receptor agonist (FOB02-04) significantly reduced impulsive choices in high-impulsive rats but had no effects in low-impulsive rats. Conversely, a selective D3 receptor antagonist (VK4-116) produced increased both impulsive and omission choices in both groups of rats. These findings suggest that impulsive decision-making is associated with a reduction in D3 receptor expression in the NAc. Selective D3 receptor agonists, but not antagonists, may hold therapeutic potentials for mitigating impulsivity in high-impulsive subjects.
Collapse
Affiliation(s)
- Hui Shen
- Neuroimaging Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Zilu Ma
- Neuroimaging Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Emma Hans
- Neuroimaging Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Ying Duan
- Neuroimaging Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Guo-Hua Bi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Yurim C Chae
- Neuroimaging Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Alessandro Bonifazi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Francisco O Battiti
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Amy Hauck Newman
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Zheng-Xiong Xi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA.
| | - Yihong Yang
- Neuroimaging Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA.
| |
Collapse
|
8
|
Doyle MR, Beltran NM, Bushnell MSA, Syed M, Acosta V, Desai M, Rice KC, Serafine KM, Gould GG, Daws LC, Collins GT. Effects of access condition on substance use disorder-like phenotypes in male and female rats self-administering MDPV or cocaine. Drug Alcohol Depend 2024; 263:112408. [PMID: 39141975 PMCID: PMC11714059 DOI: 10.1016/j.drugalcdep.2024.112408] [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: 03/19/2024] [Revised: 07/23/2024] [Accepted: 08/06/2024] [Indexed: 08/16/2024]
Abstract
Substance use disorder (SUD) is a heterogeneous disorder, where severity, symptoms, and patterns of use vary across individuals. Yet, when rats self-administer cocaine under short-access conditions, their behavior tends to be well-regulated, though individual differences can emerge with long- or intermittent-access. In contrast, significant individual differences emerge when rats self-administer 3,4-methylenedioxypyrovalerone (MDPV), even under short-access conditions, wherein ~30 % of rats exhibit high levels of drug-taking. This study assessed SUD-like phenotypes of male and female rats self-administering MDPV or cocaine by comparing level of drug intake, responding during periods of signaled drug unavailability, and sensitivity to footshock punishment to determine whether: (1) under short-access conditions, rats that self-administer MDPV will exhibit a more robust SUD-like phenotype than rats that self-administer cocaine; (2) female rats will have a more severe phenotype than male rats; and (3) compared to short-access, long- and intermittent-access to MDPV or cocaine self-administration will result in a more robust SUD-like phenotype. Compared to cocaine, rats that self-administered MDPV exhibited a more severe phenotype, even under short-access conditions. Long- and intermittent-access to cocaine and MDPV temporarily altered drug-taking patterns but did not systematically change SUD-like phenotypes. Behavioral and quantitative autoradiography studies suggest phenotypic differences are not due to expression of dopamine transporter, dopamine D2 or D3 receptors, or 5-HT1B, 5-HT2A, or 5-HT2C receptors. This study suggests individuals who use synthetic cathinones may be at greater risk for developing a SUD, and short-access MDPV self-administration may provide a useful method to study the transition to disordered substance use in humans.
Collapse
Affiliation(s)
- Michelle R Doyle
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Nina M Beltran
- Department of Psychology, University of Texas at El Paso, El Paso, TX, USA
| | - Mark S A Bushnell
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Maaz Syed
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Valeria Acosta
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Marisa Desai
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Kenner C Rice
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism - Intramural Research Program, Bethesda, MD, USA
| | | | - Georgianna G Gould
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Lynette C Daws
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA; Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Gregory T Collins
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA; South Texas Veterans Health Care System, San Antonio, TX, USA.
| |
Collapse
|
9
|
Ballester J, Marchand WR, Philip NS. Transcranial magnetic stimulation for methamphetamine use disorder: A scoping review within the neurocircuitry model of addiction. Psychiatry Res 2024; 338:115995. [PMID: 38852478 PMCID: PMC11209858 DOI: 10.1016/j.psychres.2024.115995] [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: 03/05/2024] [Revised: 05/21/2024] [Accepted: 05/28/2024] [Indexed: 06/11/2024]
Abstract
The use of methamphetamine in the United States is increasing, contributing now to the "fourth wave" in the national opioid epidemic crisis. People who suffer from methamphetamine use disorder (MUD) have a higher risk of death. No pharmacological interventions are approved by the FDA and psychosocial interventions are only moderately effective. Transcranial Magnetic Stimulation (TMS) is a relatively novel FDA-cleared intervention for the treatment of Major Depressive Disorder (MDD) and other neuropsychiatric conditions. Several lines of research suggest that TMS could be useful for the treatment of addictive disorders, including MUD. We will review those published clinical trials that show potential effects on craving reduction of TMS when applied over the dorsolateral prefrontal cortex (DLPFC) also highlighting some limitations that affect their generalizability and applicability. We propose the use of the Koob and Volkow's neurocircuitry model of addiction as a frame to explain the brain effects of TMS in patients with MUD. We will finally discuss new venues that could lead to a more individualized and effective treatment of this complex disorder including the use of neuroimaging, the exploration of different areas of the brain such as the frontopolar cortex or the salience network and the use of biomarkers.
Collapse
Affiliation(s)
- J Ballester
- Substance Abuse Residential Rehabilitation Treatment Program, VA Salt Lake City Health Care System, 500 Foothill Drive, Salt Lake City, UT 84148, USA; Department of Psychiatry, School of Medicine, University of Utah, 501 Chipeta Way, Salt Lake City, UT 84108, USA.
| | - W R Marchand
- Department of Psychiatry, School of Medicine, University of Utah, 501 Chipeta Way, Salt Lake City, UT 84108, USA; VISN-19 Whole Health Flagship Site, VA Salt Lake City Health Care System, 500 Foothill Drive, Salt Lake City, UT 84148, USA; Animal, Dairy and Veterinary Sciences, Utah State University, 4815 Old Main Hill, Logan, UT 84322, USA
| | - N S Philip
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, RI, USA; VA RR&D Center for Neurorestoration and Neurotechnology, VA Providence Healthcare System, Providence, RI, USA
| |
Collapse
|
10
|
Pochapski JA, Gómez-A A, Stringfield SJ, Jaggers H, Boettiger CA, Da Cunha C, Robinson DL. Adolescent alcohol exposure persistently alters orbitofrontal cortical encoding of Pavlovian conditional stimulus components in female rats. Sci Rep 2024; 14:13775. [PMID: 38877100 PMCID: PMC11178901 DOI: 10.1038/s41598-024-64036-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 06/04/2024] [Indexed: 06/16/2024] Open
Abstract
Exposure to alcohol during adolescence impacts cortical and limbic brain regions undergoing maturation. In rodent models, long-term effects on behavior and neurophysiology have been described after adolescent intermittent ethanol (AIE), especially in males. We hypothesized that AIE in female rats increases conditional approach to a reward-predictive cue and corresponding neuronal activity in the orbitofrontal cortex (OFC) and nucleus accumbens (NAc). We evaluated behavior and neuronal firing after AIE (5 g/kg intragastric) or water (CON) in adult female rats. Both AIE and CON groups expressed a ST phenotype, and AIE marginally increased sign-tracking (ST) and decreased goal-tracking (GT) metrics. NAc neurons exhibited phasic firing patterns to the conditional stimulus (CS), with no differences between groups. In contrast, neuronal firing in the OFC of AIE animals was greater at CS onset and offset than in CON animals. During reward omission, OFC responses to CS offset normalized to CON levels, but enhanced OFC firing to CS onset persisted in AIE. We suggest that the enhanced OFC neural activity observed in AIE rats to the CS could contribute to behavioral inflexibility. Ultimately, AIE persistently impacts the neurocircuitry of reward-motivated behavior in female rats.
Collapse
Affiliation(s)
- Jose A Pochapski
- Laboratorio de Fisiologia e Farmacologia do Sistema Nervoso Central, Department of Pharmacology, Universidade Federal do Parana, Curitiba, PR, Brazil
| | - Alexander Gómez-A
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Hannah Jaggers
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Charlotte A Boettiger
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Claudio Da Cunha
- Laboratorio de Fisiologia e Farmacologia do Sistema Nervoso Central, Department of Pharmacology, Universidade Federal do Parana, Curitiba, PR, Brazil
- Department of Biochemistry, Universidade Federal do Parana, Curitiba, PR, Brazil
| | - Donita L Robinson
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
11
|
Matt SM, Nolan R, Manikandan S, Agarwal Y, Channer B, Oteju O, Daniali M, Canagarajah JA, LuPone T, Mompho K, Runner K, Nickoloff-Bybel E, Li B, Niu M, Schlachetzki JCM, Fox HS, Gaskill PJ. Dopamine-driven Increase in IL-1β in Myeloid Cells is Mediated by Differential Dopamine Receptor Expression and Exacerbated by HIV. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.09.598137. [PMID: 38915663 PMCID: PMC11195146 DOI: 10.1101/2024.06.09.598137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
The catecholamine neurotransmitter dopamine is classically known for regulation of central nervous system (CNS) functions such as reward, movement, and cognition. Increasing evidence also indicates that dopamine regulates critical functions in peripheral organs and is an important immunoregulatory factor. We have previously shown that dopamine increases NF-κB activity, inflammasome activation, and the production of inflammatory cytokines such as IL-1β in human macrophages. As myeloid lineage cells are central to the initiation and resolution of acute inflammatory responses, dopamine-mediated dysregulation of these functions could both impair the innate immune response and exacerbate chronic inflammation. However, the exact pathways by which dopamine drives myeloid inflammation are not well defined, and studies in both rodent and human systems indicate that dopamine can impact the production of inflammatory mediators through both D1-like dopamine receptors (DRD1, DRD5) and D2-like dopamine receptors (DRD2, DRD3, and DRD4). Therefore, we hypothesized that dopamine-mediated production of IL-1β in myeloid cells is regulated by the ratio of different dopamine receptors that are activated. Our data in primary human monocyte-derived macrophages (hMDM) indicate that DRD1 expression is necessary for dopamine-mediated increases in IL-1β, and that changes in the expression of DRD2 and other dopamine receptors can alter the magnitude of the dopamine-mediated increase in IL-1β. Mature hMDM have a high D1-like to D2-like receptor ratio, which is different relative to monocytes and peripheral blood mononuclear cells (PBMCs). We further confirm in human microglia cell lines that a high ratio of D1-like to D2-like receptors promotes dopamine-induced increases in IL-1β gene and protein expression using pharmacological inhibition or overexpression of dopamine receptors. RNA-sequencing of dopamine-treated microglia shows that genes encoding functions in IL-1β signaling pathways, microglia activation, and neurotransmission increased with dopamine treatment. Finally, using HIV as an example of a chronic inflammatory disease that is substantively worsened by comorbid substance use disorders (SUDs) that impact dopaminergic signaling, we show increased effects of dopamine on inflammasome activation and IL-1β in the presence of HIV in both human macrophages and microglia. These data suggest that use of addictive substances and dopamine-modulating therapeutics could dysregulate the innate inflammatory response and exacerbate chronic neuroimmunological conditions like HIV. Thus, a detailed understanding of dopamine-mediated changes in inflammation, in particular pathways regulating IL-1β, will be critical to effectively tailor medication regimens.
Collapse
|
12
|
Ghaderi S, Amani Rad J, Hemami M, Khosrowabadi R. Dysfunctional feedback processing in male methamphetamine abusers: Evidence from neurophysiological and computational approaches. Neuropsychologia 2024; 197:108847. [PMID: 38460774 DOI: 10.1016/j.neuropsychologia.2024.108847] [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: 08/07/2023] [Revised: 01/24/2024] [Accepted: 02/28/2024] [Indexed: 03/11/2024]
Abstract
Methamphetamine use disorder (MUD) as a major public health risk is associated with dysfunctional neural feedback processing. Although dysfunctional feedback processing in people who are substance dependent has been explored in several behavioral, computational, and electrocortical studies, this mechanism in MUDs requires to be well understood. Furthermore, the current understanding of latent components of their behavior such as learning speed and exploration-exploitation dilemma is still limited. In addition, the association between the latent cognitive components and the related neural mechanisms also needs to be explored. Therefore, in this study, the underlying neurocognitive mechanisms of feedback processing of such impairment, and age/gender-matched healthy controls are evaluated within a probabilistic learning task with rewards and punishments. Mathematical modeling results based on the Q-learning paradigm suggested that MUDs show less sensitivity in distinguishing optimal options. Additionally, it may be worth noting that MUDs exhibited a slight decrease in their ability to learn from negative feedback compared to healthy controls. Also through the lens of underlying neural mechanisms, MUDs showed lower theta power at the medial-frontal areas while responding to negative feedback. However, other EEG measures of reinforcement learning including feedback-related negativity, parietal-P300, and activity flow from the medial frontal to lateral prefrontal regions, remained intact in MUDs. On the other hand, the elimination of the linkage between value sensitivity and medial-frontal theta activity in MUDs was observed. The observed dysfunction could be due to the adverse effects of methamphetamine on the cortico-striatal dopamine circuit, which is reflected in the anterior cingulate cortex activity as the most likely region responsible for efficient behavior adjustment. These findings could help us to pave the way toward tailored therapeutic approaches.
Collapse
Affiliation(s)
- Sadegh Ghaderi
- Institute for Cognitive and Brain Sciences, Shahid Beheshti University, Tehran, Iran
| | - Jamal Amani Rad
- Institute for Cognitive and Brain Sciences, Shahid Beheshti University, Tehran, Iran.
| | - Mohammad Hemami
- Institute for Cognitive and Brain Sciences, Shahid Beheshti University, Tehran, Iran
| | - Reza Khosrowabadi
- Institute for Cognitive and Brain Sciences, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
13
|
Singh N. Neurobiological basis for the application of yoga in drug addiction. Front Psychiatry 2024; 15:1373866. [PMID: 38699450 PMCID: PMC11064691 DOI: 10.3389/fpsyt.2024.1373866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 04/04/2024] [Indexed: 05/05/2024] Open
|
14
|
Heinz A, Gutwinski S, Bahr NS, Spanagel R, Di Chiara G. Does compulsion explain addiction? Addict Biol 2024; 29:e13379. [PMID: 38588458 PMCID: PMC11001268 DOI: 10.1111/adb.13379] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/18/2024] [Indexed: 04/10/2024]
Abstract
One of the leading drug addiction theories states that habits and the underlying neural process of a ventral to dorsal striatal shift are the building blocks of compulsive drug-seeking behaviour and that compulsion is the maladaptive persistence of responding despite adverse consequences. Here we discuss that compulsive behaviour as defined primarily from the perspective of animal experimentation falls short of the clinical phenomena and their neurobiological correlates. Thus for the human condition, the concept of compulsive habbits should be critically addressed and potentially revised.
Collapse
Affiliation(s)
- Andreas Heinz
- Department of Psychiatry and Neuroscience|CCM, NeuroCure Clinical Research Center, Berlin Institute of Health CCM, Charité‐Universitätsmedizin Berlin, Freie Universität BerlinHumboldt‐Universität zu BerlinBerlinGermany
- German Center for Mental Health (DZPG)Berlin‐Potsdam
| | - Stefan Gutwinski
- Department of Psychiatry and Neuroscience|CCM, NeuroCure Clinical Research Center, Berlin Institute of Health CCM, Charité‐Universitätsmedizin Berlin, Freie Universität BerlinHumboldt‐Universität zu BerlinBerlinGermany
| | - Nadja Samia Bahr
- Department of Psychiatry and Neuroscience|CCM, NeuroCure Clinical Research Center, Berlin Institute of Health CCM, Charité‐Universitätsmedizin Berlin, Freie Universität BerlinHumboldt‐Universität zu BerlinBerlinGermany
- German Center for Mental Health (DZPG)Berlin‐Potsdam
| | - Rainer Spanagel
- Institute for Psychopharmacology, Medical Faculty Mannheim, Central Institute of Mental Health (CIMH)Heidelberg UniversityMannheimGermany
| | - Gaetano Di Chiara
- Department of Biomedical Sciences, University of CagliariCittadella Universitaria di MonserratoCagliariItaly
- Neuroscience InstituteNational Research Council of Italy (CNR)CagliariItaly
| |
Collapse
|
15
|
Doyle MR, Beltran NM, Bushnell MSA, Syed M, Acosta V, Desai M, Rice KC, Serafine KM, Gould GG, Daws LC, Collins GT. Effects of access condition on substance use disorder-like phenotypes in male and female rats self-administering MDPV or cocaine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.04.583431. [PMID: 38496609 PMCID: PMC10942381 DOI: 10.1101/2024.03.04.583431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Substance use disorder (SUD) is a heterogeneous disorder, where severity, symptoms, and patterns of substance use vary across individuals. Yet, when rats are allowed to self-administer drugs such as cocaine under short-access conditions, their behavior tends to be well-regulated and homogeneous in nature; though individual differences can emerge when rats are provided long- or intermittent-access to cocaine. In contrast to cocaine, significant individual differences emerge when rats are allowed to self-administer 3,4-methylenedioxypyrovalerone (MDPV), even under short-access conditions, wherein ~30% of rats rapidly transition to high levels of drug-taking. This study assessed the SUD-like phenotypes of male and female Sprague Dawley rats self-administering MDPV (0.032 mg/kg/infusion) or cocaine (0.32 mg/kg/infusion) by comparing level of drug intake, responding during periods of signaled drug unavailability, and sensitivity to footshock punishment to test the hypotheses that: (1) under short-access conditions, rats that self-administer MDPV will exhibit a more robust SUD-like phenotype than rats that self-administered cocaine; (2) female rats will have a more severe phenotype than male rats; and (3) compared to short-access, long- and intermittent-access to MDPV or cocaine self-administration will result in a more robust SUD-like phenotype. After short-access, rats that self-administered MDPV exhibited a more severe phenotype than rats that self-administered cocaine. Though long- and intermittent-access to cocaine and MDPV self-administration altered drug-taking patterns, manipulating access conditions did not systematically alter their SUD-like phenotype. Evidence from behavioral and quantitative autoradiography studies suggest that these differences are unlikely due to changes in expression levels of dopamine transporter, dopamine D2 or D3 receptors, or 5-HT1B, 5-HT2A, or 5-HT2C receptors, though these possibilities cannot be ruled out. These results show that the phenotype exhibited by rats self-administering MDPV differs from that observed for rats self-administering cocaine, and suggests that individuals that use MDPV and/or related cathinones may be at greater risk for developing a SUD, and that short-access MDPV self-administration may provide a useful method to understand the factors that mediate the transition to problematic or disordered substance use in humans.
Collapse
Affiliation(s)
- Michelle R. Doyle
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Nina M. Beltran
- Department of Psychology, University of Texas at El Paso, El Paso, TX, USA
| | - Mark S. A. Bushnell
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Maaz Syed
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Valeria Acosta
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Marisa Desai
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Kenner C. Rice
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism - Intramural Research Program, Bethesda, MD
| | | | - Georgianna G. Gould
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Lynette C. Daws
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Gregory T. Collins
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- South Texas Veterans Health Care System, San Antonio, TX, USA
| |
Collapse
|
16
|
Stark AJ, Song AK, Petersen KJ, Hay KR, Lin YC, Trujillo P, Kang H, Collazzo JM, Donahue MJ, Zald DH, Claassen DO. Accentuated Paralimbic and Reduced Mesolimbic D 2/3-Impulsivity Associations in Parkinson's Disease. J Neurosci 2023; 43:8733-8743. [PMID: 37852792 PMCID: PMC10727183 DOI: 10.1523/jneurosci.1037-23.2023] [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: 05/29/2023] [Revised: 07/31/2023] [Accepted: 10/10/2023] [Indexed: 10/20/2023] Open
Abstract
Impulsivity is a behavioral trait that is elevated in many neuropsychiatric disorders. Parkinson's disease (PD) patients can exhibit a specific pattern of reward-seeking impulsive-compulsive behaviors (ICBs), as well as more subtle changes to generalized trait impulsivity. Prior studies in healthy controls (HCs) suggest that trait impulsivity is regulated by D2/3 autoreceptors in mesocorticolimbic circuits. While altered D2/3 binding is noted in ICB+ PD patients, there is limited prior assessment of the trait impulsivity-D2/3 relationship in PD, and no prior direct comparison with patterns in HCs. We examined 54 PD (36 M; 18 F) and 31 sex- and age-matched HC (21 M; 10 F) subjects using [18F]fallypride, a high-affinity D2/3 receptor ligand, to measure striatal and extrastriatal D2/3 nondisplaceable binding potential (BPND). Subcortical and cortical assessment exclusively used ROI or exploratory-voxelwise methods, respectively. All completed the Barratt Impulsiveness Scale, a measure of trait impulsivity. Subcortical ROI analyses indicated a negative relationship between trait impulsivity and D2/3 BPND in the ventral striatum and amygdala of HCs but not in PD. By contrast, voxelwise methods demonstrated a positive trait impulsivity-D2/3 BPND correlation in ventral frontal olfactocentric-paralimbic cortex of subjects with PD but not HCs. Subscale analysis also highlighted different aspects of impulsivity, with significant interactions between group and motor impulsivity in the ventral striatum, and attentional impulsivity in the amygdala and frontal paralimbic cortex. These results suggest that dopamine functioning in distinct regions of the mesocorticolimbic circuit influence aspects of impulsivity, with the relative importance of regional dopamine functions shifting in the neuropharmacological context of PD.SIGNIFICANCE STATEMENT The biological determinants of impulsivity have broad clinical relevance, from addiction to neurodegenerative disorders. Here, we address biomolecular distinctions in Parkinson's disease. This is the first study to evaluate a large cohort of Parkinson's disease patients and age-matched healthy controls with a measure of trait impulsivity and concurrent [18F]fallypride PET, a method that allows quantification of D2/3 receptors throughout the mesocorticolimbic network. We demonstrate widespread differences in the trait impulsivity-dopamine relationship, including (1) loss of subcortical relationships present in the healthy brain and (2) emergence of a new relationship in a limbic cortical area. This illustrates the loss of mechanisms of behavioral regulation present in the healthy brain while suggesting a potential compensatory response and target for future investigation.
Collapse
Affiliation(s)
- Adam J Stark
- School of Medicine, Vanderbilt University, Nashville, Tennessee 37232
| | - Alexander K Song
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Kalen J Petersen
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63310
| | - Kaitlyn R Hay
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Ya-Chen Lin
- Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Paula Trujillo
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Hakmook Kang
- Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Jenna M Collazzo
- School of Medicine, Temple University, Philadelphia, Pennsylvania 19140
| | - Manus J Donahue
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - David H Zald
- Department of Psychiatry, Rutgers University, Piscataway, New Jersey 08901
| | - Daniel O Claassen
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| |
Collapse
|
17
|
Grunze H. The role of the D3 dopamine receptor and its partial agonist cariprazine in patients with schizophrenia and substance use disorder. Expert Opin Pharmacother 2023; 24:1985-1992. [PMID: 37817489 DOI: 10.1080/14656566.2023.2266359] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 09/29/2023] [Indexed: 10/12/2023]
Abstract
INTRODUCTION Comorbidity of substance use disorder (SUD) with schizophrenia, referred to as dual disorder (DD), significantly increases morbidity and mortality compared to schizophrenia alone. A dopaminergic dysregulation seems to be a common pathophysiological basis of the comorbidity. AREAS COVERED This article reports the current evidence on the role of dopamine dysregulations in DD, the pharmacological profile of cariprazine, a partial agonist of D3 and D2 dopamine receptors, and first clinical observations that may support its usefulness in the therapy of DD. PubMed/MEDLINE was searched for the keywords 'cariprazine,' 'schizophrenia,' 'dual disorder,' 'dopamine,' and 'dopamine receptor.' Preclinical and clinical studies, and reviews published in English were retrieved. EXPERT OPINION Although the management of DD remains challenging, and the evidence for pharmacologic treatments is still unsatisfactory, cariprazine may be a candidate medication in DD due to its unique mechanism of action. Preliminary clinical experiences suggest that cariprazine has both antipsychotic and anticraving properties and should be considered early in patients with DD.
Collapse
Affiliation(s)
- Heinz Grunze
- Psychiatrie Schwäbisch Hall, Schwäbisch Hall, Germany
- Department of Psychiatry, Paracelsus Medical University Nuremberg, Nuremberg, Germany
| |
Collapse
|
18
|
Turan Ç, Şenormancı G, Neşelioğlu S, Budak Y, Erel Ö, Şenormancı Ö. Oxidative Stress and Inflammatory Biomarkers in People with Methamphetamine Use Disorder. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2023; 21:572-582. [PMID: 37424424 PMCID: PMC10335902 DOI: 10.9758/cpn.22.1047] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/03/2023] [Accepted: 03/06/2023] [Indexed: 07/11/2023]
Abstract
Objective This study aimed to investigate the blood serum levels of biomarkers specifying oxidative stress status and systemic inflammation between people using methamphetamine (METH) and the control group (CG). Serum thiol/disulfide balance and ischemia-modified albumin levels were studied to determine oxidative stress, and serum interleukin-6 (IL-6) levels and complete blood count (CBC) were to assess inflammation. Methods Fifty patients with METH use disorder (MUD) and 36 CG participants were included in the study. Two tubes of venous blood samples were taken to measure oxidative stress, serum thiol/disulfide balance, ischemia-modified albumin, and IL-6 levels between groups. The correlation of parameters measuring oxidative stress and inflammation between groups with sociodemographic data was investigated. Results In this study, serum total thiol, free thiol levels, disulfide/native thiol percentage ratios, and serum ischemia- modified albumin levels of the patients were statistically significantly higher than the healthy controls. No difference was observed between the groups in serum disulfide levels and serum IL-6 levels. Considering the regression analysis, only the duration of substance use was a statistically significant factor in explaining serum IL-6 levels. The parameters showing inflammation in the CBC were significantly higher in the patients than in the CG. Conclusion CBC can be used to evaluate systemic inflammation in patients with MUD. Parameters measuring thiol/disulfide homeostasis and ischemia-modified albumin can be, also, used to assess oxidative stress.
Collapse
Affiliation(s)
- Çetin Turan
- Department of Psychiatry, University of Health Sciences Bursa Yuksek Ihtisas Training and Research Hospital, Bursa, Turkey
| | - Güliz Şenormancı
- Department of Psychiatry, University of Health Sciences Bursa Yuksek Ihtisas Training and Research Hospital, Bursa, Turkey
| | - Salim Neşelioğlu
- Clinic of Clinical Biochemistry, Yıldırım Beyazıt University, Ankara City Hospital, Ankara, Turkey
| | - Yasemin Budak
- Department of Biochemistry, University of Health Sciences Bursa Yuksek Ihtisas Training and Research Hospital, Bursa, Turkey
| | - Özcan Erel
- Clinic of Clinical Biochemistry, Yıldırım Beyazıt University, Ankara City Hospital, Ankara, Turkey
| | - Ömer Şenormancı
- Department of Clinical Psychology, University of Beykent, Istanbul, Turkey
| |
Collapse
|
19
|
Towers EB, Williams IL, Qillawala EI, Lynch WJ. Role of nucleus accumbens dopamine 2 receptors in motivating cocaine use in male and female rats prior to and following the development of an addiction-like phenotype. Front Pharmacol 2023; 14:1237990. [PMID: 37564182 PMCID: PMC10411909 DOI: 10.3389/fphar.2023.1237990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 07/19/2023] [Indexed: 08/12/2023] Open
Abstract
A hallmark of cocaine use disorder (CUD) is dysfunction of dopamine signaling in the mesolimbic pathway, including impaired dopamine 2 (D2) receptor signaling. One of the most replicated findings in human imagining studies is decreased striatal D2 receptor binding in individuals with a substance use disorder relative to healthy controls; however, the vast majority of the data is from males, and findings in smokers suggest this molecular shift may not translate to females. The goal of this study was to determine whether there are sex differences in the role of D2 receptors in motivating cocaine use prior to and following the development of an addiction-like phenotype (defined by an enhanced motivation for cocaine relative to the short-access, ShA, group). Here, male and female rats were given ShA (20 infusions/day, 3 days) or extended-access (ExA; 24h/day, 96 infusions/day, 10 days) to cocaine self-administration and then following 14 days of withdrawal, were tested under a progressive-ratio schedule to assess motivation for cocaine use. Once a stable level of motivation was established, the effect of NAc-infusions of the D2 receptor antagonist eticlopride (0-3.0 µg/side) were examined. We found that in males, eticlopride was less effective at decreasing motivation for cocaine following ExA versus ShA self-administration, particularly at low eticlopride doses. In contrast, in females, there were no differences in the effectiveness of eticlopride between ExA and ShA. These findings indicate that males, but not females, become less sensitive to NAc-D2 receptor antagonism with the development of an addiction-like phenotype.
Collapse
Affiliation(s)
- Eleanor Blair Towers
- Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA, United States
- Medical Scientist Training Program, University of Virginia, Charlottesville, VA, United States
| | - Ivy L. Williams
- Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA, United States
| | - Emaan I. Qillawala
- Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA, United States
| | - Wendy J. Lynch
- Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
20
|
Hersey M, Bartole MK, Jones CS, Newman AH, Tanda G. Are There Prevalent Sex Differences in Psychostimulant Use Disorder? A Focus on the Potential Therapeutic Efficacy of Atypical Dopamine Uptake Inhibitors. Molecules 2023; 28:5270. [PMID: 37446929 PMCID: PMC10343811 DOI: 10.3390/molecules28135270] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Psychostimulant use disorders (PSUD) affect a growing number of men and women and exert sizable public health and economic burdens on our global society. Notably, there are some sex differences in the onset of dependence, relapse rates, and treatment success with PSUD observed in preclinical and clinical studies. The subtle sex differences observed in the behavioral aspects of PSUD may be associated with differences in the neurochemistry of the dopaminergic system between sexes. Preclinically, psychostimulants have been shown to increase synaptic dopamine (DA) levels and may downregulate the dopamine transporter (DAT). This effect is greatest in females during the high estradiol phase of the estrous cycle. Interestingly, women have been shown to be more likely to begin drug use at younger ages and report higher levels of desire to use cocaine than males. Even though there is currently no FDA-approved medication, modafinil, a DAT inhibitor approved for use in the treatment of narcolepsy and sleep disorders, has shown promise in the treatment of PSUD among specific populations of affected individuals. In this review, we highlight the therapeutic potential of modafinil and other atypical DAT inhibitors focusing on the lack of sex differences in the actions of these agents.
Collapse
Affiliation(s)
| | | | | | | | - Gianluigi Tanda
- Medication Development Program, NIDA IRP, Baltimore, MD 21224, USA; (M.H.); (M.K.B.); (C.S.J.); (A.H.N.)
| |
Collapse
|
21
|
Zeng J, You L, Sheng H, Luo Y, Yang X. The differential neural substrates for reward choice under gain-loss contexts and risk in alcohol use disorder: Evidence from a voxel-based meta-analysis. Drug Alcohol Depend 2023; 248:109912. [PMID: 37182355 DOI: 10.1016/j.drugalcdep.2023.109912] [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: 01/17/2023] [Revised: 04/15/2023] [Accepted: 04/30/2023] [Indexed: 05/16/2023]
Abstract
BACKGROUND Making a risky decision is a complex process that involves the evaluation of both the values of the options and the associated risk level; this process is distinct from reward processing in gain versus loss contexts. Although disrupted reward processing in mesolimbic dopamine circuitry is suggested to underlie pathological incentive processing in patients with alcohol use disorder (AUD), the differential neural processes subserving these motivational tendencies for risk situations or gain/loss choices in decision-making have not been identified. METHODS To examine the common or distinct neural mechanisms in the evaluation of risk versus outcomes for AUD, we conducted two separate coordinate-based meta-analyses of functional neuroimaging studies by using Seed-Based d Mapping software to evaluate 13 studies investigating gain and loss processing and 10 studies investigating risky decision-making. RESULTS During gain and loss processing, relative to healthy controls, AUD patients showed reduced activation in the mesocortical-limbic circuit, including the orbital prefrontal cortex (OFC), dorsal striatum, insula, hippocampus, cerebellum, cuneus cortex and superior temporal gyrus, but hyperactivation in the inferior temporal gyrus and paracentral lobule (extending to the middle cingulate cortex (MCC) and precuneus). During decision-making under risk, AUD patients exhibited hypoactivity of the prefrontal and cingulate cortices, including the posterior cingulate cortex (extending to the MCC), middle frontal gyrus, medial prefrontal cortex, dorsolateral prefrontal cortex, OFC and anterior cingulate cortex. CONCLUSIONS Our results extend existing neurological evidence by showing that a reduced response in the mesocortical-limbic circuit is found in gain versus loss processing, with decreased responsivity in cortical regions in risk decision-making. Our results implicate dissociable neural circuit responses for gain-loss processing and risk decision-making, which contribute to a better understanding of the pathophysiological mechanism underlying nondrug incentive and risk processing in individuals with AUD.
Collapse
Affiliation(s)
- Jianguang Zeng
- School of Economics and Business Administration, Chongqing University, Chongqing, China
| | - Lantao You
- School of Economics and Business Administration, Chongqing University, Chongqing, China
| | - Haoxuan Sheng
- School of Public Policy and Administration, Chongqing University, Chongqing, China
| | - Ya Luo
- Department of Psychiatry, State Key Lab of Biotherapy, West China Hospital of Sichuan University, Chengdu, China
| | - Xun Yang
- School of Public Policy and Administration, Chongqing University, Chongqing, China.
| |
Collapse
|
22
|
Zanda MT, Floris G, Daws SE. Orbitofrontal cortex microRNAs support long-lasting heroin seeking behavior in male rats. Transl Psychiatry 2023; 13:117. [PMID: 37031193 PMCID: PMC10082780 DOI: 10.1038/s41398-023-02423-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 03/23/2023] [Accepted: 03/29/2023] [Indexed: 04/10/2023] Open
Abstract
Recovery from opioid use disorder (OUD) and maintenance of abstinence from opioid use is hampered by perseverant drug cravings that may persist for months after cessation of drug use. Drug cravings can intensify during the abstinence period, a phenomenon referred to as the 'incubation of craving' that has been well-described in preclinical studies. We previously reported that animals that self-administered heroin at a dosage of 0.075 mg/kg/infusion (HH) paired with discrete drug cues displayed robust incubation of heroin craving behavior after 21 days (D) of forced abstinence, an effect that was not observed with a lower dosage (0.03 mg/kg/infusion; HL). Here, we sought to elucidate molecular mechanisms underlying long-term heroin seeking behavior by profiling microRNA (miRNA) pathways in the orbitofrontal cortex (OFC), a brain region that modulates incubation of heroin seeking. miRNAs are small noncoding RNAs with long half-lives that have emerged as critical regulators of drug seeking behavior but their expression in the OFC has not been examined in any drug exposure paradigm. We employed next generation sequencing to detect OFC miRNAs differentially expressed after 21D of forced abstinence between HH and HL animals, and proteomics analysis to elucidate miRNA-dependent translational neuroadaptations. We identified 55 OFC miRNAs associated with incubation of heroin craving, including miR-485-5p, which was significantly downregulated following 21D forced abstinence in HH but not HL animals. We bidirectionally manipulated miR-485-5p in the OFC to demonstrate that miR-485-5p can regulate long-lasting heroin seeking behavior after extended forced abstinence. Proteomics analysis identified 45 proteins selectively regulated in the OFC of HH but not HL animals that underwent 21D forced abstinence, of which 7 were putative miR-485-5p target genes. Thus, the miR-485-5p pathway is dysregulated in animals with a phenotype of persistent heroin craving behavior and OFC miR-485-5p pathways may function to support long-lasting heroin seeking.
Collapse
Affiliation(s)
- Mary Tresa Zanda
- Center for Substance Abuse Research, Temple University, Philadelphia, PA, USA
- Department of Neural Sciences, Temple University, Philadelphia, PA, USA
| | - Gabriele Floris
- Center for Substance Abuse Research, Temple University, Philadelphia, PA, USA
- Department of Neural Sciences, Temple University, Philadelphia, PA, USA
| | - Stephanie E Daws
- Center for Substance Abuse Research, Temple University, Philadelphia, PA, USA.
- Department of Neural Sciences, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
23
|
Ribeiro G, Maia A, Cotovio G, Oliveira FPM, Costa DC, Oliveira-Maia AJ. Striatal dopamine D2-like receptors availability in obesity and its modulation by bariatric surgery: a systematic review and meta-analysis. Sci Rep 2023; 13:4959. [PMID: 36973321 PMCID: PMC10042861 DOI: 10.1038/s41598-023-31250-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 03/08/2023] [Indexed: 03/29/2023] Open
Abstract
There is significant evidence linking a 'reward deficiency syndrome' (RDS), comprising decreased availability of striatal dopamine D2-like receptors (DD2lR) and addiction-like behaviors underlying substance use disorders and obesity. Regarding obesity, a systematic review of the literature with a meta-analysis of such data is lacking. Following a systematic review of the literature, we performed random-effects meta-analyses to determine group differences in case-control studies comparing DD2lR between individuals with obesity and non-obese controls and prospective studies of pre- to post-bariatric surgery DD2lR changes. Cohen's d was used to measure effect size. Additionally, we explored factors potentially associated with group differences in DD2lR availability, such as obesity severity, using univariate meta-regression. In a meta-analysis including positron emission tomography (PET) and single-photon emission computed tomography (SPECT) studies, striatal DD2lR availability did not significantly differ between obesity and controls. However, in studies comprising patients with class III obesity or higher, group differences were significant, favoring lower DD2lR availability in the obesity group. This effect of obesity severity was corroborated by meta-regressions showing inverse associations between the body mass index (BMI) of the obesity group and DD2lR availability. Post-bariatric changes in DD2lR availability were not found, although a limited number of studies were included in this meta-analysis. These results support lower DD2lR in higher classes of obesity which is a more targeted population to explore unanswered questions regarding the RDS.
Collapse
Affiliation(s)
- Gabriela Ribeiro
- Champalimaud Research and Clinical Centre, Champalimaud Foundation, Av. de Brasília, Doca de Pedrouços, 1400-038, Lisboa, Portugal
- Lisbon Academic Medical Centre PhD Program, Faculdade de Medicina da Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
- Nova Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, 1169-056, Lisboa, Portugal
- Nutrition and Metabolism Department, Nova Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, 1169-056, Lisboa, Portugal
| | - Ana Maia
- Champalimaud Research and Clinical Centre, Champalimaud Foundation, Av. de Brasília, Doca de Pedrouços, 1400-038, Lisboa, Portugal
- Nova Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, 1169-056, Lisboa, Portugal
- Department of Psychiatry and Mental Health, Centro Hospitalar de Lisboa Ocidental, Rua da Junqueira, 126, 1340-019, Lisboa, Portugal
| | - Gonçalo Cotovio
- Champalimaud Research and Clinical Centre, Champalimaud Foundation, Av. de Brasília, Doca de Pedrouços, 1400-038, Lisboa, Portugal
- Nova Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, 1169-056, Lisboa, Portugal
- Department of Psychiatry and Mental Health, Centro Hospitalar de Lisboa Ocidental, Rua da Junqueira, 126, 1340-019, Lisboa, Portugal
| | - Francisco P M Oliveira
- Champalimaud Research and Clinical Centre, Champalimaud Foundation, Av. de Brasília, Doca de Pedrouços, 1400-038, Lisboa, Portugal
| | - Durval C Costa
- Champalimaud Research and Clinical Centre, Champalimaud Foundation, Av. de Brasília, Doca de Pedrouços, 1400-038, Lisboa, Portugal
| | - Albino J Oliveira-Maia
- Champalimaud Research and Clinical Centre, Champalimaud Foundation, Av. de Brasília, Doca de Pedrouços, 1400-038, Lisboa, Portugal.
- Nova Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, 1169-056, Lisboa, Portugal.
| |
Collapse
|
24
|
Beck A, Ebrahimi C, Rosenthal A, Charlet K, Heinz A. The Dopamine System in Mediating Alcohol Effects in Humans. Curr Top Behav Neurosci 2023. [PMID: 36705911 DOI: 10.1007/7854_2022_415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Brain-imaging studies show that the development and maintenance of alcohol use disorder (AUD) is determined by a complex interaction of different neurotransmitter systems and multiple psychological factors. In this context, the dopaminergic reinforcement system appears to be of fundamental importance. We focus on the excitatory and depressant effects of acute versus chronic alcohol intake and its impact on dopaminergic neurotransmission. Furthermore, we describe alterations in dopaminergic neurotransmission as associated with symptoms of alcohol dependence. We specifically focus on neuroadaptations to chronic alcohol consumption and their effect on central processing of alcohol-associated and reward-related stimuli. Altered reward processing, complex conditioning processes, impaired reinforcement learning, and increased salience attribution to alcohol-associated stimuli enable alcohol cues to drive alcohol seeking and consumption. Finally, we will discuss how the neurobiological and neurochemical mechanisms of alcohol-associated alterations in reward processing and learning can interact with stress, cognition, and emotion processing.
Collapse
Affiliation(s)
- Anne Beck
- Faculty of Health, Health and Medical University, Potsdam, Germany
| | - Claudia Ebrahimi
- Department of Psychiatry and Neurosciences, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt- Universität zu Berlin, Berlin, Germany
| | - Annika Rosenthal
- Department of Psychiatry and Neurosciences, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt- Universität zu Berlin, Berlin, Germany
| | - Katrin Charlet
- Department of Psychiatry and Neurosciences, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt- Universität zu Berlin, Berlin, Germany
| | - Andreas Heinz
- Department of Psychiatry and Neurosciences, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt- Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
25
|
Li Y, Cheng P, Liang L, Dong H, Liu H, Shen W, Zhou W. Abnormal resting-state functional connectome in methamphetamine-dependent patients and its application in machine-learning-based classification. Front Neurosci 2022; 16:1014539. [DOI: 10.3389/fnins.2022.1014539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/04/2022] [Indexed: 11/18/2022] Open
Abstract
Brain resting-state functional connectivity (rsFC) has been widely analyzed in substance use disorders (SUDs), including methamphetamine (MA) dependence. Most of these studies utilized Pearson correlation analysis to assess rsFC, which cannot determine whether two brain regions are connected by direct or indirect pathways. Moreover, few studies have reported the application of rsFC-based graph theory in MA dependence. We evaluated alterations in Tikhonov regularization-based rsFC and rsFC-based topological attributes in 46 MA-dependent patients, as well as the correlations between topological attributes and clinical variables. Moreover, the topological attributes selected by least absolute shrinkage and selection operator (LASSO) were used to construct a support vector machine (SVM)-based classifier for MA dependence. The MA group presented a subnetwork with increased rsFC, indicating overactivation of the reward circuit that makes patients very sensitive to drug-related visual cues, and a subnetwork with decreased rsFC suggesting aberrant synchronized spontaneous activity in subregions within the orbitofrontal cortex (OFC) system. The MA group demonstrated a significantly decreased area under the curve (AUC) for the clustering coefficient (Cp) (Pperm < 0.001), shortest path length (Lp) (Pperm = 0.007), modularity (Pperm = 0.006), and small-worldness (σ, Pperm = 0.004), as well as an increased AUC for global efficiency (E.glob) (Pperm = 0.009), network strength (Sp) (Pperm = 0.009), and small-worldness (ω, Pperm < 0.001), implying a shift toward random networks. MA-related increased nodal efficiency (E.nodal) and altered betweenness centrality were also discovered in several brain regions. The AUC for ω was significantly positively associated with psychiatric symptoms. An SVM classifier trained by 36 features selected by LASSO from all topological attributes achieved excellent performance, cross-validated prediction area under the receiver operating characteristics curve, accuracy, sensitivity, specificity, and kappa of 99.03 ± 1.79, 94.00 ± 5.78, 93.46 ± 8.82, 94.52 ± 8.11, and 87.99 ± 11.57%, respectively (Pperm < 0.001), indicating that rsFC-based topological attributes can provide promising features for constructing a high-efficacy classifier for MA dependence.
Collapse
|
26
|
Dai KZ, Choi IB, Levitt R, Blegen MB, Kaplan AR, Matsui A, Shin JH, Bocarsly ME, Simpson EH, Kellendonk C, Alvarez VA, Dobbs LK. Dopamine D2 receptors bidirectionally regulate striatal enkephalin expression: Implications for cocaine reward. Cell Rep 2022; 40:111440. [PMID: 36170833 PMCID: PMC9620395 DOI: 10.1016/j.celrep.2022.111440] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 08/04/2022] [Accepted: 09/09/2022] [Indexed: 11/24/2022] Open
Abstract
Low dopamine D2 receptor (D2R) availability in the striatum can predispose for cocaine abuse; though how low striatal D2Rs facilitate cocaine reward is unclear. Overexpression of D2Rs in striatal neurons or activation of D2Rs by acute cocaine suppresses striatal Penk mRNA. Conversely, low D2Rs in D2-striatal neurons increases striatal Penk mRNA and enkephalin peptide tone, an endogenous mu-opioid agonist. In brain slices, met-enkephalin and inhibition of enkephalin catabolism suppresses intra-striatal GABA transmission. Pairing cocaine with intra-accumbens met-enkephalin during place conditioning facilitates acquisition of preference, while mu-opioid receptor antagonist blocks preference in wild-type mice. We propose that heightened striatal enkephalin potentiates cocaine reward by suppressing intra-striatal GABA to enhance striatal output. Surprisingly, a mu-opioid receptor antagonist does not block cocaine preference in mice with low striatal D2Rs, implicating other opioid receptors. The bidirectional regulation of enkephalin by D2R activity and cocaine offers insights into mechanisms underlying the vulnerability for cocaine abuse. Low striatal D2 receptor levels are associated with cocaine abuse. Dai et al. bidirectionally alter striatal D2 receptor levels to probe the downstream mechanisms underlying this abuse liability. They provide evidence that enhanced enkephalin tone resulting from low D2 receptors is associated with suppressed intra-striatal GABA and potentiated cocaine reward.
Collapse
Affiliation(s)
- Kathy Z Dai
- Laboratory on Neurobiology of Compulsive Behaviors, NIAAA, IRP, NIH, Bethesda, MD, USA
| | - In Bae Choi
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Ryan Levitt
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Mariah B Blegen
- Laboratory on Neurobiology of Compulsive Behaviors, NIAAA, IRP, NIH, Bethesda, MD, USA
| | - Alanna R Kaplan
- Laboratory on Neurobiology of Compulsive Behaviors, NIAAA, IRP, NIH, Bethesda, MD, USA
| | - Aya Matsui
- Laboratory on Neurobiology of Compulsive Behaviors, NIAAA, IRP, NIH, Bethesda, MD, USA
| | - J Hoon Shin
- Laboratory on Neurobiology of Compulsive Behaviors, NIAAA, IRP, NIH, Bethesda, MD, USA
| | - Miriam E Bocarsly
- Laboratory on Neurobiology of Compulsive Behaviors, NIAAA, IRP, NIH, Bethesda, MD, USA; Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Rutgers Brain Health Institute, Newark, NJ, USA
| | - Eleanor H Simpson
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA; Division of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, USA
| | - Christoph Kellendonk
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA; Department of Molecular Pharmacology and Therapeutics, Columbia University Medical Center, New York, NY, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Veronica A Alvarez
- Laboratory on Neurobiology of Compulsive Behaviors, NIAAA, IRP, NIH, Bethesda, MD, USA; Center on Compulsive Behaviors, IRP, NIH, Bethesda, MD, USA
| | - Lauren K Dobbs
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX, USA; Department of Neuroscience, Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
27
|
Biochemical Neuroadaptations in the Rat Striatal Dopaminergic System after Prolonged Exposure to Methamphetamine Self-Administration. Int J Mol Sci 2022; 23:ijms231710092. [PMID: 36077488 PMCID: PMC9456063 DOI: 10.3390/ijms231710092] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/29/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
Perturbations in striatal dopamine (DA) homeostasis might underlie the behavioral and pathobiological consequences of METH use disorder in humans. To identify potential consequences of long-term METH exposure, we modeled the adverse consequence DSM criterion of substance use disorders by giving footshocks to rats that had escalated their intake of METH during a drug self-administration procedure. Next, DA D1 receptor antagonist, SCH23390 was injected. Thereafter, rats were euthanized to measure several indices of the striatal dopaminergic system. Footshocks split the METH rats into two phenotypes: (i) shock-sensitive that decreased their METH-intake and (ii) shock-resistant that continued their METH intake. SCH23390 caused substantial dose-dependent reduction of METH taking in both groups. Stopping SCH23390 caused re-emergence of compulsive METH taking in shock-resistant rats. Compulsive METH takers also exhibited greater incubation of METH seeking than non-compulsive rats during withdrawal from METH SA. Analyses of DA metabolism revealed non-significant decreases (about 35%) in DA levels in resistant and sensitive rats. However, striatal contents of the deaminated metabolites, DOPAL and DOPAC, were significantly increased in sensitive rats. VMAT2 and DAT protein levels were decreased in both phenotypes. Moreover, protein expression levels of the D1-like DA receptor, D5R, and D2-like DA receptors, D3R and D4R, were significantly decreased in the compulsive METH takers. Our results parallel findings in post-mortem striatal tissues of human METH users who develop Parkinsonism after long-term METH intake and support the use of this model to investigate potential therapeutic interventions for METH use disorder.
Collapse
|
28
|
Armenta-Resendiz M, Assali A, Tsvetkov E, Cowan CW, Lavin A. Repeated methamphetamine administration produces cognitive deficits through augmentation of GABAergic synaptic transmission in the prefrontal cortex. Neuropsychopharmacology 2022; 47:1816-1825. [PMID: 35788684 PMCID: PMC9372065 DOI: 10.1038/s41386-022-01371-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/14/2022] [Accepted: 06/22/2022] [Indexed: 11/08/2022]
Abstract
Methamphetamine (METH) abuse is associated with the emergence of cognitive deficits and hypofrontality, a pathophysiological marker of many neuropsychiatric disorders that is produced by altered balance of local excitatory and inhibitory synaptic transmission. However, there is a dearth of information regarding the cellular and synaptic mechanisms underlying METH-induced cognitive deficits and associated hypofrontal states. Using PV-Cre transgenic rats that went through a METH sensitization regime or saline (SAL) followed by 7-10 days of home cage abstinence combined with cognitive tests, chemogenetic experiments, and whole-cell patch recordings on the prelimbic prefrontal cortex (PFC), we investigated the cellular and synaptic mechanisms underlying METH-induce hypofrontality. We report here that repeated METH administration in rats produces deficits in working memory and increases in inhibitory synaptic transmission onto pyramidal neurons in the PFC. The increased PFC inhibition is detected by an increase in spontaneous and evoked inhibitory postsynaptic synaptic currents (IPSCs), an increase in GABAergic presynaptic function, and a shift in the excitatory-inhibitory balance onto PFC deep-layer pyramidal neurons. We find that pharmacological blockade of D1 dopamine receptor function reduces the METH-induced augmentation of IPSCs, suggesting a critical role for D1 dopamine signaling in METH-induced hypofrontality. In addition, repeated METH administration increases the intrinsic excitability of parvalbumin-positive fast spiking interneurons (PV + FSIs), a key local interneuron population in PFC that contributes to the control of inhibitory tone. Using a cell type-specific chemogenetic approach, we show that increasing PV + FSIs activity in the PFC is necessary and sufficient to cause deficits in temporal order memory similar to those induced by METH. Conversely, reducing PV + FSIs activity in the PFC of METH-exposed rats rescues METH-induced temporal order memory deficits. Together, our findings reveal that repeated METH exposure increases PFC inhibitory tone through a D1 dopamine signaling-dependent potentiation of inhibitory synaptic transmission, and that reduction of PV + FSIs activity can rescue METH-induced cognitive deficits, suggesting a potential therapeutic approach to treating cognitive symptoms in patients suffering from METH use disorder.
Collapse
Affiliation(s)
| | - Ahlem Assali
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Evgeny Tsvetkov
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Christopher W Cowan
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Antonieta Lavin
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
29
|
Highgate Q, Abadey AA, Schenk S. Repeated eticlopride administration increases dopamine D 2 receptor expression and restores behavioral flexibility disrupted by methamphetamine exposure to male rats. Behav Brain Res 2022; 435:114064. [PMID: 35987306 DOI: 10.1016/j.bbr.2022.114064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 08/16/2022] [Accepted: 08/16/2022] [Indexed: 11/15/2022]
Abstract
Repeated methamphetamine exposure impairs reversal learning in laboratory animals and downregulates dopamine D2 receptor expression. In the present study, we tested the possibility that repeated exposure to the dopamine D2 antagonist, eticlopride, would increase D2 receptor expression, improve behavioral flexibility and restore behavioral flexibility that was disrupted by exposure to methamphetamine in rats. Male Sprague-Dawley rats received repeated daily pretreatment with the dopamine D2 antagonist, eticlopride (0.0 or 0.3 mg/kg/day, 14 days). Three days after the last treatment, whole brain (minus olfactory bulbs and cerebellum) dopamine D2 receptor expression was measured using flow cytometry in one group and reversal learning performance was measured in another group. Reversal learning was also measured in other groups prior to and after methamphetamine exposure (0.0 or 2.0 mg/kg, 4 injections, 2 h apart, 1 day) followed by repeated eticlopride (0.0 or 0.3 mg/kg, 14 days) treatment. Eticlopride treatment increased D2 receptor expression and improved reversal learning performance. Methamphetamine impaired reversal learning performance and eticlopride treatment reversed the deficit. These results suggest that repeated administration of eticlopride can restore behavioral flexibility and that upregulation of D2 receptors might be an effective adjunct to treatment of methamphetamine misuse.
Collapse
Affiliation(s)
- Quenten Highgate
- School of Psychology, Victoria University of Wellington, Wellington, New Zealand
| | - Afnan Al Abadey
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Susan Schenk
- School of Psychology, Victoria University of Wellington, Wellington, New Zealand; Department of Zoology, University of Otago, Box 56, Dunedin 9054, New Zealand.
| |
Collapse
|
30
|
Kozarzewski L, Maurer L, Mähler A, Spranger J, Weygandt M. Computational approaches to predicting treatment response to obesity using neuroimaging. Rev Endocr Metab Disord 2022; 23:773-805. [PMID: 34951003 PMCID: PMC9307532 DOI: 10.1007/s11154-021-09701-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/02/2021] [Indexed: 12/11/2022]
Abstract
Obesity is a worldwide disease associated with multiple severe adverse consequences and comorbid conditions. While an increased body weight is the defining feature in obesity, etiologies, clinical phenotypes and treatment responses vary between patients. These variations can be observed within individual treatment options which comprise lifestyle interventions, pharmacological treatment, and bariatric surgery. Bariatric surgery can be regarded as the most effective treatment method. However, long-term weight regain is comparably frequent even for this treatment and its application is not without risk. A prognostic tool that would help predict the effectivity of the individual treatment methods in the long term would be essential in a personalized medicine approach. In line with this objective, an increasing number of studies have combined neuroimaging and computational modeling to predict treatment outcome in obesity. In our review, we begin by outlining the central nervous mechanisms measured with neuroimaging in these studies. The mechanisms are primarily related to reward-processing and include "incentive salience" and psychobehavioral control. We then present the diverse neuroimaging methods and computational prediction techniques applied. The studies included in this review provide consistent support for the importance of incentive salience and psychobehavioral control for treatment outcome in obesity. Nevertheless, further studies comprising larger sample sizes and rigorous validation processes are necessary to answer the question of whether or not the approach is sufficiently accurate for clinical real-world application.
Collapse
Affiliation(s)
- Leonard Kozarzewski
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Clinic of Endocrinology, Diabetes and Metabolism, 10117, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité Center for Cardiovascular Research, 10117, Berlin, Germany
| | - Lukas Maurer
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Clinic of Endocrinology, Diabetes and Metabolism, 10117, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité Center for Cardiovascular Research, 10117, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Anja Mähler
- Max Delbrück Center for Molecular Medicine and Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center (ECRC), 13125, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, NeuroCure Clinical Research Center, 10117, Berlin, Germany
| | - Joachim Spranger
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Clinic of Endocrinology, Diabetes and Metabolism, 10117, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité Center for Cardiovascular Research, 10117, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 10117, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Martin Weygandt
- Max Delbrück Center for Molecular Medicine and Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center (ECRC), 13125, Berlin, Germany.
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, NeuroCure Clinical Research Center, 10117, Berlin, Germany.
| |
Collapse
|
31
|
Caffeine-Induced Acute and Delayed Responses in Cerebral Metabolism of Control and Schizophrenia-Like Wisket Rats. Int J Mol Sci 2022; 23:ijms23158186. [PMID: 35897774 PMCID: PMC9331118 DOI: 10.3390/ijms23158186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/21/2022] [Accepted: 07/23/2022] [Indexed: 12/04/2022] Open
Abstract
Recently, morphological impairments have been detected in the brain of a triple-hit rat schizophrenia model (Wisket), and delayed depressive effects of caffeine treatment in both control and Wisket animals have also been shown. The aims of this study were to determine the basal and caffeine-induced acute (30 min) and delayed (24 h) changes in the cerebral 18fluorodeoxyglucose (18F-FDG) uptake by positron emission tomography (PET) in control and Wisket rats. No significant differences were identified in the basal whole-brain metabolism between the two groups, and the metabolism was not modified acutely by a single intraperitoneal caffeine (20 mg/kg) injection in either group. However, one day after caffeine administration, significantly enhanced 18F-FDG uptake was detected in the whole brain and the investigated areas (hippocampus, striatum, thalamus, and hypothalamus) in the control group. Although the Wisket animals showed only moderate enhancements in the 18F-FDG uptake, significantly lower brain metabolism was observed in this group than in the caffeine-treated control group. This study highlights that the basal brain metabolism of Wisket animals was similar to control rats, and that was not influenced acutely by single caffeine treatment at the whole-brain level. Nevertheless, the distinct delayed responsiveness to this psychostimulant in Wisket model rats suggests impaired control of the cerebral metabolism.
Collapse
|
32
|
Becker-Krail DD, Walker WH, Nelson RJ. The Ventral Tegmental Area and Nucleus Accumbens as Circadian Oscillators: Implications for Drug Abuse and Substance Use Disorders. Front Physiol 2022; 13:886704. [PMID: 35574492 PMCID: PMC9094703 DOI: 10.3389/fphys.2022.886704] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/04/2022] [Indexed: 12/15/2022] Open
Abstract
Circadian rhythms convergently evolved to allow for optimal synchronization of individuals’ physiological and behavioral processes with the Earth’s 24-h periodic cycling of environmental light and temperature. Whereas the suprachiasmatic nucleus (SCN) is considered the primary pacemaker of the mammalian circadian system, many extra-SCN oscillatory brain regions have been identified to not only exhibit sustainable rhythms in circadian molecular clock function, but also rhythms in overall region activity/function and mediated behaviors. In this review, we present the most recent evidence for the ventral tegmental area (VTA) and nucleus accumbens (NAc) to serve as extra-SCN oscillators and highlight studies that illustrate the functional significance of the VTA’s and NAc’s inherent circadian properties as they relate to reward-processing, drug abuse, and vulnerability to develop substance use disorders (SUDs).
Collapse
Affiliation(s)
- Darius D Becker-Krail
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - William H Walker
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - Randy J Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
33
|
van der Eijk Y, Chen JIP. Case for raising the minimum legal age of tobacco sale to 25. Tob Control 2022; 31:487-492. [PMID: 33414266 DOI: 10.1136/tobaccocontrol-2020-055964] [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: 05/28/2020] [Revised: 10/15/2020] [Accepted: 11/17/2020] [Indexed: 11/03/2022]
Abstract
Restricting youth access to tobacco is an essential component of a comprehensive tobacco control policy. While there has been a growing movement to raise the minimum legal age (MLA) of purchasing tobacco from 18 to 21, more restrictive measures, such as raising the MLA to 25 (MLA25), have been criticised as being overly restrictive on adult's free choice. We argue that, even within a policy approach that prioritises freedom of choice, there is a strong case for MLA25 in view of neurobiological evidence which shows that, before age 25, people are neurobiologically vulnerable to developing an addiction. We discuss further considerations for an MLA25 policy, in particular its potential impact on the free choice of young adults to start or quit smoking, potential public health impact and potential effectiveness considering that most underage youth source cigarettes from older peers.
Collapse
Affiliation(s)
- Yvette van der Eijk
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Jacinta I-Pei Chen
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| |
Collapse
|
34
|
Askari N, Mousavi A, Vaez-Mahdavi MR. Maternal deprivation effect on morphine-induced CPP is related to changes in Opioid receptors in selected rat brain regions (hippocampus, prefrontal cortex, and nucleus accumbens). Behav Processes 2022; 197:104607. [PMID: 35218881 DOI: 10.1016/j.beproc.2022.104607] [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: 09/19/2021] [Revised: 02/19/2022] [Accepted: 02/20/2022] [Indexed: 11/16/2022]
Abstract
Early-life environmental conditions affect offspring's development. Maternal deprivation (MD) can induce persistent changes that give rise to neuropsychiatric diseases including substance abuse disorders. However, long-lasting mechanisms that determine vulnerability to drug addiction remain unknown. We hypothesized that MD could induce changes in Opioid system, HPA (hypothalamic-pituitary-adrenal) axis, and BDNF (brain-derived neurotrophic factor), so may be involved in the drug abuse in later life. Male offspring of Wistar rats (n=8 per group) were subjected to 3h of daily MD during postnatal days 1-14. In adulthood, morphine-induced CPP (conditioned place preference) was investigated using two doses of morphine (3 and 5mg/kg). Serum corticosterone level was measured by ELISA method. The expression level of genes in selected brain regions (hippocampus, prefrontal cortex, and nucleus accumbens) was determined by qPCR (quantitative PCR). A greater morphine-induced CPP was observed in MD rats with 3 and 5mg/kg morphine compared to controls. MD group had a higher corticosterone level. A significant decrease was observed in the expression of BDNF gene (in all of the selected brain regions) and GR (glucocorticoid receptor) gene (in the hippocampus and nucleus accumbens) in MD rats. Also, a significant increase in the expression of μ Opioid receptor (in all of the selected brain regions) and κ Opioid receptor (in the prefrontal cortex and nucleus accumbens) was observed in MD rats. Our results suggest that MD induces alterations in the HPA axis function, BDNF level, and Opioid receptors system that enhance vulnerability to morphine at adulthood.
Collapse
Affiliation(s)
- Nayere Askari
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, I.R. Iran; Immunoregulation Research Center, Shahed University, Tehran, I.R. Iran.
| | - Ali Mousavi
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, I.R. Iran
| | | |
Collapse
|
35
|
Nie L, Wen X, Luo W, Ju T, Ren A, Wu B, Li J, Hu J. Disruption of regional homogeneity in the brains of chronic methamphetamine users. Brain Imaging Behav 2022; 16:1605-1613. [PMID: 35175550 DOI: 10.1007/s11682-022-00637-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2022] [Indexed: 02/08/2023]
Abstract
Previous studies have reported evidence supporting structural and functional alterations in the brains of methamphetamine (MA) users. The aim of the present study was to extend current knowledge regarding brain function(s) in MA users by examining regional homogeneity (ReHo). Chronic MA users (51 male, 46 female), who were undergoing supervised abstinence for 12 to 621 days, and 79 healthy controls (43 male, 36 female) underwent resting-state functional brain magnetic resonance imaging. Voxel-wise whole-brain scale group differences in ReHo were examined. The mean ReHo values of significant clusters were extracted, and linear regression was used to identify factors that contributed to these mean ReHo values. MA users exhibited lower ReHo values in the left orbital part of the inferior frontal gyrus extending to the left insula and left temporal pole, left amygdala, and left fusiform gyrus. MA users also exhibited greater ReHo values in the bilateral pre- and postcentral gyri and right cerebellum. Characteristics of MA use, including duration, duration of abstinence from MA, and age at onset of MA use, demonstrated no reliable contribution to ReHo of the significant clusters. Findings of the present study demonstrated that chronic MA use was associated with regional specific disruption of ReHo, which is relatively independent of structural and functional alterations and, apparently, does not recover after relatively long-term abstinence. This disruption may underlie overall neurocognitive deficits in MA users, which is difficult to recover.
Collapse
Affiliation(s)
- Lili Nie
- School of Psychology, Liaoning Normal University, Dalian, 116029, China
| | - Xiantao Wen
- Sichuan Provincial Compulsory Drug Addiction Treatment Agency for Males, Ziyang, China
| | - Wei Luo
- Sichuan Provincial Compulsory Drug Addiction Treatment Agency for Females, Deyang, China
| | - Tao Ju
- Hospital of Sichuan Provincial Compulsory Drug Addiction Treatment Agency for Females, Deyang, China
| | - Anlian Ren
- Sichuan Provincial Compulsory Drug Addiction Treatment Agency for Males, Ziyang, China
| | - Binbin Wu
- Hospital of Sichuan Provincial Compulsory Drug Addiction Treatment Agency for Females, Deyang, China
| | - Jing Li
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, 610041, China.
| | - Jinsheng Hu
- School of Psychology, Liaoning Normal University, Dalian, 116029, China.
| |
Collapse
|
36
|
Prenatal Amphetamine-Induced Dopaminergic Alteration in a Gender- and Estrogen-Dependent Manner. Neurochem Res 2022; 47:1317-1328. [PMID: 35147850 DOI: 10.1007/s11064-022-03531-1] [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: 10/15/2021] [Revised: 01/11/2022] [Accepted: 01/15/2022] [Indexed: 10/19/2022]
Abstract
Prenatal exposure to amphetamine induces changes in dopamine receptors in mesolimbic areas and alters locomotor response to amphetamine during adulthood. Sex differences have been reported in amphetamine-induced brain activity and stress sensitivity. We evaluated the effects of prenatal amphetamine exposure on locomotor activity, dopamine receptors and tyrosine hydroxylase mRNA expression in nucleus accumbens and caudate-putamen in response to amphetamine challenge in adult female and male rats. The role of estrogen in the response to restraint stress was analyzed in ovariectomized, prenatally amphetamine-exposed rats. Pregnant rats were treated with D-amphetamine during days 15-21 of gestation. Nucleus accumbens and caudate-putamen were processed for mRNA determination by real-time PCR. In nucleus accumbens, higher mRNA dopamine (D3) receptor expression was found in basal and D-amphetamine-challenge conditions in female than male, and prenatal amphetamine increased the difference. No sex differences were observed in caudate-putamen. Basal saline-treated females showed higher locomotor activity than males. Amphetamine challenge in prenatally amphetamine-exposed rats increased locomotor activity in males and reduced it in females. In nucleus accumbens, estrogen diminished mRNA D1, D2 and D3 receptor expression in basal, and D1 and D3 in ovariectomized stressed rats. Estrogen prevented the increase in tyrosine hydroxylase expression induced by stress in ovariectomized prenatally exposed rats. In conclusion, estrogen modulates mRNA levels of D1, D2 and D3 receptors and tyrosine hydroxylase expression in nucleus accumbens; prenatal amphetamine-exposure effects on D3 receptors and behavioral responses were gender dependent.
Collapse
|
37
|
Kim SJ, Eom H, Hoon Jung Y, Kim MK, Kim E, Kim JJ. Brain functional connectivity during and after imagery of gaming and alternative leisure activities in patients with internet gaming disorder. Neurosci Lett 2022; 772:136451. [PMID: 35041909 DOI: 10.1016/j.neulet.2022.136451] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/10/2022] [Accepted: 01/10/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND The effect of gaming cue exposure on brain activity in patients with internet gaming disorder (IGD) has been investigated a lot, but the effect on brain connectivity has not. This study aimed to investigate the effects of imageries of gaming and alternative leisure activities on functional connectivity during the during-task and post-task states in patients with IGD. METHODS Twenty-nine patients and 20 healthy controls were scanned in the 6-min states before, during, and after the imagery tasks for gaming and alternative leisure behaviors using fMRI. Seed-based functional connectivity during and after the tasks were analyzed. The seeds were the nucleus accumbens (NAcc), ventral tegmental area (VTA), caudate, putamen, anterior cingulate cortex (ACC), and posterior cingulate cortex. RESULTS The group-by-state interaction effects for the during-tasks were found in caudate-, putamen-, and ACC-based connectivity, whereas those for the post-tasks were shown only in NAcc-based connectivity. In particular, patients showed that caudate-right parahippocampal gyrus connectivity and putamen-right orbitofrontal cortex connectivity increased during gaming and decreased during alternative, whereas NAcc-right precuneus connectivity decreased at baseline, increased in post-gaming, and were not different in post-alternative. CONCLUSION Differences in during-task connectivity of the habit/motor and salience networks and post-task resting-state connectivity of the reward and limbic networks between the two imagery tasks may differ between the groups. In the treatment of IGD, when these network connections are reactive to alternative leisure activity, just as to gaming activity, they seem to be freed from gaming addiction.
Collapse
Affiliation(s)
- Soo-Jeong Kim
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyojung Eom
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Hoon Jung
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min-Kyeong Kim
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eunjoo Kim
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Psychiatry, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae-Jin Kim
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Psychiatry, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
38
|
Francis AM, Bissonnette JN, MacNeil SE, Crocker CE, Tibbo PG, Fisher DJ. Interaction of sex and cannabis in adult in vivo brain imaging studies: A systematic review. Brain Neurosci Adv 2022; 6:23982128211073431. [PMID: 35097219 PMCID: PMC8793398 DOI: 10.1177/23982128211073431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 12/22/2021] [Indexed: 12/21/2022] Open
Abstract
Cannabis has been shown to cause structural and functional neurocognitive changes in heavy users. Cannabis use initiation aligns with brain development trajectories; therefore, it is imperative that the potential neurological implications of cannabis use are understood. Males and females reach neurodevelopmental milestones at different rates making it necessary to consider biological sex in all cannabis and brain-based research. Through use of a systamatic review in accordance with PRISMA guidelines, we aimed to understand the interaction between biological sex and cannabis use on brain-based markers. In total, 18 articles containing a sex-based analysis of cannabis users were identified. While the majority of studies (n = 11) reported no sex by cannabis use interactions on brain-based markers, those that reported findings (n = 8) suggest females may be more susceptible to cannabis' neurotoxic effects. Unfortunately, a large portion of the literature was excluded due to no sex-based analysis. In addition, studies that reported no sex differences often contained a reduced number of females which may result in some studies being underpowered for sex-based analyses, making it difficult to draw firm conclusions. Suggestions to improve cannabis and sex-based reseach are proposed.
Collapse
Affiliation(s)
- Ashley M. Francis
- Department of Psychology, Saint Mary’s University, Halifax, NS, Canada
| | - Jenna N. Bissonnette
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
- Department of Psychology, Mount Saint Vincent University, Halifax, NS, Canada
| | - Sarah E. MacNeil
- Department of Psychology, Mount Saint Vincent University, Halifax, NS, Canada
| | - Candice E. Crocker
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
- Department of Diagnostic Radiology, Dalhousie University, Halifax, NS, Canada
| | - Philip G. Tibbo
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Derek J. Fisher
- Department of Psychology, Saint Mary’s University, Halifax, NS, Canada
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
- Department of Psychology, Mount Saint Vincent University, Halifax, NS, Canada
| |
Collapse
|
39
|
Grunze H, Csehi R, Born C, Barabássy Á. Reducing Addiction in Bipolar Disorder via Hacking the Dopaminergic System. Front Psychiatry 2021; 12:803208. [PMID: 34970175 PMCID: PMC8712474 DOI: 10.3389/fpsyt.2021.803208] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 11/23/2021] [Indexed: 02/05/2023] Open
Abstract
The dopaminergic system plays a central and decisive role in substance use disorder (SUD), bipolar disorder (BD), and possibly in a subgroup of patients with refractory depression. Common genetic markers and underlying cellular processes, such as kindling, support the close link between these disorders, which is also expressed by the high rate of comorbidity. Although partial dopamine agonists/antagonists acting on D2 and D3 receptors have an established role in treating BD, their usefulness in SUD is less clear. However, dopamine D3 receptors were shown to play a central role in SUD and BD, making D2/D3 partial agonists/antagonists a potential target for both disorders. This narrative review examines whether these substances bear the promise of a future therapeutic approach especially in patients with comorbid BD and SUD.
Collapse
Affiliation(s)
- Heinz Grunze
- Psychiatrie Schwäbisch Hall, Schwäbisch Hall, Germany
- Paracelsus Medical University, Nuremberg, Germany
| | - Réka Csehi
- Gedeon Richter Plc, Medical Division, Budapest, Hungary
| | - Christoph Born
- Psychiatrie Schwäbisch Hall, Schwäbisch Hall, Germany
- Paracelsus Medical University, Nuremberg, Germany
| | | |
Collapse
|
40
|
Sørensen G, Rickhag M, Leo D, Lycas MD, Ridderstrøm PH, Weikop P, Lilja JH, Rifes P, Herborg F, Woldbye D, Wörtwein G, Gainetdinov RR, Fink-Jensen A, Gether U. Disruption of the PDZ domain-binding motif of the dopamine transporter uniquely alters nanoscale distribution, dopamine homeostasis, and reward motivation. J Biol Chem 2021; 297:101361. [PMID: 34756883 PMCID: PMC8648841 DOI: 10.1016/j.jbc.2021.101361] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 10/21/2021] [Accepted: 10/26/2021] [Indexed: 11/04/2022] Open
Abstract
The dopamine (DA) transporter (DAT) is part of a presynaptic multiprotein network involving interactions with scaffold proteins via its C-terminal PDZ domain-binding sequence. Using a mouse model expressing DAT with mutated PDZ-binding sequence (DAT-AAA), we previously demonstrated the importance of this binding sequence for striatal expression of DAT. Here, we show by application of direct stochastic reconstruction microscopy not only that the striatal level of transporter is reduced in DAT-AAA mice but also that the nanoscale distribution of this transporter is altered with a higher propensity of DAT-AAA to localize to irregular nanodomains in dopaminergic terminals. In parallel, we observe mesostriatal DA adaptations and changes in DA-related behaviors distinct from those seen in other genetic DAT mouse models. DA levels in the striatum are reduced to ∼45% of that of WT, accompanied by elevated DA turnover. Nonetheless, fast-scan cyclic voltammetry recordings on striatal slices reveal a larger amplitude and prolonged clearance rate of evoked DA release in DAT-AAA mice compared with WT mice. Autoradiography and radioligand binding show reduced DA D2 receptor levels, whereas immunohistochemistry and autoradiography show unchanged DA D1 receptor levels. In behavioral experiments, we observe enhanced self-administration of liquid food under both a fixed ratio of one and progressive ratio schedule of reinforcement but a reduction compared with WT when using cocaine as reinforcer. In summary, our data demonstrate how disruption of PDZ domain interactions causes changes in DAT expression and its nanoscopic distribution that in turn alter DA clearance dynamics and related behaviors.
Collapse
Affiliation(s)
- Gunnar Sørensen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Laboratory of Neuropsychiatry, Psychiatric Center Copenhagen, Mental Health Center & University of Copenhagen, Copenhagen, Denmark
| | - Mattias Rickhag
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Damiana Leo
- Neuroscience and Brain Technologies Department, Italian Institute of Technology, Genoa, Italy
| | - Matthew D Lycas
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pernille Herrstedt Ridderstrøm
- Laboratory of Neuropsychiatry, Psychiatric Center Copenhagen, Mental Health Center & University of Copenhagen, Copenhagen, Denmark
| | - Pia Weikop
- Laboratory of Neuropsychiatry, Psychiatric Center Copenhagen, Mental Health Center & University of Copenhagen, Copenhagen, Denmark
| | - Jamila H Lilja
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pedro Rifes
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Freja Herborg
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - David Woldbye
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gitta Wörtwein
- Laboratory of Neuropsychiatry, Psychiatric Center Copenhagen, Mental Health Center & University of Copenhagen, Copenhagen, Denmark
| | - Raul R Gainetdinov
- Institute of Translational Biomedicine and Saint-Petersburg University Hospital, Saint-Petersburg State University, Saint-Petersburg, Russia
| | - Anders Fink-Jensen
- Laboratory of Neuropsychiatry, Psychiatric Center Copenhagen, Mental Health Center & University of Copenhagen, Copenhagen, Denmark
| | - Ulrik Gether
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
41
|
Seo EH, Yang HJ, Kim SG, Park SC, Lee SK, Yoon HJ. A Literature Review on the Efficacy and Related Neural Effects of Pharmacological and Psychosocial Treatments in Individuals With Internet Gaming Disorder. Psychiatry Investig 2021; 18:1149-1163. [PMID: 34872237 PMCID: PMC8721297 DOI: 10.30773/pi.2021.0207] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/05/2021] [Accepted: 10/27/2021] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Internet gaming disorder (IGD) has attracted considerable attention as a serious mental and public health issue worldwide. Currently, there are no established treatment guidelines for IGD. Herein, we review the latest findings on the efficacy and related neural effects of pharmacological and psychosocial treatments for individuals with IGD. METHODS A database search of relevant studies published between 2007 and 2020 was conducted using PubMed and Google Scholar. Twenty-seven studies were reviewed for current evidence related to the efficacy and neural effects of pharmacological and psychosocial IGD treatments. RESULTS Pharmacological studies suggest that bupropion may play a significant role in IGD. Additionally, nuclear imaging studies on IGD have demonstrated functional impairment of the dopamine system, providing a neurobiological basis for the efficacy of dopamineenhancing drugs. Among the various psychosocial interventions, current evidence suggests that cognitive behavioral therapy may be an effective intervention for IGD. Cognitive behavioral therapy and bupropion were found to influence resting-state functional connectivity within the cortico-subcortical circuit and default mode network, suggesting a possible neural mechanism. Innovative approaches, including virtual reality treatment, residential camps, voluntary abstinence, and transcranial direct current stimulation, have shown promising results. However, methodological limitations, such as the absence of proper controls, small sample sizes, short duration, inconsistency of inclusion criteria across studies, and self-report measures of outcome, hamper conclusions regarding the efficacy of treatments. CONCLUSION Ongoing basic research and clinical trials overcoming these limitations could add to the existing knowledge on IGD and contribute to the development of evidence-based treatments.
Collapse
Affiliation(s)
- Eun Hyun Seo
- Premedical Science, College of Medicine, Chosun University, Gwangju, Republic of Korea
| | - Hae-Jung Yang
- Department of Psychiatry, Chosun University Hospital, Gwangju, Republic of Korea
| | - Seung-Gon Kim
- Department of Psychiatry, Chosun University Hospital, Gwangju, Republic of Korea
- Department of Psychiatry, College of Medicine, Chosun University, Gwangju, Republic of Korea
| | - Seon-Cheol Park
- Department of Psychiatry, Hanyang University Guri Hospital, Guri, Republic of Korea
| | - Sang-Kyu Lee
- Department of Psychiatry, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Hyung-Jun Yoon
- Department of Psychiatry, Chosun University Hospital, Gwangju, Republic of Korea
- Department of Psychiatry, College of Medicine, Chosun University, Gwangju, Republic of Korea
| |
Collapse
|
42
|
Loganathan K. Value-based cognition and drug dependency. Addict Behav 2021; 123:107070. [PMID: 34359016 DOI: 10.1016/j.addbeh.2021.107070] [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: 04/13/2021] [Revised: 07/03/2021] [Accepted: 07/26/2021] [Indexed: 10/20/2022]
Abstract
Value-based decision-making is thought to play an important role in drug dependency. Achieving elevated levels of euphoria or ameliorating dysphoria/pain may motivate goal-directed drug consumption in both drug-naïve and long-time users. In other words, drugs become viewed as the preferred means of attaining a desired internal state. The bias towards choosing drugs may affect one's cognition. Observed biases in learning, attention and memory systems within the brain gradually focus one's cognitive functions towards drugs and related cues to the exclusion of other stimuli. In this narrative review, the effects of drug use on learning, attention and memory are discussed with a particular focus on changes across brain-wide functional networks and the subsequent impact on behaviour. These cognitive changes are then incorporated into the cycle of addiction, an established model outlining the transition from casual drug use to chronic dependency. If drug use results in the elevated salience of drugs and their cues, the studies highlighted in this review strongly suggest that this salience biases cognitive systems towards the motivated pursuit of addictive drugs. This bias is observed throughout the cycle of addiction, possibly contributing to the persistent hold that addictive drugs have over the dependent. Taken together, the excessive valuation of drugs as the preferred means of achieving a desired internal state affects more than just decision-making, but also learning, attentional and mnemonic systems. This eventually narrows the focus of one's thoughts towards the pursuit and consumption of addictive drugs.
Collapse
|
43
|
Chand S, Gowen A, Savine M, Moore D, Clark A, Huynh W, Wu N, Odegaard K, Weyrich L, Bevins RA, Fox HS, Pendyala G, Yelamanchili SV. A comprehensive study to delineate the role of an extracellular vesicle-associated microRNA-29a in chronic methamphetamine use disorder. J Extracell Vesicles 2021; 10:e12177. [PMID: 34913274 PMCID: PMC8674191 DOI: 10.1002/jev2.12177] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 10/27/2021] [Accepted: 11/25/2021] [Indexed: 12/17/2022] Open
Abstract
Extracellular vesicles (EVs), which express a repertoire of cargo molecules (cf. proteins, microRNA, lipids, etc.), have been garnering a prominent role in the modulation of several cellular processes. Here, using both non-human primate and rodent model systems, we provide evidence that brain-derived EV (BDE) miRNA, miR-29a-3p (mir-29a), is significantly increased during chronic methamphetamine (MA) exposure. Further, miR-29a levels show significant increase both with drug-seeking and reinstatement in a rat MA self-administration model. We also show that EV-associated miR-29a is enriched in EV pool comprising of small EVs and exomeres and further plays a critical role in MA-induced inflammation and synaptodendritic damage. Furthermore, treatment with the anti-inflammatory drug ibudilast (AV411), which is known to reduce MA relapse, decreased the expression of miR-29a and subsequently attenuated inflammation and rescued synaptodendritic injury. Finally, using plasma from MUD subjects, we provide translational evidence that EV-miR29a could potentially serve as a biomarker to detect neuronal damage in humans diagnosed with MA use disorder (MUD). In summary, our work suggests that EV-associated miR-29a-3p plays a crucial role in MUD and might be used as a potential blood-based biomarker for detecting chronic inflammation and synaptic damage.
Collapse
Affiliation(s)
- Subhash Chand
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Austin Gowen
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Mason Savine
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Dalia Moore
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Alexander Clark
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Wendy Huynh
- Department of PsychologyUniversity of Nebraska–Lincoln (UNL)LincolnNebraskaUSA
| | - Niming Wu
- Department of Pharmacology and Experimental NeuroscienceUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Katherine Odegaard
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | | | - Rick A. Bevins
- Department of PsychologyUniversity of Nebraska–Lincoln (UNL)LincolnNebraskaUSA
| | - Howard S. Fox
- Department of Neurological SciencesUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Gurudutt Pendyala
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Sowmya V. Yelamanchili
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| |
Collapse
|
44
|
Miller DR, Bu M, Gopinath A, Martinez LR, Khoshbouei H. Methamphetamine Dysregulation of the Central Nervous System and Peripheral Immunity. J Pharmacol Exp Ther 2021; 379:372-385. [PMID: 34535563 PMCID: PMC9351721 DOI: 10.1124/jpet.121.000767] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 09/16/2021] [Indexed: 11/22/2022] Open
Abstract
Methamphetamine (METH) is a potent psychostimulant that increases extracellular monoamines, such as dopamine and norepinephrine, and affects multiple tissue and cell types in the central nervous system (CNS) and peripheral immune cells. The reinforcing properties of METH underlie its significant abuse potential and dysregulation of peripheral immunity and central nervous system functions. Together, the constellation of METH's effects on cellular targets and regulatory processes has led to immune suppression and neurodegeneration in METH addicts and animal models of METH exposure. Here we extensively review many of the cell types and mechanisms of METH-induced dysregulation of the central nervous and peripheral immune systems. SIGNIFICANCE STATEMENT: Emerging research has begun to show that methamphetamine regulates dopaminergic neuronal activity. In addition, METH affects non-neuronal brain cells, such as microglia and astrocytes, and immunological cells of the periphery. Concurrent disruption of bidirectional communication between dopaminergic neurons and glia in the CNS and peripheral immune cell dysregulation gives rise to a constellation of dysfunctional neuronal, cell, and tissue types. Therefore, understanding the pathophysiology of METH requires consideration of the multiple targets at the interface between basic and clinical neuroscience.
Collapse
Affiliation(s)
- Douglas R Miller
- Department of Neuroscience, College of Medicine (D.R.M., M.B., A.G., H.K.), and Department of Oral Biology, College of Dentistry (L.R.M.), University of Florida, Gainesville, Florida
| | - Mengfei Bu
- Department of Neuroscience, College of Medicine (D.R.M., M.B., A.G., H.K.), and Department of Oral Biology, College of Dentistry (L.R.M.), University of Florida, Gainesville, Florida
| | - Adithya Gopinath
- Department of Neuroscience, College of Medicine (D.R.M., M.B., A.G., H.K.), and Department of Oral Biology, College of Dentistry (L.R.M.), University of Florida, Gainesville, Florida
| | - Luis R Martinez
- Department of Neuroscience, College of Medicine (D.R.M., M.B., A.G., H.K.), and Department of Oral Biology, College of Dentistry (L.R.M.), University of Florida, Gainesville, Florida
| | - Habibeh Khoshbouei
- Department of Neuroscience, College of Medicine (D.R.M., M.B., A.G., H.K.), and Department of Oral Biology, College of Dentistry (L.R.M.), University of Florida, Gainesville, Florida
| |
Collapse
|
45
|
Srinivasan C, Phan BN, Lawler AJ, Ramamurthy E, Kleyman M, Brown AR, Kaplow IM, Wirthlin ME, Pfenning AR. Addiction-Associated Genetic Variants Implicate Brain Cell Type- and Region-Specific Cis-Regulatory Elements in Addiction Neurobiology. J Neurosci 2021; 41:9008-9030. [PMID: 34462306 PMCID: PMC8549541 DOI: 10.1523/jneurosci.2534-20.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 06/18/2021] [Accepted: 07/10/2021] [Indexed: 12/14/2022] Open
Abstract
Recent large genome-wide association studies have identified multiple confident risk loci linked to addiction-associated behavioral traits. Most genetic variants linked to addiction-associated traits lie in noncoding regions of the genome, likely disrupting cis-regulatory element (CRE) function. CREs tend to be highly cell type-specific and may contribute to the functional development of the neural circuits underlying addiction. Yet, a systematic approach for predicting the impact of risk variants on the CREs of specific cell populations is lacking. To dissect the cell types and brain regions underlying addiction-associated traits, we applied stratified linkage disequilibrium score regression to compare genome-wide association studies to genomic regions collected from human and mouse assays for open chromatin, which is associated with CRE activity. We found enrichment of addiction-associated variants in putative CREs marked by open chromatin in neuronal (NeuN+) nuclei collected from multiple prefrontal cortical areas and striatal regions known to play major roles in reward and addiction. To further dissect the cell type-specific basis of addiction-associated traits, we also identified enrichments in human orthologs of open chromatin regions of female and male mouse neuronal subtypes: cortical excitatory, D1, D2, and PV. Last, we developed machine learning models to predict mouse cell type-specific open chromatin, enabling us to further categorize human NeuN+ open chromatin regions into cortical excitatory or striatal D1 and D2 neurons and predict the functional impact of addiction-associated genetic variants. Our results suggest that different neuronal subtypes within the reward system play distinct roles in the variety of traits that contribute to addiction.SIGNIFICANCE STATEMENT We combine statistical genetic and machine learning techniques to find that the predisposition to for nicotine, alcohol, and cannabis use behaviors can be partially explained by genetic variants in conserved regulatory elements within specific brain regions and neuronal subtypes of the reward system. Our computational framework can flexibly integrate open chromatin data across species to screen for putative causal variants in a cell type- and tissue-specific manner for numerous complex traits.
Collapse
Affiliation(s)
- Chaitanya Srinivasan
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213
| | - BaDoi N Phan
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213
- Medical Scientist Training Program, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213
| | - Alyssa J Lawler
- Department of Biological Sciences, Mellon College of Science, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213
| | - Easwaran Ramamurthy
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213
| | - Michael Kleyman
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213
| | - Ashley R Brown
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213
| | - Irene M Kaplow
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213
| | - Morgan E Wirthlin
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213
| | - Andreas R Pfenning
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213
- Department of Biological Sciences, Mellon College of Science, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213
| |
Collapse
|
46
|
Klausen MK, Thomsen M, Wortwein G, Fink-Jensen A. The role of glucagon-like peptide 1 (GLP-1) in addictive disorders. Br J Pharmacol 2021; 179:625-641. [PMID: 34532853 DOI: 10.1111/bph.15677] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/21/2021] [Accepted: 06/19/2021] [Indexed: 11/29/2022] Open
Abstract
Drug-, alcohol- and tobacco use disorders are a global burden affecting millions of people. Despite decades of research, treatment options are sparse or missing, and relapse rates are high. Glucagon-like peptide-1 (GLP-1) is released in the small intestines, promotes blood glucose homeostasis, slows gastric emptying, and reduces appetite. GLP-1 receptor agonists approved for treating type 2 diabetes mellitus and obesity, have received attention as a potential anti-addiction treatment. Studies in rodents and non-human primates have demonstrated a reduction in intake of alcohol and drugs of abuse, and clinical trials have been initiated to investigate whether the preclinical findings can be translated to patients. This review will give an overview of current findings and discuss the possible mechanisms of action. We suggest that effects of GLP-1 in alcohol- and substance use disorder is mediated centrally, at least partly through dopamine signalling, but precise mechanisms are still to be uncovered.
Collapse
Affiliation(s)
- Mette Kruse Klausen
- Psychiatric Centre Copenhagen and University Hospital of Copenhagen, Copenhagen, Denmark
| | - Morgane Thomsen
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen and University Hospital of Copenhagen, Copenhagen, Denmark
| | - Gitta Wortwein
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen and University Hospital of Copenhagen, Copenhagen, Denmark
| | - Anders Fink-Jensen
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen and University Hospital of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
47
|
Jiang P, Sun J, Zhou X, Lu L, Li L, Huang X, Li J, Kendrick K, Gong Q. Functional connectivity abnormalities underlying mood disturbances in male abstinent methamphetamine abusers. Hum Brain Mapp 2021; 42:3366-3378. [PMID: 33939234 PMCID: PMC8249885 DOI: 10.1002/hbm.25439] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/09/2021] [Accepted: 03/26/2021] [Indexed: 02/05/2023] Open
Abstract
Anxiety and depression are the most common withdrawal symptoms of methamphetamine (METH) abuse, which further exacerbate relapse of METH abuse. To date, no effective pharmacotherapy exists for METH abuse and its withdrawal symptoms. Therefore, understanding the neuromechanism underlying METH abuse and its withdrawal symptoms is essential for developing clinical strategies and improving patient care. The aims of this study were to investigate brain network abnormalities in METH abusers (MAs) and their associations with affective symptoms. Forty-eight male abstinent MAs and 48 age-gender matched healthy controls were recruited and underwent resting state functional magnetic resonance imaging (fMRI). The severity of patient anxiety and depressive symptoms were measured by Hamilton anxiety and depression rating scales, which decreased across the duration of abstinence. Independent component analysis was used to investigate the brain network functional connectivity (FC) properties. Compared with healthy controls, MAs demonstrated hypo-intra-network FC in the cerebellar network and hyper-intra-network FC in the posterior salience network. A whole-brain regression analysis revealed that FC strength of clusters located in the right rostral anterior cingulate cortex (rACC) within the ventromedial network (VMN) was associated with affective symptoms in the patients. Importantly, the intra-network FC strength of the rACC in VMN mediated the association between abstinence duration and the severity level of affective symptoms. Our results demonstrate alterations in brain functional networks underlying METH abuse, and that the FC of rACC within VMN serve as a neural substrate in the association between abstinence length and affective symptom severity in the MAs.
Collapse
Affiliation(s)
- Ping Jiang
- Huaxi MR Research Center (HMRRC), Department of RadiologyWest China Hospital of Sichuan UniversityChengduChina
- Research Unit of PsychoradiologyChinese Academy of Medical SciencesChengduChina
- Functional and Molecular Imaging Key Laboratory of Sichuan ProvinceChengduChina
| | - Jiayu Sun
- Department of RadiologyWest China Hospital of Sichuan UniversityChengduChina
| | - Xiaobo Zhou
- Department of PsychosomaticsAcademy of Medical Sciences & Sichuan Provincial People's HospitalChengduSichuanChina
| | - Lu Lu
- Huaxi MR Research Center (HMRRC), Department of RadiologyWest China Hospital of Sichuan UniversityChengduChina
- Research Unit of PsychoradiologyChinese Academy of Medical SciencesChengduChina
- Functional and Molecular Imaging Key Laboratory of Sichuan ProvinceChengduChina
| | - Lei Li
- Huaxi MR Research Center (HMRRC), Department of RadiologyWest China Hospital of Sichuan UniversityChengduChina
- Research Unit of PsychoradiologyChinese Academy of Medical SciencesChengduChina
- Functional and Molecular Imaging Key Laboratory of Sichuan ProvinceChengduChina
| | - Xiaoqi Huang
- Huaxi MR Research Center (HMRRC), Department of RadiologyWest China Hospital of Sichuan UniversityChengduChina
- Research Unit of PsychoradiologyChinese Academy of Medical SciencesChengduChina
- Functional and Molecular Imaging Key Laboratory of Sichuan ProvinceChengduChina
| | - Jing Li
- Mental Health CenterWest China Hospital of Sichuan UniversityChengduChina
| | - Keith Kendrick
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for NeuroInformationUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of RadiologyWest China Hospital of Sichuan UniversityChengduChina
- Research Unit of PsychoradiologyChinese Academy of Medical SciencesChengduChina
- Functional and Molecular Imaging Key Laboratory of Sichuan ProvinceChengduChina
| |
Collapse
|
48
|
Zhang X, Talpos J, Berridge MS, Apana SM, Slikker W, Wang C, Paule MG. MicroPET/CT assessment of neurochemical effects in the brain after long-term methylphenidate treatment in nonhuman primates. Neurotoxicol Teratol 2021; 87:107017. [PMID: 34265415 DOI: 10.1016/j.ntt.2021.107017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 01/23/2023]
Abstract
Methylphenidate (MPH) is a psychostimulant approved by the FDA to treatment Attention-Deficit Hyperactivity Disorder (ADHD). MPH is believed to exert its pharmacological effects via preferential blockade of the dopamine transporter (DAT) and the norepinephrine transporter (NET), resulting in increased monoamine levels in the synapse. We used a quantitative non-invasive PET imaging technique to study the effects of long-term methylphenidate use on the central nervous system (CNS). We conducted microPET/CT scans on young adult male rhesus monkeys to monitor changes in the dopaminergic system. We used [18F] AV-133, a ligand for the vesicular monoamine transporter 2 (VMAT2), and [18F]FESP a ligand for the D2 and 5HT2 receptors. In this study we evaluated the effects if chronic MPH treatment in the nonhuman primates (NHP). Two-year-old, male rhesus monkeys were orally administered MPH diluted in the electrolyte replenisher, Prang, twice a day, five days per week (M-F) over an 8-year period. The dose of MPH was gradually escalated from 0.15 mg/kg initially to 2.5 mg/kg/dose for the low dose group, and 1.5 mg/kg to 12.5 mg/kg/dose for the high dose group (Rodriguez et al., 2010). Scans were performed on Mondays, about 60 h after their last treatment, to avoid the acute effects of MPH. Tracers were injected intravenously ten minutes before microPET/CT scanning. Sessions lasted about 120 min. The Logan reference tissue model was used to determine the Binding Potential (BP) of each tracer in the striatum with the cerebellar cortex time activity curve as an input function. Both MP treatment groups had a lower [18F] AV-133 BP, although this failed to reach statistical significance. MPH treatment did not have a significant effect on The BP of [18F] FESP in the striatum. Long-term administration of MPH did not significant change any of the marker of monoamine function used here. These data suggest that, despite lingering concerns, long-term use of methylphenidate does not negatively impact monoamine function. This study also demonstrates that microPET imaging can distinguish differences in binding potentials of a variety of radiotracers in the CNS of NHPs. This approach may provide minimally-invasive biomarkers of neurochemical processes associated with chronic exposure to CNS medications. (Supported by NCTR).
Collapse
Affiliation(s)
- X Zhang
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, United States of America.
| | - J Talpos
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, United States of America
| | - M S Berridge
- 3D Imaging, LLC, Little Rock, AR 72113 and University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States of America
| | - S M Apana
- 3D Imaging, LLC, Little Rock, AR 72113 and University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States of America
| | - W Slikker
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, United States of America
| | - C Wang
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, United States of America
| | - M G Paule
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, United States of America
| |
Collapse
|
49
|
Robinson AH, Perales JC, Volpe I, Chong TT, Verdejo‐Garcia A. Are methamphetamine users compulsive? Faulty reinforcement learning, not inflexibility, underlies decision making in people with methamphetamine use disorder. Addict Biol 2021; 26:e12999. [PMID: 33393187 DOI: 10.1111/adb.12999] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 01/12/2023]
Abstract
Methamphetamine use disorder involves continued use of the drug despite negative consequences. Such 'compulsivity' can be measured by reversal learning tasks, which involve participants learning action-outcome task contingencies (acquisition-contingency) and then updating their behaviour when the contingencies change (reversal). Using these paradigms, animal models suggest that people with methamphetamine use disorder (PwMUD) may struggle to avoid repeating actions that were previously rewarded but are now punished (inflexibility). However, difficulties in learning task contingencies (reinforcement learning) may offer an alternative explanation, with meaningful treatment implications. We aimed to disentangle inflexibility and reinforcement learning deficits in 35 PwMUD and 32 controls with similar sociodemographic characteristics, using novel trial-by-trial analyses on a probabilistic reversal learning task. Inflexibility was defined as (a) weaker reversal phase performance, compared with the acquisition-contingency phases, and (b) persistence with the same choice despite repeated punishments. Conversely, reinforcement learning deficits were defined as (a) poor performance across both acquisition-contingency and reversal phases and (b) inconsistent postfeedback behaviour (i.e., switching after reward). Compared with controls, PwMUD exhibited weaker learning (odds ratio [OR] = 0.69, 95% confidence interval [CI] [0.63-0.77], p < .001), though no greater accuracy reduction during reversal. Furthermore, PwMUD were more likely to switch responses after one reward/punishment (OR = 0.83, 95% CI [0.77-0.89], p < .001; OR = 0.82, 95% CI [0.72-0.93], p = .002) but just as likely to switch after repeated punishments (OR = 1.03, 95% CI [0.73-1.45], p = .853). These results indicate that PwMUD's reversal learning deficits are driven by weaker reinforcement learning, not inflexibility.
Collapse
Affiliation(s)
- Alex H. Robinson
- Turner Institute for Brain and Mental Health Monash University Melbourne Victoria Australia
| | - José C. Perales
- Department of Experimental Psychology, Mind, Brain, and Behavior Research Center (CIMCYC) University of Granada Granada Spain
| | - Isabelle Volpe
- Clinical and Social Research Team Turning Point, Eastern Health Melbourne Victoria Australia
- Eastern Health Clinical School Monash University Melbourne Victoria Australia
- Monash Addiction Research Centre Monash University Melbourne Victoria Australia
| | - Trevor T.‐J. Chong
- Turner Institute for Brain and Mental Health Monash University Melbourne Victoria Australia
| | - Antonio Verdejo‐Garcia
- Turner Institute for Brain and Mental Health Monash University Melbourne Victoria Australia
| |
Collapse
|
50
|
Limanaqi F, Busceti CL, Celli R, Biagioni F, Fornai F. Autophagy as a gateway for the effects of methamphetamine: From neurotransmitter release and synaptic plasticity to psychiatric and neurodegenerative disorders. Prog Neurobiol 2021; 204:102112. [PMID: 34171442 DOI: 10.1016/j.pneurobio.2021.102112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/27/2021] [Accepted: 06/18/2021] [Indexed: 02/07/2023]
Abstract
As a major eukaryotic cell clearing machinery, autophagy grants cell proteostasis, which is key for neurotransmitter release, synaptic plasticity, and neuronal survival. In line with this, besides neuropathological events, autophagy dysfunctions are bound to synaptic alterations that occur in mental disorders, and early on, in neurodegenerative diseases. This is also the case of methamphetamine (METH) abuse, which leads to psychiatric disturbances and neurotoxicity. While consistently altering the autophagy machinery, METH produces behavioral and neurotoxic effects through molecular and biochemical events that can be recapitulated by autophagy blockade. These consist of altered physiological dopamine (DA) release, abnormal stimulation of DA and glutamate receptors, as well as oxidative, excitotoxic, and neuroinflammatory events. Recent molecular insights suggest that METH early impairs the autophagy machinery, though its functional significance remains to be investigated. Here we discuss evidence suggesting that alterations of DA transmission and autophagy are intermingled within a chain of events underlying behavioral alterations and neurodegenerative phenomena produced by METH. Understanding how METH alters the autophagy machinery is expected to provide novel insights into the neurobiology of METH addiction sharing some features with psychiatric disorders and parkinsonism.
Collapse
Affiliation(s)
- Fiona Limanaqi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma, 55, 56126, Pisa, PI, Italy
| | | | - Roberta Celli
- IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli, IS, Italy
| | | | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma, 55, 56126, Pisa, PI, Italy; IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli, IS, Italy.
| |
Collapse
|