Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Integr Neurosci ; 20(3): 613-622, 2021 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-34645094

RESUMEN

As a gamma-aminobutyric acid type A receptor agonist sevoflurane is a common general anesthetic used in anesthesia and affects the neural development in offspring. We hypothesized that sevoflurane could regulate interneurons via the neuregulin-1-epidermal growth factor receptor-4 (NRG1-ErbB4) pathway in the entorhinal cortex (ECT) of the middle pregnancy. Six female rats in middle pregnancy (14.5 days of pregnancy) were randomly and equally divided into sevoflurane (SeV) and control groups. The rats in the SeV group were exposed to 4% sevoflurane for 3 hours. The expression levels of NRG1 and ErbB4, parvalbumin (PV) and glutamic acid decarboxylase (GAD67), and N-methyl-D-aspartate receptor subunit 2A (NR2A) and subunit 2B (NR2B) in offspring were examined through immunohistochemistry. The pyramidal neurons in the ECT were examined via Golgi staining. The levels of NRG1 and ErbB4 were significantly decreased (P < 0.01) and the levels of PV and GAD67 (interneurons) were found to be decreased in the SeV group (P < 0.01). The level of NR2B was found to be increased while the level of NR2A being decreased in the SeV group (P < 0.01). The development of pyramidal neurons was abnormal in the SeV group (P < 0.05). Conclusively, prenatal sevoflurane exposure could lead to the disturbance of the interneurons by activating the NRG1-ErbB4 pathway and subsequently result in abnormal development of pyramidal neurons in middle pregnancy. Prenatal sevoflurane exposure in middle pregnancy could be potentially harmful to the neural development of rat offspring. This study may reveal a novel pathway in the influence mechanism of sevoflurane on rat offspring.


Asunto(s)
Corteza Entorrinal/efectos de los fármacos , Agonistas de Receptores de GABA-A/farmacología , Interneuronas/efectos de los fármacos , Neurregulina-1/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Células Piramidales/efectos de los fármacos , Receptor ErbB-4/efectos de los fármacos , Sevoflurano/farmacología , Animales , Modelos Animales de Enfermedad , Femenino , Agonistas de Receptores de GABA-A/administración & dosificación , Embarazo , Ratas , Sevoflurano/administración & dosificación
2.
Neuropsychopharmacol Rep ; 41(1): 14-25, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33259705

RESUMEN

AIMS: Benzodiazepine receptor agonists (BZ-RAs) are frequently prescribed to treat insomnia; however, their long-term use is not recommended. To introduce an appropriate pharmaco-therapy, the current state and background factors of BZ-RAs' dependence must be elucidated. In this study, we developed a Japanese version of the Benzodiazepine Dependence Self-Report Questionnaire (Bendep-SRQ-J) and conducted a study of BZ-RAs' use disorder. METHODS: The Bendep-SRQ-J was created with permission from the original developer. Subjects were inpatients and outpatients receiving BZ-RAs between 2012 and 2013. Clinical data collected were Bendep-SRQ-J scores, sleep disorders for which BZ-RAs were prescribed, physical comorbidities, psychotropic drugs, and lifestyle factors. Logistic analysis was performed to extract factors associated with severe symptoms. RESULTS: Of the 707 patients prescribed BZ-RAs, 324 had voluntarily tapered or discontinued their drugs. Logistic analysis showed that the total number of drugs administered in the last 6 months correlated with both worsening of symptoms or conditions. This was more notable among younger patients, and the proportion of patients with severe symptoms or conditions increased with the increasing number of drugs. CONCLUSION: Using the Bendep-SRQ-J, we elucidated the current state of BZ-RA dependence. Nearly half of the patients were non-compliant. The proportion of patients with severe symptoms or disease conditions increased with the increase in the number of drugs administered. These findings highlight the need for clinicians to be aware of the likelihood of benzodiazepine dependence, especially in young patients and patients prescribed multiple hypnotics.


Asunto(s)
Ansiolíticos/administración & dosificación , Benzodiazepinas/administración & dosificación , Reducción Gradual de Medicamentos , Agonistas de Receptores de GABA-A/administración & dosificación , Hipnóticos y Sedantes/administración & dosificación , Trastornos Mentales/tratamiento farmacológico , Cooperación del Paciente , Polifarmacia , Psicometría/instrumentación , Trastornos Relacionados con Sustancias/diagnóstico , Adulto , Anciano , Anciano de 80 o más Años , Estudios Transversales , Reducción Gradual de Medicamentos/estadística & datos numéricos , Femenino , Encuestas Epidemiológicas , Humanos , Japón/epidemiología , Masculino , Trastornos Mentales/epidemiología , Persona de Mediana Edad , Cooperación del Paciente/estadística & datos numéricos , Autoinforme , Índice de Severidad de la Enfermedad , Síndrome de Abstinencia a Sustancias/diagnóstico , Síndrome de Abstinencia a Sustancias/epidemiología , Trastornos Relacionados con Sustancias/epidemiología , Adulto Joven
3.
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
4.
Sci Rep ; 10(1): 11401, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32647241

RESUMEN

Urodynamic studies, used to understand bladder function, diagnose bladder disease, and develop treatments for dysfunctions, are ideally performed with awake subjects. However, in small and medium-sized animal models, anesthesia is often required for these procedures and can be a research confounder. This study compared the effects of select survival agents (dexmedetomidine, alfaxalone, and propofol) on urodynamic (Δpressure, bladder capacity, bladder compliance, non-voiding contractions, bladder pressure slopes) and anesthetic (change in heart rate [∆HR], average heart rate [HR], reflexes, induction/recovery times) parameters in repeated cystometrograms across five adult male cats. The urodynamic parameters under isoflurane and α-chloralose were also examined in terminal procedures for four cats. Δpressure was greatest with propofol, bladder capacity was highest with α-chloralose, non-voiding contractions were greatest with α-chloralose. Propofol and dexmedetomidine had the highest bladder pressure slopes during the initial and final portions of the cystometrograms respectively. Cats progressed to a deeper plane of anesthesia (lower HR, smaller ΔHR, decreased reflexes) under dexmedetomidine, compared to propofol and alfaxalone. Time to induction was shortest with propofol, and time to recovery was shortest with dexmedetomidine. These agent-specific differences in urodynamic and anesthetic parameters in cats will facilitate appropriate study-specific anesthetic choices.


Asunto(s)
Anestésicos/farmacología , Gatos/fisiología , Urodinámica/efectos de los fármacos , Periodo de Recuperación de la Anestesia , Anestésicos/administración & dosificación , Animales , Cloralosa/farmacología , Dexmedetomidina/administración & dosificación , Dexmedetomidina/farmacología , Relación Dosis-Respuesta a Droga , Agonistas de Receptores de GABA-A/administración & dosificación , Agonistas de Receptores de GABA-A/farmacología , Frecuencia Cardíaca/efectos de los fármacos , Isoflurano/farmacología , Masculino , Modelos Animales , Contracción Muscular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Pregnanodionas/administración & dosificación , Pregnanodionas/farmacología , Presión , Propofol/administración & dosificación , Propofol/farmacología , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/fisiología
5.
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
7.
Res Social Adm Pharm ; 14(1): 18-25, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28214150

RESUMEN

BACKGROUND: Although polypharmacy is associated with significant morbidity, deprescribing can be challenging. In particular, clinicians express difficulty with their ability to deprescribe (i.e. reduce or stop medications that are potentially inappropriate). Evidence-based deprescribing guidelines are designed to help clinicians take action on reducing or stopping medications that may be causing more harm than benefit. OBJECTIVES: Determine if implementation of evidence-based guidelines increases self-efficacy for deprescribing proton pump inhibitor (PPI), benzodiazepine receptor agonist (BZRA) and antipsychotic (AP) drug classes. METHODS: A deprescribing self-efficacy survey was developed and administered to physicians, nurse practitioners and pharmacists at 3 long-term care (LTC) and 3 Family Health Teams in Ottawa, Canada at baseline and approximately 6 months after sequential implementation of each guideline. For each drug class, overall and domain-specific self-efficacy mean scores were calculated. The effects of implementation of each guideline on self-efficacy were tested by estimating the difference in scores using paired t-test. A linear mixed-effects model was used to investigate change over time and over practice sites. RESULTS: Of eligible clinicians, 25, 21, 18 and 13 completed the first, second, third and fourth survey respectively. Paired t-tests compared 14 participants for PPI and BZRA, and 9 for AP. Overall self-efficacy score increased for AP only (95% confidence intervals (CI) 0.32 to 19.79). Scores for domain 2 (develop a plan to deprescribe) increased for PPI (95% CI 0.52 to 24.12) and AP guidelines (95% CI 2.46 to 18.11); scores for domain 3 (implement the plan for deprescribing) increased for the PPI guideline (95% CI 0.55 to 14.24). Longitudinal analysis showed an increase in non-class specific scores, with a more profound effect for clinicians in LTC where guidelines were routinely used. CONCLUSION: Implementation of evidence-based deprescribing guidelines appears to increase clinicians' self-efficacy in developing and implementing a deprescribing plan for specific drug classes.


Asunto(s)
Deprescripciones , Enfermeras Practicantes/estadística & datos numéricos , Farmacéuticos/estadística & datos numéricos , Médicos/estadística & datos numéricos , Adulto , Anciano , Antipsicóticos/administración & dosificación , Antipsicóticos/efectos adversos , Práctica Clínica Basada en la Evidencia , Femenino , Agonistas de Receptores de GABA-A/administración & dosificación , Agonistas de Receptores de GABA-A/efectos adversos , Encuestas de Atención de la Salud , Humanos , Masculino , Persona de Mediana Edad , Ontario , Polifarmacia , Guías de Práctica Clínica como Asunto , Inhibidores de la Bomba de Protones/administración & dosificación , Inhibidores de la Bomba de Protones/efectos adversos , Autoeficacia
8.
Neuroscience ; 346: 349-364, 2017 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-28126369

RESUMEN

Painful events early in life have been shown to increase the incidence of interstitial cystitis/painful bladder syndrome in adulthood. However, the intrinsic mechanism is not well studied. We previously reported that neonatal bladder inflammation causes chronic visceral hypersensitivity along with molecular disruption of spinal GABAergic system in rats. The present study investigates whether these molecular changes affect the integrative function and responses of bladder-sensitive primary afferent and spinal neurons. Neonatal bladder inflammation was induced by intravesicular injection of zymosan during postnatal (P) days 14-16. In adulthood (P60), the viscero-motor response (VMR) to visceral stimuli was significantly inhibited by intrathecal (i.t) HZ166 (GABAAα-2 agonist) only in neonatally saline-treated, but not in neonatally zymosan-treated rats. HZ166 significantly inhibited the responses of bladder-responsive lumbosacral (LS) spinal neurons to urinary bladder distension (UBD) and slow infusion (SI) in neonatally saline-treated rats. Similar results were also observed in naïve adult rats where HZ166 produced significant inhibition of bladder-responsive spinal neurons. However, HZ166 did not inhibit responses of UBD-responsive spinal neurons from neonatally zymosan-treated rats. The drug did not attenuate the responses of UBD-sensitive pelvic nerve afferent (PNA) fibers to UBD and SI in either group of rats tested. Immunohistochemical studies showed a significantly lower level of GABAAα-2 receptor expression in the LS spinal cord of neonatally zymosan-treated rats compared to saline-treated rats. These findings indicate that neonatal bladder inflammation leads to functional and molecular alteration of spinal GABAAα-2 receptor subtypes, which may result in chronic visceral hyperalgesia in adulthood.


Asunto(s)
Cistitis Intersticial/fisiopatología , Neuronas/fisiología , Receptores de GABA-A/fisiología , Médula Espinal/fisiopatología , Dolor Visceral/fisiopatología , Animales , Animales Recién Nacidos , Benzodiazepinas/administración & dosificación , Colon/fisiopatología , Cistitis Intersticial/inducido químicamente , Cistitis Intersticial/complicaciones , Femenino , Agonistas de Receptores de GABA-A/administración & dosificación , Imidazoles/administración & dosificación , Región Lumbosacra , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/metabolismo , Médula Espinal/metabolismo , Vejiga Urinaria/inervación , Vejiga Urinaria/fisiopatología , Dolor Visceral/complicaciones , Zimosan
9.
PM R ; 9(8): 831-833, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28093373

RESUMEN

Patients with disorders of consciousness (DOC) have profound functional limitations with few treatment options for improving arousal and quality of life. Zolpidem is a nonbenzodiazepine hypnotic used to treat insomnia that has also been observed to paradoxically improve arousal in those with DOC, such as the vegetative or minimally conscious states. Little information exists on its use in patients with DOC who have intracranial space-occupying lesions. We present a case of a 24-year-old man in a minimally conscious state due to central nervous system lymphoma who was observed to have increased arousal and improved motor function after the administration of zolpidem. LEVEL OF EVIDENCE: V.


Asunto(s)
Trastornos de la Conciencia/diagnóstico por imagen , Trastornos de la Conciencia/tratamiento farmacológico , Agonistas de Receptores de GABA-A/administración & dosificación , Estado Vegetativo Persistente/tratamiento farmacológico , Piridinas/administración & dosificación , Nivel de Alerta/efectos de los fármacos , Neoplasias Encefálicas/complicaciones , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/patología , Trastornos de la Conciencia/etiología , Estudios de Seguimiento , Humanos , Imagen por Resonancia Magnética/métodos , Masculino , Estado Vegetativo Persistente/etiología , Estado Vegetativo Persistente/fisiopatología , Medición de Riesgo , Índice de Severidad de la Enfermedad , Resultado del Tratamiento , Adulto Joven , Zolpidem
10.
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
11.
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
12.
Neuroscience ; 293: 123-35, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25766937

RESUMEN

A GABA-site agonist gaboxadol (4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridin-3-ol) at 3 mg/kg induces strong anxiolytic response in a transgenic Thy1α6 mouse line ectopically expressing the GABA(A) receptor α6 subunit gene under the Thy-1.2 promoter. Now, we compared brain activation patterns between Thy1α6 and wild-type mice to identify brain structures potentially mediating this anxiolytic response. Acutely efficient anxiolytics such as benzodiazepines typically depress most brain regions while activating specifically neurons within the central extended amygdala. Gaboxadol treatment (3 mg/kg, i.p., 2 h) induced a significant increase in c-Fos expression selectively in many Thy1α6 brain regions including the limbic cortex, anterior olfactory nucleus, septal area and central and basolateral nuclei of amygdala. It failed to activate the lateral part of mediodorsal thalamic nucleus (MDL) in the Thy1α6 mice that was activated in the wild-type mice. Detailed mapping of the α6 subunit mRNA by in situ hybridization revealed expression in the middle layers of the isocortex, olfactory areas, hippocampal formation and basolateral nucleus of amygdala (BLA) in the Thy1α6 forebrain. The ligand autoradiographies (t-butylbicyclophosphoro[(35)S]thionate ([(35)S]TBPS) and [(3)H]Ro 15-4513) revealed high levels of pharmacologically active extrasynaptic α6ß and α6ßγ2 GABA(A) receptors in these same areas. However, c-Fos induction by gaboxadol treatment in Thy1α6 brain was not restricted to areas highly expressing the α6-containing GABA(A) receptors suggesting that indirect pathways lead to the paradoxically widespread activation. Interestingly, the activation pattern by gaboxadol at the dose that is anxiolytic in Thy1α6 mice resembled closely that observed after various fear- and stress-provoking challenges. However, our results are consistent with a recent observation that optogenetic activation of specific neuronal pathways in the extended amygdala mediates anxiolytic responses. Our results suggest that the widespread neuronal inhibition as typically associated with benzodiazepines is not the exclusive mechanism of anxiolysis.


Asunto(s)
Agonistas de Receptores de GABA-A/administración & dosificación , Isoxazoles/administración & dosificación , Inhibición Neural/efectos de los fármacos , Prosencéfalo/metabolismo , Receptores de GABA-A/metabolismo , Animales , Ansiolíticos/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Prosencéfalo/diagnóstico por imagen , Prosencéfalo/efectos de los fármacos , Subunidades de Proteína/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Cintigrafía
13.
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
14.
Physiol Res ; 62(3): 285-95, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23489191

RESUMEN

Clinical studies showed that GABA(B) receptor agonists improve symptoms in patients with gastroesophageal reflux disease. One proposed mechanism of this effect is direct inhibition of the gastroesophageal vagal tension mechanosensors by GABA(B) agonists leading to reduction of reflux. In addition to tension mechanosensors, the vagal nodose ganglion supplies the esophagus with nociceptive C-fibers that likely contribute to impairment of esophageal reflex regulation in diseases. We hypothesized that GABA(B) agonists inhibit mechanically-induced activation of vagal esophageal nodose C-fibers in baseline and/or in sensitized state induced by inflammatory mediators. Ex vivo extracellular recordings were made from the esophageal nodose C-fibers in the isolated vagally-innervated guinea pig esophagus. We found that the selective GABA(B) agonist baclofen (100-300 microM) did not inhibit activation of esophageal nodose C-fibers evoked by esophageal distention (10-60 mmHg). The mechanical response of esophageal nodose C-fibers can be sensitized by different pathways including the stimulation of the histamine H(1) receptor and the stimulation the adenosine A(2A) receptor. Baclofen failed to inhibit mechanical sensitization of esophageal nodose C-fibers induced by histamine (100 microM) or the selective adenosine A(2A) receptor agonist CGS21680 (3 nM). Our data suggest that the direct mechanical inhibition of nodose C-fibers in the esophagus is unlikely to contribute to beneficial effects of GABA(B) agonists in patients with esophageal diseases.


Asunto(s)
Vías Aferentes/fisiología , Baclofeno/administración & dosificación , Esófago/inervación , Esófago/fisiología , Agonistas de Receptores de GABA-A/administración & dosificación , Inhibición Neural/fisiología , Ganglio Nudoso/fisiología , Vías Aferentes/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Esófago/efectos de los fármacos , Cobayas , Masculino , Inhibición Neural/efectos de los fármacos , Ganglio Nudoso/efectos de los fármacos
15.
Gut ; 62(9): 1248-55, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22730470

RESUMEN

OBJECTIVE: Lesogaberan (AZD3355) is a novel γ-aminobutyric acid B-type receptor agonist designed to treat gastro-oesophageal reflux disease (GERD) by inhibiting transient lower oesophageal sphincter relaxations. A randomised, double-blind, placebo-controlled, multi-centre phase IIb study was performed to assess the efficacy and safety of lesogaberan as an add-on to proton pump inhibitor (PPI) therapy in patients with GERD who are partially responsive to PPI therapy (ClinicalTrials.gov reference: NCT01005251). DESIGN: In total, 661 patients were randomised to receive 4 weeks of placebo or 60, 120, 180 or 240 mg of lesogaberan twice daily, in addition to ongoing PPI therapy. Symptoms were measured using the Reflux Symptom Questionnaire electronic Diary. Response to treatment was defined as having an average of ≥ 3 additional days per week of not more than mild GERD symptoms during treatment compared with baseline. RESULTS: In the primary analysis, 20.9%, 25.6%, 23.5% and 26.2% of patients responded to the 60, 120, 180 and 240 mg twice daily lesogaberan doses, respectively, and 17.9% responded to placebo. The response to the 240 mg twice daily dose was statistically significantly greater than the response to placebo using a one-sided test at the predefined significance level of p < 0.1. However, the absolute increases in the proportions of patients who responded to lesogaberan compared with placebo were low. Lesogaberan was generally well tolerated, although six patients receiving lesogaberan developed reversible elevated alanine transaminase levels. CONCLUSIONS: In patients with GERD symptoms partially responsive to PPI therapy, lesogaberan was only marginally superior to placebo in achieving an improvement in symptoms.


Asunto(s)
Esfínter Esofágico Inferior/efectos de los fármacos , Reflujo Gastroesofágico/tratamiento farmacológico , Ácidos Fosfínicos/administración & dosificación , Propilaminas/administración & dosificación , Inhibidores de la Bomba de Protones/administración & dosificación , Adulto , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Monitoreo de Drogas , Quimioterapia Combinada , Esfínter Esofágico Inferior/fisiopatología , Femenino , Agonistas de Receptores de GABA-A/administración & dosificación , Reflujo Gastroesofágico/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , Resultado del Tratamiento
16.
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
17.
Eur Neuropsychopharmacol ; 21(4): 316-24, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20627472

RESUMEN

The activity of dentate gyrus granule neurons is under a strong GABAergic tonic inhibitory control which contributes to the sparse activation pattern of these neurons after environmental stimuli. Previously, we reported that in sparse dentate gyrus neurons such stimuli evoke Ser-10 (S10) phosphorylation and Lys-14 (K14) acetylation in the nucleosomal protein histone H3 (H3S10p-K14ac) resulting in the induction of c-Fos. We hypothesized that GABA is an important modulator of novelty stress-evoked epigenomic mechanisms in rat dentate neurons. As reported previously, exposure to novelty (30min in new cage) evoked a significant increase in H3S10p-K14ac-and c-Fos-positive neuron numbers in the dentate gyrus. Pre-treatment of rats with the benzo Lorazepam, an indirect GABA-A receptor agonist, had no effects on baseline levels of H3S10p-K14ac and c-Fos but dose-dependently inhibited the novelty-induced epigenomic effects. At the applied doses (0.1-0.3mg/kg), Lorazepam's effects on behavior were mainly anxiolytic-like. To examine the effects of attenuated GABAergic inhibition on dentate granule neurons we applied the partial inverse GABA-A agonist FG-7142. This drug profoundly enhanced baseline levels as well as novelty-induced increases in the number of H3S10p-K14ac- and c-Fos-positive dentate neurons. Furthermore, FG-7142 evoked behavior in the novel cage congruous with increased anxiety and hyper-vigilance. Interestingly, the FG-7142-evoked enhancements in epigenomic changes were completely blocked by the NMDA receptor antagonist MK-801. We conclude that GABA tonically controls epigenomic responses to psychologically salient events in dentate gyrus granule neurons. Furthermore, GABA appears to exert its controller activity through modulation of NMDA receptor function. These findings may be of significance for the elucidation of anxiety disorders especially PTSD.


Asunto(s)
Giro Dentado/efectos de los fármacos , Conducta Exploratoria/efectos de los fármacos , Histonas/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Estrés Psicológico/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Acetilación/efectos de los fármacos , Animales , Ansiolíticos/administración & dosificación , Ansiolíticos/uso terapéutico , Ansiedad/inducido químicamente , Ansiedad/tratamiento farmacológico , Ansiedad/metabolismo , Conducta Animal/efectos de los fármacos , Recuento de Células , Giro Dentado/metabolismo , Giro Dentado/patología , Relación Dosis-Respuesta a Droga , Agonismo Inverso de Drogas , Agonistas de Receptores de GABA-A/administración & dosificación , Agonistas de Receptores de GABA-A/uso terapéutico , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Ratas Wistar , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Estrés Psicológico/tratamiento farmacológico
18.
Gynecol Endocrinol ; 27(12): 1087-93, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21190418

RESUMEN

Animal studies suggest regulatory effects on the hypothalamic-pituitary-gonad axis by allopregnanolone, an endogenous gamma-aminobutyric acid A (GABA(A)) receptor agonist. Elevated levels of allopregnanolone in women with hypothalamic amenorrhea have been seen. Isoallopregnanolone is an isomer to allopregnanolone, but without GABA(A) receptor effects. The purpose of this study was to investigate effects of allopregnanolone and isoallopregnanolone on gonadotropin levels in healthy women of fertile age. Ten women were given allopregnanolone and five women isoallopregnanolone intravenously in follicular phase. Repeated blood samples were drawn during the test day. Main outcomes were changes in serum levels of follicle-stimulating hormone (FSH), luteinising hormone (LH), oestradiol, and progesterone. Serum-FSH decreased between 5 and 105 min after the allopregnanolone injection (F(16,144)=2.18, p=0.008). Serum-LH was reduced between 5 and 35 min following the allopregnanolone injection (F(16,144)=2.63, p=0.001). Serum-oestradiol and -progesterone were not significantly changed after allopregnanolone injections. No effect on gonadotropin levels were seen after administration of isoallopregnanolone. Allopregnanolone reduces FSH and LH levels in women and the effect might be mediated via a specific GABA(A) receptor activation since isoallopregnanolone lacked this effect. Although the number of women was small, the results suggest a regulatory mechanism on the hypothalamic-pituitary-gonadal axis by allopregnanolon.


Asunto(s)
Gonadotropinas/sangre , Pregnanolona/farmacología , Adulto , Regulación hacia Abajo/efectos de los fármacos , Estradiol/sangre , Femenino , Hormona Folículo Estimulante/sangre , Fase Folicular/sangre , Fase Folicular/efectos de los fármacos , Agonistas de Receptores de GABA-A/administración & dosificación , Agonistas de Receptores de GABA-A/farmacocinética , Agonistas de Receptores de GABA-A/farmacología , Humanos , Inyecciones Intravenosas , Hormona Luteinizante/sangre , Proyectos Piloto , Pregnanolona/administración & dosificación , Pregnanolona/análogos & derivados , Pregnanolona/farmacocinética , Progesterona/sangre , Factores de Tiempo , Adulto Joven
19.
Pharmacol Biochem Behav ; 98(1): 81-6, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21182854

RESUMEN

Gamma aminobutyric acid (GABA)-A receptor modulators constitute the majority of clinically relevant sedative-hypnotics. Zolpidem (Ambien) is a nonbenzodiazepine GABA-A receptor modulator that binds with high affinity to GABA-A receptors expressing alpha-1 subunits. The present study examined the effects of a new approach to the oral administration of zolpidem on locomotor activity, body weight, food intake, relative food intake, feed efficiency, anxiety, and visceral adiposity in rats. Effects of withdrawal associated with cessation of the drug were also recorded. A daily chronically administered oral 10 mg/kg dose of zolpidem caused a decrease in locomotor activity, an increase in food intake and relative food intake, and a more positive feed efficiency during the drug-administration period. Anxiety and visceral adiposity also increased in animals receiving the drug. During withdrawal of zolpidem, there was a decrease in body weight, food intake, relative food intake, and anxiety, as well as a negative feed efficiency. These results suggest that zolpidem can modulate locomotor activity, metabolism, and anxiety-related behavior. A highly positive feed efficiency and increased visceral adiposity associated with zolpidem intake were unique findings of this study.


Asunto(s)
Ansiedad/inducido químicamente , Actividad Motora/efectos de los fármacos , Piridinas/farmacología , Adiposidad/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Agonistas de Receptores de GABA-A/administración & dosificación , Agonistas de Receptores de GABA-A/farmacología , Agonistas de Receptores de GABA-A/toxicidad , Humanos , Hipnóticos y Sedantes/administración & dosificación , Hipnóticos y Sedantes/farmacología , Hipnóticos y Sedantes/toxicidad , Masculino , Modelos Animales , Piridinas/administración & dosificación , Piridinas/toxicidad , Ratas , Zolpidem
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA