Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Addict Res Ther ; 7(4)2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-28078167

RESUMEN

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.

2.
J Cereb Blood Flow Metab ; 34(8): 1315-20, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24824914

RESUMEN

Systemic lupus erythematosus (SLE) is characterized by multiorgan inflammation, neuropsychiatric disorders (NPSLE), and anti-nuclear antibodies. We previously identified a subset of anti-DNA antibodies (DNRAb) cross-reactive with the N-methyl-D-aspartate receptor, present in 30% to 40% of patients, able to enhance excitatory post-synaptic potentials and trigger neuronal apoptosis. DNRAb+ mice exhibit memory impairment or altered fear response, depending on whether the antibody penetrates the hippocampus or amygdala. Here, we used 18F-fluorodeoxyglucose (FDG) microPET to plot changes in brain metabolism after regional blood-brain barrier (BBB) breach. In DNRAb+ mice, metabolism declined at the site of BBB breach in the first 2 weeks and increased over the next 2 weeks. In contrast, DNRAb- mice exhibited metabolic increases in these regions over the 4 weeks after the insult. Memory impairment was present in DNRAb+ animals with hippocampal BBB breach and altered fear conditioning in DNRAb+ mice with amygdala BBB breach. In DNRAb+ mice, we observed an inverse relationship between neuron number and regional metabolism, while a positive correlation was observed in DNRAb- mice. These findings suggest that local metabolic alterations in this model take place through different mechanisms with distinct time courses, with important implications for the interpretation of imaging data in SLE subjects.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Anticuerpos Antinucleares/metabolismo , Barrera Hematoencefálica/metabolismo , Hipocampo/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Amígdala del Cerebelo/diagnóstico por imagen , Amígdala del Cerebelo/patología , Animales , Conducta Animal , Barrera Hematoencefálica/diagnóstico por imagen , Barrera Hematoencefálica/patología , Modelos Animales de Enfermedad , Femenino , Fluorodesoxiglucosa F18 , Hipocampo/diagnóstico por imagen , Hipocampo/patología , Lupus Eritematoso Sistémico/diagnóstico por imagen , Lupus Eritematoso Sistémico/patología , Lupus Eritematoso Sistémico/psicología , Ratones , Ratones Endogámicos BALB C , Neuronas/metabolismo , Neuronas/patología , Tomografía de Emisión de Positrones
3.
Mol Imaging ; 12(7): 1-15, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23920252

RESUMEN

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.


Asunto(s)
Diagnóstico por Imagen , Imagen Molecular , Edición , Proyectos de Investigación , Animales , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Ratones , Ratones Desnudos , Imagen Molecular/métodos , Revisión de la Investigación por Pares , Cintigrafía , Ratas , Tomografía Computarizada por Rayos X , Ultrasonografía
4.
Curr Top Behav Neurosci ; 11: 93-115, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22411423

RESUMEN

Behavioral neuroimaging is a rapidly evolving discipline that represents a marriage between the fields of behavioral neuroscience and preclinical molecular imaging. This union highlights the changing role of imaging in translational research. Techniques developed for humans are now widely applied in the study of animal models of brain disorders such as drug addiction. Small animal or preclinical imaging allows us to interrogate core features of addiction from both behavioral and biological endpoints. Snapshots of brain activity allow us to better understand changes in brain function and behavior associated with initial drug exposure, the emergence of drug escalation, and repeated bouts of drug withdrawal and relapse. Here we review the development and validation of new behavioral imaging paradigms and several clinically relevant radiotracers used to capture dynamic molecular events in behaving animals. We will discuss ways in which behavioral imaging protocols can be optimized to increase throughput and quantitative methods. Finally, we discuss our experience with the practical aspects of behavioral neuroimaging, so investigators can utilize effective animal models to better understand the addicted brain and behavior.


Asunto(s)
Encéfalo/patología , Modelos Animales de Enfermedad , Neuroimagen , Trastornos Relacionados con Sustancias , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/efectos de los fármacos , Dopamina/metabolismo , Fluorodesoxiglucosa F18/farmacocinética , Humanos , Racloprida/farmacocinética , Cintigrafía , Trastornos Relacionados con Sustancias/diagnóstico por imagen , Trastornos Relacionados con Sustancias/patología , Trastornos Relacionados con Sustancias/fisiopatología
5.
FEBS J ; 279(5): 871-81, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22240000

RESUMEN

There is increasing evidence that hyperoxia, particularly at the time of birth, may result in neurological injury, in particular to the susceptible vasculature of these tissues. This study was aimed at determining whether overexpression of extracellular superoxide dismutase (EC-SOD) is protective against brain injury induced by hyperoxia. Transgenic (TG) mice (with an extra copy of the human extracellular superoxide dismutase gene) and wild-type (WT) neonate mice were exposed to hyperoxia (95% of F(i) o(2) ) for 7 days after birth versus the control group in room air. Brain positron emission tomography (PET) scanning with fludeoxyglucose (FDG) isotope uptake was performed after exposure. To assess apoptosis induced by hyperoxia exposure, caspase 3 ELISA and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining were performed. Quantitative western blot for the following inflammatory markers was performed: glial fibrillary acidic protein, ionized calcium-binding adaptor molecule 1, macrophage-inhibiting factor, and phospho-AMP-activated protein kinase. PET scanning with FDG isotope uptake showed significantly higher uptake in the WT hyperoxia neonate brain group (0.14 ± 0.03) than in both the TG group (0.09 ± 0.01) and the control group (0.08 ± 0.02) (P< 0.05). Histopathological investigation showed more apoptosis and dead neurons in hippocampus and cerebellum brain sections of WT neonate mice after exposure to hyperoxia than in TG mice; this finding was also confirmed by TUNEL staining. The caspase 3 assay confirmed the finding of more apoptosis in WT hyperoxia neonates (0.814 ± 0.112) than in the TG hyperoxic group (0.579 ± 0.144) (P < 0.05); this finding was also confirmed by TUNEL staining. Quantitative western blotting for the inflammatory and metabolic markers showed significantly higher expression in the WT group than in the TG and control groups. Thus, overexpression of EC-SOD in the neonate brain offers significant protection against hyperoxia-induced brain damage.


Asunto(s)
Lesiones Encefálicas/enzimología , Lesiones Encefálicas/prevención & control , Hiperoxia/complicaciones , Superóxido Dismutasa/metabolismo , Animales , Animales Recién Nacidos , Apoptosis , Biomarcadores/metabolismo , Western Blotting , Lesiones Encefálicas/etiología , Ensayo de Inmunoadsorción Enzimática , Humanos , Hiperoxia/patología , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Estrés Oxidativo , Tomografía de Emisión de Positrones , Superóxido Dismutasa/genética
6.
Neuroimage ; 59(3): 2689-99, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21767654

RESUMEN

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.


Asunto(s)
Procesamiento de Imagen Asistido por Computador/métodos , Neurotransmisores/fisiología , Tomografía de Emisión de Positrones/métodos , Tomografía de Emisión de Positrones/estadística & datos numéricos , Algoritmos , Animales , Simulación por Computador , Dopamina/metabolismo , Antagonistas de Dopamina , Espacio Extracelular/efectos de los fármacos , Espacio Extracelular/metabolismo , Modelos Lineales , Microdiálisis , Modelos Neurológicos , Modelos Estadísticos , Racloprida , Radiofármacos , Ratas
7.
J Med Chem ; 55(1): 357-66, 2012 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-22128851

RESUMEN

Vigabatrin, a GABA aminotransferase (GABA-AT) inactivator, is used to treat infantile spasms and refractory complex partial seizures and is in clinical trials to treat addiction. We evaluated a novel GABA-AT inactivator (1S, 3S)-3-amino-4-difluoromethylenyl-1-cyclopentanoic acid (CPP-115, compound 1) and observed that it does not exhibit other GABAergic or off-target activities and is rapidly and completely orally absorbed and eliminated. By use of in vivo microdialysis techniques in freely moving rats and microPET imaging techniques, 1 produced similar inhibition of cocaine-induced increases in extracellular dopamine and in synaptic dopamine in the nucleus accumbens at (1)/(300) to (1)/(600) the dose of vigabatrin. It also blocks expression of cocaine-induced conditioned place preference at a dose (1)/(300) that of vigabatrin. Electroretinographic (ERG) responses in rats treated with 1, at doses 20-40 times higher than those needed to treat addiction in rats, exhibited reductions in ERG responses, which were less than the reductions observed in rats treated with vigabatrin at the same dose needed to treat addiction in rats. In conclusion, 1 can be administered at significantly lower doses than vigabatrin, which suggests a potential new treatment for addiction with a significantly reduced risk of visual field defects.


Asunto(s)
4-Aminobutirato Transaminasa/metabolismo , Ácidos Carboxílicos/síntesis química , Trastornos Relacionados con Cocaína/tratamiento farmacológico , Ciclopentanos/síntesis química , Animales , Disponibilidad Biológica , Ácidos Carboxílicos/farmacología , Ácidos Carboxílicos/toxicidad , Trastornos Relacionados con Cocaína/metabolismo , Trastornos Relacionados con Cocaína/psicología , Ciclopentanos/farmacología , Ciclopentanos/toxicidad , Perros , Dopamina/metabolismo , Electrorretinografía , Femenino , Proteínas Transportadoras de GABA en la Membrana Plasmática/fisiología , Inhibidores de Recaptación de GABA/síntesis química , Inhibidores de Recaptación de GABA/farmacología , Inhibidores de Recaptación de GABA/toxicidad , Humanos , Masculino , Ratones , Microdiálisis , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Oocitos/efectos de los fármacos , Oocitos/fisiología , Tomografía de Emisión de Positrones , Prolina/análogos & derivados , Ensayo de Unión Radioligante , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Receptores de GABA/metabolismo , Retina/efectos de los fármacos , Retina/fisiología , Estereoisomerismo , Distribución Tisular , Vigabatrin/farmacología , Xenopus laevis
8.
Proc Natl Acad Sci U S A ; 108(16): 6638-43, 2011 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-21464304

RESUMEN

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.


Asunto(s)
Encéfalo , Distonía , Vías Eferentes , Enfermedades Genéticas Congénitas , Chaperonas Moleculares/metabolismo , Trastornos del Movimiento , Alelos , Animales , Secuencia de Bases , Encéfalo/anomalías , Encéfalo/metabolismo , Distonía/genética , Distonía/metabolismo , Distonía/patología , Vías Eferentes/anomalías , Vías Eferentes/metabolismo , Técnicas de Sustitución del Gen , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/metabolismo , Enfermedades Genéticas Congénitas/patología , Humanos , Ratones , Ratones Transgénicos , Chaperonas Moleculares/genética , Trastornos del Movimiento/genética , Trastornos del Movimiento/metabolismo , Trastornos del Movimiento/patología , Eliminación de Secuencia
9.
Eur J Neurosci ; 30(8): 1565-75, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19821842

RESUMEN

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.


Asunto(s)
Anestésicos/farmacología , Encéfalo/efectos de los fármacos , Cloralosa/farmacología , Cocaína/farmacocinética , Isoflurano/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Volumen Sanguíneo/efectos de los fármacos , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Mapeo Encefálico/métodos , Isótopos de Carbono/farmacocinética , Circulación Cerebrovascular/efectos de los fármacos , Electrocardiografía/métodos , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Oxihemoglobinas/metabolismo , Tomografía de Emisión de Positrones/métodos , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
10.
J Neurosci ; 29(19): 6176-85, 2009 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-19439595

RESUMEN

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.


Asunto(s)
Trastornos Relacionados con Cocaína/fisiopatología , Cocaína , Cuerpo Estriado/fisiopatología , Señales (Psicología) , Dopamina/metabolismo , Animales , Radioisótopos de Carbono , Trastornos Relacionados con Cocaína/diagnóstico por imagen , Condicionamiento Psicológico , Cuerpo Estriado/diagnóstico por imagen , Antagonistas de Dopamina/farmacología , Antagonistas de los Receptores de Dopamina D2 , Masculino , Actividad Motora , Tomografía de Emisión de Positrones , Racloprida/metabolismo , Racloprida/farmacología , Ratas , Ratas Sprague-Dawley , Percepción Espacial/efectos de los fármacos
11.
Mol Imaging Biol ; 11(3): 137-43, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19132449

RESUMEN

PURPOSE: Salvinorin A (SA) is a potent and highly selective kappa-opioid receptor (KOR) agonist with rapid kinetics and commensurate behavioral effects; however, brain regions associated with these effects have not been determined. PROCEDURES: Freely moving adult male rats were given SA intraperitoneally during uptake and trapping of the brain metabolic radiotracer, 2-deoxy-2-[F-18]fluoro-D: -glucose (FDG), followed by image acquisition in a dedicated animal positron emission tomography (PET) system. Age-matched control animals received vehicle treatment. Animal behavior during FDG uptake was recorded digitally and later analyzed for locomotion. Group differences in regional FDG uptake normalized to whole brain were determined using Statistical Parametric Mapping (SPM) and verified by region of interest (ROI) analysis. RESULTS: SA-treated animals demonstrated significant increases in FDG uptake compared to controls in several brain regions associated with the distribution of KOR such as the periaqueductal grey, bed nucleus of the stria terminalis and the cerebellar vermis, as well as in the hypothalamus. Significant bilateral activations were also observed in the auditory, sensory, and frontal cortices. Regional decreases in metabolic demand were observed bilaterally in the dorsolateral striatum and hippocampus. Locomotor activity did not differ between SA and vehicle during FDG uptake. CONCLUSIONS: We have provided the first extensive maps of cerebral metabolic activation due to the potent kappa-opioid agonist, salvinorin A. A major finding from our small animal PET studies using FDG was that neural circuits affected by SA may not be limited to direct activation or inhibition of kappa-receptor-expressing cells. Instead, salvinorin A may trigger brain circuits that mediate the effects of the drug on cognition, mood, fear and anxiety, and motor output.


Asunto(s)
Encéfalo/metabolismo , Diterpenos de Tipo Clerodano/administración & dosificación , Receptores Opioides kappa/agonistas , Afecto , Animales , Conducta Animal/efectos de los fármacos , Mapeo Encefálico , Cognición , Fluorodesoxiglucosa F18/farmacocinética , Locomoción/efectos de los fármacos , Masculino , Metabolismo , Actividad Motora , Tomografía de Emisión de Positrones , Ratas
12.
Synapse ; 63(2): 87-94, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19016239

RESUMEN

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.


Asunto(s)
Trastornos Relacionados con Anfetaminas/prevención & control , Estimulantes del Sistema Nervioso Central/efectos adversos , GABAérgicos/farmacología , Metanfetamina/efectos adversos , Vigabatrin/farmacología , Animales , Condicionamiento Clásico/efectos de los fármacos , Extinción Psicológica/efectos de los fármacos , Isomerismo , Masculino , Ratas , Ratas Sprague-Dawley , Recurrencia
13.
Synapse ; 62(11): 870-2, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18720383

RESUMEN

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.


Asunto(s)
Vigabatrin/administración & dosificación , Vigabatrin/farmacología , Pérdida de Peso/efectos de los fármacos , Factores de Edad , Animales , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Cronoterapia/métodos , Inyecciones Intraperitoneales , Masculino , Obesidad/tratamiento farmacológico , Obesidad/fisiopatología , Ratas , Ratas Sprague-Dawley , Ratas Zucker , Vigabatrin/fisiología , Pérdida de Peso/fisiología
14.
Neuroimage ; 41(3): 1044-50, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18434204

RESUMEN

Salvia divinorum, a mint plant originally used by the Mazatecs of Oaxaca, Mexico in spiritual rituals has gained popularity, in smoked form, as a legal hallucinogen in the United States and Europe. Abuse results in rapid onset and short-lasting effects that include visual hallucinations and motor-function impairment. Salvinorin A, the psychoactive component of S. divinorum, is a uniquely potent agonist at kappa-opioid receptors, targets for new therapeutic drugs. We labeled salvinorin A with C-11 by acylation of salvinorin B with [11C]-acetyl chloride to study whether its kinetic behavior in the brain parallels its uniquely fast, yet brief physiological effects. Positron emission tomography (PET) studies performed in 6 adult female baboons indicated extremely rapid brain uptake reaching a peak accounting for 3.3% of the total administered dose in 40 s and clearing with a half-life of 8 min. [11C]-salvinorin A was distributed throughout the brain with the highest concentration in the cerebellum and a notable concentration in the visual cortex, perhaps accounting for its physiological effects when smoked. Naloxone administration did not reduce the overall concentration of [11C]-salvinorin A significantly nor did it change its regional distribution. Peripheral organ kinetics suggested at least two modes of metabolism and excretion occur: through the renal and biliary systems. Our findings have revealed that the exceptionally rapid uptake and brief duration of salvinorin A in the brain match the time-course of visual hallucinations for S. divinorum when smoked. The effects of salvinorin A may occur at <10 mug in the human brain, emphasizing its remarkable potency.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Diterpenos de Tipo Clerodano/farmacocinética , Alucinógenos/farmacocinética , Animales , Radioisótopos de Carbono/farmacocinética , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador , Papio , Tomografía de Emisión de Positrones
15.
Neuroimage ; 41(3): 1051-66, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18442926

RESUMEN

We investigated an imaging strategy that provides simultaneous measurements of radiotracer binding and behavior in awake, freely moving animals. In this strategy, animals are injected intravenously (i.v.) through a catheterized line and permitted to move freely for 30 min during uptake of the imaging agent, in this case 11C-raclopride. After this Awake Uptake period, animals are anesthetized and scanned for 25 min. We tested the utility of this strategy for measuring changes in striatal 11C-raclopride binding under control conditions (awake and freely moving in the home cage) and with several drug challenges: a loading dose of unlabeled raclopride, pretreatment with methamphetamine (METH) or pretreatment with gamma-vinyl-GABA [S+-GVG] followed by METH. An additional group of animals underwent a stress paradigm that we have previously shown increases brain dopamine. For drug challenge experiments, the change in 11C-raclopride binding was compared to data from animals that were anesthetized for the uptake period ("Anesthetized Uptake") and full time activity curves were used to calculate 11C-raclopride binding. Regardless of the drug treatment protocol, there was no difference in 11C-raclopride striatum to cerebellum ratio between the Awake versus the Anesthetized Uptake conditions. Awake and Anesthetized groups demonstrated over 90% occupancy of dopamine receptors with a loading dose of cold raclopride, both groups demonstrated approximately 30% reduction in 11C-raclopride binding from METH pretreatment and this effect was modulated to the same degree by GVG under both uptake conditions. Restraint during Awake Uptake decreased 11C-raclopride binding by 29%. These studies support a unique molecular imaging strategy in which radiotracer uptake occurs in freely moving animals, after which they are anesthetized and scanned. This imaging strategy extends the applicability of small animal PET to include functional neurotransmitter imaging and the neurochemical correlates of behavioral tasks.


Asunto(s)
Encéfalo/metabolismo , Antagonistas de Dopamina/metabolismo , Dopamina/metabolismo , Tomografía de Emisión de Positrones/métodos , Racloprida/metabolismo , Animales , Encéfalo/efectos de los fármacos , Radioisótopos de Carbono/metabolismo , Dopaminérgicos/farmacología , GABAérgicos/farmacología , Procesamiento de Imagen Asistido por Computador , Masculino , Metanfetamina/farmacología , Movimiento/fisiología , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Distribución Tisular , Vigabatrin/farmacología
16.
Mol Imaging Biol ; 10(2): 67-73, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18176804

RESUMEN

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.


Asunto(s)
Dopamina/metabolismo , Metanfetamina/farmacología , Microdiálisis/métodos , Neurotransmisores/metabolismo , Tomografía de Emisión de Positrones/métodos , Animales , Bombas de Infusión , Masculino , Metanfetamina/administración & dosificación , Racloprida/metabolismo , Ratas , Ratas Sprague-Dawley , Estándares de Referencia
17.
Nucl Med Biol ; 34(7): 833-47, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17921034

RESUMEN

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.


Asunto(s)
Encéfalo/diagnóstico por imagen , Sistemas de Liberación de Medicamentos/métodos , Técnicas de Sonda Molecular , Tomografía de Emisión de Positrones/métodos , Radiofármacos , Trastornos Relacionados con Sustancias/diagnóstico por imagen , Humanos , Pronóstico
18.
J Nucl Med ; 48(10): 1724-32, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17873134

RESUMEN

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.


Asunto(s)
Encéfalo/metabolismo , Cocaína/farmacocinética , Metanfetamina/farmacocinética , Papio/metabolismo , Animales , Encéfalo/diagnóstico por imagen , Tasa de Depuración Metabólica , Tomografía de Emisión de Positrones/métodos , Distribución Tisular
19.
Neuroimage ; 38(1): 34-42, 2007 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17707126

RESUMEN

Medically refractory seizures cause inflammation and neurodegeneration. Seizure initiation thresholds have been linked in mice to the serine protease tissue plasminogen activator (tPA); mice lacking tPA exhibit resistance to seizure induction, and the ensuing inflammation and neurodegeneration are similarly suppressed. Seizure foci in humans can be examined using PET employing 2-deoxy-2[(18)F]fluoro-d-glucose ((18)FDG) as a tracer to visualize metabolic dysfunction. However, there currently exist no such methods in mice to correlate measures of brain activation with behavior. Using a novel method for small animal PET data analysis, we examine patterns of (18)FDG uptake in wild-type and tPA(-/-) mice and find that they correlate with the severity of drug-induced seizure initiation. Furthermore, we report unexpected activations that may underlie the tPA modulation of seizure susceptibility. The methods described here should be applicable to other mouse models of human neurological disease.


Asunto(s)
Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Fluorodesoxiglucosa F18/farmacocinética , Convulsiones/diagnóstico por imagen , Convulsiones/metabolismo , Activador de Tejido Plasminógeno/metabolismo , Animales , Conducta Animal , Tasa de Depuración Metabólica , Ratones , Ratones Noqueados , Tomografía de Emisión de Positrones/métodos , Radiofármacos/farmacocinética , Activador de Tejido Plasminógeno/genética
20.
Brain Res ; 1144: 209-18, 2007 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-17346680

RESUMEN

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.


Asunto(s)
Conducta Animal/fisiología , Síntomas Conductuales/etiología , Encefalopatías Metabólicas/etiología , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/etiología , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Neurocirugia , Animales , Mapeo Encefálico , Fluorodesoxiglucosa F18 , Masculino , Actividad Motora/fisiología , Tomografía de Emisión de Positrones/métodos , Radiofármacos/metabolismo , Ratas , Ratas Sprague-Dawley , Reconocimiento en Psicología/fisiología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...