14
Staurosporine modulates radiosensitivity and radiation-induced apoptosis in U937 cells HOW-RAN GUO 1 , CHIA-HSIN CHEN 1 , SHENG-YOW HO 2,3 , YUAN-SOON HO 4 , RONG-JANE CHEN 1 & YING-JAN WANG 1 1 Department of Environmental and Occupational Health, and 2 Institute of Basic Medical Sciences, National Cheng Kung University, Medical College, Tainan, 3 Sinlau Christian Hospital, Tainan, Taiwan, and 4 Institute of Biomedical Technology, Taipei Medical University, Taipei, Taiwan (Received 24 July 2005; revised 20 January 2006; accepted 23 January 2006) Abstract Purpose: The present study aims at investigating the involvement of several genes in the cell cycle distribution and apoptosis in U937 cells, a cell line lacking functional p53 protein, after combined treatment with staurosporine and irradiation. Materials and methods: Using a DNA fragmentation assay, flow cytometry and western blot analysis, the molecular basis for the effects of staurosporine in combination with the irradiation of leukemia cells was investigated. Results: Our results indicated that combined treatment led to an increased apoptotic cell death in U937 cells, which is correlated with the phosphorylation of the V-Jun sarcoma virus 17 oncogene homolog (c-JUN) NH 2 -terminal kinase protein (JNK), the activation of caspases, the increase in B cell leukemia/lymphoma 2 (Bcl-2) associated X protein (Bax), the decrease in Bcl xL protein (Bcl-XL) levels, the loss of mitochondria membrane potential and the release of cytochrome c. Conclusions: Abrogation of the G2 checkpoint should be an effective strategy against p53-deficient leukemia cells to irradiation-induced cell killing. Keywords: Apoptosis, cell cycle checkpoints, chemical modifiers, leukaemias, radiotherapy Introduction Ionizing radiation (IR) is one of the most effective tools in the clinical treatment of cancer. However, the success of radiotherapy is far from assured. Increasing the sensitivity of tumor cells to the lethal effects of radiation has the potential to improve the efficacy of radiotherapy (Pawlik & Keyomarsi 2004). The induction of DNA double-strand breaks was considered the major mechanism of IR-induced cell death. The cellular response to DNA damage involves a cell-cycle arrest at both the G1/S and G2/M transitions; these checkpoints maintain viabi- lity by preventing the replication or segregation of damaged DNA. The G1 arrest involves the p53- mediated induction of p21 Wild-type p53-activated Fragment 1/Cyclin-dependent kinase inhibitor (p21WAF1/CIP1), whereas the G2 arrest involves the inactivation of cell cycle p34 cdc2 kinase protein (p34cdc2 kinase) (Maity et al. 1994). Following DNA damage, p53-deficient cells fail to arrest at G1 and accumulate at the G2/M transition. The status of p53 is pivotal for the response of tumor cells to IR. Irradiation of cells with wild-type p53 gene elevates the level of cellular p53 protein and regulates the expression of a variety of down- stream effector genes. Mutations in the p53 gene are involved in acquired and intrinsic treatment resis- tance in human tumors and render tumor cells refractory to many anticancer therapies (Kinzler & Vogelstein 1996, Giaccia & Kastan 1998). The radioresistance of tumor cells lacking p53 may be a consequence of a diminished ability to undergo apoptosis in vitro and in vivo (Lowe et al. 1994). Thus, the use of chemical modifiers as radio- sensitizers in combination with low-dose irradiation may increase the therapeutic effect by overcoming a high apoptotic threshold. Correspondence: Dr Ying-Jan Wang, Department of Environmental and Occupational Health, National Cheng Kung University Medical College, 138 Sheng-Li Road, Tainan, Taiwan 704. Tel: 886 6 235 3535 ext. 5804. Fax: 886 6 2752484. E-mail: [email protected] Int. J. Radiat. Biol., Vol. 82, No. 2, February 2006, pp. 97 – 109 ISSN 0955-3002 print/ISSN 1362-3095 online Ó 2006 Taylor & Francis DOI: 10.1080/09553000600589149 Int J Radiat Biol Downloaded from informahealthcare.com by Technische Universiteit Eindhoven on 11/23/14 For personal use only.

Staurosporine modulates radiosensitivity and radiation-induced apoptosis in U937 cells

Embed Size (px)

Citation preview

Page 1: Staurosporine modulates radiosensitivity and radiation-induced apoptosis in U937 cells

Staurosporine modulates radiosensitivity and radiation-inducedapoptosis in U937 cells

HOW-RAN GUO1, CHIA-HSIN CHEN1, SHENG-YOW HO2,3, YUAN-SOON HO4,

RONG-JANE CHEN1 & YING-JAN WANG1

1Department of Environmental and Occupational Health, and 2Institute of Basic Medical Sciences, National Cheng Kung

University, Medical College, Tainan, 3Sinlau Christian Hospital, Tainan, Taiwan, and 4Institute of Biomedical Technology,

Taipei Medical University, Taipei, Taiwan

(Received 24 July 2005; revised 20 January 2006; accepted 23 January 2006)

AbstractPurpose: The present study aims at investigating the involvement of several genes in the cell cycle distribution and apoptosisin U937 cells, a cell line lacking functional p53 protein, after combined treatment with staurosporine and irradiation.Materials and methods: Using a DNA fragmentation assay, flow cytometry and western blot analysis, the molecular basis forthe effects of staurosporine in combination with the irradiation of leukemia cells was investigated.Results: Our results indicated that combined treatment led to an increased apoptotic cell death in U937 cells, which iscorrelated with the phosphorylation of the V-Jun sarcoma virus 17 oncogene homolog (c-JUN) NH2-terminal kinase protein(JNK), the activation of caspases, the increase in B cell leukemia/lymphoma 2 (Bcl-2) associated X protein (Bax), thedecrease in Bcl xL protein (Bcl-XL) levels, the loss of mitochondria membrane potential and the release of cytochrome c.Conclusions: Abrogation of the G2 checkpoint should be an effective strategy against p53-deficient leukemia cells toirradiation-induced cell killing.

Keywords: Apoptosis, cell cycle checkpoints, chemical modifiers, leukaemias, radiotherapy

Introduction

Ionizing radiation (IR) is one of the most effective

tools in the clinical treatment of cancer. However,

the success of radiotherapy is far from assured.

Increasing the sensitivity of tumor cells to the lethal

effects of radiation has the potential to improve the

efficacy of radiotherapy (Pawlik & Keyomarsi 2004).

The induction of DNA double-strand breaks was

considered the major mechanism of IR-induced cell

death. The cellular response to DNA damage

involves a cell-cycle arrest at both the G1/S and

G2/M transitions; these checkpoints maintain viabi-

lity by preventing the replication or segregation of

damaged DNA. The G1 arrest involves the p53-

mediated induction of p21 Wild-type p53-activated

Fragment 1/Cyclin-dependent kinase inhibitor

(p21WAF1/CIP1), whereas the G2 arrest involves

the inactivation of cell cycle p34 cdc2 kinase protein

(p34cdc2 kinase) (Maity et al. 1994). Following

DNA damage, p53-deficient cells fail to arrest at G1

and accumulate at the G2/M transition.

The status of p53 is pivotal for the response of

tumor cells to IR. Irradiation of cells with wild-type

p53 gene elevates the level of cellular p53 protein

and regulates the expression of a variety of down-

stream effector genes. Mutations in the p53 gene are

involved in acquired and intrinsic treatment resis-

tance in human tumors and render tumor cells

refractory to many anticancer therapies (Kinzler &

Vogelstein 1996, Giaccia & Kastan 1998). The

radioresistance of tumor cells lacking p53 may be a

consequence of a diminished ability to undergo

apoptosis in vitro and in vivo (Lowe et al. 1994).

Thus, the use of chemical modifiers as radio-

sensitizers in combination with low-dose irradiation

may increase the therapeutic effect by overcoming a

high apoptotic threshold.

Correspondence: Dr Ying-Jan Wang, Department of Environmental and Occupational Health, National Cheng Kung University Medical College, 138

Sheng-Li Road, Tainan, Taiwan 704. Tel: 886 6 235 3535 ext. 5804. Fax: 886 6 2752484. E-mail: [email protected]

Int. J. Radiat. Biol., Vol. 82, No. 2, February 2006, pp. 97 – 109

ISSN 0955-3002 print/ISSN 1362-3095 online � 2006 Taylor & Francis

DOI: 10.1080/09553000600589149

Int J

Rad

iat B

iol D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y T

echn

isch

e U

nive

rsite

it E

indh

oven

on

11/2

3/14

For

pers

onal

use

onl

y.

Page 2: Staurosporine modulates radiosensitivity and radiation-induced apoptosis in U937 cells

Depending on the stimulus that initiates apoptosis,

different caspase cascades are activated (Rocha et al.

2000). Among the different caspases, caspase-3 plays

a major role in the effector phase of apoptosis

induced by a variety of stimuli (Abu-Qare & Abou-

Donia 2001). It has been demonstrated that the

activation of caspase-3 is regulated by at least two

mechanisms: One involves a direct pathway from

caspase-8 and the other is mediated by capase-9

following the release of cytochrome c from mito-

chondria (Shimizu et al. 1999). Upstream of the

caspase cascades pathway, mitogen-activated protein

kinase (MAPK) signal transduction pathways have

been shown to play a key role in the cellular response

to extracellular stimuli (Davis 1994, Cano &

Mahadevan 1995, Herskowitz 1995, Waskiewicz &

Cooper 1995). Three major mammalian MAPK

subgroups have been identified, including extra-

cellular signal-regulated kinase (ERK), c-JUN (V-Jun

sarcoma virus 17 oncogene homolog) NH2-terminal

kinase (JNK), and p38 MAPK (Ip & Davis 1998).

Among these, the JNK pathway is involved in many

forms of stress-induced apoptosis and is the pathway

most strongly activated by stress stimuli, such as

ultraviolet (UV) and IR (Johnson et al. 1996).

Abrogation of the G2 checkpoint has been

associated with the sensitivity of tumor cells to

DNA-damaging agents (Powell et al. 1995, Bracey

et al. 1997). Staurosporine (STP) was originally

isolated from a Streptomyces species as an inhibitor

of protein kinase C (PKC) (Omura et al. 1977). STP

and its analogues have anti-tumor properties alone

and have also been shown to abrogate the G2

checkpoint and to sensitize tumor cells to DNA-

damaging agents (Gil et al. 2003). It has been

reported that the abrogation of the radiation-induced

G2 checkpoint by STP and its analogues is

associated with radiosensitivity in several tumor cell

lines, including colorectal tumor cells, fibrosarcoma

cells and ovarian carcinoma cells (Heerdt et al. 2000,

Wang et al. 2001, Zaugg et al. 2001, Playle et al.

2002). However, the molecular basis for the effects of

STP in combination with the irradiation of leukemia

cells has seldom been thoroughly studied. In this

study, we investigated the involvement of several

genes in the cell cycle distribution and apoptosis in

U937 cells, a cell line lacking functional p53 protein,

after combined treatment with STP and IR.

Materials and methods

Cell culture, drug treatment, and irradiation conditions

U937 cells, a human pre-monocytic leukemia cell

line, were obtained from the American Type Culture

Collection. U937 cells were cultured in RPMI

1640 medium (developed by Moore et al. at Roswell

Park Memorial Institute) (Life Technology, Grand

Island, NY, USA) supplemented with antibiotics

containing 100 U/ml penicillin, 100 mg/ml strepto-

mycin (Life Technology, Grand Island, NY, USA),

and 10% heat-inactivated fetal calf serum (FCS)

(HyClone, South Logan, UT, USA.), at 378C in a

5% carbon dioxide atmosphere. For exposure to

staurosporine (Sigma Chemical Co. St Louis, MO,

USA.), the reagent was added in concentrated form

to the culture medium and mixed gently. The

cultures were then incubated for the times indicated

in the figures. Irradiation was performed with 6 MV

X-rays using a linear accelerator (Digital M Meva-

tron Accelerator, Siemens Medical Systems, CA,

USA) at a dose rate of 5 Gy/min. An additional 2 cm

of tissue-equivalent bolus was placed on the top of

a plastic tissue culture flask to ensure electronic

equilibrium, and 10 cm tissue-equivalent material

was placed under the flask to obtain full back-scatter.

DNA fragmentation assay

The control and treated cells were grown in 75-T

culture flasks. After treatment, both groups of cells

were harvested, washed twice with ice-cold phosphate

buffered saline (PBS), suspended in TNE (10 mM

tris[Hydroxymethyl]aminomethane, HCl [Tris-HCl],

pH 7.6; 140 mM sodium chloride; and 1 mM

Disodium ethylenediaminetetraacetate [EDTA]),

and lysed at 378C in 4 ml of extraction buffer

(10 mM Tris-HCl, pH 8.0; 0.1 M EDTA, pH 8.0;

20 mg/ml pancreatic RNase; and 0.5% Sodium dode-

cyl sulphate [SDS]). (All chemicals are purchased

from Sigma Chemical Co. St Louis, MO, USA). After

2 h, proteinase K was added to a final concentration of

100 mg/ml, and the mixture was incubated for another

3 h at 508C. The DNA was extracted twice with equal

volumes of phenol, and once with chloroform-isoamyl

alcohol (24:1 V:V). The DNA was then precipitated

with 0.2 volumes of sodium acetate, pH 4.8, and

2.5 volumes of ethanol at 7208C overnight, then

pelleted at 13000 g for 1 h. The samples underwentelectrophoresis in a 1.5% agarose gel. The DNA wasmade visible by ethidium bromide (EtBr) staining.

Flow cytometry

1.56106 human pre-monocytic leukemia U937 cells

were suspended with ice-cold PBS and fixed in 70%

ethanol at 7208C for at least 1 h. After fixation, the

cells were washed twice, incubated in 0.5 ml of 0.5%

Triton X-100/PBS at 378C for 30 min with 1 mg/ml

of RNase A, and stained with 0.5 ml of 50 mg/ml

propidium iodide for 10 min. Fluorescence emitted

from the propidium iodide-DNA complex was

analysed at 488 nm/600 nm (excitation/emission

wavelength) by fluorescence-activated cell sorter

98 H.-R. Guo et al.

Int J

Rad

iat B

iol D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y T

echn

isch

e U

nive

rsite

it E

indh

oven

on

11/2

3/14

For

pers

onal

use

onl

y.

Page 3: Staurosporine modulates radiosensitivity and radiation-induced apoptosis in U937 cells

(FACScan flow cytometry). The population of nuclei

in each phase of the cell cycle was determined using

established CellFIT DNA analysis software (Becton

Dickenson, San Jose, CA, USA).

Western blot analysis

Treated and untreated cells were rinsed three times

with ice-cold PBS pelleted at 8006g for 5 min, and

lysed in 500 ml of freshly prepared extraction buffer

(10 mM Tris-HCl pH 7, 140 mM sodium chloride,

3 mM magnesium chloride, 0.5% [v/v] Nonidet P-

40 [NP-40], 2 mM phenylmethylsulfonyl fluoride,

1% [w/v] aprotinin, and 5 mM dithiothreitol

[DTT]), for 20 min on ice. The extracts were

centrifuged for 30 min at 10,0006g. Proteins were

loaded at 50 mg/lane on 12% [w/v] SDS-polyacryla-

mide gel (SDS-PAGE), blotted, and probed using

specific antibodies, including p19, p21/Cip1, p27,

proliferating cell nuclear antigen (PCNA), Retino-

blastoma-binding protein (RBBP), B-cell leukemia/

lymphoma 2 (bcl-2), bax, bcl-xl, cyclin A, cyclin B,

cyclin E, cdc2, cyclin dependent kinase 2 (cdk2),

retinoblastoma (RB2), cyclin D3, cdk4, caspase 3,

caspase 8, caspase 9, poly(ADP-ribose) polymerase

(PARP), cytochrom C, JNK, phospho-JNK (Trans-

duction Laboratories, Lexington, KY), and

phospho-cdc2 (specific for phospho-Tyr 15, Cell

Signaling, Beverly, MA, USA). Caspase 3, caspase 9,

PARP, phospho- cdc2, JNK and phospho-JNK were

detected using a chemiluminescence (ECL) detec-

tion system (Amersham Life Science, Arlington

Heights, IL, USA). Others immunoreactive bands

were visualized through incubation with the colori-

genic substrates, nitro blue tetrazolium, and 5-

bromo-4-chloro-3-indolyl-phosphate (NBT/BCIP)

(Sigma Chemical Co.) The expression of Glycer-

aldehyde-3-phosphate dehydrogenase (GAPDH)

was used as the control for equal protein loading.

Analysis of mitochondrial transmembrane potential

The change in mitochondrial transmembrane potential

was monitored by flow cytometry. Briefly, U937 cells

were collected and suspended in 1 ml PBS. Mito-

chondrial transmembrane potential was measured

directly using 40 nM 3,30- dihexyloxacarbocyanine

(DiOC6(3); Molecular Probes, Eugene, Ore., USA).

Fluorescence was measured after staining the cells for

15 min at 378C.

Statistical analysis

Data are expressed as mean+SD. Statistical

significance was determined by using the Student’s

t-test for comparison between the means. Difference

was considered significant when p5 0.05.

Results

STP enhances apoptotic cell death and abrogates the G2

arrest induced by 5 Gy IR

As shown in Figure 1A, treatment with 5 Gy IR

alone for 24 h did not induce DNA fragmentation in

U937 cells. In contrast, pretreatment with a low dose

Figure 1. Characterization of apoptosis in U937 cells treated with

low-dose of STP combined with IR. (A) Time course of DNA

fragmentation after treatment with STP 10 nM and STP 50 nM

for 24 h as the positive control was analysed by 1.8% agarose gel

electrophoresis. (B) Cells were pre-treated with 10 nM STP for 1

or 4 h before 5 Gy IR for various time periods. IR alone was

treated for 24 h. (C) Quantification of apoptotic cells treated with

10 nM STP and 5 Gy IR alone or combination for 18 h. Apoptotic

fraction was recognized as sub-G1 population of cell cycle

measured by flow cytometry. (C: control; IR: 5Gy irradiation);

*p5 0.05 vs. STP.

Staurosporine modulates radiosensitivity in U937 cells 99

Int J

Rad

iat B

iol D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y T

echn

isch

e U

nive

rsite

it E

indh

oven

on

11/2

3/14

For

pers

onal

use

onl

y.

Page 4: Staurosporine modulates radiosensitivity and radiation-induced apoptosis in U937 cells

of STP (10nM), 1 or 4 h before 5 Gy IR induced

DNA fragmentation at 18 and 15 h, respectively.

DNA fragmentation was observed at 24 h in the

positive control group (treated with STP 50 nM),

whereas STP 10 nM alone did not induce DNA

fragmentation in a time-dependent manner (Figure

1B). Quantitative analyses of DNA fragmentation

showed that combined treatment with STP and 5 Gy

IR induced an approximate 20% apoptotic cell death

rate, whereas, STP or 5 Gy IR alone induced less

than a 10% of apoptosis rate (Figure 1C).

Figure 2 shows the cell cycle progression in U937

cells treated with STP and 5 Gy IR. When cells were

treated with 5 Gy IR alone, 62% of the cells were in

G2/M phase. When treated with 10 nM STP and 5

Gy IR, 20% of the cells were in G2/M phase, and

21% of the cells were in sub-G1 phase, respectively.

In addition, 46% of the cells were in G1 phase of

combined treatment, significantly more than those

treated with 5 Gy IR alone (24%). These results

suggest that combined treatment can override

IR-induced G2/M arrest and increase the number

of apoptotic cells through increasing G1 phase

progression.

Expression of cell cycle regulators in U937 cells treated

with STP and IR

To examine the molecular mechanisms of the

abrogation of G2/M arrest and the increase in the

G1 phase in U937 cells treated with STP and IR, we

studied the expression of cell cycle G1/S and G2/M

regulatory proteins. Figure 3 illustrates that 5 Gy IR

alone resulted in a remarkable increase in the

accumulation of cyclin A and cyclin B as well as a

slight accumulation of cdc2. The observations

suggested that 5 Gy IR may lead to phosphorylation

of cdc2 on Thr-14 and/or Tyr-15 and thus result in a

G2/M arrest. Figure 3 shows that phosphorylation of

cdc2 on Tyr-15 could be detected in cells treated

with 5 Gy IR, whereas STP in combination with IR

inhibited the phosphorylation of cdc2 stimulated by

Figure 2. Cell cycle progression in U937 cells treated with STP

and IR. Proportion (A) and quantification (B) of cell cycle phase

was analyzed by flow cytomety. U937 cells were treated with STP,

IR or combination for 18h. #p5 0.05 vs. 5 Gy alone.

Figure 3. Effects of combined treatment with STP and IR on the

expression of cell cycle regulators. Cells were treated with 10 nM

STP, 5 Gy IR alone or combined treatment for 18 h. (A) The

expression of G2/M-relating proteins was shown. (B) The

expression of G1/S-relating proteins was shown. The level of total

GAPDH protein was used as controls for equal loading of protein

in different lanes.

100 H.-R. Guo et al.

Int J

Rad

iat B

iol D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y T

echn

isch

e U

nive

rsite

it E

indh

oven

on

11/2

3/14

For

pers

onal

use

onl

y.

Page 5: Staurosporine modulates radiosensitivity and radiation-induced apoptosis in U937 cells

IR alone. These results indicated that STP may

abrogate the G2/M arrest induced by 5 Gy IR

through decreasing phosphorylation of cdc2 on Tyr-

15. Figure 3 also showed that combined treatment

with STP and 5 Gy IR increased G1/S regulatory

proteins such as p21, cyclin E and Rb2, as opposed

to 5 Gy IR alone. The expression of CDK4 and p27

decreased slightly when compared to 5 Gy IR alone.

These results indicated that combined treatment

could override G2/M arrest through decreasing the

expression of G2/M-related proteins such as cyclin

A, cyclin B, cdc2 and reducing phosphorylation of

cdc2 on Tyr-15, whereas an increased G1 phase

occurred through regulatory proteins such as cyclin

E, p21, and Rb2.

Cytochrome c-mediated caspases activation in U937 cells

treated with STP and IR

Figure 4A shows the western blot analysis of the pro-

caspase enzymes and their activated cleavage forms.

All the caspase-3, caspase-9, and caspase-8 cleavage

forms could be detected in U937 cells with

combined STP and IR treatment. Poly (ADP-ribose)

polymerase, PARP, has been identified as a substrate

for caspase-3. The cleavage of PARP by the activated

caspase-3 results in the formation of an 85 kDa C-

terminal fragment. Our results also showed that the

specific cleavage of PARP could be found in cells

with combined treatment (Figure 4B). To further

confirm the contribution of caspase activation in the

induction of apoptosis in cells, a selective peptide

inhibitor for activated caspase-3 was used. We found

that the pre-incubation of cells with inhibitor

Z-VAD-FMK (a broad inhibitor of caspase-3, -6, -7)

could attenuate the apoptotic cell death of combined

treatment with STP and IR (Figure 4C).

The fluorescent probe 3,30- dihexyloxacarbocy-

nine was used to measure the mitochondrial

transmembrane potential (DCm), and the release of

mitochondrial cytochrome c was detected by Wes-

tern blotting. Figure 5 shows that minor changes in

DCm and a slight release of cytochrome c could be

observed in cells treated with either STP or IR alone.

However, combined treatment caused an obvious

change in the mitochondrial potential (DCm) and in

the release of cytochrome c, when compared to the

control and STP or IR alone (Figure 5A). Mean-

while, the expression levels of the Bax proteins were

elevated in the cells with combined treatment when

compared to treatment with STP or IR alone,

whereas the expression levels of the Bcl-xL proteins

declined with combined treatment (Figure 5B).

These results suggested that cytochrome c-mediated

the caspase-9 and caspase-3 activation involved in

the induction of apoptosis in cells with combined

treatment.

Activation of JNK signaling pathway in U937 cells

treated with STP and IR

To investigate whether the JNK signaling pathway

was involved in U937 cells with combined treatment,

phosphorylated (active form) JNK protein was

detected by western blotting. Our results showed

that the activation of JNK increased slightly at

15 – 30 min when treated with 5 Gy IR and 10 nM

STP alone, whereas, a prolonged activation of JNK

could be observed from 30 – 120 min after combined

treatment (Figure 6A, 6B). We used the specific

inhibitor of JNK (SP600125) to further confirm the

involvement of the JNK signaling pathway in the

apoptosis and cell cycle regulation altered by STP

combined with 5 Gy IR. Pretreatment of the U937

cells with SP600125 resulted in an alteration of cell

cycle progression and an inhibition of apoptotic cells.

As shown in Figure 6C, when U937 cells treated

with SP600125 alone or in combination with 5 Gy

IR, the proportion of cells in G2/M phase increased

to 65.74% and 59.44%, respectively. These results

supported the theory that basal JNK plays an

important role in G2/M transition. In addition, a

diminution of apoptotic cells was observed in cells

pretreated with SP600129 in comparison with

combined treatment (4.8% vs. 20.1%). The propor-

tion of cells at G2/M phase arrest was 36.7% when

pretreated with SP600125 (Figure 6C), and was

20.75% without the pretreatment (Figure 6D).

Prolonged activation of the JNK pathway could

trigger an increased apoptosis in U937 cells treated

with STP combined with 5 Gy IR.

Discussion

G2/M arrest and apoptosis are commonly observed

in cells treated with DNA-damaging agents, includ-

ing irradiation (Miyata et al. 2001b). It has been

shown that early apoptosis could be induced in U937

cells by a high dose of irradiation, whereas G2/M

arrest was induced by a low dose of radiation.

Shinomiya et al. (2000) found that U937 cells

treated with low dose (5 Gy) IR could induce G2/

M arrest at 12 – 36 h, then cells were released from

the blockade and entered G1 phase at 36 – 48 h,

when sub-G1 fraction, namely apoptosis, become

obvious. However, after high dose (20 Gy) IR

treatment, the S and G2/M phase fractions rapidly

disappeared, and an increase of sub-G1 fraction was

detectable in 6 h. They suggested that an execution

of apoptosis after high dose (20 Gy) IR is an early

and premitotic event, whereas apoptosis following

low dose (5 Gy) IR occurs after the release of G2/M

blockage (postmitotic event) and may be executed at

G1 phase. Moreover, many studies indicated that

cancer cells with mutant or deleted p53 such as

Staurosporine modulates radiosensitivity in U937 cells 101

Int J

Rad

iat B

iol D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y T

echn

isch

e U

nive

rsite

it E

indh

oven

on

11/2

3/14

For

pers

onal

use

onl

y.

Page 6: Staurosporine modulates radiosensitivity and radiation-induced apoptosis in U937 cells

U937 cells, exploit the absence of G1/S checkpoint

but accumulate in G2/M phase in response to

irradiation (Koniaras et al. 2001, Matsui et al.

2001). The G2/M arrest means the existence of

DNA damage repair prior to potential activation

of apoptosis and may be a crucial determinant of

radioresistance. Some studies implicated that

chemical compounds capable of abrogating G2/M

arrest and stimulating apoptosis are clinical avail-

able to override radioresistance (Yao et al. 1996,

Miyata et al. 2001a). According to these studies, we

combined the treatment of low-does STP with low

dose irradiations in the current study to overcome

the radioresistance and G2/M arrest induced by low

Figure 4. Caspases related mechanisms of apoptosis in U937 cells. (A) Western blot analyses of caspase- 3, -8, -9 cleavage forms in U937

cells treated with STP(10nM), IR (5 Gy) alone, or combined treatment for 18 h. Cleavage forms of caspases proteins represent its activation.

(B) The changes of PARP protein levels were detected by Western blotting. The level of total GAPDH protein was used as controls for equal

loading of protein in different lanes. (C) Apoptosis of U937 cells was assessed 18 h after combined treatment with STP (10 nM) and IR

(5 Gy), with or without preincubation with caspases inhibitors Z-VAD-FMK (40 mM), ZB4 (25 mM), Z-IETD-FMK (25 mM);

*p50.05 vs. combined treatment; #p50.05 vs. control.

102 H.-R. Guo et al.

Int J

Rad

iat B

iol D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y T

echn

isch

e U

nive

rsite

it E

indh

oven

on

11/2

3/14

For

pers

onal

use

onl

y.

Page 7: Staurosporine modulates radiosensitivity and radiation-induced apoptosis in U937 cells

does of irradiation in p53 deleted cancer cells. A

study, that irradiated U937 cells with 5 Gy IR and

observed a prominent G2/M arrest in 12 – 36 h when

apoptosis fraction was not obvious (Shinomiya et al.

2000). Our results indicated that the combined

treatment with 10 nM STP and 5 Gy IR for 18 h

could induce remarkable apoptosis when compared

to STP or IR treatment alone (Figure 1). Meanwhile,

G2/M arrest was overridden, and the G1 phase

proportion was increased (Figure 2). In addition,

pretreatment with STP followed by 5 Gy IR for 18 h

induced more apoptotic cells compared to STP or 5

Gy IR alone (Figure 1). We believe that STP can

override the G2/M arrest induced by low dose (5 Gy)

IR and thus lead to earlier apoptosis. Our data

implicated that the balance between the extent of

DNA damage and the duration of G2/M arrest might

determine whether irradiated cells would survive or

undergo apoptosis.

STP is a PKC inhibitor and also a strong inducer

of apoptosis at doses 25 – 100 times higher than the

current study, when applied as a single agent (Bossy-

Wetzel et al. 1998). It has been hypothesized that a

strategy could be developed that would permit the

exploitation of the G2 checkpoint to obtain a

therapeutic index in the treatment of cancers lacking

a G1 checkpoint which requires functional p53. The

lack of a G1 checkpoint is common in more than

50% of cancers containing p53 mutation (Levine

1997), which is a critical component for the

induction of apoptosis in response to DNA damage

(Lowe et al. 1993a, 1993b). Using this strategy,

normal cells would arrest in the G1 after DNA

damage from irradiation or chemotherapy, whereas

Figure 5. (A) Induction of mitochondrial dysfunction in U937 cells. Cells were treated with STP (10 nM), IR (5 Gy) alone or combined

treatment for 18 h. Then, cells were incubated with 3,30- dihexyloxacarbocynine and analyzed by flow cytometry. (B) Induction of

cytochrome C release in U937 cells treated with STP, IR alone and combined treatment for 18 h. Expressions of Bax, and Bcl-xL protein

levels were detected by Western blot using specific antibodies.

Staurosporine modulates radiosensitivity in U937 cells 103

Int J

Rad

iat B

iol D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y T

echn

isch

e U

nive

rsite

it E

indh

oven

on

11/2

3/14

For

pers

onal

use

onl

y.

Page 8: Staurosporine modulates radiosensitivity and radiation-induced apoptosis in U937 cells

cancer cells with a defective G1 checkpoint would

progress through the S-phase and into the G2 phase.

Therefore, abrogation of the G2 checkpoint would

be more detrimental to the cancer than the normal

cells. STP and its analogs have been reported to

increase the radiosensitivity of p53-deficient cancer

cells by the abrogation of the G2 checkpoint (Yao

et al. 1996, Busby et al. 2000, Kihara et al. 2000,

Wang et al. 2001). However, the mechanisms by

which the cell cycle or other key factors override G2/

M arrest in U937 cells with combined STP and IR

treatment have seldom been completely elucidated.

It has been reported that STP inhibits cell growth

at both G1 (low concentration) and G2/M (high

concentration) phases, and/or induces apoptosis

in human cancer cells (Bossy-Wetzel et al. 1998).

Figure 6. The activation of JNK in U937 cells. (A and B) Cells were treated with 5 Gy IR, 10 nM STP alone or combined treatment for

various time period. Expression of JNK phosphorylation was detected by Western blot using specific phospho- JNK antibody. The level of

total GAPDH protein was used as controls for equal loading of protein in different lanes. (C) Cell cycle progression and apoptosis after

treated with 5 Gy IR, 10 nM STP, 25 mM JNK inhibitor SP600125 alone or combined treatment with or without pretreatment of 25 mM

SP600125. Proportion of cell cycle phase and apoptosis were measured by flow cytometry for 18 h. The proportion of cell cycle phase and

apoptosis after different treatment was indicated in (D); #p5 0.05 vs. combined treatment.

104 H.-R. Guo et al.

Int J

Rad

iat B

iol D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y T

echn

isch

e U

nive

rsite

it E

indh

oven

on

11/2

3/14

For

pers

onal

use

onl

y.

Page 9: Staurosporine modulates radiosensitivity and radiation-induced apoptosis in U937 cells

A study on murine embryonic fibroblasts (MEF)

found that p53 and p16 functions were not essential

for STP induced G1 arrest, whereas Rb played an

important role in determining the degree of G1 arrest

observed in the first cell cycle following the exposure

to STP. In addition, MEF from mice lacking Rb

genes showed approximately a 70% reduction in the

capacity to arrest in the G1 phase following STP

treatment (Orr et al. 1998). Schnier et al. showed

that STP might promote the hypophosphorylation of

Rb leading to G1 arrest in bladder carcinoma cells

and found that levels of CDK inhibitor proteins p21

and p27 levels were increased. They concluded that

G1 arrest at the Rb dependent checkpoint may

prevent the activation of cyclin E/CDK2 through

stabilizing its interaction with inhibitor proteins p21

and p27 (Schnier et al. 1996). These studies

suggested that the G1 arrest induced by low dose

STP (less than 10 nM) may result from the

accumulation of Rb and p21, which is consistent

with our observation as shown in Figure 3B. We

suggested that Rb accumulation (Figure 3B) may also

play an important role in STP-triggered overriding of

G2/M arrest induced by 5 Gy IR. A more recent

report indicated that exposure of U937 cells ectopi-

cally-expressing Bcl-2 to the combination of 7-

hydroxystaurosporine (UCN-01) plus 2 Gy IR leads

to a reduction in cell proliferation, and that this

phenomenon appears to involve a non-apoptotic

mechanism (Cartee et al. 2002). Despite failing to

enhance apoptosis, UCN-01 treatment abrogated IR-

induced G2/M arrest, enhanced activation of CDK1,

promotion of G0/G1 arrest, and dephosphorylation

of Rb which is similar to our current study. The

expression of Bcl-2 has been shown to prevent the

induction of apoptosis by a variety of stimuli,

suggesting that Bcl-2 protein seems likely to function

as an antagonist of a central mechanism operative in

cell death (Hockenbery et al. 1993, Zhong et al. 1993,

Lin et al. 2004). These might contribute to the non-

apoptotic mechanism in Bcl-2 over-expressed U937

cells exposed to UCN-01 plus IR.

The CDK were recognized as key regulators of cell

cycle progression through their association with

regulatory subunits called cyclins (Pietenpol &

Stewart 2002, Maggiorella et al. 2003). Deregulation

of CDK activation or overexpression of cyclin D and

cyclin E has been frequently found in human cancers

(Pietenpol & Stewart 2002). p21 is a down stream

effector of p53 that mediates both G1 and G2/M

phase arrest (Harada & Ogden 2000, Ando et al.

2001). Nevertheless, there is sufficient evidence

showed that the up-regulation of p21 can be

independent of functional p53 protein (Ding et al.

2001, Sato et al. 2002). In the present study, we

found an obvious induction of p21 expression,

however, no significant change was found in the

expression of PCNA protein (Figure 3B). One study

has shown that the ovarian carcinoma cell line

SKOV-3, with a mutation of p53, is radioresistant

because of a loss of radiation-induced p21up-

regulation (Fan et al. 1998). CDKIs p27 and p21

regulate the G1/S transition of the cell cycle by

inhibiting cyclins D, E, and A. It also has been

reported that the human glioblastoma multiform

lines which were sensitive to ionizing radiation

showed a transient increase in the CDKIs p27 and

p21 within 24 h after exposure to radiation (Yao

et al. 2003). In our experiment, we observed a

marked induction of p21 and an inhibition of cyclin

A in cells with combined treatment. Meanwhile, the

G1 phase was increased. Cyclin E is essential for

progression through the G1 phase of the cell cycle

and initiates DNA replication by interacting with and

activating its catalytic partner, CDK2. The reduced

expression of p27Kip1 has been reported to correlate

with tumor progression and poor survival (Catzavelos

et al. 1997, Mori et al. 1997). Our observations imp-

licated that, when combined with 5 Gy IR, the induc-

tion of p21 by STP may result in the reduction in

radioresistance of U937 cells through overriding G2/

M arrest and enhancing early apoptosis (Figure 3B).

In addition, the kinase activity associated with Cyclin

B-cdc2 is critical for the G2/M transition, and the

activity of Cyclin B-cdc2 complex is regulated by the

positive regulator cdc25c and the two negative

regulators Wee1 and Myt1. The protein kinase

Wee1 phosphorylates cdc2 on Tyr-15, and the kinase

Myt1 phosphorylates cdc2 on Thr-14 and Tyr-15.

Both kinases are therefore capable of inactivating the

Cyclin B-cdc2 complex, leading to G2/M arrest

(O’Connell et al. 2000). Therefore, an increase in

Tyr-15 phosphorylated cdc2 represented one of the

definite markers indicating G2/M arrest. A significant

increase in the phosphorylation of cdc2 on Tyr-

15 was observed in cells treated with 5 Gy IR, which

resulted in decreased kinase activity and, therefore, a

G2/M arrest (Figure 3A). However, the phosphoryla-

tion of cdc2 on Tyr-15 was reduced through STP

combined with IR, providing the evidence of the

involvement of p21 in G1 phase arrest induced by

STP. Taken together, we suggest that STP combined

with IR enhances apoptosis and alters the cell cycle

progression through the induction of p21, the most

obvious change found in our study. Since the U937

cells used in the current study had a mutated, inactive

p53 gene, the increase in G1 by the induction of

p21 might be independent of the effect of p53 (Zaugg

et al. 2001).

Caspases are synthesized as pro-enzymes, which

are activated by the cleavage of their subunit.

Caspases-2, -8, -9, and caspase-10, termed apical

caspases, are earlier stimulated in the apoptotic

process and then activate their effector caspases

Staurosporine modulates radiosensitivity in U937 cells 105

Int J

Rad

iat B

iol D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y T

echn

isch

e U

nive

rsite

it E

indh

oven

on

11/2

3/14

For

pers

onal

use

onl

y.

Page 10: Staurosporine modulates radiosensitivity and radiation-induced apoptosis in U937 cells

(namely caspase-3, -6, and -7) (Hengartner 2000).

Among the apical caspases, caspase-9 is activated in

response to internal insults such as DNA damage,

whereas caspase-8 and caspase-10 are effectors of the

death receptor-mediated apoptotic signaling path-

way, which is initiated by Fas (Hengartner 2000). In

addition, interactions among the Bcl-2 family pro-

teins (Bax, Bak, Bcl-2, Bcl-xl, ect.) stimulate the

release of cytochrome c to induce apoptosis (Gross

et al. 1999, Huang & Strasser 2000). Overexpression

of Bcl-2 or Bcl-xL blocks the release of cytochrome c

from mitochondria and inhibits apoptosis (Kluck

et al. 1997, Verheij & Bartelink 2000). In U937 cells,

expression of caspase-3 precursor product appeared

to be higher than caspase-8 and caspase-9 precursor

products. Our results suggest that combined treat-

ment manipulates multiple cellular targets that

trigger different apoptotic cascades: One leads to

the release of mitochondrial cytochrome c and

activates caspase-9, while the other causes activation

of the membrane death receptors thus resulting in

the activation of caspase-8 (Figure 4). Similar results

have been observed in Jurket cells and normal T

lymphocytes, in which the activation of caspase-8 by

PUVA led to a consequent activation of caspase-9

and caspase-3 (Martelli et al. 2004).We suggest

combined treatment can result in the activation of

two apoptotic pathways, caspase-8 and caspase-9.

The levels of caspases (caspase -1, -2, -3, -6, -7, -8,

-9, and -10) expression in cancer cell lines and

neoplastic tissues were lower than those in the

control specimens. It has been demonstrated that

impaired expression of caspases is due to gene

deletion, mutation, and hypermethylation (Philchen-

kov et al. 2004). Mutation or hypermethylation of

caspase-8 gene has been reported to be associated

with no or low protein expression in some cancer cell

lines such as head and neck cancer cells and

neuroblastomas (Mandruzzato et al. 1997, Teitz

et al. 2001, van Noesel et al. 2003). A study by Teitz

et al. (2002) also implicated that gene deletion,

mutation, and methylation result in caspase-9

inactivation. They found caspase-9 was either de-

leted or translocated to another chromosome in

neuroblastoma cell lines with N-Myc gene amplifica-

tion. Although there is no sufficient evidence

showing deletion, mutation, or hypermethylation of

caspase-8 and -9 genes in U 937 cells, we found the

basal levels of procaspase-8 and -9 pretty low. STP

alone or in combination with IR could increase the

expressions and activities of procaspase-8 and -9.

STP enhanced procaspase-9 expression and activity

has been reported in neuroblastoma cell lines with

remaining caspase-9 allel(s) (Teitz et al. 2002),

which is similar to our results (Figure 4A). Further-

more, Watson et al. also indicated that STP was able

to decrease DNA methylation status in H4IIE Rat

hepatoma cells through unknown mechanism (Wat-

son et al. 2004). However, further investigation

leading to a comprehensive understanding of the

caspase activation induced by the combined treat-

ment is needed.

Mitochondria play a central role in both extrinsic

and intrinsic apoptotic pathways (Gil et al. 2003).

They play an important role in apoptosis by releasing

of cytochrome c from themselves into the cytoplasm,

leading to the activation of the caspase-cascade

system (Liu et al. 1996, Zhivotovsky et al. 1998).

Figure 5 shows that minor changes in DCm and a

slight release of cytochrome c could be observed in

cells with STP and IR alone, compared to the control.

However, combined treatment caused an obvious

change in the mitochondrial potential (DCm) and the

release of cytochrome c, when compared to the

control and STP or IR alone. The Bcl-2 family

includes both death antagonists such as Bcl-2 and

Bcl-xL, and death agonists such as Bax, Bak, and Bad

(Cory & Adams 2002). The expression levels of Bax

proteins were elevated in cells with combined

treatment when compared to STP or IR alone,

whereas the expression levels of Bcl-xL proteins

declined with combined treatment (Figure 5B).

These results suggest that combined treatment with

STP and IR caused a significant change in trans-

membrane potential and subsequently increased the

release of mitochondrial cytochrome c in U937 cells.

The mitogen-activated protein kinase (MAPK)

signal transduction pathways are conserved in

eukaryotic cells (Cano & Mahadevan 1995). The

JNK group is strongly activated by pro-inflammatory

cytokines (Ip & Davis 1998) or by extracellular stress

such as irradiation and heat shock (Enomoto et al.

2000, 2001). Previous studies have shown that

phosphorylation and activation of JNK was related

to the activation of caspase in X-ray and heat-

induced apoptosis (Enomoto et al. 2000, 2001). Our

results indicated that the prolonged activation of

JNK found in cells with combined treatment might

be involved in the changes in G2/M phase arrest

induced by 5Gy IR alone (Figure 6). Some studies

demonstrated that prolonged activation of MAPK

increased the expression of p21 protein. For exam-

ple, low-dose irradiation led to prolonged activation

of the MAPK cascade in lung cancer cells, and the

ability of radiation to increase p21 was dependent on

the MAPK cascade (Carter et al. 1998). It has been

suggested that the basal level of JNK plays an

important role during G2/M transit, and that JNK-

mediated apoptosis is cell cycle-dependent but p53-

independent (Du et al. 2004, Mingo-Sion et al.

2004). Thus, p21 may be an important target for

JNK cell cycle effects in mutant p53 cells. These

studies correlated well with our current results that

combined treatment of STP and IR could override

106 H.-R. Guo et al.

Int J

Rad

iat B

iol D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y T

echn

isch

e U

nive

rsite

it E

indh

oven

on

11/2

3/14

For

pers

onal

use

onl

y.

Page 11: Staurosporine modulates radiosensitivity and radiation-induced apoptosis in U937 cells

G2/M arrest via increasing the expression of p21

protein. However, the precise roles of p21 in

modulating this complex mechanism still need to

be further investigated.

Acknowledgements

This study was supported by the National Science

Council (NSC 93-2320-B-006-031 and NSC 93-2320-

B-006-049).

References

Abu-Qare AW, Abou-Donia MB. 2001. Biomarkers of apoptosis:

Release of cytochrome c, activation of caspase-3, induction of

8-hydroxy-20-deoxyguanosine, increased 3-nitrotyrosine, and

alteration of p53 gene. Journal of Toxicology and Environ-

mental Health B Critical Reviews 4:313 – 332.

Ando T, Kawabe T, Ohara H, Ducommun B, Itoh M, Okamoto

T. 2001. Involvement of the interaction between p21 and

proliferating cell nuclear antigen for the maintenance of G2/M

arrest after DNA damage. Journal of Biological Chemistry

276:42971 – 42977.

Bossy-Wetzel E, Newmeyer DD, Green DR. 1998. Mitochondrial

cytochrome c release in apoptosis occurs upstream of DEVD-

specific caspase activation and independently of mitochondrial

transmembrane depolarization. EMBO Journal 17:37 – 49.

Bracey TS, Williams AC, Paraskeva C. 1997. Inhibition of

radiation-induced G2 delay potentiates cell death by apoptosis

and/or the induction of giant cells in colorectal tumor cells with

disrupted p53 function. Clinical Cancer Research 3:1371 –

1381.

Busby EC, Leistritz DF, Abraham RT, Karnitz LM, Sarkaria JN.

2000. The radiosensitizing agent 7-hydroxystaurosporine

(UCN-01) inhibits the DNA damage checkpoint kinase

hChk1. Cancer Research 60:2108 – 2112.

Cano E, Mahadevan LC. 1995. Parallel signal processing among

mammalian MAPKs. Trends in Biochemical Sciences

20:117 – 122.

Cartee L, Sankala H, Davis C, Smith R, Maggio S, Lin PS, Dent

P, Grant S. 2002. 7-hydroxystaurosporine (UCN-01) and

ionizing radiation combine to inhibit the growth of Bcl-2-

overexpressing U937 leukemia cells through a non-apoptotic

mechanism. International Journal of Oncology 21:351 – 359.

Carter S, Auer KL, Reardon DB, Birrer M, Fisher PB, Valerie K,

Schmidt-Ullrich R, Mikkelsen R, Dent P. 1998. Inhibition of

the mitogen activated protein (MAP) kinase cascade potenti-

ates cell killing by low dose ionizing radiation in A431 human

squamous carcinoma cells. Oncogene 16:2787 – 2796.

Catzavelos C, Bhattacharya N, Ung YC, Wilson JA, Roncari L,

Sandhu C, Shaw P, Yeger H, Morava-Protzner I, Kapusta L,

Franssen E, Pritchard KI, Slingerland JM. 1997. Decreased

levels of the cell-cycle inhibitor p27Kip1 protein: prognostic

implications in primary breast cancer. Nature Medicine

3:227 – 230.

Cory S, Adams JM. 2002. The Bcl2 family: Regulators of the

cellular life-or-death switch. National Review of Cancer

2:647 – 656.

Davis RJ. 1994. MAPKs: New JNK expands the group. Trends in

Biochemical Sciences 19:470 – 473.

Ding Z, Parchment RE, LoRusso PM, Zhou JY, Li J, Lawrence

TS, Sun Y, Wu GS. 2001. The investigational new drug

XK469 induces G(2)-M cell cycle arrest by p53-dependent

and -independent pathways. Clinical Cancer Research

7:3336 – 3342.

Du L, Lyle CS, Obey TB, Gaarde WA, Muir JA, Bennett BL,

Chambers TC. 2004. Inhibition of cell proliferation and cell

cycle progression by specific inhibition of basal JNK activity:

Evidence that mitotic Bcl-2 phosphorylation is JNK-indepen-

dent. Journal of Biological Chemistry 279:11957 – 11966.

Enomoto A, Suzuki N, Hirano K, Matsumoto Y, Morita A, Sakai

K, Koyama H. 2000. Involvement of SAPK/JNK pathway in

X-ray-induced rapid cell death of human T-cell leukemia cell

line MOLT-4. Cancer Letters 155:137 – 144.

Enomoto A, Suzuki N, Liu C, Kang Y, Zhu J, Serizawa S,

Matsumoto Y, Morita A, Ito M, Hosoi Y. 2001. Involvement

of c-Jun NH2-terminal kinase-1 in heat-induced apoptotic cell

death of human monoblastic leukaemia U937 cells. Interna-

tional Journal of Radiation Biology 77:867 – 874.

Fan S, Twu NF, Wang JA, Yuan RQ, Andres J, Goldberg ID,

Rosen EM. 1998. Down-regulation of BRCA1 and BRCA2 in

human ovarian cancer cells exposed to adriamycin and

ultraviolet radiation. International Journal of Cancer 77:600 –

609.

Giaccia AJ, Kastan MB. 1998. The complexity of p53 modulation:

Emerging patterns from divergent signals. Genes & Develop-

ment 12:2973 – 2983.

Gil J, Almeida S, Oliveira CR, Rego AC. 2003. Cytosolic and

mitochondrial ROS in staurosporine-induced retinal cell

apoptosis. Free Radicals Biology and Medicine 35:1500 –

1514.

Gross A, McDonnell JM, Korsmeyer SJ. 1999. BCL-2 family

members and the mitochondria in apoptosis. Genes &

Development 13:1899 – 1911.

Harada K, Ogden GR. 2000. An overview of the cell cycle arrest

protein, p21(WAF1). Oral Oncology 36:3 – 7.

Heerdt BG, Houston MA, Mariadason JM, Augenlicht LH. 2000.

Dissociation of staurosporine-induced apoptosis from G2-M

arrest in SW620 human colonic carcinoma cells: Initiation of

the apoptotic cascade is associated with elevation of the

mitochondrial membrane potential (deltapsim). Cancer

Research 60:6704 – 6713.

Hengartner MO. 2000. The biochemistry of apoptosis. Nature

407:770 – 776.

Herskowitz I. 1995. MAP kinase pathways in yeast: For mating

and more. Cell 80:187 – 197.

Hockenbery DM, Oltvai ZN, Yin XM, Milliman CL, Korsmeyer

SJ. 1993. Bcl-2 functions in an antioxidant pathway to prevent

apoptosis. Cell 75:241 – 251.

Huang DC, Strasser A. 2000. BH3-Only proteins-essential

initiators of apoptotic cell death. Cell 103:839 – 842.

Ip YT, Davis RJ. 1998. Signal transduction by the c-Jun

N-terminal kinase (JNK) – from inflammation to development.

Current Opinion Cell Biology 10:205 – 219.

Johnson NL, Gardner AM, Diener KM, Lange-Carter CA, Gleavy

J, Jarpe MB, Minden A, Karin M, Zon LI, Johnson GL. 1996.

Signal transduction pathways regulated by mitogen-activated/

extracellular response kinase kinase kinase induce cell death.

Journal of Biological Chemistry 271:3229 – 3237.

Kihara C, Seki T, Furukawa Y, Yamana H, Kimura Y, van

Schaardenburgh P, Hirata K, Nakamura Y. 2000. Mutations in

zinc-binding domains of p53 as a prognostic marker of

esophageal-cancer patients. Japanese Journal of Cancer

Research 91:190 – 198.

Kinzler KW, Vogelstein B. 1996. Life (and death) in a malignant

tumour. Nature 379:19 – 20.

Kluck RM, Bossy-Wetzel E, Green DR, Newmeyer DD. 1997.

The release of cytochrome c from mitochondria: A primary site

for Bcl-2 regulation of apoptosis. Science 275:1132 – 1136.

Koniaras K, Cuddihy AR, Christopoulos H, Hogg A, O’Connell

MJ. 2001. Inhibition of Chk1-dependent G2 DNA damage

checkpoint radiosensitizes p53 mutant human cells. Oncogene

20:7453 – 7463.

Staurosporine modulates radiosensitivity in U937 cells 107

Int J

Rad

iat B

iol D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y T

echn

isch

e U

nive

rsite

it E

indh

oven

on

11/2

3/14

For

pers

onal

use

onl

y.

Page 12: Staurosporine modulates radiosensitivity and radiation-induced apoptosis in U937 cells

Levine AJ. 1997. p53, the cellular gatekeeper for growth and

division. Cell 88:323 – 331.

Lin YP, Zhu BZ, Yang MC, Frei B, Pan MH, Lin JK, Wang YJ.

2004. Bcl-2 overexpression inhibits tetrachlorohydroquinone-

induced apoptosis in NIH3T3 cells: A possible mechanism for

tumor promotion. Molecular Carcinogenesis 40:24 – 33.

Liu X, Kim CN, Yang J, Jemmerson R, Wang X. 1996. Induction

of apoptotic program in cell-free extracts: requirement for

dATP and cytochrome c. Cell 86:147 – 157.

Lowe SW, Bodis S, McClatchey A, Remington L, Ruley HE,

Fisher DE, Housman DE, Jacks T. 1994. p53 status and the

efficacy of cancer therapy in vivo. Science 266:807 – 810.

Lowe SW, Ruley HE, Jacks T, Housman DE. 1993a. p53-

dependent apoptosis modulates the cytotoxicity of anticancer

agents. Cell 74:957 – 967.

Lowe SW, Schmitt EM, Smith SW, Osborne BA, Jacks T. 1993b.

p53 is required for radiation-induced apoptosis in mouse

thymocytes. Nature 362:847 – 849.

Maggiorella L, Deutsch E, Frascogna V, Chavaudra N, Jeanson L,

Milliat F, Eschwege F, Bourhis J. 2003. Enhancement of

radiation response by roscovitine in human breast carcinoma

in vitro and in vivo. Cancer Research 63:2513 – 2517.

Maity A, McKenna WG, Muschel RJ. 1994. The molecular basis

for cell cycle delays following ionizing radiation: A review.

Radiotherapy and Oncology 31:1 – 13.

Mandruzzato S, Brasseur F, Andry G, Boon T, van der Bruggen

P. 1997. A CASP-8 mutation recognized by cytolytic T

lymphocytes on a human head and neck carcinoma. Journal

of Experimental Medicine 186:785 – 793.

Martelli AM, Cappellini A, Tazzari PL, Billi AM, Tassi C, Ricci F,

Fala F, Conte R. 2004. Caspase-9 is the upstream caspase

activated by 8-methoxypsoralen and ultraviolet-A radiation

treatment of Jurkat T leukemia cells and normal T lympho-

cytes. Haematologica 89:471 – 479.

Matsui Y, Tsuchida Y, Keng PC. 2001. Effects of p53 mutations

on cellular sensitivity to ionizing radiation. American Journal of

Clinical Oncology 24:486 – 490.

Mingo-Sion AM, Marietta PM, Koller E, Wolf DM, Van Den

Berg CL. 2004. Inhibition of JNK reduces G2/M transit

independent of p53, leading to endoreduplication, decreased

proliferation, and apoptosis in breast cancer cells. Oncogene

23:596 – 604.

Miyata H, Doki Y, Yamamoto H, Kishi K, Takemoto H, Fujiwara

Y, Yasuda T, Yano M, Inoue M, Shiozaki H, Weinstein IB,

Monden M. 2001a. Overexpression of CDC25B overrides

radiation-induced G2-M arrest and results in increased apoptosis

in esophageal cancer cells. Cancer Research 61:3188 – 3193.

Miyata H, Doki Y, Yamamoto H, Kishi K, Takemoto H, Fujiwara

Y, Yasuda T, Yano M, Inoue M, Shiozaki H, Weinstein IB,

Monden M. 2001b. Overexpression of CDC25B overrides

radiation-induced G2-M arrest and results in increased

apoptosis in esophageal cancer cells. Cancer Research

61:3188 – 3193.

Mori M, Mimori K, Shiraishi T, Tanaka S, Ueo H, Sugimachi K,

Akiyoshi T. 1997. p27 expression and gastric carcinoma.

Natural Medicine 3:593.

O’Connell MJ, Walworth NC, Carr AM. 2000. The G2-phase

DNA-damage checkpoint. Trends in Cell Biology 10:296 –

303.

Omura S, Iwai Y, Hirano A, Nakagawa A, Awaya J, Tsuchya H,

Takahashi Y, Masuma R. 1977. A new alkaloid AM-2282 OF

Streptomyces origin. Taxonomy, fermentation, isolation and

preliminary characterization. Journal of Antibiotics (Tokyo)

30:275 – 282.

Orr MS, Reinhold W, Yu L, Schreiber-Agus N, O’Connor PM.

1998. An important role for the retinoblastoma protein in

staurosporine-induced G1 arrest in murine embryonic fibro-

blasts. Journal of Biological Chemistry 273:3803 – 3807.

Pawlik TM, Keyomarsi K. 2004. Role of cell cycle in mediating

sensitivity to radiotherapy. International Journal of Radiation,

Oncology, Biology and Physics 59:928 – 942.

Philchenkov A, Zavelevich M, Kroczak TJ, Los M. 2004. Caspases

and cancer: mechanisms of inactivation and new treatment

modalities. Experimental Oncology 26:82 – 97.

Pietenpol JA, Stewart ZA. 2002. Cell cycle checkpoint signaling:

Cell cycle arrest versus apoptosis. Toxicology 181 – 182, 475 –

481.

Playle LC, Hicks DJ, Qualtrough D, Paraskeva C. 2002.

Abrogation of the radiation-induced G2 checkpoint by the

staurosporine derivative UCN-01 is associated with radio-

sensitisation in a subset of colorectal tumour cell lines. British

Journal of Cancer 87:352 – 358.

Powell SN, DeFrank JS, Connell P, Eogan M, Preffer F,

Dombkowski D, Tang W, Friend S. 1995. Differential

sensitivity of p53(7) and p53(þ) cells to caffeine-induced

radiosensitization and override of G2 delay. Cancer Research

55:1643 – 1648.

Rocha S, Soengas MS, Lowe SW, Glanzmann C, Fabbro D,

Winterhalter K, Bodis S, Pruschy M. 2000. Protein kinase C

inhibitor and irradiation-induced apoptosis: relevance of the

cytochrome c-mediated caspase-9 death pathway. Cell Growth

& Differentiation 11:491 – 499.

Sato T, Koseki T, Yamato K, Saiki K, Konishi K, Yoshikawa M,

Ishikawa I, Nishihara T. 2002. p53-independent expression of

p21(CIP1/WAF1) in plasmacytic cells during G(2) cell cycle

arrest induced by Actinobacillus actinomycetemcomitans cyto-

lethal distending toxin. Infection and Immunity 70:528 – 534.

Schnier JB, Nishi K, Goodrich DW, Bradbury EM. 1996. G1

arrest and down-regulation of cyclin E/cyclin-dependent kinase

2 by the protein kinase inhibitor staurosporine are dependent

on the retinoblastoma protein in the bladder carcinoma cell

line 5637. Proceedings of the National Academy of Sciences of

the United States of America 93:5941 – 5946.

Shimizu S, Narita M, Tsujimoto Y. 1999. Bcl-2 family proteins

regulate the release of apoptogenic cytochrome c by the

mitochondrial channel VDAC. Nature 399:483 – 487.

Shinomiya N, Kuno Y, Yamamoto F, Fukasawa M, Okumura A,

Uefuji M, Rokutanda M. 2000. Different mechanisms between

premitotic apoptosis and postmitotic apoptosis in X-irradiated

U937 cells. International Journal of Radiation, Oncology,

Biology and Physics 47:767 – 777.

Teitz T, Lahti JM, Kidd VJ. 2001. Aggressive childhood

neuroblastomas do not express caspase-8: an important

component of programmed cell death. Journal of Molecular

Medicine 79:428 – 436.

Teitz T, Wei T, Liu D, Valentine V, Valentine M, Grenet J, Lahti

JM, Kidd VJ. 2002. Caspase-9 and Apaf-1 are expressed and

functionally active in human neuroblastoma tumor cell lines

with 1p36 LOH and amplified MYCN. Oncogene 21:1848 –

1858.

van Noesel MM, van Bezouw S, Voute PA, Herman JG, Pieters R,

Versteeg R. 2003. Clustering of hypermethylated genes in

neuroblastoma. Genes, Chromosomes & Cancer 38:226 – 233.

Verheij M, Bartelink H. 2000. Radiation-induced apoptosis. Cell

and Tissue Research 301:133 – 142.

Wang Y, Li J, Booher RN, Kraker A, Lawrence T, Leopold WR,

Sun Y. 2001. Radiosensitization of p53 mutant cells by

PD0166285, a novel G(2) checkpoint abrogator. Cancer

Research 61:8211 – 8217.

Waskiewicz AJ, Cooper JA. 1995. Mitogen and stress response

pathways: MAP kinase cascades and phosphatase regulation in

mammals and yeast. Current Opinion in Cell Biology 7:798 –

805.

Watson RE, McKim JM, Cockerell GL, Goodman JI. 2004. The

value of DNA methylation analysis in basic, initial toxicity

assessments. Toxicological Sciences 79:178 – 188.

108 H.-R. Guo et al.

Int J

Rad

iat B

iol D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y T

echn

isch

e U

nive

rsite

it E

indh

oven

on

11/2

3/14

For

pers

onal

use

onl

y.

Page 13: Staurosporine modulates radiosensitivity and radiation-induced apoptosis in U937 cells

Yao KC, Komata T, Kondo Y, Kanzawa T, Kondo S, Germano

IM. 2003. Molecular response of human glioblastoma multi-

forme cells to ionizing radiation: cell cycle arrest, modulation

of the expression of cyclin-dependent kinase inhibitors, and

autophagy. Journal of Neurosurgery 98:378 – 384.

Yao SL, Akhtar AJ, McKenna KA, Bedi GC, Sidransky D, Mabry

M, Ravi R, Collector MI, Jones RJ, Sharkis SJ, Fuchs EJ, Bedi

A. 1996. Selective radiosensitization of p53-deficient cells by

caffeine-mediated activation of p34cdc2 kinase. Nature Med-

icine 2:1140 – 1143.

Zaugg K, Rocha S, Resch H, Hegyi I, Oehler C, Glanzmann C,

Fabbro D, Bodis S, Pruschy M. 2001. Differential p53-

dependent mechanism of radiosensitization in vitro and in vivo

by the protein kinase C-specific inhibitor PKC412. Cancer

Resarch 61:732 – 738.

Zhivotovsky B, Orrenius S, Brustugun OT, Doskeland SO. 1998.

Injected cytochrome c induces apoptosis. Nature 391:449 –

450.

Zhong LT, Sarafian T, Kane DJ, Charles AC, Mah SP, Edwards

RH, Bredesen DE. 1993. bcl-2 inhibits death of central neural

cells induced by multiple agents. Proceedings of the National

Academy of Sciences of the United States of America

90:4533 – 4537.

Staurosporine modulates radiosensitivity in U937 cells 109

Int J

Rad

iat B

iol D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y T

echn

isch

e U

nive

rsite

it E

indh

oven

on

11/2

3/14

For

pers

onal

use

onl

y.

Page 14: Staurosporine modulates radiosensitivity and radiation-induced apoptosis in U937 cells

Int J

Rad

iat B

iol D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y T

echn

isch

e U

nive

rsite

it E

indh

oven

on

11/2

3/14

For

pers

onal

use

onl

y.