Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Anesthesiology ; 125(1): 133-46, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27183169

RESUMO

BACKGROUND: Animals exposed to sevoflurane during development sustain neuronal cell death in their developing brains. In vivo micro-positron emission tomography (PET)/computed tomography imaging has been utilized as a minimally invasive method to detect anesthetic-induced neuronal adverse effects in animal studies. METHODS: Neonatal rhesus monkeys (postnatal day 5 or 6, 3 to 6 per group) were exposed for 8 h to 2.5% sevoflurane with or without acetyl-L-carnitine (ALC). Control monkeys were exposed to room air with or without ALC. Physiologic status was monitored throughout exposures. Depth of anesthesia was monitored using quantitative electroencephalography. After the exposure, microPET/computed tomography scans using F-labeled fluoroethoxybenzyl-N-(4-phenoxypyridin-3-yl) acetamide (FEPPA) were performed repeatedly on day 1, 1 and 3 weeks, and 2 and 6 months after exposure. RESULTS: Critical physiologic metrics in neonatal monkeys remained within the normal range during anesthetic exposures. The uptake of [F]-FEPPA in the frontal and temporal lobes was increased significantly 1 day or 1 week after exposure, respectively. Analyses of microPET images recorded 1 day after exposure showed that sevoflurane exposure increased [F]-FEPPA uptake in the frontal lobe from 0.927 ± 0.04 to 1.146 ± 0.04, and in the temporal lobe from 0.859 ± 0.05 to 1.046 ± 0.04 (mean ± SE, P < 0.05). Coadministration of ALC effectively blocked the increase in FEPPA uptake. Sevoflurane-induced adverse effects were confirmed by histopathologic evidence as well. CONCLUSIONS: Sevoflurane-induced general anesthesia during development increases glial activation, which may serve as a surrogate for neurotoxicity in the nonhuman primate brain. ALC is a potential protective agent against some of the adverse effects associated with such exposures.


Assuntos
Anestésicos Inalatórios/efeitos adversos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/diagnóstico por imagem , Éteres Metílicos/efeitos adversos , Tomografia por Emissão de Pósitrons/métodos , Anestesia Geral , Anilidas , Animais , Animais Recém-Nascidos , Eletroencefalografia/efeitos dos fármacos , Feminino , Lobo Frontal/diagnóstico por imagem , Lobo Frontal/metabolismo , Processamento de Imagem Assistida por Computador , Macaca mulatta , Masculino , Piridinas , Compostos Radiofarmacêuticos , Sevoflurano , Lobo Temporal/diagnóstico por imagem , Lobo Temporal/metabolismo , Tomografia Computadorizada por Raios X
2.
J Appl Toxicol ; 35(3): 261-72, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24871937

RESUMO

Typically, time-consuming standard toxicological assays using the zebrafish (Danio rerio) embryo model evaluate mortality and teratogenicity after exposure during the first 2 days post-fertilization. Here we describe an automated image-based high content screening (HCS) assay to identify the teratogenic/embryotoxic potential of compounds in zebrafish embryos in vivo. Automated image acquisition was performed using a high content microscope system. Further automated analysis of embryo length, as a statistically quantifiable endpoint of toxicity, was performed on images post-acquisition. The biological effects of ethanol, nicotine, ketamine, caffeine, dimethyl sulfoxide and temperature on zebrafish embryos were assessed. This automated developmental toxicity assay, based on a growth-retardation endpoint should be suitable for evaluating the effects of potential teratogens and developmental toxicants in a high throughput manner. This approach can significantly expedite the screening of potential teratogens and developmental toxicants, thereby improving the current risk assessment process by decreasing analysis time and required resources.


Assuntos
Embrião não Mamífero/efeitos dos fármacos , Desenvolvimento Embrionário/efeitos dos fármacos , Ensaios de Triagem em Larga Escala/métodos , Teratogênicos/toxicidade , Testes de Toxicidade/métodos , Peixe-Zebra/embriologia , Animais , Embrião não Mamífero/patologia , Processamento de Imagem Assistida por Computador
3.
Front Neurol ; 5: 234, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25452743

RESUMO

Neural progenitor cell expansion is critical for normal brain development and an appropriate response to injury. During the brain growth spurt, exposures to general anesthetics, which either block the N-methyl-d-aspartate receptor or enhance the γ-aminobutyric acid receptor type A can disturb neuronal transduction. This effect can be detrimental to brain development. Until now, the effects of anesthetic exposure on neural progenitor cell expansion in vivo had seldom been reported. Here, minimally invasive micro positron emission tomography (microPET) coupled with 3'-deoxy-3' [(18)F] fluoro-l-thymidine ([(18)F]FLT) was utilized to assess the effects of sevoflurane exposure on neural progenitor cell proliferation. FLT, a thymidine analog, is taken up by proliferating cells and phosphorylated in the cytoplasm, leading to its intracellular trapping. Intracellular retention of [(18)F]FLT, thus, represents an observable in vivo marker of cell proliferation. Here, postnatal day 7 rats (n = 11/group) were exposed to 2.5% sevoflurane or room air for 9 h. For up to 2 weeks following the exposure, standard uptake values (SUVs) for [(18)F]-FLT in the hippocampal formation were significantly attenuated in the sevoflurane-exposed rats (p < 0.0001), suggesting decreased uptake and retention of [(18)F]FLT (decreased proliferation) in these regions. Four weeks following exposure, SUVs for [(18)F]FLT were comparable in the sevoflurane-exposed rats and in controls. Co-administration of 7-nitroindazole (30 mg/kg, n = 5), a selective inhibitor of neuronal nitric oxide synthase, significantly attenuated the SUVs for [(18)F]FLT in both the air-exposed (p = 0.00006) and sevoflurane-exposed rats (p = 0.0427) in the first week following the exposure. These findings suggested that microPET in couple with [(18)F]FLT as cell proliferation marker could be used as a non-invasive modality to monitor the sevoflurane-induced inhibition of neural progenitor cell proliferation in vivo.

4.
Nanomedicine (Lond) ; 7(6): 835-46, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22339089

RESUMO

AIM: The purpose of the current study was to determine whether copper nanoparticles (Cu-NPs) can induce the release of proinflammatory mediators that influence the restrictive characteristics of the blood-brain barrier. MATERIAL & METHODS: Confluent rat brain microvessel endothelial cells (rBMECs) were treated with well-characterized Cu-NPs (40 or 60 nm). Cytotoxicity of the Cu-NPs was evaluated by cell proliferation assay (1.5-50 µg/ml). The extracellular concentrations of proinflammatory mediators (IL-1ß, IL-2, TNF-α and prostaglandin E(2)) were evaluated by ELISA. RESULTS: The exposure of Cu-NPs at low concentrations increases cellular proliferation of rBMECs, by contrast, high concentrations induce toxicity. Prostaglandin E(2) release was significantly increased (threefold; 8 h) for Cu-NPs (40 and 60 nm). The extracellular levels of both TNF-α and IL-1ß were significantly elevated following exposure to Cu-NPs. The P-apparent ratio, as an indicator of increased permeability of rBMEC was approximately twofold for Cu-NPs (40 and 60 nm). CONCLUSION: These data suggest that Cu-NPs can induce rBMEC, proliferation at low concentrations and/or induce blood-brain barrier toxicity and potential neurotoxicity at high concentrations.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/imunologia , Cobre/imunologia , Nanopartículas/química , Animais , Barreira Hematoencefálica/citologia , Permeabilidade da Membrana Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Cobre/química , Cobre/toxicidade , Dinoprostona/imunologia , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Interleucina-1beta/imunologia , Interleucina-2/imunologia , Nanopartículas/toxicidade , Nanopartículas/ultraestrutura , Fragmentos de Peptídeos/imunologia , Ratos , Fator de Necrose Tumoral alfa/imunologia
5.
Neurotoxicol Teratol ; 33(2): 288-96, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21216281

RESUMO

Amyloid-beta peptide (Aß) deposition is assumed to play a pathogenic role in the brain of Alzheimer's disease patients. To date, the precise mechanisms underlying Aß toxicity are not fully understood. A recent hypothesis suggesting that the Receptor-for-Advanced-Glycation-End-Products (RAGE)-a trans-membrane protein signaling for oxidative stress-is involved in Aß toxicity is gaining attention. Early Aß toxicity could indeed help to explain the deleterious events further produced by this molecule in the brain. In this work, we evaluated the pattern of early expression of RAGE in the toxic model induced by Aß25₋35 in rat CA1 region. Intrahippocampal injections of Aß25₋35 in rats increased the RAGE expression at 24 h post-injection; this event was accompanied by increased components of RAGE downstream signaling in hippocampal cells, such as enhanced expression of the pro-apoptotic factor NF-κB, increased nitric oxide production, LDH leakage, mitochondrial dysfunction, increased TNF-α expression, antioxidant genes down-regulation, and augmented neurodegeneration. Our findings support an active role of RAGE during the early stages of Aß25₋35 toxicity in the hippocampus.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Região CA1 Hipocampal/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Fragmentos de Peptídeos/toxicidade , Receptores Imunológicos/genética , Animais , Antioxidantes/metabolismo , Western Blotting , Região CA1 Hipocampal/enzimologia , Região CA1 Hipocampal/metabolismo , Região CA1 Hipocampal/patologia , Morte Celular/efeitos dos fármacos , Masculino , Microinjeções , Microscopia de Fluorescência , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Óxido Nítrico/biossíntese , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Ratos , Ratos Sprague-Dawley , Receptor para Produtos Finais de Glicação Avançada , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Nanotoxicology ; 5(4): 479-92, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21175299

RESUMO

This report examined blood-brain barrier (BBB) related proinflammatory mediators and permeability changes in response to various sized gold nanoparticles (Au-NPs) (3, 5, 7, 10, 30 and 60 nm) in vitro using primary rat brain microvessel endothelial cells (rBMEC). The Au-NPs were characterized by transmission electron microscopy (TEM), dynamic light scattering (DLS) and laser Doppler velocimetry (LDV). The accumulation of Au-NPs was determined spectrophotometrically. The rBMEC cytotoxicity of Au-NPs was evaluated by cell proliferation assay (XTT) (concentration range 0.24-15.63 µg/cm², for 24 h). The time-dependent changes (0, 2, 4 and 8 h) of several proinflammatory mediators (IL-1ß, IL-2, TNFα and PGE2) were evaluated by ELISA. The smaller Au-NPs (3-7 nm) showed higher rBMEC accumulation compared to larger Au-NPs (10-60 nm), while only moderate decreased cell viability was observed with small Au-NPs (3 nm) at high concentrations (≥ 7.8 µg/cm²). Even though slight changes in cell viability were observed with small Au-NPs, the basal levels of the various proinflammatory mediators remained unchanged with all treatments except LPS (positive control). rBMEC morphology appeared unaffected 24 h after exposure to Au-NPs with only mild changes in fluorescein permeability indicating BBB integrity was unaltered. Together, these data suggest the responses of the cerebral microvasculature to Au-NPs have a significant relationship with the Au-NPs unique size-dependent physiochemical properties.


Assuntos
Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Ouro/farmacologia , Mediadores da Inflamação/metabolismo , Nanopartículas Metálicas/química , Animais , Permeabilidade da Membrana Celular/efeitos dos fármacos , Proliferação de Células , Forma Celular , Sobrevivência Celular , Células Cultivadas , Citocinas/metabolismo , Dinoprostona/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Fluoresceína , Ouro/farmacocinética , Fluxometria por Laser-Doppler , Luz , Microscopia Eletrônica de Transmissão , Microvasos/citologia , Microvasos/efeitos dos fármacos , Tamanho da Partícula , Ratos , Espalhamento de Radiação
7.
Toxicol Sci ; 118(1): 160-70, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20713472

RESUMO

The current report examines the interactions of silver nanoparticles (Ag-NPs) with the cerebral microvasculature to identify the involvement of proinflammatory mediators that can increase blood-brain barrier (BBB) permeability. Primary rat brain microvessel endothelial cells (rBMEC) were isolated from adult Sprague-Dawley rats for an in vitro BBB model. The Ag-NPs were characterized by transmission electron microscopy (TEM), dynamic light scattering, and laser Doppler velocimetry. The cellular accumulation, cytotoxicity (6.25-50 µg/cm(3)) and potential proinflammatory mediators (interleukin [IL]-1ß, IL-2, tumor necrosis factor [TNF] α, and prostaglandin E(2) [PGE(2)]) of Ag-NPs (25, 40, or 80 nm) were determined spectrophotometrically, cell proliferation assay (2,3-bis[2-methoxy-4-nitro-5-sulfophenyl]-2H-tetrazolium-5-carboxanilide) and ELISA. The results show Ag-NPs-induced cytotoxic responses at lower concentrations for 25 and 40 nm when compared with 80-nm Ag-NPs. The proinflammatory responses in this study demonstrate both Ag-NPs size and time-dependent profiles, with IL-1B preceding both TNF and PGE(2) for 25 nm. However, larger Ag-NPs (40 and 80 nm) induced significant TNF responses at 4 and 8 h, with no observable PGE(2) response. The increased fluorescein transport observed in this study clearly indicates size-dependent increases in BBB permeability correlated with the severity of immunotoxicity. Together, these data clearly demonstrate that larger Ag-NPs (80 nm) had significantly less effect on rBMEC, whereas the smaller particles induced significant effects on all the end points at lower concentrations and/or shorter times. Further, this study suggests that Ag-NPs may interact with the cerebral microvasculature producing a proinflammatory cascade, if left unchecked; these events may further induce brain inflammation and neurotoxicity.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/irrigação sanguínea , Endotélio Vascular/efeitos dos fármacos , Inflamação/patologia , Nanopartículas Metálicas/toxicidade , Microvasos/efeitos dos fármacos , Prata/toxicidade , Animais , Biomarcadores/metabolismo , Velocidade do Fluxo Sanguíneo , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Permeabilidade Capilar/efeitos dos fármacos , Endotélio Vascular/metabolismo , Mediadores da Inflamação/metabolismo , Nanopartículas Metálicas/análise , Nanopartículas Metálicas/ultraestrutura , Microscopia Eletrônica de Transmissão , Microvasos/metabolismo , Tamanho da Partícula , Ratos , Ratos Sprague-Dawley , Espalhamento de Radiação
8.
Neurotoxicology ; 30(6): 926-33, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19781568

RESUMO

Nanoparticles have received a great deal of attention for producing new engineering applications due to their novel physicochemical characteristics. However, the broad application of nanomaterials has also produced concern for nanoparticle toxicity due to increased exposure from large-scale industry production. This study was conducted to investigate the potential neurotoxicity of manganese (Mn), silver (Ag), and copper (Cu) nanoparticles using the dopaminergic neuronal cell line, PC12. Selective genes associated with the dopaminergic system were investigated for expression changes and their correlation with dopamine depletion. PC12 cells were treated with 10 microg/ml Mn-40 nm, Ag-15 nm, or Cu-90 nm nanoparticles for 24 h. Cu-90 nanoparticles induced dopamine depletion in PC12 cells, which is similar to the effect induced by Mn-40 shown in a previous study. The expression of 11 genes associated with the dopaminergic system was examined using real-time RT-PCR. The expression of Txnrd1 was up-regulated after the Cu-90 treatment and the expression of Gpx1 was down-regulated after Ag-15 or Cu-90 treatment. These alterations are consistent with the oxidative stress induced by metal nanoparticles. Mn-40 induced a down-regulation of the expression of Th; Cu-90 induced an up-regulation of the expression of Maoa. This indicates that besides the oxidation mechanism, enzymatic alterations may also play important roles in the induced dopamine depletion. Mn-40 also induced a down-regulation of the expression of Park2; while the expression of Snca was up-regulated after Mn-40 or Cu-90 treatment. These data suggest that Mn and Cu nanoparticles-induced dopaminergic neurotoxicity may share some common mechanisms associated with neurodegeneration.


Assuntos
Cobre/farmacologia , Dopamina/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Manganês/farmacologia , Nanopartículas Metálicas , Prata/farmacologia , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Animais , Proteínas de Ligação ao Cálcio/genética , Catecol O-Metiltransferase/genética , Dopamina/metabolismo , Glutationa Peroxidase/genética , Glutationa Sintase/genética , Ácido Homovanílico/metabolismo , Fluxometria por Laser-Doppler/métodos , Proteínas de Membrana/genética , Nanopartículas Metálicas/ultraestrutura , Microscopia Eletrônica de Transmissão/métodos , Monoaminoxidase/genética , Proteínas Musculares/genética , Proteínas do Tecido Nervoso/genética , Células PC12/efeitos dos fármacos , Células PC12/metabolismo , Ratos , Receptores Acoplados a Proteínas G/genética , Tiorredoxina Redutase 1/genética , Tirosina 3-Mono-Oxigenase/genética , Ubiquitina-Proteína Ligases/genética , Proteínas Vesiculares de Transporte de Monoamina/genética , Glutationa Peroxidase GPX1
9.
Toxicol Sci ; 111(2): 355-61, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19638431

RESUMO

It has been reported that suppression of N-methyl-D-aspartate (NMDA) receptor function by ketamine may trigger apoptosis of neurons when given repeatedly during the brain growth spurt period. Because microPET scans can provide in vivo molecular imaging at sufficient resolution, it has been proposed as a minimally invasive method for detecting apoptosis using the tracer (18)F-labeled annexin V. In this study, the effect of ketamine on the metabolism and integrity of the rat brain were evaluated by investigating the uptake and retention of (18)F-fluorodeoxyglucose (FDG) and (18)F-annexin V using microPET imaging. On postnatal day (PND) 7, rat pups in the experimental group were exposed to six injections of ketamine (20 mg/kg at 2-h intervals) and control rat pups received six injections of saline. On PND 35, 37 MBq (1 mCi) of (18)F-FDG or (18)F-annexin V was injected into the tail vein of treated and control rats, and static microPET images were obtained over 1 (FDG) and 2 h (annexin V) following the injection. No significant difference was found in (18)F-FDG uptake in the regions of interest (ROIs) in the brains of ketamine-treated rats compared with saline-treated controls. The uptake of (18)F-annexin V, however, was significantly increased in the ROI of ketamine-treated rats. Additionally, the duration of annexin V tracer washout was prolonged in the ketamine-treated animals. These results demonstrate that microPET imaging is capable of distinguishing differences in retention of (18)F-annexin V in different brain regions and suggests that this approach may provide a minimally invasive biomarker of neuronal apoptosis in rats.


Assuntos
Anestésicos Dissociativos/toxicidade , Anexina A5/metabolismo , Apoptose/efeitos dos fármacos , Biomarcadores/metabolismo , Radioisótopos de Flúor/metabolismo , Ketamina/toxicidade , Neurônios/efeitos dos fármacos , Tomografia por Emissão de Pósitrons/métodos , Animais , Western Blotting , Feminino , Masculino , Neurônios/citologia , Ratos , Ratos Sprague-Dawley
10.
Neurochem Int ; 52(6): 1037-43, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18069091

RESUMO

Parkinson's disease (PD) is a common neurodegenerative disease characterized by progressive loss of midbrain dopaminergic neurons with unknown etiology. MPP+ (1-methyl-4-phenylpyridinium ion) is the active metabolite of the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), which induces Parkinson's-like symptoms in humans and animals. MPTP/MPP+ produces selective dopaminergic neuronal degeneration, therefore, these agents are commonly used to study the pathogenesis of PD. However, the mechanisms of their toxicity have not been fully elucidated. Recently, we reported in a microarray study using a midbrain-derived dopaminergic neuronal cell line, MN9D, that MPP+ induced significant changes in a number of genes known to be associated with the dopaminergic system. In this study, we investigated the expression time courses of six genes using real-time RT-PCR, and compared them with the progressive dopaminergic depletion caused by MPP+. Our data showed that dopamine content was significantly decreased after 0.5h of MPP+ (200 microM) exposure and was completely depleted after 40 h. The expression of Gpr37, which is closely related to the pathogenesis of autosomal recessive juvenile Parkinsonism, was up-regulated after 0.5h, and stayed up-regulated up to 48 h. Txnip, which is critical to the adjustment of cellular redox status, was down-regulated after 1h and stayed down-regulated up to 48 h. Ldh1 and Cdo1, which are also involved in oxidative stress, were down-regulated after 16 h and stayed down-regulated up to 48 h. Two pro-apoptotic genes, Egln3 and Bnip3, were down-regulated after 2 and 4h, and stayed down-regulated up to 48 h. These findings suggested that the time course of expression for multiple genes correlated with the dopaminergic depletion; and MPP+-induced neurotoxicity in MN9D cells could be used as a model to further explore the roles of these and other genes in the pathogenesis and possible treatment of PD.


Assuntos
1-Metil-4-fenilpiridínio/toxicidade , Dopamina/deficiência , Regulação da Expressão Gênica/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Transtornos Parkinsonianos/genética , Animais , Proteínas de Transporte/genética , Linhagem Celular , Proteínas de Ligação a DNA/genética , Progressão da Doença , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Regulação da Expressão Gênica/genética , Herbicidas/toxicidade , Prolina Dioxigenases do Fator Induzível por Hipóxia , Proteínas Imediatamente Precoces/genética , Isoenzimas/genética , L-Lactato Desidrogenase/genética , Proteínas de Membrana/genética , Camundongos , Proteínas Mitocondriais/genética , Neurônios/metabolismo , Transtornos Parkinsonianos/metabolismo , Transtornos Parkinsonianos/fisiopatologia , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G/genética , Substância Negra/efeitos dos fármacos , Substância Negra/metabolismo , Substância Negra/fisiopatologia , Tiorredoxinas/genética , Fatores de Tempo
11.
Neurotoxicology ; 24(3): 379-90, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12782103

RESUMO

Plasma levels of parent compounds and metabolites were determined in adult rhesus monkeys after doses of either 5mg/kg d-fenfluramine (FEN) or 10mg/kg d-3, 4-methylenedioxymethamphetamine (MDMA) i.m. twice daily for four consecutive days. These treatment regimens have been previously shown to produce long-term serotonin (5-HT) depletions. Peak plasma levels of 2.0+/-0.4 microM FEN were reached within 40min after the first dose of FEN, and then declined rapidly, while peak plasma levels (0.4+/-0.1 microM) of the metabolite norfenfluramine (NFEN) were not reached until 6h after dosing. After the seventh (next to last) dose of FEN, peak plasma levels of FEN were 35% greater than after the first dose while peak NFEN-levels were 500% greater. The t(1/2) for FEN was 2.6+/-0.3h after the first dose and 3.2+/-0.2h after the seventh. The estimated t(1/2) for NFEN was more than 37.6+/-20.5h. Peak plasma levels of 9.5+/-2.5 microM MDMA were reached within 20min after the first dose of MDMA, and then declined rapidly, while peak plasma levels (0.9+/-0.2 microM) of the metabolite 3,4-methylenedioxyamphetamine (MDA) were not reached until 3-6h after dosing. After the seventh (next to last) dose of MDMA, peak plasma levels of MDMA were 30% greater than the first dose while peak MDA levels were elevated over 200%. The t(1/2) for MDMA was 2.8+/-0.4h after the first and 3.9+/-1.1h after the seventh dose. The estimated t(1/2) for MDA was about 8.3+/-1.0h. Variability in plasma levels of MDMA and MDA between subjects was much greater than that for FEN and NFEN. This variability in MDMA and MDA exposure levels may have lead to variability in the subsequent disruption of some behaviors seen in these same subjects. There were 80% reductions in the plasma membrane-associated 5-HT transporters 6 months after either the FEN or MDMA dosing regimen indicating that both treatments produced long-term serotonergic effects.


Assuntos
Fenfluramina/administração & dosagem , Fenfluramina/metabolismo , Proteínas de Membrana Transportadoras , N-Metil-3,4-Metilenodioxianfetamina/administração & dosagem , N-Metil-3,4-Metilenodioxianfetamina/metabolismo , Proteínas do Tecido Nervoso , Serotonina/metabolismo , Animais , Temperatura Corporal/efeitos dos fármacos , Temperatura Corporal/fisiologia , Proteínas de Transporte/sangue , Proteínas de Transporte/metabolismo , Fenfluramina/sangue , Macaca mulatta , Masculino , Glicoproteínas de Membrana/sangue , Glicoproteínas de Membrana/metabolismo , N-Metil-3,4-Metilenodioxianfetamina/sangue , Serotonina/sangue , Proteínas da Membrana Plasmática de Transporte de Serotonina , Tempo
12.
Ann N Y Acad Sci ; 965: 204-13, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12105096

RESUMO

Methamphetamine (METH) is a widely abused psychomotor stimulant known to cause dopaminergic neurotoxicity in rodents, nonhuman primates, and humans. METH administration selectively damages the dopaminergic nerve terminals, which is hypothesized to be due to release of dopamine from synaptic vesicles within the terminals. This process is believed to be mediated by the production of free radicals. The current study evaluates METH-induced dopaminergic toxicity in pheochromocytoma 12 (PC12) cells cultured in the presence or absence of nerve growth factor (NGF). Dopaminergic changes and the formation of 3-nitrotyrosine (3-NT), a marker for peroxynitrite production, were studied in PC12 cell cultures grown in the presence or absence of NGF after different doses of METH (100-1,000 microM). METH exposure did not cause significant alterations in cell viability and did not produce significant dopaminergic changes or 3-NT production in PC12 cells grown in NGF-negative media after 24 hours. However, cell viability of PC12 cells grown in NGF-positive media was decreased by 45%, and significant dose-dependent dopaminergic alteration and 3-NT production were observed 24 hours after exposure to METH. The current study supports the hypothesis that METH acts at the dopaminergic nerve terminals and produces dopaminergic damage by the production of free radical peroxynitrite.


Assuntos
Dopamina/metabolismo , Metanfetamina/toxicidade , Fator de Crescimento Neural/farmacologia , Neurotoxinas , Ácido Peroxinitroso/metabolismo , Tirosina/análogos & derivados , Animais , Diferenciação Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Cinética , Células PC12 , Feocromocitoma , Ratos , Fatores de Tempo , Tirosina/metabolismo
13.
Am J Primatol ; 2(4): 385-399, 1982.
Artigo em Inglês | MEDLINE | ID: mdl-32188175

RESUMO

The placental transfer of conjugated and nonconjugated estrogens was compared in the pregnant rhesus monkey. Placement of catheters in the maternal and fetal circulation allowed for the sampling of blood after the administration of radiolabeled naturally occurring and synthetic estrogens to mother or fetus. In all cases, nonconjugated-estrogen placental transfer was greater than conjugated-estrogen transfer. Comparison of the conjugated estrogens suggested that diethylstilbestrol-monoglucuronide (DESG) was transferred less efficiently than the estrone-sulfate (E, S). High-performance liquid chromatog-raphy (HPLC) of selected plasma samples revealed that 50-90% of the E, S observed in the maternal circulation was cleaved after fetal administration. In contrast, HPLC of maternal or fetal plasma samples after DESG administration revealed only intact DESG. These results emphasize differences in the placental transfer of the synthetic and naturally occurring estrogen hormones.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...