바로가기메뉴

본문 바로가기 주메뉴 바로가기

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

Tuberculosis & Respiratory Diseases / Tuberculosis & Respiratory Diseases,
2016, v.79 no.3, pp.143-152












  • Downloaded
  • Viewed

Abstract

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 β1 (TGF-β1)–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-β1–induced EMT in experimental lung fibrosis and clarify its mechanism of action. Methods: The A549 alveolar epithelial cell line was treated with TGF-β1 with or without ApoA1. Morphological changes and expression of EMT-related markers, including E-cadherin, N-cadherin, and α-smooth muscle actin were evaluated. Expressions of Smad and non-Smad mediators and TGF-β1 receptor type 1 (TβRI) and type 2 (TβRII) were measured. The silica-induced lung fibrosis model was established using ApoA1 overexpressing transgenic mice. Results: TGF-β1–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-β1–induced change of the EMT phenotype. ApoA1 inhibited the TGF-β1–induced increase in the phosphorylation of Smad2 and 3 as well as that of ERK and p38 mitogenactivated protein kinase mediators. In addition, ApoA1 reduced the TGF-β1–induced increase in TβRI and TβRII 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-β1–induced EMT in experimental lung fibrosis.

keywords
Apolipoprotein A-1, Transforming Growth Factor Beta1, Epithelial-Mesenchymal Transition, Pulmonary Fibrosis

Reference

1.

1. Selman M, Pardo A. Idiopathic pulmonary fibrosis: an epithelial/ fibroblastic cross-talk disorder. Respir Res 2002;3:3.

2.

2. Harari S, Caminati A. Idiopathic pulmonary fibrosis. Allergy 2005;60:421-35.

3.

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.

4.

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.

5. Lin SL, Kisseleva T, Brenner DA, Duffield JS. Pericytes and perivascular fibroblasts are the primary source of collagenproducing cells in obstructive fibrosis of the kidney. Am J Pathol 2008;173:1617-27.

6.

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.

7.

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.

8.

8. Kasai H, Allen JT, Mason RM, Kamimura T, Zhang Z. TGFbeta1 induces human alveolar epithelial to mesenchymal cell transition (EMT). Respir Res 2005;6:56.

9.

9. Willis BC, duBois RM, Borok Z. Epithelial origin of myofibroblasts during fibrosis in the lung. Proc Am Thorac Soc 2006;3: 377-82.

10.

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.

11.

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.

12.

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.

13.

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.

14.

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.

15.

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.

16.

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.

17.

17. Heldin CH, Moustakas A. Role of Smads in TGFbeta signaling. Cell Tissue Res 2012;347:21-36.

18.

18. Mu Y, Gudey SK, Landstrom M. Non-Smad signaling pathways. Cell Tissue Res 2012;347:11-20.

19.

19. Heldin CH, Landstrom M, Moustakas A. Mechanism of TGFbeta signaling to growth arrest, apoptosis, and epithelialmesenchymal transition. Curr Opin Cell Biol 2009;21:166-76.

20.

20. King TE Jr, Pardo A, Selman M. Idiopathic pulmonary fibrosis. Lancet 2011;378:1949-61.

21.

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.

22.

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.

23.

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.

24.

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.

25.

25. Derynck R, Zhang YE. Smad-dependent and Smad-independent pathways in TGF-beta family signalling. Nature 2003; 425:577-84.

26.

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.

27.

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.

28.

28. Scotton CJ, Chambers RC. Molecular targets in pulmonary fibrosis: the myofibroblast in focus. Chest 2007;132:1311-21.

29.

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.

30.

30. Gomer RH, Lupher ML Jr. Investigational approaches to therapies for idiopathic pulmonary fibrosis. Expert Opin Investig Drugs 2010;19:737-45.

31.

31. Datta A, Scotton CJ, Chambers RC. Novel therapeutic approaches for pulmonary fibrosis. Br J Pharmacol 2011;163:141- 72.

32.

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.

33.

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.

34.

34. Osei-Hwedieh DO, Amar M, Sviridov D, Remaley AT. Apolipoprotein mimetic peptides: mechanisms of action as antiatherogenic agents. Pharmacol Ther 2011;130:83-91.

35.

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.

Tuberculosis & Respiratory Diseases