106
Views
8
CrossRef citations to date
0
Altmetric
Original Research

Dihydroartemisinin Inhibits TGF-β-Induced Fibrosis in Human Tenon Fibroblasts via Inducing Autophagy

, , &
Pages 973-981 | Published online: 03 Mar 2021

References

  • LiN, ZhangR. Silencing of p53 reduces cell migration in human tenon’s fibroblasts induced by TGF-beta. Int Ophthalmol. 2020;40(6):1509–1516. doi:10.1007/s10792-020-01320-932124130
  • MarchesiniA, De FrancescoF, Mattioli-BelmonteM, et al. A new animal model for pathological subcutaneous fibrosis: surgical technique and in vitro analysis. Front Cell Dev Biol. 2020;8:542. doi:10.3389/fcell.2020.0054232850775
  • WietechaMS, PensalfiniM, CangkramaM, et al. Activin-mediated alterations of the fibroblast transcriptome and matrisome control the biomechanical properties of skin wounds. Nat Commun. 2020;11(1):2604. doi:10.1038/s41467-020-16409-z32451392
  • ZahorecP, SarkozyovaN, FerancikovaN, et al. Autologous mesenchymal stem cells application in post-burn scars treatment: a preliminary study. Cell Tissue Bank. 2021;22(1):39–46. doi:10.1007/s10561-020-09862-z32862394
  • ChenX, HeLY, LaiS, HeY. Dihydroartemisinin inhibits the migration of esophageal cancer cells by inducing autophagy. Oncol Lett. 2020;20(4):94. doi:10.3892/ol.2020.1195532831913
  • YangB, GaoX, SunY, et al. Dihydroartemisinin alleviates high glucose-induced vascular smooth muscle cells proliferation and inflammation by depressing the miR-376b-3p/KLF15 pathway. Biochem Biophys Res Commun. 2020;530(3):574–580. doi:10.1016/j.bbrc.2020.07.09532753318
  • LeiZ, YangY, LiuS, et al. Dihydroartemisinin ameliorates dextran sulfate sodium induced inflammatory bowel diseases in mice. Bioorg Chem. 2020;100:103915. doi:10.1016/j.bioorg.2020.10391532450383
  • ZhangB, LiuP, ZhouY, et al. Dihydroartemisinin attenuates renal fibrosis through regulation of fibroblast proliferation and differentiation. Life Sci. 2019;223:29–37. doi:10.1016/j.lfs.2019.03.02030862567
  • YangDX, QiuJ, ZhouHH, et al. Dihydroartemisinin alleviates oxidative stress in bleomycin-induced pulmonary fibrosis. Life Sci. 2018;205:176–183. doi:10.1016/j.lfs.2018.05.02229752961
  • LiuQ, LuoL, RenC, et al. The opposing roles of the mTOR signaling pathway in different phases of human umbilical cord blood-derived CD34(+) cell erythropoiesis. Stem Cells. 2020. doi:10.1002/stem.3268
  • PantazopoulouM, BrembatiV, KanellidiA, et al. Distinct alpha-synuclein species induced by seeding are selectively cleared by the lysosome or the proteasome in neuronally differentiated SH-SY5Y cells. J Neurochem. 2020. doi:10.1111/jnc.15174
  • MiyakeT, SakaiN, TamaiA, et al. Trehalose ameliorates peritoneal fibrosis by promoting snail degradation and inhibiting mesothelial-to-mesenchymal transition in mesothelial cells. Sci Rep. 2020;10(1):14292. doi:10.1038/s41598-020-71230-432868830
  • XuY, SunQ, YuanF, et al. RND2 attenuates apoptosis and autophagy in glioblastoma cells by targeting the p38 MAPK signalling pathway. J Exp Clin Cancer Res. 2020;39(1):174. doi:10.1186/s13046-020-01671-232867814
  • ShiW, WuY, BianD. p75NTR silencing inhibits proliferation, migration and extracellular matrix deposition of hypertrophic scar fibroblasts by activating autophagy through inhibiting PI3K/Akt/mTOR pathway. Can J Physiol Pharmacol. 2020. doi:10.1139/cjpp-2020-0219
  • ShiJ, XiaoH, LiJ, et al. Wild-type p53-modulated autophagy and autophagic fibroblast apoptosis inhibit hypertrophic scar formation. Lab Invest. 2018;98(11):1423–1437. doi:10.1038/s41374-018-0099-330089855
  • FangS, HuC, XuL, et al. All-trans-retinoic acid inhibits the malignant behaviors of hepatocarcinoma cells by regulating autophagy. Am J Transl Res. 2020;12(10):6793–6810.33194073
  • LuYT, XiaoYF, LiYF, et al. Sulfuretin protects hepatic cells through regulation of ROS levels and autophagic flux. Acta Pharmacol Sin. 2019;40(7):908–918. doi:10.1038/s41401-018-0193-530560904
  • TrelfordCB, DenstedtJT, ArmstrongJJ, HutnikCML. The pro-fibrotic behavior of human tenon’s capsule fibroblasts in medically treated glaucoma patients. Clin Ophthalmol. 2020;14:1391–1402. doi:10.2147/OPTH.S24591532546947
  • TongJ, ChenF, DuW, ZhuJ, XieZ. TGF-beta1 induces human tenon’s fibroblasts fibrosis via miR-200b and its suppression of PTEN signaling. Curr Eye Res. 2019;44(4):360–367. doi:10.1080/02713683.2018.154926130512998
  • LinLT, ChenJT, LuDW, et al. Antifibrotic role of low-dose mitomycin-c-induced cellular senescence in trabeculectomy models. PLoS One. 2020;15(6):e0234706. doi:10.1371/journal.pone.023470632574191
  • FengL, SunZG, LiuQW, et al. Propofol inhibits the expression of Abelson nonreceptor tyrosine kinase without affecting learning or memory function in neonatal rats. Brain Behav. 2020;e01810.32869521
  • QinX, WuK, ZuoC, LinM. The expression and role of hypoxia-induced factor-1alpha in human tenon’s capsule fibroblasts under hypoxia. Curr Eye Res. 2020;1–9. doi:10.1080/02713683.2020.1805470
  • ShenW, WangY, WangD, et al. miR-145-5p attenuates hypertrophic scar via reducing Smad2/Smad3 expression. Biochem Biophys Res Commun. 2020;521(4):1042–1048. doi:10.1016/j.bbrc.2019.11.04031732152
  • QiJ, LiuY, HuK, et al. MicroRNA-205-5p regulates extracellular matrix production in hyperplastic scars by targeting Smad2. Exp Ther Med. 2019;17(3):2284–2290. doi:10.3892/etm.2019.718730867712
  • XiaoY. MiR-486-5p inhibits the hyperproliferation and production of collagen in hypertrophic scar fibroblasts via IGF1/PI3K/AKT pathway. J Dermatolog Treat. 2020;1–10. doi:10.1080/09546634.2020.1728210
  • ShiX, LiS, WangL, et al. RalB degradation by dihydroartemisinin induces autophagy and IFI16/caspase-1 inflammasome depression in the human laryngeal squamous cell carcinoma. Chin Med. 2020;15(1):64. doi:10.1186/s13020-020-00340-y32577124
  • TangM, WangR, FengP, et al. Dihydroartemisinin attenuates pulmonary hypertension via inhibition of pulmonary vascular remodeling in rats. J Cardiovasc Pharmacol. 2020;76(3):337–348. doi:10.1097/FJC.000000000000086232569012
  • DuX, WangL, LiQ, et al. miR-130a/TGF-beta1 axis is involved in sow fertility by controlling granulosa cell apoptosis. Theriogenology. 2020;157:407–417. doi:10.1016/j.theriogenology.2020.08.01532871445
  • QiC, LiuX, XiongT, WangD. Tempol prevents isoprenaline-induced takotsubo syndrome via the reactive oxygen species/mitochondrial/anti-apoptosis/p38 MAPK pathway. Eur J Pharmacol. 2020;886:173439. doi:10.1016/j.ejphar.2020.17343932871175
  • RenP, XingL, HongX, ChangL, ZhangH. LncRNA PITPNA-AS1 boosts the proliferation and migration of lung squamous cell carcinoma cells by recruiting TAF15 to stabilize HMGB3 mRNA. Cancer Med. 2020;9(20):7706–7716. doi:10.1002/cam4.326832871048
  • ShababS, GholamnezhadZ, MahmoudabadyM. Protective effects of medicinal plant against diabetes induced cardiac disorder: a review. J Ethnopharmacol. 2021;265:113328. doi:10.1016/j.jep.2020.11332832871233
  • LvW, JiangJ, LiY, et al. MiR-302a-3p aggravates myocardial ischemia-reperfusion injury by suppressing mitophagy via targeting FOXO3. Exp Mol Pathol. 2020;117:104522. doi:10.1016/j.yexmp.2020.10452232866521
  • YinF, WangWY, MaoLC, CaiQQ, JiangWH. Effect of human umbilical cord mesenchymal stem cells transfected with HGF on TGF-beta1/Smad signaling pathway in CCl4-induced liver fibrosis rats. Stem Cells Dev. 2020;29(21):1395–1406. doi:10.1089/scd.2020.006032867602
  • CheM, KweonSM, TeoJL, et al. Targeting the CBP/beta-catenin interaction to suppress activation of cancer-promoting pancreatic stellate cells. Cancers (Basel). 2020;12(6):1476. doi:10.3390/cancers12061476
  • LouH, LianC, ShiF, et al. The petri dish-N2B27 culture condition maintains rpe phenotype by inhibiting cell proliferation and mTOR activation. J Ophthalmol. 2020;2020:4892978. doi:10.1155/2020/489297832855817
  • CavalleroS, Neves GranitoR, StockholmD, et al. Exposure of human skin organoids to low genotoxic stress can promote epithelial-to-mesenchymal transition in regenerating keratinocyte precursor cells. Cells. 2020;9(8):1912. doi:10.3390/cells9081912
  • CaoC, WangW, LuL, et al. Inactivation of beclin-1-dependent autophagy promotes ursolic acid-induced apoptosis in hypertrophic scar fibroblasts. Exp Dermatol. 2018;27(1):58–63. doi:10.1111/exd.1341028767174
  • ShiJH, HuDH, ZhangZF, et al. Reduced expression of microtubule-associated protein 1 light chain 3 in hypertrophic scars. Arch Dermatol Res. 2012;304(3):209–215. doi:10.1007/s00403-012-1204-x22237724