#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Deletion of a Conserved -Element in the Locus Highlights the Role of Acute Histone Acetylation in Modulating Inducible Gene Transcription


Differentiation-dependent regulation of the Ifng cytokine gene locus in T helper (Th) cells has emerged as an excellent model for functional study of distal elements that control lineage-specific gene expression. We previously identified a cis-regulatory element located 22 kb upstream of the Ifng gene (Conserved Non-coding Sequence -22, or CNS-22) that is a site for recruitment of the transcription factors T-bet, Runx3, NF-κB and STAT4, which act to regulate transcription of the Ifng gene in Th1 cells. Here, we report the generation of mice with a conditional deletion of CNS-22 that has enabled us to define the epigenetic and functional consequences of its absence. Deletion of CNS-22 led to a defect in induction of Ifng by the cytokines IL-12 and IL-18, with a more modest effect on induction via T-cell receptor activation. To better understand how CNS-22 and other Ifng CNSs regulated Ifng transcription in response to these distinct stimuli, we examined activation-dependent changes in epigenetic modifications across the extended Ifng locus in CNS-22-deficient T cells. We demonstrate that in response to both cytokine and TCR driven activation signals, CNS-22 and other Ifng CNSs recruit increased activity of histone acetyl transferases (HATs) that transiently enhance levels of histones H3 and H4 acetylation across the extended Ifng locus. We also demonstrate that activation-responsive increases in histone acetylation levels are directly linked to the ability of Ifng CNSs to acutely enhance Pol II recruitment to the Ifng promoter. Finally, we show that impairment in IL-12+IL-18 dependent induction of Ifng stems from the importance of CNS-22 in coordinating locus-wide levels of histone acetylation in response to these cytokines. These findings identify a role for acute histone acetylation in the enhancer function of distal conserved cis-elements that regulate of Ifng gene expression.


Vyšlo v časopise: Deletion of a Conserved -Element in the Locus Highlights the Role of Acute Histone Acetylation in Modulating Inducible Gene Transcription. PLoS Genet 10(1): e32767. doi:10.1371/journal.pgen.1003969
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003969

Souhrn

Differentiation-dependent regulation of the Ifng cytokine gene locus in T helper (Th) cells has emerged as an excellent model for functional study of distal elements that control lineage-specific gene expression. We previously identified a cis-regulatory element located 22 kb upstream of the Ifng gene (Conserved Non-coding Sequence -22, or CNS-22) that is a site for recruitment of the transcription factors T-bet, Runx3, NF-κB and STAT4, which act to regulate transcription of the Ifng gene in Th1 cells. Here, we report the generation of mice with a conditional deletion of CNS-22 that has enabled us to define the epigenetic and functional consequences of its absence. Deletion of CNS-22 led to a defect in induction of Ifng by the cytokines IL-12 and IL-18, with a more modest effect on induction via T-cell receptor activation. To better understand how CNS-22 and other Ifng CNSs regulated Ifng transcription in response to these distinct stimuli, we examined activation-dependent changes in epigenetic modifications across the extended Ifng locus in CNS-22-deficient T cells. We demonstrate that in response to both cytokine and TCR driven activation signals, CNS-22 and other Ifng CNSs recruit increased activity of histone acetyl transferases (HATs) that transiently enhance levels of histones H3 and H4 acetylation across the extended Ifng locus. We also demonstrate that activation-responsive increases in histone acetylation levels are directly linked to the ability of Ifng CNSs to acutely enhance Pol II recruitment to the Ifng promoter. Finally, we show that impairment in IL-12+IL-18 dependent induction of Ifng stems from the importance of CNS-22 in coordinating locus-wide levels of histone acetylation in response to these cytokines. These findings identify a role for acute histone acetylation in the enhancer function of distal conserved cis-elements that regulate of Ifng gene expression.


Zdroje

1. OngCT, CorcesVG (2011) Enhancer function: new insights into the regulation of tissue-specific gene expression. Nat Rev Genet 12: 283–293.

2. FrazerKA, PachterL, PoliakovA, RubinEM, DubchakI (2004) VISTA: computational tools for comparative genomics. Nucleic Acids Res 32: W273–279.

3. LootsGG, OvcharenkoI, PachterL, DubchakI, RubinEM (2002) rVista for comparative sequence-based discovery of functional transcription factor binding sites. Genome Res 12: 832–839.

4. BoyleAP, DavisS, ShulhaHP, MeltzerP, MarguliesEH, et al. (2008) High-resolution mapping and characterization of open chromatin across the genome. Cell 132: 311–322.

5. CrawfordGE, HoltIE, WhittleJ, WebbBD, TaiD, et al. (2006) Genome-wide mapping of DNase hypersensitive sites using massively parallel signature sequencing (MPSS). Genome Res 16: 123–131.

6. BarskiA, ZhaoK (2009) Genomic location analysis by ChIP-Seq. J Cell Biochem 107: 11–18.

7. KieferCM, HouC, LittleJA, DeanA (2008) Epigenetics of beta-globin gene regulation. Mutation research 647: 68–76.

8. AnselKM, DjureticI, TanasaB, RaoA (2006) Regulation of Th2 differentiation and Il4 locus accessibility. Annu Rev Immunol 24: 607–656.

9. SproulD, GilbertN, BickmoreWA (2005) The role of chromatin structure in regulating the expression of clustered genes. Nature reviews Genetics 6: 775–781.

10. ZhuJ, YamaneH, PaulWE (2010) Differentiation of effector CD4 T cell populations. Annu Rev Immunol 28: 445–489.

11. RowellE, MerkenschlagerM, WilsonCB (2008) Long-range regulation of cytokine gene expression. Curr Opin Immunol 20: 272–280.

12. LeeGR, KimST, SpilianakisCG, FieldsPE, FlavellRA (2006) T helper cell differentiation: regulation by cis elements and epigenetics. Immunity 24: 369–379.

13. BalasubramaniA, MukasaR, HattonRD, WeaverCT (2010) Regulation of the Ifng locus in the context of T-lineage specification and plasticity. Immunol Rev 238: 216–232.

14. WilsonCB, RowellE, SekimataM (2009) Epigenetic control of T-helper-cell differentiation. Nat Rev Immunol 9: 91–105.

15. SouttoM, ZhangF, EnersonB, TongY, BoothbyM, et al. (2002) A minimal IFN-γ promoter confers Th1 selective expression. J Immunol 169: 4205–4212.

16. SouttoM, ZhouW, AuneTM (2002) Cutting edge: distal regulatory elements are required to achieve selective expression of IFN-γ in Th1/Tc1 effector cells. J Immunol 169: 6664–6667.

17. HarringtonLE, JanowskiKM, OliverJR, ZajacAJ, WeaverCT (2008) Memory CD4 T cells emerge from effector T-cell progenitors. Nature 452: 356–360.

18. HattonRD, HarringtonLE, LutherRJ, WakefieldT, JanowskiKM, et al. (2006) A distal conserved sequence element controls Ifng gene expression by T cells and NK cells. Immunity 25: 717–729.

19. HadjurS, WilliamsLM, RyanNK, CobbBS, SextonT, et al. (2009) Cohesins form chromosomal cis-interactions at the developmentally regulated IFNG locus. Nature 460: 410–413.

20. SekimataM, Perez-MelgosaM, MillerSA, WeinmannAS, SaboPJ, et al. (2009) CCCTC-binding factor and the transcription factor T-bet orchestrate T helper 1 cell-specific structure and function at the interferon-γ locus. Immunity 31: 551–564.

21. SchoenbornJR, DorschnerMO, SekimataM, SanterDM, ShnyrevaM, et al. (2007) Comprehensive epigenetic profiling identifies multiple distal regulatory elements directing transcription of the gene encoding interferon-γ. Nat Immunol 8: 732–742.

22. ShnyrevaM, WeaverWM, BlanchetteM, TaylorSL, TompaM, et al. (2004) Evolutionarily conserved sequence elements that positively regulate IFN-γ expression in T cells. Proc Natl Acad Sci U S A 101: 12622–12627.

23. LeeDU, AvniO, ChenL, RaoA (2004) A distal enhancer in the interferon-γ (IFN-γ) locus revealed by genome sequence comparison. J Biol Chem 279: 4802–4810.

24. ChangS, AuneTM (2007) Dynamic changes in histone-methylation ‘marks’ across the locus encoding interferon-γ during the differentiation of T helper type 2 cells. Nat Immunol 8: 723–731.

25. MukasaR, BalasubramaniA, LeeYK, WhitleySK, WeaverBT, et al. (2010) Epigenetic instability of cytokine and transcription factor gene loci underlies plasticity of the T helper 17 cell lineage. Immunity 32: 616–627.

26. ChangS, AuneTM (2005) Histone hyperacetylated domains across the Ifng gene region in natural killer cells and T cells. Proc Natl Acad Sci U S A 102: 17095–17100.

27. WeiL, VahediG, SunHW, WatfordWT, TakatoriH, et al. (2010) Discrete roles of STAT4 and STAT6 transcription factors in tuning epigenetic modifications and transcription during T helper cell differentiation. Immunity 32: 840–851.

28. YagiR, JunttilaIS, WeiG, UrbanJFJr, ZhaoK, et al. (2010) The transcription factor GATA3 actively represses RUNX3 protein-regulated production of interferon-gamma. Immunity 32: 507–517.

29. BalasubramaniA, ShibataY, CrawfordGE, BaldwinAS, HattonRD, et al. (2010) Modular utilization of distal cis-regulatory elements controls Ifng gene expression in T cells activated by distinct stimuli. Immunity 33: 35–47.

30. CollinsPL, ChangS, HendersonM, SouttoM, DavisGM, et al. (2010) Distal regions of the human IFNG locus direct cell type-specific expression. J Immunol 185: 1492–1501.

31. CollinsPL, HendersonMA, AuneTM (2012) Diverse functions of distal regulatory elements at the IFNG locus. Journal of immunology 188: 1726–1733.

32. LiuP, JenkinsNA, CopelandNG (2003) A highly efficient recombineering-based method for generating conditional knockout mutations. Genome Res 13: 476–484.

33. WarmingS, CostantinoN, CourtDL, JenkinsNA, CopelandNG (2005) Simple and highly efficient BAC recombineering using galK selection. Nucleic Acids Res 33: e36.

34. WangZ, ZangC, RosenfeldJA, SchonesDE, BarskiA, et al. (2008) Combinatorial patterns of histone acetylations and methylations in the human genome. Nat Genet 40: 897–903.

35. CreyghtonMP, ChengAW, WelsteadGG, KooistraT, CareyBW, et al. (2010) Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc Natl Acad Sci USA 107: 21931–21936.

36. GhislettiS, BarozziI, MiettonF, PollettiS, De SantaF, et al. (2010) Identification and characterization of enhancers controlling the inflammatory gene expression program in macrophages. Immunity 32: 317–328.

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

38. VigneauS, RohrlichPS, BrahicM, BureauJF (2003) Tmevpg1, a candidate gene for the control of Theiler's virus persistence, could be implicated in the regulation of gamma interferon. J Virol 77: 5632–5638.

39. GomezJA, WapinskiOL, YangYW, BureauJF, GopinathS, et al. (2013) The NeST long ncRNA controls microbial susceptibility and epigenetic activation of the interferon-γ locus. Cell 152: 743–754.

40. CollierSP, CollinsPL, WilliamsCL, BoothbyMR, AuneTM (2012) Cutting edge: influence of Tmevpg1, a long intergenic noncoding RNA, on the expression of Ifng by Th1 cells. J Immunol 189: 2084–2088.

41. KaplanMH, SunYL, HoeyT, GrusbyMJ (1996) Impaired IL-12 responses and enhanced development of Th2 cells in Stat4-deficient mice. Nature 382: 174–177.

42. JacobsonNG, SzaboSJ, Weber-NordtRM, ZhongZ, SchreiberRD, et al. (1995) Interleukin 12 signaling in T helper type 1 (Th1) cells involves tyrosine phosphorylation of signal transducer and activator of transcription (Stat)3 and Stat4. J Exp Med 181: 1755–1762.

43. RobinsonD, ShibuyaK, MuiA, ZoninF, MurphyE, et al. (1997) IGIF does not drive Th1 development but synergizes with IL-12 for interferon-γ production and activates IRAK and NFκB. Immunity 7: 571–581.

44. LeeYK, TurnerH, MaynardCL, OliverJR, ChenD, et al. (2009) Late developmental plasticity in the T helper 17 lineage. Immunity 30: 92–107.

45. HarringtonLE, HattonRD, ManganPR, TurnerH, MurphyTL, et al. (2005) Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat Immunol 6: 1123–1132.

46. ScacheriPC, CrawfordGE, DavisS (2006) Statistics for ChIP-chip and DNase hypersensitivity experiments on NimbleGen arrays. Methods Enzymol 411: 270–282.

47. ScacheriPC, CrawfordGE, DavisS (2006) Statistics for ChIP-chip and DNase hypersensitivity experiments on NimbleGen arrays. Meth Enzymol 411: 270–282.

48. NicolJW, HeltGA, BlanchardSG, RajaA, LoraineAE (2009) The Integrated Genome Browser: free software for distribution and exploration of genome-scale datasets. Bioinformatics 25: 2730–2731.

49. CrawfordGE, DavisS, ScacheriPC, RenaudG, HalawiMJ, et al. (2006) DNase-chip: a high-resolution method to identify DNase I hypersensitive sites using tiled microarrays. Nat Methods 3: 503–509.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2014 Číslo 1
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#