2,216
Views
4
CrossRef citations to date
0
Altmetric
Research Paper

Protective effect of dexmedetomidine in cecal ligation perforation-induced acute lung injury through HMGB1/RAGE pathway regulation and pyroptosis activation

, , , , &
Pages 10608-10623 | Received 13 Jul 2021, Accepted 27 Oct 2021, Published online: 07 Dec 2021

References

  • Bersten AD, Edibam C, Hunt T, et al. Incidence and mortality of acute lung injury and the acute respiratory distress syndrome in three Australian States. Am J Respir Crit Care Med. 2002;165(4):443–448.
  • Mowery NT, Terzian WTH, Nelson AC. Acute lung injury. Curr Probl Surg. 2020;57(5):4.
  • Parcha V, Kalra R, Bhatt SP, et al. Trends and Geographic Variation In Acute Respiratory Failure and ARDS Mortality in the United States. Chest. 2021; 159(4):1460-1472.
  • Galam L, Failla A, Soundararajan R, et al. 4-hydroxynonenal regulates mitochondrial function in human small airway epithelial cells. Oncotarget. 2015;6(39):41508–41521.
  • Singh G, Gladdy G, Chandy TT, et al. Incidence and outcome of acute lung injury and acute respiratory distress syndrome in the surgical intensive care unit. Indian J Crit Care Med. 2014;18(10):659–665.
  • Mogensen TH. Pathogen recognition and inflammatory signaling in innate immune defenses. Clin Microbiol Rev. 2009;22:240–273.
  • Gong T, Liu L, Jiang W, et al. DAMP-sensing receptors in sterile inflammation and inflammatory diseases. Nat Rev Immunol. 2020;20(2):95–112.
  • Zheng D, Liwinski T, Elinav E. Inflammasome activation and regulation: toward a better understanding of complex mechanisms. Cell Discov. 2020;6:020–0167.
  • Kumar V. Pulmonary innate immune response determines the outcome of inflammation during pneumonia and sepsis-associated acute lung injury. front immunol. 2020;11:1722.
  • Park JS, Svetkauskaite D, He Q, et al. Involvement of toll-like receptors 2 and 4 in cellular activation by high mobility group box 1 protein. J Biol Chem. 2004;279(9):7370–7377.
  • Qu D, Ling Z, Tan X, et al. High mobility group protein B1 (HMGB1) interacts with receptor for advanced glycation end products (RAGE) to promote airway smooth muscle cell proliferation through ERK and NF-κB pathways. Int J Clin Exp Pathol. 2019;12:3268–3278.
  • Aglietti RA, Dueber EC. Recent insights into the molecular mechanisms underlying pyroptosis and gasdermin family functions. Trends Immunol. 2017;38(4):261–271.
  • Xu J, Jiang Y, Wang J, et al. Macrophage endocytosis of high-mobility group box 1 triggers pyroptosis. Cell Death Differ. 2014;21(8):1229–1239.
  • Chen L, Zhao Y, Lai D, et al. Neutrophil extracellular traps promote macrophage pyroptosis in sepsis. Cell Death Dis. 2018;9:018–0538.
  • Wang Y, Zhang H, Chen Q, et al. TNF-α/HMGB1 inflammation signalling pathway regulates pyroptosis during liver failure and acute kidney injury. Cell Prolif. 2020;53(6):17.
  • Sui X, Liu W, Liu Z. Exosomal lncRNA-p21 derived from mesenchymal stem cells protects epithelial cells during LPS-induced acute lung injury by sponging miR-181. Acta Biochim Biophys Sin (Shanghai). 2021;53(6):748–757.
  • Xiao K, He W, Guan W, et al. Mesenchymal stem cells reverse EMT process through blocking the activation of NF-kappaB and Hedgehog pathways in LPS-induced acute lung injury. Cell Death Dis. 2020;11(10):863.
  • Zhuang R, Yang X, Cai W, et al. MCTR3 reduces LPS-induced acute lung injury in mice via the ALX/PINK1 signaling pathway. Int Immunopharmacol. 2021;90:107142.
  • Hu H, Shi D, Hu C, et al. Dexmedetomidine mitigates CLP-stimulated acute lung injury via restraining the RAGE pathway. Am J Transl Res. 2017;9:5245–5258.
  • Li J, Chen Q, He X, et al. Dexmedetomidine attenuates lung apoptosis induced by renal ischemia-reperfusion injury through alpha2AR/PI3K/Akt pathway. J Transl Med. 2018;16(1):78.
  • Liang S, Wang Y, Liu Y. Dexmedetomidine alleviates lung ischemia-reperfusion injury in rats by activating PI3K/Akt pathway. Eur Rev Med Pharmacol Sci. 2019;23:370–377.
  • Liu J, Huang X, Hu S, et al. Dexmedetomidine attenuates lipopolysaccharide induced acute lung injury in rats by inhibition of caveolin-1 downstream signaling. Biomed Pharmacother. 2019;118:109314.
  • Zhang Y, Wang X, Liu Z, et al. Dexmedetomidine attenuates lipopolysaccharide induced acute lung injury by targeting NLRP3 via miR-381. J Biochem Mol Toxicol. 2018;32(11):e22211.
  • Kawasaki T, Kawasaki C, Ueki M, et al. Dexmedetomidine suppresses proinflammatory mediator production in human whole blood in vitro. J Trauma Acute Care Surg. 2013;74(5):1370–1375.
  • Tan F, Chen Y, Yuan D, et al. Dexmedetomidine protects against acute kidney injury through downregulating inflammatory reactions in endotoxemia rats. Biomed Rep. 2015;3(3):365–370.
  • Xu L, Bao H, Si Y, et al. Effects of dexmedetomidine on early and late cytokines during polymicrobial sepsis in mice. Inflamm Res. 2013;62(5):507–514.
  • Chang Y, Huang X, Liu Z, et al. Dexmedetomidine inhibits the secretion of high mobility group box 1 from lipopolysaccharide-activated macrophages in vitro. J Surg Res. 2013;181(2):308–314.
  • Li P, Gu L, Bian Q, et al. Long non-coding RNA MALAT1 enhances the protective effect of dexmedetomidine on acute lung injury by sponging miR-135a-5p to downregulate the ratio of X-box binding proteins XBP-1S/XBP-1U. Bioengineered. 2021;12(1):6377–6389.
  • Yang CH, Tsai PS, Wang TY, et al. Dexmedetomidine-ketamine combination mitigates acute lung injury in haemorrhagic shock rats. Resuscitation. 2009;80(10):1204–1210.
  • Liu J, Huang X, Hu S, et al. Dexmedetomidine attenuates lipopolysaccharide induced acute lung injury in rats by inhibition of caveolin-1 downstream signaling. Biomed Pharmacother. 2019;118:10.
  • Bhadade RR, de Souza RA, Harde MJ, et al. Clinical characteristics and outcomes of patients with acute lung injury and ARDS. J Postgrad Med. 2011;57(4):286–290.
  • Agouridakis P, Kyriakou D, Alexandrakis MG, et al. The predictive role of serum and bronchoalveolar lavage cytokines and adhesion molecules for acute respiratory distress syndrome development and outcome. Respir Res. 2002;3(1):23.
  • Meduri GU, Annane D, Chrousos GP, et al. Activation and regulation of systemic inflammation in ARDS: rationale for prolonged glucocorticoid therapy. Chest. 2009;136(6):1631–1643.
  • Jing W, Chunhua M, Shumin W. Effects of acteoside on lipopolysaccharide-induced inflammation in acute lung injury via regulation of NF-κB pathway in vivo and in vitro. Toxicol Appl Pharmacol. 2015;285(2):128–135.
  • Hirano Y, Aziz M, Yang WL, et al. Neutralization of osteopontin attenuates neutrophil migration in sepsis-induced acute lung injury. Crit Care. 2015;19(1):015–0782.
  • Duan Q, Jia Y, Qin Y, et al. Narciclasine attenuates LPS-induced acute lung injury in neonatal rats through suppressing inflammation and oxidative stress. Bioengineered. 2020;11(1):801–810.
  • Levy M, Thaiss CA, Elinav E. Taming the inflammasome. Nat Med. 2015;21(3):213–215.
  • Kelley N, Jeltema D, Duan Y, et al. The NLRP3 inflammasome: an overview of mechanisms of activation and regulation. Int J Mol Sci. 2019;20(13):3328.
  • He Y, Hara H, Núñez G. Mechanism and Regulation of NLRP3 Inflammasome Activation. Trends Biochem Sci. 2016;41(12):1012–1021.
  • Yang JW, Mao B, Tao RJ, et al. Corticosteroids alleviate lipopolysaccharide-induced inflammation and lung injury via inhibiting NLRP3-inflammasome activation. J Cell Mol Med. 2020;24(21):12716–12725.
  • Hosseinian N, Cho Y, Lockey RF, et al. The role of the NLRP3 inflammasome in pulmonary diseases. Ther Adv Respir Dis. 2015;9(4):188–197.
  • Grailer JJ, Canning BA, Kalbitz M, et al. Critical role for the NLRP3 inflammasome during acute lung injury. J Immunol. 2014;192(12):5974–5983.
  • Liu Z, Zhao H, Liu W, et al. NLRP3 inflammasome activation is essential for paraquat-induced acute lung injury. Inflammation. 2015;38(1):433–444.
  • Qu L, Chen C, Chen Y, et al. High-Mobility group box 1 (HMGB1) and autophagy in Acute Lung Injury (ALI): a review. Med Sci Monit. 2019;25:1828–1837.
  • Entezari M, Javdan M, Antoine DJ, et al. Inhibition of extracellular HMGB1 attenuates hyperoxia-induced inflammatory acute lung injury. Redox Biol. 2014;2:314–322.
  • Lee S, Piao C, Kim G, et al. Production and application of HMGB1 derived recombinant RAGE-antagonist peptide for anti-inflammatory therapy in acute lung injury. Eur J Pharm Sci. 2018;114:275–284.
  • Meng L, Li L, Lu S, et al. The protective effect of dexmedetomidine on LPS-induced acute lung injury through the HMGB1-mediated TLR4/NF-κB and PI3K/Akt/mTOR pathways. Mol Immunol. 2017;94:7–17.
  • Liu J, Huang X, Hu S, et al. Dexmedetomidine attenuates lipopolysaccharide induced acute lung injury in rats by inhibition of caveolin-1 downstream signaling. Biomed Pharmacother. 2019;118:109314.
  • Rouhiainen A, Kuja-Panula J, Tumova S, et al. RAGE-mediated cell signaling. Methods Mol Biol. 2013;963:239–263.
  • Sims GP, Rowe DC, Rietdijk ST, et al. HMGB1 and RAGE in inflammation and cancer. Annu Rev Immunol. 2010;28(1):367–388.
  • Bonaldi T, Talamo F, Scaffidi P, et al. Monocytic cells hyperacetylate chromatin protein HMGB1 to redirect it towards secretion. Embo J. 2003;22(20):5551–5560.
  • Deng M, Scott MJ, Fan J, et al. Location is the key to function: HMGB1 in sepsis and trauma-induced inflammation. J Leukoc Biol. 2019;106:161–169.
  • Schulman IG, Wang T, Wu M, et al. Macronuclei and micronuclei in Tetrahymena thermophila contain high-mobility-group-like chromosomal proteins containing a highly conserved eleven-amino-acid putative DNA-binding sequence. Mol Cell Biol. 1991;11:166–174.
  • Evankovich J, Cho SW, Zhang R, et al. High mobility group box 1 release from hepatocytes during ischemia and reperfusion injury is mediated by decreased histone deacetylase activity. J Biol Chem. 2010;285(51):39888–39897.
  • Lu B, Antoine DJ, Kwan K, et al. JAK/STAT1 signaling promotes HMGB1 hyperacetylation and nuclear translocation. Proc Natl Acad Sci U S A. 2014;111(8):3068–3073.
  • Dhupar R, Klune JR, Evankovich J, et al. Interferon regulatory factor 1 mediates acetylation and release of high mobility group box 1 from hepatocytes during murine liver ischemia-reperfusion injury. Shock. 2011;35(3):293–301.
  • Liu B, He R, Zhang L, et al. Inflammatory Caspases Drive Pyroptosis in Acute Lung Injury. Front Pharmacol. 2021;12:631256.
  • Man SM, Karki R, Kanneganti TD. Molecular mechanisms and functions of pyroptosis, inflammatory caspases and inflammasomes in infectious diseases. Immunol Rev. 2017;277(1):61–75.
  • Kayagaki N, Stowe IB, Lee BL, et al. Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature. 2015;526(7575):666–671.
  • Shi J, Gao W, Shao F. Pyroptosis: gasdermin-mediated programmed necrotic cell death. Trends Biochem Sci. 2017;42(4):245–254.
  • Deng M, Tang Y, Li W, et al. The endotoxin delivery protein HMGB1 mediates caspase-11-dependent lethality in sepsis. Immunity. 2018;49(740–53.e7):740–753.e7.
  • Ji X, Guo Y, Zhou G, et al. Dexmedetomidine protects against high mobility group box 1-induced cellular injury by inhibiting pyroptosis. Cell Biol Int. 2019;43(6):651–657.
  • Sun Y-B, Zhao H, Mu D-L, et al. Dexmedetomidine inhibits astrocyte pyroptosis and subsequently protects the brain in in vitro and in vivo models of sepsis. Cell Death Dis. 2019;10(3):167.