Reviews

A review on regulation of Rev-erbs in energy homeostasis and research prospect in sports science

  • SHEN Youqing ,
  • HUANG Guoyuan
Expand
  • 1. Department of Physical Education, Hubei University of Education, Wuhan 430205, China;
    2. Department of Sport Science, University of Southern Indiana, Evansville 47712, USA

Received date: 2015-11-30

  Revised date: 2016-10-18

  Online published: 2017-02-07

Abstract

Nuclear receptor Rev-erbs have mainly two subtypes, Rev-erbα and Rev-erbβ, which are widely expressed but mainly prevalent in tissues and organs of mammalians that have predominantly metabolic functions. The expression level of Rev-erbs can be influenced by various factors such as high calorie diet, medicine, and exercise intervention. Rev-erbs are essential to regulate glucose and lipid metabolism, insulin secretion, and blood glucose homeostasis, and are the therapeutic target of chronic diseases such as obesity and diabetes. Rev-erbs may maintain energy homeostasis through related signaling pathway of glucose and lipid metabolism. Recent researches indicate that exercise regiments stimulate Rev-erbs expressions in liver and skeletal muscle and maintain lipid homeostasis, which would be the core mechanism of exercise intervention to chronic diseases. This review analyzes the bioactivity and the mechanism of Rev-erbs, and proposes future studies on Rev-erbs in the sphere of sports science.

Cite this article

SHEN Youqing , HUANG Guoyuan . A review on regulation of Rev-erbs in energy homeostasis and research prospect in sports science[J]. Science & Technology Review, 2016 , 34(24) : 56 -61 . DOI: 10.3981/j.issn.1000-7857.2016.24.007

References

[1] Raghuram S, Stayrook K R, Huang P, et al. Identification of heme as the ligand for the orphan nuclear receptors Rev-erbα and Rev-erbβ[J]. Nature Structural & Molecular Biology, 2007, 14(12):1207-1213.
[2] Solt L A, Wang Y J, Banerjee S, et al. Regulation of circadian behav-iour and metabolism by synthetic Rev-erb agonists[J]. Nature, 2012, 485(7396):62-68.
[3] Zhang Y, Fang B, Emmett M J, et al. Discrete functions of nuclear re-ceptor Rev-erbα couple metabolism to the clock[J]. Science, 2015, 348(6462):1488-1492.
[4] Woldt E, Sebti Y, Solt L A, et al. Rev-erbα modulates skeletal muscle oxidative capacity by regulating mitochondrial biogenesis and autophagy[J]. Nature Medicine, 2013, 19(8):1039-1046.
[5] 王嘉东. 核激素孤儿受体Rev-erbβ对载脂蛋白apoC-III转录调控机制[D]. 上海:复旦大学, 2007. Wang Jiadong. Study on the regulation mechanisms of the nuclear hor-mone receptor Rev-erbβ on aplipoprotein C-III[D]. Shanghai:Fudan University, 2007.
[6] De Mei C, Ercolani L, Parodi C, et al. Dual inhibition of Rev-erbβ and autophagy as a novel pharmacological approach to induce cytotoxicity in cancer cells[J]. Oncogene, 2015, 34(20):2597-2608.
[7] Li T, Eheim A L, Klein S, et al. Novel role of nuclear receptor Reverbα in hepatic stellate cell activation:potential therapeutic target for liver injury[J]. Hepatology, 2014, 59(6):2383-2396.
[8] Chopin-Delannoy S, Thenot S, Delaunay F, et al. A specific and unusu-al nuclear localization signal in the DNA binding domain of the Reverb orphan receptors[J]. Journal of Molecular Endocrinology, 2003, 30(2):197-211.
[9] Lazar M A, Hodin R A, Darling D S, et al. A novel member of the thy-roid/steroid hormone receptor family is encoded by the opposite strand of the rat c-erbA α transcriptional unit[J]. Molecular and Cellular Biol-ogy, 1989, 9(3):1128-1136.
[10] Bonnelye E, Vanacker J M, Desbiens X, et al. Rev-erbβ, a new mem-ber of the nuclear receptor superfamily, is expressed in the nervous system during chicken development[J]. Cell Growth & Differentiation, 1994, 5(12):1357-1365.
[11] Burris T P. Nuclear hormone receptors for heme:Rev-erbα and Reverbβ are ligand-regulated components of the mammalian clock[J]. Mo-lecular Endocrinology, 2008, 22(7):1509-1520.
[12] Ramakrishnan S N, Muscat G E. The orphan Rev-erb nuclear recep-tors:a link between metabolism, circadian rhythm and inflammation[J]. Nuclear Receptor Signaling, 2006, 4:e009.
[13] Bugge A, Feng D, Everett L J, et al. Rev-erbα and Rev-erbβ coordi-nately protect the circadian clock and normal metabolic function[J]. Genes & Development, 2012, 26(7):657-667.
[14] Austin S, Medvedev A, Yan Z H, et al. Induction of the nuclear or-phan receptor RORgamma during adipocyte differentiation of D1 and 3T3-L1 cells[J]. Cell Growth & Differentiation, 1998, 9(3):267-276.
[15] Kojetin D J, Burris T P. A role for Rev-erbα ligands in regulation of adipogenesis[J]. Current Pharmaceutical Design, 2011, 17(4):320-324.
[16] Matta-Camacho E, Banerjee S, Hughes T S, et al. Structure of Reverbβ ligand-binding domain bound to a porphyrin antagonist[J]. Jour-nal of Biological Chemistry, 2014, 289(29):20054-20066.
[17] 商占仙. 生物节律紊乱动脉粥样硬化小鼠中钟基因、脂代谢相关基因改变的研究[D]. 上海:复旦大学, 2011. Shang Zhanxian. Study on the changes of clock genes, lipid metabo-lism-related genes in atherosclerotic mouse with circadian rhythm dis-order[D]. Shanghai:Fudan University, 2011.
[18] Vieira E, Marroquí L, Batista T M, et al. The clock gene Rev-erbα regulates pancreatic β-cell function:Modulation by leptin and highfat diet[J]. Endocrinology, 2012, 153(2):592-601.
[19] 殷洪山. 生物钟紊乱及吸烟致动脉粥样硬化机制的研究[D]. 石家庄:河北医科大学, 2015. Yin Hongshan. Study on the mechanisms of circadian disorders and smoking cause atherosclerosis[D]. Shijiazhuang:Hebei Medical Univer-sity, 2015.
[20] 黄洁. 高脂对小鼠心肌细胞生物钟基因表达的影响[D]. 上海:复旦大学, 2010. Huang Jie. Effects of high fat on the expression of circadian genes in mouse cardiomyocytes[D]. Shanghai:Fudan University, 2010.
[21] Torra I P, Tsibulsky V, Delaunay F, et al. Circadian and glucocorti-coid regulation of Rev-erbα expression in liver[J]. Endocrinology, 2000, 141(10):3799-3806.
[22] Duez H, Staels B. Rev-erbα:An integrator of circadian rhythms and metabolism[J]. Journal of Applied Physiology, 2009, 107(6):1972-1980.
[23] Gervois P, Chopin-Delannoy S, Fadel A, et al. Fibrates increase hu-man Rev-erbα expression in liver via a novel peroxisome proliferatoractivated receptor response element[J]. Molecular Endocrinology, 1999, 13(3):400-409.
[24] Duez H, Staels B. The nuclear receptors Rev-erbs and RORs inte-grate circadian rhythms and metabolism[J]. Diabetes & Vascular Dis-ease Research, 2008, 5(2):82-88.
[25] Kojetin D, Wang Y, Kamenecka T M, et al. Identification of SR8278, a synthetic antagonist of the nuclear heme receptor Rev-erb[J]. ACS Chemical Biology, 2011, 6(2):131-134.
[26] 沈友青, 黄国源, 王配才, 等. 大强度间歇运动对高脂喂养大鼠代谢表征和骨骼肌脂代谢基因表达的影响[J]. 中国体育科技, 2016, 52(1):84-91. Shen Youqing, Huang Guoyuan, Wang Peicai, et al. The effects of high-intensity interval training on metabolic profiles and gene expres-sion of muscle lipid metabolism in high-fat diet rat[J]. China Sport Science and Technology, 2016, 52(1):84-91.
[27] Etain B, Milhiet V, Bellivier F, et al. Genetics of circadian rhythms and mood spectrum disorders[J]. European Neuropsychopharmacology, 2011, 21(S4):S676-682.
[28] Carter R, Mouralidarane A, Soeda J, et al. Non-alcoholic fatty pancre-as disease pathogenesis:A role for developmental programming and al-tered circadian rhythms[J]. PLOS One, 2014, 9(3):e89505.
[29] Marciano D P, Chang M R, Corzo C A, et al. The therapeutic potential of nuclear receptor modulators for treatment of metabolic disorders:PPARγ, RORs, and Rev-erbs[J]. Cell Metabolism, 2014, 19(2):193-208.
[30] Keith D, Finlay L, Butler J, et al. Lipoic acid entrains the hepatic cir-cadian clock and lipid metabolic proteins that have been desynchro-nized with advanced age[J]. Biochemical and Biophysical Research Communications, 2014, 450(1):324-329.
[31] Cho H, Zhao X, Hatori M, et al. Regulation of circadian behaviour and metabolism by Rev-erbα and Rev-erbβ[J]. Nature, 2012, 485(7396):123-127.
[32] Anzulovich A, Mir A, Brewer M, et al. Elovl3:A model gene to dis-sect homeostatic links between the circadian clock and nutritional sta-tus[J]. Journal of Lipid Research, 2006, 47(12):2690-2700.
[33] Sitaula S, Billon C, Kamenecka T M, et al. Suppression of atheroscle-rosis by synthetic Rev-erb agonist[J]. Biochemical and Biophysical Re-search Communications, 2015, 460(3):566-571.
[34] Coste H, Rodriguez J C. Orphan nuclear hormone receptor Rev-erbα regulates the human apolipoprotein CIII promoter[J]. Journal of Biolog-ical Chemistry, 2002, 277(30):27120-27129.
[35] Garaulet M, Smith C E, Gomez-Abellán P, et al. Rev-erb-ALPHA cir-cadian gene variant associates with obesity in two independent popula-tions:Mediterranean and North American[J]. Molecular Nutrition & Food Research, 2014, 58(4):821-829.
[36] Goumidi L, Grechez A, Dumont J, et al. Impact of Rev-erbα gene polymorphisms on obesity phenotypes in adult and adolescent samples[J]. International Journal of Obesity, 2013, 37(5):666-672.
[37] Lee P, Bova R, Schofield L, et al. Brown Adipose Tissue Exhibits a Glucose-Responsive Thermogenic Biorhythm in Humans[J]. Cell Me-tabolism, 2016, 23(4):602-609.
[38] Ramakrishnan S N, Lau P, Burke L J, et al. Rev-erbβ regulates the expression of genes involved in lipid absorption in skeletal muscle cells:evidence for cross-talk between orphan nuclear receptors and myokines[J]. Journal of Biological Chemistry, 2005, 280(10):8651-8659.
[39] Raspé E, Duez H, Mansén A, et al. Identification of Rev-erbα as a physiological repressor of apoC-III gene transcription[J]. Journal of Lipid Research, 2002, 43(12):2172-2179.
[40] Yin L, Wu N, Curtin J C, et al. Rev-erbα, a heme sensor that coordi-nates metabolic and circadian pathways[J]. Science, 2007, 318(5857):1786-1789.
[41] Gatfield D, Le Martelot G, Vejnar C E, et al. Integration of microRNA miR-122 in hepatic circadian gene expression[J]. Genes & Develop-ment, 2009, 23(11):1313-1326.
[42] Estall J L, Ruas J L, Choi C S, et al. PGC-1α negatively regulates he-patic FGF21 expression by modulating the heme/Rev-erbα axis[J]. Proceedings of the National Academy of Sciences of the United States of America, 2009, 106(52):22510-22515.
[43] Le Martelot G, Claudel T, Gatfield D, et al. Rev-erbα participates in circadian SREBP signaling and bile acid homeostasis[J]. PLOS Biolo-gy, 2009, 7(9):e1000181.
[44] Lefebvre P, Cariou B, Lien F, et al. Role of bile acids and bile acid re-ceptors in metabolic regulation[J]. Physiological Reviews, 2009, 89(1):147-191.
[45] Lin J D, Handschin C, Spiegelman B M. Metabolic control through the PGC-1 family of transcription coactivators[J]. Cell Metabolism, 2005, 1(6):361-370.
[46] Shen Y Q, Xu X F, Yue K, et al. Effect of different exercise protocols on metabolic profiles and fatty acid metabolism in skeletal muscle in high-fat diet-fed rats[J]. Obesity, 2015, 23(5):1001-1006.
[47] 沈友青, 宋涛, 徐国栋. 持续运动和间歇运动对高脂喂养大鼠肝脏Rev-erbα和SCD1表达的影响[J]. 西安体育学院学报, 2015, 32(5):606-610. Shen Youqing, Song Tao, Xu Guodong. Effect of mild-intensity endur-ance and heavy-intensity interval exercise on hepatic Rev-erbα and SCD1 in rats fed with high-fat fiet[J]. Journal of Xi'an Physical Educa-tion University, 2015, 32(5):606-610.
[48] Pircher P, Chomez P, Yu F, et al. Aberrant expression of myosin iso-forms in skeletal muscles from mice lacking the Rev-erbα orphan re-ceptor gene[J]. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology, 2005, 288(2):R482-490.
Outlines

/