#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Computational Identification of Diverse Mechanisms Underlying Transcription Factor-DNA Occupancy


ChIP-based genome-wide assays of transcription factor (TF) occupancy have emerged as a powerful, high-throughput method to understand transcriptional regulation, especially on a global scale. This has led to great interest in the underlying biochemical mechanisms that direct TF-DNA binding, with the ultimate goal of computationally predicting a TF's occupancy profile in any cellular condition. In this study, we examined the influence of various potential determinants of TF-DNA binding on a much larger scale than previously undertaken. We used a thermodynamics-based model of TF-DNA binding, called “STAP,” to analyze 45 TF-ChIP data sets from Drosophila embryonic development. We built a cross-validation framework that compares a baseline model, based on the ChIP'ed (“primary”) TF's motif, to more complex models where binding by secondary TFs is hypothesized to influence the primary TF's occupancy. Candidates interacting TFs were chosen based on RNA-SEQ expression data from the time point of the ChIP experiment. We found widespread evidence of both cooperative and antagonistic effects by secondary TFs, and explicitly quantified these effects. We were able to identify multiple classes of interactions, including (1) long-range interactions between primary and secondary motifs (separated by ≤150 bp), suggestive of indirect effects such as chromatin remodeling, (2) short-range interactions with specific inter-site spacing biases, suggestive of direct physical interactions, and (3) overlapping binding sites suggesting competitive binding. Furthermore, by factoring out the previously reported strong correlation between TF occupancy and DNA accessibility, we were able to categorize the effects into those that are likely to be mediated by the secondary TF's effect on local accessibility and those that utilize accessibility-independent mechanisms. Finally, we conducted in vitro pull-down assays to test model-based predictions of short-range cooperative interactions, and found that seven of the eight TF pairs tested physically interact and that some of these interactions mediate cooperative binding to DNA.


Vyšlo v časopise: Computational Identification of Diverse Mechanisms Underlying Transcription Factor-DNA Occupancy. PLoS Genet 9(8): e32767. doi:10.1371/journal.pgen.1003571
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003571

Souhrn

ChIP-based genome-wide assays of transcription factor (TF) occupancy have emerged as a powerful, high-throughput method to understand transcriptional regulation, especially on a global scale. This has led to great interest in the underlying biochemical mechanisms that direct TF-DNA binding, with the ultimate goal of computationally predicting a TF's occupancy profile in any cellular condition. In this study, we examined the influence of various potential determinants of TF-DNA binding on a much larger scale than previously undertaken. We used a thermodynamics-based model of TF-DNA binding, called “STAP,” to analyze 45 TF-ChIP data sets from Drosophila embryonic development. We built a cross-validation framework that compares a baseline model, based on the ChIP'ed (“primary”) TF's motif, to more complex models where binding by secondary TFs is hypothesized to influence the primary TF's occupancy. Candidates interacting TFs were chosen based on RNA-SEQ expression data from the time point of the ChIP experiment. We found widespread evidence of both cooperative and antagonistic effects by secondary TFs, and explicitly quantified these effects. We were able to identify multiple classes of interactions, including (1) long-range interactions between primary and secondary motifs (separated by ≤150 bp), suggestive of indirect effects such as chromatin remodeling, (2) short-range interactions with specific inter-site spacing biases, suggestive of direct physical interactions, and (3) overlapping binding sites suggesting competitive binding. Furthermore, by factoring out the previously reported strong correlation between TF occupancy and DNA accessibility, we were able to categorize the effects into those that are likely to be mediated by the secondary TF's effect on local accessibility and those that utilize accessibility-independent mechanisms. Finally, we conducted in vitro pull-down assays to test model-based predictions of short-range cooperative interactions, and found that seven of the eight TF pairs tested physically interact and that some of these interactions mediate cooperative binding to DNA.


Zdroje

1. RoyS, ErnstJ, KharchenkoPV, KheradpourP, NegreN, et al. (2010) Identification of functional elements and regulatory circuits by Drosophila modENCODE. Science 330: 1787–1797.

2. MyersRM, StamatoyannopoulosJ, SnyderM, DunhamI, HardisonRC, et al. (2011) A user's guide to the encyclopedia of DNA elements (ENCODE). PLoS Biol 9: e1001046.

3. YipKY, ChengC, BhardwajN, BrownJB, LengJ, et al. (2012) Classification of human genomic regions based on experimentally determined binding sites of more than 100 transcription-related factors. Genome Biol 13: R48.

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

5. Yanez-CunaJO, DinhHQ, KvonEZ, ShlyuevaD, StarkA (2012) Uncovering cis-regulatory sequence requirements for context-specific transcription factor binding. Genome Res 22: 2018–2030.

6. KaplanT, LiXY, SaboPJ, ThomasS, StamatoyannopoulosJA, et al. (2011) Quantitative models of the mechanisms that control genome-wide patterns of transcription factor binding during early Drosophila development. PLoS Genet 7: e1001290.

7. Pique-RegiR, DegnerJF, PaiAA, GaffneyDJ, GiladY, et al. (2011) Accurate inference of transcription factor binding from DNA sequence and chromatin accessibility data. Genome Res 21: 447–455.

8. BergerMF, BadisG, GehrkeAR, TalukderS, PhilippakisAA, et al. (2008) Variation in homeodomain DNA binding revealed by high-resolution analysis of sequence preferences. Cell 133: 1266–1276.

9. NoyesMB, ChristensenRG, WakabayashiA, StormoGD, BrodskyMH, et al. (2008) Analysis of homeodomain specificities allows the family-wide prediction of preferred recognition sites. Cell 133: 1277–1289.

10. RockelS, GeertzM, MaerklSJ (2012) MITOMI: a microfluidic platform for in vitro characterization of transcription factor-DNA interaction. Methods Mol Biol 786: 97–114.

11. StoltenburgR, ReinemannC, StrehlitzB (2007) SELEX–a (r)evolutionary method to generate high-affinity nucleic acid ligands. Biomol Eng 24: 381–403.

12. SlatteryM, RileyT, LiuP, AbeN, Gomez-AlcalaP, et al. (2011) Cofactor binding evokes latent differences in DNA binding specificity between Hox proteins. Cell 147: 1270–1282.

13. PagansS, PineyroD, KosoyA, BernuesJ, AzorinF (2004) Repression by TTK69 of GAGA-mediated activation occurs in the absence of TTK69 binding to DNA and solely requires the contribution of the POZ/BTB domain of TTK69. J Biol Chem 279: 9725–9732.

14. ZhouX, O'SheaEK (2011) Integrated approaches reveal determinants of genome-wide binding and function of the transcription factor Pho4. Mol Cell 42: 826–836.

15. WorkmanJL, KingstonRE (1992) Nucleosome core displacement in vitro via a metastable transcription factor-nucleosome complex. Science 258: 1780–1784.

16. MorseRH (1993) Nucleosome disruption by transcription factor binding in yeast. Science 262: 1563–1566.

17. HughesAL, JinY, RandoOJ, StruhlK (2012) A Functional Evolutionary Approach to Identify Determinants of Nucleosome Positioning: A Unifying Model for Establishing the Genome-wide Pattern. Mol Cell 48: 5–15.

18. WassonT, HarteminkAJ (2009) An ensemble model of competitive multi-factor binding of the genome. Genome Res 19: 2101–2112.

19. 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.

20. ZhuLJ, ChristensenRG, KazemianM, HullCJ, EnuamehMS, et al. (2011) FlyFactorSurvey: a database of Drosophila transcription factor binding specificities determined using the bacterial one-hybrid system. Nucleic Acids Res 39: D111–117.

21. GraveleyBR, BrooksAN, CarlsonJW, DuffMO, LandolinJM, et al. (2011) The developmental transcriptome of Drosophila melanogaster. Nature 471: 473–479.

22. HeX, ChenCC, HongF, FangF, SinhaS, et al. (2009) A biophysical model for analysis of transcription factor interaction and binding site arrangement from genome-wide binding data. PLoS ONE 4: e8155.

23. RoiderHG, KanhereA, MankeT, VingronM (2007) Predicting transcription factor affinities to DNA from a biophysical model. Bioinformatics 23: 134–141.

24. ZinzenRP, GirardotC, GagneurJ, BraunM, FurlongEE (2009) Combinatorial binding predicts spatio-temporal cis-regulatory activity. Nature 462: 65–70.

25. StormoGD, FieldsDS (1998) Specificity, free energy and information content in protein-DNA interactions. Trends Biochem Sci 23: 109–113.

26. NibuY, ZhangH, LevineM (2001) Local action of long-range repressors in the Drosophila embryo. EMBO J 20: 2246–2253.

27. SagaiT, HosoyaM, MizushinaY, TamuraM, ShiroishiT (2005) Elimination of a long-range cis-regulatory module causes complete loss of limb-specific Shh expression and truncation of the mouse limb. Development 132: 797–803.

28. ZaretKS, CarrollJS (2011) Pioneer transcription factors: establishing competence for gene expression. Genes Dev 25: 2227–2241.

29. NienCY, LiangHL, ButcherS, SunY, FuS, et al. (2011) Temporal coordination of gene networks by Zelda in the early Drosophila embryo. PLoS Genet 7: e1002339.

30. HarrisonMM, LiXY, KaplanT, BotchanMR, EisenMB (2011) Zelda binding in the early Drosophila melanogaster embryo marks regions subsequently activated at the maternal-to-zygotic transition. PLoS Genet 7: e1002266.

31. SatijaR, BradleyRK (2012) The TAGteam motif facilitates binding of 21 sequence-specific transcription factors in the Drosophila embryo. Genome Res 22: 656–665.

32. KvonEZ, StampfelG, Yanez-CunaJO, DicksonBJ, StarkA (2012) HOT regions function as patterned developmental enhancers and have a distinct cis-regulatory signature. Genes Dev 26: 908–913.

33. GranokH, LeibovitchBA, ShafferCD, ElginSC (1995) Chromatin. Ga-ga over GAGA factor. Curr Biol 5: 238–241.

34. LehmannM (2004) Anything else but GAGA: a nonhistone protein complex reshapes chromatin structure. Trends Genet 20: 15–22.

35. LeibovitchBA, LuQ, BenjaminLR, LiuY, GilmourDS, et al. (2002) GAGA factor and the TFIID complex collaborate in generating an open chromatin structure at the Drosophila melanogaster hsp26 promoter. Mol Cell Biol 22: 6148–6157.

36. OkadaM, HiroseS (1998) Chromatin remodeling mediated by Drosophila GAGA factor and ISWI activates fushi tarazu gene transcription in vitro. Mol Cell Biol 18: 2455–2461.

37. MakeevVJ, LifanovAP, NazinaAG, PapatsenkoDA (2003) Distance preferences in the arrangement of binding motifs and hierarchical levels in organization of transcription regulatory information. Nucleic Acids Res 31: 6016–6026.

38. Barrios-RodilesM, BrownKR, OzdamarB, BoseR, LiuZ, et al. (2005) High-throughput mapping of a dynamic signaling network in mammalian cells. Science 307: 1621–1625.

39. Vizoso PintoMG, VillegasJM, PeterJ, HaaseR, HaasJ, et al. (2009) LuMPIS–a modified luminescence-based mammalian interactome mapping pull-down assay for the investigation of protein-protein interactions encoded by GC-low ORFs. Proteomics 9: 5303–5308.

40. WisotzkeyRG, MehraA, SutherlandDJ, DobensLL, LiuX, et al. (1998) Medea is a Drosophila Smad4 homolog that is differentially required to potentiate DPP responses. Development 125: 1433–1445.

41. InoueH, ImamuraT, IshidouY, TakaseM, UdagawaY, et al. (1998) Interplay of signal mediators of decapentaplegic (Dpp): molecular characterization of mothers against dpp, Medea, and daughters against dpp. Mol Biol Cell 9: 2145–2156.

42. GaoS, SteffenJ, LaughonA (2005) Dpp-responsive silencers are bound by a trimeric Mad-Medea complex. J Biol Chem 280: 36158–36164.

43. GaoS, LaughonA (2007) Flexible interaction of Drosophila Smad complexes with bipartite binding sites. Biochim Biophys Acta 1769: 484–496.

44. CapovillaM, EldonED, PirrottaV (1992) The giant gene of Drosophila encodes a b-ZIP DNA-binding protein that regulates the expression of other segmentation gap genes. Development 114: 99–112.

45. LelliKM, SlatteryM, MannRS (2012) Disentangling the many layers of eukaryotic transcriptional regulation. Annu Rev Genet 46: 43–68.

46. HallikasO, TaipaleJ (2006) High-throughput assay for determining specificity and affinity of protein-DNA binding interactions. Nat Protoc 1: 215–222.

47. ShandalaT, KortschakRD, SaintR (2002) The Drosophila retained/dead ringer gene and ARID gene family function during development. Int J Dev Biol 46: 423–430.

48. GregorySL, KortschakRD, KalionisB, SaintR (1996) Characterization of the dead ringer gene identifies a novel, highly conserved family of sequence-specific DNA-binding proteins. Mol Cell Biol 16: 792–799.

49. AndersonDM, BeresBJ, Wilson-RawlsJ, RawlsA (2009) The homeobox gene Mohawk represses transcription by recruiting the sin3A/HDAC co-repressor complex. Dev Dyn 238: 572–580.

50. ChinnaduraiG (2002) CtBP, an unconventional transcriptional corepressor in development and oncogenesis. Mol Cell 9: 213–224.

51. HolmqvistPH, BoijaA, PhilipP, CronaF, StenbergP, et al. (2012) Preferential genome targeting of the CBP co-activator by Rel and Smad proteins in early Drosophila melanogaster embryos. PLoS Genet 8: e1002769.

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

53. SorianoNS, RussellS (1998) The Drosophila SOX-domain protein Dichaete is required for the development of the central nervous system midline. Development 125: 3989–3996.

54. JolmaA, YanJ, WhitingtonT, ToivonenJ, NittaKR, et al. (2013) DNA-binding specificities of human transcription factors. Cell 152: 327–339.

55. AlleyneTM, Pena-CastilloL, BadisG, TalukderS, BergerMF, et al. (2009) Predicting the binding preference of transcription factors to individual DNA k-mers. Bioinformatics 25: 1012–1018.

56. BigginMD (2011) Animal transcription networks as highly connected, quantitative continua. Dev Cell 21: 611–626.

57. Moyle-HeyrmanG, TimsHS, WidomJ (2011) Structural constraints in collaborative competition of transcription factors against the nucleosome. J Mol Biol 412: 634–646.

58. MirnyLA (2010) Nucleosome-mediated cooperativity between transcription factors. Proc Natl Acad Sci U S A 107: 22534–22539.

59. GordanR, HarteminkAJ, BulykML (2009) Distinguishing direct versus indirect transcription factor-DNA interactions. Genome Res 19: 2090–2100.

60. MacArthurS, LiXY, LiJ, BrownJB, ChuHC, et al. (2009) Developmental roles of 21 Drosophila transcription factors are determined by quantitative differences in binding to an overlapping set of thousands of genomic regions. Genome Biol 10: R80.

61. CelnikerSE, DillonLA, GersteinMB, GunsalusKC, HenikoffS, et al. (2009) Unlocking the secrets of the genome. Nature 459: 927–930.

62. SchuettengruberB, GanapathiM, LeblancB, PortosoM, JaschekR, et al. (2009) Functional anatomy of polycomb and trithorax chromatin landscapes in Drosophila embryos. PLoS Biol 7: e13.

63. BuchlerNE, GerlandU, HwaT (2003) On schemes of combinatorial transcription logic. Proc Natl Acad Sci U S A 100: 5136–5141.

64. Boris Iglewicz DCH (1993) How to detect and handle outliers. Milwaukee: ASQC Quality Press.

65. GrantCharles E, TimothyL, Baileya, NobleWS (2011) FIMO: scanning for occurrences of a given motif. Bioinformatics 27: 1017–1018.

66. Abdi H (2007) Part (Semi Partial) and Partial Regression Coefficients. In: Salkind NJ, editor. Encyclopedia of Measurement and Statistics. New York: SAGE Publications.

67. YuC, WanKH, HammondsAS, StapletonM, CarlsonJW, et al. (2011) Development of expression-ready constructs for generation of proteomic libraries. Methods Mol Biol 723: 257–272.

68. LiangHL, NienCY, LiuHY, MetzsteinMM, KirovN, et al. (2008) The zinc-finger protein Zelda is a key activator of the early zygotic genome in Drosophila. Nature 456: 400–403.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 8
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#