#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

P-TEFb, the Super Elongation Complex and Mediator Regulate a Subset of Non-paused Genes during Early Embryo Development


Embryo development involves formation of various cell types through the regulation of gene transcription, resulting in expression of cell type specific RNAs and proteins. A key regulatory step in transcription of animal genes involves the transition of RNA polymerase II (Pol II) into active elongation. At many genes, Pol II is transiently paused approximately 50 basepairs downstream of the transcription start site. Release from this promoter-proximal pausing involves the kinase P-TEFb, which phosphorylates negative elongation factors, allowing Pol II to enter into productive elongation. In this work, we have depleted a considerable amount of P-TEFb from early Drosophila embryos. We find that several genes with paused Pol II can be expressed relatively normally in P-TEFb depleted embryos, whereas expression of some non-paused genes is substantially reduced. This result suggests that also non-paused genes transit through a P-TEFb-dependent checkpoint before entering active elongation. Unexpectedly, we find less Pol II associated with these non-paused genes in P-TEFb embryos. We demonstrate that a protein complex involved in recruitment of Pol II to promoters, the Mediator complex, show the same morphological and gene expression phenotypes as P-TEFb. We propose that Mediator and P-TEFb function together in recruiting Pol II to a subset of developmental genes.


Vyšlo v časopise: P-TEFb, the Super Elongation Complex and Mediator Regulate a Subset of Non-paused Genes during Early Embryo Development. PLoS Genet 11(2): e32767. doi:10.1371/journal.pgen.1004971
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004971

Souhrn

Embryo development involves formation of various cell types through the regulation of gene transcription, resulting in expression of cell type specific RNAs and proteins. A key regulatory step in transcription of animal genes involves the transition of RNA polymerase II (Pol II) into active elongation. At many genes, Pol II is transiently paused approximately 50 basepairs downstream of the transcription start site. Release from this promoter-proximal pausing involves the kinase P-TEFb, which phosphorylates negative elongation factors, allowing Pol II to enter into productive elongation. In this work, we have depleted a considerable amount of P-TEFb from early Drosophila embryos. We find that several genes with paused Pol II can be expressed relatively normally in P-TEFb depleted embryos, whereas expression of some non-paused genes is substantially reduced. This result suggests that also non-paused genes transit through a P-TEFb-dependent checkpoint before entering active elongation. Unexpectedly, we find less Pol II associated with these non-paused genes in P-TEFb embryos. We demonstrate that a protein complex involved in recruitment of Pol II to promoters, the Mediator complex, show the same morphological and gene expression phenotypes as P-TEFb. We propose that Mediator and P-TEFb function together in recruiting Pol II to a subset of developmental genes.


Zdroje

1. Corden JL (1990) Tails of RNA polymerase II. Trends Biochem Sci 15: 383–387. 2251729

2. Hsin JP, Manley JL (2012) The RNA polymerase II CTD coordinates transcription and RNA processing. Genes Dev 26: 2119–2137. doi: 10.1101/gad.200303.112 23028141

3. Guenther MG, Levine SS, Boyer LA, Jaenisch R, Young RA (2007) A chromatin landmark and transcription initiation at most promoters in human cells. Cell 130: 77–88. 17632057

4. Muse GW, Gilchrist DA, Nechaev S, Shah R, Parker JS, et al. (2007) RNA polymerase is poised for activation across the genome. Nature Genetics 39: 1507–1511. 17994021

5. Zeitlinger J, Stark A, Kellis M, Hong JW, Nechaev S, et al. (2007) RNA polymerase stalling at developmental control genes in the Drosophila melanogaster embryo. Nature Genetics 39: 1512–1516. 17994019

6. Adelman K, Lis JT (2012) Promoter-proximal pausing of RNA polymerase II: emerging roles in metazoans. Nature Reviews Genetics 13: 720–731. doi: 10.1038/nrg3293 22986266

7. Saunders A, Core LJ, Sutcliffe C, Lis JT, Ashe HL (2013) Extensive polymerase pausing during Drosophila axis patterning enables high-level and pliable transcription. Genes Dev 27: 1146–1158. doi: 10.1101/gad.215459.113 23699410

8. Gilmour DS (2009) Promoter proximal pausing on genes in metazoans. Chromosoma 118: 1–10. doi: 10.1007/s00412-008-0182-4 18830703

9. Peterlin BM, Price DH (2006) Controlling the elongation phase of transcription with P-TEFb. Mol Cell 23: 297–305. 16885020

10. Zhou Q, Li TD, Price DH (2012) RNA Polymerase II Elongation Control. Ann Rev Biochem 81: 119–143. doi: 10.1146/annurev-biochem-052610-095910 22404626

11. Henriques T, Gilchrist DA, Nechaev S, Bern M, Muse GW, et al. (2013) Stable pausing by RNA polymerase II provides an opportunity to target and integrate regulatory signals. Mol Cell 52: 517–528. doi: 10.1016/j.molcel.2013.10.001 24184211

12. Jonkers I, Kwak H, Lis JT (2014) Genome-wide dynamics of Pol II elongation and its interplay with promoter proximal pausing, chromatin, and exons. eLife 3: e02407. doi: 10.7554/eLife.02407 24843027

13. Nguyen D, Krueger BJ, Sedore SC, Brogie JE, Rogers JT, et al. (2012) The Drosophila 7SK snRNP and the essential role of dHEXIM in development. Nucleic Acids Research 40: 5283–5297. doi: 10.1093/nar/gks191 22379134

14. Luo Z, Lin C, Shilatifard A (2012) The super elongation complex (SEC) family in transcriptional control. Nat Rev Mol Cell Biol 13: 543–547. doi: 10.1038/nrm3417 22895430

15. Smith ER, Lin C, Garrett AS, Thornton J, Mohaghegh N, et al. (2011) The little elongation complex regulates small nuclear RNA transcription. Mol Cell 44: 954–965. doi: 10.1016/j.molcel.2011.12.008 22195968

16. Lin C, Garrett AS, De Kumar B, Smith ER, Gogol M, et al. (2011) Dynamic transcriptional events in embryonic stem cells mediated by the super elongation complex (SEC). Genes Dev 25: 1486–1498. doi: 10.1101/gad.2059211 21764852

17. Takahashi H, Parmely TJ, Sato S, Tomomori-Sato C, Banks CAS, et al. (2011) Human Mediator Subunit MED26 Functions as a Docking Site for Transcription Elongation Factors. Cell 146: 92–104. doi: 10.1016/j.cell.2011.06.005 21729782

18. Sullivan W, Theurkauf WE (1995) The cytoskeleton and morphogenesis of the early Drosophila embryo. Curr Opin Cell Biol 7: 18–22. 7755985

19. Tadros W, Lipshitz HD (2009) The maternal-to-zygotic transition: a play in two acts. Development 136: 3033–3042. doi: 10.1242/dev.033183 19700615

20. Mazumdar A, Mazumdar M (2002) How one becomes many: blastoderm cellularization in Drosophila melanogaster. BioEssays 24: 1012–1022. 12386932

21. Sokac AM, Wieschaus E (2008) Local actin-dependent endocytosis is zygotically controlled to initiate Drosophila cellularization. Dev Cell 14: 775–786. doi: 10.1016/j.devcel.2008.02.014 18477459

22. Wenzl C, Yan S, Laupsien P, Grosshans J (2010) Localization of RhoGEF2 during Drosophila cellularization is developmentally controlled by Slam. Mech Dev 127: 371–384. doi: 10.1016/j.mod.2010.01.001 20060902

23. Liang HL, Nien CY, Liu HY, Metzstein MM, Kirov N, et al. (2008) The zinc-finger protein Zelda is a key activator of the early zygotic genome in Drosophila. Nature 456: 400–403. doi: 10.1038/nature07388 18931655

24. Tang AH, Neufeld TP, Rubin GM, Muller HA (2001) Transcriptional regulation of cytoskeletal functions and segmentation by a novel maternal pair-rule gene, lilliputian. Development 128: 801–813. 11171404

25. Chen K, Johnston J, Shao W, Meier S, Staber C, et al. (2013) A global change in RNA polymerase II pausing during the Drosophila midblastula transition. Elife 2: e00861. doi: 10.7554/eLife.00861 23951546

26. Rothe M, Pehl M, Taubert H, Jackle H (1992) Loss of gene function through rapid mitotic cycles in the Drosophila embryo. Nature 359: 156–159. 1522901

27. Shermoen AW, O'Farrell PH (1991) Progression of the cell cycle through mitosis leads to abortion of nascent transcripts. Cell 67: 303–310. 1680567

28. Eissenberg JC, Shilatifard A, Dorokhov N, Michener DE (2007) Cdk9 is an essential kinase in Drosophila that is required for heat shock gene expression, histone methylation and elongation factor recruitment. Molecular Genetics and Genomics 277: 101–114. 17001490

29. Chopra VS, Hong JW, Levine M (2009) Regulation of Hox gene activity by transcriptional elongation in Drosophila. Curr Biol 19: 688–693. doi: 10.1016/j.cub.2009.02.055 19345103

30. Ni JQ, Zhou R, Czech B, Liu LP, Holderbaum L, et al. (2011) A genome-scale shRNA resource for transgenic RNAi in Drosophila. Nat Methods 8: 405–407. doi: 10.1038/nmeth.1592 21460824

31. Staller MV, Yan D, Randklev S, Bragdon MD, Wunderlich ZB, et al. (2013) Depleting gene activities in early Drosophila embryos with the "maternal-Gal4-shRNA" system. Genetics 193: 51–61. doi: 10.1534/genetics.112.144915 23105012

32. Gerber M, Ma J, Dean K, Eissenberg JC, Shilatifard A (2001) Drosophila ELL is associated with actively elongating RNA polymerase II on transcriptionally active sites in vivo. EMBO J 20: 6104–6114. 11689450

33. St Pierre SE, Ponting L, Stefancsik R, McQuilton P (2014) FlyBase 102—advanced approaches to interrogating FlyBase. Nucleic Acids Research 42: D780–788. doi: 10.1093/nar/gkt1092 24234449

34. Baldarelli RM, Mahoney PA, Salas F, Gustavson E, Boyer PD, et al. (1988) Transcripts of the Drosophila blastoderm-specific locus, terminus, are concentrated posteriorly and encode a potential DNA-binding finger. Developmental Biology 125: 85–95. 3334721

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

36. Shim EY, Walker AK, Shi Y, Blackwell TK (2002) CDK-9/cyclin T (P-TEFb) is required in two postinitiation pathways for transcription in the C-elegans embryo. Genes Dev 16: 2135–2146. 12183367

37. Eissenberg JC, Ma J, Gerber MA, Christensen A, Kennison JA, et al. (2002) dELL is an essential RNA polymerase II elongation factor with a general role in development. Proceedings of the National Academy of Sciences of the United States of America 99: 9894–9899. 12096188

38. Carlsten JO, Zhu X, Gustafsson CM (2013) The multitalented Mediator complex. Trends Biochem Sci 38: 531–537. doi: 10.1016/j.tibs.2013.08.007 24074826

39. Wang W, Yao X, Huang Y, Hu X, Liu R, et al. (2013) Mediator MED23 regulates basal transcription in vivo via an interaction with P-TEFb. Transcription 4: 39–51. doi: 10.4161/trns.22874 23340209

40. Galbraith MD, Allen MA, Bensard CL, Wang X, Schwinn MK, et al. (2013) HIF1A employs CDK8-mediator to stimulate RNAPII elongation in response to hypoxia. Cell 153: 1327–1339. doi: 10.1016/j.cell.2013.04.048 23746844

41. Nakamura A, Shirae-Kurabayashi M, Hanyu-Nakamura K (2010) Repression of early zygotic transcription in the germline. Curr Opin Cell Biol 22: 709–714. doi: 10.1016/j.ceb.2010.08.012 20817425

42. Hanyu-Nakamura K, Sonobe-Nojima H, Tanigawa A, Lasko P, Nakamura A (2008) Drosophila Pgc protein inhibits P-TEFb recruitment to chromatin in primordial germ cells. Nature 451: 730–733. doi: 10.1038/nature06498 18200011

43. Martinho RG, Kunwar PS, Casanova J, Lehmann R (2004) A noncoding RNA is required for the repression of RNApolII-dependent transcription in primordial germ cells. Curr Biol 14: 159–165. 14738740

44. Batchelder C, Dunn MA, Choy B, Suh Y, Cassie C, et al. (1999) Transcriptional repression by the Caenorhabditis elegans germ-line protein PIE-1. Genes Dev 13: 202–212. 9925644

45. Ghosh D, Seydoux G (2008) Inhibition of transcription by the Caenorhabditis elegans germline protein PIE-1: genetic evidence for distinct mechanisms targeting initiation and elongation. Genetics 178: 235–243. doi: 10.1534/genetics.107.083212 18202370

46. Seydoux G, Dunn MA (1997) Transcriptionally repressed germ cells lack a subpopulation of phosphorylated RNA polymerase II in early embryos of Caenorhabditis elegans and Drosophila melanogaster. Development 124: 2191–2201. 9187145

47. Bowman EA, Bowman CR, Ahn JH, Kelly WG (2013) Phosphorylation of RNA polymerase II is independent of P-TEFb in the C. elegans germline. Development 140: 3703–3713. doi: 10.1242/dev.095778 23903194

48. Sullivan W, Daily DR, Fogarty P, Yook KJ, Pimpinelli S (1993) Delays in anaphase initiation occur in individual nuclei of the syncytial Drosophila embryo. Mol Biol Cell 4: 885–896. 8257792

49. Bischof J, Maeda RK, Hediger M, Karch F, Basler K (2007) An optimized transgenesis system for Drosophila using germ-line-specific phiC31 integrases. Proceedings of the National Academy of Sciences of the United States of America 104: 3312–3317. 17360644

50. Ejsmont RK, Sarov M, Winkler S, Lipinski KA, Tomancak P (2009) A toolkit for high-throughput, cross-species gene engineering in Drosophila. Nature Methods 6: 435–U452. doi: 10.1038/nmeth.1334 19465918

51. Thomason LC, Costantino N, Shaw DV, Court DL (2007) Multicopy plasmid modification with phage lambda red recombineering. Plasmid 58: 148–158. 17434584

52. Chou TB, Perrimon N (1996) The autosomal FLP-DFS technique for generating germline mosaics in Drosophila melanogaster. Genetics 144: 1673–1679. 8978054

53. Jiang J, Hoey T, Levine M (1991) Autoregulation of a Segmentation Gene in Drosophila—Combinatorial Interaction of the Even-Skipped Homeo Box Protein with a Distal Enhancer Element. Genes Dev 5: 265–277. 1671662

54. Tautz D, Pfeifle C (1989) A Non-Radioactive Insitu Hybridization Method for the Localization of Specific Rnas in Drosophila Embryos Reveals Translational Control of the Segmentation Gene Hunchback. Chromosoma 98: 81–85. 2476281

55. Vandesompele J, De Preter K, Pattyn F, Poppe B, Van Roy N, et al. (2002) Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biology 3: RESEARCH0034. 12184808

56. Qi D, Bergman M, Aihara H, Nibu Y, Mannervik M (2008) Drosophila Ebi mediates Snail-dependent transcriptional repression through HDAC3-induced histone deacetylation. EMBO J 27: 898–909. doi: 10.1038/emboj.2008.26 18309295

57. Holmqvist PH, Boija A, Philip P, Crona F, Stenberg P, et al. (2012) Preferential genome targeting of the CBP co-activator by Rel and Smad proteins in early Drosophila melanogaster embryos. PLoS Genetics 8: e1002769. doi: 10.1371/journal.pgen.1002769 22737084

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2015 Číslo 2
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#