1
|
Xiao N, Yin L, Teopiz KM, Kwan ATH, Le GH, Wong S, Valentino K, Choi H, Rosenblat JD, Ho R, Lee S, McIntyre RS. The sigma-1 receptor: a mechanistically-informed therapeutic target for antidepressants. Expert Opin Ther Targets 2025. [PMID: 40298911 DOI: 10.1080/14728222.2025.2500424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025]
Abstract
INTRODUCTION The mechanism of action of antidepressants is not fully ascertained. In addition to monoamines, disparate other effectors are also implicated in the molecular and cellular effects of chronic stress including neurogenesis, neurodifferentiation, and neuroplasticity. Evidence suggests sigma-1 receptors (S1R) as a putative target and possible mediator of antidepressant activity. AREAS COVERED Data from preclinical and clinical trials was synthesized from inception to August 2024. Results showed that S1R regulate neurotransmitter availability and release (e.g. monoamines, glutamate), and influence intracellular Ca2+ levels as well as immune inflammatory responses. The introduction of the N-Methyl-D-aspartic Acid (NMDA) antagonist/S1R agonist dextromethorphan-bupropion in August of 2022 represented the first time the Food and Drug Administration (FDA) permitted language that the hypothesized mechanism of an antidepressant involved activity at S1R. We also describe the physiology, pathophysiology, and function of S1R. EXPERT OPINION Sigma-1 modulation is relevant to the mechanism of action of agents currently FDA-approved in major depressive disorder (MDD) (dextromethorphan-bupropion). Modulating sigma-1 systems is fit for purpose as it relates to future therapeutic discoveries and development in depressive and other mental disorders. Whether sigma-1 modulation is uniquely relevant to targeting dimensions of psychopathology that are more difficult to treat (i.e. anhedonia) awaits determination.
Collapse
Affiliation(s)
- Naomi Xiao
- Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada
- Department of Health Sciences, Queen's University, Kingston, Ontario, Canada
| | - Liyang Yin
- Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada
| | - Kayla M Teopiz
- Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada
| | - Angela T H Kwan
- Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Gia Han Le
- Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada
- Mood Disorder Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Sabrina Wong
- Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada
- Mood Disorder Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Kyle Valentino
- Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada
- Mood Disorder Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Hayun Choi
- Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Department of Psychiatry, Veteran Health Service Medical Center, Seoul, Republic of Korea
| | - Joshua D Rosenblat
- Mood Disorder Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Roger Ho
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore
- Division of Life Science (LIFS), Hong Kong University of Science and Technology (HKUST), Clear Water Bay, Hong Kong
| | - Serene Lee
- Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada
- Department of Health Sciences, Queen's University, Kingston, Ontario, Canada
| | - Roger S McIntyre
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Piechal A, Blecharz-Klin K, Jakimiuk A, Pyrzanowska J, Joniec-Maciejak I, Mirowska-Guzel D, Widy-Tyszkiewicz E. The effect of 3-di-o-tolylguanidine on the level of neurotransmitters in the cerebellum and related disorders of social behavior. Neuroscience 2025; 565:549-557. [PMID: 39694318 DOI: 10.1016/j.neuroscience.2024.12.017] [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: 07/01/2024] [Revised: 12/01/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024]
Abstract
It is common knowledge that the cerebellum is a structure of the central nervous system that influences the processes of balance and motor coordination. Recently its influence on social interactions has also been emphasized. The sigma receptor agonist: 3-di-o-tolylguanidine (DTG) is characterized by high affinity for sigma 1 and sigma 2 receptors, widely distributed in the cerebellum. In the experiment we assessed the effect of long term administration of DTG to adult male Sprague Dawley rats on social behavior and the concentration of neurotransmitters in the cerebellum. DTG was administered orally at a dose of 3 mg/kg body weight (bw) (DTG3), 10 mg/kg bw (DTG10) and 30 mg/kg bw (DTG30) for 9 weeks before the behavioral test. After the experiment, the concentration of catecholamines and amino acids in the cerebellum was assessed using high performance liquid chromatography (HPLC). Treatment groups showed reductions in social interactions such as grooming, sniffing and total time spent interacting. At the same time, it was shown that in the group receiving the lowest dose of the drug, a decrease in the concentration of dopamine and serotonin in the cerebellum was observed. Furthermore, changes in the concentration of taurine, alanine, glutamic acid and gamma-aminobutyric acid were observed in the treated groups. We found that long term administration of DTG disturbs animals' social interactions and the concentration of neurotransmitters in the cerebellum.
Collapse
Affiliation(s)
- Agnieszka Piechal
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1B, 02-097 Warsaw, Poland
| | - Kamilla Blecharz-Klin
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1B, 02-097 Warsaw, Poland.
| | - Alicja Jakimiuk
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1B, 02-097 Warsaw, Poland
| | - Justyna Pyrzanowska
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1B, 02-097 Warsaw, Poland
| | - Ilona Joniec-Maciejak
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1B, 02-097 Warsaw, Poland
| | - Dagmara Mirowska-Guzel
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1B, 02-097 Warsaw, Poland
| | - Ewa Widy-Tyszkiewicz
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1B, 02-097 Warsaw, Poland
| |
Collapse
|
3
|
Winge T, Schepmann D, Schmidt J, Wünsch B. Synthesis and structure-affinity relationships of spirocyclic σ 1 receptor ligands with tetrahydropyran scaffold. Eur J Med Chem 2025; 281:117002. [PMID: 39547080 DOI: 10.1016/j.ejmech.2024.117002] [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: 09/06/2024] [Revised: 10/16/2024] [Accepted: 10/25/2024] [Indexed: 11/17/2024]
Abstract
The σ1 receptor plays a key role in the regulation of various processes in the human body; it is involved in the development of neurodegenerative and neuropsychiatric diseases and is overexpressed in several human tumors rendering it an important target for potential drug candidates. In this project, spirocyclic σ1 receptor ligands with different substituents in 4- and 9-position were synthesized and investigated for their σ1 receptor affinity and selectivity over related targets. The σ1 affinity of the ligands was correlated with their lipophilicity (logD7.4 value) giving insight into their lipophilic ligand efficiency (LLE). The (pyridin-3-yl)methyl derivative 5i showed a promising balance of high σ1 affinity (Ki(σ1) = 3.9 nM) and selectivity (>250-fold) as well as high LLE of 5.8. 5i has a high plasma protein binding (89 %) and promising metabolic stability in the presence of mouse liver microsomes and NADPH (83 % intact after 90 min). Increasing the size of the piperidine ring of the spirocyclic ligands 5 to an azepane ring led to considerably increased σ1 affinity (Ki(5a) = 1.2 nM, Ki(23a) = 0.42 nM) and selectivity over σ2 receptors (5a: 45-fold, 23a: 150-fold).
Collapse
Affiliation(s)
- Tobias Winge
- Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, D-48149, Münster, Germany
| | - Dirk Schepmann
- Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, D-48149, Münster, Germany
| | - Judith Schmidt
- Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, D-48149, Münster, Germany
| | - Bernhard Wünsch
- Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, D-48149, Münster, Germany.
| |
Collapse
|
4
|
Verma K, Prasanth MI, Tencomnao T, Brimson JM. Ligand docking in the sigma-1 receptor compared to the sigma-1 receptor-BiP complex and the effects of agonists and antagonists on C. elegans lifespans. Biomed Pharmacother 2025; 182:117783. [PMID: 39729653 DOI: 10.1016/j.biopha.2024.117783] [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: 10/08/2024] [Revised: 12/15/2024] [Accepted: 12/20/2024] [Indexed: 12/29/2024] Open
Abstract
Model organisms are commonly used to study human diseases; we set out to understand the relevance of several model organisms with relation to the σ1R protein. The study explored the interactions of σ1R with various agonists, antagonists across different species. Ligand and protein-protein (σ1R-BiP) docking approaches were used to understand the significance of σ1R in modulating neuroprotective mechanisms and its potential role in Alzheimer's. Ligand docking revealed that common σ1R antagonists generally exhibited stronger σ1R binding than commonly used agonists. Human σ1R showed high binding affinity for S1RA and NE100. Orthologs in yeast, slime mold, and C. elegans displayed varied binding affinities, indicating evolutionary adaptation in their binding pockets. We evaluated the relevance of σ1R-ligand interactions in C. elegans, measuring life-spans showing the impact of ligands on lifespan depends on genetic background and amyloid-beta pathology. Haloperidol (5-10 mM) extended wild-type worms' lifespan, but this effect was absent in the σ1R-KO, suggesting at least a partial role for the σ1R. Fluoxetine (5-10 mM) also promoted a small increase in longevity in wild-type worms but was not seen in the σ1R-KO strain. BD1047 (5 & 10 mM) reduced the lifespan of amyloid-beta-expressing transgenic worms, whereas dipentylamine (DPA) (5 mM) significantly increased the lifespan in a σ1R antagonist-sensitive manner. These findings highlight the importance of the σ1R in neurodegeneration and suggest that ligand interactions are modulated by BiP. Further research using in-vitro and in-vivo models is needed to clarify σ1R's therapeutic potential in neurodegenerative diseases, where modulating σ1R could provide neuroprotective effects.
Collapse
Affiliation(s)
- Kanika Verma
- Center of Excellence on Natural Products for Neuroprotection and Anti-Ageing, Chulalongkorn University, Bangkok 10330, Thailand; Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Mani Iyer Prasanth
- Center of Excellence on Natural Products for Neuroprotection and Anti-Ageing, Chulalongkorn University, Bangkok 10330, Thailand; Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Tewin Tencomnao
- Center of Excellence on Natural Products for Neuroprotection and Anti-Ageing, Chulalongkorn University, Bangkok 10330, Thailand; Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - James Michael Brimson
- Center of Excellence on Natural Products for Neuroprotection and Anti-Ageing, Chulalongkorn University, Bangkok 10330, Thailand; Research, Innovation and International Affairs, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
5
|
Fernandes-Nascimento MH, Negrão AB, Viana-Ferreira K, Chaves BDR, Wang YP. Three Decades of Research on the Development of Ibogaine Treatment of Substance Use Disorders: A Scientometric Analysis. J Psychoactive Drugs 2024; 56:670-680. [PMID: 37927218 DOI: 10.1080/02791072.2023.2276230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 08/24/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023]
Abstract
Ibogaine is a natural psychoactive drug that has been investigated for its potential role in the treatment of substance use disorders since the mid-1960s. To evaluate the interest in ibogaine's use as a therapeutic agent, we performed a scientometric analysis covering the last three decades (1993-2002, 2003-2012, and 2013-2022). A complementary analysis was performed to select and describe published clinical trials and meta-analyses. A total of 1523 references were found. Linear growth of publications in the first and third decades were identified, and the average number of publications from 1993 to 2002 was lower than that in the other two decades. Researchers from five continents were identified. Globally, academic research centers in the United States and Canada were the most productive. Cocaine, tobacco, morphine, and alcohol prevailed as major keywords in the first two decades and opioids and psychedelics were included in the third decade. A few key authors were the most co-referenced. One preclinical meta-analysis and no meta-analysis in humans were found. Research trends for ibogaine are widespread, growing, and consonant with current attentiveness in drug abuse. Our findings support the pressing need for rigorous clinical research on ibogaine to evaluate its efficacy and safety.
Collapse
Affiliation(s)
- Maria Helha Fernandes-Nascimento
- Programa de Pós-Graduação, Fisiopatologia Experimental, Departamento de Patologia, Faculdade Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - André Brooking Negrão
- Instituto Perdizes, Hospital das Clínicas (HCFMUSP), Faculdade Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Karine Viana-Ferreira
- Programa de Pós-Graduação, Fisiopatologia Experimental, Departamento de Patologia, Faculdade Medicina, Universidade de São Paulo, São Paulo, Brazil
| | | | - Yuan-Pang Wang
- Instituto de Psiquiatria (LIM-23), Hospital das Clínicas (HCFMUSP), Faculdade Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
6
|
Zhou JT, Xu Y, Liu XH, Cheng C, Fan JN, Li X, Yu J, Li S. Single-cell RNA-seq Reveals the Inhibitory Effect of Methamphetamine on Liver Immunity with the Involvement of Dopamine Receptor D1. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae060. [PMID: 39196711 PMCID: PMC11576359 DOI: 10.1093/gpbjnl/qzae060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/25/2024] [Accepted: 08/22/2024] [Indexed: 08/30/2024]
Abstract
Methamphetamine (METH) is a highly addictive psychostimulant that causes physical and psychological damage and immune system disorder, especially in the liver which contains a significant number of immune cells. Dopamine, a key neurotransmitter in METH addiction and immune regulation, plays a crucial role in this process. Here, we developed a chronic METH administration model and conducted single-cell RNA sequencing (scRNA-seq) to investigate the effect of METH on liver immune cells and the involvement of dopamine receptor D1 (DRD1). Our findings reveal that chronic exposure to METH induces immune cell identity shifts from IFITM3+ macrophage (Mac) and CCL5+ Mac to CD14+ Mac, as well as from FYN+CD4+ T effector (Teff), CD8+ T, and natural killer T (NKT) to FOS+CD4+ T and RORα+ group 2 innate lymphoid cell (ILC2), along with the suppression of multiple functional immune pathways. DRD1 is implicated in regulating certain pathways and identity shifts among the hepatic immune cells. Our results provide valuable insights into the development of targeted therapies to mitigate METH-induced immune impairment.
Collapse
Affiliation(s)
- Jin-Ting Zhou
- Key Laboratory of National Health Commission for Forensic Sciences, Xi’an Jiaotong University, Xi'an 710061, China
- National Biosafety Evidence Foundation, Bio-evidence Sciences Academy, Xi'an Jiaotong University, Xi'an 710115, China
| | - Yungang Xu
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Xiao-Huan Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Cheng Cheng
- Key Laboratory of National Health Commission for Forensic Sciences, Xi’an Jiaotong University, Xi'an 710061, China
- National Biosafety Evidence Foundation, Bio-evidence Sciences Academy, Xi'an Jiaotong University, Xi'an 710115, China
| | - Jing-Na Fan
- Key Laboratory of National Health Commission for Forensic Sciences, Xi’an Jiaotong University, Xi'an 710061, China
- National Biosafety Evidence Foundation, Bio-evidence Sciences Academy, Xi'an Jiaotong University, Xi'an 710115, China
| | - Xiaoming Li
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, Precision Medical Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Jun Yu
- National Biosafety Evidence Foundation, Bio-evidence Sciences Academy, Xi'an Jiaotong University, Xi'an 710115, China
- OneHealth Technology Company, Xi'an 710000, China
| | - Shengbin Li
- Key Laboratory of National Health Commission for Forensic Sciences, Xi’an Jiaotong University, Xi'an 710061, China
- National Biosafety Evidence Foundation, Bio-evidence Sciences Academy, Xi'an Jiaotong University, Xi'an 710115, China
| |
Collapse
|
7
|
Fan R, Gong X, Yu Z, Lin S, Ruan Y, Qian L, Si Z, Li L, Zhou W, Liu Y. The role of heterodimers formed by histamine H3 receptors and dopamine D1 receptors on the methamphetamine-induced conditioned place preference. Eur J Pharmacol 2024; 981:176866. [PMID: 39089461 DOI: 10.1016/j.ejphar.2024.176866] [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: 08/14/2023] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
RATIONALE The rewarding effect of Methamphetamine (METH) is commonly believed to play an important role in METH use disorder. The altered expression of dopamine D1 receptor (D1R) has been suggested to be essential to the rewarding effect of METH. Notably, D1R could interact with histamine H3 receptors (H3R) by forming a H3R-D1R heteromer (H3R-D1R). OBJECTIVES This study was designed to specifically investigate the involvement of H3R-D1R in the rewarding effect of METH. METHODS C57BL/6 mice were treated with intraperitoneal injections of a selective H3R antagonist (Thioperamide, THIO; 20 mg/kg), an H1R antagonist (Pyrilamine, PYRI; 10 mg/kg), or microinjections of cytomegalovirus (CMV)-transmembrane domain 5 (TM5) into the nucleus accumbens (NAc). The animal model of Conditioned Place Preference (CPP) was applied to determine the impact of H3R-D1R on the rewarding effect of METH. RESULTS METH resulted in a significant preference for the drug-associated chamber, in conjunction with increased H3R and decreased D1R expression in both NAc and the ventral tegmental area (VTA). THIO significantly attenuated the rewarding effect of METH, accompanied by decreased H3R and increased D1R expression. In contrast, pyrilamine failed to produce the similar effects. Moreover, the inhibitory effect of THIO on METH-induced CPP was reversed by SKF38393, a D1R agonist. Furthermore, SCH23390, a D1R antagonist, counteracted the ameliorative effect of SKF38393 on THIO. Co-immunoprecipitation (CO-IP) experiments further demonstrated the specific interaction between H3R and D1R in METH CPP mice. The rewarding effect of METH was also significantly blocked by the interruption of CMV-transmembrane domain 5 (TM5), but not CMV-transmembrane domain 7 (TM7) in NAc. CONCLUSION These results suggest that modulating the activity of H3R-D1R complex holds promise for regulating METH use disorder and serves as a potential drug target for its treatment.
Collapse
Affiliation(s)
- Runyue Fan
- School of Public Health, Health Science Center, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, PR China
| | - Xinshuang Gong
- School of Public Health, Health Science Center, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, PR China
| | - Zhaoyin Yu
- School of Teaching and Education, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, PR China
| | - Shujun Lin
- School of Teaching and Education, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, PR China
| | - Yuer Ruan
- School of Teaching and Education, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, PR China
| | - Liyin Qian
- School of Public Health, Health Science Center, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, PR China
| | - Zizhen Si
- Department of Physiology and Pharmacology, School of Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, PR China, Ningbo
| | - Longhui Li
- Ningbo Kangning Hospital, 1 South Zhuangyu Road, Ningbo, Zhejiang, 315201, PR China
| | - Wenhua Zhou
- Ningbo Kangning Hospital, 1 South Zhuangyu Road, Ningbo, Zhejiang, 315201, PR China
| | - Yu Liu
- Department of Physiology and Pharmacology, School of Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, PR China, Ningbo.
| |
Collapse
|
8
|
Lin Y, Lin C, Chang JHY, Chiang DL, Lai F, Lin CJ. Psychomotor Symptoms, Cognitive Impairments, and Suicidal Thoughts after COVID-19 Infection: A Case Report and Possible Allostatic Mechanism. Ann Geriatr Med Res 2024; 28:370-374. [PMID: 38757260 PMCID: PMC11467520 DOI: 10.4235/agmr.24.0037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/30/2024] [Accepted: 05/04/2024] [Indexed: 05/18/2024] Open
Abstract
Although neuropsychiatric manifestations are common in survivors of coronavirus disease 2019 (COVID-19), the pathophysiology is not yet elucidated. Here we describe the case of a geriatric inpatient who developed post-COVID depression with psychomotor retardation, anxiety, hopelessness, executive function problems, and suicidal ideations. The language problems and cognitive impairments coemerged with the motor problems. We propose a mechanism associated with problems in energy prediction and regulation in which the coronavirus infection, which causes neuroinflammation and viral activity in the nervous system, interferes with the reward pathway and sensory prediction process. Sigma-1 receptor agonists such as sertraline may regulate energy expenditure and, thus, be beneficial to the process. The treatment improvements in our patient included those in the autonomic nervous system, activity, and circadian rhythm.
Collapse
Affiliation(s)
- Ying Lin
- Department of Psychiatry, MacKay Memorial Hospital, Taipei, Taiwan
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Chen Lin
- Department of Biomedical Sciences and Engineering, National Central University, Chunli, Taiwan
| | | | - Dai-Lun Chiang
- Financial Technology Applications Program, Ming Chuan University, Taoyuan, Taiwan
| | - Feipei Lai
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Chen-Ju Lin
- Department of Psychiatry, MacKay Memorial Hospital, Taipei, Taiwan
- Institute of Health and Welfare Policy, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
| |
Collapse
|
9
|
Miao L, Wang H, Li Y, Huang J, Wang C, Teng H, Xu L, Yang X, Tian Y, Yang G, Li J, Zeng X. Mechanisms and treatments of methamphetamine and HIV-1 co-induced neurotoxicity: a systematic review. Front Immunol 2024; 15:1423263. [PMID: 39224601 PMCID: PMC11366655 DOI: 10.3389/fimmu.2024.1423263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
Combination antiretroviral therapy (cART) has dramatically reduced mortality in people with human immunodeficiency virus (HIV), but it does not completely eradicate the virus from the brain. Patients with long-term HIV-1 infection often show neurocognitive impairment, which severely affects the quality of life of those infected. Methamphetamine (METH) users are at a significantly higher risk of contracting HIV-1 through behaviors such as engaging in high-risk sex or sharing needles, which can lead to transmission of the virus. In addition, HIV-1-infected individuals who abuse METH exhibit higher viral loads and more severe cognitive dysfunction, suggesting that METH exacerbates the neurotoxicity associated with HIV-1. Therefore, this review focuses on various mechanisms underlying METH and HIV-1 infection co-induced neurotoxicity and existing interventions targeting the sigma 1 receptor, dopamine transporter protein, and other relevant targets are explored. The findings of this review are envisaged to systematically establish a theoretical framework for METH abuse and HIV-1 infection co-induced neurotoxicity, and to suggest novel clinical treatment targets.
Collapse
Affiliation(s)
- Lin Miao
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Haowei Wang
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Yi Li
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Jian Huang
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Chan Wang
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Hanxin Teng
- Department of Pathogen Biology and Immunology, School of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Lisha Xu
- Department of Pathogen Biology and Immunology, School of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Xue Yang
- Department of Pathogen Biology and Immunology, School of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Yunqing Tian
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Genmeng Yang
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Juan Li
- Department of Pathogen Biology and Immunology, School of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Xiaofeng Zeng
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| |
Collapse
|
10
|
Young EJ, Radnai L, Prikhodko V, Miller CA. Novel therapeutics in development for the treatment of stimulant-use disorder. Curr Opin Neurobiol 2024; 87:102898. [PMID: 39096558 DOI: 10.1016/j.conb.2024.102898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 07/14/2024] [Accepted: 07/14/2024] [Indexed: 08/05/2024]
Abstract
Misuse and accidental overdoses attributed to stimulants are escalating rapidly. These stimulants include methamphetamine, cocaine, amphetamine, ecstasy-type drugs, and prescription stimulants such as methylphenidate. Unlike opioids and alcohol, there are no therapies approved by the US Food and Drug Administration (FDA) to treat stimulant-use disorder. The high rate of relapse among this population highlights the insufficiency of current treatment options, which are limited to abstinence support programs and behavioral modification therapies. Here, we briefly outline recent regulatory actions taken by FDA to help support the development of new stimulant use disorder treatments and highlight several new therapeutics in the clinical development pipeline.
Collapse
Affiliation(s)
- Erica J Young
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA; Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA; Myosin Therapeutics, Jupiter, FL, USA
| | - Laszlo Radnai
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA; Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | | | - Courtney A Miller
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA; Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA.
| |
Collapse
|
11
|
Zhang KK, Yang JZ, Cheng CH, Wan JY, Chen YC, Zhou HQ, Zheng DK, Lan ZX, You QH, Wang Q, Sun J. Short-chain fatty acids mitigate Methamphetamine-induced hepatic injuries in a Sigma-1 receptor-dependent manner. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 280:116538. [PMID: 38833980 DOI: 10.1016/j.ecoenv.2024.116538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/08/2024] [Accepted: 05/31/2024] [Indexed: 06/06/2024]
Abstract
Methamphetamine (Meth) is a potent psychostimulant with well-established hepatotoxicity. Gut microbiota-derived short-chain fatty acids (SCFAs) have been reported to yield beneficial effects on the liver. In this study, we aim to further reveal the mechanisms of Meth-induced hepatic injuries and investigate the potential protective effects of SCFAs. Herein, mice were intraperitoneally injected with 15 mg/kg Meth to induce hepatic injuries. The composition of fecal microbiota and SCFAs was profiled using 16 S rRNA sequencing and Gas Chromatography/Mass Spectrometry (GC/MS) analysis, respectively. Subsequently, SCFAs supplementation was performed to evaluate the protective effects against hepatic injuries. Additionally, Sigma-1 receptor knockout (S1R-/-) mice and fluvoxamine (Flu), an agonist of S1R, were introduced to investigate the mechanisms underlying the protective effects of SCFAs. Our results showed that Meth activated S1R and induced hepatic autophagy, inflammation, and oxidative stress by stimulating the MAPK/ERK pathway. Meanwhile, Meth disrupted SCFAs product-related microbiota, leading to a reduction in fecal SCFAs (especially Acetic acid and Propanoic acid). Accompanied by the optimization of gut microbiota, SCFAs supplementation normalized S1R expression and ameliorated Meth-induced hepatic injuries by repressing the MAPK/ERK pathway. Effectively, S1R knockout repressed Meth-induced activation of the MAPK/ERK pathway and further ameliorated hepatic injuries. Finally, the overexpression of S1R stimulated the MAPK/ERK pathway and yielded comparable adverse phenotypes to Meth administration. These findings suggest that Meth-induced hepatic injuries relied on the activation of S1R, which could be alleviated by SCFAs supplementation. Our study confirms the crucial role of S1R in Meth-induced hepatic injuries for the first time and provides a potential preemptive therapy.
Collapse
Affiliation(s)
- Kai-Kai Zhang
- State Key Laboratory of Organ Failure Research; Key Laboratory of Infectious Diseases Research in South China, Ministry of Education; Guangdong Provincial Key Laboratory of Viral Hepatitis Research; Guangdong Provincial Clinical Research Center for Viral Hepatitis; Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China; Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jian-Zheng Yang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Chang-Hao Cheng
- State Key Laboratory of Organ Failure Research; Key Laboratory of Infectious Diseases Research in South China, Ministry of Education; Guangdong Provincial Key Laboratory of Viral Hepatitis Research; Guangdong Provincial Clinical Research Center for Viral Hepatitis; Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jia-Yuan Wan
- State Key Laboratory of Organ Failure Research; Key Laboratory of Infectious Diseases Research in South China, Ministry of Education; Guangdong Provincial Key Laboratory of Viral Hepatitis Research; Guangdong Provincial Clinical Research Center for Viral Hepatitis; Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yu-Chuan Chen
- State Key Laboratory of Organ Failure Research; Key Laboratory of Infectious Diseases Research in South China, Ministry of Education; Guangdong Provincial Key Laboratory of Viral Hepatitis Research; Guangdong Provincial Clinical Research Center for Viral Hepatitis; Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - He-Qi Zhou
- State Key Laboratory of Organ Failure Research; Key Laboratory of Infectious Diseases Research in South China, Ministry of Education; Guangdong Provincial Key Laboratory of Viral Hepatitis Research; Guangdong Provincial Clinical Research Center for Viral Hepatitis; Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - De-Kai Zheng
- State Key Laboratory of Organ Failure Research; Key Laboratory of Infectious Diseases Research in South China, Ministry of Education; Guangdong Provincial Key Laboratory of Viral Hepatitis Research; Guangdong Provincial Clinical Research Center for Viral Hepatitis; Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zhi-Xian Lan
- State Key Laboratory of Organ Failure Research; Key Laboratory of Infectious Diseases Research in South China, Ministry of Education; Guangdong Provincial Key Laboratory of Viral Hepatitis Research; Guangdong Provincial Clinical Research Center for Viral Hepatitis; Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Qiu-Hong You
- State Key Laboratory of Organ Failure Research; Key Laboratory of Infectious Diseases Research in South China, Ministry of Education; Guangdong Provincial Key Laboratory of Viral Hepatitis Research; Guangdong Provincial Clinical Research Center for Viral Hepatitis; Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Qi Wang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Jian Sun
- State Key Laboratory of Organ Failure Research; Key Laboratory of Infectious Diseases Research in South China, Ministry of Education; Guangdong Provincial Key Laboratory of Viral Hepatitis Research; Guangdong Provincial Clinical Research Center for Viral Hepatitis; Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
12
|
Wang KC, Ojeda NB, Wang H, Chiang HS, Tucci MA, Lee JW, Wei HC, Kaizaki-Mitsumoto A, Tanaka S, Dankhara N, Tien LT, Fan LW. Neonatal brain inflammation enhances methamphetamine-induced reinstated behavioral sensitization in adult rats analyzed with explainable machine learning. Neurochem Int 2024; 176:105743. [PMID: 38641026 PMCID: PMC11102812 DOI: 10.1016/j.neuint.2024.105743] [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: 12/18/2023] [Revised: 03/15/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024]
Abstract
Neonatal brain inflammation produced by intraperitoneal (i.p.) injection of lipopolysaccharide (LPS) results in long-lasting brain dopaminergic injury and motor disturbances in adult rats. The goal of the present work is to investigate the effect of neonatal systemic LPS exposure (1 or 2 mg/kg, i.p. injection in postnatal day 5, P5, male rats)-induced dopaminergic injury to examine methamphetamine (METH)-induced behavioral sensitization as an indicator of drug addiction. On P70, subjects underwent a treatment schedule of 5 once daily subcutaneous (s.c.) administrations of METH (0.5 mg/kg) (P70-P74) to induce behavioral sensitization. Ninety-six hours following the 5th treatment of METH (P78), the rats received one dose of 0.5 mg/kg METH (s.c.) to reintroduce behavioral sensitization. Hyperlocomotion is a critical index caused by drug abuse, and METH administration has been shown to produce remarkable locomotor-enhancing effects. Therefore, a random forest model was used as the detector to extract the feature interaction patterns among the collected high-dimensional locomotor data. Our approaches identified neonatal systemic LPS exposure dose and METH-treated dates as features significantly associated with METH-induced behavioral sensitization, reinstated behavioral sensitization, and perinatal inflammation in this experimental model of drug addiction. Overall, the analysis suggests that the implementation of machine learning strategies is sensitive enough to detect interaction patterns in locomotor activity. Neonatal LPS exposure also enhanced METH-induced reduction of dopamine transporter expression and [3H]dopamine uptake, reduced mitochondrial complex I activity, and elevated interleukin-1β and cyclooxygenase-2 concentrations in the P78 rat striatum. These results indicate that neonatal systemic LPS exposure produces a persistent dopaminergic lesion leading to a long-lasting change in the brain reward system as indicated by the enhanced METH-induced behavioral sensitization and reinstated behavioral sensitization later in life. These findings indicate that early-life brain inflammation may enhance susceptibility to drug addiction development later in life, which provides new insights for developing potential therapeutic treatments for drug addiction.
Collapse
Affiliation(s)
- Kuo-Ching Wang
- Department of Anesthesiology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei City, Taiwan
| | - Norma B Ojeda
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA; Department of Advanced Biomedical Education, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Haifeng Wang
- Department of Industrial and Systems Engineering, Mississippi State University, Mississippi State, MS, 39762, USA
| | - Han-Sun Chiang
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist, New Taipei City, 24205, Taiwan
| | - Michelle A Tucci
- Department of Anesthesiology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Jonathan W Lee
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Han-Chi Wei
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist, New Taipei City, 24205, Taiwan
| | - Asuka Kaizaki-Mitsumoto
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA; Department of Toxicology, Showa University Graduate School of Pharmacy, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Sachiko Tanaka
- Center for Research and Development in Pharmacy Education, School of Pharmacy, Nihon University, Funabashi, Chiba, 274-8555, Japan
| | - Nilesh Dankhara
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Lu-Tai Tien
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist, New Taipei City, 24205, Taiwan.
| | - Lir-Wan Fan
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA.
| |
Collapse
|
13
|
White E, Kennedy T, Ruffell S, Perkins D, Sarris J. Ayahuasca and Dimethyltryptamine Adverse Events and Toxicity Analysis: A Systematic Thematic Review. Int J Toxicol 2024; 43:327-339. [PMID: 38363085 PMCID: PMC11088222 DOI: 10.1177/10915818241230916] [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: 02/17/2024]
Abstract
The objective of this paper is to conduct a systematic thematic review of adverse events, safety, and toxicity of traditional ayahuasca plant preparations and its main psychoactive alkaloids (dimethyltryptamine [DMT], harmine, harmaline, and tetrahydroharmine), including discussing clinical considerations (within clinical trials or approved settings). A systematic literature search of preclinical, clinical, epidemiological, and pharmacovigilance data (as well as pertinent reviews and case studies) was conducted for articles using the electronic databases of PubMed and Web of Science (to 6 July 2023) and PsycINFO, ClinicalTrials.gov, and Embase (to 21 September 2022) and included articles in English in peer-reviewed journals. Additionally, reference lists were searched. Due to the breadth of the area covered, we presented the relevant data in a thematic format. Our searches revealed 78 relevant articles. Data showed that ayahuasca or DMT is generally safe; however, some adverse human events have been reported. Animal models using higher doses of ayahuasca have shown abortifacient and teratogenic effects. Isolated harmala alkaloid studies have also revealed evidence of potential toxicity at higher doses, which may increase with co-administration with certain medications. Harmaline revealed the most issues in preclinical models. Nevertheless, animal models involving higher-dose synthetic isolates may not necessarily be able to be extrapolated to human use of therapeutic doses of plant-based extracts. Serious adverse effects are rarely reported within healthy populations, indicating an acceptable safety profile for the traditional use of ayahuasca and DMT in controlled settings. Further randomized, controlled trials with judicious blinding, larger samples, and longer duration are needed.
Collapse
Affiliation(s)
- Eleanor White
- Biomedicine Discovery Institute, Department of Physiology, Monash University, Melbourne, VIC, Australia
| | - Tom Kennedy
- The University of Queensland, Brisbane, QLD, Australia
| | - Simon Ruffell
- Psychae Institue, Melbourne, VIC, Australia
- Onaya Science, Iquitos, Peru
| | - Daniel Perkins
- Psychae Institue, Melbourne, VIC, Australia
- School of Population and Global Health, University of Melbourne, Melbourne, VIC, Australia
- School of Social and Political Science, University of Melbourne, Melbourne, VIC, Australia
- Centre for Mental Health, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Jerome Sarris
- Psychae Institue, Melbourne, VIC, Australia
- Centre for Mental Health, Swinburne University of Technology, Melbourne, VIC, Australia
- NICM Health Research Institute, Western Sydney University, Westmead, NSW, Australia
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
14
|
Hersey M, Mereu M, Jones CS, Bartole MK, Chen AY, Cao J, Hiranita T, Chun LE, Lopez JP, Katz JL, Newman AH, Tanda G. Dual DAT and sigma receptor inhibitors attenuate cocaine effects on nucleus accumbens dopamine dynamics in rats. Eur J Neurosci 2024; 59:2436-2449. [PMID: 38444104 PMCID: PMC11108740 DOI: 10.1111/ejn.16293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 01/10/2024] [Accepted: 02/07/2024] [Indexed: 03/07/2024]
Abstract
Psychostimulant use disorders (PSUD) are prevalent; however, no FDA-approved medications have been made available for treatment. Previous studies have shown that dual inhibitors of the dopamine transporter (DAT) and sigma receptors significantly reduce the behavioral/reinforcing effects of cocaine, which have been associated with stimulation of extracellular dopamine (DA) levels resulting from DAT inhibition. Here, we employ microdialysis and fast scan cyclic voltammetry (FSCV) procedures to investigate the effects of dual inhibitors of DAT and sigma receptors in combination with cocaine on nucleus accumbens shell (NAS) DA dynamics in naïve male Sprague Dawley rats. In microdialysis studies, administration of rimcazole (3, 10 mg/kg; i.p.) or its structural analog SH 3-24 (1, 3 mg/kg; i.p.), compounds that are dual inhibitors of DAT and sigma receptors, significantly reduced NAS DA efflux stimulated by increasing doses of cocaine (0.1, 0.3, 1.0 mg/kg; i.v.). Using the same experimental conditions, in FSCV tests, we show that rimcazole pretreatments attenuated cocaine-induced stimulation of evoked NAS DA release but produced no additional effect on DA clearance rate. Under the same conditions, JJC8-091, a modafinil analog and dual inhibitor of DAT and sigma receptors, similarly attenuated cocaine-induced stimulation of evoked NAS DA release but produced no additional effect on DA clearance rate. Our results provide the neurochemical groundwork towards understanding actions of dual inhibitors of DAT and sigma receptors on DA dynamics that likely mediate the behavioral effects of psychostimulants like cocaine.
Collapse
Affiliation(s)
- Melinda Hersey
- Medication Development Program, NIDA IRP, Baltimore, MD 21224, USA
| | - Maddalena Mereu
- Medication Development Program, NIDA IRP, Baltimore, MD 21224, USA
| | - Claire S. Jones
- Medication Development Program, NIDA IRP, Baltimore, MD 21224, USA
| | | | - Andy Y. Chen
- Medication Development Program, NIDA IRP, Baltimore, MD 21224, USA
| | - Jianjing Cao
- Medicinal Chemistry Section, NIDA IRP, Baltimore, MD 21224, USA
| | | | - Lauren E. Chun
- Medication Development Program, NIDA IRP, Baltimore, MD 21224, USA
| | - Jessica P. Lopez
- Medication Development Program, NIDA IRP, Baltimore, MD 21224, USA
| | | | - Amy Hauck Newman
- Medication Development Program, NIDA IRP, Baltimore, MD 21224, USA
- Medicinal Chemistry Section, NIDA IRP, Baltimore, MD 21224, USA
| | - Gianluigi Tanda
- Medication Development Program, NIDA IRP, Baltimore, MD 21224, USA
| |
Collapse
|
15
|
Martin SL, Uribe C, Strafella AP. PET imaging of synaptic density in Parkinsonian disorders. J Neurosci Res 2024; 102:e25253. [PMID: 37814917 DOI: 10.1002/jnr.25253] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/31/2023] [Accepted: 09/21/2023] [Indexed: 10/11/2023]
Abstract
Synaptic dysfunction and altered synaptic pruning are present in people with Parkinsonian disorders. Dopamine loss and alpha-synuclein accumulation, two hallmarks of Parkinson's disease (PD) pathology, contribute to synaptic dysfunction and reduced synaptic density in PD. Atypical Parkinsonian disorders are likely to have unique spatiotemporal patterns of synaptic density, differentiating them from PD. Therefore, quantification of synaptic density has the potential to support diagnoses, monitor disease progression, and treatment efficacy. Novel radiotracers for positron emission tomography which target the presynaptic vesicle protein SV2A have been developed to quantify presynaptic density. The radiotracers have successfully investigated synaptic density in preclinical models of PD and people with Parkinsonian disorders. Therefore, this review will summarize the preclinical and clinical utilization of SV2A radiotracers in people with Parkinsonian disorders. We will evaluate how SV2A abundance is associated with other imaging modalities and the considerations for interpreting SV2A in Parkinsonian pathology.
Collapse
Affiliation(s)
- Sarah L Martin
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Carme Uribe
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Unitat de Psicologia Medica, Departament de Medicina, Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
| | - Antonio P Strafella
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Edmond J. Safra Parkinson Disease Program, Neurology Division, Toronto Western Hospital & Krembil Brain Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
16
|
Knowles LG, Armanious AJ, Peng Y, Welsh WJ, James MH. Recent advances in drug discovery efforts targeting the sigma 1 receptor system: Implications for novel medications designed to reduce excessive drug and food seeking. ADDICTION NEUROSCIENCE 2023; 8:100126. [PMID: 37753198 PMCID: PMC10519676 DOI: 10.1016/j.addicn.2023.100126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
Psychiatric disorders characterized by uncontrolled reward seeking, such as substance use disorders (SUDs), alcohol use disorder (AUD) and some eating disorders, impose a significant burden on individuals and society. Despite their high prevalence and substantial morbidity and mortality rates, treatment options for these disorders remain limited. Over the past two decades, there has been a gradual accumulation of evidence pointing to the sigma-1 receptor (S1R) system as a promising target for therapeutic interventions designed to treat these disorders. S1R is a chaperone protein that resides in the endoplasmic reticulum, but under certain conditions translocates to the plasma membrane. In the brain, S1Rs are expressed in several regions important for reward, and following translocation, they physically associate with several reward-related GPCRs, including dopamine receptors 1 and 2 (D1R and D2R). Psychostimulants, alcohol, as well as palatable foods, all alter expression of S1R in regions important for motivated behavior, and S1R antagonists generally decrease behavioral responses to these rewards. Recent advances in structural modeling have permitted the development of highly-selective S1R antagonists with favorable pharmacokinetic profiles, thus providing a therapeutic avenue for S1R-based medications. Here, we provide an up-to-date overview of work linking S1R with motivated behavior for drugs of abuse and food, as well as evidence supporting the clinical utility of S1R antagonists to reduce their excessive consumption. We also highlight potential challenges associated with targeting the S1R system, including the need for a more comprehensive understanding of the underlying neurobiology and careful consideration of the pharmacological properties of S1R-based drugs.
Collapse
Affiliation(s)
- Liam G. Knowles
- Harpur School of Arts and Sciences, Binghamton University, Vestal, NY, USA
| | - Abanoub J. Armanious
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University and Rutgers Biomedical Health Sciences, Piscataway, NJ, USA
- Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA
| | - Youyi Peng
- Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - William J. Welsh
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers University and Rutgers Biomedical Health Sciences, Piscataway, NJ, USA
| | - Morgan H. James
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University and Rutgers Biomedical Health Sciences, Piscataway, NJ, USA
- Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA
| |
Collapse
|
17
|
Ona G, Reverte I, Rossi GN, Dos Santos RG, Hallak JE, Colomina MT, Bouso JC. Main targets of ibogaine and noribogaine associated with its putative anti-addictive effects: A mechanistic overview. J Psychopharmacol 2023; 37:1190-1200. [PMID: 37937505 DOI: 10.1177/02698811231200882] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
BACKGROUND There is a growing interest in studying ibogaine (IBO) as a potential treatment for substance use disorders (SUDs). However, its clinical use has been hindered for mainly two reasons: First, the lack of randomized, controlled studies informing about its safety and efficacy. And second, IBO's mechanisms of action remain obscure. It has been challenging to elucidate a predominant mechanism of action responsible for its anti-addictive effects. OBJECTIVE To describe the main targets of IBO and its main metabolite, noribogaine (NOR), in relation to their putative anti-addictive effects, reviewing the updated literature available. METHODS A comprehensive search involving MEDLINE and Google Scholar was undertaken, selecting papers published until July 2022. The inclusion criteria were both theoretical and experimental studies about the pharmacology of IBO. Additional publications were identified in the references of the initial papers. RESULTS IBO and its main metabolite, NOR, can modulate several targets associated with SUDs. Instead of identifying key targets, the action of IBO should be understood as a complex modulation of multiple receptor systems, leading to potential synergies. The elucidation of IBO's pharmacology could be enhanced through the application of methodologies rooted in the polypharmacology paradigm. Such approaches possess the capability to describe multifaceted patterns within multi-target drugs. CONCLUSION IBO displays complex effects through multiple targets. The information detailed here should guide future research on both mechanistic and therapeutic studies.
Collapse
Affiliation(s)
- Genís Ona
- International Center for Ethnobotanical Education, Research, and Service (ICEERS), Barcelona, Spain
- Department of Psychology and Research Center for Behavior Assessment (CRAMC), Universitat Rovira i Virgili, Tarragona, Spain
- Medical Anthropology Research Center (MARC), Universitat Rovira i Virgili, Tarragona, Spain
| | - Ingrid Reverte
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), Rome, Italy
| | - Giordano N Rossi
- Department of Neurosciences and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Rafael G Dos Santos
- Department of Neurosciences and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- National Institute for Translational Medicine (INCT-TM), CNPq, Ribeirão Preto (SP), Brazil
| | - Jaime Ec Hallak
- Department of Neurosciences and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- National Institute for Translational Medicine (INCT-TM), CNPq, Ribeirão Preto (SP), Brazil
| | - Maria Teresa Colomina
- Department of Psychology and Research Center for Behavior Assessment (CRAMC), Universitat Rovira i Virgili, Tarragona, Spain
| | - José Carlos Bouso
- International Center for Ethnobotanical Education, Research, and Service (ICEERS), Barcelona, Spain
- Medical Anthropology Research Center (MARC), Universitat Rovira i Virgili, Tarragona, Spain
- Department of Neurosciences and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
18
|
Damiri B, Mousa A, Helou MA. Amphetamine-type stimulant (ATS) use and association with concurrent use of benzodiazepines, barbiturates and THC in the West Bank among male Palestinians: a cross-sectional study. BMJ PUBLIC HEALTH 2023; 1:e000537. [PMID: 40017840 PMCID: PMC11812734 DOI: 10.1136/bmjph-2023-000537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/15/2023] [Indexed: 03/01/2025]
Abstract
ABSTRACT Introduction Amphetamine-type stimulants (ATSs) are becoming a growing concern in the West Bank. Despite the focus on marijuana, statistics show that ATS seizures and clandestine laboratories indicate increased use of this highly addictive chemical among Palestinians. Shifting from marijuana to ATS can have severe clinical and social consequences, yet insufficient studies have been conducted. Our study aimed to evaluate the prevalence of ATS use and its correlation with other drug use in male Palestinians living in the northern West Bank. Methods A cross-sectional study was conducted among Palestinian men aged ≥18 years from the north of the West Bank in 2022-2023. A self-administered questionnaire and urine analysis test were used to determine drug use. ATS users in this study tested positive for at least one of the following: amphetamines, ecstasy or methamphetamines. Results Out of 787 participants, 129 (16.4%) were drug users. The prevalence of drug use among drug users was as follows: benzodiazepines (55.8%), amphetamines (41.1%), tetrahydrocannabinol (THC) (38.8%), methamphetamines (38.0%), barbiturates (9.3%), phencyclidine (1.6%) and ATS (48.8%); 84.1% were amphetamine and 77.7% were methamphetamine users. Most ATS users (68.3%) were polydrug users; 57.1% were benzodiazepine, 30.2% were THC and 12.7% were barbiturate users. The adjusted binary logistic regression revealed that ATS users were more likely to be benzodiazepine (OR=11.059, p<0.001), THC (OR=5.520, p<0.001) and barbiturate users (OR=17.109, p=0.01). Conclusion The study found that ATS use is widespread among Palestinian men and is associated with other drug use, including benzodiazepines, barbiturates and THC. The study recommends implementing better strategies and policies for controlling and confiscating illicit substances to address the severe medical and sociological problems that can arise from drug dependence. Additionally, the study highlights the risks associated with transitioning from marijuana to ATS, which can have severe clinical and social consequences.
Collapse
Affiliation(s)
- Basma Damiri
- Drugs and Toxicology, An-Najah National University Faculty of Medicine and Health Sciences, Nablus, Palestine, State of
| | - Adnan Mousa
- Medicine, An-Najah National University Faculty of Medicine and Health Sciences, Nablus, Palestine, State of
| | - Maroun Amin Helou
- Medicine, An-Najah National University Faculty of Medicine and Health Sciences, Nablus, Palestine, State of
| |
Collapse
|
19
|
Berkowitz RL, Bluhm AP, Knox GW, McCurdy CR, Ostrov DA, Norris MH. Sigma Receptor Ligands Prevent COVID Mortality In Vivo: Implications for Future Therapeutics. Int J Mol Sci 2023; 24:15718. [PMID: 37958703 PMCID: PMC10647780 DOI: 10.3390/ijms242115718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/26/2023] [Accepted: 10/28/2023] [Indexed: 11/15/2023] Open
Abstract
The emergence of lethal coronaviruses follows a periodic pattern which suggests a recurring cycle of outbreaks. It remains uncertain as to when the next lethal coronavirus will emerge, though its eventual emergence appears to be inevitable. New mutations in evolving SARS-CoV-2 variants have provided resistance to current antiviral drugs, monoclonal antibodies, and vaccines, reducing their therapeutic efficacy. This underscores the urgent need to investigate alternative therapeutic approaches. Sigma receptors have been unexpectedly linked to the SARS-CoV-2 life cycle due to the direct antiviral effect of their ligands. Coronavirus-induced cell stress facilitates the formation of an ER-derived complex conducive to its replication. Sigma receptor ligands are believed to prevent the formation of this complex. Repurposing FDA-approved drugs for COVID-19 offers a timely and cost-efficient strategy to find treatments with established safety profiles. Notably, diphenhydramine, a sigma receptor ligand, is thought to counteract the virus by inhibiting the creation of ER-derived replication vesicles. Furthermore, lactoferrin, a well-characterized immunomodulatory protein, has shown antiviral efficacy against SARS-CoV-2 both in laboratory settings and in living organisms. In the present study, we aimed to explore the impact of sigma receptor ligands on SARS-CoV-2-induced mortality in ACE2-transgenic mice. We assessed the effects of an investigational antiviral drug combination comprising a sigma receptor ligand and an immunomodulatory protein. Mice treated with sigma-2 receptor ligands or diphenhydramine and lactoferrin exhibited improved survival rates and rapid rebound in mass following the SARS-CoV-2 challenge compared to mock-treated animals. Clinical translation of these findings may support the discovery of new treatment and research strategies for SARS-CoV-2.
Collapse
Affiliation(s)
- Reed L. Berkowitz
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (R.L.B.); (D.A.O.)
| | - Andrew P. Bluhm
- Spatial Epidemiology and Ecology Research Laboratory, Department of Geography, College of Liberal Arts and Sciences, University of Florida, Gainesville, FL 32611, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, FL 32601, USA
| | - Glenn W. Knox
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (R.L.B.); (D.A.O.)
| | - Christopher R. McCurdy
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
- Translational Drug Development Core, Clinical and Translational Sciences Institute, University of Florida, Gainesville, FL 32610, USA
| | - David A. Ostrov
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (R.L.B.); (D.A.O.)
| | - Michael H. Norris
- Emerging Pathogens Institute, University of Florida, Gainesville, FL 32601, USA
- School of Life Sciences, University of Hawaiʻi at Mānoa, Honolulu, HI 96822, USA
| |
Collapse
|
20
|
Davis SE, Cirincione AB, Jimenez-Torres AC, Zhu J. The Impact of Neurotransmitters on the Neurobiology of Neurodegenerative Diseases. Int J Mol Sci 2023; 24:15340. [PMID: 37895020 PMCID: PMC10607327 DOI: 10.3390/ijms242015340] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Neurodegenerative diseases affect millions of people worldwide. Neurodegenerative diseases result from progressive damage to nerve cells in the brain or peripheral nervous system connections that are essential for cognition, coordination, strength, sensation, and mobility. Dysfunction of these brain and nerve functions is associated with Alzheimer's disease, Parkinson's disease, Huntington's disease, Amyotrophic lateral sclerosis, and motor neuron disease. In addition to these, 50% of people living with HIV develop a spectrum of cognitive, motor, and/or mood problems collectively referred to as HIV-Associated Neurocognitive Disorders (HAND) despite the widespread use of a combination of antiretroviral therapies. Neuroinflammation and neurotransmitter systems have a pathological correlation and play a critical role in developing neurodegenerative diseases. Each of these diseases has a unique pattern of dysregulation of the neurotransmitter system, which has been attributed to different forms of cell-specific neuronal loss. In this review, we will focus on a discussion of the regulation of dopaminergic and cholinergic systems, which are more commonly disturbed in neurodegenerative disorders. Additionally, we will provide evidence for the hypothesis that disturbances in neurotransmission contribute to the neuronal loss observed in neurodegenerative disorders. Further, we will highlight the critical role of dopamine as a mediator of neuronal injury and loss in the context of NeuroHIV. This review will highlight the need to further investigate neurotransmission systems for their role in the etiology of neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | | | - Jun Zhu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA; (S.E.D.); (A.B.C.); (A.C.J.-T.)
| |
Collapse
|
21
|
Shi TS, Li WY, Chen YM, Huang J, Guan W, Xu DW, Jiang B. The antidepressant-like effects of escitalopram in mice require salt-inducible kinase 1 and CREB-regulated transcription co-activator 1 in the paraventricular nucleus of the hypothalamus. J Affect Disord 2023; 338:228-238. [PMID: 37257779 DOI: 10.1016/j.jad.2023.05.085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023]
Abstract
BACKGROUND The salt-inducible kinase 1 (SIK1)-CREB-regulated transcription co-activator 1 (CRTC1) system in the paraventricular nucleus (PVN) of the hypothalamus has been demonstrated to participate in not only depression neurobiology but also the antidepressant mechanisms of fluoxetine, paroxetine, venlafaxine, and duloxetine. Like fluoxetine and paroxetine, escitalopram is also a well-known selective serotonin (5-HT) reuptake inhibitor (SSRI). However, recently it has been found that escitalopram can modulate a lot of targets other than the 5-HT system. Here, we speculate that escitalopram produces effects on the SIK1-CRTC1 system in the PVN. METHODS Two mice models of depression (chronic social defeat stress (CSDS) and chronic unpredictable mild stress (CUMS)), various behavioral tests, enzyme linked immunosorbent assay (ELISA), western blotting, co-immunoprecipitation (Co-IP), quantitative real-time reverse transcription PCR (qRT-PCR), immunofluorescence, and adeno-associated virus (AAV)-mediated gene transfer were used together in the present study. RESULTS It was found that escitalopram administration not only significantly prevented the hyperactivity of the hypothalamic-pituitary-adrenal (HPA) axis induced by CSDS and CUMS, but also notably reversed the effects of CSDS and CUMS on SIK1, CRTC1, and CRTC1-CREB binding in the PVN of mice. AAV-based genetic knock-down of SIK1 in PVN neurons evidently abolished the antidepressant-like effects of escitalopram in mice. LIMITATION A shortage of this study is that only rodent models of depression were used, while human samples were not included. CONCLUSIONS In summary, regulating the SIK1-CRTC1 system in the PVN participates in the antidepressant mechanism of escitalopram, which extends the knowledge of the pharmacological actions of escitalopram.
Collapse
Affiliation(s)
- Tian-Shun Shi
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Wei-Yu Li
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Yan-Mei Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Jie Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Wei Guan
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Da-Wei Xu
- Department of Orthopedics, Affiliated Hospital 2 of Nantong University, Nantong 226006, Jiangsu, China.
| | - Bo Jiang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China.
| |
Collapse
|
22
|
Jahanabadi S, Amiri S, Karkeh-Abadi M, Razmi A. Natural psychedelics in the treatment of depression; a review focusing on neurotransmitters. Fitoterapia 2023; 169:105620. [PMID: 37490982 DOI: 10.1016/j.fitote.2023.105620] [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: 04/04/2023] [Revised: 07/21/2023] [Accepted: 07/22/2023] [Indexed: 07/27/2023]
Abstract
Natural psychedelic compounds are emerging as potential novel therapeutics in psychiatry. This review will discuss how natural psychedelics exert their neurobiological therapeutic effects, and how different neurotransmission systems mediate the effects of these compounds. Further, current therapeutic strategies for depression, and novel mechanism of action of natural psychedelics in the treatment of depression will be discussed. In this review, our focus will be on N, N-dimethyltryptamine (DMT), reversible type A monoamine oxidase inhibitors, mescaline-containing cacti, psilocybin/psilocin-containing mushrooms, ibogaine, muscimol extracted from Amanita spp. mushrooms and ibotenic acid.
Collapse
Affiliation(s)
- Samane Jahanabadi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Pharmaceutical Science Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Shayan Amiri
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada; Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada.
| | - Mehdi Karkeh-Abadi
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Ali Razmi
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran.
| |
Collapse
|
23
|
Rafcikova J, Novakova M, Stracina T. Exploring the Association between Schizophrenia and Cardiovascular Diseases: Insights into the Role of Sigma 1 Receptor. Physiol Res 2023; 72:S113-S126. [PMID: 37565416 PMCID: PMC10660581 DOI: 10.33549/physiolres.935099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/15/2023] [Indexed: 12/01/2023] Open
Abstract
Contemporary society is characterized by rapid changes. Various epidemiological, political and economic crises represent a burden to mental health of nowadays population, which may at least partially explain the increasing incidence of mental disorders, including schizophrenia. Schizophrenia is associated with premature mortality by at least 13-15 years. The leading cause of premature mortality in schizophrenia patients is high incidence of cardiovascular diseases. The specific-cause mortality risk for cardiovascular diseases in schizophrenia patients is more than twice higher as compared to the general population. Several factors are discussed as the factor of cardiovascular diseases development. Intensive efforts to identify possible link between schizophrenia and cardiovascular diseases are made. It seems that sigma 1 receptor may represent such link. By modulation of the activity of several neurotransmitter systems, including dopamine, glutamate, and GABA, sigma 1 receptor might play a role in pathophysiology of schizophrenia. Moreover, significant roles of sigma 1 receptor in cardiovascular system have been repeatedly reported. The detailed role of sigma 1 receptor in both schizophrenia and cardiovascular disorders development however remains unclear. The article presents an overview of current knowledge about the association between schizophrenia and cardiovascular diseases and proposes possible explanations with special emphasis on the role of the sigma 1 receptor.
Collapse
Affiliation(s)
- J Rafcikova
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| | | | | |
Collapse
|
24
|
Corsi S, Scheggi S, Pardu A, Braccagni G, Caruso D, Cioffi L, Diviccaro S, Gentile M, Fanni S, Stancampiano R, Gambarana C, Melcangi RC, Frau R, Carta M. Pregnenolone for the treatment of L-DOPA-induced dyskinesia in Parkinson's disease. Exp Neurol 2023; 363:114370. [PMID: 36878398 DOI: 10.1016/j.expneurol.2023.114370] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 02/02/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023]
Abstract
Growing preclinical and clinical evidence highlights neurosteroid pathway imbalances in Parkinson's Disease (PD) and L-DOPA-induced dyskinesias (LIDs). We recently reported that 5α-reductase (5AR) inhibitors dampen dyskinesias in parkinsonian rats; however, unraveling which specific neurosteroid mediates this effect is critical to optimize a targeted therapy. Among the 5AR-related neurosteroids, striatal pregnenolone has been shown to be increased in response to 5AR blockade and decreased after 6-OHDA lesions in the rat PD model. Moreover, this neurosteroid rescued psychotic-like phenotypes by exerting marked antidopaminergic activity. In light of this evidence, we investigated whether pregnenolone might dampen the appearance of LIDs in parkinsonian drug-naïve rats. We tested 3 escalating doses of pregnenolone (6, 18, 36 mg/kg) in 6-OHDA-lesioned male rats and compared the behavioral, neurochemical, and molecular outcomes with those induced by the 5AR inhibitor dutasteride, as positive control. The results showed that pregnenolone dose-dependently countered LIDs without affecting L-DOPA-induced motor improvements. Post-mortem analyses revealed that pregnenolone significantly prevented the increase of validated striatal markers of dyskinesias, such as phospho-Thr-34 DARPP-32 and phospho-ERK1/2, as well as D1-D3 receptor co-immunoprecipitation in a fashion similar to dutasteride. Moreover, the antidyskinetic effect of pregnenolone was paralleled by reduced striatal levels of BDNF, a well-established factor associated with the development of LIDs. In support of a direct pregnenolone effect, LC/MS-MS analyses revealed that striatal pregnenolone levels strikingly increased after the exogenous administration, with no significant alterations in downstream metabolites. All these data suggest pregnenolone as a key player in the antidyskinetic properties of 5AR inhibitors and highlight this neurosteroid as an interesting novel tool to target LIDs in PD.
Collapse
Affiliation(s)
- Sara Corsi
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy
| | - Simona Scheggi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, SI, Italy
| | - Alessandra Pardu
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy
| | - Giulia Braccagni
- Department of Molecular and Developmental Medicine, University of Siena, Siena, SI, Italy
| | - Donatella Caruso
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, MI, Italy
| | - Lucia Cioffi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, MI, Italy
| | - Silvia Diviccaro
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, MI, Italy
| | - Mauro Gentile
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy
| | - Silvia Fanni
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy; Basal Ganglia Pathophysiology Unit, Department Experimental Medical Science, Lund University, Sweden
| | | | - Carla Gambarana
- Department of Molecular and Developmental Medicine, University of Siena, Siena, SI, Italy
| | - Roberto Cosimo Melcangi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, MI, Italy
| | - Roberto Frau
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy; "Guy Everett Laboratory", Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy.
| | - Manolo Carta
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy.
| |
Collapse
|
25
|
VanderZwaag J, Halvorson T, Dolhan K, Šimončičová E, Ben-Azu B, Tremblay MÈ. The Missing Piece? A Case for Microglia's Prominent Role in the Therapeutic Action of Anesthetics, Ketamine, and Psychedelics. Neurochem Res 2023; 48:1129-1166. [PMID: 36327017 DOI: 10.1007/s11064-022-03772-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 08/25/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022]
Abstract
There is much excitement surrounding recent research of promising, mechanistically novel psychotherapeutics - psychedelic, anesthetic, and dissociative agents - as they have demonstrated surprising efficacy in treating central nervous system (CNS) disorders, such as mood disorders and addiction. However, the mechanisms by which these drugs provide such profound psychological benefits are still to be fully elucidated. Microglia, the CNS's resident innate immune cells, are emerging as a cellular target for psychiatric disorders because of their critical role in regulating neuroplasticity and the inflammatory environment of the brain. The following paper is a review of recent literature surrounding these neuropharmacological therapies and their demonstrated or hypothesized interactions with microglia. Through investigating the mechanism of action of psychedelics, such as psilocybin and lysergic acid diethylamide, ketamine, and propofol, we demonstrate a largely under-investigated role for microglia in much of the emerging research surrounding these pharmacological agents. Among others, we detail sigma-1 receptors, serotonergic and γ-aminobutyric acid signalling, and tryptophan metabolism as pathways through which these agents modulate microglial phagocytic activity and inflammatory mediator release, inducing their therapeutic effects. The current review includes a discussion on future directions in the field of microglial pharmacology and covers bidirectional implications of microglia and these novel pharmacological agents in aging and age-related disease, glial cell heterogeneity, and state-of-the-art methodologies in microglial research.
Collapse
Affiliation(s)
- Jared VanderZwaag
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Torin Halvorson
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Kira Dolhan
- Department of Psychology, University of Victoria, Vancouver, BC, Canada
- Department of Biology, University of Victoria, Vancouver, BC, Canada
| | - Eva Šimončičová
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Benneth Ben-Azu
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Marie-Ève Tremblay
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada.
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Département de médecine moléculaire, Université Laval, Québec City, QC, Canada.
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada.
- Neurology and Neurosurgery Department, McGill University, Montreal, QC, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
- Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
26
|
Piechal A, Jakimiuk A, Pyrzanowska J, Blecharz-Klin K, Joniec-Maciejak I, Wiercińska-Drapało A, Mirowska-Guzel D, Widy-Tyszkiewicz E. Long-term Administration of 3-Di-O-Tolylguanidine Modulates Spatial Learning and Memory in Rats and Causes Transition in the Concentration of Neurotransmitters in the Hippocampus, Prefrontal Cortex and Striatum. Neuroscience 2023; 510:129-146. [PMID: 36493909 DOI: 10.1016/j.neuroscience.2022.11.036] [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: 07/31/2022] [Revised: 11/14/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
The sigma-1 and sigma-2 (σ1 and σ2) receptors are found in high concentrations in the brain, and their altered expression leads to a variety of neuropsychiatric disorders. 3-di-tolylguanidine (DTG) stimulates the activity of both of these receptors. We assessed the effects of administering DTG to adult male Sprague Dawley rats on learning and memory consolidation processes and on the levels of neurotransmitters in selected brain structures. Spatial learning and memory were evaluated in the water maze test. The DTG was administered orally at daily doses of 3 mg/kg (DTG3), 10 mg/kg (DTG10) or 30 mg/kg (DTG30) for 10 weeks before and during the water-maze test. After completion of the experiment, the concentration of monoamines and their metabolites as well as amino acids in structures involved in cognitive performance - the hippocampus, prefrontal cortex, and striatum - were determined using high performance liquid chromatography (HPLC). The DTG10 group showed an improvement in memory processes related to the "new" platform location, whereas the DTG30 group was worse at finding the "old" platform location. Since the administration of DTG led to differences in dopaminergic transmission, it was assumed to influence memory processes in this way. Changes in histidine, serine, alanine, taurine, and glutamic acid levels in selected structures of the brains of rats with memory impairment were also observed. We conclude that long-term administration of DTG modulates spatial learning and memory in rats and changes the concentrations of neurotransmitters in the hippocampus, prefrontal cortex, and striatum..
Collapse
Affiliation(s)
- Agnieszka Piechal
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1B, 02-097 Warsaw, Poland
| | - Alicja Jakimiuk
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1B, 02-097 Warsaw, Poland
| | - Justyna Pyrzanowska
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1B, 02-097 Warsaw, Poland.
| | - Kamilla Blecharz-Klin
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1B, 02-097 Warsaw, Poland
| | - Ilona Joniec-Maciejak
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1B, 02-097 Warsaw, Poland
| | - Alicja Wiercińska-Drapało
- Department of Infectious and Tropical Diseases and Hepatology, Medical University of Warsaw, Wolska 37, 01-201 Warsaw, Poland
| | - Dagmara Mirowska-Guzel
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1B, 02-097 Warsaw, Poland
| | - Ewa Widy-Tyszkiewicz
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Centre for Preclinical Research and Technology CePT, Banacha 1B, 02-097 Warsaw, Poland
| |
Collapse
|
27
|
Frau R, Melis M. Sex-specific susceptibility to psychotic-like states provoked by prenatal THC exposure: Reversal by pregnenolone. J Neuroendocrinol 2023; 35:e13240. [PMID: 36810840 DOI: 10.1111/jne.13240] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 01/23/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023]
Abstract
Sociocultural attitudes towards cannabis legalization contribute to the common misconception that it is a relatively safe drug and its use during pregnancy poses no risk to the fetus. However, longitudinal studies demonstrate that maternal cannabis exposure results in adverse outcomes in the offspring, with a heightened risk for developing psychopathology. One of the most reported psychiatric outcomes is the proneness to psychotic-like experiences during childhood. How exposure to cannabis during gestation increases psychosis susceptibility in children and adolescents remains elusive. Preclinical research has indicated that in utero exposure to the major psychoactive component of cannabis, delta-9-tetrahydrocannabinol (THC), deranges brain developmental trajectories towards vulnerable psychotic-like endophenotypes later in life. Here, we present how prenatal THC exposure (PCE) deregulates mesolimbic dopamine development predisposing the offspring to schizophrenia-relevant phenotypes, exclusively when exposed to environmental challenges, such as stress or THC. Detrimental effects of PCE are sex-specific because female offspring do not display psychotic-like outcomes upon exposure to these challenges. Moreover, we present how pregnenolone, a neurosteroid that showed beneficial properties on the effects elicited by cannabis intoxication, normalizes mesolimbic dopamine function and rescues psychotic-like phenotypes. We, therefore, suggest this neurosteroid as a safe "disease-modifying" aid to prevent the onset of psychoses in vulnerable individuals. Our findings corroborate clinical evidence and highlight the relevance of early diagnostic screening and preventative strategies for young individuals at risk for mental diseases, such as male PCE offspring.
Collapse
Affiliation(s)
- Roberto Frau
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato, Italy
- The Guy Everett Laboratory for Neuroscience, University of Cagliari, Cagliari, Italy
| | - Miriam Melis
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato, Italy
| |
Collapse
|
28
|
Liang M, Chen G, Xi Z, Qian H, Shang Q, Gao B, An R, Shao G, Wang Z, Wang J, Xiao J, Li T, Liu X. The roles of K +-dependent Na +/Ca 2+ exchanger 2 (NCKX2) in methamphetamine-induced behavioral sensitization and conditioned place preference in mice. Neurosci Lett 2023; 792:136952. [PMID: 36336087 DOI: 10.1016/j.neulet.2022.136952] [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: 02/13/2022] [Revised: 08/27/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
Drug addiction, including methamphetamine (METH) addiction, is a significant public health and social issue. Perturbations in intracellular Ca2+ homeostasis are associated with drug addiction. K+-dependent Na+/Ca2+ exchanger 2 (NCKX2) is located on neuronal cell membranes and constitutes a Ca2+ clearance mechanism, with key roles in synaptic plasticity. NCKX2 is associated with motor learning, memory, and cognitive functions. However, the role of NCKX2 in METH addiction remains unclear. In this study, we investigated the expression levels of NCKX2 in four addiction-related brain regions: the prefrontal cortex (PFc), nucleus accumbens (NAc), dorsal striatum (DS), and hippocampus (Hip) in a C57/BL6 mouse model of METH-induced conditioned place preference (CPP) and behavioral sensitization. Levels of NCKX2 were unchanged in these brain regions in mice with METH-induced CPP but were decreased in the PFc and NAc of mice with METH-induced behavioral sensitization. Adeno-associated virus (AAV)-mediated overexpression of NCKX2 in the PFc attenuated the expression phase of METH-induced behavioral sensitization in mice, whereas AAV-mediated knockdown of NCKX2 enhanced the effects of METH. Collectively, our results suggest that NCKX2 is involved in METH-induced behavioral sensitization but does not affect conditioned reward-related memory, highlighting the potential of NCKX2 as a molecular target for studying the mechanisms underscoring METH addiction.
Collapse
Affiliation(s)
- Min Liang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Gang Chen
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China; Department of Forensic Medicine, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China.
| | - Zhijia Xi
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Hongyan Qian
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Qing Shang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Baoyao Gao
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Ran An
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Gaojie Shao
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Zhirong Wang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Jing Wang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Jing Xiao
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Tao Li
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Xinshe Liu
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China.
| |
Collapse
|
29
|
Machado da Silva MC, Iglesias LP, Candelario-Jalil E, Khoshbouei H, Moreira FA, de Oliveira ACP. Role of Microglia in Psychostimulant Addiction. Curr Neuropharmacol 2023; 21:235-259. [PMID: 36503452 PMCID: PMC10190137 DOI: 10.2174/1570159x21666221208142151] [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: 05/13/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 12/14/2022] Open
Abstract
The use of psychostimulant drugs can modify brain function by inducing changes in the reward system, mainly due to alterations in dopaminergic and glutamatergic transmissions in the mesocorticolimbic pathway. However, the etiopathogenesis of addiction is a much more complex process. Previous data have suggested that microglia and other immune cells are involved in events associated with neuroplasticity and memory, which are phenomena that also occur in addiction. Nevertheless, how dependent is the development of addiction on the activity of these cells? Although the mechanisms are not known, some pathways may be involved. Recent data have shown psychoactive substances may act directly on immune cells, alter their functions and induce various inflammatory mediators that modulate synaptic activity. These could, in turn, be involved in the pathological alterations that occur in substance use disorder. Here, we extensively review the studies demonstrating how cocaine and amphetamines modulate microglial number, morphology, and function. We also describe the effect of these substances in the production of inflammatory mediators and a possible involvement of some molecular signaling pathways, such as the toll-like receptor 4. Although the literature in this field is scarce, this review compiles the knowledge on the neuroimmune axis that is involved in the pathogenesis of addiction, and suggests some pharmacological targets for the development of pharmacotherapy.
Collapse
Affiliation(s)
- Maria Carolina Machado da Silva
- Department of Pharmacology, Neuropharmacology Laboratory, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil;
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Lia Parada Iglesias
- Department of Pharmacology, Neuropsychopharmacology Laboratory, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Habibeh Khoshbouei
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Fabrício Araujo Moreira
- Department of Pharmacology, Neuropsychopharmacology Laboratory, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | |
Collapse
|
30
|
Colwell MJ, Tagomori H, Chapman S, Gillespie AL, Cowen PJ, Harmer CJ, Murphy SE. Pharmacological targeting of cognitive impairment in depression: recent developments and challenges in human clinical research. Transl Psychiatry 2022; 12:484. [PMID: 36396622 PMCID: PMC9671959 DOI: 10.1038/s41398-022-02249-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 11/18/2022] Open
Abstract
Impaired cognition is often overlooked in the clinical management of depression, despite its association with poor psychosocial functioning and reduced clinical engagement. There is an outstanding need for new treatments to address this unmet clinical need, highlighted by our consultations with individuals with lived experience of depression. Here we consider the evidence to support different pharmacological approaches for the treatment of impaired cognition in individuals with depression, including treatments that influence primary neurotransmission directly as well as novel targets such as neurosteroid modulation. We also consider potential methodological challenges in establishing a strong evidence base in this area, including the need to disentangle direct effects of treatment on cognition from more generalised symptomatic improvement and the identification of sensitive, reliable and objective measures of cognition.
Collapse
Affiliation(s)
- Michael J Colwell
- University Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Hosana Tagomori
- University Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Sarah Chapman
- University Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
| | - Amy L Gillespie
- University Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Philip J Cowen
- University Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Catherine J Harmer
- University Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Susannah E Murphy
- University Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK.
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK.
| |
Collapse
|
31
|
Evaluation of Adenosine A2A receptor gene polymorphisms as risk factors of methamphetamine use disorder susceptibility and predictors of craving degree. Psychiatry Res 2022; 316:114790. [PMID: 35987070 DOI: 10.1016/j.psychres.2022.114790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/01/2022] [Accepted: 08/12/2022] [Indexed: 11/24/2022]
Abstract
The adenosine A2A receptor (ADORA2A) is highly expressed in the central nervous system and plays vital roles in drug addiction. In this study, we aimed to explore the susceptibility of ADORA2A to methamphetamine use disorder (MUD) and the craving degree based on a two-stage association analysis. A total of 3,542 (1,216 patients with MUD and 2,326 controls) and 1,740 participants (580 patients with MUD and 1,160 controls) were recruited in discovery and replication stage, respectively. Significant SNPs identified in the discovery stage were genotyped in the replication samples. Serum levels of ADORA2A were measured using enzyme-linked immunosorbent assay kits. The genetic association signal of each SNP was examined using Plink. A linear model was fitted to investigate the relationship between craving scores and genotypes of significant SNPs. SNP rs5751876 was significantly associated with MUD in the discovery samples and this association signal was then further replicated in the replication samples. Significant associations were also identified between serum levels of ADORA2A and the genotypes of rs5751876 (P = 0.0002). The craving scores in patients with MUD were strongly correlated with rs5751876 genotypes. Our results suggest that polymorphisms of the ADORA2A gene could affect the susceptibility to MUD and its craving degree.
Collapse
|
32
|
Marrero-Cristobal G, Gelpi-Dominguez U, Morales-Silva R, Alvarado-Torres J, Perez-Torres J, Perez-Perez Y, Sepulveda-Orengo M. Aerobic exercise as a promising nonpharmacological therapy for the treatment of substance use disorders. J Neurosci Res 2022; 100:1602-1642. [PMID: 34850988 PMCID: PMC9156662 DOI: 10.1002/jnr.24990] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 10/06/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022]
Abstract
Despite the prevalence and public health impact of substance use disorders (SUDs), effective long-term treatments remain elusive. Aerobic exercise is a promising, nonpharmacological treatment currently under investigation as a strategy for preventing drug relapse. Aerobic exercise could be incorporated into the comprehensive treatment regimens for people with substance abuse disorders. Preclinical studies of SUD with animal models have shown that aerobic exercise diminishes drug-seeking behavior, which leads to relapse, in both male and female rats. Nevertheless, little is known regarding the effects of substance abuse-induced cellular and physiological adaptations believed to be responsible for drug-seeking behavior. Accordingly, the overall goal of this review is to provide a summary and an assessment of findings to date, highlighting evidence of the molecular and neurological effects of exercise on adaptations associated with SUD.
Collapse
Affiliation(s)
| | - Ursula Gelpi-Dominguez
- School of Behavioral and Brain Sciences, Ponce Health Sciences University, Ponce, PR, USA
| | - Roberto Morales-Silva
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR, USA
| | - John Alvarado-Torres
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR, USA
| | - Joshua Perez-Torres
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR, USA
| | - Yobet Perez-Perez
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR, USA
| | - Marian Sepulveda-Orengo
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR, USA
| |
Collapse
|
33
|
Xu Z, Lei Y, Qin H, Zhang S, Li P, Yao K. Sigma-1 Receptor in Retina: Neuroprotective Effects and Potential Mechanisms. Int J Mol Sci 2022; 23:7572. [PMID: 35886921 PMCID: PMC9321618 DOI: 10.3390/ijms23147572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/03/2022] [Accepted: 07/05/2022] [Indexed: 02/04/2023] Open
Abstract
Retinal degenerative diseases are the major factors leading to severe visual impairment and even irreversible blindness worldwide. The therapeutic approach for retinal degenerative diseases is one extremely urgent and hot spot in science research. The sigma-1 receptor is a novel, multifunctional ligand-mediated molecular chaperone residing in endoplasmic reticulum (ER) membranes and the ER-associated mitochondrial membrane (ER-MAM); it is widely distributed in numerous organs and tissues of various species, providing protective effects on a variety of degenerative diseases. Over three decades, considerable research has manifested the neuroprotective function of sigma-1 receptor in the retina and has attempted to explore the molecular mechanism of action. In the present review, we will discuss neuroprotective effects of the sigma-1 receptor in retinal degenerative diseases, mainly in aspects of the following: the localization in different types of retinal neurons, the interactions of sigma-1 receptors with other molecules, the correlated signaling pathways, the influence of sigma-1 receptors to cellular functions, and the potential therapeutic effects on retinal degenerative diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Kai Yao
- Institute of Visual Neuroscience and Stem Cell Engineering, College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China; (Z.X.); (Y.L.); (H.Q.); (S.Z.); (P.L.)
| |
Collapse
|
34
|
Ren P, Wang J, Li N, Li G, Ma H, Zhao Y, Li Y. Sigma-1 Receptors in Depression: Mechanism and Therapeutic Development. Front Pharmacol 2022; 13:925879. [PMID: 35784746 PMCID: PMC9243434 DOI: 10.3389/fphar.2022.925879] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/26/2022] [Indexed: 12/26/2022] Open
Abstract
Depression is the most common type of neuropsychiatric illness and has increasingly become a major cause of disability. Unfortunately, the recent global pandemic of COVID-19 has dramatically increased the incidence of depression and has significantly increased the burden of mental health care worldwide. Since full remission of the clinical symptoms of depression has not been achieved with current treatments, there is a constant need to discover new compounds that meet the major clinical needs. Recently, the roles of sigma receptors, especially the sigma-1 receptor subtype, have attracted increasing attention as potential new targets and target-specific drugs due to their translocation property that produces a broad spectrum of biological functions. Even clinical first-line antidepressants with or without affinity for sigma-1 receptors have different pharmacological profiles. Thus, the regulatory role of sigma-1 receptors might be useful in treating these central nervous system (CNS) diseases. In addition, long-term mental stress disrupts the homeostasis in the CNS. In this review, we discuss the topical literature concerning sigma-1 receptor antidepressant mechanism of action in the regulation of intracellular proteostasis, calcium homeostasis and especially the dynamic Excitatory/Inhibitory (E/I) balance in the brain. Furthermore, based on these discoveries, we discuss sigma-1 receptor ligands with respect to their promise as targets for fast-onset action drugs in treating depression.
Collapse
Affiliation(s)
- Peng Ren
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jingya Wang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Nanxi Li
- Department of Pharmaceutical Sciences, Beijng Institute of Radiation Medicine, Beijing, China
| | - Guangxiang Li
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Hui Ma
- Beijing Institute of Basic Medical Sciences, Beijing, China
- *Correspondence: Hui Ma, ; Yongqi Zhao, ; Yunfeng Li,
| | - Yongqi Zhao
- Beijing Institute of Basic Medical Sciences, Beijing, China
- *Correspondence: Hui Ma, ; Yongqi Zhao, ; Yunfeng Li,
| | - Yunfeng Li
- Beijing Institute of Basic Medical Sciences, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
- *Correspondence: Hui Ma, ; Yongqi Zhao, ; Yunfeng Li,
| |
Collapse
|
35
|
Gursoy M, Gul Z, Buyukuysal RL. Sigma receptor ligands haloperidol and ifenprodil attenuate hypoxia induced dopamine release in rat striatum. Neurol Res 2022; 44:927-936. [DOI: 10.1080/01616412.2022.2072100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Murat Gursoy
- Bursa Provincial Health Directorate, T.C. Ministry of Health, Bursa, Turkey
| | - Zulfiye Gul
- Department of Medical Pharmacology, Faculty of Medicine, Bahcesehir University, Istanbul, Turkey
| | - R. Levent Buyukuysal
- Department of Medical Pharmacology, Faculty of Medicine, Uludag University, Bursa, Turkey
| |
Collapse
|
36
|
Shen B, Zhang D, Zeng X, Guan L, Yang G, Liu L, Huang J, Li Y, Hong S, Li L. Cannabidiol inhibits methamphetamine-induced dopamine release via modulation of the DRD1-MeCP2-BDNF-TrkB signaling pathway. Psychopharmacology (Berl) 2022; 239:1521-1537. [PMID: 34997862 DOI: 10.1007/s00213-021-06051-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 12/20/2021] [Indexed: 12/17/2022]
Abstract
RATIONALE Adaptive alteration of dopamine (DA) system in mesocorticolimbic circuits is an extremely intricate and dynamic process, which contributes to maintaining methamphetamine (METH)-related disorders. There are no approved pharmacotherapies for METH-related disorders. Cannabidiol (CBD), a major non-psychoactive constituent of cannabis, has received attention for its therapeutic potential in treating METH-related disorders. However, the major research obstacles of CBD are the yet to be clarified mechanisms behind its therapeutic potential. Recent evidence showed that DA system may be active target of CBD. CBD could be a promising dopaminergic medication for METH-related disorders. OBJECTIVES We investigated the role of the DA receptor D1 (DRD1)-methyl-CpG-binding protein 2 (MeCP2)-brain-derived neurotrophic factor (BDNF)-tyrosine receptor kinase B (TrkB) signaling pathway in DA release induced by METH. Investigating the intervention effects of CBD on the DRD1-MeCP2-BDNF-TrkB signaling pathway could help clarify the underlying mechanisms and therapeutic potential of CBD in METH-related disorders. RESULTS METH (400 μM) significantly increased DA release from primary neurons in vitro, which was blocked by CBD (1 μM) pretreatment. METH (400 μM) significantly increased the expression levels of DRD1, BDNF, and TrkB, but decreased the expression of MeCP2 in the neurons, whereas CBD (1 μM) pretreatment notably inhibited the protein changes induced by METH. In addition, DRD1 antagonist SCH23390 (10 μM) inhibited the DA release and protein change induced by METH in vitro. However, DRD1 agonist SKF81297 (10 μM) induced DA release and protein change in vitro, which was also blocked by CBD (1 μM) pretreatment. METH (2 mg/kg) significantly increased the DA level in the nucleus accumbens (NAc) of rats with activation of the DRD1-MeCP2-BDNF-TrkB signaling pathway, but these changes were blocked by CBD (40 or 80 mg/kg) pretreatment. CONCLUSIONS This study indicates that METH induces DA release via the DRD1-MeCP2-BDNF-TrkB signaling pathway. Furthermore, CBD significantly inhibits DA release induced by METH through modulation of this pathway.
Collapse
Affiliation(s)
- Baoyu Shen
- School of Forensic Medicine, Key Laboratory of Drug Addiction Medicine of National Health Commission (NHC), Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Dongxian Zhang
- School of Forensic Medicine, Key Laboratory of Drug Addiction Medicine of National Health Commission (NHC), Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Xiaofeng Zeng
- School of Forensic Medicine, Key Laboratory of Drug Addiction Medicine of National Health Commission (NHC), Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Lina Guan
- School of Forensic Medicine, Key Laboratory of Drug Addiction Medicine of National Health Commission (NHC), Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Genmeng Yang
- School of Forensic Medicine, Key Laboratory of Drug Addiction Medicine of National Health Commission (NHC), Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Liu Liu
- School of Forensic Medicine, Key Laboratory of Drug Addiction Medicine of National Health Commission (NHC), Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Jian Huang
- School of Forensic Medicine, Key Laboratory of Drug Addiction Medicine of National Health Commission (NHC), Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Yuanyuan Li
- School of Forensic Medicine, Key Laboratory of Drug Addiction Medicine of National Health Commission (NHC), Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Shijun Hong
- School of Forensic Medicine, Key Laboratory of Drug Addiction Medicine of National Health Commission (NHC), Kunming Medical University, Kunming, 650500, Yunnan, China.
| | - Lihua Li
- School of Forensic Medicine, Key Laboratory of Drug Addiction Medicine of National Health Commission (NHC), Kunming Medical University, Kunming, 650500, Yunnan, China.
| |
Collapse
|
37
|
Salguero A, Marengo L, Portillo-Salido E, Ruiz-Leyva L, Cendán CM, Morón I, Marcos Pautassi R. Administration of the sigma-1 receptor agonist PRE-084 at emerging adulthood, but not at early adolescence, attenuated ethanol-induced conditioned taste aversion in female rats. Neurosci Lett 2022; 778:136585. [PMID: 35318075 DOI: 10.1016/j.neulet.2022.136585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 03/07/2022] [Accepted: 03/18/2022] [Indexed: 11/17/2022]
Abstract
Ethanol-induced conditioned taste aversion (CTA) is greater in late adolescence or young adulthood than in early adolescence. The role of the sigma receptor system in this age-related difference has not been extensively explored, particularly in female rats. This study assessed the effects of the activation of sigma-1 receptors (S1-R), via the selective S1-R agonist PRE-084, on ethanol-induced CTA at early or at terminal adolescence/emerging adulthood (28 or 56 days-old at the beginning of the procedures, respectively) in female Wistar rats. The modulation of binge-like ethanol intake by PRE-084 was assessed at terminal adolescence. S1-R activation at the acquisition of ethanol-induced CTA attenuated such learning at terminal but not at early adolescence. PRE-084 did not significantly affect ethanol binge drinking in the terminal adolescents. These results highlight the role of S1-R in ethanol-induced CTA and suggest that differential functionality of this transmitter system may underlie age-specific sensitivities to the aversive effects of ethanol.
Collapse
Affiliation(s)
- Agustín Salguero
- Instituto de Investigación Médica M. y M. Ferreyra (INIMEC-CONICET-Universidad Nacional de Córdoba), Córdoba C.P. 5000, Argentina
| | - Leonardo Marengo
- Instituto de Investigación Médica M. y M. Ferreyra (INIMEC-CONICET-Universidad Nacional de Córdoba), Córdoba C.P. 5000, Argentina
| | | | - Leandro Ruiz-Leyva
- Department of Pharmacology, Institute of Neuroscience, Biomedical Research Center (CIBM) Faculty of Medicine, University of Granada and Biosanitary Research Institute ibs.GRANADA, Granada, Spain
| | - Cruz Miguel Cendán
- Department of Pharmacology, Institute of Neuroscience, Biomedical Research Center (CIBM) Faculty of Medicine, University of Granada and Biosanitary Research Institute ibs.GRANADA, Granada, Spain
| | - Ignacio Morón
- Department of Psychobiology and Centre of Investigation of Mind, Brain, and Behaviour (CIMCYC), University of Granada, Spain.
| | - Ricardo Marcos Pautassi
- Instituto de Investigación Médica M. y M. Ferreyra (INIMEC-CONICET-Universidad Nacional de Córdoba), Córdoba C.P. 5000, Argentina; Facultad de Psicología, Universidad Nacional de Córdoba, Córdoba, C.P. 5000, Argentina.
| |
Collapse
|
38
|
Deng B, Zhang Z, Zhou H, Zhang X, Niu S, Yan X, Yan J. MicroRNAs in Methamphetamine-Induced Neurotoxicity and Addiction. Front Pharmacol 2022; 13:875666. [PMID: 35496314 PMCID: PMC9046672 DOI: 10.3389/fphar.2022.875666] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/31/2022] [Indexed: 12/21/2022] Open
Abstract
Methamphetamine (METH) abuse remains a significant public health concern globally owing to its strong addictive properties. Prolonged abuse of the drug causes irreversible damage to the central nervous system. To date, no efficient pharmacological interventions are available, primarily due to the unclear mechanisms underlying METH action in the brain. Recently, microRNAs (miRNAs) have been identified to play critical roles in various cellular processes. The expression levels of some miRNAs are altered after METH administration, which may influence the transcription of target genes to regulate METH toxicity or addiction. This review summarizes the miRNAs in the context of METH use, discussing their role in the reward effect and neurotoxic sequelae. Better understanding of the molecular mechanisms involved in METH would be helpful for the development of new therapeutic strategies in reducing the harm of the drug.
Collapse
Affiliation(s)
- Bi Deng
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhirui Zhang
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
| | - Huixuan Zhou
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Xinran Zhang
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
| | - Shuliang Niu
- School of Basic Medical Science, Xinjiang Medical University, Urumqi, China
| | - Xisheng Yan
- Department of Cardiovascular Medicine, Wuhan Third Hospital and Tongren Hospital of Wuhan University, Wuhan, China
| | - Jie Yan
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
- School of Basic Medical Science, Xinjiang Medical University, Urumqi, China
- *Correspondence: Jie Yan,
| |
Collapse
|
39
|
Icariside II Attenuates Methamphetamine-Induced Neurotoxicity and Behavioral Impairments via Activating the Keap1-Nrf2 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8400876. [PMID: 35387263 PMCID: PMC8979738 DOI: 10.1155/2022/8400876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/26/2021] [Accepted: 03/11/2022] [Indexed: 12/15/2022]
Abstract
Chronic and long-term methamphetamine (METH) abuse is bound to cause damages to multiple organs and systems, especially the central nervous system (CNS). Icariside II (ICS), a type of flavonoid and one of the main active ingredients of the traditional Chinese medicine Epimedium, exhibits a variety of biological and pharmacological properties such as anti-inflammatory, antioxidant, and anticancer activities. However, whether ICS could protect against METH-induced neurotoxicity remains unknown. Based on a chronic METH abuse mouse model, we detected the neurotoxicity after METH exposure and determined the intervention effect of ICS and the potential mechanism of action. Here, we found that METH could trigger neurotoxicity, which was characterized by loss of dopaminergic neurons, depletion of dopamine (DA), activation of glial cells, upregulation of α-synuclein (α-syn), abnormal dendritic spine plasticity, and dysfunction of motor coordination and balance. ICS treatment, however, alleviated the above-mentioned neurotoxicity elicited by METH. Our data also indicated that when ICS combated METH-induced neurotoxicity, it was accompanied by partial correction of the abnormal Kelch 2 like ECH2 associated protein 1 (Keap1)-nuclear factor erythroid-2-related factor 2 (Nrf2) pathway and oxidative stress response. In the presence of ML385, an inhibitor of Nrf2, ICS failed to activate the Nrf2-related protein expression and reduce the oxidative stress response. More importantly, ICS could not attenuate METH-induced dopaminergic neurotoxicity and behavioral damage when the Nrf2 was inhibited, suggesting that the neuroprotective effect of ICS on METH-induced neurotoxicity was dependent on activating the Keap1-Nrf2 pathway. Although further research is needed to dig deeper into the actual molecular targets of ICS, it is undeniable that the current results imply the potential value of ICS to reduce the neurotoxicity of METH abusers.
Collapse
|
40
|
Crouzier L, Danese A, Yasui Y, Richard EM, Liévens JC, Patergnani S, Couly S, Diez C, Denus M, Cubedo N, Rossel M, Thiry M, Su TP, Pinton P, Maurice T, Delprat B. Activation of the sigma-1 receptor chaperone alleviates symptoms of Wolfram syndrome in preclinical models. Sci Transl Med 2022; 14:eabh3763. [PMID: 35138910 PMCID: PMC9516885 DOI: 10.1126/scitranslmed.abh3763] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The Wolfram syndrome is a rare autosomal recessive disease affecting many organs with life-threatening consequences; currently, no treatment is available. The disease is caused by mutations in the WSF1 gene, coding for the protein wolframin, an endoplasmic reticulum (ER) transmembrane protein involved in contacts between ER and mitochondria termed as mitochondria-associated ER membranes (MAMs). Inherited mutations usually reduce the protein's stability, altering its homeostasis and ultimately reducing ER to mitochondria calcium ion transfer, leading to mitochondrial dysfunction and cell death. In this study, we found that activation of the sigma-1 receptor (S1R), an ER-resident protein involved in calcium ion transfer, could counteract the functional alterations of MAMs due to wolframin deficiency. The S1R agonist PRE-084 restored calcium ion transfer and mitochondrial respiration in vitro, corrected the associated increased autophagy and mitophagy, and was able to alleviate the behavioral symptoms observed in zebrafish and mouse models of the disease. Our findings provide a potential therapeutic strategy for treating Wolfram syndrome by efficiently boosting MAM function using the ligand-operated S1R chaperone. Moreover, such strategy might also be relevant for other degenerative and mitochondrial diseases involving MAM dysfunction.
Collapse
Affiliation(s)
- Lucie Crouzier
- MMDN, Univ Montpellier, EPHE, INSERM, Montpellier, France
| | - Alberto Danese
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Yuko Yasui
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, 333 Cassell Drive, Baltimore, MD 21224, USA
| | | | | | - Simone Patergnani
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Simon Couly
- MMDN, Univ Montpellier, EPHE, INSERM, Montpellier, France
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Camille Diez
- MMDN, Univ Montpellier, EPHE, INSERM, Montpellier, France
| | - Morgane Denus
- MMDN, Univ Montpellier, EPHE, INSERM, Montpellier, France
| | - Nicolas Cubedo
- MMDN, Univ Montpellier, EPHE, INSERM, Montpellier, France
| | | | - Marc Thiry
- Laboratoire de Biologie Cellulaire, Université de Liège, GIGA-Neurosciences, Quartier Hopital, Avenue Hippocrate 15, 4000 Liege 1, Belgium
| | - Tsung-Ping Su
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Paolo Pinton
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Tangui Maurice
- MMDN, Univ Montpellier, EPHE, INSERM, Montpellier, France
| | | |
Collapse
|
41
|
Wang X, Tong B, Hui R, Hou C, Zhang Z, Zhang L, Xie B, Ni Z, Cong B, Ma C, Wen D. The Role of Hyperthermia in Methamphetamine-Induced Depression-Like Behaviors: Protective Effects of Coral Calcium Hydride. Front Mol Neurosci 2022; 14:808807. [PMID: 35058751 PMCID: PMC8764150 DOI: 10.3389/fnmol.2021.808807] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/06/2021] [Indexed: 12/15/2022] Open
Abstract
Methamphetamine (METH) abuse causes irreversible damage to the central nervous system and leads to psychiatric symptoms including depression. Notably, METH-induced hyperthermia is a crucial factor in the development of these symptoms, as it aggravates METH-induced neurotoxicity. However, the role of hyperthermia in METH-induced depression-like behaviors needs to be clarified. In the present study, we treated mice with different doses of METH under normal (NAT) or high ambient temperatures (HAT). We found that HAT promoted hyperthermia after METH treatment and played a key role in METH-induced depression-like behaviors in mice. Intriguingly, chronic METH exposure (10 mg/kg, 7 or 14 days) or administration of an escalating-dose (2 ∼ 15 mg/kg, 3 days) of METH under NAT failed to induce depression-like behaviors. However, HAT aggravated METH-induced damage of hippocampal synaptic plasticity, reaction to oxidative stress, and neuroinflammation. Molecular hydrogen acts as an antioxidant and anti-inflammatory agent and has been shown to have preventive and therapeutic applicability in a wide range of diseases. Coral calcium hydride (CCH) is a newly identified hydrogen-rich powder which produces hydrogen gas gradually when exposed to water. Herein, we found that CCH pretreatment significantly attenuated METH-induced hyperthermia, and administration of CCH after METH exposure also inhibited METH-induced depression-like behaviors and reduced the hippocampal synaptic plasticity damage. Moreover, CCH effectively reduced the activity of lactate dehydrogenase and decreased malondialdehyde, TNF-α and IL-6 generation in hippocampus. These results suggest that CCH is an efficient hydrogen-rich agent, which has a potential therapeutic applicability in the treatment of METH abusers.
Collapse
Affiliation(s)
- Xintao Wang
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Bonan Tong
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Rongji Hui
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Congcong Hou
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Zilu Zhang
- The First Clinical Medical College of Peking University Health Science Center, Peking University, Beijing, China
| | - Ludi Zhang
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Bing Xie
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Zhiyu Ni
- School of Basic Medical Sciences, Hebei University, Baoding, China
| | - Bin Cong
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Chunling Ma
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
- *Correspondence: Chunling Ma,
| | - Di Wen
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
- Di Wen,
| |
Collapse
|
42
|
Veerappa A, Pendyala G, Guda C. A systems omics-based approach to decode substance use disorders and neuroadaptations. Neurosci Biobehav Rev 2021; 130:61-80. [PMID: 34411560 PMCID: PMC8511293 DOI: 10.1016/j.neubiorev.2021.08.016] [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: 05/21/2021] [Revised: 07/23/2021] [Accepted: 08/14/2021] [Indexed: 11/15/2022]
Abstract
Substance use disorders (SUDs) are a group of neuropsychiatric conditions manifesting due to excessive dependence on potential drugs of abuse such as psychostimulants, opioids including prescription opioids, alcohol, inhalants, etc. Experimental studies have generated enormous data in the area of SUDs, but outcomes from such data have remained largely fragmented. In this review, we attempt to coalesce these data points providing an important first step towards our understanding of the etiology of SUDs. We propose and describe a 'core addictome' pathway that behaves central to all SUDs. Besides, we also have made some notable observations paving way for several hypotheses; MECP2 behaves as a master switch during substance use; five distinct gene clusters were identified based on respective substance addiction; a central cluster of genes serves as a hub of the addiction pathway connecting all other substance addiction clusters. In addition to describing these findings, we have emphasized the importance of some candidate genes that are of substantial interest for further investigation and serve as high-value targets for translational efforts.
Collapse
Affiliation(s)
- Avinash Veerappa
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Gurudutt Pendyala
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Center for Biomedical Informatics Research and Innovation, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
43
|
Zhao W, Zhao YL, Liu M, Liu L, Wang Y. Possible repair mechanisms of renin-angiotensin system inhibitors, matrix metalloproteinase-9 inhibitors and protein hormones on methamphetamine-induced neurotoxicity. Mol Biol Rep 2021; 48:7509-7516. [PMID: 34623593 DOI: 10.1007/s11033-021-06741-y] [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: 06/12/2021] [Accepted: 09/08/2021] [Indexed: 12/31/2022]
Abstract
Methamphetamine is a highly addictive central stimulant with extensive and strong neurotoxicity. The neurotoxicity of methamphetamine is closely related to the imbalance of dopamine levels and the destruction of the blood-brain barrier. An increase in dopamine may induce adverse effects such as behavioral sensitization and excessive locomotion. Damage to the blood-brain barrier can cause toxic or harmful substances to leak to the central nervous system, leading to neurotoxicity. The renin-angiotensin system is essential for the regulation of dopamine levels in the brain. Matrix metalloproteinase-9 causes reward effects and behavioral sensitization by inducing dopamine release. Prolactin has been shown to be involved in the regulation of tight junction proteins and the integrity of the blood-brain barrier. At present, the treatment of methamphetamine detoxification is still based on psychotherapy, and there is no specific medicine. With the rapid increase in global seizures of methamphetamine, the treatment of its toxicity has attracted more and more attention. This review intends to summarize the therapeutic mechanisms of renin-angiotensin inhibitors, matrix metalloproteinase-9 inhibitors and protein hormones (prolactin) on methamphetamine neurotoxicity. The repair effects of these three on methamphetamine may be related to the maintenance of brain dopamine balance and the integrity of the blood-brain barrier. This review is expected to provide the new therapeutic strategy of methamphetamine toxicity.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Drug Control, Criminal Investigation Police University of China, Shenyang, 110854, Liaoning, People's Republic of China.,Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China
| | - Yuan-Ling Zhao
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China
| | - Ming Liu
- Department of Drug Control, Criminal Investigation Police University of China, Shenyang, 110854, Liaoning, People's Republic of China
| | - Lian Liu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China
| | - Yun Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China.
| |
Collapse
|
44
|
Wang M, Wan C, He T, Han C, Zhu K, Waddington JL, Zhen X. Sigma-1 receptor regulates mitophagy in dopaminergic neurons and contributes to dopaminergic protection. Neuropharmacology 2021; 196:108360. [PMID: 33122030 DOI: 10.1016/j.neuropharm.2020.108360] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 10/12/2020] [Accepted: 10/16/2020] [Indexed: 12/26/2022]
Abstract
Mitochondria are essential for neuronal survival and function, and mitochondrial dysfunction plays a critical role in the pathological development of Parkinson's disease (PD). Mitochondrial quality control is known to contribute to the survival of dopaminergic (DA) neurons, with mitophagy being a key regulator of the quality control system. In this study, we show that mitophagy is impaired in the substantia nigra pars compacta (SNc) of the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD. Treatment with the sigma-1 receptor (Sig 1R) agonist 2-morpholin-4-ylethyl 1-phenylcyclohexane-1-carboxylate (PRE-084) reduced loss of DA neurons, restored motor ability and MPTP-induced damage to mitophagy activity in the SNc of PD-like mice. Additionally, knockdown of Sig 1R in SH-SY5Y DA cells inhibited mitophagy and enhanced 1-methyl-4-phenylpyridinium ion (MPP+) neurotoxicity, whereas application of the Sig 1R selective agonist SKF10047 promoted clearance of damaged mitochondria. Moreover, knockdown of Sig 1R in SH-SY5Y cells resulted in decreased levels of p-ULK1 (Unc-51 Like Autophagy Activating Kinase 1) (Ser555), p-TBK1 (TANK Binding Kinase 1) (Ser172), p-ubiquitin (Ub) (Ser65), Parkin recruitment, and stabilization of PTEN-induced putative kinase 1 (PINK1) in mitochondria. The present data provide the first evidence for potential roles of PINK1/Parkin in Sig 1R-modulated mitophagy in DA neurons.
Collapse
Affiliation(s)
- Mingmei Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China; College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou, China
| | - Chunlei Wan
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China; College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou, China
| | - Tao He
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China; College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou, China
| | - Chaojun Han
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China; College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou, China
| | - Kailian Zhu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China; College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou, China
| | - John L Waddington
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China; School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Xuechu Zhen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China; College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou, China.
| |
Collapse
|
45
|
Sanjari Moghaddam H, Mobarak Abadi M, Dolatshahi M, Bayani Ershadi S, Abbasi-Feijani F, Rezaei S, Cattarinussi G, Aarabi MH. Effects of Prenatal Methamphetamine Exposure on the Developing Human Brain: A Systematic Review of Neuroimaging Studies. ACS Chem Neurosci 2021; 12:2729-2748. [PMID: 34297546 PMCID: PMC8763371 DOI: 10.1021/acschemneuro.1c00213] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
![]()
Methamphetamine
(MA) can cross the placenta in pregnant women and
cause placental abruption and developmental alterations in offspring.
Previous studies have found prenatal MA exposure effects on the social
and cognitive performance of children. Recent studies reported some
alterations in structural and functional magnetic resonance imaging
(MRI) of prenatal MA-exposed offspring. In this study, we aimed to
investigate the effect of prenatal MA exposure on brain development
using recently published structural, metabolic, and functional MRI
studies. According to the Preferred Reporting Items for Systematic
Reviews and Meta-Analyses (PRISMA) guidelines, we searched PubMed
and SCOPUS databases for articles that used each brain imaging modality
in prenatal MA-exposed children. Seventeen studies were included in
this study. We investigated brain imaging alterations using 17 articles
with four different modalities, including structural MRI, diffusion
tensor imaging (DTI), magnetic resonance spectroscopy (MRS), and functional
MRI (fMRI). The participants’ age range was from infancy to
15 years. Our findings demonstrated that prenatal MA exposure is associated
with macrostructural, microstructural, metabolic, and functional deficits
in both cortical and subcortical areas. However, the most affected
regions were the striatum, frontal lobe, thalamus and the limbic system,
and white matter (WM) fibers connecting these regions. The findings
from our study might have valuable implications for targeted treatment
of neurocognitive and behavioral deficits in children with prenatal
MA exposure. Even so, our results should be interpreted cautiously
due to the heterogeneity of the included studies in terms of study
populations and methods of analysis.
Collapse
Affiliation(s)
| | | | - Mahsa Dolatshahi
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Sahar Rezaei
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Giulia Cattarinussi
- Department of Neuroscience and Padova Neuroscience Center (PNC), University of Padova, 35131 Padova, Italy
| | - Mohammad Hadi Aarabi
- Department of Neuroscience and Padova Neuroscience Center (PNC), University of Padova, 35131 Padova, Italy
| |
Collapse
|
46
|
Aishwarya R, Abdullah CS, Morshed M, Remex NS, Bhuiyan MS. Sigmar1's Molecular, Cellular, and Biological Functions in Regulating Cellular Pathophysiology. Front Physiol 2021; 12:705575. [PMID: 34305655 PMCID: PMC8293995 DOI: 10.3389/fphys.2021.705575] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/07/2021] [Indexed: 12/11/2022] Open
Abstract
The Sigma 1 receptor (Sigmar1) is a ubiquitously expressed multifunctional inter-organelle signaling chaperone protein playing a diverse role in cellular survival. Recessive mutation in Sigmar1 have been identified as a causative gene for neuronal and neuromuscular disorder. Since the discovery over 40 years ago, Sigmar1 has been shown to contribute to numerous cellular functions, including ion channel regulation, protein quality control, endoplasmic reticulum-mitochondrial communication, lipid metabolism, mitochondrial function, autophagy activation, and involved in cellular survival. Alterations in Sigmar1’s subcellular localization, expression, and signaling has been implicated in the progression of a wide range of diseases, such as neurodegenerative diseases, ischemic brain injury, cardiovascular diseases, diabetic retinopathy, cancer, and drug addiction. The goal of this review is to summarize the current knowledge of Sigmar1 biology focusing the recent discoveries on Sigmar1’s molecular, cellular, pathophysiological, and biological functions.
Collapse
Affiliation(s)
- Richa Aishwarya
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Chowdhury S Abdullah
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Mahboob Morshed
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Naznin Sultana Remex
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Md Shenuarin Bhuiyan
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States.,Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| |
Collapse
|
47
|
Romeo G, Bonanno F, Wilson LL, Arena E, Modica MN, Pittalà V, Salerno L, Prezzavento O, McLaughlin JP, Intagliata S. Development of New Benzylpiperazine Derivatives as σ 1 Receptor Ligands with in Vivo Antinociceptive and Anti-Allodynic Effects. ACS Chem Neurosci 2021; 12:2003-2012. [PMID: 34019387 PMCID: PMC8291485 DOI: 10.1021/acschemneuro.1c00106] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/10/2021] [Indexed: 12/22/2022] Open
Abstract
σ-1 receptors (σ1R) modulate nociceptive signaling, driving the search for selective antagonists to take advantage of this promising target to treat pain. In this study, a new series of benzylpiperazinyl derivatives has been designed, synthesized, and characterized for their affinities toward σ1R and selectivity over the σ-2 receptor (σ2R). Notably, 3-cyclohexyl-1-{4-[(4-methoxyphenyl)methyl]piperazin-1-yl}propan-1-one (15) showed the highest σ1R receptor affinity (Ki σ1 = 1.6 nM) among the series with a significant improvement of the σ1R selectivity (Ki σ2/Ki σ1= 886) compared to the lead compound 8 (Ki σ2/Ki σ1= 432). Compound 15 was further tested in a mouse formalin assay of inflammatory pain and chronic nerve constriction injury (CCI) of neuropathic pain, where it produced dose-dependent (3-60 mg/kg, i.p.) antinociception and anti-allodynic effects. Moreover, compound 15 demonstrated no significant effects in a rotarod assay, suggesting that this σ1R antagonist did not produce sedation or impair locomotor responses. Overall, these results encourage the further development of our benzylpiperazine-based σ1R antagonists as potential therapeutics for chronic pain.
Collapse
Affiliation(s)
- Giuseppe Romeo
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| | - Federica Bonanno
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| | - Lisa L. Wilson
- Department
of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Emanuela Arena
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| | - Maria N. Modica
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| | - Valeria Pittalà
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| | - Loredana Salerno
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| | - Orazio Prezzavento
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| | - Jay P. McLaughlin
- Department
of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Sebastiano Intagliata
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| |
Collapse
|
48
|
Xu L, Chen LY. Association of sigma-1 receptor with dopamine transporter attenuates the binding of methamphetamine via distinct helix-helix interactions. Chem Biol Drug Des 2021; 97:1194-1209. [PMID: 33754484 PMCID: PMC8113090 DOI: 10.1111/cbdd.13841] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/23/2021] [Accepted: 03/07/2021] [Indexed: 02/06/2023]
Abstract
Dopamine transporter (DAT) and sigma-1 receptor (σ1R) are potential therapeutic targets to reduce the psychostimulant effects induced by methamphetamine (METH). Interaction of σ1R with DAT could modulate the binding of METH, but the molecular basis of the association of the two transmembrane proteins and how their interactions mediate the binding of METH to DAT or σ1R remain unclear. Here, we characterize the protein-ligand and protein-protein interactions at a molecular level by various theoretical approaches. The present results show that METH adopts a different binding pose in the binding pocket of σ1R and is more likely to act as an agonist. The relatively lower binding affinity of METH to σ1R supports the role of antagonists as inhibitors that protect against METH-induced effects. We demonstrate that σ1R could bind to Drosophila melanogaster DAT (dDAT) through interactions with either the transmembrane helix α12 or α5 of dDAT. Our results showed that the truncated σ1R displays stronger association with dDAT than the full-length σ1R. Although different helix-helix interactions between σ1R and dDAT lead to distinct effects on the dynamics of individual protein, both associations attenuate the binding affinity of METH to dDAT, particularly in the interactions with the helix α5 of dDAT. Together, the present study provides the first computational investigation on the molecular mechanism of coupling METH binding and the association of σ1R with dDAT.
Collapse
Affiliation(s)
- Liang Xu
- Department of Physics and Astronomy, University of Texas at San Antonio, San Antonio, TX, USA
| | - Liao Y Chen
- Department of Physics and Astronomy, University of Texas at San Antonio, San Antonio, TX, USA
| |
Collapse
|
49
|
Wang SM, Goguadze N, Kimura Y, Yasui Y, Pan B, Wang TY, Nakamura Y, Lin YT, Hogan QH, Wilson KL, Su TP, Wu HE. Genomic Action of Sigma-1 Receptor Chaperone Relates to Neuropathic Pain. Mol Neurobiol 2021; 58:2523-2541. [PMID: 33459966 PMCID: PMC8128747 DOI: 10.1007/s12035-020-02276-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 12/28/2020] [Indexed: 11/16/2022]
Abstract
Sigma-1 receptors (Sig-1Rs) are endoplasmic reticulum (ER) chaperones implicated in neuropathic pain. Here we examine if the Sig-1R may relate to neuropathic pain at the level of dorsal root ganglia (DRG). We focus on the neuronal excitability of DRG in a "spare nerve injury" (SNI) model of neuropathic pain in rats and find that Sig-1Rs likely contribute to the genesis of DRG neuronal excitability by decreasing the protein level of voltage-gated Cav2.2 as a translational inhibitor of mRNA. Specifically, during SNI, Sig-1Rs translocate from ER to the nuclear envelope via a trafficking protein Sec61β. At the nucleus, the Sig-1R interacts with cFos and binds to the promoter of 4E-BP1, leading to an upregulation of 4E-BP1 that binds and prevents eIF4E from initiating the mRNA translation for Cav2.2. Interestingly, in Sig-1R knockout HEK cells, Cav2.2 is upregulated. In accordance with those findings, we find that intra-DRG injection of Sig-1R agonist (+)pentazocine increases frequency of action potentials via regulation of voltage-gated Ca2+ channels. Conversely, intra-DRG injection of Sig-1R antagonist BD1047 attenuates neuropathic pain. Hence, we discover that the Sig-1R chaperone causes neuropathic pain indirectly as a translational inhibitor.
Collapse
MESH Headings
- Animals
- Calcium Channels, N-Type/genetics
- Calcium Channels, N-Type/metabolism
- Endoplasmic Reticulum/metabolism
- Eukaryotic Initiation Factor-4E/metabolism
- Ganglia, Spinal/metabolism
- Gene Expression Regulation
- Genome
- HEK293 Cells
- Humans
- Intracellular Signaling Peptides and Proteins/metabolism
- Male
- Nerve Tissue/injuries
- Nerve Tissue/pathology
- Neuralgia/genetics
- Nuclear Envelope/metabolism
- Promoter Regions, Genetic/genetics
- Protein Biosynthesis
- Proto-Oncogene Proteins c-fos/metabolism
- RNA Caps/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Sprague-Dawley
- Receptors, sigma/agonists
- Receptors, sigma/genetics
- Receptors, sigma/metabolism
- SEC Translocation Channels/metabolism
- Transcription, Genetic
- Sigma-1 Receptor
- Rats
Collapse
Affiliation(s)
- Shao-Ming Wang
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Nino Goguadze
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Yuriko Kimura
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Yuko Yasui
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Bin Pan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Tzu-Yun Wang
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
- Department of Psychiatry, College of Medicine, National Cheng Kung University, Tainan City, 70101, Taiwan
| | - Yoki Nakamura
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
- Department of Pharmacology, Graduate School of Biomedical & Health Science, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Yu-Ting Lin
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Quinn H Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Katherine L Wilson
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Tsung-Ping Su
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA.
| | - Hsiang-En Wu
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
| |
Collapse
|
50
|
Pino JA, Nuñez-Vivanco G, Hidalgo G, Reyes Parada M, Khoshbouei H, Torres GE. Identification of Critical Residues in the Carboxy Terminus of the Dopamine Transporter Involved in the G Protein βγ-Induced Dopamine Efflux. Front Pharmacol 2021; 12:642881. [PMID: 33841159 PMCID: PMC8025876 DOI: 10.3389/fphar.2021.642881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/11/2021] [Indexed: 11/13/2022] Open
Abstract
The dopamine transporter (DAT) plays a crucial role in the regulation of brain dopamine (DA) homeostasis through the re-uptake of DA back into the presynaptic terminal. In addition to re-uptake, DAT is also able to release DA through a process referred to as DAT-mediated DA efflux. This is the mechanism by which potent and highly addictive psychostimulants, such as amphetamine (AMPH) and its analogues, increase extracellular DA levels in motivational and reward areas of the brain. Recently, we discovered that G protein βγ subunits (Gβγ) binds to the DAT, and that activation of Gβγ results in DAT-mediated efflux - a similar mechanism as AMPH. Previously, we have shown that Gβγ binds directly to a stretch of 15 residues within the intracellular carboxy terminus of DAT (residues 582-596). Additionally, a TAT peptide containing residues 582 to 596 of DAT was able to block the Gβγ-induced DA efflux through DAT. Here, we use a combination of computational biology, mutagenesis, biochemical, and functional assays to identify the amino acid residues within the 582-596 sequence of the DAT carboxy terminus involved in the DAT-Gβγ interaction and Gβγ-induced DA efflux. Our in-silico protein-protein docking analysis predicted the importance of F587 and R588 residues in a network of interactions with residues in Gβγ. In addition, we observed that mutating R588 to alanine residue resulted in a mutant DAT which exhibited attenuated DA efflux induced by Gβγ activation. We demonstrate that R588, and to a lesser extent F5837, located within the carboxy terminus of DAT play a critical role in the DAT-Gβγ physical interaction and promotion of DA efflux. These results identify a potential new pharmacological target for the treatment of neuropsychiatric conditions in which DAT functionality is implicated including ADHD and substance use disorder.
Collapse
Affiliation(s)
- José A Pino
- Departamento de Medicina, Facultad de Medicina, Universidad de Atacama, Copiapó, Chile
| | - Gabriel Nuñez-Vivanco
- Centro de Bioinformática, Simulación y Modelado, Facultad de Ingeniería, Universidad de Talca, Talca, Chile
| | - Gabriela Hidalgo
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Miguel Reyes Parada
- Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Santiago, Chile.,Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| | - Habibeh Khoshbouei
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States
| | - Gonzalo E Torres
- Department of Molecular, Cellular and Biomedical Sciences, CUNY School of Medicine at City College of New York, New York, NY, United States
| |
Collapse
|