Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros











Intervalo de año de publicación
1.
Nat Commun ; 11(1): 4929, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33004789

RESUMEN

Non-invasive, molecularly-specific, focal modulation of brain circuits with low off-target effects can lead to breakthroughs in treatments of brain disorders. We systemically inject engineered ultrasound-controllable drug carriers and subsequently apply a novel two-component Aggregation and Uncaging Focused Ultrasound Sequence (AU-FUS) at the desired targets inside the brain. The first sequence aggregates drug carriers with millimeter-precision by orders of magnitude. The second sequence uncages the carrier's cargo locally to achieve high target specificity without compromising the blood-brain barrier (BBB). Upon release from the carriers, drugs locally cross the intact BBB. We show circuit-specific manipulation of sensory signaling in motor cortex in rats by locally concentrating and releasing a GABAA receptor agonist from ultrasound-controlled carriers. Our approach uses orders of magnitude (1300x) less drug than is otherwise required by systemic injection and requires very low ultrasound pressures (20-fold below FDA safety limits for diagnostic imaging). We show that the BBB remains intact using passive cavitation detection (PCD), MRI-contrast agents and, importantly, also by sensitive fluorescent dye extravasation and immunohistochemistry.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encefalopatías/tratamiento farmacológico , Portadores de Fármacos/efectos de la radiación , Agonistas de Receptores de GABA-A/administración & dosificación , Ultrasonografía Intervencional/métodos , Animales , Barrera Hematoencefálica/diagnóstico por imagen , Barrera Hematoencefálica/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Femenino , Agonistas de Receptores de GABA-A/farmacocinética , Humanos , Imagen por Resonancia Magnética , Modelos Animales , Muscimol/administración & dosificación , Muscimol/farmacocinética , Ratas , Técnicas Estereotáxicas , Ondas Ultrasónicas
2.
J Neurosci ; 40(12): 2485-2497, 2020 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-32051327

RESUMEN

We recently developed a rat model of relapse to drug seeking after food choice-induced voluntary abstinence. Here, we used this model to study the role of the orbitofrontal cortex (OFC) and its afferent projections in relapse to fentanyl seeking. We trained male and female rats to self-administer palatable food pellets for 6 d (6 h/d) and intravenous fentanyl (2.5 µg/kg/infusion) for 12 d (6 h/d). We assessed relapse to fentanyl seeking after 13-14 voluntary abstinence days, achieved through a discrete choice procedure between fentanyl infusions and palatable food (20 trials/d). In both sexes, relapse after food choice-induced abstinence was associated with increased expression of the activity marker Fos in the OFC. Pharmacological inactivation of the OFC with muscimol plus baclofen (50 + 50 ng/side) decreased relapse to fentanyl seeking. We then determined projection-specific activation of OFC afferents during the relapse test by using Fos plus the retrograde tracer cholera toxin B (injected into the OFC). Relapse to fentanyl seeking was associated with increased Fos expression in the piriform cortex (Pir) neurons projecting to the OFC, but not in projections from the basolateral amygdala and thalamus. Pharmacological inactivation of the Pir with muscimol plus baclofen decreased relapse to fentanyl seeking after voluntary abstinence. Next, we used an anatomical disconnection procedure to determine whether projections between the Pir and OFC are critical for relapse to fentanyl seeking. Unilateral muscimol plus baclofen injections into the Pir in one hemisphere plus unilateral muscimol plus baclofen injections into the OFC in the contralateral, but not ipsilateral, hemisphere decreased relapse. Our results identify Pir-OFC projections as a new motivation-related pathway critical to relapse to opioid seeking after voluntary abstinence.SIGNIFICANCE STATEMENT There are few preclinical studies of fentanyl relapse, and these studies have used experimenter-imposed extinction or forced abstinence procedures. In humans, however, abstinence is often voluntary, with drug available in the drug environment but forgone in favor of nondrug alternative reinforcers. We recently developed a rat model of drug relapse after palatable food choice-induced voluntary abstinence. Here, we used classical pharmacology, immunohistochemistry, and retrograde tracing to demonstrate a critical role of the piriform and orbitofrontal cortices in relapse to opioid seeking after voluntary abstinence.


Asunto(s)
Analgésicos Opioides , Comportamiento de Búsqueda de Drogas , Fentanilo , Trastornos Relacionados con Opioides/fisiopatología , Trastornos Relacionados con Opioides/psicología , Corteza Piriforme/fisiopatología , Corteza Prefrontal/fisiopatología , Animales , Baclofeno/administración & dosificación , Baclofeno/farmacología , Conducta de Elección , Femenino , Preferencias Alimentarias , Agonistas del GABA/administración & dosificación , Agonistas del GABA/farmacología , Expresión Génica/efectos de los fármacos , Genes fos , Masculino , Microinyecciones , Muscimol/administración & dosificación , Muscimol/farmacología , Ratas , Ratas Sprague-Dawley , Recurrencia , Autoadministración
3.
Front Endocrinol (Lausanne) ; 11: 552805, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33505354

RESUMEN

Progestogens' (e.g., progesterone and its neuroactive metabolite, allopregnanolone), cognitive effects and mechanisms among males are not well-understood. We hypothesized if progestogen's effects on cognitive performance are through its metabolite allopregnanolone, and not actions via binding to traditional progestin receptors (PRs), then progesterone administration would enhance performance in tasks mediated by the hippocampus and cortex, coincident with increasing allopregnanolone concentrations, brain derived neurotrophic factor (BDNF) and/or muscimol binding of PR knock out (PRKO) and wild-type PR replete mice. Experiment 1: Progesterone (4 mg/kg, subcutaneously (SC; n = 12/grp), or oil vehicle control, was administered to gonadally-intact adult male mice PRKO mice and their wild-type counterparts and cognitive behaviors in object recognition, T-maze and water maze was examined. Progesterone, compared to vehicle, when administered post-training increased time investigating novel objects by the PRKO and wild-type mice in the object recognition task. In the T-maze task, progesterone administration to wild-type and PRKO mice had significantly greater number of spontaneous alternations compared to their vehicle-administered counterparts. In the water maze task, PRKO mice administered vehicle spent significantly fewer seconds in the quadrant associated with the escape platform on testing compared to all other groups. Experiment 2: Progesterone administered to wild-type and PRKO mice increased plasma progesterone and allopregnanolone levels (n = 5/group). PRKO mice had higher allopregnanolone levels in plasma and hippocampus, but not cortex, when administered progesterone and compared to wild-type mice. Experiment 3: Assessment of PR binding revealed progesterone administered wild-type mice had significantly greater levels of PRs in the hippocampus and cortex, compared to all other groups (n = 5/group). Wild-type mice administered progesterone, but not vehicle, had increased BDNF levels in the hippocampus, but not the cortex, compared to PRKOs. Wild-type as well as PRKO mice administered progesterone experienced significant increases in maximal GABAA agonist, muscimol, binding in hippocampus and cortex, compared to their vehicle-administered counterparts. Thus, adult male mice can be responsive to progesterone for cognitive performance, and such effects may be independent of PRs trophic actions of BDNF levels in the hippocampus and/or increases in GABAA activity in the hippocampus and cortex.


Asunto(s)
Corteza Cerebral/fisiología , Hipocampo/fisiología , Aprendizaje por Laberinto/fisiología , Progesterona/administración & dosificación , Receptores de GABA-A/fisiología , Receptores de Progesterona/fisiología , Memoria Espacial/fisiología , Animales , Factor Neurotrófico Derivado del Encéfalo/fisiología , Agonistas de Receptores de GABA-A/administración & dosificación , Masculino , Ratones Noqueados , Muscimol/administración & dosificación , Receptores de Progesterona/genética
4.
Neurosurgery ; 85(1): E4-E15, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30407567

RESUMEN

BACKGROUND: Minimally invasive therapies for drug-resistant epilepsy (DRE) have been advocated. A study of convection-enhanced delivery (CED) of muscimol, a GABAA receptor agonist, was previously completed in non-human primates. OBJECTIVE: To investigate the safety and anti-epileptic effects of intracerebral muscimol infusion into the epileptic focus of patients with DRE. METHODS: In this phase 1 clinical trial, 3 adult patients with DRE underwent CED into the seizure focus of artificial CSF vehicle followed by muscimol for 12 to 24 h each using a crossover design. Basic pathophysiology of the epileptic focus was examined by assessing the infusions' effects on seizure frequency, electroencephalogram (EEG) spike-wave activity, and power-spectral EEG frequency. RESULTS: Inter-ictal neurological function remained normal in all patients. Pathological examination of resected specimens showed no infusion-related brain injuries. Seizure frequency decreased in 1 of 3 patients during muscimol infusion but was unchanged in all patients during vehicle infusion. Mean beta frequencies did not differ significantly before, during, or after infusion periods. Infused fluid provided insufficient MRI-signal to track infusate distribution. In the 2 yr after standard epilepsy surgery, 1 patient had temporary reduction in seizure frequency and 2 patients were seizure-free. CONCLUSION: CED of muscimol into the epileptic focus of patients with DRE did not damage adjacent brain parenchyma or adversely affect seizure surgery outcome. This study did not confirm that intracerebral muscimol infusion effectively suppressed seizures. A surrogate tracer is recommended to track infusion distribution to the epileptic focus and surrounding structures in future studies using CED to suppress the seizure focus.


Asunto(s)
Anticonvulsivantes/administración & dosificación , Encéfalo/efectos de los fármacos , Epilepsia Refractaria/tratamiento farmacológico , Infusiones Intraventriculares , Muscimol/administración & dosificación , Adulto , Convección , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino
5.
J Neurosci ; 37(38): 9320-9331, 2017 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-28821646

RESUMEN

The transition from wakefulness to general anesthesia is widely attributed to suppressive actions of anesthetic molecules distributed by the systemic circulation to the cerebral cortex (for amnesia and loss of consciousness) and to the spinal cord (for atonia and antinociception). An alternative hypothesis proposes that anesthetics act on one or more brainstem or diencephalic nuclei, with suppression of cortex and spinal cord mediated by dedicated axonal pathways. Previously, we documented induction of an anesthesia-like state in rats by microinjection of small amounts of GABAA-receptor agonists into an upper brainstem region named the mesopontine tegmental anesthesia area (MPTA). Correspondingly, lesioning this area rendered animals resistant to systemically delivered anesthetics. Here, using rats of both sexes, we applied a modified microinjection method that permitted localization of the anesthetic-sensitive neurons with much improved spatial resolution. Microinjected at the MPTA hotspot identified, exposure of 1900 or fewer neurons to muscimol was sufficient to sustain whole-body general anesthesia; microinjection as little as 0.5 mm off-target did not. The GABAergic anesthetics pentobarbital and propofol were also effective. The GABA-sensitive cell cluster is centered on a tegmental (reticular) field traversed by fibers of the superior cerebellar peduncle. It has no specific nuclear designation and has not previously been implicated in brain-state transitions.SIGNIFICANCE STATEMENT General anesthesia permits pain-free surgery. Furthermore, because anesthetic agents have the unique ability to reversibly switch the brain from wakefulness to a state of unconsciousness, knowing how and where they work is a potential route to unraveling the neural mechanisms that underlie awareness itself. Using a novel method, we have located a small, and apparently one of a kind, cluster of neurons in the mesopontine tegmentum that are capable of effecting brain-state switching when exposed to GABAA-receptor agonists. This action appears to be mediated by a network of dedicated axonal pathways that project directly and/or indirectly to nearby arousal nuclei of the brainstem and to more distant targets in the forebrain and spinal cord.


Asunto(s)
Anestésicos Generales/administración & dosificación , Mesencéfalo/efectos de los fármacos , Puente/efectos de los fármacos , Puente/fisiología , Inconsciencia/inducido químicamente , Inconsciencia/fisiopatología , Animales , Femenino , Masculino , Muscimol/administración & dosificación , Neuronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Ratas Wistar
6.
Brain Struct Funct ; 222(1): 113-129, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-26951288

RESUMEN

Previous studies from our group have shown that cytotoxic lesions in the ventral portion of the anteromedial thalamic nucleus (AMv), one of the main targets of the hypothalamic predator-responsive circuit, strongly impairs contextual fear responses to an environment previously associated with a predator. The AMv is in a position to convey information to cortico-hippocampal-amygdalar circuits involved in the processing of fear memory. However, it remains to be determined whether the nucleus is involved in the acquisition or subsequent expression of contextual fear. In the present investigation, we addressed this question by inactivating the rat AMv with muscimol either prior to cat exposure or prior to exposure to the cat-related context. Accordingly, AMv pharmacological inactivation prior to cat exposure did not interfere with innate fear responses, but it drastically reduced contextual conditioning to the predator-associated environment. On the other hand, AMv inactivation prior to exposure to the environment associated with the predator threat did not affect contextual fear responses. The behavioral results were further supported by the demonstration that AMv inactivation prior to cat exposure also blocked the activation of sites critically involved in the expression of anti-predatory contextual defensive responses (i.e., the dorsal premammillary nucleus and the dorsolateral periaqueductal gray) in animals exposed to the predator-associated context. The AMv projections were also examined, and the results of this investigation outline important paths that can influence hippocampal circuitry and raise new ideas for anterior thalamic-hippocampal paths involved in emotional learning.


Asunto(s)
Núcleos Talámicos Anteriores/fisiología , Miedo/fisiología , Memoria/fisiología , Animales , Núcleos Talámicos Anteriores/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Gatos , Condicionamiento Psicológico/efectos de los fármacos , Condicionamiento Psicológico/fisiología , Miedo/efectos de los fármacos , Agonistas de Receptores de GABA-A/administración & dosificación , Hipotálamo Posterior/efectos de los fármacos , Hipotálamo Posterior/fisiología , Masculino , Memoria/efectos de los fármacos , Muscimol/administración & dosificación , Sustancia Gris Periacueductal/efectos de los fármacos , Sustancia Gris Periacueductal/fisiología , Conducta Predatoria , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Wistar
7.
Sleep ; 39(6): 1293-303, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27091518

RESUMEN

STUDY OBJECTIVES: To examine the REM sleep response to stress and fearful memories as a potential marker of stress resilience and vulnerability and to assess the role of the basolateral amygdala (BLA) in mediating the effects of fear memory on sleep. METHODS: Outbred Wistar rats were surgically implanted with electrodes for recording EEG and EMG and with bilateral guide cannulae directed at the BLA. Data loggers were placed intraperitoneally to record core body temperature. After recovery from surgery, the rats received shock training (ST: 20 footshocks, 0.8 mA, 0.5-s duration, 60-s interstimulus interval) and afterwards received microinjections of the GABAA agonist muscimol (MUS; 1.0 µM) to inactivate BLA or microinjections of vehicle (VEH) alone. Subsequently, the rats were separated into 4 groups (VEH-vulnerable (VEH-Vul; n = 14), VEH-resilient (VEH-Res; n = 13), MUS-vulnerable (MUS-Vul; n = 8), and MUS-resilient (MUS-Res; n = 11) based on whether or not REM was decreased, compared to baseline, during the first 4 h following ST. We then compared sleep, freezing, and the stress response (stress-induced hyperthermia, SIH) across groups to determine the effects of ST and fearful context re-exposure alone (CTX). RESULTS: REM was significantly reduced on the ST day in both VEH-Vul and MUS-Vul rats; however, post-ST MUS blocked the reduction in REM on the CTX day in the MUS-Vul group. The VEH-Res and MUS-Res rats showed similar levels of REM on both ST and CTX days. The effects of post-ST inactivation of BLA on freezing and SIH were minimal. CONCLUSIONS: Outbred Wistar rats can show significant individual differences in the effects of stress on REM that are mediated by BLA. These differences in REM can be independent of behavioral fear and the peripheral stress response, and may be an important biomarker of stress resilience and vulnerability.


Asunto(s)
Adaptación Psicológica/fisiología , Amígdala del Cerebelo/fisiología , Condicionamiento Psicológico/fisiología , Miedo/fisiología , Individualidad , Resiliencia Psicológica , Sueño REM/fisiología , Estrés Psicológico/fisiopatología , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/fisiopatología , Animales , Modelos Animales de Enfermedad , Electroencefalografía , Electromiografía , Miedo/efectos de los fármacos , Fiebre/fisiopatología , Reacción Cataléptica de Congelación/fisiología , Memoria/efectos de los fármacos , Memoria/fisiología , Muscimol/administración & dosificación , Muscimol/farmacología , Ratas , Ratas Wistar , Sueño REM/efectos de los fármacos
8.
Alcohol Clin Exp Res ; 40(3): 572-82, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26876382

RESUMEN

BACKGROUND: The aversive properties of ethanol (EtOH) that limit its intake are poorly understood. There is an increasing interest in the role of the rostromedial tegmental nucleus (RMTg), because it encodes aversion signals and inhibits motivated behaviors. It is also a major source of inhibitory GABAergic inputs to the midbrain dopamine neurons. Up to this time, the role of the RMTg in EtOH-drinking behaviors has not been well explored. METHODS: Male Long-Evans rats were trained either to drink EtOH under the intermittent 2-bottle-choice protocol or to self-administer EtOH in operant chambers under fixed-ratio-3 schedules. Changes in drinking behaviors induced by the bilateral infusion into the RMTg of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), an agonist of AMPA-type glutamate receptors, or muscimol, an agonist of GABAA receptors, were measured. RESULTS: Consumption and preference for EtOH, numbers of active lever pressing, and head entrance to the EtOH port were all significantly decreased upon activation of the RMTg by the infusion of AMPA, but were increased upon inhibition of the RMTg by the infusion of muscimol. By contrast, intra-RMTg infusion of these agents did not change sucrose consumption. CONCLUSIONS: These data show for the first time that EtOH-drinking and EtOH-seeking behaviors of rats changed inversely with RMTg function, supporting the idea that the RMTg plays a crucial role in EtOH-drinking behaviors.


Asunto(s)
Consumo de Bebidas Alcohólicas/psicología , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Operante/fisiología , Etanol/administración & dosificación , Núcleo Tegmental Pedunculopontino/efectos de los fármacos , Núcleo Tegmental Pedunculopontino/fisiología , Consumo de Bebidas Alcohólicas/tratamiento farmacológico , Animales , Masculino , Microinyecciones , Muscimol/administración & dosificación , Ratas , Ratas Long-Evans , Autoadministración , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/administración & dosificación
9.
Can J Physiol Pharmacol ; 94(1): 18-27, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26497164

RESUMEN

The changes in urine flow and renal sympathetic nerve activity (RSNA) due to pulmonary lymphatic obstruction (PLO) were examined in anesthetized, artificially ventilated New Zealand white rabbits. PLO was produced by pressurizing an isolated pouch created in the right external jugular vein at the points of entry of the right lymphatic ducts. During this maneuver, urine flow increased from 8.5 ± 0.3 mL/10 min to 12 ± 0.5 mL/10 min (P < 0.0001) and RSNA increased from 24.0 ± 4 to 40.0 ± 5 µV·s (P < 0.0001). Bilateral lesioning of the paraventricular nucleus (PVN) of the hypothalamus or cervical vagotomy abolished these responses. PLO increased c-fos gene expression in the PVN. The increase in urine flow due to PLO was attenuated by muscimol and abolished by kynurenic acid microinjections into the PVN. The results show that (i) neurons in the PVN are an important relay site in the reflex arc, which is activated by PLO; and (ii) this activation is regulated by glutamatergic and partly by GABAergic input to the PVN.


Asunto(s)
Diuresis/fisiología , Enfermedades Pulmonares/fisiopatología , Enfermedades Linfáticas/fisiopatología , Núcleo Hipotalámico Paraventricular/fisiopatología , Animales , Modelos Animales de Enfermedad , Diuresis/efectos de los fármacos , Expresión Génica , Genes fos , Riñón/inervación , Ácido Quinurénico/administración & dosificación , Enfermedades Pulmonares/genética , Enfermedades Linfáticas/genética , Vasos Linfáticos/fisiopatología , Microinyecciones , Muscimol/administración & dosificación , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Conejos , Reflejo/efectos de los fármacos , Reflejo/fisiología , Sistema Nervioso Simpático/fisiopatología , Vagotomía
10.
Brain Struct Funct ; 221(1): 91-102, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25260556

RESUMEN

Systems-level consolidation models propose that recent memories are initially hippocampus-dependent. When remote, they are partially or completely dependent upon the medial prefrontal cortex (mPFC). An implication of the mPFC in recent memory, however, is still debated. Different amounts of muscimol (MSCI 0, 30, 50, 80 and 250 ng in 1 µL PBS) were used to assess the impact of inactivation of the dorsal hippocampus (dHip) or the mPFC (targeting the prelimbic cortex) on a 24-h delayed retrieval of a platform location that rats had learned drug-free in a water maze. The two smallest amounts of MSCI (30 and 50 ng) did not affect recall, whatever the region. 80 ng MSCI infused into the dHip disrupted spatial memory retrieval, as did the larger amount. Infusion of MSCI into the mPFC did not alter performance in the 0-80 ng range. At 250 ng, it induced an as dramatic memory impairment as after efficient dHip inactivation. Stereological quantifications showed that 80 ng MSCI in the dHip and 250 ng MSCI in the mPFC induced a more than 80% reduction of c-Fos expression, suggesting that, beyond the amounts infused, it is the magnitude of the neuronal activity decrease which is determinant as to the functional outcome of the inactivation. Because, based on the literature, even 250 ng MSCI is a small amount, our results point to a contribution of the mPFC to the recall of a recently acquired spatial memory and thereby extend our knowledge about the functions of this major actor of cognition.


Asunto(s)
Hipocampo/fisiología , Corteza Prefrontal/fisiología , Memoria Espacial/fisiología , Animales , Agonistas de Receptores de GABA-A/administración & dosificación , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Recuerdo Mental/efectos de los fármacos , Recuerdo Mental/fisiología , Muscimol/administración & dosificación , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Long-Evans , Memoria Espacial/efectos de los fármacos
11.
Behav Brain Res ; 270: 287-94, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24875771

RESUMEN

We have previously shown a functional motor interaction between ethanol and Δ(9)-tetrahydrocannabinol (Δ(9)-THC) that involved cerebellar adenosinergic A1 and GABAergic A receptor modulation. We now report the development of cross-tolerance between intracerebellar Δ(9)-THC and intraperitoneal ethanol using ataxia as the test response in male CD-1 mice. The drugs [Δ(9)-THC (20 µg), N(6)-cyclohexyladenosine, CHA (12 ng), muscimol (20 ng)] used in the study were directly microinfused stereotaxically via guide cannulas into the cerebellum except ethanol. Δ(9)-THC, infused once daily for 5 days followed 16 h after the last infusion by acute ethanol (2g/kg) and Rotorod evaluation, virtually abolished ethanol ataxia indicating development of cross-tolerance. The cross-tolerance was also observed when the order of ethanol and Δ(9)-THC treatment was reversed, i.e., ethanol injected once daily for 5 days followed 16 h after the last ethanol injection by Δ(9)-THC infusion. The cross-tolerance appeared within 24-48 h, lasted over 72 h and was maximal in 5-day ethanol/Δ(9)-THC-treated animals. Finally, tolerance in chronic ethanol/Δ(9)-THC/-treated animals developed not only to ethanol/Δ(9)-THC-induced ataxia, respectively, but also to the ataxia potentiating effect of CHA and muscimol, indicating modulation by cerebellar adenosinergic A1 and GABAA receptors. A practical implication of these results could be that marijuana smokers may experience little or no negative effects such as ataxia following alcohol consumption. Clinically, such antagonism of ethanol-induced ataxia can be observed in marijuana users thereby encouraging more alcohol consumption and thus may represent a risk factor for the development of alcoholism in this segment of population.


Asunto(s)
Ataxia Cerebelosa/inducido químicamente , Cerebelo/efectos de los fármacos , Dronabinol/análogos & derivados , Tolerancia a Medicamentos , Etanol/farmacología , Receptor de Adenosina A1/metabolismo , Receptores de GABA-A/metabolismo , Adenosina/administración & dosificación , Adenosina/análogos & derivados , Adenosina/farmacología , Animales , Agonistas de Receptores de Cannabinoides/administración & dosificación , Agonistas de Receptores de Cannabinoides/farmacología , Depresores del Sistema Nervioso Central/administración & dosificación , Depresores del Sistema Nervioso Central/farmacología , Ataxia Cerebelosa/fisiopatología , Cerebelo/fisiopatología , Dronabinol/administración & dosificación , Dronabinol/farmacología , Interacciones Farmacológicas , Etanol/administración & dosificación , Agonistas de Receptores de GABA-A/administración & dosificación , Agonistas de Receptores de GABA-A/farmacología , Masculino , Ratones , Ratones Endogámicos , Microinyecciones , Muscimol/administración & dosificación , Muscimol/farmacología , Agonistas del Receptor Purinérgico P1/administración & dosificación , Agonistas del Receptor Purinérgico P1/farmacología , Receptor de Adenosina A1/efectos de los fármacos , Receptores de GABA-A/efectos de los fármacos
12.
Ann Transplant ; 18: 299-313, 2013 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-23792534

RESUMEN

BACKGROUND: Gamma-aminobutyric acid (GABA) is found throughout the body. The regulation of GABA receptor (GABAR) reduces oxidative stress (OS). Ischemia/reperfusion injury after orthotopic liver transplantation (OLT) causes OS-induced graft damage. The effects of GABAR regulation in donors in vivo were investigated. MATERIAL AND METHODS: Donor rats received saline, a GABAR agonist or GABAR antagonist 4 h before surgery. Recipient rats were divided into four groups according to the donor treatments: laparotomy, OLT with saline, OLT with GABAR agonist and OLT with GABAR antagonist. Histopathological, biochemical and immunohistological examinations were performed at 6, 12 and 24 h after OLT. Protein assays were performed at 6 h after OLT. The 4-hydroxynonenal (4-HNE), ataxia-telangiectasia mutated kinase (ATM), phosphorylated histone H2AX (gammaH2AX), phosphatidylinositol-3 kinase (PI3K), Akt and superoxide dismutase (SOD) were assessed by western blot analysis. RESULTS: In the univariate analysis, histopathological and biochemical profiles verified that the GABAR agonist reduced graft damage. Immunohistology revealed that the GABAR agonist prevented the induction of apoptosis. Measurement of 4-4-HNE levels confirmed OS-induced damage after OLT, and the GABAR agonist improved this damage. In the gammaH2AX, PI3K, Akt and antioxidant enzymes (SODs), ATM and H2AX were greatly increased after OLT, and were reduced by the GABAR agonist. In the multivariate analyses between multiple groups, histopathological assessment, aspartate aminotransferase level, immunohistological examinations for apoptotic induction and gammaH2AX showed statistical differences. CONCLUSIONS: A specific agonist demonstrated regulation of GABAR in vivo in the liver. This activation in vivo reduced OS after OLT via the ATM/H2AX pathway.


Asunto(s)
Agonistas del GABA/administración & dosificación , Antagonistas del GABA/administración & dosificación , Trasplante de Hígado/métodos , Daño por Reperfusión/prevención & control , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Bicuculina/administración & dosificación , Isquemia Fría/efectos adversos , Reparación del ADN/efectos de los fármacos , Histonas/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Masculino , Muscimol/administración & dosificación , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Endogámicas Lew , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Transducción de Señal/efectos de los fármacos , Isquemia Tibia/efectos adversos
13.
J Neurosurg ; 117(1): 162-75, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22577746

RESUMEN

OBJECT: The authors evaluated the extent to which the Subdural Pharmacotherapy Device (SPD), chronically implanted over the frontal cortex to perform periodic, localized muscimol-delivery/CSF removal cycles, affects overall behavior, motor performance, electroencephalography (EEG) activity, and blood and CSF neurochemistry in macaque monkeys. METHODS: Two monkeys were used to adjust methodology and 4 monkeys were subjected to comprehensive testing. Prior to surgery, the animals' behavior in a large test chamber was monitored, and the motor skills required to remove food pellets from food ports located on the walls of the chamber were determined. The monkeys underwent implantation of the subdural and extracranial SPD units. The subdural unit, a silicone strip integrating EEG electrodes and fluid-exchange ports, was positioned over the right frontal cortex. The control unit included a battery-powered, microprocessor-regulated dual minipump and radiofrequency module secured to the cranium. After implantation, the SPD automatically performed periodic saline or muscimol (1.0 mM) deliveries at 12-hour intervals, alternating with local CSF removals at 6-hour intervals. The antiepileptic efficacy of this muscimol concentration was verified by demonstrating its ability to prevent focal acetylcholine-induced seizures. During SPD treatment, the monkeys' behavior and motor performance were again monitored, and the power spectrum of their radiofrequency-transmitted EEG recordings was analyzed. Serum and CSF muscimol levels were measured with high-performance liquid chromatography electrochemical detection, and CSF protein levels were measured with turbidimetry. RESULTS: The SPD was well tolerated in all monkeys for up to 11 months. The behavioral study revealed that during both saline and muscimol SPD treatment, the monkeys could achieve the maximum motor performance of 40 food-pellet removals per session, as before surgery. The EEG study showed that local EEG power spectra were not affected by muscimol treatment with SPD. The neurochemical study demonstrated that the administration of 1.0 mM muscimol into the neocortical subarachnoid space led to no detectable levels of this compound in the blood and cisternal CSF, as measured 1-125 minutes after delivery. Total protein levels were within the normal range in the cisternal CSF, but protein levels in the cortical-site CSF were significantly higher than normal: 361 ± 81.6 mg/dl. Abrupt discontinuation of 3-month, periodic, subdural muscimol treatments induced withdrawal seizures, which could be completely prevented by gradually tapering off the subdural muscimol concentration from 1.0 mM to 0.12-0.03 mM over a period of 2 weeks. The monkeys' general health and weight were maintained. Infection occurred only in one monkey 9 months after surgery. CONCLUSIONS: Long-term, periodic, transmeningeal muscimol delivery with the SPD is essentially a safe procedure. If further improved and successfully adapted for use in humans, the SPD can be used for the treatment of intractable focal neocortical epilepsy affecting approximately 150,000 patients in the US.


Asunto(s)
Anticonvulsivantes/administración & dosificación , Anticonvulsivantes/uso terapéutico , Conducta Animal/efectos de los fármacos , Agonistas del GABA/administración & dosificación , Agonistas del GABA/uso terapéutico , Muscimol/administración & dosificación , Muscimol/uso terapéutico , Animales , Anticonvulsivantes/efectos adversos , Peso Corporal/fisiología , Proteínas del Líquido Cefalorraquídeo/análisis , Cromatografía Líquida de Alta Presión , Convulsivantes , Implantes de Medicamentos , Electroencefalografía , Electrofisiología , Epilepsia del Lóbulo Frontal/tratamiento farmacológico , Lóbulo Frontal/cirugía , Agonistas del GABA/efectos adversos , Macaca radiata , Masculino , Muscimol/efectos adversos , Procedimientos Neuroquirúrgicos/métodos , Convulsiones/prevención & control , Programas Informáticos , Espacio Subdural/fisiología , Espacio Subdural/cirugía , Síndrome de Abstinencia a Sustancias
14.
Brain Res Bull ; 88(6): 617-23, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22613773

RESUMEN

Previous studies have demonstrated that kainic acid (KA)-induced seizures can cause the enhancement of excitation and lead to neuronal death in rat hippocampus. Co-activation of the inhibitory GABA receptors can attenuate the excitatory JNK3 apoptotic signaling pathway via inhibiting the increased assembly of the GluR6-PSD-95-MLK3 signaling module induced by KA in epileptic rat hippocampal CA1 and CA3 regions. Caspase-3 is a cysteine protease located in both the cytoplasm and mitochondrial intermembrane space that is a central effector of many apoptotic pathways. We designed experiments to elucidate the underlying molecular mechanisms of procaspase-3 activation and neuroprotection of co-activation of GABA receptors against neuronal death induced by KA. In this study, we show that co-activation of GABA receptors can attenuate the Fas/FasL apoptotic signaling pathway and inhibit the increased of thioredoxin reductase activity induced by KA, subsequently inhibit the activation of procaspase-3 by diminishing the denitrosylation of its active-site thiol and decreasing the cleavage of the caspase-3 zymogen to its active subunits. These results indicate that co-activation of GABA receptors results in neuroprotection by preventing caspase-3 denitrosylation in KA-induced seizure of rats.


Asunto(s)
Baclofeno/uso terapéutico , Inhibidores de Caspasas , Convulsivantes/toxicidad , Agonistas de Receptores de GABA-A/uso terapéutico , Agonistas de Receptores GABA-B/uso terapéutico , Ácido Kaínico/toxicidad , Muscimol/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Neurotoxinas/toxicidad , Receptores de GABA/fisiología , Convulsiones/prevención & control , Animales , Apoptosis/efectos de los fármacos , Baclofeno/administración & dosificación , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/patología , Región CA3 Hipocampal/efectos de los fármacos , Región CA3 Hipocampal/metabolismo , Región CA3 Hipocampal/patología , Caspasa 3/metabolismo , Sinergismo Farmacológico , Quimioterapia Combinada , Activación Enzimática/efectos de los fármacos , Proteína Ligando Fas/metabolismo , Agonistas de Receptores de GABA-A/administración & dosificación , Agonistas de Receptores GABA-B/administración & dosificación , Masculino , Muscimol/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Nitrosación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de GABA/efectos de los fármacos , Convulsiones/inducido químicamente , Convulsiones/metabolismo , Compuestos de Sulfhidrilo/metabolismo , Reductasa de Tiorredoxina-Disulfuro/metabolismo , Receptor fas/metabolismo
15.
Pain ; 153(1): 129-141, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22054599

RESUMEN

Impairment of spinal GABAergic inhibition is demonstrated to contribute to pathologic chronic pain states. We investigated spinal and peripheral GABAergic regulation of incisional pain in rats. We found that intrathecal but not peripheral administration of muscimol (GABA-A receptor agonist) and baclofen (GABA-B receptor agonist) reduced mechanical and thermal hyperalgesia after plantar incision in rats. Nonevoked pain behavior after incision was unaffected by these agonists. Similarly, nociception in unincised rats was not reduced by the same dose of agonists. Thus, GABA-A and GABA-B receptors are involved in mediating incision-induced hyperalgesia (but not nonevoked pain). Intrathecal and systemic application of L-838,417, a subtype-selective benzodiazepine site agonist (α2, α3, α5), reduced mechanical and heat hyperalgesia after incision, indicating a role of these subunits in mediating incision-induced hyperalgesia. Interestingly, the effects of all agonists were more intense and prolonged on the day after surgery than on the day of incision. Similarly, spinally administered GABA-A and GABA-B antagonists increased pain behavior, again with a greater effect 1 day after incision. One possible explanation for this finding might be that an incision modulates GABA-mediated inhibition 1 day after incision. However, expression of GABA-A receptor subunits α2 and α3 and GABA-B receptor subunits within the dorsal horn of the spinal cord were unchanged after incision, indicating that receptor expression cannot explain a possible modulation of GABAergic inhibition after incision. Thus, other mechanisms need to be considered. In conclusion, GABA-A and GABA-B receptors are promising targets for postoperative, incisional pain in humans.


Asunto(s)
Dolor Crónico/tratamiento farmacológico , Hiperalgesia/tratamiento farmacológico , Dimensión del Dolor/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo , Animales , Baclofeno/administración & dosificación , Conducta Animal/efectos de los fármacos , Dolor Crónico/metabolismo , Hiperalgesia/metabolismo , Muscimol/administración & dosificación , Ratas , Ratas Sprague-Dawley , Médula Espinal/metabolismo
16.
Ann N Y Acad Sci ; 1233: 187-93, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21950992

RESUMEN

The visual and oculomotor systems of primates are immature at birth and sensitive to injury. If synergistic interactions between visual and oculomotor systems are compromised during the first months of life, disorders in eye alignment, gaze holding, and smooth pursuit (SP) follow. Here we consider some potential neural mechanisms supporting SP and associated vestibular ocular reflex (VOR) behavior in normal and strabismic monkeys. Experimental strabismus was created by prism goggle wearing or eye muscle surgery in rhesus monkeys (Macaca mulatta). SP and cancellation of the VOR were highly asymmetric in strabismic monkeys during monocular viewing conditions. Similar asymmetric SP and VOR cancellation could be produced in normal monkeys by delivering unilateral muscimol injections to the dorsolateral pontine nucleus (DLPN). We suggest that failure to develop balanced cortical-brainstem circuits in strabismus accounts for many of the components of infantile strabismus syndrome.


Asunto(s)
Seguimiento Ocular Uniforme/fisiología , Estrabismo/fisiopatología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Esotropía/etiología , Esotropía/fisiopatología , Macaca mulatta , Muscimol/administración & dosificación , Músculos Oculomotores/fisiopatología , Músculos Oculomotores/cirugía , Puente/efectos de los fármacos , Puente/fisiopatología , Seguimiento Ocular Uniforme/efectos de los fármacos , Reflejo Vestibuloocular/efectos de los fármacos , Reflejo Vestibuloocular/fisiología , Estrabismo/etiología
17.
Biol Psychiatry ; 68(3): 265-71, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20299008

RESUMEN

BACKGROUND: Nicotine is the principal component of tobacco smoke, resulting in addiction, and recent evidence suggests that damage to the insular cortex (insula) disrupts tobacco addiction in human smokers. However, the effect of an inactivation of this structure in an animal model of nicotine addiction has yet to be evaluated. METHODS: To study this question, we investigated the effects of reversible inactivation of the granular insula by local injection of a gamma-aminobutyric acid agonists mixture (baclofen/muscimol) on nicotine self-administration (SA) under fixed and progressive ratio and on reinstatement of nicotine seeking induced by nicotine priming or nicotine-associated cues in rats. We also evaluated the effects of granular insula inactivation on food SA and relapse as a control. RESULTS: The inactivation of the granular insula decreased nicotine SA under both fixed and progressive ratios without affecting the SA of food under the same schedules of reinforcement. This inactivation also prevented the reinstatement, after extinction, of nicotine seeking induced by nicotine-associated cues or nicotine priming without modifying the reinstatement of food seeking. CONCLUSIONS: Our study indicates that the integrity of the granular insula is necessary for exhibiting motivation to take nicotine and to relapse to nicotine seeking but not for consuming food pellets or to relapse for food seeking. Indeed, it might be interesting to study the effect of methods that are able to modulate the activity of the insula--such as repetitive transcranial magnetic stimulation or deep brain stimulation--on tobacco addiction and relapse in humans.


Asunto(s)
Conducta Adictiva/prevención & control , Corteza Cerebral/efectos de los fármacos , Agonistas del GABA/administración & dosificación , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Tabaquismo/prevención & control , Análisis de Varianza , Animales , Aprendizaje por Asociación/efectos de los fármacos , Baclofeno/administración & dosificación , Conducta Animal/efectos de los fármacos , Corteza Cerebral/citología , Corteza Cerebral/fisiología , Condicionamiento Clásico/efectos de los fármacos , Modelos Animales de Enfermedad , Combinación de Medicamentos , Extinción Psicológica/efectos de los fármacos , Masculino , Microinyecciones , Motivación/fisiología , Muscimol/administración & dosificación , Ratas , Esquema de Refuerzo , Autoadministración
18.
Am J Physiol Regul Integr Comp Physiol ; 298(1): R130-40, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19923355

RESUMEN

Previous studies suggest that sympathetic responses evoked from the preoptic area in anesthetized rats require activation of neurons in the dorsomedial hypothalamus. Disinhibition of neurons in the dorsomedial hypothalamus in conscious rats produces physiological and behavioral changes resembling those evoked by microinjection of muscimol, a GABA(A) receptor agonist and neuronal inhibitor, into the medial preoptic area. We tested the hypothesis that all of these effects evoked from the medial preoptic area are mediated through neurons in the dorsomedial hypothalamus by assessing the effect of bilateral microinjection of muscimol into the DMH on these changes. After injection of vehicle into the dorsomedial hypothalamus, injection of muscimol into the medial preoptic area elicited marked increases in heart rate, arterial pressure, body temperature, plasma ACTH, and locomotor activity and also increased c-Fos expression in the hypothalamic paraventricular nucleus, a region known to control the release of ACTH from the adenohypophysis. Prior bilateral microinjection of muscimol into the dorsomedial hypothalamus produced a modest depression of baseline heart rate and body temperature but completely abolished all changes evoked from the medial preoptic area. Microinjection of muscimol just anterior to the dorsomedial hypothalamus had no effect on autonomic and neuroendocrine changes evoked from the medial preoptic area. Thus, activity of neurons in the dorsomedial hypothalamus mediates a diverse array of physiological and behavioral responses elicited from the medial preoptic area, suggesting that the latter region represents an important source of inhibitory tone to key neurons in the dorsomedial hypothalamus.


Asunto(s)
Sistema Nervioso Autónomo/fisiología , Hipotálamo/fisiología , Actividad Motora/fisiología , Sistemas Neurosecretores/fisiología , Área Preóptica/fisiología , Hormona Adrenocorticotrópica/sangre , Animales , Presión Sanguínea/fisiología , Temperatura Corporal/fisiología , Agonistas del GABA/administración & dosificación , Agonistas del GABA/farmacología , Frecuencia Cardíaca/fisiología , Hipotálamo/efectos de los fármacos , Masculino , Microinyecciones , Modelos Animales , Muscimol/administración & dosificación , Muscimol/farmacología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley
19.
Neuroscience ; 161(3): 718-33, 2009 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-19336249

RESUMEN

Mu opioid receptor (MOR) signaling in the nucleus accumbens (NAcc) elicits marked increases in the consumption of palatable tastants. However, the mechanism and circuitry underlying this effect are not fully understood. Multiple downstream target regions have been implicated in mediating this effect but the role of the ventral pallidum (VP), a primary target of NAcc efferents, has not been well defined. To probe the mechanisms underlying increased consumption, we identified behavioral changes in rats' licking patterns following NAcc MOR stimulation. Because the temporal structure of licking reflects the physiological substrates modulating consumption, these measures provide a useful tool in dissecting the cause of increased consumption following NAcc MOR stimulation. Next, we used a combination of pharmacological inactivation and lesions to define the role of the VP in hyperphagia following infusion of the MOR-specific agonist [D-Ala2, N-MePhe4, Gly-ol]-enkephalin (DAMGO) into the NAcc. In agreement with previous studies, results from lick microstructure analysis suggest that NAcc MOR stimulation augments intake through a palatability-driven mechanism. Our results also demonstrate an important role for the VP in normal feeding behavior: pharmacological inactivation of the VP suppresses baseline and NAcc DAMGO-induced consumption. However, this interaction does not occur through a serial circuit requiring direct projections from the NAcc to the VP. Rather, our results indicate that NAcc and VP circuits converge on a common downstream target that regulates food intake.


Asunto(s)
Ingestión de Alimentos/fisiología , Globo Pálido/fisiología , Núcleo Accumbens/fisiología , Receptores Opioides mu/metabolismo , Animales , Cateterismo , Ingestión de Alimentos/efectos de los fármacos , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Conducta Alimentaria/efectos de los fármacos , Conducta Alimentaria/fisiología , Agonistas del GABA/administración & dosificación , Globo Pálido/efectos de los fármacos , Hiperfagia/inducido químicamente , Hiperfagia/fisiopatología , Masculino , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Muscimol/administración & dosificación , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/fisiología , Neurotoxinas/toxicidad , Neurotransmisores/administración & dosificación , Núcleo Accumbens/efectos de los fármacos , Ácido Quinolínico/toxicidad , Distribución Aleatoria , Ratas , Ratas Long-Evans , Receptores Opioides mu/agonistas , Factores de Tiempo
20.
Braz. j. med. biol. res ; 42(1): 114-121, Jan. 2009. ilus
Artículo en Inglés | LILACS | ID: lil-505427

RESUMEN

We investigated the involvement of GABAergic mechanisms of the central amygdaloid nucleus (CeA) in unanesthetized rats subjected to acute isotonic or hypertonic blood volume expansion (BVE). Male Wistar rats bearing cannulas unilaterally implanted in the CeA were treated with vehicle, muscimol (0.2 nmol/0.2 µL) or bicuculline (1.6 nmol/0.2 µL) in the CeA, followed by isotonic or hypertonic BVE (0.15 or 0.3 M NaCl, 2 mL/100 g body weight over 1 min). The vehicle-treated group showed an increase in sodium excretion, urinary volume, plasma oxytocin (OT), and atrial natriuretic peptide (ANP) levels compared to control rats. Muscimol reduced the effects of BVE on sodium excretion (isotonic: 2.4 ± 0.3 vs vehicle: 4.8 ± 0.2 and hypertonic: 4.0 ± 0.7 vs vehicle: 8.7 ± 0.6 µEq·100 g-1·40 min-1); urinary volume after hypertonic BVE (83.8 ± 10 vs vehicle: 255.6 ± 16.5 µL·100 g-1·40 min-1); plasma OT levels (isotonic: 15.3 ± 0.6 vs vehicle: 19.3 ± 1 and hypertonic: 26.5 ± 2.6 vs vehicle: 48 ± 3 pg/mL), and ANP levels (isotonic: 97 ± 12.8 vs vehicle: 258.3 ± 28.1 and hypertonic: 160 ± 14.6 vs vehicle: 318 ± 16.3 pg/mL). Bicuculline reduced the effects of isotonic or hypertonic BVE on urinary volume and ANP levels compared to vehicle-treated rats. However, bicuculline enhanced the effects of hypertonic BVE on plasma OT levels. These data suggest that CeA GABAergic mechanisms are involved in the control of ANP and OT secretion, as well as in sodium and water excretion in response to isotonic or hypertonic blood volume expansion.


Asunto(s)
Animales , Masculino , Ratas , Amígdala del Cerebelo/efectos de los fármacos , Bicuculina/farmacología , Volumen Sanguíneo/efectos de los fármacos , Agonistas del GABA/farmacología , Antagonistas del GABA/farmacología , Muscimol/farmacología , Amígdala del Cerebelo/fisiología , Factor Natriurético Atrial/sangre , Bicuculina/administración & dosificación , Volumen Sanguíneo/fisiología , Diuresis/efectos de los fármacos , Diuresis/fisiología , Agonistas del GABA/administración & dosificación , Antagonistas del GABA/administración & dosificación , Muscimol/administración & dosificación , Oxitocina/sangre , Ratas Wistar , Sodio/orina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA