#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Integrative proteomic and phosphoproteomic profiling of prostate cell lines


Autoři: Maria Katsogiannou aff001;  Jean-Baptiste Boyer aff001;  Alberto Valdeolivas aff003;  Elisabeth Remy aff003;  Laurence Calzone aff006;  Stéphane Audebert aff001;  Palma Rocchi aff001;  Luc Camoin aff001;  Anaïs Baudot aff003
Působiště autorů: Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France aff001;  Obstetrics and Gynecology department, Hôpital Saint Joseph, Marseille, France aff002;  Aix Marseille Univ, CNRS, Centrale Marseille, I2M, Marseille, France aff003;  Aix Marseille Univ, INSERM, MMG, Marseille, France aff004;  ProGeLife, Marseille, France aff005;  Mines Paris Tech, Institut Curie, PSL Research University, Paris, France aff006
Vyšlo v časopise: PLoS ONE 14(11)
Kategorie: Research Article
doi: https://doi.org/10.1371/journal.pone.0224148

Souhrn

Background

Prostate cancer is a major public health issue, mainly because patients relapse after androgen deprivation therapy. Proteomic strategies, aiming to reflect the functional activity of cells, are nowadays among the leading approaches to tackle the challenges not only of better diagnosis, but also of unraveling mechanistic details related to disease etiology and progression.

Methods

We conducted here a large SILAC-based Mass Spectrometry experiment to map the proteomes and phosphoproteomes of four widely used prostate cell lines, namely PNT1A, LNCaP, DU145 and PC3, representative of different cancerous and hormonal status.

Results

We identified more than 3000 proteins and phosphosites, from which we quantified more than 1000 proteins and 500 phosphosites after stringent filtering. Extensive exploration of this proteomics and phosphoproteomics dataset allowed characterizing housekeeping as well as cell-line specific proteins, phosphosites and functional features of each cell line. In addition, by comparing the sensitive and resistant cell lines, we identified protein and phosphosites differentially expressed in the resistance context. Further data integration in a molecular network highlighted the differentially expressed pathways, in particular migration and invasion, RNA splicing, DNA damage repair response and transcription regulation.

Conclusions

Overall, this study proposes a valuable resource toward the characterization of proteome and phosphoproteome of four widely used prostate cell lines and reveals candidates to be involved in prostate cancer progression for further experimental validation.

Klíčová slova:

Biomarkers – Membrane proteins – Phosphorylation – Prostate cancer – Protein expression – Proteomes – Proteomic databases – DU145 cells


Zdroje

1. Fusi A, Procopio G, Della Torre S, Ricotta R, Bianchini G, Salvioni R, et al. Treatment options in hormone-refractory metastatic prostate carcinoma. Tumori. 2004;90(6):535–46. doi: 10.1177/030089160409000601 15762353

2. Karantanos T, Evans CP, Tombal B, Thompson TC, Montironi R, Isaacs WB. Understanding the mechanisms of androgen deprivation resistance in prostate cancer at the molecular level. Eur Urol. 2015;67(3):470–9. doi: 10.1016/j.eururo.2014.09.049 25306226

3. Katsogiannou M, Ziouziou H, Karaki S, Andrieu C, Henry de Villeneuve M, Rocchi P. The hallmarks of castration-resistant prostate cancers. Cancer Treat Rev. 2015;41(7):588–97. doi: 10.1016/j.ctrv.2015.05.003 25981454

4. Galsky MD, Small AC, Tsao CK, Oh WK. Clinical development of novel therapeutics for castration-resistant prostate cancer: historic challenges and recent successes. CA Cancer J Clin. 2012;62(5):299–308. doi: 10.3322/caac.21141 22535487

5. Suzman DL, Antonarakis ES. Castration-resistant prostate cancer: latest evidence and therapeutic implications. Ther Adv Med Oncol. 2014;6(4):167–79. doi: 10.1177/1758834014529176 25057303

6. Iglesias-Gato D, Thysell E, Tyanova S, Crnalic S, Santos A, Lima TS, et al. The Proteome of Prostate Cancer Bone Metastasis Reveals Heterogeneity with Prognostic Implications. Clin Cancer Res. 2018;24(21):5433–5444. doi: 10.1158/1078-0432.CCR-18-1229 30042207

7. Iglesias-Gato D, Wikstrom P, Tyanova S, Lavallee C, Thysell E, Carlsson J, et al. The Proteome of Primary Prostate Cancer. Eur Urol. 2016;69(5):942–52. doi: 10.1016/j.eururo.2015.10.053 26651926

8. Kuruma H, Egawa S, Oh-Ishi M, Kodera Y, Maeda T. Proteome analysis of prostate cancer. Prostate Cancer Prostatic Dis. 2005;8(1):14–21. doi: 10.1038/sj.pcan.4500764 15477873

9. Kuruma H, Egawa S, Oh-Ishi M, Kodera Y, Satoh M, Chen W, et al. High molecular mass proteome of androgen-independent prostate cancer. Proteomics. 2005;5(4):1097–112. doi: 10.1002/pmic.200401115 15712236

10. Ornstein DK, Tyson DR. Proteomics for the identification of new prostate cancer biomarkers. Urol Oncol. 2006;24(3):231–6. doi: 10.1016/j.urolonc.2005.11.035 16678055

11. Johnson LN. The regulation of protein phosphorylation. Biochem Soc Trans. 2009;37(Pt 4):627–41. doi: 10.1042/BST0370627 19614568

12. Cans C, Mangano R, Barila D, Neubauer G, Superti-Furga G. Nuclear tyrosine phosphorylation: the beginning of a map. Biochem Pharmacol. 2000;60(8):1203–15. doi: 10.1016/s0006-2952(00)00434-2 11007959

13. Lopez Villar E, Madero L, J ALP, W CC. Study of phosphorylation events for cancer diagnoses and treatment. Clin Transl Med. 2015;4(1):59. doi: 10.1186/s40169-015-0059-0 26055493

14. Harsha HC, Pandey A. Phosphoproteomics in cancer. Mol Oncol. 2010;4(6):482–95. doi: 10.1016/j.molonc.2010.09.004 20937571

15. Lescarbeau RM, Kaplan DL. Quantitative analysis of castration resistant prostate cancer progression through phosphoproteome signaling. BMC Cancer. 2014;14:325. doi: 10.1186/1471-2407-14-325 24885093

16. Jiang N, Hjorth-Jensen K, Hekmat O, Iglesias-Gato D, Kruse T, Wang C, et al. In vivo quantitative phosphoproteomic profiling identifies novel regulators of castration-resistant prostate cancer growth. Oncogene. 2015;34(21):2764–76. doi: 10.1038/onc.2014.206 25065596

17. Wang X, Stewart PA, Cao Q, Sang QX, Chung LW, Emmett MR, et al. Characterization of the phosphoproteome in androgen-repressed human prostate cancer cells by Fourier transform ion cyclotron resonance mass spectrometry. J Proteome Res. 2011;10(9):3920–8. doi: 10.1021/pr2000144 21786837

18. Drake JM, Paull EO, Graham NA, Lee JK, Smith BA, Titz B, et al. Phosphoproteome Integration Reveals Patient-Specific Networks in Prostate Cancer. Cell. 2016;166(4):1041–54. doi: 10.1016/j.cell.2016.07.007 27499020

19. Geiger T, Cox J, Ostasiewicz P, Wisniewski JR, Mann M. Super-SILAC mix for quantitative proteomics of human tumor tissue. Nat Methods. 2010;7(5):383–5. doi: 10.1038/nmeth.1446 20364148

20. Geiger T, Wisniewski JR, Cox J, Zanivan S, Kruger M, Ishihama Y, et al. Use of stable isotope labeling by amino acids in cell culture as a spike-in standard in quantitative proteomics. Nat Protoc. 2011;6(2):147–57. doi: 10.1038/nprot.2010.192 21293456

21. Shevchenko A, Wilm M, Vorm O, Jensen ON, Podtelejnikov AV, Neubauer G, et al. A strategy for identifying gel-separated proteins in sequence databases by MS alone. Biochem Soc Trans. 1996;24(3):893–6. doi: 10.1042/bst0240893 8878870

22. Deutsch EW, Csordas A, Sun Z, Jarnuczak A, Perez-Riverol Y, Ternent T, et al. The ProteomeXchange consortium in 2017: supporting the cultural change in proteomics public data deposition. Nucleic Acids Res. 2017;45(D1):D1100–D1106. doi: 10.1093/nar/gkw936 27924013

23. Cox J, Matic I, Hilger M, Nagaraj N, Selbach M, Olsen JV, et al. A practical guide to the MaxQuant computational platform for SILAC-based quantitative proteomics. Nat Protoc. 2009;4(5):698–705. doi: 10.1038/nprot.2009.36 19373234

24. Cox J, Neuhauser N, Michalski A, Scheltema RA, Olsen JV, Mann M. Andromeda: a peptide search engine integrated into the MaxQuant environment. J Proteome Res. 2011;10(4):1794–805. doi: 10.1021/pr101065j 21254760

25. Olsen JV, Ong SE, Mann M. Trypsin cleaves exclusively C-terminal to arginine and lysine residues. Mol Cell Proteomics. 2004;3(6):608–14. doi: 10.1074/mcp.T400003-MCP200 15034119

26. Cox J, Mann M. 1D and 2D annotation enrichment: a statistical method integrating quantitative proteomics with complementary high-throughput data. BMC Bioinformatics. 2012;13 Suppl 16:S12. doi: 10.1186/1471-2105-13-S16-S12 23176165

27. RCoreTeam. R: A language and environment for statistical computing.; 2015.

28. Benjamini Y, Hochberg Y. Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing. Journal of the Royal Statistical Society Series B (Methodological). 1995;Vol. 57(N°1):289–300. doi: 10.1111/j.2517-6161.1995.tb02031.x

29. Reimand J, Arak T, Adler P, Kolberg L, Reisberg S, Peterson H, et al. g:Profiler-a web server for functional interpretation of gene lists (2016 update). Nucleic Acids Res. 2016;44(W1):W83–9. doi: 10.1093/nar/gkw199 27098042

30. Martignetti L, Calzone L, Bonnet E, Barillot E, Zinovyev A. ROMA: Representation and Quantification of Module Activity from Target Expression Data. Front Genet. 2016;7:18. doi: 10.3389/fgene.2016.00018 26925094

31. Fabregat A, Sidiropoulos K, Garapati P, Gillespie M, Hausmann K, Haw R, et al. The Reactome pathway Knowledgebase. Nucleic Acids Res. 2016;44(D1):D481–7. doi: 10.1093/nar/gkv1351 26656494

32. Liberzon A, Birger C, Thorvaldsdottir H, Ghandi M, Mesirov JP, Tamayo P. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 2015;1(6):417–425. doi: 10.1016/j.cels.2015.12.004 26771021

33. Casado P, Rodriguez-Prados JC, Cosulich SC, Guichard S, Vanhaesebroeck B, Joel S, et al. Kinase-substrate enrichment analysis provides insights into the heterogeneity of signaling pathway activation in leukemia cells. Sci Signal. 2013;6(268):rs6. doi: 10.1126/scisignal.2003573 23532336

34. Ruepp A, Waegele B, Lechner M, Brauner B, Dunger-Kaltenbach I, Fobo G, et al. CORUM: the comprehensive resource of mammalian protein complexes–2009. Nucleic Acids Res. 2010;38(Database issue):D497–501. doi: 10.1093/nar/gkp914 19884131

35. Drew K, Lee C, Huizar RL, Tu F, Borgeson B, McWhite CD, et al. Integration of over 9,000 mass spectrometry experiments builds a global map of human protein complexes. Mol Syst Biol. 2017;13(6):932. doi: 10.15252/msb.20167490 28596423

36. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504. doi: 10.1101/gr.1239303 14597658

37. Avances C, Georget V, Terouanne B, Orio F, Cussenot O, Mottet N, et al. Human prostatic cell line PNT1A, a useful tool for studying androgen receptor transcriptional activity and its differential subnuclear localization in the presence of androgens and antiandrogens. Mol Cell Endocrinol. 2001;184(1-2):13–24. doi: 10.1016/s0303-7207(01)00669-4 11694337

38. Russell PJ, Kingsley EA. Human prostate cancer cell lines. Methods Mol Med. 2003;81:21–39. doi: 10.1385/1-59259-372-0:21 12725112

39. Webber MM, Bello D, Quader S. Immortalized and tumorigenic adult human prostatic epithelial cell lines: characteristics and applications Part 2. Tumorigenic cell lines. Prostate. 1997;30(1):58–64. doi: 10.1002/(sici)1097-0045(19970101)30:1<58::aid-pros9>3.0.co;2-h 9018337

40. Mitchell S, Abel P, Ware M, Stamp G, Lalani E. Phenotypic and genotypic characterization of commonly used human prostatic cell lines. BJU Int. 2000;85(7):932–44. doi: 10.1046/j.1464-410x.2000.00606.x 10792179

41. Mann M. Functional and quantitative proteomics using SILAC. Nat Rev Mol Cell Biol. 2006;7(12):952–8. doi: 10.1038/nrm2067 17139335

42. Ong SE, Blagoev B, Kratchmarova I, Kristensen DB, Steen H, Pandey A, et al. Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics. Mol Cell Proteomics. 2002;1(5):376–86. doi: 10.1074/mcp.m200025-mcp200 12118079

43. Nagaraj N, Wisniewski JR, Geiger T, Cox J, Kircher M, Kelso J, et al. Deep proteome and transcriptome mapping of a human cancer cell line. Mol Syst Biol. 2011;7:548. doi: 10.1038/msb.2011.81 22068331

44. Degeorges A, Hoffschir F, Cussenot O, Gauville C, Le Duc A, Dutrillaux B, et al. Recurrent cytogenetic alterations of prostate carcinoma and amplification of c-myc or epidermal growth factor receptor in subclones of immortalized PNT1 human prostate epithelial cell line. Int J Cancer. 1995;62(6):724–31. doi: 10.1002/ijc.2910620613 7558421

45. Uhlen M, Bjorling E, Agaton C, Szigyarto CA, Amini B, Andersen E, et al. A human protein atlas for normal and cancer tissues based on antibody proteomics. Mol Cell Proteomics. 2005;4(12):1920–32. doi: 10.1074/mcp.M500279-MCP200 16127175

46. Gilad R, Meir K, Stein I, German L, Pikarsky E, Mabjeesh NJ. High SEPT9_i1 protein expression is associated with high-grade prostate cancers. PLoS One. 2015;10(4):e0124251. doi: 10.1371/journal.pone.0124251 25898316

47. Leung WK, Ching AK, Chan AW, Poon TC, Mian H, Wong AS, et al. A novel interplay between oncogenic PFTK1 protein kinase and tumor suppressor TAGLN2 in the control of liver cancer cell motility. Oncogene. 2011;30(44):4464–75. doi: 10.1038/onc.2011.161 21577206

48. Garber K. Energy deregulation: licensing tumors to grow. Science. 2006;312(5777):1158–9. doi: 10.1126/science.312.5777.1158 16728625

49. Jakobsen KR, Sorensen E, Brondum KK, Daugaard TF, Thomsen R, Nielsen AL. Direct RNA sequencing mediated identification of mRNA localized in protrusions of human MDA-MB-231 metastatic breast cancer cells. J Mol Signal. 2013;8(1):9. doi: 10.1186/1750-2187-8-9 24004954

50. Mayor R, Carmona-Fontaine C. Keeping in touch with contact inhibition of locomotion. Trends Cell Biol. 2010;20(6):319–28. doi: 10.1016/j.tcb.2010.03.005 20399659

51. Azmi AS, Bao B, Sarkar FH. Exosomes in cancer development, metastasis, and drug resistance: a comprehensive review. Cancer Metastasis Rev. 2013;32(3-4):623–42. doi: 10.1007/s10555-013-9441-9 23709120

52. Rauschenberger L, Staar D, Thom K, Scharf C, Venz S, Homuth G, et al. Exosomal particles secreted by prostate cancer cells are potent mRNA and protein vehicles for the interference of tumor and tumor environment. Prostate. 2016;76(4):409–24. doi: 10.1002/pros.23132 26643154

53. Yam AY, Xia Y, Lin HT, Burlingame A, Gerstein M, Frydman J. Defining the TRiC/CCT interactome links chaperonin function to stabilization of newly made proteins with complex topologies. Nat Struct Mol Biol. 2008;15(12):1255–62. doi: 10.1038/nsmb.1515 19011634

54. Roh SH, Kasembeli M, Bakthavatsalam D, Chiu W, Tweardy DJ. Contribution of the Type II Chaperonin, TRiC/CCT, to Oncogenesis. Int J Mol Sci. 2015;16(11):26706–20. doi: 10.3390/ijms161125975 26561808

55. Trinidad AG, Muller PA, Cuellar J, Klejnot M, Nobis M, Valpuesta JM, et al. Interaction of p53 with the CCT complex promotes protein folding and wild-type p53 activity. Mol Cell. 2013;50(6):805–17. doi: 10.1016/j.molcel.2013.05.002 23747015

56. Bi J, Huang A, Liu T, Zhang T, Ma H. Expression of DNA damage checkpoint 53BP1 is correlated with prognosis, cell proliferation and apoptosis in colorectal cancer. Int J Clin Exp Pathol. 2015;8(6):6070–82. 26261485

57. Harding SM, Bristow RG. Discordance between phosphorylation and recruitment of 53BP1 in response to DNA double-strand breaks. Cell Cycle. 2012;11(7):1432–44. doi: 10.4161/cc.19824 22421153

58. Savitsky K, Ziv Y, Bar-Shira A, Gilad S, Tagle DA, Smith S, et al. A human gene (DDX10) encoding a putative DEAD-box RNA helicase at 11q22-q23. Genomics. 1996;33(2):199–206. doi: 10.1006/geno.1996.0184 8660968

59. Yang L, Lin C, Liu ZR. Phosphorylations of DEAD box p68 RNA helicase are associated with cancer development and cell proliferation. Mol Cancer Res. 2005;3(6):355–63. doi: 10.1158/1541-7786.MCR-05-0022 15972854

60. Gustafson EA, Wessel GM. DEAD-box helicases: posttranslational regulation and function. Biochem Biophys Res Commun. 2010;395(1):1–6. doi: 10.1016/j.bbrc.2010.02.172 20206133

61. Lassi K, Dawson NA. Update on castrate-resistant prostate cancer: 2010. Curr Opin Oncol. 2010;22(3):263–7. doi: 10.1097/CCO.0b013e3283380939 20177381

62. Liberti MV, Locasale JW. The Warburg Effect: How Does it Benefit Cancer Cells? Trends Biochem Sci. 2016;41(3):211–8. doi: 10.1016/j.tibs.2015.12.001 26778478

63. Thiery JP, Sleeman JP. Complex networks orchestrate epithelial-mesenchymal transitions. Nat Rev Mol Cell Biol. 2006;7(2):131–42. doi: 10.1038/nrm1835 16493418

64. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74. doi: 10.1016/j.cell.2011.02.013 21376230

65. Yang CS, Melhuish TA, Spencer A, Ni L, Hao Y, Jividen K, et al. The protein kinase C super-family member PKN is regulated by mTOR and influences differentiation during prostate cancer progression. Prostate. 2017;77(15):1452–1467. doi: 10.1002/pros.23400 28875501

66. Williams K, Ghosh R, Giridhar PV, Gu G, Case T, Belcher SM, et al. Inhibition of stathmin1 accelerates the metastatic process. Cancer Res. 2012;72(20):5407–17. doi: 10.1158/0008-5472.CAN-12-1158 22915755

67. Plouffe SW, Meng Z, Lin KC, Lin B, Hong AW, Chun JV, et al. Characterization of Hippo Pathway Components by Gene Inactivation. Mol Cell. 2016;64(5):993–1008. doi: 10.1016/j.molcel.2016.10.034 27912098

68. Park JM, Park JH, Mun DG, Bae J, Jung JH, Back S, et al. Integrated analysis of global proteome, phosphoproteome, and glycoproteome enables complementary interpretation of disease-related protein networks. Scientific Reports. 2015;5:18189. doi: 10.1038/srep18189 26657352

69. Tan H, Yang K, Li Y, Shaw TI, Wang Y, Blanco DB, et al. Integrative Proteomics and Phosphoproteomics Profiling Reveals Dynamic Signaling Networks and Bioenergetics Pathways Underlying T Cell Activation. Immunity. 2017;46(3):488–503. doi: 10.1016/j.immuni.2017.02.010 28285833

70. Katada K, Tomonaga T, Satoh M, Matsushita K, Tonoike Y, Kodera Y, et al. Plectin promotes migration and invasion of cancer cells and is a novel prognostic marker for head and neck squamous cell carcinoma. J Proteomics. 2012;75(6):1803–15. doi: 10.1016/j.jprot.2011.12.018 22245045

71. Sutoh Yoneyama M, Hatakeyama S, Habuchi T, Inoue T, Nakamura T, Funyu T, et al. Vimentin intermediate filament and plectin provide a scaffold for invadopodia, facilitating cancer cell invasion and extravasation for metastasis. Eur J Cell Biol. 2014;93(4):157–69. doi: 10.1016/j.ejcb.2014.03.002 24810881

72. Burch TC, Watson MT, Nyalwidhe JO. Variable metastatic potentials correlate with differential plectin and vimentin expression in syngeneic androgen independent prostate cancer cells. PLoS One. 2013;8(5):e65005. doi: 10.1371/journal.pone.0065005 23717685

73. Luo Y, Kong F, Wang Z, Chen D, Liu Q, Wang T, et al. Loss of ASAP3 destabilizes cytoskeletal protein ACTG1 to suppress cancer cell migration. Mol Med Rep. 2014;9(2):387–94. doi: 10.3892/mmr.2013.1831 24284654

74. Bretscher A, Edwards K, Fehon RG. ERM proteins and merlin: integrators at the cell cortex. Nat Rev Mol Cell Biol. 2002;3(8):586–99. doi: 10.1038/nrm882 12154370

75. Saygideger-Kont Y, Minas TZ, Jones H, Hour S, Celik H, Temel I, et al. Ezrin Enhances EGFR Signaling and Modulates Erlotinib Sensitivity in Non-Small Cell Lung Cancer Cells. Neoplasia. 2016;18(2):111–20. doi: 10.1016/j.neo.2016.01.002 26936397

76. Soave D, Corvol H, Panjwani N, Gong J, Li W, Boelle PY, et al. A Joint Location-Scale Test Improves Power to Detect Associated SNPs, Gene Sets, and Pathways. Am J Hum Genet. 2015;97(1):125–38. doi: 10.1016/j.ajhg.2015.05.015 26140448

77. Kuang XY, Jiang HS, Li K, Zheng YZ, Liu YR, Qiao F, et al. The phosphorylation-specific association of STMN1 with GRP78 promotes breast cancer metastasis. Cancer Lett. 2016;377(1):87–96. doi: 10.1016/j.canlet.2016.04.035 27130664

78. Germann S, Gratadou L, Dutertre M, Auboeuf D. Splicing programs and cancer. J Nucleic Acids. 2012;2012:269570. doi: 10.1155/2012/269570 22132318

79. Munkley J, Livermore K, Rajan P, Elliott DJ. RNA splicing and splicing regulator changes in prostate cancer pathology. Hum Genet. 2017;. doi: 10.1007/s00439-017-1792-9 28382513

80. Chen J, Zhou J, Sanders CK, Nolan JP, Cai H. A surface display yeast two-hybrid screening system for high-throughput protein interactome mapping. Anal Biochem. 2009;390(1):29–37. doi: 10.1016/j.ab.2009.03.013 19298787

81. Naro C, Sette C. Phosphorylation-Mediated Regulation of Alternative Splicing in Cancer. Int J Cell Biol. 2013;2013:151839. doi: 10.1155/2013/151839 24069033

82. Molina H, Horn DM, Tang N, Mathivanan S, Pandey A. Global proteomic profiling of phosphopeptides using electron transfer dissociation tandem mass spectrometry. Proc Natl Acad Sci U S A. 2007;104(7):2199–204. doi: 10.1073/pnas.0611217104 17287340

83. Mermoud JE, Cohen P, Lamond AI. Ser/Thr-specific protein phosphatases are required for both catalytic steps of pre-mRNA splicing. Nucleic Acids Res. 1992;20(20):5263–9. doi: 10.1093/nar/20.20.5263 1331983

84. Broderick S, Rehmet K, Concannon C, Nasheuer HP. Eukaryotic single-stranded DNA binding proteins: central factors in genome stability. Subcell Biochem. 2010;50:143–63. 20012581

85. Dhawan M, Ryan CJ, Ashworth A. DNA Repair Deficiency Is Common in Advanced Prostate Cancer: New Therapeutic Opportunities. Oncologist. 2016;21(8):940–5. doi: 10.1634/theoncologist.2016-0135 27317574

86. Montecucco A, Biamonti G. Pre-mRNA processing factors meet the DNA damage response. Front Genet. 2013;4:102. doi: 10.3389/fgene.2013.00102 23761808

87. Jacq X, Kemp M, Martin NM, Jackson SP. Deubiquitylating enzymes and DNA damage response pathways. Cell Biochem Biophys. 2013;67(1):25–43. doi: 10.1007/s12013-013-9635-3 23712866

88. Yuan F, Li G, Tong T. Nucleolar and coiled-body phosphoprotein 1 (NOLC1) regulates the nucleolar retention of TRF2. Cell Death Discov. 2017;3:17043. doi: 10.1038/cddiscovery.2017.43 28875039

89. Fagerlund R, Kinnunen L, Kohler M, Julkunen I, Melen K. NF-kappaB is transported into the nucleus by importin alpha3 and importin alpha4. J Biol Chem. 2005;280(16):15942–51. doi: 10.1074/jbc.M500814200 15677444

90. Jin RJ, Lho Y, Connelly L, Wang Y, Yu X, Saint Jean L, et al. The nuclear factor-kappaB pathway controls the progression of prostate cancer to androgen-independent growth. Cancer Res. 2008;68(16):6762–9. doi: 10.1158/0008-5472.CAN-08-0107 18701501

91. Lai AY, Wade PA. Cancer biology and NuRD: a multifaceted chromatin remodelling complex. Nat Rev Cancer. 2011;11(8):588–96. doi: 10.1038/nrc3091 21734722

92. Spruijt CG, Luijsterburg MS, Menafra R, Lindeboom RG, Jansen PW, Edupuganti RR, et al. ZMYND8 Co-localizes with NuRD on Target Genes and Regulates Poly(ADP-Ribose)-Dependent Recruitment of GATAD2A/NuRD to Sites of DNA Damage. Cell Rep. 2016;17(3):783–798. doi: 10.1016/j.celrep.2016.09.037 27732854

93. Heeboll S, Borre M, Ottosen PD, Andersen CL, Mansilla F, Dyrskjot L, et al. SMARCC1 expression is upregulated in prostate cancer and positively correlated with tumour recurrence and dedifferentiation. Histol Histopathol. 2008;23(9):1069–76. doi: 10.14670/HH-23.1069 18581278

94. Hu R, Lu C, Mostaghel EA, Yegnasubramanian S, Gurel M, Tannahill C, et al. Distinct transcriptional programs mediated by the ligand-dependent full-length androgen receptor and its splice variants in castration-resistant prostate cancer. Cancer Res. 2012;72(14):3457–62. doi: 10.1158/0008-5472.CAN-11-3892 22710436

95. Wang Q, Li W, Zhang Y, Yuan X, Xu K, Yu J, et al. Androgen receptor regulates a distinct transcription program in androgen-independent prostate cancer. Cell. 2009;138(2):245–56. doi: 10.1016/j.cell.2009.04.056 19632176

96. Faivre EJ, Wilcox D, Lin X, Hessler P, Torrent M, He W, et al. Exploitation of Castration-Resistant Prostate Cancer Transcription Factor Dependencies by the Novel BET Inhibitor ABBV-075. Mol Cancer Res. 2017;15(1):35–44. doi: 10.1158/1541-7786.MCR-16-0221 27707886

97. Pienta KJ, Abate-Shen C, Agus DB, Attar RM, Chung LW, Greenberg NM, et al. The current state of preclinical prostate cancer animal models. Prostate. 2008;68(6):629–39. doi: 10.1002/pros.20726 18213636

98. Cunningham D, You Z. In vitro and in vivo model systems used in prostate cancer research. J Biol Methods. 2015;2(1). doi: 10.14440/jbm.2015.63 26146646

99. Ardito F, Giuliani M, Perrone D, Troiano G, Lo Muzio L. The crucial role of protein phosphorylation in cell signaling and its use as targeted therapy (Review). Int J Mol Med. 2017;40(2):271–280. doi: 10.3892/ijmm.2017.3036 28656226


Článek vyšel v časopise

PLOS One


2019 Číslo 11
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

KOST
Koncepce osteologické péče pro gynekology a praktické lékaře
nový kurz
Autoři: MUDr. František Šenk

Sekvenční léčba schizofrenie
Autoři: MUDr. Jana Hořínková

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

Svět praktické medicíny 5/2023 (znalostní test z časopisu)

Imunopatologie? … a co my s tím???
Autoři: doc. MUDr. Helena Lahoda Brodská, Ph.D.

Všechny kurzy
Kurzy Podcasty Doporučená témata Časopisy
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#