Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Mol Cell Endocrinol ; 529: 111269, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33819522

RESUMEN

Prolactin (PRL), mainly synthesized and secreted by the lactotrophs and somatolactotrophs of the anterior pituitary, is a pleiotropic hormone that regulates lactation. In the last decade, nesfatin-1 (NESF) and NESF-like peptide (NLP), encoded in nucleobindin 1 and 2 (NUCB1 and NUCB2), respectively, were characterized as metabolic factors with a potential role in the control of pituitary hormones. We hypothesized that NUCBs and their encoded peptides (NESF and NLP) suppress PRL transcription in the pituitary. The main objective of this research was to determine whether exogenous NESF and NLP, and/or endogenous NUCB1 and NUCB2 regulate the expression of prl and preb mRNAs. Using immortalized rat somatolactotrophs (GH3 cells), dose-response studies were performed to test whether NESF and NLP affect prl and preb. Moreover, the ability of these peptides to modulate the effects of the PRL stimulator thyrotropin releasing hormone (TRH) was studied. Besides, the effects of siRNA-mediated knockdown of endogenous NUCBs on prl and preb mRNAs were determined. NESF and NLP reduced the transcription of prl and preb in GH3 cells. Both NESF and NLP also prevented the stimulatory effects of TRH prl and preb expression. The knockdown of endogenous NUCB1 attenuates both basal prl and TRH-induced expression of prl and preb, while the silencing of NUCBs did not affect the actions of exogenous NESF or NLP. Overall, this work reveals that NUCBs and encoded-peptides are novel regulators of PRL. Future research should test whether the effects observed here in GH3 cells are preserved both in vivo and at the post-transcriptional level.


Asunto(s)
Proteínas de Unión al ADN/genética , Factores de Intercambio de Guanina Nucleótido/genética , Lactotrofos/efectos de los fármacos , Nucleobindinas/farmacología , Péptidos/farmacología , Prolactina/genética , Somatotrofos/efectos de los fármacos , Factores de Transcripción/genética , Animales , Línea Celular Transformada , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido/metabolismo , Lactotrofos/citología , Lactotrofos/metabolismo , Nucleobindinas/antagonistas & inhibidores , Nucleobindinas/genética , Nucleobindinas/metabolismo , Prolactina/antagonistas & inhibidores , Prolactina/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Transducción de Señal , Somatotrofos/citología , Somatotrofos/metabolismo , Hormona Liberadora de Tirotropina/genética , Hormona Liberadora de Tirotropina/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo
2.
Growth Horm IGF Res ; 51: 65-74, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32146343

RESUMEN

OBJECTIVE: Nicotine, a toxic component of smoking, adversely affects animal growth and reproduction by decreasing secretion of anterior pituitary hormones. However, it has not been clarified whether nicotine inhibits the supply of endocrine cells in the pituitary gland. The present study investigated short- and long-term effects of persistent nicotine exposure on the pituitary glands of young animals. DESIGN: Three-week-old male Wistar rats were exposed to nicotine (1 mg/kg body weight/day) for 7 days, and gene expression, cell numbers, and DNA methylation status were analyzed on the following day and 4 weeks after final treatments. RESULTS: The expression level of the stem cell marker Sox2 was not changed by nicotine exposure throughout the experiment. On the other hand, nicotine inhibited expression of a progenitor cell marker, Prrx1, and growth hormone (Gh). Immunohistochemical analysis showed that the SOX2-positive cells positive for PRRX1 in nicotine-treated groups decreased to 61% (4-week-old) and 70% (8-week-old) of the saline-treated controls. In addition, the proportion of GH-positive cells in nicotine-treated group was 14% lower than that of saline-treated controls. Furthermore, first intron hypermethylation of Prrx1 was detected by a bisulfite sequence of genomic DNA from the anterior lobe of the rat pituitary gland. CONCLUSIONS: We show that persistent nicotine exposure in young animals inhibits expression of Prrx1 in pituitary stem/progenitor cells through epigenetic regulation, leading to a delayed supply of GH-producing cells.


Asunto(s)
Epigénesis Genética/efectos de los fármacos , Hormona del Crecimiento/efectos de los fármacos , Proteínas de Homeodominio/efectos de los fármacos , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Somatotrofos/efectos de los fármacos , Células Madre/efectos de los fármacos , Animales , Recuento de Células , Metilación de ADN/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hormona del Crecimiento/metabolismo , Proteínas de Homeodominio/genética , Intrones , Masculino , Hipófisis/citología , Hipófisis/efectos de los fármacos , Hipófisis/metabolismo , Ratas , Ratas Wistar , Somatotrofos/citología , Somatotrofos/metabolismo , Células Madre/citología , Células Madre/metabolismo
3.
Int J Mol Sci ; 21(4)2020 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-32093314

RESUMEN

UCL-2077 (triphenylmethylaminomethyl)pyridine) was previously reported to suppress slow afterhyperpolarization in neurons. However, the information with respect to the effects of UCL-2077 on ionic currents is quite scarce. The addition of UCL-2077 decreased the amplitude of erg-mediated K+ current (IK(erg)) together with an increased deactivation rate of the current in pituitary GH3 cells. The IC50 and KD values of UCL-2077-induced inhibition of IK(erg) were 4.7 and 5.1 µM, respectively. UCL-2077 (10 µM) distinctly shifted the midpoint in the activation curve of IK(erg) to less hyperpolarizing potentials by 17 mV. Its presence decreased the degree of voltage hysteresis for IK(erg) elicitation by long-lasting triangular ramp pulse. It also diminished the probability of the opening of intermediate-conductance Ca2+-activated K+ channels. In cell-attached current recordings, UCL-2077 raised the frequency of action currents. When KCNH2 mRNA was knocked down, a UCL-2077-mediated increase in AC firing was attenuated. Collectively, the actions elaborated herein conceivably contribute to the perturbating effects of this compound on electrical behaviors of excitable cells.


Asunto(s)
Bencilaminas/farmacología , Canal de Potasio ERG1/metabolismo , Potasio/metabolismo , Piridinas/farmacología , Somatotrofos/metabolismo , Línea Celular , Canal de Potasio ERG1/genética , Técnicas de Silenciamiento del Gen , Humanos , Transporte Iónico/efectos de los fármacos , Transporte Iónico/genética , Somatotrofos/citología
4.
Growth Horm IGF Res ; 50: 9-22, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31809882

RESUMEN

OBJECTIVE: To revisit a finding, first described in 1978, which documented existence of a pituitary growth factor that escaped detection by immunoassay, but which was active in the established rat tibia GH bioassay. METHODS: We present a narrative review of the evolution of growth hormone complexity, and its bio-detectability, from a historical perspective. RESULTS: In humans under the age of 60, physical training (i.e. aerobic endurance and resistance training) are stressors which preferentially stimulate release of bioactive GH (bGH) into the blood. Neuroanatomical studies indicate a) that nerve fibers directly innervate the human anterior pituitary and b) that hind limb muscle afferents, in both humans and rats, also modulate plasma bGH. In the pituitary gland itself, molecular variants of GH, somatotroph heterogeneity and cell plasticity all appear to play a role in regulation of this growth factor. CONCLUSION: This review considers more recent findings on this often forgotten/neglected subject. Comparison testing of a) human plasma samples, b) sub-populations of separated rat pituitary somatotrophs or c) purified human pituitary peptides by GH bioassay vs immunoassay consistently yield conflicting results.


Asunto(s)
Ejercicio Físico/fisiología , Hormona de Crecimiento Humana/sangre , Somatotrofos/metabolismo , Vías Aferentes , Animales , Bioensayo/métodos , Plasticidad de la Célula , Entrenamiento Aeróbico , Hormona del Crecimiento/sangre , Hormona del Crecimiento/metabolismo , Hormona de Crecimiento Humana/metabolismo , Humanos , Hipotálamo/metabolismo , Inmunoensayo/métodos , Músculo Esquelético/inervación , Condicionamiento Físico Animal/fisiología , Adenohipófisis/inervación , Ratas , Entrenamiento de Fuerza , Somatotrofos/citología
5.
Curr Protein Pept Sci ; 21(1): 52-65, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31702489

RESUMEN

Hormones are known to influence various body systems that include skeletal, cardiac, digestive, excretory, and immune systems. Emerging investigations suggest the key role played by secretions of endocrine glands in immune cell differentiation, proliferation, activation, and memory attributes of the immune system. The link between steroid hormones such as glucocorticoids and inflammation is widely known. However, the role of peptide hormones and amino acid derivatives such as growth and thyroid hormones, prolactin, dopamine, and thymopoietin in regulating the functioning of the immune system remains unclear. Here, we reviewed the findings pertinent to the functional role of hormone-immune interactions in health and disease and proposed perspective directions for translational research in the field.


Asunto(s)
Enfermedades del Sistema Endocrino/metabolismo , Sistema Endocrino/metabolismo , Hormona del Crecimiento/metabolismo , Enfermedades del Sistema Inmune/metabolismo , Sistema Inmunológico/metabolismo , Prolactina/metabolismo , Timocitos/metabolismo , Animales , Comunicación Celular , Citocinas/genética , Citocinas/inmunología , Citocinas/metabolismo , Dopamina/genética , Dopamina/inmunología , Dopamina/metabolismo , Sistema Endocrino/citología , Sistema Endocrino/inmunología , Enfermedades del Sistema Endocrino/genética , Enfermedades del Sistema Endocrino/inmunología , Enfermedades del Sistema Endocrino/patología , Glucocorticoides/genética , Glucocorticoides/inmunología , Glucocorticoides/metabolismo , Hormona del Crecimiento/genética , Hormona del Crecimiento/inmunología , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Enfermedades del Sistema Inmune/genética , Enfermedades del Sistema Inmune/inmunología , Enfermedades del Sistema Inmune/patología , Lactotrofos/citología , Lactotrofos/inmunología , Lactotrofos/metabolismo , Prolactina/genética , Prolactina/inmunología , Receptores Dopaminérgicos/genética , Receptores Dopaminérgicos/inmunología , Receptores Dopaminérgicos/metabolismo , Somatotrofos/citología , Somatotrofos/inmunología , Somatotrofos/metabolismo , Timocitos/citología , Timocitos/inmunología , Hormonas Tiroideas/genética , Hormonas Tiroideas/inmunología , Hormonas Tiroideas/metabolismo
6.
Int J Mol Sci ; 20(23)2019 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-31801305

RESUMEN

Electrical activity in neurons and other excitable cells is a result of complex interactions between the system of ion channels, involving both global coupling (e.g., via voltage or bulk cytosolic Ca2+ concentration) of the channels, and local coupling in ion channel complexes (e.g., via local Ca2+ concentration surrounding Ca2+ channels (CaVs), the so-called Ca2+ nanodomains). We recently devised a model of large-conductance BKCa potassium currents, and hence BKCa-CaV complexes controlled locally by CaVs via Ca2+ nanodomains. We showed how different CaV types and BKCa-CaV stoichiometries affect whole-cell electrical behavior. Ca2+ nanodomains are also important for triggering exocytosis of hormone-containing granules, and in this regard, we implemented a strategy to characterize the local interactions between granules and CaVs. In this study, we coupled electrical and exocytosis models respecting the local effects via Ca2+ nanodomains. By simulating scenarios with BKCa-CaV complexes with different stoichiometries in pituitary cells, we achieved two main electrophysiological responses (continuous spiking or bursting) and investigated their effects on the downstream exocytosis process. By varying the number and distance of CaVs coupled with the granules, we found that bursting promotes exocytosis with faster rates than spiking. However, by normalizing to Ca2+ influx, we found that bursting is only slightly more efficient than spiking when CaVs are far away from granules, whereas no difference in efficiency between bursting and spiking is observed with close granule-CaV coupling.


Asunto(s)
Potenciales de Acción/fisiología , Canales de Calcio/metabolismo , Calcio/metabolismo , Exocitosis/fisiología , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Somatotrofos/metabolismo , Animales , Simulación por Computador , Gránulos Citoplasmáticos/química , Gránulos Citoplasmáticos/metabolismo , Humanos , Activación del Canal Iónico/fisiología , Cinética , Modelos Biológicos , Hipófisis/citología , Hipófisis/metabolismo , Somatotrofos/citología
7.
J Neuroendocrinol ; 31(8): e12764, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31251840

RESUMEN

The roles of epidermal growth factor (EGF) in the regulation of prolactin (PRL) gene expression in the normal pituitary gland remain poorly understood. In the present study, the effects of EGF and an inhibitor of the EGF receptor, erlotinib, on PRL gene expression were examined both in the pituitary tumour cell line GH3 and in a primary culture of the mouse pituitary gland under similar experimental conditions. The results showed that EGF stimulated PRL expression in GH3 cells, but not in normal cells. Erlotinib was found to counteract EGF in GH3 cells inhibiting the PRL expression enhanced by EGF. By contrast, erlotinib induced an elevation in the PRL mRNA levels in the primary culture of the adult pituitary gland and the initiation of PRL production in the culture of the foetal pituitary gland in which PRL production had not yet occurred. Western blot analyses showed that EGF induced and erlotinib inhibited the activation of extracellular regulated protein kinase equally in GH3 and normal cells. These results suggest that the consequences of EGF receptor activation in normal PRL cells contradict those in adenomatous PRL cells.


Asunto(s)
Receptores ErbB/fisiología , Prolactina/genética , Animales , Células Cultivadas , Embrión de Mamíferos , Factor de Crecimiento Epidérmico/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos ICR , Hipófisis/citología , Hipófisis/efectos de los fármacos , Hipófisis/metabolismo , Embarazo , Cultivo Primario de Células , Prolactina/metabolismo , Somatotrofos/citología
8.
Tissue Cell ; 58: 93-98, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31133252

RESUMEN

This study aimed to propose a simple and practical method for culturing primary rat somatotropic cells in vitro free of pericytes contamination. Rat adenohypophyses were randomly divided into two groups. An improved method was used in group A (digesting adenohypophysis with 0.25% trypsin-EDTA, followed by removing pericytes by double filtration and using serum-free medium for culturing somatotropic cells). The traditional method was used in group B (digesting adenohypophysis with 0.35% collagenase, using serum medium for culturing somatotropic cells, and removing pericytes by changing the culture dish). The numbers and viability of somatotropic cells were higher in group A than in group B after 6 days. GH secretion of somatotropic cells was also higher in group A than in group B. Besides, the pericytes grew rapidly only in group B after 3 days. α-SMA, type I collagen, and type III collagen had weaker expression in group A. Also, the viability of pericytes decreased in group A. The improved method could solve the problem of pericytes contamination, and the culture of primary rat somatotropic cells in vitro was successful. This method can be used for other primary cultures with pericytes contamination.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Separación Celular , Somatotrofos/citología , Animales , Supervivencia Celular , Medio de Cultivo Libre de Suero/química , Medio de Cultivo Libre de Suero/farmacología , Masculino , Ratas , Ratas Sprague-Dawley , Somatotrofos/metabolismo
9.
Mol Cell Endocrinol ; 482: 37-44, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30543878

RESUMEN

Growth Hormone (GH) deficiency is frequent in HIV-infected patients treated with antiretroviral therapy. We treated GH3 cells with antiretrovirals (nevirapine, ritonavir or abacavir sulfate; 100 pM-1 mM range), after transfection with human growth hormone releasing hormone (GHRH) receptor cDNA. Cells viability, intracellular cAMP, phosphorylation of CREB and calcium increase, GH production and secretion were evaluated both in basal condition and after GHRH, using MTT, bioluminescence resonance energy transfer, western blotting and ELISA. Antiretroviral treatment did not affect GHRH 50% effective dose (EC50) calculated for 30-min intracellular cAMP increase (Mann-Whitney's U test; p ≥ 0.05; n = 4) nor 15-min CREB phosphorylation. The kinetics of GHRH-mediated, rapid intracellular calcium increase was perturbed by pre-incubation with drugs, while GHRH failed to induce the ion increase in ritonavir pre-treated cells (ANOVA; p < 0.05; n = 3). Antiretrovirals did not impact 24-h intracellular and extracellular GH levels (ANOVA; p ≥ 0.05; n = 3). We demonstrated the association between antiretrovirals and intracellular calcium increase, without consequences on somatotrope cells viability and GH synthesis. Overall, these results suggest that antiretrovirals may not directly impact on GH axis in HIV-infected patients.


Asunto(s)
Antirretrovirales/farmacología , Calcio/metabolismo , Hormona Liberadora de Hormona del Crecimiento/genética , Hormona de Crecimiento Humana/metabolismo , Somatotrofos/citología , Línea Celular , Supervivencia Celular/efectos de los fármacos , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Didesoxinucleósidos/farmacología , Hormona Liberadora de Hormona del Crecimiento/metabolismo , Humanos , Nevirapina/farmacología , Fosforilación , Ritonavir/farmacología , Somatotrofos/efectos de los fármacos , Somatotrofos/metabolismo , Transfección
10.
Endocrinology ; 159(9): 3287-3305, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30085028

RESUMEN

The pituitary gland is a critical organ that is necessary for many physiological processes, including growth, reproduction, and stress response. The secretion of pituitary hormones from specific cell types regulates these essential processes. Pituitary hormone cell types arise from a common pool of pituitary progenitors, and mutations that disrupt the formation and differentiation of pituitary progenitors result in hypopituitarism. Canonical WNT signaling through CTNNB1 (ß-catenin) is known to regulate the formation of the POU1F1 lineage of pituitary cell types. When ß-catenin is deleted during the initial formation of the pituitary progenitors, Pou1f1 is not transcribed, which leads to the loss of the POU1F1 lineage. However, when ß-catenin is deleted after lineage specification, there is no observable effect. Similarly, the generation of a ß-catenin gain-of-function allele in early pituitary progenitors or stem cells results in the formation of craniopharyngiomas, whereas stimulating ß-catenin in differentiated cell types has no effect. PROP1 is a pituitary-specific transcription factor, and the peak of PROP1 expression coincides with a critical time point in pituitary organogenesis-that is, after pituitary progenitor formation but before lineage specification. We used a Prop1-cre to conduct both loss- and gain-of-function studies on ß-catenin during this critical time point. Our results demonstrate that pituitary progenitors remain sensitive to both loss and gain of ß-catenin at this time point, and that either manipulation results in hypopituitarism.


Asunto(s)
Craneofaringioma/genética , Hipopituitarismo/genética , Hipófisis/embriología , Células Madre/metabolismo , Factor de Transcripción Pit-1/genética , beta Catenina/genética , Animales , Linaje de la Célula , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Lactotrofos/citología , Ratones , Organogénesis , Hipófisis/metabolismo , Somatotrofos/citología , Células Madre/citología , Tirotrofos/citología , Factor de Transcripción Pit-1/metabolismo , Vía de Señalización Wnt
11.
Mol Cell Biol ; 38(19)2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29987187

RESUMEN

Peptidylarginine deiminase (PAD) enzymes convert histone arginine residues into citrulline to modulate chromatin organization and gene expression. Although PADs are expressed in anterior pituitary gland cells, their functional role and expression in pituitary adenomas are unknown. To begin to address these issues, we first examined normal human pituitaries and pituitary adenomas and found that PAD2, PAD4, and citrullinated histones are highest in prolactinomas and somatoprolactinomas. In the somatoprolactinoma-derived GH3 cell line, PADs citrullinate histone H3, which is attenuated by a pan-PAD inhibitor. RNA sequencing and chromatin immunoprecipitation (ChIP) studies show that the expression of microRNAs (miRNAs) let-7c-2, 23b, and 29c is suppressed by histone citrullination. Our studies demonstrate that these miRNAs directly target the mRNA of the oncogenes encoding HMGA, insulin-like growth factor 1 (IGF-1), and N-MYC, which are highly implicated in human prolactinoma/somatoprolactinoma pathogenesis. Our results are the first to define a direct role for PAD-catalyzed histone citrullination in miRNA expression, which may underlie the etiology of prolactinoma and somatoprolactinoma tumors through regulation of oncogene expression.


Asunto(s)
Histonas/metabolismo , Lactotrofos/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Oncogenes , ARN Mensajero/genética , ARN Mensajero/metabolismo , Somatotrofos/metabolismo , Células Cultivadas , Citrulinación/genética , Epigénesis Genética , Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas HMGA/genética , Proteínas HMGA/metabolismo , Código de Histonas/genética , Histonas/química , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Lactotrofos/citología , Modelos Biológicos , Proteína Proto-Oncogénica N-Myc/genética , Proteína Proto-Oncogénica N-Myc/metabolismo , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/metabolismo , Prolactinoma/genética , Prolactinoma/metabolismo , Arginina Deiminasa Proteína-Tipo 2 , Arginina Deiminasa Proteína-Tipo 4 , Desiminasas de la Arginina Proteica/antagonistas & inhibidores , Desiminasas de la Arginina Proteica/genética , Desiminasas de la Arginina Proteica/metabolismo , Somatotrofos/citología
12.
Endocrinology ; 159(8): 2891-2904, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29796621

RESUMEN

The process by which the somatotrope lineage emerges in the developing pituitary is regulated by the activity of specific signaling and transcription factors expressed during development. We set out to understand the contribution of FOXO1 to that process by using a mouse model in which FOXO1 is prematurely expressed in the pituitary primordium. Expression of FOXO1 in the oral ectoderm as early as embryonic day (e)9.5 resulted in pituitary gland hypoplasia and reduced expression of anterior lobe hormone transcripts at e18.5. Of note, the relative numbers of somatotropes and thyrotropes were also decreased at e18.5. LHX3 and PITX2, markers of pituitary identity, were present in a reduced number of cells during the formation of the Rathke pouch. Thus, premature expression of FOXO1 may affect adoption of pituitary identity during differentiation. Our results demonstrate that the timing of FOXO1 activation affects its role in pituitary gland organogenesis and somatotrope differentiation.


Asunto(s)
Proteína Forkhead Box O1/genética , Regulación del Desarrollo de la Expresión Génica , Organogénesis/genética , Adenohipófisis/embriología , Animales , Diferenciación Celular/genética , Linaje de la Célula , Ectodermo/embriología , Ectodermo/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Ratones , Tamaño de los Órganos , Hipófisis/citología , Hipófisis/embriología , Hipófisis/metabolismo , Hipófisis/patología , Adenohipófisis/citología , Adenohipófisis/metabolismo , Adenohipófisis/patología , Somatotrofos/citología , Somatotrofos/metabolismo , Tirotrofos/citología , Tirotrofos/metabolismo , Factores de Tiempo , Factores de Transcripción/metabolismo , Proteína del Homeodomínio PITX2
13.
PLoS One ; 11(10): e0164131, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27706259

RESUMEN

Aryl hydrocarbon receptor interacting protein (AIP) is thought to be a tumor suppressor gene, as indicated by a mutational analysis of pituitary somatotroph adenomas. However, the physiological significance of AIP inactivation in somatotroph cells remains unclear. Using CRISPR/Cas9, we identified a GH3 cell clone (termed GH3-FTY) in which Aip was genetically disrupted, and subsequently investigated its character with respect to growth hormone (Gh) synthesis and proliferation. Compared with GH3, GH3-FTY cells showed remarkably increased Gh production and a slight increase in cell proliferation. Gh-induced Stat3 phosphorylation is known to be a mechanism of Gh oversecretion in GH3. Interestingly, phosphorylated-Stat3 expression in GH3-FTY cells was increased more compared with GH3 cells, suggesting a stronger drive for this mechanism in GH3-FTY. The phenotypes of GH3-FTY concerning Gh overproduction, cell proliferation, and increased Stat3 phosphorylation were significantly reversed by the exogenous expression of Aip. GH3-FTY cells were less sensitive to somatostatin than GH3 cells in the suppression of cell proliferation, which might be associated with the reduced expression of somatostatin receptor type 2. GH3-FTY xenografts in BALB/c nude mice (GH3-FTY mice) formed more mitotic somatotroph tumors than GH3 xenografts (GH3 mice), as also evidenced by increased Ki67 scores. GH3-FTY mice were also much larger and had significantly higher plasma Gh levels than GH3 mice. Furthermore, GH3-FTY mice showed relative insulin resistance compared with GH3 mice. In conclusion, we established a somatotroph cell line, GH3-FTY, which possessed prominent Gh secretion and mitotic features associated with the disruption of Aip.


Asunto(s)
Adenoma/patología , Adenoma Hipofisario Secretor de Hormona del Crecimiento/patología , Hormona del Crecimiento/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Factor de Transcripción STAT3/metabolismo , Somatotrofos/citología , Animales , Línea Celular , Proliferación Celular , Silenciador del Gen , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Fosforilación , Ratas , Somatotrofos/metabolismo , Somatotrofos/trasplante , Regulación hacia Arriba
14.
Endocrinology ; 157(11): 4351-4363, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27631552

RESUMEN

The etiology for half of congenital hypopituitarism cases is unknown. Our long-term goal is to expand the molecular diagnoses for congenital hypopituitarism by identifying genes that contribute to this condition. We have previously shown that the forkhead box transcription factor, FOXO1, is present in approximately half of somatotropes at embryonic day (e) 18.5, suggesting it may have a role in somatotrope differentiation or function. To elucidate the role of FOXO1 in somatotrope differentiation and function, Foxo1 was conditionally deleted from the anterior pituitary (Foxo1Δpit). Uncommitted progenitor cells are maintained and able to commit to the somatotrope lineage normally based on the expression patterns of Sox2, a marker of uncommitted pituitary progenitors, and Pou1f1 (also known as Pit1), which marks committed progenitors. Interestingly, Foxo1Δpit embryonic mice exhibit delayed somatotrope differentiation as evidenced by an almost complete absence of GH immunoreactivity at e16.5 and reduced expression of Gh at e18.5 and postnatal day (P) 3. Consistent with this conclusion, expression of GHRH receptor, a marker of terminally differentiated somatotropes, is significantly reduced at e18.5 and P3 in the absence of FOXO1. The mechanism of FOXO1 regulation of somatotrope differentiation may involve the basic helix-loop-helix transcription factor, Neurod4, which has been implicated in somatotrope differentiation and is significantly reduced in Foxo1Δpit mice. Foxo1Δpit mice do not exhibit growth defects, and at P21 their pituitary glands exhibit a normal distribution of somatotropes. These studies demonstrate that FOXO1 is important for initial somatotrope specification embryonically but is dispensable for postnatal somatotrope expansion and growth.


Asunto(s)
Proteína Forkhead Box O1/metabolismo , Somatotrofos/citología , Somatotrofos/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Embrión de Mamíferos/metabolismo , Exones/genética , Femenino , Hormona Folículo Estimulante/genética , Hormona Folículo Estimulante/metabolismo , Proteína Forkhead Box O1/genética , Inmunohistoquímica , Hibridación in Situ , Hormona Luteinizante/genética , Hormona Luteinizante/metabolismo , Ratones , Hipófisis/citología , Hipófisis/metabolismo , Embarazo , Células Madre/citología , Células Madre/metabolismo , Factores de Tiempo
15.
Endocrinology ; 157(7): 2735-49, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27183316

RESUMEN

p53 is a well-known tumor suppressor that plays multiple biological roles, including the capacity to modulate metabolism at different levels. However, its metabolic role in brown adipose tissue (BAT) remains largely unknown. Herein we sought to investigate the physiological role of endogenous p53 in BAT and its implication on BAT thermogenic activity and energy balance. To this end, we generated and characterized global p53-null mice and mice lacking p53 specifically in BAT. Additionally we performed gain-and-loss-of-function experiments in the BAT of adult mice using virogenetic and pharmacological approaches. BAT was collected and analyzed by immunohistochemistry, thermography, real-time PCR, and Western blot. p53-deficient mice were resistant to diet-induced obesity due to increased energy expenditure and BAT activity. However, the deletion of p53 in BAT using a Myf5-Cre driven p53 knockout did not show any changes in body weight or the expression of thermogenic markers. The acute inhibition of p53 in the BAT of adult mice slightly increased body weight and inhibited BAT thermogenesis, whereas its overexpression in the BAT of diet-induced obese mice reduced body weight and increased thermogenesis. On the other hand, pharmacological activation of p53 improves body weight gain due to increased BAT thermogenesis by sympathetic nervous system in obese adult wild-type mice but not in p53(-/-) animals. These results reveal that p53 regulates BAT metabolism by coordinating body weight and thermogenesis, but these metabolic actions are tissue specific and also dependent on the developmental stage.


Asunto(s)
Tejido Adiposo Pardo/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Obesidad/genética , Termogénesis/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética , Tejido Adiposo Pardo/metabolismo , Animales , Composición Corporal/efectos de los fármacos , Composición Corporal/genética , Peso Corporal/genética , Línea Celular , Doxorrubicina/farmacología , Masculino , Ratones , Ratones Noqueados , Obesidad/metabolismo , Ratas , Somatotrofos/citología , Somatotrofos/efectos de los fármacos , Somatotrofos/metabolismo , Termogénesis/genética , Proteína p53 Supresora de Tumor/agonistas , Proteína p53 Supresora de Tumor/metabolismo
16.
Cell Signal ; 27(9): 1720-30, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26022182

RESUMEN

The transduction pathway mediating the inhibitory effect that TRH exerts on r-ERG channels has been thoroughly studied in GH3 rat pituitary cells but some elements have yet to be discovered, including those involved in a phosphorylation event(s). Using a quantitative phosphoproteomic approach we studied the changes in phosphorylation caused by treatment with 1µM TRH for 5min in GH3 cells. The activating residues of Erk2 and Erk1 undergo phosphorylation increases of 5.26 and 4.87 fold, respectively, in agreement with previous reports of ERK activation by TRH in GH3 cells. Thus, we studied the possible involvement of ERK pathway in the signal transduction from TRH receptor to r-ERG channels. The MEK inhibitor U0126 at 0.5µM caused no major blockade of the basal r-ERG current, but impaired the TRH inhibitory effect on r-ERG. Indeed, the TRH effect on r-ERG was also reduced when GH3 cells were transfected with siRNAs against either Erk1 or Erk2. Using antibodies, we found that TRH treatment also causes activating phosphorylation of Rsk. The TRH effect on r-ERG current was also impaired when cells were transfected with any of two different siRNAs mixtures against Rsk1. However, treatment of GH3 cells with 20nM EGF for 5min, which causes ERK and RSK activation, had no effect on the r-ERG currents. Therefore, we conclude that in the native GH3 cell system, ERK and RSK are involved in the pathway linking TRH receptor to r-ERG channel inhibition, but additional components must participate to cause such inhibition.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Somatotrofos/metabolismo , Animales , Línea Celular , Canales de Potasio Éter-A-Go-Go/genética , Transporte Iónico/efectos de los fármacos , Transporte Iónico/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa 3 Activada por Mitógenos/genética , Ratas , Receptores de Hormona Liberadora de Tirotropina/genética , Receptores de Hormona Liberadora de Tirotropina/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética , Somatotrofos/citología , Hormona Liberadora de Tirotropina/genética , Hormona Liberadora de Tirotropina/metabolismo
17.
PLoS One ; 10(3): e0120010, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25822178

RESUMEN

To test the role of wtPIT-1 (PITWT) or PIT-1 (R271W) (PIT271) in somatolactotroph cells, we established, using inducible lentiviral vectors, sublines of GH4C1 somatotroph cells that allow the blockade of the expression of endogenous PIT-1 and/or the expression of PITWT or PIT271, a dominant negative mutant of PIT-1 responsible for Combined Pituitary Hormone Deficiency in patients. Blocking expression of endogenous PIT-1 induced a marked decrease of cell proliferation. Overexpressing PITWT twofold led also to a dose-dependent decrease of cell proliferation that was accompanied by cell death. Expression of PIT271 induced a strong dose-dependent decrease of cell proliferation accompanied by a very pronounced cell death. These actions of PIT271 are independent of its interaction/competition with endogenous PIT-1, as they were unchanged when expression of endogenous PIT-1 was blocked. All these actions are specific for somatolactotroph cells, and could not be observed in heterologous cells. Cell death induced by PITWT or by PIT271 was accompanied by DNA fragmentation, but was not inhibited by inhibitors of caspases, autophagy or necrosis, suggesting that this cell death is a caspase-independent apoptosis. Altogether, our results indicate that under normal conditions PIT-1 is important for the maintenance of cell proliferation, while when expressed at supra-normal levels it induces cell death. Through this dual action, PIT-1 may play a role in the expansion/regression cycles of pituitary lactotroph population during and after lactation. Our results also demonstrate that the so-called "dominant-negative" action of PIT271 is independent of its competition with PIT-1 or a blockade of the actions of the latter, and are actions specific to this mutant variant of PIT-1.


Asunto(s)
Somatotrofos/fisiología , Factor de Transcripción Pit-1/fisiología , Adenoma/genética , Adenoma/patología , Adenoma/fisiopatología , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Apoptosis/fisiología , Secuencia de Bases , Línea Celular , Proliferación Celular/fisiología , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Chlorocebus aethiops , Humanos , Lactotrofos/citología , Lactotrofos/fisiología , Datos de Secuencia Molecular , Proteínas Mutantes/genética , Proteínas Mutantes/fisiología , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/patología , Neoplasias Hipofisarias/fisiopatología , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Ratas , Somatotrofos/citología , Factor de Transcripción Pit-1/antagonistas & inhibidores , Factor de Transcripción Pit-1/genética , Células Tumorales Cultivadas
18.
Endocrine ; 48(3): 929-36, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25151402

RESUMEN

A variety of ion channels are expressed in the plasma membrane of somatotropes within the anterior pituitary gland. Modification of these channels is linked to intracellular Ca2+ levels and therefore to hormone secretion. Previous investigations have shown that the gut-derived orexigenic peptide hormone ghrelin and synthetic GH-releasing peptides (GHRPs) stimulate release of growth hormone (GH) and increase the number of functional voltage-gated Ca2+ and Na+ channels in the membrane of clonal GC somatotropes. Here, we reveal that chronic treatment with ghrelin and its synthetic analog GHRP-6 also increases GH release from bovine pituitary somatotropes in culture, and that this action is associated with a significant increase in Na+ macroscopic current. Consistent with this, Na+ current blockade with tetrodotoxin (TTX) abolished the ghrelin- and GHRP-6-induced increase in GH release. Furthermore, semi-quantitative and real-time RT-PCR analysis revealed an upregulation in the transcript levels of GH, as well as of NaV1.1 and NaV1.2, two isoforms of TTX-sensitive Na+ channels expressed in somatotropes, after treatment with ghrelin or GHRP-6. These findings improve our knowledge on (i) the cellular mechanisms involved in the control of GH secretion, (ii) the molecular diversity of Na+ channels in pituitary somatotropes, and (iii) the regulation of GH and Na+ channel gene expression by ghrelin and GHRPs.


Asunto(s)
Ghrelina/farmacología , Hormona del Crecimiento/biosíntesis , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Canal de Sodio Activado por Voltaje NAV1.2/metabolismo , Somatotrofos/efectos de los fármacos , Animales , Bovinos , Células Cultivadas , Masculino , Canal de Sodio Activado por Voltaje NAV1.1/genética , Canal de Sodio Activado por Voltaje NAV1.2/genética , Oligopéptidos/farmacología , Hipófisis/citología , Hipófisis/efectos de los fármacos , Hipófisis/metabolismo , Somatotrofos/citología , Somatotrofos/metabolismo , Regulación hacia Arriba/efectos de los fármacos
19.
Am J Physiol Endocrinol Metab ; 307(3): E326-34, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24939736

RESUMEN

Klotho is a transmembranal protein highly expressed in the kidneys, choroid plexus, and anterior pituitary. Klotho can also be cleaved and shed and acts as a circulating hormone. Klotho-deficient mice (kl/kl mice) develop a phenotype resembling early aging. Several lines of evidence suggest a role for klotho in the regulation of growth hormone (GH) secretion. The kl/kl mice are smaller compared with their wild-type counterparts, and their somatotropes show reduced numbers of secretory granules. Moreover, klotho is a potent inhibitor of the IGF-I pathway, a negative regulator of GH secretion. Therefore, we hypothesized that klotho may enhance GH secretion. The effect of klotho on GH secretion was examined in GH3 rat somatotrophs, cultured rat pituitaries, and cultured human GH-secreting adenomas. In all three models, klotho treatment increased GH secretion. Prolonged treatment of mice with intraperitoneal klotho injections increased mRNA levels of IGF-I and IGF-I-binding protein-3 mRNA in the liver, reflecting increased serum GH levels. In accord with its ability to inhibit the IGF-I pathway, klotho partially restored the inhibitory effect of IGF-I on GH secretion. Klotho is known to be a positive regulator of basic bFGF signaling. We studied rat pituitaries and human adenoma cultures and noted that bFGF increased GH secretion and stimulated ERK1/2 phosphorylation. Both effects were augmented following treatment with klotho. Taken together, our data indicate for the first time that klotho is a positive regulator of GH secretion and suggest the IGF-I and bFGF pathways as potential mediators of this effect.


Asunto(s)
Envejecimiento , Glucuronidasa/metabolismo , Hormona del Crecimiento/metabolismo , Hígado/metabolismo , Somatotrofos/metabolismo , Adenoma/metabolismo , Adenoma/patología , Animales , Línea Celular , Células Cultivadas , Regulación de la Expresión Génica , Glucuronidasa/genética , Hormona del Crecimiento/sangre , Adenoma Hipofisario Secretor de Hormona del Crecimiento/metabolismo , Adenoma Hipofisario Secretor de Hormona del Crecimiento/patología , Humanos , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Proteínas Klotho , Masculino , Ratones , Ratones Desnudos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Proteínas del Tejido Nervioso/administración & dosificación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Ratas , Proteínas Recombinantes/metabolismo , Somatotrofos/citología , Somatotrofos/patología , Células Tumorales Cultivadas
20.
Mol Cell Biochem ; 391(1-2): 67-75, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24553818

RESUMEN

This study investigated the roles of Ras, ERK, and Akt in the glucocorticoid-induced differentiation of growth hormone-producing pituitary cells in vitro. Pituicytes isolated from day-18 rat embryos were cultured with 50 mM dexamethasone in addition to specific inhibitors of Ras (manumycin; 0.5, 5, 50 nM), ERK (U0126, 10 µM), or Akt (LY294002, 25 µM). Differentiation was assessed using immunofluorescent staining of intracellular growth hormone. Radioimmunoassay and Western blot analyses were used to determine levels of secreted and intracellular growth hormone, respectively. Manumycin reduced the fraction of growth hormone-positive cells and dexamethasone-induced growth hormone secretion in a dose-dependent manner (both P < 0.001). In the absence of dexamethasone, LY294002 and U0126 did not alter the fraction of growth hormone-positive cells or intracellular growth hormone protein expression or secretion. Both LY294002 and U0126 alone significantly attenuated the fraction of dexamethasone-treated GH-positive cells and the secretion of GH compared to those of cells treated only with dexamethasone (50 nM for 44 h or 48 h) (all P < 0.05). Dexamethasone treatment alone did not change GH protein levels. Treatment of cells with a combination of LY294402 and U0126 significantly attenuated the fraction of dexamethasone-treated GH-positive cells, GH protein levels, and GH secretion compared to cells treated with dexamethasone alone (all P < 0.05). Moreover, dexamethasone-induced phosphorylation of GTP-Ras, ERK, and Akt was significantly attenuated by exposure to the respective inhibitors (P < 0.05). Taken together, our results indicate that Ras, ERK, and Akt are key effectors in the glucocorticoid-induced differentiation of growth hormone-secreting cells.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Embrión de Mamíferos/citología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glucocorticoides/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Somatotrofos/citología , Proteínas ras/metabolismo , Animales , Butadienos/farmacología , Supervivencia Celular/efectos de los fármacos , Cromonas/farmacología , Dexametasona/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Hormona del Crecimiento/metabolismo , Morfolinas/farmacología , Nitrilos/farmacología , Fosforilación/efectos de los fármacos , Polienos/farmacología , Alcamidas Poliinsaturadas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Ratas Sprague-Dawley , Somatotrofos/efectos de los fármacos , Somatotrofos/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA