Download PDF
Review  |  Open Access  |  30 Aug 2022

Reactive oxygen species in the progression and treatment of malignant mesothelioma

Views: 911 |  Downloads: 594 |  Cited:   0
J Cancer Metastasis Treat 2022;8:36.
10.20517/2394-4722.2022.41 |  © The Author(s) 2022.
Author Information
Article Notes
Cite This Article

Abstract

Malignant mesothelioma (MM) is an aggressive cancer that affects the pleural and peritoneal mesothelial lining of the lungs and abdomen. Survival rates for patients with MM remain extremely low and effective treatments are limited. MM tumors harbor both genotypic and phenotypic features that indicate MM tumor cells are under increased oxidative stress, similar to other aggressive cancers. This increased oxidative stress in MM cells supports aggressive growth while providing a therapeutic vulnerability exploitable by redox-modulating compounds. MM tumor cells also exhibit altered mitochondrial structure and function that contribute to the disease through perturbations in metabolism and reactive oxygen species (ROS) production and metabolism. Targeting the altered redox status in cancer through increasing cellular ROS levels directly or inhibiting cellular antioxidant pathways and disrupting ROS scavenging mechanisms has become an exciting area for therapeutic intervention. This review discusses ROS sources and signaling, mitochondrial structure and function and targeting mitochondria ROS as a therapeutic approach for the treatment of MM.

Keywords

Malignant mesothelioma, targeting cellular redox status, pro-oxidant therapy

INTRODUCTIONS

Malignant mesothelioma (MM) is an aggressive form of cancer that primarily affects the pleural and peritoneal mesothelial lining of the lungs and abdomen. Although asbestos exposure is a major risk factor for the development of MM, little is known about the etiology of the disease[1]. Similar to the majority of other solid tumors, MM is strongly linked to altered metabolism, changes in mitochondrial dynamics and an imbalance in the production and clearance of reactive oxygen species (ROS). Because of this, ROS production and metabolism have become an exciting target for cancer treatments[2] including MM[3]. Numerous approaches currently used in the clinic to treat MM, including cisplatin, have also been shown to modulate ROS levels[4]. Recently, thiostrepton (TS), an inhibitor of mitochondrial peroxiredoxin 3 that induces elevated ROS levels and cell death[5,6], entered phase 1/2 clinical trials (NCT05278975), providing a new and exciting redox-dependent therapy for the treatment of MM. This review will discuss the role of ROS in cancer development and the unique mitochondrial dynamics and redox status in MM that may be an effective target for anticancer therapies.

CANCER AND ROS

Ros in cell signaling

The role of ROS in normal and cancer cells is far more than just damaging or a byproduct of oxidative metabolism. ROS contribute to cell signaling cascades through a process termed “redox-dependent signaling”[7]. ROS can directly or indirectly oxidize cysteine residues in proteins[8] through hydrogen peroxide (H2O2)-mediated oxidation of target proteins or through peroxiredoxin (Prx)-dependent redox relays[9]. ROS are produced by intracellular and extracellular sources, including asbestos, and dynamically regulate numerous cell signaling pathways[10]. Intracellular sources of ROS, such as NADPH oxidases and the mitochondrial electron transport chain, participate in redox-dependent signaling spatially and temporally[11,12] [Figure 1]. Control over the amount, timing and location of ROS contributes to specific redox signaling events, akin to cellular control over protein phosphorylation cascades[13]. One well-known redox signaling event is in the cell’s response to hypoxia, which is mediated by the stabilization of hypoxia-inducible factors (HIFs). Under normal conditions, prolyl hydroxylase domain protein 2 (PHD2) prevents HIF stabilization by hydroxylating two of its proline residues, marking it for degradation[14]. PHD2 is deactivated at low oxygen levels, allowing HIF stabilization. A study performed by Chandel et al. found that the production of mROS was required for HIF stabilization under hypoxia, though the mechanism is still unclear[15,16]. In cancer cells, HIF stabilization stimulates angiogenesis, glycolysis, and cell survival, key hallmarks of tumorigenesis[2].

Reactive oxygen species in the progression and treatment of malignant mesothelioma

Figure 1. The redox landscape of MM tumors and methods of targeting ROS: ROS are primarily derived from asbestos fibers, NADPH Oxidases (NOXs) and mitochondria in MM tumor cells. ROS levels are balanced by the expression of ROS scavenging pathways, including the TR-TRX-PRX antioxidant axis, and increased GSH synthesis through Cystine (Cys-Cys) import via the SLC7A11 uniporter. FOXM1 and NRF2 are redox-responsive transcription factors that support ROS scavenging gene expression. BAP1 alters cellular redox status through downregulation of the SLC7A11 uniporter and disruption of mitochondrial bioenergetics. ROS inhibit PTEN phosphatase activity driving increased PI3K activity that supports tumor cell proliferation and survival. Cisplatin (CDDP) targets DNA, TRX and GSH which lead to increased ROS levels. Thiostrepton (TS) inhibits mitochondrial PRX3 which lead to increased mitochondrial ROS. High ROS levels are incompatible with cell survival.

ROS IN TUMORIGENESIS AND TUMOR CELL RESPONCE TO ROS

The increased production of ROS in tumor cells is described as a “double-edged sword” in the process of tumorigenesis[17]. Increased ROS, often driven by oncogene activation[18], must be managed by cancer cells by upregulating various antioxidant networks, as excessive oxidative stress would normally induce senescence and/or apoptosis in cells[19] [Figure 1]. Conversely, ROS is also thought to promote cell proliferation by inducing DNA mutations and activating redox-dependent signaling pathways[17]. One specific way ROS may promote tumorigenesis is by activating the phosphoinositide 3-kinase (PI3K) pathway. This pathway is upregulated in cancer cells and promotes cell proliferation, survival, and mobility[20]. It is also known that ROS inhibits phosphatase and tensin homolog (PTEN) activity, which allows for constitutive expression of PI3K when inactivated[21,22] [Figure 1]. Increased ROS levels in human MM cells enhance the expression of the oncogenic transcription factor FOXM1 which supports cell cycle progression and escape from oxidative stress[23-25] [Figure 1]. ROS also have the ability to alter metabolism, an example of which is by oxidation of key cysteine residues in pyruvate kinase M2 (PKM2). The oxidation of Cys358 on PKM2 is thought to increase pentose phosphate pathway flux and cell proliferation in hypoxic conditions[26]. Inhibition of PKM2 has been associated with increased tumorigenesis[27,28].

Tumor cells must balance the increased levels of ROS associated with transformation and therefore acquire genetic and phenotypic features to survive under otherwise inhospitable redox conditions[29]. The transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) is considered to be the predominant regulator of antioxidant enzyme expression. NRF2 is activated under conditions of oxidative stress through a mechanism involving the redox-dependent release of KEAP1, allowing NRF2 translocation to the nucleus and regulation of antioxidant genes through antioxidant response element (ARE) binding[30]. NRF2 is responsible for producing and regenerating glutathione (GSH), a major antioxidant cofactor in the cell[31]. NRF2 also has a role in the production of NADPH, which is used to regenerate many antioxidant enzymes[17,32] including the thioredoxin reductase (TR) - thioredoxin (TRX) - peroxiredoxin (PRX) antioxidant network[33] [Figure 1].

The forkhead box (FOX) family of proteins also plays a significant role in tumor cell escape from oxidative stress. The FOX family of proteins plays an important role in cell proliferation, cell metabolism, stress responses, and aging[17]. FOXO specifically seems to regulate the expression of superoxide dismutases (SODs), catalase and sestrin 3 under conditions of oxidative stress[34]. FOXM1 expression is increased in H-RAS transformed cells and is required for balancing cellular ROS levels and escaping from oncogene-induced senescence[25]. FOXM1 expression is upregulated in MM and plays an important role in MM cell survival (see below). p53, a prominent tumor suppressor gene, is also thought to have a regulatory role in antioxidant gene expression, though its expression has been thought to produce both pro- and antioxidant responses[35]. It is thought to promote the production of glutamate which is required for GSH synthesis[36]. p53 also promotes sestrin 1 and 2 expression[37]. Experiments with mutated forms of p53 that prevent its ability to induce apoptosis and cell cycle arrest showed that it still retained a tumor suppressive ability[38]. This suggests that its function appears to be partly due to its ability to suppress ROS production[39]. Interestingly, p53, although mutated in many forms of cancer, is almost always a wild type in MM cell lines. Its function, however, is usually inhibited by other gene deletions frequently found in MM cells[40].

ROS IN MALIGNANT MESOTHELIOMA

Human MM cell lines demonstrate elevated levels of antioxidant enzymes such as manganese superoxide dismutase (MnSOD)[41], peroxiredoxins (PRXs) and the mitochondrial thioredoxin reductase 2 (TrxR2) - thioredoxin 2 (Trx2) - peroxiredoxin 3 (Prx3) antioxidant network[42] compared to normal mesothelial cells. The upregulation of MnSOD is significant as it is the only antioxidant induced by asbestos exposure[43] and contributes to the high resistance to oxidant chemotherapeutic drugs displayed by MM cells[44]. MM cells also display increased expression of many PRXs[45], a family of H2O2 scavenging enzymes found in the cytoplasm (PRX1, PRX2), mitochondria (PRX3, PRX5) and endoplasmic reticulum (PRX4). Some members have notable functions, such as PRX2, which induces cell proliferation and protects cells from undergoing oxidative stress-induced apoptosis[46]. PRX3 protects tumor cells from apoptosis[47] and knockdown of PRX3 in human MM cells leads to reduced proliferation and altered cell cycle progression[5,23]. The role of PRX3 in cancer is still being uncovered, but its importance in maintaining mitochondrial redox status is clear as PRX3 is estimated to metabolize ~90% of mitochondrial H2O2[48]. Targeting PRX3 for the treatment of MM is a new avenue being explored in clinical trials (elaborated below).

Disruption of cellular redox status via depletion of glutathione (GSH) and iron-dependent lipid oxidation contributes to a type of programmed cell death termed ferroptosis[49]. Inactivation of NF2, LATS1, or LATS2, genes involved in signaling cascades that prevent ferroptosis, are recently identified mutations in MM cells that sensitize them to ferroptosis[50]. Approaches to targeting these mutations and taking advantage of this ferroptosis sensitivity, as well as looking into other possible pathways involved in ferroptosis, are an avenue for therapeutic intervention in MM. One proposed target is BAP1, the most commonly mutated gene in MM, which can inhibit ferroptosis when mutated[51,52]. Interestingly, fibroblasts harboring heterozygous BAP1 mutations show altered metabolite profiles and mitochondrial respiration, including reduced citric acid cycle metabolites and mitochondrial oxygen consumption[53]. It was concluded that BAP1 mutant fibroblasts are preferentially undergoing aerobic glycolysis (Warburg effect). Additional studies in a mesothelioma cell line (NCI-H226), which does not express BAP1, showed that reconstitution of BAP1 into these cell lines promoted ROS production, gene-expression changes indicative of increased cellular oxidative stress and increased sensitivity to exogenous H2O2[54] [Figure 1]. These results provide a path forward for looking into BAP1 status in the context of redox-dependent therapies.

FOX protein family members, including FOXM1, are thought to mediate tumorigenesis and promote the survival of MM cells[24]. High FOXM1 expression is found in MM cells, and tumors and knockdown of FOXM1 with siRNA slows the growth of MM cells[55,56]. FOXM1 also regulates the expression of genes involved in cell survival and cell cycle progression. The two oncogenic isoforms of the protein, FOXM1B and FOXM1C, act as transcriptional activators and are both upregulated in MM cancer cells, suggesting that FOXM1 may be a viable target for cancer treatment[24,57]. The expression of FOXM1 is induced by the production of H2O2[25,58]. FOXM1 counteracts oxidative stress by upregulating the expression of antioxidant enzymes such as SODs, catalase, and PRX3, which suppresses oncogene-induced senescence and supports tumor cell growth[25] [Figure 1]. PRX3 functions by metabolizing H2O2, which causes catalytic cysteine residues in PRX3 to form a disulfide bond. This bond is reduced by thioredoxin 2 (TRX2), regenerating PRX3 to its active conformation and able to metabolize another molecule of H2O2[59]. TRX2 is subsequently regenerated by TR2 using NADPH as the reductant[60]. Notably, TR2 and TRX2 are also upregulated in cancer cells, and measuring the activity of TR2 is proposed to be a useful way to monitor the growth of MM cell lines[61]. The TR2-TRX2-PRX3 system has been identified as a pathway for therapeutic intervention in preclinical MM cell and animals models, which will be discussed below.

DISRUPTION OF MITOCHONDRIAL DYNAMICS IN MALIGNANT MESOTHELIOMA

The mitochondria are thought of as a dynamic network that fuse (fusion) and divide (fission) in order to adapt to the metabolic needs of the cell[62]. The process of fission is mediated by the GTPases dynamin-related protein1 (Drp1) and dynamin 2 (Dnm2)[63]. Similarly, mitochondrial fusion is mediated by mitofusins Mfn1 and Mfn2 that help perform the fusion of two mitochondrial membranes[64]. In cancerous cells, the mitochondrial network is often fragmented, and redox-dependent signaling is believed to be key to these shape changes[65,66]. One of these pathways is hypoxia-induced mitochondrial fission, which occurs when HIFs induce Drp1 expression. In addition to mitochondrial fragmentation, HIF expression is linked to increased metastatic activity. This suggests that HIFs are an important mediator for both mitochondrial fission and cancer cell proliferation[67,68]. Another important mediator is the p38 mitogen-activated protein kinase (MAPK), which stimulates stress response pathways upon receiving redox signals[69]. One direct way is to phosphorylate Drp1 directly to induce mitochondrial fission[70].

The transcription factor nuclear factor κB (NF-κB), which plays a role in inflammation and is upregulated in cancer, is also thought to alter mitochondrial dynamics. Elevated NF-κB levels in cigarette smoke-induced mitochondrial fragmentation were found to correlate with increased expression of Drp1 and decreased expression of Mfn2[71]. More recent research has also shown that the principal component of that pathway is the NF-κB inducing kinase (NIK). In addition to having pro-fission activity, NIK was also shown to promote mitochondrial migration towards the cell periphery, which was shown to correlate with increased tumor invasiveness[72,73]. NF-κB activity is upregulated following asbestos exposure[74], and sustained NF-κB activity has been observed in MM cells[75]. Downregulation of NF-κB with Onconase decreased MM proliferation and invasion[76]. Although mitochondrial defects were not reported in these studies, the cellular responses observed warrant evaluating strategies targeting NF-κB with disruption of mitochondrial dynamics.

MM cells also appear to demonstrate a difference in mitochondrial morphologies. A study performed by Lennon et al. evaluated the fission/fusion rate of various MM cells using fractal dimension and lacunarity measurements to characterize mitochondrial architectures[77]. They found that MM cell lines displayed a low fractal dimension and high lacunarity compared to control mesothelial cells. The low fractal dimension and high lacunarity both indicate a high rate of mitochondrial fission[77]. DRP1 is upregulated in many cancer types and supports tumor growth and metastasis[78]. Mitochondrial dynamics, including fission, appear to be linked to cell cycle at the G2/M phase, as it is an important checkpoint to ensure an even distribution of mitochondria. Drp1, therefore, plays an important role in mediating cell cycle progression[79]. Interestingly, we found that PRX3 expression in MM cells mediates Drp1 expression and provides evidence that this affects mitochondrial dynamics through activation of Drp1. This was demonstrated in an experiment where cells failed to progress through the G2/M phase when PRX3 expression was inhibited and DRP1 phosphorylation at a key regulatory serine was coincidingly reduced[23]. Disabling the receptor tyrosine kinase MET with MGCD516 in MM cells led to a loss of DRP1 activity that accompanied reduced viability, migration, and invasion[80]. Combining MGCD516 with the proposed mitochondrial fission inhibitor Mdivi-1 induced cell death to a greater extent than either drug used alone[80]. Given that DRP1 expression is upregulated in MM[81], the unique mitochondrial morphologies of MM cells[77] and the connection between mitochondrial redox status, DRP1 activity and cell cycle progression[23] targeting mitochondrial morphology in MM may be a viable therapeutic approach. Lennon et al. (2016) also studied the effects of mitochondrial inhibitors metformin and Mdivi-1[77]. Metformin, which inhibits complex I of the electron transport chain, appeared to be highly effective against MM cell lines with a low fractal dimension and a high lacunarity. Mdivi-1, which inhibits Drp1, was most effective on cell lines with a high lacunarity. This study did not find any correlation with their mitochondrial metabolism measurements. Although the use of treatments like metformin is largely unproven, this suggests that mitochondrial dynamics may be a promising indicator of the effectiveness of cancer treatments[77]. These results suggest that mitochondrial dynamics are a useful target for MM treatment, as well as for other tumors characterized by high mitochondrial oxidant production.

STRATEGIES IN MODULATING MITOCHONDRIA AND REDOX SYSTEMS FOR THERAPEUTIC INTERVENTION

Several studies recognize the difficulty in treating cancer by downregulating ROS production for the same reasons that they are controversial in the role of cancer development. Suppressing ROS with general antioxidants, such as β-carotene and vitamin A, was found to enhance tumor growth[82,83]. Several other studies have criticized the commercial use of antioxidants as a means of cancer prevention. There have been few population-based studies supporting their use[17]. Inducing oxidative stress by selectively increasing cellular ROS or specifically targeting key antioxidant enzymes seems to be a more viable option [Table 1]. One approach is by directly increasing ROS levels within the cell. Many well-known cancer treatments, such as chemotherapy and radiation, already work by inducing oxidative stress[17]. However, caution must be exercised as inducing ROS in cells nonspecifically may induce chemoresistance[84,85]. Additional studies have shown that some ROS-inducing agents may be able to sensitize cancer cells to treatments like radiation therapy. One, in particular, is vitamin C (ascorbate) which acts as a pro-oxidant at higher doses[86] and has been shown to enhance sensitivity to radiation therapy in pancreatic cancer[87,88]. Other studies have shown that depletion of arginine, a critical amino acid in the biosynthesis of proteins, nitric oxide, and polyamine is an actionable approach for therapeutic intervention in argininosuccinate synthase I (ASSI) - negative tumors, including MM[89,90]. Depletion of arginine leads to mitochondrial dysfunction and increased ROS levels[91,92]. Pegargiminase (ADI-PEG 20; ADI) acts to degrade arginine and shows potent activity in ASS1 deficient MM tumors[93]. Given the potent effects on mitochondrial activity, including increased oxygen consumption and ROS levels following arginine depletion, or treatment with ADI-PEG 20, it will be interesting to determine the redox-dependent activity of this approach in the therapeutic response observed[92,94].

Table 1

Targeting ROS in cancers including malignant mesothelioma

Source citationTopicPMID
Chen et al.[86] (2007)
Alexander et al.[87] (2018)
Mehdi et al.[88] (2021)
Vitamin C (ascorbate)(17502596) (30254147)
(34639220)
Cunniff et al.[5] (2015)
Nelson et al.[6] (2021)
Newick et al.[55] (2012)
Cunniff et al.[56] (2013)
Modulating PRX3 and/or FOXM1(26011724) (33498547) (22761781)
(23018647)
Scalcon et al.[59] (2018)
Zhang et al.[99] (2011)
Thioredoxin and thioredoxin reductase inhibitors(29596885) (21215310)
Omenn et al.[82] (1996)
Alpha-Tocopherol et al.[83] (1994)
Vitamin E and Beta Carotene(8602180) (8127329)
Gorrini et al.[17] (2013)
Jezek et al.[65] (2021)
Reviews(24287781) (33418995)

A more specific approach to increasing ROS is by targeting antioxidant pathways. As many antioxidant systems are upregulated in cancer cells, they have been identified as important targets for treatment that selectively targets cancer. Thioredoxin (TRX) pathway inhibitors are an important target, as it is suggested that overexpression of TRX leads to chemoresistance to pro-oxidant therapies[59]. For example, cis-diamminedichloroplatinum (II)[45] (CDDP, cisplatin) is one of the few chemotherapeutic agents approved for use in MM treatment[95], and its cytotoxicity is partly attributed to its effects on TRX activity. Studies showed that cisplatin cytotoxicity was strongly correlated with thioredoxin reductase (TR) inhibition. This study also showed that an increase in TR expression correlated with cisplatin resistance[96]. Although cisplatin activity is attributed to DNA damage, most of the intracellular platinum content reacts with GSH, forming a bis-(glutathione)-platinum (GS-Pt) complex[97], which, notably, also demonstrated inhibitory effects on TR[98].

Triphenylmethanes, like brilliant green and gentian violet (GV), appear to act by inhibiting the mitochondrial thioredoxin isoform(Trx2)[99]. GV was shown to have potent cytotoxic activity against MM cells in culture and a xenograft model of MM[5]. These studies corroborated initial studies that GV was a potent TRX2 inhibitor as loss of TRX2 expression correlated with cytotoxicity[99]. Additionally, treatment of cells with GV led to significant increases in disulfide-bonded dimers of PRX3, the molecular species reduced by TRX2 and a significant increase in mitochondrial ROS levels[55].

Studies from our group identified the increased expression of FOXM1 in MM tumors and MM cell lines[55]. This observation led to testing the proposed FOXM1 inhibitor, TS, in preclinical models of MM. TS shows potent anticancer activity in a variety of tumor cell lines[100] and has been proposed to exert its anticancer activity through inhibition of FOXM1[101], the proteasome[102,103] and PRX3 activity[5,6]. Our group has been investigating the molecular mechanism and anticancer activity of TS in preclinical cell and animal models of MM and have deduced that PRX3 is a primary molecular target of TS[5,6,23,55,56,61] [Figure 1]. MM cells are more sensitive to TS compared to normal primary and immortalized mesothelial cell lines, and TS has potent in vivo activity in xenografts of human MM cells engrafted to the peritoneal cavity of immunocompromised mice. These studies have collectively shown that TS covalently crosslinks the active site Cys 108 and Cys 229 residues, inducing a stable covalent adduct across the dimer-dimer interface. Crosslinking of PRX3 increases cellular and mitochondrial ROS levels that can be inhibited by pre-treatment with the ROS scavenger N-acetylcysteine (NAC), indicating the redox dependency of TS cytotoxicity. The crosslinking of PRX3 by TS was detectable in tissue resected from mice harboring MM xenografts, providing evidence that the mechanism of PRX3 crosslinking by TS is preserved in vivo. Our recent work uncovered the specificity of TS for mitochondrial PRX3 versus the cytosolic peroxiredoxins PRX1 and PRX2. Structural transitions of PRX3, dependent on its oxidation status and the local pH environment of the mitochondrial matrix, support preferential adduction of PRX3 in MM cells. TS treatment of MM cells also leads to a loss in FOXM1 expression. The interplay between TS, PRX3, mROS and FOXM1 remains unclear as knockdown of PRX3 reduces FOXM1 levels and treatment of MM cells with mROS inducing agents (rotenone) leads to loss of FOXM1. Although more research is necessary to dissect this interplay, targeting PRX3 and FOXM1 with TS is an exciting therapeutic approach. TS is currently being tested in the MITOPE phase 1/2 clinical trial to evaluate activity in patients with malignant pleural effusion (MPE) arising from metastatic disease or M (NCT05278975).

CONCLUSIONS

ROS have a complicated role in the development of MM and many other cancers. Although a potent and cancer promoting signaling molecule, increased ROS and adaptations to oxidative stress in cancer cells, including MM, provide a redox vulnerability exploitable through redox-dependent therapies. Several preclinical and established cancer treatments exploit the increased ROS production observed in cancer by directly inducing oxidative stress or targeting complex cellular antioxidant networks. A secondary and complementary approach is targeting mitochondrial dynamics, as they are intertwined with many of the same redox processes. MM cell lines display both increased ROS production and altered mitochondrial dynamics; therefore, further evaluating these strategies is warranted for the treatment of MM.

DECLARATIONS

Authors’ contributions

Wrote and edited the manuscript: Cote A, Messier T, Cunniff B

Availability of data and materials

Not applicable.

Financial support and sponsorship

Brian Cunniff receives funding from RS Oncology, LLC through a Sponsored Research Agreement between the University of Vermont and RS Oncology, LLC.

Conflicts of interest

Brian Cunniff and Terri Messier are paid consultants for RS Oncology, LLC. Brian Cunniff is an equity holder in RS Oncology, LLC.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2022.

REFERENCES

1. Janes SM, Alrifai D, Fennell DA. Perspectives on the treatment of malignant pleural mesothelioma. N Engl J Med 2021;385:1207-18.

2. Sena LA, Chandel NS. Physiological roles of mitochondrial reactive oxygen species. Mol Cell 2012;48:158-67.

3. Urso L, Cavallari I, Sharova E, Ciccarese F, Pasello G, Ciminale V. Metabolic rewiring and redox alterations in malignant pleural mesothelioma. Br J Cancer 2020;122:52-61.

4. Kleih M, Böpple K, Dong M, et al. Direct impact of cisplatin on mitochondria induces ROS production that dictates cell fate of ovarian cancer cells. Cell Death Dis 2019;10:851.

5. Cunniff B, Newick K, Nelson KJ, et al. Disabling mitochondrial peroxide metabolism via combinatorial targeting of peroxiredoxin 3 as an effective therapeutic approach for malignant mesothelioma. PLoS One 2015;10:e0127310.

6. Nelson KJ, Messier T, Milczarek S, et al. Unique cellular and biochemical features of human mitochondrial peroxiredoxin 3 establish the molecular basis for its specific reaction with thiostrepton. Antioxidants (Basel) 2021;10:150.

7. Reczek CR, Chandel NS. ROS-dependent signal transduction. Curr Opin Cell Biol 2015;33:8-13.

8. Finkel T. Signal transduction by reactive oxygen species. J Cell Biol 2011;194:7-15.

9. Ježek P, Holendová B, Plecitá-Hlavatá L. Redox Signaling from mitochondria: signal propagation and its targets. Biomolecules 2020;10:93.

10. Hamanaka RB, Chandel NS. Mitochondrial reactive oxygen species regulate cellular signaling and dictate biological outcomes. Trends Biochem Sci 2010;35:505-13.

11. Enyedi B, Zana M, Donkó Á, Geiszt M. Spatial and temporal analysis of NADPH oxidase-generated hydrogen peroxide signals by novel fluorescent reporter proteins. Antioxid Redox Signal 2013;19:523-34.

12. Dröse S. Differential effects of complex II on mitochondrial ROS production and their relation to cardioprotective pre- and postconditioning. Biochim Biophys Acta 2013;1827:578-87.

13. Marzo N, Chisci E, Giovannoni R. The Role of hydrogen peroxide in redox-dependent signaling: homeostatic and pathological responses in mammalian cells. Cells 2018;7:156.

14. Kaelin WG Jr, Ratcliffe PJ. Oxygen sensing by metazoans: the central role of the HIF hydroxylase pathway. Mol Cell 2008;30:393-402.

15. Chandel NS, McClintock DS, Feliciano CE, et al. Reactive oxygen species generated at mitochondrial complex III stabilize hypoxia-inducible factor-1alpha during hypoxia: a mechanism of O2 sensing. J Biol Chem 2000;275:25130-8.

16. Kumar A, Vaish M, Karuppagounder SS, et al. HIF1α stabilization in hypoxia is not oxidant-initiated. Elife 2021;10:e72873.

17. Gorrini C, Harris IS, Mak TW. Modulation of oxidative stress as an anticancer strategy. Nat Rev Drug Discov 2013;12:931-47.

18. Ogrunc M, Di Micco R, Liontos M, et al. Oncogene-induced reactive oxygen species fuel hyperproliferation and DNA damage response activation. Cell Death Differ 2014;21:998-1012.

19. Vakifahmetoglu-Norberg H, Ouchida AT, Norberg E. The role of mitochondria in metabolism and cell death. Biochem Biophys Res Commun 2017;482:426-31.

20. Cantley LC. The phosphoinositide 3-kinase pathway. Science 2002;296:1655-7.

21. Lee SR, Yang KS, Kwon J, Lee C, Jeong W, Rhee SG. Reversible inactivation of the tumor suppressor PTEN by H2O2. J Biol Chem 2002;277:20336-42.

22. Leslie NR, Bennett D, Lindsay YE, Stewart H, Gray A, Downes CP. Redox regulation of PI 3-kinase signalling via inactivation of PTEN. EMBO J 2003;22:5501-10.

23. Cunniff B, Wozniak AN, Sweeney P, DeCosta K, Heintz NH. Peroxiredoxin 3 levels regulate a mitochondrial redox setpoint in malignant mesothelioma cells. Redox Biol 2014;3:79-87.

24. Laoukili J, Stahl M, Medema RH. FoxM1: at the crossroads of ageing and cancer. Biochim Biophys Acta 2007;1775:92-102.

25. Park HJ, Carr JR, Wang Z, et al. FoxM1, a critical regulator of oxidative stress during oncogenesis. EMBO J 2009;28:2908-18.

26. Anastasiou D, Poulogiannis G, Asara JM, et al. Inhibition of pyruvate kinase M2 by reactive oxygen species contributes to cellular antioxidant responses. Science 2011;334:1278-83.

27. Hitosugi T, Kang S, Vander Heiden MG, et al. Tyrosine phosphorylation inhibits PKM2 to promote the Warburg effect and tumor growth. Sci Signal 2009;2:ra73.

28. Christofk HR, Vander Heiden MG, Harris MH, et al. The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature 2008;452:230-3.

29. Diehn M, Cho RW, Lobo NA, et al. Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature 2009;458:780-3.

30. Rushmore TH, Morton MR, Pickett CB. The antioxidant responsive element. Activation by oxidative stress and identification of the DNA consensus sequence required for functional activity. J Biol Chem 1991;266:11632-9.

31. Meister A. Selective modification of glutathione metabolism. Science 1983;220:472-7.

32. Mitsuishi Y, Taguchi K, Kawatani Y, et al. Nrf2 redirects glucose and glutamine into anabolic pathways in metabolic reprogramming. Cancer Cell 2012;22:66-79.

33. Kim YJ, Ahn JY, Liang P, Ip C, Zhang Y, Park YM. Human prx1 gene is a target of Nrf2 and is up-regulated by hypoxia/reoxygenation: implication to tumor biology. Cancer Res 2007;67:546-54.

34. Nogueira V, Park Y, Chen CC, et al. Akt determines replicative senescence and oxidative or oncogenic premature senescence and sensitizes cells to oxidative apoptosis. Cancer Cell 2008;14:458-70.

35. Vousden KH, Ryan KM. p53 and metabolism. Nat Rev Cancer 2009;9:691-700.

36. Suzuki S, Tanaka T, Poyurovsky MV, et al. Phosphate-activated glutaminase (GLS2), a p53-inducible regulator of glutamine metabolism and reactive oxygen species. Proc Natl Acad Sci USA 2010;107:7461-6.

37. Budanov AV, Karin M. p53 target genes sestrin1 and sestrin2 connect genotoxic stress and mTOR signaling. Cell 2008;134:451-60.

38. Li T, Kon N, Jiang L, et al. Tumor suppression in the absence of p53-mediated cell-cycle arrest, apoptosis, and senescence. Cell 2012;149:1269-83.

39. Sullivan LB, Chandel NS. Mitochondrial reactive oxygen species and cancer. Cancer Metab 2014;2:17.

40. Chew SH, Toyokuni S. Malignant mesothelioma as an oxidative stress-induced cancer: An update. Free Radic Biol Med 2015;86:166-78.

41. Kinnula VL, Pietarinen-runtti P, Raivio K, et al. Manganese superoxide dismutase in human pleural mesothelioma cell lines. Free Radic Biol Med 1996;21:527-32.

42. Kahlos K, Soini Y, Saily M, et al. Up-regulation of thioredoxin and thioredoxin reductase in human malignant pleural mesothelioma. Int J Cancer 2001;95:198-204.

43. Janssen YM, Marsh JP, Driscoll KE, Borm PJ, Oberdörster G, Mossman BT. Increased expression of manganese-containing superoxide dismutase in rat lungs after inhalation of inflammatory and fibrogenic minerals. Free Radic Biol Med 1994;16:315-22.

44. Kinnula K, Linnainmaa K, Raivio KO, Kinnula VL. Endogenous antioxidant enzymes and glutathione S-transferase in protection of mesothelioma cells against hydrogen peroxide and epirubicin toxicity. Br J Cancer 1998;77:1097-102.

45. Kinnula VL, Lehtonen S, Sormunen R, et al. Overexpression of peroxiredoxins I, II, III, V, and VI in malignant mesothelioma. J Pathol 2002;196:316-23.

46. Kim H, Lee TH, Park ES, et al. Role of peroxiredoxins in regulating intracellular hydrogen peroxide and hydrogen peroxide-induced apoptosis in thyroid cells. J Biol Chem 2000;275:18266-70.

47. Wang YG, Li L, Liu CH, Hong S, Zhang MJ. Peroxiredoxin 3 is resistant to oxidation-induced apoptosis of Hep-3b cells. Clin Transl Oncol 2014;16:561-6.

48. Cox AG, Winterbourn CC, Hampton MB. Mitochondrial peroxiredoxin involvement in antioxidant defence and redox signalling. Biochem J 2009;425:313-25.

49. Stockwell BR, Friedmann Angeli JP, Bayir H, et al. Ferroptosis: a regulated cell death nexus linking metabolism, redox biology, and disease. Cell 2017;171:273-85.

50. Wu J, Minikes AM, Gao M, et al. Publisher correction: intercellular interaction dictates cancer cell ferroptosis via NF2-YAP signalling. Nature 2019;572:E20.

51. Fennell D. Cancer-cell death ironed out. Nature 2019;572:314-5.

52. Zhang Y, Shi J, Liu X, et al. BAP1 links metabolic regulation of ferroptosis to tumour suppression. Nat Cell Biol 2018;20:1181-92.

53. Bononi A, Yang H, Giorgi C, et al. Germline BAP1 mutations induce a Warburg effect. Cell Death Differ 2017;24:1694-704.

54. Hebert L, Bellanger D, Guillas C, et al. Modulating BAP1 expression affects ROS homeostasis, cell motility and mitochondrial function. Oncotarget 2017;8:72513-27.

55. Newick K, Cunniff B, Preston K, et al. Peroxiredoxin 3 is a redox-dependent target of thiostrepton in malignant mesothelioma cells. PLoS One 2012;7:e39404.

56. Cunniff B, Benson K, Stumpff J, et al. Mitochondrial-targeted nitroxides disrupt mitochondrial architecture and inhibit expression of peroxiredoxin 3 and FOXM1 in malignant mesothelioma cells. J Cell Physiol 2013;228:835-45.

57. Kong X, Li L, Li Z, et al. Dysregulated expression of FOXM1 isoforms drives progression of pancreatic cancer. Cancer Res 2013;73:3987-96.

58. Schiavello M, Gazzano E, Bergandi L, et al. Identification of redox-sensitive transcription factors as markers of malignant pleural mesothelioma. Cancers (Basel) 2021;13:1138.

59. Scalcon V, Bindoli A, Rigobello MP. Significance of the mitochondrial thioredoxin reductase in cancer cells: An update on role, targets and inhibitors. Free Radic Biol Med 2018;127:62-79.

60. Lu J, Holmgren A. The thioredoxin antioxidant system. Free Radic Biol Med 2014;66:75-87.

61. Cunniff B, Snider GW, Fredette N, Hondal RJ, Heintz NH. A direct and continuous assay for the determination of thioredoxin reductase activity in cell lysates. Anal Biochem 2013;443:34-40.

62. Schrepfer E, Scorrano L. Mitofusins, from Mitochondria to Metabolism. Mol Cell 2016;61:683-94.

63. Lee JE, Westrate LM, Wu H, Page C, Voeltz GK. Multiple dynamin family members collaborate to drive mitochondrial division. Nature 2016;540:139-43.

64. Eura Y, Ishihara N, Yokota S, Mihara K. Two mitofusin proteins, mammalian homologues of FZO, with distinct functions are both required for mitochondrial fusion. J Biochem 2003;134:333-44.

65. Ježek J, Cooper KF, Strich R. The Impact of mitochondrial fission-stimulated ROS Production on pro-apoptotic chemotherapy. Biology (Basel) 2021;10:33.

66. Ježek J, Cooper KF, Strich R. Reactive Oxygen species and mitochondrial dynamics: the yin and yang of mitochondrial dysfunction and cancer progression. Antioxidants (Basel) 2018;7:13.

67. Han XJ, Yang ZJ, Jiang LP, et al. Mitochondrial dynamics regulates hypoxia-induced migration and antineoplastic activity of cisplatin in breast cancer cells. Int J Oncol 2015;46:691-700.

68. Wan YY, Zhang JF, Yang ZJ, et al. Involvement of Drp1 in hypoxia-induced migration of human glioblastoma U251 cells. Oncol Rep 2014;32:619-26.

69. Latimer HR, Veal EA. Peroxiredoxins in regulation of MAPK signalling pathways; sensors and barriers to signal transduction. Mol Cells 2016;39:40-5.

70. Ko SH, Choi GE, Oh JY, et al. Author correction: succinate promotes stem cell migration through the GPR91-dependent regulation of DRP1-mediated mitochondrial fission. Sci Rep 2018;8:13326.

71. Aravamudan B, Kiel A, Freeman M, et al. Cigarette smoke-induced mitochondrial fragmentation and dysfunction in human airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2014;306:L840-54.

72. Jung JU, Ravi S, Lee DW, et al. NIK/MAP3K14 regulates mitochondrial dynamics and trafficking to promote cell invasion. Curr Biol 2016;26:3288-302.

73. Liu TL, Upadhyayula S, Milkie DE, et al. Observing the cell in its native state: Imaging subcellular dynamics in multicellular organisms. Science 2018;360:eaaq1392.

74. Iwahori K, Serada S, Fujimoto M, et al. SOCS-1 gene delivery cooperates with cisplatin plus pemetrexed to exhibit preclinical antitumor activity against malignant pleural mesothelioma. Int J Cancer 2013;132:459-71.

75. Nishikawa S, Tanaka A, Matsuda A, et al. A molecular targeting against nuclear factor-κB, as a chemotherapeutic approach for human malignant mesothelioma. Cancer Med 2014;3:416-25.

76. Goparaju CM, Blasberg JD, Volinia S, et al. Onconase mediated NFKβ downregulation in malignant pleural mesothelioma. Oncogene 2011;30:2767-77.

77. Lennon FE, Cianci GC, Kanteti R, et al. Unique fractal evaluation and therapeutic implications of mitochondrial morphology in malignant mesothelioma. Sci Rep 2016;6:24578.

78. Lima AR, Santos L, Correia M, et al. Dynamin-related protein 1 at the crossroads of cancer. Genes (Basel) 2018;9:115.

79. Qian W, Choi S, Gibson GA, Watkins SC, Bakkenist CJ, Van Houten B. Mitochondrial hyperfusion induced by loss of the fission protein Drp1 causes ATM-dependent G2/M arrest and aneuploidy through DNA replication stress. J Cell Sci 2012;125:5745-57.

80. Wang J, Mirzapoiazova T, Carol Tan YH, et al. Inhibiting crosstalk between MET signaling and mitochondrial dynamics and morphology: a novel therapeutic approach for lung cancer and mesothelioma. Cancer Biol Ther 2018;19:1023-32.

81. Grosso S, Marini A, Gyuraszova K, et al. The pathogenesis of mesothelioma is driven by a dysregulated translatome. Nat Commun 2021;12:4920.

82. Omenn GS, Goodman GE, Thornquist MD, et al. Effects of a combination of beta carotene and vitamin A on lung cancer and cardiovascular disease. N Engl J Med 1996;334:1150-5.

83. The effect of vitamin E and beta carotene on the incidence of lung cancer and other cancers in male smokers. N Engl J Med 1994;330:1029-35.

84. Tai DJ, Jin WS, Wu CS, et al. Changes in intracellular redox status influence multidrug resistance in gastric adenocarcinoma cells. Exp Ther Med 2012;4:291-6.

85. Hwang IT, Chung YM, Kim JJ, et al. Drug resistance to 5-FU linked to reactive oxygen species modulator 1. Biochem Biophys Res Commun 2007;359:304-10.

86. Chen Q, Espey MG, Sun AY, et al. Ascorbate in pharmacologic concentrations selectively generates ascorbate radical and hydrogen peroxide in extracellular fluid in vivo. Proc Natl Acad Sci USA 2007;104:8749-54.

87. Alexander MS, Wilkes JG, Schroeder SR, et al. Pharmacologic ascorbate reduces radiation-induced normal tissue toxicity and enhances tumor radiosensitization in pancreatic cancer. Cancer Res 2018;78:6838-51.

88. Mehdi Z, Petronek MS, Stolwijk JM, et al. Utilization of pharmacological ascorbate to enhance hydrogen peroxide-mediated radiosensitivity in cancer therapy. Int J Mol Sci 2021;22:10880.

89. Beddowes E, Spicer J, Chan PY, et al. Phase 1 dose-escalation study of pegylated arginine deiminase, cisplatin, and pemetrexed in patients with argininosuccinate synthetase 1-deficient thoracic cancers. J Clin Oncol 2017;35:1778-85.

90. Szlosarek PW, Luong P, Phillips MM, et al. Metabolic response to pegylated arginine deiminase in mesothelioma with promoter methylation of argininosuccinate synthetase. J Clin Oncol 2013;31:e111-3.

91. Kung HJ, Changou CA, Li CF, Ann DK. Chromatophagy: autophagy goes nuclear and captures broken chromatin during arginine-starvation. Autophagy 2015;11:419-21.

92. Cheng CT, Qi Y, Wang YC, et al. Arginine starvation kills tumor cells through aspartate exhaustion and mitochondrial dysfunction. Commun Biol 2018;1:178.

93. Szlosarek PW, Klabatsa A, Pallaska A, et al. In vivo loss of expression of argininosuccinate synthetase in malignant pleural mesothelioma is a biomarker for susceptibility to arginine depletion. Clin Cancer Res 2006;12:7126-31.

94. Kremer JC, Prudner BC, Lange SES, et al. Arginine deprivation inhibits the warburg effect and upregulates glutamine anaplerosis and serine biosynthesis in ASS1-deficient cancers. Cell Rep 2017;18:991-1004.

95. Vogelzang NJ, Rusthoven JJ, Symanowski J, et al. Phase III study of pemetrexed in combination with cisplatin versus cisplatin alone in patients with malignant pleural mesothelioma. J Clin Oncol 2003;21:2636-44.

96. Sasada T, Nakamura H, Ueda S, et al. Possible involvement of thioredoxin reductase as well as thioredoxin in cellular sensitivity to cis-diamminedichloroplatinum (II). Free Radic Biol Med 1999;27:504-14.

97. Ishikawa T, Ali-osman F. Glutathione-associated cis-diamminedichloroplatinum(II) metabolism and ATP-dependent efflux from leukemia cells. Molecular characterization of glutathione-platinum complex and its biological significance. J Biol Chem 1993;268:20116-25.

98. Arnér ES, Nakamura H, Sasada T, Yodoi J, Holmgren A, Spyrou G. Analysis of the inhibition of mammalian thioredoxin, thioredoxin reductase, and glutaredoxin by cis -diamminedichloroplatinum (II) and its major metabolite, the glutathione-platinum complex. Free Radic Biol Med 2001;31:1170-8.

99. Zhang X, Zheng Y, Fried LE, et al. Disruption of the mitochondrial thioredoxin system as a cell death mechanism of cationic triphenylmethanes. Free Radic Biol Med 2011;50:811-20.

100. Corsello SM, Nagari RT, Spangler RD, et al. Discovering the anti-cancer potential of non-oncology drugs by systematic viability profiling. Nat Cancer 2020;1:235-48.

101. Hegde NS, Sanders DA, Rodriguez R, Balasubramanian S. The transcription factor FOXM1 is a cellular target of the natural product thiostrepton. Nat Chem 2011;3:725-31.

102. Bhat UG, Halasi M, Gartel AL. FoxM1 is a general target for proteasome inhibitors. PLoS One 2009;4:e6593.

103. Bird KE, Xander C, Murcia S, et al. Thiopeptides Induce Proteasome-Independent Activation of Cellular Mitophagy. ACS Chem Biol 2020;15:2164-74.

Cite This Article

Export citation file: BibTeX | RIS

OAE Style

Cote A, Messier T, Cunniff B. Reactive oxygen species in the progression and treatment of malignant mesothelioma. J Cancer Metastasis Treat 2022;8:36. http://dx.doi.org/10.20517/2394-4722.2022.41

AMA Style

Cote A, Messier T, Cunniff B. Reactive oxygen species in the progression and treatment of malignant mesothelioma. Journal of Cancer Metastasis and Treatment. 2022; 8: 36. http://dx.doi.org/10.20517/2394-4722.2022.41

Chicago/Turabian Style

Cote, Ava, Terri Messier, Brian Cunniff. 2022. "Reactive oxygen species in the progression and treatment of malignant mesothelioma" Journal of Cancer Metastasis and Treatment. 8: 36. http://dx.doi.org/10.20517/2394-4722.2022.41

ACS Style

Cote, A.; Messier T.; Cunniff B. Reactive oxygen species in the progression and treatment of malignant mesothelioma. J. Cancer. Metastasis. Treat. 2022, 8, 36. http://dx.doi.org/10.20517/2394-4722.2022.41

About This Article

Special Issue

This article belongs to the Special Issue Research Advances on Mesothelioma
© The Author(s) 2022. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Data & Comments

Data

Views
911
Downloads
594
Citations
0
Comments
0
2

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Cite This Article 14 clicks
Like This Article 2 likes
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Journal of Cancer Metastasis and Treatment
ISSN 2454-2857 (Online) 2394-4722 (Print)

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/