1,966
Views
19
CrossRef citations to date
0
Altmetric
Research Article

Beneficial anti-inflammatory effect of paeonol self-microemulsion-loaded colon-specific capsules on experimental ulcerative colitis rats

, , , , , & show all
Pages 324-335 | Received 02 Dec 2017, Accepted 28 Dec 2017, Published online: 09 Jan 2018

References

  • Yang Y, He J, Suo Y, et al. Tauroursodeoxycholate improves 2,4,6-trinitrobenzenesulfonic acid-induced experimental acute ulcerative colitis in mice. Int Immunopharmacol. 2016;36:271–276.
  • Li Q, Zhai W, Jiang Q, et al. Curcumin–piperine mixtures in self-microemulsifying drug delivery system for ulcerative colitis therapy. Int J Pharm. 2015;490:22–31.
  • Agis ER, Savas B, Melli M. Impact of colonic mucosal lipoxin A4 synthesis capacity on healing in rats with dextran sodium sulfate-induced colitis. Prostaglandins Other Lipid Mediat. 2015;121:63–69.
  • Trivedi PP, Jena GB. Ulcerative colitis-induced hepatic damage in mice: studies on inflammation, fibrosis, oxidative DNA damage and GST-P expression. Chem Biol Interact. 2013;201:19–30.
  • Wang YH, Ge B, Yang XL, et al. Proanthocyanidins from grape seeds modulates the nuclear factor-kappa B signal transduction pathways in rats with TNBS-induced recurrent ulcerative colitis. Int Immunopharmacol. 2011;11:1620–1627.
  • Samsami-Kor M, Daryani NE, Asl PR, et al. Anti-inflammatory effects of resveratrol in patients with ulcerative colitis: a randomized, double-blind, placebo-controlled pilot study. Arch Med Res. 2015;46:280–285.
  • Vong LB, Mo J, Abrahamsson B, et al. Specific accumulation of orally administered redox nanotherapeutics in the inflamed colon reducing inflammation with dose-response efficacy. J Control Release: Off J Control Release Soc. 2015;210:19–25.
  • Yun CS, Choi YG, Jeong MY, et al. Moutan cortex radicis inhibits inflammatory changes of gene expression in lipopolysaccharide-stimulated gingival fibroblasts. J Nat Med. 2013;67:576–589.
  • Jamal J, Mustafa MR, Wong PF. Paeonol protects against premature senescence in endothelial cells by modulating Sirtuin 1 pathway. J Ethnopharmacol. 2014;154:428–436.
  • Gong X, Yang Y, Huang L, et al. Antioxidation, anti-inflammation and anti-apoptosis by paeonol in LPS/d-GalN-induced acute liver failure in mice. Int Immunopharmacol. 2017;46:124–132.
  • Fu PK, Yang CY, Huang SC, et al. Evaluation of LPS-induced acute lung injury attenuation in rats by aminothiazole-paeonol derivatives. Molecules. 2017;22:1605.
  • Meng Y, Wang M, Xie X, et al. Paeonol ameliorates imiquimod-induced psoriasis-like skin lesions in BALB/c mice by inhibiting the maturation and activation of dendritic cells. Int J Mol Med. 2017;39:1101–1110.
  • Zhang L, Tao L, Shi T, et al. Paeonol inhibits B16F10 melanoma metastasis in vitro and in vivo via disrupting proinflammatory cytokines-mediated NF-κB and STAT3 pathways. IUBMB Life. 2015;67:778–788.
  • Li M, Tan SY, Wang XF. Paeonol exerts an anticancer effect on human colorectal cancer cells through inhibition of PGE2 synthesis and COX-2 expression. Oncol Rep. 2014;32:2845–2853.
  • Liu J, Wang S, Feng L, et al. Hypoglycemic and antioxidant activities of paeonol and its beneficial effect on diabetic encephalopathy in streptozotocin-induced diabetic rats. J Med Food. 2013;16:577–586.
  • Ye M, Yi Y, Wu S, et al. Role of paeonol in an astrocyte model of Parkinson’s disease. Med Sci Monit. 2017;23:4740–4748.
  • Shi X, Chen YH, Liu H, et al. Therapeutic effects of paeonol on methyl-4-phenyl-1,2,3,6-tetrahydropyridine/probenecid-induced Parkinson’s disease in mice. Mol Med Rep. 2016;14:2397–2404.
  • Lyu ZK, Li CL, Jin Y, et al. Paeonol exerts potential activities to inhibit the growth, migration and invasion of human gastric cancer BGC823 cells via downregulating MMP2 and MMP9. Mol Med Rep. 2017;16:7513–7519.
  • Zhou HM, Sun QX, Cheng Y. Paeonol enhances the sensitivity of human ovarian cancer cells to radiotherapy-induced apoptosis due to downregulation of the phosphatidylinositol-3-kinase/Akt/phosphatase and tensin homolog pathway and inhibition of vascular endothelial growth factor. Exp Ther Med. 2017;14:3213–3220.
  • Xu Y, Zhu JY, Lei ZM, et al. Anti-proliferative effects of paeonol on human prostate cancer cell lines DU145 and PC-3. J Physiol Biochem. 2017;73:157–165.
  • Zhai KF, Duan H, Luo L, et al. Protective effects of paeonol on inflammatory response in IL-1beta-induced human fibroblast-like synoviocytes and rheumatoid arthritis progression via modulating NF-kappaB pathway. Inflammopharmacology 2017;25:523–532.
  • Liu N, Feng X, Wang W, et al. Paeonol protects against TNF-alpha-induced proliferation and cytokine release of rheumatoid arthritis fibroblast-like synoviocytes by upregulating FOXO3 through inhibition of miR-155 expression. Inflamm Res. 2017;66:603–610.
  • Lou Y, Wang C, Tang Q, et al. Paeonol inhibits IL-1beta-induced inflammation via PI3K/Akt/NF-kappaB pathways: in vivo and vitro studies. Inflammation. 2017;40:1698–1706.
  • Choy KW, Lau YS, Murugan D, et al. Chronic treatment with paeonol improves endothelial function in mice through inhibition of endoplasmic reticulum stress-mediated oxidative stress. PLoS One. 2017;12:e0178365.
  • Ma L, Chuang CC, Weng W, et al. Paeonol protects rat heart by improving regional blood perfusion during no-reflow. Front Physiol. 2016;7:298.
  • Zong SY, Pu YQ, Xu BL, et al. Study on the physicochemical properties and anti-inflammatory effects of paeonol in rats with TNBS-induced ulcerative colitis. Int Immunopharmacol. 2017;42:32–38.
  • Liu MH, Lin AH, Ko HK, et al. Prevention of bleomycin-induced pulmonary inflammation and fibrosis in mice by paeonol. Front Physiol. 2017;8:193.
  • Xue P, Wang Y, Zeng F, et al. Paeonol suppresses solar ultraviolet-induced skin inflammation by targeting T-LAK cell-originated protein kinase. Oncotarget. 2017;8:27093–27104.
  • Wu J, Xu L, Sun C, et al. Paeonol alleviates epirubicin-induced renal injury in mice by regulating Nrf2 and NF-kappaB pathways. Eur J Pharmacol. 2017;795:84–93.
  • Fan HY, Qi D, Yu C, et al. Paeonol protects endotoxin-induced acute kidney injury: potential mechanism of inhibiting TLR4-NF-kappaB signal pathway. Oncotarget. 2016;7:39497–39510.
  • He LX, Tong X, Zeng J, et al. Paeonol suppresses neuroinflammatory responses in LPS-activated microglia cells. Inflammation. 2016;39:1904–1917.
  • Lin C, Lin HY, Chen JH, et al. Effects of paeonol on anti-neuroinflammatory responses in microglial cells. Int J Mol Sci. 2015;16:8844–8860.
  • Chang CY, Fu E, Chiang CY, et al. Effect of paeonol on tissue destruction in experimental periodontitis of rats. Am J Chin Med. 2014;42:361–374.
  • Ishiguro K, Ando T, Maeda O, et al. Paeonol attenuates TNBS-induced colitis by inhibiting NF-kappaB and STAT1 transactivation. Toxicol Appl Pharmacol. 2006;217:35–42.
  • Jin X, Wang J, Xia ZM, et al. Anti-inflammatory and anti-oxidative activities of paeonol and its metabolites through blocking MAPK/ERK/p38 signaling pathway. Inflammation. 2016;39:434–446.
  • Li M, Tan SY, Zhang J, et al. Effects of paeonol on intracellular calcium concentration and expression of RUNX3 in LoVo human colon cancer cells. Mol Med Rep. 2013;7:1425–1430.
  • Harada M, Yamashita A. [Pharmacological studies on the root bark of paeonia moutan. I. Central effects of paeonol]. Yakugaku Zasshi. 1969;89:1205–1211.
  • Luo JY, Zhong Y, Cao JC, et al. Efficacy of oral colon-specific delivery capsule of low-molecular-weight heparin on ulcerative colitis. Biomed Pharmacother. 2011;65:111–117.
  • Yehia SA, Elshafeey AH, Elsayed I. Pulsatile systems for colon targeting of budesonide: in vitro and in vivo evaluation. Drug Deliv. 2011;18:620–630.
  • Chen C, Jia F, Hou Z, et al. Delivery of paeonol by nanoparticles enhances its in vitro and in vivo antitumor effects. Int J Nanomed. 2017;12:6605–6616.
  • Chen ZX, Li B, Liu T, et al. Evaluation of paeonol-loaded transethosomes as transdermal delivery carriers. Eur J Pharm Sci. 2017;99:240–245.
  • Yao J, Zeng D, Zhang Y, et al. Effect of solvents on forming poly(butyl-2-cyanoacrylate) encapsulated paeonol nanocapsules. J Biomater Sci Polym Ed. 2017;28:240–256.
  • Yao J, Zhang Y, Hu Q, et al. Optimization of paeonol-loaded poly(butyl-2-cyanoacrylate) nanocapsules by central composite design with response surface methodology together with the antibacterial properties. Eur J Pharm Sci. 2017;101:189–199.
  • Li SS, Li GF, Liu L, et al. Optimization of paeonol-loaded microparticle formulation by response surface methodology. J Microencapsul. 2015;32:677–686.
  • Li SS, Li GF, Liu L, et al. Evaluation of paeonol skin-target delivery from its microsponge formulation: in vitro skin permeation and in vivo microdialysis. PLoS One. 2013;8:e79881.
  • Lin C, Miao Y, Qian M, et al. Enantioselective metabolism of flufiprole in rat and human liver microsomes. J Agric Food Chem. 2016;64:2371–2376.
  • Hu T, Zhou X, Wang L, et al. Effects of tanshinones from Salvia miltiorrhiza on CYP2C19 activity in human liver microsomes: enzyme kinetic and molecular docking studies. Chem Biol Interact. 2015;230:1–8.
  • Wang Y, Xie G, Liu Q, et al. Pharmacokinetics, tissue distribution, and plasma protein binding study of chicoric acid by HPLC-MS/MS. J Chromatogr B: Analyt Technol Biomed Life Sci. 2016;1031:139–145.
  • Waters NJ, Jones R, Williams G, et al. Validation of a rapid equilibrium dialysis approach for the measurement of plasma protein binding. J Pharm Sci. 2008;97:4586–4595.
  • Wang B, Pu Y, Xu B, et al. Self-microemulsifying drug delivery system improved oral bioavailability of 20(S)-protopanaxadiol: from preparation to evaluation. Chem Pharm Bull. 2015;63:688–693.
  • Laxmi M, Bhardwaj A, Mehta S, et al. Development and characterization of nanoemulsion as carrier for the enhancement of bioavailability of artemether. Artif Cells Nanomed Biotechnol. 2015;43:334–344.
  • Li F, Song S, Guo Y, et al. Preparation and pharmacokinetics evaluation of oral self-emulsifying system for poorly water-soluble drug Lornoxicam. Drug Deliv. 2015;22:487–498.
  • Xiao Y, Zhang YH, Sheng YX, et al. LC-MS determination and pharmacokinetic studies of paeonol in rat plasma after administration of different compatibility of Su-Xiao-Xin-Tong prescriptions. Biomed Chromatogr. 2008;22:527–534.
  • Xie Y, Zhou H, Wong YF, et al. Study on the pharmacokinetics and metabolism of paeonol in rats treated with pure paeonol and an herbal preparation containing paeonol by using HPLC-DAD-MS method. J Pharm Biomed Anal. 2008;46:748–756.
  • Bhandari S, Bhandari V, Sood J, et al. Improved pharmacokinetic and pharmacodynamic attributes of artemether-lumefantrine-loaded solid SMEDDS for oral administration. J Pharm Pharmacol. 2017;69:1437–1446.
  • Yeom DW, Son HY, Kim JH, et al. Development of a solidified self-microemulsifying drug delivery system (S-SMEDDS) for atorvastatin calcium with improved dissolution and bioavailability. Int J Pharm. 2016;506:302–311.
  • Baheti A, Srivastava S, Sahoo D, et al. Development and pharmacokinetic evaluation of industrially viable Self-microemulsifying Drug Delivery Systems (SMEDDS) for terbinafine. Curr Drug Deliv. 2016;13:65–75.
  • Mekjaruskul C, Yang YT, Leed MG, et al. Novel formulation strategies for enhancing oral delivery of methoxyflavones in Kaempferia parviflora by SMEDDS or complexation with 2-hydroxypropyl-β-cyclodextrin. Int J Pharm. 2013;445:1–11.
  • Constantinides PP. Lipid microemulsions for improving drug dissolution and oral absorption: physical and biopharmaceutical aspects. Pharm Res. 1995;12:1561–1572.
  • Zhang L, Zhu W, Yang C, et al. A novel folate-modified self-microemulsifying drug delivery system of curcumin for colon targeting. Int J Nanomed. 2012;7:151–162.
  • Zhou J, Tan L, Xie J, et al. Characterization of brusatol self-microemulsifying drug delivery system and its therapeutic effect against dextran sodium sulfate-induced ulcerative colitis in mice. Drug Deliv. 2017;24:1667–1679.
  • Li Y, Ma H, Wan Y, et al. Volatile organic compounds emissions from luculia pinceana flower and its changes at different stages of flower development. Molecules. 2016;21:531.
  • Xiumin LI, Man GE, Minzi LU, et al. The in vitro and in vivo evaluation of fenofibrate with a self- microemulsifying formulation. Curr Drug Deliv. 2015;12:308–313.
  • Duc Hanh N, Mitrevej A, Sathirakul K, et al. Development of phyllanthin-loaded self-microemulsifying drug delivery system for oral bioavailability enhancement. Drug Dev Ind Pharm. 2015;41:207–217.
  • Taha E, Ghorab D, Zaghloul AA. Bioavailability assessment of vitamin A self-nanoemulsified drug delivery systems in rats: a comparative study. Med Princ Pract. 2007;16:355–359.
  • Patel MM. Formulation and development of di-dependent microparticulate system for colon-specific drug delivery. Drug Deliv Transl Res. 2017;7:312–324.
  • Singh S, Kotla NG, Tomar S, et al. A nanomedicine-promising approach to provide an appropriate colon-targeted drug delivery system for 5-fluorouracil. Int J Nanomed. 2015;10:7175–7182.
  • Wachsmann P, Moulari B, Beduneau A, et al. Surfactant-dependence of nanoparticle treatment in murine experimental colitis. J Control Release. 2013;172:62–68.
  • Xu L, Shen P, Bi Y, et al. Danshen injection ameliorates STZ-induced diabetic nephropathy in association with suppression of oxidative stress, pro-inflammatory factors and fibrosis. Int Immunopharmacol. 2016;38:385–394.
  • Wang X, Yang X, Wen C, et al. Grass carp TGF-β1 impairs IL-1β signaling in the inflammatory responses: evidence for the potential of TGF-β1 to antagonize inflammation in fish. Dev Comp Immunol. 2016;59:121–127.
  • Zhang M, Zhou Q, Dorfman RG, et al. Butyrate inhibits interleukin-17 and generates tregs to ameliorate colorectal colitis in rats. BMC Gastroenterol. 2016;16:84.
  • Zhu XM, Huang YM, Fan JF, et al. Aberrant frequency of IL-10-producing B cells and its association with the balance of Treg/Th17 in children with inflammatory bowel disease. Pharmazie. 2015;70:656–660.
  • Eastaff-Leung N, Mabarrack N, Barbour A, et al. Foxp3+ regulatory T cells, Th17 effector cells, and cytokine environment in inflammatory bowel disease. J Clin Immunol. 2010;30:80–89.
  • Onishi RM, Gaffen SL. Interleukin-17 and its target genes: mechanisms of interleukin-17 function in disease. Immunology. 2010;129:311–321.
  • Park JS, Oh Y, Park O, et al. PEGylated TRAIL ameliorates experimental inflammatory arthritis by regulation of Th17 cells and regulatory T cells. J Control Release: *off J Control Release Soc. 2017;267:163–171.
  • Ma A, Yang Y, Wang Q, et al. Antiinflammatory effects of oxymatrine on rheumatoid arthritis in rats via regulating the imbalance between Treg and Th17 cells. Mol Med Rep. 2017;15:3615–3622.
  • Yuan X, Tong B, Dou Y, et al. Tetrandrine ameliorates collagen-induced arthritis in mice by restoring the balance between Th17 and Treg cells via the aryl hydrocarbon receptor. Biochem Pharmacol. 2016;101:87–99.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.