Skip to content
2000
Volume 31, Issue 40
  • ISSN: 0929-8673
  • E-ISSN: 1875-533X

Abstract

Kidney diseases, particularly Acute Kidney Injury (AKI) and Chronic Kidney Disease (CKD), are identified as global public health issues affecting millions of individuals. In addition, the frequency of renal diseases in the population has increased dramatically and rapidly in recent years. Renal disorders have become a significant public health burden. The pathophysiology of renal diseases is significantly connected with renal cell death, including apoptosis, necrosis, necroptosis, ferroptosis, pyroptosis, and autophagy, as is now recognized. Unlike other forms of cell death, pyroptosis is a unique planned cell death (PCD). Scientists have proven that pyroptosis is crucial in developing various disorders, and this phenomenon is gaining increasing attention. It is considered a novel method of inflammatory cell death. Intriguingly, inflammation is among the most significant pathological characteristics of renal disease. This study investigates the effects of pyroptosis on Acute Kidney Injury (AKI), Chronic Kidney Disease (CKD), Diabetic Nephropathy (DN), Immunoglobulin A (IgA) Nephropathy, and Lupus Nephritis (LN) to identify novel therapeutic targets for kidney diseases.

Loading

Article metrics loading...

/content/journals/cmc/10.2174/0109298673255656231003111621
2023-10-11
2024-11-01
Loading full text...

Full text loading...

References

  1. de VasconcelosN.M. LamkanfiM. Recent insights on inflammasomes, gasdermin pores, and pyroptosis.Cold Spring Harb. Perspect. Biol.2020125a03639210.1101/cshperspect.a03639231570336
    [Google Scholar]
  2. XiaS. HollingsworthL.R.IV WuH. Mechanism and regulation of gasdermin-mediated cell death.Cold Spring Harb. Perspect. Biol.2020123a03640010.1101/cshperspect.a03640031451512
    [Google Scholar]
  3. GalluzziL. VitaleI. AaronsonS.A. AbramsJ.M. AdamD. AgostinisP. AlnemriE.S. AltucciL. AmelioI. AndrewsD.W. Annicchiarico-PetruzzelliM. AntonovA.V. AramaE. BaehreckeE.H. BarlevN.A. BazanN.G. BernassolaF. BertrandM.J.M. BianchiK. BlagosklonnyM.V. BlomgrenK. BornerC. BoyaP. BrennerC. CampanellaM. CandiE. Carmona-GutierrezD. CecconiF. ChanF.K.M. ChandelN.S. ChengE.H. ChipukJ.E. CidlowskiJ.A. CiechanoverA. CohenG.M. ConradM. Cubillos-RuizJ.R. CzabotarP.E. D’AngiolellaV. DawsonT.M. DawsonV.L. De LaurenziV. De MariaR. DebatinK.M. DeBerardinisR.J. DeshmukhM. Di DanieleN. Di VirgilioF. DixitV.M. DixonS.J. DuckettC.S. DynlachtB.D. El-DeiryW.S. ElrodJ.W. FimiaG.M. FuldaS. García-SáezA.J. GargA.D. GarridoC. GavathiotisE. GolsteinP. GottliebE. GreenD.R. GreeneL.A. GronemeyerH. GrossA. HajnoczkyG. HardwickJ.M. HarrisI.S. HengartnerM.O. HetzC. IchijoH. JäätteläM. JosephB. JostP.J. JuinP.P. KaiserW.J. KarinM. KaufmannT. KeppO. KimchiA. KitsisR.N. KlionskyD.J. KnightR.A. KumarS. LeeS.W. LemastersJ.J. LevineB. LinkermannA. LiptonS.A. LockshinR.A. López-OtínC. LoweS.W. LueddeT. LugliE. MacFarlaneM. MadeoF. MalewiczM. MalorniW. ManicG. MarineJ.C. MartinS.J. MartinouJ.C. MedemaJ.P. MehlenP. MeierP. MelinoS. MiaoE.A. MolkentinJ.D. MollU.M. Muñoz-PinedoC. NagataS. NuñezG. OberstA. OrenM. OverholtzerM. PaganoM. PanaretakisT. PasparakisM. PenningerJ.M. PereiraD.M. PervaizS. PeterM.E. PiacentiniM. PintonP. PrehnJ.H.M. PuthalakathH. RabinovichG.A. RehmM. RizzutoR. RodriguesC.M.P. RubinszteinD.C. RudelT. RyanK.M. SayanE. ScorranoL. ShaoF. ShiY. SilkeJ. SimonH.U. SistiguA. StockwellB.R. StrasserA. SzabadkaiG. TaitS.W.G. TangD. TavernarakisN. ThorburnA. TsujimotoY. TurkB. Vanden BergheT. VandenabeeleP. Vander HeidenM.G. VillungerA. VirginH.W. VousdenK.H. VucicD. WagnerE.F. WalczakH. WallachD. WangY. WellsJ.A. WoodW. YuanJ. ZakeriZ. ZhivotovskyB. ZitvogelL. MelinoG. KroemerG. Molecular mechanisms of cell death: Recommendations of the nomenclature committee on cell death 2018.Cell Death Differ.201825348654110.1038/s41418‑017‑0012‑429362479
    [Google Scholar]
  4. KayagakiN. StoweI.B. LeeB.L. O’RourkeK. AndersonK. WarmingS. CuellarT. HaleyB. Roose-GirmaM. PhungQ.T. LiuP.S. LillJ.R. LiH. WuJ. KummerfeldS. ZhangJ. LeeW.P. SnipasS.J. SalvesenG.S. MorrisL.X. FitzgeraldL. ZhangY. BertramE.M. GoodnowC.C. DixitV.M. Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling.Nature2015526757566667110.1038/nature1554126375259
    [Google Scholar]
  5. LinJ. ChengA. ChengK. DengQ. ZhangS. LanZ. WangW. ChenJ. New insights into the mechanisms of pyroptosis and implications for diabetic kidney disease.Int. J. Mol. Sci.20202119705710.3390/ijms2119705732992874
    [Google Scholar]
  6. GuoH. XieM. ZhouC. ZhengM. The relevance of pyroptosis in the pathogenesis of liver diseases.Life Sci.2019223697310.1016/j.lfs.2019.02.06030831126
    [Google Scholar]
  7. CuevasS. PelegrínP. Pyroptosis and redox balance in kidney diseases.Antioxid. Redox Signal.2021351406010.1089/ars.2020.824333559516
    [Google Scholar]
  8. YangC. LongJ. ShiY. ZhouZ. WangJ. ZhaoM.H. WangH. ZhangL. CoreshJ. Healthcare resource utilisation for chronic kidney disease and other major non-communicable chronic diseases in China: A cross-sectional study.BMJ Open2022121e05188810.1136/bmjopen‑2021‑05188835027417
    [Google Scholar]
  9. BaoY.W. YuanY. ChenJ.H. LinW.Q. Kidney disease models: Tools to identify mechanisms and potential therapeutic targets.Zool. Res.2018392728629515089
    [Google Scholar]
  10. AndreevaL. DavidL. RawsonS. ShenC. PasrichaT. PelegrinP. WuH. NLRP3 cages revealed by full-length mouse NLRP3 structure control pathway activation.Cell20211842662996312.e2210.1016/j.cell.2021.11.01134861190
    [Google Scholar]
  11. NiuT. De RosnyC. ChautardS. ReyA. PatoliD. GroslambertM. CossonC. LagrangeB. ZhangZ. VisvikisO. HacotS. HologneM. WalkerO. WongJ. WangP. RicciR. HenryT. BoyerL. PetrilliV. PyB.F. NLRP3 phosphorylation in its LRR domain critically regulates inflammasome assembly.Nat. Commun.2021121586210.1038/s41467‑021‑26142‑w34615873
    [Google Scholar]
  12. XiaS. ZhangZ. MagupalliV.G. PabloJ.L. DongY. VoraS.M. WangL. FuT.M. JacobsonM.P. GrekaA. LiebermanJ. RuanJ. WuH. Gasdermin D pore structure reveals preferential release of mature interleukin-1.Nature2021593786060761110.1038/s41586‑021‑03478‑333883744
    [Google Scholar]
  13. LeveyA.S. JamesM.T. Acute kidney injury.Ann. Intern. Med.20171679ITC66ITC8010.7326/AITC20171107029114754
    [Google Scholar]
  14. FaubelS. EdelsteinC.L. Mechanisms and mediators of lung injury after acute kidney injury.Nat. Rev. Nephrol.2016121486010.1038/nrneph.2015.15826434402
    [Google Scholar]
  15. SawhneyS. TanZ. BlackC. MarksA. MclernonD.J. RonksleyP. JamesM.T. Validation of risk prediction models to inform clinical decisions after acute kidney injury.Am. J. Kidney Dis.2021781283710.1053/j.ajkd.2020.12.00833428996
    [Google Scholar]
  16. OzkokA. EdelsteinC.L. Pathophysiology of cisplatin-induced acute kidney injury.BioMed Res. Int.2014201411710.1155/2014/96782625165721
    [Google Scholar]
  17. GómezH. KellumJ.A. Sepsis-induced acute kidney injury.Curr. Opin. Crit. Care201622654655310.1097/MCC.000000000000035627661757
    [Google Scholar]
  18. FählingM. SeeligerE. PatzakA. PerssonP.B. Understanding and preventing contrast-induced acute kidney injury.Nat. Rev. Nephrol.201713316918010.1038/nrneph.2016.19628138128
    [Google Scholar]
  19. YangK. LiW.F. YuJ.F. YiC. HuangW.F. Diosmetin protects against ischemia/reperfusion-induced acute kidney injury in mice.J. Surg. Res.2017214697810.1016/j.jss.2017.02.06728624062
    [Google Scholar]
  20. XiaoC. ZhaoH. ZhuH. ZhangY. SuQ. ZhaoF. WangR. Tisp40 induces tubular epithelial cell GSDMD-mediated pyroptosis in renal ischemia-reperfusion injury via NF-κB signaling.Front. Physiol.20201190610.3389/fphys.2020.0090632903383
    [Google Scholar]
  21. XiaW. LiY. WuM. JinQ. WangQ. LiS. HuangS. ZhangA. ZhangY. JiaZ. Gasdermin E deficiency attenuates acute kidney injury by inhibiting pyroptosis and inflammation.Cell Death Dis.202112213910.1038/s41419‑021‑03431‑233542198
    [Google Scholar]
  22. ZangZ-D. YanJ. An analysis of clinical characteristics of septic acute kidney injury by using criteria of kidney disease: Improving Global Outcomes.Zhonghua Nei Ke Za Zhi201352429930423925356
    [Google Scholar]
  23. WangZ. GuZ. HouQ. ChenW. MuD. ZhangY. LiuQ. LiuZ. YangD. Zebrafish GSDMEb cleavage- gated pyroptosis drives septic acute kidney injury in vivo. J. Immunol.202020471929194210.4049/jimmunol.190145632111733
    [Google Scholar]
  24. DaiX.G. LiQ. LiT. HuangW.B. ZengZ.H. YangY. DuanZ.P. WangY.J. AiY.H. The interaction between C/EBPβ and TFAM promotes acute kidney injury via regulating NLRP3 inflammasome-mediated pyroptosis.Mol. Immunol.202012713614510.1016/j.molimm.2020.08.02332971400
    [Google Scholar]
  25. TamuraA. HiraiH. YokotaA. KamioN. SatoA. ShojiT. KashiwagiT. TorikoshiY. MiuraY. TenenD.G. MaekawaT. C/EBPβ is required for survival of Ly6C− monocytes.Blood2017130161809181810.1182/blood‑2017‑03‑77296228807982
    [Google Scholar]
  26. GuoJ. WangR. LiuD. Bone marrow-derived mesenchymal stem cells ameliorate sepsis-induced acute kidney injury by promoting mitophagy of renal tubular epithelial cells via the SIRT1/Parkin axis.Front. Endocrinol.20211263916510.3389/fendo.2021.63916534248837
    [Google Scholar]
  27. ChenB. NiY. LiuJ. ZhangY. YanF. Bone marrow-derived mesenchymal stem cells exert diverse effects on different macrophage subsets.Stem Cells Int.201820181910.1155/2018/834812130140291
    [Google Scholar]
  28. JuanC.X. MaoY. CaoQ. ChenY. ZhouL.B. LiS. ChenH. ChenJ.H. ZhouG.P. JinR. Exosome-mediated pyroptosis of miR-93-TXNIP-NLRP3 leads to functional difference between M1 and M2 macrophages in sepsis-induced acute kidney injury.J. Cell. Mol. Med.202125104786479910.1111/jcmm.1644933745232
    [Google Scholar]
  29. SunJ. GeX. WangY. NiuL. TangL. PanS. USF2 knockdown downregulates THBS1 to inhibit the TGF-β signaling pathway and reduce pyroptosis in sepsis-induced acute kidney injury.Pharmacol. Res.202217610596210.1016/j.phrs.2021.10596234756923
    [Google Scholar]
  30. WangQ.L. XingW. YuC. GaoM. DengL.T. ROCK1 regulates sepsis-induced acute kidney injury via TLR2-mediated endoplasmic reticulum stress/pyroptosis axis.Mol. Immunol.20211389910910.1016/j.molimm.2021.07.02234365196
    [Google Scholar]
  31. ChenB. HuangS. SuY. WuY.J. HannaA. BrickshawanaA. GraffJ. FrangogiannisN.G. Macrophage Smad3 protects the infarcted heart, stimulating phagocytosis and regulating inflammation.Circ. Res.20191251557010.1161/CIRCRESAHA.119.31506931092129
    [Google Scholar]
  32. ZhouM. YangL. ZhuoY. LiD. ZhangL. CuiL. LiJ. Effect of Liangxue Huoxue decoction on intestinal flora and NLRP3/caspase-1/GSDMD signaling pathway in mice model of sepsis-induced acute kidney injury.Zhonghua Wei Zhong Bing Ji Jiu Yi Xue202335325025536916336
    [Google Scholar]
  33. BattistoneM.A. MendelsohnA.C. SpallanzaniR.G. AllegrettiA.S. LibermanR.N. SesmaJ. KalimS. WallS.M. BonventreJ.V. LazarowskiE.R. BrownD. BretonS. Proinflammatory P2Y14 receptor inhibition protects against ischemic acute kidney injury in mice.J. Clin. Invest.202013073734374910.1172/JCI13479132287042
    [Google Scholar]
  34. ShigeokaA.A. MuellerJ.L. KamboA. MathisonJ.C. KingA.J. HallW.F. CorreiaJ.S. UlevitchR.J. HoffmanH.M. McKayD.B. An inflammasome-independent role for epithelial-expressed Nlrp3 in renal ischemia-reperfusion injury.J. Immunol.2010185106277628510.4049/jimmunol.100233020962258
    [Google Scholar]
  35. YangJ.R. YaoF.H. ZhangJ.G. JiZ.Y. LiK.L. ZhanJ. TongY.N. LinL.R. HeY.N. Ischemia-reperfusion induces renal tubule pyroptosis via the CHOP-caspase-11 pathway.Am. J. Physiol. Renal Physiol.20143061F75F8410.1152/ajprenal.00117.201324133119
    [Google Scholar]
  36. BaiT. CuiY. YangX. CuiX. YanC. TangY. CaoX. DongC. miR-302a-3p targets FMR1 to regulate pyroptosis of renal tubular epithelial cells induced by hypoxia–reoxygenation injury.Exp. Physiol.2021106122531254110.1113/EP08988734605097
    [Google Scholar]
  37. WangR. ZhaoH. ZhangY. ZhuH. SuQ. QiH. DengJ. XiaoC. Identification of MicroRNA-92a-3p as an essential regulator of tubular epithelial cell pyroptosis by targeting Nrf1 via HO-1.Front. Genet.20211161694710.3389/fgene.2020.61694733505436
    [Google Scholar]
  38. TajimaT. YoshifujiA. MatsuiA. ItohT. UchiyamaK. KandaT. TokuyamaH. WakinoS. ItohH. β-hydroxybutyrate attenuates renal ischemia-reperfusion injury through its anti-pyroptotic effects.Kidney Int.20199551120113710.1016/j.kint.2018.11.03430826015
    [Google Scholar]
  39. PangY. ZhangP. LuR. LiH. LiJ. FuH. CaoY.W. FangG. LiuB. WuJ. ZhouJ. ZhouY. Andrade-oliveira salvianolic acid B modulates caspase-1–mediated pyroptosis in renal ischemia-reperfusion injury via Nrf2 pathway.Front. Pharmacol.20201154142610.3389/fphar.2020.54142633013384
    [Google Scholar]
  40. MiaoN. YinF. XieH. WangY. XuY. ShenY. XuD. YinJ. WangB. ZhouZ. ChengQ. ChenP. XueH. ZhouL. LiuJ. WangX. ZhangW. LuL. The cleavage of gasdermin D by caspase-11 promotes tubular epithelial cell pyroptosis and urinary IL-18 excretion in acute kidney injury.Kidney Int.20199651105112010.1016/j.kint.2019.04.03531405732
    [Google Scholar]
  41. ZhangZ. ShaoX. JiangN. MouS. GuL. LiS. LinQ. HeY. ZhangM. ZhouW. NiZ. Caspase-11- mediated tubular epithelial pyroptosis underlies contrast-induced acute kidney injury.Cell Death Dis.201891098310.1038/s41419‑018‑1023‑x30250284
    [Google Scholar]
  42. Abu JawdehB.G. KansoA.A. SchellingJ.R. Evidence-based approach for prevention of radiocontrast-induced nephropathy.J. Hosp. Med.20094850050610.1002/jhm.47719824094
    [Google Scholar]
  43. MorcosR. KucharikM. BansalP. Al TaiiH. ManamR. CasaleJ. KhaliliH. MainiB. Contrast-induced acute kidney injury: Review and practical update.Clin. Med. Insights Cardiol.201913117954681987868010.1177/117954681987868031700251
    [Google Scholar]
  44. ChenF. LuJ. YangX. XiaoB. ChenH. PeiW. JinY. WangM. LiY. ZhangJ. LiuF. GuG. CuiW. Acetylbritannilactone attenuates contrast-induced acute kidney injury through its anti-pyroptosis effects.Biosci. Rep.2020402BSR2019325310.1042/BSR2019325331998952
    [Google Scholar]
  45. VilaysaneA. ChunJ. SeamoneM.E. WangW. ChinR. HirotaS. LiY. ClarkS.A. TschoppJ. TrpkovK. HemmelgarnB.R. BeckP.L. MuruveD.A. The NLRP3 inflammasome promotes renal inflammation and contributes to CKD.J. Am. Soc. Nephrol.201021101732174410.1681/ASN.201002014320688930
    [Google Scholar]
  46. MulayS.R. Multifactorial functions of the inflammasome component NLRP3 in pathogenesis of chronic kidney diseases.Kidney Int.2019961586610.1016/j.kint.2019.01.01430922667
    [Google Scholar]
  47. KongY. FengW. ZhaoX. ZhangP. LiS. LiZ. LinY. LiangB. LiC. WangW. HuangH. Statins ameliorate cholesterol-induced inflammation and improve AQP2 expression by inhibiting NLRP3 activation in the kidney.Theranostics20201023104151043310.7150/thno.4960332929357
    [Google Scholar]
  48. ZhuY. HuangG. YangY. YongC. YuX. WangG. YiL. GaoK. TianF. QianS. ZhouE. ZouY. Chinese herbal medicine suyin detoxification granule inhibits pyroptosis and epithelial-mesenchymal transition by downregulating MAVS/NLRP3 to alleviate renal injury.J. Inflamm. Res.2021146601661810.2147/JIR.S34159834908861
    [Google Scholar]
  49. GengW. TuC. ChenD. LuZ. MaoW. ZhuH. Huaier attenuates the adverse effects of pyroptosis by regulating the methylation of rat mesangial cells: An in vitro study.BMC Complem. Med. Therap.20222219210.1186/s12906‑022‑03559‑435351070
    [Google Scholar]
  50. PangQ. WangP. PanY. DongX. ZhouT. SongX. ZhangA. Irisin protects against vascular calcification by activating autophagy and inhibiting NLRP3-mediated vascular smooth muscle cell pyroptosis in chronic kidney disease.Cell Death Dis.202213328310.1038/s41419‑022‑04735‑735354793
    [Google Scholar]
  51. SigristM.K. TaalM.W. BungayP. McIntyreC.W. Progressive vascular calcification over 2 years is associated with arterial stiffening and increased mortality in patients with stages 4 and 5 chronic kidney disease.Clin. J. Am. Soc. Nephrol.2007261241124810.2215/CJN.0219050717928470
    [Google Scholar]
  52. MiaoN. XieH. XuD. YinJ. WangY. WangB. YinF. ZhouZ. ChengQ. ChenP. ZhouL. XueH. ZhangW. WangX. LiuJ. LuL. Caspase-11 promotes renal fibrosis by stimulating IL-1β maturation via activating caspase-1.Acta Pharmacol. Sin.201940679080010.1038/s41401‑018‑0177‑530382182
    [Google Scholar]
  53. ZhangH. WangZ. Effect and regulation of the NLRP3 inflammasome during renal fibrosis.Front. Cell Dev. Biol.2020737910.3389/fcell.2019.0037932039201
    [Google Scholar]
  54. GuoH. BiX. ZhouP. ZhuS. DingW. NLRP3 deficiency attenuates renal fibrosis and ameliorates mitochondrial dysfunction in a mouse unilateral ureteral obstruction model of chronic kidney disease.Mediators Inflamm.2017201711010.1155/2017/831656028348462
    [Google Scholar]
  55. AndersH.J. Suarez-AlvarezB. GrigorescuM. Foresto-NetoO. SteigerS. DesaiJ. MarschnerJ.A. HonarpishehM. ShiC. JordanJ. MüllerL. BurzlaffN. BäuerleT. MulayS.R. The macrophage phenotype and inflammasome component NLRP3 contributes to nephrocalcinosis-related chronic kidney disease independent from IL-1–mediated tissue injury.Kidney Int.201893365666910.1016/j.kint.2017.09.02229241624
    [Google Scholar]
  56. WuM. XiaW. JinQ. ZhouA. WangQ. LiS. HuangS. ZhangA. ZhangY. LiY. JiaZ. GasderminE. Gasdermin E deletion attenuates ureteral obstruction and 5/6 nephrectomy-induced renal fibrosis and kidney dysfunction.Front. Cell Dev. Biol.2021975413410.3389/fcell.2021.75413434746148
    [Google Scholar]
  57. TangY.S. ZhaoY.H. ZhongY. LiX.Z. PuJ.X. LuoY.C. ZhouQ.L. Neferine inhibits LPS-ATP-induced endothelial cell pyroptosis via regulation of ROS/NLRP3/Caspase-1 signaling pathway.Inflamm. Res.201968972773810.1007/s00011‑019‑01256‑631172209
    [Google Scholar]
  58. FlyvbjergA. The role of the complement system in diabetic nephropathy.Nat. Rev. Nephrol.201713531131810.1038/nrneph.2017.3128262777
    [Google Scholar]
  59. KeR. WangY. HongS. XiaoL. Endoplasmic reticulum stress related factor IRE1α regulates TXNIP/NLRP3- mediated pyroptosis in diabetic nephropathy.Exp. Cell Res.2020396211229310.1016/j.yexcr.2020.11229332950473
    [Google Scholar]
  60. ChengQ. PanJ. ZhouZ. YinF. XieH. ChenP. LiJ. ZhengP. ZhouL. ZhangW. LiuJ. LuL. Caspase-11/4 and gasdermin D-mediated pyroptosis contributes to podocyte injury in mouse diabetic nephropathy.Acta Pharmacol. Sin.202142695496310.1038/s41401‑020‑00525‑z32968210
    [Google Scholar]
  61. ShahzadK. BockF. DongW. WangH. KopfS. KohliS. Al-DabetM.M. RanjanS. WolterJ. WackerC. BiemannR. StoyanovS. ReymannK. SöderkvistP. GroßO. SchwengerV. PahernikS. NawrothP.P. GröneH.J. MadhusudhanT. IsermannB. Nlrp3-inflammasome activation in non-myeloid-derived cells aggravates diabetic nephropathy.Kidney Int.2015871748410.1038/ki.2014.27125075770
    [Google Scholar]
  62. ChenX. HeW. HuL. LiJ. FangY. WangX. XuX. WangZ. HuangK. HanJ. Pyroptosis is driven by non-selective gasdermin-D pore and its morphology is different from MLKL channel-mediated necroptosis.Cell Res.20162691007102010.1038/cr.2016.10027573174
    [Google Scholar]
  63. WangY. ZhuX. YuanS. WenS. LiuX. WangC. QuZ. LiJ. LiuH. SunL. LiuF. TLR4/NF-κB signaling induces GSDMD-related pyroptosis in tubular cells in diabetic kidney disease.Front. Endocrinol.20191060310.3389/fendo.2019.0060331608008
    [Google Scholar]
  64. LiuP. ZhangZ. LiY. Relevance of the pyroptosis-related inflammasome pathway in the pathogenesis of diabetic kidney disease.Front. Immunol.20211260341610.3389/fimmu.2021.60341633692782
    [Google Scholar]
  65. ZhanJ.F. HuangH.W. HuangC. HuL.L. XuW.W. Long Non-CodingR.N.A. Long Non-Coding RNA NEAT1 regulates pyroptosis in diabetic nephropathy via mediating the miR-34c/NLRP3 Axis.Kidney Blood Press. Res.202045458960210.1159/00050837232721950
    [Google Scholar]
  66. WangJ. ZhaoS.M. LncRNA-antisense non-coding RNA in the INK4 locus promotes pyroptosis via miR-497/thioredoxin-interacting protein axis in diabetic nephropathy.Life Sci.202126411872810.1016/j.lfs.2020.11872833160992
    [Google Scholar]
  67. DengJ. TanW. LuoQ. LinL. ZhengL. YangJ. Long non-coding RNA MEG3 promotes renal tubular epithelial cell pyroptosis by regulating the miR-18a-3p/GSDMD pathway in lipopolysaccharide-induced acute kidney injury.Front. Physiol.20211266321610.3389/fphys.2021.66321634012408
    [Google Scholar]
  68. DingX. JingN. ShenA. GuoF. SongY. PanM. MaX. ZhaoL. ZhangH. WuL. QinG. ZhaoY. MiR-21-5p in macrophage-derived extracellular vesicles affects podocyte pyroptosis in diabetic nephropathy by regulating A20.J. Endocrinol. Invest.20214461175118410.1007/s40618‑020‑01401‑732930981
    [Google Scholar]
  69. XieC. WuW. TangA. LuoN. TanY. lncRNA GAS5/miR-452-5p reduces oxidative stress and pyroptosis of high-glucose-stimulated renal tubular cells.Diabetes Metab. Syndr. Obes.2019122609261710.2147/DMSO.S22865431849505
    [Google Scholar]
  70. ZhuB. ChengX. JiangY. ChengM. ChenL. BaoJ. TangX. Silencing of KCNQ1OT1 decreases oxidative stress and pyroptosis of renal tubular epithelial cells.Diabetes Metab. Syndr. Obes.20201336537510.2147/DMSO.S22579132104033
    [Google Scholar]
  71. BaiY. MuQ. BaoX. ZuoJ. FangX. HuaJ. ZhangD. JiangG. LiP. GaoS. ZhaoD. Targeting NLRP3 inflammasome in the treatment of diabetes and diabetic complications: Role of natural compounds from herbal medicine.Aging Dis.20211271587160410.14336/AD.2021.031834631209
    [Google Scholar]
  72. WenS. LiS. LiL. FanQ. circACTR2: A novel mechanism regulating high glucose-induced fibrosis in renal tubular cells via pyroptosis.Biol. Pharm. Bull.202043355856410.1248/bpb.b19‑0090132115515
    [Google Scholar]
  73. RamC. JhaA.K. GhoshA. GairolaS. SyedA.M. MurtyU.S. NaiduV.G.M. SahuB.D. Targeting NLRP3 inflammasome as a promising approach for treatment of diabetic nephropathy: Preclinical evidences with therapeutic approaches.Eur. J. Pharmacol.202088517350310.1016/j.ejphar.2020.17350332858047
    [Google Scholar]
  74. GuJ. HuangW. ZhangW. ZhaoT. GaoC. GanW. RaoM. ChenQ. GuoM. XuY. XuY.H. Sodium butyrate alleviates high-glucose-induced renal glomerular endothelial cells damage via inhibiting pyroptosis.Int. Immunopharmacol.20197510583210.1016/j.intimp.2019.10583231473434
    [Google Scholar]
  75. WangB. DaiZ. GaoQ. LiuY. GuG. ZhengH. Spop ameliorates diabetic nephropathy through restraining NLRP3 inflammasome.Biochem. Biophys. Res. Commun.202259413113810.1016/j.bbrc.2021.12.06835081502
    [Google Scholar]
  76. DingT. WangS. ZhangX. ZaiW. FanJ. ChenW. BianQ. LuanJ. ShenY. ZhangY. JuD. MeiX. Kidney protection effects of dihydroquercetin on diabetic nephropathy through suppressing ROS and NLRP3 inflammasome.Phytomedicine201841455310.1016/j.phymed.2018.01.02629519318
    [Google Scholar]
  77. SamraY.A. SaidH.S. ElsherbinyN.M. LiouG.I. El-ShishtawyM.M. EissaL.A. Cepharanthine and piperine ameliorate diabetic nephropathy in rats: Role of NF-κB and NLRP3 inflammasome.Life Sci.201615718719910.1016/j.lfs.2016.06.00227266851
    [Google Scholar]
  78. SongW. WeiL. DuY. WangY. JiangS. Protective effect of ginsenoside metabolite compound K against diabetic nephropathy by inhibiting NLRP3 inflammasome activation and NF-κB/p38 signaling pathway in high-fat diet/streptozotocin-induced diabetic mice.Int. Immunopharmacol.20186322723810.1016/j.intimp.2018.07.02730107367
    [Google Scholar]
  79. ZhuY. ZhuC. YangH. DengJ. FanD. Protective effect of ginsenoside Rg5 against kidney injury via inhibition of NLRP3 inflammasome activation and the MAPK signaling pathway in high-fat diet/streptozotocin-induced diabetic mice.Pharmacol. Res.202015510474610.1016/j.phrs.2020.10474632156650
    [Google Scholar]
  80. KaS.M. LinJ.C. LinT.J. LiuF.C. ChaoL.K. HoC.L. YehL.T. SytwuH.K. HuaK.F. ChenA. Citral alleviates an accelerated and severe lupus nephritis model by inhibiting the activation signal of NLRP3 inflammasome and enhancing Nrf2 activation.Arthritis Res. Ther.201517133110.1186/s13075‑015‑0844‑626584539
    [Google Scholar]
  81. PengX. YangT. LiuG. LiuH. PengY. HeL. Piperine ameliorated lupus nephritis by targeting AMPK-mediated activation of NLRP3 inflammasome.Int. Immunopharmacol.20186544845710.1016/j.intimp.2018.10.02530388519
    [Google Scholar]
  82. HeJ. SunM. TianS. Procyanidin B2 prevents lupus nephritis development in mice by inhibiting NLRP3 inflammasome activation.Innate Immun.201824530731510.1177/175342591878098529874961
    [Google Scholar]
  83. ZhaoJ. WangJ. ZhouM. LiM. LiM. TanH. Curcumin attenuates murine lupus via inhibiting NLRP3 inflammasome.Int. Immunopharmacol.20196921321610.1016/j.intimp.2019.01.04630738291
    [Google Scholar]
  84. CheY. LiY. ZhengF. ZouK. LiZ. ChenM. HuS. TianC. YuW. GuoW. LuoM. DengW. ZouL. TRIP4 promotes tumor growth and metastasis and regulates radiosensitivity of cervical cancer by activating MAPK, PI3K/AKT, and hTERT signaling.Cancer Lett.201945211310.1016/j.canlet.2019.03.01730905820
    [Google Scholar]
  85. HuangJ. AnQ. JuB. ZhangJ. FanP. HeL. WangL. Role of vitamin D/VDR nuclear translocation in down-regulation of NF-κB/NLRP3/caspase-1 axis in lupus nephritis.Int. Immunopharmacol.202110010813110.1016/j.intimp.2021.10813134536747
    [Google Scholar]
  86. BonominiF. Dos SantosM. VeroneseF.V. RezzaniR. NLRP3 inflammasome modulation by melatonin supplementation in chronic pristane-induced lupus nephritis.Int. J. Mol. Sci.20192014346610.3390/ijms2014346631311094
    [Google Scholar]
  87. ZhangL. WangX.Z. LiY.S. ZhangL. HaoL.R. Icariin ameliorates IgA nephropathy by inhibition of nuclear factor kappa b/Nlrp3 pathway.FEBS Open Bio201771546310.1002/2211‑5463.1216128097088
    [Google Scholar]
  88. YangS.M. KaS.M. HuaK.F. WuT.H. ChuangY.P. LinY.W. YangF.L. WuS.H. YangS.S. LinS.H. ChangJ.M. ChenA. Antroquinonol mitigates an accelerated and progressive IgA nephropathy model in mice by activating the Nrf2 pathway and inhibiting T cells and NLRP3 inflammasome.Free Radic. Biol. Med.20136128529710.1016/j.freeradbiomed.2013.03.02423567192
    [Google Scholar]
  89. WuC.Y. HuaK.F. HsuW.H. SuzukiY. ChuL.J. LeeY.C. TakahataA. LeeS.L. WuC.C. Nikolic-PatersonD.J. KaS.M. ChenA. IgA nephropathy benefits from compound K treatment by inhibiting NF-κB/NLRP3 inflammasome and enhancing autophagy and SIRT1.J. Immunol.2020205120221210.4049/jimmunol.190028432482710
    [Google Scholar]
  90. HuaK.F. YangS.M. KaoT.Y. ChangJ.M. ChenH.L. TsaiY.J. ChenA. YangS.S. ChaoL.K. KaS.M. Osthole mitigates progressive IgA nephropathy by inhibiting reactive oxygen species generation and NF-κB/NLRP3 pathway.PLoS One2013810e7779410.1371/journal.pone.007779424204969
    [Google Scholar]
  91. LiH. LuR. PangY. LiJ. CaoY. FuH. FangG. ChenQ. LiuB. WuJ. ZhouY. ZhouJ. Zhen-Wu-Tang protects IgA nephropathy in rats by regulating exosomes to inhibit NF-κB/NLRP3 pathway.Front. Pharmacol.202011108010.3389/fphar.2020.0108032765277
    [Google Scholar]
  92. GoldwichA. BurkardM. ÖlkeM. DanielC. AmannK. HugoC. KurtsC. SteinkassererA. GessnerA. Podocytes are nonhematopoietic professional antigen-presenting cells.J. Am. Soc. Nephrol.201324690691610.1681/ASN.201202013323539760
    [Google Scholar]
  93. ZhangW. CaiY. XuW. YinZ. GaoX. XiongS. AIM2 facilitates the apoptotic DNA-induced systemic lupus erythematosus via arbitrating macrophage functional maturation.J. Clin. Immunol.201333592593710.1007/s10875‑013‑9881‑623479181
    [Google Scholar]
  94. GuoC. FuR. ZhouM. WangS. HuangY. HuH. ZhaoJ. GaskinF. YangN. FuS.M. Pathogenesis of lupus nephritis: RIP3 dependent necroptosis and NLRP3 inflammasome activation.J. Autoimmun.201910310228610.1016/j.jaut.2019.05.01431133359
    [Google Scholar]
  95. MistryP. KaplanM.J. Cell death in the pathogenesis of systemic lupus erythematosus and lupus nephritis.Clin. Immunol.2017185597310.1016/j.clim.2016.08.01027519955
    [Google Scholar]
  96. ZhangH. LiuL. LiL. Lentivirus-mediated knockdown of FcγRI (CD64) attenuated lupus nephritis via inhibition of NF-κB regulating NLRP3 inflammasome activation in MRL/lpr mice.J. Pharmacol. Sci.2018137434234910.1016/j.jphs.2018.05.01230190171
    [Google Scholar]
  97. ZhaoJ. WangH. DaiC. WangH. ZhangH. HuangY. WangS. GaskinF. YangN. Man FuS. P2X7 blockade attenuates murine lupus nephritis by inhibiting activation of the NLRP3/ASC/caspase 1 pathway.Arthritis Rheum.201365123176318510.1002/art.3817424022661
    [Google Scholar]
  98. MagistroniR. D’AgatiV.D. AppelG.B. KirylukK. New developments in the genetics, pathogenesis, and therapy of IgA nephropathy.Kidney Int.201588597498910.1038/ki.2015.25226376134
    [Google Scholar]
  99. ChunJ. ChungH. WangX. BarryR. TaheriZ.M. PlatnichJ.M. AhmedS.B. TrpkovK. HemmelgarnB. BenediktssonH. JamesM.T. MuruveD.A. NLRP3 localizes to the tubular epithelium in human kidney and correlates with outcome in IgA nephropathy.Sci. Rep.2016612466710.1038/srep2466727093923
    [Google Scholar]
  100. PétrilliV. DostertC. MuruveD.A. TschoppJ. The inflammasome: A danger sensing complex triggering innate immunity.Curr. Opin. Immunol.200719661562210.1016/j.coi.2007.09.00217977705
    [Google Scholar]
  101. MizushimaN. KomatsuM. Autophagy: renovation of cells and tissues.Cell2011147472874110.1016/j.cell.2011.10.02622078875
    [Google Scholar]
  102. SunQ. FanJ. BilliarT.R. ScottM.J. Inflammasome and autophagy regulation: A two-way street.Mol. Med.201723118819510.2119/molmed.2017.0007728741645
    [Google Scholar]
  103. ChangY.P. KaS.M. HsuW.H. ChenA. ChaoL.K. LinC.C. HsiehC.C. ChenM.C. ChiuH.W. HoC.L. ChiuY.C. LiuM.L. HuaK.F. Resveratrol inhibits NLRP3 inflammasome activation by preserving mitochondrial integrity and augmenting autophagy.J. Cell. Physiol.201523071567157910.1002/jcp.2490325535911
    [Google Scholar]
  104. TsaiY.L. HuaK.F. ChenA. WeiC.W. ChenW.S. WuC.Y. ChuC.L. YuY.L. LoC.W. KaS.M. NLRP3 inflammasome: Pathogenic role and potential therapeutic target for IgA nephropathy.Sci. Rep.2017714112310.1038/srep4112328117341
    [Google Scholar]
  105. PengW. PeiG. TangY. TanL. QinW. IgA1 deposition may induce NLRP3 expression and macrophage transdifferentiation of podocyte in IgA nephropathy.J. Transl. Med.201917140610.1186/s12967‑019‑02157‑231796125
    [Google Scholar]
/content/journals/cmc/10.2174/0109298673255656231003111621
Loading
/content/journals/cmc/10.2174/0109298673255656231003111621
Loading

Data & Media loading...

This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error
Please enter a valid_number test