university of groningen mechanisms of drug-induced ... · chapter 2 tetrahydroxyquinone induces...

25
University of Groningen Mechanisms of drug-induced apoptosis in human leukemia Cavagis, Alexandre Donizeti Martins IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below. Document Version Publisher's PDF, also known as Version of record Publication date: 2006 Link to publication in University of Groningen/UMCG research database Citation for published version (APA): Cavagis, A. D. M. (2006). Mechanisms of drug-induced apoptosis in human leukemia. s.n. Copyright Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons). Take-down policy If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim. Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum. Download date: 18-03-2020

Upload: others

Post on 15-Mar-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

University of Groningen

Mechanisms of drug-induced apoptosis in human leukemiaCavagis, Alexandre Donizeti Martins

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite fromit. Please check the document version below.

Document VersionPublisher's PDF, also known as Version of record

Publication date:2006

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):Cavagis, A. D. M. (2006). Mechanisms of drug-induced apoptosis in human leukemia. s.n.

CopyrightOther than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of theauthor(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policyIf you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediatelyand investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons thenumber of authors shown on this cover page is limited to 10 maximum.

Download date: 18-03-2020

Chapter 2

Tetrahydroxyquinone induces apoptosis of

leukemia cells through diminished survival

signaling

Alexandre D. Martins Cavagis1,2, Carmen Veríssima Ferreira1,2,

Henri H. Versteeg2, Cristiane Fernandes Assis1, Carina L. Bos2, Sylvia A.

Bleuming2, Sander H. Diks3, Hiroshi Aoyama1,

Maikel P. Peppelenbosch3,4

1 Departamento de Bioquímica, Instituto de Biologia, Universidade Estadual de

Campinas (UNICAMP), Campinas, São Paulo, Brazil. 2 Department of Experimental Internal Medicine, Academic Medical Center (AMC)

University of Amsterdam, The Netherlands. 3Department of Cell Biology, University of Groningen, Antonius Deusinglaan 1

NL-9713 AV Groningen, The Netherlands

4corresponding author (M.P.Peppelenbosch)

Tel: 31-50-363 2522 Fax: 31-50-363 2512

e-mail address: [email protected]

Experimental Hemato logy (2006) 34 (2) 188-196.

Chapter 2

28

ABSTRACT Objective Tetrahydroxyquinone is a molecule best known as a primitive anti-cataract drug, but is

also a highly redox active molecule which can take part in a redox cycle with

semiquinone radicals, leading to the formation of reactive oxygen species (ROS). Its

potential as an anticancer drug has not been investigated.

Methods The effects of tetrahydroxyquinone on HL60 leukaemia cells are investigated using

FACS-dependent detection of phosphatidylserine exposure combined with 7-amino-

actinomycin D (7-AAD) exclusion, via Western blotting using phosphospecific

antibodies, and by transfection of constitutively active protein kinase B (PKB).

Results We observe that in HL60 leukaemia cells tetrahydroxyquinone causes ROS production

followed by apoptosis through the mitochondrial pathway, whereas cellular physiology

of normal human blood leukocytes was not affected by tetrahydroxyquinone. The anti-

leukaemic effect of tetrahydroxyquinone is accompanied by reduced activity of various

anti-apoptotic survival molecules including the protein kinase B pathway. Importantly,

transfection of protein kinase B into HL60 cells and thus artificially increasing protein

kinase B activity inhibits tetrahydroxyquinone-dependent cytotoxicity.

Conclusion

Tetrahydroxyquinone provokes cytotoxic effects on leukaemia cells by reduced protein

kinase B-dependent survival signalling followed by apoptosis through the

mitochondrial pathway. Thus, tetrahydroxyquinone may be representative of a novel

class of chemotherapeutic drugs, inducing apoptosis in cancer cells through diminished

survival signalling possibly as a consequence of ROS-generation.

Keywords: HL60 cells; tetrahydroxyquinone; protein phosphatases; MAPKs; oxidative stress.

THQ induces apoptosis of leukemia cells through diminished survival signaling

29

INTRODUCTION

Chemotherapy for the treatment of some types of neoplastic disease has been

one of the success stories of medicine. However, the chemotherapeutic treatment

outcome of most adult acute myeloid leukemia (AML) remains unacceptable [1].

Among AMLs, acute promyelocytic leukemia can be successfully treated with all-trans-

retinoic acid (ATRA). The development, however, of resistance to a wide spectrum of

cytotoxic drugs frequently impedes the successful treatment of AML either in the

beginning of disease or following primary or subsequent relapses. Moreover, ATRA

resistance in acute promyelocytic leukemia is rare but markedly increases in frequency

after relapses from chemotherapy-induced clinical remission [2, 3]. Hence, novel

avenues for the treatment of AML are required.

It is now generally recognised that the reactive oxygen species (ROS) play an

important role as regulatory mediators in signalling processes [4]. Accordingly, it has

now been shown that a multitude of physiological processes are under the direct control

of ROS, the most important being the regulation of vascular tone, the sensing of oxygen

tension, the enhancement of leukocyte signal transduction and the induction of

apoptosis, the latter as an essential component of the tumour necrosis factor α-

dependent signal transduction [5-9]. Therefore, ROS generation is an important element

in the control of cellular biochemical processes. In addition, ROS generation is

important for inducing cytotoxicity in cancer cells, but the molecular mechanism

underlying these cytotoxic effects remains unclear, hampering the development of more

effective drugs.

ROS formation depends on the univalent reduction of triplet-state molecular

oxygen [10]. This process is mediated by enzymes such as NAD(P)H oxidases and

xanthine oxidase or nonenzymatic by redox-reactive compounds such as

tetrahydroxyquinone [11-13]. Superoxide dismutases convert superoxide enzymatically

into hydrogen peroxide or non-enzymatically into non-radical species, hydrogen

peroxide and singlet oxygen. Hydrogen peroxide may be converted into water by the

enzymes catalase or glutathione peroxidase. This latter enzyme oxidizes glutathione to

glutathione disulfide, which can be converted back to glutathione by glutathione

reductase in an NADPH-consuming process [14, 15]. Thus, the biochemistry of ROS

generation is relatively well understood.

Chapter 2

30

The biological effects of ROS production, however, are less clear. At high

concentrations ROS are dangerous for living organisms, damaging virtually all cellular

constituents [10]. Nevertheless, at moderate concentrations, ROS play important roles in

the control of various cellular functions and cellular generation of ROS is actively

induced under various conditions [4, 10]. The archetypal example is NADPH oxidase

activation upon immune stimulation of the phagocytic cells of the myeloid lineage,

resulting ROS production which, apart from its bactericidal function, is also

instrumental for the induction of pro-inflammatory gene transcription. Hence,

phagocyte ROS production is pivotal for proper function of the innate immune system

[16-18].

Various studies have shown that ROS producing drugs can exert important

cytotoxic effects in leukaemia cells, although the molecular details by which such drugs

mediate cancer cell death remain obscure [15]. This consideration prompted us to test

the effect of as a cytotoxic agent for HL60 leukaemia cells. Tetrahydroxyquinone is a

compound best known as a primitive anti-cataract drug, but expected to act as a redox

active benzoquinone [13]. In the present study we observe that tetrahydroxyquinone

indeed efficiently induces ROS generation in turn responsible for HL60 cell apoptosis

through the mitochondrial pathway. This apoptosis is accompanied by reduced activity

of the anti-apoptotic PKB [14] and nuclear factor (NF)-κB pathways [15] while

concomitantly specific activation of Jun-N-terminal kinase and protein phosphatases

(PPs) is observed [16]. Importantly, forced expression of PKB counteracts the effect of

tetrahydroxyquinone on HL60 cell apoptosis. Thus the diminished survival signalling is

essential for tetrahydroxyquinone-dependent cytotoxity and tetrahydroxyquinone may

be representative of a novel class of chemotherapeutic drugs, inducing apoptosis in

cancer cells through diminished survival signalling. The tetrahydroxyquinone pathways

mediating this effect involve at least in part ROS-generation.

THQ induces apoptosis of leukemia cells through diminished survival signaling

31

MATERIALS AND METHODS

Cell line and reagents

HL60 cells were purchased from the American Type Culture Collection

(ATCC, Rock-ville, MD). Polyclonal antibodies anti-caspase 3, anti-BAD, anti-

phospho-BAD, anti-phospho-p38 MAPK, anti-phospho-IκB, anti-IκB, anti-phospho-

PKC delta, anti-phospho-PKB, anti-phospho-Raf, anti-phospho-p42/44, anti-phospho-

JNK, anti-phospho-CREB, anti-rabbit and anti-mouse peroxidase-conjugated anti-

bodies were purchased from Cell Signaling Tech-nology (Beverly, MA). The antibodies

against phos-pho-PP2A, phosphotyrosine, phospho-threonine and NF-κB (p50 and p65)

were purchased from Santa Cruz Biotechnology (Santa Cruz, CA). Tetrahydroxy -

quinone was purchased from Sigma Chemical Com-pany.

Leukocyte Culture

Human blood was collected from healthy donors and human peripheral blood

mononuclear cells were isolated by Ficoll/Hypaque gradient centrifugation. Leukocytes

were cultured at the same conditions described for HL60 cells, the only difference was

the addition of 5 µg/ml phytohemaglutinin in each well. Cells were plated at density of 1

x 106 plating/ml in 24-well plate. The medium was removed 48h after cell seeding and

replaced with medium containing tetrahydroxyquinone.

Cell Culture and viability assays

HL60 cells were routinely grown in suspension in RPMI 1640 medium

supplemented with 2 mM glutamine, 100 IU/ml penicillin, 100 µg/ml streptomycin and

10% heat-inactivated fetal bovine serum, at 37oC in a 5% CO2 humidified atmosphere.

In all experiments, 3 x 105 cells/ml were seeded and, after 72 h, treated with different

tetra-hydroxyquinone concentrations for the specified periods of time.

Cell viability was assessed based on trypan blue dye exclusion and three

additional parameters: MTT reduction, protein phosphatase activity and determination

of the total protein amount.

Chapter 2

32

MTT reduction assay

The medium containing tetrahydroxyqui-none was removed and 1.0 ml of

MTT solution (0.5 mg MTT/ml of culture medium) was added to each well. After

incubation for 4 h at 37oC, the medium was removed and the formazan released by

solubilisation in 1.0 ml of ethanol. The plate was shaken for 5 min on a plate shaker and

the absorbance was measured at 570 nm (15, 16).

Protein phosphatase assay

The phosphatase extract was obtained by lysing the cells with acetate buffer

0.1 mM (pH 5.5). Then, the enzyme activity was measured in a reaction medium (final

volume, 1.0 ml) containing 100 mM acetate buffer (pH 5.5), 5.0 mM pNPP and cell

extract enzyme. After a 30 min incubation at 37°C, the reaction was stopped by adding

1.0 ml of 1.0 M NaOH. The amount of pNP released was measured at 405 nm (17).

Protein quantification

Protein concentrations were determined by a modification of the Lowry

method as described by Hartree (18).

Reduced glutathione determination

The concentration of reduced glutathione in HL60 cells was determined after

treatment of the cells for 24 h. The cells (3×105/ml) were washed with physiological

solution and lysed with water; 3 ml of precipitant solution (1.67 g glacial

metaphosphoric acid, 0.2 g ethylenediaminetetraacetic acid (EDTA) and 30 g NaCl in

100 ml MilliQ water) was added to the lysate (2 ml). After 5 min, this mixture was

centrifuged and 0.4 ml of the supernatant was added to 1.6 ml of reaction medium

(0.2 M Na2HPO4 buffer, pH 8.0; 0.5 mM DTNB dissolved in 1% sodium citrate).

Subsequently, the absorbance of the product (NTB) was measured at 412 nm and

reduced glutathione concentration calculated using the extinction coefficient E =

13.6 mol-1 cm-1 (19).

THQ induces apoptosis of leukemia cells through diminished survival signaling

33

Spectrofluorometric determination of ROS

HL60 cells were treated with 100 µM tetra-hydroxyquinone or the

combinations tetrahydroxy -quinone/10 mM glutathione and tetrahydroxyqui-none/15

mM N-acetyl-L-cysteine for 24 h in RPMI 1640 (without phenol red) containing 5%

serum. Afterwards, the cells (2×106/10 ml) were washed with PBS and resuspended in

RPMI 1640 (without phenol red, with 5% FBS) containing 20 µM DCFH-DA

(dichlorofluorescein diacetate). After 30 min of incubation at 37°C, the cells were

washed three times and resuspended in RPMI 1640 (without phenol red, with 5% FBS).

The fluorescence of the suspension was measured using a RF-5300 PC Shimadzu

spectrofluorometer with excitation at 485 nm and emission at 530 nm.

DNA fragmentation analysis

The cell pellets (5x106) were lysed in 0.5 ml of lysis buffer containing 5 mM

Tris -HCl, 20 mM ethilenediaminetetraacetic acid (EDTA) and 0.5% Triton X 100. After

centrifugation at 1,500 x g for 10 min, the pellets were resuspended in 250 µL of lysis

buffer and, to the supernatants (S), 20 µL of 6 M perchloric acid was added. Then, 500

µL of 10% trichloroacetic acid (TCA) were added to the pellets (P), the samples were

centrifuged for 10 min at 5,000 rpm and the pellets were resuspended in 250 µL of 5%

TCA followed by incubation at 100oC for 15 min. Subsequently, to each sample, 500

µL of solution (15 mg/ml DPA in glacial acetic acid), 15 µL/ml of sulfuric acid and 15

µg/ml acetaldehyde were added and incubated at 37 oC for 18 h (20). The proportion of

fragmented DNA was calculated from the absorbance at 594 nm using the following

formula:

Fragmented DNA (%) = 100 x (amount of the fragmented DNA in the

supernatant) / (amount of the fragmented DNA in the supernatant + amount DNA in the

pellets).

Western blotting

Cells (3 x 107) were lysed in 200 µL of lysis buffer [50 mM Tris –HCl (pH

7.4), 1% Tween 20, 0.25% sodium deoxycholate, 150 mM NaCl, 1 mM EGTA, 1 mM

Na3 VO4, 1 mM NaF and protease inhibitors (1 µg/ml aprotinin and 1 µg/ml 4-(2-

aminoethyl) benzenesulfonyl fluoride)] for 2 h on ice. Protein extracts were cleared by

Chapter 2

34

centrifugation and protein concentrations were determined using a Lowry protein assay.

An equal volume of 2X sodium dodecyl sulfate (SDS) gel loading buffer [100 mM

Tris–HCl (pH 6.8), 200 mM DTT, 4% SDS, 0.1% bromophenol blue and 20% glycerol]

was added to the samples, which were subsequently boiled for 10 min. Afterwards, cell

extracts were resolved by SDS-polyacry lamide gel (12%) electrophoresis (PAGE) and

transferred to PVDF membranes. Membranes were blocked in 1% fat-free dried milk or

bovine serum albumin (2%) in Tris -buffered saline (TBS)-Tween 20 (0.05%) and

incubated overnight at 4°C with appropriate primary antibody at 1:1000 dilution. After

washing in TBS-Tween 20 (0.05%), mem-branes were incubated with anti-rabbit or

anti-mouse horseradish peroxidase conjugated secondary anti-bodies, at 1:2000

dilutions, in blocking buffer for 1h. Detection was performed using enhanced

chemiluminescence (ECL).

Mitochondrial extract preparation (cytochrome c release)

For cytochrome c analysis, the cells were washed with ice cold PBS and

resuspended in lysis buffer containing 220 mM mannitol, 68 mM sucrose, 50 mM

Hepes–NaOH, pH 7.4, 50 mM KCl, 5 mM EGTA, 2 mM MgCl2, 1 mM DTT, and

protease inhibitors (1 µg/ml aprotinin, 10 µg/ml leupeptin and 1mM 4-(2-amino-ethyl)-

benzolsulfonyl-fluoride-hy-drochloride). After incubation on ice for 1 h, the lysate was

centrifuged at 14,000 x g for 15 min. Then, the supernatant was resolved by SDS-

PAGE.

Annexin V and 7-amino-actinomycin D assays

Control and tetrahydroxyquinone-treated cells were collected and resuspended

in 1X binding buffer (0.01 M Hepes/NaOH, pH 7.4, 0.14 mM NaCl and 2.5 mM CaCl2)

at a concentration of 1 x 106 cells/ml. Subsequently, 100 µl of cell suspension were

transfered to a 5 ml tube and Annexin V FITC (5 µl) and 7-amino-actinomycin D (7-

AAD) - (10 µl) were added. The cells were incubated at room temperature for 15 min,

after which 400 µl of 1X binding buffer was added and apoptosis then analyzed by flow

cytometry.

THQ induces apoptosis of leukemia cells through diminished survival signaling

35

Transient transfection of HL60 cells

HL60 cells (4 x 106) were transfected with 0.4 µg of a plasmid expressing

GAG-PKB, a constitutive form of PKB. Cells were cotransfected with 0.1 µg of a pUT-

galactosidase to normalize for transfection efficiency. After transfection, the cells were

cultured for 24h, harvested, lysed in commer-cially available reporter lysis buffer

(Promega) and β-galactosidase activity was determined using chloro-phenol red-β-D-

galactopyranoside (Roche) as subs-trate.

Transfected cells were treated with tetrahydroxyquinone for 24 h and the cell

viability was assessed by MTT reduction. The expression of PKB was evaluated by

western blotting.

Statistical evaluation

All experiments were performed in triplicate and the results shown in the

graphs represent the mean and standard deviation. Cell viability data were expressed as

the means ± standard errors of three independent experiments carried out in triplicates.

Data from each assay were analyzed statistically by ANOVA followed by a Dunnett´s

test. Multiple comparisons among group mean differences were checked with Tukey

post hoc test.

Differences were considered significant when the p value was less than 0.05.

Western blottings represent three independent experiments.

RESULTS

Tetrahydroxyquinone is cytotoxic for HL60 leukaemia cells

As evidence has been presented that the redox state-altering agents are potent

cytotoxic agents in leukaemia cells, although acting through as yet unknown molecular

mechanisms [23, 24], we decided to test the possible cytotoxic effects of

tetrahydroxyquinone on HL60 cells. To this end, HL60 were treated with various

concentrations of tetrahydroxyquinone for 24 hours and cell viability was determined

using total protein content, cellular phosphatase activity, or mitochondrial function

(MTT reduction, Fig. 1A) as a measure. It appeared that using either measure,

tetrahydroxyquinone was highly cytotoxic to the leukaemia cells, the apparent IC50 of

Chapter 2

36

0 100 200 300 400 5000

20

40

60

80

100

120

140

160

Protein MTT phosphatase

Cel

l via

bilit

y (%

)0 100 200 300 400 500

0

20

40

60

80

100

120

140

160

Protein MTT phosphatase

Cel

l via

bilit

y (%

)

Human leukaemia cells Human peripheral bloodmononuclear cells

C

WithROSscavenger

WithoutROSscavenger

tetrahydroxyquinone-induced cytotoxicity being similar whether assessed by total

protein content (IC50 20 µM), phosphatase activity (IC50 40 µM), or by MTT assay

(IC50 45 µM).

Figure 1. Effect of tetrahydroxyquinone on HL60 cell viability versus effects on

untransformed cells. HL60 cell viability was evaluated using three different parameters (protein

content, MTT reduction and phosphatase activity) after treatment with tetrahydroxyquinone for

24 h, in the absence (A) or presence of the ROS scavenger gluthathion (10 mM; B). The lack of

effect of tetrahydroxyquinone on normal peripheral blood mononuclear cell leukocytes is also

depicted (C). Each point represents the mean ± standard deviation of three independent

experiments.

THQ induces apoptosis of leukemia cells through diminished survival signaling

37

Importantly, when healthy human leukocytes were exposed to tetrahydroxy -

quinone no apparent toxicity was apparent, even at concentrations 10 times as high as

the IC50 for leukaemia cells, when assayed by protein content, phosphatase activity, or

MTT (fig 1C). Thus tetrahydroxyquinone is specifically cytotoxic for HL60 leukaemia

cells but not for the corresponding untransformed counterparts.

Tetrahydroxyquinone-dependent ROS generation mediates cytotoxity

Tetrahydroxyquinone is a highly redox active molecule, expected to induce the

formation of ROS by taking part in a redox cycle with semiquinone radicals. We

decided to investigate whether the tetrahydroxyquinone-mediated cytotoxic effects are

mediated by ROS generation. In agreement with a role of ROS generation in

tetrahydroxyquinone cytotoxicity, we observed that the compound substantially

increases the cellular levels of ROS, as determined by dichlorofluorescein diacetate-

dependent spectrophotometry (Fig. 2). Importantly, treatment with reduced glutathione

or N-acetyl-L-cysteine abolished the capacity of HL60 cells to react to

tetrahydroxyquinone with ROS production (Fig. 2). This allowed us test the importance

of tetrahydroxyquinone-induced ROS formation for its cytotoxic effects, and a

significant rightward shift of the dose-response curve with respect to

tetrahydroxyquinone-induced cytotoxicity was observed in the presence of ROS

generation inhibitors whether assessed by total protein content (IC50 from 20 µM to 45

µM ), phosphatase activity (IC50 from 40 µM to 140 µM), or by MTT assay (IC50 from

45 µM to 140 µM) (Fig. 1A,B). Thus tetrahydroxyquinone is an efficient inducer of

ROS production in HL60 leukaemia cells and ROS generation is essential for the

cytotoxic effect of this compound.

Chapter 2

38

RO

S p

rod

uct

ion

Figure 2. Tetrahydroxyquinone causes production of reactive oxygen species. The

production of reactive oxygen metabolites was determined using dichlorofluorescein diacetate-

loaded cells and a spectrofluorometer (excitation at 485 nm and emission at 530 nm). 1 - Non-

treated HL60 cells (control); 2 – tetrahydroxyquinone (100 µM) - treated cells; 3 - HL60 cells

pre- treated with N-acetyl-L-cysteine (10 mM) and 4 - HL60 cells pre-treated with reduced

glutathione (10 mM) for 30 min. For conditions 3 and 4, the cultures were further incubated with

100 µM tetrahydroxyquinone for 24h. Afterwards, a 30 min incubation with DCFH-D was carried

out and the fluorescence intensity was recorded by spectrofluorometry. Results were expressed as

the relative fluorescence intensity with respect to untreated cells. 5 - As a positive control - H2O2-

treated cells were used to monitor the level of ROS. Bars represent mean ± standard deviation.

Tetrahydroxyquinone induces cell death by apoptosis

Generally speaking cell death is brought about either via necrosis or via

apoptosis. The former process is associated with relatively large damage to the

surrounding tissue. The latter is associated with controlled elimination of cancer cells.

Thus for the possible treatment of leukaemia cytotoxic compounds should preferentially

act via apoptosis. To address the question whether tetrahydroxyquinone induces cell

death via apoptosis we measured three cellular processes associated with apoptosis

rather as necrosis: caspase 3 activation, DNA fragmentation, and phosphatidylserine

exposure.

THQ induces apoptosis of leukemia cells through diminished survival signaling

39

Figure 3. Apoptosis induction by tetrahydroxyquinone. (A) Cells were treated with

tetrahydroxyquinone as indicated and lysates were resolved by immunoblotting as described in

methods to assess the level of active caspase 3. (B) The samples were prepared as described by

Zhu and collaborators (23) and the proportion of fragmented DNA was calculated from

absorbance at 594 nm using the approach described in the method section. The bars represent the

mean ± standard deviation of three experiments. (C) Cell samples were prepared as described in

Methods and Annexin V-positive, 7-AAD-positive and Annexin V/7-AAD-positive populations

were analyzed by flow cytometry.

It appeared that tetrahydroxyquinone efficiently activates caspase 3 (Fig. 3A)

in concentration in excess of 25 µM, stimulates DNA fragmentation at the same

concentration (Fig. 3B) and provoke phosphatidylserine exposure (Fig. 3C) when

applied in a concentration of 25 µM or more. Importantly, induction of apoptosis as

measured by DNA fragmentation was not detected when the formation of ROS was

blocked (Fig 3B). Thus apoptosis is the main route to cell death in tetrahydroxyquinone-

treated cells.

Chapter 2

40

JNK and protein phosphatases activation in HL60 cells

To investigate the molecular mechanism underlying tetrahydroxyquinone-

dependent apoptosis induction we studied the activation status of the MAP kinase

family, since this family of kinases is well known to be involved in the control of a

variety of cell survival-controlling pathways [25]. In myeloid leukemia cell lines the

p42/p44 MAP kinase cascade positively regulates differentiation into the monocyte

lineage and is instrumental for phorbolester-dependent differentiation of this cell type,

while inhibition of the JNK has been implicated in 1,25-dihydroxyvitamin D3-

dependent HL60 cell differentiation. Conversely, in HL60 cells JNK activation is linked

to apoptosis [23,24]. We observed that tetrahydroxyquinone treatment strongly

activated JNK. Unexpectedly, only a modest increase in the phosphorylation of p38

MAP kinase with little effect on the activation state of p42/44 MAP kinase was

observed, even at concentrations as high as 100 µM (Fig. 4A). Thus,

tetrahydroxyquinone-induced changes in the activation of MAP kinase family members

are discordant with an effect on differentiation induction, and is consistent with an

effect on apoptosis in this cell type [23, 24].

Despite the induction of oxidative stress by tetrahydroxyquinone, unusual

activations of both protein tyrosine phosphatases and protein serine/threonine

phosphatases coinciding with the activation of PP2A were observed (Fig. 4B).

THQ induces apoptosis of leukemia cells through diminished survival signaling

41

Figure 4. Effect of tetrahydroxyquinone on MAP kinase and protein phosphatase activities.

The phosphorylation and total protein level of MAPK family members (A), phosphoprotein

profiles (employing anti-phospho-amino acid antibodies and phospho-PP2A (B) were evaluated

by immunoblotting. The panel shows tetrahydroxyquine-induced chances in phosphorylation state

(see arrows).

Chapter 2

42

Tetrahydroxyquinone-induced apoptosis coincides with activation of the mitochondrial pathway through diminished protein kinase B activation

Apoptosis can be excecuted through two basic signalling pathways: the

extrinsic pathway and the mitochondrial intrinsic pathway. We observed that

application of tetrahydroxyquinone induced the release of cytochrome c from the

mitochondria at concentration as low as 25 µM (Fig. 5), demonstrating that

tetrahydroxyquinone activates the mitochondrial pathway. Importantly, the

phosphorylation of Bad Ser112 (which leads to cell survival by inhibiting the

mitochondrial pathway) was concomitantly decreased. In agreement,

tetrahydroxyquinone treatment also caused increase of phosphorylation of Ser473 in

protein kinase B (the Bad kinase for Ser112), demonstrating that tetrahydroxyquinone

signalling reduces activity of this anti-apoptotic kinase and, consequently, leads to

apoptosis.

Figure 5. Participation of mitochondrial pathway of apoptosis in tetrahydroxyquinone-

dependent cytotoxicity. Cells were treated with tetrahydroxyquinone as indicated and lysates

were prepared as appropriate (see methods) and analysed by immunoblotting for PKB, phospho-

Bad and total Bad proteinlevels.

THQ induces apoptosis of leukemia cells through diminished survival signaling

43

The importance of mitochondria for apoptosis -induction by

tetrahydroxyquinone was also confirmed by a dramatic increase in total Bad protein

levels (Bad phosphorylation is followed by its ubiquitination and proteolysis and

mediates the inhibitory effect of protein kinase B on apoptosis through the

mitochondrial pathway). In addition, inhibition of two other kinases involved in the

survival signalling: Raf and PKC delta, reinforced our hypothesis that

tetrahydroxyquinone inhibits cell survival signalling pathways (Fig. 5). Furthermore,

additional evidence for survival pathway inhibition by tetrahydroxyquinone was

provided through down regulation of NF-κBp65 and a decrease of phosphorylated IκB

(Fig. 6).

Figure 6. Diminished levels of nuclear factor kappa B by tetrahydroxyquinone. HL60 cells

were treated with tetrahydroxyquinone as indicated and the level of both subunits of NF-κB (p65

and p50) and its phosphorylated inhibitory protein (IκB) were analysed by western blotting

Independent support for this notion was obtained in experiments in which the

protein kinase B activity was artificially increased by transfection with GAG-protein

kinase B, a constitutive active form of protein kinase B, expression construct. As it has

been shown in the Figure 7, HL60 cells transfected with protein kinase B became

resistant to tetrahydroxyquinone, even when treated with 500 µM of the compound.

Chapter 2

44

Figure 7. Viability analysis of HL60 cells transfected with PKB. HL60 cell viability was

evaluated using MTT assay after treatment with tetrahydroxyquinone for 24 h in cell transfected

with a PKB expression construct (filled circles) or empty vector (open circles).

DISCUSSION It is now generally recognised that novel compounds are called for treating

leukaemic disease. This consideration prompted us to investigate the consequences of

tetrahydroxyquinone on HL60 leukaemia cells, a compound most readily known as a

primitive anti-cataract drug, but also expected to act as a redox active molecule. In

agreement with the latter notion, we observed that tetrahydroxyquinone led to ROS

production that coincided with apoptosis through the mitochondrial pathway, and

corresponded with reduced activity of various anti-apoptotic survival molecules

including the protein kinase B pathway in HL60 leukaemia cells but not their

untransformed counterparts. Importantly, transfection of protein kinase B into HL60

cells and thus artificially increasing protein kinase B activity inhibited ROS-dependent

cytotoxicity. We concluded that the remarkable cytotoxic effects tetrahydroxyquinone

on HL60 leukaemia cells are dependent on reduced protein kinase B-dependent survival

THQ induces apoptosis of leukemia cells through diminished survival signaling

45

PKBPKB

BadBad BadP

BadBadP

BadP

BadBadP

Bad

destructionin the proteosome

mitochondrialpathway

apoptosis

NF-κBJNKJNK

Tetrahydroxyquinone

PKCδ

signalling followed by apoptosis through the mitochondrial pathway, probably as a

consequence of tetrahydroxyquinone-dependent ROS generation. Thus tetrahydroxyqui-

none may be representative of a novel class of chemotherapeutic drugs, inducing

apoptosis in cancer cells through diminished survival signalling as a consequence of

ROS production. Proof of this notion awaits in vivo experiments in which the direct

potential of tetrahydroxyquinone as an anticancer drug is directly demonstrated.

Figure 8. Schematic representation of tetrahydroxyquinone effects on HL60 leukaemia cells.

Tetrahydroxyquinone inhibits the anti-apoptotic PKB/Bad, NF-κB, and PKCδ signalling cassettes

and stimulates the pro-apoptotic JNK cassette. Of these effects transfection studies reveal

diminished activation of the anti-apoptotic PKB/Bad signalling cassette to be most important

effect.

Increased ROS production by NADPH oxidase upon macrophage activation is

well established. Earlier studies have shown that such ROS production upon immune

stimulation of the phagocytic cells, apart from its bactericidal function, is also

instrumental for the induction of pro-inflammatory gene transcription [16-18]. Among

the cellular responses to macrophage activation is also cell death, a response probably

required to rid the body from cells damaged by the immune response. HL60 cells share

important characteristics with monocytes and macrophages and the data obtained in the

Chapter 2

46

present study may indicate that this apoptosis is a direct consequence of ROS

production followed by reduced survival signalling and activation of mitochondrial

pathway of cell death, is also important in the induction of apoptosis following

macrophage activation. Final proof of this notion, however, awaits experiments in

which activation-induced cell death is investigated in the presence and absence of

glutathione and N-acetyl-L-cysteine.

The molecular mechanisms by which ROS participate in inflammatory gene

expression are obscure at best. In the present study we observed that ROS reduced the

apparent activity of the NF-κB pathway, as deduced from the decrease in expression of

both the ratio between p65 expression and I-κB as well as the reduced phosphorylation

of I-κB. As the NF-κB pathway is a potent survival signal, this down regulation

corresponds well with the reduced survival signalling induced by ROS formation

detected in this study. It is, however, more difficult to reconcile with the role of ROS

generation in mediating inflammatory gene expression, as NF-kB is in general a strong

pro-inflammatory transcription factor. Importantly, however, a recent study by

Blanchette et al. [29] indicates that the contribution of NF-κB to macrophage activation-

associated gene expression is minimal, hence our results are not necessarily at bay with

the established role of ROS in the induction of inflammatory gene expression.

Interestingly, increased JNK activity has recently come up as an important mediator of

inflammatory gene expression in inflammatory bowel disease in vivo [30]. Hence, the

ROS-induced activation of JNK may function in the induction of inflammatory gene

expression.

Despite the intricacies of ROS action in cellular physiology, the present study

has shown a remarkable action of tetrahydroxyquinone on HL60 leukaemia cells, acting

through a novel survival signalling-inhibiting mechanism. Recent data indicate that

impaired survival signalling through PKB/Akt has substantial clinical promise for the

treatment of chemotherapy-resistant leukaemia [31]. Together, these observations make

tetrahydroxyquinone a candidate drug for the treatment of leukaemia and further studies

are under progress addressing its potential usefulness for combating disease.

THQ induces apoptosis of leukemia cells through diminished survival signaling

47

REFERENCES

1. Larson RA, Daley GQ, Schiffer CA, et al. Treatment by design in leukemia, a meeting

report, Philadelphia, Pennsylvania, December 2002. Leukemia 2003;17: 2358-2382.

2. Hisatake J, O´Kelly J, Uskokovic MR, Tomoyasu S, Koeffler P. Novel vitamin D3 analog,

21-(3-methyl-3-hydroxy -butyl)-19-nor D3, that modulates cell growth, differentiation,

apoptosis, cell cycle, and induction of PTEN in leukemic cells. Blood 2001;97:2427-2433.

3. Tsuzuki S, Kitajima K, Nakano T, Glasow A, Zelent A, Enver T. Cross talk between retinoic

acid signaling and transcription factor GATA-2. Mol Cell Biol. 2004; 24:6824-6836.

4. Allen RG, Tresini M. Oxidative stress and gene regulation.

Free Radic Biol Med. 2000;28(3):463-499.

5. MacDonald GM, Steenhuis JJ, and Barry BA. A difference Fourier transform infrared

spectroscopic study of chlorophyll oxidation in hydroxylamine-treated photosystem II. J Biol

Chem. 1995;270:8420-8428.

6. Daou GB, Srivastava AK. Reactive oxygen species mediate Endothelin-1-induced activation

of ERK1/2, PKB and Pyk2 signalling, as well as protein synthesis, in vascular smooth

muscle cells. Free Radic Biol Med. 2004;37:208-215.

7. Shen HM, Lin Y, Choksi S, et al. Essential Roles of Receptor-Interacting Protein and

TRAF2 in Oxidative Stress-Induced Cell Death. Mol Cell Biol. 2004;24(13):5914-5922.

8. Jang BC, Paik JH, Jeong HY, et al. Leptomycin B-induced apoptosis is mediated through

caspase activation and down-regulation of Mcl-1 and XIAP expression, but not through the

generation of ROS in U937 leukemia cells. Biochem Pharmacol. 2004;68(2):263-274.

9. Murray J, Walmsley SR, Mecklenburgh KI, et al. Hypoxic regulation of neutrophil apoptosis

role: of reactive oxygen intermediates in constitutive and tumor necrosis factor alpha-

induced cell death. Ann NY Acad Sci. 2003;1010:417-425.

10. Ryter SW, Tyrrell RM. Singlet molecular oxygen ((1)O2): a possible effector of eukaryotic

gene expression. Free Radic Biol Med. 1998:24(9):1520-1534.

11. Wiemels J, Wiencke JK, Varykoni A, Smith MT. Modulation of the toxicity and

macromolecular binding of benzene metabolites by NAD(P)H:Quinone oxidoreductase in

transfected HL-60 cells. Chem Res Toxicol. 1999:12(6):467-475.

12. Chen Q, Vázquez EJ, Moghaddas S, Hoppel CL, Lesnefsky EJ. Production of reactive

oxygen species by mitochondria: central role of complex III. J Biol Chem.

2003;278(38):36027-36031.

13. Tudor G, Gutierrez P, Aguilera-Gutierrez A, Sausville EA. Cytotoxicity and apoptosis of

benzoquinones: redox cycling, cytochrome c release, and BAD protein expression. Biochem

Pharmacol. 2003;65:1061-1075.

Chapter 2

48

14. Anderson ME. Glutathione: an overview of biosynthesis and modulation. Chem Biol

Interact. 1998;111-112:1-14.

15. Matés JM, Sánchez-Jiménez FM. Role of reactive oxygen species in apoptosis: implications

for cancer therapy. Int J Biochem Cell Biol. 2000;32(2):157-170.

16. Schoonbroodt S, Piette J. Oxidative stress interference with the nuclear factor-kappa B

activation pathways. Biochem Pharmacol. 2000;60(8):1075-1083.

17. Victor VM, De la Fuente M. N-acetylcysteine improves in vitro the function of

macrophages from mice with endotoxin-induced oxidative stress. Free Radic Res.

2002;36(1):33-45.

18. Ryan KA, Smith MF Jr, Sanders MK, Ernst PB. Reactive oxygen and nitrogen species

differentially regulate Toll-like receptor 4-mediated activation of NF-kappa B and

interleukin-8 expression. Infect Immun. 2004;72(4):2123-2130.

19. Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to

proliferation and cytotoxicity assay. J Immunol Methods 1983;65:55-63.

20. Denizot F, Lang R. Rapid colorimetric assay for cell growth and survival. modifications to

the tetrazolium dye procedure giving improved sensitivity and reliability. J Immunol

Methods 1986;89:271-277.

21. Aoyama H, Melo PS, Granjeiro PA, Haun M and Ferreira CV. Cytotoxicity of okadaic acid

and kinetic characterization of protein tyrosine phosphatase activity in V79 fibroblasts.

Pharm Pharmacol Comm. 2000;6:331-334.

22. Hartree EF. Determination of proteins: a modification of the Lowry method that gives a

linear photometric response. Anal Biochem. 1972;48:422-427.

23. Zhu W-H., Majluf-Cruz A, Omburo GA. Cyclic AMP-specific phosphodiesterase inhibitor

rolipram and RO-20-1724 promoted apoptosis in HL-60 promyelocytic leukemic cells via

cyclic AMP-independent mechanism. Life Sci. 1998;63(4):265-274.

24. Freire AG, Melo PS, Haun M, Dúran N, Aoyama H, Ferreira CV. Cytotoxic effect of the

diterpene lactone dehydrocrotonin from Croton cajucara on human promyelocytic leukemia

cells. Planta Medica 2003;69:67-69.

25. Versteeg HH, Evertzen MW, van Deventer SJ, Peppelenbosch MP. The role of

phosphatidylinositide-3-kinase in basal mitogen-activated protein kinase activity and cell

survival. FEBS Lett. 2000 465:69-73

26. Wang XN, Studzinski GP. Inhibition of p38MAP kinase potentiates the JNK/SAPK pathway

and AP-1 activity in monocytic but not in macrophage or granulocytic differentiation of

HL60 cells. J Cell Biochem. 2001;82:68-77.

THQ induces apoptosis of leukemia cells through diminished survival signaling

49

27. Sthadeim TA, Kucera GL. c-Jun N-terminal kinase/stress-activated protein kinase

(JNK/SAPK) is required for mitoxantrone and anisomycin-induced apoptosis in HL-60 cells.

Leuk Res. 2002;26:55-65.

28. Chénais B, Andriollo M, Guiraud P, Belhoussine R, Jeannesson P. Oxidative stress

involvement in chemically induced differentiation of K562 cells. Free Radical Biol Med.

2000;28:18-27.

29. Blanchette J, Jaramillo M, Olivier M. Signalling events involved in interferon-gamma-

inducible macrophage nitric oxide generation. Immunol. 2003; 108:513-22.

30. Hommes D, van den Blink B, Plasse T, Bartelsman J, Xu C, Macpherson B, Tytgat G,

Peppelenbosch M, Van Deventer S. Inhibition of stress-activated MAP kinases induces

clinical improvement in moderate to severe Crohn's disease. Gastroenterology. 2002 122:7-

14

31. Martelli AM, Tazzari PL, Tabellini G, et al. A new selective AKT pharmacological inhibitor

reduces resistance to chemotherapeutic drugs, TRAIL, all-trans-retinoic acid, and ionizing

radiation of human leukemia cells. Leukemia 2003;17:1794-1805.

Chapter 2

50