Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Base de datos
Tipo del documento
Intervalo de año de publicación
1.
Oncogene ; 37(11): 1430-1444, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29321662

RESUMEN

Development of human tumors is driven by accumulation of alterations in tumor suppressor genes and oncogenes in cells. The POU1F1 transcription factor (also known Pit-1) is expressed in the mammary gland and its overexpression induces profound phenotypic changes in proteins involved in breast cancer progression. Patients with breast cancer and elevated expression of Pit-1 show a positive correlation with the occurrence of distant metastasis and poor overall survival. However, some mediators of Pit-1 actions are still unknown. Here, we show that CXCR4 chemokine receptor and its ligand CXCL12 play a critical role in the pro-tumoral process induced by Pit-1. We found that Pit-1 increases mRNA and protein in both CXCR4 and CXCL12. Knock-down of CXCR4 reduces tumor growth and spread of Pit-1 overexpressing cells in a zebrafish xenograft model. Furthermore, we described for the first time pro-angiogenic effects of Pit-1 through the CXCL12-CXCR4 axis, and that extravasation of Pit-1 overexpressing breast cancer cells is strongly reduced in CXCL12-deprived target tissues. Finally, in breast cancer patients, expression of Pit-1 in primary tumors was found to be positively correlated with CXCR4 and CXCL12, with specific metastasis in liver and lung, and with clinical outcome. Our results suggest that Pit-1-CXCL12-CXCR4 axis could be involved in chemotaxis guidance during the metastatic process, and may represent prognostic and/or therapeutic targets in breast tumors.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Quimiocina CXCL12/fisiología , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/secundario , Receptores CXCR4/fisiología , Factor de Transcripción Pit-1/fisiología , Animales , Movimiento Celular/genética , Proliferación Celular/genética , Células Cultivadas , Quimiocina CXCL12/genética , Embrión no Mamífero , Femenino , Regulación Neoplásica de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Neoplasias Hepáticas/genética , Neoplasias Pulmonares/genética , Células MCF-7 , Invasividad Neoplásica , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Receptores CXCR4/genética , Transducción de Señal/fisiología , Factor de Transcripción Pit-1/genética , Pez Cebra
2.
Pediatr Endocrinol Rev ; 12(3): 290-6, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25962206

RESUMEN

Various hypothalamic-pituitary diseases cause hypopituitarism. Inflammation related to autoimmunity also causes hypopituitarism. Hypophysitis is a representative disease caused by autoimmunity. Generally, anterior pituitary hormones are non-specifically impaired in this condition, but specific hormone defects have been reported in some cases. Anti-PIT-1 (pituitary-specific transcription factor 1) antibody syndrome is a novel clinical entity that presents an acquired combined pituitary hormone deficiency characterized by a specific defect in growth hormone, prolactin, and thyroid-stimulating hormone. Circulating anti-PIT-1 antibody along with various autoantibodies are detected with multiple endocrine organopathy, meeting the definition of autoimmune polyglandular syndrome. Mechanistically, cytotoxic T lymphocytes that specifically react with PIT-1 protein play an important role in the development of this syndrome.


Asunto(s)
Autoanticuerpos/inmunología , Hipopituitarismo/inmunología , Factor de Transcripción Pit-1/inmunología , Humanos , Hipopituitarismo/complicaciones , Hipopituitarismo/genética , Poliendocrinopatías Autoinmunes/genética , Poliendocrinopatías Autoinmunes/inmunología , Síndrome , Factor de Transcripción Pit-1/fisiología
3.
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
4.
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
5.
Aging Cell ; 11(4): 668-74, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22577861

RESUMEN

Previous work has shown that primary skin-derived fibroblasts from long-lived pituitary dwarf mutants resist the lethal effects of many forms of oxidative and nonoxidative stress. We hypothesized that increased autophagy may protect fibroblasts of Pit-1(dw/dw) (Snell dwarf) mice from multiple forms of stress. We found that dwarf-derived fibroblasts had higher levels of autophagy, using LC3 and p62 as markers, in response to amino acid deprivation, hydrogen peroxide, and paraquat. Fibroblasts from dwarf mice also showed diminished phosphorylation of mTOR, S6K, and 4EBP1, consistent with the higher levels of autophagy in these cells after stress. Similar results were also observed in fibroblasts from mutant mice lacking growth hormone receptor (GHRKO mice) after amino acid withdrawal. Our results suggested that increased autophagy, regulated by TOR-dependent processes, may contribute to stress resistance in fibroblasts from long-lived mutant mice.


Asunto(s)
Autofagia/genética , Longevidad/genética , Longevidad/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Autofagia/efectos de los fármacos , Autofagia/fisiología , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Medios de Cultivo , Factores Eucarióticos de Iniciación , Femenino , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Peróxido de Hidrógeno/toxicidad , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Ratones Mutantes , Estrés Oxidativo , Paraquat/toxicidad , Fosfoproteínas/metabolismo , Fosforilación , Receptores de Somatotropina/deficiencia , Receptores de Somatotropina/genética , Receptores de Somatotropina/fisiología , Proteínas Quinasas S6 Ribosómicas/metabolismo , Factor de Transcripción Pit-1/deficiencia , Factor de Transcripción Pit-1/genética , Factor de Transcripción Pit-1/fisiología
6.
Am J Physiol Regul Integr Comp Physiol ; 302(5): R606-19, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22160539

RESUMEN

Glucocorticoid (GC) treatment of rat or chicken embryonic pituitary (CEP) cells induces premature production of growth hormone (GH). GC induction of the GH gene requires ongoing protein synthesis, and the GH genes lack a canonical GC response element (GRE). To characterize cis-acting elements and identify trans-acting proteins involved in this process, we characterized the regulation of a luciferase reporter containing a fragment of the chicken GH gene (-1727/+48) in embryonic day 11 CEP cells. Corticosterone (Cort) increased luciferase activity and mRNA expression, and mRNA induction was blocked by protein synthesis inhibition. Through deletion analysis, we identified a GC-responsive region (GCRR) at -1045 to -954. The GCRR includes an ETS-1 binding site and a degenerate GRE (dGRE) half site. Nuclear proteins, including ETS-1, bound to a GCRR probe in electrophoretic mobility shift assays, and Cort regulated protein binding. Using chromatin immunoprecipitation, we found that ETS-1 and GC receptor (GR) were associated with the GCRR in CEP cells, and Cort increased GR recruitment to the GCRR. Mutation of the ETS-1 site or dGRE site in the -1045/+48 GH reporter abolished Cort responsiveness. We conclude that GC regulation of the GH gene during development requires cis-acting elements in the GCRR and involves ETS-1 and GR binding to these elements. Similar ETS-1 elements/dGREs are located in the 5'-flanking regions of GH genes in mammals, including rodents and humans. This is the first study to demonstrate involvement of ETS-1 in GC regulation of the GH gene during embryonic development in any species, enhancing our understanding of GH regulation in vertebrates.


Asunto(s)
Expresión Génica/efectos de los fármacos , Glucocorticoides/farmacología , Hormona del Crecimiento/genética , Hormona del Crecimiento/metabolismo , Hipófisis/metabolismo , Elementos de Respuesta/fisiología , Animales , Células Cultivadas , Embrión de Pollo , Eliminación de Gen , Expresión Génica/fisiología , Luciferasas/genética , Luciferasas/metabolismo , Modelos Animales , Mutación/genética , Hipófisis/citología , Hipófisis/embriología , Regiones Promotoras Genéticas/genética , Proteína Proto-Oncogénica c-ets-1/genética , Proteína Proto-Oncogénica c-ets-1/fisiología , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/fisiología , Elementos de Respuesta/genética , Factor de Transcripción Pit-1/genética , Factor de Transcripción Pit-1/fisiología
7.
J Dairy Sci ; 94(8): 4220-3, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21787958

RESUMEN

Early embryo loss is a major contributing factor to cow infertility and that 70 to 80% of this loss occurs between d 8 and 16 postfertilization. However, little is known about the molecular mechanisms and the nature of genes involved in normal and abnormal embryonic development. Moreover, information is limited on the contributions of the genomes of dams and of embryos to the development and survival of preimplantation embryos. We hypothesized that proper gene expression level in the developing embryo is essential for embryo survival and pregnancy success. As such, the characterization of expression profiles in early embryos could lead to a better understanding of the mechanisms involved in normal and abnormal embryo development. To test this hypothesis, 2 d-8 embryo populations (degenerate embryos and blastocysts) that differed in morphology and developmental status were investigated. Expression levels of POU1F1 pathway genes were estimated in 4 sets of biological replicate pools of degenerate embryos and blastocysts. The OPN and STAT5A genes were found to be upregulated in degenerate embryos compared with blastocysts, whereas STAT5B showed similar expression levels in both embryo groups. Analysis of splice variants of OPN and STAT5A revealed expression patterns different from the total expression values of these genes. As such, measuring expression of individual transcripts should be considered in gene expression studies.


Asunto(s)
Empalme Alternativo/genética , Blastocisto/metabolismo , Bovinos/genética , Expresión Génica/genética , Factor de Transcripción Pit-1/genética , Animales , Femenino , Fertilidad/genética , Fertilización/genética , Genes/genética , Genotipo , Embarazo , Factor de Transcripción Pit-1/fisiología
8.
Front Horm Res ; 38: 127-138, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20616503

RESUMEN

The RET receptor is a tyrosine kinase receptor implicated in kidney and neural development. In the adenopituitary RET and the co-receptor GFRa1 are expressed exclusively in the somatotrophs secreting GH. RET is implicated in a clever pathway to maintain at physiological levels the number of somatotrophs and the GH production. Thus, in absence of its ligand GDNF, RET induces apoptosis through massive expression of Pit-1 leading to p53 accumulation. In the presence of the ligand GDNF, RET activates its tyrosine kinase and promotes survival at the expense of reducing Pit-1 expression and downregulating GH. Recent data suggest that RET can also have a second role in pituitary plasticity through a second co-receptor GFRa2.


Asunto(s)
Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/fisiología , Factores Neurotróficos Derivados de la Línea Celular Glial/fisiología , Hipófisis/fisiología , Proteínas Proto-Oncogénicas c-ret/fisiología , Animales , Humanos , Regiones Promotoras Genéticas , Transducción de Señal , Factor de Transcripción Pit-1/fisiología , Proteína p53 Supresora de Tumor/fisiología
9.
Endocr Relat Cancer ; 17(1): 73-85, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19808898

RESUMEN

The transcription factor Pit-1/Pou1f1 regulates GH and prolactin (PRL) secretion in the pituitary gland. Pit-1 expression and GH regulation by Pit-1 have also been demonstrated in mammary gland. However, no data are available on the role of Pit-1 on breast PRL. To evaluate this role, several human breast cancer cell lines were transfected with either the Pit-1 expression vector or a Pit-1 small interference RNA construct, followed by PRL mRNA and protein evaluation. In addition, transient transfection of MCF-7 cells by a reporter construct containing the proximal PRL promoter, and ChIP assays were performed. Our data indicate that Pit-1 regulates mammary PRL at transcriptional level by binding to the proximal PRL promoter. We also found that Pit-1 raises cyclin D1 expression before increasing PRL levels, suggesting a PRL-independent effect of Pit-1 on cell proliferation. By using immunohistochemistry, we found a significant correlation between Pit-1 and PRL expression in 94 human breast invasive ductal carcinomas. Considering the possible role of PRL in breast cancer disorders, the function of Pit-1 in breast should be the focus of further research.


Asunto(s)
Neoplasias de la Mama/genética , Carcinoma Ductal de Mama/genética , Proteínas de Neoplasias/fisiología , Prolactina/biosíntesis , Factor de Transcripción Pit-1/fisiología , Animales , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/patología , División Celular , Línea Celular Tumoral/efectos de los fármacos , Ciclina D1/biosíntesis , Femenino , Regulación Neoplásica de la Expresión Génica , Genes bcl-1 , Humanos , Ratones , Mutagénesis Sitio-Dirigida , Células 3T3 NIH/efectos de los fármacos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Prolactina/genética , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Factor de Transcripción Pit-1/antagonistas & inhibidores , Factor de Transcripción Pit-1/genética , Transcripción Genética
10.
Genesis ; 47(12): 858-63, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19882669

RESUMEN

Accelerated vascular calcification occurs in several human diseases including diabetes and chronic kidney disease (CKD). In patients with CKD, vascular calcification is highly correlated with elevated serum phosphate levels. In vitro, elevated concentrations of phosphate induced vascular smooth muscle cell matrix mineralization, and the inorganic phosphate transporter-1 (PiT-1), was shown to be required. To determine the in vivo role of PiT-1, mouse conditional and null alleles were generated. Here we show that the conditional allele, PiT-1(flox), which has loxP sites flanking exons 3 and 4, is homozygous viable. Cre-mediated recombination resulted in a null allele that is homozygous lethal. Examination of early embryonic development revealed that the PiT-1(Deltae3,4/Deltae3,4) embryos displayed anemia, a defect in yolk sac vasculature, and arrested growth. Thus, conditional and null PiT-1 mouse alleles have been successfully generated and PiT-1 has a necessary, nonredundant role in embryonic development.


Asunto(s)
Alelos , Regulación del Desarrollo de la Expresión Génica , Mutación , Factor de Transcripción Pit-1/genética , Animales , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Endogámicos DBA , Ratones Endogámicos , Ratones Noqueados , Ratones Transgénicos , Embarazo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Factor de Transcripción Pit-1/fisiología
11.
J Mol Biol ; 390(1): 26-44, 2009 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-19427323

RESUMEN

The POU domain transcription factor Pit-1 is expressed in somatotropes, lactotropes, and thyrotropes of the anterior pituitary. Pit-1 is essential for the establishment of these lineages during development and regulates the expression of genes encoding the peptide hormones secreted by each cell type, including the growth hormone gene expressed in somatotropes. In contrast to rodent growth hormone loci, the human growth hormone (hGH) locus is regulated by a distal locus control region (LCR), which is required in cis for the proper expression of the hGH gene cluster in transgenic mice. The hGH LCR mediates a domain of histone acetylation targeted to the hGH locus that is associated with distal hGH-N activation, and the discrete determinants of this activity coincide with DNaseI hypersensitive site (HS) I of the LCR. The identification of three in vitro Pit-1 binding sites within the HS-I region suggested a model in which Pit-1 binding at HS-I initiates the chromatin modification mechanism associated with hGH LCR activity. To test this hypothesis directly and to determine whether Pit-1 expression is sufficient to confer hGH locus histone acetylation and activate hGH-N transcription from an inactive locus, we expressed Pit-1 in nonpituitary cell types. We show that Pit-1 expression established a domain of histone hyperacetylation at the LCR and hGH-N promoter in these cells similar to that observed in pituitary chromatin. This was accompanied by the activation of hGH-N transcription and an increase in intergenic and CD79b transcripts proximal to HS-I. These effects were coincident with Pit-1 occupancy at HS-I and the hGH-N promoter and were observed irrespective of the basal histone modification status of HS-I in the heterologous cell line. These findings are consistent with a role for Pit-1 as an initiating factor in hGH locus activation during somatotrope ontogeny, acting through binding sites at HS-I of the hGH LCR.


Asunto(s)
Histonas/metabolismo , Hormona de Crecimiento Humana/biosíntesis , Hormona de Crecimiento Humana/genética , Región de Control de Posición , Factor de Transcripción Pit-1/fisiología , Transcripción Genética , Acetilación , Antígenos CD79/biosíntesis , Antígenos CD79/genética , Línea Celular , Humanos , Activación Transcripcional
12.
Mol Biol Rep ; 36(6): 1371-4, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18654839

RESUMEN

POU1F1 is a positive regulator for prolactin (PRL) whose metabolites may directly or indirectly affect some aspects of the hair growth cycle, therefore, POU1F1 gene is an important candidate gene for cashmere traits selection through marker-assisted selection (MAS). Hence, in this study, the PCR-RFLP method was applied to detect a T>C transition determining a PstI polymorphism at the 3'UTR of POU1F1 locus and evaluate its associations with cashmere traits in 847 Inner Mongolia White Cashmere goats. In the analyzed population, the allelic frequencies for the T and C alleles are 0.959 and 0.041, respectively and the genotypic frequencies are in Hardy-Weinberg equilibrium (P > 0.05). Moreover, significant statistical relationships between the PstI polymorphism of POU1F1 gene and goat cashmere yields were found (*P < 0.05). When compared with TC genotype, TT genotype was associated with superior cashmere yields in 2, 4, and 5 years old individuals, as well as with average cashmere yield. Hence, TT genotype is suggested to be a molecular marker for senior cashmere yield.


Asunto(s)
Regiones no Traducidas 3'/genética , Desoxirribonucleasas de Localización Especificada Tipo II , Cabello/crecimiento & desarrollo , Polimorfismo Genético , Factor de Transcripción Pit-1/genética , Factores de Edad , Animales , Biomarcadores , Genotipo , Cabras , Polimorfismo de Nucleótido Simple , Factor de Transcripción Pit-1/fisiología
13.
J Mol Endocrinol ; 42(3): 225-37, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19103719

RESUMEN

Thyrotropin (TSH) is a heterodimer consisting of alpha and beta chains, and the beta chain (TSHbeta) is specific to TSH. The coexistence of two transcription factors, PIT1 and GATA2, is known to be essential for TSHbeta expression. Using kidney-derived CV1 cells, we investigated the role of PIT1 in the expression of Tshb gene. GATA2 Zn finger domain, which is known to recognize GATA-responsive elements (GATA-REs), is essential for cooperation by PIT1. Transactivation of TSHbeta promoter requires PIT1-binding site upstream to GATA-REs (PIT1-US), and the spacing between PIT1-US and GATA-REs strictly determines the cooperation between PIT1 and GATA2. Moreover, truncation of the sequence downstream to GATA-REs enabled GATA2 to transactivate the TSHbeta promoter without PIT1. The deleted region (nt -82/-52) designated as a suppressor region (SR) was considered to inhibit transactivation by GATA2. The cooperation of PIT1 with GATA2 was not conventional synergism but rather counteracted SR-induced suppression (derepression). The minimal sequence for SR was mapped to the 9 bp sequence downstream to GATA-REs. Electrophoretic mobility shift assay suggested that some nuclear factor exists in CV1 cells, which binds with SR and this interaction was blocked by recombinant PIT1. Our study indicates that major activator for the TSHbeta promoter is GATA2 and that PIT1 protects the function of GATA2 from the inhibition by SR-binding protein.


Asunto(s)
Factor de Transcripción GATA2/metabolismo , Regiones Promotoras Genéticas/genética , Tirotropina de Subunidad beta/genética , Factor de Transcripción Pit-1/metabolismo , Factor de Transcripción Pit-1/fisiología , Activación Transcripcional/fisiología , Animales , Sitios de Unión , Línea Celular , Chlorocebus aethiops , ADN/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Factor de Transcripción GATA2/genética , Modelos Biológicos , Unión Proteica , Factor de Transcripción Pit-1/genética , Activación Transcripcional/genética
14.
Genesis ; 46(1): 37-42, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18196598

RESUMEN

Tissue-specific expression of the Cre recombinase is a well-established genetic tool to analyze gene function in specific tissues and cell types. In this report, we describe the generation of a new transgenic line that expresses Cre under the control of the rat growth hormone releasing hormone receptor (rGhrhr) promoter. This promoter, chosen to target the anterior pituitary, drives cre-mediated recombination in cells of the Pit1 lineage, including somatotrophs, lactotrophs, and thyrotrophs. Cre activity is first detected at embryonic day 13.5, and gradually increases to reach high level expression by postnatal day 2. In addition to the pituitary, rGhrhr-cre expression was detected in vibrissae and in hair follicles of the proximal limb, but not in other tissues. The rGhrhr-cre line will be a valuable tool for the study of the development of the pituitary Pit1 lineage and for the study of tumorigenesis involving these cells.


Asunto(s)
Extremidades , Regulación del Desarrollo de la Expresión Génica , Técnicas Genéticas , Integrasas/metabolismo , Hipófisis/metabolismo , Factor de Transcripción Pit-1/fisiología , Alelos , Animales , Linaje de la Célula , Femenino , Masculino , Ratones , Hormonas Hipofisarias/metabolismo , Regiones Promotoras Genéticas , Factores de Tiempo
15.
Curr Opin Nephrol Hypertens ; 16(4): 325-8, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17565274

RESUMEN

PURPOSE OF REVIEW: Vascular calcification is associated with cardiovascular events in patients with end-stage renal disease and diabetes. Hyperphosphatemia is a risk factor for vascular calcification in these patients. Sodium-dependent phosphate cotransporters are required for cellular phosphate uptake. This review focuses on the potential role of phosphate transport and type III sodium-dependent phosphate cotransporters in the process of vascular calcification. RECENT FINDINGS: Consistent with clinical and animal studies, elevated phosphate induces mineralization of cultured smooth muscle cells in vitro. Calcification is concomitant with osteochondrogenic phenotype change in smooth muscle cells characterized by induction of osteochondrogenic differentiation marker, Runx2, and inhibition of smooth muscle cell lineage marker, SM22. Inhibition of the type III sodium-dependent phosphate cotransporter, Pit-1, blocks phosphate-induced smooth muscle cell calcification. Moreover, the phosphate-induced osteochondrogenic phenotype modulation is also abrogated by Pit-1 inhibition. Pit-1 is upregulated by several calcification-promoting factors, including tumor necrosis factor-alpha, bone morphogenetic protein 2, platelet-derived growth factor and elevated calcium. SUMMARY: Phosphate uptake via Pit-1 is required for osteochondrogenic phenotypic change and calcification of vascular smooth muscle cells in vitro. Modulation of Pit-1 expression or its transport activity may provide a novel therapeutic target for intervention of vascular calcification.


Asunto(s)
Calcinosis/etiología , Enfermedades Cardiovasculares/etiología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/fisiología , Factor de Transcripción Pit-1/fisiología , Animales , Humanos , Fallo Renal Crónico/fisiopatología , Ratones , Fosfatos/metabolismo , Ratas
16.
Mol Endocrinol ; 21(8): 1969-83, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17536003

RESUMEN

The GHRH receptor is expressed in the somatotroph cell of the anterior pituitary, where it functions to mediate GHRH-stimulated GH release. To study pituitary and somatotroph cell-specific expression of this gene, a transgenic mouse model and complementary cell culture experiments were developed. The activity of the 1.6-kb proximal rat GHRH receptor promoter was examined in vivo by generating transgenic mice with the promoter directing expression of a luciferase reporter. The promoter directs tissue-specific expression; luciferase is highly expressed in the pituitary but absent from 14 other tissues. Immunocytochemistry experiments show that transgene expression is targeted to GH-expressing somatotroph cells. The transgene is 5-fold more highly expressed in males than females, and there is an increase in transgene expression leading up to the onset of puberty. The 1.6-kb promoter was further examined in cell culture experiments, which revealed that the promoter is selectively activated in pituitary cells and that promoter-reporter expression in nonpituitary cells can be enhanced by the pituitary-specific transcription factor Pit-1. EMSAs identified 10 short regions that specifically bind Pit-1 with highly variable relative affinities. The highest affinity site was previously identified and is required for Pit-1 activation of the promoter. Four additional sites contribute to Pit-1 regulation of the promoter and are important to achieving full activation of the gene. The results show that the 1.6-kb promoter is sufficient to direct tissue- and cell-specific expression in vivo and is regulated by Pit-1.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Adenohipófisis/metabolismo , Receptores de Neuropéptido/biosíntesis , Receptores de Neuropéptido/genética , Receptores de Hormona Reguladora de Hormona Hipofisaria/biosíntesis , Receptores de Hormona Reguladora de Hormona Hipofisaria/genética , Factor de Transcripción Pit-1/fisiología , Animales , Animales Modificados Genéticamente , Genes Reporteros , Células HeLa , Humanos , Ratones , Ratones Transgénicos , Ratas , Transgenes
17.
Mol Endocrinol ; 21(1): 172-85, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17021049

RESUMEN

The POU-homeodomain transcription factor Pit-1 governs ontogeny and cell-specific gene expression of pituitary lactotropes, somatotropes, and thyrotropes. The splice isoform, Pit-1beta, inserts a 26-amino acid (AA) repressor at AA48 in the Pit-1 transcription activation domain (TAD). The Pit-1 TAD contains a basal regulatory subregion, R1 (AA1-45), and a basal and Ras-responsive region, R2 (AA46-80). To precisely map these activities, we generated GAL4-Pit-1/Pit-1betaTAD fusions and, in full-length HA-Pit-1, a series of substitution mutants of R2. Analysis in GH4 cells identified an activation domain at AA50-70, followed by an overlapping, dual-function, Ras-responsive-inhibitory domain, located from AA60-80. In contrast, GAL4-Pit-1betaTAD repressed both basal and Ras-mediated TAD activity. To determine the functional interplay between TAD subregions and the beta-domain, we inserted the beta-domain every 10 AA across the 80-AA Pit-1 TAD. Like wild-type Pit-1beta, each construct retained transcriptional activity in HeLa cells and repressed the Ras response in GH4 cells. However, beta-domain insertion at AA61 and 71 resulted in greater repression of Ras responsiveness, defining a critical R2 TAD spanning AA61-71 of Pit-1. Furthermore, Ras activation is augmented by steroid receptor coactivator 1, whereas cAMP response element binding protein-binding protein is not a Ras mediator in this system. In summary, the Pit-1/Pit-1beta TADs are composed of multiple, modular, and transferable subdomains, including a regulatory R1 domain, a basal activation region, a selective inhibitory-Ras-responsive segment, and a beta-specific repressor domain. These data provide novel insights into the mechanisms by which the Pit-1 TAD integrates DNA binding, protein partner interactions, and distinct signaling pathways to fine-tune Pit-1 activity.


Asunto(s)
Factor de Transcripción Pit-1/fisiología , Activación Transcripcional , Proteínas ras/fisiología , Animales , Sitios de Unión , Células HeLa , Humanos , Modelos Biológicos , Mutagénesis , Fosforilación , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ratas , Transducción de Señal , Proteínas ras/metabolismo
18.
Mol Endocrinol ; 20(12): 3212-27, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16901973

RESUMEN

The anterior pituitary-specific transcription factor Pit-1 was initially identified and cloned as a transactivator of the prolactin (PRL) and GH genes and later as a regulator of the TSHb gene. It was found to be a major developmental regulator, because natural Pit-1 gene mutations cause a dwarf phenotype in mice and cause combined pituitary hormone deficiency associated with pituitary hypoplasia in humans. To further investigate the growth-promoting effects of Pit-1, we used a strategy based on the use of dominant-negative Pit-1 mutants as an alternative means of inactivating endogenous Pit-1 functions. R271W, a Pit-1 mutant identified in one allele in patients with severe combined pituitary hormone deficiency, and Pit-1Delta1-123, a deletion mutant in which only the DNA binding domain of Pit-1 is conserved, were generated, and their ability to abolish the effects of the endogenous native Pit-1 in the differentiated proliferating somatolactotrope GH4C1 cell line was investigated. Enforced expression of the dominant-negative mutants in GH4C1 cells using recombinant lentiviral vectors decreased the levels of expression of known Pit-1 target genes such as PRL and GH, abolished the hormone release, and reduced cell viability by decreasing the growth rate and inducing apoptosis via a caspase-independent pathway. These results show for the first time that the growth-promoting effects of Pit-1 are at least partly due to the fact that this transcription factor prevents apoptotic cell death.


Asunto(s)
Apoptosis/genética , Enanismo Hipofisario/genética , Regulación de la Expresión Génica , Hormonas Hipofisarias/deficiencia , Factor de Transcripción Pit-1/fisiología , Muerte Celular/genética , Proliferación Celular , Células Cultivadas , Técnicas de Transferencia de Gen , Humanos , Lentivirus/genética , Mutación , Hormonas Hipofisarias/metabolismo , Factor de Transcripción Pit-1/genética , Transfección
19.
J Bone Miner Res ; 21(5): 674-83, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16734382

RESUMEN

UNLABELLED: Pi handling by osteogenic cells is important for bone mineralization. The role of Pi transport in BMP-2-induced matrix calcification was studied. BMP-2 enhances Pit-1 Pi transporters in osteogenic cells. Experimental analysis suggest that this response is required for bone matrix calcification. INTRODUCTION: Bone morphogenetic proteins (BMPs) are produced by osteogenic cells and play an important role in bone formation. Inorganic phosphate (Pi) is a fundamental constituent of hydroxyapatite, and its transport by osteogenic cells is an important function for primary calcification of the bone matrix. In this study, we investigated the role of Pi transport in BMP-2-induced matrix mineralization. MATERIALS AND METHODS: Confluent MC3T3-E1 osteoblast-like cells were exposed to BMP-2 for various time periods. Pi and alanine transport was determined using radiolabeled substrate, Pit-1 and Pit-2 expression by Northern blot analysis, cell differentiation by alkaline phosphatase activity, matrix mineralization by alizarin red staining, and the characteristics of mineral deposited in the matrix by transmission electron microscopy, electron diffraction analysis, and Fourier transformed infrared resolution (FTIR). RESULTS: BMP-2 time- and dose-dependently stimulated Na-dependent Pi transport in MC3T3-E1 cells by increasing the V(max) of the transport system. This effect was preceded by an increase in mRNA encoding Pit-1 but not Pit-2. BMP-2 also dose-dependently enhanced extracellular matrix mineralization, an effect blunted by either phosphonoformic acid or expression of antisense Pit-1. Enhanced Pi transport and matrix mineralization induced by BMP-2 were blunted by a specific inhibitor of the c-Jun-N-terminal kinase (JNK) pathway. CONCLUSIONS: Results presented in this study indicate that, in addition to its well-known effect on several markers of the differentiation of osteoblastic cells, BMP-2 also stimulates Pi transport activity through a selective increase in expression of type III Pi transporters Pit-1. In MC3T3-E1 cells, this effect is mediated by the JNK pathway and plays an essential role in bone matrix calcification induced by BMP-2.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Calcificación Fisiológica/fisiología , Osteoblastos/metabolismo , Factor de Transcripción Pit-1/fisiología , Factor de Crecimiento Transformador beta/fisiología , Células 3T3 , Animales , Secuencia de Bases , Proteína Morfogenética Ósea 2 , Cartilla de ADN , Ratones , Osteoblastos/citología
20.
Peptides ; 27(5): 1020-35, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16515822

RESUMEN

Neuropeptide FF (NPFF) is an RF-amide peptide with pleiotropic functions in the mammalian central nervous system, including pain modulation, opiate interactions, cardiovascular regulation and neuroendocrine effects. To gain insights into the transcriptional mechanisms that regulate NPFF gene expression, we cloned and sequenced 9.8 and 1.5 kb of the mouse and rat NPFF 5'-flanking region, respectively. Regions with high sequence homology between mouse, rat and human were expected to have high probability to interact with regulatory proteins and were studied further. Electromobility shift assays revealed one region that may interact with the homeobox proteins Oct-1, PDX1, Pit-1 and MEIS and two consensus DRE sites that bind a nuclear protein, which was identified as the downstream regulatory element antagonistic modulator DREAM by supershift assays. The distribution of NPFF gene expression was examined in the mouse using in situ hybridization and RT-PCR. NPFF expression was also evident during mouse embryogenesis. A fixed transcription initiation site for the mouse NPFF gene was found. A novel splice variant with a retained intron of the NPFF gene was characterized. Chimeric luciferase reporter gene constructs for the mouse NPFF gene revealed a minimal promoter region and a region with transcriptional suppressor features. An NGF responsive area was found using mouse NPFF reporter gene constructs. We postulate that Oct-1, PDX1, Pit-1, MEIS and DREAM are likely transcriptional regulators of NPFF gene expression.


Asunto(s)
Regulación de la Expresión Génica , Oligopéptidos/genética , Factores de Transcripción/genética , Región de Flanqueo 5'/genética , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Sistema Nervioso Central/metabolismo , Mapeo Cromosómico/veterinaria , Clonación Molecular , Ensayo de Cambio de Movilidad Electroforética , Proteínas de Homeodominio/fisiología , Humanos , Proteínas de Interacción con los Canales Kv/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Proteínas de Neoplasias/fisiología , Factor 1 de Transcripción de Unión a Octámeros/fisiología , Ratas , Alineación de Secuencia , Transactivadores/fisiología , Factor de Transcripción Pit-1/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA