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.
Nat Commun ; 14(1): 5915, 2023 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-37739964

RESUMEN

The development of genetically encoded dopamine sensors such as dLight has provided a new approach to measuring slow and fast dopamine dynamics both in brain slices and in vivo, possibly enabling dopamine measurements in areas like the dorsolateral striatum (DLS) where previously such recordings with fast-scan cyclic voltammetry (FSCV) were difficult. To test this, we first evaluated dLight photometry in mouse brain slices with simultaneous FSCV and found that both techniques yielded comparable results, but notable differences in responses to dopamine transporter inhibitors, including cocaine. We then used in vivo fiber photometry with dLight in mice to examine responses to cocaine in DLS. We also compared dopamine responses during Pavlovian conditioning across the striatum. We show that dopamine increases were readily detectable in DLS and describe transient dopamine kinetics, as well as slowly developing signals during conditioning. Overall, our findings indicate that dLight photometry is well suited to measuring dopamine dynamics in DLS.


Asunto(s)
Cocaína , Dopamina , Animales , Ratones , Cuerpo Estriado , Neostriado , Encéfalo , Cocaína/farmacología , Colorantes
2.
Front Neurosci ; 17: 1160185, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37260840

RESUMEN

Fetal Alcohol Spectrum Disorder (FASD) comprises a group of neurodevelopmental deficits caused by alcohol exposure during pregnancy. Clinical studies suggest that while the male progeny experiences serious neurodevelopmental defects, female patients have more severe cognitive, social, and affective symptoms. Other than sex, dose, frequency, and timing of exposure determine the neurobehavioral outcomes in young and adult progeny. In this regard, human studies indicate that some individuals relapse during late-term gestational periods. In mice, this interval corresponds to the first 10 days after birth (postnatal, P0-P10). In our model of postnatal ethanol exposure (PEEP0-P10), we tested whether adult female and male offspring show deficits in sociability, anxiety-like, reward consumption, and action-outcome associations. We report that female PEEP0-P10 offspring have mild social impairments and altered extinction of operant responding in the absence of anxiety-like traits and reward consumption defects. None of these deficits were detected in the male PEEP0-P10 offspring. Our data provide novel information on sex-specific neurobehavioral outcomes of postnatal ethanol exposure in female adult offspring.

3.
Neuropsychopharmacology ; 48(12): 1808-1820, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37188849

RESUMEN

Fetal alcohol exposure has deleterious consequences on the motor skills of patients affected by Fetal Alcohol Spectrum Disorder (FASD) and in pre-clinical models of gestational ethanol exposure (GEE). Deficits in striatal cholinergic interneurons (CINs) and dopamine function impair action learning and execution, yet the effects of GEE on acetylcholine (ACh) and striatal dopamine release remain unexplored. Here, we report that alcohol exposure during the first ten postnatal days (GEEP0-P10), which mimics ethanol consumption during the last gestational trimester in humans, induces sex-specific anatomical and motor skill deficits in female mice during adulthood. Consistent with these behavioral impairments, we observed increased stimulus evoked-dopamine levels in the dorsolateral striatum (DLS) of GEEP0-P10 female, but not male, mice. Further experiments revealed sex-specific deficits in ß2-containing nicotinic ACh receptor (nAChR)-modulation of electrically evoked dopamine release. Moreover, we found a reduced decay of ACh transients and a decreased excitability of striatal CINs in DLS of GEEP0-P10 females, indicating striatal CIN dysfunctions. Finally, the administration of varenicline, a ß2-containing nAChR partial agonist, and chemogenetic-mediated increase in CIN activity improved motor performance in adult GEEP0-P10 females. Altogether, these data shed new light on GEE-induced striatal deficits and establish potential pharmacological and circuit-specific interventions to ameliorate motor symptoms of FASD.


Asunto(s)
Dopamina , Trastornos del Espectro Alcohólico Fetal , Humanos , Masculino , Embarazo , Ratones , Femenino , Animales , Adulto , Dopamina/farmacología , Acetilcolina/farmacología , Destreza Motora , Etanol/farmacología , Cuerpo Estriado , Agonistas Nicotínicos/farmacología
4.
Int J Mol Sci ; 23(19)2022 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-36232321

RESUMEN

Alcohol use disorder (AUD) is characterized by escalating alcohol consumption, preoccupation with alcohol, and continued alcohol consumption despite adverse consequences. Dopamine has been implicated in neural and behavioral processes involved in reward and reinforcement and is a critical neurotransmitter in AUD. Clinical and preclinical research has shown that long-term ethanol exposure can alter dopamine release, though most of this work has focused on nucleus accumbens (NAc). Like the NAc, the dorsal striatum (DS) is implicated in neural and behavioral processes in AUD. However, little work has examined chronic ethanol effects on DS dopamine dynamics. Therefore, we examined the effect of ethanol consumption and withdrawal on dopamine release and its presynaptic regulation with fast-scan cyclic voltammetry in C57BL/6J mice. We found that one month of ethanol consumption did not alter maximal dopamine release or dopamine tissue content. However, we did find that D2 dopamine autoreceptors were sensitized. We also found a decrease in cholinergic control of dopamine release via ß2-containing nAChRs on dopamine axons. Interestingly, both effects were reversed following withdrawal, raising the possibility that some of the neuroadaptations in AUD might be reversible in abstinence. Altogether, this work elucidates some of the chronic alcohol-induced neurobiological dysfunctions in the dopamine system.


Asunto(s)
Autorreceptores , Dopamina , Consumo de Bebidas Alcohólicas , Animales , Colinérgicos/farmacología , Dopamina/farmacología , Etanol/farmacología , Ratones , Ratones Endogámicos C57BL , Núcleo Accumbens
5.
Neuropsychopharmacology ; 47(8): 1537-1549, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35478010

RESUMEN

Withdrawal symptoms are observed upon cessation of cannabis use in humans. Although animal studies have examined withdrawal symptoms following exposure to delta-9-tetrahydrocannabinol (THC), difficulties in obtaining objective measures of spontaneous withdrawal using paradigms that mimic cessation of use in humans have slowed research. The neuromodulator dopamine (DA) is affected by chronic THC treatment and plays a role in many behaviors related to human THC withdrawal symptoms. These symptoms include sleep disturbances that often drive relapse, and emotional behaviors like irritability and anhedonia. We examined THC withdrawal-induced changes in striatal DA release and the extent to which sleep disruption and behavioral maladaptation manifest during abstinence in a mouse model of chronic THC exposure. Using a THC treatment regimen known to produce tolerance, we measured electrically elicited DA release in acute brain slices from different striatal subregions during early and late THC abstinence. Long-term polysomnographic recordings from mice were used to assess vigilance state and sleep architecture before, during, and after THC treatment. We additionally assessed how behaviors that model human withdrawal symptoms are altered by chronic THC treatment in early and late abstinence. We detected altered striatal DA release, sleep disturbances that mimic clinical observations, and behavioral maladaptation in mice following tolerance to THC. Altered striatal DA release, sleep, and affect-related behaviors associated with spontaneous THC abstinence were more consistently observed in male mice. These findings provide a foundation for preclinical study of directly translatable non-precipitated THC withdrawal symptoms and the neural mechanisms that affect them.


Asunto(s)
Dronabinol , Síndrome de Abstinencia a Sustancias , Animales , Agonistas de Receptores de Cannabinoides , Dopamina , Dronabinol/farmacología , Femenino , Masculino , Ratones , Sueño , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico
6.
Small ; 18(16): e2106570, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35263020

RESUMEN

Manganese ferrite nanoparticles display interesting features in bioimaging and catalytic therapies. They have been recently used in theranostics as contrast agents in magnetic resonance imaging (MRI), and as catalase-mimicking nanozymes for hypoxia alleviation. These promising applications encourage the development of novel synthetic procedures to enhance the bioimaging and catalytic properties of these nanomaterials simultaneously. Herein, a cost-efficient synthetic microwave method is developed to manufacture ultrasmall manganese ferrite nanoparticles as advanced multimodal contrast agents in MRI and positron emission tomography (PET), and improved nanozymes. Such a synthetic method allows doping ferrites with Mn in a wide stoichiometric range (Mnx Fe3-x O4 , 0.1 ≤ x ≤ 2.4), affording a library of nanoparticles with different magnetic relaxivities and catalytic properties. These tuned magnetic properties give rise to either positive or dual-mode MRI contrast agents. On the other hand, higher levels of Mn doping enhance the catalytic efficiency of the resulting nanozymes. Finally, through their intracellular catalase-mimicking activity, these ultrasmall manganese ferrite nanoparticles induce an unprecedented tumor growth inhibition in a breast cancer murine model. All of these results show the robust characteristics of these nanoparticles for nanobiotechnological applications.


Asunto(s)
Medios de Contraste , Nanopartículas , Animales , Catalasa , Compuestos Férricos , Imagen por Resonancia Magnética/métodos , Compuestos de Manganeso , Ratones
7.
Nat Rev Neurosci ; 23(4): 191-203, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35228740

RESUMEN

Central nervous system neurons communicate via fast synaptic transmission mediated by ligand-gated ion channel (LGIC) receptors and slower neuromodulation mediated by G protein-coupled receptors (GPCRs). These receptors influence many neuronal functions, including presynaptic neurotransmitter release. Presynaptic LGIC and GPCR activation by locally released neurotransmitters influences neuronal communication in ways that modify effects of somatic action potentials. Although much is known about presynaptic receptors and their mechanisms of action, less is known about when and where these receptor actions alter release, especially in vivo. This Review focuses on emerging evidence for important local presynaptic receptor actions and ideas for future studies in this area.


Asunto(s)
Comunicación Celular , Receptores Presinapticos , Potenciales de Acción , Humanos , Neuronas , Transmisión Sináptica
8.
Neuropsychopharmacology ; 46(8): 1432-1441, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33452430

RESUMEN

The dorsal striatum (DS) is implicated in behavioral and neural processes including action control and reinforcement. Alcohol alters these processes in rodents, and it is believed that the development of alcohol use disorder involves changes in DS dopamine signaling. In nonhuman primates, the DS can be divided into caudate and putamen subregions. As part of a collaborative effort examining the effects of long-term alcohol self-administration in rhesus macaques, we examined DS dopamine signaling using fast-scan cyclic voltammetry. We found that chronic alcohol self-administration resulted in several dopamine system adaptations. Most notably, dopamine release was altered in a sex- and region-dependent manner. Following long-term alcohol consumption, male macaques, regardless of abstinence status, had reduced dopamine release in putamen, while only male macaques in abstinence had reduced dopamine release in caudate. In contrast, female macaques had enhanced dopamine release in the caudate, but not putamen. Dopamine uptake was also enhanced in females, but not males (regardless of abstinence state). We also found that dopamine D2/3 autoreceptor function was reduced in male, but not female, alcohol drinkers relative to control groups. Finally, we found that blockade of nicotinic acetylcholine receptors inhibited evoked dopamine release in nonhuman primates. Altogether, our findings demonstrate that long-term alcohol consumption can sex-dependently alter dopamine release, as well as its feedback control mechanisms in both DS subregions.


Asunto(s)
Dopamina , Receptores de Dopamina D2 , Consumo de Bebidas Alcohólicas , Animales , Cuerpo Estriado/metabolismo , Femenino , Macaca mulatta/metabolismo , Masculino , Putamen/metabolismo , Receptores de Dopamina D2/metabolismo
9.
Neuropsychopharmacology ; 45(9): 1454-1462, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31995814

RESUMEN

Dorsal striatal manipulations including stimulation of dopamine release and activation of medium spiny neurons (MSNs) are sufficient to drive reinforcement-based learning. Glutamatergic innervation of the striatum by the cortex and thalamus is a critical determinant of MSN activity and local regulation of dopamine release. However, the relationship between striatal glutamatergic afferents and behavioral reinforcement is not well understood. We evaluated the reinforcing properties of optogenetic stimulation of thalamostriatal terminals, which are associated with vesicular glutamate transporter 2 (Vglut2) expression, in the dorsomedial striatum (DMS), a region implicated in goal-directed behaviors. In mice expressing channelrhodopsin-2 (ChR2) under control of the Vglut2 promoter, optical stimulation of the DMS reinforced operant lever-pressing behavior. Mice also acquired operant self-stimulation of thalamostriatal terminals when ChR2 expression was virally targeted to the intralaminar thalamus. Stimulation trains that supported operant responding evoked dopamine release in the DMS and excitatory postsynaptic currents in DMS MSNs. Our previous work demonstrated that the presynaptic G protein-coupled receptor metabotropic glutamate receptor 2 (mGlu2) robustly inhibits glutamate and dopamine release induced by activation of thalamostriatal afferents. Thus, we examined the regulation of thalamostriatal self-stimulation by mGlu2. Administration of an mGlu2/3 agonist or an mGlu2-selective positive allosteric modulator reduced self-stimulation. Conversely, blockade of these receptors increased thalamostriatal self-stimulation, suggesting that endogenous activation of these receptors negatively modulates the reinforcing properties of thalamostriatal activity. These findings demonstrate that stimulation of thalamic terminals in the DMS is sufficient to reinforce a self-initiated action, and that thalamostriatal reinforcement is constrained by mGlu2 activation.


Asunto(s)
Cuerpo Estriado , Receptores de Glutamato Metabotrópico , Animales , Cuerpo Estriado/metabolismo , Ratones , Tálamo/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo
10.
Eur J Neurosci ; 50(3): 2023-2035, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30218623

RESUMEN

There is a compelling evidence that midbrain dopamine (DA) neurons and their projections to the ventral striatum provide a mechanism for motivating reward-seeking behavior, and for utilizing information about unexpected reward prediction errors (RPEs) to guide behavior based on current, rather than historical, outcomes. When this mechanism is compromised in addictions, it may produce patterns of maladaptive behavior that remain obdurate in the face of contrary information and even adverse consequences. Nonetheless, DAergic contributions to performance on behavioral tasks that rely on the ability to flexibly update stimulus-reward relationships remains incompletly understood. In the current study, we used a discrimination and reversal paradigm to monitor subsecond DA release in mouse NAc core (NAc) using in vivo fast-scan cyclic voltammetry (FSCV). We observed post-choice elevations in phasic NAc DA release; however, increased DA transients were only evident during early reversal when mice made responses at the newly rewarded stimulus. Based on this finding, we used in vivo optogenetic (eNpHR) photosilencing and (Channelrhodopsin2 [ChR2]) photostimulation to assess the effects of manipulating VTA-DAergic fibers in the NAc on reversal performance. Photosilencing the VTA â†’ NAc DAergic pathway during early reversal increased errors, while photostimulation did not demonstrably affect behavior. Taken together, these data provide additional evidence of the importance of NAc DA release as a neural substrate supporting adjustments in learned behavior after a switch in expected stimulus-reward contingencies. These findings have possible implications for furthering understanding the role of DA in persistent, maladaptive decision-making characterizing addictions.


Asunto(s)
Cognición/fisiología , Condicionamiento Operante/fisiología , Núcleo Accumbens/fisiología , Recompensa , Animales , Neuronas Dopaminérgicas/metabolismo , Masculino , Ratones Endogámicos C57BL , Motivación/fisiología , Área Tegmental Ventral/fisiología
11.
Neuropsychopharmacology ; 43(10): 2056-2063, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29925886

RESUMEN

Prominent motor deficits (e.g., chorea) that typify Huntington's disease (HD) arise following a prolonged prodromal stage characterized by psychiatric disturbances. Apathy, a disorder of motivation characterized by diminished goal-directed behavior, is one of the earliest and most common psychiatric symptoms in HD, but the underlying neurobiology is unclear and treatment options are limited. Alterations in the endocannabinoid (eCB) and dopamine systems represent prominent pathophysiological markers in HD that-similar to motivational deficits-present early and decline across disease progression. Whether changes in dopamine and eCB systems are associated with specific behavioral impairments in HD and whether these deficits are amenable to viable treatments is unknown. Here, we show that dopaminergic encoding of effortful drive progressively declines with age in an HD mouse model, and is restored by elevating tissue levels of the eCB 2-arachidonoylglycerol (2-AG) through targeted inhibition of its enzymatic degradation. This work supports aberrant dopaminergic encoding of reward as a neurobiological correlate of apathy in HD, and indicates that cannabinoid receptor-based therapies may benefit neuropsychiatric care for HD.


Asunto(s)
Dopamina/metabolismo , Endocannabinoides/metabolismo , Enfermedad de Huntington/psicología , Motivación/efectos de los fármacos , Envejecimiento/psicología , Animales , Apatía , Ácidos Araquidónicos/farmacología , Benzodioxoles/farmacología , Condicionamiento Operante/efectos de los fármacos , Progresión de la Enfermedad , Endocannabinoides/agonistas , Endocannabinoides/farmacología , Glicéridos/farmacología , Masculino , Ratones , Piperidinas/farmacología , Pirazoles/farmacología , Recompensa
12.
Neuropsychopharmacology ; 43(6): 1445-1456, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29362511

RESUMEN

Whereas cortical GAD67 reduction and subsequent GABA level decrease are consistently observed in schizophrenia and depression, it remains unclear how these GABAergic abnormalities contribute to specific symptoms. We modeled cortical GAD67 reduction in mice, in which the Gad1 gene is genetically ablated from ~50% of cortical and hippocampal interneurons. Mutant mice showed a reduction of tissue GABA in the hippocampus and cortex including mPFC, and exhibited a cluster of effort-based behavior deficits including decreased home-cage wheel running and increased immobility in both tail suspension and forced swim tests. Since saccharine preference, progressive ratio responding to food, and learned helplessness task were normal, such avolition-like behavior could not be explained by anhedonia or behavioral despair. In line with the prevailing view that dopamine in anterior cingulate cortex (ACC) plays a role in evaluating effort cost for engaging in actions, we found that tail-suspension triggered dopamine release in ACC of controls, which was severely attenuated in the mutant mice. Conversely, ACC dopamine release by progressive ratio responding to reward, during which animals were allowed to effortlessly perform the nose-poking, was not affected in mutants. These results suggest that cortical GABA reduction preferentially impairs the effort-based behavior which requires much effort with little benefit, through a deficit of ACC dopamine release triggered by high-effort cost behavior, but not by reward-seeking behavior. Collectively, a subset of negative symptoms with a reduced willingness to expend costly effort, often observed in patients with schizophrenia and depression, may be attributed to cortical GABA level reduction.


Asunto(s)
Corteza Cerebral/metabolismo , Glutamato Descarboxilasa/deficiencia , Hipocampo/metabolismo , Interneuronas/metabolismo , Motivación/fisiología , Ácido gamma-Aminobutírico/deficiencia , Animales , Reacción de Prevención/fisiología , Epilepsia/metabolismo , Femenino , Glutamato Descarboxilasa/genética , Masculino , Ratones Noqueados , Actividad Motora/fisiología , Fenotipo , Recompensa , Conducta Sexual Animal/fisiología , Conducta Social , Transmisión Sináptica/fisiología , Técnicas de Cultivo de Tejidos
13.
Neuron ; 96(5): 1112-1126.e5, 2017 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-29216450

RESUMEN

Dopamine (DA) transmission mediates numerous aspects of behavior. Although DA release is strongly linked to firing of DA neurons, recent developments indicate the importance of presynaptic modulation at striatal dopaminergic terminals. The endocannabinoid (eCB) system regulates DA release and is a canonical gatekeeper of goal-directed behavior. Here we report that extracellular DA increases induced by selective optogenetic activation of cholinergic neurons in the nucleus accumbens (NAc) are inhibited by CB1 agonists and eCBs. This modulation requires CB1 receptors on cortical glutamatergic afferents. Dopamine increases driven by optogenetic activation of prefrontal cortex (PFC) terminals in the NAc are similarly modulated by activation of these CB1 receptors. We further demonstrate that this same population of CB1 receptors modulates optical self-stimulation sustained by activation of PFC afferents in the NAc. These results establish local eCB actions on PFC terminals within the NAc that inhibit mesolimbic DA release and constrain reward-driven behavior.


Asunto(s)
Dopamina/metabolismo , Endocannabinoides/farmacología , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Corteza Prefrontal/efectos de los fármacos , Terminales Presinápticos/efectos de los fármacos , Terminales Presinápticos/metabolismo , Animales , Fenómenos Electrofisiológicos/efectos de los fármacos , Espacio Extracelular/efectos de los fármacos , Espacio Extracelular/metabolismo , Glutamatos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Núcleo Accumbens/citología , Corteza Prefrontal/citología , Receptor Cannabinoide CB1/agonistas , Recompensa , Autoestimulación , Transmisión Sináptica/efectos de los fármacos
14.
Neuropharmacology ; 124: 52-61, 2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28450060

RESUMEN

Dopamine (DA) is a major catecholamine neurotransmitter in the mammalian brain that controls neural circuits involved in the cognitive, emotional, and motor aspects of goal-directed behavior. Accordingly, perturbations in DA neurotransmission play a central role in several neuropsychiatric disorders. Somewhat surprisingly given its prominent role in numerous behaviors, DA is released by a relatively small number of densely packed neurons originating in the midbrain. The dopaminergic midbrain innervates numerous brain regions where extracellular DA release and receptor binding promote short- and long-term changes in postsynaptic neuron function. Striatal forebrain nuclei receive the greatest proportion of DA projections and are a predominant hub at which DA influences behavior. A number of excitatory, inhibitory, and modulatory inputs orchestrate DA neurotransmission by controlling DA cell body firing patterns, terminal release, and effects on postsynaptic sites in the striatum. The endocannabinoid (eCB) system serves as an important filter of afferent input that acts locally at midbrain and terminal regions to shape how incoming information is conveyed onto DA neurons and to output targets. In this review, we aim to highlight existing knowledge regarding how eCB signaling controls DA neuron function through modifications in synaptic strength at midbrain and striatal sites, and to raise outstanding questions on this topic. This article is part of the Special Issue entitled "A New Dawn in Cannabinoid Neurobiology".


Asunto(s)
Neuronas Dopaminérgicas/fisiología , Endocannabinoides/fisiología , Transmisión Sináptica/fisiología , Animales , Cuerpo Estriado/fisiología , Humanos , Mesencéfalo/fisiología
15.
Neuropharmacology ; 117: 114-123, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28159646

RESUMEN

The striatum plays critical roles in action control and cognition, and activity of striatal neurons is driven by glutamatergic input. Inhibition of glutamatergic inputs to projection neurons and interneurons of the striatum by presynaptic G protein-coupled receptors (GPCRs) stands to modulate striatal output and striatum-dependent behaviors. Despite knowledge that a substantial number of glutamatergic inputs to striatal neurons originate in the thalamus, most electrophysiological studies assessing GPCR modulation do not differentiate between effects on corticostriatal and thalamostriatal transmission, and synaptic inhibition is frequently assumed to be mediated by activation of GPCRs on corticostriatal terminals. We used optogenetic techniques and recently-discovered pharmacological tools to dissect the effects of a prominent presynaptic GPCR, metabotropic glutamate receptor 2 (mGlu2), on corticostriatal vs. thalamostriatal transmission. We found that an agonist of mGlu2 and mGlu3 induces long-term depression (LTD) at synapses onto MSNs from both the cortex and the thalamus. Thalamostriatal LTD is selectively blocked by an mGlu2-selective negative allosteric modulator and reversed by application of an antagonist following LTD induction. Activation of mGlu2/3 also induces LTD of thalamostriatal transmission in striatal cholinergic interneurons (CINs), and pharmacological activation of mGlu2/3 or selective activation of mGlu2 inhibits CIN-mediated dopamine release evoked by selective stimulation of thalamostriatal inputs. Thus, mGlu2 activation exerts effects on striatal physiology that extend beyond modulation of corticostriatal synapses, and has the potential to influence cognition and striatum-related disorders via inhibition of thalamus-derived glutamate and dopamine release.


Asunto(s)
Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Ácido Glutámico/metabolismo , Receptores de Glutamato Metabotrópico/fisiología , Tálamo/fisiología , Animales , Neuronas Colinérgicas/fisiología , Interneuronas/fisiología , Depresión Sináptica a Largo Plazo/efectos de los fármacos , Masculino , Ratones , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores
16.
Neuropharmacology ; 108: 275-83, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27036891

RESUMEN

The striatum is typically classified according to its major output pathways, which consist of dopamine D1 and D2 receptor-expressing neurons. The striatum is also divided into striosome and matrix compartments, based on the differential expression of a number of proteins, including the mu opioid receptor, dopamine transporter (DAT), and Nr4a1 (nuclear receptor subfamily 4, group A, member 1). Numerous functional differences between the striosome and matrix compartments are implicated in dopamine-related neurological disorders including Parkinson's disease and addiction. Using Nr4a1-eGFP mice, we provide evidence that electrically evoked dopamine release differs between the striosome and matrix compartments in a regionally-distinct manner. We further demonstrate that this difference is not due to differences in inhibition of dopamine release by dopamine autoreceptors or nicotinic acetylcholine receptors. Furthermore, cocaine enhanced extracellular dopamine in striosomes to a greater degree than in the matrix and concomitantly inhibited dopamine uptake in the matrix to a greater degree than in striosomes. Importantly, these compartment differences in cocaine sensitivity were limited to the dorsal striatum. These findings demonstrate a level of exquisite microanatomical regulation of dopamine by the DAT in striosomes relative to the matrix.


Asunto(s)
Cocaína/farmacología , Cuerpo Estriado/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/biosíntesis , Dopamina/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/biosíntesis , Receptores de Dopamina D2/biosíntesis , Animales , Cuerpo Estriado/efectos de los fármacos , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Técnicas de Cultivo de Órganos
17.
Psychopharmacology (Berl) ; 233(8): 1435-43, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26892380

RESUMEN

RATIONALE: Hypofunction of striatal dopamine neurotransmission, or hypodopaminergia, is a consequence of excessive ethanol use and is hypothesized to be a critical component of alcoholism, driving alcohol intake in an attempt to restore dopamine levels; however, the neurochemical mechanisms involved in these dopaminergic deficiencies are not fully understood. OBJECTIVE: Here we examined the specific dopaminergic adaptations that produce hypodopaminergia and contribute to alcohol use disorders using direct, sub-second measurements of dopamine signaling in nonhuman primates following chronic ethanol self-administration. METHODS: Female rhesus macaques completed 1 year of daily (22 h/day) ethanol self-administration. Subsequently, fast-scan cyclic voltammetry was used in nucleus accumbens core brain slices to determine alterations in dopamine terminal function, including release and uptake kinetics, and sensitivity to quinpirole (D2/D3 dopamine receptor agonist) and U50,488 (kappa opioid receptor agonist) induced inhibition of dopamine release. RESULTS: Ethanol drinking greatly increased uptake rates, which were positively correlated with lifetime ethanol intake. Furthermore, the sensitivity of dopamine D2/D3 autoreceptors and kappa opioid receptors, which both act as negative regulators of presynaptic dopamine release, was moderately and robustly enhanced in ethanol drinkers. CONCLUSIONS: Greater uptake rates and sensitivity to D2-type autoreceptor and kappa opioid receptor agonists could converge to drive a hypodopaminergic state, characterized by reduced basal dopamine and an inability to mount appropriate dopaminergic responses to salient stimuli. Together, we outline the specific alterations to dopamine signaling that may drive ethanol-induced hypofunction of the dopamine system and suggest that the dopamine and dynorphin/kappa opioid receptor systems may be efficacious pharmacotherapeutic targets in the treatment of alcohol use disorders.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Autorreceptores/fisiología , Dopamina/metabolismo , Etanol/administración & dosificación , Núcleo Accumbens/metabolismo , Transmisión Sináptica/fisiología , Animales , Femenino , Macaca mulatta , Núcleo Accumbens/efectos de los fármacos , Quinpirol/farmacología , Receptores de Dopamina D2/agonistas , Receptores de Dopamina D2/metabolismo , Autoadministración , Transmisión Sináptica/efectos de los fármacos
18.
Drug Alcohol Depend ; 158: 159-63, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26627912

RESUMEN

BACKGROUND: Given the high level of homology between nonhuman primates and humans in regard to anatomy, physiology and ethanol drinking patterns, nonhuman primates represent an unparalleled preclinical model for examining the neurobiological basis of ethanol abuse. METHODS: Here we examined the neurochemical consequences of chronic daily ethanol use using fast-scan cyclic voltammetry in brain slices containing the nucleus accumbens core or dorsolateral caudate taken from male cynomolgus macaques following ethanol drinking. RESULTS: We found that in both regions the ability of ethanol to decrease dopamine release was unchanged, indicating that ethanol self-administration does not produce tolerance or sensitization to ethanol effects on dopamine release at the dopamine terminal at this time point. We also found that in the nucleus accumbens core, autoregulation of dopamine release was shifted from equal D2 and D3 receptor involvement in control animals to primarily D2 receptor-mediated in drinkers. Specifically, the effect quinpirole, a D2/D3 receptor agonist, on dopamine release was equal across groups; however, dopamine signals were reversed to a greater extent by the selective D3 receptor antagonist SB-277,011A in control animals, indicating a greater contribution of D2 receptors in quinpirole-induced inhibition following ethanol self-administration. In the dorsolateral caudate, the effects of quinpirole and reversal with SB-277,011A was not different between ethanol and control slices. CONCLUSIONS: This work provides novel insight into the dopaminergic adaptations resulting from chronic ethanol use in nonhuman primates and indicates that alterations in D2/D3 dopamine autoreceptor signaling may be an important neurochemical adaptation to ethanol consumption during early use.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Dopamina/metabolismo , Etanol/administración & dosificación , Retroalimentación Fisiológica/fisiología , Núcleo Accumbens/metabolismo , Receptores de Dopamina D2/metabolismo , Animales , Retroalimentación Fisiológica/efectos de los fármacos , Macaca , Macaca fascicularis , Masculino , Núcleo Accumbens/efectos de los fármacos , Quinpirol/farmacología , Receptores de Dopamina D2/agonistas , Autoadministración
19.
Hum Mol Genet ; 24(18): 5299-312, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26123485

RESUMEN

Preferential dysfunction/degeneration of midbrain substantia nigra pars compacta (SNpc) dopaminergic (DA) neurons contributes to the main movement symptoms manifested in Parkinson's disease (PD). Although the Leucine-rich repeat kinase 2 (LRRK2) G2019S missense mutation (LRRK2 G2019S) is the most common causative genetic factor linked to PD, the effects of LRRK2 G2019S on the function and survival of SNpc DA neurons are poorly understood. Using a binary gene expression system, we generated transgenic mice expressing either wild-type human LRRK2 (WT mice) or the LRRK2 G2019S mutation (G2019S mice) selectively in the midbrain DA neurons. Here we show that overexpression of LRRK2 G2019S did not induce overt motor abnormalities or substantial SNpc DA neuron loss. However, the LRRK2 G2019S mutation impaired dopamine homeostasis and release in aged mice. This reduction in dopamine content/release coincided with the degeneration of DA axon terminals and decreased expression of DA neuron-enriched genes tyrosine hydroxylase (TH), vesicular monoamine transporter 2, dopamine transporter and aldehyde dehydrogenase 1. These factors are responsible for dopamine synthesis, transport and degradation, and their expression is regulated by transcription factor paired-like homeodomain 3 (PITX3). Levels of Pitx3 mRNA and protein were similarly decreased in the SNpc DA neurons of aged G2019S mice. Together, these findings suggest that PITX3-dependent transcription regulation could be one of the many potential mechanisms by which LRRK2 G2019S acts in SNpc DA neurons, resulting in downregulation of its downstream target genes critical for dopamine homeostasis and release.


Asunto(s)
Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Regulación de la Expresión Génica , Mutación Missense , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Factores de Edad , Animales , Conducta Animal , Modelos Animales de Enfermedad , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Mesencéfalo/metabolismo , Mesencéfalo/patología , Ratones , Ratones Transgénicos , Actividad Motora , Degeneración Nerviosa/genética , Enfermedad de Parkinson/patología , Sustancia Negra/metabolismo , Sustancia Negra/patología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
20.
Eur J Neurosci ; 39(4): 548-56, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24236977

RESUMEN

Dopamine (DA) plays an important role in integrative functions contributing to adaptive behaviors. In support of this essential function, DA modulates synaptic plasticity in different brain areas, including the striatum. Many drugs used for cognitive enhancement are psychostimulants, such as methylphenidate (MPH), which enhance DA levels. MPH treatment is of interest during adolescence, a period of enhanced neurodevelopment during which the DA system is in a state of flux. Recent epidemiological studies report the co-abuse of MPH and ethanol in adolescents and young adults. Although repeated MPH treatment produces enduring changes that affect subsequent behavioral responses to other psychostimulants, few studies have investigated the interactions between MPH and ethanol. Here we addressed whether chronic therapeutic exposure to MPH during adolescence predisposed mice to an altered response to ethanol and whether this was accompanied by altered DA release and striatal plasticity. C57BL/6J mice were administered MPH (3-6 mg/kg/day) via the drinking water between post-natal days 30 and 60. Voltammetry experiments showed that sufficient brain MPH concentrations were achieved during adolescence in mice to increase the DA clearance in adulthood. The treatment also increased long-term depression and reduced the effects of ethanol on striatal synaptic responses. Although the injection of 0.4 or 2 g/kg ethanol dose-dependently decreased locomotion in control mice, only the higher dose decreased locomotion in MPH-treated mice. These results suggested that the administration of MPH during development promoted long-term effects on synaptic plasticity in forebrain regions targeted by DA. These changes in plasticity might, in turn, underlie alterations in behaviors controlled by these brain regions into adulthood.


Asunto(s)
Estimulantes del Sistema Nervioso Central/farmacología , Cuerpo Estriado/efectos de los fármacos , Etanol/farmacología , Depresión Sináptica a Largo Plazo , Metilfenidato/farmacología , Sinapsis/efectos de los fármacos , Animales , Cuerpo Estriado/crecimiento & desarrollo , Cuerpo Estriado/fisiología , Locomoción , Masculino , Ratones , Ratones Endogámicos C57BL , Sinapsis/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA