#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

A Novel Mechanism Inducing Genome Instability in Kaposi's Sarcoma-Associated Herpesvirus Infected Cells


The hallmarks of cancer comprise the essential elements that permit the formation and development of human tumours. Genome instability is an enabling characteristic that allows the progression of tumorigenesis through genetic mutation and therefore, understanding the molecular causes of genome instability in all cancers is essential for development of therapeutics. The Kaposi's sarcoma-associated herpesvirus (KSHV) is an important human pathogen that causes multiple AIDS-related cancers. Recent studies have shown that during KSHV infection, cells show an increase in a double-strand DNA break marker, signifying a severe form of genome instability. Herein, we show that KSHV infection does cause DNA strand breaks. Moreover, we describe a novel molecular mechanism for genome instability involving the KSHV ORF57 protein interacting with the mRNA export complex, hTREX. We demonstrate that over-expression of ORF57 results in the formation of RNA:DNA hybrids, or R-loops, that lead to an increase in genome instability. DNA strand breaks have been previously reported in herpes simplex, cytomegalovirus and Epstein-Barr virus infected cells. Therefore, as this work describes for the first time the mechanism of R-loop induced genome instability involving a conserved herpesvirus protein, it may have far-reaching implications for other viral RNA export factors.


Vyšlo v časopise: A Novel Mechanism Inducing Genome Instability in Kaposi's Sarcoma-Associated Herpesvirus Infected Cells. PLoS Pathog 10(5): e32767. doi:10.1371/journal.ppat.1004098
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004098

Souhrn

The hallmarks of cancer comprise the essential elements that permit the formation and development of human tumours. Genome instability is an enabling characteristic that allows the progression of tumorigenesis through genetic mutation and therefore, understanding the molecular causes of genome instability in all cancers is essential for development of therapeutics. The Kaposi's sarcoma-associated herpesvirus (KSHV) is an important human pathogen that causes multiple AIDS-related cancers. Recent studies have shown that during KSHV infection, cells show an increase in a double-strand DNA break marker, signifying a severe form of genome instability. Herein, we show that KSHV infection does cause DNA strand breaks. Moreover, we describe a novel molecular mechanism for genome instability involving the KSHV ORF57 protein interacting with the mRNA export complex, hTREX. We demonstrate that over-expression of ORF57 results in the formation of RNA:DNA hybrids, or R-loops, that lead to an increase in genome instability. DNA strand breaks have been previously reported in herpes simplex, cytomegalovirus and Epstein-Barr virus infected cells. Therefore, as this work describes for the first time the mechanism of R-loop induced genome instability involving a conserved herpesvirus protein, it may have far-reaching implications for other viral RNA export factors.


Zdroje

1. HanahanD, WeinbergRA (2011) Hallmarks of Cancer: The Next Generation. Cell 144: 646–674.

2. HanahanD, WeinbergRA (2000) The Hallmarks of Cancer. Cell 100: 57–70.

3. FriedbergEC (2003) DNA damage and repair. Nature 421: 436–440.

4. FleckO, NielsenO (2004) DNA repair. J Cell Sci 117: 515–517.

5. IshinoY, NishinoT, MorikawaK (2006) Mechanisms of Maintaining Genetic Stability by Homologous Recombination. Chem Rev 106: 324–339.

6. JacksonSP (2002) Sensing and repairing DNA double-strand breaks. Carcinogenesis 23: 687–696.

7. CrastaK, GanemNJ, DagherR, LantermannAB, IvanovaEV, et al. (2012) DNA breaks and chromosome pulverization from errors in mitosis. Nature 482: 53–58.

8. StephensPJ, GreenmanCD, FuB, YangF, BignellGR, et al. (2011) Massive Genomic Rearrangement Acquired in a Single Catastrophic Event during Cancer Development. Cell 144: 27–40.

9. FormentJV, KaidiA, JacksonSP (2012) Chromothripsis and cancer: causes and consequences of chromosome shattering. Nat Rev Cancer 12: 663–670.

10. ThompsonLH, SchildD (2002) Recombinational DNA repair and human disease. Mutat Res 509: 49–78.

11. ElliottB, JasinM (2002) Human Genome and Diseases: Double-strand breaks and translocations in cancer. Cell Mol Life Sci 59: 373–385.

12. ThompsonS, ComptonD (2011) Chromosomes and cancer cells. Chromosome Res 19: 433–444.

13. GanemD (2010) KSHV and the pathogenesis of Kaposi sarcoma: listening to human biology and medicine. J Clin Invest 120: 939–949.

14. ChangY, CesarmanE, PessinMS, LeeF, CulpepperJ, et al. (1994) Identification of herpesvirus-like DNA sequences in AIDS-associated Kaposi's sarcoma. Science 266: 1865–1869.

15. YeF, LeiX, GaoSJ (2011) Mechanisms of Kaposi's Sarcoma-Associated Herpesvirus Latency and Reactivation. Adv Virol 2011 Article ID 193860.

16. GanemD (2006) KSHV infection and the pathogenesis of Kaposi's sarcoma. Ann Rev Pathol Mech Dis 1: 273.

17. XiaoY, ChenJ, LiaoQ, WuY, PengC, et al. (2013) Lytic infection of Kaposi's Sarcoma-Associated Herpesvirus induces DNA double-strand breaks and impairs NHEJ. J Gen Virol 94: 1870–1875.

18. PanH, ZhouF, GaoS-J (2004) Kaposi's Sarcoma-Associated Herpesvirus Induction of Chromosome Instability in Primary Human Endothelial Cells. Cancer Res 64: 4064–4068.

19. PyakurelP, PakF, MwakigonjaA, KaayaE, BiberfeldP (2007) KSHV/HHV-8 and HIV infection in Kaposi's sarcoma development. Infect Agent Cancer 2: 4.

20. PopescuN, ZimonjicD, Leventon-KrissS, BryantJ, Lunardi-IskandarY, et al. (1996) Deletion and translocation involving chromosome 3 (p14) in two tumorigenic Kaposi's sarcoma cell lines. J Natl Cancer Inst 88: 450–455.

21. CasaloneR, AlbiniA, RighiR, GranataP, TonioloA (2001) Nonrandom chromosome changes in Kaposi sarcoma: cytogenetic and FISH results in a new cell line (KS-IMM) and literature review. Cancer Genet Cytogenet 124: 16–19.

22. PyakurelP, MontagU, Castanos-VelezE, KaayaE, ChristenssonB, et al. (2006) CGH of microdissected Kaposi's sarcoma lesions reveals recurrent loss of chromosome Y in early and additional chromosomal changes in late tumor stages. AIDS 20: 1805–1812.

23. Kiuru-KuhlefeltS, Sarlomo-RikalaM, LarramendyM, SoderlundM, HedmanK, et al. (2000) FGF4 and INT2 oncogenes are amplified and expressed in Kaposi's sarcoma. Mod Pathol 13: 433–437.

24. NairP, PanH, StallingsRL, GaoS-J (2006) Recurrent genomic imbalances in primary effusion lymphomas. Cancer Genet Cytogenet 171: 119–121.

25. SiddiquiN, BordenKLB (2012) mRNA export and cancer. Wiley Interdiscip Rev RNA 3: 13–25.

26. ReedR, ChengH (2005) TREX, SR proteins and export of mRNA. Curr Opin Cell Biol 17: 269–273.

27. LunaR, RondónAG, AguileraA (2012) New clues to understand the role of THO and other functionally related factors in mRNP biogenesis. Biochim Biophys Acta 1819: 514–520.

28. SchumannS, JacksonBR, Baquero-PerezB, WhitehouseA (2013) Kaposi's sarcoma-associated herpesvirus ORF57 protein: exploiting all stages of viral mRNA processing. Viruses 5: 1901–1923.

29. Culjkovic-KraljacicB, BordenKLB (2013) Aiding and abetting cancer: mRNA export and the nuclear pore. Trends Cell Biol 23: 328–335.

30. Dominguez-SanchezM, SaezC, JaponM, AguileraA, LunaR (2011) Differential expression of THOC1 and ALY mRNP biogenesis/export factors in human cancers. BMC Cancer 11: 77.

31. YamazakiT, FujiwaraN, YukinagaH, EbisuyaM, ShikiT, et al. (2010) The Closely Related RNA helicases, UAP56 and URH49, Preferentially Form Distinct mRNA Export Machineries and Coordinately Regulate Mitotic Progression. Mol Biol Cell 21: 2953–2965.

32. HuertasP, AguileraA (2003) Cotranscriptionally Formed DNA:RNA Hybrids Mediate Transcription Elongation Impairment and Transcription-Associated Recombination. Mol Cell 12: 711–721.

33. Dominguez-SanchezMS, BarrosoS, Gomez-GonzalezB, LunaR, AguileraA (2011) Genome instability and transcription elongation impairment in human cells depleted of THO/TREX. PLoS Genet 7: e1002386.

34. JacksonBR, NoerenbergM, WhitehouseA (2012) The Kaposi's sarcoma-associated herpesvirus ORF57 protein and its multiple roles in mRNA biogenesis. Frontiers Micro 3: 59.

35. Sandri-GoldinRM (2008) The many roles of the regulatory protein ICP27 during herpes simplex virus infection. Front Biosci 13: 5241–5256.

36. TothZ, StammingerT (2008) The human cytomegalovirus regulatory protein UL69 and its effect on mRNA export. Front Biosci 13: 2939–2949.

37. GoodwinDJ, HallKT, GilesMS, CalderwoodMA, MarkhamAF, et al. (2000) The carboxy terminus of the herpesvirus saimiri ORF 57 gene contains domains that are required for transactivation and transrepression. J Gen Virol 81: 2253–2265.

38. MalikP, BlackbournDJ, ChengMF, HaywardGS, ClementsJB (2004) Functional co-operation between the Kaposi's sarcoma-associated herpesvirus ORF57 and ORF50 regulatory proteins. J Gen Virol 85: 2155–2166.

39. PalmeriD, SpadavecchiaS, CarrollKD, LukacDM (2007) Promoter- and cell-specific transcriptional transactivation by the Kaposi's sarcoma-associated herpesvirus ORF57/Mta protein. J Virol 81: 13299–13314.

40. MajerciakV, YamanegiK, AllemandE, KruhlakM, KrainerAR, et al. (2008) Kaposi's sarcoma-associated herpesvirus ORF57 functions as a viral splicing factor and promotes expression of intron-containing viral lytic genes in spliceosome-mediated RNA splicing. J Virol 82: 2792–2801.

41. BoyneJR, JacksonBR, TaylorA, MacnabSA, WhitehouseA (2010) Kaposi's sarcoma-associated herpesvirus ORF57 protein interacts with PYM to enhance translation of viral intronless mRNAs. Embo J 29: 1851–1864.

42. BoyneJR, JacksonBR, WhitehouseA (2010) ORF57: Master regulator of KSHV mRNA biogenesis. Cell Cycle 9: 2702–2703.

43. BoyneJR, WhitehouseA (2006) gamma-2 Herpes virus post-transcriptional gene regulation. Clin Microbiol Infect 12: 110–117.

44. MassimelliMJ, KangJ-G, MajerciakV, LeS-Y, LiewehrD, et al. (2011) Stability of a long noncoding viral RNA depends on a 9-nt core element at the RNA 5′ end to interact with viral ORF57 and cellular PABPC1. Int J Biol Sci 7: 1145–1160.

45. MassimelliMJ, MajerciakV, KruhlakM, ZhengZ-M (2013) Interplay between Polyadenylate-Binding Protein 1 and Kaposi's Sarcoma-Associated Herpesvirus ORF57 in Accumulation of Polyadenylated Nuclear RNA, a Viral Long Noncoding RNA. J Virol 87: 243–256.

46. SeiE, ConradNK (2011) Delineation of a core RNA element required for Kaposi's sarcoma-associated herpesvirus ORF57 binding and activity. Virology 419: 107–116.

47. JacksonBR, BoyneJR, NoerenbergM, TaylorA, HautbergueGM, et al. (2011) An interaction between KSHV ORF57 and UIF provides mRNA-adaptor redundancy in herpesvirus intronless mRNA export. PLoS Pathog 7: e1002138.

48. MalikP, BlackbournDJ, ClementsJB (2004) The evolutionarily conserved Kaposi's sarcoma-associated herpesvirus ORF57 protein interacts with REF protein and acts as an RNA export factor. J Biol Chem 279: 33001–33011.

49. BoyneJR, ColganKJ, WhitehouseA (2008) Recruitment of the complete hTREX complex is required for Kaposi's sarcoma-associated herpesvirus intronless mRNA nuclear export and virus replication. PLoS Pathog 4: e1000194.

50. HanZ, SwaminathanS (2006) Kaposi's sarcoma-associated herpesvirus lytic gene ORF57 is essential for infectious virion production. J Virol 80: 5251–5260.

51. NakamuraH, LuM, GwackY, SouvlisJ, ZeichnerSL, et al. (2003) Global Changes in Kaposi's Sarcoma-Associated Virus Gene Expression Patterns following Expression of a Tetracycline-Inducible Rta Transactivator. J Virol 77: 4205–4220.

52. YuanJ, AdamskiR, ChenJ (2010) Focus on histone variant H2AX: To be or not to be. FEBS Lett 584: 3717–3724.

53. MajerciakV, KruhlakM, DagurPK, McCoyJPJr, ZhengZM (2010) Caspase-7 cleavage of Kaposi sarcoma-associated herpesvirus ORF57 confers a cellular function against viral lytic gene expression. J Biol Chem 285: 11297–11307.

54. JhaHC, UpadhyaySK, AJMP, LuJ, CaiQ, et al. (2013) H2AX Phosphorylation Is Important for LANA-Mediated Kaposi's Sarcoma-Associated Herpesvirus Episome Persistence. J Virol 87: 5255–5269.

55. OngS-E, BlagoevB, KratchmarovaI, KristensenDB, SteenH, et al. (2002) Stable Isotope Labeling by Amino Acids in Cell Culture, SILAC, as a Simple and Accurate Approach to Expression Proteomics. Mol Cell Proteomics 1: 376–386.

56. MundayDC, SurteesR, EmmottE, DoveBK, DigardP, et al. (2012) Using SILAC and quantitative proteomics to investigate the interactions between viral and host proteomes. Proteomics 12: 666–672.

57. CoxJ, MannM (2008) MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat Biotech 26: 1367–1372.

58. CoxJr, NeuhauserN, MichalskiA, ScheltemaRA, OlsenJV, et al. (2011) Andromeda: A Peptide Search Engine Integrated into the MaxQuant Environment. J Proteome Res 10: 1794–1805.

59. FenechM, Kirsch-VoldersM, NatarajanAT, SurrallesJ, CrottJW, et al. (2011) Molecular mechanisms of micronucleus, nucleoplasmic bridge and nuclear bud formation in mammalian and human cells. Mutagenesis 26: 125–132.

60. VieiraJ, O'HearnPM (2004) Use of the red fluorescent protein as a marker of Kaposi's sarcoma-associated herpesvirus lytic gene expression. Virology 325: 225–240.

61. TaylorA, JacksonBR, NoerenbergM, HughesDJ, BoyneJR, et al. (2011) Mutation of a C-terminal motif affects KSHV ORF57 RNA binding, nuclear trafficking and multimerisation. J Virol 85: 7881–7891.

62. Gomez-GonzalezB, Felipe-AbrioI, AguileraA (2009) The S-phase checkpoint is required to respond to R-loops accumulated in THO mutants. Mol Cell Biol 29: 5203–5213.

63. WahbaL, AmonJD, KoshlandD, Vuica-RossM (2011) RNase H and Multiple RNA Biogenesis Factors Cooperate to Prevent RNA:DNA Hybrids from Generating Genome Instability. Mol Cell 44: 978–988.

64. DickersonSK, MarketE, BesmerE, PapavasiliouFN (2003) AID Mediates Hypermutation by Deaminating Single Stranded DNA. J Exp Med 197: 1291–1296.

65. TianX, Devi-RaoG, GolovanovAP, Sandri-GoldinRM (2013) The Interaction of the Cellular Export Adaptor Protein Aly/REF with ICP27 Contributes to the Efficiency of Herpes Simplex Virus 1 mRNA export. J Virol 87: 7210–7217.

66. VolcyK, FraserN (2013) DNA damage promotes herpes simplex virus-1 protein expression in a neuroblastoma cell line. J Neurovirol 19: 57–64.

67. TunnicliffeRB, HautbergueGM, KalraP, JacksonBR, WhitehouseA, et al. (2011) Structural basis for the recognition of cellular mRNA export factor REF by herpes viral proteins HSV-1 ICP27 and HVS ORF57. PLoS Pathog 7: e1001244.

68. BoyneJR, WhitehouseA (2009) Nucleolar disruption impairs Kaposi's sarcoma-associated herpesvirus ORF57-mediated nuclear export of intronless viral mRNAs. FEBS Lett 583: 3549–3556.

69. KoopalS, FuruhjelmJH, JärviluomaA, JäämaaS, PyakurelP, et al. (2007) Viral Oncogene–Induced DNA Damage Response Is Activated in Kaposi Sarcoma Tumorigenesis. PLoS Pathog 3: e140.

70. SinghVV, DuttaD, AnsariMA, DuttaS, ChandranB (2014) Kaposi's Sarcoma-Associated Herpesvirus Induces the ATM and H2AX DNA Damage Response Early during De Novo Infection of Primary Endothelial Cells, Which Play Roles in Latency Establishment. J Virol 88: 2821–2834.

71. ChandranB (2010) Early Events in Kaposi's Sarcoma-Associated Herpesvirus Infection of Target Cells. J Virol 84: 2188–2199.

72. MajerciakV, PripuzovaN, McCoyJP, GaoSJ, ZhengZM (2007) Targeted disruption of Kaposi's sarcoma-associated herpesvirus ORF57 in the viral genome is detrimental for the expression of ORF59, K8alpha, and K8.1 and the production of infectious virus. J Virol 81: 1062–1071.

73. KrishnanHH, NaranattPP, SmithMS, ZengL, BloomerC, et al. (2004) Concurrent Expression of Latent and a Limited Number of Lytic Genes with Immune Modulation and Antiapoptotic Function by Kaposi's Sarcoma-Associated Herpesvirus Early during Infection of Primary Endothelial and Fibroblast Cells and Subsequent Decline of Lytic Gene Expression. J Virol 78: 3601–3620.

74. ShirataN, KudohA, DaikokuT, TatsumiY, FujitaM, et al. (2005) Activation of Ataxia Telangiectasia-mutated DNA Damage Checkpoint Signal Transduction Elicited by Herpes Simplex Virus Infection. J Biol Chem 280: 30336–30341.

75. KudohA, FujitaM, ZhangL, ShirataN, DaikokuT, et al. (2005) Epstein-Barr Virus Lytic Replication Elicits ATM Checkpoint Signal Transduction While Providing an S-phase-like Cellular Environment. J Biol Chem 280: 8156–8163.

76. GasparM, ShenkT (2006) Human cytomegalovirus inhibits a DNA damage response by mislocalizing checkpoint proteins. Proc Natl Acad Sci U S A 103: 2821–2826.

77. LischkaP, TothZ, ThomasM, MuellerR, StammingerT (2006) The UL69 Transactivator Protein of Human Cytomegalovirus Interacts with DEXD/H-Box RNA Helicase UAP56 To Promote Cytoplasmic Accumulation of Unspliced RNA. Mol Cell Biol 26: 1631–1643.

78. HiriartE, FarjotG, GruffatH, NguyenMVC, SergeantA, et al. (2003) A Novel Nuclear Export Signal and a REF Interaction Domain Both Promote mRNA Export by the Epstein-Barr Virus EB2 Protein. J Biol Chem 278: 335–342.

79. BoyneJR, ColganKJ, WhitehouseA (2008) Herpesvirus saimiri ORF57: a post-transcriptional regulatory protein. Front Biosci 13: 2928–2938.

80. RaoCV, YangY-M, SwamyMV, LiuT, FangY, et al. (2005) Colonic tumorigenesis in BubR1+/–ApcMin/+ compound mutant mice is linked to premature separation of sister chromatids and enhanced genomic instability. Proc Natl Acad Sci U S A 102: 4365–4370.

81. Hatch EM, Fischer AH, Deerinck TJ, Hetzer MW (2013) Catastrophic Nuclear Envelope Collapse in Cancer Cell Micronuclei. Cell 154: 47–60.

82. ten AsbroekALMA, van GroenigenM, NooijM, BaasF (2002) The involvement of human ribonucleases H1 and H2 in the variation of response of cells to antisense phosphorothioate oligonucleotides. Euro J Biochem 269: 583–592.

83. GriffithsDA, Abdul-SadaH, KnightLM, JacksonBR, RichardsK, et al. (2013) Merkel cell polyomavirus small T antigen targets the NEMO adaptor protein to disrupt inflammatory signalling. J Virol 87: 13853–13867.

84. GouldF, HarrisonSM, HewittEW, WhitehouseA (2009) Kaposi's sarcoma-associated herpesvirus RTA promotes degradation of the Hey1 repressor protein through the ubiquitin proteasome pathway. J Virol 83: 6727–6738.

85. GoodwinDJ, WhitehouseA (2001) A gamma-2 herpesvirus nucleocytoplasmic shuttle protein interacts with importin alpha 1 and alpha 5. J Biol Chem 276: 19905–19912.

86. GriffithsR, WhitehouseA (2007) Herpesvirus saimiri episomal persistence is maintained via interaction between open reading frame 73 and the cellular chromosome-associated protein MeCP2. J Virol 81: 4021–4032.

87. HallKT, GilesMS, CalderwoodMA, GoodwinDJ, MatthewsDA, et al. (2002) The Herpesvirus Saimiri Open Reading Frame 73 Gene Product Interacts with the Cellular Protein p32. J Virol 76: 11612–11622.

88. StevensonAJ, ClarkeD, MeredithDM, KinseySE, WhitehouseA, et al. (2000) Herpesvirus saimiri-based gene delivery vectors maintain heterologous expression throughout mouse embryonic stem cell differentiation in vitro. Gene Ther 7: 464–471.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 Číslo 5
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#