Next Article in Journal
Toll-like Receptor 4 Inflammatory Perspective on Doxorubicin-Induced Cardiotoxicity
Previous Article in Journal
Efficiency of the Enzymatic Conversion of Flavone Glycosides Isolated from Carrot Leaves and Anti-Inflammatory Effects of Enzyme-Treated Carrot Leaves
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Cancer Prevention and Therapy by Targeting Oxidative Stress Pathways

1
Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura 281406, India
2
Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy
3
Department of Biochemistry, Faculty of Pharmacy, Ege University, Bornova, Izmir 35100, Turkey
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(11), 4293; https://doi.org/10.3390/molecules28114293
Submission received: 25 April 2023 / Revised: 28 April 2023 / Accepted: 22 May 2023 / Published: 24 May 2023

1. Introduction

Oxidative stress arises from the inadequate production of reactive oxygen species (ROS) which couldn’t be neutralized by antioxidant defense. Cells maintain this balance of oxidants and antioxidants by different biochemical, metabolic and genetic mechanisms and in case of imbalance, several pathophysiological consequences can occur [1]. The cross-talk between cancer and oxidative stress is contradictory, either promoting tumorigenesis and cancer cell proliferation or stimulating apoptosis. The proliferation of cancer cells is accompanied by ROS overproduction; however, tumor cells balance this ROS threshold via an array of antioxidant systems to avoid ferroptosis, apoptosis or senescence [2]. On one hand, ROS scavenging by antioxidants could increase apoptosis and thus, deprives cancer cells of energy in early tumorigenesis. On the other hand, ROS production could selectively kill tumor cells by apoptosis, autophagy, necrosis and ferroptosis [3]. Therefore, the fine tuning between the ROS production and clearance is highly crucial. Among the key antioxidant systems, glutathione and thioredoxin pathways and Nrf2/Keap1 signaling systems have been reviewed in cancer [3]. In this context, peroxidation of the polyunsaturated fatty acids in the lipid bilayer of the cell membranes also gives rise to 4-hydroxynonenal (4-HNE), also known as the second messenger of ROS, which has been shown to exhibit a crucial role in cancer [4]. 4-HNE reportedly promotes cell proliferation, however, in some conditions it can stimulate apoptosis or necrosis of specific cancer cells [5]. 4-HNE exerts its effects by either binding to the arginine, histidine and cysteine residues of proteins, or with DNA leading to their modifications [6,7].
It is assumed that some non-cancer cells also associate with tumors, such as cancer-associated fibroblasts (CAFs), which cooperate to maintain the tumor homeostasis and stimulate the proliferation, progression and invasion of tumor cells. CAFs and ROS have a bilateral relationship, ROS, particularly H2O2, attack fibroblasts and convert them into active CAFs by upregulation of the expression of HIF1α [8]. On the other hand, an increase in ROS-generated CAFs promotes cancer growth and invasiveness, which in turn leads to the upregulation in the antioxidant gene expressions [9]. Accumulating evidence suggests that the oncogenes raise ROS production. Thus, RAS and STAT3 genes activates NOX2 and NOX4 gene by altering mitochondrial membrane potential [10,11]. Similarly, BCL-2 and MYC are also responsible for ROS production by altering mitochondrial function [12,13]. Also, the down-regulation of tumor suppressor mediated gene expressions of antioxidant genes such as SOD2, GPX1, SESN1 and SESN2, also promote the generation of ROS in tumor cells [14]. Moreover, tumor cells are also stimulated by the TNFα released from the immune cells [15]. For instance, breast cancer cells activate Ca2+ signaling via redox sensitive TRPA1 channels, which in turn activates PI3K-PKB/Akt pathways and oxidative stress tolerance [16]. On the other hand, TRPA1 gene expression is also controlled by Nrf2 pathway, suggesting the possible link between Ca2+ signaling and oxidative stress [16].
Nrf2 (NF-E2-related factor 2) is a transcription factor, which belongs to the group of Cap’n’collar (CNC) family of bZIP transcription factors [17]. Nrf2 along with its negative regulator, the E3 ligase adaptor Kelch-like ECH-associated protein 1 (Keap1), cooperate to regulate the intracellular redox homeostasis in cancer and other diseases [17]. In normal circumstances, Keap1 binds with Neh2 domain of Nrf2 via ETGE and DLG motifs, leading to the localization of Nrf2 in cytoplasm [18]. However, with oxidative stress, Nrf2 doesn’t interact with ubiquitin-conjugating system due to a conformational change in the E3-ligase complex [18]. This results the Nrf2 release from the complex, leading to the translocation of the Nrf2 into the nucleus, heterodimer formation with sMaf protein and ultimately leads to the ARE activation, which further regulates cell protective mechanisms and antioxidant proteins expressions [19]. Nrf2 pathway promotes the expression of genes including TXN, G6PD, GSTA2, NQO1, and HMOX1 which are related to NADH regeneration and redox detoxification [20]. This special issue shares insights on the therapeutic strategies by targeting redox signaling pathways in cancer. The study by González-Montero et al. [21] documents the role of ascorbate as a potent therapeutic agent in cancer. The authors discuss the paradoxical effects of ascorbate, which induces oxidative stress at high concentration by interacting with iron. Also, it was suggested that ascorbate could also act as an adjuvant in different cancer therapies by inducing apoptosis and ferroptosis.
We consider this Special issue to bring together a wide range of review and research articles contributing to an understanding of the cancer therapies based on redox state.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hayes, J.D.; Dinkova-Kostova, A.T.; Tew, K.D. Oxidative Stress in Cancer. Cancer Cell 2020, 38, 167–197. [Google Scholar] [CrossRef] [PubMed]
  2. Dodson, M.; Castro-Portuguez, R.; Zhang, D.D. NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biol. 2019, 23, 101107. [Google Scholar] [CrossRef] [PubMed]
  3. Ulker, O.C.; Panieri, E.; Suzen, S.; Jaganjac, M.; Zarkovic, N.; Saso, L. Short overview on the relevance of microRNA–reactive oxygen species (ROS) interactions and lipid peroxidation for modulation of oxidative stress-mediated signalling pathways in cancer treatment. J. Pharm. Pharmacol. 2022, 74, 503–515. [Google Scholar] [CrossRef] [PubMed]
  4. Milkovic, L.; Gasparovic, A.C.; Zarkovic, N. Overview on major lipid peroxidation bioactive factor 4-hydroxynonenal as pluripotent growth regulating factor. Free Radic. Res. 2015, 49, 850–860. [Google Scholar] [CrossRef]
  5. Zarkovic, K.; Jakovcevic, A.; Zarkovic, N. Contribution of the HNE-immunohistochemistry to modern pathological concepts of major human diseases. Free Radic. Biol. Med. 2017, 111, 110–126. [Google Scholar] [CrossRef]
  6. Vistoli, G.; De Maddis, D.; Cipak, A.; Zarkovic, N.; Carini, M.; Aldini, G. Advanced glycoxidation and lipoxidation end products (AGEs and ALEs): An overview of their mechanisms of formation. Free Radic. Res. 2013, 47, 3–27. [Google Scholar] [CrossRef]
  7. Gentile, F.; Arcaro, A.; Pizzimenti, S.; Daga, M.; Cetrangolo, G.P.; Dianzani, C.; Lepore, A.; Graf, M.; Ames, P.R.J.; Barrera, G. DNA damage by lipid peroxidation products: Implications in cancer, inflammation and autoimmunity. AIMS Genet. 2017, 4, 103–137. [Google Scholar] [CrossRef]
  8. Chan, J.S.; Tan, M.J.; Sng, M.K.; Teo, Z.; Phua, T.; Choo, C.C.; Li, L.; Zhu, P.; Tan, N.S. Cancer-associated fibroblasts enact field cancerization by promoting extratumoral oxidative stress. Cell Death Dis. 2017, 8, e2562. [Google Scholar] [CrossRef]
  9. Holmstrom, K.M.; Finkel, T. Cellular mechanisms and physiological consequences of redox-dependent signaling. Nat. Rev. Mol. Cell Biol. 2014, 15, 411–421. [Google Scholar] [CrossRef]
  10. Chong, S.J.F.; Lai, J.X.H.; Eu, J.Q.; Bellot, G.L.; Pervaiz, S. Reactive oxygen species and oncoprotein signaling—A dangerous liaison. Antioxid. Redox Signal. 2018, 29, 1553–1588. [Google Scholar] [CrossRef]
  11. Igelmann, S.; Neubauer, H.A.; Ferbeyre, G. STAT3 and STAT5 activation in solid cancers. Cancers 2019, 11, 1428. [Google Scholar] [CrossRef]
  12. Satoh, K.; Yachida, S.; Sugimoto, M.; Oshima, M.; Nakagawa, T.; Akamoto, S.; Tabata, S.; Saitoh, K.; Kato, K.; Sato, S.; et al. Global metabolic reprogramming of colorectal cancer occurs at adenoma stage and is induced by MYC. Proc. Natl. Acad. Sci. USA 2017, 114, E7697–E7706. [Google Scholar] [CrossRef] [PubMed]
  13. Marcar, L.; Bardhan, K.; Gheorghiu, L.; Dinkelborg, P.; Pfäffle, H.; Liu, Q.; Wang, M.; Piotrowska, Z.; Sequist, L.V.; Borgmann, K.; et al. Acquired resistance of EGFR-mutated lung cancer to tyrosine kinase inhibitor treatment promotes PARP inhibitor sensitivity. Cell Rep. 2019, 27, 3422–3432.e4. [Google Scholar] [CrossRef]
  14. Gorrini, C.; Harris, I.S.; Mak, T.W. Modulation of oxidative stress as an anticancer strategy. Nat. Rev. Drug Discov. 2013, 12, 931–947. [Google Scholar] [CrossRef] [PubMed]
  15. He, C.; Danes, J.M.; Hart, P.C.; Zhu, Y.; Huang, Y.; de Abreu, A.L.; O’Brien, J.; Mathison, A.J.; Tang, B.; Frasor, J.M.; et al. SOD2 acetylation on lysine 68 promotes stem cell reprogramming in breast cancer. Proc. Natl. Acad. Sci. USA 2019, 116, 23534–23541. [Google Scholar] [CrossRef] [PubMed]
  16. Takahashi, N.; Chen, H.Y.; Harris, I.S.; Stover, D.G.; Selfors, L.M.; Bronson, R.T.; Deraedt, T.; Cichowski, K.; Welm, A.L.; Mori, Y.; et al. Cancer Cells Co-opt the neuronal redox-sensing channel TRPA1 to promote oxidative-stress tolerance. Cancer Cell 2018, 33, 985–1003.e7. [Google Scholar] [CrossRef]
  17. Saha, S.; Buttari, B.; Panieri, E.; Profumo, E.; Saso, L. An overview of Nrf2 signaling pathway and its role in inflammation. Molecules 2020, 25, 5474. [Google Scholar] [CrossRef]
  18. Kansanen, E.; Kuosmanen, S.M.; Leinonen, H.; Levonen, A. The Keap1-Nrf2pathway: Mechanisms of activation and dysregulation in cancer. Redox Biol. 2013, 1, 45–49. [Google Scholar] [CrossRef]
  19. Baburina, Y.; Krestinin, R.; Odinokova, I.; Fadeeva, I.; Sotnikova, L.; Krestinina, O. The identification of prohibitin in the rat heart mitochondria in heart failure. Biomedicines 2021, 9, 1793. [Google Scholar] [CrossRef]
  20. Bendavit, G.; Aboulkassim, T.; Hilmi, K.; Shah, S.; Batist, G. Nrf2 transcription factor can directly regulate mTOR: Linking cytoprotective gene expression to a major metabolic regulator that generates redox activity. J. Biol. Chem. 2016, 291, 25476–25488. [Google Scholar] [CrossRef]
  21. González-Montero, J.; Chichiarelli, S.; Eufemi, M.; Altieri, F.; Saso, L.; Rodrigo, R. Ascorbate as a bioactive compound in cancer therapy: The old classic strikes back. Molecules 2022, 27, 3818. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Saha, S.; Saso, L.; Armagan, G. Cancer Prevention and Therapy by Targeting Oxidative Stress Pathways. Molecules 2023, 28, 4293. https://doi.org/10.3390/molecules28114293

AMA Style

Saha S, Saso L, Armagan G. Cancer Prevention and Therapy by Targeting Oxidative Stress Pathways. Molecules. 2023; 28(11):4293. https://doi.org/10.3390/molecules28114293

Chicago/Turabian Style

Saha, Sarmistha, Luciano Saso, and Guliz Armagan. 2023. "Cancer Prevention and Therapy by Targeting Oxidative Stress Pathways" Molecules 28, no. 11: 4293. https://doi.org/10.3390/molecules28114293

Article Metrics

Back to TopTop