Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
FASEB J ; 37(5): e22913, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37078545

RESUMEN

The Multiple Endocrine Neoplasia I (MEN1) locus encodes the protein MENIN, which functions as a tumor suppressor protein in neuroendocrine tissues. Gastrinomas are neuroendocrine neoplasms that overproduce the hormone gastrin and can arise sporadically or as part of the MEN1 syndrome, in which mutations in the MEN1 gene lead to loss or inactivation of MENIN protein. Gastrin is a peptide hormone that is primarily synthesized in the gastric antrum and stimulates the secretion of histamine from enterochromaffin-like (ECL) cells and subsequently acid from parietal cells in the gastric corpus. In addition, gastrin exerts a mitogenic function primarily on ECL cells and progenitor cells in the gastric isthmus. Current studies seek to understand how MEN1 mutations generate a mutant MENIN protein that abrogates its tumor suppressor function. Mutations in the MEN1 gene are broadly distributed throughout its nine protein-coding exons, making it difficult to correlate protein structure with its function. Although disruption of the Men1 locus in mice causes functional neuroendocrine tumors in the pituitary and pancreas, gastrinomas do not develop in these transgenic animal models. Prior studies of human gastrinomas suggest that tissue-specific microenvironmental cues in the submucosal foregut may contribute to tumorigenesis by reprogramming of epithelial cells toward the neuroendocrine phenotype. Accordingly, recent studies suggest that neural crest-derived cells are also sensitive to reprogramming when MEN1 is deleted or mutated. Thus, the goal of this report is to review our current understanding of how MENIN modulates gastrin gene expression while highlighting its role in the prevention/suppression of neuroendocrine cell transformation.


Asunto(s)
Gastrinoma , Neoplasia Endocrina Múltiple Tipo 1 , Neoplasias Pancreáticas , Humanos , Animales , Ratones , Gastrinoma/genética , Gastrinoma/patología , Gastrinas/genética , Gastrinas/metabolismo , Neoplasia Endocrina Múltiple Tipo 1/genética , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Factores de Transcripción/genética , Neoplasias Pancreáticas/patología , Expresión Génica , Proteínas Proto-Oncogénicas/genética
2.
BMC Endocr Disord ; 22(1): 64, 2022 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-35287658

RESUMEN

BACKGROUND: MEN1 mutations can inactivate or disrupt menin function and are leading to multiple endocrine neoplasia type 1, a rare heritable tumor syndrome. CASE PRESENTATION: We report on a MEN1 family with a novel heterozygous germline mutation, c.674delG; p.Gly225Aspfs*56 in exon 4 of the MEN1 gene. Diagnosis and clinical phenotyping of MEN1 was established by laboratory tests, ultrasound, biopsy, MRI imaging and endosonography. The clinical course of the disease was followed in the index patient and her family members for eight years. The mutation was associated with distinct clinical phenotypes in the index patient and three family members harboring p.Gly225Aspfs*56. Family members affected showed primary hyperparathyroidism but variable patterns of associated endocrine tumors, adrenal cortical adenomas, prolactinoma, multifocal pancreatic neuroendocrine tumors, insulinoma and nonsecretory neuroendocrine tumors of the pancreas. The mutation c.674delG; p.Gly225Aspfs*56 leads to a frameshift from codon 225 with early truncation of the menin protein. In silico analysis predicts loss of multiple protein-menin interactions in p.Gly225Aspfs*56, potentially rendering menin insufficient to control cell division and replication. However, no aggressive neuroendocrine tumors were observed in the follow-up of this family. CONCLUSIONS: We report a novel heterozygous MEN1 frameshift mutation, potentially causing (at least partial) inactivation of menin tumor suppression potential but lacking a genotype-phenotype correlation. Our study highlights the importance of personalized care with appropriate testing and counseling in MEN1 families.


Asunto(s)
Neoplasia Endocrina Múltiple Tipo 1 , Proteínas Proto-Oncogénicas/genética , Mutación del Sistema de Lectura , Humanos , Neoplasia Endocrina Múltiple Tipo 1/genética , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Neoplasia Endocrina Múltiple Tipo 1/patología , Linaje , Fenotipo
3.
Int J Mol Sci ; 23(10)2022 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-35628193

RESUMEN

Pulmonary fibrosis therapy is limited by the unclear mechanism of its pathogenesis. C57BL/6 mice were used to construct the pulmonary fibrosis model in this study. The results showed that Men1, which encodes menin protein, was significantly downregulated in bleomycin (BLM)-induced pulmonary fibrosis. Mice were made to overexpress or had Men1 knockdown with adeno-associated virus (AAV) infection and then induced with pulmonary fibrosis. BLM-induced pulmonary fibrosis was attenuated by Men1 overexpression and exacerbated by Men1 knockdown. Further analysis revealed the distinct roles of Men1 in fibroblasts and macrophages. Men1 inhibited fibroblast activation and extracellular matrix (ECM) protein expression while promoting macrophages to be profibrotic (M2) phenotype and enhancing their migration. Accordingly, pyroptosis was potentiated by Men1 in mouse peritoneal macrophages (PMCs) and lung tissues upon BLM stimulation. Furthermore, the expression of profibrotic factor OPN was positively regulated by menin in Raw264.7 cells and lung tissues by binding to the OPN promoter region. Taken together, although Men1 showed antifibrotic properties in BLM-induced pulmonary fibrosis mice, conflictive roles of Men1 were displayed in fibroblasts and macrophages. The profibrotic role of Men1 in macrophages may occur via the regulation of macrophage pyroptosis and OPN expression. This study extends the current pathogenic understanding of pulmonary fibrosis.


Asunto(s)
Neoplasia Endocrina Múltiple Tipo 1 , Proteínas Proto-Oncogénicas , Fibrosis Pulmonar , Animales , Bleomicina/toxicidad , Fibroblastos/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/metabolismo
4.
Horm Metab Res ; 53(4): 245-256, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33853118

RESUMEN

Multiple Endocrine Neoplasia type-1 (MEN1) is an autosomal dominant disorder with a combined occurrence of tumours of parathyroid glands, pancreatic islets, and anterior pituitary. About 90% of these patients carry mutations in the MEN1 gene, though the spectrum is not well defined in India. Forty clinically suspected cases of MEN1 were enrolled prospectively over six years; 32 patients (23 index-cases and nine affected relatives) with≥2 classical endocrine tumours of MEN1 were considered definite, and eight were categorised as 'MEN1-like'. Details of their clinical presentation, treatment and mutational analysis including MEN1 gene, 3' and 5' untranslated regions (UTR) of MEN1, CDKN1B, and CaSR genes were collated. Asymptomatic first-degree relatives were also screened. Among the 32 definite MEN1 patients, all had primary hyperparathyroidism, 22 (68.7%) had gastroentero-pancreatic neuroendocrine tumours, and 21 (66%) had pituitary adenoma. Of the 23 definite index-cases, 13 (56.5%) carried mutations in the MEN1 gene. Five of nine affected first-degree relatives (55.5%), and four of 10 asymptomatic relatives (40%) also had MEN1 mutations. Seven of 10 MEN1 mutation-negative definite index-cases harboured p.V109G polymorphism in the CDKN1B gene. All eight MEN1-like cases were negative for mutations and large deletions in MEN1, mutations in 3' and 5' UTR of MEN1, CaSR and CDKN1B genes. The study has helped to clearly document the pattern of mutations among Indian MEN1 patients. However, the absence of MEN1 mutation in ~44% of cases and the presence of p.V109G polymorphism in CDKN1B gene raise the question whether such polymorphisms could independently contribute to pathogenesis.


Asunto(s)
Neoplasia Endocrina Múltiple Tipo 1/genética , Mutación , Proteínas Proto-Oncogénicas/genética , Adolescente , Adulto , Anciano , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Análisis Mutacional de ADN , Femenino , Humanos , India , Masculino , Persona de Mediana Edad , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Linaje , Estudios Prospectivos , Proteínas Proto-Oncogénicas/metabolismo , Receptores Sensibles al Calcio/genética , Regiones no Traducidas , Adulto Joven
5.
Int J Mol Sci ; 22(21)2021 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-34769484

RESUMEN

Multiple endocrine neoplasia type 1 (MEN1) is a rare tumor syndrome that manifests differently among various patients. Despite the mutations in the MEN1 gene that commonly predispose tumor development, there are no obvious phenotype-genotype correlations. The existing animal and in vitro models do not allow for studies of the molecular genetics of the disease in a human-specific context. We aimed to create a new human cell-based model, which would consider the variability in genetic or environmental factors that cause the complexity of MEN1 syndrome. Here, we generated patient-specific induced pluripotent stem cell lines carrying the mutation c.1252G>T, D418Y in the MEN1 gene. To reduce the genetically determined variability of the existing cellular models, we created an isogenic cell system by modifying the target allele through CRISPR/Cas9 editing with great specificity and efficiency. The high potential of these cell lines to differentiate into the endodermal lineage in defined conditions ensures the next steps in the development of more specialized cells that are commonly affected in MEN1 patients, such as parathyroid or pancreatic islet cells. We anticipate that this isogenic system will be broadly useful to comprehensively study MEN1 gene function across different contexts, including in vitro modeling of MEN1 syndrome.


Asunto(s)
Fibroblastos/metabolismo , Edición Génica/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Neoplasia Endocrina Múltiple Tipo 1/patología , Mutación , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Adulto , Sistemas CRISPR-Cas , Células Cultivadas , Femenino , Humanos , Neoplasia Endocrina Múltiple Tipo 1/genética , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Proteínas Proto-Oncogénicas/genética
6.
Int J Mol Sci ; 22(14)2021 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-34298972

RESUMEN

Multiple endocrine neoplasia type 1 (MEN1) is a rare autosomal dominant inherited multiple cancer syndrome of neuroendocrine tissues. Tumors are caused by an inherited germinal heterozygote inactivating mutation of the MEN1 tumor suppressor gene, followed by a somatic loss of heterozygosity (LOH) of the MEN1 gene in target neuroendocrine cells, mainly at parathyroids, pancreas islets, and anterior pituitary. Over 1500 different germline and somatic mutations of the MEN1 gene have been identified, but the syndrome is completely missing a direct genotype-phenotype correlation, thus supporting the hypothesis that exogenous and endogenous factors, other than MEN1 specific mutation, are involved in MEN1 tumorigenesis and definition of individual clinical phenotype. Epigenetic factors, such as microRNAs (miRNAs), are strongly suspected to have a role in MEN1 tumor initiation and development. Recently, a direct autoregulatory network between miR-24, MEN1 mRNA, and menin was demonstrated in parathyroids and endocrine pancreas, showing a miR-24-induced silencing of menin expression that could have a key role in initiation of tumors in MEN1-target neuroendocrine cells. Here, we review the current knowledge on the post-transcriptional regulation of MEN1 and menin expression by miR-24, and its possible direct role in MEN1 syndrome, describing the possibility and the potential approaches to target and silence this miRNA, to permit the correct expression of the wild type menin, and thereby prevent the development of cancers in the target tissues.


Asunto(s)
Terapia Genética , MicroARNs/genética , Terapia Molecular Dirigida , Neoplasia Endocrina Múltiple Tipo 1/genética , Regiones no Traducidas 3' , Animales , Antagomirs/farmacología , Antagomirs/uso terapéutico , Cromosomas Humanos Par 19/genética , Cromosomas Humanos Par 9/genética , Daño del ADN , Retroalimentación Fisiológica , Predicción , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Neoplasia Endocrina Múltiple Tipo 1/terapia , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Isoformas de Proteínas/genética , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/genética , ARN Neoplásico/antagonistas & inhibidores , ARN Neoplásico/genética , Ratas
7.
Int J Mol Sci ; 21(20)2020 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-33066578

RESUMEN

Multiple endocrine neoplasia type 1 (MEN1) is a rare inherited tumor syndrome, characterized by the development of multiple neuroendocrine tumors (NETs) in a single patient. Major manifestations include primary hyperparathyroidism, gastro-entero-pancreatic neuroendocrine tumors, and pituitary adenomas. In addition to these main NETs, various combinations of more than 20 endocrine and non-endocrine tumors have been described in MEN1 patients. Despite advances in diagnostic techniques and treatment options, which are generally similar to those of sporadic tumors, patients with MEN1 have a poor life expectancy, and the need for targeted therapies is strongly felt. MEN1 is caused by germline heterozygous inactivating mutations of the MEN1 gene, which encodes menin, a tumor suppressor protein. The lack of a direct genotype-phenotype correlation does not permit the determination of the exact clinical course of the syndrome. One of the possible causes of this lack of association could be ascribed to epigenetic factors, including microRNAs (miRNAs), single-stranded non-coding small RNAs that negatively regulate post-transcriptional gene expression. Some miRNAs, and their deregulation, have been associated with MEN1 tumorigenesis. Recently, an extracellular class of miRNAs has also been identified (c-miRNAs); variations in their levels showed association with various human diseases, including tumors. The aim of this review is to provide a general overview on the involvement of miRNAs in MEN1 tumor development, to be used as possible targets for novel molecular therapies. The potential role of c-miRNAs as future non-invasive diagnostic and prognostic biomarkers of MEN1 will be discussed as well.


Asunto(s)
Biomarcadores de Tumor/genética , MicroARN Circulante/genética , Neoplasia Endocrina Múltiple Tipo 1/genética , Animales , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/metabolismo , MicroARN Circulante/sangre , MicroARN Circulante/metabolismo , Humanos , Neoplasia Endocrina Múltiple Tipo 1/diagnóstico , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Neoplasia Endocrina Múltiple Tipo 1/terapia , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo
8.
Clin Endocrinol (Oxf) ; 91(5): 633-638, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31348545

RESUMEN

CONTEXT: Multiple endocrine neoplasia 1 (MEN 1) is an autosomal dominant disease presenting as hyperplasia and neoplasia of parathyroid, pituitary and enteropancreatic tissues. Over 90% of gene carriers develop phenotypic disease by age 30 years, potentially with onset of asymptomatic disease during childhood and adolescence. OBJECTIVE: To describe the paediatric and young adult manifestations of MEN 1. DESIGN: Descriptive retrospective study of 180 patients with a common MEN1 genotype. The paediatric and young adult (age <22 years) manifestations were determined using hospital records and disease surveillance data. RESULTS: Primary hyperparathyroidism (PHPT) was identified in 42 patients (mean age 17.2 ± 3.3 years). Parathyroidectomy was performed in 16 (38.1%; mean age 17.8 ± 3.2). Four patients experienced recurrent PHPT (25%), and six (37.5%) developed permanent hypoparathyroidism. Pituitary disease was identified in 13 patients. Prolactinoma was found in nine patients (mean age 16.6 ± 2.6 years) of whom four (44.4%) had macroprolactinoma. Two patients required surgical intervention; dopamine agonists showed efficacy in six patients. Two patients with Cushing's disease were successfully treated surgically. Three patients with nonfunctioning pituitary microadenoma managed conservatively. Pancreatic neuroendocrine neoplasms (pNENs) were diagnosed in 12 patients (mean age 17.0 ± 2.6 years): three patients with insulinoma successfully resected (two resected and one exhibiting perineural invasion) and nine patients with nonfunctioning adenomas (NFAs). CONCLUSION: Pituitary adenomas, PHPT and pNENs are encountered in the paediatric and young adult MEN 1 population. Successful outcomes are typically achieved using standard medical and surgical paradigms; however, parathyroidectomy was associated with a substantial complication rate.


Asunto(s)
Neoplasia Endocrina Múltiple Tipo 1/patología , Adolescente , Adulto , Agonistas de Dopamina/uso terapéutico , Femenino , Genotipo , Humanos , Masculino , Neoplasia Endocrina Múltiple Tipo 1/tratamiento farmacológico , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Enfermedades de las Paratiroides/metabolismo , Enfermedades de las Paratiroides/patología , Fenotipo , Enfermedades de la Hipófisis/tratamiento farmacológico , Enfermedades de la Hipófisis/metabolismo , Enfermedades de la Hipófisis/patología , Prolactinoma/metabolismo , Prolactinoma/patología , Estudios Retrospectivos , Análisis de Supervivencia , Adulto Joven
9.
World J Surg ; 43(3): 831-838, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30600364

RESUMEN

OBJECTIVE: Long-acting synthetic somatostatin analogues (SSA) are an essential part of the treatment of neuroendocrine neoplasms. We evaluated the chemopreventive effects of a long-acting somatostatin analogue on the development of pancreatic neuroendocrine neoplasms (pNENs) in a genetically engineered MEN1 knockout mouse model. MATERIALS AND METHODS: Heterozygote MEN1 knockout mice were injected every 28 days subcutaneously with the somatostatin analogue lanreotide (Somatuline Autogel©; Ipsen Pharma) or a placebo starting at day 35 after birth. Mice were euthanized after 6, 9, 12, 15 and 18 months, and the size and number of pNENs were measured due histological analysis and compared to the placebo group. RESULTS: The median tumor size of pNENs was statistically significantly smaller after 9 (control group vs. SSA group; 706.476 µm2 vs. 195.271 µm2; p = 0.0012), 12 (placebo group vs. SSA group 822.022 vs. 255.482; p ≤ 0.001), 15 (placebo group vs. SSA group 1192.568 vs. 273.533; p ≤ 0.001) and after 18 months (placebo group vs. SSA group 1328.299 vs. 864.587; p ≤ 0.001) in the SSA group. Comparing the amount of tumors in both groups, a significant reduction was achieved in treated Men1(+/-) mice (41%, p = 0.002). Immunostaining showed, however, no significant difference in the expression of the apoptosis marker caspase-3, but a significant difference in Ki67 index as a marker for tumor cell proliferation (p ≤ 0.005). CONCLUSION: Long-acting somatostatin analogues may be an effective chemopreventive approach to delay the progression of MEN1-associated pNENs. After our preclinical results, we would recommend to evaluate the effects of long-acting SSA in a prospective clinical trial.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasia Endocrina Múltiple Tipo 1/prevención & control , Tumores Neuroendocrinos/prevención & control , Neoplasias Pancreáticas/prevención & control , Péptidos Cíclicos/uso terapéutico , Somatostatina/análogos & derivados , Animales , Caspasa 3/metabolismo , Proliferación Celular , Quimioprevención , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Antígeno Ki-67/metabolismo , Ratones , Ratones Noqueados , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Neoplasia Endocrina Múltiple Tipo 1/patología , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas/genética , Somatostatina/uso terapéutico , Carga Tumoral
10.
J Pathol ; 242(1): 90-101, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28188614

RESUMEN

Foxa2, known as one of the pioneer factors, plays a crucial role in islet development and endocrine functions. Its expression and biological functions are regulated by various factors, including, in particular, insulin and glucagon. However, its expression and biological role in adult pancreatic α-cells remain elusive. In the current study, we showed that Foxa2 was overexpressed in islets from α-cell-specific Men1 mutant mice, at both the transcriptional level and the protein level. More importantly, immunostaining analyses showed its prominent nuclear accumulation, specifically in α-cells, at a very early stage after Men1 disruption. Similar nuclear FOXA2 expression was also detected in a substantial proportion (12/19) of human multiple endocrine neoplasia type 1 (MEN1) glucagonomas. Interestingly, our data revealed an interaction between Foxa2 and menin encoded by the Men1 gene. Furthermore, using several approaches, we demonstrated the relevance of this interaction in the regulation of two tested Foxa2 target genes, including the autoregulation of the Foxa2 promoter by Foxa2 itself. The current study establishes menin, a novel protein partner of Foxa2, as a regulator of Foxa2, the biological functions of which extend beyond the pancreatic endocrine cells. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Glucagonoma/metabolismo , Factor Nuclear 3-beta del Hepatocito/biosíntesis , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Neoplasias Pancreáticas/metabolismo , Animales , Regulación Neoplásica de la Expresión Génica , Glucagonoma/genética , Factor Nuclear 3-beta del Hepatocito/genética , Humanos , Ratones Transgénicos , Neoplasia Endocrina Múltiple Tipo 1/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/genética , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Transfección , Células Tumorales Cultivadas
11.
Trends Biochem Sci ; 38(8): 394-402, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23850066

RESUMEN

The protein menin is encoded by the MEN1 gene, which is mutated in patients with multiple endocrine neoplasia type 1 (MEN1) syndrome. Although menin acts as a tumor suppressor in endocrine organs, it is required for leukemic transformation in mouse models. Menin possesses these dichotomous functions probably because it can both positively and negatively regulate gene expression, as well as interact with a multitude of proteins with diverse functions. Here, we review the recent progress in understanding the molecular mechanisms by which menin functions. The crystal structures of menin with different binding partners reveal that menin is a key scaffold protein that functionally crosstalks with various partners to regulate gene transcription and interplay with multiple signaling pathways.


Asunto(s)
Regulación de la Expresión Génica , Modelos Biológicos , Matriz Nuclear/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Animales , Regulación hacia Abajo , Glándulas Endocrinas/metabolismo , Humanos , Neoplasia Endocrina Múltiple Tipo 1/genética , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Mutación , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/genética
12.
Gene Expr ; 17(3): 251-263, 2017 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-28485270

RESUMEN

Multiple endocrine neoplasia type 1 (MEN1) is a familial cancer syndrome with neuroendocrine tumorigenesis of the parathyroid glands, pituitary gland, and pancreatic islet cells. The MEN1 gene codes for the canonical tumor suppressor protein, menin. Its protein structure has recently been crystallized, and it has been investigated in a multitude of other tissues. In this review, we summarize recent advancements in understanding the structure of the menin protein and its function as a scaffold protein in histone modification and epigenetic gene regulation. Furthermore, we explore its role in hepatobiliary autoimmune diseases, cancers, and metabolic diseases. In particular, we discuss how menin expression and function are regulated by extracellular signaling factors and nuclear receptor activation in various hepatic cell types. How the many signaling pathways and tissue types affect menin's diverse functions is not fully understood. We show that small-molecule inhibitors affecting menin function can shed light on menin's broad role in pathophysiology and elucidate distinct menin-dependent processes. This review reveals menin's often dichotomous function through analysis of its role in multiple disease processes and could potentially lead to novel small-molecule therapies in the treatment of cholangiocarcinoma or biliary autoimmune diseases.


Asunto(s)
Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Enfermedades Autoinmunes/metabolismo , Transformación Celular Neoplásica , Colangiocarcinoma/metabolismo , Epigénesis Genética , Fibrosis , Regulación Neoplásica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina , Histonas , Humanos , Leucemia Bifenotípica Aguda/metabolismo , Hígado/metabolismo , Enfermedades Metabólicas/metabolismo , MicroARNs/metabolismo , Neoplasia Endocrina Múltiple Tipo 1/genética , Páncreas/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
13.
J Biol Chem ; 290(7): 3910-24, 2015 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-25538250

RESUMEN

Menin, the product of the multiple endocrine neoplasia type 1 (Men1) tumor suppressor gene, mediates the cell proliferation and differentiation actions of transforming growth factor-ß (TGF-ß) ligand family members. In vitro, menin modulates osteoblastogenesis and osteoblast differentiation promoted and sustained by bone morphogenetic protein-2 (BMP-2) and TGF-ß, respectively. To examine the in vivo function of menin in bone, we conditionally inactivated Men1 in mature osteoblasts by crossing osteocalcin (OC)-Cre mice with floxed Men1 (Men1(f/f)) mice to generate mice lacking menin in differentiating osteoblasts (OC-Cre;Men1(f/f) mice). These mice displayed significant reduction in bone mineral density, trabecular bone volume, and cortical bone thickness compared with control littermates. Osteoblast and osteoclast number as well as mineral apposition rate were significantly reduced, whereas osteocyte number was increased. Primary calvarial osteoblasts proliferated more quickly but had deficient mineral apposition and alkaline phosphatase activity. Although the mRNA expression of osteoblast marker and cyclin-dependent kinase inhibitor genes were all reduced, that of cyclin-dependent kinase, osteocyte marker, and pro-apoptotic genes were increased in isolated Men1 knock-out osteoblasts compared with controls. In contrast to the knock-out mice, transgenic mice overexpressing a human menin cDNA in osteoblasts driven by the 2.3-kb Col1a1 promoter, showed a gain of bone mass relative to control littermates. Osteoblast number and mineral apposition rate were significantly increased in the Col1a1-Menin-Tg mice. Therefore, osteoblast menin plays a key role in bone development, remodeling, and maintenance.


Asunto(s)
Desarrollo Óseo/fisiología , Huesos/fisiología , Diferenciación Celular , Osteoblastos/citología , Proteínas Proto-Oncogénicas/fisiología , Animales , Apoptosis , Western Blotting , Densidad Ósea , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Proliferación Celular , Células Cultivadas , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Osteoblastos/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
14.
Neuroendocrinology ; 103(5): 567-77, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26505735

RESUMEN

BACKGROUND/OBJECTIVE: Neuroendocrine neoplasms of the pancreas and duodenum with predominant or exclusive immunoreactivity for somatostatin (pdSOMs) are rare, and knowledge about tumour biology, treatment, survival and prognostic factors is limited. This study aims to describe clinical, pathological and biochemical features as well as treatment and prognosis of pdSOMs. DESIGN: Twenty-three patients with pdSOM (9 duodenal, 12 pancreatic and 2 unknown primary tumours) were identified from our prospective neuroendocrine tumour database, and data according to the study aims were recorded. RESULTS: Among the 9 patients with duodenal SOM, the male/female ratio was 4/5. All males and 1 female had neurofibromatosis type 1. Seven patients had stage 1A/B and 2 had stage 2B disease. The Ki-67 index was 1-5% (median 2%). Plasma somatostatin was elevated in the patients with 2B disease. Of the 14 patients with pancreatic SOM or an unknown primary tumour, the male/female ratio was 2/12. One male had multiple endocrine neoplasia type 1. Five had stage 1A/2B and 9 had stage 4. The Ki-67 index was 1-40% (median 7%). Plasma somatostatin was elevated in 7 patients. Patients reported symptoms related to the somatostatinoma syndrome, but none fulfilled the criteria for a full syndrome. Primary tumour in the pancreas, metastatic disease at diagnosis and higher tumour grade were all associated with significantly poorer survival. CONCLUSION: None of the patients with pdSOM presented with the full somatostatinoma syndrome. Prognostic factors are localisation of the primary tumour, dissemination and tumour grade. A Ki-67 index of 5% may discriminate the course of the disease.


Asunto(s)
Neoplasias Duodenales/metabolismo , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Neoplasias Pancreáticas/metabolismo , Somatostatina/metabolismo , Somatostatinoma/metabolismo , Adulto , Anciano , Neoplasias Duodenales/diagnóstico por imagen , Neoplasias Duodenales/cirugía , Femenino , Estudios de Seguimiento , Humanos , Antígeno Ki-67 , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Neoplasia Endocrina Múltiple Tipo 1/diagnóstico por imagen , Neoplasia Endocrina Múltiple Tipo 1/cirugía , Neoplasias Pancreáticas/diagnóstico por imagen , Neoplasias Pancreáticas/cirugía , Estudios Retrospectivos , Somatostatinoma/diagnóstico por imagen , Somatostatinoma/cirugía , Tomógrafos Computarizados por Rayos X , Organización Mundial de la Salud , Adulto Joven
15.
J Biol Chem ; 289(14): 9902-8, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-24563463

RESUMEN

Multiple endocrine neoplasia type I (MEN1) is an inherited syndrome that includes susceptibility to pancreatic islet hyperplasia. This syndrome results from mutations in the MEN1 gene, which encodes menin protein. Menin interacts with several transcription factors, including JunD, and inhibits their activities. However, the precise mechanism by which menin suppresses gene expression is not well understood. Here, we show that menin interacts with arsenite-resistant protein 2 (ARS2), a component of the nuclear RNA CAP-binding complex that is crucial for biogenesis of certain miRNAs including let-7a. The levels of primary-let-7a (pri-let-7a) are not affected by menin; however, the levels of mature let-7a are substantially decreased upon Men1 excision. Let-7a targets, including Insr and Irs2, pro-proliferative genes that are crucial for insulin-mediated signaling, are up-regulated in Men1-excised cells. Inhibition of let-7a using anti-miRNA in wild type cells is sufficient to enhance the expression of insulin receptor substrate 2 (IRS2) to levels observed in Men1-excised cells. Depletion of menin does not affect the expression of Drosha and CBP80, but substantially impairs the processing of pri-miRNA to pre-miRNA. Ars2 knockdown decreased let-7a processing in menin-expressing cells but had little impact on let-7a levels in menin-excised cells. As IRS2 is known to mediate insulin signaling and insulin/mitogen-induced cell proliferation, these findings collectively unravel a novel mechanism whereby menin suppresses cell proliferation, at least partly by promoting the processing of certain miRNAs, including let-7a, leading to suppression of Irs2 expression and insulin signaling.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , MicroARNs/biosíntesis , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Procesamiento Postranscripcional del ARN , ARN Neoplásico/biosíntesis , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Proliferación Celular , Proteínas de Unión al ADN , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Ratones , MicroARNs/genética , Neoplasia Endocrina Múltiple Tipo 1/genética , Neoplasia Endocrina Múltiple Tipo 1/patología , Proteínas de Neoplasias/genética , Complejo Proteico Nuclear de Unión a la Caperuza/genética , Complejo Proteico Nuclear de Unión a la Caperuza/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/genética , ARN Neoplásico/genética , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Ribonucleasa III/biosíntesis , Ribonucleasa III/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
16.
Hum Mol Genet ; 22(10): 1940-8, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23376981

RESUMEN

Multiple endocrine neoplasia syndrome type 1 (MEN1), which is secondary to mutation of the MEN1 gene, is a rare autosomal-dominant disease that predisposes mutation carriers to endocrine tumors. Although genotype-phenotype studies have so far failed to identify any statistical correlations, some families harbor recurrent tumor patterns. The function of MENIN is unclear, but has been described through the discovery of its interacting partners. Mutations in the interacting domains of MENIN functional partners have been shown to directly alter its regulation abilities. We report on a cohort of MEN1 patients from the Groupe d'étude des Tumeurs Endocrines. Patients with a molecular diagnosis and a clinical follow-up, totaling 262 families and 806 patients, were included. Associations between mutation type, location or interacting factors of the MENIN protein and death as well as the occurrence of MEN1-related tumors were tested using a frailty Cox model to adjust for potential heterogeneity across families. Accounting for the heterogeneity across families, the overall risk of death was significantly higher when mutations affected the JunD interacting domain (adjusted HR = 1.88: 95%-CI = 1.15-3.07). Patients had a higher risk of death from cancers of the MEN1 spectrum (HR = 2.34; 95%-CI = 1.23-4.43). This genotype-phenotype correlation study confirmed the lack of direct genotype-phenotype correlations. However, patients with mutations affecting the JunD interacting domain had a higher risk of death secondary to a MEN1 tumor and should thus be considered for surgical indications, genetic counseling and follow-up.


Asunto(s)
Neoplasia Endocrina Múltiple Tipo 1/genética , Neoplasia Endocrina Múltiple Tipo 1/mortalidad , Mutación , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas/genética , Familia , Femenino , Estudios de Seguimiento , Humanos , Masculino , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Factores de Riesgo
17.
Am J Pathol ; 182(6): 2355-67, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23583653

RESUMEN

Vascular therapeutic targeting requires thorough evaluation of the mechanisms activated in the specific context of each particular tumor type. We highlight structural, molecular, and functional microvascular aberrations contributing to development and maintenance of pancreatic neuroendocrine tumors (NETs), with special reference to multiple endocrine neoplasia 1 (MEN1) syndrome, using a Men1 mouse model. Tissue samples were analyzed by immunofluorescence to detect vessel density and pericyte distribution within the endocrine pancreas; expression of angiogenic factors was assessed by immunohistochemistry and quantitative real-time PCR in isolated islets and adenomas cultured under normoxic or hypoxic conditions. The increased vascular density of pancreatic NETs developed in Men1 mice was paralleled by an early and extensive redistribution of pericytes within endocrine tissue. These morphological alterations are supported by, and in some cases preceded by, fine-tuned variations in expression of several angiogenic regulators and are further potentiated by hypoxia. By combining two novel ex vivo and in vivo single-islet and tumor perfusion techniques, we demonstrated that both vascular reactivity and blood perfusion of tumor arterioles are significantly altered in response to glucose and L-nitro-arginine methyl ester. Our findings unravel multiple potential molecular and physiological targets differentially activated in the endocrine pancreas of Men1 mice and highlight the need for in-depth functional studies to fully understand the contribution of each component to development of pancreatic NETs in MEN1 syndrome.


Asunto(s)
Islotes Pancreáticos/irrigación sanguínea , Neoplasia Endocrina Múltiple Tipo 1/irrigación sanguínea , Neovascularización Patológica/patología , Neoplasias Pancreáticas/irrigación sanguínea , Inductores de la Angiogénesis/metabolismo , Animales , Arteriolas/fisiología , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Islotes Pancreáticos/metabolismo , Ratones , Ratones Noqueados , Microvasos/patología , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Neoplasia Endocrina Múltiple Tipo 1/patología , Proteínas de Neoplasias/metabolismo , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Pericitos/patología , Proteínas Proto-Oncogénicas/metabolismo , ARN Mensajero/genética
18.
Cancer Cell ; 10(5): 352-4, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17097557

RESUMEN

The p27Kip1 cyclin-dependent kinase inhibitor is considered to be a tumor suppressor even though somatic mutations in p27Kip1 are only rarely detected in human tumors. On the other hand, overwhelming evidence indicates that its hemizygous or posttranscriptional loss plays an important role in tumorigenesis. Based on these data, p27Kip1 was classified as a haploinsufficient tumor suppressor whose protein level has to be fine-tuned for optimal function. However, a recent study links germline mutations in p27Kip1 to multiple endocrine neoplasia syndrome in rats and humans, thus establishing p27Kip1 as a bona fide tumor suppressor gene.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Genes Supresores de Tumor , Neoplasia Endocrina Múltiple Tipo 1 , Neoplasias/metabolismo , Animales , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Histonas/metabolismo , Humanos , Masculino , Ratones , Neoplasia Endocrina Múltiple Tipo 1/genética , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Mutación , Inhibidores de Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Ratas
19.
Adv Sci (Weinh) ; 11(5): e2305659, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38044302

RESUMEN

Dysfunction of parvalbumin (PV) neurons is closely involved in depression, however, the detailed mechanism remains unclear. Based on the previous finding that multiple endocrine neoplasia type 1 (Protein: Menin; Gene: Men1) mutation (G503D) is associated with a higher risk of depression, a Menin-G503D mouse model is generated that exhibits heritable depressive-like phenotypes and increases PV expression in brain. This study generates and screens a serial of neuronal specific Men1 deletion mice, and found that PV interneuron Men1 deletion mice (PcKO) exhibit increased cortical PV levels and depressive-like behaviors. Restoration of Menin, knockdown PV expression or inhibition of PV neuronal activity in PV neurons all can ameliorate the depressive-like behaviors of PcKO mice. This study next found that ketamine stabilizes Menin by inhibiting protein kinase A (PKA) activity, which mediates the anti-depressant function of ketamine. These results demonstrate a critical role for Menin in depression, and prove that Menin is key to the antidepressant function of ketamine.


Asunto(s)
Antidepresivos , Ketamina , Neoplasia Endocrina Múltiple Tipo 1 , Animales , Ratones , Ketamina/farmacología , Neoplasia Endocrina Múltiple Tipo 1/genética , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Mutación , Parvalbúminas/genética , Parvalbúminas/metabolismo , Proteínas Proto-Oncogénicas/genética , Factores de Transcripción/genética , Antidepresivos/farmacología
20.
J Biol Chem ; 287(11): 8584-97, 2012 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-22275377

RESUMEN

Multiple endocrine neoplasia type 1 (MEN1) is characterized by tumors of the parathyroid, enteropancreas, and anterior pituitary. The MEN1 gene encodes the tumor suppressor menin of 610 amino acids that has multiple protein partners and activities. The particular pathways that, when lost, lead to tumorigenesis are not known. We demonstrated that members of a three-generation MEN1 kindred are heterozygous for a donor splice site mutation at the beginning of intron 3 (IVS3 + 1G→A). Lymphoblastoid cells of a mutant gene carrier had, in addition to the wild-type menin transcript, an aberrant transcript resulting from use of a cryptic splice site within exon III that splices to the start of exon IV. The predicted menin Δ(184-218) mutant has an in-frame deletion of 35 amino acids but is otherwise of wild-type sequence. The transfected menin Δ(184-218) mutant was well expressed and fully able to mediate the normal inhibition of the activity of the transcriptional regulators JunD and NF-κB. However, it was defective in mediating TGF-ß-stimulated Smad3 action in promoter-reporter assays in insulinoma cells. Importantly, lymphoblastoid cells from an individual heterozygous for the mutation had reduced TGF-ß-induced (Smad3) transcriptional activity but normal JunD and NF-κB function. In addition, the mutant gene carrier lymphoblastoid cells proliferated faster and were less responsive to the cytostatic effects of TGF-ß than cells from an unaffected family member. In conclusion, the menin mutant exhibits selective loss of the TGF-ß signaling pathway and loss of cell proliferation control contributing to the development of MEN1.


Asunto(s)
Secuencia de Aminoácidos/genética , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Sitios de Empalme de ARN/genética , Eliminación de Secuencia , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Adolescente , Adulto , Anciano de 80 o más Años , Animales , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Intrones , Masculino , Persona de Mediana Edad , Neoplasia Endocrina Múltiple Tipo 1/genética , Neoplasia Endocrina Múltiple Tipo 1/patología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ratas , Proteína smad3/genética , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA