To the Editor:

p53 mutations have been detected in about 10% of all acute myeloid leukemia (AML) patients, mostly in patients with 17p monosomy.1-3 The scarcity of p53 mutations in AML could mean that, in the vast majority of AML patients, loss of p53 protein function is not required for the development of this disease. Alternatively, it is possible that inactivation of the p53 growth regulatory pathway is important and that this can occur either through disruption of downstream effector molecules or through epigenetic mechanisms that regulate p53 protein function. It has been suggested, for example, that inactivation of wild-type p53 protein in AML occurs through a mechanism involving conformational change of the protein4,5 or through binding to MDM2 protein.6-8 We have examined the functional status of the wild-type p53 protein expressed in cell lines derived from AML blasts on the basis of site-specific DNA binding activity, transactivation of p53-responsive genes, and ability to promote cell cycle arrest in G1 in response to γ-irradiation.9 The first two properties of p53 protein are strongly associated with its tumor suppressor function.10 11 

Nucleotide sequence analysis of the entire p53 coding region in four p53-expressing AML cell lines (OCI/AML-2, -3, -4, and -5)12 showed wild-type sequence. The site-specific DNA binding activity of p53 protein expressed in OCI/AML-3 and OCI/AML-5 cells was examined using an electrophoretic mobility shift assay (EMSA). Nuclear protein extracts were prepared from γ-irradiated or untreated cells and mixed with a 32P-labeled double-stranded oligonucleotide containing a p53 binding consensus sequence, p53CON.13 DNA damage increases the intracellular concentration of p53 protein and is also believed to activate the latent, sequence-specific DNA binding activity of p53. Whereas little, if any, DNA binding activity was detected in the nonirradiated extracts, the formation of a p53:DNA complex was evident when extracts were prepared from irradiated cells (Fig1). Inclusion of the p53-specific monoclonal antibody PAb421 in the binding reaction resulted in a supershifted p53:DNA complex and served to confirm the presence of p53 protein in the protein:DNA complex. DNA binding was not observed when an extract from the p53-negative cell line Lan1 was used in the EMSA.

Fig. 1.

DNA binding activity of p53 protein in AML cell lines. Nuclear extracts prepared from untreated or γ-irradiated OCI/AML-5 (A) and OCI/AML-3 (B) cells were incubated with a32P-labeled double-stranded oligonucleotide containing the p53 consensus sequence (p53CON) with (+) or without (−) the p53-specific monoclonal antibody PAb421 and analyzed by EMSA. Lan1 cells, which lack p53 protein, were used as a negative control. The OCI/AML-5 and Lan1 extracts were prepared 3 hours after γ-irradiation with a dose of 6 Gy. The OCI/AML-3 extracts were prepared at the times indicated after γ-irradiation with a dose of 2 Gy. The arrow labeled B points to the p53:DNA complex, and the arrow labeled A points to the supershifted antibody:p53:DNA complex.

Fig. 1.

DNA binding activity of p53 protein in AML cell lines. Nuclear extracts prepared from untreated or γ-irradiated OCI/AML-5 (A) and OCI/AML-3 (B) cells were incubated with a32P-labeled double-stranded oligonucleotide containing the p53 consensus sequence (p53CON) with (+) or without (−) the p53-specific monoclonal antibody PAb421 and analyzed by EMSA. Lan1 cells, which lack p53 protein, were used as a negative control. The OCI/AML-5 and Lan1 extracts were prepared 3 hours after γ-irradiation with a dose of 6 Gy. The OCI/AML-3 extracts were prepared at the times indicated after γ-irradiation with a dose of 2 Gy. The arrow labeled B points to the p53:DNA complex, and the arrow labeled A points to the supershifted antibody:p53:DNA complex.

Close modal

Activation of p21WAF1 gene transcription after γ-irradiation depends on wild-type p53 protein, and the p21WAF1 gene has been proposed to be a critical downstream effector in the p53-specific pathway of growth control in mammalian cells.14-17 Northern blot analysis (Fig2) indicated that the basal level of p21WAF1 mRNA was ninefold higher in OCI/AML-5 than in the mutant p53-expressing human erythroleukemia cell line OCIM2. Furthermore, 3.5 hours after irradiation with 6 Gy, p21WAF1mRNA levels increased ninefold in OCI/AML-5 and about threefold in OCIM2 cells. Irradiated OCI/AML-5 cells contained about 30-fold more p21WAF1 mRNA than did irradiated OCIM2 cells. No further increase in p21WAF1 mRNA levels was noted at later times after irradiation. p21WAF1 induction was also observed in irradiated OCI/AML-3 and OCI/AML-4 cell lines.18 The mRNA levels for GADD45 and MDM2, two other genes known to be transcriptionally regulated by p53 in response to DNA damage, also increased after γ-irradiation of AML cell lines (data not shown).

Fig. 2.

Expression of p21WAF1 mRNA in γ-irradiated OCI/AML-5 cells. Samples of total RNA (20 μg) prepared from cells at different times after exposure to 6 Gy of γ-irradiation were fractioned on an agarose-formaldehyde gel, transferred to a nylon membrane, and hybridized sequentially with 32P-labeled probes for human p21WAF1 cDNA (A) and 18S ribosomal RNA (B). OCIM2 cells, which express mutant p53 protein, were used as a control. Signal intensities were quantitated on a phosphorimager. The ratio of the p21WAF1 RNA signal to the 18S ribosomal RNA signal in the OCIM2 sample (0 hours) was arbitrarily set to 1.0 and the normalized values of p21WAF1 mRNA are shown at the bottom of (A).

Fig. 2.

Expression of p21WAF1 mRNA in γ-irradiated OCI/AML-5 cells. Samples of total RNA (20 μg) prepared from cells at different times after exposure to 6 Gy of γ-irradiation were fractioned on an agarose-formaldehyde gel, transferred to a nylon membrane, and hybridized sequentially with 32P-labeled probes for human p21WAF1 cDNA (A) and 18S ribosomal RNA (B). OCIM2 cells, which express mutant p53 protein, were used as a control. Signal intensities were quantitated on a phosphorimager. The ratio of the p21WAF1 RNA signal to the 18S ribosomal RNA signal in the OCIM2 sample (0 hours) was arbitrarily set to 1.0 and the normalized values of p21WAF1 mRNA are shown at the bottom of (A).

Close modal

OCI/AML-3, OCI/AML-5, and OCIM2 cells were irradiated with a dose of 6 Gy and cell proliferation was assessed 16 hours later by propidium iodide staining and flow cytometry. Both OCI/AML-3 and OCI/AML-5 cells were blocked in the G1 and G2 phases of the cell cycle with little, if any, cells present in S phase. In contrast, the mutant p53-expressing OCIM2 cells accumulated in G2 and showed no evidence of a block in G1 (Fig 3). The failure of OCIM2 cells to arrest in G1 after γ-irradiation suggests that the G1 cell cycle block observed in irradiated OCI/AML-3 and OCI/AML-5 cells is likely to be dependent on functional p53 protein. Irradiation-induced G1 arrest was confirmed by dual-parameter flow cytometry after pulse labeling cells with BrdU and staining for DNA content with propidium iodide and for BrdU incorporation with a fluorescein isothiocyanate (FITC)-conjugated antibody for BrdU. OCI/AML-3 and OCI/AML-5 showed a ninefold and sixfold increase in the G1:S ratio 16 hours after γ-irradiation (6 Gy), respectively. An increase in the G1:S ratio provides a good indicator of G1 delay.

Fig. 3.

Cell cycle changes in AML cells after exposure to γ-irradiation (6 Gy). The DNA content was determined by staining the cells with propidium iodide and the resulting profiles resulting from propidium iodide fluorescence are shown. For the irradiated cells, the cell cycle analyses were performed 16 hours after irradiation. OCIM2 cells were used as a control.

Fig. 3.

Cell cycle changes in AML cells after exposure to γ-irradiation (6 Gy). The DNA content was determined by staining the cells with propidium iodide and the resulting profiles resulting from propidium iodide fluorescence are shown. For the irradiated cells, the cell cycle analyses were performed 16 hours after irradiation. OCIM2 cells were used as a control.

Close modal

Our results indicate that p53 function (DNA binding, transactivation, and G1 checkpoint) is not lost during the development of AML or in the establishment of these AML cell lines. Functional p53 protein has also been demonstrated in human neuroblastoma,19 non-Hodgkin’s lymphoma,20 and even in certain HPV-positive cancer cell lines21 that contain wild-type p53 alleles. Hence, loss of p53 function or inactivation of the p53-dependent growth arrest pathway is not required for the development of certain malignancies, including AML.

Supported by grants from the Medical Research Council of Canada and the National Cancer Institute of Canada.

1
Lai
JL
Preudhomme
C
Zandecki
M
Flactif
M
Vanrumbeke
M
Lepelley
P
Wattel
E
Fenaux
P
Myelodysplastic syndromes and acute myeloid leukemia with 17p deletion. An entity characterized by specific dysgranulopoiesis and a high incidence of p53 mutation.
Leukemia
9
1995
370
2
Fenaux
P
Jonveaux
P
Quiquandon
I
Lai
JL
Pignon
JM
Loucheux-Lefebvre
MH
Bauters
F
Berger
R
Kerckaert
JP
p53 gene mutations in acute myeloid leukemia with 17p monosomy.
Blood
78
1991
1652
3
Soenen
V
Preudhomme
C
Rournier
C
Daudignon
A
Lai
JL
Fenaux
P
17p deletion in acute myeloid leukemia with myelodysplastic syndrome. Analysis of breakpoints and deleted segments by fluorescence in situ.
Blood
91
1998
1008
4
Zhu
YM
Bradbury
D
Russell
N
Expression of different conformations of p53 in the blast cells of acute myeloblastic leukaemia is related to in vitro growth characteristics.
Br J Cancer
68
1993
851
5
Zhang
W
Hu
G
Estey
E
Hester
J
Deisseroth
A
Altered conformation of the p53 protein in myeloid leukemia cells and mitogen-stimulated normal blood cells.
Oncogene
7
1992
1645
6
Bueso-Ramos
CE
Yang
Y
deLeon
E
McCowan
P
Stass
SA
Albitar
M
The human MDM-2 oncogene is overexpressed in leukemias.
Blood
82
1993
2617
7
Watanabe
T
Ichikawa
A
Saito
H
Hotta
T
Overexpression of the MDM2 oncogene in leukemia and lymphoma.
Leuk Lymphoma
21
1996
391
8
Seliger
B
Papadileris
S
Vogel
D
Hess
G
Brendel
C
Storkel
S
Ortel
J
Kolbe
K
Huber
C
Huhn
D
Neubauer
A
Analysis of the p53 and MDM-2 gene in acute myeloid leukemia.
Eur J Haematol
57
1996
230
9
Kuerbitz
SJ
Plunkett
BS
Walsh
WV
Kastan
MB
Wild-type p53 is a cell cycle checkpoint determinant following irradiation.
Proc Natl Acad Sci USA
89
1992
7491
10
Pietenpol
JA
Tokino
T
Thiagalingam
S
El-Deiry
WS
Kinzler
KW
Vogelstein
B
Sequence-specific transcriptional activation is essential for growth suppression by p53.
Proc Natl Acad Sci USA
91
1994
1998
11
Crook
T
Marston
NJ
Sara
EA
Vousden
KH
Transcriptional activation by p53 correlates with suppression of growth but not transformation.
Cell
79
1994
817
12
Yang
GS
Minden
MD
McCulloch
EA
Influence of schedule on regulated sensitivity of AML blasts to cytosine arabinoside.
Leukemia
7
1993
1012
13
Funk
WD
Pak
DT
Karas
RH
Wright
WE
Shay
JW
A transcriptionally active DNA-binding site for human p53 protein complexes.
Mol Cell Biol
12
1992
2866
14
Di Leonardo
A
Linke
SP
Clarkin
K
Wahl
GM
DNA damage triggers a prolonged p53-dependent G1 arrest and long-term induction of Cip1 in normal human fibroblasts.
Genes Dev
8
1994
2540
15
El-Deiry
WS
Harper
JW
O’Connor
PM
Velculescu
VE
Canman
CE
Jackman
J
Pietenpol
JA
Burrell
M
Hill
DE
Wang
Y
Wiman
KG
Mercer
WE
Kastan
MB
Kohn
KW
Elledge
SJ
Kinzler
KW
Vogelstein
B
WAF1/CIP1 is induced in p53-mediated G1 arrest and apoptosis.
Cancer Res
54
1994
1169
16
Dulic
V
Kaufmann
WK
Wilson
SJ
Tlsty
TD
Lees
E
Harper
JW
Elledge
SJ
Reed
SI
p53-dependent inhibition of cyclin-dependent kinase activities in human fibroblasts during radiation-induced G1 arrest.
Cell
76
1994
1013
17
Slebos
RJ
Lee
MH
Plunkett
BS
Kessis
TD
Williams
BO
Jacks
T
Hedrick
L
Kastan
MB
Cho
KR
p53-dependent G1 arrest involves pRB-related proteins and is disrupted by the human papillomavirus 16 E7 oncoprotein.
Proc Natl Acad Sci USA
91
1994
5320
18
Fu
L
Benchimol
S
Participation of the human p53 3′UTR in translational repression and activation following γ-irradiation.
EMBO J
16
1997
4117
19
Goldman
SC
Chen
CY
Lansing
TJ
Gilmer
TM
Kastan
MB
The p53 signal transduction pathway is intact in human neuroblastoma despite cytoplasmic localization.
Am J Pathol
148
1996
1381
20
Maestro
R
Gloghini
A
Doglioni
C
Piccinin
S
Vukosavljevic
T
Gasparotto
D
Carbone
A
Boiocchi
M
Human non-Hodgkin’s lymphomas overexpress a wild-type form of p53 which is a functional transcription activator of the cyclin-dependent kinase inhibitor p21.
Blood
89
1997
2523
21
Butz
K
Shahabeddin
L
Geisen
C
Spitkovsky
D
Ullmann
A
Hoppe-Seyler
F
Functional p53 protein in human papillomavirus-positive cancer cells.
Oncogene
10
1995
927
Sign in via your Institution