Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 10(7): e1004488, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25058410

RESUMEN

We carried out an admixture analysis of a sample comprising 1,019 individuals from all the provinces of Cuba. We used a panel of 128 autosomal Ancestry Informative Markers (AIMs) to estimate the admixture proportions. We also characterized a number of haplogroup diagnostic markers in the mtDNA and Y-chromosome in order to evaluate admixture using uniparental markers. Finally, we analyzed the association of 16 single nucleotide polymorphisms (SNPs) with quantitative estimates of skin pigmentation. In the total sample, the average European, African and Native American contributions as estimated from autosomal AIMs were 72%, 20% and 8%, respectively. The Eastern provinces of Cuba showed relatively higher African and Native American contributions than the Western provinces. In particular, the highest proportion of African ancestry was observed in the provinces of Guantánamo (40%) and Santiago de Cuba (39%), and the highest proportion of Native American ancestry in Granma (15%), Holguín (12%) and Las Tunas (12%). We found evidence of substantial population stratification in the current Cuban population, emphasizing the need to control for the effects of population stratification in association studies including individuals from Cuba. The results of the analyses of uniparental markers were concordant with those observed in the autosomes. These geographic patterns in admixture proportions are fully consistent with historical and archaeological information. Additionally, we identified a sex-biased pattern in the process of gene flow, with a substantially higher European contribution from the paternal side, and higher Native American and African contributions from the maternal side. This sex-biased contribution was particularly evident for Native American ancestry. Finally, we observed that SNPs located in the genes SLC24A5 and SLC45A2 are strongly associated with melanin levels in the sample.


Asunto(s)
Flujo Génico/genética , Genética de Población , Haplotipos/genética , Pigmentación/genética , Población Negra/genética , Cromosomas Humanos Y/genética , Cuba , ADN Mitocondrial/genética , Hispánicos o Latinos/genética , Humanos , Indígenas Norteamericanos/genética , Polimorfismo de Nucleótido Simple/genética , Población Blanca/genética
2.
Cancer Lett ; 584: 216604, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38244911

RESUMEN

Novel biomarkers and therapeutic strategies for prostate-cancer (PCa) are required to overcome its lethal progression. The dysregulation/implication of the RNA-Exosome-complex (REC; cellular machinery controlling the 3'-5'processing/degradation of most RNAs) in different cancer-types, including PCa, is poorly known. Herein, different cellular/molecular/preclinical approaches with human PCa-samples (tissues and/or plasma of 7 independent cohorts), and in-vitro/in-vivo PCa-models were used to comprehensively characterize the REC-profile and explore its role in PCa. Moreover, isoginkgetin (REC-inhibitor) effects were evaluated on PCa-cells. We demonstrated a specific dysregulation of the REC-components in PCa-tissues, identifying the Poly(A)-Binding-Protein-Nuclear 1 (PABPN1) factor as a critical regulator of major cancer hallmarks. PABPN1 is consistently overexpressed in different human PCa-cohorts and associated with poor-progression, invasion and metastasis. PABPN1 silencing decreased relevant cancer hallmarks in multiple PCa-models (proliferation/migration/tumourspheres/colonies, etc.) through the modulation of key cancer-related lncRNAs (PCA3/FALEC/DLEU2) and mRNAs (CDK2/CDK6/CDKN1A). Plasma PABPN1 levels were altered in patients with metastatic and tumour-relapse. Finally, pharmacological inhibition of REC-activity drastically inhibited PCa-cell aggressiveness. Altogether, the REC is drastically dysregulated in PCa, wherein this novel molecular event/mechanism, especially PABPN1 alteration, may be potentially exploited as a novel prognostic and therapeutic tool for PCa.


Asunto(s)
Exosomas , Neoplasias de la Próstata , Masculino , Humanos , Complejo Multienzimático de Ribonucleasas del Exosoma , Exosomas/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Recurrencia Local de Neoplasia , Neoplasias de la Próstata/patología , ARN Mensajero , Proteína I de Unión a Poli(A)/metabolismo
3.
Sci Adv ; 10(40): eado8231, 2024 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-39356765

RESUMEN

Despite novel therapeutic strategies, advanced-stage prostate cancer (PCa) remains highly lethal, pointing out the urgent need for effective therapeutic strategies. While dysregulation of the splicing process is considered a cancer hallmark, the role of certain splicing factors remains unknown in PCa. This study focuses on characterizing the levels and role of SRSF6 in this disease. Comprehensive analyses of SRSF6 alterations (copy number/mRNA/protein) were conducted across eight well-characterized PCa cohorts and the Hi-MYC transgenic model. SRSF6 was up-regulated in PCa samples, correlating with adverse clinical parameters. Functional assays, both in vitro (cell proliferation, migration, colony, and tumorsphere formation) and in vivo (xenograft tumors), demonstrated the impact of SRSF6 modulation on critical cancer hallmarks. Mechanistically, SRSF6 regulates the splicing pattern of the histone-chaperone HIRA, consequently affecting the activity of H3.3 in PCa and breast cancer cell models and disrupting pivotal oncogenic pathways (AR and E2F) in PCa cells. These findings underscore SRSF6 as a promising therapeutic target for PCa/advanced-stage PCa.


Asunto(s)
Chaperonas de Histonas , Neoplasias de la Próstata , Factores de Empalme Serina-Arginina , Humanos , Factores de Empalme Serina-Arginina/metabolismo , Factores de Empalme Serina-Arginina/genética , Masculino , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Animales , Línea Celular Tumoral , Chaperonas de Histonas/metabolismo , Chaperonas de Histonas/genética , Receptores Androgénicos/metabolismo , Receptores Androgénicos/genética , Regulación Neoplásica de la Expresión Génica , Ratones , Empalme del ARN , Proliferación Celular , Histonas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Fosfoproteínas
4.
Wiley Interdiscip Rev RNA ; 14(3): e1760, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36063028

RESUMEN

Bladder cancer is the most common malignancy of the urinary tract worldwide. The therapeutic options to tackle this disease comprise surgery, intravesical or systemic chemotherapy, and immunotherapy. Unfortunately, a wide number of patients ultimately become resistant to these treatments and develop aggressive metastatic disease, presenting a poor prognosis. Therefore, the identification of novel therapeutic approaches to tackle this devastating pathology is urgently needed. However, a significant limitation is that the progression and drug response of bladder cancer is strongly associated with its intrinsic molecular heterogeneity. In this sense, RNA splicing is recently gaining importance as a critical hallmark of cancer since can have a significant clinical value. In fact, a profound dysregulation of the splicing process has been reported in bladder cancer, especially in the expression of certain key splicing variants and circular RNAs with a potential clinical value as diagnostic/prognostic biomarkers or therapeutic targets in this pathology. Indeed, some authors have already evidenced a profound antitumor effect by targeting some splicing factors (e.g., PTBP1), mRNA splicing variants (e.g., PKM2, HYAL4-v1), and circular RNAs (e.g., circITCH, circMYLK), which illustrates new possibilities to significantly improve the management of this pathology. This review represents the first detailed overview of the splicing process and its alterations in bladder cancer, and highlights opportunities for the development of novel diagnostic/prognostic biomarkers and their clinical potential for the treatment of this devastating cancer type. This article is categorized under: RNA Processing > Splicing Regulation/Alternative Splicing RNA in Disease and Development > RNA in Disease.


Asunto(s)
Empalme Alternativo , Neoplasias de la Vejiga Urinaria , Humanos , Empalme Alternativo/genética , ARN Circular/genética , ARN Circular/metabolismo , Neoplasias de la Vejiga Urinaria/diagnóstico , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/terapia , ARN/metabolismo , Biomarcadores/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Proteína de Unión al Tracto de Polipirimidina/genética , Proteína de Unión al Tracto de Polipirimidina/metabolismo
5.
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
6.
Gastroenterology ; 136(5): 1783-95.e10, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19208340

RESUMEN

BACKGROUND & AIMS: Despite high morbidity and mortality of alcoholic liver disease worldwide, the molecular mechanisms underlying alcohol-induced liver cell death are not fully understood. Transglutaminase 2 (TG2) is a cross-linking enzyme implicated in apoptosis. TG2 levels and activity are increased in association with various types of liver injury. However, how TG2 induces hepatic apoptosis is not known. METHODS: Human hepatic cells or primary hepatocytes from rats or TG2+/+ and TG2-/- mice were treated with ethanol. Mice were administered anti-Fas antibody or alcohol. Liver sections were prepared from patients with alcoholic steatohepatitis. Changes in TG2 levels, Sp1 cross-linking and its activities, expression of hepatocyte growth factor receptor, c-Met, and hepatic apoptosis were measured. RESULTS: Ethanol induced apoptosis in hepatic cells, enhanced activity and nuclear accumulation of TG2 as well as accumulation of cross-linked and inactivated Sp1, and reduced expression of the Sp1-responsive gene, c-Met. These effects were rescued by TG2 knockdown, restoration of functional Sp1, or addition of hepatocyte growth factor, whereas apoptosis was reproduced by Sp1 knockdown or TG2 overexpression. Compared with TG2+/+ mice, TG2-/- mice showed markedly reduced hepatocyte apoptosis and Sp1 cross-linking following ethanol or anti-Fas treatment. Treatment of TG2+/+ mice with the TG2 inhibitors putrescine or cystamine blocked anti-Fas-induced hepatic apoptosis and Sp1 silencing. Moreover, enhanced expression of cross-linked Sp1 and TG2 was evident in hepatocyte nuclei of patients with alcoholic steatohepatitis. CONCLUSIONS: TG2 induces hepatocyte apoptosis via Sp1 cross-linking and inactivation, with resultant inhibition of the expression of c-Met required for hepatic cell viability.


Asunto(s)
Etanol/farmacología , Hígado Graso Alcohólico/metabolismo , Proteínas de Unión al GTP/metabolismo , Silenciador del Gen , Factor de Transcripción Sp1/metabolismo , Transglutaminasas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Hígado Graso Alcohólico/patología , Cobayas , Hepatocitos/metabolismo , Humanos , Ratones , Ratones Endogámicos , Proteína Glutamina Gamma Glutamiltransferasa 2 , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Transcripción Sp1/genética , Activación Transcripcional , Transfección
7.
Science ; 369(6502): 456-460, 2020 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-32499399

RESUMEN

The Caribbean was one of the last regions of the Americas to be settled by humans, but where they came from and how and when they reached the islands remain unclear. We generated genome-wide data for 93 ancient Caribbean islanders dating between 3200 and 400 calibrated years before the present and found evidence of at least three separate dispersals into the region, including two early dispersals into the Western Caribbean, one of which seems connected to radiation events in North America. This was followed by a later expansion from South America. We also detected genetic differences between the early settlers and the newcomers from South America, with almost no evidence of admixture. Our results add to our understanding of the initial peopling of the Caribbean and the movements of Archaic Age peoples in the Americas.


Asunto(s)
Genética de Población , Migración Humana , Región del Caribe , Etnicidad/genética , Genómica , Humanos
8.
Mol Pharmacol ; 76(1): 58-67, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19389922

RESUMEN

Kisspeptins, the natural ligands of the G protein-coupled receptor KISS1R, comprise a family of related peptides derived from the proteolytic processing of a common precursor encoded by the KISS1 gene. Among those, Kisspeptin-10 (Kp-10) contains the basic residues to retain full functional activity and exhibits higher receptor affinity and biopotency than longer forms of the peptide. Although kisspeptins were first characterized by their ability to inhibit tumor metastasis, recent studies have revealed that the KISS1/KISS1R system plays an essential role in the neuroendocrine control of the reproductive axis. In this context, development and functional analysis of Kp-10 analogs may help in the search for new agonists and antagonists as valuable tools to manipulate the KISS1/KISS1R system and hence fertility. We report herein functional and structural analyses of a series of Ala-substituted rat kp-10 analogs, involving [Ca(2+)](i) responses in rat kiss1r-transfected Chinese hamster ovary cells, dynamic luteinizing hormone (LH) responses in vivo, and NMR structural studies. In vitro assays revealed that Ala substitutions in positions 6 or 10 of kp-10 resulted in a significant increase in EC(50) values (>6.46 x 10(-6) M versus 1.54 to 2.6 x 10(-8) M for rat and human Kp-10, respectively) and a substantial decrease in the proportion of responsive cells coupled to a marked increase in the time required to reach maximal response. In vivo assays showed that Ala(6) substitution diminished and Ala(10) substitution eliminated LH secretory responses, whereas coadministration of each analog failed to affect the LH-releasing ability of kp-10. Molecular modeling under NMR restraints revealed that kp-10 exhibits a helicoidal structure between the Asn(4) and Tyr(10) residues, with mixed alpha- and 3(10)-helix characteristics. Ala(6) substitution induced limited destabilization of the helix around the position of the substitution. Ala(10) substitution was found to totally disrupt the helical structure in the C-terminal region of the molecule. Taken together, our results indicate that positions 6 and 10 are critical for kp-10 action at kiss1r and suggest that modifications in these positions could lead to the generation of new kisspeptin agonists and/or antagonists with altered functional and perhaps binding properties. Furthermore, they emphasize the importance of using combined, multidisciplinary approaches, including in vivo studies, to reliably evaluate structure function properties of novel kisspeptin analogs.


Asunto(s)
Oligopéptidos/química , Oligopéptidos/farmacología , Secuencia de Aminoácidos , Animales , Células CHO , Cricetinae , Cricetulus , Kisspeptinas , Hormona Luteinizante/metabolismo , Espectroscopía de Resonancia Magnética , Masculino , Datos de Secuencia Molecular , Conformación Proteica , Ratas , Ratas Wistar , Relación Estructura-Actividad
9.
Peptides ; 30(1): 10-5, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18775460

RESUMEN

Kisspeptins, a family of peptide products derived from the KiSS-1 gene, activate their cognate receptor GPR54 in various target tissues to exert disparate functions, including inhibition of tumor metastasis and control of reproductive function. In contrast to the plethora of studies that have analyzed in recent years the regulatory functions of the KiSS-1/GPR54 system, only a limited number of reports have been primarily focused on delineating the intracellular signaling pathways involved. Nevertheless, there is solid evidence indicating that kisspeptin can activate a wide variety of signals via GPR54. These include typical G-protein (Galphaq/11)-coupled cascades, such as activation of phospholipase C (PLC), and subsequent accumulation of inositol-(1,4,5)-triphosphate (IP3), intracellular Ca(2+) mobilization, and activation of protein kinase C. However, kisspeptin also activates pathways related to mitogen activated protein kinases (MAPK), especially ERK1/2, and p38 and phosphatidylinositol-3-kinase (PI3K)/Akt. Additionally, the kisspeptin/GPR54 pair can also influence cell signaling by interacting with other receptors, such as chemokine receptor CXCR4, and GnRH receptor. Kisspeptin can also affect other signaling events, like expression of matrix metalloproteinase 9 (via NFkappaB), and that of calcineurin. The information gathered hitherto clearly indicates that activation of a specific set of interconnected signals is selectively triggered by kisspeptin via GPR54 in a cell type-dependent manner to precisely regulate functions as distinct as hormone release and cell migration. In this scenario, it will be important to decipher kisspeptin/GPR54 signaling mechanisms in reproductive and non-reproductive tissues by studying additional models, especially on natural kisspeptin targets expressing endogenous GPR54.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología , Proteínas Supresoras de Tumor/metabolismo , Secuencia de Aminoácidos , Animales , Calcio/metabolismo , Quimiocinas/metabolismo , Humanos , Kisspeptinas , Metaloproteinasa 9 de la Matriz/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Receptores de Kisspeptina-1 , Alineación de Secuencia , Fosfolipasas de Tipo C/metabolismo
10.
BMC Evol Biol ; 8: 213, 2008 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-18644108

RESUMEN

BACKGROUND: Before the arrival of Europeans to Cuba, the island was inhabited by two Native American groups, the Tainos and the Ciboneys. Most of the present archaeological, linguistic and ancient DNA evidence indicates a South American origin for these populations. In colonial times, Cuban Native American people were replaced by European settlers and slaves from Africa. It is still unknown however, to what extent their genetic pool intermingled with and was 'diluted' by the arrival of newcomers. In order to investigate the demographic processes that gave rise to the current Cuban population, we analyzed the hypervariable region I (HVS-I) and five single nucleotide polymorphisms (SNPs) in the mitochondrial DNA (mtDNA) coding region in 245 individuals, and 40 Y-chromosome SNPs in 132 male individuals. RESULTS: The Native American contribution to present-day Cubans accounted for 33% of the maternal lineages, whereas Africa and Eurasia contributed 45% and 22% of the lineages, respectively. This Native American substrate in Cuba cannot be traced back to a single origin within the American continent, as previously suggested by ancient DNA analyses. Strikingly, no Native American lineages were found for the Y-chromosome, for which the Eurasian and African contributions were around 80% and 20%, respectively. CONCLUSION: While the ancestral Native American substrate is still appreciable in the maternal lineages, the extensive process of population admixture in Cuba has left no trace of the paternal Native American lineages, mirroring the strong sexual bias in the admixture processes taking place during colonial times.


Asunto(s)
Cromosomas Humanos Y/genética , ADN Mitocondrial/genética , Variación Genética , Filogenia , Cuba/etnología , Femenino , Genética de Población , Haplotipos , Humanos , Masculino , Polimorfismo de Nucleótido Simple/genética , Grupos Raciales/genética
11.
J Clin Endocrinol Metab ; 93(6): 2269-76, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18349058

RESUMEN

CONTEXT: Rab proteins regulate the sequential steps of intracellular membrane transport. Alterations of these GTPases and their associated proteins are emerging as the underlying cause for several human diseases involving dysregulated secretory activities. OBJECTIVE: Herein we investigated the role of Rab18, which negatively regulates hormone secretion by interacting with secretory granules, in relation to the altered functioning of tumoral pituitary somatotropes causing acromegaly. PATIENTS: A total of 18 patients diagnosed with pituitary tumors causing acromegaly (nine patients) or nonfunctioning adenomas (nine patients) underwent endoscopic transsphenoidal surgery. Adenomas were subsequently processed to evaluate Rab18 production in relation to GH secretion. RESULTS: We found that somatotropinoma cells are characterized by a high secretory activity concomitantly with a remarkably reduced Rab18 expression (15%) and protein content levels (30%), as compared with cells from nonfunctioning pituitary adenomas derived from patients with normal or reduced GH plasma levels (100%). Furthermore, immunoelectron microscopy revealed that Rab18 association with the surface of GH-containing secretory granules was significantly lower in somatotropes from acromegalies than nonfunctioning pituitary adenomas. Finally, we provide evidence that modulation of Rab18 gene expression can revert substantially the hypersecretory activity of cells because Rab18 overexpression reduced by 40% the capacity of cells from acromegalies to respond to GHRH stimulation. CONCLUSION: These results suggest that molecular alterations affecting individual components of the secretory granule traffic machinery can contribute to maintain a high level of GH in plasma. Accordingly, Rab18 constitutes a valuable target as a diagnostic, prognostic, and/or therapeutic tool for human acromegaly.


Asunto(s)
Acromegalia/genética , Adenoma/genética , Adenoma Hipofisario Secretor de Hormona del Crecimiento/genética , Hormona de Crecimiento Humana/metabolismo , Proteínas de Unión al GTP rab/genética , Acromegalia/etiología , Adenoma/metabolismo , Membrana Celular/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Adenoma Hipofisario Secretor de Hormona del Crecimiento/metabolismo , Humanos , ARN Mensajero/metabolismo , Vesículas Secretoras/metabolismo , Somatotrofos/metabolismo , Distribución Tisular , Transfección , Células Tumorales Cultivadas , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rab/fisiología
12.
Peptides ; 29(5): 711-20, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18289730

RESUMEN

The UII and urotensin II-related peptide (URP) genes belong to the same superfamily as the somatostatin gene. It has been previously shown that somatostatin activates the UII-receptor (UTR). In contrast, the possible interaction between UII and URP and somatostatin receptors has remained scarcely analyzed. Herein, we have investigated the effects of UII and URP on cell proliferation and free cytosolic Ca2+ concentration ([Ca2+]i) in CHO-K1 cells stably expressing the porcine somatostatin receptor subtypes sst2 and sst5. Results show that both UII and URP induce stimulation of cell proliferation mediated by sst2 receptors and UII provokes inhibition of cell proliferation mediated by sst5 receptors. UII and URP also provoked an increase of [Ca2+]i in both sst2- and sst5-transfected cells. Together, our present data demonstrate that UII and URP directly activate sst2 and sst5 and thus mimic the effect of somatostatin on its cognate receptors.


Asunto(s)
Proliferación Celular , Hormonas Peptídicas/metabolismo , Receptores de Somatostatina/metabolismo , Urotensinas/metabolismo , Animales , Células CHO , Calcio/metabolismo , Cricetinae , Cricetulus , Humanos , Quinolinas/metabolismo , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/genética , Somatostatina/agonistas , Somatostatina/metabolismo , Porcinos , Urea/análogos & derivados , Urea/metabolismo
13.
Clin Transl Gastroenterol ; 9(10): 196, 2018 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-30297816

RESUMEN

OBJECTIVES: The association between the presence and alterations of the components of the ghrelin system and the development and progression of neuroendocrine tumors (NETs) is still controversial and remains unclear. METHODS: Here, we systematically evaluated the expression levels (by quantitative-PCR) of key ghrelin system components of in gastroenteropancreatic (GEP)-NETs, as compared to non-tumor adjacent (NTA; n = 42) and normal tissues (NT; n = 14). Then, we analyzed their putative associations with clinical-histological characteristics. RESULTS: The results indicate that ghrelin and its receptor GHSR1a are present in a high proportion of normal tissues, while the enzyme ghrelin-O-acyltransferase (GOAT) and the splicing variants In1-ghrelin and GHSR1b were present in a lower proportion of normal tissues. In contrast, all ghrelin system components were present in a high proportion of tumor and NTA tissues. GOAT was significantly overexpressed (by quantitative-PCR (qPCR)) in tumor samples compared to NTA, while a trend was found for ghrelin, In1-ghrelin and GHSR1a. In addition, expression of these components displayed significant correlations with key clinical parameters. The marked overexpression of GOAT in tumor samples compared to NTA regions was confirmed by IHC, revealing that this enzyme is particularly overexpressed in gastrointestinal NETs, where it is directly correlated with tumor diameter. CONCLUSIONS: These results provide novel information on the presence and potential pathophysiological implications of the ghrelin system components in GEP-NETs, wherein GOAT might represent a novel diagnostic biomarker.


Asunto(s)
Aciltransferasas/metabolismo , Biomarcadores de Tumor/metabolismo , Ghrelina/metabolismo , Neoplasias Intestinales/metabolismo , Tumores Neuroendocrinos/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptores de Ghrelina/metabolismo , Neoplasias Gástricas/metabolismo , Aciltransferasas/genética , Adulto , Anciano , Biomarcadores de Tumor/genética , Estudios de Casos y Controles , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Femenino , Ghrelina/genética , Humanos , Neoplasias Intestinales/genética , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Tumores Neuroendocrinos/genética , Neoplasias Pancreáticas/genética , Valor Predictivo de las Pruebas , Receptores de Ghrelina/genética , Neoplasias Gástricas/genética
14.
Endocrinology ; 148(1): 411-21, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17053026

RESUMEN

Somatostatin (SRIF) exerts its multiple actions, including inhibition of GH secretion and of tumoral growth, through a family of five receptor subtypes (sst1-sst5). We recently reported that an sst2-selective agonist markedly decreases GH release from pig somatotropes, suggesting important roles for this scarcely explored receptor, psst2. Here, functional expression of psst2 in Chinese hamster ovary-K1 and human embryonic kidney-293-AD cell lines was employed to determine its pharmacological features and functional ability to reduce cAMP, and to examine its homodimerization and internalization dynamics in real time in single living cells. Results show that psst2 is a high-affinity receptor (dissociation constant = 0.27 nM) displaying a typical sst2 profile (nM affinity for SRIF-14> or =SRIF-28>cortistatin>MK678>octreotide) and high selectivity (EC(50) = 1.1 nM) for the sst2 agonist l-779,976, but millimolar or undetectable affinity to other sst-specific agonists (sst3>sst1>sst5>>>sst4). Accordingly, SRIF dose-dependently inhibited forskolin-stimulated cAMP with high potency (EC(50) = 6.55 pm) and modest efficacy (maximum 29.1%) via psst2. Cotransfection of human embryonic kidney-293 and Chinese hamster ovary-K1 cells with two receptor constructs modified with distinct fluorescent tags (psst2-YFP/psst2-CFP) enabled fluorescence resonance energy transfer measurement of physical interaction between psst2 receptors and also receptor internalization in single living cells. This revealed that under basal conditions, psst2 forms constitutive homodimers/homomultimers, which dissociate immediately (11 sec) upon SRIF binding. Interestingly, contrary to human sst2, psst2 rapidly reassociates (110.5 sec) during a subsequent process that temporally overlaps with receptor internalization (half-maximal = 95.1 sec). Therefore, psst2 is a potent inhibitory receptor displaying a unique set of interrelated dynamic features of agonist-dependent dimerization, dissociation, internalization, and reassociation, a cascade of events that might be critical for receptor function.


Asunto(s)
Hipófisis/citología , Hipófisis/fisiología , Receptores de Somatostatina/química , Receptores de Somatostatina/metabolismo , Animales , Antineoplásicos Hormonales/farmacología , Células CHO , Calcio/metabolismo , Clonación Molecular , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Dimerización , Endocitosis/fisiología , Femenino , Transferencia Resonante de Energía de Fluorescencia , Antagonistas de Hormonas/farmacología , Neuropéptidos/farmacología , Octreótido/farmacología , Péptidos Cíclicos/farmacología , Ensayo de Unión Radioligante , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/genética , Somatostatina/análogos & derivados , Somatostatina/metabolismo , Somatostatina/farmacología , Sus scrofa
15.
Endocrinology ; 148(1): 401-10, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17038552

RESUMEN

Adiponectin is a member of the family of adipose tissue-related hormones known as adipokines, which exerts antidiabetic, antiatherogenic, antiinflammatory, and antiangiogenic properties. Adiponectin actions are primarily mediated through binding to two receptors expressed in several tissues, AdipoR1 and AdipoR2. Likewise, adiponectin expression has been detected in adipocytes as well as in a variety of extra-adipose tissues, including the chicken pituitary. Interestingly, adiponectin secretion and adiponectin receptor expression in adipocytes have been shown to be regulated by pituitary hormones. These observations led us to investigate whether adiponectin, like the adipokine leptin, regulates pituitary hormone production. Specifically, we focused our analysis on somatotrophs and gonadotrophs because of the relationship between the control of energy metabolism, growth and reproduction. To this end, the effects of adiponectin on both GH and LH secretion as well as its interaction with major stimulatory regulators of somatotrophs (ghrelin and GHRH) and gonadotrophs (GnRH) and with their corresponding receptors (GHS-R, GHRH-R, and GnRH-R), were evaluated in rat pituitary cell cultures. Results show that adiponectin inhibits GH and LH release as well as both ghrelin-induced GH release and GnRH-stimulated LH secretion in short-term (4 h) treated cell cultures, wherein the adipokine also increases GHRH-R and GHS-R mRNA content while decreasing that of GnRH-R. Additionally, we demonstrate that the pituitary expresses both adiponectin and adiponectin receptors under the regulation of the adipokine. In sum, our data indicate that adiponectin, either locally produced or from other sources, may play a neuroendocrine role in the control of both somatotrophs and gonadotrophs.


Asunto(s)
Adiponectina/genética , Adiponectina/farmacología , Adenohipófisis/citología , Adenohipófisis/fisiología , Adiponectina/metabolismo , Animales , Células Cultivadas , Expresión Génica/efectos de los fármacos , Hormona del Crecimiento/metabolismo , Hormona Luteinizante/metabolismo , Masculino , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Adiponectina , Receptores de Superficie Celular/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Ghrelina , Receptores de Neuropéptido/genética , Receptores de Hormona Reguladora de Hormona Hipofisaria/genética
16.
J Clin Endocrinol Metab ; 91(6): 2225-31, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16551736

RESUMEN

CONTEXT: In Cushing's disease, ACTH hypersecretion by pituitary corticotrope adenoma cells and resulting hypercortisolism is accompanied by a severely blunted GH secretory response. Interestingly, in Cushing's disease, ghrelin markedly increases plasma ACTH, whereas its stimulatory action on GH secretion is reduced. Although the reported expression of ghrelin receptors (GHS-R) in corticotrope tumors offers a potential mechanism for ghrelin-induced ACTH hypersecretion, studies on the direct effects of synthetic GH secretagogues on corticotropinoma cells offered contradictory results. OBJECTIVE AND DESIGN: To evaluate the direct action of ghrelin on corticotropinoma cells from two patients with Cushing's disease, we measured its effect on free cytosolic calcium concentration ([Ca(2+)](i)). Additionally, expression of GHS-R and its ligand ghrelin was examined in these cells and in five additional corticotropinomas. RESULTS: Ghrelin (10(-6) m) induced a marked [Ca(2+)](i) increase in 89.5% (case 1; n = 19 cells) and 85% (case 2; n = 13 cells) of corticotropinoma cells. Moreover, RT-PCR showed that expression of GHS-R isoforms is accompanied by that of ghrelin in all seven corticotrope adenomas examined. Importantly, double immunogold electron microscopy revealed that ghrelin is costored within ACTH secretory vesicles in densely granulated adenomatous corticotropes. CONCLUSIONS: These results constitute the first demonstration that ghrelin acts directly on corticotrope tumor cells derived from patients with Cushing's disease. The presence of ghrelin and GHS-R suggests that pituitary ghrelin may play an autocrine/paracrine role in regulating ACTH release in Cushing's disease. Our findings provide a plausible cellular basis for the exaggerated ACTH response to ghrelin in Cushing's disease and suggest novel research strategies to develop medical treatments for this disease.


Asunto(s)
Adenoma/metabolismo , Hormona Adrenocorticotrópica/metabolismo , Hormonas Peptídicas/fisiología , Neoplasias Hipofisarias/metabolismo , Adulto , Calcio/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Ghrelina , Humanos , Inmunohistoquímica , Proopiomelanocortina/genética , ARN Mensajero/análisis , Receptores Acoplados a Proteínas G/genética , Receptores de Ghrelina
17.
Mol Cell Endocrinol ; 438: 100-106, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27477782

RESUMEN

Kisspeptin (Kiss1) is an amidated neurohormone that belongs to the RF-amide peptide family, which has a key role in the control of reproduction. Specifically, kisspeptin regulates reproductive events, including puberty and ovulation, primarily by activating the surface receptor Kiss1r (aka GPR54), at hypothalamic gonadotropin-releasing hormone (GnRH) neurons. More recently, it has been found that kisspeptin peptide is present in the hypophyseal portal circulation and that the Kiss1/Kiss1r system is expressed in pituitary cells, which suggest that kisspeptin could exert an endocrine, paracrine or even autocrine role at the pituitary gland level. Indeed, mounting evidence is pointing towards a direct role of kisspeptin in the control of not only gonadotropins but also other pituitary secretions such as growth hormone or prolactin. In this review, we summarize the most recent advances in the study of the role that the Kiss/Kiss1r system plays in the control of pituitary gland function, paying special attention to the direct role of this neuropeptide on pituitary cells and its interactions with other relevant regulators.


Asunto(s)
Kisspeptinas/metabolismo , Hipófisis/citología , Hipófisis/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Humanos , Modelos Biológicos , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología
18.
Mol Endocrinol ; 18(7): 1808-17, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15184526

RESUMEN

The characteristic pulsatile secretion of GnRH from hypothalamic neurons is dependent on an autocrine interaction between GnRH and its receptors expressed in GnRH-producing neurons. The ontogeny and function of this autoregulatory process were investigated in studies on the properties of GnRH neurons derived from the olfactory placode of the fetal rat. An analysis of immunocytochemically identified, laser-captured fetal rat hypothalamic GnRH neurons, and olfactory placode-derived GnRH neurons identified by differential interference contrast microscopy, demonstrated coexpression of mRNAs encoding GnRH and its type I receptor. Both placode-derived and immortalized GnRH neurons (GT1-7 cells) exhibited spontaneous electrical activity that was stimulated by GnRH agonist treatment. This evoked response, as well as basal neuronal firing, was abolished by treatment with a GnRH antagonist. GnRH stimulation elicited biphasic intracellular calcium ([Ca2+]i) responses, and both basal and GnRH-stimulated [Ca2+]i levels were reduced by antagonist treatment. Perifused cultures released GnRH in a pulsatile manner that was highly dependent on extracellular Ca2+. The amplitude of GnRH pulses was increased by GnRH agonist stimulation and was diminished during GnRH antagonist treatment. These findings demonstrate that expression of GnRH receptor, GnRH-dependent activation of Ca2+ signaling, and autocrine regulation of GnRH release are characteristics of early fetal GnRH neurons and could provide a mechanism for gene expression and regulated GnRH secretion during embryonic migration.


Asunto(s)
Membrana Celular/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/embriología , Neuronas/metabolismo , Neurosecreción/fisiología , Vías Olfatorias/embriología , Receptores LHRH/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Señalización del Calcio , Células Cultivadas , Femenino , Regulación del Desarrollo de la Expresión Génica , Hormona Liberadora de Gonadotropina/agonistas , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Hormona Liberadora de Gonadotropina/genética , Hipotálamo/citología , Hipotálamo/metabolismo , Neuronas/fisiología , Vías Olfatorias/citología , Vías Olfatorias/metabolismo , Embarazo , Ratas , Ratas Sprague-Dawley , Receptores LHRH/genética
19.
Mol Endocrinol ; 17(12): 1792-1804, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14680004

RESUMEN

Immortalized GnRH neurons (GT1-7) express receptors for estrogen [estrogen receptor-alpha and-13(ERa and ERI3)] and progesterone (progesterone receptor A) and exhibit positive immunostaining for both intracellular and plasma membrane ERs. Exposure of GT1-7 cells to picomolar estradiol concentrations for 5-60 min caused rapid, sustained,and dose-dependent inhibition of cAMP production. In contrast, treatment with nanomolar estradiol concentrations for 60 min increased cAMP production. The inhibitory and stimulatory actions of estradiol on cAMP formation were abolished by the ER antagonist, ICI 182,780. The estradiol-induced inhibition of cAMP production was prevented by treatment with pertussis toxin, consistent with coupling of the plasma membrane ER to an inhibitory G protein. Coimmunoprecipitation studies demonstrated an estradiol-regulated stimulatory interaction between ERa and G,3 that was prevented by the ER antagonist, ICI 182,780. Exposure of perifused GT1-7 cells and hypothalamic neurons to picomolar estradiol levels increased the GnRH peak interval, shortened peak duration, and increased peak amplitude. These findings indicate that occupancy of the plasma membrane-associated ERs expressed in GT1-7 neurons by physio-logical estradiol levels causes activation of a G, protein and modulates cAMP signaling and neuropeptide secretion.


Asunto(s)
AMP Cíclico/fisiología , Hipotálamo/embriología , Neuronas/fisiología , Receptores de Estrógenos/genética , Receptores de Progesterona/genética , Transducción de Señal/fisiología , Animales , Sitios de Unión , Células Cultivadas , Cartilla de ADN , Estradiol/farmacología , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Estrógenos/metabolismo , Femenino , Feto , Regulación del Desarrollo de la Expresión Génica , Hormona Liberadora de Gonadotropina/efectos de los fármacos , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/fisiología , Inmunohistoquímica , Neuronas/citología , Neuronas/efectos de los fármacos , Nervio Óptico/fisiología , Embarazo , Progesterona/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Estrógenos/metabolismo , Transducción de Señal/efectos de los fármacos , Útero/fisiología
20.
Mol Endocrinol ; 17(9): 1792-804, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12819297

RESUMEN

Immortalized GnRH neurons (GT1-7) express receptors for estrogen [estrogen receptor-alpha and -beta(ERalpha and ERbeta)] and progesterone (progesterone receptor A) and exhibit positive immunostaining for both intracellular and plasma membrane ERs. Exposure of GT1-7 cells to picomolar estradiol concentrations for 5-60 min caused rapid, sustained, and dose-dependent inhibition of cAMP production. In contrast, treatment with nanomolar estradiol concentrations for 60 min increased cAMP production. The inhibitory and stimulatory actions of estradiol on cAMP formation were abolished by the ER antagonist, ICI 182,780. The estradiol-induced inhibition of cAMP production was prevented by treatment with pertussis toxin, consistent with coupling of the plasma membrane ER to an inhibitory G protein. Coimmunoprecipitation studies demonstrated an estradiol-regulated stimulatory interaction between ERalpha and Galphai3 that was prevented by the ER antagonist, ICI 182,780. Exposure of perifused GT1-7 cells and hypothalamic neurons to picomolar estradiol levels increased the GnRH peak interval, shortened peak duration, and increased peak amplitude. These findings indicate that occupancy of the plasma membrane-associated ERs expressed in GT1-7 neurons by physiological estradiol levels causes activation of a Gi protein and modulates cAMP signaling and neuropeptide secretion.


Asunto(s)
Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Neuronas/metabolismo , Neurosecreción/fisiología , Receptores de Estrógenos/metabolismo , Animales , Retículo Endoplásmico/metabolismo , Femenino , Feto/metabolismo , Hipotálamo/metabolismo , Inmunohistoquímica , Embarazo , Ratas , Receptores de Progesterona/metabolismo , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA