Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
BMC Complement Altern Med ; 14: 256, 2014 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-25038801

RESUMEN

BACKGROUND: Rikkunshito is a traditional Japanese herbal medicine that is used to treat appetite loss associated with cancer and other disorders. The formulation contains various constituents that influence cell signaling, and rikkunshito may accordingly affect human homeostasis through multiple regulatory pathways, including those governed by the endocrine system. We investigated the actions of rikkunshito on catecholamine release from PC12 cells, an adrenal chromaffin cell line. METHODS: The actions of rikkunshito on PC12 cells were evaluated by measuring intracellular cAMP levels, tyrosine hydroxylase (TH) and vasoactive intestinal peptide (VIP) mRNA expression levels, and catecholamine levels in the culture medium. The transcriptional activation of VIP gene by rikkunshito was assessed by using a VIP promoter-driven reporter gene assay. RESULTS: Rikkunshito dose-dependently enhanced forskolin-induced elevations in cAMP in PC12 cells, and also increased the gene expression of TH and VIP. The transcriptional activation of VIP gene by rikkunshito was confirmed. Norepinephrine and dopamine secretion into the culture medium of PC12 cells were also dose-dependently augmented by rikkunshito and/or forskolin, but experiments with a protein kinase C (PKC) activator and a phosphodiesterase inhibitor revealed that the effects of rikkunshito were not simply due to the modulation of PKC or phosphodiesterase activity. CONCLUSIONS: These findings suggest that rikkunshito enhances the release of catecholamines by a novel mechanism involving cAMP.


Asunto(s)
Catecolaminas/metabolismo , Células Cromafines/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Animales , Células Cromafines/metabolismo , AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Células PC12 , Regiones Promotoras Genéticas/efectos de los fármacos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Transducción de Señal/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Tirosina 3-Monooxigenasa/biosíntesis , Tirosina 3-Monooxigenasa/genética , Péptido Intestinal Vasoactivo/biosíntesis , Péptido Intestinal Vasoactivo/genética
2.
Proc Natl Acad Sci U S A ; 111(23): E2404-13, 2014 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-24912192

RESUMEN

The molecular mechanisms underlying the development of pancreatic neuroendocrine tumors (PanNETs) have not been well defined. We report here that the genomic region of the PHLDA3 gene undergoes loss of heterozygosity (LOH) at a remarkably high frequency in human PanNETs, and this genetic change is correlated with disease progression and poor prognosis. We also show that the PHLDA3 locus undergoes methylation in addition to LOH, suggesting that a two-hit inactivation of the PHLDA3 gene is required for PanNET development. We demonstrate that PHLDA3 represses Akt activity and Akt-regulated biological processes in pancreatic endocrine tissues, and that PHLDA3-deficient mice develop islet hyperplasia. In addition, we show that the tumor-suppressing pathway mediated by MEN1, a well-known tumor suppressor of PanNETs, is dependent on the pathway mediated by PHLDA3, and inactivation of PHLDA3 and MEN1 cooperatively contribute to PanNET development. Collectively, these results indicate the existence of a novel PHLDA3-mediated pathway of tumor suppression that is important in the development of PanNETs.


Asunto(s)
Genes Supresores de Tumor , Pérdida de Heterocigocidad , Tumores Neuroendocrinos/genética , Proteínas Nucleares/genética , Neoplasias Pancreáticas/genética , Animales , Apoptosis/genética , Western Blotting , Línea Celular Tumoral , Células Cultivadas , Metilación de ADN , Humanos , Hiperplasia , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Masculino , Ratones , Ratones Noqueados , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/patología , Proteínas Nucleares/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Pronóstico , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Endogámicas Lew , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
3.
Surg Today ; 43(8): 894-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23052745

RESUMEN

PURPOSE: To examine the clinical characteristics and survival outcomes of patients with primary hyperparathyroidism (PHPT) in multiple endocrine neoplasia type 1 (MEN1) in relation to the MEN1 gene mutation. METHODS: The study population included the patients, positive for the MEN1 gene mutation, who underwent parathyroidectomy between 1983 and 2009 at a single tertiary referral center. Manifestations of the syndrome, other tumors and causes of death were retrospectively correlated with the specific types and locations of MEN1 gene mutations. RESULTS: Thirty-two patients from 19 families were diagnosed as having MEN1 on genetic examinations. Mutations were most common in exons 2, 7 and 10. A phenotypic analysis of the main MEN1 tumor types among the 32 patients revealed that PHPT was the most common (100 %), followed in order by pancreatic neuroendocrine tumors (PNETs) (53 %) and pituitary tumors (38 %). Death due to MEN1-related disease occurred in five patients (16 %), including malignant PNET in three cases (exons 2, 3), pituitary crisis in one case (exon 2) and thymic cancer in one case (large deletion). CONCLUSIONS: Premature deaths related to MEN1 are due to the development of malignant PNET, pituitary crisis or thymic tumors associated with mutations in exons 2, 3 and a large deletion.


Asunto(s)
Hiperparatiroidismo Secundario/genética , Hiperparatiroidismo Secundario/mortalidad , Neoplasia Endocrina Múltiple Tipo 1/complicaciones , Neoplasia Endocrina Múltiple Tipo 1/genética , Mutación , Proteínas Proto-Oncogénicas/genética , Adulto , Anciano , Exones/genética , Femenino , Genotipo , Humanos , Hiperparatiroidismo Secundario/etiología , Hiperparatiroidismo Secundario/cirugía , Masculino , Persona de Mediana Edad , Paratiroidectomía/mortalidad , Fenotipo , Estudios Retrospectivos , Tasa de Supervivencia , Resultado del Tratamiento , Adulto Joven
4.
Endocr J ; 59(12): 1093-8, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22878668

RESUMEN

Germline MEN1 mutation analysis is a powerful tool for an early diagnosis of multiple endocrine neoplasia type 1 (MEN1), an autosomal dominant familial cancer syndrome characterized by the parathyroid, pituitary and gastroenteropancreatic endocrine tumors. However, the clinical significance of MEN1 gene variants, especially missense and in-frame mutations as well as some splicing mutations, is not always obvious. We have previously shown that mutant menin proteins associated with MEN1 are rapidly degraded by the ubiquitin-proteasome pathway. We also demonstrated by a fluorescent immunocytochemical stability test that the stability of missense and in-frame deletion mutants varies widely but that unstable mutants were found only in MEN1 and related disorders and not in normal polymorphisms. In the present study, we evaluated by this stability test the pathogenicity of a novel MEN1 missense mutation, c.1118C>T, encoding a P373L mutant menin, identified in a suspected MEN1 patient. The results demonstrated that the mutant menin is highly unstable, indicating that this mutation is causative for MEN1. These findings encouraged us to proceed with presymptomatic genetic screening for this mutation among the family members, which resulted in the identification of asymptomatic mutation carriers. Thus, the information from the menin stability test was useful for genetic diagnosis and counseling of MEN1 in the case with a previously unreported MEN1 missense mutation.


Asunto(s)
Pruebas Genéticas/métodos , Neoplasia Endocrina Múltiple Tipo 1/diagnóstico , Mutación Missense , Proteínas Proto-Oncogénicas/genética , Adulto , Anciano de 80 o más Años , Análisis Mutacional de ADN/métodos , Femenino , Inestabilidad Genómica/genética , Humanos , Espacio Intracelular/genética , Masculino , Persona de Mediana Edad , Neoplasia Endocrina Múltiple Tipo 1/genética , Linaje , Valor Predictivo de las Pruebas , Adulto Joven
5.
Endocr J ; 59(6): 523-30, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22447146

RESUMEN

Heterozygous germline mutation of the tumor suppressor gene MEN1 is responsible for multiple endocrine neoplasia type 1 (MEN1), a familial cancer syndrome characterized by pituitary, parathyroid and enteropancreatic tumors. Various mutations have been identified throughout the entire gene region in patients with MEN1 and its incomplete forms often manifested as familial isolated hyperparathyroidism and apparently sporadic parathyroid tumor. Mutation analysis of the MEN1 gene is a powerful tool for the early diagnosis of MEN1; however, the clinical significance of the identified mutations is not always obvious. In this study, a previously unreported missense MEN1 mutation, c.824G>T was identified in a patient with primary hyperparathyroidism and evaluated for its pathogenicity. This mutation was predicted to generate a putative missense menin protein, R275M. A stability test of the menin protein demonstrated that the stability of R275M mutant was reduced only slightly as compared with wild type menin, and therefore could not preclude the possibility that it was a rare benign polymorphism. However, further analysis of leukocyte mRNA and minigene experiments indicated that the mutant c.824G>T allele gives rise to abnormally spliced menin mRNA, and thereby confirmed that c.824G>T mutation is causative for MEN1. Thus, leukocyte mRNA analysis has been demonstrated useful to identify a splicing mutation of the MEN1 gene.


Asunto(s)
Hiperparatiroidismo Primario/genética , Neoplasias de las Paratiroides/genética , Complicaciones Neoplásicas del Embarazo/genética , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/análisis , Adulto , Análisis Mutacional de ADN , Femenino , Mutación de Línea Germinal , Heterocigoto , Humanos , Mutación Missense , Embarazo , Estabilidad Proteica
6.
Eur J Endocrinol ; 166(3): 383-90, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22127489

RESUMEN

OBJECTIVE: Dopamine agonists normalize prolactin (PRL) levels and reduce tumour size in responsive prolactinoma. However, several cases have shown resistance to dopamine agonists upon initial treatment. Infrequently, prolactinoma initially responds, but then becomes refractory to prolonged treatment (secondary resistance). We investigated the possible mechanisms of resistance to dopamine agonists. SUBJECTS AND METHODS: Twelve cases of prolactinoma were surgically resected and classified according to the responsiveness of PRL levels and tumour size to dopamine agonists: good responders (n = 5), poor responders (n = 5), or secondary resistance (n = 2). We examined the expression of dopamine D(2) receptor (D(2)R) isoform (short: D(2)S and long: D(2)L) mRNA and protein. We investigated DNA methylation patterns in the promoter region of the DRD2 gene. RESULTS: The predominant D(2)R isoform expressed in prolactinoma was D(2)L. Levels of D(2)L mRNA were significantly lower in secondary resistance and poor responders than in good responders. Expression of D(2)R protein was variable among cases. Almost no CpG sites of the DRD2 gene promoter region were methylated. CONCLUSION: Resistance of prolactinoma to dopamine agonists is correlated with a reduction in D(2)L isoform mRNA levels. Silencing of the DRD2 gene by methylation in the promoter region is unlikely to play a role in dopamine agonist resistance in prolactinoma.


Asunto(s)
Agonistas de Dopamina/uso terapéutico , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/metabolismo , Prolactinoma/genética , Prolactinoma/metabolismo , Isoformas de ARN/genética , ARN Mensajero/genética , Receptores de Dopamina D2/genética , Adulto , Antagonistas de los Receptores de Dopamina D2 , Resistencia a Antineoplásicos/genética , Femenino , Silenciador del Gen , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Hipofisarias/tratamiento farmacológico , Prolactinoma/tratamiento farmacológico , Regiones Promotoras Genéticas/genética , Isoformas de ARN/antagonistas & inhibidores , Isoformas de ARN/biosíntesis , ARN Mensajero/antagonistas & inhibidores , Receptores de Dopamina D2/biosíntesis , Adulto Joven
7.
Cancer Sci ; 102(11): 2097-102, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21819486

RESUMEN

Germline mutations of the tumor suppressor gene MEN1 are found not only in typical multiple endocrine neoplasia type 1 (MEN1) but also in its incomplete forms such as familial isolated hyperparathyroidism (FIHP) and apparently sporadic parathyroid tumor (ASPT). No definitive genotype-phenotype correlation has been established between these clinical forms and MEN1 gene mutations. We previously demonstrated that mutant menin proteins associated with MEN1 are rapidly degraded by the ubiquitin-proteasome pathway. To examine whether the intracellular stability of mutant menin is correlated with clinical phenotypes, we developed a method of evaluating menin stability and examined 20 mutants associated with typical MEN1 (17 missense, two in-frame deletion, one nonsense) and 21 mutants associated with FIHP or ASPT (19 missense, two in-frame deletion). All tested mutants associated with typical MEN1 showed reduced stability. Some missense and in-frame deletion mutants (G28A, R171W, T197I, E255K, E274A, Y353del and E366D) associated with FIHP or ASPT were almost as stable as or only slightly less stable than wild-type menin, while others were as unstable as those associated with typical MEN1. Some stable mutants exhibited substantial biological activities when tested by JunD-dependent transactivation assay. These findings suggest that certain missense and in-frame mutations are fairly stable and retain intrinsic biological activity, and might be specifically associated with incomplete clinical phenotypes. The menin stability test will provide useful information for the management of patients carrying germline MEN1 mutations especially when they have missense or in-frame variants of ambiguous clinical significance.


Asunto(s)
Hiperparatiroidismo Primario/genética , Neoplasia Endocrina Múltiple Tipo 1/genética , Mutación , Neoplasias de las Paratiroides/genética , Proteínas Proto-Oncogénicas/genética , Sustitución de Aminoácidos , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Codón sin Sentido , Heterogeneidad Genética , Genotipo , Mutación de Línea Germinal , Humanos , Mutación Missense , Fenotipo , Mutación Puntual , Estabilidad Proteica , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas c-jun/fisiología , Proteínas Recombinantes de Fusión/biosíntesis , Eliminación de Secuencia , Activación Transcripcional
9.
Endocr J ; 57(9): 825-32, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20616437

RESUMEN

Menin is lost by the sequential inactivation of both MEN1 alleles in subsets of non-hereditary endocrine tumors as well as those associated with multiple endocrine neoplasia type 1 (MEN1), an autosomal dominant hereditary cancer syndrome characterized by multiple tumors including parathyroid, pituitary and enteropancreatic endocrine tumors. Loss of menin has been reported to be associated with lowered caspase 8 expression and resistance to apoptosis in murine fibroblasts and in pancreatic islet tumors arising in heterozygous MEN1 gene knockout mice, the animal model of the human MEN1 syndrome. We confirmed by menin-knockdown experiments with specific siRNA that menin is crucial for caspase 8 expression in human culture cells while overexpression of menin did not increase caspase 8 protein over basal levels. We then examined expression of menin, caspase 8 and cyclin-dependent kinase inhibitors p27(Kip1) and p15(Ink4b) by Western blotting in human parathyroid tumors surgically resected from patients with MEN1 and those with non-hereditary primary hyperparathyroidism. The menin and p27(Kip1) expression levels were correlated with MEN1 mutation status that was confirmed by DNA analysis. The caspase 8 and p15(Ink4b) protein levels were variable among tumors, and were not correlated with menin protein levels. These findings suggest that human endocrine tumors lacking menin may not always exhibit lowered caspase 8 expression and hence may not be resistant to apoptosis-inducing therapy.


Asunto(s)
Caspasa 8/biosíntesis , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Neoplasias de las Paratiroides/fisiopatología , Proteínas Proto-Oncogénicas/biosíntesis , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/biosíntesis , Células HEK293 , Humanos , Neoplasia Endocrina Múltiple Tipo 1/genética , Proteínas Proto-Oncogénicas/genética
10.
Immunity ; 31(4): 609-20, 2009 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-19800266

RESUMEN

Naturally arising regulatory T (Treg) cells express the transcription factor FoxP3, which critically controls the development and function of Treg cells. FoxP3 interacts with another transcription factor Runx1 (also known as AML1). Here, we showed that Treg cell-specific deficiency of Cbfbeta, a cofactor for all Runx proteins, or that of Runx1, but not Runx3, induced lymphoproliferation, autoimmune disease, and hyperproduction of IgE. Cbfb-deleted Treg cells exhibited impaired suppressive function in vitro and in vivo, with altered gene expression profiles including attenuated expression of FoxP3 and high expression of interleukin-4. The Runx complex bound to more than 3000 gene loci in Treg cells, including the Foxp3 regulatory regions and the Il4 silencer. In addition, knockdown of RUNX1 showed that RUNX1 is required for the optimal regulation of FoxP3 expression in human T cells. Taken together, our results indicate that the Runx1-Cbfbeta heterodimer is indispensable for in vivo Treg cell function, in particular, suppressive activity and optimal expression of FoxP3.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Subunidad beta del Factor de Unión al Sitio Principal/metabolismo , Factores de Transcripción Forkhead/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Enfermedades Autoinmunes/metabolismo , Colon/inmunología , Colon/patología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/inmunología , Subunidad beta del Factor de Unión al Sitio Principal/genética , Subunidad beta del Factor de Unión al Sitio Principal/inmunología , Factores de Transcripción Forkhead/inmunología , Expresión Génica/genética , Expresión Génica/inmunología , Perfilación de la Expresión Génica , Inmunoglobulina E/biosíntesis , Inmunoglobulina E/inmunología , Interleucina-4/biosíntesis , Interleucina-4/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Estómago/inmunología , Estómago/patología , Linfocitos T Reguladores/metabolismo
11.
World J Surg ; 33(11): 2343-54, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19350316

RESUMEN

BACKGROUND: The goal of the present study was to make our medical practice evidence-based for patients with parathyroid carcinoma. METHODS: We posed six clinical questions relevant to the management of parathyroid cancer. A comprehensive search and critical appraisal of the literature was then carried out. RESULTS: Most of the literature retrieved was retrospective in design and differed in the definition of carcinoma. The distinction between unequivocal and equivocal carcinoma (or atypical adenoma) was not always made for the study populations. None of the studies indicated reproducibility of outcome measures. Of the histopathological features described in the literature based on the description of Schantz and Castleman, capsular/vascular invasions and trabecular growth pattern were the most specific, and fibrous bands were the most sensitive. None of the patients with "atypical adenoma" developed recurrence, whereas 25% of those with "equivocal carcinoma" did. Mutations in HRPT2, the gene responsible for hereditary hyperparathyroidism with jaw-tumor syndrome, were strongly associated with sporadic parathyroid carcinoma. Severe hypercalcemia and its related clinical symptoms, extremely high levels of parathyroid hormone, osteitis fibrosa cystica, a palpable neck mass, and a relatively large depth-width ratio on ultrasonography, are the important features of parathyroid carcinoma. Disease-specific survival rates reported in the literature were varied, reflecting the differences in the definitions of carcinoma, study populations, and interventions. CONCLUSIONS: To establish valid evidence for patient management in the future, a collaboration of endocrine specialists is essential to conduct well-designed clinical studies for this rare disease.


Asunto(s)
Recurrencia Local de Neoplasia/terapia , Neoplasias de las Paratiroides/diagnóstico , Neoplasias de las Paratiroides/terapia , Proteínas Supresoras de Tumor/genética , Adenoma , Humanos , Mutación , Oportunidad Relativa , Neoplasias de las Paratiroides/etiología , Neoplasias de las Paratiroides/mortalidad , Pronóstico , Factores de Riesgo , Sensibilidad y Especificidad , Análisis de Supervivencia
12.
Cancer Sci ; 100(2): 209-15, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19068082

RESUMEN

Heterozygous germline mutations of the tumor-suppressor gene MEN1 are responsible for multiple endocrine neoplasia type 1 (MEN1), a dominantly inherited familial cancer syndrome characterized by pituitary, parathyroid, and enteropancreatic tumors. Various mutations have been identified throughout the entire gene region in patients with MEN1 and related disorders. Neither mutation hot spot nor phenotype­genotype correlation has been established in MEN1 although some missense mutations may be specifically associated with a phenotype of familial isolated hyperparathyroidism. The gene product menin has been implicated in multiple roles, including gene transcription, maintenance of genomic integrity, and control of cell division and differentiation. These multiple functions are likely to be conferred by association with multiple protein factors. Occurrence of MEN1-causing missense mutations throughout menin also suggests the requirement of multiple binding factors for its full tumor-suppressive activity. The effect of menin depletion is highly tissue specific, but its underlying mechanism remains to be elucidated. A DNA test for MEN1 germline mutations is a useful tool for diagnosis of MEN1 although it needs further improvements


Asunto(s)
Neoplasia Endocrina Múltiple Tipo 1/genética , Mutación/genética , Proteínas Proto-Oncogénicas/genética , Humanos , Neoplasia Endocrina Múltiple Tipo 1/patología , Fenotipo
13.
J Cell Biochem ; 105(3): 785-800, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18680143

RESUMEN

In extraskeletal myxoid chondrosarcoma, a chromosomal translocation creates a gene fusion between EWS and an orphan nuclear receptor, NOR1. The resulting fusion protein EWS/NOR1 has been believed to lead to malignant transformation by functioning as a transactivator for NOR1-target genes. By comparing the gene expression profiles of NOR1- and EWS/NOR1-overexpressing cells, we found that they largely shared up-regulated genes, but no significant correlation was observed with respect to the transactivation levels of each gene. In addition, the proteins associated with NOR1 and EWS/NOR1 were mostly the same in these cells. The results suggest that these proteins differentially transactivate overlapping target genes through a similar transcriptional machinery. To clarify the mechanisms underlying the transcriptional divergence between NOR1 and EWS/NOR1, we searched for alternatively associated proteins, and identified poly(ADP-ribose) polymerase I (PARP-1) as an NOR1-specific binding protein. Consistent with its binding properties, PARP-1 acted as a transcriptional repressor of NOR1, but not EWS/NOR1, in a luciferase reporter assay employing PARP-1(-/-) fibroblasts. Interestingly, suppressive activity of PARP-1 was observed in a DNA response element-specific manner, and in a subtype-specific manner toward the NR4A family (Nur77, Nurr1, and NOR1), suggesting that PARP-1 plays a role in the diversity of transcriptional regulation mediated by the NR4A family in normal cells. Altogether, our findings suggest that NOR1 and EWS/NOR1 regulate overlapping target genes differently by utilizing associated proteins, including PARP-1; and that EWS/NOR1 may acquire oncogenic activities by avoiding (or gaining) transcription factor-specific modulation by the associated proteins.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Receptores de Esteroides/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Activación Transcripcional/genética , Animales , Células COS , Chlorocebus aethiops , Proteínas de Unión al ADN/genética , Células HeLa , Humanos , Ratones , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Proteínas de Fusión Oncogénica/genética , Poli(ADP-Ribosa) Polimerasa-1 , Proteína EWS de Unión a ARN/genética , Proteína EWS de Unión a ARN/metabolismo , Receptores de Esteroides/genética , Receptores de Hormona Tiroidea/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética
14.
Nature ; 446(7136): 685-9, 2007 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-17377532

RESUMEN

Naturally arising CD25+CD4+ regulatory T cells (T(R) cells) are engaged in the maintenance of immunological self-tolerance and immune homeostasis by suppressing aberrant or excessive immune responses, such as autoimmune disease and allergy. T(R) cells specifically express the transcription factor Foxp3, a key regulator of T(R)-cell development and function. Ectopic expression of Foxp3 in conventional T cells is indeed sufficient to confer suppressive activity, repress the production of cytokines such as interleukin-2 (IL-2) and interferon-gamma (IFN-gamma), and upregulate T(R)-cell-associated molecules such as CD25, cytotoxic T-lymphocyte-associated antigen-4, and glucocorticoid-induced TNF-receptor-family-related protein. However, the method by which Foxp3 controls these molecular events has yet to be explained. Here we show that the transcription factor AML1 (acute myeloid leukaemia 1)/Runx1 (Runt-related transcription factor 1), which is crucially required for normal haematopoiesis including thymic T-cell development, activates IL-2 and IFN-gamma gene expression in conventional CD4+ T cells through binding to their respective promoters. In natural T(R) cells, Foxp3 interacts physically with AML1. Several lines of evidence support a model in which the interaction suppresses IL-2 and IFN-gamma production, upregulates T(R)-cell-associated molecules, and exerts suppressive activity. This transcriptional control of T(R)-cell function by an interaction between Foxp3 and AML1 can be exploited to control physiological and pathological T-cell-mediated immune responses.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Factores de Transcripción Forkhead/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Subunidad alfa 2 del Factor de Unión al Sitio Principal/antagonistas & inhibidores , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica , Humanos , Interleucina-2/genética , Interleucina-2/metabolismo , Activación de Linfocitos , Ratones , Fenotipo , Regiones Promotoras Genéticas/genética , Unión Proteica
16.
Jpn J Clin Oncol ; 36(11): 739-44, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17000701

RESUMEN

Multiple endocrine neoplasia type 1 is an autosomal dominant cancer syndrome characterized by pituitary, parathyroid and enteropancreatic endocrine tumors, which is caused by germline mutations of the tumor suppressor gene MEN1. In the case reported here, the patient had family with this disease whose germline MEN1 mutation was undetectable by conventional sequencing analysis. Further investigations involving polymorphism analyses, gene dose assay and nucleotide sequencing identified a large germline deletion of approximately 29 kilobase pairs spanning the whole MEN1 gene. The deletion was flanked by Alu repetitive sequences, suggesting unequal homologous recombination as the deletion mechanism. The polymorphism linkage data suggested that an asymptomatic son of the proband did not carry the family mutation. More direct evidence was obtained by gene dose assay and deletion-specific polymerase chain reaction, which demonstrated the normal MEN1 gene dosage and the absence of the deletion breakpoints in this asymptomatic subject and thus definitely excluded the possibility of disease predisposition.


Asunto(s)
Elementos Alu/genética , Mutación de Línea Germinal , Neoplasia Endocrina Múltiple Tipo 1/genética , Eliminación de Gen , Dosificación de Gen , Genes Supresores de Tumor , Humanos , Masculino , Repeticiones de Microsatélite , Persona de Mediana Edad , Linaje , Polimorfismo Genético
17.
Endocrine ; 29(3): 485-90, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16943588

RESUMEN

In some patients with multiple endocrine neoplasia type 1 (MEN1) it is not possible to identify a germline mutation in the MEN1 gene. We sought to document the loss of expression and function of the MEN1 gene product, menin, in the tumors of such a patient. The proband is an elderly female patient with primary hyperparathyroidism, pancreatic islet tumor, and breast cancer. Her son has primary hyperparathyroidism. No germline MEN1 mutation was identified in the proband or her son. However, loss of heterozygosity at the MEN1 locus and complete lack of menin expression were demonstrated in the proband's tumor tissue. The proband's cultured parathyroid cells lacked the normal reduction in proliferation and parathyroid hormone secretion in response to transforming growth factor- beta. This assessment provided insight into the molecular pathogenesis of the patient and provides evidence for a critical requirement for menin in the antiproliferative action of transforming growth factor-beta.


Asunto(s)
Cromosomas Humanos Par 11 , Mutación de Línea Germinal/fisiología , Pérdida de Heterocigocidad , Neoplasia Endocrina Múltiple Tipo 1/genética , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Anciano , Femenino , Humanos , Hipercalcemia/etiología , Hipertiroidismo/diagnóstico , Hipertiroidismo/genética , Neoplasias Hepáticas/diagnóstico , Repeticiones de Microsatélite , Neoplasia Endocrina Múltiple Tipo 1/patología , Neoplasia Endocrina Múltiple Tipo 1/secundario , Hormona Paratiroidea/metabolismo , Neoplasias de las Paratiroides/metabolismo , Neoplasias de las Paratiroides/patología , Polimorfismo Genético , Proteínas Proto-Oncogénicas/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Células Tumorales Cultivadas
18.
Int J Oncol ; 29(2): 413-21, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16820884

RESUMEN

Familial adenomatous polyposis (FAP) is an autosomal dominant familial cancer syndrome caused by germline mutations of the tumor suppressor adenomatous polyposis coli (APC) gene. Heterozygous apc mutations have been identified in the majority of classical FAP patients who develop more than 100 colorectal adenomas. However, classical FAP patients often fail to display germline APC mutations detectable by routine mutation analysis. These apparently mutation-negative cases may be caused by heterozygous large genomic deletions. In the present study, FAP patients who showed no APC germline mutation detectable by the protein truncation assay and direct sequencing of protein coding exons were screened for APC gene deletion by a gene dose assay based on double competitive polymerase chain reaction. Gene dosage measurements within exon 15 of the APC gene identified two patients with gene deletion and one with possible gene duplication among 41 apparently mutation-negative patients. The deleted sequences in the two patients were determined by fine gene dose mapping around the APC gene and nucleotide sequencing of the deletion breakpoints. They were approximately 435-kilobase pair (kb) and 737-kb regions including the whole APC gene and flanked by a 4-base pair repeat and LINE-1 repetitive sequences, respectively. The chimeric LINE-1 element created at the breakpoint in the latter case also contained a short sequence derived from another LINE-1 element, suggesting a complex unequal homologous recombination event. These findings indicate that this gene dose assay is a useful technique to detect large gene deletions of the APC gene and to determine their genomic breakpoints.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/biosíntesis , Poliposis Adenomatosa del Colon/genética , Genes APC , Predisposición Genética a la Enfermedad , Secuencia de Bases , Exones , Eliminación de Gen , Heterocigoto , Humanos , Repeticiones de Microsatélite , Modelos Genéticos , Datos de Secuencia Molecular , Mutación , Recombinación Genética
20.
Biol Pharm Bull ; 28(9): 1603-7, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16141523

RESUMEN

Within the nuclear receptor superfamily, Nur77, Nurr1, and NOR1 constitute the nuclear receptor subfamily 4 group A. Modulation of NOR1 function would be therapeutic potential for diseases related to dysfunction of NOR1, including extraskeletal myxoid chondrosarcoma and autoimmune diseases. By screening arachidonate metabolites for their capacity of transcriptional activation, we have identified prostaglandin (PG) A2 as a transactivator for NOR1. PGA2 acted as a potent activator of NOR1-dependent transcription through the GAL4-based reporter system. The putative ligand-binding domain (LBD) of the receptor directly bound PGA2, and LBD-deleted receptor showed little transcriptional activation by PGA2. Primary cultured spleen cells derived from transgenic mice overexpressing NOR1, showed higher sensitivity to PGA2 compared to those from wild-type mice. These observations suggest that PGA2 can serve as a transactivator of NOR1, and thus suggest a possibility of pharmacological modulation of the NOR1 pathways by PGA2-related compounds.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Prostaglandinas A/fisiología , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Células Cultivadas , Proteínas de Unión al ADN/genética , Genes Reporteros , Glutatión Transferasa/metabolismo , Células Híbridas , Luciferasas/genética , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Receptores de Esteroides , Receptores de Hormona Tiroidea , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/citología , Bazo/metabolismo , Resonancia por Plasmón de Superficie , Activación Transcripcional , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA