+ All Categories
Home > Documents > Anti Ox Id Ante

Anti Ox Id Ante

Date post: 05-Apr-2018
Category:
Upload: alene-vanessa-azevedo-santos
View: 215 times
Download: 0 times
Share this document with a friend

of 12

Transcript
  • 8/2/2019 Anti Ox Id Ante

    1/12

    Original Contribution

    Bilirubin inhibits Plasmodium falciparum growth through

    the generation of reactive oxygen species

    Sanjay Kumara, Mithu Guha b, Vinay Choubey a, Pallab Maity b, Kumkum Srivastava c,Sunil K. Puri c, Uday Bandyopadhyay b,

    aDrug Target Discovery and Development Division, Central Drug Research Institute, Chatter Manzil Palace, Mahatma Gandhi Marg,

    Lucknow 226001, Uttar Pradesh, Indiab Division of Infectious Diseases and Immunology, Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India

    c Parasitology Division, Central Drug Research Institute, Chatter Manzil Palace, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India

    Received 3 July 2007; revised 28 September 2007; accepted 17 October 2007Available online 17 November 2007

    Abstract

    Free heme is very toxic because it generates highly reactive hydroxyl radicals (U

    OH) to cause oxidative damage. Detoxification of free heme by

    the heme oxygenase (HO) system is a very common phenomenon by which free heme is catabolized to form bilirubin as an end product.

    Interestingly, the malaria parasite, Plasmodium falciparum, lacks an HO system, but it forms hemozoin, mainly to detoxify free heme. Here, we

    report that bilirubin significantly induces oxidative stress in the parasite as evident from the increased formation of lipid peroxide, decrease in

    glutathione content, and increased formation of H2O2 andU

    OH. Bilirubin can effectively inhibit hemozoin formation also. Furthermore, results

    indicate that bilirubin inhibits parasite growth and induces caspase-like protease activity, up-regulates the expression of apoptosis-related protein

    (Gene ID PFI0450c), and reduces the mitochondrial membrane potential.U

    OH scavengers such as mannitol, as well as the spin trap -phenyl-n-

    tert-butylnitrone, effectively protect the parasite from bilirubin-induced oxidative stress and growth inhibition. These findings suggest that

    bilirubin, through the development of oxidative stress, induces P. falciparum cell death and that the malaria parasite lacks an HO system probablyto protect itself from bilirubin-induced cell death as a second line of defense.

    2007 Elsevier Inc. All rights reserved.

    Keywords: Bilirubin; Hydroxyl radical; Apoptosis; Malaria parasite; Hemozoin; Free radicals

    The malaria parasite (Plasmodium spp.), during intraery-

    throcytic stages, digests huge quantities of hemoglobin and

    releases a large amount of highly toxic redox-active free heme

    [1]. To overcome the free heme toxicity, the malaria parasite is

    equipped with unique heme detoxification systems. Amongthese, hemozoin formation is considered to be the main heme

    detoxification system and the inhibition of hemozoin formation

    leads to parasite death [1,2]. In contrast, detoxification of free

    heme by the heme oxygenase-1 (HO-1) system is a very

    common process in higher eukaryotes, including mammals [3].

    To degrade heme, HO-1 requires the microsomal NADPH-

    cytochrome P-450 reductase [4] and shunts reducing equivalents

    from NADPH-cytochrome P-450 reductase to the -methene

    bridge and cleaves the tetrapyrrolic ring of heme, causing the

    liberation of CO plus an equimolar amount of biliverdin.

    Biliverdin is converted into bilirubin by biliverdin reductase [5].Interestingly, the malaria parasite lacks this common heme

    oxygenase (www.plasmodb.org) to catabolize free heme.

    Bilirubin has both antioxidant and pro-oxidant properties [6].

    Bilirubin, at low physiological concentrations (0.0110 M),

    scavenges reactive oxygen species (ROS), reduces oxidant-

    induced cellular injury, and attenuates oxidative stress [7,8].

    Unconjugated bilirubin (UCB) is a scavenger of ROS such as

    hydrogen peroxide, peroxynitrite, and peroxyl radicals, both in

    vivo and in vitro, and plays a key physiological role in cyto-

    protection against oxidant-mediated cell damage [9,10]. In

    contrast, elevated concentrations (N20 M) of UCB have

    Available online at www.sciencedirect.com

    Free Radical Biology & Medicine 44 (2008) 602613www.elsevier.com/locate/freeradbiomed

    Abbreviations: HO, Heme oxygenase; UOH, Hydroxyl radical; PBN,

    -phenyl-tert-butylnitrone. Corresponding author. Fax: +91 33 24730284.

    E-mail address: [email protected] (U. Bandyopadhyay).

    0891-5849/$ - see front matter 2007 Elsevier Inc. All rights reserved.doi:10.1016/j.freeradbiomed.2007.10.057

    http://www.plasmodb.org/mailto:[email protected]://dx.doi.org/10.1016/j.freeradbiomed.2007.10.057http://dx.doi.org/10.1016/j.freeradbiomed.2007.10.057mailto:[email protected]://www.plasmodb.org/
  • 8/2/2019 Anti Ox Id Ante

    2/12

    deleterious effects in both neuronal and nonneuronal tissues

    [11]. It develops oxidative stress by generating intracellular

    ROS in Hepa1c1c7 cells and causes lipid peroxidation [12]. It

    causes brain damage in newborn piglets by increasing lipid

    peroxidation [13]. Oxidative stress is frequently associated with

    the induction of apoptosis [14,15]. UCB induces apoptosis in

    astrocytes at moderately elevated concentrations [16]. UCB-mediated programmed cell death was also reported in cultured

    rat aortic smooth muscle cells and bovine brain endothelial cells

    [17,18]. Thus the concentration as well as the type of cell is vital

    for bilirubin to exert when or where it will be pro-oxidant or

    antioxidant. Here, we report that bilirubin develops oxidative

    stress in Plasmodium falciparum and inhibits parasite growth.

    Further, we give evidence that bilirubin-induced oxidative

    stress-mediated parasite death is associated with the induction

    of caspase-like protease activity, up-regulation of the expression

    of apoptosis-related protein (Gene ID PFI0450c), and reduction

    of the mitochondrial membrane potential (m).

    Materials and methods

    Hemin, RPMI 1640, saponin, glutathione (GSH), caspase-3

    assay kit, Triton X-100, proteinase K, mannitol, -phenyl-n-

    tert-butylnitrone (PBN), Nonidet P-40, dichlorofluorescein

    diacetate, thiobarbituric acid, trichloroacetic acid (TCA),

    tetraethoxypropane, antimycin A, Hoechst 33342, and bilirubin

    were purchased from Sigma (St. Louis, MO, USA). Albumax II

    was procured from Gibco BRL (Grand Island, NY, USA).

    Giemsa stain was purchased from Qualigens Fine Chemicals

    (India). [3H]Hypoxanthine and Ready to Go RT-PCR beads

    were purchased from Amersham Biosciences (Arlington

    Heights, IL, USA). RNeasy kit was purchased from Qiagen(Valencia, CA, USA). 5,5,6,6-tetrachloro-1,1,3,3-tetraethyl-

    benzimidazolcarbocyanine iodide (JC1) was purchased from

    Molecular Probes (Eugene, OR, USA). All other chemicals

    were of analytical grade purity. Bilirubin (Sigma Chemical Co.)

    was further purified to remove biliary lipids as described earlier

    [19]. The purified bilirubin was dissolved in 0.01 N NaOH

    followed by dilution with triple-distilled water to make 1 ml

    of a 1 mM stock and prepared fresh each time before use.

    Immediately after the preparation, the bilirubin stock solution

    was used. The stock solution was kept on ice protected from

    light and all incubations containing bilirubin were carried out in

    the dark to avoid degradation.

    Parasite culture

    The ring-synchronized P. falciparum (clone NF-54) was

    grown as described [20] at a hematocrit level of 5% in complete

    RPMI medium (CRPMI; RPMI 1640 medium supplemented

    with 25 mM Hepes, 50 g ml1 gentamycin, 370 M hypo-

    xanthine, and 0.5% (w/v) Albumax II) in tissue-culture flasks

    (25 and 75 cm2) with loose screw caps. Used medium was

    replaced with fresh medium once in 24 h and the culture was

    routinely monitored through Giemsa staining of thin smears.

    The ring-synchronized P. falciparum was usually cultured for

    48 h to complete one cycle from ring to schizont stage [21]. As

    the effect of drug or agent may be stage specific, to see the effect

    of bilirubin on the growth of the parasite at any stage, it was

    cultured for 48 h in the presence or absence of different con-

    centrations of bilirubin.

    Free (unbound) bilirubin in culture medium containing Albu-

    max II (0.5% (w/v); a lipid-rich albumin from bovine serum,

    generally used to culture P. falciparum) [22] was measured atdifferent total bilirubin levels using the peroxidase method as

    described [23] in a Shimadzu UV/Vis 1700 spectrophotometer.

    Isolation of parasites from infected erythrocytes and

    preparation of parasite lysate

    Parasites were isolated as described previously [24]. Briefly,

    erythrocytes with10% parasitemia were centrifuged at 800 g

    for 5 min, washed, and resuspended in cold phosphate-buffered

    saline (PBS) (137 mM NaCl, 2.7 mM KCl, 5.3 mM Na 2HPO4,

    and 1.8 mM KH2PO4). An equal volume of 0.5% saponin in

    PBS (final concentration 0.25%) was added to the erythrocytesuspension and kept on ice for 15 min. It was centrifuged at

    1300 gfor 5 min to get the parasite pellet, and finally the pellet

    was washed with PBS and the isolated parasites were lysed in

    PBS by mild sonication (30-s pulse, bath-type sonicator) at 4C.

    The whole lysate was stored at 20C for future use. The

    protein concentration in the parasite lysate was estimated as

    described [25].

    Measurement of lipid peroxidation as an index of oxidative

    damage

    P. falciparum culture (4% parasitemia, ring synchronized)

    was incubated in the absence and presence of differentconcentrations of bilirubin for 48 h. After incubation, parasites

    were isolated and resuspended in PBS (500 l) to prepare

    parasite lysate as described above and the lipid peroxidation

    product from the lysate was measured as described earlier

    [26,27]. In brief, an aliquot (50 l) of the parasite lysate was

    allowed to react with 100 l of trichloroacetic acidthiobarbi-

    turic acidHCl reagent containing 0.01% butylated hydroxyto-

    luene, heated in a boiling water bath for 15 min, cooled, and

    centrifuged, and the supernatant was used for thiobarbituric

    acid-reactive substances determination at 535 nm using tetra-

    ethoxypropane as standard and expressed as nanomoles of lipid

    peroxide/milligram of lysate protein.

    Measurement of reduced glutathione

    P. falciparum (4% parasitemia) was cultured in the presence

    or absence of different concentrations of bilirubin. After 48 h of

    treatment, the culture was washed twice with PBS and the

    parasite was isolated from the infected erythrocytes as described

    above. GSH content from control and bilirubin-treated parasites

    was determined as described earlier[15,27,28]. In brief, parasite

    lysate (50 l) was mixed with an equal volume of 10% TCA and

    the protein precipitate was removed by centrifugation. The

    supernatant was added to an equal volume of 0.8 M TrisCl, pH

    9, containing 20 mM 5,5-dithionitrobenzoic acid to yield the

    603S. Kumar et al. / Free Radical Biology & Medicine 44 (2008) 602613

    http://dx.doi.org/10.1016/j.freeradbiomed.2007.10.057
  • 8/2/2019 Anti Ox Id Ante

    3/12

    yellow chromophore of thionitrobenzoic acid, which was

    measured at 412 nm. GSH content was measured as nmol/mg

    parasite lysate protein.

    Measurement of intraparasitic H2O2

    P. falciparum (2% parasitemia) was cultured in the presenceor absence of different concentrations of bilirubin for a period

    of 48 h and intraparasitic H2O2 was measured as described [29].

    In brief, the culture (after bilirubin treatment) was further

    incubated for 30 min in CRPMI containing 2,7-dichlorofluor-

    escein diacetate (10 M). The culture was then washed twice

    with PBS and the parasites were isolated from control and

    treated groups and isolated parasites were lysed as described

    above. H2O2 was measured in control or bilirubin-treated

    parasites by measuring the fluorescent dichlorofluorescein

    formed. Fluorescence intensities were recorded from the lysates

    in a PerkinElmer LS 50B spectrofluorimeter in a 5-mm path-

    length quartz cell in a total volume of 1 ml at wavelengths 502and 523 nm for excitation and emission, respectively. H2O2content was expressed as fluorescence intensity/milligram of

    parasite lysate protein.

    Measurement of hydroxyl radical (UOH) generation

    UOH generated in the P. falciparum after bilirubin treatment

    at various concentrations was measured using dimethyl

    sulfoxide (DMSO) as a UOH scavenger [27,30]. In brief,

    P. falciparum culture (200 l) (2% parasitemia, ring+ early

    trophozoites stage) was grown in multiwell plates in the

    presence or absence of different concentrations of bilirubin

    containing 20 l of 25% DMSO for 48 h. DMSO (20 l) wasadded along with the stipulated concentrations of bilirubin each

    time when the medium was changed (once in 24 h). A negative

    control (parasite only) was made without DMSO and bilirubin

    as a reagent blank. After 48 h, the culture was centrifuged at

    800 g for 5 min, washed, and resuspended in cold PBS. The

    parasites were isolated as described above and the isolated

    parasites were lysed in triple-distilled water and processed for

    the extraction of methanesulfinic acid formed by the reaction ofUOH with DMSO. Methanesulfinic acid formed was allowed to

    react with Fast Blue BB salt and the intensity of the resulting

    yellow chromophore was measured at 425 nm using benzene-

    sulfinic acid as standard.

    Effect of bilirubin on hemozoin formation

    Hemozoin (-hematin) formation catalyzed by parasite

    lysate in vitro was assayed in the presence or absence of

    different concentrations of bilirubin by following the method

    described earlier[31,32]. The assay mixture contained in a final

    volume of 1 ml: 100 mM sodium acetate buffer, pH 5.2, 100 M

    hemin, and parasite lysate (20 l) in the absence or presence of

    various concentrations of bilirubin. The reaction was initiated

    by the addition of hemin and incubated for 12 h at 37C. The

    reaction was terminated by centrifugation at 15,000g for

    10 min at room temperature. The pellet was washed twice with

    100 mM TrisHCl buffer, pH 7.8, containing 2.5% SDS and

    finally with 100 mM bicarbonate buffer, pH 9.2. The insoluble

    pellet (hemozoin) was solubilized in 50 l of 2 N NaOH

    and diluted further to 1 ml with 2.5% SDS. The absorbance

    of the solution was recorded at 400 nm and an extinction

    coefficient of 91 mM1 cm1 [33] was used to quantitate hemo-

    zoin formation. In P. falciparum, the amount of hemozoinformed in the presence or absence of bilirubin was also measured

    as described earlier [34].

    Measurement of heme content

    Heme content in control and bilirubin-treated P. falciparum

    was measured as described earlier [35]. In brief, P. falciparum

    was cultured in the presence or absence of varying concentra-

    tions of bilirubin for 48 h. Then the culture was centrifuged

    to pellet the cells and the cell pellet was washed in PBS to

    remove the bilirubin. Then concentrated formic acid (1 ml)

    was added to solubilize each pellet and the heme concentrationof the formic acid solution was determined in a Shimadzu

    UV/Vis 1700 spectrophotometer at 398 nm (extinction coeffi-

    cient 1.56105 M1 cm1). Heme content was expressed as

    nmol/mg of cell protein.

    In vivo growth of P. yoelii and isolation of trophozoite-infected

    red cells

    Mice (Swiss albino, 1820 g) were infected with P. yoelii

    by intraperitoneal passage of 1 106 infected erythrocytes [36]

    and parasitemia was monitored by microscopic examination of

    Giemsa-stained thin blood smear. After 4 days of inoculation,

    blood was collected in acid citrate dextrose (0.0347 M citricacid, 0.0748 M sodium citrate, 0.1359 M dextrose) at

    approximately 50% parasitemia. Infected blood was passed

    through CF-11 cellulose (Whatman) to remove white blood

    cells [37]. The collected red blood cells (RBC) were washed and

    trophozoite-rich infected RBC were isolated as described [38].

    Measurement of mitochondrial transmembrane potential

    Isolated trophozoite-rich infected RBC at a concentration of

    5106/ml in RPMI 1640 medium containing 1% fetal bovine

    serum were incubated in the presence or absence of bilirubin

    (40 M) for 1 h at room temperature (30C). For positivecontrol, the same number of cells were incubated with anti-

    mycin A. The infected RBC were washed (three times) in PBS

    to remove excess bilirubin or antimycin A and the cell pellet

    was suspended in 1 ml of CRPMI. Then JC1 (153 nM) was

    added to each cell suspension and incubated for 10 min in the

    dark at 25C. At the end of the incubation, the fluorescence was

    recorded in a PerkinElmer Life Sciences Lambda L.S 50B

    spectrofluorimeter (excitation 490; emission 590 nm) [39]. The

    JC1 uptake (J-aggregate formation) was also analyzed by

    fluorescence microscopy using JC1-treated cell suspension. In

    brief, the JC1-treated cell suspension was washed (three times)

    in CRPMI and the resulting infected cell pellet was suspended

    in 100 l CRPMI. Twenty microliters of this cell suspension

    604 S. Kumar et al. / Free Radical Biology & Medicine 44 (2008) 602613

  • 8/2/2019 Anti Ox Id Ante

    4/12

    was used to analyze the formation of J-aggregate (emission

    590 nm, TX2 green filter) as a measure of mitochondrial uptake

    and monomer (emission 530 nm, I3 blue filter) quickly under

    100 oil immersion lens in a Leica DM LB 2 fluorescence

    microscope.

    Assay of caspase-like (DEVDase) activity

    To study the activation of caspase, caspase-3-like activity

    was measured in whole P. falciparum lysate, using commer-

    cially available kits and according to the manufacturer's pro-

    tocol (Sigma) as described earlier [15]. In brief, P. falciparum

    culture (4% parasitemia) was incubated in the absence or

    presence of bilirubin (40 M) for 48 h at 37C in the dark.

    After incubation, parasites were isolated and sonicated in

    PBS (100 l) to prepare parasite lysate. Parasite lysate (10 l

    containing 30 g protein) was mixed with assay buffer and

    caspase-3 substrate (Ac-DEVD-pNA) in a microtiter plate.

    After 24 h of incubation at 37C, the absorbance of pNAreleased was measured at 405 nm in a microtiter plate reader.

    Caspase-3-like activity was confirmed using the caspase-3

    inhibitor provided in the kit.

    Semiquantitative RT-PCR for apoptosis-related protein

    P. falciparum culture (2% parasitemia) was incubated in

    the absence or presence of bilirubin (40 M) for 48 h. After

    incubation, the culture was pelleted and washed twice with

    PBS. The parasite pellet was used for RNA isolation using the

    RNeasy kit (Qiagen). Freshly isolated parasites were immedi-

    ately suspended in RNA Later solution and RNA was extracted

    from the parasites using the Qiagen RNeasy Protect kit ac-cording to the manufacturer's instructions. Nucleic acid bound

    to the RNeasy column was incubated with 5 kunitz units of

    RNase-free DNase in 50 mM TrisHCl (pH 7.5) and 10 mM

    MgCl2 for 20 min at 37C to remove DNA contamination in the

    RNA preparation. The purity of the RNA was checked in 1%

    agarose gel and quantitated by measuring the OD at 260 nm.

    An equal amount of RNA (1.5 g) was used for RT-PCR of

    apoptosis-related protein (Gene ID PFI0450c) using the

    following sets of primers: forward primer (1 M final con-

    centration), 5-ATGAATATTGAAAAAGCCG-3; reverse pri-

    mer (1 M final concentration), 5-CATATAATCTTCTTCG-

    TTGAAATC-3 [40]. RT-PCR was performed using Ready toGo RT-PCR beads (Amersham Pharmacia) with the following

    PCR program: cDNA synthesis at 42C for 30 min; 94C for

    2 min for initial denaturation; then 35 cycles of denaturation at

    94C for 1 min, annealing at 55C for 1 min, extension at 72C

    for 1.5 min; and then 72C for 7 min. Simultaneously, positive

    control primers for seryl-tRNA synthetase were added in each

    RT-PCR. Seryl-tRNA synthetase is expressed equally in each

    stage of the P. falciparum [41]. Primer sequences used for seryl-

    tRNA synthetase were 5-GAGGAATTTTACGTGTTCAT-

    CAA-3 (forward) and 5-GATTACTTGTAGGAAAGAATCC-

    TTC-3 (reverse). RT-PCR products were analyzed through

    electrophoresis on 1% agarose gel in TAE buffer at 10 V/cm and

    documented.

    Nuclear morphology by Hoechst staining

    P. falciparum culture (2% parasitemia, ring stage) was

    incubated in the absence or presence of bilirubin (40 M)

    for 48 h. After incubation, P. falciparum culture (control and

    bilirubin (40 M)-treated) was centrifuged at 2500 rpm for

    5 min to remove the medium and the pellet containing parasite-infected red cells was suspended in PBS. Hoechst 33342 (10 g/

    ml) was added to it and kept for 10 min in the dark at 37C.

    Then the cells were washed three times in PBS to remove

    the excess fluorescent stains and fixed with 3.7% paraformal-

    dehyde in PBS and the parasite was then visualized at 100

    with oil using a UV filter under a Leica DM LB 2 fluorescence

    microscope.

    Assay of antimalarial activity of bilirubin

    Inhibition ofP. falciparum growth was studied by following

    [

    3

    H]hypoxanthine uptake as described earlier [42]. Synchroni-zation of the parasites to uniform ring stage was achieved using

    5% aqueous D -sorbitol as described earlier [43]. To see the

    effect of bilirubin, the ring-synchronized P. falciparum (para-

    sitemia 0.51%) was cultured in multiwell (200 l/well) plates

    in the presence or absence of different concentrations of

    bilirubin. After 48 h [3H]hypoxanthine (0.7 Ci/well) was

    added to each well and further cultured for 48 h to monitor

    parasite viability by measuring incorporation of [3H]hypox-

    anthine in parasite nucleic acids. P. falciparum culture was

    harvested and washed twice in PBS. The parasite pellet was

    dissolved in 100 l of 3 N NaOH by keeping it at 37C for 6 h

    followed by scintillation counting for [3H]hypoxanthine uptake

    measurement.

    Statistical analysis

    Data shown are means SEM. Statistical analysis for

    parametric data was calculated using Student's t test or

    ANOVA wherever applicable. The ANOVA was followed by

    post hoc analysis (multiple comparison ttest) for the evaluation

    of the difference between individual groups. For nonparametric

    data analysis, the MannWhitney U test and KruskalWallis

    ANOVA (wherever applicable) were performed. Kruskal

    Wallis ANOVA was followed by multiple comparison test

    (Holm-Sidak methods). A pb0.05 was considered statisticallysignificant.

    Results

    Bilirubin induces oxidative stress in P. falciparum

    GSH level and the formation of lipid peroxidation product

    were measured after incubation of P. falciparum with various

    concentrations of bilirubin (Fig. 1). Bilirubin decreased GSH

    levels (Fig. 1A) in a concentration-dependent manner. GSH

    levels decreased 50 and 80% at 10 and 30 M bilirubin,

    respectively (Fig. 1A, inset). The decrease in GSH level was

    also associated with the increased formation of lipid peroxide in

    605S. Kumar et al. / Free Radical Biology & Medicine 44 (2008) 602613

  • 8/2/2019 Anti Ox Id Ante

    5/12

    the parasite (Fig. 1B). Bilirubin increased lipid peroxidation by

    65% at 10 M and 150% at 30 M compared with the control

    value (Fig. 1B, inset). In order to assess whether increased lipid

    peroxidation and GSH depletion, the indicators of oxidative

    stress, are due to generation of H2O2, the intraparasitic H2O2

    level was measured after incubation with various concentrationsof bilirubin. Interestingly, bilirubin increased the generation of

    intraparasitic H2O2 in a concentration-dependent manner (Fig.

    2A), causing a 200% increase at 10 M and 400% increase at

    30 M bilirubin (Fig. 2A, inset). Furthermore, the parasite,

    when incubated with bilirubin, showed gradual increased

    generation of UOH as a function of bilirubin concentration

    (Fig. 2B and inset).

    Bilirubin inhibits hemozoin formation

    As free heme is one of the major sources of ROS generation,

    we expected that it should accumulate to cause oxidative stress

    if hemozoin formation is inhibited in the parasite in the presence

    of bilirubin. Hemozoin formation was followed using lysates of

    the multidrug-resistant P. yoelii (Fig. 3A) and chloroquine-

    sensitive P. falciparum (Fig. 3B). The results clearly indicate

    that bilirubin prevents hemozoin formation in a concentration-

    dependent manner in both cases (Figs. 3A and B). IC50 values

    were found to be 14 M forP. yoelii (Fig. 3A, inset) and 10 M

    forP. falciparum (Fig. 3B, inset) lysates. Bilirubin also inhibitshemozoin formation in P. falciparum in culture with an IC50 of

    8 M (Fig. 3C and inset).

    The inhibition of hemozoin formation in the parasite may

    lead to the accumulation of heme. The data indicate that

    bilirubin concentration-dependently increases the heme content

    in the parasite (Fig. 4). Thus, the pro-oxidant effect of bilirubin

    is probably due to the inhibition of hemozoin formation and

    subsequent accumulation of toxic heme.

    It is accepted that the free or unbound form of bilirubin

    mainly mediates the biological effect. Because P. falciparum

    was cultured for 48 h (for one full intraerythrocytic cycle from

    ring to schizont stage) in the presence of various concentrationsof bilirubin to follow oxidative stress and hemozoin formation

    in the parasite, the free bilirubin (Bf) concentration may likely

    Fig. 1. Bilirubin develops oxidative stress in P. falciparum. (A) GSH and (B)

    lipid peroxide were measured in P. falciparum in the presence or absence of

    various concentrations of bilirubin. The data presented are meansSEM (n =6).

    *pb0.05 vs control, ***pb0.001 vs control.

    Fig. 2. Bilirubin stimulates the generation of intraparasitic H2O2 and hydroxyl

    radical (U

    OH). (A) H2O2 and (B)U

    OH were measured in P. falciparum in the

    presence or absence of various concentrations of bilirubin as described

    under Materials and methods. The data presented are means SEM (n =6).***pb0.001 vs control.

    606 S. Kumar et al. / Free Radical Biology & Medicine 44 (2008) 602613

  • 8/2/2019 Anti Ox Id Ante

    6/12

    decline during the 48 h of incubation due to oxidative

    degradation. Therefore, the Bf level was measured after 48 h.

    The data indicated that the Bf level was decreased significantly

    and the Bfconcentration, which was initially 40 M, was found

    to be 9 M (Fig. 5). Because bilirubin offers maximum in-

    hibition of hemozoin formation in the parasite in culture at

    40 M (initial level), we used this concentration in most of the

    experiments later on.

    Bilirubin reduces mitochondrial transmembrane potential of

    the malaria parasite

    The generation of ROS and the associated oxidative stress

    activate the mitochondrial pathway of apoptosis [14] or cause

    mitochondrial dysfunction [44]. In order to investigate whether

    oxidative stress induced by bilirubin can lead to mitochondrial

    dysfunction in the malaria parasite, alteration of the mitochon-

    drial m, a marker for dysfunction, was measured. The mwas measured by the fluorescence change in the membrane-

    potential-sensitive dye, JC1. In intact healthy mitochondria

    with higherm, JC1 would accumulate in the mitochondrial

    matrix to form the J-aggregate, showing intense fluorescence

    at 590 nm. Mitochondria with open transition pores have low

    m and would accumulate less JC1, leading to lower for-

    mation of aggregates, thereby showing weak fluorescence at590 nm. Bilirubin can effectively decrease m as indicated by

    spectrofluorimetric analysis (Fig. 6A) and fluorescence micro-

    scopic studies (Fig. 6B). Antimycin A, a known probe used to

    decrease mitochondrial membrane potential [45], was used as a

    positive control. Fluorescence microscopic analysis clearly

    indicates that the formation of J-aggregates (red fluorescence,

    590 nm) (Fig. 6B, images a) or the ratio of 590 nm/530 nm

    (Fig. 6B, images c) in the trophozoite-infected red cell (control)

    was higher than in bilirubin-or antimycin A-treated trophozoite-

    infected red cells. In contrast, the green fluorescence (JC1Fig. 3. Inhibition of hemozoin formation by bilirubin. Hemozoin formation was

    followed in the presence or absence of different concentrations of bilirubin as

    indicated. (A) Formation of hemozoin using P. yoelii lysate. (B) Formation ofhemozoin using P. falciparum lysate. (C) Hemozoin content ofP. falciparum in

    culture in the presence or absence of various concentrations of bilirubin as

    indicated. The data presented are meansSEM (n =6). *pb0.05, **pb0.01,

    ***pb0.001 vs control.

    Fig. 4. Bilirubin increases heme content in the parasite. Heme content was

    measured as described under Materials and methods. The data presented are

    meansSEM (n =6). **pb0.01, ***pb0.001 vs control.

    Fig. 5. Measurement of free bilirubin in culture after 48 h of incubation. Free

    bilirubin concentration was measured as described under Materials and methods.The data presented are meansSEM (n =6). *pb0.05, ***pb0.001.

    607S. Kumar et al. / Free Radical Biology & Medicine 44 (2008) 602613

  • 8/2/2019 Anti Ox Id Ante

    7/12

    monomer, 530 nm) was much lower in the control compared

    with bilirubin-or antimycin A-treated cells (Fig. 6B, images b).

    Thus, bilirubin alters mitochondrial membrane potential of the

    malaria parasite.

    Bilirubin induces an apoptosis-like phenomenon in the malaria

    parasite

    Oxidative stress may lead to the induction of apoptosis in

    many cells. In order to investigate whether bilirubin can induce

    apoptosis or pseudoapoptosis [46] or an apoptosis-likephenomenon in the malaria parasite via the development of

    oxidative stress, caspase-3-like protease (DEVDase) activity,

    expression of a putative apoptosis-related protein (Gene ID

    PFI0450c), and nuclear morphology were measured. Although

    the P. falciparum genome lacks classical caspase-3 (www.

    plasmodb.org), the cytosolic fraction of bilirubin-treated

    parasites significantly cleaved the caspase-3 substrate, DEVD-

    pNA, whereas the same fraction of control parasite showed very

    little activity (Fig. 7A). The result showed that bilirubin caused

    a sixfold activation of caspase-3-like proteases over the control

    value. Moreover, the activity of caspase-3 was significantly

    inhibited in the presence of Ac-DEVD-CHO, a potent inhibitorof caspase-3, indicating that P. falciparum contains a protease

    having caspase-3-like activity. In the P. falciparum genome, a

    gene for a putative apoptosis-related protein (PfARP) was

    predicted and annotated in chromosome 9 (Gene ID PFI0450c;

    www. plasmodb.org). RT-PCR analysis indicates that bilirubin

    causes significant up-regulation of the expression of the PfARP

    gene (Figs. 7B and C) over the control. The expression pattern

    of the seryl-tRNA synthetase (internal control) was, however,

    not affected (Figs. 7B and C). To check apoptosis-like events in

    the parasite, the nuclear morphology of the parasite was

    observed by Hoechst staining. The result indicated that bilirubin

    caused the condensation of parasite chromatin as evident from

    intense fluorescence compared to the control parasite (Fig. 8).

    Fig. 6. Effect of bilirubin on mitochondrial transmembrane potential of Plasmodium. (A) Measurement of mitochondrial transmembrane potential in control and

    bilirubin-(40M) and antimycin A-(10 M) treated trophozoite-infected red cells by spectrofluorimetry as described under Materials and methods. (B) Fluorescence

    microscopic analysis of transmembrane potential in control and bilirubin-(40 M) and antimycin A-(10 M) treated trophozoite-infected red cells. (a) J-aggregate

    formation (emission, 590 nm), (b) JC1 monomer (emission 530 nm), and (c) 590 nm/530 nm ratio (merged).

    Fig. 7. Effect of bilirubin on caspase-like (DEVDase) activity, expression of

    PfARP, and DNA damage in P. falciparum. (A) The caspase-3-like activity was

    measured in the cytosolic fraction (30g protein) frombilirubin-(40M) treated

    or control parasites. (B) RT-PCR analysis to follow the expression of PfARP. (C)

    Histogram representing the densitometric analysis of PfARP and seryl-tRNA

    synthetase expression (fold of PfARP expression relative to seryl-tRNA

    synthetase expression) in P. falciparum in the presence or absence of bilirubin.***pb0.01 vs control, **pb0.01 vs control, *pb0.01 vs bilirubin.

    608 S. Kumar et al. / Free Radical Biology & Medicine 44 (2008) 602613

    http://www.plasmodb.org/http://www.plasmodb.org/http://www.plasmodb.org/http://www.plasmodb.org/
  • 8/2/2019 Anti Ox Id Ante

    8/12

    Thus, bilirubin induces an apoptosis-like phenomenon in the

    malaria parasite.

    Effect of bilirubin on P. falciparum growth in culture

    Now the question arises whether bilirubin-induced oxidative

    stress or the associated apoptosis-like phenomenon is linked

    to parasite death? The data show that bilirubin inhibits

    P. falciparum growth in vitro in a concentration-dependent

    manner as evident from the decreased hypoxanthine uptake

    (Table 1). Bilirubin inhibits P. falciparum growth by 18 and

    80% at 10 and 80 M, respectively.

    Antioxidant treatment protects P. falciparum from

    bilirubin-induced oxidative stress and growth inhibition

    In order to assess whether the oxidative stress induced by

    bilirubin is responsible for the inhibition of parasite growth, the

    effects of antioxidants or ROS scavengers (UOH scavengers,

    such as mannitol and spin traps like PBN) were studied on

    lipid peroxidation, H2O2 generation, and P. falciparum growth.

    Results indicate that these antioxidants significantly decreased

    bilirubin-induced lipid peroxidation (Fig. 9A) and the genera-

    tion of intraparasitic H2O2 (Fig. 9B). Moreover, the well-knownUOH scavenger, mannitol, protected against the fall in JC1

    uptake (mitochondrial potential) in P. falciparum by bilirubin

    (Fig. 10A). Again, both mannitol and PBN significantly prevent

    bilirubin-induced activation of caspase-like activity (Fig. 10B)

    as well as induction of ARP expression (Figs. 10C and D). The

    expression pattern of the seryl-tRNA synthetase (internal

    control) was, however, not affected. Finally, we monitored

    parasite growth by following [3H]hypoxanthine uptake in the

    presence or absence of mannitol or caspase-3 inhibitor (DEVD-

    CHO) to test whether bilirubin-induced oxidative stress is

    mainly responsible for the growth inhibition of P. falciparum

    via the induction of apoptosis-like events. Interestingly, both

    mannitol and DEVD-CHO significantly protected P. falciparumfrom bilirubin-induced growth inhibition (Fig. 11).

    Discussion

    Evidence has been presented to show that bilirubin

    effectively induces oxidative stress, reduces mitochondrial

    membrane potential, up-regulates the expression of PfARP in

    the malaria parasite, and finally inhibits P. falciparum growth.

    The possible mechanism by which bilirubin develops

    oxidative stress and parasite death is mediated through its

    inhibitory effect on hemozoin formation, leading to excess

    accumulation of free heme in the parasite [47,48], which

    may stimulate the generation of ROS. Inhibition of heme

    Fig. 8. Effect of bilirubin on P. falciparum nuclear morphology. Hoechst

    staining was done as described under Materials and methods.

    Table 1

    Effect of bilirubin on P. falciparum growth in culture

    [3H]Hypoxanthine uptake

    (% inhibition, mean SEM)

    Control 0

    +Bilirubin (1.0 M) 4 1

    +Bilirubin (10 M) 18 2

    +Bilirubin (20 M) 40 3

    +Bilirubin (30 M) 48 5

    +Bilirubin (40 M) 65 5

    +Bilirubin (80 M) 80 8

    pb0.05 vs control.

    Fig. 9. Effects of antioxidants on bilirubin-induced oxidative stress and

    P. falciparum growth. (A) Lipid peroxide and (B) H2O2 were measured in the

    presence or absence ofU

    OH scavengers during bilirubin (40 M) treatment as

    described under Material and methods. P. falciparum culture (4% parasitemia)

    was treated with bilirubin along with PBN (40 mM) or mannitol (10 mM) for

    48 h. The data presented are meansSEM (n =6). ***pb0.001 vs control,###pb0.01 vs bilirubin.

    609S. Kumar et al. / Free Radical Biology & Medicine 44 (2008) 602613

  • 8/2/2019 Anti Ox Id Ante

    9/12

    detoxification function is known to kill the parasite through

    membrane lysis and interference of other vital functions[2,49,50]. It is already known that bilirubin can prevent heme

    crystal growth in vitro by forming complexes with heme [51].

    Moreover, different heme catabolic products and protoporphyr-

    ins are known to inhibit hemozoin formation by coordinating

    with hematin via cofacial links [52] and offer antimalarial

    activity [53]. Bilirubin may inhibit hemozoin formation by

    following the same mechanism of noniron porphyrins, but

    further studies are required to confirm it. The malaria parasite isvery much susceptible to oxidative stress [54,55] and bilirubin,

    by inhibiting hemozoin formation, may develop oxidative stress

    and inhibit parasite growth. However, bilirubin can generate

    ROS in many mammalian cells, in which mitochondria are a

    major source of ROS [11,12] and in which hemozoin formation

    is completely absent. Therefore, we cannot exclude other

    possible routes for the generation of ROS in the parasite.

    Jaundice in malaria is mostly due to hemolysis, and the range of

    total serum bilirubin is 348.2 mg% (50830 M) [56].

    Antimalarial treatment brings the elevated level of bilirubin in

    malaria to normal range [0.51.5 mg%] [57,58]. Very high

    concentrations of bilirubin formed during malaria may be pro-oxidant in nature. Although at this concentration it can damage

    various host cells, it may simultaneously protect the host from

    parasite burden by inhibiting its growth. We kept total bilirubin

    concentration to between 10 and 40 M (which allowed a

    moderate parasitemia) just to see the effect of bilirubin on

    P. falciparum growth, avoiding its nonspecific effects at a very

    high concentration.

    Our studies indicate that bilirubin-induced oxidative stress

    is associated with the reduction of m. The loss of mis considered one of the most significant events in oxidative

    stress-mediated activation of mitochondrial pathway of apop-

    tosis [11,59]. Bilirubin is known to induce apoptosis through

    the mitochondrial pathway by developing oxidative stress

    Fig. 11. Effects of antioxidant and DEVD-CHO on bilirubin-induced

    P. falciparum growth inhibition. P. falciparum culture (4% parasitemia) was

    treated with bilirubin along with mannitol (10 mM) or DEVD-CHO (100 M)

    for 48 h. P. falciparum growth was monitored by following [3H]hypoxanthine

    uptake as described under Material and methods. ***pb

    0.05 vs control,#pb0.01 vs bilirubin, ##pb0.05 vs bilirubin.

    Fig. 10. Effects of antioxidants on bilirubin-induced changes in mitochondrial potential, caspase-like activity, and expression of apoptosis-related protein (ARP).

    P. falciparum culture (4% parasitemia) was treated with bilirubin along with PBN (40 mM) or mannitol (10 mM) for 48 h. Then (A) JC1 uptake, (B) caspase-like

    activity, and (C, D) ARP expression were measured as described under Material and methods. ***pb0.01 vs control, #pb0.01 vs bilirubin, ##pb0.05 vs bilirubin.

    610 S. Kumar et al. / Free Radical Biology & Medicine 44 (2008) 602613

  • 8/2/2019 Anti Ox Id Ante

    10/12

    [11,12,59,60]. Our studies indicate that bilirubin induces an

    apoptosis-like phenomenon in P. falciparum probably by up-

    regulating the expression of PfARP and activating a caspase-

    like protease. Hoechst staining also indicates the occurrence of

    chromatin condensation in the parasite by bilirubin. The role of

    PfARP in P. falciparum apoptosis is not known; nevertheless,

    from the evidence presented it can be proposed that bilirubininduces apoptosis-like events in the parasite to cause cell death.

    Recently, by measuring the conventional parameters for

    apoptosis, it has been claimed that death caused by most of

    the antimalarials is not associated with classical apoptosis [61],

    except for atovaquone, which reduces mitochondrial membrane

    potential and respiration [45,61], vital parameters for mitochon-

    drial pathway of apoptosis. Apoptosis in any form in the malaria

    parasite is not well documented. However, it has been reported

    that chloroquine causes apoptosis in a chloroquine-sensitive

    strain of P. falciparum by DNA fragmentation [62,63].

    Caspase-like activity has also been observed in Plasmodium

    berghei ookinetes, which results in the apoptosis-like death ofmore than 50% of the parasites at this stage in the mosquito

    midgut [64]. The parasite i tself dies by a process of

    programmed cell death in the lumen of the midgut before

    invasion has occurred. Caspase-like activity was detected in the

    cytoplasm of the ookinetes, despite the absence of genes

    homologous to caspases in the genome of Plasmodium or any

    unicellular eukaryote [65]. Interestingly, P. falciparum contains

    metacaspase (Gene ID PF14_0363) and various cysteine

    proteases (PFB0325c, PFB0330c, PFB0335c, PFB0340c,

    PFB0345c). Oxidative stress is known to induce apoptosis in

    the unicellular parasite Leishmania donovani, which lacks

    classical caspases and in which the activation of caspase-3-like

    proteases has been reported [66]. However, further studies arenecessary to confirm whether the observed caspase activity

    originates from metacaspase (Gene ID PF14_0363) or from any

    cysteine protease. In the P. falciparum genome, a putative gene

    for PfARP (Gene ID PFI0450c) is annotated on chromosome 9

    (www.plasmodb.org). PfARP has the highest degree of

    homology to mammalian TF-1 cell apoptosis-related gene-19

    (TFAR-19). The growth factor or serum deprivation leads to

    overexpression of TFAR-19 in TF-1 cells, which results in

    apoptosis in those cells. Moreover, PfARP also has a close

    resemblance to PDCD5, which has been reported to have a

    regulatory role in paraptosis, a form of programmed cell death

    distinct from apoptosis [67]. Similarity search results pointtoward diverse possibilities for the functional roles of PfARP in

    P. falciparum, such as some role in classical apoptosis or in

    nonclassical programmed cell death. Interestingly, bilirubin up-

    regulates the expression of PfARP in P. falciparum. But it

    remains to be established whether this protein functions as a

    helper in the death process. If bilirubin inhibits P. falciparum

    growth through the induction of oxidative stress, antioxidant

    treatment should protect the parasite from bilirubin-induced cell

    death. UOH scavengers and spin traps remarkably inhibited

    bilirubin-induced oxidative stress in the parasite and both

    antioxidant and caspase inhibitor (DEVD-CHO) significantly

    protected P. falciparum from bilirubin-induced growth inhibi-

    tion. This suggests that bilirubin-induced oxidative stress is

    responsible for the inhibition ofP. falciparum growth. We thus

    conclude that bilirubin induces oxidative stress, which stimu-

    lates an apoptosis-like death in the malaria parasite.

    It can be suggested that the increased formation of bilirubin

    by the host during malaria, due to hemolysis, may aggravate the

    oxidative damage to the host red cells and neurons, but may also

    be protective, in part, by enhancing the oxidative destruction ofthe parasites. Moreover, this study provides a logical explana-

    tion for why this parasite does not have an HO-1 system. As

    bilirubin is toxic to the malaria parasite, it lacks a conventional

    HO-1 system to protect itself from bilirubin-induced cell death

    as a second line of defense.

    Acknowledgment

    Sanjay Kumar gratefully acknowledges the Council of

    Scientific and Industrial Research, New Delhi, for providing a

    Senior Research Fellowship to carry out this work and

    providing fund from Suprainstitutional project (SIP0007).

    References

    [1] Rosenthal, P. J.; Meshnick, S. R. Hemoglobin processing and the metabo-

    lism of amino acids, heme and iron. In: Sherman, I.W., ed. Malaria:

    parasite biology, pathogenesis and protection. Washington, DC: ASM

    Press; 1998:145158

    [2] Tekwani, B. L.; Walker, L. A. Targeting the hemozoin synthesis pathway

    for new antimalarial drug discovery: technologies for in vitro beta-hematin

    formation assay. Comb. Chem. High Throughput Screen 8:6379; 2005.

    [3] Maines, M. D. Heme oxygenase: function, multiplicity, regulatory

    mechanisms, and clinical applications. FASEB J. 2:25572568; 1988.

    [4] Docherty, J. C.; Firneisz, G. D.; Schacter, B. A. Methene bridge carbon

    atom elimination in oxidative heme degradation catalyzed by heme

    oxygenase and NADPH-cytochrome P-450 reductase. Arch. Biochem.

    Biophys. 235:657664; 1984.

    [5] Tenhunen, R.; Marver, H. S.; Schmid, R. Microsomal heme oxygenase:

    characterization of the enzyme. J. Biol. Chem. 244:63886394; 1969.

    [6] Asad, S. F.; Singh, S.;Ahmad, A.; Khan, N. U.; Hadi, S. M. Prooxidant and

    antioxidant activities of bilirubin and its metabolic precursor biliverdin: a

    structure-activity study. Chem. Biol. Interact. 137:5974; 2001.

    [7] Stocker, R.; Yamamoto, Y.; McDonagh, A. F.; Glazer, A. N.; Ames, B. N.

    Bilirubin is an antioxidant of possible physiological importance. Science

    235:10431046; 1987.

    [8] Clark, J. E.; Foresti, R.; Green, C. J.; Motterlini, R. Dynamics of haem

    oxygenase-1 expression and bilirubin production in cellular protection

    against oxidative stress. Biochem. J. 348 (Pt 3):615619; 2000.

    [9] Minetti, M.; Mallozzi, C.; Di Stasi, A. M.; Pietraforte, D. Bilirubin is an

    effective antioxidant of peroxynitrite-mediated protein oxidation in human

    blood plasma. Arch. Biochem. Biophys. 352:165174; 1998.

    [10] Tomaro, M. L.; Batlle, A. M. Bilirubin: its role in cytoprotection against

    oxidative stress. Int. J. Biochem. Cell Biol. 34:216220; 2002.

    [11] Oakes, G. H.; Bend, J. R. Early steps in bilirubin-mediated apoptosis in

    murine hepatoma (Hepa 1c1c7) cells are characterized by aryl hydrocarbon

    receptor-independent oxidative stress and activation of the mitochondrial

    pathway. J. Biochem. Mol. Toxicol. 19:244255; 2005.

    [12] Seubert, J. M.; Darmon, A. J.; El-Kadi, A. O.; D'Souza, S. J.; Bend, J. R.

    Apoptosis in murine hepatoma hepa 1c1c7 wild-type, C12, and C4 cells

    mediated by bilirubin. Mol. Pharmacol. 62:257264; 2002.

    [13] Park, W. S.; Chang, Y. S.; Lee, M. Effect of 7-nitroindazole on bilirubin-

    induced changes in brain cell membrane function and energy metabolism

    in newborn piglets. Biol. Neonate 82:6165; 2002.

    [14] Simon, H. U.; Haj-Yehia, A.; Levi-Schaffer, F. Role of reactive oxygenspecies (ROS) in apoptosis induction. Apoptosis 5:415418; 2000.

    611S. Kumar et al. / Free Radical Biology & Medicine 44 (2008) 602613

    http://www.plasmodb.org/http://www.plasmodb.org/
  • 8/2/2019 Anti Ox Id Ante

    11/12

    [15] Guha, M.; Kumar, S.; Choubey, V.; Maity, P.; Bandyopadhyay, U.

    Apoptosis in liver during malaria: role of oxidative stress and implication

    of mitochondrial pathway. FASEB J. 20:12241226; 2006.

    [16] Ostrow, J. D.; Pascolo, L.; Tiribelli, C. Reassessment of the unbound

    concentrations of unconjugated bilirubin in relation to neurotoxicity in

    vitro. Pediatr. Res. 54:98104; 2003.

    [17] Liu, X. M.; Chapman, G. B.; Wang, H.; Durante, W. Adenovirus-mediated

    heme oxygenase-1 gene expression stimulates apoptosis in vascularsmooth muscle cells. Circulation 105:7984; 2002.

    [18] Akin, E.; Clower, B.; Tibbs, R.; Tang, J.; Zhang, J. Bilirubin produces

    apoptosis in cultured bovine brain endothelial cells. Brain Res.

    931:168175; 2002.

    [19] McDonagh, A. F.; Assisi, F. The ready isomerization of bilirubin IXIn

    aqueous solution. Biochem. J. 129:797800; 1972.

    [20] Trager, W.; Jensen, J. B. Human malaria parasites in continuous culture.

    Science 193:673675; 1976.

    [21] Glushakova, S.; Yin, D.; Gartner, N.; Zimmerberg, J. Quantification of

    malaria parasite release from infected erythrocytes: inhibition by protein-

    free media. Malar. J. 6:61; 2007.

    [22] Hoppe, H. C.; van Schalkwyk, D. A.; Wiehart, U. I.; Meredith, S. A.; Egan,

    J.; Weber, B. W. Antimalarial quinolines and artemisinin inhibit endo-

    cytosis in Plasmodium falciparum. Antimicrob. Agents Chemother.

    48:23702378; 2004.[23] Roca, L.; Calligaris, S.; Wennberg, R. P.; Ahlfors, C. E.; Malik, S. G.;

    Ostrow, J. D.; Tiribelli, C. Factors affecting the binding of bilirubin to

    serum albumins: validation and application of the peroxidase method.

    Pediatr. Res. 60:724728; 2006.

    [24] Choubey, V.; Guha, M.; Maity, P.; Kumar, S.; Raghunandan, R.; Maulik,

    P. R.; Mitra, K.; Halder, U. C.; Bandyopadhyay, U. Molecular charac-

    terization and localization of Plasmodium falciparum choline kinase.

    Biochim. Biophys. Acta 1760:10271038; 2006.

    [25] Lowry, O. H.; Rosebrough, N. J.; Farr, A. L.; Randall, R. J. Protein

    measurement with the Folin phenol reagent. J. Biol. Chem. 193:265275;

    1951.

    [26] Buege, J. A.; Aust, S. D. Microsomal lipid peroxidation. Methods

    Enzymol. 52:302310; 1978.

    [27] Biswas, K.; Bandyopadhyay, U.; Chattopadhyay, I.; Varadaraj, A.; Ali, E.;

    Banerjee, R. K. A novel antioxidant and antiapoptotic role of omeprazoleto block gastric ulcer through scavenging of hydroxyl radical. J. Biol.

    Chem. 278:1099311001; 2003.

    [28] Chattopadhyay, I.; Bandyopadhyay, U.; Biswas, K.; Maity, P.; Banerjee,

    R. K. Indomethacin inactivates gastric peroxidase to induce reactive-

    oxygen-mediated gastric mucosal injury and curcumin protects it by

    preventing peroxidase inactivation and scavenging reactive oxygen. Free

    Radic. Biol. Med. 40:13971408; 2006.

    [29] Trivedi, V.; Chand, P.; Srivastava, K.; Puri, S. K.; Maulik, P. R.;

    Bandyopadhyay, U. Clotrimazole inhibits hemoperoxidase of Plasmodium

    falciparum and induces oxidative stress: proposed antimalarial mechanism

    of clotrimazole. J. Biol. Chem. 280:4112941136; 2005.

    [30] Babbs, C. F.; Steiner, M. G. Detection and quantitation of hydroxyl radical

    using dimethyl sulfoxide as molecular probe. Methods Enzymol.

    186:137147; 1990.

    [31] Trivedi, V.; Chand, P.; Maulik, P. R.; Bandyopadhyay, U. Mechanism ofhorseradish peroxidase-catalyzed heme oxidation and polymerization

    (beta-hematin formation). Biochim. Biophys. Acta 1723:221228; 2005.

    [32] Sullivan, D. J., Jr.; Gluzman, I. Y.; Goldberg, D. E. Plasmodium hemozoin

    formation mediated by histidine-rich proteins. Science 271:219222;

    1996.

    [33] Pandey, A. V.; Singh, N.; Tekwani, B. L.; Puri, S. K.; Chauhan, V. S. Assay

    of beta-hematin formation by malaria parasite. J. Pharm. Biomed. Anal.

    20:203207; 1999.

    [34] Coban, C.; Ishii, K. J.; Sullivan, D. J.; Kumar, N. Purified malaria pigment

    (hemozoin) enhances dendritic cell maturation and modulates the isotype

    of antibodies induced by a DNA vaccine. Infect. Immun. 70:39393943;

    2002.

    [35] Motterlini, R.; Foresti, R.; Vandegriff, K.; Intaglietta, M.; Winslow, R. M.

    Oxidative-stress response in vascular endothelial cells exposed to acellular

    hemoglobin solutions. Am. J. Physiol. 269:H648H655; 1995.

    [36] Solomon, V. R.; Puri, S. K.; Srivastava, K.; Katti, S. B. Design and

    synthesis of new antimalarial agents from 4-aminoquinoline. Bioorg. Med.

    Chem. 13:21572165; 2005.

    [37] Goldman, I. F.; Qari, S. H.; Skinner, J.; Oliveira, S.; Nascimento, J. M.;

    Povoa, M. M.; Collins, W. E.; Lal, A. A. Use of glass beads and CF 11

    cellulose for removal of leukocytes from malaria-infected human blood

    in field settings. Mem. Inst. Oswaldo Cruz 87:583587; 1992.

    [38] McNally, J.; O'Donovan, S. M.; Dalton, J. P. Plasmodium berghei andPlasmodium chabaudi chabaudi: development of simple in vitro eryth-

    rocyte invasion assays. Parasitology 105 (Pt 3):355362; 1992.

    [39] Cossarizza, A.; Baccarani-Contri, M.; Kalashnikova, G.; Franceschi, C. A

    new method for the cytofluorimetric analysis of mitochondrial membrane

    potential using the J-aggregate forming lipophilic cation 5,5,6,6-tetra-

    chloro-1,1,3,3-tetraethylbenzimidazolcarbocyanine iodide (JC-1). Bio-

    chem. Biophys. Res. Commun. 197:4045; 1993.

    [40] Guha, M.; Choubey, V.; Maity, P.; Kumar, S.; Shrivastava, K.; Puri, S. K.;

    Bandyopadhyay, U. Overexpression, purification, and localization of

    apoptosis-related protein from Plasmodium falciparum. Protein Expr.

    Purif. 52:363372; 2007.

    [41] Ben Mamoun, C.; Gluzman, I. Y.; Hott, C.; MacMillan, S. K.; Amarakone,

    A. S.; Anderson, D. L.; Carlton, J. M.; Dame, J. B.; Chakrabarti, D.;

    Martin, R. K.; Brownstein, B. H.; Goldberg, D. E. Co-ordinated

    programme of gene expression during asexual intraerythrocytic develop-ment of the human malaria parasite Plasmodium falciparum revealed by

    microarray analysis. Mol. Microbiol. 39:2636; 2001.

    [42] Desjardins, R. E.; Canfield, C. J.; Haynes, J. D.; Chulay, J. D. Quantitative

    assessment of antimalarial activity in vitro by a semiautomated micro-

    dilution technique. Antimicrob. Agents Chemother. 16:710718; 1979.

    [43] Lambros, C.; Vanderberg, J. P. Synchronization of Plasmodium falciparum

    erythrocytic stages in culture. J. Parasitol. 65:418420; 1979.

    [44] Begriche, K.; Igoudjil, A.; Pessayre, D.; Fromenty, B. Mitochondrial dys-

    function in NASH: causes, consequences and possible means to prevent it.

    Mitochondrion 6:128; 2006.

    [45] Srivastava, I. K.; Vaidya, A. B. A mechanism for the synergistic anti-

    malarialaction of atovaquone and proguanil.Antimicrob.Agents Chemother.

    43:13341339; 1999.

    [46] Mackenzie, A. B.; Young, M. T.; Adinolfi, E.; Surprenant, A. Pseudo-

    apoptosis induced by brief activation of ATP-gated P2X7 receptors.J. Biol. Chem. 280:3396833976; 2005.

    [47] Zhang, J.; Krugliak, M.; Ginsburg, H. The fate of ferriprotoporphyrin IX

    in malaria infected erythrocytes in conjunction with the mode of action

    of antimalarial drugs. Mol. Biochem. Parasitol. 99:129141; 1999.

    [48] Famin, O.; Ginsburg, H. Differential effects of 4-aminoquinoline-contain-

    ing antimalarial drugs on hemoglobin digestion in Plasmodium falci-

    parum-infected erythrocytes. Biochem. Pharmacol. 63:393398; 2002.

    [49] Francis, S. E.; Sullivan, D. J., Jr.; Goldberg, D. E. Hemoglobin metabolism

    in the malaria parasite Plasmodium falciparum. Annu. Rev. Microbiol.

    51:97123; 1997.

    [50] Orjih, A. U.; Banyal, H. S.; Chevli, R.; Fitch, C. D. Hemin lyses malaria

    parasites. Science 214:667669; 1981.

    [51] Chong, C. R.; Sullivan, D. J., Jr. Inhibition of heme crystal growth

    by antimalarials and other compounds: implications for drug discovery.

    Biochem. Pharmacol. 66:22012212; 2003.[52] Basilico, N.; Monti, D.; Olliaro, P.; Taramelli, D. Non-iron porphyrins

    inhibit beta-haematin (malaria pigment) polymerisation. FEBS Lett.

    409:297299; 1997.

    [53] Begum, K.; Kim, H. S.; Kumar, V.; Stojiljkovic, I.; Wataya, Y. In vitro

    antimalarial activity of metalloporphyrins against Plasmodium falciparum.

    Parasitol. Res. 90:221224; 2003.

    [54] Friedman, M. J. Oxidant damage mediates variant red cell resistance to

    malaria. Nature 280:245247; 1979.

    [55] Clark, I. A.; Hunt, N. H. Evidence for reactive oxygen intermediates

    causing hemolysis and parasite death in malaria. Infect. Immun. 39:16;

    1983.

    [56] Kochar, D. K.; Agarwal, P.; Kochar, S. K.; Jain, R.; Rawat, N.; Pokharna,

    R. K.; Kachhawa, S.; Srivastava, T. Hepatocyte dysfunction and hepatic

    encephalopathy in Plasmodium falciparum malaria. QJM 96:505512;

    2003.

    612 S. Kumar et al. / Free Radical Biology & Medicine 44 (2008) 602613

  • 8/2/2019 Anti Ox Id Ante

    12/12

    [57] Mandal, P. K.; Sarkar, N.; Pal, A. Efficacy of arteether in chloroquine

    resistant falciparum malaria in eastern India. Indian J. Med. Res.

    119:2832; 2004.

    [58] Tangpukdee, N.; Thanachartwet, V.; Krudsood, S.; Luplertlop, N.;

    Pornpininworakij, K.; Chalermrut, K.; Phokham, S.; Kano, S.; Looar-

    eesuwan, S.; Wilairatana, P. Minor liver profile dysfunctions in Plas-

    modium vivax, P. malaria and P. ovale patients and normalization after

    treatment. Korean J. Parasitol. 44:295302; 2006.[59] Rodrigues, C. M.; Sola, S.; Brites, D. Bilirubin induces apoptosis via the

    mitochondrial pathway in developing rat brain neurons. Hepatology

    35:11861195; 2002.

    [60] Keshavan, P.; Schwemberger, S. J.; Smith, D. L.; Babcock, G. F.; Zucker,

    S. D. Unconjugated bilirubin induces apoptosis in colon cancer cells

    by triggering mitochondrial depolarization. Int. J. Cancer 112:433445;

    2004.

    [61] Nyakeriga, A. M.; Perlmann, H.; Hagstedt, M.; Berzins, K.; Troye-

    Blomberg, M.; Zhivotovsky, B.; Perlmann, P.; Grandien, A. Drug-induced

    death of the asexual blood stages of Plasmodium falciparum occurs

    without typical signs of apoptosis. Microbes Infect. 8:15601568; 2006.

    [62] Picot, S.; Burnod, J.; Bracchi, V.; Chumpitazi, B. F.; Ambroise-Thomas,

    P. Apoptosis related to chloroquine sensitivity of the human malaria

    parasite Plasmodium falciparum. Trans. R. Soc. Trop. Med. Hyg.

    91:590591; 1997.

    [63] Meslin, B.; Barnadas, C.; Boni, V.; Latour, C.; De Monbrison, F.; Kaiser,

    K.; Picot, S. Features of apoptosis in Plasmodium falciparum erythrocytic

    stage through a putative role of PfMCA1 metacaspase-like protein.

    J. Infect. Dis. 195:18521859; 2007.

    [64] Al-Olayan, E. M.; Williams, G. T.; Hurd, H. Apoptosis in the malaria

    protozoan, Plasmodium berghei: a possible mechanism for limitingintensity of infection in the mosquito. Int. J. Parasitol. 32:11331143;

    2002.

    [65] Hurd, H.; Carter, V.; Nacer, A. Interactions between malaria and mos-

    quitoes: the role of apoptosis in parasite establishment and vector response

    to infection. Curr. Top. Microbiol. Immunol. 289:185217; 2005.

    [66] Sen, N.; Das, B. B.; Ganguly, A.; Mukherjee, T.; Bandyopadhyay, S.;

    Majumder, H. K. Camptothecin-induced imbalance in intracellular cation

    homeostasis regulates programmed cell death in unicellular hemoflagellate

    Leishmania donovani. J. Biol. Chem. 279:5236652375; 2004.

    [67] Uren, A. G.; O'Rourke, K.; Aravind, L. A.; Pisabarro, M. T.; Seshagiri, S.;

    Koonin, E. V.; Dixit, V. M. Identification of paracaspases and meta-

    caspases: two ancient families of caspase-like proteins, one of which plays

    a key role in MALT lymphoma. Mol. Cell 6:961967; 2000.

    613S. Kumar et al. / Free Radical Biology & Medicine 44 (2008) 602613


Recommended