#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Epstein-Barr Virus Proteins EBNA3A and EBNA3C Together Induce Expression of the Oncogenic MicroRNA Cluster miR-221/miR-222 and Ablate Expression of Its Target p57


A relatively unbiased screen of human microRNAs (miRs) revealed that in EBV-transformed B cells, a miR cluster, miR-221/miR-222, that is frequently up-regulated in cancer, is induced by the latent EBV only if the viral nuclear proteins EBNA3A and EBNA3C are both expressed. The same two EBV proteins silence a tumour-suppressor miR cluster miR-143/miR-145. The induction of miR-221/miR-222 results from the activation of a long non-coding primary RNA (pri-miR) via long-range chromatin looping between enhancer elements that bind EBNA3A and EBNA3C and the transcription start site of the pri-miR. A well-established target of miR-221/miR-222 is the cyclin-dependent kinase (CDK) inhibitor p57KIP2, which, because it can inactivate various CDKs, can inhibit cell proliferation—but might have additional functions in B cells. Since EBNA3A and EBNA3C also cooperate to repress the expression of at least two other inhibitors of CDKs (p16INK4a and p15INK4b), this implies a degree of functional redundancy in the deregulation of cell cycle checkpoints by latent EBV. This study has shown for the first time that this capacity to reduce expression of multiple cell cycle inhibitors results not only from direct repression of protein-encoding genes, but also the activation of a long non-coding RNA and cluster of oncogenic miRs.


Vyšlo v časopise: Epstein-Barr Virus Proteins EBNA3A and EBNA3C Together Induce Expression of the Oncogenic MicroRNA Cluster miR-221/miR-222 and Ablate Expression of Its Target p57. PLoS Pathog 11(7): e32767. doi:10.1371/journal.ppat.1005031
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1005031

Souhrn

A relatively unbiased screen of human microRNAs (miRs) revealed that in EBV-transformed B cells, a miR cluster, miR-221/miR-222, that is frequently up-regulated in cancer, is induced by the latent EBV only if the viral nuclear proteins EBNA3A and EBNA3C are both expressed. The same two EBV proteins silence a tumour-suppressor miR cluster miR-143/miR-145. The induction of miR-221/miR-222 results from the activation of a long non-coding primary RNA (pri-miR) via long-range chromatin looping between enhancer elements that bind EBNA3A and EBNA3C and the transcription start site of the pri-miR. A well-established target of miR-221/miR-222 is the cyclin-dependent kinase (CDK) inhibitor p57KIP2, which, because it can inactivate various CDKs, can inhibit cell proliferation—but might have additional functions in B cells. Since EBNA3A and EBNA3C also cooperate to repress the expression of at least two other inhibitors of CDKs (p16INK4a and p15INK4b), this implies a degree of functional redundancy in the deregulation of cell cycle checkpoints by latent EBV. This study has shown for the first time that this capacity to reduce expression of multiple cell cycle inhibitors results not only from direct repression of protein-encoding genes, but also the activation of a long non-coding RNA and cluster of oncogenic miRs.


Zdroje

1. Young LS, Rickinson AB (2004) Epstein-Barr virus: 40 years on. Nat Rev Cancer 4: 757–768. 15510157

2. Thorley-Lawson DA, Gross A (2004) Persistence of the Epstein-Barr virus and the origins of associated lymphomas. N Engl J Med 350: 1328–1337. 15044644

3. Thorley-Lawson DA, Hawkins JB, Tracy SI, Shapiro M (2013) The pathogenesis of Epstein-Barr virus persistent infection. Current opinion in virology.

4. Forte E, Luftig MA (2011) The role of microRNAs in Epstein-Barr virus latency and lytic reactivation. Microbes and infection / Institut Pasteur 13: 1156–1167. doi: 10.1016/j.micinf.2011.07.007 21835261

5. Baer R, Bankier AT, Biggin MD, Deininger PL, Farrell PJ, et al. (1984) DNA sequence and expression of the B95-8 Epstein-Barr virus genome. Nature 310: 207–211. 6087149

6. Hennessy K, Wang F, Bushman EW, Kieff E (1986) Definitive identification of a member of the Epstein-Barr virus nuclear protein 3 family. Proceedings of the National Academy of Sciences of the United States of America 83: 5693–5697. 3016714

7. Tomkinson B, Kieff E (1992) Use of second-site homologous recombination to demonstrate that Epstein-Barr virus nuclear protein 3B is not important for lymphocyte infection or growth transformation in vitro. J Virol 66: 2893–2903. 1313908

8. Tomkinson B, Kieff E (1992) Second-site homologous recombination in Epstein-Barr virus: insertion of type 1 EBNA 3 genes in place of type 2 has no effect on in vitro infection. Journal of virology 66: 780–789. 1309912

9. Maruo S, Wu Y, Ishikawa S, Kanda T, Iwakiri D, et al. (2006) Epstein-Barr virus nuclear protein EBNA3C is required for cell cycle progression and growth maintenance of lymphoblastoid cells. Proc Natl Acad Sci U S A 103: 19500–19505. 17159137

10. Maruo S, Johannsen E, Illanes D, Cooper A, Kieff E (2003) Epstein-Barr Virus nuclear protein EBNA3A is critical for maintaining lymphoblastoid cell line growth. J Virol 77: 10437–10447. 12970429

11. Hertle ML, Popp C, Petermann S, Maier S, Kremmer E, et al. (2009) Differential gene expression patterns of EBV infected EBNA-3A positive and negative human B lymphocytes. PLoS Pathog 5: e1000506. doi: 10.1371/journal.ppat.1000506 19578441

12. Skalska L, White RE, Franz M, Ruhmann M, Allday MJ (2010) Epigenetic repression of p16(INK4A) by latent Epstein-Barr virus requires the interaction of EBNA3A and EBNA3C with CtBP. PLoS pathogens 6: e1000951. doi: 10.1371/journal.ppat.1000951 20548956

13. White RE, Ramer PC, Naresh KN, Meixlsperger S, Pinaud L, et al. (2012) EBNA3B-deficient EBV promotes B cell lymphomagenesis in humanized mice and is found in human tumors. The Journal of clinical investigation 122: 1487–1502. doi: 10.1172/JCI58092 22406538

14. White RE, Groves IJ, Turro E, Yee J, Kremmer E, et al. (2010) Extensive co-operation between the Epstein-Barr virus EBNA3 proteins in the manipulation of host gene expression and epigenetic chromatin modification. PloS one 5: e13979. doi: 10.1371/journal.pone.0013979 21085583

15. Robertson ES, Grossman S, Johannsen E, Miller C, Lin J, et al. (1995) Epstein-Barr virus nuclear protein 3C modulates transcription through interaction with the sequence-specific DNA-binding protein J kappa. Journal of virology 69: 3108–3116. 7707539

16. Robertson ES, Lin J, Kieff E (1996) The amino-terminal domains of Epstein-Barr virus nuclear proteins 3A, 3B, and 3C interact with RBPJ(kappa). Journal of virology 70: 3068–3074. 8627785

17. Jimenez-Ramirez C, Brooks AJ, Forshell LP, Yakimchuk K, Zhao B, et al. (2006) Epstein-Barr virus EBNA-3C is targeted to and regulates expression from the bidirectional LMP-1/2B promoter. Journal of virology 80: 11200–11208. 16956945

18. Zhao B, Sample CE (2000) Epstein-barr virus nuclear antigen 3C activates the latent membrane protein 1 promoter in the presence of Epstein-Barr virus nuclear antigen 2 through sequences encompassing an spi-1/Spi-B binding site. Journal of virology 74: 5151–5160. 10799590

19. Jiang S, Willox B, Zhou H, Holthaus AM, Wang A, et al. (2014) Epstein-Barr virus nuclear antigen 3C binds to BATF/IRF4 or SPI1/IRF4 composite sites and recruits Sin3A to repress CDKN2A. Proceedings of the National Academy of Sciences of the United States of America 111: 421–426. doi: 10.1073/pnas.1321704111 24344258

20. McClellan MJ, Wood CD, Ojeniyi O, Cooper TJ, Kanhere A, et al. (2013) Modulation of enhancer looping and differential gene targeting by Epstein-Barr virus transcription factors directs cellular reprogramming. PLoS pathogens 9: e1003636. doi: 10.1371/journal.ppat.1003636 24068937

21. Harth-Hertle ML, Scholz BA, Erhard F, Glaser LV, Dolken L, et al. (2013) Inactivation of intergenic enhancers by EBNA3A initiates and maintains polycomb signatures across a chromatin domain encoding CXCL10 and CXCL9. PLoS pathogens 9: e1003638. doi: 10.1371/journal.ppat.1003638 24068939

22. Cotter MA 2nd, Robertson ES (2000) Modulation of histone acetyltransferase activity through interaction of epstein-barr nuclear antigen 3C with prothymosin alpha. Molecular and cellular biology 20: 5722–5735. 10891508

23. Hickabottom M, Parker GA, Freemont P, Crook T, Allday MJ (2002) Two nonconsensus sites in the Epstein-Barr virus oncoprotein EBNA3A cooperate to bind the co-repressor carboxyl-terminal-binding protein (CtBP). J Biol Chem 277: 47197–47204. 12372828

24. Knight JS, Lan K, Subramanian C, Robertson ES (2003) Epstein-Barr virus nuclear antigen 3C recruits histone deacetylase activity and associates with the corepressors mSin3A and NCoR in human B-cell lines. J Virol 77: 4261–4272. 12634383

25. Radkov SA, Touitou R, Brehm A, Rowe M, West M, et al. (1999) Epstein-Barr virus nuclear antigen 3C interacts with histone deacetylase to repress transcription. J Virol 73: 5688–5697. 10364319

26. Touitou R, Hickabottom M, Parker G, Crook T, Allday MJ (2001) Physical and functional interactions between the corepressor CtBP and the Epstein-Barr virus nuclear antigen EBNA3C. J Virol 75: 7749–7755. 11462050

27. Paschos K, Parker GA, Watanatanasup E, White RE, Allday MJ (2012) BIM promoter directly targeted by EBNA3C in polycomb-mediated repression by EBV. Nucleic acids research 40: 7233–7246. doi: 10.1093/nar/gks391 22584624

28. McClellan MJ, Khasnis S, Wood CD, Palermo RD, Schlick SN, et al. (2012) Downregulation of integrin receptor-signaling genes by Epstein-Barr virus EBNA 3C via promoter-proximal and-distal binding elements. Journal of virology 86: 5165–5178. doi: 10.1128/JVI.07161-11 22357270

29. Schmidt SC, Jiang S, Zhou H, Willox B, Holthaus AM, et al. (2015) Epstein-Barr virus nuclear antigen 3A partially coincides with EBNA3C genome-wide and is tethered to DNA through BATF complexes. Proceedings of the National Academy of Sciences of the United States of America 112: 554–559. doi: 10.1073/pnas.1422580112 25540416

30. Skalska L, White RE, Parker GA, Sinclair AJ, Paschos K, et al. (2013) Induction of p16(INK4a) is the major barrier to proliferation when Epstein-Barr virus (EBV) transforms primary B cells into lymphoblastoid cell lines. PLoS pathogens 9: e1003187. doi: 10.1371/journal.ppat.1003187 23436997

31. Allday MJ (2013) EBV finds a polycomb-mediated, epigenetic solution to the problem of oncogenic stress responses triggered by infection. Frontiers in genetics 4: 212. doi: 10.3389/fgene.2013.00212 24167519

32. Bazot Q, Deschamps T, Tafforeau L, Siouda M, Leblanc P, et al. (2014) Epstein-Barr virus nuclear antigen 3A protein regulates CDKN2B transcription via interaction with MIZ-1. Nucleic acids research 42: 9700–9716. doi: 10.1093/nar/gku697 25092922

33. Fabian MR, Sonenberg N, Filipowicz W (2010) Regulation of mRNA translation and stability by microRNAs. Annual review of biochemistry 79: 351–379. doi: 10.1146/annurev-biochem-060308-103103 20533884

34. Wilczynska A, Bushell M (2015) The complexity of miRNA-mediated repression. Cell death and differentiation 22: 22–33. doi: 10.1038/cdd.2014.112 25190144

35. Lim LP, Lau NC, Garrett-Engele P, Grimson A, Schelter JM, et al. (2005) Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature 433: 769–773. 15685193

36. Calin GA, Croce CM (2006) MicroRNA signatures in human cancers. Nature reviews Cancer 6: 857–866. 17060945

37. Esquela-Kerscher A, Slack FJ (2006) Oncomirs—microRNAs with a role in cancer. Nature reviews Cancer 6: 259–269. 16557279

38. Feldman ER, Kara M, Coleman CB, Grau KR, Oko LM, et al. (2014) Virus-encoded microRNAs facilitate gammaherpesvirus latency and pathogenesis in vivo. mBio 5: e00981–00914. doi: 10.1128/mBio.00981-14 24865551

39. Zhao Y, Xu H, Yao Y, Smith LP, Kgosana L, et al. (2011) Critical role of the virus-encoded microRNA-155 ortholog in the induction of Marek's disease lymphomas. PLoS pathogens 7: e1001305. doi: 10.1371/journal.ppat.1001305 21383974

40. Kuzembayeva M, Hayes M, Sugden B (2014) Multiple functions are mediated by the miRNAs of Epstein-Barr virus. Current opinion in virology 7: 61–65. doi: 10.1016/j.coviro.2014.04.003 24814666

41. Moody R, Zhu Y, Huang Y, Cui X, Jones T, et al. (2013) KSHV microRNAs mediate cellular transformation and tumorigenesis by redundantly targeting cell growth and survival pathways. PLoS pathogens 9: e1003857. doi: 10.1371/journal.ppat.1003857 24385912

42. Linnstaedt SD, Gottwein E, Skalsky RL, Luftig MA, Cullen BR (2010) Virally induced cellular microRNA miR-155 plays a key role in B-cell immortalization by Epstein-Barr virus. Journal of virology 84: 11670–11678. doi: 10.1128/JVI.01248-10 20844043

43. Sun T, Yang M, Kantoff P, Lee GS (2009) Role of microRNA-221/-222 in cancer development and progression. Cell cycle 8: 2315–2316. 19625765

44. Pallante P, Visone R, Ferracin M, Ferraro A, Berlingieri MT, et al. (2006) MicroRNA deregulation in human thyroid papillary carcinomas. Endocrine-related cancer 13: 497–508. 16728577

45. Ciafre SA, Galardi S, Mangiola A, Ferracin M, Liu CG, et al. (2005) Extensive modulation of a set of microRNAs in primary glioblastoma. Biochemical and biophysical research communications 334: 1351–1358. 16039986

46. Galardi S, Mercatelli N, Giorda E, Massalini S, Frajese GV, et al. (2007) miR-221 and miR-222 expression affects the proliferation potential of human prostate carcinoma cell lines by targeting p27Kip1. The Journal of biological chemistry 282: 23716–23724. 17569667

47. Galardi S, Mercatelli N, Farace MG, Ciafre SA (2011) NF-kB and c-Jun induce the expression of the oncogenic miR-221 and miR-222 in prostate carcinoma and glioblastoma cells. Nucleic acids research 39: 3892–3902. doi: 10.1093/nar/gkr006 21245048

48. Gottardo F, Liu CG, Ferracin M, Calin GA, Fassan M, et al. (2007) Micro-RNA profiling in kidney and bladder cancers. Urologic oncology 25: 387–392. 17826655

49. Lee EJ, Gusev Y, Jiang J, Nuovo GJ, Lerner MR, et al. (2007) Expression profiling identifies microRNA signature in pancreatic cancer. International journal of cancer Journal international du cancer 120: 1046–1054. 17149698

50. Pineau P, Volinia S, McJunkin K, Marchio A, Battiston C, et al. (2010) miR-221 overexpression contributes to liver tumorigenesis. Proceedings of the National Academy of Sciences of the United States of America 107: 264–269. doi: 10.1073/pnas.0907904107 20018759

51. Rommer A, Steinleitner K, Hackl H, Schneckenleithner C, Engelmann M, et al. (2013) Overexpression of primary microRNA 221/222 in acute myeloid leukemia. BMC cancer 13: 364. doi: 10.1186/1471-2407-13-364 23895238

52. Lawrie CH, Soneji S, Marafioti T, Cooper CD, Palazzo S, et al. (2007) MicroRNA expression distinguishes between germinal center B cell-like and activated B cell-like subtypes of diffuse large B cell lymphoma. International journal of cancer Journal international du cancer 121: 1156–1161. 17487835

53. Malumbres R, Sarosiek KA, Cubedo E, Ruiz JW, Jiang X, et al. (2009) Differentiation stage-specific expression of microRNAs in B lymphocytes and diffuse large B-cell lymphomas. Blood 113: 3754–3764. doi: 10.1182/blood-2008-10-184077 19047678

54. Andrade TA, Evangelista AF, Campos AH, Poles WA, Borges NM, et al. (2014) A microRNA signature profile in EBV+ diffuse large B-cell lymphoma of the elderly. Oncotarget 5: 11813–11826. 25544772

55. Wang J, Liu S, Sun GP, Wang F, Zou YF, et al. (2014) Prognostic significance of microRNA-221/222 expression in cancers: evidence from 1,204 subjects. The International journal of biological markers 29: e129–141. doi: 10.5301/jbm.5000058 24474451

56. Visone R, Russo L, Pallante P, De Martino I, Ferraro A, et al. (2007) MicroRNAs (miR)-221 and miR-222, both overexpressed in human thyroid papillary carcinomas, regulate p27Kip1 protein levels and cell cycle. Endocrine-related cancer 14: 791–798. 17914108

57. Fornari F, Gramantieri L, Ferracin M, Veronese A, Sabbioni S, et al. (2008) MiR-221 controls CDKN1C/p57 and CDKN1B/p27 expression in human hepatocellular carcinoma. Oncogene 27: 5651–5661. doi: 10.1038/onc.2008.178 18521080

58. le Sage C, Nagel R, Egan DA, Schrier M, Mesman E, et al. (2007) Regulation of the p27(Kip1) tumor suppressor by miR-221 and miR-222 promotes cancer cell proliferation. The EMBO journal 26: 3699–3708. 17627278

59. Medina R, Zaidi SK, Liu CG, Stein JL, van Wijnen AJ, et al. (2008) MicroRNAs 221 and 222 bypass quiescence and compromise cell survival. Cancer research 68: 2773–2780. doi: 10.1158/0008-5472.CAN-07-6754 18413744

60. Elia L, Quintavalle M, Zhang J, Contu R, Cossu L, et al. (2009) The knockout of miR-143 and -145 alters smooth muscle cell maintenance and vascular homeostasis in mice: correlates with human disease. Cell death and differentiation 16: 1590–1598. doi: 10.1038/cdd.2009.153 19816508

61. Kent OA, Chivukula RR, Mullendore M, Wentzel EA, Feldmann G, et al. (2010) Repression of the miR-143/145 cluster by oncogenic Ras initiates a tumor-promoting feed-forward pathway. Genes & development 24: 2754–2759.

62. Bonifacio LN, Jarstfer MB (2010) MiRNA profile associated with replicative senescence, extended cell culture, and ectopic telomerase expression in human foreskin fibroblasts. PloS one 5.

63. Iio A, Nakagawa Y, Hirata I, Naoe T, Akao Y (2010) Identification of non-coding RNAs embracing microRNA-143/145 cluster. Molecular cancer 9: 136. doi: 10.1186/1476-4598-9-136 20525177

64. Takagi T, Iio A, Nakagawa Y, Naoe T, Tanigawa N, et al. (2009) Decreased expression of microRNA-143 and -145 in human gastric cancers. Oncology 77: 12–21. doi: 10.1159/000218166 19439999

65. Akao Y, Nakagawa Y, Hirata I, Iio A, Itoh T, et al. (2010) Role of anti-oncomirs miR-143 and -145 in human colorectal tumors. Cancer gene therapy 17: 398–408. doi: 10.1038/cgt.2009.88 20094072

66. Lui WO, Pourmand N, Patterson BK, Fire A (2007) Patterns of known and novel small RNAs in human cervical cancer. Cancer research 67: 6031–6043. 17616659

67. Vosa U, Vooder T, Kolde R, Vilo J, Metspalu A, et al. (2013) Meta-analysis of microRNA expression in lung cancer. International journal of cancer Journal international du cancer 132: 2884–2893. doi: 10.1002/ijc.27981 23225545

68. Zhou F, Li S, Meng HM, Qi LQ, Gu L (2013) MicroRNA and histopathological characterization of pure mucinous breast carcinoma. Cancer biology & medicine 10: 22–27.

69. Chen HC, Chen GH, Chen YH, Liao WL, Liu CY, et al. (2009) MicroRNA deregulation and pathway alterations in nasopharyngeal carcinoma. British journal of cancer 100: 1002–1011. doi: 10.1038/sj.bjc.6604948 19293812

70. Yoshino H, Seki N, Itesako T, Chiyomaru T, Nakagawa M, et al. (2013) Aberrant expression of microRNAs in bladder cancer. Nature reviews Urology 10: 396–404. doi: 10.1038/nrurol.2013.113 23712207

71. Szczyrba J, Loprich E, Wach S, Jung V, Unteregger G, et al. (2010) The microRNA profile of prostate carcinoma obtained by deep sequencing. Molecular cancer research: MCR 8: 529–538. doi: 10.1158/1541-7786.MCR-09-0443 20353999

72. Iorio MV, Visone R, Di Leva G, Donati V, Petrocca F, et al. (2007) MicroRNA signatures in human ovarian cancer. Cancer research 67: 8699–8707. 17875710

73. Xing AY, Wang B, Shi DB, Zhang XF, Gao C, et al. (2013) Deregulated expression of miR-145 in manifold human cancer cells. Experimental and molecular pathology 95: 91–97. doi: 10.1016/j.yexmp.2013.05.003 23714355

74. Akao Y, Nakagawa Y, Kitade Y, Kinoshita T, Naoe T (2007) Downregulation of microRNAs-143 and -145 in B-cell malignancies. Cancer science 98: 1914–1920. 17892514

75. Rangrez AY, Massy ZA, Metzinger-Le Meuth V, Metzinger L (2011) miR-143 and miR-145: molecular keys to switch the phenotype of vascular smooth muscle cells. Circulation Cardiovascular genetics 4: 197–205. doi: 10.1161/CIRCGENETICS.110.958702 21505201

76. Zhang C, Zhang J, Zhang A, Wang Y, Han L, et al. (2010) PUMA is a novel target of miR-221/222 in human epithelial cancers. International journal of oncology 37: 1621–1626. 21042732

77. Phatnani HP, Greenleaf AL (2006) Phosphorylation and functions of the RNA polymerase II CTD. Genes & development 20: 2922–2936.

78. Maruo S, Zhao B, Johannsen E, Kieff E, Zou J, et al. (2011) Epstein-Barr virus nuclear antigens 3C and 3A maintain lymphoblastoid cell growth by repressing p16INK4A and p14ARF expression. Proceedings of the National Academy of Sciences of the United States of America 108: 1919–1924. doi: 10.1073/pnas.1019599108 21245331

79. Pombo A, Dillon N (2015) Three-dimensional genome architecture: players and mechanisms. Nature reviews Molecular cell biology 16: 245–257. doi: 10.1038/nrm3965 25757416

80. Hutzinger R, Mrazek J, Vorwerk S, Huttenhofer A (2010) NcRNA-microchip analysis: a novel approach to identify differential expression of noncoding RNAs. RNA biology 7: 586–595. 21037422

81. Cameron JE, Fewell C, Yin Q, McBride J, Wang X, et al. (2008) Epstein-Barr virus growth/latency III program alters cellular microRNA expression. Virology 382: 257–266. doi: 10.1016/j.virol.2008.09.018 18950829

82. Cameron JE, Yin Q, Fewell C, Lacey M, McBride J, et al. (2008) Epstein-Barr virus latent membrane protein 1 induces cellular MicroRNA miR-146a, a modulator of lymphocyte signaling pathways. Journal of virology 82: 1946–1958. 18057241

83. Gunasekharan V, Laimins LA (2013) Human papillomaviruses modulate microRNA 145 expression to directly control genome amplification. Journal of virology 87: 6037–6043. doi: 10.1128/JVI.00153-13 23468503

84. Godshalk SE, Bhaduri-McIntosh S, Slack FJ (2008) Epstein-Barr virus-mediated dysregulation of human microRNA expression. Cell cycle 7: 3595–3600. 19001862

85. Tursiella ML, Bowman ER, Wanzeck KC, Throm RE, Liao J, et al. (2014) Epstein-Barr Virus Nuclear Antigen 3A Promotes Cellular Proliferation by Repression of the Cyclin-Dependent Kinase Inhibitor p21WAF1/CIP1. PLoS pathogens 10: e1004415. doi: 10.1371/journal.ppat.1004415 25275486

86. Banerjee S, Lu J, Cai Q, Sun Z, Jha HC, et al. (2014) EBNA3C augments Pim-1 mediated phosphorylation and degradation of p21 to promote B-cell proliferation. PLoS pathogens 10: e1004304. doi: 10.1371/journal.ppat.1004304 25121590

87. Sherr CJ (2012) Ink4-Arf Locus in Cancer and Aging. Wiley interdisciplinary reviews Developmental biology 1: 731–741. 22960768

88. Kelly G, Bell A, Rickinson A (2002) Epstein-Barr virus-associated Burkitt lymphomagenesis selects for downregulation of the nuclear antigen EBNA2. Nat Med 8: 1098–1104. 12219084

89. Feil R, Wagner J, Metzger D, Chambon P (1997) Regulation of Cre recombinase activity by mutated estrogen receptor ligand-binding domains. Biochemical and biophysical research communications 237: 752–757. 9299439

90. Delecluse HJ, Hilsendegen T, Pich D, Zeidler R, Hammerschmidt W (1998) Propagation and recovery of intact, infectious Epstein-Barr virus from prokaryotic to human cells. Proc Natl Acad Sci U S A 95: 8245–8250. 9653172

91. Anderton E, Yee J, Smith P, Crook T, White RE, et al. (2008) Two Epstein-Barr virus (EBV) oncoproteins cooperate to repress expression of the proapoptotic tumour-suppressor Bim: clues to the pathogenesis of Burkitt's lymphoma. Oncogene 27: 421–433. 17653091

92. Hagege H, Klous P, Braem C, Splinter E, Dekker J, et al. (2007) Quantitative analysis of chromosome conformation capture assays (3C-qPCR). Nature protocols 2: 1722–1733. 17641637

93. Tempera I, Klichinsky M, Lieberman PM (2011) EBV latency types adopt alternative chromatin conformations. PLoS pathogens 7: e1002180. doi: 10.1371/journal.ppat.1002180 21829357

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2015 Číslo 7
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

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

Všetky kurzy
Prihlásenie
Zabudnuté heslo

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

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#