Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Oncogene ; 40(45): 6354-6368, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34588620

RESUMEN

It is unclear how loss-of-function germline mutations in the widely-expressed co-chaperone AIP, result in young-onset growth hormone secreting pituitary tumours. The RET receptor, uniquely co-expressed in somatotrophs with PIT1, induces apoptosis when unliganded, while RET supports cell survival when it is bound to its ligand. We demonstrate that at the plasma membrane, AIP is required to form a complex with monomeric-intracellular-RET, caspase-3 and PKCδ resulting in PIT1/CDKN2A-ARF/p53-apoptosis pathway activation. AIP-deficiency blocks RET/caspase-3/PKCδ activation preventing PIT1 accumulation and apoptosis. The presence or lack of the inhibitory effect on RET-induced apoptosis separated pathogenic AIP variants from non-pathogenic ones. We used virogenomics in neonatal rats to demonstrate the effect of mutant AIP protein on the RET apoptotic pathway in vivo. In adult male rats altered AIP induces elevated IGF-1 and gigantism, with pituitary hyperplasia through blocking the RET-apoptotic pathway. In females, pituitary hyperplasia is induced but IGF-1 rise and gigantism are blunted by puberty. Somatotroph adenomas from pituitary-specific Aip-knockout mice overexpress the RET-ligand GDNF, therefore, upregulating the survival pathway. Somatotroph adenomas from patients with or without AIP mutation abundantly express GDNF, but AIP-mutated tissues have less CDKN2A-ARF expression. Our findings explain the tissue-specific mechanism of AIP-induced somatotrophinomas and provide a previously unknown tumorigenic mechanism, opening treatment avenues for AIP-related tumours.


Asunto(s)
Acromegalia/genética , Mutación de Línea Germinal , Gigantismo/genética , Adenoma Hipofisario Secretor de Hormona del Crecimiento/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Acromegalia/metabolismo , Animales , Animales Recién Nacidos , Apoptosis , Línea Celular , Femenino , Técnicas de Inactivación de Genes , Gigantismo/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Adenoma Hipofisario Secretor de Hormona del Crecimiento/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones , Especificidad de Órganos , Proteínas Proto-Oncogénicas c-ret/metabolismo , Ratas , Transducción de Señal
3.
Cell Metab ; 33(9): 1820-1835.e9, 2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34343501

RESUMEN

Appropriate cristae remodeling is a determinant of mitochondrial function and bioenergetics and thus represents a crucial process for cellular metabolic adaptations. Here, we show that mitochondrial cristae architecture and expression of the master cristae-remodeling protein OPA1 in proopiomelanocortin (POMC) neurons, which are key metabolic sensors implicated in energy balance control, is affected by fluctuations in nutrient availability. Genetic inactivation of OPA1 in POMC neurons causes dramatic alterations in cristae topology, mitochondrial Ca2+ handling, reduction in alpha-melanocyte stimulating hormone (α-MSH) in target areas, hyperphagia, and attenuated white adipose tissue (WAT) lipolysis resulting in obesity. Pharmacological blockade of mitochondrial Ca2+ influx restores α-MSH and the lipolytic program, while improving the metabolic defects of mutant mice. Chemogenetic manipulation of POMC neurons confirms a role in lipolysis control. Our results unveil a novel axis that connects OPA1 in POMC neurons with mitochondrial cristae, Ca2+ homeostasis, and WAT lipolysis in the regulation of energy balance.


Asunto(s)
Lipólisis , Proopiomelanocortina , Tejido Adiposo/metabolismo , Animales , GTP Fosfohidrolasas , Homeostasis , Ratones , Neuronas/metabolismo , Proopiomelanocortina/metabolismo
4.
Artículo en Inglés | MEDLINE | ID: mdl-33071961

RESUMEN

The RET tyrosine kinase receptor is expressed by the endocrine somatotroph cells of the pituitary where it has important functions regulating survival/apoptosis. However, RET is also expressed by the GPS pituitary stem cells localized in a niche between the adenopituitary and the intermediate lobe. To bind any of its four ligands, RET needs one of four co-receptors called GFRα1-4. It has been previously shown that GFRα1 is expressed by somatotroph cells and acromegaly tumors. GFRα2 was shown to be expressed by pituitary stem cells. GFRα4 was proposed as not expressed in the pituitary. Here we study the RNA and protein expression of the four GFRα co-receptors for RET in rat and human pituitary. The four co-receptors were abundantly expressed at the RNA level both in rat and human pituitary, although GFRα4 was the less abundant. Multiple immunofluorescence for each co-receptor and ß-catenin, a marker of stem cell niche was performed. The four GFRα co-receptors were co-expressed by the GPS cells at the niche colocalizing with ß-catenin. Isolated individual scattered cells positive for one or other receptor could be found through the adenopituitary with low ß-catenin expression. Some of them co-express GFRα1 and PIT1. Immunohistochemistry in normal human pituitary confirmed the data. Our data suggest that the redundancy of GFRα co-expression is a self-supportive mechanism which ensures niche maintenance and proper differentiation.


Asunto(s)
Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Hipófisis/metabolismo , Proteínas Proto-Oncogénicas c-ret/metabolismo , Células Madre/metabolismo , Animales , Femenino , Humanos , Masculino , Ratas , Nicho de Células Madre
5.
EBioMedicine ; 43: 537-552, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30975543

RESUMEN

BACKGROUND: Acromegaly is produced by excess growth hormone secreted by a pituitary adenoma of somatotroph cells (ACRO). First-line therapy, surgery and adjuvant therapy with somatostatin analogs, fails in 25% of patients. There is no predictive factor of resistance to therapy. New therapies are investigated using few dispersed tumor cells in acute primary cultures in standard conditions where the cells do not grow, or using rat pituitary cell lines that do not maintain the full somatotroph phenotype. The RET/PIT1/p14ARF/p53 pathway regulates apoptosis in normal pituitary somatotrophs whereas the RET/GDNF pathway regulates survival, controlling PIT1 levels and blocking p14ARF (ARF) and p53 expression. METHODS: We investigated these two RET pathways in a prospective series of 32 ACRO and 63 non-functioning pituitary adenomas (NFPA), studying quantitative RNA and protein gene expression for molecular-clinical correlations and how the RET pathway might be implicated in therapeutic success. Clinical data was collected during post-surgical follow-up. We also established new'humanized' pituitary cultures, allowing 20 repeated passages and maintaining the pituitary secretory phenotype, and tested five multikinase inhibitors (TKI: Vandetanib, Lenvatinib, Sunitinib, Cabozantinib and Sorafenib) potentially able to act on the GDNF-induced RET dimerization/survival pathway. Antibody arrays investigated intracellular molecular pathways. FINDINGS: In ACRO, there was specific enrichment of all genes in both RET pathways, especially GDNF. ARF and GFRA4 gene expression were found to be opposing predictors of response to first-line therapy. ARF cut-off levels, calculated categorizing by GNAS mutation, were predictive of good response (above) or resistance (below) to therapy months later. Sorafenib, through AMPK, blocked the GDNF/AKT survival action without altering the RET apoptotic pathway. INTERPRETATION: Tumor ARF mRNA expression measured at the time of the surgery is a prognosis factor in acromegaly. The RET inhibitor, Sorafenib, is proposed as a potential treatment for resistant ACRO. FUND: This project was supported by national grants from Agencia Estatal de Investigación (AEI) and Instituto Investigación Carlos III, with participation of European FEDER funds, to IB (PI150056) and CVA (BFU2016-76973-R). It was also supported initially by a grant from the Investigator Initiated Research (IIR) Program (WI177773) and by a non-restricted Research Grant from Pfizer Foundation to IB. Some of the pituitary acromegaly samples were collected in the framework of the Spanish National Registry of Acromegaly (REMAH), partially supported by an unrestricted grant from Novartis to the Spanish Endocrine Association (SEEN). CVA is also supported from a grant of Medical Research Council UK MR/M018539/1.


Asunto(s)
Acromegalia/diagnóstico , Acromegalia/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Proteínas Proto-Oncogénicas c-ret/metabolismo , Factor de Transcripción Pit-1/metabolismo , Proteína p14ARF Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Acromegalia/genética , Acromegalia/terapia , Animales , Apoptosis/genética , Biomarcadores , Terapia Combinada , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Humanos , Inmunohistoquímica , Modelos Biológicos , Mutación , Neoplasias Hipofisarias/diagnóstico , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/metabolismo , Pronóstico , Proteínas Proto-Oncogénicas c-ret/genética , Ratas , Transducción de Señal , Factor de Transcripción Pit-1/genética , Resultado del Tratamiento , Proteína p14ARF Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/genética
6.
Sci Rep ; 7: 42937, 2017 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-28262687

RESUMEN

IGSF1 (Immunoglobulin Superfamily 1) gene defects cause central hypothyroidism and macroorchidism. However, the pathogenic mechanisms of the disease remain unclear. Based on a patient with a full deletion of IGSF1 clinically followed from neonate to adulthood, we investigated a common pituitary origin for hypothyroidism and macroorchidism, and the role of IGSF1 as regulator of pituitary hormone secretion. The patient showed congenital central hypothyroidism with reduced TSH biopotency, over-secretion of FSH at neonatal minipuberty and macroorchidism from 3 years of age. His markedly elevated inhibin B was unable to inhibit FSH secretion, indicating a status of pituitary inhibin B resistance. We show here that IGSF1 is expressed both in thyrotropes and gonadotropes of the pituitary and in Leydig and germ cells in the testes, but at very low levels in Sertoli cells. Furthermore, IGSF1 stimulates transcription of the thyrotropin-releasing hormone receptor (TRHR) by negative modulation of the TGFß1-Smad signaling pathway, and enhances the synthesis and biopotency of TSH, the hormone secreted by thyrotropes. By contrast, IGSF1 strongly down-regulates the activin-Smad pathway, leading to reduced expression of FSHB, the hormone secreted by gonadotropes. In conclusion, two relevant molecular mechanisms linked to central hypothyroidism and macroorchidism in IGSF1 deficiency are identified, revealing IGSF1 as an important regulator of TGFß/Activin pathways in the pituitary.


Asunto(s)
Activinas/metabolismo , Hormona Folículo Estimulante de Subunidad beta/metabolismo , Hipotiroidismo/patología , Inmunoglobulinas/genética , Proteínas de la Membrana/genética , Receptores de Hormona Liberadora de Tirotropina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Análisis Mutacional de ADN , Hormona Folículo Estimulante de Subunidad beta/genética , Estudios de Seguimiento , Eliminación de Gen , Humanos , Hipotiroidismo/genética , Recién Nacido , Masculino , Ratones , Hipófisis/metabolismo , Hipófisis/patología , Regiones Promotoras Genéticas , Ratas , Ratas Wistar , Receptores de Hormona Liberadora de Tirotropina/genética , Proteínas Smad/metabolismo , Testículo/metabolismo , Testículo/patología
7.
Neuroendocrinology ; 101(3): 175-92, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25662152

RESUMEN

The recent demonstration using genetic tracing that in the adult pituitary stem cells are normally recruited from the niche in the marginal zone and differentiate into secretory cells in the adenopituitary has elegantly confirmed the proposal made when the pituitary stem cell niche was first discovered 5 years ago. Some of the early controversies have also been resolved. However, many questions remain, such as which are the markers that make a pituitary stem cell truly unique and the exact mechanisms that trigger recruitment from the niche. Little is known about the processes of commitment and differentiation once a stem cell has left the niche. Moreover, the acceptance that pituitary cells are renewed by stem cells implies the existence of regulated mechanisms of cell death in differentiated cells which must themselves be explained. The demonstration of an apoptotic pathway mediated by RET/caspase 3/Pit-1/Arf/p53 in normal somatotrophs is therefore an important step towards understanding how pituitary cell number is regulated. Further work will elucidate how the rates of the three processes of cell renewal, differentiation and apoptosis are balanced in tissue homeostasis after birth, but altered in pituitary hyperplasia in response to physiological stimuli such as puberty and lactation. Thus, we can aim to understand the mechanisms underlying human disease due to insufficient (hypopituitarism) or excess (pituitary tumor) cell numbers.


Asunto(s)
Células Madre Adultas/fisiología , Hipófisis/fisiología , Nicho de Células Madre , Células Madre Adultas/citología , Animales , Apoptosis , Diferenciación Celular , Humanos , Modelos Animales , Hipófisis/citología
8.
Endocrinology ; 155(11): 4329-40, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25137025

RESUMEN

Acromegaly is caused by somatotroph cell adenomas (somatotropinomas [ACROs]), which secrete GH. Human and rodent somatotroph cells express the RET receptor. In rodents, when normal somatotrophs are deprived of the RET ligand, GDNF (Glial Cell Derived Neurotrophic Factor), RET is processed intracellularly to induce overexpression of Pit1 [Transcription factor (gene : POUF1) essential for transcription of Pituitary hormones GH, PRL and TSHb], which in turn leads to p19Arf/p53-dependent apoptosis. Our purpose was to ascertain whether human ACROs maintain the RET/Pit1/p14ARF/p53/apoptosis pathway, relative to nonfunctioning pituitary adenomas (NFPAs). Apoptosis in the absence and presence of GDNF was studied in primary cultures of 8 ACROs and 3 NFPAs. Parallel protein extracts were analyzed for expression of RET, Pit1, p19Arf, p53, and phospho-Akt. When GDNF deprived, ACRO cells, but not NFPAs, presented marked level of apoptosis that was prevented in the presence of GDNF. Apoptosis was accompanied by RET processing, Pit1 accumulation, and p14ARF and p53 induction. GDNF prevented all these effects via activation of phospho-AKT. Overexpression of human Pit1 (hPit1) directly induced p19Arf/p53 and apoptosis in a pituitary cell line. Using in silico studies, 2 CCAAT/enhancer binding protein alpha (cEBPα) consensus-binding sites were found to be 100% conserved in mouse, rat, and hPit1 promoters. Deletion of 1 cEBPα site prevented the RET-induced increase in hPit1 promoter expression. TaqMan qRT-PCR (real time RT-PCR) for RET, Pit1, Arf, TP53, GDNF, steroidogenic factor 1, and GH was performed in RNA from whole ACRO and NFPA tumors. ACRO but not NFPA adenomas express RET and Pit1. GDNF expression in the tumors was positively correlated with RET and negatively correlated with p53. In conclusion, ACROs maintain an active RET/Pit1/p14Arf/p53/apoptosis pathway that is inhibited by GDNF. Disruption of GDNF's survival function might constitute a new therapeutic route in acromegaly.


Asunto(s)
Adenoma/patología , Apoptosis/efectos de los fármacos , Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Adenoma Hipofisario Secretor de Hormona del Crecimiento/patología , Neoplasias Hipofisarias/patología , Adenoma/genética , Animales , Apoptosis/genética , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Adenoma Hipofisario Secretor de Hormona del Crecimiento/genética , Humanos , Neoplasias Hipofisarias/genética , Proteínas Proto-Oncogénicas c-ret/fisiología , Ratas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factor de Transcripción Pit-1/fisiología , Células Tumorales Cultivadas , Proteína p14ARF Supresora de Tumor/fisiología , Proteína p53 Supresora de Tumor/fisiología
9.
J Clin Endocrinol Metab ; 98(6): 2431-41, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23539720

RESUMEN

CONTEXT: Mechanisms of thyroid physiology and cancer are principally studied in follicular cell lines. However, human thyroid cancer lines were found to be heavily contaminated by other sources, and only one supposedly normal-thyroid cell line, immortalized with SV40 antigen, is available. In primary culture, human follicular cultures lose their phenotype after passage. We hypothesized that the loss of the thyroid phenotype could be related to culture conditions in which human cells are grown in medium optimized for rodent culture, including hormones with marked differences in its affinity for the relevant rodent/human receptor. OBJECTIVE: The objective of the study was to define conditions that allow the proliferation of primary human follicular thyrocytes for many passages without losing phenotype. METHODS: Concentrations of hormones, transferrin, iodine, oligoelements, antioxidants, metabolites, and ethanol were adjusted within normal homeostatic human serum ranges. Single cultures were identified by short tandem repeats. Human-rodent interspecies contamination was assessed. RESULTS: We defined an humanized 7 homeostatic additives medium enabling growth of human thyroid cultures for more than 20 passages maintaining thyrocyte phenotype. Thyrocytes proliferated and were grouped as follicle-like structures; expressed Na+/I- symporter, pendrin, cytokeratins, thyroglobulin, and thyroperoxidase showed iodine-uptake and secreted thyroglobulin and free T3. Using these conditions, we generated a bank of thyroid tumors in culture from normal thyroids, Grave's hyperplasias, benign neoplasms (goiter, adenomas), and carcinomas. CONCLUSIONS: Using appropriate culture conditions is essential for phenotype maintenance in human thyrocytes. The bank of thyroid tumors in culture generated under humanized humanized 7 homeostatic additives culture conditions will provide a much-needed tool to compare similarly growing cells from normal vs pathological origins and thus to elucidate the molecular basis of thyroid disease.


Asunto(s)
Técnicas de Cultivo de Célula , Medios de Cultivo , Glándula Tiroides/citología , Neoplasias de la Tiroides/patología , Animales , Línea Celular Tumoral , Proliferación Celular , Humanos , Fenotipo , Ratas , Tiroglobulina/metabolismo , Triyodotironina/metabolismo
10.
J Mol Endocrinol ; 49(2): R89-111, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22822049

RESUMEN

Embryonic, adult, artificially reprogrammed, and cancer…- there are various types of cells associated with stemness. Do they have something fundamental in common? Are we applying a common name to very different entities? In this review, we will revisit the characteristics that define 'pluripotency', the main property of stem cells (SCs). For each main type of physiological (embryonic and adult) or synthetic (induced pluripotent) SCs, markers and functional behavior in vitro and in vivo will be described. We will review the pioneering work that has led to obtaining human SC lines, together with the problems that have arisen, both in a biological context (DNA alterations, heterogeneity, tumors, and immunogenicity) and with regard to ethical concerns. Such problems have led to proposals for new operative procedures for growing human SCs of sufficiently high quality for use as models of disease and in human therapy. Finally, we will review the data from the first clinical trials to use various types of SCs.


Asunto(s)
Trasplante de Células Madre , Células Madre/fisiología , Adulto , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Línea Celular , Ensayos Clínicos como Asunto , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Rechazo de Injerto , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/fisiología , Ratones , Proteína Homeótica Nanog , Células Madre Pluripotentes/metabolismo , Regiones Promotoras Genéticas , Transducción de Señal , Células Madre/citología , beta Catenina/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...