Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Dev Biol ; 475: 131-144, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33484706

RESUMEN

Coup-TF, a member of the nuclear receptor super-family, is present in the pool of maternal mRNAs and proteins in the sea urchin egg. The presence of this protein seems to be essential for the execution of the early developmental program, leading to all three embryonic layers. Our results demonstrate that Pl-Coup-TF morphants, i.e. Pl-Coup-TF morpholino knockdown embryos, resemble blastulae that lack archenteron at 24 hpf (hours post fertilization), a stage at which normal embryos reach the end of gastrulation in Paracentrotus lividus. At 48 hpf, when normal embryos reach the pluteus larva stage, the morphants are seemingly underdeveloped and lack the characteristic skeletal rods. Nevertheless, the morphant embryos express vegetal endomesodermal marker genes, such as Pl-Blimp1, Pl-Endo16, Pl-Alx1 and Pl-Tbr as judged by in situ hybridization experiments. The anterior neuroectoderm genes, Pl-FoxQ2, Pl-Six3 and Pl-Pax6, are also expressed in the morphant embryos, but Pl-Hbn and Pl-Fez mRNAs, which encode proteins significant for the differentiation of serotonergic neurons, are not detected. Consequently, Pl-Coup-TF morphants at 48 hpf lack serotonergic neurons, whereas normal 48 hpf plutei exhibit the formation of two bilateral pairs of such neurons in the apical organ. Furthermore, genes indicative of the ciliary band formation, Pl-Hnf6, Pl-Dri, Pl-FoxG and Pl-Otx, are not expressed in Pl-Coup-TF morphants, suggesting the disruption of this neurogenic territory as well. In addition, the Pl-SynB gene, a marker of differentiated neurons, is silent leading to the hypothesis that Pl-Coup-TF morphants might lack all types of neurons. On the contrary, the genes expressing signaling molecules, which establish the ventral/dorsal axis, Pl-Nodal and Pl-Lefty show the characteristic ventral lateral expression pattern, Pl-Bmp2/4, which activates the dorsal ectoderm GRN is down-regulated and Pl-Chordin is aberrantly over-expressed in the entire ectoderm. The identity of ectodermal cells in Pl-Coup-TF morphant embryos, was probed for expression of the ventral marker Pl-Gsc which was over-expressed and dorsal markers, Pl-IrxA and Pl-Hox7, which were silent. Therefore, we propose that maternal Pl-Coup-TF is essential for correct dissemination of the early embryonic signaling along both animal/vegetal and ventral/dorsal axes. Limiting Pl-Coup-TF's quantity, results in an embryo without digestive and nervous systems, skeleton and ciliary band that cannot survive past the initial 48 h of development.


Asunto(s)
Tipificación del Cuerpo/genética , Factores de Transcripción COUP/metabolismo , Paracentrotus/embriología , Animales , Blástula/metabolismo , Factores de Transcripción COUP/genética , Factores de Transcripción COUP/fisiología , Diferenciación Celular/genética , Ectodermo/metabolismo , Embrión no Mamífero/metabolismo , Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/genética , Factor de Apareamiento/genética , Factor de Apareamiento/metabolismo , Placa Neural/metabolismo , Paracentrotus/genética , Erizos de Mar/embriología , Erizos de Mar/metabolismo , Transducción de Señal/fisiología
2.
Curr Top Dev Biol ; 125: 275-301, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28527575

RESUMEN

COUP-TFI and -TFII are members of the steroid/thyroid nuclear receptor superfamily. Recent clinical studies reveal that COUP-TFI gene mutations are associated with Bosch-Boonstra-Schaaf optic atrophy syndrome displaying symptoms of optic atrophy, intellectual disability, hypotonia, seizure, autism spectrum disorders, oromotor dysfunction, thin corpus callosum, or hearing defects, and COUP-TFII gene mutations lead to congenital heart defects and/or congenital diaphragmatic hernia with developmental delay and mental defects. In this review, we first describe the functions of COUP-TF genes in the morphogenesis of mouse forebrain including cerebral cortex, hippocampus, amygdala complex, hypothalamus, and cortical interneuron. Then, we address their roles in the development of cerebellum, glial cells, neural crest cells, and adult neuronal stem cells. Clearly, the investigations on the functions of COUP-TF genes in the developing mouse central nervous system will benefit not only the understanding of neurodevelopment, but also the etiology of human mental diseases.


Asunto(s)
Factores de Transcripción COUP/genética , Sistema Nervioso Central , Regulación del Desarrollo de la Expresión Génica , Animales , Encefalopatías/genética , Sistema Nervioso Central/embriología , Sistema Nervioso Central/fisiología , Modelos Animales de Enfermedad , Humanos , Ratones , Organogénesis
3.
Adv Exp Med Biol ; 949: 287-310, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27714695

RESUMEN

During development and through adulthood, differentiation of diverse cell types is controlled by specific genetic and molecular programs for which transcription factors are master regulators of gene expression. Here, we present an overview of the role of nuclear receptors and their selective pharmacological modulators in oligodendrocytes linage, their role in myelination and remyelination and their potential use as a therapeutic strategy for demyelinating diseases. We discuss several aspects of nuclear receptors including: (1) the biochemistry of nuclear receptors superfamily; (2) their role on stem cells physiology, focusing in differentiation and cell removal; (3) the role of nuclear receptor in the oligodendrocytes cell linage, from oligodendrocyte progenitors cells to mature myelinating cells; and (4) the therapeutics opportunities of nuclear receptors for specific demyelinating diseases.


Asunto(s)
Adrenoleucodistrofia/genética , Enfermedad de Alzheimer/genética , Oligodendroglía/metabolismo , Receptores de Ácido Retinoico/genética , Células Madre/metabolismo , Adrenoleucodistrofia/tratamiento farmacológico , Adrenoleucodistrofia/metabolismo , Adrenoleucodistrofia/patología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Factores de Transcripción COUP/agonistas , Factores de Transcripción COUP/genética , Factores de Transcripción COUP/metabolismo , Diferenciación Celular , Linaje de la Célula , Drogas en Investigación/uso terapéutico , Regulación de la Expresión Génica , Humanos , Receptores X del Hígado/agonistas , Receptores X del Hígado/genética , Receptores X del Hígado/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Oligodendroglía/efectos de los fármacos , Oligodendroglía/patología , Receptores Nucleares Huérfanos , Receptores Activados del Proliferador del Peroxisoma/agonistas , Receptores Activados del Proliferador del Peroxisoma/genética , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Ácido Retinoico/agonistas , Receptores de Ácido Retinoico/metabolismo , Células Madre/efectos de los fármacos , Células Madre/patología
4.
J Steroid Biochem Mol Biol ; 157: 7-12, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26658017

RESUMEN

Cell fate specification is a critical process to generate cells with a wide range of characteristics from stem and progenitor cells. Emerging evidence demonstrates that the orphan nuclear receptor COUP-TFII serves as a key regulator in determining the cell identity during embryonic development. The present review summarizes our current knowledge on molecular mechanisms by which COUP-TFII employs to define the cell fates, with special emphasis on cardiovascular and renal systems. These novel insights pave the road for future studies of regenerative medicine.


Asunto(s)
Vasos Sanguíneos/citología , Factores de Transcripción COUP/metabolismo , Riñón/citología , Miocardio/citología , Células Madre/citología , Células Madre/fisiología , Animales , Factores de Transcripción COUP/genética , Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Atrios Cardíacos/citología , Atrios Cardíacos/embriología , Humanos , Riñón/embriología
5.
Cell Rep ; 12(12): 2072-85, 2015 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-26387951

RESUMEN

Nuclear receptor subfamily 2, group F, member 6 (NR2F6) is an orphan member of the nuclear receptor superfamily. Here, we show that genetic ablation of Nr2f6 significantly improves survival in the murine transgenic TRAMP prostate cancer model. Furthermore, Nr2f6(-/-) mice spontaneously reject implanted tumors and develop host-protective immunological memory against tumor rechallenge. This is paralleled by increased frequencies of both CD4(+) and CD8(+) T cells and higher expression levels of interleukin 2 and interferon γ at the tumor site. Mechanistically, CD4(+) and CD8(+) T cell-intrinsic NR2F6 acts as a direct repressor of the NFAT/AP-1 complex on both the interleukin 2 and the interferon γ cytokine promoters, attenuating their transcriptional thresholds. Adoptive transfer of Nr2f6-deficient T cells into tumor-bearing immunocompetent mice is sufficient to delay tumor outgrowth. Altogether, this defines NR2F6 as an intracellular immune checkpoint in effector T cells, governing the amplitude of anti-cancer immunity.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Factores de Transcripción COUP/genética , Vigilancia Inmunológica , Inmunoterapia Adoptiva/métodos , Neoplasias de la Próstata/terapia , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/trasplante , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/trasplante , Factores de Transcripción COUP/deficiencia , Factores de Transcripción COUP/inmunología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Memoria Inmunológica , Interferón gamma/agonistas , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-2/agonistas , Interleucina-2/genética , Interleucina-2/inmunología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/mortalidad , Proteínas Represoras , Transducción de Señal , Análisis de Supervivencia , Factor de Necrosis Tumoral alfa/agonistas , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
6.
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
7.
PLoS One ; 9(11): e109274, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25386650

RESUMEN

Coup-TF, an orphan member of the nuclear receptor super family, has a fundamental role in the development of metazoan embryos. The study of the gene's regulatory circuit in the sea urchin embryo will facilitate the placement of this transcription factor in the well-studied embryonic Gene Regulatory Network (GRN). The Paracentrotus lividus Coup-TF gene (PlCoup-TF) is expressed throughout embryonic development preferentially in the oral ectoderm of the gastrula and the ciliary band of the pluteus stage. Two overlapping λ genomic clones, containing three exons and upstream sequences of PlCoup-TF, were isolated from a genomic library. The transcription initiation site was determined and 5' deletions and individual segments of a 1930 bp upstream region were placed ahead of a GFP reporter cassette and injected into fertilized P.lividus eggs. Module a (-532 to -232), was necessary and sufficient to confer ciliary band expression to the reporter. Comparison of P.lividus and Strongylocentrotus purpuratus upstream Coup-TF sequences, revealed considerable conservation, but none within module a. 5' and internal deletions into module a, defined a smaller region that confers ciliary band specific expression. Putative regulatory cis-acting elements (RE1, RE2 and RE3) within module a, were specifically bound by proteins in sea urchin embryonic nuclear extracts. Site-specific mutagenesis of these elements resulted in loss of reporter activity (RE1) or ectopic expression (RE2, RE3). It is proposed that sea urchin transcription factors, which bind these three regulatory sites, are necessary for spatial and quantitative regulation of the PlCoup-TF gene at pluteus stage sea urchin embryos. These findings lead to the future identification of these factors and to the hierarchical positioning of PlCoup-TF within the embryonic GRN.


Asunto(s)
Factores de Transcripción COUP/genética , Regulación del Desarrollo de la Expresión Génica/genética , Paracentrotus/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Sitio de Iniciación de la Transcripción , Animales , Secuencia de Bases , Clonación Molecular , Proteínas de Unión al ADN/genética , Ensayo de Cambio de Movilidad Electroforética , Embrión no Mamífero , Paracentrotus/embriología , Receptores Citoplasmáticos y Nucleares/genética , Eliminación de Secuencia/genética , Strongylocentrotus purpuratus/genética , Iniciación de la Transcripción Genética
8.
Cell Commun Signal ; 12: 38, 2014 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-24919548

RESUMEN

Members of the evolutionarily conserved family of the chicken ovalbumin upstream promoter transcription factor NR2F/COUP-TF orphan receptors have been implicated in lymphocyte biology, ranging from activation to differentiation and elicitation of immune effector functions. In particular, a CD4+ T cell intrinsic and non-redundant function of NR2F6 as a potent and selective repressor of the transcription of the pro-inflammatory cytokines interleukin (Il) 2, interferon y (ifng) and consequently of T helper (Th)17 CD4+ T cell-mediated autoimmune disorders has been discovered. NR2F6 serves as an antigen receptor signaling threshold-regulated barrier against autoimmunity where NR2F6 is part of a negative feedback loop that limits inflammatory tissue damage induced by weakly immunogenic antigens such as self-antigens. Under such low affinity antigen receptor stimulation, NR2F6 appears as a prototypical repressor that functions to "lock out" harmful Th17 lineage effector transcription. Mechanistically, only sustained high affinity antigen receptor-induced protein kinase C (PKC)-mediated phosphorylation has been shown to inactivate NR2F6, thereby displacing pre-bound NR2F6 from the DNA and, subsequently, allowing for robust NFAT/AP-1- and RORγt-mediated cytokine transcription. The NR2F6 target gene repertoire thus identifies a general anti-inflammatory gatekeeper role for this orphan receptor. Investigating these signaling pathway(s) will enable a greater knowledge of the genetic, immune, and environmental mechanisms that lead to chronic inflammation and of certain autoimmune disorders in a given individual.


Asunto(s)
Factores de Transcripción COUP/metabolismo , Células Th17/metabolismo , Animales , Autoinmunidad , Factores de Transcripción COUP/química , Factores de Transcripción COUP/genética , Citocinas/genética , Citocinas/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Células Th17/inmunología , Transcripción Genética
9.
PLoS One ; 9(1): e85594, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24465611

RESUMEN

Nuclear hormone receptors (NHRs) are transcription factors that regulate carbohydrate and lipid metabolism, immune responses, and inflammation. Although several NHRs, including peroxisome proliferator-activated receptor-γ (PPARγ) and PPARα, demonstrate a renoprotective effect in the context of diabetic nephropathy (DN), the expression and role of other NHRs in the kidney are still unrecognized. To investigate potential roles of NHRs in the biology of the kidney, we used quantitative real-time polymerase chain reaction to profile the expression of all 49 members of the mouse NHR superfamily in mouse kidney tissue (C57BL/6 and db/m), and cell lines of mesangial (MES13), podocyte (MPC), proximal tubular epithelial (mProx24) and collecting duct (mIMCD3) origins in both normal and high-glucose conditions. In C57BL/6 mouse kidney cells, hepatocyte nuclear factor 4α, chicken ovalbumin upstream promoter transcription factor II (COUP-TFII) and COUP-TFIII were highly expressed. During hyperglycemia, the expression of the NHR 4A subgroup including neuron-derived clone 77 (Nur77), nuclear receptor-related factor 1, and neuron-derived orphan receptor 1 significantly increased in diabetic C57BL/6 and db/db mice. In renal cell lines, PPARδ was highly expressed in mesangial and proximal tubular epithelial cells, while COUP-TFs were highly expressed in podocytes, proximal tubular epithelial cells, and collecting duct cells. High-glucose conditions increased the expression of Nur77 in mesangial and collecting duct cells, and liver x receptor α in podocytes. These data demonstrate NHR expression in mouse kidney cells and cultured renal cell lines and suggest potential therapeutic targets in the kidney for the treatment of DN.


Asunto(s)
Nefropatías Diabéticas/genética , Expresión Génica , Riñón/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Animales , Factor de Transcripción COUP II/genética , Factor de Transcripción COUP II/metabolismo , Factores de Transcripción COUP/genética , Factores de Transcripción COUP/metabolismo , Línea Celular , Células Cultivadas , Nefropatías Diabéticas/metabolismo , Riñón/citología , Túbulos Renales/citología , Túbulos Renales/metabolismo , Masculino , Células Mesangiales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Microscopía Fluorescente , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Podocitos/metabolismo , Receptores Citoplasmáticos y Nucleares/clasificación , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Represoras , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
J Autoimmun ; 39(4): 428-40, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22921335

RESUMEN

Interleukin-17A (IL-17A) is the signature cytokine produced by Th17 CD4(+) T cells and has been tightly linked to autoimmune pathogenesis. In particular, the transcription factors NFAT and RORγt are known to activate Il17a transcription, although the detailed mechanism of action remains incompletely understood. Here, we show that the nuclear orphan receptor NR2F6 can attenuate the capacity of NFAT to bind to critical regions of the Il17a gene promoter. In addition, because NR2F6 binds to defined hormone response elements (HREs) within the Il17a locus, it interferes with the ability of RORγt to access the DNA. Consistently, NFAT and RORγt binding within the Il17a locus were enhanced in Nr2f6-deficient CD4(+) Th17 cells but decreased in Nr2f6-overexpressing transgenic CD4(+) Th17 cells. Taken together, our findings uncover an example of antagonistic regulation of Il17a transcription through the direct reciprocal actions of NR2F6 versus NFAT and RORγt.


Asunto(s)
Factores de Transcripción COUP/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Interleucina-17/inmunología , Factores de Transcripción NFATC/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Células Th17/inmunología , Animales , Sitios de Unión , Unión Competitiva , Factores de Transcripción COUP/deficiencia , Factores de Transcripción COUP/genética , ADN/inmunología , ADN/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Regulación de la Expresión Génica/inmunología , Interleucina-17/genética , Interleucina-17/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Unión Proteica , Proteínas Represoras , Elementos de Respuesta/inmunología , Transducción de Señal , Células Th17/metabolismo , Células Th17/patología , Transcripción Genética
11.
Dev Dyn ; 241(10): 1603-15, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22836912

RESUMEN

BACKGROUND: The vertebrate nuclear receptor subfamily 2, group f (nr2f) genes encode orphan receptors that have the capacity to act as negative regulators of retinoic acid (RA) signaling. RESULTS: We describe embryonic and larval expression of four of the six zebrafish nr2f genes, nr2f1a, nr2f1b, nr2f2, and nr2f5. These genes show highly regulated patterns of expression within the central nervous system, including in the developing hindbrain, as well as in the mesoderm and endoderm. We also investigated the role of RA and fibroblast growth factor (Fgf) signaling in regulating early nr2f gene expression. RA is not required for nr2f expression in the hindbrain; however, exogenous RA can repress this expression. Conversely, we find that RA positively regulates nr2f1a expression in trunk endoderm and mesoderm. Fgf signaling is not required for nr2f expression onset in the hindbrain; however, it may play a role in maintaining rhombomere-specific expression. CONCLUSIONS: We report detailed expression analysis of four nr2f genes in all three germ layers. The onset of nr2f expression in the hindbrain does not require RA or Fgf signals. Our finding that RA positively regulates nr2f1a expression in the trunk supports the possibility that Nr2fs function in a negative feedback loop to modulate RA signaling in this region.


Asunto(s)
Factores de Transcripción COUP/metabolismo , Sistema Nervioso Central/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Transducción de Señal/fisiología , Tretinoina/metabolismo , Pez Cebra/embriología , Animales , Factores de Transcripción COUP/genética , Cartilla de ADN/genética , Perfilación de la Expresión Génica , Hibridación in Situ
12.
Thromb Haemost ; 108(2): 277-83, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22628013

RESUMEN

Hereditary factor VII (FVII) deficiency is a rare autosomal recessive disorder. Deleterious mutations that prevent the synthesis of any amount of functional FVII have been associated with life-threatening haemorrhage in neonates. Here we report two infants, of Maghrebian origin, who suffered a fatal spontaneous cerebral haemorrhage. Investigation of the molecular basis for their severe FVII deficiency revealed novel mutations in a homozygous state within the F7 gene promoter: a single nucleotide substitution (c.-65G>C) and a 2bp deletion (c.-60_-59delTT). To determine whether these promoter variants were responsible for the FVII deficiency, computer-assisted sequence analyses were performed. The data predicted a disrupted binding of both HNF4 and COUP-TF transcription factors with each variant. Concordantly, experimental results revealed an altered HNF4-induced transactivation in the promoter mutated variants. The execution of functional tests is critical to ensuring a complete understanding of the effect of any promoter mutant on FVII deficiency. Only then can an accurate molecular diagnosis be made and further genetic counselling and prenatal diagnosis be offered.


Asunto(s)
Hemorragia Cerebral/genética , Deficiencia del Factor VII/genética , Factor VII/genética , Mutación , Argelia , Coagulación Sanguínea , Factores de Transcripción COUP/genética , Femenino , Genes Reporteros , Asesoramiento Genético , Vectores Genéticos , Genotipo , Células Hep G2 , Factor Nuclear 4 del Hepatocito/genética , Humanos , Lactante , Recién Nacido , Regiones Promotoras Genéticas , Unión Proteica , Transcripción Genética , Transfección
13.
Am J Physiol Renal Physiol ; 302(8): F1025-33, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22278040

RESUMEN

Understanding the transcriptional mechanisms of renin expression is key to understanding the regulation of the renin-angiotensin system. We previously identified the nuclear receptors RAR/RXR and Nr2f6 (EAR2) as positive and negative transcriptional regulators of renin expression, respectively (Liu X, Huang X, Sigmund CD. Circ Res 92: 1033-1040, 2003). Both mediate their effects through a hormone response element (HRE) within the renin enhancer. Here, we determined whether another nuclear receptor, Nr2f2 (Coup-TFII, Arp-1), identified in a screen of proteins that bind the HRE, also regulates renin expression. Luciferase assays indicate that Nr2f2 negatively regulates the renin promoter more potently than Nr2f6. Gel-shift and chromatin immunoprecipitation (ChIP) indicate that Nr2f2 and Nr2f6 can bind directly to the renin enhancer through the HRE. Surprisingly, baseline expression of endogenous renin was not effected when Nr2f2 was knocked down in As4.1 cells, whereas knockdown of Nr2f6 increased renin expression twofold. Interestingly, however, knockdown of Nr2f2 augmented the induction of renin expression caused by retinoic acid. These data indicate that both Nr2f6 and Nr2f2 can negatively regulate the renin promoter, under baseline conditions and in response to physiological queues, respectively. Therefore, Nr2f2 may require an initiating signal that results in a change at the chromatin level or activation of another transcription factor to exert its effects. We conclude that both Nr2f2 and Nr2f6 negatively regulate renin promoter activity, but may do so by divergent mechanisms.


Asunto(s)
Factor de Transcripción COUP II/metabolismo , Factores de Transcripción COUP/metabolismo , Regulación de la Expresión Génica , Renina/genética , Animales , Secuencia de Bases , Factor de Transcripción COUP II/genética , Factores de Transcripción COUP/genética , Línea Celular , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Ratones , Regiones Promotoras Genéticas , Proteínas Represoras , Tretinoina/farmacología
14.
PLoS One ; 6(4): e19443, 2011 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-21552541

RESUMEN

BACKGROUND: COUP transcription factors are required for the regulation of gene expression underlying development, differentiation, and homeostasis. They have an evolutionarily conserved function, being a known marker for neurogenesis from cnidarians to vertebrates. A homologue of this gene was shown previously to be a neuronal and nematocyte differentiation marker in Hydra. However, COUP-TFs had not previously been studied in a colonial cnidarian. METHODOLOGY/PRINCIPAL FINDINGS: We cloned a COUP-TF homologue from the colonial marine cnidarian Hydractinia echinata. Expression of the gene was analysed during normal development, allorecognition events and ectopic Wnt activation, using in situ hybridisation and quantitative PCR. During normal Hydractinia development, the gene was first expressed in post-gastrula stages. It was undetectable in larvae, and its mRNA was present again in putative differentiating neurons and nematocytes in post-metamorphic stages. Global activation of canonical Wnt signalling in adult animals resulted in the upregulation of COUP-TF. We also monitored a strong COUP-TF upregulation in stolons undergoing allogeneic interactions. COUP-TF mRNA was most concentrated in the tissues that contacted allogeneic, non-self tissues, and decreased in a gradient away from the contact area. Interestingly, the gene was transiently upregulated during initial contact of self stolons, but dissipated rapidly following self recognition, while in non-self contacts high expression levels were maintained. CONCLUSIONS/SIGNIFICANCE: We conclude that COUP-TF is likely involved in neuronal/nematocyte differentiation in a variety of contexts. This has now been shown to include allorecognition, where COUP-TF is thought to have been co-opted to mediate allorejection by recruiting stinging cells that are the effectors of cytotoxic rejection of allogeneic tissue. Our findings that Wnt activation upregulates COUP-TF expression suggests that Wnts' role in neuronal differentiation could be mediated through COUP-TF.


Asunto(s)
Factores de Transcripción COUP/genética , Regulación del Desarrollo de la Expresión Génica , Hidrozoos/citología , Hidrozoos/genética , Transducción de Señal , Proteínas Wnt/metabolismo , Secuencia de Aminoácidos , Animales , Factores de Transcripción COUP/química , Evolución Molecular , Humanos , Hidrozoos/crecimiento & desarrollo , Hidrozoos/inmunología , Datos de Secuencia Molecular , Neurogénesis/genética , Regulación hacia Arriba
15.
BMC Mol Biol ; 12: 4, 2011 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-21247419

RESUMEN

BACKGROUND: The Rasd1 protein is a dexamethasone induced monomeric Ras-like G protein that oscillates in the suprachiasmatic nucleus (SCN). Previous studies have shown that Rasd1 modulates multiple signaling cascades. However, it is still unclear exactly how Rasd1 carries out its function. Studying protein-protein interactions involving Rasd1 may provide insights into its biological functions in different contexts. RESULTS: To further explore the molecular function of Rasd1, we performed a yeast two-hybrid screen and identified Ear2, a negative regulator of renin transcription, as an interaction partner of Rasd1. We validated the interaction in vitro and in transfected COS-7 cells. We further confirmed the interaction of endogenous Rasd1 and Ear2 from HEK293T cell and mouse brain extract. Rasd1 inhibited transcriptional repression by Ear2 on a renin promoter-luciferase reporter construct both in the presence and absence of all-trans-retinoic acid. Moreover, real-time RT-PCR showed upregulation of endogenous renin transcription in As4.1 cells over-expressing Rasd1. We demonstrated that the ligand binding domain of Ear2 is required for physical and functional interaction between the two proteins. In addition, we demonstrated that shRNA-mediated knockdown of Rasd1 results in further repression of Ear2-mediated renin transcription, whereas induction of Rasd1 by dexamethasone counteracts the effects of shRNA-mediated Rasd1 knockdown. Finally, our study showed that Rasd1 missense mutations not only attenuate their physical interaction with Ear2 but also abolish their ability to counteract repression of renin transcription mediated by Ear2. CONCLUSIONS: Our study provides evidence for physical and functional interactions between Rasd1 and Ear2. The results suggest that their interactions are involved in renin transcriptional regulation. These findings not only reveal a novel role for Rasd1-medated signaling but also provide the basis for potential intervention of renin expression.


Asunto(s)
Factores de Transcripción COUP/genética , Receptores de Esteroides/genética , Renina/genética , Proteínas ras/genética , Animales , Células COS , Chlorocebus aethiops , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genes Reporteros , Células HEK293 , Humanos , Ratones , ARN Interferente Pequeño/metabolismo , Renina/metabolismo , Proteínas Represoras , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Transcripción Genética , Tretinoina/metabolismo , Técnicas del Sistema de Dos Híbridos
16.
Cereb Cortex ; 21(6): 1311-30, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21060110

RESUMEN

A hallmark of mammalian evolution is the structural and functional complexity of the cerebral cortex. Within the cerebral cortex, the neocortex, or isocortex, is a 6-layered complexly organized structure that is comprised of multiple interconnected sensory and motor areas. These areas and their precise patterns of connections arise during development, through a process termed arealization. Intrinsic, activity-independent and extrinsic, activity-dependent mechanisms are involved in the development of neocortical areas and their connections. The intrinsic molecular mechanisms involved in the establishment of this sophisticated network are not fully understood. In this report (I) and the companion report (II), we present the first lifespan analysis of ipsilateral intraneocortical connections (INCs) among multiple sensory and motor regions, from the embryonic period to adulthood in the mouse. Additionally, we characterize the neocortical expression patterns of several developmentally regulated genes that are of central importance to studies investigating the molecular control of arealization from embryonic day 13.5 to postnatal day (P) 3 (I) and P6 to 50 (II). In this analysis, we utilize novel methods to correlate the boundaries of gene expression with INCs and developing areal boundaries, in order to better understand the nature of gene-areal relationships during development.


Asunto(s)
Corteza Cerebral/embriología , Corteza Cerebral/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica/fisiología , Expresión Génica/fisiología , Factores de Edad , Aminoácidos/metabolismo , Animales , Animales Recién Nacidos , Mapeo Encefálico , Factores de Transcripción COUP/genética , Factores de Transcripción COUP/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Corteza Cerebral/citología , Embrión de Mamíferos , Efrina-A5/genética , Efrina-A5/metabolismo , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteína 2 Inhibidora de la Diferenciación/genética , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Proteínas con Homeodominio LIM , Ratones , Vías Nerviosas/embriología , Vías Nerviosas/crecimiento & desarrollo , Vías Nerviosas/metabolismo , Miembro 2 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 2 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Embarazo , Compuestos de Piridinio/metabolismo , Receptor EphA7/genética , Receptor EphA7/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
Cereb Cortex ; 21(6): 1331-50, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21060113

RESUMEN

The mammalian neocortex contains an intricate processing network of multiple sensory and motor areas that allows the animal to engage in complex behaviors. These anatomically and functionally unique areas and their distinct connections arise during early development, through a process termed arealization. Both intrinsic, activity-independent and extrinsic, activity-dependent mechanisms drive arealization, much of which occurs during the areal patterning period (APP) from late embryogenesis to early postnatal life. How areal boundaries and their connections develop and change from infancy to adulthood is not known. Additionally, the adult patterns of sensory and motor ipsilateral intraneocortical connections (INCs) have not been thoroughly characterized in the mouse. In this report and its companion (I), we present the first lifespan analysis of ipsilateral INCs among multiple sensory and motor regions in mouse. We describe the neocortical expression patterns of several developmentally regulated genes that are of central importance to studies investigating the molecular regulation of arealization, from postnatal day (P) 6 to P50. In this study, we correlate the boundaries of gene expression patterns with developing areal boundaries across a lifespan, in order to better understand the nature of gene-areal relationships from early postnatal life to adulthood.


Asunto(s)
Mapeo Encefálico , Corteza Cerebral/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica/fisiología , Expresión Génica/fisiología , Vías Nerviosas/crecimiento & desarrollo , Factores de Edad , Aminoácidos/metabolismo , Animales , Animales Recién Nacidos , Factores de Transcripción COUP/genética , Factores de Transcripción COUP/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Corteza Cerebral/citología , Embrión de Mamíferos , Efrina-A5/genética , Efrina-A5/metabolismo , Femenino , Lateralidad Funcional , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteína 2 Inhibidora de la Diferenciación/genética , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Proteínas con Homeodominio LIM , Ratones , Vías Nerviosas/metabolismo , Miembro 2 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 2 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Embarazo , Compuestos de Piridinio/metabolismo , Receptor EphA7/genética , Receptor EphA7/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
18.
Fertil Steril ; 94(7): 2521-7, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20430378

RESUMEN

OBJECTIVE: To study the effect of peritoneal fluid from women with (PF-E) and without (PF-C) endometriosis on P(450)Arom expression in endometrial cells. DESIGN: Experimental study. SETTING: University research unit. PATIENT(S): Forty women of reproductive age with (n = 22) or without (control; n = 18) endometriosis. INTERVENTION(S): Peritoneal fluid and eutopic endometrial samples were obtained during surgery from women with (n = 13 and 9, respectively) and without (n = 4 and 14, respectively) endometriosis. MAIN OUTCOME MEASURE(S): Expression study for P(450)Arom, steroid factor 1 (SF-1), chicken ovalbumin upstream transcription factor I (COUP-TFI), and COUP-TFII messenger RNA (reverse transcriptase-polymerase chain reacion) and/or protein (immunoblot) in isolated endometrial epithelial cells transfected or not with expression vector containing SF-1, COUP-TFI, or COUP-TFII complementary DNAs. RESULT(S): Basal messenger RNA and/or protein expression of P(450)Arom and SF-1 were augmented in endometriosis, and that of COUP-TF was diminished. In control cells, (Bu)(2)cAMP and PF-E increased P(450)Arom and SF-1 expression (but not COUP-TF expression) in a dose-dependent way, an effect not observed with PF-C, adsorbed PF-E, or 10(-5) M indomethacin. Transfected cells confirmed these results. Any treatments modified the studied molecules in endometriosis cells. CONCLUSION(S): These data indicate that molecules contained in PF-E favor an estrogenic microenvironment, suggesting a role in the etiopathogenesis of endometriosis enabling the survival, maintenance, and growth of endometrial implants in the ectopic locations.


Asunto(s)
Aromatasa/biosíntesis , Líquido Ascítico/patología , Líquido Ascítico/fisiología , Endometriosis/patología , Endometrio/metabolismo , Enfermedades Peritoneales/patología , Adulto , Aromatasa/genética , Factores de Transcripción COUP/genética , Factores de Transcripción COUP/metabolismo , Estudios de Casos y Controles , Separación Celular , Células Cultivadas , Endometriosis/metabolismo , Endometrio/citología , Endometrio/efectos de los fármacos , Endometrio/enzimología , Inducción Enzimática , Femenino , Humanos , Persona de Mediana Edad , Enfermedades Peritoneales/metabolismo , Factor Esteroidogénico 1/genética , Factor Esteroidogénico 1/metabolismo
19.
Development ; 137(5): 725-34, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20147377

RESUMEN

Transcriptional networks, which are initiated by secreted proteins, cooperate with each other to orchestrate eye development. The establishment of dorsal/ventral polarity, especially dorsal specification in the optic vesicle, is poorly understood at a molecular and cellular level. Here, we show that COUP-TFI (Nr2f1) and COUP-TFII (Nr2f2) are highly expressed in the progenitor cells in the developing murine eye. Phenotype analysis of COUP-TFI and COUP-TFII single-gene conditional knockout mouse models suggests that COUP-TFs compensate for each other to maintain morphogenesis of the eye. However, in eye-specific COUP-TFI/TFII double-knockout mice, progenitor cells at the dorso-distal optic vesicle fail to differentiate appropriately, causing the retinal pigmented epithelium cells to adopt a neural retina fate and abnormal differentiation of the dorsal optic stalk; the development of proximo-ventral identities, neural retina and ventral optic stalk is also compromised. These cellular defects in turn lead to congenital ocular colobomata and microphthalmia. Immunohistochemical and in situ hybridization assays reveal that the expression of several regulatory genes essential for early optic vesicle development, including Pax6, Otx2, Mitf, Pax2 and Vax1/2, is altered in the corresponding compartments of the mutant eye. Using ChIP assay, siRNA treatment and transient transfection in ARPE-19 cells in vitro, we demonstrate that Pax6 and Otx2 are directly regulated by COUP-TFs. Taken together, our findings reveal novel and distinct cell-intrinsic mechanisms mediated by COUP-TF genes to direct the specification and differentiation of progenitor cells, and that COUP-TFs are crucial for dorsalization of the eye.


Asunto(s)
Factores de Transcripción COUP/fisiología , Proteínas del Ojo/genética , Ojo/embriología , Regulación del Desarrollo de la Expresión Génica , Morfogénesis/genética , Animales , Factores de Transcripción COUP/genética , Células Cultivadas , Coloboma/embriología , Coloboma/genética , Embrión de Mamíferos/metabolismo , Ojo/metabolismo , Anomalías del Ojo/embriología , Anomalías del Ojo/genética , Proteínas del Ojo/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Transgénicos , Microftalmía/embriología , Microftalmía/genética , Modelos Biológicos , Factores de Transcripción Otx/genética , Factores de Transcripción Otx/metabolismo , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Retina/embriología , Retina/metabolismo , Vías Visuales/embriología
20.
Gene ; 439(1-2): 63-70, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19341784

RESUMEN

Although the ovalbumin (Ov) gene has served as a model to study tissue-specific, steroid hormone-induced gene expression in vertebrates for decades, the mechanisms responsible for regulating this gene remain elusive. Ov is repressed in non-oviduct tissue and in estrogen-deprived oviduct by a strong repressor site located from -130 to -100 and designated CAR for COUP-TF adjacent repressor. The goal of this study was to identify the CAR binding protein(s). A transcription factor database search revealed that a putative interferon-stimulated response element (ISRE), which binds interferon regulatory factors (IRFs), is located in this region. Gel mobility shift assays demonstrated that the protein(s) binding to the CAR site is recognized by an IRF antibody and that mutations in the ISRE abolish that binding. In hopes of identifying the IRF(s) responsible for the tissue-specific regulation of Ov, mRNA levels for IRFs-4, -8, and -10 were measured in seven tissues from chicks treated with or without estrogen. PCR experiments showed that both IRF-8 and -10 are expressed in all chick tissues tested whereas IRF-4 has a much more limited expression pattern. Transfection experiments with OvCAT (chloramphenicol acetyltransferase) reporter constructs demonstrated that both IRF-4 and IRF-10 are capable of repressing the Ov gene even in the presence of steroid hormones and that nucleotides in the ISRE are required for repression. These experiments indicate that the repressor activity associated with the CAR site is mediated by IRF family members and suggest that IRF members also repress Ov in non-oviduct tissues.


Asunto(s)
Factores Reguladores del Interferón/fisiología , Ovalbúmina/metabolismo , Proteínas Represoras/fisiología , Animales , Sitios de Unión , Factores de Transcripción COUP/genética , Células Cultivadas , Pollos , Cloranfenicol O-Acetiltransferasa/genética , Estrógenos/farmacología , Femenino , Regulación de la Expresión Génica , Genes Reporteros , Factores Reguladores del Interferón/genética , Mutación , Especificidad de Órganos , Ovalbúmina/genética , Oviductos/citología , Regiones Promotoras Genéticas , Proteínas Represoras/genética , Elementos de Respuesta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...