Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Proc Natl Acad Sci U S A ; 108(16): 6638-43, 2011 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-21464304

RESUMO

The factors that determine symptom penetrance in inherited disease are poorly understood. Increasingly, magnetic resonance diffusion tensor imaging (DTI) and PET are used to separate alterations in brain structure and function that are linked to disease symptomatology from those linked to gene carrier status. One example is DYT1 dystonia, a dominantly inherited movement disorder characterized by sustained muscle contractions, postures, and/or involuntary movements. This form of dystonia is caused by a 3-bp deletion (i.e., ΔE) in the TOR1A gene that encodes torsinA. Carriers of the DYT1 dystonia mutation, even if clinically nonpenetrant, exhibit abnormalities in cerebellothalamocortical (CbTC) motor pathways. However, observations in human gene carriers may be confounded by variability in genetic background and age. To address this problem, we implemented a unique multimodal imaging strategy in a congenic line of DYT1 mutant mice that contain the ΔE mutation in the endogenous mouse torsinA allele (i.e., DYT1 knock-in). Heterozygous knock-in mice and littermate controls underwent microPET followed by ex vivo high-field DTI and tractographic analysis. Mutant mice, which do not display abnormal movements, exhibited significant CbTC tract changes as well as abnormalities in brainstem regions linking cerebellar and basal ganglia motor circuits highly similar to those identified in human nonmanifesting gene carriers. Moreover, metabolic activity in the sensorimotor cortex of these animals was closely correlated with individual measures of CbTC pathway integrity. These findings further link a selective brain circuit abnormality to gene carrier status and demonstrate that DYT1 mutant torsinA has similar effects in mice and humans.


Assuntos
Encéfalo , Distonia , Vias Eferentes , Doenças Genéticas Inatas , Chaperonas Moleculares/metabolismo , Transtornos dos Movimentos , Alelos , Animais , Sequência de Bases , Encéfalo/anormalidades , Encéfalo/metabolismo , Distonia/genética , Distonia/metabolismo , Distonia/patologia , Vias Eferentes/anormalidades , Vias Eferentes/metabolismo , Técnicas de Introdução de Genes , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/metabolismo , Doenças Genéticas Inatas/patologia , Humanos , Camundongos , Camundongos Transgênicos , Chaperonas Moleculares/genética , Transtornos dos Movimentos/genética , Transtornos dos Movimentos/metabolismo , Transtornos dos Movimentos/patologia , Deleção de Sequência
2.
Mol Imaging ; 12(7): 1-15, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23920252

RESUMO

Preclinical molecular imaging is a rapidly growing field, where new imaging systems, methods, and biological findings are constantly being developed or discovered. Imaging systems and the associated software usually have multiple options for generating data, which is often overlooked but is essential when reporting the methods used to create and analyze data. Similarly, the ways in which animals are housed, handled, and treated to create physiologically based data must be well described in order that the findings be relevant, useful, and reproducible. There are frequently new developments for metabolic imaging methods. Thus, specific reporting requirements are difficult to establish; however, it remains essential to adequately report how the data have been collected, processed, and analyzed. To assist with future manuscript submissions, this article aims to provide guidelines of what details to report for several of the most common imaging modalities. Examples are provided in an attempt to give comprehensive, succinct descriptions of the essential items to report about the experimental process.


Assuntos
Diagnóstico por Imagem , Imagem Molecular , Editoração , Projetos de Pesquisa , Animais , Processamento de Imagem Assistida por Computador , Imageamento por Ressonância Magnética , Camundongos , Camundongos Nus , Imagem Molecular/métodos , Revisão da Pesquisa por Pares , Cintilografia , Ratos , Tomografia Computadorizada por Raios X , Ultrassonografia
3.
Neuroimage ; 59(3): 2689-99, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21767654

RESUMO

The parametric ntPET model (p-ntPET) estimates the kinetics of neurotransmitter release from dynamic PET data with receptor-ligand radiotracers. Here we introduce a linearization (lp-ntPET) that is computationally efficient and can be applied to single scan data. lp-ntPET employs a non-invasive reference region input function and extends the LSRRM of Alpert et al. (2003) using basis functions to characterize the time course of neurotransmitter activation. In simulation studies, the temporal precision of neurotransmitter profiles estimated by lp-ntPET was similar to that of p-ntPET (standard deviation ~3 min for responses early in the scan) while computation time was reduced by several orders of magnitude. Violations of model assumptions such as activation-induced changes in regional blood flow or specific binding in the reference tissue have negligible effects on lp-ntPET performance. Application of the lp-ntPET method is demonstrated on [11C]raclopride data acquired in rats receiving methamphetamine, which yielded estimated response functions that were in good agreement with simultaneous microdialysis measurements of extracellular dopamine concentration. These results demonstrate that lp-ntPET is a computationally efficient, linear variant of ntPET that can be applied to PET data from single or multiple scan designs to estimate the time course of neurotransmitter activation.


Assuntos
Processamento de Imagem Assistida por Computador/métodos , Neurotransmissores/fisiologia , Tomografia por Emissão de Pósitrons/métodos , Tomografia por Emissão de Pósitrons/estatística & dados numéricos , Algoritmos , Animais , Simulação por Computador , Dopamina/metabolismo , Antagonistas de Dopamina , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Modelos Lineares , Microdiálise , Modelos Neurológicos , Modelos Estatísticos , Racloprida , Compostos Radiofarmacêuticos , Ratos
4.
J Neurosci ; 29(19): 6176-85, 2009 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-19439595

RESUMO

Positron emission tomography studies in drug-addicted patients have shown that exposure to drug-related cues increases striatal dopamine, which displaces binding of the D(2) ligand, [(11)C]-raclopride. However, it is not known if animals will also show cue-induced displacement of [(11)C]-raclopride binding. In this study, we use [(11)C]-raclopride imaging in awake rodents to capture cue-induced changes in dopamine release associated with the conditioned place preference model of drug craving. Ten animals were conditioned to receive cocaine in a contextually distinct environment from where they received saline. Following conditioning, each animal was tested for preference and then received two separate [(11)C]-raclopride scans. For each scan, animals were confined to the cocaine and/or the saline-paired environment for the first 25 min of uptake, after which they were anesthetized and scanned. [(11)C]-raclopride uptake in the saline-paired environment served as a within-animal control for uptake in the cocaine-paired environment. Cocaine produced a significant place preference (p = 0.004) and exposure to the cocaine-paired environment decreased [(11)C]-raclopride binding relative to the saline-paired environment in both the dorsal (20%; p < 0.002) and ventral striatum (22%; p < 0.05). The change in [(11)C]-raclopride binding correlated with preference in the ventral striatum (R(2) = -0.87; p = 0.003). In this region, animals who showed little or no preference exhibited little or no change in [(11)C]-raclopride binding in the cocaine-paired environment. This noninvasive procedure of monitoring neurochemical events in freely moving, behaving animals advances preclinical molecular imaging by interrogating the degree to which animal models reflect the human condition on multiple dimensions, both biological and behavioral.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/fisiopatologia , Cocaína , Corpo Estriado/fisiopatologia , Sinais (Psicologia) , Dopamina/metabolismo , Animais , Radioisótopos de Carbono , Transtornos Relacionados ao Uso de Cocaína/diagnóstico por imagem , Condicionamento Psicológico , Corpo Estriado/diagnóstico por imagem , Antagonistas de Dopamina/farmacologia , Antagonistas dos Receptores de Dopamina D2 , Masculino , Atividade Motora , Tomografia por Emissão de Pósitrons , Racloprida/metabolismo , Racloprida/farmacologia , Ratos , Ratos Sprague-Dawley , Percepção Espacial/efeitos dos fármacos
5.
Eur J Neurosci ; 30(8): 1565-75, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19821842

RESUMO

Most studies of the effect of cocaine on brain activity in laboratory animals are preformed under anesthesia, which could potentially affect the physiological responses to cocaine. Here we assessed the effects of two commonly used anesthetics [alpha-chloralose (alpha-CHLOR) and isofluorane (ISO)] on the effects of acute cocaine (1 mg/kg i.v.) on cerebral blood flow (CBF), cerebral blood volume (CBV), and tissue hemoglobin oxygenation (S(t)O(2)) using optical techniques and cocaine's pharmacokinetics (PK) and binding in the rat brain using (PET) and [(11)C]cocaine. We showed that acute cocaine at a dose abused by cocaine abusers decreased CBF, CBV and S(t)O(2) in rats anesthetized with ISO, whereas it increased these parameters in rats anesthetized with alpha-CHLOR. Importantly, in ISO-anesthetized animals cocaine-induced changes in CBF and S(t)O(2) were coupled, whereas for alpha-CHLOR these measures were uncoupled. Moreover, the clearance of [(11)C]cocaine from the brain was faster for ISO (peak half-clearance 15.8 +/- 2.8 min) than for alpha-CHLOR (27.5 +/- 0.6 min), and the ratio of specific to non-specific binding of [(11)C]cocaine in the brain was higher for ISO- (3.37 +/- 0.32) than for alpha-CHLOR-anesthetized rats (2.24 +/- 0.4). For both anesthetics, cocaine-induced changes in CBF followed the fast uptake of [(11)C]cocaine in the brain (peaking at approximately 2.5-4 min), but only for ISO did the duration of the CBV and S(t)O(2) changes correspond to the rate of [(11)C]cocaine's clearance from the brain. These results demonstrate that anesthetics influence cocaine's hemodynamic and metabolic changes in the brain, and its binding and PK, which highlights the need to better understand the interactions between anesthetics and pharmacological challenges in brain functional imaging studies.


Assuntos
Anestésicos/farmacologia , Encéfalo/efeitos dos fármacos , Cloralose/farmacologia , Cocaína/farmacocinética , Isoflurano/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Volume Sanguíneo/efeitos dos fármacos , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Mapeamento Encefálico/métodos , Isótopos de Carbono/farmacocinética , Circulação Cerebrovascular/efeitos dos fármacos , Eletrocardiografia/métodos , Feminino , Frequência Cardíaca/efeitos dos fármacos , Oxiemoglobinas/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
6.
Synapse ; 63(2): 87-94, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19016239

RESUMO

Preventing relapse poses a significant challenge to the successful management of methamphetamine (METH) dependence. Although no effective medication currently exists for its treatment, racemic gamma vinyl-GABA (R,S-GVG, vigabatrin) shows enormous potential as it blocks both the neurochemical and behavioral effects of a variety of drugs, including METH, heroin, morphine, ethanol, nicotine, and cocaine. Using the reinstatement of a conditioned place preference (CPP) as an animal model of relapse, the present study specifically investigated the ability of an acute dose of R,S-GVG to block METH-triggered reinstatement of a METH-induced CPP. Animals acquired a METH CPP following a 20-day-period of conditioning, in which they received 10 pairings of alternating METH and saline injections. During conditioning, rats were assigned to one of four METH dosage groups: 1.0, 2.5, 5.0, or 10.0 mg/kg (i.p., n = 8/group). Animals in all dosage groups demonstrated a robust and consistent CPP. This CPP was subsequently extinguished in each dosage group with repeated saline administration. Upon extinction, all groups reinstated following an acute METH challenge. On the following day, an acute dose of R,S-GVG (300 mg/kg, i.p.) was administered 2.5 h prior to an identical METH challenge. R,S-GVG blocked METH-triggered reinstatement in all four groups. Given that drug re-exposure may potentiate relapse to drug-seeking behavior, the ability of R,S-GVG to block METH-triggered reinstatement offers further support for its use in the successful management of METH dependence.


Assuntos
Transtornos Relacionados ao Uso de Anfetaminas/prevenção & controle , Estimulantes do Sistema Nervoso Central/efeitos adversos , GABAérgicos/farmacologia , Metanfetamina/efeitos adversos , Vigabatrina/farmacologia , Animais , Condicionamento Clássico/efeitos dos fármacos , Extinção Psicológica/efeitos dos fármacos , Isomerismo , Masculino , Ratos , Ratos Sprague-Dawley , Recidiva
7.
Mol Imaging Biol ; 10(2): 67-73, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18176804

RESUMO

A recently introduced mathematical method for extracting temporal characteristics of neurotransmitter release from dynamic positron emission tomography (PET) data was tested. The method was developed with the hope that by uncovering temporal information about neurotransmitter (nt) dynamics in PET data, researchers could shed new light on mechanisms of psychiatric diseases such as drug abuse and its treatment. In this study, we apply our model-based method, "ntPET", to (11)C-raclopride PET scans of rats in which the dopaminergic response to a microinfusion of methamphetamine in one striatum was assayed simultaneously by microdialysis and PET. Uptake of (11)C-raclopride into the untreated contralateral striatum was used as an input to the ntPET model. Direct comparisons of the model-based ntPET analysis and the microdialysis measurements confirmed that ntPET produced dopamine curves that were very similar in timing (takeoff and peak times) to the microdialysis curves. Variances in takeoff and peak times were comparable for the two methods. Neither method detected a false dopamine response to drug in a control animal. The high degree of correspondence between ntPET estimates and microdialysis measurements lends strong support to the idea that temporal information regarding dopamine release exists in dynamic (11)C-raclopride PET data and that it can be estimated reliably via ntPET. The method is entirely translatable to human PET imaging.


Assuntos
Dopamina/metabolismo , Metanfetamina/farmacologia , Microdiálise/métodos , Neurotransmissores/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Animais , Bombas de Infusão , Masculino , Metanfetamina/administração & dosagem , Racloprida/metabolismo , Ratos , Ratos Sprague-Dawley , Padrões de Referência
8.
Synapse ; 62(11): 870-2, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18720383

RESUMO

Given the growing obesity epidemic, pressure to develop an effective pharmacologic treatment is mounting. Following the completion of a randomized, double-blind, placebo controlled trial as well as two small open label trials, gamma vinyl-GABA (GVG) has been shown to be safe and effective for treating cocaine and/or methamphetamine dependence. In an extension of these findings, the present study examined whether GVG could produce weight loss in adolescent as well as genetically obese animals. Specifically, adolescent Sprague Dawley and adolescent and adult Zucker fatty rats received GVG at various doses (75-300 mg/kg, i.p., racemic) for treatment periods lasting no longer than 14 consecutive days. GVG produced significant weight loss in a dose dependent fashion in all groups. These effects were marked, as average decreases of 12-20% of original body weight were observed. These findings suggest that GVG may be useful as a treatment for obesity. Further, that these results occurred in genetically obese animals offers the possibility that GVG may even help manage severe obesity resulting from binge-eating, a disorder involving food consumption in a pattern similar to the compulsive drug-seeking behavior observed in cocaine and methamphetamine dependent subjects.


Assuntos
Vigabatrina/administração & dosagem , Vigabatrina/farmacologia , Redução de Peso/efeitos dos fármacos , Fatores Etários , Animais , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Cronoterapia/métodos , Injeções Intraperitoneais , Masculino , Obesidade/tratamento farmacológico , Obesidade/fisiopatologia , Ratos , Ratos Sprague-Dawley , Ratos Zucker , Vigabatrina/fisiologia , Redução de Peso/fisiologia
9.
J Nucl Med ; 48(2): 277-87, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17268026

RESUMO

UNLABELLED: Small-animal PET provides the opportunity to image brain activation during behavioral tasks in animal models of human conditions. The present studies aimed to simplify behavioral imaging procedures without a loss of quantitation by using an intraperitoneal route of administration (no cannulation, no anesthesia) and using a standardized uptake value (SUV) to reduce scan duration. METHODS: Sixteen animals with carotid artery cannulations were studied with 18F-FDG small-animal PET accompanied by serial arterial blood sampling. Ten of these animals were anesthetized and were inside the tomograph during 18F-FDG uptake, whereas 6 animals were awake in their home cages and scanned after 60 min of uptake. Of the 10 anesthetized animals, 6 received intraperitoneal 18F-FDG, whereas 4 received intravenous 18F-FDG, and all 6 awake animals received intraperitoneal 18F-FDG. Intravenously injected animals were positioned far enough inside the tomograph to obtain region-of-interest-based measures from the heart and the brain. In all animals, a full arterial input function and plasma glucose levels were obtained. To establish the optimal time during 18F-FDG uptake for blood sampling when using an SUV, a Patlak kinetic model was used to derive absolute rates of glucose metabolism and compared with SUVs calculated using different plasma points from the arterial input function. RESULTS: A single plasma point taken at 60 min after injection for intraperitoneal injections or 45 min after injection for intravenous injections provides a sensitive index of glucose metabolic rate with the highest correlation with data obtained from a fully quantitative input function. CONCLUSION: These studies support an experimental protocol in which animals can receive the 18F-FDG tracer injection intraperitoneally, away from the small-animal tomograph and with minimal impact on behavior. Further, animals can occupy the tomograph bed for a 10- to 30-min scan with a consequent increase in animal throughput.


Assuntos
Comportamento Animal/fisiologia , Encéfalo/diagnóstico por imagem , Fluordesoxiglucose F18/administração & dosagem , Compostos Radiofarmacêuticos/administração & dosagem , Algoritmos , Animais , Glicemia/metabolismo , Artérias Carótidas , Cateterismo , Interpretação Estatística de Dados , Processamento de Imagem Assistida por Computador , Injeções Intraperitoneais , Injeções Intravenosas , Masculino , Tomografia por Emissão de Pósitrons , Ratos , Ratos Sprague-Dawley , Receptores de Glutamato Metabotrópico/metabolismo
10.
J Nucl Med ; 48(10): 1724-32, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17873134

RESUMO

UNLABELLED: The methamphetamine molecule has a chiral center and exists as 2 enantiomers, d-methamphetamine (the more active enantiomer) and l-methamphetamine (the less active enantiomer). d-Methamphetamine is associated with more intense stimulant effects and higher abuse liability. The objective of this study was to measure the pharmacokinetics of d-methamphetamine for comparison with both l-methamphetamine and (-)-cocaine in the baboon brain and peripheral organs and to assess the saturability and pharmacologic specificity of binding. METHODS: d- and l-methamphetamine and (-)-cocaine were labeled with (11)C via alkylation of the norprecursors with (11)C-methyl iodide using literature methods. Six different baboons were studied in 11 PET sessions at which 2 radiotracer injections were administered 2-3 h apart to determine the distribution and kinetics of (11)C-d-methamphetamine in brain and peripheral organs. Saturability and pharmacologic specificity were assessed using pretreatment with d-methamphetamine, methylphenidate, and tetrabenazine. (11)C-d-Methamphetamine pharmacokinetics were compared with (11)C-l-methamphetamine and (11)C-(-)-cocaine in both brain and peripheral organs in the same animal. RESULTS: (11)C-d- and l-methamphetamine both showed high uptake and widespread distribution in the brain. Pharmacokinetics did not differ between enantiomers, and the cerebellum peaked earlier and cleared more quickly than the striatum for both. (11)C-d-Methamphetamine distribution volume ratio was not substantially affected by pretreatment with methamphetamine, methylphenidate, or tetrabenazine. Both enantiomers showed rapid, high uptake and clearance in the heart and lungs and slower uptake and clearance in the liver and kidneys. A comparison of (11)C-d-methamphetamine and (11)C-(-)-cocaine showed that (11)C-d-methamphetamine peaked later in the brain than did (11)C-(-)-cocaine and cleared more slowly. The 2 drugs showed similar behavior in all peripheral organs examined except the kidneys and pancreas, which showed higher uptake for (11)C-d-methamphetamine. CONCLUSION: Brain pharmacokinetics did not differ between d-and l-methamphetamine and thus cannot account for the more intense stimulant effects of d-methamphetamine. Lack of pharmacologic blockade by methamphetamine indicates that the PET image represents nonspecific binding, though the fact that methamphetamine is both a transporter substrate and an inhibitor may also play a role. A comparison of (11)C-d-methamphetamine and (11)C-(-)-cocaine in the same animal showed that the slower clearance of methamphetamine is likely to contribute to its previously reported longer-lasting stimulant effects relative to those of (-)-cocaine. High kidney uptake of d-methamphetamine or its labeled metabolites may account for the reported renal toxicity of d-methamphetamine in humans.


Assuntos
Encéfalo/metabolismo , Cocaína/farmacocinética , Metanfetamina/farmacocinética , Papio/metabolismo , Animais , Encéfalo/diagnóstico por imagem , Taxa de Depuração Metabólica , Tomografia por Emissão de Pósitrons/métodos , Distribuição Tecidual
11.
Brain Res ; 1144: 209-18, 2007 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-17346680

RESUMO

To test the hypothesis that functional metabolic deficits observed following surgical brain injury are associated with changes in cognitive performance in rodents, we performed serial imaging studies in parallel with behavioral measures in control animals and in animals with surgical implants. Memory function was assessed using the novel object recognition (NOR) test, administered 3 days prior to and 3, 7, 14 and 56 days after surgery. At each time point, general locomotion was also measured. Metabolic imaging with 18F-fluorodeoxyglucose ([18F]FDG) occurred 28 and 58 days after surgery. Animals with surgical implants performed significantly worse on tests of object recognition, while general locomotion was unaffected by the implant. There was a significant decrease in glucose uptake after surgery in most of the hemisphere ipsilateral to the implant relative to the contralateral hemisphere. At both time points, the most significant metabolic deficits occurred in the primary motor cortex (-25%; p<0.001), sensory cortex (-15%, p<0.001) and frontal cortex (-12%; p<0.001). Ipsilateral areas further from the site of insertion became progressively worse, including the sensory cortex, dorsal striatum and thalamus. These data was supported by a voxel-based analysis of the PET data, which revealed again a unilateral decrease in [18F]FDG uptake that extended throughout the ipsilateral cortex and persisted for the duration of the 58-day study. Probe implantation in the striatum results in a widespread and long-lasting decline in cortical glucose metabolism together with a persistent, injury-related deficit in the performance of a cognitive (object recognition) task in rats.


Assuntos
Comportamento Animal/fisiologia , Sintomas Comportamentais/etiologia , Encefalopatias Metabólicas/etiologia , Lesões Encefálicas/complicações , Lesões Encefálicas/etiologia , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Neurocirurgia , Animais , Mapeamento Encefálico , Fluordesoxiglucose F18 , Masculino , Atividade Motora/fisiologia , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos/metabolismo , Ratos , Ratos Sprague-Dawley , Reconhecimento Psicológico/fisiologia , Fatores de Tempo
12.
Nucl Med Biol ; 34(7): 833-47, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17921034

RESUMO

Although imaging studies in and of themselves have significant contributions to the study of human behavior, imaging in drug abuse has a much broader agenda. Drugs of abuse bind to molecules in specific parts of the brain in order to produce their effects. Positron emission tomography (PET) provides a unique opportunity to track this process, capturing the kinetics with which an abused compound is transported to its site of action. The specific examples discussed here were chosen to illustrate how PET can be used to map the regional distribution and kinetics of compounds that may or may not have abuse liability. We also discussed some morphological and functional changes associated with drug abuse and different stages of recovery following abstinence. PET measurements of functional changes in the brain have also led to the development of several treatment strategies, one of which is discussed in detail here. Information such as this becomes more than a matter of academic interest. Such knowledge can provide the bases for anticipating which compounds may be abused and which may not. It can also be used to identify biological markers or changes in brain function that are associated with progression from drug use to drug abuse and also to stage the recovery process. This new knowledge can guide legislative initiatives on the optimal duration of mandatory treatment stays, promoting long-lasting abstinence and greatly reducing the societal burden of drug abuse. Imaging can also give some insights into potential pharmacotherapeutic targets to manage the reinforcing effects of addictive compounds, as well as into protective strategies to minimize their toxic consequences.


Assuntos
Encéfalo/diagnóstico por imagem , Sistemas de Liberação de Medicamentos/métodos , Técnicas de Sonda Molecular , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos , Transtornos Relacionados ao Uso de Substâncias/diagnóstico por imagem , Humanos , Prognóstico
13.
Psychopharmacology (Berl) ; 186(2): 159-67, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16703400

RESUMO

RATIONALE: Children and adolescents will readily abuse household products that contain solvents such as toluene. It is likely that reinforcing exposures to toluene alter brain glucose metabolism. OBJECTIVE: Using an animal model of drug reinforcement, we sought to identify a metabolic signature of toluene abuse in the adolescent rodent brain. Small animal PET (microPET), in combination with the glucose analog radiotracer, (18)FDG, were used to evaluate the metabolic consequences of inhaled toluene. METHODS: The exposure protocol paralleled our previously established method for assessing the conditioned reinforcing effects of toluene (5,000 ppm) using the conditioned place preference (CPP) paradigm. Animals were scanned at baseline and 2 h after the last exposure. Follow-up (18)FDG scans occurred 1 day, 3 weeks, and 2 months later. RESULTS: After six pairings, 38% of the animals preferred the toluene paired chamber and 25% were averse. The immediate metabolic effect in toluene-exposed animals was a 20% decline in whole brain (18)FDG uptake. Twenty-four hours following the last exposure, the whole brain decline was 40%, and 2 months later, the decline was 30% of pretoluene levels. A region-by-region analysis demonstrated significant additional decreases in the pons, cerebellum, striatum, midbrain, temporal cortex, and hippocampus. Two months after toluene cessation, regions of complete metabolic recovery were the thalamus and cerebellum; however, the temporal cortex did not recover. CONCLUSIONS: Brain uptake of (18)FDG appears to be a useful tool for examining the metabolic impact of toluene abuse, which include a profound decline followed by region-specific recovery after cessation.


Assuntos
Encéfalo/metabolismo , Exposição por Inalação/efeitos adversos , Solventes/toxicidade , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Tolueno/toxicidade , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Mapeamento Encefálico , Modelos Animais de Doenças , Fluordesoxiglucose F18 , Masculino , Ratos , Ratos Sprague-Dawley , Reforço Psicológico , Transtornos Relacionados ao Uso de Substâncias/diagnóstico por imagem , Transtornos Relacionados ao Uso de Substâncias/etiologia , Transtornos Relacionados ao Uso de Substâncias/psicologia , Tomografia Computadorizada de Emissão
14.
J Neurosci Methods ; 155(2): 272-84, 2006 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16519945

RESUMO

Despite the widespread use of chronic brain implants in experimental and clinical settings, the effects of these long-term procedures on brain metabolism and receptor expression remain largely unknown. Under the hypothesis that intracerebral microdialysis transiently alters tissue metabolism, we performed a series of 18FDG microPET scans prior to and following surgical implantation of microdialysis cannulae. Parallel microPET measures using the competitive dopamine (DA) D2 receptor antagonist, 11C-raclopride, provided an assay of DA stability in these same animals. 18FDG scans were performed prior to microdialysis cannulation and again at 2, 12, 24, 48, 120, 168, 360 and 500 h (0.2, 0.5, 1, 2, 5, 7, 15 and 25 days). Separate animals received a sham surgery and the control group had no surgical intervention. For the first 24 h (scans at 2, 12 and 24 h post-surgery) uptake was reduced in both hemispheres. However, by 48 h, contralateral uptake had returned to pre-surgical levels. The striking finding was that from 48 to 500 h, the microdialysis cannulation produced widespread ipsilateral reductions in 18FDG uptake that encompassed the entire hemisphere. Despite the extent and persistence of these reductions, 11C-raclopride binding and ECF DA concentrations remained stable.


Assuntos
Encéfalo/diagnóstico por imagem , Microdiálise/métodos , Tomografia por Emissão de Pósitrons , Animais , Encéfalo/anatomia & histologia , Encéfalo/efeitos dos fármacos , Mapeamento Encefálico , Antagonistas de Dopamina/farmacocinética , Fluordesoxiglucose F18/metabolismo , Masculino , Racloprida/farmacocinética , Compostos Radiofarmacêuticos/farmacocinética , Ratos , Fatores de Tempo
15.
Drug Alcohol Depend ; 85(1): 87-90, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16675162

RESUMO

OBJECTIVES: Toluene is present in many commercial products and is subject to abuse by inhalation. The goal of this study was to extend previous reports indicating that rats will exhibit a positive conditioned place preference to inhaled toluene vapors and to determine the dose-response relationship for inhaled toluene in terms of exposure concentration and number of exposures. For the conditioned place preference experiments rats were exposed to toluene vapors at concentrations of 800, 2000, 3000 or 5000 ppm in one compartment of a three-compartment box. RESULTS: Following six conditioning sessions with toluene, a significant place preference was obtained at 2000 and 3000 ppm, but not at 800 or 5000 ppm. Extending the number of toluene pairings at the 2000 and 3000 ppm concentration to 12 significantly enhanced the place preference compared to that at six pairings. CONCLUSIONS: These experiments extend our previous finding that rats will show a conditioned place preference to inhaled toluene, and indicate that a reinforcing "dose" of toluene depends on both the concentration and number of pairings.


Assuntos
Comportamento de Escolha , Condicionamento Psicológico , Solventes/administração & dosagem , Transtornos Relacionados ao Uso de Substâncias/epidemiologia , Tolueno/administração & dosagem , Administração por Inalação , Animais , Comportamento Animal , Relação Dose-Resposta a Droga , Masculino , Ratos , Ratos Sprague-Dawley , Volatilização
16.
Nucl Med Biol ; 33(1): 71-9, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16459261

RESUMO

INTRODUCTION: One strength of small animal imaging is the ability to obtain longitudinal measurements within the same animal, effectively reducing the number of animals needed and increasing statistical power. However, the variability of within-rodent brain glucose uptake after an intraperitoneal injection across an extended time has not been measured. METHODS: Small animal imaging with 2-deoxy-2-[(18)F]-fluoro-D-glucose ((18)FDG) was used to determine the variability of a 50-min brain (18)FDG uptake following an intraperitoneal injection over time in awake male and female Sprague-Dawley rodents. RESULTS: After determining the variability of an intraperitoneal injection in the awake rat, we found that normalization of brain (18)FDG uptake for (1) injected dose and body weight or (2) body weight, plasma glucose concentration and injected dose resulted in a coefficient of variation (CV) of 15%. However, if we normalized regional uptake to whole brain to compare relative regional changes, the CV was less than 5%. Normalized cerebral (18)FDG uptake values were reproducible for a 2-week period in young adult animals. After 1 year, both male and female animals had reduced whole-brain uptake, as well as reduced regional hippocampal and striatal (18)FDG uptake. CONCLUSION: Overall, our results were similar to findings in previous rodent and human clinical populations; thus, using a high throughput study with intraperitoneal (18)FDG is a promising preclinical model for clinical populations. This is particularly relevant for measuring changes in brain function after experimental manipulation, such as long-term pharmacological administration.


Assuntos
Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Fluordesoxiglucose F18/farmacocinética , Animais , Feminino , Fluordesoxiglucose F18/administração & dosagem , Injeções Intraperitoneais , Cinética , Masculino , Taxa de Depuração Metabólica , Cintilografia , Compostos Radiofarmacêuticos/farmacocinética , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Fatores Sexuais
17.
J Addict Res Ther ; 7(4)2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-28078167

RESUMO

The rate of Neonatal Abstinence Syndrome (NAS) has drastically increased over the past decade. The average hospital expense per NAS patient has tripled, while the number of babies born to opioid-dependent mothers has increased to 5 in 1000 births. Current treatment options are limited to opioid replacement and tapering. Consequently, we examined the efficacy of prenatal, low-dose and short-term vigabatrin (γ-vinyl GABA, GVG) exposure for attenuating these symptoms as well as the metabolic changes observed in the brains of these animals upon reaching adolescence. Pregnant Sprague-Dawley rats were treated in one of four ways: 1) saline; 2) morphine alone; 3) morphine+GVG at 25 mg/kg; 4) morphine+GVG at 50 mg/kg. Morphine was administered throughout gestation, while GVG administration occurred only during the last 5 days of gestation. On post-natal day 1, naloxone-induced withdrawal behaviours were recorded in order to obtain a gross behaviour score. Approximately 28 days following birth, 18FDG microPET scans were obtained on these same animals (Groups 1, 2, and 4). Morphine-treated neonates demonstrated significantly higher withdrawal scores than saline controls. However, GVG at 50 but not 25 mg/kg/day significantly attenuated them. Upon reaching adolescence, morphine treated animals showed regionally specific changes in 18FDG uptake. Again, prenatal GVG exposure blocked them. These data demonstrate that low-dose, short-term prenatal GVG administration blocks naloxone-induced withdrawal in neonates. Taken together, these preliminary findings suggest that GVG may provide an alternative and long-lasting pharmacologic approach for the management of neonatal and adolescent symptoms associated with NAS.

18.
Drug Discov Today ; 10(8): 547-62, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15837598

RESUMO

Neurochemical imaging studies can identify molecular targets of abused drugs and link them to the underlying pathology associated with behaviors such as drug dependence, addiction and withdrawal. positron emission tomography (PET) is opening new avenues for the investigation of the neurochemical disturbances underlying drug abuse and addiction and the in vivo mechanisms by which medications might ameliorate these conditions. PET can identify vulnerable human populations, treatment strategies and monitor treatment efficacy. Thus, with this tool and the knowledge it provides, the potential for developing novel drugs and treatment strategies for drug addiction is now close at hand.


Assuntos
Tomografia por Emissão de Pósitrons/estatística & dados numéricos , Transtornos Relacionados ao Uso de Substâncias/diagnóstico , Animais , Previsões , Humanos , Tomografia por Emissão de Pósitrons/instrumentação , Tomografia por Emissão de Pósitrons/tendências , Terminologia como Assunto
19.
J Neurosci Methods ; 144(1): 25-34, 2005 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15848236

RESUMO

In the field of small animal positron emission tomography (PET), the assumptions underlying human and primate kinetic models may not be sustained in rodents. That is, the threshold dose at which a pharmacologic response occurs may be lower in small animals. In order to define this relationship, we combined microPET imaging using 11C-raclopride with microdialysis measures of extracellular fluid (ECF) dopamine (DA). In addition, we performed a series of studies in which a known mass of raclopride was microinfused into one striatum prior to a high specific activity (SA) systemic injection of 11C-raclopride. This single-injection approach provided a high and low SA region of radiotracer binding in the same animal during the same scanning session. Our data demonstrate that the binding potential (BP) declines above 3.5 pmol/ml (0.35 microg), with an ED50 of 8.55+/-5.62 pmol/ml. These data also provide evidence that BP may be compromised by masses of raclopride below 2.0 pmol/ml (0.326 microg). Increases in ECF DA were produced by mass doses of raclopride over 3.9 pmol/ml (0.329 microg) with an ED50 of 8.53+/-2.48 pmol/ml. Taken together, it appears that an optimal range of raclopride mass exists between 2.0 and 3.5 pmol/ml, around which the measured BP can be compromised by system sensitivity, endogenous DA, or excessive competition with unlabeled compound.


Assuntos
Encéfalo/diagnóstico por imagem , Isótopos de Carbono/farmacocinética , Antagonistas de Dopamina/farmacocinética , Microdiálise/métodos , Tomografia por Emissão de Pósitrons/métodos , Racloprida/farmacocinética , Animais , Ligação Competitiva/efeitos dos fármacos , Encéfalo/anatomia & histologia , Encéfalo/metabolismo , Mapeamento Encefálico , Isótopos de Carbono/sangue , Antagonistas de Dopamina/sangue , Relação Dose-Resposta a Droga , Lateralidade Funcional , Masculino , Racloprida/sangue , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
20.
Neuropsychopharmacology ; 28(12): 2192-8, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12888780

RESUMO

Positron emission tomography (PET), in combination with (11)C-raclopride, was used to examine the effects of phencyclidine (PCP) on dopamine (DA) in the primate striatum. In addition, we explored the hypotheses that GABAergic pathways as well as molecular targets beyond the N-methyl-D-aspartate (NMDA) receptor complex (ie dopamine transporter proteins, DAT) contribute to PCP's effects. In the first series of experiments, (11)C-raclopride was administered at baseline and 30 min following intravenous PCP administration. In the second series of studies, gamma-vinyl GABA (GVG) was used to assess whether enhanced GABAergic tone altered NMDA antagonist-induced changes in DA. Animals received an initial PET scan followed by pretreatment with GVG (300 mg/kg), then PCP 30 min prior to a second scan. Finally, we explored the possible contributions of DAT blockade to PCP-induced increases in DA. By examining (11)C-cocaine binding a paradigm in which PCP was coadministered with the radiotracer, we assessed the direct competition between these two compounds for the DAT. At 0.1, 0.5, and 1.0 mg/kg, PCP decreased (11)C-raclopride binding by 2.1, 14.9+/-2.2 and 8.18+/-1.1%, respectively. These effects were completely attenuated by GVG (3.38+/-3.1% decrease in (11)C-raclopride binding). Finally, PCP (0.5 mg/kg) decreased (11)C-cocaine binding by 25.5+/-4.3%, while at 1.0 mg/kg this decrease was 13.5%, consistent with a competitive interaction at the DAT. These results suggest that PCP may be exerting some direct effects through the DAT and that GABA partially modulates NMDA-antagonist-induced increases in striatal DA.


Assuntos
Corpo Estriado/efeitos dos fármacos , Dopamina/metabolismo , Antagonistas de Aminoácidos Excitatórios/farmacologia , Fenciclidina/farmacologia , Tomografia Computadorizada de Emissão , Análise de Variância , Animais , Anticonvulsivantes/farmacologia , Antipsicóticos/farmacocinética , Isótopos de Carbono/farmacocinética , Cerebelo/diagnóstico por imagem , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Cocaína/farmacocinética , Corpo Estriado/diagnóstico por imagem , Corpo Estriado/metabolismo , Relação Dose-Resposta a Droga , Interações Medicamentosas , Feminino , Papio , Racloprida/farmacocinética , Fatores de Tempo , Vigabatrina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA