#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

DNA Damage Regulates Translation through β-TRCP Targeting of CReP


Approximately 600 human genes encode enzymes that act as ubiquitin ligases, which facilitate the transfer of the small protein ubiquitin to thousands of substrate proteins; “tagging” with ubiquitin often promotes the degradation of the substrate by the proteasome. In this paper, we adapt a technique called Ligase Trapping for use in mammalian cells. Ligase Trapping is a highly accurate method for determining which substrates are targeted by a ubiquitin ligase. Here we use it to identify new substrates of the human cell cycle regulator βTRCP. Our screen was indeed highly accurate, as we were able to validate 88% of the candidate substrates we identified by mass spectrometry. Some of these new substrates were unstable proteins that were stabilized by inhibition of βTRCP, or of the entire class of ubiquitin ligases of which βTRCP is a part. However, others appear to be stable or redundantly-targeted substrates, which have been more difficult to identify with current techniques. This suggests that Ligase Trapping will be able to reliably identify new substrates of human ubiquitin ligases. Further, one of the new βTRCP substrates, CReP, is specifically depleted upon DNA damage, and depletion of CReP contributes to inactivation of the translational machinery upon DNA damage.


Vyšlo v časopise: DNA Damage Regulates Translation through β-TRCP Targeting of CReP. PLoS Genet 11(6): e32767. doi:10.1371/journal.pgen.1005292
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1005292

Souhrn

Approximately 600 human genes encode enzymes that act as ubiquitin ligases, which facilitate the transfer of the small protein ubiquitin to thousands of substrate proteins; “tagging” with ubiquitin often promotes the degradation of the substrate by the proteasome. In this paper, we adapt a technique called Ligase Trapping for use in mammalian cells. Ligase Trapping is a highly accurate method for determining which substrates are targeted by a ubiquitin ligase. Here we use it to identify new substrates of the human cell cycle regulator βTRCP. Our screen was indeed highly accurate, as we were able to validate 88% of the candidate substrates we identified by mass spectrometry. Some of these new substrates were unstable proteins that were stabilized by inhibition of βTRCP, or of the entire class of ubiquitin ligases of which βTRCP is a part. However, others appear to be stable or redundantly-targeted substrates, which have been more difficult to identify with current techniques. This suggests that Ligase Trapping will be able to reliably identify new substrates of human ubiquitin ligases. Further, one of the new βTRCP substrates, CReP, is specifically depleted upon DNA damage, and depletion of CReP contributes to inactivation of the translational machinery upon DNA damage.


Zdroje

1. Benanti JA, Cheung SK, Brady MC, Toczyski DP (2007) A proteomic screen reveals SCFGrr1 targets that regulate the glycolytic-gluconeogenic switch. Nat Cell Biol 9: 1184–1191. 17828247

2. Yen HC, Elledge SJ (2008) Identification of SCF ubiquitin ligase substrates by global protein stability profiling. Science 322: 923–929. doi: 10.1126/science.1160462 18988848

3. Yen HC, Xu Q, Chou DM, Zhao Z, Elledge SJ (2008) Global protein stability profiling in mammalian cells. Science 322: 918–923. doi: 10.1126/science.1160489 18988847

4. Emanuele MJ, Elia AE, Xu Q, Thoma CR, Izhar L, et al. (2011) Global identification of modular cullin-RING ligase substrates. Cell 147: 459–474. doi: 10.1016/j.cell.2011.09.019 21963094

5. Kim W, Bennett EJ, Huttlin EL, Guo A, Li J, et al. (2011) Systematic and quantitative assessment of the ubiquitin-modified proteome. Mol Cell 44: 325–340. doi: 10.1016/j.molcel.2011.08.025 21906983

6. Dorrello NV, Peschiaroli A, Guardavaccaro D, Colburn NH, Sherman NE, et al. (2006) S6K1- and betaTRCP-mediated degradation of PDCD4 promotes protein translation and cell growth. Science 314: 467–471. 17053147

7. Peschiaroli A, Dorrello NV, Guardavaccaro D, Venere M, Halazonetis T, et al. (2006) SCFbetaTrCP-mediated degradation of Claspin regulates recovery from the DNA replication checkpoint response. Mol Cell 23: 319–329. 16885022

8. Sowa ME, Bennett EJ, Gygi SP, Harper JW (2009) Defining the human deubiquitinating enzyme interaction landscape. Cell 138: 389–403. doi: 10.1016/j.cell.2009.04.042 19615732

9. Vashisht AA, Zumbrennen KB, Huang X, Powers DN, Durazo A, et al. (2009) Control of iron homeostasis by an iron-regulated ubiquitin ligase. Science 326: 718–721. doi: 10.1126/science.1176333 19762596

10. Davis MA, Larimore EA, Fissel BM, Swanger J, Taatjes DJ, et al. (2013) The SCF-Fbw7 ubiquitin ligase degrades MED13 and MED13L and regulates CDK8 module association with Mediator. Genes Dev 27: 151–156. doi: 10.1101/gad.207720.112 23322298

11. Tan MK, Lim HJ, Bennett EJ, Shi Y, Harper JW (2013) Parallel SCF adaptor capture proteomics reveals a role for SCFFBXL17 in NRF2 activation via BACH1 repressor turnover. Mol Cell 52: 9–24. doi: 10.1016/j.molcel.2013.08.018 24035498

12. Landry BD, Doyle JP, Toczyski DP, Benanti JA (2012) F-box protein specificity for g1 cyclins is dictated by subcellular localization. PLoS Genet 8: e1002851. doi: 10.1371/journal.pgen.1002851 22844257

13. Mark KG, Simonetta M, Maiolica A, Seller CA, Toczyski DP (2014) Ubiquitin ligase trapping identifies an SCF(Saf1) pathway targeting unprocessed vacuolar/lysosomal proteins. Mol Cell 53: 148–161. doi: 10.1016/j.molcel.2013.12.003 24389104

14. Skaar JR, Pagan JK, Pagano M (2013) Mechanisms and function of substrate recruitment by F-box proteins. Nat Rev Mol Cell Biol 14: 369–381. doi: 10.1038/nrm3582 23657496

15. Willems AR, Schwab M, Tyers M (2004) A hitchhiker's guide to the cullin ubiquitin ligases: SCF and its kin. Biochim Biophys Acta 1695: 133–170. 15571813

16. Deshaies RJ (1999) SCF and Cullin/Ring H2-based ubiquitin ligases. Annu Rev Cell Dev Biol 15: 435–467. 10611969

17. Reed SI (2006) The ubiquitin-proteasome pathway in cell cycle control. Results Probl Cell Differ 42: 147–181. 16903211

18. Frescas D, Pagano M (2008) Deregulated proteolysis by the F-box proteins SKP2 and beta-TrCP: tipping the scales of cancer. Nat Rev Cancer 8: 438–449. doi: 10.1038/nrc2396 18500245

19. Watanabe N, Arai H, Nishihara Y, Taniguchi M, Watanabe N, et al. (2004) M-phase kinases induce phospho-dependent ubiquitination of somatic Wee1 by SCFbeta-TrCP. Proc Natl Acad Sci U S A 101: 4419–4424. 15070733

20. Busino L, Donzelli M, Chiesa M, Guardavaccaro D, Ganoth D, et al. (2003) Degradation of Cdc25A by beta-TrCP during S phase and in response to DNA damage. Nature 426: 87–91. 14603323

21. Jin J, Shirogane T, Xu L, Nalepa G, Qin J, et al. (2003) SCFbeta-TRCP links Chk1 signaling to degradation of the Cdc25A protein phosphatase. Genes Dev 17: 3062–3074. 14681206

22. Mamely I, van Vugt MA, Smits VA, Semple JI, Lemmens B, et al. (2006) Polo-like kinase-1 controls proteasome-dependent degradation of Claspin during checkpoint recovery. Curr Biol 16: 1950–1955. 16934469

23. Rada P, Rojo AI, Chowdhry S, McMahon M, Hayes JD, et al. (2011) SCF/{beta}-TrCP promotes glycogen synthase kinase 3-dependent degradation of the Nrf2 transcription factor in a Keap1-independent manner. Mol Cell Biol 31: 1121–1133. doi: 10.1128/MCB.01204-10 21245377

24. Lassot I, Segeral E, Berlioz-Torrent C, Durand H, Groussin L, et al. (2001) ATF4 degradation relies on a phosphorylation-dependent interaction with the SCF(betaTrCP) ubiquitin ligase. Mol Cell Biol 21: 2192–2202. 11238952

25. Kanemori Y, Uto K, Sagata N (2005) Beta-TrCP recognizes a previously undescribed nonphosphorylated destruction motif in Cdc25A and Cdc25B phosphatases. Proc Natl Acad Sci U S A 102: 6279–6284. 15845771

26. Uchida S, Watanabe N, Kudo Y, Yoshioka K, Matsunaga T, et al. (2011) SCFbeta(TrCP) mediates stress-activated MAPK-induced Cdc25B degradation. J Cell Sci 124: 2816–2825. doi: 10.1242/jcs.083931 21807946

27. Wang Z, Zhong J, Gao D, Inuzuka H, Liu P, et al. (2012) DEPTOR ubiquitination and destruction by SCF(beta-TrCP). Am J Physiol Endocrinol Metab 303: E163–169. doi: 10.1152/ajpendo.00105.2012 22454292

28. Magliozzi R, Low TY, Weijts BG, Cheng T, Spanjaard E, et al. (2013) Control of epithelial cell migration and invasion by the IKKbeta- and CK1alpha-mediated degradation of RAPGEF2. Dev Cell 27: 574–585. doi: 10.1016/j.devcel.2013.10.023 24290981

29. Margottin-Goguet F, Hsu JY, Loktev A, Hsieh HM, Reimann JD, et al. (2003) Prophase destruction of Emi1 by the SCF(betaTrCP/Slimb) ubiquitin ligase activates the anaphase promoting complex to allow progression beyond prometaphase. Dev Cell 4: 813–826. 12791267

30. Yang X, Wood PA, Ansell CM, Ohmori M, Oh EY, et al. (2009) Beta-catenin induces beta-TrCP-mediated PER2 degradation altering circadian clock gene expression in intestinal mucosa of ApcMin/+ mice. J Biochem 145: 289–297. doi: 10.1093/jb/mvn167 19106159

31. Huang W, Lv X, Liu C, Zha Z, Zhang H, et al. (2012) The N-terminal phosphodegron targets TAZ/WWTR1 protein for SCFbeta-TrCP-dependent degradation in response to phosphatidylinositol 3-kinase inhibition. J Biol Chem 287: 26245–26253. doi: 10.1074/jbc.M112.382036 22692215

32. D'Annibale S, Kim J, Magliozzi R, Low TY, Mohammed S, et al. (2014) Proteasome-dependent degradation of transcription factor activating enhancer-binding protein 4 (TFAP4) controls mitotic division. J Biol Chem 289: 7730–7737. doi: 10.1074/jbc.M114.549535 24500709

33. Kim J, D'Annibale S, Magliozzi R, Low TY, Jansen P, et al. (2014) USP17- and SCFbetaTrCP--regulated degradation of DEC1 controls the DNA damage response. Mol Cell Biol 34: 4177–4185. doi: 10.1128/MCB.00530-14 25202122

34. Jousse C, Oyadomari S, Novoa I, Lu P, Zhang Y, et al. (2003) Inhibition of a constitutive translation initiation factor 2alpha phosphatase, CReP, promotes survival of stressed cells. J Cell Biol 163: 767–775. 14638860

35. Pavitt GD, Ron D (2012) New insights into translational regulation in the endoplasmic reticulum unfolded protein response. Cold Spring Harb Perspect Biol 4.

36. Holcik M, Sonenberg N (2005) Translational control in stress and apoptosis. Nat Rev Mol Cell Biol 6: 318–327. 15803138

37. Wu G, Xu G, Schulman BA, Jeffrey PD, Harper JW, et al. (2003) Structure of a beta-TrCP1-Skp1-beta-catenin complex: destruction motif binding and lysine specificity of the SCF(beta-TrCP1) ubiquitin ligase. Mol Cell 11: 1445–1456. 12820959

38. Rual JF, Venkatesan K, Hao T, Hirozane-Kishikawa T, Dricot A, et al. (2005) Towards a proteome-scale map of the human protein-protein interaction network. Nature 437: 1173–1178. 16189514

39. Kim TY, Siesser PF, Rossman KL, Goldfarb D, Mackinnon K, et al. (2015) Substrate Trapping Proteomics Reveals Targets of the betaTrCP2/FBXW11 Ubiquitin Ligase. Mol Cell Biol 35: 167–181. doi: 10.1128/MCB.00857-14 25332235

40. Low TY, Peng M, Magliozzi R, Mohammed S, Guardavaccaro D, et al. (2014) A systems-wide screen identifies substrates of the SCFbetaTrCP ubiquitin ligase. Sci Signal 7: rs8. doi: 10.1126/scisignal.2005882 25515538

41. Latres E, Chiaur DS, Pagano M (1999) The human F box protein beta-Trcp associates with the Cul1/Skp1 complex and regulates the stability of beta-catenin. Oncogene 18: 849–854. 10023660

42. Qu Z, Qing G, Rabson A, Xiao G (2004) Tax deregulation of NF-kappaB2 p100 processing involves both beta-TrCP-dependent and-independent mechanisms. J Biol Chem 279: 44563–44572. 15310758

43. Kruiswijk F, Yuniati L, Magliozzi R, Low TY, Lim R, et al. (2012) Coupled activation and degradation of eEF2K regulates protein synthesis in response to genotoxic stress. Sci Signal 5: ra40. doi: 10.1126/scisignal.2002718 22669845

44. Guardavaccaro D, Frescas D, Dorrello NV, Peschiaroli A, Multani AS, et al. (2008) Control of chromosome stability by the beta-TrCP-REST-Mad2 axis. Nature 452: 365–369. doi: 10.1038/nature06641 18354482

45. Westbrook TF, Hu G, Ang XL, Mulligan P, Pavlova NN, et al. (2008) SCFbeta-TRCP controls oncogenic transformation and neural differentiation through REST degradation. Nature 452: 370–374. doi: 10.1038/nature06780 18354483

46. Paul M, Jabbar MA (1997) Phosphorylation of both phosphoacceptor sites in the HIV-1 Vpu cytoplasmic domain is essential for Vpu-mediated ER degradation of CD4. Virology 232: 207–216. 9185604

47. Coyaud E, Mis M, Laurent EM, Dunham WH, Couzens AL, et al. (2015) BioID-based identification of SCF beta-TrCP1/2 E3 ligase substrates. Mol Cell Proteomics.

48. Inuzuka H, Tseng A, Gao D, Zhai B, Zhang Q, et al. (2010) Phosphorylation by casein kinase I promotes the turnover of the Mdm2 oncoprotein via the SCF(beta-TRCP) ubiquitin ligase. Cancer Cell 18: 147–159. doi: 10.1016/j.ccr.2010.06.015 20708156

49. Harding HP, Novoa I, Zhang Y, Zeng H, Wek R, et al. (2000) Regulated translation initiation controls stress-induced gene expression in mammalian cells. Mol Cell 6: 1099–1108. 11106749

50. Marciniak SJ, Yun CY, Oyadomari S, Novoa I, Zhang Y, et al. (2004) CHOP induces death by promoting protein synthesis and oxidation in the stressed endoplasmic reticulum. Genes Dev 18: 3066–3077. 15601821

51. Harding HP, Zhang Y, Scheuner D, Chen JJ, Kaufman RJ, et al. (2009) Ppp1r15 gene knockout reveals an essential role for translation initiation factor 2 alpha (eIF2alpha) dephosphorylation in mammalian development. Proc Natl Acad Sci U S A 106: 1832–1837. doi: 10.1073/pnas.0809632106 19181853

52. Novoa I, Zeng H, Harding HP, Ron D (2001) Feedback inhibition of the unfolded protein response by GADD34-mediated dephosphorylation of eIF2alpha. J Cell Biol 153: 1011–1022. 11381086

53. Fornace AJ Jr, Jackman J, Hollander MC, Hoffman-Liebermann B, Liebermann DA (1992) Genotoxic-stress-response genes and growth-arrest genes. gadd, MyD, and other genes induced by treatments eliciting growth arrest. Ann N Y Acad Sci 663: 139–153. 1482047

54. Schmidt EK, Clavarino G, Ceppi M, Pierre P (2009) SUnSET, a nonradioactive method to monitor protein synthesis. Nat Methods 6: 275–277. doi: 10.1038/nmeth.1314 19305406

55. Kudo T, Ikeda M, Nishikawa M, Yang Z, Ohno K, et al. (2012) The RASSF3 candidate tumor suppressor induces apoptosis and G1-S cell-cycle arrest via p53. Cancer Res 72: 2901–2911. doi: 10.1158/0008-5472.CAN-12-0572 22593196

56. Muratani M, Kung C, Shokat KM, Tansey WP (2005) The F box protein Dsg1/Mdm30 is a transcriptional coactivator that stimulates Gal4 turnover and cotranscriptional mRNA processing. Cell 120: 887–899. 15797387

57. Deng J, Harding HP, Raught B, Gingras AC, Berlanga JJ, et al. (2002) Activation of GCN2 in UV-irradiated cells inhibits translation. Curr Biol 12: 1279–1286. 12176355

58. Powley IR, Kondrashov A, Young LA, Dobbyn HC, Hill K, et al. (2009) Translational reprogramming following UVB irradiation is mediated by DNA-PKcs and allows selective recruitment to the polysomes of mRNAs encoding DNA repair enzymes. Genes Dev 23: 1207–1220. doi: 10.1101/gad.516509 19451221

59. Zhang P, Gao K, Tang Y, Jin X, An J, et al. (2014) Destruction of DDIT3/CHOP Protein by Wild-Type SPOP but Not Prostate Cancer-Associated Mutants. Hum Mutat 35: 1142–1151. doi: 10.1002/humu.22614 24990631

60. Brush MH, Shenolikar S (2008) Control of cellular GADD34 levels by the 26S proteasome. Mol Cell Biol 28: 6989–7000. doi: 10.1128/MCB.00724-08 18794359

61. Popov N, Schulein C, Jaenicke LA, Eilers M (2010) Ubiquitylation of the amino terminus of Myc by SCF(beta-TrCP) antagonizes SCF(Fbw7)-mediated turnover. Nat Cell Biol 12: 973–981. doi: 10.1038/ncb2104 20852628

62. Kaiser P, Wohlschlegel J (2005) Identification of ubiquitination sites and determination of ubiquitin-chain architectures by mass spectrometry. Methods in Enzymology 399: 266–277. 16338362

63. Wohlschlegel JA (2009) Identification of SUMO-conjugated proteins and their SUMO attachment sites using proteomic mass spectrometry. Methods in molecular biology 497: 33–49. doi: 10.1007/978-1-59745-566-4_3 19107409

64. Kelstrup CD, Young C, Lavallee R, Nielsen ML, Olsen JV (2012) Optimized fast and sensitive acquisition methods for shotgun proteomics on a quadrupole orbitrap mass spectrometer. Journal of proteome research 11: 3487–3497. doi: 10.1021/pr3000249 22537090

65. Michalski A, Damoc E, Hauschild JP, Lange O, Wieghaus A, et al. (2011) Mass spectrometry-based proteomics using Q Exactive, a high-performance benchtop quadrupole Orbitrap mass spectrometer. Molecular & cellular proteomics: MCP 10: M111 011015.

66. Beausoleil SA, Villen J, Gerber SA, Rush J, Gygi SP (2006) A probability-based approach for high-throughput protein phosphorylation analysis and site localization. Nature biotechnology 24: 1285–1292. 16964243

67. Cociorva D, D LT, Yates JR (2007) Validation of tandem mass spectrometry database search results using DTASelect. Curr Protoc Bioinformatics Chapter 13: Unit 13 14.

68. Tabb DL, McDonald WH, Yates JR 3rd (2002) DTASelect and Contrast: tools for assembling and comparing protein identifications from shotgun proteomics. J Proteome Res 1: 21–26. 12643522

69. Xu T, Venable JD, Park SK, Cociorva D, Lu B, et al. (2006) ProLuCID, A Fast and Sensitive Tandem Mass Spectra-based Protein Identification Programs. Molecular & Cellular Proteomics 5: S174.

70. Elias JE, Gygi SP (2007) Target-decoy search strategy for increased confidence in large-scale protein identifications by mass spectrometry. Nat Methods 4: 207–214. 17327847

71. Florens L, Carozza MJ, Swanson SK, Fournier M, Coleman MK, et al. (2006) Analyzing chromatin remodeling complexes using shotgun proteomics and normalized spectral abundance factors. Methods 40: 303–311. 17101441

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2015 Číslo 6
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#