Deferoxamine (DFO) represents a widely used iron chelator for the treatment of iron overload. Here we describe the use of mitochondrially targeted deferoxamine (mitoDFO) as a novel approach to preferentially target cancer cells. The agent showed marked cytostatic, cytotoxic, and migrastatic properties in vitro, and it significantly suppressed tumor growth and metastasis in vivo. The underlying molecular mechanisms included (i) impairment of iron-sulfur [Fe-S] cluster/heme biogenesis, leading to destabilization and loss of activity of [Fe-S] cluster/heme containing enzymes, (ii) inhibition of mitochondrial respiration leading to mitochondrial reactive oxygen species production, resulting in dysfunctional mitochondria with markedly reduced supercomplexes, and (iii) fragmentation of the mitochondrial network and induction of mitophagy. Mitochondrial targeting of deferoxamine represents a way to deprive cancer cells of biologically active iron, which is incompatible with their proliferation and invasion, without disrupting systemic iron metabolism. Our findings highlight the importance of mitochondrial iron metabolism for cancer cells and demonstrate repurposing deferoxamine into an effective anticancer drug via mitochondrial targeting.

Significance:

These findings show that targeting the iron chelator deferoxamine to mitochondria impairs mitochondrial respiration and biogenesis of [Fe-S] clusters/heme in cancer cells, which suppresses proliferation and migration and induces cell death.

Iron represents a vital element required for almost all forms of life (1). Because of its ability to transfer electrons by shuttling between ferrous and ferric forms, it is a crucial component participating in the electron transport chain within mitochondria, in DNA repair and replication (2), chromatin remodeling and epigenetic changes (3) and cellular metabolism (4).

In biological systems, iron can be incorporated into iron-sulfur [Fe-S] clusters and the heme molecule (2, 5). Both of these iron-containing structures serve as cofactors of many enzymes and are partially synthesized inside mitochondria, making it the central organelle in the cellular iron metabolism. At the same time, mitochondria are important for ATP production, metabolic reactions, calcium homeostasis, and programmed cell death (2, 4). Dysfunctional mitochondria are degraded by a specific process termed mitophagy, which is induced by iron deficiency (6, 7).

Iron is crucial for the rapidly proliferating cancer cells (8) and also for the cancer stem-like cells (9). Therefore, attempts to target iron metabolism to suppress tumor growth have been made (10). In certain cancer types, such as bladder cancer, gallium nitrate has been successfully applied, competitively replacing iron (11). Other approaches including antibodies against transferrin receptor, application of iron chelators, or a combination of both approaches have shown promising results in vitro (10, 12–14). Yet, the main remaining drawbacks of such strategies are the nonselective nature of the treatment and the associated perturbation of the systemic iron metabolism (14).

In recent years, a strategy for targeting small molecules into mitochondria via a triphenylphosphonium group (TPP+) proposed by Murphy and colleagues (15) has been applied for several compounds. Moreover, preferential accumulation of the drugs in cancer cells has been described, due to their higher inner mitochondrial membrane potential (16–19).

In this study, we have modified the classical iron chelator deferoxamine (DFO) by tagging it with the TPP+ group, thus repurposing an iron chelator used for treatment of iron overload into an anticancer agent. We demonstrate that such modification results in a preferential targeting of cancer cells, halting their proliferation and inducing cell death via interfering with their iron metabolism and destabilizing their mitochondria. This represents a novel approach towards targeting of cancer cells, which is of clinical relevance.

Cell culture

Cell lines were obtained from ATCC and used within 3 months from their thawing. Cells were tested for Mycoplasma (MycoAlert PLUS detection Kit; Lonza) and authenticated by short tandem repeat analysis (Generi Biotech).

mitoDFO and mitoDFO-FITC synthesis

mitoDFO—triphenyl (3,14,25-trihydroxy-2,10,13,21,24-pentaoxo-31-(10-(triphenyl phosphonio)decyl)-3,9,14,20,25,31-hexaazahentetracontan-41-yl) phosphonium chloride was prepared according to Scheme 1 described together with nuclear magnetic resonance results in Supplementary Data. mitoDFO-fluorescein isothiocyanate was prepared according to the protocol provided in Supplementary data. Both mitoDFO and mitoDFO-fluorescein isothiocyanate (mitoDFO-FITC) were dissolved in DMSO (10 mmol/L stock) and added to cells (max 0.4% DMSO). Under control conditions, the same volume of DMSO was added.

SDS-PAGE and Western blot analysis

SDS-PAGE and Western blot analysis were performed as reported previously (9). Cells were lysed in RIPA buffer and protein content assessed by bicinchoninic acid method (Thermo Fisher Scientific). The list of primary antibodies is available as Supplementary Table S3.

Aconitase activity

The activity of aconitase was assessed by the Aconitase Activity Assay Kit (Sigma-Aldrich) according to the manufacturer's instructions. Aconitase activity is expressed as percentage of control cells treated with the solvent (DMSO). Control (lysate) and mitoDFO (lysate) columns correspond to the activity observed in the cell lysate treated with either DMSO or mitoDFO at 5 μmol/L. In addition, in-gel aconitase activity was assessed according to a published protocol (20).

Heme content

Cellular heme content was evaluated as described previously (21), by boiling whole-cell lysate with 2 mol/L oxalic acid. Results are expressed as the difference between total and free protoporphyrin, relative to control.

Autoradiography

Detailed description of autoradiography is provided in Supplementary Materials and Methods.

Oxygen consumption and extracellular acidification measurement

The oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) were measured by the Seahorse XFe96 Analyzer (Agilent Technologies) according to the manufacturer's instructions (details in Supplementary Data).

Mitochondrial respiration in permeabilized cells was assessed using the high-resolution Oxygraph-2k respirometer (Oroboros Instruments), according to the standard procedure (22).

Isolation and solubilization of mitochondria

Mitochondria were isolated by differential centrifugation, and solubilized with digitonin, as described previously (23).

Native electrophoresis

Blue native electrophoresis (BNE) and high resolution clear native electrophoresis (hrCNE) were used to assess the level and activity of mitochondrial respiratory complexes, as described in ref. 23. All antibodies used are listed in the Supplementary Table S3.

Cellular ATP content

Total cellular ATP was assessed by the Cell Titer-Glo Luminescent Cell Viability Assay Kit (Promega) according to the manufacturer's instructions. Results are presented as percentage of control.

Reactive oxygen species levels and mitochondrial membrane potential

Mitochondrial superoxide levels were determined using the fluorescent probe MitoSOX Red (Thermo Fisher Scientific) and total cellular reactive oxygen species (ROS) by 2′,7′-dichlorodihydrofluorescein-diacetate (DCF-DA; Sigma-Aldrich). Mitochondrial membrane potential was assessed using tetramethylrhodamine methyl ester (TMRM; Sigma-Aldrich). Results are expressed as percentage of relative fluorescence units relative to the control.

Confocal microscopy

Confocal microscopy images were acquired using a 63× water immersion objective in a Leica SP8 confocal microscope (Leica Microsystems; details in Supplementary Data).

KEIMA protein detection

Acidification of mitochondria was detected using the fluorescent protein mitoKEIMA as described previously (24). Results are expressed as the ratio between fluorescence intensity at 440 and 586 nm, relative to control.

Crystal violet staining

The number of viable cells showing combined cytostatic and cytotoxic effect was assessed by crystal violet staining. IC50 values for each condition were calculated by plotting percentage of viable cells versus compound concentration. After transforming x values to logarithmic scale, values were fitted to dose–response inhibition curves with normalized y values in the GraphPad Prism 5.0 software, and IC50 values were interpolated and expressed as mean ± SEM.

Real-time cell monitoring

The xCELLigence real-time cell analysis (RTCA) SP station (ACEA Biosciences) was used to determine the effect of mitoDFO and deferoxamine on cell growth. Impedance was monitored for 48 hours in 15-minute intervals. Results were analyzed using the RTCA software and are shown as cell index over time.

Similarly, the effect of the compounds on cellular viability was analyzed by the JuLI FL Fluorescence Live Cell Analyzer (NanoEnTek) for 48 hours. Results were analyzed with the JuLI FL PC software and are shown as percentage of confluence over time.

Cell death assessment

Cell death was assessed using the annexin V/propidium iodide (AV/PI) assay, as described previously (25). Results are expressed as percentage of dead cells (sum of AV+/PI, AV/PI+ and AV+/PI+ cells).

Cell cycle

Cell cycle measurement was performed using Vybrant DyeCycle Violet Stain (Thermo Fisher Scientific), according to the manufacturer's instructions.

Three-dimensional invasion spheroid assay

MDA-MB-231 and 4T1 cells were grown as spheroids for 2 days using three-dimensional (3D) Petri Dish (Microtissues; #12-81 large spheroids) according to the manufacturer's instructions. The invasion assay was performed as described previously (26).

Animal studies

Balb/c mice were injected with 1 × 106 4T1 cells, and NSG mice with either 1 × 106 MDA-MB-231 cells or 3 × 106 Bx-PC3 cells. When tumors reached the volume of 30–50 mm3 (quantified by ultrasound imaging, USI), each group was divided into two subgroups and treated intraperitoneally with either mitoDFO (1, 8, or 12 mg/kg), deferoxamine (8 or 80 mg/kg), or vehicle (2.5% DMSO in corn oil, 100 μL per dose) twice per week. Tumor volume was monitored by the USI instrument Vevo770 (VisualSonics). Results are presented as relative tumor size. Animal ethics was approved by the Czech Academy of Sciences and animal experiments were performed according to the Czech Republic Council guidelines for the Care and Use of Animals in Research and Teaching.

Assessment of lung metastases

Lung metastases were evaluated as described previously (27), by selection of 4T1 cells with 60 μmol/L 6-thioguanine (details in Supplementary Materials and Methods).

Liver, spleen, and tumor iron levels

Tissue iron levels were determined by the method of Torrance and Bothwell (28) in mg per μg of wet tissue. Iron content was calculated on the basis of a standard curve of ferric ammonium citrate.

Serum iron levels

Serum iron levels were assessed by the Liquid Fe 200 Kit (Erba Lachema) according to the manufacturer's instructions. Samples for the analysis were acquired by cardiac puncture, blood was clotted for 30 minutes and spun, and the resulting serum was analyzed. Hemolytic samples were excluded from the analysis.

Hematologic parameters

Blood was collected into heparinized tubes and hematologic parameters measured at the Czech Centre for Phenogenomics, using the Mindray Bc-5300 Vet.

Ki67 IHC

Tumors were fixed in 4% paraformaldehyde and analyzed by the Czech Centre for Phenogenomics principally as shown before (29). Images were acquired using the AxioScan.Z1 automated slide scanner (Carl Zeiss) and analyzed by the ImageJ software.

Statistical analysis

All results are expressed as mean ± SEM of at least three independent experiments. The comparison between experimental groups and control was performed by one- or two-way ANOVA followed by Sidak post test, or by Student t test using the GraphPad Prism 5.0 software. Differences were considered significant at P < 0.05.

mitoDFO destabilizes iron metabolism in vitro

Deferoxamine, a cell-impermeable iron chelator, has been shown to inhibit proliferation of cancer cells both in vitro and in vivo (13, 14, 30). Aiming to direct it into mitochondria, we tagged it with two TPP+ moieties via 10-carbon linker chains, referring to it as mitochondrially targeted deferoxamine (mitoDFO; for structure see Fig. 1A).

Figure 1.

mitoDFO reduces mitochondrial biogenesis of [Fe-S] clusters and heme. A, Structure of mitoDFO. B, Western blot images of mitochondrial ETC [Fe-S] cluster/heme-containing subunits and lipoylated proteins in MCF7 and MDA-MB-231 human breast cancer cells exposed to mitoDFO or deferoxamine for 16 hours. C, Western blot images of mitochondrial (ISCU) and cytosolic (CIAO1, MMS19, NUBP1) [Fe-S] cluster biogenesis-related proteins in MCF7 and MDA-MB-231 cells exposed to mitoDFO or deferoxamine (DFO) for 16 hours. D, In-gel enzymatic activity and Western blot images of mitochondrial and cytosolic aconitase in MCF7 and MDA-MB-231 cells exposed to DFO or mitoDFO for 6 hours. E, Western blot images of iron metabolism-related proteins in MCF7 and MDA-MB-231 cells exposed to mitoDFO or deferoxamine for 16 hours. F,55Fe autoradiography of mitochondrial and cytosolic fractions from MCF7 cells preincubated with 55Fe for 72 hours and exposed to mitoDFO 5 μmol/L or deferoxamine 50 μmol/L for 24 hours in fresh media. G, Western blot images of heme-related proteins in MCF7 and MDA-MB-231 cells exposed to mitoDFO or deferoxamine for 16 hours. H, Heme content in MCF7 and MDA-MB-231 cells exposed to mitoDFO or deferoxamine for 16 hours. Values are normalized to protein content and represent the mean of at least three independent experiments. Numbers below each Western blot band represent the mean relative intensity compared with control obtained from at least three independent experiments. P values were calculated by one-way ANOVA followed by Sidak multiple comparisons test. *, P < 0.05 versus Control (B, C, E, G, and H).

Figure 1.

mitoDFO reduces mitochondrial biogenesis of [Fe-S] clusters and heme. A, Structure of mitoDFO. B, Western blot images of mitochondrial ETC [Fe-S] cluster/heme-containing subunits and lipoylated proteins in MCF7 and MDA-MB-231 human breast cancer cells exposed to mitoDFO or deferoxamine for 16 hours. C, Western blot images of mitochondrial (ISCU) and cytosolic (CIAO1, MMS19, NUBP1) [Fe-S] cluster biogenesis-related proteins in MCF7 and MDA-MB-231 cells exposed to mitoDFO or deferoxamine (DFO) for 16 hours. D, In-gel enzymatic activity and Western blot images of mitochondrial and cytosolic aconitase in MCF7 and MDA-MB-231 cells exposed to DFO or mitoDFO for 6 hours. E, Western blot images of iron metabolism-related proteins in MCF7 and MDA-MB-231 cells exposed to mitoDFO or deferoxamine for 16 hours. F,55Fe autoradiography of mitochondrial and cytosolic fractions from MCF7 cells preincubated with 55Fe for 72 hours and exposed to mitoDFO 5 μmol/L or deferoxamine 50 μmol/L for 24 hours in fresh media. G, Western blot images of heme-related proteins in MCF7 and MDA-MB-231 cells exposed to mitoDFO or deferoxamine for 16 hours. H, Heme content in MCF7 and MDA-MB-231 cells exposed to mitoDFO or deferoxamine for 16 hours. Values are normalized to protein content and represent the mean of at least three independent experiments. Numbers below each Western blot band represent the mean relative intensity compared with control obtained from at least three independent experiments. P values were calculated by one-way ANOVA followed by Sidak multiple comparisons test. *, P < 0.05 versus Control (B, C, E, G, and H).

Close modal

We first tested whether mitoDFO affects cellular iron metabolism. Our results show that mitoDFO induces a dysfunction in mitochondrial [Fe-S] cluster assembly, as the levels of [Fe-S] cluster-containing mitochondrial proteins NDUFA9, SDHB, UQCRFS1, and mtCO1 are markedly reduced upon mitoDFO treatment of MCF7 and MDA-MB-231 breast cancer cells (Fig. 1B). Furthermore, the levels of lipoylated proteins, which are dependent on the [Fe-S] cluster-containing lipoyl synthase (31), were diminished as a response to mitoDFO treatment (Fig. 1B). Interestingly, deferoxamine treatment (100 μmol/L) showed a much lower effect in both cell lines (Fig. 1B). We then studied the levels of enzymes responsible for [Fe-S] cluster synthesis. Our results show that while the level of the mitochondrially localized enzyme ISCU was greatly diminished by mitoDFO, the cytosolic enzymes were either not affected (CIAO1, MMS19) or even slightly increased (NUBP1 in MCF7 cells; Fig. 1C).

To identify the mechanism behind the decrease in the protein levels, we first analyzed the mRNA expression of iron-related genes. Our results show that mitoDFO and deferoxamine cause a small, yet significant reduction in the expression of genes related to [Fe-S] cluster biogenesis and utilization, as well as heme biosynthesis and degradation. On the other hand, the level of mRNA of genes related to iron uptake and storage was significantly upregulated (Supplementary Fig. S1A). It is of note that transcripts such as ACO1, ACO2, COX1, and TFRC clearly showed differential response to deferoxamine and mitoDFO (Supplementary Fig. S1A), documenting that the effect of the two chelators is not identical. Nevertheless, changes in gene expression were rather mild (2-fold), while changes in the corresponding protein levels were more profound especially for the mitochondrial proteins NDUFA9, SDHB, UQCRFS1, mtCO1, ISCU, and FECH, and these changes were not replicated with 100 μmol/L deferoxamine (Fig. 1B, C, E, and F). We thus propose that the main effect of mitoDFO occurs at the level of protein degradation, and we show that cotreatment with mitoDFO and the proteasome inhibitor MG-132 (5 μmol/L; 16 hours) prevented the decrease in protein levels of the proteasome-degraded ISCU (32), SDHB (33), FECH (34) and mtCO1 (35). On the other hand, the effect of mitoDFO on the level of NDUFA9 and UQCRFS1 was not affected by proteasome inhibition (Supplementary Fig. S1B), as both proteins are degraded by mitochondrial proteases (36, 37).

Another enzyme that depends on the correct assembly of [Fe-S] clusters is aconitase. In agreement with our previous results, mitoDFO significantly reduced the activity of both forms of aconitase (cytosolic and mitochondrial; Fig. 1D; Supplementary Fig. S1C). Interestingly, while deferoxamine had a similar effect on the activity of cytosolic ACO1, it had a much lower effect on the activity of mitochondrial ACO2 (Fig. 1D). Our results also show that mitoDFO not only affected aconitase activity when added to live cells, but decreased it even when added to whole-cell lysates, suggesting a direct effect of the compound on the aconitase proteins (Supplementary Fig. S1D). Aconitase 1 that lacks its [Fe-S] cluster serves as iron regulatory protein 1 (IRP1) and has affinity for the iron-responsive elements (IRE) on the mRNA of several iron regulators (38). Our results suggest that mitoDFO promotes increased binding of IRP1 to IREs and activates the IRP/IRE system, as the level of transferrin receptor 1 is increased, while the level of ferritin is decreased although the extent of the activation is lower compared with deferoxamine (Fig. 1E), being in line with only a slight change in the protein level of IRE-containing ferroportin and ACO2 (Fig. 1D and E). Similarly, the level of the cytosolic iron sensor FBXL5 was not changed or only slightly increased in response to mitoDFO, suggesting that the free labile iron pool in the cytosol is not diminished (Fig. 1E).

To further define the effect of mitoDFO on iron-containing proteins, we incubated cells loaded with 55Fe with mitoDFO and deferoxamine, and visualized mitochondrial and cytosolic iron-containing proteins by autoradiography. Our data show that 24-hour incubation with 5 μmol/L mitoDFO led to a dramatic decrease in bands corresponding to respiratory supercomplexes, a general decrease of the 55Fe signal in mitochondrial fraction, and a decrease in cytosolic ferritin (Fig. 1F). Deferoxamine treatment (50 μmol/L) did not affect mitochondrial proteins but led to a marked decrease in cytosolic ferritin (Fig. 1F). Long-term incubation with 1 μmol/L mitoDFO (60 hours) led to a decrease (MCF7) or absence (MDA-MB-231) of bands corresponding to iron-containing native proteins (Fig. 1F; Supplementary Fig. S1E) with the exception of ferritin, which was considerably increased (Supplementary Fig. S1E).

The second iron-containing cofactor is heme. Our results show that mitoDFO significantly reduced the level of mitochondrial FECH responsible for the last step of heme synthesis, while not affecting the levels of HMOX2 responsible for heme degradation. In line with this, phosphorylation of EIF2α induced by the heme-regulated inhibitor kinase (HRI) upon heme deficiency (39) was upregulated by mitoDFO (Fig. 1G). Although similar changes were observed with deferoxamine, they required much higher concentration (100 μmol/L; Fig. 1G). Furthermore, the total heme level (Fig. 1H) was lower upon treatment with mitoDFO, which is likely linked to lower levels of [Fe-S] cluster-containing ferrochelatase, resulting in lower heme biosynthesis, and diminished iron bioavailability. Once again, deferoxamine required a 10-fold higher concentration to reach a similar effect.

mitoDFO impairs mitochondrial OCR

One of the main processes within mitochondria involves the transfer of electrons along the electron transport chain (ETC). Because the function of the ETC highly relies on [Fe-S] clusters and heme, we assessed the effect of mitoDFO on ETC-dependent oxygen consumption. Our results show that mitoDFO (10 μmol/L, 1 hour) completely suppressed mitochondrial respiration (Fig. 2A; Supplementary Fig. S2A) and enhanced basal ECAR (a surrogate marker of the glycolytic state; Fig. 2B; Supplementary Fig. S2B) in all cell lines studied.

Figure 2.

mitoDFO inhibits mitochondrial respiration and enhances glycolysis. A and B, Profile of OCR (A) and ECAR (B) in MCF7 and MDA-MB-231 cells exposed to 10 μmol/L mitoDFO for 1 hour, assessed using the Seahorse XF analyzer. OCR was evaluated before and after addition of oligomycin (Omy, CV inhibitor), CCCP (an uncoupler of OXPHOS), and rotenone + antimycin A (Rot + AA, CI and CIII inhibitor, respectively). ECAR was evaluated before and after addition of glucose (Glc), oligomycin, and 2-deoxyglucose (2-DG, hexokinase-II inhibitor). All values are mean ± SEM of three independent experiments with at least five replicates each. C, ATP levels in MCF7 and MDA-MB-231 cells exposed to 10 μmol/L mitoDFO for 4 hours in the absence or presence of 2-DG or Omy. Each value represents mean ± SEM of at least three independent experiments with at least six replicates each. D, Western blot images after BNE of mitochondrial lysates of MCF7 and MDA-MB-231 cells exposed to 5 μmol/L mitoDFO for 16 hours. HSP60 served as a loading control. P values were calculated by one-way ANOVA followed by Sidak multiple comparisons test. *, P < 0.05 versus Control; #, P < 0.05 vs. untreated (C).

Figure 2.

mitoDFO inhibits mitochondrial respiration and enhances glycolysis. A and B, Profile of OCR (A) and ECAR (B) in MCF7 and MDA-MB-231 cells exposed to 10 μmol/L mitoDFO for 1 hour, assessed using the Seahorse XF analyzer. OCR was evaluated before and after addition of oligomycin (Omy, CV inhibitor), CCCP (an uncoupler of OXPHOS), and rotenone + antimycin A (Rot + AA, CI and CIII inhibitor, respectively). ECAR was evaluated before and after addition of glucose (Glc), oligomycin, and 2-deoxyglucose (2-DG, hexokinase-II inhibitor). All values are mean ± SEM of three independent experiments with at least five replicates each. C, ATP levels in MCF7 and MDA-MB-231 cells exposed to 10 μmol/L mitoDFO for 4 hours in the absence or presence of 2-DG or Omy. Each value represents mean ± SEM of at least three independent experiments with at least six replicates each. D, Western blot images after BNE of mitochondrial lysates of MCF7 and MDA-MB-231 cells exposed to 5 μmol/L mitoDFO for 16 hours. HSP60 served as a loading control. P values were calculated by one-way ANOVA followed by Sidak multiple comparisons test. *, P < 0.05 versus Control; #, P < 0.05 vs. untreated (C).

Close modal

To confirm this finding, we assessed the levels of ATP after the incubation of cells with mitoDFO in the absence or presence of the glycolysis inhibitor 2-deoxyglucose (2-DG). Surprisingly, mitoDFO (10 μmol/L, 1 hour) alone did not have any effect on the ATP content (Fig. 2C). However, the combination of mitoDFO and 2-DG dropped ATP levels significantly more than 2-DG alone. Such effect was not observed when cells were coincubated with mitoDFO and the complex V inhibitor oligomycin (2 μmol/L; Fig. 2C). Of note, mitoDFO treatment also induced an increase in protein level of some key glycolytic enzymes (HKII, LDHA) while simultaneously decreasing the citric acid cycle enzyme PDH-E1 (Supplementary Fig. S2C).

Efficient ETC-linked respiration mainly relies on the assembly of respiratory complexes (RC) and supercomplexes (SC). The most known SC, referred to as respirasome, is composed of complex I (CI), CIII, and CIV (40). Given the profound effect of mitoDFO on the level of individual [Fe-S] cluster-containing subunits of the ETC and on mitochondrial respiration, we analyzed the level and organization of RCs and SCs, using blue native electrophoresis. A strong decrease in the level of the respirasome was observed in all cell lines after their exposure to 5 μmol/L mitoDFO for 16 hours (Fig. 2D; Supplementary Fig. S2D) while deferoxamine showed no effect (Supplementary Fig. S2E). This change was not accompanied by a decrease in lower forms of SCs, such as the SC composed of only CIII and CIV. On the other hand, it appears that an intermediate SC of CIII, migrating above CIII2/CIV, is enhanced (Fig. 2D). Regarding individual RCs, we observed a minor decrease in the levels of CIII and CIV and no change in CV. CII, however, was significantly decreased in response to mitoDFO treatment. We also performed clear native electrophoresis to assess in-gel activity of complex I in mitoDFO-treated cells. Our results show that the activity of CI was lower in the respirasome as well as in other SCs comprising CI and in CI itself (Supplementary Fig. S3A).

Therefore, we studied the effect of short-term mitoDFO incubation on respiration via individual complexes using the high-resolution respirometer Oxygraph-2k. Supplementary Fig. S3B shows that 1-hour incubation of MCF7 cells with 10 μmol/L mitoDFO lowered CI-dependent respiration, while it did not significantly change CII-dependent respiration.

mitoDFO accumulates in mitochondria and induces mitochondrial dysfunction

Given the differential effect of mitoDFO on mitochondria compared with deferoxamine, we next sought to demonstrate that our compound accumulates within mitochondria. To do so, we attached a FITC moiety to mitoDFO, and our results show that in MCF7 and MDA-MB-231 cells mitoDFO-FITC signal highly colocalizes with that of MitoTracker Deep Red (Fig. 3A).

Figure 3.

mitoDFO accumulates in mitochondria and increases ROS. A, Representative confocal images of MCF7 and MDA-MB-231 cells incubated with mitoDFO-FITC (10 μmol/L) and MitoTracker Deep Red (5 nmol/L) for 1 hour, with added Hoechst 33342 at 2 μg/mL before imaging. Enlarged panel corresponds to ×10 magnification of the “merge” panel. Manders coefficients for green-to-red colocalization were 0.86 ± 0.05 for MCF7 cells and 0.67 ± 0.08 for MDA-MB-231 cells. B–D, Quantification of TMRM fluorescence (mitochondrial membrane potential; B), mitoSOX fluorescence (mitochondrial superoxide; C), and DCF-DA fluorescence (cellular ROS; D) after exposure of MCF7, MDA-MB-231, Bx-PC3, and OVCAR-3 cells to 10 μmol/L mitoDFO. E, Quantification of cell death after exposure of MCF7, MDA-MB-231, Bx-PC3, and OVCAR-3 cells to mitoDFO in the absence or presence of NAC for 48 hours. All values represent mean ± SEM of three independent experiments with two replicates each. P values were calculated by one-way ANOVA followed by Sidak multiple comparisons test.*, P < 0.05 versus control (BD); #, P < 0.05 relative to the same concentration of mitoDFO without NAC (E).

Figure 3.

mitoDFO accumulates in mitochondria and increases ROS. A, Representative confocal images of MCF7 and MDA-MB-231 cells incubated with mitoDFO-FITC (10 μmol/L) and MitoTracker Deep Red (5 nmol/L) for 1 hour, with added Hoechst 33342 at 2 μg/mL before imaging. Enlarged panel corresponds to ×10 magnification of the “merge” panel. Manders coefficients for green-to-red colocalization were 0.86 ± 0.05 for MCF7 cells and 0.67 ± 0.08 for MDA-MB-231 cells. B–D, Quantification of TMRM fluorescence (mitochondrial membrane potential; B), mitoSOX fluorescence (mitochondrial superoxide; C), and DCF-DA fluorescence (cellular ROS; D) after exposure of MCF7, MDA-MB-231, Bx-PC3, and OVCAR-3 cells to 10 μmol/L mitoDFO. E, Quantification of cell death after exposure of MCF7, MDA-MB-231, Bx-PC3, and OVCAR-3 cells to mitoDFO in the absence or presence of NAC for 48 hours. All values represent mean ± SEM of three independent experiments with two replicates each. P values were calculated by one-way ANOVA followed by Sidak multiple comparisons test.*, P < 0.05 versus control (BD); #, P < 0.05 relative to the same concentration of mitoDFO without NAC (E).

Close modal

Targeting TPP+ compounds to the inner mitochondrial membrane (IMM) is driven by their positive charge and the negative potential at the matrix face of the IMM (15). However, the presence of such compounds might lead to the dissipation of mitochondrial membrane potential (MMP), mitochondrial depolarization, and dysfunction (16, 17, 19). Our data show that addition of mitoDFO to cells led to a rapid and significant decrease of tetramethylrhodamine (TMRM) fluorescence, indicating a loss of MMP (Fig. 3B).

Mitochondria are known to be the main intracellular source of ROS (41), and mitochondrial depolarization and dysfunction highly increase their production. Therefore, we assessed the ability of mitoDFO to generate oxidative stress and found that it induced a rapid and substantial increase in mitochondrial superoxide and cellular ROS levels (Fig. 3C and D). Furthermore, coincubation of cells with mitoDFO and the antioxidant N-acetyl cysteine (NAC, 10 mmol/L) significantly suppressed cell death induced by mitoDFO (Fig. 3E) and the levels of intrinsic antioxidant defense enzymes CAT and SOD2 were slightly induced by a 16 hours treatment with mitoDFO (Supplementary Fig. S3C).

mitoDFO induces mitochondrial fragmentation and mitophagy

Mitochondria are highly dynamic organelles able to form a vast network. Such network is controlled by fusion, rendering highly efficient, nuclei-associated elongated mitochondria (42), and fission, resulting in small, fragmented, radially located mitochondria. The increased fission is a hallmark of mitochondrial damage and increased oxidative stress (41). Therefore, we assessed the status of the mitochondrial network by confocal microscopy and analyzed the intracellular distribution of mitochondria. Figure 4A shows that mitoDFO treatment (5 μmol/L, 24 hours) induces a slight, yet significant increase of radially localized mitochondria at the expense of their perinuclear localization in MCF7 cells. Furthermore, significant decrease in average mitochondrial volume and increase in the number of individual mitochondria in each cell were observed upon treatment with mitoDFO (Fig. 4B), consistent with more prominent mitochondrial fission. Of note, such increase in the number of individual mitochondria was not caused by increase in cellular size, as cell area was not affected by mitoDFO (Fig. 4B).

Figure 4.

mitoDFO induces mitochondrial fragmentation and mitophagy. A and B, Representative confocal images and quantification of mitochondrial distribution (A) and mitochondrial number, average size, and cell area (B) of MCF7 cells exposed to 5 μmol/L mitoDFO for 24 hours. Mitochondria were visualized by stably transfected mitochondrially-targeted GFP, and nuclei were stained with Hoechst 33342 at 2 μg/mL. C and D, Confocal microscopy images (C) and their quantification in MCF7 and MDA-MB-231 (D) cells exposed to 5 μmol/L mitoDFO for 24 hours and stained using a Mitophagy kit for visualization of acidified mitochondria. Microscopy analyses were performed in three independent samples with at least five fields of view each. E, Quantification of mitoKEIMA fluorescence assessed by flow cytometry in MCF7, MDA-MB-231, and 4T1 cells exposed to mitoDFO or deferoxamine for 24 hours. Cells were stably transfected with mitoKEIMA. All data are represented as mean ± SEM of three independent experiments with at least two replicates each. F, Western blot images of mitophagy markers in MCF7 and MDA-MB-231 cells exposed to mitoDFO for 16 hours. Numbers below each Western blot band represent the relative intensity compared with the control and were obtained from at least three independent experiments. P values were calculated by unpaired Student t test (A, B, and D) or by one-way ANOVA followed by Sidak multiple comparisons test (E and F). *, P < 0.05 versus control (AF).

Figure 4.

mitoDFO induces mitochondrial fragmentation and mitophagy. A and B, Representative confocal images and quantification of mitochondrial distribution (A) and mitochondrial number, average size, and cell area (B) of MCF7 cells exposed to 5 μmol/L mitoDFO for 24 hours. Mitochondria were visualized by stably transfected mitochondrially-targeted GFP, and nuclei were stained with Hoechst 33342 at 2 μg/mL. C and D, Confocal microscopy images (C) and their quantification in MCF7 and MDA-MB-231 (D) cells exposed to 5 μmol/L mitoDFO for 24 hours and stained using a Mitophagy kit for visualization of acidified mitochondria. Microscopy analyses were performed in three independent samples with at least five fields of view each. E, Quantification of mitoKEIMA fluorescence assessed by flow cytometry in MCF7, MDA-MB-231, and 4T1 cells exposed to mitoDFO or deferoxamine for 24 hours. Cells were stably transfected with mitoKEIMA. All data are represented as mean ± SEM of three independent experiments with at least two replicates each. F, Western blot images of mitophagy markers in MCF7 and MDA-MB-231 cells exposed to mitoDFO for 16 hours. Numbers below each Western blot band represent the relative intensity compared with the control and were obtained from at least three independent experiments. P values were calculated by unpaired Student t test (A, B, and D) or by one-way ANOVA followed by Sidak multiple comparisons test (E and F). *, P < 0.05 versus control (AF).

Close modal

When fragmented mitochondria are not able to recover from an insult, the process of mitophagy is activated and damaged mitochondria are engulfed by lysosomes, where they are degraded (6, 43, 44). By using a pH-sensitive mitochondrial probe, we were able to study the acidification that occurs in mitochondria upon their engulfment. Our results show that incubation of cells with mitoDFO (5 μmol/L, 24 hours) led to increased mitochondrial acidification (Fig. 4C and D). Another indicator of mitochondrial acidification is the reporter protein mitoKEIMA, with excitation fluorescence dependent on the pH value (24). Consistent with the mitophagy-inducing effect of mitoDFO, mitoKEIMA-expressing cancer cells showed a lower ratio of neutral-to-acid-related fluorescence following exposure to the agent (Fig. 4E). Furthermore, we observed increase in PINK1 and BNIP3 levels, decrease in p62 and processing of LC3B upon addition of mitoDFO to cancer cells (Fig. 4F), pointing to mitophagy activation (44).

Mitochondrial targeting of deferoxamine significantly enhances its antiproliferative and cytotoxic effects

Mitochondrial dysfunction, increased oxidative stress and enhanced mitophagy can halt the cell cycle and eventually cause cell death. Therefore, we evaluated the effect of mitoDFO on cellular proliferation and viability. Our results show that mitoDFO is at least 40-fold more potent than the parental deferoxamine in decreasing the number of viable cancer cells of breast, ovarian, and pancreatic origin (Fig. 5A; Supplementary Table S1). To further understand the inhibitory effect of mitoDFO, we performed real-time monitoring assays with low concentrations of mitoDFO (1–5 μmol/L). The results show that at concentrations ≤ 2 μmol/L, the effect of mitoDFO is primarily cytostatic, with cell death appearing at concentrations of 5 μmol/L (Fig. 5B; Supplementary Fig. S3D and S3E). Once again, real-time monitoring confirmed that mitoDFO is more potent than deferoxamine in inhibiting cellular proliferation and inducing cell death, as deferoxamine increased proliferation of cells at low micromolar concentrations and induced cell death only at a concentration of 100 μmol/L (Fig. 5B, top right).

Figure 5.

Mitochondrial targeting of deferoxamine enhances its cytostatic and cytotoxic activity. A, Quantification of the number of viable cells by crystal violet after exposure of MCF7 (malignant) and BJ (nonmalignant) cells to increasing concentrations of mitoDFO and for 48 hours. B, Proliferation curves for MCF7, MDA-MB-231, and BJ cells exposed to mitoDFO or deferoxamine. Growth was monitored by electrical impedance using the xCELLigence real-time cell analysis (RTCA) SP station. C, Quantification of cell-cycle distribution of MCF7, MDA-MB-231, and BJ cells exposed to mitoDFO or deferoxamine for 24 hours. D and E, Quantification of cell death after exposure of human breast (MCF7, MDA-MB-231), pancreatic (Bx-PC3), and ovarian (OVCAR-3) cancer cells, and normal fibroblasts (BJ, MRC-5, and HFP1) to mitoDFO or deferoxamine for 48 hours. Cell death was assessed by the AV/PI method. F, Quantification of cell death of MCF7, MDA-MB-231, Bx-PC3, and OVCAR-3 cells exposed to mitoDFO alone or after preincubation with Fe3+ for 48 hours. All data represent mean ± SEM of three independent experiments with at least two replicates each. P values were calculated by two-way (C) or one-way ANOVA (D–F) followed by Sidak multiple comparisons test. $, P < 0.05 relative to G1 phase in the Control; @, P < 0.05 relative to S-phase in the Control (C); *, P < 0.05 relative to the Control (D and E); #, P < 0.05 relative to the same concentration of mitoDFO without Fe3+ (F).

Figure 5.

Mitochondrial targeting of deferoxamine enhances its cytostatic and cytotoxic activity. A, Quantification of the number of viable cells by crystal violet after exposure of MCF7 (malignant) and BJ (nonmalignant) cells to increasing concentrations of mitoDFO and for 48 hours. B, Proliferation curves for MCF7, MDA-MB-231, and BJ cells exposed to mitoDFO or deferoxamine. Growth was monitored by electrical impedance using the xCELLigence real-time cell analysis (RTCA) SP station. C, Quantification of cell-cycle distribution of MCF7, MDA-MB-231, and BJ cells exposed to mitoDFO or deferoxamine for 24 hours. D and E, Quantification of cell death after exposure of human breast (MCF7, MDA-MB-231), pancreatic (Bx-PC3), and ovarian (OVCAR-3) cancer cells, and normal fibroblasts (BJ, MRC-5, and HFP1) to mitoDFO or deferoxamine for 48 hours. Cell death was assessed by the AV/PI method. F, Quantification of cell death of MCF7, MDA-MB-231, Bx-PC3, and OVCAR-3 cells exposed to mitoDFO alone or after preincubation with Fe3+ for 48 hours. All data represent mean ± SEM of three independent experiments with at least two replicates each. P values were calculated by two-way (C) or one-way ANOVA (D–F) followed by Sidak multiple comparisons test. $, P < 0.05 relative to G1 phase in the Control; @, P < 0.05 relative to S-phase in the Control (C); *, P < 0.05 relative to the Control (D and E); #, P < 0.05 relative to the same concentration of mitoDFO without Fe3+ (F).

Close modal

Furthermore, there is a difference in the cytostatic effect of mitoDFO between most malignant and normal cells, as cancer cells in general respond by considerably lower proliferation while nonmalignant BJ fibroblasts continue to grow under the same conditions (Fig. 5B; Supplementary Fig. S3D). In addition, nonmalignant cell lines BJ, MRC5 and HFP1 show higher IC50 values for mitoDFO compared with malignant cell lines tested, with several exceptions (Supplementary Table S1), and we observed accumulation of cells in the G1-phase in MCF7 and MDA-MB-231 cells, but not in normal BJ fibroblasts, upon exposure to mitoDFO for 24 hours (Fig. 5C). However, such effect was not observed in OVCAR-3 or Bx-PC3 cells incubated with mitoDFO (Supplementary Fig. S3F), probably because at 24 hours mitoDFO showed very little cytostatic effect in OVCAR-3 cells, and already a strong cytotoxic effect in Bx-PC3 cells (Supplementary Fig. S3E).

Importantly, all tested cancer cell lines (even those with lower cytostatic effect of mitoDFO) showed enhanced cytotoxic effect of the compound observed by annexin V/PI staining at concentrations ranging from 5 to 40 μmol/L compared with their nonmalignant counterparts that exhibited pronounced cell death only at 40 μmol/L (Fig. 5D and E). Interestingly, deferoxamine did not show any effect on cell-cycle distribution when added at 5 μmol/L, and was only effective in stopping proliferation or inducing cytotoxicity at concentrations of 100–200 μmol/L, where it also affected nonmalignant BJ fibroblasts (Fig. 5B, C, and E; Supplementary Fig. S3D).

Finally, to determine whether the iron-chelating ability of mitoDFO was important for the cytotoxic effect, we exposed cells to mitoDFO preincubated with equimolar concentrations of iron citrate. Our data show that the complex of mitoDFO with iron is much less cytotoxic to malignant cells than the agent alone, confirming the importance of the iron-chelating properties of mitoDFO for cell death induction (Fig. 5F).

mitoDFO inhibits tumor growth

Our in vitro data provided a solid evidence that mitoDFO is effective in suppressing cellular proliferation and inducing death of cancer cells. Therefore, we next tested its efficacy in vivo using both syngeneic and xenograft mice models. In the syngeneic model, we found that mitoDFO (8 mg/kg, i.p.) considerably suppressed the growth of tumors, in some cases causing even a decrease in tumor size (Fig. 6A). Furthermore, Ki67 staining was markedly reduced in mitoDFO-treated tumors (Supplementary Figs. S4A and S4B). In contrast, deferoxamine failed to affect tumor progression, even when administered at a 10-fold higher dose (8 and 80 mg/kg, i.p; Fig. 6A).

Figure 6.

mitoDFO inhibits tumor growth. A–C, Tumor growth curves of mice injected subcutaneously with 4T1 (A), MDA-MB-231 (B), and Bx-PC3 (C) cells and treated with mitoDFO or deferoxamine (2 doses/week; n = 5 for 4T1; n = 7 for MDA-MB-231 and Bx-PC3). Treatment was started once the tumors reached approximately 30–50 mm3 and continued until the first tumor reached 1,000 mm3. D, Western blot images of mitophagy and iron metabolism markers, and [Fe-S] cluster-containing enzymes in 4T1 tumors treated with 8 mg/kg of mitoDFO twice per week for 18 days. E, Quantification of iron content in 4T1, MDA-MB-231, and Bx-PC3 tumors treated with mitoDFO (8 mg/kg for 4T1 and MDA-MB-231; 12 mg/kg for Bx-PC3) twice per week. F–H, Quantification of iron content in serum (F), spleen (G), and liver (H) of Balb/c and NSG tumor-bearing mice treated with mitoDFO (8 mg/kg) twice per week. I and J, Quantification of total red blood cell count (I) and hemoglobin content (J) of Balb/c tumor-bearing mice treated with mitoDFO (8 mg/kg) twice per week. All data are presented as mean ± SEM of at least five animals or as representative image from four animals' tumor lysates (D). P values were calculated by unpaired Student t test. *, P < 0.05 versus Control (AC).

Figure 6.

mitoDFO inhibits tumor growth. A–C, Tumor growth curves of mice injected subcutaneously with 4T1 (A), MDA-MB-231 (B), and Bx-PC3 (C) cells and treated with mitoDFO or deferoxamine (2 doses/week; n = 5 for 4T1; n = 7 for MDA-MB-231 and Bx-PC3). Treatment was started once the tumors reached approximately 30–50 mm3 and continued until the first tumor reached 1,000 mm3. D, Western blot images of mitophagy and iron metabolism markers, and [Fe-S] cluster-containing enzymes in 4T1 tumors treated with 8 mg/kg of mitoDFO twice per week for 18 days. E, Quantification of iron content in 4T1, MDA-MB-231, and Bx-PC3 tumors treated with mitoDFO (8 mg/kg for 4T1 and MDA-MB-231; 12 mg/kg for Bx-PC3) twice per week. F–H, Quantification of iron content in serum (F), spleen (G), and liver (H) of Balb/c and NSG tumor-bearing mice treated with mitoDFO (8 mg/kg) twice per week. I and J, Quantification of total red blood cell count (I) and hemoglobin content (J) of Balb/c tumor-bearing mice treated with mitoDFO (8 mg/kg) twice per week. All data are presented as mean ± SEM of at least five animals or as representative image from four animals' tumor lysates (D). P values were calculated by unpaired Student t test. *, P < 0.05 versus Control (AC).

Close modal

A similar growth-slowing effect of mitoDFO was observed in the xenograft models of breast and pancreatic cancer. In the first case, while a dose of 1 mg/kg had no effect, a dose of 8 mg/kg mitoDFO (i.p.) significantly decreased the growth rate of MDA-MB-231 tumors, although with less potency than in the syngeneic model (Fig. 6B). Similarly, the Bx-PC3 xenografts showed that mitoDFO (12 mg/kg i.p.) significantly inhibited the growth of tumors (Fig. 6C).

mitoDFO induces mitophagy, reduces [Fe-S] cluster biogenesis in vivo, and does not affect systemic iron homeostasis

To assess whether the induction of mitophagy observed in cultured cells also occurs in vivo, we analyzed the expression of several mitophagy markers in 4T1-derived tumors. Our results show that mitoDFO treatment increased expression of PINK and BNIP3 while reducing the levels of p62 (Fig. 6D). Similarly, we observed an increase in transferrin receptor 1 and a decrease in FECH and SDHB, consistent with in vitro results. These changes were not detected in the liver tissue of the same animals (Supplementary Fig. S4C). Furthermore, mice weight was not affected by mitoDFO treatment (Supplementary Figs. S4D and S4E).

To define whether mitoDFO preferentially targets cancer cells in vivo and does not significantly affect the systemic iron metabolism, we assessed the level of iron in the tumor, serum, spleen and liver in treated and control tumor-bearing Balb/c and NSG mice. None of these parameters was significantly different between the control and treated animals (Fig. 6E–H). Similarly, the number of red blood cells, their hemoglobin content, hematocrit and mean corpuscular volume in Balb/c mice did not differ between the two groups (Fig. 6I and J; Supplementary Fig. S4F and S4G). In addition, white blood cells (WBC) were assessed and, although mitoDFO-treated animals showed a general increase in WBC number (Supplementary Fig. S4H), the percentage of each individual type of cell was similar in both groups (Supplementary Fig. S4I).

mitoDFO diminishes invasion ability in vitro and metastasis in vivo

We have shown that mitoDFO effectively inhibits growth of cancer cells, highlighting its potential for cancer treatment. However, the most life-threatening aspect of cancer is development of metastases, depending on the ability of cancer cells to invade surrounding tissues (45). Therefore, we tested the ability of mitoDFO to affect the invasive behavior of cancer cells using a 3D collagen invasion assay (26). Our results show that mitoDFO is highly efficient in inhibiting cell invasion at noncytotoxic concentrations (Fig. 7A and B). Again, the effect of mitoDFO was much more potent than that of deferoxamine.

Figure 7.

mitoDFO impairs cellular invasion and metastasis. A and B, Representative images and quantification of migration index of 4T1 (A) and MDA-MB-231 (B) cells exposed to mitoDFO or deferoxamine (DFO) for 48 hours. Invasion index was calculated as the ratio of the area after 48 hours of treatment and the initial area, relative to control. Data are represented as mean ± SEM of four independent experiments with at least six replicates each. C, Representative pictures and quantification of primary culture colonies from whole lung homogenates of Balb/c mice injected with 4T1 cells and treated with mitoDFO (8 mg/kg), deferoxamine (8 mg/kg; DFO Low) or DFO (80 mg/kg; DFO High). Treatment started when mice showed tumors of at least 30–50 mm3 and lasted 18 days. Whole lungs from each mice were minced and digested with a collagenase IV/elastase cocktail for 1 hour. Resulting homogenates were cultivated for 2 weeks in the presence of 6-thioguanine for selection of 4T1 cells. Colonies were stained with crystal violet and counted with the ImageJ software. P values were calculated by one-way ANOVA followed by Sidak multiple comparisons test. *, P < 0.05 relative to Control (A–C).

Figure 7.

mitoDFO impairs cellular invasion and metastasis. A and B, Representative images and quantification of migration index of 4T1 (A) and MDA-MB-231 (B) cells exposed to mitoDFO or deferoxamine (DFO) for 48 hours. Invasion index was calculated as the ratio of the area after 48 hours of treatment and the initial area, relative to control. Data are represented as mean ± SEM of four independent experiments with at least six replicates each. C, Representative pictures and quantification of primary culture colonies from whole lung homogenates of Balb/c mice injected with 4T1 cells and treated with mitoDFO (8 mg/kg), deferoxamine (8 mg/kg; DFO Low) or DFO (80 mg/kg; DFO High). Treatment started when mice showed tumors of at least 30–50 mm3 and lasted 18 days. Whole lungs from each mice were minced and digested with a collagenase IV/elastase cocktail for 1 hour. Resulting homogenates were cultivated for 2 weeks in the presence of 6-thioguanine for selection of 4T1 cells. Colonies were stained with crystal violet and counted with the ImageJ software. P values were calculated by one-way ANOVA followed by Sidak multiple comparisons test. *, P < 0.05 relative to Control (A–C).

Close modal

We also observed a strong anti-metastatic effect of mitoDFO in vivo in the syngeneic model of 4T1 lung metastatic breast cancer cells. We assessed the total number of lung-metastasized 4T1 cells in control and mitoDFO-treated tumor-bearing animals (8 mg/kg i.p.), and observed an almost complete inhibition of lung metastases in mitoDFO-treated mice (Fig. 7C). On the other hand, deferoxamine treatment (8 and 80 mg/kg) had no effect on the number of lung metastases (Fig. 7C).

Iron, an essential micronutrient, has a role in many cellular processes, such as DNA replication, cellular metabolism, and mitochondrial respiration (1, 2). Cancer cells show higher demand for iron due to their proliferative nature and altered metabolic needs, (8–10, 46). Several reports have shown that higher iron uptake and decreased iron export are associated with increased tumorigenicity and poor clinical outcome (9, 46). Therefore, iron chelators have been tested as possible anticancer agents (8, 10, 12–14, 30, 47–49), and shown to inhibit proliferation of cancer cells in vitro (13) and delay the growth of tumors (10, 12, 49). However, low intracellular availability of the agents and concomitant side effects linked to considerable alterations in systemic iron metabolism have been observed (14).

Within cells, iron is primarily present either in heme or [Fe-S] clusters (5). Because both are synthesized by mitochondria, these organelles are crucial for cellular iron metabolism (2, 4). We have, therefore, modified the structure of the iron chelator deferoxamine by tagging it with two triphenylphosphonium groups (TPP+), to target it to mitochondria (15). Our data show that mitoDFO accumulates in mitochondria, as 1-hour incubation with fluorescently labeled mitoDFO-FITC was sufficient to observe almost exclusive mitochondrial localization of FITC staining.

When designing mitoDFO, we tested deferoxamine tagged with one or two TPP+ groups and found that mitoDFO with two TPP+ moieties is more potent (Supplementary Table S2). Therefore, this structure was used in all further experiments. The likely explanation of its enhanced potency is that a more lipophilic double-TPP+ mitoDFO is better at carrying the highly hydrophilic deferoxamine moiety across the membrane. On the other hand, the addition of the FITC moiety to mitoDFO decreased its effect, probably due to its bigger size and decreased lipophilicity (Supplementary Table S2). Of note, mitoDFO presented here is different to the mitochondrially targeted deferoxamine synthesized by Alta and colleagues (referred to as TPP-DFO; ref. 50), who reported it as a nontoxic chelator with antioxidant and protective activity against iron overload, with some evidence of its biological effects and virtually no results on its anticancer effect. Furthermore, Alta's structure presents a two-carbon linker between the TPP group and the mother compound, and contains a single TPP group, making it much less lipophilic when compared with mitoDFO. Similarly, TPP-DFO was designed so that the labile imine bond can be hydrolyzed under physiologic conditions, potentially releasing free deferoxamine inside cells, whereas mitoDFO comprises an amine bond that is much less prone to intracellular hydrolysis. These differences point to the fact that both compounds have distinct mechanisms of action, and opposite effects on cells, that is, TPP-DFO is a protective compound while mitoDFO is a toxic agent in cancer cells. In fact, the IC50 reported for TPP-DFO in A2780 human cells is 135 μmol/L, while IC50 values of mitoDFO are in the range of 2–7 μmol/L. This suggests that the length and identity of the linker, the chemical structure of the TPP+ attachment and the number of TPP+ groups present, considerably affect the activity of the synthesized compounds.

As deferoxamine is known to induce transferrin receptor 1 and reduce cytosolic ferritin as a consequence of iron starvation, we tested whether mitoDFO exerts a similar effect. Furthermore, we tested whether mitoDFO interferes with iron-dependent processes inside mitochondria, including [Fe-S] cluster and heme biosynthesis. Our results show that mitoDFO diminishes (i) mitochondrial [Fe-S] cluster biogenesis as evidenced by the decrease in ISCU, (ii) heme synthesis by downregulation of the last enzyme of heme biosynthesis, FECH, and (iii) the levels of [Fe-S] cluster/heme-containing subunits of individual mitochondrial respiratory complexes: NDUFA9 (CI), SDHB (CII), UQCRFS1 (CIII), and mtCO1 (CIV). The decrease in SDHB, ISCU, and FECH has already been reported in situations where [Fe-S] cluster biogenesis is impaired (51, 52) and thus is in agreement with our observations. Moreover, the decrease in [Fe-S] cluster availability was further confirmed by the aconitase in-gel activity assay, which showed a marked decrease in enzymatic activity in mitoDFO-treated cells. Of note, the enzymatic activity of mitochondrial aconitase (ACO2) was strongly reduced by mitoDFO, while such effect was much milder with deferoxamine. Aconitase 1 that lacks the [Fe-S] cluster behaves as IRP1 and binds the IRE sequences of genes related to iron metabolism, posttranscriptionally decreasing their translation (ferritin, FTH) or stabilizing their mRNA (transferrin receptor 1, TFR1). We observed that mitoDFO increased the level of TFR1 and decreased the level of FTH, although the effect was less prominent compared with deferoxamine. Moreover, mitoDFO also decreased the level of lipoylated proteins, possibly due to [Fe-S] cluster-dependent activity of lipoyl synthase (31, 52, 53), thus linking [Fe-S] cluster deficiency with other metabolic pathways, in agreement with low PDH E2 and αKGDH E2 following repression of [Fe-S] cluster biogenesis by lipopolysaccharide (51). Similarly, phosphorylation of EIF2α was increased by mitoDFO treatment, probably due to activation of the HRI kinase resulting from mitoDFO-induced decrease in heme levels.

Furthermore, we observed that mitoDFO treatment resulted in lower level of 55Fe-labeled proteins within mitochondria and the cytosol, and extended incubation (60 hours) led to an even stronger effect. An exception was the storage protein ferritin, which accumulated 55Fe, likely representing a “sink” for iron that cannot be incorporated into [Fe-S] clusters and heme.

Taken together, our data show that mitoDFO disturbs the [Fe-S] cluster biogenesis, leading to a decrease in the level and activity of [Fe-S] cluster/heme-containing proteins, confirming that mitoDFO deprives cancer cells of biologically active iron.

Mitochondrial ETC is instrumental for generation of the electrochemical gradient coupled with ATP synthesis and is also the main source of ROS (41, 54). We found that mitoDFO almost completely abolished oxygen consumption and led to a significant destabilization and disassembly of SCs including the respirasome, which is essential for CI-dependent respiration (40) an effect not observed in the case of deferoxamine. This is consistent with our previous reports on several TPP+-containing compounds that inhibit CI in malignant cells by blocking the movement of electrons from the catalytic site of CI to its coenzyme Q binding domain (16, 18). Of note, the effect of mitoDFO on the assembly of SCs is unlikely to explain the fast decrease in the OCR described above, because long time incubation (≥6 hours) is required to affect SC assembly while a decrease in OCR is evident within minutes. Importantly, we also show that mitoDFO leads to significant increase in ECAR and combination of the glycolysis inhibitor 2-DG with mitoDFO resulted in profound decrease in cellular ATP. This suggests that cancer cells, in response to mitoDFO, switch their bioenergetics toward glycolysis in order to generate ATP.

Because mitochondria are the main cellular source of ROS (41, 54), we assessed the effect of mitoDFO on ROS level and found that both total cellular ROS and mitochondrial superoxide levels were significantly increased and the antioxidant NAC was able to significantly protect against mitoDFO-induced cytotoxicity, confirming the role of ROS in the response to mitoDFO. The incomplete protection could reflect the fact that NAC is a water-soluble general ROS scavenger not directed to mitochondria.

Dysfunctional mitochondria differ in their intracellular localization and architecture (6, 55) and are divided into smaller organelles by the process of fission (56). The resulting mitochondrial fragments could then be degraded by mitophagy, thus maintaining a healthy mitochondrial pool (6, 42, 44). Our results show that mitoDFO induces mitochondrial fragmentation and decreases their perinuclear localization, increasing also mitochondrial acidification and PINK1-dependent mitophagy, supporting the finding that intracellular iron chelation induces engulfment of mitochondria by lysosomes (7). Because mitophagy maintains a pool of healthy mitochondria, it is likely that the observed induction of mitophagy is an adaptive response of cancer cells that are trying to maintain functional mitochondria and reduce generation of ROS (57). A report by Seebacher and colleagues linked novel cytotoxic iron chelators with lysosomal destabilization (58), which could also impact the process of mitophagy. However, we did not detect any effect of mitoDFO on lysosomal permeability assessed by the acridine orange method (59).

Delocalized lipophilic cations have been reported to accumulate preferentially in cancer cells (16–19, 60) due to their higher mitochondrial membrane potential (61, 62). Our results show that by tagging deferoxamine with TPP+, we increased its antiproliferative ability over 40-fold in several cancer cell lines. The IC50 values were in the range of 1–8 μmol/L (with two exceptions being higher than 25 μmol/L), and normal fibroblasts were more resistant to the agent than most breast, ovarian, or pancreatic cancer cells. A detailed analysis by real-time cell monitoring revealed that mitoDFO exerted a potent cytostatic effect at concentrations lower than 5 μmol/L, consistent with accumulation of cells in G1-phase of cell cycle, while nonmalignant fibroblasts were unaffected. Furthermore, the cytotoxic effect seen at higher concentrations was considerably more pronounced in cancer cells than in their nonmalignant counterparts and was present even in cell lines that were less responsive to the cytostatic effect of mitoDFO. Importantly, the cytotoxic effect elicited by mitoDFO was dependent on the iron-chelating activity as mitoDFO precomplexed with iron was significantly less toxic toward malignant cells.

Because the in vitro data suggested a profound cytostatic and cytotoxic effect of mitoDFO on cancer cells, the next step was to investigate the effect of the agent on tumor growth and metastasis in vivo. Our data show a significant antitumor effect of mitoDFO in both immuno-competent (Balb/c) and immuno-suppressed (NSG) mice, with a significant reduction of Ki67 staining in the treated syngeneic tumors, documenting an effect of mitoDFO on the proliferation of cancer cells in vivo. Furthermore, our analysis showed decreased level of ISCU, SDHB, and FECH proteins in mitoDFO-treated tumors, while transferrin receptor 1 was increased, suggesting the inhibition of [Fe-S] cluster and heme biogenesis and activation of the IRP/IRE system similarly to the in vitro data. Moreover, we also detected the induction of PINK1, suggesting active mitophagy. Such changes were not observed in liver tissue of mitoDFO-treated mice, and our results illustrate that mitoDFO does not affect systemic iron metabolism as serum, spleen, and liver iron and standard hematologic parameters were not changed. Furthermore, mice weight was not affected by mitoDFO treatment. This strengthens the premise that mitoDFO preferentially affects malignant cells while sparing normal cells, and allows us to postulate that mitoDFO may be a lead compound in the design of novel anticancer therapies with high efficacy and lower general toxicity.

As cancer spreading and poor clinical outcome is connected with metastasis formation (45, 63), we analyzed the migrastatic properties of mitoDFO. Our results show significantly reduced cellular invasion in the 3D in vitro spheroid model and a strong migrastatic activity in the mouse model of lung metastasis formation by 4T1 cells (27) while deferoxamine failed to show such effect. This confirms the critical importance of effective mitochondrial metabolism for the energy demanding process of cancer cell invasiveness (64). It is also possible that highly migratory cells possess higher ROS level and mitoDFO-induced increase in ROS exceeds the sustainable ROS level, leading to cytostatic and cytotoxic effect, thus stopping cellular migration.

Interestingly, our results show that mitoDFO is more efficient in delaying the tumor growth of mouse 4T1 cells in immunocompetent Balb/c mice compared with immunocompromised animals with breast or pancreatic human cells. It is possible that the in vivo effect of mitoDFO could be partially dependent on immune system, in agreement with finding that induction of mitophagy increases antigen presentation of malignant cells in vivo, resulting in immune system-mediated recognition and elimination of cancer cells (65).

In summary, mitoDFO represents a novel approach that targets iron metabolism of cancer cells, representing an agent with considerable cytostatic, cytotoxic, and migrastatic properties derived from an FDA-approved drug for treating iron overload. Targeting mitochondrial iron metabolism thus represents one of the possible strategies that might be the basis for the development of next-generation antineoplastic treatments. Of importance, mitoDFO targets a critical process of the cell, the synthesis of biologically active iron-containing cofactors, required for a wide range of cancer cells to maintain high rates of proliferation and the ensuing tumor progression. This also means that our new compound does not target a single protein or pathway, by virtue of which, frequent complications that compromise cancer therapy, such as genetic instability and multiple mutations, are unlikely to affect the response to mitoDFO.

J. Stursa reports a patent for PCT/CZ2018/050036, WO/2019/015701, EP3655414, US20200078379 pending. L. Werner reports grants from Czech Academy of Sciences during the conduct of the study; in addition, L. Werner has a patent for PCT/CZ2018/050036, WO/2019/015701, EP3655414, US20200078379 (deferoxamine derivatives as medicaments) pending. J. Truksa reports grants from Czech Science Foundation and Ministry of Education, Youth, and Sport of the Czech Republic during the conduct of the study; in addition, J. Truksa has a patent for PCT/CZ2018/050036, WO/2019/015701, EP3655414, US20200078379 (deferoxamine derivatives as medicaments) pending. No disclosures were reported by the other authors.

C. Sandoval-Acuña: Conceptualization, data curation, formal analysis, supervision, investigation, methodology, writing–original draft, writing–review and editing. N. Torrealba: Data curation, formal analysis, investigation, methodology, writing–review and editing. V. Tomkova: Investigation, methodology, writing–review and editing. S.B. Jadhav: Investigation, methodology. K. Blazkova: Investigation, methodology. L. Merta: Investigation, methodology. S. Lettlova: Investigation, methodology, writing–review and editing. M.K. Adamcová: Resources. D. Rosel: Supervision, validation, methodology, writing–review and editing. J. Brábek: Supervision, validation, methodology, writing–review and editing. J. Neuzil: Conceptualization, funding acquisition, methodology, project administration, writing–review and editing. J. Stursa: Funding acquisition, investigation, methodology, writing–original draft, writing–review and editing. L. Werner: Resources, funding acquisition, investigation, methodology, project administration, writing–review and editing. J. Truksa: Conceptualization, resources, data curation, supervision, funding acquisition, validation, investigation, methodology, writing–original draft, project administration, writing–review and editing.

This work has been supported by GACR grants 16-12816S and 18-13103S, and the institutional support of the Czech Academy of Sciences RVO: 86652036 to J. Truksa, and GACR grants 17-01192J and 18-10832S, an AZV grant 17-30138A, and funding from the Australian Research Council to J. Neuzil It was further supported by the MEYS of CR within the LQ1604 National Sustainability Program II (Project BIOCEV-FAR). The results were obtained in cooperation with Czech Centre for Phenogenomics supported by the MEYS of CR no. LM2018126 and by MEYS and ERDF OP VaVpI CZ.1.05/2.1.00/19.039. This publication is a result of the project implementation: “The equipment for metabolomic and cell analyses,” registration number CZ.1.05/2.1.00/19.0400, supported by Research and Development for Innovations Operational Programme (RDIOP) cofinanced by European regional development fund and the state budget of the Czech Republic. The CMS-Biocev (Biophysical techniques/Crystallization/Diffraction/Structural mass spectrometry) was supported by MEYS CR (LM2015043). L. Werner was supported in part by the Research Programme 18—Strategy AV21 of the Czech Academy of Sciences.

The authors thank Roberto Bravo-Sagua from INTA, University of Chile, for his cordial assistance with microscopy images processing and analysis. The authors acknowledge Imaging Methods Core Facility at BIOCEV, institution supported by the MEYS CR (LM2018129 Czech-BioImaging) and ERDF (project No. CZ.02.1.01/0.0/0.0/16_013/0001775 and CZ.02.1.01/0.0/0.0/18_046/0016045) for their support with obtaining imaging and flow cytometry data presented in this article. Graphical abstract was created using BioRender.

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.
Sheftel
AD
,
Mason
AB
,
Ponka
P
. 
The long history of iron in the Universe and in health and disease
.
Biochim Biophys Acta
2012
;
1820
:
161
87
.
2.
Paul
VD
,
Lill
R
. 
Biogenesis of cytosolic and nuclear iron-sulfur proteins and their role in genome stability
.
Biochim Biophys Acta
2015
;
1853
:
1528
39
.
3.
Gallagher
PS
,
Larkin
M
,
Thillainadesan
G
,
Dhakshnamoorthy
J
,
Balachandran
V
,
Xiao
H
, et al
Iron homeostasis regulates facultative heterochromatin assembly in adaptive genome control
.
Nat Struct Mol Biol
2018
;
25
:
372
83
.
4.
Huang
ML
,
Lane
DJ
,
Richardson
DR
. 
Mitochondrial mayhem: the mitochondrion as a modulator of iron metabolism and its role in disease
.
Antioxid Redox Signal
2011
;
15
:
3003
19
.
5.
Ponka
P
. 
Cell biology of heme
.
Am J Med Sci
1999
;
318
:
241
56
.
6.
Kubli
DA
,
Gustafsson
AB
. 
Mitochondria and mitophagy: the yin and yang of cell death control
.
Circ Res
2012
;
111
:
1208
21
.
7.
Allen
GF
,
Toth
R
,
James
J
,
Ganley
IG
. 
Loss of iron triggers PINK1/Parkin-independent mitophagy
.
EMBO Rep
2013
;
14
:
1127
35
.
8.
Richardson
DR
,
Kalinowski
DS
,
Lau
S
,
Jansson
PJ
,
Lovejoy
DB
. 
Cancer cell iron metabolism and the development of potent iron chelators as anti-tumour agents
.
Biochim Biophys Acta
2009
;
1790
:
702
17
.
9.
Rychtarcikova
Z
,
Lettlova
S
,
Tomkova
V
,
Korenkova
V
,
Langerova
L
,
Simonova
E
, et al
Tumor-initiating cells of breast and prostate origin show alterations in the expression of genes related to iron metabolism
.
Oncotarget
2017
;
8
:
6376
98
.
10.
Lui
GY
,
Kovacevic
Z
,
Richardson
V
,
Merlot
AM
,
Kalinowski
DS
,
Richardson
DR
. 
Targeting cancer by binding iron: Dissecting cellular signaling pathways
.
Oncotarget
2015
;
6
:
18748
79
.
11.
Chitambar
CR
. 
The therapeutic potential of iron-targeting gallium compounds in human disease: From basic research to clinical application
.
Pharmacol Res
2017
;
115
:
56
64
.
12.
Lui
GY
,
Obeidy
P
,
Ford
SJ
,
Tselepis
C
,
Sharp
DM
,
Jansson
PJ
, et al
The iron chelator, deferasirox, as a novel strategy for cancer treatment: oral activity against human lung tumor xenografts and molecular mechanism of action
.
Mol Pharmacol
2013
;
83
:
179
90
.
13.
Kovar
J
,
Stunz
LL
,
Stewart
BC
,
Kriegerbeckova
K
,
Ashman
RF
,
Kemp
JD
. 
Direct evidence that iron deprivation induces apoptosis in murine lymphoma 38C13
.
Pathobiology
1997
;
65
:
61
8
.
14.
Kemp
JD
,
Cardillo
T
,
Stewart
BC
,
Kehrberg
E
,
Weiner
G
,
Hedlund
B
, et al
Inhibition of lymphoma growth in vivo by combined treatment with hydroxyethyl starch deferoxamine conjugate and IgG monoclonal antibodies against the transferrin receptor
.
Cancer Res
1995
;
55
:
3817
24
.
15.
Murphy
MP
,
Smith
RA
. 
Targeting antioxidants to mitochondria by conjugation to lipophilic cations
.
Annu Rev Pharmacol Toxicol
2007
;
47
:
629
56
.
16.
Rohlenova
K
,
Sachaphibulkij
K
,
Stursa
J
,
Bezawork-Geleta
A
,
Blecha
J
,
Endaya
B
, et al
Selective disruption of respiratory supercomplexes as a new strategy to suppress Her2high breast cancer
.
Antioxid Redox Signal
2017
;
26
:
84
103
.
17.
Truksa
J
,
Dong
LF
,
Rohlena
J
,
Stursa
J
,
Vondrusova
M
,
Goodwin
J
, et al
Mitochondrially targeted vitamin E succinate modulates expression of mitochondrial DNA transcripts and mitochondrial biogenesis
.
Antioxid Redox Signal
2015
;
22
:
883
900
.
18.
Boukalova
S
,
Stursa
J
,
Werner
L
,
Ezrova
Z
,
Cerny
J
,
Bezawork-Geleta
A
, et al
Mitochondrial targeting of metformin enhances its activity against pancreatic cancer
.
Mol Cancer Ther
2016
;
15
:
2875
86
.
19.
Sandoval-Acuna
C
,
Fuentes-Retamal
S
,
Guzman-Rivera
D
,
Peredo-Silva
L
,
Madrid-Rojas
M
,
Rebolledo
S
, et al
Destabilization of mitochondrial functions as a target against breast cancer progression: role of TPP(+)-linked-polyhydroxybenzoates
.
Toxicol Appl Pharmacol
2016
;
309
:
2
14
.
20.
Tong
WH
,
Rouault
TA
. 
Functions of mitochondrial ISCU and cytosolic ISCU in mammalian iron-sulfur cluster biogenesis and iron homeostasis
.
Cell Metab
2006
;
3
:
199
210
.
21.
Morrison
GR
. 
Fluorometric microdetermination of heme protein
.
Anal Chem
1965
;
37
:
1124
6
.
22.
Pesta
D
,
Gnaiger
E
. 
High-resolution respirometry: OXPHOS protocols for human cells and permeabilized fibers from small biopsies of human muscle
.
Methods Mol Biol
2012
;
810
:
25
58
.
23.
Vondrusova
M
,
Bezawork-Geleta
A
,
Sachaphibulkij
K
,
Truksa
J
,
Neuzil
J
. 
The effect of mitochondrially targeted anticancer agents on mitochondrial (super)complexes
.
Methods Mol Biol
2015
;
1265
:
195
208
.
24.
Yamashita
SI
,
Kanki
T
. 
Detection of hypoxia-induced and iron depletion-induced mitophagy in mammalian cells
.
Methods Mol Biol
2018
;
1759
:
141
9
.
25.
Tomkova
V
,
Sandoval-Acuna
C
,
Torrealba
N
,
Truksa
J
. 
Mitochondrial fragmentation, elevated mitochondrial superoxide and respiratory supercomplexes disassembly is connected with the tamoxifen-resistant phenotype of breast cancer cells
.
Free Radic Biol Med
2019
;
143
:
510
21
.
26.
Jobe
NP
,
Zivicova
V
,
Mifkova
A
,
Rosel
D
,
Dvorankova
B
,
Kodet
O
, et al
Fibroblasts potentiate melanoma cells in vitro invasiveness induced by UV-irradiated keratinocytes
.
Histochem Cell Biol
2018
;
149
:
503
16
.
27.
Pulaski
BA
,
Ostrand-Rosenberg
S
. 
Mouse 4T1 breast tumor model
.
Curr Protoc Immunol
2000
;
Unit 20.2
: 20.2.1–20.2.16
28.
Torrance
JD
,
Bothwell
TH
. 
Tissue iron stores
.
In
:
Cook
JD
, editor.
Methods in hematology
.
New York
:
Churchill Livingstone
; 
1980
. p.
90
115
.
29.
Bayram
J
,
Malcova
I
,
Sinkovec
L
,
Holubova
J
,
Streparola
G
,
Jurnecka
D
, et al
Cytotoxicity of the effector protein BteA was attenuated in Bordetella pertussis by insertion of an alanine residue
.
PLoS Pathog
2020
;
16
:
e1008512
.
30.
Richardson
DR
,
Tran
EH
,
Ponka
P
. 
The potential of iron chelators of the pyridoxal isonicotinoyl hydrazone class as effective antiproliferative agents
.
Blood
1995
;
86
:
4295
306
.
31.
Harmer
JE
,
Hiscox
MJ
,
Dinis
PC
,
Fox
SJ
,
Iliopoulos
A
,
Hussey
JE
, et al
Structures of lipoyl synthase reveal a compact active site for controlling sequential sulfur insertion reactions
.
Biochem J
2014
;
464
:
123
33
.
32.
La
P
,
Yang
G
,
Dennery
PA
. 
Mammalian target of rapamycin complex 1 (mTORC1)-mediated phosphorylation stabilizes ISCU protein: implications for iron metabolism
.
J Biol Chem
2013
;
288
:
12901
9
.
33.
Yang
C
,
Matro
JC
,
Huntoon
KM
,
Ye
DY
,
Huynh
TT
,
Fliedner
SM
, et al
Missense mutations in the human SDHB gene increase protein degradation without altering intrinsic enzymatic function
.
FASEB J
2012
;
26
:
4506
16
.
34.
Crooks
DR
,
Ghosh
MC
,
Haller
RG
,
Tong
WH
,
Rouault
TA
. 
Posttranslational stability of the heme biosynthetic enzyme ferrochelatase is dependent on iron availability and intact iron-sulfur cluster assembly machinery
.
Blood
2010
;
115
:
860
9
.
35.
Collins
DW
,
Gudiseva
HV
,
Chavali
VRM
,
Trachtman
B
,
Ramakrishnan
M
,
Merritt
WT
 III
, et al
The MT-CO1 V83I polymorphism is a risk factor for primary open-angle glaucoma in African American men
.
Invest Ophthalmol Vis Sci
2018
;
59
:
1751
9
.
36.
Bottani
E
,
Cerutti
R
,
Harbour
ME
,
Ravaglia
S
,
Dogan
SA
,
Giordano
C
, et al
TTC19 plays a husbandry role on UQCRFS1 turnover in the biogenesis of mitochondrial respiratory complex III
.
Mol Cell
2017
;
67
:
96
105
.
37.
Pryde
KR
,
Taanman
JW
,
Schapira
AH
. 
A LON-ClpP proteolytic axis degrades complex I to extinguish ROS production in depolarized mitochondria
.
Cell Rep
2016
;
17
:
2522
31
.
38.
Hentze
MW
,
Kuhn
LC
. 
Molecular control of vertebrate iron metabolism: mRNA-based regulatory circuits operated by iron, nitric oxide, and oxidative stress
.
Proc Natl Acad Sci U S A
1996
;
93
:
8175
82
.
39.
Zhang
Q
,
Du
R
,
Reis Monteiro Dos Santos
GR
,
Yefidoff-Freedman
R
,
Bohm
A
,
Halperin
J
, et al
New activators of eIF2alpha Kinase Heme-Regulated Inhibitor (HRI) with improved biophysical properties
.
Eur J Med Chem
2020
;
187
:
111973
.
40.
Letts
JA
,
Fiedorczuk
K
,
Sazanov
LA
. 
The architecture of respiratory supercomplexes
.
Nature
2016
;
537
:
644
8
.
41.
Murphy
MP
. 
How mitochondria produce reactive oxygen species
.
Biochem J
2009
;
417
:
1
13
.
42.
Scheibye-Knudsen
M
,
Fang
EF
,
Croteau
DL
,
Wilson
DM
, 3rd,
Bohr
VA
. 
Protecting the mitochondrial powerhouse
.
Trends Cell Biol
2015
;
25
:
158
70
.
43.
Thomas
RL
,
Kubli
DA
,
Gustafsson
AB
. 
Bnip3-mediated defects in oxidative phosphorylation promote mitophagy
.
Autophagy
2011
;
7
:
775
7
.
44.
Ding
WX
,
Yin
XM
. 
Mitophagy: mechanisms, pathophysiological roles, and analysis
.
Biol Chem
2012
;
393
:
547
64
.
45.
Gandalovicova
A
,
Rosel
D
,
Fernandes
M
,
Vesely
P
,
Heneberg
P
,
Cermak
V
, et al
Migrastatics-anti-metastatic and anti-invasion drugs: promises and challenges
.
Trends Cancer
2017
;
3
:
391
406
.
46.
Torti
SV
,
Torti
FM
. 
Iron and cancer: more ore to be mined
.
Nat Rev Cancer
2013
;
13
:
342
55
.
47.
Richardson
DR
. 
Iron chelators as therapeutic agents for the treatment of cancer
.
Crit Rev Oncol Hematol
2002
;
42
:
267
81
.
48.
Buss
JL
,
Torti
FM
,
Torti
SV
. 
The role of iron chelation in cancer therapy
.
Curr Med Chem
2003
;
10
:
1021
34
.
49.
Harima
H
,
Kaino
S
,
Takami
T
,
Shinoda
S
,
Matsumoto
T
,
Fujisawa
K
, et al
Deferasirox, a novel oral iron chelator, shows antiproliferative activity against pancreatic cancer in vitro and in vivo
.
BMC Cancer
2016
;
16
:
702
.
50.
Alta
RYP
,
Vitorino
HA
,
Goswami
D
,
Teresa Machini
M
,
Esposito
BP
. 
Triphenylphosphonium-desferrioxamine as a candidate mitochondrial iron chelator
.
Biometals
2017
;
30
:
709
18
.
51.
Tong
WH
,
Maio
N
,
Zhang
DL
,
Palmieri
EM
,
Ollivierre
H
,
Ghosh
MC
, et al
TLR-activated repression of Fe-S cluster biogenesis drives a metabolic shift and alters histone and tubulin acetylation
.
Blood Adv
2018
;
2
:
1146
56
.
52.
Maio
N
,
Kim
KS
,
Holmes-Hampton
G
,
Singh
A
,
Rouault
TA
. 
Dimeric ferrochelatase bridges ABCB7 and ABCB10 homodimers in an architecturally defined molecular complex required for heme biosynthesis
.
Haematologica
2019
;
104
:
1756
67
.
53.
McCarthy
EL
,
Booker
SJ
. 
Destruction and reformation of an iron-sulfur cluster during catalysis by lipoyl synthase
.
Science
2017
;
358
:
373
7
.
54.
Giulivi
C
,
Boveris
A
,
Cadenas
E
. 
Hydroxyl radical generation during mitochondrial electron transfer and the formation of 8-hydroxydesoxyguanosine in mitochondrial DNA
.
Arch Biochem Biophys
1995
;
316
:
909
16
.
55.
Ploumi
C
,
Daskalaki
I
,
Tavernarakis
N
. 
Mitochondrial biogenesis and clearance: a balancing act
.
FEBS J
2017
;
284
:
183
95
.
56.
Kiryu-Seo
S
,
Tamada
H
,
Kato
Y
,
Yasuda
K
,
Ishihara
N
,
Nomura
M
, et al
Mitochondrial fission is an acute and adaptive response in injured motor neurons
.
Sci Rep
2016
;
6
:
28331
.
57.
Chourasia
AH
,
Boland
ML
,
Macleod
KF
. 
Mitophagy and cancer
.
Cancer Metab
2015
;
3
:
4
.
58.
Seebacher
NA
,
Richardson
DR
,
Jansson
PJ
. 
A mechanism for overcoming P-glycoprotein-mediated drug resistance: novel combination therapy that releases stored doxorubicin from lysosomes via lysosomal permeabilization using Dp44mT or DpC
.
Cell Death Dis
2016
;
7
:
e2510
.
59.
Repnik
U
,
Cesen
MH
,
Turk
B
. 
The use of lysosomotropic dyes to exclude lysosomal membrane permeabilization
.
Cold Spring Harb Protoc
2016
;
2016
. DOI: 10.1101/pdb.prot087106.
60.
Dong
LF
,
Jameson
VJ
,
Tilly
D
,
Prochazka
L
,
Rohlena
J
,
Valis
K
, et al
Mitochondrial targeting of alpha-tocopheryl succinate enhances its pro-apoptotic efficacy: a new paradigm for effective cancer therapy
.
Free Radic Biol Med
2011
;
50
:
1546
55
.
61.
Modica-Napolitano
JS
,
Aprille
JR
. 
Delocalized lipophilic cations selectively target the mitochondria of carcinoma cells
.
Adv Drug Deliv Rev
2001
;
49
:
63
70
.
62.
Gogvadze
V
,
Orrenius
S
,
Zhivotovsky
B
. 
Mitochondria in cancer cells: what is so special about them?
Trends Cell Biol
2008
;
18
:
165
73
.
63.
Mehlen
P
,
Puisieux
A
. 
Metastasis: a question of life or death
.
Nat Rev Cancer
2006
;
6
:
449
58
.
64.
Zanotelli
MR
,
Goldblatt
ZE
,
Miller
JP
,
Bordeleau
F
,
Li
J
,
VanderBurgh
JA
, et al
Regulation of ATP utilization during metastatic cell migration by collagen architecture
.
Mol Biol Cell
2018
;
29
:
1
9
.
65.
Ziegler
PK
,
Bollrath
J
,
Pallangyo
CK
,
Matsutani
T
,
Canli
O
,
De Oliveira
T
, et al
Mitophagy in intestinal epithelial cells triggers adaptive immunity during tumorigenesis
.
Cell
2018
;
174
:
88
101
.