#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Transposable Elements Are Major Contributors to the Origin, Diversification, and Regulation of Vertebrate Long Noncoding RNAs


Advances in vertebrate genomics have uncovered thousands of loci encoding long noncoding RNAs (lncRNAs). While progress has been made in elucidating the regulatory functions of lncRNAs, little is known about their origins and evolution. Here we explore the contribution of transposable elements (TEs) to the makeup and regulation of lncRNAs in human, mouse, and zebrafish. Surprisingly, TEs occur in more than two thirds of mature lncRNA transcripts and account for a substantial portion of total lncRNA sequence (∼30% in human), whereas they seldom occur in protein-coding transcripts. While TEs contribute less to lncRNA exons than expected, several TE families are strongly enriched in lncRNAs. There is also substantial interspecific variation in the coverage and types of TEs embedded in lncRNAs, partially reflecting differences in the TE landscapes of the genomes surveyed. In human, TE sequences in lncRNAs evolve under greater evolutionary constraint than their non–TE sequences, than their intronic TEs, or than random DNA. Consistent with functional constraint, we found that TEs contribute signals essential for the biogenesis of many lncRNAs, including ∼30,000 unique sites for transcription initiation, splicing, or polyadenylation in human. In addition, we identified ∼35,000 TEs marked as open chromatin located within 10 kb upstream of lncRNA genes. The density of these marks in one cell type correlate with elevated expression of the downstream lncRNA in the same cell type, suggesting that these TEs contribute to cis-regulation. These global trends are recapitulated in several lncRNAs with established functions. Finally a subset of TEs embedded in lncRNAs are subject to RNA editing and predicted to form secondary structures likely important for function. In conclusion, TEs are nearly ubiquitous in lncRNAs and have played an important role in the lineage-specific diversification of vertebrate lncRNA repertoires.


Vyšlo v časopise: Transposable Elements Are Major Contributors to the Origin, Diversification, and Regulation of Vertebrate Long Noncoding RNAs. PLoS Genet 9(4): e32767. doi:10.1371/journal.pgen.1003470
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003470

Souhrn

Advances in vertebrate genomics have uncovered thousands of loci encoding long noncoding RNAs (lncRNAs). While progress has been made in elucidating the regulatory functions of lncRNAs, little is known about their origins and evolution. Here we explore the contribution of transposable elements (TEs) to the makeup and regulation of lncRNAs in human, mouse, and zebrafish. Surprisingly, TEs occur in more than two thirds of mature lncRNA transcripts and account for a substantial portion of total lncRNA sequence (∼30% in human), whereas they seldom occur in protein-coding transcripts. While TEs contribute less to lncRNA exons than expected, several TE families are strongly enriched in lncRNAs. There is also substantial interspecific variation in the coverage and types of TEs embedded in lncRNAs, partially reflecting differences in the TE landscapes of the genomes surveyed. In human, TE sequences in lncRNAs evolve under greater evolutionary constraint than their non–TE sequences, than their intronic TEs, or than random DNA. Consistent with functional constraint, we found that TEs contribute signals essential for the biogenesis of many lncRNAs, including ∼30,000 unique sites for transcription initiation, splicing, or polyadenylation in human. In addition, we identified ∼35,000 TEs marked as open chromatin located within 10 kb upstream of lncRNA genes. The density of these marks in one cell type correlate with elevated expression of the downstream lncRNA in the same cell type, suggesting that these TEs contribute to cis-regulation. These global trends are recapitulated in several lncRNAs with established functions. Finally a subset of TEs embedded in lncRNAs are subject to RNA editing and predicted to form secondary structures likely important for function. In conclusion, TEs are nearly ubiquitous in lncRNAs and have played an important role in the lineage-specific diversification of vertebrate lncRNA repertoires.


Zdroje

1. PheasantM, MattickJS (2007) Raising the estimate of functional human sequences. Genome Res 17: 1245–1253.

2. PontingCP, HardisonRC (2011) What fraction of the human genome is functional? Genome Res 21: 1769–1776.

3. GersteinMB, KundajeA, HariharanM, LandtSG, YanKK, et al. (2012) Architecture of the human regulatory network derived from ENCODE data. Nature 489: 91–100.

4. DingerME, AmaralPP, MercerTR, MattickJS (2009) Pervasive transcription of the eukaryotic genome: functional indices and conceptual implications. Brief Funct Genomic Proteomic 8: 407–423.

5. MercerTR, DingerME, MattickJS (2009) Long non-coding RNAs: insights into functions. Nat Rev Genet 10: 155–159.

6. PontingCP, OliverPL, ReikW (2009) Evolution and functions of long noncoding RNAs. Cell 136: 629–641.

7. RinnJL, ChangHY (2012) Genome regulation by long noncoding RNAs. Annu Rev Biochem 81: 145–166.

8. JiaH, OsakM, BoguGK, StantonLW, JohnsonR, et al. (2010) Genome-wide computational identification and manual annotation of human long noncoding RNA genes. RNA 16: 1478–1487.

9. CabiliMN, TrapnellC, GoffL, KoziolM, Tazon-VegaB, et al. (2011) Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev 25: 1915–1927.

10. DerrienT, JohnsonR, BussottiG, TanzerA, DjebaliS, et al. (2012) The GENCODE v7 catalog of human long noncoding RNAs: Analysis of their gene structure, evolution, and expression. Genome Res 22: 1775–1789.

11. DingerME, AmaralPP, MercerTR, PangKC, BruceSJ, et al. (2008) Long noncoding RNAs in mouse embryonic stem cell pluripotency and differentiation. Genome Res 18: 1433–1445.

12. GuttmanM, AmitI, GarberM, FrenchC, LinMF, et al. (2009) Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature 458: 223–227.

13. GuttmanM, GarberM, LevinJZ, DonagheyJ, RobinsonJ, et al. (2010) Ab initio reconstruction of cell type-specific transcriptomes in mouse reveals the conserved multi-exonic structure of lincRNAs. Nat Biotechnol 28: 503–510.

14. UlitskyI, ShkumatavaA, JanCH, SiveH, BartelDP (2011) Conserved function of lincRNAs in vertebrate embryonic development despite rapid sequence evolution. Cell 147: 1537–1550.

15. BuD, YuK, SunS, XieC, SkogerboG, et al. (2012) NONCODE v3.0: integrative annotation of long noncoding RNAs. Nucleic Acids Res 40: D210–215.

16. KutterC, WattS, StefflovaK, WilsonMD, GoncalvesA, et al. (2012) Rapid turnover of long noncoding RNAs and the evolution of gene expression. PLoS Genet 8: e1002841 doi:10.1371/journal.pgen.1002841.

17. YoungRS, MarquesAC, TibbitC, HaertyW, BassettAR, et al. (2012) Identification and properties of 1,119 candidate lincRNA loci in the Drosophila melanogaster genome. Genome Biol Evol 4: 427–442.

18. NamJW, BartelD (2012) Long non-coding RNAs in C. elegans. Genome Res

19. LiuJ, JungC, XuJ, WangH, DengS, et al. (2012) Genome-Wide Analysis Uncovers Regulation of Long Intergenic Noncoding RNAs in Arabidopsis. Plant Cell

20. MercerTR, DingerME, SunkinSM, MehlerMF, MattickJS (2008) Specific expression of long noncoding RNAs in the mouse brain. Proc Natl Acad Sci U S A 105: 716–721.

21. PangKC, DingerME, MercerTR, MalquoriL, GrimmondSM, et al. (2009) Genome-wide identification of long noncoding RNAs in CD8+ T cells. J Immunol 182: 7738–7748.

22. SunwooH, DingerME, WiluszJE, AmaralPP, MattickJS, et al. (2009) MEN epsilon/beta nuclear-retained non-coding RNAs are up-regulated upon muscle differentiation and are essential components of paraspeckles. Genome Res 19: 347–359.

23. GuttmanM, DonagheyJ, CareyBW, GarberM, GrenierJK, et al. (2011) lincRNAs act in the circuitry controlling pluripotency and differentiation. Nature 477: 295–300.

24. PauliA, ValenE, LinMF, GarberM, VastenhouwNL, et al. (2012) Systematic identification of long noncoding RNAs expressed during zebrafish embryogenesis. Genome Res 22: 577–591.

25. HungT, WangY, LinMF, KoegelAK, KotakeY, et al. (2011) Extensive and coordinated transcription of noncoding RNAs within cell-cycle promoters. Nat Genet 43: 621–629.

26. CawleyS, BekiranovS, NgHH, KapranovP, SekingerEA, et al. (2004) Unbiased mapping of transcription factor binding sites along human chromosomes 21 and 22 points to widespread regulation of noncoding RNAs. Cell 116: 499–509.

27. WuSC, KallinEM, ZhangY (2010) Role of H3K27 methylation in the regulation of lncRNA expression. Cell Res 20: 1109–1116.

28. KapranovP, ChengJ, DikeS, NixDA, DuttaguptaR, et al. (2007) RNA maps reveal new RNA classes and a possible function for pervasive transcription. Science 316: 1484–1488.

29. PonjavicJ, PontingCP, LunterG (2007) Functionality or transcriptional noise? Evidence for selection within long noncoding RNAs. Genome Res 17: 556–565.

30. MarquesAC, PontingCP (2009) Catalogues of mammalian long noncoding RNAs: modest conservation and incompleteness. Genome Biol 10: R124.

31. JinG, SunJ, IsaacsSD, WileyKE, KimST, et al. (2011) Human polymorphisms at long non-coding RNAs (lncRNAs) and association with prostate cancer risk. Carcinogenesis 32: 1655–1659.

32. WardLD, KellisM (2012) Evidence of abundant purifying selection in humans for recently acquired regulatory functions. Science 337: 1675–1678.

33. ChenG, QiuC, ZhangQ, LiuB, CuiQ (2012) Genome-Wide Analysis of Human SNPs at Long Intergenic Noncoding RNAs. Hum Mutat

34. ChurchDM, GoodstadtL, HillierLW, ZodyMC, GoldsteinS, et al. (2009) Lineage-specific biology revealed by a finished genome assembly of the mouse. PLoS Biol 7: e1000112 doi:10.1371/journal.pbio.1000112.

35. ChodroffRA, GoodstadtL, SireyTM, OliverPL, DaviesKE, et al. (2010) Long noncoding RNA genes: conservation of sequence and brain expression among diverse amniotes. Genome Biol 11: R72.

36. RinnJL, KerteszM, WangJK, SquazzoSL, XuX, et al. (2007) Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell 129: 1311–1323.

37. KhalilAM, GuttmanM, HuarteM, GarberM, RajA, et al. (2009) Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression. Proc Natl Acad Sci U S A 106: 11667–11672.

38. TsaiMC, ManorO, WanY, MosammaparastN, WangJK, et al. (2010) Long noncoding RNA as modular scaffold of histone modification complexes. Science 329: 689–693.

39. OromUA, DerrienT, BeringerM, GumireddyK, GardiniA, et al. (2010) Long noncoding RNAs with enhancer-like function in human cells. Cell 143: 46–58.

40. GuptaRA, ShahN, WangKC, KimJ, HorlingsHM, et al. (2010) Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature 464: 1071–1076.

41. LoewerS, CabiliMN, GuttmanM, LohYH, ThomasK, et al. (2010) Large intergenic non-coding RNA-RoR modulates reprogramming of human induced pluripotent stem cells. Nat Genet 42: 1113–1117.

42. HuarteM, GuttmanM, FeldserD, GarberM, KoziolMJ, et al. (2010) A large intergenic noncoding RNA induced by p53 mediates global gene repression in the p53 response. Cell 142: 409–419.

43. NgSY, JohnsonR, StantonLW (2012) Human long non-coding RNAs promote pluripotency and neuronal differentiation by association with chromatin modifiers and transcription factors. EMBO J 31: 522–533.

44. ChuC, QuK, ZhongFL, ArtandiSE, ChangHY (2011) Genomic maps of long noncoding RNA occupancy reveal principles of RNA-chromatin interactions. Mol Cell 44: 667–678.

45. DuretL, ChureauC, SamainS, WeissenbachJ, AvnerP (2006) The Xist RNA gene evolved in eutherians by pseudogenization of a protein-coding gene. Science 312: 1653–1655.

46. ElisaphenkoEA, KolesnikovNN, ShevchenkoAI, RogozinIB, NesterovaTB, et al. (2008) A dual origin of the Xist gene from a protein-coding gene and a set of transposable elements. PLoS ONE 3: e2521 doi:10.1371/journal.pone.0002521.

47. LanderES, LintonLM, BirrenB, NusbaumC, ZodyMC, et al. (2001) Initial sequencing and analysis of the human genome. Nature 409: 860–921.

48. de KoningAP, GuW, CastoeTA, BatzerMA, PollockDD (2011) Repetitive elements may comprise over two-thirds of the human genome. PLoS Genet 7: e1002384 doi:10.1371/journal.pgen.1002384.

49. FeschotteC (2008) Transposable elements and the evolution of regulatory networks. Nat Rev Genet 9: 397–405.

50. BourqueG (2009) Transposable elements in gene regulation and in the evolution of vertebrate genomes. Curr Opin Genet Dev 19: 607–612.

51. RebolloR, RomanishMT, MagerDL (2012) Transposable Elements: An Abundant and Natural Source of Regulatory Sequences for Host Genes. Annu Rev Genet

52. FeschotteC, GilbertC (2012) Endogenous viruses: insights into viral evolution and impact on host biology. Nat Rev Genet 13: 283–296.

53. JordanIK, RogozinIB, GlazkoGV, KooninEV (2003) Origin of a substantial fraction of human regulatory sequences from transposable elements. Trends Genet 19: 68–72.

54. FaulknerGJ, KimuraY, DaubCO, WaniS, PlessyC, et al. (2009) The regulated retrotransposon transcriptome of mammalian cells. Nat Genet 41: 563–571.

55. WangT, ZengJ, LoweCB, SellersRG, SalamaSR, et al. (2007) Species-specific endogenous retroviruses shape the transcriptional network of the human tumor suppressor protein p53. Proc Natl Acad Sci U S A 104: 18613–18618.

56. BourqueG, LeongB, VegaVB, ChenX, LeeYL, et al. (2008) Evolution of the mammalian transcription factor binding repertoire via transposable elements. Genome Res 18: 1752–1762.

57. KunarsoG, ChiaNY, JeyakaniJ, HwangC, LuX, et al. (2010) Transposable elements have rewired the core regulatory network of human embryonic stem cells. Nat Genet 42: 631–634.

58. BejeranoG, LoweCB, AhituvN, KingB, SiepelA, et al. (2006) A distal enhancer and an ultraconserved exon are derived from a novel retroposon. Nature 441: 87–90.

59. LynchVJ, MayG, WagnerGP (2011) Regulatory evolution through divergence of a phosphoswitch in the transcription factor CEBPB. Nature 480: 383–386.

60. SchmidtD, SchwaliePC, WilsonMD, BallesterB, GoncalvesA, et al. (2012) Waves of retrotransposon expansion remodel genome organization and CTCF binding in multiple mammalian lineages. Cell 148: 335–348.

61. WangJ, LunyakVV, JordanIK (2012) Genome-wide prediction and analysis of human chromatin boundary elements. Nucleic Acids Res 40: 511–529.

62. VolffJN (2006) Turning junk into gold: domestication of transposable elements and the creation of new genes in eukaryotes. Bioessays 28: 913–922.

63. SorekR (2007) The birth of new exons: mechanisms and evolutionary consequences. RNA 13: 1603–1608.

64. NekrutenkoA, LiWH (2001) Transposable elements are found in a large number of human protein-coding genes. Trends Genet 17: 619–621.

65. van de LagemaatLN, LandryJR, MagerDL, MedstrandP (2003) Transposable elements in mammals promote regulatory variation and diversification of genes with specialized functions. Trends Genet 19: 530–536.

66. ZhangXH, ChasinLA (2006) Comparison of multiple vertebrate genomes reveals the birth and evolution of human exons. Proc Natl Acad Sci U S A 103: 13427–13432.

67. SelaN, MerschB, Gal-MarkN, Lev-MaorG, Hotz-WagenblattA, et al. (2007) Comparative analysis of transposed element insertion within human and mouse genomes reveals Alu's unique role in shaping the human transcriptome. Genome Biol 8: R127.

68. SorekR, AstG, GraurD (2002) Alu-containing exons are alternatively spliced. Genome Res 12: 1060–1067.

69. LevyA, SelaN, AstG (2008) TranspoGene and microTranspoGene: transposed elements influence on the transcriptome of seven vertebrates and invertebrates. Nucleic Acids Res 36: D47–52.

70. KrullM, PetrusmaM, MakalowskiW, BrosiusJ, SchmitzJ (2007) Functional persistence of exonized mammalian-wide interspersed repeat elements (MIRs). Genome Res 17: 1139–1145.

71. SmalheiserNR, TorvikVI (2005) Mammalian microRNAs derived from genomic repeats. Trends Genet 21: 322–326.

72. PiriyapongsaJ, Marino-RamirezL, JordanIK (2007) Origin and evolution of human microRNAs from transposable elements. Genetics 176: 1323–1337.

73. HarrowJ, FrankishA, GonzalezJM, TapanariE, DiekhansM, et al. (2012) GENCODE: The reference human genome annotation for The ENCODE Project. Genome Res 22: 1760–1774.

74. DjebaliS, DavisCA, MerkelA, DobinA, LassmannT, et al. (2012) Landscape of transcription in human cells. Nature 489: 101–108.

75. WangF, LiX, XieX, ZhaoL, ChenW (2008) UCA1, a non-protein-coding RNA up-regulated in bladder carcinoma and embryo, influencing cell growth and promoting invasion. FEBS Lett 582: 1919–1927.

76. PanzittK, TschernatschMM, GuellyC, MoustafaT, StradnerM, et al. (2007) Characterization of HULC, a novel gene with striking up-regulation in hepatocellular carcinoma, as noncoding RNA. Gastroenterology 132: 330–342.

77. PrensnerJR, IyerMK, BalbinOA, DhanasekaranSM, CaoQ, et al. (2011) Transcriptome sequencing across a prostate cancer cohort identifies PCAT-1, an unannotated lincRNA implicated in disease progression. Nat Biotechnol 29: 742–749.

78. CartaultF, MunierP, BenkoE, DesguerreI, HaneinS, et al. (2012) Mutation in a primate-conserved retrotransposon reveals a noncoding RNA as a mediator of infantile encephalopathy. Proc Natl Acad Sci U S A 109: 4980–4985.

79. FlockhartRJ, WebsterDE, QuK, MascarenhasN, KovalskiJ, et al. (2012) BRAFV600E remodels the melanocyte transcriptome and induces BANCR to regulate melanoma cell migration. Genome Res 22: 1006–1014.

80. GroteP, WittlerL, HendrixD, KochF, WahrischS, et al. (2013) The Tissue-Specific lncRNA Fendrr Is an Essential Regulator of Heart and Body Wall Development in the Mouse. Dev Cell 24: 206–214.

81. ThurmanRE, RynesE, HumbertR, VierstraJ, MauranoMT, et al. (2012) The accessible chromatin landscape of the human genome. Nature 489: 75–82.

82. DunhamI, KundajeA, AldredSF, CollinsPJ, DavisCA, et al. (2012) An integrated encyclopedia of DNA elements in the human genome. Nature 489: 57–74.

83. HesselberthJR, ChenX, ZhangZ, SaboPJ, SandstromR, et al. (2009) Global mapping of protein-DNA interactions in vivo by digital genomic footprinting. Nat Methods 6: 283–289.

84. XieD, ChenCC, PtaszekLM, XiaoS, CaoX, et al. (2010) Rewirable gene regulatory networks in the preimplantation embryonic development of three mammalian species. Genome Res 20: 804–815.

85. BonnetE, WuytsJ, RouzeP, Van de PeerY (2004) Evidence that microRNA precursors, unlike other non-coding RNAs, have lower folding free energies than random sequences. Bioinformatics 20: 2911–2917.

86. IzsvakZ, IvicsZ, ShimodaN, MohnD, OkamotoH, et al. (1999) Short inverted-repeat transposable elements in teleost fish and implications for a mechanism of their amplification. J Mol Evol 48: 13–21.

87. KentWJ (2002) BLAT–the BLAST-like alignment tool. Genome Res 12: 656–664.

88. BassBL (2002) RNA editing by adenosine deaminases that act on RNA. Annu Rev Biochem 71: 817–846.

89. KimDD, KimTT, WalshT, KobayashiY, MatiseTC, et al. (2004) Widespread RNA editing of embedded alu elements in the human transcriptome. Genome Res 14: 1719–1725.

90. LevanonEY, EisenbergE, YelinR, NemzerS, HalleggerM, et al. (2004) Systematic identification of abundant A-to-I editing sites in the human transcriptome. Nat Biotechnol 22: 1001–1005.

91. AthanasiadisA, RichA, MaasS (2004) Widespread A-to-I RNA editing of Alu-containing mRNAs in the human transcriptome. PLoS Biol 2: e391 doi:10.1371/journal.pbio.0020391.

92. ChenLL, CarmichaelGG (2008) Gene regulation by SINES and inosines: biological consequences of A-to-I editing of Alu element inverted repeats. Cell Cycle 7: 3294–3301.

93. KiranA, BaranovPV (2010) DARNED: a DAtabase of RNa EDiting in humans. Bioinformatics 26: 1772–1776.

94. KelleyDR, RinnJL (2012) Transposable elements reveal a stem cell specific class of long noncoding RNAs. Genome Biol 13: R107.

95. WaterstonRH, Lindblad-TohK, BirneyE, RogersJ, AbrilJF, et al. (2002) Initial sequencing and comparative analysis of the mouse genome. Nature 420: 520–562.

96. RoweHM, TronoD (2011) Dynamic control of endogenous retroviruses during development. Virology 411: 273–287.

97. LeungDC, LorinczMC (2012) Silencing of endogenous retroviruses: when and why do histone marks predominate? Trends Biochem Sci 37: 127–133.

98. SeifarthW, FrankO, ZeilfelderU, SpiessB, GreenwoodAD, et al. (2005) Comprehensive analysis of human endogenous retrovirus transcriptional activity in human tissues with a retrovirus-specific microarray. J Virol 79: 341–352.

99. ConleyAB, PiriyapongsaJ, JordanIK (2008) Retroviral promoters in the human genome. Bioinformatics 24: 1563–1567.

100. CohenCJ, LockWM, MagerDL (2009) Endogenous retroviral LTRs as promoters for human genes: a critical assessment. Gene 448: 105–114.

101. CordauxR, UditS, BatzerMA, FeschotteC (2006) Birth of a chimeric primate gene by capture of the transposase gene from a mobile element. Proc Natl Acad Sci U S A 103: 8101–8106.

102. Lev-MaorG, SorekR, ShomronN, AstG (2003) The birth of an alternatively spliced exon: 3′ splice-site selection in Alu exons. Science 300: 1288–1291.

103. ZhaoJ, OhsumiTK, KungJT, OgawaY, GrauDJ, et al. (2010) Genome-wide identification of polycomb-associated RNAs by RIP-seq. Mol Cell 40: 939–953.

104. GongC, MaquatLE (2011) lncRNAs transactivate STAU1-mediated mRNA decay by duplexing with 3′ UTRs via Alu elements. Nature 470: 284–288.

105. CarrieriC, CimattiL, BiagioliM, BeugnetA, ZucchelliS, et al. (2012) Long non-coding antisense RNA controls Uchl1 translation through an embedded SINEB2 repeat. Nature 491: 454–457.

106. Smit AFA, Hubley R, Green P (1996–2010) RepeatMasker, version 3.0. Available: http://www.repeatmasker.org.

107. KentWJ, SugnetCW, FureyTS, RoskinKM, PringleTH, et al. (2002) The human genome browser at UCSC. Genome Res 12: 996–1006.

108. GoecksJ, NekrutenkoA, TaylorJ (2010) Galaxy: a comprehensive approach for supporting accessible, reproducible, and transparent computational research in the life sciences. Genome Biol 11: R86.

109. BlankenbergD, Von KusterG, CoraorN, AnandaG, LazarusR, et al. (2010) Galaxy: a web-based genome analysis tool for experimentalists. Curr Protoc Mol Biol Chapter 19: Unit 19 10 11–21.

110. GiardineB, RiemerC, HardisonRC, BurhansR, ElnitskiL, et al. (2005) Galaxy: a platform for interactive large-scale genome analysis. Genome Res 15: 1451–1455.

111. QuinlanAR, HallIM (2010) BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26: 841–842.

112. Team RDC (2012) R: A language and environment for statistical computing, version 2.15. Available: http://www.R-project.org.

113. NephS, VierstraJ, StergachisAB, ReynoldsAP, HaugenE, et al. (2012) An expansive human regulatory lexicon encoded in transcription factor footprints. Nature 489: 83–90.

114. JurkaJ, KapitonovVV, PavlicekA, KlonowskiP, KohanyO, et al. (2005) Repbase Update, a database of eukaryotic repetitive elements. Cytogenet Genome Res 110: 462–467.

115. HofackerIL, StadlerPF (2006) Memory efficient folding algorithms for circular RNA secondary structures. Bioinformatics 22: 1172–1176.

116. StickneyHL, SchmutzJ, WoodsIG, HoltzerCC, DicksonMC, et al. (2002) Rapid mapping of zebrafish mutations with SNPs and oligonucleotide microarrays. Genome Res 12: 1929–1934.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 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#