351
|
Deblois G, Giguère V. Oestrogen-related receptors in breast cancer: control of cellular metabolism and beyond. Nat Rev Cancer 2013; 13:27-36. [PMID: 23192231 DOI: 10.1038/nrc3396] [Citation(s) in RCA: 181] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oestrogen-related receptors (ERRs) are orphan nuclear receptors that were initially investigated in breast cancer because of their structural relationship to oestrogen receptors. Recent data have shown that the ERRs control vast gene networks that are involved in glycolysis, glutaminolysis, oxidative phosphorylation, nutrient sensing and biosynthesis pathways. In the context of breast cancer, the ERRs affect cellular metabolism in a manner that promotes a Warburg-like phenotype. The ERRs also modulate breast cancer cell metabolism, growth and proliferation through the regulation of key oncoproteins. We discuss the value but also the implications of the complexity of targeting the ERRs for the development of cancer therapeutics.
Collapse
Affiliation(s)
- Geneviève Deblois
- Goodman Cancer Research Centre and Department of Biochemistry, McGill University, Montreal, Quebec H3A 1A3, Canada
| | | |
Collapse
|
352
|
Gaete L, Tchernitchin AN, Bustamante R, Villena J, Lemus I, Gidekel M, Cabrera G, Astorga P. Daidzein-estrogen interaction in the rat uterus and its effect on human breast cancer cell growth. J Med Food 2012; 15:1081-90. [PMID: 23216111 PMCID: PMC3523250 DOI: 10.1089/jmf.2011.0322] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 07/20/2012] [Indexed: 11/12/2022] Open
Abstract
Sex hormone replacement therapy provides several advantages in the quality of life for climacteric women. However, estrogen-induced cell proliferation in the uterus and mammary gland increases the risk of cancer development in these organs. The lower incidence of mammary cancer in Asian women as compared with Western women has been attributed to high intake of soy isoflavones, including genistein. We have previously shown that genistein induces an estradiol-like hypertrophy of uterine cells, but does not induce cell proliferation, uterine eosinophilia, or endometrial edema. It also inhibits estradiol-induced mitosis in uterine cells and hormone-induced uterine eosinophilia and endometrial edema. Nevertheless, genistein stimulates growth of human breast cancer cells in culture; therefore, it is not an ideal estrogen for use in hormone replacement therapy (HRD). The present study investigated the effect of another soy isoflavone, daidzein (subcutaneous, 0.066 mg/kg body weight), in the same animal model, and its effect on responses induced by subsequent treatment (1 h later) with estradiol-17β (E(2); subcutaneous, 0.33 mg/kg body weight). In addition, we investigated the effects of daidzein (1 μg/mL) or E(2) on the growth of human breast cancer cells in culture. Results indicate that daidzein stimulates growth of breast cancer cells and potentiates estrogen-induced cell proliferation in the uterus. We suggest caution for the use of daidzein or formulas containing this compound in HRD. Future research strategies should be addressed in the search for new phytoestrogens that selectively inhibit cell proliferation in the uterus and breast.
Collapse
Affiliation(s)
- Leonardo Gaete
- Institute of Biomedical Sciences (ICBM), School of Medicine, University of Chile, Santiago, Chile
| | - Andrei N. Tchernitchin
- Institute of Biomedical Sciences (ICBM), School of Medicine, University of Chile, Santiago, Chile
| | - Rodrigo Bustamante
- Institute of Biomedical Sciences (ICBM), School of Medicine, University of Chile, Santiago, Chile
| | - Joan Villena
- School of Medicine, Universidad de Valparaiso, Valparaiso, Chile
| | - Igor Lemus
- School of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | | | | | - Paola Astorga
- Institute of Biomedical Sciences (ICBM), School of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
353
|
Pérez-Schindler J, Summermatter S, Salatino S, Zorzato F, Beer M, Balwierz PJ, van Nimwegen E, Feige JN, Auwerx J, Handschin C. The corepressor NCoR1 antagonizes PGC-1α and estrogen-related receptor α in the regulation of skeletal muscle function and oxidative metabolism. Mol Cell Biol 2012; 32:4913-24. [PMID: 23028049 PMCID: PMC3510532 DOI: 10.1128/mcb.00877-12] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 09/23/2012] [Indexed: 01/08/2023] Open
Abstract
Skeletal muscle exhibits a high plasticity and accordingly can quickly adapt to different physiological and pathological stimuli by changing its phenotype largely through diverse epigenetic mechanisms. The nuclear receptor corepressor 1 (NCoR1) has the ability to mediate gene repression; however, its role in regulating biological programs in skeletal muscle is still poorly understood. We therefore studied the mechanistic and functional aspects of NCoR1 function in this tissue. NCoR1 muscle-specific knockout mice exhibited a 7.2% higher peak oxygen consumption (VO(2peak)), a 11% reduction in maximal isometric force, and increased ex vivo fatigue resistance during maximal stimulation. Interestingly, global gene expression analysis revealed a high overlap between the effects of NCoR1 deletion and peroxisome proliferator-activated receptor gamma (PPARγ) coactivator 1α (PGC-1α) overexpression on oxidative metabolism in muscle. Importantly, PPARβ/δ and estrogen-related receptor α (ERRα) were identified as common targets of NCoR1 and PGC-1α with opposing effects on the transcriptional activity of these nuclear receptors. In fact, the repressive effect of NCoR1 on oxidative phosphorylation gene expression specifically antagonizes PGC-1α-mediated coactivation of ERRα. We therefore delineated the molecular mechanism by which a transcriptional network controlled by corepressor and coactivator proteins determines the metabolic properties of skeletal muscle, thus representing a potential therapeutic target for metabolic diseases.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Mice, Knockout
- Mice, Transgenic
- Models, Biological
- Muscle Contraction/genetics
- Muscle Contraction/physiology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/physiology
- Nuclear Receptor Co-Repressor 1/deficiency
- Nuclear Receptor Co-Repressor 1/genetics
- Nuclear Receptor Co-Repressor 1/metabolism
- Oxidative Phosphorylation
- Oxygen Consumption
- PPAR delta/metabolism
- PPAR-beta/metabolism
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Estrogen/antagonists & inhibitors
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Trans-Activators/antagonists & inhibitors
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcription Factors
- ERRalpha Estrogen-Related Receptor
Collapse
Affiliation(s)
| | | | | | - Francesco Zorzato
- Departments of Anesthesia and Biomedicine, Basel University Hospital, Basel, Switzerland
- Department of Experimental and Diagnostic Medicine, University of Ferrara, Ferrara, Italy
| | - Markus Beer
- Biozentrum, University of Basel, Basel, Switzerland
| | - Piotr J. Balwierz
- Biozentrum, University of Basel, Basel, Switzerland
- Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Erik van Nimwegen
- Biozentrum, University of Basel, Basel, Switzerland
- Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Jérôme N. Feige
- Novartis Institute for Biomedical Research, Basel, Switzerland
- Laboratory for Integrative and Systems Physiology, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory for Integrative and Systems Physiology, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | | |
Collapse
|
354
|
Chang CY, McDonnell DP. Molecular pathways: the metabolic regulator estrogen-related receptor α as a therapeutic target in cancer. Clin Cancer Res 2012; 18:6089-95. [PMID: 23019305 PMCID: PMC3500439 DOI: 10.1158/1078-0432.ccr-11-3221] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The estrogen-related receptor α (ERRα) is an orphan member of the nuclear receptor superfamily of transcription factors whose activity is regulated by the expression level and/or activity of its obligate coregulators, peroxisome proliferator-activated receptor γ coactivator-1 α and β (PGC-1α or PGC-1β). Under normal physiologic conditions, and in responding to different environmental stimuli, the ERRα/PGC-1 complex is involved in regulating metabolic homeostasis under conditions of high energy demand in brown adipocytes, proliferating T cells, and muscle. Interestingly, increased expression and activity of the ERRα/PGC-1 axis has also been shown to correlate with unfavorable clinical outcomes in both breast and ovarian tumors. The observation that ERRα activity is manifest in all breast tumor subtypes with particularly high activity being evident in ERα-negative, HER2-positive, and triple-negative breast cancers has raised significant interest in targeting this receptor for the treatment of those breast cancers for which therapeutic options are limited.
Collapse
Affiliation(s)
- Ching-yi Chang
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | | |
Collapse
|
355
|
Liao XM, Leung KN. Indirubin-3'-oxime induces mitochondrial dysfunction and triggers growth inhibition and cell cycle arrest in human neuroblastoma cells. Oncol Rep 2012; 29:371-9. [PMID: 23117445 DOI: 10.3892/or.2012.2094] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 09/20/2012] [Indexed: 11/06/2022] Open
Abstract
Neuroblastoma is the most common extracranial solid tumor found in infancy and childhood. Current multimodal therapies such as surgery, chemotherapy, radiotherapy and stem cell transplantation often cause inevitable severe side-effects, therefore, it is necessary to develop novel drugs with higher efficacy on neuroblastoma cells and minimal side-effects on normal cells. Indirubin-3'-oxime (I3M), an indigo alkaloid, was found to exhibit potent antitumor activities on various types of cancer cells. However, its modulatory effects on human neuroblastoma and the underlying mechanisms remain poorly understood. As mitochondrial biogenesis and function play critical roles in cell growth and survival, in the present study the effects of I3M on mitochondrial functions and their correlation to the anticancer effect of I3M on human neuroblastoma cells were investigated. I3M was found to inhibit the growth of the human neuroblastoma LA-N-1, SH-SY5Y and SK-N-DZ cells in a dose- and time-dependent manner, but exhibited little, if any, direct cytotoxicity on normal cells. Mechanistic studies showed that I3M specifically decreased the expression of the mitochondrial regulators ERRγ and PGC-1β and resulted in decreased mitochondrial mass and altered mitochondrial function characterized by a reduction in mitochondrial membrane potential and elevation of reactive oxygen species levels in LA-N-1 cells. I3M also increased the level of CDK inhibitor p27Kip1 and reduced the levels of CDK2 and cyclin E in LA-N-1 cells, leading to cell cycle arrest at the G0/G1 phase. Collectively, these results suggest that mitochondrial dysfunction might be an important mechanism underlying the I3M-induced cell cycle arrest.
Collapse
Affiliation(s)
- Xue-Mei Liao
- Biochemistry Programme, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, PR China
| | | |
Collapse
|
356
|
Percharde M, Lavial F, Ng JH, Kumar V, Tomaz RA, Martin N, Yeo JC, Gil J, Prabhakar S, Ng HH, Parker MG, Azuara V. Ncoa3 functions as an essential Esrrb coactivator to sustain embryonic stem cell self-renewal and reprogramming. Genes Dev 2012; 26:2286-98. [PMID: 23019124 DOI: 10.1101/gad.195545.112] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Embryonic stem cell (ESC) pluripotency depends on a well-characterized gene regulatory network centered on Oct4, Sox2, and Nanog. In contrast, little is known about the identity of the key coregulators and the mechanisms by which they may potentiate transcription in ESCs. Alongside core transcription factors, the orphan nuclear receptor Esrrb (estrogen-related receptor β) is vital for the maintenance of ESC identity and furthermore is uniquely associated with the basal transcription machinery. Here, we show that Ncoa3, an essential coactivator, is required to mediate Esrrb function in ESCs. Ncoa3 interacts with Esrrb via its ligand-binding domain and bridges Esrrb to RNA polymerase II complexes. Functionally, Ncoa3 is critical for both the induction and maintenance of pluripotency. Through chromatin immunoprecipitation (ChIP) sequencing and microarray experiments, we further demonstrate that Ncoa3 shares overlapping gene regulatory functions with Esrrb and cooperates genome-wide with the Oct4-Sox2-Nanog circuitry at active enhancers to up-regulate genes involved in self-renewal and pluripotency. We propose an integrated model of transcriptional and coactivator control, mediated by Ncoa3, for the maintenance of ESC self-renewal and somatic cell reprogramming.
Collapse
Affiliation(s)
- Michelle Percharde
- Institute of Reproductive and Developmental Biology, Imperial College, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
357
|
Lee JH, Kim EJ, Kim DK, Lee JM, Park SB, Lee IK, Harris RA, Lee MO, Choi HS. Hypoxia induces PDK4 gene expression through induction of the orphan nuclear receptor ERRγ. PLoS One 2012; 7:e46324. [PMID: 23050013 PMCID: PMC3457976 DOI: 10.1371/journal.pone.0046324] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 08/29/2012] [Indexed: 01/06/2023] Open
Abstract
Multiple cellular signaling pathways that control metabolism and survival are activated when cell are incubated under hypoxic conditions. Activation of the hypoxia inducible factor (HIF)-1 promotes expression of genes that increase the capacity to cope with the stress imposed by a reduced oxygen environment. Here we show that the orphan nuclear receptor estrogen related receptor γ (ERRγ) plays a critical role in hypoxia–mediated activation of pyruvate dehydrogenase kinase 4 (PDK4) gene expression. ERRγ mRNA and protein levels were increased by hypoxia or desferrioxamine (DFO) treatment in hepatoma cell lines. Co-expression of HIF-1α and β increased ERRγ promoter activity as well as mRNA expression, while knockdown of endogenous HIF-1α reduced the hypoxia-mediated induction of ERRγ. In addition, hypoxia also increased the promoter activity and mRNA level of PDK4 in HepG2 cells. Adenovirus mediated-overexpression of ERRγ specifically increased PDK4 gene expression, while ablation of endogenous ERRγ significantly decreased hypoxia-mediated induction of PDK4 gene expression. Finally, GSK5182, an inverse agonist of ERRγ, strongly inhibited the hypoxia-mediated induction of PDK4 protein and promoter activity. Regulation of the transcriptional activity of ERRγ may provide a therapeutic approach for the regulation of PDK4 gene expression under hypoxia.
Collapse
Affiliation(s)
- Ja Hee Lee
- National Creative Research Initiatives Center for Nuclear Receptor Signals and Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Eun-Jin Kim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Don-Kyu Kim
- National Creative Research Initiatives Center for Nuclear Receptor Signals and Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Ji-Min Lee
- National Creative Research Initiatives Center for Nuclear Receptor Signals and Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Seung Bum Park
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
- Department of Biophysics and Chemical Biology, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - In-Kyu Lee
- World Class University Program, Research Institute of Aging and Metabolism, Kyungpook National University School of Medicine, Daegu, Republic of Korea
- Department of Endocrinology and Metabolism, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Robert A. Harris
- World Class University Program, Department of Biochemistry and Molecular Biology, Indiana University School of Medicine and the Roudebush VA Medical Center, Indianapolis, Indiannapolis, United States of America
| | - Mi-Ock Lee
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
- * E-mail: (MOL); (HSC)
| | - Hueng-Sik Choi
- National Creative Research Initiatives Center for Nuclear Receptor Signals and Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
- Research Institute of Medical Sciences, Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju, Republic of Korea
- * E-mail: (MOL); (HSC)
| |
Collapse
|
358
|
Hawk JD, Bookout AL, Poplawski SG, Bridi M, Rao AJ, Sulewski ME, Kroener BT, Manglesdorf DJ, Abel T. NR4A nuclear receptors support memory enhancement by histone deacetylase inhibitors. J Clin Invest 2012; 122:3593-602. [PMID: 22996661 DOI: 10.1172/jci64145] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 07/26/2012] [Indexed: 02/06/2023] Open
Abstract
The formation of a long-lasting memory requires a transcription-dependent consolidation period that converts a short-term memory into a long-term memory. Nuclear receptors compose a class of transcription factors that regulate diverse biological processes, and several nuclear receptors have been implicated in memory formation. Here, we examined the potential contribution of nuclear receptors to memory consolidation by measuring the expression of all 49 murine nuclear receptors after learning. We identified 13 nuclear receptors with increased expression after learning, including all 3 members of the Nr4a subfamily. These CREB-regulated Nr4a genes encode ligand-independent "orphan" nuclear receptors. We found that blocking NR4A activity in memory-supporting brain regions impaired long-term memory but did not impact short-term memory in mice. Further, expression of Nr4a genes increased following the memory-enhancing effects of histone deacetylase (HDAC) inhibitors. Blocking NR4A signaling interfered with the ability of HDAC inhibitors to enhance memory. These results demonstrate that the Nr4a gene family contributes to memory formation and is a promising target for improving cognitive function.
Collapse
Affiliation(s)
- Joshua D Hawk
- Neuroscience Graduate Group and Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
359
|
Shearer KE, Rickert EL, Peterson AC, Weatherman RV. Dissecting rapid estrogen signaling with conjugates. Steroids 2012; 77:968-73. [PMID: 22417626 PMCID: PMC3382051 DOI: 10.1016/j.steroids.2012.02.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Revised: 02/18/2012] [Accepted: 02/24/2012] [Indexed: 10/28/2022]
Abstract
Hypothesizing that rapid estrogen signaling could be modulated from different estrogen receptors with unique localization patterns, a number of groups have attempted to design drug conjugates that target or restrict compounds to specific subcellular compartments. This article will briefly discuss the history of using conjugates to dissect rapid estrogen signaling and different strategies to attempt to target estrogens and antiestrogens to different locations. It will also detail some of the potential issues that can arise with different types of conjugates, using examples drawn from the authors' own work.
Collapse
Affiliation(s)
- Kati E. Shearer
- Department of Chemistry and Biochemistry, Rose-Hulman Institute of Technology, Terre Haute, IN. USA
| | - Emily L. Rickert
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN. USA
| | - Anton C. Peterson
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN. USA
| | - Ross V. Weatherman
- Department of Chemistry and Biochemistry, Rose-Hulman Institute of Technology, Terre Haute, IN. USA
| |
Collapse
|
360
|
Gacias M, Pérez-Martí A, Pujol-Vidal M, Marrero PF, Haro D, Relat J. PGC-1β regulates mouse carnitine-acylcarnitine translocase through estrogen-related receptor α. Biochem Biophys Res Commun 2012; 423:838-43. [PMID: 22713466 DOI: 10.1016/j.bbrc.2012.06.051] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 06/12/2012] [Indexed: 11/25/2022]
Abstract
Carnitine/acylcarnitine translocase (CACT) is a mitochondrial-membrane carrier proteins that mediates the transport of acylcarnitines into the mitochondrial matrix for their oxidation by the mitochondrial fatty acid-oxidation pathway. CACT deficiency causes a variety of pathological conditions, such as hypoketotic hypoglycemia, cardiac arrest, hepatomegaly, hepatic dysfunction and muscle weakness, and it can be fatal in newborns and infants. Here we report that expression of the Cact gene is induced in mouse skeletal muscle after 24h of fasting. To gain insight into the control of Cact gene expression, we examine the transcriptional regulation of the mouse Cact gene. We show that the 5'-flanking region of this gene is transcriptionally active and contains a consensus sequence for the estrogen-related receptor (ERR), a member of the nuclear receptor family of transcription factors. This sequence binds ERRαin vivo and in vitro and is required for the activation of Cact expression by the peroxisome proliferator-activated receptor gamma coactivator (PGC)-1/ERR axis. We also demonstrate that XTC790, the inverse agonist of ERRα, specifically blocks Cact activation by PGC-1β in C2C12 cells.
Collapse
Affiliation(s)
- Mar Gacias
- Department of Biochemistry and Molecular Biology, School of Pharmacy and the Institute of Biomedicine of the University of Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
361
|
Poidatz D, Dos Santos E, Brulé A, De Mazancourt P, Dieudonné MN. Estrogen-related receptor gamma modulates energy metabolism target genes in human trophoblast. Placenta 2012; 33:688-95. [PMID: 22763271 DOI: 10.1016/j.placenta.2012.06.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 05/23/2012] [Accepted: 06/07/2012] [Indexed: 11/25/2022]
Abstract
Placenta growth and functions depend on correct trophoblast migration, proliferation, and differentiation. The placenta has a critical role in gas and nutrient transport. To accomplish these numerous functions, the placenta depends on a highly efficient energy metabolism control. Recent studies showed that the orphan nuclear receptor Estrogen-Related Receptor gamma (ERRγ) is highly expressed in human placentas. As ERRγ has been described as a major energy metabolism regulator, we investigated ERRγ expression and putative roles on energy homeostasis in human trophoblast from first trimester placentas. First, we showed that ERRγ expression level increased during pregnancy and that ERRγ was more abundant in villous than in extravillous trophoblasts. We also observed that ERRγ expression increased during trophoblast differentiation. Second, we demonstrated that mitochondrial biogenesis and expression of some energy metabolism target genes decreased when ERRγ expression was impaired. Altogether, these results suggest that ERRγ could be implicated in the energy metabolism regulation of human trophoblasts.
Collapse
Affiliation(s)
- D Poidatz
- Université de Versailles-St Quentin, Service de Biochimie et Biologie Moléculaire, UPRES-EA 2493, Faculté de Médecine Paris-Ile de France Ouest, PRES Université Paris Grand Ouest (UPGO), Centre Hospitalier de Poissy-Saint Germain, 78303 Poissy Cedex, France
| | | | | | | | | |
Collapse
|
362
|
Bianco S, Sailland J, Vanacker JM. ERRs and cancers: effects on metabolism and on proliferation and migration capacities. J Steroid Biochem Mol Biol 2012; 130:180-5. [PMID: 21414406 DOI: 10.1016/j.jsbmb.2011.03.014] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 01/20/2011] [Accepted: 03/07/2011] [Indexed: 01/06/2023]
Abstract
ERRs are orphan members of the nuclear receptor superfamily which, at least for ERRα and ERRγ display important roles in the control of various metabolic processes. On other hand, correlations have been found between the expression of ERRα and γ and diverse parameters of tumor progression in human cancers. Whereas it is tempting to speculate that ERR receptors act in tumors through the regulation of metabolism, recent data have suggested that they also may directly regulate tumor proliferation and progression independently of their effects on metabolism. The two aspects of tumoral functions of ERR receptors are the purpose of the present review.
Collapse
Affiliation(s)
- Stéphanie Bianco
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, Ecole Normale Supérieure de Lyon, 46 allée d'Italie, 69364 Lyon cedex 07, France
| | | | | |
Collapse
|
363
|
Abstract
Estrogen is a potent steroid with pleiotropic effects, which have yet to be fully elucidated. Estrogen has both nuclear and non-nuclear effects. The rapid response to estrogen, which involves a membrane associated estrogen receptor(ER) and is protective, involves signaling through PI3K, Akt, and ERK 1/2. The nuclear response is much slower, as the ER-estrogen complex moves to the nucleus, where it functions as a transcription factor, both activating and repressing gene expression. Several different ERs regulate the specificity of response to estrogen, and appear to have specific effects in cardiac remodeling and the response to injury. However, much remains to be understood about the selectivity of these receptors and their specific effects on gene expression. Basic studies have demonstrated that estrogen treatment prevents apoptosis and necrosis of cardiac and endothelial cells. Estrogen also attenuates pathologic cardiac hypertrophy. Estrogen may have great benefit in aging as an anti-inflammatory agent. However, clinical investigations of estrogen have had mixed results, and not shown the clear-cut benefit of more basic investigations. This can be explained in part by differences in study design: in basic studies estrogen treatment was used immediately or shortly after ovariectomy, while in some key clinical trials, estrogen was given years after menopause. Further basic research into the underlying molecular mechanisms of estrogen's actions is essential to provide a better comprehension of the many properties of this powerful hormone.
Collapse
Affiliation(s)
- A A Knowlton
- Molecular and Cellular Cardiology, Department of Medicine, University of California, Davis, CA 95616, USA.
| | | |
Collapse
|
364
|
Lee VHY, Lam IPY, Choi HS, Chow BKC, Lee LTO. The estrogen-related receptor alpha upregulates secretin expressions in response to hypertonicity and angiotensin II stimulation. PLoS One 2012; 7:e39913. [PMID: 22761926 PMCID: PMC3382582 DOI: 10.1371/journal.pone.0039913] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 06/03/2012] [Indexed: 01/17/2023] Open
Abstract
Osmoregulation via maintenance of water and salt homeostasis is a vital process. In the brain, a functional secretin (SCT) and secretin receptor (SCTR) axis has recently been shown to mediate central actions of angiotensin II (ANGII), including initiation of water intake and stimulation of vasopressin (VP) expression and release. In this report, we provide evidence that estrogen-related receptor α (ERRα, NR3B1), a transcription factor mainly involved in metabolism, acts as an upstream activator of the SCT gene. In vitro studies using mouse hypothalamic cell line N-42 show that ERRα upregulates SCT promoter and gene expression. More importantly, knockdown of endogenous ERRα abolishes SCT promoter activation in response to hypertonic and ANGII stimulations. In mouse brain, ERRα coexpresses with SCT in various osmoregulatory brain regions, including the lamina terminalis and the paraventricular nucleus of the hypothalamus, and its expression is induced by hyperosmotic and ANGII treatments. Based on our data, we propose that both the upregulation of ERRα and/or the increased binding of ERRα to the mouse SCT promoter are two possible mechanisms for the elevated SCT expression upon hyperosmolality and central ANGII stimulation.
Collapse
Affiliation(s)
- Vien H. Y. Lee
- School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Ian P. Y. Lam
- School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Hueng-Sik Choi
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Billy K. C. Chow
- School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Leo T. O. Lee
- School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- * E-mail:
| |
Collapse
|
365
|
Crawford PA, Schaffer JE. Metabolic stress in the myocardium: adaptations of gene expression. J Mol Cell Cardiol 2012; 55:130-8. [PMID: 22728216 DOI: 10.1016/j.yjmcc.2012.06.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 06/05/2012] [Accepted: 06/13/2012] [Indexed: 12/28/2022]
Abstract
The heart is one of the highest ATP consuming organs in mammalian organisms. Its metabolic function has evolved a remarkable degree of efficiency to meet high demand and plasticity in response to varying changes in energy substrate supply. Given the high flux of energy substrates and the centrality of their appropriate use for optimal cardiac function, it is not surprising that the heart has intricate signaling mechanisms through which it responds to metabolic stress. This review focuses on the changes in gene expression in myocardial and vascular tissues during metabolic stress that affect mRNAs and subsequent protein synthesis with an eye toward understanding the manner in which these changes effect adaptive and maladaptive responses of the heart. This article is part of a Special Issue entitled "Focus on Cardiac Metabolism".
Collapse
Affiliation(s)
- Peter A Crawford
- Diabetic Cardiovascular Disease Center, Cardiovascular Division, Washington University School of Medicine, USA.
| | | |
Collapse
|
366
|
Perry DJ, Yin Y, Telarico T, Baker HV, Dozmorov I, Perl A, Morel L. Murine lupus susceptibility locus Sle1c2 mediates CD4+ T cell activation and maps to estrogen-related receptor γ. THE JOURNAL OF IMMUNOLOGY 2012; 189:793-803. [PMID: 22711888 DOI: 10.4049/jimmunol.1200411] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Sle1c is a sublocus of the NZM2410-derived Sle1 major lupus susceptibility locus. We have shown previously that Sle1c contributes to lupus pathogenesis by conferring increased CD4(+) T cell activation and increased susceptibility to chronic graft-versus-host disease (cGVHD), which mapped to the centromeric portion of the locus. In this study, we have refined the centromeric sublocus to a 675-kb interval, termed Sle1c2. Mice from recombinant congenic strains expressing Sle1c2 exhibited increased CD4(+) T cell intrinsic activation and cGVHD susceptibility, similar to mice with the parental Sle1c. In addition, B6.Sle1c2 mice displayed a robust expansion of IFN-γ-expressing T cells. NZB complementation studies showed that Sle1c2 expression exacerbated B cell activation, autoantibody production, and renal pathology, verifying that Sle1c2 contributes to lupus pathogenesis. The Sle1c2 interval contains two genes, only one of which, Esrrg, is expressed in T cells. B6.Sle1c2 CD4(+) T cells expressed less Esrrg than B6 CD4(+) T cells, and Esrrg expression was correlated negatively with CD4(+) T cell activation. Esrrg encodes an orphan nuclear receptor that regulates oxidative metabolism and mitochondrial functions. In accordance with reduced Esrrg expression, B6.Sle1c2 CD4(+) T cells present reduced mitochondrial mass and altered mitochondrial functions as well as altered metabolic pathway utilization when compared with B6 CD4(+) T cells. Taken together, we propose Esrrg as a novel lupus susceptibility gene regulating CD4(+) T cell function through their mitochondrial metabolism.
Collapse
Affiliation(s)
- Daniel J Perry
- Department of Pathology, University of Florida, Gainesville, FL, USA
| | | | | | | | | | | | | |
Collapse
|
367
|
Zhao Y, Li Y, Lou G, Zhao L, Xu Z, Zhang Y, He F. MiR-137 targets estrogen-related receptor alpha and impairs the proliferative and migratory capacity of breast cancer cells. PLoS One 2012; 7:e39102. [PMID: 22723937 PMCID: PMC3377602 DOI: 10.1371/journal.pone.0039102] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Accepted: 05/16/2012] [Indexed: 12/05/2022] Open
Abstract
ERRα is an orphan nuclear receptor emerging as a novel biomarker of breast cancer. Over-expression of ERRα in breast tumor is considered as a prognostic factor of poor clinical outcome. The mechanisms underlying the dysexpression of this nuclear receptor, however, are poorly understood. MicroRNAs (miRNAs) regulate gene expression at the post-transcriptional level and play important roles in tumor initiation and progression. In the present study, we have identified that the expression of ERRα is regulated by miR-137, a potential tumor suppressor microRNA. The bioinformatics search revealed two putative and highly conserved target-sites for miR-137 located within the ERRα 3'UTR at nt 480-486 and nt 596-602 respectively. Luciferase-reporter assay demonstrated that the two predicted target sites were authentically functional. They mediated the repression of reporter gene expression induced by miR-137 in an additive manner. Moreover, ectopic expression of miR-137 down-regulated ERRα expression at both protein level and mRNA level, and the miR-137 induced ERRα-knockdown contributed to the impaired proliferative and migratory capacity of breast cancer cells. Furthermore, transfection with miR-137 mimics suppressed at least two downstream target genes of ERRα-CCNE1 and WNT11, which are important effectors of ERRα implicated in tumor proliferation and migration. Taken together, our results establish a role of miR-137 in negatively regulating ERRα expression and breast cancer cell proliferation and migration. They suggest that manipulating the expression level of ERRα by microRNAs has the potential to influence breast cancer progression.
Collapse
Affiliation(s)
- Yuanyin Zhao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Yuping Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Guiyu Lou
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Li Zhao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Zhizhen Xu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Yan Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Fengtian He
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| |
Collapse
|
368
|
Abstract
Cancer cells typically display altered glucose metabolism characterized by a preference of aerobic glycolysis, known as the Warburg effect, which facilitates cell proliferation. Hypoxia-inducible factor (HIF) and oncoprotein Myc are two prominent transcription factors that drive glycolysis. Previously, we reported that the estrogen-related receptors (ERRs) act as cofactors of HIF and enhance HIF-dependent transcription of glycolytic genes under hypoxia. ERRs are orphan nuclear receptors and key regulators of energy metabolism by orchestrating mitochondrial biogenesis, fatty acid oxidation (FAO) and oxidative phosphorylation. Here, we show that ERRs also stimulate glycolysis under normoxia. ERRs directly bind to and activate promoters of many genes encoding glycolytic enzymes, and the ERR-binding sites in such promoters are essential for ERR-mediated transcriptional activation. ERRs interact with Myc, and the two factors synergistically activate transcription of glycolytic genes. Furthermore, overexpression of ERRs increases glycolytic gene expression and lactate production. Conversely, depletion of ERRs in cancer cells reduces expression of glycolytic genes and glucose uptake, resulting in decreased aerobic glycolysis and cell growth. Taken together, these results suggest that ERRs are important transcriptional activators of the glycolytic pathway and contribute to the Warburg effect in cancer cells.
Collapse
|
369
|
Reactive oxygen species-mediated control of mitochondrial biogenesis. Int J Cell Biol 2012; 2012:403870. [PMID: 22693510 PMCID: PMC3369472 DOI: 10.1155/2012/403870] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 03/29/2012] [Accepted: 03/31/2012] [Indexed: 11/17/2022] Open
Abstract
Mitochondrial biogenesis is a complex process. It necessitates the contribution of both the nuclear and the mitochondrial genomes and therefore crosstalk between the nucleus and mitochondria. It is now well established that cellular mitochondrial content can vary according to a number of stimuli and physiological states in eukaryotes. The knowledge of the actors and signals regulating the mitochondrial biogenesis is thus of high importance. The cellular redox state has been considered for a long time as a key element in the regulation of various processes. In this paper, we report the involvement of the oxidative stress in the regulation of some actors of mitochondrial biogenesis.
Collapse
|
370
|
Burgueño AL, Cabrerizo R, Gonzales Mansilla N, Sookoian S, Pirola CJ. Maternal high-fat intake during pregnancy programs metabolic-syndrome-related phenotypes through liver mitochondrial DNA copy number and transcriptional activity of liver PPARGC1A. J Nutr Biochem 2012; 24:6-13. [PMID: 22658649 DOI: 10.1016/j.jnutbio.2011.12.008] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 11/19/2011] [Accepted: 12/23/2011] [Indexed: 01/04/2023]
Abstract
UNLABELLED In this study, we contrasted the hypothesis that maternal diet during pregnancy has an impact on fetal metabolic programming through changes in liver mitochondrial DNA (mtDNA) content and transcriptional activity of Ppargc1a and that these effects are sex specific. METHODS Rats were fed either high-fat (HFD) or standard chow diet (SCD) during gestation and lactation. The resulting adult male and female offspring were fed either HFD or SCD for an 18-week period, generating eight experimental groups. RESULTS Maternal HFD feeding during pregnancy is associated with a decreased liver mtDNA copy number (P<.008). This effect was independent of the offspring sex or diet, and was significantly associated with fatty liver when dams were fed HFD (P<.05, adjusted by homeostasis model assessment of insulin resistance, HOMA-IR). We also found that maternal HFD feeding results in a male-specific significant reduction of the liver abundance of Ppargc1a mRNA (P<.004), which significantly impacted peripheral insulin resistance. Liver expression of Ppargc1a was inversely correlated with HOMA-IR (R=-0.53, P<.0003). Only male offspring exposed to a chronic metabolic insult in adult life were insulin resistant and hyperleptinemic, and showed abnormal liver and abdominal fat accumulation. Liver abundance of Tfam, Nrf1, Hnf4a, Pepck and Ppparg mRNA was not associated with maternal programming. In conclusion, maternal high-fat diet feeding during pregnancy programs liver mtDNA content and the transcriptional activity of Ppargc1a, which strongly modulates, in a sex-specific manner, glucose homeostasis and organ fat accumulation in adult life after exposure to a nutritional insult.
Collapse
Affiliation(s)
- Adriana Laura Burgueño
- Department of Molecular Genetics and Biology of Complex Diseases, Institute of Medical Research A. Lanari-IDIM, University of Buenos Aires-National-Council of Scientific and Technological Research (CONICET), Ciudad Autónoma de Buenos Aires-1427, Argentina
| | | | | | | | | |
Collapse
|
371
|
Zengin A, Nguyen AD, Wong IPL, Zhang L, Enriquez RF, Eisman JA, Herzog H, Baldock PA, Sainsbury A. Neuropeptide Y mediates the short-term hypometabolic effect of estrogen deficiency in mice. Int J Obes (Lond) 2012; 37:390-8. [PMID: 22565420 DOI: 10.1038/ijo.2012.71] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Estrogen deficiency increases body weight or total and central adiposity and decreases energy expenditure. Hypothalamic neuropeptide Y (NPY) expression is altered by estrogen deficiency in rodents, but the long-term consequences on energy homeostasis are unknown. OBJECTIVE To investigate the role of NPY in the changes in energy expenditure and physical activity, as well as the associated changes in body weight and composition in response to short-term and long-term estrogen deficiency. DESIGN Sham and ovariectomy (OVX) operations were performed at 8 weeks of age in wild-type (WT) and NPY(-/-) mice. Energy expenditure, physical activity, body composition and weight, as well as food intake were measured at 10-18 days (short-term) and 46-54 days (long-term) after OVX. RESULTS OVX influences energy homeostasis differently at early compared with later time-points. At the early but not the late time point, OVX in WT mice reduced oxygen consumption and energy expenditure and tended to reduce resting metabolic rate. Interestingly, these effects of short-term estrogen deficiency were ablated by NPY deletion, with NPY(-/-) mice exhibiting significant increases in energy expenditure and resting metabolic rate. In addition to these hypermetabolic effects, OVX NPY(-/-) mice exhibited significantly lower body weight and whole-body fat mass relative to OVX WT controls at the short-term but not the long-term time point. Food intake and physical activity were unaltered by OVX, but NPY(-/-) mice exhibited significant reductions in these parameters relative to WT. CONCLUSION The effects of estrogen deficiency to reduce energy metabolism are transient, and NPY is critical to this effect as well as the early OVX-induced obesity.
Collapse
Affiliation(s)
- A Zengin
- Osteoporosis and Bone Biology, Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, New South Wales, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
372
|
Kim DK, Ryu D, Koh M, Lee MW, Lim D, Kim MJ, Kim YH, Cho WJ, Lee CH, Park SB, Koo SH, Choi HS. Orphan nuclear receptor estrogen-related receptor γ (ERRγ) is key regulator of hepatic gluconeogenesis. J Biol Chem 2012; 287:21628-39. [PMID: 22549789 DOI: 10.1074/jbc.m111.315168] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Glucose homeostasis is tightly controlled by hormonal regulation of hepatic glucose production. Dysregulation of this system is often associated with insulin resistance and diabetes, resulting in hyperglycemia in mammals. Here, we show that the orphan nuclear receptor estrogen-related receptor γ (ERRγ) is a novel downstream mediator of glucagon action in hepatic gluconeogenesis and demonstrate a beneficial impact of the inverse agonist GSK5182. Hepatic ERRγ expression was increased by fasting-dependent activation of the cAMP-response element-binding protein-CRTC2 pathway. Overexpression of ERRγ induced Pck1 and G6PC gene expression and glucose production in primary hepatocytes, whereas abolition of ERRγ gene expression attenuated forskolin-mediated induction of gluconeogenic gene expression. Deletion and mutation analyses of the Pck1 promoter showed that ERRγ directly regulates the Pck1 gene transcription via ERR response elements of the Pck1 promoter as confirmed by ChIP assay and in vivo imaging analysis. We also demonstrate that GSK5182, an inverse agonist of ERRγ, specifically inhibits the transcriptional activity of ERRγ in a PGC-1α dependent manner. Finally, the ERRγ inverse agonist ameliorated hyperglycemia through inhibition of hepatic gluconeogenesis in db/db mice. Control of hepatic glucose production by an ERRγ-specific inverse agonist is a new potential therapeutic approach for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Don-Kyu Kim
- National Creative Research Initiatives Center for Nuclear Receptor Signals, School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
373
|
Meermeier N, Krishnan N. Circadian regulation of cellular homeostasis--implications for cell metabolism and clinical diseases. Med Hypotheses 2012; 79:17-24. [PMID: 22521428 DOI: 10.1016/j.mehy.2012.03.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 03/25/2012] [Indexed: 01/07/2023]
Abstract
The major pathways involving nutrient and energy metabolism including cellular homeostasis are profoundly impacted by the circadian clock, which orchestrates diurnal rhythms in physiology and behavior. While the links between circadian and metabolic rhythms are unclear, recent studies imply a close link between the two with one feeding back on the other. In this discussion, we present the hypothesis that circadian clocks likely contribute to cellular homeostasis, especially proteostasis, through regulation of metabolic rhythms, which in turn feed-back on circadian oscillators. The disruption of circadian clocks leads to altered metabolic rhythms and metabolic disease states as a result of altered cellular homeostasis.
Collapse
Affiliation(s)
- N Meermeier
- Department of Microbiology, 220 Nash Hall, Oregon State University, Corvallis, OR 97331, United States.
| | | |
Collapse
|
374
|
Abstract
Cancer cells exhibit altered glucose metabolism characterized by a preference for aerobic glycolysis or the Warburg effect, and the cells resist matrix detachment-induced apoptosis, which is called anoikis, a barrier to metastasis. It remains largely unclear whether tumor metabolism influences anoikis and metastasis. Here we show that when detached from the matrix, untransformed mammary epithelial cells undergo metabolic reprogramming by markedly upregulating pyruvate dehydrogenase (PDH) kinase 4 (PDK4) through estrogen-related receptor gamma (ERRγ), thereby inhibiting PDH and attenuating the flux of glycolytic carbon into mitochondrial oxidation. To decipher the significance of this metabolic response, we found that depletion of PDK4 or activation of PDH increased mitochondrial respiration and oxidative stress in suspended cells, resulting in heightened anoikis. Conversely, overexpression of PDKs prolonged survival of cells in suspension. Therefore, decreased glucose oxidation following cell detachment confers anoikis resistance. Unlike untransformed cells, most cancer cells demonstrate reduced glucose oxidation even under attached conditions, and thus they inherently possess a survival advantage when suspended. Normalization of glucose metabolism by stimulating PDH in cancer cells restores their susceptibility to anoikis and impairs their metastatic potential. These results suggest that the Warburg effect, more specifically, diminished glucose oxidation, promotes anoikis resistance and metastasis and that PDKs are potential targets for antimetastasis therapy.
Collapse
|
375
|
Matsakas A, Yadav V, Lorca S, Evans RM, Narkar VA. Revascularization of ischemic skeletal muscle by estrogen-related receptor-γ. Circ Res 2012; 110:1087-96. [PMID: 22415017 DOI: 10.1161/circresaha.112.266478] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
RATIONALE Oxidative myofibers in the skeletal muscles express high levels of angiogenic factors, have dense vasculature, and promptly revascularize during ischemia. Estrogen-related receptor-gamma (ERRγ) activates genes that govern metabolic and vascular features typical to oxidative myofibers. Therefore, ERRγ-dependent remodeling of the myofibers may promote neoangiogenesis and restoration of blood perfusion in skeletal muscle ischemia. OBJECTIVE To investigate the muscle fiber type remodeling by ERRγ and its role in the vascular recovery of ischemic muscle. METHODS AND RESULTS Using immunohistology, we show that skeletal muscle-specific transgenic overexpression of ERRγ increases the proportions of oxidative and densely vascularized type IIA and IIX myofibers and decreases glycolytic and less vascularized type IIB myofibers. This myofiber remodeling results in a higher basal blood flow in the transgenic skeletal muscle. By applying unilateral hind limb ischemia to transgenic and wild-type mice, we found accelerated revascularization (fluorescent microangiography), restoration of blood perfusion (laser Doppler flowmetry), and muscle repair (Evans blue dye exclusion) in transgenic compared to wild-type ischemic muscles. This ameliorative effect is linked to enhanced neoangiogenesis (CD31 staining and microfil perfusion) by ERRγ. Using cultured muscle cells in which ERRγ is inactivated, we show that the receptor is dispensable for the classical hypoxic response of transcriptional upregulation and secretion of vascular endothelial growth factor A. Rather, the ameliorative effect of ERRγ is linked to the receptor-mediated increase in oxidative myofibers that inherently express and secrete high levels of angiogenic factors. CONCLUSIONS The ERRγ is a hypoxia-independent inducer of neoangiogenesis that can promote reparative revascularization.
Collapse
Affiliation(s)
- Antonios Matsakas
- Center for Metabolic and Degenerative Diseases, Brown Foundation Institute of Molecular Medicine, The University of Texas Medical School at Houston, 77030, USA
| | | | | | | | | |
Collapse
|
376
|
Ju D, He J, Zhao L, Zheng X, Yang G. Estrogen related receptor α-induced adipogenesis is PGC-1β-dependent. Mol Biol Rep 2012; 39:3343-54. [PMID: 21732060 DOI: 10.1007/s11033-011-1104-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2011] [Accepted: 06/15/2011] [Indexed: 11/25/2022]
Abstract
Previous report showed that Estrogen related receptor α (ERRα) knockout mice had a significant reduction in adipose tissue deposition. Although it was reported that ERRα could promote adipogenesis in several immortalized preadipocytes cell lines, the mechanism behind which is still unclear to date. Besides, the expression pattern of ERRα in white adipose tissue is rarely examined. Here, we show that the expression of ERRα in primary cultured adipocytes is closely associated with adipogenesis. Besides, we found that peroxisome proliferator-activated receptor-γ coactivator 1β (PGC-1β) play an important role in regulating ERRα-induced adipogenesis. ERRα-induced adipogenesis was greatly attenuated when knocking down PGC-1β expression, while rescued by overexpression of PGC-1β. However, PGC-1β could still promote adipogenesis when suppressing ERRα expression. Furthermore, we demonstrated that ERRα could transcriptionally active PGC-1β expression and then enhance the formation of sterol-regulatory-element-binding protein-1c (SREBP-1c)/PGC-1β complex and peroxisome proliferator-activated receptor-γ (PPARγ)/PGC-1β complex. Taken together, these results indicate that ERRα-induced adipogenesis is triggered by modulating the expression of PGC-1β.
Collapse
Affiliation(s)
- Dapeng Ju
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, People's Republic of China
| | | | | | | | | |
Collapse
|
377
|
Kawashima H, Nakatani T. Involvement of estrogen receptors in prostatic diseases. Int J Urol 2012; 19:512-22; author reply 522-3. [PMID: 22375605 DOI: 10.1111/j.1442-2042.2012.02987.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Accumulating evidence shows that estrogens participate in the pathogenesis and development of benign prostatic hyperplasia and prostate cancer by activating estrogen receptor α. In contrast, estrogen receptor β is involved in the differentiation and maturation of prostatic epithelial cells, and thus possesses antitumor effects in prostate cancer. However, the natural ligands of estrogen receptor β are not fully understood, and its mode of action according to its ligands and the binding sites located in the promoter regions of downstream genes remains to be elucidated. Here, we review recent experimental investigations of estrogen receptors and their urological relevance. Estrogen receptor-mediated signaling in the prostate is essential together with the androgen receptor-mediated pathway, providing a new therapeutic target for prostatic diseases.
Collapse
Affiliation(s)
- Hidenori Kawashima
- Department of Urology, Osaka City University Graduate School of Medicine, Osaka, Japan.
| | | |
Collapse
|
378
|
Um SJ, Youn H, Kim EJ. Negative regulation of ERRα by a novel nucleolar protein. Biochem Biophys Res Commun 2012; 418:290-5. [PMID: 22266318 DOI: 10.1016/j.bbrc.2012.01.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Accepted: 01/05/2012] [Indexed: 01/13/2023]
Abstract
The regulation of estrogen-related receptor (ERR) transcriptional activity is poorly understood. To explore the underlying mechanism, we sought to isolate ERRα-binding protein(s). In a yeast two-hybrid screen, we identified a novel protein that has been characterized as a retinoic acid resistance factor (RaRF) (manuscript in-preparation). A specific interaction between RaRF and ERRα was confirmed in a GST pull-down assay in vitro and immunoprecipitation (IP) in mammalian cells. Further yeast two-hybrid assays and IP analyses indicated that the C-terminus of ERRα is required for RaRF binding. Consistent with our interaction data, transfection of RaRF significantly reduced the ability of ERRα, but not ERRγ, to transactivate an ERR-responsive luciferase reporter. In contrast, down-regulation of RaRF using shRNA increased ERRα activity without affecting that of ERRγ. RaRF was subsequently shown to repress the expression of the ERR target gene pS2. Further fluorescence microscopy revealed that ERRα or ERRγ is normally expressed in the nucleoplasm, with ERRα, but not ERRγ, translocating to the nucleolus when RaRF is expressed. Taken together, our data suggest that RaRF sequesters ERRα in the nucleolus through a specific interaction, thereby inhibiting its transcriptional activity.
Collapse
Affiliation(s)
- Soo-Jong Um
- Department of Bioscience & Biotechnology/Institute of Bioscience, BK21 Graduate Program, Sejong University, Seoul 143-747, Republic of Korea
| | | | | |
Collapse
|
379
|
Gibson DA, Saunders PTK. Estrogen dependent signaling in reproductive tissues - a role for estrogen receptors and estrogen related receptors. Mol Cell Endocrinol 2012; 348:361-72. [PMID: 21964318 DOI: 10.1016/j.mce.2011.09.026] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Revised: 09/03/2011] [Accepted: 09/12/2011] [Indexed: 12/20/2022]
Abstract
Estrogens play a fundamental role in the development and normal physiological function of multiple tissue systems and have been implicated in the ontogeny of cancers. The biological effects of estrogens are classically mediated via interaction with cognate nuclear receptors. The relative expression of ER subtypes/variants varies between cells within different tissues and this alters the response to natural and synthetic ligands. This review focuses on the role of estrogen and estrogen related receptors in reproductive tissues.
Collapse
Affiliation(s)
- Douglas A Gibson
- MRC/UoE Centre for Reproductive Health, The University of Edinburgh, The Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | | |
Collapse
|
380
|
Ranhotra HS. The estrogen-related receptors: orphans orchestrating myriad functions. J Recept Signal Transduct Res 2012; 32:47-56. [PMID: 22268851 DOI: 10.3109/10799893.2011.647350] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Coordinated and tight regulation of gene expression in metazoans is essential for cellular homeostasis and functions. Tissue- and cell-specific regulatory factors are indispensable and a wide variety of them exist to regulate genes. A family of transcriptional factors was identified in the past two decades through gene cloning studies and was informally referred as "orphan receptors", as appropriate endogenous ligands for such receptors were unknown. One of the subclasses of such receptors is known as the estrogen-related receptors (ERRs), which include three isoforms, namely ERRα, ERRβ and ERRγ. Over the past one decade, unprecedented knowledge about the ERRs biology has been generated, indicating their vital roles in various metabolic and physiological activities in animals. The ERRs cellular action is largely attributed to its interaction with a wide variety of other nuclear receptors, including some orphan nuclear receptors, and thereby can modulate diverse array of genes involved in metabolism and animal physiology. Studies using genome-wide location analyses, microarray and functional genomics, including ERR-specific null mice have revealed a number of pathways controlled by the ERRs. In this context, new and recent information on the biological functions of ERRs are being reviewed.
Collapse
Affiliation(s)
- Harmit S Ranhotra
- Department of Biochemistry, Orphan Nuclear Receptors Laboratory, St. Edmund's College, Shillong, India.
| |
Collapse
|
381
|
NCoR1 is a conserved physiological modulator of muscle mass and oxidative function. Cell 2012; 147:827-39. [PMID: 22078881 DOI: 10.1016/j.cell.2011.10.017] [Citation(s) in RCA: 208] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 08/09/2011] [Accepted: 10/06/2011] [Indexed: 02/08/2023]
Abstract
Transcriptional coregulators control the activity of many transcription factors and are thought to have wide-ranging effects on gene expression patterns. We show here that muscle-specific loss of nuclear receptor corepressor 1 (NCoR1) in mice leads to enhanced exercise endurance due to an increase of both muscle mass and of mitochondrial number and activity. The activation of selected transcription factors that control muscle function, such as MEF2, PPARβ/δ, and ERRs, underpins these phenotypic alterations. NCoR1 levels are decreased in conditions that require fat oxidation, resetting transcriptional programs to boost oxidative metabolism. Knockdown of gei-8, the sole C. elegans NCoR homolog, also robustly increased muscle mitochondria and respiration, suggesting conservation of NCoR1 function. Collectively, our data suggest that NCoR1 plays an adaptive role in muscle physiology and that interference with NCoR1 action could be used to improve muscle function.
Collapse
|
382
|
|
383
|
Devarakonda S, Gupta K, Chalmers MJ, Hunt JF, Griffin PR, Van Duyne GD, Spiegelman BM. Disorder-to-order transition underlies the structural basis for the assembly of a transcriptionally active PGC-1α/ERRγ complex. Proc Natl Acad Sci U S A 2011; 108:18678-83. [PMID: 22049338 PMCID: PMC3219099 DOI: 10.1073/pnas.1113813108] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Peroxisome proliferator activated receptor (PPAR) γ coactivator-1α (PGC-1α) is a potent transcriptional coactivator of oxidative metabolism and is induced in response to a variety of environmental cues. It regulates a broad array of target genes by coactivating a whole host of transcription factors. The estrogen-related receptor (ERR) family of nuclear receptors are key PGC-1α partners in the regulation of mitochondrial and tissue-specific oxidative metabolic pathways; these receptors also demonstrate strong physical and functional interactions with this coactivator. Here we perform comprehensive biochemical, biophysical, and structural analyses of the complex formed between PGC-1α and ERRγ. PGC-1α activation domain (PGC-1α(2-220)) is intrinsically disordered with limited secondary and no defined tertiary structure. Complex formation with ERRγ induces significant changes in the conformational mobility of both partners, highlighted by significant stabilization of the ligand binding domain (ERRγLBD) as determined by HDX (hydrogen/deuterium exchange) and an observed disorder-to-order transition in PGC-1α(2-220). Small-angle X-ray scattering studies allow for modeling of the solution structure of the activation domain in the absence and presence of ERRγLBD, revealing a stable and compact binary complex. These data show that PGC-1α(2-220) undergoes a large-scale conformational change when binding to the ERRγLBD, leading to substantial compaction of the activation domain. This change results in stable positioning of the N-terminal part of the activation domain of PGC-1α, favorable for assembly of an active transcriptional complex. These data also provide structural insight into the versatile coactivation profile of PGC-1α and can readily be extended to understand other transcriptional coregulators.
Collapse
Affiliation(s)
- Srikripa Devarakonda
- Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| | - Kushol Gupta
- Department of Biochemistry and Biophysics and Howard Hughes Medical Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Michael J. Chalmers
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, FL 33458; and
| | - John F. Hunt
- Department of Biological Sciences and Northeast Structural Genomics Consortium, Columbia University, New York, NY 10027
| | - Patrick R. Griffin
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, FL 33458; and
| | - Gregory D. Van Duyne
- Department of Biochemistry and Biophysics and Howard Hughes Medical Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Bruce M. Spiegelman
- Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
384
|
Baron D, Magot A, Ramstein G, Steenman M, Fayet G, Chevalier C, Jourdon P, Houlgatte R, Savagner F, Pereon Y. Immune response and mitochondrial metabolism are commonly deregulated in DMD and aging skeletal muscle. PLoS One 2011; 6:e26952. [PMID: 22096509 PMCID: PMC3212519 DOI: 10.1371/journal.pone.0026952] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 10/06/2011] [Indexed: 01/12/2023] Open
Abstract
Duchenne Muscular Dystrophy (DMD) is a complex process involving multiple pathways downstream of the primary genetic insult leading to fatal muscle degeneration. Aging muscle is a multifactorial neuromuscular process characterized by impaired muscle regeneration leading to progressive atrophy. We hypothesized that these chronic atrophying situations may share specific myogenic adaptative responses at transcriptional level according to tissue remodeling. Muscle biopsies from four young DMD and four AGED subjects were referred to a group of seven muscle biopsies from young subjects without any neuromuscular disorder and explored through a dedicated expression microarray. We identified 528 differentially expressed genes (out of 2,745 analyzed), of which 328 could be validated by an exhaustive meta-analysis of public microarray datasets referring to DMD and Aging in skeletal muscle. Among the 328 validated co-expressed genes, 50% had the same expression profile in both groups and corresponded to immune/fibrosis responses and mitochondrial metabolism. Generalizing these observed meta-signatures with large compendia of public datasets reinforced our results as they could be also identified in other pathological processes and in diverse physiological conditions. Focusing on the common gene signatures in these two atrophying conditions, we observed enrichment in motifs for candidate transcription factors that may coordinate either the immune/fibrosis responses (ETS1, IRF1, NF1) or the mitochondrial metabolism (ESRRA). Deregulation in their expression could be responsible, at least in part, for the same transcriptome changes initiating the chronic muscle atrophy. This study suggests that distinct pathophysiological processes may share common gene responses and pathways related to specific transcription factors.
Collapse
|
385
|
Michalek RD, Gerriets VA, Nichols AG, Inoue M, Kazmin D, Chang CY, Dwyer MA, Nelson ER, Pollizzi KN, Ilkayeva O, Giguere V, Zuercher WJ, Powell JD, Shinohara ML, McDonnell DP, Rathmell JC. Estrogen-related receptor-α is a metabolic regulator of effector T-cell activation and differentiation. Proc Natl Acad Sci U S A 2011; 108:18348-53. [PMID: 22042850 PMCID: PMC3215012 DOI: 10.1073/pnas.1108856108] [Citation(s) in RCA: 172] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Stimulation of resting CD4(+) T lymphocytes leads to rapid proliferation and differentiation into effector (Teff) or inducible regulatory (Treg) subsets with specific functions to promote or suppress immunity. Importantly, Teff and Treg use distinct metabolic programs to support subset specification, survival, and function. Here, we describe that the orphan nuclear receptor estrogen-related receptor-α (ERRα) regulates metabolic pathways critical for Teff. Resting CD4(+) T cells expressed low levels of ERRα protein that increased on activation. ERRα deficiency reduced activated T-cell numbers in vivo and cytokine production in vitro but did not seem to modulate immunity through inhibition of activating signals or viability. Rather, ERRα broadly affected metabolic gene expression and glucose metabolism essential for Teff. In particular, up-regulation of Glut1 protein, glucose uptake, and mitochondrial processes were suppressed in activated ERRα(-/-) T cells and T cells treated with two chemically independent ERRα inhibitors or by shRNAi. Acute ERRα inhibition also blocked T-cell growth and proliferation. This defect appeared as a result of inadequate glucose metabolism, because provision of lipids, but not increased glucose uptake or pyruvate, rescued ATP levels and cell division. Additionally, we have shown that Treg requires lipid oxidation, whereas Teff uses glucose metabolism, and lipid addition selectively restored Treg--but not Teff--generation after acute ERRα inhibition. Furthermore, in vivo inhibition of ERRα reduced T-cell proliferation and Teff generation in both immunization and experimental autoimmune encephalomyelitis models. Thus, ERRα is a selective transcriptional regulator of Teff metabolism that may provide a metabolic means to modulate immunity.
Collapse
Affiliation(s)
- Ryan D. Michalek
- Department of Pharmacology and Cancer Biology
- Department of Immunology, and
| | | | | | | | | | | | | | | | - Kristen N. Pollizzi
- The Sidney–Kimmel Comprehensive Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Olga Ilkayeva
- The Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27710
| | - Vincent Giguere
- Goodman Cancer Research Centre and Department of Biochemistry, McGill University, Montreal, QC, Canada H3A 1A3; and
| | - William J. Zuercher
- Discovery Medicinal Chemistry, GlaxoSmithKline, Research Triangle Park, NC 27709
| | - Jonathan D. Powell
- The Sidney–Kimmel Comprehensive Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | | | | | - Jeffrey C. Rathmell
- Department of Pharmacology and Cancer Biology
- Department of Immunology, and
- The Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
386
|
Teng CT, Li Y, Stockton P, Foley J. Fasting induces the expression of PGC-1α and ERR isoforms in the outer stripe of the outer medulla (OSOM) of the mouse kidney. PLoS One 2011; 6:e26961. [PMID: 22073226 PMCID: PMC3208565 DOI: 10.1371/journal.pone.0026961] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Accepted: 10/07/2011] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Peroxisome proliferator-activated receptor-γ co-activator-1α (PGC-1α) is a member of the transcriptional coactivator family that plays a central role in the regulation of cellular energy metabolism under various physiological stimuli. During fasting, PGC-1α is induced in the liver and together with estrogen-related receptor a and γ (ERRα and ERRγ, orphan nuclear receptors with no known endogenous ligand, regulate sets of genes that participate in the energy balance program. We found that PGC-1α, ERRα and ERRγ was highly expressed in human kidney HK2 cells and that PGC-1α induced dynamic protein interactions on the ERRα chromatin. However, the effect of fasting on the expression of endogenous PGC-1α, ERRα and ERRγ in the kidney is not known. METHODOLOGY/PRINCIPAL FINDINGS In this study, we demonstrated by qPCR that the expression of PGC-1α, ERRα and ERRγ was increased in the mouse kidney after fasting. By using immunohistochemistry (IHC), we showed these three proteins are co-localized in the outer stripe of the outer medulla (OSOM) of the mouse kidney. We were able to collect this region from the kidney using the Laser Capture Microdissection (LCM) technique. The qPCR data showed significant increase of PGC-1α, ERRα and ERRγ mRNA in the LCM samples after fasting for 24 hours. Furthermore, the known ERRα target genes, mitochondrial oxidative phosphorylation gene COX8H and the tricarboxylic acid (TCA) cycle gene IDH3A also showed an increase. Taken together, our data suggest that fasting activates the energy balance program in the OSOM of the kidney.
Collapse
Affiliation(s)
- Christina T Teng
- Biomolecular Screening Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, United States of America.
| | | | | | | |
Collapse
|
387
|
Scarpulla RC. Nucleus-encoded regulators of mitochondrial function: integration of respiratory chain expression, nutrient sensing and metabolic stress. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2011; 1819:1088-97. [PMID: 22080153 DOI: 10.1016/j.bbagrm.2011.10.011] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 10/28/2011] [Indexed: 12/23/2022]
Abstract
Nucleus-encoded regulatory factors are major contributors to mitochondrial biogenesis and function. Several act within the organelle to regulate mitochondrial transcription and translation while others direct the expression of nuclear genes encoding the respiratory chain and other oxidative functions. Loss-of-function studies for many of these factors reveal a wide spectrum of phenotypes. These range from embryonic lethality and severe respiratory chain deficiency to relatively mild mitochondrial defects seen only under conditions of physiological stress. The PGC-1 family of regulated coactivators (PGC-1α, PGC-1β and PRC) plays an important integrative role through their interactions with transcription factors (NRF-1, NRF-2, ERRα, CREB, YY1 and others) that control respiratory gene expression. In addition, recent evidence suggests that PGC-1 coactivators may balance the cellular response to oxidant stress by promoting a pro-oxidant environment or by orchestrating an inflammatory response to severe metabolic stress. These pathways may serve as essential links between the energy generating functions of mitochondria and the cellular REDOX environment associated with longevity, senescence and disease. This article is part of a Special Issue entitled: Mitochondrial Gene Expression.
Collapse
Affiliation(s)
- Richard C Scarpulla
- Department of Cell and Molecular Biology, Northwestern Medical School, Chicago, IL 60611, USA.
| |
Collapse
|
388
|
Oka S, Alcendor R, Zhai P, Park JY, Shao D, Cho J, Yamamoto T, Tian B, Sadoshima J. PPARα-Sirt1 complex mediates cardiac hypertrophy and failure through suppression of the ERR transcriptional pathway. Cell Metab 2011; 14:598-611. [PMID: 22055503 PMCID: PMC3217210 DOI: 10.1016/j.cmet.2011.10.001] [Citation(s) in RCA: 156] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 07/08/2011] [Accepted: 10/05/2011] [Indexed: 02/08/2023]
Abstract
High energy production in mitochondria is essential for maintaining cardiac contraction in the heart. Genes regulating mitochondrial function are commonly downregulated during heart failure. Here we show that both PPARα and Sirt1 are upregulated by pressure overload in the heart. Haploinsufficiency of either PPARα or Sirt1 attenuated pressure overload-induced cardiac hypertrophy and failure, whereas simultaneous upregulation of PPARα and Sirt1 exacerbated the cardiac dysfunction. PPARα and Sirt1 coordinately suppressed genes involved in mitochondrial function that are regulated by estrogen-related receptors (ERRs). PPARα bound and recruited Sirt1 to the ERR response element (ERRE), thereby suppressing ERR target genes in an RXR-independent manner. Downregulation of ERR target genes was also observed during fasting, and this appeared to be an adaptive response of the heart. These results suggest that suppression of the ERR transcriptional pathway by PPARα/Sirt1, a physiological fasting response, is involved in the progression of heart failure by promoting mitochondrial dysfunction.
Collapse
Affiliation(s)
- Shinichi Oka
- Department of Cell Biology and Molecular Medicine, University of Medicine and Dentistry of New Jersey, Newark, NJ 07103, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
389
|
Peng L, Gao X, Duan L, Ren X, Wu D, Ding K. Identification of Pyrido[1,2-α]pyrimidine-4-ones as New Molecules Improving the Transcriptional Functions of Estrogen-Related Receptor α. J Med Chem 2011; 54:7729-33. [DOI: 10.1021/jm200976s] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Lijie Peng
- Key Laboratory of Regenerative Biology and Institute of Chemical Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China
- Graduate School of Chinese Academy of Sciences, No. 19 Yuquan Road, Beijing 100049, China
| | - Xuefei Gao
- Key Laboratory of Regenerative Biology and Institute of Chemical Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Lei Duan
- Key Laboratory of Regenerative Biology and Institute of Chemical Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Xiaomei Ren
- Key Laboratory of Regenerative Biology and Institute of Chemical Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China
- Zhongshan Medical School, Sun Yat-Sen University, No. 74 2nd Zhongshan Road, Guangzhou 510080 China
| | - Donghai Wu
- Key Laboratory of Regenerative Biology and Institute of Chemical Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Ke Ding
- Key Laboratory of Regenerative Biology and Institute of Chemical Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China
| |
Collapse
|
390
|
An CI, Dong Y, Hagiwara N. Genome-wide mapping of Sox6 binding sites in skeletal muscle reveals both direct and indirect regulation of muscle terminal differentiation by Sox6. BMC DEVELOPMENTAL BIOLOGY 2011; 11:59. [PMID: 21985497 PMCID: PMC3239296 DOI: 10.1186/1471-213x-11-59] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2011] [Accepted: 10/10/2011] [Indexed: 01/06/2023]
Abstract
Background Sox6 is a multi-faceted transcription factor involved in the terminal differentiation of many different cell types in vertebrates. It has been suggested that in mice as well as in zebrafish Sox6 plays a role in the terminal differentiation of skeletal muscle by suppressing transcription of slow fiber specific genes. In order to understand how Sox6 coordinately regulates the transcription of multiple fiber type specific genes during muscle development, we have performed ChIP-seq analyses to identify Sox6 target genes in mouse fetal myotubes and generated muscle-specific Sox6 knockout (KO) mice to determine the Sox6 null muscle phenotype in adult mice. Results We have identified 1,066 Sox6 binding sites using mouse fetal myotubes. The Sox6 binding sites were found to be associated with slow fiber-specific, cardiac, and embryonic isoform genes that are expressed in the sarcomere as well as transcription factor genes known to play roles in muscle development. The concurrently performed RNA polymerase II (Pol II) ChIP-seq analysis revealed that 84% of the Sox6 peak-associated genes exhibited little to no binding of Pol II, suggesting that the majority of the Sox6 target genes are transcriptionally inactive. These results indicate that Sox6 directly regulates terminal differentiation of muscle by affecting the expression of sarcomere protein genes as well as indirectly through influencing the expression of transcription factors relevant to muscle development. Gene expression profiling of Sox6 KO skeletal and cardiac muscle revealed a significant increase in the expression of the genes associated with Sox6 binding. In the absence of the Sox6 gene, there was dramatic upregulation of slow fiber-specific, cardiac, and embryonic isoform gene expression in Sox6 KO skeletal muscle and fetal isoform gene expression in Sox6 KO cardiac muscle, thus confirming the role Sox6 plays as a transcriptional suppressor in muscle development. Conclusions Our present data indicate that during development, Sox6 functions as a transcriptional suppressor of fiber type-specific and developmental isoform genes to promote functional specification of muscle which is critical for optimum muscle performance and health.
Collapse
Affiliation(s)
- Chung-Il An
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, One Shields Avenue, Davis, California 95616, USA
| | | | | |
Collapse
|
391
|
Hu X, Xu X, Lu Z, Zhang P, Fassett J, Zhang Y, Xin Y, Hall JL, Viollet B, Bache RJ, Huang Y, Chen Y. AMP activated protein kinase-α2 regulates expression of estrogen-related receptor-α, a metabolic transcription factor related to heart failure development. Hypertension 2011; 58:696-703. [PMID: 21825219 PMCID: PMC3182261 DOI: 10.1161/hypertensionaha.111.174128] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The normal expression of myocardial mitochondrial enzymes is essential to maintain the cardiac energy reserve and facilitate responses to stress, but the molecular mechanisms to maintain myocardial mitochondrial enzyme expression have been elusive. Here we report that congestive heart failure is associated with a significant decrease of myocardial estrogen-related receptor-α (ERRα), but not peroxisome proliferator-activated receptor-γ coactivator 1α, in human heart failure samples. In addition, chronic pressure overload in mice caused a decrease of ERRα expression that was significantly correlated to the degree of left ventricular dysfunction, pulmonary congestion, and decreases of a group of myocardial energy metabolism-related genes. We found that the metabolic sensor AMP activated protein kinase (AMPK) regulates ERRα expression in vivo and in vitro. AMPKα2 knockout decreased myocardial ERRα (both mRNA and protein) and its downstream targets under basal conditions, with no change in myocardial peroxisome proliferator-activated receptor-γ coactivator 1α expression. Using cultured rat neonatal cardiac myocytes, we found that overexpression of constitutively active AMPKα significantly induced ERRα mRNA, protein, and promoter activity. Conversely, selective gene silencing of AMPKα2 repressed ERRα and its target gene levels, indicating that AMPKα2 is involved in the regulation of ERRα expression. In addition, overexpression of ERRα in AMPKα2 knockout neonatal cardiac myocytes partially rescued the repressed expression of some energy metabolism-related genes. These data support an important role for AMPKα2 in regulating the expression of myocardial ERRα and its downstream mitochondrial enzymes.
Collapse
Affiliation(s)
- Xinli Hu
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
- Lillehei Heart Institute; University of Minnesota, Minneapolis, MN 55455, USA
| | - Xin Xu
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
| | - Zhongbing Lu
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ping Zhang
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
- Lillehei Heart Institute; University of Minnesota, Minneapolis, MN 55455, USA
| | - John Fassett
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ying Zhang
- Molecular and Cellular Biology Laboratory, Beijing Anzhen Hospital, Capital Medical University, Anzhenli, Chaoyang District, Beijing 100029, China
| | - Yi Xin
- Molecular and Cellular Biology Laboratory, Beijing Anzhen Hospital, Capital Medical University, Anzhenli, Chaoyang District, Beijing 100029, China
| | - Jennifer L. Hall
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
- Lillehei Heart Institute; University of Minnesota, Minneapolis, MN 55455, USA
| | - Benoit Viollet
- Inserm, U1016, Institut Cochin; Cnrs, UMR8104; University Paris Descartes, Paris, France
| | - Robert J. Bache
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
- Lillehei Heart Institute; University of Minnesota, Minneapolis, MN 55455, USA
| | - Yimin Huang
- Molecular and Cellular Biology Laboratory, Beijing Anzhen Hospital, Capital Medical University, Anzhenli, Chaoyang District, Beijing 100029, China
| | - Yingjie Chen
- Cardiovascular Division, University of Minnesota, Minneapolis, MN 55455, USA
- Lillehei Heart Institute; University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
392
|
Murray J, Huss JM. Estrogen-related receptor α regulates skeletal myocyte differentiation via modulation of the ERK MAP kinase pathway. Am J Physiol Cell Physiol 2011; 301:C630-45. [PMID: 21562305 PMCID: PMC3174569 DOI: 10.1152/ajpcell.00033.2011] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Accepted: 05/06/2011] [Indexed: 02/06/2023]
Abstract
Myocyte differentiation involves complex interactions between signal transduction pathways and transcription factors. The estrogen-related receptors (ERRs) regulate energy substrate uptake, mitochondrial respiration, and biogenesis and may target structural gene programs in striated muscle. However, ERRα's role in regulating myocyte differentiation is not known. ERRα and peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) are coordinately upregulated with metabolic and skeletal muscle-specific genes early in myogenesis. We analyzed effects of ERRα overexpression and loss of function in myogenic models. In C2C12 myocytes ERRα overexpression accelerated differentiation, whereas XCT790 treatment delayed myogenesis and resulted in myotubes with fewer mitochondria and disorganized sarcomeres. ERRα-/- primary myocytes showed delayed myogenesis, resulting in structurally immature myotubes with reduced sarcomeric assembly and mitochondrial function. However, sarcomeric and metabolic gene expression was unaffected or upregulated in ERRα-/- cells. Instead, ERRα-/- myocytes exhibited aberrant ERK activation early in myogenesis, consistent with delayed myotube formation. XCT790 treatment also increased ERK phosphorylation in C2C12, whereas ERRα overexpression decreased early ERK activation, consistent with the opposing effects of these treatments on differentiation. The transient induction of MAP kinase phosphatase-1 (MKP-1), which mediates ERK dephosphorylation at the onset of myogenesis, was lost in ERRα-/- myocytes and in XCT790-treated C2C12. The ERRα-PGC-1α complex activates the Dusp1 gene, which encodes MKP-1, and ERRα occupies the proximal 5' regulatory region during early differentiation in C2C12 myocytes. Finally, treatment of ERRα-/- myocytes with MEK inhibitors rescued normal ERK signaling and myogenesis. Collectively, these data demonstrate that ERRα is required for normal skeletal myocyte differentiation via modulation of MAP kinase signaling.
Collapse
MESH Headings
- Animals
- Butadienes/pharmacology
- Cell Differentiation/drug effects
- Cell Differentiation/physiology
- Creatine Kinase, Mitochondrial Form/genetics
- Dual Specificity Phosphatase 1/genetics
- Dual Specificity Phosphatase 1/metabolism
- Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Flavonoids/pharmacology
- Gene Expression/drug effects
- Gene Expression/genetics
- Kinetics
- MAP Kinase Signaling System/drug effects
- MAP Kinase Signaling System/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria/metabolism
- Mitochondria/pathology
- Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors
- Mitogen-Activated Protein Kinase Kinases/metabolism
- Muscle Development/drug effects
- Muscle Development/physiology
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/enzymology
- Muscle Fibers, Skeletal/metabolism
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Myoblasts, Skeletal/cytology
- Myoblasts, Skeletal/drug effects
- Myoblasts, Skeletal/enzymology
- Myoblasts, Skeletal/metabolism
- Myogenin/genetics
- Myosin Heavy Chains/genetics
- Nitriles/pharmacology
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha
- Phosphorylation/drug effects
- Protein Binding/genetics
- Protein Serine-Threonine Kinases/genetics
- Pyruvate Dehydrogenase Acetyl-Transferring Kinase
- Receptors, Estrogen/antagonists & inhibitors
- Receptors, Estrogen/deficiency
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Ribosomal Protein S6 Kinases, 90-kDa/metabolism
- Sarcomeres/pathology
- Thiazoles/pharmacology
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcription Factors
- Transduction, Genetic
- Troponin I/genetics
- Troponin I/metabolism
- Up-Regulation/drug effects
- Up-Regulation/genetics
- p38 Mitogen-Activated Protein Kinases/metabolism
- ERRalpha Estrogen-Related Receptor
Collapse
Affiliation(s)
- Jennifer Murray
- Division of Gene Regulation and Drug Discovery, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope, Duarte, California, USA
| | | |
Collapse
|
393
|
Lu N, Wang W, Liu J, Wong CW. Protein kinase C epsilon affects mitochondrial function through estrogen-related receptor alpha. Cell Signal 2011; 23:1473-8. [PMID: 21596133 DOI: 10.1016/j.cellsig.2011.04.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 04/23/2011] [Accepted: 04/30/2011] [Indexed: 11/27/2022]
Abstract
Members of the protein kinase C (PKC) family have been implicated in controlling cell proliferation, differentiation, and motility. Many of these processes are energy demanding. How PKCs affect mitochondrial function to regulate energy production is not well defined. Using an inhibitor Gö6983 with broad specificity, we found that inhibiting PKCs reduced mitochondrial mass and altered mitochondrial function characterized by elevations in mitochondrial membrane potential (Δψm) and reactive oxygen species (ROS) levels. These alterations indicated that Gö6983 suppressed the activities of mitochondrial regulators such as estrogen-related receptor α (ERRα). Indeed, Gö6983 dose-dependently suppressed the expression levels of ERRα-target genes peroxisome proliferator-activated receptor α (PPARα) and medium-chain acyl-CoA dehydrogenase (MCAD). Conversely, PKC activator phorbol ester (PMA) enhanced the expression level of another ERRα-target gene pyruvate dehydrogenase kinase 4 (PDK4). This PMA-mediated induction of PDK4 was blunted by an ERRα inverse agonist XCT-790, suggesting that ERRα plays a role in mediating the effects of PKCs on mitochondrial function. By over-expressing constitutively active forms of PKCs, we found that PKCε preferentially stimulated the transcription activity of ERRα. Through mutating residues on ERRα, we established that this PKCε-induced ERRα activity involves threonine 106, serine 110, and threonine 124 of ERRα. Collectively, these pieces of evidence suggest that ERRα plays an important role down-stream of PKCε to regulate mitochondrial homeostasis.
Collapse
Affiliation(s)
- Na Lu
- Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou 510530, China
| | | | | | | |
Collapse
|
394
|
Deblois G, Giguère V. Functional and physiological genomics of estrogen-related receptors (ERRs) in health and disease. Biochim Biophys Acta Mol Basis Dis 2011; 1812:1032-40. [DOI: 10.1016/j.bbadis.2010.12.009] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Revised: 12/09/2010] [Accepted: 12/10/2010] [Indexed: 12/11/2022]
|
395
|
Sun Z, Singh N, Mullican SE, Everett LJ, Li L, Yuan L, Liu X, Epstein JA, Lazar MA. Diet-induced lethality due to deletion of the Hdac3 gene in heart and skeletal muscle. J Biol Chem 2011; 286:33301-9. [PMID: 21808063 DOI: 10.1074/jbc.m111.277707] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Many human diseases result from the influence of the nutritional environment on gene expression. The environment interacts with the genome by altering the epigenome, including covalent modification of nucleosomal histones. Here, we report a novel and dramatic influence of diet on the phenotype and survival of mice in which histone deacetylase 3 (Hdac3) is deleted postnatally in heart and skeletal muscle. Although embryonic deletion of myocardial Hdac3 causes major cardiomyopathy that reduces survival, we found that excision of Hdac3 in heart and muscle later in development leads to a much milder phenotype and does not reduce survival when mice are fed normal chow. Remarkably, upon switching to a high fat diet, the mice begin to die within weeks and display signs of severe hypertrophic cardiomyopathy and heart failure. Down-regulation of myocardial mitochondrial bioenergetic genes, specifically those involved in lipid metabolism, precedes the full development of cardiomyopathy, suggesting that HDAC3 is important in maintaining proper mitochondrial function. These data suggest that loss of the epigenomic modifier HDAC3 causes dietary lethality by compromising the ability of cardiac mitochondria to respond to changes of nutritional environment. In addition, this study provides a mouse model for diet-inducible heart failure.
Collapse
Affiliation(s)
- Zheng Sun
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine and Department of Genetics, and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
396
|
Huang C, Zhou T, Chen Y, Sun T, Zhang S, Chen G. Estrogen-related receptor ERRα-mediated downregulation of human hydroxysteroid sulfotransferase (SULT2A1) in Hep G2 cells. Chem Biol Interact 2011; 192:264-71. [PMID: 21513704 PMCID: PMC3111048 DOI: 10.1016/j.cbi.2011.04.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 03/30/2011] [Accepted: 04/07/2011] [Indexed: 01/14/2023]
Abstract
Hydroxysteroid sulfotransferase SULT2A1 catalyzes the sulfation of hydroxysteroids and xenobiotics. It plays an important role in the detoxification of hydroxyl-containing xenobiotics and in the regulation of the biological activities of hydroxysteroids. ERRα is an orphan member of the nuclear receptor superfamily that is closely related to estrogen receptor alpha (ERα). Here we report that the mRNA expression of human SULT2A1 was suppressed by ERRα in Hep G2 cells. To investigate the mechanisms of this regulation, the effects of ERRα on human SULT2A1 promoter transcription in Hep G2 cells were investigated. Reporter luciferase assay results showed that ERRα significantly represses human SULT2A1 promoter transcription in Hep G2 cells. Deletion analysis indicated that human SULT2A1 promoter region between positions -188 and -130 is necessary for its repression by ERRα in Hep G2 cells. The 5' DNA -188 to -130 region of human SULT2A1 contains IR2 and DR4 hormone response elements and two putative ERRα response elements (ERREs) (ERRE188: GCAAGCTCA and ERRE155: ATAAGTTCA). Interestingly, ERRE188 overlaps with the IR2 element and ERRE155 overlaps with the DR4 element. Our further investigation demonstrated that ERRα represses human SULT2A1 promoter transcription by competing with other nuclear receptors for binding to IR2 or DR4 elements. The interaction of ERRE188 and ERRE155 elements with ERRα was confirmed by electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) analysis. Our results suggest that ERRα may play an important role in regulating the metabolism of drugs and xenobiotics and in regulating endogenous hydroxysteroid activities via the regulation of SULT2A1.
Collapse
Affiliation(s)
- Chaoqun Huang
- Department of Physiological Sciences, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, 74078 USA
| | - Tianyan Zhou
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100083, China
| | - Yue Chen
- Department of Physiological Sciences, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, 74078 USA
| | - Teng Sun
- Department of Physiological Sciences, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, 74078 USA
| | - Shufen Zhang
- Department of Physiological Sciences, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, 74078 USA
| | - Guangping Chen
- Department of Physiological Sciences, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, 74078 USA
| |
Collapse
|
397
|
Scarpulla RC. Metabolic control of mitochondrial biogenesis through the PGC-1 family regulatory network. BIOCHIMICA ET BIOPHYSICA ACTA 2011; 1813:1269-78. [PMID: 20933024 PMCID: PMC3035754 DOI: 10.1016/j.bbamcr.2010.09.019] [Citation(s) in RCA: 887] [Impact Index Per Article: 68.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Revised: 09/14/2010] [Accepted: 09/27/2010] [Indexed: 12/23/2022]
Abstract
The PGC-1 family of regulated coactivators, consisting of PGC-1α, PGC-1β and PRC, plays a central role in a regulatory network governing the transcriptional control of mitochondrial biogenesis and respiratory function. These coactivators target multiple transcription factors including NRF-1, NRF-2 and the orphan nuclear hormone receptor, ERRα, among others. In addition, they themselves are the targets of coactivator and co-repressor complexes that regulate gene expression through chromatin remodeling. The expression of PGC-1 family members is modulated by extracellular signals controlling metabolism, differentiation or cell growth and in some cases their activities are known to be regulated by post-translational modification by the energy sensors, AMPK and SIRT1. Recent gene knockout and silencing studies of many members of the PGC-1 network have revealed phenotypes of wide ranging severity suggestive of complex compensatory interactions or broadly integrative functions that are not exclusive to mitochondrial biogenesis. The results point to a central role for the PGC-1 family in integrating mitochondrial biogenesis and energy production with many diverse cellular functions. This article is part of a Special Issue entitled: Mitochondria and Cardioprotection.
Collapse
Affiliation(s)
- Richard C Scarpulla
- Department of Cell and Molecular Biology, Northwestern Medical School, 303 East Chicago Avenue, Chicago, IL 60611, USA.
| |
Collapse
|
398
|
Dwivedi SKD, Singh N, Kumari R, Mishra JS, Tripathi S, Banerjee P, Shah P, Kukshal V, Tyagi AM, Gaikwad AN, Chaturvedi RK, Mishra DP, Trivedi AK, Sanyal S, Chattopadhyay N, Ramachandran R, Siddiqi MI, Bandyopadhyay A, Arora A, Lundåsen T, Anakk SP, Moore DD, Sanyal S. Bile acid receptor agonist GW4064 regulates PPARγ coactivator-1α expression through estrogen receptor-related receptor α. Mol Endocrinol 2011; 25:922-32. [PMID: 21493670 PMCID: PMC5417256 DOI: 10.1210/me.2010-0512] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Accepted: 03/29/2011] [Indexed: 01/08/2023] Open
Abstract
Peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) is induced in energy-starved conditions and is a key regulator of energy homeostasis. This makes PGC-1α an attractive therapeutic target for metabolic syndrome and diabetes. In our effort to identify new regulators of PGC-1α expression, we found that GW4064, a widely used synthetic agonist for the nuclear bile acid receptor [farnesoid X receptor (FXR)] strongly enhances PGC-1α promoter reporter activity, mRNA, and protein expression. This induction in PGC-1α concomitantly enhances mitochondrial mass and expression of several PGC-1α target genes involved in mitochondrial function. Using FXR-rich or FXR-nonexpressing cell lines and tissues, we found that this effect of GW4064 is not mediated directly by FXR but occurs via activation of estrogen receptor-related receptor α (ERRα). Cell-based, biochemical and biophysical assays indicate GW4064 as an agonist of ERR proteins. Interestingly, FXR disruption alters GW4064 induction of PGC-1α mRNA in a tissue-dependent manner. Using FXR-null [FXR knockout (FXRKO)] mice, we determined that GW4064 induction of PGC-1α expression is not affected in oxidative soleus muscles of FXRKO mice but is compromised in the FXRKO liver. Mechanistic studies to explain these differences revealed that FXR physically interacts with ERR and protects them from repression by the atypical corepressor, small heterodimer partner in liver. Together, this interplay between ERRα-FXR-PGC-1α and small heterodimer partner offers new insights into the biological functions of ERRα and FXR, thus providing a knowledge base for therapeutics in energy balance-related pathophysiology.
Collapse
MESH Headings
- Animals
- Binding Sites
- Cell Line
- Genes, Reporter
- Humans
- Isoxazoles/chemistry
- Isoxazoles/pharmacology
- Liver/metabolism
- Luciferases/genetics
- Luciferases/metabolism
- Mice
- Mice, Knockout
- Muscle, Skeletal/metabolism
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha
- Promoter Regions, Genetic
- Rats
- Receptors, Cytoplasmic and Nuclear/agonists
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Estrogen/chemistry
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Thermodynamics
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcription Factors
- Transcription, Genetic/drug effects
- ERRalpha Estrogen-Related Receptor
Collapse
Affiliation(s)
- Shailendra Kumar Dhar Dwivedi
- Endocrinology Division, Institute of Toxicological Research, Council of Scientific and Industrial Research, Lucknow 226001, India
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
399
|
Gao M, Wang J, Lu N, Fang F, Liu J, Wong CW. Mitogen-activated protein kinase kinases promote mitochondrial biogenesis in part through inducing peroxisome proliferator-activated receptor γ coactivator-1β expression. BIOCHIMICA ET BIOPHYSICA ACTA 2011; 1813:1239-44. [PMID: 21458501 DOI: 10.1016/j.bbamcr.2011.03.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 03/22/2011] [Accepted: 03/23/2011] [Indexed: 01/08/2023]
Abstract
Growth factor activates mitogen-activated protein kinase kinases to promote cell growth. Mitochondrial biogenesis is an integral part of cell growth. How growth factor regulates mitochondrial biogenesis is not fully understood. In this study, we found that mitochondrial mass was specifically reduced upon serum starvation and induced upon re-feeding with serum. Using mitogen-activated protein kinase kinases inhibitor U0126, we found that the mRNA expression levels of ATP synthase, cytochrome-C, mitochondrial transcription factor A, and mitofusin 2 were reduced. Since the transcriptional levels of these genes are under the control of peroxisome proliferator-activated receptor γ coactivator-1α and -1β (PGC-1α and PGC-1β), we examined and found that only the mRNA and protein levels of PGC-1β were suppressed. Importantly, over-expression of PGC-1β partially reversed the reduction of mitochondrial mass upon U0126 treatment. Thus, we conclude that mitogen-activated protein kinase kinases direct mitochondrial biogenesis through selectively inducing PGC-1β expression.
Collapse
Affiliation(s)
- Minghui Gao
- Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, 510530 China
| | | | | | | | | | | |
Collapse
|
400
|
Dufour CR, Levasseur MP, Pham NHH, Eichner LJ, Wilson BJ, Charest-Marcotte A, Duguay D, Poirier-Héon JF, Cermakian N, Giguère V. Genomic convergence among ERRα, PROX1, and BMAL1 in the control of metabolic clock outputs. PLoS Genet 2011; 7:e1002143. [PMID: 21731503 PMCID: PMC3121748 DOI: 10.1371/journal.pgen.1002143] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Accepted: 05/03/2011] [Indexed: 01/12/2023] Open
Abstract
Metabolic homeostasis and circadian rhythms are closely intertwined biological processes. Nuclear receptors, as sensors of hormonal and nutrient status, are actively implicated in maintaining this physiological relationship. Although the orphan nuclear receptor estrogen-related receptor α (ERRα, NR3B1) plays a central role in the control of energy metabolism and its expression is known to be cyclic in the liver, its role in temporal control of metabolic networks is unknown. Here we report that ERRα directly regulates all major components of the molecular clock. ERRα-null mice also display deregulated locomotor activity rhythms and circadian period lengths under free-running conditions, as well as altered circulating diurnal bile acid and lipid profiles. In addition, the ERRα-null mice exhibit time-dependent hypoglycemia and hypoinsulinemia, suggesting a role for ERRα in modulating insulin sensitivity and glucose handling during the 24-hour light/dark cycle. We also provide evidence that the newly identified ERRα corepressor PROX1 is implicated in rhythmic control of metabolic outputs. To help uncover the molecular basis of these phenotypes, we performed genome-wide location analyses of binding events by ERRα, PROX1, and BMAL1, an integral component of the molecular clock. These studies revealed the existence of transcriptional regulatory loops among ERRα, PROX1, and BMAL1, as well as extensive overlaps in their target genes, implicating these three factors in the control of clock and metabolic gene networks in the liver. Genomic convergence of ERRα, PROX1, and BMAL1 transcriptional activity thus identified a novel node in the molecular circuitry controlling the daily timing of metabolic processes.
Collapse
Affiliation(s)
| | - Marie-Pier Levasseur
- Goodman Cancer Research Centre, Montréal, Canada
- Department of Biochemistry, McGill University, Montréal, Canada
| | | | - Lillian J. Eichner
- Goodman Cancer Research Centre, Montréal, Canada
- Department of Biochemistry, McGill University, Montréal, Canada
| | | | - Alexis Charest-Marcotte
- Goodman Cancer Research Centre, Montréal, Canada
- Department of Biochemistry, McGill University, Montréal, Canada
| | - David Duguay
- Laboratory of Molecular Chronobiology, Douglas Mental Health University Institute, Montréal, Canada
- Department of Psychiatry, McGill University, Montréal, Canada
| | - Jean-François Poirier-Héon
- Laboratory of Molecular Chronobiology, Douglas Mental Health University Institute, Montréal, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, Canada
| | - Nicolas Cermakian
- Laboratory of Molecular Chronobiology, Douglas Mental Health University Institute, Montréal, Canada
- Department of Psychiatry, McGill University, Montréal, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, Canada
| | - Vincent Giguère
- Goodman Cancer Research Centre, Montréal, Canada
- Department of Biochemistry, McGill University, Montréal, Canada
- Department of Medicine, McGill University, Montréal, Canada
- Department of Oncology, McGill University, Montréal, Canada
| |
Collapse
|