#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Chromatin Landscapes of Retroviral and Transposon Integration Profiles


Retroviruses and transposons are widely used in cancer research and gene therapy. However, these systems show integration biases that may strongly affect results. To address this issue, we generated very large datasets consisting of to unselected integrations for the Sleeping Beauty and piggyBac transposons, and the Mouse Mammary Tumor Virus (MMTV). We analyzed (epi)genomic features to generate bias maps at local and genome-wide scales. MMTV showed a remarkably uniform distribution of integrations across the genome, and a striking similarity was observed between piggyBac and the Murine Leukemia Virus. Moreover, we find that target site selection is directed at multiple scales. At larger scales, it is similar across systems, and directed by a set of domain-oriented features, including chromatin compaction, replication timing, and CpG islands. Notable differences between systems are defined at smaller scales by a diverse range of epigenetic features. As a practical application of our findings, we determined that three recent insertional mutagenesis screens - commonly used for cancer gene discovery - contained 7%–33% putative false positive integration hotspots.


Vyšlo v časopise: Chromatin Landscapes of Retroviral and Transposon Integration Profiles. PLoS Genet 10(4): e32767. doi:10.1371/journal.pgen.1004250
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004250

Souhrn

Retroviruses and transposons are widely used in cancer research and gene therapy. However, these systems show integration biases that may strongly affect results. To address this issue, we generated very large datasets consisting of to unselected integrations for the Sleeping Beauty and piggyBac transposons, and the Mouse Mammary Tumor Virus (MMTV). We analyzed (epi)genomic features to generate bias maps at local and genome-wide scales. MMTV showed a remarkably uniform distribution of integrations across the genome, and a striking similarity was observed between piggyBac and the Murine Leukemia Virus. Moreover, we find that target site selection is directed at multiple scales. At larger scales, it is similar across systems, and directed by a set of domain-oriented features, including chromatin compaction, replication timing, and CpG islands. Notable differences between systems are defined at smaller scales by a diverse range of epigenetic features. As a practical application of our findings, we determined that three recent insertional mutagenesis screens - commonly used for cancer gene discovery - contained 7%–33% putative false positive integration hotspots.


Zdroje

1. CartierN, Hacein-Bey-AbinaS, BartholomaeCC, VeresG, SchmidtM, et al. (2009) Hematopoietic stem cell gene therapy with a lentiviral vector in X-Linked adrenoleukodystrophy. Science 326: 818–823.

2. FischerA, Hacein-Bey-AbinaS, Cavazzana-CalvoM (2010) 20 years of gene therapy for SCID. Nat Immunol 11: 457–460.

3. UrenAG, KoolJ, MatentzogluK, de RidderJ, MattisonJ, et al. (2008) Large-scale mutagenesis in p19(ARF)- and p53-deficient mice identifies cancer genes and their collaborative networks. Cell 133: 727–741.

4. MattisonJ, KoolJ, UrenAG, de RidderJ, WesselsL, et al. (2010) Novel candidate cancer genes identified by a large-scale cross-species comparative oncogenomics approach. Cancer Res 70: 883–895.

5. AkhtarW, de JongJ, PindyurinAV, PagieL, MeulemanW, et al. (2013) Chromatin position effects assayed by thousands of reporters integrated in parallel. Cell 154: 914–27.

6. RufS, SymmonsO, UsluVV, DolleD, HotC, et al. (2011) Large-scale analysis of the regulatory architecture of the mouse genome with a transposon-associated sensor. Nat Genet 43: 379–386.

7. MillerEH, ObernostererG, RaabenM, HerbertAS, DeffieuMS, et al. (2012) Ebola virus entry requires the host-programmed recognition of an intracellular receptor. EMBO J 31 (8) 1947–60.

8. BouwmanP, AlyA, EscandellJM, PieterseM, BartkovaJ, et al. (2010) 53BP1 loss rescues BRCA1 deficiency and is associated with triple-negative and BRCA-mutated breast cancers. Nat Struct & Mol Biol 17: 688–695.

9. Hacein-Bey-AbinaS, GarrigueA, WangGP, SoulierJ, LimA, et al. (2008) Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of scid-x1. J Clin Invest 118: 3132–3142.

10. LiuG, GeurtsAM, YaeK, SrinivasanAR, FahrenkrugSC, et al. (2005) Target-site preferences of sleeping beauty transposons. J Mol Biol 346: 161–73.

11. GeurtsAM, HackettCS, BellJB, BergemannTL, CollierLS, et al. (2006) Structure-based prediction of insertionsite preferences of transposons into chromosomes. Nucleic acids research 34: 2803–2811.

12. CopelandNG, JenkinsNA (2010) Harnessing transposons for cancer gene discovery. Nat Rev Cancer 10: 696–706.

13. VandendriesscheT, IvicsZ, IzsvakZ, ChuahMK (2009) Emerging potential of transposons for gene therapy and generation of induced pluripotent stem cells. Blood 114: 1461–1468.

14. LiangQ, KongJ, StalkerJ, BradleyA (2009) Chromosomal mobilization and reintegration of sleeping beauty and piggybac transposons. Genesis 47: 404–8.

15. BerryC, HannenhalliS, LeipzigJ, BushmanFD (2006) Selection of target sites for mobile DNA integration in the human genome. PLoS Comput Biol 2: e157.

16. BaluB, ChauhanC, MaherS, ShoueD, KissingerJ, et al. (2009) piggyBac is an effective tool for functional analysis of the plasmodium falciparum genome. BMC Microbiol 9: 83+.

17. GalvanDL, NakazawaY, KajaA, KettlunC, CooperLJ, et al. (2009) Genome-wide mapping of PiggyBac transposon integrations in primary human T cells. J Immunother 32: 837–844.

18. LiMA, PettittSJ, EckertS, NingZ, RiceS, et al. (2013) The piggybac transposon displays local and distant reintegration preferences and can cause mutations at non-canonical integration sites. Mol Cell Biol 33 (7) 1317–30.

19. WangW, LinC, LuD, NingZ, CoxT, et al. (2008) Chromosomal transposition of PiggyBac in mouse embryonic stem cells. P Natl Acad of Sci USA 105: 9290–9295.

20. FaschingerA, RouaultF, JohannesS, LukasA, SalmonsB, et al. (2007) Mouse Mammary Tumor Virus integration site selection in human and mouse genomes. J Virol 82: 1360–1367.

21. FeliceB, CattoglioC, CittaroD, TestaA, MiccioA, et al. (2009) Transcription factor binding sites are genetic determinants of retroviral integration in the human genome. PLoS One 4: e4571.

22. SantoniFA, HartleyO, LubanJ (2010) Deciphering the code for retroviral integration target site selection. PLoS Comput Biol 6: e1001008.

23. BradyT, LeeYN, RonenK, MalaniN, BerryCC, et al. (2009) Integration target site selection by a resurrected human endogenous retrovirus. Genes & Development 23: 633–642.

24. KodamaY, ShumwayM, LeinonenR (2012) International Nucleotide Sequence Database Collaboration (2012) The sequence read archive: explosive growth of sequencing data. Nucleic acids research 40: D54–6.

25. BergemannTL, StarrTK, YuH, SteinbachM, ErdmannJ, et al. (2012) New methods for finding common insertion sites and co-occurring common insertion sites in transposon- and virus-based genetic screens. Nucleic Acids Res 40: 3822–33.

26. AbelU, DeichmannA, NowrouziA, GabrielR, BartholomaeCC, et al. (2011) Analyzing the number of common integration sites of viral vectors new methods and computer programs. PLoS ONE 6: e24247.

27. BiffiA, BartolomaeC, CesanaD, CartierN, AubourgP, et al. (2011) Lentiviral vector common integration sites in preclinical models and a clinical trial reect a benign integration bias and not oncogenic selection. Blood 117: 5332–5339.

28. WuX, LukeB, BurgessS (2006) Redefining the common insertion site. Virology 344: 292–295.

29. StarrTK, AllaeiR, SilversteinKAT, StaggsRA, SarverAL, et al. (2009) A Transposon-Based genetic screen in mice identifies genes altered in colorectal cancer. Science 323: 1747–1750.

30. de JongJ, de RidderJ, van der WeydenL, SunN, van UitertM, et al. (2011) Computational identification of insertional mutagenesis targets for cancer gene discovery. Nucleic Acids Res 39: e105.

31. KoudijsMJ, KlijnC, van der WeydenL, KoolJ, ten HoeveJ, et al. (2011) High-throughput semiquantitative analysis of insertional mutations in heterogeneous tumors. Genome Res 21: 2181–2189.

32. RadR, RadL, WangW, CadinanosJ, VassiliouG, et al. (2010) PiggyBac transposon mutagenesis: A tool for cancer gene discovery in mice. Science 330: 1104–1107.

33. de RidderJ, UrenA, KoolJ, ReindersM, WesselsL (2006) Detecting statistically significant common insertion sites in retroviral insertional mutagenesis screens. PLoS Comput Biol 2: e166.

34. HeinzS, BennerC, SpannN, BertolinoE, LinYC, et al. (2010) Simple combinations of Lineage-Determining transcription factors prime cis-Regulatory elements required for macrophage and b cell identities. Mol Cell 38: 576–589.

35. MitchellRS, BeitzelBF, SchroderAR, ShinnP, ChenH, et al. (2004) Retroviral DNA integration: ASLV, HIV, and MLV show distinct target site preferences. PLoS Biol 2: e234.

36. WuX, LiY, CriseB, BurgessSM (2003) Transcription Start Regions in the Human Genome Are Favored Targets for MLV Integration. Science 300: 1749–1751.

37. KoolJ, UrenAG, MartinsCP, SieD, de RidderJ, et al. (2010) Insertional mutagenesis in mice deficient for p15Ink4b, p16Ink4a, p21Cip1, and p27Kip1 reveals cancer gene interactions and correlations with tumor phenotypes. Cancer Res 70: 520–531.

38. BeltonJMM, McCordRPP, GibcusJHH, NaumovaN, ZhanY, et al. (2012) Hi-C: A comprehensive technique to capture the conformation of genomes. Methods 58: 268–276.

39. DixonJR, SelvarajS, YueF, KimA, LiY, et al. (2012) Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature 485: 376–380.

40. HarteminkAJ (2005) Reverse engineering gene regulatory networks. Nat Biotechnol 23: 554–555.

41. Pearl J (1985) Bayesian networks: A model of self-activated memory for evidential reasoning. In: Proceedings of the 7th Conference of the Cognitive Science Society, University of California, Irvine. pp. 329–334.

42. MikkersH, AllenJ, KnipscheerP, RomeijnL, HartA, et al. (2002) High-throughput retroviral tagging to identify components of specific signaling pathways in cancer. Nat Genet 32: 153–159.

43. LundAH, TurnerG, TrubetskoyA, VerhoevenE, WientjensE, et al. (2002) Genome-wide retro-viral insertional tagging of genes involved in cancer in Cdkn2a-deficient mice. Nat Genet 32: 160–165.

44. UrenAG, KoolJ, BernsA, van LohuizenM (2005) Retroviral insertional mutagenesis: past, present and future. Oncogene 24: 7656–7672.

45. CollierLS, CarlsonCM, RavimohanS, DupuyAJ, LargaespadaDA (2005) Cancer gene discovery in solid tumours using transposon-based somatic mutagenesis in the mouse. Nature 436: 272–276.

46. Robertson ES (2011) Cancer Associated Viruses. Current Cancer Research. Springer.

47. TheodorouV, KimmMA, BoerM, WesselsL, TheelenW, et al. (2007) MMTV insertional mutagenesis identifies genes, gene families and pathways involved in mammary cancer. Nature Genetics 39: 759–769.

48. KageyMH, NewmanJJ, BilodeauS, ZhanY, OrlandoDA, et al. (2010) Mediator and cohesin connect gene expression and chromatin architecture. Nature 467: 430–435.

49. MikkelsenTS, KuM, JaffeDB, IssacB, LiebermanE, et al. (2007) Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature 448: 553–560.

50. Bilodeau S, Young R (2010) ChIPSeq for H3K79me2 (unpublished). GSE accession: GSE26680.

51. CreyghtonMP, ChengAW, WelsteadGG, KooistraT, CareyBW, et al. (2010) Histone H3K27ac separates active from poised enhancers and predicts developmental state. P Natl Acad Sci USA 107: 21931–21936.

52. ShenY, YueF, McClearyDF, YeZ, EdsallL, et al. (2012) A map of the cis-regulatory sequences in the mouse genome. Nature 488: 116–120.

53. MasuiS, ShimosatoD, ToyookaY, YagiR, TakahashiK, et al. (2005) An efficient system to establish multiple embryonic stem cell lines carrying an inducible expression unit. Nucleic Acids Res 33: e43.

54. YingQL, WrayJ, NicholsJ, Batlle-MoreraL, DobleB, et al. (2008) The ground state of embryonic stem cell self-renewal. Nature 453: 519–23.

55. WangW, YangJ, LiuH, LuD, ChenX, et al. (2011) Rapid and efficient reprogramming of somatic cells to induced pluripotent stem cells by retinoic acid receptor gamma and liver receptor homolog 1. P Natl Acad Sci USA

56. CadinanosJ, BradleyA (2007) Generation of an inducible and optimized piggybac transposon system. Nucleic Acids Res 35: e87.

57. MatesL, ChuahMKL, BelayE, JerchowB, ManojN, et al. (2009) Molecular evolution of a novel hyperactive sleeping beauty transposase enables robust stable gene transfer in vertebrates. Nat Genet 41: 753–761.

58. LangmeadB, SalzbergSL (2012) Fast gapped-read alignment with bowtie 2. Nat Meth 9: 357–359.

59. LangmeadB, TrapnellC, PopM, SalzbergS (2009) Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol 10: R25–10.

60. BarrettT, TroupDB, WilhiteSE, LedouxP, EvangelistaC, et al. (2010) NCBI GEO: archive for functional genomics data sets–10 years on. Nucleic Acids Res 39: D1005–D1010.

61. XuY, WuF, TanL, KongL, XiongL, et al. (2011) Genome-wide regulation of 5hmC, 5mC, and gene expression by tet1 hydroxylase in mouse embryonic stem cells. Mol Cell 42: 451–464.

62. PedersenB, HsiehTF, IbarraC, FischerRL (2011) MethylCoder: software pipeline for bisulfitetreated sequences. Bioinformatics 27: 2435–2436.

63. TrapnellC, WilliamsBA, PerteaG, MortazaviA, KwanG, et al. (2010) Transcript assembly and quantification by RNA-seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat Biotechnol 28: 511–515.

64. LienertF, MohnF, TiwariVK, BaubecT, RoloffTC, et al. (2011) Genomic prevalence of heterochromatic H3K9me2 and transcription do not discriminate pluripotent from terminally differentiated cells. PLoS Genet 7: e1002090+.

65. HirataniI, RybaT, ItohM, YokochiT, SchwaigerM, et al. (2008) Global reorganization of replication domains during embryonic stem cell differentiation. PLoS Biol 6: e245+.

66. Peric-HupkesD, MeulemanW, PagieL, BruggemanSWM, SoloveiI, et al. (2010) Molecular maps of the reorganization of genome-nuclear lamina interactions during differentiation. Mol Cell 38: 603–613.

67. ChangC, YangX, PursellB, MercurioAM (2013) Id2 complexes with the snag domain of snai1 inhibiting snai1-mediated repression of integrin beta-4. Mol Cell Biol 33 (19) 3795–804.

68. FlicekP, AmodeMR, BarrellD, BealK, BrentS, et al. (2012) Ensembl 2012. Nucleic Acids Res 40: D84–D90.

69. StamatoyannopoulosJ, SnyderM, HardisonR, RenB, GingerasT, et al. (2012) An encyclopedia of mouse DNA elements (mouse ENCODE). Genome Biol 13: 418+.

70. KarimiMM, GoyalP, MaksakovaIA, BilenkyM, LeungD, et al. (2011) DNA methylation and SETDB1/H3K9me3 regulate predominantly distinct sets of genes, retroelements, and chimeric transcripts in mESCs. Cell Stem Cell 8: 676–687.

71. XiaoS, XieD, CaoX, YuP, XingX, et al. (2012) Comparative epigenomic annotation of regulatory DNA. Cell 149: 1381–1392.

72. LawMJ, LowerKM, VoonHPJ, HughesJR, GarrickD, et al. (2010) ATR-x syndrome protein targets tandem repeats and inuences Allele-Specific expression in a Size-Dependent manner. Cell 143: 367–378.

73. HoL, JothiR, RonanJL, CuiK, ZhaoK, et al. (2009) An embryonic stem cell chromatin remodeling complex, esBAF, is an essential component of the core pluripotency transcriptional network. P Natl Acad Sci USA 106: 5187–5191.

74. ChenX, XuH, YuanP, FangF, HussM, et al. (2008) Integration of external signaling pathways with the core transcriptional network in embryonic stem cells. Cell 133: 1106–1117.

75. MarsonA, LevineSS, ColeMF, FramptonGM, BrambrinkT, et al. (2008) Connecting microRNA genes to the core transcriptional regulatory circuitry of embryonic stem cells. Cell 134: 521–533.

76. KuM, KocheRP, RheinbayE, MendenhallEM, EndohM, et al. (2008) Genomewide analysis of PRC1 and PRC2 occupancy identifies two classes of bivalent domains. PLoS Genet 4: e1000242+.

77. BilodeauS, KageyMH, FramptonGM, RahlPB, YoungRA (2009) SetDB1 contributes to repression of genes encoding developmental regulators and maintenance of ES cell state. Gene Dev 23: 2484–2489.

78. LiG, MargueronR, KuM, ChambonP, BernsteinBE, et al. (2010) Jarid2 and PRC2, partners in regulating gene expression. Gene Dev 24: 368–380.

79. HanJ, YuanP, YangH, ZhangJ, SohBS, et al. (2010) Tbx3 improves the germ-line competency of induced pluripotent stem cells. Nature 463: 1096–100.

80. RahlPB, LinCY, SeilaAC, FlynnRA, McCuineS, et al. (2010) c-Myc regulates transcriptional pause release. Cell 141: 432–445.

81. KrebsAR, DemmersJ, KarmodiyaK, ChangNC, ChangAC, et al. (2010) ATAC and mediator coactivators form a stable complex and regulate a set of non-coding RNA genes. EMBO Rep 11: 541–7.

82. SchnetzMP, HandokoL, Akhtar-ZaidiB, BartelsCF, PereiraCF, et al. (2010) CHD7 targets active gene enhancer elements to modulate ES cell-specific gene expression. PLoS Genet 6.

83. MendenhallEM, KocheRP, TruongT, ZhouVW, IssacB, et al. (2010) GC-rich sequence elements recruit PRC2 in mammalian ES cells. PLoS Genet 6: e1001244+.

84. HandokoL, XuH, LiG, NganCYY, ChewE, et al. (2011) CTCF-mediated functional chromatin interactome in pluripotent cells. Nat Genet 43: 630–638.

85. HezroniH, SailajaBS, MeshorerE (2011) Pluripotency-related, valproic acid (VPA)-induced genome-wide histone h3 lysine 9 (H3K9) acetylation patterns in embryonic stem cells. J Biol Chem 286: 35977–35988.

86. LiuZ, ScannellDR, EisenMB, TjianR (2011) Control of embryonic stem cell lineage commitment by core promoter factor, TAF3. Cell 146: 720–731.

87. SchmitzSU, AlbertM, MalatestaM, MoreyL, JohansenJV, et al. (2011) Jarid1b targets genes regulating development and is involved in neural differentiation. EMBO J 30 (22) 4586–600.

88. MullenAC, OrlandoDA, NewmanJJ, LovénJ, KumarRM, et al. (2011) Master transcription factors determine cell-type-specific responses to TGF-? signaling. Cell 147: 565–576.

89. QuennevilleS, VerdeG, CorsinottiA, KapopoulouA, JakobssonJ, et al. (2011) In embryonic stem cells, ZFP57/KAP1 recognize a methylated hexanucleotide to affect chromatin and DNA methylation of imprinting control regions. Mol Cell 44: 361–372.

90. StadlerMB, MurrR, BurgerL, IvanekR, LienertF, et al. (2011) DNA-binding factors shape the mouse methylome at distal regulatory regions. Nature 480: 490–495.

91. CreppeC, JanichP, CantarinoN, NogueraM, ValeroV, et al. (2012) Macroh2a1 regulates the balance between self-renewal and differentiation commitment in embryonic and adult stem cells. Mol Cell Biol 32: 1442–52.

92. LiM, HeY, DuboisW, WuX, ShiJ, et al. (2012) Distinct regulatory mechanisms and functions for p53-Activated and p53-Repressed DNA damage response genes in embryonic stem cells. Mol Cell 46: 30–42.

93. TavaresL, DimitrovaE, OxleyD, WebsterJ, PootR, et al. (2012) RYBP-PRC1 complexes mediate H2A ubiquitylation at polycomb target sites independently of PRC2 and H3K27me3. Cell 148: 664–678.

94. WhyteWA, BilodeauS, OrlandoDA, HokeHA, FramptonGM, et al. (2012) Enhancer decommissioning by LSD1 during embryonic stem cell differentiation. Nature 482 (7384) 221–5.

95. JiangH, ShuklaA, WangX, ChenWy, BernsteinBE, et al. (2011) Role for dpy-30 in ES Cell-Fate specification by regulation of H3K4 methylation within bivalent domains. Cell 144: 825.

96. YildirimO, LiR, HungJHH, ChenPB, DongX, et al. (2011) Mbd3/NURD complex regulates expression of 5-hydroxymethylcytosine marked genes in embryonic stem cells. Cell 147: 1498–1510.

97. Young R (2011) ChIPSeq for Mcaf1 (unpublished). GSE accession: GSE26680.

98. Young R (2010) ChIPSeq for Tbp (unpublished). GSE accession: GSE22303.

99. Young R (2010) ChIPSeq for PolII-Ser18P (unpublished). GSE accession: GSE21917.

100. SmithER, LinC, GarrettAS, ThorntonJ, MohagheghN, et al. (2011) The little elongation complex regulates small nuclear RNA transcription. Mol Cell 44: 954–965.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2014 Číslo 4
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#