To the editor:

Mabaera et al1  addressed the role of DNA hypomethylation in the induction of γ-globin expression by 5-azacytidine. On the basis of the failure of siRNA-mediated down-regulation of DNMT1 to induce expression of γ-globin in cultured human erythroid progenitors, the authors suggest “that the decrease in γ-globin promoter methylation seen with 5-azacytidine is not the primary cause of γ gene induction and fetal hemoglobin (HbF) production, but is a secondary effect related to gene activation by some other mechanism.” As an alternative, the authors propose changes in posttranscriptional RNA stability and translational efficiency possibly related to 5-azacytidine incorporation into RNA transcripts (5-azacytidine is a ribose). In proposing this mechanism, the authors fail to consider that decitabine (5-aza-2′-deoxycytidine), a deoxyribose analog of 5-azacytidine that is primarily incorporated into DNA,2,4  produces equivalent elevations of HbF in baboons and in patients with sickle cell disease at molar doses 10% to 20% that of 5-azacytidine5,,,,,,12  (corresponding to the approximate 10% of 5-azacytidine that is incorporated into DNA2 ). Furthermore, the ribonucleotide reductase inhibitor hydroxyurea, which would be expected to block conversion of 5-azacytidine into the deoxyribose form and its incorporation into DNA but not into RNA, blocks the ability of 5-azacytidine to induce HbF in baboons.7  These results are most consistent with a requirement for 5-azacytidine incorporation into DNA, necessary for DNA methyltransferase inhibition, in its mechanism of action.

Consistent with a transcription activation mechanism of action, γ-globin promoter hypomethylation produced by decitabine treatment of baboons resulted in recruitment of RNA polymerase II and acetylation of histone H3.13  Although these results are from baboon erythroid cells, β-globin locus structure and developmental globin switching are similar in baboons and man and 5-azacytidine– and decitabine–related observations from this model have successfully translated into the clinic.5,,,,,,12  Furthermore, observations with primary erythroid cells isolated after in vivo decitabine treatment may be more representative of clinical effects than treatment of human cells in vitro, using high levels of fetal bovine serum and growth factors. In the baboon model, γ-globin promoter methylation during HbF expression was significantly lower than that observed by Mabaera et al.1  We suggest that the siRNA and shRNA directed to DNMT1 may not have reduced γ-globin promoter DNA methylation to sufficient levels at an appropriate stage of erythroid differentiation to increase γ-globin expression. While the authors state that shRNA treatment reduced γ-globin methylation to levels observed in fetal liver erythroid cells (20%), this was only demonstrated on d17 of culture (Figure 6C1 ), a time beyond peak globin gene transcription. At earlier times (d9, d13) γ-globin methylation was significantly higher (50%-60%). Other DNMTs may assist or act in a redundant manner to maintain γ-globin gene DNA methylation and knock-down of DNMT1 alone may be inadequate to mimic the effect of 5-azacytidine or decitabine.14,15 

In conclusion, we suggest that the experimental evidence presented in the Mabaera paper is insufficient to support the conclusion that, with regards to the mechanism of induction of γ-globin expression by 5-azacytidine, decreased methylation of the γ-globin promoter is an unimportant secondary effect.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Donald Lavelle, Department of Medicine, University of Illinois at Chicago, VA Chicago, Westside Division MP151C, 820 S Damen Avenue, Chicago, IL 60612; e-mail: dlavelle@uic.edu.

1
Mabaera
R
Greene
MR
Richardson
CA
Conine
S
Kozul
CD
Lowrey
CH
Neither DNA hypomethylation nor changes in the kinetics of erythroid differentiation explain 5-azacytidine's ability to induce human fetal hemoglobin.
Blood
2008
111
411
420
2
Li
LH
Olin
EJ
Buskirk
HH
Reineke
LM
Cytotoxicity and mode of action of 5-azacytidine on L1210 leukemia.
Cancer Res
1970
30
2760
2769
3
Vesely
J
Cihak
A
Incorporation of a potent antileukemia agent, 5-aza-2′-deoxycytiidine, into DNA of cells from leukemic mice.
Cancer Res
1977
37
3684
3689
4
Momparler
RL
Vesely
J
Momparler
LF
Rivard
GE
Synergistic action of 5-aza-2′-deoxycytidine and 3-deazauridine on L1210 leukemic cells and EMT6 tumor cells.
Cancer Res
1979
39
3822
3827
5
DeSimone
J
Heller
P
Molokie
RE
Hall
L
Zwiers
D
Tetrahydrouridine, cytidine analogues, and hemoglobin F.
Am J Hematol
1985
18
283
288
6
Saunthararajah
Y
Hillery
CA
Lavelle
D
et al
Effects of 5-aza-2′-deoxycytidine on fetal hemoglobin levels, red cell adhesion, and hematopoietic differentiation in patients with sickle cell disease.
Blood
2003
102
3865
3870
7
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
7
4428
4431
8
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
1469
1476
9
Ley
TJ
DeSimone
J
Noguchi
CT
et al
5-azacytidine increases gamma-globin synthesis and reduces the proportion of dense cells in patients with sickle cell anemia.
Blood
1983
62
370
380
10
Lowrey
CH
Nienhuis
AW
Treatment with 5-azacitidine of patients with end-stage β-thalassemia.
N Engl J Med
1993
329
845
848
11
Koshy
M
Dorn
L
Bressler
L
et al
2-deoxycytidine-5-azacytidine and fetal hemoglobin induction in sickle cell anemia.
Blood
2000
96
2379
2384
12
DeSimone
J
Koshy
M
Dorn
L
et al
Maintenance of elevated fetal hemoglobin levels by decitabine during dose interval treatment of sickle cell anemia.
Blood
2002
99
3905
3908
13
Lavelle
D
Vaitkus
K
Hankewych
M
Singh
M
DeSimone
J
Effect of 5-aza-2′-deoxycytidine (Dacogen) on covalent histone modifications of chromatin associated with the ε-, γ-, and β-globin gene promoters in Papio anubis.
Exp Hematol
2006
34
339
347
14
Rhee
I
Jair
KW
Yen
RW
et al
CpG methylation is maintained in human cancer cells lacking DNMT1.
Nature
2000
404
1003
1007
15
Ting
AH
Jair
KW
Schubel
KE
Baylin
SB
Differential requirement for DNA methyltransferase I in maintaining human cancer cell gene promoter hypermethylation.
Cancer Res
2006
66
729
735
Sign in via your Institution