Cell Sorting Experiments Link Persistent Mitochondrial DNA Damage with Loss of Mitochondrial Membrane Potential and Apoptotic Cell Death*

Janine Hertzog SantosDagger , L'uba HunakovaDagger §, Yiming ChenDagger , Carl Bortner, and Bennett Van HoutenDagger ||

From the Dagger  Laboratory of Molecular Genetics and  Laboratory of Signal Transduction, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709 and § Cancer Research Institute, 833 91 Bratislava, Slovakia

Received for publication, August 27, 2002, and in revised form, October 15, 2002

    ABSTRACT
TOP
ABSTRACT
INTRODUCTION
EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
REFERENCES

In order to understand the molecular events following oxidative stress, which lead to persistence of lesions in the mtDNA, experiments were performed on normal human fibroblast (NHF) expressing human telomerase reverse transcriptase (hTERT). The formation and repair of H2O2-induced DNA lesions were examined using quantitative PCR. It was found that NHF hTERTs show extensive mtDNA damage (~4 lesions/10 kb) after exposure to 200 µM H2O2, which is partially repaired during a recovery period of 6 h. At the same time, the nDNA seemed to be completely resistant to damage. Cell sorting experiments revealed persistent mtDNA damage at 24 h only in the fraction of cells with low mitochondrial membrane potential (Delta Psi m). Further analysis also showed increased production of H2O2 by these cells, which subsequently undergo apoptosis. This work supports a hypothesis for a feed-forward cascade of reactive oxygen species generation and mtDNA damage and also suggested a possible mechanism for persistence of lesions in the mtDNA involving a drop in Delta Psi m, compromised protein import, secondary reactive oxygen species generation, and loss of repair capacity.

    INTRODUCTION
TOP
ABSTRACT
INTRODUCTION
EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
REFERENCES

Reactive oxygen species (ROS)1 are ubiquitous toxicants generated primarily by the mitochondrial respiration and by other biochemical processes within cells (1). In addition to these sources, ROS are also produced in pathophysiological conditions such as inflammation and ischemic reperfusion injury (2). Furthermore, certain environmental pollutants (like pesticides and herbicides), as well as smoking, can induce ROS either directly or indirectly (1, 3, 4). It is well known that ROS can damage biomolecules and have been postulated as an important cause of aging, cancer, and various human degenerative diseases (3, 5). One of the most widely used compounds to produce oxidative stress, both in vitro and in vivo biological studies, is hydrogen peroxide (H2O2). Unlike other ROS, H2O2 is not charged and is therefore freely diffusible within the cell. It can arise spontaneously or be generated from superoxide by superoxide dismutase. More importantly, in the presence of metal ions such as copper or iron, H2O2 can undergo Fenton chemistry and give rise to the potent hydroxyl radical. This radical is extremely reactive and can damage different components of the cell (6). The responses of cultured cells to H2O2 vary according to cell type and are known to be both time- and, especially, dose-dependent (for review see Ref. 7). For example, low concentrations (3-15 µM) of H2O2 were shown to activate a mitogenic response (8), whereas higher doses can have either cytostatic or cytotoxic effects. It was observed that concentrations ranging from 50 to 150 µM promote DNA damage (9, 10) replicative senescence (11-13), sustained p21 levels, cell cycle arrest, transient elevation of p53 protein (14), and temporary growth arrest followed by increased resistance to subsequent oxidative stress (8, 15). In contrast, H2O2 doses of 200 µM and higher were demonstrated to induce apoptosis (16), necrosis (16, 17), protein oxidation (18), as well as lesions in both nuclear and mitochondrial genomes (9, 10).

With regard to DNA damage, we and others have demonstrated that the mtDNA of different cell types and organisms is more prone to oxidative injury than the nDNA (9, 10, 19-22). Additionally, Yakes and Van Houten (9) demonstrated that depending on the intensity of the damage (dose and time of exposure), nuclear and mitochondrial genomes are differentially damaged and repaired. Although the precise molecular reason(s) that lead to the persistence of lesions in the mitochondrial genome is not clear, several explanations have been offered to clarify the increased vulnerability of the mtDNA to damage. These include the lack of a compact nucleosome structure as compared with the nDNA, the limited mitochondrial repair pathways, as well as the proximity of the mtDNA to the main source of ROS generation (9, 23). More work is needed to understand the molecular events underlying the susceptibility of the mtDNA to oxidative damage.

Whereas the initial results obtained regarding the kinetics of damage and repair in both mitochondria and nuclear genomes were based on SV40-transformed fibroblasts (9), data from the literature raise concerns as to the suitability of the SV40-transformed cell model. In fact, a significant number of physiological changes after viral transformation have been reported. For example, loss of p53 function with consequent alteration in cell cycle control (24) decreased DNA repair (25, 26), and diminished catalase activity (27) was found.

In an effort to better understand the basis of the increased susceptibility and persistent damage observed in mtDNA after oxidative stress, we have used a non-transformed diploid fibroblast cell line expressing the telomerase gene (NHF hTERT) to follow the formation and removal of H2O2-induced lesions. With this approach it was observed that these cells accumulate large amounts of mtDNA damage, which can be completely repaired after a 15-min treatment but not a 60-min treatment. Cell sorting experiments revealed that persistent lesions in the mtDNA correlate with loss of Delta psi m, increased ROS generation, and cell death. Interestingly, we also show that the nDNA of the NHF hTERTs seems totally resistant to H2O2-induced damage, suggesting that SV40-transformation makes the nuclear genome more prone to oxidative injury.

    EXPERIMENTAL PROCEDURES
TOP
ABSTRACT
INTRODUCTION
EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
REFERENCES

Cells and Cell Culture-- The normal human fibroblasts transfected with the telomerase gene (NHF hTERT) used throughout this study were a gift from Dr. Carl Barrett (NCI, National Institutes of Health). The cells were routinely grown and sub-cultured in 75-cm2 flasks at 37 °C and 5% CO2 in DMEM/F-12 medium (Invitrogen) supplemented with 10% fetal bovine serum (HyClone), SAP (0.2 mM serine, 0.1 mM aspartic acid, and 1.0 mM pyruvate), 50 units/ml penicillin/streptomycin (Invitrogen), and 800 µg/ml G418 (Invitrogen) for selection of the stable transfectants.

H2O2 Treatments-- Cells were seeded in 100-mm Petri dishes 15-18 h prior to the experiments. Immediately before treatment cells were washed once with DMEM/F-12 without any supplements, and the conditioned medium was saved for later use. A 30 mM stock of H2O2 (30% Sigma) was prepared in PBS (Invitrogen) and used to generate the 200 µM solution (in DMEM/F-12 alone) with which the cells were challenged. Monolayer cultures were exposed to H2O2 for 15 or 60 min and harvested immediately or allowed to recover for up to 24 h (in this case, in the original conditioned medium).

DNA Isolation, QPCR Assay, and DNA Damage Analysis-- High molecular weight DNA was extracted, and QPCR was performed and analyzed as described elsewhere (22). Briefly, genomic DNA was isolated, and specific primers were used to amplify a fragment of the mitochondrial and/or nDNA. The assay is based on the premise that DNA lesions (including oxidative damage such as strand breaks, base modifications, and abasic sites) block the progression of the polymerase so that only undamaged templates can participate in the PCR. Thus, amplification is inversely proportional to DNA damage: the more lesions encountered on the target DNA, the less the amplification. Note that for the mitochondrial genome both a short (221 bp) and a long fragment (8.9 kb) are routinely amplified. The rationale underlying this procedure is that the probability of introducing a lesion in a short segment is low, and therefore amplification of this segment gives an accurate estimation of the copy number of mtDNA in the sample. The data obtained from the small fragment were subsequently used to normalize the results of the 8.9-kb target (more details can be obtained in Ref. 22). Amplification of treated samples was then compared with controls, and the relative amplification was calculated. These values were next used to estimate the average number of lesions per 10 kb of the genome, using a Poisson distribution (22, 28). It is important to mention that a "negative" number of lesions was obtained when amplification of the treated sample was higher than that of the matched control. This phenomenon can occur by means of induced repair activity in the treated sample (9, 29). Results presented here are the mean of two sets of PCR for each target gene of at least three different biological experiments. Student's unpaired t test was performed to evaluate statistical significance.

Mitochondrial Membrane Potential Analysis-- Changes in the Delta psi m were measured by flow cytometry using JC-1 (5,5',6,6'-tetrachloro-1,1',3,3'-tetra-ethylbenzimidazolylcarbocyanine iodide; Molecular Probes). JC-1 was chosen because of its specificity regarding the mitochondrial membrane (30-32). At various time points (every 2 h up to 24 h and then at 36 h and 48 h) after the challenge with H2O2, control and treated cells were harvested, washed twice with PBS, and incubated with 10 µM JC-1 for 10 min (37 °C; 5% CO2 atmosphere). For each sample, 10,000 cells were examined on an FL-1 (530 nm) versus FL-2 (585 nm) dot plot using a BD Biosciences FACSort. JC-1 has dual emission depending on the state of the Delta psi m, forming aggregates with a high 585 nm fluorescence in cells (which indicates a normal Delta psi m). Loss of mitochondrial membrane integrity results in reduction in 585 nm fluorescence with a concurrent gain in 530 nm fluorescence as the dye shifts from an aggregate to a monomeric state (33). Therefore, retention of the dye in the cell can be monitored. The data were converted to density plots using the CellQuest software.

Cell Cycle Analysis with Propidium Iodide-- Cells were treated as described above and at various times after the treatments were subjected to cell cycle analysis by flow cytometry using propidium iodide (PI, Molecular Probes). Briefly, at each time point both control and treated cells were harvested, washed twice with PBS, and incubated at room temperature for 15 min with 1 mg/ml RNase, 1 µl of Triton X-100(Sigma), and 20 µg/ml of PI. Samples were then immediately analyzed by fluorescence-activated cell sorter.

Caspase Activation Assay-- Measurement of caspase 3 activation was performed fluorimetrically using the Bio-Rad Apopain Kit, according to instructions of the manufacturer. Briefly, 24 h after the challenge with H2O2 cells were harvested and subjected to total protein extraction using CHAPS buffer with 5 mM dithiothreitol (Cell Signaling) and 1 mM phenylmethylsulfonyl fluoride. After 5 rounds of quick-freezing (dry ice embedded in ethanol) and thawing (37 °C bath), protein concentrations were determined, and subsequently, 50 µl of each sample was used in the Apopain assay. Pro-caspase/apopain activity was monitored using a fluorogenic peptide substrate, Ac-DEVD-AFC. Apopain enzymatically cleaved the AFC from the peptide and released it in a free form, which upon exposure to near UV light produced a blue-green fluorescence (detected at 500-550 nm). A calibration curve was generated first by measuring fluorescence of AFC released from a serial dilution of the standard substrate (AC-DEVD-AFC). A positive control consisting of protein extract of Jurkat cells treated with a Fas ligand, a model system for apoptosis, was also included. Duplicate fluorescence readings of the samples were taken every 30 min after reaction was started for up to 3 h until the readings stabilized. Caspase activity was calculated comparing results from the samples to the calibration curve. Data were reported as fluorescence readings.

Sorting Procedure Based on JC-1 Staining and Flow Cytometry-- Sorting of the normal and depolarized (low Delta psi m) JC-1-stained cells was accomplished using a BD Biosciences FACSVantage SE. In these experiments, both control and 60-min treated cells were initially gated in forward-scatter versus a side-scatter dot plot to exclude debris. Sort gates were set up on an FL-1 (530:30 nm) versus FL-2 (575:26 nm) dot plot with increased stringency for only the depolarized cells. An all-sort cell population (cells gated on the initial forward-scatter versus side-scatter dot plot) was also collected to control for the introduction of DNA lesions and/or any change in the Delta psi m because of the sorting procedure. The data were converted to density plots using CellQuest software. Individual sorted cell populations were then immediately subjected to DNA extraction and QPCR analysis or placed back into culture for viability assays.

H2O2 Detection from Sorted Cells-- Cells were sorted as described above, and supernatants were immediately collected by centrifugation and subjected to analysis using the Amplex Red Kit (Molecular Probes). The assay is based on the detection of H2O2 using 10-acetyl-7-hydroxiphenoxazine (Amplex Red reagent), a highly stable and specific probe for this compound. In the presence of horseradish peroxidase, Amplex Red reacts with H2O2 at a 1:1 stoichiometry, producing a highly fluorescent product, resorufin, which can be detected using excitation and emission wavelengths of 530 and 590 nm, respectively (34). Briefly, 100 µl of the supernatants was loaded in a 96-well microtiter plate and incubated for 30 min (protected from light and at room temperature) in the presence of phosphate buffer containing 400 µM of Amplex Red reagent and 2 units/ml horseradish peroxidase. Background fluorescence was corrected by subtracting the values derived from PBS alone. Controls including catalase were also included to ensure that the fluorescent signal was due to H2O2. For each sample, all measurements were made in duplicate. The mean of those readings was then used to estimate H2O2 concentration using a standard H2O2 curve.

    RESULTS
TOP
ABSTRACT
INTRODUCTION
EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
REFERENCES

The purpose of this study was to investigate the formation and repair of H2O2-induced lesions in both nuclear and mitochondrial genomes of normal human fibroblasts transfected with the telomerase gene. In addition, we also wanted to understand the apparent loss of DNA repair activity in cells showing persistent mtDNA damage and to gain some insights into the biological outcome(s) of this permanent damage.

H2O2-induced Damage-- Quantitation of lesions introduced both in the mitochondrial and nuclear genomes was performed using QPCR, and the details of the assay have been published previously (9, 22, 28). Fig. 1A shows the relative amplification for the mtDNA of NHF hTERT cells after treatment with 200 µM H2O2 and repair periods of up to 24 h. As can be seen, a 15-min exposure to H2O2 gives rise to an ~70% decrease in amplification as compared with the untreated control. Note that amplification was completely restored after a 1.5-h incubation in conditioned medium. In contrast, cells challenged for 60 min showed a marked reduction in amplification, corresponding to ~4 lesions/10 kb. Unlike the 15-min treatment, the 60-min treatment leads to lesions that persisted even after long recovery periods (24 h). During the first 6 h, ~50% of the damage from a 60-min treatment could be repaired. However, no further removal could be observed up to 24 h. These data suggest that the 60-min treatment passed some threshold of damage, such that some other deleterious events have taken place inside the mitochondria.


View larger version (32K):
[in this window]
[in a new window]
 
Fig. 1.   The mitochondrial genome of NHF hTERT cells is extremely sensitive to oxidative stress, whereas the nuclear DNA is resistant to damage. Kinetics of damage and repair of the mitochondrial fragment (8.9 kb) of cells treated with 200 µM H2O2 for 15 min or 60 min and allowed to recover for the times indicated (A). B depicts relative amplification of the nuclear beta -globin gene immediately after H2O2 treatments and at various times for repair (similar results were obtained for the HPRT gene, not shown). The decrease in amplification was calculated comparing treated samples to undamaged control (dashed line). These values were then used to determine the lesion frequency using a Poisson distribution as described under "Experimental Procedures." Data are expressed as the mean ± S.E. of three biological experiments in which two PCRs were performed per experiment. Student's t test was performed comparing treated samples (time 0) to untreated control or time 0 to each recovery period (*, significance stands for p < 0.05; **, p < 0.01).

Surprisingly, the nDNA of NHF hTERT cells did not show any decrease in amplification (when compared with untreated controls) even after a 60-min exposure to H2O2 (Fig. 1B). This result suggested an absence of lesions detected in the two genomic regions (beta -globin and HPRT) studied. These data differ significantly from the findings of Yakes and Van Houten (9), which demonstrated the same type of treatment to introduce 0.5 lesions/10 kb into the nuclear genome of SV40-transformed fibroblasts. Interestingly, we were able to perceive a very low amount of nDNA damage 24 h after the treatments, which was coupled to the observation that a vast proportion of cells (~50%, data not shown) were detaching from the dishes. Together, these results indicate that the biology of a specific population of cells has changed after H2O2 treatment, culminating in cell death. Moreover, the absence of nDNA damage suggests that, in this scenario, the cell death is exclusively a consequence of persistent lesions in the mtDNA.

Mitochondrial Membrane Potential as an Indicator of Apoptosis-- We next sought to determine whether the fraction of cells that were detaching from the dishes was dying by apoptotic death. We reasoned that cells with extensive and persistent mtDNA damage might not be able to maintain the Delta psi m across the inner membrane, believed to play a role in the apoptotic process (reviewed in Ref. 35). The integrity of the Delta psi m was evaluated using JC-1, a fluorophore recognized as the most specific probe for the detection of changes in the Delta psi m of different cell types as compared with other available dyes (30-32). Cells were treated as described under "Experimental Procedures" for 15 or 60 min and were analyzed every 2 h after the exposure to H2O2. No differences in the Delta psi m were observed when comparing the 15-min with the control at any time monitored (Fig. 2A, upper right panel). However, a decline in the Delta psi m was detected at around 14 h in cells treated with H2O2 for 60 min, with the highest proportion of cells (~70%) with decreased Delta psi m values observed at 24 h (Fig. 2A). These data suggest that the cells treated for long periods of time with H2O2 might be undergoing apoptosis.


View larger version (44K):
[in this window]
[in a new window]
 
Fig. 2.   Markers of apoptosis observed in cells treated with 200 µM H2O2 for 15 or 60 min. A, cells were incubated with JC-1 for 10 min (37 °C) and analyzed by flow cytometry at various times after treatment (14 and 24 h, for 60 min of treatment; 24 h for 15 min of treatment) to follow the Delta Psi m. Drop in Delta Psi m is identified as a change in JC-1 properties from aggregate to monomeric form, only detected in cells treated for 60 min. B represents the fluorimetric analysis of caspase 3 activation (details of the assay are described under "Experimental Procedures"); , control; open circle , cells treated for 15 min; triangle , cells treated for 60 min; and black-triangle, positive control (Jurkat cells treated with a Fas ligand). Data representative of three different biological experiments. C, detection of apoptotic population by means of PI staining, analyzed by flow cytometry, after 24, 48, and 72 h of the treatments. R1 gates the apoptotic population, which is translated in percentage of total events analyzed.

Caspase Activation and Apoptotic Population Detection by PI-- To confirm that a fraction of the cells in our experiments were undergoing apoptotic death, other apoptosis-specific assays were conducted including analysis of caspase-3 activation as well as the staining of the nDNA with PI.

Caspase 3 activation was followed using a fluorimetric assay as described under "Experimental Procedures." Fig. 2B shows that only cells that were exposed for 60 min to H2O2 had increased caspase-3 activation as compared with the negative control. DNA staining with PI also indicated a significant percentage of apoptotic cells beginning at 24 h with a peak at 72 h (Fig. 2C). Taken together, these data indicate that a fraction of NHF hTERTs, when exposed to H2O2 for 60 min, undergoes apoptosis; this process was exclusively a consequence of extensive damage to the mitochondrial genome. Like many other studies with fibroblasts, these same cells did not show DNA laddering at any time monitored (24, 36, 48, and 72 h; data not shown).

Sorting Procedure by Flow Cytometry and JC-1 Staining-- Integrity of the Delta psi m plays a role not only in apoptosis but also in trafficking various proteins into the mitochondria. In fact, nuclear encoded proteins that act in this organelle have to be targeted, unfolded, and refolded inside the mitochondrion by different protein complexes attached to the outer and inner mitochondrial membranes. This import mechanism also depended on the maintenance of a high Delta psi m (reviewed in Ref. 36). Considering that repair proteins that act in mitochondria are nuclear encoded, we next sought to evaluate whether the persistent mtDNA damage observed only in the cells treated for 60 min correlated with the drop in Delta psi m. To determine whether this was the case, a procedure relying on JC-1 staining and flow cytometry was adopted.

By using a FACSVantage SE flow cytometer and JC-1, both control and H2O2-treated cells were sorted into separate populations based on having low (depolarized cells) or high Delta psi m values (polarized cells). Fig. 3 shows the gating scheme used to individually isolate polarized and depolarized populations. Immediately after sorting, DNA was isolated, and damage was evaluated with the QPCR assay. Fig. 4A illustrates the results from the mtDNA damage analysis on the various sorted populations of cells. Interestingly, only the depolarized population from the treated cells showed significant mtDNA damage. This is the first evidence that directly links depolarization of the Delta psi m with persistent lesions in this genome. As described under "Experimental Procedures," an all-sort cell population where cells passed through the sorter but were not physically separated was included. Note that no significant differences in the amount of mtDNA damage were observed between the depolarized and all-sort treated cells, which is probably due to stringency of the gated depolarized population that excluded some depolarized cells (those falling between the two sort gates) that were included in the all-sort. In addition, it is important to notice that the majority of the treated (>= 60%) cells were falling in the depolarized gate (example depicted in Fig. 3).


View larger version (31K):
[in this window]
[in a new window]
 
Fig. 3.   Flow cytometric parameters used for separation of distinct population of cells showing high (polarized) and low (depolarized) Delta Psi m based on JC-1 staining. Upper panel represents control cells, and the bottom panel represents the 60-min treated populations. Left-hand side of the panels depicts polarized cells, and the right-hand side represents the depolarized counterparts.


View larger version (13K):
[in this window]
[in a new window]
 
Fig. 4.   Persistent mitochondrial DNA damage is restricted to treated depolarized cells, which also show significantly increased levels of ROS. A, QPCR analysis of the mitochondrial target of NHF hTERT cells after the sorting procedure. Cells were treated as described previously, allowed to repair for 24 h, stained with JC-1 for 10 min, and subjected to flow cytometric sorting using the FACSVantage SE. Controls included 1) cells harvested immediately after the treatments (data not shown); 2) aliquot harvested 24 h after the challenge and kept with JC-1 for the same period as sorted cells but not submitted to this procedure (data not shown); and 3) cells that passed through the sorter but no separation of the two distinct populations conducted (all-sort). Data represent the amount of lesions per 10 kb in the populations that were sorted (n = 2). B, H2O2 detection in sorted cells. After sorting, cells were harvested, and the supernatant was collected for H2O2 measurements. To ensure comparability of results, the ratio between cell number and volume collected was constant so that the dilution was the same in all samples (i.e. the volume of flow cytometer fluid per cell was identical). Aliquots of the supernatant were then subjected to the Amplex Red coupled to horseradish peroxidase assay and H2O2 concentration calculated based on a standard H2O2 curve. Numbers represent the mean ± S.E. (n = 2). Unpaired Student's t test applied for statistical significance comparing depolarized samples to polarized counterparts (p values < 0.05).

H2O2 Released from Sorted Cells and Viability Assessment-- Our current working hypothesis predicts that a damaged mitochondrion generates more ROS than an intact organelle. The mtDNA encodes 13 polypeptides involved in the function of the electron transport chain (ETC). Incomplete flow of electrons could be caused by the lack of expression of vital mitochondrial genes from a damaged genome and would be expected to give rise to increased levels of ROS. In this way, a high steady-state level of ROS could be generated inside the organelle, which may in turn continuously injure the mtDNA in a feed-forward cascade. Eventually, this vicious cycle results in loss of mitochondria function and apoptotic cell death. Thus we next sought to determine whether ROS were being released from the sorted cells, especially from the population showing both low Delta psi m and permanent mtDNA damage. As described under "Experimental Procedures," supernatants of sorted cells were collected and used to perform the Amplex Red assay, which is specific for H2O2. As can be observed, treated depolarized cells release significantly more H2O2 than polarized counterparts 24 h after the treatments (Fig. 4B). It could be argued that this result reflects the presence of the initially supplied H2O2; however, the exogenous compound showed a half-life of 30 min in the presence of NHF hTERT cells. Moreover, H2O2 detection reached background levels by 4 h after the treatments (data not shown).

Finally, it was important to confirm that the cells with persistent mtDNA damage were the inviable cells after the treatments. In an effort to investigate this phenomenon, sterile sorting was conducted, and both H2O2-treated and non-treated polarized and depolarized populations were re-cultured to access viability. Cells were monitored for the next 72 h and counted with a Coulter counter. It was found that 80% of the control polarized cells were able to attach and grow, whereas the portion of treated cells with normal Delta psi m showed ~66% attachment. Only 5% of the depolarized treated cells was able to attach as compared with 13% of the depolarized controls (data not shown), suggesting that these populations are mainly composed of non-viable cells.

    DISCUSSION
TOP
ABSTRACT
INTRODUCTION
EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
REFERENCES

The increased susceptibility of the mtDNA to H2O2-induced damage, as compared with the nDNA, has been documented previously (9, 10, 23); however, the specific molecular causes of these phenomena are not well understood. It has been hypothesized that a combination of biological determinants, such as the proximity of the mtDNA to ROS, the lack of a compact nucleosome structure to protect the genome, and a paucity of mtDNA damage-processing pathways relative to those present in the nucleus play a role (reviewed in Ref. 23). In an attempt to understand these phenomena better, we have used telomerase-expressing normal diploid fibroblasts and found that repair of ROS-induced lesions in the mitochondrial genome depends on a threshold of dose and time and that persistent mtDNA damage leads to loss of Delta psi m and subsequent cell death.

The present study confirmed earlier findings (9, 10), H2O2 induces a large amount of mtDNA lesions (Fig. 1A) and little or no nDNA damage (Fig. 1B). Surprisingly, the mitochondrial genome of hTERT-transfected cells appeared to be more vulnerable to H2O2 than previously reported (9, 10, 19-22) for several other cell types, whereas the nuclear genome of the same cells was completely resistant to damage. This is likely a reflection of the fact that hTERT-transfected fibroblasts have increased levels of intracellular iron, making the mtDNA more prone to damage.2 The fact that the lesions induced in the mitochondrial genome after a 60-min treatment are repaired up to 6 h (~50% of lesions are removed) agrees with previous reports (23, 37-39) suggesting that repair in this organelle is capable of removing oxidative DNA lesions. However, these data also indicate that removal of damage is clearly dependent on the total amount of lesions induced over a period of time, after which additional events take place in the mitochondrion that seem to compromise repair function.

DNA damage caused by ROS is removed by the base excision repair (BER) machinery both in the nucleus and in the mitochondria. Although oxidative stress induces a wide variety of lesions, many are removed from the mtDNA through the action of a glycosylase with an associated abasic site-lyase activity, followed by the activity of gamma -polymerase and DNA ligase (reviewed in Refs. 23 and 37-39). Because the mitochondrial genome encodes polypeptides exclusively involved in the production of ATP through oxidative phosphorylation, BER proteins (as well as the other 1000 estimated proteins that function in this organelle) are nuclear encoded and therefore must be targeted to the mitochondrion.

Trafficking of nuclear encoded proteins from the cytosol to the mitochondria is achieved by the concerted action of at least four translocation complexes (see Ref. 36 for review). In this scenario, the Delta psi m provides one of the major driving forces for the vectorial transport of polypeptide chains by these import machines (40). The results obtained during this study provide evidence that a majority (~70%) of cells exposed to H2O2 for 60 min have lost their Delta psi m within 24 h of the treatments (Fig. 2A). This led us to hypothesize that the presence of mtDNA lesions and the loss of Delta psi m are intimately linked and may result in lack of repair activity through a compromised import mechanism. Data to support this concept were obtained from our sorting experiments (Fig. 4A). In fact, this approach clearly showed that only those cells that have low Delta psi m also showed mtDNA damage, suggesting that a compromised import mechanism was involved in the lack of repair activity in mitochondria.

However, one cannot rule out other putative causes that may impair the overall repair activity in the mitochondria. For example, Graziewicz et al. (18) showed that the human gamma -polymerase, the only polymerase present and active in mitochondria, is prone to oxidative damage. This might also be the case in our experiments. Along the same line, another BER protein that could be the target of oxidative attack is the human endonuclease III homologue hNTH1 (41). Although there is no direct evidence that hNTH1 is sensitive to oxidative damage, one could envision a mechanism analogous to the inactivation of aconitase. This protein localizes in the mitochondrial matrix and, like hNTH1, has an iron-sulfur [4Fe-4S] cluster in its active site. Yan et al. (42) showed that aconitase is modified and inactivated by oxidative stress, which is believed to occur because of the release of one iron atom from the cluster (43). It is worth mentioning that if oxidation of hNTH1 is taking place in our experiments, one would expect not only the enzyme to decrease repair efficiency but also to release iron, which could further contribute to Fenton chemistry and higher the levels of mtDNA damage. In this context, preliminary data following mitochondrial protein oxidation revealed that several proteins are oxidized in this organelle in NHF hTERT cells exposed to H2O2 (data not shown).

Alternatively, saturation of repair activity by increased promotion of lesions, even in the presence of functional repair machinery, could be responsible for the persistence of lesions in the mitochondrial genome. Inappropriate electron flow on an already damaged ETC represents a potential source of additional waves of ROS (9). In this case, a vicious cycle is established where the basal level of ROS generation as well as of mtDNA lesions is enhanced, possibly saturating the active repair system. The fact that the depolarized sorted cells (those that also show the mtDNA damage) release increased amounts of H2O2 as compared with the polarized counterparts (Fig. 4B) is consistent with this model.

It is worth pointing out that regardless of the mechanism(s) contributing to the maintenance of lesions in the mitochondrial genome, our experiments indicate that the biological effect of these persistent lesions is apoptosis (Fig. 2, A-C). Accordingly, Dobson et al. (44) demonstrated that targeting an 8-oxoguanine-specific glycosylase not only enhances the repair of menadione-induced mtDNA lesions but also the viability of treated cells. In addition, it has been proposed that the differential susceptibility of glial cell types to oxidative stress and apoptosis correlates with increased oxidative mtDNA damage (45). Although reports are available showing that H2O2 induces apoptotic death, the mechanisms through which this oxygen byproduct promotes apoptosis are still debated (see Ref. 7 for review). Interestingly, down-regulation of mitochondrially encoded mRNA, rRNA, and DNA was reported in apoptotic cells (46-48). All these events are consistent with lesions being present in the mitochondrial genome, as previously proposed by Yakes and Van Houten (9).

A striking finding in this study is the total absence of nDNA damage in the same cells that show a large number of mtDNA lesions. In fact, detectable levels of nDNA damage were only observed 24 h after the treatments, which is when apoptosis has been initiated. Because the mtDNA showed increased susceptibility to oxidative damage in hTERT-transfected cells, one might also expect the nDNA to be more prone to insult. It might be argued that the nuclear genome, unlike the mitochondrial one, is better protected by histones and, thus, less accessible to oxidative attack. In addition, the presence of additional and overlapping repair pathways (such as the nucleotide excision repair) (49) could account for the lack of damage accumulation. An appealing alternative relates to the fact that the cells used in this study are not virus-transformed and, therefore, have intact p53 function. Although p53 has been claimed to interact directly and promote the activity of the BER machinery (50), we have no evidence that this is the case in our experiments. One could envision that an active p53 is necessary and sufficient to decrease nDNA damage resulting from oxidative stress. Further p53 analysis should help to shed light onto this problem.

In summary, the findings of the sorting experiments lead us to suggest a chain of events, involving persistent mtDNA damage, drop in Delta psi m, loss of repair activity and apoptosis. Because mtDNA lesions were observed within 5 min of H2O2 exposure (data not shown), it is most likely that mtDNA damage is the initial event triggering this cascade. Although our results do not implicate a drop in the Delta psi m as the sole event to impair mitochondrial repair activity, they provide a novel insight to account for the increased susceptibility of the mitochondrial genome to oxidative attack as compared with the nDNA. An increased understanding of the kinetics of damage and repair (or lack of repair) in the mitochondrial genome should provide new insights into the biological/physiological significance of mtDNA damage and, eventually, mutagenesis.

    ACKNOWLEDGEMENTS

We thank Drs. Milan Skorvaga and Bhaskar Mandavilli for fruitful discussions in the laboratory; Dr. Julie Horton for help with the cell counting; Dr. Maria Anna Graziewicz for assistance in the protein oxidation assay, and also Drs. William Copeland, Julie Horton, and Michael Resnick for critical review of the manuscript.

    FOOTNOTES

* The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

|| To whom correspondence should be addressed: Laboratory of Molecular Genetics, NIEHS, 111 Alexander Dr., P. O. Box 12233, Research Triangle Park, NC 27709. Tel.: 919-541-7752; Fax: 919-541-7593, E-mail: vanhout1@niehs.nih.gov.

Published, JBC Papers in Press, November 6, 2002, DOI 10.1074/jbc.M208752200

2 J. H. Santos and B. Van Houten, manuscript in preparation.

    ABBREVIATIONS

The abbreviations used are: ROS, reactive oxygen species; H2O2, hydrogen peroxide; QPCR, quantitative polymerase chain reaction; NHF, normal human fibroblast; hTERT, human telomerase reverse transcriptase; Delta psi m, mitochondrial membrane potential; ETC, electron transport chain; CHAPS, 3-[(3-cholamidopropyl)dimethylammonio]-1-propanesulfonic acid; PBS, phosphate-buffered saline; DMEM, Dulbecco's modified Eagle's medium; BER, base excision repair; AFC, 7-amino-4-trifluoromethylcoumarin; PI, propidium iodide; mtDNA, mito- chondrial DNA; nDNA, nuclear DNA.

    REFERENCES
TOP
ABSTRACT
INTRODUCTION
EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
REFERENCES

1. Cadenas, E. (1989) Annu. Rev. Biochem. 58, 79-110[CrossRef][Medline] [Order article via Infotrieve]
2. Kehrer, J. P. (1989) Free Radic. Res. Commun. 6, 305-314
3. Ames, B. N., Shigenaga, M. K., and Hagen, T. M. (1993) Proc. Natl. Acad. Sci. U. S. A. 90, 7915-7922[Abstract/Free Full Text]
4. Halliwell, B., and Cross, C. E. (1994) Environ. Health Perspect. 10 (suppl.), 5-12
5. Berlett, B. S., and Stadtman, E. R. (1997) J. Biol. Chem. 272, 20313-20336[Free Full Text]
6. Fridovich, E. (1998) J. Exp. Biol. 201, 1203-1209[Abstract/Free Full Text]
7. Davies, K. J. (1999) IUBMB Life 48, 41-47[CrossRef][Medline] [Order article via Infotrieve]
8. Wiese, A. G., Pacifici, R. E., and Davies, K. J. (1995) Arch. Biochem. Biophys. 318, 231-240[CrossRef][Medline] [Order article via Infotrieve]
9. Yakes, F. M., and Van Houten, B. (1997) Proc. Natl. Acad. Sci. U. S. A. 94, 514-519[Abstract/Free Full Text]
10. Salazar, J. J., and Van Houten, B. (1997) Mutat. Res. 385, 139-149[Medline] [Order article via Infotrieve]
11. Chen, Q., and Ames, B. N. (1994) Proc. Natl. Acad. Sci. U. S. A. 91, 4130-4134[Abstract]
12. Chen, Q. M. (2000) Ann. N. Y. Acad. Sci. 908, 111-125[Abstract/Free Full Text]
13. Frippiat, C., Chen, Q. M., Zdanov, S., Magalhaes, J.-P., Remacle, J., and Toussaint, O. (2001) J. Biol. Chem. 276, 2531-2537[Abstract/Free Full Text]
14. Chen, Q. M., Bartholomew, J. C., Campisi, C., Acosta, M., Reagan, J. D., and Ames, B. N. (1998) Biochem. J. 332, 43-50[Medline] [Order article via Infotrieve]
15. Crawford, D. R., Schools, G. P., and Davies, K. J. (1996) Arch. Biochem. Biophys. 329, 137-144[CrossRef][Medline] [Order article via Infotrieve]
16. Chen, Q., Liu, B., and Merrett, J. B. (2000) Biochem. J. 347, 543-551[CrossRef][Medline] [Order article via Infotrieve]
17. Bladier, C., Wolvetang, E. J., Hutchinson, P., de Haan, J. B., and Kola, I. (1997) Cell Growth Differ. 8, 589-598[Abstract]
18. Graziewicz, M. A., Day, B. J., and Copeland, W. C. (2002) Nucleic Acids Res. 30, 2817-2824[Abstract/Free Full Text]
19. Ballinger, S. W., Van Houten, B., Coklin, C. A., Jin, A., and Godley, B. (1999) Exp. Eye. Res. 68, 765-772[CrossRef][Medline] [Order article via Infotrieve]
20. Deng, G., Su, J. H., Ivins, K. J., Van Houten, B., and Cottman, C. (1999) Exp. Neurol. 159, 309-318[CrossRef][Medline] [Order article via Infotrieve]
21. Mandavilli, B. S., Ali, S. F., and Van Houten, B. (2000) Brain Res. 885, 45-52[CrossRef][Medline] [Order article via Infotrieve]
22. Santos, J. H., Mandavilli, B. S., and Van Houten, B. (2002) Methods Mol. Biol. 197, 159-176[Medline] [Order article via Infotrieve]
23. Sawyer, D. E., and Van Houten, B. (1999) Mutat. Res. 434, 161-176[Medline] [Order article via Infotrieve]
24. Garbe, J., Wong, M., Wigington, D., Yaswen, P., and Stampfer, M. R. (1999) Oncogene 18, 2169-2180[CrossRef][Medline] [Order article via Infotrieve]
25. Bowman, K. K., Sicard, D. M., Ford, J. M., and Hanawalt, P. C. (2000) Mol. Carcinog. 1, 17-24[CrossRef]
26. Ford, J. M., and Hanawalt, P. C. (1997) J. Biol. Chem. 272, 28073-28080[Abstract/Free Full Text]
27. Hoffschir, F., Vuillaume, M., Sabatier, L., Ricoul, M., Daya-Grosjean, L., Estrade, S., Cassingena, R., Calvayrac, R., Sarasin, A., and Dutrillaux, B. (1993) Carcinogenesis 14, 1569-1572[Abstract]
28. Ayala-Torres, S., Chen, Y., Svoboda, T., Rosenblatt, J., and Van Houten, B. (2000) Methods (Orlando) 22, 135-147[CrossRef]
29. Chen, H.-K., Yakes, F. M., Srivastava, D. K., Singhal, R. K., Sobol, R. W., Horton, J. K., Van Houten, B., and Wilson, S. H. (1998) Nucleic Acids Res. 26, 2001-2007[Abstract/Free Full Text]
30. Salvioli, S., Ardizzoni, A., Franceschi, C., and Cossarizza, A. (1997) FEBS Lett. 411, 77-82[CrossRef][Medline] [Order article via Infotrieve]
31. Bortner, C. D., and Cidlowski, J. A. (1999) J. Biol. Chem. 274, 21953-21962[Abstract/Free Full Text]
32. Mathur, A., Hong, Y., Kemp, B. K., Barrientos, A. A., and Erusalimsky, J. D. (2000) Cardiovasc. Res. 46, 126-138[CrossRef][Medline] [Order article via Infotrieve]
33. Haugland, R. (1996) in Handbook of Fluorescent Probes and Research Chemicals (Spence, M. T. Z., ed) , p. 269, Molecular Probes, Eugene, OR
34. Zhou, M., Diwu, Z., Panchuk-Voloshina, N., and Haugland, R. P. (1997) Annu. Rev. Biochem. 253, 162-168
35. Waterhouse, N. J., Ricci, J.-E., and Green, D. R. (2002) Biochimie (Paris) 84, 113-121
36. Neupert, W. (1997) Annu. Rev. Biochem. 66, 863-917[CrossRef][Medline] [Order article via Infotrieve]
37. Bohr, V. A., and Anson, R. M. (1999) J. Bioenerg. Biomembr. 31, 391-398[CrossRef][Medline] [Order article via Infotrieve]
38. Croteau, D. L., Stierum, R. H., and Bohr, V. A. (1999) Mutat. Res. 434, 137-148[Medline] [Order article via Infotrieve]
39. LeDoux, S. P., Driggers, W. J., Hollensworth, B. S., and Wilson, G. L. (1999) Mutat. Res. 434, 149-159[Medline] [Order article via Infotrieve]
40. Herrmann, J. M., and Neupert, W. (2000) Biochim. Biophys. Acta 1459, 331-338[Medline] [Order article via Infotrieve]
41. Hilbert, T. P., Chaung, W., Boorstein, R. J., Cunningham, R. P., and Teebor, G. W. (1997) J. Biol. Chem. 272, 6733-6740[Abstract/Free Full Text]
42. Yan, L.-J., Levine, R. L., and Sohal, R. S. (1997) Proc. Natl. Acad. Sci. U. S. A. 94, 11168-11172[Abstract/Free Full Text]
43. Flint, D. H., Smyk, R. E., Tuminello, J. F., Draczynska, L. B., and Brown, O. R. (1993) J. Biol. Chem. 268, 25547-25552[Abstract/Free Full Text]
44. Dobson, A. W., Xu, Y., Kelly, M. R., LeDoux, S. P., and Wilson, G. L. (2000) J. Biol. Chem. 275, 37518-37523[Abstract/Free Full Text]
45. Hollensworth, S. B., Shen, C., Sim, J. E., Spitz, D. R., Wilson, G. L., and LeDoux, S. P. (2000) Free Radic. Biol. Med. 28, 1161-1174[CrossRef][Medline] [Order article via Infotrieve]
46. Crawford, D. R., Wang, Y., Schools, G. P., Kochheiser, J., and Davies, J. K. (1997) Free Radic. Biol. Med. 22, 551-559[CrossRef][Medline] [Order article via Infotrieve]
47. Crawford, D. R., Lauzon, R. J., Wang, Y., Mazurkiewicz, J. E., Schools, G. P., and Davies, J. K. (1997) Free Radic. Biol. Med. 22, 1295-1300[CrossRef][Medline] [Order article via Infotrieve]
48. Crawford, D. R., Abramova, N. E., and Davies, K. J. (1998) Free Radic. Biol. Med. 25, 1106-1111[CrossRef][Medline] [Order article via Infotrieve]
49. Doetsch, P., Morey, N. J., Swanson, R. L., and Jinks-Robertson, S. (2001) Prog. Nucleic Acids Res. Mol. Biol. 68, 29-39[Medline] [Order article via Infotrieve]
50. Zhou, J., Ahn, J., Wilson, S. H., and Prives, C. (2001) EMBO J. 20, 914-923[Abstract/Free Full Text]


Copyright © 2003 by The American Society for Biochemistry and Molecular Biology, Inc.