#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

The Catalytic and Non-catalytic Functions of the Chromatin-Remodeling Protein Collaborate to Fine-Tune Circadian Transcription in


The circadian clock is an endogenous timing system that enables organisms to anticipate daily changes in their external environment and temporally coordinate key biological functions that are important to their survival. Central to Drosophila clockwork is a key transcription factor CLOCK (CLK). CLK activates expression of target genes only during specific parts of the day, thereby orchestrating rhythmic expression of hundreds of clock-controlled genes, which consequently manifest into daily rhythms in physiology and behavior. In this study, we demonstrated that the Brahma (Brm) chromatin-remodeling protein interacts with CLK and fine-tune the levels of CLK-dependent transcription to maintain the robustness of the circadian clock. Specifically, we uncovered two distinct but collaborative functions of Brm. Brm possesses a non-catalytic function that negatively regulates the binding of CLK to target genes and limits transcriptional output, likely by recruiting repressive protein complexes. Catalytically, Brm functions by condensing the chromatin at CLK target genes, specifically when transcription is active. This serves to precisely control the level of repressive factors likely recruited by Brm as well as other transcriptional regulators. By disentangling these two roles of Brm, our study uncovered a multi-layered mechanism in which a chromatin remodeler regulates the circadian clock.


Vyšlo v časopise: The Catalytic and Non-catalytic Functions of the Chromatin-Remodeling Protein Collaborate to Fine-Tune Circadian Transcription in. PLoS Genet 11(7): e32767. doi:10.1371/journal.pgen.1005307
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1005307

Souhrn

The circadian clock is an endogenous timing system that enables organisms to anticipate daily changes in their external environment and temporally coordinate key biological functions that are important to their survival. Central to Drosophila clockwork is a key transcription factor CLOCK (CLK). CLK activates expression of target genes only during specific parts of the day, thereby orchestrating rhythmic expression of hundreds of clock-controlled genes, which consequently manifest into daily rhythms in physiology and behavior. In this study, we demonstrated that the Brahma (Brm) chromatin-remodeling protein interacts with CLK and fine-tune the levels of CLK-dependent transcription to maintain the robustness of the circadian clock. Specifically, we uncovered two distinct but collaborative functions of Brm. Brm possesses a non-catalytic function that negatively regulates the binding of CLK to target genes and limits transcriptional output, likely by recruiting repressive protein complexes. Catalytically, Brm functions by condensing the chromatin at CLK target genes, specifically when transcription is active. This serves to precisely control the level of repressive factors likely recruited by Brm as well as other transcriptional regulators. By disentangling these two roles of Brm, our study uncovered a multi-layered mechanism in which a chromatin remodeler regulates the circadian clock.


Zdroje

1. Hardin PE, Panda S. Circadian timekeeping and output mechanisms in animals. Curr Opin Neurobiol. 2013;23: 724–31. doi: 10.1016/j.conb.2013.02.018 23731779

2. Hsu PY, Harmer SL. Wheels within wheels: the plant circadian system. Trends Plant Sci. 2013;19: 240–9. doi: 10.1016/j.tplants.2013.11.007 24373845

3. Baker CL, Loros JJ, Dunlap JC. The circadian clock of Neurospora crassa. FEMS Microbiol Rev. 2012;36: 95–110. doi: 10.1111/j.1574-6976.2011.00288.x 21707668

4. Cyran SA, Buchsbaum AM, Reddy KL, Lin MC, Glossop NR, Hardin PE, et al. Vrille, Pdp1, and dClock form a second feedback loop in the Drosophila circadian clock. Cell. 2003;112: 329–41. 12581523

5. Glossop NR, Houl JH, Zheng H, Ng FS, Dudek SM, Hardin PE. WRILLE feeds back to control circadian transcription of Clock in the Drosophila circadian oscillator. Neuron. 2003;37: 249–61. 12546820

6. Martinek S, Inonog S, Manoukian AS, Young MW. A role for the segment polarity gene shaggy/GSK-3 in the Drosophila circadian clock. Cell. 2001;105: 769–79. 11440719

7. Sathyanarayanan S, Zheng X, Xiao R, Sehgal A. Posttranslational regulation of Drosophila PERIOD protein by protein phosphatase 2A. Cell. 2004;116: 603–15. 14980226

8. Cyran SA, Yiannoulos G, Buchsbaum AM, Saez L, Young MW, Blau J. The double-time protein kinase regulates the subcellular localization of the Drosophila clock protein. J Neurosci. 2005;25: 5430–7. 15930393

9. Fang Y, Sathyanarayanan S, Seghal A. Post-translational regulation of the Drosophila circadian clock requires protein phosphatase 1 (PP1). Genes Dev. 2007;21: 1506–18. 17575052

10. Chiu JC, Ko HW, Edery I. NEMO/NLK phosphorylates PERIOD to initiate a time-delay phosphorylation circuit that sets circadian clock speed. Cell. 2011;145: 357–70. doi: 10.1016/j.cell.2011.04.002 21514639

11. Kim EY, Jeong EH, Park S, Jeong HJ, Edery I, Cho JW. A role for O-GlcNAcylation in setting circadian clock speed. Genes Dev. 2012;26: 490–502. doi: 10.1101/gad.182378.111 22327476

12. Lim C, Allada R. ATAXIN-2 activates PERIOD translation to sustain circadian rhythms in Drosophila. Science. 2013;340: 875–9. doi: 10.1126/science.1234785 23687047

13. Zhang Y, Ling J, Yuan C, Dubruille R, Emery P. A role for Drosophila ATX2 in activation of PER translation and circadian behavior. Science. 2013;340: 879–882. doi: 10.1126/science.1234746 23687048

14. Koh K, Zheng X, Sehgal A. JETLAG resets the Drosophila circadian clock by promoting light-induced degradation of TIMELESS. Science. 2006;312: 1809–1812. 16794082

15. Ko HW, Jiang J, Edery I. A role for Slimb in the degradation of Drosophila PERIOD protein phosphorylated by DOUBLETIME. Nature. 2002;420: 673–678. 12442174

16. Grima B, Lamouroux A, Chelot E, Papin C, Limbourg-Bouchon B, Rouyer F. The F-box protein Slimb controls the levels of clock proteins Period and Timeless. Nature. 2002;420: 178–82. 12432393

17. Lim C, Chung BY, Pitman JL, McGill JJ, Pradhan S, Lee J, et al. clockwork orange encodes a transcriptional repressor important for circadian-clock amplitude in Drosophila. Curr Biol. 2007;17: 1802–9.

18. Kadener S, Stoleru D, McDonald M, Nawathean P, Rosbash M. Clockwork Orange is a transcriptional repressor and a new Drosophila circadian pacemaker component. Genes Dev. 2007;21: 1675–86. 17578907

19. Hung HC, Maurer C, Kay SA, Weber F. Circadian transcription depends on limiting amounts of the transcription co-activator nejire/CBP. J Biol Chem. 2007;282: 31349–57. 17635913

20. Lim C, Lee J, Choi C, Kim J, Doh E, Choe J. Functional role of CREB-binding protein in the circadian clock system of Drosophila melanogaster. Mol Cell Biol. 2007;27: 4876–90. 17452464

21. Abruzzi KC, Rodriguez J, Menet JS, Desrochers J, Zadina A, Luo W, et al. Drosophila CLOCK target gene characterization: implications for circadian tissue-specific gene expression. Genes Dev. 2011;25: 2374–2386. doi: 10.1101/gad.174110.111 22085964

22. Crosio C, Cermakian N, Allis CD, Sassone-Corsi P. Light induces chromatin modification in cells of the mammalian circadian clock. Nat Neurosci. 2000;3: 1241–7. 11100144

23. Etchegaray JP, Lee C, Wade PA, Reppert SM. Rhythmic histone acetylation underlies transcription in the mammalian clock. Nature. 2003;421: 177–82. 12483227

24. Eckel-Mahan K, Sassone-Corsi P. Epigenetic regulation of the molecular clockwork. Prog Mol Biol Transl Sci. 2013;119: 29–50. doi: 10.1016/B978-0-12-396971-2.00002-6 23899593

25. Barneche F, Malapeira J, Mas P. The impact of chromatin dynamics on plant night responses and circadian clock function. J Exp Biol. 2014; 63: 2895–913.

26. Taylor P, Hardin PE. Rhythmic E-Box binding by CLK-CYC controls daily cycles in per and tim transcription and chromatin modifications. Mol Cell Biol. 2008;28: 4642–52. doi: 10.1128/MCB.01612-07 18474612

27. Koike N, Yoo SH, Huang HC, Kumar V, Lee C, Kim TK, et al. Transcriptional architecture and chromatin landscape of the core circadian clock in mammals. Science. 2012;338: 349–54. doi: 10.1126/science.1226339 22936566

28. Menet JS, Pescatore S, Rosbash M. CLOCK:BMAL1 is a pioneer-like transcription factor. Genes Dev. 2014;28: 8–13. doi: 10.1101/gad.228536.113 24395244

29. Clapier CR, Cairns BR. The biology of chromatin remodeling complexes. Annu Rev Biochem. 2009;78: 273–304. doi: 10.1146/annurev.biochem.77.062706.153223 19355820

30. Hota SK, Bartholomew B. Diversity of operation in ATP-dependent chromatin remodelers. Biochim Biophys Acta. 2011;1809: 476–87. doi: 10.1016/j.bbagrm.2011.05.007 21616185

31. Petty E, Pillus L. Balancing chromatin remodeling and histone modifications in transcription. Trends Genet. 2013;29: 621–9. doi: 10.1016/j.tig.2013.06.006 23870137

32. Belden WJ, Loros JJ, Dunlap JC. Execution of the circadian negative feedback loop in Neurospora requires the ATP-dependent chromatin-remodeling enzyme CLOCKSWITCH. Mol Cell. 2007;25: 587–600. 17317630

33. Belden WJ, Lewis ZA, Selker EU, Loros JJ, Dunlap JC. CHD1 remodels chromatin and influences transient DNA methylation at the clock gene frequency. PLoS Genet. 2011;7: e1002166. doi: 10.1371/journal.pgen.1002166 21811413

34. Cha J, Zhou M, Liu Y. CATP is a critical component of the Neurospora circadian clock by regulating the nucleosome occupancy rhythm at the frequency locus. EMBO Rep. 2013;14: 923–30. doi: 10.1038/embor.2013.131 23958634

35. Wang B, Kettenbach AN, Gerber SA, Loros JJ, Dunlap JC. Neurospora WC-1 recruits SWI/SNF to remodel frequency and initiate a circadian cycle. PLoS Genet. 2014;10: e1004599. doi: 10.1371/journal.pgen.1004599 25254987

36. Dubruille R, Murad A, Rosbash M, Emery P. A constant light-genetic screen identifies KISMET as a regulator of circadian photoresponses. PLoS Genet. 2009;5: e1000787. doi: 10.1371/journal.pgen.1000787 20041201

37. Tamkun JW, Deuring R, Scott MP, Kissinger M, Pattatucci AM, Laufman TC, et al. brahma: a regulator of Drosophila homeotic genes structurally related to the yeast transcriptional activator SNF2/SWI2. Cell. 1992;68: 561–572. 1346755

38. Zraly CB, Marenda DR, Nanchal R, Cavalli G, Muchardt C, Dingwall AK. SNR1 is an essential subunit in a subset of Drosophila brm complexes, targeting specific functions during development. Dev Biol. 2003;253: 291–308. 12645932

39. Dhalluin C, Carlson JE, Zeng L, He C, Aggarwal AK, Zhou MM. Structure and ligand of a histone acetyltransferase bromodomain. Nature. 1999;399: 491–6. 10365964

40. Owen DJ, Omaghi P, Yang JC, Lowe N, Evans PR, Ballario P, et al. The structural basis for the recognition of acetyl histone H4 by the bromodomain of histone acetyltransferase gnc5p. EMBO J. 2000;19: 6141–6149. 11080160

41. Marenda DR, Zraly CB, Dingwall AK. The Drosophila Brahma (SWI/SNF) chromatin remodeling complex exhibits cell-type specific activation and repression functions. Dev Biol. 2004;267: 279–93. 15013794

42. Bae K, Lee C, Hardin PE, Edery I. dCLOCK is present in limiting amounts and likely mediates daily interactions between the dCLOCK-CYC transcription factor and the PER-TIM complex. J Neurosci. 2000;20: 1746–53. 10684876

43. Kilman VL and Allada R. Genetic analysis of ectopic circadian clock induction in Drosophila. J Biol Rhythms. 2009;24: 368–378. doi: 10.1177/0748730409343761 19755582

44. Darlington TK, Wager-Smith K, Ceriani MF, Staknis D, Gekakis N, Steeves TD, et al. Closing the circadian loop: CLOCK-induced transcription of its own inhibitors per and tim. Science. 1998;280: 1599–603. 9616122

45. Blau J, Young MW. Cycling vrille expression is required for a functional Drosophila clock. Cell. 1999;99: 661–71. 10612401

46. Yu W, Zheng H, Houl JH, Daudwalder B, Hardin PE. PER-dependent rhythms in CLK phosphorylation and E-box binding regulate circadian transcription. Genes Dev. 2006;20: 723–733. 16543224

47. Mahesh G, Jeong E, Ng FS, Liu Y, Gunawardhana K, Houl JH, et al. Phosphorylation of the transcriptional activator CLOCK regulates progression through a ~24-h feedback loops to influence the circadian period in Drosophila. J Biol Chem. 2014;289: 19681–93. doi: 10.1074/jbc.M114.568493 24872414

48. Hung MC, Maurer C, Zorn D, Chang WL, Weber F. Sequential and compartment-specific phosphorylation controls the life cycle of the circadian CLOCK protein. J Biol Chem. 2009;284: 23734–42. doi: 10.1074/jbc.M109.025064 19564332

49. Marenda DR, Zraly CB, Feng Y, Egan S, Dingwall AK. The Drosophila SNF1 (SNF5/INI1) subunit directs essential developmental functions of the Brahma chromatin remodeling complex. Mol Cell Biol. 2003;23: 189–305.

50. Elfring LK, Daniel C, Papoulas O, Deuring R, Sarte M, Moseley S, et al. Genetic analysis of brahma: the Drosophila homolog of the yeast chromatin remodeling factor SWI2/SNF2. Genetics. 1998;148: 251–265. 9475737

51. Menet JS, Abruzzi KC, Desrochers J, Rodriguez J, and Rosbash M. Dynamic PER repression mechanisms in the Drosophila circadian clock: from on-DNA to off-DNA. Genes Dev. 2010;24: 358–67. doi: 10.1101/gad.1883910 20159956

52. Hao H, Allen DL, Hardin PE. A circadian enhancer mediates PER-dependent mRNA cycling in Drosophila melanogaster. Mol Cell Biol. 1997;17: 3687–93. 9199302

53. Hao H, Glossop NR, Lyons L, Qiu J, Morrish B, Cheng Y, et al. The 69 bp circadian regulatory sequence (CRS) mediates per-like developmental, spatial, and circadian expression and behavioral rescue in Drosophila. J Neurosci. 1999;19: 987–94. 9920662

54. Zraly CB and Dingwall AK. The chromatin remodeling and mRNA splicing functions of the Brahma (SWI/SNF) complex are mediated by the SNR1/SNF5 regulatory subunit. Nucleic Acids Res. 2012;40: 5975–87. doi: 10.1093/nar/gks288 22467207

55. Tolstorukov MY, Sansam CG, Lu P, Koellhoffer EC, Helming KC, Alver BH, et al. Swi/Snf chromatin remodeling/tumor suppressor complex establishes nucleosome occupancy at target promoters. Proc Natl Acad Sci USA. 2013;110: 10165–70. doi: 10.1073/pnas.1302209110 23723349

56. Zraly CB, Middleton FA, Dingwall AK. Hormone-response genes are direct in vivo regulatory targets of Brahma (SWI/SNF) complex function. J Biol Chem. 2006;281: 35305–15. 16990270

57. Curtis BJ, Zraly CB, Marenda DR, Dingwall AK. Histone lysine demethylases function as co-repressors of SWI/SNF remodeling activities during Drosophila wing development. Dev Biol. 2011;350: 534–47. doi: 10.1016/j.ydbio.2010.12.001 21146519

58. Thurman RE, Rynes E, Humbert R, Vierstra J, Maurano MT, Haugen E, et al. The accessible chromatin landscape of the human genome. Nature. 2012;489: 75–82. doi: 10.1038/nature11232 22955617

59. Curtin KD, Huang ZJ, Rosbash M. Temporally regulated nuclear entry of the Drosophila period protein contributes to the circadian clock. Neuron. 1995;14: 365–72. 7857645

60. So WV and Rosbash M. Post-transcriptional regulation contributes to Drosophila clock gene mRNA cycling. EMBO. 1997;16: 7146–55.

61. McDonald MJ, Rosbash M. Microarray analysis and organization of circadian gene expression in Drosophila. Cell. 2001;107: 567–578. 11733057

62. Vorobyeva NE, Nikolenko JV, Nabirochkina EN, Krasnov AN, Shidlovskii YV, Georgieva SG. SAYP and Brahma are important for ‘repressive’ and ‘transient’ Pol II pausing. Nucleic Acids Res. 2012;40: 7319–31. doi: 10.1093/nar/gks472 22638575

63. Adelman K, Lis JT. Promoter-proximal pausing of RNA polymerase II: emerging roles in metazoans. Nat Rev Genet. 2012;13: 720–31. doi: 10.1038/nrg3293 22986266

64. Gilchrist DA, Dos Santos G, Fargo DC, Xie B, Gao Y, Li L, et al. Pausing of RNA polymerase II disrupts DNA-specified nucleosome organization to enable precise gene regulation. Cell. 2010;143: 540–551. doi: 10.1016/j.cell.2010.10.004 21074046

65. Czudnochowski N, Bosken CA, Geyer M. Serine-7 but not serine-5 phosphorylation primes RNA polymerase II CTD for P-TEFb recognition. Nat Commun. 2012;3: 842. doi: 10.1038/ncomms1846 22588304

66. Tyagi A, Ryme J, Brodin D, Ostlund Farrants AK, Visa N. SWI/SNF associates with nascent pre-mRNPs and regulates alternative pre = mRNA processing. PLoS Genet. 2009;5: e1000470. doi: 10.1371/journal.pgen.1000470 19424417

67. Cheng Y, Gvakharia B, Hardin PE. Two alternatively spliced transcripts from the Drosophila period gene rescue rhythms having different molecular and behavioral characteristics. Mol Cell Biol. 1998;18: 6505–14. 9774666

68. Majercak J, Sidote D, Hardin PE, Edery I. How a circadian clock adapts to seasonal decrease in temperature and day length. Neuron. 1999;24: 219–30. 10677039

69. Mavrich TN, Jiang C, Ioshikhes IP, Li X, Venters BJ, Zanton SJ, et al. Nucleosome organization in the Drosophila genome. Nature. 2008;453: 358–364. doi: 10.1038/nature06929 18408708

70. Jiang C, Pugh BF. Nucleosome positioning and gene regulation: advances through genomics. Nat Rev Genetics. 2009;10: 161–172.

71. Underhill C, Qutob MS, Yee SP, Torchia J. A novel nuclear receptor corerepressor complex, N-CoR, contains components of the mammalian SWI/SNF complex and the corepressor KAP-1. J Biol Chem. 2000;275: 40463–70. 11013263

72. Sif S, Saurin AJ, Imbalzano AN, Kingston RE. Purification and characterization of mSin3A-containing BRG1 and hBRM chromatin remodeling complexes. Genes Dev. 2001;15: 603–18. 11238380

73. Kuzmichev A, Zhang Y, Erdjument-Bromage H, Tempst P, Reinberg D. Role of the Sin3-histone deacetylase complex in growth regulation by the candidate tumor suppressor p33(ING1). Mol Cell Biol. 2002;22: 835–48. 11784859

74. Metivier R, Penot G, Hubner MR, Reid G, Brand H, Kos M, et al. Estrogen Receptor-alpha directs ordered, cyclical, and combinatorial recruitment of cofactors on a natural target promoter. Cell. 2003;115: 751–763. 14675539

75. Tie F, Banerjee R, Conrad PA, Scacheri PC, Harte PJ. Histone demethylase UTX and chromatin remodeler BRM bind directly to CBP and modulate acetylation of histone H3 lysine 27. Mol Cell Biol. 2012;32: 2323–2334. doi: 10.1128/MCB.06392-11 22493065

76. Shi J, Zheng M, Ye Y, Li M, Chen X, Hu X, et al. Drosophila Brahma complex remodels nucleosome organizations in multiple aspects. Nucleic Acids Res. 2014;42: 9730–9. doi: 10.1093/nar/gku717 25081211

77. Zhang Y, Iratni R, Erdjument-Bromage H, Tempst P, Reinberg D. Histone deacetylases and SAP18, a novel polypeptide, are components of a human Sin3 complex. Cell. 1997;89: 357–64 9150135

78. Duong HA, Robles MS, Knutti D, Weitz CJ. A molecular mechanism for circadian clock negative feedback. Science. 2011;332: 1436–39. doi: 10.1126/science.1196766 21680841

79. Gilchrist DA, Nechaez V, Lee C, Ghosh SK, Collins JB, Li L, et al. NELF-mediated stalling of Pol II can enhance gene expression by blocking promoter-proximal nucleosome assembly. Genes Dev. 2008;22: 1921–33 doi: 10.1101/gad.1643208 18628398

80. Skene PJ, Hernandez AE, Groudine M, Henikoff S. The nucleosome barrier to promoter escape by RNA polymerase II is overcome by the chromatin remodeler Chd1. Elife. 2014;3: e02042. doi: 10.7554/eLife.02042 24737864

81. Brand AH, Perrimon N. Target gene expression as a means of altering cell fates and generating dominant phenotypes. Development. 1993;118: 401–15. 8223268

82. Chiu JC, Low KH, Pike DH, Yildirim E, Edery I. Assaying locomoter activity to study circadian rhythms and sleep parameters in Drosophila. J Vis Exp. 2010;43: 2157. doi: 10.3791/2157 20972399

83. Kim EY, Ko HW, Yu W, Hardin PE, Edery I. DOUBLETIME kinase binding domain on the Drosophila PERIOD protein is essential for its hyperphosphorylation, transcriptional repression, and circadian clock function. Mol Cell Biol. 2007;27: 5014–28. 17452449

84. Kim EY, Edery I. Balance between DBT/CKIε kinase and protein phosphatase activities regulate phosphorylation and stability of Drosophila CLOCK protein. Proc Natl Acad Sci USA. 2006;103: 6178–83. 16603629

85. Sidote D, Majercak J, Parikh V, Edery I. Differential effects on light and heat on Drosophila circadian clock proteins PER and TIM. Mol Cell Biol. 1998;18: 2004–13. 9528772

86. Hamby KA, Kwok RS, Zalom FG, Chiu JC. Integrating circadian activity and gene expression profiles to predict chronotoxicity of Drosophila suzukii response to insecticides. PLoS ONE. 2013;8: e68472. doi: 10.1371/journal.pone.0068472 23861907

87. Khodor YL, Rodriguez J, Abruzzi KC, Tang CH, Marr MT II, Rosbash M. Nascent-seq indicates widespread cotranscriptional pre-mRNA splicing in Drosophila. Genes Dev. 2011;25: 2502–12. doi: 10.1101/gad.178962.111 22156210

88. Menet JS, Rodriguez J, Abruzzi KC, Rosbash M. Nascent-Seq reveals novel features of mouse circadian transcriptional regulation. eLife. 2012;1: e00011. doi: 10.7554/eLife.00011 23150795

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2015 Číslo 7
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#