Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 149
Filtrar
1.
Cell ; 163(4): 960-74, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26544941

RESUMEN

Alterations in estrogen-mediated cellular signaling play an essential role in the pathogenesis of endometriosis. In addition to higher estrogen receptor (ER) ß levels, enhanced ERß activity was detected in endometriotic tissues, and the inhibition of enhanced ERß activity by an ERß-selective antagonist suppressed mouse ectopic lesion growth. Notably, gain of ERß function stimulated the progression of endometriosis. As a mechanism to evade endogenous immune surveillance for cell survival, ERß interacts with cellular apoptotic machinery in the cytoplasm to inhibit TNF-α-induced apoptosis. ERß also interacts with components of the cytoplasmic inflammasome to increase interleukin-1ß and thus enhance its cellular adhesion and proliferation properties. Furthermore, this gain of ERß function enhances epithelial-mesenchymal transition signaling, thereby increasing the invasion activity of endometriotic tissues for establishment of ectopic lesions. Collectively, we reveal how endometrial tissue generated by retrograde menstruation can escape immune surveillance and develop into sustained ectopic lesions via gain of ERß function.


Asunto(s)
Endometriosis/patología , Receptor beta de Estrógeno/metabolismo , Inflamasomas/metabolismo , Menstruación/metabolismo , Animales , Apoptosis , Adhesión Celular , Proliferación Celular , Endometriosis/metabolismo , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Vigilancia Inmunológica , Interleucina-1beta/metabolismo , Ratones , Factor de Necrosis Tumoral alfa/metabolismo
2.
Mol Cell ; 78(6): 1114-1132.e10, 2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32446320

RESUMEN

Bromodomain-containing protein 4 (BRD4) is a cancer therapeutic target in ongoing clinical trials disrupting primarily BRD4-regulated transcription programs. The role of BRD4 in cancer has been attributed mainly to the abundant long isoform (BRD4-L). Here we show, by isoform-specific knockdown and endogenous protein detection, along with transgene expression, the less abundant BRD4 short isoform (BRD4-S) is oncogenic while BRD4-L is tumor-suppressive in breast cancer cell proliferation and migration, as well as mammary tumor formation and metastasis. Through integrated RNA-seq, genome-wide ChIP-seq, and CUT&RUN association profiling, we identify the Engrailed-1 (EN1) homeobox transcription factor as a key BRD4-S coregulator, particularly in triple-negative breast cancer. BRD4-S and EN1 comodulate the extracellular matrix (ECM)-associated matrisome network, including type II cystatin gene cluster, mucin 5, and cathepsin loci, via enhancer regulation of cancer-associated genes and pathways. Our work highlights the importance of targeted therapies for the oncogenic, but not tumor-suppressive, activity of BRD4.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiología , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Animales , Neoplasias de la Mama/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Genes Homeobox , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Invasividad Neoplásica , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas/antagonistas & inhibidores , Proteínas/metabolismo , Transcripción Genética/genética , Neoplasias de la Mama Triple Negativas/genética
3.
Mol Cell ; 70(4): 679-694.e7, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29775582

RESUMEN

Enhancers are thought to activate transcription by physically contacting promoters via looping. However, direct assays demonstrating these contacts are required to mechanistically verify such cellular determinants of enhancer function. Here, we present versatile cell-free assays to further determine the role of enhancer-promoter contacts (EPCs). We demonstrate that EPC is linked to mutually stimulatory transcription at the enhancer and promoter in vitro. SRC-3 was identified as a critical looping determinant for the estradiol-(E2)-regulated GREB1 locus. Surprisingly, the GREB1 enhancer and promoter contact two internal gene body SRC-3 binding sites, GBS1 and GBS2, which stimulate their transcription. Utilizing time-course 3C assays, we uncovered SRC-3-dependent dynamic chromatin interactions involving the enhancer, promoter, GBS1, and GBS2. Collectively, these data suggest that the enhancer and promoter remain "poised" for transcription via their contacts with GBS1 and GBS2. Upon E2 induction, GBS1 and GBS2 disengage from the enhancer, allowing direct EPC for active transcription.


Asunto(s)
Neoplasias de la Mama/genética , Cromatina/metabolismo , Estrógenos/farmacología , Regulación Neoplásica de la Expresión Génica , Coactivador 3 de Receptor Nuclear/metabolismo , Transcripción Genética , Sitios de Unión , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Cromatina/genética , Elementos de Facilitación Genéticos , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Coactivador 3 de Receptor Nuclear/genética , Regiones Promotoras Genéticas , Unión Proteica , Células Tumorales Cultivadas
4.
Nature ; 556(7700): 249-254, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29615789

RESUMEN

Alterations in both cell metabolism and transcriptional programs are hallmarks of cancer that sustain rapid proliferation and metastasis 1 . However, the mechanisms that control the interaction between metabolic reprogramming and transcriptional regulation remain unclear. Here we show that the metabolic enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 4 (PFKFB4) regulates transcriptional reprogramming by activating the oncogenic steroid receptor coactivator-3 (SRC-3). We used a kinome-wide RNA interference-based screening method to identify potential kinases that modulate the intrinsic SRC-3 transcriptional response. PFKFB4, a regulatory enzyme that synthesizes a potent stimulator of glycolysis 2 , is found to be a robust stimulator of SRC-3 that coregulates oestrogen receptor. PFKFB4 phosphorylates SRC-3 at serine 857 and enhances its transcriptional activity, whereas either suppression of PFKFB4 or ectopic expression of a phosphorylation-deficient Ser857Ala mutant SRC-3 abolishes the SRC-3-mediated transcriptional output. Functionally, PFKFB4-driven SRC-3 activation drives glucose flux towards the pentose phosphate pathway and enables purine synthesis by transcriptionally upregulating the expression of the enzyme transketolase. In addition, the two enzymes adenosine monophosphate deaminase-1 (AMPD1) and xanthine dehydrogenase (XDH), which are involved in purine metabolism, were identified as SRC-3 targets that may or may not be directly involved in purine synthesis. Mechanistically, phosphorylation of SRC-3 at Ser857 increases its interaction with the transcription factor ATF4 by stabilizing the recruitment of SRC-3 and ATF4 to target gene promoters. Ablation of SRC-3 or PFKFB4 suppresses breast tumour growth in mice and prevents metastasis to the lung from an orthotopic setting, as does Ser857Ala-mutant SRC-3. PFKFB4 and phosphorylated SRC-3 levels are increased and correlate in oestrogen receptor-positive tumours, whereas, in patients with the basal subtype, PFKFB4 and SRC-3 drive a common protein signature that correlates with the poor survival of patients with breast cancer. These findings suggest that the Warburg pathway enzyme PFKFB4 acts as a molecular fulcrum that couples sugar metabolism to transcriptional activation by stimulating SRC-3 to promote aggressive metastatic tumours.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , Glucosa/metabolismo , Coactivador 3 de Receptor Nuclear/metabolismo , Fosfofructoquinasa-2/metabolismo , Activación Transcripcional , Factor de Transcripción Activador 4/metabolismo , Animales , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Glucólisis , Humanos , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Ratones , Metástasis de la Neoplasia , Vía de Pentosa Fosfato , Fosforilación , Fosfoserina/metabolismo , Pronóstico , Purinas/biosíntesis , Purinas/metabolismo , Interferencia de ARN , Receptores de Estrógenos/metabolismo , Transcetolasa/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
PLoS Genet ; 16(6): e1008868, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32579581

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disorder featuring progressive loss of midbrain dopaminergic (DA) neurons that leads to motor symptoms. The etiology and pathogenesis of PD are not clear. We found that expression of COUP-TFII, an orphan nuclear receptor, in DA neurons is upregulated in PD patients through the analysis of public datasets. We show here that through epigenetic regulation, COUP-TFII contributes to oxidative stress, suggesting that COUP-TFII may play a role in PD pathogenesis. Elevated COUP-TFII expression specifically in DA neurons evokes DA neuronal loss in mice and accelerates the progression of phenotypes in a PD mouse model, MitoPark. Compared to control mice, those with elevated COUP-TFII expression displayed reduced cristae in mitochondria and enhanced cellular electron-dense vacuoles in the substantia nigra pars compacta. Mechanistically, we found that overexpression of COUP-TFII disturbs mitochondrial pathways, resulting in mitochondrial dysfunction. In particular, there is repressed expression of genes encoding cytosolic aldehyde dehydrogenases, which could enhance oxidative stress and interfere with mitochondrial function via 3,4-dihydroxyphenylacetaldehyde (DOPAL) buildup in DA neurons. Importantly, under-expression of COUP-TFII in DA neurons slowed the deterioration in motor functions of MitoPark mice. Taken together, our results suggest that COUP-TFII may be an important contributor to PD development and a potential therapeutic target.


Asunto(s)
Factor de Transcripción COUP II/metabolismo , Neuronas Dopaminérgicas/patología , Mitocondrias/patología , Enfermedad de Parkinson/genética , Ácido 3,4-Dihidroxifenilacético/análogos & derivados , Ácido 3,4-Dihidroxifenilacético/metabolismo , Aldehído Deshidrogenasa , Animales , Encéfalo/citología , Encéfalo/patología , Línea Celular , Línea Celular Tumoral , Estudios de Cohortes , Conjuntos de Datos como Asunto , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Neuronas Dopaminérgicas/citología , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Estrés Oxidativo/genética , Enfermedad de Parkinson/patología , Cultivo Primario de Células , RNA-Seq , Ratas , Regulación hacia Arriba
6.
Ecotoxicol Environ Saf ; 263: 115358, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37595350

RESUMEN

Seasonal effects on subclinical cardiovascular functions (CVFs) are an important emerging health issue for people living in urban environment. The objectives of this study were to demonstrate the effects of seasonal variations of temperature, relative humidity, and PM2.5 air pollution on CVFs. A total of 86 office workers in Taipei City were recruited, their arterial pressure waveform was recorded by cuff sphygmomanometer using an oscillometric blood pressure (BP) device for CVFs assessment. Results of paried t-test with Bonferroni correction showed significantly increased systolic and diastolic BP (SBP, DBP), central end-systolic and diastolic BP (cSBP, cDBP) and systemic vascular resistance, but decreased heart rate (HR), stroke volume (SV), cardio output (CO), and cardiac index in winter compared with other seasons. After controlling for related confounding factors, SBP, DBP, cSBP, cDBP, LV dp/dt max, and brachial-ankle pulse wave velocity (baPWV) were negatively associated with, and SV was positively associated with seasonal temperature changes. Seasonal changes of air pollution in terms of PM2.5 were significantly positively associated with DBP and cDBP, as well as negatively associated with HR and CO. Seasonal changes of relative humidity were significantly negatively associated with DBP, and cDBP, as well as positively associated with HR, CO, and baPWV. This study provides evidence of greater susceptibility to cardiovascular events in winter compared with other seasons, with ambient temperature, relative humidity, and PM2.5 as the major factors of seasonal variation of CVFs.


Asunto(s)
Contaminación del Aire , Índice Tobillo Braquial , Humanos , Estaciones del Año , Temperatura , Humedad , Análisis de la Onda del Pulso , Contaminación del Aire/efectos adversos , Material Particulado
7.
Mol Cell ; 51(2): 185-99, 2013 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-23850489

RESUMEN

Chromatin immunoprecipitation studies have mapped protein occupancies at many genomic loci. However, a detailed picture of the complexity of coregulators (CoRs) bound to a defined enhancer along with a transcription factor is missing. To address this, we used biotin-DNA pull-down assays coupled with mass spectrometry-immunoblotting to identify at least 17 CoRs from nuclear extracts bound to 17ß-estradiol (E2)-liganded estrogen receptor-α on estrogen response elements (EREs). Unexpectedly, these complexes initially are biochemically stable and contain certain atypical corepressors. Addition of ATP dynamically converts these complexes to an "activated" state by phosphorylation events, primarily mediated by DNA-dependent protein kinase. Importantly, a "natural" ERE-containing enhancer and nucleosomal EREs recruit similar complexes. We further discovered the mechanism whereby H3K4me3 stimulates ERα-mediated transcription as compared with unmodified nucleosomes. H3K4me3 templates promote specific CoR dynamics in the presence of ATP and AcCoA, as manifested by CBP/p300 and SRC-3 dismissal and SAGA and TFIID stabilization/recruitment.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Receptor alfa de Estrógeno/metabolismo , Regulación Neoplásica de la Expresión Génica , Nucleosomas/metabolismo , Proteómica , Elementos de Respuesta/genética , Neoplasias de la Mama/genética , Inmunoprecipitación de Cromatina , ADN/genética , Proteína Quinasa Activada por ADN/genética , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteína p300 Asociada a E1A/genética , Proteína p300 Asociada a E1A/metabolismo , Receptor alfa de Estrógeno/genética , Estrógenos/farmacología , Femenino , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Células HeLa , Humanos , Células MCF-7 , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Coactivador 3 de Receptor Nuclear/genética , Coactivador 3 de Receptor Nuclear/metabolismo , Nucleosomas/genética , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Sialoglicoproteínas/genética , Sialoglicoproteínas/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Transactivadores , Transcripción Genética , Activación Transcripcional
8.
FASEB J ; 33(6): 7375-7386, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30866655

RESUMEN

Polycystic ovary syndrome (PCOS) is a hypothalamic-pituitary-gonadal (HPG) axis disorder. PCOS symptoms most likely result from a disturbance in the complex feedback regulation system of the HPG axis, which involves gonadotrophic hormones and ovarian steroid hormones. However, the nature of this complex and interconnecting feedback regulation makes it difficult to dissect the molecular mechanisms responsible for PCOS phenotypes. Global estrogen receptor α (ERα) knockout (KO) mice exhibit a disruption of the HPG axis, resulting in hormonal dysregulation in which female ERα KO mice have elevated levels of serum estradiol (E2), testosterone, and LH. The ERα KO females are anovulatory and develop cystic hemorrhagic ovaries that are thought to be due to persistently high circulating levels of LH from the pituitary. However, the role of ERα in the pituitary is still controversial because of the varied phenotypes reported in pituitary-specific ERα KO mouse models. Therefore, we developed a mouse model where ERα is reintroduced to be exclusively expressed in the pituitary on the background of a global ERα-null (PitERtgKO) mouse. Serum E2 and LH levels were normalized in PitERtgKO females and were comparable to wild-type serum levels. However, the ovaries of PitERtgKO adult mice displayed a more overt cystic and hemorrhagic phenotype when compared with ERα KO littermates. We determined that anomalous sporadic LH secretion caused the severe ovarian phenotype of PitERtgKO females. Our observations suggest that pituitary ERα is involved in the estrogen negative feedback regulation, whereas hypothalamic ERα is necessary for the precise control of LH secretion. Uncontrolled, irregular LH secretion may be the root cause of the cystic ovarian phenotype with similarities to PCOS.-Arao, Y., Hamilton, K. J., Wu, S.-P., Tsai, M.-J., DeMayo, F. J., Korach, K. S. Dysregulation of hypothalamic-pituitary estrogen receptor α-mediated signaling causes episodic LH secretion and cystic ovary.


Asunto(s)
Receptor alfa de Estrógeno/fisiología , Hipotálamo/fisiopatología , Hormona Luteinizante/metabolismo , Ovario/fisiopatología , Adenohipófisis/fisiopatología , Síndrome del Ovario Poliquístico/fisiopatología , Animales , Modelos Animales de Enfermedad , Estradiol/fisiología , Receptor alfa de Estrógeno/deficiencia , Receptor alfa de Estrógeno/genética , Retroalimentación Fisiológica , Femenino , Hemorragia/etiología , Humanos , Sistema Hipotálamo-Hipofisario/fisiopatología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Especificidad de Órganos , Ovario/patología , Síndrome del Ovario Poliquístico/sangre , Síndrome del Ovario Poliquístico/patología , Proteínas Recombinantes/metabolismo
9.
Nature ; 493(7431): 236-40, 2013 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-23201680

RESUMEN

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


Asunto(s)
Factor de Transcripción COUP II/metabolismo , Transformación Celular Neoplásica , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Transducción de Señal , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Factor de Transcripción COUP II/deficiencia , Factor de Transcripción COUP II/genética , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Eliminación de Gen , Humanos , Masculino , Ratones , Metástasis de la Neoplasia , Fosfohidrolasa PTEN/deficiencia , Fosfohidrolasa PTEN/genética , Modelos de Riesgos Proporcionales , Próstata/metabolismo , Próstata/patología , Proteína Smad4/deficiencia , Proteína Smad4/genética , Proteína Smad4/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
10.
J Biol Chem ; 292(35): 14456-14472, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28717009

RESUMEN

The transition from transcription initiation to elongation is a key regulatory step in gene expression, which requires RNA polymerase II (pol II) to escape promoter proximal pausing on chromatin. Although elongation factors promote pause release leading to transcription elongation, the role of epigenetic modifications during this critical transition step is poorly understood. Two histone marks on histone H3, lysine 4 trimethylation (H3K4me3) and lysine 9 acetylation (H3K9ac), co-localize on active gene promoters and are associated with active transcription. H3K4me3 can promote transcription initiation, yet the functional role of H3K9ac is much less understood. We hypothesized that H3K9ac may function downstream of transcription initiation by recruiting proteins important for the next step of transcription. Here, we describe a functional role for H3K9ac in promoting pol II pause release by directly recruiting the super elongation complex (SEC) to chromatin. H3K9ac serves as a substrate for direct binding of the SEC, as does acetylation of histone H4 lysine 5 to a lesser extent. Furthermore, lysine 9 on histone H3 is necessary for maximal pol II pause release through SEC action, and loss of H3K9ac increases the pol II pausing index on a subset of genes in HeLa cells. At select gene promoters, H3K9ac loss or SEC depletion reduces gene expression and increases paused pol II occupancy. We therefore propose that an ordered histone code can promote progression through the transcription cycle, providing new mechanistic insight indicating that SEC recruitment to certain acetylated histones on a subset of genes stimulates the subsequent release of paused pol II needed for transcription elongation.


Asunto(s)
Ensamble y Desensamble de Cromatina , Histonas/metabolismo , Lisina/metabolismo , Modelos Biológicos , Procesamiento Proteico-Postraduccional , Elongación de la Transcripción Genética , Iniciación de la Transcripción Genética , Acetilación , Sustitución de Aminoácidos , Animales , Drosophila , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Epigénesis Genética , Células HeLa , Histonas/antagonistas & inhibidores , Histonas/química , Histonas/genética , Humanos , Mutación , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Dominios y Motivos de Interacción de Proteínas , Interferencia de ARN , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
11.
Mol Cell ; 37(3): 321-32, 2010 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-20159552

RESUMEN

EGF induces signal transduction between EGFR and FAK, and FAK is required for EGF-induced cell migration. It is unknown, however, what factor mediates the interaction between EGFR and FAK and leads to EGF-induced FAK phosphorylation. Here, we identify SRC-3Delta4, a splicing isoform of the SRC-3 oncogene, as a signaling adaptor that links EGFR and FAK and promotes EGF-induced phosphorylations of FAK and c-Src. We identify three PAK1-mediated phosphorylations in SRC-3Delta4 that promote the localization of SRC-3Delta4 to the plasma membrane and mediate the interactions with EGFR and FAK. Importantly, overexpression of SRC-3Delta4 promotes MDA-MB231-induced breast tumor metastasis. Our findings identify phosphorylated SRC-3Delta4 as a missing adaptor between EGFR and its downstream signaling molecule FAK to coordinately regulate EGF-induced cell migration. Our study also reveals that a nuclear receptor coactivator can act in the periphery of a cell to directly mediate activation of an enzyme.


Asunto(s)
Movimiento Celular/fisiología , Receptores ErbB/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Coactivador 3 de Receptor Nuclear/fisiología , Animales , Línea Celular Tumoral , Femenino , Quinasa 1 de Adhesión Focal/análisis , Humanos , Neoplasias Pulmonares/secundario , Ganglios Linfáticos/patología , Metástasis Linfática , Ratones , Metástasis de la Neoplasia , Coactivador 3 de Receptor Nuclear/análisis , Coactivador 3 de Receptor Nuclear/genética , Fosforilación , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Transducción de Señal , Quinasas p21 Activadas/metabolismo , Familia-src Quinasas/metabolismo , Familia-src Quinasas/fisiología
12.
PLoS Genet ; 11(4): e1005116, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25830341

RESUMEN

Ever since we developed mitochondria to generate ATP, eukaryotes required intimate mito-nuclear communication. In addition, since reactive oxygen species are a cost of mitochondrial oxidative phosphorylation, this demands safeguards as protection from these harmful byproducts. Here we identified a critical transcriptional integrator which eukaryotes share to orchestrate both nutrient-induced mitochondrial energy metabolism and stress-induced nuclear responses, thereby maintaining carbon-nitrogen balance, and preserving life span and reproductive capacity. Inhibition of nutrient-induced expression of CAPER arrests nutrient-dependent cell proliferation and ATP generation and induces autophagy-mediated vacuolization. Nutrient signaling to CAPER induces mitochondrial transcription and glucose-dependent mitochondrial respiration via coactivation of nuclear receptor ERR-α-mediated Gabpa transcription. CAPER is also a coactivator for NF-κB that directly regulates c-Myc to coordinate nuclear transcriptome responses to mitochondrial stress. Finally, CAPER is responsible for anaplerotic carbon flux into TCA cycles from glycolysis, amino acids and fatty acids in order to maintain cellular energy metabolism to counter mitochondrial stress. Collectively, our studies reveal CAPER as an evolutionarily conserved 'master' regulatory mechanism by which eukaryotic cells control vital homeostasis for both ATP and antioxidants via CAPER-dependent coordinated control of nuclear and mitochondrial transcriptomic programs and their metabolisms. These CAPER dependent bioenergetic programs are highly conserved, as we demonstrated that they are essential to preserving life span and reproductive capacity in human cells-and even in C. elegans.


Asunto(s)
Metabolismo Energético , Factor de Transcripción de la Proteína de Unión a GA/metabolismo , Glucosa/metabolismo , Mitocondrias/metabolismo , Estrés Oxidativo , Proteínas de Unión al ARN/metabolismo , Receptores de Estrógenos/metabolismo , Transactivadores/metabolismo , Adaptación Fisiológica , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Línea Celular , Factor de Transcripción de la Proteína de Unión a GA/genética , Homeostasis , Humanos , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo , Oxidación-Reducción , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas de Unión al ARN/genética , Receptores de Estrógenos/genética , Transactivadores/genética , Receptor Relacionado con Estrógeno ERRalfa
13.
Am J Hum Genet ; 94(2): 303-9, 2014 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-24462372

RESUMEN

Optic nerve atrophy and hypoplasia can be primary disorders or can result from trans-synaptic degeneration arising from cerebral visual impairment (CVI). Here we report six individuals with CVI and/or optic nerve abnormalities, born after an uneventful pregnancy and delivery, who have either de novo heterozygous missense mutations in NR2F1, also known as COUP-TFI, or deletions encompassing NR2F1. All affected individuals show mild to moderate intellectual impairment. NR2F1 encodes a nuclear receptor protein that regulates transcription. A reporter assay showed that missense mutations in the zinc-finger DNA-binding domain and the putative ligand-binding domain decrease NR2F1 transcriptional activity. These findings indicate that NR2F1 plays an important role in the neurodevelopment of the visual system and that its disruption can lead to optic atrophy with intellectual disability.


Asunto(s)
Factor de Transcripción COUP I/genética , Discapacidad Intelectual/genética , Atrofia Óptica/genética , Adolescente , Adulto , Secuencia de Aminoácidos , Factor de Transcripción COUP I/metabolismo , Niño , Preescolar , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Genotipo , Humanos , Discapacidad Intelectual/patología , Masculino , Datos de Secuencia Molecular , Mutación Missense , Atrofia Óptica/patología , Fenotipo , Adulto Joven , Dedos de Zinc/genética
14.
Biol Reprod ; 96(2): 313-326, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28203817

RESUMEN

The precise timing of progesterone signaling through its cognate receptor, the progesterone receptor (PGR), is critical for the establishment and maintenance of pregnancy. Loss of PGR expression in the murine uterine epithelium during the preimplantation period is a marker for uterine receptivity and embryo attachment. We hypothesized that the decrease in progesterone receptor A (PGRA) expression is necessary for successful embryo implantation. To test this hypothesis, a mouse model constitutively expressing PGRA (mPgrALsL/+) was generated. Expression of PGRA in all uterine compartments (Pgrcre) or uterine epithelium (Wnt7acre) resulted in infertility with defects in embryo attachment and stromal decidualization. Expression of critical PGRA target genes, indian hedgehog, and amphiregulin (Areg), was maintained through the window of receptivity while the estrogen receptor target gene, the leukemia inhibitory factor (Lif), a key regulator of embryo receptivity, was decreased. Transcriptomic and cistromic analyses of the mouse uterus at day 4.5 of pregnancy identified an altered group of genes regulating molecular transport in the control of fluid and ion levels within the uterine interstitial space. Additionally, LIF and its cognate receptor, the leukemia inhibitory factor receptor (LIFR), exhibited PGR-binding events in regions upstream of the transcriptional start sites, suggesting PGRA is inhibiting transcription at these loci. Therefore, downregulation of the PGRA isoform at the window of receptivity is necessary for the attenuation of hedgehog signaling, transcriptional activation of LIF signaling, and modulation of solutes and fluid, producing a receptive environment for the attaching embryo.


Asunto(s)
Implantación del Embrión , Endometrio , Progesterona/metabolismo , Receptores de Progesterona/metabolismo , Alelos , Animales , Clonación Molecular , Regulación hacia Abajo , Femenino , Regulación de la Expresión Génica/fisiología , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Masculino , Ratones Transgénicos , Receptores OSM-LIF/genética , Receptores OSM-LIF/metabolismo , Receptores de Progesterona/genética , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
15.
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
16.
Mol Cell ; 31(6): 835-49, 2008 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-18922467

RESUMEN

SRC-3/AIB1 is a master growth coactivator and oncogene, and phosphorylation activates it into a powerful coregulator. Dephosphorylation is a potential regulatory mechanism for SRC-3 function, but the identity of such phosphatases remains unexplored. Herein, we report that, using functional genomic screening of human Ser/Thr phosphatases targeting SRC-3's known phosphorylation sites, the phosphatases PDXP, PP1, and PP2A were identified to be key negative regulators of SRC-3 transcriptional coregulatory activity in steroid receptor signalings. PDXP and PP2A dephosphorylate SRC-3 and inhibit its ligand-dependent association with estrogen receptor. PP1 stabilizes SRC-3 protein by blocking its proteasome-dependent turnover through dephosphorylation of two previously unidentified phosphorylation sites (Ser101 and S102) required for activity. These two sites are located within a degron of SRC-3 and are primary determinants of SRC-3 turnover. Moreover, PP1 regulates the oncogenic cell proliferation and invasion functions of SRC-3 in breast cancer cells.


Asunto(s)
Fosfoproteínas/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Factores de Transcripción/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Regulación de la Expresión Génica , Genoma/genética , Células HeLa , Humanos , Coactivador 3 de Receptor Nuclear , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación , Fosfoserina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Proteína Fosfatasa 1/metabolismo , Proteína Fosfatasa 2/metabolismo , ARN Interferente Pequeño/metabolismo , Receptores de Estrógenos/metabolismo , Transducción de Señal , Termodinámica , Factores de Transcripción/genética , Transcripción Genética
17.
Mol Cell ; 29(4): 465-76, 2008 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-18313384

RESUMEN

SRC-3/AIB1 is a steroid receptor coactivator with potent growth-promoting activity, and its overexpression is sufficient to induce tumorigenesis. Previous studies indicate that the cellular level of SRC-3 is tightly regulated by both ubiquitin-dependent and ubiquitin-independent proteasomal degradation pathways. Atypical protein kinase C (aPKC) is frequently overexpressed in cancers. In the present study, we show that aPKC phosphorylates and specifically stabilizes SRC-3 in a selective ER-dependent manner. We further demonstrate that an acidic residue-rich region in SRC-3 is an important determinant for aPKC-mediated phosphorylation and stabilization. The mechanism of the aPKC-mediated stabilization appears due to a decreased interaction between SRC-3 and the C8 subunit of the 20S core proteasome, thus preventing SRC-3 degradation. Our results demonstrate a potent signaling mechanism for regulating SRC-3 levels in cells by coordinate enzymatic inhibition of both ubiquitin-dependent and ubiquitin-independent proteolytic pathways.


Asunto(s)
Histona Acetiltransferasas/metabolismo , Isoenzimas/metabolismo , Proteína Quinasa C/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Neoplasias de la Mama/metabolismo , Línea Celular , Retículo Endoplásmico/metabolismo , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Estrógenos/metabolismo , Femenino , Regulación de la Expresión Génica , Histona Acetiltransferasas/química , Histona Acetiltransferasas/genética , Humanos , Isoenzimas/genética , Ratones , Datos de Secuencia Molecular , Coactivador 3 de Receptor Nuclear , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteína Quinasa C/genética , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Alineación de Secuencia , Transactivadores/química , Transactivadores/genética , Factores de Transcripción/genética
18.
Semin Cell Dev Biol ; 24(10-12): 687-93, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23978678

RESUMEN

The formation of complex organisms is highly dependent on the differentiation of specialized mature cells from common stem/progenitor cells. The orphan nuclear receptors chicken ovalbumin upstream promoter transcription factors (COUP-TFs) are broadly, but not ubiquitously, expressed in multiple tissues throughout embryonic development and COUP-TFs are indispensible for proper organogenesis. Recently, growing evidence suggests a critical role of COUP-TFs in multiple aspects of stem/progenitor cell biology. In this review, we highlight the progress of COUP-TFs function and its underlying mechanism in driving stem/progenitor cell self-renewal, lineage specification, differentiation, maintenance, and cell identity in diverse tissue types. These studies provide novel insights into future clinical utilities of COUP-TFs in stem cell based therapies and in the management of diseases.


Asunto(s)
Factores de Transcripción COUP/metabolismo , Desarrollo Embrionario , Células Madre/metabolismo , Animales , Humanos
19.
Development ; 139(9): 1630-9, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22492355

RESUMEN

The development of the progenitor zones in the pallium, lateral ganglionic eminence (LGE) and medial ganglionic eminence (MGE) in the subpallium has been well studied; however, so far the role of the caudal ganglionic eminence (CGE), a posterior subpallial domain, in telencephalon patterning remains poorly understood. COUP-TFII, an orphan nuclear receptor, is preferentially expressed in the CGE. We generated COUP-TFII mouse mutants, using Rx-Cre (RxCre;COUP-TFII(F/F)), to study its function in telencephalon development. In these mutants, we found severe defects in the formation of the amygdala complex, including the lateral (LA), basolateral (BLA) and basomedial (BMA) amygdala nuclei. Molecular analysis provided evidence that the migration of CGE-derived Pax6(+) cells failed to settle into the BMA nucleus, owing to reduced expression of neuropilin 1 (Nrp1) and Nrp2, two semaphorin receptors that regulate neuronal cell migration and axon guidance. Our ChIP assays revealed that Nrp1 and Nrp2 genes are the direct targets of COUP-TFII in the telencephalon in vivo. Furthermore, our results showed that the coordinated development between the CGE originated subpallial population (Pax6(+) cells) and pallial populations (Tbr1(+) and Lhx2(+) cells) was essential for patterning the amygdala assembly. Our study presented novel genetic evidence that the caudal ganglionic eminence, a distinct subpallial progenitor zone, contributes cells to the basal telencephalon, such as the BMA nucleus.


Asunto(s)
Amígdala del Cerebelo/embriología , Factor de Transcripción COUP II/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Neuropilina-1/metabolismo , Neuropilina-2/metabolismo , Animales , Factor de Transcripción COUP II/genética , Inmunoprecipitación de Cromatina , Cartilla de ADN/genética , Regulación del Desarrollo de la Expresión Génica/genética , Inmunohistoquímica , Ratones , Ratones Mutantes , Reacción en Cadena en Tiempo Real de la Polimerasa
20.
Development ; 139(13): 2330-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22669823

RESUMEN

Development of the metanephric kidney in mammals requires complex reciprocal tissue interactions between the ureteric epithelium and the mesenchyme. It is believed that Gdnf, produced in the metanephric mesenchyme, activates Ret signaling in the Wolffian duct to initiate the formation of the metanephros. However, the molecular mechanism for induction of Gdnf in the metanephric mesenchyme is not completely defined. Previous studies demonstrated that during the early stages of kidney development, loss of Osr1, Eya1, Pax2 or Wt1 gene function in the metanephric mesenchyme compromises the formation of the kidney. Moreover, it has been shown that the Hox11-Eya1-Pax2 complex activates the expression of Six2 and Gdnf in the metanephric mesenchyme to drive nephrogenesis. Here, we demonstrate that the orphan nuclear receptor chicken ovalbumin upstream promoter transcription factor II (COUP-TFII, also known as Nr2f2) is required for the specification of the metanephric mesenchyme. Deletion of COUP-TFII at E7.5 results in improper differentiation of the metanephric mesenchyme and absence of essential developmental regulators, such as Eya1, Six2, Pax2 and Gdnf. Importantly, we show that COUP-TFII directly regulates the expression of both Eya1 and Wt1 in the metanephric mesenchyme. Our findings reveal, for the first time, that COUP-TFII plays a central role in the specification of metanephric fate and in the maintenance of metanephric mesenchyme proliferation and survival by acting as a crucial regulator of Eya1 and Wt1 expression.


Asunto(s)
Factor de Transcripción COUP II/fisiología , Riñón/crecimiento & desarrollo , Células Madre Mesenquimatosas/fisiología , Mesodermo/crecimiento & desarrollo , Animales , Factor de Transcripción COUP II/genética , Diferenciación Celular/fisiología , Supervivencia Celular/fisiología , Desarrollo Embrionario , Femenino , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Factor Neurotrófico Derivado de la Línea Celular Glial/análisis , Proteínas de Homeodominio/biosíntesis , Péptidos y Proteínas de Señalización Intracelular , Riñón/metabolismo , Masculino , Mesodermo/metabolismo , Ratones , Proteínas Nucleares/biosíntesis , Organogénesis/fisiología , Factor de Transcripción PAX2/análisis , Embarazo , Proteínas Tirosina Fosfatasas/biosíntesis , Factores de Transcripción/biosíntesis
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda