To the editor:

Ten-Eleven-Translocation 2 (TET2) is one of the most frequently mutated genes in various hematologic malignancies.1,2  Li et al have previously reported that the deletion of Tet2 in mice led to dysregulated hematopoietic stem cells (HSCs) and subsequent development of myeloid malignancies.3  We and others have also reported similar findings using different strains of Tet2-mutant mice.4-7  These observations led us to speculate that enhanced HSC function of Tet2-mutant mice sets a critical background for malignant transformation, and such dysregulated HSCs are the cell of origin for myeloid malignancies. However, leukemic stem cells (LSCs) of myeloid leukemia have been shown to emerge from committed progenitors, such as common myeloid progenitors (CMPs) or granulocyte-monocyte progenitors with enhanced self-renewal capacity conferred by leukemia-associated oncogenes.8  Because Tet2 mutation enhances self-renewal of HSCs and causes expansion of myeloid progenitors, it is reasonably speculated that committed myeloid progenitors may also aberrantly acquire stem cell function by Tet2 mutation, and this in collaboration with their inherent high proliferative capacity leads to the development of LSCs. However, none of the reports so far have tested stem cell capacity of myeloid progenitors in Tet2-deficient mice.

To address this issue, we examined HSCs and myeloid progenitors in fetal livers (FLs) from hypomorphic Tet2 gene-trap (gt) mice.4  Serial replating colony formation assay showed increased replating capacity of Tet2gt/gt FL-LineageSca-1+c-Kit+ (LSK) cells as compared with wild-type (WT) ones as reported previously (Figure 1A, upper panel). Surprisingly, Tet2gt/gt FL-CMPs also showed enhanced serial replating capacity to the same level as Tet2gt/gt FL-LSKs, whereas WT FL-CMPs could not be replated more than 4 times (Figure 1A, lower panel). These data clearly indicate that functional loss of Tet2 confers aberrant in vitro self-renewal capacity, one of the hallmarks of LSCs, to FL-CMPs.

Figure 1

In vitro and in vivo self-renewal capacity of Tet2-mutant FL hematopoietic stem/ progenitor cells and myeloid progenitors. FL cells were obtained from E14.5 embryos of WT (Tet2+/+) and Tet2gt/gt mice. (A) LSK and CMP (LinSca-1c-Kit+IL7RαCD34+FcγRII/IIIlow) cells were sorted by flow cytometry (fluorescence-activated cell sorter [FACS]) and plated in methylcellulose media (MethoCult M3434; StemCell Technologies). Cells were cultured for 7 days before enumeration of colony numbers. Numbers of colonies from 500 FL-LSK cells (upper panel) or 1000 FL-CMP cells (lower panel) are shown. Upon replating, cells were recovered from dishes, counted, and plated in methylcellulose at 500 cells per dish for the LSK group and 1000 cells per dish for the CMP group, respectively. Data are the mean ± standard deviation (SD) (n = 3). (B) Five hundred CD150+LSK cells (Ly5.2) (upper panel) or 5000 CMP cells (Ly5.2) (lower panel) were sorted by FACS and transplanted into lethally (950 rads) irradiated primary recipients (Ly5.1) with 2 × 105 competitor bone marrow (BM) cells (Ly5.1). For secondary and tertiary transplantation, 2 × 106 whole BM cells taken from the primary or secondary recipients at 12 weeks after transplantation were transplanted into lethally irradiated recipient mice (Ly5.1). Percentages of donor chimerism in recipients’ peripheral blood (PB) were assessed by FACS at 4, 8, and 12 weeks after transplantation. Data are shown as the mean ± SD (n = 3-5). Statistical analyses were performed by unpaired Student t test. P < .05 was considered statistically significant. N.S., not significant.

Figure 1

In vitro and in vivo self-renewal capacity of Tet2-mutant FL hematopoietic stem/ progenitor cells and myeloid progenitors. FL cells were obtained from E14.5 embryos of WT (Tet2+/+) and Tet2gt/gt mice. (A) LSK and CMP (LinSca-1c-Kit+IL7RαCD34+FcγRII/IIIlow) cells were sorted by flow cytometry (fluorescence-activated cell sorter [FACS]) and plated in methylcellulose media (MethoCult M3434; StemCell Technologies). Cells were cultured for 7 days before enumeration of colony numbers. Numbers of colonies from 500 FL-LSK cells (upper panel) or 1000 FL-CMP cells (lower panel) are shown. Upon replating, cells were recovered from dishes, counted, and plated in methylcellulose at 500 cells per dish for the LSK group and 1000 cells per dish for the CMP group, respectively. Data are the mean ± standard deviation (SD) (n = 3). (B) Five hundred CD150+LSK cells (Ly5.2) (upper panel) or 5000 CMP cells (Ly5.2) (lower panel) were sorted by FACS and transplanted into lethally (950 rads) irradiated primary recipients (Ly5.1) with 2 × 105 competitor bone marrow (BM) cells (Ly5.1). For secondary and tertiary transplantation, 2 × 106 whole BM cells taken from the primary or secondary recipients at 12 weeks after transplantation were transplanted into lethally irradiated recipient mice (Ly5.1). Percentages of donor chimerism in recipients’ peripheral blood (PB) were assessed by FACS at 4, 8, and 12 weeks after transplantation. Data are shown as the mean ± SD (n = 3-5). Statistical analyses were performed by unpaired Student t test. P < .05 was considered statistically significant. N.S., not significant.

Close modal

Next, we assessed long-term repopulation and self-renewal capacity of FL-CMPs in vivo by serial transplantation assays. Consistent with previous reports, Tet2gt/gt CD150+LSK cells, highly purified FL-HSC fraction, showed significantly higher engraftment in the secondary and the tertiary recipients as compared with WT (Figure 1B, upper panel). In contrast, however, both WT and Tet2gt/gt FL-CMPs did not engraft for long-term in the primary recipients and completely lost repopulation in the secondary recipients (Figure 1B, lower panel). These data indicate that Tet2 loss did not confer FL-CMPs with in vivo stem cell capacity.

Taken together, these data seem to support the current notion that abnormal HSCs with TET2 mutation are the cell of origin for myeloid malignancies.9,10  However, they also raise a possibility that TET2-mutated myeloid progenitors are prone to additional mutations because of their enhanced self-renewal and behave as premalignant cells. In addition, enhanced self-renewal potential may contribute to the progression of myeloid disease by expanding TET2-mutated progenitors. It should be mentioned that our Tet2 gt mice are slightly different from other published models, and that we examined cells from FL, not from BM. Nonetheless, our observation calls for further investigations on the role of TET2-mutated myeloid progenitors in the development of myeloid disease.

Acknowledgments: The authors thank Akiko Ito and Junko Kawakita for excellent technical assistance, and S. Suzuki (FACS Core Laboratory, Keio University School of Medicine) for FACS sorting. This work was supported in part by a grant from the Ministry of Education, Culture, Sports, Science and Technology of Japan and Keio University Special Grant-in-Aid for Innovative Collaborative Research Projects.

Contribution: H.K. performed research, analyzed the data, and wrote the manuscript; Y.F., M.S., and K.T. performed research; S.O. supervised the study; and H.N. designed research, analyzed the data, and wrote the manuscript.

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

Correspondence: Hideaki Nakajima, Division of Hematology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan; e-mail: hnakajim@z2.keio.jp.

1
Delhommeau
 
F
Dupont
 
S
Della Valle
 
V
, et al. 
Mutation in TET2 in myeloid cancers.
N Engl J Med
2009
, vol. 
360
 
22
(pg. 
2289
-
2301
)
2
Couronné
 
L
Bastard
 
C
Bernard
 
OA
TET2 and DNMT3A mutations in human T-cell lymphoma.
N Engl J Med
2012
, vol. 
366
 
1
(pg. 
95
-
96
)
3
Li
 
Z
Cai
 
X
Cai
 
CL
, et al. 
Deletion of Tet2 in mice leads to dysregulated hematopoietic stem cells and subsequent development of myeloid malignancies.
Blood
2011
, vol. 
118
 
17
(pg. 
4509
-
4518
)
4
Kunimoto
 
H
Fukuchi
 
Y
Sakurai
 
M
, et al. 
Tet2 disruption leads to enhanced self-renewal and altered differentiation of fetal liver hematopoietic stem cells.
Sci Rep
2012
, vol. 
2
 pg. 
273
 
5
Moran-Crusio
 
K
Reavie
 
L
Shih
 
A
, et al. 
Tet2 loss leads to increased hematopoietic stem cell self-renewal and myeloid transformation.
Cancer Cell
2011
, vol. 
20
 
1
(pg. 
11
-
24
)
6
Quivoron
 
C
Couronné
 
L
Della Valle
 
V
, et al. 
TET2 inactivation results in pleiotropic hematopoietic abnormalities in mouse and is a recurrent event during human lymphomagenesis.
Cancer Cell
2011
, vol. 
20
 
1
(pg. 
25
-
38
)
7
Ko
 
M
Bandukwala
 
HS
An
 
J
, et al. 
Ten-Eleven-Translocation 2 (TET2) negatively regulates homeostasis and differentiation of hematopoietic stem cells in mice.
Proc Natl Acad Sci USA
2011
, vol. 
108
 
35
(pg. 
14566
-
14571
)
8
Huntly
 
BJ
Shigematsu
 
H
Deguchi
 
K
, et al. 
MOZ-TIF2, but not BCR-ABL, confers properties of leukemic stem cells to committed murine hematopoietic progenitors.
Cancer Cell
2004
, vol. 
6
 
6
(pg. 
587
-
596
)
9
Jan
 
M
Snyder
 
TM
Corces-Zimmerman
 
MR
, et al. 
Clonal evolution of preleukemic hematopoietic stem cells precedes human acute myeloid leukemia.
Sci Transl Med
2012
, vol. 
4
 
149
pg. 
149ra118
 
10
Busque
 
L
Patel
 
JP
Figueroa
 
ME
, et al. 
Recurrent somatic TET2 mutations in normal elderly individuals with clonal hematopoiesis.
Nat Genet
2012
, vol. 
44
 
11
(pg. 
1179
-
1181
)
Sign in via your Institution