Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 3770 2022-12-08 10:20:18 |
2 format correct Meta information modification 3770 2022-12-09 02:13:10 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Pan, Y.;  Wang, X.;  Liu, X.;  Shen, L.;  Chen, Q.;  Shu, Q. Targeting Ferroptosis for Ischemia-Reperfusion Injury. Encyclopedia. Available online: https://encyclopedia.pub/entry/38297 (accessed on 27 July 2024).
Pan Y,  Wang X,  Liu X,  Shen L,  Chen Q,  Shu Q. Targeting Ferroptosis for Ischemia-Reperfusion Injury. Encyclopedia. Available at: https://encyclopedia.pub/entry/38297. Accessed July 27, 2024.
Pan, Yihang, Xueke Wang, Xiwang Liu, Lihua Shen, Qixing Chen, Qiang Shu. "Targeting Ferroptosis for Ischemia-Reperfusion Injury" Encyclopedia, https://encyclopedia.pub/entry/38297 (accessed July 27, 2024).
Pan, Y.,  Wang, X.,  Liu, X.,  Shen, L.,  Chen, Q., & Shu, Q. (2022, December 08). Targeting Ferroptosis for Ischemia-Reperfusion Injury. In Encyclopedia. https://encyclopedia.pub/entry/38297
Pan, Yihang, et al. "Targeting Ferroptosis for Ischemia-Reperfusion Injury." Encyclopedia. Web. 08 December, 2022.
Targeting Ferroptosis for Ischemia-Reperfusion Injury
Edit

Ischemia-reperfusion (I/R) injury is a major challenge in perioperative medicine that contributes to pathological damage in various conditions, including ischemic stroke, myocardial infarction, acute lung injury, liver transplantation, acute kidney injury and hemorrhagic shock. I/R damage is often irreversible, and current treatments for I/R injury are limited. Ferroptosis, a type of regulated cell death characterized by the iron-dependent accumulation of lipid hydroperoxides, has been implicated in multiple diseases, including I/R injury. Emerging evidence suggests that ferroptosis can serve as a therapeutic target to alleviate I/R injury, and pharmacological strategies targeting ferroptosis have been developed in I/R models.

ischemia-reperfusion injury ferroptosis iron antioxidant therapeutic strategies

1. Introduction

Ischemia-reperfusion (I/R) is a pathological condition characterized by the initial restriction of the blood supply to organs or tissues, followed by the restoration of blood flow and reoxygenation. Insufficient blood supply leads to tissue hypoxia and cellular metabolic imbalance, and subsequent reperfusion and reoxygenation cause excessive inflammatory responses and exacerbate ischemic tissue damage, which is known as I/R injury [1]. I/R injury is intrinsically associated with oxidative damage, and multiple pathological processes contribute to this damage, including impaired endothelial barrier function, mitochondrial dysfunction, activation of the cell death program, calcium overload, sterile inflammation and autoimmune responses [2]. However, the precise molecular mechanism of I/R injury has not been fully elucidated and targeted therapies are still limited. Overall, therapeutic strategies for this condition need to be developed, and examining new therapeutic targets to manage I/R injury is a top priority.
Cell death is a stable pathological indicator of I/R injury. Emerging evidence has revealed a novel therapeutic concept to target regulated cell death (RCD) to counteract I/R injury, although the role of RCD in I/R injury has only recently become apparent [3]. Different forms of RCD have been identified in I/R injury, including autophagy, necroptosis, apoptosis, pyroptosis, parthanatos and ferroptosis [2]. Recent studies suggest that targeting RCD exerts beneficial effects against I/R injury; in particular, RCD in parenchymal and endothelial cells is recognized as a promising intervention target. I/R injury leads to RCD of parenchymal and endothelial cells, and apoptosis, necroptosis and autophagy are the most common types [4]. Generally, regulating I/R-induced RCD has been recognized as a new therapeutic strategy against I/R injury, but effective interventions are rarely summarized.
Recently, ferroptosis, a form of RCD characterized by iron-dependent lipid peroxidation, glutathione (GSH) depletion and glutathione peroxidase 4 (GPX4) inactivation [5], has received great attention in I/R events [6]. Ferroptosis occurs during the reperfusion but not the ischemic phase, as the levels of two key enzymes in ferroptosis, GPX4 and long-chain-fatty-acid-CoA ligase 4 (ACSL4) in tissues were significantly regulated only during reperfusion, accompanied by elevated iron concentration and malondialdehyde (MDA) levels [7]. During the reperfusion phase, mitochondrial respiration is enhanced, which consequently triggers reactive oxygen species (ROS) explosion and ferroptosis [8]. Notably, decreased mitochondrial membrane potential (MMP) can be observed, which indicates increased mitochondrial outer membrane permeability, a characteristic of mitochondrial-mediated apoptosis [9]. Although caspases are activated during this process, interventions targeting apoptosis do not completely prevent cell death. Thus, although activated in I/R, apoptosis is not essential for subsequent cell death, suggesting the existence of other mechanisms governing cell death [10], such as ferroptosis. And indeed, mitochondria play a much more active role in ferroptosis than in apoptosis (17). Moreover, some morphological characteristics of mitochondria during I/R, including reduced mitochondrial volume, reduced or even lost mitochondrial cristae, and condensed mitochondrial membrane densities, are not associated with other forms of cell death, further emphasizing the relevance of ferroptosis [8].

References

  1. Eltzschig, H.K.; Eckle, T. Ischemia and reperfusion—From mechanism to translation. Nat. Med. 2011, 17, 1391–1401.
  2. Chen, D.Q.; Guo, Y.; Li, X.; Zhang, G.Q.; Li, P. Small molecules as modulators of regulated cell death against ischemia/reperfusion injury. Med. Res. Rev. 2022, 42, 2067–2101.
  3. Lee, H.; Zandkarimi, F.; Zhang, Y.; Meena, J.K.; Kim, J.; Zhuang, L.; Tyagi, S.; Ma, L.; Westbrook, T.F.; Steinberg, G.R.; et al. Energy-stress-mediated ampk activation inhibits ferroptosis. Nat. Cell. Biol. 2020, 22, 225–234.
  4. Heusch, G. Myocardial ischaemia-reperfusion injury and cardioprotection in perspective. Nat. Rev. Cardiol. 2020, 17, 773–789.
  5. Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell 2012, 149, 1060–1072.
  6. Yan, H.F.; Tuo, Q.Z.; Yin, Q.Z.; Lei, P. The pathological role of ferroptosis in ischemia/reperfusion-related injury. Zool Res. 2020, 41, 220–230.
  7. Tang, L.J.; Luo, X.J.; Tu, H.; Chen, H.; Xiong, X.M.; Li, N.S.; Peng, J. Ferroptosis occurs in phase of reperfusion but not ischemia in rat heart following ischemia or ischemia/reperfusion. Naunyn Schmiedebergs Arch. Pharmacol. 2021, 394, 401–410.
  8. Wang, H.; Liu, C.; Zhao, Y.; Gao, G. Mitochondria regulation in ferroptosis. Eur. J. Cell. Biol. 2020, 99, 151058.
  9. Gao, M.; Yi, J.; Zhu, J.; Minikes, A.M.; Monian, P.; Thompson, C.B.; Jiang, X. Role of mitochondria in ferroptosis. Mol. Cell. 2019, 73, 354–363.e3.
  10. Loor, G.; Kondapalli, J.; Iwase, H.; Chandel, N.S.; Waypa, G.B.; Guzy, R.D.; Hoek, T.L.V.; Schumacker, P.T. Mitochondrial oxidant stress triggers cell death in simulated ischemia-reperfusion. Biochim. Biophys. Acta 2011, 1813, 1382–1394.
  11. Hankey, G.J. Stroke. Lancet 2017, 389, 641–654.
  12. Park, U.J.; Lee, Y.A.; Won, S.M.; Lee, J.H.; Kang, S.H.; Springer, J.E.; Lee, Y.B.; Gwag, B.J. Blood-derived iron mediates free radical production and neuronal death in the hippocampal ca1 area following transient forebrain ischemia in rat. Acta Neuropathol. 2011, 121, 459–473.
  13. Won, S.M.; Lee, J.H.; Park, U.J.; Gwag, J.; Gwag, B.J.; Lee, Y.B. Iron mediates endothelial cell damage and blood-brain barrier opening in the hippocampus after transient forebrain ischemia in rats. Exp. Mol. Med. 2011, 43, 121–128.
  14. Dávalos, A.; Fernandez-Real, J.M.; Ricart, W.; Soler, S.; Molins, A.; Planas, E.; Genís, D. Iron-related damage in acute ischemic stroke. Stroke 1994, 25, 1543–1546.
  15. Castellanos, M.; Puig, N.; Carbonell, T.; Castillo, J.; Martinez, J.; Rama, R.; Dávalos, A. Iron intake increases infarct volume after permanent middle cerebral artery occlusion in rats. Brain Res. 2002, 952, 1–6.
  16. Prass, K.; Ruscher, K.; Karsch, M.; Isaev, N.; Megow, D.; Priller, J.; Scharff, A.; Dirnagl, U.; Meisel, A. Desferrioxamine induces delayed tolerance against cerebral ischemia in vivo and in vitro. J. Cereb. Blood Flow Metab. 2002, 22, 520–525.
  17. Doll, S.; Conrad, M. Iron and ferroptosis: A still ill-defined liaison. IUBMB Life 2017, 69, 423–434.
  18. Lei, P.; Ayton, S.; Appukuttan, A.T.; Moon, S.; Duce, J.A.; Volitakis, I.; Cherny, R.; Wood, S.J.; Greenough, M.; Berger, G.; et al. Lithium suppression of tau induces brain iron accumulation and neurodegeneration. Mol. Psychiatry 2017, 22, 396–406.
  19. Tuo, Q.Z.; Lei, P.; Jackman, K.A.; Li, X.L.; Xiong, H.; Li, X.L.; Liuyang, Z.Y.; Roisman, L.; Zhang, S.T.; Ayton, S.; et al. Tau-mediated iron export prevents ferroptotic damage after ischemic stroke. Mol. Psychiatry 2017, 22, 1520–1530.
  20. Pham, C.G.; Bubici, C.; Zazzeroni, F.; Papa, S.; Jones, J.; Alvarez, K.; Jayawardena, S.; de Smaele, E.; Cong, R.; Beaumont, C.; et al. Ferritin heavy chain upregulation by nf-kappab inhibits tnfalpha-induced apoptosis by suppressing reactive oxygen species. Cell 2004, 119, 529–542.
  21. Hou, W.; Xie, Y.; Song, X.; Sun, X.; Lotze, M.T.; Zeh, H.J., 3rd; Kang, R.; Tang, D. Autophagy promotes ferroptosis by degradation of ferritin. Autophagy 2016, 12, 1425–1428.
  22. Chen, W.; Jiang, L.; Hu, Y.; Tang, N.; Liang, N.; Li, X.F.; Chen, Y.W.; Qin, H.; Wu, L. Ferritin reduction is essential for cerebral ischemia-induced hippocampal neuronal death through p53/slc7a11-mediated ferroptosis. Brain Res. 2021, 1752, 147216.
  23. Wang, Y.Q.; Chang, S.Y.; Wu, Q.; Gou, Y.J.; Jia, L.; Cui, Y.M.; Yu, P.; Shi, Z.H.; Wu, W.S.; Gao, G.; et al. The protective role of mitochondrial ferritin on erastin-induced ferroptosis. Front. Aging Neurosci. 2016, 8, 308.
  24. Wang, P.; Cui, Y.; Ren, Q.; Yan, B.; Zhao, Y.; Yu, P.; Gao, G.; Shi, H.; Chang, S.; Chang, Y.Z. Mitochondrial ferritin attenuates cerebral ischaemia/reperfusion injury by inhibiting ferroptosis. Cell. Death Dis. 2021, 12, 447.
  25. Mancias, J.D.; Vaites, L.P.; Nissim, S.; Biancur, D.E.; Kim, A.J.; Wang, X.; Liu, Y.; Goessling, W.; Kimmelman, A.C.; Harper, J.W. Ferritinophagy via ncoa4 is required for erythropoiesis and is regulated by iron dependent herc2-mediated proteolysis. Elife 2015, 4, e10308.
  26. Mancias, J.D.; Wang, X.; Gygi, S.P.; Harper, J.W.; Kimmelman, A.C. Quantitative proteomics identifies ncoa4 as the cargo receptor mediating ferritinophagy. Nature 2014, 509, 105–109.
  27. Gao, M.; Monian, P.; Pan, Q.; Zhang, W.; Xiang, J.; Jiang, X. Ferroptosis is an autophagic cell death process. Cell. Res. 2016, 26, 1021–1032.
  28. Li, C.; Sun, G.; Chen, B.; Xu, L.; Ye, Y.; He, J.; Bao, Z.; Zhao, P.; Miao, Z.; Zhao, L.; et al. Nuclear receptor coactivator 4-mediated ferritinophagy contributes to cerebral ischemia-induced ferroptosis in ischemic stroke. Pharmacol. Res. 2021, 174, 105933.
  29. Ingold, I.; Berndt, C.; Schmitt, S.; Doll, S.; Poschmann, G.; Buday, K.; Roveri, A.; Peng, X.; Freitas, F.P.; Seibt, T.; et al. Selenium utilization by gpx4 is required to prevent hydroperoxide-induced ferroptosis. Cell 2018, 172, 409–422.e21.
  30. Stockwell, B.R.; Angeli, J.P.F.; Bayir, H.; Bush, A.I.; Conrad, M.; Dixon, S.J.; Fulda, S.; Gascón, S.; Hatzios, S.K.; Kagan, V.E.; et al. Ferroptosis: A regulated cell death nexus linking metabolism, redox biology, and disease. Cell 2017, 171, 273–285.
  31. Alim, I.; Caulfield, J.T.; Chen, Y.; Swarup, V.; Geschwind, D.H.; Ivanova, E.; Seravalli, J.; Ai, Y.; Sansing, L.H.; Marie, E.J.S.; et al. Selenium drives a transcriptional adaptive program to block ferroptosis and treat stroke. Cell 2019, 177, 1262–1279.e25.
  32. Tuo, Q.Z.; Masaldan, S.; Southon, A.; Mawal, C.; Ayton, S.; Bush, A.I.; Lei, P.; Belaidi, A.A. Characterization of selenium compounds for anti-ferroptotic activity in neuronal cells and after cerebral ischemia-reperfusion injury. Neurotherapeutics 2021, 18, 2682–2691.
  33. Shi, Y.; Han, L.; Zhang, X.; Xie, L.; Pan, P.; Chen, F. Selenium alleviates cerebral ischemia/reperfusion injury by regulating oxidative stress, mitochondrial fusion and ferroptosis. Neurochem. Res. 2022, 47, 2992–3002.
  34. Chen, W.; Xu, B.; Xiao, A.; Liu, L.; Fang, X.; Liu, R.; Turlova, E.; Barszczyk, A.; Zhong, X.; Sun, C.L.; et al. Trpm7 inhibitor carvacrol protects brain from neonatal hypoxic-ischemic injury. Mol Brain 2015, 8, 11.
  35. Guan, X.; Li, X.; Yang, X.; Yan, J.; Shi, P.; Ba, L.; Cao, Y.; Wang, P. The neuroprotective effects of carvacrol on ischemia/reperfusion-induced hippocampal neuronal impairment by ferroptosis mitigation. Life Sci. 2019, 235, 116795.
  36. Fu, C.; Wu, Y.; Liu, S.; Luo, C.; Lu, Y.; Liu, M.; Wang, L.; Zhang, Y.; Liu, X. Rehmannioside a improves cognitive impairment and alleviates ferroptosis via activating pi3k/akt/nrf2 and slc7a11/gpx4 signaling pathway after ischemia. J. Ethnopharmacol. 2022, 289, 115021.
  37. Guan, X.; Li, Z.; Zhu, S.; Cheng, M.; Ju, Y.; Ren, L.; Yang, G.; Min, D. Galangin attenuated cerebral ischemia-reperfusion injury by inhibition of ferroptosis through activating the slc7a11/gpx4 axis in gerbils. Life Sci. 2021, 264, 118660.
  38. Guo, H.; Zhu, L.; Tang, P.; Chen, D.; Li, Y.; Li, J.; Bao, C. Carthamin yellow improves cerebral ischemia-reperfusion injury by attenuating inflammation and ferroptosis in rats. Int. J. Mol. Med. 2021, 47, 52.
  39. Yuan, Y.; Zhai, Y.; Chen, J.; Xu, X.; Wang, H. Kaempferol ameliorates oxygen-glucose deprivation/reoxygenation-induced neuronal ferroptosis by activating nrf2/slc7a11/gpx4 axis. Biomolecules 2021, 11, 923.
  40. Arrigo, M.; Price, S.; Baran, D.A.; Pöss, J.; Aissaoui, N.; Bayes-Genis, A.; Bonello, L.; François, B.; Gayat, E.; Gilard, M.; et al. Optimising clinical trials in acute myocardial infarction complicated by cardiogenic shock: A statement from the 2020 critical care clinical trialists workshop. Lancet Respir. Med. 2021, 9, 1192–1202.
  41. Ibáñez, B.; Heusch, G.; Ovize, M.; van de Werf, F. Evolving therapies for myocardial ischemia/reperfusion injury. J. Am. Coll. Cardiol. 2015, 65, 1454–1471.
  42. Li, J.Y.; Liu, S.Q.; Yao, R.Q.; Tian, Y.P.; Yao, Y.M. A novel insight into the fate of cardiomyocytes in ischemia-reperfusion injury: From iron metabolism to ferroptosis. Front. Cell. Dev. Biol. 2021, 9, 799499.
  43. Lillo-Moya, J.; Rojas-Solé, C.; Muñoz-Salamanca, D.; Panieri, E.; Saso, L.; Rodrigo, R. Targeting ferroptosis against ischemia/reperfusion cardiac injury. Antioxidants 2021, 10, 667.
  44. Gao, M.; Monian, P.; Quadri, N.; Ramasamy, R.; Jiang, X. Glutaminolysis and transferrin regulate ferroptosis. Mol. Cell. 2015, 59, 298–308.
  45. Lin, Z.; Yang, H.; Kong, Q.; Li, J.; Lee, S.M.; Gao, B.; Dong, H.; Wei, J.; Song, J.; Zhang, D.D.; et al. Usp22 antagonizes p53 transcriptional activation by deubiquitinating sirt1 to suppress cell apoptosis and is required for mouse embryonic development. Mol. Cell. 2012, 46, 484–494.
  46. Ma, S.; Sun, L.; Wu, W.; Wu, J.; Sun, Z.; Ren, J. Usp22 protects against myocardial ischemia-reperfusion injury via the sirt1-p53/slc7a11-dependent inhibition of ferroptosis-induced cardiomyocyte death. Front. Physiol. 2020, 11, 551318.
  47. Tang, L.J.; Zhou, Y.J.; Xiong, X.M.; Li, N.S.; Zhang, J.J.; Luo, X.J.; Peng, J. Ubiquitin-specific protease 7 promotes ferroptosis via activation of the p53/tfr1 pathway in the rat hearts after ischemia/reperfusion. Free Radic. Biol. Med. 2021, 162, 339–352.
  48. Lakhal-Littleton, S.; Wolna, M.; Carr, C.A.; Miller, J.J.; Christian, H.C.; Ball, V.; Santos, A.; Diaz, R.; Biggs, D.; Stillion, R.; et al. Cardiac ferroportin regulates cellular iron homeostasis and is important for cardiac function. Proc. Natl. Acad. Sci. USA 2015, 112, 3164–3169.
  49. Harada, N.; Kanayama, M.; Maruyama, A.; Yoshida, A.; Tazumi, K.; Hosoya, T.; Mimura, J.; Toki, T.; Maher, J.M.; Yamamoto, M.; et al. Nrf2 regulates ferroportin 1-mediated iron efflux and counteracts lipopolysaccharide-induced ferroportin 1 mrna suppression in macrophages. Arch. Biochem. Biophys. 2011, 508, 101–109.
  50. Zhang, B.; Zhai, M.; Li, B.; Liu, Z.; Li, K.; Jiang, L.; Zhang, M.; Yi, W.; Yang, J.; Yi, D.; et al. Honokiol ameliorates myocardial ischemia/reperfusion injury in type 1 diabetic rats by reducing oxidative stress and apoptosis through activating the sirt1-nrf2 signaling pathway. Oxid. Med. Cell. Longev. 2018, 2018, 3159801.
  51. Tian, H.; Xiong, Y.; Zhang, Y.; Leng, Y.; Tao, J.; Li, L.; Qiu, Z.; Xia, Z. Activation of nrf2/fpn1 pathway attenuates myocardial ischemia-reperfusion injury in diabetic rats by regulating iron homeostasis and ferroptosis. Cell. Stress Chaperones 2021, 27, 149–164.
  52. Li, W.; Li, W.; Wang, Y.; Leng, Y.; Xia, Z. Inhibition of dnmt-1 alleviates ferroptosis through ncoa4 mediated ferritinophagy during diabetes myocardial ischemia/reperfusion injury. Cell. Death Discov. 2021, 7, 267.
  53. Yeang, C.; Hasanally, D.; Que, X.; Hung, M.Y.; Stamenkovic, A.; Chan, D.; Chaudhary, R.; Margulets, V.; Edel, A.L.; Hoshijima, M.; et al. Reduction of myocardial ischaemia-reperfusion injury by inactivating oxidized phospholipids. Cardiovasc. Res. 2019, 115, 179–189.
  54. Stamenkovic, A.; O’Hara, K.A.; Nelson, D.C.; Maddaford, T.G.; Edel, A.L.; Maddaford, G.; Dibrov, E.; Aghanoori, M.; Kirshenbaum, L.A.; Fernyhough, P.; et al. Oxidized phosphatidylcholines trigger ferroptosis in cardiomyocytes during ischemia-reperfusion injury. Am. J. Physiol. Heart Circ. Physiol. 2021, 320, H1170–H1184.
  55. Simone, L.E.; Keene, J.D. Mechanisms coordinating elav/hu mrna regulons. Curr. Opin. Genet. Dev. 2013, 23, 35–43.
  56. de Bruin, R.G.; Rabelink, T.J.; van Zonneveld, A.J.; van der Veer, E.P. Emerging roles for rna-binding proteins as effectors and regulators of cardiovascular disease. Eur. Heart J. 2017, 38, 1380–1388.
  57. Zhang, Z.; Yao, Z.; Wang, L.; Ding, H.; Shao, J.; Chen, A.; Zhang, F.; Zheng, S. Activation of ferritinophagy is required for the rna-binding protein elavl1/hur to regulate ferroptosis in hepatic stellate cells. Autophagy 2018, 14, 2083–2103.
  58. Chen, H.Y.; Xiao, Z.Z.; Ling, X.; Xu, R.N.; Zhu, P.; Zheng, S.Y. Elavl1 is transcriptionally activated by foxc1 and promotes ferroptosis in myocardial ischemia/reperfusion injury by regulating autophagy. Mol. Med. 2021, 27, 14.
  59. Xu, Y.; Li, X.; Cheng, Y.; Yang, M.; Wang, R. Inhibition of acsl4 attenuates ferroptotic damage after pulmonary ischemia-reperfusion. FASEB J. 2020, 34, 16262–16275.
  60. Li, Y.; Cao, Y.; Xiao, J.; Shang, J.; Tan, Q.; Ping, F.; Huang, W.; Wu, F.; Zhang, H.; Zhang, X. Inhibitor of apoptosis-stimulating protein of p53 inhibits ferroptosis and alleviates intestinal ischemia/reperfusion-induced acute lung injury. Cell. Death Differ. 2020, 27, 2635–2650.
  61. Wei, S.; Bi, J.; Yang, L.; Zhang, J.; Wan, Y.; Chen, X.; Wang, Y.; Wu, Z.; Lv, Y.; Wu, R. Serum irisin levels are decreased in patients with sepsis, and exogenous irisin suppresses ferroptosis in the liver of septic mice. Clin. Transl. Med. 2020, 10, e173.
  62. Chen, K.; Xu, Z.; Liu, Y.; Wang, Z.; Li, Y.; Xu, X.; Chen, C.; Xia, T.; Liao, Q.; Yao, Y.; et al. Irisin protects mitochondria function during pulmonary ischemia/reperfusion injury. Sci. Transl. Med. 2017, 9, eaao6298.
  63. Yamada, N.; Karasawa, T.; Wakiya, T.; Sadatomo, A.; Ito, H.; Kamata, R.; Watanabe, S.; Komada, T.; Kimura, H.; Sanada, Y.; et al. Iron overload as a risk factor for hepatic ischemia-reperfusion injury in liver transplantation: Potential role of ferroptosis. Am. J. Transplant. 2020, 20, 1606–1618.
  64. Mao, L.; Zhao, T.; Song, Y.; Lin, L.; Fan, X.; Cui, B.; Feng, H.; Wang, X.; Yu, Q.; Zhang, J.; et al. The emerging role of ferroptosis in non-cancer liver diseases: Hype or increasing hope? Cell. Death Dis. 2020, 11, 518.
  65. Wu, S.; Yang, J.; Sun, G.; Hu, J.; Zhang, Q.; Cai, J.; Yuan, D.; Li, H.; Hei, Z.; Yao, W. Macrophage extracellular traps aggravate iron overload-related liver ischaemia/reperfusion injury. Br. J. Pharmacol. 2021, 178, 3783–3796.
  66. Zhong, Q.; Gao, W.; Du, F.; Wang, X. Mule/arf-bp1, a bh3-only e3 ubiquitin ligase, catalyzes the polyubiquitination of mcl-1 and regulates apoptosis. Cell 2005, 121, 1085–1095.
  67. Wu, Y.; Jiao, H.; Yue, Y.; He, K.; Jin, Y.; Zhang, J.; Zhang, J.; Wei, Y.; Luo, H.; Hao, Z.; et al. Ubiquitin ligase e3 huwe1/mule targets transferrin receptor for degradation and suppresses ferroptosis in acute liver injury. Cell. Death Differ. 2022, 29, 1705–1718.
  68. Ghosh, M.C.; Tong, W.H.; Zhang, D.; Ollivierre-Wilson, H.; Singh, A.; Krishna, M.C.; Mitchell, J.B.; Rouault, T.A. Tempol-mediated activation of latent iron regulatory protein activity prevents symptoms of neurodegenerative disease in irp2 knockout mice. Proc. Natl. Acad. Sci. USA 2008, 105, 12028–12033.
  69. Li, X.; Wu, L.; Tian, X.; Zheng, W.; Yuan, M.; Tian, X.; Zuo, H.; Song, H.; Shen, Z. Mir-29a-3p in exosomes from heme oxygenase-1 modified bone marrow mesenchymal stem cells alleviates steatotic liver ischemia-reperfusion injury in rats by suppressing ferroptosis via iron responsive element binding protein 2. Oxid. Med. Cell. Longev. 2022, 2022, 6520789.
  70. Friedmann Angeli, J.P.; Schneider, M.; Proneth, B.; Tyurina, Y.Y.; Tyurin, V.A.; Hammond, V.J.; Herbach, N.; Aichler, M.; Walch, A.; Eggenhofer, E.; et al. Inactivation of the ferroptosis regulator gpx4 triggers acute renal failure in mice. Nat. Cell. Biol. 2014, 16, 1180–1191.
  71. Polimeno, L.; Pesetti, B.; de Santis, F.; Resta, L.; Rossi, R.; de Palma, A.; Girardi, B.; Amoruso, A.; Francavilla, A. Decreased expression of the augmenter of liver regeneration results in increased apoptosis and oxidative damage in human-derived glioma cells. Cell. Death Dis. 2012, 3, e289.
  72. Huang, L.L.; Liao, X.H.; Sun, H.; Jiang, X.; Liu, Q.; Zhang, L. Augmenter of liver regeneration protects the kidney from ischaemia-reperfusion injury in ferroptosis. J. Cell. Mol. Med. 2019, 23, 4153–4164.
  73. Jankowski, J.; Perry, H.M.; Medina, C.B.; Huang, L.; Yao, J.; Bajwa, A.; Lorenz, U.M.; Rosin, D.L.; Ravichandran, K.S.; Isakson, B.E.; et al. Epithelial and endothelial pannexin1 channels mediate aki. J. Am. Soc. Nephrol. 2018, 29, 1887–1899.
  74. Su, L.; Jiang, X.; Yang, C.; Zhang, J.; Chen, B.; Li, Y.; Yao, S.; Xie, Q.; Gomez, H.; Murugan, R.; et al. Pannexin 1 mediates ferroptosis that contributes to renal ischemia/reperfusion injury. J. Biol. Chem. 2019, 294, 19395–19404.
  75. Sui, M.; Xu, D.; Zhao, W.; Lu, H.; Chen, R.; Duan, Y.; Li, Y.; Zhu, Y.; Zhang, L.; Zeng, L. Cirbp promotes ferroptosis by interacting with elavl1 and activating ferritinophagy during renal ischaemia-reperfusion injury. J. Cell. Mol. Med. 2021, 25, 6203–6216.
  76. Miller, G.; Matthews, S.P.; Reinheckel, T.; Fleming, S.; Watts, C. Asparagine endopeptidase is required for normal kidney physiology and homeostasis. FASEB J. 2011, 25, 1606–1617.
  77. Chen, C.; Wang, D.; Yu, Y.; Zhao, T.; Min, N.; Wu, Y.; Kang, L.; Zhao, Y.; Du, L.; Zhang, M.; et al. Legumain promotes tubular ferroptosis by facilitating chaperone-mediated autophagy of gpx4 in aki. Cell. Death Dis. 2021, 12, 65.
  78. Mohib, K.; Wang, S.; Guan, Q.; Mellor, A.L.; Sun, H.; Du, C.; Jevnikar, A.M. Indoleamine 2,3-dioxygenase expression promotes renal ischemia-reperfusion injury. Am. J. Physiol.-Renal Physiol. 2008, 295, F226–F234.
  79. Eleftheriadis, T.; Pissas, G.; Golfinopoulos, S.; Liakopoulos, V.; Stefanidis, I. Role of indoleamine 2,3-dioxygenase in ischemia-reperfusion injury of renal tubular epithelial cells. Mol. Med. Rep. 2021, 23, 472.
  80. Feng, R.; Xiong, Y.; Lei, Y.; Huang, Q.; Liu, H.; Zhao, X.; Chen, Z.; Chen, H.; Liu, X.; Wang, L.; et al. Lysine-specific demethylase 1 aggravated oxidative stress and ferroptosis induced by renal ischemia and reperfusion injury through activation of tlr4/nox4 pathway in mice. J. Cell. Mol. Med. 2022, 26, 4254–4267.
  81. Zou, Y.F.; Wen, D.; Zhao, Q.; Shen, P.Y.; Shi, H.; Zhao, Q.; Chen, Y.X.; Zhang, W. Urinary microrna-30c-5p and microrna-192-5p as potential biomarkers of ischemia-reperfusion-induced kidney injury. Exp. Biol. Med. 2017, 242, 657–667.
  82. Wilflingseder, J.; Sunzenauer, J.; Toronyi, E.; Heinzel, A.; Kainz, A.; Mayer, B.; Perco, P.; Telkes, G.; Langer, R.M.; Oberbauer, R. Molecular pathogenesis of post-transplant acute kidney injury: Assessment of whole-genome mrna and mirna profiles. PLoS ONE 2014, 9, e104164.
  83. Wilflingseder, J.; Jelencsics, K.; Bergmeister, H.; Sunzenauer, J.; Regele, H.; Eskandary, F.; Reindl-Schwaighofer, R.; Kainz, A.; Oberbauer, R. Mir-182-5p inhibition ameliorates ischemic acute kidney injury. Am. J. Pathol. 2017, 187, 70–79.
  84. Zhang, X.; Wang, S.; Wang, H.; Cao, J.; Huang, X.; Chen, Z.; Xu, P.; Sun, G.; Xu, J.; Lv, J.; et al. Circular rna circnrip1 acts as a microrna-149-5p sponge to promote gastric cancer progression via the akt1/mtor pathway. Mol. Cancer 2019, 18, 20.
  85. Ding, C.; Ding, X.; Zheng, J.; Wang, B.; Li, Y.; Xiang, H.; Dou, M.; Qiao, Y.; Tian, P.; Xue, W. Mir-182-5p and mir-378a-3p regulate ferroptosis in i/r-induced renal injury. Cell. Death Dis. 2020, 11, 929.
  86. Tang, Z.; Ju, Y.; Dai, X.; Ni, N.; Liu, Y.; Zhang, D.; Gao, H.; Sun, H.; Zhang, J.; Gu, P. Ho-1-mediated ferroptosis as a target for protection against retinal pigment epithelium degeneration. Redox. Biol. 2021, 43, 101971.
  87. Tao, W.; Liu, F.; Zhang, J.; Fu, S.; Zhan, H.; Qian, K. Mir-3587 inhibitor attenuates ferroptosis following renal ischemia-reperfusion through ho-1. Front. Mol. Biosci. 2021, 8, 789927.
  88. Linkermann, A.; Skouta, R.; Himmerkus, N.; Mulay, S.R.; Dewitz, C.; de Zen, F.; Prokai, A.; Zuchtriegel, G.; Krombach, F.; Welz, P.S.; et al. Synchronized renal tubular cell death involves ferroptosis. Proc. Natl. Acad. Sci. USA 2014, 111, 16836–16841.
  89. Zhao, Z.; Wu, J.; Xu, H.; Zhou, C.; Han, B.; Zhu, H.; Hu, Z.; Ma, Z.; Ming, Z.; Yao, Y.; et al. Xjb-5-131 inhibited ferroptosis in tubular epithelial cells after ischemia-reperfusion injury. Cell. Death Dis. 2020, 11, 629.
  90. Wang, Y.; Quan, F.; Cao, Q.; Lin, Y.; Yue, C.; Bi, R.; Cui, X.; Yang, H.; Yang, Y.; Birnbaumer, L.; et al. Quercetin alleviates acute kidney injury by inhibiting ferroptosis. J. Adv. Res. 2021, 28, 231–243.
  91. Tonnus, W.; Meyer, C.; Steinebach, C.; Belavgeni, A.; von Mässenhausen, A.; Gonzalez, N.Z.; Maremonti, F.; Gembardt, F.; Himmerkus, N.; Latk, M.; et al. Dysfunction of the key ferroptosis-surveilling systems hypersensitizes mice to tubular necrosis during acute kidney injury. Nat. Commun. 2021, 12, 4402.
  92. Zhou, L.; Xue, X.; Hou, Q.; Dai, C. Targeting ferroptosis attenuates interstitial inflammation and kidney fibrosis. Kidney Dis. 2022, 8, 57–71.
  93. Zhang, J.; Bi, J.; Ren, Y.; Du, Z.; Li, T.; Wang, T.; Zhang, L.; Wang, M.; Wei, S.; Lv, Y.; et al. Involvement of gpx4 in irisin’s protection against ischemia reperfusion-induced acute kidney injury. J. Cell. Physiol. 2021, 236, 931–945.
  94. Jiang, G.P.; Liao, Y.J.; Huang, L.L.; Zeng, X.J.; Liao, X.H. Effects and molecular mechanism of pachymic acid on ferroptosis in renal ischemia reperfusion injury. Mol. Med. Rep. 2021, 23, 63.
  95. Yang, J.; Sun, X.; Huang, N.; Li, P.; He, J.; Jiang, L.; Zhang, X.; Han, S.; Xin, H. Entacapone alleviates acute kidney injury by inhibiting ferroptosis. FASEB J. 2022, 36, e22399.
  96. Li, Y.; Feng, D.; Wang, Z.; Zhao, Y.; Sun, R.; Tian, D.; Liu, D.; Zhang, F.; Ning, S.; Yao, J.; et al. Ischemia-induced acsl4 activation contributes to ferroptosis-mediated tissue injury in intestinal ischemia/reperfusion. Cell. Death Differ. 2019, 26, 2284–2299.
  97. Yang, W.S.; SriRamaratnam, R.; Welsch, M.E.; Shimada, K.; Skouta, R.; Viswanathan, V.S.; Cheah, J.H.; Clemons, P.A.; Shamji, A.F.; Clish, C.B.; et al. Regulation of ferroptotic cancer cell death by gpx4. Cell 2014, 156, 317–331.
  98. Cai, M.; Ma, Y.; Zhang, W.; Wang, S.; Wang, Y.; Tian, L.; Peng, Z.; Wang, H.; Qingrong, T. Apigenin-7-o-β-d-(-6’’-p-coumaroyl)-glucopyranoside treatment elicits neuroprotective effect against experimental ischemic stroke. Int. J. Biol. Sci. 2016, 12, 42–52.
  99. Feng, Y.D.; Ye, W.; Tian, W.; Meng, J.R.; Zhang, M.; Sun, Y.; Zhang, H.N.; Wang, S.J.; Wu, K.H.; Liu, C.X.; et al. Old targets, new strategy: Apigenin-7-o-β-d-(-6″-p-coumaroyl)-glucopyranoside prevents endothelial ferroptosis and alleviates intestinal ischemia-reperfusion injury through ho-1 and mao-b inhibition. Free Radic. Biol. Med. 2022, 184, 74–88.
  100. Deng, F.; Zhao, B.C.; Yang, X.; Lin, Z.B.; Sun, Q.S.; Wang, Y.F.; Yan, Z.Z.; Liu, W.F.; Li, C.; Hu, J.J.; et al. The gut microbiota metabolite capsiate promotes gpx4 expression by activating trpv1 to inhibit intestinal ischemia reperfusion-induced ferroptosis. Gut Microbes 2021, 13, 1902719.
  101. Dong, H.; Qiang, Z.; Chai, D.; Peng, J.; Xia, Y.; Hu, R.; Jiang, H. Nrf2 inhibits ferroptosis and protects against acute lung injury due to intestinal ischemia reperfusion via regulating slc7a11 and ho-1. Aging 2020, 12, 12943–12959.
  102. Qiang, Z.; Dong, H.; Xia, Y.; Chai, D.; Hu, R.; Jiang, H. Nrf2 and stat3 alleviates ferroptosis-mediated iir-ali by regulating slc7a11. Oxid. Med. Cell. Longev. 2020, 2020, 5146982.
  103. Dong, H.; Xia, Y.; Jin, S.; Xue, C.; Wang, Y.; Hu, R.; Jiang, H. Nrf2 attenuates ferroptosis-mediated iir-ali by modulating tert and slc7a11. Cell. Death Dis. 2021, 12, 1027.
  104. Zhongyin, Z.; Wei, W.; Juan, X.; Guohua, F. Isoliquiritin apioside relieves intestinal ischemia/reperfusion-induced acute lung injury by blocking hif-1α-mediated ferroptosis. Int. Immunopharmacol. 2022, 108, 108852.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , ,
View Times: 326
Revisions: 2 times (View History)
Update Date: 09 Dec 2022
1000/1000
Video Production Service