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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neurochem ; 165(5): 660-681, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36648143

RESUMEN

Schwann cells (SCs) are known to produce myelin for saltatory nerve conduction in the peripheral nervous system (PNS). Schwann cell differentiation and myelination processes are controlled by several transcription factors including Sox10, Oct6/Pou3f1, and Krox20/Egr2. Chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII/NR2F2) is an orphan receptor that plays a role in the development and differentiation. However, the role of COUP-TFII in the transcriptional regulatory network of SC differentiation has not been fully identified yet. Thus, the objective of this study was to investigate the role and molecular hierarchy of COUP-TFII during cAMP-induced SC differentiation. Our results showed that dibutyryl-cAMP (db-cAMP) increased expression levels of COUP-TFII along with the expressions of Oct6, Krox20, and myelin-related genes known to be related to SC differentiation. Our mechanistic studies showed that COUP-TFII acted downstream of Hsp90/ErbB2/Gab1/ERK-AKT pathway during db-cAMP-induced SC differentiation. In addition, we found that COUP-TFII induced Krox20 expression by directly binding to Krox20-MSE8 as revealed by chromatin immunoprecipitation assay and promoter activity assay. In line with this, the expression of COUP-TFII was increased before up-regulation of Oct6, Krox20, and myelin-related genes in the sciatic nerves during early postnatal myelination period. Finally, COUP-TFII knockdown by COUP-TFII siRNA or via AAV-COUP-TFII shRNA in SCs inhibited db-cAMP-induced SC differentiation and in vitro myelination of sensory axons, respectively. Taken together, these findings indicate that COUP-TFII might be involved in postnatal myelination through induction of Krox20 in SCs. Our results present a new insight into the transcriptional regulatory mechanism in SC differentiation and myelination.


Asunto(s)
Factor de Transcripción COUP II , Proteína 2 de la Respuesta de Crecimiento Precoz , Células de Schwann , Animales , Ratas , Diferenciación Celular , Células Cultivadas , Factor de Transcripción COUP II/genética , Factor de Transcripción COUP II/metabolismo , AMP Cíclico/metabolismo , Regulación de la Expresión Génica , Vaina de Mielina/metabolismo , Células de Schwann/citología , Células de Schwann/metabolismo , Nervio Ciático/metabolismo , Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo
2.
EMBO Rep ; 22(6): e51169, 2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-34031962

RESUMEN

Recent studies demonstrate that metabolic disturbance, such as augmented glycolysis, contributes to fibrosis. The molecular regulation of this metabolic perturbation in fibrosis, however, has been elusive. COUP-TFII (also known as NR2F2) is an important regulator of glucose and lipid metabolism. Its contribution to organ fibrosis is undefined. Here, we found increased COUP-TFII expression in myofibroblasts in human fibrotic kidneys, lungs, kidney organoids, and mouse kidneys after injury. Genetic ablation of COUP-TFII in mice resulted in attenuation of injury-induced kidney fibrosis. A non-biased proteomic study revealed the suppression of fatty acid oxidation and the enhancement of glycolysis pathways in COUP-TFII overexpressing fibroblasts. Overexpression of COUP-TFII in fibroblasts also induced production of alpha-smooth muscle actin (αSMA) and collagen 1. Knockout of COUP-TFII decreased glycolysis and collagen 1 levels in fibroblasts. Chip-qPCR revealed the binding of COUP-TFII on the promoter of PGC1α. Overexpression of COUP-TFII reduced the cellular level of PGC1α. Targeting COUP-TFII serves as a novel treatment approach for mitigating fibrosis in chronic kidney disease and potentially fibrosis in other organs.


Asunto(s)
Factor de Transcripción COUP II , Receptores Nucleares Huérfanos , Animales , Factor de Transcripción COUP II/genética , Factor de Transcripción COUP II/metabolismo , Fibrosis , Glucólisis/genética , Riñón , Ratones , Ratones Noqueados , Miofibroblastos , Receptores Nucleares Huérfanos/metabolismo , Proteómica
3.
Proc Natl Acad Sci U S A ; 117(41): 25859-25868, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-33004630

RESUMEN

Endometriosis is a highly prevalent gynecological disease with severe negative impacts on life quality and financial burden. Unfortunately, there is no cure for this disease, which highlights the need for further investigation about the pathophysiology of this disease to provide clues for developing novel therapeutic regimens. Herein, we identified that vascular endothelial growth factor (VEGF)-C, a potent lymphangiogenic factor, is up-regulated in endometriotic cells and contributes to increased lymphangiogenesis. Bioinformatic analysis and molecular biological characterization revealed that VEGF-C is negatively regulated by an orphan nuclear receptor, chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII). Further studies demonstrated that proinflammatory cytokines, via suppression of COUP-TFII level, induce VEGF-C overexpression. More importantly, we show that functional VEGF-C is transported by extracellular vesicles (EVs) to enhance the lymphangiogenic ability of lymphatic endothelial cells. Autotransplanted mouse model of endometriosis showed lenvatinib treatment abrogated the increased lymphatic vessels development in the endometriotic lesion, enlarged retroperitoneal lymph nodes, and immune cells infiltration, indicating that blocking VEGF-C signaling can reduce local chronic inflammation and concomitantly endometriosis development. Evaluation of EV-transmitted VEGF-C from patients' sera demonstrates it is a reliable noninvasive way for clinical diagnosis. Taken together, we identify the vicious cycle of inflammation, COUP-TFII, VEGF-C, and lymphangiogenesis in the endometriotic microenvironment, which opens up new horizons in understanding the pathophysiology of endometriosis. VEGF-C not only can serve as a diagnostic biomarker but also a molecular target for developing therapeutic regimens.


Asunto(s)
Endometriosis/inmunología , Vesículas Extracelulares/inmunología , Sistema Inmunológico/inmunología , Linfangiogénesis , Factor C de Crecimiento Endotelial Vascular/inmunología , Animales , Factor de Transcripción COUP II/genética , Factor de Transcripción COUP II/inmunología , Citocinas/genética , Citocinas/inmunología , Endometriosis/genética , Endometriosis/fisiopatología , Células Endoteliales/inmunología , Vesículas Extracelulares/genética , Femenino , Humanos , Vasos Linfáticos/inmunología , Ratones , Factor C de Crecimiento Endotelial Vascular/genética
4.
Biochem Biophys Res Commun ; 611: 183-189, 2022 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-35490658

RESUMEN

We previously reported that the canonical Wnt signaling pathway is activated during compensatory islet hyperplasia in prediabetic mice. Here, we aimed to expand our knowledge concerning the Wnt signaling partners and modulators involved in this process. We report here that Axin1, Axin2, and DACT1, inhibitors of the canonical Wnt signaling pathway, displayed no change in their expression, while GSK-3ß, a multi-functional kinase that acts as a negative regulator of this pathway as well as affects insulin secretion/action, was up-regulated in hyperplastic islets of prediabetic mice. We also observed that COUP-TFII, a protein that acts positively on Wnt-target genes related to cell proliferation, displays a significant increase in gene expression and protein content and is highly immunolabeled in islet cell nuclei of prediabetic mice compared to control islets. These findings suggest that GSK-3ß and COUP-TFII may play a role in beta-cell dysfunction and hyperplasia during type 2 prediabetes.


Asunto(s)
Estado Prediabético , Vía de Señalización Wnt , Animales , Proliferación Celular , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Hiperplasia , Ratones , Estado Prediabético/genética , Vía de Señalización Wnt/fisiología , beta Catenina/metabolismo
5.
Int J Mol Sci ; 23(3)2022 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-35163483

RESUMEN

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor known for mediating the toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and related compounds. Although the canonical mechanism of AhR activation involves heterodimerization with the aryl hydrocarbon receptor nuclear translocator, other transcriptional regulators that interact with AhR have been identified. Enrichment analysis of motifs in AhR-bound genomic regions implicated co-operation with COUP transcription factor (COUP-TF) and hepatocyte nuclear factor 4 (HNF4). The present study investigated AhR, HNF4α and COUP-TFII genomic binding and effects on gene expression associated with liver-specific function and cell differentiation in response to TCDD. Hepatic ChIPseq data from male C57BL/6 mice at 2 h after oral gavage with 30 µg/kg TCDD were integrated with bulk RNA-sequencing (RNAseq) time-course (2-72 h) and dose-response (0.01-30 µg/kg) datasets to assess putative AhR, HNF4α and COUP-TFII interactions associated with differential gene expression. Functional enrichment analysis of differentially expressed genes (DEGs) identified differential binding enrichment for AhR, COUP-TFII, and HNF4α to regions within liver-specific genes, suggesting intersections associated with the loss of liver-specific functions and hepatocyte differentiation. Analysis found that the repression of liver-specific, HNF4α target and hepatocyte differentiation genes, involved increased AhR and HNF4α binding with decreased COUP-TFII binding. Collectively, these results suggested TCDD-elicited loss of liver-specific functions and markers of hepatocyte differentiation involved interactions between AhR, COUP-TFII and HNF4α.


Asunto(s)
Factores de Transcripción COUP/metabolismo , Secuenciación de Inmunoprecipitación de Cromatina , Genoma , Factor Nuclear 4 del Hepatocito/metabolismo , Hígado/metabolismo , Dibenzodioxinas Policloradas/toxicidad , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Secuencia de Bases , Bases de Datos Genéticas , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Masculino , Ratones Endogámicos C57BL , Motivos de Nucleótidos/genética , Unión Proteica , RNA-Seq , Transcripción Genética
6.
Biol Reprod ; 105(5): 1283-1306, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34225363

RESUMEN

In males, Leydig cells are the main producers of testosterone and insulin-like 3 (INSL3), two hormones essential for sex differentiation and reproductive functions. Chicken ovalbumin upstream promoter-transcription factors I (COUP-TFI/NR2F1) and COUP-TFII (NR2F2) belong to the steroid/thyroid hormone nuclear receptor superfamily of transcription factors. In the testis, COUP-TFII is expressed and plays a role in the differentiation of cells committed to give rise to fully functional steroidogenic adult Leydig cells. Steroid production has also been shown to be diminished in COUP-TFII-depleted Leydig cells, indicating an important functional role in steroidogenesis. Until now, only a handful of target genes have been identified for COUP-TFII in Leydig cells. To provide new information into the mechanism of action of COUP-TFII in Leydig cells, we performed microarray analyses of COUP-TFII-depleted MA-10 Leydig cells. We identified 262 differentially expressed genes in COUP-TFII-depleted MA-10 cells. Many of the differentially expressed genes are known to be involved in lipid biosynthesis, lipid metabolism, male gonad development, and steroidogenesis. We validated the microarray data for a subset of the modulated genes by RT-qPCR. Downregulated genes included hydroxy-delta-5-steroid dehydrogenase, 3 beta- and steroid delta-isomerase 1 (Hsd3b1), cytochrome P450, family 11, subfamily a, polypeptide 1 (Cyp11a1), prolactin receptor (Prlr), nuclear receptor subfamily 0, group B, member 2 (Shp/Nr0b2), ferredoxin 1 (Fdx1), scavenger receptor class B, member 1 (Scarb1), inhibin alpha (Inha), and glutathione S-transferase, alpha 3 (Gsta3). Finally, analysis of the Gsta3 and Inha gene promoters showed that at least two of the downregulated genes are potentially new direct targets for COUP-TFII. These data provide new evidence that further strengthens the important nature of COUP-TFII in steroidogenesis, androgen homeostasis, cellular defense, and differentiation in mouse Leydig cells.


Asunto(s)
Factor de Transcripción COUP II/genética , Regulación de la Expresión Génica , Células Intersticiales del Testículo/metabolismo , Transducción de Señal , Animales , Factor de Transcripción COUP II/metabolismo , Línea Celular , Masculino , Ratones
7.
EMBO Rep ; 20(6)2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30988000

RESUMEN

Oncogenic signals contribute to enhanced glycolysis and mTORC1 activity, leading to rapid cell proliferation in cancer. Regulation of glycolysis and mTORC1 by PI3K/Akt signaling is well established, but how KRAS-induced MEK signaling regulates these pathways remains poorly understood. Here, we report a role for MEK-driven lactate production in mTORC1 activation in KRAS-activated cells. KRAS/MEK-induced upregulation of the chicken ovalbumin upstream promoter transcriptional factor II (COUP-TFII) increases the expression of lactate dehydrogenase A (LDHA), resulting in lactate production and mTORC1 activation. Further, lactate inhibits the interaction of TSC2 and Rheb, leading to the cellular activation of mTORC1 irrespective of growth factor stimulation. These findings suggest that COUP-TFII is a novel oncogenic mediator, connecting KRAS signaling and glycolysis, and leading to mTORC1 activation and cellular growth.


Asunto(s)
Factor de Transcripción COUP II/metabolismo , Ácido Láctico/biosíntesis , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Transducción de Señal , Factor de Transcripción COUP II/genética , Línea Celular Tumoral , Expresión Génica , Técnicas de Silenciamiento del Gen , Glucólisis , Humanos , Modelos Biológicos , Proteína Homóloga de Ras Enriquecida en el Cerebro/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo
8.
Endocr J ; 67(3): 283-293, 2020 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-31801919

RESUMEN

The chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII) plays essential roles in organogenesis of embryos. Recently COUP-TFII is also implicated in several diseases in adults. Here we focus on the role of COUP-TFII in cisplatin-induced acute kidney injury (AKI). COUP-TFII was the most abundantly expressed in the kidney among organs. Male tamoxifen-inducible COUP-TFII-knockout mice or control mice were intraperitoneally treated with 30 mg/kg body weight of cisplatin at 12 weeks old to induce AKI. The kidney samples were subject to morphological studies, terminal deoxynucleotidyl transferase-mediated deoxyuridine nick-end labeling (TUNEL) assay, immunohistochemistry and RT-qPCR. Serum levels of creatinine, blood urea nitrogen (BUN) and tumor necrosis factor alpha (TNF-α) were measured. Administration of cisplatin induced a more severe AKI in adult COUP-TFII-knockout mice. An increase in dead cells in both the proximal tubules and thick ascending limb of Henle's loop (TAL) was observed in the knockout mouse kidney. The expression levels of COUP-TFII decreased in the TAL by cisplatin administration. There was no difference in the expression levels of transporter mRNAs responsible for cellular cisplatin uptake between control and knockout mouse kidney. COUP-TFII-knockout mice and COUP-TFII-depleted cells exhibited an elevation in TNF-α levels, suggesting the involvement of the TNF-α pathway. Chromatin immunoprecipitation showed that COUP-TFII was enriched in the potential binding site, suggesting that COUP-TFII might directly suppress the TNF-α gene at transcriptional level. These results indicate the involvement of COUP-TFII in the pathophysiology of AKI and COUP-TFII may be a potential therapeutic target for AKI.


Asunto(s)
Lesión Renal Aguda/genética , Antineoplásicos/efectos adversos , Factor de Transcripción COUP II/genética , Cisplatino/efectos adversos , Riñón/patología , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Factor de Transcripción COUP II/metabolismo , Humanos , Riñón/metabolismo , Masculino , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Factor de Necrosis Tumoral alfa/sangre
9.
Acta Biochim Biophys Sin (Shanghai) ; 52(11): 1247-1256, 2020 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-33166992

RESUMEN

Intrahepatic cholangiocarcinoma (ICC) arises from cholangiocytes in the intrahepatic bile duct and is the second most common type of liver cancer. The overexpression of COUP-TFII has been observed in several types of malignancies. However, its role in ICC progression remains unclear. In this study, we found that the protein level of COUP-TFII was increased, but the mRNA level was unchanged in ICC tissues. High protein expression was positively associated with tumor size, lymph node metastasis, and poor prognosis in ICC patients. Furthermore, the overexpression of COUP-TFII promoted the proliferation, migration, and invasion of ICC cells in vitro and enhanced tumor growth and metastasis in nude mouse models. Mechanistic studies revealed that COUP-TFII induced epithelial-to-mesenchymal transition in ICC cells by upregulating Snail expression. Moreover, the activation of PI3K/AKT signaling led to the upregulation of COUP-TFII protein expression in ICC. Together, these findings indicate that COUP-TFII promotes epithelial-to-mesenchymal transition and metastasis in ICC and suggest that this protein is a potential target for adjuvant therapy for these patients.


Asunto(s)
Neoplasias de los Conductos Biliares/genética , Factor de Transcripción COUP II/genética , Factor de Transcripción COUP II/metabolismo , Colangiocarcinoma/genética , Transición Epitelial-Mesenquimal/genética , Factores de Transcripción de la Familia Snail/genética , Anciano , Animales , Neoplasias de los Conductos Biliares/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Colangiocarcinoma/diagnóstico , Colangiocarcinoma/patología , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Metástasis de la Neoplasia/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Pronóstico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Regulación hacia Arriba
10.
Development ; 143(10): 1753-65, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27034423

RESUMEN

GABAergic interneurons are highly heterogeneous and originate in the subpallium mainly from the medial (MGE) and caudal (CGE) ganglionic eminences according to a precise temporal sequence. MGE-derived cells disperse dorsally and migrate towards all regions of the cortex, but little is known about how CGE-derived cells reach their targets during development. Here, we unravel the existence of two novel CGE caudo-rostral migratory streams, one located laterally (LMS) and the other one more medially (MMS), that, together with the well-known caudal migratory stream (CMS), contribute to populate the neocortex, hippocampus and amygdala. These paths appear in a precise temporal sequence and express a distinct combination of transcription factors, such as SP8, PROX1, COUP-TFI and COUP-TFII. By inactivating COUP-TFI in developing interneurons, the lateral and medial streams are perturbed and expression of SP8 and COUP-TFII affected. As a consequence, adult mutant neocortices have laminar-specific alterations of distinct cortical interneuron subtypes. Overall, we propose that the existence of spatially and temporally regulated migratory paths in the subpallium contributes to the laminar distribution and specification of distinct interneuron subpopulations in the adult brain.


Asunto(s)
Encéfalo/citología , Encéfalo/embriología , Movimiento Celular , Interneuronas/citología , Eminencia Media/citología , Envejecimiento/metabolismo , Animales , Recuento de Células , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Interneuronas/metabolismo , Ratones Transgénicos , Modelos Biológicos , Mutación/genética , Factores de Tiempo , Factores de Transcripción/metabolismo
11.
Development ; 142(9): 1593-605, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25922524

RESUMEN

Neural stem cells (NSCs) persist in the adult mammalian subventricular zone (SVZ) of the lateral ventricle. Primary NSCs generate rapidly dividing intermediate progenitor cells, which in turn generate neuroblasts that migrate along the rostral migratory stream (RMS) to the olfactory bulb (OB). Here, we have examined the role of the COUP-TFI and COUP-TFII orphan nuclear receptor transcription factors in mouse OB interneuron development. We observed that COUP-TFI is expressed in a gradient of low rostral to high caudal within the postnatal SVZ neural stem/progenitor cells. COUP-TFI is also expressed in a large number of migrating neuroblasts in the SVZ and RMS, and in mature interneurons in the OB. By contrast, very few COUP-TFII-expressing (+) cells exist in the SVZ-RMS-OB pathway. Conditional inactivation of COUP-TFI resulted in downregulation of tyrosine hydroxylase expression in the OB periglomerular cells and upregulation of COUP-TFII expression in the SVZ, RMS and OB deep granule cell layer. In COUP-TFI/COUP-TFII double conditional mutant SVZ, cell proliferation was increased through the upregulation of the proneural gene Ascl1. Furthermore, COUP-TFI/II-deficient neuroblasts had impaired migration, resulting in ectopic accumulation of calretinin (CR)+ and NeuN+ cells, and an increase in apoptotic cell death in the SVZ. Finally, we found that most Pax6+ and a subset of CR+ granular cells were lost in the OB. Taken together, these results suggest that COUP-TFI/II coordinately regulate the proliferation, migration and survival of a subpopulation of Pax6+ and CR+ granule cells in the OB.


Asunto(s)
Factor de Transcripción COUP II/metabolismo , Factor de Transcripción COUP I/metabolismo , Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Interneuronas/fisiología , Células-Madre Neurales/fisiología , Bulbo Olfatorio/citología , Animales , Bromodesoxiuridina , Movimiento Celular/fisiología , Colágeno , Combinación de Medicamentos , Galactósidos , Inmunohistoquímica , Indoles , Interneuronas/metabolismo , Laminina , Ratones , Microscopía Fluorescente , Proteoglicanos , Tamoxifeno , Tirosina 3-Monooxigenasa/metabolismo
12.
Hum Reprod ; 33(8): 1517-1527, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29982401

RESUMEN

STUDY QUESTION: How does hypoxia-mediated downregulation of chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII) promote angiogenesis in endometriosis? SUMMARY ANSWER: Suppression of COUP-TFII by hypoxia stimulates angiogenesis through induction of angiogenin (ANG). WHAT IS KNOWN ALREADY: The level of COUP-TFII is downregulated in endometriotic tissues, and downregulation of COUP-TFII contributes to the development of endometriosis. STUDY DESIGN, SIZE, DURATION: Twenty-seven patients of reproductive age with endometriosis were recruited in this study. Eutopic endometrial and ectopic endometriotic stromal cells were isolated, cultured and subjected to various treatments. PARTICIPANTS/MATERIALS, SETTING, METHODS: Microarray hybridization, quantitative RT-PCR, and Western blot were used to detect gene expression in normal and endometriotic samples. A luciferase reporter assay and chromatin immunoprecipitation in normoxia- or hypoxia-treated primary cultures of human endometrial stromal cells were performed. Tube formation analysis was performed using primary human umbilical vein endothelial cells (HUVECs). MAIN RESULTS AND THE ROLE OF CHANCE: Protein level of COUP-TFII was downregulated by hypoxia (P < 0.05, normoxia versus hypoxia). Loss of COUP-TFII increased the angiogenic capacity of endometrial stromal cells (P < 0.05, COUP-TFII knockdown versus knockdown control). A novel COUP-TFII target gene, ANG, was identified through microarray analysis. Chromatin immunoprecipitation and promoter activity assays demonstrated that the ANG promoter was bound and suppressed by COUP-TFII (P < 0.05, COUP-TFII overexpression versus empty vector). The levels of ANG mRNA and protein were elevated in ectopic endometriotic stromal cells and negatively correlated with COUP-TFII (P < 0.05, endometrial versus endometriotic tissues/stromal cells). Both knockdown and forced-expression of COUP-TFII further demonstrated that ANG expression and ANG-mediated angiogenic activity were negatively regulated by COUP-TFII (P < 0.05, COUP-TFII knockdown versus knockdown control, and COUP-TFII overexpression versus empty vector). LIMITATIONS, REASONS FOR CAUTION: This study was conducted in primary human endometrial stromal cell cultures and HUVECs, therefore, may not fully reflect the situation in vivo. LARGE SCALE DATA: The raw data were submitted to Gene Expression Omnibus (GSE107469). WIDER IMPLICATIONS OF THE FINDINGS: This is the first study to highlight that the aberrant expression of ANG in endometriotic lesions is mediated by hypoxia-suppressed COUP-TFII expression, which reveals an as yet unidentified molecular pathogenesis of endometriosis. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by research grants (MOST 105-2314-B-006-059-MY3 to M.H.W. and MOST 104-2320-B-006-036-MY3 to S.J.T.) from the Ministry of Science and Technology, Taiwan. The authors declare that there is no conflict of interest.


Asunto(s)
Factor de Transcripción COUP II/metabolismo , Endometriosis/metabolismo , Endometrio/irrigación sanguínea , Endometrio/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neovascularización Patológica , Comunicación Paracrina , Ribonucleasa Pancreática/metabolismo , Células del Estroma/metabolismo , Factor de Transcripción COUP II/genética , Estudios de Casos y Controles , Hipoxia de la Célula , Células Cultivadas , Microambiente Celular , Endometriosis/genética , Endometriosis/patología , Endometrio/patología , Femenino , Humanos , Cultivo Primario de Células , Ribonucleasa Pancreática/genética , Transducción de Señal , Células del Estroma/patología
13.
Proc Natl Acad Sci U S A ; 112(36): E4985-94, 2015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-26305926

RESUMEN

The preoptic area (POa) of the rostral diencephalon supplies the neocortex and the amygdala with GABAergic neurons in the developing mouse brain. However, the molecular mechanisms that determine the pathway and destinations of POa-derived neurons have not yet been identified. Here we show that Chicken ovalbumin upstream promoter transcription factor II (COUP-TFII)-induced expression of Neuropilin-2 (Nrp2) and its down-regulation control the destination of POa-derived GABAergic neurons. Initially, a majority of the POa-derived migrating neurons express COUP-TFII and form a caudal migratory stream toward the caudal subpallium. When a subpopulation of cells steers toward the neocortex, they exhibit decreased expression of COUP-TFII and Nrp2. The present findings show that suppression of COUP-TFII/Nrp2 changed the destination of the cells into the neocortex, whereas overexpression of COUP-TFII/Nrp2 caused cells to end up in the medial part of the amygdala. Taken together, these results reveal that COUP-TFII/Nrp2 is a molecular switch determining the pathway and destination of migrating GABAergic neurons born in the POa.


Asunto(s)
Encéfalo/metabolismo , Factor de Transcripción COUP II/metabolismo , Diencéfalo/metabolismo , Neuronas GABAérgicas/metabolismo , Neuropilina-2/metabolismo , Amígdala del Cerebelo/embriología , Amígdala del Cerebelo/metabolismo , Animales , Western Blotting , Encéfalo/embriología , Factor de Transcripción COUP II/genética , Movimiento Celular/genética , Diencéfalo/embriología , Neuronas GABAérgicas/citología , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Ratones Endogámicos ICR , Ratones Noqueados , Microscopía Confocal , Neocórtex/embriología , Neocórtex/metabolismo , Neuropilina-2/genética , Área Preóptica/embriología , Área Preóptica/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Técnicas de Cultivo de Tejidos
14.
Development ; 141(6): 1228-38, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24523456

RESUMEN

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.


Asunto(s)
Proteínas de Homeodominio/fisiología , Linfangiogénesis/fisiología , Proteínas Supresoras de Tumor/fisiología , Proteínas de Pez Cebra/fisiología , Pez Cebra/crecimiento & desarrollo , Animales , Animales Modificados Genéticamente , Factor de Transcripción COUP II/deficiencia , Factor de Transcripción COUP II/genética , Factor de Transcripción COUP II/metabolismo , Diferenciación Celular , Linaje de la Célula , Células Endoteliales/citología , Células Endoteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Linfangiogénesis/genética , Vasos Linfáticos/citología , Vasos Linfáticos/metabolismo , Ratones , Ratones Noqueados , Mutación , Factores de Transcripción SOXF/deficiencia , Factores de Transcripción SOXF/genética , Factores de Transcripción SOXF/metabolismo , Especificidad de la Especie , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Pez Cebra/genética , Pez Cebra/fisiología , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
15.
Development ; 141(5): 1120-8, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24550118

RESUMEN

Coordinated arterial-venous differentiation is crucial for vascular development and function. The origin of the cardinal vein (CV) in mammals is unknown, while conflicting theories have been reported in chick and zebrafish. Here, we provide the first molecular characterization of endothelial cells (ECs) expressing venous molecular markers, or venous-fated ECs, within the emergent dorsal aorta (DA). These ECs, expressing the venous molecular markers Coup-TFII and EphB4, cohabited the early DA with ECs expressing the arterial molecular markers ephrin B2, Notch and connexin 40. These mixed ECs in the early DA expressed either the arterial or venous molecular marker, but rarely both. Subsequently, the DA exhibited uniform arterial markers. Real-time imaging of mouse embryos revealed EC movement from the DA to the CV during the stage when venous-fated ECs occupied the DA. We analyzed mutants for EphB4, which encodes a receptor tyrosine kinase for the ephrin B2 ligand, as we hypothesized that ephrin B2/EphB4 signaling may mediate the repulsion of venous-fated ECs from the DA to the CV. Using an EC quantification approach, we discovered that venous-fated ECs increased in the DA and decreased in the CV in the mutants, whereas the rest of the ECs in each vessel were unaffected. This result suggests that the venous-fated ECs were retained in the DA and missing in the CV in the EphB4 mutant, and thus that ephrin B2/EphB4 signaling normally functions to clear venous-fated ECs from the DA to the CV by cell repulsion. Therefore, our cellular and molecular evidence suggests that the DA harbors venous progenitors that move to participate in CV formation, and that ephrin B2/EphB4 signaling regulates this aortic contribution to the mammalian CV.


Asunto(s)
Aorta/citología , Células Madre/citología , Venas/citología , Animales , Células Endoteliales/citología , Células Endoteliales/metabolismo , Ratones , Ratones Transgénicos , Neovascularización Fisiológica/fisiología , Transducción de Señal/fisiología , Células Madre/metabolismo
16.
Biochim Biophys Acta ; 1849(2): 201-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24878540

RESUMEN

Recent studies reveal that COUP-TF genes are essential for neural development, cardiovascular development, energy metabolism and adipogenesis, as well as for organogenesis of multiple systems. In this review, we mainly describe the COUP-TF genes, molecular mechanisms of COUP-TF action, and their crucial functions in the morphogenesis of the murine eye. Mutations of COUP-TF genes lead to the congenital coloboma and/or optic atrophy in both mouse and human, indicating that the study on COUP-TFs and the eye will benefit our understanding of the etiology of human ocular diseases. This article is part of a Special Issue entitled: Nuclear receptors in animal development.


Asunto(s)
Factores de Transcripción COUP/fisiología , Ojo/embriología , Organogénesis/genética , Animales , Factores de Transcripción COUP/genética , Drosophila/embriología , Drosophila/genética , Ojo/metabolismo , Oftalmopatías/genética , Humanos , Ratones/embriología , Ratones/genética , Xenopus/embriología , Xenopus/genética , Pez Cebra/embriología , Pez Cebra/genética
17.
Int J Cancer ; 139(7): 1574-85, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27193872

RESUMEN

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.


Asunto(s)
Proteína BRCA1/genética , Factor de Transcripción COUP II/genética , Carcinoma de Células Renales/genética , Neoplasias Renales/genética , Animales , Apoptosis/inmunología , Proteína BRCA1/biosíntesis , Factor de Transcripción COUP II/biosíntesis , Factor de Transcripción COUP II/deficiencia , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Proliferación Celular/genética , Técnicas de Silenciamiento del Gen , Genes BRCA1 , Xenoinjertos , Humanos , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Regulación hacia Arriba
18.
Am J Med Genet A ; 170(9): 2457-61, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27363585

RESUMEN

COUP-TFII (NR2F2) is mapped to the 15q26 deletion hotspot associated with the common and highly morbid congenital diaphragmatic hernia (CDH). Conditional homozygous deletions of COUP-TFII in mice result in diaphragmatic defects analogous to the human Bochdalek-type hernia phenotype. Despite evidence from animal models however, mutations in the coding sequence of COUP-TFII have not been reported in patients, prompting the speculation that additional coding or non-coding sequences in the 15q26 locus are necessary for diaphragmatic hernias to develop. In this report, we describe a case of a patient with a heterozygous de novo COUP-TFII frameshift mutation, presenting with CDH and an atrial septal defect. The p.Pro33AlafsTer77 mutation specifically disrupts protein isoform 1 which contains the DNA binding domain. In addition, we review other COUP-TFII sequence variations and deletions that have been described in cases of CDH. We conclude that COUP-TFII mutations can cause diaphragmatic hernias, and should be included in the differential diagnosis of CDH patients, particularly those with comorbid congenital heart defects. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Factor de Transcripción COUP II/genética , Mutación del Sistema de Lectura , Estudios de Asociación Genética , Hernias Diafragmáticas Congénitas/diagnóstico , Hernias Diafragmáticas Congénitas/genética , Fenotipo , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Alelos , Análisis Mutacional de ADN , Femenino , Genotipo , Humanos , Recién Nacido , Masculino , Linaje , Polimorfismo de Nucleótido Simple
19.
Dev Biol ; 390(2): 116-25, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24699544

RESUMEN

Initial embryonic determination of artery or vein identity is regulated by genetic factors that work in concert to specify the endothelial cell׳s (EC) fate, giving rise to two structurally unique components of the circulatory loop. The Shh/VEGF/Notch pathway is critical for arterial specification, while the orphan receptor nr2f2 (COUP-TFII) has been implicated in venous specification. Studies in mice have shown that nr2f2 is expressed in venous but not arterial ECs, and that it preferentially induces markers of venous cell fate. We have examined the role of nr2f2 during early arterial-venous development in the zebrafish trunk. We show that expression of a subset of markers of venous endothelial identity requires nr2f2, while the expression of nr2f2 itself requires sox7 and sox18 gene function. However, while sox7 and sox18 are expressed in both the cardinal vein and the dorsal aorta during early trunk development, nr2f2 is expressed only in the cardinal vein. We show that Notch signaling activity present in the dorsal aorta suppresses expression of nr2f2, restricting nr2f2-dependent promotion of venous differentiation to the cardinal vein.


Asunto(s)
Vasos Sanguíneos/embriología , Factor de Transcripción COUP II/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Receptores Notch/metabolismo , Factores de Transcripción SOXF/metabolismo , Transducción de Señal/fisiología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Factor de Transcripción COUP II/genética , Diferenciación Celular/fisiología , Clonación Molecular , Cartilla de ADN/genética , Regulación del Desarrollo de la Expresión Génica/genética , Inmunohistoquímica , Hibridación in Situ , Microscopía Confocal , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción SOXF/genética , Transgenes/genética , Venas/citología , Venas/embriología , Proteínas de Pez Cebra/genética
20.
Biol Reprod ; 91(1): 26, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24899578

RESUMEN

Testosterone production is dependent on cholesterol transport within the mitochondrial matrix, an essential step mediated by a protein complex containing the steroidogenic acute regulatory (STAR) protein. In steroidogenic Leydig cells, Star expression is hormonally regulated and involves several transcription factors. NR2F2 (COUP-TFII) is an orphan nuclear receptor that plays critical roles in cell differentiation and lineage determination. Conditional NR2F2 knockout prior to puberty leads to male infertility due to insufficient testosterone production, suggesting that NR2F2 could positively regulate steroidogenesis and Star expression. In this study we found that NR2F2 is expressed in the nucleus of some peritubular myoid cells and in interstitial cells, mainly in steroidogenically active adult Leydig cells. In MA-10 and MLTC-1 Leydig cells, small interfering RNA (siRNA)-mediated NR2F2 knockdown reduces basal steroid production without affecting hormone responsiveness. Consistent with this, we found that STAR mRNA and protein levels were reduced in NR2F2-depleted MA-10 and MLTC-1 cells. Transient transfections of Leydig cells revealed that a -986 bp mouse Star promoter construct was activated 3-fold by NR2F2. Using 5' progressive deletion constructs, we mapped the NR2F2-responsive element between -131 and -95 bp. This proximal promoter region contains a previously uncharacterized direct repeat 1 (DR1)-like element to which NR2F2 is recruited and directly binds. Mutations in the DR1-like element that prevent NR2F2 binding severely blunted NR2F2-mediated Star promoter activation. These data identify an essential role for the nuclear receptor NR2F2 as a direct activator of Star gene expression in Leydig cells, and thus in the control of steroid hormone biosynthesis.


Asunto(s)
Factor de Transcripción COUP II/metabolismo , Hormonas Esteroides Gonadales/biosíntesis , Células Intersticiales del Testículo/metabolismo , Fosfoproteínas/metabolismo , Animales , Factor de Transcripción COUP II/genética , Línea Celular , Masculino , Ratones , Fosfoproteínas/genética , Regiones Promotoras Genéticas , ARN Interferente Pequeño
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA