Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 11(1): 10962, 2021 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-34040115

RESUMEN

Neuropeptides have been reported to regulate progenitor proliferation and neurogenesis in the central nervous system. However, these studies have typically been conducted using pharmacological agents in ex vivo preparations, and in vivo evidence for their developmental function is generally lacking. Recent scRNA-Seq studies have identified multiple neuropeptides and their receptors as being selectively expressed in neurogenic progenitors of the embryonic mouse and human retina. This includes Sstr2, whose ligand somatostatin is transiently expressed by immature retinal ganglion cells. By analyzing retinal explants treated with selective ligands that target these receptors, we found that Sstr2-dependent somatostatin signaling induces a modest, dose-dependent inhibition of photoreceptor generation, while correspondingly increasing the relative fraction of primary progenitor cells. These effects were confirmed by scRNA-Seq analysis of retinal explants but abolished in Sstr2-deficient retinas. Although no changes in the relative fraction of primary progenitors or photoreceptor precursors were observed in Sstr2-deficient retinas in vivo, scRNA-Seq analysis demonstrated accelerated differentiation of neurogenic progenitors. We conclude that, while Sstr2 signaling may act to negatively regulate retinal neurogenesis in combination with other retinal ganglion cell-derived secreted factors such as Shh, it is dispensable for normal retinal development.


Asunto(s)
Proteínas del Ojo/fisiología , Neurogénesis/fisiología , Neuropéptidos/fisiología , Receptores de Somatostatina/fisiología , Retina/citología , Animales , Relación Dosis-Respuesta a Droga , Proteínas del Ojo/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Edad Gestacional , Humanos , Ligandos , Ratones , Ratones Noqueados , Neuropéptidos/agonistas , Neuropéptidos/antagonistas & inhibidores , Neuropéptidos/farmacología , Fenotipo , Células Fotorreceptoras de Vertebrados/efectos de los fármacos , Células Fotorreceptoras de Vertebrados/metabolismo , Receptores de Somatostatina/deficiencia , Receptores de Somatostatina/efectos de los fármacos , Retina/embriología , Transducción de Señal/fisiología , Análisis de la Célula Individual
2.
Int J Neuropsychopharmacol ; 23(1): 53-65, 2020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-31563948

RESUMEN

BACKGROUND: Evidence from anatomical, pharmacological, and genetic studies supports a role for the neuropeptide melanin concentrating hormone system in modulating emotional and cognitive functions. Genome-wide association studies revealed a potential association between the melanin concentrating hormone receptor (MCHR1) gene locus and schizophrenia, and the largest genome-wide association study conducted to date shows a credible genome-wide association. METHODS: We analyzed MCHR1 and pro-melanin concentrating hormone RNA-Seq expression in the prefrontal cortex in schizophrenia patients and healthy controls. Disruptions in the melanin concentrating hormone system were modeled in the mouse brain by germline deletion of MCHR1 and by conditional ablation of melanin concentrating hormone expressing neurons using a Cre-inducible diphtheria toxin system. RESULTS: MCHR1 expression is decreased in the prefrontal cortex of schizophrenia samples (false discovery rate (FDR) P < .05, CommonMind and PsychEncode combined datasets, n = 901) while pro-melanin concentrating hormone is below the detection threshold. MCHR1 expression decreased with aging (P = 6.6E-57) in human dorsolateral prefrontal cortex. The deletion of MCHR1 was found to lead to behavioral abnormalities mimicking schizophrenia-like phenotypes: hyperactivity, increased stereotypic and repetitive behavior, social impairment, impaired sensorimotor gating, and disrupted cognitive functions. Conditional ablation of pro-melanin concentrating hormone neurons increased repetitive behavior and produced a deficit in sensorimotor gating. CONCLUSIONS: Our study indicates that early disruption of the melanin concentrating hormone system interferes with neurodevelopmental processes, which may contribute to the pathogenesis of schizophrenia. Further neurobiological research on the developmental timing and circuits that are affected by melanin concentrating hormone may lead to a therapeutic target for early prevention of schizophrenia.


Asunto(s)
Hormonas Hipotalámicas/metabolismo , Melaninas/metabolismo , Trastornos de la Memoria/fisiopatología , Hormonas Hipofisarias/metabolismo , Corteza Prefrontal/metabolismo , Receptores de Somatostatina/deficiencia , Receptores de Somatostatina/metabolismo , Esquizofrenia/metabolismo , Esquizofrenia/fisiopatología , Filtrado Sensorial/fisiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Conducta Animal/fisiología , Niño , Preescolar , Modelos Animales de Enfermedad , Femenino , Feto , Humanos , Lactante , Masculino , Trastornos de la Memoria/etiología , Ratones , Ratones Noqueados , Persona de Mediana Edad , Esquizofrenia/complicaciones , Adulto Joven
3.
Artículo en Inglés | MEDLINE | ID: mdl-29409919

RESUMEN

This study investigated whether sst2 gene deletion interacts with age and chronic stress exposure to produce exacerbated emotional and cognitive ageing. Middle-aged (10-12 month) sst2 knockout (sst2KO) and wild-type (WT) mice underwent an unpredictable chronic mild stress (UCMS) procedure for 6 weeks or no stress for control groups. This was followed by a battery of tests to assess emotional and cognitive functions and neuroendocrine status (CORT level). A re-evaluation was performed 6 months later (i.e. with 18-month-old mice). UCMS reproduced neuroendocrine and behavioral features of stress-related disorders such as elevated circulating CORT levels, physical deteriorations, increased anxiety- and depressive-like behaviors and working memory impairments. sst2KO mice displayed behavioral alterations which were similar to stressed WT and exhibited exacerbated changes following UCMS exposure. The evaluations performed in the older mice showed significant long-term effects of UCMS exposure. Old sst2KO mice previously exposed to UCMS exhibited spatial learning and memory accuracy impairments and high levels of anxiety-like behaviors which drastically added to the effects of normal ageing. Spatial abilities and emotionality scores (mean z-scores) measured both at the UCMS outcome and 6 months later were correlated with the initially measured CORT levels in middle-age. The present findings indicate that the deletion of the sst2 receptor gene produces chronic hypercorticosteronemia and exacerbates sensitivity to stressors which over time, have consequences on ageing brain function processes.


Asunto(s)
Envejecimiento/metabolismo , Envejecimiento/psicología , Cognición/fisiología , Emociones/fisiología , Receptores de Somatostatina/deficiencia , Estrés Psicológico/metabolismo , Animales , Ansiedad/metabolismo , Enfermedad Crónica , Disfunción Cognitiva/metabolismo , Corticosterona/sangre , Depresión/metabolismo , Modelos Animales de Enfermedad , Eliminación de Gen , Trastornos de la Memoria/metabolismo , Memoria a Corto Plazo/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Somatostatina/genética
4.
Behav Brain Res ; 316: 271-278, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27633558

RESUMEN

In order to decipher the functional involvement of melanin-concentrating hormone 1 (MCH1) receptors in the control of feeding and foraging behaviors, mice with constitutive deletion of MCH1 receptors MCH1R -/- or knockout (KO) were studied and compared to age-matched littermate control mice (MCH1R +/+ or wildtype (WT)). Several challenges to food-motivated behaviors of food-restricted WT and KO mice were implemented. There were no differences between genotypes in the acquisition of a nose-poke response that produced food or in a discrimination between a response that produced food and one that did not. There were also no genotype differences in the rate of extinction of a food-motivated response. However, during the first day of extinction, foraging behaviors were increased significantly more in KO than in WT mice. Likewise, when the response requirement to obtain food was progressively increased, KO mice made significantly more food-directed responses than WT mice. Although adulteration of food with quinine did not suppress food-directed behavior in either genotype when the mice were food-restricted, manipulation of the degree of food-deprivation resulted in suppression of behavior of WT mice without suppressing the behavior of KO mice. Although response-produced foot shock suppressed food-maintained responding of both WT and KO mice, equipotent levels of shock (based upon psychophysical thresholds) suppressed behavior of WT mice without suppressing behavior of the KO mice. Finally, under a Vogel conflict procedure, KO mice had significantly higher levels of both punished and non-punished food maintained responding. Thus, the data from challenges with both appetitive and noxious stimulus challenges support the conclusion that mice with constitutive deletion of MCH1Rs have increased food seeking motivation that is coincident with their higher metabolism. The data also highlight important differences in the biological impact of MCH1 receptor KO and MCH1 receptor antagonism.


Asunto(s)
Ingestión de Alimentos/genética , Conducta Alimentaria/fisiología , Receptores de Somatostatina/deficiencia , Refuerzo en Psicología , Animales , Animales Recién Nacidos , Biofisica , Condicionamiento Operante/fisiología , Estimulación Eléctrica , Femenino , Alimentos , Privación de Alimentos , Masculino , Ratones , Ratones Transgénicos , Quinina/administración & dosificación , Receptores de Somatostatina/genética , Saciedad/fisiología
5.
Br J Pharmacol ; 173(18): 2739-51, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27400775

RESUMEN

BACKGROUND AND PURPOSE: Melanin-concentrating hormone (MCH) is an orexigen, and while rodents express one MCH receptor (MCH1 receptor), humans, non-human primates and dogs express two MCH receptors (MCH1 and MCH2 ). MCH1 receptor antagonists have been developed for the treatment of obesity and lower body weight in rodents. However, the mechanisms for the body weight loss and whether MCH1 receptor antagonism can lower body weight in species expressing both MCH receptors are not fully understood. EXPERIMENTAL APPROACH: A novel recently identified potent MCH1 receptor antagonist, AZD1979, was studied in wild type and Mchr1 knockout (KO) mice and by using pair-feeding and indirect calorimetry in diet-induced obese (DIO) mice. The effect of AZD1979 on body weight was also studied in beagle dogs. KEY RESULTS: AZD1979 bound to MCH1 receptors in the CNS and dose-dependently reduced body weight in DIO mice leading to improved homeostasis model assessment-index of insulin sensitivity. AZD1979 did not affect food intake or body weight in Mchr1 KO mice demonstrating specificity for the MCH1 receptor mechanism. In DIO mice, initial AZD1979-mediated body weight loss was driven by decreased food intake, but an additional component of preserved energy expenditure was apparent in pair-feeding and indirect calorimetry studies. AZD1979 also dose-dependently reduced body weight in dogs. CONCLUSION AND IMPLICATIONS: AZD1979 is a novel potent MCH1 receptor antagonist that affects both food intake and energy expenditure. That AZD1979 also lowers body weight in a species expressing both MCH receptors holds promise for the use of MCH1 receptor antagonists for the treatment of human obesity.


Asunto(s)
Azetidinas/farmacología , Peso Corporal/efectos de los fármacos , Homeostasis/efectos de los fármacos , Oxadiazoles/farmacología , Receptores de Somatostatina/antagonistas & inhibidores , Animales , Azetidinas/administración & dosificación , Azetidinas/química , Perros , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estructura Molecular , Oxadiazoles/administración & dosificación , Oxadiazoles/química , Receptores de Somatostatina/deficiencia , Relación Estructura-Actividad
6.
J Med Chem ; 59(3): 1116-39, 2016 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-26736071

RESUMEN

Aiming to discover melanin-concentrating hormone receptor 1 (MCHR1) antagonists with improved safety profiles, we hypothesized that the aliphatic amine employed in most antagonists reported to date could be removed if the bicyclic motif of the compound scaffold interacted with Asp123 and/or Tyr272 of MCHR1. We excluded aliphatic amines from our compound designs, with a cutoff value of pK(a) < 8, and explored aliphatic amine-free MCHR1 antagonists in a CNS-oriented chemical space limited by four descriptors (TPSA, ClogP, MW, and HBD count). Screening of novel bicyclic motifs with high intrinsic binding affinity for MCHR1 identified the imidazo[1,2-a]pyridine ring (represented in compounds 6a and 6b), and subsequent cyclization of the central aliphatic amide linkage led to the discovery of a potent, orally bioavailable MCHR1 antagonist 4-[(4-chlorobenzyl)oxy]-1-(2-cyclopropyl-3-methylimidazo[1,2-a]pyridin-6-yl)pyridin-2(1H)-one 10a. It exhibited low potential for hERG inhibition and phospholipidosis induction as well as sufficient brain concentration to exert antiobesity effects in diet-induced obese rats.


Asunto(s)
Fármacos Antiobesidad/farmacología , Imidazoles/química , Imidazoles/farmacología , Obesidad/tratamiento farmacológico , Piridonas/química , Piridonas/farmacología , Receptores de Somatostatina/antagonistas & inhibidores , Animales , Fármacos Antiobesidad/síntesis química , Fármacos Antiobesidad/química , Células CHO , Cricetulus , Relación Dosis-Respuesta a Droga , Humanos , Imidazoles/síntesis química , Masculino , Estructura Molecular , Piridonas/síntesis química , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley , Receptores de Somatostatina/deficiencia , Relación Estructura-Actividad
7.
Physiol Behav ; 152(Pt B): 402-7, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26048303

RESUMEN

Exposure to environmental cues associated with food can evoke eating behavior in the absence of hunger. This capacity for reward cues to promote feeding behaviors under sated conditions can be examined in the laboratory using cue-potentiated feeding (CPF). The orexigenic neuropeptide Melanin Concentrating Hormone (MCH) is expressed throughout brain circuitry critical for CPF. We examined whether deletion of the MCH receptor, MCH-1R, would in KO mice disrupt overeating in the presence of a Pavlovian CS+ associated with sucrose delivery. While both wild-type controls and KO mice showed comparable food magazine approach responses during the CPF test, MCH-1R deletion significantly impaired the ability of the CS+ to evoke overeating of sucrose under satiety. Through the use of a refined analysis of meal intake, it was revealed that this disruption to overeating behavior in KO mice reflected a reduction in the capacity for the CS+ to initiate and maintain bursts of licking behavior. These findings suggest that overeating during CPF requires intact MCH-1R signaling and may be due to an influence of the CS+ on the palatability of food and on regulatory mechanisms of peripheral control. Thus, disruptions to MCH-1R signaling may be a useful pharmacological tool to inhibit this form of overeating behavior.


Asunto(s)
Señales (Psicología) , Conducta Alimentaria/fisiología , Alimentos , Hiperfagia/fisiopatología , Receptores de Somatostatina/deficiencia , Animales , Condicionamiento Clásico/fisiología , Sacarosa en la Dieta , Conducta Alimentaria/psicología , Hiperfagia/psicología , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/fisiología , Boca/fisiología , Receptores de Somatostatina/genética , Saciedad/fisiología , Eliminación de Secuencia
9.
Gastroenterology ; 148(7): 1452-65, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25683115

RESUMEN

BACKGROUND & AIMS: The KRAS gene is mutated in most pancreatic ductal adenocarcinomas (PDAC). Expression of this KRAS oncoprotein in mice is sufficient to initiate carcinogenesis but not progression to cancer. Activation of phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) is required for KRAS for induction and maintenance of PDAC in mice. The somatostatin receptor subtype 2 (sst2) inhibits PI3K, but sst2 expression is lost during the development of human PDAC. We investigated the effects of sst2 loss during KRAS-induced PDAC development in mice. METHODS: We analyzed tumor growth in mice that expressed the oncogenic form of KRAS (KRAS(G12D)) in pancreatic precursor cells, as well as sst2+/- and sst2-/-, and in crossed KRAS(G12D);sst2+/- and KRAS(G12D);sst2-/- mice. Pancreatic tissues and acini were collected and assessed by histologic, immunoblot, immunohistochemical, and reverse-transcription polymerase chain reaction analyses. We also compared protein levels in paraffin-embedded PDAC samples from patients vs heathy pancreatic tissues from individuals without pancreatic cancer. RESULTS: In sst2+/- mice, PI3K was activated and signaled via AKT (PKB; protein kinase B); when these mice were crossed with KRAS(G12D) mice, premalignant lesions, tumors, and lymph node metastases developed more rapidly than in KRAS(G12D) mice. In crossed KRAS(G12D);sst2+/- mice, activation of PI3K signaling via AKT resulted in activation of nuclear factor-κB (NF-κB), which increased KRAS activity and its downstream pathways, promoting initiation and progression of neoplastic lesions. We found this activation loop to be mediated by PI3K-induced production of the chemokine CXCL16. Administration of a CXCL16-neutralizing antibody to KRAS(G12D) mice reduced activation of PI3K signaling to AKT and NF-κB, blocking carcinogenesis. Levels of CXCL16 and its receptor CXCR6 were significantly higher in PDAC tissues and surrounding acini than in healthy pancreatic tissues from mice or human beings. In addition, expression of sst2 was progressively lost, involving increased PI3K activity, in mouse lesions that expressed KRAS(G12D) and progressed to PDAC. CONCLUSIONS: Based on analyses of mice, loss of sst2 from pancreatic tissues activates PI3K signaling via AKT, leading to activation of NF-κB, amplification of oncogenic KRAS signaling, increased expression of CXCL16, and pancreatic tumor formation. CXCL16 might be a therapeutic target for PDAC.


Asunto(s)
Carcinoma Ductal Pancreático/enzimología , Proliferación Celular , Quimiocina CXCL6/metabolismo , Mutación , Neoplasias Pancreáticas/enzimología , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Receptores de Somatostatina/deficiencia , Transducción de Señal , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/secundario , Estudios de Casos y Controles , Línea Celular Tumoral , Quimiocina CXCL16 , Quimiocinas CXC/metabolismo , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Humanos , Metástasis Linfática , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Fenotipo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Depuradores/metabolismo , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Factores de Tiempo , Transfección , Carga Tumoral , Regulación hacia Arriba
10.
PLoS One ; 9(11): e112109, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25427253

RESUMEN

We have generated a novel monoclonal antibody targeting human FGFR1c (R1c mAb) that caused profound body weight and body fat loss in diet-induced obese mice due to decreased food intake (with energy expenditure unaltered), in turn improving glucose control. R1c mAb also caused weight loss in leptin-deficient ob/ob mice, leptin receptor-mutant db/db mice, and in mice lacking either the melanocortin 4 receptor or the melanin-concentrating hormone receptor 1. In addition, R1c mAb did not change hypothalamic mRNA expression levels of Agrp, Cart, Pomc, Npy, Crh, Mch, or Orexin, suggesting that R1c mAb could cause food intake inhibition and body weight loss via other mechanisms in the brain. Interestingly, peripherally administered R1c mAb accumulated in the median eminence, adjacent arcuate nucleus and in the circumventricular organs where it activated the early response gene c-Fos. As a plausible mechanism and coinciding with the initiation of food intake suppression, R1c mAb induced hypothalamic expression levels of the cytokines Monocyte chemoattractant protein 1 and 3 and ERK1/2 and p70 S6 kinase 1 activation.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Órganos Circunventriculares/efectos de los fármacos , Intolerancia a la Glucosa/tratamiento farmacológico , Hipotálamo/efectos de los fármacos , Obesidad/tratamiento farmacológico , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Núcleo Arqueado del Hipotálamo/fisiopatología , Quimiocina CCL2/agonistas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocina CCL7/agonistas , Quimiocina CCL7/genética , Quimiocina CCL7/metabolismo , Órganos Circunventriculares/metabolismo , Órganos Circunventriculares/fisiopatología , Ingestión de Alimentos/efectos de los fármacos , Metabolismo Energético , Femenino , Regulación de la Expresión Génica , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/fisiopatología , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiopatología , Leptina/deficiencia , Leptina/genética , Ratones , Ratones Noqueados , Ratones Obesos , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Obesidad/genética , Obesidad/metabolismo , Obesidad/fisiopatología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor de Melanocortina Tipo 4/deficiencia , Receptor de Melanocortina Tipo 4/genética , Receptores de Somatostatina/deficiencia , Receptores de Somatostatina/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Factor de Respuesta Sérica/agonistas , Factor de Respuesta Sérica/genética , Factor de Respuesta Sérica/metabolismo , Transducción de Señal
11.
PLoS One ; 9(9): e108146, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25268135

RESUMEN

Hair cells and spiral ganglion neurons of the mammalian auditory system do not regenerate, and their loss leads to irreversible hearing loss. Aminoglycosides induce auditory hair cell death in vitro, and evidence suggests that phosphatidylinositol-3-kinase/Akt signaling opposes gentamicin toxicity via its downstream target, the protein kinase Akt. We previously demonstrated that somatostatin-a peptide with hormone/neurotransmitter properties-can protect hair cells from gentamicin-induced hair cell death in vitro, and that somatostatin receptors are expressed in the mammalian inner ear. However, it remains unknown how this protective effect is mediated. In the present study, we show a highly significant protective effect of octreotide (a drug that mimics and is more potent than somatostatin) on gentamicin-induced hair cell death, and increased Akt phosphorylation in octreotide-treated organ of Corti explants in vitro. Moreover, we demonstrate that somatostatin receptor-1 knockout mice overexpress somatostatin receptor-2 in the organ of Corti, and are less susceptible to gentamicin-induced hair cell loss than wild-type or somatostatin-1/somatostatin-2 double-knockout mice. Finally, we show that octreotide affects auditory hair cells, enhances spiral ganglion neurite number, and decreases spiral ganglion neurite length.


Asunto(s)
Células Ciliadas Auditivas/metabolismo , Pérdida Auditiva/genética , Receptores de Somatostatina/genética , Ganglio Espiral de la Cóclea/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Regulación de la Expresión Génica , Gentamicinas , Células Ciliadas Auditivas/efectos de los fármacos , Células Ciliadas Auditivas/patología , Pérdida Auditiva/inducido químicamente , Pérdida Auditiva/fisiopatología , Pérdida Auditiva/prevención & control , Ratones , Ratones Noqueados , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Neuritas/ultraestructura , Octreótido/farmacología , Técnicas de Cultivo de Órganos , Fosforilación/efectos de los fármacos , Sustancias Protectoras/farmacología , Proteínas Proto-Oncogénicas c-akt/agonistas , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Somatostatina/deficiencia , Transducción de Señal , Ganglio Espiral de la Cóclea/efectos de los fármacos , Ganglio Espiral de la Cóclea/ultraestructura
12.
Cell Tissue Res ; 358(3): 717-27, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25149275

RESUMEN

Somatostatin (SST) is a peptide hormone that exerts inhibitory effects mediated through binding to specific cell surface G protein-coupled receptors, of which five distinct subtypes (SSTR1-SSTR5) have been characterized. Our study performed on mouse cochlear hair cells shows the expression and localization of the three receptors (SSTR3-SSTR5) in wild-type (WT), single-knockout (SSTR1 KO) and double-knockout SSTR1/SSTR2 (DKO) mice. Similar SSTRs expression were observed in the inner hair cells (IHC), outer hair cells (OHC) and supporting cells of cultivated P7 mouse organ of Corti (OC) explants as well as in cultivated cochlear neuroepithelial supporting cells (NEsc). We found differences in the expression of SSTR3-5 in WT, SSTR1 KO and DKO mouse cochlea, which might be explained as a compensatory effect in the cochlea after the loss of SSTR1 and/or SSTR2.


Asunto(s)
Cóclea/metabolismo , Receptores de Somatostatina/deficiencia , Receptores de Somatostatina/metabolismo , Envejecimiento/metabolismo , Animales , Cóclea/embriología , Embrión de Mamíferos/metabolismo , Técnica del Anticuerpo Fluorescente , Regulación del Desarrollo de la Expresión Génica , Ratones Endogámicos C57BL , Ratones Noqueados , Células Neuroepiteliales/metabolismo , Órgano Espiral/metabolismo , Receptores de Somatostatina/genética
13.
PLoS One ; 9(7): e100469, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25010045

RESUMEN

Somatostatin receptor subtype 2 (SSTR2) is the most frequently expressed SSTR subtype in normal human tissues. SSTR2 expression is differentially regulated in various tumor types and therapeutic somatostatin analogs binding to SSTR2 are in clinical use. In prostate cancers highly contradictory results in terms of SSTR2 expression and its consequences have been published over the past years. The aim of this study was to clarify prevalence and clinical significance of SSTR2 expression in prostate cancer. Therefore, quantitative immunohistochemistry (IHC) using a tissue microarray containing samples from 3,261 prostate cancer patients with extensive clinical and molecular cancer characteristics and oncological follow-up data was performed. IHC data was compared to publicly available Gene Expression Omnibus datasets of human prostate cancer gene expression arrays. While membranous SSTR2 staining was always seen in normal prostate epithelium, SSTR2 staining was absent in more than half (56.1%) of 2,195 interpretable prostate cancer samples. About 13% of all analyzed prostate cancers showed moderate to strong cytoplasmic and membranous SSTR2 staining. Staining intensities were inversely correlated with high Gleason grade, advanced pT category, high tumor cell proliferation (p<0.0001 each), high pre-operative PSA levels, (p = 0.0011) and positive surgical margins (p = 0.006). In silico analysis confirmed lower SSTR2 gene expression in prostate cancers vs. normal adjacent tissue (p = 0.0424), prostate cancer metastases vs. primary cancers (p = 0.0011) and recurrent vs. non-recurrent prostate cancers (p = 0.0438). PSA-free survival gradually declined with SSTR2 staining intensity (p<0.0001). SSTR2-negative cancers were more likely to develop metastases over time (p<0.05). In conclusion, most prostate cancers are indeed SSTR2-negative and loss of SSTR2 strongly predicts an unfavorable tumor phenotype and poor prognosis. Therefore, SSTR2 expression seems an important factor in the pathogenesis of prostate cancer and re-introduction of the receptor in SSTR2-negative prostate cancers may feature a promising target for novel gene therapy approaches.


Asunto(s)
Fenotipo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Receptores de Somatostatina/deficiencia , Anciano , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Neoplasias de la Próstata/diagnóstico , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Recurrencia
14.
Mol Pain ; 10: 12, 2014 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-24521084

RESUMEN

BACKGROUND: Somatostatin (SST) and some of its receptor subtypes have been implicated in pain signaling at the spinal level. In this study we have investigated the role of SST and its sst2A receptor (sst2A) in dorsal root ganglia (DRGs) and spinal cord. RESULTS: SST and sst2A protein and sst2 transcript were found in both mouse and human DRGs, sst2A-immunoreactive (IR) cell bodies and processes in lamina II in mouse and human spinal dorsal horn, and sst2A-IR nerve terminals in mouse skin. The receptor protein was associated with the cell membrane. Following peripheral nerve injury sst2A-like immunoreactivity (LI) was decreased, and SST-LI increased in DRGs. sst2A-LI accumulated on the proximal and, more strongly, on the distal side of a sciatic nerve ligation. Fluorescence-labeled SST administered to a hind paw was internalized and retrogradely transported, indicating that a SST-sst2A complex may represent a retrograde signal. Internalization of sst2A was seen in DRG neurons after systemic treatment with the sst2 agonist octreotide (Oct), and in dorsal horn and DRG neurons after intrathecal administration. Some DRG neurons co-expressed sst2A and the neuropeptide Y Y1 receptor on the cell membrane, and systemic Oct caused co-internalization, hypothetically a sign of receptor heterodimerization. Oct treatment attenuated the reduction of pain threshold in a neuropathic pain model, in parallel suppressing the activation of p38 MAPK in the DRGs CONCLUSIONS: The findings highlight a significant and complex role of the SST system in pain signaling. The fact that the sst2A system is found also in human DRGs and spinal cord, suggests that sst2A may represent a potential pharmacologic target for treatment of neuropathic pain.


Asunto(s)
Ganglios Espinales/patología , Receptores de Somatostatina/metabolismo , Ciática/metabolismo , Ciática/patología , Células Receptoras Sensoriales/metabolismo , Somatostatina/metabolismo , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Lateralidad Funcional/genética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Glutamato Descarboxilasa/genética , Proteínas Fluorescentes Verdes/deficiencia , Proteínas Fluorescentes Verdes/genética , Humanos , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Octreótido/uso terapéutico , Oligopéptidos/farmacología , Umbral del Dolor/efectos de los fármacos , Umbral del Dolor/fisiología , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , Receptores de Somatostatina/antagonistas & inhibidores , Receptores de Somatostatina/deficiencia , Receptores de Somatostatina/genética , Ciática/complicaciones , Ciática/tratamiento farmacológico , Células Receptoras Sensoriales/efectos de los fármacos , Somatostatina/genética
15.
Nat Neurosci ; 16(7): 845-7, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23708141
16.
Dev Neurosci ; 34(4): 342-53, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22986312

RESUMEN

The neuropeptide somatostatin (SST) exerts several important physiological actions in the adult central nervous system through interactions with membrane-bound receptors. Transient expression of SST and its receptors has been described in several brain areas during early ontogeny. It is therefore believed that SST may play a role in neural maturation. The present study provides the first evidence for the developmental expression of SST receptors in the mammalian cochlea, emphasizing their possible roles in cochlear maturation. In the developing mouse cochlea, cells immunoreactive to somatostatin receptor 1 (SSTR1) and somatostatin receptor 2 (SSTR2) were located in the embryonic cochlear duct on Kolliker's organ as early as embryonic day (E) 14 (E14). At E17, the expression of both receptors was high and already located at the hair cells and supporting cells along the length of the cochlear duct, which have become arranged into the characteristic pattern for the organ of Corti (OC) at this stage. At birth, SSTR1- and SSTR2-containing cells were only localized in the OC. In general, immunoreactivity for both receptors increased in the mouse cochlea from postnatal day (P) 0 (P0) to P10; the majority of immunostained cells were inner hair cells, outer hair cells, and supporting cells. Finally, a peak in the mRNA and protein expression of both receptors is present near the time when they respond to physiological hearing (i.e., hearing of airborne sound) at P14. At P21, SSTR1 and SSTR2 levels decrease dramatically. A similar developmental pattern was observed for SSTR1 and SSTR2 mRNA, suggesting that the expression of the SSTR1 and SSTR2 genes is controlled at the transcriptional level throughout development. In addition, we observed reduced levels of phospho-Akt and total Akt in SSTR1 knockout and SSTR1/SSTR2 double-knockout mice compared with wild-type mice. We know from previous studies that Akt is involved in hair cell survival. Taken together, the dynamic nature of SSTR1 and SSTR2 expression at a time of major developmental changes in the cochlea suggests that SSTR1 and SSTR2 (and possibly other members of this family) are involved in the maturation of the mammalian cochlea.


Asunto(s)
Cóclea/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas del Tejido Nervioso/biosíntesis , Receptores de Somatostatina/biosíntesis , Animales , Cóclea/embriología , Cóclea/crecimiento & desarrollo , Conducto Coclear/citología , Conducto Coclear/embriología , Conducto Coclear/crecimiento & desarrollo , Conducto Coclear/metabolismo , Células Epiteliales/metabolismo , Femenino , Edad Gestacional , Células Ciliadas Auditivas/metabolismo , Audición/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Órgano Espiral/citología , Órgano Espiral/embriología , Órgano Espiral/crecimiento & desarrollo , Órgano Espiral/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de Somatostatina/deficiencia , Receptores de Somatostatina/genética , Transcripción Genética
17.
J Neurochem ; 120(5): 818-29, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22168912

RESUMEN

In a retinal ischemic ex vivo model, we have reported protective effects of somatostatin (SRIF) receptor 2 (sst(2) ). As an ischemic condition not only causes cell death but also induces a vascular response, we asked whether vascular endothelial growth factor (VEGF) is altered in this model and whether its expression, release or localization are affected by sst(2) activation. Ex vivo retinas of wild-type (WT) and sst(1) KO mice (which over-express sst(2) ) were incubated in ischemic conditions with SRIF, octreotide (OCT) or a VEGF trap. Ischemia in WT retinas caused increase of VEGF release and decrease of VEGF mRNA. Both effects were counteracted by SRIF or OCT. VEGF immunoreactivity was in retinal neurons and scarcely in vessels. Ischemia caused a significant shift of VEGF immunoreactivity from neurons to vessels. The increase of vascular VEGF was reduced in sst(1) KO retinas and in WT retinas treated with SRIF or OCT. VEGF trap also limited this increase, demonstrating that vascular VEGF was of extracellular origin. Together, the data show a VEGF response to ischemia, in which VEGF released by damaged neurons reaches the retinal capillaries. The activation of sst(2) protects neurons from ischemic damage, thereby limiting VEGF release and the VEGF response.


Asunto(s)
Isquemia/patología , Retina/metabolismo , Somatostatina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Proteínas del Ojo/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Octreótido/efectos adversos , ARN Mensajero , Receptores de Somatostatina/deficiencia , Retina/patología , Vasos Retinianos/metabolismo , Somatostatina/farmacología , Factor A de Crecimiento Endotelial Vascular/genética
18.
PLoS One ; 6(9): e24467, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21912697

RESUMEN

BACKGROUND: Selective degeneration of medium spiny neurons and preservation of medium sized aspiny interneurons in striatum has been implicated in excitotoxicity and pathophysiology of Huntington's disease (HD). However, the molecular mechanism for the selective sparing of medium sized aspiny neurons and vulnerability of projection neurons is still elusive. The pathological characteristic of HD is an extensive reduction of the striatal mass, affecting caudate putamen. Somatostatin (SST) positive neurons are selectively spared in HD and Quinolinic acid/N-methyl-D-aspartic acid induced excitotoxicity, mimic the model of HD. SST plays neuroprotective role in excitotoxicity and the biological effects of SST are mediated by five somatostatin receptor subtypes (SSTR1-5). METHODS AND FINDINGS: To delineate subtype selective biological responses we have here investigated changes in SSTR1 and 5 double knockout mice brain and compared with HD transgenic mouse model (R6/2). Our study revealed significant loss of dopamine and cAMP regulated phosphoprotein of 32 kDa (DARPP-32) and comparable changes in SST, N-methyl-D-aspartic acid receptors subtypes, calbindin and brain nitric oxide synthase expression as well as in key signaling proteins including calpain, phospho-extracellular-signal-regulated kinases1/2, synapsin-IIa, protein kinase C-α and calcineurin in SSTR1/5(-/-) and R6/2 mice. Conversely, the expression of somatostatin receptor subtypes, enkephalin and phosphatidylinositol 3-kinases were strain specific. SSTR1/5 appears to be important in regulating NMDARs, DARPP-32 and signaling molecules in similar fashion as seen in HD transgenic mice. CONCLUSIONS: This is the first comprehensive description of disease related changes upon ablation of G- protein coupled receptor gene. Our results indicate that SST and SSTRs might play an important role in regulation of neurodegeneration and targeting this pathway can provide a novel insight in understanding the pathophysiology of Huntington's disease.


Asunto(s)
Encéfalo/metabolismo , Técnicas de Inactivación de Genes , Enfermedad de Huntington/metabolismo , Receptores de Somatostatina/deficiencia , Receptores de Somatostatina/genética , Animales , Encéfalo/patología , Calbindinas , Modelos Animales de Enfermedad , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Regulación de la Expresión Génica , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Masculino , Ratones , Ratones Transgénicos , Neostriado/metabolismo , Neuronas/metabolismo , Neuronas/patología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Proteína G de Unión al Calcio S100/metabolismo , Transducción de Señal/genética
19.
Neurosci Lett ; 484(2): 104-7, 2010 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-20709149

RESUMEN

The Melanin Concentrating Hormone (MCH) system is widely expressed throughout the central nervous system and regulates a variety of physiological functions. It has been reported that acute central administration of MCH inhibits pentylenetetrazol (PTZ)-induced seizures in rats. In the present study MCH(1) receptor knockout mice (MCH(1)R-KO) were used to investigate the role of MCH signaling in modulating seizure susceptibility. Seizure behaviors were compared between MCH(1)R-KO and wild type (MCH(1)R-WT) mice following administration of the convulsant compounds PTZ or pilocarpine. PTZ injection induced clonic seizures in MCH(1)R-WT mice but failed to induce them in MCH(1)R-KO mice. More than twice as many injections of intermittently administered low dose PTZ were required to induce clonic seizures in MCH(1)R-KO mice than in MCH(1)R-WT mice. Following pilocarpine injection, MCH(1)R-WT mice experienced clonic seizures and most had tonic seizures and entered status epilepticus, while all MCH(1)R-KO mice were completely resistant to these effects. MCH(1)R-KO mice were also observed to be strongly protected from the development of PTZ kindling. Genetic deletion of MCH(1)R conferred resistance to all seizure models tested in this study. The data indicate that the MCH system is involved in the regulation of PTZ and pilocarpine seizure threshold.


Asunto(s)
Receptores de Somatostatina/deficiencia , Convulsiones/genética , Animales , Modelos Animales de Enfermedad , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , N-Metilescopolamina , Pentilenotetrazol , Pilocarpina , Receptores de Somatostatina/genética , Convulsiones/inducido químicamente , Convulsiones/mortalidad , Convulsiones/patología
20.
J Neurophysiol ; 104(3): 1417-25, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20592115

RESUMEN

The hypothalamic neuropeptide melanin-concentrating hormone (MCH) plays important roles in energy homeostasis, anxiety, and sleep regulation. Since the MCH receptor-1 (MCH-R1), the only functional receptor that mediates MCH functions in rodents, facilitates behavioral performance in hippocampus-dependent learning tasks, we investigated whether glutamatergic transmission in CA1 pyramidal cells could be modulated in mice lacking the MCH-R1 gene (MCH-R1(-/-)). We found that both α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and N-methyl-d-aspartate (NMDA) receptor-mediated transmissions were diminished in the mutant mice compared with their controls. This deficit was explained, at least in part, by a postsynaptic down-regulation of these receptors since the amplitude of miniature excitatory postsynaptic currents and the NMDA/AMPA ratio were decreased. Long-term synaptic potentiation (LTP) was also impaired in MCH-R1(-/-) mice. This was due to an altered induction, rather than an impaired, expression because repeating the induction stimulus restored LTP to a normal magnitude. In addition, long-term synaptic depression was strongly diminished in MCH-R1(-/-) mice. These results suggest that MCH exerts a facilitatory effect on CA1 glutamatergic synaptic transmission and long-term synaptic plasticity. Recently, it has been shown that MCH neurons fire exclusively during sleep and mainly during rapid eye movement sleep. Thus these findings provide a mechanism by which sleep might facilitate memory consolidation.


Asunto(s)
Ácido Glutámico/fisiología , Hipocampo/fisiología , Plasticidad Neuronal/fisiología , Receptores de Somatostatina/fisiología , Sinapsis/fisiología , Transmisión Sináptica/fisiología , Animales , Potenciales Postsinápticos Excitadores/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Somatostatina/deficiencia , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA