• Runx3 conditional knockout mice develop a myeloproliferative disorder when aged.

  • Runx3-deficient cells show hypersensitivity to G-CSF.

The RUNX family genes encode transcription factors that are involved in development and human diseases. RUNX1 is one of the most frequently mutated genes in human hematological malignancies and is a critical factor for the generation and maintenance of hematopoietic stem cells. Another Runx family gene, Runx3, is known to be expressed in hematopoietic cells. However, its involvement in hematopoiesis remains unclear. Here we show the hematopoietic phenotypes in Runx3 conditional knockout (KO) mice (Runx3fl/fl;Mx1-Cre+): whereas young Runx3 KO mice did not exhibit any significant hematopoietic defects, aged Runx3 KO mice developed a myeloproliferative disorder characterized by myeloid-dominant leukocytosis, splenomegaly, and an increase of hematopoietic stem/progenitor cells (HSPCs). Notably, Runx3-deficient cells showed hypersensitivity to granulocyte-colony stimulating factor, suggesting enhanced proliferative and mobilization capability of Runx3-deficient HSPCs when stimulated. These results suggest that, besides Runx1, Runx3 also plays a role in hematopoiesis.

The Runx genes belong to the Polyomavirus enhancer binding protein 2/core binding factor family of heterodimeric transcription factors that are important for development and are implicated in various human diseases.1-3  Of 3 family genes, RUNX1 is a well-documented hematopoietic and leukemia factor. RUNX1 is abrogated by multiple types of genetic alterations in human hematological malignancies3-5  and is critical for the generation and maintenance of hematopoietic stem cells (HSCs).6-10 Runx1 regulates the expression of stemness- and niche-related factors, such as Bmi1, Cxcr4, and integrin α2, deregulation of which in adult Runx1 knockout (KO) mice led to an expanded hematopoietic stem/progenitor cell (HSPC) compartment and subsequent stem cell exhaustion.6-12  In addition, adult Runx1 KO mice show abnormal megakaryocytic differentiation and defective lymphoid development.11-14  In contrast, the role of Runx3 in hematopoiesis remains elusive, despite the fact that Runx3 is expressed in various hematopoietic tissues in adult mice.15  We therefore sought to examine its effects on hematopoiesis in Runx3-deficient mice.

Mice

Runx3fl/fl chimeric mice obtained from genetically manipulated 129P2/O1aHsd-derived E14 embryonic stem cells injected into C57BL/6 blastocysts16  were backcrossed onto C57BL/6 inbred mice for ≥3 generations before being used for subsequent crosses with Mx1-Cre+ mice. For induction of the Mx-Cre transgene, 4-week-old Runx3fl/fl;Mx1-Cre+ mice were injected intraperitoneally with 600 µg of polyinosinic-polycytidylic acid (pIpC; Sigma-Aldrich) on 7 alternate days.

G-CSF stimulation

Mice were subcutaneously injected with 300 μg/kg/day murine granulocyte-colony stimulating factor (G-CSF; PeproTech) daily for 4 days. Twenty microliters of peripheral blood (PB) was obtained for the colony-forming unit culture (CFU-C) assay.

Additional procedures

For complete information on hematological analyses, flow cytometric analyses, CFU-C assay, bone marrow transplantation (BMT) procedures, and quantitative real-time polymerase chain reaction analysis, see supplemental Methods on the Blood website.

Generation of Runx3fl/fl;Mx1-Cre+ mice

Due to the neonatal lethality of conventional Runx3−/− mice, conditional Runx3 KO mice were generated. To induce Cre expression in hematopoietic cells, Runx3fl/fl;Mx1-Cre+ mice and Runx3fl/fl;Mx1-Cre littermates were injected with pIpC (hitherto referred to as Runx3 KO and Runx3 wild-type [WT] mice, respectively). Deletion of the Runx3 locus was nearly complete in the BM of Runx3 KO mice (supplemental Figure 1A).

Runx3 inactivation in young adult mice does not result in gross hematological abnormalities but leads to enhanced proliferative and mobilization ability of HSPCs when stimulated

Young Runx3 KO mice (6-8 weeks old) did not display gross hematopoietic abnormalities (supplemental Figure 2), evidenced by normal white blood cell (WBC), hemoglobin (Hb), and platelet (Plt) counts. Consistent with these findings, flow cytometric analysis showed no significant changes in various lineage populations, namely the myeloid, lymphoid, and erythroid lineages. There were no significant differences in BM cellularity, spleen weight, and thymus weight between Runx3 KO and Runx3 WT mice.

The HSPC compartment, immunophenotypically defined as a c-Kit+Sca-1+Lin (KSL) population, in the BM of Runx3 KO mice exhibited a slight increase, although this was statistically not significant (P = .11, Student t test; supplemental Figure 2D). In addition, a trend toward an increase in short-term HSC frequency with a marginal decrease in long-term HSC frequency in Runx3 KO mice was observed (supplemental Figure 2E). Interestingly, a significant increase in CFU-C activity was observed in cells from Runx3 KO mice (Figure 1A-E). The increased CFU-C activity was further pronounced after the replating assay (Figure 1B-D). Notably, Runx3 KO cells do not replate for a longer period of time compared with the WT cells. These results suggest that that Runx3 KO mice have enhanced CFU-C forming capacity of HSPCs compared with their WT littermates.

Figure 1

HSPCs from young Runx3 KO mice show enhanced proliferative and mobilization ability when stimulated. (A) Schematic diagram showing an overview of the CFU-C assay, including serial replating. Colonies were scored 7 days after plating. (B,D) Colony-forming potential of (B) whole bone marrow (BM) cells and (D) BM KSL cells. Mean ± standard deviation (SD) is shown (n = 2/genotype). All colonies were counted from triplicate samples. Two independent experiments were performed. Asterisks represent significant differences (**P < .01; ***P < .001, Student t test). (C,E) Morphology of colonies formed on each plate. Representative plates in first and second platings from B and D are shown. (F-G) Flow cytometric analysis of the KSL and c-Kit+Sca-1lineage (myeloid progenitor) compartment in the spleen at 6 weeks after pIpC treatment. Mean ± SD of percentage of (F) KSL and (G) myeloid progenitors in spleen is shown (n = 5/genotype). Three independent experiments were performed. n.s., no significant difference (Student t test). (H) G-CSF mobilization assay of cells from Runx3 KO mice (n = 7) and Runx3 WT mice (n = 5). Time course of progenitor cell numbers in 20 µL PB after in vivo G-CSF stimulation (300 μg/kg/day for 4 days) is shown. Mean ± SD of CFU-C numbers is shown. Asterisks represent significant differences (*P < .05; ***P < .001, Student t test).

Figure 1

HSPCs from young Runx3 KO mice show enhanced proliferative and mobilization ability when stimulated. (A) Schematic diagram showing an overview of the CFU-C assay, including serial replating. Colonies were scored 7 days after plating. (B,D) Colony-forming potential of (B) whole bone marrow (BM) cells and (D) BM KSL cells. Mean ± standard deviation (SD) is shown (n = 2/genotype). All colonies were counted from triplicate samples. Two independent experiments were performed. Asterisks represent significant differences (**P < .01; ***P < .001, Student t test). (C,E) Morphology of colonies formed on each plate. Representative plates in first and second platings from B and D are shown. (F-G) Flow cytometric analysis of the KSL and c-Kit+Sca-1lineage (myeloid progenitor) compartment in the spleen at 6 weeks after pIpC treatment. Mean ± SD of percentage of (F) KSL and (G) myeloid progenitors in spleen is shown (n = 5/genotype). Three independent experiments were performed. n.s., no significant difference (Student t test). (H) G-CSF mobilization assay of cells from Runx3 KO mice (n = 7) and Runx3 WT mice (n = 5). Time course of progenitor cell numbers in 20 µL PB after in vivo G-CSF stimulation (300 μg/kg/day for 4 days) is shown. Mean ± SD of CFU-C numbers is shown. Asterisks represent significant differences (*P < .05; ***P < .001, Student t test).

Close modal

HSPC analysis in the spleen of Runx3 KO mice revealed a marginal increase in the KSL and myeloid progenitor (c-Kit+Sca-1Lin) populations (Figure 1F-G). As increased HSPC numbers in the spleen is indicative of an enhanced mobilization ability, a mobilization assay was conducted using G-CSF, which is widely known to enhance the mobilization of HSPCs.17  The result showed that the number of CFU-Cs in the PB of Runx3 KO mice was significantly greater than that in Runx3 WT mice at all time points (Figure 1H). Therefore, Runx3 KO HSPCs appear to be easily mobilized into the periphery by G-CSF.

Aged Runx3 KO mice show enhanced myeloproliferation with an expanded HSPC compartment

To explore whether the hematopoietic abnormalities will be enhanced over time, Runx3 KO mice were aged to 18 months old. Deletion of the Runx3 locus in BM cells from aged Runx3 KO mice remained almost complete (supplemental Figure 1B). There was a significant increase in WBC counts in aged Runx3 KO mice compared with WT controls, whereas Hb and Plt counts remained unchanged (Figure 2A). Notably, the leukocytosis was dominated by a population of myeloid cells, characterized by abundant granulocytic cells in the PB and an increase in Mac-1+ and Gr-1+ populations in the BM and spleen of aged Runx3 KO mice (Figure 2B-C and data not shown). Analysis of other hematopoietic lineages in the BM revealed a significant decrease in the erythroid lineage (Figure 2C). These changes were accompanied by significant increases in BM cellularity and spleen weight (Figure 2D-E). HSPC analysis in aged Runx3 KO mice showed an increase of KSL and myeloid progenitor populations in BM and spleen compared with aged Runx3 WT mice (Figure 2F-I). Although the frequency of KSL in BM remained unchanged, which is thought to be attributable to the decreased Lin fraction in Runx3 KO BM, the absolute number of KSL cells was significantly increased in the aged Runx3 KO mice (supplemental Figure 3A-C). Analysis of the proliferative ability of KSL cells by Ki-67 staining revealed a tendency toward more Ki-67+ cells in Runx3 KO mice, suggesting that the increased HSPCs in aged mice is probably due to enhanced proliferation in a subtle but sustained manner (supplemental Figure 3D). Together, these results suggest that the increased WBC count and increased BM cellularity were due to an expansion of KSL and myeloid progenitor compartments and myeloid cells. Of note, development of spontaneous leukemia was not observed in aged Runx3 KO mice.

Figure 2

Aged Runx3 KO mice shows enhanced myeloproliferation with an expanded HSPC compartment. (A) Complete blood counts performed on 18-month-old Runx3 WT (n = 9) and Runx3 KO mice (n = 21). WBC, Hb, and Plt counts are shown. The statistical significance (P value, Student t test) is shown at the top. (B) May-Grünwald-Giemsa staining of PB cells in aged mice. Representative pictures of cells are shown. (C) Number of cells of each lineage in BM of 18-month-old mice. Lineage markers: myeloid, Mac-1+ and Gr-1+; B-cells, B220+CD19+; T cells, CD3+; erythroid, Ter119+. Mean ± standard deviation (SD) is shown (WT, n = 2; KO, n = 4). Three independent experiments were performed. Asterisks represent significant difference (**P < .01, Student t test). (D) BM cellularity of aged mice. Mean ± SD of the numbers of cells in BM are shown (WT, n = 2; KO, n = 4). Asterisks represent significant difference (**P < .01, Student t test). (E) Spleen of aged Runx3 KO mice. (Left) Graphical representation of spleen weight in the aged mice. Mean ± SD is shown (WT, n = 2; KO, n = 4). Asterisk represents significant difference (*P < .05, Student t test). Three independent experiments were performed. (Right) Representative pictures of spleens in the aged mice. (F,H) Flow cytometric analysis of the HSPC compartment in (F) BM and (H) spleen of 18-month-old mice. Representative FACS plots of 200 000 cells gated on viable Lineage cells are shown. (G,I) Graphical representations of results presented in F and H are shown in G and I, respectively. Mean ± SD of percentage of c-Kit+Sca-1Lineage (myeloid progenitors) and KSL within the BM Lineage population is shown (WT: n = 2; KO: n = 4). Three independent experiments were performed. (J) Time course PB chimerism analysis of recipient mice after BMT. CD45.1/CD45.2 competitive cells were cotransplanted with Runx3 WT or KO BM cells (CD45.2/CD45.2) into CD45.1/CD45.1 sublethally irradiated (8 Gy) mice in a 1:1 ratio. Percentage contribution of CD45.2/CD45.2 donor cells to total PB, myeloid, B-cell, and T-cell populations at indicated time points after BMT are shown. Mean ± SD is shown (WT, n = 3; KO, n = 6). Five of 8 mice receiving Runx3 WT donor cells and 3 of 9 mice receiving Runx3 KO donor cells that failed to show contribution of CD45.2/CD45.2 cells to PB of recipient mice are excluded. Asterisk represents significant differences (*P < .05, Student t test).

Figure 2

Aged Runx3 KO mice shows enhanced myeloproliferation with an expanded HSPC compartment. (A) Complete blood counts performed on 18-month-old Runx3 WT (n = 9) and Runx3 KO mice (n = 21). WBC, Hb, and Plt counts are shown. The statistical significance (P value, Student t test) is shown at the top. (B) May-Grünwald-Giemsa staining of PB cells in aged mice. Representative pictures of cells are shown. (C) Number of cells of each lineage in BM of 18-month-old mice. Lineage markers: myeloid, Mac-1+ and Gr-1+; B-cells, B220+CD19+; T cells, CD3+; erythroid, Ter119+. Mean ± standard deviation (SD) is shown (WT, n = 2; KO, n = 4). Three independent experiments were performed. Asterisks represent significant difference (**P < .01, Student t test). (D) BM cellularity of aged mice. Mean ± SD of the numbers of cells in BM are shown (WT, n = 2; KO, n = 4). Asterisks represent significant difference (**P < .01, Student t test). (E) Spleen of aged Runx3 KO mice. (Left) Graphical representation of spleen weight in the aged mice. Mean ± SD is shown (WT, n = 2; KO, n = 4). Asterisk represents significant difference (*P < .05, Student t test). Three independent experiments were performed. (Right) Representative pictures of spleens in the aged mice. (F,H) Flow cytometric analysis of the HSPC compartment in (F) BM and (H) spleen of 18-month-old mice. Representative FACS plots of 200 000 cells gated on viable Lineage cells are shown. (G,I) Graphical representations of results presented in F and H are shown in G and I, respectively. Mean ± SD of percentage of c-Kit+Sca-1Lineage (myeloid progenitors) and KSL within the BM Lineage population is shown (WT: n = 2; KO: n = 4). Three independent experiments were performed. (J) Time course PB chimerism analysis of recipient mice after BMT. CD45.1/CD45.2 competitive cells were cotransplanted with Runx3 WT or KO BM cells (CD45.2/CD45.2) into CD45.1/CD45.1 sublethally irradiated (8 Gy) mice in a 1:1 ratio. Percentage contribution of CD45.2/CD45.2 donor cells to total PB, myeloid, B-cell, and T-cell populations at indicated time points after BMT are shown. Mean ± SD is shown (WT, n = 3; KO, n = 6). Five of 8 mice receiving Runx3 WT donor cells and 3 of 9 mice receiving Runx3 KO donor cells that failed to show contribution of CD45.2/CD45.2 cells to PB of recipient mice are excluded. Asterisk represents significant differences (*P < .05, Student t test).

Close modal

The Runx family genes show a high degree of conservation in their amino acid sequences within the family and share the same capability to bind to their consensus DNA sequence. Therefore, the observation that Runx3 function was not fully unveiled in young Runx3 KO mice may be attributable to functional compensation among Runx family genes. However, Runx1 expression levels were not significantly altered in both young and aged Runx3 KO mice (supplemental Figure 3E). However, aged Runx3 KO mice partially but clearly phenocopied Runx1 KO mice, implying that Runx3 per se is also a critical factor for hematopoiesis. Although a clear increase of KSL fraction was observed in young Runx1 KO mice,6  a mild myeloid proliferation phenotype was only occasionally observed in aged Runx1 KO mice.11,13  In contrast, all aged Runx3 KO mice examined showed a myeloproliferative status accompanied by a modest HSPC expansion. Consistent with the higher penetrance of MPD in Runx3 KO mice than in Runx1 KO mice, HSC exhaustion was not observed in the Runx3 KO mice, unlike Runx1 KO mice, as Runx3 KO BM cells transplanted into WT recipient mice were not outcompeted by WT competitor cells at different doses (test: competitor ratios of 1:1, 2:1, and 4:1) in BMT experiments. In fact, transplantation with Runx3 KO donor cells resulted in greater chimerisms and higher WBC counts in the recipient mice, as opposed to mice transplanted with Runx3 WT BM cells (Figure 2J; supplemental Figure 3F; data not shown), implicating that Runx1 and Runx3 seem to play distinct roles in regulating the HSC behavior. The BMT result also suggests that the increase of phenotypic HSCs in aged Runx3 KO mice corresponds to an increase of functional HSCs and that the phenotypes observed were due to cell autonomous mechanisms. Differentiation blocks in megakaryocytic and lymphoid lineages, which were observed in Runx1 KO mice, were not evident in Runx3 KO mice. Intriguingly, Runx3-deficient HSPCs showed enhanced HSPC mobilization suggested by hypersensitivity to G-CSF, like RUNX1 haploinsufficient status, which is known to cause familial Plt disorder with predisposition to acute myeloid leukemia (AML).18 

Human data further implicate RUNX3 in leukemogenesis. Deletion in chromosome 1p36, containing the RUNX3 locus, is frequently observed in human leukemia cells.19-21  Methylation of the RUNX3 promoter in AML has also been reported,22  and RUNX3 expression is down-regulated in AML with t(8;21) or inv(16).23,24  Collectively, our work represents the first direct evidence for the pivotal role of Runx3 in hematopoiesis. Further analysis of Runx3 would provide us with deeper insight into leukemogenesis and may lead to future drug development.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The authors thank K. Rajewshky for Mx-Cre Tg mice and members of the Biological Resource Center, Biopolis, and MD2 Vivarium, NUS, for mouse husbandry.

This work was supported by the A*STAR (Agency of Science, Technology and Research), Biomedical Research Council, National Medical Research Council, Singapore National Research Foundation, and the Ministry of Education under the Research Center of Excellence Programme.

Contribution: C.Q.W. performed experiments, analyzed data, and wrote the manuscript; L.M. performed experiments; M.S., Y.I., and I.T. provided research tools; M.O. and V.T. designed research and wrote the manuscript; and C.Q.W., L.M., M.S., Y.I., I.T., M.O., and V.T. reviewed and approved the manuscript.

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

Correspondence: Motomi Osato, Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Dr, Singapore 117599; e-mail: csimo@nus.edu.sg; and Vinay Tergaonkar, Institute of Molecular and Cell Biology, 61 Biopolis Dr, Singapore 138673; e-mail: vinayt@imcb.a-star.edu.sg.

1
Ito
 
Y
RUNX genes in development and cancer: regulation of viral gene expression and the discovery of RUNX family genes.
Adv Cancer Res
2008
, vol. 
99
 (pg. 
33
-
76
)
2
Ito
 
Y
Oncogenic potential of the RUNX gene family: ‘overview’.
Oncogene
2004
, vol. 
23
 
24
(pg. 
4198
-
4208
)
3
Speck
 
NA
Gilliland
 
DG
Core-binding factors in haematopoiesis and leukaemia.
Nat Rev Cancer
2002
, vol. 
2
 
7
(pg. 
502
-
513
)
4
Osato
 
M
Point mutations in the RUNX1/AML1 gene: another actor in RUNX leukemia.
Oncogene
2004
, vol. 
23
 
24
(pg. 
4284
-
4296
)
5
Osato
 
M
Asou
 
N
Abdalla
 
E
, et al. 
Biallelic and heterozygous point mutations in the runt domain of the AML1/PEBP2alphaB gene associated with myeloblastic leukemias.
Blood
1999
, vol. 
93
 
6
(pg. 
1817
-
1824
)
6
Motoda
 
L
Osato
 
M
Yamashita
 
N
, et al. 
Runx1 protects hematopoietic stem/progenitor cells from oncogenic insult.
Stem Cells
2007
, vol. 
25
 
12
(pg. 
2976
-
2986
)
7
Jacob
 
B
Osato
 
M
Yamashita
 
N
, et al. 
Stem cell exhaustion due to Runx1 deficiency is prevented by Evi5 activation in leukemogenesis.
Blood
2010
, vol. 
115
 
8
(pg. 
1610
-
1620
)
8
Jacob
 
B
Osato
 
M
Stem cell exhaustion and leukemogenesis.
J Cell Biochem
2009
, vol. 
107
 
3
(pg. 
393
-
399
)
9
Motoda
 
L
Osato
 
M
Ito
 
Y
Runx1/AML1 is a guardian of hematopoietic stem cells.
Curr Cancer Therap Rev
2008
, vol. 
4
 
3
(pg. 
188
-
195
)
10
Wang
 
CQ
Jacob
 
B
Nah
 
GS
Osato
 
M
Runx family genes, niche, and stem cell quiescence.
Blood Cells Mol Dis
2010
, vol. 
44
 
4
(pg. 
275
-
286
)
11
Growney
 
JD
Shigematsu
 
H
Li
 
Z
, et al. 
Loss of Runx1 perturbs adult hematopoiesis and is associated with a myeloproliferative phenotype.
Blood
2005
, vol. 
106
 
2
(pg. 
494
-
504
)
12
Ichikawa
 
M
Asai
 
T
Saito
 
T
, et al. 
AML-1 is required for megakaryocytic maturation and lymphocytic differentiation, but not for maintenance of hematopoietic stem cells in adult hematopoiesis.
Nat Med
2004
, vol. 
10
 
3
(pg. 
299
-
304
)
13
Putz
 
G
Rosner
 
A
Nuesslein
 
I
Schmitz
 
N
Buchholz
 
F
AML1 deletion in adult mice causes splenomegaly and lymphomas.
Oncogene
2006
, vol. 
25
 
6
(pg. 
929
-
939
)
14
Taniuchi
 
I
Osato
 
M
Egawa
 
T
, et al. 
Differential requirements for Runx proteins in CD4 repression and epigenetic silencing during T lymphocyte development.
Cell
2002
, vol. 
111
 
5
(pg. 
621
-
633
)
15
Ng
 
CE
Osato
 
M
Tay
 
BH
Venkatesh
 
B
Ito
 
Y
cDNA cloning of Runx family genes from the pufferfish (Fugu rubripes).
Gene
2007
, vol. 
399
 
2
(pg. 
162
-
173
)
16
Naoe
 
Y
Setoguchi
 
R
Akiyama
 
K
, et al. 
Repression of interleukin-4 in T helper type 1 cells by Runx/Cbf beta binding to the Il4 silencer.
J Exp Med
2007
, vol. 
204
 
8
(pg. 
1749
-
1755
)
17
Wilson
 
A
Trumpp
 
A
Bone-marrow haematopoietic-stem-cell niches.
Nat Rev Immunol
2006
, vol. 
6
 
2
(pg. 
93
-
106
)
18
Cottles
 
GM
Osato
 
M
Yokomizo
 
T
, et al. 
Hematological analysis of AML1/PEBP2 alpha B+/− mice: A possible predisposition to myeloid leukemia.
Blood
1999
, vol. 
94
 
10
pg. 
218b
 
19
Mitelman
 
F
Mertens
 
F
Johansson
 
B
A breakpoint map of recurrent chromosomal rearrangements in human neoplasia.
Nat Genet
1997
, vol. 
15
 
Spec No
(pg. 
417
-
474
)
20
Mori
 
N
Morosetti
 
R
Lee
 
S
, et al. 
Allelotype analysis in the evolution of chronic myelocytic leukemia.
Blood
1997
, vol. 
90
 
5
(pg. 
2010
-
2014
)
21
Xie
 
D
Hofmann
 
WK
Mori
 
N
Miller
 
CW
Hoelzer
 
D
Koeffler
 
HP
Allelotype analysis of the myelodysplastic syndrome.
Leukemia
2000
, vol. 
14
 
5
(pg. 
805
-
810
)
22
Maddipoti
 
SC
Bueso-Ramos
 
C
Yang
 
H
, et al. 
 
Epigenetic silencing of the RUNX3 gene by promoter hypermethylation in patients with acute myeloid leukemia. Blood. 2008;112(11):3341
23
Cheng
 
CK
Li
 
L
Cheng
 
SH
, et al. 
Transcriptional repression of the RUNX3/AML2 gene by the t(8;21) and inv(16) fusion proteins in acute myeloid leukemia.
Blood
2008
, vol. 
112
 
8
(pg. 
3391
-
3402
)
24
Gutiérrez
 
NC
López-Pérez
 
R
Hernández
 
JM
, et al. 
Gene expression profile reveals deregulation of genes with relevant functions in the different subclasses of acute myeloid leukemia.
Leukemia
2005
, vol. 
19
 
3
(pg. 
402
-
409
)
Sign in via your Institution