#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Genome Wide Analysis Reveals Zic3 Interaction with Distal Regulatory Elements of Stage Specific Developmental Genes in Zebrafish


Zic3 regulates early embryonic patterning in vertebrates. Loss of Zic3 function is known to disrupt gastrulation, left-right patterning, and neurogenesis. However, molecular events downstream of this transcription factor are poorly characterized. Here we use the zebrafish as a model to study the developmental role of Zic3 in vivo, by applying a combination of two powerful genomics approaches – ChIP-seq and microarray. Besides confirming direct regulation of previously implicated Zic3 targets of the Nodal and canonical Wnt pathways, analysis of gastrula stage embryos uncovered a number of novel candidate target genes, among which were members of the non-canonical Wnt pathway and the neural pre-pattern genes. A similar analysis in zic3-expressing cells obtained by FACS at segmentation stage revealed a dramatic shift in Zic3 binding site locations and identified an entirely distinct set of target genes associated with later developmental functions such as neural development. We demonstrate cis-regulation of several of these target genes by Zic3 using in vivo enhancer assay. Analysis of Zic3 binding sites revealed a distribution biased towards distal intergenic regions, indicative of a long distance regulatory mechanism; some of these binding sites are highly conserved during evolution and act as functional enhancers. This demonstrated that Zic3 regulation of developmental genes is achieved predominantly through long distance regulatory mechanism and revealed that developmental transitions could be accompanied by dramatic changes in regulatory landscape.


Vyšlo v časopise: Genome Wide Analysis Reveals Zic3 Interaction with Distal Regulatory Elements of Stage Specific Developmental Genes in Zebrafish. PLoS Genet 9(10): e32767. doi:10.1371/journal.pgen.1003852
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003852

Souhrn

Zic3 regulates early embryonic patterning in vertebrates. Loss of Zic3 function is known to disrupt gastrulation, left-right patterning, and neurogenesis. However, molecular events downstream of this transcription factor are poorly characterized. Here we use the zebrafish as a model to study the developmental role of Zic3 in vivo, by applying a combination of two powerful genomics approaches – ChIP-seq and microarray. Besides confirming direct regulation of previously implicated Zic3 targets of the Nodal and canonical Wnt pathways, analysis of gastrula stage embryos uncovered a number of novel candidate target genes, among which were members of the non-canonical Wnt pathway and the neural pre-pattern genes. A similar analysis in zic3-expressing cells obtained by FACS at segmentation stage revealed a dramatic shift in Zic3 binding site locations and identified an entirely distinct set of target genes associated with later developmental functions such as neural development. We demonstrate cis-regulation of several of these target genes by Zic3 using in vivo enhancer assay. Analysis of Zic3 binding sites revealed a distribution biased towards distal intergenic regions, indicative of a long distance regulatory mechanism; some of these binding sites are highly conserved during evolution and act as functional enhancers. This demonstrated that Zic3 regulation of developmental genes is achieved predominantly through long distance regulatory mechanism and revealed that developmental transitions could be accompanied by dramatic changes in regulatory landscape.


Zdroje

1. WareSM, HarutyunyanKG, BelmontJW (2006) Zic3 is critical for early embryonic patterning during gastrulation. Dev Dyn 235: 776–785.

2. ArugaJ (2004) The role of Zic genes in neural development. Mol Cell Neurosci 26: 205–221.

3. GrinblatY, SiveH (2001) zic Gene expression marks anteroposterior pattern in the presumptive neurectoderm of the zebrafish gastrula. Dev Dyn 222: 688–693.

4. NagaiT, ArugaJ, TakadaS, GuntherT, SporleR, et al. (1997) The expression of the mouse Zic1, Zic2, and Zic3 gene suggests an essential role for Zic genes in body pattern formation. Dev Biol 182: 299–313.

5. ArugaJ, NagaiT, TokuyamaT, HayashizakiY, OkazakiY, et al. (1996) The mouse zic gene family. Homologues of the Drosophila pair-rule gene odd-paired. J Biol Chem 271: 1043–1047.

6. BenedykMJ, MullenJR, DiNardoS (1994) odd-paired: a zinc finger pair-rule protein required for the timely activation of engrailed and wingless in Drosophila embryos. Genes Dev 8: 105–117.

7. MerzdorfCS (2007) Emerging roles for zic genes in early development. Dev Dyn 236: 922–940.

8. GrinbergI, MillenKJ (2005) The ZIC gene family in development and disease. Clin Genet 67: 290–296.

9. GebbiaM, FerreroGB, PiliaG, BassiMT, AylsworthA, et al. (1997) X-linked situs abnormalities result from mutations in ZIC3. Nat Genet 17: 305–308.

10. CastAE, GaoC, AmackJD, WareSM (2012) An essential and highly conserved role for Zic3 in left-right patterning, gastrulation and convergent extension morphogenesis. Dev Biol 364: 22–31.

11. KitaguchiT, MizugishiK, HatayamaM, ArugaJ, MikoshibaK (2002) Xenopus Brachyury regulates mesodermal expression of Zic3, a gene controlling left-right asymmetry. Dev Growth Differ 44: 55–61.

12. KitaguchiT, NagaiT, NakataK, ArugaJ, MikoshibaK (2000) Zic3 is involved in the left-right specification of the Xenopus embryo. Development 127: 4787–4795.

13. CampioneM, SteinbeisserH, SchweickertA, DeisslerK, van BebberF, et al. (1999) The homeobox gene Pitx2: mediator of asymmetric left-right signaling in vertebrate heart and gut looping. Development 126: 1225–1234.

14. RyanAK, BlumbergB, Rodriguez-EstebanC, Yonei-TamuraS, TamuraK, et al. (1998) Pitx2 determines left-right asymmetry of internal organs in vertebrates. Nature 394: 545–551.

15. SampathK, RubinsteinAL, ChengAM, LiangJO, FekanyK, et al. (1998) Induction of the zebrafish ventral brain and floorplate requires cyclops/nodal signalling. Nature 395: 185–189.

16. FujimiTJ, HatayamaM, ArugaJ (2012) Xenopus Zic3 controls notochord and organizer development through suppression of the Wnt/beta-catenin signaling pathway. Dev Biol 361: 220–231.

17. PurandareSM, WareSM, KwanKM, GebbiaM, BassiMT, et al. (2002) A complex syndrome of left-right axis, central nervous system and axial skeleton defects in Zic3 mutant mice. Development 129: 2293–2302.

18. NakataK, NagaiT, ArugaJ, MikoshibaK (1997) Xenopus Zic3, a primary regulator both in neural and neural crest development. Proc Natl Acad Sci U S A 94: 11980–11985.

19. MarchalL, LuxardiG, ThomeV, KodjabachianL (2009) BMP inhibition initiates neural induction via FGF signaling and Zic genes. Proc Natl Acad Sci U S A 106: 17437–17442.

20. WeberJR, SokolSY (2003) Identification of a phylogenetically conserved activin-responsive enhancer in the Zic3 gene. Mech Dev 120: 955–964.

21. ENCODE Project Consortium (2004) The ENCODE (ENCyclopedia Of DNA Elements) Project. Science 306: 636–640.

22. SanyalA, LajoieBR, JainG, DekkerJ (2012) The long-range interaction landscape of gene promoters. Nature 489: 109–113.

23. FarnhamPJ (2009) Insights from genomic profiling of transcription factors. Nat Rev Genet 10: 605–616.

24. SpitzF, FurlongEE (2012) Transcription factors: from enhancer binding to developmental control. Nat Rev Genet 13: 613–626.

25. WederellED, BilenkyM, CullumR, ThiessenN, DagpinarM, et al. (2008) Global analysis of in vivo Foxa2-binding sites in mouse adult liver using massively parallel sequencing. Nucleic Acids Res 36: 4549–4564.

26. 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.

27. CarrollJS, LiuXS, BrodskyAS, LiW, MeyerCA, et al. (2005) Chromosome-wide mapping of estrogen receptor binding reveals long-range regulation requiring the forkhead protein FoxA1. Cell 122: 33–43.

28. ParinovS, KondrichinI, KorzhV, EmelyanovA (2004) Tol2 transposon-mediated enhancer trap to identify developmentally regulated zebrafish genes in vivo. Dev Dyn 231: 449–459.

29. AanesH, WinataCL, LinCH, ChenJP, SrinivasanKG, et al. (2011) Zebrafish mRNA sequencing deciphers novelties in transcriptome dynamics during maternal to zygotic transition. Genome Res 21: 1328–1338.

30. SchmitzB, PapanC, Campos-OrtegaJA (1993) Neurulation in the anterior trunk region of the zebrafish Brachydanio rerio. Roux's Arch Dev Biol 202: 250–259.

31. WooK, FraserSE (1995) Order and coherence in the fate map of the zebrafish nervous system. Development 121: 2595–2609.

32. GrinblatY, GamseJ, PatelM, SiveH (1998) Determination of the zebrafish forebrain: induction and patterning. Development 125: 4403–4416.

33. SagerstromCG, GrinbaltY, SiveH (1996) Anteroposterior patterning in the zebrafish, Danio rerio: an explant assay reveals inductive and suppressive cell interactions. Development 122: 1873–1883.

34. KondrychynI, Garcia-LeceaM, EmelyanovA, ParinovS, KorzhV (2009) Genome-wide analysis of Tol2 transposon reintegration in zebrafish. BMC Genomics 10: 418.

35. McLeanCY, BristorD, HillerM, ClarkeSL, SchaarBT, et al. (2010) GREAT improves functional interpretation of cis-regulatory regions. Nat Biotechnol 28: 495–501.

36. ArugaJ, MikoshibaK (2011) Role of BMP, FGF, calcium signaling, and Zic proteins in vertebrate neuroectodermal differentiation. Neurochem Res 36: 1286–1292.

37. AppelB (2000) Zebrafish neural induction and patterning. Dev Dyn 219: 155–168.

38. LimLS, HongFH, KunarsoG, StantonLW (2010) The pluripotency regulator Zic3 is a direct activator of the Nanog promoter in ESCs. Stem Cells 28: 1961–1969.

39. NewburgerDE, BulykML (2009) UniPROBE: an online database of protein binding microarray data on protein-DNA interactions. Nucleic Acids Res 37: D77–82.

40. MizugishiK, ArugaJ, NakataK, MikoshibaK (2001) Molecular properties of Zic proteins as transcriptional regulators and their relationship to GLI proteins. J Biol Chem 276: 2180–2188.

41. KoyabuY, NakataK, MizugishiK, ArugaJ, MikoshibaK (2001) Physical and functional interactions between Zic and Gli proteins. J Biol Chem 276: 6889–6892.

42. ThisseC, ThisseB (1999) Antivin, a novel and divergent member of the TGFbeta superfamily, negatively regulates mesoderm induction. Development 126: 229–240.

43. FaucourtM, HoulistonE, BesnardeauL, KimelmanD, LepageT (2001) The pitx2 homeobox protein is required early for endoderm formation and nodal signaling. Dev Biol 229: 287–306.

44. FeldmanB, DouganST, SchierAF, TalbotWS (2000) Nodal-related signals establish mesendodermal fate and trunk neural identity in zebrafish. Curr Biol 10: 531–534.

45. FeldmanB, GatesMA, EganES, DouganST, RennebeckG, et al. (1998) Zebrafish organizer development and germ-layer formation require nodal-related signals. Nature 395: 181–185.

46. BisgroveBW, EssnerJJ, YostHJ (1999) Regulation of midline development by antagonism of lefty and nodal signaling. Development 126: 3253–3262.

47. EkkerM, AkimenkoMA, AllendeML, SmithR, DrouinG, et al. (1997) Relationships among msx gene structure and function in zebrafish and other vertebrates. Mol Biol Evol 14: 1008–1022.

48. WodaJM, PastagiaJ, MercolaM, ArtingerKB (2003) Dlx proteins position the neural plate border and determine adjacent cell fates. Development 130: 331–342.

49. de la Calle-MustienesE, GlavicA, ModolellJ, Gomez-SkarmetaJL (2002) Xiro homeoproteins coordinate cell cycle exit and primary neuron formation by upregulating neuronal-fate repressors and downregulating the cell-cycle inhibitor XGadd45-gamma. Mech Dev 119: 69–80.

50. LecaudeyV, AnselmeI, DildropR, RutherU, Schneider-MaunouryS (2005) Expression of the zebrafish Iroquois genes during early nervous system formation and patterning. J Comp Neurol 492: 289–302.

51. KorzhV, SleptsovaI, LiaoJ, HeJ, GongZ (1998) Expression of zebrafish bHLH genes ngn1 and nrd defines distinct stages of neural differentiation. Dev Dyn 213: 92–104.

52. WallingfordJB, HarlandRM (2002) Neural tube closure requires Dishevelled-dependent convergent extension of the midline. Development 129: 5815–5825.

53. SongH, HuJ, ChenW, ElliottG, AndreP, et al. (2010) Planar cell polarity breaks bilateral symmetry by controlling ciliary positioning. Nature 466: 378–382.

54. ThirietN, AgasseF, NicoleauC, GueganC, ValletteF, et al. (2011) NPY promotes chemokinesis and neurogenesis in the rat subventricular zone. J Neurochem 116: 1018–1027.

55. YehCM, LiuYC, ChangCJ, LaiSL, HsiaoCD, et al. (2011) Ptenb mediates gastrulation cell movements via Cdc42/AKT1 in zebrafish. PLoS One 6: e18702.

56. GhignaC, GiordanoS, ShenH, BenvenutoF, CastiglioniF, et al. (2005) Cell motility is controlled by SF2/ASF through alternative splicing of the Ron protooncogene. Mol Cell 20: 881–890.

57. SeuxM, PeugetS, MonteroMP, SiretC, RigotV, et al. (2011) TP53INP1 decreases pancreatic cancer cell migration by regulating SPARC expression. Oncogene 30: 3049–3061.

58. GilbertMM, RobinsonBS, MobergKH (2009) Functional interactions between the erupted/tsg101 growth suppressor gene and the DaPKC and rbf1 genes in Drosophila imaginal disc tumors. PLoS One 4: e7039.

59. WangY, KanekoN, AsaiN, EnomotoA, Isotani-SakakibaraM, et al. (2011) Girdin is an intrinsic regulator of neuroblast chain migration in the rostral migratory stream of the postnatal brain. J Neurosci 31: 8109–8122.

60. OharaK, EnomotoA, KatoT, HashimotoT, Isotani-SakakibaraM, et al. (2012) Involvement of Girdin in the determination of cell polarity during cell migration. PLoS One 7: e36681.

61. FisherS, GriceEA, VintonRM, BesslingSL, UrasakiA, et al. (2006) Evaluating the biological relevance of putative enhancers using Tol2 transposon-mediated transgenesis in zebrafish. Nat Protoc 1: 1297–1305.

62. LiG, RuanX, AuerbachRK, SandhuKS, ZhengM, et al. (2012) Extensive promoter-centered chromatin interactions provide a topological basis for transcription regulation. Cell 148: 84–98.

63. TenaJJ, AlonsoME, de la Calle-MustienesE, SplinterE, de LaatW, et al. (2011) An evolutionarily conserved three-dimensional structure in the vertebrate Irx clusters facilitates enhancer sharing and coregulation. Nat Commun 2: 310.

64. EngstromPG, FredmanD, LenhardB (2008) Ancora: a web resource for exploring highly conserved noncoding elements and their association with developmental regulatory genes. Genome Biol 9: R34.

65. 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.

66. SandelinA, BaileyP, BruceS, EngstromPG, KlosJM, et al. (2004) Arrays of ultraconserved non-coding regions span the loci of key developmental genes in vertebrate genomes. BMC Genomics 5: 99.

67. WoolfeA, GoodsonM, GoodeDK, SnellP, McEwenGK, et al. (2005) Highly conserved non-coding sequences are associated with vertebrate development. PLoS Biol 3: e7.

68. VenkateshB, KirknessEF, LohYH, HalpernAL, LeeAP, et al. (2006) Ancient noncoding elements conserved in the human genome. Science 314: 1892.

69. BejeranoG, PheasantM, MakuninI, StephenS, KentWJ, et al. (2004) Ultraconserved elements in the human genome. Science 304: 1321–1325.

70. BlowMJ, McCulleyDJ, LiZ, ZhangT, AkiyamaJA, et al. (2010) ChIP-Seq identification of weakly conserved heart enhancers. Nat Genet 42: 806–810.

71. SchmidtD, WilsonMD, BallesterB, SchwaliePC, BrownGD, et al. (2010) Five-vertebrate ChIP-seq reveals the evolutionary dynamics of transcription factor binding. Science 328: 1036–1040.

72. LeeAP, KerkSY, TanYY, BrennerS, VenkateshB (2011) Ancient vertebrate conserved noncoding elements have been evolving rapidly in teleost fishes. Mol Biol Evol 28: 1205–1215.

73. ChristoffelsA, KohEG, ChiaJM, BrennerS, AparicioS, et al. (2004) Fugu genome analysis provides evidence for a whole-genome duplication early during the evolution of ray-finned fishes. Mol Biol Evol 21: 1146–1151.

74. HoeggS, BrinkmannH, TaylorJS, MeyerA (2004) Phylogenetic timing of the fish-specific genome duplication correlates with the diversification of teleost fish. J Mol Evol 59: 190–203.

75. CrowKD, StadlerPF, LynchVJ, AmemiyaC, WagnerGP (2006) The “fish-specific” Hox cluster duplication is coincident with the origin of teleosts. Mol Biol Evol 23: 121–136.

76. LaneyJD, BigginMD (1997) Zeste-mediated activation by an enhancer is independent of cooperative DNA binding in vivo. Proc Natl Acad Sci U S A 94: 3602–3604.

77. CookPR (1999) The organization of replication and transcription. Science 284: 1790–1795.

78. OuyangZ, ZhouQ, WongWH (2009) ChIP-Seq of transcription factors predicts absolute and differential gene expression in embryonic stem cells. Proc Natl Acad Sci U S A 106: 21521–21526.

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

80. LimLS, LohYH, ZhangW, LiY, ChenX, et al. (2007) Zic3 is required for maintenance of pluripotency in embryonic stem cells. Mol Biol Cell 18: 1348–1358.

81. OrianA, van SteenselB, DelrowJ, BussemakerHJ, LiL, et al. (2003) Genomic binding by the Drosophila Myc, Max, Mad/Mnt transcription factor network. Genes Dev 17: 1101–1114.

82. FernandezPC, FrankSR, WangL, SchroederM, LiuS, et al. (2003) Genomic targets of the human c-Myc protein. Genes Dev 17: 1115–1129.

83. ZeitlingerJ, ZinzenRP, StarkA, KellisM, ZhangH, et al. (2007) Whole-genome ChIP-chip analysis of Dorsal, Twist, and Snail suggests integration of diverse patterning processes in the Drosophila embryo. Genes Dev 21: 385–390.

84. SandmannT, GirardotC, BrehmeM, TongprasitW, StolcV, et al. (2007) A core transcriptional network for early mesoderm development in Drosophila melanogaster. Genes Dev 21: 436–449.

85. RobertsonG, HirstM, BainbridgeM, BilenkyM, ZhaoY, et al. (2007) Genome-wide profiles of STAT1 DNA association using chromatin immunoprecipitation and massively parallel sequencing. Nat Methods 4: 651–657.

86. CaoY, YaoZ, SarkarD, LawrenceM, SanchezGJ, et al. (2010) Genome-wide MyoD binding in skeletal muscle cells: a potential for broad cellular reprogramming. Dev Cell 18: 662–674.

87. MacQuarrieKL, FongAP, MorseRH, TapscottSJ (2011) Genome-wide transcription factor binding: beyond direct target regulation. Trends Genet 27: 141–148.

88. LiXY, ThomasS, SaboPJ, EisenMB, StamatoyannopoulosJA, et al. (2011) The role of chromatin accessibility in directing the widespread, overlapping patterns of Drosophila transcription factor binding. Genome Biol 12: R34.

89. FujiokaM, WuX, JaynesJB (2009) A chromatin insulator mediates transgene homing and very long-range enhancer-promoter communication. Development 136: 3077–3087.

90. ChepelevI, WeiG, WangsaD, TangQ, ZhaoK (2012) Characterization of genome-wide enhancer-promoter interactions reveals co-expression of interacting genes and modes of higher order chromatin organization. Cell Res 22: 490–503.

91. Carmany-RampeyA, SchierAF (2001) Single-cell internalization during zebrafish gastrulation. Curr Biol 11: 1261–1265.

92. MyersDC, SepichDS, Solnica-KrezelL (2002) Convergence and extension in vertebrate gastrulae: cell movements according to or in search of identity? Trends Genet 18: 447–455.

93. Solnica-KrezelL (2006) Gastrulation in zebrafish – all just about adhesion? Curr Opin Genet Dev 16: 433–441.

94. RohdeLA, HeisenbergCP (2007) Zebrafish gastrulation: cell movements, signals, and mechanisms. Int Rev Cytol 261: 159–192.

95. BisgroveBW, EssnerJJ, YostHJ (2000) Multiple pathways in the midline regulate concordant brain, heart and gut left-right asymmetry. Development 127: 3567–3579.

96. DanosMC, YostHJ (1996) Role of notochord in specification of cardiac left-right orientation in zebrafish and Xenopus. Dev Biol 177: 96–103.

97. MorganD, TurnpennyL, GoodshipJ, DaiW, MajumderK, et al. (1998) Inversin, a novel gene in the vertebrate left-right axis pathway, is partially deleted in the inv mouse. Nat Genet 20: 149–156.

98. OttoEA, SchermerB, ObaraT, O'TooleJF, HillerKS, et al. (2003) Mutations in INVS encoding inversin cause nephronophthisis type 2, linking renal cystic disease to the function of primary cilia and left-right axis determination. Nat Genet 34: 413–420.

99. OkadaY, TakedaS, TanakaY, Izpisua BelmonteJC, HirokawaN (2005) Mechanism of nodal flow: a conserved symmetry breaking event in left-right axis determination. Cell 121: 633–644.

100. HashimotoM, ShinoharaK, WangJ, IkeuchiS, YoshibaS, et al. (2010) Planar polarization of node cells determines the rotational axis of node cilia. Nat Cell Biol 12: 170–176.

101. WangG, CadwalladerAB, JangDS, TsangM, YostHJ, et al. (2011) The Rho kinase Rock2b establishes anteroposterior asymmetry of the ciliated Kupffer's vesicle in zebrafish. Development 138: 45–54.

102. WatanabeD, SaijohY, NonakaS, SasakiG, IkawaY, et al. (2003) The left-right determinant Inversin is a component of node monocilia and other 9+0 cilia. Development 130: 1725–1734.

103. May-SimeraHL, KaiM, HernandezV, OsbornDP, TadaM, et al. (2010) Bbs8, together with the planar cell polarity protein Vangl2, is required to establish left-right asymmetry in zebrafish. Dev Biol 345: 215–225.

104. RossAJ, May-SimeraH, EichersER, KaiM, HillJ, et al. (2005) Disruption of Bardet-Biedl syndrome ciliary proteins perturbs planar cell polarity in vertebrates. Nat Genet 37: 1135–1140.

105. HeisenbergCP, TadaM (2002) Wnt signalling: a moving picture emerges from van gogh. Curr Biol 12: R126–128.

106. YenHJ, TayehMK, MullinsRF, StoneEM, SheffieldVC, et al. (2006) Bardet-Biedl syndrome genes are important in retrograde intracellular trafficking and Kupffer's vesicle cilia function. Hum Mol Genet 15: 667–677.

107. ArugaJ, TohmondaT, HommaS, MikoshibaK (2002) Zic1 promotes the expansion of dorsal neural progenitors in spinal cord by inhibiting neuronal differentiation. Dev Biol 244: 329–341.

108. Westerfield M (2000) The zebrafish book. A guide for the laboratory use of zebrafish (Danio rerio). 4th edition. Eugene: Univ. of Oregon Press.

109. KimmelCB, BallardWW, KimmelSR, UllmannB, SchillingTF (1995) Stages of embryonic development of the zebrafish. Dev Dyn 203: 253–310.

110. ValouevA, JohnsonDS, SundquistA, MedinaC, AntonE, et al. (2008) Genome-wide analysis of transcription factor binding sites based on ChIP-Seq data. Nat Methods 5: 829–834.

111. Huang daW, ShermanBT, LempickiRA (2009) Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 4: 44–57.

112. Huang daW, ShermanBT, LempickiRA (2009) Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res 37: 1–13.

113. BaileyTL, ElkanC (1994) Fitting a mixture model by expectation maximization to discover motifs in biopolymers. Proc Int Conf Intell Syst Mol Biol 2: 28–36.

114. TusherVG, TibshiraniR, ChuG (2001) Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci U S A 98: 5116–5121.

115. DorskyRI, SheldahlLC, MoonRT (2002) A transgenic Lef1/beta-catenin-dependent reporter is expressed in spatially restricted domains throughout zebrafish development. Dev Biol 241: 229–237.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 10
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#