Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Psychosom Med ; 81(4): 352-362, 2019 05.
Article in English | MEDLINE | ID: mdl-30855555

ABSTRACT

OBJECTIVE: Depression in patients with cardiovascular disease is associated with increased risk of adverse clinical outcomes. Investigators have searched for potential biobehavioral explanations for this increased risk. Platelet activation and response to serotonin is an attractive potential mechanism. The aim of the study was to examine platelet serotonin signaling in a group of patients with coronary artery disease (CAD) and comorbid depression to define the relationship between platelet serotonin signaling and cardiovascular complications. METHODS: A total of 300 patients with CAD were enrolled (145 with acute coronary syndrome and 155 with stable CAD). Depression was assessed using the Structured Clinical Interview for DSM-IV as well as Beck Depression Inventory II in a dichotomous and continuous manner. Platelet serotonin response was measured by serotonin augmented aggregation, direct platelet serotonin activation, platelet serotonin receptor density, and platelet serotonin uptake. Cardiovascular outcomes were assessed at 12-month follow-up. RESULTS: One third of enrolled participants had at least minimal depressive symptoms and 13.6% had major depressive disorder. Depressed cardiovascular patients had significantly higher incidence of major (odds ratio = 3.43, 95% confidence interval = 1.49-7.91, p = .004) and minor (odds ratio = 2.42, 95% confidence interval = 1.41-4.13, p = .001) adverse cardiac events. Platelet serotonin response was not significantly different in patients with depression. Participants with major depressive disorder had higher serotonin receptor density (997.5 ± 840.8 vs 619.3 ± 744.3 fmol/ug, p = .009) primarily found in ACS patients. Depressed patients with minor adverse cardiac events had increased platelet response to serotonin. CONCLUSIONS: Depressed cardiovascular patients had higher serotonin receptor density and significantly higher incidence of major and minor cardiac adverse events. Future studies with larger sample sizes including patients with more severe depression are needed to expand on the present hypothesis-generating findings.


Subject(s)
Blood Platelets/metabolism , Cardiovascular Diseases/complications , Depression/complications , Serotonin/metabolism , Cardiovascular Diseases/blood , Cardiovascular Diseases/psychology , Depression/blood , Female , Humans , Male , Middle Aged , Platelet Activation , Receptors, Serotonin/metabolism
2.
Curr Drug Abuse Rev ; 8(1): 15-25, 2015.
Article in English | MEDLINE | ID: mdl-25731754

ABSTRACT

Ecstasy or 3,4-methylenedioxymethamphetamine (MDMA) is a popular drug of abuse. In the animal studies MDMA has been shown to have deleterious effects on the serotonergic neurotransmitter system. Understanding the adverse effects of MDMA on human brain function is of considerable importance owing to the rising number of MDMA users. Various neuroimaging studies have investigated the structural, chemical and functional differences in the brain integrity of chronic MDMA users. Various neurocognitive domains like working memory, episodic memory, semantic memory, visual stimulation, motor function and impulsivity have been compared between chronic MDMA users and nonusers using fMRI. The fMRI studies remain much more sensitive in studying the neurological deficits associated with chronic MDMA use as compared to the cognitive studies alone and therefore they serve as a prelude in our understanding of MDMA induced neurotoxicity. However they still face certain limitations contributing to inconsistency in the results and further research is needed before we can draw definitive conclusions regarding the neurotoxic effects of MDMA.


Subject(s)
Hallucinogens/adverse effects , Magnetic Resonance Imaging/methods , N-Methyl-3,4-methylenedioxyamphetamine/adverse effects , Amphetamine-Related Disorders/complications , Animals , Brain/drug effects , Brain/pathology , Cognition/drug effects , Cognition Disorders/chemically induced , Humans , Neurotoxicity Syndromes/diagnosis , Neurotoxicity Syndromes/etiology
3.
Int J Neuropsychopharmacol ; 16(4): 791-801, 2013 May.
Article in English | MEDLINE | ID: mdl-22824226

ABSTRACT

Repeated doses of the popular recreational drug methylenedioxymethamphetamine (MDMA, 'Ecstasy') are known to produce neurotoxic effects on brain serotonin (5-HT) neurons but it is widely believed that typical single oral doses of MDMA are free of neurotoxic risk. Experimental and therapeutic trials with MDMA in humans are underway. The mechanisms by which MDMA produces neurotoxic effects are not understood but drug metabolites have been implicated. The aim of the present study was to assess the neurotoxic potential of a range of clinically relevant single oral doses of MDMA in a non-human primate species that metabolizes MDMA in a manner similar to humans, the squirrel monkey. A secondary objective was to explore the relationship between plasma MDMA and metabolite concentrations and lasting serotonergic deficits. Single oral doses of MDMA produced lasting dose-related serotonergic neurochemical deficits in the brains of squirrel monkeys. Notably, even the lowest dose of MDMA tested (5.7 mg/kg, estimated to be equivalent to 1.6 mg/kg in humans) produced significant effects in some brain regions. Plasma levels of MDMA engendered by neurotoxic doses of MDMA were on the order of those found in humans. Serotonergic neurochemical markers were inversely correlated with plasma concentrations of MDMA, but not with those of its major metabolites, 3,4-dihydroxymethamphetamine and 4-hydroxy-3-methoxymethamphetamine. These results suggest that single oral doses of MDMA in the range of those used by humans pose a neurotoxic risk and implicate the parent compound (MDMA), rather than one of its metabolites, in MDMA-induced 5-HT neural injury.


Subject(s)
Brain/metabolism , N-Methyl-3,4-methylenedioxyamphetamine/administration & dosage , N-Methyl-3,4-methylenedioxyamphetamine/metabolism , Serotonin Agents/administration & dosage , Serotonin Agents/metabolism , Serotonin/metabolism , Administration, Oral , Animals , Brain/drug effects , Dose-Response Relationship, Drug , Female , Male , N-Methyl-3,4-methylenedioxyamphetamine/blood , Primates , Saimiri , Serotonin Agents/blood
4.
J Pharmacol Exp Ther ; 344(2): 479-88, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23209329

ABSTRACT

The neurotoxicity of (±)-3,4-methylenedioxymethamphetamine (MDMA; "Ecstasy") is influenced by temperature and varies according to species. The mechanisms underlying these two features of MDMA neurotoxicity are unknown, but differences in MDMA metabolism have recently been implicated in both. The present study was designed to 1) assess the effect of hypothermia on MDMA metabolism, 2) determine whether the neuroprotective effect of hypothermia is related to inhibition of MDMA metabolism, and 3) determine if different neurotoxicity profiles in mice and rats are related to differences in MDMA metabolism and/or disposition in the two species. Rats and mice received single neurotoxic oral doses of MDMA at 25°C and 4°C, and body temperature, pharmacokinetic parameters, and serotonergic and dopaminergic neuronal markers were measured. Hypothermia did not alter MDMA metabolism in rats and only modestly inhibited MDMA metabolism in mice; however, it afforded complete neuroprotection in both species. Rats and mice metabolized MDMA in a similar pattern, with 3,4-methylenedioxyamphetamine being the major metabolite, followed by 4-hydroxy-3-methoxymethamphetamine and 3,4-dihydroxymethamphetamine, respectively. Differences between MDMA pharmacokinetics in rats and mice, including faster elimination in mice, did not account for the different profile of MDMA neurotoxicity in the two species. Taken together, the results of these studies indicate that inhibition of MDMA metabolism is not responsible for the neuroprotective effect of hypothermia in rodents, and that different neurotoxicity profiles in rats and mice are not readily explained by differences in MDMA metabolism or disposition.


Subject(s)
Hypothermia, Induced , N-Methyl-3,4-methylenedioxyamphetamine/metabolism , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/prevention & control , Administration, Oral , Animals , Biotransformation , Body Temperature , Brain/drug effects , Brain/metabolism , Dopamine/metabolism , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred Strains , N-Methyl-3,4-methylenedioxyamphetamine/pharmacokinetics , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Neurons/drug effects , Neurons/metabolism , Neurotoxicity Syndromes/etiology , Rats , Rats, Sprague-Dawley , Serotonin/metabolism , Species Specificity , Time Factors , Tissue Distribution
5.
J Neuropsychiatry Clin Neurosci ; 24(3): 309-15, 2012.
Article in English | MEDLINE | ID: mdl-23037644

ABSTRACT

There are currently no known early neuroanatomical markers predictive of the development of major depression or depressive symptoms after mild traumatic brain injury (mTBI). The authors conducted a 1-year longitudinal pilot study to determine whether diffusion tensor imaging (DTI) measures collected within 1 month of mTBI could predict incident depression. Of the 14 subjects who met study inclusion criteria, 4 (28.6%) developed major depression over the follow-up period. Compared with the nondepressed group, those who developed depression had white-matter abnormalities in the fronto-temporal regions measured by DTI. These preliminary results highlight the need for additional studies, including studies using a larger sample and appropriate controls.


Subject(s)
Brain Injuries/complications , Brain/pathology , Depressive Disorder, Major/diagnosis , Depressive Disorder, Major/etiology , Diffusion Tensor Imaging , Adult , Anisotropy , Female , Follow-Up Studies , Glasgow Coma Scale , Humans , Image Processing, Computer-Assisted , Linear Models , Male , Middle Aged , Psychiatric Status Rating Scales , Young Adult
6.
Exp Clin Psychopharmacol ; 20(1): 28-39, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21928913

ABSTRACT

The extended-release formulation of zolpidem (Ambien CR) is approved for the treatment of insomnia without a treatment duration limit. Acutely zolpidem impairs performance, and no research to date has examined whether tolerance develops to these performance impairments during nighttime awakening. The present double-blind, placebo-controlled study examined whether tolerance develops to zolpidem-induced acute performance impairment after repeated (22-30 days) nightly use. Effects of bedtime administration of zolpidem extended-release (ZOL; 12.5 mg) were tested on a battery of performance measures assessed during a forced nighttime awakening in 15 healthy male volunteers who completed overnight polysomnographic recording sessions in our laboratory at baseline and after approximately a month of at-home ZOL. As expected, bedtime ZOL administration was associated with changes in sleep architecture and impairments across all performance domains during nighttime testing (psychomotor function, attention, working memory, episodic memory, metacognition) with no residual next morning impairment. Tolerance did not develop to the observed ZOL-related impairments on any outcome. Possible evidence of acute abstinence effects after discontinuation of ZOL was observed on some performance and sleep outcomes. Overall, these findings suggest that performance is significantly impaired during nighttime awakening even after a month of nightly ZOL administration, and these impairments could significantly impact safety should nighttime awakening require unimpaired functioning (e.g., driving; combat-related activities in the military).


Subject(s)
Cognition/drug effects , Hypnotics and Sedatives/administration & dosage , Psychomotor Performance/drug effects , Pyridines/administration & dosage , Sleep/drug effects , Adult , Attention/drug effects , Delayed-Action Preparations/administration & dosage , Double-Blind Method , Humans , Male , Memory, Episodic , Polysomnography/methods , Time Factors , Young Adult , Zolpidem
7.
Psychopharmacology (Berl) ; 217(4): 475-84, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21603895

ABSTRACT

RATIONALE: (±)3,4-Methylenedioxymethamphetamine (MDMA) is a popular recreational drug that has potential to damage brain serotonin (5-HT) neurons in humans. Brain 5-HT neurons play a role in pain modulation, yet little is known about long-term effects of MDMA on pain function. Notably, MDMA users have been shown to have altered sleep, a phenomenon that can lead to altered pain modulation. OBJECTIVES: This study sought to assess pain processing in MDMA users using objective methods, and explore potential relationships between pain processing and sleep indices. METHODS: Forty-two abstinent MDMA users and 43 age-matched controls participated in a 5-day inpatient study. Outcome measures included standardized measures of pain, sleep polysomnograms, and power spectral measures of the sleep EEG. When differences in psychophysiological measures of pain were found, the relationship between pain and sleep measures was explored. RESULTS: MDMA users demonstrated lower pressure pain thresholds, increased cold pain ratings, increased pain ratings during testing of diffuse noxious inhibitory control, and decreased Stage 2 sleep. Numerous significant relationships between sleep and pain measures were identified, but differences in sleep between the two groups were not found to mediate altered pain perception in MDMA users. CONCLUSIONS: Abstinent MDMA users have altered pain perception and sleep architecture. Although pain and sleep outcomes were related, differences in sleep architecture in MDMA users did not mediate altered pain responses. It remains to be determined whether alterations in pain perception in MDMA users are secondary to neurotoxicity of 5-HT-mediated pain pathways or alterations in other brain processes that modulate pain perception.


Subject(s)
N-Methyl-3,4-methylenedioxyamphetamine/adverse effects , Pain Threshold/psychology , Sleep/drug effects , Substance Withdrawal Syndrome/psychology , Adult , Brain/drug effects , Brain/metabolism , Female , Humans , Male , Neuropsychological Tests , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/psychology , Pain Measurement , Physical Stimulation , Regression Analysis , Serotonin/metabolism , Substance Withdrawal Syndrome/metabolism , Young Adult
8.
J Pharmacol Exp Ther ; 338(1): 310-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21493752

ABSTRACT

The baboon is potentially an attractive animal for modeling 3,4-methylenedioxymethamphetamine (MDMA) effects in humans. Baboons self-administer MDMA, are susceptible to MDMA neurotoxicity, and are suitable for positron emission tomography, the method most often used to probe for MDMA neurotoxicity in humans. Because pharmacokinetic equivalence is a key feature of a good predictive animal model, we compared the pharmacokinetics of MDMA in baboons and humans. Baboons were trained to orally consume MDMA. Then, pharmacokinetic profiles of MDMA and its major metabolites were determined after various oral MDMA doses using the same analytical method recently used to perform similar studies in humans. Results indicate that MDMA pharmacokinetics after oral ingestion differ markedly between baboons and humans. Baboons had little or no MDMA in their plasma but had high plasma concentrations of 3,4-dihydroxymethamphetamine (HHMA), pointing to much more extensive first-pass metabolism of MDMA in baboons than in humans. Other less prominent differences included less O-methylation of HHMA to 4-hydroxy-3-methoxymethamphetamine, greater N-demethylation of MDMA to 3,4-methylenedioxyamphetamine, and a shorter half-life of HHMA in the baboon. To our knowledge, this is the first study to characterize MDMA metabolism and disposition in the baboon. Differences in MDMA pharmacokinetics between baboons and humans suggest that the baboon may not be ideal for modeling human MDMA exposure. However, the unusually rapid conversion of MDMA to HHMA in the baboon may render this animal uniquely useful for clarifying the relative role of the parent compound (MDMA) versus metabolites (particularly HHMA) in the biological actions of MDMA.


Subject(s)
N-Methyl-3,4-methylenedioxyamphetamine/administration & dosage , N-Methyl-3,4-methylenedioxyamphetamine/blood , Papio hamadryas/blood , Administration, Oral , Animals , Dose-Response Relationship, Drug , Humans , Male , N-Methyl-3,4-methylenedioxyamphetamine/pharmacokinetics , Species Specificity , Tissue Distribution/drug effects , Tissue Distribution/physiology
9.
Synapse ; 65(10): 983-90, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21360595

ABSTRACT

3,4-Methylenedioxymethamphetamine (MDMA)'s O-demethylenated metabolite, 3,4-dihydroxymethamphetamine (HHMA), has been hypothesized to serve as a precursor for the formation of toxic catechol-thioether metabolites (e.g., 5-N-acetylcystein-S-yl-HHMA) that mediate MDMA neurotoxicity. To further test this hypothesis, HHMA formation was blocked with dextromethorphan (DXM), which competitively inhibits cytochrome P450 enzyme-mediated O-demethylenation of MDMA to HHMA. In particular, rats were randomly assigned to one of four treatment groups (n = 9-12 per group): (1) Saline/MDMA; (2) DXM/MDMA; (3) DXM/Saline; (4) Saline/Saline. During drug exposure, time-concentration profiles of MDMA and its metabolites were determined, along with body temperature. One week later, brain serotonin (5-HT) neuronal markers were measured in the same animals. DXM did not significantly alter core temperature in MDMA-treated animals. A large (greater than 70%) decrease in HHMA formation had no effect on the magnitude of MDMA neurotoxicity. These results cast doubt on the role of HHMA-derived catechol-thioether metabolites in the mechanism of MDMA neurotoxicity.


Subject(s)
3,4-Methylenedioxyamphetamine/analogs & derivatives , Deoxyepinephrine/analogs & derivatives , Neurotoxicity Syndromes/metabolism , Neurotoxins/toxicity , Serotonin/toxicity , 3,4-Methylenedioxyamphetamine/antagonists & inhibitors , 3,4-Methylenedioxyamphetamine/pharmacokinetics , 3,4-Methylenedioxyamphetamine/toxicity , Animals , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Deoxyepinephrine/antagonists & inhibitors , Deoxyepinephrine/pharmacokinetics , Deoxyepinephrine/toxicity , Dextromethorphan/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Hydroxyindoleacetic Acid/metabolism , Male , Neurotoxins/metabolism , Rats , Rats, Sprague-Dawley , Serotonin/metabolism , Sulfides/chemistry , Sulfides/metabolism
10.
Drug Alcohol Depend ; 117(1): 38-44, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21296508

ABSTRACT

BACKGROUND: Sleep difficulty is a common symptom of cannabis withdrawal, but little research has objectively measured sleep or explored the effects of hypnotic medication on sleep during cannabis withdrawal. METHODS: Twenty daily cannabis users completed a within-subject crossover study. Participants alternated between periods of ad libitum cannabis use and short-term cannabis abstinence (3 days). Placebo was administered at bedtime during one abstinence period (withdrawal test) and extended-release zolpidem, a non-benzodiazepine GABA(A) receptor agonist, was administered during the other. Polysomnographic (PSG) sleep architecture measures, subjective ratings, and cognitive performance effects were assessed each day. RESULTS: During the placebo-abstinence period, participants had decreased sleep efficiency, total sleep time, percent time spent in Stage 1 and Stage 2 sleep, REM latency and subjective sleep quality, as well as increased sleep latency and time spent in REM sleep compared with when they were using cannabis. Zolpidem attenuated the effects of abstinence on sleep architecture and normalized sleep efficiency scores, but had no effect on sleep latency. Zolpidem was not associated with any significant side effects or next-day cognitive performance impairments. CONCLUSIONS: These data extend prior research that indicates abrupt abstinence from cannabis can lead to clinically significant sleep disruption in daily users. The findings also indicate that sleep disruption associated with cannabis withdrawal can be attenuated by zolpidem, suggesting that hypnotic medications might be useful adjunct pharmacotherapies in the treatment of cannabis use disorders.


Subject(s)
Cannabinoids/pharmacology , Hypnotics and Sedatives/therapeutic use , Pyridines/therapeutic use , Sleep Initiation and Maintenance Disorders/drug therapy , Sleep/drug effects , Substance Withdrawal Syndrome/pathology , Adult , Cannabis/metabolism , Control Groups , Cross-Over Studies , Delayed-Action Preparations , Double-Blind Method , Female , Humans , Male , Middle Aged , Placebos , Polysomnography , Sleep Initiation and Maintenance Disorders/complications , Sleep, REM/drug effects , Substance Withdrawal Syndrome/complications , Substance Withdrawal Syndrome/metabolism , Young Adult , Zolpidem
11.
J Clin Psychiatry ; 72(4): 539-47, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21034691

ABSTRACT

OBJECTIVE: Previous studies have suggested a link between heart rate (HR) following trauma and the development of posttraumatic stress disorder (PTSD). This study expands on previous work by evaluating HR in burn patients followed longitudinally for symptoms of acute stress disorder (ASD) and PTSD. METHOD: Data were collected from consecutive patients admitted to the Johns Hopkins Burn Center, Baltimore, Maryland, between 1997 and 2002. Patients completed the Stanford Acute Stress Reaction Questionnaire (n = 157) to assess symptoms of ASD. The Davidson Trauma Scale was completed at 1 (n = 145), 6 (n = 106), 12 (n = 94), and 24 (n = 66) months postdischarge to assess symptoms of PTSD. Heart rate in the ambulance, emergency room, and burn unit were obtained by retrospective medical chart review. RESULTS: Pearson correlations revealed a significant relationship between HR in the ambulance (r = 0.32, P = .016) and burn unit (r = 0.30, P = .001) and ASD scores at baseline. Heart rate in the ambulance was related to PTSD avoidance cluster scores at 1, 6, 12, and 24 months. In women, HR in the ambulance was correlated with PTSD scores at 6 (r = 0.65, P = .005) and 12 (r = 0.78, P = .005) months. When covariates (gender, ß-blockers, Brief Symptom Inventory Global Severity Index score) were included in multivariate linear regression analyses, ambulance HR was associated with ASD and PTSD scores at baseline and 1 month, and the interaction of ambulance HR and gender was associated with PTSD scores at 6 and 12 months. Multivariate logistic regression results were similar at baseline and 12 months, which included an HR association yet no interaction at 6 months and a marginal interaction at 1 month. CONCLUSIONS: While peritraumatic HR is most robustly associated with PTSD symptom severity, HR on admission to burn unit also predicts the development of ASD. Gender and avoidance symptoms appear particularly salient in this relationship, and these factors may aid in the identification of subgroups for which HR serves as a biomarker for PTSD. Future work may identify endophenotypic measures of increased risk for PTSD, targeting subgroups for early intervention.


Subject(s)
Burns/psychology , Heart Rate/physiology , Stress Disorders, Post-Traumatic/etiology , Adult , Burns/physiopathology , Female , Humans , Injury Severity Score , Linear Models , Logistic Models , Male , Psychiatric Status Rating Scales , Retrospective Studies , Stress Disorders, Post-Traumatic/physiopathology , Stress Disorders, Post-Traumatic/psychology , Surveys and Questionnaires
12.
J Neurochem ; 114(4): 1135-42, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20533999

ABSTRACT

It is widely believed that dopamine (DA) mediates methamphetamine (METH)-induced toxicity to brain dopaminergic neurons, because drugs that interfere with DA neurotransmission decrease toxicity, whereas drugs that increase DA neurotransmission enhance toxicity. However, temperature effects of drugs that have been used to manipulate brain DA neurotransmission confound interpretation of the data. Here we show that the recently reported ability of L-dihydroxyphenylalanine to reverse the protective effect of alpha-methyl-para-tyrosine on METH-induced DA neurotoxicity is also confounded by drug effects on body temperature. Further, we show that mice genetically engineered to be deficient in brain DA develop METH neurotoxicity, as long as the thermic effects of METH are preserved. In addition, we demonstrate that mice genetically engineered to have unilateral brain DA deficits develop METH-induced dopaminergic deficits that are of comparable magnitude on both sides of the brain. Taken together, these findings demonstrate that DA is not essential for the development of METH-induced dopaminergic neurotoxicity and suggest that mechanisms independent of DA warrant more intense investigation.


Subject(s)
Dopamine/deficiency , Methamphetamine/toxicity , Neurotoxicity Syndromes/etiology , Adrenergic Agents/toxicity , Amphetamine-Related Disorders/complications , Amphetamine-Related Disorders/metabolism , Amphetamine-Related Disorders/physiopathology , Animals , Body Temperature/drug effects , Body Temperature/physiology , Dihydroxyphenylalanine/pharmacology , Disease Models, Animal , Dopamine/genetics , Functional Laterality/drug effects , Functional Laterality/physiology , Male , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/antagonists & inhibitors , Neurotoxicity Syndromes/genetics , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , alpha-Methyltyrosine/antagonists & inhibitors
13.
Neurology ; 73(23): 2011-7, 2009 Dec 08.
Article in English | MEDLINE | ID: mdl-19955499

ABSTRACT

BACKGROUND: Methylenedioxymethamphetamine (MDMA, "ecstasy") is a popular recreational drug of abuse and a selective brain serotonin neurotoxin. Functional consequences of MDMA neurotoxicity have defied ready characterization. Obstructive sleep apnea (OSA) is a common form of sleep-disordered breathing in which brain serotonin dysfunction may play a role. The present study sought to determine whether abstinent recreational MDMA users have an increased prevalence of OSA. METHODS: We studied 71 medically healthy recreational MDMA users and 62 control subjects using all-night sleep polysomnography in a controlled inpatient research setting. Rates of apneas, hypopneas, and apnea hypopnea indices were compared in the 2 groups, controlling for body mass index, age, race, and gender. RESULTS: Recreational MDMA users who had been drug free for at least 2 weeks had significantly increased rates of obstructive sleep apnea and hypopnea compared with controls. The odds ratio (95% confidence interval) for sleep apnea (mild, moderate, and severe combined) in MDMA users during non-REM sleep was 8.5 (2.4-30.4), which was greater than that associated with obesity [6.9 (1.7-28.2)]. Severity of OSA was significantly related to lifetime MDMA exposure. CONCLUSIONS: These findings suggest that prior recreational methylenedioxymethamphetamine use increases the risk for obstructive sleep apnea and lend support to the notion that brain serotonin neuronal dysfunction plays a role in the pathophysiology of sleep apnea.


Subject(s)
Illicit Drugs , N-Methyl-3,4-methylenedioxyamphetamine , Sleep Apnea Syndromes/etiology , Substance-Related Disorders/complications , Substance-Related Disorders/etiology , Adolescent , Adult , Age Factors , Female , Humans , Illicit Drugs/adverse effects , Male , Middle Aged , N-Methyl-3,4-methylenedioxyamphetamine/adverse effects , Polysomnography , Serotonin/physiology , Sleep Apnea Syndromes/physiopathology , Substance-Related Disorders/physiopathology , Young Adult
15.
J Neurosci ; 29(44): 14050-6, 2009 Nov 04.
Article in English | MEDLINE | ID: mdl-19890014

ABSTRACT

Methylenedioxymethamphetamine (MDMA; "Ecstasy") is a popular recreational drug and brain serotonin (5-HT) neurotoxin. Neuroimaging data indicate that some human MDMA users develop persistent deficits in brain 5-HT neuronal markers. Although the consequences of MDMA-induced 5-HT neurotoxicity are not fully understood, abstinent MDMA users have been found to have subtle cognitive deficits and altered sleep architecture. The present study sought to test the hypothesis that sleep disturbance plays a role in cognitive deficits in MDMA users. Nineteen abstinent MDMA users and 21 control subjects participated in a 5 d inpatient study in a clinical research unit. Baseline sleep quality was measured using the Pittsburgh Sleep Quality Inventory. Cognitive performance was tested three times daily using a computerized cognitive battery. On the third day of admission, subjects began a 40 h sleep deprivation period and continued cognitive testing using the same daily schedule. At baseline, MDMA users performed less accurately than controls on a task of working memory and more impulsively on four of the seven computerized tests. During sleep deprivation, MDMA users, but not controls, became increasingly impulsive, performing more rapidly at the expense of accuracy on tasks of working and short-term memory. Tests of mediation implicated baseline sleep disturbance in the cognitive decline seen during sleep deprivation. These findings are the first to demonstrate that memory problems in MDMA users may be related, at least in part, to sleep disturbance and suggest that cognitive deficits in MDMA users may become more prominent in situations associated with sleep deprivation.


Subject(s)
Cognition Disorders/physiopathology , N-Methyl-3,4-methylenedioxyamphetamine , Psychomotor Performance/physiology , Sleep Deprivation/physiopathology , Substance-Related Disorders/physiopathology , Adult , Cognition/physiology , Cognition Disorders/etiology , Female , Humans , Male , N-Methyl-3,4-methylenedioxyamphetamine/administration & dosage , Sleep Deprivation/complications , Substance-Related Disorders/complications , Young Adult
16.
Drug Metab Dispos ; 37(10): 2079-86, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19628751

ABSTRACT

The mechanism by which the recreational drug (+/-)-3,4-methylenedioxymethamphetamine (MDMA) destroys brain serotonin (5-HT) axon terminals is not understood. Recent studies have implicated MDMA metabolites, but their precise role remains unclear. To further evaluate the relative importance of metabolites versus the parent compound in neurotoxicity, we explored the relationship between pharmacokinetic parameters of MDMA, 3,4-methylenedioxyamphetamine (MDA), 3,4-dihydroxymethamphetamine (HHMA), and 4-hydroxy-3-methoxymethamphetamine (HMMA) and indexes of serotonergic neurotoxicity in the same animals. We also further evaluated the neurotoxic potential of 5-(N-acetylcystein-S-yl)-HHMA (5-NAC-HHMA), an MDMA metabolite recently implicated in 5-HT neurotoxicity. Lasting serotonergic deficits correlated strongly with pharmacokinetic parameters of MDMA (C(max) and area under the concentration-time curve), more weakly with those of MDA, and not at all with those of HHMA or HMMA (total amounts of the free analytes obtained after conjugate cleavage). HHMA and HMMA could not be detected in the brains of animals with high brain MDMA concentrations and high plasma HHMA and HMMA concentrations, suggesting that HHMA and HMMA do not readily penetrate the blood-brain barrier (either in their free form or as sulfate or glucuronic conjugates) and that little or no MDMA is metabolized to HHMA or HMMA in the brain. Repeated intraparenchymal administration of 5-NAC-HHMA did not produce significant lasting serotonergic deficits in the rat brain. Taken together, these results indicate that MDMA and, possibly, MDA are more important determinants of brain 5-HT neurotoxicity in the rat than HHMA and HMMA and bring into question the role of metabolites (including 5-NAC-HHMA) in MDMA neurotoxicity.


Subject(s)
N-Methyl-3,4-methylenedioxyamphetamine/metabolism , Neurotoxicity Syndromes/complications , Serotonin/metabolism , 3,4-Methylenedioxyamphetamine/metabolism , 3,4-Methylenedioxyamphetamine/pharmacology , Animals , Deoxyepinephrine/analogs & derivatives , Deoxyepinephrine/metabolism , Deoxyepinephrine/pharmacology , Disease Models, Animal , Male , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Neurotoxicity Syndromes/blood , Neurotoxicity Syndromes/urine , Rats , Rats, Sprague-Dawley
17.
Ther Drug Monit ; 31(3): 367-73, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19417716

ABSTRACT

The present study compared the disposition and metabolism of the recreational drug (+/-) 3,4-methylenedioxymethamphetamine (MDMA, "ecstasy") in squirrel monkeys and humans because the squirrel monkey has been extensively studied for MDMA neurotoxicity. A newly developed liquid chromatography-mass spectrometric procedure for simultaneous measurement of MDMA, 3,4-dihydroxymethamphetamine, 4-hydroxy-3-methoxymethamphetamine, and 3,4-methylenedioxyamphetamine was employed. In both humans and squirrel monkeys, a within-subject design permitted testing of different doses in the same subjects. Humans and squirrel monkeys were found to metabolize MDMA in similar, but not identical, pathways and proportions. In particular, amounts of 3,4-dihydroxymethamphetamine (after conjugate cleavage) and 3,4-methylenedioxyamphetamine were similar in the 2 species, but formation of 4-hydroxy-3-methoxymethamphetamine was greater in squirrel monkeys than in humans. Both species demonstrated nonlinear MDMA pharmacokinetics at comparable plasma MDMA concentrations (125-150 ng/mL and above). The elimination half-life of MDMA was considerably shorter in squirrel monkeys than in humans (2-3 versus 6-9 hours). In both species, there was substantial individual variability. These results suggest that the squirrel monkey may be a useful model for predicting outcomes of MDMA exposure in humans, although this will also depend on the degree to which MDMA pharmacodynamics in the squirrel monkey parallels that in humans.


Subject(s)
3,4-Methylenedioxyamphetamine/blood , Brain/metabolism , N-Methyl-3,4-methylenedioxyamphetamine/blood , Adolescent , Animals , Chromatography, High Pressure Liquid , Drug Monitoring , Female , Haplorhini , Humans , Male , Metabolic Networks and Pathways , N-Methyl-3,4-methylenedioxyamphetamine/pharmacokinetics , Neurotoxicity Syndromes , Saimiri , Serotonin/metabolism , Spectrometry, Mass, Electrospray Ionization , Young Adult
18.
J Pharmacol Exp Ther ; 327(1): 38-44, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18591215

ABSTRACT

At certain doses, the psychoactive drug (+/-)3,4-methylenedioxymethamphetamine (MDMA, "Ecstasy") destroys brain serotonin axon terminals. By causing increases in plasma MDMA concentrations that exceed those predicted by the increase in dose, nonlinear pharmacokinetics has the potential to narrow the range between safe and neurotoxic doses of MDMA. The present study sought to determine whether the pharmacokinetics of MDMA in nonhuman primates are nonlinear and, if they are, to identify plasma concentrations of MDMA at which nonlinear accumulation of MDMA occurs. Four different oral doses of MDMA were tested in the same six squirrel monkeys in random order. At each dose, pharmacokinetic parameters for MDMA and its metabolites 3,4-dihydroxymethamphetamine (HHMA), 4-hydroxy-3-methoxymethamphetamine (HMMA), and 3,4-methylenedioxyamphetamine were determined. Doses were selected to be equivalent to 0.4, 0.8, 1.6, and 2.8 mg/kg doses in humans. The maximal concentration (C(max)) and area under the curve (AUC) of MDMA increased nonlinearly with dose, whereas the C(max) and AUC of the metabolites HHMA and HMMA remained relatively constant. Nonlinear MDMA pharmacokinetics occurred at plasma MDMA concentrations of 100 to 300 ng/ml and above. The half-life (T(1/2)) of MDMA and its metabolites also increased with dose. These results firmly establish nonlinear pharmacokinetics for MDMA in squirrel monkeys and indicate that nonlinear MDMA accumulation occurs at plasma MDMA concentrations that develop in humans taking typical doses. By raising MDMA concentrations and prolonging its action, nonlinear pharmacokinetics and T(1/2) prolongation, respectively, may influence the likelihood and severity of MDMA toxicities (including brain serotonin neurotoxicity).


Subject(s)
Deoxyepinephrine/analogs & derivatives , Methamphetamine/analogs & derivatives , N-Methyl-3,4-methylenedioxyamphetamine/pharmacokinetics , Animals , Area Under Curve , Deoxyepinephrine/pharmacokinetics , Half-Life , Male , Methamphetamine/pharmacokinetics , Saimiri , Species Specificity
19.
Psychopharmacology (Berl) ; 200(3): 439-50, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18661256

ABSTRACT

BACKGROUND: (+/-)3,4-Methylenedioxymethamphetamine (MDMA, "ecstasy") is a recreational drug and brain serotonin (5-HT) neurotoxin. Under certain conditions, MDMA can also damage brain dopamine (DA) neurons, at least in rodents. Human MDMA users have been found to have reduced brain 5-HT transporter (SERT) density and cognitive deficits, although it is not known whether these are related. This study sought to determine whether MDMA users who take closely spaced sequential doses, which engender high plasma MDMA concentrations, develop DA transporter (DAT) deficits, in addition to SERT deficits, and whether there is a relationship between transporter binding and cognitive performance. MATERIALS AND METHODS: Sixteen abstinent MDMA users with a history of using sequential MDMA doses (two or more doses over a 3- to 12-h period) and 16 age-, gender-, and education-matched controls participated. Subjects underwent positron emission tomography with the DAT and SERT radioligands, [11C]WIN 35,428 and [11C]DASB, respectively. Subjects also underwent formal neuropsychiatric testing. RESULTS: MDMA users had reductions in SERT binding in multiple brain regions but no reductions in striatal DAT binding. Memory performance in the aggregate subject population was correlated with SERT binding in the dorsolateral prefrontal cortex, orbitofrontal cortex, and parietal cortex, brain regions implicated in memory function. Prior exposure to MDMA significantly diminished the strength of this relationship. CONCLUSIONS: Use of sequential MDMA doses is associated with lasting decreases in brain SERT, but not DAT. Memory performance is associated with SERT binding in brain regions involved in memory function. Prior MDMA exposure appears to disrupt this relationship. These data are the first to directly relate memory performance to brain SERT density.


Subject(s)
Amphetamine-Related Disorders/diagnostic imaging , Brain/diagnostic imaging , Cognition/drug effects , Dopamine Plasma Membrane Transport Proteins/metabolism , N-Methyl-3,4-methylenedioxyamphetamine , Positron-Emission Tomography , Serotonin Agents , Serotonin Plasma Membrane Transport Proteins/metabolism , Adolescent , Adult , Amphetamine-Related Disorders/rehabilitation , Brain/drug effects , Brain Mapping , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/drug effects , Female , Humans , Male , Mental Recall/drug effects , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Neuropsychological Tests , Serotonin Agents/toxicity , Young Adult
20.
Synapse ; 62(2): 91-100, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17992686

ABSTRACT

BACKGROUND: Studies in abstinent methamphetamine (METH) users have demonstrated reductions in brain dopamine transporter (DAT) binding potential (BP), as well as cognitive and motor deficits, but it is not yet clear whether cognitive deficits and brain DAT reductions fully reverse with sustained abstinence, or whether behavioral deficits in METH users are related to dopamine (DA) deficits. This study was conducted to further investigate potential persistent psychomotor deficits secondary to METH abuse, and their relationship to brain DAT availability, as measured using quantitative PET methods with [(11)C]WIN 35428. METHODS: Twenty-two abstinent METH users and 17 healthy non-METH using controls underwent psychometric testing to test the hypothesis that METH users would demonstrate selective deficits in neuropsychiatric domains known to involve DA neurons (e.g., working memory, executive function, motor function). A subset of subjects also underwent PET scanning with [(11)C]WIN 35428. RESULTS: METH users were found to have modest deficits in short-term memory, executive function, and manual dexterity. Exploratory correlational analyses revealed that deficits in memory, but not those in executive or motor function, were associated with decreases in striatal DAT BP. CONCLUSIONS: These results suggest a possible relationship between DAT BP and memory deficits in abstinent METH users, and lend support to the notion that METH produces lasting effects on central DA neurons in humans. As METH can also produce toxic effects on serotonin (5-HT) neurons, further study is needed to address the potential role of brain 5-HT depletion in cognitive deficits in abstinent METH users.


Subject(s)
Amphetamine-Related Disorders/metabolism , Amphetamine-Related Disorders/physiopathology , Cognition/drug effects , Dopamine Uptake Inhibitors/pharmacology , Dopamine/metabolism , Methamphetamine/administration & dosage , Adult , Amphetamine-Related Disorders/diagnostic imaging , Analysis of Variance , Cocaine/analogs & derivatives , Cocaine/metabolism , Corpus Striatum/diagnostic imaging , Corpus Striatum/drug effects , Female , Humans , Male , Middle Aged , Neuropsychological Tests , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/diagnostic imaging , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/physiopathology , Positron-Emission Tomography/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...