Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Myelodysplastic syndrome

The functional mechanisms of mutations in myelodysplastic syndrome

Abstract

Overlapping spectrum of mutated genes affected in myelodysplastic syndrome (MDS) and primary acute myeloid leukemia suggest common pathogenic mechanisms. However, the frequencies of specific mutations are significantly different between them, which implies they might determine specific disease phenotype. For instance, there are overrepresentations of mutations in RNA splicing factors or epigenetic regulators in MDS. We provide an overview of recent advances in our understanding of the biology of MDS and related disorders. Our focus is how mutations of in splicing factors or epigenetic regulators identified in MDS patients demonstrate phenotypes in knockin/knockout mouse models. For instance, mutant Srsf2 mice could alter Srsf2’s normal sequence-specific RNA binding activity. It exhibited changing in the recognition of specific exonic splicing enhancer motifs to drive recurrent missplicing of Ezh2, which reduces Ezh2 expression by promoting nonsense-mediated decay. Consistent with this, SRSF2 mutations are mutually exclusive with EZH2 loss-of-function mutations in MDS patients. We also review how gene editing technology identified unique associations between pathogenic mechanisms and targeted therapy using lenalidomide, including: (i) how haploinsufficiency of the genes located in the commonly deleted region in del(5q) MDS patients promotes MDS; (ii) how lenalidomide causes selective elimination of del(5q) MDS cells; and (iii) why del(5q) MDS patients become resistant to lenalidomide. Thus, this review describes our current understanding of the mechanistic and biological effects of mutations in spliceosome and epigenetic regulators by comparing wild-type normal to mutant function as well as a brief overview of the recent progresses in MDS biology.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

References

  1. Tefferi A, Vardiman JW. Myelodysplastic syndromes. N Engl J Med. 2009;361:1872–85.

    Article  CAS  PubMed  Google Scholar 

  2. Nimer SD. Myelodysplastic syndromes. Blood. 2008;111:4841–51.

    Article  CAS  PubMed  Google Scholar 

  3. Yoshida K, Sanada M, Shiraishi Y, Nowak D, Nagata Y, Yamamoto R, et al. Frequent pathway mutations of splicing machinery in myelodysplasia. Nature. 2011;478:64–9.

    Article  CAS  PubMed  Google Scholar 

  4. Papaemmanuil E, Cazzola M, Boultwood J, Malcovati L, Vyas P, Bowen D, et al. Somatic SF3B1 mutation in myelodysplasia with ring sideroblasts. New Engl J Med. 2011;365:1384–95.

    Article  CAS  PubMed  Google Scholar 

  5. Haferlach T, Nagata Y, Grossmann V, Okuno Y, Bacher U, Nagae G, et al. Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia. 2014;28:241–7.

    CAS  PubMed  Google Scholar 

  6. Tefferi A, Lim KH, Levine R. Mutation in TET2 in myeloid cancers. N Engl J Med. 2009;361:1117.

    Article  CAS  PubMed  Google Scholar 

  7. Nikoloski G, Langemeijer SM, Kuiper RP, Knops R, Massop M, Tönnissen ER, et al. Somatic mutations of the histone methyltransferase gene EZH2 in myelodysplastic syndromes. Nat Genet. 2010;42:665–7.

    Article  CAS  PubMed  Google Scholar 

  8. Sugimoto K, Hirano N, Toyoshima H, Chiba S, Mano H, Takaku F, et al. Mutations of the p53 gene in myelodysplastic syndrome (MDS) and MDS-derived leukemia. Blood. 1993;81:3022–6.

    Article  CAS  PubMed  Google Scholar 

  9. Sanada M, Suzuki T, Shih LY, Otsu M, Kato M, Yamazaki S, et al. Gain-of-function of mutated C-CBL tumour suppressor in myeloid neoplasms. Nature. 2009;460:904–8.

    Article  CAS  PubMed  Google Scholar 

  10. Osato M. Point mutations in the RUNX1/AML1 gene: another actor in RUNX leukemia. Oncogene. 2004;23:4284–96.

    Article  CAS  PubMed  Google Scholar 

  11. Makishima H, Yoshizato T, Yoshida K, Sekeres MA, Radivoyevitch T, Suzuki H, et al. Dynamics of clonal evolution in myelodysplastic syndromes. Nat Genet. 2017;49:204–12.

    Article  CAS  PubMed  Google Scholar 

  12. Graubert TA, Shen D, Ding L, Okeyo-Owuor T, Lunn CL, Shao J, et al. Recurrent mutations in the U2AF1 splicing factor in myelodysplastic syndromes. Nat Genet. 2011;44:53–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Delhommeau F, Dupont S, Valle VD, James C, Trannoy S, Massé A, et al. Mutation in TET2 in myeloid cancers. New Engl J Med. 2009;360:2289–301.

    Article  PubMed  Google Scholar 

  14. Ley TJ, Ding L, Walter MJ, McLellan MD, Lamprecht T, Larson DE, et al. DNMT3A mutations in acute myeloid leukemia. New Engl J Med. 2010;363:2424–33.

    Article  CAS  PubMed  Google Scholar 

  15. Mardis ER, Ding L, Dooling DJ, Larson DE, McLellan MD, Chen K, et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. N Engl J Med. 2009;361:1058–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ogawa S. Genetics of MDS. Blood. 2019;133:1049–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Wahl MC, Will CL, Lührmann R. The spliceosome: design principles of a dynamic RNP machine. Cell. 2009;136:701–18.

    Article  CAS  PubMed  Google Scholar 

  18. Wang ET, Sandberg R, Luo S, Khrebtukova I, Zhang L, Mayr C, et al. Alternative isoform regulation in human tissue transcriptomes. Nature. 2008;456:470–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Fu XD, Ares M. Context-dependent control of alternative splicing by RNA-binding proteins. Nat Rev Genet. 2014;15:689–701.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Inoue D, Bradley RK, Abdel-Wahab O. Spliceosomal gene mutations in myelodysplasia: molecular links to clonal abnormalities of hematopoiesis. Genes Dev. 2016;30:989–1001. 05

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Malcovati L, Papaemmanuil E, Bowen DT, Boultwood J, Della Porta MG, Pascutto C, et al. Clinical significance of SF3B1 mutations in myelodysplastic syndromes and myelodysplastic/myeloproliferative neoplasms. Blood. 2011;118:6239–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Dolatshad H, Pellagatti A, Fernandez-Mercado M, Yip BH, Malcovati L, Attwood M, et al. Disruption of SF3B1 results in deregulated expression and splicing of key genes and pathways in myelodysplastic syndrome hematopoietic stem and progenitor cells. Leukemia. 2015;29:1798.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Dolatshad H, Pellagatti A, Liberante FG, Llorian M, Repapi E, Steeples V, et al. Cryptic splicing events in the iron transporter ABCB7 and other key target genes in SF3B1-mutant myelodysplastic syndromes. Leukemia. 2016;30:2322–31. 12

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Alsafadi S, Houy A, Battistella A, Popova T, Wassef M, Henry E, et al. Cancer-associated SF3B1 mutations affect alternative splicing by promoting alternative branchpoint usage. Nat Commun. 2016;7:10615.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Obeng EA, Chappell RJ, Seiler M, Chen MC, Campagna DR, Schmidt PJ, et al. Physiologic expression of Sf3b1(K700E) causes impaired erythropoiesis, aberrant splicing, and sensitivity to therapeutic spliceosome modulation. Cancer Cell. 2016;30:404–17. 09

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Darman RB, Seiler M, Agrawal AA, Lim KH, Peng S, Aird D, et al. Cancer-associated SF3B1 hotspot mutations induce cryptic 3′ splice site selection through use of a different branch point. Cell Rep. 2015;13:1033–45.

    Article  CAS  PubMed  Google Scholar 

  27. DeBoever C, Ghia EM, Shepard PJ, Rassenti L, Barrett CL, Jepsen K, et al. Transcriptome sequencing reveals potential mechanism of cryptic 3′ splice site selection in SF3B1-mutated cancers. PLoS Comput Biol. 2015;11:e1004105.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Mupo A, Seiler M, Sathiaseelan V, Pance A, Yang Y, Agrawal AA, et al. Hemopoietic-specific Sf3b1-K700E knock-in mice display the splicing defect seen in human MDS but develop anemia without ring sideroblasts. Leukemia. 2017;31:720–7. 03

    Article  CAS  PubMed  Google Scholar 

  29. Inoue D, Abdel-Wahab O. Modeling SF3B1 mutations in cancer: advances, challenges, and opportunities. Cancer Cell. 2016;30:371–3. 09

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Daubner GM, Cléry A, Jayne S, Stevenin J, Allain FH. A syn-anti conformational difference allows SRSF2 to recognize guanines and cytosines equally well. EMBO J. 2012;31:162–74.

    Article  CAS  PubMed  Google Scholar 

  31. Papaemmanuil E, Gerstung M, Malcovati L, Tauro S, Gundem G, Van Loo P, et al. Clinical and biological implications of driver mutations in myelodysplastic syndromes. Blood. 2013;122:3616–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Kim E, Ilagan JO, Liang Y, Daubner GM, Lee SC, Ramakrishnan A, et al. SRSF2 mutations contribute to myelodysplasia by mutant-specific effects on exon recognition. Cancer Cell. 2015;27:617–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Kon A, Yamazaki S, Nannya Y, Kataoka K, Ota Y, Nakagawa MM, et al. Physiological Srsf2 P95H expression causes impaired hematopoietic stem cell functions and aberrant RNA splicing in mice. Blood. 2018;131:621–35. 02

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Smeets MF, Tan SY, Xu JJ, Anande G, Unnikrishnan A, Chalk AM, et al. initiates myeloid bias and myelodysplastic/myeloproliferative syndrome from hemopoietic stem cells. Blood. 2018;132:608–21.

    Article  CAS  PubMed  Google Scholar 

  35. Shirai CL, Ley JN, White BS, Kim S, Tibbitts J, Shao J, et al. Mutant U2AF1 expression alters hematopoiesis and Pre-mRNA splicing in vivo. Cancer Cell. 2015;27:631–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Fei DL, Zhen T, Durham B, Ferrarone J, Zhang T, Garrett L, et al. Impaired hematopoiesis and leukemia development in mice with a conditional knock-in allele of a mutant splicing factor gene. Proc Natl Acad Sci USA. 2018;115:E10437–46. 10

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Przychodzen B, Jerez A, Guinta K, Sekeres MA, Padgett R, Maciejewski JP, et al. Patterns of missplicing due to somatic U2AF1 mutations in myeloid neoplasms. Blood. 2013;122:999–1006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Okeyo-Owuor T, White BS, Chatrikhi R, Mohan DR, Kim S, Griffith M, et al. U2AF1 mutations alter sequence specificity of pre-mRNA binding and splicing. Leukemia. 2015;29:909–17.

    Article  CAS  PubMed  Google Scholar 

  39. Ilagan JO, Ramakrishnan A, Hayes B, Murphy ME, Zebari AS, Bradley P, et al. U2AF1 mutations alter splice site recognition in hematological malignancies. Genome Res. 2015;25:14–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Chen H, Ruiz PD, Novikov L, Casill AD, Park JW, Gamble MJ. MacroH2A1.1 and PARP-1 cooperate to regulate transcription by promoting CBP-mediated H2B acetylation. Nat Struct Mol Biol. 2014;21:981–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Novikov L, Park JW, Chen H, Klerman H, Jalloh AS, Gamble MJ. QKI-mediated alternative splicing of the histone variant MacroH2A1 regulates cancer cell proliferation. Mol Cell Biol. 2011;31:4244–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Zhang J, Lieu YK, Ali AM, Penson A, Reggio KS, Rabadan R, et al. Disease-associated mutation in SRSF2 misregulates splicing by altering RNA-binding affinities. Proc Natl Acad Sci USA. 2015;112:E4726–34.

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Shen H, Zheng X, Luecke S, Green MR. The U2AF35-related protein Urp contacts the 3′ splice site to promote U12-type intron splicing and the second step of U2-type intron splicing. Genes Dev. 2010;24:2389–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Niemelä EH, Frilander MJ. Regulation of gene expression through inefficient splicing of U12-type introns. RNA Biol. 2014;11:1325–9.

    Article  PubMed  Google Scholar 

  45. Alioto TS. U12DB: a database of orthologous U12-type spliceosomal introns. Nucleic Acids Res. 2007;35(Jan):D110–5.

    Article  CAS  PubMed  Google Scholar 

  46. Madan V, Kanojia D, Li J, Okamoto R, Sato-Otsubo A, Kohlmann A, et al. Aberrant splicing of U12-type introns is the hallmark of ZRSR2 mutant myelodysplastic syndrome. Nat Commun. 2015;6:6042.

    Article  CAS  PubMed  Google Scholar 

  47. Ehrlich M. DNA hypomethylation in cancer cells. Epigenomics. 2009;1:239–59.

    Article  CAS  PubMed  Google Scholar 

  48. Silverman LR, Demakos EP, Peterson BL, Kornblith AB, Holland JC, Odchimar-Reissig R, et al. Randomized controlled trial of azacitidine in patients with the myelodysplastic syndrome: a study of the cancer and leukemia group B. J Clin Oncol. 2002;20:2429–40.

    Article  CAS  PubMed  Google Scholar 

  49. Navada SC, Steinmann J, Lübbert M, Silverman LR. Clinical development of demethylating agents in hematology. J Clin Invest. 2014;124:40–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Santini V. How I treat MDS after hypomethylating agent failure. Blood. 2019;133:521–9.

    Article  CAS  PubMed  Google Scholar 

  51. Herceg Z, Hainaut P. Genetic and epigenetic alterations as biomarkers for cancer detection, diagnosis and prognosis. Mol Oncol. 2007;1:26–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Schuettengruber B, Chourrout D, Vervoort M, Leblanc B, Cavalli G. Genome regulation by polycomb and trithorax proteins. Cell. 2007;128:735–45.

    Article  CAS  PubMed  Google Scholar 

  53. Comet I, Riising EM, Leblanc B, Helin K. Maintaining cell identity: PRC2-mediated regulation of transcription and cancer. Nat Rev Cancer. 2016;16:803–10. 12

    Article  CAS  PubMed  Google Scholar 

  54. Wang H, Wang L, Erdjument-Bromage H, Vidal M, Tempst P, Jones RS, et al. Role of histone H2A ubiquitination in Polycomb silencing. Nature. 2004;431:873–8.

    Article  CAS  PubMed  Google Scholar 

  55. Laugesen A, Højfeldt JW, Helin K. Molecular Mechanisms Directing PRC2 Recruitment and H3K27 Methylation. Mol Cell. 2019;74:8–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Cao R, Wang L, Wang H, Xia L, Erdjument-Bromage H, Tempst P, et al. Role of histone H3 lysine 27 methylation in polycomb-group silencing. Science. 2002;298:1039–43.

    Article  CAS  PubMed  Google Scholar 

  57. Boyer LA, Plath K, Zeitlinger J, Brambrink T, Medeiros LA, Lee TI, et al. Polycomb complexes repress developmental regulators in murine embryonic stem cells. Nature. 2006;441:349–53.

    Article  CAS  PubMed  Google Scholar 

  58. Francis NJ, Kingston RE, Woodcock CL. Chromatin compaction by a polycomb group protein complex. Science. 2004;306:1574–7.

    Article  CAS  PubMed  Google Scholar 

  59. Tavares L, Dimitrova E, Oxley D, Webster J, Poot R, Demmers J, et al. RYBP-PRC1 complexes mediate H2A ubiquitylation at polycomb target sites independently of PRC2 and H3K27me3. Cell. 2012;148:664–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Simon JA, Kingston RE. Occupying chromatin: Polycomb mechanisms for getting to genomic targets, stopping transcriptional traffic, and staying put. Mol Cell. 2013;49:808–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Aoyama K, Oshima M, Koide S, Suzuki E, Mochizuki-Kashio M, Kato Y, et al. Ezh1 targets bivalent genes to maintain self-renewing stem cells in Ezh2-insufficient myelodysplastic syndrome. iScience. 2018;9:161–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Bejar R, Stevenson K, Abdel-Wahab O, Galili N, Nilsson B, Garcia-Manero G, et al. Clinical effect of point mutations in myelodysplastic syndromes. New Engl J Med. 2011;364:2496–506.

    Article  CAS  PubMed  Google Scholar 

  63. Sashida G, Harada H, Matsui H, Oshima M, Yui M, Harada Y, et al. Ezh2 loss promotes development of myelodysplastic syndrome but attenuates its predisposition to leukaemic transformation. Nat Commun. 2014;5:4177.

    Article  CAS  PubMed  Google Scholar 

  64. Yap DB, Chu J, Berg T, Schapira M, Cheng SW, Moradian A, et al. Somatic mutations at EZH2 Y641 act dominantly through a mechanism of selectively altered PRC2 catalytic activity, to increase H3K27 trimethylation. Blood. 2011;117:2451–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. McCabe MT, Ott HM, Ganji G, Korenchuk S, Thompson C, Van Aller GS, et al. EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations. Nature. 2012;492:108–12.

    Article  CAS  PubMed  Google Scholar 

  66. Muto T, Sashida G, Oshima M, Wendt GR, Mochizuki-Kashio M, Nagata Y, et al. Concurrent loss of Ezh2 and Tet2 cooperates in the pathogenesis of myelodysplastic disorders. J Exp Med. 2013;210:2627–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Sashida G, Oshima M, Iwama A Deregulated polycomb functions in myeloproliferative neoplasms. Int J Hematol. 2019;110:170–8.

    Article  CAS  PubMed  Google Scholar 

  68. Mochizuki-Kashio M, Aoyama K, Sashida G, Oshima M, Tomioka T, Muto T, et al. Ezh2 loss in hematopoietic stem cells predisposes mice to develop heterogeneous malignancies in an Ezh1-dependent manner. Blood. 2015;126:1172–83.

    Article  CAS  PubMed  Google Scholar 

  69. Hasegawa N, Oshima M, Sashida G, Matsui H, Koide S, Saraya A, et al. Impact of combinatorial dysfunctions of Tet2 and Ezh2 on the epigenome in the pathogenesis of myelodysplastic syndrome. Leukemia. 2017;31:861–71. 04

    Article  CAS  PubMed  Google Scholar 

  70. Inoue D, Kitaura J, Togami K, Nishimura K, Enomoto Y, Uchida T, et al. Myelodysplastic syndromes are induced by histone methylation–altering ASXL1 mutations. J Clin Invest. 2013;123:4627–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Park UH, Yoon SK, Park T, Kim EJ, Um SJ. Additional sex comb-like (ASXL) proteins 1 and 2 play opposite roles in adipogenesis via reciprocal regulation of peroxisome proliferator-activated receptor {gamma}. J Biol Chem. 2011;286:1354–63.

    Article  CAS  PubMed  Google Scholar 

  72. Asada S, Kitamura T Aberrant histone modifications induced by mutant ASXL1 in myeloid neoplasms. Int J Hematol. 2019;110:179–86.

    Article  PubMed  CAS  Google Scholar 

  73. Inoue D, Fujino T, Kitamura T. ASXL1 as a critical regulator of epigenetic marks and therapeutic potential of mutated cells. Oncotarget. 2018;9:35203–4.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Jaiswal S, Fontanillas P, Flannick J, Manning A, Grauman PV, Mar BG, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med. 2014;371:2488–98.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. Abdel-Wahab O, Adli M, LaFave LM, Gao J, Hricik T, Shih AH, et al. ASXL1 mutations promote myeloid transformation through loss of PRC2-mediated gene repression. Cancer Cell. 2012;22:180–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Nagase R, Inoue D, Pastore A, Fujino T, Hou HA, Yamasaki N, et al. Expression of mutant Asxl1 perturbs hematopoiesis and promotes susceptibility to leukemic transformation. J Exp Med. 2018;215:1729–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Hsu YC, Chiu YC, Lin CC, Kuo YY, Hou HA, Tzeng YS, et al. The distinct biological implications of Asxl1 mutation and its roles in leukemogenesis revealed by a knock-in mouse model. J Hematol Oncol. 2017;10:139. 07

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Uni M, Masamoto Y, Sato T, Kamikubo Y, Arai S, Hara E, et al. Modeling ASXL1 mutation revealed impaired hematopoiesis caused by derepression of p16Ink4a through aberrant PRC1-mediated histone modification. Leukemia. 2019;33:191–204.

    Article  CAS  PubMed  Google Scholar 

  79. Asada S, Goyama S, Inoue D, Shikata S, Takeda R, Fukushima T, et al. Mutant ASXL1 cooperates with BAP1 to promote myeloid leukaemogenesis. Nat Commun. 2018;9:2733. 07

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  80. Saxonov S, Berg P, Brutlag DL. A genome-wide analysis of CpG dinucleotides in the human genome distinguishes two distinct classes of promoters. Proc Natl Acad Sci USA. 2006;103:1412–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Herman JG, Baylin SB. Gene silencing in cancer in association with promoter hypermethylation. N Engl J Med. 2003;349:2042–54.

    Article  CAS  PubMed  Google Scholar 

  82. Ko M, Huang Y, Jankowska AM, Pape UJ, Tahiliani M, Bandukwala HS, et al. Impaired hydroxylation of 5-methylcytosine in myeloid cancers with mutant TET2. Nature. 2010;468:839–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Tsukada Y, Fang J, Erdjument-Bromage H, Warren ME, Borchers CH, Tempst P, et al. Histone demethylation by a family of JmjC domain-containing proteins. Nature. 2006;439:811–6.

    Article  CAS  PubMed  Google Scholar 

  84. Yamane K, Toumazou C, Tsukada Y, Erdjument-Bromage H, Tempst P, Wong J, et al. JHDM2A, a JmjC-containing H3K9 demethylase, facilitates transcription activation by androgen receptor. Cell. 2006;125:483–95.

    Article  CAS  PubMed  Google Scholar 

  85. Guryanova OA, Shank K, Spitzer B, Luciani L, Koche RP, Garrett-Bakelman FE, et al. DNMT3A mutations promote anthracycline resistance in acute myeloid leukemia via impaired nucleosome remodeling. Nat Med. 2016;22:1488–95. 12

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Chiba S. Dysregulation of TET2 in hematologic malignancies. Int J Hematol. 2017;105:17–22.

    Article  CAS  PubMed  Google Scholar 

  87. Li Z, Cai X, Cai CL, Wang J, Zhang W, Petersen BE, et al. Deletion of Tet2 in mice leads to dysregulated hematopoietic stem cells and subsequent development of myeloid malignancies. Blood. 2011;118:4509–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Kats LM, Reschke M, Taulli R, Pozdnyakova O, Burgess K, Bhargava P, et al. Proto-oncogenic role of mutant IDH2 in leukemia initiation and maintenance. Cell Stem Cell. 2014;14:329–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. List A, Ebert BL, Fenaux P. A decade of progress in myelodysplastic syndrome with chromosome 5q deletion. Leukemia. 2018;32:1493–9. 07

    Article  PubMed  Google Scholar 

  90. Van den Berghe H, Cassiman JJ, David G, Fryns JP, Michaux JL, Sokal G. Distinct haematological disorder with deletion of long arm of no. 5 chromosome. Nature. 1974;251:437–8.

    Article  PubMed  Google Scholar 

  91. List A, Dewald G, Bennett J, Giagounidis A, Raza A, Feldman E, et al. Lenalidomide in the myelodysplastic syndrome with chromosome 5q deletion. New Engl J Med. 2006;355:1456–65.

    Article  CAS  PubMed  Google Scholar 

  92. Ribezzo F, Snoeren IAM, Ziegler S, Stoelben J, Olofsen PA, Henic A, et al. Rps14, Csnk1a1 and miRNA145/miRNA146a deficiency cooperate in the clinical phenotype and activation of the innate immune system in the 5q- syndrome. Leukemia 2019;33:1759–72.

    Article  CAS  PubMed  Google Scholar 

  93. Schneider RK, Schenone M, Ferreira MV, Kramann R, Joyce CE, Hartigan C, et al. Rps14 haploinsufficiency causes a block in erythroid differentiation mediated by S100A8 and S100A9. Nat Med. 2016;22:288–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Boultwood J, Pellagatti A, Cattan H, Lawrie CH, Giagounidis A, Malcovati L, et al. Gene expression profiling of CD34+ cells in patients with the 5q- syndrome. Br J Haematol. 2007;139:578–89.

    Article  CAS  PubMed  Google Scholar 

  95. Fink EC, Ebert BL. The novel mechanism of lenalidomide activity. Blood. 2015;126:2366–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Caceres G, McGraw K, Yip BH, Pellagatti A, Johnson J, Zhang L, et al. TP53 suppression promotes erythropoiesis in del(5q) MDS, suggesting a targeted therapeutic strategy in lenalidomide-resistant patients. Proc Natl Acad Sci USA. 2013;110:16127–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Jädersten M, Saft L, Smith A, Kulasekararaj A, Pomplun S, Göhring G, et al. TP53 mutations in low-risk myelodysplastic syndromes with del(5q) predict disease progression. J Clin Oncol. 2011;29:1971–9.

    Article  PubMed  Google Scholar 

  98. Bello E, Pellagatti A, Shaw J, Mecucci C, Kušec R, Killick S, et al. CSNK1A1 mutations and gene expression analysis in myelodysplastic syndromes with del(5q). Br J Haematol. 2015;171:210–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Hayashi Y, Zhang Y, Yokota A, Yan X, Liu J, Choi K, et al. Pathobiological pseudohypoxia as a putative mechanism underlying myelodysplastic syndromes. Cancer Disco. 2018;8:1438–57.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by US National Institute of Health (NIH) grants R35 HL135795, R01HL123904, R01 HL118281, R01 HL128425, R01 HL132071, a grant from Edward P. Evans Foundation (Maciejewski) and a grant JSPS Overseas Research Fellow (Nagata).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Yasunobu Nagata or Jaroslaw P. Maciejewski.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nagata, Y., Maciejewski, J.P. The functional mechanisms of mutations in myelodysplastic syndrome. Leukemia 33, 2779–2794 (2019). https://doi.org/10.1038/s41375-019-0617-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41375-019-0617-3

This article is cited by

Search

Quick links