#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

RNA Polymerase II CTD phosphatase Rtr1 fine-tunes transcription termination


Autoři: Jose F. Victorino aff001;  Melanie J. Fox aff001;  Whitney R. Smith-Kinnaman aff001;  Sarah A. Peck Justice aff001;  Katlyn H. Burriss aff001;  Asha K. Boyd aff001;  Megan A. Zimmerly aff001;  Rachel R. Chan aff001;  Gerald O. Hunter aff001;  Yunlong Liu aff002;  Amber L. Mosley aff001
Působiště autorů: Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America aff001;  Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America aff002;  Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America aff003
Vyšlo v časopise: RNA Polymerase II CTD phosphatase Rtr1 fine-tunes transcription termination. PLoS Genet 16(3): e32767. doi:10.1371/journal.pgen.1008317
Kategorie: Research Article
doi: https://doi.org/10.1371/journal.pgen.1008317

Souhrn

RNA Polymerase II (RNAPII) transcription termination is regulated by the phosphorylation status of the C-terminal domain (CTD). The phosphatase Rtr1 has been shown to regulate serine 5 phosphorylation on the CTD; however, its role in the regulation of RNAPII termination has not been explored. As a consequence of RTR1 deletion, interactions within the termination machinery and between the termination machinery and RNAPII were altered as quantified by Disruption-Compensation (DisCo) network analysis. Of note, interactions between RNAPII and the cleavage factor IA (CF1A) subunit Pcf11 were reduced in rtr1Δ, whereas interactions with the CTD and RNA-binding termination factor Nrd1 were increased. Globally, rtr1Δ leads to decreases in numerous noncoding RNAs that are linked to the Nrd1, Nab3 and Sen1 (NNS) -dependent RNAPII termination pathway. Genome-wide analysis of RNAPII and Nrd1 occupancy suggests that loss of RTR1 leads to increased termination at noncoding genes. Additionally, premature RNAPII termination increases globally at protein-coding genes with a decrease in RNAPII occupancy occurring just after the peak of Nrd1 recruitment during early elongation. The effects of rtr1Δ on RNA expression levels were lost following deletion of the exosome subunit Rrp6, which works with the NNS complex to rapidly degrade a number of noncoding RNAs following termination. Overall, these data suggest that Rtr1 restricts the NNS-dependent termination pathway in WT cells to prevent premature termination of mRNAs and ncRNAs. Rtr1 facilitates low-level elongation of noncoding transcripts that impact RNAPII interference thereby shaping the transcriptome.

Klíčová slova:

Messenger RNA – Non-coding RNA sequences – Phosphatases – RNA sequencing – Transcriptional control – Transcriptional termination – Yeast – ChIP exo


Zdroje

1. Herzel L, Straube K, Neugebauer KM. Long-read sequencing of nascent RNA reveals coupling among RNA processing events. Genome Res. 2018;28(7):1008–19. Epub 2018/06/16. doi: 10.1101/gr.232025.117 29903723; PubMed Central PMCID: PMC6028129.

2. Peck SA, Hughes KD, Victorino JF, Mosley AL. Writing a wrong: Coupled RNA polymerase II transcription and RNA quality control. Wiley Interdiscip Rev RNA. 2019;10(4):e1529. Epub 2019/03/09. doi: 10.1002/wrna.1529 30848101; PubMed Central PMCID: PMC6570551.

3. Kamieniarz-Gdula K, Proudfoot NJ. Transcriptional Control by Premature Termination: A Forgotten Mechanism. Trends Genet. 2019. Epub 2019/06/20. doi: 10.1016/j.tig.2019.05.005 31213387.

4. Kamieniarz-Gdula K, Gdula MR, Panser K, Nojima T, Monks J, Wisniewski JR, et al. Selective Roles of Vertebrate PCF11 in Premature and Full-Length Transcript Termination. Mol Cell. 2019;74(1):158–72 e9. Epub 2019/03/02. doi: 10.1016/j.molcel.2019.01.027 30819644; PubMed Central PMCID: PMC6458999.

5. Schlackow M, Nojima T, Gomes T, Dhir A, Carmo-Fonseca M, Proudfoot NJ. Distinctive Patterns of Transcription and RNA Processing for Human lincRNAs. Mol Cell. 2017;65(1):25–38. Epub 2016/12/27. doi: 10.1016/j.molcel.2016.11.029 28017589; PubMed Central PMCID: PMC5222723.

6. Dhir A, Dhir S, Proudfoot NJ, Jopling CL. Microprocessor mediates transcriptional termination of long noncoding RNA transcripts hosting microRNAs. Nat Struct Mol Biol. 2015;22(4):319–27. Epub 2015/03/03. doi: 10.1038/nsmb.2982 25730776; PubMed Central PMCID: PMC4492989.

7. Wagschal A, Rousset E, Basavarajaiah P, Contreras X, Harwig A, Laurent-Chabalier S, et al. Microprocessor, Setx, Xrn2, and Rrp6 co-operate to induce premature termination of transcription by RNAPII. Cell. 2012;150(6):1147–57. Epub 2012/09/18. doi: 10.1016/j.cell.2012.08.004 22980978; PubMed Central PMCID: PMC3595997.

8. Fusby B, Kim S, Erickson B, Kim H, Peterson ML, Bentley DL. Coordination of RNA Polymerase II Pausing and 3' End Processing Factor Recruitment with Alternative Polyadenylation. Molecular and cellular biology. 2016;36(2):295–303. Epub 2015/11/04. doi: 10.1128/MCB.00898-15 26527620; PubMed Central PMCID: PMC4719304.

9. Brody Y, Neufeld N, Bieberstein N, Causse SZ, Bohnlein EM, Neugebauer KM, et al. The in vivo kinetics of RNA polymerase II elongation during co-transcriptional splicing. PLoS Biol. 2011;9(1):e1000573. Epub 2011/01/26. doi: 10.1371/journal.pbio.1000573 21264352; PubMed Central PMCID: PMC3019111.

10. Liu X, Freitas J, Zheng D, Oliveira MS, Hoque M, Martins T, et al. Transcription elongation rate has a tissue-specific impact on alternative cleavage and polyadenylation in Drosophila melanogaster. Rna. 2017;23(12):1807–16. Epub 2017/08/31. doi: 10.1261/rna.062661.117 28851752; PubMed Central PMCID: PMC5689002.

11. Corden JL. Tails of RNA polymerase II. Trends Biochem Sci. 1990;15(10):383–7. Epub 1990/10/01. doi: 10.1016/0968-0004(90)90236-5 2251729.

12. Buratowski S. Progression through the RNA polymerase II CTD cycle. Mol Cell. 2009;36(4):541–6. Epub 2009/11/28. doi: 10.1016/j.molcel.2009.10.019 19941815; PubMed Central PMCID: PMC3232742.

13. Mosley AL, Pattenden SG, Carey M, Venkatesh S, Gilmore JM, Florens L, et al. Rtr1 is a CTD phosphatase that regulates RNA polymerase II during the transition from serine 5 to serine 2 phosphorylation. Mol Cell. 2009;34(2):168–78. doi: 10.1016/j.molcel.2009.02.025 19394294; PubMed Central PMCID: PMC2996052.

14. Zhang DW, Mosley AL, Ramisetty SR, Rodriguez-Molina JB, Washburn MP, Ansari AZ. Ssu72 phosphatase-dependent erasure of phospho-Ser7 marks on the RNA polymerase II C-terminal domain is essential for viability and transcription termination. J Biol Chem. 2012;287(11):8541–51. Epub 2012/01/12. doi: 10.1074/jbc.M111.335687 22235117; PubMed Central PMCID: PMC3318730.

15. Jeronimo C, Collin P, Robert F. The RNA Polymerase II CTD: The Increasing Complexity of a Low-Complexity Protein Domain. J Mol Biol. 2016;428(12):2607–22. Epub 2016/02/16. doi: 10.1016/j.jmb.2016.02.006 26876604.

16. Bataille AR, Jeronimo C, Jacques PE, Laramee L, Fortin ME, Forest A, et al. A universal RNA polymerase II CTD cycle is orchestrated by complex interplays between kinase, phosphatase, and isomerase enzymes along genes. Mol Cell. 2012;45(2):158–70. Epub 2012/01/31. doi: 10.1016/j.molcel.2011.11.024 22284676.

17. Schreieck A, Easter AD, Etzold S, Wiederhold K, Lidschreiber M, Cramer P, et al. RNA polymerase II termination involves C-terminal-domain tyrosine dephosphorylation by CPF subunit Glc7. Nat Struct Mol Biol. 2014;21(2):175–9. Epub 2014/01/15. doi: 10.1038/nsmb.2753 24413056; PubMed Central PMCID: PMC3917824.

18. Nedea E, He X, Kim M, Pootoolal J, Zhong G, Canadien V, et al. Organization and function of APT, a subcomplex of the yeast cleavage and polyadenylation factor involved in the formation of mRNA and small nucleolar RNA 3'-ends. J Biol Chem. 2003;278(35):33000–10. Epub 2003/06/24. doi: 10.1074/jbc.M304454200 12819204.

19. Kong SE, Kobor MS, Krogan NJ, Somesh BP, Sogaard TM, Greenblatt JF, et al. Interaction of Fcp1 phosphatase with elongating RNA polymerase II holoenzyme, enzymatic mechanism of action, and genetic interaction with elongator. J Biol Chem. 2005;280(6):4299–306. Epub 2004/11/26. doi: 10.1074/jbc.M411071200 15563457.

20. Kobor MS, Archambault J, Lester W, Holstege FC, Gileadi O, Jansma DB, et al. An unusual eukaryotic protein phosphatase required for transcription by RNA polymerase II and CTD dephosphorylation in S. cerevisiae. Mol Cell. 1999;4(1):55–62. Epub 1999/08/13. doi: 10.1016/s1097-2765(00)80187-2 10445027.

21. Schwer B, Ghosh A, Sanchez AM, Lima CD, Shuman S. Genetic and structural analysis of the essential fission yeast RNA polymerase II CTD phosphatase Fcp1. Rna. 2015;21(6):1135–46. Epub 2015/04/18. doi: 10.1261/rna.050286.115 25883047; PubMed Central PMCID: PMC4436666.

22. Ghosh A, Shuman S, Lima CD. The structure of Fcp1, an essential RNA polymerase II CTD phosphatase. Mol Cell. 2008;32(4):478–90. Epub 2008/11/26. doi: 10.1016/j.molcel.2008.09.021 19026779; PubMed Central PMCID: PMC2645342.

23. Hsu PL, Yang F, Smith-Kinnaman W, Yang W, Song JE, Mosley AL, et al. Rtr1 is a dual specificity phosphatase that dephosphorylates Tyr1 and Ser5 on the RNA polymerase II CTD. J Mol Biol. 2014;426(16):2970–81. Epub 2014/06/22. doi: 10.1016/j.jmb.2014.06.010 24951832; PubMed Central PMCID: PMC4119023.

24. Krishnamurthy S, He X, Reyes-Reyes M, Moore C, Hampsey M. Ssu72 Is an RNA polymerase II CTD phosphatase. Mol Cell. 2004;14(3):387–94. Epub 2004/05/06. doi: 10.1016/s1097-2765(04)00235-7 15125841.

25. Meinhart A, Silberzahn T, Cramer P. The mRNA transcription/processing factor Ssu72 is a potential tyrosine phosphatase. J Biol Chem. 2003;278(18):15917–21. Epub 2003/02/28. doi: 10.1074/jbc.M301643200 12606538.

26. Hunter GO, Fox MJ, Smith-Kinnaman WR, Gogol M, Fleharty B, Mosley AL. The phosphatase Rtr1 regulates global levels of serine 5 RNA Polymerase II C-terminal domain phosphorylation and cotranscriptional histone methylation. Molecular and cellular biology. 2016. doi: 10.1128/MCB.00870-15 27247267.

27. Lunde BM, Reichow SL, Kim M, Suh H, Leeper TC, Yang F, et al. Cooperative interaction of transcription termination factors with the RNA polymerase II C-terminal domain. Nat Struct Mol Biol. 2010;17(10):1195–201. Epub 2010/09/08. doi: 10.1038/nsmb.1893 20818393; PubMed Central PMCID: PMC2950884.

28. Vasiljeva L, Kim M, Mutschler H, Buratowski S, Meinhart A. The Nrd1-Nab3-Sen1 termination complex interacts with the Ser5-phosphorylated RNA polymerase II C-terminal domain. Nat Struct Mol Biol. 2008;15(8):795–804. Epub 2008/07/29. doi: 10.1038/nsmb.1468 18660819; PubMed Central PMCID: PMC2597375.

29. Kim M, Vasiljeva L, Rando OJ, Zhelkovsky A, Moore C, Buratowski S. Distinct pathways for snoRNA and mRNA termination. Mol Cell. 2006;24(5):723–34. Epub 2006/12/13. doi: 10.1016/j.molcel.2006.11.011 17157255.

30. Vasiljeva L, Buratowski S. Nrd1 interacts with the nuclear exosome for 3' processing of RNA polymerase II transcripts. Mol Cell. 2006;21(2):239–48. Epub 2006/01/24. doi: 10.1016/j.molcel.2005.11.028 16427013.

31. Creamer TJ, Darby MM, Jamonnak N, Schaughency P, Hao H, Wheelan SJ, et al. Transcriptome-wide binding sites for components of the Saccharomyces cerevisiae non-poly(A) termination pathway: Nrd1, Nab3, and Sen1. PLoS Genet. 2011;7(10):e1002329. Epub 2011/10/27. doi: 10.1371/journal.pgen.1002329 22028667; PubMed Central PMCID: PMC3197677.

32. Jamonnak N, Creamer TJ, Darby MM, Schaughency P, Wheelan SJ, Corden JL. Yeast Nrd1, Nab3, and Sen1 transcriptome-wide binding maps suggest multiple roles in post-transcriptional RNA processing. Rna. 2011;17(11):2011–25. Epub 2011/09/29. doi: 10.1261/rna.2840711 21954178; PubMed Central PMCID: PMC3198594.

33. Hobor F, Pergoli R, Kubicek K, Hrossova D, Bacikova V, Zimmermann M, et al. Recognition of transcription termination signal by the nuclear polyadenylated RNA-binding (NAB) 3 protein. J Biol Chem. 2011;286(5):3645–57. Epub 2010/11/19. doi: 10.1074/jbc.M110.158774 21084293; PubMed Central PMCID: PMC3030368.

34. Carroll KL, Ghirlando R, Ames JM, Corden JL. Interaction of yeast RNA-binding proteins Nrd1 and Nab3 with RNA polymerase II terminator elements. Rna. 2007;13(3):361–73. Epub 2007/01/24. doi: 10.1261/rna.338407 17237360; PubMed Central PMCID: PMC1800511.

35. Loya TJ, O'Rourke TW, Degtyareva N, Reines D. A network of interdependent molecular interactions describes a higher order Nrd1-Nab3 complex involved in yeast transcription termination. J Biol Chem. 2013. Epub 2013/10/09. doi: 10.1074/jbc.M113.516765 24100036.

36. Loya TJ, O'Rourke TW, Reines D. The hnRNP-like Nab3 termination factor can employ heterologous prion-like domains in place of its own essential low complexity domain. PLoS One. 2017;12(10):e0186187. Epub 2017/10/13. doi: 10.1371/journal.pone.0186187 29023495; PubMed Central PMCID: PMC5638401.

37. Loya TJ, O'Rourke TW, Reines D. Yeast Nab3 protein contains a self-assembly domain found in human heterogeneous nuclear ribonucleoprotein-C (hnRNP-C) that is necessary for transcription termination. J Biol Chem. 2013;288(4):2111–7. Epub 2012/11/30. doi: 10.1074/jbc.M112.430678 23192344; PubMed Central PMCID: PMC3554884.

38. O'Rourke TW, Reines D. Determinants of Amyloid Formation for the Yeast Termination Factor Nab3. PLoS One. 2016;11(3):e0150865. Epub 2016/03/10. doi: 10.1371/journal.pone.0150865 26954508; PubMed Central PMCID: PMC4783047.

39. Wlotzka W, Kudla G, Granneman S, Tollervey D. The nuclear RNA polymerase II surveillance system targets polymerase III transcripts. Embo J. 2011;30(9):1790–803. Epub 2011/04/05. doi: 10.1038/emboj.2011.97 21460797; PubMed Central PMCID: PMC3102002.

40. Bacikova V, Pasulka J, Kubicek K, Stefl R. Structure and semi-sequence-specific RNA binding of Nrd1. Nucleic Acids Res. 2014;42(12):8024–38. Epub 2014/05/27. doi: 10.1093/nar/gku446 24860164; PubMed Central PMCID: PMC4081072.

41. Tudek A, Porrua O, Kabzinski T, Lidschreiber M, Kubicek K, Fortova A, et al. Molecular basis for coordinating transcription termination with noncoding RNA degradation. Mol Cell. 2014;55(3):467–81. Epub 2014/07/30. doi: 10.1016/j.molcel.2014.05.031 25066235; PubMed Central PMCID: PMC4186968.

42. Kim K, Heo DH, Kim I, Suh JY, Kim M. Exosome Cofactors Connect Transcription Termination to RNA Processing by Guiding Terminated Transcripts to the Appropriate Exonuclease within the Nuclear Exosome. J Biol Chem. 2016;291(25):13229–42. Epub 2016/04/15. doi: 10.1074/jbc.M116.715771 27076633; PubMed Central PMCID: PMC4933236.

43. Zhang Y, Chun Y, Buratowski S, Tong L. Identification of Three Sequence Motifs in the Transcription Termination Factor Sen1 that Mediate Direct Interactions with Nrd1. Structure. 2019;27(7):1156–61 e4. Epub 2019/05/21. doi: 10.1016/j.str.2019.04.005 31104813; PubMed Central PMCID: PMC6610696.

44. Arigo JT, Carroll KL, Ames JM, Corden JL. Regulation of yeast NRD1 expression by premature transcription termination. Mol Cell. 2006;21(5):641–51. Epub 2006/03/02. doi: 10.1016/j.molcel.2006.02.005 16507362.

45. Loya TJ, O'Rourke TW, Reines D. A genetic screen for terminator function in yeast identifies a role for a new functional domain in termination factor Nab3. Nucleic Acids Res. 2012;40(15):7476–91. Epub 2012/05/09. doi: 10.1093/nar/gks377 22564898; PubMed Central PMCID: PMC3424548.

46. Jenks MH, O'Rourke TW, Reines D. Properties of an intergenic terminator and start site switch that regulate IMD2 transcription in yeast. Molecular and cellular biology. 2008;28(12):3883–93. Epub 2008/04/23. doi: 10.1128/MCB.00380-08 18426909; PubMed Central PMCID: PMC2423123.

47. Kopcewicz KA, O'Rourke TW, Reines D. Metabolic regulation of IMD2 transcription and an unusual DNA element that generates short transcripts. Molecular and cellular biology. 2007;27(8):2821–9. Epub 2007/02/14. doi: 10.1128/MCB.02159-06 17296737; PubMed Central PMCID: PMC1899919.

48. Grzechnik P, Gdula MR, Proudfoot NJ. Pcf11 orchestrates transcription termination pathways in yeast. Genes Dev. 2015;29(8):849–61. Epub 2015/04/17. doi: 10.1101/gad.251470.114 25877920; PubMed Central PMCID: PMC4403260.

49. Mayer A, Heidemann M, Lidschreiber M, Schreieck A, Sun M, Hintermair C, et al. CTD tyrosine phosphorylation impairs termination factor recruitment to RNA polymerase II. Science. 2012;336(6089):1723–5. Epub 2012/06/30. doi: 10.1126/science.1219651 22745433.

50. Kim M, Krogan NJ, Vasiljeva L, Rando OJ, Nedea E, Greenblatt JF, et al. The yeast Rat1 exonuclease promotes transcription termination by RNA polymerase II. Nature. 2004;432(7016):517–22. Epub 2004/11/27. doi: 10.1038/nature03041 15565157.

51. Fong N, Brannan K, Erickson B, Kim H, Cortazar MA, Sheridan RM, et al. Effects of Transcription Elongation Rate and Xrn2 Exonuclease Activity on RNA Polymerase II Termination Suggest Widespread Kinetic Competition. Mol Cell. 2015;60(2):256–67. Epub 2015/10/17. doi: 10.1016/j.molcel.2015.09.026 26474067; PubMed Central PMCID: PMC4654110.

52. Pearson EL, Moore CL. Dismantling promoter-driven RNA polymerase II transcription complexes in vitro by the termination factor Rat1. J Biol Chem. 2013;288(27):19750–9. Epub 2013/05/22. doi: 10.1074/jbc.M112.434985 23689372; PubMed Central PMCID: PMC3707679.

53. Baejen C, Andreani J, Torkler P, Battaglia S, Schwalb B, Lidschreiber M, et al. Genome-wide Analysis of RNA Polymerase II Termination at Protein-Coding Genes. Mol Cell. 2017;66(1):38–49 e6. Epub 2017/03/21. doi: 10.1016/j.molcel.2017.02.009 28318822.

54. Park J, Kang M, Kim M. Unraveling the mechanistic features of RNA polymerase II termination by the 5'-3' exoribonuclease Rat1. Nucleic Acids Res. 2015;43(5):2625–37. Epub 2015/02/28. doi: 10.1093/nar/gkv133 25722373; PubMed Central PMCID: PMC4357727.

55. Proudfoot NJ. Transcriptional termination in mammals: Stopping the RNA polymerase II juggernaut. Science. 2016;352(6291):aad9926. Epub 2016/06/11. doi: 10.1126/science.aad9926 27284201; PubMed Central PMCID: PMC5144996.

56. Eaton JD, West S. An end in sight? Xrn2 and transcriptional termination by RNA polymerase II. Transcription. 2018;9(5):321–6. Epub 2018/07/24. doi: 10.1080/21541264.2018.1498708 30035655; PubMed Central PMCID: PMC6150625.

57. Fox MJ, Gao H, Smith-Kinnaman WR, Liu Y, Mosley AL. The exosome component Rrp6 is required for RNA polymerase II termination at specific targets of the Nrd1-Nab3 pathway. PLoS Genet. 2015;11(2):e1004999. Epub 2015/02/14. doi: 10.1371/journal.pgen.1004999 25680078; PubMed Central PMCID: PMC4378619.

58. Lemay JF, Bachand F. Fail-safe transcription termination: Because one is never enough. RNA Biol. 2015;12(9):927–32. Epub 2015/08/15. doi: 10.1080/15476286.2015.1073433 26273910; PubMed Central PMCID: PMC4615224.

59. Lemay JF, Larochelle M, Marguerat S, Atkinson S, Bahler J, Bachand F. The RNA exosome promotes transcription termination of backtracked RNA polymerase II. Nat Struct Mol Biol. 2014;21(10):919–26. Epub 2014/09/23. doi: 10.1038/nsmb.2893 25240800.

60. Smith-Kinnaman WR, Berna MJ, Hunter GO, True JD, Hsu P, Cabello GI, et al. The interactome of the atypical phosphatase Rtr1 in Saccharomyces cerevisiae. Mol Biosyst. 2014;10(7):1730–41. Epub 2014/03/29. doi: 10.1039/c4mb00109e 24671508; PubMed Central PMCID: PMC4074173.

61. Gibney PA, Fries T, Bailer SM, Morano KA. Rtr1 is the Saccharomyces cerevisiae homolog of a novel family of RNA polymerase II-binding proteins. Eukaryot Cell. 2008;7(6):938–48. Epub 2008/04/15. doi: 10.1128/EC.00042-08 18408053; PubMed Central PMCID: PMC2446653.

62. Gudipati RK, Villa T, Boulay J, Libri D. Phosphorylation of the RNA polymerase II C-terminal domain dictates transcription termination choice. Nat Struct Mol Biol. 2008;15(8):786–94. Epub 2008/07/29. doi: 10.1038/nsmb.1460 18660821.

63. Vasiljeva L, Kim M, Mutschler H, Buratowski S, Meinhart A. The Nrd1-Nab3-Sen1 termination complex interacts with the Ser5-phosphorylated RNA polymerase II C-terminal domain. Nature structural & molecular biology. 2008;15(8):795–804. Epub 2008/07/29. doi: 10.1038/nsmb.1468 18660819; PubMed Central PMCID: PMC2597375.

64. Arndt KM, Reines D. Termination of Transcription of Short Noncoding RNAs by RNA Polymerase II. Annual review of biochemistry. 2015;84:381–404. doi: 10.1146/annurev-biochem-060614-034457 25747400.

65. Porrua O, Libri D. Transcription termination and the control of the transcriptome: why, where and how to stop. Nature reviews Molecular cell biology. 2015;16(3):190–202. doi: 10.1038/nrm3943 25650800.

66. Sahni N, Yi S, Zhong Q, Jailkhani N, Charloteaux B, Cusick ME, et al. Edgotype: a fundamental link between genotype and phenotype. Curr Opin Genet Dev. 2013;23(6):649–57. Epub 2013/11/30. doi: 10.1016/j.gde.2013.11.002 24287335; PubMed Central PMCID: PMC3902775.

67. Charloteaux B, Zhong Q, Dreze M, Cusick ME, Hill DE, Vidal M. Protein-protein interactions and networks: forward and reverse edgetics. Methods Mol Biol. 2011;759:197–213. Epub 2011/08/25. doi: 10.1007/978-1-61779-173-4_12 21863489.

68. Choi H, Larsen B, Lin ZY, Breitkreutz A, Mellacheruvu D, Fermin D, et al. SAINT: probabilistic scoring of affinity purification-mass spectrometry data. Nat Methods. 2011;8(1):70–3. Epub 2010/12/07. doi: 10.1038/nmeth.1541 nmeth.1541 [pii]. 21131968; PubMed Central PMCID: PMC3064265.

69. Teo G, Liu G, Zhang J, Nesvizhskii AI, Gingras AC, Choi H. SAINTexpress: improvements and additional features in Significance Analysis of INTeractome software. J Proteomics. 2014;100:37–43. Epub 2014/02/12. doi: 10.1016/j.jprot.2013.10.023 S1874-3919(13)00538-1 [pii]. 24513533; PubMed Central PMCID: PMC4102138.

70. Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019;47(D1):D607–D13. Epub 2018/11/27. doi: 10.1093/nar/gky1131 30476243; PubMed Central PMCID: PMC6323986.

71. Casanal A, Kumar A, Hill CH, Easter AD, Emsley P, Degliesposti G, et al. Architecture of eukaryotic mRNA 3'-end processing machinery. Science. 2017;358(6366):1056–9. Epub 2017/10/28. doi: 10.1126/science.aao6535 29074584; PubMed Central PMCID: PMC5788269.

72. Bedard LG, Dronamraju R, Kerschner JL, Hunter GO, Axley ED, Boyd AK, et al. Quantitative Analysis of Dynamic Protein Interactions during Transcription Reveals a Role for Casein Kinase II in Polymerase-associated Factor (PAF) Complex Phosphorylation and Regulation of Histone H2B Monoubiquitylation. J Biol Chem. 2016;291(26):13410–20. Epub 2016/05/05. doi: 10.1074/jbc.M116.727735 27143358; PubMed Central PMCID: PMC4919428.

73. Mosley AL, Sardiu ME, Pattenden SG, Workman JL, Florens L, Washburn MP. Highly reproducible label free quantitative proteomic analysis of RNA polymerase complexes. Mol Cell Proteomics. 2011;10(2):M110 000687. Epub 2010/11/05. doi: 10.1074/mcp.M110.000687 M110.000687 [pii]. 21048197; PubMed Central PMCID: PMC3033667.

74. Jiang L, Schlesinger F, Davis CA, Zhang Y, Li R, Salit M, et al. Synthetic spike-in standards for RNA-seq experiments. Genome Res. 2011;21(9):1543–51. Epub 2011/08/06. doi: 10.1101/gr.121095.111 21816910; PubMed Central PMCID: PMC3166838.

75. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26(1):139–40. Epub 2009/11/17. doi: 10.1093/bioinformatics/btp616 19910308; PubMed Central PMCID: PMC2796818.

76. Neil H, Malabat C, d'Aubenton-Carafa Y, Xu Z, Steinmetz LM, Jacquier A. Widespread bidirectional promoters are the major source of cryptic transcripts in yeast. Nature. 2009;457(7232):1038–42. Epub 2009/01/27. doi: 10.1038/nature07747 19169244.

77. Schulz D, Schwalb B, Kiesel A, Baejen C, Torkler P, Gagneur J, et al. Transcriptome surveillance by selective termination of noncoding RNA synthesis. Cell. 2013;155(5):1075–87. Epub 2013/11/12. doi: 10.1016/j.cell.2013.10.024 24210918.

78. Merran J, Corden JL. Yeast RNA-Binding Protein Nab3 Regulates Genes Involved in Nitrogen Metabolism. Molecular and cellular biology. 2017;37(18). Epub 2017/07/05. doi: 10.1128/MCB.00154-17 28674185; PubMed Central PMCID: PMC5574042.

79. Kuehner JN, Brow DA. Regulation of a eukaryotic gene by GTP-dependent start site selection and transcription attenuation. Mol Cell. 2008;31(2):201–11. Epub 2008/07/29. doi: 10.1016/j.molcel.2008.05.018 18657503.

80. Steinmetz EJ, Warren CL, Kuehner JN, Panbehi B, Ansari AZ, Brow DA. Genome-wide distribution of yeast RNA polymerase II and its control by Sen1 helicase. Mol Cell. 2006;24(5):735–46. Epub 2006/12/13. doi: 10.1016/j.molcel.2006.10.023 17157256.

81. Hyle JW, Shaw RJ, Reines D. Functional distinctions between IMP dehydrogenase genes in providing mycophenolate resistance and guanine prototrophy to yeast. J Biol Chem. 2003;278(31):28470–8. Epub 2003/05/15. doi: 10.1074/jbc.M303736200 12746440; PubMed Central PMCID: PMC3367515.

82. Arigo JT, Eyler DE, Carroll KL, Corden JL. Termination of cryptic unstable transcripts is directed by yeast RNA-binding proteins Nrd1 and Nab3. Mol Cell. 2006;23(6):841–51. Epub 2006/09/16. doi: 10.1016/j.molcel.2006.07.024 16973436.

83. Rhee HS, Pugh BF. ChIP-exo method for identifying genomic location of DNA-binding proteins with near-single-nucleotide accuracy. Curr Protoc Mol Biol. 2012;Chapter 21:Unit 21 4. Epub 2012/10/03. doi: 10.1002/0471142727.mb2124s100 23026909; PubMed Central PMCID: PMC3813302.

84. Thiebaut M, Colin J, Neil H, Jacquier A, Seraphin B, Lacroute F, et al. Futile cycle of transcription initiation and termination modulates the response to nucleotide shortage in S. cerevisiae. Mol Cell. 2008;31(5):671–82. Epub 2008/09/09. doi: 10.1016/j.molcel.2008.08.010 18775327.

85. Hazelbaker DZ, Marquardt S, Wlotzka W, Buratowski S. Kinetic competition between RNA Polymerase II and Sen1-dependent transcription termination. Mol Cell. 2013;49(1):55–66. Epub 2012/11/28. doi: 10.1016/j.molcel.2012.10.014 23177741; PubMed Central PMCID: PMC3545030.

86. Vasiljeva L, Kim M, Terzi N, Soares LM, Buratowski S. Transcription termination and RNA degradation contribute to silencing of RNA polymerase II transcription within heterochromatin. Mol Cell. 2008;29(3):313–23. Epub 2008/02/19. doi: 10.1016/j.molcel.2008.01.011 18280237.

87. Fasken MB, Laribee RN, Corbett AH. Nab3 facilitates the function of the TRAMP complex in RNA processing via recruitment of Rrp6 independent of Nrd1. PLoS Genet. 2015;11(3):e1005044. Epub 2015/03/17. doi: 10.1371/journal.pgen.1005044 25775092; PubMed Central PMCID: PMC4361618.

88. Lemay JF, D'Amours A, Lemieux C, Lackner DH, St-Sauveur VG, Bahler J, et al. The nuclear poly(A)-binding protein interacts with the exosome to promote synthesis of noncoding small nucleolar RNAs. Mol Cell. 2010;37(1):34–45. Epub 2010/02/05. doi: 10.1016/j.molcel.2009.12.019 20129053.

89. LaCava J, Houseley J, Saveanu C, Petfalski E, Thompson E, Jacquier A, et al. RNA degradation by the exosome is promoted by a nuclear polyadenylation complex. Cell. 2005;121(5):713–24. Epub 2005/06/07. doi: 10.1016/j.cell.2005.04.029 15935758.

90. Shen L, Shao N, Liu X, Nestler E. ngs.plot: Quick mining and visualization of next-generation sequencing data by integrating genomic databases. BMC genomics. 2014;15:284. doi: 10.1186/1471-2164-15-284 24735413; PubMed Central PMCID: PMC4028082.

91. Nagalakshmi U, Wang Z, Waern K, Shou C, Raha D, Gerstein M, et al. The transcriptional landscape of the yeast genome defined by RNA sequencing. Science. 2008;320(5881):1344–9. Epub 2008/05/03. doi: 10.1126/science.1158441 18451266; PubMed Central PMCID: PMC2951732.

92. Webb S, Hector RD, Kudla G, Granneman S. PAR-CLIP data indicate that Nrd1-Nab3-dependent transcription termination regulates expression of hundreds of protein coding genes in yeast. Genome Biol. 2014;15(1):R8. Epub 2014/01/08. doi: 10.1186/gb-2014-15-1-r8 24393166; PubMed Central PMCID: PMC4053934.

93. Honorine R, Mosrin-Huaman C, Hervouet-Coste N, Libri D, Rahmouni AR. Nuclear mRNA quality control in yeast is mediated by Nrd1 co-transcriptional recruitment, as revealed by the targeting of Rho-induced aberrant transcripts. Nucleic Acids Res. 2011;39(7):2809–20. Epub 2010/11/30. doi: 10.1093/nar/gkq1192 21113025; PubMed Central PMCID: PMC3074134.

94. Marquardt S, Hazelbaker DZ, Buratowski S. Distinct RNA degradation pathways and 3' extensions of yeast non-coding RNA species. Transcription. 2011;2(3):145–54. Epub 2011/08/10. doi: 10.4161/trns.2.3.16298 21826286; PubMed Central PMCID: PMC3149692.

95. Gudipati RK, Xu Z, Lebreton A, Seraphin B, Steinmetz LM, Jacquier A, et al. Extensive degradation of RNA precursors by the exosome in wild-type cells. Mol Cell. 2012;48(3):409–21. Epub 2012/09/25. doi: 10.1016/j.molcel.2012.08.018 23000176; PubMed Central PMCID: PMC3496076.

96. Davis CA, Ares M Jr., Accumulation of unstable promoter-associated transcripts upon loss of the nuclear exosome subunit Rrp6p in Saccharomyces cerevisiae. Proc Natl Acad Sci U S A. 2006;103(9):3262–7. Epub 2006/02/18. doi: 10.1073/pnas.0507783103 16484372; PubMed Central PMCID: PMC1413877.

97. Castelnuovo M, Rahman S, Guffanti E, Infantino V, Stutz F, Zenklusen D. Bimodal expression of PHO84 is modulated by early termination of antisense transcription. Nat Struct Mol Biol. 2013;20(7):851–8. Epub 2013/06/19. doi: 10.1038/nsmb.2598 23770821.

98. Mostovoy Y, Thiemicke A, Hsu TY, Brem RB. The Role of Transcription Factors at Antisense-Expressing Gene Pairs in Yeast. Genome Biol Evol. 2016;8(6):1748–61. Epub 2016/05/18. doi: 10.1093/gbe/evw104 27190003; PubMed Central PMCID: PMC4943177.

99. Cakiroglu SA, Zaugg JB, Luscombe NM. Backmasking in the yeast genome: encoding overlapping information for protein-coding and RNA degradation. Nucleic Acids Res. 2016;44(17):8065–72. Epub 2016/08/06. doi: 10.1093/nar/gkw683 27492286; PubMed Central PMCID: PMC5041482.

100. Conrad NK, Wilson SM, Steinmetz EJ, Patturajan M, Brow DA, Swanson MS, et al. A yeast heterogeneous nuclear ribonucleoprotein complex associated with RNA polymerase II. Genetics. 2000;154(2):557–71. Epub 2000/02/03. 10655211; PubMed Central PMCID: PMC1460961.

101. Chen X, Poorey K, Carver MN, Muller U, Bekiranov S, Auble DT, et al. Transcriptomes of six mutants in the Sen1 pathway reveal combinatorial control of transcription termination across the Saccharomyces cerevisiae genome. PLoS Genet. 2017;13(6):e1006863. Epub 2017/07/01. doi: 10.1371/journal.pgen.1006863 28665995; PubMed Central PMCID: PMC5513554.

102. Ganem C, Devaux F, Torchet C, Jacq C, Quevillon-Cheruel S, Labesse G, et al. Ssu72 is a phosphatase essential for transcription termination of snoRNAs and specific mRNAs in yeast. Embo J. 2003;22(7):1588–98. Epub 2003/03/28. doi: 10.1093/emboj/cdg141 12660165; PubMed Central PMCID: PMC152886.

103. Winzeler EA, Shoemaker DD, Astromoff A, Liang H, Anderson K, Andre B, et al. Functional characterization of the S. cerevisiae genome by gene deletion and parallel analysis. Science. 1999;285(5429):901–6. Epub 1999/08/07. doi: 10.1126/science.285.5429.901 10436161.

104. Washburn MP, Wolters D, Yates JR 3rd. Large-scale analysis of the yeast proteome by multidimensional protein identification technology. Nat Biotechnol. 2001;19(3):242–7. Epub 2001/03/07. doi: 10.1038/85686 85686 [pii]. 11231557.

105. Mellacheruvu D, Wright Z, Couzens AL, Lambert JP, St-Denis NA, Li T, et al. The CRAPome: a contaminant repository for affinity purification-mass spectrometry data. Nat Methods. 2013;10(8):730–6. Epub 2013/08/08. doi: 10.1038/nmeth.2557 23921808; PubMed Central PMCID: PMC3773500.

106. Knight JDR, Choi H, Gupta GD, Pelletier L, Raught B, Nesvizhskii AI, et al. ProHits-viz: a suite of web tools for visualizing interaction proteomics data. Nat Methods. 2017;14(7):645–6. Epub 2017/07/01. doi: 10.1038/nmeth.4330 28661499; PubMed Central PMCID: PMC5831326.

107. Wickham H. ggplot2—Elegant Graphics for Data Analysis: Springer-Verlag, New York.; 2016. 260 p.

108. Burriss KH, Mosley AL. Methods review: Mass spectrometry analysis of RNAPII complexes. Methods. 2019;159–160:105–14. Epub 2019/03/25. doi: 10.1016/j.ymeth.2019.03.013 30902665.

109. Wal M, Pugh BF. Genome-wide mapping of nucleosome positions in yeast using high-resolution MNase ChIP-Seq. Methods Enzymol. 2012;513:233–50. Epub 2012/08/30. doi: 10.1016/B978-0-12-391938-0.00010-0 22929772; PubMed Central PMCID: PMC4871120.

110. Homer NM, Barry; Nelson, Stanley F. BFAST: an alignment tool for large scale genome resequencing. PLoS One. 2009;4(11):e7767. doi: 10.1371/journal.pone.0007767 19907642

111. Breese MR, Liu Y. NGSUtils: a software suite for analyzing and manipulating next-generation sequencing datasets. Bioinformatics. 2013;29(4):494–6. Epub 2013/01/15. doi: 10.1093/bioinformatics/bts731 23314324; PubMed Central PMCID: PMC3570212.

112. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29(1):15–21. Epub 2012/10/30. doi: 10.1093/bioinformatics/bts635 23104886; PubMed Central PMCID: PMC3530905.

113. Robinson JT, Thorvaldsdottir H, Winckler W, Guttman M, Lander ES, Getz G, et al. Integrative genomics viewer. Nat Biotechnol. 2011;29(1):24–6. Epub 2011/01/12. doi: 10.1038/nbt.1754 21221095; PubMed Central PMCID: PMC3346182.

114. Ching T, Huang S, Garmire LX. Power analysis and sample size estimation for RNA-Seq differential expression. Rna. 2014;20(11):1684–96. doi: 10.1261/rna.046011.114 25246651; PubMed Central PMCID: PMC4201821.


Článek vyšel v časopise

PLOS Genetics


2020 Číslo 3
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

Hypertenze a hypercholesterolémie – synergický efekt léčby
nový kurz
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Multidisciplinární zkušenosti u pacientů s diabetem
Autoři: Prof. MUDr. Martin Haluzík, DrSc., prof. MUDr. Vojtěch Melenovský, CSc., prof. MUDr. Vladimír Tesař, DrSc.

Úloha kombinovaných preparátů v léčbě arteriální hypertenze
Autoři: prof. MUDr. Martin Haluzík, DrSc.

Halitóza
Autoři: MUDr. Ladislav Korábek, CSc., MBA

Terapie roztroušené sklerózy v kostce
Autoři: MUDr. Dominika Šťastná, Ph.D.

Všechny kurzy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#