Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 156
Filtrar
1.
J Exp Biol ; 223(Pt 4)2020 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-31988165

RESUMEN

The physiological roles of corticotropin-releasing factor (CRF) have recently been extended to cytoprotection. Here, to determine whether CRF is neuroprotective in fish, the effects of CRF against high environmental ammonia (HEA)-mediated neurogenic impairment and cell death were investigated in zebrafish. In vivo, exposure of 1 day post-fertilization (dpf) embryos to HEA only reduced the expression of the determined neuron marker neurod1 In contrast, in 5 dpf larvae, HEA increased the expression of nes and sox2, neural progenitor cell markers, and reduced the expression of neurog1, gfap and mbpa, proneuronal cell, radial glia and oligodendrocyte markers, respectively, and neurod1 The N-methyl-d-aspartate (NMDA) receptor inhibitor MK801 rescued the HEA-induced reduction in neurod1 in 5 dpf larvae but did not affect the HEA-induced transcriptional changes in other neural cell types, suggesting that hyperactivation of NMDA receptors specifically contributes to the deleterious effects of HEA in determined neurons. As observed in vivo, HEA exposure elicited marked changes in the expression of cell type-specific markers in isolated 5 dpf larval brains. The addition of CRF reversed the in vitro effects of HEA on neurod1 expression and prevented an HEA-induced increase in cell death. Finally, the protective effects of CRF against HEA-mediated neurogenic impairment and cell death were prevented by the CRF type 1 receptor selective antagonist antalarmin. Together, these results provide novel evidence that HEA has developmental time- and cell type-specific neurotoxic effects, that NMDA receptor hyperactivation contributes to HEA-mediated impairment of determined neurons, and that CRF has neuroprotective properties in the larval zebrafish brain.


Asunto(s)
Amoníaco/toxicidad , Hormona Liberadora de Corticotropina/farmacología , Pez Cebra/embriología , Animales , Encéfalo/metabolismo , Muerte Celular/efectos de los fármacos , Maleato de Dizocilpina/farmacología , Regulación del Desarrollo de la Expresión Génica , Larva/efectos de los fármacos , Larva/metabolismo , Pirimidinas/farmacología , Pirroles/farmacología , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Pez Cebra/genética , Pez Cebra/metabolismo
2.
Addict Biol ; 25(3): e12769, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31099135

RESUMEN

Adolescence represents increased susceptibility to stress that increases risk for nicotine dependence. The present study examined the interactive effects of brief exposure to stress (shipping/transportation or experimentally induced) and chronic nicotine during adolescence on cognitive function and stress reactivity in adulthood. Adolescent (P31), but not young adult (P47), C57BL/6J mice had higher levels of corticosterone after shipping vs mice bred onsite. Shipped preadolescent (P23) and adolescent (P38) mice, but not those bred onsite, exposed to nicotine showed deficits in contextual fear learning when tested in adulthood. Adult learning deficits were replicated in adolescent mice bred onsite, exposed to experimentally induced stress, and administered chronic nicotine. Stress and nicotine during adolescence resulted in higher expression of hippocampal glucocorticoid receptors and corticotropin-releasing factor receptors and blunted restraint induced CORT release in adulthood. Importantly, studies examining adolescent behavior in mice should consider stress influences outcomes.


Asunto(s)
Cognición/efectos de los fármacos , Hipocampo/efectos de los fármacos , Aprendizaje/efectos de los fármacos , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Estrés Psicológico/fisiopatología , Animales , Cognición/fisiología , Corticosterona/metabolismo , Miedo , Hipocampo/fisiopatología , Aprendizaje/fisiología , Ratones , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Receptores de Glucocorticoides/efectos de los fármacos , Receptores de Glucocorticoides/metabolismo , Estrés Psicológico/metabolismo
3.
J Neurophysiol ; 122(6): 2636-2642, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31800349

RESUMEN

Gastric vagal afferent (GVA) sensing of food-related mechanical stimuli is a crucial mechanism in the control of feeding behavior and gastric function. Stress is an important factor contributing to eating disorders and gastric diseases. Chronic stress has been shown to increase the mechanosensitivity of GVAs in mice and to reduce food intake and body weight. Whether the mechanosensitivity of GVAs is modulated by stress hormones is not known. This study aimed to determine the effect of stress hormones on GVA mechanosensitivity. The expression of stress hormone receptors in GVA cell bodies was determined in 8-wk-old male C57BL/6 mice using quantitative RT-PCR combined with laser capture microdissection. The mechanosensitivity of GVAs was determined in the absence and presence of stress hormones using an in vitro single-fiber recording preparation. NR3C1 and CRHR2 (mRNA isoforms of glucocorticoid receptor and CRF2 receptor, respectively) were expressed in GVA neurons. The glucocorticoid receptor agonist corticosterone had no effect on the mechanosensitivity of either tension or mucosal GVAs. Activation of CRF2 receptor by its specific analog, urocortin 3, significantly increased the mechanosensitivity of both tension and mucosal GVAs, an effect prevented by the CRF2 receptor antagonist astressin 2B. In conclusion, activation of CRF2 receptor increases the mechanosensitivity of GVAs. This may contribute to the stress- and CRF2 receptor-associated changes in feeding behavior and gastric function, possibly contributing to the hypersensitivity of GVAs in chronic stress conditions.NEW & NOTEWORTHY Gastric vagal afferents (GVAs) relay food-related signals to the central nervous system, where they are processed, eventually leading to modulation of food intake and gastric function. GVA signaling can be modulated by an array of hormones. Stress has been shown to induce GVA hypersensitivity. This study demonstrates that GVA neurons express subtypes of stress hormone receptors, specifically CRF2. Furthermore, activation of CRF2 receptor increases GVA mechanosensitivity, which could have implications for food intake and gastric function.


Asunto(s)
Mecanorreceptores/fisiología , Neuronas Aferentes/fisiología , Receptores de Hormona Liberadora de Corticotropina/fisiología , Estómago/inervación , Nervio Vago/fisiología , Animales , Corticosterona/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos , Receptores de Glucocorticoides/metabolismo , Urocortinas/farmacología
4.
Cell Mol Neurobiol ; 38(3): 627-633, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28608000

RESUMEN

The anorexigenic molecule nesfatin-1 has recently been taken as a potential mood regulator, but the potential mechanisms remain unknown. Results of our previous study have demonstrated that nesfatin-1 could induce anxiety- and depression-like behaviors in rats, accompanied by the hyperactivity of the hypothalamic-pituitary-adrenal axis and the imbalanced mRNA expression of synaptic vesicle proteins. To explore the potential neurobiological mechanism underlying the effect of nesfatin-1 on the synaptic plasticity, the human neuroblastoma SH-SY5Y cells were cultured and treated with different concentrations of nesfatin-1 in the present study. The mRNA and protein expressions of corticotropin-releasing hormone (CRH) were measured via real-time fluorescent quantitative PCR and western blot, respectively. The protein expressions of extracellular signal-regulated kinase 1/2 (ERK1/2), phosphorylated-ERK1/2 (p-ERK1/2), and synapsin I were detected via western blot. The results confirmed that nesfatin-1 (10-9~10-7 mol/L) could up-regulate the expression of CRH. Moreover, nesfatin-1 (10-9~10-7 mol/L) could also increase the protein expressions of p-ERK1/2 and synapsin I, and these effects could be blocked by CP376395, a selective antagonist of CRH type 1 receptor (CRHR1). Furthermore, the increased expression of synapsin I induced by nesfatin-1 could also be reversed by PD98059, a specific inhibitor of the p-ERK. These results indicated that CRHR1 might mediate the effect of nesfatin-1 on the expressions of synapsin I via ERK1/2 signaling pathway.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al ADN/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas del Tejido Nervioso/metabolismo , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Sinapsinas/efectos de los fármacos , Aminopiridinas/farmacología , Línea Celular , Hormona Liberadora de Corticotropina/efectos de los fármacos , Hormona Liberadora de Corticotropina/metabolismo , Flavonoides/farmacología , Humanos , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Nucleobindinas , Fosforilación , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/metabolismo , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos , Sinapsinas/metabolismo , Regulación hacia Arriba/efectos de los fármacos
5.
J Neurosci ; 36(42): 10729-10741, 2016 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-27798128

RESUMEN

The corticotropin releasing factor (CRF) system in the central amygdala (CeA) has been implicated in the effects of acute ethanol and the development of alcohol dependence. We previously demonstrated that CRF receptor 1 (CRF1) neurons comprise a specific component of the CeA microcircuitry that is selectively engaged by acute ethanol. To investigate the impact of chronic ethanol exposure on inhibitory signaling in CRF1+ CeA neurons, we used CRF1:GFP mice subjected to chronic intermittent ethanol (CIE) inhalation and examined changes in local inhibitory control, the effects of acute ethanol, and the output of these neurons from the CeA. Following CIE, CRF1+ neurons displayed decreased phasic inhibition and a complete loss of tonic inhibition that persisted into withdrawal. CRF1- neurons showed a cell type-specific upregulation of both phasic and tonic signaling with CIE, the latter of which persists into withdrawal and is likely mediated by δ subunit-containing GABAA receptors. The loss of tonic inhibition with CIE was seen in CRF1+ and CRF1- neurons that project out of the CeA and into the bed nucleus of the stria terminalis. CRF1+ projection neurons displayed an increased baseline firing rate and loss of sensitivity to acute ethanol following CIE. These data demonstrate that chronic ethanol exposure produces profound and long-lasting changes in local inhibitory control of the CeA, resulting in an increase in the output of the CeA and the CRF1 receptor system, in particular. These cellular changes could underlie the behavioral manifestations of alcohol dependence and potentially contribute to the pathology of addiction. SIGNIFICANCE STATEMENT: The corticotropin releasing factor (CRF) system in the central amygdala (CeA) has been implicated in the effects of acute and chronic ethanol. We showed previously that CRF receptor 1-expressing (CRF1+) neurons in the CeA are under tonic inhibitory control and are differentially regulated by acute ethanol (Herman et al., 2013). Here we show that the inhibitory control of CRF1+ CeA neurons is lost with chronic ethanol exposure, likely by a functional switch in local tonic signaling. The loss of tonic inhibition is seen in CRF1+ projection neurons, suggesting that a critical consequence of chronic ethanol exposure is an increase in the output of the CeA CRF1 system, a neuroadaptation that may contribute to the behavioral consequences of alcohol dependence.


Asunto(s)
Núcleo Amigdalino Central/efectos de los fármacos , Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Vías Nerviosas/efectos de los fármacos , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos , Ácido gamma-Aminobutírico/fisiología , Animales , Técnicas In Vitro , Masculino , Ratones , Ratones Transgénicos , Técnicas de Placa-Clamp , Receptores de Hormona Liberadora de Corticotropina/genética , Receptores de GABA-A/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos
6.
J Neurosci ; 34(19): 6659-67, 2014 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-24806691

RESUMEN

The nature of neuroadaptations in the genesis of escalated cocaine taking remains a topic of considerable interest. Intermittent social defeat stress induces both locomotor and dopaminergic cross-sensitization to cocaine, as well as escalated cocaine self-administration. The current study examines the role of corticotropin releasing factor receptor subtypes 1 and 2 (CRFR1, CRFR2) within the ventral tegmental area (VTA) during social defeat stress. This study investigated whether injecting either a CRFR1 or CRFR2 antagonist directly into the VTA before each social defeat would prevent the development of later (1) locomotor sensitization, (2) dopaminergic sensitization, and (3) escalated cocaine self-administration in rats. CRFR1 antagonist CP376395 (50 or 500 ng/side), CRFR2 antagonist Astressin2-B (100 or 1000 ng/side), or vehicle (aCSF) was microinjected into the VTA 20 min before social defeat stress (or handling) on days 1, 4, 7, and 10. Ten days later, rats were injected with cocaine (10 mg/kg, i.p.) and assessed for either locomotor sensitization, measured by walking activity, or dopaminergic sensitization, measured by extracellular dopamine (DA) in the nucleus accumbens shell (NAcSh) through in vivo microdialysis. Locomotor sensitization testing was followed by intravenous cocaine self-administration. Intra-VTA antagonism of CRFR1, but not CRFR2, inhibited the induction of locomotor cross-sensitization to cocaine, whereas both prevented dopaminergic cross-sensitization and escalated cocaine self-administration during a 24 h "binge." This may suggest dissociation between locomotor sensitization and cocaine taking. These data also suggest that interactions between CRF and VTA DA neurons projecting to the NAcSh are essential for the development of dopaminergic cross-sensitization to cocaine.


Asunto(s)
Trastornos Relacionados con Cocaína/psicología , Cocaína/farmacología , Hormona Liberadora de Corticotropina/farmacología , Inhibidores de Captación de Dopamina/farmacología , Dopamina/farmacología , Medio Social , Estrés Psicológico/psicología , Área Tegmental Ventral/efectos de los fármacos , Aminopiridinas/farmacología , Animales , Conducta Animal/efectos de los fármacos , Cocaína/administración & dosificación , Inhibidores de Captación de Dopamina/administración & dosificación , Masculino , Microdiálisis , Microinyecciones , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Fragmentos de Péptidos/farmacología , Ratas , Ratas Long-Evans , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos , Autoadministración , Conducta Social , Abuso de Sustancias por Vía Intravenosa
7.
Am J Physiol Endocrinol Metab ; 307(10): E944-53, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25269482

RESUMEN

Energy expenditure is determined by metabolic rate and diet-induced thermogenesis. Normally, energy expenditure increases due to neural mechanisms that sense plasma levels of ingested nutrients/hormones and reflexively increase sympathetic nerve activity (SNA). Here, we investigated neural mechanisms of glucose-driven sympathetic activation by determining contributions of neuronal activity in the hypothalamic paraventricular nucleus (PVN) and activation of corticotropin-releasing factor (CRF) receptors in the rostral ventrolateral medulla (RVLM). Glucose was infused intravenously (150 mg/kg, 10 min) in male rats to raise plasma glucose concentration to a physiological postprandial level. In conscious rats, glucose infusion activated CRF-containing PVN neurons and TH-containing RVLM neurons, as indexed by c-Fos immunofluorescence. In α-chloralose/urethane-anesthetized rats, glucose infusion increased lumbar and splanchnic SNA, which was nearly prevented by prior RVLM injection of the CRF receptor antagonist astressin (10 pmol/50 nl). This cannot be attributed to a nonspecific effect, as sciatic afferent stimulation increased SNA and ABP equivalently in astressin- and aCSF-injected rats. Glucose-stimulated sympathoexcitation was largely reversed during inhibition of PVN neuronal activity with the GABA-A receptor agonist muscimol (100 pmol/50 nl). The effects of astressin to prevent glucose-stimulated sympathetic activation appear to be specific to interruption of PVN drive to RVLM because RVLM injection of astressin prior to glucose infusion effectively prevented SNA from rising and prevented any fall of SNA in response to acute PVN inhibition with muscimol. These findings suggest that activation of SNA, and thus energy expenditure, by glucose is initiated by activation of CRF receptors in RVLM by descending inputs from PVN.


Asunto(s)
Metabolismo Energético , Glucosa/metabolismo , Bulbo Raquídeo/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Sistema Nervioso Simpático/metabolismo , Animales , Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Hormona Liberadora de Corticotropina/farmacología , Agonistas de Receptores de GABA-A/farmacología , Masculino , Bulbo Raquídeo/efectos de los fármacos , Muscimol/farmacología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos , Sistema Nervioso Simpático/efectos de los fármacos
8.
Gastroenterology ; 145(6): 1253-61.e1-3, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23954313

RESUMEN

BACKGROUND & AIMS: Alterations in central corticotropin-releasing factor signaling pathways have been implicated in the pathophysiology of anxiety disorders and irritable bowel syndrome (IBS). We aimed to characterize the effects of the corticotropin-releasing factor receptor 1 (CRF-R1) antagonist, GW876008, on brain and skin conductance responses during acquisition and extinction of conditioned fear to the threat of abdominal pain in subjects with IBS and healthy individuals (controls). METHODS: We performed a single-center, randomized, double-blind, 3-period crossover study of 11 women with IBS (35.50 ± 12.48 years old) and 15 healthy women (controls) given a single oral dose (20 mg or 200 mg) of the CRF-R1 antagonist or placebo. Blood-oxygen level-dependent responses were analyzed using functional magnetic resonance imaging in a tertiary care setting. RESULTS: Controls had greater skin conductance responses during acquisition than extinction, validating the fear-conditioning paradigm. In contrast, during extinction, women with IBS had greater skin conductance responses than controls-an effect normalized by administration of a CRF-R1 antagonist. Although the antagonist significantly reduced activity in the thalamus in patients with IBS and controls during acquisition, the drug produced greater suppression of blood-oxygen level-dependent activity in a wide range of brain regions in IBS patients during extinction, including the medial prefrontal cortex, pons, hippocampus, and anterior insula. CONCLUSIONS: Although CRF signaling via CRF-R1 is involved in fear acquisition and extinction learning related to expected abdominal pain in patients with IBS and controls, this system appears to be up-regulated in patients with IBS. This up-regulation might contribute to the previously reported abnormal brain responses to expected abdominal pain.


Asunto(s)
Trastornos de Ansiedad/fisiopatología , Hormona Liberadora de Corticotropina/fisiología , Extinción Psicológica/fisiología , Síndrome del Colon Irritable/fisiopatología , Receptores de Hormona Liberadora de Corticotropina/fisiología , Transducción de Señal/fisiología , Dolor Abdominal/fisiopatología , Dolor Abdominal/psicología , Adulto , Trastornos de Ansiedad/psicología , Encéfalo/fisiología , Mapeo Encefálico , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Estudios Cruzados , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Miedo/fisiología , Miedo/psicología , Femenino , Respuesta Galvánica de la Piel/efectos de los fármacos , Respuesta Galvánica de la Piel/fisiología , Humanos , Persona de Mediana Edad , Pirazoles/farmacología , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
9.
Am J Physiol Regul Integr Comp Physiol ; 306(10): R722-7, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24598461

RESUMEN

Nesfatin-1 is produced in the periphery and in the brain where it has been demonstrated to regulate appetite, stress hormone secretion, and cardiovascular function. The anorexigenic action of central nesfatin-1 requires recruitment of neurons producing the melanocortins and centrally projecting oxytocin (OT) and corticotropin-releasing hormone (CRH) neurons. We previously have shown that two components of this pathway, the central melanocortin and oxytocin systems, contribute to the hypertensive action of nesfatin-1 as well. We hypothesized that the cardiovascular effect of nesfatin-1 also was dependent on activation of neurons expressing CRH receptors, and that the order of activation of the melanocortin-CRH-oxytocin circuit was preserved for both the anorexigenic and hypertensive actions of the peptide. Pretreatment of male rats with the CRH-2 receptor antagonist astressin2B abrogated nesfatin-1-induced increases in mean arterial pressure (MAP). Furthermore, the hypertensive action of CRH was blocked by pretreatment with an oxytocin receptor antagonist ornithine vasotocin (OVT), indicating that the hypertensive effect of nesfatin-1 may require activation of oxytocinergic (OTergic) neurons in addition to recruitment of CRH neurons. Interestingly, we found that the hypertensive effect of α-melanocyte stimulating hormone (α-MSH) itself was not blocked by either astressin2B or OVT. These data suggest that while α-MSH-producing neurons are part of a core melanocortin-CRH-oxytocin circuit regulating food intake, and a subpopulation of melanocortin neurons activated by nesfatin-1 do mediate the hypertensive action of the peptide, α-MSH can signal independently from this circuit to increase MAP.


Asunto(s)
Presión Sanguínea/fisiología , Proteínas de Unión al Calcio/fisiología , Proteínas de Unión al ADN/fisiología , Hormonas/fisiología , Hipertensión/fisiopatología , Red Nerviosa/fisiología , Proteínas del Tejido Nervioso/fisiología , Animales , Presión Sanguínea/efectos de los fármacos , Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Hormona Liberadora de Corticotropina/fisiología , Modelos Animales de Enfermedad , Masculino , Melanocortinas/fisiología , Hormonas Estimuladoras de los Melanocitos/farmacología , Nucleobindinas , Oxitocina/fisiología , Fragmentos de Péptidos/farmacología , Péptidos Cíclicos/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos , Receptores de Hormona Liberadora de Corticotropina/fisiología , Receptores de Oxitocina/antagonistas & inhibidores , Receptores de Oxitocina/efectos de los fármacos , Receptores de Oxitocina/fisiología , Vasotocina/farmacología , alfa-MSH/farmacología
10.
J Cell Physiol ; 228(6): 1295-303, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23168683

RESUMEN

Urocortin (Ucn1), a member of corticotrophin-releasing hormone (CRH) family, has been reported to be upregulated in inflammatory diseases and function as an autocrine or paracrine inflammatory mediator. Growing evidence shows that Ucn1 increases the endothelial permeability in inflammatory conditions; however, the detailed mechanisms are not clear. In the present study, we investigated the mechanisms of increased endothelial permeability by Ucn1 in human umbilical vein endothelial cells (HUVECs) exposed to lipopolysaccharide (LPS). Pretreatment of HUVECs with Ucn1 increased the endothelial cell permeability, which was augmented by LPS synergistically. Significant downregulation of VE-cadherin expression was also observed. Moreover, Ucn1 increased phosphorylation of protein kinase D (PKD) and heat shock protein 27 (HSP27) in a time- and CRHR(2) -dependent manner. Inhibition of PKD and HSP27 drastically attenuated Ucn1-induced downregulation of VE-cadherin expression. Further investigations demonstrated that Ucn1 phosphorylated ß-catenin at Ser552 to disrupt the cadherin-catenin complex and hence promote the disassociation of ß-catenin and VE-cadherin. Disassociation of ß-catenin and VE-cadherin resulted in decreased VE-cadherin expression while on the contrary ß-catenin was increased, which may due to the inactivation of GSK-3ß. Increased ß-catenin translocated into the nucleus and subsequently bound to TCF/LEF site, contributing to the elevated expression of vascular endothelial growth factor (VEGF). The above effects of Ucn1 were completely reversed by CRHR(2) receptor blocker, antisauvagine-30. Taken together, our data suggest that Ucn1 increase LPS-induced endothelial permeability by disrupting the VE-cadherin-ß-catenin complex via activation of CRHR(2) and PKD-HSP27 signaling pathway.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Lipopolisacáridos/farmacología , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos , Urocortinas/metabolismo , beta Catenina/metabolismo , Animales , Antígenos CD/genética , Cadherinas/genética , Células Cultivadas , Regulación de la Expresión Génica , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Proteínas de Choque Térmico HSP27/metabolismo , Proteínas de Choque Térmico , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Chaperonas Moleculares , Complejos Multiproteicos , Fragmentos de Péptidos/farmacología , Fosforilación , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Interferencia de ARN , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
Am J Physiol Heart Circ Physiol ; 305(2): H182-91, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23686711

RESUMEN

The presence of urocortins (UCNs) and corticotropin-releasing factor (CRF) receptors has been reported in the hypothalamic arcuate nucleus (ARCN). We have previously reported that UCNs are involved in central cardiovascular regulation. Based on this information, we hypothesized that the ARCN may be one of the sites where UCNs exert their central cardiovascular actions. Experiments were done in artificially ventilated, adult male Wistar rats anesthetized with urethane. Unilateral microinjections (30 nl) of UCN1 (0.12-2 mM) elicited decreases in mean arterial pressure (MAP) and heart rate (HR). Maximum cardiovascular responses were elicited by a 1 mM concentration of UCN1. Microinjections of UCN2 and UCN3 (1 mM each) into the ARCN elicited similar decreases in MAP and HR. UCN1 was used as a prototype for the other experiments described below. HR responses elicited by UCN1 were significantly attenuated by bilateral vagotomy. Prior microinjections of NBI-27914 (CRF-1 receptor antagonist) and astressin (CRF-1 receptor and CRF-2 receptor antagonist) (1 mM each) into the ARCN significantly attenuated the cardiovascular responses elicited by UCN1 microinjections at the same site. Microinjections of UCN1 into the ARCN decreased efferent renal sympathetic nerve activity. It was concluded that microinjections of UCN1, UCN2, and UCN3 into the ARCN elicited decreases in MAP and HR. Decreases in MAP, HR, and renal sympathetic nerve activity elicited by UCN1 microinjections into the ARCN were mediated via CRF receptors. Bradycardic responses to UCN1 were mediated via the activation of vagus nerves, and decreases in MAP may be mediated via decreases in sympathetic nerve activity.


Asunto(s)
Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Sistema Cardiovascular/efectos de los fármacos , Sistema Cardiovascular/inervación , Hemodinámica/efectos de los fármacos , Sistema Nervioso Simpático/efectos de los fármacos , Urocortinas/administración & dosificación , Nervio Vago/efectos de los fármacos , Anestesia General , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Presión Arterial/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Frecuencia Cardíaca/efectos de los fármacos , Antagonistas de Hormonas/administración & dosificación , Riñón/efectos de los fármacos , Riñón/inervación , Masculino , Microinyecciones , Antagonistas de Narcóticos/administración & dosificación , Ratas , Ratas Wistar , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Receptores Ionotrópicos de Glutamato/efectos de los fármacos , Receptores Ionotrópicos de Glutamato/metabolismo , Respiración Artificial , Vagotomía , Nervio Vago/cirugía
12.
Gastroenterology ; 140(5): 1586-96.e6, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21277852

RESUMEN

BACKGROUND & AIMS: Corticotropin-releasing factor receptor-1 (CRF(1)) mediates the stress-induced colonic motor activity. Less is known about the role of CRF(2) in the colonic response to stress. METHODS: We studied colonic contractile activity in rats and CRF(2)-/-, CRF-overexpressing, and wild-type mice using still manometry; we analyzed defecation induced by acute partial-restraint stress (PRS), and/or intraperitoneal injection of CRF ligands. In rats, we monitored activation of the colonic longitudinal muscle myenteric plexus (LMMP) neurons and localization of CRF(1) and CRF(2) using immunohistochemical and immunoblot analyses. We measured phosphorylation of extracellular signal-regulated kinase 1/2 by CRF ligands in primary cultures of LMMP neurons (PC-LMMPn) and cyclic adenosine monophosphate (cAMP) production in human embryonic kidney-293 cells transfected with CRF(1) and/or CRF(2). RESULTS: In rats, a selective agonist of CRF(2) (urocortin 2) reduced CRF-induced defecation (>50%), colonic contractile activity, and Fos expression in the colonic LMMP. A selective antagonist of CRF(2) (astressin(2)-B) increased these responses. Urocortin 2 reduced PRS-induced colonic contractile activity in wild-type and CRF-overexpressing mice, whereas disruption of CRF(2) increased PRS-induced colonic contractile activity and CRF-induced defecation. CRF(2) colocalized with CRF(1) and neuronal nitric oxide synthase in the rat colon, LMMP, and PC-LMMPn. CRF-induced phosphorylation of extracellular signal-regulated kinase in PC-LMMPn; this was inhibited or increased by a selective antagonist of CRF(1) (NBI35965) or astressin(2)-B, respectively. The half maximal effective concentration, EC(50), for the CRF-induced cAMP response was 8.6 nmol/L in human embryonic kidney-293 cells that express only CRF(1); this response was suppressed 10-fold in cells that express CRF(1) and CRF(2). CONCLUSIONS: In colon tissues of rodents, CRF(2) activation inhibits CRF(1) signaling in myenteric neurons and the stress-induced colonic motor responses. Disruption of CRF(2) function impairs colonic coping responses to stress.


Asunto(s)
Colon/fisiopatología , Motilidad Gastrointestinal/fisiología , Plexo Mientérico/metabolismo , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Estrés Fisiológico , Enfermedad Aguda , Animales , Colon/metabolismo , Colon/patología , Modelos Animales de Enfermedad , Femenino , Motilidad Gastrointestinal/efectos de los fármacos , Humanos , Inmunohistoquímica , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos , Plexo Mientérico/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos , Urocortinas/administración & dosificación
13.
Addict Biol ; 17(4): 694-705, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21955024

RESUMEN

The neuropeptide galanin and its three receptor subtypes (GalR1-3) are expressed in the central amygdala (CeA), a brain region involved in stress- and anxiety-related behaviors, as well as alcohol dependence. Galanin also has been suggested to play a role in alcohol intake and alcohol dependence. We examined the effects of galanin in CeA slices from wild-type and knockout (KO) mice deficient of GalR2 and both GalR1 and GalR2 receptors. Galanin had dual effects on gamma-aminobutyric acid (GABA)-ergic transmission, decreasing the amplitudes of pharmacologically isolated GABAergic inhibitory postsynaptic potentials (IPSPs) in over half of CeA neurons but augmenting IPSPs in the others. The increase in IPSP size was absent after superfusion of the GalR3 antagonist SNAP 37889, whereas the IPSP depression was absent in CeA neurons of GalR1 × GalR2 double KO and GalR2 KO mice. Paired-pulse facilitation studies showed weak or infrequent effects of galanin on GABA release. Thus, galanin may act postsynaptically through GalR3 to augment GABAergic transmission in some CeA neurons, whereas GalR2 receptors likely are involved in the depression of IPSPs. Co-superfusion of ethanol, which augments IPSPs presynaptically, together with galanin caused summated effects of ethanol and galanin in those CeA neurons showing galanin-augmented IPSPs, suggesting the two agents act via different mechanisms in this population. However, in neurons showing IPSP-diminishing galanin effects, galanin blunted the ethanol effects, suggesting a preemptive effect of galanin. These findings may increase understanding of the complex cellular mechanisms that underlie the anxiety-related behavioral effects of galanin and ethanol in CeA.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Galanina/farmacología , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos , Animales , Ansiedad/etiología , Depresores del Sistema Nervioso Central/farmacología , Interacciones Farmacológicas , Etanol/farmacología , Potenciales Evocados/efectos de los fármacos , Neuronas GABAérgicas/efectos de los fármacos , Indoles/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Estrés Psicológico/etiología , Ácido gamma-Aminobutírico/efectos de los fármacos
14.
Gen Comp Endocrinol ; 170(3): 613-21, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21130092

RESUMEN

The present study, for the first time in ectothermic vertebrates, reports the immunoregulatory role of urotensins I and II (UI and UII). Urotensins decreased the phagocytosis and nitrite production by splenic phagocytes. On superoxide production, UI had stimulatory while UII showed inhibitory effect. UI exerted its effect on phagocytes through corticotrophin-releasing factor (CRF) receptor as its non-specific antagonist astressin completely blocked the effect of UI on phagocytosis, nitrite release and superoxide production. Among the antagonists for specific CRF receptor 1 and 2, only CRF receptor 1 antagonist NBI 27914 abolished the effect of urotensin I. On the other hand, UII mediated its effect through urotensin receptor (UT receptor) since its antagonist urantide antagonized the effect of UII on phagocytosis, superoxide and nitrite release. These findings provide the direct evidence on physiological role of UI and UII through CRF receptor 1 and UT receptor, respectively in control of fish immune responses.


Asunto(s)
Factores Inmunológicos/fisiología , Perciformes/fisiología , Urotensinas/fisiología , Animales , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos , Receptores de Hormona Liberadora de Corticotropina/fisiología , Receptores Acoplados a Proteínas G/fisiología
15.
Neuropeptides ; 88: 102162, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34062382

RESUMEN

Corticotropin releasing factor (CRF) is a neuropeptide widely distributed in the brain as a hormonal modulator and neurotransmitter. The best known behavioral function of CRF is activation of stress and anxiety via the hypothalamus and limbic structures but the role of CRF in the cortex is still poorly understood. Our previous studies have shown anxiolytic-like effects of high doses of CRF injected into the Fr2 frontal cortex and involvement of CRF1 receptors (R) in that effect. These results seemed to be controversial as most other studies suggested anxiogenic and not anxiolytic effects of CRF1R stimulation. Since stress is associated with adrenergic system, in the present study, we focused on participation of alpha1 and alpha2 or beta adrenergic receptors in the anxiolytic-like effect of CRF. Moreover, we verified whether these effects of CRF in the Fr2 were really connected with CRF1R. Male Wistar rats were bilaterally microinjected with CRF in a dose of 0.2 µg/1 µl/site or with the specific agonist of CRF1R, stressin 1 (0.2-0.0125 µg/1 µl/site) into the Fr2 area. The elevated plus maze (EPM) test was performed 30 min later to assess the anxiolysis. An involvement of noradrenergic receptors in the CRF induced anxiolytic-like effect in the Fr2 was studied by pretreatment with the alpha1 antagonist prazosin, alpha2 agonist clonidine, alpha2 antagonist RS 79948 or beta antagonist propranolol, 20-30 min before CRF. The influence on anxiety was assessed in the EPM test. The results show that anxiolytic behavior after CRF microinjection into the Fr2 area seems to be mainly connected with the CRF1R activation because a similar effect was observed after stressin 1 administration and it was blocked by CRF1R antagonist. The results observed after administration of noradrenergic ligands indicated that anxiolytic effects of CRF in the Fr2 engaged the alpha1 and alpha2 adrenergic receptors but not beta receptors.


Asunto(s)
Ansiolíticos/farmacología , Ansiedad/tratamiento farmacológico , Conducta Animal/efectos de los fármacos , Hormona Liberadora de Corticotropina/farmacología , Lóbulo Frontal/efectos de los fármacos , Animales , Hormona Liberadora de Corticotropina/administración & dosificación , Isoquinolinas/farmacología , Naftiridinas/farmacología , Ratas , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos
16.
Neuropeptides ; 88: 102147, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33932861

RESUMEN

Corticotropin-releasing factor (CRF) and the urocortins (Ucn1, Ucn2 and Ucn3) are structurally related neuropeptides which act via two distinct CRF receptors, CRF1 and CRF2, with putatively antagonistic effects in the brain. CRF and Ucn1 activate both CRF1 and CRF2, while Ucn2 and Ucn3 activate selectively CRF2. The aim of the present study was to investigate the effects of CRF, Ucn1, Ucn2 and Ucn3 on the hippocampal acetylcholine release through which they may modulate cognitive functions, including attention, learning and memory. In this purpose male Wistar rats were used, their hippocampus was isolated, dissected, incubated, superfused and stimulated electrically. The hippocampal slices were first pretreated with selective CRF1 antagonist antalarmin or selective CRF2 antagonist astressin2B, and then treated with non-selective CRF1 agonists, CRF or Ucn1, and selective CRF2 agonists, Ucn2 or Ucn3. The hippocampal acetylcholine release was increased significantly by CRF and Ucn1 and decreased significantly by Ucn2 and Ucn3. The increasing effect of CRF and Ucn1 was reduced significantly by antalarmin, but not astressin2B. In contrast, the decreasing effect of Ucn2 and Ucn3 was reversed significantly by the selective CRF2, but not the selective CRF1 antagonist. Our results demonstrate that CRF and Ucn1 stimulate the hippocampal acetylcholine release through CRF1, whereas Ucn2 and Ucn3 inhibit the hippocampal acetylcholine release through CRF2. Therefore, the present study suggests the existence of two apparently opposing CRF systems in the hippocampus, through which CRF and the urocortins might modulate cholinergic activity and thereby cognitive functions.


Asunto(s)
Acetilcolina/metabolismo , Hormona Liberadora de Corticotropina/farmacología , Hipocampo/efectos de los fármacos , Urocortinas/farmacología , Animales , Hormona Liberadora de Corticotropina/metabolismo , Hipocampo/metabolismo , Fragmentos de Péptidos/metabolismo , Ratas Wistar , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Urocortinas/metabolismo
17.
Neuropharmacology ; 200: 108819, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34610289

RESUMEN

The basolateral amygdala (BLA) is a critical brain region for cocaine-memory reconsolidation. Corticotropin-releasing factor receptor type 1 (CRFR1) is densely expressed in the BLA, and CRFR1 stimulation can activate intra-cellular signaling cascades that mediate memory reconsolidation. Hence, we tested the hypothesis that BLA CRFR1 stimulation is necessary and sufficient for cocaine-memory reconsolidation. Using an instrumental model of drug relapse, male and female Sprague-Dawley rats received cocaine self-administration training in a distinct environmental context over 10 days followed by extinction training in a different context over 7 days. Next, rats were re-exposed to the cocaine-paired context for 15 min to initiate cocaine-memory retrieval and destabilization. Immediately or 6 h after this session, the rats received bilateral vehicle, antalarmin (CRFR1 antagonist; 500 ng/hemisphere), or corticotropin-releasing factor (CRF; 0.2, 30 or 500 ng/hemisphere) infusions into the BLA. Resulting changes in drug context-induced cocaine seeking (index of context-cocaine memory strength) were assessed three days later. Female rats self-administered more cocaine infusions and exhibited more extinction responding than males. Intra-BLA antalarmin treatment immediately after memory retrieval (i.e., when cocaine memories were labile), but not 6 h later (i.e., after memory reconsolidation), attenuated drug context-induced cocaine seeking at test independent of sex, relative to vehicle. Conversely, intra-BLA CRF treatment increased this behavior selectively in females, in a U-shaped dose-dependent fashion. In control experiments, a high (behaviorally ineffective) dose of CRF treatment did not reduce BLA CRFR1 cell-surface expression in females. Thus, BLA CRFR1 signaling is necessary and sufficient, in a sex-dependent manner, for regulating cocaine-memory strength.


Asunto(s)
Complejo Nuclear Basolateral/efectos de los fármacos , Trastornos Relacionados con Cocaína/patología , Cocaína/farmacología , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Memoria/efectos de los fármacos , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos , Animales , Hormona Liberadora de Corticotropina/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Pirimidinas/farmacología , Pirroles/farmacología , Ratas , Ratas Sprague-Dawley
18.
Behav Brain Res ; 399: 113015, 2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33212086

RESUMEN

Post-traumatic stress disorder (PTSD) is a psychological disorder affecting many around the world. Growing evidence suggests that orexin-A is involved in the pathophysiology of depression and panic anxiety disorder. However, the role of orexin-A in PTSD remains unclear. Therefore, pharmacological manipulation of orexin-A can be a potential approach for the treatment of PTSD. Male Wistar rats were subjected to stress re-stress (SRS) by restraining them for 2 h followed by foot shock (FS) and halothane exposure on day-2 (D-2). Then the rats were weekly exposed to FS as re-stress cue . Suvorexant, an orexin antagonist (10, 20 and 30 mg/kg p.o.) and paroxetine (10 mg/kg p.o.) were administered from D-8 to D-32. Plasma and cerebrospinal fluid (CSF) were collected for corticosterone and orexin-A measurement. The analysis of serotonin and corticotropin-releasing factor receptor-1 (CRF-R1) were performed in the amygdalar tissue. SRS-induced PTSD-like symptoms like fear response, anxiety-like behaviour and hypocorticosteronism were attenuated by suvorexant and paroxetine. Interestingly, SRS exposed rats showed activation of orexin-A and serotonergic systems, which were also attenuated by suvorexant. Additionally, suvorexant ameliorated the extrahypothalamic induced upregulation of CRH-R1 in SRS-exposed rats. Therefore, orexin-A may be considered as a neurochemical-marker for PTSD and suvorexant alleviated PTSD-like symptoms through modulating orexinergic, serotonergic and neuroendocrine systems.


Asunto(s)
Amígdala del Cerebelo , Azepinas/farmacología , Corticosterona , Antagonistas de los Receptores de Orexina/farmacología , Orexinas , Receptores de Hormona Liberadora de Corticotropina , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Serotonina , Trastornos por Estrés Postraumático/tratamiento farmacológico , Triazoles/farmacología , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Azepinas/administración & dosificación , Corticosterona/sangre , Corticosterona/líquido cefalorraquídeo , Modelos Animales de Enfermedad , Masculino , Antagonistas de los Receptores de Orexina/administración & dosificación , Orexinas/sangre , Orexinas/líquido cefalorraquídeo , Orexinas/efectos de los fármacos , Paroxetina/farmacología , Ratas , Ratas Wistar , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Serotonina/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Trastornos por Estrés Postraumático/etiología , Triazoles/administración & dosificación
19.
Mol Pharmacol ; 77(5): 864-73, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20159948

RESUMEN

The role of stress in drug addiction is well established. The negative affective states of withdrawal most probably involve recruitment of brain stress neurocircuitry [e.g., induction of hypothalamo-pituitary-adrenocortical (HPA) axis, noradrenergic activity, and corticotropin-releasing factor (CRF) activity]. The present study investigated t$he role of CRF receptor-1 subtype (CRF1R) on the response of brain stress system to morphine withdrawal. The effects of naloxone-precipitated morphine withdrawal on noradrenaline (NA) turnover in the paraventricular nucleus (PVN), HPA axis activity, signs of withdrawal, and c-Fos expression were measured in rats pretreated with vehicle, CP-154526 [N-butyl-N-ethyl-2,5-dimethyl-7-(2,4,6-trimethylphenyl)pyrrolo[3,2-e]pyrimidin-4-amine], or antalarmin (selective CRF1R antagonists). Tyrosine hydroxylase-positive neurons expressing CRF1R were seen at the level of the nucleus tractus solitarius-A(2) cell group in both control and morphine-withdrawn rats. CP-154526 and antalarmin attenuated the increases in body weight loss and irritability that were seen during naloxone-induced morphine withdrawal. Pretreatment with CRF1R antagonists resulted in no significant modification of the increased NA turnover at PVN, plasma corticosterone levels, or c-Fos expression that was seen during naloxone-induced morphine withdrawal. However, blockade of CRF1R significantly reduced morphine withdrawal-induced increases in plasma adrenocorticotropin levels. These results suggest that the CRF1R subtype may be involved in the behavioral and somatic signs and in adrenocorticotropin release (partially) during morphine withdrawal. However, CRF1R activation may not contribute to the functional interaction between NA and CRF systems in mediating morphine withdrawal-activation of brain stress neurocircuitry.


Asunto(s)
Encéfalo/fisiopatología , Morfina/farmacología , Receptores de Hormona Liberadora de Corticotropina/fisiología , Síndrome de Abstinencia a Sustancias/fisiopatología , Hormona Adrenocorticotrópica/sangre , Animales , Encéfalo/efectos de los fármacos , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/fisiopatología , Corticosterona/sangre , Radioisótopos de Yodo , Masculino , Metoxihidroxifenilglicol/metabolismo , Morfina/sangre , Norepinefrina/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Radioinmunoensayo , Ratas , Ratas Sprague-Dawley , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos , Estrés Psicológico/fisiopatología , Trastornos Relacionados con Sustancias/fisiopatología , Tirosina 3-Monooxigenasa/metabolismo
20.
J Neurochem ; 115(3): 795-803, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20807310

RESUMEN

The roles of two subtypes of corticotrophin-releasing factor (CRF) receptor in corticostriatal synaptic plasticity under cocaine withdrawal condition were examined in this study. Neither the resting membrane potential and input resistance of striatal neurons nor the long-term potentiation (LTP) of corticostriatal slices were affected by cocaine withdrawal. CRF dose-dependently enhanced in vitro corticostriatal LTP in rats from both cocaine-withdrawal and saline-control groups. Yet, the enhancement of corticostriatal LTP by CRF (20, 40, 80 nM) was significantly greater in the cocaine-withdrawal group than in the control group. CRF(1)-selective antagonist (NBI 27914, 100 nM) attenuated the CRF-induced enhancement of corticostriatal LTP in both groups, whereas the CRF(2)-selective antagonist (astression2B, 100 nM) attenuated the enhanced corticostriatal LTP only in the cocaine-withdrawal group. Importantly, urocortin2 (a CRF(2)-selective agonist, 40 nM) selectively increased corticostriatal LTP in the cocaine-withdrawal group, but not in the saline controls. The urocortin2-induced enhancement of LTP was totally blocked by astression2B (100 nM). These results suggest that the CRF system modulate neuroadaptive changes in the corticostriatal circuit during cocaine withdrawal, and the CRF(2) in this area mediate an important mechanism that contributes to the relapse of cocaine addiction.


Asunto(s)
Corteza Cerebral/fisiología , Cocaína/efectos adversos , Cuerpo Estriado/fisiología , Potenciación a Largo Plazo/fisiología , Receptores de Hormona Liberadora de Corticotropina/fisiología , Síndrome de Abstinencia a Sustancias/fisiopatología , Animales , Hormona Liberadora de Corticotropina/metabolismo , Electrofisiología , Masculino , Técnicas de Placa-Clamp , Ratas , Ratas Wistar , Receptores de Hormona Liberadora de Corticotropina/efectos de los fármacos , Síndrome de Abstinencia a Sustancias/psicología , Sinapsis/fisiología , Urocortinas/farmacología , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA