#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

HDAC7 Is a Repressor of Myeloid Genes Whose Downregulation Is Required for Transdifferentiation of Pre-B Cells into Macrophages


: B lymphopoiesis is the result of several cell-commitment, lineage-choice, and differentiation processes. Every differentiation step is characterized by the activation of a new, lineage-specific, genetic program and the extinction of the previous one. To date, the central role of specific transcription factors in positively regulating these distinct differentiation processes to acquire a B cell–specific genetic program is well established. However, the existence of specific transcriptional repressors responsible for the silencing of lineage inappropriate genes remains elusive. Here we addressed the molecular mechanism behind repression of non-lymphoid genes in B cells. We report that the histone deacetylase HDAC7 was highly expressed in pre-B cells but dramatically down-regulated during cellular lineage conversion to macrophages. Microarray analysis demonstrated that HDAC7 re-expression interfered with the acquisition of the gene transcriptional program characteristic of macrophages during cell transdifferentiation; the presence of HDAC7 blocked the induction of key genes for macrophage function, such as immune, inflammatory, and defense response, cellular response to infections, positive regulation of cytokines production, and phagocytosis. Moreover, re-introduction of HDAC7 suppressed crucial functions of macrophages, such as the ability to phagocytose bacteria and to respond to endotoxin by expressing major pro-inflammatory cytokines. To gain insight into the molecular mechanisms mediating HDAC7 repression in pre-B cells, we undertook co-immunoprecipitation and chromatin immunoprecipitation experimental approaches. We found that HDAC7 specifically interacted with the transcription factor MEF2C in pre-B cells and was recruited to MEF2 binding sites located at the promoters of genes critical for macrophage function. Thus, in B cells HDAC7 is a transcriptional repressor of undesirable genes. Our findings uncover a novel role for HDAC7 in maintaining the identity of a particular cell type by silencing lineage-inappropriate genes.


Vyšlo v časopise: HDAC7 Is a Repressor of Myeloid Genes Whose Downregulation Is Required for Transdifferentiation of Pre-B Cells into Macrophages. PLoS Genet 9(5): e32767. doi:10.1371/journal.pgen.1003503
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003503

Souhrn

: B lymphopoiesis is the result of several cell-commitment, lineage-choice, and differentiation processes. Every differentiation step is characterized by the activation of a new, lineage-specific, genetic program and the extinction of the previous one. To date, the central role of specific transcription factors in positively regulating these distinct differentiation processes to acquire a B cell–specific genetic program is well established. However, the existence of specific transcriptional repressors responsible for the silencing of lineage inappropriate genes remains elusive. Here we addressed the molecular mechanism behind repression of non-lymphoid genes in B cells. We report that the histone deacetylase HDAC7 was highly expressed in pre-B cells but dramatically down-regulated during cellular lineage conversion to macrophages. Microarray analysis demonstrated that HDAC7 re-expression interfered with the acquisition of the gene transcriptional program characteristic of macrophages during cell transdifferentiation; the presence of HDAC7 blocked the induction of key genes for macrophage function, such as immune, inflammatory, and defense response, cellular response to infections, positive regulation of cytokines production, and phagocytosis. Moreover, re-introduction of HDAC7 suppressed crucial functions of macrophages, such as the ability to phagocytose bacteria and to respond to endotoxin by expressing major pro-inflammatory cytokines. To gain insight into the molecular mechanisms mediating HDAC7 repression in pre-B cells, we undertook co-immunoprecipitation and chromatin immunoprecipitation experimental approaches. We found that HDAC7 specifically interacted with the transcription factor MEF2C in pre-B cells and was recruited to MEF2 binding sites located at the promoters of genes critical for macrophage function. Thus, in B cells HDAC7 is a transcriptional repressor of undesirable genes. Our findings uncover a novel role for HDAC7 in maintaining the identity of a particular cell type by silencing lineage-inappropriate genes.


Zdroje

1. ParraM (2009) Epigenetic events during B lymphocyte development. Epigenetics 4: 462–468.

2. MercerEM, LinYC, MurreC (2011) Factors and networks that underpin early hematopoiesis. Semin Immunol 23: 317–325.

3. AdolfssonJ, ManssonR, Buza-VidasN, HultquistA, LiubaK, et al. (2005) Identification of Flt3+ lympho-myeloid stem cells lacking erythro-megakaryocytic potential a revised road map for adult blood lineage commitment. Cell 121: 295–306.

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

5. Stehling-SunS, DadeJ, NuttSL, DeKoterRP, CamargoFD (2009) Regulation of lymphoid versus myeloid fate ‘choice’ by the transcription factor Mef2c. Nat Immunol 10: 289–296.

6. BainG, MaandagEC, IzonDJ, AmsenD, KruisbeekAM, et al. (1994) E2A proteins are required for proper B cell development and initiation of immunoglobulin gene rearrangements. Cell 79: 885–892.

7. BainG, Robanus MaandagEC, te RieleHP, FeeneyAJ, SheehyA, et al. (1997) Both E12 and E47 allow commitment to the B cell lineage. Immunity 6: 145–154.

8. ZhuangY, SorianoP, WeintraubH (1994) The helix-loop-helix gene E2A is required for B cell formation. Cell 79: 875–884.

9. LinH, GrosschedlR (1995) Failure of B-cell differentiation in mice lacking the transcription factor EBF. Nature 376: 263–267.

10. DenglerHS, BarachoGV, OmoriSA, BrucknerS, ArdenKC, et al. (2008) Distinct functions for the transcription factor Foxo1 at various stages of B cell differentiation. Nat Immunol 9: 1388–1398.

11. UrbanekP, WangZQ, FetkaI, WagnerEF, BusslingerM (1994) Complete block of early B cell differentiation and altered patterning of the posterior midbrain in mice lacking Pax5/BSAP. Cell 79: 901–912.

12. DeloguA, SchebestaA, SunQ, AschenbrennerK, PerlotT, et al. (2006) Gene repression by Pax5 in B cells is essential for blood cell homeostasis and is reversed in plasma cells. Immunity 24: 269–281.

13. SchebestaA, McManusS, SalvagiottoG, DeloguA, BusslingerGA, et al. (2007) Transcription factor Pax5 activates the chromatin of key genes involved in B cell signaling, adhesion, migration, and immune function. Immunity 27: 49–63.

14. PridansC, HolmesML, PolliM, WettenhallJM, DakicA, et al. (2008) Identification of Pax5 target genes in early B cell differentiation. J Immunol 180: 1719–1728.

15. ParraM, VerdinE (2010) Regulatory signal transduction pathways for class IIa histone deacetylases. Curr Opin Pharmacol 10: 454–460.

16. MartinM, KettmannR, DequiedtF (2009) Class IIa histone deacetylases: conducting development and differentiation. Int J Dev Biol 53: 291–301.

17. BussmannLH, SchubertA, Vu ManhTP, De AndresL, DesbordesSC, et al. (2009) A robust and highly efficient immune cell reprogramming system. Cell Stem Cell 5: 554–566.

18. Di TullioA, ManhTP, SchubertA, ManssonR, GrafT (2011) CCAAT/enhancer binding protein alpha (C/EBP(alpha))-induced transdifferentiation of pre-B cells into macrophages involves no overt retrodifferentiation. Proc Natl Acad Sci U S A 108: 17016–17021.

19. Rodriguez-UbrevaJ, CiudadL, Gomez-CabreroD, ParraM, BussmannLH, et al. (2011) Pre-B cell to macrophage transdifferentiation without significant promoter DNA methylation changes. Nucleic Acids Res

20. Perez-LlamasC, Lopez-BigasN (2011) Gitools: analysis and visualisation of genomic data using interactive heat-maps. PLoS ONE 6: e19541 doi:10.1371/journal.pone.0019541.

21. LinYC, JhunjhunwalaS, BennerC, HeinzS, WelinderE, et al. (2010) A global network of transcription factors, involving E2A, EBF1 and Foxo1, that orchestrates B cell fate. Nat Immunol 11: 635–643.

22. SwansonBJ, JackHM, LyonsGE (1998) Characterization of myocyte enhancer factor 2 (MEF2) expression in B and T cells: MEF2C is a B cell-restricted transcription factor in lymphocytes. Mol Immunol 35: 445–458.

23. WilkerPR, KohyamaM, SandauMM, AlbringJC, NakagawaO, et al. (2008) Transcription factor Mef2c is required for B cell proliferation and survival after antigen receptor stimulation. Nat Immunol 9: 603–612.

24. KhiemD, CysterJG, SchwarzJJ, BlackBL (2008) A p38 MAPK-MEF2C pathway regulates B-cell proliferation. Proc Natl Acad Sci U S A 105: 17067–17072.

25. GekasC, RhodesKE, GereigeLM, HelgadottirH, FerrariR, et al. (2009) Mef2C is a lineage-restricted target of Scl/Tal1 and regulates megakaryopoiesis and B-cell homeostasis. Blood 113: 3461–3471.

26. DequiedtF, KaslerH, FischleW, KiermerV, WeinsteinM, et al. (2003) HDAC7, a thymus-specific class II histone deacetylase, regulates Nur77 transcription and TCR-mediated apoptosis. Immunity 18: 687–698.

27. KaslerHG, VerdinE (2007) Histone deacetylase 7 functions as a key regulator of genes involved in both positive and negative selection of thymocytes. Mol Cell Biol 27: 5184–5200.

28. KaslerHG, YoungBD, MottetD, LimHW, CollinsAM, et al. (2011) Histone deacetylase 7 regulates cell survival and TCR signaling in CD4/CD8 double-positive thymocytes. J Immunol 186: 4782–4793.

29. DequiedtF, Van LintJ, LecomteE, Van DuppenV, SeufferleinT, et al. (2005) Phosphorylation of histone deacetylase 7 by protein kinase D mediates T cell receptor-induced Nur77 expression and apoptosis. J Exp Med 201: 793–804.

30. NavarroMN, GoebelJ, Feijoo-CarneroC, MorriceN, CantrellDA (2011) Phosphoproteomic analysis reveals an intrinsic pathway for the regulation of histone deacetylase 7 that controls the function of cytotoxic T lymphocytes. Nat Immunol 12: 352–361.

31. ParraM, KaslerH, McKinseyTA, OlsonEN, VerdinE (2005) Protein kinase D1 phosphorylates HDAC7 and induces its nuclear export after T-cell receptor activation. J Biol Chem 280: 13762–13770.

32. RafatiH, ParraM, HakreS, MoshkinY, VerdinE, et al. (2011) Repressive LTR nucleosome positioning by the BAF complex is required for HIV latency. PLoS Biol 9: e1001206 doi:10.1371/journal.pbio.1001206.

33. GentlemanRC, CareyVJ, BatesDM, BolstadB, DettlingM, et al. (2004) Bioconductor: open software development for computational biology and bioinformatics. Genome Biol 5: R80.

34. GautierL, CopeL, BolstadBM, IrizarryRA (2004) affy–analysis of Affymetrix GeneChip data at the probe level. Bioinformatics 20: 307–315.

35. KauffmannA, GentlemanR, HuberW (2009) arrayQualityMetrics–a bioconductor package for quality assessment of microarray data. Bioinformatics 25: 415–416.

36. SmythGK (2004) Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol 3: Article3.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 5
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#