DOI QR코드

DOI QR Code

Apolipoprotein A1 Inhibits TGF-β1-Induced Epithelial-to-Mesenchymal Transition of Alveolar Epithelial Cells

  • Baek, Ae Rin (Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital) ;
  • Lee, Ji Min (Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital) ;
  • Seo, Hyun Jung (Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital) ;
  • Park, Jong Sook (Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital) ;
  • Lee, June Hyuk (Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital) ;
  • Park, Sung Woo (Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital) ;
  • Jang, An Soo (Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital) ;
  • Kim, Do Jin (Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital) ;
  • Koh, Eun Suk (Department of Pathology, Soonchunhyang University Bucheon Hospital) ;
  • Uh, Soo Taek (Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Hospital) ;
  • Kim, Yong Hoon (Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Cheonan Hospital) ;
  • Park, Choon Sik (Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital)
  • 투고 : 2015.12.19
  • 심사 : 2016.05.03
  • 발행 : 2016.07.01

초록

Background: Idiopathic pulmonary fibrosis (IPF) is a progressive and lethal lung disease characterized by the accumulation of excessive fibroblasts and myofibroblasts in the extracellular matrix. The transforming growth factor ${\beta}1$ (TGF-${\beta}1$)-induced epithelial-to-mesenchymal transition (EMT) is thought to be a possible source of fibroblasts/myofibroblasts in IPF lungs. We have previously reported that apolipoprotein A1 (ApoA1) has anti-fibrotic activity in experimental lung fibrosis. In this study, we determine whether ApoA1 modulates TGF-${\beta}1$-induced EMT in experimental lung fibrosis and clarify its mechanism of action. Methods: The A549 alveolar epithelial cell line was treated with TGF-${\beta}1$ with or without ApoA1. Morphological changes and expression of EMT-related markers, including E-cadherin, N-cadherin, and ${\alpha}$-smooth muscle actin were evaluated. Expressions of Smad and non-Smad mediators and TGF-${\beta}1$ receptor type 1 ($T{\beta}RI$) and type 2 ($T{\beta}RII$) were measured. The silica-induced lung fibrosis model was established using ApoA1 overexpressing transgenic mice. Results: TGF-${\beta}1$-treated A549 cells were changed to the mesenchymal morphology with less E-cadherin and more N-cadherin expression. The addition of ApoA1 inhibited the TGF-${\beta}1$-induced change of the EMT phenotype. ApoA1 inhibited the TGF-${\beta}1$-induced increase in the phosphorylation of Smad2 and 3 as well as that of ERK and p38 mitogen-activated protein kinase mediators. In addition, ApoA1 reduced the TGF-${\beta}1$-induced increase in $T{\beta}RI$ and $T{\beta}RII$ expression. In a mouse model of silica-induced lung fibrosis, ApoA1 overexpression reduced the silica-mediated effects, which were increased N-cadherin and decreased E-cadherin expression in the alveolar epithelium. Conclusion: Our data demonstrate that ApoA1 inhibits TGF-${\beta}1$-induced EMT in experimental lung fibrosis.

키워드

참고문헌

  1. Selman M, Pardo A. Idiopathic pulmonary fibrosis: an epithelial/fibroblastic cross-talk disorder. Respir Res 2002;3:3.
  2. Harari S, Caminati A. Idiopathic pulmonary fibrosis. Allergy 2005;60:421-35. https://doi.org/10.1111/j.1398-9995.2005.00719.x
  3. Ross R, Everett NB, Tyler R. Wound healing and collagen formation. VI. The origin of the wound fibroblast studied in parabiosis. J Cell Biol 1970;44:645-54. https://doi.org/10.1083/jcb.44.3.645
  4. Bucala R, Spiegel LA, Chesney J, Hogan M, Cerami A. Circulating fibrocytes define a new leukocyte subpopulation that mediates tissue repair. Mol Med 1994;1:71-81.
  5. Lin SL, Kisseleva T, Brenner DA, Duffield JS. Pericytes and perivascular fibroblasts are the primary source of collagen-producing cells in obstructive fibrosis of the kidney. Am J Pathol 2008;173:1617-27. https://doi.org/10.2353/ajpath.2008.080433
  6. Hashimoto N, Phan SH, Imaizumi K, Matsuo M, Nakashima H, Kawabe T, et al. Endothelial-mesenchymal transition in bleomycin-induced pulmonary fibrosis. Am J Respir Cell Mol Biol 2010;43:161-72. https://doi.org/10.1165/rcmb.2009-0031OC
  7. Liu Y. Epithelial to mesenchymal transition in renal fibrogenesis: pathologic significance, molecular mechanism, and therapeutic intervention. J Am Soc Nephrol 2004;15:1-12. https://doi.org/10.1097/01.ASN.0000106015.29070.E7
  8. Kasai H, Allen JT, Mason RM, Kamimura T, Zhang Z. TGF-beta1 induces human alveolar epithelial to mesenchymal cell transition (EMT). Respir Res 2005;6:56. https://doi.org/10.1186/1465-9921-6-56
  9. Willis BC, duBois RM, Borok Z. Epithelial origin of myofibroblasts during fibrosis in the lung. Proc Am Thorac Soc 2006;3:377-82. https://doi.org/10.1513/pats.200601-004TK
  10. Zeisberg M, Yang C, Martino M, Duncan MB, Rieder F, Tanjore H, et al. Fibroblasts derive from hepatocytes in liver fibrosis via epithelial to mesenchymal transition. J Biol Chem 2007;282:23337-47. https://doi.org/10.1074/jbc.M700194200
  11. Willis BC, Liebler JM, Luby-Phelps K, Nicholson AG, Crandall ED, du Bois RM, et al. Induction of epithelial-mesenchymal transition in alveolar epithelial cells by transforming growth factor-beta1: potential role in idiopathic pulmonary fibrosis. Am J Pathol 2005;166:1321-32. https://doi.org/10.1016/S0002-9440(10)62351-6
  12. Khalil N, O'Connor RN, Unruh HW, Warren PW, Flanders KC, Kemp A, et al. Increased production and immunohistochemical localization of transforming growth factor-beta in idiopathic pulmonary fibrosis. Am J Respir Cell Mol Biol 1991;5:155-62. https://doi.org/10.1165/ajrcmb/5.2.155
  13. Idiopathic Pulmonary Fibrosis Clinical Research Network, Raghu G, Anstrom KJ, King TE Jr, Lasky JA, Martinez FJ. Prednisone, azathioprine, and N-acetylcysteine for pulmonary fibrosis. N Engl J Med 2012;366:1968-77. https://doi.org/10.1056/NEJMoa1113354
  14. Raghu G, Rochwerg B, Zhang Y, Garcia CA, Azuma A, Behr J, et al. An official ATS/ERS/JRS/ALAT clinical practice guideline: treatment of idiopathic pulmonary fibrosis: an update of the 2011 clinical practice guideline. Am J Respir Crit Care Med 2015;192:e3-19. https://doi.org/10.1164/rccm.201506-1063ST
  15. Kim TH, Lee YH, Kim KH, Lee SH, Cha JY, Shin EK, et al. Role of lung apolipoprotein A-I in idiopathic pulmonary fibrosis: antiinflammatory and antifibrotic effect on experimental lung injury and fibrosis. Am J Respir Crit Care Med 2010;182:633-42. https://doi.org/10.1164/rccm.200905-0659OC
  16. Lee E, Lee EJ, Kim H, Jang A, Koh E, Uh ST, et al. Overexpression of apolipoprotein A1 in the lung abrogates fibrosis in experimental silicosis. PLoS One 2013;8:e55827. https://doi.org/10.1371/journal.pone.0055827
  17. Heldin CH, Moustakas A. Role of Smads in TGFbeta signaling. Cell Tissue Res 2012;347:21-36. https://doi.org/10.1007/s00441-011-1190-x
  18. Mu Y, Gudey SK, Landstrom M. Non-Smad signaling pathways. Cell Tissue Res 2012;347:11-20. https://doi.org/10.1007/s00441-011-1201-y
  19. Heldin CH, Landstrom M, Moustakas A. Mechanism of TGF-beta signaling to growth arrest, apoptosis, and epithelial-mesenchymal transition. Curr Opin Cell Biol 2009;21:166-76. https://doi.org/10.1016/j.ceb.2009.01.021
  20. King TE Jr, Pardo A, Selman M. Idiopathic pulmonary fibrosis. Lancet 2011;378:1949-61. https://doi.org/10.1016/S0140-6736(11)60052-4
  21. Spagnolo P, Sverzellati N, Rossi G, Cavazza A, Tzouvelekis A, Crestani B, et al. Idiopathic pulmonary fibrosis: an update. Ann Med 2015;47:15-27. https://doi.org/10.3109/07853890.2014.982165
  22. Noguchi S, Yamauchi Y, Takizawa H. Novel therapeutic strategies for fibrotic lung disease: a review with a focus on epithelial-mesenchymal transition. Recent Pat Inflamm Allergy Drug Discov 2014;8:9-18. https://doi.org/10.2174/1872213X07666131229131451
  23. Bartis D, Mise N, Mahida RY, Eickelberg O, Thickett DR. Epithelial-mesenchymal transition in lung development and disease: does it exist and is it important? Thorax 2014;69:760-5. https://doi.org/10.1136/thoraxjnl-2013-204608
  24. Willis BC, Borok Z. TGF-beta-induced EMT: mechanisms and implications for fibrotic lung disease. Am J Physiol Lung Cell Mol Physiol 2007;293:L525-34. https://doi.org/10.1152/ajplung.00163.2007
  25. Derynck R, Zhang YE. Smad-dependent and Smad-independent pathways in TGF-beta family signalling. Nature 2003;425:577-84. https://doi.org/10.1038/nature02006
  26. Hayashida T, Decaestecker M, Schnaper HW. Cross-talk between ERK MAP kinase and Smad signaling pathways enhances TGF-beta-dependent responses in human mesangial cells. FASEB J 2003;17:1576-8. https://doi.org/10.1096/fj.03-0037fje
  27. 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. https://doi.org/10.3904/kjim.2014.29.3.281
  28. Scotton CJ, Chambers RC. Molecular targets in pulmonary fibrosis: the myofibroblast in focus. Chest 2007;132:1311-21. https://doi.org/10.1378/chest.06-2568
  29. Gharaee-Kermani M, Hu B, Phan SH, Gyetko MR. Recent advances in molecular targets and treatment of idiopathic pulmonary fibrosis: focus on TGFbeta signaling and the myofibroblast. Curr Med Chem 2009;16:1400-17. https://doi.org/10.2174/092986709787846497
  30. Gomer RH, Lupher ML Jr. Investigational approaches to therapies for idiopathic pulmonary fibrosis. Expert Opin Investig Drugs 2010;19:737-45. https://doi.org/10.1517/13543784.2010.484018
  31. Datta A, Scotton CJ, Chambers RC. Novel therapeutic approaches for pulmonary fibrosis. Br J Pharmacol 2011;163:141-72. https://doi.org/10.1111/j.1476-5381.2011.01247.x
  32. Rojas A, Padidam M, Cress D, Grady WM. TGF-beta receptor levels regulate the specificity of signaling pathway activation and biological effects of TGF-beta. Biochim Biophys Acta 2009;1793:1165-73. https://doi.org/10.1016/j.bbamcr.2009.02.001
  33. Sherman CB, Peterson SJ, Frishman WH. Apolipoprotein A-I mimetic peptides: a potential new therapy for the prevention of atherosclerosis. Cardiol Rev 2010;18:141-7. https://doi.org/10.1097/CRD.0b013e3181c4b508
  34. Osei-Hwedieh DO, Amar M, Sviridov D, Remaley AT. Apolipoprotein mimetic peptides: mechanisms of action as anti-atherogenic agents. Pharmacol Ther 2011;130:83-91. https://doi.org/10.1016/j.pharmthera.2010.12.003
  35. Park SW, Lee EH, Lee EJ, Kim HJ, Bae DJ, Han S, et al. Apolipoprotein A1 potentiates lipoxin A4 synthesis and recovery of allergen-induced disrupted tight junctions in the airway epithelium. Clin Exp Allergy 2013;43:914-27. https://doi.org/10.1111/cea.12143

피인용 문헌

  1. Hesperidin inhibits the epithelial to mesenchymal transition induced by transforming growth factor-β1 in A549 cells through Smad signaling in the cytoplasm vol.55, pp.None, 2016, https://doi.org/10.1590/s2175-97902019000218172
  2. TGF‐β1 mediated MAPK signaling pathway promotes collagen formation induced by Nano NiO in A549 cells vol.34, pp.6, 2016, https://doi.org/10.1002/tox.22738
  3. IL-37 Attenuates Lung Fibrosis by Inducing Autophagy and Regulating TGF-β1 Production in Mice vol.203, pp.8, 2016, https://doi.org/10.4049/jimmunol.1801515
  4. Anti-Fibrosis Effects of Magnesium Lithospermate B in Experimental Pulmonary Fibrosis: By Inhibiting TGF-βRI/Smad Signaling vol.26, pp.6, 2016, https://doi.org/10.3390/molecules26061715
  5. Serum levels of small HDL particles are negatively correlated with death or lung transplantation in an observational study of idiopathic pulmonary fibrosis vol.58, pp.6, 2016, https://doi.org/10.1183/13993003.04053-2020