Pirfenidone attenuates bleomycin-induced pulmonary fibrosis in mice by regulating Nrf2/Bach1 equilibrium

Yuan Liu, Fuai Lu, Lirong Kang, Zhihua Wang, Yongfu Wang, Yuan Liu, Fuai Lu, Lirong Kang, Zhihua Wang, Yongfu Wang

Abstract

Background: Oxidative stress is one of the important factors involved in the pathogenesis of idiopathic pulmonary fibrosis (IPF). The equilibrium of Nuclear factor-erythroid-related factor 2 (Nrf2)/[BTB (broad-complex, tramtrack and bric-a-brac) and CNC (cap'n'collar protein) homology 1, Bach1] determines the expression level of antioxidant factors, further regulating the function of oxidation/antioxidation capacity. Pirfenidone (PFD) is one of two currently for IPF therapy approved drugs. PFD regulates intracellular antioxidants, inhibits secretion of inflammatory cytokines and collagen synthesis. However the mechanisms of its antioxidant effects remain elusive.

Methods: Effects of PFD treatment were studied in mouse lung fibroblasts (MLF) following induction by transforming-growth factor beta 1 (TGF-β1) and in mice following bleomycin-induced lung fibrosis. The mRNA and protein levels of oxidative stress-related factors Nrf2/Bach1 and their downstream antioxidant factors heme oxygenase-1 (Ho-1) and glutathione peroxidase 1 (Gpx1) were determined by RT-PCR and Western blot. Fibrosis-related cytokines interleukin-6 (IL-6) and myofibroblast markers type 1 collagen α1 (COL1A1) levels in supernate of MLF, serum, and bronchoalveolar lavage fluid (BALF) as well as malondialdehyde (MDA) in serum and BALF were detected by ELISA, reactive oxygen species (ROS) generation was measured by 2',7'- dichlorofluorescin diacetate (DCFH-DA) assay and lung pathological/morphological alterations in mice were observed by HE and Masson to assess the antioxidant mechanism and therapeutic effects on pulmonary fibrosis induced by bleomycin.

Results: PFD inhibited Bach1 mRNA and protein expressions in mouse lung fibroblasts induced by TGF-β1 and lung tissues with pulmonary fibrosis induced by bleomycin. Furthermore, it improved Nrf2, Ho-1 and Gpx1 mRNA and protein expressions. After PFD treatment, COL1A1and IL-6 levels in supernate of MLF, serum, and BALF as well as ROS in lung tissues and MDA in serum and BALF from a mouse with pulmonary fibrosis were significantly decreased, and the infiltration of lung inflammatory cells and fibrosis degree were alleviated.

Conclusions: Theraputic effects of PFD for IPF were involved in Nrf2/Bach1 equilibrium which regulated the capacity of oxidative stress. The study provided new insights into the antioxidant mechanism of PFD.

Keywords: Idiopathic pulmonary fibrosis; Nrf2/Bach1; Oxidative stress; Pirfenidone.

Figures

Fig. 1
Fig. 1
Expression of oxidant/antioxidant factors in Mouse lung fibroblasts (MLF) incubated for 24 h with TGF-β1 and various concentrations of pirfenidone (PFD) for 48 h. MLF cells were stimulated with TGF-β1 (5 ng/mL) for 24 h before PFD (100 μg/mL, 200 μg/mL and 500 μg/mL) treatment for 48 h. The mRNA expression of nuclear factor-erythroid-related factor 2 (Nrf2), [BTB (broad-complex, tramtrack and bric-a-brac) and CNC (cap‘n’collar protein) homology 1, Bach1], heme oxygenase-1 (HO-1), and glutathione peroxidase 1 (GPx1) in MLF cells was analyzed by reverse transcription-polymerase chain reaction (RT-PCR) (a, b, c, d). The protein levels of Nrf2, Bach1, Ho-1, and GPx1 in MLF cells were detected by Western blot (e). Data are expressed as the mean ± SD. *P < 0.05, **P < 0.01, vs. TGF-β1 group
Fig. 2
Fig. 2
Effects of PFD on oxidant/antioxidant factors in Bleomycin (BLM)-induced pulmonary fibrosis in mice (3 in each group). BLM (5 mg/kg, intratracheally instillation for 14d) administered mice were then treated with PFD (300 mg/kg/d orally administered) for 4 weeks. Control group was intratracheally administered 50 μL 0.9% saline), only PFD group was orally administrated with 300 mg/kg PFD daily for 4 weeks. The mRNA expressions of Nrf2, Bach1, HO-1, and GPx1 in the lung were analyzed by RT-PCR (a, b, c, d); protein levels of Nrf2, Bach1, HO-1, and GPx1 in the lung were detected by Western blot (e). Data are expressed as the mean ± SD. *P < 0.05, **P < 0.01, vs. BLM group
Fig. 3
Fig. 3
Analysis of type 1 collagen α1 (COL1A1) and interleukin-6 (IL-6) levels in the supernatant of cells, serum and bronchoalveolar lavage fluid (BALF). They were measured by enzyme-linked immunosorbent assay (ELISA). Data are expressed as the mean ± SD. a and b: COL1A1 and IL-6 levels in the supernatant of cells after TGF-β1 and various concentrations of PFD stimulation (*P < 0.05, **P < 0.01, vs. TGF-β1 group). c and d: Serum COL1A1 and IL-6 after BLM and PFD treatment (*P < 0.05, **P < 0.01, vs. BLM 14d and 42d group). e and f: COL1A1 and IL-6 levels in BALF after BLM and PFD treatment (*P < 0.05, **P < 0.01, vs. BLM 14d and 42d group)
Fig. 4
Fig. 4
Effect of PFD on indicators of oxidative stress such as reactive oxygen species (ROS) generation and malondialdehyde (MDA) expression. a: ROS generation (%) in lung tissue was detected with 2,7-dichlorofluorescein diacetate (DCFDA) by flow cytometric. b and c: MDA expression in serum and BALF by ELISA. Data represent as the means ± SD. *P < 0.05, **P < 0.01, vs. BLM 14d and 42d group
Fig. 5
Fig. 5
Histopathological changes of lung tissue in mice at the end point of the experiment (HE staining, ×200). BLM (5 mg/kg) intratracheally administered once for 2 weeks and then treated with pirfenidone (300 mg/kg/d orally administered) for 4 weeks. Lung histological data obtained on day 14 after BLM treatment and day 42 after PFD treatment (BLM 42 d). a: Lung inflammation, fibrosis, and integrity of the structure was evaluated by H&E (×200). Degree of fibrosis in lung tissues by Masson’s staining (×200). b and c: Degree of lung fibrosis was evaluated by inflammation and fibrosis scored. The data are expressed as mean ± SD,*P < 0.05, **P < 0.01, vs. BLM 14d and 42 d group

References

    1. Selman M, Pardo A. Role of epithelial cells in idiopathic pulmonary fibrosis: from innocent targets to serial killers. Proc Am Thorac Soc. 2006;3:364–72. doi: 10.1513/pats.200601-003TK.
    1. Yoshida M, Sakuma J, Hayashi S, Abe K, Saito I, Harada S, et al. A histologically distinctive interstitial pneumonia induced by overexpression of the interleukin 6, transforming growth factor beta 1, or platelet-derived growth factor B gene. Proc Natl Acad Sci U S A. 1995;92:9570–4. doi: 10.1073/pnas.92.21.9570.
    1. Raghu G, Collard HR, Egan JJ, Martinez FJ, Behr J, Brown KK, et al. An official ATS/ERS/JRS/ALAT statement: idiopathic pulmonary fibrosis: evidence-based guidelines for diagnosis and management. Am J Respir Crit Care Med. 2011;183:788–824. doi: 10.1164/rccm.2009-040GL.
    1. Lee CM, Park JW, Cho WK, Zhou Y, Han B, Yoon PO, et al. Modifiers of TGF-beta1 effector function as novel therapeutic targets of pulmonary fibrosis. Korean J Intern Med. 2014;29:281–90. doi: 10.3904/kjim.2014.29.3.281.
    1. Collard HR, Ryu JH, Douglas WW, Schwarz MI, Curran-Everett D, King TE, Jr, et al. Combined corticosteroid and cyclophosphamide therapy does not alter survival in idiopathic pulmonary fibrosis. Chest. 2004;125:2169–74. doi: 10.1378/chest.125.6.2169.
    1. Izumi S, Iikura M, Hirano S. Prednisone, azathioprine, and N-acetylcysteine for pulmonary fibrosis. N Engl J Med. 2012;367:870.
    1. Puglisi S, Torrisi SE, Vindigni V, Giuliano R, Palmucci S, Mule M, et al. New perspectives on management of idiopathic pulmonary fibrosis. Ther Adv Chronic Dis. 2016;7:108–20. doi: 10.1177/2040622315624276.
    1. Iyer SN, Hyde DM, Giri SN. Anti-inflammatory effect of pirfenidone in the bleomycin-hamster model of lung inflammation. Inflammation. 2000;24:477–91. doi: 10.1023/A:1007068313370.
    1. Inomata M, Kamio K, Azuma A, Matsuda K, Kokuho N, Miura Y, et al. Pirfenidone inhibits fibrocyte accumulation in the lungs in bleomycin-induced murine pulmonary fibrosis. Respir Res. 2014;15:16. doi: 10.1186/1465-9921-15-16.
    1. Noble PW, Albera C, Bradford WZ, Costabel U, du Bois RM, Fagan EA, et al. Pirfenidone for idiopathic pulmonary fibrosis: analysis of pooled data from three multinational phase 3 trials. Eur Respir J. 2016;47:243–53. doi: 10.1183/13993003.00026-2015.
    1. Giri SN, Leonard S, Shi X, Margolin SB, Vallyathan V. Effects of pirfenidone on the generation of reactive oxygen species in vitro. J Environ Pathol Toxicol Oncol. 1999;18:169–77.
    1. Macias-Barragan J, Caligiuri A, Garcia-Banuelos J, Parola M, Pinzani M, Armendariz-Borunda J. Effects of alpha lipoic acid and pirfenidone on liver cells antioxidant modulation against oxidative damage. Rev Med Chil. 2014;142:1553–64. doi: 10.4067/S0034-98872014001200009.
    1. Schunemann HJ, Muti P, Trevisan M. Serum indicators of free radical activity in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 1997;155:769. doi: 10.1164/ajrccm.155.2.9032227.
    1. Matsuzawa Y, Kawashima T, Kuwabara R, Hayakawa S, Irie T, Yoshida T, et al. Change in serum marker of oxidative stress in the progression of idiopathic pulmonary fibrosis. Pulm Pharmacol Ther. 2015;32:1–6. doi: 10.1016/j.pupt.2015.03.005.
    1. Chen JD, Evans RM. A transcriptional co-repressor that interacts with nuclear hormone receptors. Nature. 1995;377:454–7. doi: 10.1038/377454a0.
    1. Giudice A, Arra C, Turco MC. Review of molecular mechanisms involved in the activation of the Nrf2-ARE signaling pathway by chemopreventive agents. Methods Mol Biol. 2010;647:37–74. doi: 10.1007/978-1-60761-738-9_3.
    1. Jyrkkanen HK, Kuosmanen S, Heinaniemi M, Laitinen H, Kansanen E, Mella-Aho E, et al. Novel insights into the regulation of antioxidant-response-element-mediated gene expression by electrophiles: induction of the transcriptional repressor BACH1 by Nrf2. Biochem J. 2011;440:167–74. doi: 10.1042/BJ20110526.
    1. Santos-Silva MA, Pires KM, Trajano ET, Martins V, Nesi RT, Benjamin CF, et al. Redox imbalance and pulmonary function in bleomycin-induced fibrosis in C57BL/6, DBA/2, and BALB/c mice. Toxicol Pathol. 2012;40:731–41. doi: 10.1177/0192623312441404.
    1. Kikuchi N, Ishii Y, Morishima Y, Yageta Y, Haraguchi N, Itoh K, et al. Nrf2 protects against pulmonary fibrosis by regulating the lung oxidant level and Th1/Th2 balance. Respir Res. 2010;11:31. doi: 10.1186/1465-9921-11-31.
    1. Liu Y, Zheng Y. Bach1 siRNA attenuates bleomycin-induced pulmonary fibrosis by modulating oxidative stress in mice. Int J Mol Med. 2017;39:91–100.
    1. Szapiel SV, Elson NA, Fulmer JD, Hunninghake GW, Crystal RG. Bleomycin-induced interstitial pulmonary disease in the nude, athymic mouse. Am Rev Respir Dis. 1979;120:893–9.
    1. Ashcroft T, Simpson JM, Timbrell V. Simple method of estimating severity of pulmonary fibrosis on a numerical scale. J Clin Pathol. 1988;41:467–70. doi: 10.1136/jcp.41.4.467.
    1. Lasky J. Pirfenidone. IDrugs. 2004;7:166–72.
    1. Bando M, Yamauchi H, Ogura T, Taniguchi H, Watanabe K, Azuma A, et al. Clinical experience of the long-term use of pirfenidone for idiopathic pulmonary fibrosis. Intern Med. 2016;55:443–8. doi: 10.2169/internalmedicine.55.5272.
    1. Antoniu SA. Pirfenidone for the treatment of idiopathic pulmonary fibrosis. Expert Opin Investig Drugs. 2006;15:823–8. doi: 10.1517/13543784.15.7.823.
    1. Misra HP, Rabideau C. Pirfenidone inhibits NADPH-dependent microsomal lipid peroxidation and scavenges hydroxyl radicals. Mol Cell Biochem. 2000;204:119–26. doi: 10.1023/A:1007023532508.
    1. Zucker SN, Fink EE, Bagati A, Mannava S, Bianchi-Smiraglia A, Bogner PN, et al. Nrf2 amplifies oxidative stress via induction of Klf9. Mol Cell. 2014;53:916–28. doi: 10.1016/j.molcel.2014.01.033.
    1. Cho HY, Reddy SP, Kleeberger SR. Nrf2 defends the lung from oxidative stress. Antioxid Redox Signal. 2006;8:76–87. doi: 10.1089/ars.2006.8.76.
    1. Artaud-Macari E, Goven D, Brayer S, Hamimi A, Besnard V, Marchal-Somme J, et al. Nuclear factor erythroid 2-related factor 2 nuclear translocation induces myofibroblastic dedifferentiation in idiopathic pulmonary fibrosis. Antioxid Redox Signal. 2013;18:66–79. doi: 10.1089/ars.2011.4240.
    1. Ni S, Wang D, Qiu X, Pang L, Song Z, Guo K. Bone marrow mesenchymal stem cells protect against bleomycin-induced pulmonary fibrosis in rat by activating Nrf2 signaling. Int J Clin Exp Pathol. 2015;8:7752–61.
    1. Dhakshinamoorthy S, Jain AK, Bloom DA, Jaiswal AK. Bach1 competes with Nrf2 leading to negative regulation of the antioxidant response element (ARE)-mediated NAD(P)H:quinone oxidoreductase 1 gene expression and induction in response to antioxidants. J Biol Chem. 2005;280:16891–900. doi: 10.1074/jbc.M500166200.
    1. Warnatz HJ, Schmidt D, Manke T, Piccini I, Sultan M, Borodina T, et al. The BTB and CNC homology 1 (BACH1) target genes are involved in the oxidative stress response and in control of the cell cycle. J Biol Chem. 2011;286:23521–32. doi: 10.1074/jbc.M111.220178.
    1. MacLeod AK, McMahon M, Plummer SM, Higgins LG, Penning TM, Igarashi K, et al. Characterization of the cancer chemopreventive NRF2-dependent gene battery in human keratinocytes: demonstration that the KEAP1-NRF2 pathway, and not the BACH1-NRF2 pathway, controls cytoprotection against electrophiles as well as redox-cycling compounds. Carcinogenesis. 2009;30:1571–80. doi: 10.1093/carcin/bgp176.
    1. Hinz B, Phan SH, Thannickal VJ, Galli A, Bochaton-Piallat ML, Gabbiani G. The myofibroblast: one function, multiple origins. Am J Pathol. 2007;170:1807–16. doi: 10.2353/ajpath.2007.070112.
    1. Moeller A, Ask K, Warburton D, Gauldie J, Kolb M. The bleomycin animal model: a useful tool to investigate treatment options for idiopathic pulmonary fibrosis? Int J Biochem Cell Biol. 2008;40:362–82. doi: 10.1016/j.biocel.2007.08.011.
    1. Manoury B, Nenan S, Leclerc O, Guenon I, Boichot E, Planquois JM, et al. The absence of reactive oxygen species production protects mice against bleomycin-induced pulmonary fibrosis. Respir Res. 2005;6:11. doi: 10.1186/1465-9921-6-11.
    1. Cheresh P, Kim SJ, Tulasiram S, Kamp DW. Oxidative stress and pulmonary fibrosis. Biochim Biophys Acta. 2013;832:1028–40. doi: 10.1016/j.bbadis.2012.11.021.
    1. Lei Y, Wang K, Deng L, Chen Y, Nice EC, Huang C. Redox regulation of inflammation: old elements, a new story. Med Res Rev. 2015;35:306–40. doi: 10.1002/med.21330.
    1. Yu WN, Sun LF, Yang H. Inhibitory effects of astragaloside IV on bleomycin-induced pulmonary fibrosis in rats Via attenuation of oxidative stress and inflammation. Inflammation. 2016;39:1835–41. doi: 10.1007/s10753-016-0420-5.
    1. Ermis H, Parlakpinar H, Gulbas G, Vardi N, Polat A, Cetin A, et al. Protective effect of dexpanthenol on bleomycin-induced pulmonary fibrosis in rats. Naunyn Schmiedebergs Arch Pharmacol. 2013;386:1103–10. doi: 10.1007/s00210-013-0908-6.
    1. Yao R, Cao Y, He YR, Lau WB, Zeng Z, Liang ZA. Adiponectin attenuates lung fibroblasts activation and pulmonary fibrosis induced by paraquat. PLoS One. 2015;10:e0125169. doi: 10.1371/journal.pone.0125169.
    1. Degryse AL, Tanjore H, Xu XC, Polosukhin VV, Jones BR, McMahon FB, et al. Repetitive intratracheal bleomycin models several features of idiopathic pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol. 2010;299:L442–52. doi: 10.1152/ajplung.00026.2010.

Source: PubMed

3
订阅