1
|
Behavioral and Pharmacokinetics Studies of N-Methyl-2-Aminoindane (NM2AI) in Mice: An Aminoindane Briefly Used in the Illicit Drug Market. Int J Mol Sci 2023; 24:ijms24031882. [PMID: 36768197 PMCID: PMC9916073 DOI: 10.3390/ijms24031882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/10/2023] [Accepted: 01/13/2023] [Indexed: 01/20/2023] Open
Abstract
Drug forums are considered as the main platform sources that have contributed to the increase in NPS popularity, especially for those not yet known to law enforcement and therefore not yet illegal. An example is the new synthetic stimulant NM2AI, which has a very short history of human use and abuse. Little is known regarding this compound, but some information from internet forums and the scientific literature indicates NM2AI as a structural derivate of MDAI, which is known for its entactogenic activity. Indeed, the purpose of this study is to evaluate, for the first time, the in vivo acute effect induced by the intraperitoneal injection of NM2AI (1-10-30-100 mg/kg) in mice. We demonstrate the sensory (by visual placing and object tests) and physiological (core temperature measurement) function variations, nociceptor (by tail pinch test) and strength (grip test) alterations, and sensorimotor (time on rod and mobility) decrease. Moreover, we verify the mild hallucinogenic effect of NM2AI (by startle/prepulse inhibition test). Lastly, we perform a pharmacokinetic study on mice blood samples, highlighting that the main active metabolite of NM2AI is 2-aminoindane (2AI). Taken together, our data confirm the suspected entactogenic activity of NM2AI; however, these in vivo effects appear atypical and less intense with respect to those induced by the classic stimulants, in surprising analogy with what is reported by networked users.
Collapse
|
2
|
Kamp D, Paschali M, Bouanane A, Christl J, Supprian T, Meisenzahl-Lechner E, Kojda G, Lange-Asschenfeldt C. Characteristics of antipsychotic drug-induced hypothermia in psychogeriatric inpatients. Hum Psychopharmacol 2022; 37:e2816. [PMID: 34532905 DOI: 10.1002/hup.2816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 08/11/2021] [Accepted: 09/01/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Hypothermia is a potentially lethal adverse reaction to typical and atypical antipsychotic drugs (APD). Among predisposing factors are advanced age and comorbid somatic diseases. The aim of this study was to assess the incidence of hypothermia and quantify risk factors. METHOD Charts of N = 3002 psychogeriatric inpatients were screened for incidence of hypothermia (body core temperature <35.0°C). The frequency of hypothermia was compared between patients treated with versus without APD and, within the sample of APD-treated patients, for (1) specific APD, (2) sex, (3) main diagnosis, and (4) age. RESULTS N = 54 cases (2.6%) of hypothermia occurred in APD-treated patients and 12 cases (1.3%) in non-APD-treated patients (p = 0.024). In APD-treated patients, only male sex (p = 0.038) and pipamperone were associated with a higher incidence of hypothermia (p = 0.0017). Whereas the main diagnosis delirium showed a trend to significance, age did not correlate with hypothermia. CONCLUSION Medication with pipamperone was associated with an increased risk of hypothermia. The advanced age of our sample might as well explain the high incidence of hypothermia within our sample and the failure to detect high age as a risk factor due to a ceiling effect.
Collapse
Affiliation(s)
- Daniel Kamp
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Myrella Paschali
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Annabelle Bouanane
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Julia Christl
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Tillmann Supprian
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Eva Meisenzahl-Lechner
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Georg Kojda
- Department of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Christian Lange-Asschenfeldt
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,Oberberg Clinic Group, Clinics for Psychiatry, Psychosomatics and Psychotherapy, Oberberg Kliniken, Berlin, Germany
| |
Collapse
|
3
|
Docherty JR, Alsufyani HA. Pharmacology of Drugs Used as Stimulants. J Clin Pharmacol 2021; 61 Suppl 2:S53-S69. [PMID: 34396557 DOI: 10.1002/jcph.1918] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/22/2021] [Indexed: 12/21/2022]
Abstract
Psychostimulant, cardiovascular, and temperature actions of stimulants involve adrenergic (norepinephrine), dopaminergic (dopamine), and serotonergic (serotonin) pathways. Stimulants such as amphetamine, 3,4-methylenedioxymethamphetamine (MDMA), or mephedrone can act on the neuronal membrane monoamine transporters NET, DAT, and SERT and/or the vesicular monoamine transporter 2 to inhibit reuptake of neurotransmitter or cause release by reverse transport. Stimulants may have additional effects involving pre- and postsynaptic/junctional receptors for norepinephrine, dopamine, and serotonin and other receptors. As a result, stimulants may have a wide range of possible actions. Agents with cocaine or MDMA-like actions can induce serious and potentially fatal adverse events via thermodysregulatory, cardiovascular, or other mechanisms. MDMA-like stimulants may cause hyperthermia that can be life threathening. Recreational users of stimulants should be aware of the dangers of hyperthermia in a rave/club environment.
Collapse
Affiliation(s)
| | - Hadeel A Alsufyani
- Department of Physiology, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
4
|
Docherty JR, Alsufyani HA. Cardiovascular and temperature adverse actions of stimulants. Br J Pharmacol 2021; 178:2551-2568. [PMID: 33786822 DOI: 10.1111/bph.15465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/22/2021] [Accepted: 03/10/2021] [Indexed: 12/11/2022] Open
Abstract
The vast majority of illicit stimulants act at monoaminergic systems, causing both psychostimulant and adverse effects. Stimulants can interact as substrates or antagonists at the nerve terminal monoamine transporter that mediates the reuptake of monoamines across the nerve synaptic membrane and at the vesicular monoamine transporter (VMAT-2) that mediates storage of monoamines in vesicles. Stimulants can act directly at presynaptic or postsynaptic receptors for monoamines or have indirect monoamine-mimetic actions due to the release of monoamines. Cocaine and other stimulants can acutely increase the risk of sudden cardiac death. Stimulants, particularly MDMA, in hot conditions, such as that occurring at a "rave," have caused fatalities from the consequences of hyperthermia, often compounding cardiac adverse actions. This review examines the pharmacology of the cardiovascular and temperature adverse actions of stimulants.
Collapse
Affiliation(s)
- James R Docherty
- Department of Physiology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Hadeel A Alsufyani
- Department of Physiology, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
5
|
Drinking to death: Hyponatraemia induced by synthetic phenethylamines. Drug Alcohol Depend 2020; 212:108045. [PMID: 32460203 DOI: 10.1016/j.drugalcdep.2020.108045] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 11/21/2022]
Abstract
Synthetic phenethylamines are widely abused drugs, comprising new psychoactive substances such as synthetic cathinones, but also well-known amphetamines such as methamphetamine and 3,4-methylenedioxymethamphetamine (MDMA, ecstasy). Cathinones and amphetamines share many toxicodynamic mechanisms. One of their potentially life-threatening consequences, particularly of MDMA, is serotonin-mediated hyponatraemia. Herein, we review the state of the art on phenethylamine-induced hyponatremia; discuss the mechanisms involved; and present the preventive and therapeutic measures. Hyponatraemia mediated by phenethylamines results from increased secretion of antidiuretic hormone (ADH) and consequent kidney water reabsorption, additionally involving diaphoresis and polydipsia. Data for MDMA suggest that acute hyponatraemia elicited by cathinones may also be a consequence of metabolic activation. The literature often reveals hyponatraemia-associated complications such as cerebral oedema, cerebellar tonsillar herniation and coma that may evolve to a fatal outcome, particularly in women. Ready availability of fluids and the recommendation to drink copiously at the rave scene to counteract hyperthermia, often precipitate water intoxication. Users should be advised about the importance of controlling fluid intake while using phenethylamines. At early signs of adverse effects, medical assistance should be promptly sought. Severe hyponatraemia (<130 mmol sodium/L plasma) may be corrected with hypertonic saline or suppression of fluid intake. Also, clinicians should be made aware of the hyponatraemic potential of these drugs and encouraged to report future cases of toxicity to increase knowledge on this potentially lethal outcome.
Collapse
|
6
|
Shokry IM, Shields CJ, Callanan JJ, Ma Z, Tao R. Differential role of dose and environment in initiating and intensifying neurotoxicity caused by MDMA in rats. BMC Pharmacol Toxicol 2019; 20:47. [PMID: 31383036 PMCID: PMC6683525 DOI: 10.1186/s40360-019-0326-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 07/18/2019] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND MDMA causes serotonin (5-HT) syndrome immediately after administration and serotonergic injury in a few days or weeks. However, a serotonin syndrome is not always followed by serotonergic injury, indicating different mechanisms responsible for two adverse effects. The goal of present study was to determine causes for two adverse events and further test that dose and environment have a differential role in initiating and intensifying MDMA neurotoxicity. METHODS Initiation and intensification were examined by comparing neurotoxic effects of a high-dose (10 mg/kg × 3 at 2 h intervals) with a low-dose (2 mg/kg × 3) under controlled-environmental conditions. Initiation of a serotonin syndrome was estimated by measuring extracellular 5-HT, body-core temperature, electroencephalogram and MDMA concentrations in the cerebrospinal fluid, while intensification determined in rats examined under modified environment. Initiation and intensification of the serotonergic injury were assessed in rats by measuring tissue 5-HT content, SERT density and functional integrity of serotonergic retrograde transportation. RESULTS Both low- and high-dose could cause increases in extracellular 5-HT to elicit a serotonin syndrome at the same intensity. Modification of environmental conditions, which had no impact on MDMA-elicited increases in 5-HT levels, markedly intensified the syndrome intensity. Although either dose would cause the severe syndrome under modified environments, only the high-dose that resulted in high MDMA concentrations in the brain could cause serotonergic injury. CONCLUSION Our results reveal that extracellular 5-HT is the cause of a syndrome and activity of postsynaptic receptors critical for the course of syndrome intensification. Although the high-dose has the potential to initiate serotonergic injury due to high MDMA concentrations present in the brain, whether an injury is observed depends upon the drug environment via the levels of reactive oxygen species generated. This suggests that brain MDMA concentration is the determinant in the injury initiation while reactive oxygen species generation associated with the injury intensification. It is concluded that the two adverse events utilize distinctly different mediating molecules during the toxic initiation and intensification.
Collapse
Affiliation(s)
- Ibrahim M. Shokry
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431 USA
- Ross University School of Veterinary Medicine, Basseterre, Saint Kitts and Nevis
| | - Connor J. Shields
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431 USA
| | - John J. Callanan
- Ross University School of Veterinary Medicine, Basseterre, Saint Kitts and Nevis
| | - Zhiyuan Ma
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431 USA
| | - Rui Tao
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431 USA
| |
Collapse
|
7
|
Petschner P, Tamasi V, Adori C, Kirilly E, Ando RD, Tothfalusi L, Bagdy G. Gene expression analysis indicates reduced memory and cognitive functions in the hippocampus and increase in synaptic reorganization in the frontal cortex 3 weeks after MDMA administration in Dark Agouti rats. BMC Genomics 2018; 19:580. [PMID: 30071829 PMCID: PMC6090855 DOI: 10.1186/s12864-018-4929-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 07/05/2018] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND 3,4-methylenedioxymethamphetamine (MDMA, "ecstasy") is a widely used entactogenic drug known to impair cognitive functions on the long-run. Both hippocampal and frontal cortical regions have well established roles in behavior, memory formation and other cognitive tasks and damage of these regions is associated with altered behavior and cognitive functions frequently described in otherwise healthy MDMA users. Meanwhile, in post-traumatic stress disorder (PTSD) patients seem to benefit from therapeutic application of the drug, where damage in hippocampal cue extinction may play a role. The aim of this study was to examine the hippocampus, frontal cortex and dorsal raphe of Dark Agouti rats with gene expression arrays (Illumina RatRef bead arrays) looking for possible mechanisms and new candidates contributing to the consequences of a single dose of MDMA (15 mg/kg) 3 weeks earlier. RESULTS The number of differentially expressed genes in the hippocampus, frontal cortex and the dorsal raphe were 481, 155, and 15, respectively. Gene set enrichment analysis of the microarray data revealed reduced expression of 'memory' and 'cognition', 'dendrite development' and 'regulation of synaptic plasticity' gene sets in the hippocampus, parallel to the downregulation of CaMK II subunits, glutamate-, CB1 cannabinoid- and EphA4, EphA5, EphA6 receptors. Downregulated gene sets in the frontal cortex were related to protein synthesis, chromatin organization, transmembrane transport processes, while 'dendrite development', 'regulation of synaptic plasticity' and 'positive regulation of synapse assembly' gene sets were upregulated besides elevated levels of a CaMK II subunit and NMDA2B glutamate receptor. Changes in the dorsal raphe region were mild and in most cases not significant. CONCLUSION The present data raise the possibility of new synapse formation / synaptic reorganization in the frontal cortex 3 weeks after a single neurotoxic dose of MDMA. In contrast, a prolonged depression of new neurite formation in the hippocampus is proposed by downregulations of members in long-term potentiation pathway and synaptic plasticity emphasizing the particular vulnerability of this brain region and proposing a mechanism responsible for cognitive problems in healthy individuals. At the same time, these results underpin benefits of MDMA in PTSD, where the drug may help memory extinction.
Collapse
Affiliation(s)
- Peter Petschner
- Department of Pharmacodynamics, Semmelweis University, Nagyvarad ter 4., Budapest, H-1089, Hungary.,MTA-SE Neuropsychopharmacology & Neurochemistry Research Group, Nagyvarad ter 4., Budapest, H-1089, Hungary
| | - Viola Tamasi
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvarad ter 4., Budapest, H-1089, Hungary
| | - Csaba Adori
- Department of Pharmacodynamics, Semmelweis University, Nagyvarad ter 4., Budapest, H-1089, Hungary.,4 Retzius Laboratory, Department of Neuroscience, Karolinska Institutet, Retzius väg 8, 17177, Stockholm, Sweden
| | - Eszter Kirilly
- Department of Pharmacodynamics, Semmelweis University, Nagyvarad ter 4., Budapest, H-1089, Hungary
| | - Romeo D Ando
- Department of Pharmacodynamics, Semmelweis University, Nagyvarad ter 4., Budapest, H-1089, Hungary
| | - Laszlo Tothfalusi
- Department of Pharmacodynamics, Semmelweis University, Nagyvarad ter 4., Budapest, H-1089, Hungary
| | - Gyorgy Bagdy
- Department of Pharmacodynamics, Semmelweis University, Nagyvarad ter 4., Budapest, H-1089, Hungary. .,MTA-SE Neuropsychopharmacology & Neurochemistry Research Group, Nagyvarad ter 4., Budapest, H-1089, Hungary. .,NAP-2-SE New Antidepressant Target Research Group, Semmelweis University, Nagyvarad ter 4., Budapest, H-1089, Hungary.
| |
Collapse
|
8
|
Bogaty SER, Lee RSC, Hickie IB, Hermens DF. Meta-analysis of neurocognition in young psychosis patients with current cannabis use. J Psychiatr Res 2018; 99:22-32. [PMID: 29407284 DOI: 10.1016/j.jpsychires.2018.01.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 01/12/2018] [Accepted: 01/12/2018] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Adult psychosis patients (i.e. over the age of 25 years) who are also lifetime cannabis users (CANN±) appear to exhibit superior cognition compared to never-using patients (CANN-). The objective of this meta-analysis was to evaluate the cognitive differences between CANN- and patients who currently use cannabis (CANN+) (i.e. during the CANN± patients' cannabis-using stage). Specifically, focusing on young patients under the age of 25 years, the typical stage of both psychosis- and cannabis-onset. METHOD Of the 308 studies identified through database searches and secondary referencing, 14 compared neurocognition of CANN+ and CANN- in young people with psychotic disorders (mean age between 15 and 45 years). Effect sizes were extracted using neurocognitive test performance between CANN+ and CANN- and random effects modelling was conducted on pooled ES and moderator analyses. RESULTS CANN+ performed worse on several cognitive domains (i.e. premorbid IQ, current IQ, verbal learning, verbal working memory, motor inhibition) compared to CANN-. The association between age and performance in CANN+ cognition was varied, with older age predictive of worse performance in processing speed, sustained attention, verbal memory, and better performance in verbal learning and very fluency. Of note, CANN+ outperformed CANN- in tests of conceptual set-shifting. CONCLUSION These results are consistent with previous findings indicating that CANN+ demonstrate poorer neurocognition than CANN-; and that this is exacerbated with increasing age. Our findings demonstrate significant cognitive differences between patients with CANN+ versus CANN- even at early-onset psychosis, which could suggest a different underlying mechanism towards psychosis for cannabis users.
Collapse
Affiliation(s)
| | - Rico S C Lee
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia; Brain and Mental Health Laboratory, Monash University, Melbourne, VIC, Australia
| | - Ian B Hickie
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
| | - Daniel F Hermens
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia; Sunshine Coast Mind and Neuroscience Thompson Institute, University of the Sunshine Coast, Birtinya, QLD, Australia.
| |
Collapse
|
9
|
Šíchová K, Pinterová N, Židková M, Horsley RR, Lhotková E, Štefková K, Vejmola Č, Uttl L, Balíková M, Kuchař M, Páleníček T. Mephedrone (4-Methylmethcathinone): Acute Behavioral Effects, Hyperthermic, and Pharmacokinetic Profile in Rats. Front Psychiatry 2018; 8:306. [PMID: 29375408 PMCID: PMC5767720 DOI: 10.3389/fpsyt.2017.00306] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 12/20/2017] [Indexed: 12/20/2022] Open
Abstract
Mephedrone (MEPH) is a synthetic cathinone derivative with effects that mimic MDMA and/or cocaine. Our study in male Wistar rats provides detailed investigations of MEPH's and its primary metabolite nor-mephedrone's (nor-MEPH) pharmacokinetics and bio-distribution to four different substrates (serum, brain, lungs, and liver), as well as comparative analysis of their effects on locomotion [open field test (OFT)] and sensorimotor gating [prepulse inhibition of acoustic startle reaction (PPI ASR)]. Furthermore, in order to mimic the crowded condition where MEPH is typically taken (e.g., clubs), the acute effect of MEPH on thermoregulation in singly- and group-housed rats was evaluated. Pharmacokinetics of MEPH and nor-MEPH after MEPH (5 mg/kg, sc.) were analyzed over 8 h using liquid chromatography with mass spectrometry. MEPH (2.5, 5, or 20 mg/kg, sc.) and nor-MEPH (5 mg/kg, sc.) were administered 5 or 40 min before the behavioral testing in the OFT and PPI ASR; locomotion and its spatial distribution, ASR, habituation and PPI itself were quantified. The effect of MEPH on rectal temperature was measured after 5 and 20 mg/kg, sc. Both MEPH and nor-MEPH were detected in all substrates, with the highest levels detected in lungs. Mean brain: serum ratios were 1:1.19 (MEPH) and 1:1.91 (nor-MEPH), maximum concentrations were observed at 30 min; at 2 and 4 h after administration, nor-MEPH concentrations were higher compared to the parent drug. While neither of the drugs disrupted PPI, both increased locomotion and affected its spatial distribution. The effects of MEPH were dose dependent, rapid, and short-lasting, and the intensity of locomotor stimulant effects was comparable between MEPH and nor-MEPH. Despite the disappearance of behavioral effects within 40 min after administration, MEPH induced rectal temperature elevations that persisted for 3 h even in singly housed rats. To conclude, we observed a robust, short-lasting, and most likely synergistic stimulatory effect of both drugs which corresponded to brain pharmacokinetics. The dissociation between the duration of behavioral and hyperthermic effects is indicative of the possible contribution of nor-MEPH or other biologically active metabolites. This temporal dissociation may be related to the risk of prolonged somatic toxicity when stimulatory effects are no longer present.
Collapse
Affiliation(s)
- Klára Šíchová
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic
| | - Nikola Pinterová
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic
- Third Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Monika Židková
- Institute of Forensic Medicine and Toxicology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Rachel R. Horsley
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic
| | - Eva Lhotková
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic
| | - Kristýna Štefková
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic
| | - Čestmír Vejmola
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic
- Third Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Libor Uttl
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Marie Balíková
- Institute of Forensic Medicine and Toxicology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Martin Kuchař
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic
- Forensic Laboratory of Biologically Active Compounds, Department of Chemistry of Natural Compounds, University of Chemistry and Technology Prague, Prague, Czech Republic
| | - Tomáš Páleníček
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic
- Third Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| |
Collapse
|
10
|
Fonseca DA, Guerra AF, Carvalho F, Fernandes E, Ferreira LM, Branco PS, Antunes PE, Antunes MJ, Cotrim MD. Hyperthermia Severely Affects the Vascular Effects of MDMA and Metabolites in the Human Internal Mammary Artery In Vitro. Cardiovasc Toxicol 2017; 17:405-416. [PMID: 28084566 DOI: 10.1007/s12012-017-9398-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
3,4-Methylenedioxymethamphetamine (MDMA or "ecstasy") is a recreational drug used worldwide for its distinctive psychotropic effects. Although important cardiovascular effects, such as increased blood pressure and heart rate, have also been described, the vascular effects of MDMA and metabolites and their correlation with hyperthermia (major side effect of MDMA) are not yet fully understood and have not been previously reported. This study aimed at evaluating the effects of MDMA and its main catechol metabolites, alpha-methyldopamine (α-MeDA), N-methyl-alpha-methyldopamine (N-Me-α-MeDA), 5-(glutathion-S-yl)-alpha-methyldopamine [5-(GSH)-α-MeDA] and 5-(glutathion-S-yl)-N-methyl-alpha-methyldopamine [5-(GSH)-N-Me-α-MeDA], on the 5-HT-dependent vasoactivity in normothermia (37 °C) and hyperthermia (40 °C) of the human internal mammary artery (IMA) in vitro. The results showed the ability of MDMA, α-MeDA and N-Me-α-MeDA to exert vasoconstriction of the IMA which was considerably higher in hyperthermic conditions (about fourfold for MDMA and α-MeDA and twofold for N-Me-α-MeDA). The results also showed that all the compounds may influence the 5-HT-mediated concentration-dependent response of IMA, as MDMA, α-MeDA and N-Me-α-MeDA behaved as partial agonists and 5-(GSH)-α-MeDA and 5-(GSH)-N-Me-α-MeDA as antagonists. In conclusion, MDMA abuse may imply a higher cardiovascular risk associated both to MDMA and its metabolites that might be relevant in patients with underlying cardiovascular diseases, particularly in hyperthermia.
Collapse
Affiliation(s)
- D A Fonseca
- Pharmacology and Pharmaceutical Care Group, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal.
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal.
- CNC.IBILI, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal.
| | - A F Guerra
- Pharmacology and Pharmaceutical Care Group, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
| | - F Carvalho
- UCIBIO/REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313, Porto, Portugal
| | - E Fernandes
- UCIBIO/REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313, Porto, Portugal
| | - L M Ferreira
- REQUIMTE/CQFB (Centro de Química Fina e Biotecnologia), Department of Chemistry, Faculty of Sciences and Technology, University Nova of Lisbon, Quinta da Torre, 2829-516, Caparica, Portugal
| | - P S Branco
- REQUIMTE/CQFB (Centro de Química Fina e Biotecnologia), Department of Chemistry, Faculty of Sciences and Technology, University Nova of Lisbon, Quinta da Torre, 2829-516, Caparica, Portugal
| | - P E Antunes
- Center of Cardiothoracic Surgery, Coimbra University Hospitals, Praceta Prof. Mota Pinto, 3000-075, Coimbra, Portugal
| | - M J Antunes
- Center of Cardiothoracic Surgery, Coimbra University Hospitals, Praceta Prof. Mota Pinto, 3000-075, Coimbra, Portugal
| | - M D Cotrim
- Pharmacology and Pharmaceutical Care Group, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
- CNC.IBILI, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
| |
Collapse
|
11
|
Tao R, Shokry IM, Callanan JJ. Environment Influencing Serotonin Syndrome Induced by Ecstasy Abuse. ANNALS OF FORENSIC RESEARCH AND ANALYSIS 2017; 4:1039. [PMID: 29732414 PMCID: PMC5931730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Ecstasy is a recreational drug containing 3,4-methylenedioxymethamphetamine (MDMA). In the U.S., there are several millions of lifetime users, and millions each year added to the list as new users. Only several thousand every year show signs of severe toxicity and require emergency intervention. The illness is known as serotonin (5-HT) syndrome, which can be mild, moderate or severe. The relationship between mild, moderate and severe syndromes appears to be interchangeable, but the severe syndrome is life-threatening. The serotonergic mechanisms of how the mild or moderate syndrome becomes severe and life-threatening have attracted considerable attention in the last few years as an effort to explore new treatments potentially to manage illness and prevent death of patients. High levels of extracellular 5-HT in the brain produced by large doses of MDMA are not always necessary to cause a severe serotonin syndrome. Additional mechanisms may be more important. Recent research has demonstrated that environmental conditions (i.e., non-drug factors) are more critical in determining the severity of MDMA-induced serotonin syndrome than the drug dose. The purpose of the current article was to review available evidence regarding the effect of non-drug factors on serotonergic extrasynaptic receptor responsivity and the severity of MDMA-induced serotonin syndrome.
Collapse
Affiliation(s)
- Rui Tao
- Charles E. Schmidt College of Medicine, Florida Atlantic University, USA
| | - Ibrahim M. Shokry
- Charles E. Schmidt College of Medicine, Florida Atlantic University, USA
- Ross University School of Veterinary Medicine, West Indies
| | | |
Collapse
|
12
|
Popova D, Forsblad A, Hashemian S, Jacobsson SOP. Non-Serotonergic Neurotoxicity by MDMA (Ecstasy) in Neurons Derived from Mouse P19 Embryonal Carcinoma Cells. PLoS One 2016; 11:e0166750. [PMID: 27861613 PMCID: PMC5115802 DOI: 10.1371/journal.pone.0166750] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 11/03/2016] [Indexed: 11/18/2022] Open
Abstract
3,4-methylenedioxymethamphetamine (MDMA; ecstasy) is a commonly abused recreational drug that causes neurotoxic effects in both humans and animals. The mechanism behind MDMA-induced neurotoxicity is suggested to be species-dependent and needs to be further investigated on the cellular level. In this study, the effects of MDMA in neuronally differentiated P19 mouse embryonal carcinoma cells have been examined. MDMA produces a concentration-, time- and temperature-dependent toxicity in differentiated P19 neurons, as measured by intracellular MTT reduction and extracellular LDH activity assays. The P19-derived neurons express both the serotonin reuptake transporter (SERT), that is functionally active, and the serotonin metabolizing enzyme monoamine oxidase A (MAO-A). The involvement of these proteins in the MDMA-induced toxicity was investigated by a pharmacological approach. The MAO inhibitors clorgyline and deprenyl, and the SERT inhibitor fluoxetine, per se or in combination, were not able to mimic the toxic effects of MDMA in the P19-derived neurons or block the MDMA-induced cell toxicity. Oxidative stress has been implicated in MDMA-induced neurotoxicity, but pre-treatment with the antioxidants α-tocopherol or N-acetylcysteine did not reveal any protective effects in the P19 neurons. Involvement of mitochondria in the MDMA-induced cytotoxicity was also examined, but MDMA did not alter the mitochondrial membrane potential (ΔΨm) in the P19 neurons. We conclude that MDMA produce a concentration-, time- and temperature-dependent neurotoxicity and our results suggest that the mechanism behind MDMA-induced toxicity in mouse-derived neurons do not involve the serotonergic system, oxidative stress or mitochondrial dysfunction.
Collapse
Affiliation(s)
- Dina Popova
- Department of Pharmacology and Clinical Neuroscience, Umeå University, Umeå, Sweden
| | - Andréas Forsblad
- Department of Pharmacology and Clinical Neuroscience, Umeå University, Umeå, Sweden
| | - Sanaz Hashemian
- Department of Pharmacology and Clinical Neuroscience, Umeå University, Umeå, Sweden
| | - Stig O. P. Jacobsson
- Department of Pharmacology and Clinical Neuroscience, Umeå University, Umeå, Sweden
- * E-mail:
| |
Collapse
|
13
|
Riley CL, Dao C, Kenaston MA, Muto L, Kohno S, Nowinski SM, Solmonson AD, Pfeiffer M, Sack MN, Lu Z, Fiermonte G, Sprague JE, Mills EM. The complementary and divergent roles of uncoupling proteins 1 and 3 in thermoregulation. J Physiol 2016; 594:7455-7464. [PMID: 27647490 DOI: 10.1113/jp272971] [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: 06/22/2016] [Accepted: 09/15/2016] [Indexed: 01/23/2023] Open
Abstract
KEY POINTS Both uncoupling protein 1 (UCP1) and UCP3 are important for mammalian thermoregulation. UCP1 and UCP3 in brown adipose tissue mediate early and late phases of sympathomimetic thermogenesis, respectively. Lipopolysaccharide thermogenesis requires skeletal muscle UCP3 but not UCP1. Acute noradrenaline-induced hyperthermia requires UCP1 but not UCP3. Loss of both UCP1 and UCP3 accelerate the loss of body temperature compared to UCP1KO alone during acute cold exposure. ABSTRACT Uncoupling protein 1 (UCP1) is the established mediator of brown adipose tissue-dependent thermogenesis. In contrast, the role of UCP3, expressed in both skeletal muscle and brown adipose tissue, in thermoregulatory physiology is less well understood. Here, we show that mice lacking UCP3 (UCP3KO) have impaired sympathomimetic (methamphetamine) and completely abrogated lipopolysaccharide (LPS) thermogenesis, but a normal response to noradrenaline. By comparison, UCP1 knockout (UCP1KO) mice exhibit blunted methamphetamine and fully inhibited noradrenaline thermogenesis, but an increased febrile response to LPS. We further establish that mice lacking both UCP1 and 3 (UCPDK) fail to show methamphetamine-induced hyperthermia, and have a markedly accelerated loss of body temperature and survival after cold exposure compared to UCP1KO mice. Finally, we show that skeletal muscle-specific human UCP3 expression is able to significantly rescue LPS, but not sympathomimetic thermogenesis blunted in UCP3KO mice. These studies identify UCP3 as an important mediator of physiological thermogenesis and support a renewed focus on targeting UCP3 in metabolic physiology.
Collapse
Affiliation(s)
- Christopher L Riley
- Department of Molecular Biosciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Christine Dao
- Division of Pharmacy and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
| | - M Alexander Kenaston
- Division of Pharmacy and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Luigina Muto
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Cosenza, Italy
| | - Shohei Kohno
- Division of Pharmacy and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Sara M Nowinski
- Department of Biochemistry, The University of Utah, Salt Lake City, UT, 84112, USA
| | - Ashley D Solmonson
- Department of Molecular Biosciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, 78712, USA.,Division of Pharmacy and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Matthew Pfeiffer
- Division of Pharmacy and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Michael N Sack
- National Heart, Lung, and Blood Institute, Laboratory of Mitochondrial Biology and Metabolism, NIH, Bethesda, MD, 20892, USA
| | - Zhongping Lu
- Cardiovascular and Pulmonary Branch and the Department of Biochemistry and Molecular Medicine, George Washington University, and the Veterans Affairs Medical Center, Washington, DC, 20422, 20052, USA
| | - Giuseppe Fiermonte
- Department of Biosciences, Biotechnologies, and Biopharmaceutics and Center of Excellence in Comparative Genomics, University of Bari, 70125, Bari, Italy
| | - Jon E Sprague
- The Ohio Attorney General's Center for the Future of Forensic Science, Bowling Green State University, Bowling Green, OH, 43403, USA
| | - Edward M Mills
- Division of Pharmacy and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
| |
Collapse
|
14
|
Frau L, Simola N, Porceddu PF, Morelli M. Effect of crowding, temperature and age on glia activation and dopaminergic neurotoxicity induced by MDMA in the mouse brain. Neurotoxicology 2016; 56:127-138. [PMID: 27451954 DOI: 10.1016/j.neuro.2016.07.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 07/15/2016] [Accepted: 07/20/2016] [Indexed: 12/13/2022]
Abstract
3,4-methylenedyoxymethamphetamine (MDMA or "ecstasy"), a recreational drug of abuse, can induce glia activation and dopaminergic neurotoxicity. Since MDMA is often consumed in crowded environments featuring high temperatures, we studied how these factors influenced glia activation and dopaminergic neurotoxicity induced by MDMA. C57BL/6J adolescent (4 weeks old) and adult (12 weeks old) mice received MDMA (4×20mg/kg) in different conditions: 1) while kept 1, 5, or 10×cage at room temperature (21°C); 2) while kept 5×cage at either room (21°C) or high (27°C) temperature. After the last MDMA administration, immunohistochemistry was performed in the caudate-putamen for CD11b and GFAP, to mark microglia and astroglia, and in the substantia nigra pars compacta for tyrosine hydroxylase, to mark dopaminergic neurons. MDMA induced glia activation and dopaminergic neurotoxicity, compared with vehicle administration. Crowding (5 or 10 mice×cage) amplified MDMA-induced glia activation (in adult and adolescent mice) and dopaminergic neurotoxicity (in adolescent mice). Conversely, exposure to a high environmental temperature (27°C) potentiated MDMA-induced glia activation in adult and adolescent mice kept 5×cage, but not dopaminergic neurotoxicity. Crowding and exposure to a high environmental temperature amplified MDMA-induced hyperthermia, and a positive correlation between body temperature and activation of either microglia or astroglia was found in adult and adolescent mice. These results provide further evidence that the administration setting influences the noxious effects of MDMA in the mouse brain. However, while crowding amplifies both glia activation and dopaminergic neurotoxicity, a high environmental temperature exacerbates glia activation only.
Collapse
Affiliation(s)
- Lucia Frau
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Via Ospedale 72, 09124, Cagliari, Italy.
| | - Nicola Simola
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Via Ospedale 72, 09124, Cagliari, Italy.
| | - Pier Francesca Porceddu
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Via Ospedale 72, 09124, Cagliari, Italy.
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Via Ospedale 72, 09124, Cagliari, Italy; CNR, Institute of Neuroscience, Cagliari, Italy.
| |
Collapse
|
15
|
Abad S, Ramon C, Pubill D, Camarasa J, Camins A, Escubedo E. Adolescent exposure to MDMA induces dopaminergic toxicity in substantia nigra and potentiates the amyloid plaque deposition in the striatum of APPswe/PS1dE9 mice. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1815-26. [PMID: 27344237 DOI: 10.1016/j.bbadis.2016.06.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 05/24/2016] [Accepted: 06/21/2016] [Indexed: 12/20/2022]
Abstract
MDMA is one of the most used drugs by adolescents and its consumption has been associated with many psychobiological problems, among them psychomotor problems. Moreover, some authors described that early exposure to MDMA may render the dopaminergic neurons more vulnerable to the effects of future neurotoxic insults. Alzheimer disease (AD) is the main cause of dementia in the elderly and a percentage of the patients have predisposition to suffer nigrostriatal alterations, developing extrapyramidal signs. Nigrostriatal dysfunction in the brain of aged APPswe/PS1dE9 (APP/PS1), a mouse model of familiar AD (FAD), has also been described. The aim of the present study was to investigate the consequences of adolescent exposure to MDMA in APP/PS1 mice, on nigrostriatal function on early adulthood. We used a MDMA schedule simulating weekend binge abuse of this substance. Our MDMA schedule produced a genotype-independent decrease in dopaminergic neurons in the substantia nigra that remained at least 3months. Shortly after the injury, wild-type animals showed a decrease in the locomotor activity and apparent DA depletion in striatum, however in the APP/PS1 mice neither the locomotor activity nor the DA levels were modified, but a reduction in dopamine transporter (DAT) expression and a higher levels of oxidative stress were observed. We found that these disturbances are age-related characteristics that this APP/PS1 mice develops spontaneously much later. Therefore, MDMA administration seems to anticipate the striatal dopaminergic dysfunction in this FAD model. The most important outcome lies in a potentiation, by MDMA, of the amyloid beta deposition in the striatum.
Collapse
Affiliation(s)
- Sonia Abad
- Department of Pharmacology, Toxicology and Therapeutic Chemistry (Pharmacology Section) and Biomedicine Institute, University of Barcelona (IBUB), Spain
| | - Carla Ramon
- Department of Pharmacology, Toxicology and Therapeutic Chemistry (Pharmacology Section) and Biomedicine Institute, University of Barcelona (IBUB), Spain
| | - David Pubill
- Department of Pharmacology, Toxicology and Therapeutic Chemistry (Pharmacology Section) and Biomedicine Institute, University of Barcelona (IBUB), Spain
| | - Jorge Camarasa
- Department of Pharmacology, Toxicology and Therapeutic Chemistry (Pharmacology Section) and Biomedicine Institute, University of Barcelona (IBUB), Spain
| | - Antonio Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry (Pharmacology Section) and Biomedicine Institute, University of Barcelona (IBUB), Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Spain.
| | - Elena Escubedo
- Department of Pharmacology, Toxicology and Therapeutic Chemistry (Pharmacology Section) and Biomedicine Institute, University of Barcelona (IBUB), Spain
| |
Collapse
|
16
|
Moratalla R, Khairnar A, Simola N, Granado N, García-Montes JR, Porceddu PF, Tizabi Y, Costa G, Morelli M. Amphetamine-related drugs neurotoxicity in humans and in experimental animals: Main mechanisms. Prog Neurobiol 2015; 155:149-170. [PMID: 26455459 DOI: 10.1016/j.pneurobio.2015.09.011] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 09/04/2015] [Accepted: 09/15/2015] [Indexed: 12/13/2022]
Abstract
Amphetamine-related drugs, such as 3,4-methylenedioxymethamphetamine (MDMA) and methamphetamine (METH), are popular recreational psychostimulants. Several preclinical studies have demonstrated that, besides having the potential for abuse, amphetamine-related drugs may also elicit neurotoxic and neuroinflammatory effects. The neurotoxic potentials of MDMA and METH to dopaminergic and serotonergic neurons have been clearly demonstrated in both rodents and non-human primates. This review summarizes the species-specific cellular and molecular mechanisms involved in MDMA and METH-mediated neurotoxic and neuroinflammatory effects, along with the most important behavioral changes elicited by these substances in experimental animals and humans. Emphasis is placed on the neuropsychological and neurological consequences associated with the neuronal damage. Moreover, we point out the gap in our knowledge and the need for developing appropriate therapeutic strategies to manage the neurological problems associated with amphetamine-related drug abuse.
Collapse
Affiliation(s)
- Rosario Moratalla
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid, Spain; CIBERNED, ISCIII, Madrid, Spain.
| | - Amit Khairnar
- Applied Neuroscience Research Group, CEITEC - Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Nicola Simola
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| | - Noelia Granado
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid, Spain; CIBERNED, ISCIII, Madrid, Spain
| | - Jose Ruben García-Montes
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid, Spain; CIBERNED, ISCIII, Madrid, Spain
| | - Pier Francesca Porceddu
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| | - Giulia Costa
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy; Centre of Excellence for Neurobiology of Dependence, University of Cagliari, Cagliari, Italy; National Research Council (CNR), Institute of Neuroscience, Cagliari, Italy
| |
Collapse
|
17
|
Coman D, Sanganahalli BG, Jiang L, Hyder F, Behar KL. Distribution of temperature changes and neurovascular coupling in rat brain following 3,4-methylenedioxymethamphetamine (MDMA, "ecstasy") exposure. NMR IN BIOMEDICINE 2015; 28:1257-66. [PMID: 26286889 PMCID: PMC4573923 DOI: 10.1002/nbm.3375] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 06/22/2015] [Accepted: 07/19/2015] [Indexed: 05/05/2023]
Abstract
(+/-)3,4-methylenedioxymethamphetamine (MDMA, "ecstasy") is an abused psychostimulant that produces strong monoaminergic stimulation and whole-body hyperthermia. MDMA-induced thermogenesis involves activation of uncoupling proteins (UCPs), primarily a type specific to skeletal muscle (UCP-3) and absent from the brain, although other UCP types are expressed in the brain (e.g. thalamus) and might contribute to thermogenesis. Since neuroimaging of brain temperature could provide insights into MDMA action, we measured spatial distributions of systemically administered MDMA-induced temperature changes and dynamics in rat cortex and subcortex using a novel magnetic resonance method, Biosensor Imaging of Redundant Deviation in Shifts (BIRDS), with an exogenous temperature-sensitive probe (thulium ion and macrocyclic chelate 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetramethyl-1,4,7,10-tetraacetate (DOTMA(4-))). The MDMA-induced temperature rise was greater in the cortex than in the subcortex (1.6 ± 0.4 °C versus 1.3 ± 0.4 °C) and occurred more rapidly (2.0 ± 0.2 °C/h versus 1.5 ± 0.2 °C/h). MDMA-induced temperature changes and dynamics in the cortex and body were correlated, although the body temperature exceeded the cortex temperature before and after MDMA. Temperature, neuronal activity, and blood flow (CBF) were measured simultaneously in the cortex and subcortex (i.e. thalamus) to investigate possible differences of MDMA-induced warming across brain regions. MDMA-induced warming correlated with increases in neuronal activity and blood flow in the cortex, suggesting that the normal neurovascular response to increased neural activity was maintained. In contrast to the cortex, a biphasic relationship was seen in the subcortex (i.e. thalamus), with a decline in CBF as temperature and neural activity rose, transitioning to a rise in CBF for temperature above 37 °C, suggesting that MDMA affected CBF and neurovascular coupling differently in subcortical regions. Considering that MDMA effects on CBF and heat dissipation (as well as potential heat generation) may vary regionally, neuroprotection may require different cooling strategies.
Collapse
Affiliation(s)
- Daniel Coman
- Magnetic Resonance Research Center (MRRC), Yale University, New Haven, CT 06520, USA
- Core Center for Quantitative Neuroscience with Magnetic Resonance (QNMR), Yale University, New Haven, CT 06520, USA
- Department of Diagnostic Radiology, Yale University, New Haven, CT 06520, USA
| | - Basavaraju G. Sanganahalli
- Magnetic Resonance Research Center (MRRC), Yale University, New Haven, CT 06520, USA
- Core Center for Quantitative Neuroscience with Magnetic Resonance (QNMR), Yale University, New Haven, CT 06520, USA
- Department of Diagnostic Radiology, Yale University, New Haven, CT 06520, USA
| | - Lihong Jiang
- Magnetic Resonance Research Center (MRRC), Yale University, New Haven, CT 06520, USA
- Department of Diagnostic Radiology, Yale University, New Haven, CT 06520, USA
| | - Fahmeed Hyder
- Magnetic Resonance Research Center (MRRC), Yale University, New Haven, CT 06520, USA
- Core Center for Quantitative Neuroscience with Magnetic Resonance (QNMR), Yale University, New Haven, CT 06520, USA
- Department of Diagnostic Radiology, Yale University, New Haven, CT 06520, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Kevin L. Behar
- Magnetic Resonance Research Center (MRRC), Yale University, New Haven, CT 06520, USA
- Department of Psychiatry, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
18
|
Tao R, Shokry IM, Callanan JJ, Adams HD, Ma Z. Mechanisms and environmental factors that underlying the intensification of 3,4-methylenedioxymethamphetamine (MDMA, Ecstasy)-induced serotonin syndrome in rats. Psychopharmacology (Berl) 2015; 232:1245-60. [PMID: 25300903 PMCID: PMC4361258 DOI: 10.1007/s00213-014-3759-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 09/23/2014] [Indexed: 01/08/2023]
Abstract
RATIONALE Illicit use of 3,4-methylenedioxymethamphetamine (MDMA, Ecstasy) may cause a mild or severe form of the serotonin syndrome. The syndrome intensity is not just influenced by drug doses but also by environmental factors. OBJECTIVES Warm environmental temperatures and physical activity are features of raves. The purpose of this study was to assess how these two factors can potentially intensify the syndrome. METHODS Rats were administered MDMA at doses of 0.3, 1, or 3 mg/kg and examined in the absence or presence of warm temperature and physical activity. The syndrome intensity was estimated by visual scoring for behavioral syndrome and also instrumentally measuring changes in symptoms of the syndrome. RESULTS Our results showed that MDMA at 3 mg/kg, but not 0.3 or 1 mg/kg, caused a mild serotonin syndrome in rats. Each environmental factor alone moderately intensified the syndrome. When the two factors were combined, the intensification became more severe than each factor alone highlighting a synergistic effect. This intensification was blocked by the 5-HT2A receptor antagonist M100907, competitive N-methyl-D-aspartic acid (NMDA) receptor antagonist CGS19755, autonomic ganglionic blocker hexamethonium, and the benzodiazepine-GABAA receptor agonist midazolam but not by the 5-HT1A receptor antagonist WAY100635 or nicotinic receptor antagonist methyllycaconitine. CONCLUSIONS Our data suggest that, in the absence of environmental factors, the MDMA-induced syndrome is mainly mediated through the serotonergic transmission (5-hydroxytryptamine (5HT)-dependent mechanism) and therefore is relatively mild. Warm temperature and physical activity facilitate serotonergic and other neural systems such as glutamatergic and autonomic transmissions, resulting in intensification of the syndrome (non-5HT mechanisms).
Collapse
Affiliation(s)
- Rui Tao
- Charles E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Road, Boca Raton, FL, 33431, USA,
| | - Ibrahim M. Shokry
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, USA, School of Veterinary Medicine, Ross University, St. Kitts, West Indies
| | - John J. Callanan
- School of Veterinary Medicine, Ross University, St. Kitts, West Indies
| | - H. Daniel Adams
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, USA
| | - Zhiyuan Ma
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, USA
| |
Collapse
|
19
|
Rubio M, López-Arnau R, Pubill D, Escubedo E, Camarasa J. Serotonin is involved in the psychostimulant and hypothermic effect of 4-methylamphetamine in rats. Neurosci Lett 2015; 590:68-73. [PMID: 25641130 DOI: 10.1016/j.neulet.2015.01.075] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 01/26/2015] [Accepted: 01/28/2015] [Indexed: 10/24/2022]
Abstract
4-Methylamphetamine (4-MA) has recently emerged as a designer drug of abuse in Europe and it is consumed always with amphetamine. There have been reported some deaths and non-fatal intoxications related to 4-MA. We investigated the changes in locomotor activity and body temperature after 4-MA administration to male Sprague-Dawley rats. Our experiments were carried out at a normal or high ambient temperature. 4-MA (2.5-10mg/Kg, given subcutaneously) increased, in a dose-dependent manner, the horizontal locomotor activity that was significantly reduced by ketanserin, p-cholorophenylalanine (pCPA) or haloperidol, but not by pindolol. In addition, we have studied the effect of 4-MA on core body temperature by means of an implanted electronic thermograph, enabling continuous measurement of body temperature. We observed a dose-dependent hypothermic response to 4-MA that reached a maximum 45 min after a single injection. We also evidenced slight tachyphylaxis to the hypothermic effect when 4-MA was administered four times in a 2h interval. The pre-treatment of animals with pCPA or pindolol, but not with ketanserin, fully abolished the hypothermic effect of 4-MA. With all that, we conclude that hypothermia induced by 4-MA is due to the release of 5-HT which activates postsynaptic 5-HT1A receptors.
Collapse
Affiliation(s)
- Mar Rubio
- Department of Pharmacology and Therapeutic Chemistry (Pharmacology Section) and Institute of Biomedicine (IBUB), Faculty of Pharmacy, University of Barcelona, Spain
| | - Raúl López-Arnau
- Department of Pharmacology and Therapeutic Chemistry (Pharmacology Section) and Institute of Biomedicine (IBUB), Faculty of Pharmacy, University of Barcelona, Spain
| | - David Pubill
- Department of Pharmacology and Therapeutic Chemistry (Pharmacology Section) and Institute of Biomedicine (IBUB), Faculty of Pharmacy, University of Barcelona, Spain
| | - Elena Escubedo
- Department of Pharmacology and Therapeutic Chemistry (Pharmacology Section) and Institute of Biomedicine (IBUB), Faculty of Pharmacy, University of Barcelona, Spain.
| | - Jorge Camarasa
- Department of Pharmacology and Therapeutic Chemistry (Pharmacology Section) and Institute of Biomedicine (IBUB), Faculty of Pharmacy, University of Barcelona, Spain
| |
Collapse
|
20
|
Green AR, King MV, Shortall SE, Fone KCF. The preclinical pharmacology of mephedrone; not just MDMA by another name. Br J Pharmacol 2014; 171:2251-68. [PMID: 24654568 DOI: 10.1111/bph.12628] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Revised: 12/05/2013] [Accepted: 12/11/2013] [Indexed: 01/15/2023] Open
Abstract
The substituted β-keto amphetamine mephedrone (4-methylmethcathinone) was banned in the UK in April 2010 but continues to be used recreationally in the UK and elsewhere. Users have compared its psychoactive effects to those of 3,4-methylenedioxymethamphetamine (MDMA, 'ecstasy'). This review critically examines the preclinical data on mephedrone that have appeared over the last 2-3 years and, where relevant, compares the pharmacological effects of mephedrone in experimental animals with those obtained following MDMA administration. Both mephedrone and MDMA enhance locomotor activity and change rectal temperature in rodents. However, both of these responses are of short duration following mephedrone compared with MDMA probably because mephedrone has a short plasma half-life and rapid metabolism. Mephedrone appears to have no pharmacologically active metabolites, unlike MDMA. There is also little evidence that mephedrone induces a neurotoxic decrease in monoamine concentration in rat or mouse brain, again in contrast to MDMA. Mephedrone and MDMA both induce release of dopamine and 5-HT in the brain as shown by in vivo and in vitro studies. The effect on 5-HT release in vivo is more marked with mephedrone even though both drugs have similar affinity for the dopamine and 5-HT transporters in vitro. The profile of action of mephedrone on monoamine receptors and transporters suggests it could have a high abuse liability and several studies have found that mephedrone supports self-administration at a higher rate than MDMA. Overall, current data suggest that mephedrone not only differs from MDMA in its pharmacological profile, behavioural and neurotoxic effects, but also differs from other cathinones.
Collapse
Affiliation(s)
- A R Green
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | | | | | | |
Collapse
|
21
|
Protracted treatment with MDMA induces heteromeric nicotinic receptor up-regulation in the rat brain: an autoradiography study. Prog Neuropsychopharmacol Biol Psychiatry 2014; 53:1-8. [PMID: 24614096 DOI: 10.1016/j.pnpbp.2014.02.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 02/10/2014] [Accepted: 02/26/2014] [Indexed: 11/23/2022]
Abstract
Previous studies indicate that 3,4-methylenedioxy-methamphetamine (MDMA, ecstasy) can induce a heteromeric nicotinic acetylcholine receptor (nAChR, mainly of α4β2 subtype) up-regulation. In this study we treated male Sprague-Dawley rats twice-daily for 10 days with either saline or MDMA (7 mg/kg) and sacrificed them the day after to perform [(125)I]Epibatidine binding autoradiograms on serial coronal slices. MDMA induced significant increases in nAChR density in the substantia nigra, ventral tegmental area, nucleus accumbens, olfactory tubercle, anterior caudate-putamen, somatosensory, motor, auditory and retrosplenial cortex, laterodorsal thalamus nuclei, amygdala, postsubiculum and pontine nuclei. These increases ranged from 3% (retrosplenial cortex) to 30 and 34% (amygdala and substantia nigra). No increased α4 subunit immunoreactivity was found in up-regulated areas compared with saline-treated rats, suggesting a post-translational mechanism as occurs with nicotine. The heteromeric nAChR up-regulation in certain areas could account, at least in part, for the reinforcing, sensitizing and psychiatric disorders observed after long-term consumption of MDMA.
Collapse
|
22
|
Green AR, Nutt DJ. Pharmacology should be at the centre of all preclinical and clinical studies on new psychoactive substances (recreational drugs). J Psychopharmacol 2014; 28:711-8. [PMID: 24674814 DOI: 10.1177/0269881114528593] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Despite the publication of a substantial body of preclinical and clinical information on recent recreational drugs such as 3,4-methylenedioxymethamphetamine (MDMA, 'ecstasy') and cathinone compounds such as mephedrone there remains a disturbing lack of consensus as to how dangerous these compounds are to the health of the individual and to society in general. This perspective proposes that use of good pharmacological practice should be mandatory in all preclinical and clinical studies. Its use will assist both translation and reverse translation of information produced in animals and clinical subjects. We propose several basic rules to be followed in all future studies. Preclinical studies should employ pharmacokinetic-pharmacodynamic integration thereby exposing animals to known or calculable drug concentrations. This will provide results relevant to pharmacology rather than toxicology and, crucially, data relevant to human drug use. Full experimental detail should be routinely provided, to allow comparison with other similar work. In clinical studies evidence should be provided that the drug under investigation has been ingested by the subjects being examined, and details given of all other drugs being ingested. Drug-drug interactions are an unavoidable confound but studies of a size that allows reliable statistical evaluation and preferably allows sub-group analysis, particularly by using meta-analysis, should help with this problem. This may require greater collaboration between investigative groups, as routinely occurs during pharmaceutical clinical trials. Other proposals include greater integration of preclinical and clinical scientists in both preclinical and clinical studies and changes in the law regarding Good Manufacturing Process (GMP) sourcing of drug for human studies.
Collapse
Affiliation(s)
- A Richard Green
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | - David J Nutt
- Division of Neurosciences and Mental Health, Imperial College London, London, UK
| |
Collapse
|
23
|
Current preclinical studies on neuroinflammation and changes in blood-brain barrier integrity by MDMA and methamphetamine. Neuropharmacology 2014; 87:125-34. [PMID: 24594477 DOI: 10.1016/j.neuropharm.2014.02.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 02/05/2014] [Accepted: 02/20/2014] [Indexed: 11/24/2022]
Abstract
The blood-brain barrier (BBB) is essential in the maintenance of brain homeostasis both by preserving normal brain functioning and also by protecting the brain from exposure to a range of potentially harmful substances. This review presents some of the evidence of BBB disruption following exposure to the substituted amphetamines 3,4-methylenedioxymethamphetamine (MDMA, 'ecstasy') and methamphetamine (METH), two drugs of abuse which are widely consumed recreationally by younger sectors of the population. Both MDMA and METH have been shown to produce disruption of the BBB as reflected by IgG extravasation and Evans Blue leakage. In particular, METH decreases the expression of basal lamina proteins associated with an increase in matrix metalloproteinase activity. These changes in BBB integrity appear to be related to MDMA-induced activation of the mitogen-activated protein kinase (MAPK) JNK1/2. The consequences of the disruption in the BBB by these two drugs remain to be established, but there is evidence in the literature that, at least in the case of METH, increased matrix metalloproteinase (MMP) activity may be related to increased behavioural sensitization and reward perhaps because of the modification of the passage of the drug into the CNS. In addition, the high incidence of AIDS-related neurologic disease in METH users may also be related to increased entry into the brain of virally derived neurotoxic products. This article is part of the Special Issue entitled 'CNS Stimulants'.
Collapse
|
24
|
Petschner P, Tamasi V, Adori C, Kirilly E, Ando RD, Tothfalusi L, Bagdy G. Gene expression analysis indicates CB1 receptor upregulation in the hippocampus and neurotoxic effects in the frontal cortex 3 weeks after single-dose MDMA administration in Dark Agouti rats. BMC Genomics 2013; 14:930. [PMID: 24378229 PMCID: PMC3902429 DOI: 10.1186/1471-2164-14-930] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 12/23/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND 3,4-methylenedioxymethamphetamine (MDMA, "ecstasy") is a widely used recreational drug known to impair cognitive functions on the long-run. Both hippocampal and frontal cortical regions have well established roles in behavior, memory formation and other cognitive tasks and damage of these regions is associated with altered behavior and cognitive functions, impairments frequently described in heavy MDMA users. The aim of this study was to examine the hippocampus, frontal cortex and dorsal raphe of Dark Agouti rats with gene expression arrays (Illumina RatRef bead arrays) looking for possible mechanisms and new candidates contributing to the effects of a single dose of MDMA (15 mg/kg) 3 weeks earlier. RESULTS The number of differentially expressed genes in the hippocampus, frontal cortex and the dorsal raphe were 481, 155, and 15, respectively. Gene set enrichment analysis of the microarray data revealed reduced expression of 'memory' and 'cognition', 'dendrite development' and 'regulation of synaptic plasticity' gene sets in the hippocampus, parallel to the upregulation of the CB1 cannabinoid- and Epha4, Epha5, Epha6 ephrin receptors. Downregulated gene sets in the frontal cortex were related to protein synthesis, chromatin organization, transmembrane transport processes, while 'dendrite development', 'regulation of synaptic plasticity' and 'positive regulation of synapse assembly' gene sets were upregulated. Changes in the dorsal raphe region were mild and in most cases not significant. CONCLUSION The present data raise the possibility of new synapse formation/synaptic reorganization in the frontal cortex three weeks after a single neurotoxic dose of MDMA. In contrast, a prolonged depression of new neurite formation in the hippocampus is suggested by the data, which underlines the particular vulnerability of this brain region after the drug treatment. Finally, our results also suggest the substantial contribution of CB1 receptor and endocannabinoid mediated pathways in the hippocampal impairments. Taken together the present study provides evidence for the participation of new molecular candidates in the long-term effects of MDMA.
Collapse
Affiliation(s)
- Peter Petschner
- Department of Pharmacodynamics, Semmelweis University, H-1089 Nagyvarad ter 4., Budapest, Hungary
- MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Budapest, Hungary
| | - Viola Tamasi
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, H-1089 Nagyvarad ter 4., Budapest, Hungary
| | - Csaba Adori
- Department of Pharmacodynamics, Semmelweis University, H-1089 Nagyvarad ter 4., Budapest, Hungary
| | - Eszter Kirilly
- Department of Pharmacodynamics, Semmelweis University, H-1089 Nagyvarad ter 4., Budapest, Hungary
| | - Romeo D Ando
- Department of Pharmacodynamics, Semmelweis University, H-1089 Nagyvarad ter 4., Budapest, Hungary
| | - Laszlo Tothfalusi
- Department of Pharmacodynamics, Semmelweis University, H-1089 Nagyvarad ter 4., Budapest, Hungary
| | - Gyorgy Bagdy
- Department of Pharmacodynamics, Semmelweis University, H-1089 Nagyvarad ter 4., Budapest, Hungary
- MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Budapest, Hungary
| |
Collapse
|
25
|
Docherty JR, Bexis S. Influence of ketanserin on the effects of methylenedioxymethamphetamine on body temperature in the mouse. AUTONOMIC & AUTACOID PHARMACOLOGY 2013; 33:35-41. [PMID: 23906337 DOI: 10.1111/aap.12009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 04/18/2013] [Accepted: 05/13/2013] [Indexed: 06/02/2023]
Abstract
(1) We have investigated the ability of the 5HT2 -receptor antagonist ketanserin to affect the hyperthermia produced by methylenedioxymethamphetamine (MDMA) in conscious mice and examined whether α1 -adrenoceptor antagonist actions are involved. (2) Mice were implanted with intra-abdominal temperature probes under anaesthesia and allowed 2 weeks recovery. MDMA (20 mg kg(-1) ) was administered subcutaneously 30 min after vehicle or test antagonist and effects on body temperature monitored by telemetry. (3) Following vehicle, MDMA produced a slowly developing hyperthermia, reaching a maximum increase of 1.24 °C at 150 min postinjection. Ketanserin (0.5 mg kg(-1) ) revealed a significant and marked early hypothermia to MDMA, an effect that is mimicked by the α1 -adrenoceptor antagonist prazosin (0.1 mg kg(-1) ). (4) Functional studies revealed antagonist actions of ketanserin at α1 -adrenoceptors in rat aorta and rat vas deferens in vitro indicative of α1 -adrenoceptor antagonist actions at the concentration used in vivo. (5) In conclusion, ketanserin (0.5 mg kg(-1) ) modulates the hyperthermic actions of MDMA in mice. Although we cannot rule out additional actions at 5HT2 -receptors, the actions of ketanserin are consistent with α1 -adrenoceptor antagonism. There is no clear evidence from this study that 5HT2-receptors mediate the hyperthermic response to MDMA.
Collapse
Affiliation(s)
- J R Docherty
- Department of Physiology, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin, Ireland
| | | |
Collapse
|
26
|
Methamphetamine and core temperature in the rat: ambient temperature, dose, and the effect of a D2 receptor blocker. Psychopharmacology (Berl) 2013; 228:551-61. [PMID: 23732837 DOI: 10.1007/s00213-013-3059-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Accepted: 03/02/2013] [Indexed: 12/16/2022]
Abstract
RATIONALE Methamphetamine (METH) induces hyperthermia in warm and hypothermia in cool environments. Our first goal was to further study the role of ambient temperature in METH's effect on core temperature in rats. Previously, these effects were primarily demonstrated in high doses; we extended this investigation to the low-dose range (1 mg/kg METH). Our second goal was to identify the role of the D2 receptor in METH's effects in cool ambient temperatures. METHOD Rats received METH (saline, 1, 5, and 10 mg/kg), raclopride (saline, 0.3, 0.6, and 1.2 mg/kg), or a combination (all doses of raclopride combined with 10 mg/kg METH). Treatments occurred in ambient temperatures of 18, 24, or 30 °C. RESULTS AND CONCLUSIONS Consistent with prior research, 5 and 10 mg/kg METH caused hyperthermia or hypothermia in a dose- and ambient temperature-dependent manner (60 min after METH). In contrast, 1 mg/kg produced similar levels of hyperthermia at all ambient temperatures. These findings suggest that a threshold METH dose exists; below this dose, METH still changes core temperature, but CNS control over temperature regulation is left intact. In our experiments regarding D2 blockade, raclopride decreased METH-induced core temperature at 30 and 24 °C (60 min after METH), consistent with previous findings. We extended these findings by demonstrating that in a cool ambient temperature (18 °C), raclopride pretreatment also lowered the core temperature response to METH. Although the D2 receptor is known to mediate hypothermia, the combination of METH and D2 blockade suggests a complex mediation of the core temperature response, perhaps involving neurotransmitter interactions.
Collapse
|
27
|
Shortall SE, Green AR, Swift KM, Fone KCF, King MV. Differential effects of cathinone compounds and MDMA on body temperature in the rat, and pharmacological characterization of mephedrone-induced hypothermia. Br J Pharmacol 2013; 168:966-77. [PMID: 23043631 DOI: 10.1111/j.1476-5381.2012.02236.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 08/31/2012] [Accepted: 09/09/2012] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Recreational users report that mephedrone has similar psychoactive effects to 3,4-methylenedioxymethamphetamine (MDMA). MDMA induces well-characterized changes in body temperature due to complex monoaminergic effects on central thermoregulation, peripheral blood flow and thermogenesis, but there are little preclinical data on the acute effects of mephedrone or other synthetic cathinones. EXPERIMENTAL APPROACH The acute effects of cathinone, methcathinone and mephedrone on rectal and tail temperature were examined in individually housed rats, with MDMA included for comparison. Rats were killed 2 h post-injection and brain regions were collected for quantification of 5-HT, dopamine and major metabolites. Further studies examined the impact of selected α-adrenoceptor and dopamine receptor antagonists on mephedrone-induced changes in rectal temperature and plasma catecholamines. KEY RESULTS At normal room temperature, MDMA caused sustained decreases in rectal and tail temperature. Mephedrone caused a transient decrease in rectal temperature, which was enhanced by α(1) -adrenoceptor and dopamine D(1) receptor blockade, and a prolonged decrease in tail temperature. Cathinone and methcathinone caused sustained increases in rectal temperature. MDMA decreased 5-HT and/or 5-hydroxyindoleacetic acid (5-HIAA) content in several brain regions and reduced striatal homovanillic acid (HVA) levels, whereas cathinone and methcathinone increased striatal HVA and 5-HIAA. Cathinone elevated striatal and hypothalamic 5-HT. Mephedrone elevated plasma noradrenaline levels, an effect prevented by α-adrenoceptor and dopamine receptor antagonists. CONCLUSIONS AND IMPLICATIONS MDMA and cathinones have different effects on thermoregulation, and their acute effects on brain monoamines also differ. These findings suggest that the adverse effects of cathinones in humans cannot be extrapolated from previous observations on MDMA.
Collapse
Affiliation(s)
- S E Shortall
- School of Biomedical Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK
| | | | | | | | | |
Collapse
|
28
|
Frau L, Simola N, Morelli M. Contribution of Caffeine to the Psychostimulant, Neuroinflammatory and Neurotoxic Effects of Amphetamine-Related Drugs. JOURNAL OF CAFFEINE RESEARCH 2013. [DOI: 10.1089/jcr.2013.0007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Lucia Frau
- Section of Neuropsychopharmacology, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Nicola Simola
- Section of Neuropsychopharmacology, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Micaela Morelli
- Section of Neuropsychopharmacology, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
- Centre of Excellence for Neurobiology of Dependence, University of Cagliari, Cagliari, Italy
- CNR, Institute of Neuroscience, Cagliari, Italy
| |
Collapse
|
29
|
Abstract
Numerous medications and illicit drugs can predispose an individual to heat illness, primarily by altering thermoregulation by either increasing endogenous heat production or impairing heat dissipation. This study sought to determine if use of such drugs was associated with more severe illness in patients presenting with heatstroke. A case control study was conducted on adult patients (age, ≥14 years) admitted to an intensive care unit with an admitting diagnosis of heatstroke at two academic teaching hospitals in Phoenix, AZ, between 31 August 2005 through 31 July 2010. Subjects were classified as "users" if they admitted to taking a drug on a pre-defined list of drugs associated with abnormal thermal homeostasis, or if a urine test for drugs of abuse revealed the presence of an amphetamine or cocaine. Similarly, subjects who did not take such drugs were considered "non-users." Seventy-eight patients were identified, with complete medication histories available for 74 of 78 subjects. The overall prevalence of drug utilization was 41.9 % (31 of 74). The median length of stay was 3.0 days for the non-users compared with 9.0 days for "users." There was no difference between users and non-users with regard to mortality. Drugs that impair thermoregulation are frequently encountered in patients admitted for heatstroke. Patients taking such drugs may experience increased morbidity over those patients not taking such drugs.
Collapse
|
30
|
Pubill D, Garcia-Ratés S, Camarasa J, Escubedo E. 3,4-Methylenedioxy-methamphetamine induces in vivo regional up-regulation of central nicotinic receptors in rats and potentiates the regulatory effects of nicotine on these receptors. Neurotoxicology 2012; 35:41-9. [PMID: 23261423 DOI: 10.1016/j.neuro.2012.11.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 11/19/2012] [Accepted: 11/22/2012] [Indexed: 12/01/2022]
Abstract
Nicotine (NIC), the main psychostimulant compound of smoked tobacco, exerts its effects through activation of central nicotinic acetylcholine receptors (nAChR), which become up-regulated after chronic administration. Recent work has demonstrated that the recreational drug 3,4-methylenedioxy-methamphetamine (MDMA) has affinity for nAChR and also induces up-regulation of nAChR in PC 12 cells. Tobacco and MDMA are often consumed together. In the present work we studied the in vivo effect of a classic chronic dosing schedule of MDMA in rats, alone or combined with a chronic schedule of NIC, on the density of nAChR and on serotonin reuptake transporters. MDMA induced significant decreases in [(3)H]paroxetine binding in the cortex and hippocampus measured 24h after the last dose and these decreases were not modified by the association with NIC. In the prefrontal cortex, NIC and MDMA each induced significant increases in [(3)H]epibatidine binding (29.5 and 34.6%, respectively) with respect to saline-treated rats, and these increases were significantly potentiated (up to 72.1%) when the two drugs were associated. Also in this area, [(3)H]methyllycaconitine binding was increased a 42.1% with NIC+MDMA but not when they were given alone. In the hippocampus, MDMA potentiated the α7 regulatory effects of NIC (raising a 25.5% increase to 52.5%) but alone was devoid of effect. MDMA had no effect on heteromeric nAChR in striatum and a coronal section of the midbrain containing superior colliculi, geniculate nuclei, substantia nigra and ventral tegmental area. Specific immunoprecipitation of solubilised receptors suggests that the up-regulated heteromeric nAChRs contain α4 and β2 subunits. Western blots with specific α4 and α7 antibodies showed no significant differences between the groups, indicating that, as reported for nicotine, up-regulation caused by MDMA is due to post-translational events rather than increased receptor synthesis.
Collapse
Affiliation(s)
- David Pubill
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Nucli Universitari de Pedralbes, Universitat de Barcelona, Institut de Biomedicina de la UB (IBUB), 08028 Barcelona, Spain.
| | | | | | | |
Collapse
|
31
|
Vanattou-Saïfoudine N, McNamara R, Harkin A. Caffeine provokes adverse interactions with 3,4-methylenedioxymethamphetamine (MDMA, 'ecstasy') and related psychostimulants: mechanisms and mediators. Br J Pharmacol 2012; 167:946-59. [PMID: 22671762 PMCID: PMC3492978 DOI: 10.1111/j.1476-5381.2012.02065.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 05/04/2012] [Accepted: 05/18/2012] [Indexed: 11/27/2022] Open
Abstract
Concomitant consumption of caffeine with recreational psychostimulant drugs of abuse can provoke severe acute adverse reactions in addition to longer term consequences. The mechanisms by which caffeine increases the toxicity of psychostimulants include changes in body temperature regulation, cardiotoxicity and lowering of the seizure threshold. Caffeine also influences the stimulatory, discriminative and reinforcing effects of psychostimulant drugs. In this review, we consider our current understanding of such caffeine-related drug interactions, placing a particular emphasis on an adverse interaction between caffeine and the substituted amphetamine, 3,4-methylenedioxymethamphetamine (MDMA, 'ecstasy'), which has been most recently described and characterized. Co-administration of caffeine profoundly enhances the acute toxicity of MDMA in rats, as manifested by high core body temperature, tachycardia and increased mortality. In addition, co-administration of caffeine enhances the long-term serotonergic neurotoxicity induced by MDMA. Observations to date support an interactive model of drug-induced toxicity comprising MDMA-related enhancement of dopamine release coupled to a caffeine-mediated antagonism of adenosine receptors in addition to inhibition of PDE. These experiments are reviewed together with reports of caffeine-related drug interactions with cocaine, d-amphetamine and ephedrine where similar mechanisms are implicated. Understanding the underlying mechanisms will guide appropriate intervention strategies for the management of severe reactions and potential for increased drug-related toxicity, resulting from concomitant caffeine consumption.
Collapse
Affiliation(s)
- N Vanattou-Saïfoudine
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences & Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
| | | | | |
Collapse
|
32
|
Green AR, King MV, Shortall SE, Fone KCF. Lost in translation: preclinical studies on 3,4-methylenedioxymethamphetamine provide information on mechanisms of action, but do not allow accurate prediction of adverse events in humans. Br J Pharmacol 2012; 166:1523-36. [PMID: 22188379 PMCID: PMC3419898 DOI: 10.1111/j.1476-5381.2011.01819.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 11/18/2011] [Accepted: 11/29/2011] [Indexed: 11/28/2022] Open
Abstract
3,4-Methylenedioxymethamphetamine (MDMA) induces both acute adverse effects and long-term neurotoxic loss of brain 5-HT neurones in laboratory animals. However, when choosing doses, most preclinical studies have paid little attention to the pharmacokinetics of the drug in humans or animals. The recreational use of MDMA and current clinical investigations of the drug for therapeutic purposes demand better translational pharmacology to allow accurate risk assessment of its ability to induce adverse events. Recent pharmacokinetic studies on MDMA in animals and humans are reviewed and indicate that the risks following MDMA ingestion should be re-evaluated. Acute behavioural and body temperature changes result from rapid MDMA-induced monoamine release, whereas long-term neurotoxicity is primarily caused by metabolites of the drug. Therefore acute physiological changes in humans are fairly accurately mimicked in animals by appropriate dosing, although allometric dosing calculations have little value. Long-term changes require MDMA to be metabolized in a similar manner in experimental animals and humans. However, the rate of metabolism of MDMA and its major metabolites is slower in humans than rats or monkeys, potentially allowing endogenous neuroprotective mechanisms to function in a species specific manner. Furthermore acute hyperthermia in humans probably limits the chance of recreational users ingesting sufficient MDMA to produce neurotoxicity, unlike in the rat. MDMA also inhibits the major enzyme responsible for its metabolism in humans thereby also assisting in preventing neurotoxicity. These observations question whether MDMA alone produces long-term 5-HT neurotoxicity in human brain, although when taken in combination with other recreational drugs it may induce neurotoxicity.
Collapse
Affiliation(s)
- A R Green
- School of Biomedical Sciences, Queen's Medical Centre, University of Nottingham, UK.
| | | | | | | |
Collapse
|
33
|
Ros-Simó C, Ruiz-Medina J, Valverde O. Behavioural and neuroinflammatory effects of the combination of binge ethanol and MDMA in mice. Psychopharmacology (Berl) 2012; 221:511-25. [PMID: 22139453 DOI: 10.1007/s00213-011-2598-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 11/21/2011] [Indexed: 01/22/2023]
Abstract
RATIONALE Binge drinking is a common pattern of alcohol consumption among young people. Binge drinkers are especially susceptible to brain damage when other substances are co-administered, in particular, 3,4-methylendioxymethamphetamine (MDMA). OBJECTIVE To evaluate the behavioural consequences of voluntary binge ethanol consumption, alone and in combination to MDMA. Also, to elucidate the effects of the combined consumption of these two drugs on neuroinflammation. MATERIALS AND METHODS Adolescent mice received MDMA (MDMA-treated mice), ethanol (ethanol-treated mice group) or both (ethanol plus MDMA-treated mice). Drinking in the dark (DID) procedure was used as a model of binge. Body temperature, locomotor activity, motor coordination, anxiety-like and despair behaviour in adolescent mice were evaluated 48 h, 72 h, and 7 days after the treatments. Also, neuroinflammatory response to these treatments was measured in the striatum. RESULTS The hyperthermia observed in MDMA-treated mice was abolished by pre-exposition to ethanol. Ethanol plus MDMA-treated mice showed lower locomotor activity. Ethanol-treated mice showed motor coordination impairment and increased despair behaviour. Anxiety-like behaviour was only seen in animals that were treated with both drugs. Contrarily, neuroinflammation was mostly seen in animals treated only with MDMA. CONCLUSIONS Ethanol and MDMA co-administration increases the neurobehavioural changes induced by the consumption of each one of these drugs. However, as ethanol consumption did not increase neuroinflammatory responses induced by MDMA, other mechanisms, mediated by ethanol, are likely to account for this effect and need to be evaluated.
Collapse
Affiliation(s)
- Clara Ros-Simó
- Grup de Recerca en Neurobiologia del Comportament, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona, Dr Aiguader 88, 08003 Barcelona, Spain
| | | | | |
Collapse
|
34
|
Biezonski DK, Meyer JS. The Nature of 3, 4-Methylenedioxymethamphetamine (MDMA)-Induced Serotonergic Dysfunction: Evidence for and Against the Neurodegeneration Hypothesis. Curr Neuropharmacol 2011; 9:84-90. [PMID: 21886568 PMCID: PMC3137208 DOI: 10.2174/157015911795017146] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Revised: 04/17/2010] [Accepted: 05/26/2010] [Indexed: 01/18/2023] Open
Abstract
High doses of the recreational drug 3,4-methylenedioxymethamphetamine (MDMA, "Ecstasy") have been well-documented to reduce the expression of serotonergic markers in several forebrain regions of rats and nonhuman primates. Neuroimaging studies further suggest that at least one of these markers, the plasma membrane serotonin transporter (SERT), may also be reduced in heavy Ecstasy users. Such effects, particularly when observed in experimental animal models, have generally been interpreted as reflecting a loss of serotonergic fibers and terminals following MDMA exposure. This view has been challenged, however, based on the finding that MDMA usually does not elicit glial cell reactions known to occur in response to central nervous system (CNS) damage. The aim of this review is to address both sides of the MDMA-neurotoxicity controversy, including recent findings from our laboratory regarding the potential of MDMA to induce serotonergic damage in a rat binge model. Our data add to the growing literature implicating neuroregulatory mechanisms underlying MDMA-induced serotonergic dysfunction and questioning the need to invoke a degenerative response to explain such dysfunction.
Collapse
Affiliation(s)
- Dominik K Biezonski
- Neuroscience and Behavior Program, University of Massachusetts, Amherst MA 01003, USA
| | | |
Collapse
|
35
|
Vanattou-Saïfoudine N, Behan B, Harkin A. Dopamine D1 Receptor-Mediated Intracellular Responses in the Hypothalamus after Co-Administration of Caffeine with MDMA. Basic Clin Pharmacol Toxicol 2011; 110:283-9. [DOI: 10.1111/j.1742-7843.2011.00805.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
36
|
Kasai M, Shioda K, Nisijima K, Yoshino T, Iwamura T, Kato S. The effects of mirtazapine and fluoxetine on hyperthermia induced by 3,4-methylenedioxymethamphetamine (MDMA) in rats. Neurosci Lett 2011; 499:24-7. [DOI: 10.1016/j.neulet.2011.05.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 05/09/2011] [Accepted: 05/13/2011] [Indexed: 11/16/2022]
|
37
|
Rodsiri R, Spicer C, Green AR, Marsden CA, Fone KCF. Acute concomitant effects of MDMA binge dosing on extracellular 5-HT, locomotion and body temperature and the long-term effect on novel object discrimination in rats. Psychopharmacology (Berl) 2011; 213:365-76. [PMID: 20645080 DOI: 10.1007/s00213-010-1921-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Accepted: 06/09/2010] [Indexed: 11/29/2022]
Abstract
RATIONALE 3,4-methylenedioxymethamphetamine (MDMA, ecstasy) produces an acute release of 5-HT in the brain, together with increased locomotion and hyperthermia. OBJECTIVE This study examined whether the acute functional changes of locomotor activity and body temperature are related to enhanced 5-HT release induced by MDMA. METHODS We concomitantly measured changes in extraneuronal 5-HT by in vivo brain microdialysis and used radiotelemetry to measure locomotion and body temperature to establish whether any positive correlations occur between these three parameters. 'Binge-type' repeated administration of low doses of MDMA (3 and 6 mg/kg given at 2-h intervals three times) were given to provide drug exposure similar to that experienced by recreational drug users. RESULTS MDMA induced acute hyperactivity, changes in core body temperature (both hypothermia and hyperthermia) and elevation of hippocampal 5-HT overflow, all of which were dependent on the dose of MDMA administered. The change in locomotor activity and the magnitude of the hyperthermia appeared to be unrelated both to each other and to the magnitude of MDMA-induced 5-HT release. The study also found evidence of long-term disruption of novel object discrimination 2 weeks following "binge-type" repeated MDMA administration. CONCLUSIONS MDMA-induced 5-HT release in the brain was not responsible for either the hyperthermia or increased locomotor activity that occurred. Since neither dose schedule of MDMA induced a neurotoxic loss of brain 5-HT 2 weeks after its administration, the impairment of recognition memory found in novel object discrimination probably results from other long-term changes yet to be established.
Collapse
Affiliation(s)
- Ratchanee Rodsiri
- School of Biomedical Sciences, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | | | | | | | | |
Collapse
|
38
|
Vanattou-Saïfoudine N, McNamara R, Harkin A. Mechanisms mediating the ability of caffeine to influence MDMA ('Ecstasy')-induced hyperthermia in rats. Br J Pharmacol 2010; 160:860-77. [PMID: 20590585 DOI: 10.1111/j.1476-5381.2010.00660.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Caffeine exacerbates the hyperthermia associated with an acute exposure to 3,4 methylenedioxymethamphetamine (MDMA, 'Ecstasy') in rats. The present study investigated the mechanisms mediating this interaction. EXPERIMENTAL APPROACH Adult male Sprague-Dawley rats were treated with caffeine (10 mg x kg(-1); i.p.) and MDMA (15 mg x kg(-1); i.p.) alone and in combination. Core body temperatures were monitored before and after drug administration. KEY RESULTS Central catecholamine depletion blocked MDMA-induced hyperthermia and its exacerbation by caffeine. Caffeine provoked a hyperthermic response when the catecholamine releaser d-amphetamine (1 mg x kg(-1)) was combined with the 5-HT releaser D-fenfluramine (5 mg x kg(-1)) or the non-selective dopamine receptor agonist apomorphine (1 mg x kg(-1)) was combined with the 5-HT(2) receptor agonist DOI (2 mg x kg(-1)) but not following either agents alone. Pretreatment with the dopamine D(1) receptor antagonist Schering (SCH) 23390 (1 mg x kg(-1)), the 5-HT(2) receptor antagonist ketanserin (5 mg x kg(-1)) or alpha(1)-adreno- receptor antagonist prazosin (0.2 mg x kg(-1)) blocked MDMA-induced hyperthermia and its exacerbation by caffeine. Co-administration of a combination of MDMA with the PDE-4 inhibitor rolipram (0.025 mg x kg(-1)) and the adenosine A(1/2) receptor antagonist 9-chloro-2-(2-furanyl)-[1,2,4]triazolo[1,5-C]quinazolin-5-amine 15943 (10 mg x kg(-1)) or the A(2A) receptor antagonist SCH 58261 (2 mg x kg(-1)) but not the A(1) receptor antagonist DPCPX (10 mg x kg(-1)) exacerbated MDMA-induced hyperthermia. CONCLUSIONS AND IMPLICATIONS A mechanism comprising 5-HT and catecholamines is proposed to mediate MDMA-induced hyperthermia. A combination of adenosine A(2A) receptor antagonism and PDE inhibition can account for the exacerbation of MDMA-induced hyperthermia by caffeine.
Collapse
Affiliation(s)
- N Vanattou-Saïfoudine
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences & Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | | | | |
Collapse
|
39
|
Docherty JR, Green AR. The role of monoamines in the changes in body temperature induced by 3,4-methylenedioxymethamphetamine (MDMA, ecstasy) and its derivatives. Br J Pharmacol 2010; 160:1029-44. [PMID: 20590597 DOI: 10.1111/j.1476-5381.2010.00722.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Hyperthermia is probably the most widely known acute adverse event that can follow ingestion of 3,4-methylenedioxymethamphetamine (MDMA, ecstasy) by recreational users. The effect of MDMA on body temperature is complex because the drug has actions on all three major monoamine neurotransmitters [5-hydroxytryptamine (5-HT), dopamine and noradrenaline], both by amine release and by direct receptor activation. Hyperthermia and hypothermia can be induced in laboratory animals by MDMA, depending on the ambient temperature, and involve both central thermoregulation and peripheral changes in blood flow and thermogenesis. Acute 5-HT release is not directly responsible for hyperthermia, but 5-HT receptors are involved in modulating the hyperthermic response. Impairing 5-HT function with a neurotoxic dose of MDMA or p-chlorophenylalanine alters the subsequent MDMA-induced hyperthermic response. MDMA also releases dopamine, and evidence suggests that this transmitter is involved in both the hyperthermic and hypothermic effects of MDMA in rats. The noradrenergic system is also involved in the hyperthermic response to MDMA. MDMA activates central alpha(2A)-adrenoceptors and peripheral alpha(1)-adrenoceptors to produce cutaneous vasoconstriction to restrict heat loss, and beta(3)-adrenoceptors in brown adipose tissue to increase heat generation. The hyperthermia occurring in recreational users of MDMA can be fatal, but data reviewed here indicate that it is unlikely that any single pharmaceutical agent will be effective in reversing the hyperthermia, so careful body cooling remains the principal clinical approach. Crucially, educating recreational users about the potential dangers of hyperthermia and the control of ambient temperature should remain key approaches to prevent this potentially fatal problem.
Collapse
Affiliation(s)
- J R Docherty
- Department of Physiology, Royal College of Surgeons in Ireland, Ireland.
| | | |
Collapse
|
40
|
Pourahmad J, Eskandari MR, Nosrati M, Kobarfard F, Khajeamiri AR. Involvement of mitochondrial/lysosomal toxic cross-talk in ecstasy induced liver toxicity under hyperthermic condition. Eur J Pharmacol 2010; 643:162-169. [PMID: 20599925 DOI: 10.1016/j.ejphar.2010.06.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Revised: 06/09/2010] [Accepted: 06/10/2010] [Indexed: 11/30/2022]
Abstract
The initial objectives of this study were to evaluate the extent of 3, 4-methylenedioxymethamphetamine (MDMA) induced loss of cell viability (cytotoxicity), induction of reactive oxygen species formation and damage to sub-cellular organelles (e.g. mitochondria/lysosomes) in freshly isolated rat hepatocytes under normothermic conditions (37 degrees C) and to compare the results with the effects obtained under hyperthermic conditions (41 degrees C). MDMA induced cytotoxicity, reactive oxygen species formation, mitochondrial membrane potential decline and lysosomal membrane leakiness in isolated rat hepatocytes at 37 degrees C. A rise in incubation temperature from 37 degrees C to 41 degrees C had an additive/synergic effect on the oxidative stress markers. We observed variations in mitochondrial membrane potential and lysosomal membrane stability that are significantly (P<0.05) higher than those under normothermic conditions. Antioxidants, reactive oxygen species scavengers, lysosomal inactivators, mitochondrial permeability transition (MPT) pore sealing agents, NADPH P450 reductase inhibitor, and inhibitors of reduced CYP2E1 and CYP2D6 prevented all MDMA induced hepatocyte oxidative stress cytotoxicity markers. It is therefore suggested that metabolic reductive activation of MDMA by reduced cytochrome P450s and glutathione could lead to generation of some biological reactive intermediates which could activate reactive oxygen species generation and cause mitochondrial and lysosomal oxidative stress membrane damages. We finally concluded that hyperthermia could potentiate MDMA induced liver toxicity probably through a mitochondrial/lysosomal toxic cross-talk in freshly isolated rat hepatocytes.
Collapse
Affiliation(s)
- Jalal Pourahmad
- Faculty of Pharmacy, Shaheed Beheshti University of Medical Sciences, Tehran, Iran.
| | | | | | | | | |
Collapse
|
41
|
Vanattou-Saïfoudine N, McNamara R, Harkin A. Caffeine promotes dopamine D1 receptor-mediated body temperature, heart rate and behavioural responses to MDMA ('ecstasy'). Psychopharmacology (Berl) 2010; 211:15-25. [PMID: 20437223 DOI: 10.1007/s00213-010-1864-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2010] [Accepted: 04/08/2010] [Indexed: 12/17/2022]
Abstract
RATIONALE Caffeine exacerbates the acute toxicity of 3,4-methylenedioxymethamphetamine (MDMA, 'ecstasy') in rats characterised by hyperthermia, tachycardia and lethality. Depletion of central catecholamine stores and dopamine D(1) receptor blockade have been reported to attenuate the ability of caffeine to exacerbate MDMA-induced hyperthermia. OBJECTIVES Here, we investigate whether dopamine D(1) and D(2) receptors mediate the effects of caffeine on MDMA-induced changes in body temperature, heart rate and locomotor activity. METHODS All parameters were recorded continuously in individually housed rats using bioradiotelemetry from 1 h prior to 4 h following caffeine (10 mg/kg, s.c.) and/or MDMA (10 mg/kg, s.c.) administration. RESULTS Co-administration of caffeine with MDMA provoked a switch from MDMA-induced hypothermia and bradycardia to hyperthermia and tachycardia without influencing MDMA-induced hyperlocomotion. Pre-treatment with a specific dopamine D(1/5) antagonist SCH 23390 (1 mg/kg) enhanced MDMA-induced hypothermia and blocked the ability of caffeine to provoke a switch from MDMA-induced hypothermia to hyperthermia. Furthermore, SCH 23390 blocked MDMA-induced hyperactivity and the ability of caffeine to promote a tachycardic response to MDMA. By contrast, pre-treatment with the selective D(2) antagonist, sulpiride (100 mg/kg) blocked MDMA-induced hypothermia, failed to influence the ability of caffeine to promote tachycardia whilst enhancing MDMA-induced hyperactivity. CONCLUSIONS Our results highlight the importance of dopamine D(1) and D(2) receptors in shaping the behavioural and physiological response to MDMA and suggest that the ability of caffeine to provoke MDMA-induced toxicity is associated with the promotion of dopamine D(1) over D(2) receptor-related responses.
Collapse
Affiliation(s)
- Natacha Vanattou-Saïfoudine
- Neuropsychopharmacology Research Group, Trinity College Institute of Neuroscience, Trinity College of Dublin, Dublin, 2, Ireland
| | | | | |
Collapse
|
42
|
Prenatal MDMA exposure delays postnatal development in the rat: A preliminary study. Neurotoxicol Teratol 2010; 32:425-31. [DOI: 10.1016/j.ntt.2010.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2009] [Revised: 02/18/2010] [Accepted: 03/13/2010] [Indexed: 11/20/2022]
|
43
|
Docherty JR. Subtypes of functional alpha1-adrenoceptor. Cell Mol Life Sci 2010; 67:405-17. [PMID: 19862476 PMCID: PMC11115521 DOI: 10.1007/s00018-009-0174-4] [Citation(s) in RCA: 141] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 09/11/2009] [Accepted: 10/05/2009] [Indexed: 11/29/2022]
Abstract
In this review, subtypes of functional alpha1-adrenoceptor are discussed. These are cell membrane receptors, belonging to the seven-transmembrane-spanning G-protein-linked family of receptors, which respond to the physiological agonist noradrenaline. alpha1-Adrenoceptors can be divided into alpha1A-, alpha1B- and alpha1D-adrenoceptors, all of which mediate contractile responses involving Gq/11 and inositol phosphate turnover. A fourth alpha1-adrenoceptor, the alpha1L-, represents a functional phenotype of the alpha1A-adrenoceptor. alpha1-Adrenoceptor subtype knock-out mice have refined our knowledge of the functions of alpha-adrenoceptor subtypes, particuarly as subtype-selective agonists and antagonists are not available for all subtypes. alpha1-Adrenoceptors function as stimulatory receptors involved particularly in smooth muscle contraction, especially contraction of vascular smooth muscle, both in local vasoconstriction and in the control of blood pressure and temperature, and contraction of the prostate and bladder neck. Central actions are now being elucidated.
Collapse
MESH Headings
- Animals
- Blood Pressure/physiology
- Body Temperature Regulation
- Drug Inverse Agonism
- GTP-Binding Protein alpha Subunits, Gq-G11/metabolism
- Inositol Phosphates/metabolism
- Mice
- Mice, Knockout
- Muscle, Smooth/physiology
- Muscle, Smooth, Vascular/physiology
- Receptors, Adrenergic, alpha-1/classification
- Receptors, Adrenergic, alpha-1/metabolism
- Receptors, Adrenergic, alpha-1/physiology
- Second Messenger Systems/physiology
- Vasoconstriction/physiology
Collapse
Affiliation(s)
- James R Docherty
- Department of Physiology, Royal College of Surgeons in Ireland, 123, St. Stephen's Green, Dublin 2, Ireland.
| |
Collapse
|
44
|
Dumont GJH, Kramers C, Sweep FCGJ, Willemsen JJ, Touw DJ, Schoemaker RC, van Gerven JMA, Buitelaar JK, Verkes RJ. Ethanol co-administration moderates 3,4-methylenedioxymethamphetamine effects on human physiology. J Psychopharmacol 2010; 24:165-74. [PMID: 19074534 DOI: 10.1177/0269881108100020] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Alcohol is frequently used in combination with 3,4-methylenedioxymethamphetamine (MDMA). Both drugs affect cardiovascular function, hydration and temperature regulation, but may have partly opposing effects. The present study aims to assess the acute physiologic effects of (co-) administration of MDMA and ethanol over time. A four-way, double blind, randomized, crossover, placebo-controlled study in 16 healthy volunteers (9 male and 7 female) between the ages of 18 and 29. MDMA (100 mg) was given orally and blood ethanol concentration was maintained at pseudo-steady state levels of 0.6 per thousand by a three-hour 10% intravenous ethanol clamp. Cardiovascular function, temperature and hydration measures were recorded throughout the study days. Ethanol did not significantly affect physiologic function, with the exception of a short lasting increase in heart rate. MDMA potently increased heart rate and blood pressure and induced fluid retention as well as an increase in temperature. Co-administration of ethanol with MDMA did not affect cardiovascular function compared to the MDMA alone condition, but attenuated the effects of MDMA on fluid retention and showed a trend for attenuation of MDMA-induced temperature increase. In conclusion, co-administration of ethanol and MDMA did not exacerbate physiologic effects compared to all other drug conditions, and moderated some effects of MDMA alone.
Collapse
Affiliation(s)
- G J H Dumont
- Department of Psychiatry, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Bexis S, Docherty JR. Role of alpha 1- and beta 3-adrenoceptors in the modulation by SR59230A of the effects of MDMA on body temperature in the mouse. Br J Pharmacol 2009; 158:259-66. [PMID: 19422394 PMCID: PMC2795232 DOI: 10.1111/j.1476-5381.2009.00186.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2008] [Accepted: 01/07/2009] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE We have investigated the ability of the beta(3)-adrenoceptor antagonist 1-(2-ethylphenoxy)-3-[[(1S)-1,2,3,4,-tetrahydro-1-naphthalenyl]amino]-(2S)-2-propanol hydrochloride (SR59230A) to affect the hyperthermia produced by methylenedioxymethamphetamine (MDMA) in conscious mice and whether alpha(1)-adrenoceptor antagonist actions are involved. EXPERIMENTAL APPROACH Mice were implanted with temperature probes under anaesthesia, and allowed 2 week recovery. MDMA (20 mg x kg(-1)) was administered subcutaneously 30 min after vehicle or test antagonist and effects on body temperature monitored by telemetry. KEY RESULTS Following vehicle, MDMA produced a slowly developing hyperthermia, reaching a maximum increase of 1.8 degrees C at 130 min post injection. A low concentration of SR59230A (0.5 mg x kg(-1)) produced a small but significant attenuation of the slowly developing hyperthermia to MDMA. A high concentration of SR59230A (5 mg x kg(-1)) revealed a significant and marked early hypothermic reaction to MDMA, an effect that was mimicked by the alpha(1)-adrenoceptor antagonist prazosin. Functional and ligand binding studies revealed actions of SR59230A at alpha(1)-adrenoceptors. CONCLUSIONS AND IMPLICATIONS 1-(2-ethylphenoxy)-3-[[(1S)-1,2,3,4,-tetrahydro-1-naphthalenyl]amino]-(2S)-2-propanol hydrochloride in high concentrations modulates the hyperthermic actions of MDMA in mice in two ways: by blocking an early alpha(1)-adrenoceptor-mediated component to reveal a hypothermia, and by a small attenuation of the later hyperthermic component which may possibly be beta(3)-adrenoceptor-mediated (this seen with the low concentration of SR59230A). Hence, the major actions of SR59230A in modulating the actions of MDMA on temperature involve alpha(1)-adrenoceptor antagonism.
Collapse
Affiliation(s)
- Sotiria Bexis
- Department of Physiology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | |
Collapse
|
46
|
van Nieuwenhuijzen PS, McGregor IS. Sedative and hypothermic effects of gamma-hydroxybutyrate (GHB) in rats alone and in combination with other drugs: assessment using biotelemetry. Drug Alcohol Depend 2009; 103:137-47. [PMID: 19446408 DOI: 10.1016/j.drugalcdep.2009.03.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Revised: 01/22/2009] [Accepted: 03/05/2009] [Indexed: 11/19/2022]
Abstract
The recreational drug gamma-hydroxybutyrate (GHB) has euphoric effects and can induce sedation and body temperature changes. GHB is frequently combined with other recreational drugs although these interactions are not well characterised. The present study used biotelemetry to provide a fine-grained analysis of the effects of GHB on body temperature and locomotor activity in freely moving rats, and investigated interactions between GHB and 3,4-methylenedioxymethamphetamine (MDMA), methamphetamine (METH) and various antagonist drugs. GHB (1000mg/kg) caused profound sedation for more than 2h and a complex triphasic effect on body temperature: an initial hypothermia (5-40min), followed by hyperthermia (40-140min), followed again by hypothermia (140-360min). A lower GHB dose (500mg/kg) also caused sedation but only a hypothermic effect that lasted up to 6h. The dopamine D(1) receptor antagonist SCH 23390 (1mg/kg), the opioid antagonist naltrexone (1mg/kg), the benzodiazepine antagonist flumazenil (10mg/kg), and the 5-HT(2A/2C) receptor antagonist ritanserin (1mg/kg) did not prevent the overall sedative or body temperature effects of GHB (1000mg/kg). However the GABA(B) antagonist SCH 50911 (50mg/kg) prevented the hyperthermia induced by GHB (1000mg/kg). Repeated daily administration of GHB (1000mg/kg) produced tolerance to the sedative and hyperthermic effects of the drug and cross-tolerance to the sedative effects of the GABA(B) receptor agonist baclofen (10mg/kg). A high ambient temperature of 28 degrees C prevented the hypothermia obtained with GHB (500mg/kg) at 20 degrees C, while GHB (500mg/kg) reduced the hyperthermia and hyperactivity produced by co-administered doses of MDMA (5mg/kg) or METH (1mg/kg) at 28 degrees C. These results further confirm a role for GABA(B) receptors in the hypothermic and sedative effects of GHB and show an interaction between GHB and MDMA, and GHB and METH, that may be relevant to the experience of recreational users who mix these drugs.
Collapse
|
47
|
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: 201] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Accepted: 02/27/2009] [Indexed: 11/29/2022]
|
48
|
Sharma HS, Ali SF. Acute administration of 3,4-methylenedioxymethamphetamine induces profound hyperthermia, blood-brain barrier disruption, brain edema formation, and cell injury. Ann N Y Acad Sci 2008; 1139:242-58. [PMID: 18991870 DOI: 10.1196/annals.1432.052] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The psychostimulant 3,4-,ethylenedioxymethamphetamine (MDMA, "ecstasy") is known to induce hyperthermia and alterations in neurochemical metabolism in the CNS. However, the detailed cellular or molecular mechanisms behind MDMA-induced neurotoxicity are still not well known. Since MDMA induces profound hyperthermia that could lead to intense cellular stress and cause disruption of the blood-brain barrier (BBB), this investigation examined the effects of acute MDMA on BBB dysfunction, brain edema, and cell injury in rats and mice. When MDMA (40 mg/kg, i.p.) was administered to rats or mice, these animals exhibited profound behavioral disturbances (hyperactivity and hyperlocomotion) and hyperthermia (>40 to 41 degrees C) at 4 h. At this time, the leakage of Evans blue dye was evident, particularly in the cerebellum, hippocampus, cortex, thalamus, and hypothalamus. This effect was most pronounced in mice compared to rats. Marked increase in brain water along with Na(+), K(+), and Cl(-) content was also seen in the aforementioned brain regions. Presence of distorted neuronal and glial cells in brain regions associated with leakage of Evans blue is quite common in MDMA-treated animals. Increased albumin immunoreactivity, indicating breakdown of the BBB, and upregulation of glial fibrillary acidic protein (GFAP), suggesting activation of astrocytes, were seen in most brain regions showing edematous changes. Upregulation of heat-shock protein (HSP72) immunoreactivity in the nuclei and cell cytoplasm of the neurons located in the edematous brain regions are quite common. Taken together, these observations are the first to show that MDMA has the capacity to disrupt BBB permeability to proteins and to induce the formation of edema, probably by inducing hyperthermia and cellular stress, as evident with HSP overexpression leading to cell injury.
Collapse
Affiliation(s)
- Hari Shanker Sharma
- Laboratory of Neurochemistry, Division of Neurotoxicology, National Center of Toxicological Research, Food and Drug Administration, Jefferson, Arkansas, USA.
| | | |
Collapse
|
49
|
The effect of long-term repeated exposure to 3,4-methylenedioxymethamphetamine on cardiovascular and thermoregulatory changes. Psychopharmacology (Berl) 2008; 201:161-70. [PMID: 18679656 DOI: 10.1007/s00213-008-1258-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2008] [Accepted: 07/08/2008] [Indexed: 10/21/2022]
Abstract
RATIONALE 3,4-Methylenedioxymethamphetamine (MDMA, "ecstasy") disrupts thermoregulation in rats and can lead to life-threatening hyperthermia in humans. MDMA administration can also lead to long-term neurotoxicity in animals and possibly humans. OBJECTIVES The purpose of the current study was to extend previous results on the acute effects of MDMA on behavioral thermoregulation to a repeated dosing regime, simulating regular weekend use of ecstasy, on measures of thermoregulation and heart rate (HR). MATERIALS AND METHODS Sprague-Dawley rats with telemetry implants were administered 40 micromol/kg MDMA on three consecutive days each week for 1 or 6 weeks before being confined to an elevated ambient temperature (TA) (HOT; 30+/-1 degrees C) or an area at room temperature (ROOM; 21.5+/-1.5 degrees C) for 30 min. After the final drug administration, rats were placed in a thermal gradient for 4 h to allow behavioral thermoregulation. RESULTS HOT rats showed higher core temperature (TC), HR, and locomotor activity than ROOM rats during confinement to a set TA (P<0.001). HR responses to MDMA over 6 weeks at both TAs progressively decreased with repeated dosing (P<0.05). TC was significantly higher in both 6-week groups compared to the 1-week groups (P<0.05) at the end of time in the gradient. Cortical concentrations of dihydroxyphenylacetic acid (DOPAC; P<0.05) and 5-hydroxyindole acetic acid (5-HIAA; P<0.001) decreased significantly irrespective of TA, while concentrations of dopamine and 5-HT did not change. CONCLUSION Long-term treatment with MDMA resulted in apparent tolerance to the effects of the drug on HR, dysregulation of TC in thermal gradient, and depletion of cortical DOPAC and 5-HIAA.
Collapse
|
50
|
Risperidone attenuates and reverses hyperthermia induced by 3,4-methylenedioxymethamphetamine (MDMA) in rats. Neurotoxicology 2008; 29:1030-6. [PMID: 18722468 DOI: 10.1016/j.neuro.2008.07.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2007] [Revised: 07/22/2008] [Accepted: 07/22/2008] [Indexed: 11/22/2022]
Abstract
3,4-Methylenedioxymethamphetamine (MDMA, "ecstasy") is a widely used recreational drug. Despite an increase in the number of fatalities related to its use, no definite therapeutic method has been established thus far. In the present study, risperidone's ability to attenuate MDMA-induced hyperthermia and its mechanism of action were investigated in rats. The pharmacological effect of MDMA was evaluated using microdialysis. In the body temperature experiment, administration of risperidone before and after MDMA administration significantly suppressed MDMA-induced hyperthermia in a dose-dependent fashion. Furthermore, risperidone completely inhibited MDMA-induced hyperthermia at a low ambient temperature. Moreover, pretreatment with ritanserin, ketanserin, or R-96544, all of which are 5-HT(2A)-receptor antagonists, significantly prevented MDMA-induced hyperthermia. On the other hand, pretreatment with WAY-100635 (a 5-HT(1A) receptor antagonist), SB 206553 (a 5-HT(2B/2C) receptor antagonist), or SB 242084 (a 5-HT(2C) receptor antagonist) did not prevent MDMA-induced hyperthermia. Pretreatment with haloperidol, which blocks the dopamine (DA) receptors D(2) and D(1), significantly prevented MDMA-induced hyperthermia. However, sulpiride and L-741626, which are D(2) receptor blockers, did not prevent MDMA-induced hyperthermia. Pretreatment with SCH 23390 (a D(1) receptor antagonist) significantly prevented MDMA-induced hyperthermia. Furthermore, postadministration of ritanserin, haloperidol, and SCH23390 reversed MDMA-induced hyperthermia. These results demonstrate that the mechanism underlying the suppression of MDMA-induced hyperthermia by risperidone is primarily based on the drug's potent 5-HT(2A) receptor blocking effect, and to a lesser extent, on its D(1) receptor blocking effect. A microdialysis study showed that when MDMA (10mg/kg) was subcutaneously (s.c.) injected into the rats, the DA and serotonin (5-HT) levels in the anterior hypothalamus of the rats increased approximately 10- and 50-fold, respectively, as compared to their preadministration levels. These increases in the DA and 5-HT levels after MDMA injection were significantly suppressed by pretreatment with risperidone (0.5mg/kg). This suggested that both the DA and 5-HT systems were involved in the induction of hyperthermia by MDMA. Taken together, the present study's results indicate that risperidone may be an effective drug for the treatment of MDMA-induced hyperthermia in humans.
Collapse
|