#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Epstein-Barr virus subverts mevalonate and fatty acid pathways to promote infected B-cell proliferation and survival


Autoři: Liang Wei Wang aff001;  Zhonghao Wang aff002;  Ina Ersing aff002;  Luis Nobre aff006;  Rui Guo aff002;  Sizun Jiang aff002;  Stephen Trudeau aff002;  Bo Zhao aff002;  Michael P. Weekes aff006;  Benjamin E. Gewurz aff001
Působiště autorů: Graduate Program in Virology, Division of Medical Sciences, Harvard Medical School, Boston, Massachusetts, United States of America aff001;  Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America aff002;  Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America aff003;  Broad Institute of Harvard and MIT, Cambridge, Massachusetts, United States of America aff004;  Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China aff005;  Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom aff006
Vyšlo v časopise: Epstein-Barr virus subverts mevalonate and fatty acid pathways to promote infected B-cell proliferation and survival. PLoS Pathog 15(9): e32767. doi:10.1371/journal.ppat.1008030
Kategorie: Research Article
doi: https://doi.org/10.1371/journal.ppat.1008030

Souhrn

Epstein-Barr virus (EBV) causes infectious mononucleosis and is associated with multiple human malignancies. EBV drives B-cell proliferation, which contributes to the pathogenesis of multiple lymphomas. Yet, knowledge of how EBV subverts host biosynthetic pathways to transform resting lymphocytes into activated lymphoblasts remains incomplete. Using a temporal proteomic dataset of EBV primary human B-cell infection, we identified that cholesterol and fatty acid biosynthetic pathways were amongst the most highly EBV induced. Epstein-Barr nuclear antigen 2 (EBNA2), sterol response element binding protein (SREBP) and MYC each had important roles in cholesterol and fatty acid pathway induction. Unexpectedly, HMG-CoA reductase inhibitor chemical epistasis experiments revealed that mevalonate pathway production of geranylgeranyl pyrophosphate (GGPP), rather than cholesterol, was necessary for EBV-driven B-cell outgrowth, perhaps because EBV upregulated the low-density lipoprotein receptor in newly infected cells for cholesterol uptake. Chemical and CRISPR genetic analyses highlighted downstream GGPP roles in EBV-infected cell small G protein Rab activation. Rab13 was highly EBV-induced in an EBNA3-dependent manner and served as a chaperone critical for latent membrane protein (LMP) 1 and 2A trafficking and target gene activation in newly infected and in lymphoblastoid B-cells. Collectively, these studies identify highlight multiple potential therapeutic targets for prevention of EBV-transformed B-cell growth and survival.

Klíčová slova:

Biology and life sciences – Cell biology – Cellular types – Animal cells – Immune cells – Antibody-producing cells – B cells – Blood cells – White blood cells – Biochemistry – Lipids – Cholesterol – Fatty acids – Proteins – DNA-binding proteins – Transcription factors – Regulatory proteins – Biosynthesis – Organisms – Viruses – DNA viruses – Herpesviruses – Epstein-Barr virus – Microbiology – Medical microbiology – Microbial pathogens – Viral pathogens – Molecular biology – Molecular biology techniques – Molecular probe techniques – Immunoblot analysis – Genetics – Gene expression – Gene regulation – Medicine and health sciences – Immunology – Pathology and laboratory medicine – Pathogens – Research and analysis methods


Zdroje

1. Longnecker R, Kieff E and Cohen JI. Epstein-Barr Virus. In: Knipe P.M., editor. Fields Virology. 2. 6 ed. Philadelphia: Lippincott, Williams and Wilkins; 2013. p. 1898–959.

2. Shannon-Lowe C, Rickinson AB, Bell AI. Epstein-Barr virus-associated lymphomas. Philos Trans R Soc Lond B Biol Sci. 2017;372(1732).

3. LaCasce AS. Post-transplant lymphoproliferative disorders. Oncologist. 2006;11(6):674–80. doi: 10.1634/theoncologist.11-6-674 16794246

4. Green M, Michaels MG. Epstein-Barr virus infection and posttransplant lymphoproliferative disorder. Am J Transplant. 2013;13 Suppl 3:41–54; quiz

5. Makata AM, Toriyama K, Kamidigo NO, Eto H, Itakura H. The pattern of pediatric solid malignant tumors in western Kenya, east Africa, 1979–1994: an analysis based on histopathologic study. Am J Trop Med Hyg. 1996;54(4):343–7. doi: 10.4269/ajtmh.1996.54.343 8615444

6. Daniel E. Burkitt's lymphoma in Ethiopian children. Trop Geogr Med. 1990;42(3):255–60. 2293434

7. Hammerschmidt W, Sugden B. Epstein-Barr virus sustains Burkitt's lymphomas and Hodgkin's disease. Trends Mol Med. 2004;10(7):331–6. doi: 10.1016/j.molmed.2004.05.006 15242681

8. 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

9. Robinson JE, Smith D, Niederman J. Plasmacytic differentiation of circulating Epstein-Barr virus-infected B lymphocytes during acute infectious mononucleosis. J Exp Med. 1981;153(2):235–44. doi: 10.1084/jem.153.2.235 6264016

10. Taylor GS, Long HM, Brooks JM, Rickinson AB, Hislop AD. The immunology of Epstein-Barr virus-induced disease. Annu Rev Immunol. 2015;33:787–821. doi: 10.1146/annurev-immunol-032414-112326 25706097

11. Munz C. Epstein Barr virus—a tumor virus that needs cytotoxic lymphocytes to persist asymptomatically. Curr Opin Virol. 2016;20:34–9. doi: 10.1016/j.coviro.2016.08.010 27591678

12. Thorley-Lawson DA. EBV Persistence—Introducing the Virus. Curr Top Microbiol Immunol. 2015;390(Pt 1):151–209. doi: 10.1007/978-3-319-22822-8_8 26424647

13. Kang MS, Kieff E. Epstein-Barr virus latent genes. Exp Mol Med. 2015;47:e131. doi: 10.1038/emm.2014.84 25613728

14. Nikitin PA, Yan CM, Forte E, Bocedi A, Tourigny JP, White RE, et al. An ATM/Chk2-mediated DNA damage-responsive signaling pathway suppresses Epstein-Barr virus transformation of primary human B cells. Cell Host Microbe. 2010;8(6):510–22. doi: 10.1016/j.chom.2010.11.004 21147465

15. Kaiser C, Laux G, Eick D, Jochner N, Bornkamm GW, Kempkes B. The proto-oncogene c-myc is a direct target gene of Epstein-Barr virus nuclear antigen 2. J Virol. 1999;73(5):4481–4. 10196351

16. Zhao B, Maruo S, Cooper A, R Chase M, Johannsen E, Kieff E, et al. RNAs induced by Epstein-Barr virus nuclear antigen 2 in lymphoblastoid cell lines. Proc Natl Acad Sci U S A. 2006;103(6):1900–5. doi: 10.1073/pnas.0510612103 16446431

17. Zhao B, Zou J, Wang H, Johannsen E, Peng CW, Quackenbush J, et al. Epstein-Barr virus exploits intrinsic B-lymphocyte transcription programs to achieve immortal cell growth. Proc Natl Acad Sci U S A. 2011;108(36):14902–7. doi: 10.1073/pnas.1108892108 21746931

18. Stine ZE, Walton ZE, Altman BJ, Hsieh AL, Dang CV. MYC, Metabolism, and Cancer. Cancer Discov. 2015;5(10):1024–39. doi: 10.1158/2159-8290.CD-15-0507 26382145

19. Marshall D, Sample C. Epstein-Barr virus nuclear antigen 3C is a transcriptional regulator. J Virol. 1995;69(6):3624–30. 7745710

20. Robertson ES, Grossman S, Johannsen E, Miller C, Lin J, Tomkinson B, et al. Epstein-Barr virus nuclear protein 3C modulates transcription through interaction with the sequence-specific DNA-binding protein J kappa. J Virol. 1995;69(5):3108–16. 7707539

21. Wang A, Welch R, Zhao B, Ta T, Keles S, Johannsen E. Epstein-Barr Virus Nuclear Antigen 3 (EBNA3) Proteins Regulate EBNA2 Binding to Distinct RBPJ Genomic Sites. J Virol. 2015;90(6):2906–19. doi: 10.1128/JVI.02737-15 26719268

22. Schmidt SC, Jiang S, Zhou H, Willox B, Holthaus AM, Kharchenko PV, et al. Epstein-Barr virus nuclear antigen 3A partially coincides with EBNA3C genome-wide and is tethered to DNA through BATF complexes. Proc Natl Acad Sci U S A. 2015;112(2):554–9. doi: 10.1073/pnas.1422580112 25540416

23. Jiang S, Zhou H, Liang J, Gerdt C, Wang C, Ke L, et al. The Epstein-Barr Virus Regulome in Lymphoblastoid Cells. Cell Host Microbe. 2017;22(4):561–73.e4. doi: 10.1016/j.chom.2017.09.001 29024646

24. Jiang S, Willox B, Zhou H, Holthaus AM, Wang A, Shi TT, et al. Epstein-Barr virus nuclear antigen 3C binds to BATF/IRF4 or SPI1/IRF4 composite sites and recruits Sin3A to repress CDKN2A. Proc Natl Acad Sci U S A. 2014;111(1):421–6. doi: 10.1073/pnas.1321704111 24344258

25. McFadden K, Hafez AY, Kishton R, Messinger JE, Nikitin PA, Rathmell JC, et al. Metabolic stress is a barrier to Epstein-Barr virus-mediated B-cell immortalization. Proc Natl Acad Sci U S A. 2016;113(6):E782–90. doi: 10.1073/pnas.1517141113 26802124

26. Wang LW, Shen H, Nobre L, Ersing I, Paulo JA, Trudeau S, et al. Epstein-Barr-Virus-Induced One-Carbon Metabolism Drives B Cell Transformation. Cell Metab. 2019.

27. Sabatini DM. Twenty-five years of mTOR: Uncovering the link from nutrients to growth. Proc Natl Acad Sci U S A. 2017;114(45):11818–25. doi: 10.1073/pnas.1716173114 29078414

28. Wang L, Wei, Shen H, Nobre L, Ersing I, Paulo JA, et al. Epstein-Barr Virus Induced One-Carbon Metabolism Drives B-Cell Transformation. Cell Metabolism 2019.

29. Price AM, Tourigny JP, Forte E, Salinas RE, Dave SS, Luftig MA. Analysis of Epstein-Barr virus-regulated host gene expression changes through primary B-cell outgrowth reveals delayed kinetics of latent membrane protein 1-mediated NF-κB activation. J Virol. 2012;86(20):11096–106. doi: 10.1128/JVI.01069-12 22855490

30. Price AM, Messinger JE, Luftig MA. c-Myc Represses Transcription of Epstein-Barr Virus Latent Membrane Protein 1 Early after Primary B Cell Infection. J Virol. 2018;92(2).

31. Wang LW, Jiang S, Gewurz BE. Epstein-Barr Virus LMP1-Mediated Oncogenicity. J Virol. 2017;91(21).

32. Kieser A, Sterz KR. The Latent Membrane Protein 1 (LMP1). Curr Top Microbiol Immunol. 2015;391:119–49. doi: 10.1007/978-3-319-22834-1_4 26428373

33. Cen O, Longnecker R. Latent Membrane Protein 2 (LMP2). Curr Top Microbiol Immunol. 2015;391:151–80. doi: 10.1007/978-3-319-22834-1_5 26428374

34. Mancao C, Hammerschmidt W. Epstein-Barr virus latent membrane protein 2A is a B-cell receptor mimic and essential for B-cell survival. Blood. 2007;110(10):3715–21. doi: 10.1182/blood-2007-05-090142 17682125

35. Mrozek-Gorska P, Buschle A, Pich D, Schwarzmayr T, Fechtner R, Scialdone A, et al. Epstein-Barr virus reprograms human B lymphocytes immediately in the prelatent phase of infection. Proc Natl Acad Sci U S A. 2019.

36. Birsoy K, Wang T, Chen WW, Freinkman E, Abu-Remaileh M, Sabatini DM. An Essential Role of the Mitochondrial Electron Transport Chain in Cell Proliferation Is to Enable Aspartate Synthesis. Cell. 2015;162(3):540–51. doi: 10.1016/j.cell.2015.07.016 26232224

37. Goldstein JL, Brown MS. Regulation of the mevalonate pathway. Nature. 1990;343(6257):425–30. doi: 10.1038/343425a0 1967820

38. Valente AJ, Morris CJ, Walton KW. Demonstration by a rosette-forming technique of surface receptors for low-density lipoproteins on lymphoblastoid cells. Clin Sci (Lond). 1980;59(4):265–73.

39. Crumpton MJ, Owens RJ, Gallagher CJ, Davies AA. The cell surface and its metabolism. J Pathol. 1983;141(3):235–48. doi: 10.1002/path.1711410305 6363647

40. Wheeler HE, Dolan ME. Lymphoblastoid cell lines in pharmacogenomic discovery and clinical translation. Pharmacogenomics. 2012;13(1):55–70. doi: 10.2217/pgs.11.121 22176622

41. Carta G, Murru E, Banni S, Manca C. Palmitic Acid: Physiological Role, Metabolism and Nutritional Implications. Front Physiol. 2017;8:902. doi: 10.3389/fphys.2017.00902 29167646

42. Foretz M, Guichard C, Ferre P, Foufelle F. Sterol regulatory element binding protein-1c is a major mediator of insulin action on the hepatic expression of glucokinase and lipogenesis-related genes. Proc Natl Acad Sci U S A. 1999;96(22):12737–42. doi: 10.1073/pnas.96.22.12737 10535992

43. Amemiya-Kudo M, Shimano H, Hasty AH, Yahagi N, Yoshikawa T, Matsuzaka T, et al. Transcriptional activities of nuclear SREBP-1a, -1c, and -2 to different target promoters of lipogenic and cholesterogenic genes. J Lipid Res. 2002;43(8):1220–35. 12177166

44. Shimano H, Yahagi N, Amemiya-Kudo M, Hasty AH, Osuga J, Tamura Y, et al. Sterol regulatory element-binding protein-1 as a key transcription factor for nutritional induction of lipogenic enzyme genes. J Biol Chem. 1999;274(50):35832–9. doi: 10.1074/jbc.274.50.35832 10585467

45. Zeller KI, Jegga AG, Aronow BJ, O'Donnell KA, Dang CV. An integrated database of genes responsive to the Myc oncogenic transcription factor: identification of direct genomic targets. Genome Biol. 2003;4(10):R69. doi: 10.1186/gb-2003-4-10-r69 14519204

46. Lin CY, Lovén J, Rahl PB, Paranal RM, Burge CB, Bradner JE, et al. Transcriptional amplification in tumor cells with elevated c-Myc. Cell. 2012;151(1):56–67. doi: 10.1016/j.cell.2012.08.026 23021215

47. Dang CV. MYC, metabolism, cell growth, and tumorigenesis. Cold Spring Harb Perspect Med. 2013;3(8).

48. Davis CA, Hitz BC, Sloan CA, Chan ET, Davidson JM, Gabdank I, et al. The Encyclopedia of DNA elements (ENCODE): data portal update. Nucleic Acids Res. 2018;46(D1):D794–d801. doi: 10.1093/nar/gkx1081 29126249

49. Consortium EP. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489(7414):57–74. doi: 10.1038/nature11247 22955616

50. Engelking LJ, Kuriyama H, Hammer RE, Horton JD, Brown MS, Goldstein JL, et al. Overexpression of Insig-1 in the livers of transgenic mice inhibits SREBP processing and reduces insulin-stimulated lipogenesis. J Clin Invest. 2004;113(8):1168–75. doi: 10.1172/JCI20978 15085196

51. Yang T, Espenshade PJ, Wright ME, Yabe D, Gong Y, Aebersold R, et al. Crucial step in cholesterol homeostasis: sterols promote binding of SCAP to INSIG-1, a membrane protein that facilitates retention of SREBPs in ER. Cell. 2002;110(4):489–500. doi: 10.1016/s0092-8674(02)00872-3 12202038

52. Kamisuki S, Mao Q, Abu-Elheiga L, Gu Z, Kugimiya A, Kwon Y, et al. A small molecule that blocks fat synthesis by inhibiting the activation of SREBP. Chem Biol. 2009;16(8):882–92. doi: 10.1016/j.chembiol.2009.07.007 19716478

53. Arvey A, Tempera I, Tsai K, Chen HS, Tikhmyanova N, Klichinsky M, et al. An atlas of the Epstein-Barr virus transcriptome and epigenome reveals host-virus regulatory interactions. Cell Host Microbe. 2012;12(2):233–45. doi: 10.1016/j.chom.2012.06.008 22901543

54. Rowe D, Heston L, Metlay J, Miller G. Identification and expression of a nuclear antigen from the genomic region of the Jijoye strain of Epstein-Barr virus that is missing in its nonimmortalizing deletion mutant, P3HR-1. Proc Natl Acad Sci U S A. 1985;82(21):7429–33. doi: 10.1073/pnas.82.21.7429 2997790

55. Rymo L, Klein G, Ricksten A. Expression of a second Epstein-Barr virus-determined nuclear antigen in mouse cells after gene transfer with a cloned fragment of the viral genome. Proc Natl Acad Sci U S A. 1985;82(10):3435–9. doi: 10.1073/pnas.82.10.3435 2987926

56. Tsang SF, Wang F, Izumi KM, Kieff E. Delineation of the cis-acting element mediating EBNA-2 transactivation of latent infection membrane protein expression. J Virol. 1991;65(12):6765–71. 1658373

57. Wang F, Tsang SF, Kurilla MG, Cohen JI, Kieff E. Epstein-Barr virus nuclear antigen 2 transactivates latent membrane protein LMP1. J Virol. 1990;64(7):3407–16. 2352328

58. Miller G, Robinson J, Heston L, Lipman M. Differences between laboratory strains of Epstein-Barr virus based on immortalization, abortive infection, and interference. Proc Natl Acad Sci U S A. 1974;71(10):4006–10. doi: 10.1073/pnas.71.10.4006 4372601

59. Miller G, Robinson J, Heston L. Immortalizing and nonimmortalizing laboratory strains of Epstein-Barr Virus. Cold Spring Harb Symp Quant Biol. 1975;39 Pt 2:773–81.

60. Schuhmacher M, Staege MS, Pajic A, Polack A, Weidle UH, Bornkamm GW, et al. Control of cell growth by c-Myc in the absence of cell division. Curr Biol. 1999;9(21):1255–8. doi: 10.1016/s0960-9822(99)80507-7 10556095

61. Schuhmacher M, Kohlhuber F, Hölzel M, Kaiser C, Burtscher H, Jarsch M, et al. The transcriptional program of a human B cell line in response to Myc. Nucleic Acids Res. 2001;29(2):397–406. doi: 10.1093/nar/29.2.397 11139609

62. Katano H, Pesnicak L, Cohen JI. Simvastatin induces apoptosis of Epstein-Barr virus (EBV)-transformed lymphoblastoid cell lines and delays development of EBV lymphomas. Proc Natl Acad Sci U S A. 2004;101(14):4960–5. doi: 10.1073/pnas.0305149101 15041742

63. Wagner BK, Gilbert TJ, Hanai J, Imamura S, Bodycombe NE, Bon RS, et al. A small-molecule screening strategy to identify suppressors of statin myopathy. ACS Chem Biol. 2011;6(9):900–4. doi: 10.1021/cb200206w 21732624

64. Goldstein JL, Brown MS. A century of cholesterol and coronaries: from plaques to genes to statins. Cell. 2015;161(1):161–72. doi: 10.1016/j.cell.2015.01.036 25815993

65. Berndt N, Hamilton AD, Sebti SM. Targeting protein prenylation for cancer therapy. Nat Rev Cancer. 2011;11(11):775–91. doi: 10.1038/nrc3151 22020205

66. Casey PJ, Seabra MC. Protein prenyltransferases. J Biol Chem. 1996;271(10):5289–92. doi: 10.1074/jbc.271.10.5289 8621375

67. Hutagalung AH, Novick PJ. Role of Rab GTPases in membrane traffic and cell physiology. Physiol Rev. 2011;91(1):119–49. doi: 10.1152/physrev.00059.2009 21248164

68. Lackner MR, Kindt RM, Carroll PM, Brown K, Cancilla MR, Chen C, et al. Chemical genetics identifies Rab geranylgeranyl transferase as an apoptotic target of farnesyl transferase inhibitors. Cancer Cell. 2005;7(4):325–36. doi: 10.1016/j.ccr.2005.03.024 15837622

69. Nguyen UT, Guo Z, Delon C, Wu Y, Deraeve C, Franzel B, et al. Analysis of the eukaryotic prenylome by isoprenoid affinity tagging. Nat Chem Biol. 2009;5(4):227–35. doi: 10.1038/nchembio.149 19219049

70. McGuire TF, Sebti SM. Geranylgeraniol potentiates lovastatin inhibition of oncogenic H-Ras processing and signaling while preventing cytotoxicity. Oncogene. 1997;14(3):305–12. doi: 10.1038/sj.onc.1200819 9018116

71. Ma Y, Walsh MJ, Bernhardt K, Ashbaugh CW, Trudeau SJ, Ashbaugh IY, et al. CRISPR/Cas9 Screens Reveal Epstein-Barr Virus-Transformed B Cell Host Dependency Factors. Cell Host Microbe. 2017;21(5):580–91 e7. doi: 10.1016/j.chom.2017.04.005 28494239

72. Maruo S, Wu Y, Ishikawa S, Kanda T, Iwakiri D, Takada K. 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. 2006;103(51):19500–5. doi: 10.1073/pnas.0604919104 17159137

73. Zhao B, Mar JC, Maruo S, Lee S, Gewurz BE, Johannsen E, et al. Epstein-Barr virus nuclear antigen 3C regulated genes in lymphoblastoid cell lines. Proc Natl Acad Sci U S A. 2011;108(1):337–42. doi: 10.1073/pnas.1017419108 21173222

74. Longnecker R, Kieff E. A second Epstein-Barr virus membrane protein (LMP2) is expressed in latent infection and colocalizes with LMP1. J Virol. 1990;64(5):2319–26. 2157888

75. Meckes DG, Menaker NF, Raab-Traub N. Epstein-Barr virus LMP1 modulates lipid raft microdomains and the vimentin cytoskeleton for signal transduction and transformation. J Virol. 2013;87(3):1301–11. doi: 10.1128/JVI.02519-12 23152522

76. Higuchi M, Izumi KM, Kieff E. Epstein-Barr virus latent-infection membrane proteins are palmitoylated and raft-associated: protein 1 binds to the cytoskeleton through TNF receptor cytoplasmic factors. Proc Natl Acad Sci U S A. 2001;98(8):4675–80. doi: 10.1073/pnas.081075298 11296297

77. Lam N, Sugden B. LMP1, a viral relative of the TNF receptor family, signals principally from intracellular compartments. Embo j. 2003;22(12):3027–38. doi: 10.1093/emboj/cdg284 12805217

78. Lee J, Sugden B. A membrane leucine heptad contributes to trafficking, signaling, and transformation by latent membrane protein 1. J Virol. 2007;81(17):9121–30. doi: 10.1128/JVI.00136-07 17581993

79. Lange PT, Lagunoff M, Tarakanova VL. Chewing the Fat: The Conserved Ability of DNA Viruses to Hijack Cellular Lipid Metabolism. Viruses. 2019;11(2).

80. Sychev ZE, Hu A, DiMaio TA, Gitter A, Camp ND, Noble WS, et al. Integrated systems biology analysis of KSHV latent infection reveals viral induction and reliance on peroxisome mediated lipid metabolism. PLoS Pathog. 2017;13(3):e1006256. doi: 10.1371/journal.ppat.1006256 28257516

81. Rodriguez-Sanchez I, Munger J. Meal for Two: Human Cytomegalovirus-Induced Activation of Cellular Metabolism. Viruses. 2019;11(3).

82. Shenk T, Alwine JC. Human Cytomegalovirus: Coordinating Cellular Stress, Signaling, and Metabolic Pathways. Annu Rev Virol. 2014;1(1):355–74. doi: 10.1146/annurev-virology-031413-085425 26958726

83. Purdy JG, Shenk T, Rabinowitz JD. Fatty acid elongase 7 catalyzes lipidome remodeling essential for human cytomegalovirus replication. Cell Rep. 2015;10(8):1375–85. doi: 10.1016/j.celrep.2015.02.003 25732827

84. Jean Beltran PM, Cook KC, Hashimoto Y, Galitzine C, Murray LA, Vitek O, et al. Infection-Induced Peroxisome Biogenesis Is a Metabolic Strategy for Herpesvirus Replication. Cell Host Microbe. 2018;24(4):526–41.e7. doi: 10.1016/j.chom.2018.09.002 30269970

85. Sanchez EL, Lagunoff M. Viral activation of cellular metabolism. Virology. 2015;479–480:609–18. doi: 10.1016/j.virol.2015.02.038 25812764

86. Greseth MD, Traktman P. De novo fatty acid biosynthesis contributes significantly to establishment of a bioenergetically favorable environment for vaccinia virus infection. PLoS Pathog. 2014;10(3):e1004021. doi: 10.1371/journal.ppat.1004021 24651651

87. Singh RK, Lang F, Pei Y, Jha HC, Robertson ES. Metabolic reprogramming of Kaposi's sarcoma associated herpes virus infected B-cells in hypoxia. PLoS Pathog. 2018;14(5):e1007062. doi: 10.1371/journal.ppat.1007062 29746587

88. Zhu Y, Li T, Ramos da Silva S, Lee JJ, Lu C, Eoh H, et al. A Critical Role of Glutamine and Asparagine gamma-Nitrogen in Nucleotide Biosynthesis in Cancer Cells Hijacked by an Oncogenic Virus. MBio. 2017;8(4).

89. Zhu Y, Ramos da Silva S, He M, Liang Q, Lu C, Feng P, et al. An Oncogenic Virus Promotes Cell Survival and Cellular Transformation by Suppressing Glycolysis. PLoS Pathog. 2016;12(5):e1005648. doi: 10.1371/journal.ppat.1005648 27187079

90. Li X, Wu JB, Li Q, Shigemura K, Chung LW, Huang WC. SREBP-2 promotes stem cell-like properties and metastasis by transcriptional activation of c-Myc in prostate cancer. Oncotarget. 2016;7(11):12869–84. doi: 10.18632/oncotarget.7331 26883200

91. Wu Y, Chen K, Liu X, Huang L, Zhao D, Li L, et al. Srebp-1 Interacts with c-Myc to Enhance Somatic Cell Reprogramming. Stem Cells. 2016;34(1):83–92. doi: 10.1002/stem.2209 26388522

92. Lo AK, Lung RW, Dawson CW, Young LS, Ko C, Yeung WW, et al. Activation of sterol regulatory element‐binding protein 1 (SREBP1)‐mediated lipogenesis by the Epstein–Barr virus‐encoded latent membrane protein 1 (LMP1) promotes cell proliferation and progression of nasopharyngeal carcinoma. J Pathol. 2462018. p. 180–90.

93. Moody CA, Scott RS, Amirghahari N, Nathan CO, Young LS, Dawson CW, et al. Modulation of the cell growth regulator mTOR by Epstein-Barr virus-encoded LMP2A. J Virol. 2005;79(9):5499–506. doi: 10.1128/JVI.79.9.5499-5506.2005 15827164

94. Ikeda M, Longnecker R. Cholesterol is critical for Epstein-Barr virus latent membrane protein 2A trafficking and protein stability. Virology. 2007;360(2):461–8. doi: 10.1016/j.virol.2006.10.046 17150237

95. Verweij FJ, van Eijndhoven MA, Hopmans ES, Vendrig T, Wurdinger T, Cahir-McFarland E, et al. LMP1 association with CD63 in endosomes and secretion via exosomes limits constitutive NF-κB activation. EMBO J. 2011;30(11):2115–29. doi: 10.1038/emboj.2011.123 21527913

96. Shimabukuro-Vornhagen A, Zoghi S, Liebig TM, Wennhold K, Chemitz J, Draube A, et al. Inhibition of protein geranylgeranylation specifically interferes with CD40-dependent B cell activation, resulting in a reduced capacity to induce T cell immunity. J Immunol. 2014;193(10):5294–305. doi: 10.4049/jimmunol.1203436 25311809

97. Ageberg M, Rydström K, Lindén O, Linderoth J, Jerkeman M, Drott K. Inhibition of geranylgeranylation mediates sensitivity to CHOP-induced cell death of DLBCL cell lines. Exp Cell Res. 2011;317(8):1179–91. doi: 10.1016/j.yexcr.2011.02.006 21324313

98. van de Donk NW, Schotte D, Kamphuis MM, van Marion AM, van Kessel B, Bloem AC, et al. Protein geranylgeranylation is critical for the regulation of survival and proliferation of lymphoma tumor cells. Clin Cancer Res. 2003;9(15):5735–48. 14654559

99. van de Donk NW, Lokhorst HM, Nijhuis EH, Kamphuis MM, Bloem AC. Geranylgeranylated proteins are involved in the regulation of myeloma cell growth. Clin Cancer Res. 2005;11(2 Pt 1):429–39.

100. Fuchs D, Berges C, Opelz G, Daniel V, Naujokat C. HMG-CoA reductase inhibitor simvastatin overcomes bortezomib-induced apoptosis resistance by disrupting a geranylgeranyl pyrophosphate-dependent survival pathway. Biochem Biophys Res Commun. 2008;374(2):309–14. doi: 10.1016/j.bbrc.2008.07.012 18625202

101. Deng B, Zhu X, Zhao Y, Zhang D, Pannu A, Chen L, et al. PKC and Rab13 mediate Ca. Biochem Biophys Res Commun. 2018;495(2):1956–63. doi: 10.1016/j.bbrc.2017.12.064 29247648

102. Yeo JC, Wall AA, Luo L, Stow JL. Sequential recruitment of Rab GTPases during early stages of phagocytosis. Cell Logist. 2016;6(1):e1140615. doi: 10.1080/21592799.2016.1140615 27217977

103. Condon ND, Heddleston JM, Chew TL, Luo L, McPherson PS, Ioannou MS, et al. Macropinosome formation by tent pole ruffling in macrophages. J Cell Biol. 2018;217(11):3873–85. doi: 10.1083/jcb.201804137 30150290

104. Zhang L, Dai F, Cui L, Zhou B, Guo Y. Up-regulation of the active form of small GTPase Rab13 promotes macroautophagy in vascular endothelial cells. Biochim Biophys Acta Mol Cell Res. 2017;1864(4):613–24. doi: 10.1016/j.bbamcr.2017.01.003 28087344

105. Nishikimi A, Ishihara S, Ozawa M, Etoh K, Fukuda M, Kinashi T, et al. Rab13 acts downstream of the kinase Mst1 to deliver the integrin LFA-1 to the cell surface for lymphocyte trafficking. Sci Signal. 2014;7(336):ra72. doi: 10.1126/scisignal.2005199 25074980

106. Ioannou MS, Bell ES, Girard M, Chaineau M, Hamlin JN, Daubaras M, et al. DENND2B activates Rab13 at the leading edge of migrating cells and promotes metastatic behavior. J Cell Biol. 2015;208(5):629–48. doi: 10.1083/jcb.201407068 25713415

107. Köhler K, Louvard D, Zahraoui A. Rab13 regulates PKA signaling during tight junction assembly. J Cell Biol. 2004;165(2):175–80. doi: 10.1083/jcb.200312118 15096524

108. Ioannou MS, McPherson PS. Regulation of Cancer Cell Behavior by the Small GTPase Rab13. J Biol Chem. 2016;291(19):9929–37. doi: 10.1074/jbc.R116.715193 27044746

109. Hurwitz SN, Nkosi D, Conlon MM, York SB, Liu X, Tremblay DC, et al. CD63 Regulates Epstein-Barr Virus LMP1 Exosomal Packaging, Enhancement of Vesicle Production, and Noncanonical NF-κB Signaling. J Virol. 2017;91(5).

110. Liu HP, Wu CC, Chang YS. PRA1 promotes the intracellular trafficking and NF-kappaB signaling of EBV latent membrane protein 1. EMBO J. 2006;25(17):4120–30. doi: 10.1038/sj.emboj.7601282 16917502

111. Verweij FJ, de Heus C, Kroeze S, Cai H, Kieff E, Piersma SR, et al. Exosomal sorting of the viral oncoprotein LMP1 is restrained by TRAF2 association at signalling endosomes. J Extracell Vesicles. 2015;4:26334. doi: 10.3402/jev.v4.26334 25865256

112. Nkosi D, Howell LA, Cheerathodi MR, Hurwitz SN, Tremblay DC, Liu X, et al. Transmembrane Domains Mediate Intra- and Extracellular Trafficking of Epstein-Barr Virus Latent Membrane Protein 1. J Virol. 2018;92(17).

113. Ioannou MS, Girard M, McPherson PS. Rab13 Traffics on Vesicles Independent of Prenylation. J Biol Chem. 2016;291(20):10726–35. doi: 10.1074/jbc.M116.722298 26969162

114. Lynch DT, Zimmerman JS, Rowe DT. Epstein-Barr virus latent membrane protein 2B (LMP2B) co-localizes with LMP2A in perinuclear regions in transiently transfected cells. J Gen Virol. 2002;83(Pt 5):1025–35. doi: 10.1099/0022-1317-83-5-1025 11961256

115. Dawson CW, George JH, Blake SM, Longnecker R, Young LS. The Epstein-Barr virus encoded latent membrane protein 2A augments signaling from latent membrane protein 1. Virology. 2001;289(2):192–207. doi: 10.1006/viro.2001.1142 11689042

116. Morimoto S, Nishimura N, Terai T, Manabe S, Yamamoto Y, Shinahara W, et al. Rab13 mediates the continuous endocytic recycling of occludin to the cell surface. J Biol Chem. 2005;280(3):2220–8. doi: 10.1074/jbc.M406906200 15528189

117. Martincic I, Peralta ME, Ngsee JK. Isolation and characterization of a dual prenylated Rab and VAMP2 receptor. J Biol Chem. 1997;272(43):26991–8. doi: 10.1074/jbc.272.43.26991 9341137

118. Hutt DM, Da-Silva LF, Chang LH, Prosser DC, Ngsee JK. PRA1 inhibits the extraction of membrane-bound rab GTPase by GDI1. J Biol Chem. 2000;275(24):18511–9. doi: 10.1074/jbc.M909309199 10751420

119. Sivars U, Aivazian D, Pfeffer SR. Yip3 catalyses the dissociation of endosomal Rab-GDI complexes. Nature. 2003;425(6960):856–9. doi: 10.1038/nature02057 14574414

120. Stunz LL, Bishop GA. Latent membrane protein 1 and the B lymphocyte-a complex relationship. Crit Rev Immunol. 2014;34(3):177–98. 24941072

121. Küppers R, Klein U, Schwering I, Distler V, Bräuninger A, Cattoretti G, et al. Identification of Hodgkin and Reed-Sternberg cell-specific genes by gene expression profiling. J Clin Invest. 1112003. p. 529–37.

122. DeKroon RM, Gunawardena HP, Edwards R, Raab-Traub N. Global Proteomic Changes Induced by the Epstein-Barr Virus Oncoproteins Latent Membrane Protein 1 and 2A. MBio. 2018;9(3).

123. Thibault A, Samid D, Tompkins AC, Figg WD, Cooper MR, Hohl RJ, et al. Phase I study of lovastatin, an inhibitor of the mevalonate pathway, in patients with cancer. Clin Cancer Res. 1996;2(3):483–91. 9816194

124. Munz C. Humanized mouse models for Epstein Barr virus infection. Curr Opin Virol. 2017;25:113–8. doi: 10.1016/j.coviro.2017.07.026 28837889

125. Calderwood MA, Holthaus AM, Johannsen E. The Epstein-Barr virus LF2 protein inhibits viral replication. J Virol. 2008;82(17):8509–19. doi: 10.1128/JVI.00315-08 18562535

126. Johannsen E, Luftig M, Chase MR, Weicksel S, Cahir-McFarland E, Illanes D, et al. Proteins of purified Epstein-Barr virus. Proc Natl Acad Sci U S A. 2004;101(46):16286–91. doi: 10.1073/pnas.0407320101 15534216

127. Ersing I, Nobre L, Wang LW, Soday L, Ma Y, Paulo JA, et al. A Temporal Proteomic Map of Epstein-Barr Virus Lytic Replication in B Cells. Cell Rep. 2017;19(7):1479–93. doi: 10.1016/j.celrep.2017.04.062 28514666

128. Jiang S, Wang LW, Walsh MJ, Trudeau SJ, Gerdt C, Zhao B, et al. CRISPR/Cas9-Mediated Genome Editing in Epstein-Barr Virus-Transformed Lymphoblastoid B-Cell Lines. Curr Protoc Mol Biol. 2018;121:31.12.1–31.12.23.

129. Joberty G, Tavitian A, Zahraoui A. Isoprenylation of Rab proteins possessing a C-terminal CaaX motif. FEBS letters. 1993;330(3):323–8. doi: 10.1016/0014-5793(93)80897-4 8375503

130. Huang da W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009;4(1):44–57. doi: 10.1038/nprot.2008.211 19131956

131. Portal D, Zhou H, Zhao B, Kharchenko PV, Lowry E, Wong L, et al. Epstein-Barr virus nuclear antigen leader protein localizes to promoters and enhancers with cell transcription factors and EBNA2. Proc Natl Acad Sci U S A. 2013;110(46):18537–42. doi: 10.1073/pnas.1317608110 24167291

132. Ohashi M, Holthaus AM, Calderwood MA, Lai CY, Krastins B, Sarracino D, et al. The EBNA3 family of Epstein-Barr virus nuclear proteins associates with the USP46/USP12 deubiquitination complexes to regulate lymphoblastoid cell line growth. PLoS Pathog. 2015;11(4):e1004822. doi: 10.1371/journal.ppat.1004822 25855980

133. Langmead B, Trapnell C, Pop M, Salzberg SL. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 2009;10(3):R25. doi: 10.1186/gb-2009-10-3-r25 19261174

134. Zhou X, Lowdon RF, Li D, Lawson HA, Madden PA, Costello JF, et al. Exploring long-range genome interactions using the WashU Epigenome Browser. Nature methods. 2013;10(5):375–6. doi: 10.1038/nmeth.2440 23629413

135. Gordon J, Walker L, Guy G, Brown G, Rowe M, Rickinson A. Control of human B-lymphocyte replication. II. Transforming Epstein-Barr virus exploits three distinct viral signals to undermine three separate control points in B-cell growth. Immunology. 1986;58(4):591–5. 2426189

136. Wang F, Gregory CD, Rowe M, Rickinson AB, Wang D, Birkenbach M, et al. Epstein-Barr virus nuclear antigen 2 specifically induces expression of the B-cell activation antigen CD23. Proc Natl Acad Sci U S A. 1987;84(10):3452–6. doi: 10.1073/pnas.84.10.3452 3033649

137. Thorley-Lawson DA, Nadler LM, Bhan AK, Schooley RT. BLAST-2 [EBVCS], an early cell surface marker of human B cell activation, is superinduced by Epstein Barr virus. J Immunol. 1985;134(5):3007–12. 2984280

Štítky
Hygiena a epidemiologie Infekční lékařství Laboratoř

Článek vyšel v časopise

PLOS Pathogens


2019 Číslo 9
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#