Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Rev Endocr Metab Disord ; 24(2): 267-282, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36418657

RESUMEN

Neuroendocrine neoplasms (NENs) comprise a highly heterogeneous group of tumors arising from the diffuse neuroendocrine system. NENs mainly originate in gastrointestinal, pancreatic, and pulmonary tissues, and despite being rare, show rising incidence. The molecular mechanisms underlying NEN development are still poorly understood, although recent studies are unveiling their genomic, epigenomic and transcriptomic landscapes. RNA was originally considered as an intermediary between DNA and protein. Today, compelling evidence underscores the regulatory relevance of RNA processing, while new RNA molecules emerge with key functional roles in core cell processes. Indeed, correct functioning of the interrelated complementary processes comprising RNA biology, its processing, transport, and surveillance, is essential to ensure adequate cell homeostasis, and its misfunction is related to cancer at multiple levels. This review is focused on the dysregulation of RNA biology in NENs. In particular, we survey alterations in the splicing process and available information implicating the main RNA species and processes in NENs pathology, including their role as biomarkers, and their functionality and targetability. Understanding how NENs precisely (mis)behave requires a profound knowledge at every layer of their heterogeneity, to help improve NEN management. RNA biology provides a wide spectrum of previously unexplored processes and molecules that open new avenues for NEN detection, classification and treatment. The current molecular biology era is rapidly evolving to facilitate a detailed comprehension of cancer biology and is enabling the arrival of personalized, predictive and precision medicine to rare tumors like NENs.


Asunto(s)
Tumores Neuroendocrinos , ARN , Humanos , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/patología
2.
FASEB J ; 31(11): 4682-4696, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28705809

RESUMEN

sst5TMD4, a splice variant of the sst5 gene, is overexpressed and associated with aggressiveness in various endocrine-related tumors, but its presence, functional role, and mechanisms of actions in prostate cancer (PCa)-the most common cancer type in males-is completely unexplored. In this study, formalin-fixed, paraffin-embedded prostate pieces from patients with localized PCa, which included tumoral and nontumoral adjacent regions (n = 45), fresh biopsies from patients with high-risk PCa (n = 52), and healthy fresh prostates from cystoprostatectomies (n = 14) were examined. In addition, PCa cell lines and xenograft models were used to determine the presence and functional role of sst5TMD4. Results demonstrated that sst5TMD4 is overexpressed (mRNA/protein) in PCa samples, and this is especially drastic in metastatic and/or high Gleason score tumor samples. Remarkably, sst5TMD4 expression was associated with an altered frequency of 2 single-nucleotide polymorphisms: rs197055 and rs12599155. In addition, PCa cell lines and xenograft models were used to demonstrate that sst5TMD4 overexpression increases cell proliferation and migration in PCa cells and induces larger tumors in nude mice, whereas its silencing decreased proliferation and migration. Remarkably, sst5TMD4 overexpression activated multiple intracellular pathways (ERK/JNK, MYC/MAX, WNT, retinoblastoma), altered oncogenes and tumor suppressor gene expression, and disrupted the normal response to somatostatin analogs in PCa cells. Altogether, we demonstrate that sst5TMD4 is overexpressed in PCa, especially in those patients with a worse prognosis, and plays an important pathophysiologic role in PCa, which suggesting its potential as a biomarker and/or therapeutic target.-Hormaechea-Agulla, D., Jiménez-Vacas, J. M., Gómez-Gómez, E., L.-López, F., Carrasco-Valiente, J., Valero-Rosa, J., Moreno, M. M., Sánchez-Sánchez, R., Ortega-Salas, R., Gracia-Navarro, F., Culler, M. D., Ibáñez-Costa, A., Gahete, M. D., Requena, M. J., Castaño, J. P., Luque, R. M. The oncogenic role of the spliced somatostatin receptor sst5TMD4 variant in prostate cancer.


Asunto(s)
Empalme Alternativo , Regulación Neoplásica de la Expresión Génica , Sistema de Señalización de MAP Quinasas , Proteínas Oncogénicas , Neoplasias de la Próstata , Receptores de Somatostatina , Vía de Señalización Wnt , Anciano , Animales , Línea Celular Tumoral , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Metástasis de la Neoplasia , Trasplante de Neoplasias , Proteínas Oncogénicas/biosíntesis , Proteínas Oncogénicas/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Receptores de Somatostatina/biosíntesis , Receptores de Somatostatina/genética
3.
Am J Physiol Endocrinol Metab ; 306(5): E483-93, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24368669

RESUMEN

The majority of the biological actions attributed to somatostatin (SST) are thought to be mediated by SST receptor 2 (sst2), the most ubiquitous sst, and, to a lesser extent, by sst5. However, a growing body of evidence suggests a relevant role of sst1 in mediating SST actions in (patho)physiological situations (i.e., endometriosis, type 2 diabetes). Moreover, sst1 together with sst2 and sst5 is involved in the well-known actions of SST on pituitary somatotropes in pig and primates. Here, we cloned the porcine sst1 (psst1) and performed a structural and functional characterization using both primary and heterologous models. The psst1 sequence presents the majority of signature motifs shared among G protein-coupled receptors and, specifically, among ssts and exhibits a high homology with other mammalian sst1, with only minor differences in the amino-terminal domain, reinforcing the idea of an early evolutive divergence between mammalian and nonmammalian sst1s. psst1 is functional in terms of decreasing cAMP levels in response to SST when transfected in heterologous models. The psst1 receptor is expressed in several tissues, and analyses of gene cis elements predict regulation by multiple transcription factors and metabolic stimuli. Finally, psst1 is coexpressed with other sst subtypes in various tissues, and in vitro data demonstrate that psst1 can interact with itself forming homodimers and with other ssts forming heterodimers. These data highlight the functional importance of sst1 on the SST-mediated effects and its functional interaction with different ssts, which point out the necessity of exploring the consequences of such interactions.


Asunto(s)
Inflamación/metabolismo , Hipófisis/metabolismo , Receptores de Somatostatina/metabolismo , Somatostatina/metabolismo , Animales , Sitios de Unión , Regulación de la Expresión Génica , Inflamación/genética , Regiones Promotoras Genéticas , Receptores de Somatostatina/genética , Porcinos
4.
Mol Ther Nucleic Acids ; 35(1): 102090, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38187140

RESUMEN

Pancreatic neuroendocrine tumors (PanNETs) comprise a heterogeneous group of tumors with growing incidence. Recent molecular analyses provided a precise picture of their genomic and epigenomic landscape. Splicing dysregulation is increasingly regarded as a novel cancer hallmark influencing key tumor features. We have previously demonstrated that splicing machinery is markedly dysregulated in PanNETs. Here, we aimed to elucidate the molecular and functional implications of CUGBP ELAV-like family member 4 (CELF4), one of the most altered splicing factors in PanNETs. CELF4 expression was determined in 20 PanNETs, comparing tumor and non-tumoral adjacent tissue. An RNA sequencing (RNA-seq) dataset was analyzed to explore CELF4-linked interrelations among clinical features, gene expression, and splicing events. Two PanNET cell lines were employed to assess CELF4 function in vitro and in vivo. PanNETs display markedly upregulated CELF4 expression, which is closely associated with malignancy features, altered expression of key tumor players, and distinct splicing event profiles. Modulation of CELF4 influenced proliferation in vitro and reduced in vivo xenograft tumor growth. Interestingly, functional assays and RNA-seq analysis revealed that CELF4 silencing altered mTOR signaling pathway, enhancing the effect of everolimus. We demonstrate that CELF4 is dysregulated in PanNETs, where it influences tumor development and aggressiveness, likely by modulating the mTOR pathway, suggesting its potential as therapeutic target.

5.
Biochem J ; 443(2): 387-96, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22250954

RESUMEN

Golgi-associated long coiled-coil proteins, often referred to as golgins, are involved in the maintenance of the structural organization of the Golgi apparatus and the regulation of membrane traffic events occurring in this organelle. Little information is available on the contribution of golgins to Golgi function in cells specialized in secretion such as endocrine cells or neurons. In the present study, we characterize the intracellular distribution as well as the biochemical and functional properties of a novel long coiled-coil protein present in neuroendocrine tissues, NECC1 (neuroendocrine long coiled-coil protein 1). The present study shows that NECC1 is a peripheral membrane protein displaying high stability to detergent extraction, which distributes across the Golgi apparatus in neuroendocrine cells. In addition, NECC1 partially localizes to post-Golgi carriers containing secretory cargo in PC12 cells. Overexpression of NECC1 resulted in the formation of juxtanuclear aggregates together with a slight fragmentation of the Golgi and a decrease in K+-stimulated hormone release. In contrast, NECC1 silencing did not alter Golgi architecture, but enhanced K+-stimulated hormone secretion in PC12 cells. In all, the results of the present study identify NECC1 as a novel component of the Golgi matrix and support a role for this protein as a negative modulator of the regulated trafficking of secretory cargo in neuroendocrine cells.


Asunto(s)
Aparato de Golgi/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Transporte Biológico , Silenciador del Gen , Proteínas de Homeodominio/genética , Proteínas de la Membrana/genética , Células Neuroendocrinas/metabolismo , Células PC12 , Ratas
6.
Am J Physiol Endocrinol Metab ; 303(11): E1325-34, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23032684

RESUMEN

Somatostatin (SST) and its related peptide cortistatin (CORT) exert their multiple actions through binding to the SST receptor (sst) family, generally considered to comprise five G protein-coupled receptors with seven transmembrane domains (TMD), named sst1-sst5, plus a splice sst2B variant. However, we recently discovered that human and rodent sst5 gene expression also generates, through noncanonical alternative splicing, novel truncated albeit functional sst5 variants with less than seven TMD. Here, we cloned and characterized for the first time the porcine wild-type sst5 (psst5, full-length) and identified two novel truncated psst5 variants with six and three TMD, thus termed psst5TMD6 and psst5TMD3, respectively. In line with that observed in human and rodent truncated sst5 variants, psst5TMD6 and psst5TMD3 are functional (e.g., activate calcium signaling), selectively respond to SST and CORT, respectively, and exhibit specific tissue expression profiles that differ from full-length psst5 and often overlaps with psst2 expression. Moreover, fluorescence resonance energy transfer analysis shows that psst5 truncated variants physically interact with psst2, thereby altering their localization at the plasma membrane and specifically disrupting the cellular response to SST and/or CORT. These results represent the first characterization of a key porcine SST receptor, psst5, and, together with our previous results, provide strong evidence that alternative splicing-derived, truncated sst5 variants with distinct functional capacities exist in the mammalian lineage, where they can act as dominant-negative receptors, by interacting directly with long, seven TMD variants, potentially contributing to modulate normal and pathological SST and CORT signaling.


Asunto(s)
Señalización del Calcio/fisiología , Estructura Terciaria de Proteína/fisiología , Receptores de Somatostatina/fisiología , Transducción de Señal/fisiología , Empalme Alternativo , Animales , Células CHO , Clonación Molecular , Cricetinae , Humanos , Fragmentos de Péptidos , Ingeniería de Proteínas , Isoformas de Proteínas/fisiología , Porcinos , Distribución Tisular
7.
Gen Comp Endocrinol ; 175(1): 1-9, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21907200

RESUMEN

The regulated secretory pathway is a hallmark of endocrine and neuroendocrine cells. This process comprises different sequential steps, including ER-associated protein synthesis, ER-to-Golgi protein transport, Golgi-associated posttranslational modification, sorting and packing of secretory proteins into carrier granules, cytoskeleton-based granule transport towards the plasma membrane and tethering, docking and fusion of granules with specialized releasing zones in the plasma membrane. Each one of these steps is tightly regulated by a large number of factors that function in a spatially and temporarily coordinated fashion. During the past three decades, much effort has been devoted to characterize the precise role of the yet-known proteins participating in the different steps of this process and to identify new regulatory factors in order to obtain a unifying picture of the secretory pathway. In spite of this and given the enormous complexity of the process, certain steps are not fully understood yet and many players remain to be identified. In this review, we offer a summary of the current knowledge on the main molecular mechanisms that govern and ensure the correct release of secretory proteins. In addition, we have integrated the advance on the field made possible by studies carried out in non-mammalian vertebrates, which, although not very numerous, have substantially contributed to acquire a mechanistic understanding of the regulated secretory pathway.


Asunto(s)
Sistema Endocrino/fisiología , Células Neuroendocrinas/fisiología , Vías Secretoras/fisiología , Animales , Anuros , Retículo Endoplásmico/fisiología , Aparato de Golgi/fisiología , Humanos , Vesículas Secretoras/fisiología
8.
Endocrinology ; 147(6): 2902-8, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16543371

RESUMEN

It is well established that somatostatin acts through G protein-coupled receptors, termed sst, to inhibit GH release. However in pigs somatostatin can stimulate or inhibit in vitro GH secretion in a dose- and somatotrope subpopulation-dependent manner. We report herein that somatostatin-stimulated GH release is blocked by pretreatment with GTPgamma-S, suggesting an involvement of G protein-coupled receptors. Consistent with this, an sst5 selective agonist stimulated spontaneous GH secretion at doses ranging 10(-13) to 10(-9) m, without influencing GHRH-induced GH release. Conversely, sst1-, sst2-, sst3-, and sst4-specific agonists inhibited GHRH-evoked GH release but not basal GH secretion. Examination of the effects of sst-specific agonists on two subpopulations of somatotrope cells separated by density gradient centrifugation [low- (LD) and high-density (HD) cells] showed that only a low dose of the sst5 agonist stimulated GH release in LD somatotropes, whereas both low and high doses of this agonist stimulated GH release in HD cells. In marked contrast, sst1 and sst2 agonists blocked GHRH-stimulated GH release in LD cells at all doses tested, whereas only a high dose of the sst2 agonist inhibited GHRH-induced GH release in HD somatotropes. Interestingly, sst expression pattern in these subpopulations correlates with the distinct actions of sst-selective agonists; specifically, sst5 is more abundant in HD somatotropes, whereas sst1 and sst2 mRNA predominate in LD cells. These results indicate that in the pig, sst1 and sst2 are the primary mediators of the inhibitory effects of somatostatin, whereas sst5 or an sst5-related mechanism mediates the stimulatory action of somatostatin on GH release.


Asunto(s)
Hormona del Crecimiento/metabolismo , Receptores de Somatostatina/fisiología , Somatostatina/farmacología , Animales , Femenino , Hormona Liberadora de Hormona del Crecimiento/farmacología , Guanosina 5'-O-(3-Tiotrifosfato)/farmacología , ARN Mensajero/análisis , Receptores de Somatostatina/clasificación , Receptores de Somatostatina/genética , Porcinos
9.
Oncotarget ; 7(37): 60110-60122, 2016 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-27507050

RESUMEN

The truncated somatostatin receptor sst5TMD4 is associated with poor prognosis in breast cancer and increases breast cancer cell malignancy. Here, we examined the cellular/molecular mechanisms underlying this association, aiming to identify new molecular tools to improve diagnosis, prognosis or therapy. A gene expression array comparing sst5TMD4 stably-transfected MCF-7 cells and their controls (empty-plasmid) revealed the existence of profound alterations in the expression of genes involved in key tumoral processes, such as cell survival or angiogenesis. Moreover, sst5TMD4-overexpressing MCF-7 and MDA-MB-231 cells demonstrated increased expression/production of pro-angiogenic factors and enhanced capacity to form mammospheres. Consistently, sst5TMD4-expressing MCF-7 cells induced xenografted tumors with higher VEGF levels and elevated number of blood vessels. Importantly, sst5TMD4 was expressed in a subset of breast cancers, where it correlated with angiogenic markers, lymphatic metastasis, and reduced disease-free survival. These results, coupled to our previous data, support a relevant role of sst5TMD4 in the angiogenic process and reinforce the role of sst5TMD4 in breast cancer malignancy and metastatic potential, supporting its possible utility to develop new molecular biomarkers and drug therapies for these tumors.


Asunto(s)
Neoplasias de la Mama/genética , Mutación , Neovascularización Patológica/genética , Receptores de Somatostatina/genética , Adulto , Anciano , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Metástasis Linfática , Células MCF-7 , Ratones Desnudos , Persona de Mediana Edad , Neovascularización Patológica/metabolismo , Receptores de Somatostatina/metabolismo , Trasplante Heterólogo
10.
Endocrinology ; 145(7): 3182-9, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15044357

RESUMEN

Secretion of GH by pituitary somatotropes is primarily stimulated by the hypothalamic GHRH through the activation of a specific G protein-coupled receptor, GHRH receptor (GHRH-R). GH is also released in response to ghrelin, a peptide produced in the stomach, hypothalamus, and pituitary that activates somatotropes via a distinct G protein-coupled receptor, referred to as the GH secretagogue receptor (GHS-R). Here, we have analyzed the expression of both GHRH-R and GHS-R (by multiplex RT-PCR) in porcine pituitary cell cultures, after acute (4 h) treatment with GHRH or ghrelin as well as with other regulators of somatotropes (somatostatin, dexamethasone). Exposure of cultures to GHRH decreased GHRH-R mRNA content and also diminished GHS-R transcript levels. Likewise, ghrelin down-regulated both GHS-R and GHRH-R expression. Interestingly, administration of the activator of adenylate cyclase, forskolin, decreased GHRH-R mRNA levels but had no effect on GHS-R, thus suggesting a distinct contribution of the various intracellular signals operating in somatotropes to the regulation of the expression of these receptors. Accordingly, an atypical activator of adenylate cyclase in the pig somatotrope is low-dose (10(-13) m) somatostatin, which also suppressed GHRH-R mRNA levels without altering GHS-R expression. Finally, dexamethasone did not modify GHRH-R or GHS-R expression. In summary, our data show for the first time that ghrelin, as well as GHRH, mediates homologous and heterologous down-regulation of their own receptor synthesis. However, our results also indicate that the expression of porcine GHRH-R and GHS-R is regulated by distinct signals that may differ from those reported in other mammalian species.


Asunto(s)
Hipófisis/fisiología , Receptores Acoplados a Proteínas G/genética , Receptores de Neuropéptido/genética , Receptores de Hormona Reguladora de Hormona Hipofisaria/genética , Animales , Células Cultivadas , Colforsina/farmacología , Dexametasona/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Ghrelina , Glucocorticoides/farmacología , Hormona Liberadora de Hormona del Crecimiento/farmacología , Hormonas Peptídicas/farmacología , Hipófisis/citología , ARN Mensajero/análisis , Receptores de Ghrelina , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/normas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Somatostatina/farmacología , Sus scrofa
11.
Endocrinology ; 144(12): 5372-80, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12960033

RESUMEN

Ghrelin is a newly discovered peptide that binds the receptor for GH secretagogues (GHS-R). The presence of both ghrelin and GHS-Rs in the hypothalamic-pituitary system, together with the ability of ghrelin to increase GH release, suggests a hypophysiotropic role for this peptide. To ascertain the intracellular mechanisms mediating the action of ghrelin in somatotropes, we evaluated ghrelin-induced GH release from pig pituitary cells both under basal conditions and after specific blockade of key steps of cAMP-, inositol phosphate-, and Ca2+-dependent signaling routes. Ghrelin stimulated GH release at concentrations ranging from 10-10 to 10-6 m. Its effects were comparable with those exerted by GHRH or the GHS L-163,255. Combined treatment with ghrelin and GHRH or L-163,255 did not cause further increases in GH release, whereas somatostatin abolished the effect of ghrelin. Blockade of phospholipase C or protein kinase C inhibited ghrelin-induced GH secretion, suggesting a requisite role for this route in ghrelin action. Unexpectedly, inhibition of either adenylate cyclase or protein kinase A also suppressed ghrelin-induced GH release. In addition, ghrelin stimulated cAMP production and also had an additive effect with GHRH on cAMP accumulation. Ghrelin also increased free intracellular Ca2+ levels in somatotropes. Moreover, ghrelin-induced GH release was entirely dependent on extracellular Ca2+ influx through L-type voltage-sensitive channels. These results indicate that ghrelin exerts a direct stimulatory action on porcine GH release that is not additive with that of GHRH and requires the contribution of a multiple, complex set of interdependent intracellular signaling pathways.


Asunto(s)
Hormona del Crecimiento/metabolismo , Hormonas Peptídicas/farmacología , Hipófisis/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Adenilil Ciclasas/metabolismo , Animales , Calcio/metabolismo , Calcio/farmacología , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Interacciones Farmacológicas , Femenino , Ghrelina , Hormona Liberadora de Hormona del Crecimiento/farmacología , Hipófisis/citología , Hipófisis/efectos de los fármacos , Proteína Quinasa C/metabolismo , Somatostatina/farmacología , Sus scrofa , Fosfolipasas de Tipo C/metabolismo
12.
Endocrinology ; 143(3): 889-97, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11861510

RESUMEN

Somatostatin (SRIF) inhibits GH release from rat somatotropes by reducing adenylate cyclase (AC) activity and the free cytosolic calcium concentration ([Ca(2+)](i)). In contrast, we have reported that SRIF can stimulate GH release in vitro from pig somatotropes. Specifically, 10(-7) and 10(-15) M SRIF stimulate GH release from a subpopulation of high density (HD) somatotropes isolated by Percoll gradient centrifugation, whereas in low density (LD) somatotropes only 10(-15) M SRIF induces such an effect. To ascertain the signaling pathways underlying this phenomenon, we assessed SRIF effects on second messengers in cultured LD and HD cells by measuring cAMP, IP turnover, and [Ca(2+)](i). Likewise, contribution of the corresponding signaling pathways to SRIF-induced GH release was evaluated by blocking AC, PLC, extracellular Ca(2+) influx, or intracellular Ca(2+) mobilization. Both 10(-7) and 10(-15) M SRIF increased cAMP, IP turnover, and [Ca(2+)](i) in HD cells. Conversely, in LD cells 10(-7) M SRIF reduced [Ca(2+)](i), but did not alter cAMP or IP, and 10(-15) M SRIF was without effect. Interestingly, SRIF-stimulated GH release was abolished in both subpopulations by AC blockade, but not by PLC inhibition. Furthermore, SRIF-induced GH release was not reduced by blockade of extracellular Ca(2+) influx through voltage-sensitive channels or by depletion of thapsigargin-sensitive intracellular Ca(2+) stores. Therefore, SRIF stimulates GH secretion from cultured porcine somatotrope subpopulations through an AC/cAMP pathway-dependent mechanism that is seemingly independent of net increases in IP turnover or [Ca(2+)](i). These novel actions challenge classic views of SRIF as a mere inhibitor for somatotropes and suggest that it may exert a more complex, dual function in the control of porcine GH release, wherein molecular heterogeneity of somatotropes would play a critical role.


Asunto(s)
AMP Cíclico/fisiología , Hormona del Crecimiento/metabolismo , Hipófisis/metabolismo , Transducción de Señal/fisiología , Somatostatina/farmacología , Animales , Calcio/metabolismo , Calcio/fisiología , Bloqueadores de los Canales de Calcio/farmacología , Células Cultivadas , Femenino , Inositol/metabolismo , Hipófisis/citología , Hipófisis/efectos de los fármacos , Porcinos
13.
Regul Pept ; 116(1-3): 43-52, 2003 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-14599714

RESUMEN

Substance P has been previously shown to stimulate luteinizing hormone (LH) secretion and synergistically enhance gonadotropin-releasing hormone (GnRH)-evoked LH release from cultured pig pituitary cells. To investigate the mechanisms involved in these responses, the effects of substance P (100 nM; 4 h) and/or GnRH (10 nM, 4 h) on LH release, LH intracellular content, and betaLH mRNA accumulation were evaluated in the absence or presence of extracellular Ca(2+). Likewise, the effects of substance P on the dynamics of cytosolic free Ca(2+) concentration ([Ca(2+)](i)) were examined in single cells. Extracellular Ca(2+) deprivation abolished both substance P- and GnRH-stimulated LH release, as well as their synergistic interaction. The substance P antagonist D-Arg1,D-Phe5,-Trp7,9,Leu11-substance P (100 nM) blocked the stimulatory effect of substance P on LH release and its interaction with GnRH without affecting GnRH-induced LH secretion. Whereas substance P did not modify betaLH transcript levels, GnRH stimulated betaLH mRNA accumulation through a mechanism dependent upon extracellular Ca(2+). Substance P directly increased [Ca(2+)](i) in a 30% of gonadotropes by causing two distinct types of response kinetics with single-peak (predominant, 83.3%) or sustained-plateau profiles. Reduction of external [Ca(2+)] decreased by half the percent of responsive cells, which only showed single-peak profiles. Taken together, our results demonstrate that the ability of substance P to stimulate basal and GnRH-induced LH release is exerted directly upon gonadotropes, is extracellular Ca(2+)-dependent and does not seem to require net increases in betaLH mRNA levels. Moreover, [Ca(2+)](i) measurements revealed that although substance P action in pig gonadotropes is strongly dependent on extracellular Ca(2+) influx, it would also involve intracellular Ca(2+) mobilization. Finally, extracellular Ca(2+) also plays a requisite role to sustain GnRH-stimulated increases in both betaLH mRNA and LH release.


Asunto(s)
Calcio/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Hipófisis/efectos de los fármacos , Sustancia P/farmacología , Animales , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Femenino , Hormona Luteinizante de Subunidad beta/genética , Hormona Luteinizante de Subunidad beta/metabolismo , Hipófisis/citología , Hipófisis/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Porcinos
14.
J Endocrinol ; 220(1): R1-24, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24194510

RESUMEN

Ghrelin is a 28-amino acid acylated hormone, highly expressed in the stomach, which binds to its cognate receptor (GHSR1a) to regulate a plethora of relevant biological processes, including food intake, energy balance, hormonal secretions, learning, inflammation, etc. However, ghrelin is, in fact, the most notorious component of a complex, intricate regulatory system comprised of a growing number of alternative peptides (e.g. obestatin, unacylated ghrelin, and In1-ghrelin, etc.), known (GHSRs) and, necessarily unknown receptors, as well as modifying enzymes (e.g. ghrelin-O-acyl-transferase), which interact among them as well as with other regulatory systems in order to tightly modulate key (patho)-physiological processes. This multiplicity of functions and versatility of the ghrelin system arise from a dual, genetic and functional, complexity. Importantly, a growing body of evidence suggests that dysregulation in some of the components of the ghrelin system can lead to or influence the development and/or progression of highly concerning pathologies such as endocrine-related tumors, inflammatory/cardiovascular diseases, and neurodegeneration, wherein these altered components could be used as diagnostic, prognostic, or therapeutic targets. In this context, the aim of this review is to integrate and comprehensively analyze the multiple components and functions of the ghrelin system described to date in order to define and understand its biological and (patho)-physiological significance.


Asunto(s)
Aciltransferasas/metabolismo , Ghrelina/metabolismo , Receptores de Ghrelina/metabolismo , Transducción de Señal , Aciltransferasas/genética , Sistema Endocrino/metabolismo , Sistema Endocrino/patología , Sistema Endocrino/fisiopatología , Ghrelina/genética , Humanos , Modelos Biológicos , Receptores de Ghrelina/genética
15.
Mol Nutr Food Res ; 58(9): 1897-906, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24995559

RESUMEN

SCOPE: Dietary fat influences systemic inflammatory status, which determines the progression of age-associated diseases. Since somatostatin (SST), cortistatin (CORT), and ghrelin systems modulate inflammatory response, we aim to comprehensively characterize the presence and regulation of the components of these systems in the peripheral blood mononuclear cells (PMBCs), a subset of white blood cells placed at the crossroad between diet and inflammation, in response to diets with different fat composition, and during the postprandial phase in elderly subjects. METHODS AND RESULTS: The applied nutrigenomic, inflammation-related PBMC-based approach revealed that the majority of components of SST/CORT and ghrelin systems are present in the human PBMCs. Particularly, CORT, SST/CORT receptors (sst2, sst3, sst5, and sst5TMD4), ghrelin, its acylating enzyme (GOAT), In1-ghrelin variant, and GHSR1b were detected in PBMCs. Their expression was altered in the long-term by diet composition, and in the short-term, during the postprandial phase. Of particular relevance is the postprandial elevation of CORT, sst2, and sst5 expression in PBMCs of subjects under n-3 PUFAs-enriched diet. CONCLUSION: Our results suggest a potential relevant role of CORT/ssts and ghrelin systems in regulating PBMCs response to nutrient intake, which could help to explain the positive effects of n-3 PUFAs-enriched diets in reducing the inflammatory response.


Asunto(s)
Grasas de la Dieta/farmacología , Ghrelina/sangre , Leucocitos Mononucleares/efectos de los fármacos , Neuropéptidos/sangre , Periodo Posprandial/efectos de los fármacos , Anciano , Dieta Mediterránea , Dieta Occidental , Ácidos Grasos Omega-3/farmacología , Femenino , Ghrelina/metabolismo , Humanos , Inflamación/metabolismo , Inflamación/prevención & control , Leucocitos Mononucleares/fisiología , Masculino , Neuropéptidos/genética , Neuropéptidos/metabolismo , Nutrigenómica/métodos , Receptores de Somatostatina/genética , Somatostatina/genética
16.
Endocrinology ; 155(4): 1407-17, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24484169

RESUMEN

Obestatin is a 23-amino-acid amidated peptide that is encoded by the ghrelin gene. Previous studies have shown obestatin can modulate the hypothalamic neuronal circuitry that regulates pituitary function, perhaps by modulating the actions of ghrelin. However, the direct actions of obestatin on pituitary function remain controversial. Here, primary pituitary cell cultures from a nonhuman primate (baboon) and mice were used to test the effects of obestatin on pituitary hormone expression and secretion. In pituitary cultures from both species, obestatin had no effect on prolactin, LH, FSH, or TSH expression/release. Conversely, obestatin stimulated proopiomelanocortin expression and ACTH release and inhibited GH expression/release in vitro, actions that were also observed in vivo in mice treated with obestatin. In vitro, obestatin inhibited the stimulatory actions of ghrelin on GH but not ACTH release. The inhibitory effect of obestatin on somatotrope function was associated with an overall reduction in pituitary transcription factor-1 and GHRH receptor mRNA levels in vitro and in vivo as well as a reduction in hypothalamic GHRH and ghrelin expression in vivo. The stimulatory effect of obestatin on ACTH was associated with an increase in pituitary CRF receptors. Obestatin also reduced the expression of pituitary somatostatin receptors (sst1/sst2), which could serve to modify its impact on hormone secretion. The in vitro actions of obestatin on both GH and ACTH release required the adenylyl cyclase and MAPK routes. Taken together, our results provide evidence that obestatin can act directly at the pituitary to control somatotrope and corticotrope function, and these effects are conserved across species.


Asunto(s)
Corticotrofos/metabolismo , Regulación de la Expresión Génica , Ghrelina/metabolismo , Hipófisis/metabolismo , Somatotrofos/metabolismo , Animales , Supervivencia Celular , Femenino , Ghrelina/química , Hormona del Crecimiento/metabolismo , Hormonas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Papio anubis , Proopiomelanocortina/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal , Somatostatina/química
17.
PLoS One ; 8(2): e57834, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23469081

RESUMEN

Ghrelin-system components [native ghrelin, In1-ghrelin, Ghrelin-O-acyltransferase enzyme (GOAT) and receptors (GHS-Rs)] are expressed in a wide variety of tissues, including the pancreas, where they exert different biological actions including regulation of neuroendocrine secretions, food intake and pancreatic function. The expression of ghrelin system is regulated by metabolic conditions (fasting/obesity) and is associated with the progression of obesity and insulin resistance. Cortistatin (CORT), a neuropeptide able to activate GHS-R, has emerged as an additional link in gut-brain interplay. Indeed, we recently reported that male CORT deficient mice (cort-/-) are insulin-resistant and present a clear dysregulation in the stomach ghrelin-system. The present work was focused at analyzing the expression pattern of ghrelin-system components at pancreas level in cort-/- mice and their control littermates (cort +/+) under low- or high-fat diet. Our data reveal that all the ghrelin-system components are expressed at the mouse pancreatic level, where, interestingly, In1-ghrelin was expressed at higher levels than native-ghrelin. Thus, GOAT mRNA levels were significantly lower in cort-/- mice compared with controls while native ghrelin, In1-ghrelin and GHS-R transcript levels remained unaltered under normal metabolic conditions. Moreover, under obese condition, a significant increase in pancreatic expression of native-ghrelin, In1-ghrelin and GHS-R was observed in obese cort+/+ but not in cort-/- mice. Interestingly, insulin expression and release was elevated in obese cort+/+, while these changes were not observed in obese cort-/- mice. Altogether, our results indicate that the ghrelin-system expression is clearly regulated in the pancreas of cort+/+ and cort -/- under normal and/or obesity conditions suggesting that this system may play relevant roles in the endocrine pancreas. Most importantly, our data demonstrate, for the first time, that endogenous CORT is essential for the obesity-induced changes in insulin expression/secretion observed in mice, suggesting that CORT is a key regulatory component of the pancreatic function.


Asunto(s)
Regulación de la Expresión Génica , Ghrelina/metabolismo , Islotes Pancreáticos/metabolismo , Neuropéptidos/metabolismo , Obesidad/metabolismo , Animales , Metabolismo Basal , Técnicas de Inactivación de Genes , Insulina/sangre , Masculino , Ratones , Neuropéptidos/deficiencia , Neuropéptidos/genética
18.
PLoS One ; 8(9): e73668, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24040018

RESUMEN

TrkA-mediated NGF signaling in PC12 cells has been shown to be compartimentalized in specialized microdomains of the plasma membrane, the caveolae, which are organized by scaffold proteins including the member of the caveolin family of proteins, caveolin-1. Here, we characterize the intracellular distribution as well as the biochemical and functional properties of the neuroendocrine long coiled-coil protein 2 (NECC2), a novel long coiled-coil protein selectively expressed in neuroendocrine tissues that contains a predicted caveolin-binding domain and displays structural characteristics of a scaffolding factor. NECC2 distributes in caveolae, wherein it colocalizes with the TrkA receptor, and behaves as a caveolae-associated protein in neuroendocrine PC12 cells. In addition, stimulation of PC12 cells with nerve growth factor (NGF) increased the expression and regulated the distribution of NECC2. Interestingly, knockdown as well as overexpression of NECC2 resulted in a reduction of NGF-induced phosphorylation of the TrkA downstream effector extracellular signal-regulated kinases 1 and 2 (ERK1/ERK2) but not of Akt. Altogether, our results identify NECC2 as a novel component of caveolae in PC12 cells and support the contribution of this protein in the maintenance of TrkA-mediated NGF signaling.


Asunto(s)
Caveolas/metabolismo , Proteínas de la Membrana/metabolismo , Factor de Crecimiento Nervioso/farmacología , Receptor trkA/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Caveolina 1/genética , Caveolina 1/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Immunoblotting , Proteínas de la Membrana/genética , Microscopía Confocal , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Células PC12 , Fosforilación/efectos de los fármacos , Interferencia de ARN , Ratas
19.
Artículo en Inglés | MEDLINE | ID: mdl-23162532

RESUMEN

Somatostatin (SST), cortistatin (CORT), and its receptors (sst1-5), and ghrelin and its receptors (GHS-R) are two highly interrelated neuropeptide systems with a broad range of overlapping biological actions at central, cardiovascular, and immune levels among others. Besides their potent regulatory role on GH release, its endocrine actions are highlighted by SST/CORT and ghrelin influence on insulin secretion, glucose homeostasis, and insulin resistance. Interestingly, most components of these systems are expressed at the endocrine pancreas and are actively involved in the modulation of pancreatic islet function and, consequently influence glucose homeostasis. In addition, some of them also participate in islet survival and regeneration. Furthermore, under severe metabolic condition as well as in endocrine pathologies, their expression profile is severely deregulated. These findings suggest that SST/CORT and ghrelin systems could play a relevant role in pancreatic function under metabolic and endocrine pathologies. Accordingly, these systems have been therapeutically targeted for the prevention or amelioration of certain metabolic conditions (obesity) as well as for tumor growth inhibition and/or hormonal regulation in endocrine pathologies (neuroendocrine tumors). This review focuses on the interrelationship between SST/CORT and ghrelin systems and their role in severe metabolic conditions and some endocrine disorders.

20.
PLoS One ; 6(7): e22931, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21829560

RESUMEN

Lipid droplets (LDs) are organelles that coordinate lipid storage and mobilization, both processes being especially important in cells specialized in managing fat, the adipocytes. Proteomic analyses of LDs have consistently identified the small GTPase Rab18 as a component of the LD coat. However, the specific contribution of Rab18 to adipocyte function remains to be elucidated. Herein, we have analyzed Rab18 expression, intracellular localization and function in relation to the metabolic status of adipocytes. We show that Rab18 production increases during adipogenic differentiation of 3T3-L1 cells. In addition, our data show that insulin induces, via phosphatidylinositol 3-kinase (PI3K), the recruitment of Rab18 to the surface of LDs. Furthermore, Rab18 overexpression increased basal lipogenesis and Rab18 silencing impaired the lipogenic response to insulin, thereby suggesting that this GTPase promotes fat accumulation in adipocytes. On the other hand, studies of the ß-adrenergic receptor agonist isoproterenol confirmed and extended previous evidence for the participation of Rab18 in lipolysis. Together, our data support the view that Rab18 is a common mediator of lipolysis and lipogenesis and suggests that the endoplasmic reticulum (ER) is the link that enables Rab18 action on these two processes. Finally, we describe, for the first time, the presence of Rab18 in human adipose tissue, wherein the expression of this GTPase exhibits sex- and depot-specific differences and is correlated to obesity. Taken together, these findings indicate that Rab18 is involved in insulin-mediated lipogenesis, as well as in ß-adrenergic-induced lipolysis, likely facilitating interaction of LDs with ER membranes and the exchange of lipids between these compartments. A role for Rab18 in the regulation of adipocyte biology under both normal and pathological conditions is proposed.


Asunto(s)
Adipocitos/metabolismo , Lipogénesis , Lipólisis , Obesidad/fisiopatología , Proteínas de Unión al GTP rab/fisiología , Células 3T3-L1 , Adipocitos/citología , Adipocitos/efectos de los fármacos , Adiponectina/metabolismo , Animales , Western Blotting , Células Cultivadas , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Femenino , Humanos , Hipoglucemiantes/farmacología , Técnicas para Inmunoenzimas , Insulina/farmacología , Metabolismo de los Lípidos , Masculino , Ratones , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA