Page 56 - 2019_12-Haematologica-web
P. 56

X. Yu et al.
is the fact that there are no CpG rich regions in the proxi- mal g-globin promoter, and in fact the entire β-globin locus is CpG poor. Previous work has shown that MBD2 does not bind directly to sequences in the β-globin gene locus in β-YAC transgenic mice;32 this is expected since there are no CpG rich regions in the locus. This suggests that MBD2-NuRD may exert its effect through regulation of other silencers. Here we investigated whether MBD2 depletion changes the expression of known g-globin silencers, and found that MBD2KO does not affect the expression of LRF and actually increases expression of BCL11A and KLF1. It remains possible that BCL11A inter- acts functionally with MBD2-NuRD in human erythroid cells, as depletion of either results in high levels of HbF without impairing differentiation. Current studies to iden- tify targets and interaction partners of MBD2 through which g-globin silencing is mediated are ongoing.
The data presented here firmly establish that MBD2- NuRD is a potent repressor of HbF expression in adult human erythroid cells, while MBD3-NuRD is not. Specific mutations in the intrinsically disordered region and CC domains of MBD2 necessary for association of other NuRD components that recruit HDAC and chro- matin remodeling sub-complexes abrogate the silencing
effect of NuRD on the g-globin gene (Figure 7). Finally, we re-enforce the finding that MBD2 knockdown in pri- mary human erythroid cells results in an 8-10-fold increase in %g/g+β mRNA expression without affecting erythroid differentiation. The fact that MBD2 null mice show only minor phenotypic abnormalities (mild deficits in maternal nurturing, a lower than normal body weight, and altered B-cell differentiation) but are otherwise fully viable and fertile49,50 suggests that therapies targeting MBD2, especially in somatic tissues, may have acceptable side effects.
Acknowledgments
The authors wish to thank Amy Jones and Lindsey Paisley for expert assistance in preparation of this manuscript.
Funding
This work was supported by National Institutes of Health grants; R56 DK29902 (GDG) and R01 DK115563 (GDG and DCW) from the National Institute of Diabetes, Digestive and Kidney Diseases, R01 GM098264 (DCW) from the National Institute of General Medical Sciences, and P30 CA16059 (VCU Massey Cancer Center Core Grant) from the National Cancer Institute.
References
1. Lettre G, Bauer DE. Fetal haemoglobin in sickle-cell disease: from genetic epidemiol- ogy to new therapeutic strategies. Lancet. 2016;387(10037):2554-2564.
2. Modell B, Darlison M. Global epidemiolo- gy of haemoglobin disorders and derived service indicators. Bull World Health Organ. 2008;86(6):480-487.
3. Vichinsky EP. Changing patterns of tha- lassemia worldwide. Ann N Y Acad Sci. 2005;1054:18-24.
4. Gaziev J, Lucarelli G. Stem cell transplanta- tion for hemoglobinopathies. Curr Opin Pediatr. 2003;15(1):24-31.
5. Olivieri NF. Reactivation of fetal hemoglo- bin in patients with beta-thalassemia. Semin Hematol. 1996;33(1):24-42.
6. Wood WG, Clegg JB, Weatherall DJ. Hereditary persistence of fetal haemoglo- bin (HPFH) and delta beta thalassaemia. Br J Haematol. 1979;43(4):509-520.
7. Platt OS, Brambilla DJ, Rosse WF, et al. Mortality in sickle cell disease. Life expectancy and risk factors for early death. N Engl J Med. 1994;330(23):1639-1644.
8. Sankaran VG, Orkin SH. The switch from fetal to adult hemoglobin. Cold Spring Harb Perspect Med. 2013;3(1):a011643.
9. Amaya M, Desai M, Gnanapragasam MN, et al. Mi2beta-mediated silencing of the fetal gamma-globin gene in adult erythroid cells. Blood. 2013;121(17):3493-3501.
10. Borg J, Patrinos GP, Felice AE, Philipsen S. Erythroid phenotypes associated with KLF1 mutations. Haematologica. 2011; 96(5):635-638.
11. Canver MC, Smith EC, Sher F, et al. BCL11A enhancer dissection by Cas9- mediated in situ saturating mutagenesis. Nature. 2015;527(7577):192-197.
12. Ginder GD. Epigenetic regulation of fetal
globin gene expression in adult erythroid
cells. Transl Res. 2015;165(1):115-125.
13. Masuda T, Wang X, Maeda M, et al. Transcription factors LRF and BCL11A independently repress expression of fetal hemoglobin. Science. 2016;351(6270):285-
289.
14. Sankaran VG, Menne TF, Xu J, et al. Human
fetal hemoglobin expression is regulated by the developmental stage-specific repressor BCL11A. Science. 2008;322(5909):1839- 1842.
15. Shi L, Cui S, Engel JD, Tanabe O. Lysine- specific demethylase 1 is a therapeutic tar- get for fetal hemoglobin induction. Nat Med. 2013;19(3):291-294.
16. Zhou D, Liu K, Sun CW, Pawlik KM, Townes TM. KLF1 regulates BCL11A expression and gamma- to beta-globin gene switching. Nat Genet. 2010;42(9):742-744.
17. Kurita R, Suda N, Sudo K, et al. Establishment of immortalized human ery- throid progenitor cell lines able to produce enucleated red blood cells. PLoS One. 2013;8(3):e59890.
18. Charache S, Dover G, Smith K, Talbot CC Jr., Moyer M, Boyer S. Treatment of sickle cell anemia with 5-azacytidine results in increased fetal hemoglobin production and is associated with nonrandom hypomethy- lation of DNA around the gamma-delta- beta-globin gene complex. Proc Natl Acad Sci U S A. 1983;80(15):4842-4846.
19. DeSimone J, Heller P, Hall L, Zwiers D. 5- Azacytidine stimulates fetal hemoglobin synthesis in anemic baboons. Proc Natl Acad Sci U S A. 1982;79(14):4428-4431.
20. Ginder GD, Whitters MJ, Pohlman JK. Activation of a chicken embryonic globin gene in adult erythroid cells by 5-azacyti- dine and sodium butyrate. Proc Natl Acad Sci U S A. 1984;81(13):3954-3958.
21. Ley TJ, DeSimone J, Anagnou NP, et al. 5-
azacytidine selectively increases gamma- globin synthesis in a patient with beta+ thalassemia. N Engl J Med. 1982;307(24): 1469-1475.
22. Perrine SP, Ginder GD, Faller DV, et al. A short-term trial of butyrate to stimulate fetal-globin-gene expression in the beta- globin disorders. N Engl J Med. 1993; 328(2):81-86.
23. Witt O, Monkemeyer S, Ronndahl G, et al. Induction of fetal hemoglobin expression by the histone deacetylase inhibitor api- cidin. Blood. 2003;101(5):2001-2007.
24. Cramer JM, Scarsdale JN, Walavalkar NM, Buchwald WA, Ginder GD, Williams DC, Jr. Probing the dynamic distribution of bound states for methylcytosine-binding domains on DNA. J Biol Chem. 2014;289(3):1294-1302.
25. Desai MA, Webb HD, Sinanan LM, et al. An intrinsically disordered region of methyl-CpG binding domain protein 2 (MBD2) recruits the histone deacetylase core of the NuRD complex. Nucleic Acids Res. 2015;43(6):3100-3113.
26. Ginder GD, Williams DC Jr. Readers of DNA methylation, the MBD family as potential therapeutic targets. Pharmacol Ther. 2018;184:98-111.
27. Gnanapragasam MN, Scarsdale JN, Amaya ML, et al. p66Alpha-MBD2 coiled-coil interaction and recruitment of Mi-2 are crit- ical for globin gene silencing by the MBD2- NuRD complex. Proc Natl Acad Sci U S A. 2011;108(18):7487-7492.
28. Ramirez J, Dege C, Kutateladze TG, Hagman J. MBD2 and multiple domains of CHD4 are required for transcriptional repression by Mi-2/NuRD complexes. Mol Cell Biol. 2012;32(24):5078-5088.
29. Feng Q, Zhang Y. The MeCP1 complex represses transcription through preferential binding, remodeling, and deacetylating
2370
haematologica | 2019; 104(12)


































































































   54   55   56   57   58