#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Clustering of Tissue-Specific Sub-TADs Accompanies the Regulation of Genes in Developing Limbs


HoxA genes exhibit central roles during development and causal mutations have been found in several human syndromes including limb malformation. Despite their importance, information on how these genes are regulated is lacking. Here, we report on the first identification of bona fide transcriptional enhancers controlling HoxA genes in developing limbs and show that these enhancers are grouped into distinct topological domains at the sub-megabase scale (sub-TADs). We provide evidence that target genes and regulatory elements physically interact with each other through contacts between sub-TADs rather than by the formation of discreet “DNA loops”. Interestingly, there is no obvious relationship between the functional domains of the enhancers within the limb and how they are partitioned among the topological domains, suggesting that sub-TAD formation does not rely on enhancer activity. Moreover, we show that suppressing the transcriptional activity of enhancers does not abrogate their contacts with HoxA genes. Based on these data, we propose a model whereby chromatin architecture defines the functional landscapes of enhancers. From an evolutionary standpoint, our data points to the convergent evolution of HoxA and HoxD regulation in the fin-to-limb transition, one of the major morphological innovations in vertebrates.


Vyšlo v časopise: Clustering of Tissue-Specific Sub-TADs Accompanies the Regulation of Genes in Developing Limbs. PLoS Genet 9(12): e32767. doi:10.1371/journal.pgen.1004018
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004018

Souhrn

HoxA genes exhibit central roles during development and causal mutations have been found in several human syndromes including limb malformation. Despite their importance, information on how these genes are regulated is lacking. Here, we report on the first identification of bona fide transcriptional enhancers controlling HoxA genes in developing limbs and show that these enhancers are grouped into distinct topological domains at the sub-megabase scale (sub-TADs). We provide evidence that target genes and regulatory elements physically interact with each other through contacts between sub-TADs rather than by the formation of discreet “DNA loops”. Interestingly, there is no obvious relationship between the functional domains of the enhancers within the limb and how they are partitioned among the topological domains, suggesting that sub-TAD formation does not rely on enhancer activity. Moreover, we show that suppressing the transcriptional activity of enhancers does not abrogate their contacts with HoxA genes. Based on these data, we propose a model whereby chromatin architecture defines the functional landscapes of enhancers. From an evolutionary standpoint, our data points to the convergent evolution of HoxA and HoxD regulation in the fin-to-limb transition, one of the major morphological innovations in vertebrates.


Zdroje

1. KmitaM, TarchiniB, ZakanyJ, LoganM, TabinCJ, et al. (2005) Early developmental arrest of mammalian limbs lacking HoxA/HoxD gene function. Nature 435: 1113–1116.

2. ScottiM, KmitaM (2012) Recruitment of 5′ Hoxa genes in the allantois is essential for proper extra-embryonic function in placental mammals. Development 139(4): 731–9.

3. Medina-MartinezO, BradleyA, Ramirez-SolisR (2000) A large targeted deletion of Hoxb1-Hoxb9 produces a series of single-segment anterior homeotic transformations. Dev Biol 222: 71–83.

4. SpitzF, GonzalezF, PeichelC, VogtTF, DubouleD, et al. (2001) Large scale transgenic and cluster deletion analysis of the HoxD complex separate an ancestral regulatory module from evolutionary innovations. Genes Dev 15: 2209–2214.

5. SuemoriH, NoguchiS (2000) Hox C cluster genes are dispensable for overall body plan of mouse embryonic development. Dev Biol 220: 333–342.

6. Fromental-RamainC, WarotX, MessadecqN, LeMeurM, DolleP, et al. (1996) Hoxa-13 and Hoxd-13 play a crucial role in the patterning of the limb autopod. Development 122: 2997–3011.

7. ShautCA, KeeneDR, SorensenLK, LiDY, StadlerHS (2008) HOXA13 Is essential for placental vascular patterning and labyrinth endothelial specification. PLoS Genet 4: e1000073.

8. Fromental-RamainC, WarotX, LakkarajuS, FavierB, HaackH, et al. (1996) Specific and redundant functions of the paralogous Hoxa-9 and Hoxd-9 genes in forelimb and axial skeleton patterning. Development 122: 461–472.

9. WellikDM, CapecchiMR (2003) Hox10 and Hox11 genes are required to globally pattern the mammalian skeleton. Science 301: 363–367.

10. DavisAP, WitteDP, Hsieh-LiHM, PotterSS, CapecchiMR (1995) Absence of radius and ulna in mice lacking hoxa-11 and hoxd-11. Nature 375: 791–795.

11. BouletAM, CapecchiMR (2004) Multiple roles of Hoxa11 and Hoxd11 in the formation of the mammalian forelimb zeugopod. Development 131: 299–309.

12. ZakanyJ, DubouleD (2007) The role of Hox genes during vertebrate limb development. Current opinion in genetics & development 17: 359–366.

13. TarchiniB, DubouleD (2006) Control of Hoxd genes' collinearity during early limb development. Dev Cell 10: 93–103.

14. MontavonT, DubouleD (2012) Landscapes and archipelagos: spatial organization of gene regulation in vertebrates. Trends Cell Biol 22: 347–354.

15. MontavonT, SoshnikovaN, MascrezB, JoyeE, ThevenetL, et al. (2011) A regulatory archipelago controls Hox genes transcription in digits. Cell 147: 1132–1145.

16. LehoczkyJA, WilliamsME, InnisJW (2004) Conserved expression domains for genes upstream and within the HoxA and HoxD clusters suggests a long-range enhancer existed before cluster duplication. Evolution & development 6: 423–430.

17. LehoczkyJA, InnisJW (2008) BAC transgenic analysis reveals enhancers sufficient for Hoxa13 and neighborhood gene expression in mouse embryonic distal limbs and genital bud. Evolution & development 10: 421–432.

18. DostieJ, BickmoreWA (2012) Chromosome organization in the nucleus - charting new territory across the Hi-Cs. Curr Opin Genet Dev 22: 125–131.

19. EthierSD, MiuraH, DostieJ (2012) Discovering genome regulation with 3C and 3C-related technologies. Biochimica et biophysica acta 1819: 401–410.

20. WilliamsonI, EskelandR, LetticeLA, HillAE, BoyleS, et al. (2012) Anterior-posterior differences in HoxD chromatin topology in limb development. Development 139: 3157–3167.

21. AmanoT, SagaiT, TanabeH, MizushinaY, NakazawaH, et al. (2009) Chromosomal dynamics at the Shh locus: limb bud-specific differential regulation of competence and active transcription. Dev Cell 16: 47–57.

22. MeloCA, DrostJ, WijchersPJ, van de WerkenH, de WitE, et al. (2013) eRNAs are required for p53-dependent enhancer activity and gene transcription. Mol Cell 49: 524–535.

23. VakocCR, LettingDL, GheldofN, SawadoT, BenderMA, et al. (2005) Proximity among distant regulatory elements at the beta-globin locus requires GATA-1 and FOG-1. Mol Cell 17: 453–462.

24. DengW, LeeJ, WangH, MillerJ, ReikA, et al. (2012) Controlling long-range genomic interactions at a native locus by targeted tethering of a looping factor. Cell 149: 1233–1244.

25. RenX, SiegelR, KimU, RoederRG (2011) Direct interactions of OCA-B and TFII-I regulate immunoglobulin heavy-chain gene transcription by facilitating enhancer-promoter communication. Mol Cell 42: 342–355.

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

27. ViselA, BlowMJ, LiZ, ZhangT, AkiyamaJA, et al. (2009) ChIP-seq accurately predicts tissue-specific activity of enhancers. Nature 457: 854–858.

28. CotneyJ, LengJ, OhS, DeMareLE, ReillySK, et al. (2012) Chromatin state signatures associated with tissue-specific gene expression and enhancer activity in the embryonic limb. Genome research 22: 1069–1080.

29. DostieJ, DekkerJ (2007) Mapping networks of physical interactions between genomic elements using 5C technology. Nature protocols 2: 988–1002.

30. FraserJ, EthierSD, MiuraH, DostieJ (2012) A Torrent of Data: Mapping Chromatin Organization Using 5C and High-Throughput Sequencing. Methods in enzymology 513: 113–141.

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

32. NoraEP, LajoieBR, SchulzEG, GiorgettiL, OkamotoI, et al. (2012) Spatial partitioning of the regulatory landscape of the X-inactivation centre. Nature 485: 381–385.

33. Phillips-CreminsJE, SauriaME, SanyalA, GerasimovaTI, LajoieBR, et al. (2013) Architectural Protein Subclasses Shape 3D Organization of Genomes during Lineage Commitment. Cell 153: 1281–1295.

34. MerkenschlagerM, OdomDT (2013) CTCF and cohesin: linking gene regulatory elements with their targets. Cell 152: 1285–1297.

35. DeMareLE, LengJ, CotneyJ, ReillySK, YinJ, et al. (2013) The genomic landscape of cohesin-associated chromatin interactions. Genome Res 23: 1224–1234.

36. LitingtungY, DahnRD, LiY, FallonJF, ChiangC (2002) Shh and Gli3 are dispensable for limb skeleton formation but regulate digit number and identity. Nature 418: 979–983.

37. te WelscherP, ZunigaA, KuijperS, DrenthT, GoedemansHJ, et al. (2002) Progression of vertebrate limb development through SHH-mediated counteraction of GLI3. Science 298: 827–830.

38. MethotN, BaslerK (1999) Hedgehog controls limb development by regulating the activities of distinct transcriptional activator and repressor forms of Cubitus interruptus. Cell 96: 819–831.

39. VokesSA, JiH, WongWH, McMahonAP (2008) A genome-scale analysis of the cis-regulatory circuitry underlying sonic hedgehog-mediated patterning of the mammalian limb. Genes & development 22: 2651–2663.

40. ChiangC, LitingtungY, LeeE, YoungKE, CordenJL, et al. (1996) Cyclopia and defective axial patterning in mice lacking Sonic hedgehog gene function. Nature 383: 407–413.

41. ShethR, GregoireD, DumouchelA, ScottiM, PhamJM, et al. (2013) Decoupling the function of Hox and Shh in developing limb reveals multiple inputs of Hox genes on limb growth. Development 140: 2130–2138.

42. SextonT, YaffeE, KenigsbergE, BantigniesF, LeblancB, et al. (2012) Three-dimensional folding and functional organization principles of the Drosophila genome. Cell 148: 458–472.

43. NoraEP, DekkerJ, HeardE (2013) Segmental folding of chromosomes: A basis for structural and regulatory chromosomal neighborhoods? Bioessays 35(9): 818–28.

44. CavalliG, MisteliT (2013) Functional implications of genome topology. Nat Struct Mol Biol 20: 290–299.

45. AndreyG, MontavonT, MascrezB, GonzalezF, NoordermeerD, et al. (2013) A switch between topological domains underlies HoxD genes collinearity in mouse limbs. Science 340: 1234167.

46. BoettigerAN, LevineM (2009) Synchronous and stochastic patterns of gene activation in the Drosophila embryo. Science 325: 471–473.

47. PerryMW, BoettigerAN, BothmaJP, LevineM (2010) Shadow enhancers foster robustness of Drosophila gastrulation. Curr Biol 20: 1562–1567.

48. SpitzF, GonzalezF, DubouleD (2003) A global control region defines a chromosomal regulatory landscape containing the HoxD cluster. Cell 113: 405–417.

49. GonzalezF, DubouleD, SpitzF (2007) Transgenic analysis of Hoxd gene regulation during digit development. Dev Biol 306: 847–859.

50. DolleP, DierichA, LeMeurM, SchimmangT, SchuhbaurB, et al. (1993) Disruption of the Hoxd-13 gene induces localized heterochrony leading to mice with neotenic limbs. Cell 75: 431–441.

51. KmitaM, FraudeauN, HeraultY, DubouleD (2002) Serial deletions and duplications suggest a mechanism for the collinearity of Hoxd genes in limbs. Nature 420: 145–150.

52. MontavonT, Le GarrecJF, KerszbergM, DubouleD (2008) Modeling Hox gene regulation in digits: reverse collinearity and the molecular origin of thumbness. Genes Dev 22: 346–359.

53. SoshnikovaN, DewaeleR, JanvierP, KrumlaufR, DubouleD (2013) Duplications of hox gene clusters and the emergence of vertebrates. Dev Biol 378: 194–199.

54. WilliamsME, LehoczkyJA, InnisJW (2006) A group 13 homeodomain is neither necessary nor sufficient for posterior prevalence in the mouse limb. Dev Biol 297: 493–507.

55. KondoT, ZakanyJ, DubouleD (1998) Control of colinearity in AbdB genes of the mouse HoxD complex. Molecular cell 1: 289–300.

56. LeeTI, JohnstoneSE, YoungRA (2006) Chromatin immunoprecipitation and microarray-based analysis of protein location. Nature protocols 1: 729–748.

57. MiuraH, TomaruY, NakanishiM, KondoS, HayashizakiY, et al. (2009) Identification of DNA regions and a set of transcriptional regulatory factors involved in transcriptional regulation of several human liver-enriched transcription factor genes. Nucleic acids research 37: 778–792.

58. LangmeadB, TrapnellC, PopM, SalzbergSL (2009) Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome biology 10: R25.

59. ZhangY, LiuT, MeyerCA, EeckhouteJ, JohnsonDS, et al. (2008) Model-based analysis of ChIP-Seq (MACS). Genome biology 9: R137.

60. FerraiuoloMA, RousseauM, MiyamotoC, ShenkerS, WangXQ, et al. (2010) The three-dimensional architecture of Hox cluster silencing. Nucleic acids research 38: 7472–7484.

61. LajoieBR, van BerkumNL, SanyalA, DekkerJ (2009) My5C: web tools for chromosome conformation capture studies. Nature methods 6: 690–691.

62. FraserJ, RousseauM, BlanchetteM, DostieJ (2010) Computing chromosome conformation. Methods in molecular biology 674: 251–268.

63. DostieJ, ZhanY, DekkerJ (2007) Chromosome conformation capture carbon copy technology. Curr Protoc Mol Biol Chapter 21: Unit 21 14.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 12
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#