#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

The IKAROS Interaction with a Complex Including Chromatin Remodeling and Transcription Elongation Activities Is Required for Hematopoiesis


Perturbation of the expression level of IKAROS, a transcription factor critical during hematopoiesis, is associated with malignant transformation in mice and humans. The importance of IKAROS expression levels for the control of target-gene regulation was addressed in hematopoietic progenitor cells. The collaboration between IKAROS and the Nucleosome Remodeling and Deacetylase (NuRD) complex can promote gene activation or repression. IKAROS can also interact with the Positive-Transcription Elongation Factor b (P-TEFb) and the Protein Phosphatase 1 (PP1), an important P-TEFb regulator. Immunoaffinity purification of IKAROS interacting proteins and Fast Protein Liquid Chromatography analysis revealed a dynamic interaction between IKAROS, PP1 and the newly defined NuRD-P-TEFb complex. This complex can be targeted to specific genes in cells expressing high levels of IKAROS to promote productive transcription elongation. Based on our results we suggest that, in addition to P-TEFb, the NuRD complex and PP1 are required to facilitate transcription elongation of IKAROS-target genes and normal differentiation of hematopoietic progenitor cells.


Vyšlo v časopise: The IKAROS Interaction with a Complex Including Chromatin Remodeling and Transcription Elongation Activities Is Required for Hematopoiesis. PLoS Genet 10(12): e32767. doi:10.1371/journal.pgen.1004827
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004827

Souhrn

Perturbation of the expression level of IKAROS, a transcription factor critical during hematopoiesis, is associated with malignant transformation in mice and humans. The importance of IKAROS expression levels for the control of target-gene regulation was addressed in hematopoietic progenitor cells. The collaboration between IKAROS and the Nucleosome Remodeling and Deacetylase (NuRD) complex can promote gene activation or repression. IKAROS can also interact with the Positive-Transcription Elongation Factor b (P-TEFb) and the Protein Phosphatase 1 (PP1), an important P-TEFb regulator. Immunoaffinity purification of IKAROS interacting proteins and Fast Protein Liquid Chromatography analysis revealed a dynamic interaction between IKAROS, PP1 and the newly defined NuRD-P-TEFb complex. This complex can be targeted to specific genes in cells expressing high levels of IKAROS to promote productive transcription elongation. Based on our results we suggest that, in addition to P-TEFb, the NuRD complex and PP1 are required to facilitate transcription elongation of IKAROS-target genes and normal differentiation of hematopoietic progenitor cells.


Zdroje

1. HeizmannB, KastnerP, ChanS (2013) Ikaros is absolutely required for pre-B cell differentiation by attenuating IL-7 signals. J Exp Med 210: 2823–2832.

2. JohnLB, WardAC (2011) The Ikaros gene family: transcriptional regulators of hematopoiesis and immunity. Mol Immunol 48: 1272–1278.

3. MalingeS, ThiollierC, ChlonTM, DoreLC, DieboldL, et al. (2013) Ikaros inhibits megakaryopoiesis through functional interaction with GATA-1 and NOTCH signaling. Blood 121: 2440–51.

4. NgSY, YoshidaT, ZhangJ, GeorgopoulosK (2009) Genome-wide lineage-specific transcriptional networks underscore Ikaros-dependent lymphoid priming in hematopoietic stem cells. Immunity 30: 493–507.

5. ZhangJ, JacksonAF, NaitoT, DoseM, SeavittJ, et al. (2012) Harnessing of the nucleosome-remodeling-deacetylase complex controls lymphocyte development and prevents leukemogenesis. Nat Immunol 13: 86–94.

6. WangJH, NichogiannopoulouA, WuL, SunL, SharpeAH, et al. (1996) Selective defects in the development of the fetal and adult lymphoid system in mice with an Ikaros null mutation. Immunity 5: 537–549.

7. BottardiS, RossJ, BourgoinV, Fotouhi-ArdakaniN, Affar elB, et al. (2009) Ikaros and GATA-1 combinatorial effect is required for silencing of human gamma-globin genes. Mol Cell Biol 29: 1526–1537.

8. ChoSJ, HuhJE, SongJ, RheeDK, PyoS (2008) Ikaros negatively regulates inducible nitric oxide synthase expression in macrophages: involvement of Ikaros phosphorylation by casein kinase 2. Cell Mol Life Sci 65: 3290–3303.

9. DijonM, BardinF, MuratiA, BatozM, ChabannonC, et al. (2008) The role of Ikaros in human erythroid differentiation. Blood 111: 1138–1146.

10. DumortierA, KirstetterP, KastnerP, ChanS (2003) Ikaros regulates neutrophil differentiation. Blood 101: 2219–2226.

11. PapathanasiouP, AttemaJL, KarsunkyH, HosenN, SontaniY, et al. (2009) Self-renewal of the long-term reconstituting subset of hematopoietic stem cells is regulated by Ikaros. Stem Cells 27: 3082–3092.

12. RossJ, MavoungouL, BresnickEH, MilotE (2012) GATA-1 utilizes Ikaros and polycomb repressive complex 2 to suppress Hes1 and to promote erythropoiesis. Mol Cell Biol 32: 3624–3638.

13. YoshidaT, NgSY, GeorgopoulosK (2010) Awakening lineage potential by Ikaros-mediated transcriptional priming. Curr Opin Immunol 22: 154–160.

14. GeorgopoulosK (2002) Haematopoietic cell-fate decisions, chromatin regulation and ikaros. Nat Rev Immunol 2: 162–174.

15. Ferreiros-VidalI, CarrollT, TaylorB, TerryA, LiangZ, et al. (2013) Genome-wide identification of Ikaros targets elucidates its contribution to mouse B-cell lineage specification and pre-B-cell differentiation. Blood 121: 1769–1782.

16. SchwickertTA, TagohH, GultekinS, DakicA, AxelssonE, et al. (2014) Stage-specific control of early B cell development by the transcription factor Ikaros. Nat Immunol 15: 283–293.

17. KimJ, SifS, JonesB, JacksonA, KoipallyJ, et al. (1999) Ikaros DNA-binding proteins direct formation of chromatin remodeling complexes in lymphocytes. Immunity 10: 345–355.

18. O'NeillDW, SchoetzSS, LopezRA, CastleM, RabinowitzL, et al. (2000) An ikaros-containing chromatin-remodeling complex in adult-type erythroid cells. Mol Cell Biol 20: 7572–7582.

19. MiccioA, WangY, HongW, GregoryGD, WangH, et al. (2010) NuRD mediates activating and repressive functions of GATA-1 and FOG-1 during blood development. EMBO J 29: 442–456.

20. ReynoldsN, LatosP, Hynes-AllenA, LoosR, LeafordD, et al. (2012) NuRD suppresses pluripotency gene expression to promote transcriptional heterogeneity and lineage commitment. Cell Stem Cell 10: 583–594.

21. WhyteWA, BilodeauS, OrlandoDA, HokeHA, FramptonGM, et al. (2012) Enhancer decommissioning by LSD1 during embryonic stem cell differentiation. Nature 482: 221–225.

22. YildirimO, LiR, HungJH, ChenPB, DongX, et al. (2011) Mbd3/NURD complex regulates expression of 5-hydroxymethylcytosine marked genes in embryonic stem cells. Cell 147: 1498–1510.

23. BottardiS, ZmiriFA, BourgoinV, RossJ, MavoungouL, et al. (2011) Ikaros interacts with P-TEFb and cooperates with GATA-1 to enhance transcription elongation. Nucleic Acids Res 39: 3505–3519.

24. HarkerN, NaitoT, CortesM, HostertA, HirschbergS, et al. (2002) The CD8alpha gene locus is regulated by the Ikaros family of proteins. Mol Cell 10: 1403–1415.

25. KoipallyJ, HellerEJ, SeavittJR, GeorgopoulosK (2002) Unconventional potentiation of gene expression by Ikaros. J Biol Chem 277: 13007–13015.

26. QuirionMR, GregoryGD, UmetsuSE, WinandyS, BrownMA (2009) Cutting edge: Ikaros is a regulator of Th2 cell differentiation. J Immunol 182: 741–745.

27. BottardiS, MavoungouL, BourgoinV, MashtalirN, Affar elB, et al. (2013) Direct Protein Interactions Are Responsible for Ikaros-GATA and Ikaros-Cdk9 Cooperativeness in Hematopoietic Cells. Mol Cell Biol 33: 3064–3076.

28. ZhouQ, LiT, PriceDH (2012) RNA polymerase II elongation control. Annu Rev Biochem 81: 119–143.

29. ChenR, LiuM, LiH, XueY, RameyWN, et al. (2008) PP2B and PP1alpha cooperatively disrupt 7SK snRNP to release P-TEFb for transcription in response to Ca2+ signaling. Genes Dev 22: 1356–1368.

30. NekhaiS, PetukhovM, BreuerD (2014) Regulation of CDK9 activity by phosphorylation and dephosphorylation. Biomed Res Int 2014: 964964.

31. D'OrsoI, FrankelAD (2010) HIV-1 Tat: Its Dependence on Host Factors is Crystal Clear. Viruses 2: 2226–2234.

32. JangMK, MochizukiK, ZhouM, JeongHS, BradyJN, et al. (2005) The bromodomain protein Brd4 is a positive regulatory component of P-TEFb and stimulates RNA polymerase II-dependent transcription. Mol Cell 19: 523–534.

33. YangZ, YikJH, ChenR, HeN, JangMK, et al. (2005) Recruitment of P-TEFb for stimulation of transcriptional elongation by the bromodomain protein Brd4. Mol Cell 19: 535–545.

34. CeulemansH, BollenM (2004) Functional diversity of protein phosphatase-1, a cellular economizer and reset button. Physiol Rev 84: 1–39.

35. PopescuM, GurelZ, RonniT, SongC, HungKY, et al. (2009) Ikaros stability and pericentromeric localization are regulated by protein phosphatase 1. J Biol Chem 284: 13869–13880.

36. SridharanR, SmaleST (2007) Predominant interaction of both Ikaros and Helios with the NuRD complex in immature thymocytes. J Biol Chem 282: 30227–30238.

37. RomanoG (2013) Deregulations in the cyclin-dependent kinase-9-related pathway in cancer: implications for drug discovery and development. ISRN Oncol 2013: 305371.

38. LuoZ, LinC, ShilatifardA (2012) The super elongation complex (SEC) family in transcriptional control. Nat Rev Mol Cell Biol 13: 543–547.

39. LinC, SmithER, TakahashiH, LaiKC, Martin-BrownS, et al. (2010) AFF4, a component of the ELL/P-TEFb elongation complex and a shared subunit of MLL chimeras, can link transcription elongation to leukemia. Mol Cell 37: 429–437.

40. WuSY, LeeAY, LaiHT, ZhangH, ChiangCM (2013) Phospho switch triggers Brd4 chromatin binding and activator recruitment for gene-specific targeting. Mol Cell 49: 843–857.

41. WinandyS, WuP, GeorgopoulosK (1995) A dominant mutation in the Ikaros gene leads to rapid development of leukemia and lymphoma. Cell 83: 289–299.

42. NichogiannopoulouA, TrevisanM, NebenS, FriedrichC, GeorgopoulosK (1999) Defects in hemopoietic stem cell activity in Ikaros mutant mice. J Exp Med 190: 1201–1214.

43. WeissMJ, YuC, OrkinSH (1997) Erythroid-cell-specific properties of transcription factor GATA-1 revealed by phenotypic rescue of a gene-targeted cell line. Mol Cell Biol 17: 1642–1651.

44. JingH, VakocCR, YingL, MandatS, WangH, et al. (2008) Exchange of GATA factors mediates transitions in looped chromatin organization at a developmentally regulated gene locus. Mol Cell 29: 232–242.

45. NovershternN, SubramanianA, LawtonLN, MakRH, HainingWN, et al. (2011) Densely interconnected transcriptional circuits control cell states in human hematopoiesis. Cell 144: 296–309.

46. RahlPB, LinCY, SeilaAC, FlynnRA, McCuineS, et al. (2010) c-Myc regulates transcriptional pause release. Cell 141: 432–445.

47. ReppasNB, WadeJT, ChurchGM, StruhlK (2006) The transition between transcriptional initiation and elongation in E. coli is highly variable and often rate limiting. Mol Cell 24: 747–757.

48. DengC, LiY, LiangS, CuiK, SalzT, et al. (2013) USF1 and hSET1A mediated epigenetic modifications regulate lineage differentiation and HoxB4 transcription. PLoS Genet 9: e1003524.

49. ChaoSH, FujinagaK, MarionJE, TaubeR, SausvilleEA, et al. (2000) Flavopiridol inhibits P-TEFb and blocks HIV-1 replication. J Biol Chem 275: 28345–28348.

50. SambolEB, AmbrosiniG, GehaRC, KennealeyPT, DecarolisP, et al. (2006) Flavopiridol targets c-KIT transcription and induces apoptosis in gastrointestinal stromal tumor cells. Cancer Res 66: 5858–5866.

51. YoshidaT, NgSY, Zuniga-PfluckerJC, GeorgopoulosK (2006) Early hematopoietic lineage restrictions directed by Ikaros. Nat Immunol 7: 382–391.

52. OrphanidesG, LeRoyG, ChangCH, LuseDS, ReinbergD (1998) FACT, a factor that facilitates transcript elongation through nucleosomes. Cell 92: 105–116.

53. KimT, XuZ, Clauder-MunsterS, SteinmetzLM, BuratowskiS (2012) Set3 HDAC mediates effects of overlapping noncoding transcription on gene induction kinetics. Cell 150: 1158–1169.

54. LiB, GogolM, CareyM, LeeD, SeidelC, et al. (2007) Combined action of PHD and chromo domains directs the Rpd3S HDAC to transcribed chromatin. Science 316: 1050–1054.

55. HuG, WadePA (2012) NuRD and pluripotency: a complex balancing act. Cell Stem Cell 10: 497–503.

56. MullighanCG, GoorhaS, RadtkeI, MillerCB, Coustan-SmithE, et al. (2007) Genome-wide analysis of genetic alterations in acute lymphoblastic leukaemia. Nature 446: 758–764.

57. MullighanCG, MillerCB, RadtkeI, PhillipsLA, DaltonJ, et al. (2008) BCR-ABL1 lymphoblastic leukaemia is characterized by the deletion of Ikaros. Nature 453: 110–114.

58. MullighanCG, SuX, ZhangJ, RadtkeI, PhillipsLA, et al. (2009) Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia. N Engl J Med 360: 470–480.

59. AnguitaE, HughesJ, HeyworthC, BlobelGA, WoodWG, et al. (2004) Globin gene activation during haemopoiesis is driven by protein complexes nucleated by GATA-1 and GATA-2. Embo J 23: 2841–2852.

60. BottardiS, RossJ, Pierre-CharlesN, BlankV, MilotE (2006) Lineage-specific activators affect beta-globin locus chromatin in multipotent hematopoietic progenitors. Embo J 25: 3586–3595.

61. CaloE, WysockaJ (2013) Modification of enhancer chromatin: what, how, and why? Mol Cell 49: 825–837.

62. JimenezG, GriffithsSD, FordAM, GreavesMF, EnverT (1992) Activation of the beta-globin locus control region precedes commitment to the erythroid lineage. Proc Natl Acad Sci U S A 89: 10618–10622.

63. 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.

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

65. YeM, IwasakiH, LaiosaCV, StadtfeldM, XieH, et al. (2003) Hematopoietic stem cells expressing the myeloid lysozyme gene retain long-term, multilineage repopulation potential. Immunity 19: 689–699.

66. LarochelleS, AmatR, Glover-CutterK, SansoM, ZhangC, et al. (2012) Cyclin-dependent kinase control of the initiation-to-elongation switch of RNA polymerase II. Nat Struct Mol Biol 19: 1108–1115.

67. Gomez-del ArcoP, MakiK, GeorgopoulosK (2004) Phosphorylation controls Ikaros's ability to negatively regulate the G(1)-S transition. Mol Cell Biol 24: 2797–2807.

68. GurelZ, RonniT, HoS, KucharJ, PayneKJ, et al. (2008) Recruitment of ikaros to pericentromeric heterochromatin is regulated by phosphorylation. J Biol Chem 283: 8291–8300.

69. AmmosovaT, ObukhovY, KotelkinA, BreuerD, BeullensM, et al. (2011) Protein phosphatase-1 activates CDK9 by dephosphorylating Ser175. PLoS One 6: e18985.

70. MisteliT, SpectorDL (1996) Serine/threonine phosphatase 1 modulates the subnuclear distribution of pre-mRNA splicing factors. Mol Biol Cell 7: 1559–1572.

71. ShiY, ReddyB, ManleyJL (2006) PP1/PP2A phosphatases are required for the second step of Pre-mRNA splicing and target specific snRNP proteins. Mol Cell 23: 819–829.

72. KimuraY, DingB, ImaiN, NolanDJ, ButlerJM, et al. (2011) c-Kit-mediated functional positioning of stem cells to their niches is essential for maintenance and regeneration of adult hematopoiesis. PLoS One 6: e26918.

73. MassonK, RonnstrandL (2009) Oncogenic signaling from the hematopoietic growth factor receptors c-Kit and Flt3. Cell Signal 21: 1717–1726.

74. PapathanasiouP, PerkinsAC, CobbBS, FerriniR, SridharanR, et al. (2003) Widespread failure of hematolymphoid differentiation caused by a recessive niche-filling allele of the Ikaros transcription factor. Immunity 19: 131–144.

75. ShinJY, HuW, NaramuraM, ParkCY (2014) High c-Kit expression identifies hematopoietic stem cells with impaired self-renewal and megakaryocytic bias. J Exp Med

76. NakataniY, OgryzkoV (2003) Immunoaffinity purification of mammalian protein complexes. Methods Enzymol 370: 430–444.

77. KroonE, KroslJ, ThorsteinsdottirU, BabanS, BuchbergAM, et al. (1998) Hoxa9 transforms primary bone marrow cells through specific collaboration with Meis1a but not Pbx1b. EMBO J 17: 3714–3725.

78. HahmK, ErnstP, LoK, KimGS, TurckC, et al. (1994) The lymphoid transcription factor LyF-1 is encoded by specific, alternatively spliced mRNAs derived from the Ikaros gene. Mol Cell Biol 14: 7111–7123.

79. MolnarA, GeorgopoulosK (1994) The Ikaros gene encodes a family of functionally diverse zinc finger DNA-binding proteins. Mol Cell Biol 14: 8292–8303.

80. SunL, LiuA, GeorgopoulosK (1996) Zinc finger-mediated protein interactions modulate Ikaros activity, a molecular control of lymphocyte development. Embo J 15: 5358–5369.

81. CarottaS, DakicA, D'AmicoA, PangSH, GreigKT, et al. (2010) The transcription factor PU.1 controls dendritic cell development and Flt3 cytokine receptor expression in a dose-dependent manner. Immunity 32: 628–641.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2014 Číslo 12
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
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#