Oxidative Stress: Signaling Pathways, Biological Functions, and Disease

Sixuan Liu , Jiachen Liu , Yinhuai Wang , Fei Deng , Zebin Deng

MedComm ›› 2025, Vol. 6 ›› Issue (7) : e70268

PDF
MedComm ›› 2025, Vol. 6 ›› Issue (7) : e70268 DOI: 10.1002/mco2.70268
REVIEW

Oxidative Stress: Signaling Pathways, Biological Functions, and Disease

Author information +
History +
PDF

Abstract

The dysregulated accumulation of reactive oxygen species (ROS) and reactive nitrogen species disrupts redox homeostasis, triggering oxidative stress (OS) and driving pathophysiological changes across multiple organ systems. OS modulates critical signaling pathways, induces inflammation, impairs mitochondrial function, alters metabolic homeostasis, and dysregulates autophagy, contributing to disease progression. While prior research has largely focused on OS within single-organ diseases (e.g., neurodegenerative, cardiovascular, and oncological disorders), the systemic role of OS in pan-organ diseases and interorgan communication remains insufficiently explored. This review integrates multidisciplinary evidence to elucidate the biological functions of OS in cellular signaling, homeostasis, and cross-organ crosstalk. It systematically dissects OS-driven molecular mechanisms and pathophysiological networks across 10 major organ systems, including the nervous, cardiovascular, oncological, hepatic, and renal systems. Furthermore, it critically examines OS-related therapeutic targets, including antioxidant and ROS-generating enzymes, and explores synergistic redox-based therapeutic strategies. By moving beyond traditional single-organ paradigms, this review constructs a holistic framework to decode the systemic impact of OS, offering novel insights into disease mechanisms and therapeutic innovations. Ultimately, it lays the foundation for precision medicine approaches aimed at mitigating OS-driven diseases and improving multiorgan health outcomes.

Keywords

inflammation / mitochondrial dysfunction / oxidative stress / reactive oxygen species / redox homeostasis

Cite this article

Download citation ▾
Sixuan Liu, Jiachen Liu, Yinhuai Wang, Fei Deng, Zebin Deng. Oxidative Stress: Signaling Pathways, Biological Functions, and Disease. MedComm, 2025, 6(7): e70268 DOI:10.1002/mco2.70268

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

D. B. Zorov, M. Juhaszova, and S. J. Sollott, “Mitochondrial Reactive Oxygen Species (Ros) and Ros-Induced Ros Release,” Physiological Reviews 94, no. 3 (2014): 909-950.

[2]

L. Milkovic, A. Cipak Gasparovic, M. Cindric, P. Mouthuy, and N. Zarkovic, “Short Overview of ROS as Cell Function Regulators and Their Implications in Therapy Concepts,” Cells 8, no. 8 (2019): 793.

[3]

H. Sies, R. J. Mailloux, and U. Jakob, “Fundamentals of Redox Regulation in Biology,” Nature Reviews Molecular Cell Biology 25, no. 9 (2024): 701-719.

[4]

T. Hussain, B. Tan, Y. Yin, et al., “Oxidative Stress and Inflammation: What Polyphenols Can Do for Us?” Oxidative Medicine and Cellular Longevity 2016 (2016): 7432797.

[5]

J. Papaconstantinou, “The Role of Signaling Pathways of Inflammation and Oxidative Stress in Development of Senescence and Aging Phenotypes in Cardiovascular Disease,” Cells 8, no. 11 (2019): 1383.

[6]

J. N. Peoples, A. Saraf, N. Ghazal, T. T. Pham, and J. Q. Kwong, “Mitochondrial Dysfunction and Oxidative Stress in Heart Disease,” Experimental & Molecular Medicine 51, no. 12 (2019): 1-13.

[7]

D. B. Zorov, M. Juhaszova, and S. J. Sollott, “Mitochondrial Reactive Oxygen Species (ROS) and ROS-induced ROS Release,” Physiological Reviews 94, no. 3 (2014): 909-950.

[8]

Y. Liu, Y. Huang, C. Xu, et al., “Mitochondrial Dysfunction and Therapeutic Perspectives in Cardiovascular Diseases,” International Journal of Molecular Sciences 23, no. 24 (2022): 16053.

[9]

Y. Mei, M. D. Thompson, R. A. Cohen, and X. Tong, “Autophagy and Oxidative Stress in Cardiovascular Diseases,” Biochimica Et Biophysica Acta 1852, no. 2 (2015): 243-251.

[10]

Y. Gu, J. Han, C. Jiang, and Y. Zhang, “Biomarkers, Oxidative Stress and Autophagy in Skin Aging,” Ageing Research Reviews 59 (2020): 101036.

[11]

W. Ornatowski, Q. Lu, M. Yegambaram, et al., “Complex Interplay between Autophagy and Oxidative Stress in the Development of Pulmonary Disease,” Redox Biology 36 (2020): 101679.

[12]

D. A. Averill-Bates, “The Antioxidant Glutathione,” Vitamins and Hormones 121 (2023): 109-141.

[13]

G. Bresciani, I. B. M. Da Cruz, and J. González-Gallego, “Manganese Superoxide Dismutase and Oxidative Stress Modulation,” Advances in Clinical Chemistry 68 (2015): 87-130.

[14]

L. Chen, Y. Liu, Y. Zhang, et al., “Superoxide Dismutase Ameliorates Oxidative Stress and Regulates Liver Transcriptomics to Provide Therapeutic Benefits in Hepatic Inflammation,” PeerJ 11 (2023): e15829.

[15]

E. Niki, “Oxidative Stress and Aging,” Internal Medicine 39, no. 4 (2000): 324-326.

[16]

R. G. Cutler, “Oxidative Stress and Aging: Catalase Is a Longevity Determinant Enzyme,” Rejuvenation Research 8, no. 3 (2005): 138-140.

[17]

L. Gebicka, J. Krych-Madej, “The Role of Catalases in the Prevention/Promotion of Oxidative Stress,” Journal of Inorganic Biochemistry 197 (2019): 110699.

[18]

L. Ma, F. Wu, Q. Shao, et al., “Baicalin Alleviates Oxidative Stress and Inflammation in Diabetic Nephropathy via Nrf2 and MAPK Signaling Pathway,” Drug Design, Development and Therapy 15 (2021): 3207-3221.

[19]

T. Behl, T. Upadhyay, S. Singh, et al., “Polyphenols Targeting MAPK Mediated Oxidative Stress and Inflammation in Rheumatoid Arthritis,” Molecules (Basel, Switzerland) 26, no. 21 (2021): 6570.

[20]

F. Xu, J. Xu, X. Xiong, and Y. Deng, “Salidroside Inhibits MAPK, NF-κB, and STAT3 Pathways in Psoriasis-associated Oxidative Stress via SIRT1 Activation,” Redox Report: Communications in Free Radical Research 24, no. 1 (2019): 70-74.

[21]

B. M. Hybertson, B. Gao, S. K. Bose, and J. M. McCord, “Oxidative Stress in Health and Disease: the Therapeutic Potential of Nrf2 Activation,” Molecular Aspects of Medicine 32, no. 4-6 (2011): 234-246.

[22]

Q. Ma, “Role of nrf2 in Oxidative Stress and Toxicity,” Annual Review of Pharmacology and Toxicology 53 (2013): 401-426.

[23]

E. O. O. Olufunmilayo, M. B. B. Gerke-Duncan, and R. M. D. Holsinger, “Oxidative Stress and Antioxidants in Neurodegenerative Disorders,” Antioxidants (Basel) 12, no. 2 (2023): 517.

[24]

Q. Yan, S. Liu, Y. Sun, et al., “Targeting Oxidative Stress as a Preventive and Therapeutic Approach for Cardiovascular Disease,” Journal of translational medicine 21, no. 1 (2023): 519.

[25]

C. Gorrini, I. S. Harris, and T. W. Mak, “Modulation of Oxidative Stress as an Anticancer Strategy,” Nat Rev Drug Discovery 12, no. 12 (2013): 931-947.

[26]

D. Li, Q. Yu, R. Wu, et al., “Interactions between Oxidative Stress and Senescence in Cancer: Mechanisms, Therapeutic Implications, and Future Perspectives,” Redox Biology 73 (2024): 103208.

[27]

L. Sun, X. Wang, J. Saredy, et al., “Innate-adaptive Immunity Interplay and Redox Regulation in Immune Response,” Redox Biology 37 (2020): 101759.

[28]

O. A. Castaneda, S. C. Lee, C. T. Ho, and T. C. Huang, “Macrophages in Oxidative Stress and Models to Evaluate the Antioxidant Function of Dietary Natural Compounds,” Journal of Food and Drug Analysis 25, no. 1 (2017): 111-118.

[29]

A. M. Fresneda, Z. McLaren, and H. L. Wright, “Neutrophils in the Pathogenesis of Rheumatoid Arthritis and Systemic Lupus Erythematosus: Same Foe Different M.O,” Frontiers in Immunology 12 (2021): 649693.

[30]

C. Li, C. Deng, S. Wang, et al., “A Novel Role for the ROS-ATM-Chk2 Axis Mediated Metabolic and Cell Cycle Reprogramming in the M1 Macrophage Polarization,” Redox Biology 70 (2024): 103059.

[31]

I. Cotzomi-Ortega, O. Nieto-Yañez, I. Juárez-Avelar, et al., “Autophagy Inhibition in Breast Cancer Cells Induces ROS-mediated MIF Expression and M1 Macrophage Polarization,” Cell Signalling 86 (2021): 110075.

[32]

C. Liu, F. Hu, G. Jiao, et al., “Dental Pulp Stem Cell-derived Exosomes Suppress M1 Macrophage Polarization through the ROS-MAPK-NFκB P65 Signaling Pathway after Spinal Cord Injury,” Journal of Nanobiotechnology 20, no. 1 (2022): 65.

[33]

M. Liu, X. Liu, Y. Wang, et al., “Intrinsic ROS Drive Hair Follicle Cycle Progression by Modulating DNA Damage and Repair and Subsequently Hair Follicle Apoptosis and Macrophage Polarization,” Oxidative Medicine and Cellular Longevity 2022 (2022): 8279269.

[34]

J. Tschopp, K. Schroder, “NLRP3 inflammasome Activation: the Convergence of Multiple Signalling Pathways on ROS Production?” Nature Reviews Immunology 10, no. 3 (2010): 210-215.

[35]

H. Sun, Z. Sun, X. Xu, et al., “Blocking TRPV4 Ameliorates Osteoarthritis by Inhibiting M1 Macrophage Polarization via the ROS/NLRP3 Signaling Pathway,” Antioxidants (Basel) 11, no. 12 (2022): 2315.

[36]

J. Wang, Y. Yin, Q. Zhang, et al., “HgCl2 exposure Mediates Pyroptosis of HD11 Cells and Promotes M1 Polarization and the Release of Inflammatory Factors through ROS/Nrf2/NLRP3,” Ecotoxicol. Environ. Saf. 269 (2024): 115779.

[37]

J. Liu, Y. Wei, W. Jia, et al., “Chenodeoxycholic Acid Suppresses AML Progression through Promoting Lipid Peroxidation via ROS/p38 MAPK/DGAT1 Pathway and Inhibiting M2 Macrophage Polarization,” Redox Biology 56 (2022): 102452.

[38]

H. Jeong, H. Yoon, Y. Lee, et al., “SOCS3 Attenuates Dexamethasone-Induced M2 Polarization by Down-Regulation of GILZ via ROS- and p38 MAPK-Dependent Pathways,” Immune Network 22, no. 4 (2022): e33.

[39]

M. Tsai, Y. Tsai, Y. Lin, et al., “IL-25 Induced ROS-Mediated M2 Macrophage Polarization via AMPK-Associated Mitophagy,” International Journal of Molecular Sciences 23, no. 1 (2021): 3.

[40]

E. Vergadi, E. Ieronymaki, K. Lyroni, K. Vaporidi, and C. Tsatsanis, “Akt Signaling Pathway in Macrophage Activation and M1/M2 Polarization,” Journal of Immunology 198, no. 3 (2017): 1006-1014.

[41]

W. Sha, B. Zhao, H. Wei, et al., “Astragalus Polysaccharide Ameliorates Vascular Endothelial Dysfunction by Stimulating Macrophage M2 Polarization via Potentiating Nrf2/HO-1 Signaling Pathway,” Phytomedicine 112 (2023): 154667.

[42]

D. C. Thomas, “The Phagocyte respiratory Burst: Historical Perspectives and Recent Advances,” Immunology Letters 192 (2017): 88-96.

[43]

M. B. Hampton, A. J. Kettle, and C. C. Winterbourn, “Inside the Neutrophil Phagosome: Oxidants, Myeloperoxidase, and Bacterial Killing,” Blood 92, no. 9 (1998): 3007-3017.

[44]

J. El-Benna, M. Hurtado-Nedelec, V. Marzaioli, et al., “Priming of the Neutrophil respiratory Burst: Role in Host Defense and Inflammation,” Immunological Reviews 273, no. 1 (2016): 180-193.

[45]

T. A. Fuchs, U. Abed, C. Goosmann, et al., “Novel Cell Death Program Leads to Neutrophil Extracellular Traps,” Journal of Cell Biology 176, no. 2 (2007): 231-241.

[46]

T. Kirchner, S. Moeller, M. Klinger, et al., “The Impact of Various Reactive Oxygen Species on the Formation of Neutrophil Extracellular Traps,” Mediators of Inflammation 2012 (2012): 849136.

[47]

A. Hakkim, T. A. Fuchs, N. E. Martinez, et al., “Activation of the Raf-MEK-ERK Pathway Is Required for Neutrophil Extracellular Trap Formation,” Nature Chemical Biology 7, no. 2 (2011): 75-77.

[48]

D. N. Douda, M. A. Khan, H. Grasemann, and N. Palaniyar, “SK3 channel and Mitochondrial ROS Mediate NADPH Oxidase-independent NETosis Induced by Calcium Influx,” PNAS 112, no. 9 (2015): 2817-2822.

[49]

X. Zhou, L. Yang, X. Fan, et al., “Neutrophil Chemotaxis and NETosis in Murine Chronic Liver Injury via Cannabinoid Receptor 1/Gα(i/o)/ ROS/ p38 MAPK Signaling Pathway,” Cells 9, no. 2 (2020): 373.

[50]

M. A. Khan, A. Farahvash, D. N. Douda, et al., “JNK Activation Turns on LPS- and Gram-Negative Bacteria-Induced NADPH Oxidase-Dependent Suicidal NETosis,” Scientific Reports 7, no. 1 (2017): 3409.

[51]

N. V. Vorobjeva, B. V. Chernyak, “NETosis: Molecular Mechanisms, Role in Physiology and Pathology,” Biochemistry (Moscow) 85, no. 10 (2020): 1178-1190.

[52]

S. Zheng, Y. Chen, Z. Wang, et al., “Combination of Matrine and Tacrolimus Alleviates Acute Rejection in Murine Heart Transplantation by Inhibiting DCs Maturation through ROS/ERK/NF-κB Pathway,” International Immunopharmacology 101, no. Pt B (2021): 108218.

[53]

L. M. Paardekooper, W. Vos, and G. van den Bogaart, “Oxygen in the Tumor Microenvironment: Effects on Dendritic Cell Function,” Oncotarget 10, no. 8 (2019): 883-896.

[54]

Q. Liu, J. Zhang, X. Liu, and J. Gao, “Role of Growth Hormone in Maturation and Activation of Dendritic Cells via miR-200a and the Keap1/Nrf2 Pathway,” Cell Proliferation 48, no. 5 (2015): 573-581.

[55]

H. X. A. Yeang, J. M. Hamdam, L. M. A. Al-Huseini, et al., “Loss of Transcription Factor Nuclear Factor-erythroid 2 (NF-E2) p45-related Factor-2 (Nrf2) Leads to Dysregulation of Immune Functions, Redox Homeostasis, and Intracellular Signaling in Dendritic Cells,” Journal of Biological Chemistry 287, no. 13 (2012): 10556-10564.

[56]

J. Ma, K. Wei, H. Zhang, et al., “Mechanisms by Which Dendritic Cells Present Tumor Microparticle Antigens to CD8+ T Cells,” Cancer Immunology Research 6, no. 9 (2018): 1057-1068.

[57]

M. Oberkampf, C. Guillerey, J. Mouriès, et al., “Mitochondrial Reactive Oxygen Species Regulate the Induction of CD8(+) T Cells by Plasmacytoid Dendritic Cells,” Nature Communications 9, no. 1 (2018): 2241.

[58]

H. Peng, J. Lucavs, D. Ballard, et al., “Metabolic Reprogramming and Reactive Oxygen Species in T Cell Immunity,” Frontiers in Immunology 12 (2021): 652687.

[59]

J. D. Graves, A. Craxton, and E. A. Clark, “Modulation and Function of Caspase Pathways in B Lymphocytes,” Immunological Reviews 197 (2004): 129-146.

[60]

F. Macian, “NFAT Proteins: Key Regulators of T-cell Development and Function,” Nature Reviews Immunology 5, no. 6 (2005): 472-484.

[61]

M. M. Kaminski, S. W. Sauer, C. Klemke, et al., “Mitochondrial Reactive Oxygen Species Control T Cell Activation by Regulating IL-2 and IL-4 Expression: Mechanism of Ciprofloxacin-mediated Immunosuppression,” Journal of Immunology 184, no. 9 (2010): 4827-4841.

[62]

M. M. Kamiński, S. W. Sauer, M. Kamiński, et al., “T Cell Activation Is Driven by an ADP-dependent Glucokinase Linking Enhanced Glycolysis with Mitochondrial Reactive Oxygen Species Generation,” Cell Reports 2, no. 5 (2012): 1300-1315.

[63]

M. M. Kamiński, D. Röth, P. H. Krammer, and K. Gülow, “Mitochondria as Oxidative Signaling Organelles in T-cell Activation: Physiological Role and Pathological Implications,” Archivum Immunologiae Et Therapiae Experimentalis 61, no. 5 (2013): 367-384.

[64]

L. A. Sena, S. Li, A. Jairaman, et al., “Mitochondria Are Required for Antigen-Specific T Cell Activation through Reactive Oxygen Species Signaling,” Immunity 38, no. 2 (2013): 225-236.

[65]

T. W. Mak, M. Grusdat, G. S. Duncan, et al., “Glutathione Primes T Cell Metabolism for Inflammation,” Immunity 46, no. 4 (2017): 675-689.

[66]

G. Angelini, S. Gardella, M. Ardy, et al., “Antigen-presenting Dendritic Cells Provide the Reducing Extracellular Microenvironment Required for T Lymphocyte Activation,” PNAS 99, no. 3 (2002): 1491-1496.

[67]

M. Matsushita, S. Freigang, C. Schneider, et al., “T Cell Lipid Peroxidation Induces Ferroptosis and Prevents Immunity to Infection,” Journal of Experimental Medicine 212, no. 4 (2015): 555-568.

[68]

M. Matsushita, S. Freigang, C. Schneider, et al., “T Cell Lipid Peroxidation Induces Ferroptosis and Prevents Immunity to Infection,” Journal of Experimental Medicine 212, no. 4 (2015): 555-568.

[69]

C. Xu, S. Sun, T. Johnson, et al., “The Glutathione Peroxidase Gpx4 Prevents Lipid Peroxidation and Ferroptosis to Sustain Treg Cell Activation and Suppression of Antitumor Immunity,” Cell Reports 35, no. 11 (2021): 109235.

[70]

Y. Feng, M. Tang, M. Suzuki, et al., “Essential Role of NADPH Oxidase-Dependent Production of Reactive Oxygen Species in Maintenance of Sustained B Cell Receptor Signaling and B Cell Proliferation,” Journal of Immunology 202, no. 9 (2019): 2546-2557.

[71]

K. Kläsener, P. C. Maity, E. Hobeika, J. Yang, and M. Reth, “B Cell Activation Involves Nanoscale Receptor Reorganizations and inside-out Signaling by Syk,” Elife 3 (2014): e02069.

[72]

V. Rolli, M. Gallwitz, T. Wossning, et al., “Amplification of B Cell Antigen Receptor Signaling by a Syk/ITAM Positive Feedback Loop,” Molecular Cell 10, no. 5 (2002): 1057-1069.

[73]

M. Capasso, M. K. Bhamrah, T. Henley, et al., “HVCN1 modulates BCR Signal Strength via Regulation of BCR-dependent Generation of Reactive Oxygen Species,” Nature Immunology 11, no. 3 (2010): 265-272.

[74]

C. Nathan, A. Cunningham-Bussel, “Beyond Oxidative Stress: an Immunologist's Guide to Reactive Oxygen Species,” Nature Reviews Immunology 13, no. 5 (2013): 349-361.

[75]

C. Wu, L. Wang, S. Chen, et al., “Iron Induces B Cell Pyroptosis through Tom20-Bax-caspase-gasdermin E Signaling to Promote Inflammation Post-spinal Cord Injury,” Journal of Neuroinflammation 20, no. 1 (2023): 171.

[76]

C. Paiva, J. C. Godbersen, R. S. Soderquist, et al., “Cyclin-Dependent Kinase Inhibitor P1446A Induces Apoptosis in a JNK/p38 MAPK-Dependent Manner in Chronic Lymphocytic Leukemia B-Cells,” PLoS ONE 10, no. 11 (2015): e0143685.

[77]

G. B. Park, Y. Choi, Y. S. Kim, et al., “ROS-mediated JNK/p38-MAPK Activation Regulates Bax Translocation in Sorafenib-induced Apoptosis of EBV-transformed B Cells,” International Journal of Oncology 44, no. 3 (2014): 977-985.

[78]

C. F. Nathan, R. K. Root, “Hydrogen Peroxide Release from Mouse Peritoneal Macrophages: Dependence on Sequential Activation and Triggering,” Journal of Experimental Medicine 146, no. 6 (1977): 1648-1662.

[79]

M. Y. Zeng, I. Miralda, C. L. Armstrong, S. M. Uriarte, and J. Bagaitkar, “The Roles of NADPH Oxidase in Modulating Neutrophil Effector Responses,” Molecular Oral Microbiology 34, no. 2 (2019): 27-38.

[80]

C. Dunnill, T. Patton, J. Brennan, et al., “Reactive Oxygen Species (ROS) and Wound Healing: the Functional Role of ROS and Emerging ROS-modulating Technologies for Augmentation of the Healing Process,” International Wound Journal 14, no. 1 (2017): 89-96.

[81]

O. Sul, S. W. Ra, “Quercetin Prevents LPS-Induced Oxidative Stress and Inflammation by Modulating NOX2/ROS/NF-kB in Lung Epithelial Cells,” Molecules (Basel, Switzerland) 26, no. 22 (2021): 6949.

[82]

J. Liu, X. Han, T. Zhang, et al., “Reactive Oxygen Species (ROS) Scavenging Biomaterials for Anti-inflammatory Diseases: from Mechanism to Therapy,” Journal of Hematology & Oncology 16, no. 1 (2023): 116.

[83]

T. Luo, X. Zhou, M. Qin, et al., “Corilagin Restrains NLRP3 Inflammasome Activation and Pyroptosis through the ROS/TXNIP/NLRP3 Pathway to Prevent Inflammation,” Oxidative Medicine and Cellular Longevity 2022 (2022): 1652244.

[84]

L. Lian, Z. Le, Z. Wang, et al., “SIRT1 Inhibits High Glucose-Induced TXNIP/NLRP3 Inflammasome Activation and Cataract Formation,” Investigative Ophthalmology & Visual Science 64, no. 3 (2023): 16.

[85]

Q. Li, Y. Sun, B. Liu, et al., “ACT001 modulates the NF-κB/MnSOD/ROS Axis by Targeting IKKβ to Inhibit Glioblastoma Cell Growth,” Journal of Molecular Medicine (Berl) 98, no. 2 (2020): 263-277.

[86]

L. Huangfu, J. Wang, D. Li, et al., “Fraxetin Inhibits IKKβ, Blocks NF-κB Pathway and NLRP3 Inflammasome Activation, and Alleviates Spleen Injury in Sepsis,” Chemico-Biological Interactions 408 (2025): 111406.

[87]

Y. Wang, D. Han, Y. Huang, et al., “Oral Administration of punicalagin Attenuates Imiquimod-induced Psoriasis by Reducing ROS Generation and Inflammation via MAPK/ERK and NF-κB Signaling Pathways,” Phytotherapy Research 38, no. 2 (2024): 713-726.

[88]

L. Gong, Y. Lei, Y. Liu, et al., “Vaccarin Prevents Ox-LDL-induced HUVEC EndMT, Inflammation and Apoptosis by Suppressing ROS/p38 MAPK Signaling,” American Journal of Translational Research 11, no. 4 (2019): 2140-2154.

[89]

S. Yang, X. Li, M. Xiu, et al., “Flos Puerariae Ameliorates the Intestinal Inflammation of Drosophila via Modulating the Nrf2/Keap1, JAK-STAT and Wnt Signaling,” Frontiers in Pharmacology 13 (2022): 893758.

[90]

S. Li, P. Wen, D. Zhang, et al., “PGAM5 expression Levels in Heart Failure and Protection ROS-induced Oxidative Stress and Ferroptosis by Keap1/Nrf2,” Clinical and Experimental Hypertension 45, no. 1 (2023): 2162537.

[91]

L. Kong, J. Deng, X. Zhou, et al., “Sitagliptin Activates the p62-Keap1-Nrf2 Signalling Pathway to Alleviate Oxidative Stress and Excessive Autophagy in Severe Acute Pancreatitis-related Acute Lung Injury,” Cell Death & Disease 12, no. 10 (2021): 928.

[92]

K. Taguchi, H. Motohashi, and M. Yamamoto, “Molecular Mechanisms of the Keap1-Nrf2 Pathway in Stress Response and Cancer Evolution,” Genes to Cells 16, no. 2 (2011): 123-140.

[93]

W. Tu, H. Wang, S. Li, Q. Liu, and H. Sha, “The Anti-Inflammatory and Anti-Oxidant Mechanisms of the Keap1/Nrf2/ARE Signaling Pathway in Chronic Diseases,” Aging and Disease 10, no. 3 (2019): 637-651.

[94]

S. Kasai, S. Shimizu, Y. Tatara, J. Mimura, and K. Itoh, “Regulation of Nrf2 by Mitochondrial Reactive Oxygen Species in Physiology and Pathology,” Biomolecules 10, no. 2 (2020): 320.

[95]

S. Lo, M. Hannink, “PGAM5 tethers a Ternary Complex Containing Keap1 and Nrf2 to Mitochondria,” Experimental Cell Research 314, no. 8 (2008): 1789-1803.

[96]

A. Zeb, V. Choubey, R. Gupta, et al., “A Novel Role of KEAP1/PGAM5 Complex: ROS Sensor for Inducing Mitophagy,” Redox Biology 48 (2021): 102186.

[97]

E. Panieri, S. A. Pinho, G. J. M. Afonso, et al., “NRF2 and Mitochondrial Function in Cancer and Cancer Stem Cells,” Cells 11, no. 15 (2022): 2401.

[98]

Y. Qiu, B. Wan, G. Liu, et al., “Nrf2 protects against Seawater Drowning-induced Acute Lung Injury via Inhibiting Ferroptosis,” Respiratory Research 21, no. 1 (2020): 232.

[99]

L. Zhang, J. Zhang, Y. Jin, et al., “Nrf2 Is a Potential Modulator for Orchestrating Iron Homeostasis and Redox Balance in Cancer Cells,” Frontiers in Cell and Developmental Biology 9 (2021): 728172.

[100]

Z. Wang, Y. Chang, Y. Liu, et al., “Inhibition of the lncRNA MIAT Prevents Podocyte Injury and Mitotic Catastrophe in Diabetic Nephropathy,” Molecular Therapy Nucleic Acids 28 (2022): 136-153.

[101]

R. Radhakrishnan, R. A. Kowluru, “Long Noncoding RNA MALAT1 and Regulation of the Antioxidant Defense System in Diabetic Retinopathy,” Diabetes 70, no. 1 (2021): 227-239.

[102]

J. Zhang, H. Sun, L. Zhu, et al., “Micro Ribonucleic Acid 27a Aggravates Ferroptosis During Early Ischemic Stroke of Rats through Nuclear Factor Erythroid-2-Related Factor 2,” Neuroscience 504 (2022): 10-20.

[103]

S. Basak, A. Hoffmann, “Crosstalk via the NF-kappaB Signaling System,” Cytokine & Growth Factor Reviews 19, no. 3-4 (2008): 187-197.

[104]

M. J. Morgan, Z. Liu, “Crosstalk of Reactive Oxygen Species and NF-κB Signaling,” Cell Research 21, no. 1 (2011): 103-115.

[105]

Y. Liu, D. Xiang, H. Zhang, H. Yao, and Y. Wang, “Hypoxia-Inducible Factor-1: a Potential Target to Treat Acute Lung Injury,” Oxidative Medicine and Cellular Longevity 2020 (2020): 8871476.

[106]

Y. Watanabe, C. E. Murdoch, S. Sano, et al., “Glutathione Adducts Induced by Ischemia and Deletion of Glutaredoxin-1 Stabilize HIF-1α and Improve Limb Revascularization,” PNAS 113, no. 21 (2016): 6011-6016.

[107]

A. L. Orr, L. Vargas, C. N. Turk, et al., “Suppressors of Superoxide Production from Mitochondrial Complex III,” Nature Chemical Biology 11, no. 11 (2015): 834-836.

[108]

N. S. Chandel, D. S. McClintock, C. E. Feliciano, et al., “Reactive Oxygen Species Generated at Mitochondrial Complex III Stabilize Hypoxia-inducible Factor-1alpha during Hypoxia: a Mechanism of O2 Sensing,” Journal of Biological Chemistry 275, no. 33 (2000): 25130-25138.

[109]

D. R. Calnan, A. Brunet, “The FoxO Code,” Oncogene 27, no. 16 (2008): 2276-2288.

[110]

Y. Yan, H. Huang, “Interplay among PI3K/AKT, PTEN/FOXO and AR Signaling in Prostate Cancer,” Advances in Experimental Medicine and Biology 1210 (2019): 319-331.

[111]

E. Nishida, Y. Gotoh, “The MAP Kinase Cascade Is Essential for Diverse Signal Transduction Pathways,” Trends in Biochemical Sciences 18, no. 4 (1993): 128-131.

[112]

K. Z. Guyton, Y. Liu, M. Gorospe, Q. Xu, and N. J. Holbrook, “Activation of Mitogen-activated Protein Kinase by H2O2. Role in Cell Survival Following Oxidant Injury,” Journal of Biological Chemistry 271, no. 8 (1996): 4138-4142.

[113]

M. A. G. Essers, S. Weijzen, A. M. M. de Vries-Smits, et al., “FOXO Transcription Factor Activation by Oxidative Stress Mediated by the Small GTPase Ral and JNK,” Embo Journal 23, no. 24 (2004): 4802-4812.

[114]

J. Lewandowska, A. Bartoszek, “DNA Methylation in Cancer Development, Diagnosis and Therapy-multiple Opportunities for Genotoxic Agents to Act as Methylome Disruptors or Remediators,” Mutagenesis 26, no. 4 (2011): 475-487.

[115]

N. Chia, L. Wang, X. Lu, et al., “Hypothesis: Environmental Regulation of 5-hydroxymethylcytosine by Oxidative Stress,” Epigenetics 6, no. 7 (2011): 853-856.

[116]

N. E. Simpson, V. P. Tryndyak, M. Pogribna, F. A. Beland, and I. P. Pogribny, “Modifying Metabolically Sensitive Histone Marks by Inhibiting Glutamine Metabolism Affects Gene Expression and Alters Cancer Cell Phenotype,” Epigenetics 7, no. 12 (2012): 1413-1420.

[117]

M. D. Jelic, A. D. Mandic, S. M. Maricic, and B. U. Srdjenovic, “Oxidative Stress and Its Role in Cancer,” Journal of Cancer Research and Therapeutics 17, no. 1 (2021): 22-28.

[118]

P. W. Turk, A. Laayoun, S. S. Smith, and S. A. Weitzman, “DNA Adduct 8-hydroxyl-2'-deoxyguanosine (8-hydroxyguanine) Affects Function of human DNA Methyltransferase,” Carcinogenesis 16, no. 5 (1995): 1253-1255.

[119]

V. Valinluck, H. Tsai, D. K. Rogstad, et al., “Oxidative Damage to Methyl-CpG Sequences Inhibits the Binding of the Methyl-CpG Binding Domain (MBD) of Methyl-CpG Binding Protein 2 (MeCP2),” Nucleic Acids Research 32, no. 14 (2004): 4100-4108.

[120]

Y. Zhang, Z. Xu, J. Ding, et al., “HZ08 suppresses RelB-activated MnSOD Expression and Enhances Radiosensitivity of Prostate Cancer Cells,” Journal of Experimental & Clinical Cancer Research 37, no. 1 (2018): 174.

[121]

D. Peng, T. Hu, A. Jiang, et al., “Location-specific Epigenetic Regulation of the Metallothionein 3 Gene in Esophageal Adenocarcinomas,” PLoS ONE 6, no. 7 (2011): e22009.

[122]

M. Tada, O. Yokosuka, K. Fukai, et al., “Hypermethylation of NAD(P)H: Quinone Oxidoreductase 1 (NQO1) Gene in human Hepatocellular Carcinoma,” Journal of Hepatology 42, no. 4 (2005): 511-519.

[123]

A. Penn, N. McPherson, T. Fullston, B. Arman, and D. Zander-Fox, “Maternal High-fat Diet Changes DNA Methylation in the Early Embryo by Disrupting the TCA Cycle Intermediary Alpha Ketoglutarate,” Reproduction (Cambridge, England) 165, no. 4 (2023): 347-362.

[124]

I. Martínez-Reyes, N. S. Chandel, “Mitochondrial TCA Cycle Metabolites Control Physiology and Disease,” Nature Communications 11, no. 1 (2020): 102.

[125]

Y. Zhao, X. Fan, Q. Wang, et al., “ROS Promote Hyper-methylation of NDRG2 Promoters in a DNMTS-dependent Manner: Contributes to the Progression of Renal Fibrosis,” Redox Biology 62 (2023): 102674.

[126]

S. Guan, L. Zhao, and R. Peng, “Mitochondrial Respiratory Chain Supercomplexes: from Structure to Function,” International Journal of Molecular Sciences 23, no. 22 (2022): 13880.

[127]

P. Hernansanz-Agustin, J. A. Enriquez, “Generation of Reactive Oxygen Species by Mitochondria,” Antioxidants (Basel) 10, no. 3 (2021): 415.

[128]

T. Chen, H. Wang, C. Chang, and S. Lee, “Mitochondrial Glutathione in Cellular Redox Homeostasis and Disease Manifestation,” International Journal of Molecular Sciences 25, no. 2 (2024): 1314.

[129]

Y. Wang, R. Branicky, A. Noë, and S. Hekimi, “Superoxide Dismutases: Dual Roles in Controlling ROS Damage and Regulating ROS Signaling,” Journal of Cell Biology 217, no. 6 (2018): 1915-1928.

[130]

S. W. Caito, M. Aschner, “Mitochondrial Redox Dysfunction and Environmental Exposures,” Antioxid Redox Signaling 23, no. 6 (2015): 578-595.

[131]

V. G. Grivennikova, A. D. Vinogradov, “Generation of Superoxide by the Mitochondrial Complex I,” Biochimica Et Biophysica Acta 1757, no. 5-6 (2006): 553-561.

[132]

P. Willems, R. Rossignol, C. E. J. Dieteren, M. P. Murphy, and W. J. H. Koopman, “Redox Homeostasis and Mitochondrial Dynamics,” Cell Metabolism 22, no. 2 (2015): 207-218.

[133]

D. G. Hardie, F. A. Ross, and S. A. Hawley, “AMPK: a Nutrient and Energy Sensor That Maintains Energy Homeostasis,” Nature Reviews Molecular Cell Biology 13, no. 4 (2012): 251-262.

[134]

J. Mars, B. Culjkovic-Kraljacic, and K. L. B. Borden, “eIF4E orchestrates mRNA Processing, RNA Export and Translation to Modify Specific Protein Production,” Nucleus 15, no. 1 (2024): 2360196.

[135]

A. Ghosh, N. Shcherbik, “Effects of Oxidative Stress on Protein Translation: Implications for Cardiovascular Diseases,” International Journal of Molecular Sciences 21, no. 8 (2020): 2661.

[136]

C. G. Proud, “eIF2 and the Control of Cell Physiology,” Seminars in Cell & Developmental Biology 16, no. 1 (2005): 3-12.

[137]

T. Adomavicius, M. Guaita, Y. Zhou, et al., “The Structural Basis of Translational Control by eIF2 Phosphorylation,” Nature Communications 10, no. 1 (2019): 2136.

[138]

C. Picazo, M. Molin, “Impact of Hydrogen Peroxide on Protein Synthesis in Yeast,” Antioxidants (Basel) 10, no. 6 (2021): 952.

[139]

V. Jha, T. Kumari, V. Manickam, et al., “ERO1-PDI Redox Signaling in Health and Disease,” Antioxid Redox Signaling 35, no. 13 (2021): 1093-1115.

[140]

A. G. Shergalis, S. Hu, A. R. Bankhead, and N. Neamati, “Role of the ERO1-PDI Interaction in Oxidative Protein Folding and Disease,” Pharmacology & Therapeutics 210 (2020): 107525.

[141]

D. Eletto, E. Chevet, Y. Argon, and C. Appenzeller-Herzog, “Redox Controls UPR to Control Redox,” Journal of Cell Science 127, no. Pt 17 (2014): 3649-3658.

[142]

Z. Zhang, L. Zhang, L. Zhou, et al., “Redox Signaling and Unfolded Protein Response Coordinate Cell Fate Decisions under ER Stress,” Redox Biology 25 (2019): 101047.

[143]

T. Grune, B. Catalgol, A. Licht, et al., “HSP70 mediates Dissociation and Reassociation of the 26S Proteasome during Adaptation to Oxidative Stress,” Free Radical Biology and Medicine 51, no. 7 (2011): 1355-1364.

[144]

M. Demasi, L. E. S. Netto, G. M. Silva, et al., “Redox Regulation of the Proteasome via S-glutathionylation,” Redox Biology 2 (2013): 44-51.

[145]

J. Zhang, X. Wang, V. Vikash, et al., “ROS and ROS-Mediated Cellular Signaling,” Oxidative Medicine and Cellular Longevity 2016 (2016): 4350965.

[146]

G. Filomeni, D. De Zio, and F. Cecconi, “Oxidative Stress and Autophagy: the Clash between Damage and Metabolic Needs,” Cell Death and Differentiation 22, no. 3 (2015): 377-388.

[147]

B. Carroll, E. G. Otten, D. Manni, et al., “Oxidation of SQSTM1/p62 Mediates the Link between Redox state and Protein Homeostasis,” Nature Communications 9, no. 1 (2018): 256.

[148]

K. Frudd, T. Burgoyne, and J. R. Burgoyne, “Oxidation of Atg3 and Atg7 Mediates Inhibition of Autophagy,” Nature Communications 9, no. 1 (2018): 95.

[149]

L. Gao, W. Fu, Y. Liu, et al., “Albiflorin Inhibits Advanced Glycation End Products-Induced ROS and MMP-1 Expression in Gingival Fibroblasts,” Current Stem Cell Research & Therapy (2024).

[150]

L. Cao, J. Liu, L. Zhang, X. Xiao, and W. Li, “Curcumin Inhibits H2O2-induced Invasion and Migration of human Pancreatic Cancer via Suppression of the ERK/NF-κB Pathway,” Oncology Reports 36, no. 4 (2016): 2245-2251.

[151]

V. Ibáñez Gaspar, T. McMorrow, “The Curcuminoid EF24 in Combination with TRAIL Reduces Human Renal Cancer Cell Migration by Decreasing MMP-2/MMP-9 Activity through a Reduction in H(2)O(2),” International Journal of Molecular Sciences 24, no. 2 (2023): 1043.

[152]

H. Jang, G. Kim, J. Kim, et al., “Fisetin Inhibits UVA-Induced Expression of MMP-1 and MMP-3 through the NOX/ROS/MAPK Pathway in Human Dermal Fibroblasts and Human Epidermal Keratinocytes,” International Journal of Molecular Sciences 24, no. 24 (2023): 17358.

[153]

J. H. Kim, H. D. Jeong, M. J. Song, et al., “SOD3 Suppresses the Expression of MMP-1 and Increases the Integrity of Extracellular Matrix in Fibroblasts,” Antioxidants (Basel) 11, no. 5 (2022): 928.

[154]

T. Fukai, M. Ushio-Fukai, “Cross-Talk between NADPH Oxidase and Mitochondria: Role in ROS Signaling and Angiogenesis,” Cells 9, no. 8 (2020): 1849.

[155]

D. C. Stieg, Y. Wang, L. Liu, and B. Jiang, “ROS and miRNA Dysregulation in Ovarian Cancer Development, Angiogenesis and Therapeutic Resistance,” International Journal of Molecular Sciences 23, no. 12 (2022): 6702.

[156]

Y. Huang, G. Nan, “Oxidative Stress-induced Angiogenesis,” Journal of Clinical Neuroscience 63 (2019): 13-16.

[157]

Y. Kim, S. Kim, R. Tatsunami, et al., “ROS-induced ROS Release Orchestrated by Nox4, Nox2, and Mitochondria in VEGF Signaling and Angiogenesis,” American Journal of Physiology. Cell Physiology 312, no. 6 (2017): C749-C764.

[158]

A. Vermot, I. Petit-Härtlein, S. M. E. Smith, and F. Fieschi, “NADPH Oxidases (NOX): an Overview from Discovery, Molecular Mechanisms to Physiology and Pathology,” Antioxidants (Basel) 10, no. 6 (2021): 890.

[159]

J. El-Benna, P. M. Dang, “Gougerot-Pocidalo M. Priming of the Neutrophil NADPH Oxidase Activation: Role of p47phox Phosphorylation and NOX2 Mobilization to the Plasma Membrane,” Seminars in Immunopathology 30, no. 3 (2008): 279-289.

[160]

S. Lu, Y. Liang, S. Yang, et al., “Stachydrine Hydrochloride Regulates the NOX2-ROS-Signaling Axis in Pressure-Overload-Induced Heart Failure,” International Journal of Molecular Sciences 24, no. 18 (2023): 14369.

[161]

H. S. Park, S. H. Lee, D. Park, et al., “Sequential Activation of Phosphatidylinositol 3-kinase, Beta Pix, Rac1, and Nox1 in Growth Factor-induced Production of H2O2,” Molecular and Cellular Biology 24, no. 10 (2004): 4384-4394.

[162]

H. Lo, Y. Tsai, W. Du, C. Tsou, and W. Wu, “A Naturally Occurring Carotenoid, Lutein, Reduces PDGF and H₂O₂ Signaling and Compromised Migration in Cultured Vascular Smooth Muscle Cells,” Journal of Biomedical Science 19, no. 1 (2012): 18.

[163]

P. A. Tyurin-Kuzmin, N. D. Zhdanovskaya, A. A. Sukhova, et al., “Nox4 and Duox1/2 Mediate Redox Activation of Mesenchymal Cell Migration by PDGF,” PLoS ONE 11, no. 4 (2016): e0154157.

[164]

L. Valgimigli, “Lipid Peroxidation and Antioxidant Protection,” Biomolecules 13, no. 9 (2023): 1291.

[165]

H. Yin, L. Xu, and N. A. Porter, “Free Radical Lipid Peroxidation: Mechanisms and Analysis,” Chemical Reviews 111, no. 10 (2011): 5944-5972.

[166]

A. Gęgotek, E. Skrzydlewska, “Lipid Peroxidation Products' role in Autophagy Regulation,” Free Radical Biology and Medicine 212 (2024): 375-383.

[167]

L. Su, J. Zhang, H. Gomez, et al., “Reactive Oxygen Species-Induced Lipid Peroxidation in Apoptosis, Autophagy, and Ferroptosis,” Oxidative Medicine and Cellular Longevity 2019 (2019): 5080843.

[168]

D. Li, G. Zhang, Z. Wang, et al., “Idebenone Attenuates Ferroptosis by Inhibiting Excessive Autophagy via the ROS-AMPK-mTOR Pathway to Preserve Cardiac Function after Myocardial Infarction,” European Journal of Pharmacology 943 (2023): 175569.

[169]

D. Han, L. Jiang, X. Gu, et al., “SIRT3 deficiency Is Resistant to Autophagy-dependent Ferroptosis by Inhibiting the AMPK/mTOR Pathway and Promoting GPX4 Levels,” Journal of Cellular Physiology 235, no. 11 (2020): 8839-8851.

[170]

C. Liu, K. Yao, Q. Tian, et al., “CXCR4-BTK Axis Mediate Pyroptosis and Lipid Peroxidation in Early Brain Injury after Subarachnoid Hemorrhage via NLRP3 Inflammasome and NF-κB Pathway,” Redox Biology 68 (2023): 102960.

[171]

T. Xu, J. Cui, R. Xu, J. Cao, and M. Guo, “Microplastics Induced Inflammation and Apoptosis via Ferroptosis and the NF-κB Pathway in Carp,” Aquatic Toxicology 262 (2023): 106659.

[172]

M. M. Gaschler, B. R. Stockwell, “Lipid Peroxidation in Cell Death,” Biochemical and Biophysical Research Communications 482, no. 3 (2017): 419-425.

[173]

Z. Feng, F. Meng, F. Huo, et al., “Inhibition of Ferroptosis Rescues M2 Macrophages and Alleviates Arthritis by Suppressing the HMGB1/TLR4/STAT3 Axis in M1 Macrophages,” Redox Biology 75 (2024): 103255.

[174]

A. Ashraf, J. Jeandriens, H. G. Parkes, and P. So, “Iron Dyshomeostasis, Lipid Peroxidation and Perturbed Expression of Cystine/Glutamate Antiporter in Alzheimer's Disease: Evidence of Ferroptosis,” Redox Biology 32 (2020): 101494.

[175]

T. J. Montine, M. D. Neely, J. F. Quinn, et al., “Lipid Peroxidation in Aging Brain and Alzheimer's Disease,” Free Radical Biology and Medicine 33, no. 5 (2002): 620-626.

[176]

N. T. Moldogazieva, S. P. Zavadskiy, D. V. Astakhov, and A. A. Terentiev, “Lipid Peroxidation: Reactive Carbonyl Species, Protein/DNA Adducts, and Signaling Switches in Oxidative Stress and Cancer,” Biochemical and Biophysical Research Communications 687 (2023): 149167.

[177]

G. Pistritto, D. Trisciuoglio, C. Ceci, A. Garufi, and G. D'Orazi, “Apoptosis as Anticancer Mechanism: Function and Dysfunction of Its Modulators and Targeted Therapeutic Strategies,” Aging (Albany NY) 8, no. 4 (2016): 603-619.

[178]

G. Yu, H. Luo, N. Zhang, et al., “Loss of p53 Sensitizes Cells to Palmitic Acid-Induced Apoptosis by Reactive Oxygen Species Accumulation,” International Journal of Molecular Sciences 20, no. 24 (2019): 6268.

[179]

W. Yu, C. Lu, B. Wang, X. Ren, and K. Xu, “Effects of Rapamycin on Osteosarcoma Cell Proliferation and Apoptosis by Inducing Autophagy,” European Review for Medical and Pharmacological Sciences 24, no. 2 (2020): 915-921.

[180]

A. Z. Spitz, E. Gavathiotis, “Physiological and Pharmacological Modulation of BAX,” Trends in Pharmacological Sciences 43, no. 3 (2022): 206-220.

[181]

D. Gradinaru, C. Borsa, C. Ionescu, and G. I. Prada, “Oxidized LDL and NO Synthesis-Biomarkers of Endothelial Dysfunction and Ageing,” Mechanisms of Ageing and Development 151 (2015): 101-113.

[182]

S. Parthasarathy, A. Raghavamenon, M. O. Garelnabi, and N. Santanam, “Oxidized Low-density Lipoprotein,” Methods in Molecular Biology 610 (2010): 403-417.

[183]

A. Hartley, D. Haskard, and R. Khamis, “Oxidized LDL and Anti-oxidized LDL Antibodies in Atherosclerosis—Novel Insights and Future Directions in Diagnosis and Therapy<Sup/>” Trends in Cardiovascular Medicine 29, no. 1 (2019): 22-26.

[184]

A. R. Monteiro, D. J. Barbosa, F. Remião, and R. Silva, “Alzheimer's disease: Insights and New Prospects in Disease Pathophysiology, Biomarkers and Disease-modifying Drugs,” Biochemical Pharmacology 211 (2023): 115522.

[185]

E. J. Henriksen, M. K. Diamond-Stanic, and E. M. Marchionne, “Oxidative Stress and the Etiology of Insulin Resistance and Type 2 Diabetes,” Free Radical Biology and Medicine 51, no. 5 (2011): 993-999.

[186]

J. Cadet, K. J. A. Davies, M. H. Medeiros, P. Di Mascio, and J. R. Wagner, “Formation and Repair of Oxidatively Generated Damage in Cellular DNA,” Free Radical Biology and Medicine 107 (2017): 13-34.

[187]

J. Cadet, M. Berger, T. Douki, and J. L. Ravanat, “Oxidative Damage to DNA: Formation, Measurement, and Biological Significance,” Reviews of Physiology, Biochemistry and Pharmacology 131 (1997): 1-87.

[188]

Z. Li, A. H. Pearlman, and P. Hsieh, “DNA Mismatch Repair and the DNA Damage Response,” Dna Repair 38 (2016): 94-101.

[189]

T. Hemnani, M. S. Parihar, “Reactive Oxygen Species and Oxidative DNA Damage,” Indian Journal of Physiology and Pharmacology 42, no. 4 (1998): 440-452.

[190]

M. J. Iqbal, A. Kabeer, Z. Abbas, et al., “Interplay of Oxidative Stress, Cellular Communication and Signaling Pathways in Cancer,” Cell Communication and Signaling 22, no. 1 (2024): 7.

[191]

S. Reuter, S. C. Gupta, M. M. Chaturvedi, and B. B. Aggarwal, “Oxidative Stress, Inflammation, and Cancer: How Are They Linked?” Free Radical Biology and Medicine 49, no. 11 (2010): 1603-1616.

[192]

R. Bai, J. Guo, X. Ye, Y. Xie, and T. Xie, “Oxidative Stress: the Core Pathogenesis and Mechanism of Alzheimer's Disease,” Ageing Research Reviews 77 (2022): 101619.

[193]

R. X. Santos, S. C. Correia, X. Zhu, et al., “Mitochondrial DNA Oxidative Damage and Repair in Aging and Alzheimer's Disease,” Antioxid Redox Signaling 18, no. 18 (2013): 2444-2457.

[194]

M. Kieroń, C. Żekanowski, A. Falk, and M. Wężyk, “Oxidative DNA Damage Signalling in Neural Stem Cells in Alzheimer's Disease,” Oxidative Medicine and Cellular Longevity 2019 (2019): 2149812.

[195]

F. J. Bock, S. W. G. Tait, “Mitochondria as Multifaceted Regulators of Cell Death,” Nature Reviews Molecular Cell Biology 21, no. 2 (2020): 85-100.

[196]

L. A. Pradelli, M. Bénéteau, and J. Ricci, “Mitochondrial Control of Caspase-dependent and -independent Cell Death,” Cellular and Molecular Life Sciences 67, no. 10 (2010): 1589-1597.

[197]

J. Estaquier, F. Vallette, J. Vayssiere, and B. Mignotte, “The Mitochondrial Pathways of Apoptosis,” Advances in Experimental Medicine and Biology 942 (2012): 157-183.

[198]

W. Zhang, Y. Liu, Z. An, et al., “Mediating Effect of ROS on mtDNA Damage and Low ATP Content Induced by Arsenic Trioxide in Mouse Oocytes,” Toxicology in Vitro 25, no. 4 (2011): 979-984.

[199]

K. N. Belosludtsev, N. V. Belosludtseva, and M. V. Dubinin, “Diabetes Mellitus, Mitochondrial Dysfunction and Ca(2+)-Dependent Permeability Transition Pore,” International Journal of Molecular Sciences 21, no. 18 (2020): 6559.

[200]

W. Cai, K. Chong, Y. Huang, C. Huang, and L. Yin, “Empagliflozin Improves Mitochondrial Dysfunction in Diabetic Cardiomyopathy by Modulating Ketone Body Metabolism and Oxidative Stress,” Redox Biology 69 (2024): 103010.

[201]

R. Blake, I. A. Trounce, “Mitochondrial dysfunction and complications associated With diabetes,” Biochimica Et Biophysica Acta 1840, no. 4 (2014): 1404-1412.

[202]

R. Veluthakal, D. Esparza, J. M. Hoolachan, et al., “Mitochondrial Dysfunction, Oxidative Stress, and Inter-Organ Miscommunications in T2D Progression,” International Journal of Molecular Sciences 25, no. 3 (2024): 1504.

[203]

M. Camacho-Encina, L. K. Booth, R. E. Redgrave, et al., “Cellular Senescence, Mitochondrial Dysfunction, and Their Link to Cardiovascular Disease,” Cells 13, no. 4 (2024): 353.

[204]

D. L. Kirkman, A. T. Robinson, M. J. Rossman, D. R. Seals, and D. G. Edwards, “Mitochondrial Contributions to Vascular Endothelial Dysfunction, Arterial Stiffness, and Cardiovascular Diseases,” American Journal of Physiology. Heart and Circulatory Physiology 320, no. 5 (2021): H2080-H2100.

[205]

D. A. Chistiakov, T. P. Shkurat, A. A. Melnichenko, A. V. Grechko, and A. N. Orekhov, “The Role of Mitochondrial Dysfunction in Cardiovascular Disease: A Brief Review,” Annals of Medicine 50, no. 2 (2018): 121-127.

[206]

S. Yoo, J. Park, S. Kim, and Y. Jung, “Emerging Perspectives on Mitochondrial Dysfunction and Inflammation in Alzheimer's Disease,” BMB Reports 53, no. 1 (2020): 35-46.

[207]

M. T. Islam, “Oxidative Stress and Mitochondrial Dysfunction-Linked Neurodegenerative Disorders,” Neurological Research 39, no. 1 (2017): 73-82.

[208]

T. Ashleigh, R. H. Swerdlow, and M. F. Beal, “The Role of Mitochondrial Dysfunction in Alzheimer's Disease Pathogenesis,” Alzheimer's & Dementia 19, no. 1 (2023): 333-342.

[209]

T. Alqahtani, S. L. Deore, A. A. Kide, et al., “Mitochondrial Dysfunction and Oxidative Stress in Alzheimer's Disease, and Parkinson's Disease, Huntington's Disease and Amyotrophic Lateral Sclerosis-An Updated Review,” Mitochondrion 71 (2023): 83-92.

[210]

P. H. Reddy, D. M. Oliver, “Amyloid Beta and Phosphorylated Tau-Induced Defective Autophagy and Mitophagy in Alzheimer's Disease,” Cells 8, no. 5 (2019): 488.

[211]

B. Li, P. Zhou, K. Xu, et al., “Metformin induces cell cycle arrest, apoptosis and autophagy Through ROS/JNK signaling pathway in human osteosarcoma,” International Journal of Biological Sciences 16, no. 1 (2020): 74-84.

[212]

X. Jia, Q. Zhang, L. Xu, W. Yao, and L. Wei, “Lotus leaf flavonoids induce apoptosis of human lung cancer A549 cells Through the ROS/p38 MAPK pathway,” Biological Research 54, no. 1 (2021): 7.

[213]

N. Kelley, D. Jeltema, Y. Duan, and Y. He, “The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation,” International Journal of Molecular Sciences 20, no. 13 (2019): 3328.

[214]

E. Kropf, M. Fahnestock, “Effects of Reactive Oxygen and Nitrogen Species on TrkA Expression and Signalling: Implications for proNGF in Aging and Alzheimer's Disease,” Cells 10, no. 8 (2021): 1983.

[215]

N. Üremiş, M. M. Üremiş, “Oxidative/Nitrosative Stress, Apoptosis, and Redox Signaling: Key Players in Neurodegenerative Diseases,” Journal of Biochemical and Molecular Toxicology 39, no. 1 (2025): e70133.

[216]

Y. Wang, T. Veremeyko, A. H. Wong, et al., “Downregulation of miR-132/212 Impairs S-Nitrosylation Balance and Induces Tau Phosphorylation in Alzheimer's Disease,” Neurobiology of Aging 51 (2017): 156-166.

[217]

M. R. Reynolds, J. F. Reyes, Y. Fu, et al., “Tau Nitration Occurs at Tyrosine 29 in the Fibrillar Lesions of Alzheimer's Disease and Other Tauopathies,” Journal of Neuroscience 26, no. 42 (2006): 10636-10645.

[218]

J. F. Reyes, Y. Fu, L. Vana, N. M. Kanaan, and L. I. Binder, “Tyrosine nitration Within the proline-rich region of Tau in Alzheimer's disease,” American Journal of Pathology 178, no. 5 (2011): 2275-2285.

[219]

Z. Liu, Q. Liu, B. Zhang, et al., “Blood-Brain Barrier Permeable and NO-Releasing Multifunctional Nanoparticles for Alzheimer's Disease Treatment: Targeting NO/cGMP/CREB Signaling Pathways,” Journal of Medicinal Chemistry 64, no. 18 (2021): 13853-13872.

[220]

M. R. Tropea, W. Gulisano, V. Vacanti, et al., “Nitric Oxide/cGMP/CREB Pathway and Amyloid-Beta Crosstalk: From Physiology to Alzheimer's Disease,” Free Radical Biology and Medicine 193, no. Pt 2 (2022): 657-668.

[221]

Y. Zhang, M. Wang, and W. Chang, “Iron Dyshomeostasis and Ferroptosis in Alzheimer's Disease: Molecular Mechanisms of Cell Death and Novel Therapeutic Drugs and Targets for AD,” Frontiers in Pharmacology 13 (2022): 983623.

[222]

Q. Han, L. Sun, and K. Xiang, “Research Progress of Ferroptosis in Alzheimer Disease: A Review,” Medicine 102, no. 36 (2023): e35142.

[223]

P. Baruah, H. Moorthy, M. Ramesh, D. Padhi, and T. Govindaraju, “A Natural Polyphenol Activates and Enhances GPX4 to Mitigate Amyloid-β Induced Ferroptosis in Alzheimer's Disease,” Chemical Science 14, no. 35 (2023): 9427-9438.

[224]

Y. Yong, L. Yan, J. Wei, et al., “A Novel Ferroptosis Inhibitor, Thonningianin A, Improves Alzheimer's Disease by Activating GPX4,” Theranostics 14, no. 16 (2024): 6161-6184.

[225]

M. W. Park, H. W. Cha, J. Kim, et al., “NOX4 Promotes Ferroptosis of Astrocytes by Oxidative Stress-Induced Lipid Peroxidation via the Impairment of Mitochondrial Metabolism in Alzheimer's Diseases,” Redox Biology 41 (2021): 101947.

[226]

L. Mahoney-Sánchez, H. Bouchaoui, S. Ayton, et al., “Ferroptosis and its Potential Role in the Physiopathology of Parkinson's Disease,” Progress in Neurobiology 196 (2021): 101890.

[227]

P. Dusek, T. Hofer, J. Alexander, P. M. Roos, and J. O. Aaseth, “Cerebral Iron Deposition in Neurodegeneration,” Biomolecules 12, no. 5 (2022): 714.

[228]

J. Wang, N. Song, H. Jiang, J. Wang, and J. Xie, “Pro-Inflammatory Cytokines Modulate Iron Regulatory Protein 1 Expression and Iron Transportation Through Reactive Oxygen/nitrogen Species Production in Ventral Mesencephalic Neurons,” Biochimica Et Biophysica Acta 1832, no. 5 (2013): 618-625.

[229]

I. Villalón-García, S. Povea-Cabello, M. Álvarez-Córdoba, et al., “Vicious Cycle of Lipid Peroxidation and Iron Accumulation in Neurodegeneration,” Neural Regeneration Research 18, no. 6 (2023): 1196-1202.

[230]

S. Mehra, S. Sahay, and S. K. Maji, “α-Synuclein Misfolding and Aggregation: Implications in Parkinson's Disease Pathogenesis,” Biochimica et Biophysica Acta (BBA) - Proteins and Proteomics 1867, no. 10 (2019): 890-908.

[231]

X. Dong-Chen, C. Yong, X. Yang, S. Chen-Yu, and P. Li-Hua, “Signaling Pathways in Parkinson's Disease: Molecular Mechanisms and Therapeutic Interventions,” Signal Transduction and Targeted Therapy 8, no. 1 (2023): 73.

[232]

J. Sun, X. Lin, D. Lu, et al., “Midbrain Dopamine Oxidation Links Ubiquitination of Glutathione Peroxidase 4 to Ferroptosis of Dopaminergic Neurons,” Journal of Clinical Investigation 133, no. 10 (2023): e165228.

[233]

K. M. Lohr, S. T. Masoud, A. Salahpour, and G. W. Miller, “Membrane Transporters as Mediators of Synaptic Dopamine Dynamics: Implications for Disease,” European Journal of Neuroscience 45, no. 1 (2017): 20-33.

[234]

C. Mytilineou, B. C. Kramer, and J. A. Yabut, “Glutathione Depletion and Oxidative Stress,” Parkinsonism & Related Disorders 8, no. 6 (2002): 385-387.

[235]

G. Bjorklund, M. Peana, M. Maes, M. Dadar, and B. Severin, “The Glutathione System in Parkinson's Disease and its Progression,” Neuroscience and Biobehavioral Reviews 120 (2021): 470-478.

[236]

S. R. Subramaniam, M. Chesselet, “Mitochondrial Dysfunction and Oxidative Stress in Parkinson's Disease,” Progress in Neurobiology 106-107 (2013): 17-32.

[237]

D. J. Surmeier, J. N. Guzman, J. Sanchez-Padilla, and P. T. Schumacker, “The Role of Calcium and Mitochondrial Oxidant Stress in the Loss of Substantia Nigra Pars Compacta Dopaminergic Neurons in Parkinson's Disease,” Neuroscience 198 (2011): 221-231.

[238]

V. Zaman, D. C. Shields, R. Shams, et al., “Cellular and Molecular Pathophysiology in the Progression of Parkinson's Disease,” Metabolic Brain Disease 36, no. 5 (2021): 815-827.

[239]

C. Munteanu, A. I. Galaction, M. Turnea, et al., “Redox Homeostasis, Gut Microbiota, and Epigenetics in Neurodegenerative Diseases: A Systematic Review,” Antioxidants (Basel) 13, no. 9 (2024): 1062.

[240]

M. G. Stykel, S. D. Ryan, “Nitrosative Stress in Parkinson's Disease,” NPJ Parkinson's Disease 8, no. 1 (2022): 104.

[241]

S. Aras, G. Tanriover, M. Aslan, P. Yargicoglu, and A. Agar, “The Role of Nitric Oxide on Visual-Evoked Potentials in MPTP-Induced Parkinsonism in Mice,” Neurochemistry International 72 (2014): 48-57.

[242]

D. Tripathy, J. Chakraborty, and K. P. Mohanakumar, “Antagonistic Pleiotropic Effects of Nitric Oxide in the Pathophysiology of Parkinson's Disease,” Free Radical Research 49, no. 9 (2015): 1129-1139.

[243]

J. Zheng, J. Winderickx, V. Franssens, and B. Liu, “A Mitochondria-Associated Oxidative Stress Perspective on Huntington's Disease,” Frontiers in Molecular Neuroscience 11 (2018): 329.

[244]

A. Singh, R. Kukreti, L. Saso, and S. Kukreti, “Oxidative Stress: A Key Modulator in Neurodegenerative Diseases,” Molecules (Basel, Switzerland) 24, no. 8 (2019): 1583.

[245]

D. M. Teleanu, A. Niculescu, I. I. Lungu, et al., “An Overview of Oxidative Stress, Neuroinflammation, and Neurodegenerative Diseases,” International Journal of Molecular Sciences 23, no. 11 (2022): 5938.

[246]

D. A. Simmons, M. Casale, B. Alcon, et al., “Ferritin Accumulation in Dystrophic Microglia is an Early Event in the Development of Huntington's Disease,” Glia 55, no. 10 (2007): 1074-1084.

[247]

Y. Mi, X. Gao, H. Xu, et al., “The Emerging Roles of Ferroptosis in Huntington's Disease,” Neuromolecular Medicine 21, no. 2 (2019): 110-119.

[248]

J. Chen, E. Marks, B. Lai, et al., “Iron Accumulates in Huntington's Disease Neurons: Protection by Deferoxamine,” PLoS ONE 8, no. 10 (2013): e77023.

[249]

U. P. Shirendeb, M. J. Calkins, M. Manczak, et al., “Mutant Huntingtin's Interaction With Mitochondrial Protein Drp1 Impairs Mitochondrial Biogenesis and Causes Defective Axonal Transport and Synaptic Degeneration in Huntington's Disease,” Human Molecular Genetics 21, no. 2 (2012): 406-420.

[250]

R. Skouta, S. J. Dixon, J. Wang, et al., “Ferrostatins Inhibit Oxidative Lipid Damage and Cell Death in Diverse Disease Models,” Journal of the American Chemical Society 136, no. 12 (2014): 4551-4556.

[251]

L. Wang, R. Xu, C. Huang, et al., “Targeting the Ferroptosis Crosstalk: Novel Alternative Strategies for the Treatment of Major Depressive Disorder,” General Psychiatry 36, no. 5 (2023): e101072.

[252]

P. K. Maurya, C. Noto, L. B. Rizzo, et al., “The Role of Oxidative and Nitrosative Stress in Accelerated Aging and Major Depressive Disorder,” Progress in Neuro-Psychopharmacology & Biological Psychiatry 65 (2016): 134-144.

[253]

M. Jazvinšćak Jembrek, N. Oršolić, D. Karlović, and V. Peitl, “Flavonols in Action: Targeting Oxidative Stress and Neuroinflammation in Major Depressive Disorder,” International Journal of Molecular Sciences 24, no. 8 (2023): 6888.

[254]

Y. Mu, Y. Mei, Y. Chen, et al., “Perisciatic Nerve Dexmedetomidine Alleviates Spinal Oxidative Stress and Improves Peripheral Mitochondrial Dynamic Equilibrium in a Neuropathic Pain Mouse Model in an AMPK-Dependent Manner,” Disease Markers 2022 (2022): 6889676.

[255]

Y. Huang, X. Zhang, Y. Zou, et al., “Quercetin Ameliorates Neuropathic Pain After Brachial Plexus Avulsion via Suppressing Oxidative Damage Through Inhibition of PKC/MAPK/NOX Pathway,” Current Neuropharmacology 21, no. 11 (2023): 2343-2361.

[256]

C. Dai, Y. Guo, and X. Chu, “Neuropathic Pain: The Dysfunction of Drp1, Mitochondria, and ROS Homeostasis,” Neurotoxicity Research 38, no. 3 (2020): 553-563.

[257]

L. Teixeira-Santos, A. Albino-Teixeira, and D. Pinho, “Neuroinflammation, Oxidative Stress and Their Interplay in Neuropathic Pain: Focus on Specialized pro-Resolving Mediators and NADPH Oxidase Inhibitors as Potential Therapeutic Strategies,” Pharmacological Research 162 (2020): 105280.

[258]

M. Nazıroğlu, D. M. Dikici, and S. Dursun, “Role of Oxidative Stress and Ca2⁺ Signaling on Molecular Pathways of Neuropathic Pain in Diabetes: Focus on TRP Channels,” Neurochemical Research 37, no. 10 (2012): 2065-2075.

[259]

P. Zhao, W. Shi, Y. Ye, et al., “Atox1 Protects Hippocampal Neurons After Traumatic Brain Injury via DJ-1 Mediated Anti-Oxidative Stress and Mitophagy,” Redox Biology 72 (2024): 103156.

[260]

A. Fesharaki-Zadeh, “Oxidative Stress in Traumatic Brain Injury,” International Journal of Molecular Sciences 23, no. 21 (2022): 13000.

[261]

D. Pavlovic, S. Pekic, M. Stojanovic, and V. Popovic, “Traumatic Brain Injury: Neuropathological, Neurocognitive and Neurobehavioral Sequelae,” Pituitary 22, no. 3 (2019): 270-282.

[262]

M. A. Ansari, K. N. Roberts, and S. W. Scheff, “A Time Course of Contusion-Induced Oxidative Stress and Synaptic Proteins in Cortex in a rat Model of TBI,” Journal of Neurotrauma 25, no. 5 (2008): 513-526.

[263]

X. Dong, L. Deng, Y. Su, et al., “Curcumin Alleviates Traumatic Brain Injury Induced by gas Explosion Through Modulating gut Microbiota and Suppressing the LPS/TLR4/MyD88/NF-κB Pathway,” Environmental Science and Pollution Research International 31, no. 1 (2024): 1094-1113.

[264]

T. Farkhondeh, S. Samarghandian, B. Roshanravan, and L. Peivasteh-Roudsari, “Impact of Curcumin on Traumatic Brain Injury and Involved Molecular Signaling Pathways,” Recent Patents on Food, Nutrition & Agriculture 11, no. 2 (2020): 137-144.

[265]

M. Yu, Z. Wang, D. Wang, et al., “Oxidative Stress Following Spinal Cord Injury: From Molecular Mechanisms to Therapeutic Targets,” Journal of Neuroscience Research 101, no. 10 (2023): 1538-1554.

[266]

S. Fakhri, F. Abbaszadeh, S. Z. Moradi, et al., “Effects of Polyphenols on Oxidative Stress, Inflammation, and Interconnected Pathways During Spinal Cord Injury,” Oxidative Medicine and Cellular Longevity 2022 (2022): 8100195.

[267]

T. Huang, J. Shen, B. Bao, et al., “Mitochondrial-Targeting Antioxidant MitoQ Modulates Angiogenesis and Promotes Functional Recovery After Spinal Cord Injury,” Brain Research 1786 (2022): 147902.

[268]

Z. He, C. Zhang, J. Liang, et al., “Targeting Mitochondrial Oxidative Stress: Potential Neuroprotective Therapy for Spinal Cord Injury,” Journal of Integrative Neuroscience 22, no. 6 (2023): 153.

[269]

M. A. Nieto, R. Y. Huang, R. A. Jackson, and J. P. Thiery, “EMT: 2016,” Cell 166, no. 1 (2016): 21-45.

[270]

M. Fedele, R. Sgarra, S. Battista, L. Cerchia, and G. Manfioletti, “The Epithelial-Mesenchymal Transition at the Crossroads Between Metabolism and Tumor Progression,” International Journal of Molecular Sciences 23, no. 2 (2022): 800.

[271]

J. Lee, J. H. You, M. Kim, and J. Roh, “Epigenetic Reprogramming of Epithelial-Mesenchymal Transition Promotes Ferroptosis of Head and Neck Cancer,” Redox Biology 37 (2020): 101697.

[272]

Y. Ren, X. Mao, H. Xu, et al., “Ferroptosis and EMT: Key Targets for Combating Cancer Progression and Therapy Resistance,” Cellular and Molecular Life Sciences 80, no. 9 (2023): 263.

[273]

M. Wang, S. Li, Y. Wang, et al., “Gambogenic Acid Induces Ferroptosis in Melanoma Cells Undergoing Epithelial-To-Mesenchymal Transition,” Toxicology and Applied Pharmacology 401 (2020): 115110.

[274]

S. Ju, M. K. Singh, S. Han, et al., “Oxidative Stress and Cancer Therapy: Controlling Cancer Cells Using Reactive Oxygen Species,” International Journal of Molecular Sciences 25, no. 22 (2024): 12387.

[275]

G. Zhou, L. A. Dada, M. Wu, et al., “Hypoxia-Induced Alveolar Epithelial-Mesenchymal Transition Requires Mitochondrial ROS and Hypoxia-Inducible Factor 1,” American Journal of Physiology. Lung Cellular and Molecular Physiology 297, no. 6 (2009): L1120-L1130.

[276]

L. Varisli, V. Tolan, “Increased ROS Alters E-/N-Cadherin Levels and Promotes Migration in Prostate Cancer Cells,” Bratislavske Lekarske Listy 123, no. 10 (2022): 752-757.

[277]

D. Qiu, S. Song, N. Chen, et al., “NQO1 Alleviates Renal Fibrosis by Inhibiting the TLR4/NF-κB and TGF-β/Smad Signaling Pathways in Diabetic Nephropathy,” Cell. Signalling 108 (2023): 110712.

[278]

G. Shu, A. Yusuf, C. Dai, H. Sun, and X. Deng, “Piperine Inhibits AML-12 Hepatocyte EMT and LX-2 HSC Activation and Alleviates Mouse Liver Fibrosis Provoked by CCl(4): Roles in the Activation of the Nrf2 Cascade and Subsequent Suppression of the TGF-β1/Smad Axis,” Food and Function 12, no. 22 (2021): 11686-11703.

[279]

D. F. Higgins, K. Kimura, W. M. Bernhardt, et al., “Hypoxia Promotes Fibrogenesis in Vivo via HIF-1 Stimulation of Epithelial-To-Mesenchymal Transition,” Journal of Clinical Investigation 117, no. 12 (2007): 3810-3820.

[280]

R. Chatterjee, J. Chatterjee, “ROS and Oncogenesis With Special Reference to EMT and Stemness,” European Journal of Cell Biology 99, no. 2-3 (2020): 151073.

[281]

J. Fan, D. Ren, J. Wang, et al., “Bruceine D Induces Lung Cancer Cell Apoptosis and Autophagy via the ROS/MAPK Signaling Pathway in Vitro and in Vivo,” Cell Death & Disease 11, no. 2 (2020): 126.

[282]

X. Su, Z. Shen, Q. Yang, et al., “Vitamin C Kills Thyroid Cancer Cells Through ROS-Dependent Inhibition of MAPK/ERK and PI3K/AKT Pathways via Distinct Mechanisms,” Theranostics 9, no. 15 (2019): 4461-4473.

[283]

B. Sun, P. Ding, Y. Song, et al., “FDX1 Downregulation Activates Mitophagy and the PI3K/AKT Signaling Pathway to Promote Hepatocellular Carcinoma Progression by Inducing ROS Production,” Redox Biology 75 (2024): 103302.

[284]

Y. Gong, S. Luo, P. Fan, et al., “Growth Hormone Activates PI3K/Akt Signaling and Inhibits ROS Accumulation and Apoptosis in Granulosa Cells of Patients With Polycystic Ovary Syndrome,” Reproductive Biology and Endocrinology [Electronic Resource]: RB&E 18, no. 1 (2020): 121.

[285]

T. Zhang, X. Zhu, H. Wu, et al., “Targeting the ROS/PI3K/AKT/HIF-1α/HK2 Axis of Breast Cancer Cells: Combined Administration of Polydatin and 2-Deoxy-D-Glucose,” Journal of Cellular and Molecular Medicine 23, no. 5 (2019): 3711-3723.

[286]

X. Zheng, W. Chen, J. Yi, et al., “Apolipoprotein C1 Promotes Glioblastoma Tumorigenesis by Reducing KEAP1/NRF2 and CBS-Regulated Ferroptosis,” Acta Pharmacologica Sinica 43, no. 11 (2022): 2977-2992.

[287]

Y. Lu, C. Zhu, Y. Ding, et al., “Cepharanthine, a Regulator of Keap1-Nrf2, Inhibits Gastric Cancer Growth Through Oxidative Stress and Energy Metabolism Pathway,” Cell Death Discovery 9, no. 1 (2023): 450.

[288]

Z. Li, R. Mo, J. Gong, et al., “Dihydrotanshinone I Inhibits Gallbladder Cancer Growth by Targeting the Keap1-Nrf2 Signaling Pathway and Nrf2 Phosphorylation,” Phytomedicine 129 (2024): 155661.

[289]

W. Ge, K. Zhao, X. Wang, et al., “iASPP Is an Antioxidative Factor and Drives Cancer Growth and Drug Resistance by Competing With Nrf2 for Keap1 Binding,” Cancer Cell 32, no. 5 (2017): 561-573. e6.

[290]

J. Zhou, T. Li, H. Chen, et al., “ADAMTS10 Inhibits Aggressiveness via JAK/STAT/c-Myc Pathway and Reprograms Macrophage to Create an Anti-Malignant Microenvironment in Gastric Cancer,” Gastric Cancer 25, no. 6 (2022): 1002-1016.

[291]

K. Gałczyńska, A. Węgierek-Ciuk, K. Durlik-Popińska, et al., “Copper(II) Complex With 1-Allylimidazole Induces G2/M Cell Cycle Arrest and Suppresses A549 Cancer Cell Growth by Attenuating Wnt, JAK-STAT, and TGF-β Signaling Pathways,” Journal of Inorganic Biochemistry 264 (2025): 112791.

[292]

R. Shekh, R. K. Tiwari, A. Ahmad, et al., “Ethanolic Extract of Coleus Aromaticus Leaves Impedes the Proliferation and Instigates Apoptotic Cell Death in Liver Cancer HepG2 Cells Through Repressing JAK/STAT Cascade,” Journal of Food Biochemistry 46, no. 10 (2022): e14368.

[293]

S. Bakrim, N. Benkhaira, I. Bourais, et al., “Health Benefits and Pharmacological Properties of Stigmasterol,” Antioxidants (Basel) 11, no. 10 (2022): 1912.

[294]

A. Vallée, Y. Lecarpentier, “Crosstalk Between Peroxisome Proliferator-Activated Receptor Gamma and the Canonical WNT/β-Catenin Pathway in Chronic Inflammation and Oxidative Stress During Carcinogenesis,” Frontiers in Immunology 9 (2018): 745.

[295]

J. Wang, X. Wang, X. Wang, et al., “Curcumin Inhibits the Growth via Wnt/β-Catenin Pathway in non-Small-Cell Lung Cancer Cells,” European Review for Medical and Pharmacological Sciences 22, no. 21 (2018): 7492-7499.

[296]

H. You, D. Wang, L. Wei, et al., “Deferoxamine Inhibits Acute Lymphoblastic Leukemia Progression Through Repression of ROS/HIF-1α, Wnt/β-Catenin, and p38MAPK/ERK Pathways,” Journal of Oncology 2022 (2022): 8281267.

[297]

H. Bo, Q. Wu, C. Zhu, et al., “PIEZO1 Acts as a Cancer Suppressor by Regulating the ROS/Wnt/β-Catenin Axis,” Thoracic Cancer 15, no. 12 (2024): 1007-1016.

[298]

X. Liu, Y. Liu, Z. Liu, et al., “CircMYH9 Drives Colorectal Cancer Growth by Regulating Serine Metabolism and Redox Homeostasis in a p53-Dependent Manner,” Molecular Cancer 20, no. 1 (2021): 114.

[299]

J. Cao, X. Liu, Y. Yang, et al., “Decylubiquinone Suppresses Breast Cancer Growth and Metastasis by Inhibiting Angiogenesis via the ROS/p53/BAI1 Signaling Pathway,” Angiogenesis 23, no. 3 (2020): 325-338.

[300]

L. Jiang, N. Kon, T. Li, et al., “Ferroptosis as a p53-Mediated Activity During Tumour Suppression,” Nature 520, no. 7545 (2015): 57-62.

[301]

P. Jin, X. Feng, C. Huang, et al., “Oxidative Stress and Cellular Senescence: Roles in Tumor Progression and Therapeutic Opportunities,” Medcomm-Oncology 3, no. 4 (2024): 44-52.

[302]

S. O. Ebrahimi, S. Reiisi, and S. Shareef, “miRNAs, Oxidative Stress, and Cancer: A Comprehensive and Updated Review,” Journal of Cellular Physiology 235, no. 11 (2020): 8812-8825.

[303]

C. Chang, S. Pauklin, “ROS and TGFβ: From Pancreatic Tumour Growth to Metastasis,” Journal of Experimental & Clinical Cancer Research 40, no. 1 (2021): 152.

[304]

A. T. Dharmaraja, “Role of Reactive Oxygen Species (ROS) in Therapeutics and Drug Resistance in Cancer and Bacteria,” Journal of Medicinal Chemistry 60, no. 8 (2017): 3221-3240.

[305]

Q. Cui, J. Wang, Y. G. Assaraf, et al., “Modulating ROS to Overcome Multidrug Resistance in Cancer,” Drug Resistance Updates 41 (2018): 1-25.

[306]

X. Zhou, B. An, Y. Lin, et al., “Molecular Mechanisms of ROS-Modulated Cancer Chemoresistance and Therapeutic Strategies,” Biomedicine & Pharmacotherapy 165 (2023): 115036.

[307]

Q. Ou, L. Cheng, Y. Chang, J. Liu, and S. Zhang, “Jianpi Jiedu Decoction Reverses 5-Fluorouracil Resistance in Colorectal Cancer by Suppressing the xCT/GSH/GPX4 Axis to Induce Ferroptosis,” Heliyon 10, no. 5 (2024): e27082.

[308]

X. Yan, Y. Liu, C. Li, et al., “Pien-Tze-Huang Prevents Hepatocellular Carcinoma by Inducing Ferroptosis via Inhibiting SLC7A11-GSH-GPX4 Axis,” Cancer Cell International 23, no. 1 (2023): 109.

[309]

M. Zhang, H. Zheng, X. Zhu, et al., “Synchronously Evoking Disulfidptosis and Ferroptosis via Systematical Glucose Deprivation Targeting SLC7A11/GSH/GPX4 Antioxidant Axis,” ACS Nano 19, no. 14 (2025): 14233-14248.

[310]

P. Koppula, L. Zhuang, and B. Gan, “Cystine Transporter SLC7A11/xCT in Cancer: Ferroptosis, Nutrient Dependency, and Cancer Therapy,” Protein Cell 12, no. 8 (2021): 599-620.

[311]

M. Lei, Y. Zhang, F. Huang, et al., “Gankyrin Inhibits Ferroptosis Through the p53/SLC7A11/GPX4 Axis in Triple-Negative Breast Cancer Cells,” Scientific Reports 13, no. 1 (2023): 21916.

[312]

R. Kang, G. Kroemer, and D. Tang, “The Tumor Suppressor Protein p53 and the Ferroptosis Network,” Free Radical Biology and Medicine 133 (2019): 162-168.

[313]

P. Liao, W. Wang, W. Wang, et al., “CD8(+) T Cells and Fatty Acids Orchestrate Tumor Ferroptosis and Immunity via ACSL4,” Cancer Cell 40, no. 4 (2022): 365-378. e6.

[314]

L. Zhu, Y. Liao, and B. Jiang, “Role of ROS and Autophagy in the Pathological Process of Atherosclerosis,” Journal of Physiology and Biochemistry 80, no. 4 (2024): 743-756.

[315]

M. A. Gimbrone, G. Garcia-Cardena, “Endothelial Cell Dysfunction and the Pathobiology of Atherosclerosis,” Circulation Research 118, no. 4 (2016): 620-636.

[316]

M. Batty, M. R. Bennett, and E. Yu, “The Role of Oxidative Stress in Atherosclerosis,” Cells 11, no. 23 (2022): 3843.

[317]

K. Solanki, E. Bezsonov, A. Orekhov, et al., “Effect of Reactive Oxygen, Nitrogen, and Sulfur Species on Signaling Pathways in Atherosclerosis,” Vascular Pharmacology 154 (2024): 107282.

[318]

M. Ushio-Fukai, D. Ash, S. Nagarkoti, et al., “Interplay Between Reactive Oxygen/Reactive Nitrogen Species and Metabolism in Vascular Biology and Disease,” Antioxid Redox Signaling 34, no. 16 (2021): 1319-1354.

[319]

P. D. Reaven, “Mechanisms of Atherosclerosis: Role of LDL Oxidation,” Advances in Experimental Medicine and Biology 366 (1994): 113-128.

[320]

J. Pedro-Botet, E. Climent, and D. Benaiges, “LDL Cholesterol as a Causal Agent of Atherosclerosis,” Clinica e Investigacion En Arteriosclerosis: Publicacion Oficial De La Sociedad Espanola De Arteriosclerosis 36, no. Suppl 1 (2024): S3-S8.

[321]

P. Marchio, S. Guerra-Ojeda, J. M. Vila, et al., “Targeting Early Atherosclerosis: A Focus on Oxidative Stress and Inflammation,” Oxidative Medicine and Cellular Longevity 2019 (2019): 8563845.

[322]

S. Tavakoli, R. Asmis, “Reactive Oxygen Species and Thiol Redox Signaling in the Macrophage Biology of Atherosclerosis,” Antioxid Redox Signaling 17, no. 12 (2012): 1785-1795.

[323]

E. Galkina, K. Ley, “Immune and Inflammatory Mechanisms of Atherosclerosis,” Annual Review of Immunology 27 (2009): 165-197.

[324]

A. J. Kattoor, N. V. K. Pothineni, D. Palagiri, and J. L. Mehta, “Oxidative Stress in Atherosclerosis,” Current Atherosclerosis Reports 19, no. 11 (2017): 42.

[325]

C. Yeh, J. Wu, G. Lee, et al., “Vanadium Derivative Exposure Promotes Functional Alterations of VSMCs and Consequent Atherosclerosis via ROS/p38/NF-κB-Mediated IL-6 Production,” International Journal of Molecular Sciences 20, no. 24 (2019): 6115.

[326]

A. E. Vendrov, N. R. Madamanchi, X. Niu, et al., “NADPH Oxidases Regulate CD44 and Hyaluronic Acid Expression in Thrombin-Treated Vascular Smooth Muscle Cells and in Atherosclerosis,” Journal of Biological Chemistry 285, no. 34 (2010): 26545-26557.

[327]

Z. B. Wang, M. R. Castresana, and W. H. Newman, “Reactive Oxygen Species-Sensitive p38 MAPK Controls Thrombin-Induced Migration of Vascular Smooth Muscle Cells,” Journal of Molecular and Cellular Cardiology 36, no. 1 (2004): 49-56.

[328]

C. Chen, H. Li, Y. Leu, et al., “Corylin Inhibits Vascular Cell Inflammation, Proliferation and Migration and Reduces Atherosclerosis in ApoE-Deficient Mice,” Antioxidants (Basel) 9, no. 4 (2020): 275.

[329]

S. H. Yu, J. M. Yu, H. J. Yoo, et al., “Anti-Proliferative Effects of Rutin on OLETF Rat Vascular Smooth Muscle Cells Stimulated by Glucose Variability,” Yonsei Medical Journal 57, no. 2 (2016): 373-381.

[330]

H. M. A. Zeeshan, G. H. Lee, H. Kim, and H. Chae, “Endoplasmic Reticulum Stress and Associated ROS,” International Journal of Molecular Sciences 17, no. 3 (2016): 327.

[331]

C. D. Ochoa, R. F. Wu, and L. S. Terada, “ROS Signaling and ER Stress in Cardiovascular Disease,” Molecular Aspects of Medicine 63 (2018): 18-29.

[332]

L. Hang, Y. Peng, R. Xiang, X. Li, and Z. Li, “Ox-Ldl Causes Endothelial Cell Injury Through ASK1/NLRP3-Mediated Inflammasome Activation via Endoplasmic Reticulum Stress,” Drug Design, Development and Therapy 14 (2020): 731-743.

[333]

K. Malekmohammad, R. D. E. Sewell, and M. Rafieian-Kopaei, “Antioxidants and Atherosclerosis: Mechanistic Aspects,” Biomolecules 9, no. 8 (2019): 301.

[334]

A. van der Pol, W. H. van Gilst, A. A. Voors, and P. van der Meer, “Treating Oxidative Stress in Heart Failure: Past, Present and Future,” European Journal of Heart Failure 21, no. 4 (2019): 425-435.

[335]

K. F. Ayoub, N. V. K. Pothineni, J. Rutland, Z. Ding, and J. L. Mehta, “Immunity, Inflammation, and Oxidative Stress in Heart Failure: Emerging Molecular Targets,” Cardiovascular Drugs and Therapy 31, no. 5-6 (2017): 593-608.

[336]

K. Wróbel-Nowicka, C. Wojciechowska, W. Jacheć, M. Zalewska, and E. Romuk, “The Role of Oxidative Stress and Inflammatory Parameters in Heart Failure,” Medicina (Kaunas, Lithuania) 60, no. 5 (2024): 760.

[337]

A. Aimo, V. Castiglione, C. Borrelli, et al., “Oxidative Stress and Inflammation in the Evolution of Heart Failure: From Pathophysiology to Therapeutic Strategies,” European Journal of Preventive Cardiology 27, no. 5 (2020): 494-510.

[338]

S. Shi, Y. Chen, Z. Luo, G. Nie, and Y. Dai, “Role of Oxidative Stress and Inflammation-Related Signaling Pathways in Doxorubicin-Induced Cardiomyopathy,” Cell Communication and Signaling 21, no. 1 (2023): 61.

[339]

Y. Chen, S. Shi, and Y. Dai, “Research Progress of Therapeutic Drugs for Doxorubicin-Induced Cardiomyopathy,” Biomedicine & Pharmacotherapy 156 (2022): 113903.

[340]

L. Cheng, M. Zhu, X. Xu, et al., “AMPD3 Promotes Doxorubicin-Induced Cardiomyopathy Through HSP90a-Mediated a- Mediated Ferroptosis,” Iscience 27, no. 10 (2024): 111005.

[341]

D. Li, X. Liu, W. Pi, et al., “Fisetin Attenuates Doxorubicin-Induced Cardiomyopathy In Vivo and In Vitro by Inhibiting Ferroptosis Through SIRT1/Nrf2 Signaling Pathway Activation,” Frontiers in Pharmacology 12 (2022): 808480.

[342]

M. Zhang, X. Li, Z. Zhang, et al., “Elabela Blunts Doxorubicin-Induced Oxidative Stress and Ferroptosis in rat Aortic Adventitial Fibroblasts by Activating the KLF15/GPX4 Signaling,” Cell Stress & Chaperones 28, no. 1 (2023): 91-103.

[343]

W. Wu, Y. Cui, Y. Hong, et al., “Doxorubicin Cardiomyopathy is Ameliorated by Acacetin via Sirt1-Mediated Activation of AMPK/Nrf2 Signal Molecules,” Journal of Cellular and Molecular Medicine 24, no. 20 (2020): 12141-12153.

[344]

K. K. S. Nordgren, K. B. Wallace, “Disruption of the Keap1/Nrf2-Antioxidant Response System After Chronic Doxorubicin Exposure In Vivo,” Cardiovascular Toxicology 20, no. 6 (2020): 557-570.

[345]

Y. Wu, L. Zhang, Z. Wu, T. Shan, and C. Xiong, “Berberine Ameliorates Doxorubicin-Induced Cardiotoxicity via a SIRT1/p66Shc-Mediated Pathway,” Oxidative Medicine and Cellular Longevity 2019 (2019): 2150394.

[346]

Y. Liu, Y. Li, J. Ni, et al., “MiR-124 Attenuates Doxorubicin-Induced Cardiac Injury via Inhibiting p66Shc-Mediated Oxidative Stress,” Biochemical and Biophysical Research Communications 521, no. 2 (2020): 420-426.

[347]

Q. Liu, S. Wang, and L. Cai, “Diabetic Cardiomyopathy and its Mechanisms: Role of Oxidative Stress and Damage,” Journal of Diabetes Investigation 5, no. 6 (2014): 623-634.

[348]

Z. Chen, Z. Jin, J. Cai, et al., “Energy Substrate Metabolism and Oxidative Stress in Metabolic Cardiomyopathy,” Journal of Molecular Medicine (Berlin) 100, no. 12 (2022): 1721-1739.

[349]

M. Chen, X. Meng, Y. Han, et al., “Profile of Crosstalk Between Glucose and Lipid Metabolic Disturbance and Diabetic Cardiomyopathy: Inflammation and Oxidative Stress,” Front Endocrinol (Lausanne) 13 (2022): 983713.

[350]

I. D. Kyriazis, M. Hoffman, L. Gaignebet, et al., “KLF5 Is Induced by FOXO1 and Causes Oxidative Stress and Diabetic Cardiomyopathy,” Circulation Research 128, no. 3 (2021): 335-357.

[351]

D. Wang, J. Li, G. Luo, et al., “Nox4 as a Novel Therapeutic Target for Diabetic Vascular Complications,” Redox Biology 64 (2023): 102781.

[352]

S. Cadenas, “ROS and Redox Signaling in Myocardial Ischemia-Reperfusion Injury and Cardioprotection,” Free Radical Biology and Medicine 117 (2018): 76-89.

[353]

T. Hao, M. Qian, Y. Zhang, et al., “An Injectable Dual-Function Hydrogel Protects Against Myocardial Ischemia/Reperfusion Injury by Modulating ROS/NO Disequilibrium,” Advanced Science (Weinheim) 9, no. 15 (2022): e2105408.

[354]

P. Huang, C. Qu, Z. Rao, D. Wu, and J. Zhao, “Bidirectional Regulation Mechanism of TRPM2 Channel: Role in Oxidative Stress, Inflammation and Ischemia-Reperfusion Injury,” Frontiers in immunology 15 (2024): 1391355.

[355]

M. Wu, G. Yiang, W. Liao, et al., “Current Mechanistic Concepts in Ischemia and Reperfusion Injury,” Cellular Physiology and Biochemistry 46, no. 4 (2018): 1650-1667.

[356]

G. M. Pieper, V. Nilakantan, T. K. Nguyen, et al., “Reactive Oxygen and Reactive Nitrogen as Signaling Molecules for Caspase 3 Activation in Acute Cardiac Transplant Rejection,” Antioxid Redox Signaling 10, no. 6 (2008): 1031-1040.

[357]

C. Kaltenmeier, R. Wang, B. Popp, et al., “Role of Immuno-Inflammatory Signals in Liver Ischemia-Reperfusion Injury,” Cells 11, no. 14 (2022): 2222.

[358]

S. Xu, X. Han, X. Wang, et al., “The Role of Oxidative Stress in Aortic Dissection: A Potential Therapeutic Target,” Frontiers in Cardiovascular Medicine 11 (2024): 1410477.

[359]

W. Zeng, Y. Liang, S. Huang, et al., “Ciprofloxacin Accelerates Angiotensin-Ii-Induced Vascular Smooth Muscle Cells Senescence Through Modulating AMPK/ROS Pathway in Aortic Aneurysm and Dissection,” Cardiovascular Toxicology 24, no. 9 (2024): 889-903.

[360]

L. Xia, C. Sun, H. Zhu, et al., “Melatonin Protects Against Thoracic Aortic Aneurysm and Dissection Through SIRT1-Dependent Regulation of Oxidative Stress and Vascular Smooth Muscle Cell Loss,” Journal of Pineal Research 69, no. 1 (2020): e12661.

[361]

L. Qiu, S. Yi, T. Yu, and Y. Hao, “Sirt3 Protects Against Thoracic Aortic Dissection Formation by Reducing Reactive Oxygen Species, Vascular Inflammation, and Apoptosis of Smooth Muscle Cells,” Frontiers in Cardiovascular Medicine 8 (2021): 675647.

[362]

S. Zhou, B. Ma, and M. Luo, “Matrix Metalloproteinases in Aortic Dissection,” Vascular Pharmacology 156 (2024): 107420.

[363]

L. Zhang, C. Wang, Z. Xi, D. Li, and Z. Xu, “Mercaptoethanol Protects the Aorta From Dissection by Inhibiting Oxidative Stress, Inflammation, and Extracellular Matrix Degeneration in a Mouse Model,” Medical Science Monitor 24 (2018): 1802-1812.

[364]

K. M. Wales, K. Kavazos, M. Nataatmadja, et al., “N-3 PUFAs Protect Against Aortic Inflammation and Oxidative Stress in Angiotensin II-Infused Apolipoprotein E-/- Mice,” PLoS ONE 9, no. 11 (2014): e112816.

[365]

K. Zhao, H. Zhu, X. He, et al., “Senkyunolide I Ameliorates Thoracic Aortic Aneurysm and Dissection in Mice via Inhibiting the Oxidative Stress and Apoptosis of Endothelial Cells,” Biochimica et Biophysica Acta: Molecular Basis of Disease 1869, no. 7 (2023): 166819.

[366]

X. Xu, Y. Yu, M. Ling, et al., “Oxidative Stress and Mitochondrial Damage in Lambda-Cyhalothrin Toxicity: A Comprehensive Review of Antioxidant Mechanisms,” Environmental Pollution 338 (2023): 122694.

[367]

L. Zhang, L. Yang, Y. Luo, L. Dong, and F. Chen, “Acrylamide-Induced Hepatotoxicity Through Oxidative Stress: Mechanisms and Interventions,” Antioxid Redox Signaling 38, no. 16-18 (2023): 1122-1137.

[368]

W. W. Yew, K. C. Chang, and D. P. Chan, “Oxidative Stress and First-Line Antituberculosis Drug-Induced Hepatotoxicity,” Antimicrobial Agents and Chemotherapy 62, no. 8 (2018): e02637.

[369]

S. Wang, J. Bao, J. Li, et al., “Fraxinellone Induces Hepatotoxicity in Zebrafish Through Oxidative Stress and the Transporters Pathway,” Molecules (Basel, Switzerland) 27, no. 9 (2022): 2647.

[370]

Y. M. Yang, Y. E. Cho, and S. Hwang, “Crosstalk Between Oxidative Stress and Inflammatory Liver Injury in the Pathogenesis of Alcoholic Liver Disease,” International Journal of Molecular Sciences 23, no. 2 (2022): 774.

[371]

H. Aghara, P. Chadha, D. Zala, and P. Mandal, “Stress Mechanism Involved in the Progression of Alcoholic Liver Disease and the Therapeutic Efficacy of Nanoparticles,” Frontiers in Immunology 14 (2023): 1205821.

[372]

S. Liu, I. Tsai, and Y. Hsu, “Alcohol-Related Liver Disease: Basic Mechanisms and Clinical Perspectives,” International Journal of Molecular Sciences 22, no. 10 (2021): 5170.

[373]

W. Lai, J. Zhang, J. Sun, et al., “Oxidative Stress in Alcoholic Liver Disease, Focusing on Proteins, Nucleic Acids, and Lipids: A Review,” International Journal of Biological Macromolecules 278, no. Pt 3 (2024): 134809.

[374]

S. Li, H. Tan, N. Wang, et al., “The Role of Oxidative Stress and Antioxidants in Liver Diseases,” International Journal of Molecular Sciences 16, no. 11 (2015): 26087-26124.

[375]

J. Bai, B. Qian, T. Cai, et al., “Aloin Attenuates Oxidative Stress, Inflammation, and CCl(4)-Induced Liver Fibrosis in Mice: Possible Role of TGF-β/Smad Signaling,” Journal of Agricultural and Food Chemistry 71, no. 49 (2023): 19475-19487.

[376]

D. Dhar, J. Baglieri, T. Kisseleva, and D. A. Brenner, “Mechanisms of Liver Fibrosis and its Role in Liver Cancer,” Experimental Biology and Medicine (Maywood, N.J.) 245, no. 2 (2020): 96-108.

[377]

N. Roehlen, E. Crouchet, and T. F. Baumert, “Liver Fibrosis: Mechanistic Concepts and Therapeutic Perspectives,” Cells 9, no. 4 (2020): 875.

[378]

J. Liu, K. Man, “Mechanistic Insight and Clinical Implications of Ischemia/Reperfusion Injury Post Liver Transplantation,” Cellular and Molecular Gastroenterology and Hepatology 15, no. 6 (2023): 1463-1474.

[379]

S. Shi, L. Wang, L. J. W. van der Laan, Q. Pan, and M. M. A. Verstegen, “Mitochondrial Dysfunction and Oxidative Stress in Liver Transplantation and Underlying Diseases: New Insights and Therapeutics,” Transplantation 105, no. 11 (2021): 2362-2373.

[380]

N. Alva, A. Panisello-Roselló, M. Flores, J. Roselló-Catafau, and T. Carbonell, “Ubiquitin-Proteasome System and Oxidative Stress in Liver Transplantation,” World Journal of Gastroenterology 24, no. 31 (2018): 3521-3530.

[381]

S. Wang, Y. Xu, Y. Weng, et al., “Astilbin Ameliorates Cisplatin-Induced Nephrotoxicity Through Reducing Oxidative Stress and Inflammation,” Food and Chemical Toxicology 114 (2018): 227-236.

[382]

W. Dong, J. Wan, H. Yu, et al., “Nrf2 Protects Against Methamphetamine-Induced Nephrotoxicity by Mitigating Oxidative Stress and Autophagy in Mice,” Toxicology Letters 384 (2023): 136-148.

[383]

E. Dil, A. Topcu, T. Mercantepe, et al., “Agomelatine on Cisplatin-Induced Nephrotoxicity via Oxidative Stress and Apoptosis,” Naunyn-Schmiedebergs Archives of Pharmacology 396, no. 10 (2023): 2753-2764.

[384]

L. Tang, J. Yu, S. Zhuge, et al., “Oxidative Stress and Cx43-Mediated Apoptosis are Involved in PFOS-Induced Nephrotoxicity,” Toxicology 478 (2022): 153283.

[385]

Q. Wu, X. Wang, E. Nepovimova, et al., “Mechanism of Cyclosporine A Nephrotoxicity: Oxidative Stress, Autophagy, and Signalings,” Food and Chemical Toxicology 118 (2018): 889-907.

[386]

P. Laorodphun, R. Cherngwelling, A. Panya, and P. Arjinajarn, “Curcumin Protects Rats Against Gentamicin-Induced Nephrotoxicity by Amelioration of Oxidative Stress, Endoplasmic Reticulum Stress and Apoptosis,” Pharmaceutical Biology 60, no. 1 (2022): 491-500.

[387]

M. O. Cumaoglu, M. Makav, S. Dag, et al., “Combating Oxidative Stress and Inflammation in Gentamicin-Induced Nephrotoxicity Using Hydrogen-Rich Water,” Tissue & Cell 91 (2024): 102604.

[388]

S. Qu, C. Dai, F. Lang, et al., “Rutin Attenuates Vancomycin-Induced Nephrotoxicity by Ameliorating Oxidative Stress, Apoptosis, and Inflammation in Rats,” Antimicrobial Agents and Chemotherapy 63, no. 1 (2019): e01545.

[389]

N. Akaras, C. Gur, S. Kucukler, and F. M. Kandemir, “Zingerone Reduces Sodium Arsenite-Induced Nephrotoxicity by Regulating Oxidative Stress, Inflammation, Apoptosis and Histopathological Changes,” Chemico-Biological Interactions 374 (2023): 110410.

[390]

J. C. Jha, C. Banal, B. S. M. Chow, M. E. Cooper, and K. Jandeleit-Dahm, “Diabetes and Kidney Disease: Role of Oxidative Stress,” Antioxid Redox Signaling 25, no. 12 (2016): 657-684.

[391]

N. Samsu, “Diabetic Nephropathy: Challenges in Pathogenesis, Diagnosis, and Treatment,” BioMed Research International 2021 (2021): 1497449.

[392]

M. Darenskaya, S. Kolesnikov, N. Semenova, and L. Kolesnikova, “Diabetic Nephropathy: Significance of Determining Oxidative Stress and Opportunities for Antioxidant Therapies,” International Journal of Molecular Sciences 24, no. 15 (2023): 12378.

[393]

J. A. Østergaard, M. E. Cooper, and K. A. M. Jandeleit-Dahm, “Targeting Oxidative Stress and Anti-Oxidant Defence in Diabetic Kidney Disease,” Journal of Nephrology 33, no. 5 (2020): 917-929.

[394]

Q. Jin, T. Liu, Y. Qiao, et al., “Oxidative Stress and Inflammation in Diabetic Nephropathy: Role of Polyphenols,” Frontiers in Immunology 14 (2023): 1185317.

[395]

X. Zhang, E. Agborbesong, and X. Li, “The Role of Mitochondria in Acute Kidney Injury and Chronic Kidney Disease and Its Therapeutic Potential,” International Journal of Molecular Sciences 22, no. 20 (2021): 11253.

[396]

L. K. Billingham, J. S. Stoolman, K. Vasan, et al., “Mitochondrial Electron Transport Chain is Necessary for NLRP3 Inflammasome Activation,” Nature Immunology 23, no. 5 (2022): 692-704.

[397]

H. Liu, X. Wu, J. Luo, et al., “Pterostilbene Attenuates Astrocytic Inflammation and Neuronal Oxidative Injury After Ischemia-Reperfusion by Inhibiting NF-κB Phosphorylation,” Frontiers in Immunology 10 (2019): 2408.

[398]

S. Peerapornratana, C. L. Manrique-Caballero, H. Gómez, and J. A. Kellum, “Acute Kidney Injury From Sepsis: Current Concepts, Epidemiology, Pathophysiology, Prevention and Treatment,” Kidney International 96, no. 5 (2019): 1083-1099.

[399]

H. Gomez, C. Ince, D. De Backer, et al., “A Unified Theory of Sepsis-Induced Acute Kidney Injury: Inflammation, Microcirculatory Dysfunction, Bioenergetics, and the Tubular Cell Adaptation to Injury,” Shock (Augusta, Ga.) 41, no. 1 (2014): 3-11.

[400]

M. Mittal, M. R. Siddiqui, K. Tran, S. P. Reddy, and A. B. Malik, “Reactive Oxygen Species in Inflammation and Tissue Injury,” Antioxid Redox Signaling 20, no. 7 (2014): 1126-1167.

[401]

K. Hosohata, “Role of Oxidative Stress in Drug-Induced Kidney Injury,” International Journal of Molecular Sciences 17, no. 11 (2016): 1826.

[402]

M. E. Franco, G. E. Sutherland, and R. Lavado, “Xenobiotic Metabolism in the Fish Hepatic Cell Lines Hepa-E1 and RTH-149, and the Gill Cell Lines RTgill-W1 and G1B: Biomarkers of CYP450 Activity and Oxidative Stress,” Comparative Biochemistry and Physiology. Toxicology & Pharmacology 206-207 (2018): 32-40.

[403]

L. Su, J. Zhang, H. Gomez, J. A. Kellum, and Z. Peng, “Mitochondria ROS and Mitophagy in Acute Kidney Injury,” Autophagy 19, no. 2 (2023): 401-414.

[404]

Y. Wang, M. Pu, J. Yan, et al., “1,2-Bis(2-Aminophenoxy)ethane-N,N,N',N'-Tetraacetic Acid Acetoxymethyl Ester Loaded Reactive Oxygen Species Responsive Hyaluronic Acid-Bilirubin Nanoparticles for Acute Kidney Injury Therapy via Alleviating Calcium Overload Mediated Endoplasmic Reticulum Stress,” ACS Nano 17, no. 1 (2023): 472-491.

[405]

Y. Liu, H. Zhang, S. Duan, et al., “Mitofusin2 Ameliorated Endoplasmic Reticulum Stress and Mitochondrial Reactive Oxygen Species Through Maintaining Mitochondria-Associated Endoplasmic Reticulum Membrane Integrity in Cisplatin-Induced Acute Kidney Injury,” Antioxid Redox Signaling 40, no. 1-3 (2024): 16-39.

[406]

M. Wang, H. Xiong, H. Chen, Q. Li, and X. Z. Ruan, “Renal Injury by SARS-Cov-2 Infection: A Systematic Review,” Kidney Diseases (Basel) 7, no. 2 (2021): 100-110.

[407]

J. Foo, G. Bellot, S. Pervaiz, and S. Alonso, “Mitochondria-Mediated Oxidative Stress During Viral Infection,” Trends in Microbiology 30, no. 7 (2022): 679-692.

[408]

A. Srivastava, B. Tomar, D. Sharma, and S. K. Rath, “Mitochondrial Dysfunction and Oxidative Stress: Role in Chronic Kidney Disease,” Life Sciences 319 (2023): 121432.

[409]

H. Ho, H. Shirakawa, “Oxidative Stress and Mitochondrial Dysfunction in Chronic Kidney Disease,” Cells 12, no. 1 (2022): 88.

[410]

J. D. Malhotra, R. J. Kaufman, “Endoplasmic Reticulum Stress and Oxidative Stress: A Vicious Cycle or a Double-Edged Sword?” Antioxid Redox Signaling 9, no. 12 (2007): 2277-2293.

[411]

S. S. Cao, R. J. Kaufman, “Endoplasmic Reticulum Stress and Oxidative Stress in Cell Fate Decision and Human Disease,” Antioxid Redox Signaling 21, no. 3 (2014): 396-413.

[412]

A. V. Cybulsky, “Endoplasmic Reticulum Stress, the Unfolded Protein Response and Autophagy in Kidney Diseases,” Nature Reviews Nephrology 13, no. 11 (2017): 681-696.

[413]

K. L. Nowak, A. Jovanovich, H. Farmer-Bailey, et al., “Vascular Dysfunction, Oxidative Stress, and Inflammation in Chronic Kidney Disease,” Kidney360 1, no. 6 (2020): 501-509.

[414]

M. Muñoz, M. E. López-Oliva, C. Rodríguez, et al., “Differential Contribution of Nox1, Nox2 and Nox4 to Kidney Vascular Oxidative Stress and Endothelial Dysfunction in Obesity,” Redox Biology 28 (2020): 101330.

[415]

W. Feng, N. Zhu, Y. Xia, et al., “Melanin-Like Nanoparticles Alleviate Ischemia-Reperfusion Injury in the Kidney by Scavenging Reactive Oxygen Species and Inhibiting Ferroptosis,” Iscience 27, no. 4 (2024): 109504.

[416]

B. R. Stockwell, J. P. Friedmann Angeli, H. Bayir, et al., “Ferroptosis: A Regulated Cell Death Nexus Linking Metabolism, Redox Biology, and Disease,” Cell 171, no. 2 (2017): 273-285.

[417]

Y. Qi, M. Hu, Y. Qiu, et al., “Mitoglitazone Ameliorates Renal Ischemia/reperfusion Injury by Inhibiting Ferroptosis via Targeting MitoNEET,” Toxicology and Applied Pharmacology 465 (2023): 116440.

[418]

A. Rossi, A. Asthana, C. Riganti, et al., “Mitochondria Transplantation Mitigates Damage in an In Vitro Model of Renal Tubular Injury and in an Ex Vivo Model of DCD Renal Transplantation,” Annals of Surgery 278, no. 6 (2023): e1313-e1326.

[419]

Y. Chen, Z. Li, H. Zhang, et al., “Mitochondrial Metabolism and Targeted Treatment Strategies in Ischemic-Induced Acute Kidney Injury,” Cell Death Discovery 10, no. 1 (2024): 69.

[420]

S. Granata, V. Votrico, F. Spadaccino, et al., “Oxidative Stress and Ischemia/Reperfusion Injury in Kidney Transplantation: Focus on Ferroptosis, Mitophagy and New Antioxidants,” Antioxidants (Basel) 11, no. 4 (2022): 769.

[421]

M. Kwiatkowska, U. Oldakowska-Jedynak, E. Wojtaszek, T. Glogowski, and J. Malyszko, “Potential Effects of Immunosuppression on Oxidative Stress and Atherosclerosis in Kidney Transplant Recipients,” Oxidative Medicine and Cellular Longevity 2021 (2021): 6660846.

[422]

A. Tejani, L. Emmett, “Acute and Chronic Rejection,” Seminars in Nephrology 21, no. 5 (2001): 498-507.

[423]

Y. W. Ng, M. Singh, and M. M. Sarwal, “Antibody-Mediated Rejection in Pediatric Kidney Transplantation: Pathophysiology, Diagnosis, and Management,” Drugs 75, no. 5 (2015): 455-472.

[424]

M. Bruschi, G. Candiano, A. Petretto, et al., “Antibodies Against Anti-Oxidant Enzymes in Autoimmune Glomerulonephritis and in Antibody-Mediated Graft Rejection,” Antioxidants (Basel) 13, no. 12 (2024): 1519.

[425]

J. Ros, I. Matos, and J. Martin-Liberal, “Immunotherapy in Organ-Transplanted Cancer Patients: Efficacy and Risk of Organ Rejection,” Annals of Oncology 30, no. 7 (2019): 1173-1177.

[426]

X. Lai, X. Zheng, J. M. Mathew, et al., “Tackling Chronic Kidney Transplant Rejection: Challenges and Promises,” Frontiers in Immunology 12 (2021): 661643.

[427]

H. Yaribeygi, T. Sathyapalan, S. L. Atkin, and A. Sahebkar, “Molecular Mechanisms Linking Oxidative Stress and Diabetes Mellitus,” Oxidative Medicine and Cellular Longevity 2020 (2020): 8609213.

[428]

J. L. Rains, S. K. Jain, “Oxidative Stress, Insulin Signaling, and Diabetes,” Free Radical Biology and Medicine 50, no. 5 (2011): 567-575.

[429]

J. Wada, A. Nakatsuka, “Mitochondrial Dynamics and Mitochondrial Dysfunction in Diabetes,” Acta Medica Okayama 70, no. 3 (2016): 151-158.

[430]

W. H. Kim, J. W. Lee, B. Gao, and M. H. Jung, “Synergistic Activation of JNK/SAPK Induced by TNF-Alpha and IFN-Gamma: Apoptosis of Pancreatic Beta-Cells via the p53 and ROS Pathway,” Cell. Signalling 17, no. 12 (2005): 1516-1532.

[431]

D. L. Eizirik, M. L. Colli, and F. Ortis, “The Role of Inflammation in Insulitis and Beta-Cell Loss in Type 1 Diabetes,” Nature reviews Endocrinology 5, no. 4 (2009): 219-226.

[432]

D. J. Porte, S. E. Kahn, “beta-Cell Dysfunction and Failure in Type 2 Diabetes: Potential Mechanisms,” Diabetes 50, no. Suppl 1 (2001): S160-S163.

[433]

C. Wysham, J. Shubrook, “Beta-Cell Failure in Type 2 Diabetes: Mechanisms, Markers, and Clinical Implications,” Postgraduate Medicine 132, no. 8 (2020): 676-686.

[434]

Y. Zhang, X. Fang, J. Wei, et al., “PDX-1: A Promising Therapeutic Target to Reverse Diabetes,” Biomolecules 12, no. 12 (2022): 1785.

[435]

K. Bennett, C. James, and K. Hussain, “Pancreatic β-Cell KATP Channels: Hypoglycaemia and Hyperglycaemia,” Reviews in Endocrine & Metabolic Disorders 11, no. 3 (2010): 157-163.

[436]

A. C. Codo, G. G. Davanzo, L. D. B. Monteiro, et al., “Elevated Glucose Levels Favor SARS-Cov-2 Infection and Monocyte Response Through a HIF-1α/Glycolysis-Dependent Axis,” Cell Metabolism 32, no. 3 (2020): 437-446. e5.

[437]

M. Guasch-Ferré, J. L. Santos, M. A. Martínez-González, et al., “Glycolysis/gluconeogenesis- And Tricarboxylic Acid Cycle-Related Metabolites, Mediterranean Diet, and Type 2 Diabetes,” American Journal of Clinical Nutrition 111, no. 4 (2020): 835-844.

[438]

F. A. Alhumaydhi, D. de O Lopes, D. L. Bordin, et al., “Alkyladenine DNA Glycosylase Deficiency Uncouples Alkylation-Induced Strand Break Generation From PARP-1 Activation and Glycolysis Inhibition,” Scientific Reports 10, no. 1 (2020): 2209.

[439]

D. H. Hu, J. Peng, X. Zhang, et al., “Thyroid Hormone Exacerbates Vasoconstriction in Insulin Resistance: The Role of ONOO(-),” European Journal of Pharmacology 730 (2014): 41-50.

[440]

G. Pilon, A. Charbonneau, P. J. White, et al., “Endotoxin Mediated-Inos Induction Causes Insulin Resistance via ONOO⁻ Induced Tyrosine Nitration of IRS-1 in Skeletal Muscle,” PLoS ONE 5, no. 12 (2010): e15912.

[441]

S. K. Masenga, L. S. Kabwe, M. Chakulya, and A. Kirabo, “Mechanisms of Oxidative Stress in Metabolic Syndrome,” International Journal of Molecular Sciences 24, no. 9 (2023): 7898.

[442]

P. V. Dludla, B. B. Nkambule, B. Jack, et al., “Inflammation and Oxidative Stress in an Obese State and the Protective Effects of Gallic Acid,” Nutrients 11, no. 1 (2018): 23.

[443]

V. Rani, G. Deep, R. K. Singh, K. Palle, and U. C. S. Yadav, “Oxidative Stress and Metabolic Disorders: Pathogenesis and Therapeutic Strategies,” Life Sciences 148 (2016): 183-193.

[444]

J. Kim, X. Jia, P. D. Buckett, et al., “Iron Loading Impairs Lipoprotein Lipase Activity and Promotes Hypertriglyceridemia,” Faseb Journal 27, no. 4 (2013): 1657-1663.

[445]

X. Wu, J. Pan, J. J. Yu, et al., “DiDang Decoction Improves Mitochondrial Function and Lipid Metabolism via the HIF-1 Signaling Pathway to Treat Atherosclerosis and Hyperlipidemia,” Journal of Ethnopharmacology 308 (2023): 116289.

[446]

X. Jin, W. Fu, J. Zhou, et al., “Oxymatrine Attenuates Oxidized low‑density Lipoprotein‑induced HUVEC Injury by Inhibiting NLRP3 Inflammasome‑mediated Pyroptosis via the Activation of the SIRT1/Nrf2 Signaling Pathway,” International Journal of Molecular Medicine 48, no. 4 (2021): 187.

[447]

Z. Chen, R. Tian, Z. She, J. Cai, and H. Li, “Role of Oxidative Stress in the Pathogenesis of Nonalcoholic Fatty Liver Disease,” Free Radical Biology and Medicine 152 (2020): 116-141.

[448]

G. Paradies, V. Paradies, F. M. Ruggiero, and G. Petrosillo, “Oxidative Stress, Cardiolipin and Mitochondrial Dysfunction in Nonalcoholic Fatty Liver Disease,” World Journal of Gastroenterology 20, no. 39 (2014): 14205-14218.

[449]

L. Zhu, A. Wang, P. Luo, et al., “Mindin/Spondin 2 Inhibits Hepatic Steatosis, Insulin Resistance, and Obesity via Interaction With Peroxisome Proliferator-Activated Receptor α in Mice,” Journal of Hepatology 60, no. 5 (2014): 1046-1054.

[450]

B. Kim, M. Woo, C. Park, et al., “Hovenia Dulcis Extract Reduces Lipid Accumulation in Oleic Acid-Induced Steatosis of Hep G2 Cells via Activation of AMPK and PPARα/CPT-1 Pathway and in Acute Hyperlipidemia Mouse Model,” Phytotherapy Research 31, no. 1 (2017): 132-139.

[451]

C. M. Quiñonez-Flores, S. A. González-Chávez, D. Del Río Nájera, and C. Pacheco-Tena, “Oxidative Stress Relevance in the Pathogenesis of the Rheumatoid Arthritis: A Systematic Review,” BioMed Research International 2016 (2016): 6097417.

[452]

A. Phull, B. Nasir, I. U. Haq, and S. J. Kim, “Oxidative Stress, Consequences and ROS Mediated Cellular Signaling in Rheumatoid Arthritis,” Chemico-Biological Interactions 281 (2018): 121-136.

[453]

A. M. Abdallah, A. H. A. Naiem, S. R. Abdelraheim, et al., “Pyrazole Derivatives Ameliorate Synovial Inflammation in Collagen-Induced Arthritis Mice Model via Targeting p38 MAPK and COX-2,” Naunyn-Schmiedebergs Archives of Pharmacology (2024).

[454]

A. H. Eid, A. M. Gad, E. M. Fikry, and H. H. Arab, “Venlafaxine and Carvedilol Ameliorate Testicular Impairment and Disrupted Spermatogenesis in Rheumatoid Arthritis by Targeting AMPK/ERK and PI3K/AKT/mTOR Pathways,” Toxicology and Applied Pharmacology 364 (2019): 83-96.

[455]

R. Kumar, Y. K. Gupta, S. Singh, and A. Patil, “Glorisa Superba Hydroalcoholic Extract From Tubers Attenuates Experimental Arthritis by Downregulating Inflammatory Mediators, and Phosphorylation of ERK/JNK,” Immunological Investigations 45, no. 7 (2016): 603-618.

[456]

I. Ikonomidis, S. Tzortzis, I. Andreadou, et al., “Increased Benefit of Interleukin-1 Inhibition on Vascular Function, Myocardial Deformation, and Twisting in Patients With Coronary Artery Disease and Coexisting Rheumatoid Arthritis,” Circulation: Cardiovascular Imaging 7, no. 4 (2014): 619-628.

[457]

S. Sakaguchi, N. Mikami, J. B. Wing, et al., “Regulatory T Cells and Human Disease,” Annual Review of Immunology 38 (2020): 541-566.

[458]

S. Sakaguchi, T. Yamaguchi, T. Nomura, and M. Ono, “Regulatory T Cells and Immune Tolerance,” Cell 133, no. 5 (2008): 775-787.

[459]

J. Tao, M. Cheng, J. Tang, et al., “Foxp3, Regulatory T Cell, and Autoimmune Diseases,” Inflammation 40, no. 1 (2017): 328-339.

[460]

J. B. Wing, A. Tanaka, and S. Sakaguchi, “Human FOXP3(+) Regulatory T Cell Heterogeneity and Function in Autoimmunity and Cancer,” Immunity 50, no. 2 (2019): 302-316.

[461]

J. Ma, W. Hu, Y. Liu, et al., “CD226 Maintains Regulatory T Cell Phenotype Stability and Metabolism by the mTOR/Myc Pathway Under Inflammatory Conditions,” Cell Reports 42, no. 10 (2023): 113306.

[462]

R. Vallion, J. Divoux, S. Glauzy, et al., “Regulatory T Cell Stability and Migration Are Dependent on mTOR,” Journal of Immunology 205, no. 7 (2020): 1799-1809.

[463]

P. Wang, Q. Zhang, L. Tan, et al., “The Regulatory Effects of mTOR Complexes in the Differentiation and Function of CD4(+) T Cell Subsets,” Journal of Immunology Research 2020 (2020): 3406032.

[464]

S. Cassano, V. Pucino, C. La Rocca, et al., “Leptin Modulates Autophagy in Human CD4+CD25- Conventional T Cells,” Metabolism 63, no. 10 (2014): 1272-1279.

[465]

N. Scheinfeld, V. A. Deleo, “Photosensitivity in Lupus Erythematosus,” Photodermatology, Photoimmunology & Photomedicine 20, no. 5 (2004): 272-279.

[466]

A. Kuhn, S. Beissert, “Photosensitivity in Lupus Erythematosus,” Autoimmunity 38, no. 7 (2005): 519-529.

[467]

A. Kuhn, J. Wenzel, and H. Weyd, “Photosensitivity, Apoptosis, and Cytokines in the Pathogenesis of Lupus Erythematosus: A Critical Review,” Clinical Reviews in Allergy & Immunology 47, no. 2 (2014): 148-162.

[468]

M. O. Johnson, M. M. Wolf, M. Z. Madden, et al., “Distinct Regulation of Th17 and Th1 Cell Differentiation by Glutaminase-Dependent Metabolism,” Cell 175, no. 7 (2018): 1780-1795. e19.

[469]

M. Pandit, M. Timilshina, and J. Chang, “LKB1-Pten Axis Controls Th1 and Th17 Cell Differentiation via Regulating mTORC1,” Journal of Molecular Medicine (Berlin) 99, no. 8 (2021): 1139-1150.

[470]

Q. Yang, X. Liu, Q. Liu, et al., “Roles of mTORC1 and mTORC2 in Controlling γδ T1 and γδ T17 Differentiation and Function,” Cell Death and Differentiation 27, no. 7 (2020): 2248-2262.

[471]

M. G. Danieli, E. Antonelli, E. Longhi, S. Gangemi, and A. Allegra, “The Role of Microbiota and Oxidative Stress Axis and the Impact of Intravenous Immunoglobulin in Systemic Lupus Erythematosus,” Autoimmunity Reviews 23, no. 9 (2024): 103607.

[472]

B. Zhang, M. Zeng, Q. Zhang, et al., “Ephedrae Herba Polysaccharides Inhibit the Inflammation of Ovalbumin Induced Asthma by Regulating Th1/Th2 and Th17/Treg Cell Immune Imbalance,” Molecular Immunology 152 (2022): 14-26.

[473]

S. Tan, Z. Zheng, T. Liu, et al., “Schisandrin B Induced ROS-Mediated Autophagy and Th1/Th2 Imbalance via Selenoproteins in Hepa1-6 Cells,” Frontiers in Immunology 13 (2022): 857069.

[474]

B. Richardson, L. Scheinbart, J. Strahler, et al., “Evidence for Impaired T Cell DNA Methylation in Systemic Lupus Erythematosus and Rheumatoid Arthritis,” Arthritis and Rheumatism 33, no. 11 (1990): 1665-1673.

[475]

D. P. Kadam, A. N. Suryakar, R. D. Ankush, C. Y. Kadam, and K. H. Deshpande, “Role of Oxidative Stress in Various Stages of Psoriasis,” Indian Journal of Clinical Biochemistry 25, no. 4 (2010): 388-392.

[476]

H. Nemati, R. Khodarahmi, M. Sadeghi, et al., “Antioxidant Status in Patients With Psoriasis,” Cell Biochemistry and Function 32, no. 3 (2014): 268-273.

[477]

K. Rutault, C. Alderman, B. M. Chain, and D. R. Katz, “Reactive Oxygen Species Activate Human Peripheral Blood Dendritic Cells,” Free Radical Biology and Medicine 26, no. 1-2 (1999): 232-238.

[478]

R. Singh, S. Koppu, P. O. Perche, and S. R. Feldman, “The Cytokine Mediated Molecular Pathophysiology of Psoriasis and Its Clinical Implications,” International Journal of Molecular Sciences 22, no. 23 (2021): 12793.

[479]

M. Kamata, Y. Tada, “Crosstalk: Keratinocytes and Immune Cells in Psoriasis,” Frontiers in Immunology 14 (2023): 1286344.

[480]

Z. Tang, K. Zhang, S. Lv, et al., “LncRNA MEG3 Suppresses PI3K/AKT/mTOR Signalling Pathway to Enhance Autophagy and Inhibit Inflammation in TNF-α-Treated Keratinocytes and Psoriatic Mice,” Cytokine 148 (2021): 155657.

[481]

K. Furue, T. Ito, G. Tsuji, T. Kadono, and M. Furue, “Psoriasis and the TNF/IL23/IL17 Axis,” Giornale Italiano Di Dermatologia E Venereologia 154, no. 4 (2019): 418-424.

[482]

Y. Kim, T. V. Byzova, “Oxidative Stress in Angiogenesis and Vascular Disease,” Blood 123, no. 5 (2014): 625-631.

[483]

D. Bai, X. Cheng, Q. Li, et al., “Eupatilin Inhibits Keratinocyte Proliferation and Ameliorates Imiquimod-Induced Psoriasis-Like Skin Lesions in Mice via the p38 MAPK/NF-κB Signaling Pathway,” Immunopharmacology and Immunotoxicology 45, no. 2 (2023): 133-139.

[484]

A. Mavropoulos, E. I. Rigopoulou, C. Liaskos, D. P. Bogdanos, and L. I. Sakkas, “The Role of p38 MAPK in the Aetiopathogenesis of Psoriasis and Psoriatic Arthritis,” Clinical & Developmental Immunology 2013 (2013): 569751.

[485]

Y. Pan, Y. You, L. Sun, et al., “The STING Antagonist H-151 Ameliorates Psoriasis via Suppression of STING/NF-κB-Mediated Inflammation,” British Journal of Pharmacology 178, no. 24 (2021): 4907-4922.

[486]

P. Lin, H. Shi, Y. Lu, and J. Lin, “Centella Asiatica Alleviates Psoriasis Through JAK/STAT3-Mediated Inflammation: An in Vitro and in Vivo Study,” Journal of Ethnopharmacology 317 (2023): 116746.

[487]

R. Pandey, M. Bakay, and H. Hakonarson, “SOCS-Jak-STAT Inhibitors and SOCS Mimetics as Treatment Options for Autoimmune Uveitis, Psoriasis, Lupus, and Autoimmune Encephalitis,” Frontiers in Immunology 14 (2023): 1271102.

[488]

M. Y. Ansari, N. Ahmad, and T. M. Haqqi, “Oxidative Stress and Inflammation in Osteoarthritis Pathogenesis: Role of Polyphenols,” Biomedicine & Pharmacotherapy 129 (2020): 110452.

[489]

J. Riegger, A. Schoppa, L. Ruths, M. Haffner-Luntzer, and A. Ignatius, “Oxidative Stress as a key Modulator of Cell Fate Decision in Osteoarthritis and Osteoporosis: A Narrative Review,” Cellular & Molecular Biology Letters 28, no. 1 (2023): 76.

[490]

S. Zhang, J. Xu, H. Si, et al., “The Role Played by Ferroptosis in Osteoarthritis: Evidence Based on Iron Dyshomeostasis and Lipid Peroxidation,” Antioxidants (Basel) 11, no. 9 (2022): 1668.

[491]

F. Werry, E. Mazur, L. F. H. Theyse, and F. Edlich, “Apoptosis Regulation in Osteoarthritis and the Influence of Lipid Interactions,” International Journal of Molecular Sciences 24, no. 17 (2023): 13028.

[492]

J. Bao, Z. Chen, L. Xu, L. Wu, and Y. Xiong, “Rapamycin Protects Chondrocytes Against IL-18-Induced Apoptosis and Ameliorates rat Osteoarthritis,” Aging (Albany NY) 12, no. 6 (2020): 5152-5167.

[493]

J. Lu, H. Zhang, J. Pan, et al., “Fargesin Ameliorates Osteoarthritis via Macrophage Reprogramming by Downregulating MAPK and NF-κB Pathways,” Arthritis Research & Therapy 23, no. 1 (2021): 142.

[494]

F. Zhou, J. Mei, X. Han, et al., “Kinsenoside Attenuates Osteoarthritis by Repolarizing Macrophages Through Inactivating NF-κB/MAPK Signaling and Protecting Chondrocytes,” Acta Pharmaceutica Sinica B 9, no. 5 (2019): 973-985.

[495]

A. Mohammadalipour, S. P. Dumbali, and P. L. Wenzel, “Mitochondrial Transfer and Regulators of Mesenchymal Stromal Cell Function and Therapeutic Efficacy,” Frontiers in Cell and Developmental Biology 8 (2020): 603292.

[496]

S. Paliwal, R. Chaudhuri, A. Agrawal, and S. Mohanty, “Regenerative Abilities of Mesenchymal Stem Cells Through Mitochondrial Transfer,” Journal of Biomedical Science 25, no. 1 (2018): 31.

[497]

I. V. Zhivodernikov, T. V. Kirichenko, Y. V. Markina, A. Y. Postnov, and A. M. Markin, “Molecular and Cellular Mechanisms of Osteoporosis,” International Journal of Molecular Sciences 24, no. 21 (2023): 15772.

[498]

Y. Guo, X. Jia, Y. Cui, et al., “Sirt3-Mediated Mitophagy Regulates AGEs-Induced BMSCs Senescence and Senile Osteoporosis,” Redox Biology 41 (2021): 101915.

[499]

T. Iantomasi, C. Romagnoli, G. Palmini, et al., “Oxidative Stress and Inflammation in Osteoporosis: Molecular Mechanisms Involved and the Relationship With MicroRNAs,” International Journal of Molecular Sciences 24, no. 4 (2023): 3772.

[500]

V. Domazetovic, G. Marcucci, T. Iantomasi, M. L. Brandi, and M. T. Vincenzini, “Oxidative Stress in Bone Remodeling: Role of Antioxidants,” Clinical Cases in Mineral and Bone Metabolism 14, no. 2 (2017): 209-216.

[501]

S. Hong, J. Lee, D. Seo, et al., “Euphorbia Factor L1 Inhibits Osteoclastogenesis by Regulating Cellular Redox Status and Induces Fas-Mediated Apoptosis in Osteoclast,” Free Radical Biology and Medicine 112 (2017): 191-199.

[502]

J. S. Kimball, J. P. Johnson, and D. A. Carlson, “Oxidative Stress and Osteoporosis,” Journal of Bone and Joint Surgery. American Volume 103, no. 15 (2021): 1451-1461.

[503]

X. Rong, Y. Kou, Y. Zhang, et al., “ED-71 Prevents Glucocorticoid-Induced Osteoporosis by Regulating Osteoblast Differentiation via Notch and Wnt/β-Catenin Pathways,” Drug Design, Development and Therapy 16 (2022): 3929-3946.

[504]

P. Llanos, J. Palomero, “Reactive Oxygen and Nitrogen Species (RONS) and Cytokines-Myokines Involved in Glucose Uptake and Insulin Resistance in Skeletal Muscle,” Cells 11, no. 24 (2022): 4008.

[505]

L. M. Leitner, R. J. Wilson, Z. Yan, and A. Gödecke, “Reactive Oxygen Species/Nitric Oxide Mediated Inter-Organ Communication in Skeletal Muscle Wasting Diseases,” Antioxid Redox Signaling 26, no. 13 (2017): 700-717.

[506]

M. J. Jackson, D. Pye, and J. Palomero, “The Production of Reactive Oxygen and Nitrogen Species by Skeletal Muscle,” Journal of Applied Physiology (1985) 102, no. 4 (2007): 1664-1670.

[507]

A. Espinosa, A. Leiva, M. Peña, et al., “Myotube Depolarization Generates Reactive Oxygen Species Through NAD(P)H Oxidase; ROS-Elicited Ca2+ Stimulates ERK, CREB, Early Genes,” Journal of Cellular Physiology 209, no. 2 (2006): 379-388.

[508]

G. Cherednichenko, A. V. Zima, W. Feng, et al., “NADH Oxidase Activity of rat Cardiac Sarcoplasmic Reticulum Regulates Calcium-Induced Calcium Release,” Circulation Research 94, no. 4 (2004): 478-486.

[509]

E. Marzetti, R. Calvani, M. Cesari, et al., “Mitochondrial Dysfunction and Sarcopenia of Aging: From Signaling Pathways to Clinical Trials,” International Journal of Biochemistry & Cell Biology 45, no. 10 (2013): 2288-2301.

[510]

L. Larsson, H. Degens, M. Li, et al., “Sarcopenia: Aging-Related Loss of Muscle Mass and Function,” Physiological Reviews 99, no. 1 (2019): 427-511.

[511]

X. Xu, Z. Wen, “The Mediating Role of Inflammaging Between Mitochondrial Dysfunction and Sarcopenia in Aging: A Review,” American Journal of Clinical and Experimental Immunology 12, no. 6 (2023): 109-126.

[512]

C. M. Lee, M. E. Lopez, R. Weindruch, and J. M. Aiken, “Association of age-Related Mitochondrial Abnormalities With Skeletal Muscle Fiber Atrophy,” Free Radical Biology and Medicine 25, no. 8 (1998): 964-972.

[513]

Y. Huang, B. Wu, D. Shen, et al., “Ferroptosis in a Sarcopenia Model of Senescence Accelerated Mouse Prone 8 (SAMP8),” International Journal of Biological Sciences 17, no. 1 (2021): 151-162.

[514]

Y. Wang, Z. Zhang, W. Jiao, et al., “Ferroptosis and its Role in Skeletal Muscle Diseases,” Frontiers in Molecular Biosciences 9 (2022): 1051866.

[515]

Y. Chen, Y. Zhang, S. Zhang, and H. Ren, “Molecular Insights Into Sarcopenia: Ferroptosis-Related Genes as Diagnostic and Therapeutic Targets,” Journal of Biomolecular Structure & Dynamics (2024): 1-19.

[516]

L. Ju, J. Diao, J. Zhang, et al., “Shenshuai Yingyang Jiaonang Ameliorates Chronic Kidney Disease-Associated Muscle Atrophy in Rats by Inhibiting Ferroptosis Mediated by the HIF-1α/SLC7A11 Pathway,” Heliyon 10, no. 8 (2024): e29093.

[517]

T. N. Stitt, D. Drujan, B. A. Clarke, et al., “The IGF-1/PI3K/Akt Pathway Prevents Expression of Muscle Atrophy-Induced Ubiquitin Ligases by Inhibiting FOXO Transcription Factors,” Molecular Cell 14, no. 3 (2004): 395-403.

[518]

S. Tabak, S. Schreiber-Avissar, and E. Beit-Yannai, “Crosstalk Between MicroRNA and Oxidative Stress in Primary Open-Angle Glaucoma,” International Journal of Molecular Sciences 22, no. 5 (2021): 2421.

[519]

A. Izzotti, A. Bagnis, and S. C. Saccà, “The Role of Oxidative Stress in Glaucoma,” Mutation Research 612, no. 2 (2006): 105-114.

[520]

L. Xu, X. Zhang, Y. Zhao, et al., “Metformin Protects Trabecular Meshwork Against Oxidative Injury via Activating Integrin/ROCK Signals,” Elife 12 (2023): e81198.

[521]

H. Wu, Y. Shui, Y. Liu, X. Liu, and C. J. Siegfried, “Trabecular Meshwork Mitochondrial Function and Oxidative Stress: Clues to Racial Disparities of Glaucoma,” Ophthalmology Science 2, no. 1 (2022): 100107.

[522]

T. Yang, E. Y. Kang, P. Lin, et al., “Mitochondria in Retinal Ganglion Cells: Unraveling the Metabolic Nexus and Oxidative Stress,” International Journal of Molecular Sciences 25, no. 16 (2024): 8626.

[523]

Z. Zeng, M. You, C. Fan, et al., “Pathologically High Intraocular Pressure Induces Mitochondrial Dysfunction Through Drp1 and Leads to Retinal Ganglion Cell PANoptosis in Glaucoma,” Redox Biology 62 (2023): 102687.

[524]

R. Fuchshofer, E. R. Tamm, “The Role of TGF-β in the Pathogenesis of Primary Open-Angle Glaucoma,” Cell and Tissue Research 347, no. 1 (2012): 279-290.

[525]

G. J. Villarreal, D. Oh, M. H. Kang, and D. J. Rhee, “Coordinated Regulation of Extracellular Matrix Synthesis by the MicroRNA-29 Family in the Trabecular Meshwork,” Investigative Ophthalmology & Visual Science 52, no. 6 (2011): 3391-3397.

[526]

C. Luna, G. Li, J. Qiu, D. L. Epstein, and P. Gonzalez, “Role of miR-29B on the Regulation of the Extracellular Matrix in Human Trabecular Meshwork Cells Under Chronic Oxidative Stress,” Molecular Vision 15 (2009): 2488-2497.

[527]

L. Cheng, K. Li, N. Yi, et al., “miRNA-141 Attenuates UV-Induced Oxidative Stress via Activating Keap1-Nrf2 Signaling in Human Retinal Pigment Epithelium Cells and Retinal Ganglion Cells,” Oncotarget 8, no. 8 (2017): 13186-13194.

[528]

Y. Wang, F. Li, and S. Wang, “MicroRNA‑93 is Overexpressed and Induces Apoptosis in Glaucoma Trabecular Meshwork Cells,” Molecular Medicine Reports 14, no. 6 (2016): 5746-5750.

[529]

L. Wang, X. Wei, “Exosome-Based Crosstalk in Glaucoma Pathogenesis: A Focus on Oxidative Stress and Neuroinflammation,” Frontiers in immunology 14 (2023): 1202704.

[530]

J. Liu, F. Jiang, Y. Jiang, et al., “Roles of Exosomes in Ocular Diseases,” International Journal of Nanomedicine 15 (2020): 10519-10538.

[531]

E. Kang, J. Wu, N. M. Gutierrez, et al., “Mitochondrial Replacement in Human Oocytes Carrying Pathogenic Mitochondrial DNA Mutations,” Nature 540, no. 7632 (2016): 270-275.

[532]

E. Jemt, Ö. Persson, Y. Shi, et al., “Regulation of DNA Replication at the end of the Mitochondrial D-Loop Involves the Helicase TWINKLE and a Conserved Sequence Element,” Nucleic Acids Res. 43, no. 19 (2015): 9262-9275.

[533]

M. Al-Shabrawey, S. Smith, “Prediction of Diabetic Retinopathy: Role of Oxidative Stress and Relevance of Apoptotic Biomarkers,” EPMA J 1, no. 1 (2010): 56-72.

[534]

S. J. Riedl, Y. Shi, “Molecular Mechanisms of Caspase Regulation During Apoptosis,” Nature Reviews Molecular Cell Biology 5, no. 11 (2004): 897-907.

[535]

M. Wu, G. Yiang, T. Lai, and C. Li, “The Oxidative Stress and Mitochondrial Dysfunction During the Pathogenesis of Diabetic Retinopathy,” Oxidative Medicine and Cellular Longevity 2018 (2018): 3420187.

[536]

M. Lorenzi, “The Polyol Pathway as a Mechanism for Diabetic Retinopathy: Attractive, Elusive, and Resilient,” Experimental Diabetes Research 2007 (2007): 61038.

[537]

C. Altmann, M. H. H. Schmidt, “The Role of Microglia in Diabetic Retinopathy: Inflammation, Microvasculature Defects and Neurodegeneration,” International Journal of Molecular Sciences 19, no. 1 (2018): 110.

[538]

R. E. Schmidt, D. A. Dorsey, L. N. Beaudet, et al., “A Potent Sorbitol Dehydrogenase Inhibitor Exacerbates Sympathetic Autonomic Neuropathy in Rats With Streptozotocin-Induced Diabetes,” Experimental Neurology 192, no. 2 (2005): 407-419.

[539]

L. Zheng, T. S. Kern, “Role of Nitric Oxide, Superoxide, Peroxynitrite and PARP in Diabetic Retinopathy,” Frontiers in Bioscience (Landmark Ed) 14, no. 10 (2009): 3974-3987.

[540]

D. R. Jones, W. Keune, K. E. Anderson, et al., “The Hexosamine Biosynthesis Pathway and O-Glcnacylation Maintain Insulin-Stimulated PI3K-PKB Phosphorylation and Tumour Cell Growth After Short-Term Glucose Deprivation,” Febs Journal 281, no. 16 (2014): 3591-3608.

[541]

X. Du, T. Matsumura, D. Edelstein, et al., “Inhibition of GAPDH Activity by Poly(ADP-Ribose) Polymerase Activates Three Major Pathways of Hyperglycemic Damage in Endothelial Cells,” Journal of Clinical Investigation 112, no. 7 (2003): 1049-1057.

[542]

M. Brownlee, “The Pathobiology of Diabetic Complications: A Unifying Mechanism,” Diabetes 54, no. 6 (2005): 1615-1625.

[543]

M. Capitão, R. Soares, “Angiogenesis and Inflammation Crosstalk in Diabetic Retinopathy,” Journal of Cellular Biochemistry 117, no. 11 (2016): 2443-2453.

[544]

C. Zhang, L. Gu, H. Xie, et al., “Glucose Transport, Transporters and Metabolism in Diabetic Retinopathy,” Biochimica et Biophysica Acta: Molecular Basis of Disease 1870, no. 3 (2024): 166995.

[545]

M. I. L. Gálvez, “Rubosixtaurin and Other PKC Inhibitors in Diabetic Retinopathy and Macular Edema,” Current Diabetes Reviews 5, no. 1 (2009): 14-17.

[546]

L. P. Aiello, “The Potential Role of PKC Beta in Diabetic Retinopathy and Macular Edema,” Survey of Ophthalmology 47, no. Suppl 2 (2002): S263-269.

[547]

W. Liang, H. Yang, H. Liu, W. Qian, and X. Chen, “HMGB1 Upregulates NF-Kb by Inhibiting IKB-α and Associates With Diabetic Retinopathy,” Life Sciences 241 (2020): 117146.

[548]

L. Tang, C. Zhang, L. Lu, et al., “Melatonin Maintains Inner Blood-Retinal Barrier by Regulating Microglia via Inhibition of PI3K/Akt/Stat3/NF-κB Signaling Pathways in Experimental Diabetic Retinopathy,” Frontiers in Immunology 13 (2022): 831660.

[549]

J. Peng, S. Xiong, L. Ding, J. Peng, and X. Xia, “Diabetic Retinopathy: Focus on NADPH Oxidase and its Potential as Therapeutic Target,” European Journal of Pharmacology 853 (2019): 381-387.

[550]

J. L. Wilkinson-Berka, I. Rana, R. Armani, and A. Agrotis, “Reactive Oxygen Species, Nox and Angiotensin II in Angiogenesis: Implications for Retinopathy,” Clinical Science (London, England: 1979) 124, no. 10 (2013): 597-615.

[551]

Y. Ruan, S. Jiang, and A. Gericke, “Age-Related Macular Degeneration: Role of Oxidative Stress and Blood Vessels,” International Journal of Molecular Sciences 22, no. 3 (2021): 1296.

[552]

Z. Zhang, X. Bao, Y. Cong, B. Fan, and G. Li, “Autophagy in Age-Related Macular Degeneration: A Regulatory Mechanism of Oxidative Stress,” Oxidative Medicine and Cellular Longevity 2020 (2020): 2896036.

[553]

J. Hanus, C. Anderson, and S. Wang, “RPE Necroptosis in Response to Oxidative Stress and in AMD,” Ageing Research Reviews 24, no. Pt B (2015): 286-298.

[554]

K. Kaarniranta, H. Uusitalo, J. Blasiak, et al., “Mechanisms of Mitochondrial Dysfunction and Their Impact on age-Related Macular Degeneration,” Progress in Retinal and Eye Research 79 (2020): 100858.

[555]

S. K. Mitter, C. Song, X. Qi, et al., “Dysregulated Autophagy in the RPE is Associated With Increased Susceptibility to Oxidative Stress and AMD,” Autophagy 10, no. 11 (2014): 1989-2005.

[556]

M. Nita, A. Grzybowski, “Interplay Between Reactive Oxygen Species and Autophagy in the Course of age-Related Macular Degeneration,” EXCLI journal 19 (2020): 1353-1371.

[557]

X. Wang, Y. Gao, Q. Yuan, and M. Zhang, “NLRP3 and Autophagy in Retinal Ganglion Cell Inflammation in age-Related Macular Degeneration: Potential Therapeutic Implications,” International Journal of Ophthalmology 17, no. 8 (2024): 1531-1544.

[558]

S. Zhang, B. Fan, Y. L. Li, Z. Zuo, and G. Li, “Oxidative Stress-Involved Mitophagy of Retinal Pigment Epithelium and Retinal Degenerative Diseases,” Cellular and Molecular Neurobiology 43, no. 7 (2023): 3265-3276.

[559]

L. You, H. Peng, J. Liu, et al., “Catalpol Protects ARPE-19 Cells Against Oxidative Stress via Activation of the Keap1/Nrf2/ARE Pathway,” Cells 10, no. 10 (2021): 2635.

[560]

S. Sajadimajd, M. Khazaei, “Oxidative Stress and Cancer: The Role of Nrf2,” Current Cancer Drug Targets 18, no. 6 (2018): 538-557.

[561]

S. Li, Y. Ma, S. Ye, et al., “Endogenous Hydrogen Sulfide Counteracts Polystyrene Nanoplastics-Induced Mitochondrial Apoptosis and Excessive Autophagy via Regulating Nrf2 and PGC-1α Signaling Pathway in Mouse Spermatocyte-Derived GC-2spd(ts) Cells,” Food and Chemical Toxicology 164 (2022): 113071.

[562]

P. Yang, K. Cheng, J. Huang, et al., “Sulforaphane Inhibits Blue Light-Induced Inflammation and Apoptosis by Upregulating the SIRT1/PGC-1α/Nrf2 Pathway and Autophagy in Retinal Pigment Epithelial Cells,” Toxicology and Applied Pharmacology 421 (2021): 115545.

[563]

R. R. Henkel, “Leukocytes and Oxidative Stress: Dilemma for Sperm Function and Male Fertility,” Asian Journal of Andrology 13, no. 1 (2011): 43-52.

[564]

N. B. Takalani, E. M. Monageng, K. Mohlala, et al., “Role of Oxidative Stress in Male Infertility,” Reproduction and Fertility 4, no. 3 (2023): e230024.

[565]

A. T. Alahmar, “Role of Oxidative Stress in Male Infertility: An Updated Review,” Journal of Human Reproductive Sciences 12, no. 1 (2019): 4-18.

[566]

R. J. Aitken, J. R. Drevet, “The Importance of Oxidative Stress in Determining the Functionality of Mammalian Spermatozoa: A Two-Edged Sword,” Antioxidants (Basel) 9, no. 2 (2020): 111.

[567]

K. Nowicka-Bauer, B. Nixon, “Molecular Changes Induced by Oxidative Stress That Impair Human Sperm Motility,” Antioxidants (Basel) 9, no. 2 (2020): 134.

[568]

M. B. Hosen, M. R. Islam, F. Begum, Y. Kabir, and M. Z. H. Howlader, “Oxidative Stress Induced Sperm DNA Damage, a Possible Reason for Male Infertility,” Iranian Journal of Reproductive Medicine 13, no. 9 (2015): 525-532.

[569]

C. González-Marín, J. Gosálvez, and R. Roy, “Types, Causes, Detection and Repair of DNA Fragmentation in Animal and Human Sperm Cells,” International Journal of Molecular Sciences 13, no. 11 (2012): 14026-14052.

[570]

L. Rashki Ghaleno, A. Alizadeh, J. R. Drevet, A. Shahverdi, and M. R. Valojerdi, “Oxidation of Sperm DNA and Male Infertility,” Antioxidants (Basel) 10, no. 1 (2021): 97.

[571]

A. Agarwal, G. Virk, C. Ong, and S. S. du Plessis, “Effect of Oxidative Stress on Male Reproduction,” World Journal of Men's Health 32, no. 1 (2014): 1-17.

[572]

P. Piomboni, R. Focarelli, A. Stendardi, A. Ferramosca, and V. Zara, “The Role of Mitochondria in Energy Production for Human Sperm Motility,” International Journal of Andrology 35, no. 2 (2012): 109-124.

[573]

A. Agarwal, A. Aponte-Mellado, B. J. Premkumar, A. Shaman, and S. Gupta, “The Effects of Oxidative Stress on Female Reproduction: A Review,” Reproductive Biology and Endocrinology [Electronic Resource]: RB&E 10 (2012): 49.

[574]

H. R. Behrman, P. H. Kodaman, S. L. Preston, and S. Gao, “Oxidative Stress and the Ovary,” Journal of the Society for Gynecologic Investigation 8, no. 1 Suppl Proceedings (2001): S40-S42.

[575]

J. Lu, Z. Wang, J. Cao, Y. Chen, and Y. Dong, “A Novel and Compact Review on the Role of Oxidative Stress in Female Reproduction,” Reproductive Biology and Endocrinology [Electronic Resource]: RB&E 16, no. 1 (2018): 80.

[576]

T. Fukai, M. Ushio-Fukai, “Superoxide Dismutases: Role in Redox Signaling, Vascular Function, and Diseases,” Antioxid Redox Signaling 15, no. 6 (2011): 1583-1606.

[577]

W. Verreth, D. De Keyzer, P. C. Davey, et al., “Rosuvastatin Restores Superoxide Dismutase Expression and Inhibits Accumulation of Oxidized LDL in the Aortic Arch of Obese Dyslipidemic Mice,” British Journal of Pharmacology 151, no. 3 (2007): 347-355.

[578]

B. Zhao, J. Peng, C. Chen, et al., “Innovative Engineering of Superoxide Dismutase for Enhanced Cardioprotective Biocatalysis in Myocardial Ischemia-Reperfusion Injury,” International Journal of Biological Macromolecules 286 (2025): 137656.

[579]

J. Feng, X. Chen, R. Liu, et al., “Melatonin Protects Against Myocardial Ischemia-Reperfusion Injury by Elevating Sirtuin3 Expression and Manganese Superoxide Dismutase Activity,” Free Radical Research 52, no. 8 (2018): 840-849.

[580]

K. Zhao, J. Tang, H. Xie, et al., “Nicotinamide Riboside Attenuates Myocardial Ischemia-Reperfusion Injury via Regulating SIRT3/SOD2 Signaling Pathway,” Biomedicine & Pharmacotherapy 175 (2024): 116689.

[581]

A. Furuta, D. L. Price, C. A. Pardo, et al., “Localization of Superoxide Dismutases in Alzheimer's Disease and Down's Syndrome Neocortex and Hippocampus,” American Journal of Pathology 146, no. 2 (1995): 357-367.

[582]

L. Esposito, J. Raber, L. Kekonius, et al., “Reduction in Mitochondrial Superoxide Dismutase Modulates Alzheimer's Disease-Like Pathology and Accelerates the Onset of Behavioral Changes in Human Amyloid Precursor Protein Transgenic Mice,” Journal of Neuroscience 26, no. 19 (2006): 5167-5179.

[583]

H. Lee, N. Pancholi, L. Esposito, et al., “Early Induction of Oxidative Stress in Mouse Model of Alzheimer Disease With Reduced Mitochondrial Superoxide Dismutase Activity,” PLoS ONE 7, no. 1 (2012): e28033.

[584]

P. A. Dionísio, J. D. Amaral, and C. M. P. Rodrigues, “Oxidative Stress and Regulated Cell Death in Parkinson's Disease,” Ageing Research Reviews 67 (2021): 101263.

[585]

A. Yoritaka, N. Hattori, H. Mori, K. Kato, and Y. Mizuno, “An Immunohistochemical Study on Manganese Superoxide Dismutase in Parkinson's Disease,” Journal of the Neurological Sciences 148, no. 2 (1997): 181-186.

[586]

J. Tomkins, S. J. Banner, C. J. McDermott, and P. J. Shaw, “Mutation Screening of Manganese Superoxide Dismutase in Amyotrophic Lateral Sclerosis,” Neuroreport 12, no. 11 (2001): 2319-2322.

[587]

O. A. Andreassen, R. J. Ferrante, P. Klivenyi, et al., “Partial Deficiency of Manganese Superoxide Dismutase Exacerbates a Transgenic Mouse Model of Amyotrophic Lateral Sclerosis,” Annals of Neurology 47, no. 4 (2000): 447-455.

[588]

H. Chen, X. Li, and P. N. Epstein, “MnSOD and Catalase Transgenes Demonstrate That Protection of Islets From Oxidative Stress Does not Alter Cytokine Toxicity,” Diabetes 54, no. 5 (2005): 1437-1446.

[589]

S. Bertera, M. L. Crawford, A. M. Alexander, et al., “Gene Transfer of Manganese Superoxide Dismutase Extends Islet Graft Function in a Mouse Model of Autoimmune Diabetes,” Diabetes 52, no. 2 (2003): 387-393.

[590]

Y. D. Liu, L. Yu, L. Ying, et al., “Toll-Like Receptor 2 Regulates Metabolic Reprogramming in Gastric Cancer via Superoxide Dismutase 2,” International Journal of Cancer 144, no. 12 (2019): 3056-3069.

[591]

B. Faubert, A. Solmonson, and R. J. DeBerardinis, “Metabolic Reprogramming and Cancer Progression,” Science 368, no. 6487 (2020): eaaw5473.

[592]

C. Zhou, L. Lyu, H. Miao, et al., “Redox Regulation by SOD2 Modulates Colorectal Cancer Tumorigenesis Through AMPK-Mediated Energy Metabolism,” Molecular Carcinogenesis 59, no. 5 (2020): 545-556.

[593]

K. Loh, H. Deng, A. Fukushima, et al., “Reactive Oxygen Species Enhance Insulin Sensitivity,” Cell metabolism 10, no. 4 (2009): 260-272.

[594]

H. Yan, Q. Tuo, Q. Yin, and P. Lei, “The Pathological Role of Ferroptosis in Ischemia/reperfusion-Related Injury,” Zoological Research 41, no. 3 (2020): 220-230.

[595]

X. Li, N. Ma, J. Xu, et al., “Targeting Ferroptosis: Pathological Mechanism and Treatment of Ischemia-Reperfusion Injury,” Oxidative Medicine and Cellular Longevity 2021 (2021): 1587922.

[596]

X. Wu, Y. Li, S. Zhang, and X. Zhou, “Ferroptosis as a Novel Therapeutic Target for Cardiovascular Disease,” Theranostics 11, no. 7 (2021): 3052-3059.

[597]

W. Tonnus, C. Meyer, C. Steinebach, et al., “Dysfunction of the key Ferroptosis-Surveilling Systems Hypersensitizes Mice to Tubular Necrosis During Acute Kidney Injury,” Nature Communications 12, no. 1 (2021): 4402.

[598]

J. P. Friedmann Angeli, M. Schneider, B. Proneth, et al., “Inactivation of the Ferroptosis Regulator Gpx4 Triggers Acute Renal Failure in Mice,” Nature Cell Biology 16, no. 12 (2014): 1180-1191.

[599]

J. Zhang, J. Bi, Y. Ren, et al., “Involvement of GPX4 in Irisin's Protection Against Ischemia Reperfusion-Induced Acute Kidney Injury,” Journal of Cellular Physiology 236, no. 2 (2021): 931-945.

[600]

W. Li, W. Li, Y. Leng, Y. Xiong, and Z. Xia, “Ferroptosis Is Involved in Diabetes Myocardial Ischemia/Reperfusion Injury Through Endoplasmic Reticulum Stress,” Dna and Cell Biology 39, no. 2 (2020): 210-225.

[601]

J. M. Hollander, K. M. Lin, B. T. Scott, and W. H. Dillmann, “Overexpression of PHGPx and HSP60/10 Protects Against Ischemia/reoxygenation Injury,” Free Radical Biology and Medicine 35, no. 7 (2003): 742-751.

[602]

L. Yang, B. Du, S. Zhang, and M. Wang, “RXRγ Attenuates Cerebral Ischemia-Reperfusion Induced Ferroptosis in Neurons in Mice Through Transcriptionally Promoting the Expression of GPX4,” Metabolic Brain Disease 37, no. 5 (2022): 1351-1363.

[603]

Z. Zhang, Y. Wu, S. Yuan, et al., “Glutathione Peroxidase 4 Participates in Secondary Brain Injury Through Mediating Ferroptosis in a rat Model of Intracerebral Hemorrhage,” Brain Research 1701 (2018): 112-125.

[604]

X. Guan, X. Li, X. Yang, et al., “The Neuroprotective Effects of Carvacrol on Ischemia/reperfusion-Induced Hippocampal Neuronal Impairment by Ferroptosis Mitigation,” Life Sciences 235 (2019): 116795.

[605]

M. Yoo, X. Gu, X. Xu, et al., “Delineating the Role of Glutathione Peroxidase 4 in Protecting Cells Against Lipid Hydroperoxide Damage and in Alzheimer's Disease,” Antioxid Redox Signaling 12, no. 7 (2010): 819-827.

[606]

W. S. Hambright, R. S. Fonseca, L. Chen, R. Na, and Q. Ran, “Ablation of Ferroptosis Regulator Glutathione Peroxidase 4 in Forebrain Neurons Promotes Cognitive Impairment and Neurodegeneration,” Redox Biology 12 (2017): 8-17.

[607]

X. Qu, J. He, Z. Cui, T. Yang, and X. Sun, “PPAR-α Agonist GW7647 Protects Against Oxidative Stress and Iron Deposit via GPx4 in a Transgenic Mouse Model of Alzheimer's Diseases,” Acs Chemical Neuroscience 13, no. 2 (2022): 207-216.

[608]

V. Casañas-Sánchez, J. A. Pérez, N. Fabelo, et al., “Addition of Docosahexaenoic Acid, but not Arachidonic Acid, Activates Glutathione and Thioredoxin Antioxidant Systems in Murine Hippocampal HT22 Cells: Potential Implications in Neuroprotection,” Journal of Neurochemistry 131, no. 4 (2014): 470-483.

[609]

V. Casañas-Sánchez, J. A. Pérez, N. Fabelo, D. Quinto-Alemany, and M. L. Díaz, “Docosahexaenoic (DHA) Modulates Phospholipid-Hydroperoxide Glutathione Peroxidase (Gpx4) Gene Expression to Ensure Self-Protection From Oxidative Damage in Hippocampal Cells,” Frontiers in Physiology 6 (2015): 203.

[610]

Q. Wang, C. Bin, Q. Xue, et al., “GSTZ1 Sensitizes Hepatocellular Carcinoma Cells to Sorafenib-Induced Ferroptosis via Inhibition of NRF2/GPX4 Axis,” Cell Death & Disease 12, no. 5 (2021): 426.

[611]

D. Shin, E. H. Kim, J. Lee, and J. Roh, “Nrf2 Inhibition Reverses Resistance to GPX4 Inhibitor-Induced Ferroptosis in Head and Neck Cancer,” Free Radical Biology and Medicine 129 (2018): 454-462.

[612]

X. Song, X. Wang, Z. Liu, and Z. Yu, “Role of GPX4-Mediated Ferroptosis in the Sensitivity of Triple Negative Breast Cancer Cells to Gefitinib,” Frontiers in Oncology 10 (2020): 597434.

[613]

H. Zhao, B. Ji, J. Chen, Q. Huang, and X. Lu, “Gpx 4 is Involved in the Proliferation, Migration and Apoptosis of Glioma Cells,” Pathology, Research and Practice 213, no. 6 (2017): 626-633.

[614]

M. Jia, D. Qin, C. Zhao, et al., “Redox Homeostasis Maintained by GPX4 Facilitates STING Activation,” Nature Immunology 21, no. 7 (2020): 727-735.

[615]

Y. Zhao, J. Lian, Z. Lan, et al., “Ferroptosis Promotes Anti-Tumor Immune Response by Inducing Immunogenic Exposure in HNSCC,” Oral Diseases 29, no. 3 (2023): 933-941.

[616]

B. E. Holbein, C. Lehmann, “Dysregulated Iron Homeostasis as Common Disease Etiology and Promising Therapeutic Target,” Antioxidants (Basel) 12, no. 3 (2023): 671.

[617]

A. R. Bogdan, M. Miyazawa, K. Hashimoto, and Y. Tsuji, “Regulators of Iron Homeostasis: New Players in Metabolism, Cell Death, and Disease,” Trends in Biochemical Sciences 41, no. 3 (2016): 274-286.

[618]

T. R. Daniels, E. Bernabeu, J. A. Rodríguez, et al., “The Transferrin Receptor and the Targeted Delivery of Therapeutic Agents Against Cancer,” Biochimica Et Biophysica Acta 1820, no. 3 (2012): 291-317.

[619]

S. L. Byrne, P. D. Buckett, J. Kim, et al., “Ferristatin II Promotes Degradation of Transferrin Receptor-1 in Vitro and in Vivo,” PLoS ONE 8, no. 7 (2013): e70199.

[620]

G. Y. L. Lui, Z. Kovacevic, V. Richardson, et al., “Targeting Cancer by Binding Iron: Dissecting Cellular Signaling Pathways,” Oncotarget 6, no. 22 (2015): 18748-18779.

[621]

J. H. Ohyashiki, C. Kobayashi, R. Hamamura, et al., “The Oral Iron Chelator Deferasirox Represses Signaling Through the mTOR in Myeloid Leukemia Cells by Enhancing Expression of REDD1,” Cancer Science 100, no. 5 (2009): 970-977.

[622]

G. Y. L. Lui, P. Obeidy, S. J. Ford, 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,” Molecular Pharmacology 83, no. 1 (2013): 179-190.

[623]

A. Terman, T. Kurz, “Lysosomal Iron, Iron Chelation, and Cell Death,” Antioxid Redox Signaling 18, no. 8 (2013): 888-898.

[624]

J. Benadiba, C. Rosilio, M. Nebout, et al., “Iron Chelation: An Adjuvant Therapy to Target Metabolism, Growth and Survival of Murine PTEN-Deficient T Lymphoma and Human T Lymphoblastic Leukemia/lymphoma,” Leukemia & Lymphoma 58, no. 6 (2017): 1433-1445.

[625]

Y. Luo, X. Gao, L. Zou, et al., “Bavachin Induces Ferroptosis Through the STAT3/P53/SLC7A11 Axis in Osteosarcoma Cells,” Oxidative Medicine and Cellular Longevity 2021 (2021): 1783485.

[626]

Y. Yu, Y. Yan, F. Niu, et al., “Ferroptosis: A Cell Death Connecting Oxidative Stress, Inflammation and Cardiovascular Diseases,” Cell Death Discovery 7, no. 1 (2021): 193.

[627]

X. Fang, H. Ardehali, J. Min, and F. Wang, “The Molecular and Metabolic Landscape of Iron and Ferroptosis in Cardiovascular Disease,” Nature Reviews Cardiology 20, no. 1 (2023): 7-23.

[628]

T. Bai, M. Li, Y. Liu, Z. Qiao, and Z. Wang, “Inhibition of Ferroptosis Alleviates Atherosclerosis Through Attenuating Lipid Peroxidation and Endothelial Dysfunction in Mouse Aortic Endothelial Cell,” Free Radical Biology and Medicine 160 (2020): 92-102.

[629]

Z. Xiao, B. Kong, J. Fang, et al., “Ferrostatin-1 Alleviates Lipopolysaccharide-Induced Cardiac Dysfunction,” Bioengineered 12, no. 2 (2021): 9367-9376.

[630]

Y. Feng, N. B. Madungwe, A. D. Imam Aliagan, N. Tombo, and J. C. Bopassa, “Liproxstatin-1 Protects the Mouse Myocardium Against Ischemia/reperfusion Injury by Decreasing VDAC1 Levels and Restoring GPX4 Levels,” Biochemical and Biophysical Research Communications 520, no. 3 (2019): 606-611.

[631]

C. Zou, X. Liu, R. Xie, et al., “Deferiprone Attenuates Inflammation and Myocardial Fibrosis in Diabetic Cardiomyopathy Rats,” Biochemical and Biophysical Research Communications 486, no. 4 (2017): 930-936.

[632]

H. Chang, R. Wu, M. Shang, et al., “Reduction in Mitochondrial Iron Alleviates Cardiac Damage During Injury,” EMBO Molecular Medicine 8, no. 3 (2016): 247-267.

[633]

N. Li, W. Jiang, W. Wang, et al., “Ferroptosis and its Emerging Roles in Cardiovascular Diseases,” Pharmacological Research 166 (2021): 105466.

[634]

N. Sumneang, N. Siri-Angkul, S. Kumfu, S. C. Chattipakorn, and N. Chattipakorn, “The Effects of Iron Overload on Mitochondrial Function, Mitochondrial Dynamics, and Ferroptosis in Cardiomyocytes,” Archives of Biochemistry and Biophysics 680 (2020): 108241.

[635]

J. Wang, J. Fu, Y. Zhao, et al., “Iron and Targeted Iron Therapy in Alzheimer's Disease,” International Journal of Molecular Sciences 24, no. 22 (2023): 16353.

[636]

A. Martin-Bastida, R. J. Ward, R. Newbould, et al., “Brain Iron Chelation by Deferiprone in a Phase 2 Randomised Double-Blinded Placebo Controlled Clinical Trial in Parkinson's Disease,” Scientific Reports 7, no. 1 (2017): 1398.

[637]

A. Elkouzi, V. Vedam-Mai, R. S. Eisinger, and M. S. Okun, “Emerging Therapies in Parkinson Disease—repurposed Drugs and new Approaches,” Nature reviews Neurology 15, no. 4 (2019): 204-223.

[638]

M. T. Nuñez, P. Chana-Cuevas, “New Perspectives in Iron Chelation Therapy for the Treatment of Neurodegenerative Diseases,” Pharmaceuticals (Basel) 11, no. 4 (2018): 109.

[639]

R. Calderon Moreno, A. Navas-Acien, E. Escolar, et al., “Potential Role of Metal Chelation to Prevent the Cardiovascular Complications of Diabetes,” Journal of Clinical Endocrinology and Metabolism 104, no. 7 (2019): 2931-2941.

[640]

M. Brownlee, H. Vlassara, A. Kooney, P. Ulrich, and A. Cerami, “Aminoguanidine Prevents Diabetes-Induced Arterial Wall Protein Cross-Linking,” Science 232, no. 4758 (1986): 1629-1632.

[641]

B. I. Freedman, J. P. Wuerth, K. Cartwright, et al., “Design and Baseline Characteristics for the Aminoguanidine Clinical Trial in Overt Type 2 Diabetic Nephropathy (ACTION II),” Controlled Clinical Trials 20, no. 5 (1999): 493-510.

[642]

T. Miyata, C. van Ypersele De Strihou, Y. Ueda, et al., “Angiotensin II Receptor Antagonists and Angiotensin-Converting Enzyme Inhibitors Lower in Vitro the Formation of Advanced Glycation end Products: Biochemical Mechanisms,” Journal of the American Society of Nephrology 13, no. 10 (2002): 2478-2487.

[643]

Y. Huang, W. Li, Z. Su, and A. T. Kong, “The Complexity of the Nrf2 Pathway: Beyond the Antioxidant Response,” Journal of Nutritional Biochemistry 26, no. 12 (2015): 1401-1413.

[644]

S. Kim, H. Lee, S. Park, et al., “Keap1 Cysteine 288 as a Potential Target for Diallyl Trisulfide-Induced Nrf2 Activation,” PLoS ONE 9, no. 1 (2014): e85984.

[645]

C. Tsai, C. Wang, T. Lai, et al., “Antioxidant Effects of Diallyl Trisulfide on High Glucose-Induced Apoptosis are Mediated by the PI3K/Akt-Dependent Activation of Nrf2 in Cardiomyocytes,” International Journal of Cardiology 168, no. 2 (2013): 1286-1297.

[646]

S. Lee, I. Lee, and W. Mar, “Inhibition of Inducible Nitric Oxide Synthase and Cyclooxygenase-2 Activity by 1,2,3,4,6-Penta-O-Galloyl-Beta-D-Glucose in Murine Macrophage Cells,” Archives of Pharmacal Research 26, no. 10 (2003): 832-839.

[647]

N. Khan, H. Mukhtar, “Multitargeted Therapy of Cancer by Green tea Polyphenols,” Cancer Letters 269, no. 2 (2008): 269-280.

[648]

R. Gold, R. A. Linker, and M. Stangel, “Fumaric Acid and its Esters: An Emerging Treatment for Multiple Sclerosis With Antioxidative Mechanism of Action,” Clinical Immunology 142, no. 1 (2012): 44-48.

[649]

R. H. Scannevin, S. Chollate, M. Jung, et al., “Fumarates Promote Cytoprotection of Central Nervous System Cells Against Oxidative Stress via the Nuclear Factor (erythroid-Derived 2)-Like 2 Pathway,” Journal of Pharmacology and Experimental Therapeutics 341, no. 1 (2012): 274-284.

[650]

L. Cheng, H. Zhang, F. Wu, et al., “Role of Nrf2 and Its Activators in Cardiocerebral Vascular Disease,” Oxidative Medicine and Cellular Longevity 2020 (2020): 4683943.

[651]

G. Bresciani, F. Manai, S. Davinelli, et al., “Novel Potential Pharmacological Applications of Dimethyl Fumarate-An Overview and Update,” Frontiers in Pharmacology 14 (2023): 1264842.

[652]

S. D. Thomas, N. K. Jha, B. Sadek, and S. Ojha, “Repurposing Dimethyl Fumarate for Cardiovascular Diseases: Pharmacological Effects, Molecular Mechanisms, and Therapeutic Promise,” Pharmaceuticals (Basel) 15, no. 5 (2022): 497.

[653]

A. Boivin, M. Hanot, C. Malesys, et al., “Transient Alteration of Cellular Redox Buffering Before Irradiation Triggers Apoptosis in Head and Neck Carcinoma Stem and non-Stem Cells,” PLoS ONE 6, no. 1 (2011): e14558.

[654]

M. Li, Z. Zhang, D. L. Hill, H. Wang, and R. Zhang, “Curcumin, a Dietary Component, has Anticancer, Chemosensitization, and Radiosensitization Effects by Down-Regulating the MDM2 Oncogene Through the PI3K/mTOR/ETS2 Pathway,” Cancer Research 67, no. 5 (2007): 1988-1996.

[655]

L. Li, B. Ahmed, K. Mehta, and R. Kurzrock, “Liposomal Curcumin With and Without Oxaliplatin: Effects on Cell Growth, Apoptosis, and Angiogenesis in Colorectal Cancer,” Molecular Cancer Therapeutics 6, no. 4 (2007): 1276-1282.

[656]

C. Wang, S. Chen, H. Guo, et al., “Forsythoside A Mitigates Alzheimer's-Like Pathology by Inhibiting Ferroptosis-Mediated Neuroinflammation via Nrf2/GPX4 Axis Activation,” International Journal of Biological Sciences 18, no. 5 (2022): 2075-2090.

[657]

X. Wang, X. Chen, W. Zhou, et al., “Ferroptosis is Essential for Diabetic Cardiomyopathy and is Prevented by Sulforaphane via AMPK/NRF2 Pathways,” Acta Pharmaceutica Sinica B 12, no. 2 (2022): 708-722.

[658]

Y. Luo, S. Sun, Y. Zhang, et al., “Effects of Oltipraz on the Glycolipid Metabolism and the Nrf2/HO-1 Pathway in Type 2 Diabetic Mice,” Drug Design, Development and Therapy 18 (2024): 5685-5700.

[659]

Y. Tuo, Z. Liu, J. Chen, et al., “NADPH Oxidase Inhibitor Improves Outcome of Mechanical Reperfusion by Suppressing Hemorrhagic Transformation,” Journal of NeuroInterventional Surgery 9, no. 5 (2017): 492-498.

[660]

D. Gebremedhin, M. Terashvili, N. Wickramasekera, et al., “Redox Signaling via Oxidative Inactivation of PTEN Modulates Pressure-Dependent Myogenic Tone in rat Middle Cerebral Arteries,” PLoS ONE 8, no. 7 (2013): e68498.

[661]

T B. A. Hart, S. Copray, and I. Philippens, “Apocynin, a low Molecular Oral Treatment for Neurodegenerative Disease,” BioMed Research International 2014 (2014): 298020.

[662]

A. M. Mahmoud, M. M. Ali, E. R. Miranda, et al., “Nox2 Contributes to Hyperinsulinemia-Induced Redox Imbalance and Impaired Vascular Function,” Redox Biology 13 (2017): 288-300.

[663]

D. S. de Jesus, P. Bargi-Souza, V. Cruzat, et al., “BMAL1 Modulates ROS Generation and Insulin Secretion in Pancreatic β-Cells: An Effect Possibly Mediated via NOX2,” Molecular and Cellular Endocrinology 555 (2022): 111725.

[664]

N. Quilaqueo-Millaqueo, D. A. Brown-Brown, J. A. Vidal-Vidal, and I. Niechi, “NOX Proteins and ROS Generation: Role in Invadopodia Formation and Cancer Cell Invasion,” Biological Research 57, no. 1 (2024): 98.

[665]

A. Czigler, L. Toth, N. Szarka, et al., “Hypertension Exacerbates Cerebrovascular Oxidative Stress Induced by Mild Traumatic Brain Injury: Protective Effects of the Mitochondria-Targeted Antioxidative Peptide SS-31,” Journal of Neurotrauma 36, no. 23 (2019): 3309-3315.

[666]

Z. Hao, Y. Huang, M. Wang, et al., “SS31 Ameliorates age-Related Activation of NF-κB Signaling in Senile Mice Model, SAMP8,” Oncotarget 8, no. 2 (2017): 1983-1992.

[667]

A. M. James, H. M. Cochemé, R. A. J. Smith, and M. P. Murphy, “Interactions of Mitochondria-Targeted and Untargeted Ubiquinones With the Mitochondrial Respiratory Chain and Reactive Oxygen Species. Implications for the use of Exogenous Ubiquinones as Therapies and Experimental Tools,” Journal of Biological Chemistry 280, no. 22 (2005): 21295-21312.

[668]

G. F. Kelso, C. M. Porteous, C. V. Coulter, et al., “Selective Targeting of a Redox-Active Ubiquinone to Mitochondria Within Cells: Antioxidant and Antiapoptotic Properties,” Journal of Biological Chemistry 276, no. 7 (2001): 4588-4596.

[669]

A. M. James, M. S. Sharpley, A. B. Manas, et al., “Interaction of the Mitochondria-Targeted Antioxidant MitoQ With Phospholipid Bilayers and Ubiquinone Oxidoreductases,” Journal of Biological Chemistry 282, no. 20 (2007): 14708-14718.

[670]

A. O. Oyewole, M. A. Birch-Machin, “Mitochondria-Targeted Antioxidants,” Faseb Journal 29, no. 12 (2015): 4766-4771.

[671]

R. A. Gioscia-Ryan, T. J. LaRocca, A. L. Sindler, et al., “Mitochondria-Targeted Antioxidant (MitoQ) Ameliorates age-Related Arterial Endothelial Dysfunction in Mice,” The Journal of Physiology 592, no. 12 (2014): 2549-2561.

[672]

B. J. Snow, F. L. Rolfe, M. M. Lockhart, et al., “A Double-Blind, Placebo-Controlled Study to Assess the Mitochondria-Targeted Antioxidant MitoQ as a Disease-Modifying Therapy in Parkinson's Disease,” Movement Disorders 25, no. 11 (2010): 1670-1674.

[673]

E. J. Anderson, M. E. Lustig, K. E. Boyle, et al., “Mitochondrial H2O2 Emission and Cellular Redox State Link Excess fat Intake to Insulin Resistance in Both Rodents and Humans,” Journal of Clinical Investigation 119, no. 3 (2009): 573-581.

[674]

X. Wang, Z. Liu, P. Peng, et al., “Astaxanthin Attenuates Osteoarthritis Progression via Inhibiting Ferroptosis and Regulating Mitochondrial Function in Chondrocytes,” Chemico-Biological Interactions 366 (2022): 110148.

[675]

Y. Nishida, A. Nawaz, K. Hecht, and K. Tobe, “Astaxanthin as a Novel Mitochondrial Regulator: A New Aspect of Carotenoids, Beyond Antioxidants,” Nutrients 14, no. 1 (2021): 107.

[676]

G. B. Kubat, E. Bouhamida, O. Ulger, et al., “Mitochondrial Dysfunction and Skeletal Muscle Atrophy: Causes, Mechanisms, and Treatment Strategies,” Mitochondrion 72 (2023): 33-58.

[677]

S. Upadhayay, P. Kumar, “Mitochondrial Targeted Antioxidants as Potential Therapy for Huntington's Disease,” Pharmacological Reports 76, no. 4 (2024): 693-713.

[678]

Z. Xun, P. Wipf, and C. T. McMurray, “XJB-5-131 Is a Mild Uncoupler of Oxidative Phosphorylation,” Journal of Huntington's Disease 11, no. 2 (2022): 141-151.

[679]

Z. Fisar, J. Hroudova, “CoQ10 and Mitochondrial Dysfunction in Alzheimer's Disease,” Antioxidants (Basel) 13, no. 2 (2024): 191.

[680]

M. Sanoobar, S. Eghtesadi, A. Azimi, et al., “Coenzyme Q10 Supplementation Ameliorates Inflammatory Markers in Patients With Multiple Sclerosis: A Double Blind, Placebo, Controlled Randomized Clinical Trial,” Nutritional Neuroscience 18, no. 4 (2015): 169-176.

[681]

L. D. Coles, P. J. Tuite, G. Oz, et al., “Repeated-Dose Oral N-Acetylcysteine in Parkinson's Disease: Pharmacokinetics and Effect on Brain Glutathione and Oxidative Stress,” Journal of Clinical Pharmacology 58, no. 2 (2018): 158-167.

[682]

M. Millán, N. DeGregorio-Rocasolano, N. Pérez De La Ossa, et al., “Targeting Pro-Oxidant Iron With Deferoxamine as a Treatment for Ischemic Stroke: Safety and Optimal Dose Selection in a Randomized Clinical Trial,” Antioxidants (Basel) 10, no. 8 (2021): 1270.

[683]

F. Shakeri, S. Kiani, G. Rahimi, and M. H. Boskabady, “Anti-Inflammatory, Antioxidant, and Immunomodulatory Effects of Berberis Vulgaris and its Constituent Berberine, Experimental and Clinical, a Review,” Phytotherapy Research 38, no. 4 (2024): 1882-1902.

[684]

E. Tian, G. Sharma, and C. Dai, “Neuroprotective Properties of Berberine: Molecular Mechanisms and Clinical Implications,” Antioxidants (Basel) 12, no. 10 (2023): 1883.

[685]

S. Naz, T. Mahmood, R. Gupta, et al., “Clinical Manifestation of AGE-Rage Axis in Neurodegenerative and Cognitive Impairment Disorders,” Drug Research (Stuttgart) 73, no. 06 (2023): 309-317.

[686]

D. Yesilbursa, A. Serdar, T. Senturk, et al., “Effect of N-Acetylcysteine on Oxidative Stress and Ventricular Function in Patients With Myocardial Infarction,” Heart and Vessels 21, no. 1 (2006): 33-37.

[687]

S. Pasupathy, R. Tavella, S. Grover, et al., “Early Use of N-Acetylcysteine With Nitrate Therapy in Patients Undergoing Primary Percutaneous Coronary Intervention for ST-Segment-Elevation Myocardial Infarction Reduces Myocardial Infarct Size (the NACIAM Trial [N-Acetylcysteine in Acute Myocardial Infarction]),” Circulation 136, no. 10 (2017): 894-903.

[688]

F. Dehghani, M. Vafa, A. Ebrahimkhani, M. Găman, and S. H. Sezavar Seyedi Jandaghi, “Effects of Quercetin Supplementation on Endothelial Dysfunction Biomarkers and Depression in Post-Myocardial Infarction Patients: A Double-Blind, Placebo-Controlled, Randomized Clinical Trial,” Clinical Nutrition ESPEN 56 (2023): 73-80.

[689]

R. T. Ras, P. L. Zock, Y. Zebregs, et al., “Effect of Polyphenol-Rich Grape Seed Extract on Ambulatory Blood Pressure in Subjects With pre- and Stage I Hypertension,” British Journal of Nutrition 110, no. 12 (2013): 2234-2241.

[690]

S. Dey, D. DeMazumder, A. Sidor, D. B. Foster, and B. O'Rourke, “Mitochondrial ROS Drive Sudden Cardiac Death and Chronic Proteome Remodeling in Heart Failure,” Circulation Research 123, no. 3 (2018): 356-371.

[691]

R. A. Kloner, S. L. Hale, W. Dai, et al., “Reduction of Ischemia/reperfusion Injury With Bendavia, a Mitochondria-Targeting Cytoprotective Peptide,” Journal of the American Heart Association 1, no. 3 (2012): e001644.

[692]

D. A. Brown, S. L. Hale, C. P. Baines, et al., “Reduction of Early Reperfusion Injury With the Mitochondria-Targeting Peptide Bendavia,” Journal of Cardiovascular Pharmacology and Therapeutics 19, no. 1 (2014): 121-132.

[693]

J. Cho, K. Won, D. Wu, et al., “Potent Mitochondria-Targeted Peptides Reduce Myocardial Infarction in Rats,” Coronary Artery Disease 18, no. 3 (2007): 215-220.

[694]

D. Dai, E. J. Hsieh, T. Chen, et al., “Global Proteomics and Pathway Analysis of Pressure-Overload-Induced Heart Failure and its Attenuation by Mitochondrial-Targeted Peptides,” Circulation: Heart Failure 6, no. 5 (2013): 1067-1076.

[695]

H. Liu, Y. Huang, S. Cheng, Y. Huang, and P. Lin, “Effects of Coenzyme Q10 Supplementation on Antioxidant Capacity and Inflammation in Hepatocellular Carcinoma Patients After Surgery: A Randomized, Placebo-Controlled Trial,” Nutrition Journal 15, no. 1 (2016): 85.

[696]

K. R. Patel, V. A. Brown, D. J. L. Jones, et al., “Clinical Pharmacology of Resveratrol and its Metabolites in Colorectal Cancer Patients,” Cancer Research 70, no. 19 (2010): 7392-7399.

[697]

J. Huober, W. Fett, A. Nusch, et al., “A Multicentric Observational Trial of Pegylated Liposomal Doxorubicin for Metastatic Breast Cancer,” BMC cancer 10 (2010): 2.

[698]

H. Ozkaya, T. Omma, Y. M. Bag, et al., “Topical and Systemic Effects of N-Acetyl Cysteine on Wound Healing in a Diabetic Rat Model,” Wounds 31, no. 4 (2019): 91-96.

[699]

M. Hu, B. Liu, “Resveratrol via Activation of LKB1-Ampk Signaling Suppresses Oxidative Stress to Prevent Endothelial Dysfunction in Diabetic Mice,” Clinical and Experimental Hypertension 38, no. 4 (2016): 381-387.

[700]

W. Mahjabeen, D. A. Khan, and S. A. Mirza, “Role of Resveratrol Supplementation in Regulation of Glucose Hemostasis, Inflammation and Oxidative Stress in Patients With Diabetes Mellitus Type 2: A Randomized, Placebo-Controlled Trial,” Complementary Therapies in Medicine 66 (2022): 102819.

[701]

Q. Wang, M. Zhang, G. Torres, et al., “Metformin Suppresses Diabetes-Accelerated Atherosclerosis via the Inhibition of Drp1-Mediated Mitochondrial Fission,” Diabetes 66, no. 1 (2017): 193-205.

[702]

J. Hou, X. Wang, Y. Li, et al., “Positive Regulation of Endothelial Tom70 by Metformin as a new Mechanism Against Cardiac Microvascular Injury in Diabetes,” Mitochondrion 65 (2022): 150-160.

[703]

V. K. P. Venu, M. Saifeddine, K. Mihara, et al., “Metformin Prevents Hyperglycemia-Associated, Oxidative Stress-Induced Vascular Endothelial Dysfunction: Essential Role for the Orphan Nuclear Receptor Human Nuclear Receptor 4A1 (Nur77),” Molecular Pharmacology 100, no. 5 (2021): 428-455.

[704]

W. C. Knowler, E. Barrett-Connor, S. E. Fowler, et al., “Reduction in the Incidence of Type 2 Diabetes With Lifestyle Intervention or Metformin,” New England Journal of Medicine 346, no. 6 (2002): 393-403.

[705]

Q. Li, Y. Lin, S. Wang, L. Zhang, and L. Guo, “GLP-1 Inhibits High-Glucose-Induced Oxidative Injury of Vascular Endothelial Cells,” Scientific Reports 7, no. 1 (2017): 8008.

[706]

Y. Yang, Q. Zhao, “Exenatide Regulates Inflammation and the Production of Reactive Oxygen Species via Inhibition of S1PR2 Synthesis,” Advances in Clinical and Experimental Medicine: Official Organ Wroclaw Medical University 30, no. 5 (2021): 555-561.

[707]

X. Luo, Y. Hu, S. He, et al., “Dulaglutide Inhibits High Glucose- Induced Endothelial Dysfunction and NLRP3 Inflammasome Activation,” Archives of Biochemistry and Biophysics 671 (2019): 203-209.

[708]

W. T. Garvey, A. L. Birkenfeld, D. Dicker, et al., “Efficacy and Safety of Liraglutide 3.0 mg in Individuals With Overweight or Obesity and Type 2 Diabetes Treated With Basal Insulin: The SCALE Insulin Randomized Controlled Trial,” Diabetes Care 43, no. 5 (2020): 1085-1093.

[709]

Y. Zhou, S. Tai, N. Zhang, L. Fu, and Y. Wang, “Dapagliflozin Prevents Oxidative Stress-Induced Endothelial Dysfunction via Sirtuin 1 Activation,” Biomedicine & Pharmacotherapy 165 (2023): 115213.

[710]

M. El-Daly, V. K. Pulakazhi Venu, M. Saifeddine, et al., “Hyperglycaemic Impairment of PAR2-Mediated Vasodilation: Prevention by Inhibition of Aortic Endothelial Sodium-Glucose-Co-Transporter-2 and Minimizing Oxidative Stress,” Vascular Pharmacology 109 (2018): 56-71.

[711]

A. T. Dinkova-Kostova, W. D. Holtzclaw, R. N. Cole, et al., “Direct Evidence That Sulfhydryl Groups of Keap1 are the Sensors Regulating Induction of Phase 2 Enzymes That Protect Against Carcinogens and Oxidants,” PNAS 99, no. 18 (2002): 11908-11913.

[712]

M. Xue, Q. Qian, A. Adaikalakoteswari, et al., “Activation of NF-E2-Related Factor-2 Reverses Biochemical Dysfunction of Endothelial Cells Induced by Hyperglycemia Linked to Vascular Disease,” Diabetes 57, no. 10 (2008): 2809-2817.

[713]

Y. Chai, D. Sheng, X. Ji, et al., “Developmental and Neurobehavioral Toxicity of 2,2'-Methylenebis(6-Tert-Butyl-4-Methylphenol) (antioxidant AO2246) During the Early Life Stage of Zebrafish,” Science of the Total Environment 899 (2023): 166306.

[714]

S. Di Meo, P. Venditti, “Evolution of the Knowledge of Free Radicals and Other Oxidants,” Oxidative Medicine and Cellular Longevity 2020 (2020): 9829176.

[715]

Y. Dong, Q. Yuan, J. Ren, et al., “Identification and Characterization of a Novel Molecular Classification Incorporating Oxidative Stress and Metabolism-Related Genes for Stomach Adenocarcinoma in the Framework of Predictive, Preventive, and Personalized Medicine,” Frontiers in Endocrinology (Lausanne) 14 (2023): 1090906.

[716]

Y. Cui, Q. Yuan, J. Chen, et al., “Determination and Characterization of Molecular Heterogeneity and Precision Medicine Strategies of Patients With Pancreatic Cancer and Pancreatic Neuroendocrine Tumor Based on Oxidative Stress and Mitochondrial Dysfunction-Related Genes,” Frontiers in Endocrinology (Lausanne) 14 (2023): 1127441.

[717]

X. Zhou, N. Wang, B. Zhao, Z. Liu, and P. Yu, “Precision Medicine for Acute Kidney Injury: Baicalein-Nanodrug Delivery System Combat Oxidative Stress and Repair Mitochondrial Dysfunction,” International Journal of Pharmaceutics 678 (2025): 125694.

[718]

J. Huang, D. Zhou, W. Luo, et al., “Integrating Oxidative-Stress Biomarkers Into a Precision Oncology Risk-Stratification Model for Bladder Cancer Prognosis and Therapy,” Frontiers in Cell and Developmental Biology 12 (2024): 1453448.

[719]

J. Shi, B. Peng, R. Xu, et al., “Exploration Oxidative Stress Underlying Gastroesophageal Reflux Disease and Therapeutic Targets Identification: A Multi-Omics Mendelian Randomization Study,” Postgraduate Medical Journal (2024).

[720]

H. Wang, H. Guo, J. Sun, and Y. Wang, “Multi-Omics Analyses Based on Genes Associated With Oxidative Stress and Phospholipid Metabolism Revealed the Intrinsic Molecular Characteristics of Pancreatic Cancer,” Scientific Reports 13, no. 1 (2023): 13564.

[721]

S. Zhao, Q. Hu, H. Jiang, et al., “Multi-Omics Analysis of Oxidative Stress and Apoptosis in Hepatopancreas Cells Induced by Polyascus Gregaria Parasitizing the Eriocheir Sinensis,” Fish & Shellfish Immunology 143 (2023): 109180.

[722]

S. Chen, J. Sun, W. Wen, Z. Chen, and Z. Yu, “Integrative Multi-Omics Summary-Based Mendelian Randomization Identifies key Oxidative Stress-Related Genes as Therapeutic Targets for Atrial Fibrillation and Flutter,” Frontiers in Genetics 15 (2024): 1447872.

RIGHTS & PERMISSIONS

2025 The Author(s). MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.

AI Summary AI Mindmap
PDF

10

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/