#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

A Systems Approach Identifies Essential FOXO3 Functions at Key Steps of Terminal Erythropoiesis


Red blood cells (RBCs) are highly specialized cells that transport oxygen throughout the body and are essential for survival. However, RBCs have a limited lifespan and need to be replenished continuously by stem cells in the bone marrow. Mammalian RBCs are unique in that in order to fully mature they exclude their nucleus and other organelles. Mechanisms involved in these processes are not well understood at the molecular level. Defects in any of the these processes may lead to red blood cell defects, a decreased capacity to transport oxygen and/or a block in red blood cell production in vitro. Therefore, understanding how these processes are regulated at the molecular level can lead to promising new therapies for red blood cell defects and improved methods of generating red blood cells in a dish. Here, using an integrated computational and experimental biology approach, we found that the nuclear factor FOXO3 is a crucial regulator of red blood cell production by coordinating the expression of many of the genes specific for terminal maturation of red blood cells. Furthermore we found that FOXO3 can even increase the production of normal red blood cells in culture raising the possibility that enhancing FOXO3 may have a therapeutic use. Our studies identify FOXO3 as a novel regulator of RBC enucleation and terminal erythropoiesis.


Vyšlo v časopise: A Systems Approach Identifies Essential FOXO3 Functions at Key Steps of Terminal Erythropoiesis. PLoS Genet 11(10): e32767. doi:10.1371/journal.pgen.1005526
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1005526

Souhrn

Red blood cells (RBCs) are highly specialized cells that transport oxygen throughout the body and are essential for survival. However, RBCs have a limited lifespan and need to be replenished continuously by stem cells in the bone marrow. Mammalian RBCs are unique in that in order to fully mature they exclude their nucleus and other organelles. Mechanisms involved in these processes are not well understood at the molecular level. Defects in any of the these processes may lead to red blood cell defects, a decreased capacity to transport oxygen and/or a block in red blood cell production in vitro. Therefore, understanding how these processes are regulated at the molecular level can lead to promising new therapies for red blood cell defects and improved methods of generating red blood cells in a dish. Here, using an integrated computational and experimental biology approach, we found that the nuclear factor FOXO3 is a crucial regulator of red blood cell production by coordinating the expression of many of the genes specific for terminal maturation of red blood cells. Furthermore we found that FOXO3 can even increase the production of normal red blood cells in culture raising the possibility that enhancing FOXO3 may have a therapeutic use. Our studies identify FOXO3 as a novel regulator of RBC enucleation and terminal erythropoiesis.


Zdroje

1. Lapillonne H, Kobari L, Mazurier C, Tropel P, Giarratana MC, et al. (2010) Red blood cell generation from human induced pluripotent stem cells: perspectives for transfusion medicine. Haematologica 95: 1651–1659. doi: 10.3324/haematol.2010.023556 20494935

2. Ji P, Murata-Hori M, Lodish HF (2011) Formation of mammalian erythrocytes: chromatin condensation and enucleation. Trends Cell Biol 21: 409–415. doi: 10.1016/j.tcb.2011.04.003 21592797

3. Kerenyi MA, Orkin SH (2010) Networking erythropoiesis. J Exp Med 207: 2537–2541. doi: 10.1084/jem.20102260 21098097

4. Kang YA, Sanalkumar R, O'Geen H, Linnemann AK, Chang CJ, et al. (2012) Autophagy driven by a master regulator of hematopoiesis. Mol Cell Biol 32: 226–239. doi: 10.1128/MCB.06166-11 22025678

5. McIver SC, Kang YA, DeVilbiss AW, O'Driscoll CA, Ouellette JN, et al. (2014) The exosome complex establishes a barricade to erythroid maturation. Blood 124: 2285–2297. doi: 10.1182/blood-2014-04-571083 25115889

6. Li L, Freudenberg J, Cui K, Dale R, Song SH, et al. (2013) Ldb1-nucleated transcription complexes function as primary mediators of global erythroid gene activation. Blood 121: 4575–4585. doi: 10.1182/blood-2013-01-479451 23610375

7. Eijkelenboom A, Burgering BM (2013) FOXOs: signalling integrators for homeostasis maintenance. Nat Rev Mol Cell Biol 14: 83–97. doi: 10.1038/nrm3507 23325358

8. Pourfarzad F, von Lindern M, Azarkeivan A, Hou J, Kia SK, et al. (2013) Hydroxyurea responsiveness in beta-thalassemic patients is determined by the stress response adaptation of erythroid progenitors and their differentiation propensity. Haematologica 98: 696–704. doi: 10.3324/haematol.2012.074492 23100274

9. Franco SS, De Falco L, Ghaffari S, Brugnara C, Sinclair DA, et al. (2014) Resveratrol accelerates erythroid maturation by activation of FoxO3 and ameliorates anemia in beta-thalassemic mice. Haematologica 99: 267–275. doi: 10.3324/haematol.2013.090076 23975182

10. Sheehan VA, Crosby JR, Sabo A, Howard TA, Muzny DM, et al. (2013) FOXO3 Variants Are Associated With Lower Fetal Hemoglobin Levels In Children With Sickle Cell Disease Blooda 122.

11. Salih DA, Brunet A (2008) FoxO transcription factors in the maintenance of cellular homeostasis during aging. Curr Opin Cell Biol 20: 126–136. doi: 10.1016/j.ceb.2008.02.005 18394876

12. Miyamoto K, Araki KY, Naka K, Arai F, Takubo K, et al. (2007) Foxo3a is essential for maintenance of the hematopoietic stem cell pool. Cell Stem Cell 1: 101–112. doi: 10.1016/j.stem.2007.02.001 18371339

13. Tothova Z, Gilliland DG (2007) FoxO transcription factors and stem cell homeostasis: insights from the hematopoietic system. Cell Stem Cell 1: 140–152. doi: 10.1016/j.stem.2007.07.017 18371346

14. Yalcin S, Zhang X, Luciano JP, Mungamuri SK, Marinkovic D, et al. (2008) Foxo3 Is Essential for the Regulation of Ataxia Telangiectasia Mutated and Oxidative Stress-mediated Homeostasis of Hematopoietic Stem Cells. J Biol Chem 283: 25692–25705. doi: 10.1074/jbc.M800517200 18424439

15. Paik JH, Ding Z, Narurkar R, Ramkissoon S, Muller F, et al. (2009) FoxOs cooperatively regulate diverse pathways governing neural stem cell homeostasis. Cell Stem Cell 5: 540–553. doi: 10.1016/j.stem.2009.09.013 19896444

16. Renault VM, Rafalski VA, Morgan AA, Salih DA, Brett JO, et al. (2009) FoxO3 regulates neural stem cell homeostasis. Cell Stem Cell 5: 527–539. doi: 10.1016/j.stem.2009.09.014 19896443

17. Zhang X, Yalcin S, Lee DF, Yeh TY, Lee SM, et al. (2011) FOXO1 is an essential regulator of pluripotency in human embryonic stem cells. Nat Cell Biol 13: 1092–1099. doi: 10.1038/ncb2293 21804543

18. Rimmele P, Bigarella CL, Liang R, Izac B, Dieguez-Gonzalez R, et al. (2014) Aging-like Phenotype and Defective Lineage Specification in SIRT1-Deleted Hematopoietic Stem and Progenitor Cells. Stem Cell Reports 3: 44–59. doi: 10.1016/j.stemcr.2014.04.015 25068121

19. Giannakou ME, Goss M, Junger MA, Hafen E, Leevers SJ, et al. (2004) Long-lived Drosophila with overexpressed dFOXO in adult fat body. Science 305: 361. 15192154

20. Partridge L, Bruning JC (2008) Forkhead transcription factors and ageing. Oncogene 27: 2351–2363. doi: 10.1038/onc.2008.28 18391977

21. Castrillon DH, Miao L, Kollipara R, Horner JW, DePinho RA (2003) Suppression of ovarian follicle activation in mice by the transcription factor Foxo3a. Science 301: 215–218. 12855809

22. Sykes SM, Lane SW, Bullinger L, Kalaitzidis D, Yusuf R, et al. (2011) AKT/FOXO Signaling Enforces Reversible Differentiation Blockade in Myeloid Leukemias. Cell 146: 697–708. doi: 10.1016/j.cell.2011.07.032 21884932

23. Naka K, Hoshii T, Muraguchi T, Tadokoro Y, Ooshio T, et al. (2010) TGF-beta-FOXO signalling maintains leukaemia-initiating cells in chronic myeloid leukaemia. Nature 463: 676–680. doi: 10.1038/nature08734 20130650

24. Willcox BJ, Donlon TA, He Q, Chen R, Grove JS, et al. (2008) FOXO3A genotype is strongly associated with human longevity. Proc Natl Acad Sci U S A 105: 13987–13992. doi: 10.1073/pnas.0801030105 18765803

25. Demontis F, Perrimon N (2010) FOXO/4E-BP signaling in Drosophila muscles regulates organism-wide proteostasis during aging. Cell 143: 813–825. doi: 10.1016/j.cell.2010.10.007 21111239

26. Zhang P, Judy M, Lee SJ, Kenyon C (2013) Direct and indirect gene regulation by a life-extending FOXO protein in C. elegans: roles for GATA factors and lipid gene regulators. Cell Metab 17: 85–100. doi: 10.1016/j.cmet.2012.12.013 23312285

27. Lee JC, Espeli M, Anderson CA, Linterman MA, Pocock JM, et al. (2013) Human SNP links differential outcomes in inflammatory and infectious disease to a FOXO3-regulated pathway. Cell 155: 57–69. doi: 10.1016/j.cell.2013.08.034 24035192

28. Bakker WJ, Blazquez-Domingo M, Kolbus A, Besooyen J, Steinlein P, et al. (2004) FoxO3a regulates erythroid differentiation and induces BTG1, an activator of protein arginine methyl transferase 1. J Cell Biol 164: 175–184. 14734530

29. Marinkovic D, Zhang X, Yalcin S, Luciano JP, Brugnara C, et al. (2007) Foxo3 is required for the regulation of oxidative stress in erythropoiesis. J Clin Invest 117: 2133–2144. 17671650

30. Yu D, dos Santos CO, Zhao G, Jiang J, Amigo JD, et al. (2010) miR–451 protects against erythroid oxidant stress by repressing 14-3-3zeta. Genes Dev 24: 1620–1633. doi: 10.1101/gad.1942110 20679398

31. Zhang X, Camprecios G, Rimmele P, Liang R, Yalcin S, et al. (2014) FOXO3-mTOR metabolic cooperation in the regulation of erythroid cell maturation and homeostasis. Am J Hematol 89: 954–963. doi: 10.1002/ajh.23786 24966026

32. Wong P, Hattangadi SM, Cheng AW, Frampton GM, Young RA, et al. (2011) Gene induction and repression during terminal erythropoiesis are mediated by distinct epigenetic changes. Blood 118: e128–138. doi: 10.1182/blood-2011-03-341404 21860024

33. An X, Schulz VP, Li J, Wu K, Liu J, et al. (2014) Global transcriptome analyses of human and murine terminal erythroid differentiation. Blood 123: 3466–3477. doi: 10.1182/blood-2014-01-548305 24637361

34. Chen K, Liu J, Heck S, Chasis JA, An X, et al. (2009) Resolving the distinct stages in erythroid differentiation based on dynamic changes in membrane protein expression during erythropoiesis. Proc Natl Acad Sci U S A 106: 17413–17418. doi: 10.1073/pnas.0909296106 19805084

35. Chen EY, Tan CM, Kou Y, Duan Q, Wang Z, et al. (2013) Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics 14: 128. doi: 10.1186/1471-2105-14-128 23586463

36. Lachmann A, Xu H, Krishnan J, Berger SI, Mazloom AR, et al. (2010) ChEA: transcription factor regulation inferred from integrating genome-wide ChIP-X experiments. Bioinformatics 26: 2438–2444. doi: 10.1093/bioinformatics/btq466 20709693

37. Zhang J, Randall MS, Loyd MR, Dorsey FC, Kundu M, et al. (2009) Mitochondrial clearance is regulated by Atg7-dependent and -independent mechanisms during reticulocyte maturation. Blood 114: 157–164. doi: 10.1182/blood-2008-04-151639 19417210

38. Peslak SA, Wenger J, Bemis JC, Kingsley PD, Koniski AD, et al. (2012) EPO-mediated expansion of late-stage erythroid progenitors in the bone marrow initiates recovery from sublethal radiation stress. Blood 120: 2501–2511. 22889760

39. Pereira R, Quang CT, Lesault I, Dolznig H, Beug H, et al. (1999) FLI–1 inhibits differentiation and induces proliferation of primary erythroblasts. Oncogene 18: 1597–1608. 10102630

40. Rekhtman N, Radparvar F, Evans T, Skoultchi AI (1999) Direct interaction of hematopoietic transcription factors PU.1 and GATA–1: functional antagonism in erythroid cells. Genes Dev 13: 1398–1411. 10364157

41. Stadhouders R, Thongjuea S, Andrieu-Soler C, Palstra RJ, Bryne JC, et al. (2012) Dynamic long-range chromatin interactions control Myb proto-oncogene transcription during erythroid development. EMBO J 31: 986–999. doi: 10.1038/emboj.2011.450 22157820

42. Morera D, Roher N, Ribas L, Balasch JC, Donate C, et al. (2011) RNA-Seq reveals an integrated immune response in nucleated erythrocytes. PLoS One 6: e26998. doi: 10.1371/journal.pone.0026998 22046430

43. Greenfest-Allen E, Malik J, Palis J, Stoeckert CJ Jr. (2013) Stat and interferon genes identified by network analysis differentially regulate primitive and definitive erythropoiesis. BMC Syst Biol 7: 38. doi: 10.1186/1752-0509-7-38 23675896

44. Litvak V, Ratushny AV, Lampano AE, Schmitz F, Huang AC, et al. (2012) A FOXO3-IRF7 gene regulatory circuit limits inflammatory sequelae of antiviral responses. Nature 490: 421–425. doi: 10.1038/nature11428 22982991

45. Yang Z, Klionsky DJ (2010) Mammalian autophagy: core molecular machinery and signaling regulation. Curr Opin Cell Biol 22: 124–131. doi: 10.1016/j.ceb.2009.11.014 20034776

46. Schweers RL, Zhang J, Randall MS, Loyd MR, Li W, et al. (2007) NIX is required for programmed mitochondrial clearance during reticulocyte maturation. Proc Natl Acad Sci U S A 104: 19500–19505. 18048346

47. Kundu M, Lindsten T, Yang CY, Wu J, Zhao F, et al. (2008) Ulk1 plays a critical role in the autophagic clearance of mitochondria and ribosomes during reticulocyte maturation. Blood 112: 1493–1502. doi: 10.1182/blood-2008-02-137398 18539900

48. Sandoval H, Thiagarajan P, Dasgupta SK, Schumacher A, Prchal JT, et al. (2008) Essential role for Nix in autophagic maturation of erythroid cells. Nature 454: 232–235. doi: 10.1038/nature07006 18454133

49. Bakker WJ, van Dijk TB, Parren-van Amelsvoort M, Kolbus A, Yamamoto K, et al. (2007) Differential regulation of Foxo3a target genes in erythropoiesis. Mol Cell Biol 27: 3839–3854. 17353275

50. Mammucari C, Milan G, Romanello V, Masiero E, Rudolf R, et al. (2007) FoxO3 controls autophagy in skeletal muscle in vivo. Cell Metab 6: 458–471. 18054315

51. Xiong X, Tao R, DePinho RA, Dong XC (2012) The autophagy-related gene 14 (Atg14) is regulated by forkhead box O transcription factors and circadian rhythms and plays a critical role in hepatic autophagy and lipid metabolism. J Biol Chem 287: 39107–39114. doi: 10.1074/jbc.M112.412569 22992773

52. Klionsky DJ (2012) A human autophagy interaction network. Autophagy 8: 439–441. doi: 10.4161/auto.19926 22781101

53. Mizushima N, Yoshimori T (2007) How to interpret LC3 immunoblotting. Autophagy 3: 542–545. 17611390

54. Oeste CL, Seco E, Patton WF, Boya P, Perez-Sala D (2013) Interactions between autophagic and endo-lysosomal markers in endothelial cells. Histochem Cell Biol 139: 659–670. doi: 10.1007/s00418-012-1057-6 23203316

55. Honda S, Arakawa S, Nishida Y, Yamaguchi H, Ishii E, et al. (2014) Ulk1-mediated Atg5-independent macroautophagy mediates elimination of mitochondria from embryonic reticulocytes. Nat Commun 5: 4004. doi: 10.1038/ncomms5004 24895007

56. Nishida Y, Arakawa S, Fujitani K, Yamaguchi H, Mizuta T, et al. (2009) Discovery of Atg5/Atg7-independent alternative macroautophagy. Nature 461: 654–658. doi: 10.1038/nature08455 19794493

57. Mortensen M, Ferguson DJ, Simon AK (2010) Mitochondrial clearance by autophagy in developing erythrocytes: clearly important, but just how much so? Cell Cycle 9: 1901–1906. 20495377

58. Konstantinidis DG, Pushkaran S, Johnson JF, Cancelas JA, Manganaris S, et al. (2012) Signaling and cytoskeletal requirements in erythroblast enucleation. Blood 119: 6118–6127. doi: 10.1182/blood-2011-09-379263 22461493

59. Keerthivasan G, Wickrema A, Crispino JD (2011) Erythroblast enucleation. Stem Cells Int 2011: 139851. doi: 10.4061/2011/139851 22007239

60. Zhang L, Flygare J, Wong P, Lim B, Lodish HF (2011) miR–191 regulates mouse erythroblast enucleation by down-regulating Riok3 and Mxi1. Genes Dev 25: 119–124. doi: 10.1101/gad.1998711 21196494

61. Ji P, Jayapal SR, Lodish HF (2008) Enucleation of cultured mouse fetal erythroblasts requires Rac GTPases and mDia2. Nat Cell Biol 10: 314–321. doi: 10.1038/ncb1693 18264091

62. Ji P, Lodish HF (2010) Rac GTPases play multiple roles in erythropoiesis. Haematologica 95: 2–4. doi: 10.3324/haematol.2009.015511 20065075

63. Thom CS, Traxler EA, Khandros E, Nickas JM, Zhou OY, et al. (2014) Trim58 Degrades Dynein and Regulates Terminal Erythropoiesis. Dev Cell.

64. McGrath KE, Kingsley PD, Koniski AD, Porter RL, Bushnell TP, et al. (2008) Enucleation of primitive erythroid cells generates a transient population of "pyrenocytes" in the mammalian fetus. Blood 111: 2409–2417. 18032705

65. Sui Z, Nowak RB, Bacconi A, Kim NE, Liu H, et al. (2014) Tropomodulin3-null mice are embryonic lethal with anemia due to impaired erythroid terminal differentiation in the fetal liver. Blood 123: 758–767. doi: 10.1182/blood-2013-03-492710 24159174

66. Goehring NW, Grill SW (2013) Cell polarity: mechanochemical patterning. Trends Cell Biol 23: 72–80. doi: 10.1016/j.tcb.2012.10.009 23182746

67. Wang L, Yang L, Filippi MD, Williams DA, Zheng Y (2006) Genetic deletion of Cdc42GAP reveals a role of Cdc42 in erythropoiesis and hematopoietic stem/progenitor cell survival, adhesion, and engraftment. Blood 107: 98–105. 16174757

68. England SJ, McGrath KE, Frame JM, Palis J (2011) Immature erythroblasts with extensive ex vivo self-renewal capacity emerge from the early mammalian fetus. Blood 117: 2708–2717. doi: 10.1182/blood-2010-07-299743 21127173

69. Hattangadi SM, Martinez-Morilla S, Patterson HC, Shi J, Burke K, et al. (2014) Histones to the cytosol: Exportin 7 is essential for normal terminal erythroid nuclear maturation. Blood.

70. Ghaffari S, Jagani Z, Kitidis C, Lodish HF, Khosravi-Far R (2003) Cytokines and BCR-ABL mediate suppression of TRAIL-induced apoptosis through inhibition of forkhead FOXO3a transcription factor. Proc Natl Acad Sci U S A 100: 6523–6528. 12750477

71. Ghaffari S, Kitidis C, Zhao W, Marinkovic D, Fleming MD, et al. (2006) AKT induces erythroid-cell maturation of JAK2-deficient fetal liver progenitor cells and is required for Epo regulation of erythroid-cell differentiation. Blood 107: 1888–1891. 16254141

72. Zhao W, Kitidis C, Fleming MD, Lodish HF, Ghaffari S (2006) Erythropoietin stimulates phosphorylation and activation of GATA–1 via the PI3-kinase/AKT signaling pathway. Blood 107: 907–915. 16204311

73. Wang J, Ramirez T, Ji P, Jayapal SR, Lodish HF, et al. (2012) Mammalian erythroblast enucleation requires PI3K-dependent cell polarization. J Cell Sci 125: 340–349. doi: 10.1242/jcs.088286 22331356

74. Sandri M, Sandri C, Gilbert A, Skurk C, Calabria E, et al. (2004) Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin–1 and cause skeletal muscle atrophy. Cell 117: 399–412. 15109499

75. Zhao J, Brault JJ, Schild A, Cao P, Sandri M, et al. (2007) FoxO3 coordinately activates protein degradation by the autophagic/lysosomal and proteasomal pathways in atrophying muscle cells. Cell Metab 6: 472–483. 18054316

76. Warr MR, Binnewies M, Flach J, Reynaud D, Garg T, et al. (2013) FOXO3A directs a protective autophagy program in haematopoietic stem cells. Nature.

77. Eijkelenboom A, Mokry M, de Wit E, Smits LM, Polderman PE, et al. (2013) Genome-wide analysis of FOXO3 mediated transcription regulation through RNA polymerase II profiling. Mol Syst Biol 9: 638. doi: 10.1038/msb.2012.74 23340844

78. Eijkelenboom A, Mokry M, Smits LM, Nieuwenhuis EE, Burgering BM (2013) FOXO3 selectively amplifies enhancer activity to establish target gene regulation. Cell Rep 5: 1664–1678. doi: 10.1016/j.celrep.2013.11.031 24360957

79. Li L, Jothi R, Cui K, Lee JY, Cohen T, et al. (2011) Nuclear adaptor Ldb1 regulates a transcriptional program essential for the maintenance of hematopoietic stem cells. Nat Immunol 12: 129–136. doi: 10.1038/ni.1978 21186366

80. Novershtern N, Subramanian A, Lawton LN, Mak RH, Haining WN, et al. (2011) Densely interconnected transcriptional circuits control cell states in human hematopoiesis. Cell 144: 296–309. doi: 10.1016/j.cell.2011.01.004 21241896

81. Xu J, Shao Z, Glass K, Bauer DE, Pinello L, et al. (2012) Combinatorial assembly of developmental stage-specific enhancers controls gene expression programs during human erythropoiesis. Dev Cell 23: 796–811. doi: 10.1016/j.devcel.2012.09.003 23041383

82. Trapnell C, Pachter L, Salzberg SL (2009) TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25: 1105–1111. doi: 10.1093/bioinformatics/btp120 19289445

83. Trapnell C, Roberts A, Goff L, Pertea G, Kim D, et al. (2012) Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat Protoc 7: 562–578. doi: 10.1038/nprot.2012.016 22383036

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2015 Číslo 10
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#