• AML1-ETO-W692A loses interaction between NHR4 and N-CoR, decreases AML1-ETO cellular dysregulation, and promotes leukemia development in mice.

AML1-ETO (RUNX1-ETO) fusion proteins are generated by the 8;21 translocation, commonly found in acute myeloid leukemia, which fuses the AML1 (RUNX1) and ETO (MTG8, RUNX1T1) genes. Previous studies have shown that AML1-ETO interferes with AML1 function but requires additional cooperating mutations to induce leukemia development. In mouse models, AML1-ETO forms lacking the C-terminus have been shown to have greatly enhanced leukemogenic potential. Here, we investigate the role of 2 AML1-ETO C-terminal-interacting proteins, N-CoR, a transcriptional corepressor, and SON, a splicing/transcription factor required for cell cycle progression, in AML1-ETO-induced leukemia development. AML1-ETO-W692A loses N-CoR binding at NHR4, displays attenuated transcriptional repression ability and decreased cellular dysregulation, and promotes leukemia in vivo. These results support the importance of the degree of dysregulation by AML1-ETO in cellular transformation and demonstrate that AML1-ETO-W692A can be used as an effective experimental model for determining which factors compromise the leukemogenic potential of AML1-ETO.

t(8;21), one of the most common chromosomal abnormalities associated with acute myeloid leukemia (AML),1-4  causes expression of fusion protein AML1-ETO. AML1-ETO alone is insufficient to induce leukemia in mouse models,5  and additional cooperating mutations are required.6  If AML1-ETO is C-terminally truncated through mutation or alternative splicing, it rapidly induces leukemia.7,8 

AML1-ETO C-terminus interacts with several factors, including nuclear receptor corepressors N-CoR/SMRT9  and the large nuclear protein SON.10  N-CoR associates with mSin3A and histone deacetylases,11,12  and the N-CoR/AML1-ETO interaction mediates aberrant transcriptional repression of AML1 target genes.13-15  SON is a DNA- and RNA-binding protein, is involved in regulation of gene transcription and RNA splicing, and is required for cell cycle progression.16,17  Blocking protein interactions with AML1-ETO C-terminus by overexpression of a SON fragment is sufficient to rescue cells from AML1-ETO-induced growth arrest.10 

C-terminal C663S mutation of AML1-ETO reportedly destroyed the zinc-chelating structure of the NHR4 domain and disrupted N-CoR interaction with NHR4.14,18  We previously showed that AML1-ETO-C663S had similar leukemogenic potential to C-terminally truncated AML1-ETO and that ETO-C663S mutation eliminated SON interaction but retained N-CoR binding.10  The interaction between N-CoR and ETO-C663S is likely a result of N-CoR and Sin3A binding to other ETO domains besides NHR4.14  As stated in our previous report, although we hypothesized that the SON/NHR4 interaction may be one factor inhibiting AML1-ETO leukemogenic potential, we could not exclude the possibility that reduced N-CoR recruitment to AML1-ETO-C663S or other factors also contributes to leukemogenesis.10  As both aberrant transcriptional regulation and cell cycle progression could conceivably attenuate AML1-ETO leukemogenicity, it is important to determine which process, recruitment of N-CoR or inactivation/sequestration of SON, was responsible. Using AML1-ETO mutants to differentiate N-CoR and SON binding, we aim here to delineate the contributions of these 2 proteins in decreasing AML1-ETO leukemogenicity, as they each potentially contribute through very different mechanisms requiring different therapeutic strategies for effective treatment of AML. We demonstrate that AML1-ETO-W692A mutation loses NHR4/N-CoR interaction, attenuates AML1-ETO cellular effects, and promotes leukemogenesis, supporting a new model for study of AML1-ETO transformation and leukemia.

Plasmids, immunoprecipitation, and immunoblotting

NHR2-4 constructs expressed from pCMV2 (Sigma-Aldrich). Amino acid mutations generated by site-directed mutagenesis (Agilent). Immunoprecipitation: precleared lysate incubated 4 h-overnight at 4°C with antibody and washed 5 times with lysis buffer.16  Antibodies: N-CoR (Abcam); Flag, α-tubulin (Sigma-Aldrich); CDK4, Cyclin A, Cyclin D3 (Santa Cruz); p27 (BD Biosciences); HA (Covance).

Luciferase reporter assay

Performed as previously described.19  Significance determined by Student t test.

Apoptosis and MTS assays

K562 or primary bone marrow cells transduced with virus produced by cotransfection of packaging vector and MSCV-IRES-Puror containing indicated AML1-ETO constructs in 293T cells, selected 2 to 3 d in 2 μg/mL puromycin, and subjected to Annexin V Apoptosis Kit (BD Pharmingen) or CellTiter 96 AQueous One Solution Cell Proliferation Assay (Promega) per manufacturers’ instructions.

Animals

C57BL/6 mice housed in a pathogen-free facility. Procedures approved by Institutional Animal Care and Use Committee of University of California San Diego.

Mouse survival

Kaplan-Meier survival curves generated using GraphPad Prism4 (GraphPad Software, San Diego, CA).

Fetal liver-cell isolation, infection, and transplantation

See supplemental Methods.

Three groups have reported that N-CoR/SMRT directly interacts with NHR4,13-15  although AML1-ETO without the NHR4 domain can still interact with N-CoR via another site (Figure 1A). C663S mutation is predicted to disrupt the zinc-chelating structure of NHR4 and thereby block all its protein-protein interactions. W692A retains NHR4 structure but is predicted to lose N-CoR interaction.18  To determine whether W692A disrupts N-CoR binding to NHR4, we coimmunoprecipitated endogenous N-CoR and transiently transfected ETO fragments beginning at NHR2, to allow proper tetramerization,20  and continuing through ETO C-terminus (Figure 1A, ‘NHR2-4’), thereby eliminating the additional N-CoR binding site between NHR1 and NHR221  and allowing us to examine specifically the N-CoR interaction at NHR4. Endogenous N-CoR interacts with wild-type, but not W692A-mutant, NHR2-4 (Figure 1B). Conversely, transiently transfected SON retains interaction with W692A (Figure 1C). To substantiate this result further, we confirmed that endogenous SON also retains its interaction with Flag-NHR2-4-W692A (supplemental Figure 1). Combined, these results demonstrate that NHR4-W692A loses N-CoR, but retains SON, binding.

Figure 1

Characterization of NHR4/N-CoR and AML1-ETO/SON binding and effects on gene repression. (A) Structures of AML1-ETO and the NHR2-4 constructs used in the co-IP experiments, with location of N-CoR binding sites indicated. (B) Endogenous N-CoR interacts with WT, but not W692A, NHR2-4. Cell lysates were immunoprecipitated with N-CoR or isotype control (ctrl) antibodies, and N-CoR and NHR2-4 were detected with N-CoR and Flag antibodies, respectively. Tubulin serves as a loading control. (C) SON interacts with both WT and W692A NHR2-4. Cell lysates were immunoprecipitated with HA antibody, and SON and NHR2-4 were detected using HA and Flag antibodies, respectively. (D) SON interacts with full-length AML1-ETO (AE), but not when the NHR3 domain is deleted (ΔNHR3) or when amino acids 629-634 ‘TERAKM’ in NHR3 are mutated to ‘AAAAAA’ (mutNHR3). Immunoprecipitation is as performed in (C). (E) Endogenous N-CoR retains interaction with NHR2-4 containing mutNHR3 alanine mutations. Immunoprecipitation performed as in (B). (F) W692A decreases ability of AML1-ETO to repress a luciferase reporter; mutNHR3 does not. 293T cells were cotransfected with firefly luciferase reporter containing tandem AML1 binding sites upstream of the basal TK promoter, Renilla control luciferase and either empty vector (control) or indicated AML1-ETO construct. Firefly luciferase expression was normalized to Renilla expression and control was set to 1. Equal expression of AML1-ETO constructs was confirmed by immunoblot using HA antibody (data not shown). Data show averages and standard deviations of 3 independent experiments, each performed in duplicate. EV, empty vector; WCL, whole cell lysate.

Figure 1

Characterization of NHR4/N-CoR and AML1-ETO/SON binding and effects on gene repression. (A) Structures of AML1-ETO and the NHR2-4 constructs used in the co-IP experiments, with location of N-CoR binding sites indicated. (B) Endogenous N-CoR interacts with WT, but not W692A, NHR2-4. Cell lysates were immunoprecipitated with N-CoR or isotype control (ctrl) antibodies, and N-CoR and NHR2-4 were detected with N-CoR and Flag antibodies, respectively. Tubulin serves as a loading control. (C) SON interacts with both WT and W692A NHR2-4. Cell lysates were immunoprecipitated with HA antibody, and SON and NHR2-4 were detected using HA and Flag antibodies, respectively. (D) SON interacts with full-length AML1-ETO (AE), but not when the NHR3 domain is deleted (ΔNHR3) or when amino acids 629-634 ‘TERAKM’ in NHR3 are mutated to ‘AAAAAA’ (mutNHR3). Immunoprecipitation is as performed in (C). (E) Endogenous N-CoR retains interaction with NHR2-4 containing mutNHR3 alanine mutations. Immunoprecipitation performed as in (B). (F) W692A decreases ability of AML1-ETO to repress a luciferase reporter; mutNHR3 does not. 293T cells were cotransfected with firefly luciferase reporter containing tandem AML1 binding sites upstream of the basal TK promoter, Renilla control luciferase and either empty vector (control) or indicated AML1-ETO construct. Firefly luciferase expression was normalized to Renilla expression and control was set to 1. Equal expression of AML1-ETO constructs was confirmed by immunoblot using HA antibody (data not shown). Data show averages and standard deviations of 3 independent experiments, each performed in duplicate. EV, empty vector; WCL, whole cell lysate.

Close modal

To find a mutant of AML1-ETO with the opposite interaction pattern, we mapped the SON binding site through deletion and alanine mutation. We previously reported that NHR4 C663S mutation impairs SON interaction (supplemental Figure 1).10  Because disruption of NHR4 structure may also affect protein-protein interactions with neighboring regions of AML1-ETO, we examined whether SON interacts with adjacent domains. Interestingly, deletion of either NHR3 or NHR4 blocks SON interaction with AML1-ETO (supplemental Figure 2). Furthermore, mutation of 6 amino acids (TERAKM) within NHR3 to alanines (AE-mutNHR3) blocks AML1-ETO interaction with SON (Figure 1D), but not N-CoR (Figure 1E).

AML1-ETO downregulates AML1 target genes via interaction with N-CoR/SMRT and associated HDAC complexes,14,15  and loss of N-CoR/NHR4 interaction attenuates the function of AML1-ETO.18  To examine the effect of W692A and NHR3 mutation on AML1-ETO transcriptional repression, we cotransfected AML1-ETO constructs with an AML1 target promoter-luciferase reporter.19  As expected, AML1-ETO strongly repressed luciferase expression up to 80% of control, whereas AML1-ETO9a, which lacks NHR3 and NHR4 domains,8  displayed attenuated repression (Figure 1F). Interestingly, AE-mutNHR3 behaved like full-length AML1-ETO, but AE-W692A displayed impaired repression, similar to AML1-ETO9a (Figure 1F). This same pattern of gene dysregulation was also observed at endogenous AML1-ETO targets in primary murine bone marrow cells (supplemental Figure 3).18  AE-W692A thereby decreases AML1-ETO transcriptional repression, correlating the number of N-CoR binding sites with repression ability.

We next tested the functional effects of these mutants on cellular dysregulation. AE-mutNHR3 increased apoptosis and decreased proliferation in both primary murine bone marrow (Figures 2A-C) and K562 cells (supplemental Figure 4). AE-mutNHR3 also increased p27 levels and decreased expression of CDK4 and cyclins A and D3 (Figure 2D). These results mirror those obtained with full-length AML1-ETO and demonstrate the negative effect of these proteins on survival and cell cycle. AE-W692A, however, behaved similarly to the leukemogenic AE9a, demonstrating significantly reduced defects in the previously mentioned assays (Figures 2A-D; supplemental Figure 4). These results clearly indicate stronger negative cellular effects of AE-mutNHR3 compared to AE-W692A.

Figure 2

Effects of AML1-ETO–mutNHR3 and –W692A on cellular dysregulation. (A) Effects of AML1-ETO mutants on apoptosis in primary bone marrow cells. Murine bone marrow cells transduced with the indicated AML1-ETO constructs were harvested after 2 d of selection, stained with 7-AAD and Annexin V-APC, and analyzed by flow cytometry. Gating controls are shown at left. Data are representative of 3 independent experiments. (B) Averages and standard deviations of 3 independent experiments from (A). (C) Effects of AE constructs on cellular metabolic activity. Primary bone marrow cells were transduced and selected as in (A), seeded in duplicate at 10 000 cells/well in a 96-well plate, and assayed every 2 d by MTS assay. Data are representative of 3 independent experiments. (D) Immunoblot analysis of CDK4, cyclins A and D3, and p27 expression in K562 cells infected and selected as in supplemental Figure 4A. Data are representative of 4 independent experiments. (E) W692A increases leukemogenic potential of AML1-ETO. Kaplan-Meier survival curves of mice transplanted with hematopoietic cells expressing the indicated AML1-ETO construct. Data combined from 3 independent transplantations. (F) Presence of hematopoietic blast cells in tissues of mice transplanted with AML1-ETO-W692A expressing cells. Peripheral blood smear and cytocentrifugation of bone marrow and spleen cells were stained with Wright-Giemsa solutions.

Figure 2

Effects of AML1-ETO–mutNHR3 and –W692A on cellular dysregulation. (A) Effects of AML1-ETO mutants on apoptosis in primary bone marrow cells. Murine bone marrow cells transduced with the indicated AML1-ETO constructs were harvested after 2 d of selection, stained with 7-AAD and Annexin V-APC, and analyzed by flow cytometry. Gating controls are shown at left. Data are representative of 3 independent experiments. (B) Averages and standard deviations of 3 independent experiments from (A). (C) Effects of AE constructs on cellular metabolic activity. Primary bone marrow cells were transduced and selected as in (A), seeded in duplicate at 10 000 cells/well in a 96-well plate, and assayed every 2 d by MTS assay. Data are representative of 3 independent experiments. (D) Immunoblot analysis of CDK4, cyclins A and D3, and p27 expression in K562 cells infected and selected as in supplemental Figure 4A. Data are representative of 4 independent experiments. (E) W692A increases leukemogenic potential of AML1-ETO. Kaplan-Meier survival curves of mice transplanted with hematopoietic cells expressing the indicated AML1-ETO construct. Data combined from 3 independent transplantations. (F) Presence of hematopoietic blast cells in tissues of mice transplanted with AML1-ETO-W692A expressing cells. Peripheral blood smear and cytocentrifugation of bone marrow and spleen cells were stained with Wright-Giemsa solutions.

Close modal

SON is also expressed as an isoform lacking exon 3 (SONΔE3), which is expressed at very low levels in blood cell lines, is not detectable by Northern blot, and loses interaction with both AE-W692A and AE-mutNHR3 (supplemental Figure 5). Because these two mutants have very different effects on AML1-ETO function (Figure 2), it is unlikely that loss of SONΔE3 binding contributes to AE-W692A phenotypes.

Because AML1-ETO-W692A behaved similarly to AE9a in vitro, we tested whether it also induced leukemia in a mouse model. AML1-ETO-C663S rapidly induced leukemia, as previously reported,10  and AML1-ETO-W692A induced oligoclonal leukemia with indistinguishable latency (Figure 2E; supplemental Figure 6), with increased numbers of blast cells in peripheral blood, bone marrow, and spleen (Figure 2F), confirming that single W692A substitution is sufficient to enable AML1-ETO-induced leukemogenesis. These phenotypes are similar to those of AML1-ETO9a-induced leukemia.8 

In conclusion, we have shown that W692A substitution disrupts N-CoR/NHR4 binding, but interacts with SON. Conversely, AE-mutNHR3 disrupts SON binding, but not N-CoR. W692A substitution correlates with attenuated transcriptional repression and reduced negative effects of AML1-ETO in vitro. Loss of SON binding causes no such change in in vitro phenotypes. Finally, AML1-ETO-W692A is sufficient to induce leukemia in vivo. Therefore, between SON and N-CoR, SON is not the factor that interacts with NHR4 and diminishes the leukemogenic potential of AML1-ETO via NHR4 domain. In contrast, it is more likely that N-CoR binding to NHR4 enhances the transcription repression activity of AML1-ETO and reduces the leukemogenic potential of AML1-ETO. However, it is also possible that other N-CoR unrelated factor(s) interact with AML1-ETO but not with AML1-ETO-W692A, and they regulate the leukemogenic potential of AML1-ETO.

Attenuation of AML1-ETO-driven transcriptional repression and cellular dysregulation correlates with enhanced leukemogenic potential. Although seemingly counterintuitive, this is not unprecedented. Mice with 50% to 80% (but not complete) loss of Pu.1 expression developed a precancerous state predisposing them to leukemia.22  Similarly, histone acetyltransferase mutations correlated with leukemia and lymphoma development were usually monoallelic or otherwise did not completely block HAT function, indicating that decreased dosage or attenuated function is sufficient for disease progression.23,24  We propose a similar model for the contribution of AML1-ETO to leukemia development, in which the attenuation of AML1-ETO function, through mutations that either directly affect the interaction of N-CoR or other proteins at W692 or reduce the downstream effects of these interactions, allows cells to overcome the negative effects of AML1-ETO and to promote leukemia 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 Dr John Bushweller and all members of the D.-E.Z. laboratory for valuable discussions.

This work was supported by National Institutes of Health grants CA096735 (D.-E.Z.) and 5T32GM007240.

Contribution: R.C.D. designed and performed the research and wrote the paper; M.Y., E.-Y.A., and W.-J.S. designed and performed the research; N.A.S. provided critical reagents and aided in manuscript preparation; and D.-E.Z. supervised experimental design, data analysis, and manuscript preparation.

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

The current affiliation for E.-Y.A. is Mitchell Cancer Institute, University of South Alabama, Mobile, AL.

Correspondence: Dong-Er Zhang, Mail Code 0815, Room 5328, Moores UCSD Cancer Center, University of California San Diego, 3855 Health Sciences Dr, La Jolla, CA 92093; e-mail: d7zhang@ucsd.edu.

1
Nucifora
 
G
Rowley
 
JD
AML1 and the 8;21 and 3;21 translocations in acute and chronic myeloid leukemia.
Blood
1995
, vol. 
86
 
1
(pg. 
1
-
14
)
2
Langabeer
 
SE
Walker
 
H
Rogers
 
JR
, et al. 
MRC Adult Leukaemia Working Party
Incidence of AML1/ETO fusion transcripts in patients entered into the MRC AML trials.
Br J Haematol
1997
, vol. 
99
 
4
(pg. 
925
-
928
)
3
Rege
 
K
Swansbury
 
GJ
Atra
 
AA
, et al. 
Disease features in acute myeloid leukemia with t(8;21)(q22;q22). Influence of age, secondary karyotype abnormalities, CD19 status, and extramedullary leukemia on survival.
Leuk Lymphoma
2000
, vol. 
40
 
1-2
(pg. 
67
-
77
)
4
Rowe
 
D
Cotterill
 
SJ
Ross
 
FM
, et al. 
Cytogenetically cryptic AML1-ETO and CBF beta-MYH11 gene rearrangements: incidence in 412 cases of acute myeloid leukaemia.
Br J Haematol
2000
, vol. 
111
 
4
(pg. 
1051
-
1056
)
5
Peterson
 
LF
Boyapati
 
A
Ahn
 
EY
, et al. 
Acute myeloid leukemia with the 8q22;21q22 translocation: secondary mutational events and alternative t(8;21) transcripts.
Blood
2007
, vol. 
110
 
3
(pg. 
799
-
805
)
6
Müller
 
AM
Duque
 
J
Shizuru
 
JA
Lübbert
 
M
Complementing mutations in core binding factor leukemias: from mouse models to clinical applications.
Oncogene
2008
, vol. 
27
 
44
(pg. 
5759
-
5773
)
7
Yan
 
M
Burel
 
SA
Peterson
 
LF
, et al. 
Deletion of an AML1-ETO C-terminal NcoR/SMRT-interacting region strongly induces leukemia development.
Proc Natl Acad Sci USA
2004
, vol. 
101
 
49
(pg. 
17186
-
17191
)
8
Yan
 
M
Kanbe
 
E
Peterson
 
LF
, et al. 
A previously unidentified alternatively spliced isoform of t(8;21) transcript promotes leukemogenesis.
Nat Med
2006
, vol. 
12
 
8
(pg. 
945
-
949
)
9
Peterson
 
LF
Zhang
 
DE
The 8;21 translocation in leukemogenesis.
Oncogene
2004
, vol. 
23
 
24
(pg. 
4255
-
4262
)
10
Ahn
 
EY
Yan
 
M
Malakhova
 
OA
, et al. 
Disruption of the NHR4 domain structure in AML1-ETO abrogates SON binding and promotes leukemogenesis.
Proc Natl Acad Sci USA
2008
, vol. 
105
 
44
(pg. 
17103
-
17108
)
11
Hörlein
 
AJ
Näär
 
AM
Heinzel
 
T
, et al. 
Ligand-independent repression by the thyroid hormone receptor mediated by a nuclear receptor co-repressor.
Nature
1995
, vol. 
377
 
6548
(pg. 
397
-
404
)
12
Heinzel
 
T
Lavinsky
 
RM
Mullen
 
TM
, et al. 
A complex containing N-CoR, mSin3 and histone deacetylase mediates transcriptional repression.
Nature
1997
, vol. 
387
 
6628
(pg. 
43
-
48
)
13
Wang
 
J
Hoshino
 
T
Redner
 
RL
Kajigaya
 
S
Liu
 
JM
ETO, fusion partner in t(8;21) acute myeloid leukemia, represses transcription by interaction with the human N-CoR/mSin3/HDAC1 complex.
Proc Natl Acad Sci USA
1998
, vol. 
95
 
18
(pg. 
10860
-
10865
)
14
Gelmetti
 
V
Zhang
 
J
Fanelli
 
M
Minucci
 
S
Pelicci
 
PG
Lazar
 
MA
Aberrant recruitment of the nuclear receptor corepressor-histone deacetylase complex by the acute myeloid leukemia fusion partner ETO.
Mol Cell Biol
1998
, vol. 
18
 
12
(pg. 
7185
-
7191
)
15
Lutterbach
 
B
Westendorf
 
JJ
Linggi
 
B
, et al. 
ETO, a target of t(8;21) in acute leukemia, interacts with the N-CoR and mSin3 corepressors.
Mol Cell Biol
1998
, vol. 
18
 
12
(pg. 
7176
-
7184
)
16
Ahn
 
EY
DeKelver
 
RC
Lo
 
MC
, et al. 
SON controls cell-cycle progression by coordinated regulation of RNA splicing.
Mol Cell
2011
, vol. 
42
 
2
(pg. 
185
-
198
)
17
Ahn
 
EE
Higashi
 
T
Yan
 
M
, et al. 
SON regulates GATA-2 through transcriptional control of the miR-23a∼27a∼24-2 cluster.
J Biol Chem
2013
, vol. 
288
 
8
(pg. 
5381
-
5388
)
18
Liu
 
Y
Chen
 
W
Gaudet
 
J
, et al. 
Structural basis for recognition of SMRT/N-CoR by the MYND domain and its contribution to AML1/ETO’s activity.
Cancer Cell
2007
, vol. 
11
 
6
(pg. 
483
-
497
)
19
Okumura
 
AJ
Peterson
 
LF
Okumura
 
F
Boyapati
 
A
Zhang
 
DE
t(8;21)(q22;q22) Fusion proteins preferentially bind to duplicated AML1/RUNX1 DNA-binding sequences to differentially regulate gene expression.
Blood
2008
, vol. 
112
 
4
(pg. 
1392
-
1401
)
20
Liu
 
Y
Cheney
 
MD
Gaudet
 
JJ
, et al. 
The tetramer structure of the Nervy homology two domain, NHR2, is critical for AML1/ETO’s activity.
Cancer Cell
2006
, vol. 
9
 
4
(pg. 
249
-
260
)
21
Amann
 
JM
Nip
 
J
Strom
 
DK
, et al. 
ETO, a target of t(8;21) in acute leukemia, makes distinct contacts with multiple histone deacetylases and binds mSin3A through its oligomerization domain.
Mol Cell Biol
2001
, vol. 
21
 
19
(pg. 
6470
-
6483
)
22
Rosenbauer
 
F
Wagner
 
K
Kutok
 
JL
, et al. 
Acute myeloid leukemia induced by graded reduction of a lineage-specific transcription factor, PU.1.
Nat Genet
2004
, vol. 
36
 
6
(pg. 
624
-
630
)
23
Mullighan
 
CG
Zhang
 
J
Kasper
 
LH
, et al. 
CREBBP mutations in relapsed acute lymphoblastic leukaemia.
Nature
2011
, vol. 
471
 
7337
(pg. 
235
-
239
)
24
Pasqualucci
 
L
Dominguez-Sola
 
D
Chiarenza
 
A
, et al. 
Inactivating mutations of acetyltransferase genes in B-cell lymphoma.
Nature
2011
, vol. 
471
 
7337
(pg. 
189
-
195
)
25
Rhoades
 
KL
Hetherington
 
CJ
Harakawa
 
N
, et al. 
Analysis of the role of AML1-ETO in leukemogenesis, using an inducible transgenic mouse model.
Blood
2000
, vol. 
96
 
6
(pg. 
2108
-
2115
)
Sign in via your Institution