We reported in Ran et al that ectopic expression of RUNX1a, a naturally occurring isoform of RUNX1 that contains a DNA binding domain but lacks both transactivation and inhibitory domains of RUNX1b/c (Figure 1), enhanced the production of CD34+CD45+ hematopoietic stem/progenitor cells (HSPCs) from human embryonic stem cells and induced pluripotent stem cells (ES/iPS).1  HSPCs produced from ES/iPS cells transduced with lentiviruses overexpressing RUNX1a engrafted NSG mice and produced both myeloid and lymphoid cells. In the “Letter to the Editor” by Real et al,2  a valid point was raised that the transformation potential of RUNX1a could partially contribute to the enhancement of HSPC formation by RUNX1a, although no overexpansion or any other clear signs of transformation were observed throughout the 9 weeks post-transplantation in our studies. It should be noted that in addition to the dominant negative effect mentioned by Real et al, RUNX1a can be an activator or repressor in gene expression, but loses certain regulatory functions due to its lack of interaction with some positive and negative cofactors (Figure 1).3,4  In mouse models, overexpression of RUNX1a results in expansion of hematopoietic cells,5  lymphoid leukemia,6  and enhanced engraftment upon transplantation.5,7  In contrast, overexpression of RUNX1b/c promotes p53-dependent senescence,8,9  hematopoietic cell differentiation,10  and the loss of transplanted blood cells.5,11  Using RUNX1a, but not RUNX1c, in our studies is based on these previous discoveries.

Figure 1

Three isoforms of RUNX1 and interacting proteins. Selected domains and isoform-specific regions indicated in key. Coactivator interaction partners are in red. Co-repressor interaction partners are in green. Numbering of domains refers to the RUNX1b isoform.

Figure 1

Three isoforms of RUNX1 and interacting proteins. Selected domains and isoform-specific regions indicated in key. Coactivator interaction partners are in red. Co-repressor interaction partners are in green. Numbering of domains refers to the RUNX1b isoform.

Close modal

We agree that the potential for hematopoietic cell transformation due to long-term overexpression of RUNX1a is a concern. However, regulated transient expression of RUNX1a during hematopoietic development of ES/iPS cells could be very useful for expanding a rare population of HSPCs. This same principle is illustrated by the use of the very potent proto-oncogene c-Myc to generate iPS cells. In the “Discussion” section, we suggested the use of cell-permeable transcription factors as an alternative to lentiviral transduction and expression of RUNX1a.1  Although we suggested this strategy to avoid the inappropriate expression of endogenous genes via lentiviral integration, transient expression strategies would also eliminate the potentially negative impact of long-term overexpression of RUNX1a on HSPCs. We thank Real et al for raising this important issue, and giving us the opportunity to clarify our argument.

Regarding the expression of 3 isoforms of RUNX1, our data agree with the finding of Real et al that the expression of RUNX1a and RUNX1b/c is increased during the hematopoietic differentiation of human ES/iPS cells, and that RUNX1b/c expression is always higher than RUNX1a expression. This was illustrated in Ran et al,1 Figure 1A-B, and supplemental Figure 1.

Finally, Real et al2  questioned whether the engraftment we observed by CD45+ CD34+ HSPCs derived from RUNX1a-expressing human ES cells was due to an intrinsic feature of the HSPCs, or simply because we transplanted an unusually large number of HSPCs. At present, we cannot distinguish between those 2 possibilities. However, regardless of the mechanism, overexpression of RUNX1a permitted engraftment, either by promoting expansion of HSPCs in vitro, or by altering the properties of HSPCs in vivo; determining which is the case will be a focus of future studies.

In short, we demonstrate a positive effect of RUNX1a on promoting hematopoiesis from human pluripotent stem cells, which provides a potential novel avenue for generating therapeutic HSCs. Additional studies are necessary to examine its possible transforming ability and to create inducible expression systems for using RUNX1a in regenerative medicine.

Acknowledgments: The authors thank Dr Nancy Speck for valuable discussion and critical suggestions.

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

Correspondence: Dong-Er Zhang, Moores UCSD Cancer Center, University of California San Diego, La Jolla, CA 92093; e-mail: d7zhang@ucsd.edu.

1
Ran
 
D
Shia
 
WJ
Lo
 
MC
, et al. 
RUNX1a enhances hematopoietic lineage commitment from human embryonic stem cells and inducible pluripotent stem cells.
Blood
2013
, vol. 
121
 
15
(pg. 
2882
-
2890
)
2
Real
 
PJ
Navarro-Montero
 
O
Ramos-Mejia
 
V
Ayllón
 
V
Bueno
 
C
Menendez
 
P
The role of RUNX1 isoforms in hematopoietic commitment of human pluripotent stem cells.
Blood
2013
, vol. 
121
 
26
(pg. 
5250
-
5252
)
3
Lam
 
K
Zhang
 
DE
RUNX1 and RUNX1-ETO: roles in hematopoiesis and leukemogenesis.
Front Biosci
2012
, vol. 
17
 (pg. 
1120
-
1139
)
4
Petrovick
 
MS
Hiebert
 
SW
Friedman
 
AD
Hetherington
 
CJ
Tenen
 
DG
Zhang
 
DE
Multiple functional domains of AML1: PU.1 and C/EBPalpha synergize with different regions of AML1.
Mol Cell Biol
1998
, vol. 
18
 
7
(pg. 
3915
-
3925
)
5
Tsuzuki
 
S
Hong
 
D
Gupta
 
R
Matsuo
 
K
Seto
 
M
Enver
 
T
Isoform-specific potentiation of stem and progenitor cell engraftment by AML1/RUNX1.
PLoS Med
2007
, vol. 
4
 
5
pg. 
e172
 
6
Liu
 
X
Zhang
 
Q
Zhang
 
DE
, et al. 
Overexpression of an isoform of AML1 in acute leukemia and its potential role in leukemogenesis.
Leukemia
2009
, vol. 
23
 
4
(pg. 
739
-
745
)
7
Tsuzuki
 
S
Seto
 
M
Expansion of functionally defined mouse hematopoietic stem and progenitor cells by a short isoform of RUNX1/AML1.
Blood
2012
, vol. 
119
 
3
(pg. 
727
-
735
)
8
Wolyniec
 
K
Wotton
 
S
Kilbey
 
A
, et al. 
RUNX1 and its fusion oncoprotein derivative, RUNX1-ETO, induce senescence-like growth arrest independently of replicative stress.
Oncogene
2009
, vol. 
28
 
27
(pg. 
2502
-
2512
)
9
Wotton
 
SF
Blyth
 
K
Kilbey
 
A
, et al. 
RUNX1 transformation of primary embryonic fibroblasts is revealed in the absence of p53.
Oncogene
2004
, vol. 
23
 
32
(pg. 
5476
-
5486
)
10
Guo
 
H
Ma
 
O
Speck
 
NA
Friedman
 
AD
Runx1 deletion or dominant inhibition reduces Cebpa transcription via conserved promoter and distal enhancer sites to favor monopoiesis over granulopoiesis.
Blood
2012
, vol. 
119
 
19
(pg. 
4408
-
4418
)
11
Challen
 
GA
Goodell
 
MA
Runx1 isoforms show differential expression patterns during hematopoietic development but have similar functional effects in adult hematopoietic stem cells.
Exp Hematol
2010
, vol. 
38
 
5
(pg. 
403
-
416
)
Sign in via your Institution