Anda di halaman 1dari 10

Am J Physiol Heart Circ Physiol 284: H549H558, 2003.

First published October 10, 2002; 10.1152/ajpheart.00708.2002.

Cell death during ischemia: relationship to mitochondrial


depolarization and ROS generation
JACQUES LEVRAUT, HIROTARO IWASE, Z.-H. SHAO,
TERRY L. VANDEN HOEK, AND PAUL T. SCHUMACKER
Pulmonary and Critical Care Medicine, University of Chicago, Chicago, Illinois 60637
Submitted 22 August 2002; accepted in final form 7 October 2002

Levraut, Jacques, Hirotaro Iwase, Z.-H. Shao, Terry we observed (32, 33) an increase in ROS generation
L. Vanden Hoek, and Paul T. Schumacker. Cell death during ischemia followed by a large burst of oxidant

Downloaded from http://ajpheart.physiology.org/ by 10.220.33.2 on September 18, 2016


during ischemia: relationship to mitochondrial depolariza- production during the first few minutes after reoxygen-
tion and ROS generation. Am J Physiol Heart Circ Physiol ation. In that model, antioxidants were more protective
284: H549H558, 2003. First published October 10, 2002; when given throughout the experiment than when
10.1152/ajpheart.00708.2002.Ischemia-reperfusion injury
given only at reperfusion, which supports the idea that
induces cell death, but the responsible mechanisms are not
understood. This study examined mitochondrial depolariza- oxidants generated during the ischemic phase contrib-
tion and cell death during ischemia and reperfusion. Con- ute to cell injury and are important determinants of
tracting cardiomyocytes were subjected to 60-min ischemia cell survival and recovery of function (2, 35).
followed by 3-h reperfusion. Mitochondrial membrane poten- ROS generation cannot occur during ischemia unless
tial (m) was assessed with tetramethylrhodamine methyl some residual O2 is still present. Previous studies us-
ester. During ischemia, m decreased to 24 5.5% of ing cardiomyocytes revealed that trace levels of O2 are
baseline, but no recovery was evident during reperfusion. still detectable during simulated ischemia (PO2 57
Cell death assessed by Sytox Green was minimal during mmHg). During ischemia, indexes of oxidant stress
ischemia but averaged 66 7% after 3-h reperfusion. Cyclo- were attenuated by mitochondrial electron transport
sporin A, an inhibitor of mitochondrial permeability transi- inhibitors, suggesting that the ROS are generated by
tion, was not protective. However, pharmacological antioxi- mitochondria (2, 33, 34). Collectively, these observa-
dants attenuated the fall in m during ischemia and cell
tions support the notion that superoxide is generated
death after reperfusion and decreased lipid peroxidation as
assessed with C11-BODIPY. Cell death was also attenuated
during ischemia despite the conditions of low O2 con-
when residual O2 was scavenged from the perfusate, creating centration ([O2]) (11), and they suggest that these
anoxic ischemia. These results suggested that reactive oxy- oxidants may play an important role in determining
gen species (ROS) were important for the decrease in m cell survival during I/R.
during ischemia. Finally, 143B-0 osteosarcoma cells lacking Although previous studies indicate that oxidants
a mitochondrial electron transport chain failed to demon- generated during ischemia may contribute to cell dam-
strate a depletion of m during ischemia and were sig- age, the specific mechanism by which these ROS dis-
nificantly protected against cell death during reperfusion. rupt cellular function is not known. The present study
Collectively, these studies identify a central role for mito- sought to clarify the physiological consequences of ox-
chondrial ROS generation during ischemia in the mitochon- idants generated during ischemia before reperfusion.
drial depolarization and subsequent cell death induced by We hypothesized that oxidant stress generated at the
ischemia and reperfusion in this model. mitochondria during ischemia could contribute to a
reactive oxygen species; hypoxia; oxidants loss of mitochondrial membrane potential (m),
which in turn could contribute to the overall cellular
injury and survival.
REACTIVE OXYGEN SPECIES (ROS) have been implicated as
participants in the myocardial damage induced by is- MATERIALS AND METHODS
chemia-reperfusion (I/R) (1, 4, 9, 17, 21, 25). Most
studies have focused on the importance of oxidant Cell culture and perfusion system. Embryonic chick cardi-
stress generated during reperfusion, when a burst of omyocytes were prepared as previously described (35) and
were grown on glass coverslips in a humidified incubator.
ROS is generated after oxygen is reintroduced into the Experiments were performed on spontaneously contracting
system after a prolonged period of ischemia (32, 39). cells at 35 days after isolation, under controlled O2-CO2
However, growing evidence suggests that oxidant conditions at 37C on an inverted microscope. A flow-through
stress begins during ischemia before reperfusion. For chamber was created by clamping a stainless steel spacer
example, in cardiomyocytes subjected to simulated I/R, ring between two coverslips, allowing perfusion of the space

Address for reprint requests and other correspondence: P. T. Schu- The costs of publication of this article were defrayed in part by the
macker, Dept. of Medicine MC6026, The Univ. of Chicago, 5841 payment of page charges. The article must therefore be hereby
South Maryland Ave., Chicago, IL 60637 (E-mail: pschumac marked advertisement in accordance with 18 U.S.C. Section 1734
@medicine.bsd.uchicago.edu). solely to indicate this fact.

http://www.ajpheart.org 0363-6135/03 $5.00 Copyright 2003 the American Physiological Society H549
H550 MITOCHONDRIAL DEPOLARIZATION DURING ISCHEMIA

between with buffered salt solutions (BSS; 0.5 ml/min) equil- electron transport system and cannot generate ATP or ROS
ibrated with O2-CO2 gas mixtures in a water-jacketed col- in mitochondria. Cells were maintained in medium supple-
umn. Stainless steel tubing connecting the column to the mented with pyruvate (2 mM), uridine (50 g/ml), and 5-bro-
chamber prevented diffusive entry of ambient O2 through the mouridine (0.015 mg/ml). Loss of mitochondrial DNA was
tubing wall. confirmed by semiquantitative PCR and by loss of cell viabil-
I/R model. Cells were equilibrated for 30 min by superfu- ity when uridine supplements to the media were withdrawn.
sion with BSS (in mM: 120 NaCl, 18 NaHCO3, 4 KCl, 1 Reagents and analysis. Reagents were obtained from
MgSO4, 0.8 NaH2PO4, 1.4 CaCl2, and 5.6 glucose; 5% CO2, Sigma, and fluorophores were obtained from Molecular
pH 7.35). During simulated ischemia, cells were superfused Probes. Replicate experiments were carried out with sepa-
with a variation of BSS containing 20 mM 2-deoxyglucose rate coverslips. Treatment and control group were matched
(2-DOG) to inhibit glycolysis, zero glucose, 8 mM K, 5 mM by using cells isolated on the same day.
lactate, and low [O2] and hypercarbia (pH 6.8) obtained by
bubbling with 80% N2-20% CO2. During hypoxic acidosis,
cells were superfused with BSS bubbled with 80% N2-20% RESULTS
CO2. In other experiments, complete anoxia was achieved in To assess changes in m, contracting cardiomyo-
the ischemic or hypoxic medium by adding EC-Oxyrase (10
l/ml), an oxidase mixture that reduces O2 to H2O. Anoxia cytes on coverslips were loaded with TMRE, placed in

Downloaded from http://ajpheart.physiology.org/ by 10.220.33.2 on September 18, 2016


was confirmed with an optical phosphorescence quenching a flow-through chamber on an inverted microscope,
method using a porphryin probe in solution to measure PO2 and superfused with BSS (37C) equilibrated with nor-
within the perfusion chamber (Oxyspot) (24). After ischemia moxic (21% O2, 5% CO2) gas. TMRE loading properties
or hypoxia/anoxia (1 h), reperfusion was carried out with in cardiomyocytes, and the nonquenching characteris-
normoxic BSS (3 h). Tetramethylrhodamine methyl ester tics of this fluorophore in assessing membrane poten-
(TMRE) measurements were obtained during baseline, tial under nonquenching conditions were reported pre-
throughout ischemia, and during the first 90 min of reperfu- viously (6). Stable levels of fluorescence were typically
sion.
Mitochondrial membrane potential. m was assessed
observed under baseline conditions (Fig. 1A). This flu-
with TMRE. This cationic dye enters the cells and accumu- orescence was rapidly dissipated on addition of the
lates within mitochondria according to the Nernst equation protonophore FCCP, consistent with the expected loss
(12). Cells were loaded with TMRE (100 nM) at 37C for 45 of m induced by this uncoupling agent.
min; studies were carried out in the continued presence of the To assess the effects of ischemia on m, cells loaded
dye (10 nM). Mean fluorescence intensity was measured with TMRE were studied for 30 min under baseline
every minute (excitation 535 nm, emission 610 nm) for a field normoxic conditions followed by 60 min of simulated
of cells. Under these nonquenching conditions, mitochondrial ischemia. During ischemia, a progressive decrease in
depolarization with FCCP causes an immediate decrease in
TMRE fluorescence was observed, reaching 24.0
TMRE fluorescence, whereas oligomycin causes an immedi-
ate increase (6). Fluorescence intensity is expressed as the 5.5% of the initial intensity after 1 h. Minimal recovery
percentage of initial brightness after background subtrac- of TMRE fluorescence was seen after return to nor-
tion. moxia (reperfusion; Fig. 1B). Cell death within the
Cell viability. Viability was measured in the same field of same field of cells was minimal (5%) at the end of
cells used to assess m. Dead cells were identified with ischemia but increased significantly during 3-h reper-
Sytox Green, a membrane-impermeant dye that is excluded fusion (Fig. 1C). Inspection of TMRE fluorescence im-
from cells when the plasma membrane is intact. In dying ages revealed that the majority of cells lost all fluores-
cells with increased plasma membrane permeability, nuclear
cence during ischemia and some cells retained some
fluorescence becomes apparent. To assess cell death in a field
of cells, a fluorescent image (10 objective) was acquired and fluorescence at an attenuated level. Reperfusion was
the number of fluorescent nuclei was counted (Metamorph; not associated with significant recovery of fluorescence
Universal Imaging). Digitonin (300 M) was added to per- in either case.
meabilize all cells in the field. Cell counts were then re- Mitochondrial depolarization could conceivably be
peated, and the previous counts were normalized to that caused by activation of the mitochondrial permeability
value. transition (MPT) pore, a high-conductance putative
Lipid peroxidation assay. Cardiomyocytes were loaded channel in the inner mitochondrial membrane (10).
with C11-BODIPY (10 M), and fluorescence was measured The opening of the MPT pore has been suggested to
during baseline, ischemia, and reperfusion. Because of its
lipophilic nature, this fluorophore localizes to cell mem- contribute to the decrease in m during I/R injury
branes and can be used to assess oxidative stress in that (18). Cyclosporin A inhibits the opening of this pore
environment (27). On oxidation, the fluorescence (excitation and was therefore used to evaluate its contribution to
480 nm, emission 525 nm) of C11-BODIPY increases. membrane depolarization and cell death during I/R.
Generation of respiration-deficient 0-cells. To clarify the Cyclosporin A (0.2 M) had no significant effect on
significance of mitochondrial ROS generation for membrane membrane depolarization during simulated ischemia
damage and cell death during ischemia, mitochondria-defi- (Fig. 2A) and had no significant effect on cell death
cient cells (0-cells) were generated from wild-type 143B after 3-h reperfusion (Fig. 2B). Additional studies us-
osteosarcoma cells (American Type Culture Collection) (22).
The 0-cells were generated by incubating rapidly dividing
ing a higher concentration (0.5 M) also failed to abol-
wild-type cells with ethidium bromide (50 ng/ml), which ish the fall in TMRE fluorescence (data not shown).
inhibits replication of mitochondrial DNA (8). The mitochon- These results suggest that opening of the MPT pore
drial DNA encodes specific subunits that are critical for does not contribute significantly to the depolarization
electron transport, so 0-cells do not possess a functional and cell death in this model.
AJP-Heart Circ Physiol VOL 284 FEBRUARY 2003 www.ajpheart.org
MITOCHONDRIAL DEPOLARIZATION DURING ISCHEMIA H551

Downloaded from http://ajpheart.physiology.org/ by 10.220.33.2 on September 18, 2016


Fig. 2. A: effects of cyclosporin A (CyA) on mitochondrial potential in
cardiomyocytes as assessed with TMRE fluorescence during base-
line, ischemia, and reperfusion in a flow-through chamber. Cyclo-
sporin A was added to the buffer perfusate throughout the experi-
ment. B: cell death in the same experiments as assessed with the
membrane-impermeable probe Sytox Green.

subsequent cell death in this model, pharmacological


antioxidants were added to the perfusate throughout
the experiment and the effects on depolarization and
cell death were assessed (Table 1). The thiol reductants
2-mercaptopropionyl glycine (2-MPG; 400 M) and pyr-
rolidine dithiocarbamate (PDTC, 10 M) significantly

Table 1. Effect of antioxidants on mitochondrial


Fig. 1. A: representative experiment showing tetramethylrhodam- membrane potential at end of ischemia and
ine methyl ester (TMRE) fluorescence under nonquenching condi- cell death after reperfusion
tions to assess mitochondrial membrane potential (m). B: m in
cardiomyocytes as assessed with TMRE fluorescence during base- Fall in m (end Cell Death (end
line, ischemia, and reperfusion in a flow-through chamber. C: cell n of ischemia), % of reperfusion), %
death in the same population of cells assessed with the membrane-
impermeant probe Sytox Green during baseline, simulated ischemia, Control 9 76 5.5 66 7
and reperfusion. (mean values SE, n 9). 2-MPG (0.4 mM) 4 31 13 18 15
NAC (0.5 mM) 4 34 15 15 5
PDTC (10 M) 4 31 10 17 5
We previously found (33) evidence of mitochondrial 1,10-Phenanthroline (10 M) 4 20 10 61
oxidant stress during ischemia, before the start of Values are means SE for n fields of cells. m, mitochondrial
reperfusion. To determine whether ROS generation potential; 2-MPG, 2-mercaptopropionyl glycine; NAC, N-acetylcys-
during ischemia contributed to the fall in m and tome; PDTC, pyrrolidine dithiocarbamate.

AJP-Heart Circ Physiol VOL 284 FEBRUARY 2003 www.ajpheart.org


H552 MITOCHONDRIAL DEPOLARIZATION DURING ISCHEMIA

attenuated the fall in m at the end of ischemia, as did


N-acetyl-L-cysteine (NAC; 0.5 mM). Likewise, the metal
chelator 1,10-phenanthroline significantly attenuated
the decrease in m during ischemia. These antioxidant
compounds also significantly lessened cell death after
3-h reperfusion. Collectively, these studies suggested
that oxidant stress during ischemia contributes to the
fall in m.
ROS generated from the mitochondrial electron
transport chain can induce cardiolipin oxidation within
the inner mitochondrial membrane (29). This oxidative
damage could contribute to the observed membrane
depolarization by compromising the integrity of the
inner membrane. To detect lipid peroxidation, cardio-
myocytes loaded with the lipophilic probe C11-BODIPY

Downloaded from http://ajpheart.physiology.org/ by 10.220.33.2 on September 18, 2016


were subjected to simulated I/R. During ischemia, a sig-
nificant increase in fluorescence was observed, consistent
with an increase in the oxidation of this fluorophore. If
H2O2 is required for the oxidative damage during ische-
mia, then lipid peroxidation should be attenuated if SOD
is inhibited. Accordingly, the Cu,Zn-SOD inhibitor dieth-
yldithiocarbamate (DDC; 1 mM) was used to attenuate
SOD activity. This caused a significant attenuation in
C11-BODIPY fluorescence (Fig. 3A). In replicate experi-
ments, the rate of increase in fluorescence during ische-
mia (0.77 0.19 arbitrary units (a.u./min); n 6) was
significantly greater (P 0.01) than during baseline
(0.04 0.09 a.u./min; n 6). Administration of DDC
significantly decreased the slope of this relationship
(0.43 0.13, n 3; P 0.01), suggesting that H2O2
contributes to lipid peroxidation. If free iron in the cell
contributes to hydroxyl radical generation by the Fenton
reaction, then iron chelation should also attenuate lipid
peroxidation. Addition of the chelator 1,10-phenanthro-
line (10 M) during ischemia caused a significant de-
crease in the fluorescence signal (Fig. 3B). In replicate
experiments, 1,10-phenanthroline administration signif-
icantly decreased the rate of fluorescence increase
(0.63 0.13 a.u./min, n 3; P 0.01) compared with
ischemia. To determine whether mitochondria are the
source of these oxidants, the electron transport inhibitor
myxothiazol was added during ischemia to inhibit ROS
generation by complex III (30). Myxothiazol blocks elec-
tron transfer from ubiquinol to the Rieske iron-sulfur
center in complex III, thereby preventing the generation
of ubisemiquinone, which is a major source of superoxide
generation. Myxothiazol (2 M) significantly attenuated
the rate of increase in fluorescence during ischemia
(0.026 0.106 a.u./min, n 3). Addition of DMSO, the
solvent used in myxothiazol experiments, produced no
detectable effect.
ROS generation during ischemia most likely begins
Fig. 3. Representative tracing of cell lipid peroxidation as assessed
with univalent electron transfer to O2, thereby gener- with the lipophilic probe C11-BODIPY in cardiomyocytes during
ating superoxide. This process requires that some re- baseline and simulated ischemia. A: diethyldithiocarbamate (DDC),
sidual O2 must still be present to provide substrate for an inhibitor of Cu,Zn-SOD, was added during ischemia to attenuate
that reaction. In previous studies (33) we found that the production of hydrogen peroxide. B: 1,10-phenanthroline, an iron
low levels of O2 were present during ischemia in this chelator, was added during ischemia to attenuate the availability of
free iron. C: myxothiazol, a mitochondrial electron transport inhibi-
model. In the present study, O2 tension within the tor, was added during ischemia to block reactive oxygen species
flow-through chamber was assessed with a phospho- production from complex III. a.u., Arbitrary units.
rescence quenching technique previously shown to be
accurate at low [O2] (24, 37). During simulated ische-
AJP-Heart Circ Physiol VOL 284 FEBRUARY 2003 www.ajpheart.org
MITOCHONDRIAL DEPOLARIZATION DURING ISCHEMIA H553

effects on membrane potential and cell death (data not


shown). These findings suggested that residual O2 con-
tributes to cell death and the irreversible decline in
m during I/R.
During reperfusion after anoxic ischemia, TMRE flu-
orescence increased progressively, indicating a resto-

Downloaded from http://ajpheart.physiology.org/ by 10.220.33.2 on September 18, 2016


Fig. 4. Representative tracing showing PO2 in buffer perfusate
within the flow-through chamber during standard hypoxic ischemia.
Note that anoxic conditions were not achieved despite vigorous
bubbling with N2-CO2 gas mixtures in the equilibration column.

mia the O2 tension decreased progressively, reaching a


value of 7 mmHg within 10 min (Fig. 4). Therefore,
it is possible that superoxide could be generated using
residual O2 that is present during ischemia.
If residual O2 contributes to the generation of super-
oxide during ischemia, and if these oxidants contribute
to mitochondrial depolarization and cell death, then
significant protection should ensue if residual O2 is
scavenged from the system during ischemia. To test
this, an enzymatic O2 scavenger was added to the
perfusate to create anoxic conditions during ischemia,
thereby limiting the availability of O2 as an electron
acceptor. Anoxic conditions (PO2 0 mmHg) were
created by adding EC-Oxyrase to the ischemia buffer
after it had been equilibrated with 80% N2-20% CO2.
Measurements confirmed that this decreased the PO2
within the chamber from 7 to 0.1 mmHg during
ischemia. Heat inactivation of EC-Oxyrase (100C for
15 min) abolished its O2-scavenging properties (data
not shown). During anoxic ischemia (PO2 0 mmHg),
the decrease in m was attenuated (43 7% de-
crease; Fig. 5A) and cell death after reperfusion was
lessened (12 6%; P 0.001) compared with standard
hypoxic ischemia (66 7%; PO2 7 mmHg) (Fig. 5C).
Heat treatment of EC-Oxyrase abolished its protective

Fig. 5. A: m in cardiomyocytes as assessed with TMRE fluores-


cence during baseline, ischemia, and reperfusion in a flow-through
chamber. Standard hypoxic ischemia produced PO2 of 7 mmHg in the
flow-through chamber. Anoxic ischemia, generated by adding the
enzymatic O2 scavenger EC-Oxyrase to the ischemic perfusate, pro-
duced an PO2 of 0.1 mmHg in the flow-through chamber (mean
values SE, n 4 each). B: m assessed with TMRE fluorescence
during anoxic acidosis. Anoxic acidosis was generated by adding
EC-Oxyrase to standard buffered salt solution (BSS, containing
glucose) bubbled with 80% N2-20% CO2. In 1 group, oligomycin was
added to the perfusate only during anoxic acidosis. (mean values
SE, n 4 each) C: cell death after 3-h reperfusion with normoxic BSS
in the same populations of cells represented in A and B.

AJP-Heart Circ Physiol VOL 284 FEBRUARY 2003 www.ajpheart.org


H554 MITOCHONDRIAL DEPOLARIZATION DURING ISCHEMIA

ration of m (Fig. 5A). By contrast, minimal evidence


of recovery was seen during reperfusion after standard
hypoxic ischemia. This suggested that the mechanism
responsible for loss of m may be different between
hypoxic and anoxic ischemia. Normally, m reflects a
balance between the rate of proton extrusion from the
mitochondrial matrix (a function of the rate of electron
transport) and the rate at which protons reenter the
matrix (a function of ATP synthase activity and/or ion
leaks). During anoxia, electron flux should cease.
Therefore, m should decrease unless glycolytic ATP
is available to maintain m through reverse opera-
tion of the ATP synthase. During ischemia in our
model, inhibition of glycolysis by 2-DOG may have
prevented reverse operation of the ATP synthase. To

Downloaded from http://ajpheart.physiology.org/ by 10.220.33.2 on September 18, 2016


explore the mechanism responsible for the fall in m
during anoxia, cells were subjected to anoxia under the Fig. 6. Cell death during reperfusion as a function of the decrease in
same acidic conditions used for ischemia (20% CO2), m during ischemia in cardiomyocytes studied in a flow-through
except that glucose was added to the perfusate and chamber. The decrease in m was adjusted by lessening the sever-
2-DOG was omitted to permit glycolysis to continue. ity of hypoxia during the ischemia. Each data point represents an
independent experiment.
During anoxic acidosis, no decrease in m was ob-
served (Fig. 5B) and minimal cell was evident after
reperfusion (Fig. 5C), which suggested that m was
maintained during anoxia by reverse operation of the I/R, 0-cells were loaded with TMRE and studied dur-
ATP synthase. To confirm that reverse operation of the ing simulated ischemia. Despite a lack of electron
ATP synthase was responsible for sustaining m dur- transport, 0-cells maintain m by ATP/ADP ex-
ing anoxia when glycolysis remained functional, oligo- change via the adenine nucleotide translocator in the
mycin (10 M) was added to inhibit the ATP synthase inner membrane (5). Wild-type 143B osteosarcoma
during anoxic acidosis. Under those conditions, m cells demonstrated a marked depletion of m during
decreased significantly (Fig. 5B). During reperfusion ischemia that was qualitatively similar to that seen in
(21% O2, 5% CO2) without oligomycin, clear evidence of cardiomyocytes (Fig. 7). However, ischemia failed to
mitochondrial repolarization was evident because pro- produce a similar depletion of TMRE fluorescence in
ton pumping was restored when electron transport 0-cells. Cell death in wild-type cells averaged 82.2
resumed and the mitochondria membrane integrity 9.9% vs. 28.7 7.5% in the mitochondria-deficient cells
was not compromised. The decrease in m caused by (P 0.001). Thus the 0-cells were protected against
anoxic acidosis with oligomycin was associated with mitochondrial depolarization and subsequent cell
relatively low cell death (Fig. 5C). These findings indi- death compared with wild-type cells.
cate that low levels of residual O2 during ischemia are DISCUSSION
injurious because they contribute to an irreversible
decline in m. By contrast, the decline in m caused These studies demonstrate that mitochondria un-
by anoxia plus glucose deprivation is reversible and dergo a significant and irreversible decrease in poten-
associated with minimal cell death. tial during ischemia. The degree of depolarization cor-
Preliminary studies of standard hypoxic ischemia relates with the extent of cell death during reperfusion.
suggested that a correlation may exist between the However, mitochondrial depolarization by itself does
magnitude of the decrease in m and subsequent cell not cause cell death, because administration of anoxia
death. To determine whether such a dose-response plus oligomycin caused a reversible mitochondrial de-
relationship exists, we experimentally varied the se- polarization without causing significant cell death. Mi-
verity of ischemia by adjusting the residual level of O2 tochondrial depolarization during ischemia was trig-
during ischemia without changing its duration (1 h). In gered by ROS generated from the mitochondrial
these experiments the PO2 during ischemia was in- electron transport chain despite the low [O2] condi-
creased from 7 mmHg (hypoxic ischemia) to 15 tions. These oxidants appear to initiate a cascade of
mmHg. Subsequent cell death was measured after-3 h lipid peroxidation that disrupts the integrity of the
reperfusion. As shown in Fig. 6, the extent of cell death inner mitochondrial membrane, thereby preventing re-
during reperfusion was significantly attenuated when polarization during reperfusion. Activation of the MPT
the fall in m during ischemia was less severe. This pore apparently did not contribute to this process,
suggested that the magnitude of mitochondrial depo- because attempts to inhibit the activation of that pore
larization during ischemia might contribute mechanis- failed to prevent depolarization or cell death. By con-
tically to the cell death measured at the end of reper- trast, a variety of pharmacological antioxidant com-
fusion. pounds attenuated both the fall in m and subse-
To determine the significance of mitochondrial ROS quent cell death. Furthermore, scavenging of residual
generation for the depolarization and cell death during O2 during ischemia prevented the depletion of m
AJP-Heart Circ Physiol VOL 284 FEBRUARY 2003 www.ajpheart.org
MITOCHONDRIAL DEPOLARIZATION DURING ISCHEMIA H555

ischemic preconditioning (13, 26, 31). The activation of


preconditioning confers significant protection against
subsequent lethal ischemia. In contrast, higher levels
of oxidant stress are observed at the start of reperfu-
sion, when a transient burst of ROS generation is
observed that correlates with subsequent cell death
(32). During ischemia before reperfusion, ROS are gen-
erated by the electron transport chain of mitochondria
(33, 34), although the significance of these oxidants in
cell injury is not fully understood. The present study
focused on the relationship between oxidant stress
during ischemia, cell survival, and the fall in m in a
cardiomyocyte model.
Relationship between ischemic ROS and m. m
is normally maintained by proton pumping, which is

Downloaded from http://ajpheart.physiology.org/ by 10.220.33.2 on September 18, 2016


linked to the rate of electron transport. If the O2 ten-
sion in the cell falls below a critical level of 14 mmHg,
m should decrease because electron transport be-
comes limited by the availability of O2 at cytochrome
oxidase. In our study, large decreases in TMRE fluo-
rescence were observed during ischemia, indicating
that a significant fall in m must have occurred.
However, the fall in m could not be explained by a
lack of O2, because the O2 tension did not fall below the
critical level during the ischemic exposure (19). More-
over, m failed to show significant recovery during
reperfusion, which suggests that the mitochondria sus-
tained an irreversible injury during the ischemic expo-
sure. The extent of cell death after 3-h reperfusion
correlated significantly with the extent of the loss in
m, which suggests that the mitochondrial injury
sustained during ischemia could contribute to cell
death during reperfusion.
Several observations support the conclusion that
ROS contribute importantly to the decline in m

Fig. 7. A: m in wild-type 143B osteosarcoma cells as assessed with


TMRE fluorescence during baseline, ischemia, and reperfusion in a
flow-through chamber (n 5). B: mitochondrial potential in mutant
0-143B osteosarcoma cells as assessed with TMRE fluorescence
during baseline, ischemia, and reperfusion in a flow-through cham-
ber (n 5).

and significantly protected cells. Finally, 143B cells


lacking a mitochondrial electron transport chain failed
to demonstrate a depletion of m during ischemia
and were significantly protected against cell death
during reperfusion. Collectively these studies identify
a central role for mitochondrial oxidant generation
during ischemia in the irreversible mitochondrial de-
polarization and subsequent cell death induced by is-
chemia and reperfusion in this model (Fig. 8).
Role of ROS during I/R. ROS have long been associ-
ated with I/R injury. It is increasingly evident that
ROS play diverse roles in I/R, ranging from protective
effects at one extreme to damage-inducing effects at
the other. For example, low levels of oxidants appear to Fig. 8. Proposed scheme of cellular injury during ischemia before
function as signaling agents during the induction of reperfusion. ROS, reactive oxygen species.

AJP-Heart Circ Physiol VOL 284 FEBRUARY 2003 www.ajpheart.org


H556 MITOCHONDRIAL DEPOLARIZATION DURING ISCHEMIA

during ischemia. First, a variety of chemically dissim- The data suggest that H2O2 and hydroxyl radicals,
ilar antioxidant compounds given during ischemia rather than superoxide, are responsible for the oxida-
were able to attenuate the fall in m and to signifi- tive damage to mitochondria. Normally, superoxide
cantly lessen cell death after reperfusion. Second, the degradation by SOD is an important step in preventing
scavenging of residual O2 during anoxic ischemia ab- oxidant injury by that radical, so it is surprising that
rogated both the fall in m and later cell death. Both SOD inhibition is protective during ischemia. One ex-
the enzymatic activity of EC-Oxyrase and its protective planation is that H2O2 may pose an unusual threat to
effects were abolished by heat denaturation, which the cell under conditions of ischemia, when release of
indicates that the protection it provided was due to its iron from sites where it is normally chelated could
O2 scavenging properties. The studies with anoxic is- facilitate hydroxyl radical generation via the Fenton
chemia indicate that low residual levels of O2 during reaction. Relative to hydroxyl radical, superoxide is far
ischemia are important for mitochondrial depolariza- less reactive and may be less injurious during rela-
tion and cell death because they act as a substrate for tively short periods of ischemia.
the generation of ROS. Third, the results with C11- Relationship between mitochondrial depolarization
BODIPY indicate that lipid peroxidation occurs during and cell death. We observed a significant correlation

Downloaded from http://ajpheart.physiology.org/ by 10.220.33.2 on September 18, 2016


ischemia and that ROS originate from the mito- between the fall in TMRE fluorescence and cell death.
chondrial electron transport chain. Fourth, the 0-cells When the magnitude of the fall in m was manipu-
lacking an electron transport chain failed to exhibit lated by adjusting the severity of the ischemia, cell
mitochondrial depolarization and were significantly survival was found to be worse in experiments where
protected against cell death during reperfusion. We the fall in m was larger. Previous studies in cardio-
conclude that mitochondrial ROS generation during myocytes demonstrated that ROS generation tends to
ischemia contributes importantly to the fall in m. increase as O2 tension is lowered from 35 to 7 mmHg
This response is likely due to the formation of lipid (6). We therefore suggest that milder ischemia was
peroxides, which could undermine m by destabiliz- protective in the present study because of the lesser
ing the inner membrane. A loss of membrane integrity oxidant stress it generated and the associated decrease
could also explain why m failed to recover when in oxidant damage to membranes. An oxidant-medi-
ated disruption of mitochondrial inner membrane in-
normal O2 levels were restored during reperfusion.
tegrity during ischemia could conceivably lead to cell
This conclusion is consistent with the findings of
death by promoting matrix swelling, release of cyto-
Lesnefsky et al. (23), who demonstrated that cardio-
chrome c to the cytosol, and activation of the cell
lipin levels decrease during ischemia in subsarcolem-
apoptotic machinery (36). However, mitochondrial de-
mal mitochondria. Cardiolipin is a membrane phospho-
polarization by itself was not lethal to these cells, as
lipid found in high abundance within mitochondria
evidenced by the minimal extent of death observed
that interacts with electron transport proteins (14, 15) when m was depleted with anoxic acidosis plus
and may also be important for maintaining the integ- oligomycin. For a given degree of depolarization, it is
rity of the inner membrane in terms of its ability to conceivable that matrix swelling caused by lipid per-
support the transmembrane potential. Mitochondrial oxidation and loss of membrane integrity is more se-
oxidant generation during ischemia may explain the vere than when caused simply by electron transport
loss of cardiolipin, which could contribute to the mito- inhibition. In the former case, reoxygenation would not
chondrial dysfunction associated with I/R (28). Alter- promote recovery of m because the loss of membrane
natively, oxidants could contribute to mitochondrial integrity would defeat the effects of proton pumping. In
damage through direct oxidation of other lipids and the latter case, a restoration of electron transport dur-
proteins or by promoting the opening of the MPT pore ing reoxygenation would allow m to recover.
(3). Each of these mechanisms could contribute to mi- Interestingly, hypoxia alone (PO2 7 mmHg) was
tochondrial depolarization because of their effects on not sufficient to induce significant cell death in this
electron transport and/or mitochondrial membrane in- study. Likewise, hypercapnic acidosis (20% CO2) was
tegrity. However, our findings suggest that the de- well tolerated under normoxic conditions. However,
crease in m was not a result of MPT pore opening, when the two conditions were combined, 60% cell
because cyclosporin A treatment had no significant death resulted. Acidosis may increase cell death by
effect on the decrease in membrane potential during causing the release of Fe2 from intracellular sites
ischemia or the extent of cell death. This conclusion is where it is normally chelated. The ROS released in
consistent with previous studies suggesting that MPT response to hypoxia alone appear to be well tolerated
pore opening is unlikely to occur under the low pH by cells (7). However, in a setting where acidosis causes
conditions of ischemia and is more likely to occur after release of iron ions, these ROS may lead to generation
reperfusion (18, 20); indeed, we observed a fall in m of hydroxyl radicals as a consequence of Fenton inter-
during ischemia before reperfusion. However, other actions. The resulting loss of membrane integrity could
investigators have used higher (10, 16) or lower (38) explain the observed decrease in m and subsequent
concentrations of cyclosporin A to inhibit the opening failure to recover during reoxygenation. This interpre-
of that pore, so it is not clear whether protection would tation is consistent with our observation that an iron
have been observed at different concentrations or with chelator was most protective of m and cell viability.
other inhibitors of the MPT pore. Although the importance of the Fenton reaction in I/R
AJP-Heart Circ Physiol VOL 284 FEBRUARY 2003 www.ajpheart.org
MITOCHONDRIAL DEPOLARIZATION DURING ISCHEMIA H557

injury is not new (1, 4, 9, 17, 21, 25), its potential 14. Fry M, Blondin GA, and Green DE. The localization of tightly
involvement in cellular injury during ischemia before bound cardiolipin in cytochrome oxidase. J Biol Chem 255:
99679970, 1980.
reperfusion has not been explored previously, to our 15. Fry M and Green DE. Cardiolipin requirement for electron
knowledge. transfer in complex I and III of the mitochondrial respiratory
In summary, these studies reveal that ROS gener- chain. J Biol Chem 256: 18741880, 1981.
ated during ischemia contribute to the irreversible loss 16. Gibson EM, Henson ES, Villanueva J, and Gibson SB. MEK
of m. The extent of this damage correlates with the kinase 1 induces mitochondrial permeability transition leading
to apoptosis independent of cytochrome c release. J Biol Chem
extent of cell death during reperfusion, and interven- 277: 1057310580, 2002.
tions that minimize the oxidative stress during ische- 17. Granger DN and Korthuis RJ. Physiologic mechanisms of
mia also attenuate the loss of m. These findings postischemic tissue injury. Annu Rev Physiol 57: 311332, 1995.
therefore suggest that oxidant generation during ische- 18. Halestrap AP, Kerr PM, Javadov S, and Woodfield KY.
mia before reperfusion plays a significant role in deter- Elucidating the molecular mechanism of the permeability tran-
sition pore and its role in reperfusion injury of the heart. Bio-
mining cell death during reperfusion. chim Biophys Acta 1366: 7994, 1998.
19. Kennedy FG and Jones DP. Oxygen dependence of mitochon-
This study was supported by National Heart, Lung, and Blood
drial function in isolated rat cardiac myocytes. Am J Physiol Cell

Downloaded from http://ajpheart.physiology.org/ by 10.220.33.2 on September 18, 2016


Institute Grants HL-32646 and HL-35440. J. Levraut was supported
Physiol 250: C374C383, 1986.
by a grant from the Socie te de Re animation de Langue Franc aise.
20. Kerr PM, Suleiman MS, and Halestrap AP. Reversal of
permeability transition during recovery of hearts from ischemia
REFERENCES
and its enhancement by pyruvate. Am J Physiol Heart Circ
1. Ambrosio G, Zweier JL, Duilio C, Kuppusamy P, Santoro Physiol 276: H496H502, 1999.
G, Elia PP, Tritto I, Cirillo P, Condorelli M, and Chiariello 21. Kim MS and Akera T. O2 free radicals: cause of ischemia-
M. Evidence that mitochondrial respiration is a source of poten- reperfusion injury to cardiac Na K ATPase. Am J Physiol
tially toxic oxygen free radicals in intact rabbit hearts subjected Heart Circ Physiol 252: H252H257, 1987.
to ischemia and reflow. J Biol Chem 268: 1853218541, 1993. 22. King MP and Attardi G. Human cells lacking mtDNA: repopu-
2. Becker LB, Vanden Hoek TL, Shao ZH, Li CQ, and Schu- lation with exogenous mitochondria by complementation. Sci-
macker PT. Generation of superoxide in cardiomyocytes during ence 246: 500503, 1989.
ischemia before reperfusion. Am J Physiol Heart Circ Physiol 23. Lesnefsky EJ, Slabe TJ, Stoll MSK, Minkler PE, and Hop-
277: H2240H2246, 1999. pel CL. Myocardial ischemia selectively depletes cardiolipin in
3. Bernardi P. Mitochondrial transport of cations: channels, ex- rabbit heart subsarcolemmal mitochondria. Am J Physiol Heart
changers, and permeability transition. Physiol Rev 79: 1127 Circ Physiol 280: H2770H2778, 2001.
1155, 1999. 24. Lo LW, Koch CJ, and Wilson DF. Calibration of oxygen-
4. Bolli R, Jeroudi MO, Patel BS, DuBose CM, Lai EK, Rob- dependent quenching of the phosphorescence of Pd-meso-tetra
erts R, and McCay PB. Direct evidence that oxygen-derived (4-carboxyphenyl) porphine: a phosphor with general application
free radicals contribute to postischemic myocardial dysfunction for measuring oxygen concentration in biological systems. Anal
in the intact dog. Proc Natl Acad Sci USA 86: 46954699, 1989. Biochem 236: 153160, 1996.
5. Buchet K and Godinot C. Functional F1-ATPase essential in 25. Opie LH. Reperfusion injury and its pharmacologic modifica-
maintaining growth and membrane potential of human mito- tion. Circulation 80: 10491062, 1989.
chondrial DNA-depleted rho cells. J Biol Chem 273: 22983 26. Pain T, Yang XM, Critz SD, Yue Y, Nakano A, Liu GS,
22989, 1998. Heusch G, Cohen MV, and Downey JM. Opening of mito-
6. Budinger GRS, Duranteau J, Chandel NS, and Schu- chondrial KATP channels triggers the preconditioned state by
macker PT. Hibernation during hypoxia in cardiomyocytes: role generating free radicals. Circ Res 87: 460466, 2000.
of mitochondria as the O2 sensor. J Biol Chem 273: 33203326, 27. Pap EHW, Drummen GPC, Post JA, Rijken PJ, and Wirtz
1998.
KWA. Fluorescent fatty acid to monitor reactive oxygen in single
7. Chandel NS, McClintock DS, Feliciano CE, Wood TM, Me-
cells. Methods Enzymol 319: 603612, 2000.
lendez JA, Rodriguez AM, and Schumacker PT. Reactive
28. Paradies G, Petrosillo G, Pistolese M, Di Venosa N, Serena
oxygen species generated at mitochondrial Complex III stabilize
D, and Ruggiero FM. Lipid peroxidation and alterations to
HIF-1- during hypoxia: a mechanism of O2 sensing. J Biol
oxidative metabolism in mitochondria isolated from rat heart
Chem 275: 2513025138, 2000.
subjected to ischemia and reperfusion. Free Radic Biol Med 27:
8. Chandel NS and Schumacker PT. Cells depleted of mitochon-
drial DNA (0) yield insight into physiological mechanisms. 4250, 1999.
FEBS Lett 454: 173176, 1999. 29. Petrosillo G, Ruggiero FM, Pistolese M, and Paradies G.
9. Cordis GA, Maulik G, Bagchi D, Riedel W, and Das DK. Reactive oxygen species generated from the mitochondrial elec-
Detection of oxidative DNA damage to ischemic reperfused rat tron transport chain induce cytochrome c dissociation from beef-
hearts by 8-hydroxydeoxyguanosine formation. J Mol Cell Car- heart submitochondrial particles via cardiolipin peroxidation.
diol 30: 19391944, 1998. Possible role in the apoptosis. FEBS Lett 509: 435438, 2001.
10. Di Lisa F, Menabo R, Canton M, Barile M, and Bernardi P. 30. Turrens JF, Alexandre A, and Lehninger AL. Ubisemiqui-
Opening of the mitochondrial permeability transition pore none is the electron donor for superoxide formation by complex
causes depletion of mitochondrial and cytosolic NAD and is a III of heart mitochondria. Arch Biochem Biophys 237: 408414,
causative event in the death of myocytes in postischemic reper- 1985.
fusion of the heart. J Biol Chem 276: 25712575, 2001. 31. Vanden Hoek TL, Becker LB, Shao Z, Li C, and Schu-
11. Duranteau J, Chandel NS, Kulisz A, Shao Z, and Schu- macker PT. Reactive oxygen species released from mitochon-
macker PT. Intracellular signaling by reactive oxygen species dria during brief hypoxia induce preconditioning in cardiomyo-
during hypoxia in cardiomyocytes. J Biol Chem 273: 11619 cytes. J Biol Chem 273: 1809218098, 1998.
11624, 1998. 32. Vanden Hoek TL, Becker LB, Shao ZH, Li CQ, and Schu-
12. Ehrenberg B, Montana V, Wei MD, Wuskell JP, and Loew macker PT. Preconditioning in cardiomyocytes protects by at-
LM. Membrane potential can be determined in individual cells tenuating oxidant stress at reperfusion. Circ Res 86: 541548,
from the Nernstian distribution of cationic dyes. Biophys J 53: 2000.
785794, 1988. 33. Vanden Hoek TL, Li C, Shao Z, Schumacker PT, and
13. Forbes RA, Steenbergen C, and Murphy E. Diazoxide-in- Becker LB. Significant levels of oxidants are generated by
duced cardioprotection requires signaling through a redox-sen- isolated cardiomyocytes during ischemia prior to reperfusion. J
sitive mechanism. Circ Res 88: 802809, 2001. Mol Cell Cardiol 29: 25712583, 1997.

AJP-Heart Circ Physiol VOL 284 FEBRUARY 2003 www.ajpheart.org


H558 MITOCHONDRIAL DEPOLARIZATION DURING ISCHEMIA

34. Vanden Hoek TL, Shao Z, Li C, Schumacker PT, and 37. Wilson DF, Rumsey WL, Green TJ, and Vanderkooi JM.
Becker LB. Mitochondrial electron transport can become a The oxygen dependence of mitochondrial oxidative phosphoryla-
significant source of oxidative injury in cardiomyocytes. J Mol tion measured by a new optical method for measuring oxygen
Cell Cardiol 29: 24412450, 1997. concentration. J Biol Chem 263: 27122718, 1988.
35. Vanden Hoek TL, Shao Z, Li C, Zak R, Schumacker PT, 38. Woodfield K, Rueck A, Brdiczka D, and Halestrap AP.
and Becker LB. Reperfusion injury in cardiac myocytes after Direct demonstration of a specific interaction between cyclophi-
simulated ischemia. Am J Physiol Heart Circ Physiol 270: lin-D and the adenine nucleotide translocase confirms their role
H1334H1341, 1996. in the mitochondrial permeability transition. Biochem J 336:
36. Vander Heiden MG, Chandel NS, Li XX, Schumacker PT, 287290, 1998.
Colombini M, and Thompson CB. Outer mitochondrial mem- 39. Zweier JL, Flaherty JT, and Weisfeldt ML. Direct measure-
brane permeability can regulate coupled respiration and cell ment of free radical generation following reperfusion of ischemic
survival. Proc Natl Acad Sci USA 97: 46664671, 2000. myocardium. Proc Natl Acad Sci USA 84: 14041407, 1987.

Downloaded from http://ajpheart.physiology.org/ by 10.220.33.2 on September 18, 2016

AJP-Heart Circ Physiol VOL 284 FEBRUARY 2003 www.ajpheart.org

Anda mungkin juga menyukai