DOI QR코드

DOI QR Code

The role of FGF-2 in smoke-induced emphysema and the therapeutic potential of recombinant FGF-2 in patients with COPD

  • Kim, You-Sun (Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine) ;
  • Hong, Goohyeon (Department of Internal Medicine, Dankook University College of Medicine) ;
  • Kim, Doh Hyung (Department of Internal Medicine, Dankook University College of Medicine) ;
  • Kim, Young Min (Department of Internal Medicine, Dankook University College of Medicine) ;
  • Kim, Yoon-Keun (Institute of MD Healthcare, Inc) ;
  • Oh, Yeon-Mok (Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine) ;
  • Jee, Young-Koo (Department of Internal Medicine, Dankook University College of Medicine)
  • Received : 2018.03.04
  • Accepted : 2018.09.04
  • Published : 2018.11.30

Abstract

Although the positive effects of recombinant fibroblast growth factor-2 (rFGF-2) in chronic obstructive pulmonary disease (COPD) have been implicated in previous studies, knowledge of its role in COPD remains limited. The mechanism of FGF2 in a COPD mouse model and the therapeutic potential of rFGF-2 were investigated in COPD. The mechanism and protective effects of rFGF-2 were evaluated in cigarette smoke-exposed or elastase-induced COPD animal models. Inflammation was assessed in alveolar cells and lung tissues from mice. FGF-2 was decreased in the lungs of cigarette smoke-exposed mice. Intranasal use of rFGF-2 significantly reduced macrophage-dominant inflammation and alveolar destruction in the lungs. In the elastase-induced emphysema model, rFGF-2 improved regeneration of the lungs. In humans, plasma FGF-2 was decreased significantly in COPD compared with normal subjects (10 subjects, P = 0.037). The safety and efficacy of inhaled rFGF-2 use was examined in COPD patients, along with changes in respiratory symptoms and pulmonary function. A 2-week treatment with inhaled rFGF-2 in COPD (n = 6) resulted in significantly improved respiratory symptoms compared with baseline levels (P < 0.05); however, the results were not significant compared with the placebo. The pulmonary function test results of COPD improved numerically compared with those in the placebo, but the difference was not statistically significant. No serious adverse events occurred during treatment with inhaled rFGF-2. The loss of FGF-2 production is an important mechanism in the development of COPD. Inhaling rFGF-2 may be a new therapeutic option for patients with COPD because rFGF-2 decreases inflammation in lungs exposed to cigarette smoke.

Keywords

Acknowledgement

Supported by : Ministry of Health and Welfare

References

  1. Global Strategy for the Diagnosis, Management and Prevention of COPD, Global Initiative for Chronic Obstructive Lung Disease (GOLD) 2018. Available at: http://www.goldcopd.org/.
  2. Wedzicha, J. A. et al. Indacaterol-glycopyrronium versus salmeterol-fluticasone for COPD. N. Engl. J. Med 374, 2222-2234 (2016). https://doi.org/10.1056/NEJMoa1516385
  3. Janson, C. et al. Pneumonia and pneumonia related mortality in patients with COPD treated with fixed combinations of inhaled corticosteroid and long acting ${\beta}$2 agonist: observational matched cohort study (PATHOS). BMJ 346, f3306 (2013). https://doi.org/10.1136/bmj.f3306
  4. Morse,D. & Rosas, I. O. Tobacco smoke-induced lung fibrosis and emphysema. Annu Rev. Physiol. 76, 493-513 (2014). https://doi.org/10.1146/annurev-physiol-021113-170411
  5. Talikka, M. et al. Genomic impact of cigarette smoke, with application to three smoking-related diseases. Crit. Rev. Toxicol. 42, 877-889 (2012). https://doi.org/10.3109/10408444.2012.725244
  6. MacNee, W. Oxidants/antioxidants and COPD. Chest 117(5Suppl 1), 303S-317SS (2000).
  7. Celedon, J. C. et al. The transforming growth factor-beta1 (TGFB1) gene is associated with chronic obstructive pulmonary disease (COPD). Hum. Mol. Genet 13, 1649-1656 (2004). https://doi.org/10.1093/hmg/ddh171
  8. Chen, P. Y. et al. Fibroblast growth factor (FGF) signaling regulates transforming growth factor beta (TGF${\beta}$)-dependent smooth muscle cell phenotype modulation. Sci. Rep. 6, 33407 (2016). https://doi.org/10.1038/srep33407
  9. Lee, B. J. et al. Protective effects of basic fibroblast growth factor in the development of emphysema induced by interferon-${\gamma}$. Exp. Mol. Med. 43, 169-178 (2011). https://doi.org/10.3858/emm.2011.43.4.018
  10. Bikfalvi, A., Klein, S., Pintucci, G. & Rifkin, D. B. Biological roles of fibroblast growth factor-2. Endocr. Rev. 18, 26-45 (1997).
  11. Nugent, M. A. & Iozzo, R. V. Fibroblast growth factor-2. Int. J. Biochem. Cell Biol. 32, 115-120 (2000). https://doi.org/10.1016/S1357-2725(99)00123-5
  12. Burgess, J. K. The role of the extracellular matrix and specific growth factors in the regulation of inflammation and remodelling in asthma. Pharmacol. Ther. 122, 19-29 (2009). https://doi.org/10.1016/j.pharmthera.2008.12.002
  13. Hoshino, M., Takahashi, M. & Aoike, N. Expression of vascular endothelial growth factor, basic fibroblast growth factor, and angiogenin immunoreactivity in asthmatic airways and its relationship to angiogenesis. J. Allergy Clin. Immunol. 107, 295-301 (2001). https://doi.org/10.1067/mai.2001.111928
  14. Redington, A. E. et al. Basic fibroblast growth factor in asthma: measurement in bronchoalveolar lavage fluid basally and following allergen challenge. J. Allergy Clin. Immunol. 107, 384-387 (2001). https://doi.org/10.1067/mai.2001.112268
  15. Shute, J. K. et al. Epithelial expression and release of FGF-2 from heparan sulphate binding sites in bronchial tissue in asthma. Thorax 59, 557-562 (2004). https://doi.org/10.1136/thx.2002.002626
  16. Chang, S. S., Yokomise, H., Matsuura, N., Gotoh, M. & Tabata, Y. Novel therapeutic approach for pulmonary emphysema using gelatin microspheres releasing basic fibroblast growth factor in a canine model. Surg. Today 44, 1536-1541 (2014). https://doi.org/10.1007/s00595-014-0864-x
  17. Morino, S. et al. Fibroblast growth factor-2 promotes recovery of pulmonary function in a canine models of elastase-induced emphysema. Exp. Lung Res. 33, 15-26 (2007). https://doi.org/10.1080/01902140601113070
  18. Morino, S. et al. Fibroblast growth factor-2 induces recovery of pulmonary blood flow in canine emphysema models. Chest 128, 920-926 (2005). https://doi.org/10.1378/chest.128.2.920
  19. Kawago, M. et al. Intrapleural administration of gelatin-embedded, sustainedrelease basic fibroblast growth factor for the regeneration of emphysematous lungs in rats. J. Thorac. Cardiovasc. Surg. 147, 1644-1649 (2014). https://doi.org/10.1016/j.jtcvs.2013.07.039
  20. Jeon, S. G. et al. Recombinant basic fibroblast growth factor inhibits the airway hyperresponsiveness, mucus production, and lung inflammation induced by an allergen challenge. J. Allergy Clin. Immunol. 119, 831-837 (2007). https://doi.org/10.1016/j.jaci.2006.12.653
  21. Kim, Y. S. et al. The safety and efficacy of recombinant fibroblast growth factor 2 in human asthmatics: a pilot study. Allergy Asthma Respir. Dis. 2, 200-207 (2014). https://doi.org/10.4168/aard.2014.2.3.200
  22. Huh, J. W. et al. Bone marrow cells repair cigarette smoke-induced emphysema in rats. Am. J. Physiol. Lung Cell. Mol. Physiol. 301, L255-L266 (2011). https://doi.org/10.1152/ajplung.00253.2010
  23. Kim, Y. S. et al. Extracellular vesicles, especially derived from Gram-negative bacteria, in indoor dust induce neutrophilic pulmonary inflammation associated with both Th1 and Th17 cell responses. Clin. Exp. Allergy 43, 443-454 (2013). https://doi.org/10.1111/cea.12085
  24. Lazarous, D. F. et al. Effects of chronic systemic administration of basic fibroblast growth factor on collateral development in the canine heart. Circulation 91, 145-153 (1995). https://doi.org/10.1161/01.CIR.91.1.145
  25. Schaper, W., De Brabander, M. & Lewi, P. DNA synthesis and mitoses in coronary collateral vessels of the dog. Circ. Res. 28, 671-679 (1971). https://doi.org/10.1161/01.RES.28.6.671
  26. Partridge, M. R. et al. Development and validation of the capacity of daily living during the morning questionnaire and the Global Chest Symptoms Questionnaire in COPD. Eur. Respir. J. 36, 96-104 (2010). https://doi.org/10.1183/09031936.00123709
  27. Lommatzsch, M. et al. Acute effects of tobacco smoke on human airway dendritic cells in vivo. Eur. Respir. J. 35, 1130-1136 (2010). https://doi.org/10.1183/09031936.00090109
  28. Costabel, U. & Guzman, J. Effect of smoking on bronchoalveolar lavage constituents. Eur. Respir. J. 5, 776-779 (1992).
  29. Kuschner, W. G., D'Alessandro, A., Wong, H. & Blanc, P. D. Dose-dependent cigarette smoking-related inflammatory responses in healthy adults. Eur. Respir. J. 9, 1989-1994 (1996). https://doi.org/10.1183/09031936.96.09101989
  30. Burke, W. M. et al. Smoking-induced changes in epithelial lining fluid volume, cell density and protein. Eur. Respir. J. 5, 780-784 (1992).
  31. Pardo, O. E. et al. Fibroblast growth factor-2 induces translational regulation of Bcl-XL and Bcl-2 via a MEK-dependent pathway: correlation with resistance to etoposide-induced apoptosis. J. Biol. Chem. 277, 12040-12046 (2002). https://doi.org/10.1074/jbc.M109006200
  32. Pardo, O. E. et al. Fibroblast growth factor 2-mediated translational control of IAPs blocks mitochondrial release of Smac/DIABLO and apoptosis in small cell lung cancer cells. Mol. Cell Biol. 23, 7600-7610 (2003). https://doi.org/10.1128/MCB.23.21.7600-7610.2003
  33. Aubry, M. C.,Wright, J. L. &Myers, J. L. The pathology of smoking related lung diseases. Clin. Chest Med. 21, 11-35 (2000). vii. https://doi.org/10.1016/S0272-5231(05)70005-8
  34. Sun, C. et al. LL-37 secreted by epithelium promotes fibroblast collagen production: a potential mechanism of small airway remodeling in chronic obstructive pulmonary disease. Lab. Invest. 94, 991-1002 (2014). https://doi.org/10.1038/labinvest.2014.86
  35. Kim, S. H. et al. Perceptions of severe asthma and asthma-COPD overlap syndrome among specialists: a questionnaire survey. Allergy Asthma Immunol. Res. 10, 225-235 (2018). https://doi.org/10.4168/aair.2018.10.3.225
  36. Krimmer, D. I., Burgess, J. K., Wooi, T. K., Black, J. L. & Oliver, B. G.Matrix proteins from smoke-exposed fibroblasts are pro-proliferative. Am. J. Respir. Cell Mol. Biol. 46, 34-39 (2012). https://doi.org/10.1165/rcmb.2010-0426OC
  37. Hogg, J. C. et al. The nature of small-airway obstruction in chronic obstructive pulmonary disease. N. Engl. J. Med. 350, 2645-2653 (2004). https://doi.org/10.1056/NEJMoa032158
  38. James, A. L. & Wenzel, S. Clinical relevance of airway remodelling in airway diseases. Eur. Respir. J. 30, 134-155 (2007). https://doi.org/10.1183/09031936.00146905
  39. Turner, N. & Grose, R. Fibroblast growth factor signalling: from development to cancer. Nat. Rev. Cancer 10, 116-129 (2010). https://doi.org/10.1038/nrc2780

Cited by

  1. Adverse early-life environment impairs postnatal lung development in mice vol.51, pp.9, 2018, https://doi.org/10.1152/physiolgenomics.00016.2019
  2. Mesenchymal WNT-5A/5B Signaling Represses Lung Alveolar Epithelial Progenitors vol.8, pp.10, 2019, https://doi.org/10.3390/cells8101147
  3. FGF2, an Immunomodulatory Factor in Asthma and Chronic Obstructive Pulmonary Disease (COPD) vol.8, pp.None, 2018, https://doi.org/10.3389/fcell.2020.00223
  4. Rho-Kinase 1/2 Inhibition Prevents Transforming Growth Factor-β-Induced Effects on Pulmonary Remodeling and Repair vol.11, pp.None, 2018, https://doi.org/10.3389/fphar.2020.609509
  5. FGF/FGFR signaling: From lung development to respiratory diseases vol.62, pp.None, 2021, https://doi.org/10.1016/j.cytogfr.2021.09.002