14
ORIGINAL ARTICLE: RESEARCH Arsenic trioxide cooperates with all-trans retinoic acid to enhance mitogen-activated protein kinase activation and differentiation in PML–RARa negative human myeloblastic leukemia cells SATYAPRAKASH NAYAK 1 , MIAOQING SHEN 2 , RODICA P. BUNACIU 2 , STEPHEN E. BLOOM 3 , JEFFREY D. VARNER 1 , & ANDREW YEN 2 1 School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA, 2 Department of Biomedical Sciences, Cornell University, Ithaca, NY, USA, and 3 Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA (Received 12 May 2010; revised 4 June 2010; accepted 10 June 2010) Abstract Arsenic trioxide (ATO) synergistically promotes all-trans retinoic acid (ATRA)-induced differentiation of PML–RARa negative HL-60 myeloblastic leukemia cells. In PML–RARa positive myeloid leukemia cells, ATO is known to cause degradation of PML–RARa with subsequent induced myeloid differentiation. We found that ATO by itself does not cause differentiation of the PML–RARa negative HL-60 cells, but enhances ATRA’s capability to cause differentiation. ATO augmented ATRA-induced RAF/MEK/ERK axis signaling, expression of CD11b and p47 PHOX , and inducible oxidative metabolism. ATO enhanced ATRA-induced population growth retardation without evidence of apoptosis or enhanced G1/ G0 growth arrest. Compared to ATRA-treated cells, the ATRA plus ATO-treated cells progressed more slowly through the cell cycle as detected by a slower rate of accumulation in G2/M following nocodazole treatment. Hoechst/PI staining showed that low-dose ATO did not induce apoptosis. In summary, our results indicate that ATO in conjunction with ATRA is of potential chemotherapeutic use in PML–RARa negative leukemias. Keywords: HL-60, all-trans retinoic acid, arsenic trioxide, MAPK, differentiation Introduction Arsenic has historically been used as a medicinal agent for a variety of diseases such as psoriasis, eczema, asthma, malaria, ulcers, syphilis, and leuke- mia. Arsenic and its derivatives have been used as antiseptics, antispasmodics, antipyretics, sedatives, and tonics, etc., especially in traditional Chinese medicine [1]. The most common compound of arsenic used in medicine is arsenic trioxide. Arsenic trioxide (As 2 O 3 ) or ATO is the active ingredient in Fowler’s solution developed in the 18th century. It has been used to treat different malignancies for over 100 years. In the 1930s, before the introduction of chemotherapy and radiation therapy, arsenic was used in one of the standard treatments for chronic myeloid leukemia and other leukemias [1,2]. The modern use of arsenic trioxide as a therapy for acute promyelocytic leukemia (APL) originated in China in the 1990s [3]. In a study at the Shanghai Institute of Hematology (SIH) in 1999, patients with relapsed APL were treated with ATO. Complete remission (CR) was achieved in 9/10 of the cases treated with ATO alone and 5/5 cases treated with ATO plus chemotherapy (CT) or retinoic acid [3–5]. This suggests that ATO is potent as a single agent in APL, a disease cytogenetically characterized by a t(15;17) translocation that gives rise to the PML–RARa fusion protein (promyelocytic leukemia–retinoic acid receptor a), which is a rogue transcription factor. This was further proved by relatively high 2- and 3-year disease-free survival (DFS) rates seen recently in newly diagnosed leukemias [6]. Correspondence: Prof. Andrew Yen, Department of Biomedical Sciences, T4-008, Veterinary Research Tower (VRT), Cornell University, Ithaca, NY 14853, USA. Tel: (607)253-3354. Fax: (607)253-3317. E-mail: [email protected] Leukemia & Lymphoma, September 2010; 51(9): 1734–1747 ISSN 1042-8194 print/ISSN 1029-2403 online Ó 2010 Informa UK, Ltd. DOI: 10.3109/10428194.2010.501535 Leuk Lymphoma Downloaded from informahealthcare.com by Procter & Gamble Pharmaceuticals on 03/14/11 For personal use only.

Arsenic trioxide cooperates with all trans retinoic acid to enhance mitogen-activated protein kinase activation and differentiation in PML–RARα negative human myeloblastic leukemia

Embed Size (px)

Citation preview

ORIGINAL ARTICLE: RESEARCH

Arsenic trioxide cooperates with all-trans retinoic acid to enhancemitogen-activated protein kinase activation and differentiation inPML–RARa negative human myeloblastic leukemia cells

SATYAPRAKASH NAYAK1, MIAOQING SHEN2, RODICA P. BUNACIU2,

STEPHEN E. BLOOM3, JEFFREY D. VARNER1, & ANDREW YEN2

1School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA, 2Department of Biomedical

Sciences, Cornell University, Ithaca, NY, USA, and 3Department of Microbiology and Immunology, Cornell University,

Ithaca, NY, USA

(Received 12 May 2010; revised 4 June 2010; accepted 10 June 2010)

AbstractArsenic trioxide (ATO) synergistically promotes all-trans retinoic acid (ATRA)-induced differentiation of PML–RARanegative HL-60 myeloblastic leukemia cells. In PML–RARa positive myeloid leukemia cells, ATO is known to causedegradation of PML–RARa with subsequent induced myeloid differentiation. We found that ATO by itself does not causedifferentiation of the PML–RARa negative HL-60 cells, but enhances ATRA’s capability to cause differentiation. ATOaugmented ATRA-induced RAF/MEK/ERK axis signaling, expression of CD11b and p47PHOX, and inducible oxidativemetabolism. ATO enhanced ATRA-induced population growth retardation without evidence of apoptosis or enhanced G1/G0 growth arrest. Compared to ATRA-treated cells, the ATRA plus ATO-treated cells progressed more slowly through thecell cycle as detected by a slower rate of accumulation in G2/M following nocodazole treatment. Hoechst/PI staining showedthat low-dose ATO did not induce apoptosis. In summary, our results indicate that ATO in conjunction with ATRA is ofpotential chemotherapeutic use in PML–RARa negative leukemias.

Keywords: HL-60, all-trans retinoic acid, arsenic trioxide, MAPK, differentiation

Introduction

Arsenic has historically been used as a medicinal

agent for a variety of diseases such as psoriasis,

eczema, asthma, malaria, ulcers, syphilis, and leuke-

mia. Arsenic and its derivatives have been used as

antiseptics, antispasmodics, antipyretics, sedatives,

and tonics, etc., especially in traditional Chinese

medicine [1]. The most common compound of

arsenic used in medicine is arsenic trioxide. Arsenic

trioxide (As2O3) or ATO is the active ingredient in

Fowler’s solution developed in the 18th century. It

has been used to treat different malignancies for over

100 years. In the 1930s, before the introduction of

chemotherapy and radiation therapy, arsenic was

used in one of the standard treatments for chronic

myeloid leukemia and other leukemias [1,2]. The

modern use of arsenic trioxide as a therapy for

acute promyelocytic leukemia (APL) originated in

China in the 1990s [3]. In a study at the Shanghai

Institute of Hematology (SIH) in 1999, patients

with relapsed APL were treated with ATO.

Complete remission (CR) was achieved in 9/10 of

the cases treated with ATO alone and 5/5 cases

treated with ATO plus chemotherapy (CT) or

retinoic acid [3–5]. This suggests that ATO is potent

as a single agent in APL, a disease cytogenetically

characterized by a t(15;17) translocation that gives

rise to the PML–RARa fusion protein (promyelocytic

leukemia–retinoic acid receptor a), which is a rogue

transcription factor. This was further proved by

relatively high 2- and 3-year disease-free survival

(DFS) rates seen recently in newly diagnosed

leukemias [6].

Correspondence: Prof. Andrew Yen, Department of Biomedical Sciences, T4-008, Veterinary Research Tower (VRT), Cornell University, Ithaca, NY 14853,

USA. Tel: (607)253-3354. Fax: (607)253-3317. E-mail: [email protected]

Leukemia & Lymphoma, September 2010; 51(9): 1734–1747

ISSN 1042-8194 print/ISSN 1029-2403 online � 2010 Informa UK, Ltd.

DOI: 10.3109/10428194.2010.501535

Leu

k L

ymph

oma

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Proc

ter

& G

ambl

e Ph

arm

aceu

tical

s on

03/

14/1

1Fo

r pe

rson

al u

se o

nly.

All-trans retinoic acid (ATRA) has been used in

differentiation induction therapy for APL since the

early 1980s [7,8]. A recent clinical study of APL

treatment with ATRA differentiation therapy in Japan

resulted in a CR rate of 94%, a 6-year DFS rate of

68.5%, and a 6-year overall survival (OS) rate

of 83.9% [9]. Clinical studies using a combination of

ATRA and ATO showed that the time required to

achieve CR with ATRA–ATO was statistically sig-

nificantly different (25.5 days) from that with ATRA

alone (40.5 days) or ATO alone (31 days). The

median DFS rate was also increased to 20 months for

the combination treatment as compared to ATRA

alone (13 months) and ATO alone (16 months) [10].

Furthermore, the disease burden, determined by the

fold change in PML–RARa, was much shorter in the

combination treatment compared to monotherapy.

Clinical studies thus show a synergistic effect of ATRA

and ATO treatments, motivating interest in the

biochemical cause of this synergy. Both ATRA and

ATO have been found to cause degradation of the

PML–RARa fusion protein, suggesting a mechanism

of action whereby degradation of the transforming

protein relieves the block in differentiation and permits

progression of differentiation along the myeloid line-

age. ATO has been found to bind the PML–RARa and

promote its SUMOylation and degradation [11].

However, ATO may have many other actions, as is

the case for ATRA, and their collaboration may be

attributable to other biochemical mechanisms they

share as targets. It is thus of interest to determine

whether they collaborate in a setting with no PML–

RARa. If so, this may suggest therapeutic potential for

the combination of RA and ATO in tumors other than

PML–RARa positive APL.

HL-60 is a human myeloblastic leukemia cell line

that has been extensively used as a model for

pharmacologically induced differentiation. HL-60

cells undergo either myeloid or monocytic differ-

entiation, and G1/G0 growth arrest, when treated

with ATRA or 1-25-dihydroxyvitamin D3 (vitamin

D3), respectively. Previous studies in our laboratory

have shown that ATRA- or vitamin D3-induced

differentiation and G1/G0 cell arrest require sus-

tained activation of mitogen-activated protein kinase

(MAPK) signaling along the RAF/MEK/ERK axis

[12,13]. ATRA induces the MAPK-dependent ex-

pression of cell surface differentiation markers such

as CD38 and CD11b, as well as functional differ-

entiation markers such as inducible oxidative meta-

bolism that become progressively expressed as the

cells undergo myeloid differentiation.

HL-60 cells are negative for the t(15;17) chromo-

somal translocation that defines APL and gives rise to

the PML–RARa fusion protein. If the ability of ATO

to promote differentiation is solely through

degradation of the PML–RARa fusion protein, then

we should not see any enhanced differentiation for

the combination treatment of ATRA and ATO.

However, if ATO action collaborates with the

mechanism of ATRA-induced differentiation, e.g.

by promoting sustained activation of MAPK, then

addition of ATO to ATRA treatment should

augment the differentiation of HL-60 cells.

Previous studies on the effects of ATO show that

high concentrations of ATO were able to induce

apoptosis in drug-resistant HL-60 cell lines [14].

Experiments by Jiang et al. showed that the effect of

ATO may be mediated by c-Myc down-regulation,

whereas Herst et al. showed that adding ATO led to

apoptosis after 5 days [15,16]. The mechanism of

apoptosis induction was discovered to be via the

death receptor pathway rather than the intrinsic

(mitochondrial) pathway. The present studies using

low doses of ATO showed that, compared to cells

treated with ATRA alone, HL-60 cells treated with a

combination of ATRA and ATO exhibited augmen-

ted differentiation propelled by an enhanced activa-

tion of MAPK signaling. While the addition of ATO

to ATRA treatment enhanced population growth

retardation, there was no corresponding increase

seen in the percentage of cells undergoing G0-

specific cell cycle arrest, but rather an apparent

overall retardation of cell cycle progression. How-

ever, ATO alone did not cause differentiation in HL-

60 cells, as indicated before for PML–RARa positive

cells [17]. The results show that ATO can enhance

the differentiation-inducing effects of ATRA in

PML–RARa negative cells, consistent with a para-

digm whereby ATO enhances the ATRA-induced

MAPK signaling that propels induced differentia-

tion. Since ATO is already Food and Drug Admin-

istration (FDA) approved (Trisenox), along with

ATRA differentiation therapy for APL, a more

thorough understanding of the mechanism of action

of ATO, especially on the MAPK and related

pathways, may be of therapeutic value in treating

other leukemias, or even non-hematologic cancers.

Materials and methods

Cell culture

HL-60 human myeloblastic leukemia cells were

cultured in RPMI 1640 medium supplemented with

5% heat-inactivated fetal bovine serum (Invitrogen,

Carlsbad, CA, USA) in a 5% CO2 humidified

environment at 378C, and were maintained in an

exponential growth phase. All the experimental

cultures were initiated at a density of 0.26106 cells/

mL. Cell viability was monitored by Trypan Blue Stain

(Invitrogen) and consistently exceeded 95%.

Arsenic trioxide in PML–RARa (7) cells 1735

Leu

k L

ymph

oma

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Proc

ter

& G

ambl

e Ph

arm

aceu

tical

s on

03/

14/1

1Fo

r pe

rson

al u

se o

nly.

Reagents used

All-trans retinoic acid (Sigma-Aldrich, St. Louis, MO,

USA) was dissolved in 100% ethanol to make a stock

solution concentration of 5 mM. The concentration of

ATRA used in cultures was 1 mM. Arsenic trioxide

(Sigma-Aldrich) was diluted in water to make a stock

concentration of 5 mM. ATO was used at a final

concentration of 2 mM, and added at the same time as

ATRA to the cultures of HL-60 cells. NB4 cells,

however, were treated with 0.25 mM ATRA and

0.5 mM ATO. Mitomycin C (MMC) (Sigma-Aldrich)

was used at a final concentration of 100 mM.

Antibodies for Western analysis against ERK1/2,

phosphorylated ERK-1/2 (Thr-202/Tyr-204), MEK1/

2, phosphorylated MEK1/2 (Ser-217/221), p47PHOX,

and GAPDH (glyceraldehyde 3-phosphate dehydro-

genase), as well as MEK1 inhibitor PD98059, were

obtained from Cell Signaling (Beverly, MA, USA).

PD98059 was dissolved in dimethylsulfoxide (DMSO)

to make a stock solution of 2 mM. PD98059 was

added 16 h before the addition of ATRA and ATO to

make its concentration in culture 1 mM. It was also re-

added along with ATRA and ATO (at 0 h) and finally

at 16 h after the addition of ATRA and ATO. The

detailed procedure for the addition of PD98059 has

been explained elsewhere [12]. Nocodazole was

purchased from Sigma.

Procedure for preparing total cell lysates and Western blots

Total cell lysates were prepared as follows. Cells were

harvested at the 48 h time point, and washed with

phosphate-buffered saline (PBS) and centrifuged

three times to wash away any culture medium. After

washing away the medium, cells were re-suspended

in 100 mL M-PER Mammalian Protein Extraction

Reagent (Invitrogen) buffer mixed with a phospha-

tase and protease inhibitor. Western analysis was

done by resolving the lysates on 12% Tris-HCl gels

(Bio Rad), and the membrane was blocked overnight

with 5% w/v non-fat milk. The membrane was then

probed using primary and secondary antibodies as

described above in ‘Reagents used.’ All Western

blots shown are typical of at least three repeats.

CD38 and CD11b expression studies by flow cytometry

At the 24 h and 48 h time points, 0.56 106 cells

were collected, centrifuged to a pellet, and resus-

pended in 100 mL PBS containing 5 mL allophyco-

cyanin (APC)-conjugated CD11b and 5 mL

phycoerythrin (PE)-conjugated CD38 antibodies.

The cells were then incubated for 1 h in a humidified

atmosphere of 5% CO2 at 378C, and expression

levels were measured by flow cytometry (BD LSR II

flow cytometer; Becton Dickinson, Mountain View,

CA, USA). CD11b expression was measured by

633 nm red laser excitation and emission reflected

from a 735 long-pass dichroic mirror and collected

through a 660/20 band-pass filter. CD38 expression

was measured with 488 nm laser excitation and

emission through a 550 long-pass dichroic mirror

and 576/26 band-pass filter. Gates to determine the

shift in cellular fluorescence intensity were set to

exclude 95% of the control (untreated) cells.

Measurement of inducible oxidative metabolism

Approximately 0.56 106 cells were collected at the

48 h and 72 h time points, centrifuged to a pellet,

and resuspended in 200 mL PBS at 378C containing

5 mmol/L 5- (and 6-) chloromethyl-20,70-dichlorodi-

hydrofluorescein diacetate acetyl ester (H2DCF-DA;

Molecular Probes) without or with 0.2 mg/mL 12-O-

tetradecanoylphorbol-13-acetate (TPA; Sigma).

H2DCF-DA and TPA stock solutions were made

in DMSO at concentrations of 0.2 mg/mL and

0.5 mg/mL, respectively. Cells were incubated for

20 min in a humidified atmosphere of 5% CO2 at

378C. Flow cytometric analysis was done (BD LSR

II flow cytometer) using 488 nm excitation, and

emission from oxidized DCF was collected through a

505 long-pass dichroic mirror and 530/30 band pass

filter. The shift in fluorescence intensity in response

to TPA was used to determine the percentage of cells

capable of inducible oxidative metabolism. Gates to

determine the percentage of positive cells were set to

exclude 95% of the same cells not treated with TPA.

Hoechst/propidium iodide assay to determine extent of

apoptosis in culture

A volume of 250 mL of cell culture at the 72 h time

point was taken and placed in a tube. Then 25 mL of

Hoechst 33342 and 25 mL of propidium iodide

working solutions were added to the cell suspension.

The concentration of Hoechst stock solution was

1 mM, which was diluted 1:4 in sterile water to obtain

the working solution. Propidium iodide stock solution

was prepared at a concentration of 200 mg/mL, which

was diluted 1:9 to obtain the working solution. The

cells were mixed very gently by tapping, and incubated

in a water bath for 15 min at 378C. Next, 25 mL of

stained cells were loaded onto a microscope slide and

placed on the fluorescence microscope for counting.

Round blue cells with no condensation of chromatin

or small masses inside the cells were treated as live,

viable cells. Cells that had small, blue masses inside

them were counted as apoptotic, whereas pink cells

with non-condensed chromatin were treated as necro-

tic. One hundred cells per slide were counted and

1736 S. Nayak et al.

Leu

k L

ymph

oma

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Proc

ter

& G

ambl

e Ph

arm

aceu

tical

s on

03/

14/1

1Fo

r pe

rson

al u

se o

nly.

segregated into viable, apoptotic, or necrotic cate-

gories.

Cell cycle analysis

About 0.56106 cells were harvested at 72 h by

centrifugation and resuspended in 200 mL hypotonic

staining solution (at 48C) containing 50 mg/mL propi-

dium iodide (PI), 1 mL Triton X-100, and 1 mg/mL

sodium citrate. Cells were incubated at room tem-

perature for 1 h and analyzed using flow cytometry

(BD LSR II). Excitation was provided by a 488 nm

laser and the emission was collected using a 550 long-

pass dichroic mirror and a 576/26 band-pass filter.

Cell cycle progression and accumulation in G2/M

during mitotic block were measured using 100 ng/

mL nocodazole added to cultures that were un-

treated, or treated with ATRA, ATO, or ATRA plus

ATO for 24 h. The cells were harvested after 0, 6,

12, and 18 h and stained with propidium iodide for

cell cycle analysis. Nocodazole was added to the

cultures at the same time as ATO and ATRA.

Statistical analysis

Statistical analysis was done using MATLAB’s

statistical toolbox. Three independent repeats were

performed in each case. Means of treatment cases

were compared using paired-samples t-test, using the

ttest2 function in MATLAB’s statistical toolbox. The

bar graphs show means of three independent repeats

along with standard errors.

Results

Arsenic trioxide leads to enhanced sustained activation of

ATRA-induced MAPK proteins

It is well known that transient activation of MAPK leads

to proliferation, whereas a sustained activation of

MAPK, especially ERK1/2, has been thought to drive

differentiation in a variety of cell lines, including HL-60

cells [12,13]. It is also known that ATO can activate

MAPK pathways as part of a stress response [18,19].

We studied the effect of ATO on MAPK signaling, in

particular the potential synergistic effects of ATRA and

ATO on MEK and ERK. Treating cells with ATO plus

ATRA enhanced the increase in pMEK1/2 (Ser-217/

221) [Figure 1(A)] and pERK1/2 (Thr-202/Tyr-204)

[Figure 1(B)] induced by ATRA alone (Western blot

for MEK, pMEK, ERK, and pERK was done after

48 h of ATRA/ATO treatment). Interestingly, there

was also increased activation of ERK1/2 (Thr-202/Tyr-

204) in the ATO-alone treatment case compared to the

Figure 1. Western blot analysis of MAPK proteins shows enhanced activation in the combination treatment of ATRA and ATO compared to

ATRA treatment. (A, B) The effect of the different treatments on expression and activation levels of MEK1/2 and ERK1/2, respectively; (C)

the effect in the presence of MEK1 inhibitor PD98059. The left columns show the effect on expression levels at 48 h, whereas the right

columns show the effect on activation of MAPK proteins. The lanes show control (untreated) cells, cells treated with ATRA, ATRA plus

ATO, and ATO only. Increased activation was seen for MEK1/2 as well as ERK1/2 at 48 h. We did not see a large increase in total

expression of MEK1/2 and ERK1/2 for any of the treatment cases (see left column) at 48 h. This suggests that ATO (in combination or

alone) only affects activation (via phosphorylation) of MAPK proteins, without affecting their total expression levels. (C) The effect of adding

MEK1 inhibitor (PD98059) on p-MEK and p-ERK at 48 h time-point is shown.

Arsenic trioxide in PML–RARa (7) cells 1737

Leu

k L

ymph

oma

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Proc

ter

& G

ambl

e Ph

arm

aceu

tical

s on

03/

14/1

1Fo

r pe

rson

al u

se o

nly.

control (untreated) case. The ATO-induced increase

could be blunted by addition of a known MEK1

inhibitor, PD98059 [Figure 1(C)]. Western analysis of

ERK1/2 expression showed that there was no increase

in the total expression of ERK1/2 in any of the

treatment cases. The expression levels of ERK1/2

were thus unaffected by treatment, although their

activation increased. Western analysis of total MEK1/

2 [Figure 1(A)] also showed no effect of either ATRA

or ATO treatment on MEK1/2 expression levels.

Similar to ERK, the increased phosphorylated MEK1/

2 with the combination treatment is thus attributable to

increased activation and not to increased expression.

Arsenic trioxide enhances ATRA-induced CD11b but not

CD38 expression

Having established enhanced activation of MAPK

signaling in the combination treatment, we then

proceeded to investigate whether this augmentation

translated into enhanced differentiation in HL-60

cells. A visual inspection of different treatments for

96 h suggested that there was a synergy in the

combination treatment which led to enhanced

differentiation (Figure 2). Cells treated with ATO

alone did not cause any apparent differentiation, but

did exhibit a decrease in growth rate. To investigate

whether combination treatment led to enhanced

differentiation, we started by assessing the cell

surface markers of differentiation, CD38 and

CD11b.

CD38 is a 45 kDa ectoenzyme type II transmem-

brane receptor. The gene is transcriptionally regu-

lated by a retinoic acid response element (RARE) in

its first intron, which confers response to ATRA [20].

It has been shown that CD38 can elicit the MAPK

cascade, and that one component of the signaling

machinery is the c-Cbl adaptor [21]. To determine

the effects of ATO and ATRA on CD38 expression,

we measured expression of CD38 at an early time

point (6 h) and at 24 h after various treatments,

using a PE-conjugated CD38 antibody and flow

cytometry. As seen in Figure 3, treatment with

ATRA induced progressively increasing CD38

Figure 2. Change in cellular morphology induced by different treatments. Wright’s stained images showing morphological changes at 96 h

after different treatments. (A) In the control (untreated) case, mostly round cells were observed. Addition of ATRA led to differentiated cells,

seen here as distorted cells (C). Differentiation was more apparent in the combination treatment of ATRA and ATO, whereas the ATO-alone

treatment case shows a morphology similar to the control case, although a smaller number of cells are seen in this case (D and B,

respectively).

1738 S. Nayak et al.

Leu

k L

ymph

oma

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Proc

ter

& G

ambl

e Ph

arm

aceu

tical

s on

03/

14/1

1Fo

r pe

rson

al u

se o

nly.

expression that was indistinguishable from treatment

with ATRA plus ATO. There was no increase in

CD38 expression in the cells treated with ATO

alone. These results indicated that ATO had no

effect on CD38 expression, either in combination

with ATRA or by itself. Since CD38 has retinoic acid

response elements in its promoter, RA-induced

expression will not be affected unless ATO affects

that transcriptional activation.

Another cell surface marker, CD11b, an integrin

receptor subunit, was used to characterize the effect

of ATO on ATRA-induced cell differentiation.

Expression of CD11b typically marks the progression

of ATRA-induced differentiation beyond the stage of

CD38 expression, which is an early marker. CD11b

expression was measured by flow cytometry using an

APC-conjugated CD11b antibody [22,23]. The

effect of ATO and ATRA, alone and in combination,

on CD11b expression was measured at 48 and 72 h.

Compared to untreated control cells, ATRA-treated

cells showed an increase in CD11b expression, as

expected. However, CD11b expression was consis-

tently higher in the case of the ATRA–ATO

combination when compared with the ATRA-alone

case [Figures 3(B) and 3(C)]. The difference

between CD11b expressions was statistically signifi-

cant at both 48 h (p¼ 0.004) and 72 h (p¼ 0.016),

indicating that the differentiation was affected by

ATO during its both early and late progression. In

contrast, the cells treated with ATO alone did not

show an increase when compared with the untreated

control case, indicating that ATO alone does not

affect CD11b expression.

Arsenic trioxide enhances ATRA-induced differentiation

by up-regulating p47PHOX

Inducible oxidative metabolism marked by an

increase in reactive oxygen species (ROS) is one of

the functional markers of differentiation into mature

Figure 3. ATO enhances ATRA-induced CD11b expression, but not CD38 expression. (A) There was no enhancement in CD38 expression

in the combination treatment, but CD11b (B) expression showed a synergistic increase in the ATRA–ATO combined treatment case. CD38

expression was analyzed with PE-conjugated CD38 antibody at 6 h and 24 h and CD11b expression was analyzed with APC-conjugated

CD11b antibody at 48 h and 72 h by flow cytometry. The y-axis shows the percentage of CD38 positive cells (A) and CD11b positive cells

(B) at indicated time points when compared with untreated cells, with gates set to exclude 95% of cells in the control case. The data show

mean values+ standard errors. * and # indicate statistically significant differences compared to control. ## and ** indicate statistically

significant differences compared to ATRA alone. For example, CD11b expression in cells treated with ATRA–ATO was statistically

significantly different from that in cells treated with ATRA alone at both 48 h (p¼0.004) and 72 h (p¼0.016). (C) Representative CD11b

histograms of untreated (control) cells and cells treated with ATRA, ATRA–ATO combination, and ATO alone at 48 h.

Arsenic trioxide in PML–RARa (7) cells 1739

Leu

k L

ymph

oma

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Proc

ter

& G

ambl

e Ph

arm

aceu

tical

s on

03/

14/1

1Fo

r pe

rson

al u

se o

nly.

myelo-monocytic cells. p47PHOX is one of the

essential components of the NADPH (reduced

nicotinamide adenine dinucleotide phosphate)–oxi-

dase complex which generates ROS when activated

in differentiated cells. We measured the effect of

ATO and ATRA separately and in combination on

p47PHOX expression. The untreated control cells and

ATO-treated cells did not show p47PHOX expression

at 48 h [Figure 4(A)]. Consistent with the trend seen

in MAPK activation, enhanced expression was seen

in the ATRA–ATO combination treatment com-

pared to treatment with ATRA alone. This result

suggests that ATO synergizes with ATRA to induce

expression of p47PHOX, although ATO by itself does

not induce p47PHOX expression. Also, this enhance-

ment in p47PHOX was abrogated in the presence of

MEK1 inhibitor PD98059 [Figure 4(B)]. This is

consistent with the suggestion that the enhancement

in differentiation in the combined treatment case is

through the MAPK axis, as inhibition of MEK1/2

activation leads to decreased differentiation.

Arsenic trioxide enhances ATRA-induced functional

differentiation in HL-60 and NB4 cells

To confirm the functional significance of the induced

p47PHOX expression, TPA inducible oxidative meta-

bolism, a late functional differentiation marker that

characterizes mature myelo-monocytic cells, was

measured. When treated with ATRA, HL-60 cells

typically exhibit a functional inducible ROS re-

sponse. As before, cells were treated with ATO or

ATRA alone or in combination. Inducible oxidative

metabolism was measured by flow cytometry using

H2DCF-DA, a cell-permeant indicator of reactive

oxygen species that is non-fluorescent until the

acetate groups are removed by intercellular esterases

when oxidation takes place within the cell. As shown

by Figure 5(A), HL-60 cells treated with a combina-

tion of ATRA–ATO showed increased functional

differentiation at both 48 h and 72 h time points

when compared with cells treated with ATRA alone.

For example, at 72 h, 85.3% of cells treated with

ATO plus ATRA were able to produce ROS induced

by TPA, compared to 60.9% of cells treated with

ATRA alone. The enhancement in ROS in the case

of ATO–ATRA was statistically significantly different

from that of ATRA alone with 95% confidence

(p¼ 0.024) at 72 h. In the case of cells treated with

ATO alone, there was no significant increase in ROS

induction upon TPA treatment, indicating that ATO

alone was unable to induce functional differentiation

of HL-60 cells. This is consistent with the inability of

ATO to induce the other differentiation markers

measured. These results indicate that, even though

ATO by itself is unable to induce differentiation of

HL-60 cells, it works synergistically to enhance

ATRA-induced differentiation.

Similar enhancement in differentiation was ob-

tained for NB4 cells with the combination treatment.

There were some key differences, however, between

the responses of HL60 cells and NB4 cells, an APL-

derived cell line. For example, NB4 cells showed

much higher sensitivity toward ATO, and massive

apoptosis was observed in cultures with ATO

concentration more than 0.5 mM. Therefore, a lower

Figure 4. (A) ATO leads to an increase in ATRA-induced expression in p47PHOX. Western analysis of total expression of p47PHOX is shown

at 48 h time point. An up-regulation in expression of p47PHOX is seen in ATRA–ATO treatment when compared with ATRA-alone

treatment. Protein lysates were extracted from untreated cells, and cells treated with ATRA, ATRA–ATO combination, and ATO alone.

Loading control is GAPDH. (B) The effect on p47PHOX upon addition of MEK inhibitor PD98059. As seen from the figure, addition of

PD98059 leads to a decrease in the expression of p47PHOX.

1740 S. Nayak et al.

Leu

k L

ymph

oma

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Proc

ter

& G

ambl

e Ph

arm

aceu

tical

s on

03/

14/1

1Fo

r pe

rson

al u

se o

nly.

dose of ATO (0.5 mM) and ATRA (0.25 mM) was

administered to NB4 cells in culture. This dosage

preserved the same ratio of ATRA to ATO as in HL-

60 cells (2:1). Another difference in the responses of

the two cell lines was that almost 100% of the cells

showed terminal differentiation at 72 h in NB4 cells.

Therefore, the inducible oxidative mechanism was

measured at earlier time points, i.e. 24 h and 48 h.

An increase in terminal differentiation was observed

at 24 h and 48 h in the combination treatment case,

when compared to the ATRA-alone case. Using a

one-tailed t-test, we concluded that differentiation in

the ATRA–ATO case was statistically more than that

in the ATO-alone case at 48 h (p¼ 0.046) [Figure

5(B)]. This result suggests that mechanisms involved

in the enhancement of differentiation in the combi-

nation are not HL-60 specific. The APL-derived

NB4 cells were, however, more ATO sensitive.

Arsenic trioxide leads to reduced population growth

HL-60 cells treated with ATO had slower population

growth rates compared to untreated cells as well as

ATRA-treated cells, as seen in Figure 6. Addition of

ATRA alone decreased the population growth rate

compared to untreated cells. However, addition of

ATO had a more drastic effect on the growth rate of

cells than ATRA. Cultures with ATO alone or the

combination treatment of ATRA–ATO grew much

slower than cultures with ATRA alone. Indeed, the

differences in growth rates between control and cell

lines with ATO were statistically significant after just

24 h (p5 0.05). Of note, growth of ATO plus

ATRA-treated populations was slower than that of

ATRA-treated populations. The difference in growth

rates of ATRA-alone and ATRA–ATO treatment

was statistically significant at both 48 h (p¼ 0.006)

Figure 5. Arsenic trioxide enhances ATRA-induced differentiation

of HL-60 cells as well as NB4 cells. (A) The percentage of cells (y-

axis) with capability for inducible oxidative metabolism at

indicated time points. Cells were treated with TPA to induce

oxidative metabolism. Using a functional marker of differentiation,

i.e. generation of ROS, we measured terminal differentiation in

untreated cells, cells treated with ATRA, a combination of ATRA–

ATO, and ATO alone. The percentage of positive cells with TPA,

when gates were set to exclude 95% of control cells (without

TPA), is shown. * indicates statistically significant difference

compared to control. ** indicates statistically significant difference

compared to ATRA alone. There was no statistically significant

difference in the percentage of differentiated cells in the ATRA-

alone versus ATRA–ATO combination treatments at 48 h.

However, at 72 h the difference in DCF positive % cells was

statistically significant between ATRA and ATRA–ATO combina-

tion treatment (p¼0.024). (B) The same is shown for NB4 cells.

There is an enhancement in differentiation seen in NB4 cells also;

however, almost all the cells are terminally differentiated at the

72 h time-point. # indicates that the percentage of DCF positive

cells are more in the ATRA case when compared to control. ##

denotes that the percentage of DCF positive cells was statistically

more (using a one-tailed t-test) in the ATRA–ATO combination

case when compared with the ATRA-alone case at 48 h with 95%

confidence (p¼0.046). The background ROS activity in all

samples was measured by cells treated with DMSO carrier blank.

Figure 6. There is a decrease in cell growth rate upon addition of

ATO. Cell growth rates were measured over a period of 72 h of

treatment. Cells were seeded at an initial concentration of 0.2

million cells/mL. The y-axis shows cell density in the culture in

million cells/mL whereas the x-axis shows time points at which the

cells were counted. Addition of ATRA also leads to decreased

growth rate; however, ATO addition has a more significant effect

by itself or in combination with ATRA. * and ** indicate that the

cell growth rates between ATO and ATRA–ATO combination

treatment cases were statistically significantly different at both 48 h

(p¼ 0.006) and 72 h (p¼ 0.013). { and {{ indicate that growth

rates in cultures with ATO alone also were statistically significantly

different from the control case at 24 h, 48 h, and 72 h time-points.

The ATO cultures did not show an increase in dead cells or debris.

Arsenic trioxide in PML–RARa (7) cells 1741

Leu

k L

ymph

oma

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Proc

ter

& G

ambl

e Ph

arm

aceu

tical

s on

03/

14/1

1Fo

r pe

rson

al u

se o

nly.

and 72 h (p¼ 0.013). Microscopic images of the

cultures at 96 h showed morphologically differen-

tiated cells in the ATRA- and ATRA–ATO (Figure

2) combination-treated cultures, whereas cells in the

ATO-alone cultures were not obviously different

from the untreated cells, morphologically. Taken

with the differentiation data, the population growth

results suggest that treatment with ATO alone slows

down population growth without inducing differen-

tiation.

Three possible causes for the decreased growth

were hypothesized and tested. A decrease in popula-

tion growth rate could be due to apoptosis upon

addition of ATO. Alternatively, an increase in cells

arresting in the G1/G0 phase could lead to an overall

decrease in population growth rates due to a

diminishing growth fraction. Finally, an overall

decrease in biosynthetic activity could cause a general

dilatation of the cell cycle and decreased cell-

doubling rates. We performed the Hoechst/propi-

dium iodide assay to seek evidence of apoptosis in

various treatment cases, cell cycle distribution by

flow cytometry for evidence of G0 cell cycle arrest,

and cell cycle transit rates measured by accumulation

in G2/M after mitotic block for evidence of cell cycle

transit slow-down.

Addition of arsenic trioxide with or without ATRA does

not lead to apoptosis

To test the first hypothesis, namely that reduced

growth rates are due to increased apoptosis, we

performed a Hoechst/PI assay to determine the

number of apoptotic and necrotic versus viable cells

in the culture. The Hoechst assay, as developed by

Bloom et al. [24], is a double fluorescence staining

procedure for the simultaneous diagnosis of apopto-

sis and necrosis in cell culture. The procedure uses

the DNA-specific fluorochrome Hoechst 33342, a

vital stain that fluoresces blue when bound to DNA

within the normal chromatin of nuclei. The cells are

also simultaneously stained in propidium iodide,

which necrotic cells cannot exclude and fluoresces

red when bound to DNA. DNA in apoptotic cells is

cleaved into smaller fragments, and there is a loss of

normal chromatin architecture. Chromatin in apop-

totic cells is segregated into variable numbers of

round masses having uniform bright (blue-white)

fluorescence. Normal cells and cells in early stages of

apoptosis take up the Hoechst dye but not the

propidium iodide. Cells having damaged membranes

do take up propidium iodide and stain pink. Necrotic

cells appear swollen and have pinkish nuclei. Figures

7(A), 7(C), and 7(D) show a typical viable cell, an

apoptotic cell, and a necrotic cell, respectively.

Figure 7(B) shows the average number of viable,

apoptotic, and necrotic cells and standard errors

obtained after 72 h of different treatments. Treat-

ment with 100 mM MMC (at 16 h after treatment)

was used as a positive control. The data show that

there is no apparent increase in apoptosis upon the

addition of ATO, when administered with or without

ATRA. The percentage of apoptotic cells seen in

cultures with MMC is consistent with previous data

from Yen et al. [25]. The results are also consistent

with the observations on trypan blue exclusion,

which showed no evidence of induced cell death

when there is arsenic trioxide in the culture. We thus

conclude that the decrease in population growth rates

seen in the ATRA–ATO combination treatment or in

the ATO-alone treatment case is not due to

apoptosis. We next tested whether the decrease in

growth rates was attributable to an increase in the

percentage of cells arresting in the G1/G0 phase.

Arsenic trioxide does not accelerate ATRA-induced G0

arrest

ATRA treatment of HL-60 cells led to myeloid

differentiation as well as G1/G0 arrest. To determine

whether the ATO-induced growth retardation was

attributable to G1/G0 arrest, the effect of ATO on

the percentage of G1/G0 cells was measured by flow

cytometry for untreated cells and cells treated with

ATRA, ATRA–ATO combined, and ATO alone.

Treatment with ATRA caused G1/G0 arrest that was

evidenced by an increase in the percentage of cells in

G1/G0 phase, as seen before [23]. Treatment with

ATO alone, however, did not show an increase in the

percentage of cells arrested, as compared to the

control case, at 72 h [Figure 8(A)]. Treatment with

ATO plus ATRA resulted in an increase in the

percentage of G1/G0 cells, which was indistinguish-

able from that caused by ATRA alone. The effect of

ATO treatment on cell cycle arrest was also

measured at earlier time points (24 h and 48 h,

results not shown), and similar results were obtained.

Thus, although ATO enhances ATRA-induced

differentiation, it does not enhance ATRA-induced

growth inhibition through enhancing G1/G0 arrest.

Arsenic trioxide slows cell cycle transit

To determine whether ATO slowed progression

through the cell cycle, the effect of ATO on the

rate at which cells accumulated in G2/M in cultures

with mitosis blocked was measured. Cells that were

untreated or treated with ATRA, ATO, or a

combination thereof for 24 h were treated with

nocodazole, a microtubule disruptor blocking cells

in M phase. The rate of accumulation of cells in G2/

M and depletion of cells from G1 reflect the rate of

1742 S. Nayak et al.

Leu

k L

ymph

oma

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Proc

ter

& G

ambl

e Ph

arm

aceu

tical

s on

03/

14/1

1Fo

r pe

rson

al u

se o

nly.

cell cycle transit. ATO slowed the accumulation of

cells in G2/M and the depletion of G1 cells as

measured by flow cytometry. As shown in Figure 9,

cells treated with a combination of ATO plus ATRA

went through the cell cycle more slowly than cells

that were untreated or treated with just ATRA.

Addition of ATO alone or in combination with

ATRA led to statistically significantly different

percentages of cells (p5 0.05) in G2/M phase

between the control case and treatments containing

ATO, at 12 h as well as 18 h [Figure 9(A)].

Similarly, significant differences were obtained in

the percentages of cells in G1 phase when ATO

treatment was compared with the control case at 18 h

[Figure 9(B)]. Comparing Figures 8 and 9, it is of

interest to note that the rate of transit through the cell

cycle is slower (Figure 9) and the probability of arrest

is higher (Figure 8) in the ATRA plus ATO co-

treatment group. Figure 8 shows arrest after 72 h,

whereas Figure 9 shows the rate of transit during the

first 18 h of treatment.

Discussion

There are a number of potential mechanisms by

which ATO may cause or facilitate leukemic cell

differentiation. A number of regulatory molecules,

pathways, and their downstream cellular outcomes

have been studied. One of the potentially most

significant is the capability of ATO, like ATRA, to

cause degradation of PML–RARa, the fusion protein

resulting from the t(15;17) translocation that is

putatively the seminal cytogenetic cause of APL

[26]. In the NB4 APL cell line, ATO caused

degradation of the PML–RARa fusion protein, but

not the mRNA, in both ATRA sensitive and resistant

variant lines [26]. ATO also affects the spatial

localization and organization of PML and PML–

RARa, altering PML bodies within the nucleus of

NB4 cells [19,26]. Clinical correlates have also been

reported [5]. Degradation of PML–RARa is thus a

prominent potential mechanism of action of ATO.

ATO has also been found to affect signaling

pathways that have been implicated in ATRA-

induced differentiation, as well as growth regulation

and apoptosis. ATO can evoke a cellular oxidative

stress response [27,28]. The glutathione (GSH)

redox system is a critical modulator of cellular

response in oxidative stress. As such it can regulate

apoptosis, a possible sequela of ATO treatment. A

function of the GSH redox system is to scavenge free

radicals. The GSH redox system is targeted by ATO

in various APL cell lines. NB4 cells have one of the

lowest levels of basal GSH in leukemic cell lines, and

Figure 7. Addition of ATO to the culture does not lead to an increase in apoptosis. (A, C, D) Representative viable, apoptotic, and necrotic

cells as seen in the Hoechst/PI assay. (B) The number of apoptotic, necrotic, and viable cells in a representative sample when 100 cells were

counted for the different treatment cases. A positive control in the form of MMC-treated cells was also included in the experiment. As seen in

the figure, addition of ATO did not lead to an increase in apoptosis in the presence or absence of RA.

Arsenic trioxide in PML–RARa (7) cells 1743

Leu

k L

ymph

oma

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Proc

ter

& G

ambl

e Ph

arm

aceu

tical

s on

03/

14/1

1Fo

r pe

rson

al u

se o

nly.

were found to be most sensitive to arsenic treatment

[29]. In NB4 cells, ATO treatment modulated the

GSH levels, which sensitized the cells toward

apoptosis [29,30]. There were dose-dependent

growth inhibitory, differentiation enhancing, or

apoptosis enhancing effects of ATO [17,31]. Pre-

vious studies have shown that ATO causes activation

of the stress-activated protein kinase, c-jun N-

terminal kinase (JNK) [28]. This activation is directly

related to apoptosis, as inhibition of p38/JNK was

seen to reduce ATO-induced apoptosis in NB4 cells.

In the case of MAPK signaling, the effect of ATO on

p38 has been widely studied, too. Verma et al.

showed that p38 is phosphorylated and activated by

ATO in a sustained manner, and that this activation

is downstream of redox events activated by ATO [32]

in NB4 cells. Activation of other MAPK proteins,

such as Mkk 3 and Mkk 6, was also seen in NB4 cells

and in chronic myelogenous leukemia (CML)-

derived KT-1 cells, and found to be essential for

p38MAPK activation. Signaling molecules and path-

ways that are in common downstream of ATO and

ATRA may provide a basis for their potential

synergistic interaction. Clinically, a synergistic effect

has been reported for ATRA–ATO combination

treatment, when compared with either ATRA or

ATO monotherapy [10]. Other studies of the

biochemical effects of ATRA–ATO combination

treatment have revealed additive effects in JNK/

p38, tumor necrosis factor (TNF), and cyclic

adenosine monophosphate/protein kinase A (cAMP/

PKA) pathways in APL cell lines [33,34]. For

example, in the cAMP/PKA study, it was found

that cAMP, which can induce monocytic differentia-

tion in HL-60 cells, was synergistically induced by

ATRA and low doses of ATO [33,35].

The results of the present study showed that for the

PML–RARa negative HL-60 cells, ATO did not

induce differentiation, although it did slow cell

proliferation. It did enhance the differentiation

induction capability of ATRA. Although ATO did

not augment ATRA-induced CD38 expression, a

marker of the earliest stages of ATRA-induced

progression along the myeloid lineage, it did enhance

ATRA-induced expression of CD11b, an integrin

receptor subunit that marks further progression of

differentiation, as well as p47PHOX, which is a

component of the NAPDH–oxidase complex used

for the inducible oxidative metabolism characteristic

of mature myeloid cells. ATO also enhanced the

ATRA-induced functional differentiation marker,

inducible oxidative metabolism. While ATO by itself

Figure 8. (A) Arsenic trioxide does not accelerate ATRA-induced G0 arrest. The percentage of G1/G0, S, and G2/M cells at 72 h after

treatment is shown for control (untreated), ATRA, ATRA–ATO combination treated, and ATO-alone treated cells. The y-axis here shows

the percentage of cells in the different cell-cycle phases at 72 h. The x-axis shows the different treatment groups. Cells stained with hypotonic

PI solution were incubated for 1 h and analyzed by flow cytometry. * indicates that treatment with ATRA or ATRA–ATO led to a statistically

significant increase in the percentage of cells in G1/G0 with respect to control, as expected. Similar DNA histograms were generated for

earlier time points also, and we did not see a significant difference in percentages of cells arrested in G1/G0 phase between the ATRA–ATO

case and ATRA-alone case. (B) Representative DNA histograms for untreated (control) cells, cells treated with ATRA, cells treated with

ATRA–ATO combination, and cells treated with ATO alone are shown at 72 h.

1744 S. Nayak et al.

Leu

k L

ymph

oma

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Proc

ter

& G

ambl

e Ph

arm

aceu

tical

s on

03/

14/1

1Fo

r pe

rson

al u

se o

nly.

thus could not cause differentiation, it enhanced

ATRA-induced differentiation, indicating that it

affected pathways of importance to propelling differ-

entiation. One such pathway is activation of the RAF/

MEK/ERK axis in MAPK signaling [12,13]. Activa-

tion of this axis was enhanced in cells treated with the

combination of ATRA plus ATO, compared to

ATRA alone. Although phosphorylation of MEK

and ERK betraying activation was enhanced, their

expression levels were not detectably affected. The

activation and expression patterns of MAPK proteins

suggest that the synergy between ATRA and ATO is

at the protein–protein interaction level, rather than at

the transcriptional level. This agrees with a recent

systems-level study which indicated that while the

effects of ATRA involved transcriptional remodeling,

ATO effects were seen at a proteome level, although

the study was done for NB4 cells [34].

The present results also showed that ATO

retarded population growth, and that populations

Figure 9. Addition of ATO leads to a general dilatation in cell cycle. The percentage of cells with G2/M DNA (A) or G1 DNA (B) for cells

that were untreated or treated with ATRA, ATO, or ATRA plus ATO for 24 h and 100 ng/mL nocodazole thereafter for 0, 6, 12, 18 h is

shown. (A) Treatment with ATO (with or without ATRA) leads to a statistically significantly (* and **, p50.05) different percentage of cells

in G2/M phase at 18 h when compared with the control case. # and ## indicate statistically significant differences for 12 h. (B) { and {{indicate that the percentage of cells in G1 phase is statistically different from the control case at 18 h when ATO (with or without ATRA) is

added to the medium. { and {{ show statistically significantly different cases at 12 h, when compared to control. Representative DNA

histograms of control (untreated), ATRA-, ATO-, and ATRA plus ATO-treated populations after 18 h in nocodazole are shown (C).

Arsenic trioxide in PML–RARa (7) cells 1745

Leu

k L

ymph

oma

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Proc

ter

& G

ambl

e Ph

arm

aceu

tical

s on

03/

14/1

1Fo

r pe

rson

al u

se o

nly.

treated with ATO plus ATRA were more growth-

inhibited than those treated with ATRA alone. The

viability of cultures detected by trypan blue exclusion

routinely exceeded 95%, and as seen conclusively

from the Hoechst assay there was no increase in

apoptosis upon ATO addition. There was also no

extra accumulation of cells in G0 in ATO plus

ATRA-treated cells compared to ATRA-treated

cells. There was an ATO-induced retardation of

cell cycle progression.

In summary, in PML–RARa negative HL-60

human myeloblastic leukemia cells, ATO causes

MAPK signaling and augments ATRA-induced

differentiation. The effects are more prominent as

the MAPK signal augmentation increases, enhancing

expression of later differentiation markers, but not

the earliest detected. A further potential contributor

to the enhanced differentiation by ATO is the

retardation of growth, possibly slowing down the

self-renewal capacity of these progenitor cells and

favoring differentiation. The results show that ATO

may be of therapeutic use to augment the effects of

ATRA in a PML–RARa negative context.

Acknowledgements

The authors would like to thank Dr. Deanna

Schaefer for her kind help in Wright’s staining of

slides. Additionally, we acknowledge the careful

review of the manuscript by Ryan Tasseff of Prof.

Jeffrey Varner’s research group.

Declaration of interest: This work was supported

in part by grants from NIH (USPS) (CA 033505),

NYSTEM, NY Department of Health, and NSF

CAREER #CBET-0846876.

References

1. Waxman S, Anderson KC. History of the development of

arsenic derivatives in cancer therapy. Oncologist 2001;6:3–10.

2. Evens AM, Tallman MS, Gartenhaus RB. The potential of

arsenic trioxide in the treatment of malignant disease: past,

present, and future. Leuk Res 2004;28:891–900.

3. Wang ZY, Chen Z. Acute promyelocytic leukemia: from

highly fatal to highly curable. Blood 2008;111:2505–2515.

4. Niu C, Yan H, Yu T, et al. Studies on treatment of acute

promyelocytic leukemia with arsenic trioxide: remission

induction, follow-up, and molecular monitoring in 11 newly

diagnosed and 47 relapsed acute promyelocytic leukemia

patients. Blood 1999;94:3315–3324.

5. Shen ZX, Chen GQ, Ni JH, et al. Use of arsenic trioxide

(As2O3) in the treatment of acute promyelocytic leukemia

(APL): II. Clinical efficacy and pharmacokinetics in relapsed

patients. Blood 1997;89:3354–3360.

6. Mathews V, George B, Lakshmi KM, et al. Single-agent

arsenic trioxide in the treatment of newly diagnosed acute

promyelocytic leukemia: durable remissions with minimal

toxicity. Blood 2006;107:2627–2632.

7. Breitman T, Collins S, Keene B. Terminal differentiation of

human promyelocytic leukemic cells in primary culture in

response to retinoic acid. Blood 1981;57:1000–1004.

8. Huang M, Ye Y, Chen S, et al. Use of all-trans retinoic acid in

the treatment of acute promyelocytic leukemia. Blood

1988;72:567–572.

9. Asou N, Kishimoto Y, Kiyoi H, et al. A randomized

study with or without intensified maintenance chemo-

therapy in patients with acute promyelocytic leukemia

who have become negative for PML-RARalpha transcript

after consolidation therapy: The Japan Adult Leukemia

Study Group (JALSG) APL97 study. Blood 2007;110:59–

66.

10. Shen ZX, Shi ZZ, Fang J, et al. All-trans retinoic acid/As2O3

combination yields a high quality remission and survival in

newly diagnosed acute promyelocytic leukemia. Proc Natl

Acad Sci USA 2004;101:5328–5335.

11. Zhang XW, Yan XJ, Zhou ZR, et al. Arsenic trioxide controls

the fate of the PML–RARa oncoprotein by directly binding

PML. Science 2010;328:240–243.

12. Yen A, Roberson MS, Varvayanis S, Lee AT. Retinoic acid

induced mitogen-activated protein (MAP)/extracellular signal-

regulated kinase (ERK) kinase-dependent MAP kinase activa-

tion needed to elicit HL-60 cell differentiation and growth

arrest. Cancer Res 1998;58:3163–3172.

13. Wang J, Yen A. A MAPK-positive feedback mechanism

for BLR1 signaling propels retinoic acid-triggered differ-

entiation and cell cycle arrest. J Biol Chem 2008;283:4375–

4386.

14. Perkins C, Kim C, Fang G, Bhalla K. Arsenic induces

apoptosis of multidrug-resistant human myeloid leukemia

cells that express Bcr-Abl or overexpress MDR, MRP, Bcl-2,

or Bcl-xL. Blood 2000;95:1014–1022.

15. Herst P, Hesketh E, Ritchie D, Berridge M. Glycolytic

metabolism confers resistance to combined all-trans retinoic

acid and arsenic trioxide-induced apoptosis in HL60rho0

cells. Leuk Res 2008;32:327–333.

16. Jiang G, Albihn A, Tang T, Tian Z, Henriksson M. Role of

Myc in differentiation and apoptosis in HL60 cells after

exposure to arsenic trioxide or all-trans retinoic acid. Leuk Res

2008;32:297–307.

17. Chen GQ, Zhu J, Shi XG, et al. In vitro studies on cellular and

molecular mechanisms of arsenic trioxide (As2O3) in the

treatment of acute promyelocytic leukemia: As2O3 induces

NB4 cell apoptosis with downregulation of Bcl-2 expression

and modulation of PML-RAR alpha/PML proteins. Blood

1996;88:1052–1061.

18. Fernandez C, Ramos AM, Sancho P, Amran D, de Blas E,

Aller P. 12-O-Tetradecanoylphorbol-13-acetate may both

potentiate and decrease the generation of apoptosis by the

antileukemic agent arsenic trioxide in human promonocytic

cells. J Biol Chem 2004;279:3877–3884.

19. Huang C, Ma W, Li J, Dong Z. Arsenic induces apoptosis

through a c-Jun NH2-terminal kinase-dependent, p53-inde-

pendent pathway. Cancer Res 1999;59:3053–3058.

20. Kishimoto H, Hoshino S, Ohori M, et al. Molecular

mechanism of human CD38 gene expression by retinoic

acid identification of retinoic acid response element in the first

intron. J Biol Chem 1998;273:15429–15434.

21. Shen M, Yen A. c-Cbl interacts with CD38 and promotes

retinoic acid-induced differentiation and G0 arrest of human

myeloblastic leukemia cells. Cancer Res 2008;68:8761–

8769.

22. Reiterer G, Bunaciu RP, Smith JL, Yen A. Inhibiting the

platelet derived growth factor receptor increases signs of

retinoic acid syndrome in myeloid differentiated HL-60 cells.

FEBS Lett 2008;582:2508–2514.

1746 S. Nayak et al.

Leu

k L

ymph

oma

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Proc

ter

& G

ambl

e Ph

arm

aceu

tical

s on

03/

14/1

1Fo

r pe

rson

al u

se o

nly.

23. Shen M, Yen A. Nicotinamide cooperates with retinoic acid

and 1,25-dihydroxyvitamin D3 to regulate cell differentiation

and cell cycle arrest of human myeloblastic leukemia cells.

Oncology 2009;76:91–100.

24. Muscarella DE, Rachlinski MK, Sotiriadis J, Bloom SE.

contribution of gene-specific lesions, DNA-replication-asso-

ciated damage, and subsequent transcriptional inhibition-

mediated apoptosis in lymphoma cells. Exp Cell Res

1998;238:155–167.

25. Lamkin TJ, Chin V, Varvayanis S, et al. Retinoic acid-induced

CD38 expression in HL-60 myeloblastic leukemia cells

regulates cell differentiation of viability depending on expres-

sion levels. J Cell Biochem 2006;97:1328–1338.

26. Shao W. Arsenic trioxide as an inducer of apoptosis and loss of

PML/RAR alpha protein in acute promyelocytic leukemia

cells. J Natl Cancer Inst 1998;90:124–133.

27. Jing Y, Dai J, Chalmers-Redman RME, Tatton WG, Waxman

S. Arsenic trioxide selectively induces acute promyelocytic

leukemia cell apoptosis via a hydrogen peroxide-dependent

pathway. Blood 1999;94:2102–2111.

28. Davison K, Mann KK, Waxman S, Miller WH. JNK activation

is a mediator of arsenic trioxide-induced apoptosis in acute

promyelocytic leukemia cells. Blood 2004;103:3496–3502.

29. Dai J, Weinberg RS, Waxman S, Jing Y. Malignant cells can

be sensitized to undergo growth inhibition and apoptosis by

arsenic trioxide through modulation of the glutathione redox

system. Blood 1999;93:268–277.

30. Ramos AM, Fernandez C, Amran D, Sancho P, de Blas E,

Aller P. Pharmacologic inhibitors of PI3K/Akt potentiate

the apoptotic action of the antileukemic drug arsenic

trioxide via glutathione depletion and increased peroxide

accumulation in myeloid leukemia cells. Blood 2005;105:

4013–4020.

31. Chen GQ, Shi XG, Tang W, et al. Use of arsenic trioxide

(As2O3) in the treatment of acute promyelocytic leukemia

(APL): I. As2O3 exerts dose-dependent dual effects on APL

cells. Blood 1997;89:3345–3353.

32. Verma A, Mohindru M, Deb DK, et al. Activation of Rac1

and the p38 mitogen-activated protein kinase pathway in

response to arsenic trioxide. J Biol Chem 2002;277:44988–

44995.

33. Zhao Q, Tao J, Zhu Q, et al. Rapid induction of cAMP/

PKA pathway during retinoic acid-induced acute promyelo-

cytic leukemia cell differentiation. Leukemia 2003;18:285–

292.

34. Zheng PZ, Wang KK, Zhang QY, et al. Systems analysis

of transcriptome and proteome in retinoic acid/arsenic

trioxide-induced cell differentiation/apoptosis of promyelo-

cytic leukemia. Proc Natl Acad Sci USA 2005;102:7653–

7658.

35. Zhu Q, Zhang JW, Zhu HQ, et al. Synergic effects of arsenic

trioxide and cAMP during acute promyelocytic leukemia cell

maturation subtends a novel signaling cross-talk. Blood 2002;

99:1014–1022.

Arsenic trioxide in PML–RARa (7) cells 1747

Leu

k L

ymph

oma

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Proc

ter

& G

ambl

e Ph

arm

aceu

tical

s on

03/

14/1

1Fo

r pe

rson

al u

se o

nly.