151
|
Baal N, Cunningham S, Obermann HL, Thomas J, Lippitsch A, Dietert K, Gruber AD, Kaufmann A, Michel G, Nist A, Stiewe T, Rupp O, Goesmann A, Zukunft S, Fleming I, Bein G, Lohmeyer J, Bauer S, Hackstein H. ADAR1 Is Required for Dendritic Cell Subset Homeostasis and Alveolar Macrophage Function. THE JOURNAL OF IMMUNOLOGY 2019; 202:1099-1111. [DOI: 10.4049/jimmunol.1800269] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 12/07/2018] [Indexed: 11/19/2022]
|
152
|
Huang B, Luo N, Wu X, Xu Z, Wang X, Pan X. The modulatory role of low concentrations of bisphenol A on tamoxifen-induced proliferation and apoptosis in breast cancer cells. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:2353-2362. [PMID: 30467747 DOI: 10.1007/s11356-018-3780-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 11/15/2018] [Indexed: 06/09/2023]
Abstract
Selective estrogen receptor modulators such as tamoxifen (TAM) significantly reduce the risks of developing estrogen receptor-positive (ER+) breast cancer. Low concentrations (nanomolar range) of bisphenol A (BPA) shows estrogenic effects and further promotes the proliferation of hormone-dependent breast cancer cells. However, whether or not BPA can influence TAM-treatment resistance in breast cancer has not drawn much attention. In the current study, low concentrations of BPA reduced TAM-induced cytotoxicity of MCF-7 cells, which was proved by the suppression of cell apoptosis, transition of cell cycle from G1 to S phase, and upregulation of cyclin D1 and ERα. Simultaneously, the mRNA levels of estrogen-related receptor γ (ERRγ) and its coactivators, peroxisome proliferation-activated receptor γ coactivator-1α (PGC-1α), and PGC-1β, were increased. However, the similar effects were not observed in MDA-MB-231 cells. Our results indicated that low concentrations of BPA decreased the sensitivity of TAM in MCF-7 cells rather than in MDA-MB-231 cells. These different actions likely involved the interaction of relative receptors and coactivators. This study provided a possible support that the exposure of BPA in environmental media may potentially induce TAM resistance to breast cancer treatment.
Collapse
Affiliation(s)
- Bin Huang
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Nao Luo
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Xinhao Wu
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Zhixiang Xu
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China.
| | - Xiaoxia Wang
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Xuejun Pan
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China.
| |
Collapse
|
153
|
Anelli GM, Mandò C, Letizia T, Mazzocco MI, Novielli C, Lisso F, Personeni C, Vago T, Cetin I. Placental ESRRG-CYP19A1 Expressions and Circulating 17-Beta Estradiol in IUGR Pregnancies. Front Pediatr 2019; 7:154. [PMID: 31069202 PMCID: PMC6491753 DOI: 10.3389/fped.2019.00154] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/01/2019] [Indexed: 12/04/2022] Open
Abstract
Introduction: Sex steroids are regulating factors for intrauterine growth. 17-β Estradiol (E2) is particularly critical to a physiological pregnancy, as increased maternal E2 was correlated to lower fetal weight at delivery. The placenta itself is a primary source of estrogens, synthetized from cholesterol precursors. Cytochrome P450 aromatase (encoded by CYP19A1 gene) is a rate-limiting enzyme for E2 biosynthesis. CYP19A1 transcription is supported by Estrogen Related-Receptor Gamma (ERRγ- ESRRG gene), which thus has an indirect role in placental steroidogenesis. Here we investigated maternal E2 levels and placental CYP19A1 and ESRRG expressions in pregnancies with IntraUterine Growth Restriction (IUGR). Methods: Singleton pregnancies were studied. E2 was measured in maternal plasma by electrochemiluminescence in 16 term controls and 11 IUGR (classified by umbilical artery doppler pulsatility index) at elective cesarean section, and also in 13 controls during pregnancy at a gestational age comparable to IUGR. CYP19A1 and ESRRG expressions were analyzed in placental tissue. Maternal/fetal characteristics, placental and molecular data were compared among study groups and tested for correlations. Results: Maternal E2 plasma concentrations were significantly decreased in IUGR compared to controls at delivery. When analyzing normal pregnancies at a gestational age similar to IUGR, E2 levels were not different to pathological cases. However, E2 levels at delivery positively correlated with placental efficiency. Placental CYP19A1 levels were significantly higher in IUGR placental tissue vs. controls, and specifically increased in female IUGR placentas. ESRRG expression was not different among groups. Discussion: We report a positive correlation between 17-β Estradiol levels and placental efficiency, that might indicate a disrupted steroidogenesis in IUGR pregnancies. Moreover, we show alterations of CYP19A1 expression in IUGR placentas, possibly indicating a compensatory effect to the adverse IUGR intrauterine environment, also depending on fetal sex. Further studies are needed to deeper investigate IUGR alterations in the complex interaction among molecules involved in placental steroidogenesis.
Collapse
Affiliation(s)
- Gaia Maria Anelli
- Unit of Obstetrics and Gynecology, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco University Hospital, University of Milan, Milan, Italy
| | - Chiara Mandò
- Unit of Obstetrics and Gynecology, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco University Hospital, University of Milan, Milan, Italy
| | - Teresa Letizia
- Endocrinology Laboratory, Sacco University Hospital, Milan, Italy
| | - Martina Ilaria Mazzocco
- Unit of Obstetrics and Gynecology, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco University Hospital, University of Milan, Milan, Italy
| | - Chiara Novielli
- Unit of Obstetrics and Gynecology, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco University Hospital, University of Milan, Milan, Italy
| | - Fabrizia Lisso
- Unit of Obstetrics and Gynecology, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco University Hospital, University of Milan, Milan, Italy
| | - Carlo Personeni
- Unit of Obstetrics and Gynecology, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco University Hospital, University of Milan, Milan, Italy
| | - Tarcisio Vago
- Endocrinology Laboratory, Sacco University Hospital, Milan, Italy
| | - Irene Cetin
- Unit of Obstetrics and Gynecology, Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco University Hospital, University of Milan, Milan, Italy.,Unit of Obstetrics and Gynecology, Buzzi University Hospital, Milan, Italy
| |
Collapse
|
154
|
Liu G, Sun P, Dong B, Sehouli J. Key regulator of cellular metabolism, estrogen-related receptor α, a new therapeutic target in endocrine-related gynecological tumor. Cancer Manag Res 2018; 10:6887-6895. [PMID: 30588094 PMCID: PMC6296681 DOI: 10.2147/cmar.s182466] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The estrogen-related receptor α (ERRα), is an orphan transcription factor. Recently, many studies have reported its regulatory mechanisms and transcriptional targets after identification. Therefore, it may be eligible to join the rank of other nuclear receptors that control almost all aspects of cell metabolism. Cellular metabolism reprogramming plays a key role in fueling malignant change. The purpose of this review was to demonstrate that the ERRα plays an important role in the association between gynecological endocrine-related tumors and energy metabolism. Furthermore, regulation of ERRα may represent a promising strategy to induce cellular metabolic vulnerability of cancer from different origins. Thus, a comprehensive understanding of current treatment strategies may be achieved.
Collapse
Affiliation(s)
- GuiFen Liu
- Laboratory of Gynaecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, People's Republic of China,
| | - PengMing Sun
- Laboratory of Gynaecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, People's Republic of China, .,Department of Gynaecology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, People's Republic of China,
| | - BinHua Dong
- Laboratory of Gynaecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, People's Republic of China,
| | - Jalid Sehouli
- Department of Gynaecologic Oncology and Gynaecology, Charité/Campus Virchow-Klinikum, European Competence Centre for Ovarian Cancer University of Berlin, Berlin 13353, Germany
| |
Collapse
|
155
|
Stevens JF, Revel JS, Maier CS. Mitochondria-Centric Review of Polyphenol Bioactivity in Cancer Models. Antioxid Redox Signal 2018; 29:1589-1611. [PMID: 29084444 PMCID: PMC6207154 DOI: 10.1089/ars.2017.7404] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 10/28/2017] [Indexed: 12/17/2022]
Abstract
SIGNIFICANCE Humans are exposed daily to polyphenols in milligram-to-gram amounts through dietary consumption of fruits and vegetables. Polyphenols are also available as components of dietary supplements for improving general health. Although polyphenols are often advertised as antioxidants to explain health benefits, experimental evidence shows that their beneficial cancer preventing and controlling properties are more likely due to stimulation of pro-oxidant and proapoptotic pathways. Recent Advances: The understanding of the biological differences between cancer and normal cell, and especially the role that mitochondria play in carcinogenesis, has greatly advanced in recent years. These advances have resulted in a wealth of new information on polyphenol bioactivity in cell culture and animal models of cancer. Polyphenols appear to target oxidative phosphorylation and regulation of the mitochondrial membrane potential (MMP), glycolysis, pro-oxidant pathways, and antioxidant (adaptive) stress responses with greater selectivity in tumorigenic cells. CRITICAL ISSUES The ability of polyphenols to dissipate the MMP (Δψm) by a protonophore mechanism has been known for more than 50 years. However, researchers focus primarily on the downstream molecular effects of Δψm dissipation and mitochondrial uncoupling. We argue that the physicochemical properties of polyphenols are responsible for their anticancer properties by virtue of their protonophoric and pro-oxidant properties rather than their specific effects on downstream molecular targets. FUTURE DIRECTIONS Polyphenol-induced dissipation of Δψm is a physicochemical process that cancer cells cannot develop resistance against by gene mutation. Therefore, polyphenols should receive more attention as agents for cotherapy with cancer drugs to gain synergistic activity. Antioxid. Redox Signal.
Collapse
Affiliation(s)
- Jan F. Stevens
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, Oregon
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon
| | - Johana S. Revel
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, Oregon
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon
- Department of Chemistry, Oregon State University, Corvallis, Oregon
| | - Claudia S. Maier
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon
- Department of Chemistry, Oregon State University, Corvallis, Oregon
| |
Collapse
|
156
|
Khalturin K, Billas IML, Chebaro Y, Reitzel AM, Tarrant AM, Laudet V, Markov GV. NR3E receptors in cnidarians: A new family of steroid receptor relatives extends the possible mechanisms for ligand binding. J Steroid Biochem Mol Biol 2018; 184:11-19. [PMID: 29940311 PMCID: PMC6240368 DOI: 10.1016/j.jsbmb.2018.06.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 06/20/2018] [Accepted: 06/22/2018] [Indexed: 01/21/2023]
Abstract
Steroid hormone receptors are important regulators of development and physiology in bilaterian animals, but the role of steroid signaling in cnidarians has been contentious. Cnidarians produce steroids, including A-ring aromatic steroids with a side-chain, but these are probably made through pathways different than the one used by vertebrates to make their A-ring aromatic steroids. Here we present comparative genomic analyses indicating the presence of a previously undescribed nuclear receptor family within medusozoan cnidarians, that we propose to call NR3E. This family predates the diversification of ERR/ER/SR in bilaterians, indicating that the first NR3 evolved in the common ancestor of the placozoan and cnidarian-bilaterian with lineage-specific loss in the anthozoans, even though multiple species in this lineage have been shown to produce aromatic steroids, whose function remain unclear. We discovered serendipitously that a cytoplasmic factor within epidermal cells of transgenic Hydra vulgaris can trigger the nuclear translocation of heterologously expressed human ERα. This led us to hypothesize that aromatic steroids may also be present in the medusozoan cnidarian lineage, which includes Hydra, and may explain the translocation of human ERα. Docking experiments with paraestrol A, a cnidarian A-ring aromatic steroid, into the ligand-binding pocket of Hydra NR3E indicates that, if an aromatic steroid is indeed the true ligand, which remains to be demonstrated, it would bind to the pocket through a partially distinct mechanism from the manner in which estradiol binds to vertebrate ER.
Collapse
Affiliation(s)
- Konstantin Khalturin
- Marine Genomics Unit, Okinawa Institute of Science and Technology, 1919-1 Tancha, Onna-son, Kunigami-gun, Okinawa 904-0495, Japan
| | - Isabelle M L Billas
- Centre for Integrative Biology (CBI), Department of Integrated Structural Biology, IGBMC (Institute of Genetics and of Molecular and Cellular Biology), Illkirch, France; Centre National de la Recherche Scientifique (CNRS) UMR 7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U964, Illkirch, France; Université de Strasbourg, Strasbourg, France
| | - Yassmine Chebaro
- Centre for Integrative Biology (CBI), Department of Integrated Structural Biology, IGBMC (Institute of Genetics and of Molecular and Cellular Biology), Illkirch, France; Centre National de la Recherche Scientifique (CNRS) UMR 7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U964, Illkirch, France; Université de Strasbourg, Strasbourg, France
| | - Adam M Reitzel
- Department of Biological Sciences, University of North Carolina, Charlotte, 9201 University City Blvd, Charlotte, NC 28223, USA
| | - Ann M Tarrant
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, USA
| | - Vincent Laudet
- Sorbonne Université, CNRS, Observatoire océanologique de Banyuls-sur-mer, Avenue de Fontaule, 66650 Banyuls-sur-mer, France
| | - Gabriel V Markov
- Sorbonne Université, CNRS, UMR 8227 Integrative Biology of Marine Models, Station Biologique de Roscoff, Place Georges Teissier, CS 90074, 29688 Roscoff Cedex, France.
| |
Collapse
|
157
|
Xu Z, Wang Y, Xiao ZG, Zou C, Zhang X, Wang Z, Wu D, Yu S, Chan FL. Nuclear receptor ERRα and transcription factor ERG form a reciprocal loop in the regulation of TMPRSS2:ERG fusion gene in prostate cancer. Oncogene 2018; 37:6259-6274. [PMID: 30042415 PMCID: PMC6265259 DOI: 10.1038/s41388-018-0409-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 05/17/2018] [Accepted: 06/19/2018] [Indexed: 11/08/2022]
Abstract
The TMPRSS2:ERG (T:E) fusion gene is generally believed to be mainly regulated by the activated androgen receptor (AR) signaling in androgen-dependent prostate cancer. However, its persistent expression in castration-resistant and neuroendocrine prostate cancers implies that other transcription factors might also regulate its expression. Here, we showed that up-regulation of nuclear receptor estrogen-related receptor alpha (ERRα) was closely associated with the oncogenic transcription factor ERG expression in prostate cancer, and their increased coexpression patterns were closely associated with high Gleason scores and metastasis in patients. Both ERRα and ERG exhibited a positive expression correlation in a castration-resistant prostate cancer (CRPC) xenograft model VCaP-CRPC. We showed that ERRα could directly transactivate T:E fusion gene in both AR-positive and -negative prostate cancer cells via both ERR-binding element- and AR-binding element-dependent manners. Ectopic T:E expression under ERRα regulation could promote both in vitro invasion and in vivo metastasis capacities of AR-negative prostatic cells. Intriguingly, ERG expressed by the T:E fusion could also transactivate the ERRα (ESRRA) gene. Hereby, ERRα and ERG can synergistically regulate each other and form a reciprocal regulatory loop to promote the advanced growth of prostate cancer. Inhibition of ERRα activity by ERRα inverse agonist could suppress T:E expression in prostate cancer cells, implicating that targeting ERRα could be a potential therapeutic strategy for treating the aggressive T:E-positive prostate cancer.
Collapse
Affiliation(s)
- Zhenyu Xu
- Department of Pharmacy, Yijishan Affiliated Hospital, Wannan Medical College, Wuhu, Anhui, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yuliang Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhan Gang Xiao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Department of Pharmacology, Southwest Medical University, Luzhou, China
| | - Chang Zou
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Clinical Medical Research Center, Shenzhen People's Hospital, Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Xian Zhang
- Department of Pharmacy, Yijishan Affiliated Hospital, Wannan Medical College, Wuhu, Anhui, China
| | - Zhu Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Dinglan Wu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Shan Yu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| | - Franky Leung Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
158
|
Ampawong S, Isarangkul D, Reamtong O, Aramwit P. Adaptive effect of sericin on hepatic mitochondrial conformation through its regulation of apoptosis, autophagy and energy maintenance: a proteomics approach. Sci Rep 2018; 8:14943. [PMID: 30297713 PMCID: PMC6175853 DOI: 10.1038/s41598-018-33372-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/27/2018] [Indexed: 12/22/2022] Open
Abstract
We recently demonstrated that in addition to its protective effect on pancreatic and adrenal biosynthesis, antioxidant properties of sericin decrease blood cholesterol levels and improve the liver mitochondrial architecture. However, little is known about the detailed mechanisms underlying these effects. Using proteomics and electron microscopy, we identified mitochondrial proteins that play important roles in the preservation of the mitochondrial ultrastructure and cholesterol-lowering properties of sericin. Our results showed that sericin maintains the mitochondrial architecture during conditions of high blood cholesterol by regulating apoptotic (NADH-ubiquinone oxidoreductase 75 kDa subunit) and autophagic (mitochondrial elongation factor Tu and prohibitin-2) proteins as well as energy maintenance proteins [haloacid dehalogenase-like hydrolase domain-containing protein 3, succinate dehydrogenase (ubiquinone) flavoprotein subunit, ATP synthase-α subunit precursor, enoyl-CoA hydratase domain-containing protein 3 and electron transfer flavoprotein subunit-α]. Sericin also exerts anti-oxidative properties via aconitate hydratase and Chain A, crystal structure of rat carnitine palmitoyltrasferase 2 proteins. Together, these activities may reduce hepatocytic triglyceride deposition, thereby decreasing steatosis, as demonstrated by the modulatory effects on ornithine aminotransferase, mitochondrial aspartate aminotransferase, acyl-CoA synthase, hydroxyacyl-CoA dehydrogenase and D-beta-hydroxybutyrate dehydrogenase. Sericin activity further balanced nitrogenous waste detoxification, characterised by carbamoyl-phosphate synthase (ammonia), aldehyde dehydrogenase and uricase, or folate biosynthesis via sarcosine dehydrogenase and dimethyl glycine dehydrogenase. These results suggest that sericin maintains the hepatic mitochondrial architecture through apoptotic, autophagic, energy maintenance and anti-oxidative mitochondrial proteins for alleviating hepatic steatosis and promoting liver function under conditions of hypercholesterolaemia.
Collapse
Affiliation(s)
- Sumate Ampawong
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Ratchawithi Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Duangnate Isarangkul
- Department of Microbiology, Faculty of Science, Mahidol University, 272, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetic, Faculty of Tropical Medicine, Mahidol University, Ratchawithi Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Pornanong Aramwit
- Bioactive Resources for Innovative Clinical Applications Research Unit and Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Chulalongkorn University, PhayaThai Road, Phatumwan, Bangkok, 10330, Thailand.
| |
Collapse
|
159
|
Moulton VR. Sex Hormones in Acquired Immunity and Autoimmune Disease. Front Immunol 2018; 9:2279. [PMID: 30337927 PMCID: PMC6180207 DOI: 10.3389/fimmu.2018.02279] [Citation(s) in RCA: 329] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 09/13/2018] [Indexed: 12/15/2022] Open
Abstract
Women have stronger immune responses to infections and vaccination than men. Paradoxically, the stronger immune response comes at a steep price, which is the high incidence of autoimmune diseases in women. The reasons why women have stronger immunity and higher incidence of autoimmunity are not clear. Besides gender, sex hormones contribute to the development and activity of the immune system, accounting for differences in gender-related immune responses. Both innate and adaptive immune systems bear receptors for sex hormones and respond to hormonal cues. This review focuses on the role of sex hormones particularly estrogen, in the adaptive immune response, in health, and autoimmune disease with an emphasis on systemic lupus erythematosus.
Collapse
Affiliation(s)
- Vaishali R Moulton
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
160
|
Zhang JJ, Xi GS. Morphological changes in different caste adult ant specificities of Polyrhachis vicina Roger (Hymenoptera, Formicidae) caused in estrogen-related receptor. Gen Comp Endocrinol 2018; 266:29-37. [PMID: 29746854 DOI: 10.1016/j.ygcen.2018.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 03/11/2018] [Accepted: 04/02/2018] [Indexed: 11/29/2022]
Abstract
The estrogen-related receptor (ERR) gene is a member of the nuclear receptor subfamily. Previous studies have indicated that ERR plays important roles in regulating insect growth and development. How ERR is associated with ant caste specificities remains unclear. In this study, we attempted to identify the role of ERR in the regulation of different adult caste specificities of Polyrhachis vicina Roger. Significant variations were detected in the ants including PvERR expressions, some physiological indexes and morphological traits including survival rate, body weight, body length, head width and abdominal appearance by different techniques. The results revealed that when PvERR expressions is up-regulated, boundaries of the abdominal segments were indistinct on the ventral side of the abdomen in males. Down-regulation of PvERR expressions caused abdominal swelling in males and a distended ventral abdomen in females and workers. Variation in PvERR expressions led to a remarkable decline in ant survival rates, particularly for males. These results indicated that different caste adults appeared to have different degrees of sensitivity in physiological response and morphological changes caused by variation in PvERR expressions. Thus, our data demonstrate that PvERR plays an important role in regulating the different adult caste specificities of P. vicina.
Collapse
Affiliation(s)
- Juan-Juan Zhang
- Institute of Zoology, Department of Life Science, Shaanxi Normal University, Xi'an, Shaanxi Province 710119, PR China
| | - Geng-Si Xi
- Institute of Zoology, Department of Life Science, Shaanxi Normal University, Xi'an, Shaanxi Province 710119, PR China.
| |
Collapse
|
161
|
He X, Zeng H, Chen JX. Emerging role of SIRT3 in endothelial metabolism, angiogenesis, and cardiovascular disease. J Cell Physiol 2018; 234:2252-2265. [PMID: 30132870 DOI: 10.1002/jcp.27200] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 07/12/2018] [Indexed: 12/11/2022]
Abstract
Sirtuin 3 (SIRT3) a mitochondrial enzyme that plays an important role in energy homeostasis, cardiac remodeling, and heart failure (HF). The expression of SIRT3 declines with advanced age, cardiovascular, and metabolic diseases. Accumulating evidence suggests that SIRT3 plays a critical role in protecting the heart from cardiac hypertrophy, cardiac dysfunction associated with HF, and in the protection of cardiac cells from stress-mediated cell death. Clinical studies have demonstrated that HF with preserved ejection fraction (HFpEF) in patients present with abnormalities in coronary microcirculation related to endothelial dysfunction and coronary microvascular rarefaction. Although SIRT3-mediated regulation of mitochondrial homeostasis and heart function has been intensively investigated, the effect of SIRT3 on endothelial cell (EC) glycolytic metabolism and microvascular function has not been well studied. ECs utilize glycolysis for generating ATP rather than oxidative phosphorylation to maintain their normal functions and promote angiogenesis and EC-cardiomyocyte interactions. Emerging evidence indicates that SIRT3 is involved in the regulation of endothelial metabolism and angiogenesis and thus affects the development of cardiovascular diseases associated with aging. This review will discuss the current knowledge of SIRT3 and its functional role on endothelial metabolism, cardiac function, and cardiovascular diseases.
Collapse
Affiliation(s)
- Xiaochen He
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Heng Zeng
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jian-Xiong Chen
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
162
|
Gan Z, Fu T, Kelly DP, Vega RB. Skeletal muscle mitochondrial remodeling in exercise and diseases. Cell Res 2018; 28:969-980. [PMID: 30108290 DOI: 10.1038/s41422-018-0078-7] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 07/27/2018] [Indexed: 12/18/2022] Open
Abstract
Skeletal muscle fitness and plasticity is an important determinant of human health and disease. Mitochondria are essential for maintaining skeletal muscle energy homeostasis by adaptive re-programming to meet the demands imposed by a myriad of physiologic or pathophysiological stresses. Skeletal muscle mitochondrial dysfunction has been implicated in the pathogenesis of many diseases, including muscular dystrophy, atrophy, type 2 diabetes, and aging-related sarcopenia. Notably, exercise counteracts the effects of many chronic diseases on skeletal muscle mitochondrial function. Recent studies have revealed a finely tuned regulatory network that orchestrates skeletal muscle mitochondrial biogenesis and function in response to exercise and in disease states. In addition, increasing evidence suggests that mitochondria also serve to "communicate" with the nucleus and mediate adaptive genomic re-programming. Here we review the current state of knowledge relevant to the dynamic remodeling of skeletal muscle mitochondria in response to exercise and in disease states.
Collapse
Affiliation(s)
- Zhenji Gan
- The State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, 210061, Nanjing, China.
| | - Tingting Fu
- The State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, 210061, Nanjing, China
| | - Daniel P Kelly
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Rick B Vega
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, FL, 32804, USA.
| |
Collapse
|
163
|
Cao LY, Zheng Z, Ren XM, Andersson PL, Guo LH. Structure-Dependent Activity of Polybrominated Diphenyl Ethers and Their Hydroxylated Metabolites on Estrogen Related Receptor γ: in Vitro and in Silico Study. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2018; 52:8894-8902. [PMID: 30005570 DOI: 10.1021/acs.est.8b02509] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Estrogen-related receptor γ (ERRγ) is an orphan nuclear receptor having functional cross-talk with classical estrogen receptors. Here, we investigated whether ERRγ is a potential target of polybrominated diphenyl ethers (PBDEs) and their hydroxylated metabolites (OH-PBDEs). By using a fluorescence competitive binding method established in our laboratory, the binding potencies of 30 PBDEs/OH-PBDEs with ERRγ were determined for the first time. All of the tested OH-PBDEs and some PBDEs bound to ERRγ with Kd values ranging from 0.13-13.61 μM. The OH-PBDEs showed much higher binding potency than their parent PBDEs. A quantitative structure-activity relationship (QSAR) model was developed to analyze the chemical binding potencies in relation to their structural and chemical characteristics. The QSAR model indicated that the molecular size, relative ratios of aromatic atoms, and hydrogen bond donors and acceptors were crucial factors for PBDEs/OH-PBDEs binding. By using a reporter gene assay, we found that most of the low-brominated PBDEs/OH-PBDEs exerted agonistic activity toward ERRγ, while high-brominated PBDEs/OH-PBDEs had no effect on the basal ERRγ activity. The docking results showed that the low-brominated PBDEs/OH-PBDEs tended to take an agonistic binding mode while the high-brominated ones tended to take an antagonistic binding mode. Overall, our results suggest ERRγ to be a potential novel target for PBDEs/OH-PBDEs.
Collapse
Affiliation(s)
- Lin-Ying Cao
- State Key Laboratory of Environmental Chemistry and Eco-toxicology, Research Center for Eco-environmental Sciences , Chinese Academy of Sciences , 18 Shuangqing Road , P.O. Box 2871, Beijing 100085 , China
- College of Resources and Environment , University of Chinese Academy of Sciences , Beijing 100039 , People's Republic of China
| | - Ziye Zheng
- Department of Chemistry , Umeå University , SE-901 87 Umeå , Sweden
| | - Xiao-Min Ren
- State Key Laboratory of Environmental Chemistry and Eco-toxicology, Research Center for Eco-environmental Sciences , Chinese Academy of Sciences , 18 Shuangqing Road , P.O. Box 2871, Beijing 100085 , China
| | | | - Liang-Hong Guo
- State Key Laboratory of Environmental Chemistry and Eco-toxicology, Research Center for Eco-environmental Sciences , Chinese Academy of Sciences , 18 Shuangqing Road , P.O. Box 2871, Beijing 100085 , China
- College of Resources and Environment , University of Chinese Academy of Sciences , Beijing 100039 , People's Republic of China
| |
Collapse
|
164
|
Kim H, Kim BK, Ohk B, Yoon H, Kang WY, Cho S, Seong SJ, Lee HW, Yoon Y. Estrogen‐related receptor γ negatively regulates osteoclastogenesis and protects against inflammatory bone loss. J Cell Physiol 2018; 234:1659-1670. [DOI: 10.1002/jcp.27035] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 06/25/2018] [Indexed: 01/01/2023]
Affiliation(s)
- Hyun‐Ju Kim
- Department of Molecular Medicine Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University Daegu Republic of Korea
- Clinical Trial Center, Kyungpook National University Hospital Daegu Republic of Korea
| | - Bo Kyung Kim
- Department of Molecular Medicine Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University Daegu Republic of Korea
- Clinical Trial Center, Kyungpook National University Hospital Daegu Republic of Korea
| | - Boram Ohk
- Department of Molecular Medicine Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University Daegu Republic of Korea
- Clinical Trial Center, Kyungpook National University Hospital Daegu Republic of Korea
| | - Hye‐Jin Yoon
- Clinical Trial Center, Kyungpook National University Hospital Daegu Republic of Korea
| | - Woo Youl Kang
- Clinical Trial Center, Kyungpook National University Hospital Daegu Republic of Korea
| | - Seungil Cho
- Clinical Trial Center, Kyungpook National University Hospital Daegu Republic of Korea
| | - Sook Jin Seong
- Clinical Trial Center, Kyungpook National University Hospital Daegu Republic of Korea
| | - Hae Won Lee
- Clinical Trial Center, Kyungpook National University Hospital Daegu Republic of Korea
| | - Young‐Ran Yoon
- Department of Molecular Medicine Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University Daegu Republic of Korea
- Clinical Trial Center, Kyungpook National University Hospital Daegu Republic of Korea
| |
Collapse
|
165
|
Senga S, Kawaguchi K, Kobayashi N, Ando A, Fujii H. A novel fatty acid-binding protein 5-estrogen-related receptor α signaling pathway promotes cell growth and energy metabolism in prostate cancer cells. Oncotarget 2018; 9:31753-31770. [PMID: 30167092 PMCID: PMC6114981 DOI: 10.18632/oncotarget.25878] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 07/21/2018] [Indexed: 01/16/2023] Open
Abstract
Epidermal or cutaneous fatty acid-binding protein is an intracellular lipid-binding protein, also known as FABP5, and its expression level is closely related to cancer cell proliferation and metastatic activities in various types of carcinoma. However, the molecular mechanisms of FABP5 in cancer cell proliferation and its other functions have remained unclear. In the present study, we have clearly revealed that FABP5 activated expression of metabolic genes (ATP5B, LCHAD, ACO2, FH and MFN2) via a novel signaling pathway in an ERRα (estrogen-related receptor α)-dependent manner in prostate cancer cell lines. To clarify the novel function of FABP5, we examined the activation mechanisms of the ERRα target genes via FABP5. A direct protein-protein interaction between FABP5 and ERRα was demonstrated by immunoprecipitation and GST pull-down assays. We have clearly revealed that FABP5 interacted directly with transcriptional complex containing ERRα and its co-activator PGC-1β to increase expression of the ERRα target genes. In addition, we have shown that FABP5 knockdown induced high energy stress leading to induction of apoptosis and cell cycle arrest via AMPK-FOXO3A signaling pathway in prostate cancer cells, suggesting that FABP5 plays an important role in cellular energy status directing metabolic adaptation to support cellular proliferation and survival.
Collapse
Affiliation(s)
- Shogo Senga
- Interdisciplinary Graduate School of Science and Technology, Shinshu University, Minami-minowa, Kami-ina, Nagano, 399-4598, Japan
| | - Koichiro Kawaguchi
- Interdisciplinary Graduate School of Science and Technology, Shinshu University, Minami-minowa, Kami-ina, Nagano, 399-4598, Japan
| | - Narumi Kobayashi
- Department of Biomedical Engineering, Graduate School of Science and Technology, Shinshu University, Minami-minowa, Kami-ina, Nagano, 399-4598, Japan
| | - Akira Ando
- Department of Biomedical Engineering, Graduate School of Science and Technology, Shinshu University, Minami-minowa, Kami-ina, Nagano, 399-4598, Japan
| | - Hiroshi Fujii
- Department of Interdisciplinary Genome Sciences and Cell Metabolism, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting-Edge Research, Shinshu University, Minami-minowa, Kami-ina, Nagano, 399-4598, Japan
| |
Collapse
|
166
|
Guan L, Chen Y, Wang Y, Zhang H, Fan S, Gao Y, Jiao T, Fu K, Sun J, Yu A, Huang M, Bi H. Effects of carnitine palmitoyltransferases on cancer cellular senescence. J Cell Physiol 2018; 234:1707-1719. [DOI: 10.1002/jcp.27042] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/25/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Lihuan Guan
- Institute of Clinical Pharmacology and Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat‐sen University Guangzhou China
| | - Yixin Chen
- Institute of Clinical Pharmacology and Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat‐sen University Guangzhou China
| | - Yongtao Wang
- Institute of Clinical Pharmacology and Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat‐sen University Guangzhou China
| | - Huizhen Zhang
- Institute of Clinical Pharmacology and Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat‐sen University Guangzhou China
| | - Shicheng Fan
- Institute of Clinical Pharmacology and Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat‐sen University Guangzhou China
| | - Yue Gao
- Institute of Clinical Pharmacology and Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat‐sen University Guangzhou China
| | - Tingying Jiao
- Institute of Clinical Pharmacology and Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat‐sen University Guangzhou China
| | - Kaili Fu
- Institute of Clinical Pharmacology and Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat‐sen University Guangzhou China
| | - Jiahong Sun
- Institute of Clinical Pharmacology and Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat‐sen University Guangzhou China
| | - Aiming Yu
- Department of Biochemistry & Molecular Medicine Comprehensive Cancer Center, UC Davis School of Medicine Sacramento California
| | - Min Huang
- Institute of Clinical Pharmacology and Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat‐sen University Guangzhou China
| | - Huichang Bi
- Institute of Clinical Pharmacology and Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat‐sen University Guangzhou China
| |
Collapse
|
167
|
Shen G, Wu J, Han C, Liu H, Xu Y, Zhang H, Lin Y, Xia Q. Oestrogen-related receptor reduces vitellogenin expression by crosstalk with the ecdysone receptor pathway in female silkworm, Bombyx mori. INSECT MOLECULAR BIOLOGY 2018; 27:454-463. [PMID: 29603466 DOI: 10.1111/imb.12385] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Oestrogen-related receptor (ERR) is involved in oestrogen receptor (ER) signalling pathways owing to its similarity to ER in terms of domain structure and co-activator and response elements. Although insects lack ER, they harbour an ERR gene that is thought to modulate metabolism and energy conversion via an unknown mechanism. The present study investigated the function of ERR in insects using female silkworm (Bombyx mori, Bm). We found that the expression of B. mori vitellogenin (BmVg) and B. mori ERR (BmERR) in the fat bodies of female silkworms at different stages of development exhibited alternating patterns, and RNA interference of BmERR in females induced BmVg transcription, resulting in an increase in egg weight relative to the control. Furthermore, BmERR was found to be involved in regulating the transcription of BmVg through an oestrogen-related receptor response element (ERRE) in the promoter of the BmVg gene, as demonstrated by electrophoretic mobility shift assay, cell transfection assay and chromatin immunoprecipitation. In summary, our results indicate that BmERR bound to the ERRE motif in the BmVg promoter reducing the expression of BmVg in the fat body of the female silkworm. To our surprise, the ERRE also showed the ability to bind the ecdysone receptor (BmEcR) and ultraspiracle complex. Thus, we surmise that ERR participates in steroid hormone signalling by engaging in crosstalk with the ER pathway in vertebrates and with the EcR pathway in insects.
Collapse
Affiliation(s)
- G Shen
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
- Chongqing Engineering and Technology Research Center for Novel Silk Materials, Chongqing, China
| | - J Wu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - C Han
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - H Liu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Y Xu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - H Zhang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Y Lin
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
- Chongqing Engineering and Technology Research Center for Novel Silk Materials, Chongqing, China
| | - Q Xia
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
- Chongqing Engineering and Technology Research Center for Novel Silk Materials, Chongqing, China
| |
Collapse
|
168
|
Zhang L, Carnesecchi J, Cerutti C, Tribollet V, Périan S, Forcet C, Wong J, Vanacker JM. LSD1-ERRα complex requires NRF1 to positively regulate transcription and cell invasion. Sci Rep 2018; 8:10041. [PMID: 29968728 PMCID: PMC6030097 DOI: 10.1038/s41598-018-27676-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 06/05/2018] [Indexed: 12/20/2022] Open
Abstract
Lysine-specific demethylase 1 (LSD1) exerts dual effects on histone H3, promoting transcriptional repression via Lys4 (H3K4) demethylation or transcriptional activation through Lys9 (H3K9) demethylation. These activities are often exerted at transcriptional start sites (TSSs) and depend on the type of enhancer-bound transcription factor (TFs) with which LSD1 interacts. In particular, the Estrogen-Receptor Related α (ERRα) TF interacts with LSD1 and switches its activities toward H3K9 demethylation, resulting in transcriptional activation of a set of common target genes. However, how are the LSD1-TF and, in particular LSD1-ERRα, complexes determined to act at TSSs is not understood. Here we show that promoter-bound nuclear respiratory factor 1 (NRF1), but not ERRα, is essential to LSD1 recruitment at the TSSs of positive LSD1-ERRα targets. In contrast to ERRα, NRF1 does not impact on the nature of LSD1 enzymatic activity. We propose a three factor model, in which the LSD1 histone modifier requires a TSS tethering factor (NRF1) as well as an activity inducer (ERRα) to transcriptionally activate common targets. The relevance of this common network is illustrated by functional data, showing that all three factors are required for cell invasion in an MMP1 (Matrix MetalloProtease 1)-dependent manner, the expression of which is regulated by NRF1/LSD1/ERRα-mediated H3K9me2 demethylation.
Collapse
Affiliation(s)
- Ling Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, F-69007, Lyon, France
| | - Julie Carnesecchi
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, F-69007, Lyon, France
- Department of Developmental Biology, Centre for Organismal Studies (COS), Heidelberg, University of Heidelberg, D-69120, Heidelberg, Germany
| | - Catherine Cerutti
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, F-69007, Lyon, France
| | - Violaine Tribollet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, F-69007, Lyon, France
| | - Séverine Périan
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, F-69007, Lyon, France
| | - Christelle Forcet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, F-69007, Lyon, France
| | - Jiemin Wong
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jean-Marc Vanacker
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, F-69007, Lyon, France.
| |
Collapse
|
169
|
Tang Y, Min Z, Xiang XJ, Liu L, Ma YL, Zhu BL, Song L, Tang J, Deng XJ, Yan Z, Chen GJ. Estrogen-related receptor alpha is involved in Alzheimer's disease-like pathology. Exp Neurol 2018; 305:89-96. [PMID: 29641978 DOI: 10.1016/j.expneurol.2018.04.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/28/2018] [Accepted: 04/06/2018] [Indexed: 12/12/2022]
Abstract
Estrogen-related receptor alpha (ERRα) is a transcriptional factor associated with mitochondrial biogenesis and energy metabolism. However, little is known about the role of ERRα in Alzheimer's disease (AD). Here, we report that in APP/PS1 mice, an animal model of AD, ERRα protein and mRNA were decreased in a region- and age-dependent manner. In HEK293 cells that stably express human full-length β-amyloid precursor protein (APP), overexpression of ERRα inhibited the amyloidogenic processing of APP and consequently reduced Aβ1-40/1-42 level. ERRα overexpression also attenuated Tau phosphorylation at selective sites, with the concomitant reduction of glycogen synthase kinase 3β (GSK3β) activity. Interestingly, alterations of APP processing and Tau phosphorylation induced by hydrogen peroxide were reversed by ERRα overexpression in HEK/APP cells. These results indicated that ERRα plays a functional role in AD pathology. By attenuating both amyloidogenesis and Tau phosphorylation, ERRα may serve as a potential therapeutic target for AD.
Collapse
Affiliation(s)
- Ying Tang
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, China
| | - Zhuo Min
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, China
| | - Xiao-Jiao Xiang
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, China
| | - Lu Liu
- Thirteenth people's Hospital of Chongqing, Chongqing 400016, China
| | - Yuan-Lin Ma
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, China
| | - Bing-Lin Zhu
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, China
| | - Li Song
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, China
| | - Jing Tang
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, China
| | - Xiao-Juan Deng
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, China
| | - Zhen Yan
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Guo-Jun Chen
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing 400016, China.
| |
Collapse
|
170
|
Singh BK, Sinha RA, Tripathi M, Mendoza A, Ohba K, Sy JAC, Xie SY, Zhou J, Ho JP, Chang CY, Wu Y, Giguère V, Bay BH, Vanacker JM, Ghosh S, Gauthier K, Hollenberg AN, McDonnell DP, Yen PM. Thyroid hormone receptor and ERRα coordinately regulate mitochondrial fission, mitophagy, biogenesis, and function. Sci Signal 2018; 11:eaam5855. [PMID: 29945885 DOI: 10.1126/scisignal.aam5855] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Thyroid hormone receptor β1 (THRB1) and estrogen-related receptor α (ESRRA; also known as ERRα) both play important roles in mitochondrial activity. To understand their potential interactions, we performed transcriptome and ChIP-seq analyses and found that many genes that were co-regulated by both THRB1 and ESRRA were involved in mitochondrial metabolic pathways. These included oxidative phosphorylation (OXPHOS), the tricarboxylic acid (TCA) cycle, and β-oxidation of fatty acids. TH increased ESRRA expression and activity in a THRB1-dependent manner through the induction of the transcriptional coactivator PPARGC1A (also known as PGC1α). Moreover, TH induced mitochondrial biogenesis, fission, and mitophagy in an ESRRA-dependent manner. TH also induced the expression of the autophagy-regulating kinase ULK1 through ESRRA, which then promoted DRP1-mediated mitochondrial fission. In addition, ULK1 activated the docking receptor protein FUNDC1 and its interaction with the autophagosomal protein MAP1LC3B-II to induce mitophagy. siRNA knockdown of ESRRA, ULK1, DRP1, or FUNDC1 inhibited TH-induced autophagic clearance of mitochondria through mitophagy and decreased OXPHOS. These findings show that many of the mitochondrial actions of TH are mediated through stimulation of ESRRA expression and activity, and co-regulation of mitochondrial turnover through the PPARGC1A-ESRRA-ULK1 pathway is mediated by their regulation of mitochondrial fission and mitophagy. Hormonal or pharmacologic induction of ESRRA expression or activity could improve mitochondrial quality in metabolic disorders.
Collapse
Affiliation(s)
- Brijesh K Singh
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore.
| | - Rohit A Sinha
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow 226014, Uttar Pradesh, India
| | - Madhulika Tripathi
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Arturo Mendoza
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Center for Life Sciences, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Kenji Ohba
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
- Department of Internal Medicine, Enshu Hospital, Hamamatsu, Shizuoka 430-0929, Japan
| | - Jann A C Sy
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Sherwin Y Xie
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Jin Zhou
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Jia Pei Ho
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Ching-Yi Chang
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, C238A Levine Science Research Center, Durham, NC 27710, USA
| | - Yajun Wu
- Department of Anatomy, Yong Loo Lin School of Medicine, NUS, Singapore
| | - Vincent Giguère
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Montreal, Québec H3A 1A3, Canada
| | - Boon-Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, NUS, Singapore
| | - Jean-Marc Vanacker
- 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
| | - Sujoy Ghosh
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Karine Gauthier
- 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
| | - Anthony N Hollenberg
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Center for Life Sciences, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Donald P McDonnell
- Department of Internal Medicine, Enshu Hospital, Hamamatsu, Shizuoka 430-0929, Japan
| | - Paul M Yen
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore.
| |
Collapse
|
171
|
Huang X, Wang X, Shang J, Zhaang Z, Cui B, Lin Y, Yang Y, Song Y, Yu S, Xia J. Estrogen related receptor alpha triggers the migration and invasion of endometrial cancer cells via up regulation of TGFB1. Cell Adh Migr 2018; 12:538-547. [PMID: 29781387 PMCID: PMC6363028 DOI: 10.1080/19336918.2018.1477901] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 05/11/2018] [Accepted: 05/14/2018] [Indexed: 12/22/2022] Open
Abstract
Estrogenic signals have been suggested to be important for the tumorigenesis and progression of endometrial cancer (EC) cells. Our present data showed that estrogen related receptor alpha (ERRα), while not ERRβ or ERRγ, was significantly elevated in EC cells and tissues when compared to their controls. Targeted inhibition of ERRα by siRNA or its inverse agonist XCT-790 can suppress the migration and invasion of EC cells. Both si-ERRα and XCT-790 decreased the expression of transforming growth factor-beta (TGF-β). ERRα can directly bind with the promoter of TGFB1 and then increase its transcription. Further, ERRα was involved in the positive self-feedback loop of TGF-β in EC cells. Targeted inhibition of ERRα/TGF-β can synergistically suppress the in vitro invasion of EC cells. Collectively, our data suggested that ERRα can trigger the cell migration and invasion via increasing the positive self-feedback regulation of TGF-β.
Collapse
Affiliation(s)
- Xiumin Huang
- Department of Gynecology and Obstetrics, Zhongshan Hospital of Xiamen University, Xiamen, China
| | - Xuelian Wang
- Department of Gynecology and Obstetrics, Zhongshan Hospital of Xiamen University, Xiamen, China
| | - Jing Shang
- Department of Gynecology and Obstetrics, Zhongshan Hospital of Xiamen University, Xiamen, China
| | - Zhiqin Zhaang
- Department of Gynecology and Obstetrics, Zhongshan Hospital of Xiamen University, Xiamen, China
| | - Binbin Cui
- Department of Gynecology and Obstetrics, Zhongshan Hospital of Xiamen University, Xiamen, China
| | - Yanzhen Lin
- Department of Gynecology and Obstetrics, Zhongshan Hospital of Xiamen University, Xiamen, China
| | - Ying Yang
- Department of Gynecology and Obstetrics, Zhongshan Hospital of Xiamen University, Xiamen, China
| | - Youyi Song
- Department of Gynecology and Obstetrics, Zhongshan Hospital of Xiamen University, Xiamen, China
| | - Shengnan Yu
- Department of Gynecology and Obstetrics, Zhongshan Hospital of Xiamen University, Xiamen, China
| | - Junjie Xia
- Organ Transplantation Institute, Xiamen University, No. 308, Xiang'an South Road, Xiamen City, Fujian Province, China
| |
Collapse
|
172
|
Madhu Krishna B, Chaudhary S, Mishra DR, Naik SK, Suklabaidya S, Adhya AK, Mishra SK. Estrogen receptor α dependent regulation of estrogen related receptor β and its role in cell cycle in breast cancer. BMC Cancer 2018; 18:607. [PMID: 29843638 PMCID: PMC5975398 DOI: 10.1186/s12885-018-4528-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 05/18/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Breast cancer (BC) is highly heterogeneous with ~ 60-70% of estrogen receptor positive BC patient's response to anti-hormone therapy. Estrogen receptors (ERs) play an important role in breast cancer progression and treatment. Estrogen related receptors (ERRs) are a group of nuclear receptors which belong to orphan nuclear receptors, which have sequence homology with ERs and share target genes. Here, we investigated the possible role and clinicopathological importance of ERRβ in breast cancer. METHODS Estrogen related receptor β (ERRβ) expression was examined using tissue microarray slides (TMA) of Breast Carcinoma patients with adjacent normal by immunohistochemistry and in breast cancer cell lines. In order to investigate whether ERRβ is a direct target of ERα, we investigated the expression of ERRβ in short hairpin ribonucleic acid knockdown of ERα breast cancer cells by western blot, qRT-PCR and RT-PCR. We further confirmed the binding of ERα by electrophoretic mobility shift assay (EMSA), chromatin immunoprecipitation (ChIP), Re-ChIP and luciferase assays. Fluorescence-activated cell sorting analysis (FACS) was performed to elucidate the role of ERRβ in cell cycle regulation. A Kaplan-Meier Survival analysis of GEO dataset was performed to correlate the expression of ERRβ with survival in breast cancer patients. RESULTS Tissue microarray (TMA) analysis showed that ERRβ is significantly down-regulated in breast carcinoma tissue samples compared to adjacent normal. ER + ve breast tumors and cell lines showed a significant expression of ERRβ compared to ER-ve tumors and cell lines. Estrogen treatment significantly induced the expression of ERRβ and it was ERα dependent. Mechanistic analyses indicate that ERα directly targets ERRβ through estrogen response element and ERRβ also mediates cell cycle regulation through p18, p21cip and cyclin D1 in breast cancer cells. Our results also showed the up-regulation of ERRβ promoter activity in ectopically co-expressed ERα and ERRβ breast cancer cell lines. Fluorescence-activated cell sorting analysis (FACS) showed increased G0/G1 phase cell population in ERRβ overexpressed MCF7 cells. Furthermore, ERRβ expression was inversely correlated with overall survival in breast cancer. Collectively our results suggest cell cycle and tumor suppressor role of ERRβ in breast cancer cells which provide a potential avenue to target ERRβ signaling pathway in breast cancer. CONCLUSION Our results indicate that ERRβ is a negative regulator of cell cycle and a possible tumor suppressor in breast cancer. ERRβ could be therapeutic target for the treatment of breast cancer.
Collapse
Affiliation(s)
- B Madhu Krishna
- Cancer Biology Lab, Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India
| | - Sanjib Chaudhary
- Cancer Biology Lab, Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India.,Present address: Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center (UNMC), Omaha, NE, USA
| | - Dipti Ranjan Mishra
- Department of Gene Function & Regulation, Institute of Life Sciences, Nalco square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India
| | - Sanoj K Naik
- Cancer Biology Lab, Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India
| | - S Suklabaidya
- Tumor Microenvironment and Animal Models Lab, Department of Translational Research and Technology Development, Institute of Life Sciences, Nalco square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India
| | - A K Adhya
- Department of Pathology, Kalinga Institute of Medical Sciences, Chandaka Industrial Estate, KIIT Rd, Patia, Bhubaneswar, Odisha, India
| | - Sandip K Mishra
- Cancer Biology Lab, Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India.
| |
Collapse
|
173
|
Zhu H, Huang L, He Z, Zou Z, Luo Y. Estrogen-related receptor γ regulates expression of 17β-hydroxysteroid dehydrogenase type 1 in fetal growth restriction. Placenta 2018; 67:38-44. [PMID: 29941172 DOI: 10.1016/j.placenta.2018.05.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 05/24/2018] [Accepted: 05/28/2018] [Indexed: 02/01/2023]
Abstract
INTRODUCTION Estrogen-related receptor γ (ERRγ) and 17β-hydroxysteroid dehydrogenase type 1 (HSD17B1) have important roles in cell invasion and in the proliferation of many types of cancer cells. However, it remains unknown whether ERRγ and HSD17B1 contribute to abnormal placental structure and dysfunction which characterize fetal growth restriction (FGR). Therefore, the aim of this study was to investigate the expression profiles of ERRγ and HSD17B1 in placenta tissues affected by FGR and to examine a possible molecular mechanism by which ERRγ is able to regulate HSD17B1 during development of FGR. METHODS Placenta tissues were collected from women affected by FGR (n = 28) and from women with appropriately gestational age (AGA) (n = 30). Relative mRNA and protein levels of ERRγ and HSD17B1 in both groups were assessed by quantitative real-time PCR, immunohistochemistry, and Western blot analyses. The effect of ERRγ on trophoblast function and its associated mechanistic details were studied in the trophoblast cell line, HTR-8/SVneo, which was transfected with small interfering RNA (siRNA) targeting ERRγ. RESULTS Both mRNA and protein levels of ERRγ and HSD17B1 were significantly lower in FGR placentae (P < 0.05). When ERRγ expression was knocked down in HTR-8/SVneo cells with siRNA, invasion and proliferation were inhibited. In addition, HSD17B1 expression was significantly decreased. In dual luciferase reporter assays, ERRγ stimulated transcription of HSD17B1 by targeting the ERRγ response element within its 5'-flanking promoter region. DISCUSSION Aberrant ERRγ expression may contribute to the pathogenesis of FGR by regulating the transcriptional activity of HSD17B1.
Collapse
Affiliation(s)
- Hui Zhu
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Linhuan Huang
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhiming He
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhiyong Zou
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yanmin Luo
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
174
|
A map of the PGC-1α- and NT-PGC-1α-regulated transcriptional network in brown adipose tissue. Sci Rep 2018; 8:7876. [PMID: 29777200 PMCID: PMC5959870 DOI: 10.1038/s41598-018-26244-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 05/04/2018] [Indexed: 11/19/2022] Open
Abstract
Transcriptional coactivator PGC-1α and its splice variant NT-PGC-1α play crucial roles in regulating cold-induced thermogenesis in brown adipose tissue (BAT). PGC-1α and NT-PGC-1α are highly induced by cold in BAT and subsequently bind to and coactivate many transcription factors to regulate expression of genes involved in mitochondrial biogenesis, fatty acid oxidation, respiration and thermogenesis. To identify the complete repertoire of PGC-1α and NT-PGC-1α target genes in BAT, we analyzed genome-wide DNA-binding and gene expression profiles. We find that PGC-1α-/NT-PGC-1α binding broadly associates with cold-mediated transcriptional activation. In addition to their known target genes in mitochondrial biogenesis and oxidative metabolism, PGC-1α and NT-PGC-1α additionally target a broad spectrum of genes involved in diverse biological pathways including ubiquitin-dependent protein catabolism, ribonucleoprotein complex biosynthesis, phospholipid biosynthesis, angiogenesis, glycogen metabolism, phosphorylation, and autophagy. Our findings expand the number of genes and biological pathways that may be regulated by PGC-1α and NT-PGC-1α and provide further insight into the transcriptional regulatory network in which PGC-1α and NT-PGC-1α coordinate a comprehensive transcriptional response in BAT in response to cold.
Collapse
|
175
|
B'chir W, Dufour CR, Ouellet C, Yan M, Tam IS, Andrzejewski S, Xia H, Nabata K, St-Pierre J, Giguère V. Divergent Role of Estrogen-Related Receptor α in Lipid- and Fasting-Induced Hepatic Steatosis in Mice. Endocrinology 2018; 159:2153-2164. [PMID: 29635284 DOI: 10.1210/en.2018-00115] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 03/28/2018] [Indexed: 01/23/2023]
Abstract
Given the increasing prevalence of obesity and the metabolic syndrome, identification of intrinsic molecular programs responsible for ensuring fuel homeostasis and preventing metabolic disease is needed. We investigated whether the orphan nuclear receptor estrogen-related receptor α (ERRα), a major regulator of energy metabolism, plays a role in lipid homeostasis and the development of nonalcoholic fatty liver disease (NAFLD) in response to chronic high-fat diet (HFD) consumption and long-term fasting. Systemic ablation of ERRα in mice demonstrated clear beneficial effects for loss of ERRα function in protection from HFD-provoked body weight gain manifested not only from a reduction in white adipose tissue stores but also from an impediment in intrahepatic lipid accumulation. The prevention of HFD-induced NAFLD in ERRα-null mice was underscored by transcriptional repression of de novo lipogenesis, which was upregulated in wild-type mice, a known contributing factor to lipid-stimulated hepatic steatosis. Surprisingly, given these findings, ERRα deficiency had no significant impact on the degree of fasting-induced NAFLD, involving the mobilization of adipocyte triglyceride (TG) stores into the liver. However, the presence of ERRα was essential for acute refeeding-mediated reversal of fasting-induced hepatic TG accretion, underpinned by impaired downregulation of adipose TG lipolysis and reduced hepatic mitochondrial oxidative activity. Taken together, the regulation of lipid handling by ERRα depended on the nutritional state, suggesting that negative modulation of ERRα activity could be envisaged to prevent lipid-induced NAFLD, whereas inducing its activity would be useful to treat and reverse the instilled disease.
Collapse
Affiliation(s)
- Wafa B'chir
- Goodman Cancer Research Centre, McGill University, Montréal, Quebec, Canada
| | - Catherine R Dufour
- Goodman Cancer Research Centre, McGill University, Montréal, Quebec, Canada
| | - Carlo Ouellet
- Goodman Cancer Research Centre, McGill University, Montréal, Quebec, Canada
| | - Ming Yan
- Goodman Cancer Research Centre, McGill University, Montréal, Quebec, Canada
| | - Ingrid S Tam
- Goodman Cancer Research Centre, McGill University, Montréal, Quebec, Canada
| | - Sylvia Andrzejewski
- Goodman Cancer Research Centre, McGill University, Montréal, Quebec, Canada
- Department of Biochemistry, McGill University, Montréal, Quebec, Canada
| | - Hui Xia
- Goodman Cancer Research Centre, McGill University, Montréal, Quebec, Canada
- Department of Biochemistry, McGill University, Montréal, Quebec, Canada
| | - Kylie Nabata
- Department of Biochemistry, McGill University, Montréal, Quebec, Canada
| | - Julie St-Pierre
- Goodman Cancer Research Centre, McGill University, Montréal, Quebec, Canada
- Department of Biochemistry, McGill University, Montréal, Quebec, Canada
| | - Vincent Giguère
- Goodman Cancer Research Centre, McGill University, Montréal, Quebec, Canada
- Department of Biochemistry, McGill University, Montréal, Quebec, Canada
- Department of Medicine, McGill University, Montréal, Quebec, Canada
- Department of Oncology, McGill University, Montréal, QC, Canada
| |
Collapse
|
176
|
Lin H, Doebelin C, Patouret R, Garcia-Ordonez RD, Chang MR, Dharmarajan V, Bayona CR, Cameron MD, Griffin PR, Kamenecka TM. Design, synthesis, and evaluation of simple phenol amides as ERRγ agonists. Bioorg Med Chem Lett 2018; 28:1313-1319. [PMID: 29548571 PMCID: PMC5893368 DOI: 10.1016/j.bmcl.2018.03.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/01/2018] [Accepted: 03/06/2018] [Indexed: 10/17/2022]
Abstract
Herein we report the design and synthesis of a series of simple phenol amide ERRγ agonists based on a hydrazone lead molecule. Our structure activity relationship studies in this series revealed the phenol portion of the molecule to be required for activity. Attempts to replace the hydrazone with more suitable chemotypes led to a simple amide as a viable alternative. Differential hydrogen-deuterium exchange experiments were used to help understand the structural basis for binding to ERRγ and aid in the development of more potent ligands.
Collapse
Affiliation(s)
- Hua Lin
- The Scripps Research Institute, Scripps Florida, Department of Molecular Medicine, 130 Scripps Way #A2A, Jupiter, FL 33458, USA
| | - Christelle Doebelin
- The Scripps Research Institute, Scripps Florida, Department of Molecular Medicine, 130 Scripps Way #A2A, Jupiter, FL 33458, USA
| | - Rémi Patouret
- The Scripps Research Institute, Scripps Florida, Department of Molecular Medicine, 130 Scripps Way #A2A, Jupiter, FL 33458, USA
| | - Ruben D Garcia-Ordonez
- The Scripps Research Institute, Scripps Florida, Department of Molecular Medicine, 130 Scripps Way #A2A, Jupiter, FL 33458, USA
| | - M R Chang
- The Scripps Research Institute, Scripps Florida, Department of Molecular Medicine, 130 Scripps Way #A2A, Jupiter, FL 33458, USA
| | - Venkatasubramanian Dharmarajan
- The Scripps Research Institute, Scripps Florida, Department of Molecular Medicine, 130 Scripps Way #A2A, Jupiter, FL 33458, USA
| | - Claudia Ruiz Bayona
- The Scripps Research Institute, Scripps Florida, Department of Molecular Medicine, 130 Scripps Way #A2A, Jupiter, FL 33458, USA
| | - Michael D Cameron
- The Scripps Research Institute, Scripps Florida, Department of Molecular Medicine, 130 Scripps Way #A2A, Jupiter, FL 33458, USA
| | - Patrick R Griffin
- The Scripps Research Institute, Scripps Florida, Department of Molecular Medicine, 130 Scripps Way #A2A, Jupiter, FL 33458, USA
| | - Theodore M Kamenecka
- The Scripps Research Institute, Scripps Florida, Department of Molecular Medicine, 130 Scripps Way #A2A, Jupiter, FL 33458, USA.
| |
Collapse
|
177
|
Emerging Roles of Estrogen-Related Receptors in the Brain: Potential Interactions with Estrogen Signaling. Int J Mol Sci 2018; 19:ijms19041091. [PMID: 29621182 PMCID: PMC5979530 DOI: 10.3390/ijms19041091] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 03/21/2018] [Accepted: 03/30/2018] [Indexed: 01/22/2023] Open
Abstract
In addition to their well-known role in the female reproductive system, estrogens can act in the brain to regulate a wide range of behaviors and physiological functions in both sexes. Over the past few decades, genetically modified animal models have greatly increased our knowledge about the roles of estrogen receptor (ER) signaling in the brain in behavioral and physiological regulations. However, less attention has been paid to the estrogen-related receptors (ERRs), the members of orphan nuclear receptors whose sequences are homologous to ERs but lack estrogen-binding ability. While endogenous ligands of ERRs remain to be determined, they seemingly share transcriptional targets with ERs and their expression can be directly regulated by ERs through the estrogen-response element embedded within the regulatory region of the genes encoding ERRs. Despite the broad expression of ERRs in the brain, we have just begun to understand the fundamental roles they play at molecular, cellular, and circuit levels. Here, we review recent research advancement in understanding the roles of ERs and ERRs in the brain, with particular emphasis on ERRs, and discuss possible cross-talk between ERs and ERRs in behavioral and physiological regulations.
Collapse
|
178
|
Wang T, Liu J, McDonald C, Lupino K, Zhai X, Wilkins BJ, Hakonarson H, Pei L. GDF15 is a heart-derived hormone that regulates body growth. EMBO Mol Med 2018; 9:1150-1164. [PMID: 28572090 PMCID: PMC5538424 DOI: 10.15252/emmm.201707604] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The endocrine system is crucial for maintaining whole-body homeostasis. Little is known regarding endocrine hormones secreted by the heart other than atrial/brain natriuretic peptides discovered over 30 years ago. Here, we identify growth differentiation factor 15 (GDF15) as a heart-derived hormone that regulates body growth. We show that pediatric heart disease induces GDF15 synthesis and secretion by cardiomyocytes. Circulating GDF15 in turn acts on the liver to inhibit growth hormone (GH) signaling and body growth. We demonstrate that blocking cardiomyocyte production of GDF15 normalizes circulating GDF15 level and restores liver GH signaling, establishing GDF15 as a bona fide heart-derived hormone that regulates pediatric body growth. Importantly, plasma GDF15 is further increased in children with concomitant heart disease and failure to thrive (FTT). Together these studies reveal a new endocrine mechanism by which the heart coordinates cardiac function and body growth. Our results also provide a potential mechanism for the well-established clinical observation that children with heart diseases often develop FTT.
Collapse
Affiliation(s)
- Ting Wang
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jian Liu
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Caitlin McDonald
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Katherine Lupino
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Xiandun Zhai
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Institute of Forensic Medicine, Henan University of Science and Technology, Luoyang Henan, China
| | - Benjamin J Wilkins
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pediatrics, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA, USA
| | - Liming Pei
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA .,Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
179
|
Abstract
The eukaryotic nuclear receptors (NRs) super-family of transcriptional factors include the estrogen-related receptors (ERRs) that have diverse roles in control of cellular energy balance, general metabolism, growth and development, immunity etc. Mouse knock-out models of specific ERR isoforms (ERRα, ERRβ and ERRγ) exhibit defects in several phenotypic traits. Newer findings indicate important roles of ERRs in the regulation of brown adipocyte tissue mitochondrial oxidative functions as well as metabolic control in association with hypoxia-inducible factors during cellular hypoxic state. Genes involved in cardiac metabolism is also influenced by ERRα and ERRγ in association with the co-activators PGC-1α and PGC-1β. On the other hand, ERRs have crucial involvement at the interface of metabolism and diseases such as cancer. Recent findings have implicated ERRα in the progression of tumor and malignancy of the breast, prostate, colon, endometrium etc. In this article, new insights into the regulatory role of ERRs in metabolism and cancer shall be reviewed.
Collapse
Affiliation(s)
- Harmit S Ranhotra
- a Department of Biochemistry , St. Edmund's College , Shillong , India
| |
Collapse
|
180
|
Baba T, Otake H, Inoue M, Sato T, Ishihara Y, Moon JY, Tsuchiya M, Miyabayashi K, Ogawa H, Shima Y, Wang L, Sato R, Yamazaki T, Suyama M, Nomura M, Choi MH, Ohkawa Y, Morohashi KI. Ad4BP/SF-1 regulates cholesterol synthesis to boost the production of steroids. Commun Biol 2018; 1:18. [PMID: 30271905 PMCID: PMC6123728 DOI: 10.1038/s42003-018-0020-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 02/14/2018] [Indexed: 11/09/2022] Open
Abstract
Housekeeping metabolic pathways such as glycolysis are active in all cell types. In addition, many types of cells are equipped with cell-specific metabolic pathways. To properly perform their functions, housekeeping and cell-specific metabolic pathways must function cooperatively. However, the regulatory mechanisms that couple metabolic pathways remain largely unknown. Recently, we showed that the steroidogenic cell-specific nuclear receptor Ad4BP/SF-1, which regulates steroidogenic genes, also regulates housekeeping glycolytic genes. Here, we identify cholesterogenic genes as the targets of Ad4BP/SF-1. Further, we reveal that Ad4BP/SF-1 regulates Hummr, a candidate mediator of cholesterol transport from endoplasmic reticula to mitochondria. Given that cholesterol is the starting material for steroidogenesis and is synthesized from acetyl-CoA, which partly originates from glucose, our results suggest that multiple biological processes involved in synthesizing steroid hormones are governed by Ad4BP/SF-1. To our knowledge, this study provides the first example where housekeeping and cell-specific metabolism are coordinated at the transcriptional level.
Collapse
Affiliation(s)
- Takashi Baba
- Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan.,Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hiroyuki Otake
- Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Miki Inoue
- Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tetsuya Sato
- Division of Bioinformatics, Medical Institute of Bioregulation, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yasuhiro Ishihara
- Laboratory of Molecular Brain Science, Graduate School of Integrated Arts and Sciences, Hiroshima University, Kagamiyama 1-7-1, Higashi-Hiroshima, 739-8521, Japan
| | - Ju-Yeon Moon
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, 02792, Korea
| | - Megumi Tsuchiya
- Nuclear Dynamics Group, Graduate School of Frontier Biosciences, Osaka University, Yamadaoka 1-3, Osaka, 565-0871, Japan
| | - Kanako Miyabayashi
- Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hidesato Ogawa
- Nuclear Dynamics Group, Graduate School of Frontier Biosciences, Osaka University, Yamadaoka 1-3, Osaka, 565-0871, Japan
| | - Yuichi Shima
- Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan.,Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan.,Department of Anatomy, Kawasaki Medical School, 577 Matsushima, Kurashiki, 701-0192, Japan
| | - Lixiang Wang
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Ryuichiro Sato
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo, Tokyo, 113-8657, Japan
| | - Takeshi Yamazaki
- Laboratory of Molecular Brain Science, Graduate School of Integrated Arts and Sciences, Hiroshima University, Kagamiyama 1-7-1, Higashi-Hiroshima, 739-8521, Japan
| | - Mikita Suyama
- Division of Bioinformatics, Medical Institute of Bioregulation, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masatoshi Nomura
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan.,Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Asahimachi 67, Kurume, 830-0011, Japan
| | - Man Ho Choi
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, 02792, Korea
| | - Yasuyuki Ohkawa
- Division of Transcritomics, Medical Institute of Bioregulation, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Ken-Ichirou Morohashi
- Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan. .,Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
181
|
Dickinson JM, D'Lugos AC, Naymik MA, Siniard AL, Wolfe AJ, Curtis DR, Huentelman MJ, Carroll CC. Transcriptome response of human skeletal muscle to divergent exercise stimuli. J Appl Physiol (1985) 2018. [PMID: 29543133 DOI: 10.1152/japplphysiol.00014.2018] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Aerobic (AE) and resistance exercise (RE) elicit unique adaptations in skeletal muscle that have distinct implications for health and performance. The purpose of this study was to identify the unique transcriptome response of skeletal muscle to acute AE and RE. In a counterbalanced, crossover design, six healthy, recreationally active young men (27 ± 3 yr) completed acute AE (40 min of cycling, ∼70% maximal HR) and RE [8 sets, 10 reps, ∼65% 1-repetition maximum (1RM)], separated by ∼1 wk. Muscle biopsies (vastus lateralis) were obtained before and at 1 and 4 h postexercise. Whole transcriptome RNA sequencing (HiSeq2500; Illumina) was performed on cDNA synthesized from skeletal muscle RNA. Sequencing data were analyzed using HTSeq, and differential gene expression was identified using DESeq2 [adjusted P value (FDR) <0.05, >1.5-fold change from preexercise]. RE resulted in a greater number of differentially expressed genes at 1 (67 vs. 48) and 4 h (523 vs. 221) compared with AE. We identified 348 genes that were differentially expressed only following RE, whereas 48 genes were differentially expressed only following AE. Gene clustering indicated that AE targeted functions related to zinc interaction, angiogenesis, and ubiquitination, whereas RE targeted functions related to transcription regulation, cytokine activity, cell adhesion, kinase activity, and the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. ESRRG and TNFSRF12A were identified as potential targets related to the specific response of skeletal muscle to AE and RE, respectively. These data describe the early postexercise transcriptome response of skeletal muscle to acute AE and RE and further highlight that different forms of exercise stimulate unique molecular activity in skeletal muscle. NEW & NOTEWORTHY Whole transcriptome RNA sequencing was used to determine the early postexercise transcriptome response of skeletal muscle to acute aerobic (AE) and resistance exercise (RE) in untrained individuals. Although a number of shared genes were stimulated following both AE and RE, several genes were uniquely responsive to each exercise mode. These findings support the need for future research focused to better identify the role of exercise mode as it relates to targeting specific cellular skeletal muscle abnormalities.
Collapse
Affiliation(s)
- Jared M Dickinson
- School of Nutrition and Health Promotion, Healthy Lifestyles Research Center, Exercise Science and Health Promotion, Arizona State University , Phoenix, Arizona
| | - Andrew C D'Lugos
- School of Nutrition and Health Promotion, Healthy Lifestyles Research Center, Exercise Science and Health Promotion, Arizona State University , Phoenix, Arizona
| | - Marcus A Naymik
- Translational Genomics Research Institute , Phoenix, Arizona
| | | | - Amanda J Wolfe
- Translational Genomics Research Institute , Phoenix, Arizona
| | | | | | - Chad C Carroll
- Midwestern University , Glendale, Arizona.,Department of Health and Kinesiology, Purdue University , West Lafayette, Indiana
| |
Collapse
|
182
|
Gu S, Cui D, Chen X, Xiong X, Zhao Y. PROTACs: An Emerging Targeting Technique for Protein Degradation in Drug Discovery. Bioessays 2018; 40:e1700247. [PMID: 29473971 DOI: 10.1002/bies.201700247] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 01/20/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Shanshan Gu
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine; 310003 Hangzhou China
- Institute of Translational Medicine, Zhejiang University School of Medicine; Hangzhou China
| | - Danrui Cui
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine; 310003 Hangzhou China
- Institute of Translational Medicine, Zhejiang University School of Medicine; Hangzhou China
| | - Xiaoyu Chen
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine; 310003 Hangzhou China
- Institute of Translational Medicine, Zhejiang University School of Medicine; Hangzhou China
| | - Xiufang Xiong
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine; 310003 Hangzhou China
- Institute of Translational Medicine, Zhejiang University School of Medicine; Hangzhou China
| | - Yongchao Zhao
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine; 310003 Hangzhou China
- Institute of Translational Medicine, Zhejiang University School of Medicine; Hangzhou China
| |
Collapse
|
183
|
Liang R, Lin Y, Yuan CL, Liu ZH, Li YQ, Luo XL, Ye JZ, Ye HH. High expression of estrogen-related receptor α is significantly associated with poor prognosis in patients with colorectal cancer. Oncol Lett 2018; 15:5933-5939. [PMID: 29552224 DOI: 10.3892/ol.2018.8011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 12/21/2017] [Indexed: 12/14/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common types of malignancy with high morbidity and mortality rates worldwide. This biologically heterogeneous disease results in diverse therapeutic responses, thus, novel prognostic biomarkers are required to improve CRC treatment. Estrogen-related receptor α (ERRα) is a nuclear orphan receptor, which is associated with estrogen receptor α. The present study aimed to investigate the expression of ERRα in patients with CRC, and explore the association between ERRα expression and clinicopathological factors, local recurrence and prognosis. In the present study, ERRα expression was detected in 15 fresh CRC tissues using quantitative real-time polymerase chain reaction (RT-qPCR) and in 128 paraffin-embedded CRC tissues using immunohistochemistry. The associations between ERRα expression and prognosis of CRC patients were evaluated by univariate, and multivariate (Cox proportional hazards model) analysis. RT-qPCR demonstrated that the mRNA expression of ERRα in CRC tissues was significantly higher compared with that in matched normal tissues. Immunohistochemistry revealed that ERRα high expression was detected in the nuclei of cancer cells from 39.1% (50/128) of CRC tissues. ERRα expression based on immunohistochemical staining was significantly associated with tumor differentiation, tumor invasion, lymph node status and Dukes stage (all P<0.05). Furthermore, patients with high ERRα expression were significantly associated with an increased risk of recurrence and poor prognosis, compared with patients with low ERRα expression. ERRα expression was identified as an independent prognostic factor for patients with CRC. In conclusion, ERRα serves important roles in the progression of CRC and is a potential prognostic factor for patients with CRC.
Collapse
Affiliation(s)
- Rong Liang
- First Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yan Lin
- First Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Chun-Ling Yuan
- First Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Zhi-Hui Liu
- First Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yong-Qiang Li
- First Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xiao-Ling Luo
- First Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Jia-Zhou Ye
- First Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Hai-Hong Ye
- Department of Hepatobilliary Surgery, Affiliated Minzu Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530001, P.R. China
| |
Collapse
|
184
|
Giguère V. Canonical signaling and nuclear activity of mTOR-a teamwork effort to regulate metabolism and cell growth. FEBS J 2018; 285:1572-1588. [PMID: 29337437 DOI: 10.1111/febs.14384] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 12/21/2017] [Accepted: 01/10/2018] [Indexed: 01/07/2023]
Abstract
Mechanistic (or mammalian) target of rapamycin (mTOR) is a kinase that regulates almost all functions related to cell growth and metabolism in response to extra- and intracellular stimuli, such as availability of nutrients, the presence of growth factors, or the energy status of the cell. As part of two distinct protein complexes, mTORC1 and mTORC2, the kinase has been shown to influence cell growth and proliferation by controlling ribosome biogenesis, mRNA translation, carbohydrate and lipid metabolism, protein degradation, autophagy as well as microtubule and actin dynamics. In addition to these well-characterized functions, mTOR can also influence gene transcription. While most studies focused on investigating how canonical mTOR signaling regulates the activity of transcription factors outside the nucleus, recent findings point to a more direct role for mTOR as a transcription factor operating on chromatin in the nucleus. In particular, recent genome-wide identification of mTOR targets on chromatin reveals that its activities in the nucleus and cytoplasm are functionally and biologically linked, thus uncovering a novel paradigm in mTOR function.
Collapse
Affiliation(s)
- Vincent Giguère
- Departments of Biochemistry, Medicine and Oncology, Faculty of Medicine, Goodman Cancer Research Centre, McGill University, Montréal, Canada
| |
Collapse
|
185
|
Zhang J, Ye ZW, Chen W, Manevich Y, Mehrotra S, Ball L, Janssen-Heininger Y, Tew KD, Townsend DM. S-Glutathionylation of estrogen receptor α affects dendritic cell function. J Biol Chem 2018; 293:4366-4380. [PMID: 29374060 DOI: 10.1074/jbc.m117.814327] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 01/18/2018] [Indexed: 12/27/2022] Open
Abstract
Glutathione S-transferase Pi (GSTP) is a thiolase that catalyzes the addition of glutathione (GSH) to receptive cysteines in target proteins, producing an S-glutathionylated residue. Accordingly, previous studies have reported that S-glutathionylation is constitutively decreased in cells from mice lacking GSTP (Gstp1/p2-/-). Here, we found that bone marrow-derived dendritic cells (BMDDCs) from Gstp1/p2-/- mice have proliferation rates that are greater than those in their WT counterparts (Gstp1/p2+/+). Moreover, Gstp1/p2-/- BMDDCs had increased reactive oxygen species (ROS) levels and decreased GSH:glutathione disulfide (GSSG) ratios. Estrogen receptor α (ERα) is linked to myeloproliferation and differentiation, and we observed that its steady-state levels are elevated in Gstp1/p2-/- BMDDCs, indicating a link between GSTP and ERα activities. BMDDCs differentiated by granulocyte-macrophage colony-stimulating factor had elevated ERα levels, which were more pronounced in Gstp1/p2-/- than WT mice. When stimulated with lipopolysaccharide for maturation, Gstp1/p2-/- BMDDCs exhibited augmented endocytosis, maturation rate, cytokine secretion, and T-cell activation; heightened glucose uptake and glycolysis; increased Akt signaling (in the mTOR pathway); and decreased AMPK-mediated phosphorylation of proteins. Of note, GSTP formed a complex with ERα, stimulating ERα S-glutathionylation at cysteines 221, 245, 417, and 447; altering ERα's binding affinity for estradiol; and reducing overall binding potential (receptor density and affinity) 3-fold. Moreover, in Gstp1/p2-/- BMDDCs, ERα S-glutathionylation was constitutively decreased. Taken together, these findings suggest that GSTP-mediated S-glutathionylation of ERα controls BMDDC differentiation and affects metabolic function in dendritic cells.
Collapse
Affiliation(s)
- Jie Zhang
- From the Departments of Cell and Molecular Pharmacology and Experimental Therapeutics
| | - Zhi-Wei Ye
- From the Departments of Cell and Molecular Pharmacology and Experimental Therapeutics
| | - Wei Chen
- Department of Infectious Disease, the Second Affiliated Hospital of Medical School of the Southeast University, 1-1 Zhongfu Road, Nanjing 210003, China, and
| | - Yefim Manevich
- From the Departments of Cell and Molecular Pharmacology and Experimental Therapeutics
| | - Shikhar Mehrotra
- Surgery, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Lauren Ball
- From the Departments of Cell and Molecular Pharmacology and Experimental Therapeutics
| | - Yvonne Janssen-Heininger
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont 05405
| | - Kenneth D Tew
- From the Departments of Cell and Molecular Pharmacology and Experimental Therapeutics,
| | | |
Collapse
|
186
|
Takada L, Barbero MMD, Oliveira HN, de Camargo GMF, Fernandes Júnior GA, Aspilcueta-Borquis RR, Souza FRP, Boligon AA, Melo TP, Regatieri IC, Feitosa FLB, Fonseca LFS, Magalhães AFB, Costa RB, Albuquerque LG. Genomic association for sexual precocity in beef heifers using pre-selection of genes and haplotype reconstruction. PLoS One 2018; 13:e0190197. [PMID: 29293544 PMCID: PMC5749767 DOI: 10.1371/journal.pone.0190197] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 12/08/2017] [Indexed: 12/23/2022] Open
Abstract
Reproductive traits are of the utmost importance for any livestock farming, but are difficult to measure and to interpret since they are influenced by various factors. The objective of this study was to detect associations between known polymorphisms in candidate genes related to sexual precocity in Nellore heifers, which could be used in breeding programs. Records of 1,689 precocious and non-precocious heifers from farms participating in the Conexão Delta G breeding program were analyzed. A subset of single nucleotide polymorphisms (SNP) located in the region of the candidate genes at a distance of up to 5 kb from the boundaries of each gene, were selected from the panel of 777,000 SNPs of the High-Density Bovine SNP BeadChip. Linear mixed models were used for statistical analysis of early heifer pregnancy, relating the trait with isolated SNPs or with haplotype groups. The model included the contemporary group (year and month of birth) as fixed effect and parent of the animal (sire effect) as random effect. The fastPHASE® and GenomeStudio® were used for reconstruction of the haplotypes and for analysis of linkage disequilibrium based on r2 statistics. A total of 125 candidate genes and 2,024 SNPs forming haplotypes were analyzed. Statistical analysis after Bonferroni correction showed that nine haplotypes exerted a significant effect (p<0.05) on sexual precocity. Four of these haplotypes were located in the Pregnancy-associated plasma protein-A2 gene (PAPP-A2), two in the Estrogen-related receptor gamma gene (ESRRG), and one each in the Pregnancy-associated plasma protein-A gene (PAPP-A), Kell blood group complex subunit-related family (XKR4) and mannose-binding lectin genes (MBL-1) genes. Although the present results indicate that the PAPP-A2, PAPP-A, XKR4, MBL-1 and ESRRG genes influence sexual precocity in Nellore heifers, further studies are needed to evaluate their possible use in breeding programs.
Collapse
Affiliation(s)
- Luciana Takada
- Departamento de Zootecnia-São Paulo State University-UNESP, Jaboticabal, São Paulo, Brazil
| | - Marina M D Barbero
- Departamento de Zootecnia-São Paulo State University-UNESP, Jaboticabal, São Paulo, Brazil
| | - Henrique N Oliveira
- Departamento de Zootecnia-São Paulo State University-UNESP, Jaboticabal, São Paulo, Brazil
| | | | | | | | - Fabio R P Souza
- Departamento de Zootecnia-São Paulo State University-UNESP, Jaboticabal, São Paulo, Brazil
| | - Arione A Boligon
- Departamento de Zootecnia-São Paulo State University-UNESP, Jaboticabal, São Paulo, Brazil
| | - Thaise P Melo
- Departamento de Zootecnia-São Paulo State University-UNESP, Jaboticabal, São Paulo, Brazil
| | - Inaê C Regatieri
- Departamento de Zootecnia-São Paulo State University-UNESP, Jaboticabal, São Paulo, Brazil
| | - Fabieli L B Feitosa
- Departamento de Zootecnia-São Paulo State University-UNESP, Jaboticabal, São Paulo, Brazil
| | - Larissa F S Fonseca
- Departamento de Zootecnia-São Paulo State University-UNESP, Jaboticabal, São Paulo, Brazil
| | - Ana F B Magalhães
- Departamento de Zootecnia-São Paulo State University-UNESP, Jaboticabal, São Paulo, Brazil
| | - Raphael B Costa
- Departamento de Zootecnia-São Paulo State University-UNESP, Jaboticabal, São Paulo, Brazil
| | - Lucia G Albuquerque
- Departamento de Zootecnia-São Paulo State University-UNESP, Jaboticabal, São Paulo, Brazil
| |
Collapse
|
187
|
Wang XX, Wang D, Luo Y, Myakala K, Dobrinskikh E, Rosenberg AZ, Levi J, Kopp JB, Field A, Hill A, Lucia S, Qiu L, Jiang T, Peng Y, Orlicky D, Garcia G, Herman-Edelstein M, D'Agati V, Henriksen K, Adorini L, Pruzanski M, Xie C, Krausz KW, Gonzalez FJ, Ranjit S, Dvornikov A, Gratton E, Levi M. FXR/TGR5 Dual Agonist Prevents Progression of Nephropathy in Diabetes and Obesity. J Am Soc Nephrol 2018; 29:118-137. [PMID: 29089371 PMCID: PMC5748904 DOI: 10.1681/asn.2017020222] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 08/23/2017] [Indexed: 12/12/2022] Open
Abstract
Bile acids are ligands for the nuclear hormone receptor farnesoid X receptor (FXR) and the G protein-coupled receptor TGR5. We have shown that FXR and TGR5 have renoprotective roles in diabetes- and obesity-related kidney disease. Here, we determined whether these effects are mediated through differential or synergistic signaling pathways. We administered the FXR/TGR5 dual agonist INT-767 to DBA/2J mice with streptozotocin-induced diabetes, db/db mice with type 2 diabetes, and C57BL/6J mice with high-fat diet-induced obesity. We also examined the individual effects of the selective FXR agonist obeticholic acid (OCA) and the TGR5 agonist INT-777 in diabetic mice. The FXR agonist OCA and the TGR5 agonist INT-777 modulated distinct renal signaling pathways involved in the pathogenesis and treatment of diabetic nephropathy. Treatment of diabetic DBA/2J and db/db mice with the dual FXR/TGR5 agonist INT-767 improved proteinuria and prevented podocyte injury, mesangial expansion, and tubulointerstitial fibrosis. INT-767 exerted coordinated effects on multiple pathways, including stimulation of a signaling cascade involving AMP-activated protein kinase, sirtuin 1, PGC-1α, sirtuin 3, estrogen-related receptor-α, and Nrf-1; inhibition of endoplasmic reticulum stress; and inhibition of enhanced renal fatty acid and cholesterol metabolism. Additionally, in mice with diet-induced obesity, INT-767 prevented mitochondrial dysfunction and oxidative stress determined by fluorescence lifetime imaging of NADH and kidney fibrosis determined by second harmonic imaging microscopy. These results identify the renal signaling pathways regulated by FXR and TGR5, which may be promising targets for the treatment of nephropathy in diabetes and obesity.
Collapse
MESH Headings
- Albuminuria/etiology
- Animals
- Bile Acids and Salts/pharmacology
- Chenodeoxycholic Acid/analogs & derivatives
- Chenodeoxycholic Acid/pharmacology
- Cholesterol/metabolism
- Cholic Acids/pharmacology
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 2/metabolism
- Diabetic Nephropathies/complications
- Diabetic Nephropathies/metabolism
- Diabetic Nephropathies/pathology
- Diabetic Nephropathies/prevention & control
- Disease Progression
- Endoplasmic Reticulum Stress
- Fibrosis
- Glomerular Mesangium/pathology
- Humans
- Kidney Tubules/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mitochondria/metabolism
- Obesity/complications
- Obesity/metabolism
- Oxidative Stress
- Podocytes/pathology
- RNA, Messenger/metabolism
- Receptors, Cytoplasmic and Nuclear/agonists
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, G-Protein-Coupled/agonists
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Signal Transduction/drug effects
- Triglycerides/metabolism
Collapse
Affiliation(s)
- Xiaoxin X Wang
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| | - Dong Wang
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| | - Yuhuan Luo
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| | - Komuraiah Myakala
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| | - Evgenia Dobrinskikh
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| | - Avi Z Rosenberg
- National Institute of Diabetes and Digestive and Kidney Diseases and
- Division of Pathology and
| | - Jonathan Levi
- National Institute of Diabetes and Digestive and Kidney Diseases and
| | - Jeffrey B Kopp
- National Institute of Diabetes and Digestive and Kidney Diseases and
| | - Amanda Field
- Center for Genetic Medicine Research, Children's Research Institute, Children's National Medical Center and the George Washington University School of Medicine and Health Sciences, Washington, DC
- Department of Nephrology and Hypertension, Rabin Medical Center, Tel Aviv, Israel
| | - Ashley Hill
- Center for Genetic Medicine Research, Children's Research Institute, Children's National Medical Center and the George Washington University School of Medicine and Health Sciences, Washington, DC
- Department of Nephrology and Hypertension, Rabin Medical Center, Tel Aviv, Israel
| | - Scott Lucia
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| | - Liru Qiu
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| | - Tao Jiang
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| | - Yingqiong Peng
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| | - David Orlicky
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| | - Gabriel Garcia
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| | - Michal Herman-Edelstein
- Department of Nephrology and Hypertension, Rabin Medical Center, Tel Aviv, Israel
- Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Vivette D'Agati
- Department of Pathology, Columbia University College of Physicians and Surgeons, New York, New York
| | - Kammi Henriksen
- Department of Pathology, University of Chicago, Chicago, Illinois
| | | | - Mark Pruzanski
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Cen Xie
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Kristopher W Krausz
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Frank J Gonzalez
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Suman Ranjit
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, California
| | - Alexander Dvornikov
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, California
| | - Enrico Gratton
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, California
| | - Moshe Levi
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| |
Collapse
|
188
|
Casaburi I, Chimento A, De Luca A, Nocito M, Sculco S, Avena P, Trotta F, Rago V, Sirianni R, Pezzi V. Cholesterol as an Endogenous ERRα Agonist: A New Perspective to Cancer Treatment. Front Endocrinol (Lausanne) 2018; 9:525. [PMID: 30254608 PMCID: PMC6141749 DOI: 10.3389/fendo.2018.00525] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/21/2018] [Indexed: 01/01/2023] Open
Abstract
The estrogen-related receptors (ERRs) are important members of nuclear receptors which contain three isoforms (α, β, and γ). ERRα is the best-characterized isoform expressed mainly in high-energy demanding tissues where it preferentially works in association with the peroxisome proliferator-activated receptor-γ co-activator 1α (PGC-1α) and PGC-1β. ERRα together with its cofactors modulates cellular metabolism, supports the growth of rapidly dividing cells, directs metabolic programs required for cell differentiation and maintains cellular energy homeostasis in differentiated cells. In cancer cells, the functional association between ERRα and PGC-1s is further influenced by oncogenic signals and induces metabolic programs favoring cell growth and proliferation as well as tumor progression. Recently, cholesterol has been identified as a natural ERRα ligand using a combined biochemical strategy. This new finding highlighted some important physiological aspects related to the use of cholesterol-lowering drugs such as statins and bisphosphonates. Even more meaningful is the link between increased cholesterol levels and certain cancer phenotypes characterized by an overexpressed ERRα such as mammary, prostatic, and colorectal cancers, where the metabolic adaptation affects many cancer processes. Moreover, high-energy demanding cancer-related processes are strictly related to the cross-talk between tumor cells and some key players of tumor microenvironment, such as tumor-associated macrophage that fuels cancer progression. Some evidence suggests that high cholesterol content and ERRα activity favor the inflammatory environment by the production of different cytokines. In this review, starting from the most recent observations on the physiological role of the new signaling activated by the natural ligand of ERRα, we propose a new hypothesis on the suitability to control cholesterol levels as a chance in modulating ERRα activity in those tumors in which its expression and activity are increased.
Collapse
|
189
|
Li W, Sivakumar R, Titov AA, Choi SC, Morel L. Metabolic Factors that Contribute to Lupus Pathogenesis. Crit Rev Immunol 2017; 36:75-98. [PMID: 27480903 DOI: 10.1615/critrevimmunol.2016017164] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease in which organ damage is mediated by pathogenic autoantibodies directed against nucleic acids and protein complexes. Studies in SLE patients and in mouse models of lupus have implicated virtually every cell type in the immune system in the induction or amplification of the autoimmune response as well as the promotion of an inflammatory environment that aggravates tissue injury. Here, we review the contribution of CD4+ T cells, B cells, and myeloid cells to lupus pathogenesis and then discuss alterations in the metabolism of these cells that may contribute to disease, given the recent advances in the field of immunometabolism.
Collapse
Affiliation(s)
- Wei Li
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610; Department of Biochemistry and Molecular Biology, Gene Engineering and Biotechnology, Beijing Key Laboratory, Beijing Normal University, Beijing 100875, People's Republic of China
| | - Ramya Sivakumar
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Anton A Titov
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Seung-Chul Choi
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Laurence Morel
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| |
Collapse
|
190
|
Velloso FJ, Bianco AFR, Farias JO, Torres NEC, Ferruzo PYM, Anschau V, Jesus-Ferreira HC, Chang THT, Sogayar MC, Zerbini LF, Correa RG. The crossroads of breast cancer progression: insights into the modulation of major signaling pathways. Onco Targets Ther 2017; 10:5491-5524. [PMID: 29200866 PMCID: PMC5701508 DOI: 10.2147/ott.s142154] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer is the disease with highest public health impact in developed countries. Particularly, breast cancer has the highest incidence in women worldwide and the fifth highest mortality in the globe, imposing a significant social and economic burden to society. The disease has a complex heterogeneous etiology, being associated with several risk factors that range from lifestyle to age and family history. Breast cancer is usually classified according to the site of tumor occurrence and gene expression profiling. Although mutations in a few key genes, such as BRCA1 and BRCA2, are associated with high breast cancer risk, the large majority of breast cancer cases are related to mutated genes of low penetrance, which are frequently altered in the whole population. Therefore, understanding the molecular basis of breast cancer, including the several deregulated genes and related pathways linked to this pathology, is essential to ensure advances in early tumor detection and prevention. In this review, we outline key cellular pathways whose deregulation has been associated with breast cancer, leading to alterations in cell proliferation, apoptosis, and the delicate hormonal balance of breast tissue cells. Therefore, here we describe some potential breast cancer-related nodes and signaling concepts linked to the disease, which can be positively translated into novel therapeutic approaches and predictive biomarkers.
Collapse
Affiliation(s)
| | | | | | | | | | - Valesca Anschau
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | | | - Ted Hung-Tse Chang
- Cancer Genomics Group, International Center for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa
| | | | - Luiz F Zerbini
- Cancer Genomics Group, International Center for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa
| | - Ricardo G Correa
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|
191
|
Andrzejewski S, Klimcakova E, Johnson RM, Tabariès S, Annis MG, McGuirk S, Northey JJ, Chénard V, Sriram U, Papadopoli DJ, Siegel PM, St-Pierre J. PGC-1α Promotes Breast Cancer Metastasis and Confers Bioenergetic Flexibility against Metabolic Drugs. Cell Metab 2017; 26:778-787.e5. [PMID: 28988825 DOI: 10.1016/j.cmet.2017.09.006] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 05/31/2017] [Accepted: 09/08/2017] [Indexed: 02/07/2023]
Abstract
Metabolic adaptations play a key role in fueling tumor growth. However, less is known regarding the metabolic changes that promote cancer progression to metastatic disease. Herein, we reveal that breast cancer cells that preferentially metastasize to the lung or bone display relatively high expression of PGC-1α compared with those that metastasize to the liver. PGC-1α promotes breast cancer cell migration and invasion in vitro and augments lung metastasis in vivo. Pro-metastatic capabilities of PGC-1α are linked to enhanced global bioenergetic capacity, facilitating the ability to cope with bioenergetic disruptors like biguanides. Indeed, biguanides fail to mitigate the PGC-1α-dependent lung metastatic phenotype and PGC-1α confers resistance to stepwise increases in metformin concentration. Overall, our results reveal that PGC-1α stimulates bioenergetic potential, which promotes breast cancer metastasis and facilitates adaptation to metabolic drugs.
Collapse
Affiliation(s)
- Sylvia Andrzejewski
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada; Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Eva Klimcakova
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Radia M Johnson
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Sébastien Tabariès
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Matthew G Annis
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Shawn McGuirk
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada; Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Jason J Northey
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Valérie Chénard
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada; Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Urshila Sriram
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada; Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 1Y6, Canada
| | - David J Papadopoli
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada; Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Peter M Siegel
- Department of Medicine, McGill University, Montreal, QC H3G 1Y6, Canada; Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 1Y6, Canada.
| | - Julie St-Pierre
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada; Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 1Y6, Canada.
| |
Collapse
|
192
|
Yan X, Zhang G, Bie F, Lv Y, Ma Y, Ma M, Wang Y, Hao X, Yuan N, Jiang X. Eugenol inhibits oxidative phosphorylation and fatty acid oxidation via downregulation of c-Myc/PGC-1β/ERRα signaling pathway in MCF10A-ras cells. Sci Rep 2017; 7:12920. [PMID: 29018241 PMCID: PMC5634997 DOI: 10.1038/s41598-017-13505-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 09/25/2017] [Indexed: 12/22/2022] Open
Abstract
Alteration in cellular energy metabolism plays a critical role in the development and progression of cancer. Targeting metabolic pathways for cancer treatment has been investigated as potential preventive or therapeutic methods. Eugenol (Eu), a major volatile constituent of clove essential oil mainly obtained from Syzygium, has been reported as a potential chemopreventive drug. However, the mechanism by which Eu regulates cellular energy metabolism is still not well defined. This study was designed to determine the effect of Eu on cellular energy metabolism during early cancer progression employing untransformed and H-ras oncogene transfected MCF10A human breast epithelial cells. Eu showed dose-dependent selective cytotoxicity toward MCF10A-ras cells but exhibited no apparent cytotoxicity in MCF10A cells. Treatment with Eu also significantly reduced intracellular ATP levels in MCF10A-ras cells but not in MCF10A cells. This effect was mediated mainly through inhibiting oxidative phosphorylation (OXPHOS) complexs and the expression of fatty acid oxidation (FAO) proteins including PPARα, MCAD and CPT1C by downregulating c-Myc/PGC-1β/ERRα pathway and decreasing oxidative stress in MCF10A-ras cells. These results indicate a novel mechanism involving the regulation of cellular energy metabolism by which Eu may prevent breast cancer progression.
Collapse
Affiliation(s)
- Xianxin Yan
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Guijuan Zhang
- The School Outpatient Department, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Fengjie Bie
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Yanhong Lv
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Yi Ma
- Bio-engineering institute of Jinan University, Guangzhou, China
| | - Min Ma
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China.
| | - Yurong Wang
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xiaoqian Hao
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Naijun Yuan
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xuefeng Jiang
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
193
|
Abstract
Mitochondria are essential organelles for many aspects of cellular homeostasis, including energy harvesting through oxidative phosphorylation. Alterations of mitochondrial function not only impact on cellular metabolism but also critically influence whole-body metabolism, health, and life span. Diseases defined by mitochondrial dysfunction have expanded from rare monogenic disorders in a strict sense to now also include many common polygenic diseases, including metabolic, cardiovascular, neurodegenerative, and neuromuscular diseases. This has led to an intensive search for new therapeutic and preventive strategies aimed at invigorating mitochondrial function by exploiting key components of mitochondrial biogenesis, redox metabolism, dynamics, mitophagy, and the mitochondrial unfolded protein response. As such, new findings linking mitochondrial function to the progression or outcome of this ever-increasing list of diseases has stimulated the discovery and development of the first true mitochondrial drugs, which are now entering the clinic and are discussed in this review.
Collapse
Affiliation(s)
- Vincenzo Sorrentino
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland;
| | - Keir J Menzies
- Interdisciplinary School of Health Sciences, University of Ottawa Brain and Mind Research Institute and Centre for Neuromuscular Disease, Ottawa K1H 8M5, Canada;
| | - Johan Auwerx
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland;
| |
Collapse
|
194
|
Guh YJ, Hwang PP. Insights into molecular and cellular mechanisms of hormonal actions on fish ion regulation derived from the zebrafish model. Gen Comp Endocrinol 2017; 251:12-20. [PMID: 27554927 DOI: 10.1016/j.ygcen.2016.08.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 08/15/2016] [Accepted: 08/18/2016] [Indexed: 02/07/2023]
Abstract
Fish have sophisticated mechanisms of ionic and acid-base regulation for maintaining body fluid homeostasis. Many hormones have been proposed to control the ionic and acid-base regulation mechanisms in fishes; however, lots of the proposed actions lack convincing cellular/molecular evidence. With the advantages of available genetic databases and molecular manipulation techniques, zebrafish has become an emerging model for research into ion transport physiology and functional regulation. Different types of ionocytes were found to transport ions through various sets of ion transporters, and the molecular mechanisms of ionocyte proliferation and differentiation have also been dissected, providing a competent platform with which to precisely study the ion transport pathways and ionocytes targeted by hormones, including isotocin, prolactin, cortisol, stanniocalcin-1, calcitonin, endothelin-1, vitamin D, parathyroid hormone 1, catecholamines, the renin-angiotensin-system, estrogen-related receptor α, and calcitonin gene-related peptide, which have been demonstrated to positively or negatively regulate ion transport through specific receptors at different molecular levels (transcriptional, translational, or posttranslational) or at different developmental stages of ionocytes (proliferation or differentiation). The knowledge obtained in zebrafish not only enhances our understanding of the hormonal control of fish ion regulation, but also informs studies on other animal species, thereby providing insights into related fields.
Collapse
Affiliation(s)
- Ying-Jey Guh
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Pung-Pung Hwang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan.
| |
Collapse
|
195
|
Ning Y, Chen H, Du Y, Ling H, Zhang L, Chen L, Qi H, Shi X, Li Q. A novel compound LingH2-10 inhibits the growth of triple negative breast cancer cells in vitro and in vivo as a selective inverse agonist of estrogen-related receptor α. Biomed Pharmacother 2017; 93:913-922. [PMID: 28715872 DOI: 10.1016/j.biopha.2017.07.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 07/04/2017] [Accepted: 07/05/2017] [Indexed: 02/07/2023] Open
Abstract
Unlike other breast cancer subtypes, targeted therapies for triple negative breast cancer (TNBC) have yet to progress past clinical trial stage to approval. Accumulating evidences demonstrated that expression of estrogen-related receptor alpha (ERRα) indicated worse prognosis and correlated with poor outcome in breast cancers including TNBC. Therefore, ERRα modulators/regulators may be potential in the therapeutic treatment of breast cancers. In the current study, we presented a novel compound LingH2-10 that bound to ERRα, as identified using a time-resolved fluorescence resonance energy transfer assay (TR-FRET) with the IC50 value of 0.64±0.12μM. Further, functional activity was determined by transient transfection luciferase reporter assay in order to validate the utility of the binding affinity in a cellular context. LingH2-10 showed selective inhibition on ERRα transcriptional activity with the IC50 value of 0.58±0.09μM in cell-based luciferase reporter assay. Moreover, representative in vitro results showed that LingH2-10 suppressed the proliferation of various human cancer cells, and inhibited the migration of triple negative breast cancer cell MDA-MB-231. In addition, our results demonstrated that well known ERRα target genes such as PDK4, Osteopontin and pS2, were all significantly down modulated by LingH2-10. In vivo experiments showed that LingH2-10 (30mg/kg, every other day) observably suppressed the growth of MDA-MB-231 xenograft tumors by 42.02% compared to untreated xenograft tumors. Taken together, all these data suggested that LingH2-10, as a selective inverse agonist of ERRα, was a lead compound of anti-cancer agents for treating TNBC patients.
Collapse
Affiliation(s)
- Yang Ning
- School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology, 3501 Daxue Road, Jinan 250353, China
| | - Haifei Chen
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai 201907, China
| | - Yongli Du
- School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology, 3501 Daxue Road, Jinan 250353, China.
| | - Hao Ling
- School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology, 3501 Daxue Road, Jinan 250353, China
| | - Liudi Zhang
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai 201907, China
| | - Lu Chen
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai 201907, China
| | - Huijie Qi
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai 201907, China
| | - Xiaojin Shi
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai 201907, China; Clinical Pharmacy Laboratory, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qunyi Li
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai 201907, China; Clinical Pharmacy Laboratory, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
196
|
Emmett MJ, Lim HW, Jager J, Richter HJ, Adlanmerini M, Peed LC, Briggs ER, Steger DJ, Ma T, Sims CA, Baur JA, Pei L, Won KJ, Seale P, Gerhart-Hines Z, Lazar MA. Histone deacetylase 3 prepares brown adipose tissue for acute thermogenic challenge. Nature 2017; 546:544-548. [PMID: 28614293 PMCID: PMC5826652 DOI: 10.1038/nature22819] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 05/03/2017] [Indexed: 12/18/2022]
Abstract
Brown adipose tissue is a thermogenic organ that dissipates chemical energy as heat to protect animals against hypothermia and to counteract metabolic disease. However, the transcriptional mechanisms that determine the thermogenic capacity of brown adipose tissue before environmental cold are unknown. Here we show that histone deacetylase 3 (HDAC3) is required to activate brown adipose tissue enhancers to ensure thermogenic aptitude. Mice with brown adipose tissue-specific genetic ablation of HDAC3 become severely hypothermic and succumb to acute cold exposure. Uncoupling protein 1 (UCP1) is nearly absent in brown adipose tissue lacking HDAC3, and there is also marked downregulation of mitochondrial oxidative phosphorylation genes resulting in diminished mitochondrial respiration. Remarkably, although HDAC3 acts canonically as a transcriptional corepressor, it functions as a coactivator of oestrogen-related receptor α (ERRα) in brown adipose tissue. HDAC3 coactivation of ERRα is mediated by deacetylation of PGC-1α and is required for the transcription of Ucp1, Ppargc1a (encoding PGC-1α), and oxidative phosphorylation genes. Importantly, HDAC3 promotes the basal transcription of these genes independently of adrenergic stimulation. Thus, HDAC3 uniquely primes Ucp1 and the thermogenic transcriptional program to maintain a critical capacity for thermogenesis in brown adipose tissue that can be rapidly engaged upon exposure to dangerously cold temperature.
Collapse
Affiliation(s)
- Matthew J. Emmett
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hee-Woong Lim
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jennifer Jager
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hannah J. Richter
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marine Adlanmerini
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lindsey C. Peed
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Erika R. Briggs
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David J. Steger
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tao Ma
- Section for Metabolic Receptology at the Novo Nordisk Foundation Center for Basic Metabolic Research, and Institute for Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, 2200, DK
| | - Carrie A. Sims
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- The Trauma Center at Penn, Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104
| | - Joseph A. Baur
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Physiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Liming Pei
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Mitochondrial and Epigenomic Medicine, Children’s Hospital of Philadelphia, and Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kyoung-Jae Won
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Patrick Seale
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zachary Gerhart-Hines
- Section for Metabolic Receptology at the Novo Nordisk Foundation Center for Basic Metabolic Research, and Institute for Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, 2200, DK
| | - Mitchell A. Lazar
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
197
|
He X, Ma S, Tian Y, Wei C, Zhu Y, Li F, Zhang P, Wang P, Zhang Y, Zhong H. ERRα negatively regulates type I interferon induction by inhibiting TBK1-IRF3 interaction. PLoS Pathog 2017; 13:e1006347. [PMID: 28591144 PMCID: PMC5476288 DOI: 10.1371/journal.ppat.1006347] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 06/19/2017] [Accepted: 04/11/2017] [Indexed: 12/21/2022] Open
Abstract
Estrogen-related receptor α (ERRα) is a member of the nuclear receptor superfamily controlling energy homeostasis; however, its precise role in regulating antiviral innate immunity remains to be clarified. Here, we showed that ERRα deficiency conferred resistance to viral infection both in vivo and in vitro. Mechanistically, ERRα inhibited the production of type-I interferon (IFN-I) and the expression of multiple interferon-stimulated genes (ISGs). Furthermore, we found that viral infection induced TBK1-dependent ERRα stabilization, which in turn associated with TBK1 and IRF3 to impede the formation of TBK1-IRF3, IRF3 phosphorylation, IRF3 dimerization, and the DNA binding affinity of IRF3. The effect of ERRα on IFN-I production was independent of its transcriptional activity and PCG-1α. Notably, ERRα chemical inhibitor XCT790 has broad antiviral potency. This work not only identifies ERRα as a critical negative regulator of antiviral signaling, but also provides a potential target for future antiviral therapy.
Collapse
Affiliation(s)
- Xiang He
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, P.R. China
| | - Shengli Ma
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, P.R. China
| | - Yinyin Tian
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, P.R. China
- Institute of Healthy Science, Anhui University, Hefei, Anhui, P.R. China
| | - Congwen Wei
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, P.R. China
| | - Yongjie Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, P.R. China
- Institute of Healthy Science, Anhui University, Hefei, Anhui, P.R. China
| | - Feng Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, P.R. China
- Institute of Healthy Science, Anhui University, Hefei, Anhui, P.R. China
| | - Pingping Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, P.R. China
| | - Penghao Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, P.R. China
| | - Yanhong Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, P.R. China
| | - Hui Zhong
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, P.R. China
| |
Collapse
|
198
|
Ye Q, Chen C, Si E, Cai Y, Wang J, Huang W, Li D, Wang Y, Chen X. Mitochondrial Effects of PGC-1alpha Silencing in MPP + Treated Human SH-SY5Y Neuroblastoma Cells. Front Mol Neurosci 2017; 10:164. [PMID: 28611589 PMCID: PMC5447087 DOI: 10.3389/fnmol.2017.00164] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/11/2017] [Indexed: 12/30/2022] Open
Abstract
The dopaminergic neuron degeneration and loss that occurs in Parkinson’s disease (PD) has been tightly linked to mitochondrial dysfunction. Although the aged-related cause of the mitochondrial defect observed in PD patients remains unclear, nuclear genes are of potential importance to mitochondrial function. Human peroxisome proliferator-activated receptor γ coactivator-1alpha (PGC-1α) is a multi-functional transcription factor that tightly regulates mitochondrial biogenesis and oxidative capacity. The goal of the present study was to explore the potential pathogenic effects of interference by the PGC-1α gene on N-methyl-4-phenylpyridinium ion (MPP+)-induced SH-SY5Y cells. We utilized RNA interference (RNAi) technology to probe the pathogenic consequences of inhibiting PGC-1α in the SH-SY5Y cell line. Remarkably, a reduction in PGC-1α resulted in the reduction of mitochondrial membrane potential, intracellular ATP content and intracellular H2O2 generation, leading to the translocation of cytochrome c (cyt c) to the cytoplasm in the MPP+-induced PD cell model. The expression of related proteins in the signaling pathway (e.g., estrogen-related receptor α (ERRα), nuclear respiratory factor 1 (NRF-1), NRF-2 and Peroxisome proliferator-activated receptor γ (PPARγ)) also decreased. Our finding indicates that small interfering RNA (siRNA) interference targeting the PGC-1α gene could inhibit the function of mitochondria in several capacities and that the PGC-1α gene may modulate mitochondrial function by regulating the expression of ERRα, NRF-1, NRF-2 and PPARγ. Thus, PGC-1α can be considered a potential therapeutic target for PD.
Collapse
Affiliation(s)
- Qinyong Ye
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union HospitalFuzhou, China.,Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical UniversityFuzhou, China
| | - Chun Chen
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union HospitalFuzhou, China
| | - Erwang Si
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union HospitalFuzhou, China
| | - Yousheng Cai
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union HospitalFuzhou, China
| | - Juhua Wang
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union HospitalFuzhou, China
| | - Wanling Huang
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union HospitalFuzhou, China
| | - Dongzhu Li
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union HospitalFuzhou, China
| | - Yingqing Wang
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union HospitalFuzhou, China
| | - Xiaochun Chen
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union HospitalFuzhou, China.,Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical UniversityFuzhou, China
| |
Collapse
|
199
|
Sone M, Morone N, Nakamura T, Tanaka A, Okita K, Woltjen K, Nakagawa M, Heuser JE, Yamada Y, Yamanaka S, Yamamoto T. Hybrid Cellular Metabolism Coordinated by Zic3 and Esrrb Synergistically Enhances Induction of Naive Pluripotency. Cell Metab 2017; 25:1103-1117.e6. [PMID: 28467928 DOI: 10.1016/j.cmet.2017.04.017] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 03/06/2017] [Accepted: 04/15/2017] [Indexed: 01/05/2023]
Abstract
Naive pluripotent stem cells (PSCs) utilize both glycolysis and oxidative phosphorylation (OXPHOS) to satisfy their metabolic demands. However, it is unclear how somatic cells acquire this hybrid energy metabolism during reprogramming toward naive pluripotency. Here, we show that when transduced with Oct4, Sox2, and Klf4 (OSK) into murine fibroblasts, Zic3 and Esrrb synergistically enhance the reprogramming efficiency by regulating cellular metabolic pathways. These two transcription factors (TFs) cooperatively activate glycolytic metabolism independently of hypoxia inducible factors (HIFs). In contrast, the regulatory modes of the TFs on OXPHOS are antagonistic: Zic3 represses OXPHOS, whereas Esrrb activates it. Therefore, when introduced with Zic3, Esrrb restores OXPHOS activity, which is essential for efficient reprogramming. In addition, Esrrb-mediated OXPHOS activation is critical for the conversion of primed PSCs into the naive state. Our study suggests that the combinatorial function of TFs achieves an appropriate balance of metabolic pathways to induce naive PSCs.
Collapse
Affiliation(s)
- Masamitsu Sone
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan; Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Nobuhiro Morone
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan; MRC Toxicology Unit, University of Leicester, Leicester, LE1 9HN, UK
| | - Tomonori Nakamura
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Akito Tanaka
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Keisuke Okita
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Knut Woltjen
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan; Hakubi Center for Advanced Research, Kyoto University, Kyoto 606-8501, Japan
| | - Masato Nakagawa
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - John E Heuser
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yasuhiro Yamada
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan; Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shinya Yamanaka
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan; Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
| | - Takuya Yamamoto
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan; Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan; AMED-CREST, AMED 1-7-1 Otemach, Chiyodaku, Tokyo, 100-0004, Japan.
| |
Collapse
|
200
|
Carnesecchi J, Forcet C, Zhang L, Tribollet V, Barenton B, Boudra R, Cerutti C, Billas IML, Sérandour AA, Carroll JS, Beaudoin C, Vanacker JM. ERRα induces H3K9 demethylation by LSD1 to promote cell invasion. Proc Natl Acad Sci U S A 2017; 114:3909-3914. [PMID: 28348226 PMCID: PMC5393192 DOI: 10.1073/pnas.1614664114] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Lysine Specific Demethylase 1 (LSD1) removes mono- and dimethyl groups from lysine 4 of histone H3 (H3K4) or H3K9, resulting in repressive or activating (respectively) transcriptional histone marks. The mechanisms that control the balance between these two antagonist activities are not understood. We here show that LSD1 and the orphan nuclear receptor estrogen-related receptor α (ERRα) display commonly activated genes. Transcriptional activation by LSD1 and ERRα involves H3K9 demethylation at the transcriptional start site (TSS). Strikingly, ERRα is sufficient to induce LSD1 to demethylate H3K9 in vitro. The relevance of this mechanism is highlighted by functional data. LSD1 and ERRα coregulate several target genes involved in cell migration, including the MMP1 matrix metallo-protease, also activated through H3K9 demethylation at the TSS. Depletion of LSD1 or ERRα reduces the cellular capacity to invade the extracellular matrix, a phenomenon that is rescued by MMP1 reexpression. Altogether our results identify a regulatory network involving a direct switch in the biochemical activities of a histone demethylase, leading to increased cell invasion.
Collapse
Affiliation(s)
- Julie Carnesecchi
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, F-69007 Lyon, France
| | - Christelle Forcet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, F-69007 Lyon, France
| | - Ling Zhang
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, F-69007 Lyon, France
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Violaine Tribollet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, F-69007 Lyon, France
| | - Bruno Barenton
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, F-69007 Lyon, France
| | - Rafik Boudra
- Genetics, Reproduction and Development, Université Blaise Pascal Clermont-Ferrand, CNRS UMR 6293, Inserm U1103, Centre de Recherche en Nutrition Humaine, F-63171 Aubière, France
| | - Catherine Cerutti
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, F-69007 Lyon, France
| | - Isabelle M L Billas
- Department of Integrative Structural Biology, Institute of Genetics and Molecular and Cellular Biology, CNRS UMR7104, Inserm U964, Université de Strasbourg, F-67404 Illkirch, France
| | - Aurélien A Sérandour
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, United Kingdom
| | - Jason S Carroll
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, United Kingdom
| | - Claude Beaudoin
- Genetics, Reproduction and Development, Université Blaise Pascal Clermont-Ferrand, CNRS UMR 6293, Inserm U1103, Centre de Recherche en Nutrition Humaine, F-63171 Aubière, France
| | - Jean-Marc Vanacker
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, F-69007 Lyon, France;
| |
Collapse
|