Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Neurobiol Dis ; 134: 104673, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31734455

RESUMEN

The recently developed DJ-1 knockout (KO) rat models the DJ-1 (or PARK7) loss-of-function mutation responsible for one form of early-onset familial Parkinson's disease (PD). Prior studies demonstrate that DJ-1 KO rats present progressive dopamine (DA) cell body degeneration in the substantia nigra pars compacta between 4 and 8 months of age. Furthermore, as some motor deficits emerge before the significant loss of DA cells, this mutation may yield a period of DA neuron dysfunction preceding cell death that may also contribute to cognitive impairments in early PD. However, cognitive functions subserved by corticostriatal circuitry, as well as additional alterations to the neurochemistry of monoamine systems, are largely uncharacterized in the DJ-1 KO rat. We therefore assessed a variety of striatally-mediated behavioral tasks, as well as the integrity of dopamine and serotonin systems, in male DJ-1 KO rats and wild-type (WT) controls at 4, 6, and 8 months of age. We demonstrate that DJ-1 KO rats exhibited motor impairments, but have intact goal-directed control over behavior in an appetitive instrumental learning task. Further, preprotachykinin mRNA expression, a post-synaptic indicator of DA signaling, was significantly decreased in 4-month DJ-1 KO rats, while DA transporter binding in the dorsal striatum did not differ between genotypes at any of the ages examined. Striatal tyrosine hydroxylase levels were significantly increased in 8-month DJ-1 KO rats and tended to be higher than WT at 4 and 6 months. Lastly, serotonin transporter binding was increased in the medial and orbitofrontal cortices of 4-month old DJ-1 KO rats. These results suggest that the nigrostriatal dopaminergic and prefrontal serotoninergic systems are altered early in the progression of DJ-1 KO pathology, despite no overt loss of the DA innervation of the striatum, and thus may be associated with early alterations in the functions of corticostriatal systems.


Asunto(s)
Conducta Animal/fisiología , Cuerpo Estriado/fisiología , Enfermedad de Parkinson/fisiopatología , Proteína Desglicasa DJ-1/fisiología , Animales , Condicionamiento Operante/fisiología , Modelos Animales de Enfermedad , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Técnicas de Inactivación de Genes , Masculino , Enfermedad de Parkinson/psicología , Corteza Prefrontal/metabolismo , Proteína Desglicasa DJ-1/genética , Precursores de Proteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Ratas Long-Evans , Taquicininas/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
2.
J Physiol ; 595(4): 1393-1412, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-27682823

RESUMEN

KEY POINTS: The lateral habenula (LHb) has been implicated in regulation of drug-seeking behaviours through aversion-mediated learning. In this study, we recorded neuronal activity in the LHb of rats during an operant task before and after ethanol-induced conditioned taste aversion (CTA) to saccharin. Ethanol-induced CTA caused significantly higher baseline firing rates in LHb neurons, as well as elevated firing rates in response to cue presentation, lever press and saccharin taste. In a separate cohort of rats, we found that bilateral LHb lesions blocked ethanol-induced CTA. Our results strongly suggest that excitation of LHb neurons is required for ethanol-induced CTA, and point towards a mechanism through which LHb firing may regulate voluntary ethanol consumption. ABSTRACT: Ethanol, like other drugs of abuse, has both rewarding and aversive properties. Previous work suggests that sensitivity to ethanol's aversive effects negatively modulates voluntary alcohol intake and thus may be important in vulnerability to developing alcohol use disorders. We previously found that rats with lesions of the lateral habenula (LHb), which is implicated in aversion-mediated learning, show accelerated escalation of voluntary ethanol consumption. To understand neural encoding in the LHb contributing to ethanol-induced aversion, we recorded neural firing in the LHb of freely behaving, water-deprived rats before and after an ethanol-induced (1.5 g kg-1 20% ethanol, i.p.) conditioned taste aversion (CTA) to saccharin taste. Ethanol-induced CTA strongly decreased motivation for saccharin in an operant task to obtain the tastant. Comparison of LHb neural firing before and after CTA induction revealed four main differences in firing properties. First, baseline firing after CTA induction was significantly higher. Second, firing evoked by cues signalling saccharin availability shifted from a pattern of primarily inhibition before CTA to primarily excitation after CTA induction. Third, CTA induction reduced the magnitude of lever press-evoked inhibition. Finally, firing rates were significantly higher during consumption of the devalued saccharin solution after CTA induction. Next, we studied sham- and LHb-lesioned rats in our operant CTA paradigm and found that LHb lesion significantly attenuated CTA effects in the operant task. Our data demonstrate the importance of LHb excitation in regulating expression of ethanol-induced aversion and suggest a mechanism for its role in modulating escalation of voluntary ethanol intake.


Asunto(s)
Consumo de Bebidas Alcohólicas/fisiopatología , Etanol/toxicidad , Potenciales Evocados Somatosensoriales , Habénula/fisiología , Neuronas/fisiología , Trastornos del Gusto/fisiopatología , Percepción del Gusto , Animales , Condicionamiento Operante , Habénula/citología , Masculino , Ratas , Ratas Long-Evans , Trastornos del Gusto/etiología
3.
Eur J Neurosci ; 45(11): 1418-1430, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28394447

RESUMEN

Opioid signaling in the nucleus accumbens shell (sNAcc) has been implicated in hedonic feeding and binge eating behavior. The sNAcc projects to the lateral hypothalamus (LH), and this pathway has been suggested to modulate palatability-driven feeding behavior. In this study, we investigated the effects of sNAcc mu opioid receptor (MOR) stimulation on firing rates of LH neurons in previously sated rats. Neural firing in the LH was recorded while food-deprived rats performed an operant task to obtain sweetened Intralipid (a 4% fat emulsion containing 5% sucrose) before and after bilateral sNAcc infusion of either a MOR agonist [D-Ala2, N-MePhe4, Gly-ol]-enkephalin (DAMGO) or a saline control solution. During sessions in which saline was infused into the sNAcc, the number of trials completed after infusion were significantly lower than the number completed before infusion, likely reflecting animals' increased satiety state. During sessions in which DAMGO was infused into the sNAcc, the decrease in the number of trials completed (comparing post- vs. pre-infusion trials) was significantly attenuated. Electrophysiological recording showed that the percentage of LH neurons showing an excitatory response due to behavioral events (cue presentation, lever press, lever retraction, and consumption) was reduced in post vs. pre-saline infusion period. However, the percentage of LH neurons showing excitatory responses to the same behavioral events was similar in pre- and post-DAMGO infusion periods. These findings suggest that MOR stimulation in sNAcc leads to an increase in stimulus-evoked excitatory signaling in LH neurons which could contribute to preventing satiety-induced decline in palatable food intake.


Asunto(s)
Conducta Alimentaria , Hipotálamo/metabolismo , Neuronas/fisiología , Núcleo Accumbens/metabolismo , Receptores Opioides mu/metabolismo , Saciedad , Animales , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Potenciales Evocados , Hipotálamo/citología , Hipotálamo/fisiología , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neurotransmisores/farmacología , Núcleo Accumbens/citología , Núcleo Accumbens/fisiología , Ratas , Ratas Long-Evans , Receptores Opioides mu/agonistas
4.
Synapse ; 70(4): 139-46, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26799527

RESUMEN

Preclinical studies demonstrate that repeated, high-dose methamphetamine administrations rapidly decrease plasmalemmal dopamine uptake, which may contribute to aberrant dopamine accumulation, reactive species generation, and long-term dopaminergic deficits. The present study extends these findings by demonstrating a heretofore unreported, epitope-specific modification in the dopamine transporter caused by a methamphetamine regimen that induces these deficits. Specifically, repeated, high-dose methamphetamine injections (4 × 10 mg/kg/injection, 2-h intervals) rapidly decreased immunohistochemical detection of striatal dopamine transporter as assessed 1 h after the final methamphetamine exposure. In contrast, neither a single high dose (1 × 10 mg/kg) nor repeated injections of a lower dose (4 × 2 mg/kg/injection) induced this change. The high-dose regimen-induced alteration was only detected using antibodies directed against the N-terminus. Immunohistochemical staining using antibodies directed against the C-terminus did not reveal any changes. The high-dose regimen also did not alter dopamine transporter expression as assessed using [(125) I]RTI-55 autoradiography. These data suggest that the repeated, high-dose methamphetamine regimen alters the N-terminus of the dopamine transporter. Further, these data may be predictive of persistent dopamine deficits caused by the stimulant. Future studies of the signaling cascades involved should provide novel insight into potential mechanisms underlying the physiological and pathophysiological regulation of the dopamine transporter.


Asunto(s)
Dopaminérgicos/farmacología , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Metanfetamina/farmacología , Secuencia de Aminoácidos , Animales , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Dopaminérgicos/administración & dosificación , Dopaminérgicos/toxicidad , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/química , Epítopos/metabolismo , Masculino , Metanfetamina/administración & dosificación , Metanfetamina/toxicidad , Datos de Secuencia Molecular , Unión Proteica , Dominios Proteicos , Ratas , Ratas Sprague-Dawley
5.
J Neurochem ; 125(4): 566-74, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23414433

RESUMEN

Neurotoxic regimens of methamphetamine (METH) result in reactive microglia and astrocytes in striatum. Prior data indicate that rats with partial dopamine (DA) loss resulting from prior exposure to METH are resistant to further decreases in striatal DA when re-exposed to METH 30 days later. Such resistant animals also do not show an activated microglia phenotype, suggesting a relation between microglial activation and METH-induced neurotoxicity. To date, the astrocyte response in such resistance has not been examined. Thus, this study examined glial-fibrillary acidic protein (GFAP) and CD11b protein expression in striata of animals administered saline or a neurotoxic regimen of METH on post-natal days 60 and/or 90 (Saline:Saline, Saline:METH, METH:Saline, METH:METH). Consistent with previous work, animals experiencing acute toxicity (Saline:METH) showed both activated microglia and astocytes, whereas those resistant to the acute toxicity (METH:METH) did not show activated microglia. Interestingly, GFAP expression remained elevated in rats exposed to METH at PND60 (METH:Saline), and was not elevated further in resistant rats treated for the second time with METH (METH:METH). These data suggest that astrocytes remain reactive up to 30 days post-METH exposure. In addition, these data indicate that astrocyte reactivity does not reflect acute, METH-induced DA terminal toxicity, whereas microglial reactivity does.


Asunto(s)
Astrocitos/fisiología , Dopamina/fisiología , Metanfetamina/toxicidad , Microglía/fisiología , Síndromes de Neurotoxicidad/fisiopatología , Trastornos Relacionados con Anfetaminas/genética , Trastornos Relacionados con Anfetaminas/fisiopatología , Animales , Astrocitos/efectos de los fármacos , Antígeno CD11b/genética , Estimulantes del Sistema Nervioso Central/toxicidad , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/fisiopatología , Fiebre/fisiopatología , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Proteína Ácida Fibrilar de la Glía/genética , Masculino , Microglía/efectos de los fármacos , Síndromes de Neurotoxicidad/genética , Ratas , Ratas Sprague-Dawley
6.
J Neurochem ; 125(4): 555-65, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23480199

RESUMEN

Methamphetamine-induced partial dopamine depletions are associated with impaired basal ganglia function, including decreased preprotachykinin mRNA expression and impaired transcriptional activation of activity-regulated, cytoskeleton-associated (Arc) gene in striatum. Recent work implicates deficits in phasic dopamine signaling as a potential mechanism linking methamphetamine-induced dopamine loss to impaired basal ganglia function. This study thus sought to establish a causal link between phasic dopamine transmission and altered basal ganglia function by determining whether the deficits in striatal neuron gene expression could be restored by increasing phasic dopamine release. Three weeks after pretreatment with saline or a neurotoxic regimen of methamphetamine, rats underwent phasic- or tonic-like stimulation of ascending dopamine neurons. Striatal gene expression was examined using in situ hybridization histochemistry. Phasic-like, but not tonic-like, stimulation induced immediate-early genes Arc and zif268 in both groups, despite the partial striatal dopamine denervation in methamphetamine-pretreated rats, with the Arc expression occurring in presumed striatonigral efferent neurons. Phasic-like stimulation also restored preprotachykinin mRNA expression. These results suggest that disruption of phasic dopamine signaling likely underlies methamphetamine-induced impairments in basal ganglia function, and that restoring phasic dopamine signaling may be a viable approach to manage long-term consequences of methamphetamine-induced dopamine loss on basal ganglia functions.


Asunto(s)
Cuerpo Estriado/fisiología , Dopamina/fisiología , Neuronas Dopaminérgicas/fisiología , Haz Prosencefálico Medial/fisiología , Metanfetamina/toxicidad , Síndromes de Neurotoxicidad/fisiopatología , Trastornos Relacionados con Anfetaminas/genética , Trastornos Relacionados con Anfetaminas/fisiopatología , Animales , Estimulantes del Sistema Nervioso Central/toxicidad , Cuerpo Estriado/efectos de los fármacos , Proteínas del Citoesqueleto/genética , Desnervación/métodos , Neuronas Dopaminérgicas/efectos de los fármacos , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Estimulación Eléctrica/métodos , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Genes Inmediatos-Precoces/genética , Masculino , Haz Prosencefálico Medial/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Síndromes de Neurotoxicidad/genética , Precursores de Proteínas/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Taquicininas/genética
7.
Eur J Neurosci ; 38(1): 2078-88, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23574406

RESUMEN

Methamphetamine (METH) is a highly addictive drug that is also neurotoxic to central dopamine (DA) systems. Although striatal DA depletions induced by METH are associated with behavioral and cognitive impairments, the link between these phenomena remains poorly understood. Previous work in both METH-pretreated animals and the 6-hydroxydopamine model of Parkinson's disease suggests that a disruption of phasic DA signaling, which is important for learning and goal-directed behavior, may be such a link. However, previous studies used electrical stimulation to elicit phasic-like DA responses and were also performed under anesthesia, which alters DA neuron activity and presynaptic function. Here we investigated the consequences of METH-induced DA terminal loss on both electrically evoked phasic-like DA signals and so-called 'spontaneous' phasic DA transients measured by voltammetry in awake rats. Not ostensibly attributable to discrete stimuli, these subsecond DA changes may play a role in enhancing reward-cue associations. METH pretreatment reduced tissue DA content in the dorsomedial striatum and nucleus accumbens by ~55%. Analysis of phasic-like DA responses elicited by reinforcing stimulation revealed that METH pretreatment decreased their amplitude and underlying mechanisms for release and uptake to a similar degree as DA content in both striatal subregions. Most importantly, characteristics of DA transients were altered by METH-induced DA terminal loss, with amplitude and frequency decreased and duration increased. These results demonstrate for the first time that denervation of DA neurons alters naturally occurring DA transients and are consistent with diminished phasic DA signaling as a plausible mechanism linking METH-induced striatal DA depletions and cognitive deficits.


Asunto(s)
Dopaminérgicos/toxicidad , Dopamina/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Metanfetamina/toxicidad , Transmisión Sináptica , Animales , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Cuerpo Estriado/fisiología , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/fisiología , Masculino , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Núcleo Accumbens/fisiología , Ratas , Ratas Sprague-Dawley
8.
J Pharmacol Exp Ther ; 344(2): 511-21, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23230214

RESUMEN

Nitric oxide is implicated in methamphetamine (METH)-induced neurotoxicity; however, the source of the nitric oxide has not been identified. Previous work has also revealed that animals with partial dopamine loss induced by a neurotoxic regimen of methamphetamine fail to exhibit further decreases in striatal dopamine when re-exposed to methamphetamine 7-30 days later. The current study examined nitric oxide synthase expression and activity and protein nitration in striata of animals administered saline or neurotoxic regimens of methamphetamine at postnatal days 60 and/or 90, resulting in four treatment groups: Saline:Saline, METH:Saline, Saline:METH, and METH:METH. Acute administration of methamphetamine on postnatal day 90 (Saline:METH and METH:METH) increased nitric oxide production, as evidenced by increased protein nitration. Methamphetamine did not, however, change the expression of endothelial or inducible isoforms of nitric oxide synthase, nor did it change the number of cells positive for neuronal nitric oxide synthase mRNA expression or the amount of neuronal nitric oxide synthase mRNA per cell. However, nitric oxide synthase activity in striatal interneurons was increased in the Saline:METH and METH:METH animals. These data suggest that increased nitric oxide production after a neurotoxic regimen of methamphetamine results from increased nitric oxide synthase activity, rather than an induction of mRNA, and that constitutively expressed neuronal nitric oxide synthase is the most likely source of nitric oxide after methamphetamine administration. Of interest, animals rendered resistant to further methamphetamine-induced dopamine depletions still show equivalent degrees of methamphetamine-induced nitric oxide production, suggesting that nitric oxide production alone in response to methamphetamine is not sufficient to induce acute neurotoxic injury.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Metanfetamina/toxicidad , Síndromes de Neurotoxicidad/enzimología , Óxido Nítrico Sintasa , Óxido Nítrico/biosíntesis , Animales , Cuerpo Estriado/enzimología , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Inducción Enzimática , Hibridación in Situ , Interneuronas/efectos de los fármacos , Interneuronas/enzimología , Interneuronas/metabolismo , Isoenzimas , Masculino , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/metabolismo , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo I/biosíntesis , Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ratas , Ratas Sprague-Dawley
9.
Front Mol Neurosci ; 16: 1160157, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37251646

RESUMEN

The shift from drug abuse to addiction is considered to arise from the transition between goal-directed and habitual control over drug behavior. Habitual responding for appetitive and skill-based behaviors is mediated by potentiated glutamate signaling in the dorsolateral striatum (DLS), but the state of the DLS glutamate system in the context of habitual drug-behavior remains undefined. Evidence from the nucleus accumbens of cocaine-experienced rats suggests that decreased transporter-mediated glutamate clearance and enhanced synaptic glutamate release contribute to the potentiated glutamate signaling that underlies the enduring vulnerability to relapse. Preliminary evidence from the dorsal striatum of cocaine-experienced rats suggests that this region exhibits similar alterations to glutamate clearance and release, but it is not known whether these glutamate dynamics are associated with goal-directed or habitual control over cocaine-seeking behavior. Therefore, we trained rats to self-administer cocaine in a chained cocaine-seeking and -taking paradigm, which yielded goal-directed, intermediate, and habitual cocaine-seeking rats. We then assessed glutamate clearance and release dynamics in the DLS of these rats using two different methods: synaptic transporter current (STC) recordings of patch-clamped astrocytes and the intensity-based glutamate sensing fluorescent reporter (iGluSnFr). While we observed a decreased rate of glutamate clearance in STCs evoked with single-pulse stimulation in cocaine-experienced rats, we did not observe any cocaine-induced differences in glutamate clearance rates from STCs evoked with high frequency stimulation (HFS) or iGluSnFr responses evoked with either double-pulse stimulation or HFS. Furthermore, GLT-1 protein expression in the DLS was unchanged in cocaine-experienced rats, regardless of their mode of control over cocaine-seeking behavior. Lastly, there were no differences in metrics of glutamate release between cocaine-experienced rats and yoked-saline controls in either assay. Together, these results suggest that glutamate clearance and release dynamics in the DLS are largely unaltered by a history of cocaine self-administration on this established cocaine seeking-taking paradigm, regardless of whether the control over the cocaine seeking behavior was habitual or goal directed.

10.
J Neurochem ; 123(5): 845-55, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22978492

RESUMEN

The immediate-early gene Arc (activity-regulated cytoskeleton-associated protein) is provocative in the context of neuroplasticity because of its experience-dependent regulation and mRNA transport to and translation at activated synapses. Normal rats have more preproenkephalin-negative (ppe-neg; presumed striatonigral) neurons with cytoplasmic Arc mRNA than ppe-positive (ppe-pos; striatopallidal) neurons, despite equivalent numbers of these neurons showing novelty-induced transcriptional activation of Arc. Furthermore, rats with partial monoamine loss induced by methamphetamine (METH) show impaired Arc mRNA expression in both ppe-neg and ppe-pos neurons relative to normal animals following response-reversal learning. In this study, Arc expression induced by exposure to a novel environment was used to assess transcriptional activation and cytoplasmic localization of Arc mRNA in striatal efferent neuron subpopulations subsequent to METH-induced neurotoxicity. Partial monoamine depletion significantly altered Arc expression. Specifically, basal Arc expression was elevated, but novelty-induced transcriptional activation was abolished. Without novelty-induced Arc transcription, METH-pre-treated rats also had fewer neurons with cytoplasmic Arc mRNA expression, with the effect being greater for ppe-neg neurons. Thus, METH-induced neurotoxicity substantially alters striatal efferent neuron function at the level of Arc transcription, suggesting a long-term shift in basal ganglia neuroplasticity processes subsequent to METH-induced neurotoxicity. Such changes potentially underlie striatally based learning deficits associated with METH-induced neurotoxicity.


Asunto(s)
Estimulantes del Sistema Nervioso Central/toxicidad , Proteínas del Citoesqueleto/biosíntesis , Dopamina/metabolismo , Metanfetamina/toxicidad , Proteínas del Tejido Nervioso/biosíntesis , Neuronas Eferentes/metabolismo , Animales , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Regulación de la Expresión Génica , Hibridación Fluorescente in Situ , Masculino , Neuronas Eferentes/efectos de los fármacos , Síndromes de Neurotoxicidad/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Transcripción Genética/efectos de los fármacos
11.
Psychopharmacology (Berl) ; 239(1): 93-104, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34985532

RESUMEN

RATIONALE: Methamphetamine (METH) exposure is associated with damage to central monoamine systems, particularly dopamine signaling. Rodent models of such damage have revealed a decrease in the amplitude of phasic dopamine signals and significant striatal dysfunction, including changes in the molecular, system, and behavioral functions of the striatum. Dopamine signaling through D1 receptors promotes corticostriatal long-term potentiation (LTP), a critical substrate of these striatal functions. OBJECTIVES: Therefore, the purpose of this study was to determine if METH-induced dopamine neurotoxicity would impair D1 receptor-dependent striatal LTP in mice. METHODS: Mice were treated with a METH binge regimen (4 × 10 mg/kg d,l-methamphetamine, s.c.) that recapitulates all of the known METH-induced neurotoxic effects observed in humans, including dopamine toxicity. Three weeks later, acute brain slices containing either the dorsomedial striatum (DMS) or dorsolateral striatum (DLS) were prepared, and plasticity was assessed using white matter, high-frequency stimulation (HFS), and striatal extracellular electrophysiology. RESULTS: Under these conditions, LTP was induced in brain slices containing the DMS from saline-pretreated mice, but not mice with METH-induced neurotoxicity. Furthermore, the LTP observed in DMS slices from saline-pretreated mice was blocked by the dopamine D1 receptor antagonist SCH23390, indicating that this LTP is dopamine D1 receptor-dependent. Finally, acute in vivo treatment of METH-pretreated mice with bupropion (50 mg/kg, i.p.) promoted LTP in DMS slices. CONCLUSIONS: Together, these studies demonstrate that METH-induced neurotoxicity impairs dopamine D1 receptor-dependent LTP within the DMS and that the FDA-approved drug bupropion restores induction of striatal LTP in mice with METH-induced dopamine neurotoxicity.


Asunto(s)
Metanfetamina , Síndromes de Neurotoxicidad , Animales , Cuerpo Estriado , Dopamina , Potenciación a Largo Plazo , Metanfetamina/toxicidad , Ratones
12.
J Neurochem ; 118(4): 668-76, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21668447

RESUMEN

Neurotoxic doses of methamphetamine (METH) are known to cause depletions in striatal dopamine (DA) tissue content. However, the effects of METH-induced insults on dopaminergic neurotransmission are not fully understood. Here, we employed fast-scan cyclic voltammetry at a carbon-fiber microelectrode in the anesthetized rat striatum to assess the effects of a neurotoxic regimen of METH on phasic and tonic modes of dopaminergic signaling and underlying mechanisms of DA release and uptake. Extracellular DA was electrically evoked by stimulation of the medial forebrain bundle mimicking tonic and phasic firing patterns for dopaminergic cells and was monitored simultaneously in both the dorsomedial and dorsolateral striatum. Kinetic analysis of evoked recordings determined parameters describing DA release and uptake. Striatal DA tissue content was quantified by high performance liquid chromatography with electrochemical detection. METH-pretreatment (four doses of 7.5 or 10.0 mg/kg s.c.) induced DA depletions of ∼ 40% on average, which are reported in both striatal subregions. METH pre-treatment significantly decreased the amplitude of signals evoked by phasic, but not tonic, stimulation. Parameters for DA release and uptake were also similarly reduced by ∼ 40%, consistent with effects on evoked phasic-like responses and DA tissue content. Taken together, these results suggest that METH-pretreatment selectively diminishes phasic, but not tonic, dopaminergic signaling in the dorsal striatum.


Asunto(s)
Estimulantes del Sistema Nervioso Central/toxicidad , Cuerpo Estriado/fisiopatología , Dopamina/fisiología , Metanfetamina/toxicidad , Síndromes de Neurotoxicidad/fisiopatología , Transducción de Señal/fisiología , Algoritmos , Animales , Dopamina/metabolismo , Estimulación Eléctrica , Fenómenos Electrofisiológicos , Espacio Extracelular/metabolismo , Cinética , Masculino , Microelectrodos , Neuronas/fisiología , Oxidopamina , Enfermedad de Parkinson Secundaria/fisiopatología , Ratas , Ratas Sprague-Dawley , Simpatectomía Química , Simpaticolíticos
13.
J Clin Invest ; 131(10)2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-33848264

RESUMEN

Opioid use disorder (OUD) has become a leading cause of death in the United States, yet current therapeutic strategies remain highly inadequate. To identify potential treatments for OUD, we screened a targeted selection of over 100 drugs using a recently developed opioid self-administration assay in zebrafish. This paradigm showed that finasteride, a steroidogenesis inhibitor approved for the treatment of benign prostatic hyperplasia and androgenetic alopecia, reduced self-administration of multiple opioids without affecting locomotion or feeding behavior. These findings were confirmed in rats; furthermore, finasteride reduced the physical signs associated with opioid withdrawal. In rat models of neuropathic pain, finasteride did not alter the antinociceptive effect of opioids and reduced withdrawal-induced hyperalgesia. Steroidomic analyses of the brains of fish treated with finasteride revealed a significant increase in dehydroepiandrosterone sulfate (DHEAS). Treatment with precursors of DHEAS reduced opioid self-administration in zebrafish in a fashion akin to the effects of finasteride. These results highlight the importance of steroidogenic pathways as a rich source of therapeutic targets for OUD and point to the potential of finasteride as a new treatment option for this disorder.


Asunto(s)
Inhibidores de 5-alfa-Reductasa/farmacología , Finasterida/farmacología , Trastornos Relacionados con Opioides/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Humanos , Masculino , Trastornos Relacionados con Opioides/fisiopatología , Ratas , Ratas Sprague-Dawley , Pez Cebra
14.
Pharmacol Biochem Behav ; 198: 173033, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32888972

RESUMEN

Instrumental actions are initially goal-directed and driven by their associated outcome. However, with repeated experience habitual actions develop which are automated and efficient, as they are instead driven by antecedent stimuli. Dopamine is thought to facilitate the transition from goal-directed to habitual actions. This idea has been largely derived from evidence that psychostimulants accelerate the development of habitual actions. In the current study, we examined the impact of L-dopa (levodopa or L-dihydroxyphenylalanine), which also potentiates dopamine activity, on habitual learning. L-dopa was systemically administered prior to training rats to press a lever for a food outcome. When tested, L-dopa exposed animals were insensitive to changes in the value of the food outcome, and hence demonstrated accelerated habitual behavioral control compared to control animals that remained goal directed. We also showed that when N-acetylcysteine (NAC), an antioxidant and regulator of glutamate activity, was co-administered with L-dopa, it prevented the transition to habitual behavior; an effect demonstrated previously for cocaine. Therefore, this study establishes similarities between L-dopa and psychostimulants in both the development and prevention of habitual actions, and supports the notion that excess dopamine potentiates habitual learning. This finding extends the limited existing knowledge of the impact of L-dopa on learning and behavior, and has implications for neurological disorders where L-dopa is the primary treatment.


Asunto(s)
Acetilcisteína/farmacología , Conducta Animal/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Levodopa/farmacología , Animales , Antioxidantes/farmacología , Conducta Adictiva/tratamiento farmacológico , Conducta Adictiva/metabolismo , Dopamina/metabolismo , Dopaminérgicos/farmacología , Ácido Glutámico/metabolismo , Hábitos , Masculino , Actividad Motora/efectos de los fármacos , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Ratas , Ratas Long-Evans
15.
Eur J Neurosci ; 28(10): 2041-52, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19046385

RESUMEN

The striatum is crucially involved in motor and cognitive function, and receives significant glutamate input from the cortex and thalamus. The corticostriatal pathway arises from diverse regions of the cortex and is thought to provide information to the basal ganglia from which motor actions are selected and modified. The thalamostriatal pathway arises from specific thalamic nuclei and is involved in attention and possibly strategy switching. Despite these fundamental functional differences, direct comparisons of the properties of these pathways are lacking. N-methyl-D-aspartate (NMDA) receptors at synapses powerfully affect postsynaptic processing, and incorporation of different NR2 subunits into NMDA receptors has profound effects on the pharmacological and biophysical properties of the receptor. Utilization of different NMDA receptors at thalamostriatal and corticostriatal synapses could allow for afferent-specific differences in information processing. We used a novel rat brain slice preparation preserving corticostriatal and thalamostriatal pathways to medium spiny neurons to examine the properties of NMDA receptor-mediated excitatory postsynaptic currents (EPSCs) recorded using the whole-cell, patch-clamp technique. Within the same neuron, the NMDA/non-NMDA ratio is greater for excitatory responses evoked from the thalamostriatal pathway than for those evoked from the corticostriatal pathway. In addition, reversal potentials and decay kinetics of the NMDA receptor-mediated EPSCs suggest that the thalamostriatal synapse is more distant on the dendritic arbor. Finally, results obtained with antagonists specific for NR2B-containing NMDA receptors imply that NMDA receptors at corticostriatal synapses contain more NR2B subunits. These synapse-specific differences in NMDA receptor content and pharmacology provide potential differential sites of action for NMDA receptor subtype-specific antagonists proposed for the treatment of Parkinson's disease.


Asunto(s)
Corteza Cerebral/metabolismo , Cuerpo Estriado/metabolismo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transmisión Sináptica/fisiología , Tálamo/metabolismo , Animales , Corteza Cerebral/citología , Cuerpo Estriado/citología , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/metabolismo , Espinas Dendríticas/ultraestructura , Potenciales Postsinápticos Excitadores/fisiología , Ácido Glutámico/metabolismo , Masculino , Vías Nerviosas/citología , Vías Nerviosas/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Sinapsis/ultraestructura , Transmisión Sináptica/efectos de los fármacos , Tálamo/citología
16.
Neurotox Res ; 14(4): 307-15, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19073434

RESUMEN

Methamphetamine abuse results in lasting, partial depletions of striatal dopamine and cognitive dysfunction. However, the effect of partial dopamine depletions on the expression of an effector immediate early gene, Arc (activity regulated, cytoskeletal-associated protein), known to be involved in synaptic modifications underlying learning and memory, has heretofore not been examined. Male Sprague-Dawley rats were pretreated with a neurotoxic regimen of methamphetamine or saline. Seven weeks later, rats were trained in a motor-response task on a T-maze for five days, and then underwent reversal training on day five. Rats were sacrificed 5 min after reaching criterion on the reversal task, and the brains were removed and processed using double-label fluorescent in situ hybridization for Arc and preproenkephalin (PPE) mRNA expression in the dorsomedial striatum. Rats pretreated with methamphetamine had an average (+/-SEM) 54.4+/-7.9% loss of dopamine in dorsomedial striatum. Interestingly, there was no difference in reversal trials to criterion in methamphetamine- vs. saline-pretreated rats. However, the expression of Arc mRNA in dorsomedial striatum was attenuated in methamphetamine-pretreated animals, particularly in PPE-negative neurons. Furthermore, the correlation between Arc mRNA expression in dorsomedial striatum and learning was abolished in methamphetamine-pretreated animals. These data suggest that methamphetamine-induced partial monoamine loss is associated with disrupted induction of the effector immediate early gene Arc during a behavioral task, particularly in PPE-negative (presumed striatonigral) neurons, as well as with disruption of the relation between Arc mRNA expression in dorsomedial striatum and reversal learning.


Asunto(s)
Proteínas del Citoesqueleto/genética , Aprendizaje por Laberinto/efectos de los fármacos , Metanfetamina/toxicidad , Proteínas del Tejido Nervioso/genética , Neuronas Eferentes/efectos de los fármacos , Neuronas Eferentes/metabolismo , Animales , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Proteínas del Citoesqueleto/efectos de los fármacos , Dopamina/metabolismo , Encefalinas/genética , Encefalinas/metabolismo , Genes Inmediatos-Precoces/efectos de los fármacos , Masculino , Proteínas del Tejido Nervioso/efectos de los fármacos , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Aprendizaje Inverso/efectos de los fármacos
17.
J Neurosci Methods ; 159(2): 224-35, 2007 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-16899300

RESUMEN

The striatum, the primary input nucleus of the basal ganglia, is crucially involved in motor and cognitive function and receives significant glutamate input from the cortex and thalamus. Increasing evidence suggests fundamental differences between these afferents, yet direct comparisons have been lacking. We describe a slice preparation that allows for direct comparison of the pharmacology and biophysics of these two pathways. Visualization of slices from animals previously injected with BDA into the parafascicular nucleus revealed the presence of axons of thalamic origin in the slice. These axons were especially well-preserved after traversing the reticular nucleus, the location chosen for stimulation of thalamostriatal afferents. Initial characterization of the two pathways revealed both non-NMDA and NMDA receptor-mediated currents at synapses from both afferents and convergence of the afferents in 51% of striatal efferent neurons. Annihilation of action potentials was not observed in collision experiments, nor was current spread from the site of stimulation to striatum found. Differences in short-term plasticity suggest that the probability of release differs for the two inputs. The present work thus provides a novel rat brain slice preparation in which the effects of selective stimulation of cortical versus thalamic afferents to striatum can be studied in the same preparation.


Asunto(s)
Cuerpo Estriado/citología , Cuerpo Estriado/fisiología , Técnicas de Cultivo de Órganos/métodos , Tálamo/citología , Tálamo/fisiología , Vías Aferentes , Animales , Estimulación Eléctrica , Electrofisiología , Potenciales Postsinápticos Excitadores/fisiología , Masculino , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/fisiología
18.
Behav Brain Res ; 328: 195-208, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28432009

RESUMEN

The lateral habenula (LHb) is an epithalamic brain region implicated in aversive processing via negative modulation of midbrain dopamine (DA) and serotonin (5-HT) systems. Given the role of the LHb in inhibiting DA and 5-HT systems, it is thought to be involved in various psychiatric pathologies, including drug addiction. In support, it has been shown that LHb plays a critical role in cocaine- and ethanol-related behaviors, most likely by mediating drug-induced aversive conditioning. In our previous work, we showed that LHb lesions increased voluntary ethanol consumption and operant ethanol self-administration and blocked yohimbine-induced reinstatement of ethanol self-administration. LHb lesions also attenuated ethanol-induced conditioned taste aversion suggesting that a mechanism for the increased intake of ethanol may be reduced aversion learning. However, whether afferents to the LHb are required for mediating effects of the LHb on these behaviors remained to be investigated. Our present results show that lesioning the fiber bundle carrying afferent inputs to the LHb, the stria medullaris (SM), increases voluntary ethanol consumption, suggesting that afferent structures projecting to the LHb are important for mediating ethanol-directed behaviors. We then chose two afferent structures as the focus of our investigation. We specifically studied the role of the inputs from the lateral hypothalamus (LH) and ventral pallidum (VP) to the LHb in ethanol-directed behaviors. Our results show that the LH-LHb projection is necessary for regulating voluntary ethanol consumption. These results are an important first step towards understanding the functional role of afferents to LHb with regard to ethanol consumption.


Asunto(s)
Consumo de Bebidas Alcohólicas/fisiopatología , Prosencéfalo Basal/fisiopatología , Habénula/fisiopatología , Área Hipotalámica Lateral/fisiopatología , Consumo de Bebidas Alcohólicas/patología , Animales , Prosencéfalo Basal/patología , Depresores del Sistema Nervioso Central/administración & dosificación , Comportamiento de Búsqueda de Drogas/fisiología , Etanol/administración & dosificación , Habénula/patología , Área Hipotalámica Lateral/patología , Masculino , Vías Nerviosas/patología , Vías Nerviosas/fisiopatología , Ratas Long-Evans , Autoadministración , Volición
19.
Eur J Pharmacol ; 532(1-2): 61-73, 2006 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-16443216

RESUMEN

Several studies have shown that psychostimulants can induce differential immediate early gene and neuropeptide expression in the patch versus matrix compartments of dorsal striatum. The patch compartment contains a high density of mu opioid receptors and activation of these receptors may contribute to psychostimulant-induced gene expression in the patch versus matrix compartments of dorsal striatum. However, the contribution of mu opioid receptor activation to psychostimulant-induced changes in gene expression in the patch compartment of dorsal striatum has not been examined. The current study examined the role of mu opioid receptors in psychostimulant induction of preprodynorphin, c-fos and zif/268 messenger RNA expression in the patch versus matrix compartments of dorsal striatum. Male Sprague-Dawley rats were treated with the mu opioid receptor antagonist, clocinnamox (1 mg/kg, s.c.), 24 h prior to treatment with cocaine (30 mg/kg, i.p.) or methamphetamine (15 mg/kg, s.c.) and sacrificed 45 min or 3 h later. Mu opioid receptor antagonism blocked psychostimulant-induced preprodynorphin messenger RNA expression only in the rostral patch compartment, whereas psychostimulant-induced zif/268 messenger RNA expression in the patch and matrix compartments was attenuated throughout the dorsal striatum. Clocinnamox pretreatment had no effect on stimulant-induced increases in c-fos expression. These data suggest that mu opioid receptor activation plays a specific role in psychostimulant-induced preprodynorphin messenger RNA expression in the rostral patch compartment and zif/268 messenger RNA expression throughout dorsal striatum.


Asunto(s)
Cinamatos/farmacología , Cuerpo Estriado/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Derivados de la Morfina/farmacología , ARN Mensajero/metabolismo , Receptores Opioides mu/antagonistas & inhibidores , Animales , Estimulantes del Sistema Nervioso Central/farmacología , Cocaína/farmacología , Cuerpo Estriado/metabolismo , Dinorfinas/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Expresión Génica/genética , Hibridación in Situ , Masculino , Metanfetamina/farmacología , Antagonistas de Narcóticos/farmacología , Precursores de Proteínas/genética , Proteínas Proto-Oncogénicas c-fos/genética , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Receptores Opioides mu/análisis , Receptores Opioides mu/fisiología , Factores de Tiempo
20.
Neurotox Res ; 29(4): 569-82, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26846719

RESUMEN

Abused amphetamines, such as d-amphetamine (AMPH) and methamphetamine (METH), are highly addictive and destructive to health and productive lifestyles. The abuse of these drugs is associated with impulsive behavior, which is likely to contribute to addiction. The amphetamines also differentially damage dopamine (DA) and serotonin (5-HT) systems, which regulate impulsive behavior; therefore, exposure to these drugs may differentially alter impulsive behavior to effect the progression of addiction. We examined the impact of neurotoxicity induced by three amphetamines on impulsive action using a stop-signal task in rats. Animals were rewarded with a food pellet after lever pressing (i.e., a go trial), unless an auditory cue was presented and withholding lever press gained reward (i.e., a stop trial). Animals were trained on the task and then exposed to a neurotoxic regimen of either AMPH, p-chloroamphetamine (PCA), or METH. These regimens preferentially reduced DA transporter levels in striatum, 5-HT transporter levels in prefrontal cortex, or both, respectively. Assessment of performance on the stop-signal task beginning 1 week after the treatment revealed that AMPH produced a deficit in go-trial performance, whereas PCA did not alter performance on either trial type. In contrast, METH produced a deficit in stop-trial performance (i.e., impulsive action) but not go-trial performance. These findings suggest that the different neurotoxic consequences of substituted amphetamines are associated with different effects on inhibitory control over behavior. Thus, the course of addiction and maladaptive behavior resulting from exposure to these substances is likely to differ.


Asunto(s)
Encéfalo/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/toxicidad , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Conducta Impulsiva/efectos de los fármacos , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/fisiopatología , Animales , Temperatura Corporal/efectos de los fármacos , Encéfalo/metabolismo , Dextroanfetamina , Modelos Animales de Enfermedad , Dopamina/metabolismo , Masculino , Metanfetamina , Actividad Motora/efectos de los fármacos , Pruebas Neuropsicológicas , Ratas , Ratas Sprague-Dawley , Serotonina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática , Factores de Tiempo , p-Cloroanfetamina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA