25
MITOCHONDRIAL OXIDANT STRESS TRIGGERS CELL DEATH IN SIMULATED ISCHEMIA-REPERFUSION Gabriel Loor 1 , Jyothisri Kondapalli 3 , Hirotaro Iwase 2 , Navdeep S. Chandel 4 , Gregory B. Waypa 3 , Robert D. Guzy 3 , Terry L. Vanden Hoek 2 , and Paul T. Schumacker 2,3,4 1 Department of Surgery, University of Chicago, Chicago IL 60637 2 Department of Medicine, University of Chicago, Chicago IL 60637 3 Department of Pediatrics, Northwestern University, Chicago, IL 60611 4 Department of Medicine, Northwestern University, Chicago, IL 60611 Abstract To clarify the relationship between reactive oxygen species (ROS) and cell death during ischemia- reperfusion (I/R), we studied cell death mechanisms in a cellular model of I/R. Oxidant stress during simulated ischemia was detected in the mitochondrial matrix using mito-roGFP, a ratiometric redox sensor, and by Mito-Sox Red oxidation. Reperfusion-induced death was attenuated by over-expression of Mn-superoxide dismutase (Mn-SOD) or mitochondrial phospholipid hydroperoxide glutathione peroxidase (mito-PHGPx), but not by catalase, mitochondria-targeted catalase, or Cu,Zn-SOD. Protection was also conferred by chemically distinct antioxidant compounds, and mito-roGFP oxidation was attenuated by NAC, or by scavenging of residual O 2 during the ischemia (anoxic ischemia). Mitochondrial permeability transition pore (mPTP) oscillation/opening was monitored by real-time imaging of mitochondrial calcein fluorescence. Oxidant stress caused release of calcein to the cytosol during ischemia, a response that was inhibited by chemically diverse antioxidants, anoxia, or over-expression of Mn- SOD or mito-PHGPx. These findings suggest that mitochondrial oxidant stress causes oscillation of the mPTP prior to reperfusion. Cytochrome c release from mitochondria to the cytosol was not detected until after reperfusion, and was inhibited by anoxic ischemia or antioxidant administration during ischemia. Although DNA fragmentation was detected after I/R, no evidence of Bax activation was detected. Over-expression of the anti-apoptotic protein Bcl-X L in cardiomyocytes did not confer protection against I/R-induced cell death. Moreover, murine embryonic fibroblasts with genetic depletion of Bax and Bak, or over-expression of Bcl-X L , failed to show protection against I/R. These findings indicate that mitochondrial ROS during ischemia triggers mPTP activation, mitochondrial depolarization, and cell death during reperfusion through a Bax/Bak-independent cell death pathway. Therefore, mitochondrial apoptosis appears to represent a redundant death pathway in this model of simulated I/R. © 2010 Elsevier B.V. All rights reserved. Correspondence: Paul T. Schumacker, Ph.D., Department of Pediatrics, Northwestern University Feinberg School of Medicine, 310 East Superior St., Morton Bldg. 4-685, Chicago, IL 60611, Tel. 312 503-1475, Fax 312 503-1181, [email protected]. Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. NIH Public Access Author Manuscript Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1. Published in final edited form as: Biochim Biophys Acta. 2011 July ; 1813(7): 1382–1394. doi:10.1016/j.bbamcr.2010.12.008. NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

Mitochondrial oxidant stress triggers cell death in simulated ischemia–reperfusion

Embed Size (px)

Citation preview

MITOCHONDRIAL OXIDANT STRESS TRIGGERS CELL DEATHIN SIMULATED ISCHEMIA-REPERFUSION

Gabriel Loor1, Jyothisri Kondapalli3, Hirotaro Iwase2, Navdeep S. Chandel4, Gregory B.Waypa3, Robert D. Guzy3, Terry L. Vanden Hoek2, and Paul T. Schumacker2,3,4

1Department of Surgery, University of Chicago, Chicago IL 606372Department of Medicine, University of Chicago, Chicago IL 606373Department of Pediatrics, Northwestern University, Chicago, IL 606114Department of Medicine, Northwestern University, Chicago, IL 60611

AbstractTo clarify the relationship between reactive oxygen species (ROS) and cell death during ischemia-reperfusion (I/R), we studied cell death mechanisms in a cellular model of I/R. Oxidant stressduring simulated ischemia was detected in the mitochondrial matrix using mito-roGFP, aratiometric redox sensor, and by Mito-Sox Red oxidation. Reperfusion-induced death wasattenuated by over-expression of Mn-superoxide dismutase (Mn-SOD) or mitochondrialphospholipid hydroperoxide glutathione peroxidase (mito-PHGPx), but not by catalase,mitochondria-targeted catalase, or Cu,Zn-SOD. Protection was also conferred by chemicallydistinct antioxidant compounds, and mito-roGFP oxidation was attenuated by NAC, or byscavenging of residual O2 during the ischemia (anoxic ischemia). Mitochondrial permeabilitytransition pore (mPTP) oscillation/opening was monitored by real-time imaging of mitochondrialcalcein fluorescence. Oxidant stress caused release of calcein to the cytosol during ischemia, aresponse that was inhibited by chemically diverse antioxidants, anoxia, or over-expression of Mn-SOD or mito-PHGPx. These findings suggest that mitochondrial oxidant stress causes oscillationof the mPTP prior to reperfusion. Cytochrome c release from mitochondria to the cytosol was notdetected until after reperfusion, and was inhibited by anoxic ischemia or antioxidantadministration during ischemia. Although DNA fragmentation was detected after I/R, no evidenceof Bax activation was detected. Over-expression of the anti-apoptotic protein Bcl-XL incardiomyocytes did not confer protection against I/R-induced cell death. Moreover, murineembryonic fibroblasts with genetic depletion of Bax and Bak, or over-expression of Bcl-XL, failedto show protection against I/R. These findings indicate that mitochondrial ROS during ischemiatriggers mPTP activation, mitochondrial depolarization, and cell death during reperfusion througha Bax/Bak-independent cell death pathway. Therefore, mitochondrial apoptosis appears torepresent a redundant death pathway in this model of simulated I/R.

© 2010 Elsevier B.V. All rights reserved.Correspondence: Paul T. Schumacker, Ph.D., Department of Pediatrics, Northwestern University Feinberg School of Medicine, 310East Superior St., Morton Bldg. 4-685, Chicago, IL 60611, Tel. 312 503-1475, Fax 312 503-1181, [email protected]'s Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to ourcustomers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review ofthe resulting proof before it is published in its final citable form. Please note that during the production process errors may bediscovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

NIH Public AccessAuthor ManuscriptBiochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

Published in final edited form as:Biochim Biophys Acta. 2011 July ; 1813(7): 1382–1394. doi:10.1016/j.bbamcr.2010.12.008.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

INTRODUCTIONTissue ischemia is characterized by severe hypoxia, acidosis, energy depletion and celldeath. Although timely restoration of blood flow is currently the most effective means ofminimizing ischemic injury, reperfusion of ischemic tissue triggers a paradoxical increase incell death [6]. Excessive oxidant stress is well accepted as an important component ofischemia-reperfusion (I/R) injury [5,6]. ROS production begins early in ischemia, and isfollowed by a large burst of oxidant stress during the first few minutes of reperfusion[2,30,41,46]. Potential sources of ROS include mitochondria, NAD(P)H oxidases, nitricoxide (NO) synthase, and xanthine oxidase, and the critical targets of oxidant stress mayinclude proteins, membrane lipids, and DNA [7,14,25,37]. Although many of the detailsregarding the sources and targets of oxidant stress during I/R are not known, a consensusregarding the importance of ROS in I/R injury has developed, based on studies showing thatcells are protected during I/R by pretreatment with antioxidants or by over-expression ofantioxidant enzymes [8,9,16,34].

Other reports have implicated mitochondrial apoptosis in the cell death triggered by I/R.This highly conserved process can be initiated by the activation of BH3-containing pro-death proteins in response to a variety of stimuli including hypoxia, nutrient deprivation,ROS and DNA damage [20]. Apoptotic triggers cause the Bcl-2 family members Bax andBak to translocate to the mitochondria and trigger the release of cytochrome c to the cytosol[19]. This leads to the activation of caspases and subsequent cell death through an ATP-dependent pathway [42,49]. Consistent with that model, transgenic mice that over-expressBcl-2, a Bax/Bak suppressor, or that are deficient in the Bax protein, have been reported toexhibit smaller infarcts compared with wild-type mice subjected to I/R [23,26,43]. Otherstudies report that pharmacological inhibition of caspases is protective against I/R injury invitro [36,40] although there is debate as to whether caspase inhibition can prevent cell deathfollowing cytochrome c release to the cytosol, due to the existence of redundant effectorpathways of apoptosis that act downstream of the mitochondria.

Although oxidant stress and apoptosis have both been implicated in I/R-induced cell death,the relationship between these processes is not clearly established. One possibility is thatROS generation during I/R leads to the BH3-dependent activation of mitochondrialapoptosis, cytochrome c release, and caspase-mediated cell death. Alternatively, oxidantstress generated during I/R could trigger opening of the mitochondrial permeabilitytransition pore, leading to cytochrome c release to the cytosol, bioenergetic failure, and celldeath by a necrotic rather than an apoptotic pathway [11,12]. In the former case cytochromec (cyt-c) release would mediate cell death, whereas in the latter case the release of cyt-cwould represent only a marker of lethal cell damage. The present study examined therelationship between oxidant stress during I/R and mitochondria-mediated cell death, using amodel of simulated I/R in which the cells were superfused with hypoxic, hypercarbic andacidotic medium lacking glucose. The data show that oxidant stress generated duringsimulated ischemia triggers oscillation of the mitochondrial permeability transition pore, andthat this response precedes the irreversible opening of the pore and cyt-c release duringreperfusion. Although cytochrome c release during reperfusion leads to caspase activation,interventions that inhibit BH3-dependent apoptosis do not protect against cell death in thismodel. These findings indicate that apoptosis becomes activated in simulated I/R, but it isnot required for subsequent cell death.

Loor et al. Page 2

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

METHODSCell culture and perfusion system

Embryonic chick cardiomyocytes were prepared as previously described and grown at adensity of 1 × 105 on 25 mm glass cover slips or 6 well plates in a humidified incubator[47]. Experiments were performed on spontaneously contracting cells at 3–5 days afterisolation. Primary mouse embryonic fibroblasts (MEFs) derived from wild type and Bax-Bak double knockout mice were immortalized with a plasmid containing SV40 genomicDNA. Immortalized wild-type MEFs were also used to develop a stable cell line over-expressing Bcl-XL. MEFs were maintained in DMEM supplemented with 10% heat-inactivated fetal bovine serum, L-glutamine (200 mmol/L) and antibiotics (pen/strep).Embryonic stem cells devoid of cytochrome c (cyt-c−/− ES cells) were cultured underconditions that compensate for their defect in mitochondrial respiration using mediasupplemented with uridine and pyruvate as previously described [31]. Wild-type embryonicstem cells containing cytochrome c were cultured in parallel with null (cyt-c−/−) ES cellsand used as controls.

Simulated ischemia-reperfusionSimulated I/R was achieved in a flow through chamber as previously described [47]. Thechamber was comprised of two cover slips separated by a stainless steel spacer ring, with thecultured cells on the lower cover slip. The space between the cover slips was perfused withbalanced salt solutions (BSS) at a flow rate of 0.5 ml/min. All solutions were equilibrated bybubbling vigorously with calibrated gases in a heated, water jacketed reservoir (RadnotiGlass Co.). The reservoir was connected to the flow-through chamber with stainless steeltubing to minimize entry of ambient oxygen. The chamber was mounted on a heated stage(37° C) on an inverted microscope (Nikon). During baseline conditions the cells weresuperfused for 20 min with BSS (NaCl 117 mmol/L, KCl 4 mmol/L, NaHCO3 18 mmol/L,MgSO4 0.76 mmol/L, NaH2PO4 1 mmol/L, CaCl2 1.21 mmol/L and glucose 5.6 mmol/L)bubbled with a normoxic gas mixture (5% CO2/21% O2/74% N2). The same solution wasused during simulated reperfusion. Simulated ischemia was achieved by perfusing thechamber with ischemic media (NaCl 108.9 mmol/L, KCl 8 mmol/L, NaHCO3 21.4 mmol/L,MgSO4 0.76 mmol/L, NaH2PO4 1 mmol/L, CaCl2 1.21 mmol/L) devoid of glucose andbubbled with 20% CO2/0 % O2/80% N2 (pH 6.8). This resulted in an extracellular PO2 ofapproximately 7 mmHg within 10 min of hypoxia as determined by a porphyrin-basedphosphorescence quenching assay [33].

Cell ViabilityCell death was assessed using propidium iodide (PI, 5 µmol/L) in the perfusate. This dye isexcluded from live cells, but enters upon disruption of the plasma membrane. Fluorescenceimages were obtained using a 10× objective (excitation: 555 nm, emission: 605 nm) atbaseline, at the end of ischemia, and at 1, 2 and 3 hrs into reperfusion. Following the 3 hrmeasurement, cells were permeabilized with digitonin (300 mmol/L) for 45 min. Cell deathduring the experiment was calculated as the percent of the post-digitonin value. In mostexperiments, cell death was assessed in 3 separate regions per cover slip using a computer-controlled motorized stage that returned to specific X-Y positions for repeatedmeasurements. An LDH cytotoxicity assay (BioVision) was used to assess cell viability inresponse to staurosporine treatment and serum withdrawal. LDH is released into theextracellular space upon loss of plasma membrane integrity. The LDH assay was usedaccording to the manufacturer’s specifications with minor modifications. Supernatants werecollected after treatment with staurosporine (1 µmol/L in 1.5 ml of serum-free media) orprolonged serum withdrawal. Cells were incubated in Triton-X (1.5 ml of 2% in serum-freemedia) for 30 min. Lysates were collected and centrifuged at 11,000 RPM for 10 min.

Loor et al. Page 3

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Samples were mixed with the reaction mixture and incubated for 30 min in a 96 well plate,at which point the absorbance was measured at 490 nm. Cytotoxicity was calculated as theabsorbance in the supernatant divided by the sum of absorbance in the supernatant andlysates.

Mitochondrial Membrane PotentialΔΨm was assessed with tetramethylrhodamine ethyl ester (TMRE) under non-quenchingconditions as previously described [30]. Cells were loaded with TMRE (100 nmol/L) at 37°C for 30 min. The media was then replaced with a maintenance concentration of TMRE (25nmol/L). Mitochondrial fluorescence intensity was assessed using excitation at 555 nm andemission at 605 nm. Images were obtained during baseline and at periodic intervalsthereafter (10× objective). Intensity values were corrected for background. Stage positionswere programmed using data acquisition software (Metamorph, Universal Imaging)allowing repeated images to be obtained from the same fields of cells throughout eachexperiment.

DNA Fragmentation assayCardiomyocytes (2.1 ×106) were disrupted using lysis buffer, and the DNA was extractedusing phenol:chloroform after 2 hrs of proteinase K digestion at 50° C. Genomic DNA (1.5µg per lane) was treated with 100 µg /ml of RNase A and run on a 2.0% agarose gelcontaining 0.06 mg/ml of ethidium bromide. Apoptotic DNA fragments consist of multimersof 180–200 base pairs and appear as a DNA ladder. DNA of non-apoptotic populations ofcells has a high molecular weight and does not migrate far into the gel.

Immunofluorescence labeling of cytochrome c and BaxTo determine subcellular localization of cytochrome c, both treated and untreated cells werefixed in 3% paraformaldehyde with 0.02% glutaraldehyde in PBS for 15 min andsubsequently permeabilized with ice-cold methanol. Cells were incubated in 0.5 mg/mlsodium borohydride (Sigma–Aldrich) solution, blocked for 30 min in PBS containing 1%goat serum, and incubated overnight at 4° C with 1:100 dilution of mouse anti-cytochrome cantibody (BD Bioscience) or mouse anti-Bax (BD Bioscience). Cells were then incubatedwith a fluorescent secondary antibody (Alexa Fluor 488-labeled anti-mouse IgG, (1:100))for 1 hr at room temperature and visualized under confocal fluorescent microscopy using a40× objective. Cells with a normal mitochondrial localization of cytochrome c exhibit afluorescence pattern that is punctuate in appearance, while cells that have undergoneredistribution of cytochrome c to the cytosol exhibit a more diffuse pattern of fluorescence.Conversely, cells with a normal cytosolic localization of Bax exhibit a fluorescence patternthat is diffuse in appearance while cells that have undergone Bax translocation to themitochondria exhibit a punctuate pattern of fluorescence. Diffuse cells were enumeratedindependently by two blinded investigators who examined three separate visual fields eachcontaining 50–60 cells. Values were expressed as percent of total cells. In separateexperiments, cardiomyocytes were transduced for 48 hrs with an adenovirus expressing aGreen Fluorescent Protein mutant (roGFP) targeted to the mitochondrial matrix using thetargeting sequence from Cytochrome oxidase subunit IV. This targeting results in expressionof the protein exclusively in the matrix compartment [48]. Cells were then subjected to I/R.They were then fixed, immunostained for cytochrome c or Bax, and were imaged using alaser scanning confocal microscope (Zeiss, 60X) to assess the extent of co-localization.

ReagentsAdenoviruses expressing catalase or mitochondria-targeted catalase were obtained from Drs.Bai and Cederbaum [3]. Adenoviruses expressing Cu,Zn-SOD (SOD I) or Mn-SOD (SOD

Loor et al. Page 4

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

II) were obtained from the Gene Transfer Vector Core Facility at the University of Iowa[50,51]. Viral doses were established in preliminary studies evaluating expression as afunction of viral dosage (multiplicity of infection, MOI, data not shown). The effectiveconcentrations of the chemical antioxidants were reported previously [29,30,41,44].Embryonic stem cells with homozygous deletion of cytochrome c were obtained fromATCC. TMRE, Calcein-AM, and Alexa-Fluor secondary antibodies were obtained fromMolecular Probes-Invitrogen. Digitonin was obtained from Aldrich. Other chemicals wereobtained from Sigma.

Statistical AnalysisData were analyzed by ANOVA, and are reported as mean values ± SEM. When astatistically significant effect was detected, individual differences were explored using aNewman-Keuls post hoc analysis. Statistical significance was determined at the 0.05 level.

RESULTSCellular response to simulated I/R

Cardiomyocytes on cover slips were subjected to simulated I/R in a flow-through chamberas previously reported [41]. Phase contrast images revealed evidence of cellular swelling,membrane irregularity, and increased PI uptake during reperfusion (Fig 1A). To clarify therole of oxidant stress in I/R, antioxidant enzymes targeted to specific intracellularcompartments were expressed in cultured cardiomyocytes using adenoviral vectors, andexamined in terms of their ability to protect against I/R-induced cell death. The adenoviraltiters needed for optimal expression of catalase, catalase targeted to the mitochondrialmatrix, Cu,Zn-SOD or Mn-SOD targeted to the mitochondrial matrix, were determined at 48hr for each adenovirus by Western blotting (Fig. 1B). Cells transduced with theseadenoviruses or an empty viral vector (Y5) were incubated for 48 hrs prior to simulated I/R.Paired control experiments were performed on the same day in order to minimize the effectsof any differences due to cell age or isolation batch. Negligible cell death was present atbaseline, and no significant increases in cell death were detected during ischemia prior toreperfusion (Fig. 1C). During 3 hrs of reperfusion, cell death increased significantly in cellsexpressing catalase, mitochondrial catalase or Cu,Zn-SOD, whereas cells over-expressingMn-SOD showed dramatic protection (9±3%) compared to Y5 transduced controls (54±4%).These findings indicate that mitochondrial superoxide, which would have been scavengedby Mn-SOD, must have been a critical contributor to the I/R-induced cell death in thecardiomyocytes.

Protection by antioxidant compoundsAddition of the antioxidant N-acetyl-L-cysteine (NAC), 2-mercaptoproprionyl glycine (2-MPG) or reduced glutathione peptide (GSH) to the perfusate throughout baseline andischemia, but not during reperfusion, resulted in significant protection from I/R-induced celldeath (Fig 2A). Addition of the O2 scavenger Oxyrase to the ischemia buffer lowers the O2tension to anoxic levels, thereby preventing ROS generation during ischemia. As seenpreviously [41], Oxyrase administration during ischemia confers significant protectionagainst later cell death during reperfusion (Fig. 2A). However, administration of NAC+2-MPG only during reperfusion did not result in significant protection compared with controlcells. Mitochondrial potential (ΔΨm), assessed using TMRE fluorescence, decreasedsignificantly during ischemia, and no recovery of TRME fluorescence was detected duringreperfusion (Fig. 2B). Similar responses have been reported previously in this model [30].

Loor et al. Page 5

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Assessment of mitochondrial oxidant stress during ischemiaTo assess oxidant stress in the mitochondrial matrix during ischemia, cells were transducedwith an adenovirus expressing mito-roGFP, a ratiometric redox-sensitive sensor [15]. Thisprotein was targeted to the mitochondrial matrix by appending the mitochondrial targetingsequence from cytochrome oxidase subunit IV. The specificity of expression was confirmedby co-localization studies comparing the distribution of mito-roGFP fluorescence andcytochrome c immunostaining, by confocal microscopy (Fig. 3A). Cardiomyocytesexpressing mito-roGFP exhibited partial oxidation of the sensor under baseline conditions(Fig. 3B). During simulated ischemia, a significant increase in oxidation was detected,which was abrogated by continuous treatment with NAC, or by anoxic ischemia. Mockischemia, wherein cells were superfused with ischemic buffer equilibrated with 20% CO2and 21% O2 (no hypoxia), was included to control for possible effects arising from the lowpH conditions during ischemia. No significant change in the mito-roGFP oxidation state wasdetected during mock ischemia, confirming that the changes observed during simulatedischemia were not an artifact arising from low pH conditions. The percent oxidation of themito-roGFP protein during ischemia was determined by calibrating the sensor at the end ofthe ischemia challenge. This was achieved by perfusing the cells with media containingdithiothreitol (1 mM), followed by t-butyl hydroperoxide (1 mM) in order to fully reduceand fully oxidize the protein, respectively (Fig. 3C). To confirm the generation of oxidantstress in the mitochondrial matrix during simulated ischemia, cells were loaded with Mito-Sox Red, a cationic probe that distributes to the mitochondrial matrix by virtue of thetransmembrane potential (Fig. 3D). Oxidation of this probe was detected during simulatedischemia, but that response was significantly attenuated by NAC administration or by Mn-SOD over-expression (Fig. 3E). Collectively, these findings reveal that simulated ischemiais associated with a significant increase in oxidant stress within the mitochondrial matrix.

Alterations in mitochondrial morphology induced by ischemic oxidant stressCells expressing mito-roGFP exhibited a reticular fluorescence pattern that was concentratedin the perinuclear region. During ischemia this pattern changed, as the fluorescence becamemore punctate in nature (Fig. 4A). Adult mouse ventricular muscle (400 µm slice) washarvested, rapidly chilled to prevent ischemic damage, fixed, and examined by transmissionelectron microscopy. Another slice was subjected to simulated ischemia in the flow-thoughchamber (30 min), followed by rapid cooling and fixation to permit analysis of theultrastructural changes associated with ischemia in the absence of reperfusion. In the normalventricular slice, cardiac muscle fibers in both longitudinal and cross sectional orientationsexhibited normal ultrastructure (Fig. 4B, left). Bundles of myofibrils arranged along thelength of each muscle fiber were interspersed with numerous mitochondria. Ventricularslices subjected to ischemia without reperfusion showed morphological changes inmitochondria that included rounding and blebbing of the mitochondrial outer membrane(Fig. 4B, right). Mitochondria were disorganized and dispersed, in comparison to the tightlypacked organization in control slices. To determine the role of ischemic oxidant stress in thealteration of mitochondrial morphology, the fraction of cells exhibiting fragmentation in thefluorescent labeling studies (Fig. 4A) was quantified (Fig. 4C). Administration of NACduring ischemia, or scavenging of residual O2 during ischemia with Oxyrase (anoxicischemia), significantly attenuated the fragmented mitochondrial appearance. These findingsimplicate ischemic oxidant stress as the cause of these changes in mitochondrialmorphology.

Role of ischemic oxidant stress in regulating the mitochondrial permeability transitionpore

Opening of the mitochondrial permeability transition pore (mPTP) at reperfusion has beenimplicated in causing cyt-c redistribution to the cytosol during I/R [4,11,27]. To determine

Loor et al. Page 6

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

whether activation of the pore is responsible for the depolarization during ischemia,mitochondria were loaded with calcein-AM as previously described [38]. After quenchingthe cytosolic fluorescence with cobalt chloride, a pattern of mitochondrial fluorescence wasobserved in the cells (Fig. 5A), that remained stable during baseline. During ischemia, themitochondrial fluorescence pattern shifted to a diffuse cytosolic distribution, which wasassociated with a significant increase in cellular fluorescence intensity (Fig. 5A,B),indicating escape of calcein from the mitochondrial matrix to the cytosol. Addition of NACor 2-MPG during baseline and ischemia abolished this response, as did anoxic ischemiaachieved using Oxyrase to scavenge O2 from the ischemia buffer. These findings implicateischemic oxidant stress in the activation of the mPTP during simulated ischemia.

To further explore the role of mitochondrial oxidant stress in the activation of the mPTP,cardiomyocytes were transduced with adenoviruses to express mitochondria-targetedcatalase, Mn-SOD, or mitochondrial phospholipid hydroperoxide glutathione peroxidase(mito-PHGPx). The increase in cytosolic calcein fluorescence during ischemia wasabrogated by mito-PHGPx and by Mn-SOD expression, but not by mito-catalase or byinfection with the empty Y5 virus (Fig. 5C). Expression of mito-PHGPx was confirmed byWestern blotting. Also, mito-PHGPx over-expression conferred protection against cell deathin this model (Fig. 5D).

Relationship between ischemia and reperfusion in terms of cytochrome c release during I/R

The release of cytochrome c from the intermembrane space to the cytosol represents athreshold event in the activation of mitochondrial apoptosis. Cardiomyocytesimmunostained for cytochrome c under baseline conditions demonstrated a punctate patternof fluorescence in confocal images, which exhibited significant co-localization withmitochondria-targeted roGFP, consistent with mitochondrial localization (Fig. 6A). Duringsimulated ischemia, no significant increase in cells exhibiting diffuse cyt-c staining wasdetected (Fig. 6B). However, a significant increase in the number of cells exhibiting adiffuse pattern of fluorescence was detected during reperfusion, which was mirrored in thecells treated with the apoptosis-inducing agent staurosporine. Treatment of cardiomyocyteswith NAC during baseline and ischemia, or with Oxyrase to scavenge O2 during ischemia,resulted in a significant decrease in the extent of cyt-c redistribution during reperfusion.These findings indicate that, despite release of calcein during the ischemia phase, cyt-credistribution does not occur until after reperfusion. Moreover, ischemic oxidant stress isrequired for both the release of calcein during ischemia, and the release of cyt-c duringreperfusion in this model.

Role of mitochondrial apoptosis in simulated I/RTo clarify the requirement for apoptosis in the response to I/R, we examined the role of BHdomain-containing proteins in the regulation of cell death. Cardiomyocytes were transducedwith an adenovirus to induce over-expression of the antiapoptotic protein Bcl-XL. WhileBcl-XL over-expression conferred significant protection against cell death induced bystaurosporine (data not shown), it did not confer protection against simulated I/R (Fig. 7A).Simulated I/R was not associated with a translocation of Bax from the cytosol to themitochondria, as evidenced by a lack of co-localization of mitochondria-targeted roGFP andactivated Bax (Fig. 7B). Similarly, there was no increase in the percentage of total cellsexhibiting a punctuate pattern of Bax immunostaining in I/R (Fig. 7C), indicating an absenceof Bax oligomerization. However, DNA harvested from cardiomyocytes subjected to I/R didshow evidence of laddering (Fig. 7D), indicating that caspase activation had occurred. Theseresults indicate that the release of cyt-c during simulated I/R is not inhibited by anti-

Loor et al. Page 7

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

apoptotic proteins, and is not associated with Bax activation and translocation to themitochondria.

Role of Bax/Bak and cytochrome c release in I/R- induced cell deathTo further explore the requirement for apoptotic cell death in the setting of I/R,immortalized mouse embryonic fibroblast cells (MEFs) with genetic deletion of theproapoptotic proteins Bax and Bak (Bax−/−/Bak−/− double knockout cells) were comparedwith immortalized MEFs from isogenic wild type animals. Wild type immortalized MEFs,immortalized Bax/Bak double knockout MEFs, and immortalized MEFs with stable over-expression of Bcl-XL were grown on glass cover slips and subjected to serum withdrawal(48 hrs). Cell death in response to serum withdrawal was significantly attenuated in Bax/Bak-deficient cells, and by over-expression of Bcl-XL (Fig. 8A). By contrast, neither Bax/Bak deletion nor Bcl-XL over-expression conferred significant protection against simulatedI/R.

As cytochrome c is released to the cytosol during simulated reperfusion (Fig. 6), we soughtto clarify the significance of this event in I/R-induced cell death. Murine embryonic cells(ES cells) devoid of cytochrome c (cyt-c −/−) were obtained from ATCC. Previous studiesdemonstrated that these cells are resistant to various apoptotic stimuli including UVirradiation, staurosporine, and serum withdrawal [31]. Cytochrome c-deficient ES cells andmurine wild type (WT ES cells) were grown on cover slips and subjected to simulated I/R.However, deletion of cytochrome c did not confer protection against I/R (Fig. 8B). Thesefindings indicate that, although apoptosis is activated during simulated I/R as evidenced bycytochrome c release and DNA laddering, cell death does not require cytochrome c releasein this model.

Temporal relationship between PI uptake and cytochrome c releaseHeLa cells stably expressing a fusion protein consisting of cyt-c and GFP [18] were used toassess the temporal relationship between cytochrome c release to the cytosol and propidiumuptake, during simulated I/R. Co-localization studies using Mitotracker Red and GFPfluorescence demonstrated significant co-localization under confocal microscopy (Fig. 9A).During ischemia, there was minimal release of cyt-c and minimal uptake of PI in these cells(Fig. 9B). Within 1 hr after reperfusion, significant PI uptake was observed, indicative ofplasma membrane disruption. By contrast, a progressive increase in cyt-c release wasobserved during 3 hr of reperfusion. These findings suggest that cyt-c release is not theprimary mechanism driving cell death in this model.

DISCUSSIONOxidant stress and mitochondrial apoptosis have both been implicated in the cell deathobserved during I/R, but the relationship between these contributing factors is not clear. Asubstantial body of evidence indicates that ROS production occurs during I/R [46,52], andthat chemical antioxidants confer protection against I/R-induced cell death [30]. However,apoptosis also has been reported to be activated by I/R, and to be important for mediating I/R-induced cell death [23,43]. It is also conceivable that oxidant stress during I/R mediatescell death by activating a programmed cell death pathway [40]. We previously reported thatoxidant stress generated during simulated ischemia is required for cell death duringreperfusion [41]. The present study sought to examine how oxidant stress generated duringischemia contributes to cell death, and the relationship between ischemic oxidant stress andthe activation of mitochondrial apoptosis.

Loor et al. Page 8

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Ischemia causes oxidant stress in the mitochondrial matrix during simulated I/RWe utilized mito-roGFP, a ratiometric redox sensor, to detect protein thiol oxidation duringischemia in the mitochondrial matrix. During simulated ischemia, oxidation of this sensorincreased from less than 50% to more than 90%, indicating that mitochondrial ROSproduction increases paradoxically during ischemia, before experiencing the ROS burst thatoccurs upon reperfusion. Ischemic oxidant production was abrogated by chemicalantioxidants, and by scavenging all of the residual O2 from the ischemia media to preventROS generation, indicating that ROS were involved. Mitochondrial ROS production wasalso detected using Mito-SOX, a superoxide-sensitive cationic dye that accumulates inmitochondria. Oxidation of Mito-SOX was attenuated by antioxidants and by Mn-SODover-expression, indicating that superoxide is the initial oxidant generated in thatcompartment. A paradoxical increase in ROS production during ischemia was originallypredicted by Misra and Fridovich, who postulated that the increase in reduction state of themitochondrial electron carriers during hypoxia would promote ROS generation despite adecrease in the availability of O2 levels [35]. We previously detected such oxidant stress inthe cytosol of cardiomyocytes in our simulated I/R model [41]. The present finding extendsthat work by showing that oxidant stress increases in the matrix as well, suggesting thatischemic oxidants originate from the electron transport chain. It seems unlikely that each ofthe chemically diverse antioxidants was able to scavenge superoxide directly. More likely,some of these agents may have acted by protecting the redox status of proteins that were thetargets of superoxide attack.

Ischemic oxidant stress is critical for cell death in simulated I/RROS production during ischemia is required for cell death during reperfusion. Thisconclusion is based on the observation that chemical antioxidants given during the ischemiawere protective, as was anoxic ischemia achieved by O2 scavenging. These findings areconsistent with our previous work [41].

Ischemic oxidant stress causes a change in mitochondrial morphology, from a reticular to apunctate or fragmented appearance. In association with this morphological change,mitochondria release calcein from the matrix to the cytosol, a response that signals theformation of a large conductance pore in the mitochondrial membranes. The most likelysource of this leak is the mPTP, which has previously been implicated in the depolarizationof mitochondria during reperfusion after ischemia [10,13]. Antioxidants and anoxicischemia prevented the calcein leak, as did over-expression of mito-PHGPx. This protein istargeted to the mitochondrial membranes and is responsible for scavenging lipidhydroperoxides [17]. Over-expression of mito-PHGPx conferred protection against I/R-induced cell death, and it abrogated the ischemia-induced calcein leakage. These resultssuggest that mitochondrial oxidant stress is responsible for triggering the mPTP opening,which is responsible for the mitochondrial depolarization and cell death. A previous reportalso noted that mito-PHGPx over-expression was protective in I/R [24].

Protection against calcein leak and cell death was also conferred by Mn-SOD, whichsuggests that superoxide production in the mitochondrial matrix is critical for regulating celldeath in this model. This finding is consistent with studies showing that protection fromhepatic and cardiac I/R injury in vivo is conferred by over-expression of Mn-SOD in mice[8]. Loss of this critical antioxidant in mice causes a severe dilated cardiomyopathy andneonatal lethality [28,32], underscoring the potential toxicity of intra-mitochondrialsuperoxide. Recently, Ago et al. reported that NOX4 may be expressed in mitochondria, andthat over-expression of NOX4 can lead to cell death in aging and hypertrophic hearts [1].While the electron transport chain has been regarded as the apparent source of superoxide inischemia, those findings raise the question of whether NOX4 contributes importantly to the

Loor et al. Page 9

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

generation of superoxide during ischemia. In either case, superoxide appears to be criticalfor the cell damage, as catalase expression in either the cytosol or in the mitochondrialmatrix failed to provide protection, which indicates that H2O2 is a less important factor inthis model. Although there is little doubt that excessive H2O2 can potentially injure a cell,our findings do suggest that superoxide in the mitochondrial matrix may act on differenttargets than does H2O2, with important consequences for the rapid initiation of cell deathpathways. Over-expression of Cu,Zn-SOD provided no significant protection in this model.Hydrogen peroxide generated in one intracellular compartment can diffuse throughmembranes to access other compartments. By contrast, superoxide generated in I/R cannoteasily cross membranes, which explains why Mn-SOD was protective in the I/R modelwhereas Cu,Zn-SOD was not.

Previous studies have suggested that mPTP opening only occurs after reperfusion in intactheart, as the low pH during the ischemic insult inhibits pore activation [11,21,22]. Althoughour cellular model of simulated ischemia includes hypercarbic acidosis, this did not preventactivation of the pore during the ischemia. Despite this difference, important similarities areseen between this model and the intact heart, including the ability to undergopreconditioning protection [45], involvement of ROS in the cell death [30], and thedevelopment of cell death after reperfusion [47]. Further studies are needed to determinewhether more severe acidosis would delay the onset of mPTP opening until after the start ofreperfusion.

Relationship between mPTP activation and cytochrome c releasePrevious studies of I/R have shown that mPTP opening at reperfusion is associated withcytochrome c release to the cytosol [11,12]. Our findings confirm that negligible cyt-crelease occurs before reperfusion, yet our results consistently show that calcein releaseoccurs in parallel with mitochondrial depolarization during ischemia, in an ROS-dependentmanner. One possibility is that mPTP activation during ischemia represents oscillation orflickering of the pore, whereas irreversible opening occurs at reperfusion. Our findings areconsistent with the mPTP oscillations reported by Di Lisa and colleagues, who concludedthat cell death was not triggered by these short mPTP openings [39]. The only differencebetween their findings and ours is that we observed release of both calcein and TMREduring this oscillation, whereas they observed release of calcein but not TMRM. In eithercase, our findings suggest that the ROS-mediated oscillation of the mPTP during ischemia isan important determinant of later reperfusion-induced cell death. We agree with theirconclusion that mPTP oscillation by itself is not lethal for the cell, based on our observationthat cyt-c release and cell death do not increase significantly until reperfusion, even whenthe ischemic insult is extensively prolonged.

Role of mitochondrial apoptosis in cell death following I/RSimulated I/R caused a redistribution of cyt-c from the intermembrane space to the cytosol.This, plus the associated DNA laddering, suggests that caspases were activated by thatevent. However, over-expression of Bcl-XL in cardiomyocytes and MEFs failed to induceprotection, and there was no evidence of Bax oligomerization in cardiomyocytes during I/R.Furthermore, MEFs derived from Bax/Bak double knockout embryos were not protectedagainst I/R, but did show protection against serum withdrawal, a potent stimulus foractivating mitochondrial apoptosis. These findings indicate that activation of BH-domainproteins is not required for I/R-induced cell death.

Activation of redundant death pathways mediated by mPTP oscillationAlthough cytochrome c release occurs in response to I/R, our data suggest that this is amarker rather than a mediator of cell death. First, genetic depletion of cytochrome c was not

Loor et al. Page 10

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

protective against cell death in this model. Second, when comparing the kinetics of PIuptake and the release of cyt-c, it is apparent that PI uptake occurs within the first hour afterreperfusion whereas cytochrome c release develops later. This suggests that cytochrome crelease is not required for the loss in plasma membrane integrity. Rather, events early inreperfusion, most likely the ROS burst, appear to be responsible for triggering both the lossof plasma membrane integrity and the later release of cytochrome c. Once the plasmamembrane integrity is lost, a cell becomes committed to death regardless of whether or notcytochrome c is later released. Our data suggest that low levels of oxidant stress duringischemia trigger oscillation of the mPTP, which causes cell death during reperfusion,possibly by initiating a lethal ROS burst. Although the specific mechanism is not fullyunderstood, we speculate that mPTP activation during ischemia may prime the cell forundergoing a burst in oxidant stress at reperfusion by releasing NAD(P)H from the matrix tothe cytosol. Di Lisa and colleagues demonstrated the feasibility of such a process byshowing that mitochondrial NAD+ release to the cytosol occurs in response to mPTPactivation [13]. A burst of ROS generation at reperfusion, resulting from the oxidation ofmitochondria-derived NADH in the cytosol, could then trigger plasma membrane disruptionand also cause irreversible mPTP opening and cytochrome c release. In that respect, oxidantstress during ischemia would represent a causative factor in the lethal ROS burst duringreperfusion, which would undermine cell survival by activating multiple redundant deathpathways. A fuller test of this hypothesis is warranted.

AcknowledgmentsThe authors thank Bhumika Sharma for general technical assistance, Danijela Dokic for assistance withimmunofluorescence studies, and Juan Li for the isolation of the cardiomyocytes. We also thank Dr. Douglas R.Green for providing the HeLa cell line with stable expression of cytochrome c-GFP, and Drs. J.X. Bai and A.I.Cederbaum for providing adenoviruses expressing catalase and mitochondrial catalase. This work was supported byHL35440, HL32646, HL66315, HL079650 and the American Heart Association, Midwest Affiliates.

References1. Ago T, Kuroda J, Pain J, Fu C, Li H, Sadoshima J. Upregulation of Nox4 by hypertrophic stimuli

promotes apoptosis and mitochondrial dysfunction in cardiac myocytes. Circ. Res. 2010; 106:1253–1264. [PubMed: 20185797]

2. Ambrosio G, Zweier JL, Duilio C, Kuppusamy P, Santoro G, Elia PP, Tritto I, Cirillo P, CondorelliM, Chiariello M. Evidence that mitochondrial respiration is a source of potentially toxic oxygen freeradicals in intact rabbit hearts subjected to ischemia and reflow. J. Biol. Chem. 1993; 268:18532–18541. [PubMed: 8395507]

3. Bai JX, Rodriguez AM, Melendez JA, Cederbaum AI. Overexpression of catalase in cytosolic ormitochondrial compartment protects HepG2 cells against oxidative injury. J. Biol. Chem. 1999;274:26217–26224. [PubMed: 10473575]

4. Bernardi P, Krauskopf A, Basso E, Petronilli V, Blachly-Dyson E, Di Lisa F, Forte MA. Themitochondrial permeability transition from in vitro artifact to disease target. FEBS J. 2006;273:2077–2099. [PubMed: 16649987]

5. Bolli R, Marban E. Molecular and cellular mechanisms of myocardial stunning. Physiol Rev. 1999;79:609–634. [PubMed: 10221990]

6. Braunwald E, Kloner RA. Myocardial reperfusion: a double-edged sword? J. Clin. Invest. 1985;76:1713–1719. [PubMed: 4056048]

7. Cave AC, Brewer AC, Narayanapanicker A, Ray R, Grieve DJ, Walker S, Shah AM. NADPHoxidases in cardiovascular health and disease. Antioxid. Redox. Signal. 2006; 8:691–728. [PubMed:16771662]

8. Chen Z, Siu B, Ho YS, Vincent R, Chua CC, Hamdy RC, Chua BH. Overexpression of MnSODprotects against myocardial ischemia/reperfusion injury in transgenic mice. J. Mol. Cell Cardiol.1998; 30:2281–2289. [PubMed: 9925365]

Loor et al. Page 11

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

9. Chen ZY, Oberley TD, Ho YS, Chua CC, Siu B, Hamdy RC, Epstein CJ, Chua BHL.Overexpression of CuZnSOD in coronary vascular cells attenuates myocardial ischemia/reperfusioninjury. Free Radic. Biol. Med. 2000; 29:589–596. [PubMed: 11033410]

10. Di Lisa F. Mitochondrial contribution in the progression of cardiac ischemic injury. IUBMB. Life.2001; 52:255–261. [PubMed: 11798040]

11. Di Lisa F, Bernardi P. Mitochondria and ischemia-reperfusion injury of the heart: fixing a hole.Cardiovasc. Res. 2006; 70:191–199. [PubMed: 16497286]

12. Di Lisa F, Menabo R, Canton M, Barile M, Bernardi P. Opening of the mitochondrial permeabilitytransition pore causes depletion of mitochondrial and cytosolic NAD+ and is a causative event inthe death of myocytes in postischemic reperfusion of the heart. J. Biol. Chem. 2001; 276:2571–2575. [PubMed: 11073947]

13. Di Lisa F, Menabo R, Canton M, Barile M, Bernardi P. Opening of the mitochondrial permeabilitytransition pore causes depletion of mitochondrial and cytosolic NAD+ and is a causative event inthe death of myocytes in postischemic reperfusion of the heart. J. Biol. Chem. 2001; 276:2571–2575. [PubMed: 11073947]

14. Dixon IM, Kaneko M, Hata T, Panagia V, Dhalla NS. Alterations in cardiac membrane Ca2+transport during oxidative stress. Mol. Cell Biochem. 1990; 99:125–133. [PubMed: 1962845]

15. Dooley CT, Dore TM, Hanson GT, Jackson WC, Remington SJ, Tsien RY. Imaging dynamicredox changes in mammalian cells with green fluorescent protein indicators. J. Biol. Chem. 2004;279:22284–22293. [PubMed: 14985369]

16. Fischer UM, Cox CS Jr, Allen SJ, Stewart RH, Mehlhorn U, Laine GA. The antioxidant N-acetylcysteine preserves myocardial function and diminishes oxidative stress after cardioplegicarrest. J. Thorac. Cardiovasc. Surg. 2003; 126:1483–1488. [PubMed: 14666023]

17. Godeas C, Sandri G, Panfili E. Distribution of phospholipid hydroperoxide glutathione peroxidase(PHGPx) in rat testis mitochondria. Biochim. Biophys. Acta. 1994; 1191:147–150. [PubMed:8155669]

18. Goldstein JC, Waterhouse NJ, Juin P, Evan GI, Green DR. The coordinate release of cytochrome cduring apoptosis is rapid, complete and kinetically invariant. Nat. Cell Biol. 2000; 2:156–162.[PubMed: 10707086]

19. Goping IS, Gross A, Lavoie JN, Nguyen M, Jemmerson R, Roth K, Korsmeyer SJ, Shore GC.Regulated targeting of BAX to mitochondria. J. Cell Biol. 1998; 143:207–215. [PubMed:9763432]

20. Green DR, Kroemer G. The pathophysiology of mitochondrial cell death. Science. 2004; 305:626–629. [PubMed: 15286356]

21. Halestrap AP. Calcium, mitochondria and reperfusion injury: a pore way to die. Biochem. Soc.Trans. 2006; 34:232–237. [PubMed: 16545083]

22. Halestrap AP, Kerr PM, Javadov S, Woodfield KY. Elucidating the molecular mechanism of thepermeability transition pore and its role in reperfusion injury of the heart. Biochim. Biophys. ActaBio. -Energetics. 1998; 1366:79–94.

23. Hochhauser E, Kivity S, Offen D, Maulik N, Otani H, Barhum Y, Pannet H, Shneyvays V,Shainberg A, Goldshtaub V, Tobar A, Vidne BA. Bax ablation protects against myocardialischemia-reperfusion injury in transgenic mice. Am. J. Physiol Heart Circ. Physiol. 2003;284:H2351–H2359. [PubMed: 12742833]

24. Hollander JM, Lin KM, Scott BT, Dillmann WH. Overexpression of PHGPx and HSP60/10protects against ischemia/reoxygenation injury. Free Radic. Biol. Med. 2003; 35:742–751.[PubMed: 14583338]

25. Hurd TR, Filipovska A, Costa NJ, Dahm CC, Murphy MP. Disulphide formation on mitochondrialprotein thiols. Biochem. Soc. Trans. 2005; 33:1390–1393. [PubMed: 16246126]

26. Imahashi K, Schneider MD, Steenbergen C, Murphy E. Transgenic expression of Bcl-2 modulatesenergy metabolism, prevents cytosolic acidification during ischemia, and reduces ischemia/reperfusion injury. Circ. Res. 2004; 95:734–741. [PubMed: 15345651]

27. Kim JS, He L, Lemasters JJ. Mitochondrial permeability transition: a common pathway to necrosisand apoptosis. Biochem. Biophys. Res. Commun. 2003; 304:463–470. [PubMed: 12729580]

Loor et al. Page 12

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

28. Lebovitz RM, Zhang H, Vogel H, Cartwright J Jr, Dionne L, Lu N, Huang S, Matzuk MM.Neurodegeneration, myocardial injury, and perinatal death in mitochondrial superoxide dismutase-deficient mice. Proc. Natl. Acad. Sci. U. S. A. 1996; 93:9782–9787. [PubMed: 8790408]

29. Lebuffe G, Schumacker PT, Shao ZH, Anderson T, Iwase H, Vanden Hoek TL. ROS and NOtrigger early preconditioning: relationship to mitochondrial KATP channel. Am. J. Physiol HeartCirc. Physiol. 2003; 284:H299–H308. [PubMed: 12388274]

30. Levraut J, Iwase H, Shao ZH, Vanden Hoek TL, Schumacker PT. Cell death during ischemia:relationship to mitochondrial depolarization and ROS generation. Am. J. Physiol Heart Circ.Physiol. 2003; 284:H549–H558. [PubMed: 12388276]

31. Li K, Li Y, Shelton JM, Richardson JA, Spencer E, Chen ZJ, Wang X, Williams RS. Cytochrome cdeficiency causes embryonic lethality and attenuates stress-induced apoptosis. Cell. 2000;101:389–399. [PubMed: 10830166]

32. Li Y, Huang TT, Carlson EJ, Melov S, Ursell PC, Olson JL, Noble LJ, Yoshimura MP, Berger C,Chan PH. Dilated cardiomyopathy and neonatal lethality in mutant mice lacking manganesesuperoxide dismutase. Nat. Genet. 1995; 11:376–381. [PubMed: 7493016]

33. Lo LW, Koch CJ, Wilson DF. Calibration of oxygen-dependent quenching of the phosphorescenceof Pd-meso-tetra (4-carboxyphenyl) porphine: a phosphor with general application for measuringoxygen concentration in biological systems. Analyt. Biochem. 1996; 236:153–160. [PubMed:8619481]

34. McDonald MC, Zacharowski K, Bowes J, Cuzzocrea S, Thiemermann C. Tempol reduces infarctsize in rodent models of regional myocardial ischemia and reperfusion. Free Radic. Biol. Med.1999; 27:493–503. [PubMed: 10490268]

35. Misra HP, Fridovich I. The univalent reduction of oxygen by reduced flavins and quinones. J. Biol.Chem. 1972; 247:188–192. [PubMed: 4401581]

36. Mueller TH, Kienle K, Beham A, Geissler EK, Jauch KW, Rentsch M. Caspase 3 inhibitionimproves survival and reduces early graft injury after ischemia and reperfusion in rat livertransplantation. Transplantation. 2004; 78:1267–1273. [PubMed: 15548962]

37. Ostadal P, Elmoselhi AB, Zdobnicka I, Lukas A, Elimban V, Dhalla NS. Role of oxidative stress inischemia-reperfusion-induced changes in Na+,K(+)-ATPase isoform expression in rat heart.Antioxid. Redox. Signal. 2004; 6:914–923. [PubMed: 15345151]

38. Petronilli V, Miotto G, Canton M, Brini M, Colonna R, Bernardi P, Di Lisa F. Transient and long-lasting openings of the mitochondrial permeability transition pore can be monitored directly inintact cells by changes in mitochondrial calcein fluorescence. Biophys. J. 1999; 76:725–734.[PubMed: 9929477]

39. Petronilli V, Penzo D, Scorrano L, Bernardi P, Di Lisa F. The mitochondrial permeabilitytransition, release of cytochrome c and cell death. Correlation with the duration of pore openingsin situ. J. Biol. Chem. 2001; 276:12030–12034. [PubMed: 11134038]

40. Qin YM, Hoek TLV, Wojcik K, Anderson T, Li CQ, Shao ZH, Becker LB, Hamann KJ. Caspase-dependent cytochrome c release and cell death in chick cardiomyocytes after simulated ischemia-reperfusion. Am. J. Physiol Heart Circ. Physiol. 2004; 286:H2280–H2286. [PubMed: 14975933]

41. Robin E, Guzy RD, Loor G, Iwase H, Waypa GB, Marks JD, Vanden Hoek TL, Schumacker PT.Oxidant stress during simulated ischemia primes cardiomyocytes for cell death during reperfusion.J. Biol. Chem. 2007; 282:19133–19143. [PubMed: 17488710]

42. Scorrano L, Korsmeyer SJ. Mechanisms of cytochrome c release by proapoptotic BCL-2 familymembers. Biochem. Biophys. Res. Commun. 2003; 304:437–444. [PubMed: 12729577]

43. Tanaka M, Nakae S, Terry PD, Mokhtari GK, Gunawan F, Balsam LB, Kaneda H, Kofidis T, TsaoPS, Robbins RC. Cardiomyocyte-specific Bcl-2 overexpression attenuates ischemia-reperfusioninjury, immune response during acute rejection, and graft coronary artery disease. Blood. 2004;104:3789–3796. [PubMed: 15280201]

44. Vanden Hoek TL, Becker LB, Shao Z, Li C, Schumacker PT. Reactive oxygen species releasedfrom mitochondria during brief hypoxia induce preconditioning in cardiomyocytes. J. Biol. Chem.1998; 273:18092–18098. [PubMed: 9660766]

Loor et al. Page 13

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

45. Vanden Hoek TL, Becker LB, Shao ZH, Li CQ, Schumacker PT. Preconditioning incardiomyocytes protects by attenuating oxidant stress at reperfusion. Circ. Res. 2000; 86:541–548.[PubMed: 10720416]

46. Vanden Hoek TL, Li C, Shao Z, Schumacker PT, Becker LB. Significant levels of oxidants aregenerated by isolated cardiomyocytes during ischemia prior to reperfusion. J. Mol. Cell. Cardiol.1997; 29:2571–2583. [PubMed: 9299379]

47. Vanden Hoek TL, Shao Z, Li C, Zak R, Schumacker PT, Becker LB. Reperfusion injury in cardiacmyocytes after simulated ischemia. Am. J. Physiol. 1996; 270:H1334–H1341. [PubMed: 8967373]

48. Waypa GB, Marks JD, Guzy R, Mungai PT, Schriewer J, Dokic D, Schumacker PT. HypoxiaTriggers Subcellular Compartmental Redox Signaling in Vascular Smooth Muscle Cells. Circ.Res. 2009; 106:526–535. [PubMed: 20019331]

49. Wei MC, Zong WX, Cheng EHY, Lindsten T, Panoutsakopoulou V, Ross AJ, Roth KA,MacCregor GR, Thompson CB, Korsmeyer SJ. Proapoptotic BAX and BAK: A requisite gatewayto mitochondrial dysfunction and death. Science. 2001; 292:727–730. [PubMed: 11326099]

50. Zwacka RM, Dudus L, Epperly MW, Greenberger JS, Engelhardt JF. Redox gene therapy protectshuman IB-3 lung epithelial cells against ionizing radiation-induced apoptosis. Hum. Gene Ther.1998; 9:1381–1386. [PubMed: 9650622]

51. Zwacka RM, Zhou W, Zhang Y, Darby CJ, Dudus L, Halldorson J, Oberley L, Engelhardt JF.Redox gene therapy for ischemia/reperfusion injury of the liver reduces AP1 and NF-kappaBactivation. Nat. Med. 1998; 4:698–704. [PubMed: 9623979]

52. Zweier JL, Flaherty JT, Weisfeldt ML. Direct measurement of free radical generation followingreperfusion of ischemic myocardium. Proc. Natl. Acad. Sci. USA. 1987; 84:1404–1407. [PubMed:3029779]

Loor et al. Page 14

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Figure 1.Cell death in simulated ischemia-reperfusion. (A) Cardiomyocytes exposed to 1 hr ofsimulated ischemia followed by 3 hrs of reperfusion in a flow-through chamber. Phaseimages show increased cellular swelling, plasma membrane irregularity and PI uptake afterI/R. (B) Western blot analysis of adenovirus-transduced cardiomyocytes showing over-expression of catalase, mitochondria-targeted catalase, Cu,Zn-SOD and Mn-SOD after 48hrs. (C) Cardiomyocytes transduced with either Y5 (1000 MOI, n=10), cytosolic catalase(1000 MOI, n=6), mitochondria-targeted catalase (500 MOI, n=6), Cu,Zn-SOD (500 MOI,n=5) or Mn-SOD (20 MOI, n=5) for 48 hrs prior to simulated I/R (* p<.05 compared withcontrols).

Loor et al. Page 15

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Figure 2.Oxidant stress and mitochondrial depolarization in simulated ischemia-reperfusion. (A) N-acetyl-L-cysteine (NAC, 500 µmol/L) or 2-MPG (2-mercaptopropionyl glycine, 400 µmol/L) added prior to and during simulated ischemia decreased cell death after 3 hrs ofreperfusion. Similar protection was conferred by anoxic ischemia, achieved by addingOxyrase to the ischemic media, or by addition of reduced glutathione (GSH, 100 µmol/L)throughout simulated I/R. The combined use of NAC and 2-MPG, beginning 10 min prior toand throughout reperfusion, did not significantly decrease cell death during I/R (Controls,n=9; NAC, 2-MPG, Oxyrase, n=4; GSH, n=3; NAC/2-MPG during reperfusion n=4; * p<.05). (B) TMRE fluorescence in cardiomyocytes, to assess changes in mitochondrialpotential. Fluorescence intensities were monitored in control cells maintained undernormoxia and in cells challenged with simulated I/R. A significant decrease in TMREfluorescence was detected during 20–40 min of simulated ischemia. Values are expressed aspercent of baseline intensity (Controls, n=6; I/R, n=4; * p<.05). White bars indicate controls;dark bars indicate I/R.

Loor et al. Page 16

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Figure 3.Detection of oxidant stress in the mitochondrial matrix during simulated ischemia. (A)Isolated cardiomyocytes were transduced with an adenovirus to express mito-roGFP in themitochondrial matrix. Cytochrome c immunostaining and merged images demonstrate theexpected co-localization within the mitochondria. (B) Ratiometric measurements madeusing the mito-roGFP sensor showed a significant increase in the oxidation state within themitochondrial matrix during simulated ischemia. This oxidation response was attenuatedduring “mock ischemia” (20%CO2, 21%O2, 59% N2), anoxic ischemia (PO2 <0.5 mmHg),or ischemia in the presence of NAC (500µmol/L) (Mock ischemia, anoxic ischemia,ischemia + NAC, n=3; Controls, n=4; * p<.05). (C) Representative tracing of ratiometricmeasurements show the dynamics of the mito-roGFP sensor in cardiomyocytes duringsimulated ischemia, followed by calibration of the sensor using dithiothreitol (DTT, 1 mmol/L) followed by t-butyl hydroperoxide (TBH 1mmol/L). (D) Mito-Sox Red staining of themitochondrial matrix in cultured cardiomyocytes. (E). Increases in Mito-Sox Redfluorescence relative to baseline values were evident within 20 min of simulated ischemia.No significant increase was detected in control cells maintained under baseline conditionsfor 20 min (Control) or in cells exposed to ischemia in the presence of NAC (Ischemia +NAC). Over-expression of Mn-SOD, induced using an adenoviral vector, significantlyattenuated the rise in Mito-Sox fluorescence observed during simulated ischemia (Ischemia+ NAC) (Control, n=6; ischemia, n=8; ischemia + NAC, n=4; ischemia + Mn-SOD, n=3; *p<.05).

Loor et al. Page 17

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Figure 4.Mitochondrial fragmentation in cardiomyocytes during simulated ischemia. (A)Cardiomyocytes were transduced with mito-roGFP and visualized under fluorescentmicroscopy (excitation: 484nm, emission: 525 nm) to detect real time changes inmitochondrial morphology. Cells perfused under baseline conditions or subjected tosimulated ischemia in the presence of N-acetylcysteine (NAC, 500µmol/L) retained anormal reticular mitochondrial morphology. Cells exposed to ischemia in the absence ofNAC demonstrated a significantly fragmented mitochondrial morphology. (B)Representative electron micrographs of mouse ventricular tissue from 400 µm slicesmaintained under baseline conditions (left) or after 30 min simulated ischemia without

Loor et al. Page 18

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

reperfusion (right). Bars are 1 µm in length. (C) Cardiomyocytes expressing mito-roGFPwere subjected to baseline conditions or simulated ischemia with or without NAC or anoxiatreatment. Images were analyzed by 2 independent observers in order to quantify the numberof cells with fragmented mitochondria, as a percent of the total cells per high power field(50–70 cells from 3 distinct regions per cover slip). NAC and anoxic ischemia usingOxyrase both significantly reduced the percentage of cells with fragmented mitochondria.(n=3 each; * p<.05 compared with Ischemia – NAC).

Loor et al. Page 19

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Figure 5.Monitoring the opening of the mitochondrial permeability transition pore (mPTP) duringsimulated ischemia using mitochondrial calcein fluorescence. (A) Cardiomyocytes wereloaded with calcein dye (1 mmol/L) in the presence of cobalt chloride (CoCl2 1 mmol/L) for30 min and then studied in the flow-through chamber. During baseline perfusion amitochondrial pattern of calcein fluorescence was observed. Within 16 min after beginningsimulated ischemia, a rapid and profound release of calcein to the cytosol was observed. (B)A significant increase in calcein fluorescence intensity was detected during simulatedischemia. This increase was significantly attenuated by NAC (500 µmol/L) or 2-MPG (400µmol/L) administration prior to and during ischemia. Anoxic ischemia using Oxyrase alsoprevented calcein release (2-MPG, n=3; controls, NAC, anoxic ischemia n=4). (C)Adenoviral over-expression of mitochondrial phospholipid hydroperoxide glutathioneperoxidase (mito-PHGPx, 1500 MOI) or manganese superoxide dismutase (Mn-SOD, 100MOI) conferred significant protection against calcein escape from mitochondria duringischemia. Mitochondrial-targeted catalase (Mito-cat, 500 MOI) did not attenuate the rise incytosolic calcein fluorescence (Controls, mito-PHGPx, n=5; Mn-SOD, n=6, Mito-cat, n=4).(D) Over-expression of mito-PHGPx also conferred significant protection against cell deathafter 1 hr of ischemia and 3 hrs of reperfusion, compared with cells transduced with Y5empty virus (n=5; *p<0.05).

Loor et al. Page 20

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Figure 6.Release of cytochrome c during simulated I/R. (A). Cardiomyocytes were transduced withmito-roGFP for 48 hrs to identify the mitochondria. Cells were fixed at baseline, after 45minutes of ischemia, or after 2 hrs of reperfusion. Cells were then immunostained forcytochrome c to assess the degree of co-localization with the mito-roGFP sensor. Significantredistribution of cytochrome c to the cytosol was noted after 2 hrs of reperfusion, while onlytrace levels of redistribution were detected during ischemia (Mag. 60X). (B) Cells showingredistribution of cytochrome c were enumerated and expressed as a percent of the total cellpopulation. Staurosporine (STS) caused a significant increase in the percentage of cells withcytochrome c release. A similar increase was seen in cells subjected to simulated I/R.Cytochrome c release was attenuated in cells treated with NAC prior to and during ischemia,and in cells challenged with anoxic ischemia using Oxyrase. No significant difference atbaseline or after (base, ischemia, ischemia/reperfusion, STS, n=4; NAC, anoxic ischemia,n=3; * p<0.05).

Loor et al. Page 21

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Figure 7.Mitochondrial apoptosis in staurosporine-treated and I/R-challenged cardiomyocytes. (A)Bcl-XL over-expression with an adenovirus (1000 MOI) for 36 hrs in cardiomyocytes wasnot protective against simulated I/R compared with cells transduced with empty virus (Y5,1000 MOI)(n=4). (B) Cardiomyocytes transduced with an adenovirus to express mito-roGFPfor 48 hrs, in order to identify mitochondria. Cell were then subjected to simulated I/R andimmunostained for Bax, in order to assess oligomerization as evidenced by co-localizationwith the mitochondrial fluorescence, by confocal microscopy. I/R did not induce significantBax activation, as seen by the lack of co-localization in this representative image (Mag.60X). (C) No significant increase in the number of cells exhibiting a mitochondriallocalization of Bax after simulated I/R was detected, indicating that I/R was not a significantstimulus for the activation of mitochondrial apoptosis (n=4). (D) Representative gel showingthe presence of DNA fragmentation after treatment with I/R (lanes: 1- Ladder, 2- Baseline,3- Ischemia, 4-Ischemia+Reperfusion (I/R), 5-Staurosporine (STS)).

Loor et al. Page 22

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Figure 8.Cell death in wild-type MEFs, Bax/Bak double knockout (KO) MEFs, Bcl-XL over-expressing MEFs, wild-type ES cells and cytochrome c deficient ES cells. (A) Bcl-XL over-expression and genetic deletion of Bax/Bak conferred protection against growth factorwithdrawal for 72 hrs as assessed by LDH cytotoxicity assays. However, in cells subjectedto 30 min of simulated ischemia followed by 3 hrs reperfusion there was no significantdifference in cell death between groups as determined by PI uptake (n=5; * p<.05 comparedwith controls). (B) Murine embryonic stem (ES) cells lacking cytochrome c (cyt-c−/−) werenot significantly protected against 60 min of simulated ischemia followed by 3 hrs ofreperfusion compared with wild-type ES cells as shown by analysis of PI uptake (n=4).

Loor et al. Page 23

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Figure 9.Cytochrome c release and PI uptake during simulated I/R in HeLa cells with stableexpression of cytochrome c-GFP. (A) Cytochrome c-GFP- expressing HeLa cells wereloaded with Mitotracker Red (100 nM) for 30 min to label mitochondria. Merged confocalimage shows co-localization of the cytochrome c-GFP fluorescence and Mitotracker Red(Mag. 60X). (B) HeLa cells challenged with simulated I/R. Live cell images were obtainedduring baseline, ischemia, and during reperfusion. Rapid and extensive dissipation ofpunctuate cyt-c-GFP fluorescence was observed within 2 hrs of reperfusion. In separateexperiments, propidium iodide was used to detect the extent of cell death in the HeLa cellsduring simulated I/R. Significantly greater cell death within 1 hr of reperfusion was

Loor et al. Page 24

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

observed using PI uptake compared with cellular cytochrome c redistribution. Thepercentage of cells demonstrating cytochrome c release approximated the percentage of cellsexhibiting PI uptake by 2 hrs of reperfusion. (Cyt-c redistribution and PI uptake, n=4;*p<0.05 compared to cyt-c release at baseline; **p<0.05 compared to PI uptake at baseline;† p<0.05, PI uptake compared to percent of cells with diffuse cyt-c staining).

Loor et al. Page 25

Biochim Biophys Acta. Author manuscript; available in PMC 2012 July 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript