AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (2 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Review | Open Access

Major roles of the circadian clock in cancer

Chen Huang1,*Chenliang Zhang2,*Yubin Cao3Jian Li4Feng Bi1 ( )
Department of Abdominal Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu 610000, China
Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu 610000, China
Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610000, China
West China School of Medicine, Sichuan University, Chengdu 610000, China

*These authors contributed equally to this work.

Show Author Information

Abstract

Circadian rhythms are natural rhythms that widely exist in all creatures, and regulate the processes and physiological functions of various biochemical reactions. The circadian clock is critical for cancer occurrence and progression. Its function is regulated by metabolic activities, and the expression and transcription of various genes. This review summarizes the composition of the circadian clock; the biological basis for its function; its relationship with, and mechanisms in, cancer; its various functions in different cancers; the effects of anti-tumor treatment; and potential therapeutic targets. Research in this area is expected to advance understanding of circadian locomotor output cycles kaput (CLOCK) and brain and muscle ARNT-like protein 1 (BMAL1) in tumor diseases, and contribute to the development of new anti-tumor treatment strategies.

Electronic Supplementary Material

Download File(s)
cbm-20-1-1_ESM.pdf (2.9 MB)

References

1

Shafi AA, Knudsen KE. Cancer and the circadian clock. Cancer Res. 2019; 79: 3806-14.

2

Ouyang Y, Andersson CR, Kondo T, Golden SS, Johnson CH. Resonating circadian clocks enhance fitness in cyanobacteria. Proc Natl Acad Sci U S A. 1998; 95: 8660-4.

3

Turek FW. Circadian clocks: not your grandfather’s clock. Science. 2016; 354: 992-3.

4

Mohawk JA, Green CB, Takahashi JS. Central and peripheral circadian clocks in mammals. Annu Rev Neurosci. 2012; 35: 445-62.

5

Sancar A, van Gelder RN. Clocks, cancer, and chronochemotherapy. Science. 2021; 371: eabb0738.

6

Partch CL, Green CB, Takahashi JS. Molecular architecture of the mammalian circadian clock. Trends Cell Biol. 2014; 24: 90-9.

7

Patke A, Young MW, Axelrod S. Molecular mechanisms and physiological importance of circadian rhythms. Nat Rev Mol Cell Biol. 2020; 21: 67-84.

8

Reppert SM, Weaver DR. Coordination of circadian timing in mammals. Nature. 2002; 418: 935-41.

9

Hastings MH, Reddy AB, Maywood ES. A clockwork web: circadian timing in brain and periphery, in health and disease. Nat Rev Neurosci. 2003; 4: 649-61.

10

Cederroth CR, Albrecht U, Bass J, Brown SA, Dyhrfjeld-Johnsen J, Gachon F, et al. Medicine in the fourth dimension. Cell Metab. 2019; 30: 238-50.

11

Lubov JE, Cvammen W, Kemp MG. The impact of the circadian clock on skin physiology and cancer development. Int J Mol Sci. 2021; 22: 6112.

12

Kume K, Zylka MJ, Sriram S, Shearman LP, Weaver DR, Jin X, et al. mCRY1 and mCRY2 are essential components of the negative limb of the circadian clock feedback loop. Cell. 1999; 98: 193-205.

13

Griffin EA, Staknis D, Weitz CJ. Light-independent role of CRY1 and CRY2 in the mammalian circadian clock. Science. 1999; 286: 768-71.

14

Sangoram AM, Saez L, Antoch MP, Gekakis N, Staknis D, Whiteley A, et al. Mammalian circadian autoregulatory loop: a timeless ortholog and mPer1 interact and negatively regulate CLOCKBMAL1-induced transcription. Neuron. 1998; 21: 1101-13.

15

Narasimamurthy R, Virshup DM. The phosphorylation switch that regulates ticking of the circadian clock. Mol Cell 2021; 81: 1133-46.

16

Kondratov RV, Chernov MV, Kondratova AA, Gorbacheva VY, Gudkov AV, Antoch MP. BMAL1-dependent circadian oscillation of nuclear CLOCK: posttranslational events induced by dimerization of transcriptional activators of the mammalian clock system. Genes Dev. 2003; 17: 1921-32.

17

Kondratov RV, Shamanna RK, Kondratova AA, Gorbacheva VY, Antoch MP. Dual role of the CLOCK/BMAL1 circadian complex in transcriptional regulation. FASEB J. 2006; 20: 530-2.

18

Xu Y, Padiath QS, Shapiro RE, Jones CR, Wu SC, Saigoh N, et al. Functional consequences of a CKIdelta mutation causing familial advanced sleep phase syndrome. Nature. 2005; 434: 640-4.

19

Etchegaray JP, Machida KK, Noton E, Constance CM, Dallmann R, Di Napoli MN, et al. Casein kinase 1 delta regulates the pace of the mammalian circadian clock. Mol Cell Biol. 2009; 29: 3853-66.

20

Philpott JM, Narasimamurthy R, Ricci CG, Freeberg AM, Hunt SR, Yee LE, et al. Casein kinase 1 dynamics underlie substrate selectivity and the PER2 circadian phosphoswitch. Elife. 2020; 9: e52343.

21

Green CB. Circadian posttranscriptional regulatory mechanisms in mammals. Cold Spring Harb Perspect Biol. 2018; 10: a030692.

22

Shilts J, Chen G, Hughey JJ. Evidence for widespread dysregulation of circadian clock progression in human cancer. PeerJ. 2018; 6: e4327.

23

Shostak A, Ruppert B, Ha N, Bruns P, Toprak UH, Eils R, et al. MYC/MIZ1-dependent gene repression inversely coordinates the circadian clock with cell cycle and proliferation. Nat Commun. 2016; 7: 11807.

24

Huber AL, Papp SJ, Chan AB, Henriksson E, Jordan SD, Kriebs A, et al. CRY2 and FBXL3 cooperatively degrade c-MYC. Mol Cell. 2016; 64: 774-89.

25

Aviram R, Manella G, Kopelman N, Neufeld-Cohen A, Zwighaft Z, Elimelech M, et al. Lipidomics analyses reveal temporal and spatial lipid organization and uncover daily oscillations in intracellular organelles. Mol Cell 2016; 62: 636-48.

26

Blask DE, Dauchy RT, Dauchy EM, Mao L, Hill SM, Greene MW, et al. Light exposure at night disrupts host/cancer circadian regulatory dynamics: impact on the Warburg effect, lipid signaling and tumor growth prevention. PLoS One. 2014; 9: e102776.

27

Gamble KL, Berry R, Frank SJ, Young ME. Circadian clock control of endocrine factors. Nat Rev Endocrinol. 2014; 10: 466-75.

28

Greene MW. Circadian rhythms and tumor growth. Cancer Lett. 2012; 318: 115-23.

29

He W, Holtkamp S, Hergenhan SM, Kraus K, de Juan A, Weber J, et al. Circadian expression of migratory factors establishes lineagespecific signatures that guide the homing of leukocyte subsets to tissues. Immunity. 2018; 49: 1175-90.e7.

30

Aiello I, Fedele MLM, Román F, Marpegan L, Caldart C, Chiesa JJ, et al. Circadian disruption promotes tumor-immune microenvironment remodeling favoring tumor cell proliferation. Sci Adv. 2020; 6: eaaz4530.

31

Hadadi E, Taylor W, Li XM, Aslan Y, Villote M, Rivière J, et al. Chronic circadian disruption modulates breast cancer stemness and immune microenvironment to drive metastasis in mice. Nat Commun. 2020; 11: 3193.

32

Wu Y, Tao B, Zhang T, Fan Y, Mao R. Pan-cancer analysis reveals disrupted circadian clock associates with T cell exhaustion. Front Immunol. 2019; 10: 2451.

33

Thorsson V, Gibbs DL, Brown SD, Wolf D, Bortone DS, Ou Yang TH, et al. The immune landscape of cancer. Immunity. 2018; 48: 812-30.e14.

34

Xie Y, Tang Q, Chen G, Xie M, Yu S, Zhao J, et al. New insights into the circadian rhythm and its related diseases. Front Physiol. 2019; 10: 682.

35

Leng Y, Musiek ES, Hu K, Cappuccio FP, Yaffe K. Association between circadian rhythms and neurodegenerative diseases. Lancet. Neurol. 2019; 18: 307-18.

36

Kinouchi K, Sassone-Corsi P. Metabolic rivalry: circadian homeostasis and tumorigenesis. Nat Rev Cancer. 2020; 20: 645-61.

37

Schibler U, Gotic I, Saini C, Gos P, Curie T, Emmenegger Y, et al. Clock-talk: interactions between central and peripheral circadian oscillators in mammals. Cold Spring Harb Symp Quant Biol. 2015; 80: 223-32.

38

Cox KH, Takahashi JS. Circadian clock genes and the transcriptional architecture of the clock mechanism. J Mol Endocrinol. 2019; 63: R93-102.

39

Finger AM, Dibner C, Kramer A. Coupled network of the circadian clocks: a driving force of rhythmic physiology. FEBS Lett. 2020; 594: 2734-69.

40

Numata M, Hirano A, Yamamoto Y, Yasuda M, Miura N, Sayama K, et al. Metastasis of breast cancer promoted by circadian rhythm disruption due to light/dark shift and its prevention by dietary quercetin in mice. J Circadian Rhythms. 2021; 19: 2.

41

Ha NH, Long J, Cai Q, Shu XO, Hunter KW. The circadian rhythm gene Arntl2 is a metastasis susceptibility gene for estrogen receptornegative breast cancer. PLoS Genetics. 2016; 12: e1006267.

42

Chen J, Liu A, Lin Z, Wang B, Chai X, Chen S, et al. Downregulation of the circadian rhythm regulator HLF promotes multiple-organ distant metastases in non-small cell lung cancer through PPAR/NF-κb signaling. Cancer Lett. 2020; 482: 56-71.

43

Wang Y, Sun N, Lu C, Bei Y, Qian R, Hua L. Upregulation of circadian gene ‘hClock’ contribution to metastasis of colorectal cancer. Int J Oncol. 2017; 50: 2191-9.

44

Koritala BSC, Porter KI, Arshad OA, Gajula RP, Mitchell HD, Arman T, et al. Night shift schedule causes circadian dysregulation of DNA repair genes and elevated DNA damage in humans. J Pineal Res. 2021; 70: e12726.

45

Dun A, Zhao X, Jin X, Wei T, Gao X, Wang Y, et al. Association between night-shift work and cancer risk: updated systematic review and meta-analysis. Front Oncol. 2020; 10: 1006.

46

Srour B, Plancoulaine S, Andreeva VA, Fassier P, Julia C, Galan P, et al. Circadian nutritional behaviours and cancer risk: new insights from the NutriNet-santé prospective cohort study: disclaimers. Int J Cancer. 2018; 143: 2369-79.

47

Lou X, Wang H, Tu Y, Tan W, Jiang C, Sun J, et al. Alterations of sleep quality and circadian rhythm genes expression in elderly thyroid nodule patients and risks associated with thyroid malignancy. Sci Rep. 2021; 11: 13682.

48

Kondratov RV, Kondratova AA, Gorbacheva VY, Vykhovanets OV, Antoch MP. Early aging and age-related pathologies in mice deficient in BMAL1, the core componentof the circadian clock. Genes Dev. 2006; 20: 1868-73.

49

Yu EA, Weaver DR. Disrupting the circadian clock: gene-specific effects on aging, cancer, and other phenotypes. Aging. 2011; 3: 479-93.

50

Papagiannakopoulos T, Bauer MR, Davidson SM, Heimann M, Subbaraj L, Bhutkar A, et al. Circadian rhythm disruption promotes lung tumorigenesis. Cell Metab. 2016; 24: 324-31.

51

Sulli G, Lam MTY, Panda S. Interplay between circadian clock and cancer: new frontiers for cancer treatment. Trends Cancer. 2019; 5: 475-94.

52

Zhou L, Luo Z, Li Z, Huang Q. Circadian clock is associated with tumor microenvironment in kidney renal clear cell carcinoma. Aging. 2020; 12: 14620-32.

53

Yang Y, Yuan G, Xie H, Wei T, Zhu D, Cui J, et al. Circadian clock associates with tumor microenvironment in thoracic cancers. Aging. 2019; 11: 11814-28.

54

Pavlova NN, Thompson CB. The emerging hallmarks of cancer metabolism. Cell Metab. 2016; 23: 27-47.

55

Chen WD, Wen MS, Shie SS, Lo YL, Wo HT, Wang CC, et al. The circadian rhythm controls telomeres and telomerase activity. Biochem Biophys Res Commun. 2014; 451: 408-14.

56

Ruan W, Yuan X, Eltzschig HK. Circadian rhythm as a therapeutic target. Nat Rev Drug Discov. 2021; 20: 287-307.

57

Chen P, Hsu WH, Han J, Xia Y, DePinho RA. Cancer stemness meets immunity: from mechanism to therapy. Cell Rep. 2021; 34: 108597.

58

Chen P, Hsu WH, Chang A, Tan Z, Lan Z, Zhou A, et al. Circadian regulator CLOCK recruits immune-suppressive microglia into the GBM tumor microenvironment. Cancer Discov. 2020; 10: 371-81.

59

Dong Z, Zhang G, Qu M, Gimple RC, Wu Q, Qiu Z, et al. Targeting glioblastoma stem cells through disruption of the circadian clock. Cancer Discov. 2019; 9: 1556-73.

60

Puram RV, Kowalczyk MS, de Boer CG, Schneider RK, Miller PG, McConkey M, et al. Core circadian clock genes regulate leukemia stem cells in AML. Cell. 2016; 165: 303-16.

61

Hu Z, Brooks SA, Dormoy V, Hsu CW, Hsu HY, Lin LT, et al. Assessing the carcinogenic potential of low-dose exposures to chemical mixtures in the environment: focus on the cancer hallmark of tumor angiogenesis. Carcinogenesis. 2015; 36 Suppl 1: S184-202.

62

Jiang X, Wang J, Deng X, Xiong F, Zhang S, Gong Z, et al. The role of microenvironment in tumor angiogenesis. J Exp Clin Cancer Res. 2020; 39: 204.

63

Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011; 144: 646-74.

64

Shalapour S, Karin M. Pas de deux: control of anti-tumor immunity by cancer-associated inflammation. Immunity. 2019; 51: 15-26.

65

Ramos CA, Ouyang C, Qi Y, Chung Y, Cheng CT, LaBarge MA, et al. A non-canonical function of BMAL1 metabolically limits obesitypromoted triple-negative breast cancer. iScience. 2020; 23: 100839.

66

Matsunaga N, Ogino T, Hara Y, Tanaka T, Koyanagi S, Ohdo S. Optimized dosing schedule based on circadian dynamics of mouse breast cancer stem cells improves the antitumor effects of aldehyde dehydrogenase inhibitor. Cancer Res. 2018; 78: 3698-708.

67

Chefetz I, Grimley E, Yang K, Hong L, Vinogradova EV, Suciu R, et al. A pan-ALDH1A inhibitor induces necroptosis in ovarian cancer stem-like cells. Cell Rep. 2019; 26: 3061-3075.e6.

68

Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature. 1999; 402: 656-60.

69

Avau B, Carbone F, Tack J, Depoortere I. Ghrelin signaling in the gut, its physiological properties, and therapeutic potential. Neurogastroenterol Motil. 2013; 25: 720-32.

70

Müller TD, Nogueiras R, Andermann ML, Andrews ZB, Anker SD, Argente J, et al. Ghrelin. Mol Metab. 2015; 4: 437-60.

71

Waseem T, Javaid Ur R, Ahmad F, Azam M, Qureshi MA. Role of ghrelin axis in colorectal cancer: a novel association. Peptides. 2008; 29: 1369-76.

72

Murata M, Okimura Y, Iida K, Matsumoto M, Sowa H, Kaji H, et al. Ghrelin modulates the downstream molecules of insulin signaling in hepatoma cells. J Biol Chem. 2002; 277: 5667-74.

73

De Vriese C, Grégoire F, De Neef P, Robberecht P, Delporte C. Ghrelin is produced by the human erythroleukemic HEL cell line and involved in an autocrine pathway leading to cell proliferation. Endocrinology. 2005; 146: 1514-22.

74

Fung JNT, Seim I, Wang D, Obermair A, Chopin LK, Chen C. Expression and in vitro functions of the ghrelin axis in endometrial cancer. Horm Cancer. 2010; 1: 245-55.

75

Duxbury MS, Waseem T, Ito H, Robinson MK, Zinner MJ, Ashley SW, et al. Ghrelin promotes pancreatic adenocarcinoma cellular proliferation and invasiveness. Biochem Biophys Res Commun. 2003; 309: 464-68.

76

Lien GS, Lin CH, Yang YL, Wu MS, Chen BC. Ghrelin induces colon cancer cell proliferation through the GHS-R, Ras, PI3K, Akt, and mTOR signaling pathways. Eur J Pharmacol. 2016; 776: 124-31.

77

Kraus D, Reckenbeil J, Wenghoefer M, Stark H, Frentzen M, Allam JP, et al. Ghrelin promotes oral tumor cell proliferation by modifying GLUT1 expression. Cell Mol Life Sci. 2016; 73: 1287-99.

78

Díaz-Lezama N, Hernández-Elvira M, Sandoval A, Monroy A, Felix R, Monjaraz E. Ghrelin inhibits proliferation and increases T-type Ca2+ channel expression in PC-3 human prostate carcinoma cells. Biochem Biophys Res Commun. 2010; 403: 24-9.

79

Yeh AH, Jeffery PL, Duncan RP, Herington AC, Chopin LK. Ghrelin and a novel preproghrelin isoform are highly expressed in prostate cancer and ghrelin activates mitogen-activated protein kinase in prostate cancer. Clin Cancer Res. 2005; 11: 8295-303.

80

Segers A, Desmet L, Sun S, Verbeke K, Tack J, Depoortere I. Nighttime feeding of Bmal1-/- mice restores SCFA rhythms and their effect on ghrelin. J Endocrinol. 2020; 245: 155-64.

81

Cummings DE, Purnell JQ, Frayo RS, Schmidova K, Wisse BE, Weigle DS. A preprandial rise in plasma ghrelin levels suggests a role in meal initiation in humans. Diabetes. 2001; 50: 1714-9.

82

Bodosi B, Gardi J, Hajdu I, Szentirmai E, Obal F, Krueger JM. Rhythms of ghrelin, leptin, and sleep in rats: effects of the normal diurnal cycle, restricted feeding, and sleep deprivation. Am J Physiol Regul Integr Comp Physiol. 2004; 287: R1071-9.

83

Yildiz BO, Suchard MA, Wong ML, McCann SM, Licinio J. Alterations in the dynamics of circulating ghrelin, adiponectin, and leptin in human obesity. Proc Natl Acad Sci U S A. 2004; 101: 10434-9.

84

Laermans J, Vancleef L, Tack J, Depoortere I. Role of the clock gene Bmal1 and the gastric ghrelin-secreting cell in the circadian regulation of the ghrelin-GOAT system. Sci Rep. 2015; 5: 16748.

85

Diehl JA, Fuchs SY, Koumenis C. The cell biology of the unfolded protein response. Gastroenterology. 2011; 141: 38-41.

86

Bu Y, Yoshida A, Chitnis N, Altman BJ, Tameire F, Oran A, et al. A PERK-miR-211 axis suppresses circadian regulators and protein synthesis to promote cancer cell survival. Nat Cell Biol. 2018; 20: 104-15.

87

Hart LS, Cunningham JT, Datta T, Dey S, Tameire F, Lehman SL, et al. ER stress-mediated autophagy promotes Myc-dependent transformation and tumor growth. J Clin Invest. 2012; 122: 4621-34.

88

Cramer T, Yamanishi Y, Clausen BE, Förster I, Pawlinski R, Mackman N, et al. HIF-1alpha is essential for myeloid cellmediated inflammation. Cell. 2003; 112: 645-57.

89

Masson N, Ratcliffe PJ. Hypoxia signaling pathways in cancer metabolism: the importance of co-selecting interconnected physiological pathways. Cancer Metab. 2014; 2: 3.

90

Semenza GL, Roth PH, Fang HM, Wang GL. Transcriptional regulation of genes encoding glycolytic enzymes by hypoxiainducible factor 1. J Biol Chem. 1994; 269: 23757-63.

91

Doedens AL, Stockmann C, Rubinstein MP, Liao D, Zhang N, DeNardo DG, et al. Macrophage expression of hypoxia-inducible factor-1 alpha suppresses T-cell function and promotes tumor progression. Cancer Res. 2010; 70: 7465-75.

92

Steggerda SM, Bennett MK, Chen J, Emberley E, Huang T, Janes JR, et al. Inhibition of arginase by CB-1158 blocks myeloid cellmediated immune suppression in the tumor microenvironment. J Immunother Cancer. 2017; 5: 101.

93

Nguyen KD, Fentress SJ, Qiu Y, Yun K, Cox JS, Chawla A. Circadian gene Bmal1 regulates diurnal oscillations of Ly6C(hi) inflammatory monocytes. Science. 2013; 341: 1483-8.

94

Alexander RK, Liou YH, Knudsen NH, Starost KA, Xu C, Hyde AL, et al. Bmal1 integrates mitochondrial metabolism and macrophage activation. ELife. 2020; 9: e54090.

95

DeMarzo AM, Nelson WG, Isaacs WB, Epstein JI. Pathological and molecular aspects of prostate cancer. Lancet (London, England). 2003; 361: 955-64.

96

Albihn A, Johnsen JI, Henriksson MA. MYC in oncogenesis and as a target for cancer therapies. Adv Cancer Res. 2010; 107: 163-224.

97

Fernandez PC, Frank SR, Wang L, Schroeder M, Liu S, Greene J, et al. Genomic targets of the human c-Myc protein. Genes Dev. 2003; 17: 1115-29.

98

Adhikary S, Eilers M. Transcriptional regulation and transformation by Myc proteins. Nat Rev Mol Cell Biol. 2005; 6: 635-45.

99

Larsson LG, Henriksson MA. The Yin and Yang functions of the Myc oncoprotein in cancer development and as targets for therapy. Exp Cell Res. 2010; 316: 1429-37.

100

Altman BJ, Hsieh AL, Sengupta A, Krishnanaiah SY, Stine ZE, Walton ZE, et al. MYC disrupts the circadian clock and metabolism in cancer cells. Cell Metab. 2015; 22: 1009-19.

101

Gery S, Komatsu N, Baldjyan L, Yu A, Koo D, Koeffler HP. The circadian gene per1 plays an important role in cell growth and DNA damage control in human cancer cells. Mol Cell. 2006; 22: 375-82.

102

Hua H, Wang Y, Wan C, Liu Y, Zhu B, Yang C, et al. Circadian gene mPer2 overexpression induces cancer cell apoptosis. Cancer Sci. 2006; 97: 589-96.

103

Yang X, Wood PA, Ansell CM, Quiton DFT, Oh EY, Du-Quiton J, et al. The circadian clock gene Per1 suppresses cancer cell proliferation and tumor growth at specific times of day. Chronobiol Int. 2009; 26: 1323-39.

104

Repouskou A, Prombona A. c-MYC targets the central oscillator gene Per1 and is regulated by the circadian clock at the posttranscriptional level. Biochim Biophys Acta. 2016; 1859: 541-52.

105

O’Connor K, Chen M. Dynamic functions of RhoA in tumor cell migration and invasion. Small GTPases. 2013; 4: 141-7.

106

Chiba S, Enami T, Ogawa S, Sakata-Yanagimoto M. G17V RHOA: Genetic evidence of GTP-unbound RHOA playing a role in tumorigenesis in T cells. Small GTPases. 2015; 6: 100-3.

107

Nomikou E, Stournaras C, Kardassis D. Functional analysis of the promoters of the small GTPases RhoA and RhoB in embryonic stem cells. Biochem Biophys Res Commun. 2017; 491: 754-9.

108

Ma TJ, Zhang ZW, Lu YL, Zhang YY, Tao DC, Liu YQ, et al. CLOCK and BMAL1 stabilize and activate RHOA to promote F-actin formation in cancer cells. Exp Mol Med. 2018; 50: 1-50.

109

Ramanathan C, Kathale ND, Liu D, Lee C, Freeman DA, Hogenesch JB, et al. mTOR signaling regulates central and peripheral circadian clock function. PLoS Genet. 2018; 14: e1007369.

110

Memmott RM, Dennis PA. Akt-dependent and -independent mechanisms of mTOR regulation in cancer. Cell Signal. 2009; 21: 656-64.

111

Chiang GG, Abraham RT. Targeting the mTOR signaling network in cancer. Trends Mol Med. 2007; 13: 433-42.

112

Alzahrani AS. PI3K/Akt/mTOR inhibitors in cancer: at the bench and bedside. Semin Cancer Biol. 2019; 59: 125-32.

113

Hardie DG. AMP-activated/SNF1 protein kinases: conserved guardians of cellular energy. Nat Rev Mol Cell Biol. 2007; 8: 774-85.

114

Lee Y, Kim EK. AMP-activated protein kinase as a key molecular link between metabolism and clockwork. Exp Mol Med. 2013; 45: e33.

115

Um JH, Yang S, Yamazaki S, Kang H, Viollet B, Foretz M, et al. Activation of 5’-AMP-activated kinase with diabetes drug metformin induces casein kinase Iepsilon (CKIepsilon)-dependent degradation of clock protein mPer2. J Biol Chem. 2007; 282: 20794-8.

116

Liang J, Mills GB. AMPK: a contextual oncogene or tumor suppressor? Cancer Res. 2013; 73: 2929-35.

117

Carling D. AMPK signalling in health and disease. Curr Opin Cell Biol. 2017; 45: 31-7.

118

Li Q, Xia D, Wang Z, Liu B, Zhang J, Peng P, et al. Circadian rhythm gene PER3 negatively regulates stemness of prostate cancer stem cells via WNT/β-catenin signaling in tumor microenvironment. Front Cell Dev Biol. 2021; 9: 656981.

119

Maiese K. Moving to the rhythm with clock (Circadian) genes, autophagy, mTOR, and SIRT1 in degenerative disease and cancer. Curr Neurovasc Res. 2017; 14: 299-304.

120

Okazaki F, Matsunaga N, Okazaki H, Azuma H, Hamamura K, Tsuruta A, et al. Circadian clock in a mouse colon tumor regulates intracellular iron levels to promote tumor progression. J Biol Chem. 2016; 291: 7017-28.

121

Shen Y, Endale M, Wang W, Morris AR, Francey LJ, Harold RL, et al. NF-κB modifies the mammalian circadian clock through interaction with the core clock protein BMAL1. PLoS Genet. 2021; 17: e1009933.

122

Kubra S, Zhang H, Si Y, Gao X, Wang T, Pan L, et al. REGγ regulates circadian clock by modulating BMAL1 protein stability. Cell Death Discov. 2021; 7: 335.

123

Moreno-Smith M, Milazzo G, Tao L, Fekry B, Zhu B, Mohammad MA, et al. Restoration of the molecular clock is tumor suppressive in neuroblastoma. Nat Commun. 2021; 12: 4006.

124

Jiang W, Zhao S, Jiang X, Zhang E, Hu G, Hu B, et al. The circadian clock gene Bmal1 acts as a potential anti-oncogene in pancreatic cancer by activating the p53 tumor suppressor pathway. Cancer Lett. 2016; 371: 314-25.

125

Liu L, Liao JZ, He XX, Li PY. The role of autophagy in hepatocellular carcinoma: friend or foe. Oncotarget. 2017; 8: 57707-22.

126

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015; 65: 87-108.

127
World Health Organization, Projections of Mortality and Causes of Death, 2016 to 2060. Geneva, Switzerland: World Health Organization; 2020.
128

Singal AG, El-Serag HB. Hepatocellular carcinoma from epidemiology to prevention: translating knowledge into practice. Clin Gastroenterol Hepatol. 2015; 13: 2140-51.

129

Fattovich G, Stroffolini T, Zagni I, Donato F. Hepatocellular carcinoma in cirrhosis: incidence and risk factors. Gastroenterology. 2004; 127(5 Suppl 1): S35-50.

130

Molina-Aguilar C, Guerrero-Carrillo MdJ, Espinosa-Aguirre JJ, Olguin-Reyes S, Castro-Belio T, Vázquez-Martínez O, et al. Time-caloric restriction inhibits the neoplastic transformation of cirrhotic liver in rats treated with diethylnitrosamine. Carcinogenesis. 2017; 38: 847-58.

131

Krizkova S, Kepinska M, Emri G, Rodrigo MAM, Tmejova K, Nerudova D, et al. Microarray analysis of metallothioneins in human diseases--A review. J Pharm Biomed Anal, 2016; 117: 464-73.

132

Fujie T, Segawa Y, Yoshida E, Kimura T, Fujiwara Y, Yamamoto C, et al. Induction of metallothionein isoforms by copper diethyldithiocarbamate in cultured vascular endothelial cells. J Toxicol Sci. 2016; 41: 225-32.

133

Klaassen CD, Liu J, Choudhuri S. Metallothionein: an intracellular protein to protect against cadmium toxicity. Annu Rev Pharmacol Toxicol. 1999; 39: 267-94.

134

Li H, Lu YF, Chen H, Liu J. Dysregulation of metallothionein and circadian genes in human hepatocellular carcinoma. Chronobiol Int. 2017; 34: 192-202.

135

Hill SM, Belancio VP, Dauchy RT, Xiang S, Brimer S, Mao L, et al. Melatonin: an inhibitor of breast cancer. Endocr Relat Cancer. 2015; 22: R183-204.

136

Reiter RJ, Rosales-Corral SA, Tan DX, Acuna-Castroviejo D, Qin L, Yang SF, et al. Melatonin, a full service anti-cancer agent: inhibition of initiation, progression and metastasis. Int J Mol Sci. 2017; 18: 843.

137

Li Y, Li S, Zhou Y, Meng X, Zhang JJ, Xu DP, et al. Melatonin for the prevention and treatment of cancer. Oncotarget. 2017; 8: 39896-921.

138

Sánchez DI, González-Fernández B, Crespo I, San-Miguel B, Álvarez M, González-Gallego J, et al. Melatonin modulates dysregulated circadian clocks in mice with diethylnitrosamineinduced hepatocellular carcinoma. J Pineal Res. 2018; 65: e12506.

139

Sladek FM, Zhong WM, Lai E, Darnell JE. Liver-enriched transcription factor HNF-4 is a novel member of the steroid hormone receptor superfamily. Genes Dev. 1990; 4: 2353-65.

140

Battle MA, Konopka G, Parviz F, Gaggl AL, Yang C, Sladek FM, et al. Hepatocyte nuclear factor 4alpha orchestrates expression of cell adhesion proteins during the epithelial transformation of the developing liver. Proc Natl Acad Sci U S A. 2006; 103: 8419-24.

141

Bonzo JA, Ferry CH, Matsubara T, Kim JH, Gonzalez FJ. Suppression of hepatocyte proliferation by hepatocyte nuclear factor 4α in adult mice. J Biol Chem. 2012; 287: 7345-56.

142

Chellappa K, Deol P, Evans JR, Vuong LM, Chen G, Briançon N, et al. Opposing roles of nuclear receptor HNF4α isoforms in colitis and colitis-associated colon cancer. ELife. 2016; 5: e10903.

143

Walesky C, Apte U. Role of hepatocyte nuclear factor 4α (HNF4α) in cell proliferation and cancer. Gene Expr. 2015; 16: 101-8.

144

Vuong LM, Chellappa K, Dhahbi JM, Deans JR, Fang B, Bolotin E, et al. Differential effects of hepatocyte nuclear factor 4α isoforms on tumor growth and T-cell factor 4/AP-1 interactions in human colorectal cancer cells. Mol Cell Biol. 2015; 35: 3471-90.

145

Ning BF, Ding J, Yin C, Zhong W, Wu K, Zeng X, et al. Hepatocyte nuclear factor 4 alpha suppresses the development of hepatocellular carcinoma. Cancer Res. 2010; 70: 7640-51.

146

Hatziapostolou M, Polytarchou C, Aggelidou E, Drakaki A, Poultsides GA, Jaeger SA, et al. An HNF4α-miRNA inflammatory feedback circuit regulates hepatocellular oncogenesis. Cell. 2011; 147: 1233-47.

147

Walesky C, Edwards G, Borude P, Gunewardena S, O’Neil M, Yoo B, et al. Hepatocyte nuclear factor 4 alpha deletion promotes diethylnitrosamine-induced hepatocellular carcinoma in rodents. Hepatology. 2013; 57: 2480-90.

148

Tanaka T, Jiang S, Hotta H, Takano K, Iwanari H, Sumi K, et al. Dysregulated expression of P1 and P2 promoter-driven hepatocyte nuclear factor-4alpha in the pathogenesis of human cancer. J Pathol. 2006; 208: 662-72.

149

Chellappa K, Jankova L, Schnabl JM, Pan S, Brelivet Y, Fung CLS, et al. Src tyrosine kinase phosphorylation of nuclear receptor HNF4α correlates with isoform-specific loss of HNF4α in human colon cancer. Proc Natl Acad Sci U S A. 2012; 109: 2302-7.

150

Fekry B, Ribas-Latre A, Baumgartner C, Deans JR, Kwok C, Patel P, et al. Incompatibility of the circadian protein BMAL1 and HNF4α in hepatocellular carcinoma. Nat Commun. 2018; 9: 4349.

151

Biller LH, Schrag D. Diagnosis and treatment of metastatic colorectal cancer: a review. JAMA. 2021; 325: 669-85.

152

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021; 71: 209-49.

153

Siegel RL, Medhanie GA, Fedewa SA, Jemal A. State variation in early-onset colorectal cancer in the United States, 1995-2015. J Natl Cancer Inst. 2019; 111: 1104-6.

154

Zhang Y, Devocelle A, Souza L, Foudi A, Tenreira Bento S, Desterke C, et al. BMAL1 knockdown triggers different colon carcinoma cell fates by altering the delicate equilibrium between AKT/mTOR and P53/P21 pathways. Aging. 2020; 12: 8067-83.

155

Stokes K, Nunes M, Trombley C, Flôres DEFL, Wu G, Taleb Z, et al. The circadian clock gene, Bmal1, regulates intestinal stem cell signaling and represses tumor initiation. Cell Mol Gastroenterol Hepatol. 2021; 12: 1847-72.

156

Liu JL, Wang CY, Cheng TY, Rixiati Y, Ji C, Deng M, et al. Circadian clock disruption suppresses PDL1 intraepithelial B cells in experimental colitis and colitis-associated colorectal cancer. Cell Mol Gastroenterol Hepatol. 2021; 12: 251-76.

157

Zhang Y, Ma J, Zhang S, Deng G, Wu X, He J, et al. A prognostic analysis of 895 cases of stage Ⅲ colon cancer in different colon subsites. Int J Colorectal Dis. 2015; 30: 1173-83.

158

Huisman SA, Oklejewicz M, Ahmadi AR, Tamanini F, Ijzermans JNM, van der Horst GTJ, et al. Colorectal liver metastases with a disrupted circadian rhythm phase shift the peripheral clock in liver and kidney. Int J Cancer. 2015; 136: 1024-32.

159

Dong P, Wang Y, Liu Y, Zhu C, Lin J, Qian R, et al. BMAL1 induces colorectal cancer metastasis by stimulating exosome secretion. Mol Biol Rep. 2022; 49: 373-84.

160

Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018; 68: 7-30.

161

Dai X, Li T, Bai Z, Yang Y, Liu X, Zhan J, et al. Breast cancer intrinsic subtype classification, clinical use and future trends. Am J Cancer Res. 2015; 5: 2929-43.

162

Xiang S, Mao L, Duplessis T, Yuan L, Dauchy R, Dauchy E, et al. Oscillation of clock and clock controlled genes induced by serum shock in human breast epithelial and breast cancer cells: regulation by melatonin. Breast Cancer (Auckl). 2012; 6: 137-50.

163

Rossetti S, Esposito J, Corlazzoli F, Gregorski A, Sacchi N. Entrainment of breast (cancer) epithelial cells detects distinct circadian oscillation patterns for clock and hormone receptor genes. Cell Cycle. 2012; 11: 350-60.

164

Lesicka M, Jabłońska E, Wieczorek E, Seroczyńska B, Siekierzycka A, Skokowski J, et al. Altered circadian genes expression in breast cancer tissue according to the clinical characteristics. PLoS One. 2018; 13: e0199622.

165

Broadberry E, McConnell J, Williams J, Yang N, Zindy E, Leek A, et al. Disrupted circadian clocks and altered tissue mechanics in primary human breast tumours. Breast Cancer Res. 2018; 20: 125.

166

Korkmaz T, Aygenli F, Emisoglu H, Ozcelik G, Canturk A, Yilmaz S, et al. Opposite carcinogenic effects of circadian clock gene BMAL1. Sci Rep. 2018; 8: 16023.

167

Kwon YJ, Seo EB, Kwon SH, Lee SH, Kim SK, Park SK, et al. Extracellular Acidosis Promotes Metastatic Potency via Decrease of the BMAL1 Circadian Clock Gene in Breast Cancer. Cells. 2020; 9: 989.

168

Sharma D, Kumar S, Narasimhan B. Estrogen alpha receptor antagonists for the treatment of breast cancer: a review. Chem Cent J. 2018; 12: 107.

169

Xie F, Wang L, Liu Y, Liu Z, Zhang Z, Pei J, et al. ASMT regulates tumor metastasis through the circadian clock system in triplenegative breast cancer. Front Oncol. 2020; 10: 537247.

170

Boese AC, Kang S. Tumor progression of breast cancer during hyperinsulinemic obesity. Trends Mol Med. 2020; 26: 354-6.

171

Hambardzumyan D, Gutmann DH, Kettenmann H. The role of microglia and macrophages in glioma maintenance and progression. Nat Neurosci. 2016; 19: 20-7.

172

Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004; 25: 677-86.

173

Verreck FAW, de Boer T, Langenberg DML, Hoeve MA, Kramer M, Vaisberg E, et al. Human IL-23-producing type 1 macrophages promote but IL-10-producing type 2 macrophages subvert immunity to (myco)bacteria. Proc Natl Acad Sci U S A. 2004; 101: 4560-5.

174

Qin C, Zhou LQ, Ma XT, Hu ZW, Yang S, Chen M, et al. Dual functions of microglia in ischemic stroke. Neurosci Bull. 2019; 35: 921-33.

175

Li X, Guan J, Jiang Z, Cheng S, Hou W, Yao J, et al. Microglial exosome miR-7239-3p promotes glioma progression by regulating circadian genes. Neurosci Bull. 2021; 37: 497-510.

176

de Assis LVM, Mendes D, Silva MM, Kinker GS, Pereira-Lima I, Moraes MN, et al. Melanopsin mediates UVA-dependent modulation of proliferation, pigmentation, apoptosis, and molecular clock in normal and malignant melanocytes. Biochim Biophys Acta Mol Cell Res. 2020; 1867: 118789.

177

de Assis LVM, Moraes MN, da Silveira Cruz-Machado S, Castrucci AML. The effect of white light on normal and malignant murine melanocytes: a link between opsins, clock genes, and melanogenesis. Biochim Biophys Acta. 2016; 1863: 1119-33.

178

Zhanfeng N, Yanhui L, Zhou F, Shaocai H, Guangxing L, Hechun X. Circadian genes Per1 and Per2 increase radiosensitivity of glioma in vivo. Oncotarget 2015; 6: 9951-8.

179

Shen H, Cook K, Gee HE, Hau E. Hypoxia, metabolism, and the circadian clock: new links to overcome radiation resistance in highgrade gliomas. J Exp Clin Cancer Res CR. 2020; 39: 129.

180

Wagner PM, Prucca CG, Velazquez FN, Sosa Alderete LG, Caputto BL, Guido ME. Temporal regulation of tumor growth in nocturnal mammals: in vivo studies and chemotherapeutical potential. FASEB J. 2021; 35: e21231.

181

Katamune C, Koyanagi S, Hashikawa KI, Kusunose N, Akamine T, Matsunaga N, et al. Mutation of the gene encoding the circadian clock component PERIOD2 in oncogenic cells confers chemoresistance by up-regulating the Aldh3a1 gene. J Biol Chem. 2019; 294: 547-58.

182

Ferrara N, Hillan KJ, Gerber HP, Novotny W. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nat Rev Drug Discov. 2004; 3: 391-400.

183

De Palma M, Biziato D, Petrova TV. Microenvironmental regulation of tumour angiogenesis. Nat Rev Cancer. 2017; 17: 457-74.

184

Koyanagi S, Kuramoto Y, Nakagawa H, Aramaki H, Ohdo S, Soeda S, et al. A molecular mechanism regulating circadian expression of vascular endothelial growth factor in tumor cells. Cancer Res. 2003; 63: 7277-83.

185

Peek CB, Levine DC, Cedernaes J, Taguchi A, Kobayashi Y, Tsai SJ, et al. Circadian clock interaction with HIF1α mediates oxygenic metabolism and anaerobic glycolysis in skeletal muscle. Cell Metabolism. 2017; 25: 86-92.

186

Ma Z, Jin X, Qian Z, Li F, Xu M, Zhang Y, et al. Deletion of clock gene Bmal1 impaired the chondrocyte function due to disruption of the HIF1α-VEGF signaling pathway. Cell Cycle. 2019; 18: 1473-89.

187

Burgermeister E, Battaglin F, Eladly F, Wu W, Herweck F, Schulte N, et al. Aryl hydrocarbon receptor nuclear translocator-like (ARNTL/ BMAL1) is associated with bevacizumab resistance in colorectal cancer via regulation of vascular endothelial growth factor A. EBioMedicine. 2019; 45: 139-54.

188

Thiery JP, Acloque H, Huang RYJ, Nieto MA. Epithelialmesenchymal transitions in development and disease. Cell. 2009; 139: 871-90.

189

Zhang J, Tian XJ, Xing J. Signal transduction pathways of EMT induced by TGF-β, SHH, and WNT and their crosstalks. J Clin Med. 2016; 5: 41.

190

Jeanes A, Gottardi CJ, Yap AS. Cadherins and cancer: how does cadherin dysfunction promote tumor progression? Oncogene. 2008; 27: 6920-9.

191

Zhang Y, Devocelle A, Desterke C, de Souza LEB, Hadadi É, Acloque H, et al. BMAL1 knockdown leans epithelial-mesenchymal balance toward epithelial properties and decreases the chemoresistance of colon carcinoma cells. Int J Mol Sci. 2021; 22: 5247.

192

Slat EA, Sponagel J, Marpegan L, Simon T, Kfoury N, Kim A, et al. Cell-intrinsic, Bmal1-dependent circadian regulation of temozolomide sensitivity in glioblastoma. J Biol Rhythms. 2017; 32: 121-9.

193

Wagner PM, Sosa Alderete LG, Gorné LD, Gaveglio V, Salvador G, Pasquaré S, et al. Proliferative glioblastoma cancer cells exhibit persisting temporal control of metabolism and display differential temporal drug susceptibility in chemotherapy. Mol Neurobiol. 2019; 56: 1276-92.

194

Jiang W, Zhao S, Shen J, Guo L, Sun Y, Zhu Y, et al. The MiR-135bBMAL1-YY1 loop disturbs pancreatic clockwork to promote tumourigenesis and chemoresistance. Cell Death Dis. 2018; 9: 149.

195

Tang Q, Cheng B, Xie M, Chen Y, Zhao J, Zhou X, et al. Circadian clock gene Bmal1 inhibits tumorigenesis and increases paclitaxel sensitivity in tongue squamous cell carcinoma. Cancer Res. 2017; 77: 532-44.

196

Trebucq LL, Cardama GA, Lorenzano Menna P, Golombek DA, Chiesa JJ, Marpegan L. Timing of novel drug 1A-116 to circadian rhythms improves therapeutic effects against glioblastoma. Pharmaceutics. 2021; 13: 1091.

197

Cardama GA, Gonzalez N, Ciarlantini M, Gandolfi Donadío L, Comin MJ, Alonso DF, et al. Proapoptotic and antiinvasive activity of Rac1 small molecule inhibitors on malignant glioma cells. OncoTargets and Therapy, 2014; 7: 2021-33.

198

Cabrera M, Echeverria E, Lenicov FR, Cardama G, Gonzalez N, Davio C, et al. Pharmacological Rac1 inhibitors with selective apoptotic activity in human acute leukemic cell lines. Oncotarget, 2017; 8: 98509-23.

199

Parri M, Chiarugi P. Rac and Rho GTPases in cancer cell motility control. Cell Commun Signal. 2010; 8: 23.

200

Ridley AJ. Rho GTPase signalling in cell migration. Curr Opin Cell Biol. 2015; 36: 103-12.

201

Teixeira AAS, Biondo LA, Silveira LS, Lima EA, Batatinha HA, Diniz TA, et al. Doxorubicin modulated clock genes and cytokines in macrophages extracted from tumor-bearing mice. Cancer Biol Ther. 2020; 21: 344-53.

202

Mendes MCS, Pimentel GD, Costa FO, Carvalheira JBC. Molecular and neuroendocrine mechanisms of cancer cachexia. J Endocrinol. 2015; 226: R29-43.

203

Kinsey E, Ajazi E, Wang X, Johnston MAM, Crawford J. Predictors of physical and functional loss in advanced-stage lung cancer patients receiving platinum chemotherapy. J Thorac Oncol. 2018; 13: 1294-301.

204

Muliawati Y, Haroen H, Rotty LWA. Cancer anorexia - cachexia syndrome. Acta Med Indones. 2012; 44: 154-62.

205

von Haehling S, Anker SD. Cachexia as major underestimated unmet medical need: facts and numbers. Int J Cardiol. 2012; 161: 121-3.

206

Colldén G, Tschöp MH, Müller TD. Therapeutic potential of targeting the ghrelin pathway. Int J Mol Sci. 2017; 18: 798.

207

Chen JA, Splenser A, Guillory B, Luo J, Mendiratta M, Belinova B, et al. Ghrelin prevents tumour- and cisplatin-induced muscle wasting: characterization of multiple mechanisms involved. J Cachexia Sarcopenia Muscle. 2015; 6: 132-3.

208

Hatanaka M, Konishi M, Ishida J, Saito M, Springer J. Novel mechanism of ghrelin therapy for cachexia. J Cachexia Sarcopenia Muscle. 2015; 6: 393.

209

Lundholm K, Gunnebo L, Körner U, Iresjö BM, Engström C, Hyltander A, et al. Effects by daily long term provision of ghrelin to unselected weight-losing cancer patients: a randomized doubleblind study. Cancer. 2010; 116: 2044-52.

210

Doruk YU, Yarparvar D, Akyel YK, Gul S, Taskin AC, Yilmaz F, et al. A CLOCK-binding small molecule disrupts the interaction between CLOCK and BMAL1 and enhances circadian rhythm amplitude. J Biol Chem. 2020; 295: 3518-31.

211

Saxton RA, Sabatini DM. mTOR signaling in growth, metabolism, and disease. Cell. 2017; 169: 361-71.

212

Shimobayashi M, Hall MN. Making new contacts: the mTOR network in metabolism and signalling crosstalk. Nat Rev Mol Cell Biol. 2014; 15: 155-62.

213

González A, Hall MN. Nutrient sensing and TOR signaling in yeast and mammals. EMBO J. 2017; 36: 397-408.

214

Hay N, Sonenberg N. Upstream and downstream of mTOR. Genes Dev. 2004; 18: 1926-45.

215

Matsumoto CS, Almeida LO, Guimarães DM, Martins MD, Papagerakis P, Papagerakis S, et al. PI3K-PTEN dysregulation leads to mTOR-driven upregulation of the core clock gene BMAL1 in normal and malignant epithelial cells. Oncotarget. 2016; 7: 42393-407.

216

Bobrovnikova-Marjon E, Grigoriadou C, Pytel D, Zhang F, Ye J, Koumenis C, et al. PERK promotes cancer cell proliferation and tumor growth by limiting oxidative DNA damage. Oncogene. 2010; 29: 3881-95.

217

Pytel D, Gao Y, Mackiewicz K, Katlinskaya YV, Staschke KA, Paredes MCG, et al. PERK is a haploinsufficient tumor suppressor: gene dose determines tumor-suppressive versus tumor promoting properties of PERK in melanoma. PLoS Genet. 2016; 12: e1006518.

218

Bhattacharya S, HuangFu WC, Dong G, Qian J, Baker DP, Karar J, et al. Anti-tumorigenic effects of Type 1 interferon are subdued by integrated stress responses. Oncogene. 2013; 32: 4214-21.

219

Lee Y, Fong SY, Shon J, Zhang SL, Brooks R, Lahens NF, et al. Timeof-day specificity of anticancer drugs may be mediated by circadian regulation of the cell cycle. Sci. Adv. 2021; 7: eabd2645.

220

Isakoff MS, Bielack SS, Meltzer P, Gorlick R. Osteosarcoma: current treatment and a collaborative pathway to success. J Clin Oncol. 2015; 33: 3029-35.

221

Tang QL, Xie XB, Wang J, Chen Q, Han AJ, Zou CY, et al. Glycogen synthase kinase-3β, NF-κB signaling, and tumorigenesis of human osteosarcoma. J Natl Cancer Ins. 2012; 104: 749-63.

222

Liu HC, Enikolopov G, Chen Y. Cul4B regulates neural progenitor cell growth. BMC Neurosci. 2012; 13: 112.

223

Soucy TA, Smith PG, Milhollen MA, Berger AJ, Gavin JM, Adhikari S, et al. An inhibitor of NEDD8-activating enzyme as a new approach to treat cancer. Nature. 2009; 458: 732-6.

224

Nawrocki ST, Griffin P, Kelly KR, Carew JS. MLN4924: a novel firstin-class inhibitor of NEDD8-activating enzyme for cancer therapy. Expert Opin Investig Drugs. 2012; 21: 1563-73.

225

Khalife J, Radomska HS, Santhanam R, Huang X, Neviani P, Saultz J, et al. Pharmacological targeting of miR-155 via the NEDD8- activating enzyme inhibitor MLN4924 (Pevonedistat) in FLT3-ITD acute myeloid leukemia. Leukemia. 2015; 29: 1981-92.

226

Chen P, Hu T, Liang Y, Jiang Y, Pan Y, Li C, et al. Synergistic inhibition of autophagy and neddylation pathways as a novel therapeutic approach for targeting liver cancer. Oncotarget. 2015; 6: 9002-17.

227

Kuo KL, Ho IL, Shi CS, Wu JT, Lin WC, Tsai YC, et al. MLN4924, a novel protein neddylation inhibitor, suppresses proliferation and migration of human urothelial carcinoma: In vitro and in vivo studies. Cancer Lett. 2015; 363: 127-36.

228

Zhang S, Zhang J, Deng Z, Liu H, Mao W, Jiang F, et al. Circadian clock components RORα and Bmal1 mediate the anti-proliferative effect of MLN4924 in osteosarcoma cells. Oncotarget. 2016; 7: 66087-99.

229

Yu X, Rollins D, Ruhn KA, Stubblefield JJ, Green CB, Kashiwada M, et al. TH17 cell differentiation is regulated by the circadian clock. Science. 2013; 342: 727-30.

230

Hand LE, Gray KJ, Dickson SH, Simpkins DA, Ray DW, Konkel JE, et al. Regulatory T cells confer a circadian signature on inflammatory arthritis. Nat Commun. 2020; 11: 1658.

231

Hu X, Liu X, Moisan J, Wang Y, Lesch CA, Spooner C, et al. Synthetic RORγ agonists regulate multiple pathways to enhance antitumor immunity. Oncoimmunology. 2016; 5: e1254854.

232

Mullins D, Proulx D, Saoudi A, Ng CE. Chronomodulation of topotecan or X-radiation treatment increases treatment efficacy without enhancing acute toxicity. Int J Radiat Oncol Biol Phys. 2005; 62: 230-7.

233

Ozturk N, Ozturk D, Pala-Kara Z, Kaptan E, Sancar-Bas S, Ozsoy N, et al. The immune system as a chronotoxicity target of the anticancer mTOR inhibitor everolimus. Chronobiol Int. 2018; 35: 705-s18.

Cancer Biology & Medicine
Pages 1-24
Cite this article:
Huang C, Zhang C, Cao Y, et al. Major roles of the circadian clock in cancer. Cancer Biology & Medicine, 2023, 20(1): 1-24. https://doi.org/10.20892/j.issn.2095-3941.2022.0474

119

Views

4

Downloads

18

Crossref

14

Web of Science

15

Scopus

Altmetrics

Received: 08 August 2022
Accepted: 04 November 2022
Published: 12 January 2023
©2023 Cancer Biology & Medicine.

Creative Commons Attribution-NonCommercial 4.0 International License

Return