#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Altered Chromatin Occupancy of Master Regulators Underlies Evolutionary Divergence in the Transcriptional Landscape of Erythroid Differentiation


The process whereby blood progenitor cells differentiate into red blood cells, known as erythropoiesis, is very similar between mice and humans. Yet, while studies of this process in mouse have substantially improved our knowledge of human erythropoiesis, recent work has shown a significant divergence in global gene expression across species, suggesting that extrapolation from mouse models to human is not always straightforward. In order to better understand these differences, we have performed a comparative epigenomic analysis of six histone modifications and four master transcription factors. By globally comparing chromatin structure across primary cells and model cell lines in both species, we discovered that while chromatin structure is well conserved at orthologous promoters, subtle changes are predictive of species-specific gene expression. Furthermore, we discovered that the genomic localizations of master transcription factors are poorly conserved, and species-specific losses or gains are associated with changes to the underlying chromatin structure and concomitant gene expression. By using our comparative epigenomics framework, we identified a putative human-specific cis-regulatory module that drives expression of human, but not mouse, GDF15, a gene implicated in iron homeostasis. Our results provide a resource to aid researchers in interpreting genetic and epigenetic differences between species.


Vyšlo v časopise: Altered Chromatin Occupancy of Master Regulators Underlies Evolutionary Divergence in the Transcriptional Landscape of Erythroid Differentiation. PLoS Genet 10(12): e32767. doi:10.1371/journal.pgen.1004890
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004890

Souhrn

The process whereby blood progenitor cells differentiate into red blood cells, known as erythropoiesis, is very similar between mice and humans. Yet, while studies of this process in mouse have substantially improved our knowledge of human erythropoiesis, recent work has shown a significant divergence in global gene expression across species, suggesting that extrapolation from mouse models to human is not always straightforward. In order to better understand these differences, we have performed a comparative epigenomic analysis of six histone modifications and four master transcription factors. By globally comparing chromatin structure across primary cells and model cell lines in both species, we discovered that while chromatin structure is well conserved at orthologous promoters, subtle changes are predictive of species-specific gene expression. Furthermore, we discovered that the genomic localizations of master transcription factors are poorly conserved, and species-specific losses or gains are associated with changes to the underlying chromatin structure and concomitant gene expression. By using our comparative epigenomics framework, we identified a putative human-specific cis-regulatory module that drives expression of human, but not mouse, GDF15, a gene implicated in iron homeostasis. Our results provide a resource to aid researchers in interpreting genetic and epigenetic differences between species.


Zdroje

1. OrkinSH, ZonLI (2008) Hematopoiesis: an evolving paradigm for stem cell biology. Cell 132: 631–644.

2. DoulatovS, NottaF, LaurentiE, DickJE (2012) Hematopoiesis: a human perspective. Cell Stem Cell 10: 120–136.

3. DzierzakE, PhilipsenS (2013) Erythropoiesis: development and differentiation. Cold Spring Harb Perspect Med 3: a011601.

4. SankaranVG, LudwigLS, SicinskaE, XuJ, BauerDE, et al. (2012) Cyclin D3 coordinates the cell cycle during differentiation to regulate erythrocyte size and number. Genes & Development 26: 2075–2087.

5. Merryweather-ClarkeAT, AtzbergerA, SonejiS, GrayN, ClarkK, et al. (2011) Global gene expression analysis of human erythroid progenitors. Blood 117: e96–108.

6. KingsleyPD, Greenfest-AllenE, FrameJM, BushnellTP, MalikJ, et al. (2013) Ontogeny of erythroid gene expression. Blood 121: e5–e13.

7. KellisM, WoldB, SnyderMP, BernsteinBE, KundajeA, et al. (2014) Defining functional DNA elements in the human genome. Proc Natl Acad Sci U S A 111: 6131–6138.

8. SankaranVG, XuJ, RagoczyT, IppolitoGC, WalkleyCR, et al. (2009) Developmental and species-divergent globin switching are driven by BCL11A. Nature 460: 1093–U1050.

9. Valverde-GardunoV, GuyotB, AnguitaE, HamlettI, PorcherC, et al. (2004) Differences in the chromatin structure and cis-element organization of the human and mouse GATA1 loci: implications for cis-element identification. Blood 104: 3106–3116.

10. PisheshaN, ThiruP, ShiJH, EngJC, SankaranVG, et al. (2014) Transcriptional divergence and conservation of human and mouse erythropoiesis. Proceedings of the National Academy of Sciences of the United States of America 111: 4103–4108.

11. AnX, SchulzVP, LiJ, WuK, LiuJ, et al. (2014) Global transcriptome analyses of human and murine terminal erythroid differentiation. Blood 123: 3466–3477.

12. SankaranVG, OrkinSH (2013) Genome-wide association studies of hematologic phenotypes: a window into human hematopoiesis. Current Opinion in Genetics & Development 23: 339–344.

13. TaoJ, ZhuM, WangH, AfelikS, VasievichMP, et al. (2012) SEC23B is required for the maintenance of murine professional secretory tissues. Proc Natl Acad Sci U S A 109: E2001–2009.

14. KhoriatyR, VasievichMP, JonesM, EverettL, ChaseJ, et al. (2014) Absence of a Red Blood Cell Phenotype in Mice with Hematopoietic Deficiency of SEC23B. Mol Cell Biol 34: 3721–3734.

15. PinelloL, XuJ, OrkinSH, YuanGC (2014) Analysis of chromatin-state plasticity identifies cell-type-specific regulators of H3K27me3 patterns. Proc Natl Acad Sci U S A 111: E344–353.

16. Hughes JR, Roberts N, McGowan S, Hay D, Giannoulatou E, et al.. (2014) Analysis of hundreds of cis-regulatory landscapes at high resolution in a single, high-throughput experiment. Nature Genetics 46: 205−+.

17. MayG, SonejiS, TippingAJ, TelesJ, McGowanSJ, et al. (2013) Dynamic analysis of gene expression and genome-wide transcription factor binding during lineage specification of multipotent progenitors. Cell Stem Cell 13: 754–768.

18. PilonAM, AjaySS, KumarSA, SteinerLA, CherukuriPF, et al. (2011) Genome-wide ChIP-Seq reveals a dramatic shift in the binding of the transcription factor erythroid Kruppel-like factor during erythrocyte differentiation. Blood 118: e139–148.

19. SuMY, SteinerLA, BogardusH, MishraT, SchulzVP, et al. (2013) Identification of biologically relevant enhancers in human erythroid cells. J Biol Chem 288: 8433–8444.

20. XuJ, ShaoZ, GlassK, BauerDE, PinelloL, et al. (2012) Combinatorial assembly of developmental stage-specific enhancers controls gene expression programs during human erythropoiesis. Dev Cell 23: 796–811.

21. KadaukeS, UdugamaMI, PawlickiJM, AchtmanJC, JainDP, et al. (2012) Tissue-specific mitotic bookmarking by hematopoietic transcription factor GATA1. Cell 150: 725–737.

22. WuW, ChengY, KellerCA, ErnstJ, KumarSA, et al. (2011) Dynamics of the epigenetic landscape during erythroid differentiation after GATA1 restoration. Genome Res 21: 1659–1671.

23. FujiwaraT, O'GeenH, KelesS, BlahnikK, LinnemannAK, et al. (2009) Discovering Hematopoietic Mechanisms through Genome-wide Analysis of GATA Factor Chromatin Occupancy. Molecular Cell 36: 667–681.

24. WongP, HattangadiSM, ChengAW, FramptonGM, YoungRA, et al. (2011) Gene induction and repression during terminal erythropoiesis are mediated by distinct epigenetic changes. Blood 118: e128–138.

25. HuGQ, SchonesDE, CuiKR, YbarraR, NorthrupD, et al. (2011) Regulation of nucleosome landscape and transcription factor targeting at tissue-specific enhancers by BRG1. Genome Research 21: 1650–1658.

26. ChengY, WuW, KumarSA, YuD, DengW, et al. (2009) Erythroid GATA1 function revealed by genome-wide analysis of transcription factor occupancy, histone modifications, and mRNA expression. Genome Res 19: 2172–2184.

27. ShyuYC, LeeTL, ChenX, HsuPH, WenSC, et al. (2014) Tight regulation of a timed nuclear import wave of EKLF by PKCtheta and FOE during Pro-E to Baso-E transition. Dev Cell 28: 409–422.

28. Kharchenko PV, Alekseyenko AA, Schwartz YB, Minoda A, Riddle NC, et al.. (2011) Comprehensive analysis of the chromatin landscape in Drosophila melanogaster. Nature 471: 480−+.

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

30. WooYH, LiWH (2012) Evolutionary conservation of histone modifications in mammals. Mol Biol Evol 29: 1757–1767.

31. KondoY, ShenL, ChengAS, AhmedS, BoumberY, et al. (2008) Gene silencing in cancer by histone H3 lysine 27 trimethylation independent of promoter DNA methylation. Nat Genet 40: 741–750.

32. Arnold CD, Gerlach D, Spies D, Matts JA, Sytnikova YA, et al.. (2014) Quantitative genome-wide enhancer activity maps for five Drosophila species show functional enhancer conservation and turnover during cis-regulatory evolution. Nat Genet.

33. OdomDT, DowellRD, JacobsenES, GordonW, DanfordTW, et al. (2007) Tissue-specific transcriptional regulation has diverged significantly between human and mouse. Nature Genetics 39: 730–732.

34. MikkelsenTS, XuZ, ZhangX, WangL, GimbleJM, et al. (2010) Comparative epigenomic analysis of murine and human adipogenesis. Cell 143: 156–169.

35. StefflovaK, ThybertD, WilsonMD, StreeterI, AleksicJ, et al. (2013) Cooperativity and rapid evolution of cobound transcription factors in closely related mammals. Cell 154: 530–540.

36. TallackMR, WhitingtonT, YuenWS, WainwrightEN, KeysJR, et al. (2010) A global role for KLF1 in erythropoiesis revealed by ChIP-seq in primary erythroid cells. Genome Res 20: 1052–1063.

37. LovePE, WarzechaC, LiL (2014) Ldb1 complexes: the new master regulators of erythroid gene transcription. Trends Genet 30: 1–9.

38. ErnstJ, KellisM (2012) ChromHMM: automating chromatin-state discovery and characterization. Nat Methods 9: 215–216.

39. KiekhaeferCM, GrassJA, JohnsonKD, BoyerME, BresnickEH (2002) Hematopoietic-specific activators establish an overlapping pattern of histone acetylation and methylation within a mammalian chromatin domain. Proc Natl Acad Sci U S A 99: 14309–14314.

40. ChengC, GersteinM (2012) Modeling the relative relationship of transcription factor binding and histone modifications to gene expression levels in mouse embryonic stem cells. Nucleic Acids Res 40: 553–568.

41. ChengY, KingDC, DoreLC, ZhangXM, ZhouYP, et al. (2008) Transcriptional enhancement by GATA1-occupied DNA segments is strongly associated with evolutionary constraint on the binding site motif. Genome Research 18: 1896–1905.

42. LiQ, PetersonKR, FangX, StamatoyannopoulosG (2002) Locus control regions. Blood 100: 3077–3086.

43. BenderMA, RagoczyT, LeeJ, ByronR, TellingA, et al. (2012) The hypersensitive sites of the murine beta-globin locus control region act independently to affect nuclear localization and transcriptional elongation. Blood 119: 3820–3827.

44. 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.

45. BauerDE, KamranSC, LessardS, XuJ, FujiwaraY, et al. (2013) An erythroid enhancer of BCL11A subject to genetic variation determines fetal hemoglobin level. Science 342: 253–257.

46. SankaranVG, MenneTF, XuJ, AkieTE, LettreG, et al. (2008) Human Fetal Hemoglobin Expression Is Regulated by the Developmental Stage-Specific Repressor BCL11A. Science 322: 1839–1842.

47. SatchwellTJ, PellegrinS, BianchiP, HawleyBR, GampelA, et al. (2013) Characteristic phenotypes associated with congenital dyserythropoietic anemia (type II) manifest at different stages of erythropoiesis. Haematologica 98: 1788–1796.

48. CasanovasG, Vujic SpasicM, CasuC, RivellaS, StrelauJ, et al. (2013) The murine growth differentiation factor 15 is not essential for systemic iron homeostasis in phlebotomized mice. Haematologica 98: 444–447.

49. TannoT, BhanuNV, OnealPA, GohSH, StakerP, et al. (2007) High levels of GDF15 in thalassemia suppress expression of the iron regulatory protein hepcidin. Nat Med 13: 1096–1101.

50. MusallamKM, TaherAT, DucaL, CesarettiC, HalawiR, et al. (2011) Levels of growth differentiation factor-15 are high and correlate with clinical severity in transfusion-independent patients with beta thalassemia intermedia. Blood Cells Mol Dis 47: 232–234.

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

52. HeQ, BardetAF, PattonB, PurvisJ, JohnstonJ, et al. (2011) High conservation of transcription factor binding and evidence for combinatorial regulation across six Drosophila species. Nat Genet 43: 414–420.

53. VillarD, FlicekP, OdomDT (2014) Evolution of transcription factor binding in metazoans - mechanisms and functional implications. Nat Rev Genet 15: 221–233.

54. SankaranVG, GhazvinianR, DoR, ThiruP, VergilioJA, et al. (2012) Exome sequencing identifies GATA1 mutations resulting in Diamond-Blackfan anemia. Journal of Clinical Investigation 122: 2439–2443.

55. JaffrayJA, MitchellWB, GnanapragasamMN, SeshanSV, GuoX, et al. (2013) Erythroid transcription factor EKLF/KLF1 mutation causing congenital dyserythropoietic anemia type IV in a patient of Taiwanese origin: review of all reported cases and development of a clinical diagnostic paradigm. Blood Cells Mol Dis 51: 71–75.

56. LudwigLS, GazdaHT, EngJC, EichhornSW, ThiruP, et al. (2014) Altered translation of GATA1 in Diamond-Blackfan anemia. Nat Med 20: 748–753.

57. CrispinoJD, WeissMJ (2014) Erythro-megakaryocytic transcription factors associated with hereditary anemia. Blood 123: 3080–3088.

58. ArnaudL, SaisonC, HeliasV, LucienN, SteschenkoD, et al. (2010) A dominant mutation in the gene encoding the erythroid transcription factor KLF1 causes a congenital dyserythropoietic anemia. Am J Hum Genet 87: 721–727.

59. SolisC, AizencangGI, AstrinKH, BishopDF, DesnickRJ (2001) Uroporphyrinogen III synthase erythroid promoter mutations in adjacent GATA1 and CP2 elements cause congenital erythropoietic porphyria. J Clin Invest 107: 753–762.

60. MacArthurDG, ManolioTA, DimmockDP, RehmHL, ShendureJ, et al. (2014) Guidelines for investigating causality of sequence variants in human disease. Nature 508: 469–476.

61. SankaranVG, GallagherPG (2013) Applications of high-throughput DNA sequencing to benign hematology. Blood 122: 3575–3582.

62. TeytelmanL, ThurtleDM, RineJ, van OudenaardenA (2013) Highly expressed loci are vulnerable to misleading ChIP localization of multiple unrelated proteins. Proc Natl Acad Sci U S A 110: 18602–18607.

63. ParkD, LeeY, BhupindersinghG, IyerVR (2013) Widespread misinterpretable ChIP-seq bias in yeast. PLoS One 8: e83506.

64. BoyleAP, ArayaCL, BrdlikC, CaytingP, ChengC, et al. (2014) Comparative analysis of regulatory information and circuits across distant species. Nature 512: 453–456.

65. HuangW, LoganantharajR, SchroederB, FargoD, LiL (2013) PAVIS: a tool for Peak Annotation and Visualization. Bioinformatics 29: 3097–3099.

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

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

68. ShenL, ShaoN, LiuX, NestlerE (2014) ngs. plot: Quick mining and visualization of next-generation sequencing data by integrating genomic databases. BMC Genomics 15: 284.

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

70. LiuT (2014) Use model-based Analysis of ChIP-Seq (MACS) to analyze short reads generated by sequencing protein-DNA interactions in embryonic stem cells. Methods Mol Biol 1150: 81–95.

71. MachanickP, BaileyTL (2011) MEME-ChIP: motif analysis of large DNA datasets. Bioinformatics 27: 1696–1697.

72. HegerA, WebberC, GoodsonM, PontingCP, LunterG (2013) GAT: a simulation framework for testing the association of genomic intervals. Bioinformatics 29: 2046–2048.

73. ErnstJ, KheradpourP, MikkelsenTS, ShoreshN, WardLD, et al. (2011) Mapping and analysis of chromatin state dynamics in nine human cell types. Nature 473: 43–49.

74. TrapnellC, RobertsA, GoffL, PerteaG, KimD, et al. (2012) Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat Protoc 7: 562–578.

75. FriedmanJ, HastieT, TibshiraniR (2010) Regularization Paths for Generalized Linear Models via Coordinate Descent. J Stat Softw 33: 1–22.

76. ThorvaldsdottirH, RobinsonJT, MesirovJP (2013) Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief Bioinform 14: 178–192.

77. BlanchetteM, KentWJ, RiemerC, ElnitskiL, SmitAF, et al. (2004) Aligning multiple genomic sequences with the threaded blockset aligner. Genome Res 14: 708–715.

78. SiepelA, BejeranoG, PedersenJS, HinrichsAS, HouM, et al. (2005) Evolutionarily conserved elements in vertebrate, insect, worm, and yeast genomes. Genome Res 15: 1034–1050.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2014 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#