메뉴 건너뛰기




Volumn 4, Issue JUN, 2017, Pages

Celastrol: A spectrum of treatment opportunities in chronic diseases

Author keywords

Cancer; Celastrol; Clinical trials; Inflammation; Neurodegenerative diseases; Toxicity; Treatment

Indexed keywords


EID: 85037138841     PISSN: None     EISSN: 2296858X     Source Type: Journal    
DOI: 10.3389/fmed.2017.00069     Document Type: Review
Times cited : (186)

References (220)
  • 1
    • 84914112012 scopus 로고    scopus 로고
    • Amelioration of mitochondrial dysfunction-induced insulin resistance in differentiated 3T3-L1 adipocytes via inhibition of NF-kappaB pathways
    • Bakar MH, Sarmidi MR, Kai CK, Huri HZ, Yaakob H. Amelioration of mitochondrial dysfunction-induced insulin resistance in differentiated 3T3-L1 adipocytes via inhibition of NF-kappaB pathways. Int J Mol Sci (2014) 15(12):22227-57. doi: 10.3390/ijms151222227
    • (2014) Int J Mol Sci , vol.15 , Issue.12 , pp. 22227-22257
    • Bakar, M.H.1    Sarmidi, M.R.2    Kai, C.K.3    Huri, H.Z.4    Yaakob, H.5
  • 2
    • 34347377577 scopus 로고    scopus 로고
    • Efficacy of T2 in active Crohn's disease: a prospective study report
    • Ren J, Tao Q, Wang X, Wang Z, Li J. Efficacy of T2 in active Crohn's disease: a prospective study report. Dig Dis Sci (2007) 52(8):1790-7. doi:10.1007/s10620-007-9747-y
    • (2007) Dig Dis Sci , vol.52 , Issue.8 , pp. 1790-1797
    • Ren, J.1    Tao, Q.2    Wang, X.3    Wang, Z.4    Li, J.5
  • 3
    • 77953145025 scopus 로고    scopus 로고
    • [Maintenance effect of polyglycosides of Tripterygium wilfordii on remission in postoperative Crohn disease]
    • Tao QS, Ren JA, Ji ZL, Li JS, Wang XB, Jiang XH. [Maintenance effect of polyglycosides of Tripterygium wilfordii on remission in postoperative Crohn disease]. Zhonghua Wei Chang Wai Ke Za Zhi (2009) 12(5):491-3
    • (2009) Zhonghua Wei Chang Wai Ke Za Zhi , vol.12 , Issue.5 , pp. 491-493
    • Tao, Q.S.1    Ren, J.A.2    Ji, Z.L.3    Li, J.S.4    Wang, X.B.5    Jiang, X.H.6
  • 4
    • 76549102074 scopus 로고    scopus 로고
    • [Efficacy of polyglycosides of Tripterygium wilfordii in preventing postoperative recurrence of Crohn disease]
    • Liao NS, Ren JA, Fan CG, Wang GF, Zhao YZ, Li JS. [Efficacy of polyglycosides of Tripterygium wilfordii in preventing postoperative recurrence of Crohn disease]. Zhonghua Wei Chang Wai Ke Za Zhi (2009) 12(2):167-9
    • (2009) Zhonghua Wei Chang Wai Ke Za Zhi , vol.12 , Issue.2 , pp. 167-169
    • Liao, N.S.1    Ren, J.A.2    Fan, C.G.3    Wang, G.F.4    Zhao, Y.Z.5    Li, J.S.6
  • 5
    • 84878416366 scopus 로고    scopus 로고
    • Prevention of postoperative recurrence of Crohn's disease: Tripterygium wilfordii polyglycoside versus mesalazine
    • Ren J, Wu X, Liao N, Wang G, Fan C, Liu S, et al. Prevention of postoperative recurrence of Crohn's disease: Tripterygium wilfordii polyglycoside versus mesalazine. J Int Med Res (2013) 41(1):176-87. doi:10.1177/0300060512474744
    • (2013) J Int Med Res , vol.41 , Issue.1 , pp. 176-187
    • Ren, J.1    Wu, X.2    Liao, N.3    Wang, G.4    Fan, C.5    Liu, S.6
  • 6
    • 84919625684 scopus 로고    scopus 로고
    • Tripterygium wilfordii Hook. F. versus azathioprine for prevention of postoperative recurrence in patients with Crohn's disease: a randomized clinical trial
    • Zhu W, Li Y, Gong J, Zuo L, Zhang W, Cao L, et al. Tripterygium wilfordii Hook. F. versus azathioprine for prevention of postoperative recurrence in patients with Crohn's disease: a randomized clinical trial. Dig Liver Dis (2015) 47(1):14-9. doi:10.1016/j.dld.2014.09.008
    • (2015) Dig Liver Dis , vol.47 , Issue.1 , pp. 14-19
    • Zhu, W.1    Li, Y.2    Gong, J.3    Zuo, L.4    Zhang, W.5    Cao, L.6
  • 7
    • 84946564827 scopus 로고    scopus 로고
    • Tripterygium wilfordii Hook F as maintenance treatment for Crohn's disease
    • Sun J, Shen X, Dong J, Wang H, Zuo L, Zhao J, et al. Tripterygium wilfordii Hook F as maintenance treatment for Crohn's disease. Am J Med Sci (2015) 350(5):345-51. doi:10.1097/MAJ.0000000000000591
    • (2015) Am J Med Sci , vol.350 , Issue.5 , pp. 345-351
    • Sun, J.1    Shen, X.2    Dong, J.3    Wang, H.4    Zuo, L.5    Zhao, J.6
  • 8
    • 84922477296 scopus 로고    scopus 로고
    • Efficacy and safety of Tripterygium wilfordii Hook F versus acitretin in moderate to severe psoriasis vulgaris: a randomized clinical trial
    • Wu C, Jin HZ, Shu D, Li F, He CX, Qiao J, et al. Efficacy and safety of Tripterygium wilfordii Hook F versus acitretin in moderate to severe psoriasis vulgaris: a randomized clinical trial. Chin Med J (Engl) (2015) 128(4):443-9. doi:10.4103/0366-6999.151069
    • (2015) Chin Med J (Engl) , vol.128 , Issue.4 , pp. 443-449
    • Wu, C.1    Jin, H.Z.2    Shu, D.3    Li, F.4    He, C.X.5    Qiao, J.6
  • 9
    • 0036065039 scopus 로고    scopus 로고
    • Benefit of an extract of Tripterygium wilfordii Hook F in patients with rheumatoid arthritis: a double-blind, placebo-controlled study
    • Tao X, Younger J, Fan FZ, Wang B, Lipsky PE. Benefit of an extract of Tripterygium wilfordii Hook F in patients with rheumatoid arthritis: a double-blind, placebo-controlled study. Arthritis Rheum (2002) 46(7):1735-43. doi:10.1002/art.10411
    • (2002) Arthritis Rheum , vol.46 , Issue.7 , pp. 1735-1743
    • Tao, X.1    Younger, J.2    Fan, F.Z.3    Wang, B.4    Lipsky, P.E.5
  • 10
    • 75649096444 scopus 로고    scopus 로고
    • [Tripterygium wilfordii extract for treating rheumatoid arthritis: systematic review]
    • Jiang Q, Cao W, Tang X, Jiao J. [Tripterygium wilfordii extract for treating rheumatoid arthritis: systematic review]. Zhongguo Zhong Yao Za Zhi (2009) 34(20):2637-43
    • (2009) Zhongguo Zhong Yao Za Zhi , vol.34 , Issue.20 , pp. 2637-2643
    • Jiang, Q.1    Cao, W.2    Tang, X.3    Jiao, J.4
  • 11
    • 69149101082 scopus 로고    scopus 로고
    • Comparison of Tripterygium wilfordii Hook F versus sulfasalazine in the treatment of rheumatoid arthritis: a randomized trial
    • Goldbach-Mansky R, Wilson M, Fleischmann R, Olsen N, Silverfield J, Kempf P, et al. Comparison of Tripterygium wilfordii Hook F versus sulfasalazine in the treatment of rheumatoid arthritis: a randomized trial. Ann Intern Med (2009) 151(4):229-40. doi:10.7326/0003-4819-151-4-200908180-00005W49-51
    • (2009) Ann Intern Med , vol.151 , Issue.4 , pp. 229-240
    • Goldbach-Mansky, R.1    Wilson, M.2    Fleischmann, R.3    Olsen, N.4    Silverfield, J.5    Kempf, P.6
  • 12
    • 84891611472 scopus 로고    scopus 로고
    • [External application of compound Tripterygium wilfordii decreased the activity of rheumatoid arthritis]
    • Jiao J, Jiang Q. [External application of compound Tripterygium wilfordii decreased the activity of rheumatoid arthritis]. Zhongguo Zhong Xi Yi Jie He Za Zhi (2012) 32(11):1470-2. 6
    • (2012) Zhongguo Zhong Xi Yi Jie He Za Zhi , vol.32 , Issue.11 , pp. 1426-1470
    • Jiao, J.1    Jiang, Q.2
  • 13
    • 0037334146 scopus 로고    scopus 로고
    • A randomized double blind, placebo controlled trial of topical Tripterygium wilfordii in rheumatoid arthritis: reanalysis using logistic regression analysis
    • Cibere J, Deng Z, Lin Y, Ou R, He Y, Wang Z, et al. A randomized double blind, placebo controlled trial of topical Tripterygium wilfordii in rheumatoid arthritis: reanalysis using logistic regression analysis. J Rheumatol (2003) 30(3):465-7
    • (2003) J Rheumatol , vol.30 , Issue.3 , pp. 465-467
    • Cibere, J.1    Deng, Z.2    Lin, Y.3    Ou, R.4    He, Y.5    Wang, Z.6
  • 14
    • 84893784248 scopus 로고    scopus 로고
    • Extracts of Tripterygium wilfordii Hook F in the treatment of rheumatoid arthritis: a systemic review and meta-analysis of randomised controlled trials
    • Liu Y, Tu S, Gao W, Wang Y, Liu P, Hu Y, et al. Extracts of Tripterygium wilfordii Hook F in the treatment of rheumatoid arthritis: a systemic review and meta-analysis of randomised controlled trials. Evid Based Complement Alternat Med (2013) 2013:410793. doi:10.1155/2013/410793
    • (2013) Evid Based Complement Alternat Med , vol.2013
    • Liu, Y.1    Tu, S.2    Gao, W.3    Wang, Y.4    Liu, P.5    Hu, Y.6
  • 15
    • 84931265721 scopus 로고    scopus 로고
    • Clinical trials of integrative medicine for rheumatoid arthritis: issues and recommendations
    • Zhang C, Jiang M, He XJ, Lu AP. Clinical trials of integrative medicine for rheumatoid arthritis: issues and recommendations. Chin J Integr Med (2015) 21(6):403-7. doi:10.1007/s11655-015-2041-5
    • (2015) Chin J Integr Med , vol.21 , Issue.6 , pp. 403-407
    • Zhang, C.1    Jiang, M.2    He, X.J.3    Lu, A.P.4
  • 16
    • 84934911857 scopus 로고    scopus 로고
    • Comparison of Tripterygium wilfordii Hook F with methotrexate in the treatment of active rheumatoid arthritis (TRIFRA): a randomised, controlled clinical trial
    • Lv QW, Zhang W, Shi Q, Zheng WJ, Li X, Chen H, et al. Comparison of Tripterygium wilfordii Hook F with methotrexate in the treatment of active rheumatoid arthritis (TRIFRA): a randomised, controlled clinical trial. Ann Rheum Dis (2015) 74(6):1078-86. doi:10.1136/annrheumdis-2013-204807
    • (2015) Ann Rheum Dis , vol.74 , Issue.6 , pp. 1078-1086
    • Lv, Q.W.1    Zhang, W.2    Shi, Q.3    Zheng, W.J.4    Li, X.5    Chen, H.6
  • 17
    • 0024400067 scopus 로고
    • Methotrexate in rheumatoid arthritis. Toxic effects as the major factor in limiting long-term treatment
    • Alarcon GS, Tracy IC, Blackburn WD Jr. Methotrexate in rheumatoid arthritis. Toxic effects as the major factor in limiting long-term treatment. Arthritis Rheum (1989) 32(6):671-6. doi:10.1002/anr.1780320603
    • (1989) Arthritis Rheum , vol.32 , Issue.6 , pp. 671-676
    • Alarcon, G.S.1    Tracy, I.C.2    Blackburn, W.D.3
  • 18
    • 0038814042 scopus 로고    scopus 로고
    • [Clinical observation on small doses Tripterygium wilfordii polyglycoside combined with methotrexate in treating rheumatoid arthritis]
    • Wu YJ, Lao ZY, Zhang ZL. [Clinical observation on small doses Tripterygium wilfordii polyglycoside combined with methotrexate in treating rheumatoid arthritis]. Zhongguo Zhong Xi Yi Jie He Za Zhi (2001) 21(12):895-6
    • (2001) Zhongguo Zhong Xi Yi Jie He Za Zhi , vol.21 , Issue.12 , pp. 895-896
    • Wu, Y.J.1    Lao, Z.Y.2    Zhang, Z.L.3
  • 19
    • 84903135857 scopus 로고    scopus 로고
    • [Etanercept combined with Tripterygium wilfordii polyglycoside for treatment of rheumatoid arthritis in the elderly: a clinical study]
    • He WZ, Yin ZH, Gao JH, Ye ZZ, Xie Y, Kong WH, et al. [Etanercept combined with Tripterygium wilfordii polyglycoside for treatment of rheumatoid arthritis in the elderly: a clinical study]. Zhongguo Zhong Xi Yi Jie He Za Zhi (2014) 34(3):267-71
    • (2014) Zhongguo Zhong Xi Yi Jie He Za Zhi , vol.34 , Issue.3 , pp. 267-271
    • He, W.Z.1    Yin, Z.H.2    Gao, J.H.3    Ye, Z.Z.4    Xie, Y.5    Kong, W.H.6
  • 20
    • 0036023433 scopus 로고    scopus 로고
    • Antiproliferative and proapoptotic activities of triptolide (PG490), a natural product entering clinical trials, on primary cultures of human prostatic epithelial cells
    • Kiviharju TM, Lecane PS, Sellers RG, Peehl DM. Antiproliferative and proapoptotic activities of triptolide (PG490), a natural product entering clinical trials, on primary cultures of human prostatic epithelial cells. Clin Cancer Res (2002) 8(8):2666-74
    • (2002) Clin Cancer Res , vol.8 , Issue.8 , pp. 2666-2674
    • Kiviharju, T.M.1    Lecane, P.S.2    Sellers, R.G.3    Peehl, D.M.4
  • 21
    • 67049110419 scopus 로고    scopus 로고
    • Phase I dose-escalation study of F60008, a novel apoptosis inducer, in patients with advanced solid tumours
    • Kitzen JJ, de Jonge MJ, Lamers CH, Eskens FA, van der Biessen D, van Doorn L, et al. Phase I dose-escalation study of F60008, a novel apoptosis inducer, in patients with advanced solid tumours. Eur J Cancer (2009) 45(10):1764-72. doi:10.1016/j.ejca.2009.01.026
    • (2009) Eur J Cancer , vol.45 , Issue.10 , pp. 1764-1772
    • Kitzen, J.J.1    de Jonge, M.J.2    Lamers, C.H.3    Eskens, F.A.4    van der Biessen, D.5    van Doorn, L.6
  • 22
    • 33746849034 scopus 로고    scopus 로고
    • Neuroprotective agents for clinical trials in ALS: a systematic assessment
    • Traynor BJ, Bruijn L, Conwit R, Beal F, O'Neill G, Fagan SC, et al. Neuroprotective agents for clinical trials in ALS: a systematic assessment. Neurology (2006) 67(1):20-7. doi:10.1212/01.wnl.0000223353.34006.54
    • (2006) Neurology , vol.67 , Issue.1 , pp. 20-27
    • Traynor, B.J.1    Bruijn, L.2    Conwit, R.3    Beal, F.4    O'Neill, G.5    Fagan, S.C.6
  • 23
    • 84922066899 scopus 로고    scopus 로고
    • [The effects and mechanism of Tripterygium wilfordii Hook F combination with irbesartan on urinary podocyte excretion in diabetic nephropathy patients]
    • Ma RX, Zhao N, Zhang W. [The effects and mechanism of Tripterygium wilfordii Hook F combination with irbesartan on urinary podocyte excretion in diabetic nephropathy patients]. Zhonghua Nei Ke Za Zhi (2013) 52(6):469-73
    • (2013) Zhonghua Nei Ke Za Zhi , vol.52 , Issue.6 , pp. 469-473
    • Ma, R.X.1    Zhao, N.2    Zhang, W.3
  • 24
    • 84878362911 scopus 로고    scopus 로고
    • Treatment of diabetic nephropathy with Tripterygium wilfordii Hook F extract: a prospective, randomized, controlled clinical trial
    • Ge Y, Xie H, Li S, Jin B, Hou J, Zhang H, et al. Treatment of diabetic nephropathy with Tripterygium wilfordii Hook F extract: a prospective, randomized, controlled clinical trial. J Transl Med (2013) 11:134. doi:10.1186/1479-5876-11-134
    • (2013) J Transl Med , vol.11 , pp. 134
    • Ge, Y.1    Xie, H.2    Li, S.3    Jin, B.4    Hou, J.5    Zhang, H.6
  • 25
    • 84883219051 scopus 로고    scopus 로고
    • Tripterygium preparations for the treatment of CKD: a systematic review and meta-analysis
    • Zhu B, Wang Y, Jardine M, Jun M, Lv JC, Cass A, et al. Tripterygium preparations for the treatment of CKD: a systematic review and meta-analysis. Am J Kidney Dis (2013) 62(3):515-30. doi:10.1053/j.ajkd.2013.02.374
    • (2013) Am J Kidney Dis , vol.62 , Issue.3 , pp. 515-530
    • Zhu, B.1    Wang, Y.2    Jardine, M.3    Jun, M.4    Lv, J.C.5    Cass, A.6
  • 26
    • 33745223816 scopus 로고    scopus 로고
    • Clinical trial of Tripterygium Wilfordii Hook F. in human kidney transplantation in China
    • Ji SM, Wang QW, Chen JS, Sha GZ, Liu ZH, Li LS. Clinical trial of Tripterygium Wilfordii Hook F. in human kidney transplantation in China. Transplant Proc (2006) 38(5):1274-9. doi:10.1016/j.transproceed.2006.03.017
    • (2006) Transplant Proc , vol.38 , Issue.5 , pp. 1274-1279
    • Ji, S.M.1    Wang, Q.W.2    Chen, J.S.3    Sha, G.Z.4    Liu, Z.H.5    Li, L.S.6
  • 27
    • 0023231271 scopus 로고
    • Tripterygium wilfordii, a Chinese herb effective in male fertility regulation
    • Qian SZ. Tripterygium wilfordii, a Chinese herb effective in male fertility regulation. Contraception (1987) 36(3):335-45. doi:10.1016/0010-7824(87)90104-1
    • (1987) Contraception , vol.36 , Issue.3 , pp. 335-345
    • Qian, S.Z.1
  • 28
    • 0027467355 scopus 로고
    • Male antifertility compounds from Tripterygium wilfordii Hook F
    • Matlin SA, Belenguer A, Stacey VE, Qian SZ, Xu Y, Zhang JW, et al. Male antifertility compounds from Tripterygium wilfordii Hook F. Contraception (1993) 47(4):387-400. doi:10.1016/0010-7824(93)90036-7
    • (1993) Contraception , vol.47 , Issue.4 , pp. 387-400
    • Matlin, S.A.1    Belenguer, A.2    Stacey, V.E.3    Qian, S.Z.4    Xu, Y.5    Zhang, J.W.6
  • 29
    • 0029188729 scopus 로고
    • [In vitro inhibition of celastrol on spermatozoa fertilization ability of guinea pig]
    • Yuan YY, Gu ZP, Shi QX, Qin GW, Xu RS, Cao L. [In vitro inhibition of celastrol on spermatozoa fertilization ability of guinea pig]. Yao Xue Xue Bao (1995) 30(5):331-5
    • (1995) Yao Xue Xue Bao , vol.30 , Issue.5 , pp. 331-335
    • Yuan, Y.Y.1    Gu, Z.P.2    Shi, Q.X.3    Qin, G.W.4    Xu, R.S.5    Cao, L.6
  • 30
    • 0342599563 scopus 로고
    • [Inhibitory effect of tripterine on activities of IL-1, IL-2 and release of PGE2]
    • Xu WM, Zhang LX, Cheng ZH, Cai WZ, Miao HH, Pan DJ. [Inhibitory effect of tripterine on activities of IL-1, IL-2 and release of PGE2]. Yao Xue Xue Bao (1991) 26(9):641-5
    • (1991) Yao Xue Xue Bao , vol.26 , Issue.9 , pp. 641-645
    • Xu, W.M.1    Zhang, L.X.2    Cheng, Z.H.3    Cai, W.Z.4    Miao, H.H.5    Pan, D.J.6
  • 32
    • 47749130766 scopus 로고    scopus 로고
    • Therapeutic effect of tripterine on adjuvant arthritis in rats
    • Li H, Zhang YY, Tan HW, Jia YF, Li D. Therapeutic effect of tripterine on adjuvant arthritis in rats. J Ethnopharmacol (2008) 118(3):479-84. doi:10.1016/j.jep.2008.05.028
    • (2008) J Ethnopharmacol , vol.118 , Issue.3 , pp. 479-484
    • Li, H.1    Zhang, Y.Y.2    Tan, H.W.3    Jia, Y.F.4    Li, D.5
  • 33
    • 68349129966 scopus 로고    scopus 로고
    • Suppression of inflammatory responses by celastrol, a quinone methide triterpenoid isolated from Celastrus regelii
    • Kim DH, Shin EK, Kim YH, Lee BW, Jun JG, Park JH, et al. Suppression of inflammatory responses by celastrol, a quinone methide triterpenoid isolated from Celastrus regelii. Eur J Clin Invest (2009) 39(9):819-27. doi:10.1111/j.1365-2362.2009.02186.x
    • (2009) Eur J Clin Invest , vol.39 , Issue.9 , pp. 819-827
    • Kim, D.H.1    Shin, E.K.2    Kim, Y.H.3    Lee, B.W.4    Jun, J.G.5    Park, J.H.6
  • 34
    • 33750008443 scopus 로고    scopus 로고
    • Inhibition of NF-kappa B activation through targeting I kappa B kinase by celastrol, a quinone methide triterpenoid
    • Lee JH, Koo TH, Yoon H, Jung HS, Jin HZ, Lee K, et al. Inhibition of NF-kappa B activation through targeting I kappa B kinase by celastrol, a quinone methide triterpenoid. Biochem Pharmacol (2006) 72(10):1311-21. doi:10.1016/j.bcp.2006.08.014
    • (2006) Biochem Pharmacol , vol.72 , Issue.10 , pp. 1311-1321
    • Lee, J.H.1    Koo, T.H.2    Yoon, H.3    Jung, H.S.4    Jin, H.Z.5    Lee, K.6
  • 35
    • 79955424712 scopus 로고    scopus 로고
    • Celastrus-derived celastrol suppresses autoimmune arthritis by modulating antigen-induced cellular and humoral effector responses
    • Venkatesha SH, Yu H, Rajaiah R, Tong L, Moudgil KD. Celastrus-derived celastrol suppresses autoimmune arthritis by modulating antigen-induced cellular and humoral effector responses. J Biol Chem (2011) 286(17):15138-46. doi:10.1074/jbc.M111.226365
    • (2011) J Biol Chem , vol.286 , Issue.17 , pp. 15138-15146
    • Venkatesha, S.H.1    Yu, H.2    Rajaiah, R.3    Tong, L.4    Moudgil, K.D.5
  • 36
    • 84865042616 scopus 로고    scopus 로고
    • Suppression of autoimmune arthritis by Celastrus-derived celastrol through modulation of pro-inflammatory chemokines
    • Venkatesha SH, Astry B, Nanjundaiah SM, Yu H, Moudgil KD. Suppression of autoimmune arthritis by Celastrus-derived celastrol through modulation of pro-inflammatory chemokines. Bioorg Med Chem (2012) 20(17):5229-34. doi:10.1016/j.bmc.2012.06.050
    • (2012) Bioorg Med Chem , vol.20 , Issue.17 , pp. 5229-5234
    • Venkatesha, S.H.1    Astry, B.2    Nanjundaiah, S.M.3    Yu, H.4    Moudgil, K.D.5
  • 38
    • 0346220027 scopus 로고    scopus 로고
    • Chemokine receptor expression and in vivo signaling pathways in the joints of rats with adjuvant-induced arthritis
    • Shahrara S, Amin MA, Woods JM, Haines GK, Koch AE. Chemokine receptor expression and in vivo signaling pathways in the joints of rats with adjuvant-induced arthritis. Arthritis Rheum (2003) 48(12):3568-83. doi:10.1002/art.11344
    • (2003) Arthritis Rheum , vol.48 , Issue.12 , pp. 3568-3583
    • Shahrara, S.1    Amin, M.A.2    Woods, J.M.3    Haines, G.K.4    Koch, A.E.5
  • 39
    • 84866558326 scopus 로고    scopus 로고
    • Effective treatment of rat adjuvant-induced arthritis by celastrol
    • Cascao R, Vidal B, Raquel H, Neves-Costa A, Figueiredo N, Gupta V, et al. Effective treatment of rat adjuvant-induced arthritis by celastrol. Autoimmun Rev (2012) 11(12):856-62. doi:10.1016/j.autrev.2012.02.022
    • (2012) Autoimmun Rev , vol.11 , Issue.12 , pp. 856-862
    • Cascao, R.1    Vidal, B.2    Raquel, H.3    Neves-Costa, A.4    Figueiredo, N.5    Gupta, V.6
  • 40
    • 84957109780 scopus 로고    scopus 로고
    • Decrease of CD68 synovial macrophages in celastrol treated arthritic rats
    • Cascao R, Vidal B, Lopes IP, Paisana E, Rino J, Moita LF, et al. Decrease of CD68 synovial macrophages in celastrol treated arthritic rats. PLoS One (2015) 10(12):e0142448. doi:10.1371/journal.pone.0142448
    • (2015) PLoS One , vol.10 , Issue.12
    • Cascao, R.1    Vidal, B.2    Lopes, I.P.3    Paisana, E.4    Rino, J.5    Moita, L.F.6
  • 41
    • 84936940989 scopus 로고    scopus 로고
    • Celastrol blocks binding of lipopolysaccharides to a toll-like receptor4/myeloid differentiation factor2 complex in a thiol-dependent manner
    • Lee JY, Lee BH, Kim ND, Lee JY. Celastrol blocks binding of lipopolysaccharides to a toll-like receptor4/myeloid differentiation factor2 complex in a thiol-dependent manner. J Ethnopharmacol (2015) 172:254-60. doi:10.1016/j.jep.2015.06.028
    • (2015) J Ethnopharmacol , vol.172 , pp. 254-260
    • Lee, J.Y.1    Lee, B.H.2    Kim, N.D.3    Lee, J.Y.4
  • 42
    • 84879801389 scopus 로고    scopus 로고
    • Celastrol inhibits lipopolysaccharide-stimulated rheumatoid fibroblast-like synoviocyte invasion through suppression of TLR4/NF-kappaB-mediated matrix metalloproteinase-9 expression
    • Li G, Liu D, Zhang Y, Qian Y, Zhang H, Guo S, et al. Celastrol inhibits lipopolysaccharide-stimulated rheumatoid fibroblast-like synoviocyte invasion through suppression of TLR4/NF-kappaB-mediated matrix metalloproteinase-9 expression. PLoS One (2013) 8(7):e68905. doi:10.1371/journal.pone.0068905
    • (2013) PLoS One , vol.8 , Issue.7
    • Li, G.1    Liu, D.2    Zhang, Y.3    Qian, Y.4    Zhang, H.5    Guo, S.6
  • 43
    • 84866975989 scopus 로고    scopus 로고
    • Celastrol inhibits interleukin-17A-stimulated rheumatoid fibroblast-like synoviocyte migration and invasion through suppression of NF-kappaB-mediated matrix metalloproteinase-9 expression
    • Li GQ, Zhang Y, Liu D, Qian YY, Zhang H, Guo SY, et al. Celastrol inhibits interleukin-17A-stimulated rheumatoid fibroblast-like synoviocyte migration and invasion through suppression of NF-kappaB-mediated matrix metalloproteinase-9 expression. Int Immunopharmacol (2012) 14(4):422-31. doi:10.1016/j.intimp.2012.08.016
    • (2012) Int Immunopharmacol , vol.14 , Issue.4 , pp. 422-431
    • Li, G.Q.1    Zhang, Y.2    Liu, D.3    Qian, Y.Y.4    Zhang, H.5    Guo, S.Y.6
  • 44
    • 84887086286 scopus 로고    scopus 로고
    • Anti-invasive effects of celastrol in hypoxia-induced fibroblast-like synoviocyte through suppressing of HIF-1alpha/CXCR4 signaling pathway
    • Li GQ, Liu D, Zhang Y, Qian YY, Zhu YD, Guo SY, et al. Anti-invasive effects of celastrol in hypoxia-induced fibroblast-like synoviocyte through suppressing of HIF-1alpha/CXCR4 signaling pathway. Int Immunopharmacol (2013) 17(4):1028-36. doi:10.1016/j.intimp.2013.10.006
    • (2013) Int Immunopharmacol , vol.17 , Issue.4 , pp. 1028-1036
    • Li, G.Q.1    Liu, D.2    Zhang, Y.3    Qian, Y.Y.4    Zhu, Y.D.5    Guo, S.Y.6
  • 45
    • 84930986439 scopus 로고    scopus 로고
    • Celastrol inhibits inflammatory stimuli-induced neutrophil extracellular trap formation
    • Yu Y, Koehn CD, Yue Y, Li S, Thiele GM, Hearth-Holmes MP, et al. Celastrol inhibits inflammatory stimuli-induced neutrophil extracellular trap formation. Curr Mol Med (2015) 15(4):401-10. doi:10.2174/1566524015666150505160743
    • (2015) Curr Mol Med , vol.15 , Issue.4 , pp. 401-410
    • Yu, Y.1    Koehn, C.D.2    Yue, Y.3    Li, S.4    Thiele, G.M.5    Hearth-Holmes, M.P.6
  • 46
    • 84924544171 scopus 로고    scopus 로고
    • Celastrol, a Chinese herbal compound, controls autoimmune inflammation by altering the balance of pathogenic and regulatory T cells in the target organ
    • Astry B, Venkatesha SH, Laurence A, Christensen-Quick A, Garzino-Demo A, Frieman MB, et al. Celastrol, a Chinese herbal compound, controls autoimmune inflammation by altering the balance of pathogenic and regulatory T cells in the target organ. Clin Immunol (2015) 157(2):228-38. doi:10.1016/j.clim.2015.01.011
    • (2015) Clin Immunol , vol.157 , Issue.2 , pp. 228-238
    • Astry, B.1    Venkatesha, S.H.2    Laurence, A.3    Christensen-Quick, A.4    Garzino-Demo, A.5    Frieman, M.B.6
  • 47
    • 78649853549 scopus 로고    scopus 로고
    • Small molecule inhibitors of IkappaB kinase signaling inhibit osteoclast formation in vitro and prevent ovariectomy-induced bone loss in vivo
    • Idris AI, Krishnan M, Simic P, Landao-Bassonga E, Mollat P, Vukicevic S, et al. Small molecule inhibitors of IkappaB kinase signaling inhibit osteoclast formation in vitro and prevent ovariectomy-induced bone loss in vivo. FASEB J (2010) 24(11):4545-55. doi:10.1096/fj.10-164095
    • (2010) FASEB J , vol.24 , Issue.11 , pp. 4545-4555
    • Idris, A.I.1    Krishnan, M.2    Simic, P.3    Landao-Bassonga, E.4    Mollat, P.5    Vukicevic, S.6
  • 48
    • 84862676391 scopus 로고    scopus 로고
    • Celastrus and its bioactive celastrol protect against bone damage in autoimmune arthritis by modulating osteoimmune cross-talk
    • Nanjundaiah SM, Venkatesha SH, Yu H, Tong L, Stains JP, Moudgil KD. Celastrus and its bioactive celastrol protect against bone damage in autoimmune arthritis by modulating osteoimmune cross-talk. J Biol Chem (2012) 287(26):22216-26. doi:10.1074/jbc.M112.356816
    • (2012) J Biol Chem , vol.287 , Issue.26 , pp. 22216-22226
    • Nanjundaiah, S.M.1    Venkatesha, S.H.2    Yu, H.3    Tong, L.4    Stains, J.P.5    Moudgil, K.D.6
  • 49
    • 84920380130 scopus 로고    scopus 로고
    • Celastrol attenuates bone erosion in collagen-Induced arthritis mice and inhibits osteoclast differentiation and function in RANKL-induced RAW264.7
    • Gan K, Xu L, Feng X, Zhang Q, Wang F, Zhang M, et al. Celastrol attenuates bone erosion in collagen-Induced arthritis mice and inhibits osteoclast differentiation and function in RANKL-induced RAW264.7. Int Immunopharmacol (2015) 24(2):239-46. doi:10.1016/j.intimp.2014.12.012
    • (2015) Int Immunopharmacol , vol.24 , Issue.2 , pp. 239-246
    • Gan, K.1    Xu, L.2    Feng, X.3    Zhang, Q.4    Wang, F.5    Zhang, M.6
  • 50
    • 84960192063 scopus 로고    scopus 로고
    • Celastrol, an NF-kappaB inhibitor, ameliorates hypercalciuria and articular cartilage lesions in a mouse model of secondary osteoporosis
    • Liu X, Cai F, Zhang Y, Yang A, Liu L. Celastrol, an NF-kappaB inhibitor, ameliorates hypercalciuria and articular cartilage lesions in a mouse model of secondary osteoporosis. J Pharmacol Sci (2016) 130(4):204-11. doi:10.1016/j.jphs.2016.02.001
    • (2016) J Pharmacol Sci , vol.130 , Issue.4 , pp. 204-211
    • Liu, X.1    Cai, F.2    Zhang, Y.3    Yang, A.4    Liu, L.5
  • 51
    • 34247164611 scopus 로고    scopus 로고
    • Ankylosing spondylitis
    • Braun J, Sieper J. Ankylosing spondylitis. Lancet (2007) 369(9570):1379-90. doi:10.1016/S0140-6736(07)60635-7
    • (2007) Lancet , vol.369 , Issue.9570 , pp. 1379-1390
    • Braun, J.1    Sieper, J.2
  • 52
    • 40549109246 scopus 로고    scopus 로고
    • Critical appraisal of assessment of structural damage in ankylosing spondylitis: implications for treatment outcomes
    • Sieper J, Appel H, Braun J, Rudwaleit M. Critical appraisal of assessment of structural damage in ankylosing spondylitis: implications for treatment outcomes. Arthritis Rheum (2008) 58(3):649-56. doi:10.1002/art.23260
    • (2008) Arthritis Rheum , vol.58 , Issue.3 , pp. 649-656
    • Sieper, J.1    Appel, H.2    Braun, J.3    Rudwaleit, M.4
  • 54
    • 13844309261 scopus 로고    scopus 로고
    • Prostaglandin E2 enhances alveolar bone formation in the rat mandible
    • Ramirez-Yanez GO, Seymour GJ, Walsh LJ, Forwood MR, Symons AL. Prostaglandin E2 enhances alveolar bone formation in the rat mandible. Bone (2004) 35(6):1361-8. doi:10.1016/j.bone.2004.08.006
    • (2004) Bone , vol.35 , Issue.6 , pp. 1361-1368
    • Ramirez-Yanez, G.O.1    Seymour, G.J.2    Walsh, L.J.3    Forwood, M.R.4    Symons, A.L.5
  • 55
    • 0026508139 scopus 로고
    • Stimulation of bone nodule formation in vitro by prostaglandins E1 and E2
    • Flanagan AM, Chambers TJ. Stimulation of bone nodule formation in vitro by prostaglandins E1 and E2. Endocrinology (1992) 130(1):443-8. doi:10.1210/endo.130.1.1309342
    • (1992) Endocrinology , vol.130 , Issue.1 , pp. 443-448
    • Flanagan, A.M.1    Chambers, T.J.2
  • 56
    • 0031982941 scopus 로고    scopus 로고
    • Effects of Tripterygium wilfordii Hook F extracts on induction of cyclooxygenase 2 activity and prostaglandin E2 production
    • Tao X, Schulze-Koops H, Ma L, Cai J, Mao Y, Lipsky PE. Effects of Tripterygium wilfordii Hook F extracts on induction of cyclooxygenase 2 activity and prostaglandin E2 production. Arthritis Rheum (1998) 41(1):130-8. doi:10.1002/1529-0131(199801)41:1<130::AID-ART16>3.3.CO;2-W
    • (1998) Arthritis Rheum , vol.41 , Issue.1 , pp. 130-138
    • Tao, X.1    Schulze-Koops, H.2    Ma, L.3    Cai, J.4    Mao, Y.5    Lipsky, P.E.6
  • 57
    • 84960421708 scopus 로고    scopus 로고
    • Celastrol inhibits prostaglandin E2-induced proliferation and osteogenic differentiation of fibroblasts isolated from ankylosing spondylitis hip tissues in vitro
    • Zou YC, Yang XW, Yuan SG, Zhang P, Li YK. Celastrol inhibits prostaglandin E2-induced proliferation and osteogenic differentiation of fibroblasts isolated from ankylosing spondylitis hip tissues in vitro. Drug Des Devel Ther (2016) 10:933-48. doi:10.2147/DDDT.S97463
    • (2016) Drug Des Devel Ther , vol.10 , pp. 933-948
    • Zou, Y.C.1    Yang, X.W.2    Yuan, S.G.3    Zhang, P.4    Li, Y.K.5
  • 58
    • 84925884782 scopus 로고    scopus 로고
    • Celastrol attenuates inflammatory and neuropathic pain mediated by cannabinoid receptor type 2
    • Yang L, Li Y, Ren J, Zhu C, Fu J, Lin D, et al. Celastrol attenuates inflammatory and neuropathic pain mediated by cannabinoid receptor type 2. Int J Mol Sci (2014) 15(8):13637-48. doi:10.3390/ijms150813637
    • (2014) Int J Mol Sci , vol.15 , Issue.8 , pp. 13637-13648
    • Yang, L.1    Li, Y.2    Ren, J.3    Zhu, C.4    Fu, J.5    Lin, D.6
  • 59
    • 0036019404 scopus 로고    scopus 로고
    • Endocannabinoids and pain: spinal and peripheral analgesia in inflammation and neuropathy
    • Rice AS, Farquhar-Smith WP, Nagy I. Endocannabinoids and pain: spinal and peripheral analgesia in inflammation and neuropathy. Prostaglandins Leukot Essent Fatty Acids (2002) 66(2-3):243-56. doi:10.1054/plef.2001.0362
    • (2002) Prostaglandins Leukot Essent Fatty Acids , vol.66 , Issue.2-3 , pp. 243-256
    • Rice, A.S.1    Farquhar-Smith, W.P.2    Nagy, I.3
  • 60
    • 0037379509 scopus 로고    scopus 로고
    • Observation on serum anti-double stranded DNA antibodies of tripterine in systemic lupus erythematosus of (NZBxW)F1 mice
    • Xu X, Wu Z, Xu C, Ren Y, Ge Y. Observation on serum anti-double stranded DNA antibodies of tripterine in systemic lupus erythematosus of (NZBxW)F1 mice. Ann Rheum Dis (2003) 62(4):377-8. doi:10.1136/ard.62.4.377
    • (2003) Ann Rheum Dis , vol.62 , Issue.4 , pp. 377-378
    • Xu, X.1    Wu, Z.2    Xu, C.3    Ren, Y.4    Ge, Y.5
  • 61
    • 17444428634 scopus 로고    scopus 로고
    • Beneficial effect of tripterine on systemic lupus erythematosus induced by active chromatin in BALB/c mice
    • Li H, Zhang YY, Huang XY, Sun YN, Jia YF, Li D. Beneficial effect of tripterine on systemic lupus erythematosus induced by active chromatin in BALB/c mice. Eur J Pharmacol (2005) 512(2-3):231-7. doi:10.1016/j.ejphar.2005.02.030
    • (2005) Eur J Pharmacol , vol.512 , Issue.2-3 , pp. 231-237
    • Li, H.1    Zhang, Y.Y.2    Huang, X.Y.3    Sun, Y.N.4    Jia, Y.F.5    Li, D.6
  • 62
    • 0037148940 scopus 로고    scopus 로고
    • Genes, microbes, and T cells-new therapeutic targets in Crohn's disease
    • Elson CO. Genes, microbes, and T cells-new therapeutic targets in Crohn's disease. N Engl J Med (2002) 346(8):614-6. doi:10.1056/NEJM200202213460812
    • (2002) N Engl J Med , vol.346 , Issue.8 , pp. 614-616
    • Elson, C.O.1
  • 63
    • 0036284984 scopus 로고    scopus 로고
    • The role of Th1/Th2 polarization in mucosal immunity
    • Neurath MF, Finotto S, Glimcher LH. The role of Th1/Th2 polarization in mucosal immunity. Nat Med (2002) 8(6):567-73. doi:10.1038/nm0602-567
    • (2002) Nat Med , vol.8 , Issue.6 , pp. 567-573
    • Neurath, M.F.1    Finotto, S.2    Glimcher, L.H.3
  • 65
    • 84892779067 scopus 로고    scopus 로고
    • Celastrol ameliorates murine colitis via modulating oxidative stress, inflammatory cytokines and intestinal homeostasis
    • Shaker ME, Ashamallah SA, Houssen ME. Celastrol ameliorates murine colitis via modulating oxidative stress, inflammatory cytokines and intestinal homeostasis. Chem Biol Interact (2014) 210:26-33. doi:10.1016/j.cbi.2013.12.007
    • (2014) Chem Biol Interact , vol.210 , pp. 26-33
    • Shaker, M.E.1    Ashamallah, S.A.2    Houssen, M.E.3
  • 66
    • 84948446834 scopus 로고    scopus 로고
    • The natural compound celastrol inhibits necroptosis and alleviates ulcerative colitis in mice
    • Jia Z, Xu C, Shen J, Xia T, Yang J, He Y. The natural compound celastrol inhibits necroptosis and alleviates ulcerative colitis in mice. Int Immunopharmacol (2015) 29(2):552-9. doi:10.1016/j.intimp.2015.09.029
    • (2015) Int Immunopharmacol , vol.29 , Issue.2 , pp. 552-559
    • Jia, Z.1    Xu, C.2    Shen, J.3    Xia, T.4    Yang, J.5    He, Y.6
  • 67
    • 80053615114 scopus 로고    scopus 로고
    • LITAF mediation of increased TNF-alpha secretion from inflamed colonic lamina propria macrophages
    • Bushell KN, Leeman SE, Gillespie E, Gower AC, Reed KL, Stucchi AF, et al. LITAF mediation of increased TNF-alpha secretion from inflamed colonic lamina propria macrophages. PLoS One (2011) 6(9):e25849. doi:10.1371/journal.pone.0025849
    • (2011) PLoS One , vol.6 , Issue.9
    • Bushell, K.N.1    Leeman, S.E.2    Gillespie, E.3    Gower, A.C.4    Reed, K.L.5    Stucchi, A.F.6
  • 68
    • 84926625663 scopus 로고    scopus 로고
    • Celastrol ameliorates experimental colitis in IL-10 deficient mice via the up-regulation of autophagy
    • Zhao J, Sun Y, Shi P, Dong JN, Zuo LG, Wang HG, et al. Celastrol ameliorates experimental colitis in IL-10 deficient mice via the up-regulation of autophagy. Int Immunopharmacol (2015) 26(1):221-8. doi:10.1016/j.intimp.2015.03.033
    • (2015) Int Immunopharmacol , vol.26 , Issue.1 , pp. 221-228
    • Zhao, J.1    Sun, Y.2    Shi, P.3    Dong, J.N.4    Zuo, L.G.5    Wang, H.G.6
  • 69
    • 84962028201 scopus 로고    scopus 로고
    • Celastrol ameliorates ulcerative colitis-related colorectal cancer in mice via suppressing inflammatory responses and epithelial-mesenchymal transition
    • Lin L, Sun Y, Wang D, Zheng S, Zhang J, Zheng C. Celastrol ameliorates ulcerative colitis-related colorectal cancer in mice via suppressing inflammatory responses and epithelial-mesenchymal transition. Front Pharmacol (2015) 6:320. doi:10.3389/fphar.2015.00320
    • (2015) Front Pharmacol , vol.6 , pp. 320
    • Lin, L.1    Sun, Y.2    Wang, D.3    Zheng, S.4    Zhang, J.5    Zheng, C.6
  • 70
    • 84966389760 scopus 로고    scopus 로고
    • A lipidomics investigation into the intervention of celastrol in experimental colitis
    • Wang R, Gu X, Dai W, Ye J, Lu F, Chai Y, et al. A lipidomics investigation into the intervention of celastrol in experimental colitis. Mol Biosyst (2016) 12(5):1436-44. doi:10.1039/c5mb00864f
    • (2016) Mol Biosyst , vol.12 , Issue.5 , pp. 1436-1444
    • Wang, R.1    Gu, X.2    Dai, W.3    Ye, J.4    Lu, F.5    Chai, Y.6
  • 71
    • 84877818621 scopus 로고    scopus 로고
    • Celastrol, an inhibitor of heat shock protein 90beta potently suppresses the expression of matrix metalloproteinases, inducible nitric oxide synthase and cyclooxygenase-2 in primary human osteoarthritic chondrocytes
    • Ding QH, Cheng Y, Chen WP, Zhong HM, Wang XH. Celastrol, an inhibitor of heat shock protein 90beta potently suppresses the expression of matrix metalloproteinases, inducible nitric oxide synthase and cyclooxygenase-2 in primary human osteoarthritic chondrocytes. Eur J Pharmacol (2013) 708(1-3):1-7. doi:10.1016/j.ejphar.2013.01.057
    • (2013) Eur J Pharmacol , vol.708 , Issue.1-3 , pp. 1-7
    • Ding, Q.H.1    Cheng, Y.2    Chen, W.P.3    Zhong, H.M.4    Wang, X.H.5
  • 72
    • 0037326942 scopus 로고    scopus 로고
    • Chemokines in asthma: cooperative interaction between chemokines and IL-13
    • Zimmermann N, Hershey GK, Foster PS, Rothenberg ME. Chemokines in asthma: cooperative interaction between chemokines and IL-13. J Allergy Clin Immunol (2003) 111(2):227-42. doi:10.1067/mai.2003.139 quiz 43
    • (2003) J Allergy Clin Immunol , vol.111 , Issue.2 , pp. 227-242
    • Zimmermann, N.1    Hershey, G.K.2    Foster, P.S.3    Rothenberg, M.E.4
  • 73
    • 0033120966 scopus 로고    scopus 로고
    • Inhibition of matrix metalloproteinases prevents allergen-induced airway inflammation in a murine model of asthma
    • Kumagai K, Ohno I, Okada S, Ohkawara Y, Suzuki K, Shinya T, et al. Inhibition of matrix metalloproteinases prevents allergen-induced airway inflammation in a murine model of asthma. J Immunol (1999) 162(7):4212-9
    • (1999) J Immunol , vol.162 , Issue.7 , pp. 4212-4219
    • Kumagai, K.1    Ohno, I.2    Okada, S.3    Ohkawara, Y.4    Suzuki, K.5    Shinya, T.6
  • 74
    • 0036679426 scopus 로고    scopus 로고
    • Overlapping and enzyme-specific contributions of matrix metalloproteinases-9 and-12 in IL-13-induced inflammation and remodeling
    • Lanone S, Zheng T, Zhu Z, Liu W, Lee CG, Ma B, et al. Overlapping and enzyme-specific contributions of matrix metalloproteinases-9 and-12 in IL-13-induced inflammation and remodeling. J Clin Invest (2002) 110(4):463-74. doi:10.1172/JCI0214136
    • (2002) J Clin Invest , vol.110 , Issue.4 , pp. 463-474
    • Lanone, S.1    Zheng, T.2    Zhu, Z.3    Liu, W.4    Lee, C.G.5    Ma, B.6
  • 75
    • 4344703841 scopus 로고    scopus 로고
    • [The experimental study on the inhibitory effect of tripterine on airway inflammation in asthmatic mice]
    • Liu RL, Liu ZL, Li Q, Qiu ZM, Lu HJ, Yang ZM, et al. [The experimental study on the inhibitory effect of tripterine on airway inflammation in asthmatic mice]. Zhonghua Jie He He Hu Xi Za Zhi (2004) 27(3):165-8
    • (2004) Zhonghua Jie He He Hu Xi Za Zhi , vol.27 , Issue.3 , pp. 165-168
    • Liu, R.L.1    Liu, Z.L.2    Li, Q.3    Qiu, Z.M.4    Lu, H.J.5    Yang, Z.M.6
  • 76
    • 65549142792 scopus 로고    scopus 로고
    • Celastrol suppresses allergen-induced airway inflammation in a mouse allergic asthma model
    • Kim DY, Park JW, Jeoung D, Ro JY. Celastrol suppresses allergen-induced airway inflammation in a mouse allergic asthma model. Eur J Pharmacol (2009) 612(1-3):98-105. doi:10.1016/j.ejphar.2009.03.078
    • (2009) Eur J Pharmacol , vol.612 , Issue.1-3 , pp. 98-105
    • Kim, D.Y.1    Park, J.W.2    Jeoung, D.3    Ro, J.Y.4
  • 77
    • 65549157628 scopus 로고    scopus 로고
    • Celastrol binds to ERK and inhibits FcepsilonRI signaling to exert an anti-allergic effect
    • Kim Y, Kim K, Lee H, Han S, Lee YS, Choe J, et al. Celastrol binds to ERK and inhibits FcepsilonRI signaling to exert an anti-allergic effect. Eur J Pharmacol (2009) 612(1-3):131-42. doi:10.1016/j.ejphar.2009.03.071
    • (2009) Eur J Pharmacol , vol.612 , Issue.1-3 , pp. 131-142
    • Kim, Y.1    Kim, K.2    Lee, H.3    Han, S.4    Lee, Y.S.5    Choe, J.6
  • 78
    • 84927611106 scopus 로고    scopus 로고
    • Celastrol induces apoptosis and autophagy via the ROS/JNK signaling pathway in human osteosarcoma cells: an in vitro and in vivo study
    • Li HY, Zhang J, Sun LL, Li BH, Gao HL, Xie T, et al. Celastrol induces apoptosis and autophagy via the ROS/JNK signaling pathway in human osteosarcoma cells: an in vitro and in vivo study. Cell Death Dis (2015) 6:e1604. doi:10.1038/cddis.2014.543
    • (2015) Cell Death Dis , vol.6
    • Li, H.Y.1    Zhang, J.2    Sun, L.L.3    Li, B.H.4    Gao, H.L.5    Xie, T.6
  • 79
    • 84922272575 scopus 로고    scopus 로고
    • Celastrol inhibits gastric cancer growth by induction of apoptosis and autophagy
    • Lee HW, Jang KS, Choi HJ, Jo A, Cheong JH, Chun KH. Celastrol inhibits gastric cancer growth by induction of apoptosis and autophagy. BMB Rep (2014) 47(12):697-702. doi:10.5483/BMBRep.2014.47.12.069
    • (2014) BMB Rep , vol.47 , Issue.12 , pp. 697-702
    • Lee, H.W.1    Jang, K.S.2    Choi, H.J.3    Jo, A.4    Cheong, J.H.5    Chun, K.H.6
  • 80
    • 84892417914 scopus 로고    scopus 로고
    • Celastrol induces apoptosis of gastric cancer cells by miR-21 inhibiting PI3K/Akt-NF-kappaB signaling pathway
    • Sha M, Ye J, Zhang LX, Luan ZY, Chen YB, Huang JX. Celastrol induces apoptosis of gastric cancer cells by miR-21 inhibiting PI3K/Akt-NF-kappaB signaling pathway. Pharmacology (2014) 93(1-2):39-46. doi:10.1159/000357683
    • (2014) Pharmacology , vol.93 , Issue.1-2 , pp. 39-46
    • Sha, M.1    Ye, J.2    Zhang, L.X.3    Luan, Z.Y.4    Chen, Y.B.5    Huang, J.X.6
  • 81
    • 84945126912 scopus 로고    scopus 로고
    • Celastrol induces cell cycle arrest by MicroRNA-21-mTOR-mediated inhibition p27 protein degradation in gastric cancer
    • Sha M, Ye J, Luan ZY, Guo T, Wang B, Huang JX. Celastrol induces cell cycle arrest by MicroRNA-21-mTOR-mediated inhibition p27 protein degradation in gastric cancer. Cancer Cell Int (2015) 15:101. doi:10.1186/s12935-015-0256-3
    • (2015) Cancer Cell Int , vol.15 , pp. 101
    • Sha, M.1    Ye, J.2    Luan, Z.Y.3    Guo, T.4    Wang, B.5    Huang, J.X.6
  • 82
    • 84948977272 scopus 로고    scopus 로고
    • Celastrol induces autophagy by targeting AR/miR-101 in prostate cancer cells
    • Guo J, Huang X, Wang H, Yang H. Celastrol induces autophagy by targeting AR/miR-101 in prostate cancer cells. PLoS One (2015) 10(10):e0140745. doi:10.1371/journal.pone.0140745
    • (2015) PLoS One , vol.10 , Issue.10
    • Guo, J.1    Huang, X.2    Wang, H.3    Yang, H.4
  • 83
    • 33749433916 scopus 로고    scopus 로고
    • Gene expression signature-based chemical genomic prediction identifies a novel class of HSP90 pathway modulators
    • Hieronymus H, Lamb J, Ross KN, Peng XP, Clement C, Rodina A, et al. Gene expression signature-based chemical genomic prediction identifies a novel class of HSP90 pathway modulators. Cancer Cell (2006) 10(4):321-30. doi:10.1016/j.ccr.2006.09.005
    • (2006) Cancer Cell , vol.10 , Issue.4 , pp. 321-330
    • Hieronymus, H.1    Lamb, J.2    Ross, K.N.3    Peng, X.P.4    Clement, C.5    Rodina, A.6
  • 84
    • 54049150008 scopus 로고    scopus 로고
    • Calpain-mediated androgen receptor breakdown in apoptotic prostate cancer cells
    • Yang H, Murthy S, Sarkar FH, Sheng S, Reddy GP, Dou QP. Calpain-mediated androgen receptor breakdown in apoptotic prostate cancer cells. J Cell Physiol (2008) 217(3):569-76. doi:10.1002/jcp.21565
    • (2008) J Cell Physiol , vol.217 , Issue.3 , pp. 569-576
    • Yang, H.1    Murthy, S.2    Sarkar, F.H.3    Sheng, S.4    Reddy, G.P.5    Dou, Q.P.6
  • 85
    • 84911420350 scopus 로고    scopus 로고
    • Celastrol stimulates hypoxia-inducible factor-1 activity in tumor cells by initiating the ROS/Akt/p70S6K signaling pathway and enhancing hypoxia-inducible factor-1alpha protein synthesis
    • Han X, Sun S, Zhao M, Cheng X, Chen G, Lin S, et al. Celastrol stimulates hypoxia-inducible factor-1 activity in tumor cells by initiating the ROS/Akt/p70S6K signaling pathway and enhancing hypoxia-inducible factor-1alpha protein synthesis. PLoS One (2014) 9(11):e112470. doi:10.1371/journal.pone.0112470
    • (2014) PLoS One , vol.9 , Issue.11
    • Han, X.1    Sun, S.2    Zhao, M.3    Cheng, X.4    Chen, G.5    Lin, S.6
  • 86
    • 38349153572 scopus 로고    scopus 로고
    • A novel Hsp90 inhibitor to disrupt Hsp90/Cdc37 complex against pancreatic cancer cells
    • Zhang T, Hamza A, Cao X, Wang B, Yu S, Zhan CG, et al. A novel Hsp90 inhibitor to disrupt Hsp90/Cdc37 complex against pancreatic cancer cells. Mol Cancer Ther (2008) 7(1):162-70. doi:10.1158/1535-7163.MCT-07-0484
    • (2008) Mol Cancer Ther , vol.7 , Issue.1 , pp. 162-170
    • Zhang, T.1    Hamza, A.2    Cao, X.3    Wang, B.4    Yu, S.5    Zhan, C.G.6
  • 87
    • 70349921403 scopus 로고    scopus 로고
    • Molecular mechanism of inhibition of the human protein complex Hsp90-Cdc37, a kinome chaperone-cochaperone, by triterpene celastrol
    • Sreeramulu S, Gande SL, Gobel M, Schwalbe H. Molecular mechanism of inhibition of the human protein complex Hsp90-Cdc37, a kinome chaperone-cochaperone, by triterpene celastrol. Angew Chem Int Ed Engl (2009) 48(32):5853-5. doi:10.1002/anie.200900929
    • (2009) Angew Chem Int Ed Engl , vol.48 , Issue.32 , pp. 5853-5855
    • Sreeramulu, S.1    Gande, S.L.2    Gobel, M.3    Schwalbe, H.4
  • 88
    • 77950485584 scopus 로고    scopus 로고
    • Celastrol inhibits Hsp90 chaperoning of steroid receptors by inducing fibrillization of the co-chaperone p23
    • Chadli A, Felts SJ, Wang Q, Sullivan WP, Botuyan MV, Fauq A, et al. Celastrol inhibits Hsp90 chaperoning of steroid receptors by inducing fibrillization of the co-chaperone p23. J Biol Chem (2010) 285(6):4224-31. doi:10.1074/jbc.M109.081018
    • (2010) J Biol Chem , vol.285 , Issue.6 , pp. 4224-4231
    • Chadli, A.1    Felts, S.J.2    Wang, Q.3    Sullivan, W.P.4    Botuyan, M.V.5    Fauq, A.6
  • 89
    • 84907882487 scopus 로고    scopus 로고
    • The effect of celastrol, a triterpene with antitumorigenic activity, on conformational and functional aspects of the human 90kDa heat shock protein Hsp90alpha, a chaperone implicated in the stabilization of the tumor phenotype
    • Zanphorlin LM, Alves FR, Ramos CH. The effect of celastrol, a triterpene with antitumorigenic activity, on conformational and functional aspects of the human 90kDa heat shock protein Hsp90alpha, a chaperone implicated in the stabilization of the tumor phenotype. Biochim Biophys Acta (2014) 1840(10):3145-52. doi:10.1016/j.bbagen.2014.06.008
    • (2014) Biochim Biophys Acta , vol.1840 , Issue.10 , pp. 3145-3152
    • Zanphorlin, L.M.1    Alves, F.R.2    Ramos, C.H.3
  • 90
    • 77951215549 scopus 로고    scopus 로고
    • Celastrol, a triterpene, enhances TRAIL-induced apoptosis through the down-regulation of cell survival proteins and up-regulation of death receptors
    • Sung B, Park B, Yadav VR, Aggarwal BB. Celastrol, a triterpene, enhances TRAIL-induced apoptosis through the down-regulation of cell survival proteins and up-regulation of death receptors. J Biol Chem (2010) 285(15):11498-507. doi:10.1074/jbc.M109.090209
    • (2010) J Biol Chem , vol.285 , Issue.15 , pp. 11498-11507
    • Sung, B.1    Park, B.2    Yadav, V.R.3    Aggarwal, B.B.4
  • 91
    • 77956407726 scopus 로고    scopus 로고
    • Up-regulation of death receptor 4 and 5 by celastrol enhances the anti-cancer activity of TRAIL/Apo-2L
    • Zhu H, Liu XW, Ding WJ, Xu DQ, Zhao YC, Lu W, et al. Up-regulation of death receptor 4 and 5 by celastrol enhances the anti-cancer activity of TRAIL/Apo-2L. Cancer Lett (2010) 297(2):155-64. doi:10.1016/j.canlet.2010.04.030
    • (2010) Cancer Lett , vol.297 , Issue.2 , pp. 155-164
    • Zhu, H.1    Liu, X.W.2    Ding, W.J.3    Xu, D.Q.4    Zhao, Y.C.5    Lu, W.6
  • 92
    • 79957961355 scopus 로고    scopus 로고
    • Celastrol induces apoptosis in non-small-cell lung cancer A549 cells through activation of mitochondria-and Fas/FasL-mediated pathways
    • Mou H, Zheng Y, Zhao P, Bao H, Fang W, Xu N. Celastrol induces apoptosis in non-small-cell lung cancer A549 cells through activation of mitochondria-and Fas/FasL-mediated pathways. Toxicol In Vitro (2011) 25(5):1027-32. doi:10.1016/j.tiv.2011.03.023
    • (2011) Toxicol In Vitro , vol.25 , Issue.5 , pp. 1027-1032
    • Mou, H.1    Zheng, Y.2    Zhao, P.3    Bao, H.4    Fang, W.5    Xu, N.6
  • 93
    • 79955855304 scopus 로고    scopus 로고
    • Celastrol targets mitochondrial respiratory chain complex I to induce reactive oxygen species-dependent cytotoxicity in tumor cells
    • Chen G, Zhang X, Zhao M, Wang Y, Cheng X, Wang D, et al. Celastrol targets mitochondrial respiratory chain complex I to induce reactive oxygen species-dependent cytotoxicity in tumor cells. BMC Cancer (2011) 11:170. doi:10.1186/1471-2407-11-170
    • (2011) BMC Cancer , vol.11 , pp. 170
    • Chen, G.1    Zhang, X.2    Zhao, M.3    Wang, Y.4    Cheng, X.5    Wang, D.6
  • 94
    • 84937200175 scopus 로고    scopus 로고
    • Celastrol induces apoptosis of human osteosarcoma cells via the mitochondrial apoptotic pathway
    • Yu X, Zhou X, Fu C, Wang Q, Nie T, Zou F, et al. Celastrol induces apoptosis of human osteosarcoma cells via the mitochondrial apoptotic pathway. Oncol Rep (2015) 34(3):1129-36. doi:10.3892/or.2015.4124
    • (2015) Oncol Rep , vol.34 , Issue.3 , pp. 1129-1136
    • Yu, X.1    Zhou, X.2    Fu, C.3    Wang, Q.4    Nie, T.5    Zou, F.6
  • 95
    • 84928476141 scopus 로고    scopus 로고
    • Celastrol induces mitochondria-mediated apoptosis in hepatocellular carcinoma Bel-7402 cells
    • Li PP, He W, Yuan PF, Song SS, Lu JT, Wei W. Celastrol induces mitochondria-mediated apoptosis in hepatocellular carcinoma Bel-7402 cells. Am J Chin Med (2015) 43(1):137-48. doi:10.1142/S0192415X15500093
    • (2015) Am J Chin Med , vol.43 , Issue.1 , pp. 137-148
    • Li, P.P.1    He, W.2    Yuan, P.F.3    Song, S.S.4    Lu, J.T.5    Wei, W.6
  • 96
    • 84926296864 scopus 로고    scopus 로고
    • Anticancer effect of celastrol on human triple negative breast cancer: possible involvement of oxidative stress, mitochondrial dysfunction, apoptosis and PI3K/Akt pathways
    • Shrivastava S, Jeengar MK, Reddy VS, Reddy GB, Naidu VG. Anticancer effect of celastrol on human triple negative breast cancer: possible involvement of oxidative stress, mitochondrial dysfunction, apoptosis and PI3K/Akt pathways. Exp Mol Pathol (2015) 98(3):313-27. doi:10.1016/j.yexmp.2015.03.031
    • (2015) Exp Mol Pathol , vol.98 , Issue.3 , pp. 313-327
    • Shrivastava, S.1    Jeengar, M.K.2    Reddy, V.S.3    Reddy, G.B.4    Naidu, V.G.5
  • 97
    • 84872054236 scopus 로고    scopus 로고
    • Celastrol inhibits growth and induces apoptotic cell death in melanoma cells via the activation ROS-dependent mitochondrial pathway and the suppression of PI3K/AKT signaling
    • Lee JH, Won YS, Park KH, Lee MK, Tachibana H, Yamada K, et al. Celastrol inhibits growth and induces apoptotic cell death in melanoma cells via the activation ROS-dependent mitochondrial pathway and the suppression of PI3K/AKT signaling. Apoptosis (2012) 17(12):1275-86. doi:10.1007/s10495-012-0767-5
    • (2012) Apoptosis , vol.17 , Issue.12 , pp. 1275-1286
    • Lee, J.H.1    Won, Y.S.2    Park, K.H.3    Lee, M.K.4    Tachibana, H.5    Yamada, K.6
  • 98
    • 82955162775 scopus 로고    scopus 로고
    • Celastrol inhibits tumor cell proliferation and promotes apoptosis through the activation of c-Jun N-terminal kinase and suppression of PI3 K/Akt signaling pathways
    • Kannaiyan R, Manu KA, Chen L, Li F, Rajendran P, Subramaniam A, et al. Celastrol inhibits tumor cell proliferation and promotes apoptosis through the activation of c-Jun N-terminal kinase and suppression of PI3 K/Akt signaling pathways. Apoptosis (2011) 16(10):1028-41. doi:10.1007/s10495-011-0629-6
    • (2011) Apoptosis , vol.16 , Issue.10 , pp. 1028-1041
    • Kannaiyan, R.1    Manu, K.A.2    Chen, L.3    Li, F.4    Rajendran, P.5    Subramaniam, A.6
  • 99
    • 84873313731 scopus 로고    scopus 로고
    • Celastrol suppresses breast cancer MCF-7 cell viability via the AMP-activated protein kinase (AMPK)-induced p53-polo like kinase 2 (PLK-2) pathway
    • Kim JH, Lee JO, Lee SK, Kim N, You GY, Moon JW, et al. Celastrol suppresses breast cancer MCF-7 cell viability via the AMP-activated protein kinase (AMPK)-induced p53-polo like kinase 2 (PLK-2) pathway. Cell Signal (2013) 25(4):805-13. doi:10.1016/j.cellsig.2012.12.005
    • (2013) Cell Signal , vol.25 , Issue.4 , pp. 805-813
    • Kim, J.H.1    Lee, J.O.2    Lee, S.K.3    Kim, N.4    You, G.Y.5    Moon, J.W.6
  • 100
    • 78751506175 scopus 로고    scopus 로고
    • Anticancer activity of celastrol in combination with ErbB2-targeted therapeutics for treatment of ErbB2-overexpressing breast cancers
    • Raja SM, Clubb RJ, Ortega-Cava C, Williams SH, Bailey TA, Duan L, et al. Anticancer activity of celastrol in combination with ErbB2-targeted therapeutics for treatment of ErbB2-overexpressing breast cancers. Cancer Biol Ther (2011) 11(2):263-76. doi:10.4161/cbt.11.2.13959
    • (2011) Cancer Biol Ther , vol.11 , Issue.2 , pp. 263-276
    • Raja, S.M.1    Clubb, R.J.2    Ortega-Cava, C.3    Williams, S.H.4    Bailey, T.A.5    Duan, L.6
  • 101
    • 78549293218 scopus 로고    scopus 로고
    • Celastrol inhibits the growth of estrogen positive human breast cancer cells through modulation of estrogen receptor alpha
    • Jang SY, Jang SW, Ko J. Celastrol inhibits the growth of estrogen positive human breast cancer cells through modulation of estrogen receptor alpha. Cancer Lett (2011) 300(1):57-65. doi:10.1016/j.canlet.2010.09.006
    • (2011) Cancer Lett , vol.300 , Issue.1 , pp. 57-65
    • Jang, S.Y.1    Jang, S.W.2    Ko, J.3
  • 102
    • 78951492587 scopus 로고    scopus 로고
    • Celastrol isolated from Tripterygium regelii induces apoptosis through both caspase-dependent and-independent pathways in human breast cancer cells
    • Yang HS, Kim JY, Lee JH, Lee BW, Park KH, Shim KH, et al. Celastrol isolated from Tripterygium regelii induces apoptosis through both caspase-dependent and-independent pathways in human breast cancer cells. Food Chem Toxicol (2011) 49(2):527-32. doi:10.1016/j.fct.2010.11.044
    • (2011) Food Chem Toxicol , vol.49 , Issue.2 , pp. 527-532
    • Yang, H.S.1    Kim, J.Y.2    Lee, J.H.3    Lee, B.W.4    Park, K.H.5    Shim, K.H.6
  • 103
    • 2842616065 scopus 로고    scopus 로고
    • Apoptosis induction in HL-60 cells and inhibition of topoisomerase II by triterpene celastrol
    • Nagase M, Oto J, Sugiyama S, Yube K, Takaishi Y, Sakato N. Apoptosis induction in HL-60 cells and inhibition of topoisomerase II by triterpene celastrol. Biosci Biotechnol Biochem (2003) 67(9):1883-7. doi:10.1271/bbb.67.1883
    • (2003) Biosci Biotechnol Biochem , vol.67 , Issue.9 , pp. 1883-1887
    • Nagase, M.1    Oto, J.2    Sugiyama, S.3    Yube, K.4    Takaishi, Y.5    Sakato, N.6
  • 104
    • 84973512749 scopus 로고    scopus 로고
    • Celastrol induces cell apoptosis and inhibits the expression of the AML1-ETO/C-KIT oncoprotein in t(8;21) leukemia
    • Yu X, Ruan X, Zhang J, Zhao Q. Celastrol induces cell apoptosis and inhibits the expression of the AML1-ETO/C-KIT oncoprotein in t(8;21) leukemia. Molecules (2016) 21:5. doi:10.3390/molecules21050574
    • (2016) Molecules , vol.21 , pp. 5
    • Yu, X.1    Ruan, X.2    Zhang, J.3    Zhao, Q.4
  • 105
    • 84864835881 scopus 로고    scopus 로고
    • Celastrol suppresses growth and induces apoptosis of human hepatocellular carcinoma through the modulation of STAT3/JAK2 signaling cascade in vitro and in vivo
    • Rajendran P, Li F, Shanmugam MK, Kannaiyan R, Goh JN, Wong KF, et al. Celastrol suppresses growth and induces apoptosis of human hepatocellular carcinoma through the modulation of STAT3/JAK2 signaling cascade in vitro and in vivo. Cancer Prev Res (Phila) (2012) 5(4):631-43. doi:10.1158/1940-6207.CAPR-11-0420
    • (2012) Cancer Prev Res (Phila) , vol.5 , Issue.4 , pp. 631-643
    • Rajendran, P.1    Li, F.2    Shanmugam, M.K.3    Kannaiyan, R.4    Goh, J.N.5    Wong, K.F.6
  • 106
    • 84960341337 scopus 로고    scopus 로고
    • Targeting acute myeloid leukemia with a small molecule inhibitor of the Myb/p300 interaction
    • Uttarkar S, Dasse E, Coulibaly A, Steinmann S, Jakobs A, Schomburg C, et al. Targeting acute myeloid leukemia with a small molecule inhibitor of the Myb/p300 interaction. Blood (2016) 127(9):1173-82. doi:10.1182/blood-2015-09-668632
    • (2016) Blood , vol.127 , Issue.9 , pp. 1173-1182
    • Uttarkar, S.1    Dasse, E.2    Coulibaly, A.3    Steinmann, S.4    Jakobs, A.5    Schomburg, C.6
  • 107
    • 84919782859 scopus 로고    scopus 로고
    • Celastrol induces unfolded protein response-dependent cell death in head and neck cancer
    • Fribley AM, Miller JR, Brownell AL, Garshott DM, Zeng Q, Reist TE, et al. Celastrol induces unfolded protein response-dependent cell death in head and neck cancer. Exp Cell Res (2015) 330(2):412-22. doi:10.1016/j.yexcr.2014.08.014
    • (2015) Exp Cell Res , vol.330 , Issue.2 , pp. 412-422
    • Fribley, A.M.1    Miller, J.R.2    Brownell, A.L.3    Garshott, D.M.4    Zeng, Q.5    Reist, T.E.6
  • 108
    • 84881045284 scopus 로고    scopus 로고
    • ER stress-mediated apoptosis induced by celastrol in cancer cells and important role of glycogen synthase kinase-3beta in the signal network
    • Feng L, Zhang D, Fan C, Ma C, Yang W, Meng Y, et al. ER stress-mediated apoptosis induced by celastrol in cancer cells and important role of glycogen synthase kinase-3beta in the signal network. Cell Death Dis (2013) 4:e715. doi:10.1038/cddis.2013.222
    • (2013) Cell Death Dis , vol.4
    • Feng, L.1    Zhang, D.2    Fan, C.3    Ma, C.4    Yang, W.5    Meng, Y.6
  • 109
    • 84901012573 scopus 로고    scopus 로고
    • Celastrol targets proteostasis and acts synergistically with a heat-shock protein 90 inhibitor to kill human glioblastoma cells
    • Boridy S, Le PU, Petrecca K, Maysinger D. Celastrol targets proteostasis and acts synergistically with a heat-shock protein 90 inhibitor to kill human glioblastoma cells. Cell Death Dis (2014) 5:e1216. doi:10.1038/cddis.2014.182
    • (2014) Cell Death Dis , vol.5
    • Boridy, S.1    Le, P.U.2    Petrecca, K.3    Maysinger, D.4
  • 110
    • 84926500135 scopus 로고    scopus 로고
    • Effect of celastrol on growth inhibition of prostate cancer cells through the regulation of hERG channel in vitro
    • Ji N, Li J, Wei Z, Kong F, Jin H, Chen X, et al. Effect of celastrol on growth inhibition of prostate cancer cells through the regulation of hERG channel in vitro. Biomed Res Int (2015) 2015:308475. doi:10.1155/2015/308475
    • (2015) Biomed Res Int , vol.2015
    • Ji, N.1    Li, J.2    Wei, Z.3    Kong, F.4    Jin, H.5    Chen, X.6
  • 111
    • 84874610297 scopus 로고    scopus 로고
    • Celastrol suppresses tumor cell growth through targeting an AR-ERG-NF-kappaB pathway in TMPRSS2/ERG fusion gene expressing prostate cancer
    • Shao L, Zhou Z, Cai Y, Castro P, Dakhov O, Shi P, et al. Celastrol suppresses tumor cell growth through targeting an AR-ERG-NF-kappaB pathway in TMPRSS2/ERG fusion gene expressing prostate cancer. PLoS One (2013) 8(3):e58391. doi:10.1371/journal.pone.0058391
    • (2013) PLoS One , vol.8 , Issue.3
    • Shao, L.1    Zhou, Z.2    Cai, Y.3    Castro, P.4    Dakhov, O.5    Shi, P.6
  • 112
    • 84878050153 scopus 로고    scopus 로고
    • Celastrol induces apoptosis of gastric cancer cells by miR-146a inhibition of NF-kappaB activity
    • Sha M, Ye J, Zhang LX, Luan ZY, Chen YB. Celastrol induces apoptosis of gastric cancer cells by miR-146a inhibition of NF-kappaB activity. Cancer Cell Int (2013) 13(1):50. doi:10.1186/1475-2867-13-50
    • (2013) Cancer Cell Int , vol.13 , Issue.1 , pp. 50
    • Sha, M.1    Ye, J.2    Zhang, L.X.3    Luan, Z.Y.4    Chen, Y.B.5
  • 113
    • 80054024030 scopus 로고    scopus 로고
    • Celastrol inhibits proliferation and induces chemosensitization through down-regulation of NF-kappaB and STAT3 regulated gene products in multiple myeloma cells
    • Kannaiyan R, Hay HS, Rajendran P, Li F, Shanmugam MK, Vali S, et al. Celastrol inhibits proliferation and induces chemosensitization through down-regulation of NF-kappaB and STAT3 regulated gene products in multiple myeloma cells. Br J Pharmacol (2011) 164(5):1506-21. doi:10.1111/j.1476-5381.2011.01449.x
    • (2011) Br J Pharmacol , vol.164 , Issue.5 , pp. 1506-1521
    • Kannaiyan, R.1    Hay, H.S.2    Rajendran, P.3    Li, F.4    Shanmugam, M.K.5    Vali, S.6
  • 114
    • 84899694806 scopus 로고    scopus 로고
    • NF-kappa B modulation is involved in celastrol induced human multiple myeloma cell apoptosis
    • Ni H, Zhao W, Kong X, Li H, Ouyang J. NF-kappa B modulation is involved in celastrol induced human multiple myeloma cell apoptosis. PLoS One (2014) 9(4):e95846. doi:10.1371/journal.pone.0095846
    • (2014) PLoS One , vol.9 , Issue.4
    • Ni, H.1    Zhao, W.2    Kong, X.3    Li, H.4    Ouyang, J.5
  • 115
    • 80053192325 scopus 로고    scopus 로고
    • Quinone methide tripterine, celastrol, induces apoptosis in human myeloma cells via NF-kappaB pathway
    • Tozawa K, Sagawa M, Kizaki M. Quinone methide tripterine, celastrol, induces apoptosis in human myeloma cells via NF-kappaB pathway. Int J Oncol (2011) 39(5):1117-22. doi:10.3892/ijo.2011.1161
    • (2011) Int J Oncol , vol.39 , Issue.5 , pp. 1117-1122
    • Tozawa, K.1    Sagawa, M.2    Kizaki, M.3
  • 116
    • 33646406554 scopus 로고    scopus 로고
    • Celastrol, a triterpene extracted from the Chinese "Thunder of God Vine," is a potent proteasome inhibitor and suppresses human prostate cancer growth in nude mice
    • Yang H, Chen D, Cui QC, Yuan X, Dou QP. Celastrol, a triterpene extracted from the Chinese "Thunder of God Vine," is a potent proteasome inhibitor and suppresses human prostate cancer growth in nude mice. Cancer Res (2006) 66(9):4758-65. doi:10.1158/0008-5472.CAN-05-4529
    • (2006) Cancer Res , vol.66 , Issue.9 , pp. 4758-4765
    • Yang, H.1    Chen, D.2    Cui, Q.C.3    Yuan, X.4    Dou, Q.P.5
  • 117
    • 78650733125 scopus 로고    scopus 로고
    • Natural proteasome inhibitor celastrol suppresses androgen-independent prostate cancer progression by modulating apoptotic proteins and NF-kappaB
    • Dai Y, Desano J, Tang W, Meng X, Meng Y, Burstein E, et al. Natural proteasome inhibitor celastrol suppresses androgen-independent prostate cancer progression by modulating apoptotic proteins and NF-kappaB. PLoS One (2010) 5(12):e14153. doi:10.1371/journal.pone.0014153
    • (2010) PLoS One , vol.5 , Issue.12
    • Dai, Y.1    Desano, J.2    Tang, W.3    Meng, X.4    Meng, Y.5    Burstein, E.6
  • 118
    • 79951851668 scopus 로고    scopus 로고
    • Effects of Celastrol on growth inhibition of U937 leukemia cells through the regulation of the Notch1/NF-kappaB signaling pathway in vitro
    • Wang XN, Wu Q, Yang X, Zhang LS, Wu YP, Lu C. Effects of Celastrol on growth inhibition of U937 leukemia cells through the regulation of the Notch1/NF-kappaB signaling pathway in vitro. Chin J Cancer (2010) 29(4):385-90. doi:10.5732/cjc.009.10526
    • (2010) Chin J Cancer , vol.29 , Issue.4 , pp. 385-390
    • Wang, X.N.1    Wu, Q.2    Yang, X.3    Zhang, L.S.4    Wu, Y.P.5    Lu, C.6
  • 119
    • 84899745281 scopus 로고    scopus 로고
    • Celastrol blocks interleukin-6 gene expression via downregulation of NF-kappaB in prostate carcinoma cells
    • Chiang KC, Tsui KH, Chung LC, Yeh CN, Chen WT, Chang PL, et al. Celastrol blocks interleukin-6 gene expression via downregulation of NF-kappaB in prostate carcinoma cells. PLoS One (2014) 9(3):e93151. doi:10.1371/journal.pone.0093151
    • (2014) PLoS One , vol.9 , Issue.3
    • Chiang, K.C.1    Tsui, K.H.2    Chung, L.C.3    Yeh, C.N.4    Chen, W.T.5    Chang, P.L.6
  • 120
    • 67650724069 scopus 로고    scopus 로고
    • Regulation and function of NF-kappaB transcription factors in the immune system
    • Vallabhapurapu S, Karin M. Regulation and function of NF-kappaB transcription factors in the immune system. Annu Rev Immunol (2009) 27:693-733. doi:10.1146/annurev.immunol.021908.132641
    • (2009) Annu Rev Immunol , vol.27 , pp. 693-733
    • Vallabhapurapu, S.1    Karin, M.2
  • 121
    • 84907087401 scopus 로고    scopus 로고
    • Release of Ca2+ from the endoplasmic reticulum and its subsequent influx into mitochondria trigger celastrol-induced paraptosis in cancer cells
    • Yoon MJ, Lee AR, Jeong SA, Kim YS, Kim JY, Kwon YJ, et al. Release of Ca2+ from the endoplasmic reticulum and its subsequent influx into mitochondria trigger celastrol-induced paraptosis in cancer cells. Oncotarget (2014) 5(16):6816-31. doi:10.18632/oncotarget.2256
    • (2014) Oncotarget , vol.5 , Issue.16 , pp. 6816-6831
    • Yoon, M.J.1    Lee, A.R.2    Jeong, S.A.3    Kim, Y.S.4    Kim, J.Y.5    Kwon, Y.J.6
  • 122
    • 79551612454 scopus 로고    scopus 로고
    • Inhibitory action of Celastrol on hypoxia-mediated angiogenesis and metastasis via the HIF-1alpha pathway
    • Huang L, Zhang Z, Zhang S, Ren J, Zhang R, Zeng H, et al. Inhibitory action of Celastrol on hypoxia-mediated angiogenesis and metastasis via the HIF-1alpha pathway. Int J Mol Med (2011) 27(3):407-15. doi:10.3892/ijmm.2011.600
    • (2011) Int J Mol Med , vol.27 , Issue.3 , pp. 407-415
    • Huang, L.1    Zhang, Z.2    Zhang, S.3    Ren, J.4    Zhang, R.5    Zeng, H.6
  • 123
    • 84903150095 scopus 로고    scopus 로고
    • Celastrol inhibits the HIF-1alpha pathway by inhibition of mTOR/p70S6K/eIF4E and ERK1/2 phosphorylation in human hepatoma cells
    • Ma J, Han LZ, Liang H, Mi C, Shi H, Lee JJ, et al. Celastrol inhibits the HIF-1alpha pathway by inhibition of mTOR/p70S6K/eIF4E and ERK1/2 phosphorylation in human hepatoma cells. Oncol Rep (2014) 32(1):235-42. doi:10.3892/or.2014.3211
    • (2014) Oncol Rep , vol.32 , Issue.1 , pp. 235-242
    • Ma, J.1    Han, L.Z.2    Liang, H.3    Mi, C.4    Shi, H.5    Lee, J.J.6
  • 124
    • 77950234271 scopus 로고    scopus 로고
    • Celastrol suppresses angiogenesis-mediated tumor growth through inhibition of AKT/mammalian target of rapamycin pathway
    • Pang X, Yi Z, Zhang J, Lu B, Sung B, Qu W, et al. Celastrol suppresses angiogenesis-mediated tumor growth through inhibition of AKT/mammalian target of rapamycin pathway. Cancer Res (2010) 70(5):1951-9. doi:10.1158/0008-5472.CAN-09-3201
    • (2010) Cancer Res , vol.70 , Issue.5 , pp. 1951-1959
    • Pang, X.1    Yi, Z.2    Zhang, J.3    Lu, B.4    Sung, B.5    Qu, W.6
  • 125
    • 42649088411 scopus 로고    scopus 로고
    • Celastrol inhibits the growth of human glioma xenografts in nude mice through suppressing VEGFR expression
    • Huang Y, Zhou Y, Fan Y, Zhou D. Celastrol inhibits the growth of human glioma xenografts in nude mice through suppressing VEGFR expression. Cancer Lett (2008) 264(1):101-6. doi:10.1016/j.canlet.2008.01.043
    • (2008) Cancer Lett , vol.264 , Issue.1 , pp. 101-106
    • Huang, Y.1    Zhou, Y.2    Fan, Y.3    Zhou, D.4
  • 126
    • 70349554345 scopus 로고    scopus 로고
    • Antiangiogenic effect of celastrol on the growth of human glioma: an in vitro and in vivo study
    • Zhou YX, Huang YL. Antiangiogenic effect of celastrol on the growth of human glioma: an in vitro and in vivo study. Chin Med J (Engl) (2009) 122(14):1666-73
    • (2009) Chin Med J (Engl) , vol.122 , Issue.14 , pp. 1666-1673
    • Zhou, Y.X.1    Huang, Y.L.2
  • 127
    • 78149300933 scopus 로고    scopus 로고
    • Low-dose radiation augments vasculogenesis signaling through HIF-1-dependent and-independent SDF-1 induction
    • Lerman OZ, Greives MR, Singh SP, Thanik VD, Chang CC, Seiser N, et al. Low-dose radiation augments vasculogenesis signaling through HIF-1-dependent and-independent SDF-1 induction. Blood (2010) 116(18):3669-76. doi:10.1182/blood-2009-03-213629
    • (2010) Blood , vol.116 , Issue.18 , pp. 3669-3676
    • Lerman, O.Z.1    Greives, M.R.2    Singh, S.P.3    Thanik, V.D.4    Chang, C.C.5    Seiser, N.6
  • 128
    • 36749029349 scopus 로고    scopus 로고
    • Significance of HIF-1-active cells in angiogenesis and radioresistance
    • Harada H, Kizaka-Kondoh S, Li G, Itasaka S, Shibuya K, Inoue M, et al. Significance of HIF-1-active cells in angiogenesis and radioresistance. Oncogene (2007) 26(54):7508-16. doi:10.1038/sj.onc.1210556
    • (2007) Oncogene , vol.26 , Issue.54 , pp. 7508-7516
    • Harada, H.1    Kizaka-Kondoh, S.2    Li, G.3    Itasaka, S.4    Shibuya, K.5    Inoue, M.6
  • 129
    • 67649525966 scopus 로고    scopus 로고
    • Celastrol potentiates radiotherapy by impairment of DNA damage processing in human prostate cancer
    • Dai Y, DeSano JT, Meng Y, Ji Q, Ljungman M, Lawrence TS, et al. Celastrol potentiates radiotherapy by impairment of DNA damage processing in human prostate cancer. Int J Radiat Oncol Biol Phys (2009) 74(4):1217-25. doi:10.1016/j.ijrobp.2009.03.057
    • (2009) Int J Radiat Oncol Biol Phys , vol.74 , Issue.4 , pp. 1217-1225
    • Dai, Y.1    DeSano, J.T.2    Meng, Y.3    Ji, Q.4    Ljungman, M.5    Lawrence, T.S.6
  • 130
    • 79551598917 scopus 로고    scopus 로고
    • Enhancement of radiation sensitivity in lung cancer cells by celastrol is mediated by inhibition of Hsp90
    • Lee JH, Choi KJ, Seo WD, Jang SY, Kim M, Lee BW, et al. Enhancement of radiation sensitivity in lung cancer cells by celastrol is mediated by inhibition of Hsp90. Int J Mol Med (2011) 27(3):441-6. doi:10.3892/ijmm.2011.601
    • (2011) Int J Mol Med , vol.27 , Issue.3 , pp. 441-446
    • Lee, J.H.1    Choi, K.J.2    Seo, W.D.3    Jang, S.Y.4    Kim, M.5    Lee, B.W.6
  • 131
    • 79959933122 scopus 로고    scopus 로고
    • Radiosensitization by celastrol is mediated by modification of antioxidant thiol molecules
    • Seo HR, Seo WD, Pyun BJ, Lee BW, Jin YB, Park KH, et al. Radiosensitization by celastrol is mediated by modification of antioxidant thiol molecules. Chem Biol Interact (2011) 193(1):34-42. doi:10.1016/j.cbi.2011.04.009
    • (2011) Chem Biol Interact , vol.193 , Issue.1 , pp. 34-42
    • Seo, H.R.1    Seo, W.D.2    Pyun, B.J.3    Lee, B.W.4    Jin, Y.B.5    Park, K.H.6
  • 132
    • 84936986395 scopus 로고    scopus 로고
    • Celastrol induces proteasomal degradation of FANCD2 to sensitize lung cancer cells to DNA crosslinking agents
    • Wang GZ, Liu YQ, Cheng X, Zhou GB. Celastrol induces proteasomal degradation of FANCD2 to sensitize lung cancer cells to DNA crosslinking agents. Cancer Sci (2015) 106(7):902-8. doi:10.1111/cas.12679
    • (2015) Cancer Sci , vol.106 , Issue.7 , pp. 902-908
    • Wang, G.Z.1    Liu, Y.Q.2    Cheng, X.3    Zhou, G.B.4
  • 133
    • 84924860249 scopus 로고    scopus 로고
    • ATF2 contributes to cisplatin resistance in non-small cell lung cancer and celastrol induces cisplatin resensitization through inhibition of JNK/ATF2 pathway
    • Lo Iacono M, Monica V, Vavala T, Gisabella M, Saviozzi S, Bracco E, et al. ATF2 contributes to cisplatin resistance in non-small cell lung cancer and celastrol induces cisplatin resensitization through inhibition of JNK/ATF2 pathway. Int J Cancer (2015) 136(11):2598-609. doi:10.1002/ijc.29302
    • (2015) Int J Cancer , vol.136 , Issue.11 , pp. 2598-2609
    • Lo Iacono, M.1    Monica, V.2    Vavala, T.3    Gisabella, M.4    Saviozzi, S.5    Bracco, E.6
  • 134
    • 76949108486 scopus 로고    scopus 로고
    • Celastrol and an EGCG pro-drug exhibit potent chemosensitizing activity in human leukemia cells
    • Davenport A, Frezza M, Shen M, Ge Y, Huo C, Chan TH, et al. Celastrol and an EGCG pro-drug exhibit potent chemosensitizing activity in human leukemia cells. Int J Mol Med (2010) 25(3):465-70
    • (2010) Int J Mol Med , vol.25 , Issue.3 , pp. 465-470
    • Davenport, A.1    Frezza, M.2    Shen, M.3    Ge, Y.4    Huo, C.5    Chan, T.H.6
  • 135
    • 77649178833 scopus 로고    scopus 로고
    • Celastrol, a novel HSP90 inhibitor, depletes Bcr-Abl and induces apoptosis in imatinib-resistant chronic myelogenous leukemia cells harboring T315I mutation
    • Lu Z, Jin Y, Qiu L, Lai Y, Pan J. Celastrol, a novel HSP90 inhibitor, depletes Bcr-Abl and induces apoptosis in imatinib-resistant chronic myelogenous leukemia cells harboring T315I mutation. Cancer Lett (2010) 290(2):182-91. doi:10.1016/j.canlet.2009.09.006
    • (2010) Cancer Lett , vol.290 , Issue.2 , pp. 182-191
    • Lu, Z.1    Jin, Y.2    Qiu, L.3    Lai, Y.4    Pan, J.5
  • 136
    • 73849110612 scopus 로고    scopus 로고
    • Celastrol synergistically enhances temozolomide cytotoxicity in melanoma cells
    • Chen M, Rose AE, Doudican N, Osman I, Orlow SJ. Celastrol synergistically enhances temozolomide cytotoxicity in melanoma cells. Mol Cancer Res (2009) 7(12):1946-53. doi:10.1158/1541-7786.MCR-09-0243
    • (2009) Mol Cancer Res , vol.7 , Issue.12 , pp. 1946-1953
    • Chen, M.1    Rose, A.E.2    Doudican, N.3    Osman, I.4    Orlow, S.J.5
  • 137
    • 84896914147 scopus 로고    scopus 로고
    • Celastrol induces apoptosis in gefitinib-resistant non-small cell lung cancer cells via caspases-dependent pathways and Hsp90 client protein degradation
    • Fan XX, Li N, Wu JL, Zhou YL, He JX, Liu L, et al. Celastrol induces apoptosis in gefitinib-resistant non-small cell lung cancer cells via caspases-dependent pathways and Hsp90 client protein degradation. Molecules (2014) 19(3):3508-22. doi:10.3390/molecules19033508
    • (2014) Molecules , vol.19 , Issue.3 , pp. 3508-3522
    • Fan, X.X.1    Li, N.2    Wu, J.L.3    Zhou, Y.L.4    He, J.X.5    Liu, L.6
  • 138
    • 84879411020 scopus 로고    scopus 로고
    • Celastrol exerts synergistic effects with PHA-665752 and inhibits tumor growth of c-Met-deficient hepatocellular carcinoma in vivo
    • Jiang HL, Jin JZ, Wu D, Xu D, Lin GF, Yu H, et al. Celastrol exerts synergistic effects with PHA-665752 and inhibits tumor growth of c-Met-deficient hepatocellular carcinoma in vivo. Mol Biol Rep (2013) 40(7):4203-9. doi:10.1007/s11033-013-2501-y
    • (2013) Mol Biol Rep , vol.40 , Issue.7 , pp. 4203-4209
    • Jiang, H.L.1    Jin, J.Z.2    Wu, D.3    Xu, D.4    Lin, G.F.5    Yu, H.6
  • 139
    • 84871424648 scopus 로고    scopus 로고
    • Upregulating Noxa by ER stress, celastrol exerts synergistic anti-cancer activity in combination with ABT-737 in human hepatocellular carcinoma cells
    • Zhu H, Yang W, He LJ, Ding WJ, Zheng L, Liao SD, et al. Upregulating Noxa by ER stress, celastrol exerts synergistic anti-cancer activity in combination with ABT-737 in human hepatocellular carcinoma cells. PLoS One (2012) 7(12):e52333. doi:10.1371/journal.pone.0052333
    • (2012) PLoS One , vol.7 , Issue.12
    • Zhu, H.1    Yang, W.2    He, L.J.3    Ding, W.J.4    Zheng, L.5    Liao, S.D.6
  • 140
    • 33947598693 scopus 로고    scopus 로고
    • Celastrol, a novel triterpene, potentiates TNF-induced apoptosis and suppresses invasion of tumor cells by inhibiting NF-kappaB-regulated gene products and TAK1-mediated NF-kappaB activation
    • Sethi G, Ahn KS, Pandey MK, Aggarwal BB. Celastrol, a novel triterpene, potentiates TNF-induced apoptosis and suppresses invasion of tumor cells by inhibiting NF-kappaB-regulated gene products and TAK1-mediated NF-kappaB activation. Blood (2007) 109(7):2727-35. doi:10.1182/blood-2006-10-050807
    • (2007) Blood , vol.109 , Issue.7 , pp. 2727-2735
    • Sethi, G.1    Ahn, K.S.2    Pandey, M.K.3    Aggarwal, B.B.4
  • 141
    • 80052081077 scopus 로고    scopus 로고
    • Celastrol inhibits breast cancer cell invasion via suppression of NF-kB-mediated matrix metalloproteinase-9 expression
    • Kim Y, Kang H, Jang SW, Ko J. Celastrol inhibits breast cancer cell invasion via suppression of NF-kB-mediated matrix metalloproteinase-9 expression. Cell Physiol Biochem (2011) 28(2):175-84. doi:10.1159/000331729
    • (2011) Cell Physiol Biochem , vol.28 , Issue.2 , pp. 175-184
    • Kim, Y.1    Kang, H.2    Jang, S.W.3    Ko, J.4
  • 142
    • 84914691713 scopus 로고    scopus 로고
    • Celastrol induces the apoptosis of breast cancer cells and inhibits their invasion via downregulation of MMP-9
    • Mi C, Shi H, Ma J, Han LZ, Lee JJ, Jin X. Celastrol induces the apoptosis of breast cancer cells and inhibits their invasion via downregulation of MMP-9. Oncol Rep (2014) 32(6):2527-32. doi:10.3892/or.2014.3535
    • (2014) Oncol Rep , vol.32 , Issue.6 , pp. 2527-2532
    • Mi, C.1    Shi, H.2    Ma, J.3    Han, L.Z.4    Lee, J.J.5    Jin, X.6
  • 143
    • 84885589594 scopus 로고    scopus 로고
    • miR-224 is critical for celastrol-induced inhibition of migration and invasion of hepatocellular carcinoma cells
    • Li H, Li Y, Liu D, Sun H, Liu J. miR-224 is critical for celastrol-induced inhibition of migration and invasion of hepatocellular carcinoma cells. Cell Physiol Biochem (2013) 32(2):448-58. doi:10.1159/000354450
    • (2013) Cell Physiol Biochem , vol.32 , Issue.2 , pp. 448-458
    • Li, H.1    Li, Y.2    Liu, D.3    Sun, H.4    Liu, J.5
  • 144
    • 84941010663 scopus 로고    scopus 로고
    • [Effect of celastrol in inhibiting metastasis of lung cancer cells by influencing Akt signaling pathway and expressing integrins]
    • Xu J, Wu CL, Huang J. [Effect of celastrol in inhibiting metastasis of lung cancer cells by influencing Akt signaling pathway and expressing integrins]. Zhongguo Zhong Yao Za Zhi (2015) 40(6):1129-33
    • (2015) Zhongguo Zhong Yao Za Zhi , vol.40 , Issue.6 , pp. 1129-1133
    • Xu, J.1    Wu, C.L.2    Huang, J.3
  • 145
    • 84941085066 scopus 로고    scopus 로고
    • [Anti-metastasis of celastrol on esophageal cancer cells and its mechanism]
    • Xu J, Wu CL. [Anti-metastasis of celastrol on esophageal cancer cells and its mechanism]. Sheng Li Xue Bao (2015) 67(3):341-7
    • (2015) Sheng Li Xue Bao , vol.67 , Issue.3 , pp. 341-347
    • Xu, J.1    Wu, C.L.2
  • 146
    • 77954917015 scopus 로고    scopus 로고
    • Celastrol acts as a potent antimetastatic agent targeting beta1 integrin and inhibiting cell-extracellular matrix adhesion, in part via the p38 mitogen-activated protein kinase pathway
    • Zhu H, Liu XW, Cai TY, Cao J, Tu CX, Lu W, et al. Celastrol acts as a potent antimetastatic agent targeting beta1 integrin and inhibiting cell-extracellular matrix adhesion, in part via the p38 mitogen-activated protein kinase pathway. J Pharmacol Exp Ther (2010) 334(2):489-99. doi:10.1124/jpet.110.165654
    • (2010) J Pharmacol Exp Ther , vol.334 , Issue.2 , pp. 489-499
    • Zhu, H.1    Liu, X.W.2    Cai, T.Y.3    Cao, J.4    Tu, C.X.5    Lu, W.6
  • 147
    • 78651319942 scopus 로고    scopus 로고
    • Celastrol suppresses invasion of colon and pancreatic cancer cells through the downregulation of expression of CXCR4 chemokine receptor
    • Yadav VR, Sung B, Prasad S, Kannappan R, Cho SG, Liu M, et al. Celastrol suppresses invasion of colon and pancreatic cancer cells through the downregulation of expression of CXCR4 chemokine receptor. J Mol Med (Berl) (2010) 88(12):1243-53. doi:10.1007/s00109-010-0669-3
    • (2010) J Mol Med (Berl) , vol.88 , Issue.12 , pp. 1243-1253
    • Yadav, V.R.1    Sung, B.2    Prasad, S.3    Kannappan, R.4    Cho, S.G.5    Liu, M.6
  • 148
    • 68849116745 scopus 로고    scopus 로고
    • Pharmacologic inhibitors of IkappaB kinase suppress growth and migration of mammary carcinosarcoma cells in vitro and prevent osteolytic bone metastasis in vivo
    • Idris AI, Libouban H, Nyangoga H, Landao-Bassonga E, Chappard D, Ralston SH. Pharmacologic inhibitors of IkappaB kinase suppress growth and migration of mammary carcinosarcoma cells in vitro and prevent osteolytic bone metastasis in vivo. Mol Cancer Ther (2009) 8(8):2339-47. doi:10.1158/1535-7163.MCT-09-0133
    • (2009) Mol Cancer Ther , vol.8 , Issue.8 , pp. 2339-2347
    • Idris, A.I.1    Libouban, H.2    Nyangoga, H.3    Landao-Bassonga, E.4    Chappard, D.5    Ralston, S.H.6
  • 149
    • 23844529468 scopus 로고    scopus 로고
    • Celastrol protects against MPTP-and 3-nitropropionic acid-induced neurotoxicity
    • Cleren C, Calingasan NY, Chen J, Beal MF. Celastrol protects against MPTP-and 3-nitropropionic acid-induced neurotoxicity. J Neurochem (2005) 94(4):995-1004. doi:10.1111/j.1471-4159.2005.03253.x
    • (2005) J Neurochem , vol.94 , Issue.4 , pp. 995-1004
    • Cleren, C.1    Calingasan, N.Y.2    Chen, J.3    Beal, M.F.4
  • 150
    • 12944335007 scopus 로고    scopus 로고
    • Reversal of a full-length mutant huntingtin neuronal cell phenotype by chemical inhibitors of polyglutamine-mediated aggregation
    • Wang J, Gines S, MacDonald ME, Gusella JF. Reversal of a full-length mutant huntingtin neuronal cell phenotype by chemical inhibitors of polyglutamine-mediated aggregation. BMC Neurosci (2005) 6:1. doi:10.1186/1471-2202-6-1
    • (2005) BMC Neurosci , vol.6 , pp. 1
    • Wang, J.1    Gines, S.2    MacDonald, M.E.3    Gusella, J.F.4
  • 151
    • 36849044616 scopus 로고    scopus 로고
    • Celastrol inhibits polyglutamine aggregation and toxicity though induction of the heat shock response
    • Zhang YQ, Sarge KD. Celastrol inhibits polyglutamine aggregation and toxicity though induction of the heat shock response. J Mol Med (Berl) (2007) 85(12):1421-8. doi:10.1007/s00109-007-0251-9
    • (2007) J Mol Med (Berl) , vol.85 , Issue.12 , pp. 1421-1428
    • Zhang, Y.Q.1    Sarge, K.D.2
  • 152
    • 77950377674 scopus 로고    scopus 로고
    • Reduction of beta-amyloid pathology by celastrol in a transgenic mouse model of Alzheimer's disease
    • Paris D, Ganey NJ, Laporte V, Patel NS, Beaulieu-Abdelahad D, Bachmeier C, et al. Reduction of beta-amyloid pathology by celastrol in a transgenic mouse model of Alzheimer's disease. J Neuroinflammation (2010) 7:17. doi:10.1186/1742-2094-7-17
    • (2010) J Neuroinflammation , vol.7 , pp. 17
    • Paris, D.1    Ganey, N.J.2    Laporte, V.3    Patel, N.S.4    Beaulieu-Abdelahad, D.5    Bachmeier, C.6
  • 153
    • 33744486642 scopus 로고    scopus 로고
    • Celastrol blocks neuronal cell death and extends life in transgenic mouse model of amyotrophic lateral sclerosis
    • Kiaei M, Kipiani K, Petri S, Chen J, Calingasan NY, Beal MF. Celastrol blocks neuronal cell death and extends life in transgenic mouse model of amyotrophic lateral sclerosis. Neurodegener Dis (2005) 2(5):246-54. doi:10.1159/000090364
    • (2005) Neurodegener Dis , vol.2 , Issue.5 , pp. 246-254
    • Kiaei, M.1    Kipiani, K.2    Petri, S.3    Chen, J.4    Calingasan, N.Y.5    Beal, M.F.6
  • 154
    • 84939563275 scopus 로고    scopus 로고
    • Celastrol ameliorates EAE induction by suppressing pathogenic T cell responses in the peripheral and central nervous systems
    • Wang Y, Cao L, Xu LM, Cao FF, Peng B, Zhang X, et al. Celastrol ameliorates EAE induction by suppressing pathogenic T cell responses in the peripheral and central nervous systems. J Neuroimmune Pharmacol (2015) 10(3):506-16. doi:10.1007/s11481-015-9598-9
    • (2015) J Neuroimmune Pharmacol , vol.10 , Issue.3 , pp. 506-516
    • Wang, Y.1    Cao, L.2    Xu, L.M.3    Cao, F.F.4    Peng, B.5    Zhang, X.6
  • 155
    • 84939569034 scopus 로고    scopus 로고
    • Modulatory effect of celastrol on Th1/Th2 cytokines profile, TLR2 and CD3+ T-lymphocyte expression in a relapsing-remitting model of multiple sclerosis in rats
    • Abdin AA, Hasby EA. Modulatory effect of celastrol on Th1/Th2 cytokines profile, TLR2 and CD3+ T-lymphocyte expression in a relapsing-remitting model of multiple sclerosis in rats. Eur J Pharmacol (2014) 742:102-12. doi:10.1016/j.ejphar.2014.09.001
    • (2014) Eur J Pharmacol , vol.742 , pp. 102-112
    • Abdin, A.A.1    Hasby, E.A.2
  • 156
    • 7944233158 scopus 로고    scopus 로고
    • Cell biology of protein misfolding: the examples of Alzheimer's and Parkinson's diseases
    • Selkoe DJ. Cell biology of protein misfolding: the examples of Alzheimer's and Parkinson's diseases. Nat Cell Biol (2004) 6(11):1054-61. doi:10.1038/ncb1104-1054
    • (2004) Nat Cell Biol , vol.6 , Issue.11 , pp. 1054-1061
    • Selkoe, D.J.1
  • 157
    • 7044238416 scopus 로고    scopus 로고
    • Neurodegenerative diseases: a decade of discoveries paves the way for therapeutic breakthroughs
    • Forman MS, Trojanowski JQ, Lee VM. Neurodegenerative diseases: a decade of discoveries paves the way for therapeutic breakthroughs. Nat Med (2004) 10(10):1055-63. doi:10.1038/nm1113
    • (2004) Nat Med , vol.10 , Issue.10 , pp. 1055-1063
    • Forman, M.S.1    Trojanowski, J.Q.2    Lee, V.M.3
  • 158
    • 11144243412 scopus 로고    scopus 로고
    • Modulation of neurodegeneration by molecular chaperones
    • Muchowski PJ, Wacker JL. Modulation of neurodegeneration by molecular chaperones. Nat Rev Neurosci (2005) 6(1):11-22. doi:10.1038/nrn1587
    • (2005) Nat Rev Neurosci , vol.6 , Issue.1 , pp. 11-22
    • Muchowski, P.J.1    Wacker, J.L.2
  • 160
    • 35348894945 scopus 로고    scopus 로고
    • Induction of heat shock proteins in differentiated human and rodent neurons by celastrol
    • Chow AM, Brown IR. Induction of heat shock proteins in differentiated human and rodent neurons by celastrol. Cell Stress Chaperones (2007) 12(3):237-44. doi:10.1379/CSC-269.1
    • (2007) Cell Stress Chaperones , vol.12 , Issue.3 , pp. 237-244
    • Chow, A.M.1    Brown, I.R.2
  • 161
    • 2942650958 scopus 로고    scopus 로고
    • Hsp70 promotes TNF-mediated apoptosis by binding IKK gamma and impairing NF-kappa B survival signaling
    • Ran R, Lu A, Zhang L, Tang Y, Zhu H, Xu H, et al. Hsp70 promotes TNF-mediated apoptosis by binding IKK gamma and impairing NF-kappa B survival signaling. Genes Dev (2004) 18(12):1466-81. doi:10.1101/gad.1188204
    • (2004) Genes Dev , vol.18 , Issue.12 , pp. 1466-1481
    • Ran, R.1    Lu, A.2    Zhang, L.3    Tang, Y.4    Zhu, H.5    Xu, H.6
  • 162
    • 34548292474 scopus 로고    scopus 로고
    • Enhanced heat shock protein 70 expression alters proteasomal degradation of IkappaB kinase in experimental acute respiratory distress syndrome
    • Weiss YG, Bromberg Z, Raj N, Raphael J, Goloubinoff P, Ben-Neriah Y, et al. Enhanced heat shock protein 70 expression alters proteasomal degradation of IkappaB kinase in experimental acute respiratory distress syndrome. Crit Care Med (2007) 35(9):2128-38. doi:10.1097/01.CCM.0000278915.78030.74
    • (2007) Crit Care Med , vol.35 , Issue.9 , pp. 2128-2138
    • Weiss, Y.G.1    Bromberg, Z.2    Raj, N.3    Raphael, J.4    Goloubinoff, P.5    Ben-Neriah, Y.6
  • 163
    • 33646580045 scopus 로고    scopus 로고
    • Hsp70 inhibits lipopolysaccharide-induced NF-kappaB activation by interacting with TRAF6 and inhibiting its ubiquitination
    • Chen H, Wu Y, Zhang Y, Jin L, Luo L, Xue B, et al. Hsp70 inhibits lipopolysaccharide-induced NF-kappaB activation by interacting with TRAF6 and inhibiting its ubiquitination. FEBS Lett (2006) 580(13):3145-52. doi:10.1016/j.febslet.2006.04.066
    • (2006) FEBS Lett , vol.580 , Issue.13 , pp. 3145-3152
    • Chen, H.1    Wu, Y.2    Zhang, Y.3    Jin, L.4    Luo, L.5    Xue, B.6
  • 164
    • 84892670789 scopus 로고    scopus 로고
    • Celastrol from 'Thunder God Vine' protects SH-SY5Y cells through the preservation of mitochondrial function and inhibition of p38 MAPK in a rotenone model of Parkinson's disease
    • Choi BS, Kim H, Lee HJ, Sapkota K, Park SE, Kim S, et al. Celastrol from 'Thunder God Vine' protects SH-SY5Y cells through the preservation of mitochondrial function and inhibition of p38 MAPK in a rotenone model of Parkinson's disease. Neurochem Res (2014) 39(1):84-96. doi:10.1007/s11064-013-1193-y
    • (2014) Neurochem Res , vol.39 , Issue.1 , pp. 84-96
    • Choi, B.S.1    Kim, H.2    Lee, H.J.3    Sapkota, K.4    Park, S.E.5    Kim, S.6
  • 165
    • 84891835356 scopus 로고    scopus 로고
    • Celastrol prevents cadmium-induced neuronal cell death via targeting JNK and PTEN-Akt/mTOR network
    • Chen S, Gu C, Xu C, Zhang J, Xu Y, Ren Q, et al. Celastrol prevents cadmium-induced neuronal cell death via targeting JNK and PTEN-Akt/mTOR network. J Neurochem (2014) 128(2):256-66. doi:10.1111/jnc.12474
    • (2014) J Neurochem , vol.128 , Issue.2 , pp. 256-266
    • Chen, S.1    Gu, C.2    Xu, C.3    Zhang, J.4    Xu, Y.5    Ren, Q.6
  • 166
    • 84907597951 scopus 로고    scopus 로고
    • Lack of neuroprotective effect of celastrol under conditions of proteasome inhibition by lactacystin in in vitro and in vivo studies: implications for Parkinson's disease
    • Konieczny J, Jantas D, Lenda T, Domin H, Czarnecka A, Kuter K, et al. Lack of neuroprotective effect of celastrol under conditions of proteasome inhibition by lactacystin in in vitro and in vivo studies: implications for Parkinson's disease. Neurotox Res (2014) 26(3):255-73. doi:10.1007/s12640-014-9477-9
    • (2014) Neurotox Res , vol.26 , Issue.3 , pp. 255-273
    • Konieczny, J.1    Jantas, D.2    Lenda, T.3    Domin, H.4    Czarnecka, A.5    Kuter, K.6
  • 167
    • 84878373373 scopus 로고    scopus 로고
    • The extent of neurodegeneration and neuroprotection in two chemical in vitro models related to Parkinson's disease is critically dependent on cell culture conditions
    • Jantas D, Roman A, Kusmierczyk J, Lorenc-Koci E, Konieczny J, Lenda T, et al. The extent of neurodegeneration and neuroprotection in two chemical in vitro models related to Parkinson's disease is critically dependent on cell culture conditions. Neurotox Res (2013) 24(1):41-54. doi:10.1007/s12640-012-9374-z
    • (2013) Neurotox Res , vol.24 , Issue.1 , pp. 41-54
    • Jantas, D.1    Roman, A.2    Kusmierczyk, J.3    Lorenc-Koci, E.4    Konieczny, J.5    Lenda, T.6
  • 168
    • 84878831944 scopus 로고    scopus 로고
    • Induction of heat shock proteins in cerebral cortical cultures by celastrol
    • Chow AM, Tang DW, Hanif A, Brown IR. Induction of heat shock proteins in cerebral cortical cultures by celastrol. Cell Stress Chaperones (2013) 18(2):155-60. doi:10.1007/s12192-012-0364-0
    • (2013) Cell Stress Chaperones , vol.18 , Issue.2 , pp. 155-160
    • Chow, A.M.1    Tang, D.W.2    Hanif, A.3    Brown, I.R.4
  • 169
    • 84897350282 scopus 로고    scopus 로고
    • Localization of heat shock proteins in cerebral cortical cultures following induction by celastrol
    • Chow AM, Tang DW, Hanif A, Brown IR. Localization of heat shock proteins in cerebral cortical cultures following induction by celastrol. Cell Stress Chaperones (2014) 19(6):845-51. doi:10.1007/s12192-014-0508-5
    • (2014) Cell Stress Chaperones , vol.19 , Issue.6 , pp. 845-851
    • Chow, A.M.1    Tang, D.W.2    Hanif, A.3    Brown, I.R.4
  • 170
    • 84907975871 scopus 로고    scopus 로고
    • Celastrol enhances cell viability and inhibits amyloid-beta production induced by lipopolysaccharide in vitro
    • Zhao Y, Zhao H, Lobo N, Guo X, Gentleman SM, Ma D. Celastrol enhances cell viability and inhibits amyloid-beta production induced by lipopolysaccharide in vitro. J Alzheimers Dis (2014) 41(3):835-44. doi:10.3233/JAD-131799
    • (2014) J Alzheimers Dis , vol.41 , Issue.3 , pp. 835-844
    • Zhao, Y.1    Zhao, H.2    Lobo, N.3    Guo, X.4    Gentleman, S.M.5    Ma, D.6
  • 171
    • 0026702955 scopus 로고
    • Microglial-produced nitric oxide and reactive nitrogen oxides mediate neuronal cell death
    • Boje KM, Arora PK. Microglial-produced nitric oxide and reactive nitrogen oxides mediate neuronal cell death. Brain Res (1992) 587(2):250-6. doi:10.1016/0006-8993(92)91004-X
    • (1992) Brain Res , vol.587 , Issue.2 , pp. 250-256
    • Boje, K.M.1    Arora, P.K.2
  • 172
    • 33748447845 scopus 로고    scopus 로고
    • Microglia, major player in the brain inflammation: their roles in the pathogenesis of Parkinson's disease
    • Kim YS, Joh TH. Microglia, major player in the brain inflammation: their roles in the pathogenesis of Parkinson's disease. Exp Mol Med (2006) 38(4):333-47. doi:10.1038/emm.2006.40
    • (2006) Exp Mol Med , vol.38 , Issue.4 , pp. 333-347
    • Kim, Y.S.1    Joh, T.H.2
  • 173
    • 38049121045 scopus 로고    scopus 로고
    • Celastrol inhibits production of nitric oxide and proinflammatory cytokines through MAPK signal transduction and NF-kappaB in LPS-stimulated BV-2 microglial cells
    • Jung HW, Chung YS, Kim YS, Park YK. Celastrol inhibits production of nitric oxide and proinflammatory cytokines through MAPK signal transduction and NF-kappaB in LPS-stimulated BV-2 microglial cells. Exp Mol Med (2007) 39(6):715-21. doi:10.1038/emm.2007.78
    • (2007) Exp Mol Med , vol.39 , Issue.6 , pp. 715-721
    • Jung, H.W.1    Chung, Y.S.2    Kim, Y.S.3    Park, Y.K.4
  • 174
    • 84935098094 scopus 로고    scopus 로고
    • Mitochondrial dysfunction as a central event for mechanisms underlying insulin resistance: the roles of long chain fatty acids
    • Hafizi Abu Bakar M, Kian Kai C, Wan Hassan WN, Sarmidi MR, Yaakob H, Zaman Huri H. Mitochondrial dysfunction as a central event for mechanisms underlying insulin resistance: the roles of long chain fatty acids. Diabetes Metab Res Rev (2015) 31(5):453-75. doi:10.1002/dmrr.2601
    • (2015) Diabetes Metab Res Rev , vol.31 , Issue.5 , pp. 453-475
    • Hafizi Abu Bakar, M.1    Kian Kai, C.2    Wan Hassan, W.N.3    Sarmidi, M.R.4    Yaakob, H.5    Zaman Huri, H.6
  • 175
    • 84929208510 scopus 로고    scopus 로고
    • Celastrol protects against antimycin A-induced insulin resistance in human skeletal muscle cells
    • Abu Bakar MH, Cheng KK, Sarmidi MR, Yaakob H, Huri HZ. Celastrol protects against antimycin A-induced insulin resistance in human skeletal muscle cells. Molecules (2015) 20(5):8242-69. doi:10.3390/molecules20058242
    • (2015) Molecules , vol.20 , Issue.5 , pp. 8242-8269
    • Abu Bakar, M.H.1    Cheng, K.K.2    Sarmidi, M.R.3    Yaakob, H.4    Huri, H.Z.5
  • 176
    • 84876817513 scopus 로고    scopus 로고
    • Celastrol, an NF-kappaB inhibitor, improves insulin resistance and attenuates renal injury in db/db mice
    • Kim JE, Lee MH, Nam DH, Song HK, Kang YS, Lee JE, et al. Celastrol, an NF-kappaB inhibitor, improves insulin resistance and attenuates renal injury in db/db mice. PLoS One (2013) 8(4):e62068. doi:10.1371/journal.pone.0062068
    • (2013) PLoS One , vol.8 , Issue.4
    • Kim, J.E.1    Lee, M.H.2    Nam, D.H.3    Song, H.K.4    Kang, Y.S.5    Lee, J.E.6
  • 177
    • 84958781237 scopus 로고    scopus 로고
    • Protective effects of celastrol on diabetic liver injury via TLR4/MyD88/NF-kappaB signaling pathway in type 2 diabetic rats
    • Han LP, Li CJ, Sun B, Xie Y, Guan Y, Ma ZJ, et al. Protective effects of celastrol on diabetic liver injury via TLR4/MyD88/NF-kappaB signaling pathway in type 2 diabetic rats. J Diabetes Res (2016) 2016:2641248. doi:10.1155/2016/2641248
    • (2016) J Diabetes Res , vol.2016
    • Han, L.P.1    Li, C.J.2    Sun, B.3    Xie, Y.4    Guan, Y.5    Ma, Z.J.6
  • 178
    • 84963533468 scopus 로고    scopus 로고
    • Celastrol attenuates oxidative stress in the skeletal muscle of diabetic rats by regulating the AMPK-PGC1alpha-SIRT3 signaling pathway
    • Guan Y, Cui ZJ, Sun B, Han LP, Li CJ, Chen LM. Celastrol attenuates oxidative stress in the skeletal muscle of diabetic rats by regulating the AMPK-PGC1alpha-SIRT3 signaling pathway. Int J Mol Med (2016) 37(5):1229-38. doi:10.3892/ijmm.2016.2549
    • (2016) Int J Mol Med , vol.37 , Issue.5 , pp. 1229-1238
    • Guan, Y.1    Cui, Z.J.2    Sun, B.3    Han, L.P.4    Li, C.J.5    Chen, L.M.6
  • 180
    • 84867524649 scopus 로고    scopus 로고
    • Oral bioavailability and gender-related pharmacokinetics of celastrol following administration of pure celastrol and its related tablets in rats
    • Zhang J, Li CY, Xu MJ, Wu T, Chu JH, Liu SJ, et al. Oral bioavailability and gender-related pharmacokinetics of celastrol following administration of pure celastrol and its related tablets in rats. J Ethnopharmacol (2012) 144(1):195-200. doi:10.1016/j.jep.2012.09.005
    • (2012) J Ethnopharmacol , vol.144 , Issue.1 , pp. 195-200
    • Zhang, J.1    Li, C.Y.2    Xu, M.J.3    Wu, T.4    Chu, J.H.5    Liu, S.J.6
  • 181
    • 84928265883 scopus 로고    scopus 로고
    • Celastrol ameliorates cytokine toxicity and pro-inflammatory immune responses by suppressing NF-kappaB activation in RINm5F beta cells
    • Ju SM, Youn GS, Cho YS, Choi SY, Park J. Celastrol ameliorates cytokine toxicity and pro-inflammatory immune responses by suppressing NF-kappaB activation in RINm5F beta cells. BMB Rep (2015) 48(3):172-7. doi:10.5483/BMBRep.2015.48.3.147
    • (2015) BMB Rep , vol.48 , Issue.3 , pp. 172-177
    • Ju, S.M.1    Youn, G.S.2    Cho, Y.S.3    Choi, S.Y.4    Park, J.5
  • 183
  • 184
    • 84943455845 scopus 로고    scopus 로고
    • Celastrol protects against obesity and metabolic dysfunction through activation of a HSF1-PGC1alpha transcriptional axis
    • Ma X, Xu L, Alberobello AT, Gavrilova O, Bagattin A, Skarulis M, et al. Celastrol protects against obesity and metabolic dysfunction through activation of a HSF1-PGC1alpha transcriptional axis. Cell Metab (2015) 22(4):695-708. doi:10.1016/j.cmet.2015.08.005
    • (2015) Cell Metab , vol.22 , Issue.4 , pp. 695-708
    • Ma, X.1    Xu, L.2    Alberobello, A.T.3    Gavrilova, O.4    Bagattin, A.5    Skarulis, M.6
  • 185
    • 84905654464 scopus 로고    scopus 로고
    • Celastrol may have an anti-atherosclerosis effect in a rabbit experimental carotid atherosclerosis model
    • Zhu F, Li C, Jin XP, Weng SX, Fan LL, Zheng Z, et al. Celastrol may have an anti-atherosclerosis effect in a rabbit experimental carotid atherosclerosis model. Int J Clin Exp Med (2014) 7(7):1684-91
    • (2014) Int J Clin Exp Med , vol.7 , Issue.7 , pp. 1684-1691
    • Zhu, F.1    Li, C.2    Jin, X.P.3    Weng, S.X.4    Fan, L.L.5    Zheng, Z.6
  • 186
    • 84879184415 scopus 로고    scopus 로고
    • Celastrol prevents atherosclerosis via inhibiting LOX-1 and oxidative stress
    • Gu L, Bai W, Li S, Zhang Y, Han Y, Gu Y, et al. Celastrol prevents atherosclerosis via inhibiting LOX-1 and oxidative stress. PLoS One (2013) 8(6):e65477. doi:10.1371/journal.pone.0065477
    • (2013) PLoS One , vol.8 , Issue.6
    • Gu, L.1    Bai, W.2    Li, S.3    Zhang, Y.4    Han, Y.5    Gu, Y.6
  • 187
    • 84942279745 scopus 로고    scopus 로고
    • Tripterine treatment improves endothelial progenitor cell function via integrin-linked kinase
    • Lu C, Yu X, Zuo K, Zhang X, Cao C, Xu J, et al. Tripterine treatment improves endothelial progenitor cell function via integrin-linked kinase. Cell Physiol Biochem (2015) 37(3):1089-103. doi:10.1159/000430234
    • (2015) Cell Physiol Biochem , vol.37 , Issue.3 , pp. 1089-1103
    • Lu, C.1    Yu, X.2    Zuo, K.3    Zhang, X.4    Cao, C.5    Xu, J.6
  • 188
    • 84921647796 scopus 로고    scopus 로고
    • Short time tripterine treatment enhances endothelial progenitor cell function via heat shock protein 32
    • Lu C, Zhang X, Zhang D, Pei E, Xu J, Tang T, et al. Short time tripterine treatment enhances endothelial progenitor cell function via heat shock protein 32. J Cell Physiol (2015) 230(5):1139-47. doi:10.1002/jcp.24849
    • (2015) J Cell Physiol , vol.230 , Issue.5 , pp. 1139-1147
    • Lu, C.1    Zhang, X.2    Zhang, D.3    Pei, E.4    Xu, J.5    Tang, T.6
  • 189
    • 33846183562 scopus 로고    scopus 로고
    • Aminoglycoside-induced ototoxicity
    • Selimoglu E. Aminoglycoside-induced ototoxicity. Curr Pharm Des (2007) 13(1):119-26. doi:10.2174/138161207779313731
    • (2007) Curr Pharm Des , vol.13 , Issue.1 , pp. 119-126
    • Selimoglu, E.1
  • 190
    • 34948879664 scopus 로고    scopus 로고
    • Ototoxicity
    • Rybak LP, Ramkumar V. Ototoxicity. Kidney Int (2007) 72(8):931-5. doi:10.1038/sj.ki.5002434
    • (2007) Kidney Int , vol.72 , Issue.8 , pp. 931-935
    • Rybak, L.P.1    Ramkumar, V.2
  • 192
    • 33646158677 scopus 로고    scopus 로고
    • Heme oxygenase-1 attenuates the cisplatin-induced apoptosis of auditory cells via down-regulation of reactive oxygen species generation
    • Kim HJ, So HS, Lee JH, Lee JH, Park C, Park SY, et al. Heme oxygenase-1 attenuates the cisplatin-induced apoptosis of auditory cells via down-regulation of reactive oxygen species generation. Free Radic Biol Med (2006) 40(10):1810-9. doi:10.1016/j.freeradbiomed.2006.01.018
    • (2006) Free Radic Biol Med , vol.40 , Issue.10 , pp. 1810-1819
    • Kim, H.J.1    So, H.S.2    Lee, J.H.3    Lee, J.H.4    Park, C.5    Park, S.Y.6
  • 193
    • 84902846582 scopus 로고    scopus 로고
    • Solid self-microemulsifying dispersible tablets of celastrol: formulation development, characterization and bioavailability evaluation
    • Qi X, Qin J, Ma N, Chou X, Wu Z. Solid self-microemulsifying dispersible tablets of celastrol: formulation development, characterization and bioavailability evaluation. Int J Pharm (2014) 472(1-2):40-7. doi:10.1016/j.ijpharm.2014.06.019
    • (2014) Int J Pharm , vol.472 , Issue.1-2 , pp. 40-47
    • Qi, X.1    Qin, J.2    Ma, N.3    Chou, X.4    Wu, Z.5
  • 194
    • 84862330042 scopus 로고    scopus 로고
    • Celastrol nanoparticles inhibit corneal neovascularization induced by suturing in rats
    • Li Z, Yao L, Li J, Zhang W, Wu X, Liu Y, et al. Celastrol nanoparticles inhibit corneal neovascularization induced by suturing in rats. Int J Nanomedicine (2012) 7:1163-73. doi:10.2147/IJN.S27860
    • (2012) Int J Nanomedicine , vol.7 , pp. 1163-1173
    • Li, Z.1    Yao, L.2    Li, J.3    Zhang, W.4    Wu, X.5    Liu, Y.6
  • 196
    • 0037424425 scopus 로고    scopus 로고
    • Effects of demethylzeylasteral and celastrol on spermatogenic cell Ca2+ channels and progesterone-induced sperm acrosome reaction
    • Bai JP, Shi YL, Fang X, Shi QX. Effects of demethylzeylasteral and celastrol on spermatogenic cell Ca2+ channels and progesterone-induced sperm acrosome reaction. Eur J Pharmacol (2003) 464(1):9-15. doi:10.1016/S0014-2999(03)01351-7
    • (2003) Eur J Pharmacol , vol.464 , Issue.1 , pp. 9-15
    • Bai, J.P.1    Shi, Y.L.2    Fang, X.3    Shi, Q.X.4
  • 197
    • 33646851612 scopus 로고    scopus 로고
    • Chronic inhibition of cardiac Kir2.1 and HERG potassium channels by celastrol with dual effects on both ion conductivity and protein trafficking
    • Sun H, Liu X, Xiong Q, Shikano S, Li M. Chronic inhibition of cardiac Kir2.1 and HERG potassium channels by celastrol with dual effects on both ion conductivity and protein trafficking. J Biol Chem (2006) 281(9):5877-84. doi:10.1074/jbc.M600072200
    • (2006) J Biol Chem , vol.281 , Issue.9 , pp. 5877-5884
    • Sun, H.1    Liu, X.2    Xiong, Q.3    Shikano, S.4    Li, M.5
  • 198
    • 0034752311 scopus 로고    scopus 로고
    • Celastrol, a potent antioxidant and anti-inflammatory drug, as a possible treatment for Alzheimer's disease
    • Allison AC, Cacabelos R, Lombardi VR, Alvarez XA, Vigo C. Celastrol, a potent antioxidant and anti-inflammatory drug, as a possible treatment for Alzheimer's disease. Prog Neuropsychopharmacol Biol Psychiatry (2001) 25(7):1341-57. doi:10.1016/S0278-5846(01)00192-0
    • (2001) Prog Neuropsychopharmacol Biol Psychiatry , vol.25 , Issue.7 , pp. 1341-1357
    • Allison, A.C.1    Cacabelos, R.2    Lombardi, V.R.3    Alvarez, X.A.4    Vigo, C.5
  • 199
    • 77955771876 scopus 로고    scopus 로고
    • Celastrol attenuates hypertension-induced inflammation and oxidative stress in vascular smooth muscle cells via induction of heme oxygenase-1
    • Yu X, Tao W, Jiang F, Li C, Lin J, Liu C. Celastrol attenuates hypertension-induced inflammation and oxidative stress in vascular smooth muscle cells via induction of heme oxygenase-1. Am J Hypertens (2010) 23(8):895-903. doi:10.1038/ajh.2010.75
    • (2010) Am J Hypertens , vol.23 , Issue.8 , pp. 895-903
    • Yu, X.1    Tao, W.2    Jiang, F.3    Li, C.4    Lin, J.5    Liu, C.6
  • 200
    • 84973293346 scopus 로고    scopus 로고
    • Exosomal formulation enhances therapeutic response of celastrol against lung cancer
    • Aqil F, Kausar H, Agrawal AK, Jeyabalan J, Kyakulaga AH, Munagala R, et al. Exosomal formulation enhances therapeutic response of celastrol against lung cancer. Exp Mol Pathol (2016) 101(1):12-21. doi:10.1016/j.yexmp.2016.05.013
    • (2016) Exp Mol Pathol , vol.101 , Issue.1 , pp. 12-21
    • Aqil, F.1    Kausar, H.2    Agrawal, A.K.3    Jeyabalan, J.4    Kyakulaga, A.H.5    Munagala, R.6
  • 201
    • 84901272511 scopus 로고    scopus 로고
    • Enhancement of oral bioavailability of tripterine through lipid nanospheres: preparation, characterization, and absorption evaluation
    • Zhang X, Zhang T, Zhou X, Liu H, Sun H, Ma Z, et al. Enhancement of oral bioavailability of tripterine through lipid nanospheres: preparation, characterization, and absorption evaluation. J Pharm Sci (2014) 103(6):1711-9. doi:10.1002/jps.23967
    • (2014) J Pharm Sci , vol.103 , Issue.6 , pp. 1711-1719
    • Zhang, X.1    Zhang, T.2    Zhou, X.3    Liu, H.4    Sun, H.5    Ma, Z.6
  • 202
    • 84946605748 scopus 로고    scopus 로고
    • Nanoencapsulation of natural triterpenoid celastrol for prostate cancer treatment
    • Sanna V, Chamcheu JC, Pala N, Mukhtar H, Sechi M, Siddiqui IA. Nanoencapsulation of natural triterpenoid celastrol for prostate cancer treatment. Int J Nanomedicine (2015) 10:6835-46. doi:10.2147/IJN.S93752
    • (2015) Int J Nanomedicine , vol.10 , pp. 6835-6846
    • Sanna, V.1    Chamcheu, J.C.2    Pala, N.3    Mukhtar, H.4    Sechi, M.5    Siddiqui, I.A.6
  • 203
    • 84868138089 scopus 로고    scopus 로고
    • Modulation of inflammatory signaling and cytokine release from microglia by celastrol incorporated into dendrimer nanocarriers
    • Boridy S, Soliman GM, Maysinger D. Modulation of inflammatory signaling and cytokine release from microglia by celastrol incorporated into dendrimer nanocarriers. Nanomedicine (Lond) (2012) 7(8):1149-65. doi:10.2217/nnm.12.16
    • (2012) Nanomedicine (Lond) , vol.7 , Issue.8 , pp. 1149-1165
    • Boridy, S.1    Soliman, G.M.2    Maysinger, D.3
  • 204
    • 84904445281 scopus 로고    scopus 로고
    • Evaluation of anticancer activity of celastrol liposomes in prostate cancer cells
    • Wolfram J, Suri K, Huang Y, Molinaro R, Borsoi C, Scott B, et al. Evaluation of anticancer activity of celastrol liposomes in prostate cancer cells. J Microencapsul (2014) 31(5):501-7. doi:10.3109/02652048.2013.879932
    • (2014) J Microencapsul , vol.31 , Issue.5 , pp. 501-507
    • Wolfram, J.1    Suri, K.2    Huang, Y.3    Molinaro, R.4    Borsoi, C.5    Scott, B.6
  • 205
    • 84862798178 scopus 로고    scopus 로고
    • Preparation, characterization, and assessment of the antiglioma effects of liposomal celastrol
    • Huang Y, Zhou D, Hang T, Wu Z, Liu J, Xu Q, et al. Preparation, characterization, and assessment of the antiglioma effects of liposomal celastrol. Anticancer Drugs (2012) 23(5):515-24. doi:10.1097/CAD.0b013e3283514b68
    • (2012) Anticancer Drugs , vol.23 , Issue.5 , pp. 515-524
    • Huang, Y.1    Zhou, D.2    Hang, T.3    Wu, Z.4    Liu, J.5    Xu, Q.6
  • 206
    • 80053277669 scopus 로고    scopus 로고
    • Formulation and evaluation of celastrol-loaded liposomes
    • Song J, Shi F, Zhang Z, Zhu F, Xue J, Tan X, et al. Formulation and evaluation of celastrol-loaded liposomes. Molecules (2011) 16(9):7880-92. doi:10.3390/molecules16097880
    • (2011) Molecules , vol.16 , Issue.9 , pp. 7880-7892
    • Song, J.1    Shi, F.2    Zhang, Z.3    Zhu, F.4    Xue, J.5    Tan, X.6
  • 207
    • 84862276090 scopus 로고    scopus 로고
    • Optimized preparation of celastrol-loaded polymeric nanomicelles using rotatable central composite design and response surface methodology
    • Peng X, Wang J, Song H, Cui D, Li L, Li J, et al. Optimized preparation of celastrol-loaded polymeric nanomicelles using rotatable central composite design and response surface methodology. J Biomed Nanotechnol (2012) 8(3):491-9. doi:10.1166/jbn.2012.1398
    • (2012) J Biomed Nanotechnol , vol.8 , Issue.3 , pp. 491-499
    • Peng, X.1    Wang, J.2    Song, H.3    Cui, D.4    Li, L.5    Li, J.6
  • 208
    • 84870361878 scopus 로고    scopus 로고
    • Antitumor activity of celastrol nanoparticles in a xenograft retinoblastoma tumor model
    • Li Z, Wu X, Li J, Yao L, Sun L, Shi Y, et al. Antitumor activity of celastrol nanoparticles in a xenograft retinoblastoma tumor model. Int J Nanomedicine (2012) 7:2389-98. doi:10.2147/IJN.S29945
    • (2012) Int J Nanomedicine , vol.7 , pp. 2389-2398
    • Li, Z.1    Wu, X.2    Li, J.3    Yao, L.4    Sun, L.5    Shi, Y.6
  • 209
    • 84887325602 scopus 로고    scopus 로고
    • Antitumor activity of tripterine via cell-penetrating peptide-coated nanostructured lipid carriers in a prostate cancer model
    • Yuan L, Liu C, Chen Y, Zhang Z, Zhou L, Qu D. Antitumor activity of tripterine via cell-penetrating peptide-coated nanostructured lipid carriers in a prostate cancer model. Int J Nanomedicine (2013) 8:4339-50. doi:10.2147/IJN.S51621
    • (2013) Int J Nanomedicine , vol.8 , pp. 4339-4350
    • Yuan, L.1    Liu, C.2    Chen, Y.3    Zhang, Z.4    Zhou, L.5    Qu, D.6
  • 210
    • 84870366629 scopus 로고    scopus 로고
    • Formulation, characterization, and evaluation of in vitro skin permeation and in vivo pharmacodynamics of surface-charged tripterine-loaded nanostructured lipid carriers
    • Chen Y, Zhou L, Yuan L, Zhang ZH, Liu X, Wu Q. Formulation, characterization, and evaluation of in vitro skin permeation and in vivo pharmacodynamics of surface-charged tripterine-loaded nanostructured lipid carriers. Int J Nanomedicine (2012) 7:3023-32. doi:10.2147/IJN.S32476
    • (2012) Int J Nanomedicine , vol.7 , pp. 3023-3032
    • Chen, Y.1    Zhou, L.2    Yuan, L.3    Zhang, Z.H.4    Liu, X.5    Wu, Q.6
  • 211
    • 84861752543 scopus 로고    scopus 로고
    • Preparation of tripterine nanostructured lipid carriers and their absorption in rat intestine
    • Zhou L, Chen Y, Zhang Z, He J, Du M, Wu Q. Preparation of tripterine nanostructured lipid carriers and their absorption in rat intestine. Pharmazie (2012) 67(4):304-10
    • (2012) Pharmazie , vol.67 , Issue.4 , pp. 304-310
    • Zhou, L.1    Chen, Y.2    Zhang, Z.3    He, J.4    Du, M.5    Wu, Q.6
  • 212
    • 84937231871 scopus 로고    scopus 로고
    • Sugar-decorated mesoporous silica nanoparticles as delivery vehicles for the poorly soluble drug celastrol enables targeted induction of apoptosis in cancer cells
    • Niemela E, Desai D, Nkizinkiko Y, Eriksson JE, Rosenholm JM. Sugar-decorated mesoporous silica nanoparticles as delivery vehicles for the poorly soluble drug celastrol enables targeted induction of apoptosis in cancer cells. Eur J Pharm Biopharm (2015) 96:11-21. doi:10.1016/j.ejpb.2015.07.009
    • (2015) Eur J Pharm Biopharm , vol.96 , pp. 11-21
    • Niemela, E.1    Desai, D.2    Nkizinkiko, Y.3    Eriksson, J.E.4    Rosenholm, J.M.5
  • 213
    • 84862779635 scopus 로고    scopus 로고
    • Targeted inhibition of tumor proliferation, survival, and metastasis by pentacyclic triterpenoids: potential role in prevention and therapy of cancer
    • Shanmugam MK, Nguyen AH, Kumar AP, Tan BK, Sethi G. Targeted inhibition of tumor proliferation, survival, and metastasis by pentacyclic triterpenoids: potential role in prevention and therapy of cancer. Cancer Lett (2012) 320(2):158-70. doi:10.1016/j.canlet.2012.02.037
    • (2012) Cancer Lett , vol.320 , Issue.2 , pp. 158-170
    • Shanmugam, M.K.1    Nguyen, A.H.2    Kumar, A.P.3    Tan, B.K.4    Sethi, G.5
  • 214
    • 84862792928 scopus 로고    scopus 로고
    • Fighting fire with fire: poisonous Chinese herbal medicine for cancer therapy
    • Wang S, Wu X, Tan M, Gong J, Tan W, Bian B, et al. Fighting fire with fire: poisonous Chinese herbal medicine for cancer therapy. J Ethnopharmacol (2012) 140(1):33-45. doi:10.1016/j.jep.2011.12.041
    • (2012) J Ethnopharmacol , vol.140 , Issue.1 , pp. 33-45
    • Wang, S.1    Wu, X.2    Tan, M.3    Gong, J.4    Tan, W.5    Bian, B.6
  • 215
    • 36749077060 scopus 로고    scopus 로고
    • Preclinical studies of celastrol and acetyl isogambogic acid in melanoma
    • Abbas S, Bhoumik A, Dahl R, Vasile S, Krajewski S, Cosford ND, et al. Preclinical studies of celastrol and acetyl isogambogic acid in melanoma. Clin Cancer Res (2007) 13(22 Pt 1):6769-78. doi:10.1158/1078-0432.CCR-07-1536
    • (2007) Clin Cancer Res , vol.13 , Issue.22 , pp. 6769-6778
    • Abbas, S.1    Bhoumik, A.2    Dahl, R.3    Vasile, S.4    Krajewski, S.5    Cosford, N.D.6
  • 216
    • 77954309816 scopus 로고    scopus 로고
    • Synthesis and preliminary evaluation of neuroprotection of celastrol analogues in PC12 cells
    • Sun H, Xu L, Yu P, Jiang J, Zhang G, Wang Y. Synthesis and preliminary evaluation of neuroprotection of celastrol analogues in PC12 cells. Bioorg Med Chem Lett (2010) 20(13):3844-7. doi:10.1016/j.bmcl.2010.05.066
    • (2010) Bioorg Med Chem Lett , vol.20 , Issue.13 , pp. 3844-3847
    • Sun, H.1    Xu, L.2    Yu, P.3    Jiang, J.4    Zhang, G.5    Wang, Y.6
  • 217
    • 84906282762 scopus 로고    scopus 로고
    • Novel celastrol derivatives inhibit the growth of hepatocellular carcinoma patient-derived xenografts
    • Wei W, Wu S, Wang X, Sun CK, Yang X, Yan X, et al. Novel celastrol derivatives inhibit the growth of hepatocellular carcinoma patient-derived xenografts. Oncotarget (2014) 5(14):5819-31. doi:10.18632/oncotarget.2171
    • (2014) Oncotarget , vol.5 , Issue.14 , pp. 5819-5831
    • Wei, W.1    Wu, S.2    Wang, X.3    Sun, C.K.4    Yang, X.5    Yan, X.6
  • 218
    • 84856211622 scopus 로고    scopus 로고
    • Terpenoids from root bark of Celastrus orbiculatus
    • Wu J, Zhou Y, Wang L, Zuo J, Zhao W. Terpenoids from root bark of Celastrus orbiculatus. Phytochemistry (2012) 75:159-68. doi:10.1016/j.phytochem.2011.11.023
    • (2012) Phytochemistry , vol.75 , pp. 159-168
    • Wu, J.1    Zhou, Y.2    Wang, L.3    Zuo, J.4    Zhao, W.5
  • 219
    • 84904808084 scopus 로고    scopus 로고
    • Design, synthesis and biological evaluation of C6-modified celastrol derivatives as potential antitumor agents
    • Tang K, Huang Q, Zeng J, Wu G, Huang J, Pan J, et al. Design, synthesis and biological evaluation of C6-modified celastrol derivatives as potential antitumor agents. Molecules (2014) 19(7):10177-88. doi:10.3390/molecules190710177
    • (2014) Molecules , vol.19 , Issue.7 , pp. 10177-10188
    • Tang, K.1    Huang, Q.2    Zeng, J.3    Wu, G.4    Huang, J.5    Pan, J.6
  • 220
    • 84925399465 scopus 로고    scopus 로고
    • Design and synthesis of celastrol derivatives as anticancer agents
    • Tang WJ, Wang J, Tong X, Shi JB, Liu XH, Li J. Design and synthesis of celastrol derivatives as anticancer agents. Eur J Med Chem (2015) 95:166-73. doi:10.1016/j.ejmech.2015.03.039
    • (2015) Eur J Med Chem , vol.95 , pp. 166-173
    • Tang, W.J.1    Wang, J.2    Tong, X.3    Shi, J.B.4    Liu, X.H.5    Li, J.6


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.