Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 193
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biol Reprod ; 110(4): 782-797, 2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38224314

RESUMEN

Defining features of polycystic ovary syndrome (PCOS) include elevated expression of steroidogenic genes, theca cell androgen biosynthesis, and peripheral levels of androgens. In previous studies, we identified vascular cell adhesion molecule 1 (VCAM1) as a selective androgen target gene in specific NR2F2/SF1 (+/+) theca cells. By deleting NR2F2 and VCAM1 selectively in CYP17A1 theca cells in mice, we documented that NR2F2 and VCAM1 impact distinct and sometimes opposing theca cell functions that alter ovarian follicular development in vivo: including major changes in ovarian morphology, steroidogenesis, gene expression profiles, immunolocalization images (NR5A1, CYP11A1, NOTCH1, CYP17A1, INSL3, VCAM1, NR2F2) as well as granulosa cell functions. We propose that theca cells impact follicle integrity by regulating androgen production and action, as well as granulosa cell differentiation/luteinization in response to androgens and gonadotropins that may underlie PCOS.


Asunto(s)
Factor de Transcripción COUP II , Síndrome del Ovario Poliquístico , Células Tecales , Molécula 1 de Adhesión Celular Vascular , Animales , Femenino , Ratones , Andrógenos/metabolismo , Factor de Transcripción COUP II/genética , Factor de Transcripción COUP II/metabolismo , Células de la Granulosa/metabolismo , Síndrome del Ovario Poliquístico/genética , Síndrome del Ovario Poliquístico/metabolismo , Células Tecales/metabolismo , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
2.
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
3.
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
4.
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
5.
Reproduction ; 164(2): 31-40, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35666805

RESUMEN

In brief: The insulin-like 3 (INSL3) hormone produced by Leydig cells is essential for proper male sex differentiation, but the regulation of Insl3 expression remains poorly understood. This study describes a new physical and functional cooperation between the nuclear receptors SF1 and COUP-TFII in Insl3 expression. Abstract: INSL3, a hormone abundantly produced by Leydig cells, is essential for testis descent during fetal life and bone metabolism in adults. The mechanisms regulating Insl3 expression in Leydig cells have been studied in several species but remain poorly understood. To date, only a handful of transcription factors are known to activate the Insl3 promoter and include the nuclear receptors AR, NUR77, COUP-TFII, and SF1, as well as the Krüppel-like factor KLF6. Some of these transcription factors are known to transcriptionally cooperate on the Insl3 promoter, but the mechanisms at play remain unknown. Here, we report that COUP-TFII and SF1 functionally cooperate on the Insl3 promoter from various species but not on the Inha, Akr1c14, Cyp17a1, Hsd3b1, Star, Gsta3, and Amhr2 promoters that are known to be regulated by COUP-TFII and/or SF1. The Insl3 promoter contains species-conserved binding sites for COUP-TFII (-91 bp) and SF1 (-134 bp). Mutation of either the COUP-TFII or the SF1 sequence had no impact on the COUP-TFII/SF1 cooperation, but the mutation of both binding sites abolished the cooperation. In agreement with this, we found that COUP-TFII and SF1 physically interact in Leydig cells. Finally, we report that the transcriptional cooperation is not limited to COUP-TFII and SF1 as it also occurred between all NR2F and NR5A family members. Our data provide new mechanistic insights into the cooperation between the orphan nuclear receptors COUP-TFII and SF1 in the regulation of Insl3 gene expression in Leydig cells.


Asunto(s)
Factor de Transcripción COUP II , Insulina , Células Intersticiales del Testículo , Proteínas , Factor Esteroidogénico 1 , Adulto , Sitios de Unión , Factor de Transcripción COUP II/genética , Factor de Transcripción COUP II/metabolismo , Humanos , Insulina/genética , Insulina/metabolismo , Células Intersticiales del Testículo/metabolismo , Masculino , Regiones Promotoras Genéticas , Proteínas/genética , Factor Esteroidogénico 1/genética , Factor Esteroidogénico 1/metabolismo , Testículo/metabolismo
6.
Circ Res ; 126(7): 875-888, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-32065070

RESUMEN

RATIONALE: Significant progress has revealed transcriptional inputs that underlie regulation of artery and vein endothelial cell fates. However, little is known concerning genome-wide regulation of this process. Therefore, such studies are warranted to address this gap. OBJECTIVE: To identify and characterize artery- and vein-specific endothelial enhancers in the human genome, thereby gaining insights into mechanisms by which blood vessel identity is regulated. METHODS AND RESULTS: Using chromatin immunoprecipitation and deep sequencing for markers of active chromatin in human arterial and venous endothelial cells, we identified several thousand artery- and vein-specific regulatory elements. Computational analysis revealed that NR2F2 (nuclear receptor subfamily 2, group F, member 2) sites were overrepresented in vein-specific enhancers, suggesting a direct role in promoting vein identity. Subsequent integration of chromatin immunoprecipitation and deep sequencing data sets with RNA sequencing revealed that NR2F2 regulated 3 distinct aspects related to arteriovenous identity. First, consistent with previous genetic observations, NR2F2 directly activated enhancer elements flanking cell cycle genes to drive their expression. Second, NR2F2 was essential to directly activate vein-specific enhancers and their associated genes. Our genomic approach further revealed that NR2F2 acts with ERG (ETS-related gene) at many of these sites to drive vein-specific gene expression. Finally, NR2F2 directly repressed only a small number of artery enhancers in venous cells to prevent their activation, including a distal element upstream of the artery-specific transcription factor, HEY2 (hes related family bHLH transcription factor with YRPW motif 2). In arterial endothelial cells, this enhancer was normally bound by ERG, which was also required for arterial HEY2 expression. By contrast, in venous endothelial cells, NR2F2 was bound to this site, together with ERG, and prevented its activation. CONCLUSIONS: By leveraging a genome-wide approach, we revealed mechanistic insights into how NR2F2 functions in multiple roles to maintain venous identity. Importantly, characterization of its role at a crucial artery enhancer upstream of HEY2 established a novel mechanism by which artery-specific expression can be achieved.


Asunto(s)
Arterias/metabolismo , Factor de Transcripción COUP II/genética , Células Endoteliales/metabolismo , Elementos de Facilitación Genéticos/genética , Perfilación de la Expresión Génica/métodos , Genómica/métodos , Venas/metabolismo , Arterias/citología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factor de Transcripción COUP II/metabolismo , Células Cultivadas , Inmunoprecipitación de Cromatina/métodos , Regulación de la Expresión Génica , Células HeLa , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Venas/citología
7.
J Periodontal Res ; 57(2): 316-323, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34910830

RESUMEN

BACKGROUND AND OBJECTIVE: This study aimed to investigate the correlation between chronic periodontitis (CP) and abnormal circular RNA (circRNA) expression and to identify the role of hsa_circ_0003948 in the progression of CP. METHODS: Next-generation sequencing was utilized to investigate abnormal expression of circRNA in gingival tissues from CP patients and healthy control subjects. Bioinformatics and luciferase reporting analyses were used to clarify the interactive relationship among circRNA, miRNA, and mRNA. Periodontal ligament cells (PDLCs) were employed to analyze proliferation and apoptosis after lipopolysaccharide (LPS) treatment using the cell counting kit 8 (CCK8) assay and flow cytometry detection. RESULTS: High-throughput sequencing and RT-qPCR analyses confirmed that hsa_circ_0003948 expression decreased dramatically in gingival samples of CP patients. Overexpression of hsa_circ_0003948 alleviated LPS-induced PDLC injury by regulating NR2F2/PTEN signaling. The miR-144-3p and NR2F2 were determined to be hsa_circ_0003948 downstream targets. NR2F2 downregulation or miR-144-3p overexpression reversed the protective effect of hsa_circ_0003948 in PDLCs after treatment with LPS. Upregulation of NR2F2 reversed the inhibitory effect of miR-144-3p on surviving PDLCs after LPS treatment. CONCLUSION: Overexpression of hsa_circ_0003948 exerts a protective effect in CP via miR-144-3p/NR2F2/PTEN signaling regulation.


Asunto(s)
Factor de Transcripción COUP II , Periodontitis Crónica , MicroARNs , Fosfohidrolasa PTEN , ARN Circular , Apoptosis/genética , Factor de Transcripción COUP II/genética , Factor de Transcripción COUP II/metabolismo , Proliferación Celular/genética , Periodontitis Crónica/genética , Periodontitis Crónica/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Fosfohidrolasa PTEN/genética , ARN Circular/genética , ARN Circular/metabolismo
8.
Int J Clin Oncol ; 27(12): 1891-1903, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36269529

RESUMEN

BACKGROUND: Multiple studies have revealed that long non-coding RNA (lncRNA) NR2F2-AS1 plays a role in affecting cancer cell proliferation and metastasis. Here, both in vitro and in vivo experiments were performed for investigating the function and mechanism of NR2F2-AS1 in human osteosarcoma (OS). METHODS: The NR2F2-AS1 level in human OS tissues and adjacent non-tumor tissues was examined by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). The NR2F2-AS1 overexpression model was constructed in OS cells, then cell proliferation, invasion, and apoptosis were monitored. The OS xenograft model was established in nude mice using NR2F2-AS1-overexpressed OS cells. The downstream target genes of NR2F2-AS1 were predicted. qRT-PCR and Western blot were implemented to validate the profiles of miR-425-5p and HMGB2. The targeting link between NR2F2-AS1 and miR-425-5p, miR-425-5p and HMGB2 was further probed by dual-luciferase reporter experiment. RESULTS: In comparison to adjacent non-tumor tissues, OS tissues showed upregulated NR2F2-AS1 expression. Higher NR2F2-AS1 level was predominantly correlated with worse clinical stages. In vivo and in vitro tests corroborated that NR2F2-AS1 overexpression spurred OS cell proliferation, growth, invasion, and choked apoptosis. Mechanistically, NR2F2-AS1 hampered miR-425-5p expression as its competitive endogenous RNA (ceRNA). Thus, NR2F2-AS1 facilitated the HMGB2 expression. However, miR-425-5p inhibited HMGB2 expression by targeting the latter. CONCLUSION: NR2F2-AS1 expedited the evolution of OS by elevating HMGB2 levels through sponging miR-425-5p. The NR2F2-AS1/miR-425-5p/HMGB2 regulatory axis is a promising target in treating human OS.


Asunto(s)
Neoplasias Óseas , Proteína HMGB2 , MicroARNs , Osteosarcoma , ARN Largo no Codificante , Animales , Humanos , Ratones , Neoplasias Óseas/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Factor de Transcripción COUP II/genética , Factor de Transcripción COUP II/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Proteína HMGB2/genética , Proteína HMGB2/metabolismo , Ratones Desnudos , MicroARNs/genética , MicroARNs/metabolismo , Osteosarcoma/genética , Osteosarcoma/patología , ARN Largo no Codificante/genética , Factores de Transcripción/genética
9.
Mediators Inflamm ; 2022: 8373389, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36081650

RESUMEN

Diabetes is well recognized to increase the risk of heart failure, which is associated with higher mortality and morbidity. It is important for the development of novel therapeutic methods targeting heart failure in diabetic patients. Ferroptosis, an iron-dependent regulated cell death, has been implicated in the progression of diabetes-induced heart failure (DIHF). This study was designed to investigate the contribution of Nr2f2 to the activation of ferroptosis and mitochondrial dysfunction in DIHF. We established a diabetic model by a high-fat feeding diet combined with an intraperitoneal injection of streptozotocin. After 16 weeks, Nr2f2 expression was increased in heart tissue of DIHF mice. In vivo, DIHF mice overexpressing Nr2f2 (AAV9-cTNT-Nr2f2) exhibited severe heart failure and enhanced cardiac ferroptosis compared with DIHF control mice (AAV9-cTNT-ctrl), accompanied by mitochondrial dysfunction and aggravated oxidative stress reaction. In vitro, Nr2f2 knockdown ameliorated ferroptosis and mitochondrial dysfunction by negatively regulating PGC-1α, a crucial metabolic regulator. PGC-1α knockdown counteracted the protective effect of Nr2f2 knockdown. These data suggest that Nr2f2 promotes heart failure and ferroptosis in DIHF by modulating the PGC-1α signaling. Our study provides a new idea for the treatment of diabetes-induced heart failure.


Asunto(s)
Factor de Transcripción COUP II , Diabetes Mellitus , Ferroptosis , Insuficiencia Cardíaca , Animales , Factor de Transcripción COUP II/genética , Factor de Transcripción COUP II/metabolismo , Diabetes Mellitus/metabolismo , Insuficiencia Cardíaca/metabolismo , Ratones , Mitocondrias/metabolismo , Estrés Oxidativo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Transducción de Señal
10.
PLoS Genet ; 15(2): e1007962, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30721228

RESUMEN

Multiple syndromes share congenital heart and craniofacial muscle defects, indicating there is an intimate relationship between the adjacent cardiac and pharyngeal muscle (PM) progenitor fields. However, mechanisms that direct antagonistic lineage decisions of the cardiac and PM progenitors within the anterior mesoderm of vertebrates are not understood. Here, we identify that retinoic acid (RA) signaling directly promotes the expression of the transcription factor Nr2f1a within the anterior lateral plate mesoderm. Using zebrafish nr2f1a and nr2f2 mutants, we find that Nr2f1a and Nr2f2 have redundant requirements restricting ventricular cardiomyocyte (CM) number and promoting development of the posterior PMs. Cre-mediated genetic lineage tracing in nr2f1a; nr2f2 double mutants reveals that tcf21+ progenitor cells, which can give rise to ventricular CMs and PM, more frequently become ventricular CMs potentially at the expense of posterior PMs in nr2f1a; nr2f2 mutants. Our studies reveal insights into the molecular etiology that may underlie developmental syndromes that share heart, neck and facial defects as well as the phenotypic variability of congenital heart defects associated with NR2F mutations in humans.


Asunto(s)
Factor de Transcripción COUP II/metabolismo , Proteínas de Unión al ADN/metabolismo , Miocitos Cardíacos/metabolismo , Músculos Faríngeos/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Tipificación del Cuerpo/genética , Factor de Transcripción COUP II/genética , Linaje de la Célula/genética , Anomalías Craneofaciales/embriología , Anomalías Craneofaciales/genética , Proteínas de Unión al ADN/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/genética , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/embriología , Ventrículos Cardíacos/metabolismo , Humanos , Mesodermo/citología , Mesodermo/embriología , Mesodermo/metabolismo , Modelos Animales , Mutación , Miocitos Cardíacos/citología , Músculos Faríngeos/citología , Músculos Faríngeos/embriología , Regiones Promotoras Genéticas , Transducción de Señal , Factores de Transcripción/genética , Tretinoina/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
11.
Am J Hum Genet ; 102(3): 487-493, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29478779

RESUMEN

Emerging evidence from murine studies suggests that mammalian sex determination is the outcome of an imbalance between mutually antagonistic male and female regulatory networks that canalize development down one pathway while actively repressing the other. However, in contrast to testis formation, the gene regulatory pathways governing mammalian ovary development have remained elusive. We performed exome or Sanger sequencing on 79 46,XX SRY-negative individuals with either unexplained virilization or with testicular/ovotesticular disorders/differences of sex development (TDSD/OTDSD). We identified heterozygous frameshift mutations in NR2F2, encoding COUP-TF2, in three children. One carried a c.103_109delGGCGCCC (p.Gly35Argfs∗75) mutation, while two others carried a c.97_103delCCGCCCG (p.Pro33Alafs∗77) mutation. In two of three children the mutation was de novo. All three children presented with congenital heart disease (CHD), one child with congenital diaphragmatic hernia (CDH), and two children with blepharophimosis-ptosis-epicanthus inversus syndrome (BPES). The three children had androgen production, virilization of external genitalia, and biochemical or histological evidence of testicular tissue. We demonstrate a highly significant association between the NR2F2 loss-of-function mutations and this syndromic form of DSD (p = 2.44 × 10-8). We show that COUP-TF2 is highly abundant in a FOXL2-negative stromal cell population of the fetal human ovary. In contrast to the mouse, these data establish COUP-TF2 as a human "pro-ovary" and "anti-testis" sex-determining factor in female gonads. Furthermore, the data presented here provide additional evidence of the emerging importance of nuclear receptors in establishing human ovarian identity and indicate that nuclear receptors may have divergent functions in mouse and human biology.


Asunto(s)
Trastornos del Desarrollo Sexual 46, XX/genética , Factor de Transcripción COUP II/genética , Mutación con Pérdida de Función/genética , Testículo/anomalías , Testículo/crecimiento & desarrollo , Secuencia de Aminoácidos , Secuencia de Bases , Factor de Transcripción COUP II/química , Niño , Femenino , Proteína Forkhead Box L2/metabolismo , Mutación del Sistema de Lectura/genética , Heterocigoto , Humanos , Masculino , Ovario/crecimiento & desarrollo , Ovario/metabolismo , Fenotipo
12.
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
13.
Haematologica ; 106(2): 474-482, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32107331

RESUMEN

The human fetal γ-globin gene is repressed in the adult stage through complex regulatory mechanisms involving transcription factors and epigenetic modifiers. Reversing γ-globin repression, or maintaining its expression by manipulating regulatory mechanisms, has become a major clinical goal in the treatment of ß-hemoglobinopathies. Here, we identify the orphan nuclear receptor Coup-TFII (NR2F2/ARP-1) as an embryonic/fetal stage activator of γ-globin expression. We show that Coup-TFII is expressed in early erythropoiesis of yolk sac origin, together with embryonic/fetal globins. When overexpressed in adult cells (including peripheral blood cells from human healthy donors and ß039 thalassemic patients) Coup-TFII activates the embryonic/fetal globins genes, overcoming the repression imposed by the adult erythroid environment. Conversely, the knock-out of Coup-TFII increases the ß/γ+ß globin ratio. Molecular analysis indicates that Coup-TFII binds in vivo to the ß-locus and contributes to its conformation. Overall, our data identify Coup-TFII as a specific activator of the γ-globin gene.


Asunto(s)
Receptores Nucleares Huérfanos , gamma-Globinas , Factor de Transcripción COUP II/genética , Factor de Transcripción COUP II/metabolismo , Proteínas Portadoras/genética , Humanos , Regiones Promotoras Genéticas , gamma-Globinas/genética
14.
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
15.
Int J Mol Sci ; 22(11)2021 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-34070923

RESUMEN

Increasing numbers of miRNAs have been observed as oncogenes or tumor suppressors in colorectal cancer (CRC). It was recently reported that hsa-miR-106b-5p (miR-106b) promoted CRC cell migration and invasion. However, there were also studies showing contradictory results. Therefore, in the present study, we further explore the role of miR-106b and its downstream networks in the carcinogenesis of CRC. We observed that the expression of miR-106b is significantly increased in Pan-Cancer and CRC tissues compared with normal tissues from The Cancer Genome Atlas (TCGA) database. Furthermore, we used Transwell, Cell Counting Kit-8, and colony formation assays to clarify that miR-106b promotes the migratory, invasive, and proliferative abilities of CRC cells. For the first time, we systematically screened the target mRNAs and lncRNAs of miR-106b using TCGA database and the bioinformatics algorithms. Dual-luciferase reporter assay confirmed that NR2F2-AS1 and PLEKHO2 are the direct targets of miR-106b. Furthermore, NR2F2-AS1 acts as a competing endogenous RNA (ceRNA) to regulate PLEKHO2 expression by sponging miR-106b. The results of Gene set enrichment analysis (GSEA) and Western blot indicated that they play important roles in CRC progression by regulating MAPK pathway. Thus, miR-106b/NR2F2-AS1/PLEKHO2/MAPK signaling axis may suggest the potential usage in CRC treatment.


Asunto(s)
Factor de Transcripción COUP II/genética , Carcinogénesis/genética , Neoplasias Colorrectales/genética , Péptidos y Proteínas de Señalización Intercelular/genética , MicroARNs/genética , ARN Largo no Codificante/genética , Atlas como Asunto , Secuencia de Bases , Sitios de Unión , Factor de Transcripción COUP II/metabolismo , Carcinogénesis/metabolismo , Carcinogénesis/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Bases de Datos Genéticas , Regulación Neoplásica de la Expresión Génica , Genes Reporteros , Células HCT116 , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Luciferasas/genética , Luciferasas/metabolismo , Sistema de Señalización de MAP Quinasas , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo
16.
Biochem Biophys Res Commun ; 529(4): 1209-1215, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32819587

RESUMEN

The nuclear receptor subfamily 2 group F member 2 (NR2F2) gene encodes a ligand-inducible transcription factor involved in angiogenesis and heart development. This study aimed to elucidate the molecular mechanism of epigenetic regulation of NR2F2 in tetralogy of Fallot (TOF) development. In the present study, immunohistochemical staining showed that NR2F2 protein expression was significantly higher in the right ventricular outflow tract (RVOT) tissues of TOF cases compared with controls. The methylation status of the CpG island shore (CGIS) of the NR2F2 gene was decreased in TOF cases, and the CpG site 3 in the CGIS region of NR2F2 promoter was a differential methylation site. Furthermore, the methylation level of the CpG site 3 and the NR2F2 protein expression were significantly negatively correlated in TOF patients. In vitro functional analysis revealed that RXRα could upregulate the NR2F2 gene by directly binding to the CGIS in the NR2F2 promoter, while hypomethylation of the NR2F2 promoter via treatment with 5-azacytidine influenced the affinity of RXRα to its binding sites, as shown by ChIP-qPCR. These findings suggest that promoter hypomethylation activates NR2F2 by enhancing RXRα binding to NR2F2 CGIS in the development of TOF.


Asunto(s)
Factor de Transcripción COUP II/metabolismo , Islas de CpG/genética , Metilación de ADN/genética , Miocardio/metabolismo , Miocardio/patología , Receptor alfa X Retinoide/metabolismo , Tetralogía de Fallot/genética , Adolescente , Animales , Secuencia de Bases , Factor de Transcripción COUP II/genética , Estudios de Casos y Controles , Niño , Preescolar , Femenino , Células HEK293 , Células HeLa , Humanos , Masculino , Ratones , Regiones Promotoras Genéticas/genética , Unión Proteica , Reproducibilidad de los Resultados , Transcripción Genética
17.
Development ; 144(15): 2837-2851, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28694260

RESUMEN

Distinct cortical interneuron (CIN) subtypes have unique circuit functions; dysfunction in specific subtypes is implicated in neuropsychiatric disorders. Somatostatin- and parvalbumin-expressing (SST+ and PV+) interneurons are the two major subtypes generated by medial ganglionic eminence (MGE) progenitors. Spatial and temporal mechanisms governing their cell-fate specification and differential integration into cortical layers are largely unknown. We provide evidence that Coup-TF1 and Coup-TF2 (Nr2f1 and Nr2f2) transcription factor expression in an arc-shaped progenitor domain within the MGE promotes time-dependent survival of this neuroepithelium and the time-dependent specification of layer V SST+ CINs. Coup-TF1 and Coup-TF2 autonomously repress PV+ fate in MGE progenitors, in part through directly driving Sox6 expression. These results have identified, in mouse, a transcriptional pathway that controls SST-PV fate.


Asunto(s)
Factor de Transcripción COUP II/metabolismo , Factor de Transcripción COUP I/metabolismo , Interneuronas/metabolismo , Neocórtex/citología , Animales , Factor de Transcripción COUP I/genética , Factor de Transcripción COUP II/genética , Células Cultivadas , Inmunoprecipitación de Cromatina , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Inmunohistoquímica , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Parvalbúminas/genética , Parvalbúminas/metabolismo , Factores de Transcripción SOXD/genética , Factores de Transcripción SOXD/metabolismo , Somatostatina/genética , Somatostatina/metabolismo
18.
BMC Cancer ; 20(1): 626, 2020 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-32631390

RESUMEN

BACKGROUND: The failure of treatment for breast cancer usually results from distant metastasis in which the epithelial-mesenchymal transition (EMT) plays a critical role. Hyperinsulinemia, the hallmark of Type 2 diabetes mellitus (T2DM), has been regarded as a key risk factor for the progression of breast cancer. Nuclear receptor subfamily 2, group F, member 2 (NR2F2) has been implicated in the development of breast cancer, however its contribution to insulin-induced EMT in breast cancer remains unclear. METHODS: Overexpression and knockdown of NR2F2 were used in two breast cancer cell lines, MCF-7 and MDA-MB-231 to investigate potential mechanisms by which NR2F2 leads to insulin-mediated EMT. To elucidate the effects of insulin and signaling events following NR2F2 overexpression and knockdown, Cells' invasion and migration capacity and changes of NR2F2, E-cadherin, N-cadherin and vimentin were investigated by real-time RT-PCR and western blot. RESULTS: Insulin stimulation of these cells increased NR2F2 expression levels and promoted cell invasion and migration accompanied by alterations in EMT-related molecular markers. Overexpression of NR2F2 and NR2F2 knockdown demonstrated that NR2F2 expression was positively correlated with cell invasion, migration and the expression of N-cadherin and vimentin. In contrast, NR2F2 had an inverse correlation with E-cadherin expression. In MDA-MB-231, both insulin-induced cell invasion and migration and EMT-related marker alteration were abolished by NR2F2 knockdown. CONCLUSIONS: These results suggest that NR2F2 plays a critical role in insulin-mediated breast cancer cell invasion, migration through its effect on EMT.


Asunto(s)
Neoplasias de la Mama/genética , Factor de Transcripción COUP II/metabolismo , Transición Epitelial-Mesenquimal/genética , Insulina/metabolismo , Neoplasias de la Mama/patología , Factor de Transcripción COUP II/genética , Movimiento Celular/genética , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/epidemiología , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Hiperinsulinismo/sangre , Hiperinsulinismo/epidemiología , Hiperinsulinismo/etiología , Hiperinsulinismo/metabolismo , Insulina/sangre , Células MCF-7 , Invasividad Neoplásica/genética , Factores de Riesgo
19.
Aging Male ; 23(5): 1073-1079, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31566058

RESUMEN

We in this study investigated the role of lncRNA NR2F2-AS1 in prostate carcinoma (PC). We showed that NR2F2-AS1 was upregulated in PC and positively correlated with CDK4. In PC cells, NR2F2-AS1 overexpression led to upregulated, while NR2F2-AS1 siRNA silencing led to downregulated CDK4. Follow-up study revealed that high levels of NR2F2-AS1 and CDK4 in PC tissues were closely correlated with the poor survival of PC patients. Cell proliferation data showed that NR2F2-AS1 overexpression led to increased, while NR2F2-AS1 siRNA silencing led to proliferation rate of PC cells. Moreover, NR2F2-AS1 also showed positive effects on cell cycle progression. In addition, CDK4 overexpression reduced the effects of NR2F2-AS1 siRNA silencing. Therefore, NR2F2-AS1 positively regulates CDK4 to promote cancer cell proliferation in PC.


Asunto(s)
Carcinoma , ARN Largo no Codificante , Factor de Transcripción COUP II/genética , Línea Celular Tumoral , Proliferación Celular , Quinasa 4 Dependiente de la Ciclina/genética , Estudios de Seguimiento , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Próstata , ARN Largo no Codificante/genética
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA