Elevated levels of DNA ligase IIIα (LigIIIα) have been identified as a biomarker of an alteration in DNA repair in cancer cells that confers hypersensitivity to a LigIIIα inhibitor, L67, in combination with a poly (ADP-ribose) polymerase inhibitor. Because LigIIIα functions in the nucleus and mitochondria, we examined the effect of L67 on these organelles. Here, we show that, although the DNA ligase inhibitor selectively targets mitochondria, cancer and nonmalignant cells respond differently to disruption of mitochondrial DNA metabolism. Inhibition of mitochondrial LigIIIα in cancer cells resulted in abnormal mitochondrial morphology, reduced levels of mitochondrial DNA, and increased levels of mitochondrially generated reactive oxygen species that caused nuclear DNA damage. In contrast, these effects did not occur in nonmalignant cells. Furthermore, inhibition of mitochondrial LigIIIα activated a caspase 1–dependent apoptotic pathway, which is known to be part of inflammatory responses induced by pathogenic microorganisms in cancer, but not nonmalignant cells. These results demonstrate that the disruption of mitochondrial DNA metabolism elicits different responses in nonmalignant and cancer cells and suggests that the abnormal response in cancer cells may be exploited in the development of novel therapeutic strategies that selectively target cancer cells. Cancer Res; 76(18); 5431–41. ©2016 AACR.

Among the ATP-dependent DNA ligases encoded by the mammalian LIG genes, LIG1, LIG3, and LIG4 (1), DNA ligase I (LigI) is primarily responsible for joining Okazaki fragments during nuclear DNA replication. However, DNA ligase IIIα (LigIIIα) is essential for DNA replication in LigI-deficient cells (2–5). LigI and LigIIIα also appear to have overlapping functions in the repair of base damage and single-strand breaks (3–8). Although DNA ligase IV is predominantly responsible for the repair of nuclear DNA double-strand breaks (DSB) by nonhomologous end joining (NHEJ), LigI and Lig IIIα participate in alternative (alt) NHEJ pathways (9, 10).

Unlike the nucleus, only one DNA ligase is present in mitochondria (3, 4, 11). Mitochondrial (mito) and nuclear (nuc) versions of LigIIIα are generated by alternative translation initiation (11). Although mito LigIIIα is required to maintain mitochondrial DNA and is essential for cell viability under normal culture conditions, this lethality can be rescued by either addition of pyruvate and uridine to the culture media or expression of mitochondrially targeted, heterologous DNA ligases, including the NAD-dependent E. coli LigA (3, 4, 12).

A subset of DNA ligase inhibitors preferentially sensitized cancer cells to DNA-damaging agents (13). Subsequently, it was shown that BCR-ABL1–positive cell lines and samples from patients with chronic myeloid leukemia, in particular leukemia cells that had acquired resistance to imatinib, were hypersensitive to the LigI/III inhibitor L67 in combination with a PARP inhibitor (14). A similar hypersensitivity was observed in breast cancer cell lines with either intrinsic or acquired resistance to antiestrogens (15). Because LigIIIα knockdown had the same effect as L67 in combination with a PARP inhibitor, it appears that L67 exerts its cancer cell–specific effect by inhibition of LigIIIα (14, 15). The hypersensitivity to the combination of L67 and a PARP inhibitor correlated with elevated expression of both LigIIIα and PARP1, and increased dependence on PARP1- and LigIIIα-dependent alt NHEJ (9, 14–16).

Although the repair inhibitor combination does inhibit alt NHEJ (14, 15), the observed synergy is unlikely to be due to the inhibition of two enzymes in the same pathway (9). Because LigIIIα has nuclear and mitochondrial functions, we examined the mechanism of L67-induced cytotoxicity. These studies revealed that L67 preferentially targets mito LigIIIα, resulting in mitochondrial dysfunction. Surprisingly, cancer cell mitochondria were more susceptible to L67 than mitochondria in nonmalignant cells. The disruption of mitochondrial function in cancer cells resulted in elevated levels of mitochondrially generated reactive oxygen species (ROS) and activation of a caspase 1–dependent apoptotic pathway that is involved in inflammatory responses induced by pathogenic microorganisms (17). In nonmalignant cells, there was no increase in mitochondrially generated ROS, but oxidative phosphorylation (OXPHOS) was completely uncoupled and the cells became senescent.

Cell lines

Human cervical (HeLa, 2012), colorectal (HCT116, 2006 and 2016), and breast (MDA-MB-231, 2008) cancers cell lines were purchased from the ATCC and grown in the recommended media. A HeLa cell line that stably expresses mitochondrially targeted E. coli LigA (mitoLigA; ref. 4) after transfection with the plasmid pCAG-mitoLigAYFP-Neo that encodes E. coli LigA fused at its N terminus to the LigIIIα mitochondrial targeting sequence and at its C terminus to EYFP. The telomerase-immortalized human fibroblast cell line HCA-Ltrt from Dr. Murnane (2010) was grown in DMEM/F12 medium with 10% FBS. Normal breast epithelium MCF10A cells from Dr. Rassool (2012) were grown using recommended medium and mixture of additives (Lonza/Clonetics Corporation) with 5% horse serum and 100 ng/mL cholera toxin. Cell lines lacking mitochondria DNA (Rho minus cells) were established as described (18, 19). The identity of commercially available cell lines was confirmed by STR profiling with the PowerPlex 1.2 System (Promega), most recently in 2016.

Colony forming and cell growth assays

To measure colony formation, cells were cultured in triplicate in 6-well plates overnight and then were incubated with different concentrations of L67 for 14 days prior to staining with crystal violet. To measure cell growth, cells incubated with L67 for 48 to 72 hours. Genomic DNA was then stained with the CyQUANT NF reagent and quantitated according to the manufacturer's protocol.

Ligation assay

Human Lig IIIα/His-tagged XRCC1 complex was expressed in and purified from insect cells (20). E. coli DNA ligase (LigA) was purchased from New England Biolabs. A labeled duplex oligonucleotide with a single ligatable nick was prepared as described previously (21). Lig IIIα/XRCC1 (0.05 pmol) and LigA (0.1 units) were incubated with L67 in the presence of either ATP (for Lig IIIα/XRCC1) or NAD (for LigA) at room temperature for 10 minutes prior to the addition of the DNA substrate (0.1 pmol) and incubation in buffer containing 50 mmol/L Tris-Cl pH 7.5, 12.5 mmol/L NaCl, 6 mmol/L MgCl2, 3% glycerol, 1 mmol/L DTT, 0.25 mg/mL BSA, and 1% DMSO at 37°C for 20 minutes in a final volume of 20 μL. After the addition of SDS to a final concetario of 0.5%, DNA was recovered with Streptavidin MagneSphere beads (Promega), suspended in formamide/dye solution and electrophoresed through a 12.5% denaturing polyacrylamide gel. Ligated and unligated DNAs were detected by autoradiography and quantitated by ImageJ (NIH).

Immunoblotting

Cells were lysed in 20 mmol/L Tris-HCl, pH 7.5, 100 mmol/L NaCl, 1 mmol/L EDTA, 5% glycerol, and 1% Triton X-100 containing a protease inhibitor cocktail (Sigma). Proteins were detected by immunoblotting using the following antibodies: actin, phospho-KAP1(serine 824), (Abcam); Chk2, KAP1 (GeneTex), phospho-Chk2, p53, phospho-p53(serine 15), PARP1, cleaved PARP1 (Cell Signaling Technology); H2AX (R&D Systems); and γ-H2AX(serine 139) (Millipore). ATM (Cell Signaling Technology) and phospho-ATM (serine 1981) (Abcam) were detected as described (22).

Measurement of γ-H2AX and mitochondrial superoxide

Overnight cultures were incubated in fresh medium containing DMSO with different concentrations of L67 for 24 hours in the absence or presence of 5 mmol/L NAC. γ-H2AX and mitochondrial superoxide (mSOX) were detected by flow cytometry using an Alexa Fluor 647–conjugated antibody specific for γ-H2AX phosphorylated on serine139 and MitoSOX Red (Molecular Probes), respectively.

Seahorse extracellular flux analysis

In experiments performed on a Seahorse XF24 Flux analyzer, cells were maintained at 5% CO2 in DMEM media at 37°C. Cells (40,000) were plated on XF24 plates using Cell-Tak Cell and Tissue Adhesive (Corning), and growth medium was replaced with bicarbonate-free modified DMEM, the “assay medium.” In experiments performed on a Seahorse XF96e Flux analyzer, cells (20,000) were placed into each well using Cell-Tak. After incubation for another 60 minutes in a 37°C incubator without CO2, oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) were measured essentially as previously described (18).

Mitochondrial morphology and DNA content

Cells were seeded on slides and grown overnight in phenol-free DMEM medium prior to incubation in fresh medium containing either DMSO or 30 μmol/L L67 for 24 hours. Mitochondria were then stained by incubation with 250 nmol/L Mitotracker Red CMXRos for 20 minutes at 37°C. After washing, cells were maintained in phenol-free medium prior to imaging with a Zeiss LSM510 confocal system on an Axio Observer inverted microscope with a 63 × 1.2 NA water immersion objective. Mitotracker Red was excited with a 543 nm HeNe laser and fluorescence emission collected with 560 nm LP filter.

Genomic DNA was isolated using the QIAamp DNA Kit (QIAGEN) and then quantitated with the Quant-iT PicoGreen dsDNA Assay Kit (Molecular Probes). Short (220 bp) and long (8.9 kb) fragments of mitochondrial DNA and a 4 kb fragment of the nuc gene encoding DNA polymerase δ were amplified using 3 ng of total genomic DNA using the primers (Supplementary Table S2), amplification conditions (Supplementary Table S3), and a KAPA LongRange HotStart PCR kit (KAPA BIOSYSTEMS). Mitochondrial DNA copy number was calculated as described (19).

Apoptosis assay

Apoptotic cells were detected by flow cytometry using the PE Annexin V apoptosis detection Kit (BD Pharmingen) according to the manufacturer's instructions. Where indicated, a caspase 1 inhibitor (50 μmol/L Ac-YVAD-cmk; Sigma-Aldrich), a caspase 3 inhibitor (50 μmol/L Z-DEVD-fmk; ApexBio), or a pan caspase inhibitor (50 μmol/L Z-VAD-FMK; R&D Systems) was added to the media with L67 prior to incubation for 24 hours.

β-Galactosidase assay

Expression of β-galactosidase was detected using the Senescence β-Galactosidase Staining Kit (Cell Signaling Technology) according to the manufacturer's instructions.

Caspase 1 activity

Caspase 1 activity was measured by flow cytometry using a FLICA 660 in vitro caspase-1 detection Kit (Immunochemistry Technologies) according to the manufacturer's instructions.

Statistical analysis

Data are expressed as mean ± SEM. For comparison of groups, we used the Student two-tailed t test. A level of P < 0.05 was regarded as statistically significant.

Cells without mitochondrial DNA are more resistant to the DNA ligase inhibitor L67

To determine the mechanisms underlying the cytotoxicity of the LigI/III inhibitor L67 (13), derivatives of cell lines established from cervical (HeLa; Supplementary Fig. S1A), breast (MDA-MB-231), and colon (HCT116) cancers that lack mito DNA (Rho minus) were selected by growth in the presence of ethidium bromide, pyruvate, and uridine (18). HeLa cells with abnormal mitochondria that lack mitochondrial DNA (Supplementary Fig. S1A and S1B) were more resistant to L67 in colony forming (Fig. 1A) and cell proliferation assays (Fig. 1B) with the absence of mitochondrial DNA increasing the IC50 from 8.2 μmol/L to 29.7 μmol/L (Supplementary Table S1). The differential effect of L67 on HeLa and HeLa Rho minus cells was also observed when HeLa cells were grown in the media containing pyruvate and uridine (Supplementary Fig. S1C). MDA-MB-231 and HCT 116 cells lacking mitochondrial DNA were also more resistant to L67 compared with their respective parental cells (Fig. 1C and D; and Supplementary Table S1), suggesting that, at lower concentrations, L67 causes cell death by targeting mitochondrial function.

Figure 1.

Cancer cells lacking mitochondrial DNA are more resistant to L67. A, colony formation by HeLa, HeLaMLigA, and HeLa Rho minus cells incubated with either vehicle control (DMSO) or the indicated concentration of L67. Left plot, the boxes indicate that the wells containing cells incubated with vehicle control were in the same plate but not contiguous with the wells containing cells incubated with L67. Right plot, cell survival results are shown graphically. B–D, effect of L67 on the proliferation of HeLa (filled circle), HeLaMLigA (gray circle), and HeLa Rho minus cells (empty circle; B); MDA-MB-231 (filled square) and MDA-MB-231 Rho minus (empty square; C); HCT116 (filled triangle) and HCT116 Rho minus cells (empty triangle; D) was determined as described in Materials and Methods. Data shown graphically are the mean ± SEM of three independent experiments and are expressed as a percentage of the values for untreated cells.

Figure 1.

Cancer cells lacking mitochondrial DNA are more resistant to L67. A, colony formation by HeLa, HeLaMLigA, and HeLa Rho minus cells incubated with either vehicle control (DMSO) or the indicated concentration of L67. Left plot, the boxes indicate that the wells containing cells incubated with vehicle control were in the same plate but not contiguous with the wells containing cells incubated with L67. Right plot, cell survival results are shown graphically. B–D, effect of L67 on the proliferation of HeLa (filled circle), HeLaMLigA (gray circle), and HeLa Rho minus cells (empty circle; B); MDA-MB-231 (filled square) and MDA-MB-231 Rho minus (empty square; C); HCT116 (filled triangle) and HCT116 Rho minus cells (empty triangle; D) was determined as described in Materials and Methods. Data shown graphically are the mean ± SEM of three independent experiments and are expressed as a percentage of the values for untreated cells.

Close modal

To confirm that the effect of L67 is dependent upon inhibition of mito LigIIIα, we stably expressed mitoLigA in HeLa cells (Supplementary Fig. S2A). Although this enzyme can substitute for mitoLigIIIα (4), it is at least 6-fold more resistant to L67 than LigIIIα (Supplementary Fig. S2B). Notably, expression of mitoLigA significantly increased the resistance of HeLa cells to L67 (Fig. 1A and B) albeit not to same level as the HeLa Rho minus cells (Supplementary Table S1). This may be due in part to a dominant-negative effect of inhibiting endogenous LigIIIα.

Inhibition of mito LigIIIα results in increased mSOX and nuclear DNA damage in HeLa cells

The differential effect of L67 on the growth of parental and Rho minus cancer cells was most pronounced in a concentration range (5 to 15 μmol/L; Fig. 1A–D) that encompasses the IC50 values for LigI (6 μmol/L) and LigIII (8 μmol/L; ref. 13). Although LigI and LigIIIα participate in nuc DNA replication (2, 5), incubation of synchronized cells with L67 at concentrations up to 15 μmol/L for 24 hours had only minor effects on cycle distribution (Supplementary Fig. S3A). Furthermore, incubation with L67 concentrations up to 10 μmol/L for 24 hours only reduced bromodeoxyuridine (BrdUrd) incorporation by 10% to 15% (Supplementary Fig. S3B). At higher concentrations of L67, there was an accumulation of HeLa cells, the derivative expressing mitoLigA, and, to a lesser extent, Rho minus cells in S phase (Supplementary Fig. S3A) that correlated with reduced BrdUrd incorporation (Supplementary Fig. S3B).

Interestingly, incubation of HeLa, but not HeLa Rho minus cells, with L67 at 10 and 15 μmol/L for 24 hours resulted in a dose-dependent increase in the formation of nuclear γH2AX foci (Supplementary Fig. S4A) and steady-state levels of γH2AX (Fig. 2A), consistent with replication stress and DSB induction in the nucleus (23). Expression of mitoLigA reduced γH2AX foci formation (Supplementary Fig. S4A) and the steady-state levels (Fig. 2A) induced by L67 compared with the parental cells, but not to the extent observed in Rho minus cells (Fig. 2A and Supplementary Fig. S4A). Because mitochondria are the major cellular source of reactive oxygen species (ROS) and disruption of mitochondrial function can lead to elevated levels of ROS (24, 25), we examined the effect of L67 on mSOX levels. Incubation with L67 for 24 hours resulted in a concentration-dependent increase in mSOX levels in HeLa cells, whereas expression of mitoLigA reduced the levels of mSOX induced by L67 and, as expected, there was no induction of mSOX in HeLa Rho minus cells (Fig. 2B).

Figure 2.

Elevated levels of γH2AX, mSOX, and ATM activation induced by L67 in HeLa cells. Effect of L67 is attenuated by either the absence of mito DNA, expression of mitoLigA, or coincubation with an antioxidant. γH2AX (A) and mSOX (B) in HeLa (filled circle), HeLaMLigA (gray circle), and HeLa Rho minus cells (empty circle) were detected by flow cytometry. C, the indicated proteins and their phosphorylated derivatives in extracts from HeLa (left) and HeLaMLigA (right) cells that had been incubated with or without (Not) L67 for 24 hours were detected by immunoblotting. Where indicated, cells were incubated with 100 μmol/L H202 for 1 hour to induce the DNA damage response. D and E, effect of the absence (black bars) or presence (white bars) of 5 mmol/L NAC on L67-induced γH2AX (D) and mSOX (E) in HeLa cells. Data shown graphically are the mean ± SEM of three independent experiments and are expressed as a percentage of the values for untreated cells. *, P < 0.05 and ***, P < 0.0001 using the unpaired two-tailed Student test.

Figure 2.

Elevated levels of γH2AX, mSOX, and ATM activation induced by L67 in HeLa cells. Effect of L67 is attenuated by either the absence of mito DNA, expression of mitoLigA, or coincubation with an antioxidant. γH2AX (A) and mSOX (B) in HeLa (filled circle), HeLaMLigA (gray circle), and HeLa Rho minus cells (empty circle) were detected by flow cytometry. C, the indicated proteins and their phosphorylated derivatives in extracts from HeLa (left) and HeLaMLigA (right) cells that had been incubated with or without (Not) L67 for 24 hours were detected by immunoblotting. Where indicated, cells were incubated with 100 μmol/L H202 for 1 hour to induce the DNA damage response. D and E, effect of the absence (black bars) or presence (white bars) of 5 mmol/L NAC on L67-induced γH2AX (D) and mSOX (E) in HeLa cells. Data shown graphically are the mean ± SEM of three independent experiments and are expressed as a percentage of the values for untreated cells. *, P < 0.05 and ***, P < 0.0001 using the unpaired two-tailed Student test.

Close modal

Because γH2AX can be generated in response to other types of cellular stress in addition to DSBs (26), we examined the phosphorylation status of the ataxia telangiectasia mutated (ATM) kinase, which is recruited to and activated by DSBs, and ATM substrates (27). Expression of mitoLigA greatly attenuated L67-induced autophosphorylation of ATM and phosphorylation of γH2AX and several other ATM substrates but not p53, which was constitutively phosphorylated (Fig. 2C, compare left and right plots). While expression of mitoLigA attenuated the effects of L67 on cell survival and mSOX levels, mitochondrial function was abnormal (Fig. 4), suggesting that p53 may be activated in response to metabolic stress in these cells (28). Although ATM can be activated by DSB-independent mechanisms, including direct activation by ROS (27, 29), incubation with L67 resulted in phosphorylation of KAP1, whose phosphorylation by ATM is DSB-dependent (Fig. 2C; ref. 30). As expected, the free radical scavenger N-acetyl cysteine (NAC) reduced mSOX (Fig. 2E), ATM activation (Supplementary Fig. S4B), and γH2AX formation (Fig. 2D) induced by L67 in HeLa cells. Together, these results indicate that the nuclear DNA damage induced by L67 in HeLa cells is due to increased generation of ROS by mitochondria as result of inhibition of mito LigIIIα.

L67 induces elevated mSOX in cancer but not nonmalignant cell lines

Similar to the cancer cell lines (Fig. 1), proliferation of a Rho minus derivative of a telomerase-immortalized fibroblast (HCA-Ltrt) was more resistant to L67 than the parental cell line (Fig. 3A). Surprisingly, while L67 had similar effects on cell-cycle distribution (Supplementary Figs. S3A and S5A) and the proliferation of cancer cell lines (Fig. 1A–D) and nonmalignant cell lines established from normal tissues (Fig. 3A), elevated levels of mSOX were not detected in nonmalignant cells, including HCA-Ltrt Rho minus cells (Fig. 3B and Supplementary Fig. S5B). Accordingly, L67 did not induce detectable phosphorylation of H2AX and KAP1 in HCA-Ltrt cells, and ATM autophosphorylation was only observed with 30 μmol/L L67 (Fig. 3C). Interestingly, phosphorylation of p53 and Chk2 occurred in the absence of detectable ATM activation (Fig. 3C), suggesting a response to a stress signal other than DNA damage (28).

Figure 3.

Effect of L67 on the proliferation of nonmalignant cells; no induction of mSOX and reduced ATM activation by L67 in nonmalignant cell lines. A, proliferation of telomerase-immortalized fibroblasts HCA-Ltrt (filled diamond), HCA-Ltrt Rho minus cells (empty diamond), and MCF10A normal breast epithelial cells (crossed gray square). B, mSOX levels in HCA-Ltrt (filled diamond), MCF10A (crossed gray square), MDA-MB-231 (filled square), and HCT116 (filled triangle) cells. Cells were incubated in the presence or absence of L67 for 24 hours. Data shown graphically are the mean ± SEM of three independent experiments and are expressed as a percentage of the values for untreated cells. C, the indicated proteins and their phosphorylated derivatives in extracts from HCA-Ltrt cells that had been incubated with or without (Not) L67 for 24 hours were detected by immunoblotting. Where indicated, cells were incubated with 100 μmol/L H202 for 1 hour to induce the DNA damage response.

Figure 3.

Effect of L67 on the proliferation of nonmalignant cells; no induction of mSOX and reduced ATM activation by L67 in nonmalignant cell lines. A, proliferation of telomerase-immortalized fibroblasts HCA-Ltrt (filled diamond), HCA-Ltrt Rho minus cells (empty diamond), and MCF10A normal breast epithelial cells (crossed gray square). B, mSOX levels in HCA-Ltrt (filled diamond), MCF10A (crossed gray square), MDA-MB-231 (filled square), and HCT116 (filled triangle) cells. Cells were incubated in the presence or absence of L67 for 24 hours. Data shown graphically are the mean ± SEM of three independent experiments and are expressed as a percentage of the values for untreated cells. C, the indicated proteins and their phosphorylated derivatives in extracts from HCA-Ltrt cells that had been incubated with or without (Not) L67 for 24 hours were detected by immunoblotting. Where indicated, cells were incubated with 100 μmol/L H202 for 1 hour to induce the DNA damage response.

Close modal

L67 induces changes in mitochondrial function in both cancer and nonmalignant cells, but alterations in mitochondrial morphology and DNA copy number occur only in cancer cells

As mentioned previously, expression of mitoLigA in HeLa cells resulted in altered mitochondrial function. The OCR was about 50% lower compared with the parental HeLa cells (Fig. 4A), whereas the proportion of oligomycin-insensitive oxygen consumption, indicative of uncoupled OXPHOS, was higher (∼40%) compared with the parental HeLa cells (∼20%). Incubation with L67 resulted in a similar reduction in OCR (40%–50%) in both cell lines with about 80% of the remaining OCR due to uncoupled OXPHOS (Fig. 4A). The changes in OCR in the HeLa cells expressing mitoLigA resulted in a compensatory increase in ECAR, a measure of glycolysis. As expected, there was an increase in ECAR following incubation with L67 (Fig. 4B).

Figure 4.

L67 causes a reduction in OCR and mitochondrial DNA, and abnormal mitochondrial morphology in HeLa but not HCA-Ltrt cells. HeLa (red bars) and HeLaMLigA (black bars) cells were incubated with (+) or without (−) 10 μmol/L L67 for 24 hours. A, OCR measured in the presence (+) or absence (−) of 1 μmol/L oligomycin (Oligo). B, ECAR, OCR, and ECAR were measured in a Seahorse Bioanalyzer as described in Materials and Methods. OCR and ECAR values for untreated HeLa and HCA-Ltrt cells values were set to 100%. Data shown graphically represent mean ± SEM of 4 to 6 replicates and 3 to 4 time points. For A and B, all treatment groups are significantly different than their untreated control with P values <0.001. C, effect of L67 on mitochondrial DNA copy number was determined in HeLa (red bars), HeLaMLigA (black bars), and HCA-Ltrt (gray bars) cells as described in Materials and Methods after incubation in the presence or absence (Not Treated) of L67 for 24 hours. Data shown graphically are the mean ± SEM of three independent experiments. *, P < 0.05; **, P < 0.001. D, mitochondria in HeLa and HeLaMLigA cells were stained with Mitotracker Red and visualized by fluorescence microscopy after incubation in the presence or absence (Not Treated) of L67 for 24 hours. Scale bars, 10 μm. OCR (E) and ECAR (F) were measured in HCA-Ltrt (gray bars) cells incubated with (+) or without (−) 10 μmol/L L67 for 24 hours as described above. E, OCR, with the exception of L67 treatment, all treatment groups are significantly different than the untreated control with a P value of <0.001 for oligo and a P value of <0.05 for oligo plus L67. F, ECAR, the treatment group is significantly different than the untreated control with a P value <0.001. G, mitochondria in HCA-Ltrt and MCF10A cells were visualized as described above after incubation in the presence or absence (Not Treated) of L67 for 24 hours.

Figure 4.

L67 causes a reduction in OCR and mitochondrial DNA, and abnormal mitochondrial morphology in HeLa but not HCA-Ltrt cells. HeLa (red bars) and HeLaMLigA (black bars) cells were incubated with (+) or without (−) 10 μmol/L L67 for 24 hours. A, OCR measured in the presence (+) or absence (−) of 1 μmol/L oligomycin (Oligo). B, ECAR, OCR, and ECAR were measured in a Seahorse Bioanalyzer as described in Materials and Methods. OCR and ECAR values for untreated HeLa and HCA-Ltrt cells values were set to 100%. Data shown graphically represent mean ± SEM of 4 to 6 replicates and 3 to 4 time points. For A and B, all treatment groups are significantly different than their untreated control with P values <0.001. C, effect of L67 on mitochondrial DNA copy number was determined in HeLa (red bars), HeLaMLigA (black bars), and HCA-Ltrt (gray bars) cells as described in Materials and Methods after incubation in the presence or absence (Not Treated) of L67 for 24 hours. Data shown graphically are the mean ± SEM of three independent experiments. *, P < 0.05; **, P < 0.001. D, mitochondria in HeLa and HeLaMLigA cells were stained with Mitotracker Red and visualized by fluorescence microscopy after incubation in the presence or absence (Not Treated) of L67 for 24 hours. Scale bars, 10 μm. OCR (E) and ECAR (F) were measured in HCA-Ltrt (gray bars) cells incubated with (+) or without (−) 10 μmol/L L67 for 24 hours as described above. E, OCR, with the exception of L67 treatment, all treatment groups are significantly different than the untreated control with a P value of <0.001 for oligo and a P value of <0.05 for oligo plus L67. F, ECAR, the treatment group is significantly different than the untreated control with a P value <0.001. G, mitochondria in HCA-Ltrt and MCF10A cells were visualized as described above after incubation in the presence or absence (Not Treated) of L67 for 24 hours.

Close modal

Because reduced mito DNA has been linked with elevated mSOX and increased sensitivity to DNA-damaging agents (31), we examined the effect of L67 on mito DNA copy number. Incubation of HeLa cells with 5 and 10 μmol/L L67 resulted in about a 25% reduction in mito DNA, whereas there was no reduction in mito DNA in derivatives expressing mitoLigA (Fig. 4C). Furthermore, incubation with 30 μmol/L L67 disrupted the filamentous network of mitochondria observed in untreated HeLa cells, resulting in a similar appearance to HeLa Rho minus cells with large irregular clumps of mitochondria and diffuse cell staining (Fig. 3D; Supplementary Fig. S1B). Thus, while expression of mitoLigA resulted in reduced OCR (Fig. 4A) and increased ECAR (Fig. 4B), it prevented the reduction of mito DNA and abnormal mitochondrial morphology induced by L67 (Fig. 4D). Notably, incubation of telomerase-immortalized fibroblasts with 10 μmol/L L67 did not cause a reduction in OCR, but OXPHOS was totally uncoupled (high OCR in presence of both L67 and oligomycin; Fig. 4E) and there was a large compensatory increase in glycolysis (Fig. 4F). In addition, there were no reductions in mitochondrial DNA copy number (Fig. 4C) or alterations in mitochondrial morphology (Fig. 4G) in nonmalignant cells incubated with L67.

Inhibition of mito LigIIIα induces apoptosis in cancer cells and senescence in nonmalignant cells

Following incubation with 10 μmol/L L67 for 24 hours, there were greater than 12-fold more apoptotic (7-AAD positive, PE Annexin V positive) HeLa and HeLa mitoLigA cells than apoptotic HeLa Rho minus cells (Fig. 5A), indicating that the majority of cell death induced by L67 is dependent upon mito DNA and presumably mitochondrial function. Although, at 10 μmol/L L67, the extent of apoptosis was similar in the HeLa and HeLa mitoLigA cells (Fig. 5A), cell death was caspase-dependent in HeLa cells (Fig. 6) and caspase-independent in the HeLa mitoLigA cells (Fig. 6A and Supplementary Fig. S6). There was greater apoptosis in the HeLa cells compared with HeLa cells expressing mitoLigA at 100 μmol/L L67 with apoptotic cells constituting about 50% of the HeLa cell population compared with 23% of the HeLa mitoLigA population (Fig. 5A), indicating that expression of mitoLigA reduces the cytotoxicity of L67. In contrast, very low levels of apoptosis were detected in HCA-Ltrt cells incubated with L67 (Fig. 5A). Because L67 reduced the proliferation of nonmalignant cells (Fig. 3A), we asked whether L67 induced senescence. As shown in Fig. 5B, incubation of HCA-Ltrt and MCF10A cells with L67 resulted in β-galactosidase expression, a marker of senescence. Thus, nonmalignant and cancer cells respond differently to inhibition of mito LigIIIα with nonmalignant cells becoming senescent, whereas cancer cells undergo apoptosis.

Figure 5.

L67 induces apoptosis in HeLa cells and senescence in nonmalignant cells. A, the fraction of late (top right quadrant) and early (bottom right) apoptotic cells in cultures of HeLa, HeLa Rho minus, HeLaMLigA, and HCA-Ltrt cells after incubation in the presence or absence (Not Treated) of L67 for 24 hours was determined by flow cytometry. B, β-galactosidase activity in HCA-Ltrt and MCF10A cells after incubation in the presence or absence (Not Treated) of L67 for 24 hours was detected as described in Materials and Methods. Scale bars, 50 μm.

Figure 5.

L67 induces apoptosis in HeLa cells and senescence in nonmalignant cells. A, the fraction of late (top right quadrant) and early (bottom right) apoptotic cells in cultures of HeLa, HeLa Rho minus, HeLaMLigA, and HCA-Ltrt cells after incubation in the presence or absence (Not Treated) of L67 for 24 hours was determined by flow cytometry. B, β-galactosidase activity in HCA-Ltrt and MCF10A cells after incubation in the presence or absence (Not Treated) of L67 for 24 hours was detected as described in Materials and Methods. Scale bars, 50 μm.

Close modal
Figure 6.

L67 induces caspase 1–dependent apoptosis in HeLa cells; attenuation of L67-induced apoptosis by loss of mitochondrial DNA, expression of mitoLigA, or inhibition of caspase 1. A, HeLa (filled circle), HeLaMLigA (gray circle), and HeLa Rho minus (empty circle). B, HCA-Ltrt (filled diamond), MCF10A (crossed gray square), MDA-MB-231 (filled square), and HCT116 (filled triangle) cells were incubated with or without L67 for 24 hours. Caspase 1 activity was measured by flow cytometry. Data shown graphically are the mean ± SEM of three independent experiments. C, the effects of a caspase 3 inhibitor, a caspase 1 inhibitor, and a pan caspase inhibitor on L67-induced apoptosis in HeLa cells.

Figure 6.

L67 induces caspase 1–dependent apoptosis in HeLa cells; attenuation of L67-induced apoptosis by loss of mitochondrial DNA, expression of mitoLigA, or inhibition of caspase 1. A, HeLa (filled circle), HeLaMLigA (gray circle), and HeLa Rho minus (empty circle). B, HCA-Ltrt (filled diamond), MCF10A (crossed gray square), MDA-MB-231 (filled square), and HCT116 (filled triangle) cells were incubated with or without L67 for 24 hours. Caspase 1 activity was measured by flow cytometry. Data shown graphically are the mean ± SEM of three independent experiments. C, the effects of a caspase 3 inhibitor, a caspase 1 inhibitor, and a pan caspase inhibitor on L67-induced apoptosis in HeLa cells.

Close modal

Inhibition of mito LigIIIα activates a caspase 1–dependent cell death pathway in cancer but not nonmalignant cells

No activation of caspases 7 and 9 that are involved in extrinsic and intrinsic apoptotic pathways was detected in HeLa cells incubated with L67 (Supplementary Fig. S6). Instead, caspase 1, which is activated in the inflammatory response elicited by pathogens (17) and by the release of ROS and mito DNA into the cytoplasm (32–34), was activated in HeLa cells but not in either the Rho minus or mitoLigA-expressing derivatives (Fig. 6A). Notably, incubation with L67 also activated caspase 1 in breast (MDA-MB-231) and colorectal (HCT116) cancer cell lines but not in nonmalignant cell lines (Fig. 6B). To determine whether cell death is dependent upon caspase 1 activation, we examined the effect of the caspase inhibitors on L67-induced apoptosis in HeLa cells. As shown in Fig. 6C, apoptosis was reduced by a caspase 1 inhibitor and a pan caspase inhibitor but not a caspase 3 inhibitor, confirming that cell death was mediated by caspase 1. Together, these results demonstrate that inhibition of mito LigIIIα selectively induces cell death in cancer cells by activating a caspase 1–dependent apoptotic pathway.

Previous studies have shown that therapy-resistant forms of breast cancer and leukemia with elevated levels of LigIIIα and PARP-1 are hypersensitive to inhibition of LigIIIα in combination with a PARP inhibitor (14, 15). Here, we have shown that the LigIIIα inhibitor, L67, used in those studies preferentially targets mito LigIIIα and, at the concentrations used to preferentially kill therapy-resistant leukemia and breast cancer cells in combination with a PARP inhibitor (14, 15), had no effect on nuclear DNA replication. Importantly, while L67 affected mitochondrial function in both cancer and nonmalignant cells, the effects and outcomes were very different. In cancer cell lines, incubation with L67 resulted in elevated levels of mitochondrially generated ROS, abnormal mitochondrial morphology, reductions in OCR and mitochondrial DNA, and caspase 1–dependent cell death. Expression of L67-resistant mitoLigA in HeLa cells partially suppressed the increase in mSOX, and completely prevented changes in mitochondrial DNA copy number and morphology, and activation of caspase 1, demonstrating that the effects of L67 were dependent upon inhibition of mito LigIIIα. In contrast to cancer cells, L67 did not alter mitochondrial morphology, mitochondrial DNA copy number, or mSOX levels and did not activate caspase 1 in nonmalignant cells. Instead of apoptosis, L67 induced significant uncoupling of mitochondria with a compensatory increase in glycolysis and senescence in nonmalignant cells. Thus, cancer cells are less able to mitigate the deleterious effects caused by disruption of mitochondrial DNA metabolism than nonmalignant cells.

Because inhibition of mito LigIIIα did not cause an increase in steady-state levels of mSOX in nonmalignant cells even though there was no reduction in the OCR, we suggest that initial increases in the levels of mito DNA damage and/or mSOX activate antioxidant responses in nonmalignant cells that prevent further increases in mSOX. Notably, we found that OXPHOS was totally uncoupled in the nonmalignant cell line incubated with L67. Because the uncoupling of oxygen consumption from electron transport reduces the generation of ROS by the electron transport chain (35), this alteration in nonmalignant cells presumably attenuates increases in mSOX. In addition, nonmalignant cells may be able to prevent the increase in mitochondrially generated ROS by removing dysfunctional, damaged mitochondria via autophagy (32, 34, 36). Although mito dysfunction caused by inhibition of mito LigIIIα did not result in elevated levels of ROS, p53 was activated, presumably in response to the alterations in cellular energy metabolism, and may contribute to cellular senescence (28). The increase in glycolysis may either be an effort to compensate for the uncoupled OXPHOS or an indicator of senescence (37).

There are conflicting reports as to the effect of LigIIIα knockdown on mitochondrial DNA (12, 38). In accord with the normal appearance of mitochondria in L67-treated nonmalignant cells, these cells were able to maintain their mitochondrial DNA copy number. Recent studies have shown that there are at least two modes of mitochondrial DNA replication, one of which involves a conventional coupled leading and lagging strand, whereas the other one involves RNA incorporation throughout the lagging strand (39). Because the latter mechanism does not require the joining of multiple Okazaki fragments (39), it is possible that nonmalignant cells are able to maintain mitochondrial DNA copy number by switching to this relatively DNA ligase–independent replication mechanism.

It is well established that cancers driven by oncogenes, such as Ras, c-Myc, BCR-ABL1, and FLT3-ITD, have elevated levels of ROS (40–42). While this increase in ROS activates signaling pathways that promote cell proliferation and metastasis and inhibit cell death pathways (43), redox homeostasis is presumably deregulated as part of the adaptation to constitutively higher ROS levels. Because treatment with an exogenous oxidizing agent induces elevated levels of mSOX and degradation of mitochondrial DNA (44–46) and reduced levels of LigIIIα increase the ROS-induced degradation of mitochondrial DNA (12), our results suggest that the elevated mSOX induced by L67 in cancer cells results from an inability to activate mechanisms, such as OXPHOS uncoupling and mitophagy, that reduce mitochondrially generated ROS (35, 36). Consequently, this initiates a vicious cycle in which elevated mSOX levels cause more DNA damage, resulting in reduced mitochondrial DNA copy number and more mito dysfunction that in turn generates more mSOX (31, 45, 47). Interestingly, expression of mitoLigA in HeLa cells did not totally prevent the increase in mSOX induced by L67, but fully maintained mitochondrial DNA copy number. Thus, our results support the idea that a combination of increased mSOX and inhibition of mito LigIIIα results in the degradation of mito DNA in HeLa cells.

L67-induced cell death in cancer cells occurred via a caspase 1–dependent apoptotic pathway. Although caspase 1 can be activated by either inflammasomes or pyroptosomes (17), it seems likely that activation of caspase 1 by L67 is mediated by the inflammasome because ROS and mitochondrial DNA have been shown to activate the NLRP3 inflammasome (32–34). Thus, we suggest that the L67-induced mitochondrial dysfunction triggers the mitochondrial permeability transition (48) in cancer cells with the subsequent release of superoxide and mito DNA into the cytoplasm, activating the inflammasome and caspase 1–dependent apoptosis (32–34).

Here, we have shown that cancer cells are unable to effectively respond to the deleterious effects caused by the inhibition of LigIIIα, a key enzyme in mitochondrial DNA metabolism. Notably, the elevated levels of mitochondrially generated ROS in cancer cells incubated with L67 result in increased nuclear DNA damage, suggesting that L67-induced mitochondria-dependent nuclear DNA damage underlies the synergistic activity of L67 with PARP inhibitors that reduce nuclear DNA repair (14, 15). Furthermore, our studies showing that regulation of mitochondrial function is abnormal in cancer cells not only provide further evidence that strategies targeting cancer cell mitochondria may have utility in cancer therapy (49, 50), but may lead to the identification of novel therapeutic approaches that exploit these differences to selectively eliminate cancer cells by activating caspase 1–dependent apoptosis.

A.E Tomkinson is Associate Editor at DNA Repair Journal, has ownership interest (including patents) in University of Maryland US patent 8,445.537 and US patent 9,132,120, and is a consultant/advisory board member for University of Maryland Cancer Center. No potential conflicts of interest were disclosed by the other authors.

Conception and design: A. Sallmyr, V. Roginskaya, B. Van Houten, A.E. Tomkinson

Development of methodology: A. Sallmyr, Y. Matsumoto, B. Van Houten

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): A. Sallmyr, V. Roginskaya, B. Van Houten

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): A. Sallmyr, V. Roginskaya, B. Van Houten, A.E. Tomkinson

Writing, review, and/or revision of the manuscript: A. Sallmyr, B. Van Houten, A.E. Tomkinson

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): V. Roginskaya

Study supervision: A. Sallmyr, B. Van Houten, A.E. Tomkinson

We thank Dr. Maria Jasin for the plasmid construct encoding mitoLigA and Dr. George Greco for the synthesis of L67.

This work was supported by US NIH Grants (R01 ES012512 and P01 CA92584 to A.E. Tomkinson). Flow cytometry and microscopy were carried out in University of New Mexico Cancer Center Shared Resources supported by NCI Cancer Center Support Grant P30 CA11800. This work was also supported in part by PA CURE from the Department of Health, PA (B. Van Houten).

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

1.
Ellenberger
T
,
Tomkinson
AE
. 
Eukaryotic DNA ligases: Structural and functional insights
.
Annu Rev Biochem
2008
;
77
:
313
38
.
2.
Arakawa
H
,
Bednar
T
,
Wang
M
,
Paul
K
,
Mladenov
E
,
Bencsik-Theilen
AA
, et al
Functional redundancy between DNA ligases I and III in DNA replication in vertebrate cells
.
Nucleic Acids Res
2012
;
40
:
2599
610
.
3.
Gao
Y
,
Katyal
S
,
Lee
Y
,
Zhao
J
,
Rehg
JE
,
Russell
HR
, et al
DNA ligase III is critical for mtDNA integrity but not Xrcc1-mediated nuclear DNA repair
.
Nature
2011
;
471
:
240
4
.
4.
Simsek
D
,
Furda
A
,
Gao
Y
,
Artus
J
,
Brunet
E
,
Hadjantonakis
AK
, et al
Crucial role for DNA ligase III in mitochondria but not in Xrcc1-dependent repair
.
Nature
2011
;
471
:
245
8
.
5.
Le Chalony
C
,
Hoffschir
F
,
Gauthier
LR
,
Gross
J
,
Biard
DS
,
Boussin
FD
, et al
Partial complementation of a DNA ligase I deficiency by DNA ligase III and its impact on cell survival and telomere stability in mammalian cells
.
Cell Mol Life Sci
2012
;
69
:
2933
49
.
6.
Barnes
DE
,
Tomkinson
AE
,
Lehmann
AR
,
Webster
AD
,
Lindahl
T
. 
Mutations in the DNA ligase I gene of an individual with immunodeficiencies and cellular hypersensitivity to DNA-damaging agents
.
Cell
1992
;
69
:
495
503
.
7.
Bentley
DJ
,
Harrison
C
,
Ketchen
AM
,
Redhead
NJ
,
Samuel
K
,
Waterfall
M
, et al
DNA ligase I null mouse cells show normal DNA repair activity but altered DNA replication and reduced genome stability
.
J Cell Sci
2002
;
115
(
Pt 7
):
1551
61
.
8.
Harrison
C
,
Ketchen
AM
,
Redhead
NJ
,
O'Sullivan
MJ
,
Melton
DW
. 
Replication failure, genome instability, and increased cancer susceptibility in mice with a point mutation in the DNA ligase I gene
.
Cancer Res
2002
;
62
:
4065
74
.
9.
Audebert
M
,
Salles
B
,
Calsou
P
. 
Involvement of poly(ADP-ribose) polymerase-1 and XRCC1/DNA ligase III in an alternative route for DNA double-strand breaks rejoining
.
J Biol Chem
2004
;
279
:
55117
26
.
10.
Simsek
D
,
Brunet
E
,
Wong
SY
,
Katyal
S
,
Gao
Y
,
McKinnon
PJ
, et al
DNA ligase III promotes alternative nonhomologous end-joining during chromosomal translocation formation
.
PLoS Genet
2011
;
7
:
e1002080
.
11.
Lakshmipathy
U
,
Campbell
C
. 
The human DNA ligase III gene encodes nuclear and mitochondrial proteins
.
Mol Cell Biol
1999
;
19
:
3869
76
.
12.
Shokolenko
IN
,
Fayzulin
RZ
,
Katyal
S
,
McKinnon
PJ
,
Wilson
GL
,
Alexeyev
MF
. 
Mitochondrial DNA ligase is dispensable for the viability of cultured cells but essential for mtDNA maintenance
.
J Biol Chem
2013
;
288
:
26594
605
.
13.
Chen
X
,
Zhong
S
,
Zhu
X
,
Dziegielewska
B
,
Ellenberger
T
,
Wilson
GM
, et al
Rational design of human DNA ligase inhibitors that target cellular DNA replication and repair
.
Cancer Res
2008
;
68
:
3169
77
.
14.
Tobin
LA
,
Robert
C
,
Rapoport
AP
,
Gojo
I
,
Baer
MR
,
Tomkinson
AE
, et al
Targeting abnormal DNA double strand break repair in tyrosine kinase inhibitor-resistant chronic myeloid leukemias
.
Oncogene
2013
;
32
:
1784
93
.
15.
Tobin
LA
,
Robert
C
,
Nagaria
P
,
Chumsri
S
,
Twaddell
W
,
Ioffe
OB
, et al
Targeting abnormal DNA repair in therapy-resistant breast cancers
.
Mol Cancer Res
2012
;
10
:
96
107
.
16.
Sallmyr
A
,
Tomkinson
AE
,
Rassool
F
. 
Up-regulation of WRN and DNA ligase IIIα in chromic myeloid leukemia: Consequences for the repair of DNA double strand breaks
.
Blood
2008
;
112
:
1413
23
.
17.
Latz
E
,
Xiao
TS
,
Stutz
A
. 
Activation and regulation of the inflammasomes
.
Nat Rev Immunol
2013
;
13
:
397
411
.
18.
Qian
W
,
Van Houten
B
. 
Alterations in bioenergetics due to changes in mitochondrial DNA copy number
.
Methods
2010
;
51
:
452
7
.
19.
Santos
JH
,
Meyer
JN
,
Mandavilli
BS
,
Van Houten
B
. 
Quantitative PCR-based measurement of nuclear and mitochondrial DNA damage and repair in mammalian cells
.
Methods Mol Biol
2006
;
314
:
183
99
.
20.
Della-Maria
J
,
Zhou
Y
,
Tsai
MS
,
Kuhnlein
J
,
Carney
JP
,
Paull
TT
, et al
Human Mre11/Human Rad50/Nbs1 and DNA ligase IIIα/XRCC1 protein complexes act together in an alternative nonhomologous end joining pathway
.
J Biol Chem
2011
;
286
:
33845
53
.
21.
Song
W
,
Levin
DS
,
Varkey
J
,
Post
S
,
Bermudez
VP
,
Hurwitz
J
, et al
A conserved
physical and functional interaction between the cell cycle checkpoint clamp loader and DNA ligase I of eukaryotes
.
J Biol Chem
2007
;
282
:
22721
30
.
22.
Gamper
AM
,
Rofougaran
R
,
Watkins
SC
,
Greenberger
JS
,
Beumer
JH
,
Bakkenist
CJ
. 
ATR kinase activation in G1 phase facilitates the repair of ionizing radiation-induced DNA damage
.
Nucleic Acids Res
2013
;
41
:
10334
44
.
23.
Bonner
WM
,
Redon
CE
,
Dickey
JS
,
Nakamura
AJ
,
Sedelnikova
OA
,
Solier
S
, et al
GammaH2AX and cancer
.
Nat Rev Cancer
2008
;
8
:
957
67
.
24.
Tann
AW
,
Boldogh
I
,
Meiss
G
,
Qian
W
,
Van Houten
B
,
Mitra
S
, et al
Apoptosis induced by persistent single-strand breaks in mitochondrial genome: Critical role of EXOG (5′-EXO/endonuclease) in their repair
.
J Biol Chem
2011
;
286
:
31975
83
.
25.
Turrens
JF
. 
Mitochondrial formation of reactive oxygen species
.
J Physiol
2003
;
552
(
Pt 2
):
335
44
.
26.
Cleaver
JE
,
Feeney
L
,
Revet
I
. 
Phosphorylated H2Ax is not an unambiguous marker for DNA double-strand breaks
.
Cell Cycle
2011
;
10
:
3223
4
.
27.
Shiloh
Y
,
Ziv
Y
. 
The ATM protein kinase: Regulating the cellular response to genotoxic stress, and more
.
Nat Rev Mol Cell Biol
2013
;
14
:
197
210
.
28.
Jones
RG
,
Plas
DR
,
Kubek
S
,
Buzzai
M
,
Mu
J
,
Xu
Y
, et al
AMP-activated protein kinase induces a p53-dependent metabolic checkpoint
.
Mol Cell
2005
;
18
:
283
93
.
29.
Guo
Z
,
Kozlov
S
,
Lavin
MF
,
Person
MD
,
Paull
TT
. 
ATM activation by oxidative stress
.
Science
2010
;
330
:
517
21
.
30.
Goodarzi
AA
,
Noon
AT
,
Deckbar
D
,
Ziv
Y
,
Shiloh
Y
,
Lobrich
M
, et al
ATM signaling facilitates repair of DNA double-strand breaks associated with heterochromatin
.
Mol Cell
2008
;
31
:
167
77
.
31.
Mei
H
,
Sun
S
,
Bai
Y
,
Chen
Y
,
Chai
R
,
Li
H
. 
Reduced mtDNA copy number increases the sensitivity of tumor cells to chemotherapeutic drugs
.
Cell Death Dis
2015
;
6
:
e1710
.
32.
Nakahira
K
,
Haspel
JA
,
Rathinam
VA
,
Lee
SJ
,
Dolinay
T
,
Lam
HC
, et al
Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome
.
Nat Immunol
2011
;
12
:
222
30
.
33.
Shimada
K
,
Crother
TR
,
Karlin
J
,
Dagvadorj
J
,
Chiba
N
,
Chen
S
, et al
Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis
.
Immunity
2012
;
36
:
401
14
.
34.
Zhou
R
,
Yazdi
AS
,
Menu
P
,
Tschopp
J
. 
A role for mitochondria in NLRP3 inflammasome activation
.
Nature
2011
;
469
:
221
5
.
35.
Echtay
KS
,
Roussel
D
,
St-Pierre
J
,
Jekabsons
MB
,
Cadenas
S
,
Stuart
JA
, et al
Superoxide activates mitochondrial uncoupling proteins
.
Nature
2002
;
415
:
96
9
.
36.
Zhong
Z
,
Umemura
A
,
Sanchez-Lopez
E
,
Liang
S
,
Shalapour
S
,
Wong
J
, et al
NF-kappaB restricts inflammasome activation via elimination of damaged mitochondria
.
Cell
2016
;
164
:
896
910
.
37.
James
EL
,
Michalek
RD
,
Pitiyage
GN
,
de Castro
AM
,
Vignola
KS
,
Jones
J
, et al
Senescent human fibroblasts show increased glycolysis and redox homeostasis with extracellular metabolomes that overlap with those of irreparable DNA damage, aging, and disease
.
J Proteome Res
2015
;
14
:
1854
71
.
38.
Lakshmipathy
U
,
Campbell
C
. 
Antisense-mediated decrease in DNA ligase III expression results in reduced mitochondrial DNA integrity
.
Nucleic Acids Res
2001
;
29
:
668
76
.
39.
Holt
IJ
,
Jacobs
HT
. 
Unique features of DNA replication in mitochondria: A functional and evolutionary perspective
.
Bioessays
2014
;
36
:
1024
31
.
40.
Sallmyr
A
,
Fan
J
,
Datta
K
,
Kim
KT
,
Grosu
D
,
Shapiro
P
, et al
Internal tandem duplication of FLT3 (FLT3/ITD) induces increased ROS production, DNA damage, and misrepair: Implications for poor prognosis in AML
.
Blood
2008
;
111
:
3173
82
.
41.
Hlavata
L
,
Aguilaniu
H
,
Pichova
A
,
Nystrom
T
. 
The oncogenic RAS2(val19) mutation locks respiration, independently of PKA, in a mode prone to generate ROS
.
EMBO J
2003
;
22
:
3337
45
.
42.
Vafa
O
,
Wade
M
,
Kern
S
,
Beeche
M
,
Pandita
TK
,
Hampton
GM
, et al
c-Myc can induce DNA damage, increase reactive oxygen species, and mitigate p53 function: A mechanism for oncogene-induced genetic instability
.
Mol Cell
2002
;
9
:
1031
44
.
43.
Sabharwal
SS
,
Schumacker
PT
. 
Mitochondrial ROS in cancer: Initiators, amplifiers or an Achilles' heel?
Nat Rev Cancer
2014
;
14
:
709
21
.
44.
Furda
AM
,
Marrangoni
AM
,
Lokshin
A
,
Van Houten
B
. 
Oxidants and not alkylating agents induce rapid mtDNA loss and mitochondrial dysfunction
.
DNA Repair
2012
;
11
:
684
92
.
45.
Yakes
FM
,
Van Houten
B
. 
Mitochondrial DNA damage is more extensive and persists longer than nuclear DNA damage in human cells following oxidative stress
.
Proc Natl Acad Sci U S A
1997
;
94
:
514
9
.
46.
Shokolenko
I
,
Venediktova
N
,
Bochkareva
A
,
Wilson
GL
,
Alexeyev
MF
. 
Oxidative stress induces degradation of mitochondrial DNA
.
Nucleic Acids Res
2009
;
37
:
2539
48
.
47.
Santos
JH
,
Hunakova
L
,
Chen
Y
,
Bortner
C
,
Van Houten
B
. 
Cell sorting experiments link persistent mitochondrial DNA damage with loss of mitochondrial membrane potential and apoptotic cell death
.
J Biol Chem
2003
;
278
:
1728
34
.
48.
Green
DR
,
Van Houten
B
. 
SnapShot: Mitochondrial quality control
.
Cell
2011
;
147
:
950
,
50 e1
.
49.
Viale
A
,
Pettazzoni
P
,
Lyssiotis
CA
,
Ying
H
,
Sanchez
N
,
Marchesini
M
, et al
Oncogene ablation-resistant pancreatic cancer cells depend on mitochondrial function
.
Nature
2014
;
514
:
628
32
.
50.
Zhang
X
,
Fryknas
M
,
Hernlund
E
,
Fayad
W
,
De Milito
A
,
Olofsson
MH
, et al
Induction of mitochondrial dysfunction as a strategy for targeting tumour cells in metabolically compromised microenvironments
.
Nat Commun
2014
;
5
:
3295
.

Supplementary data