Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Psychosom Med ; 81(4): 352-362, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30855555

RESUMO

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.


Assuntos
Plaquetas/metabolismo , Doenças Cardiovasculares/complicações , Depressão/complicações , Serotonina/metabolismo , Doenças Cardiovasculares/sangue , Doenças Cardiovasculares/psicologia , Depressão/sangue , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Ativação Plaquetária , Receptores de Serotonina/metabolismo
2.
J Pharmacol Exp Ther ; 344(2): 479-88, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23209329

RESUMO

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.


Assuntos
Hipotermia Induzida , N-Metil-3,4-Metilenodioxianfetamina/metabolismo , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/prevenção & controle , Administração Oral , Animais , Biotransformação , Temperatura Corporal , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Dopamina/metabolismo , Relação Dose-Resposta a Droga , Masculino , Camundongos , Camundongos Endogâmicos , N-Metil-3,4-Metilenodioxianfetamina/farmacocinética , N-Metil-3,4-Metilenodioxianfetamina/toxicidade , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Síndromes Neurotóxicas/etiologia , Ratos , Ratos Sprague-Dawley , Serotonina/metabolismo , Especificidade da Espécie , Fatores de Tempo , Distribuição Tecidual
3.
Int J Neuropsychopharmacol ; 16(4): 791-801, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-22824226

RESUMO

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.


Assuntos
Encéfalo/metabolismo , N-Metil-3,4-Metilenodioxianfetamina/administração & dosagem , N-Metil-3,4-Metilenodioxianfetamina/metabolismo , Serotoninérgicos/administração & dosagem , Serotoninérgicos/metabolismo , Serotonina/metabolismo , Administração Oral , Animais , Encéfalo/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Masculino , N-Metil-3,4-Metilenodioxianfetamina/sangue , Primatas , Saimiri , Serotoninérgicos/sangue
4.
J Neuropsychiatry Clin Neurosci ; 24(3): 309-15, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23037644

RESUMO

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.


Assuntos
Lesões Encefálicas/complicações , Encéfalo/patologia , Transtorno Depressivo Maior/diagnóstico , Transtorno Depressivo Maior/etiologia , Imagem de Tensor de Difusão , Adulto , Anisotropia , Feminino , Seguimentos , Escala de Coma de Glasgow , Humanos , Processamento de Imagem Assistida por Computador , Modelos Lineares , Masculino , Pessoa de Meia-Idade , Escalas de Graduação Psiquiátrica , Adulto Jovem
5.
J Pharmacol Exp Ther ; 338(1): 310-7, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21493752

RESUMO

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.


Assuntos
N-Metil-3,4-Metilenodioxianfetamina/administração & dosagem , N-Metil-3,4-Metilenodioxianfetamina/sangue , Papio hamadryas/sangue , Administração Oral , Animais , Relação Dose-Resposta a Droga , Humanos , Masculino , N-Metil-3,4-Metilenodioxianfetamina/farmacocinética , Especificidade da Espécie , Distribuição Tecidual/efeitos dos fármacos , Distribuição Tecidual/fisiologia
6.
Synapse ; 65(10): 983-90, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21360595

RESUMO

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.


Assuntos
3,4-Metilenodioxianfetamina/análogos & derivados , Desoxiepinefrina/análogos & derivados , Síndromes Neurotóxicas/metabolismo , Neurotoxinas/toxicidade , Serotonina/toxicidade , 3,4-Metilenodioxianfetamina/antagonistas & inibidores , 3,4-Metilenodioxianfetamina/farmacocinética , 3,4-Metilenodioxianfetamina/toxicidade , Animais , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450/metabolismo , Desoxiepinefrina/antagonistas & inibidores , Desoxiepinefrina/farmacocinética , Desoxiepinefrina/toxicidade , Dextrometorfano/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Ácido Hidroxi-Indolacético/metabolismo , Masculino , Neurotoxinas/metabolismo , Ratos , Ratos Sprague-Dawley , Serotonina/metabolismo , Sulfetos/química , Sulfetos/metabolismo
7.
J Neurosci ; 29(44): 14050-6, 2009 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-19890014

RESUMO

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.


Assuntos
Transtornos Cognitivos/fisiopatologia , N-Metil-3,4-Metilenodioxianfetamina , Desempenho Psicomotor/fisiologia , Privação do Sono/fisiopatologia , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , Adulto , Cognição/fisiologia , Transtornos Cognitivos/etiologia , Feminino , Humanos , Masculino , N-Metil-3,4-Metilenodioxianfetamina/administração & dosagem , Privação do Sono/complicações , Transtornos Relacionados ao Uso de Substâncias/complicações , Adulto Jovem
8.
J Neurochem ; 114(4): 1135-42, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20533999

RESUMO

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.


Assuntos
Dopamina/deficiência , Metanfetamina/toxicidade , Síndromes Neurotóxicas/etiologia , Adrenérgicos/toxicidade , Transtornos Relacionados ao Uso de Anfetaminas/complicações , Transtornos Relacionados ao Uso de Anfetaminas/metabolismo , Transtornos Relacionados ao Uso de Anfetaminas/fisiopatologia , Animais , Temperatura Corporal/efeitos dos fármacos , Temperatura Corporal/fisiologia , Di-Hidroxifenilalanina/farmacologia , Modelos Animais de Doenças , Dopamina/genética , Lateralidade Funcional/efeitos dos fármacos , Lateralidade Funcional/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/antagonistas & inibidores , Síndromes Neurotóxicas/genética , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , alfa-Metiltirosina/antagonistas & inibidores
9.
Drug Metab Dispos ; 37(10): 2079-86, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19628751

RESUMO

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.


Assuntos
N-Metil-3,4-Metilenodioxianfetamina/metabolismo , Síndromes Neurotóxicas/complicações , Serotonina/metabolismo , 3,4-Metilenodioxianfetamina/metabolismo , 3,4-Metilenodioxianfetamina/farmacologia , Animais , Desoxiepinefrina/análogos & derivados , Desoxiepinefrina/metabolismo , Desoxiepinefrina/farmacologia , Modelos Animais de Doenças , Masculino , N-Metil-3,4-Metilenodioxianfetamina/toxicidade , Síndromes Neurotóxicas/sangue , Síndromes Neurotóxicas/urina , Ratos , Ratos Sprague-Dawley
10.
Ther Drug Monit ; 31(3): 367-73, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19417716

RESUMO

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.


Assuntos
3,4-Metilenodioxianfetamina/sangue , Encéfalo/metabolismo , N-Metil-3,4-Metilenodioxianfetamina/sangue , Adolescente , Animais , Cromatografia Líquida de Alta Pressão , Monitoramento de Medicamentos , Feminino , Haplorrinos , Humanos , Masculino , Redes e Vias Metabólicas , N-Metil-3,4-Metilenodioxianfetamina/farmacocinética , Síndromes Neurotóxicas , Saimiri , Serotonina/metabolismo , Espectrometria de Massas por Ionização por Electrospray , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA