117
Views
23
CrossRef citations to date
0
Altmetric
Original Research

Resveratrol Relieves Gouty Arthritis by Promoting Mitophagy to Inhibit Activation of NLRP3 Inflammasomes

, , , &
Pages 3523-3536 | Published online: 24 Jul 2021

References

  • Cleophas MCP, Crişan TO, Klück V, et al. Romidepsin suppresses monosodium urate crystal-induced cytokine production through upregulation of suppressor of cytokine signaling 1 expression. Arthritis Res Ther. 2019;21(1):50. doi:10.1186/s13075-019-1834-x
  • Desai J, Steiger S, Anders HJ. Molecular Pathophysiology of Gout. Trends Mol Med. 2017;23(8):756–768.
  • Dalbeth N, Choi HK, Terkeltaub R. Review gout: a roadmap to approaches for improving global outcomes. Arthritis Rheumatol. 2017;69(1):22–34. doi:10.1002/art.39799
  • Gupta MK, Singh JA. Cardiovascular disease in gout and the protective effect of treatments including urate-lowering therapy. Drugs. 2019;79(5):531–541. doi:10.1007/s40265-019-01081-5
  • Liu R, Han C, Wu D, et al. Prevalence of hyperuricemia and gout in Mainland China from 2000 to 2014: a systematic review and meta-analysis. Biomed Res Int. 2015;2015:762820. doi:10.1155/2015/762820
  • Lu X, Zeng R, Lin J, et al. Pharmacological basis for use of madecassoside in gouty arthritis: anti-inflammatory, anti-hyperuricemic, and NLRP3 inhibition. Immunopharmacol Immunotoxicol. 2019;41(2):277–284. doi:10.1080/08923973.2019.1590721
  • Huang HC, Chiang HP, Hsu NW, Huang CF, Chang SH, Lin KC. Differential risk group of developing stroke among older women with gouty arthritis: a latent transition analysis. Eur J Clin Invest. 2019;49(5):e13090. doi:10.1111/eci.13090.
  • Wilson L, Saseen JJ. Gouty arthritis: a review of acute management and prevention. Pharmacotherapy. 2016;36(8):906–922. doi:10.1002/phar.1788
  • So AK, Martinon F. Inflammation in gout: mechanisms and therapeutic targets. Nat Rev Rheumatol. 2017;13(11):639–647. doi:10.1038/nrrheum.2017.155
  • Elliott EI, Sutterwala FS. Initiation and perpetuation of NLRP3 inflammasome activation and assembly. Immunol Rev. 2015;265(1):35–52. doi:10.1111/imr.12286
  • Jo EK, Kim JK, Shin DM, Sasakawa C. Molecular mechanisms regulating NLRP3 inflammasome activation. Cell Mol Immunol. 2016;13(2):148–159. doi:10.1038/cmi.2015.95
  • Kingsbury SR, Conaghan PG, McDermott MF. The role of the NLRP3 inflammasome in gout. J Inflamm Res. 2011;4:39–49. doi:10.2147/JIR.S11330
  • Klück V, Jansen TLTA, Janssen M, et al. Dapansutrile, an oral selective NLRP3 inflammasome inhibitor, for treatment of gout flares: an open-label, dose-adaptive, proof-of-concept, phase 2a trial. Lancet Rheumatol. 2020;2(5):e270–e280. doi:10.1016/s2665-9913(20)30065-5
  • Chu J, Thomas LM, Watkins SC, Franchi L, Núñez G, Salter RD. Cholesterol-dependent cytolysins induce rapid release of mature IL-1beta from murine macrophages in a NLRP3 inflammasome and cathepsin B-dependent manner. J Leukoc Biol. 2009;86(5):1227–1238. doi:10.1189/jlb.0309164
  • He Y, Hara H, Núñez G. Mechanism and regulation of NLRP3 inflammasome activation. Trends Biochem Sci. 2016;41(12):1012–1021. doi:10.1016/j.tibs.2016.09.002
  • Zhou R, Tardivel A, Thorens B, Choi I, Tschopp J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat Immunol. 2010;11(2):136–140. doi:10.1038/ni.1831
  • Shimada K, Crother TR, Karlin J, et al. Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity. 2012;36(3):401–414. doi:10.1016/j.immuni.2012.01.009
  • Zhou R, Yazdi AS, Menu P, Tschopp J. A role for mitochondria in NLRP3 inflammasome activation. Nature. 2011;469(7329):221–225. doi:10.1038/nature09663
  • Kavas GO, Ayral PA, Elhan AH. The effects of resveratrol on oxidant/antioxidant systems and their cofactors in rats. Adv Clin Exp Med. 2013;22(2):151–155.
  • de la Lastra CA, Villegas I. Resveratrol as an anti-inflammatory and anti-aging agent: mechanisms and clinical implications. Mol Nutr Food Res. 2005;49(5):405–430. doi:10.1002/mnfr.200500022
  • Bertelli AA, Ferrara F, Diana G, et al. Resveratrol, a natural stilbene in grapes and wine, enhances intraphagocytosis in human promonocytes: a co-factor in antiinflammatory and anticancer chemopreventive activity. Int J Tissue React. 1999;21(4):93–104.
  • Rauf A, Imran M, Butt MS, Nadeem M, Peters DG, Mubarak MS. Resveratrol as an anti-cancer agent: a review. Crit Rev Food Sci Nutr. 2018;58(9):1428–1447. doi:10.1080/10408398.2016.1263597
  • Shi YW, Wang CP, Liu L, et al. Antihyperuricemic and nephroprotective effects of resveratrol and its analogues in hyperuricemic mice. Mol Nutr Food Res. 2012;56(9):1433–1444. doi:10.1002/mnfr.201100828
  • Chen H, Zheng S, Wang Y, et al. The effect of resveratrol on the recurrent attacks of gouty arthritis. Clin Rheumatol. 2014. doi:10.1007/s10067-014-2826-5
  • Yang QB, He YL, Zhong XW, Xie WG, Zhou JG. Resveratrol ameliorates gouty inflammation via upregulation of sirtuin 1 to promote autophagy in gout patients. Inflammopharmacology. 2019;27(1):47–56. doi:10.1007/s10787-018-00555-4
  • Chen X, Lu J, An M, Ma Z, Zong H, Yang J. Anti-inflammatory effect of resveratrol on adjuvant arthritis rats with abnormal immunological function via the reduction of cyclooxygenase-2 and prostaglandin E2. Mol Med Rep. 2014;9(6):2592–2598. doi:10.3892/mmr.2014.2070
  • El-Ghazaly MA, Fadel NA, Abdel-Naby DH, Abd El-Rehim HA, Zaki HF, Kenawy SA. Amelioration of adjuvant-induced arthritis by exposure to low dose gamma radiation and resveratrol administration in rats. Int J Radiat Biol. 2020;96(7):857–867. doi:10.1080/09553002.2020.1748911
  • Zhou Q, Lin FF, Liu SM, Sui XF. Influence of the total saponin fraction from Dioscorea nipponica Makino on TLR2/4-IL1R receptor singnal pathway in rats of gouty arthritis. J Ethnopharmacol. 2017;206:274–282. doi:10.1016/j.jep.2017.04.024
  • Coderre TJ, Wall PD. Ankle joint urate arthritis in rats provides a useful tool for the evaluation of analgesic and anti-arthritic agents. Pharmacol Biochem Behav. 1988;29(3):461–466. doi:10.1016/0091-3057(88)90004-4
  • Chung YH, Kim HY, Yoon BR, Kang YJ, Lee WW. Suppression of Syk activation by resveratrol inhibits MSU crystal-induced inflammation in human monocytes. J Mol Med (Berl). 2019;97(3):369–383. doi:10.1007/s00109-018-01736-y
  • Martin WJ, Walton M, Harper J. Resident macrophages initiating and driving inflammation in a monosodium urate monohydrate crystal-induced murine peritoneal model of acute gout. Arthritis Rheum. 2009;60(1):281–289. doi:10.1002/art.24185
  • Mian W, Zhang M, Ma Y, et al. Chaetocin attenuates gout in mice through inhibiting HIF-1α and NLRP3 inflammasome-dependent IL-1β secretion in macrophages. Arch Biochem Biophys. 2019;670:94–103. doi:10.1016/j.abb.2019.06.010
  • Zaiss MM, Maslowski KM. In vitro inflammasome assay. Bio-Protocol. 2014;4(11):e1142. doi:10.21769/BioProtoc.1142.
  • Pietrosimone KM, Jin M, Poston B, Liu P. Collagen-induced arthritis: a model for murine autoimmune arthritis. Bio Protoc. 2015;5(20):e1626. doi:10.21769/bioprotoc.1626
  • Dalbeth N, Merriman TR, Stamp LK. Gout. Lancet. 2016;388(10055):2039–2052. doi:10.1016/S0140-6736(16)00346-9
  • Martinon F, Pétrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440(7081):237–241. doi:10.1038/nature04516
  • Gicquel T, Robert S, Loyer P, et al. IL-1β production is dependent on the activation of purinergic receptors and NLRP3 pathway in human macrophages. FASEB J. 2015;29(10):4162–4173. doi:10.1096/fj.14-267393
  • Zheng SC, Zhu XX, Xue Y, et al. Role of the NLRP3 inflammasome in the transient release of IL-1β induced by monosodium urate crystals in human fibroblast-like synoviocytes. J Inflamm (Lond). 2015;12:30. doi:10.1186/s12950-015-0070-7
  • Robinson PC, Horsburgh S. Gout: joints and beyond, epidemiology, clinical features, treatment and co-morbidities. Maturitas. 2014;78(4):245–251. doi:10.1016/j.maturitas.2014.05.001
  • Inokuchi T, Moriwaki Y, Tsutsui H, et al. Plasma interleukin (IL)-18 (interferon-gamma-inducing factor) and other inflammatory cytokines in patients with gouty arthritis and monosodium urate monohydrate crystal-induced secretion of IL-18. Cytokine. 2006;33(1):21–27. doi:10.1016/j.cyto.2005.11.010
  • Swanson KV, Deng M, Ting JP. The NLRP3 inflammasome: molecular activation and regulation to therapeutics. Nat Rev Immunol. 2019;19(8):477–489. doi:10.1038/s41577-019-0165-0
  • Joosten LA, Netea MG, Mylona E, et al. Engagement of fatty acids with Toll-like receptor 2 drives interleukin-1β production via the ASC/caspase 1 pathway in monosodium urate monohydrate crystal-induced gouty arthritis. Arthritis Rheum. 2010;62(11):3237–3248. doi:10.1002/art.27667
  • Jiang L, Zhang L, Kang K, et al. Resveratrol ameliorates LPS-induced acute lung injury via NLRP3 inflammasome modulation. Biomed Pharmacother. 2016;84:130–138. doi:10.1016/j.biopha.2016.09.020
  • He Q, Li Z, Wang Y, Hou Y, Li L, Zhao J. Resveratrol alleviates cerebral ischemia/reperfusion injury in rats by inhibiting NLRP3 inflammasome activation through Sirt1-dependent autophagy induction. Int Immunopharmacol. 2017;50:208–215. doi:10.1016/j.intimp.2017.06.029
  • Tschopp J, Schroder K. NLRP3 inflammasome activation: the convergence of multiple signalling pathways on ROS production? Nat Rev Immunol. 2010;10(3):210–215. doi:10.1038/nri2725
  • Ashrafi G, Schwarz TL. The pathways of mitophagy for quality control and clearance of mitochondria. Cell Death Differ. 2013;20(1):31–42. doi:10.1038/cdd.2012.81
  • Chen G, Han Z, Feng D, et al. A regulatory signaling loop comprising the PGAM5 phosphatase and CK2 controls receptor-mediated mitophagy. Mol Cell. 2014;54(3):362–377. doi:10.1016/j.molcel.2014.02.034
  • Zhong Z, Umemura A, Sanchez-Lopez E, et al. NF-κB restricts inflammasome activation via elimination of damaged mitochondria. Cell. 2016;164(5):896–910. doi:10.1016/j.cell.2015.12.057
  • Saitoh T, Akira S. Regulation of inflammasomes by autophagy. J Allergy Clin Immunol. 2016;138(1):28–36. doi:10.1016/j.jaci.2016.05.009
  • Wu J, Li X, Zhu G, Zhang Y, He M, Zhang J. The role of Resveratrol-induced mitophagy/autophagy in peritoneal mesothelial cells inflammatory injury via NLRP3 inflammasome activation triggered by mitochondrial ROS. Exp Cell Res. 2016;341(1):42–53. doi:10.1016/j.yexcr.2016.01.014
  • He L, Zhou Q, Huang Z, et al. PINK1/Parkin-mediated mitophagy promotes apelin-13-induced vascular smooth muscle cell proliferation by AMPKα and exacerbates atherosclerotic lesions. J Cell Physiol. 2019;234(6):8668–8682. doi:10.1002/jcp.27527
  • Nguyen TN, Padman BS, Lazarou M. Deciphering the molecular signals of PINK1/Parkin mitophagy. Trends Cell Biol. 2016;26(10):733–744. doi:10.1016/j.tcb.2016.05.008
  • Lazarou M, Sliter DA, Kane LA, et al. The ubiquitin kinase PINK1 recruits autophagy receptors to induce mitophagy. Nature. 2015;524(7565):309–314. doi:10.1038/nature14893
  • Schlesinger N. Difficult-to-treat gouty arthritis: a disease warranting better management. Drugs. 2011;71(11):1413–1439. doi:10.2165/11592290-000000000-00000
  • Shin WY, Shim DW, Kim MK, et al. Protective effects of Cinnamomum cassia (Lamaceae) against gout and septic responses via attenuation of inflammasome activation in experimental models. J Ethnopharmacol. 2017;205:173–177. doi:10.1016/j.jep.2017.03.043
  • Huang D, Chen Y, Chen W, et al. Anti-inflammatory effects of the extract of Gnaphalium affine D. Don in vivo and in vitro. J Ethnopharmacol. 2015;176:356–364. doi:10.1016/j.jep.2015.11.010