Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 566
Filtrar
1.
Neuropharmacology ; 232: 109527, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37011784

RESUMEN

Parvalbumin-expressing dorsal striatal fast-spiking interneurons, comprising ∼1% of the total dorsal striatal neuronal population, are necessary for the expression of compulsive-like ethanol consumption mice. Fast-spiking interneurons are driven to fire by glutamatergic inputs derived primarily from the cortex. However, these neurons also receive substantial GABAergic input from two sources: the globus pallidus and the reticular nucleus of the thalamus. How ethanol modulates inhibitory input onto fast-spiking neurons is unclear and, more broadly, alcohol effects on GABAergic synaptic transmission onto GABAergic interneurons are understudied. Examining this, we found that acute bath application of ethanol (50 mM) potentiated GABAergic transmission from both the globus pallidus and the reticular nucleus of the thalamus onto fast-spiking interneurons in mouse of both sexes. This ethanol-induced potentiation required postsynaptic calcium and was not accompanied by a sustained change in presynaptic GABA release probability. Examining whether this ethanol effect persisted following chronic intermittent ethanol exposure, we found attenuated acute-ethanol potentiation of GABAergic transmission from both the globus pallidus and the reticular nucleus of the thalamus onto striatal fast-spiking interneurons. These data underscore the impact of ethanol on GABAergic signaling in the dorsal striatum and support the notion that ethanol may disinhibit the dorsolateral striatum.


Asunto(s)
Cuerpo Estriado , Etanol , Neuronas GABAérgicas , Interneuronas , Animales , Femenino , Masculino , Ratones , Cuerpo Estriado/citología , Cuerpo Estriado/efectos de los fármacos , Etanol/administración & dosificación , Etanol/farmacología , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Globo Pálido/citología , Globo Pálido/efectos de los fármacos , Interneuronas/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Núcleos Talámicos/citología , Núcleos Talámicos/efectos de los fármacos , Núcleos Talámicos/metabolismo , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Calcio/metabolismo
2.
Synapse ; 74(5): e22144, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31736138

RESUMEN

Neurosteroids are potent allosteric modulators of GABAA receptors (GABAA Rs). Although the effects of exogenous neurosteroids on GABAA R function are well documented, less is known about effects of neurosteroids produced by local endogenous biosynthesis. The neurosteroidogenic enzymes 5α-reductase and 3α-hydroxysteroid dehydrogenase are expressed in two nuclei of somatosensory thalamus, the thalamic reticular nucleus (nRT) and ventrobasal nucleus (VB). Here, the effects of acute blockade of neurosteroidogenesis by the 5α-reductase inhibitor finasteride on phasic and tonic GABAA R-mediated currents were examined in nRT and VB of mice. In nRT, finasteride altered the decay and amplitude, but not the frequency, of phasic currents, with no effect on tonic inhibition. In VB neurons, by contrast, finasteride reduced both the size and frequency of phasic currents, and also reduced the degree of tonic inhibition. These studies thus provide novel evidence for endogenous modulation of GABAA R function by 5α-reduced neurosteroids in the mature thalamus.


Asunto(s)
Inhibición Neural , Neuroesteroides/metabolismo , Núcleos Talámicos/metabolismo , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/metabolismo , Inhibidores de 5-alfa-Reductasa/farmacología , Animales , Femenino , Finasterida/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de GABA-A/metabolismo , Núcleos Talámicos/efectos de los fármacos , Núcleos Talámicos/fisiología
3.
Neuroimage ; 195: 203-214, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-30946950

RESUMEN

Forepaw somatosensory stimulation induces neural activities in relay thalamic nuclei, the primary somatosensory cortex of forelimb (S1FL), and the secondary somatosensory cortex (S2). However, rodent fMRI studies of somatosensory stimulation have commonly reported BOLD changes only in S1FL, which may be due to side effects of anesthetics and/or the low sensitivity in the thalamus. Thus, we have obtained mouse BOLD fMRI under newly-adopted ketamine-xylazine anesthesia. High-resolution BOLD fMRI obtained with same imaging parameters at 9.4T versus 15.2T shows the improvement of functional detectability by ≥ 2 times at 15.2T due to higher signal intensity and larger BOLD response. The fMRI responses at 15.2T were robustly observed at well-known somatosensory networks including thalamus. Second, echo-time-dependent BOLD signals are dominant based on multi-echo fMRI data. A ratio of BOLD responses in S1FL to thalamus is ∼2, which is not related to different baseline T2∗ or different cerebral blood volume. Third, group-averaged 15.2T BOLD maps show activities in S1FL, S2, motor cortices, and thalamic nuclei, which agree well with neural tracing network data from the Allen Institute, demonstrating that fMRI detects entire somatosensory networks. Our data suggest that ultrahigh field fMRI provides a unique window into understanding functional networks in normal and transgenic mouse models noninvasively.


Asunto(s)
Mapeo Encefálico/métodos , Imagen por Resonancia Magnética/métodos , Núcleos Talámicos/fisiología , Anestésicos/farmacología , Animales , Potenciales Evocados Somatosensoriales/efectos de los fármacos , Potenciales Evocados Somatosensoriales/fisiología , Ketamina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Corteza Somatosensorial/efectos de los fármacos , Corteza Somatosensorial/fisiología , Núcleos Talámicos/efectos de los fármacos , Xilazina/farmacología
4.
Eur J Neurosci ; 50(1): 1779-1798, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30919548

RESUMEN

The mediodorsal (MD) and adjacent intralaminar (IL) and midline nuclei provide the main thalamic input to the medial prefrontal cortex (mPFC) and are critical for associative learning and decision-making. MD neurons exhibit activity related to actions and outcomes that mirror responses of mPFC neurons in rats during dynamic delayed non-match to position (dDNMTP), a variation of DNMTP where start location is varied randomly within an open octagonal arena to avoid confounding behavioral events with spatial location. To test whether the thalamus affects the expression of these responses in mPFC, we inhibited the central thalamus unilaterally by microinjecting muscimol at doses and sites found to affect decision-making when applied bilaterally. Unilateral inactivation reduced normalized task-related responses in the ipsilateral mPFC without disrupting behavior needed to characterize event-related neuronal activity. Our results extend earlier findings that focused on delay-related activity by showing that central thalamic inactivation interferes with responses related to actions and outcomes that occur outside the period of memory delay. These findings are consistent with the broad effects of central thalamic lesions on behavioral measures of reinforcement-guided responding. Most (7/8) of the prefrontal response types affected by thalamic inactivation have also been observed in MD during dDNMTP. These results support the hypothesis that MD and IL act as transthalamic gates: monitoring prefrontal activity through corticothalamic inputs; integrating this information with signals from motivational and sensorimotor systems that converge in thalamus; and acting through thalamocortical projections to enhance expression of neuronal responses in the PFC that support adaptive goal-directed behavior.


Asunto(s)
Toma de Decisiones/fisiología , Objetivos , Neuronas/fisiología , Corteza Prefrontal/fisiología , Desempeño Psicomotor/fisiología , Refuerzo en Psicología , Núcleos Talámicos/fisiología , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Toma de Decisiones/efectos de los fármacos , Masculino , Muscimol/farmacología , Neuronas/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Desempeño Psicomotor/efectos de los fármacos , Ratas , Ratas Long-Evans , Memoria Espacial/efectos de los fármacos , Memoria Espacial/fisiología , Núcleos Talámicos/efectos de los fármacos
5.
Neuroscience ; 406: 626-636, 2019 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-30825581

RESUMEN

The medial prefrontal cortex (mPFC) has been implicated in novelty detection and attention. We studied the effect of mPFC electrical stimulation on whisker responses recorded in the ventroposterior medial thalamic nucleus (VPM), the posterior thalamic nucleus (POm) and the primary somatosensory (S1) cortex in urethane anesthetized rats. Field potentials and unit recordings were performed in the VPM or POm thalamic nuclei, in S1 cortex, and in the Zona Incerta (ZI). Somatosensory evoked potentials were elicited by whisker deflections. Current pulses were delivered by bipolar stimulating electrodes aimed at the prelimbic (PL) or infralimbic (IL) areas of mPFC. PL train stimulation (50 Hz, 500 ms) induced a facilitation of whisker responses in the VPM nucleus that lasted minutes and a short inhibition in the POm nucleus. IL stimulation induced a facilitation of whisker responses in both VPM and POm nuclei. Facilitation was due to corticofugal projections because it was reduced after S1 cortical inactivation with lidocaine, and by activation of NMDA glutamatergic receptors because it was blocked by APV. Paired stimulation of mPFC and whiskers revealed an inhibitory effect at short intervals (<100 ms), which was mediated by ZI inhibitory neurons since PL stimulation induced response facilitation in the majority of ZI neurons (42%) and muscimol injection into ZI nucleus reduced inhibitory effects, suggesting that the mPFC may inhibit the POm neurons by activation of GABAergic ZI neurons. In conclusion, the mPFC may control the flow of somatosensory information through the thalamus by activation of S1 and ZI neurons.


Asunto(s)
Estimulación Física , Corteza Prefrontal/fisiopatología , Corteza Somatosensorial/fisiopatología , Tálamo/fisiopatología , Vibrisas/fisiología , Animales , Estimulación Eléctrica/métodos , Potenciales Evocados Somatosensoriales/efectos de los fármacos , Masculino , Muscimol/farmacología , Corteza Prefrontal/efectos de los fármacos , Ratas Sprague-Dawley , Corteza Somatosensorial/efectos de los fármacos , Núcleos Talámicos/efectos de los fármacos , Núcleos Talámicos/fisiopatología , Tálamo/efectos de los fármacos , Vibrisas/efectos de los fármacos , Zona Incerta/efectos de los fármacos , Zona Incerta/fisiopatología
6.
Eur J Neurosci ; 50(4): 2683-2693, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30803080

RESUMEN

Orexins/hypocretins are hypothalamic neuropeptides that have a variety of functions, including maintenance of arousal, control over the sleep/wake cycle, reward and feeding. Accumulating evidence links orexins to the time-keeping system with a documented action in the master clock-the suprachiasmatic nucleus. The intergeniculate leaflet (IGL) is a thalamic structure with the well-known function of collecting photic and non-photic cues to adjust the rhythm of the suprachiasmatic nucleus to changing environmental conditions. The IGL consists of GABAergic neurons that are intrinsically active, even in slice preparations. Our previous studies revealed the excitatory postsynaptic effects of orexins on single IGL neurons, even though the ionic mechanism underlying this effect remained elusive. Therefore, in this study, we used patch clamp electrophysiology to identify the ions and distinct ion channels responsible for the observed depolarisations. The major finding of this article is that the orexin A-evoked depolarisation of IGL neurons depends on non-selective cation channels, implicating the orexinergic tone in establishing the basal firing rate in these cells. The data presented here strengthen the mutual connections between the time-keeping and orexinergic systems.


Asunto(s)
Cuerpos Geniculados/efectos de los fármacos , Canales Iónicos/efectos de los fármacos , Neuronas/efectos de los fármacos , Orexinas/farmacología , Núcleos Talámicos/efectos de los fármacos , Animales , Fenómenos Electrofisiológicos/efectos de los fármacos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Cuerpos Geniculados/citología , Masculino , Técnicas de Placa-Clamp , Potasio/fisiología , Ratas , Ratas Wistar , Sodio/fisiología , Núcleo Supraquiasmático/efectos de los fármacos , Núcleos Talámicos/citología , Ácido gamma-Aminobutírico/fisiología
7.
Neuropharmacology ; 137: 13-23, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29702122

RESUMEN

BACKGROUND: Sub-anesthetic doses of the non-competitive N-methyl-d-aspartate receptor (NMDA-R) antagonist ketamine evoke transient psychotomimetic effects, followed by persistent antidepressant effects in treatment-resistant depressed patients and rodents through still poorly understood mechanisms. Since phencyclidine (PCP) disinhibits thalamo-cortical networks by blocking NMDA-Rs on GABAergic neurons of the reticular thalamic nucleus (RtN), we examined ketamine's actions in the same areas. METHODS: Single units and local field potentials were recorded in chloral hydrate anesthetized male Wistar rats. The effects of cumulative ketamine doses (0.25-5 mg/kg, i.v.) on neuronal discharge and oscillatory activity were examined in RtN, mediodorsal and centromedial (MD/CM) thalamic nuclei, and layer VI of the medial prefrontal cortex (mPFC). RESULTS: Ketamine (1, 2 and 5 mg/kg, i.v.) significantly decreased the discharge of MD/CM, RtN and layer VI mPFC pyramidal neurons. Simultaneously, ketamine decreased the power of low frequency oscillations in all areas examined and increased gamma oscillations in mPFC and MD/CM. Lower ketamine doses (0.25 and 0.5 mg/kg, i.v.) were ineffective. CONCLUSIONS: As observed for PCP, ketamine markedly inhibited the activity of RtN neurons. However, unlike PCP, this effect did not translate into a disinhibition of MD/CM and mPFC excitatory neurons, possibly due to a more potent and simultaneous blockade of NMDA-Rs by ketamine in MD/CM and mPFC neurons. Hence, the present in vivo results show that ketamine evokes an early transient inhibition of neuronal discharge in thalamo-cortical networks, following its rapid pharmacokinetics, which is likely associated to its psychotomimetic effects. The prolonged increase in gamma oscillations may underlie its antidepressant action.


Asunto(s)
Antagonistas de Aminoácidos Excitadores/farmacología , Ritmo Gamma/efectos de los fármacos , Ketamina/farmacología , Neuronas/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Núcleos Talámicos/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Antagonistas de Aminoácidos Excitadores/sangre , Ritmo Gamma/fisiología , Ketamina/sangre , Masculino , Inhibición Neural/efectos de los fármacos , Neuronas/fisiología , Fenciclidina/farmacología , Corteza Prefrontal/fisiología , Ratas Wistar , Núcleos Talámicos/fisiología
8.
Eur Neuropsychopharmacol ; 27(12): 1248-1257, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29128144

RESUMEN

Brexpiprazole (BREX), a recently approved antipsychotic drug in the US and Canada, improves cognitive dysfunction in animal models, by still largely unknown mechanisms. BREX is a partial agonist at 5-HT1A and D2 receptors and antagonist at α1B- and α2C-adrenergic and 5-HT2A receptors all with a similar potency. The NMDA receptor antagonist phencyclidine (PCP), used as pharmacological model of schizophrenia, activates thalamocortical networks and decreases low frequency oscillations (LFO; <4 Hz). These effects are reversed by antipsychotics. Here we assessed the ability of BREX to reverse PCP-induced hyperactivity of thalamocortical circuits, and the involvement of 5-HT1A receptors in its therapeutic action. BREX reversed PCP-induced neuronal activation at a lower dose in centromedial/mediodorsal thalamic nuclei (CM/MD; 0.5mg/kg) than in pyramidal medial prefrontal cortex neurons (mPFC, 2mg/kg), perhaps due to antagonism at α1B-adrenoceptors, abundantly expressed in the thalamus. Conversely, a cumulative 0.5 mg/kg dose reversed a PCP-induced LFO decrease in mPFC but not in CM/MD. BREX reduced LFO in both areas, yet with a different dose-response, and moderately excited mPFC neurons. The latter effect was reversed by the 5-HT1A receptor antagonist WAY-100635. Thus, BREX partly antagonizes PCP-induced thalamocortical hyperactivity, differentially in mPFC versus CM/MD. This regional selectivity may be related to the differential expression of α1B-, α2C-adrenergic and 5-HT2A receptors in both regions and/or different neuronal types. Furthermore, the pro-cognitive properties of BREX may be related to the 5-HT1A receptor-mediated increase in mPFC pyramidal neuron activity. Overall, the present data provide new insight on the brain elements involved in BREX's therapeutic actions.


Asunto(s)
Antipsicóticos/farmacología , Vías Nerviosas/efectos de los fármacos , Neuronas/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Quinolonas/farmacología , Núcleos Talámicos/efectos de los fármacos , Tiofenos/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Ondas Encefálicas/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Antagonistas de Aminoácidos Excitadores/farmacología , Análisis de Fourier , Masculino , Vías Nerviosas/fisiología , Fenciclidina/farmacología , Piperazinas/farmacología , Corteza Prefrontal/citología , Piridinas/farmacología , Ratas , Ratas Wistar , Antagonistas de la Serotonina/farmacología , Núcleos Talámicos/citología
9.
Eur J Neurosci ; 46(3): 1850-1862, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28664636

RESUMEN

Drug addiction is a chronic disease that is shaped by alterations in neuronal function within the cortical-basal ganglia-thalamic circuit. However, our understanding of how this circuit regulates drug-seeking remains incomplete, and relapse rates remain high. The midline thalamic nuclei are an integral component of the cortical-basal ganglia-thalamic circuit and are poised to mediate addiction behaviors, including relapse. It is surprising that little research has examined the contribution of midline thalamic nuclei and their efferent projections in relapse. To address this, we expressed inhibitory, Gi/o -coupled DREADDs (Designer Receptors Exclusively Activated by Designer Drugs) in a subset of the midline thalamic nuclei or in midline thalamic nuclei neurons projecting to either the nucleus accumbens or the amygdala. We examined the effect of transiently decreasing activity of these neuronal populations on cue-induced and cocaine-primed reinstatement of cocaine-seeking. Reducing activity of midline thalamic nuclei neurons attenuated both cue-induced and cocaine-primed reinstatement, but had no effect on cue-induced reinstatement of sucrose-seeking or locomotor activity. Interestingly, attenuating activity of efferent projections from the anterior portion of midline thalamic nuclei to the nucleus accumbens blocked cocaine-primed reinstatement but enhanced cue-induced reinstatement. Decreasing activity of efferent projections from either the posterior midline thalamic nuclei to the nucleus accumbens or the midline thalamic nuclei to amygdala had no effect. These results reveal a novel contribution of subsets of midline thalamic nuclei neurons in drug-seeking behaviors and suggest that modulation of midline thalamic nuclei activity may be a promising therapeutic target for preventing relapse.


Asunto(s)
Trastornos Relacionados con Cocaína/metabolismo , Comportamiento de Búsqueda de Drogas , Núcleo Accumbens/efectos de los fármacos , Receptores Acoplados a Proteínas G/genética , Núcleos Talámicos/efectos de los fármacos , Animales , Clozapina/farmacología , Trastornos Relacionados con Cocaína/fisiopatología , Señales (Psicología) , Drogas de Diseño/farmacología , Vías Eferentes/efectos de los fármacos , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Núcleo Accumbens/citología , Núcleo Accumbens/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Memoria Implícita , Núcleos Talámicos/citología , Núcleos Talámicos/metabolismo
10.
PLoS One ; 12(4): e0174774, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28445527

RESUMEN

Dopaminergic reward dysfunction in addictive behaviors is well supported in the literature. There is evidence that alterations in synchronous neural activity between brain regions subserving reward and various cognitive functions may significantly contribute to substance-related disorders. This study presents the first evidence showing that a pro-dopaminergic nutraceutical (KB220Z) significantly enhances, above placebo, functional connectivity between reward and cognitive brain areas in the rat. These include the nucleus accumbens, anterior cingulate gyrus, anterior thalamic nuclei, hippocampus, prelimbic and infralimbic loci. Significant functional connectivity, increased brain connectivity volume recruitment (potentially neuroplasticity), and dopaminergic functionality were found across the brain reward circuitry. Increases in functional connectivity were specific to these regions and were not broadly distributed across the brain. While these initial findings have been observed in drug naïve rodents, this robust, yet selective response implies clinical relevance for addicted individuals at risk for relapse, who show reductions in functional connectivity after protracted withdrawal. Future studies will evaluate KB220Z in animal models of addiction.


Asunto(s)
Encéfalo/efectos de los fármacos , Catecolaminas/farmacología , Dopaminérgicos/farmacología , Monoaminooxidasa/farmacología , Neprilisina/farmacología , Animales , Encéfalo/anatomía & histología , Encéfalo/fisiología , Mapeo Encefálico , Cognición/efectos de los fármacos , Hipocampo/anatomía & histología , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Imagen por Resonancia Magnética , Masculino , Núcleo Accumbens/anatomía & histología , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/fisiología , Efecto Placebo , Ratas , Ratas Long-Evans , Núcleos Talámicos/anatomía & histología , Núcleos Talámicos/efectos de los fármacos , Núcleos Talámicos/fisiología
11.
Neuroscience ; 343: 10-20, 2017 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-27916729

RESUMEN

The intergeniculate leaflet (IGL) is a flat thalamic nucleus implicated in the modulation of circadian rhythmicity. In rat, two main GABAergic subpopulations can be distinguished in the IGL: neurons synthesizing neuropeptide Y (NPY), which directly innervates the suprachiasmatic nuclei, and enkephalinergic cells, which connect contralaterally located leaflets. The aim of this study was to evaluate possible effects of inner IGL neurotransmitters on the spontaneous and synaptic activity of IGL neurons. The data presented in this article provide evidence that enkephalin, and not NPY, could act upon the majority of IGL neurons. Moreover, we investigated the type of opioid receptor activated by enkephalin and showed that the µ-receptor is functionally predominant in the IGL. The application of met-enkephalin not only robustly hyperpolarized IGL neurons (both putatively NPY-synthesizing and putatively enkephalinergic neurons), but it also was able to inhibit GABAergic and glutamatergic synaptic transmission. Based on this and previous studies, we hypothesize that IGL enkephalinergic neurons may act as powerful interneurons that inhibit themselves and NPY-synthesizing neurons, also in the contralaterally located IGL.


Asunto(s)
Relojes Biológicos/fisiología , Encefalinas/metabolismo , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Núcleos Talámicos/metabolismo , Animales , Relojes Biológicos/efectos de los fármacos , Encefalinas/administración & dosificación , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Ácido Glutámico/metabolismo , Inmunohistoquímica , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/fisiología , Masculino , Potenciales Postsinápticos Miniatura/efectos de los fármacos , Potenciales Postsinápticos Miniatura/fisiología , Neuronas/citología , Neuronas/efectos de los fármacos , Neurotransmisores/administración & dosificación , Técnicas de Placa-Clamp , Ratas Wistar , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Núcleos Talámicos/citología , Núcleos Talámicos/efectos de los fármacos , Técnicas de Cultivo de Tejidos , Ácido gamma-Aminobutírico/metabolismo
12.
Brain Res ; 1644: 258-66, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27208494

RESUMEN

Through GABAergic fibers, globus pallidus (GP) coordinates basal ganglia global function. Electrical activity of GP neurons depends on their membrane properties and afferent fibers, including GABAergic fibers from striatum. In pathological conditions, abnormal electrical activity of GP neurons is associated with motor deficits. There is a GABAergic pathway from the GP to the reticular thalamic nucleus (RTn) whose contribution to RTn neurons electrical activity has received little attention. This fact called our attention because the RTn controls the overall information flow of thalamic nuclei to cerebral cortex. Here, we study the spontaneous electrical activity of RTn neurons recorded in vivo in anesthetized rats and under pharmacological activation or inhibition of the GP. We found that activation of GP predominantly diminishes the spontaneous RTn neurons firing rate and its inhibition increases their firing rate; however, both activation and inhibition of GP did not modified the burst index (BI) or the coefficient of variation (CV) of RTn neurons. Moreover, stimulation of striatum predominantly diminishes the spiking rate of GP cells and increases the spiking rate in RTn neurons without modifying the BI or CV in reticular neurons. Our data suggest a GP tight control over RTn spiking activity.


Asunto(s)
Potenciales de Acción , Cuerpo Estriado/fisiología , Globo Pálido/fisiología , Neuronas/fisiología , Núcleos Talámicos/fisiología , Animales , Cuerpo Estriado/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/administración & dosificación , GABAérgicos/administración & dosificación , Globo Pálido/efectos de los fármacos , Ácido Glutámico/administración & dosificación , Masculino , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/fisiología , Neuronas/efectos de los fármacos , Ratas , Ratas Wistar , Núcleos Talámicos/efectos de los fármacos , Ácido gamma-Aminobutírico/administración & dosificación
13.
Neuroscience ; 329: 306-17, 2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-27208616

RESUMEN

The intergeniculate leaflet (IGL) is a flat retinorecipient thalamic structure implicated in orchestrating circadian rhythm, historically considered to be a subdivision of the neighboring ventrolateral geniculate nucleus (VLG). IGL consists of two main neuronal subpopulations: enkephalinergic and neuropeptide Y (NPY)-synthesizing cells. These cell types have different functions, connectivity and firing pattern in vivo, which suggest that they have different membrane currents to support their functional differences. We therefore performed patch-clamp experiments combined with immunohistochemical staining to clarify possible differences in the subthreshold currents of IGL neurons. Our results suggest that IGL neurons can be divided into two subpopulations based on two ionic currents. A T-type calcium current (IT) was identified in neurons that do not synthesise NPY, whereas all NPY-positive neurons were found to express a marked A-type potassium current (IA). Due to the fact that the clear electrophysiological discriminants between IGL and VLG are lacking, we decided to compare the amplitudes of the identified currents between those two structures. Our data suggest that VLG neurons can be characterized by a high amplitude IT and a low IA. Finally, we compared both currents with WAG/Rij rats, a well-established model of absence epilepsy, with co-occurring retinal pathologies, sleep-onset disturbances, and seizures exhibiting circadian rhythmicity. Data presented in this study uncovered pathologies in the IT exhibiting neurons of the IGL and VLG. In conclusion, the data presented here suggest that different subthreshold current expression supports the functional differences of thalamic nuclei. Those differences are promising for possible pharmacological manipulations of specified cell types in pathophysiologies including absence epilepsy.


Asunto(s)
Neuronas/citología , Neuronas/fisiología , Núcleos Talámicos/citología , Núcleos Talámicos/fisiología , Animales , Calcio/metabolismo , Modelos Animales de Enfermedad , Epilepsia Tipo Ausencia/fisiopatología , Inmunohistoquímica , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Neuronas/efectos de los fármacos , Neuropéptido Y/metabolismo , Técnicas de Placa-Clamp , Potasio/metabolismo , Ratas Wistar , Núcleos Talámicos/efectos de los fármacos , Núcleos Talámicos/fisiopatología
14.
Cell Calcium ; 59(6): 312-23, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27041217

RESUMEN

The thalamic reticular nucleus (TRN) plays a major role in modulating the transfer of information from the thalamus to the cortex. GABAergic inhibition via the TRN is differentially regulated by metabotropic glutamate receptors (mGluRs) and the effect of mGluRs on the membrane potential, on ion channels, and on the plasticity of electrical coupling of TRN neurons has been studied previously. Although mGluRs are generally known to trigger Ca(2+) transients, mGluR-mediated Ca(2+)-transients in TRN neurons have not yet been investigated. In this study, we show that mGluRs can trigger Ca(2+)-transients in TRN neurons, that these transients depend on intracellular Ca(2+)-stores, and are mediated by IP3 receptors. Ca(2+) transients caused by the group I mGluR agonist DHPG elicit a current that is sensitive to flufenamic acid and has a reversal potential around -40mV. Our results add mGluR-mediated Ca(2+)-signalling in the TRN to the state-dependent modulators of the thalamocortical system.


Asunto(s)
Señalización del Calcio , Receptores de Glutamato Metabotrópico/metabolismo , Núcleos Talámicos/metabolismo , Animales , Ácido Benzoico/farmacología , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Espacio Intracelular/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Ratas , Núcleos Talámicos/efectos de los fármacos
15.
J Physiol ; 594(7): 1875-90, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26732231

RESUMEN

KEY POINTS: Despite the clinical importance of pre-emptive analgesia, the mechanisms by which it attenuates pain associated with central sensitization are poorly understood. We find that fentanyl and the α2-adrenoceptor agonist dexmedetomidine (Dex) differ significantly in their modulatory actions on noxious mechanical and noxious heat-evoked nociception in vivo. Unlike fentanyl, Dex modified descending control of nociception by decreasing the threshold for descending inhibition and/or increasing the threshold for descending facilitation. Dex exhibited after-actions on activities of thalamus in prolongation of noxious heat-evoked paw withdrawal latency that persisted for at least 7 days. This study provides insight into the organization of thalamic modulation in pre-emptive analgesia. ABSTRACT: We investigated and compared the antinociceptive effects of intraperitoneal administration of fentanyl (2-60 µg kg(-1)) and dexmedetomidine (Dex, 1-10 µg kg(-1); a highly selective α2-adrenoceptor agonist) in the regulation of nociception assessed by measuring noxious paw withdrawal reflexes in rats. Fentanyl elevated noxious mechanical paw withdrawal threshold and prolonged paw withdrawal heat latency within 1-1.5 h (P < 0.05). Dex failed to affect the mechanical paw withdrawal threshold, yet significantly prolonged the paw withdrawal heat latency in a bi-phasic manner; a short transient 1-1.5 h period followed by a second, slowly developing increase in latency that persisted for at least 7 days (P < 0.05). Lesion of the dorsolateral funiculus (DLF) did not influence fentanyl-induced antinociceptive effects, indicating peripheral and spinal antinociceptive mechanisms. By contrast, the Dex-induced second, but not the first, phase of the prolonged paw withdrawal heat latency was significantly blocked by the lesion of either DLF or thalamic ventromedial (VM) nuclei, and was attenuated by intracerebral administration of either atipamezole (α2-adrenoceptor antagonist) or WAY-100635 (5-HT1A receptor antagonist) into the VM nuclei (P < 0.05). Upon intramuscular 5.8% saline-induced muscle nociception, pre-emptive injection of fentanyl enhanced mechanical hyperalgesia and blocked heat hypoalgesia, whereas Dex significantly prevented the occurrence of mechanical hyperalgesia and enhanced heat hypoalgesia. It is suggested that Dex, but not fentanyl, significantly enhances descending inhibition and/or decreases descending facilitation to modulate pain and nociception. The present study provides novel insight into thalamus-mediated mechanisms in pre-emptive analgesia.


Asunto(s)
Analgésicos no Narcóticos/farmacología , Analgésicos Opioides/farmacología , Dexmedetomidina/farmacología , Fentanilo/farmacología , Hiperalgesia/tratamiento farmacológico , Núcleos Talámicos/efectos de los fármacos , Analgésicos no Narcóticos/administración & dosificación , Analgésicos no Narcóticos/uso terapéutico , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/uso terapéutico , Animales , Dexmedetomidina/administración & dosificación , Dexmedetomidina/uso terapéutico , Fentanilo/administración & dosificación , Fentanilo/uso terapéutico , Imidazoles/farmacología , Masculino , Inhibición Neural , Nocicepción , Umbral del Dolor , Piperazinas/farmacología , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Núcleos Talámicos/fisiología
16.
J Neurochem ; 136(3): 526-35, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26484945

RESUMEN

Serotonin receptors are targets of drug therapies for a variety of neuropsychiatric and neurodegenerative disorders. Cocaine inhibits the re-uptake of serotonin (5-HT), dopamine, and noradrenaline, whereas caffeine blocks adenosine receptors and opens ryanodine receptors in the endoplasmic reticulum. We studied how 5-HT and adenosine affected spontaneous GABAergic transmission from thalamic reticular nucleus. We combined whole-cell patch clamp recordings of miniature inhibitory post-synaptic currents (mIPSCs) in ventrobasal thalamic neurons during local (puff) application of 5-HT in wild type (WT) or knockout mice lacking 5-HT2A receptors (5-HT2A -/-). Inhibition of mIPSCs frequency by low (10 µM) and high (100 µM) 5-HT concentrations was observed in ventrobasal neurons from 5-HT2A -/- mice. In WT mice, only 100 µM 5-HT significantly reduced mIPSCs frequency. In 5-HT2A -/- mice, NAN-190, a specific 5-HT1A antagonist, prevented the 100 µM 5-HT inhibition while blocking H-currents that prolonged inhibition during post-puff periods. The inhibitory effects of 100 µM 5-HT were enhanced in cocaine binge-treated 5-HT2A -/- mice. Caffeine binge treatment did not affect 5-HT-mediated inhibition. Our findings suggest that both 5-HT1A and 5-HT2A receptors are present in pre-synaptic thalamic reticular nucleus terminals. Serotonergic-mediated inhibition of GABA release could underlie aberrant thalamocortical physiology described after repetitive consumption of cocaine. Our findings suggest that both 5-HT1A , 5-HT2A and A1 receptors are present in pre-synaptic TRN terminals. 5-HT1A and A1 receptors would down-regulate adenylate cyclase, whereas 5-HT1A would also increase the probability of the opening of G-protein-activated inwardly rectifying K(+) channels (GIRK). Sustained opening of GIRK channels would hyperpolarize pre-synaptic terminals activating H-currents, resulting in less GABA release. 5-HT2A -would activate PLC and IP3 , increasing intracellular [Ca(2+) ] and thus facilitating GABA release.


Asunto(s)
Cafeína/farmacología , Cocaína/farmacología , Inhibidores de Captación de Dopamina/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Receptor de Serotonina 5-HT2A/metabolismo , Núcleos Talámicos/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Cloruro de Cadmio/farmacología , Relación Dosis-Respuesta a Droga , Técnicas In Vitro , Inositol 1,4,5-Trifosfato/metabolismo , Masculino , Ratones , Ratones Noqueados , Técnicas de Placa-Clamp , Receptor de Serotonina 5-HT2A/genética , Serotonina/farmacología , Serotoninérgicos/farmacología , Núcleos Talámicos/metabolismo , Fosfolipasas de Tipo C/metabolismo
17.
Brain Res ; 1629: 38-53, 2015 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-26453289

RESUMEN

The brain circuits underlying tics in Tourette׳s syndrome (TS) are unknown but thought to involve cortico/amygdalo-striato-thalamo-cortical (CSTC) loop hyperactivity. We previously engineered a transgenic mouse "circuit model" of TS by expressing an artificial neuropotentiating transgene (encoding the cAMP-elevating, intracellular A1 subunit of cholera toxin) within a small population of dopamine D1 receptor-expressing somatosensory cortical and limbic neurons that hyperactivate cortico/amygdalostriatal glutamatergic output circuits thought to be hyperactive in TS and comorbid obsessive-compulsive (OC) disorders. As in TS, these D1CT-7 ("Ticcy") transgenic mice׳s tics were alleviated by the TS drugs clonidine and dopamine D2 receptor antagonists; and their chronic glutamate-excited striatal motor output was unbalanced toward hyperactivity of the motoric direct pathway and inactivity of the cataleptic indirect pathway. Here we have examined whether these mice׳s tics are countered by drugs that "break" sequential elements of their hyperactive cortical/amygdalar glutamatergic and efferent striatal circuit: anti-serotonoceptive and anti-noradrenoceptive corticostriatal glutamate output blockers (the serotonin 5-HT2a,c receptor antagonist ritanserin and the NE alpha-1 receptor antagonist prazosin); agmatinergic striatothalamic GABA output blockers (the presynaptic agmatine/imidazoline I1 receptor agonist moxonidine); and nigrostriatal dopamine output blockers (the presynaptic D2 receptor agonist bromocriptine). Each drug class alleviates tics in the Ticcy mice, suggesting a hyperglutamatergic CSTC "tic circuit" could exist in TS wherein cortical/amygdalar pyramidal projection neurons׳ glutamatergic overexcitation of both striatal output neurons and nigrostriatal dopaminergic modulatory neurons unbalances their circuit integration to excite striatothalamic output and create tics, and illuminating new TS drug strategies.


Asunto(s)
Cuerpo Estriado/metabolismo , Ácido Glutámico/metabolismo , Corteza Somatosensorial/metabolismo , Núcleos Talámicos/metabolismo , Tics/metabolismo , Síndrome de Tourette/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Antagonistas de Receptores Adrenérgicos alfa 1/uso terapéutico , Animales , Cuerpo Estriado/efectos de los fármacos , Modelos Animales de Enfermedad , Agonistas de Dopamina/farmacología , Agonistas de Dopamina/uso terapéutico , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Red Nerviosa/efectos de los fármacos , Red Nerviosa/metabolismo , Antagonistas del Receptor de Serotonina 5-HT2/farmacología , Antagonistas del Receptor de Serotonina 5-HT2/uso terapéutico , Corteza Somatosensorial/efectos de los fármacos , Núcleos Talámicos/efectos de los fármacos , Tics/tratamiento farmacológico , Síndrome de Tourette/tratamiento farmacológico
18.
Neuroscience ; 304: 36-46, 2015 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-26189794

RESUMEN

It has been proposed that thalamic mediodorsal (MD) and ventromedial (VM) nuclei form thalamic 'nociceptive discriminators' in discrimination of nociceptive afferents, and specifically govern endogenous descending facilitation and inhibition. The present study conducted in rats was to explore the role of thalamic MD and VM nuclei in modulation of cerebral neuronal activities by means of detection of spatiotemporal variations of Fos expression within the cerebral cortex. Following a unilateral intramuscular injection of 5.8% saline into the gastrocnemius muscle, Fos expression within the bilateral, different areas of the cerebral cortex except S2 was significantly increased (P<0.05). Particularly, the increases in Fos expression within the cingulate cortex and the insular cortex occurred at 0.5h, 4h and reached the peak level at 4h, 16h, respectively. Electrolytic lesion of the contralateral thalamic MD and VM nuclei significantly blocked the 5.8% saline intramuscularly induced increases in Fos expression within the bilateral cingulate and insular cortices, respectively. Additionally, the 5.8% saline-induced Fos expression in the cingulate cortex and the insular cortex were dose-dependently attenuated by microinjection of µ-opioid antagonist ß-funaltrexamine hydrochloride into the thalamic MD and VM nuclei. It is suggested that (1) the neural circuits of 'thalamic MD nucleus - cingulate cortex' and 'thalamic VM nucleus - insular cortex' form two distinct pathways in the endogenous control of nociception, (2) mirror or contralateral pain is hypothesized to be related to cross-talk of neuronal activities within the bilateral cerebral cortices modulated by µ-opioid receptors within the thalamic MD and VM nuclei.


Asunto(s)
Corteza Cerebral/metabolismo , Mialgia/metabolismo , Dolor Nociceptivo/metabolismo , Núcleos Talámicos/metabolismo , Animales , Modelos Animales de Enfermedad , Estimulación Eléctrica , Lateralidad Funcional/fisiología , Masculino , Mialgia/tratamiento farmacológico , Naltrexona/administración & dosificación , Naltrexona/análogos & derivados , Antagonistas de Narcóticos/administración & dosificación , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/metabolismo , Dolor Nociceptivo/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas Sprague-Dawley , Receptores Opioides mu/antagonistas & inhibidores , Receptores Opioides mu/metabolismo , Solución Salina Hipertónica , Núcleos Talámicos/efectos de los fármacos , Núcleos Talámicos/lesiones , Factores de Tiempo
19.
Eur J Neurosci ; 41(1): 120-8, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25327971

RESUMEN

This study investigated the effect of repetitive cortical spreading depression (CSD) on behaviour and the anatomical and physiological patterns of cellular activation of cortical and subcortical areas in awake, moving rats. Rat behaviours in response to repetitive CSD events evoked by the application of KCl were quantified with electrophysiological recording. Immunohistochemistry was used to quantify anatomical regions of cellular activation. The effects of acute valproic acid administration on the behavioural parameters and cellular activation were evaluated. CSD significantly decreased locomotor activity and induced freezing in awake, moving rats, and stimulated c-Fos expression in the cortex, trigeminal nucleus caudalis (TNC), and amygdala. CSD also resulted in a prominent increase in c-Fos expression in the ipsilateral thalamic reticular nucleus (TRN) visual sector. Electrophysiological recordings revealed propagation of CSD into the TRN. Valproic acid pretreatment decreased the duration of CSD-induced freezing episodes and reversed the CSD-induced reduction in locomotor activity. Acute valproic acid administration also significantly blocked CSD-induced c-Fos expression in the TNC and TRN. These findings show that CSD events cause consistent behavioural responses and activate specific brain regions in awake, freely moving rats. Selective activation of TRN by CSD and the suppression of this activation by valproic acid suggest that this brain region may play an important role in migraine pathogenesis and may represent a novel target for migraine therapy.


Asunto(s)
Depresión de Propagación Cortical/efectos de los fármacos , Depresión de Propagación Cortical/fisiología , GABAérgicos/farmacología , Núcleos Talámicos/efectos de los fármacos , Núcleos Talámicos/fisiología , Ácido Valproico/farmacología , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/fisiología , Animales , Fármacos del Sistema Nervioso Central/farmacología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/fisiología , Electrodos Implantados , Reacción Cataléptica de Congelación/efectos de los fármacos , Reacción Cataléptica de Congelación/fisiología , Inmunohistoquímica , Masculino , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Cloruro de Potasio/farmacología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas Wistar , Núcleo Caudal del Trigémino/efectos de los fármacos , Núcleo Caudal del Trigémino/fisiología
20.
Nature ; 511(7510): 471-4, 2014 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-24828045

RESUMEN

During development, thalamocortical (TC) input has a critical role in the spatial delineation and patterning of cortical areas, yet the underlying cellular and molecular mechanisms that drive cortical neuron differentiation are poorly understood. In the primary (S1) and secondary (S2) somatosensory cortex, layer 4 (L4) neurons receive mutually exclusive input originating from two thalamic nuclei: the ventrobasalis (VB), which conveys tactile input, and the posterior nucleus (Po), which conveys modulatory and nociceptive input. Recently, we have shown that L4 neuron identity is not fully committed postnatally, implying a capacity for TC input to influence differentiation during cortical circuit assembly. Here we investigate whether the cell-type-specific molecular and functional identity of L4 neurons is instructed by the origin of their TC input. Genetic ablation of the VB at birth resulted in an anatomical and functional rewiring of Po projections onto L4 neurons in S1. This induced acquisition of Po input led to a respecification of postsynaptic L4 neurons, which developed functional molecular features of Po-target neurons while repressing VB-target traits. Respecified L4 neurons were able to respond both to touch and to noxious stimuli, in sharp contrast to the normal segregation of these sensory modalities in distinct cortical circuits. These findings reveal a behaviourally relevant TC-input-type-specific control over the molecular and functional differentiation of postsynaptic L4 neurons and cognate intracortical circuits, which instructs the development of modality-specific neuronal and circuit properties during corticogenesis.


Asunto(s)
Diferenciación Celular , Vías Nerviosas/fisiología , Neuronas/citología , Neuronas/fisiología , Densidad Postsináptica/fisiología , Corteza Somatosensorial/fisiología , Núcleos Talámicos/fisiología , Animales , Axones/efectos de los fármacos , Axones/fisiología , Capsaicina/farmacología , Diferenciación Celular/efectos de los fármacos , Femenino , Masculino , Ratones Endogámicos C57BL , Vías Nerviosas/efectos de los fármacos , Neuronas/efectos de los fármacos , Noxas/farmacología , Optogenética , Densidad Postsináptica/efectos de los fármacos , Corteza Somatosensorial/citología , Corteza Somatosensorial/efectos de los fármacos , Potenciales Sinápticos/efectos de los fármacos , Núcleos Talámicos/citología , Núcleos Talámicos/efectos de los fármacos , Tacto/fisiología , Vibrisas/efectos de los fármacos , Vibrisas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...