Journal List > J Korean Ophthalmol Soc > v.55(7) > 1009732

Park, Hong, Iizuka, Kim, and Seong: Cytotoxicities and Anti-Fibrotic Effects of Pirfenidone and Mitomycin C on Human Fibroblasts

Abstract

Purpose

The cytotoxicities and anti-fibrotic effects of mitomycin C and pirfenidone on human dermal fibroblast were evaluated.

Methods

Initially, 24-hour cell cultures were exposed to transforming growth factor (TGF)-β1, different concentrations of mitomycin C, and pirfenidone solutions in order to evaluate cytotoxicity. Expressions of fibronectin, collagen type 1, α smooth muscle, and β-actin were evaluated by real-time reverse transcription-polymerase chain reaction (RT-PCR) and western blot in mitomycin C solutions at concentrations of 4 μg/mL and 20 μg/mL, and in pirfenidone solutions at 250 μg/mL and 500 μg/mL.

Results

In comparison to cell cultures exposed to TGF-β1 solutions, cytotoxicities were increased in solutions of mitomycin C at 4 μg/mL, 20 μg/mL, 40 μg/mL and pirfenidone at 500 μg/mL, 750 μg/mL, 1,000 μg/mL (p < 0.05, Mann Whitney U-test). The results of real-time RT-PCR show that expressions of fibronectin, collagen type 1, and α smooth muscle were significantly more decreased in all concentrations of mitomycin C and pirfenidone compared to those in TGF-β1 solution. In western blot analysis, expressions of fibronectin and α smooth muscle were decreased in all concentrations of mitomycin C and pirfenidone compared to TGF-β1 solution.

Conclusions

Both drugs have cytotoxicities and anti-fibrotic effects, but pirfenidone was found to have less cytotoxicity and mitomycin C was found to have more anti-fibrotic effects when compared to each other.

References

1. Carter NJ. Pirfenidone: in idiopathic pulmonary fibrosis. Drugs. 2011; 71:1721–32.
2. Richeldi L, Yasothan U, Kirkpatrick P. Pirfenidone. Nat Rev Drug Discov. 2011; 10:489–90.
crossref
3. Iyer SN, Gurujeyalakshmi G, Giri SN. Effects of pirfenidone on transforming growth factor-beta gene expression at the transcriptional level in bleomycin hamster model of lung fibrosis. J Pharmacol Exp Ther. 1999; 291:367–73.
4. Hewitson TD, Kelynack KJ, Tait MG, et al. Pirfenidone reduces in vitro rat renal fibroblast activation and mitogenesis. J Nephrol. 2001; 14:453–60.
5. Gurujeyalakshmi G, Hollinger MA, Giri SN. Pirfenidone inhibits PDGF isoforms in bleomycin hamster model of lung fibrosis at the translational level. Am J Physiol. 1999; 276:L311–8.
6. Oku H, Nakazato H, Horikawa T, et al. Pirfenidone suppresses tumor necrosis factor-alpha, enhances interleukin-10 and protects mice from endotoxic shock. Eur J Pharmacol. 2002; 446:167–76.
7. Oku H, Shimizu T, Kawabata T, et al. Antifibrotic action of pirfenidone and prednisolone: different effects on pulmonary cytokines and growth factors in bleomycin-induced murine pulmonary fibrosis. Eur J Pharmacol. 2008; 590:400–8.
crossref
8. Iyer SN, Gurujeyalakshmi G, Giri SN. Effects of pirfenidone on procollagen gene expression at the transcriptional level in bleomycin hamster model of lung fibrosis. J Pharmacol Exp Ther. 1999; 289:211–8.
9. Mitani Y, Sato K, Muramoto Y, et al. Superoxide scavenging activity of pirfenidone-iron complex. Biochem Biophys Res Commun. 2008; 372:19–23.
crossref
10. Di Sario A, Bendia E, Svegliati Baroni G, et al. Effect of pirfenidone on rat hepatic stellate cell proliferation and collagen production. J Hepatol. 2002; 37:584–91.
crossref
11. Yamazaki T, Yamashita N, Izumi Y, et al. The antifibrotic agent pirfenidone inhibits angiotensin II-induced cardiac hypertrophy in mice. Hypertens Res. 2012; 35:34–40.
crossref
12. Zhang S, Shiels IA, Ambler JS, Taylor SM. Pirfenidone reduces fibronectin synthesis by cultured human retinal pigment epithelial cells. Aust NZ J Ophthalmol. 1998; 26(Suppl 1):S74–6.
crossref
13. Lin X, Yu M, Wu K, et al. Effects of pirfenidone on proliferation, migration, and collagen contraction of human Tenon's fibroblasts in vitro. Invest Ophthalmol Vis Sci. 2009; 50:3763–70.
crossref
14. Kim H, Choi YH, Park SJ, et al. Antifibrotic effect of Pirfenidone on orbital fibroblasts of patients with thyroid-associated ophthalmopathy by decreasing TIMP-1 and collagen levels. Invest Ophthalmol Vis Sci. 2010; 51:3061–6.
crossref
15. Zhong H, Sun G, Lin X, et al. Evaluation of pirfenidone as a new postoperative antiscarring agent in experimental glaucoma surgery. Invest Ophthalmol Vis Sci. 2011; 52:3136–42.
crossref
16. Lama PJ, Fechtner RD. Antifibrotics and wound healing in glaucoma surgery. Surv Ophthalmol. 2003; 48:314–46.
crossref
17. Palanca-Capistrano AM, Hall J, Cantor LB, et al. Long-term out- comes of intraoperative 5-fluoro uracil versus intraoperative mito- mycin C in primary trabeculectomy surgery. Ophthalmology. 2009; 116:185–90.
18. Franks WA, Hitchings RA. Complications of 5–fluorouracil after trabeculectomy. Eye (Lond). 1991; 5:385–9.
crossref
19. Seong GJ, Park C, Kim CY, et al. Mitomycin-C induces the apoptosis of human Tenon's capsule fibroblast by activation of c-Jun N-terminal kinase 1 and caspase-3 protease. Invest Ophthalmol Vis Sci. 2005; 46:3545–52.
20. Chen T, Kunnavatana SS, Koch RJ. Effects of mitomycin-C on normal dermal fibroblasts. Laryngoscope. 2006; 116:514–7.
crossref
21. Anderson MT, Staal FJ, Gitler C, et al. Separation of oxidant-initiated and redox-regulated steps in the NF-kappa B signal transduction pathway. Proc Natl Acad Sci U S A. 1994; 91:11527–31.
crossref
22. Yao KS, O'Dwyer PJ. Involvement of NF-kappa B in the induction of NAD(P)H:quinone oxidoreductase (DT-diaphorase) by hypo-xia, oltipraz and mitomycin C. Biochem Pharmacol. 1995; 49:275–82.
23. Baldassarre F, Mallardo M, Mezza E, et al. Regulation of NF-kappa B through the nuclear processing of p105 (NF-kappa B1) in Epstein-Barr virus-immortalized B cell lines. J Biol Chem. 1995; 270:31244–8.
24. Stahnke T, Löbler M, Kastner C, et al. Different fibroblast subpopulations of the eye: a therapeutic target to prevent postoperative fibrosis in glaucoma therapy. Exp Eye Res. 2012; 100:88–97.
crossref
25. Shihab FS, Bennett WM, Yi H, Andoh TF. Effect of pirfenidone on apoptosis-regulatory genes in chronic cyclosporine nephrotoxicity. Transplantation. 2005; 79:419–26.
crossref

Figure 1.
Result of lactate dehydrogenase (LDH) assay. M4, M5, M6, P4, P5, P6 solutions increased cytotoxicity significantly (*p < 0.05, Mann Whitney U-test). Sample C: only Dulbecco's modified eagle's medium (DMEM); T-P6: add transforming growth factor (TGF)-βl 5 ng/mL; M1-M6: add Mitomycin C, 0.00004 (Ml), 0.004 (M2), 0.4 (M3), 4 (M4), 20 (M5), 40 μg/mL (M6; P1-P6: add Pirfenidone, 10 (Pl), 100 (P2), 250 (P3), 500 (P4), 750 (P5), 1,000 μg/mL (P6). TGF = transforming growth factor; C = control; T = TGF-β 1; M = mitomycin C; P = pirfenidone.
jkos-55-1077f1.tif
Figure 2.
Result of real time reverse transcription polymerase chain reaction (RT-PCR). Mitomycin C and pirfenidone solution decreased ribonucleic acid (RNA) expression of fibronectin, collagen I, α smooth muscle, which compared to the only transforming growth factor (TGF)-β1 solution.
jkos-55-1077f2.tif
Figure 3.
Result of western blot. Mitomycin C and pirfenidone solution decreased protein expression of fibronectin, α smooth muscle, which compared to the only transforming growth factor (TGF)-β1 solution. Protein expression of collagen type I was too weak.
jkos-55-1077f3.tif
Table 1.
Primer sequences for real time reverse transcription polymerase chain reaction (RT-PCR)
Gene name Sequence
Fibronectin F 5’- CCG TGG GCA ACT CTG TC-3’
R 5’- TGC GGC AGT TGT CAC AG-3’
Collagen type 1 F 5’- CAC CAA TCA CCT GCG GTA CAG AA-3’
R 5’- CAG ATC ACG TCA TCG CAC AAC-3’
α smooth muscle F 5’- GTG TTA TGT AGC TCT GGA CTT TG AAAA-3’
R 5’- GGC AGC GGA AAC GTT CAT T-3’
β-actin F 5’- GCG GGA AAT CGT GCG TGA CAT T-3’
R 5’- GAT GGA GTT GAA GGT AGT TTC GTG-3’

F = forward; R = reverse.

TOOLS
Similar articles