The relationship between differentiation of human myeloid cells and apoptosis remains unclear. Recent studies have shown that terminal differentiation need not necessarily lead to the apoptotic demise of myeloid cells, while other studies have shown that induction of differentiation is associated with increased resistance to apoptosis-inducing agents, such as chemotherapy and γ-irradiation. Such results are pertinent to the treatment of acute myeloid leukemia (AML) and myelodysplastic syndrome, where differentiating agents and hemopoietic growth factors are being combined with chemotherapy to enhance response and limit toxicity. To elucidate the factors governing apoptosis in human AML blasts, we have studied the cytotoxic effect of idarubicin on HL60, U937 and KG1 cells, after incubation with all-trans retinoic acid (ATRA), 1,25(OH)2 D3, and granulocyte-macrophage colony-stimulating factor (GM-CSF ). We show that prior incubation of human myeloid leukemic cells with ATRA or 1,25(OH)2 D3 induced resistance to idarubicin-induced apoptosis, which was modulated by coincubation with GM-CSF. The altered chemosensitivity of cells depended on the degree of G0/G1 cell-cycle arrest induced by incubation with ATRA, 1,25(OH)2 D3, and GM-CSF and was independent of differentiation status or Bcl-2 oncoprotein expression. These findings suggest that cell-cycle arrest in human leukemic cells can be induced by exogenous agents and may promote drug resistance. Determining the mechanisms by which cell-cycle arrest is induced may permit understanding of the processes by which the cells escape cytotoxic drug-mediated apoptosis.

ACUTE MYELOID leukemia (AML) remains incurable in the majority of patients, largely due to resistance to chemotherapy. Ten percent to 20% of patients have de novo refractory disease, while greater than 40% of those who attain remission subsequently relapse.1 The first described and best characterized mechanism of resistance is the mdr1 gene product, P-glycoprotein (Pgp). This molecule spans the cell membrane and acts as an efflux pump for toxins, including chemotherapy drugs such as anthracyclines, vinca alkaloids, and topoisomerase II inhibitors.2 Unfortunately, Pgp expression is often induced by chemotherapy and cross-resistance develops to drugs not previously used,3 which severely limits therapeutic options in refractory and relapsed cases of AML.4 Such “multiple drug resistance” (MDR) has now been described in relation to a large number of other mechanisms, involving Pgp-like membrane transporters, cytoplasmic detoxification enzymes and the cellular machinery for DNA repair and apoptosis (programmed cell death).5,6 Furthermore, redundancy is apparent amongst MDR mechanisms, which limits the benefits of MDR modulation therapy.2,7-9 

The clinical limitations of the chemotherapeutic approach in AML have led to the assessment of adjuvant therapies. The underlying biology, with a block in the normal myeloid differentiation program, has led to the use of differentiating agents, either alone, or in combination with chemotherapy.10,11 In vitro studies have shown that all-trans retinoic acid (ATRA) induces differentiation of human AML cell lines, with morphological and functional changes accompanied by a loss of proliferative capacity.12,13 Differentiation responses of blasts from AML patients have been restricted to those with acute promyelocytic leukemia (APL),14 while bone marrow samples from patients with myelodysplasia (MDS) have shown increased myeloid colony formation and the development of increasingly mature granulocytic cells when liquid marrow cultures are incubated with 13-cis retinoic acid.15 ATRA has proven effective in remission induction of APL, however, these remissions are not sustained and chemotherapy is required for durable remission.16,17 Where ATRA is effective, it acts by inducing apoptosis in the leukemic clone.18 

Although the active metabolite of vitamin D, 1,25 dihydroxyvitamin D3 (1,25(OH)2 D3) induces monocytic differentiation of HL60 and U937 cells, cell lines such as KG1 generally respond less well to 1,25(OH)2 D3.19 Bone marrow cells from patients with myeloid leukemia have again shown encouraging in vitro responses with monocytoid differentiation and a decrease in myelo/monoblasts induced by various vitamin D analogues.19 Sadly, such responses have not been translated into clinical benefit: in vivo use of 1,25(OH)2 D3 is associated with dose-limiting hypercalcemia,20 and studies combining retinoids and/or vitamin D analogs with low-dose chemotherapy in MDS have failed to produce useful responses in other than occasional cases.21 

The relationship between differentiation of human myeloid cells and apoptosis remains unclear. Terminal differentiation of myeloid cells can be associated with apoptosis; Martin et al have shown that HL60 cells cultured with ATRA differentiate and die by apoptosis after 7 to 10 days.22,23 However, two studies have shown that terminal differentiation need not necessarily lead to apoptotic demise of myeloid cells: HL60 cells transfected so as to constitutively overexpress Bcl-2 showed morphological, functional, and phenotypic evidence of differentiation when cultured with ATRA, but remained viable for up to 40 days, with no evidence of apoptosis.24,25 

It is not clear what effect differentiation-induction has on the sensitivity of leukemic cells to undergo apoptosis in response to cytotoxic agents, or indeed, whether there is a rationale for using differentiating agents in combination with chemotherapy. If the prevalence of spontaneous apoptosis increases as a result of maturation, then it is possible that sensitivity to cytotoxic drugs would also increase. Previous studies have shown that induction of differentiation is associated with increased resistance to agents such as etoposide, camptothecin, and azacytidine26-30; however, these studies provide conflicting data on the determination of such cytoprotection.

To elucidate the factors governing apoptosis in human AML blasts, we have studied the cytotoxic effect of idarubicin on the AML cell lines HL60, U937 and KG1, after incubation with ATRA, 1,25(OH)2 D3, and GM-CSF. Idarubicin was chosen as it is an anthracycline, which is extensively used in the treatment of AML31 and because it is less affected by Pgp expression than other anthracyclines.32,33 Idarubicin also has the advantage of being available for oral administration, which is often appropriate for the treatment of elderly patients with AML and MDS, as a part of combination treatment regimens.34,35 

We show that prior incubation of human myeloid leukemic cells with ATRA or 1,25(OH)2 D3 induced resistance to idarubicin-induced apoptosis, which was modulated by coincubation with GM-CSF. The altered chemosensitivity of cells depended not on differentiation or Bcl-2 oncoprotein expression, but rather on the degree of G0/G1 cell-cycle arrest induced by incubation with ATRA, 1,25(OH)2 D3, and GM-CSF. These findings suggest that differentiating agents may interact in vivo with cytotoxic drugs and hemopoietic growth factors to determine apoptotic cell-death of AML blasts. Such interactions may explain mechanisms by which AML blasts become constitutively resistant to chemotherapy, and thus help to develop treatment strategies to overcome such resistance.

Cells and reagents. HL60, U937, and KG1 cells were obtained from the European Collection of Animal Cell Cultures (Salisbury, UK). Reagents were purchased from Sigma Chemical Co (Poole, UK). Cells were cultured at 37°C with CO2 enriched to 5% in RPMI, supplemented with 10% fetal calf serum, 2 mmol/L glutamine, 100 U/mL penicillin, and 100 μg/mL streptomycin. ATRA (Sigma) and 1,25(OH)2 D3 (the kind gift of Dr Kay Colston, St Georges Hospital Medical School, London, UK) were dissolved in ethanol to achieve stock solutions of 1 mmol/L and 20 μmol/L, respectively; these were stored at −20°C until required. The final concentration of ethanol in cultures did not exceed 0.001 vol/vol. GM-CSF (Sandoz, Camberley, UK) was dissolved in sterile water and stored at −20°C until required.

Incubation of cells with ATRA, 1,25(OH)2 D3, and GM-CSF. Cells in exponential growth were seeded at 0.1 × 106/mL in 25-cm2 culture flasks (Costar, High Wycombe, UK) and supplemented with 1 μmol/L ATRA or 1 nmol/L 1,25(OH)2 D3 with or without 1,000 U/mL GM-CSF. Cells were harvested after 72 hours, washed, and used for assay of idarubicin-induced apoptosis (see below); cell proliferation was assessed by counting in a Neubauer hemocytometer (Weber Scientific International, London, UK). Aliquots were analyzed for cell-cycle profile, phenotype and Bcl-2 expression.

Analysis of cell-cycle profile, phenotype and Bcl-2 expression. Cell-cycle profiles were measured by flow cytometry. Washed, pelleted cells were prepared using a Coulter DNA-Prep (Coulter Electronics, Luton, UK), which uses a nonionic detergent permeabilization stage followed by propidium iodide/RNase staining of DNA. Cells were analyzed 1 hour after processing using an EPICS Elite cytometer (Coulter Electronics). Initial gating to exclude doublets was done using peak versus integral red channel signal, such that control HL60 cells gave 2% to 5% apoptosis, which level is observed morphologically on cytospin preparations. G0/G1, S, and G2/M fractions were identified from the resulting cell-cycle profile and apoptosis was measured from the sub-G1 fraction.36 

Surface expression of the maturation antigens CD11b, CD13, CD14, CD32, and CD33 was measured using an EPICS Elite cytometer with the following conjugated primary antibodies: CD11b/phycoerythrin and CD14/phycoerythrin (Becton Dickinson, Oxford, UK); CD13/phycoerythrin and CD33/fluorescein isothiocyanate (Coulter); CD32/unconjugated (Immunotec, Marseille, France); rabbit antimouse/fluorescein isothiocyanate (Dako, High Wycombe, UK); appropriate isotype controls were from the same source as primary antibodies.

Bcl-2 protein expression was measured using a modification of the method of Delia et al37; briefly, paired tubes of washed, pelleted cells were fixed on ice with fresh 2% paraformaldehyde (10 minutes) and permeabilized with 0.05% Triton-X (10 minutes; Sigma). After two washes, one tube of each pair was incubated with anti-Bcl-2/fluorescein isothiocyanate conjugated antibody and the other with an isotype-matched control antibody. Percentage expression and mean fluorescence index were measured for the anti-Bcl-2 relative to the control for each cell type/treatment group.

Drug-induced apoptosis. After incubation with ATRA, 1,25(OH)2 D3, and GM-CSF, washed cells were incubated with idarubicin (Pharmacia, Milton Keynes, UK) at concentrations of 2,10, and 50 μg/L in 12-well plates (Costar, High Wycombe, UK); cells were seeded at 0.2 × 106/mL in 3-mL volumes and plates included control wells without idarubicin. Drug concentrations were chosen to reflect in vivo plasma levels as closely as possible: 50 μg/L corresponds to plasma levels achieved with standard doses of intravenous idarubicin, while oral dosing typically achieves levels of 2 to 10 μg/L.38 Cells were harvested after 24 hours and prepared using the DNA-Prep (as above); apoptosis was measured from the sub-G1 fraction of the resulting cell-cycle profile. Cell death was confirmed by loss of the ability to exclude trypan blue; the apoptotic nature of cell death was confirmed morphologically and by DNA fragmentation using agarose gel electrophoresis.39 

Statistical analysis. Results were analysed using paired t-test, ANOVA and Spearman Rank correlation by means of Statistica (StatSoft, Tulsa, OK) and STATA (Stata Corp, College Station, TX) software.

Differentiation status after incubation with ATRA, 1,25(OH)2 D3, and GM-CSF. 1,25(OH)2 D3 induced monocytic differentiation in the U937 cell line as documented by an increase in expression of CD14 (P < .05, Table 1). 1,25(OH)2 D3 reduced the expression of CD33 in HL60 cells (P < .05) and increased the expression of CD11b in the KG1 cell line (P < .001, Table 1). ATRA induced phenotypic evidence of granulocytic differentiation in the HL60 and KG1 cell lines, as indicated by a reduction in CD13 expression (each P < .01), and induced the expression of CD11b in U937 and KG1 cells (P < .05 and P < .01, respectively). GM-CSF had little effect on the differentiation status of HL60 and KG1 cells, either alone or in combination with ATRA or 1,25(OH)2 D3. However, GM-CSF interacted with both ATRA and 1,25(OH)2 D3 to increase monocytic differentiation of U937 in a manner that was at least additive; expression of the CD14 and CD32 antigens increased in cultures coincubated with GM-CSF (CD14, ATRA, and 1,25(OH)2 D3, P < .001; CD32, ATRA, and 1,25(OH)2 D3, P < .05, Table 1).

Table 1.

Flow-Cytometric Determination of Differentiation of HL60, U937, and KG1 Induced by 72-Hours Incubation With ATRA, 1,25(OH)2 D3, and GM-CSF

CellAntigenControlGM-CSFATRAATRA + GM-CSF1,25(OH)2 D31,25(OH)2 D3 + GM-CSF
HL60 CD13 99.88 (0.1) 99.87 (0.1) 99.95 (0.1) 99.93 (0.1) 99.90 (0.1) 99.93 (0.1) 
  85.53 (19.6) 76.13 (12.9) 37.13 (8.7)* 42.90 (15.0) 65.63 (20.2) 66.97 (23.6) 
HL60 CD33 86.87 (9.0) 94.35 (2.9) 69.40 (11.8) 71.2 (13.6) 65.77 (3.7) 66.15 (13.6) 
  6.16 (2.4) 7.51 (1.3) 4.43 (1.6) 5.41 (0.5) 4.81 (2.1)* 6.06 (1.1) 
U937 CD11b 2.80 (0.7) 5.07 (2.2) 14.57 (4.7)* 19.65 (10.2) 9.27 (5.9) 46.20 (22.1) 
  3.49 (0.6) 4.36 (1.3) 3.14 (0.7) 1.83 (0.1) 4.64 (1.1) 3.51 (0.5) 
U937 CD14 1.44 (0.4) 11.98 (5.7)* 1.80 (1.7) 39.18 (8.5) 35.04 (19.9)* 90.34 (18.1) 
  2.87 (0.7) 2.25 (2.5) 1.72 (0.7) 2.98 (1.4) 51.48 (10.8) 
U937 CD32 23.98 (2.4) 29.06 (12.7) 8.56 (7.0) 53.76 (20.6) 22.22 (14.1) 54.88 (13.9) 
  5.26 (2.8) 6.86 (3.4)* 6.62 (4.4) 5.32 (3.7) 5.99 (3.1) 7.23 (4.7) 
KG1 CD11b 32.62 (5.9) 33.77 (9.3) 70.46 (11.5)* 90.37 (3.0) 85.30 (8.6)* 95.20 (1.2) 
  4.87 (1.1) 4.50 (0.8) 4.95 (1.5) 4.72 (0.4) 9.63 (2.0)* 10.08 (2.2) 
KG1 CD13 99.83 (0.1) 100.00 (0) 99.92 (0.1) 99.93 (0.1) 99.87 (0.1) 99.90 (0.1) 
  81.12 (47.7) 139.57 (48.6) 38.98 (16.0)* 44.93 (9.2) 59.13 (29.6) 81.83 (18.8) 
KG1 CD33 53.90 (15.5) 50.45 (12.9) 68.28 (7.9) 26.95 (3.0) 26.35 (14.7) 28.20 (12.6) 
  4.45 (1.1) 4.02 (1.1) 2.93 (1.0)* 3.02 (1.1) 3.96 (1.6) 3.18 (1.6) 
CellAntigenControlGM-CSFATRAATRA + GM-CSF1,25(OH)2 D31,25(OH)2 D3 + GM-CSF
HL60 CD13 99.88 (0.1) 99.87 (0.1) 99.95 (0.1) 99.93 (0.1) 99.90 (0.1) 99.93 (0.1) 
  85.53 (19.6) 76.13 (12.9) 37.13 (8.7)* 42.90 (15.0) 65.63 (20.2) 66.97 (23.6) 
HL60 CD33 86.87 (9.0) 94.35 (2.9) 69.40 (11.8) 71.2 (13.6) 65.77 (3.7) 66.15 (13.6) 
  6.16 (2.4) 7.51 (1.3) 4.43 (1.6) 5.41 (0.5) 4.81 (2.1)* 6.06 (1.1) 
U937 CD11b 2.80 (0.7) 5.07 (2.2) 14.57 (4.7)* 19.65 (10.2) 9.27 (5.9) 46.20 (22.1) 
  3.49 (0.6) 4.36 (1.3) 3.14 (0.7) 1.83 (0.1) 4.64 (1.1) 3.51 (0.5) 
U937 CD14 1.44 (0.4) 11.98 (5.7)* 1.80 (1.7) 39.18 (8.5) 35.04 (19.9)* 90.34 (18.1) 
  2.87 (0.7) 2.25 (2.5) 1.72 (0.7) 2.98 (1.4) 51.48 (10.8) 
U937 CD32 23.98 (2.4) 29.06 (12.7) 8.56 (7.0) 53.76 (20.6) 22.22 (14.1) 54.88 (13.9) 
  5.26 (2.8) 6.86 (3.4)* 6.62 (4.4) 5.32 (3.7) 5.99 (3.1) 7.23 (4.7) 
KG1 CD11b 32.62 (5.9) 33.77 (9.3) 70.46 (11.5)* 90.37 (3.0) 85.30 (8.6)* 95.20 (1.2) 
  4.87 (1.1) 4.50 (0.8) 4.95 (1.5) 4.72 (0.4) 9.63 (2.0)* 10.08 (2.2) 
KG1 CD13 99.83 (0.1) 100.00 (0) 99.92 (0.1) 99.93 (0.1) 99.87 (0.1) 99.90 (0.1) 
  81.12 (47.7) 139.57 (48.6) 38.98 (16.0)* 44.93 (9.2) 59.13 (29.6) 81.83 (18.8) 
KG1 CD33 53.90 (15.5) 50.45 (12.9) 68.28 (7.9) 26.95 (3.0) 26.35 (14.7) 28.20 (12.6) 
  4.45 (1.1) 4.02 (1.1) 2.93 (1.0)* 3.02 (1.1) 3.96 (1.6) 3.18 (1.6) 

Upper row: mean % expn (SD); lower row: mean MFI (SD).

*

Result statistically different from control, for P values see text.

Result statistically different from ATRA/1,25(OH)2 D3 alone, eg, ATRA versus ATRA + GM-CSF, for P values see text.

Growth and cell-cycle profile after incubation with ATRA, 1,25(OH)2 D3, and GM-CSF. 1,25(OH)2 D3 inhibited proliferation of KG1 cells (P < .05, Fig 1), with a trend towards a similar effect in U937 and HL60. In contrast, ATRA inhibited the proliferation of U937 and HL60 cell lines (U937, P < .01; HL60 P < .01), with a trend towards an inhibitory response in KG1. GM-CSF had no effect on cellular proliferation when used alone, however, coincubation with ATRA synergistically inhibited the proliferation of U937 cells (P < .05). GM-CSF also reversed the growth inhibitory effect of 1,25(OH)2 D3 on KG1 cells (P < .05). In general, growth inhibition was associated with phenotypic evidence of differentiation; however, U937 cells underwent differentiation in response to 1 nmol/L 1,25(OH)2 D3 without growth inhibition.

Fig. 1.

Growth inhibition of HL60, U937, and KG1 cells after 72-hours incubation with ATRA, 1,25(OH)2 D3, and GM-CSF. Results shown are mean of n = 3 (HL60 & KG1) or n = 7 (U937) experiments; error bars omitted for clarity. Statistical significance: * result statistically different from control; # result statistically different from ATRA/1,25(OH)2 D3 alone, eg, ATRA versus ATRA+GM-CSF; for P values see text.

Fig. 1.

Growth inhibition of HL60, U937, and KG1 cells after 72-hours incubation with ATRA, 1,25(OH)2 D3, and GM-CSF. Results shown are mean of n = 3 (HL60 & KG1) or n = 7 (U937) experiments; error bars omitted for clarity. Statistical significance: * result statistically different from control; # result statistically different from ATRA/1,25(OH)2 D3 alone, eg, ATRA versus ATRA+GM-CSF; for P values see text.

Close modal

ATRA and 1,25(OH)2 D3 induced significant G0/G1 cell-cycle arrest in all three cell lines (Fig 2), the degree of G0/G1 arrest induced by ATRA being greater than that by 1,25(OH)2 D3 in each case. GM-CSF alone had no effect on cell-cycle status in HL60 and U937, but reduced the proportion of KG1 cells in G0/G1 (P < .05). Coincubation with GM-CSF increased the proportion of U937 cells arrested by 1,25(OH)2 D3 (P < .01). A trend towards a similar interaction between ATRA and GM-CSF (Fig 2) was observed.

Fig. 2.

G0/G1 arrest of HL60, U937, and KG1 cells after 72-hours incubation with ATRA, 1,25(OH)2 D3, and GM-CSF. Results shown are mean of n = 3 (HL60 & KG1) or n = 7 (U937) experiments; error bars omitted for clarity. Statistical significance: * result statistically different from control; # result statistically different from ATRA/1,25(OH)2 D3 alone, eg, ATRA versus ATRA+GM-CSF; for P values see text.

Fig. 2.

G0/G1 arrest of HL60, U937, and KG1 cells after 72-hours incubation with ATRA, 1,25(OH)2 D3, and GM-CSF. Results shown are mean of n = 3 (HL60 & KG1) or n = 7 (U937) experiments; error bars omitted for clarity. Statistical significance: * result statistically different from control; # result statistically different from ATRA/1,25(OH)2 D3 alone, eg, ATRA versus ATRA+GM-CSF; for P values see text.

Close modal

Idarubicin-induced apoptosis. Untreated control cultures in logarithmic growth demonstrated no significant apoptosis (<5% of cells) (Fig 3A). Idarubicin induced dose-dependent apoptotic cell death in all three cell lines after 24-hours exposure at concentrations of 2, 10, and 50 μg/L (data not shown). Cell death was confirmed by loss of the ability to exclude trypan blue; apoptosis was demonstrated by light microscopy and by agarose gel electrophoresis of DNA (data not shown). Apoptosis was quantitated by flow cytometry as described in Materials and Methods.

Fig. 3.

Apoptosis in HL60, U937, and KG1 cells incubated with ATRA, 1,25(OH)2 D3, and GM-CSF for 72 hours and then exposed to idarubicin 50 μg/L for 24 hours. (A) Apoptosis induced by idarubicin 50 μg/L. Results shown are mean of n = 3 (HL60 & KG1) or n = 7 (U937) experiments; error bars omitted for clarity. Statistical significance: * result statistically different from control; # result statistically different from ATRA/1,25(OH)2 D3 alone, eg, ATRA versus ATRA+GM-CSF; for P values see Table 2. (B) Representative cell-cycle profiles for U937 cells incubated with ATRA and ATRA + GM-CSF, and then exposed to idarubicin 50 μg/L; see Materials and Methods for details: X-axis, red fluorescence, Y-axis, cell number; left-hand plots, after initial incubation (ATRA ± GM-CSF, 72 hours), right-hand plots, after exposure to idarubicin (24 hours); gate B denotes apoptotic fraction, C cells in G0/G1, E S phase, and F G2/M.

Fig. 3.

Apoptosis in HL60, U937, and KG1 cells incubated with ATRA, 1,25(OH)2 D3, and GM-CSF for 72 hours and then exposed to idarubicin 50 μg/L for 24 hours. (A) Apoptosis induced by idarubicin 50 μg/L. Results shown are mean of n = 3 (HL60 & KG1) or n = 7 (U937) experiments; error bars omitted for clarity. Statistical significance: * result statistically different from control; # result statistically different from ATRA/1,25(OH)2 D3 alone, eg, ATRA versus ATRA+GM-CSF; for P values see Table 2. (B) Representative cell-cycle profiles for U937 cells incubated with ATRA and ATRA + GM-CSF, and then exposed to idarubicin 50 μg/L; see Materials and Methods for details: X-axis, red fluorescence, Y-axis, cell number; left-hand plots, after initial incubation (ATRA ± GM-CSF, 72 hours), right-hand plots, after exposure to idarubicin (24 hours); gate B denotes apoptotic fraction, C cells in G0/G1, E S phase, and F G2/M.

Close modal

Prior incubation of leukemic cells with either ATRA or 1,25(OH)2 D3 significantly inhibited the ability of idarubicin to induce apoptosis (Table 2, Fig 3A). GM-CSF had a protective effect on U937 cells exposed to idarubicin and interacted with ATRA and 1,25(OH)2 D3 to further increase the resistance of U937 to idarubicin-induced apoptosis (Fig 3B). By contrast, GM-CSF sensitized both HL60 and KG1 cells to the effects of idarubicin, and partially reversed the protective effect of ATRA and 1,25(OH)2 D3 incubation on KG1 cells.

Table 2.

Apoptosis Induced by Idarubicin 50 μg/L After 24 hours: Mean % Cells Apoptotic (SD)

Cell Line/TreatmentControlGM-CSFATRAATRA + GM-CSF1,25(OH)2 D31,25(OH)2 D3 + GM-CSF
HL60 n = 3 30.2 (9.5) 33.3 (12.3) 7.0 (1.2) 9.3 (1.6) 10.2 (5.1) 10.4 (4.7) 
U937 n = 7 27.9 (12.0) 21.6 (6.6) 13.1 (4.8) 4.2 (2.8) 21.9 (8.2) 9.2 (4.4) 
KG1 n = 3 30.1 (4.6) 32.0 (8.0) 8.3 (4.7) 15.1 (4.0) 11.8 (6.1) 16.7 (7.0) 
Cell Line/TreatmentControlGM-CSFATRAATRA + GM-CSF1,25(OH)2 D31,25(OH)2 D3 + GM-CSF
HL60 n = 3 30.2 (9.5) 33.3 (12.3) 7.0 (1.2) 9.3 (1.6) 10.2 (5.1) 10.4 (4.7) 
U937 n = 7 27.9 (12.0) 21.6 (6.6) 13.1 (4.8) 4.2 (2.8) 21.9 (8.2) 9.2 (4.4) 
KG1 n = 3 30.1 (4.6) 32.0 (8.0) 8.3 (4.7) 15.1 (4.0) 11.8 (6.1) 16.7 (7.0) 

ANOVA P values for ATRA, 1,25(OH)2 D3 and GM-CSF effects: ATRA-HL60 P = .02, U937 P < .001, KG1 P < .01; 1,25(OH)2 D3-HL60 P = .02, U937 P < .01, KG1 P < .01; GM-CSF-HL60 P = .02, U937 P < .001, KG1 P < .001.

Bcl-2 protein expression. Expression of the Bcl-2 oncoprotein has been shown to be associated with the ability of some hemopoietic cells to resist the induction of apoptosis in certain circumstances. We therefore quantitated the expression of Bcl-2 protein in leukemic cells following incubation with ATRA, 1,25(OH)2 D3, and GM-CSF to establish whether upregulation of Bcl-2 was responsible for the modulation of cytotoxicity which we observed. ATRA, but not 1,25(OH)2 D3 or GM-CSF, reduced Bcl-2 expression by HL60 cells (P < .05, Fig 4A). Neither ATRA, 1,25(OH)2 D3, nor GM-CSF, either alone or in combination, had any significant effect on Bcl-2 protein expression by KG1 cells (Fig 4B). Both ATRA and GM-CSF induced statistically significant reductions in Bcl-2 expression in U937 cells (ATRA P < .01, GM-CSF P < .05, Fig 4c), and the combination of ATRA with GM-CSF reduced Bcl-2 expression still further (P < .01).

Fig. 4.

Bcl-2 protein expression of HL60, U937, and KG1 cells after 72-hours incubation with ATRA, 1,25(OH)2 D3, and GM-CSF. (A) HL60; (B) KG1; (C) U937. Results shown are mean of n = 3 (HL60 & KG1) or n = 7 (U937) experiments; error bars represent 1 SD above the mean. Statistical significance: * result statistically different from control; # result statistically different from ATRA/1,25(OH)2 D3 alone, eg, ATRA versus ATRA + GM-CSF; for P values see text.

Fig. 4.

Bcl-2 protein expression of HL60, U937, and KG1 cells after 72-hours incubation with ATRA, 1,25(OH)2 D3, and GM-CSF. (A) HL60; (B) KG1; (C) U937. Results shown are mean of n = 3 (HL60 & KG1) or n = 7 (U937) experiments; error bars represent 1 SD above the mean. Statistical significance: * result statistically different from control; # result statistically different from ATRA/1,25(OH)2 D3 alone, eg, ATRA versus ATRA + GM-CSF; for P values see text.

Close modal

Correlation of G0/G1 arrest and apoptosis. It is clear from the data presented that susceptibility to idarubicin-induced apoptosis was not related to the differentiation status of cells following incubation with ATRA, 1,25(OH)2 D3, and GM-CSF: 1,25(OH)2 D3 induced greater differentiation of U937 than ATRA, yet ATRA afforded a greater protective effect, while both ATRA and 1,25(OH)2 D3 induced little differentiation of KG1, but both afforded protection. Levels of the Bcl-2 oncoprotein are also unlikely to explain the altered susceptibility of these cells to undergo apoptosis: over the period of incubation, Bcl-2 expression in U937 was downregulated by both ATRA and 1,25(OH)2 D3, while ATRA downregulated expression in HL60, with a trend towards reduced expression in KG1. These changes are consistent with previous observations associating differentiation with downregulation of Bcl-2 expression; however, such downregulation of expression should have increased susceptibility to apoptosis, rather than the reverse, which we observed.

Induction of apoptosis by idarubicin was, however, related to the proportion of cells induced to undergo G0/G1 arrest during incubation. Using Spearman Rank correlation analysis, a negative correlation was observed between the percentage of apoptotic cells after exposure to 50 μg/L idarubicin and the proportion of cells in G0/G1 after incubation with ATRA, 1,25(OH)2 D3, and GM-CSF (rho = −0.9092, P < .001).

We have confirmed that prior incubation with ATRA and 1,25(OH)2 D3 protects HL60 cells against drug-induced apoptosis using the anthracycline antibiotic idarubicin, and have demonstrated a similar response in the myeloid leukemic cell lines U937 and KG1, which are less differentiated than HL60.40,41 Several recent studies have shown that HL60 cells have reduced sensitivity to a range of apoptotic stimuli after induction of differentiation26-30; however, there are conflicting data regarding the mechanisms underlying this effect. Zwelling et al30 have shown a 10-fold reduction in etoposide-induced DNA damage after incubation of HL60 cells with PMA; concordant results using isolated nuclei suggested that the protective effect resided in the cell nucleus. By contrast, Solary et al29 have shown an apparent cytoplasmic localization of TPA-induced protection against a range of topoisomerase inhibitors.

We have sought to clarify the relative roles of the nuclear and cytoplasmic consequences of differentiation by comparing the effects of ATRA and 1,25(OH)2 D3 on cell-cycle distribution, Bcl-2 protein expression and immunophenotype of myeloid leukemia cells before their exposure to idarubicin. We show that neither the degree of immunophenotypic differentiation, nor the level of Bcl-2 expression, explain the observed protection of cells against idarubicin-induced apoptosis. By contrast, the degree to which cells were protected was inversely proportional to the extent to which ATRA and 1,25(OH)2 D3 induced G0/G1 cell-cycle arrest.

In this study leukemic cells underwent differentiation and growth arrest in response to 1,25(OH)2 D3 and ATRA; differentiation of U937 cells was further augmented by GM-CSF as previously described.42,43 However, resistance to idarubicin was independent of differentiation per se as there appeared to be discordance between the degree of differentiation observed by phenotypic assessment and idarubicin-associated apoptosis; indeed, ATRA alone conferred greater protection upon U937 cells without inducing detectable phenotypic maturation.

Our observations concerning Bcl-2 expression accord with previous studies that have shown downregulation of Bcl-2 protein in myeloid cells exposed to differentiating agents.26,27,37 However, these studies have shown a paradoxical decrease in drug-induced apoptosis when the reverse would be expected.26,27 It is now clear that protection from a range of apoptotic stimuli is determined not simply by Bcl-2 expression per se but by heterodimers of Bcl-2 and its homologues. Bcl-2 and Bcl-XL act as suppressors of cell death, while Bax and Bcl-XS promote induction of apoptosis.44 Although other members of the Bcl-2 family were not studied here, Sanz et al45 have shown only low-level Bcl-XL expression in parental HL60 cells, with downregulation in response to ATRA-induced differentiation. Nevertheless, it is clear that protection from idarubicin-induced apoptosis conferred by differentiating agents was not mediated by upregulation of Bcl-2 protein expression.

An alternative mechanism of differentiation-induced cytoprotection might involve factors that determine an MDR phenotype, such as Pgp. Upregulation of Pgp in association with differentiation is unlikely to be responsible for the increase in resistance to idarubicin: Xu et al26 and Del Bino et al28 attempted to induce apoptosis with cytarabine, which is not a substrate for Pgp, and with noncytotoxic inducers of apoptosis (calcium ionophore, hyperthermia, and γ-irradiation) and observed a similar protective effect after differentiation. We have confirmed these findings using cytarabine (Ketley et al, unpublished observations, March 1995). Parental HL60 cells have been shown not to express Pgp9 and preliminary data suggest that, although Pgp expression may be differentiation-dependent in K562 cells, differentiation of HL60 and U937 cells does not induce Pgp expression (X.R. Jiang personal communication, July 1995). In addition, idarubicin is not thought to be a substrate for Pgp at physiological levels of expression.32,33 

Our data strongly suggest an association between resistance to idarubicin-induced apoptosis and arrest of cell cycling in the G1 phase. This is inferred by three lines of evidence. Firstly, KG1 cells showed limited evidence of differentiation in response to ATRA or 1,25(OH)2 D3, but accumulated in G1 in response to these agents and became resistant to idarubicin. Secondly, there was a strong correlation between G1 arrest in the U937 and HL60 cell lines and protection from idarubicin. Thirdly, GM-CSF was able to alter the sensitivity of both KG1 and U937 cells to idarubicin-induced cell death after incubation with ATRA and 1,25(OH)2 D3, an effect again predicted by the ability of GM-CSF to modulate the induction of G1 arrest.

The relationship between differentiation and G1 cell-cycle arrest is crucial to understanding how the cytoprotection we have observed might be mediated. Bergh et al46 have recently shown that G1 arrest is necessary but not sufficient for differentiation. Using U937 cells transfected with an antisense oligonucleotide (ASO) to the retinoblastoma gene, they showed that ATRA and 1,25(OH)2 D3 inhibit proliferation and induce G1 arrest as in control cells, but without morphological, phenotypic, or functional evidence of differentiation.46 A potential candidate for mediating the G1 arrest seen with differentiation of leukemia cells is p21, and recent data suggest that this cyclin-dependent kinase inhibitor (CKI) may have a role in the modulation of drug-induced apoptosis and DNA repair.47-49 

The classical pathway of p21 induction is following DNA damage, which causes upregulation of p53, a transcription factor for the p21 gene.50 p21 inhibits cyclin-dependent kinases (CDKs) and hence CDK-dependent phosphorylation, thereby maintaining the retinoblastoma protein in a hypophosphorylated state, which induces G1 arrest and growth suppression in vitro (to almost the same extent as transfected p53).50-52 p21 can also be induced independently of p53, notably in association with cellular differentiation53-55: HL60 cells show p21 activation after treatment with a range of differentiating agents, including ATRA, with p21 mRNA appearing at 24 hours, before any phenotypic or cell-cycle evidence of differentiation.53 

The specificity of p21 mediation of differentiation-induced G1 arrest is supported by the finding of a vitamin D response element in the promoter region of the p21 gene and the demonstration that p21 transfection induces phenotypic differentiation, although not to the extent seen with 1,25(OH)2 D3 itself.56 Furthermore, transfection of p21 ASO is associated with decreased PMA-induced cell-cycle arrest and phenotypic differentiation, as well as an attenuation of PMA-induced inhibition of clonogenic growth.57 The relevance of these in vitro findings to the clinic is provided by Zhang et al,58 who have shown that p21 is an independent risk factor in untreated AML.58 Although the p21 protein expression was heterogeneous amongst the 100 patients studied, high levels were associated with a lower remission rate, which the authors postulated was due to quiescence-associated chemoresistance.58 

Such chemoresistance might arise in two main ways: either G1-arrested cells are damaged less by a given dose and duration of chemotherapy, or they are better at repairing that damage before it translates into apoptosis, perhaps simply by having longer in which to do so. Differentiation itself is associated with an increase in DNA strand breaks,59 as well as induction of the repair enzyme poly-ADP-ribose polymerase (PARP).60 PARP seems to act as a “molecular nick sensor,” marking sections of DNA for repair,61 but is itself a target for proteolytic cleavage during apoptosis.62 

Again, recent evidence supports a role for p21 in DNA repair following drug-induced damage. Accurate DNA repair requires the suppression of DNA replication, a balance coordinated by proliferating cell nuclear antigen (PCNA).63 Warbrick et al49 have recently demonstrated that p21 has a PCNA binding sequence, and that the formation of p21/PCNA complexes favors DNA repair over replication. The impact of p21 status on cell survival is shown by two studies: Sheikh et al48 have shown increased clonogenic survival and DNA repair of p21-transfected colorectal carcinoma cells exposed to UV irradiation,48 while Fan et al47 have shown that disruption of p21 function is associated with sensitization to cisplatinum-induced cytotoxicity and cell-cycle delay, as well as decreased DNA repair.

Although p21 is a candidate for mediating the differentiation-induced cytoprotection, which we have described, the clinical implications of our findings are complex. The finding by Zhang et al58 of an association between p21 expression and clinical remission in de novo AML suggests that quiescence of tumor cells may be a potent mechanism of chemoresistance. However, they and others have failed to detect mutations or deletions of the p21 gene in leukemia samples, suggesting that this CKI may have a pivotal role in determining tumor therapy responses.58,64 However, despite in vitro evidence for reduced drug-induced cell death after ATRA,27,65 the accumulating clinical evidence from APL trials is of greater remission induction and reduced relapse with ATRA plus chemotherapy as opposed to either modality alone.66 

Although there is little clinical data to support the potential for hemopoietic growth factors to recruit cells into cycle and increase their chemosensitivity,67,68 it may be that cell-cycle quiescence is associated with chemoresistance. Constitutive changes or external stimuli that induce cell-cycle arrest in leukemic cells may substantially increase the resistance of these cells to chemotherapy-induced apoptosis. Our data suggest that further elucidation of the mechanisms underlying differentiation-induced cytoprotection will enable novel therapeutic strategies to be developed. Furthermore, if tumor cell quiescence can be accurately assessed at presentation, stratification of chemotherapy based on predicted response may become more sophisticated. The studies of Banker et al69 where multiparametric analyses of cell-cycle characteristics and apoptosis were able to sensitively and reproducibly detect the effects of apoptosis modulators on individual patient samples may be important in this respect.

Supported by the Leukaemia Research Fund (London, UK) as a Clinical Training Fellow (to N.J.K.).

Address reprint requests to Nicolas J. Ketley, MRCP, Department of Haematology, Great Ormond Street Hospital for Children, Great Ormond St, London, WC1N 3JH, United Kingdom.

1
Greer JP, Kinney MC: Acute nonlymphocytic leukemia, in Lee GR, Bithell TC, Foerster J, Athens JW, Lukens JN (eds): Wintrobe's Clinical Hematology. Philadelphia, PA, Lea & Febiger, 1993, p 1920
2
Pinedo
HM
Giaccone
G
P-glycoprotein–a marker of cancer-cell behavior.
N Engl J Med
333
1995
1417
3
Marie J: P-glycoprotein in adult hematologic malignancies, in Fisher GA, Sikic BI (eds): Drug Resistance in Clinical Oncology and Hematology. Philadelphia, PA, Saunders, 1995, p 239
4
Nussler V, Pelka-Fleischer R, Zwiernina H, Nerl C, Beckert B, Gieseler F, Diem H, Ledderose G, Gullis E, Sauer H, Wilmanns W: P-glycoprotein expression in patients with acute leukemia–clinical relevance. Leukemia 10:S23, 1996 (suppl)
5
Filipits M, Suchomel RW, Zochbauer S, Malayeri R, Pirker R: Clinical relevance of drug resistance genes in malignant diseases. Leukemia 10:S10, 1996
6
Reed JC: BCL-2: Prevention of apoptosis as a mechanism of drug resistance, in Fisher GA, Sikic BI (eds): Drug Resistance in Clinical Oncology and Hematology. Philadelphia, PA, Saunders, 1995, p 451
7
Deffie
AM
Alam
T
Seneviratne
C
Beenken
SW
Batra
JK
Shea
TC
Henner
WD
Goldenberg
GJ
Multifactorial resistance to adriamycin: Relationship of DNA repair, glutathione transferase activity, drug efflux, and P-glycoprotein in cloned cell lines of adriamycin-sensitive and -resistant P388 leukemia.
Cancer Res
48
1988
3595
8
List
AF
Role of multidrug resistance and its pharmacological modulation in acute myeloid leukemia.
Leukemia
10
1996
937
9
Huang
Y
Ibrado
AM
Reed
JC
Bullock
G
Ray
S
Tang
C
Bhalla
K
Co-expression of several molecular mechanisms of multidrug resistance and their significance for paclitaxel cytotoxicity in human AML HL-60 cells.
Leukemia
11
1997
253
10
Koeffler
HP
Induction of differentiation of human acute myelogenous leukemia cells: Therapeutic implications.
Blood
62
1983
709
11
Tallman
MS
Differentiating therapy in acute myeloid leukemia.
Leukemia
10
1996
1262
12
Breitman
TR
Selonick
SE
Collins
SJ
Induction of differentiation of the human promyelocytic leukemia cell line (HL60) by retinoic acid.
Proc Natl Acad Sci USA
77
1980
2936
13
Olsson
IL
Breitman
TR
Induction of differentiation of the human histiocytic lymphoma cell line U937 by retinoic acid and cyclic adenosine 3′:5′-monophosphate-inducing agents.
Cancer Res
42
1982
3924
14
Breitman
TR
Collins
SJ
Keene
BR
Terminal differentiation of human promyelocytic leukemic cells in primary cuture in response to retinoic acid.
Blood
57
1981
1000
15
Fabian
I
Shvartzmayer
S
Nagler
A
In-vitro growth and differentiation of marrow cells from myelodysplastic patients in the presence of a retinoidal benzoic acid derivative.
Leuk Res
11
1987
635
16
Huang
M
Ye
Y
Chen
S
Chai
J
Lu
J
Zhoa
L
Gu
L
Wang
Z
Use of all-trans retinoic acid in the treatment of acute promyelocytic leukemia.
Blood
72
1988
567
17
Castaigne
S
Chomienne
C
Daniel
MT
Ballerini
P
Berger
R
Fenaux
P
Degos
L
All-trans retinoic acid as a differentiation therapy for acute promyelocytic leukemia. I Clinical results.
Blood
76
1990
1704
18
Chomienne C, Castaigne S, Cornic M, Lefebvre P, Balitrand N, Barbey S, De The H, Fenaux P, Degos L: Retinoids in acute promyelocytic leukaemia, in Armitage J, Burnett A, Keating A, Newland A (eds): Haematological Oncology. Cambridge, UK, Cambridge University, 1994, p 189
19
Munker
R
Norman
A
Koeffler
HP
Vitamin D compounds: Effect on clonal proliferation and differentiation of human myeloid cells.
J Clin Invest
78
1986
424
20
Koeffler
HP
Hirji
K
Itri
L
Southern
California Leukemia Group
1,25-dihydroxyvitamin D3: In vivo and in vitro effects on human preleukemic and leukemic cells.
Cancer Treat Rep
69
1985
1399
21
Clark
RE
Ismail
SAD
Jacobs
A
Payne
H
Smith
SA
A randomized trial of 13-cis retinoic acid with or without cytosine arabinoside in patients with the myelodysplastic syndrome.
Br J Haematol
66
1987
77
22
Savill
JS
Wyllie
AH
Henson
JE
Walport
MJ
Henson
PM
Haslett
C
Macrophage phagocytosis of aging neutrophils in inflammation: Programmed cell death in the neutrophil leads to its recognition by macrophages.
J Clin Invest
83
1989
865
23
Martin
SJ
Bradley
JG
Cotter
TG
HL60 cells induced to differentiate towards neutrophils subsequently die via apoptosis.
Clin Exp Immunol
79
1990
448
24
Naumovski
L
Cleary
ML
Bcl-2 inhibits apoptosis associated with terminal differentiation of HL60 myeloid leukemia cells.
Blood
83
1994
2261
25
Park
JR
Robertson
K
Hickstein
DD
Tsai
S
Hockenbery
DM
Collins
SJ
Dysregulated bcl-2 expression inhibits apoptosis but not differentiation of retinoic acid-induced HL60 granulocytes.
Blood
84
1994
440
26
Xu
H
Tepper
CG
Jones
JB
Fernandez
CE
Studzinski
GP
1,25-dihydroxyvitamin D3 protects HL60 cells against apoptosis but down-regulates the expression of the Bcl-2 gene.
Exp Cell Res
209
1993
367
27
Cotter
TG
Fernandes
RS
Verhaegen
S
McCarthy
JV
Cell death in the myeloid lineage.
Immunol Rev
142
1994
93
28
Del Bino
G
Li
X
Traganos
F
Darzynkiewicz
Z
Altered susceptibility of differentiating HL60 cells to apoptosis induced by antitumor drugs.
Leukemia
8
1994
281
29
Solary
E
Bertrand
R
Kohn
KW
Pommier
Y
Differential induction of apoptosis in undifferentiated and differentiated HL60 cells by DNA topoisomerase I and II inhibitors.
Blood
81
1993
1359
30
Zwelling
LA
Chan
D
Hinds
M
Mayes
J
Silberman
LE
Blick
M
Effect of phorbol ester treatment on drug-induced, topoisomerase II-mediated DNA cleavage in human leukemia cells.
Cancer Res
48
1988
6625
31
Wiernik
PH
Banks
PLC
Case
DCJ
Arlin
ZA
Periman
PO
Todd
MB
Ritch
PS
Enck
RE
Weitberg
AB
Cytarabine plus idarubicin or daunorubicin as induction and consolidation therapy for previously untreated adult patients with acute myeloid leukemia.
Blood
79
1992
313
32
Petrini
M
Mattii
L
Valentini
P
Sabbatini
ARM
Grassi
B
Grandi
M
Idarubicin is active on MDR cells: Evaluation of DNA synthesis inhibition on P388 cell lines.
Ann Hematol
67
1993
227
33
Berman
E
McBride
M
Comparative cellular pharmacology of daunorubicin and idarubicin in human multidrug-resistant leukemia cells.
Blood
79
1992
3267
34
Berrebi A, Polliack A: Oral idarubicin in elderly acute leukemia and refractory anemia with excess of blasts, in Buchner T, Schellong G, Hiddemann W, Ritter J (eds): Acute Leukemias II. Berlin, Germany, Springer-Verlag, 1990, p 342
35
Parapia LA, Johnson E, Barlow AM, Brown SB, Galvin M, Child JAC, Roberts BR, McEvoy M, Cuthbert AC: Oral idarubicin in myelodysplastic syndromes and leukemias, in Mandelli F (ed): Idarubicin in the Treatment of Acute Leukemia. Amsterdam, The Netherlands, Excerta Medica, 1987, p 56
36
Darzynkiewicz
Z
Bruno
S
Del Bino
G
Gorczyca
W
Hotz
MA
Lassota
P
Traganos
F
Features of apoptotic cells measured by flow cytometry.
Cytometry
13
1992
795
37
Delia
D
Aiello
A
Soligo
D
Fontanella
E
Melani
C
Pezzella
F
Pierotti
MA
Della Porta
G
Bcl-2 proto-oncogene expression in normal and neoplastic human myeloid cells.
Blood
79
1992
1291
38
Hollingshead
LM
Faulds
D
Idarubicin: A review of its pharmacodynamic and pharmacokinetic properties, and therapeutic potential in the chemotherapy of cancer.
Drugs
42
1991
690
39
Wyllie
AH
Glucocorticoid-induced thymocyte apoptosis is associated with endogenous endonuclease activation.
Nature
284
1980
555
40
Sundstrom
C
Nilsson
K
Establishment and characterization of a human histiocytic lymphoma cell line (U937).
Int J Cancer
17
1976
565
41
Koeffler
HP
Golde
DW
Acute myelogenous leukemia: A human cell line responsive to colony-stimulating activity.
Science
200
1978
1153
42
Kelsey
SM
Makin
HLJ
Newland
AC
Functional significance of induction of differentiation in human myeloid leukaemia blasts by 1,25-dihydroxyvitamin D3 and GM-CSF.
Leuk Res
16
1992
427
43
Kim
YR
Abraham
NG
Lutton
JD
Mechanisms of differentiation of U937 leukemic cells induced by GM-CSF and 1,25(OH)2 vitamin D3.
Leuk Res
15
1991
409
44
Yang
E
Korsmeyer
SJ
Molecular thanatopsis: A discourse on the BCL2 family and cell death.
Blood
88
1996
386
45
Sanz
C
Benito
A
Silva
M
Albella
B
Richard
C
Segovia
JC
Insunza
A
Bueren
JA
Fernandez-Luna
JL
The expression of bcl-X is downregulated during differentiation of human hematopoietic progenitor cells along the granulocyte but not the monocyte/macrophage lineage.
Blood
89
1997
3199
46
Bergh
G
Ehinger
M
Olofsson
T
Baldetorp
B
Johnson
E
Brycke
H
Lindgren
G
Olsson
I
Gullberg
U
Altered expression of the retinoblastoma tumor-suppressor gene in leukemic cell lines inhibits induction of differentiation but not G1-accumulation.
Blood
89
1997
2938
47
Fan
S
Chang
JK
Smith
ML
Duba
D
Fornace
AJ Jr
O'Connor
PM
Cells lacking CIP1/WAF1 genes exhibit preferential sensitivity to cisplatin and nitrogen mustard.
Oncogene
14
1997
2127
48
Sheikh
MS
Chen
YQ
Smith
ML
Fornace
AJ Jr
Role of p21Waf1/Cip1/Sdi1 in cell death and DNA repair as studied using a tetracycline-inducible system in p53-deficient cells.
Oncogene
14
1997
1875
49
Warbrick
E
Lane
DP
Glover
DM
Cox
LS
Homologous regions of Fen1 and p21Cip1 compete for binding to the same site on PCNA: A potential mechanism to co-ordinate DNA replication and repair.
Oncogene
14
1997
2313
50
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
51
El-Deiry
WS
Tokino
T
Velculescu
VE
Levy
DB
Parsons
R
Trent
JM
Lin
D
Mercer
WE
Kinzler
KW
Vogelstein
B
WAF1, a potential mediator of p53 tumor suppression.
Cell
75
1993
817
52
Harper
JW
Adami
GR
Wei
N
Keyomarsi
K
Elledge
SJ
The p21 Cdk-interacting protein Cip1 is a potent inhibitor of G1 cyclin-dependent kinases.
Cell
75
1993
805
53
Steinman
RA
Hoffman
B
Iro
A
Guillouf
C
Liebermann
DA
El-Houseini
ME
Induction of p21 (WAF-1/CIP1) during differentiation.
Oncogene
9
1994
3389
54
Jiang
H
Lin
J
Su
Z
Collart
FR
Huberman
E
Fisher
PB
Induction of differentiation in human promyelocytic HL-60 leukemia cells activates p21, WAF1/CIP1, expression in the absence of p53.
Oncogene
9
1994
3397
55
Zhang
W
Grasso
L
McClain
CD
Gambel
AM
Cha
Y
Travali
S
Deisseroth
AB
Mercer
WE
p53-independent induction of WAF1/CIP1 in human leukemia cells is correlated with growth arrest accompanying monocyte/macrophage differentiation.
Cancer Res
55
1995
668
56
Liu
M
Lee
M
Cohen
M
Bommakanti
M
Freedman
LP
Transcriptional activation of the Cdk inhibitor p21 by vitamin D3 leads to the induced differentiation of the myelomonocytic cell line U937.
Genes Dev
10
1996
142
57
Freemerman
AJ
Vrana
JA
Tombes
RM
Jiang
H
Chellappan
SP
Fisher
PB
Grant
S
Effects of antisense p21 (WAF1/CIP1/MDA6) expression on the induction of differentiation and drug-mediated apoptosis in human myeloid leukemia cells (HL-60).
Leukemia
11
1997
504
58
Zhang
W
Kornblau
SM
Kobayashi
T
Gambel
AM
Claxton
D
Deisseroth
AB
High levels of constitutive WAF1/Cip1 protein are associated with chemoresistance in acute myelogenous leukemia.
Clin Cancer Res
1
1995
1051
59
Farzaneh
F
Zalin
R
Brill
D
Shall
S
DNA strand breaks and ADP-ribosyl transferase activation during cell differentiation.
Nature
300
1982
362
60
Khan
Z
Francis
GE
Contrasting patterns of DNA strand breakage and ADP-ribosylation-dependent DNA ligation during granulocyte and monocyte differentiation.
Blood
69
1987
1114
61
Satoh
MS
Lindahl
T
Role of poly(ADP-ribose) formation in DNA repair.
Nature
356
1992
356
62
Kaufmann
SH
Desnoyers
S
Ottaviano
Y
Davidson
NE
Poirier
GG
Specific proteolytic cleavage of poly(ADP-ribose) polymerase: An early marker of chemotherapy-induced apoptosis.
Cancer Res
53
1993
3946
63
Kelman
Z
PCNA: Structure, functions and interactions.
Oncogene
14
1997
629
64
Shiohara
M
El-Deiry
WS
Wada
M
Nakamaki
T
Takeuchi
S
Yang
R
Chen
D
Vogelstein
B
Koeffler
HP
Absence on WAF1 mutations in a variety of human malignancies.
Blood
84
1994
3781
65
Yang
GS
Minden
MD
McCulloch
EA
Influence of schedule on regulated sensitivity of AML blasts to cytosine arabinoside.
Leukemia
7
1993
1012
66
Fenaux P, Degos L: Treatment of acute promyelocytic leukaemia, in Lowenberg B (ed): Acute Myelogenous Leukemia and Myelodysplasia. London, UK, Bailliere Tindall, 1996, p 107
67
Tafuri
A
Andreeff
M
Kinetic rationale for cytokine-induced recruitment of myeloblastic leukemia followed by cycle-specific chemotherapy in vitro.
Leukemia
4
1990
826
68
Stone
RM
Berg
DT
George
SL
Dodge
RK
Paciucci
PA
Schulman
P
Lee
EJ
Moore
JO
Powell
BL
Schiffer
CA
Granulocyte-macrophage colony-stimulating factor after initial chemotherapy for elderly patients with primary acute myelogenous leukemia.
N Engl J Med
332
1995
1671
69
Banker
DE
Groudine
M
Norwood
T
Appelbaum
FR
Measurement of spontaneous and therapeutic agent-induced apoptosis with BCL-2 protein expression in acute myeloid leukemia.
Blood
89
1997
243
Sign in via your Institution