Overexpression of Mitochondrial Leishmania major Ascorbate Peroxidase … · Ascorbate peroxidase...

11
EUKARYOTIC CELL, Nov. 2009, p. 1721–1731 Vol. 8, No. 11 1535-9778/09/$12.00 doi:10.1128/EC.00198-09 Copyright © 2009, American Society for Microbiology. All Rights Reserved. Overexpression of Mitochondrial Leishmania major Ascorbate Peroxidase Enhances Tolerance to Oxidative Stress-Induced Programmed Cell Death and Protein Damage Subhankar Dolai, Rajesh K. Yadav, Swati Pal, and Subrata Adak* Division of Structural Biology & Bio-informatics, Indian Institute of Chemical Biology, Council of Scientific & Industrial Research, 4, Raja S.C. Mullick Road, Kolkata 700 032, India Received 3 July 2009/Accepted 31 August 2009 Ascorbate peroxidase from Leishmania major (LmAPX) is one of the key enzymes for scavenging of reactive oxygen species generated from the mitochondrial respiratory chain. We have investigated whether mitochon- drial LmAPX has any role in oxidative stress-induced apoptosis. The measurement of reduced glutathione (GSH) and protein carbonyl contents in cellular homogenates indicates that overexpression of LmAPX protects Leishmania cells against depletion of GSH and oxidative damage of proteins by H 2 O 2 or camptothecin (CPT) treatment. Confocal microscopy and fluorescence spectroscopy data have revealed that the intracellular elevation of Ca 2 attained by the LmAPX-overexpressing cells was always below that attained in control cells. Flow cytometry assay data and confocal microscopy observation strongly suggest that LmAPX overexpression protects cells from H 2 O 2 -induced mitochondrial membrane depolarization as well as ATP decrease. Western blot data suggest that overexpression of LmAPX shields against H 2 O 2 - or CPT-induced cytochrome c and endonuclease G release from mitochondria and subsequently their accumulation in the cytoplasm. Caspase activity assay by flow cytometry shows a lower level of caspase-like protease activity in LmAPX-overexpressing cells under apoptotic stimuli. The data on phosphatidylserine exposed on the cell surface and DNA fragmen- tation results show that overexpression of LmAPX renders the Leishmania cells more resistant to apoptosis provoked by H 2 O 2 or CPT treatment. Taken together, these results indicate that constitutive overexpression of LmAPX in the mitochondria of L. major prevents cells from the deleterious effects of oxidative stress, that is, mitochondrial dysfunction and cellular death. In multicellular organisms, mitochondria are the major physiological source of reactive oxygen species (ROS) within cells and also are important checkpoints for the control of programmed cell death (27). There are increasing numbers of reports that describe apoptosis- or programmed cell death-like processes in unicellular organisms also, such as trypanosoma- tids (4, 60), bacteria (20, 25), yeasts (34), and Plasmodium (3). Among the kinetoplastid parasites, Trypanosoma and Leishma- nia are the most carefully studied genera where apoptotic features are well established (49). Several reports have shown that mitochondrial dysfunction or an imbalance of antioxidant homeostasis causes an increase in mitochondrion-generated ROS, which include H 2 O 2 , superoxide radical anions, singlet oxygen, and hydroxyl radicals. These species have all been implicated in apoptosis (16, 26, 28, 41). Increasing evidence has been presented to support that ROS homeostasis regulates two major types of important physiological processes and ex- erts diverse functions within cells. One type of function in- cludes damage or oxidation of cellular macromolecules (DNA, proteins, and lipids), which can lead to necrotic cell death or protein modification (7). The second type of function includes the activation of cellular signaling cascades that regulate pro- liferation, detoxification, DNA repair, or apoptosis (11). The detoxification of toxic mitochondrial ROS in cells occurs through a variety of cellular antioxidant enzymes, such as su- peroxide dismutase, which detoxifies cells from superoxide re- leased into the mitochondrial matrix, and several other anti- oxidant proteins, such as catalase, glutathione (GSH) peroxidase, and peroxiredoxins, which are known to catalyze further deg- radation of H 2 O 2 (44). During its life cycle, the Leishmania sp. encounters a pool of ROS that is generated either by its own physiological processes or as a result of host immune reaction and drug metabolism. However, unlike most eukaryotes, Leish- mania lacks catalase- and selenium-containing GSH peroxi- dases, enzymes that play a front-line role in detoxifying ROS. Hence, the mechanism by which it resists the toxic effects of H 2 O 2 remains poorly understood. Recently, we cloned, expressed and characterized the un- usual heme-containing ascorbate peroxidase from Leishmania major (LmAPX) and observed that the expression of LmAPX is increased when Leishmania cells are treated with exogenous H 2 O 2 (1, 18). This enzyme is a functional hybrid between cytochrome c peroxidase and APX, owing to its ability to use both ascorbate and cytochrome c as reducing electron donors (58). Colocalization studies by confocal microscopy, submito- chondrial fractionation analysis of the isolated mitochondria, and subsequent Western blot analysis with anti-LmAPX anti- body have confirmed that the mature enzyme is present in intermembrane space side of the inner membrane. It has also been shown that overexpression of LmAPX causes a decrease in the mitochondrial ROS burden, an increase in tolerance to * Corresponding author. Mailing address: Division of Structural Bi- ology & Bio-informatics, Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata 700 032, India. Phone: 91 33 2473-6793. Fax: 91 33 2473-5197. E-mail: [email protected]. † Supplemental material for this article may be found at http://ec .asm.org/. Published ahead of print on 11 September 2009. 1721 on February 8, 2021 by guest http://ec.asm.org/ Downloaded from

Transcript of Overexpression of Mitochondrial Leishmania major Ascorbate Peroxidase … · Ascorbate peroxidase...

Page 1: Overexpression of Mitochondrial Leishmania major Ascorbate Peroxidase … · Ascorbate peroxidase from Leishmania major (LmAPX) is one of the key enzymes for scavenging of reactive

EUKARYOTIC CELL, Nov. 2009, p. 1721–1731 Vol. 8, No. 111535-9778/09/$12.00 doi:10.1128/EC.00198-09Copyright © 2009, American Society for Microbiology. All Rights Reserved.

Overexpression of Mitochondrial Leishmania major AscorbatePeroxidase Enhances Tolerance to Oxidative Stress-Induced

Programmed Cell Death and Protein Damage�†Subhankar Dolai, Rajesh K. Yadav, Swati Pal, and Subrata Adak*

Division of Structural Biology & Bio-informatics, Indian Institute of Chemical Biology, Council of Scientific & Industrial Research,4, Raja S.C. Mullick Road, Kolkata 700 032, India

Received 3 July 2009/Accepted 31 August 2009

Ascorbate peroxidase from Leishmania major (LmAPX) is one of the key enzymes for scavenging of reactiveoxygen species generated from the mitochondrial respiratory chain. We have investigated whether mitochon-drial LmAPX has any role in oxidative stress-induced apoptosis. The measurement of reduced glutathione(GSH) and protein carbonyl contents in cellular homogenates indicates that overexpression of LmAPXprotects Leishmania cells against depletion of GSH and oxidative damage of proteins by H2O2 or camptothecin(CPT) treatment. Confocal microscopy and fluorescence spectroscopy data have revealed that the intracellularelevation of Ca2� attained by the LmAPX-overexpressing cells was always below that attained in control cells.Flow cytometry assay data and confocal microscopy observation strongly suggest that LmAPX overexpressionprotects cells from H2O2-induced mitochondrial membrane depolarization as well as ATP decrease. Westernblot data suggest that overexpression of LmAPX shields against H2O2- or CPT-induced cytochrome c andendonuclease G release from mitochondria and subsequently their accumulation in the cytoplasm. Caspaseactivity assay by flow cytometry shows a lower level of caspase-like protease activity in LmAPX-overexpressingcells under apoptotic stimuli. The data on phosphatidylserine exposed on the cell surface and DNA fragmen-tation results show that overexpression of LmAPX renders the Leishmania cells more resistant to apoptosisprovoked by H2O2 or CPT treatment. Taken together, these results indicate that constitutive overexpression ofLmAPX in the mitochondria of L. major prevents cells from the deleterious effects of oxidative stress, that is,mitochondrial dysfunction and cellular death.

In multicellular organisms, mitochondria are the majorphysiological source of reactive oxygen species (ROS) withincells and also are important checkpoints for the control ofprogrammed cell death (27). There are increasing numbers ofreports that describe apoptosis- or programmed cell death-likeprocesses in unicellular organisms also, such as trypanosoma-tids (4, 60), bacteria (20, 25), yeasts (34), and Plasmodium (3).Among the kinetoplastid parasites, Trypanosoma and Leishma-nia are the most carefully studied genera where apoptoticfeatures are well established (49). Several reports have shownthat mitochondrial dysfunction or an imbalance of antioxidanthomeostasis causes an increase in mitochondrion-generatedROS, which include H2O2, superoxide radical anions, singletoxygen, and hydroxyl radicals. These species have all beenimplicated in apoptosis (16, 26, 28, 41). Increasing evidencehas been presented to support that ROS homeostasis regulatestwo major types of important physiological processes and ex-erts diverse functions within cells. One type of function in-cludes damage or oxidation of cellular macromolecules (DNA,proteins, and lipids), which can lead to necrotic cell death orprotein modification (7). The second type of function includesthe activation of cellular signaling cascades that regulate pro-

liferation, detoxification, DNA repair, or apoptosis (11). Thedetoxification of toxic mitochondrial ROS in cells occursthrough a variety of cellular antioxidant enzymes, such as su-peroxide dismutase, which detoxifies cells from superoxide re-leased into the mitochondrial matrix, and several other anti-oxidant proteins, such as catalase, glutathione (GSH) peroxidase,and peroxiredoxins, which are known to catalyze further deg-radation of H2O2 (44). During its life cycle, the Leishmania sp.encounters a pool of ROS that is generated either by its ownphysiological processes or as a result of host immune reactionand drug metabolism. However, unlike most eukaryotes, Leish-mania lacks catalase- and selenium-containing GSH peroxi-dases, enzymes that play a front-line role in detoxifying ROS.Hence, the mechanism by which it resists the toxic effects ofH2O2 remains poorly understood.

Recently, we cloned, expressed and characterized the un-usual heme-containing ascorbate peroxidase from Leishmaniamajor (LmAPX) and observed that the expression of LmAPXis increased when Leishmania cells are treated with exogenousH2O2 (1, 18). This enzyme is a functional hybrid betweencytochrome c peroxidase and APX, owing to its ability to useboth ascorbate and cytochrome c as reducing electron donors(58). Colocalization studies by confocal microscopy, submito-chondrial fractionation analysis of the isolated mitochondria,and subsequent Western blot analysis with anti-LmAPX anti-body have confirmed that the mature enzyme is present inintermembrane space side of the inner membrane. It has alsobeen shown that overexpression of LmAPX causes a decreasein the mitochondrial ROS burden, an increase in tolerance to

* Corresponding author. Mailing address: Division of Structural Bi-ology & Bio-informatics, Indian Institute of Chemical Biology, 4, RajaS.C. Mullick Road, Kolkata 700 032, India. Phone: 91 33 2473-6793.Fax: 91 33 2473-5197. E-mail: [email protected].

† Supplemental material for this article may be found at http://ec.asm.org/.

� Published ahead of print on 11 September 2009.

1721

on February 8, 2021 by guest

http://ec.asm.org/

Dow

nloaded from

Page 2: Overexpression of Mitochondrial Leishmania major Ascorbate Peroxidase … · Ascorbate peroxidase from Leishmania major (LmAPX) is one of the key enzymes for scavenging of reactive

H2O2, and protection against cardiolipin oxidation under oxi-dative stress (18). Although previous studies have shown thatLeishmania species use superoxide dismutase (23), peroxire-doxins (8), intracellular thiols (14), lipophosphoglycan (13),trypanothione (5), HSP 70 (a heat shock protein) (36), trypare-doxin peroxidase (29), and APX (18) for detoxification ofROS, it is still unclear how the antioxidants protect againstoxidative stress-induced apoptotic events in the unicellular or-ganism Leishmania.

Since the LmAPX protein is localized in the mitochondria,we hypothesized that it would be a key protein for the main-tenance of mitochondrial functions due to its antioxidant prop-erties via its ROS-scavenging function (18). To test this hy-pothesis, we overexpressed LmAPX in Leishmania major cellsand investigated whether overexpression of LmAPX can con-fer resistance to oxidant-mediated mitochondrial damage aswell as oxidative stress-induced cell death. In this study, weprovide evidence that the overexpression of LmAPX in Leish-mania cells can indeed protect against camptothecin (CPT) orH2O2-mediated mitochondrial damage as measured by variousparameters, including disruption of mitochondrial membranepotential (��m), decrease of ATP production, and cytochromec and endonuclease G release from mitochondria. Cells over-expressing LmAPX were also protected against oxidativestress-induced protein carbonylation, DNA fragmentation, andapoptosis. To the best of our knowledge, this is the first reportof a mitochondrial hemeperoxidase that controls the ROS-induced mitochondrial death pathway.

MATERIALS AND METHODS

Materials. The Mitoprobe JC-1 assay kit for flow cytometry, Vibrant apoptosisassay kit no. 3 for annexin V and propidium iodide (PI), caspase 3 and 7 assay kit,Fluo 4-AM, Pluronic F127, calcium ionophore A23187, ionomycin, and fetalbovine serum were purchased from Molecular Probes (Eugene, OR). The GSHdetection kit, caspase inhibitor VAD-fmk (Val-Ala-Asp-fluoromethyl ketone),and terminal deoxynucleotidyltransferase-mediated dUTP nick-end labeling(TUNEL) kit were procured from Clontech. The Oxyblot protein oxidationdetection kit was from Millipore. CPT and all other chemicals were obtainedfrom Sigma or from sources mentioned previously (1, 18).

Parasite culture and treatments. The promastigote form of Leishmania major(5ASKH) was cultured in M199 medium supplemented with 10% heat-inacti-vated fetal bovine serum as described previously (1, 18). pXG B2863 vector alone(control) or pXG B2863 vector with LmAPX gene-transfected L. major cells wasmaintained in the presence of 200 �g/ml G418. Ectopic expression of LmAPXwas monitored regularly by Western blotting and activity assay. Western blotexperiments and flow cytometry assays (using the fluorogenic peroxidase sub-strate dihydrorhodamine 123) showed at least 4-fold- and 2.5-fold-higherLmAPX expression and peroxidase activity in the LmAPX-overexpressing par-asites compared to control cells (data not shown). For experimental purposes,3-day-old exponentially growing cultures that contained almost 100% motilepromastigotes were used. Control and LmAPX-overexpressing cells were treatedwith either 1 mM H2O2 or 5 �M CPT to induce apoptosis.

Viability assay in the presence of various concentration of H2O2. Leishmaniamajor promastigote viability during oxidative stress was measured by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) (Sigma) assay asdescribed by Gantt et al. with minor modifications (22). Exponentially growingpromastigotes (2 � 106) in M199 medium were exposed to different concentra-tions of H2O2 for 2.0 to 8.0 h. After treatment, cells were washed with ice-cold1� phosphate-buffered saline (PBS) and incubated in fresh M199 medium with10% heat-inactivated fetal bovine serum with 0.5 mg/ml MTT for 3 h. After 3 h,cells were pelleted by centrifugation (1,200 � g for 5.0 min) and washed twicewith 1� PBS, and 100 �l of 0.04 N HCl in isopropanol was added. The dark blueformazan generated from MTT by living mitochondria is soluble in acid-isopro-panol. The absorbance of these solutions was measured on a microplate readerat 570 nm. The percentage of viability was calculated from optical density read-ings in wells with H2O2 compared with those in wells without. All experiments

were performed in triplicate. The viability assay showed that 90% cellular deathoccurred in Leishmania major promastigotes as a result of treatment for 8.0 hwith 1.0 mM H2O2 (see Fig. S1 in the supplemental material).

Measurement of GSH. The cellular GSH content was measured with a GSHdetection kit (Clontech). After treatment with H2O2 or CPT, 107 cells werepelleted and lysed with 1� lysis buffer. Cell lysates were then incubated withmonochlorobimane dye (2 mM) for 1 hour at 37°C, and the GSH level wasdetected with a fluorescence plate reader at 395-nm excitation and 480-nmemission wavelengths.

Measurement of free cytosolic calcium. Cytosolic Ca2� in cells was monitoredusing Fluo 4AM (Molecular Probes, Eugene, OR). A total of 107 cells wereloaded with 5 �M Fluo-4/AM for 60 min at room temperature in the presence of10 �M pluronic acid F127. After incubation, cells were washed with fresh serum-free medium and analyzed immediately with a fluorescence spectrophotometerwith excitation at 490 nm and emission at 518 nm. The concentration of freeCa2� was calculated using the formula Kd (F � FMIN)/(FMAX � F), where Kd is345 nM, F is the fluorescence intensity of the cells, FMIN is the minimumfluorescence of the cells obtained by treating cells with 10 �M calcium ionophorein the presence of 3 mM EGTA, and FMAX is maximum fluorescence of cellsachieved in the presence of calcium ionophore. Different aliquots of same sam-ple were visualized with a Leica TCS-SP confocal microscope with excitation at488 nm and emission at 530 nm.

Subcellular fractionation and Western blot analysis for quantitation of cyto-chrome c and endonuclease G release. Subcellular fractionation was performedby hypotonic lysis followed by use of the Percoll density gradient method at 4°Cas described previously (18). The mitochondrial fraction was judged by cyto-chrome c oxidation assay and kynurenine hydroxylase (an outer membranemarker) assay. Western blot analysis was performed as described previously (18).The primary antibodies used were as follows: rabbit anti-Trypanosoma bruceicytochrome c antibody (1:500), rabbit anti-Leishmania donovani endonuclease G(1:500), goat anti-HSP 60 antibody (1:5,000), rabbit anti-Leishmania donovaniadenosine kinase (1:50), and mouse anti-�-tubulin antibody (1:5,000). The horse-radish peroxidase-conjugated secondary antibodies used were anti-rabbit (1:10,000), anti-mouse (1:6,000), and anti-goat (1:6,000). In each experiment 50 �gof total protein was loaded as described for each case. Precise quantitation wasdone by densitometric analysis to correct the expression of the protein of interestwith that of �-tubulin, HSP 60, or adenosine kinase, which were immunodetectedin the same sample. Densitometric analysis was performed by importing imagesto a personal computer using Total Lab TL 100 software (Nonlinear DynamicsLtd.).

��m measurement. ��m was assayed by flow cytometry with 5,5�,6,6�-tetra-chloro-1,1�,3,3�-tetraethylbenzimidazole carbocyanide iodide (JC-1) as a probe.JC-1 is a cationic and lipophilic vital dye that concentrates in mitochondria in apotential-dependent manner. Measurements were performed according to man-ufacturer’s instruction. Briefly, after treatment, cells were washed twice andresuspended in 1 ml PBS at 106 cells/ml. JC-1 probe was added to a 6 �M finalconcentration and incubated for 20 min at 26°C. For a positive control, 50 �M ofthe mitochondrial uncoupler carbonyl cyanide 3-chlorophenylhydrazone (CCCP)was added to nontreated control cells 15 min prior to addition of JC-1. Analysiswas performed on FacsCanto flow cytometer (Becton Dickinson) equipped with488-nm excitation and 530/610-nm emission filters for green (monomeric form)and red (J-aggregate formation) fluorescence, respectively, after appropriatefluorescence compensation. Data were analyzed with FACSDiva software.

For microscopy, JC-1-labeled equivalent cells were allowed to adhere to poly-L-lysine-coated slides and visualized with a Leica TCS-SP confocal microscope.Mitochondria with higher transmembrane potential accumulate more dye, andemission shifts from 530 nm (green fluorescence) at lower concentrations to 590nm (red fluorescence) at higher concentrations due to J-aggregate formation.

Measurement of protein carbonyl. The protein carbonyl content was detectedwith an Oxyblot protein oxidation detection kit (Chemicon International Ltd.,Hampshire, United Kingdom) according to the manufacturer’s protocol. Totalproteins were extracted from 1 � 107 cells with Laemmli buffer containing 6%sodium dodecyl sulfate. Fifty micrograms of total protein was incubated with 1�2,4-dinitrophenylhydrazine for 20 min at room temperature to form the carbonylderivative dinitrophenylhydrazone. Carbonylated proteins were detected withanti-2,4-dinitrophenol rabbit antibody (diln-200) followed by horseradish perox-idase-conjugated anti-rabbit antibody (diln-300). Blots were developed with anAmersham ECL kit.

Detection of caspase-like protease activity. The detection of caspase activity byFLICA (a caspase 3- and 7-specific fluorogenic inhibitor, FAM-DEVD-FMK) isbased on affinity labeling of the reactive-center cysteine residue of activatedcaspases by the FMK moiety of FLICA via the caspase-specific recognitionsequence aspartic acid-glutamic acid-valine-aspartic acid (DEVD). Apart from

1722 DOLAI ET AL. EUKARYOT. CELL

on February 8, 2021 by guest

http://ec.asm.org/

Dow

nloaded from

Page 3: Overexpression of Mitochondrial Leishmania major Ascorbate Peroxidase … · Ascorbate peroxidase from Leishmania major (LmAPX) is one of the key enzymes for scavenging of reactive

the FMK moiety, FLICA also contains a fluorogenic carboxyfluorescein group asa reporter molecule. Due to the cell membrane permeability, unbound FLICAmolecules diffuse out of the cell, and the green fluorescence indicates the amountof FLICA-bound active caspases within the cell. Activation of caspase-like pro-tein was detected by flow cytometry as per the manufacturer’s instructions.Briefly after treatment with H2O2 or CPT, 106 cells were resuspended in 300 �lof fresh medium and incubated for 1 h in the presence of 10 �l caspase inhibitor.Cells were further washed with 1� wash buffer and finally suspended in 500 �l1� wash buffer and analyzed immediately by flow cytometry (BD FacsCanto)with 488-nm excitation and 530-nm emission wavelengths.

TUNEL staining. Cells undergoing apoptosis produce DNA fragments in thenuclei. TUNEL staining was performed with an Apoalert DNA fragmentationassay kit (Clontech, Mountain View, CA) to detect in situ DNA fragmentationaccording to manufacturer’s manual. Briefly H2O2- or CPT-treated cells wereharvested at different time intervals, washed with PBS, and fixed with 4% form-aldehyde in PBS. Cells were then applied to poly-L-lysine-coated slides andpermeabilized with 0.2% Triton X-100 in PBS. Equilibration was performed byincubating cells in equilibration buffer for 10 min at room temperature, followedby incubation with equilibration buffer containing nucleotide mix and terminaldeoxynucleotidyl transferase enzyme for 1 h in 37°C humidified incubator. Sam-ples were counterstained with 10 �g/ml PI with 1 �g/ml RNase and visualizedunder a Leica TCS-SP confocal microscope.

Measurement of ATP. The cellular ATP concentration was measured by thebioluminescence method using an ATP determination kit (Molecular Probes).Briefly, differently treated cells (1 � 107) were mixed with reaction buffer con-taining 1 mM dithiothreitol, 0.5 mM luciferin, and 12.5 �g/ml luciferase. Theluminescence intensity was measured in a luminometer (Promega). ATP con-centrations were calculated from an ATP standard curve, and cellular ATP levelswere expressed as nmol/106 cells.

Apoptosis assessment by annexin V staining. Phosphatidylserine exposure wasassessed with the Vybrant apoptosis assay kit no. 3 (Molecular Probes). Afterincubation with H2O2 or CPT, cells were harvested by centrifugation for 5 minat 1,200 � g and washed twice with cold 1� PBS. Cells (1 � 106/ml) were thenresuspended in 100 �l 1� annexin binding buffer with 5 �l fluorescein isothio-cyanate (FITC)-conjugated annexin V and 1 �g/ml PI and incubated for 20 min.After staining, 400 �l of 1� buffer was added to the cells, and samples werestored on ice until data acquisition by flow cytometry. To eliminate the emissionspectral overlap of fluophores, fluorescence compensation was performed withunstained and 8-h-treated single-stained (with either PI or with FITC) samples.Measurements were completed within 1 h. For confocal imaging, only annexinV-bound cells were used.

Statistical analysis. All results were expressed as the mean standard error(SE) from at least three independent experiments. Statistical analysis for para-metric data was done by Student’s t test or analysis of variance wherever appli-cable using Origin 6.0 software (Microcal Software, Inc. Northampton, MA).The analysis of variance was followed by post hoc analysis (multiple-comparisont test) for the evaluation of the difference between individual groups. A P valueof less than 0.05 was considered statistically significant.

RESULTS

LmAPX prevents both cellular GSH depletion and accumu-lation of oxidized proteins. Like H2O2, CPT is an in vivoROS-producing agent that acts by inhibiting class I DNA to-poisomerase and induces apoptotic death in cultured mamma-lian cells (50). In Leishmania cells, CPT has been shown toinduce apoptosis by generation of ROS, an imbalance in cyto-solic cations, mitochondrial dysfunction, and subsequent acti-vation of caspase-like molecules (48). Depletion of cellularGSH or protein oxidation is often used as a measure of thelevel of ROS within the cell. We measured the cellular GSHcontent in control and LmAPX-overexpressing cells treated forup to 4 h with H2O2 or CPT to induce oxidative stress. Cellsoverexpressing LmAPX show more than a 2.5-fold-higher levelof GSH after 1 h of treatment and more than 1.5-fold protec-tion throughout the 4 h of treatment, indicating a less-oxidizingenvironment in overexpressing cells (Table 1). Protein car-bonyl content is the most general and well-used biomarker of

protein oxidation within cells (15). Since a decrease in ROSproduction could probably cause less oxidative damage to pro-teins, we monitored the protein carbonyl content of cellularhomogenates by derivatization of the carbonyl group with 2,4-dinitrophenylhydrazine and subsequent immunodetection ofthe resulting hydrazone with the Oxyblot protein oxidationdetection kit (Fig. 1). Treatment of H2O2 and CPT in bothcontrol and LmAPX-overexpressing cells resulted in a time-dependent increase in band intensity (Fig. 1A and C). Densi-tometry quantification of the bands revealed 16-fold and 10-fold increases in the carbonyl content of proteins in controlcells subjected to 8 h of H2O2 and CPT treatment, respectively,whereas LmAPX-overexpressing cells under identical treat-ment conditions showed increases of only 6-fold and 4-fold inprotein carbonyl content in H2O2- and CPT-treated overex-pressing cells, respectively (Fig. 1 B and D). The band intensityin LmAPX-overexpressing cells was at least 2.5-fold lower thanthat in the control cells throughout the 8 h of treatment. Theseresults indicate that overexpression of LmAPX protectedLeishmania cells against oxidative damage of proteins by H2O2

or CPT treatment.LmAPX overexpression prevents oxidative stress-induced

elevation of cytosolic calcium. It is now recognized that ROS inmitochondria play a key role in intracellular Ca2� homeostasis(38, 51). Perturbation of homeostasis and elevation of cytosolicCa2� can lead to apoptotic cell death. Hence, we askedwhether overexpression of LmAPX defended against oxidativestress-mediated elevation of cytosolic Ca2�. We measuredCa2� levels in both control cells and LmAPX-overexpressingcells after treatment with H2O2 and CPT at different timepoints by confocal microscopy and fluorescence spectroscopyusing the Fluo 4 dye as a calcium sensor (Fig. 2). Elevation ofCa2� was usually monitored by an increase in the green fluo-rescence intensity at 518 nm resulting from fluorescence emis-sion of Ca2�-bound Fluo 4 (excitation, 490 nm). The fluores-cence emission peak at 518 nm was raised when either cell typewas treated with H2O2 or CPT for 90 min. However, theintensity of the peak at 518 nm for the LmAPX-overexpressingcells was at least twofold lower than that for the control cells,indicating that overexpression of LmAPX protected cellsagainst oxidative stress-mediated elevation of cytosolic Ca2�

(Fig. 2A). Also, at other time points after H2O2 and CPT

TABLE 1. Effect of LmAPX overexpression on cellular GSH levela

Time (h)

Fluorescence units (mean SEM)b after treatment with:

H2O2 CPT

Controlcells

LmAPX-overexpressing

cells

Controlcells

LmAPX-overexpressing

cells

0 318 12 322 10 309 15 312 201 185 10* 272 13* 209 10* 296 22*2 134 8* 231 11* 172 12* 260 15*3 75 5* 139 8* 148 10* 216 14*4 62 4* 112 7* 102 4* 172 8*

a The intracellular GSH level was measured using monochlorobimane aftertreatment with either CPT (5�M) or H2O2 (1 mM). An increase in fluorescenceunits reflects a larger amount of GSH present in the cells. All experiments wereperformed in triplicate.

b �, P 0.01.

VOL. 8, 2009 ROLE OF L. MAJOR ASCORBATE PEROXIDASE IN APOPTOSIS 1723

on February 8, 2021 by guest

http://ec.asm.org/

Dow

nloaded from

Page 4: Overexpression of Mitochondrial Leishmania major Ascorbate Peroxidase … · Ascorbate peroxidase from Leishmania major (LmAPX) is one of the key enzymes for scavenging of reactive

treatment, the elevation of cytosolic Ca2� was much lower inLmAPX-overexpressing cells than in control cells (Fig. 2B).

To validate the above results by an alternative method, thelevel of cytosolic Ca2� was analyzed by Fluo 4AM fluorescenceimaging by microscopy. The data presented in Fig. 2C clearlyconfirmed the data obtained with the fluorescence spectropho-

tometer. Although the nontreated control had very low greensignals, nontreated overexpressing cells had no green signal.The treatment with H2O2 or CPT caused elevation of Ca2� at0 to 1.5 h in both control and LmAPX-overexpressing cells, asshown by an increase in fluorescence at 518 nm for control cellsand LmAPX-overexpressing cells. The level of elevation of

FIG. 1. Effect of LmAPX overexpression on cellular protein oxidation. Total cell lysates were derivatized with 2,4-dinitrophenylhydrazine asdescribed in Materials and Methods, and 20 �g of each was subjected to immunoblot analysis either with anti-2,4-dinitrophenol antibody or�-tubulin antibody (as a loading control). (A) Immunoblot of H2O2-treated control cells (lanes 3, 5, 7, and 9) or cells overexpressing LmAPX (lanes4, 6, 8, and 10). (B) The bar graph represents the percentage of carbonylated proteins in H2O2-treated cells expressed as percentage of bandintensity. CT, control cells; OE, cells overexpressing LmAPX. (C) Immunoblot of CPT-treated control cells (lanes 3, 5, 7, and 9) or cellsoverexpressing LmAPX (lanes 4, 6, 8, and 10). Lanes 1 and 2 of both panels A and C represented untreated control and LmAPX-overexpressingcells, respectively. (D) The bar graph represents the percentage of carbonylated proteins in CPT-treated cells expressed as percentage of bandintensity. Band intensity was quantified by Total Lab TL100 software, and error bars represent the SE from three independent experiments. Thepositions of molecular mass standards are indicated.

FIG. 2. LmAPX overexpression suppresses cytosolic Ca2� release. The intracellular Ca2� level was measured using Fluo 4 AM as an indicatorafter treatment with either CPT (5 �M) or H2O2. CT, control cells; OE, cells overexpressing LmAPX. (A) Changes in intracellular release of Ca2�

were compared by fluorescence spectrophotometry. (B) Analysis of time-dependent elevation of intracellular Ca2� level. The data shown aremeans SEs. (C) The increase in Ca2� level was visualized by confocal microscopy. Data are representative of at least three independentexperiments.

1724 DOLAI ET AL. EUKARYOT. CELL

on February 8, 2021 by guest

http://ec.asm.org/

Dow

nloaded from

Page 5: Overexpression of Mitochondrial Leishmania major Ascorbate Peroxidase … · Ascorbate peroxidase from Leishmania major (LmAPX) is one of the key enzymes for scavenging of reactive

Ca2� attained by the LmAPX-overexpressing cells was clearlylower than that observed in control cells from 0 to 1.5 h inter-vals.

LmAPX overexpression prevents oxidative stress-inducedloss of ��m and a decline in cellular ATP generation. Intra-cellular ROS buildup and elevation of cytosolic Ca2� can in-duce mitochondrial dysfunction, which was highly coupled witha collapse in ��m (7). To examine whether overexpression ofLmAPX shielded against oxidative stress-mediated loss of��m, we measured the ��m by flow cytometry assay and con-

focal microscopy using the potentiometric fluorescent dye JC-1(Fig. 3; see Fig. S2 in the supplemental material). A shift in thefluorescence emission from green (535 nm) to red (595 nm)indicates accumulation of JC-1 in the mitochondria, which isdependent solely on the membrane potential of the mitochon-dria. Consequently, mitochondrial membrane depolarization isusually accompanied by a decrease in the fluorescence inten-sity ratio (red/green). We observed that incubation with themitochondrial uncoupler CCCP reduced the JC-1 fluorescenceintensity ratio, indicating that the JC-1 response in Leishmania

FIG. 3. Effect of LmAPX overexpression on preservation of ��m in L. major. L. major cells (107/ml) were incubated with the potential-sensitiveprobe JC-1 (6 �M) for 15 min at 25°C to asses ��m after treatment with H2O2 (1 mM) or CPT (5 �M) for the indicated times and analyzed byflow cytometry and confocal microscopy with excitation at 488 nm. Emission was detected at 530 nm (monomer) and 590 nm (aggregate). A dropin ��m is identified as a change in JC-1 properties from forming J-aggregates (emission at 590 nm, red color) at high ��m to forming J-monomers(emission at 530 nm, green color) at low ��m. The nearly complete monomer was induced by treating cells with 50 �M CCCP, an uncoupler ofmitochondrial respiration, 15 min prior to addition of JC-1. (A) Dot plots of blank cells (cells without JC-1), cells with JC-1, H2O2-treatedJC-1-stained cells, and CCCP-treated JC-1-stained cells. CT and OE, control cells and LmAPX-overexpressing cells, respectively. (B) Effect ofH2O2 (6 h) and CCCP treatment on ��m in CT and OE cells. Bar graphs represent the ratio of mean fluorescence intensity (590/530) of total cellsanalyzed by flow cytometry. Error bars indicate the SE from three independent experiments. The asterisks indicate the level of statisticalsignificance (0.05). (C) Time-dependent analysis of 590/530 values of H2O2-treated CT and OE cells. (D) Time-dependent measurement ofintracellular ATP levels in CT and OE cells in the presence of either CPT or H2O2. ATP concentration is expressed as nanomoles of ATP/106 cells.

VOL. 8, 2009 ROLE OF L. MAJOR ASCORBATE PEROXIDASE IN APOPTOSIS 1725

on February 8, 2021 by guest

http://ec.asm.org/

Dow

nloaded from

Page 6: Overexpression of Mitochondrial Leishmania major Ascorbate Peroxidase … · Ascorbate peroxidase from Leishmania major (LmAPX) is one of the key enzymes for scavenging of reactive

cells is sensitive to changes in membrane potential (Fig. 3Aand B). Treatment with H2O2 in both control and LmAPX-overexpressing cells resulted in a time-dependent shift in thefluorescence intensity from red (595 nm) to green (535 nm).However, careful analysis of the data revealed that the red/green ratio for the LmAPX-overexpressing cells was at leasttwofold higher than that for the control cells (Fig. 3C). Thisobservation strongly suggested that LmAPX overexpressionprotects cells from H2O2-induced mitochondrial membranedepolarization.

To further substantiate the lower ��m of control cells com-pared with LmAPX-overexpressing cells, fluorescence imagingof samples analyzed by fluorescence-activated cell sorting(FACS) was performed in parallel by confocal microscopy.JC-1 fluorescence imaging (see Fig. S2 in the supplementalmaterial) clearly confirmed the data obtained by FACS. Non-treated control and LmAPX-overexpressing cells showedmarked peripheral red and green signals in a punctuated man-ner. Treatment with H2O2 caused depolarization of mitochon-dria at 0 to 8 h in both control and LmAPX-overexpressingcells, as shown by an increase in green fluorescence at 535 nmfor control cells and LmAPX-overexpressing cells. The obser-vation of lower green fluorescence in overexpressing cells in-dicated that the level of depolarization attained by theLmAPX-overexpressing cells was always below that for controlcells.

��m could affect mitochondrial generation of ATP, which isdirectly proportional to ��m (24). The ATP level is important,as progression to necrosis or apoptosis depends on the avail-ability of ATP (32). Thus, to examine whether overexpressionof LmAPX protects against H2O2- and CPT-induced loss ofATP generation, we measured the ATP content by biolumi-nescence assay using the Molecular Probes ATP determinationkit (A22066) (Fig. 3D). Although there was a gradual fall in theATP levels in both control cells and LmAPX-overexpressingcells, the ATP content for the H2O2- or CPT-treated controlcells was at least twofold lower than that for the LmAPX-overexpressing cells (Fig. 3D). At various time points,LmAPX-overexpressing cells also showed protection againstATP decrease compared to control cell.

LmAPX overexpression prevents oxidative stress-inducedrelease of cytochrome c and endonuclease G from mitochon-dria to the cytosol. Cytochrome c was identified as a factor thatis released from mitochondria to the cytoplasm in apoptoticcells (31). To investigate whether overexpression of LmAPXprotected against oxidative stress-mediated cytochrome c re-lease, we compared H2O2- or CPT-induced cytochrome c re-lease from mitochondria to the cytoplasm in control andLmAPX-overexpressing cells. Subcellular fractionation fol-lowed by Western blotting indicated that CPT and H2O2

treatment causes time-dependent cytochrome c release frommitochondria (Fig. 4A) and subsequent accumulation in thecytoplasm (Fig. 4B). Our experimental results showed that theband intensities of mitochondrial cytochrome c for the controlcells were �2-fold lower than those for the LmAPX-overex-pressing cells. Simultaneously, the band intensities of cyto-plasm cytochrome c for the control cells were �2-fold higherthan those for the LmAPX-overexpressing cells. As controlmarkers for mitochondria and cytoplasm, we used the mito-chondrial HSP 60 and cytosolic adenosine kinase, respectively.

We next tested whether overexpression of LmAPX pro-tected L. major cells against oxidative stress-mediated releaseof another cell death protein, Leishmania endonuclease G (9,21, 47), by comparing endonuclease G released from the mi-tochondria to the cytoplasm in control and LmAPX-overex-pressing cells upon treatment with cell death stimuli. Immu-noblotting with anti-endonuclease G antibody for mitochondrialand cytosolic extracts prepared from parasites showed gradualtranslocation of endonuclease G to the cytosol with time uponH2O2 or CPT treatment (Fig. 4C and D). Our immunoblottingresults showed that the band intensity of mitochondrial endo-nuclease G for the control cells was �2-fold lower than that forthe LmAPX-overexpressing cells (Fig. 4C). Concurrently, theband intensity of cytoplasm endonuclease G for the controlcells was �2-fold higher than that for the LmAPX-overex-pressing cells (Fig. 4D). Although mitochondrial endonucleaseG gradually decreased with time of treatment in both types ofcells, cytosolic endonuclease G was not increased after 4 h.These results suggested that the cytosolic endonuclease Gmight be translocated into the nucleus after 4 h of treatment(9, 21, 47). The equal band intensities corresponding to HSP 60for the mitochondrial extracts and L. donovani adenosine ki-nase for the cytoplasm extracts served as loading controls.

LmAPX-overexpressing cells show low caspase-like activityunder oxidative stress. Caspases are a class of cysteine pro-teases and are the main actors in the metazoan apoptoticpathway. Caspase-like activities have been reported in bothLeishmania donovani and Leishmania major (6, 16). To checkwhether control and LmAPX-overexpressing cells show anydifference in caspase-like activity in the presence of apoptoticstimuli, we used the caspase 3- and caspase 7-specific fluoro-genic inhibitor FAM-DEVD-FMK. Flow cytometric analysisshowed a 56% increase in caspase-like activity from a basallevel in H2O2-treated control cells, compared to only 9% inLmAPX-overexpressing cells (Table 2). CPT-treated cellsshowed 42% and 9% increases in basal activity in control andoverexpressing cells, respectively. In an assessment of caspase-like activity in the presence of a nonfluorogenic caspase inhib-itor (VAD-fmk), pretreated control cells showed almost 86 to90% inhibition of caspase-like activity. LmAPX-overexpress-ing cells showed a basal level of caspase activity in the presenceof the inhibitor (Table 2).

LmAPX overexpression prevents apoptosis. Elevation of cy-tosolic Ca2� and mitochondrial dysfunction are known to beimportant events promoting apoptosis (46). To investigatewhether overexpression of LmAPX shielded against oxidativestress-mediated apoptosis, we measured phosphatidylserineexternalization (Fig. 5) and nuclear DNA fragmentation (Fig.6) as markers of apoptosis. Representative FACS dot plots ofFITC-conjugated annexin V- and PI-labeled samples areshown in Fig. 5A and B. Both H2O2 treatment and CPT treat-ment led to a gradual conversion of viable cells (lower leftquadrant) into early apoptotic (lower right quadrant) and lateapoptotic (upper right quadrant) cells in both control andLmAPX-overexpressing cells. However, overexpression ofLmAPX clearly protected against H2O2- or CPT-induced celldeath by maintaining a higher percentage of viable cells intime-dependent manner. LmAPX overexpression also protectsagainst late apoptosis, with a minimum difference of ninefoldfor the longest time interval. This result suggested that treated

1726 DOLAI ET AL. EUKARYOT. CELL

on February 8, 2021 by guest

http://ec.asm.org/

Dow

nloaded from

Page 7: Overexpression of Mitochondrial Leishmania major Ascorbate Peroxidase … · Ascorbate peroxidase from Leishmania major (LmAPX) is one of the key enzymes for scavenging of reactive

control cells reached the late apoptotic stage faster thanLmAPX-overexpressing cells. For further validation, fluores-cence imaging of FACS-analyzed samples was performed inparallel by confocal microscopy. Annexin V-FITC fluorescenceimaging (see Fig. S3 in the supplemental material) clearlyconfirmed the data obtained by FACS.

DNA fragmentation was assessed by TUNEL staining (Fig.6). As shown in Fig. 6A and B, the formation of red color in

both nucleus and kinetoplast DNAs with PI staining indicatedthat there was no FITC-conjugated dUTP-labeled DNA ineither control or LmAPX-overexpressing cells. On the otherhand, H2O2- or CPT-treated control cells at 6 h showed sig-nificantly more dUTP-labeled green nuclei (Fig. 6C and E)than H2O2- or CPT-treated LmAPX-overexpressing cells (Fig.6D and F), indicating that a larger amount of DNA fragmen-tation occurred in H2O2- or CPT-treated control cells. On the

FIG. 4. Western blot analysis for cytosolic cytochrome c and endonuclease G release from mitochondria isolated from control cells (CT) andLmAPX-overexpressing cells (OE). The concentrations of H2O2 and CPT used were 1 mM and 5 �M, respectively. Lanes 1 and 2, untreatedcontrol and LmAPX-overexpressing cells, respectively; lanes 3, 5, 7, and 9 control cells with 1 h, 2 h, 4 h, and 6 h of treatment, respectively; lanes4, 6, 8, and 10, LmAPX-overexpressing cells with 1 h, 2 h, 4 h, and 6 h of treatment, respectively. Panels A, B, C, and D represent immunoblotand densitometric analysis of cytochrome c in the mitochondrial fraction, cytochrome c in the cytosolic fraction, endonuclease G in themitochondrial fraction, and endonuclease G in the cytosolic fraction, respectively. Band intensity is presented as the percentage of cytochrome cor endonuclease G released from untreated cells. Fifty micrograms of total protein was analyzed by 13% sodium dodecyl sulfate-polyacrylamidegel electrophoresis with antibodies specific for T. brucei cytochrome c, L. donovani endonuclease G, HSP 60 (mitochondrial matrix protein), andL. donovani adenosine kinase (cytosolic protein). No HSP 60 was detected in the cytosolic fraction. All the data are representative of at least threeindependent experiments. *, statistically significant P value of less than 0.05 (P value is less than 0.027).

VOL. 8, 2009 ROLE OF L. MAJOR ASCORBATE PEROXIDASE IN APOPTOSIS 1727

on February 8, 2021 by guest

http://ec.asm.org/

Dow

nloaded from

Page 8: Overexpression of Mitochondrial Leishmania major Ascorbate Peroxidase … · Ascorbate peroxidase from Leishmania major (LmAPX) is one of the key enzymes for scavenging of reactive

basis of dUTP-labeled cell counting, LmAPX overexpressionreduced by around 85% and 80% TUNEL-positive cells com-pared to control cells after H2O2 and CPT treatment, respec-tively (data not shown). The results for overexpressing cellswith H2O2 or CPT treatment indicated that there was 4 to 5times more dUTP-labeled DNA in control cells than inLmAPX-overexpressing cells. Furthermore, in the presence ofVAD-fmk, H2O2- or CPT-induced fragmentation of nuclearDNA was inhibited and there was less labeling with FITC-conjugated dUTP in both control and overexpressing cells(Fig. 6G and H). These results suggested that greater DNAfragmentation occurred in H2O2- or CPT-treated control cellsthan in LmAPX-overexpressing cells.

DISCUSSION

LmAPX is an important enzyme for ROS detoxification andprotection against cardiolipin oxidation under oxidative stress(18). Though plant APX has been extensively characterizedand has been shown to be responsive to several environmentalstresses (37, 43, 52, 59), the exact physiological function of themitochondrial LmAPX enzyme has not been yet clarified. Thework presented here indicates, for the first time, that LmAPXfunctions to protect the Leishmania parasite from oxidativestress-induced cell death. The protective function of LmAPX ismediated by detoxification of excess ROS burden, which is anearly and critical event in preventing release of Ca2�, loss of

TABLE 2. Depletion of drug-induced caspase-like protease activity by LmAPX

Cells

FITC green fluorescence (mean SEM)a with:

No FLICA FLICA FLICA � H2O2 FLICA � CPT H2O2 � VAD-fmk �FLICA

CPT � VAD-fmk �FLICA

Control 126 5 278 10 434 18* 397 30* 328 20* 307 22LmAPX

overexpressing142 8 258 9 281 7* 283 12* 271 10* 277 15

a Increased FLICA green fluorescence indicates increased caspase-like protease activity. Unlabeled control cells were used to set up a flow cytometer by adjustingthe forward and side scatter of the cell population. Values without FLICA indicate the background fluorescence level of unlabeled cells. The increase in fluorescenceintensity was calculated with respect to background fluorescence intensity. The fluorescence data are representative of three independent experiments. Quantitationof the relative change in fluorescence was analyzed with FACSDiVa software. �, P 0.05.

FIG. 5. Changes in plasma membrane phosphatidylserine distribution during apoptosis induced either by H2O2 (1 mM) or CPT (5 �M).Control (CT) and LmAPX-overexpressing (OE) cells were double stained with annexin V and PI and analyzed by flow cytometric analysis at theindicated times. Representative dot plots are divided in four quadrants. Viable cells that did not bind annexin V and also did not incorporate PIare represented by the lower left quadrant of each dot plot, early apoptotic cells that bind only annexin V are represented by the lower rightquadrant, late apoptotic cells that bind annexin V and incorporate PI are represented by the upper right quadrant. Percentages of cells areindicated in the corresponding quadrants. (A) Time-dependent distribution of viable, early apoptotic, and late apoptotic cells among H2O2-challenged control cells (upper panel) and LmAPX-overexpressing cells (lower panel). (B) Time-dependent distribution of viable, early apoptotic,and late apoptotic cells among CPT-treated control cells (upper panel) and LmAPX-overexpressing cells (lower panel). A total of 30,000 cells wereanalyzed in each condition. All results are representative of three independent experiments.

1728 DOLAI ET AL. EUKARYOT. CELL

on February 8, 2021 by guest

http://ec.asm.org/

Dow

nloaded from

Page 9: Overexpression of Mitochondrial Leishmania major Ascorbate Peroxidase … · Ascorbate peroxidase from Leishmania major (LmAPX) is one of the key enzymes for scavenging of reactive

mitochondrial potential, and cytochrome c and endonucleaseG release from mitochondria to the cytosol.

Considerable evidence has accumulated to suggest that ROS(e.g., hydrogen peroxide and NO) may act as mediators ofapoptosis in a variety of cell types (2, 30, 42). Although mito-chondrial respiration, enzymatic reactions (aconitase and �-ketoglutarate dehydrogenase) of the tricarboxylic acid cycle,the microsomal cytochrome P450 system, and plasma mem-brane NADPH oxidase generate intracellular ROS, the ROSfrom mitochondria are responsible for a close association be-tween the activities of mitochondria and cell death (35). In-deed, H2O2 or CPT treatment of L. donovani control cellsresults in mitochondrial dysfunction and is accompanied bycellular death (16, 38, 48). It has been previously shown thatLmAPX is localized exclusively in the mitochondria (18) andutilizes H2O2 and reduced cytochrome c in the reaction cycle;hence, it may act directly on mitochondria and preserve theseorganelles, which represent one of the main targets for ROS-induced damage.

It has long been known that catalase and classical selenium-containing GSH peroxidase, two major hydroperoxide-elimi-nating enzymes generally present in eukaryotes, are missing intrypanosomatid genome sequences. Instead, mainly three dis-tinct families of peroxidase (2-Cys peroxiredoxins, nonsele-

nium GSH peroxidase-like enzymes, and APXs) have beenshown to be crucial in elimination of hydroperoxide and per-oxynitrite in trypanosomatids (12). By knockdown analysis oftrypanothione synthetase, it was suggested that this enzyme isessential for T. brucei survival (5). The trypanothione-reducingenzyme trypanothione reductase has been shown to be criticalfor survival and/or infectivity of L. donovani and T. brucei (39).Another group of highly abundant redox proteins, tryparedox-ins, have diverse cell functions, such as peroxide metabolism,synthesis of deoxynucleotides, and regulation of mitochondrialDNA replication (19, 40). The results from studies with per-oxiredoxin-overexpressing Leishmania and Trypanosoma cruzicells and double-stranded RNA interference studies with T.brucei show that peroxiredoxins have an important role ineliminating hydroperoxides and peroxynitrite (8, 33, 55). Ineukaryotes, peroxiredoxins are considered to act as a regulatorof H2O2-mediated intracellular signaling processes and not asgeneral antioxidant devices. Considerable evidence has sug-gested that nonselenium GSH peroxidase-like enzymes intrypanosomatids metabolize fatty acid and phospholipid hy-droperoxides (56), although the activity is very low comparedto that of selenium-containing GSH peroxidase. Leishmaniaalso possess trypanothione S transferase activity associatedwith the eukaryotic translation elongation factor 1B, whichreacts preferentially with linoleic acid hydroperoxide but notwith H2O2 (54). Recent reports also demonstrate that overex-pression of APX within Leishmania and T. cruzi is more resis-tant to exogenously added H2O2 (18, 57). This heme protein isinvolved in the elimination of H2O2 with a very high catalyticefficiency (k1 � 6.7 � 107 M�1 s�1) (58), and it acts as apseudocatalase in the presence of low concentration of ascor-bate (17). Because LmAPX can oxidize ferrocytochrome c inthe presence of endogenous H2O2, it is an excellent candidatefor playing dual roles in mitochondria by elimination of bothH2O2 and superoxide.

Recently we have proposed that cyclic oxidation/reductionof the cytochrome c by the LmAPX system may function as aROS-scavenging system (18, 58). In mammalian cells, an in-crease in cellular ROS production has been claimed to beresponsible for cell death (53). From this study, it is establishedthat overexpression of LmAPX in Leishmania cells clearly pro-tects cells against H2O2 (an exogenous oxidant) or CPT (anendogenous oxidant)-mediated mitochondrial apoptotic celldeath. It is well known that apoptosis in mammalian cells hasbeen tightly linked to activation of caspases (45), which aremissing in the Leishmania genome sequence. It is notable thatROS act as mediators in many cases of caspase-like protein-dependent as well as -independent DNA fragmentation inLeishmania cells (10, 21). Another peculiar aspect emergingfrom our experiments is the fact that although the TUNELassay suggests that oxidant-induced DNA cleavage is blockedby overexpression of LmAPX, DNA degradation could not becompletely blocked by using a caspase inhibitor. The reason forthis discrepancy could be an involvement of oxidant-dependentcaspase-like protease activation on one hand (6, 16), but on theother hand it suggests the existence of some alternative oxi-dant-dependent apoptotic pathway (endonuclease G depen-dent) leading to nuclear fragmentation in Leishmania (9, 21,47). Our endonuclease G release data presented in Fig. 4C andD clearly confirmed the data obtained by TUNEL assay.

FIG. 6. TUNEL staining to determine the level of in situ oxidativestress-induced DNA fragmentation. TUNEL-staining cells were coun-terstained with PI in the presence of RNase. (A) Merged image ofcontrol cells under bright-field microscopy. (B) Merged image ofLmAPX-overexpressing cells under bright-field microscopy. (C) Con-trol cells with H2O2 treatment for 6 h. (D) Overexpressing cells withH2O2 treatment for 6 h. (E) Control cells with CPT treatment for 6 h.(F) Overexpressing cells with CPT treatment for 6 h. (G) Control cellswith H2O2 treatment for 6 h in the presence of a caspase inhibitor(VAD-fmk). (H) Overexpressing cells with H2O2 treatment for 6 h inthe presence of a caspase inhibitor (VAD-fmk). All results are repre-sentative of three independent experiments.

VOL. 8, 2009 ROLE OF L. MAJOR ASCORBATE PEROXIDASE IN APOPTOSIS 1729

on February 8, 2021 by guest

http://ec.asm.org/

Dow

nloaded from

Page 10: Overexpression of Mitochondrial Leishmania major Ascorbate Peroxidase … · Ascorbate peroxidase from Leishmania major (LmAPX) is one of the key enzymes for scavenging of reactive

Mitochondria are both producers and targets of ROS, andan increase of ROS is known to trigger cells to undergo apop-tosis through the activation of caspases, especially caspase 3(10). The prevention of phosphatidylserine exposure and DNAfragmentation in the LmAPX-overexpressing cells after expo-sure to oxidative stress indicates their ability to resist proapop-totic changes induced by oxidative stress. In the absence ofBcl2 family proteins in Leishmania that regulate apoptosisinduced by diverse stimuli in human host cells, LmAPX ap-pears to be an important protein in ROS-induced apoptosisregulation. Thus, understanding the molecular function of sin-gle-copy mitochondrial LmAPX provides opportunities for dis-covering and evaluating molecular targets for drug design,which now forms a rational basis for the development of im-proved therapy against leishmaniasis.

ACKNOWLEDGMENTS

We thank S. M. Beverley for providing pXG-B2863 vector, A. K.Datta for L. donovani adenosine kinase antibody, H. K. Majumder forL. donovani endonuclease G antibody, and Andre Schneider (Univer-sity of Berne, Switzerland) for T. brucei cytochrome c antibody. Wethank Arunima Biswas for her help with flow cytometry.

This work was supported by Council of Scientific and IndustrialResearch (CSIR) project NWP 0038 and CSIR fellowships (to S.D.,R.K.Y., and S.P.).

REFERENCES

1. Adak, S., and A. K. Datta. 2005. Leishmania major encodes an unusualperoxidase that is a close homologue of plant ascorbate peroxidase: a novelrole of the transmembrane domain. Biochem. J. 390:465–474.

2. Albina, J. E., S. Cui, R. B. Mateo, and J. S. Reichner. 1993. Nitric oxide-mediated apoptosis in murine peritoneal macrophages. J. Immunol. 150:5080–5085.

3. Al-Olayan, E. M., G. T. Williams, and H. Hurd. 2002. Apoptosis in themalaria protozoan, Plasmodium berghei: a possible mechanism for limitingintensity of infection in the mosquito. Int. J. Parasitol. 32:1133–1143.

4. Ameisen, J. C., T. Idziorek, O. Billaut-Mulot, M. Loyens, J. P. Tissier, A.Potentier, and A. Ouaissi. 1995. Apoptosis in a unicellular eukaryote(Trypanosoma cruzi): implications for the evolutionary origin and role ofprogrammed cell death in the control of cell proliferation, differentiation andsurvival. Cell Death Differ. 2:285–300.

5. Ariyanayagam, M. R., S. L. Oza, M. L. Guther, and A. H. Fairlamb. 2005.Phenotypic analysis of trypanothione synthetase knockdown in the Africantrypanosome. Biochem. J. 391:425–432.

6. Arnoult, D., K. Akarid, A. Grodet, P. X. Petit, J. Estaquier, and J. C.Ameisen. 2002. On the evolution of programmed cell death: apoptosis of theunicellular eukaryote Leishmania major involves cysteine proteinase activa-tion and mitochondrion permeabilization. Cell Death Differ. 9:65–81.

7. Balaban, R. S., S. Nemoto, and T. Finkel. 2005. Mitochondria, oxidants, andaging. Cell 120:483–495.

8. Barr, S. D., and L. Gedamu. 2003. Role of peroxidoxins in Leishmaniachagasi survival. Evidence of an enzymatic defense against nitrosative stress.J. Biol. Chem. 278:10816–10823.

9. BoseDasgupta, S., B. B. Das, S. Sengupta, A. Ganguly, A. Roy, S. Dey, G.Tripathi, B. Dinda, and H. K. Majumder. 2008. The caspase-independentalgorithm of programmed cell death in Leishmania induced by baicalein: therole of LdEndoG, LdFEN-1 and LdTatD as a DNA ‘degradesome.’ CellDeath Differ. 15:1629–1640.

10. Burkle, A. 2005. Poly(ADP-ribose). The most elaborate metabolite ofNAD�. FEBS J. 272:4576–4589.

11. Cadenas, E. 2004. Mitochondrial free radical production and cell signaling.Mol. Aspects Med. 25:17–26.

12. Castro, H., and A. M. Tomas. 2008. Peroxidases of trypanosomatids. Anti-oxid. Redox Signal. 10:1593–1606.

13. Chan, J., T. Fujiwara, P. Brennan, M. McNeil, S. J. Turco, J. C. Sibille, M.Snapper, P. Aisen, and B. R. Bloom. 1989. Microbial glycolipids: possiblevirulence factors that scavenge oxygen radicals. Proc. Natl. Acad. Sci. USA86:2453–2457.

14. Channon, J. Y., and J. M. Blackwell. 1985. A study of the sensitivity ofLeishmania donovani promastigotes and amastigotes to hydrogen peroxide.II. Possible mechanisms involved in protective H2O2 scavenging. Parasitol-ogy 91:207–217.

15. Dalle-Donne, I., D. Giustarini, R. Colombo, R. Rossi, and A. Milzani. 2003.Protein carbonylation in human diseases. Trends Mol. Med. 9:169–176.

16. Das, M., S. B. Mukherjee, and C. Shaha. 2001. Hydrogen peroxide inducesapoptosis-like death in Leishmania donovani promastigotes. J. Cell Sci. 114:2461–2469.

17. Dolai, S., R. K. Yadav, A. K. Datta, and S. Adak. 2007. Effect of thiocyanateon the peroxidase and pseudocatalase activities of Leishmania major ascor-bate peroxidase. Biochim. Biophys. Acta 1770:247–256.

18. Dolai, S., R. K. Yadav, S. Pal, and S. Adak. 2008. Leishmania major ascor-bate peroxidase overexpression protects cells against reactive oxygen species-mediated cardiolipin oxidation. Free Radic. Biol. Med. 45:1520–1529.

19. Dormeyer, M., N. Reckenfelderbaumer, H. Ludemann, and R. L. Krauth-Siegel. 2001. Trypanothione-dependent synthesis of deoxyribonucleotides byTrypanosoma brucei ribonucleotide reductase. J. Biol. Chem. 276:10602–10606.

20. Engelberg-Kulka, H., and G. Glaser. 1999. Addiction modules and pro-grammed cell death and antideath in bacterial cultures. Annu. Rev. Micro-biol. 53:43–70.

21. Gannavaram, S., C. Vedvyas, and A. Debrabant. 2008. Conservation of thepro-apoptotic nuclease activity of endonuclease G in unicellular trypanoso-matid parasites. J. Cell Sci. 121:99–109.

22. Gantt, K. R., T. L. Goldman, M. L. McCormick, M. A. Miller, S. M.Jeronimo, E. T. Nascimento, B. E. Britigan, and M. E. Wilson. 2001. Oxi-dative responses of human and murine macrophages during phagocytosis ofLeishmania chagasi. J. Immunol. 167:893–901.

23. Ghosh, S., S. Goswami, and S. Adhya. 2003. Role of superoxide dismutase insurvival of Leishmania within the macrophage. Biochem. J. 369:447–452.

24. Gottlieb, R. A. 2001. Mitochondria and apoptosis. Biol. Signals Recept.10:147–161.

25. Grassme, H., V. Jendrossek, and E. Gulbins. 2001. Molecular mechanisms ofbacteria induced apoptosis. Apoptosis 6:441–445.

26. Harder, S., M. Bente, K. Isermann, and I. Bruchhaus. 2006. Expression of amitochondrial peroxiredoxin prevents programmed cell death in Leishmaniadonovani. Eukaryot. Cell 5:861–870.

27. Hengartner, M. O. 2000. The biochemistry of apoptosis. Nature 407:770–776.

28. Hockenbery, D. M., Z. N. Oltvai, X. M. Yin, C. L. Milliman, and S. J.Korsmeyer. 1993. Bcl-2 functions in an antioxidant pathway to preventapoptosis. Cell 75:241–251.

29. Iyer, J. P., A. Kaprakkaden, M. L. Choudhary, and C. Shaha. 2008. Crucialrole of cytosolic tryparedoxin peroxidase in Leishmania donovani survival,drug response and virulence. Mol. Microbiol. 68:372–391.

30. Kane, D. J., T. A. Sarafian, R. Anton, H. Hahn, E. B. Gralla, J. S. Valentine,T. Ord, and D. E. Bredesen. 1993. Bcl-2 inhibition of neural death: decreasedgeneration of reactive oxygen species. Science 262:1274–1277.

31. Kluck, R. M., E. Bossy-Wetzel, D. R. Green, and D. D. Newmeyer. 1997. Therelease of cytochrome c from mitochondria: a primary site for Bcl-2 regula-tion of apoptosis. Science 275:1132–1136.

32. Lemasters, J. J., T. Qian, C. A. Bradham, D. A. Brenner, W. E. Cascio, L. C.Trost, Y. Nishimura, A. L. Nieminen, and B. Herman. 1999. Mitochondrialdysfunction in the pathogenesis of necrotic and apoptotic cell death.J. Bioenerg. Biomembr. 31:305–319.

33. Lin, Y. C., J. Y. Hsu, S. C. Chiang, and S. T. Lee. 2005. Distinct overexpres-sion of cytosolic and mitochondrial tryparedoxin peroxidases results in pref-erential detoxification of different oxidants in arsenite-resistant Leishmaniaamazonensis with and without DNA amplification. Mol. Biochem. Parasitol.142:66–75.

34. Madeo, F., E. Frohlich, M. Ligr, M. Grey, S. J. Sigrist, D. H. Wolf, and K. U.Frohlich. 1999. Oxygen stress: a regulator of apoptosis in yeast. J. Cell Biol.145:757–767.

35. Mignotte, B., and J. L. Vayssiere. 1998. Mitochondria and apoptosis. Eur.J. Biochem. 252:1–15.

36. Miller, M. A., S. E. McGowan, K. R. Gantt, M. Champion, S. L. Novick, K. A.Andersen, C. J. Bacchi, N. Yarlett, B. E. Britigan, and M. E. Wilson. 2000.Inducible resistance to oxidant stress in the protozoan Leishmania chagasi.J. Biol. Chem. 275:33883–33889.

37. Mittler, M., and B. A. Zilinskas. 1994. Regulation of pea cytosolic ascorbateperoxidase and other antioxidants enzymes during the progression ofdrought stress and following recovery from drought. Plant J. 5:397–405.

38. Mukherjee, S. B., M. Das, G. Sudhandiran, and C. Shaha. 2002. Increase incytosolic Ca2� levels through the activation of non-selective cation channelsinduced by oxidative stress causes mitochondrial depolarization leading toapoptosis-like death in Leishmania donovani promastigotes. J. Biol. Chem.277:24717–24727.

39. Muller, S., E. Liebau, R. D. Walter, and R. L. Krauth-Siegel. 2003. Thiol-based redox metabolism of protozoan parasites. Trends Parasitol. 19:320–328.

40. Onn, I., N. Milman-Shtepel, and J. Shlomai. 2004. Redox potential regulatesbinding of universal minicircle sequence binding protein at the kinetoplastDNA replication origin. Eukaryot. Cell 3:277–287.

41. Piacenza, L., F. Irigoin, M. N. Alvarez, G. Peluffo, M. C. Taylor, J. M. Kelly,S. R. Wilkinson, and R. Radi. 2007. Mitochondrial superoxide radicals me-diate programmed cell death in Trypanosoma cruzi: cytoprotective action of

1730 DOLAI ET AL. EUKARYOT. CELL

on February 8, 2021 by guest

http://ec.asm.org/

Dow

nloaded from

Page 11: Overexpression of Mitochondrial Leishmania major Ascorbate Peroxidase … · Ascorbate peroxidase from Leishmania major (LmAPX) is one of the key enzymes for scavenging of reactive

mitochondrial iron superoxide dismutase overexpression. Biochem. J. 403:323–334.

42. Polyak, K., Y. Xia, J. L. Zweier, K. W. Kinzler, and B. Vogelstein. 1997. Amodel for p53-induced apoptosis. Nature 389:300–305.

43. Prasad, T. K., M. D. Anderson, B. A. Martin, and C. R. Stewart. 1994.Evidence for chilling-induced oxidative stress in maize seedlings and a reg-ulatory role for hydrogen peroxide. Plant Cell 6:65–74.

44. Rhee, S. G., K. S. Yang, S. W. Kang, H. A. Woo, and T. S. Chang. 2005.Controlled elimination of intracellular H(2)O(2): regulation of peroxire-doxin, catalase, and glutathione peroxidase via post-translational modifica-tion. Antioxid. Redox Signal. 7:619–626.

45. Rich, T., C. J. Watson, and A. Wyllie. 1999. Apoptosis: the germs of death.Nat. Cell Biol. 1:E69–E71.

46. Richter, C., M. Schweizer, A. Cossarizza, and C. Franceschi. 1996. Controlof apoptosis by the cellular ATP level. FEBS Lett. 378:107–110.

47. Rico, E., J. F. Alzate, A. A. Arias, D. Moreno, J. Clos, F. Gago, I. Moreno, M.Dominguez, and A. Jimenez-Ruiz. 2009. Leishmania infantum expresses amitochondrial nuclease homologous to EndoG that migrates to the nucleusin response to an apoptotic stimulus. Mol. Biochem. Parasitol. 163:28–38.

48. Sen, N., B. B. Das, A. Ganguly, T. Mukherjee, G. Tripathi, S. Bandyo-padhyay, S. Rakshit, T. Sen, and H. K. Majumder. 2004. Camptothecininduced mitochondrial dysfunction leading to programmed cell death inunicellular hemoflagellate Leishmania donovani. Cell Death Differ. 11:924–936.

49. Shaha, C. 2006. Apoptosis in Leishmania species and its relevance to diseasepathogenesis. Indian J. Med. Res. 123:233–244.

50. Simizu, S., M. Takada, K. Umezawa, and M. Imoto. 1998. Requirement ofcaspase-3(-like) protease-mediated hydrogen peroxide production for apop-tosis induced by various anticancer drugs. J. Biol. Chem. 273:26900–26907.

51. Tagliarino, C., J. J. Pink, G. R. Dubyak, A. L. Nieminen, and D. A. Booth-man. 2001. Calcium is a key signaling molecule in beta-lapachone-mediatedcell death. J. Biol. Chem. 276:19150–19159.

52. Tanaka, K., Y. Suda, N. Kondo, and K. Sugahara. 1985. O3 tolerance andthe ascorbate-dependent H2O2 decomposing system in chloroplasts. PlantCell Physiol. 26:1425–1431.

53. Vander Heiden, M. G., N. S. Chandel, E. K. Williamson, P. T. Schumacker,and C. B. Thompson. 1997. Bcl-xL regulates the membrane potential andvolume homeostasis of mitochondria. Cell 91:627–637.

54. Vickers, T. J., S. Wyllie, and A. H. Fairlamb. 2004. Leishmania major elon-gation factor 1B complex has trypanothione S-transferase and peroxidaseactivity. J. Biol. Chem. 279:49003–49009.

55. Wilkinson, S. R., D. Horn, S. R. Prathalingam, and J. M. Kelly. 2003. RNAinterference identifies two hydroperoxide metabolizing enzymes that areessential to the bloodstream form of the African trypanosome. J. Biol. Chem.278:31640–31646.

56. Wilkinson, S. R., D. J. Meyer, and J. M. Kelly. 2000. Biochemical charac-terization of a trypanosome enzyme with glutathione-dependent peroxidaseactivity. Biochem. J. 352:755–761.

57. Wilkinson, S. R., S. O. Obado, I. L. Mauricio, and J. M. Kelly. 2002.Trypanosoma cruzi expresses a plant-like ascorbate-dependent hemoperoxi-dase localized to the endoplasmic reticulum. Proc. Natl. Acad. Sci. USA99:13453–13458.

58. Yadav, R. K., S. Dolai, S. Pal, and S. Adak. 2008. Role of tryptophan-208residue in cytochrome c oxidation by ascorbate peroxidase from Leishmaniamajor-kinetic studies on Trp208Phe mutant and wild type enzyme. Biochim.Biophys. Acta 1784:863–871.

59. Yoshimura, K., Y. Yabuta, T. Ishikawa, and S. Shigeoka. 2000. Expression ofspinach ascorbate peroxidase isoenzymes in response to oxidative stresses.Plant Physiol. 123:223–234.

60. Zangger, H., J. C. Mottram, and N. Fasel. 2002. Cell death in Leishmaniainduced by stress and differentiation: programmed cell death or necrosis?Cell Death Differ. 9:1126–1139.

VOL. 8, 2009 ROLE OF L. MAJOR ASCORBATE PEROXIDASE IN APOPTOSIS 1731

on February 8, 2021 by guest

http://ec.asm.org/

Dow

nloaded from