1
|
Shinozuka K, Tabaac BJ, Arenas A, Beutler BD, Cherian K, Evans VD, Fasano C, Muir OS. Psychedelic Therapy: A Primer for Primary Care Clinicians-3,4-Methylenedioxy-methamphetamine (MDMA). Am J Ther 2024; 31:e141-e154. [PMID: 38518271 DOI: 10.1097/mjt.0000000000001722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2024]
Abstract
BACKGROUND After becoming notorious for its use as a party drug in the 1980s, 3,4-methylenedioxy-methampetamine (MDMA), also known by its street names "molly" and "ecstasy," has emerged as a powerful treatment for post-traumatic stress disorder (PTSD). AREAS OF UNCERTAINTY There are extensive data about the risk profile of MDMA. However, the literature is significantly biased. Animal models demonstrating neurotoxic or adverse effects used doses well beyond the range that would be expected in humans (up to 40 mg/kg in rats compared with roughly 1-2 mg/kg in humans). Furthermore, human samples often comprise recreational users who took other substances in addition to MDMA, in uncontrolled settings. THERAPEUTIC ADVANCES Phase III clinical trials led by the Multidisciplinary Association for Psychedelic Studies (MAPS) have shown that MDMA-assisted psychotherapy has an effect size of d = 0.7-0.91, up to 2-3 times higher than the effect sizes of existing antidepressant treatments. 67%-71% of patients who undergo MDMA-assisted psychotherapy no longer meet the diagnostic criteria for PTSD within 18 weeks. We also describe other promising applications of MDMA-assisted psychotherapy for treating alcohol use disorder, social anxiety, and other psychiatric conditions. LIMITATIONS Thus far, almost all clinical trials on MDMA have been sponsored by a single organization, MAPS. More work is needed to determine whether MDMA-assisted therapy is more effective than existing nonpharmacological treatments such as cognitive behavioral therapy. CONCLUSIONS Phase III trials suggest that MDMA is superior to antidepressant medications for treating PTSD. Now that MAPS has officially requested the Food and Drug Administration to approve MDMA as a treatment for PTSD, legal MDMA-assisted therapy may become available as soon as 2024.
Collapse
Affiliation(s)
- Kenneth Shinozuka
- Centre for Eudaimonia and Human Flourishing, University of Oxford, Oxford, United Kingdom
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Burton J Tabaac
- University of Nevada, Reno School of Medicine, Reno, NV
- Department of Neurology, Carson Tahoe Health, Carson City, NV
| | - Alejandro Arenas
- Department of Anesthesiology, University of Washington School of Medicine, Seattle, WA
| | - Bryce D Beutler
- University of Southern California, Keck School of Medicine, Los Angeles, CA
| | - Kirsten Cherian
- Department of Psychiatry & Behavioral Sciences, Stanford University, Palo Alto, CA
| | - Viviana D Evans
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Owen S Muir
- Fermata Health, Brooklyn, NY; and
- Acacia Clinics, Sunnyvale, CA
| |
Collapse
|
2
|
Latimer D, Stocker MD, Sayers K, Green J, Kaye AM, Abd-Elsayed A, Cornett EM, Kaye AD, Varrassi G, Viswanath O, Urits I. MDMA to Treat PTSD in Adults. PSYCHOPHARMACOLOGY BULLETIN 2021; 51:125-149. [PMID: 34421149 PMCID: PMC8374929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Post-traumatic stress disorder (PTSD) has become one of the most common psychiatric diagnosis in the United States specifically within the veteran population. The current treatment options for this debilitating diagnosis include trauma-focused psychotherapies along with selective serotonin reuptake inhibitors (SSRI) and serotonin-norepinephrine reuptake inhibitors (SNRI).1 MDMA has recently been shown as a novel therapeutic agent with promisingly results in the treatment of PTSD. MDMA is a psychoactive compound traditionally categorized as a psychedelic amphetamine that deemed a Schedule I controlled substance in the 1980s. Prior to its status as a controlled substance, it was used by psychotherapists for an array of psychiatric issues. In more recent times, MDMA has resurfaced as a potential therapy for PTSD and the data produced from randomized, controlled trials back the desire for MDMA to be utilized as an effective pharmacologic therapy in conjunction with psychotherapy.2.
Collapse
Affiliation(s)
- Dustin Latimer
- Latimer, Louisiana State University Health Science Center, Department of Psychiatry and Behavioral Medicine in Baton Rouge. Michael D. Stocker, Kia Sayers, Louisiana State University New Orleans School of Medicine. Green, Visions Adolescent Treatment Center, Department of Mental Health, Los Angeles, CA. Adam M. Kaye, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Alaa Abd-Elsayed, University of Wisconsin School of Medicine and Public Health, Department of Anesthesiology, Madison, WI. Elyse M. Cornett, Alan D. Kaye, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Varrassi, Paolo Procacci Foundation, Via Tacito 7, Roma, Italy. Viswanath, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA; University of Arizona College of Medicine-Phoenix, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE; Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Ivan Urits, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA; Southcoast Health, Southcoast Health Physicians Group Pain Medicine, Wareham, MA
| | - Michael D Stocker
- Latimer, Louisiana State University Health Science Center, Department of Psychiatry and Behavioral Medicine in Baton Rouge. Michael D. Stocker, Kia Sayers, Louisiana State University New Orleans School of Medicine. Green, Visions Adolescent Treatment Center, Department of Mental Health, Los Angeles, CA. Adam M. Kaye, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Alaa Abd-Elsayed, University of Wisconsin School of Medicine and Public Health, Department of Anesthesiology, Madison, WI. Elyse M. Cornett, Alan D. Kaye, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Varrassi, Paolo Procacci Foundation, Via Tacito 7, Roma, Italy. Viswanath, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA; University of Arizona College of Medicine-Phoenix, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE; Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Ivan Urits, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA; Southcoast Health, Southcoast Health Physicians Group Pain Medicine, Wareham, MA
| | - Kia Sayers
- Latimer, Louisiana State University Health Science Center, Department of Psychiatry and Behavioral Medicine in Baton Rouge. Michael D. Stocker, Kia Sayers, Louisiana State University New Orleans School of Medicine. Green, Visions Adolescent Treatment Center, Department of Mental Health, Los Angeles, CA. Adam M. Kaye, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Alaa Abd-Elsayed, University of Wisconsin School of Medicine and Public Health, Department of Anesthesiology, Madison, WI. Elyse M. Cornett, Alan D. Kaye, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Varrassi, Paolo Procacci Foundation, Via Tacito 7, Roma, Italy. Viswanath, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA; University of Arizona College of Medicine-Phoenix, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE; Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Ivan Urits, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA; Southcoast Health, Southcoast Health Physicians Group Pain Medicine, Wareham, MA
| | - Jackson Green
- Latimer, Louisiana State University Health Science Center, Department of Psychiatry and Behavioral Medicine in Baton Rouge. Michael D. Stocker, Kia Sayers, Louisiana State University New Orleans School of Medicine. Green, Visions Adolescent Treatment Center, Department of Mental Health, Los Angeles, CA. Adam M. Kaye, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Alaa Abd-Elsayed, University of Wisconsin School of Medicine and Public Health, Department of Anesthesiology, Madison, WI. Elyse M. Cornett, Alan D. Kaye, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Varrassi, Paolo Procacci Foundation, Via Tacito 7, Roma, Italy. Viswanath, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA; University of Arizona College of Medicine-Phoenix, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE; Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Ivan Urits, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA; Southcoast Health, Southcoast Health Physicians Group Pain Medicine, Wareham, MA
| | - Adam M Kaye
- Latimer, Louisiana State University Health Science Center, Department of Psychiatry and Behavioral Medicine in Baton Rouge. Michael D. Stocker, Kia Sayers, Louisiana State University New Orleans School of Medicine. Green, Visions Adolescent Treatment Center, Department of Mental Health, Los Angeles, CA. Adam M. Kaye, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Alaa Abd-Elsayed, University of Wisconsin School of Medicine and Public Health, Department of Anesthesiology, Madison, WI. Elyse M. Cornett, Alan D. Kaye, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Varrassi, Paolo Procacci Foundation, Via Tacito 7, Roma, Italy. Viswanath, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA; University of Arizona College of Medicine-Phoenix, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE; Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Ivan Urits, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA; Southcoast Health, Southcoast Health Physicians Group Pain Medicine, Wareham, MA
| | - Alaa Abd-Elsayed
- Latimer, Louisiana State University Health Science Center, Department of Psychiatry and Behavioral Medicine in Baton Rouge. Michael D. Stocker, Kia Sayers, Louisiana State University New Orleans School of Medicine. Green, Visions Adolescent Treatment Center, Department of Mental Health, Los Angeles, CA. Adam M. Kaye, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Alaa Abd-Elsayed, University of Wisconsin School of Medicine and Public Health, Department of Anesthesiology, Madison, WI. Elyse M. Cornett, Alan D. Kaye, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Varrassi, Paolo Procacci Foundation, Via Tacito 7, Roma, Italy. Viswanath, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA; University of Arizona College of Medicine-Phoenix, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE; Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Ivan Urits, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA; Southcoast Health, Southcoast Health Physicians Group Pain Medicine, Wareham, MA
| | - Elyse M Cornett
- Latimer, Louisiana State University Health Science Center, Department of Psychiatry and Behavioral Medicine in Baton Rouge. Michael D. Stocker, Kia Sayers, Louisiana State University New Orleans School of Medicine. Green, Visions Adolescent Treatment Center, Department of Mental Health, Los Angeles, CA. Adam M. Kaye, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Alaa Abd-Elsayed, University of Wisconsin School of Medicine and Public Health, Department of Anesthesiology, Madison, WI. Elyse M. Cornett, Alan D. Kaye, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Varrassi, Paolo Procacci Foundation, Via Tacito 7, Roma, Italy. Viswanath, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA; University of Arizona College of Medicine-Phoenix, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE; Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Ivan Urits, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA; Southcoast Health, Southcoast Health Physicians Group Pain Medicine, Wareham, MA
| | - Alan D Kaye
- Latimer, Louisiana State University Health Science Center, Department of Psychiatry and Behavioral Medicine in Baton Rouge. Michael D. Stocker, Kia Sayers, Louisiana State University New Orleans School of Medicine. Green, Visions Adolescent Treatment Center, Department of Mental Health, Los Angeles, CA. Adam M. Kaye, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Alaa Abd-Elsayed, University of Wisconsin School of Medicine and Public Health, Department of Anesthesiology, Madison, WI. Elyse M. Cornett, Alan D. Kaye, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Varrassi, Paolo Procacci Foundation, Via Tacito 7, Roma, Italy. Viswanath, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA; University of Arizona College of Medicine-Phoenix, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE; Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Ivan Urits, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA; Southcoast Health, Southcoast Health Physicians Group Pain Medicine, Wareham, MA
| | - Giustino Varrassi
- Latimer, Louisiana State University Health Science Center, Department of Psychiatry and Behavioral Medicine in Baton Rouge. Michael D. Stocker, Kia Sayers, Louisiana State University New Orleans School of Medicine. Green, Visions Adolescent Treatment Center, Department of Mental Health, Los Angeles, CA. Adam M. Kaye, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Alaa Abd-Elsayed, University of Wisconsin School of Medicine and Public Health, Department of Anesthesiology, Madison, WI. Elyse M. Cornett, Alan D. Kaye, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Varrassi, Paolo Procacci Foundation, Via Tacito 7, Roma, Italy. Viswanath, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA; University of Arizona College of Medicine-Phoenix, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE; Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Ivan Urits, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA; Southcoast Health, Southcoast Health Physicians Group Pain Medicine, Wareham, MA
| | - Omar Viswanath
- Latimer, Louisiana State University Health Science Center, Department of Psychiatry and Behavioral Medicine in Baton Rouge. Michael D. Stocker, Kia Sayers, Louisiana State University New Orleans School of Medicine. Green, Visions Adolescent Treatment Center, Department of Mental Health, Los Angeles, CA. Adam M. Kaye, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Alaa Abd-Elsayed, University of Wisconsin School of Medicine and Public Health, Department of Anesthesiology, Madison, WI. Elyse M. Cornett, Alan D. Kaye, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Varrassi, Paolo Procacci Foundation, Via Tacito 7, Roma, Italy. Viswanath, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA; University of Arizona College of Medicine-Phoenix, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE; Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Ivan Urits, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA; Southcoast Health, Southcoast Health Physicians Group Pain Medicine, Wareham, MA
| | - Ivan Urits
- Latimer, Louisiana State University Health Science Center, Department of Psychiatry and Behavioral Medicine in Baton Rouge. Michael D. Stocker, Kia Sayers, Louisiana State University New Orleans School of Medicine. Green, Visions Adolescent Treatment Center, Department of Mental Health, Los Angeles, CA. Adam M. Kaye, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Department of Pharmacy Practice, Stockton, CA. Alaa Abd-Elsayed, University of Wisconsin School of Medicine and Public Health, Department of Anesthesiology, Madison, WI. Elyse M. Cornett, Alan D. Kaye, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA. Varrassi, Paolo Procacci Foundation, Via Tacito 7, Roma, Italy. Viswanath, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA; University of Arizona College of Medicine-Phoenix, Phoenix, AZ; Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE; Valley Anesthesiology and Pain Consultants-Envision Physician Services, Phoenix, AZ. Ivan Urits, Louisiana State University Shreveport, Department of Anesthesiology, Shreveport, LA; Southcoast Health, Southcoast Health Physicians Group Pain Medicine, Wareham, MA
| |
Collapse
|
3
|
Cholanians AB, Phan AV, Lau SS, Monks TJ. Concurrent Inhibition of Vesicular Monoamine Transporter 2 Does Not Protect Against 3,4-Methylenedioxymethamphetamine (Ecstasy) Induced Neurotoxicity. Toxicol Sci 2019; 170:157-166. [PMID: 30923810 DOI: 10.1093/toxsci/kfz085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
3, 4-Methylenedioxymethamphetamine (MDMA) is a hallucinogenic amphetamine derivative. The acute effects of MDMA are hyperthermia, hyperactivity, and behavioral changes, followed by long-term serotonergic neurotoxicity in rats and primates. However, the underlying mechanisms of MDMA neurotoxicity remain elusive. We reported that pretreatment of rats with Ro 4-1284, a reversible inhibitor of the vesicular monoamine transporter 2 (VMAT2), reduced MDMA-induced hyperactivity in rats, abolished the hyperthermic response, and the long-term neurotoxicity. Current studies focused on the effects of co- and/or postinhibition of VMAT2 on the acute and chronic effects of MDMA and on the dose-response relationship between MDMA-induced elevations in body temperature and subsequent reductions in indolamine concentrations. Sprague Dawley rats were treated with MDMA (20, 25, or 27.5 mg/kg sc), and either co- and/or posttreatment with the VMAT2 inhibitor (10 mg/kg ip). Rats simultaneously treated with Ro 4-1284 and MDMA exhibited a more rapid increase in body temperature compared to just MDMA. However, the duration of the elevated body temperature was significantly shortened (approximately 3 h vs approximately 8 h, respectively). A similar body temperature response was observed in rats posttreated (7 h after MDMA) with Ro 4-1284. Despite decreases in the area under the curve (Δtemp X time) of body temperature caused by Ro 4-1284, there were no significant differences in the degree of indolamine depletion between any of the MDMA-treated groups. The results suggest that the neuroprotective effects of VMAT2 inhibition is likely due to the indirect monoamine depleting effects of the Ro 4-1284 pretreatment, rather than by the direct inhibition of VMAT2 function.
Collapse
Affiliation(s)
- Aram B Cholanians
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721
| | - Andy V Phan
- Department of Radiation Oncology, School of Medicine, University of Colorado Denver, Denver, Colorado 80045
| | - Serrine S Lau
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan 48201
| | - Terrence J Monks
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan 48201
| |
Collapse
|
4
|
Dunlap LE, Andrews AM, Olson DE. Dark Classics in Chemical Neuroscience: 3,4-Methylenedioxymethamphetamine. ACS Chem Neurosci 2018; 9:2408-2427. [PMID: 30001118 PMCID: PMC6197894 DOI: 10.1021/acschemneuro.8b00155] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Better known as "ecstasy", 3,4-methylenedioxymethamphetamine (MDMA) is a small molecule that has played a prominent role in defining the ethos of today's teenagers and young adults, much like lysergic acid diethylamide (LSD) did in the 1960s. Though MDMA possesses structural similarities to compounds like amphetamine and mescaline, it produces subjective effects that are unlike any of the classical psychostimulants or hallucinogens and is one of the few compounds capable of reliably producing prosocial behavioral states. As a result, MDMA has captured the attention of recreational users, the media, artists, psychiatrists, and neuropharmacologists alike. Here, we detail the synthesis of MDMA as well as its pharmacology, metabolism, adverse effects, and potential use in medicine. Finally, we discuss its history and why it is perhaps the most important compound for the future of psychedelic science-having the potential to either facilitate new psychedelic research initiatives, or to usher in a second Dark Age for the field.
Collapse
Affiliation(s)
- Lee E Dunlap
- Department of Chemistry , University of California, Davis , One Shields Avenue , Davis , California 95616 , United States
| | - Anne M Andrews
- Departments of Psychiatry and Chemistry & Biochemistry, Semel Institute for Neuroscience and Human Behavior, and Hatos Center for Neuropharmacology , University of California , Los Angeles , California 90095 , United States
| | - David E Olson
- Department of Chemistry , University of California, Davis , One Shields Avenue , Davis , California 95616 , United States
- Department of Biochemistry & Molecular Medicine, School of Medicine , University of California, Davis , 2700 Stockton Blvd, Suite 2102 , Sacramento , California 95817 , United States
- Center for Neuroscience , University of California, Davis , 1544 Newton Ct , Davis , California 95616 , United States
| |
Collapse
|
5
|
Nakamichi N, Kato Y. Physiological Roles of Carnitine/Organic Cation Transporter OCTN1/SLC22A4 in Neural Cells. Biol Pharm Bull 2017; 40:1146-1152. [DOI: 10.1248/bpb.b17-00099] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Noritaka Nakamichi
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| | - Yukio Kato
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| |
Collapse
|
6
|
Taghizadeh G, Pourahmad J, Mehdizadeh H, Foroumadi A, Torkaman-Boutorabi A, Hassani S, Naserzadeh P, Shariatmadari R, Gholami M, Rouini MR, Sharifzadeh M. Protective effects of physical exercise on MDMA-induced cognitive and mitochondrial impairment. Free Radic Biol Med 2016; 99:11-19. [PMID: 27451936 DOI: 10.1016/j.freeradbiomed.2016.07.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 07/13/2016] [Accepted: 07/20/2016] [Indexed: 11/28/2022]
Abstract
Debate continues about the effect of 3, 4-methylenedioxymethamphetamine (MDMA) on cognitive and mitochondrial function through the CNS. It has been shown that physical exercise has an important protective effect on cellular damage and death. Therefore, we investigated the effect of physical exercise on MDMA-induced impairments of spatial learning and memory as well as MDMA effects on brain mitochondrial function in rats. Male wistar rats underwent short-term (2 weeks) or long-term (4 weeks) treadmill exercise. After completion of exercise duration, acquisition and retention of spatial memory were evaluated by Morris water maze (MWM) test. Rats were intraperitoneally (I.P) injected with MDMA (5, 10, and 15mg/kg) 30min before the first training trial in 4 training days of MWM. Different parameters of brain mitochondrial function were measured including the level of ROS production, mitochondrial membrane potential (MMP), mitochondrial swelling, mitochondrial outermembrane damage, the amount of cytochrome c release from the mitochondria, and ADP/ATP ratio. MDMA damaged the spatial learning and memory in a dose-dependent manner. Brain mitochondria isolated from the rats treated with MDMA showed significant increase in ROS formation, collapse of MMP, mitochondrial swelling, and outer membrane damage, cytochrome c release from the mitochondria, and finally increased ADP/ATP ratio. This study also found that physical exercise significantly decreased the MDMA-induced impairments of spatial learning and memory and also mitochondrial dysfunction. The results indicated that MDMA-induced neurotoxicity leads to brain mitochondrial dysfunction and subsequent oxidative stress is followed by cognitive impairments. However, physical exercise could reduce these deleterious effects of MDMA through protective effects on brain mitochondrial function.
Collapse
Affiliation(s)
- Ghorban Taghizadeh
- Department of Neuroscience, School of Advanced Science and Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jalal Pourahmad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hajar Mehdizadeh
- Department of Neuroscience, School of Advanced Science and Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Foroumadi
- Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Anahita Torkaman-Boutorabi
- Department of Neuroscience, School of Advanced Science and Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shokoufeh Hassani
- Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Parvaneh Naserzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reyhaneh Shariatmadari
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Gholami
- Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Rouini
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sharifzadeh
- Department of Neuroscience, School of Advanced Science and Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Li IH, Ma KH, Kao TJ, Lin YY, Weng SJ, Yen TY, Chen LC, Huang YS. Involvement of autophagy upregulation in 3,4-methylenedioxymethamphetamine ('ecstasy')-induced serotonergic neurotoxicity. Neurotoxicology 2015; 52:114-26. [PMID: 26610922 DOI: 10.1016/j.neuro.2015.11.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 11/16/2015] [Accepted: 11/16/2015] [Indexed: 12/20/2022]
Abstract
It has been suggested that autophagy plays pathogenetic roles in cerebral ischemia, brain trauma, and neurodegenerative disorders. 3,4-Methylenedioxymethamphetamine (MDMA or ecstasy) is an illicit drug that causes long-term serotonergic neurotoxicity in the brain. Apoptosis and necrosis have been implicated in MDMA-induced neurotoxicity, but the role of autophagy in MDMA-elicited serotonergic toxicity has not been investigated. The present study aimed to examine the contribution of autophagy to neurotoxicity in serotonergic neurons in in vitro and in vivo animal models challenged with MDMA. Here, we demonstrated that in cultured rat serotonergic neurons, MDMA exposure induced LC3B-densely stained autophagosome formation, accompanying by a decrease in neurite outgrowth. Autophagy inhibitor 3-methyladenine (3-MA) significantly attenuated MDMA-induced autophagosome accumulation, and ameliorated MDMA-triggered serotonergic neurite damage and neuron death. In contrast, enhanced autophagy flux by rapamycin or impaired autophagosome clearance by bafilomycin A1 led to more autophagosome accumulation in serotonergic neurons and aggravated neurite degeneration. In addition, MDMA-induced autophagy activation in cultured serotonergic neurons might be mediated by serotonin transporter (SERT). In an in vivo animal model administered MDMA, neuroimaging showed that 3-MA protected the serotonin system against MDMA-induced downregulation of SERT evaluated by animal-PET with 4-[(18)F]-ADAM, a SERT radioligand. Taken together, our results demonstrated that MDMA triggers upregulation of autophagy in serotonergic neurons, which appears to be detrimental to neuronal growth.
Collapse
Affiliation(s)
- I-Hsun Li
- Department of Pharmacy Practice, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Kuo-Hsing Ma
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Tzu-Jen Kao
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan
| | - Yang-Yi Lin
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Shao-Ju Weng
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Ting-Yin Yen
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Lih-Chi Chen
- Department of Pharmacy, Taipei City Hospital, Taipei, Taiwan
| | - Yuahn-Sieh Huang
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
8
|
Barbosa DJ, Capela JP, Feio-Azevedo R, Teixeira-Gomes A, Bastos MDL, Carvalho F. Mitochondria: key players in the neurotoxic effects of amphetamines. Arch Toxicol 2015; 89:1695-725. [PMID: 25743372 DOI: 10.1007/s00204-015-1478-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 02/09/2015] [Indexed: 12/21/2022]
Abstract
Amphetamines are a class of psychotropic drugs with high abuse potential, as a result of their stimulant, euphoric, emphathogenic, entactogenic, and hallucinogenic properties. Although most amphetamines are synthetic drugs, of which methamphetamine, amphetamine, and 3,4-methylenedioxymethamphetamine ("ecstasy") represent well-recognized examples, the use of natural related compounds, namely cathinone and ephedrine, has been part of the history of humankind for thousands of years. Resulting from their amphiphilic nature, these drugs can easily cross the blood-brain barrier and elicit their well-known psychotropic effects. In the field of amphetamines' research, there is a general consensus that mitochondrial-dependent pathways can provide a major understanding concerning pathological processes underlying the neurotoxicity of these drugs. These events include alterations on tricarboxylic acid cycle's enzymes functioning, inhibition of mitochondrial electron transport chain's complexes, perturbations of mitochondrial clearance mechanisms, interference with mitochondrial dynamics, as well as oxidative modifications in mitochondrial macromolecules. Additionally, other studies indicate that amphetamines-induced neuronal toxicity is closely regulated by B cell lymphoma 2 superfamily of proteins with consequent activation of caspase-mediated downstream cell death pathway. Understanding the molecular mechanisms at mitochondrial level involved in amphetamines' neurotoxicity can help in defining target pathways or molecules mediating these effects, as well as in developing putative therapeutic approaches to prevent or treat the acute- or long-lasting neuropsychiatric complications seen in human abusers.
Collapse
Affiliation(s)
- Daniel José Barbosa
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal. .,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal. .,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua do Campo Alegre 823, 4150-180, Porto, Portugal.
| | - João Paulo Capela
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal.,FP-ENAS (Unidade de Investigação UFP em energia, Ambiente e Saúde), CEBIMED (Centro de Estudos em Biomedicina), Faculdade de Ciências da Saúde, Universidade Fernando Pessoa, Rua 9 de Abril 349, 4249-004, Porto, Portugal
| | - Rita Feio-Azevedo
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Armanda Teixeira-Gomes
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Maria de Lourdes Bastos
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Félix Carvalho
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal.
| |
Collapse
|
9
|
Herndon JM, Cholanians AB, Lau SS, Monks TJ. Glial cell response to 3,4-(+/-)-methylenedioxymethamphetamine and its metabolites. Toxicol Sci 2013; 138:130-8. [PMID: 24299738 DOI: 10.1093/toxsci/kft275] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
3,4-(±)-Methylenedioxymethamphetamine (MDMA) and 3,4-(±)-methylenedioxyamphetamine (MDA), a primary metabolite of MDMA, are phenylethylamine derivatives that cause serotonergic neurotoxicity. Although several phenylethylamine derivatives activate microglia, little is known about the effects of MDMA on glial cells, and evidence of MDMA-induced microglial activation remains ambiguous. We initially determined microglial occupancy status of the parietal cortex in rats at various time points following a single neurotoxic dose of MDMA (20mg/kg, SC). A biphasic microglial response to MDMA was observed, with peak microglial occupancy occurring 12- and 72-h post-MDMA administration. Because direct injection of MDMA into the brain does not produce neurotoxicity, the glial response to MDMA metabolites was subsequently examined in vivo and in vitro. Rats were treated with MDA (20mg/kg, SC) followed by ex vivo biopsy culture to determine the activation of quiescent microglia. A reactive microglial response was observed 72 h after MDA administration that subsided by 7 days. In contrast, intracerebroventricular (ICV) administration of MDA failed to produce a microglial response. However, thioether metabolites of MDA derived from α-methyldopamine (α-MeDA) elicited a robust microglial response following icv injection. We subsequently determined the direct effects of various MDMA metabolites on primary cultures of E18 hippocampal mixed glial and neuronal cells. 5-(Glutathion-S-yl)-α-MeDA, 2,5-bis-(glutathion-S-yl)-α-MeDA, and 5-(N-acetylcystein-S-yl)-α-MeDA all stimulated the proliferation of glial fibrillary acidic protein-positive astrocytes at a dose of 10 µM. The findings indicate that glial cells are activated in response to MDMA/MDA and support a role for thioether metabolites of α-MeDA in the neurotoxicity.
Collapse
Affiliation(s)
- Joseph M Herndon
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721
| | | | | | | |
Collapse
|
10
|
Cuyas E, Robledo P, Pizarro N, Farré M, Puerta E, Aguirre N, de la Torre R. 3,4-methylenedioxymethamphetamine induces gene expression changes in rats related to serotonergic and dopaminergic systems, but not to neurotoxicity. Neurotox Res 2013; 25:161-9. [PMID: 23949956 DOI: 10.1007/s12640-013-9416-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 07/16/2013] [Accepted: 07/30/2013] [Indexed: 11/24/2022]
Abstract
3,4-Methylenedioxymethamphetamine (MDMA, ecstasy) is an amphetamine derivative widely abused by young adults. Although many studies have reported that relatively high doses of MDMA deplete serotonin (5-HT) content and decrease the availability of serotonin transporters (5-HTT), limited evidence is available as to the adaptive mechanisms taking place in gene expression levels in the brain following a dosing regimen of MDMA comparable to human consumption. In order to further clarify this issue, we used quantitative PCR to study the long-term changes induced by acute administration of MDMA (5 mg/kg × 3) in the expression of genes related to serotonergic and dopaminergic systems, as well as those related to cellular toxicity in the cortex, hippocampus, striatum, and brain stem of rats. Seven days after MDMA administration, we found a significantly lower expression of the 5-HTT (Slc6a4) and the vesicular monoamine transporter (Slc18a2) genes in the brain stem area. In the hippocampus, monoamine oxidase B (Maob) and tryptophan hydroxylase 2 (Tph2) gene expressions were increased. In the striatum, tyrosine hydroxylase (Th) expression was decreased, and a lower expression of α-synuclein (Snca) was observed in the cortex. In contrast, no significant changes were observed in the genes considered to be biomarkers of toxicity including the glial fibrillary acidic protein (Gfap) and the heat-shock 70 kD protein 1A (Hspa1a) in any of the structures assayed. These results suggest that MDMA promotes adaptive changes in genes related to serotonergic and dopaminergic functionality, but not in genes related to neurotoxicity.
Collapse
Affiliation(s)
- Elisabet Cuyas
- Human Pharmacology and Clinical Neurosciences Research Group, Neurosciences Research Program, IMIM-Hospital del Mar Medical Research Institute, Parc de Recerca Biomèdica de Barcelona, Doctor Aiguader, 88, 08003, Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
11
|
Huff C, Bhide N, Schroering A, Yamamoto BK, Gudelsky GA. Effect of repeated exposure to MDMA on the function of the 5-HT transporter as assessed by synaptosomal 5-HT uptake. Brain Res Bull 2013; 91:52-7. [PMID: 23318273 DOI: 10.1016/j.brainresbull.2013.01.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 01/04/2013] [Accepted: 01/07/2013] [Indexed: 11/28/2022]
Abstract
Recent studies have demonstrated that a preconditioning regimen (i.e., repeated low doses) of MDMA provides protection against the reductions in tissue concentrations of 5-HT and 5-HT transporter (SERT) density and/or expression produced by a subsequent binge regimen of MDMA. In the present study, the effects of preconditioning and binge treatment regimens of MDMA on SERT function were assessed by synaptosomal 5-HT uptake. Synaptosomal 5-HT uptake was reduced by 72% 7 days following the binge regimen (10 mg/kg, i.p. every 2 h for a total of 4 injections). In rats exposed to the preconditioning regimen of MDMA (daily treatment with 10 mg/kg for 4 days), the reduction in synaptosomal 5-HT uptake induced by a subsequent binge regimen was significantly less. Treatment with the preconditioning regimen alone resulted in a transient 46% reduction in 5-HT uptake that was evident 1 day, but not 7 days, following the last injection of MDMA. Furthermore, the preconditioning regimen of MDMA did not alter tissue concentrations of 5-HT, whereas the binge regimen of MDMA resulted in a long-term reduction of 40% of tissue 5-HT concentrations. The distribution of SERT immunoreactivity (ir) in membrane and endosomal fractions of the hippocampus also was evaluated following the preconditioning regimen of MDMA. There was no significant difference in the relative distribution of SERTir between these two compartments in control and preconditioned rats. The results demonstrate that SERT function is transiently reduced in response to a preconditioning regimen of MDMA, while long-term reductions in SERT function occur in response to a binge regimen of MDMA. Moreover, a preconditioning regimen of MDMA provides protection against the long-term reductions in SERT function evoked by a subsequent binge regimen of the drug. It is tempting to speculate that the neuroprotective effect of MDMA preconditioning results from a transient down-regulation in SERT function.
Collapse
Affiliation(s)
- Courtney Huff
- James Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | | | | | | | | |
Collapse
|
12
|
López-Arnau R, Martínez-Clemente J, Pubill D, Escubedo E, Camarasa J. Comparative neuropharmacology of three psychostimulant cathinone derivatives: butylone, mephedrone and methylone. Br J Pharmacol 2013; 167:407-20. [PMID: 22509960 DOI: 10.1111/j.1476-5381.2012.01998.x] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND AND PURPOSE Here, we have compared the neurochemical profile of three new cathinones, butylone, mephedrone and methylone, in terms of their potential to inhibit plasmalemmal and vesicular monoamine transporters. Their interaction with 5-HT and dopamine receptors and their psychostimulant effect was also studied. EXPERIMENTAL APPROACH Locomotor activity was recorded in mice following different doses of cathinones. Monoamine uptake assays were performed in purified rat synaptosomes. Radioligand-binding assays were carried out to assess the affinity of these compounds for monoamine transporters or receptors. KEY RESULTS Butylone, mephedrone and methylone (5-25 mg·kg(-1) ) caused hyperlocomotion, which was prevented with ketanserin or haloperidol. Methylone was the most potent compound inhibiting both [(3) H]5-HT and [(3) H]dopamine uptake with IC(50) values that correlate with its affinity for dopamine and 5-HT transporter. Mephedrone was found to be the cathinone derivative with highest affinity for vesicular monoamine transporter-2 causing the inhibition of dopamine uptake. The affinity of cathinones for 5-HT(2A) receptors was similar to that of MDMA. CONCLUSIONS AND IMPLICATIONS Butylone and methylone induced hyperlocomotion through activating 5-HT(2A) receptors and increasing extra-cellular dopamine. They inhibited 5-HT and dopamine uptake by competing with substrate. Methylone was the most potent 5-HT and dopamine uptake inhibitor and its effect partly persisted after withdrawal. Mephedrone-induced hyperlocomotion was dependent on endogenous 5-HT. Vesicular content played a key role in the effect of mephedrone, especially for 5-HT uptake inhibition. The potency of mephedrone in inhibiting noradrenaline uptake suggests a sympathetic effect of this cathinone.
Collapse
Affiliation(s)
- Raul López-Arnau
- Department of Pharmacology and Therapeutic Chemistry (Pharmacology Section) and Institute of Biomedicine (IBUB), University of Barcelona, Barcelona, Spain
| | | | | | | | | |
Collapse
|
13
|
Scheidweiler KB, Ladenheim B, Barnes AJ, Cadet JL, Huestis MA. (±)-3,4-methylenedioxymethamphetamine and metabolite disposition in plasma and striatum of wild-type and multidrug resistance protein 1a knock-out mice. J Anal Toxicol 2012; 35:470-80. [PMID: 21871156 DOI: 10.1093/anatox/35.7.470] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mice lacking multidrug resistance protein 1a (mdr1a) are protected from methylenedioxymethamphetamine (MDMA)-induced neurotoxicity, suggesting mdr1a might play an important role in this phenomenon. We characterized MDMA pharmacokinetics in murine plasma and brain to determine if mdr1a alters MDMA distribution. Wild-type (mdr1a⁺/⁺) and mdr1a knock-out (mdr1a⁻/⁻) mice received i.p. 10, 20 or 40 mg/kg MDMA. Plasma and brain specimens were collected 0.3-4 h after MDMA, and striatum were dissected. MDMA and metabolites were quantified in plasma and striatum by gas chromatography-mass spectrometry. MDMA maximum plasma concentrations (C(max)) for both strains were 916- 1363, 1833-3546, and 5979-7948 μg/L, whereas brain C(max) were 6673-14,869, 23,428-29,433, and 52,735-66,525 μg/kg after 10, 20, or 40 mg/kg MDMA, respectively. MDMA and metabolite striatum/plasma AUC ratios were similar in both strains, inconsistent with observed MDMA neuroprotective effects in mdr1a⁻/⁻ mice. Ratios of methylenedioxyamphetamine (MDA) and 4-hydroxy-3-methoxymethamphetamine (HMMA) AUCs exceeded 18% of MDMA's in plasma, suggesting substantial MDMA hepatic metabolism in mice. MDMA, MDA, HMMA, and 4-hydroxy-3-methoxyamphetamine maximum concentrations and AUCs exhibited nonlinear relationships during dose-escalation studies, consistent with impaired enzymatic demethylenation. Nonlinear increases in MDMA plasma and brain concentrations with increased MDMA dose may potentiate MDMA effects and toxicity.
Collapse
Affiliation(s)
- Karl B Scheidweiler
- Chemistry and Drug Metabolism Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Biomedical Research Center, 251 Bayview Boulevard, Baltimore, Maryland 21224, USA
| | | | | | | | | |
Collapse
|
14
|
Martinez CM, Neudörffer A, Largeron M. A convenient biomimetic synthesis of optically active putative neurotoxic metabolites of MDMA (“ecstasy”) from R-(−)- and S-(+)-N-methyl-α-methyldopamine precursors. Org Biomol Chem 2012; 10:3739-48. [DOI: 10.1039/c2ob25245g] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
15
|
Escubedo E, Abad S, Torres I, Camarasa J, Pubill D. Comparative neurochemical profile of 3,4-methylenedioxymethamphetamine and its metabolite alpha-methyldopamine on key targets of MDMA neurotoxicity. Neurochem Int 2011; 58:92-101. [DOI: 10.1016/j.neuint.2010.11.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Revised: 08/31/2010] [Accepted: 11/03/2010] [Indexed: 10/18/2022]
|
16
|
Gandy MN, McIldowie M, Lewis K, Wasik AM, Salomonczyk D, Wagg K, Millar ZA, Tindiglia D, Huot P, Johnston T, Thiele S, Nguyen B, Barnes NM, Brotchie JM, Martin-Iverson MT, Nash J, Gordon J, Piggott MJ. Redesigning the designer drug ecstasy: non-psychoactive MDMA analogues exhibiting Burkitt's lymphoma cytotoxicity. MEDCHEMCOMM 2010. [DOI: 10.1039/c0md00108b] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
17
|
Scheidweiler KB, Ladenheim B, Cadet JL, Huestis MA. Mice lacking multidrug resistance protein 1a show altered dopaminergic responses to methylenedioxymethamphetamine (MDMA) in striatum. Neurotox Res 2009; 18:200-9. [PMID: 19851718 DOI: 10.1007/s12640-009-9124-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Revised: 08/04/2009] [Accepted: 10/07/2009] [Indexed: 01/16/2023]
Abstract
Multidrug resistance protein 1a (MDR1a) potentiated methylenedioxymethamphetamine (MDMA)-induced decreases of dopamine (DA) and dopamine transport protein in mouse brain one week after MDMA administration. In the present study, we examined if mdr1a wild-type (mdr1a +/+) and knock-out (mdr1a -/-) mice differentially handle the acute effects of MDMA on the nigrostriatal DA system 0-24 h following a single drug injection. 3-way ANOVA revealed significant 2-way interactions of strain x time (F (5,152) = 32.4, P < 0.001) and strain x dose (F (3,152) = 25.8, P < 0.001) on 3,4-dihydroxyphenylacetic acid (DOPAC)/DA ratios in mdr1a +/+ and -/- mice. 0.3-3 h after 10 mg/kg MDMA, DOPAC/DA ratios were increased in mdr1a +/+ mice, but decreased 0.3-1 h after MDMA in mdr1a -/- mice. Twenty-four hours after 10 mg/kg MDMA, DOPAC/DA ratios were increased 600% in mdr1a +/+ mice compared to saline-treated control mice, while in mdr1a -/- mice DOPAC/DA ratios were unchanged. Striatal MDMA and its metabolite, methylenedioxyamphetamine, concentrations by gas chromatography-mass spectrometry were similar in both strains 0.3-4 h after MDMA, discounting the role of MDR1a-facilitated MDMA transport in observed inter-strain differences. Increased DOPAC/DA turnover in mdr1a +/+ mice following MDMA is consistent with the previous report that MDMA neurotoxicity is increased in mdr1a +/+ mice. Increased DA turnover via monoamine oxidase in mdr1a +/+ vs -/- mice might increase exposure to neurotoxic reactive oxygen species.
Collapse
Affiliation(s)
- Karl B Scheidweiler
- Chemistry and Drug Metabolism, Intramural Research Program, National Institutes of Health, Biomedical Research Center, 251 Bayview Boulevard Suite 200, Room 05A-721, Baltimore, MD 21224, USA
| | | | | | | |
Collapse
|
18
|
Meyer MR, Peters FT, Maurer HH. Investigations on the human hepatic cytochrome P450 isozymes involved in the metabolism of 3,4-methylenedioxy-amphetamine (MDA) and benzodioxolyl-butanamine (BDB) enantiomers. Toxicol Lett 2009; 190:54-60. [DOI: 10.1016/j.toxlet.2009.06.866] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2009] [Revised: 06/23/2009] [Accepted: 06/25/2009] [Indexed: 10/20/2022]
|
19
|
Bhide NS, Lipton JW, Cunningham JI, Yamamoto BK, Gudelsky GA. Repeated exposure to MDMA provides neuroprotection against subsequent MDMA-induced serotonin depletion in brain. Brain Res 2009; 1286:32-41. [PMID: 19555677 DOI: 10.1016/j.brainres.2009.06.042] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2008] [Revised: 06/12/2009] [Accepted: 06/15/2009] [Indexed: 11/26/2022]
Abstract
Repeated exposure to sub-lethal insults has been reported to result in neuroprotection against a subsequent deleterious insult. The purpose of this study was to evaluate whether repeated exposure (preconditioning) to a non-5-HT depleting dose of MDMA in adult rats provides neuroprotection against subsequent MDMA-induced 5-HT depletion. Treatment of rats with MDMA (10 mg/kg, ip every 2 h for 4 injections) resulted in a 50-65% depletion of 5-HT in the striatum, hippocampus and cortex, and these depletions were significantly attenuated in rats that received a preconditioning regimen of MDMA (10 mg/kg, ip daily for 4 days). The 5-HT depleting regimen of MDMA also resulted in a 40-80% reduction in 5-HT transporter immunoreactivity (SERT(ir)), and the reduction in SERT(ir) also was completely attenuated in MDMA-preconditioned animals. Preconditioning with MDMA (10 mg/kg, ip) daily for 4 days provided neuroprotection against methamphetamine-induced 5-HT depletion, but not dopamine depletion, in the striatum. Additional studies were conducted to exclude the possibility that alterations in MDMA pharmacokinetics or MDMA-induced hyperthermia in rats previously exposed to MDMA contribute towards neuroprotection. During the administration of the 5-HT depleting regimen of MDMA, there was no difference in the extracellular concentration of the drug in the striatum of rats that had received 4 prior, daily injections of vehicle or MDMA. Moreover, there was no difference in the hyperthermic response to the 5-HT depleting regimen of MDMA in rats that had earlier received 4 daily injections of vehicle or MDMA. Furthermore, hyperthermia induced by MDMA during preconditioning appears not to contribute towards neuroprotection, inasmuch as preconditioning with MDMA at a low ambient temperature at which hyperthermia was absent did not alter the neuroprotection provided by the preconditioning regimen. Thus, prior exposure to MDMA affords protection against the long-term depletion of brain 5-HT produced by subsequent MDMA administration. The mechanisms underlying preconditioning-induced neuroprotection for MDMA remain to be determined.
Collapse
Affiliation(s)
- Nirmal S Bhide
- College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267, USA
| | | | | | | | | |
Collapse
|
20
|
Capela JP, Carmo H, Remião F, Bastos ML, Meisel A, Carvalho F. Molecular and Cellular Mechanisms of Ecstasy-Induced Neurotoxicity: An Overview. Mol Neurobiol 2009; 39:210-71. [DOI: 10.1007/s12035-009-8064-1] [Citation(s) in RCA: 210] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Accepted: 02/27/2009] [Indexed: 11/29/2022]
|
21
|
Perfetti X, O'Mathúna B, Pizarro N, Cuyàs E, Khymenets O, Almeida B, Pellegrini M, Pichini S, Lau SS, Monks TJ, Farré M, Pascual JA, Joglar J, de la Torre R. Neurotoxic thioether adducts of 3,4-methylenedioxymethamphetamine identified in human urine after ecstasy ingestion. Drug Metab Dispos 2009; 37:1448-55. [PMID: 19349378 DOI: 10.1124/dmd.108.026393] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
3,4-Methylenedioxymethamphetamine (MDMA, Ecstasy) is a widely misused synthetic amphetamine derivative and a serotonergic neurotoxicant in animal models and possibly humans. The underlying mechanism of neurotoxicity involves the formation of reactive oxygen species although their source remains unclear. It has been postulated that MDMA-induced neurotoxicity is mediated via the formation of bioreactive metabolites. In particular, the primary catechol metabolites, 3,4-dihydroxymethamphetamine (HHMA) and 3,4-dihydroxyamphetamine (HHA), subsequently cause the formation of glutathione and N-acetylcysteine conjugates, which retain the ability to redox cycle and are serotonergic neurotoxicants in rats. Although the presence of such metabolites has been recently demonstrated in rat brain microdialysate, their formation in humans has not been reported. The present study describes the detection of 5-(N-acetylcystein-S-yl)-3,4-dihydroxymethamphetamine (N-Ac-5-Cys-HHMA) and 5-(N-acetylcystein-S-yl)-3,4-dihydroxyamphetamine (N-Ac-5-Cys-HHA) in human urine of 15 recreational users of MDMA (1.5 mg/kg) in a controlled setting. The results reveal that in the first 4 h after MDMA ingestion approximately 0.002% of the administered dose was recovered as thioether adducts. Genetic polymorphisms in CYP2D6 and catechol-O-methyltransferase expression, the combination of which are major determinants of steady-state levels of HHMA and 4-hydroxy-3-methoxyamphetamine, probably explain the interindividual variability seen in the recovery of N-Ac-5-Cys-HHMA and N-Ac-5-Cys-HHA. In summary, the formation of neurotoxic thioether adducts of MDMA has been demonstrated for the first time in humans. The findings lend weight to the hypothesis that the bioactivation of MDMA to neurotoxic metabolites is a relevant pathway to neurotoxicity in humans.
Collapse
Affiliation(s)
- Ximena Perfetti
- Human Pharmacology and Clinical Neurosciences Research Group, Institut Municipal d'Investigació Mèdica-Hospital del Mar, Dr. Aiguader 88, Barcelona, Spain 08003
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Schilt T, Koeter MWJ, de Win MML, Zinkstok JR, van Amelsvoort TA, Schmand B, den Brink WV. The effect of Ecstasy on memory is moderated by a functional polymorphism in the cathechol-O-methyltransferase (COMT) gene. Eur Neuropsychopharmacol 2009; 19:116-24. [PMID: 19042106 DOI: 10.1016/j.euroneuro.2008.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2008] [Revised: 10/20/2008] [Accepted: 10/28/2008] [Indexed: 10/21/2022]
Abstract
There is ample evidence for decreased verbal memory in heavy Ecstasy users. However, findings on the presence of a dose-response relation are inconsistent, possibly due to individual differences in genetic vulnerability. Catechol-O-methyltransferase (COMT) is involved in the catabolism of Ecstasy. Therefore, COMT gene polymorphisms may moderate this vulnerability. We prospectively assessed verbal memory in subjects with a high risk for future Ecstasy use, and compared 59 subjects after first Ecstasy use with 60 subjects that remained Ecstasy-naive. In addition, we tested the interaction effect of Ecstasy and the functional val (158)met polymorphism on verbal memory. Met-allele carriers were somewhat more sensitive to the effects of Ecstasy on verbal learning than homozygous val-subjects. After correction for the use of other substances this effect was no longer statistically significant. The findings suggest that the COMT gene moderates the negative effect of Ecstasy on memory, but also other drug use seems to play a role.
Collapse
Affiliation(s)
- Thelma Schilt
- Amsterdam Institute for Addiction Research, Department of Psychiatry, Academic Medical Center of the University of Amsterdam, 1070 AW Amsterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
23
|
Thibaut R, Porte C. Effects of fibrates, anti-inflammatory drugs and antidepressants in the fish hepatoma cell line PLHC-1: Cytotoxicity and interactions with cytochrome P450 1A. Toxicol In Vitro 2008; 22:1128-35. [DOI: 10.1016/j.tiv.2008.02.020] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2007] [Revised: 01/28/2008] [Accepted: 02/27/2008] [Indexed: 11/26/2022]
|
24
|
Hatcher JM, Delea KC, Richardson JR, Pennell KD, Miller GW. Disruption of dopamine transport by DDT and its metabolites. Neurotoxicology 2008; 29:682-90. [PMID: 18533268 PMCID: PMC4755343 DOI: 10.1016/j.neuro.2008.04.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2007] [Revised: 04/03/2008] [Accepted: 04/04/2008] [Indexed: 12/21/2022]
Abstract
Epidemiological studies suggest a link between pesticide exposure and an increased risk of developing Parkinson's disease (PD). Although studies have been unable to clearly identify specific pesticides that contribute to PD, a few human studies have reported higher levels of the organochlorine pesticides dieldrin and DDE (a metabolite of DDT) in post-mortem PD brains. Previously, we found that exposure of mice to dieldrin caused perturbations in the nigrostriatal dopamine system consistent with those seen in PD. Given the concern over the environmental persistence and reintroduction of DDT for the control of malaria-carrying mosquitoes and other pests, we sought to determine whether DDT and its two major metabolites, DDD and DDE, could damage the dopamine system. In vitro analyses in mouse synaptosomes and vesicles demonstrated that DDT and its metabolites inhibit the plasma membrane dopamine transporter (DAT) and the vesicular monoamine transporter (VMAT2). However, exposure of mice to either DDT or DDE failed to show evidence of nigrostriatal damage or behavioral abnormalities in any of the measures examined. Thus, we report that in vitro effects of DDT and its metabolites on components of the dopamine system do not translate into neurotoxicological outcomes in orally exposed mice and DDT appears to have less dopamine toxicity when compared to dieldrin. These data suggest elevated DDE levels in PD patients may represent a measure of general pesticide exposure and that other pesticides may be responsible for the association between pesticide exposure and PD.
Collapse
Affiliation(s)
- Jaime M. Hatcher
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, United States
- Department of Environmental and Occupational Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Kristin C. Delea
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, United States
- Department of Environmental and Occupational Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Jason R. Richardson
- Environmental and Occupational Health Sciences Institute and Department of Environmental and Occupational Medicine, University of Medicine and Dentistry-New Jersey/Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Kurt D. Pennell
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, United States
- School of Civil and Environmental Engineering, Georgia Institute of Technology, Atlanta, GA, United States
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, United States
| | - Gary W. Miller
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, United States
- Department of Environmental and Occupational Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| |
Collapse
|
25
|
Goni-Allo B, O Mathúna B, Segura M, Puerta E, Lasheras B, de la Torre R, Aguirre N. The relationship between core body temperature and 3,4-methylenedioxymethamphetamine metabolism in rats: implications for neurotoxicity. Psychopharmacology (Berl) 2008; 197:263-78. [PMID: 18074122 DOI: 10.1007/s00213-007-1027-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2007] [Accepted: 11/20/2007] [Indexed: 11/28/2022]
Abstract
RATIONALE A close relationship appears to exist between 3,4-methylenedioxymethamphetamine (MDMA)-induced changes in core body temperature and long-term serotonin (5-HT) loss. OBJECTIVE We investigated whether changes in core body temperature affect MDMA metabolism. MATERIALS AND METHODS Male Wistar rats were treated with MDMA at ambient temperatures of 15, 21.5, or 30 degrees C to prevent or exacerbate MDMA-induced hyperthermia. Plasma concentrations of MDMA and its main metabolites were determined for 6 h. Seven days later, animals were killed and brain indole content was measured. RESULTS The administration of MDMA at 15 degrees C blocked the hyperthermic response and long-term 5-HT depletion found in rats treated at 21.5 degrees C. At 15 degrees C, plasma concentrations of MDMA were significantly increased, whereas those of three of its main metabolites were reduced when compared to rats treated at 21.5 degrees C. By contrast, hyperthermia and indole deficits were exacerbated in rats treated at 30 degrees C. Noteworthy, plasma concentrations of MDMA metabolites were greatly enhanced in these animals. Instrastriatal perfusion of MDMA (100 microM for 5 h at 21 degrees C) did not potentiate the long-term depletion of 5-HT after systemic MDMA. Furthermore, interfering in MDMA metabolism using the catechol-O-methyltransferase inhibitor entacapone potentiated the neurotoxicity of MDMA, indicating that metabolites that are substrates for this enzyme may contribute to neurotoxicity. CONCLUSIONS This is the first report showing a direct relationship between core body temperature and MDMA metabolism. This finding has implications on both the temperature dependence of the mechanism of MDMA neurotoxicity and human use, as hyperthermia is often associated with MDMA use in humans.
Collapse
Affiliation(s)
- Beatriz Goni-Allo
- Department of Pharmacology, School of Medicine, University of Navarra, c/ Irunlarrea 1, 31008 Pamplona, Spain
| | | | | | | | | | | | | |
Collapse
|
26
|
Goñi-Allo B, Puerta E, Mathúna BO, Hervias I, Lasheras B, de la Torre R, Aguirre N. On the role of tyrosine and peripheral metabolism in 3,4-methylenedioxymethamphetamine-induced serotonin neurotoxicity in rats. Neuropharmacology 2008; 54:885-900. [PMID: 18329670 DOI: 10.1016/j.neuropharm.2008.01.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2007] [Revised: 01/11/2008] [Accepted: 01/24/2008] [Indexed: 11/25/2022]
Abstract
The mechanisms underlying 3,4-methylenedioxymethamphetamine (MDMA)-induced serotonergic (5-HT) toxicity remain unclear. It has been suggested that MDMA depletes 5-HT by increasing brain tyrosine levels, which via non-enzymatic hydroxylation leads to DA-derived free radical formation. Because this hypothesis assumes the pre-existence of hydroxyl radicals, we hypothesized that MDMA metabolism into pro-oxidant compounds is the limiting step in this process. Acute hyperthermia, plasma tyrosine levels and concentrations of MDMA and its main metabolites were higher after a toxic (15 mg/kg i.p.) vs. a non-toxic dose of MDMA (7.5mg/kg i.p.). The administration of a non-toxic dose of MDMA in combination with l-tyrosine (0.2 mmol/kg i.p.) produced a similar increase in serum tyrosine levels to those found after a toxic dose of MDMA; however, brain 5-HT content remained unchanged. The non-toxic dose of MDMA combined with a high dose of tyrosine (0.5 mmol/kg i.p.), caused long-term 5-HT depletions in rats treated at 21.5 degrees C but not in those treated at 15 degrees C, conditions known to decrease MDMA metabolism. Furthermore, striatal perfusion of MDMA (100 microM for 5h) combined with tyrosine (0.5 mmol/kg i.p.) in hyperthermic rats did not cause 5-HT depletions. By contrast, rats treated with the non-toxic dose of MDMA under heating conditions or combined with entacapone or acivicin, which interfere with MDMA metabolism or increase brain MDMA metabolite availability respectively, showed significant reductions of brain 5-HT content. Altogether, these data indicate that although tyrosine may contribute to MDMA-induced toxicity, MDMA metabolism appears to be the limiting step.
Collapse
Affiliation(s)
- Beatriz Goñi-Allo
- Department of Pharmacology, School of Medicine, University of Navarra, c/ Irunlarrea 1, 31008 Pamplona, Spain
| | | | | | | | | | | | | |
Collapse
|
27
|
Alves E, Summavielle T, Alves CJ, Gomes-da-Silva J, Barata JC, Fernandes E, de Lourdes Bastos M, Tavares MA, Carvalho F. Monoamine oxidase-B mediates ecstasy-induced neurotoxic effects to adolescent rat brain mitochondria. J Neurosci 2007; 27:10203-10. [PMID: 17881526 PMCID: PMC6672671 DOI: 10.1523/jneurosci.2645-07.2007] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
3,4-Methylenedioxymethamphetamine (MDMA)-induced neurotoxicity and the protective role of monoamine oxidase-B (MAO-B) inhibition were evaluated at the mitochondrial level in various regions of the adolescent rat brain. Four groups of adolescent male Wistar rats were used: (1) saline control, (2) exposed to MDMA (4 x 10 mg/kg, i.p.; two hourly), (3) treated with selegiline (2 mg/kg, i.p.) 30 min before the same dosing of MDMA, and (4) treated with selegiline (2 mg/kg, i.p.). Body temperatures were monitored throughout the whole experiment. Animals were killed 2 weeks later, and mitochondria were isolated from several brain regions. Our results showed that "binge" MDMA administration causes, along with sustained hyperthermia, long-term alterations in brain mitochondria as evidenced by increased levels of lipid peroxides and protein carbonyls. Additionally, analysis of mitochondrial DNA (mtDNA) revealed that NDI nicotinamide adenine dinucleotide phosphate dehydrogenase subunit I and NDII (nicotinamide adenine dinucleotide phosphate dehydrogenase subunit II) subunits of mitochondrial complex I and cytochrome c oxidase subunit I of complex IV suffered deletions in MDMA-exposed animals. Inhibition of MAO-B by selegiline did not reduce hyperthermia but reversed MDMA-induced effects in the oxidative stress markers, mtDNA, and related protein expression. These results indicate that monoamine oxidation by MAO-B with subsequent mitochondrial damage may be an important contributing factor for MDMA-induced neurotoxicity.
Collapse
Affiliation(s)
- Ema Alves
- Neurobehaviour Unit, Instituto de Biologia Molecular e Celular
- Toxicology Department, Faculty of Pharmacy, REQUIMTE
| | - Teresa Summavielle
- Neurobehaviour Unit, Instituto de Biologia Molecular e Celular
- Departamento de Ciências Biomédicas, Escola Superior de Tecnologia da Saúde, Instituto Politécnico do Porto, 4000-294 Porto, Portugal
| | | | - Joana Gomes-da-Silva
- Neurobehaviour Unit, Instituto de Biologia Molecular e Celular
- Escola de Saúde, University of Aveiro, 3180-193 Aveiro, Portugal, and
| | - José Custódio Barata
- Biochemistry Department, Faculty of Pharmacy, University of Coimbra, 3000-295 Coimbra, Portugal
| | | | | | - Maria Amélia Tavares
- Neurobehaviour Unit, Instituto de Biologia Molecular e Celular
- Institute of Anatomy, Medical School of Porto, University of Porto, 4099-002 Porto, Portugal
| | | |
Collapse
|
28
|
Erives GV, Lau SS, Monks TJ. Accumulation of Neurotoxic Thioether Metabolites of 3,4-(±)-Methylenedioxymethamphetamine in Rat Brain. J Pharmacol Exp Ther 2007; 324:284-91. [PMID: 17906065 DOI: 10.1124/jpet.107.128785] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The serotonergic neurotoxicity of 3,4-(+/-)-methylenedioxymethamphetamine (MDMA) appears dependent upon systemic metabolism because direct injection of MDMA into the brain fails to reproduce the neurotoxicity. MDMA is demethylenated to the catechol metabolite N-methyl-alpha-methyldopamine (N-Me-alpha-MeDA). Thioether (glutathione and N-acetylcysteine) metabolites of N-Me-alpha-MeDA are neurotoxic and are present in rat brain following s.c. injection of MDMA. Because multidose administration of MDMA is typical of drug intake during rave parties, the present study was designed to determine the effects of multiple doses of MDMA on the concentration of neurotoxic thioether metabolites in rat brain. Administration of MDMA (20 mg/kg s.c.) at 12-h intervals for a total of four injections led to a significant accumulation of the N-Me-alpha-MeDA thioether metabolites in striatal dialysate. The area under the curve (AUC)(0-300 min) for 5-(glutathion-S-yl)-N-Me-alpha-MeDA increased 33% between the first and fourth injections and essentially doubled for 2,5-bis-(glutathion-S-yl)-N-Me-alpha-MeDA. Likewise, accumulation of the mercapturic acid metabolites was reflected by increases in the AUC(0-300 min) for both 5-(N-acetylcystein-S-yl)-N-Me-alpha-MeDA (35%) and 2,5-bis-(N-acetylcystein-S-yl)-N-Me-alpha-MeDA (85%), probably because processes for their elimination become saturated. Indeed, the elimination half-life of 5-(N-acetylcystein-S-yl)-N-Me-alpha-MeDA and 2,5-bis-(N-acetylcystein-S-yl)-N-Me-alpha-MeDA increased by 53 and 28%, respectively, between the first and third doses. Finally, although the C(max) values for the monothioether conjugates were essentially unchanged after each injection, the values increased by 38 and approximately 50% for 2,5-bis-(glutathion-S-yl)-N-Me-alpha-MeDA and 2,5-bis-(N-acetylcystein-S-yl)-N-Me-alpha-MeDA, respectively, between the first and fourth injections. The data indicate that neurotoxic metabolites of MDMA may accumulate in brain after multiple dosing.
Collapse
Affiliation(s)
- Gladys V Erives
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona Health Sciences Center, 1703 E. Mabel Street, Tucson, AZ 85721-0207, USA.
| | | | | |
Collapse
|
29
|
Ben Hamida S, Plute E, Bach S, Lazarus C, Tracqui A, Kelche C, de Vasconcelos AP, Jones BC, Cassel JC. Ethanol-MDMA interactions in rats: the importance of interval between repeated treatments in biobehavioral tolerance and sensitization to the combination. Psychopharmacology (Berl) 2007; 192:555-69. [PMID: 17345065 DOI: 10.1007/s00213-007-0752-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2006] [Accepted: 02/19/2007] [Indexed: 10/23/2022]
Abstract
RATIONALE In our previous work, we showed that ethanol (EtOH) potentiates 3,4-methylenedioxymethamphetamine (MDMA)-induced hyperlocomotion while protecting against its hyperthermic effects. Whereas the effect on activity were found on all days (although declining over the three first days), the protection against hyperthermia completely disappeared on the second day. The latter effect was previously thought to reflect tolerance to ethanol or the combination, per se. OBJECTIVE In the present study, we changed the treatment regimen to irregular and longer intervals between treatments (48, 120, and again 48 h) to check if tolerance was still observed. RESULTS We found progressive sensitization of locomotor activity to EtOH (1.5 g/kg, i.p.)+MDMA (6.6 mg/kg, i.p.), and a partial EtOH protection against MDMA-induced hyperthermia that persisted after the first drug challenge day. When the monoamine neurotransmitters, dopamine, and serotonin were assessed 2 weeks after treatment, we found no consistent effect on the concentration of any of these neurotransmitters, whatever the treatment. Similarly, we found that regional brain concentrations of MDMA were not significantly affected by EtOH at a 45-min post-treatment delay; however, the overall ratio of the metabolite 3,4-methylenedioxyamphetamine (MDA) to MDMA was lower (overall, -16%) in animals treated with the combination compared to MDMA alone, indicating possible contribution of pharmacokinetic factors. This difference was especially marked in the striatum (-25%). CONCLUSIONS These findings shed new light on the consequences of EtOH-MDMA, taken together at a nearly normal ambient temperature, both in terms of motivation and potential risks for recreational drug users.
Collapse
Affiliation(s)
- Sami Ben Hamida
- LINC-UMR 7191, Université Louis Pasteur-CNRS, Institut Fédérératif de Recherche 37, GDR CNRS 2905, Strasbourg, France
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Goñi-Allo B, Puerta E, Hervias I, Di Palma R, Ramos M, Lasheras B, Aguirre N. Studies on the mechanisms underlying amiloride enhancement of 3,4-methylenedioxymethamphetamine-induced serotonin depletion in rats. Eur J Pharmacol 2007; 562:198-207. [PMID: 17320075 DOI: 10.1016/j.ejphar.2007.01.049] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2006] [Revised: 12/13/2006] [Accepted: 01/11/2007] [Indexed: 10/23/2022]
Abstract
Amiloride and several of its congeners known to block the Na(+)/Ca(2+) and/or Na(+)/H(+) antiporters potentiate methamphetamine-induced neurotoxicity without altering methamphetamine-induced hyperthermia. We now examine whether amiloride also exacerbates 3,4-methylenedioxymethamphetamine (MDMA)-induced long-term serotonin (5-HT) loss in rats. Amiloride (2.5 mg/kg, every 2 h x 3, i.p.) given at ambient temperature 30 min before MDMA (5 mg/kg, every 2 h x 3, i.p.), markedly exacerbated long-term 5-HT loss. However, in contrast to methamphetamine, amiloride also potentiated MDMA-induced hyperthermia. Fluoxetine (10 mg/kg i.p.) completely protected against 5-HT depletion caused by the MDMA/amiloride combination without significantly altering the hyperthermic response. By contrast, the calcium channel antagonists flunarizine or diltiazem did not afford any protection. Findings with MDMA and amiloride were extended to the highly selective Na(+)/H(+) exchange inhibitor dimethylamiloride, suggesting that the potentiating effects of amiloride are probably mediated by the blockade of Na(+)/H(+) exchange. When the MDMA/amiloride combination was administered at 15 degrees C hyperthermia did not develop and brain 5-HT concentrations remained unchanged 7 days later. Intrastriatal perfusion of MDMA (100 microM for 8 h) in combination with systemic amiloride caused a small depletion of striatal 5-HT content in animals made hyperthermic but not in the striatum of normothermic rats. These data suggest that enhancement of MDMA-induced 5-HT loss caused by amiloride or dimethylamiloride depends on their ability to enhance MDMA-induced hyperthermia. We hypothesise that blockade of Na(+)/H(+) exchange could synergize with hyperthermia to render 5-HT terminals more vulnerable to the toxic effects of MDMA.
Collapse
Affiliation(s)
- Beatriz Goñi-Allo
- Department of Pharmacology, School of Medicine, University of Navarra, C/ Irunlarrea, 1, 31008, Pamplona, Spain
| | | | | | | | | | | | | |
Collapse
|
31
|
Capela JP, Macedo C, Branco PS, Ferreira LM, Lobo AM, Fernandes E, Remião F, Bastos ML, Dirnagl U, Meisel A, Carvalho F. Neurotoxicity mechanisms of thioether ecstasy metabolites. Neuroscience 2007; 146:1743-57. [PMID: 17467183 DOI: 10.1016/j.neuroscience.2007.03.028] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2007] [Revised: 03/20/2007] [Accepted: 03/22/2007] [Indexed: 10/23/2022]
Abstract
3,4-Methylenedioxymethamphetamine (MDMA or "ecstasy"), is a widely abused, psychoactive recreational drug that is known to induce neurotoxic effects. Human and rat hepatic metabolism of MDMA involves N-demethylation to 3,4-methylenedioxyamphetamine (MDA), which is also a drug of abuse. MDMA and MDA are O-demethylenated to N-methyl-alpha-methyldopamine (N-Me-alpha-MeDA) and alpha-methyldopamine (alpha-MeDA), respectively, which are both catechols that can undergo oxidation to the corresponding ortho-quinones. Ortho-quinones may be conjugated with glutathione (GSH) to form glutathionyl adducts, which can be transported into the brain and metabolized to the correspondent N-acetylcysteine (NAC) adducts. In this study we evaluated the neurotoxicity of nine MDMA metabolites, obtained by synthesis: N-Me-alpha-MeDA, alpha-MeDA and their correspondent GSH and NAC adducts. The studies were conducted in rat cortical neuronal cultures, for a 6 h of exposure period, under normal (36.5 degrees C) and hyperthermic (40 degrees C) conditions. Our findings show that thioether MDMA metabolites are strong neurotoxins, significantly more than their correspondent parent catechols. On the other hand, N-Me-alpha-MeDA and alpha-MeDA are more neurotoxic than MDMA. GSH and NAC conjugates of N-Me-alpha-MeDA and alpha-MeDA induced a concentration dependent delayed neuronal death, accompanied by activation of caspase 3, which occurred earlier in hyperthermic conditions. Furthermore, thioether MDMA metabolites time-dependently increased the production of reactive species, concentration-dependently depleted intracellular GSH and increased protein bound quinones. Finally, thioether MDMA metabolites induced neuronal death and oxidative stress was prevented by NAC, an antioxidant and GSH precursor. This study provides new insights into the neurotoxicity mechanisms of thioether MDMA metabolites and highlights their importance in "ecstasy" neurotoxicity.
Collapse
Affiliation(s)
- J P Capela
- REQUIMTE (Rede de Química e Tecnologia), Toxicology Department, Faculty of Pharmacy, University of Porto, Rua Aníbal Cunha, 164, 4099-030 Porto, Portugal.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Quinton MS, Yamamoto BK. Causes and consequences of methamphetamine and MDMA toxicity. AAPS JOURNAL 2006; 8:E337-47. [PMID: 16796384 PMCID: PMC3231568 DOI: 10.1007/bf02854904] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Methamphetamine (METH) and its derivative 3,4-methylenedioxymethamphetamine (MDMA; ecstasy) are 2 substituted amphetamines with very high abuse liability in the United States. These amphetamine-like stimulants have been associated with loss of multiple markers for dopaminergic and serotonergic terminals in the brain. Among other causes, oxidative stress, excitotoxicity and mitochondrial dysfunction appear to play a major role in the neurotoxicity produced by the substituted amphetamines. The present review will focus on these events and how they interact and converge to produce the monoaminergic depletions that are typically observed after METH or MDMA administration. In addition, more recently identified consequences of METH or MDMA-induced oxidative stress, excitotoxicity, and mitochondrial dysfunction are described in relation to the classical markers of METH-induced damage to dopamine terminals.
Collapse
Affiliation(s)
- Maria S. Quinton
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Room L-613, 715 Albany Street, 02118 Boston, MA
| | - Bryan K. Yamamoto
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Room L-613, 715 Albany Street, 02118 Boston, MA
| |
Collapse
|
33
|
Peterson AL, Gilman TL, Banks ML, Sprague JE. Hypothyroidism alters striatal dopamine release mediated by 3,4-methylenedioxymethamphetamine (MDMA, ecstasy). Synapse 2006; 59:317-9. [PMID: 16421905 DOI: 10.1002/syn.20244] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Microdialysis cannulas were surgically implanted into the striatum of thyroparathyroidectomized (TX) and sham rats in order to determine differences in dopamine release and core body temperature following MDMA administration. Rats were subsequently treated with MDMA (10 mg/kg, s.c.), and striatal DA levels were monitored at 20 min intervals, as well as core temperature every 30 min. Sham rats responded to MDMA with a characteristic hyperthermic response and significant increases in extracellular dopamine. Conversely, TX rats responded to MDMA with a hypothermic response and failed to demonstrate a similar increase from basal dopamine levels. On the basis of these data, thyroid hormones are not only important in the thermogenic effects of MDMA but also appear to have an auxiliary role in MDMA-induced striatal dopamine release.
Collapse
Affiliation(s)
- Andrea L Peterson
- Virginia College of Osteopathic Medicine, Virginia Polytechnic Institute and State University, Blacksburg, 24060, USA
| | | | | | | |
Collapse
|
34
|
Goñi-Allo B, Ramos M, Herv'as I, Lasheras B, Aguirre N. Studies on striatal neurotoxicity caused by the 3,4-methylenedioxymethamphetamine/ malonate combination: implications for serotonin/dopamine interactions. J Psychopharmacol 2006; 20:245-56. [PMID: 16510482 DOI: 10.1177/0269881106063264] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The amphetamine derivative 3,4-methylenedioxymethamphetamine (MDMA) produces long-term toxicity to serotonin (5-HT) neurones in rats, which is exacerbated when combined with the mitochondrial inhibitor malonate. Moreover, MDMA, which does not produce dopamine depletion in the rat, potentiates malonate-induced striatal dopamine toxicity. Because the malonate/MDMA combination acutely causes a synergistic increase of 5-HT and dopamine release, in this study we sought to determine whether pharmacological blockade of MDMA- and/or malonate-induced dopamine release prevents neurotoxicity. Fluoxetine, given 30 min prior to the malonate/MDMA combination, afforded complete protection against 5-HT depletion and reversed MDMA-induced exacerbation of dopamine toxicity found in the malonate/MDMA treated rats. Protection afforded by fluoxetine was not related to changes in MDMA-induced hyperthermia. Similarly, potentiation of malonate-induced dopamine toxicity caused by MDMA was not observed in p-chlorophenylalanine-5-HT depleted rats. Finally, the dopamine transporter inhibitor GBR 12909 completely prevented dopamine neurotoxicity caused by the malonate/MDMA combination and reversed the exacerbating toxic effects of malonate on MDMA-induced 5-HT depletion without significantly altering the hyperthermic response. Overall, these results suggest that the synergic release of dopamine caused by the malonate/MDMA combination plays an important role in the long-term toxic effects. A possible mechanism of neurotoxicity and protection is proposed.
Collapse
Affiliation(s)
- Beatriz Goñi-Allo
- Department of Pharmacology, School of Medicine, University of Navarra, Pamplona, Spain
| | | | | | | | | |
Collapse
|
35
|
Darvesh AS, Gudelsky GA. Evidence for a role of energy dysregulation in the MDMA-induced depletion of brain 5-HT. Brain Res 2005; 1056:168-75. [PMID: 16098955 DOI: 10.1016/j.brainres.2005.07.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2005] [Revised: 07/07/2005] [Accepted: 07/13/2005] [Indexed: 10/25/2022]
Abstract
Although the exact mechanism involved in the long-term depletion of brain serotonin (5-HT) produced by substituted amphetamines is not completely known, evidence suggests that oxidative and/or bioenergetic stress may contribute to 3,4-methylenedioxymethamphetamine (MDMA)-induced 5-HT toxicity. In the present study, the effect of supplementing energy substrates was examined on the long-term depletion of striatal 5-HT and dopamine produced by the local perfusion of MDMA (100 microM) and malonate (100 mM) and the depletion of striatal and hippocampal 5-HT concentrations produced by the systemic administration of MDMA (10 mg/kg i.p. x4). The effect of systemic administration of MDMA on ATP levels in the striatum and hippocampus also was examined. Reverse dialysis of MDMA and malonate directly into the striatum resulted in a 55-70% reduction in striatal concentrations of 5-HT and dopamine, and these reductions were significantly attenuated when MDMA and malonate were co-perfused with nicotinamide (1 mM). Perfusion of nicotinamide or ubiquinone (100 microM) also attenuated the depletion of 5-HT in the striatum and hippocampus produced by the systemic administration of MDMA. Finally, the systemic administration of MDMA produced a 30% decrease in the concentration of ATP in the striatum and hippocampus. These results support the conclusion that MDMA produces a dysregulation of energy metabolism which contributes to the mechanism of MDMA-induced 5-HT neurotoxicity.
Collapse
Affiliation(s)
- Altaf S Darvesh
- College of Pharmacy, University of Cincinnati, 3223, Eden Ave., Cincinnati, OH 45267, USA
| | | |
Collapse
|
36
|
de la Torre R, Farré M, Monks TJ, Jones D. Response to Sprague and Nichols: Contribution of metabolic activation to MDMA neurotoxicity. Trends Pharmacol Sci 2005. [DOI: 10.1016/j.tips.2004.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
37
|
Sprague JE, Nichols DE. Neurotoxicity of MDMA (ecstasy): beyond metabolism. Trends Pharmacol Sci 2005; 26:59-60; author reply 60-1. [PMID: 15681020 DOI: 10.1016/j.tips.2004.12.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
38
|
Jones DC, Duvauchelle C, Ikegami A, Olsen CM, Lau SS, de la Torre R, Monks TJ. Serotonergic Neurotoxic Metabolites of Ecstasy Identified in Rat Brain. J Pharmacol Exp Ther 2005; 313:422-31. [PMID: 15634943 DOI: 10.1124/jpet.104.077628] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The selective serotonergic neurotoxicity of 3,4-methylenedioxyamphetamine (MDA) and 3,4-methylenedioxymethamphetamine (MDMA, ecstasy) depends on their systemic metabolism. We have recently shown that inhibition of brain endothelial cell gamma-glutamyl transpeptidase (gamma-GT) potentiates the neurotoxicity of both MDMA and MDA, indicating that metabolites that are substrates for this enzyme contribute to the neurotoxicity. Consistent with this view, glutathione (GSH) and N-acetylcysteine conjugates of alpha-methyl dopamine (alpha-MeDA) are selective neurotoxicants. However, neurotoxic metabolites of MDMA or MDA have yet to be identified in brain. Using in vivo microdialysis coupled to liquid chromatography-tandem mass spectroscopy and a high-performance liquid chromatography-coulometric electrode array system, we now show that GSH and N-acetylcysteine conjugates of N-methyl-alpha-MeDA are present in the striatum of rats administered MDMA by subcutaneous injection. Moreover, inhibition of gamma-GT with acivicin increases the concentration of GSH and N-acetylcysteine conjugates of N-methyl-alpha-MeDA in brain dialysate, and there is a direct correlation between the concentrations of metabolites in dialysate and the extent of neurotoxicity, measured by decreases in serotonin (5-HT) and 5-hydroxyindole acetic (5-HIAA) levels. Importantly, the effects of acivicin are independent of MDMA-induced hyperthermia, since acivicin-mediated potentiation of MDMA neurotoxicity occurs in the context of acivicin-mediated decreases in body temperature. Finally, we have synthesized 5-(N-acetylcystein-S-yl)-N-methyl-alpha-MeDA and established that it is a relatively potent serotonergic neurotoxicant. Together, the data support the contention that MDMA-mediated serotonergic neurotoxicity is mediated by the systemic formation of GSH and N-acetylcysteine conjugates of N-methyl-alpha-MeDA (and alpha-MeDA). The mechanisms by which such metabolites access the brain and produce selective serotonergic neurotoxicity remain to be determined.
Collapse
Affiliation(s)
- Douglas C Jones
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona Health Sciences Center, Tucson, AZ 85721-0207, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Darvesh AS, Yamamoto BK, Gudelsky GA. Evidence for the Involvement of Nitric Oxide in 3,4-Methylenedioxymethamphetamine-Induced Serotonin Depletion in the Rat Brain. J Pharmacol Exp Ther 2004; 312:694-701. [PMID: 15456837 DOI: 10.1124/jpet.104.074849] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Production of reactive oxygen and/or nitrogen species has been thought to contribute to the long-term depletion of brain dopamine and serotonin (5-HT) produced by amphetamine derivatives, i.e., methamphetamine and 3,4-methylenedioxymethamphetamine (MDMA). In the present study, the effects of nitric-oxide synthase (NOS) inhibitors were examined on the long-term depletion of striatal dopamine and/or 5-HT produced by the local perfusion of malonate and MDMA or the systemic administration of MDMA. The effect of MDMA on nitric oxide formation and nitrotyrosine concentration also was determined. Perfusion with MDMA and malonate resulted in a 34% reduction of 5-HT and 49% reduction of dopamine concentrations in the striatum. The systemic administration of NOS inhibitors, N(omega)-nitro-l-arginine methyl ester hydrochloride and S-methyl-l-thiocitrulline (S-MTC), and the peroxynitrite decomposition catalyst Fe(III) tetrakis (1-methyl-4-pyridyl) porphyrin pentachloride attenuated the MDMA- and malonate-induced depletion of striatal dopamine and 5-HT. S-MTC also attenuated the depletion of 5-HT in the striatum produced by the systemic administration of MDMA without attenuating MDMA-induced hyperthermia. Additionally, the systemic administration of MDMA significantly increased the formation of nitric oxide and the nitrotyrosine concentration in the striatum. These results support the conclusion that MDMA produces reactive nitrogen species in the rat that contribute to the neurotoxicity of this amphetamine analog.
Collapse
Affiliation(s)
- Altaf S Darvesh
- University of Cincinnati, College of Pharmacy, 3223 Eden Ave., Cincinnati, OH 45267, USA
| | | | | |
Collapse
|
40
|
Abstract
Autoxidation pathways and redox reactions of dihydroxytryptamines (5,6- and 5,7-DHT) and of 6-hydroxydopamine (6-OH-DA) are illustrated, and their potential role in aminergic neurotoxicity is discussed. It is proposed that certain aspects of the cytotoxicity of 6-OH-DA and of the DHTs, namely redox cycling of their quinone- and quinoneimine-intermediates as a source of free radicals, may also apply to quinoidal reactive intermediates and to glutathionyl- or cysteinyl conjugates ("thioether adducts") of o-dihydroxylated (catechol-like) metabolites of certain substituted amphetamines (of methylenedioxymethamphetamine (MDMA) and of methylenedioxyamphetamine (MDA)). Despite similarities in their primary interaction with the plasmalemmal (serotonergic transporter/dopamine transporter, SERT/DAT) and vesicular monoamine transporters (VMAT2), MDMA and fenfluramine (N-ethyl-meta-trifluoromethamphetamine, Fen) differ substantially in many aspects of their metabolism, pharmacokinetics, pharmacology, and neurotoxicology profile; the consequences of these differences for neuronal response patterns and long-term survival prospects are not yet fully understood. However, sustained hyperthermia appears to be a critical factor in these differences. Methodological requirements for adequate detection and description of pre- and postsynaptic forms of drug-induced neurotoxicity are exemplified using recently published accounts. The inclusion of microglial markers into research strategies has widened contemporary pathogenetic concepts on methamphetamine (MA)-induced neurotoxicity as an example of inflammatory neurodegeneration, thus complementing the traditional ROS and RNS-dependent stress models. Amphetamine-type neurotoxicity studies may assist in elaborating of preventive strategies for human neurodegenerative disorders.
Collapse
Affiliation(s)
- H G Baumgarten
- Institut für Anatomie, Charite Universitätsmedizin Berlin, Campus Benjamin Franklin, Königin-Luise-Str. 15, 14195 Berlin, Germany.
| | | |
Collapse
|