Mitochondrial Contribution to Inflammation in Diabetic Kidney Disease
Abstract
:1. Introduction
2. Mitochondrial Regulation of Inflammation
2.1. cGAS-STING Signaling
2.2. RLR Signaling
2.3. Inflammasome Signaling
2.4. TLR Signaling
2.5. NF-κB Signaling
3. Mitochondrial Regulation of Cell Death
3.1. BAX-BAK1 Signaling
3.2. Cardiolipin
3.3. Mitophagy
3.4. Pyroptosis and Ferroptosis
4. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Jha, V.; Garcia-Garcia, G.; Iseki, K.; Li, Z.; Naicker, S.; Plattner, B.; Saran, R.; Wang, A.Y.; Yang, C.W. Chronic kidney disease: Global dimension and perspectives. Lancet 2013, 382, 260–272. [Google Scholar] [CrossRef]
- USRDS. 2021 USRDS Annual Data Report: Epidemiology of kidney disease in the United States; National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases: Bethesda, MD, USA, 2021.
- De Boer, I.H.; Khunti, K.; Sadusky, T.; Tuttle, K.R.; Neumiller, J.J.; Rhee, C.M.; Rosas, S.E.; Rossing, P.; Bakris, G. Diabetes management in chronic kidney disease: A consensus report by the American Diabetes Association (ADA) and Kidney Disease: Improving Global Outcomes (KDIGO). Kidney Int. 2022, 102, 974–989. [Google Scholar] [CrossRef]
- Skrabic, R.; Kumric, M.; Vrdoljak, J.; Rusic, D.; Skrabic, I.; Vilovic, M.; Martinovic, D.; Duplancic, V.; Ticinovic Kurir, T.; Bozic, J. SGLT2 Inhibitors in Chronic Kidney Disease: From Mechanisms to Clinical Practice. Biomedicines 2022, 10, 2458. [Google Scholar] [CrossRef]
- Kroemer, G.; Galassi, C.; Zitvogel, L.; Galluzzi, L. Immunogenic cell stress and death. Nat. Immunol. 2022, 23, 487–500. [Google Scholar] [CrossRef]
- Roger, A.J.; Muñoz-Gómez, S.A.; Kamikawa, R. The Origin and Diversification of Mitochondria. Curr. Biol. 2017, 27, R1177–R1192. [Google Scholar] [CrossRef] [Green Version]
- Galluzzi, L.; Vitale, I.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Agostinis, P.; Alnemri, E.S.; Altucci, L.; Amelio, I.; Andrews, D.W.; et al. Molecular mechanisms of cell death: Recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018, 25, 486–541. [Google Scholar] [CrossRef]
- Wang, Z.; Ying, Z.; Bosy-Westphal, A.; Zhang, J.; Schautz, B.; Later, W.; Heymsfield, S.B.; Müller, M.J. Specific metabolic rates of major organs and tissues across adulthood: Evaluation by mechanistic model of resting energy expenditure. Am. J. Clin. Nutr. 2010, 92, 1369–1377. [Google Scholar] [CrossRef] [Green Version]
- Ducasa, G.M.; Mitrofanova, A.; Mallela, S.K.; Liu, X.; Molina, J.; Sloan, A.; Pedigo, C.E.; Ge, M.; Santos, J.V.; Hernandez, Y.; et al. ATP-binding cassette A1 deficiency causes cardiolipin-driven mitochondrial dysfunction in podocytes. J. Clin. Investig. 2019, 129, 3387–3400. [Google Scholar] [CrossRef] [Green Version]
- Zhan, M.; Usman, I.M.; Sun, L.; Kanwar, Y.S. Disruption of Renal Tubular Mitochondrial Quality Control by Myo-Inositol Oxygenase in Diabetic Kidney Disease. J. Am. Soc. Nephrol. 2015, 26, 1304–1321. [Google Scholar] [CrossRef] [Green Version]
- Na, K.R.; Jeong, J.Y.; Shin, J.A.; Chang, Y.K.; Suh, K.S.; Lee, K.W.; Choi, D.E. Mitochondrial Dysfunction in Podocytes Caused by CRIF1 Deficiency Leads to Progressive Albuminuria and Glomerular Sclerosis in Mice. Int. J. Mol. Sci. 2021, 22, 4827. [Google Scholar] [CrossRef]
- Galvan, D.L.; Green, N.H.; Danesh, F.R. The hallmarks of mitochondrial dysfunction in chronic kidney disease. Kidney Int. 2017, 92, 1051–1057. [Google Scholar] [CrossRef]
- Abe, Y.; Sakairi, T.; Kajiyama, H.; Shrivastav, S.; Beeson, C.; Kopp, J.B. Bioenergetic characterization of mouse podocytes. Am. J. Physiol. Cell Physiol. 2010, 299, C464–C476. [Google Scholar] [CrossRef]
- Fink, B.D.; Herlein, J.A.; O’Malley, Y.; Sivitz, W.I. Endothelial cell and platelet bioenergetics: Effect of glucose and nutrient composition. PLoS ONE 2012, 7, e39430. [Google Scholar] [CrossRef] [Green Version]
- Czajka, A.; Malik, A.N. Hyperglycemia induced damage to mitochondrial respiration in renal mesangial and tubular cells: Implications for diabetic nephropathy. Redox Biol. 2016, 10, 100–107. [Google Scholar] [CrossRef] [Green Version]
- Brinkkoetter, P.T.; Bork, T.; Salou, S.; Liang, W.; Mizi, A.; Özel, C.; Koehler, S.; Hagmann, H.H.; Ising, C.; Kuczkowski, A.; et al. Anaerobic Glycolysis Maintains the Glomerular Filtration Barrier Independent of Mitochondrial Metabolism and Dynamics. Cell Rep. 2019, 27, 1551–1566.e5. [Google Scholar] [CrossRef] [Green Version]
- Imasawa, T.; Obre, E.; Bellance, N.; Lavie, J.; Imasawa, T.; Rigothier, C.; Delmas, Y.; Combe, C.; Lacombe, D.; Benard, G.; et al. High glucose repatterns human podocyte energy metabolism during differentiation and diabetic nephropathy. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2017, 31, 294–307. [Google Scholar] [CrossRef] [Green Version]
- Yamazaki, T.; Kirchmair, A.; Sato, A.; Buqué, A.; Rybstein, M.; Petroni, G.; Bloy, N.; Finotello, F.; Stafford, L.; Navarro Manzano, E.; et al. Mitochondrial DNA drives abscopal responses to radiation that are inhibited by autophagy. Nat. Immunol. 2020, 21, 1160–1171. [Google Scholar] [CrossRef]
- Yatim, N.; Jusforgues-Saklani, H.; Orozco, S.; Schulz, O.; Barreira da Silva, R.; Reis e Sousa, C.; Green, D.R.; Oberst, A.; Albert, M.L. RIPK1 and NF-κB signaling in dying cells determines cross-priming of CD8⁺ T cells. Science 2015, 350, 328–334. [Google Scholar] [CrossRef] [Green Version]
- Bakhoum, S.F.; Ngo, B.; Laughney, A.M.; Cavallo, J.A.; Murphy, C.J.; Ly, P.; Shah, P.; Sriram, R.K.; Watkins, T.B.K.; Taunk, N.K.; et al. Chromosomal instability drives metastasis through a cytosolic DNA response. Nature 2018, 553, 467–472. [Google Scholar] [CrossRef] [Green Version]
- Li, H.; Zimmerman, S.E.; Weyemi, U. Genomic instability and metabolism in cancer. Int. Rev. Cell Mol. Biol. 2021, 364, 241–265. [Google Scholar]
- Cheng, Y.; Sun, Y.; Wang, H.; Yan, Y.; Ding, C.; Sun, J. Chicken STING Mediates Activation of the IFN Gene Independently of the RIG-I Gene. J. Immunol. 2015, 195, 3922–3936. [Google Scholar] [CrossRef]
- Zhong, B.; Yang, Y.; Li, S.; Wang, Y.Y.; Li, Y.; Diao, F.; Lei, C.; He, X.; Zhang, L.; Tien, P.; et al. The adaptor protein MITA links virus-sensing receptors to IRF3 transcription factor activation. Immunity 2008, 29, 538–550. [Google Scholar] [CrossRef] [Green Version]
- Barber, G.N. STING: Infection, inflammation and cancer. Nat. Rev. Immunol. 2015, 15, 760–770. [Google Scholar] [CrossRef] [Green Version]
- Burdette, D.L.; Vance, R.E. STING and the innate immune response to nucleic acids in the cytosol. Nat. Immunol. 2013, 14, 19–26. [Google Scholar] [CrossRef]
- Dhanwani, R.; Takahashi, M.; Sharma, S. Cytosolic sensing of immuno-stimulatory DNA, the enemy within. Curr. Opin. Immunol. 2018, 50, 82–87. [Google Scholar] [CrossRef] [Green Version]
- Chung, K.W.; Dhillon, P.; Huang, S.; Sheng, X.; Shrestha, R.; Qiu, C.; Kaufman, B.A.; Park, J.; Pei, L.; Baur, J.; et al. Mitochondrial Damage and Activation of the STING Pathway Lead to Renal Inflammation and Fibrosis. Cell Metab. 2019, 30, 784–799.e5. [Google Scholar] [CrossRef]
- Mitrofanova, A.; Fontanella, A.; Tolerico, M.; Mallela, S.K.; Molina, J.; Kim, J.J.; Burke, G.; Merscher, S.; Fornoni, A. STING activation causes proteinuria in mice and contributes to glomerular disease. Kidney Int. Rep. 2022, 7, S155–S156. [Google Scholar] [CrossRef]
- Mitrofanova, A.; Fontanella, A.; Tolerico, M.; Mallela, S.; Molina, J.; Zuo, Y.; Boulina, M.; Kim, J.-J.; Varona Santos, J.; Ge, M.; et al. Activation of Stimulator of Interferon Genes (STING) Causes Proteinuria and Contributes to Glomerular Diseases. J. Am. Soc. Nephrol. 2022, 33. [Google Scholar] [CrossRef]
- Khedr, S.; Dissanayake, L.V.; Palygin, O.; Staruschenko, A. Potential Role of cGAS-STING Pathway in the Induction of Diabetic Kidney Disease. FASEB J. 2020, 34, 1. [Google Scholar] [CrossRef]
- Sharma, K.; Karl, B.; Mathew, A.V.; Gangoiti, J.A.; Wassel, C.L.; Saito, R.; Pu, M.; Sharma, S.; You, Y.H.; Wang, L.; et al. Metabolomics reveals signature of mitochondrial dysfunction in diabetic kidney disease. J. Am. Soc. Nephrol. JASN 2013, 24, 1901–1912. [Google Scholar] [CrossRef] [Green Version]
- Al-Kafaji, G.; Aljadaan, A.; Kamal, A.; Bakhiet, M. Peripheral blood mitochondrial DNA copy number as a novel potential biomarker for diabetic nephropathy in type 2 diabetes patients. Exp. Ther. Med. 2018, 16, 1483–1492. [Google Scholar] [CrossRef]
- Cao, H.; Wu, J.; Luo, J.; Chen, X.; Yang, J.; Fang, L. Urinary mitochondrial DNA: A potential early biomarker of diabetic nephropathy. Diabetes Metab. Res. Rev. 2019, 35, e3131. [Google Scholar] [CrossRef]
- Jiang, H.; Shao, X.; Jia, S.; Qu, L.; Weng, C.; Shen, X.; Wang, Y.; Huang, H.; Wang, Y.; Wang, C.; et al. The Mitochondria-Targeted Metabolic Tubular Injury in Diabetic Kidney Disease. Cell Physiol. Biochem. 2019, 52, 156–171. [Google Scholar]
- Qi, H.; Casalena, G.; Shi, S.; Yu, L.; Ebefors, K.; Sun, Y.; Zhang, W.; D’Agati, V.; Schlondorff, D.; Haraldsson, B.; et al. Glomerular Endothelial Mitochondrial Dysfunction Is Essential and Characteristic of Diabetic Kidney Disease Susceptibility. Diabetes 2017, 66, 763–778. [Google Scholar] [CrossRef] [Green Version]
- Kakimoto, M.; Inoguchi, T.; Sonta, T.; Yu, H.Y.; Imamura, M.; Etoh, T.; Hashimoto, T.; Nawata, H. Accumulation of 8-Hydroxy-2′-Deoxyguanosine and Mitochondrial DNA Deletion in Kidney of Diabetic Rats. Diabetes 2002, 51, 1588–1595. [Google Scholar] [CrossRef] [Green Version]
- Akhtar, S.; Siragy, H.M. Pro-renin receptor suppresses mitochondrial biogenesis and function via AMPK/SIRT-1/ PGC-1α pathway in diabetic kidney. PLoS ONE 2019, 14, e0225728. [Google Scholar] [CrossRef] [Green Version]
- Ma, X.M.; Geng, K.; Law, B.Y.-K.; Wang, P.; Pu, Y.L.; Chen, Q.; Xu, H.W.; Tan, X.Z.; Jiang, Z.Z.; Xu, Y. Lipotoxicity-induced mtDNA release promotes diabetic cardiomyopathy by activating the cGAS-STING pathway in obesity-related diabetes. Cell Biol. Toxicol. 2022. [Google Scholar] [CrossRef]
- Hur, S. Double-Stranded RNA Sensors and Modulators in Innate Immunity. Annu. Rev. Immunol. 2019, 37, 349–375. [Google Scholar] [CrossRef] [PubMed]
- Dhir, A.; Dhir, S.; Borowski, L.S.; Jimenez, L.; Teitell, M.; Rötig, A.; Crow, Y.J.; Rice, G.I.; Duffy, D.; Tamby, C.; et al. Mitochondrial double-stranded RNA triggers antiviral signalling in humans. Nature 2018, 560, 238–242. [Google Scholar] [CrossRef] [PubMed]
- Tigano, M.; Vargas, D.C.; Tremblay-Belzile, S.; Fu, Y.; Sfeir, A. Nuclear sensing of breaks in mitochondrial DNA enhances immune surveillance. Nature 2021, 591, 477–481. [Google Scholar] [CrossRef]
- Di Mauro, S.; Scamporrino, A.; Filippello, A.; Di Marco, M.; Di Martino, M.T.; Scionti, F.; Di Pino, A.; Scicali, R.; Malaguarnera, R.; Purrello, F.; et al. Mitochondrial RNAs as Potential Biomarkers of Functional Impairment in Diabetic Kidney Disease. Int. J. Mol. Sci. 2022, 23, 8198. [Google Scholar] [CrossRef]
- Salem, R.M.; Todd, J.N.; Sandholm, N.; Cole, J.B.; Chen, W.-M.; Andrews, D.; Pezzolesi, M.G.; McKeigue, P.M.; Hiraki, L.T.; Qiu, C.; et al. Genome-Wide Association Study of Diabetic Kidney Disease Highlights Biology Involved in Glomerular Basement Membrane Collagen. J. Am. Soc. Nephrol. 2019, 30, 2000–2016. [Google Scholar] [CrossRef]
- Muruve, D.A.; Pétrilli, V.; Zaiss, A.K.; White, L.R.; Clark, S.A.; Ross, P.J.; Parks, R.J.; Tschopp, J. The inflammasome recognizes cytosolic microbial and host DNA and triggers an innate immune response. Nature 2008, 452, 103–107. [Google Scholar] [CrossRef]
- Shimada, K.; Crother, T.R.; Karlin, J.; Dagvadorj, J.; Chiba, N.; Chen, S.; Ramanujan, V.K.; Wolf, A.J.; Vergnes, L.; Ojcius, D.M.; et al. Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity 2012, 36, 401–414. [Google Scholar] [CrossRef] [Green Version]
- Nakahira, K.; Haspel, J.A.; Rathinam, V.A.; Lee, S.J.; Dolinay, T.; Lam, H.C.; Englert, J.A.; Rabinovitch, M.; Cernadas, M.; Kim, H.P.; et al. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat. Immunol. 2011, 12, 222–230. [Google Scholar] [CrossRef] [Green Version]
- Saxena, G.; Chen, J.; Shalev, A. Intracellular shuttling and mitochondrial function of thioredoxin-interacting protein. J. Biol. Chem. 2010, 285, 3997–4005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bauernfeind, F.; Bartok, E.; Rieger, A.; Franchi, L.; Núñez, G.; Hornung, V. Cutting edge: Reactive oxygen species inhibitors block priming, but not activation, of the NLRP3 inflammasome. J. Immunol. 2011, 187, 613–617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Billingham, L.K.; Stoolman, J.S.; Vasan, K.; Rodriguez, A.E.; Poor, T.A.; Szibor, M.; Jacobs, H.T.; Reczek, C.R.; Rashidi, A.; Zhang, P.; et al. Mitochondrial electron transport chain is necessary for NLRP3 inflammasome activation. Nat. Immunol. 2022, 23, 692–704. [Google Scholar] [CrossRef]
- Zhong, Z.; Liang, S.; Sanchez-Lopez, E.; He, F.; Shalapour, S.; Lin, X.J.; Wong, J.; Ding, S.; Seki, E.; Schnabl, B.; et al. New mitochondrial DNA synthesis enables NLRP3 inflammasome activation. Nature 2018, 560, 198–203. [Google Scholar] [CrossRef]
- Latz, E.; Xiao, T.S.; Stutz, A. Activation and regulation of the inflammasomes. Nat. Rev. Immunol. 2013, 13, 397–411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dang, E.V.; McDonald, J.G.; Russell, D.W.; Cyster, J.G. Oxysterol Restraint of Cholesterol Synthesis Prevents AIM2 Inflammasome Activation. Cell 2017, 171, 1057–1071.e11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shahzad, K.; Bock, F.; Dong, W.; Wang, H.; Kopf, S.; Kohli, S.; Al-Dabet, M.M.; Ranjan, S.; Wolter, J.; Wacker, C.; et al. Nlrp3-inflammasome activation in non-myeloid-derived cells aggravates diabetic nephropathy. Kidney Int. 2015, 87, 74–84. [Google Scholar] [CrossRef] [PubMed]
- Birnbaum, Y.; Bajaj, M.; Yang, H.C.; Ye, Y. Combined SGLT2 and DPP4 Inhibition Reduces the Activation of the Nlrp3/ASC Inflammasome and Attenuates the Development of Diabetic Nephropathy in Mice with Type 2 Diabetes. Cardiovasc. Drugs Ther. 2018, 32, 135–145. [Google Scholar] [CrossRef] [PubMed]
- Carlos, D.; Costa, F.R.; Pereira, C.A.; Rocha, F.A.; Yaochite, J.N.; Oliveira, G.G.; Carneiro, F.S.; Tostes, R.C.; Ramos, S.G.; Zamboni, D.S.; et al. Mitochondrial DNA Activates the NLRP3 Inflammasome and Predisposes to Type 1 Diabetes in Murine Model. Front. Immunol. 2017, 8, 164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pereira, C.A.; Carlos, D.; Ferreira, N.S.; Silva, J.F.; Zanotto, C.Z.; Zamboni, D.S.; Garcia, V.D.; Ventura, D.F.; Silva, J.S.; Tostes, R.C. Mitochondrial DNA Promotes NLRP3 Inflammasome Activation and Contributes to Endothelial Dysfunction and Inflammation in Type 1 Diabetes. Front. Physiol. 2019, 10, 1557. [Google Scholar] [CrossRef] [Green Version]
- Huang, Y.; Zhou, J.H.; Zhang, H.; Canfran-Duque, A.; Singh, A.K.; Perry, R.J.; Shulman, G.I.; Fernandez-Hernando, C.; Min, W. Brown adipose TRX2 deficiency activates mtDNA-NLRP3 to impair thermogenesis and protect against diet-induced insulin resistance. J. Clin. Investig. 2022, 132, e148852. [Google Scholar] [CrossRef]
- Nam, B.Y.; Jhee, J.H.; Park, J.; Kim, S.; Kim, G.; Park, J.T.; Yoo, T.-H.; Kang, S.-W.; Yu, J.-W.; Han, S.H. PGC-1α inhibits the NLRP3 inflammasome via preserving mitochondrial viability to protect kidney fibrosis. Cell Death Dis. 2022, 13, 31. [Google Scholar] [CrossRef]
- Shahzad, K.; Fatima, S.; Khawaja, H.; Elwakiel, A.; Gadi, I.; Ambreen, S.; Zimmermann, S.; Mertens, P.R.; Biemann, R.; Isermann, B. Podocyte-specific Nlrp3 inflammasome activation promotes diabetic kidney disease. Kidney Int. 2022, 102, 766–779. [Google Scholar] [CrossRef]
- Østergaard, J.A.; Jha, J.C.; Sharma, A.; Dai, A.; Choi, J.S.Y.; de Haan, J.B.; Cooper, M.E.; Jandeleit-Dahm, K. Adverse renal effects of NLRP3 inflammasome inhibition by MCC950 in an interventional model of diabetic kidney disease. Clin. Sci. 2022, 136, 167–180. [Google Scholar] [CrossRef]
- Knauf, F.; Asplin, J.R.; Granja, I.; Schmidt, I.M.; Moeckel, G.W.; David, R.J.; Flavell, R.A.; Aronson, P.S. NALP3-mediated inflammation is a principal cause of progressive renal failure in oxalate nephropathy. Kidney Int. 2013, 84, 895–901. [Google Scholar] [CrossRef] [Green Version]
- Bae, J.H.; Jo, S.I.; Kim, S.J.; Lee, J.M.; Jeong, J.H.; Kang, J.S.; Cho, N.J.; Kim, S.S.; Lee, E.Y.; Moon, J.S. Circulating Cell-Free mtDNA Contributes to AIM2 Inflammasome-Mediated Chronic Inflammation in Patients with Type 2 Diabetes. Cells 2019, 8, 328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Q.; Raoof, M.; Chen, Y.; Sumi, Y.; Sursal, T.; Junger, W.; Brohi, K.; Itagaki, K.; Hauser, C.J. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature 2010, 464, 104–107. [Google Scholar] [CrossRef] [PubMed]
- Tian, J.; Avalos, A.M.; Mao, S.Y.; Chen, B.; Senthil, K.; Wu, H.; Parroche, P.; Drabic, S.; Golenbock, D.; Sirois, C.; et al. Toll-like receptor 9-dependent activation by DNA-containing immune complexes is mediated by HMGB1 and RAGE. Nat. Immunol. 2007, 8, 487–496. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.Z.; Liu, Z.; Liu, J.; Ren, J.X.; Sun, T.S. Mitochondrial DNA induces inflammation and increases TLR9/NF-κB expression in lung tissue. Int. J. Mol. Med. 2014, 33, 817–824. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jing, R.; Hu, Z.K.; Lin, F.; He, S.; Zhang, S.S.; Ge, W.Y.; Dai, H.J.; Du, X.K.; Lin, J.Y.; Pan, L.H. Mitophagy-Mediated mtDNA Release Aggravates Stretching-Induced Inflammation and Lung Epithelial Cell Injury via the TLR9/MyD88/NF-κB Pathway. Front. Cell Dev. Biol. 2020, 8, 819. [Google Scholar] [CrossRef]
- Oka, T.; Hikoso, S.; Yamaguchi, O.; Taneike, M.; Takeda, T.; Tamai, T.; Oyabu, J.; Murakawa, T.; Nakayama, H.; Nishida, K.; et al. Mitochondrial DNA that escapes from autophagy causes inflammation and heart failure. Nature 2012, 485, 251–255. [Google Scholar] [CrossRef] [Green Version]
- Papadimitraki, E.; Tzardi, M.; Bertsias, G.; Sotsiou, E.; Boumpas, D. Glomerular expression of toll-like receptor-9 in lupus nephritis but not in normal kidneys: Implications for the amplification of the inflammatory response. Lupus 2009, 18, 831–835. [Google Scholar] [CrossRef]
- Machida, H.; Ito, S.; Hirose, T.; Takeshita, F.; Oshiro, H.; Nakamura, T.; Mori, M.; Inayama, Y.; Yan, K.; Kobayashi, N.; et al. Expression of Toll-like receptor 9 in renal podocytes in childhood-onset active and inactive lupus nephritis. Nephrol. Dial. Transplant. 2010, 25, 2430–2537. [Google Scholar] [CrossRef] [Green Version]
- Batsford, S.; Duermueller, U.; Seemayer, C.; Mueller, C.; Hopfer, H.; Mihatsch, M. Protein level expression of Toll-like receptors 2, 4 and 9 in renal disease. Nephrol. Dial. Transplant. 2011, 26, 1413–1416. [Google Scholar] [CrossRef] [Green Version]
- Frieri, M.; Samih, M.A.; Dzhindzhikhashvili, M.; Liu, H.; Balsam, L.; Rubinstein, S. Toll-like receptor 9 and vascular endothelial growth factor levels in human kidneys from lupus nephritis patients. J. Nephrol. 2012, 25, 1041–1046. [Google Scholar] [CrossRef]
- Bao, W.; Xia, H.; Liang, Y.; Ye, Y.; Lu, Y.; Xu, X.; Duan, A.; He, J.; Chen, Z.; Wu, Y.; et al. Toll-like Receptor 9 Can be Activated by Endogenous Mitochondrial DNA to Induce Podocyte Apoptosis. Sci. Rep. 2016, 6, 22579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sureshbabu, A.; Patino, E.; Ma, K.C.; Laursen, K.; Finkelsztein, E.J.; Akchurin, O.; Muthukumar, T.; Ryter, S.W.; Gudas, L.; Choi, A.M.K.; et al. RIPK3 promotes sepsis-induced acute kidney injury via mitochondrial dysfunction. JCI Insight 2018, 3, e98411. [Google Scholar] [CrossRef] [PubMed]
- Mitrofanova, A.; Fontanella, A.M.; Merscher, S.; Fornoni, A. Lipid deposition and metaflammation in diabetic kidney disease. Curr. Opin. Pharmacol. 2020, 55, 60–72. [Google Scholar] [CrossRef] [PubMed]
- Brennan, E.P.; Mohan, M.; McClelland, A.; Tikellis, C.; Ziemann, M.; Kaspi, A.; Gray, S.P.; Pickering, R.; Tan, S.M.; Ali-Shah, S.T.; et al. Lipoxins Regulate the Early Growth Response-1 Network and Reverse Diabetic Kidney Disease. J. Am. Soc. Nephrol. JASN 2018, 29, 1437–1448. [Google Scholar] [CrossRef] [Green Version]
- Woroniecka, K.I.; Park, A.S.; Mohtat, D.; Thomas, D.B.; Pullman, J.M.; Susztak, K. Transcriptome analysis of human diabetic kidney disease. Diabetes 2011, 60, 2354–2369. [Google Scholar] [CrossRef] [Green Version]
- Zhang, H.; Sun, S.C. NF-κB in inflammation and renal diseases. Cell Biosci. 2015, 5, 63. [Google Scholar] [CrossRef] [Green Version]
- Sun, S.C. The noncanonical NF-κB pathway. Immunol. Rev. 2012, 246, 125–140. [Google Scholar] [CrossRef] [Green Version]
- Giampazolias, E.; Zunino, B.; Dhayade, S.; Bock, F.; Cloix, C.; Cao, K.; Roca, A.; Lopez, J.; Ichim, G.; Proïcs, E.; et al. Mitochondrial permeabilization engages NF-κB-dependent anti-tumour activity under caspase deficiency. Nat. Cell Biol. 2017, 19, 1116–1129. [Google Scholar] [CrossRef] [Green Version]
- Foresto-Neto, O.; Albino, A.H.; Arias, S.C.A.; Faustino, V.D.; Zambom, F.F.F.; Cenedeze, M.A.; Elias, R.M.; Malheiros, D.; Camara, N.O.S.; Fujihara, C.K.; et al. NF-κB System Is Chronically Activated and Promotes Glomerular Injury in Experimental Type 1 Diabetic Kidney Disease. Front. Physiol. 2020, 11, 84. [Google Scholar] [CrossRef]
- Lee, F.T.; Cao, Z.; Long, D.M.; Panagiotopoulos, S.; Jerums, G.; Cooper, M.E.; Forbes, J.M. Interactions between angiotensin II and NF-kappaB-dependent pathways in modulating macrophage infiltration in experimental diabetic nephropathy. J. Am. Soc. Nephrol. JASN 2004, 15, 2139–2151. [Google Scholar] [CrossRef]
- Mezzano, S.; Aros, C.; Droguett, A.; Burgos, M.E.; Ardiles, L.; Flores, C.; Schneider, H.; Ruiz-Ortega, M.; Egido, J. NF-kappaB activation and overexpression of regulated genes in human diabetic nephropathy. Nephrol. Dial. Transplant. 2004, 19, 2505–2512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lin, M.; Yiu, W.H.; Wu, H.J.; Chan, L.Y.; Leung, J.C.; Au, W.S.; Chan, K.W.; Lai, K.N.; Tang, S.C. Toll-like receptor 4 promotes tubular inflammation in diabetic nephropathy. J. Am. Soc. Nephrol. JASN 2012, 23, 86–102. [Google Scholar] [CrossRef] [PubMed]
- Wei, M.; Li, Z.; Xiao, L.; Yang, Z. Effects of ROS-relative NF-κB signaling on high glucose-induced TLR4 and MCP-1 expression in podocyte injury. Mol. Immunol. 2015, 68, 261–271. [Google Scholar] [CrossRef] [PubMed]
- Bottero, V.; Busuttil, V.; Loubat, A.; Magné, N.; Fischel, J.L.; Milano, G.; Peyron, J.F. Activation of nuclear factor kappaB through the IKK complex by the topoisomerase poisons SN38 and doxorubicin: A brake to apoptosis in HeLa human carcinoma cells. Cancer Res. 2001, 61, 7785–7791. [Google Scholar] [PubMed]
- Cogswell, P.C.; Kashatus, D.F.; Keifer, J.A.; Guttridge, D.C.; Reuther, J.Y.; Bristow, C.; Roy, S.; Nicholson, D.W.; Baldwin, A.S., Jr. NF-kappa B and I kappa B alpha are found in the mitochondria. Evidence for regulation of mitochondrial gene expression by NF-kappa B. J. Biol. Chem. 2003, 278, 2963–2968. [Google Scholar] [CrossRef] [Green Version]
- Guseva, N.V.; Taghiyev, A.F.; Sturm, M.T.; Rokhlin, O.W.; Cohen, M.B. Tumor necrosis factor-related apoptosis-inducing ligand-mediated activation of mitochondria-associated nuclear factor-kappaB in prostatic carcinoma cell lines. Mol. Cancer Res. 2004, 2, 574–584. [Google Scholar] [CrossRef]
- Zamora, M.; Meroño, C.; Viñas, O.; Mampel, T. Recruitment of NF-kappaB into mitochondria is involved in adenine nucleotide translocase 1 (ANT1)-induced apoptosis. J. Biol. Chem. 2004, 279, 38415–38423. [Google Scholar] [CrossRef]
- Nan, J.; Hu, H.; Sun, Y.; Zhu, L.; Wang, Y.; Zhong, Z.; Zhao, J.; Zhang, N.; Wang, Y.; Wang, Y.; et al. TNFR2 Stimulation Promotes Mitochondrial Fusion via Stat3- and NF-kB-Dependent Activation of OPA1 Expression. Circ. Res. 2017, 121, 392–410. [Google Scholar] [CrossRef]
- Pazarentzos, E.; Mahul-Mellier, A.L.; Datler, C.; Chaisaklert, W.; Hwang, M.S.; Kroon, J.; Qize, D.; Osborne, F.; Al-Rubaish, A.; Al-Ali, A.; et al. IκΒα inhibits apoptosis at the outer mitochondrial membrane independently of NF-κB retention. EMBO J. 2014, 33, 2814–2828. [Google Scholar] [CrossRef] [Green Version]
- Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg Effect: The Metabolic Requirements of Cell Proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef] [Green Version]
- Psarra, A.-M.G.; Sekeris, C.E. Nuclear receptors and other nuclear transcription factors in mitochondria: Regulatory molecules in a new environment. Biochim. Biophys. Acta (BBA)-Mol. Cell Res. 2008, 1783, 1–11. [Google Scholar] [CrossRef] [Green Version]
- Johnson, R.F.; Witzel, I.-I.; Perkins, N.D. p53-Dependent Regulation of Mitochondrial Energy Production by the RelA Subunit of NF-κB. Cancer Res. 2011, 71, 5588–5597. [Google Scholar] [CrossRef] [PubMed]
- Zhong, Z.; Umemura, A.; Sanchez-Lopez, E.; Liang, S.; Shalapour, S.; Wong, J.; He, F.; Boassa, D.; Perkins, G.; Ali, S.R.; et al. NF-κB Restricts Inflammasome Activation via Elimination of Damaged Mitochondria. Cell 2016, 164, 896–910. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Flores-Romero, H.; Hohorst, L.; John, M.; Albert, M.C.; King, L.E.; Beckmann, L.; Szabo, T.; Hertlein, V.; Luo, X.; Villunger, A.; et al. BCL-2-family protein tBID can act as a BAX-like effector of apoptosis. EMBO J. 2022, 41, e108690. [Google Scholar] [CrossRef]
- Domizio, J.D.; Gulen, M.F.; Saidoune, F.; Thacker, V.V.; Yatim, A.; Sharma, K.; Nass, T.; Guenova, E.; Schaller, M.; Conrad, C.; et al. The cGAS-STING pathway drives type I IFN immunopathology in COVID-19. Nature 2022, 603, 145–151. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.; Gupta, R.; Blanco, L.P.; Yang, S.; Shteinfer-Kuzmine, A.; Wang, K.; Zhu, J.; Yoon, H.E.; Wang, X.; Kerkhofs, M.; et al. VDAC oligomers form mitochondrial pores to release mtDNA fragments and promote lupus-like disease. Science 2019, 366, 1531–1536. [Google Scholar] [CrossRef] [PubMed]
- Bonora, M.; Giorgi, C.; Pinton, P. Molecular mechanisms and consequences of mitochondrial permeability transition. Nat. Rev. Mol. Cell Biol. 2022, 23, 266–285. [Google Scholar] [CrossRef] [PubMed]
- Liu, L.; Zhang, L.; Zhao, J.; Guo, X.; Luo, Y.; Hu, W.; Zhao, T. Tumor Necrosis Factor Receptor-Associated Protein 1 Protects against Mitochondrial Injury by Preventing High Glucose-Induced mPTP Opening in Diabetes. Oxid. Med. Cell Longev. 2020, 2020, 6431517. [Google Scholar] [CrossRef]
- Ma, L.; Ainsworth, H.C.; Snipes, J.A.; Murea, M.; Choi, Y.A.; Langefeld, C.D.; Parks, J.S.; Bharadwaj, M.S.; Chou, J.W.; Hemal, A.K.; et al. APOL1 Kidney-Risk Variants Induce Mitochondrial Fission. Kidney Int. Rep. 2020, 5, 891–904. [Google Scholar] [CrossRef]
- Mei, S.; Li, L.; Wei, Q.; Hao, J.; Su, Y.; Mei, C.; Dong, Z. Double knockout of Bax and Bak from kidney proximal tubules reduces unilateral urethral obstruction associated apoptosis and renal interstitial fibrosis. Sci. Rep. 2017, 7, 44892. [Google Scholar] [CrossRef] [Green Version]
- Wei, Q.; Dong, G.; Chen, J.K.; Ramesh, G.; Dong, Z. Bax and Bak have critical roles in ischemic acute kidney injury in global and proximal tubule-specific knockout mouse models. Kidney Int. 2013, 84, 138–148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, J.; Mao, L.Q.; Polonsky, K.S.; Ren, D.C. Pancreatic β-Cell Death due to Pdx-1 Deficiency Requires Multi-BH Domain Protein Bax but Not Bak. J. Biol. Chem. 2016, 291, 13529–13534. [Google Scholar] [CrossRef] [PubMed]
- Lei, J.; Zhao, L.; Zhang, Y.; Wu, Y.; Liu, Y. High Glucose-Induced Podocyte Injury Involves Activation of Mammalian Target of Rapamycin (mTOR)-Induced Endoplasmic Reticulum (ER) Stress. Cell. Physiol. Biochem. 2018, 45, 2431–2443. [Google Scholar] [CrossRef]
- Liu, X.; Jiang, L.; Li, Y.; Huang, Y.; Hu, X.; Zhu, W.; Wang, X.; Wu, Y.; Meng, X.; Qi, X. Wogonin protects glomerular podocytes by targeting Bcl-2-mediated autophagy and apoptosis in diabetic kidney disease. Acta Pharmacol. Sin. 2022, 43, 96–110. [Google Scholar] [CrossRef]
- Lecocq, J.; Ballou, C.E. On the Structure Of Cardiolipin. Biochemistry 1964, 3, 976–980. [Google Scholar] [CrossRef]
- Arnarez, C.; Marrink, S.J.; Periole, X. Identification of cardiolipin binding sites on cytochrome c oxidase at the entrance of proton channels. Sci. Rep. 2013, 3, 1263. [Google Scholar] [CrossRef] [Green Version]
- Tyurina, Y.Y.; Domingues, R.M.; Tyurin, V.A.; Maciel, E.; Domingues, P.; Amoscato, A.A.; Bayir, H.; Kagan, V.E. Characterization of cardiolipins and their oxidation products by LC-MS analysis. Chem. Phys. Lipids 2014, 179, 3–10. [Google Scholar] [CrossRef] [Green Version]
- Chu, C.T.; Ji, J.; Dagda, R.K.; Jiang, J.F.; Tyurina, Y.Y.; Kapralov, A.A.; Tyurin, V.A.; Yanamala, N.; Shrivastava, I.H.; Mohammadyani, D.; et al. Cardiolipin externalization to the outer mitochondrial membrane acts as an elimination signal for mitophagy in neuronal cells. Nat. Cell Biol. 2013, 15, 1197–1205. [Google Scholar] [CrossRef] [Green Version]
- Chao, H.; Lin, C.; Zuo, Q.; Liu, Y.; Xiao, M.; Xu, X.; Li, Z.; Bao, Z.; Chen, H.; You, Y.; et al. Cardiolipin-Dependent Mitophagy Guides Outcome after Traumatic Brain Injury. J. Neurosci. 2019, 39, 1930–1943. [Google Scholar] [CrossRef] [Green Version]
- Iriondo, M.N.; Etxaniz, A.; Varela, Y.R.; Hervás, J.H.; Montes, L.R.; Goñi, F.M.; Alonso, A. LC3 subfamily in cardiolipin-mediated mitophagy: A comparison of the LC3A, LC3B and LC3C homologs. bioRxiv 2020. [Google Scholar] [CrossRef]
- Leslie, D.S.; Dascher, C.C.; Cembrola, K.; Townes, M.A.; Hava, D.L.; Hugendubler, L.C.; Mueller, E.; Fox, L.; Roura-Mir, C.; Moody, D.B.; et al. Serum lipids regulate dendritic cell CD1 expression and function. Immunology 2008, 125, 289–301. [Google Scholar] [CrossRef] [PubMed]
- Luévano-Martínez, L.A.; Forni, M.F.; dos Santos, V.T.; Souza-Pinto, N.C.; Kowaltowski, A.J. Cardiolipin is a key determinant for mtDNA stability and segregation during mitochondrial stress. Biochim. Biophys. Acta 2015, 1847, 587–598. [Google Scholar] [CrossRef] [PubMed]
- Chen, L.; Dong, J.; Liao, S.; Wang, S.; Wu, Z.; Zuo, M.; Liu, B.; Yan, C.; Chen, Y.; He, H.; et al. Loss of Sam50 in hepatocytes induces cardiolipin-dependent mitochondrial membrane remodeling to trigger mtDNA release and liver injury. Hepatology 2022, 76, 1389–1408. [Google Scholar] [CrossRef] [PubMed]
- Miyamoto, S.; Zhang, G.; Hall, D.; Oates, P.J.; Maity, S.; Madesh, M.; Han, X.; Sharma, K. Restoring mitochondrial superoxide levels with elamipretide (MTP-131) protects db/db mice against progression of diabetic kidney disease. J. Biol. Chem. 2020, 295, 7249–7260. [Google Scholar] [CrossRef] [Green Version]
- Tan, S.M.; Ziemann, M.; Thallas-Bonke, V.; Snelson, M.; Kumar, V.; Laskowski, A.; Nguyen, T.V.; Huynh, K.; Clarke, M.V.; Libianto, R.; et al. Complement C5a Induces Renal Injury in Diabetic Kidney Disease by Disrupting Mitochondrial Metabolic Agility. Diabetes 2020, 69, 83–98. [Google Scholar] [CrossRef]
- Gladkova, C.; Maslen, S.L.; Skehel, J.M.; Komander, D. Mechanism of parkin activation by PINK1. Nature 2018, 559, 410–414. [Google Scholar] [CrossRef]
- Wu, H.; Xue, D.; Chen, G.; Han, Z.; Huang, L.; Zhu, C.; Wang, X.; Jin, H.; Wang, J.; Zhu, Y.; et al. The BCL2L1 and PGAM5 axis defines hypoxia-induced receptor-mediated mitophagy. Autophagy 2014, 10, 1712–1725. [Google Scholar] [CrossRef] [Green Version]
- Chen, M.; Chen, Z.; Wang, Y.; Tan, Z.; Zhu, C.; Li, Y.; Han, Z.; Chen, L.; Gao, R.; Liu, L.; et al. Mitophagy receptor FUNDC1 regulates mitochondrial dynamics and mitophagy. Autophagy 2016, 12, 689–702. [Google Scholar] [CrossRef] [Green Version]
- Zhang, J.; Ney, P.A. Role of BNIP3 and NIX in cell death, autophagy, and mitophagy. Cell Death Differ. 2009, 16, 939–946. [Google Scholar] [CrossRef] [Green Version]
- Rogov, V.V.; Suzuki, H.; Marinković, M.; Lang, V.; Kato, R.; Kawasaki, M.; Buljubašić, M.; Šprung, M.; Rogova, N.; Wakatsuki, S.; et al. Phosphorylation of the mitochondrial autophagy receptor Nix enhances its interaction with LC3 proteins. Sci. Rep. 2017, 7, 1131. [Google Scholar] [CrossRef]
- Huang, C.; Zhang, Y.; Kelly, D.J.; Tan, C.Y.; Gill, A.; Cheng, D.; Braet, F.; Park, J.S.; Sue, C.M.; Pollock, C.A.; et al. Thioredoxin interacting protein (TXNIP) regulates tubular autophagy and mitophagy in diabetic nephropathy through the mTOR signaling pathway. Sci. Rep. 2016, 6, 29196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, K.; Dai, H.; Yuan, J.; Chen, J.; Lin, L.; Zhang, W.; Wang, L.; Zhang, J.; Li, K.; He, Y. Optineurin-mediated mitophagy protects renal tubular epithelial cells against accelerated senescence in diabetic nephropathy. Cell Death Dis. 2018, 9, 105. [Google Scholar] [CrossRef] [PubMed]
- Feng, J.; Lu, C.; Dai, Q.; Sheng, J.; Xu, M. SIRT3 Facilitates Amniotic Fluid Stem Cells to Repair Diabetic Nephropathy Through Protecting Mitochondrial Homeostasis by Modulation of Mitophagy. Cell Physiol. Biochem. 2018, 46, 1508–1524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, K.; Chen, J.; Wang, L.; Yang, J.; Xiao, F.; Wang, X.; Yuan, J.; Wang, L.; He, Y. Parkin ubiquitinates GATA4 and attenuates the GATA4/GAS1 signaling and detrimental effects on diabetic nephropathy. FASEB J. 2020, 34, 8858–8875. [Google Scholar] [CrossRef]
- Li, W.; Du, M.; Wang, Q.; Ma, X.; Wu, L.; Guo, F.; Ji, H.; Huang, F.; Qin, G. FoxO1 Promotes Mitophagy in the Podocytes of Diabetic Male Mice via the PINK1/Parkin Pathway. Endocrinology 2017, 158, 2155–2167. [Google Scholar] [CrossRef] [Green Version]
- Zhou, D.; Zhou, M.; Wang, Z.; Fu, Y.; Jia, M.; Wang, X.; Liu, M.; Zhang, Y.; Sun, Y.; Zhou, Y.; et al. Progranulin alleviates podocyte injury via regulating CAMKK/AMPK-mediated autophagy under diabetic conditions. J. Mol. Med. 2019, 97, 1507–1520. [Google Scholar] [CrossRef]
- Sun, J.; Zhu, H.; Wang, X.; Gao, Q.; Li, Z.; Huang, H. CoQ10 ameliorates mitochondrial dysfunction in diabetic nephropathy through mitophagy. J. Endocrinol. 2019, 240, 445–465. [Google Scholar] [CrossRef]
- Li, X.Y.; Wang, S.S.; Han, Z.; Han, F.; Chang, Y.P.; Yang, Y.; Xue, M.; Sun, B.; Chen, L.M. Triptolide Restores Autophagy to Alleviate Diabetic Renal Fibrosis through the miR-141-3p/PTEN/Akt/mTOR Pathway. Mol. Ther. Nucleic Acids 2017, 9, 48–56. [Google Scholar] [CrossRef] [Green Version]
- Liu, X.; Wang, W.; Song, G.; Wei, X.; Zeng, Y.; Han, P.; Wang, D.; Shao, M.; Wu, J.; Sun, H.; et al. Astragaloside IV ameliorates diabetic nephropathy by modulating the mitochondrial quality control network. PLoS ONE 2017, 12, e0182558. [Google Scholar] [CrossRef] [Green Version]
- Liu, X.; Lu, J.; Liu, S.; Huang, D.; Chen, M.; Xiong, G.; Li, S. Huangqi-Danshen decoction alleviates diabetic nephropathy in db/db mice by inhibiting PINK1/Parkin-mediated mitophagy. Am. J. Transl. Res. 2020, 12, 989–998. [Google Scholar]
- Lee, E.-J.; Kang, M.-K.; Kim, Y.-H.; Kim, D.Y.; Oh, H.; Kim, S.-I.; Oh, S.Y.; Kang, Y.-H. Dietary Chrysin Suppresses Formation of Actin Cytoskeleton and Focal Adhesion in AGE-Exposed Mesangial Cells and Diabetic Kidney: Role of Autophagy. Nutrients 2019, 11, 127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, K.; Zheng, X.; Pan, Z.; Yao, W.; Gao, X.; Wang, X.; Ding, X. Icariin Prevents Extracellular Matrix Accumulation and Ameliorates Experimental Diabetic Kidney Disease by Inhibiting Oxidative Stress via GPER Mediated p62-Dependent Keap1 Degradation and Nrf2 Activation. Front. Cell Dev. Biol. 2020, 8, 559. [Google Scholar] [CrossRef] [PubMed]
- Gao, C.; Fan, F.; Chen, J.; Long, Y.; Tang, S.; Jiang, C.; Xu, Y. FBW7 Regulates the Autophagy Signal in Mesangial Cells Induced by High Glucose. BioMed Res. Int. 2019, 2019, 6061594. [Google Scholar] [CrossRef] [PubMed]
- Lu, X.; Fan, Q.; Xu, L.; Li, L.; Yue, Y.; Xu, Y.; Su, Y.; Zhang, D.; Wang, L. Ursolic acid attenuates diabetic mesangial cell injury through the up-regulation of autophagy via miRNA-21/PTEN/Akt/mTOR suppression. PLoS ONE 2015, 10, e0117400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wen, D.; Tan, R.Z.; Zhao, C.Y.; Li, J.C.; Zhong, X.; Diao, H.; Lin, X.; Duan, D.D.; Fan, J.M.; Xie, X.S.; et al. Astragalus mongholicus Bunge and Panax notoginseng (Burkill) F.H. Chen Formula for Renal Injury in Diabetic Nephropathy-In Vivo and In Vitro Evidence for Autophagy Regulation. Front. Pharmacol. 2020, 11, 732. [Google Scholar] [CrossRef] [PubMed]
- Han, Y.-C.; Tang, S.-Q.; Liu, Y.-T.; Li, A.-M.; Zhan, M.; Yang, M.; Song, N.; Zhang, W.; Wu, X.-Q.; Peng, C.-H.; et al. AMPK agonist alleviate renal tubulointerstitial fibrosis via activating mitophagy in high fat and streptozotocin induced diabetic mice. Cell Death Dis. 2021, 12, 925. [Google Scholar] [CrossRef] [PubMed]
- Xiao, L.; Xu, X.; Zhang, F.; Wang, M.; Xu, Y.; Tang, D.; Wang, J.; Qin, Y.; Liu, Y.; Tang, C.; et al. The mitochondria-targeted antioxidant MitoQ ameliorated tubular injury mediated by mitophagy in diabetic kidney disease via Nrf2/PINK1. Redox Biol. 2017, 11, 297–311. [Google Scholar] [CrossRef]
- Jiang, X.S.; Chen, X.M.; Hua, W.; He, J.L.; Liu, T.; Li, X.J.; Wan, J.M.; Gan, H.; Du, X.G. PINK1/Parkin mediated mitophagy ameliorates palmitic acid-induced apoptosis through reducing mitochondrial ROS production in podocytes. Biochem. Biophys. Res. Commun. 2020, 525, 954–961. [Google Scholar] [CrossRef]
- Bergsbaken, T.; Cookson, B.T. Macrophage activation redirects yersinia-infected host cell death from apoptosis to caspase-1-dependent pyroptosis. PLoS Pathog. 2007, 3, e161. [Google Scholar] [CrossRef]
- Shi, J.; Zhao, Y.; Wang, K.; Shi, X.; Wang, Y.; Huang, H.; Zhuang, Y.; Cai, T.; Wang, F.; Shao, F. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature 2015, 526, 660–665. [Google Scholar] [CrossRef]
- Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, Z.W.; Zhang, J.; Li, X.; Wang, Y.; Fu, Y.H.; Gao, X.Y. A new research hot spot: The role of NLRP3 inflammasome activation, a key step in pyroptosis, in diabetes and diabetic complications. Life Sci. 2020, 240, 117138. [Google Scholar] [CrossRef] [PubMed]
- Cheng, Q.; Pan, J.; Zhou, Z.-l.; Yin, F.; Xie, H.-y.; Chen, P.-p.; Li, J.-y.; Zheng, P.-q.; Zhou, L.; Zhang, W.; et al. Caspase-11/4 and gasdermin D-mediated pyroptosis contributes to podocyte injury in mouse diabetic nephropathy. Acta Pharmacol. Sin. 2021, 42, 954–963. [Google Scholar] [CrossRef] [PubMed]
- Gao, Y.; Ma, Y.; Xie, D.; Jiang, H. ManNAc protects against podocyte pyroptosis via inhibiting mitochondrial damage and ROS/NLRP3 signaling pathway in diabetic kidney injury model. Int. Immunopharmacol. 2022, 107, 108711. [Google Scholar] [CrossRef]
- Wang, H.; Lv, D.; Jiang, S.; Hou, Q.; Zhang, L.; Li, S.; Zhu, X.; Xu, X.; Wen, J.; Zeng, C.; et al. Complement induces podocyte pyroptosis in membranous nephropathy by mediating mitochondrial dysfunction. Cell Death Dis. 2022, 13, 281. [Google Scholar] [CrossRef]
- Rogers, C.; Erkes, D.A.; Nardone, A.; Aplin, A.E.; Fernandes-Alnemri, T.; Alnemri, E.S. Gasdermin pores permeabilize mitochondria to augment caspase-3 activation during apoptosis and inflammasome activation. Nat. Commun. 2019, 10, 1689. [Google Scholar] [CrossRef] [Green Version]
- Rogers, C.; Fernandes-Alnemri, T.; Mayes, L.; Alnemri, D.; Cingolani, G.; Alnemri, E.S. Cleavage of DFNA5 by caspase-3 during apoptosis mediates progression to secondary necrotic/pyroptotic cell death. Nat. Commun. 2017, 8, 14128. [Google Scholar] [CrossRef] [Green Version]
- Wang, Y.; Gao, W.; Shi, X.; Ding, J.; Liu, W.; He, H.; Wang, K.; Shao, F. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature 2017, 547, 99–103. [Google Scholar] [CrossRef]
- Wu, W.Y.; Wang, Z.X.; Li, T.S.; Ding, X.Q.; Liu, Z.H.; Yang, J.; Fang, L.; Kong, L.D. SSBP1 drives high fructose-induced glomerular podocyte ferroptosis via activating DNA-PK/p53 pathway. Redox Biol. 2022, 52, 102303. [Google Scholar] [CrossRef]
- Zhang, Q.; Hu, Y.; Hu, J.-E.; Ding, Y.; Shen, Y.; Xu, H.; Chen, H.; Wu, N. Sp1-mediated upregulation of Prdx6 expression prevents podocyte injury in diabetic nephropathy via mitigation of oxidative stress and ferroptosis. Life Sci. 2021, 278, 119529. [Google Scholar] [CrossRef]
- Skonieczna, M.; Cieslar-Pobuda, A.; Saenko, Y.; Foksinski, M.; Olinski, R.; Rzeszowska-Wolny, J.; Wiechec, E. The Impact of DIDS-Induced Inhibition of Voltage-Dependent Anion Channels (VDAC) on Cellular Response of Lymphoblastoid Cells to Ionizing Radiation. Med. Chem. 2017, 13, 477–483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Y.; Bi, R.; Quan, F.; Cao, Q.; Lin, Y.; Yue, C.; Cui, X.; Yang, H.; Gao, X.; Zhang, D. Ferroptosis involves in renal tubular cell death in diabetic nephropathy. Eur. J. Pharmacol. 2020, 888, 173574. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Zheng, L.; Zhang, J.; Liu, X.; Wu, Z. Inhibition of ferroptosis by up-regulating Nrf2 delayed the progression of diabetic nephropathy. Free Radic. Biol. Med. 2021, 162, 435–449. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.; Kang, S.-W.; Joo, J.; Han, S.H.; Shin, H.; Nam, B.Y.; Park, J.; Yoo, T.-H.; Kim, G.; Lee, P.; et al. Characterization of ferroptosis in kidney tubular cell death under diabetic conditions. Cell Death Dis. 2021, 12, 160. [Google Scholar] [CrossRef] [PubMed]
Compound | Target | Model | Phenotype | Ref. |
---|---|---|---|---|
Metformin | HFD mice | ↓ mtROS production | ||
AMPK | STZ mice | ↑ PINK1 and Parkin protein levels | [137] | |
HK-2 cells | ↑ LC3-II and Atg5 levels | |||
Mitotempo | mtROS | HG-treated | ↓ mtROS production | [123] |
mouse primary RTECs | ↓ p16, p21, SAHF, SA-β-Gal, DcR2 | |||
MitoQ | ↓ oxidative stress | |||
Nrf2/PINK1 | db/db mice | ↓ caspase-3 expression | [138] | |
↑ Δψm | ||||
D-glucarate | HK-2 cells | Restored mitochondrial morphology | ||
MIOX | STZ mice | ↓ mtROS production | [10] | |
↓ BAX mediated apoptosis | ||||
CoQ10 | HG-treated | [128] | ||
Nrf2 | mGECs | ↑ mitophagy via PINK1 and Parkin | ||
Triptolide | ↑ autophagy and LC3-II levels | [129] | ||
PTEN/AKT/mTOR | HFD rats | ↓ p62 | ||
Palmitic acid | ↑ mitophagy and LC3 levels | |||
PINK1/Parkin | HFD rats | ↑ mtROS production | [139] | |
↑ apoptosis | ||||
Progranulin | HFD rats | [127] | ||
CAMKK-AMPK | STZ mice | ↑ autophagy and LC3 levels | ||
APF | HG-treated RMCs | [136] | ||
mTOR/PINK1/Parkin | STZ mice | ↑ mitophagy via PINK1 and Parkin | ||
Icariin | STZ rats | ↓ superoxide anion | [133] | |
Nrf2/GPER | HG-treated hGMCs | ↑ antioxidant enzymes activity | ||
Ursolic acid | ↑ LC3 levels | [135] | ||
PI3K/AKT/mTOR | HG-treated rGMCs | ↑ PTEN mRNA and protein expression | ||
Astragaloside IV | ↓ mitophagy via PINK1 and Parkin | [130] | ||
Not specified | db/db mice | ↓ renal DRP1, FIS1, MFF expression | ||
HDD | Not specified | db/db mice | ↓ mitophagy via PINK1 and Parkin | [131] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Mitrofanova, A.; Fontanella, A.M.; Burke, G.W.; Merscher, S.; Fornoni, A. Mitochondrial Contribution to Inflammation in Diabetic Kidney Disease. Cells 2022, 11, 3635. https://doi.org/10.3390/cells11223635
Mitrofanova A, Fontanella AM, Burke GW, Merscher S, Fornoni A. Mitochondrial Contribution to Inflammation in Diabetic Kidney Disease. Cells. 2022; 11(22):3635. https://doi.org/10.3390/cells11223635
Chicago/Turabian StyleMitrofanova, Alla, Antonio M. Fontanella, George W. Burke, Sandra Merscher, and Alessia Fornoni. 2022. "Mitochondrial Contribution to Inflammation in Diabetic Kidney Disease" Cells 11, no. 22: 3635. https://doi.org/10.3390/cells11223635