Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Hepatol ; 66(4): 754-764, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27866920

RESUMO

BACKGROUND & AIMS: Hepatic stellate cell (HSC) transdifferentiation into collagen-producing myofibroblasts is a key event in hepatic fibrogenesis, but the transcriptional network that controls the acquisition of the activated phenotype is still poorly understood. In this study, we explored whether the nuclear receptor chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII) is involved in HSC activation and in the multifunctional role of these cells during the response to liver injury. METHODS: COUP-TFII expression was evaluated in normal and cirrhotic livers by immunohistochemistry and Western blot. The role of COUP-TFII in HSC was assessed by gain and loss of function transfection experiments and by generation of mice with COUP-TFII deletion in HSC. Molecular changes were determined by gene expression microarray and RT-qPCR. RESULTS: We showed that COUP-TFII is highly expressed in human fibrotic liver and in mouse models of hepatic injury. COUP-TFII expression rapidly increased upon HSC activation and it was associated with the regulation of genes involved in cell motility, proliferation and angiogenesis. Inactivation of COUP-TFII impairs proliferation and invasiveness in activated HSC and COUP-TFII deletion in mice abrogate HSC activation and angiogenesis. Finally, co-culture experiments with HSC and liver sinusoidal endothelial cells (SEC) showed that COUP-TFII expression in HSC influenced SEC migration and tubulogenesis via a hypoxia-independent and nuclear factor kappaB-dependent mechanism. CONCLUSION: This study elucidates a novel transcriptional pathway in HSC that is involved in the acquisition of the proangiogenic phenotype and regulates the paracrine signals between HSC and SEC during hepatic wound healing. LAY SUMMARY: In this study, we identified an important regulator of HSC pathobiology. We showed that the orphan receptor COUP-TFII is an important player in hepatic neoangiogenesis. COUP-TFII expression in HSC controls the crosstalk between HSC and endothelial cells coordinating vascular remodelling during liver injury. TRANSCRIPT PROFILING: ArrayExpress accession E-MTAB-1795.


Assuntos
Fator II de Transcrição COUP/metabolismo , Células Estreladas do Fígado/metabolismo , Animais , Fator II de Transcrição COUP/deficiência , Fator II de Transcrição COUP/genética , Comunicação Celular , Movimento Celular , Proliferação de Células , Transdiferenciação Celular , Técnicas de Cocultura , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Células Estreladas do Fígado/citologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Hipóxia/metabolismo , Hipóxia/patologia , Fígado/lesões , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/genética , Cirrose Hepática/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Fisiológica/genética , Regulação para Cima , Cicatrização/genética , Cicatrização/fisiologia
2.
Int J Cancer ; 139(7): 1574-85, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27193872

RESUMO

COUP-TFII belongs to the nuclear receptor family, which is highly expressed in many kinds of tumors. Previous studies have shown that COUP-TFII can promote tumor progression through regulating tumor angiogenesis and cell proliferation and migration of certain cancer cells. However, the function of COUP-TFII in renal cell carcinoma (RCC) is not clear. Here, we showed that clinical RCC tumor tissues showed much higher COUP-TFII expression level than adjacent normal tissues. When COUP-TFII was knocked down in RCC 769-P and 786-O cells by siRNA or shRNA-expressing lentivirus, the cell proliferation was markedly inhibited, and apoptosis increased. Moreover, the tumor growth of COUP-TFII knockdown 769-P and 786-O xenografts in nude mice was also obviously inhibited. Using qRT-PCR and Western blot, we showed that the expression of the tumor suppressor gene BRCA1 was upregulated in COUP-TFII knockdown cells. Simultaneously knockdown of BRCA1 and COUP-TFII partially rescued the inhibited cell proliferation and increased apoptosis in COUP-TFII single knockdown cells. These results indicate that COUP-TFII may play an oncogenic role in RCC, and COUP-TFII may promote tumor progression through inhibiting BRCA1.


Assuntos
Proteína BRCA1/genética , Fator II de Transcrição COUP/genética , Carcinoma de Células Renais/genética , Neoplasias Renais/genética , Animais , Apoptose/imunologia , Proteína BRCA1/biossíntese , Fator II de Transcrição COUP/biossíntese , Fator II de Transcrição COUP/deficiência , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Técnicas de Silenciamento de Genes , Genes BRCA1 , Xenoenxertos , Humanos , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Regulação para Cima
3.
Development ; 141(6): 1228-38, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24523456

RESUMO

In mammals, the homeodomain transcription factor Prox1 acts as the central regulator of lymphatic cell fate. Its restricted expression in a subset of cardinal vein cells leads to a switch towards lymphatic specification and hence represents a prerequisite for the initiation of lymphangiogenesis. Murine Prox1-null embryos lack lymphatic structures, and sustained expression of Prox1 is indispensable for the maintenance of lymphatic cell fate even at adult stages, highlighting the unique importance of this gene for the lymphatic lineage. Whether this pre-eminent role of Prox1 within the lymphatic vasculature is conserved in other vertebrate classes has remained unresolved, mainly owing to the lack of availability of loss-of-function mutants. Here, we re-examine the role of Prox1a in zebrafish lymphangiogenesis. First, using a transgenic reporter line, we show that prox1a is initially expressed in different endothelial compartments, becoming restricted to lymphatic endothelial cells only at later stages. Second, using targeted mutagenesis, we show that Prox1a is dispensable for lymphatic specification and subsequent lymphangiogenesis in zebrafish. In line with this result, we found that the functionally related transcription factors Coup-TFII and Sox18 are also dispensable for lymphangiogenesis. Together, these findings suggest that lymphatic commitment in zebrafish and mice is controlled in fundamentally different ways.


Assuntos
Proteínas de Homeodomínio/fisiologia , Linfangiogênese/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/crescimento & desenvolvimento , Animais , Animais Geneticamente Modificados , Fator II de Transcrição COUP/deficiência , Fator II de Transcrição COUP/genética , Fator II de Transcrição COUP/metabolismo , Diferenciação Celular , Linhagem da Célula , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Linfangiogênese/genética , Vasos Linfáticos/citologia , Vasos Linfáticos/metabolismo , Camundongos , Camundongos Knockout , Mutação , Fatores de Transcrição SOXF/deficiência , Fatores de Transcrição SOXF/genética , Fatores de Transcrição SOXF/metabolismo , Especificidade da Espécie , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética , Peixe-Zebra/genética , Peixe-Zebra/fisiologia , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
4.
Nature ; 493(7431): 236-40, 2013 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-23201680

RESUMO

Mutations in phosphatase and tensin homologue (PTEN) or genomic alterations in the phosphatidylinositol-3-OH kinase-signalling pathway are the most common genetic alterations reported in human prostate cancer. However, the precise mechanism underlying how indolent tumours with PTEN alterations acquire metastatic potential remains poorly understood. Recent studies suggest that upregulation of transforming growth factor (TGF)-ß signalling triggered by PTEN loss will form a growth barrier as a defence mechanism to constrain prostate cancer progression, underscoring that TGF-ß signalling might represent a pre-invasive checkpoint to prevent PTEN-mediated prostate tumorigenesis. Here we show that COUP transcription factor II (COUP-TFII, also known as NR2F2), a member of the nuclear receptor superfamily, serves as a key regulator to inhibit SMAD4-dependent transcription, and consequently overrides the TGF-ß-dependent checkpoint for PTEN-null indolent tumours. Overexpression of COUP-TFII in the mouse prostate epithelium cooperates with PTEN deletion to augment malignant progression and produce an aggressive metastasis-prone tumour. The functional counteraction between COUP-TFII and SMAD4 is reinforced by genetically engineered mouse models in which conditional loss of SMAD4 diminishes the inhibitory effects elicited by COUP-TFII ablation. The biological significance of COUP-TFII in prostate carcinogenesis is substantiated by patient sample analysis, in which COUP-TFII expression or activity is tightly correlated with tumour recurrence and disease progression, whereas it is inversely associated with TGF-ß signalling. These findings reveal that the destruction of the TGF-ß-dependent barrier by COUP-TFII is crucial for the progression of PTEN-mutant prostate cancer into a life-threatening disease, and supports COUP-TFII as a potential drug target for the intervention of metastatic human prostate cancer.


Assuntos
Fator II de Transcrição COUP/metabolismo , Transformação Celular Neoplásica , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Transdução de Sinais , Fator de Crescimento Transformador beta/antagonistas & inibidores , Animais , Fator II de Transcrição COUP/deficiência , Fator II de Transcrição COUP/genética , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Modelos Animais de Doenças , Progressão da Doença , Deleção de Genes , Humanos , Masculino , Camundongos , Metástase Neoplásica , PTEN Fosfo-Hidrolase/deficiência , PTEN Fosfo-Hidrolase/genética , Modelos de Riscos Proporcionais , Próstata/metabolismo , Próstata/patologia , Proteína Smad4/deficiência , Proteína Smad4/genética , Proteína Smad4/metabolismo , Fator de Crescimento Transformador beta/metabolismo
5.
Arterioscler Thromb Vasc Biol ; 32(11): e135-46, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22962329

RESUMO

OBJECTIVE: Septal defects and coronary vessel anomalies are common congenital heart defects, yet their ontogeny and the underlying genetic mechanisms are not well understood. Here, we investigated the role of chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII, NR2F2) in cardiac organogenesis. METHODS AND RESULTS: We analyzed embryos deficient in COUP-TFII and observed a spectrum of cardiac defects, including atrioventricular septal defect, thin-walled myocardium, and abnormal coronary morphogenesis. We show by expression analysis that COUP-TFII is expressed in the endocardium and the epicardium but not in the myocardium of the ventricle. Using endothelial-specific COUP-TFII mutants and molecular approaches, we show that COUP-TFII deficiency resulted in endocardial cushion hypoplasia. This was attributed to the reduced growth and survival of atrioventricular cushion mesenchymal cells and defective epithelial-mesenchymal transformation (EMT) in the underlying endocardium. In addition, the endocardial EMT defect was accompanied by downregulation of Snai1, one of the master regulators of EMT, and upregulation of vascular endothelial-cadherin. Furthermore, we show that although COUP-TFII does not play a major role in the formation of epicardial cell cysts, it is critically important for the formation of epicardium. Ablation of COUP-TFII impairs epicardial EMT and coronary plexus formation. CONCLUSIONS: Our results reveal that COUP-TFII plays cell-autonomous roles in the endocardium and the epicardium for endocardial and epicardial EMT, which are required for proper valve and coronary vessel formation during heart development.


Assuntos
Fator II de Transcrição COUP/metabolismo , Vasos Coronários/embriologia , Vasos Coronários/metabolismo , Endocárdio/embriologia , Endocárdio/metabolismo , Cardiopatias Congênitas/embriologia , Cardiopatias Congênitas/metabolismo , Coração/embriologia , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Fator II de Transcrição COUP/deficiência , Fator II de Transcrição COUP/genética , Caderinas/genética , Caderinas/metabolismo , Movimento Celular , Vasos Coronários/patologia , Coxins Endocárdicos/embriologia , Coxins Endocárdicos/metabolismo , Endocárdio/patologia , Transição Epitelial-Mesenquimal , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfogênese , Miocárdio/metabolismo , Miocárdio/patologia , Neovascularização Fisiológica , Pericárdio/embriologia , Pericárdio/metabolismo , Pericárdio/patologia , Fatores de Transcrição da Família Snail , Técnicas de Cultura de Tecidos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
6.
PLoS One ; 7(5): e38278, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22693611

RESUMO

INTRODUCTION: The orphan nuclear receptor COUP-TFII plays an undefined role in breast cancer. Previously we reported lower COUP-TFII expression in tamoxifen/endocrine-resistant versus sensitive breast cancer cell lines. The identification of COUP-TFII-interacting proteins will help to elucidate its mechanism of action as a transcriptional regulator in breast cancer. RESULTS: FLAG-affinity purification and multidimensional protein identification technology (MudPIT) identified nucleolin among the proteins interacting with COUP-TFII in MCF-7 tamoxifen-sensitive breast cancer cells. Interaction of COUP-TFII and nucleolin was confirmed by coimmunoprecipitation of endogenous proteins in MCF-7 and T47D breast cancer cells. In vitro studies revealed that COUP-TFII interacts with the C-terminal arginine-glycine repeat (RGG) domain of nucleolin. Functional interaction between COUP-TFII and nucleolin was indicated by studies showing that siRNA knockdown of nucleolin and an oligonucleotide aptamer that targets nucleolin, AS1411, inhibited endogenous COUP-TFII-stimulated RARB2 expression in MCF-7 and T47D cells. Chromatin immunoprecipitation revealed COUP-TFII occupancy of the RARB2 promoter was increased by all-trans retinoic acid (atRA). RARß2 regulated gene RRIG1 was increased by atRA and COUP-TFII transfection and inhibited by siCOUP-TFII. Immunohistochemical staining of breast tumor microarrays showed nuclear COUP-TFII and nucleolin staining was correlated in invasive ductal carcinomas. COUP-TFII staining correlated with ERα, SRC-1, AIB1, Pea3, MMP2, and phospho-Src and was reduced with increased tumor grade. CONCLUSIONS: Our data indicate that nucleolin plays a coregulatory role in transcriptional regulation of the tumor suppressor RARB2 by COUP-TFII.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Fator II de Transcrição COUP/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Receptores do Ácido Retinoico/genética , Ativação Transcricional , Animais , Aptâmeros de Nucleotídeos , Neoplasias da Mama/genética , Fator II de Transcrição COUP/deficiência , Fator II de Transcrição COUP/genética , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Gradação de Tumores , Oligodesoxirribonucleotídeos/farmacologia , Fosfoproteínas/química , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Estrutura Terciária de Proteína , Proteínas de Ligação a RNA/química , Sequências Repetitivas de Ácido Nucleico , Análise Serial de Tecidos , Ativação Transcricional/efeitos dos fármacos , Tretinoína/farmacologia , Nucleolina
7.
PLoS One ; 7(5): e35810, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22606236

RESUMO

BACKGROUND: The Nuclear Receptor 2F2 (NR2F2/COUP-TFII) heterozygous knockout mice display low basal insulinemia and enhanced insulin sensitivity. We previously established that insulin represses NR2F2 gene expression in pancreatic ß-cells. The cis-regulatory region of the NR2F2 promoter is unknown and its influence on metabolism in humans is poorly understood. The present study aimed to identify the regulatory regions that control NR2F2 gene transcription and to evaluate the effect of NR2F2 promoter variation on glucose homeostasis in humans. METHODOLOGY/PRINCIPAL FINDINGS: Regulation of the NR2F2 promoter was assessed using gene reporter assays, ChIP and gel shift experiments. The effects of variation at SNP rs3743462 in NR2F2 on quantitative metabolic traits were studied in two European prospective cohorts. We identified a minimal promoter region that down-regulates NR2F2 expression by attenuating HNF4α activation in response to high glucose concentrations. Subjects of the French DESIR population, who carried the rs3743462 T-to-C polymorphism, located in the distal glucose-responsive promoter, displayed lower basal insulin levels and lower HOMA-IR index. The C-allele at rs3743462 was associated with increased NR2F2 binding and decreased NR2F2 gene expression. CONCLUSIONS/SIGNIFICANCE: The rs3743462 polymorphism affects glucose-responsive NR2F2 promoter regulation and thereby may influence whole-body insulin sensitivity, suggesting a role of NR2F2 in the control of glucose homeostasis in humans.


Assuntos
Fator II de Transcrição COUP/genética , Glucose/metabolismo , Resistência à Insulina/genética , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas , Adulto , Animais , Sequência de Bases , Glicemia/metabolismo , Fator II de Transcrição COUP/deficiência , Fator II de Transcrição COUP/metabolismo , Linhagem Celular , Estudos de Coortes , DNA/genética , Regulação da Expressão Gênica , Fator 4 Nuclear de Hepatócito/metabolismo , Humanos , Insulina/sangue , Resistência à Insulina/fisiologia , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Estudos Prospectivos , Homologia de Sequência do Ácido Nucleico
8.
Mol Endocrinol ; 24(6): 1297-304, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20382891

RESUMO

Nuclear receptors and coregulators orchestrate diverse aspects of biological functions and inappropriate expression of these factors often associates with human diseases. The present study describes a conditional overexpression system consisting of a minigene located at the Rosa26 locus in the genome of mouse embryonic stem (ES) cells. Before activation, the minigene is silent due to a floxed STOP cassette inserted between the promoter and the transgene. Upon cre-mediated excision of the STOP cassette, the minigene constitutively expresses the tagged transgene driven by the ubiquitous CAGGS promoter. Thus, this system can be used to express target gene in any tissue in a spatial and/or temporal manner if respective cre mouse lines are available. Serving as proof of principle, the CAG-S-hCOUP-TFI allele was generated in ES cells and subsequently in mice. This allele was capable of conditionally overexpressing human chicken ovalbumin upstream promoter-transcription factor I (COUP-TFI) in all tissues tested upon activation by cre drivers. This allele was further subjected to address functionality of expressed COUP-TFI and the functional similarity between COUP-TFI and COUP-TFII. Expression of COUP-TFI in COUP-TFII-ablated uterus suppressed aberrant estrogen receptor-alpha activities and rescued implantation and decidualization defects of COUP-TFII mutants, suggesting that COUP-TFI and COUP-TFII are able to functionally compensate for each other in the uterus. A toolbox currently under construction will contain ES cell lines for overexpressing all 48 nuclear receptors and selected 10 coregulators. Upon completion, it will be a very valuable resource for the scientific community. Several ES cells are currently available for distribution.


Assuntos
Células-Tronco Embrionárias/metabolismo , Coativadores de Receptor Nuclear/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Alelos , Animais , Fator I de Transcrição COUP/metabolismo , Fator II de Transcrição COUP/deficiência , Fator II de Transcrição COUP/metabolismo , Linhagem Celular , Feminino , Humanos , Camundongos , Útero/metabolismo , Útero/patologia
9.
Nature ; 463(7283): 953-7, 2010 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-20164929

RESUMO

One of the most notable features of the vertebrate body plan organization is its bilateral symmetry, evident at the level of vertebrae and skeletal muscles. Here we show that a mutation in Rere (also known as atrophin2) leads to the formation of asymmetrical somites in mouse embryos, similar to embryos deprived of retinoic acid. Furthermore, we also demonstrate that Rere controls retinoic acid signalling, which is required to maintain somite symmetry by interacting with Fgf8 in the left-right signalling pathway. Rere forms a complex with Nr2f2, p300 (also known as Ep300) and a retinoic acid receptor, which is recruited to the retinoic acid regulatory element of retinoic acid targets, such as the Rarb promoter. Furthermore, the knockdown of Nr2f2 and/or Rere decreases retinoic acid signalling, suggesting that this complex is required to promote transcriptional activation of retinoic acid targets. The asymmetrical expression of Nr2f2 in the presomitic mesoderm overlaps with the asymmetry of the retinoic acid signalling response, supporting its implication in the control of somitic symmetry. Misregulation of this mechanism could be involved in symmetry defects of the human spine, such as those observed in patients with scoliosis.


Assuntos
Padronização Corporal/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais , Somitos/embriologia , Somitos/metabolismo , Tretinoína/metabolismo , Animais , Fator II de Transcrição COUP/deficiência , Fator II de Transcrição COUP/genética , Fator II de Transcrição COUP/metabolismo , Linhagem Celular , Proteína p300 Associada a E1A/metabolismo , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Fator 8 de Crescimento de Fibroblasto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Regiões Promotoras Genéticas/genética , Receptores do Ácido Retinoico/genética , Receptores do Ácido Retinoico/metabolismo , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Elementos de Resposta/genética
10.
PLoS Genet ; 3(6): e102, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17590085

RESUMO

Progesterone and estrogen are critical regulators of uterine receptivity. To facilitate uterine remodeling for embryo attachment, estrogen activity in the uterine epithelia is attenuated by progesterone; however, the molecular mechanism by which this occurs is poorly defined. COUP-TFII (chicken ovalbumin upstream promoter transcription factor II; also known as NR2F2), a member of the nuclear receptor superfamily, is highly expressed in the uterine stroma and its expression is regulated by the progesterone-Indian hedgehog-Patched signaling axis that emanates from the epithelium. To further assess COUP-TFII uterine function, a conditional COUP-TFII knockout mouse was generated. This mutant mouse is infertile due to implantation failure, in which both embryo attachment and uterine decidualization are impaired. Using this animal model, we have identified a novel genetic pathway in which BMP2 lies downstream of COUP-TFII. Epithelial progesterone-induced Indian hedgehog regulates stromal COUP-TFII, which in turn controls BMP2 to allow decidualization to manifest in vivo. Interestingly, enhanced epithelial estrogen activity, which impedes maturation of the receptive uterus, was clearly observed in the absence of stromal-derived COUP-TFII. This finding is consistent with the notion that progesterone exerts its control of implantation through uterine epithelial-stromal cross-talk and reveals that stromal-derived COUP-TFII is an essential mediator of this complex cross-communication pathway. This finding also provides a new signaling paradigm for steroid hormone regulation in female reproductive biology, with attendant implications for furthering our understanding of the molecular mechanisms that underlie dysregulation of hormonal signaling in such human reproductive disorders as endometriosis and endometrial cancer.


Assuntos
Fator II de Transcrição COUP/fisiologia , Implantação do Embrião/fisiologia , Progesterona/fisiologia , Receptores de Estrogênio/antagonistas & inibidores , Útero/metabolismo , Animais , Fator II de Transcrição COUP/deficiência , Fator II de Transcrição COUP/genética , Comunicação Celular/genética , Epitélio/metabolismo , Epitélio/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Gravidez , Receptor Cross-Talk/fisiologia , Receptores de Estrogênio/metabolismo , Células Estromais/metabolismo , Células Estromais/fisiologia
11.
Proc Natl Acad Sci U S A ; 102(45): 16351-6, 2005 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-16251273

RESUMO

Congenital diaphragmatic hernia (CDH), a life-threatening anomaly, is a major cause of pediatric mortality. Although the disease was described >350 years ago, the etiology of CDH is poorly understood. Here, we show that tissue-specific null mutants of COUP-TFII exhibit Bochdalek-type CDH, the most common form of CDH. COUP-TFII, a member of orphan nuclear receptors, is expressed in regions critical for the formation of the diaphragm during embryonic development. Ablation of COUP-TFII in the foregut mesenchyme, including the posthepatic mesenchymal plate (PHMP), results in the malformation of the diaphragm and the failure of appropriate attachment of the PHMP to the body wall. Thus, both the stomach and liver enter the thoracic cavity, leading to lung hypoplasia and neonatal death. Recently a minimally deleted region for CDH has been identified on chromosome 15q26.1-26.2 by CGH array and FISH analysis. COUP-TFII is one of the four known genes residing within this critical region. Our finding suggests that COUP-TFII is a likely contributor to the formation of CDH in individuals with 15q deletions, and it may also be a potential contributor to some other Bochdalek-type of CDH.


Assuntos
Fator II de Transcrição COUP/genética , Modelos Animais de Doenças , Hérnias Diafragmáticas Congênitas , Mutação , Animais , Fator II de Transcrição COUP/deficiência , Fator II de Transcrição COUP/fisiologia , Diafragma/embriologia , Hérnia Diafragmática/etiologia , Hérnia Diafragmática/genética , Camundongos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA