Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Oncogene ; 43(6): 379-387, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38129506

ABSTRACT

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.


Subject(s)
ERRalpha Estrogen-Related Receptor , Neoplasms , Humans , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Transcription Factors , Neoplasms/genetics , Cell Movement/genetics
2.
Int J Mol Sci ; 24(5)2023 Feb 21.
Article in English | MEDLINE | ID: mdl-36901694

ABSTRACT

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.


Subject(s)
Gene Regulatory Networks , Receptors, Estrogen , Animals , Gene Expression Regulation , Mammals/metabolism , Receptors, Estrogen/metabolism , Transcription Factors/metabolism , Humans , ERRalpha Estrogen-Related Receptor
3.
Cells ; 12(1)2023 01 03.
Article in English | MEDLINE | ID: mdl-36611993

ABSTRACT

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.


Subject(s)
Protein Interaction Maps , Humans , Microscopy, Fluorescence/methods
4.
Endocr Relat Cancer ; 29(8): 451-465, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35583188

ABSTRACT

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.


Subject(s)
Breast Neoplasms , Proto-Oncogene Proteins , Breast Neoplasms/pathology , Estrogens/therapeutic use , Female , Gene Silencing , Humans , MCF-7 Cells , Mechanistic Target of Rapamycin Complex 1 , Oncogenes , Proto-Oncogene Proteins/genetics
5.
Cancer Gene Ther ; 29(10): 1429-1438, 2022 10.
Article in English | MEDLINE | ID: mdl-35379907

ABSTRACT

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.


Subject(s)
Actins , Focal Adhesions , Actin Depolymerizing Factors/metabolism , Actins/genetics , Actins/metabolism , Cell Movement/physiology , Focal Adhesions/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , ERRalpha Estrogen-Related Receptor
6.
Sci Rep ; 12(1): 3826, 2022 03 09.
Article in English | MEDLINE | ID: mdl-35264626

ABSTRACT

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.


Subject(s)
Breast Neoplasms , Receptors, Estrogen , Breast Neoplasms/genetics , DEAD-box RNA Helicases/genetics , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation , Histone-Lysine N-Methyltransferase/metabolism , Humans , Promoter Regions, Genetic , RNA-Binding Proteins/metabolism , Receptors, Estrogen/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , ERRalpha Estrogen-Related Receptor
7.
Article in English | MEDLINE | ID: mdl-32973678

ABSTRACT

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.


Subject(s)
Endocrine Disruptors/metabolism , Environmental Pollutants/metabolism , Gene Expression Regulation , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Humans , Receptors, Estrogen/genetics , Receptors, G-Protein-Coupled/genetics
8.
Article in English | MEDLINE | ID: mdl-32922363

ABSTRACT

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.


Subject(s)
Endocrine Disruptors/chemistry , Endocrine Disruptors/pharmacology , Fibroblasts/cytology , Receptors, Estrogen/antagonists & inhibitors , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Cells, Cultured , Endocrine Disruptors/classification , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans
9.
Cell Mol Life Sci ; 77(22): 4573-4579, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32448995

ABSTRACT

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.


Subject(s)
Gene Expression/genetics , Receptors, Estrogen/genetics , Animals , Humans , Ligands , Protein Processing, Post-Translational/genetics , Transcription, Genetic/genetics
11.
Sci Rep ; 8(1): 10041, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29968728

ABSTRACT

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.


Subject(s)
Histone Demethylases/metabolism , Nuclear Respiratory Factor 1/metabolism , Receptors, Estrogen/metabolism , Cell Line , Chromatin/metabolism , Gene Expression , Gene Expression Regulation , HEK293 Cells , Histones/metabolism , Humans , Promoter Regions, Genetic , Transcription Factors/metabolism , Transcription Initiation Site , Transcription, Genetic , Transcriptional Activation , ERRalpha Estrogen-Related Receptor
12.
Sci Signal ; 11(536)2018 06 26.
Article in English | MEDLINE | ID: mdl-29945885

ABSTRACT

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.


Subject(s)
Autophagy , Mitochondria/physiology , Mitochondrial Dynamics , Mitophagy , Receptors, Estrogen/metabolism , Thyroid Hormone Receptors beta/physiology , Animals , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Cells, Cultured , Dynamins/genetics , Dynamins/metabolism , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Phosphorylation , Receptors, Estrogen/genetics , ERRalpha Estrogen-Related Receptor
13.
PLoS One ; 12(11): e0188871, 2017.
Article in English | MEDLINE | ID: mdl-29190800

ABSTRACT

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.


Subject(s)
Breast Neoplasms/genetics , Histone Demethylases/metabolism , Receptors, Estrogen/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Demethylation , Female , Histone Demethylases/genetics , Humans , Receptors, Estrogen/genetics , ERRalpha Estrogen-Related Receptor
14.
Proc Natl Acad Sci U S A ; 114(15): 3909-3914, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28348226

ABSTRACT

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.


Subject(s)
Histone Demethylases/metabolism , Histones/metabolism , Receptors, Estrogen/metabolism , Cell Movement , Gene Expression Regulation , HEK293 Cells , Histone Demethylases/genetics , Humans , Lysine/metabolism , Matrix Metalloproteinase 1/metabolism , Methylation , Promoter Regions, Genetic , Receptors, Estrogen/genetics , Transcription Initiation Site , ERRalpha Estrogen-Related Receptor
15.
Metallomics ; 8(11): 1177-1184, 2016 11 09.
Article in English | MEDLINE | ID: mdl-27500357

ABSTRACT

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.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Hepatocellular/metabolism , Copper/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/metabolism , Liver Neoplasms/metabolism , Carcinoma, Hepatocellular/pathology , Cell Fractionation , Cell Survival , Copper Radioisotopes/metabolism , Hep G2 Cells , Humans , Liver Neoplasms/pathology
16.
Cell Mol Life Sci ; 73(20): 3781-7, 2016 10.
Article in English | MEDLINE | ID: mdl-27514376

ABSTRACT

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.


Subject(s)
Bone Resorption/drug therapy , Molecular Targeted Therapy , Receptors, Estrogen/metabolism , Animals , Bone Resorption/physiopathology , Calcification, Physiologic , Humans , Ligands , Models, Biological
17.
PLoS One ; 11(5): e0156445, 2016.
Article in English | MEDLINE | ID: mdl-27227989

ABSTRACT

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.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Neoplasm Proteins/biosynthesis , Prostatic Neoplasms/metabolism , RNA, Neoplasm/metabolism , Receptors, Estrogen/biosynthesis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Male , MicroRNAs/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Neoplasm/genetics , Receptors, Estrogen/genetics , ERRalpha Estrogen-Related Receptor
18.
Mol Cell Endocrinol ; 432: 37-43, 2016 09 05.
Article in English | MEDLINE | ID: mdl-26206717

ABSTRACT

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.


Subject(s)
Bone and Bones/cytology , Cell Lineage , Receptors, Estrogen/metabolism , Animals , Humans , Mesoderm/cytology , Models, Biological , Receptors, Cytoplasmic and Nuclear/metabolism , ERRalpha Estrogen-Related Receptor
19.
Immunity ; 43(1): 80-91, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26200012

ABSTRACT

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.


Subject(s)
Cysteine Endopeptidases/biosynthesis , Inflammation/immunology , Intracellular Signaling Peptides and Proteins/biosynthesis , Macrophages/metabolism , Receptors, Estrogen/genetics , Toll-Like Receptor 4/immunology , Acetylation , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Cells, Cultured , Cysteine Endopeptidases/genetics , Enzyme Activation/genetics , Glycolysis/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Lipopolysaccharides , Macrophages/immunology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , NAD/metabolism , Oxidative Phosphorylation , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , Shock, Septic/immunology , Signal Transduction , Sirtuin 1/metabolism , TNF Receptor-Associated Factor 6/metabolism , Transcription Factor RelA/metabolism , Transcription, Genetic/genetics , Tumor Necrosis Factor alpha-Induced Protein 3 , Ubiquitination , ERRalpha Estrogen-Related Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...