Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Oncogene ; 43(6): 379-387, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38129506

RESUMEN

Cell migration is essential throughout the life of multicellular organisms, and largely depends on the spatial and temporal regulation of cytoskeletal dynamics, cell adhesion and signal transduction. Interestingly, Estrogen-related receptor alpha (ERRα) has been identified as a major regulator of cell migration in both physiological and pathological conditions. ERRα is an orphan member of the nuclear hormone receptor superfamily of transcription factors and displays many biological functions. ERRα is a global regulator of energy metabolism, and it is also highly involved in bone homeostasis, development, differentiation, immunity and cancer progression. Importantly, in some instances, the regulation of these biological processes relies on the ability to orchestrate cell movements. Therefore, this review describes how ERRα-mediated cell migration contributes not only to tissue homeostasis but also to tumorigenesis and metastasis, and highlights the molecular and cellular mechanisms by which ERRα finely controls the cell migratory potential.


Asunto(s)
Receptor Relacionado con Estrógeno ERRalfa , Neoplasias , Humanos , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Factores de Transcripción , Neoplasias/genética , Movimiento Celular/genética
2.
Int J Mol Sci ; 24(5)2023 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-36901694

RESUMEN

Estrogen-related receptors (ERRα, ß and γ in mammals) are orphan members of the nuclear receptor superfamily acting as transcription factors. ERRs are expressed in several cell types and they display various functions in normal and pathological contexts. Amongst others, they are notably involved in bone homeostasis, energy metabolism and cancer progression. In contrast to other nuclear receptors, the activities of the ERRs are apparently not controlled by a natural ligand but they rely on other means such as the availability of transcriptional co-regulators. Here we focus on ERRα and review the variety of co-regulators that have been identified by various means for this receptor and their reported target genes. ERRα cooperates with distinct co-regulators to control the expression of distinct sets of target genes. This exemplifies the combinatorial specificity of transcriptional regulation that induces discrete cellular phenotypes depending on the selected coregulator. We finally propose an integrated view of the ERRα transcriptional network.


Asunto(s)
Redes Reguladoras de Genes , Receptores de Estrógenos , Animales , Regulación de la Expresión Génica , Mamíferos/metabolismo , Receptores de Estrógenos/metabolismo , Factores de Transcripción/metabolismo , Humanos , Receptor Relacionado con Estrógeno ERRalfa
3.
Cells ; 12(1)2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36611993

RESUMEN

Biological pathways rely on the formation of intricate protein interaction networks called interactomes. Getting a comprehensive map of interactomes implies the development of tools that allow one to capture transient and low-affinity protein-protein interactions (PPIs) in live conditions. Here we presented an experimental strategy: the Cell-PCA (cell-based protein complementation assay), which was based on bimolecular fluorescence complementation (BiFC) for ORFeome-wide screening of proteins that interact with different bait proteins in the same live cell context, by combining high-throughput sequencing method. The specificity and sensitivity of the Cell-PCA was established by using a wild-type and a single-amino-acid-mutated HOXA9 protein, and the approach was subsequently applied to seven additional human HOX proteins. These proof-of-concept experiments revealed novel molecular properties of HOX interactomes and led to the identification of a novel cofactor of HOXB13 that promoted its proliferative activity in a cancer cell context. Taken together, our work demonstrated that the Cell-PCA was pertinent for revealing and, importantly, comparing the interactomes of different or highly related bait proteins in the same cell context.


Asunto(s)
Mapas de Interacción de Proteínas , Humanos , Microscopía Fluorescente/métodos
4.
Endocr Relat Cancer ; 29(8): 451-465, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35583188

RESUMEN

Menin, encoded by the MEN1 gene, has been identified as a critical factor regulating ESR1 transcription, playing an oncogenic role in ER+ breast cancer (BC) cells. Here, we further dissected the consequences of menin inactivation in ER+ BC cells by focusing on factors within two major pathways involved in BC, mTOR and MYC. MEN1 silencing in MCF7 and T-47D resulted in an increase in phosphor-p70S6K1, phosphor-p85S6K1 and phosphor-4EBP1 expression. The use of an AKT inhibitor inhibited the activation of S6K1 and S6RP triggered by MEN1 knockdown (KD). Moreover, MEN1 silencing in ER+ BC cells led to increased formation of the eIF4E and 4G complex. Clinical studies showed that patients with menin-low breast cancer receiving tamoxifen plus everolimus displayed a trend toward better overall survival. Importantly, MEN1 KD in MCF7 and T-47D cells led to reduced MYC expression. ChIP analysis demonstrated that menin bound not only to the MYC promoter but also to its 5' enhancer. Furthermore, E2-treated MEN1 KD MCF7 cells displayed a decrease in MYC activation, suggesting its role in estrogen-mediated MYC transcription. Finally, expression data mining in tumors revealed a correlation between the expression of MEN1 mRNA and that of several mTORC1 components and targets and a significant inverse correlation between MEN1 and two MYC inhibitory factors, MYCBP2 and MYCT1, in ER+ BC. The current work thus highlights altered mTORC1 and MYC pathways after menin inactivation in ER+ BC cells, providing insight into the crosstalk between menin, mTORC1 and MYC in ER+ BC.


Asunto(s)
Neoplasias de la Mama , Proteínas Proto-Oncogénicas , Neoplasias de la Mama/patología , Estrógenos/uso terapéutico , Femenino , Silenciador del Gen , Humanos , Células MCF-7 , Diana Mecanicista del Complejo 1 de la Rapamicina , Oncogenes , Proteínas Proto-Oncogénicas/genética
5.
Cancer Gene Ther ; 29(10): 1429-1438, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35379907

RESUMEN

Cell migration depends on the dynamic organisation of the actin cytoskeleton and assembly and disassembly of focal adhesions (FAs). However, the precise mechanisms coordinating these processes remain poorly understood. We previously identified the oestrogen-related receptor α (ERRα) as a major regulator of cell migration. Here, we show that loss of ERRα leads to abnormal accumulation of actin filaments that is associated with an increased level of inactive form of the actin-depolymerising factor cofilin. We further show that ERRα depletion decreases cell adhesion and results in defective FA formation and turnover. Interestingly, specific inhibition of the RhoA-ROCK-LIMK-cofilin pathway rescues the actin polymerisation defects resulting from ERRα silencing, but not cell adhesion. Instead, we found that MAP4K4 is a direct target of ERRα and down-regulation of its activity rescues cell adhesion and FA formation in the ERRα-depleted cells. Altogether, our results highlight a crucial role of ERRα in coordinating the dynamic of actin network and FAs through the independent regulation of the RhoA and MAP4K4 pathways.


Asunto(s)
Actinas , Adhesiones Focales , Factores Despolimerizantes de la Actina/metabolismo , Actinas/genética , Actinas/metabolismo , Movimiento Celular/fisiología , Adhesiones Focales/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Serina-Treonina Quinasas , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Receptor Relacionado con Estrógeno ERRalfa
6.
Sci Rep ; 12(1): 3826, 2022 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-35264626

RESUMEN

Estrogen related receptors are orphan members of the nuclear receptor superfamily acting as transcription factors (TFs). In contrast to classical nuclear receptors, the activities of the ERRs are not controlled by a natural ligand. Regulation of their activities thus relies on availability of transcriptional co-regulators. In this paper, we focus on ERRα, whose involvement in cancer progression has been broadly demonstrated. We propose a new approach to identify potential co-activators, starting from previously identified ERRα-activated genes in a breast cancer (BC) cell line. Considering mRNA gene expression from two sets of human BC cells as major endpoint, we used sparse partial least squares modeling to uncover new transcriptional regulators associated with ERRα. Among them, DDX21, MYBBP1A, NFKB1, and SETD7 are functionally relevant in MDA-MB-231 cells, specifically activating the expression of subsets of ERRα-activated genes. We studied SET7 in more details and showed its co-localization with ERRα and its ERRα-dependent transcriptional and phenotypic effects. Our results thus demonstrate the ability of a modeling approach to identify new transcriptional partners from gene expression. Finally, experimental results show that ERRα cooperates with distinct co-regulators to control the expression of distinct sets of target genes, thus reinforcing the combinatorial specificity of transcription.


Asunto(s)
Neoplasias de la Mama , Receptores de Estrógenos , Neoplasias de la Mama/genética , ARN Helicasas DEAD-box/genética , Proteínas de Unión al ADN/genética , Femenino , Regulación de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Regiones Promotoras Genéticas , Proteínas de Unión al ARN/metabolismo , Receptores de Estrógenos/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Receptor Relacionado con Estrógeno ERRalfa
7.
Artículo en Inglés | MEDLINE | ID: mdl-32973678

RESUMEN

Endocrine-disrupting chemicals (EDCs) are exogenous chemicals that interfere with endogenous hormonal systems at various levels, resulting in adverse health effects. EDCs belong to diverse chemical families and can accumulate in the environment, diet and body fluids, with different levels of persistence. Their action can be mediated by several receptors, including members of the nuclear receptor family, such as estrogen and androgen receptors. The G protein-coupled estrogen receptor (GPER), a seven-transmembrane domain receptor, has also attracted attention as a potential target of EDCs. This review summarizes our current knowledge concerning GPER as a mediator of EDCs' effects.


Asunto(s)
Disruptores Endocrinos/metabolismo , Contaminantes Ambientales/metabolismo , Regulación de la Expresión Génica , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Humanos , Receptores de Estrógenos/genética , Receptores Acoplados a Proteínas G/genética
8.
Artículo en Inglés | MEDLINE | ID: mdl-32922363

RESUMEN

Endocrine-disrupting chemicals (EDCs) are exogenous compounds that impact endogenous hormonal systems, resulting in adverse health effects. These chemicals can exert their actions by interfering with several pathways. Simple biological systems to determine whether EDCs act positively or negatively on a given receptor are often lacking. Here we describe a low-to-middle throughput method to screen the agonist/antagonist potential of EDCs specifically on the GPER membrane estrogen receptor. Application of this assay to 23 candidate EDCs from different chemical families reveals the existence of six agonists and six antagonists.


Asunto(s)
Disruptores Endocrinos/química , Disruptores Endocrinos/farmacología , Fibroblastos/citología , Receptores de Estrógenos/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Células Cultivadas , Disruptores Endocrinos/clasificación , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos
9.
Cell Mol Life Sci ; 77(22): 4573-4579, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32448995

RESUMEN

Estrogen related receptors (ERRα, ß and γ in mammals) are orphan members of the nuclear receptor superfamily acting as transcription factors. ERRs are expressed in several tissues and cells and they display various physiological and pathological functions, controlling, amongst others and depending on the receptor, bone homeostasis, energy metabolism, embryonic stem cell pluripotency, and cancer progression. In contrast to classical nuclear receptors, the activities of the ERRs are not controlled by a natural ligand. Regulation of their activities thus rely on other means such as post-translational modification or availability of transcriptional co-regulators. In addition, regulation of their mere expression under given physiological or pathological conditions is a particularly important level of control. Here we discuss the mechanisms involved in the regulation of ERRs expression and the reported means to impact on it using pharmacological approaches.


Asunto(s)
Expresión Génica/genética , Receptores de Estrógenos/genética , Animales , Humanos , Ligandos , Procesamiento Proteico-Postraduccional/genética , Transcripción Genética/genética
11.
Sci Rep ; 8(1): 10041, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29968728

RESUMEN

Lysine-specific demethylase 1 (LSD1) exerts dual effects on histone H3, promoting transcriptional repression via Lys4 (H3K4) demethylation or transcriptional activation through Lys9 (H3K9) demethylation. These activities are often exerted at transcriptional start sites (TSSs) and depend on the type of enhancer-bound transcription factor (TFs) with which LSD1 interacts. In particular, the Estrogen-Receptor Related α (ERRα) TF interacts with LSD1 and switches its activities toward H3K9 demethylation, resulting in transcriptional activation of a set of common target genes. However, how are the LSD1-TF and, in particular LSD1-ERRα, complexes determined to act at TSSs is not understood. Here we show that promoter-bound nuclear respiratory factor 1 (NRF1), but not ERRα, is essential to LSD1 recruitment at the TSSs of positive LSD1-ERRα targets. In contrast to ERRα, NRF1 does not impact on the nature of LSD1 enzymatic activity. We propose a three factor model, in which the LSD1 histone modifier requires a TSS tethering factor (NRF1) as well as an activity inducer (ERRα) to transcriptionally activate common targets. The relevance of this common network is illustrated by functional data, showing that all three factors are required for cell invasion in an MMP1 (Matrix MetalloProtease 1)-dependent manner, the expression of which is regulated by NRF1/LSD1/ERRα-mediated H3K9me2 demethylation.


Asunto(s)
Histona Demetilasas/metabolismo , Factor Nuclear 1 de Respiración/metabolismo , Receptores de Estrógenos/metabolismo , Línea Celular , Cromatina/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Células HEK293 , Histonas/metabolismo , Humanos , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo , Sitio de Iniciación de la Transcripción , Transcripción Genética , Activación Transcripcional , Receptor Relacionado con Estrógeno ERRalfa
12.
Sci Signal ; 11(536)2018 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-29945885

RESUMEN

Thyroid hormone receptor ß1 (THRB1) and estrogen-related receptor α (ESRRA; also known as ERRα) both play important roles in mitochondrial activity. To understand their potential interactions, we performed transcriptome and ChIP-seq analyses and found that many genes that were co-regulated by both THRB1 and ESRRA were involved in mitochondrial metabolic pathways. These included oxidative phosphorylation (OXPHOS), the tricarboxylic acid (TCA) cycle, and ß-oxidation of fatty acids. TH increased ESRRA expression and activity in a THRB1-dependent manner through the induction of the transcriptional coactivator PPARGC1A (also known as PGC1α). Moreover, TH induced mitochondrial biogenesis, fission, and mitophagy in an ESRRA-dependent manner. TH also induced the expression of the autophagy-regulating kinase ULK1 through ESRRA, which then promoted DRP1-mediated mitochondrial fission. In addition, ULK1 activated the docking receptor protein FUNDC1 and its interaction with the autophagosomal protein MAP1LC3B-II to induce mitophagy. siRNA knockdown of ESRRA, ULK1, DRP1, or FUNDC1 inhibited TH-induced autophagic clearance of mitochondria through mitophagy and decreased OXPHOS. These findings show that many of the mitochondrial actions of TH are mediated through stimulation of ESRRA expression and activity, and co-regulation of mitochondrial turnover through the PPARGC1A-ESRRA-ULK1 pathway is mediated by their regulation of mitochondrial fission and mitophagy. Hormonal or pharmacologic induction of ESRRA expression or activity could improve mitochondrial quality in metabolic disorders.


Asunto(s)
Autofagia , Mitocondrias/fisiología , Dinámicas Mitocondriales , Mitofagia , Receptores de Estrógenos/metabolismo , Receptores beta de Hormona Tiroidea/fisiología , Animales , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Células Cultivadas , Dinaminas/genética , Dinaminas/metabolismo , Humanos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Fosforilación , Receptores de Estrógenos/genética , Receptor Relacionado con Estrógeno ERRalfa
13.
PLoS One ; 12(11): e0188871, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29190800

RESUMEN

The LSD1 histone demethylase is highly expressed in breast tumors where it constitutes a factor of poor prognosis and promotes traits of cancer aggressiveness such as cell invasiveness. Recent work has shown that the Estrogen-Related Receptor α (ERRα) induces LSD1 to demethylate the Lys 9 of histone H3. This results in the transcriptional activation of a number of common target genes, several of which being involved in cellular invasion. High expression of ERRα protein is also a factor of poor prognosis in breast tumors. Here we show that, independently of its demethylase activities, LSD1 protects ERRα from ubiquitination, resulting in overexpression of the latter protein. Our data also suggests that the elevation of LSD1 mRNA and protein in breast cancer (as compared to normal tissue) may be a key event to increase ERRα protein, independently of its corresponding mRNA.


Asunto(s)
Neoplasias de la Mama/genética , Histona Demetilasas/metabolismo , Receptores de Estrógenos/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Desmetilación , Femenino , Histona Demetilasas/genética , Humanos , Receptores de Estrógenos/genética , Receptor Relacionado con Estrógeno ERRalfa
14.
Proc Natl Acad Sci U S A ; 114(15): 3909-3914, 2017 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-28348226

RESUMEN

Lysine Specific Demethylase 1 (LSD1) removes mono- and dimethyl groups from lysine 4 of histone H3 (H3K4) or H3K9, resulting in repressive or activating (respectively) transcriptional histone marks. The mechanisms that control the balance between these two antagonist activities are not understood. We here show that LSD1 and the orphan nuclear receptor estrogen-related receptor α (ERRα) display commonly activated genes. Transcriptional activation by LSD1 and ERRα involves H3K9 demethylation at the transcriptional start site (TSS). Strikingly, ERRα is sufficient to induce LSD1 to demethylate H3K9 in vitro. The relevance of this mechanism is highlighted by functional data. LSD1 and ERRα coregulate several target genes involved in cell migration, including the MMP1 matrix metallo-protease, also activated through H3K9 demethylation at the TSS. Depletion of LSD1 or ERRα reduces the cellular capacity to invade the extracellular matrix, a phenomenon that is rescued by MMP1 reexpression. Altogether our results identify a regulatory network involving a direct switch in the biochemical activities of a histone demethylase, leading to increased cell invasion.


Asunto(s)
Histona Demetilasas/metabolismo , Histonas/metabolismo , Receptores de Estrógenos/metabolismo , Movimiento Celular , Regulación de la Expresión Génica , Células HEK293 , Histona Demetilasas/genética , Humanos , Lisina/metabolismo , Metaloproteinasa 1 de la Matriz/metabolismo , Metilación , Regiones Promotoras Genéticas , Receptores de Estrógenos/genética , Sitio de Iniciación de la Transcripción , Receptor Relacionado con Estrógeno ERRalfa
15.
Metallomics ; 8(11): 1177-1184, 2016 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-27500357

RESUMEN

Hepatocellular carcinoma (HCC) is the most frequent type of primary liver cancer, with increasing incidence worldwide. The unrestrained proliferation of tumour cells leads to tumour hypoxia which in turn promotes cancer aggressiveness. While changes in the concentration of copper (Cu) have long been observed upon cancerization, we have recently reported that the isotopic composition of copper is also altered in several types of cancer. In particular, we showed that in hepatocellular carcinoma, tumour tissue contains heavier copper compared to the surrounding parenchyma. However, the reasons behind such isotopic signature remained elusive. Here we show that hypoxia causes heavy copper enrichment in several human cell lines. We also demonstrate that this effect of hypoxia is pH, HIF-1 and -2 independent. Our data identify a previously unrecognized cellular process associated with hypoxia, and suggests that in vivo tumour hypoxia determines copper isotope fractionation in HCC and other solid cancers.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinoma Hepatocelular/metabolismo , Cobre/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Neoplasias Hepáticas/metabolismo , Carcinoma Hepatocelular/patología , Fraccionamiento Celular , Supervivencia Celular , Radioisótopos de Cobre/metabolismo , Células Hep G2 , Humanos , Neoplasias Hepáticas/patología
16.
Cell Mol Life Sci ; 73(20): 3781-7, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27514376

RESUMEN

Bone loss and the resulting skeletal fragility is induced by several pathological or natural conditions, the most prominent of which being aging as well as the decreased levels of circulating estrogens in post-menopause females. To date, most treatments against bone loss aim at preventing excess bone resorption. We here summarize data indicating that the estrogen-related receptors (ERRs) α and γ prevent bone formation. Inhibiting these receptors may thus constitute an anabolic approach by increasing bone formation.


Asunto(s)
Resorción Ósea/tratamiento farmacológico , Terapia Molecular Dirigida , Receptores de Estrógenos/metabolismo , Animales , Resorción Ósea/fisiopatología , Calcificación Fisiológica , Humanos , Ligandos , Modelos Biológicos
17.
PLoS One ; 11(5): e0156445, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27227989

RESUMEN

MicroRNA-135a (miR-135a) down-modulates parameters of cancer progression and its expression is decreased in metastatic breast cancers (as compared to non-metastatic tumors) as well as in prostate tumors relative to normal tissue. These expression and activity patterns are opposite to those of the Estrogen-Related Receptor α (ERRα), an orphan member of the nuclear receptor family. Indeed high expression of ERRα correlates with poor prognosis in breast and prostate cancers, and the receptor promotes various traits of cancer aggressiveness including cell invasion. Here we show that miR-135a down-regulates the expression of ERRα through specific sequences of its 3'UTR. As a consequence miR-135a also reduces the expression of downstream targets of ERRα. miR-135a also decreases cell invasive potential in an ERRα-dependent manner. Our results suggest that the decreased expression of miR-135a in metastatic tumors leads to elevated ERRα expression, resulting in increased cell invasion capacities.


Asunto(s)
Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , MicroARNs/metabolismo , Proteínas de Neoplasias/biosíntesis , Neoplasias de la Próstata/metabolismo , ARN Neoplásico/metabolismo , Receptores de Estrógenos/biosíntesis , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Masculino , MicroARNs/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , ARN Neoplásico/genética , Receptores de Estrógenos/genética , Receptor Relacionado con Estrógeno ERRalfa
18.
Mol Cell Endocrinol ; 432: 37-43, 2016 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-26206717

RESUMEN

Bone loss is naturally occurring in aging males and females and exacerbated in the latter after menopause, altogether leading to cumulative skeleton fragility and increased fracture risk. Two types of therapeutic strategies can be envisioned to counteract age- or menopause-associated bone loss, aiming at either reducing bone resorption exerted by osteoclasts or, alternatively, promoting bone formation by osteoblasts. We here summarize data suggesting that inhibition of the Estrogen-Related Receptors α and/or γ could promote bone formation and compensate for bone loss induced by ageing or estrogen-deficiency.


Asunto(s)
Huesos/citología , Linaje de la Célula , Receptores de Estrógenos/metabolismo , Animales , Humanos , Mesodermo/citología , Modelos Biológicos , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptor Relacionado con Estrógeno ERRalfa
19.
Immunity ; 43(1): 80-91, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26200012

RESUMEN

The orphan nuclear receptor estrogen-related receptor α (ERRα; NR3B1) is a key metabolic regulator, but its function in regulating inflammation remains largely unknown. Here, we demonstrate that ERRα negatively regulates Toll-like receptor (TLR)-induced inflammation by promoting Tnfaip3 transcription and fine-tuning of metabolic reprogramming in macrophages. ERRα-deficient (Esrra(-/-)) mice showed increased susceptibility to endotoxin-induced septic shock, leading to more severe pro-inflammatory responses than control mice. ERRα regulated macrophage inflammatory responses by directly binding the promoter region of Tnfaip3, a deubiquitinating enzyme in TLR signaling. In addition, Esrra(-/-) macrophages showed an increased glycolysis, but impaired mitochondrial respiratory function and biogenesis. Further, ERRα was required for the regulation of NF-κB signaling by controlling p65 acetylation via maintenance of NAD(+) levels and sirtuin 1 activation. These findings unravel a previously unappreciated role for ERRα as a negative regulator of TLR-induced inflammatory responses through inducing Tnfaip3 transcription and controlling the metabolic reprogramming.


Asunto(s)
Cisteína Endopeptidasas/biosíntesis , Inflamación/inmunología , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Macrófagos/metabolismo , Receptores de Estrógenos/genética , Receptor Toll-Like 4/inmunología , Acetilación , Animales , Calcio/metabolismo , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Células Cultivadas , Cisteína Endopeptidasas/genética , Activación Enzimática/genética , Glucólisis/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Lipopolisacáridos , Macrófagos/inmunología , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/metabolismo , NAD/metabolismo , Fosforilación Oxidativa , Regiones Promotoras Genéticas/genética , Regiones Promotoras Genéticas/inmunología , Choque Séptico/inmunología , Transducción de Señal , Sirtuina 1/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Factor de Transcripción ReIA/metabolismo , Transcripción Genética/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Ubiquitinación , Receptor Relacionado con Estrógeno ERRalfa
20.
PLoS One ; 10(3): e0120672, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25781607

RESUMEN

The post-menopausal decrease in estrogen circulating levels results in rapid skin deterioration pointing out to a protective effect exerted by these hormones. The identity of the skin cell type responding to estrogens is unclear as are the cellular and molecular processes they elicit. Here, we reported that lack of estrogens induces rapid re-organization of the human dermal fibroblast cytoskeleton resulting in striking cell shape change. This morphological change was accompanied by a spatial re-organization of focal adhesion and a substantial reduction of their number as evidenced by vinculin and actin co-staining. Cell morphology and cytoskeleton organization was fully restored upon 17ß-estradiol (E2) addition. Treatment with specific ER antagonists and cycloheximide respectively showed that the E2 acts independently of the classical Estrogen Receptors and that cell shape change is mediated by non-genomic mechanisms. E2 treatment resulted in a rapid and transient activation of ERK1/2 but not Src or PI3K. We show that human fibroblasts express the non-classical E2 receptor GPR30 and that its agonist G-1 phenocopies the effect of E2. Inhibiting GPR30 through treatment with the G-15 antagonist or specific shRNA impaired E2 effects. Altogether, our data reveal a novel mechanism by which estrogens act on skin fibroblast by regulating cell shape through the non-classical G protein-coupled receptor GPR30 and ERK1/2 activation.


Asunto(s)
Estradiol/farmacología , Estrógenos/farmacología , Fibroblastos/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Adulto , Benzodioxoles/farmacología , Dermis , Receptor beta de Estrógeno/metabolismo , Femenino , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Quinolinas/farmacología , Receptores de Estrógenos/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...