Anti-inflammatory effect of Yu-Ping-Feng-San via TGF-β1 signaling suppression in rat model of COPD

Document Type : Original Article

Authors

1 Faculty of Basic Medical Science, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan

2 Central laboratory, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan

Abstract

Objective(s): Yu-Ping-Feng-San (YPFS) is a classical traditional Chinese medicine that is widely used for treatment of the diseases in respiratory systems, including chronic obstructive pulmonary disease (COPD) recognized as chronic inflammatory disease. However, the molecular mechanism remains unclear. Here we detected the factors involved in transforming growth factor beta 1 (TGF-β1)/Smad2 signaling pathway and inflammatory cytokines, to clarify whether YPFS could attenuate inflammatory response dependent on TGF-β1/Smad2 signaling in COPD rats or cigarette smoke extract (CSE)-treated human bronchial epithelial (Beas-2B) cells. 
Materials and Methods: The COPD rat model was established by exposure to cigarette smoke and intratracheal instillation of lipopolysaccharide, YPFS was administered to the animals. The efficacy of YPFS was evaluated by comparing the severity of pulmonary pathological damage, pro-inflammation cytokines, collagen related genes and the activation of TGF-β1/Smad2 signaling pathway. Furthermore, CSE-treated cells were employed to confirm whether the effect of YPFS was dependent on the TGF-β1/Smad2 signaling via knockdown Smad2 (Si-RNA), or pretreatment with the inhibitor of TGF-β1.
Results: Administration of YPFS effectively alleviated injury of lung, suppressed releasing of pro-inflammatory cytokines and collagen deposition in COPD animals (P<0.05), whereas exogenous TGF-β1 promoted releasing of IL-1β, IL-6, TNFα (P<0.05). Administration YPFS reduced inflammatory response significantly, also down-regulated TGF-β1/Smad2 signaling in vivo and in vitro. Unexpectedly, knockdown Smad2 or inhibition of TGF-β1 abolished anti-inflammatory effect of YPFS in CSE-treated cells.
Conclusion: YPFS accomplished anti-inflammatory effects mainly by suppressing phosphorylation of Smad2, TGF-β1/Smad2 signaling pathway was required for YPFS-mediated anti-inflammation in COPD rats or CSE-treated Beas-2B cells.

Keywords


1.Hagstad S, Bjerg A, Ekerljung L, Backman H, Lindberg A, Rönmark E, et al. Passive smoking exposure is associated with increased risk of COPD in never smokers. Chest 2014; 145:1298-1304.
2.Ford ES, Murphy LB, Khavjou O, Giles WH, Holt JB, Croft JB. Total and state-specific medical and absenteeism costs of COPD among adults aged≥ 18 years in the United States for 2010 and projections through 2020. Chest 2015; 147:31-45.
3.Sethi S, Mahler DA, Marcus P, Owen CA, Yawn B, Rennard S. Inflammation in COPD: implications for management. Am J Med 2012; 125:1162-1270.
4.Chung K, Adcock I. Multifaceted mechanisms in COPD: inflammation, immunity, and tissue repair and destruction. Eur Respir J 2008; 31:1334-1356.
5.Jiménez-Ruiz CA, Andreas S, Lewis KE, Tonnesen P, van Schayck C, Hajek P, et al. Statement on smoking cessation in COPD and other pulmonary diseases and in smokers with comorbidities who find it difficult to quit. Eur Respir J 2015;46:61-79.
6.Shah S, Gangan N, Bechtol R, Vaidya V. Smoking In Chronic Obstructive Pulmonary Disease (Copd) Patients: Socio-Demographic Factors Associated With Smoking Cessation. Value Health 2013; 16:A240.
7.Rabe KF, Bateman ED, O'Donnell D, Witte S, Bredenbröker D, Bethke TD. Roflumilast-an oral anti-inflammatory treatment for chronic obstructive pulmonary disease: a randomised controlled trial. Lancet 2005; 366:563-571.
8.Günter S, John-Schuster G, Conlon TM, Hager K, Amarie OV, Eickelberg O, et al. Immunoaging augments sensitivity to cigarette smoke-induced COPD. Eur Respir J 2014; 44:3248.
9.Leila GM, Mohammad Hossein B, Mohammad RN. The effect of Zataria multiflora and its constituent, carvacrol, on tracheal responsiveness and lung pathology in guinea pig model of COPD. Phytother Res 2015; 29:730–736.
10. Mohammad Hossein B, Leila GM. Lung inflammation changes and oxidative stress induced by cigarette smoke exposure in guinea pigs affected by Zataria multiflora and its constituent, carvacrol. BMC Complement Altern Med 2015;15:1-10.
11. Brandsma C-A, Timens W, Jonker MR, Rutgers B, Noordhoek JA, Postma DS. Differential effects of fluticasone on extracellular matrix production by airway and parenchymal fibroblasts in severe COPD. Am J Physiol Lung Cell Mol Physiol 2013;305:582-589.
12. Königshoff M, Kramer M, Balsara N, Wilhelm J, Amarie OV, Jahn A, et al. WNT1-inducible signaling protein–1 mediates pulmonary fibrosis in mice and is upregulated in humans with idiopathic pulmonary fibrosis. J Clin Invest 2009; 119:772-787.
13. Wang B, Komers R, Carew R, Winbanks CE, Xu B, Herman-Edelstein M, et al. Suppression of microRNA-29 expression by TGF-β1 promotes collagen expression and renal fibrosis. J Am Soc Nephrol 2012; 23:252-265.
14. Su Zg, Wen Fq, Feng Yl, Xiao M, Wu Xl. Transforming growth factor-β1 gene polymorphisms associated with chronic obstructive pulmonary disease in Chinese population. Acta Pharmacol Sin 2005; 26:714-720.
15. Ichimaru Y, Krimmer DI, Burgess JK, Black JL, Oliver BG. TGF-β enhances deposition of perlecan from COPD airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2012; 302:325-333.
16. Warburton D. Developmental responses to lung injury: repair or fibrosis. Fibrogenesis Tissue Repair 2012; 5:S2.
17. Kitamura H, Cambier S, Somanath S, Barker T, Minagawa S, Markovics J, et al. Mouse and human lung fibroblasts regulate dendritic cell trafficking, airway inflammation, and fibrosis through integrin αvβ8–mediated activation of TGF-β.  J Clin Invest 2011; 121:2863-2875.
18. Yoshimura A, Wakabayashi Y, Mori T. Cellular and molecular basis for the regulation of inflammation by TGF-β. J Biochem 2010; 147:781-792.
19. Sharkey DJ, Tremellen KP, Jasper MJ, Gemzell-Danielsson K, Robertson SA. Seminal fluid induces leukocyte recruitment and cytokine and chemokine mRNA expression in the human cervix after coitus. J Immunol 2012; 188:2445-2454.
20. Hovhannisyan L, Mkrtchyan G, Boyajyan A, Avetyan D, Tadevosyan МY SS. Inflammatory markers in post-traumatic stress disorder. Cytokines Inflammation. 2012; 11:42-45.
21. Han G, Li F, Singh TP, Wolf P, Wang XJ. The pro-inflammatory role of TGFβ1: a paradox? Int J Biol Sci 2012;8:228-235.
22. Li T, Wang Y, Wang Y, Liang R, Zhang D, Zhang H, et al. Development of an SPE–HPLC–MS method for simultaneous determination and pharmacokinetic study of bioactive constituents of Yu Ping Feng San in rat plasma after oral administration. J Ethnopharmacol 2013;145:784-92.
23. Liao Yl, An W, Zhang W. Yu ping feng san auxiliary treatment of 30 cases of chronic obstructive pulmonary disease. Herald Med. 2011;30:63-66.
24. Ma YT, Wang DH. Yu Ping Feng San stabilization treatment of chronic obstructive pulmonary disease pulmonary qi deficiency syndrome curative effect observation. Modern Traditional Chinese Med 2009:6-8.
25. Wang CM, Jiang M, Wang HJ. Effect of NF-κB inhibitor on high‑mobility group protein B1 expression in a COPD rat model. Mol Med Rep 2013; 7:499-502.
26. Zheng H, Liu Y, Tian H, Fang Z, Li G, He S. Development and characterization of a rat model of chronic obstructive pulmonary disease (COPD) induced by sidestream cigarette smoke. Toxicol Lett 2009; 189:225-234.
27. Du CY, Choi RC, Zheng KY, Dong TT, Lau DT, Tsim KW. Yu Ping Feng San, an ancient Chinese herbal decoction containing Astragali radix, Atractylodis macrocephalae Rhizoma and Saposhnikoviae radix, regulates the release of cytokines in murine macrophages. PLoS One 2013;8: e78622.
28. Shaw LH, Lin LC, Tsai TH. HPLC–MS/MS analysis of a traditional Chinese medical formulation of Bu-Yang-Huan-Wu-Tang and its pharmacokinetics after oral administration to rats. PLoS One 2012;7:e43848.
29. Zhao HM, Huang XY, Zhou F, Tong WT, Wan PT, Huang MF, et al. Si Shen Wan inhibits mRNA expression of apoptosis-related molecules in p38 MAPK signal pathway in mice with colitis. Evid Based Complement Alternat Med 2013; 2013:432097.
30. Tony P, Sarah LP, Daniel P, Monique LVH. Cigarette smoke extract induces differential expression levels of beta-defensin peptides in human alveolar epithelial cells. Tob Induc Dis 2013; 11:10.
31. Kothari HPS. Immune-oxidative alterations in cultured human lymphocytes by cigarette smoke.Global J Multidisciplinary study. Global J of Multidisciplinary Studies 2015; 4:281-290.
32. Munder A, Wölbeling F, Kerber-Momot T, Wedekind D, Baumann U, Gulbins E, et al. Acute intratracheal Pseudomonas aeruginosa infection in cystic fibrosis mice is age-independent. Respir Res 2011; 12:148.
33. Persson C. Simvastatin inhibits smoke-induced airway epithelial injury: implications for COPD therapy. Eur Respir J 2014;43:1208-1211.
34. Gorowiec MR, Borthwick LA, Parker SM, Kirby JA, Saretzki GC, Fisher AJ. Free radical generation induces epithelial-to-mesenchymal transition in lung epithelium via a TGF-β1-dependent mechanism. Free Radic Biol Med 2012; 52:1024-1032.
35. Takizawa H, Tanaka M, Takami K, Ohtoshi T, Ito K, Satoh M, et al. Increased expression of transforming growth factor-β 1 in small airway epithelium from tobacco smokers and patients with chronic obstructive pulmonary disease (COPD). Am J Respir Criti Care Med 2001;163:1476-1483.
36. Kranenburg AR, Willems-Widyastuti A, Mooi WJ, Sterk PJ, Alagappan VK, de Boer WI, et al. Enhanced bronchial expression of extracellular matrix proteins in chronic obstructive pulmonary disease. Am J Clin Pathol 2006; 126:725-735.
37. Gu j, Jiang J, Shen C, Lu L, Dai Q, Effects of‘‘Jude-Screen Powder’’on Th1/Th2 balance in mice model of systemic allergic airway disease. ShangHai J Traditional Chinese Med 2005:50-52.
38. Shen D, Xie X, Zhu Z, Yu X, Liu H, Wang H, et al. Screening active components from Yu-Ping-Feng-San for regulating initiative key factors in allergic sensitization. PLoS One 2014; 9:e107279.
39. Du CYQ, Zheng KYZ, Bi CWC, Dong TTX, Lin HL, Tsim KWK. Yu Ping Feng San, an Ancient Chinese herbal decoction, Induces gene expression of anti-viral proteins and inhibits neuraminidase activity. Phytother Res 2015; 29:656-661.
40. Mohammad Hossein B, Leila GM. Effect of the Zataria multiflora on systemic inflammation of experimental animals model of COPD. BioMed Res Int 2014; 2014:802189.
41. Gholamnezhad Z, Mohammad Hossein B, Hosseini M. Effect of Nigella sativa on immune response in treadmill exercised rat. BMC Complement Altern Med 2014; 14:437.
42. Leila GM, Feizpour A, Kianmehr M, Soukhtanloo M, Mohammad Hossein B. The effect of carvacrol on systemic inflammation in guinea pigs model of COPD induced by cigarette smoke exposure. Pharmacol Rep 2015; 67:140-145.
43. Gu S, Yin N, Pei J, Lai L. Understanding traditional Chinese medicine anti-inflammatory herbal formulae by simulating their regulatory functions in the human arachidonic acid metabolic network. Mol Biosyst 2013; 9:1931-1938.
44. Kuzubova N, Gichkin A, Surkova E, Titova O. Left ventricular dysfunction in patients with COPD: Inflammation and endothelial dysfunction. Eur Respir J 2013; 42:178.
45. Yanbaeva DG, Dentener MA, Creutzberg EC, Wouters EF. Systemic inflammation in COPD: is genetic susceptibility a key factor? COPD 2006; 3:51-61.
46. Barnes PJ. Immunology of asthma and chronic obstructive pulmonary disease. Nat Rev Immunol. 2008; 8:183-192.
47. Calabrese F, Baraldo S, Bazzan E, Lunardi F, Rea F, Maestrelli P, et al. IL-32, a novel proinflammatory cytokine in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2008; 178:894-901.
48. Profita M, Chiappara G, Mirabella F, Di Giorgi R, Chimenti L, Costanzo G, et al. Effect of cilomilast (Ariflo) on TNF-α, IL-8, and GM-CSF release by airway cells of patients with COPD. Thorax 2003; 58:573-579.
49. O’Leary L, Tildy B, Papazoglou E, Adcock I, Chung K, Perry M. S50 Airway smooth muscle inflammation is controlled by microrna-145 targeting of smad3 in COPD. Thorax. 2014; 69:A28.
50. Cazzola M, Calzetta L, Bettoncelli G, Cricelli C, Romeo F, Matera MG, et al. Cardiovascular disease in asthma and COPD: a population-based retrospective cross-sectional study. Respir Med 2012; 106:249-256.
51. Sanchez-Muñoz F, Dominguez-Lopez A, Yamamoto-Furusho JK. Role of cytokines in inflammatory bowel disease. World J Gastroenterol 2008;14:4280-4288.
52. Baumgart DC, Carding SR. Inflammatory bowel disease: cause and immunobiology. Lancet 2007;369:1627-1640.
53. Feizpour A, Mohammad Hossein B, Ghorbani A. Adipose-derived stromal cell therapy affects lung inflammation and tracheal responsiveness in guinea pig model of COPD. PLoS One 2014; 9:e108974.
54. Bonniaud P, Margetts PJ, Kolb M, Schroeder JA, Kapoun AM, Damm D, et al. Progressive transforming growth factor β1–induced lung fibrosis is blocked by an orally active ALK5 kinase inhibitor. Am J Respir Crit Care Med 2005;171:889-898.
55. Rosendahl A, Checchin D, Fehniger TE, ten Dijke P, Heldin C-H, Sideras P. Activation of the TGF-β/activin-Smad2 pathway during allergic airway inflammation. Am J Respir Cell Mol Biol 2001; 25:60-68.
56. Biernacka A, Dobaczewski M, Frangogiannis NG. TGF-β signaling in fibrosis. Growth factors. 2011; 29:196-202.
57. Lan HY. Diverse roles of TGF-β/Smads in renal fibrosis and inflammation. Int J Biol Sci 2011; 7:1056-1067.
58. Chung AC, Huang XR, Zhou L, Heuchel R, Lai KN, Lan HY. Disruption of the Smad7 gene promotes renal fibrosis and inflammation in unilateral ureteral obstruction (UUO) in mice. Nephrol Dial Transplant 2009; 24:1443-1454.
59. Gregory LG, Mathie SA, Walker SA, Pegorier S, Jones CP, Lloyd CM. Overexpression of Smad2 drives house dust mite–mediated airway remodeling and airway hyperresponsiveness via activin and IL-25. Am J Respir Crit Care Med 2010; 182:143-54.