J Yeungnam Med Sci.  2024 Apr;41(2):61-73. 10.12701/jyms.2023.01347.

Comprehensive overview of the role of mitochondrial dysfunction in the pathogenesis of acute kidney ischemia-reperfusion injury: a narrative review

Affiliations
  • 1Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Korea
  • 2Research Institute of Aging and Metabolism, School of Medicine, Kyungpook National University, Daegu, Korea
  • 3Department of Physiology, School of Medicine, Kyungpook National University, Daegu, Korea

Abstract

Acute kidney ischemia-reperfusion (IR) injury is a life-threatening condition that predisposes individuals to chronic kidney disease. Since the kidney is one of the most energy-demanding organs in the human body and mitochondria are the powerhouse of cells, mitochondrial dysfunction plays a central role in the pathogenesis of IR-induced acute kidney injury. Mitochondrial dysfunction causes a reduction in adenosine triphosphate production, loss of mitochondrial dynamics (represented by persistent fragmentation), and impaired mitophagy. Furthermore, the pathological accumulation of succinate resulting from fumarate reduction under oxygen deprivation (ischemia) in the reverse flux of the Krebs cycle can eventually lead to a burst of reactive oxygen species driven by reverse electron transfer during the reperfusion phase. Accumulating evidence indicates that improving mitochondrial function, biogenesis, and dynamics, and normalizing metabolic reprogramming within the mitochondria have the potential to preserve kidney function during IR injury and prevent progression to chronic kidney disease. In this review, we summarize recent advances in understanding the detrimental role of metabolic reprogramming and mitochondrial dysfunction in IR injury and explore potential therapeutic strategies for treating kidney IR injury.

Keyword

Acute kidney injury; Ferroptosis; Mitochondria; Mitochondrial dynamics; Mitophagy; Reactive oxygen species; Review

Figure

  • Fig. 1. Ischemia induces mitochondrial death pathway and aberrant succinate accumulation. Ischemia significantly impacts mitochondrial functions, notably causing a decline in adenosine triphosphate (ATP) production within kidney proximal tubule cells. During reperfusion, a compromised state becomes evident with a significant increase in reactive oxygen species (ROS), which leads to mitochondrial dysfunction. This dysfunction causes a decrease in mitochondrial membrane potential (ΔΨm), subsequently triggering the opening of the mitochondrial permeability transition pore (mPTP). A crucial event in this process is the opening of the mPTP, which allows the release of cytochrome c and initiates the mitochondrial death pathway. Concurrently, hypoxia induces a reversal in the activity of complex II, leading to an excessive accumulation of succinate. Upon reperfusion, the accumulated succinate is rapidly oxidized back by succinate dehydrogenase (SDH). This phenomenon amplifies ROS generation, ultimately culminating in cellular death. Pyruvate dehydrogenase kinase 4 (PDK4) inhibition enhances pyruvate dehydrogenase complex (PDC) activity. Inhibition of the malate/aspartate shuttle (MAS) by activating the pyruvate dehydrogenase (PDH) flux can mitigate this succinate accumulation. VDAC, voltage-dependent anion channel; ANT, adenine nucleotide translocase; OMM, outer mitochondrial membrane; IMM, inner mitochondrial membrane; CypD, cyclophilin D; NAD+, oxidized nicotinamide adenine dinucleotide; NADH, reduced nicotinamide adenine dinucleotide; TCA, tricarboxylic acid cycle; MPC, mitochondrial pyruvate carrier; PC, pyruvate carboxylase.

  • Fig. 2. Mitochondrial dynamics and quality control in kidney IR injury. (A) Mitochondrial fusion-fission: mitochondria undergo fusion or division in response to stimuli like hypoxia. Dynamin-related protein 1 (Drp-1) predominantly causes fission. Importantly, OMA1-mediated proteolysis of optic atrophy 1 (OPA1) present in two main isoforms, a long form and a short form, plays a differential role in these processes. The long form of OPA1 (L-OPA1) promotes mitochondrial fusion, while the short form of OPA1 (S-OPA1) is more associated with fission. Stress can lead to inner membrane cleavage through OMA1’s action on OPA1, converting L-OPA1 to its S-OPA1 counterpart. In kidney injury, such processes are linked to tubular cell apoptosis. Under stressful conditions including IR injury, the endoplasmic reticulum (ER) also undergoes stress. Persistent ER stress causes a large amount of Ca2+ release from the ER. This released Ca2+ results in mitochondrial Ca2+ overload through the inositol 1,4,5-trisphosphate receptor (IP3R)-voltage-dependent anion channel (VDAC) 1, which forms a bridge between the two organelles at mitochondria-associated membranes. Furthermore, under such stressful conditions, Bax-interacting factor 1 (Bif-1) regulates mitochondrial membranes by interacting with prohibitin-2. This interaction leads to disruption of prohibitin (PHB) complexes, culminating in the inactivation of OPA1, particularly the L-OPA1 isoform, thereby favoring mitochondrial fission. (B) Mitochondrial quality control: mitophagy is the process of clearing damaged mitochondria from cells essential in kidney ischemia-reperfusion (IR) injury. PTEN-induced kinase 1 (PINK1) and parkin can detect mitochondrial quality, leading to removal of damaged mitochondria. Activation of mitophagy is critical for reducing IR injury. Disruption of mitophagy worsens the injury. Several proteins and pathways, such as pannexin 1 (PANX1), mammalian sterile 20-like kinase 1 (MST1), and acyl-CoA synthetase family member 2 (ACSF2), have roles in mitophagy and kidney IR injury protection. LTBP4, latent transforming growth factor-beta binding protein 4; MFN, mitofusin; OMM, outer mitochondrial membrane; IMM, inner mitochondrial membrane; VAPB, vesicle-associated membrane protein-associated protein B; GRP75, glucose-regulated protein 75; PTPIP51, protein tyrosine phosphatase interacting protein 51; ATP, adenosine triphosphate; P-mTOR, phosphorylated mammalian target of rapamycin; LC3, microtubule-associated proteins 1A/1B light chain 3; BNIP3, Bcl-2 interacting protein 3; NIX, NIP3-like protein X; AMPK, AMP-activated protein kinase; YAP, yes-associated protein.

  • Fig. 3. Ferroptosis as a central mechanism in kidney ischemia-reperfusion (IR) injury. IR injury can induce lipid peroxidation through various pathways, ultimately triggering ferroptosis and leading to acute kidney injury (AKI). Cells have various antioxidant enzymes (e.g., Catalase) and coenzymes (e.g., coenzyme Q10 [CoQ10]) to eliminate reactive oxygen species (ROS), thus maintaining cellular homeostasis. However, excessive ROS generated due to prolonged injury can eventually lead to lipid peroxidation. IR injury causes the release of Fe3+ from ruptured red blood cells, which binds to transferrin (TF) and then associates with TF receptor 1 (TfR1) on the cell membrane, forming endosomes. Within the endosome, six-transmembrane epithelial antigen of the prostate 3 (STEAP3) converts Fe3++ to Fe2+, which is subsequently released into the cytoplasm through solute carrier family 11 member 2 (SLC11A2), establishing a labile iron pool. Fe2++ is extruded from the cytoplasm to the extracellular space through cell membrane proteins such as SLC40A1, solute carrier family 40 member 1 (SCL40A1) and adaptor protein (poly(rC)-binding protein [PCBP] 2). Extracellular Fe2+ is converted back to Fe3+ by ceruloplasmin (CP). It can bind to TF, re-entering the cytoplasm by binding to TfR1, leading to endosome formation. Intracellularly, Fe2+ within the labile iron pool is regulated by PCBP1, which can store it as ferritin or convert it back to Fe2+ through nuclear receptor coactivator 4 (NCOA4). Additionally, heme oxygenase 1 (HMOX1) contributes to intracellular Fe2+ accumulation. Prolonged IR injury can lead to excessive cytoplasmic Fe2+, promoting the Fenton reaction, converting Fe2+ to Fe3+, and generating H2O2 and hydroxyl radicals (•OH) with subsequent lipid peroxidation. Intracellular Fe2+ also accumulates within mitochondria through membrane proteins such as solute carrier family 25 member 28 (SLC25A28) and solute carrier family 25 member 37 (SLC25A37), contributing to heme synthesis and Fe-S cluster formation. Fe2+ within mitochondria can accumulate ROS including peroxides (polyunsaturated fatty acid-phospholipid ethanolamine peroxyl radical, PL-OO•). Additionally, excessive succinate accumulation during IR injury can lead to ROS generation through the electron transport chain (ETC), causing lipid peroxidation both inside and outside the mitochondria. The antioxidant enzyme glutathione peroxidase 4 (GPX4) plays a crucial role in reducing lipid peroxidation by using reduced glutathione (GSH) to convert oxidized glutathione (GSSG) while countering ROS. GSH is produced intracellularly through the Xc (solute carrier family 7 member 11 [SCL7A11], solute carrier family 3 member 2 [SCL3A2]) transport system, converting cystine to cysteine. However, reduced GSH during IR injury can compromise GPX4’s activity. Increased tripartite motif-containing 21 (TRIM21) activity during IR injury can lead to GPX4 ubiquitination and degradation, intensifying ROS and lipid peroxidation. Activated inositol-requiring enzyme 1 (IRE1)/jun N-terminal kinase (JNK) pathways during IR injury can induce endoplasmic reticulum (ER) stress, promoting ferroptosis. Ferroptosis can exacerbate ER stress. IR-induced activation of indoleamine 2,3‑dioxygenase 1 (IDO1) can lead to increased conversion of intracellular tryptophan to kynurenine metabolites. These metabolites can bind to the aryl hydrocarbon receptor (AhR) in the nucleus, enhancing the expression of cytochrome P450 superfamily of enzymes (CYPs). CYPs can further increase ROS production, contributing to lipid peroxidation. AIFM2, apoptosis-inducing factor mitochondria-associated 2; CoQ10H2, reduced coenzyme Q10; NADPH, reduced nicotinamide adenine dinucleotide phosphate; NADP+, oxidized nicotinamide adenine dinucleotide phosphate; DMT1, divalent metal transporter 1; PL-OOH, polyunsaturated fatty acid containing phospholipid hydroperoxides.


Reference

References

1. Khwaja A. KDIGO clinical practice guidelines for acute kidney injury. Nephron Clin Pract. 2012; 120:c179–84.
Article
2. Rewa O, Bagshaw SM. Acute kidney injury-epidemiology, outcomes and economics. Nat Rev Nephrol. 2014; 10:193–207.
Article
3. Eltzschig HK, Eckle T. Ischemia and reperfusion--from mechanism to translation. Nat Med. 2011; 17:1391–401.
Article
4. Sabouny R, Shutt TE. The role of mitochondrial dynamics in mtDNA maintenance. J Cell Sci. 2021; 134:jcs258944.
Article
5. Javadov S, Kozlov AV, Camara AK. Mitochondria in health and diseases. Cells. 2020; 9:1177.
Article
6. Brooks C, Wei Q, Cho SG, Dong Z. Regulation of mitochondrial dynamics in acute kidney injury in cell culture and rodent models. J Clin Invest. 2009; 119:1275–85.
Article
7. Bhargava P, Schnellmann RG. Mitochondrial energetics in the kidney. Nat Rev Nephrol. 2017; 13:629–46.
Article
8. Chouchani ET, Pell VR, Gaude E, Aksentijević D, Sundier SY, Robb EL, et al. Ischaemic accumulation of succinate controls reperfusion injury through mitochondrial ROS. Nature. 2014; 515:431–5.
Article
9. Oh CJ, Kim MJ, Lee JM, Kim DH, Kim IY, Park S, et al. Inhibition of pyruvate dehydrogenase kinase 4 ameliorates kidney ischemia-reperfusion injury by reducing succinate accumulation during ischemia and preserving mitochondrial function during reperfusion. Kidney Int. 2023; 104:724–39.
Article
10. Nourbakhsh N, Singh P. Role of renal oxygenation and mitochondrial function in the pathophysiology of acute kidney injury. Nephron Clin Pract. 2014; 127:149–52.
Article
11. Yamamoto S, Yamamoto M, Nakamura J, Mii A, Yamamoto S, Takahashi M, et al. Spatiotemporal ATP dynamics during AKI predict renal prognosis. J Am Soc Nephrol. 2020; 31:2855–69.
Article
12. Nieuwenhuijs-Moeke GJ, Pischke SE, Berger SP, Sanders JS, Pol RA, Struys MM, et al. Ischemia and reperfusion injury in kidney transplantation: relevant mechanisms in injury and repair. J Clin Med. 2020; 9:253.
Article
13. Salvadori M, Rosso G, Bertoni E. Update on ischemia-reperfusion injury in kidney transplantation: pathogenesis and treatment. World J Transplant. 2015; 5:52–67.
Article
14. Park JS, Pasupulati R, Feldkamp T, Roeser NF, Weinberg JM. Cyclophilin D and the mitochondrial permeability transition in kidney proximal tubules after hypoxic and ischemic injury. Am J Physiol Renal Physiol. 2011; 301:F134–50.
Article
15. Wang C, Youle RJ. The role of mitochondria in apoptosis*. Annu Rev Genet. 2009; 43:95–118.
Article
16. Zhang J, Zhang J, Ni H, Wang Y, Katwal G, Zhao Y, et al. Downregulation of XBP1 protects kidney against ischemia-reperfusion injury via suppressing HRD1-mediated NRF2 ubiquitylation. Cell Death Discov. 2021; 7:44.
Article
17. Marin I, Boix O, Garcia-Garijo A, Sirois I, Caballe A, Zarzuela E, et al. Cellular senescence is immunogenic and promotes antitumor immunity. Cancer Discov. 2023; 13:410–31.
Article
18. Rocca C, Soda T, De Francesco EM, Fiorillo M, Moccia F, Viglietto G, et al. Mitochondrial dysfunction at the crossroad of cardiovascular diseases and cancer. J Transl Med. 2023; 21:635.
Article
19. Ma H, Guo X, Cui S, Wu Y, Zhang Y, Shen X, et al. Dephosphorylation of AMP-activated protein kinase exacerbates ischemia/reperfusion-induced acute kidney injury via mitochondrial dysfunction. Kidney Int. 2022; 101:315–30.
Article
20. Yang X, Kang A, Lu Y, Li Y, Guo L, Li R, et al. Exploratory metabolomic analysis based on UHPLC-Q-TOF-MS/MS to study hypoxia-reoxygenation energy metabolic alterations in HK-2 cells. Ren Fail. 2023; 45:2186715.
Article
21. Singh P. Reprogramming of energy metabolism in kidney disease. Nephron. 2023; 147:61–4.
Article
22. Lan R, Geng H, Singha PK, Saikumar P, Bottinger EP, Weinberg JM, et al. Mitochondrial pathology and glycolytic shift during proximal tubule atrophy after ischemic AKI. J Am Soc Nephrol. 2016; 27:3356–67.
Article
23. Zhang J, Wang YT, Miller JH, Day MM, Munger JC, Brookes PS. Accumulation of succinate in cardiac ischemia primarily occurs via canonical krebs cycle activity. Cell Rep. 2018; 23:2617–28.
Article
24. Spinelli JB, Rosen PC, Sprenger HG, Puszynska AM, Mann JL, Roessler JM, et al. Fumarate is a terminal electron acceptor in the mammalian electron transport chain. Science. 2021; 374:1227–37.
Article
25. Beach TE, Prag HA, Pala L, Logan A, Huang MM, Gruszczyk AV, et al. Targeting succinate dehydrogenase with malonate ester prodrugs decreases renal ischemia reperfusion injury. Redox Biol. 2020; 36:101640.
Article
26. Archer SL. Mitochondrial dynamics: mitochondrial fission and fusion in human diseases. N Engl J Med. 2013; 369:2236–51.
Article
27. Xiao X, Hu Y, Quirós PM, Wei Q, López-Otín C, Dong Z. OMA1 mediates OPA1 proteolysis and mitochondrial fragmentation in experimental models of ischemic kidney injury. Am J Physiol Renal Physiol. 2014; 306:F1318–26.
Article
28. Zhan M, Brooks C, Liu F, Sun L, Dong Z. Mitochondrial dynamics: regulatory mechanisms and emerging role in renal pathophysiology. Kidney Int. 2013; 83:568–81.
Article
29. McBride H, Soubannier V. Mitochondrial function: OMA1 and OPA1, the grandmasters of mitochondrial health. Curr Biol. 2010; 20:R274–6.
Article
30. Liu Z, Li H, Su J, Xu S, Zhu F, Ai J, et al. Numb depletion promotes Drp1-mediated mitochondrial fission and exacerbates mitochondrial fragmentation and dysfunction in acute kidney injury. Antioxid Redox Signal. 2019; 30:1797–816.
Article
31. Cho SG, Xiao X, Wang S, Gao H, Rafikov R, Black S, et al. Bif-1 Interacts with prohibitin-2 to regulate mitochondrial inner membrane during cell stress and apoptosis. J Am Soc Nephrol. 2019; 30:1174–91.
Article
32. Qin N, Cai T, Ke Q, Yuan Q, Luo J, Mao X, et al. UCP2-dependent improvement of mitochondrial dynamics protects against acute kidney injury. J Pathol. 2019; 247:392–405.
Article
33. Su CT, See DH, Huang YJ, Jao TM, Liu SY, Chou CY, et al. LTBP4 protects against renal fibrosis via mitochondrial and vascular impacts. Circ Res. 2023; 133:71–85.
Article
34. Ge P, Dawson VL, Dawson TM. PINK1 and Parkin mitochondrial quality control: a source of regional vulnerability in Parkinson’s disease. Mol Neurodegener. 2020; 15:20.
Article
35. Durcan TM, Fon EA. The three ‘P’s of mitophagy: PARKIN, PINK1, and post-translational modifications. Genes Dev. 2015; 29:989–99.
Article
36. Rüb C, Wilkening A, Voos W. Mitochondrial quality control by the Pink1/Parkin system. Cell Tissue Res. 2017; 367:111–23.
Article
37. Grenier K, McLelland GL, Fon EA. Parkin- and PINK1-dependent mitophagy in neurons: will the real pathway please stand up? Front Neurol. 2013; 4:100.
Article
38. Tang C, Han H, Yan M, Zhu S, Liu J, Liu Z, et al. PINK1-PRKN/PARK2 pathway of mitophagy is activated to protect against renal ischemia-reperfusion injury. Autophagy. 2018; 14:880–97.
Article
39. Li N, Wang H, Jiang C, Zhang M. Renal ischemia/reperfusion-induced mitophagy protects against renal dysfunction via Drp1-dependent-pathway. Exp Cell Res. 2018; 369:27–33.
Article
40. Livingston MJ, Wang J, Zhou J, Wu G, Ganley IG, Hill JA, et al. Clearance of damaged mitochondria via mitophagy is important to the protective effect of ischemic preconditioning in kidneys. Autophagy. 2019; 15:2142–62.
Article
41. Su L, Zhang J, Wang J, Wang X, Cao E, Yang C, et al. Pannexin 1 targets mitophagy to mediate renal ischemia/reperfusion injury. Commun Biol. 2023; 6:889.
Article
42. Feng J, Li H, Zhang Y, Wang Q, Zhao S, Meng P, et al. Mammalian STE20-like kinase 1 deletion alleviates renal ischaemia-reperfusion injury via modulating mitophagy and the AMPK-YAP signalling pathway. Cell Physiol Biochem. 2018; 51:2359–76.
Article
43. Shi H, Qi H, Xie D, Zhuang J, Qi H, Dai Y, et al. Inhibition of ACSF2 protects against renal ischemia/reperfusion injury via mediating mitophagy in proximal tubular cells. Free Radic Biol Med. 2023; 198:68–82.
Article
44. Tang C, Han H, Liu Z, Liu Y, Yin L, Cai J, et al. Activation of BNIP3-mediated mitophagy protects against renal ischemia-reperfusion injury. Cell Death Dis. 2019; 10:677.
Article
45. Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012; 149:1060–72.
Article
46. Hosohata K, Harnsirikarn T, Chokesuwattanaskul S. Ferroptosis: a potential therapeutic target in acute kidney injury. Int J Mol Sci. 2022; 23:6583.
Article
47. Eleftheriadis T, Pissas G, Filippidis G, Liakopoulos V, Stefanidis I. Reoxygenation induces reactive oxygen species production and ferroptosis in renal tubular epithelial cells by activating aryl hydrocarbon receptor. Mol Med Rep. 2021; 23:41.
48. 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.
Article
49. Sun X, Huang N, Li P, Dong X, Yang J, Zhang X, et al. TRIM21 ubiquitylates GPX4 and promotes ferroptosis to aggravate ischemia/reperfusion-induced acute kidney injury. Life Sci. 2023; 321:121608.
Article
50. Liang Y, Liu Z, Qu L, Wang Y, Zhou Y, Liang L, et al. Inhibition of the IRE1/JNK pathway in renal tubular epithelial cells attenuates ferroptosis in acute kidney injury. Front Pharmacol. 2022; 13:927641.
Article
51. 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.
Article
52. Corridon PR. Enhancing the expression of a key mitochondrial enzyme at the inception of ischemia-reperfusion injury can boost recovery and halt the progression of acute kidney injury. Front Physiol. 2023; 14:1024238.
Article
53. Liu Z, Li Y, Li C, Yu L, Chang Y, Qu M. Delivery of coenzyme Q10 with mitochondria-targeted nanocarrier attenuates renal ischemia-reperfusion injury in mice. Mater Sci Eng C Mater Biol Appl. 2021; 131:112536.
Article
54. Kubat GB, Kartal Y, Atalay O, Ulger O, Ekinci O, Celik E, et al. Investigation of the effect of isolated mitochondria transplantation on renal ischemia-reperfusion injury in rats. Toxicol Appl Pharmacol. 2021; 433:115780.
Article
55. Doulamis IP, Guariento A, Duignan T, Kido T, Orfany A, Saeed MY, et al. Mitochondrial transplantation by intra-arterial injection for acute kidney injury. Am J Physiol Renal Physiol. 2020; 319:F403–13.
Article
56. Li L, Zhang L, Cao Y, Chen X, Gong H, Ma Y, et al. NDUFV1 attenuates renal ischemia-reperfusion injury by improving mitochondrial homeostasis. J Cell Mol Med. 2023; 27:1341–52.
Article
57. Doke T, Mukherjee S, Mukhi D, Dhillon P, Abedini A, Davis JG, et al. NAD+ precursor supplementation prevents mtRNA/RIG-I-dependent inflammation during kidney injury. Nat Metab. 2023; 5:414–30.
58. Morevati M, Egstrand S, Nordholm A, Mace ML, Andersen CB, Salmani R, et al. Effect of NAD+ boosting on kidney ischemia-reperfusion injury. PLoS One. 2021; 16:e0252554.
59. Mao H, Zhang Y, Xiong Y, Zhu Z, Wang L, Liu X. Mitochondria-Targeted antioxidant mitoquinone maintains mitochondrial homeostasis through the Sirt3-dependent pathway to mitigate oxidative damage caused by renal ischemia/reperfusion. Oxid Med Cell Longev. 2022; 2022:2213503.
Article
60. Cheng L, Yang X, Jian Y, Liu J, Ke X, Chen S, et al. SIRT3 deficiency exacerbates early-stage fibrosis after ischaemia-reperfusion-induced AKI. Cell Signal. 2022; 93:110284.
Article
61. Yu X, Xu M, Meng X, Li S, Liu Q, Bai M, et al. Nuclear receptor PXR targets AKR1B7 to protect mitochondrial metabolism and renal function in AKI. Sci Transl Med. 2020; 12:eaay7591.
Article
62. Wang D, Wang Y, Zou X, Shi Y, Liu Q, Huyan T, et al. FOXO1 inhibition prevents renal ischemia-reperfusion injury via cAMP-response element binding protein/PPAR-γ coactivator-1α-mediated mitochondrial biogenesis. Br J Pharmacol. 2020; 177:432–48.
Article
63. Hurtado KA, Janda J, Schnellmann RG. Lasmiditan promotes recovery from acute kidney injury through induction of mitochondrial biogenesis. Am J Physiol Renal Physiol. 2023; 324:F56–63.
Article
64. Gibbs WS, Collier JB, Morris M, Beeson CC, Megyesi J, Schnellmann RG. 5-HT1F receptor regulates mitochondrial homeostasis and its loss potentiates acute kidney injury and impairs renal recovery. Am J Physiol Renal Physiol. 2018; 315:F1119–28.
65. Tang Y, Leng YF, Wang W, Zhang J, Yuan TL, Wang J. Protective effect of Saxagliptin on diabetic rats with renal ischemia reperfusion injury by targeting oxidative stress and mitochondrial apoptosis pathway through activating Nrf-2/HO-1 signaling. Transpl Immunol. 2023; 76:101762.
Article
66. Barati A, Rahbar Saadat Y, Meybodi SM, Nouraei S, Moradi K, Kamrani Moghaddam F, et al. Eplerenone reduces renal ischaemia/reperfusion injury by modulating Klotho, NF-κB and SIRT1/SIRT3/PGC-1α signalling pathways. J Pharm Pharmacol. 2023; 75:819–27.
Article
67. Li J, Jiang Y, Dai Q, Yu Y, Lv X, Zhang Y, et al. Protective effects of mefunidone on ischemia-reperfusion injury/folic acid-induced acute kidney injury. Front Pharmacol. 2022; 13:1043945.
Article
68. Kawabata C, Hirakawa Y, Inagi R, Nangaku M. Acetate attenuates kidney fibrosis in an oxidative stress-dependent manner. Physiol Rep. 2023; 11:e15774.
Article
69. Liu D, Shu G, Jin F, Qi J, Xu X, Du Y, et al. ROS-responsive chitosan-SS31 prodrug for AKI therapy via rapid distribution in the kidney and long-term retention in the renal tubule. Sci Adv. 2020; 6:eabb7422.
Article
70. Nowak G, Megyesi J. γ-tocotrienol protects against mitochondrial dysfunction, energy deficits, morphological damage, and decreases in renal functions after renal ischemia. Int J Mol Sci. 2021; 22:12674.
Article
71. Ding M, Tolbert E, Birkenbach M, Gohh R, Akhlaghi F, Ghonem NS. Treprostinil reduces mitochondrial injury during rat renal ischemia-reperfusion injury. Biomed Pharmacother. 2021; 141:111912.
Article
72. Ji X, Chu L, Su D, Sun J, Song P, Sun S, et al. MRPL12-ANT3 interaction involves in acute kidney injury via regulating MPTP of tubular epithelial cells. iScience. 2023; 26:106656.
Article
73. Wang J, Zhu P, Li R, Ren J, Zhang Y, Zhou H. Bax inhibitor 1 preserves mitochondrial homeostasis in acute kidney injury through promoting mitochondrial retention of PHB2. Theranostics. 2020; 10:384–97.
Article
74. Thomas K, Zondler L, Ludwig N, Kardell M, Lüneburg C, Henke K, et al. Glutamine prevents acute kidney injury by modulating oxidative stress and apoptosis in tubular epithelial cells. JCI Insight. 2022; 7:e163161.
Article
75. Wang S, Zhu H, Li R, Mui D, Toan S, Chang X, et al. DNA-PKcs interacts with and phosphorylates Fis1 to induce mitochondrial fragmentation in tubular cells during acute kidney injury. Sci Signal. 2022; 15:eabh1121.
Article
76. Shen L, Zhang Q, Tu S, Qin W. SIRT3 mediates mitofusin 2 ubiquitination and degradation to suppress ischemia reperfusion-induced acute kidney injury. Exp Cell Res. 2021; 408:112861.
Article
77. Yang W, Li X, He L, Zhu S, Lai S, Zhang X, et al. Empagliflozin improves renal ischemia-reperfusion injury by reducing inflammation and enhancing mitochondrial fusion through AMPK-OPA1 pathway promotion. Cell Mol Biol Lett. 2023; 28:42.
Article
78. Wang Y, Liu Q, Cai J, Wu P, Wang D, Shi Y, et al. Emodin prevents renal ischemia-reperfusion injury via suppression of CAMKII/DRP1-mediated mitochondrial fission. Eur J Pharmacol. 2022; 916:174603.
Article
79. Wang Q, Xu J, Li X, Liu Z, Han Y, Xu X, et al. Sirt3 modulate renal ischemia-reperfusion injury through enhancing mitochondrial fusion and activating the ERK-OPA1 signaling pathway. J Cell Physiol. 2019; 234:23495–506.
Article
80. Fu ZJ, Wang ZY, Xu L, Chen XH, Li XX, Liao WT, et al. HIF-1α-BNIP3-mediated mitophagy in tubular cells protects against renal ischemia/reperfusion injury. Redox Biol. 2020; 36:101671.
Article
81. Sun Z, Gao Z, Wu J, Zheng X, Jing S, Wang W. MSC-derived extracellular vesicles activate mitophagy to alleviate renal ischemia/reperfusion injury via the miR-223-3p/NLRP3 axis. Stem Cells Int. 2022; 2022:6852661.
Article
82. Kamarauskaite J, Baniene R, Trumbeckas D, Strazdauskas A, Trumbeckaite S. Increased succinate accumulation induces ROS generation in in vivo ischemia/reperfusion-affected rat kidney mitochondria. Biomed Res Int. 2020; 2020:8855585.
Article
83. Scantlebery AM, Tammaro A, Mills JD, Rampanelli E, Kors L, Teske GJ, et al. The dysregulation of metabolic pathways and induction of the pentose phosphate pathway in renal ischaemia-reperfusion injury. J Pathol. 2021; 253:404–14.
Article
84. Gao Z, Zhang C, Peng F, Chen Q, Zhao Y, Chen L, et al. Hypoxic mesenchymal stem cell-derived extracellular vesicles ameliorate renal fibrosis after ischemia-reperfusion injure by restoring CPT1A mediated fatty acid oxidation. Stem Cell Res Ther. 2022; 13:191.
Article
85. Kim IY, Song SH, Seong EY, Lee DW, Bae SS, Lee SB. Akt1 is involved in renal fibrosis and tubular apoptosis in a murine model of acute kidney injury-to-chronic kidney disease transition. Exp Cell Res. 2023; 424:113509.
Article
86. Lin HY, Chen Y, Chen YH, Ta AP, Lee HC, MacGregor GR, et al. Tubular mitochondrial AKT1 is activated during ischemia reperfusion injury and has a critical role in predisposition to chronic kidney disease. Kidney Int. 2021; 99:870–84.
Article
87. Granata S, Votrico V, Spadaccino F, Catalano V, Netti GS, Ranieri E, et al. Oxidative stress and ischemia/reperfusion injury in kidney transplantation: focus on ferroptosis, mitophagy and new antioxidants. Antioxidants (Basel). 2022; 11:769.
Article
88. Song J, Sheng J, Lei J, Gan W, Yang Y. Mitochondrial targeted antioxidant SKQ1 ameliorates acute kidney injury by inhibiting ferroptosis. Oxid Med Cell Longev. 2022; 2022:2223957.
Article
89. Batirel S, Bozaykut P, Mutlu Altundag E, Kartal Ozer N, Mantzoros CS. The effect of Irisin on antioxidant system in liver. Free Radic Biol Med. 2014; 75(Suppl 1):S16.
Article
90. Zhang J, Bi J, Ren Y, Du Z, Li T, Wang T, et al. Involvement of GPX4 in irisin’s protection against ischemia reperfusion-induced acute kidney injury. J Cell Physiol. 2021; 236:931–45.
Article
91. Friedmann Angeli JP, Schneider M, Proneth B, Tyurina YY, Tyurin VA, Hammond VJ, et al. Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat Cell Biol. 2014; 16:1180–91.
Article
92. Tschopp J. Mitochondria: sovereign of inflammation? Eur J Immunol. 2011; 41:1196–202.
Article
93. Su X, Liu B, Wang S, Wang Y, Zhang Z, Zhou H, et al. NLRP3 inflammasome: a potential therapeutic target to minimize renal ischemia/reperfusion injury during transplantation. Transpl Immunol. 2022; 75:101718.
Article
94. Yin L, Zhao H, Zhang H, Li Y, Dong Y, Ju H, et al. Remdesivir alleviates acute kidney injury by inhibiting the activation of NLRP3 inflammasome. Front Immunol. 2021; 12:652446.
Article
95. Cao JY, Zhou LT, Li ZL, Yang Y, Liu BC, Liu H. Dopamine D1 receptor agonist A68930 attenuates acute kidney injury by inhibiting NLRP3 inflammasome activation. J Pharmacol Sci. 2020; 143:226–33.
Article
Full Text Links
  • JYMS
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr