Yeungnam Univ J Med.  2020 Oct;37(4):269-276. 10.12701/yujm.2020.00458.

Therapeutic potential of targeting kinase inhibition in patients with idiopathic pulmonary fibrosis

Affiliations
  • 1Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu, Korea
  • 2Department of Pharmacology, Yeungnam University College of Medicine, Daegu, Korea
  • 3Department of Microbiology, Ewha Womans University College of Medicine, Seoul, Korea

Abstract

Fibrosis is characterized by excessive accumulation of extracellular matrix components. The fibrotic process ultimately leads to organ dysfunction and failure in chronic inflammatory and metabolic diseases such as pulmonary fibrosis, advanced kidney disease, and liver cirrhosis. Idiopathic pulmonary fibrosis (IPF) is a common form of progressive and chronic interstitial lung disease of unknown etiology. Pathophysiologically, the parenchyma of the lung alveoli, interstitium, and capillary endothelium becomes scarred and stiff, which makes breathing difficult because the lungs have to work harder to transfer oxygen and carbon dioxide between the alveolar space and bloodstream. The transforming growth factor beta (TGF-) signaling pathway plays an important role in the pathogenesis of pulmonary fibrosis and scarring of the lung tissue. Recent clinical trials focused on the development of pharmacological agents that either directly or indirectly target kinases for the treatment of IPF. Therefore, to develop therapeutic targets for pulmonary fibrosis, it is essential to understand the key factors involved in the pathogenesis of pulmonary fibrosis and the underlying signaling pathway. The objective of this review is to discuss the role of kinase signaling cascades in the regulation of either TGF--dependent or other signaling pathways, including Rho-associated coiled-coil kinase, c-jun N-terminal kinase, extracellular signal-regulated kinase 5, and p90 ribosomal S6 kinase pathways, and potential therapeutic targets in IPF.

Keyword

Fibrosis; Idiopathic pulmonary fibrosis; Kinase; Transforming growth factor beta

Figure

  • Fig. 1. Roles of extracellular signal-regulated kinase 5 (ERK5) and p90 ribosomal S6 kinase (p90RSK) in pulmonary fibrosis. (A) ERK5 may be activated by multiple receptors involved in pulmonary fibrosis. (B) Transforming growth factor beta (TGF-β) activates ERK5 and p90RSK in lung alveolar epithelial cells and lung fibroblasts. Activation of ERK5 or p90RSK regulates Smad3 transcriptional activity via acetylation modification. The pharmacological inhibitors of ERK5 or p90RSK reduce TGF-β-induced fibrogenic gene expression and experimental lung fibrosis. TGFR, transforming growth factor receptor; FGFR, fibroblast growth factor receptor; PDGFR, platelet-derived growth factor receptor; EGFR, epidermal growth factor receptor; VEGFR, vascular endothelial growth factor receptor; P, phosphorylation; A, acetylation; HAT, histone acetyltransferase.


Cited by  1 articles

Advances in the science and treatment of respiratory diseases
Jin Hong Chung
Yeungnam Univ J Med. 2020;37(4):251-252.    doi: 10.12701/yujm.2020.00661.


Reference

References

1. Bonnans C, Chou J, Werb Z. Remodelling the extracellular matrix in development and disease. Nat Rev Mol Cell Biol. 2014; 15:786–801.
Article
2. Iredale JP, Thompson A, Henderson NC. Extracellular matrix degradation in liver fibrosis: biochemistry and regulation. Biochim Biophys Acta. 2013; 1832:876–83.
Article
3. Borthwick LA, Wynn TA, Fisher AJ. Cytokine mediated tissue fibrosis. Biochim Biophys Acta. 2013; 1832:1049–60.
Article
4. Liu RM, Gaston Pravia KA. Oxidative stress and glutathione in TGF-beta-mediated fibrogenesis. Free Radic Biol Med. 2010; 48:1–15.
5. Rockey DC, Bell PD, Hill JA. Fibrosis: a common pathway to organ injury and failure. N Engl J Med. 2015; 372:1138–49.
Article
6. King TE Jr. Clinical advances in the diagnosis and therapy of the interstitial lung diseases. Am J Respir Crit Care Med. 2005; 172:268–79.
Article
7. Strongman H, Kausar I, Maher TM. Incidence, prevalence, and survival of patients with idiopathic pulmonary fibrosis in the UK. Adv Ther. 2018; 35:724–36.
Article
8. Hopkins RB, Burke N, Fell C, Dion G, Kolb M. Epidemiology and survival of idiopathic pulmonary fibrosis from national data in Canada. Eur Respir J. 2016; 48:187–95.
Article
9. Raghu G. Idiopathic pulmonary fibrosis: lessons from clinical trials over the past 25 years. Eur Respir J. 2017; 50:1701209.
10. Noble PW, Homer RJ. Idiopathic pulmonary fibrosis: new insights into pathogenesis. Clin Chest Med. 2004; 25:749–58.
Article
11. Thannickal VJ, Toews GB, White ES, Lynch JP 3rd, Martinez FJ. Mechanisms of pulmonary fibrosis. Annu Rev Med. 2004; 55:395–417.
Article
12. Saito A, Horie M, Nagase T. TGF-β signaling in lung health and disease. Int J Mol Sci. 2018; 19:2460.
Article
13. Flanders KC. Smad3 as a mediator of the fibrotic response. Int J Exp Pathol. 2004; 85:47–64.
Article
14. Nakerakanti S, Trojanowska M. The role of TGF-β receptors in fibrosis. Open Rheumatol J. 2012; 6:156–62.
Article
15. Gordon KJ, Blobe GC. Role of transforming growth factor-beta superfamily signaling pathways in human disease. Biochim Biophys Acta. 2008; 1782:197–228.
Article
16. Hoyt DG, Lazo JS. Alterations in pulmonary mRNA encoding procollagens, fibronectin and transforming growth factor-beta precede bleomycin-induced pulmonary fibrosis in mice. J Pharmacol Exp Ther. 1988; 246:765–71.
17. Broekelmann TJ, Limper AH, Colby TV, McDonald JA. Transforming growth factor beta 1 is present at sites of extracellular matrix gene expression in human pulmonary fibrosis. Proc Natl Acad Sci U S A. 1991; 88:6642–6.
Article
18. Wu CF, Chiang WC, Lai CF, Chang FC, Chen YT, Chou YH, et al. Transforming growth factor β-1 stimulates profibrotic epithelial signaling to activate pericyte-myofibroblast transition in obstructive kidney fibrosis. Am J Pathol. 2013; 182:118–31.
Article
19. Border WA, Noble NA, Yamamoto T, Harper JR, Yamaguchi Yu, Pierschbacher MD, et al. Natural inhibitor of transforming growth factor-beta protects against scarring in experimental kidney disease. Nature. 1992; 360:361–4.
Article
20. Isaka Y, Tsujie M, Ando Y, Nakamura H, Kaneda Y, Imai E, et al. Transforming growth factor-beta 1 antisense oligodeoxynucleotides block interstitial fibrosis in unilateral ureteral obstruction. Kidney Int. 2000; 58:1885–92.
21. Takabatake Y, Isaka Y, Mizui M, Kawachi H, Shimizu F, Ito T, et al. Exploring RNA interference as a therapeutic strategy for renal disease. Gene Ther. 2005; 12:965–73.
Article
22. Zhao J, Shi W, Wang YL, Chen H, Bringas P Jr, Datto MB, et al. Smad3 deficiency attenuates bleomycin-induced pulmonary fibrosis in mice. Am J Physiol Lung Cell Mol Physiol. 2002; 282:L585–93.
23. Zi Z, Chapnick DA, Liu X. Dynamics of TGF-β/Smad signaling. FEBS Lett. 2012; 586:1921–8.
Article
24. Zhang YE. Non-Smad pathways in TGF-beta signaling. Cell Res. 2009; 19:128–39.
25. Bringardner BD, Baran CP, Eubank TD, Marsh CB. The role of inflammation in the pathogenesis of idiopathic pulmonary fibrosis. Antioxid Redox Signal. 2008; 10:287–301.
Article
26. Selman M, Pardo A. Idiopathic pulmonary fibrosis: an epithelial/fibroblastic cross-talk disorder. Respir Res. 2002; 3:3.
Article
27. Sakai N, Tager AM. Fibrosis of two: epithelial cell-fibroblast interactions in pulmonary fibrosis. Biochim Biophys Acta. 2013; 1832:911–21.
Article
28. Camelo A, Dunmore R, Sleeman MA, Clarke DL. The epithelium in idiopathic pulmonary fibrosis: breaking the barrier. Front Pharmacol. 2014; 4:173.
Article
29. Richeldi L, du Bois RM, Raghu G, Azuma A, Brown KK, Costabel U, et al. Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. N Engl J Med. 2014; 370:2071–82.
Article
30. Ogura T, Taniguchi H, Azuma A, Inoue Y, Kondoh Y, Hasegawa Y, et al. Safety and pharmacokinetics of nintedanib and pirfenidone in idiopathic pulmonary fibrosis. Eur Respir J. 2015; 45:1382–92.
Article
31. Taniguchi H, Ebina M, Kondoh Y, Ogura T, Azuma A, Suga M, et al. Pirfenidone in idiopathic pulmonary fibrosis. Eur Respir J. 2010; 35:821–9.
Article
32. Gurujeyalakshmi G, Hollinger MA, Giri SN. Pirfenidone inhibits PDGF isoforms in bleomycin hamster model of lung fibrosis at the translational level. Am J Physiol. 1999; 276:L311–8.
33. Iyer SN, Gurujeyalakshmi G, Giri SN. Effects of pirfenidone on transforming growth factor-beta gene expression at the transcriptional level in bleomycin hamster model of lung fibrosis. J Pharmacol Exp Ther. 1999; 291:367–73.
34. Noble PW, Albera C, Bradford WZ, Costabel U, Glassberg MK, Kardatzke D, et al. Pirfenidone in patients with idiopathic pulmonary fibrosis (CAPACITY): two randomised trials. Lancet. 2011; 377:1760–9.
Article
35. Lopez-de la Mora DA, Sanchez-Roque C, Montoya-Buelna M, Sanchez-Enriquez S, Lucano-Landeros S, Macias-Barragan J, et al. Role and new insights of pirfenidone in fibrotic diseases. Int J Med Sci. 2015; 12:840–7.
Article
36. King TE Jr, Bradford WZ, Castro-Bernardini S, Fagan EA, Glaspole I, Glassberg MK, et al. A phase 3 trial of pirfenidone in patients with idiopathic pulmonary fibrosis. N Engl J Med. 2014; 370:2083–92.
Article
37. Jiang C, Huang H, Liu J, Wang Y, Lu Z, Xu Z. Adverse events of pirfenidone for the treatment of pulmonary fibrosis: a meta-analysis of randomized controlled trials. PLoS One. 2012; 7:e47024.
Article
38. Wollin L, Wex E, Pautsch A, Schnapp G, Hostettler KE, Stowasser S, et al. Mode of action of nintedanib in the treatment of idiopathic pulmonary fibrosis. Eur Respir J. 2015; 45:1434–45.
Article
39. Hostettler KE, Zhong J, Papakonstantinou E, Karakiulakis G, Tamm M, Seidel P, et al. Anti-fibrotic effects of nintedanib in lung fibroblasts derived from patients with idiopathic pulmonary fibrosis. Respir Res. 2014; 15:157.
Article
40. Wollin L, Maillet I, Quesniaux V, Holweg A, Ryffel B. Antifibrotic and anti-inflammatory activity of the tyrosine kinase inhibitor nintedanib in experimental models of lung fibrosis. J Pharmacol Exp Ther. 2014; 349:209–20.
Article
41. Richeldi L, Cottin V, Flaherty KR, Kolb M, Inoue Y, Raghu G, et al. Design of the INPULSIS™ trials: two phase 3 trials of nintedanib in patients with idiopathic pulmonary fibrosis. Respir Med. 2014; 108:1023–30.
Article
42. Meng XM, Nikolic-Paterson DJ, Lan HY. TGF-β: the master regulator of fibrosis. Nat Rev Nephrol. 2016; 12:325–38.
Article
43. Choi ME. Mechanism of transforming growth factor-β1 signaling: role of the mitogen-activated protein kinase. Kidney Int. 2000; 58(Suppl 77):S53–8.
Article
44. Leask A, Abraham DJ. All in the CCN family: essential matricellular signaling modulators emerge from the bunker. J Cell Sci. 2006; 119(Pt 23):4803–10.
Article
45. Yue X, Shan B, Lasky JA. TGF-β: titan of lung fibrogenesis. Curr Enzym Inhib. 2010; 6:10.2174/10067.
46. Varga J, Abraham D. Systemic sclerosis: a prototypic multisystem fibrotic disorder. J Clin Invest. 2007; 117:557–67.
Article
47. Derynck R, Zhang YE. Smad-dependent and Smad-independent pathways in TGF-beta family signalling. Nature. 2003; 425:577–84.
Article
48. Das F, Ghosh-Choudhury N, Venkatesan B, Li X, Mahimainathan L, Choudhury GG. Akt kinase targets association of CBP with SMAD 3 to regulate TGFbeta-induced expression of plasminogen activator inhibitor-1. J Cell Physiol. 2008; 214:513–27.
49. Munger JS, Huang X, Kawakatsu H, Griffiths MJD, Dalton SL, Wu J, et al. The integrin αvβ6 binds and activates latent TGFβ1: a mechanism for regulating pulmonary inflammation and fibrosis. Cell. 1999; 96:319–28.
Article
50. Mackinnon AC, Gibbons MA, Farnworth SL, Leffler H, Nilsson UJ, Delaine T, et al. Regulation of transforming growth factor-β1-driven lung fibrosis by galectin-3. Am J Respir Crit Care Med. 2012; 185:537–46.
Article
51. Brigstock DR. The connective tissue growth factor/cysteine-rich 61/nephroblastoma overexpressed (CCN) family. Endocr Rev. 1999; 20:189–206.
Article
52. Kondo S, Kubota S, Shimo T, Nishida T, Yosimichi G, Eguchi T, et al. Connective tissue growth factor increased by hypoxia may initiate angiogenesis in collaboration with matrix metalloproteinases. Carcinogenesis. 2002; 23:769–76.
Article
53. Shimo T, Nakanishi T, Nishida T, Asano M, Sasaki A, Kanyama M, et al. Involvement of CTGF, a hypertrophic chondrocyte-specific gene product, in tumor angiogenesis. Oncology. 2001; 61:315–22.
Article
54. Takigawa M. CTGF/Hcs24 as a multifunctional growth factor for fibroblasts, chondrocytes and vascular endothelial cells. Drug News Perspect. 2003; 16:11–21.
Article
55. Igarashi A, Okochi H, Bradham DM, Grotendorst GR. Regulation of connective tissue growth factor gene expression in human skin fibroblasts and during wound repair. Mol Biol Cell. 1993; 4:637–45.
Article
56. Grotendorst GR, Okochi H, Hayashi N. A novel transforming growth factor beta response element controls the expression of the connective tissue growth factor gene. Cell Growth Differ. 1996; 7:469–80.
57. Shimo T, Kubota S, Kondo S, Nakanishi T, Sasaki A, Mese H, et al. Connective tissue growth factor as a major angiogenic agent that is induced by hypoxia in a human breast cancer cell line. Cancer Lett. 2001; 174:57–64.
Article
58. Suzuma K, Naruse K, Suzuma I, Takahara N, Ueki K, Aiello LP, et al. Vascular endothelial growth factor induces expression of connective tissue growth factor via KDR, Flt1, and phosphatidylinositol 3-kinase-akt-dependent pathways in retinal vascular cells. J Biol Chem. 2000; 275:40725–31.
Article
59. Lau LF. Cell surface receptors for CCN proteins. J Cell Commun Signal. 2016; 10:121–7.
Article
60. Wang Q, Usinger W, Nichols B, Gray J, Xu L, Seeley TW, et al. Cooperative interaction of CTGF and TGF-β in animal models of fibrotic disease. Fibrogenesis Tissue Repair. 2011; 4:4.
Article
61. Kono M, Nakamura Y, Suda T, Kato M, Kaida Y, Hashimoto D, et al. Plasma CCN2 (connective tissue growth factor; CTGF) is a potential biomarker in idiopathic pulmonary fibrosis (IPF). Clin Chim Acta. 2011; 412:2211–5.
Article
62. Pan LH, Yamauchi K, Uzuki M, Nakanishi T, Takigawa M, Inoue H, et al. Type II alveolar epithelial cells and interstitial fibroblasts express connective tissue growth factor in IPF. Eur Respir J. 2001; 17:1220–7.
Article
63. Gabbiani G. Modulation of fibroblastic cytoskeletal features during wound healing and fibrosis. Pathol Res Pract. 1994; 190:851–3.
Article
64. Sandbo N, Dulin N. Actin cytoskeleton in myofibroblast differentiation: ultrastructure defining form and driving function. Transl Res. 2011; 158:181–96.
Article
65. Knipe RS, Tager AM, Liao JK. The Rho kinases: critical mediators of multiple profibrotic processes and rational targets for new therapies for pulmonary fibrosis. Pharmacol Rev. 2015; 67:103–17.
Article
66. Zhou Y, Huang X, Hecker L, Kurundkar D, Kurundkar A, Liu H, et al. Inhibition of mechanosensitive signaling in myofibroblasts ameliorates experimental pulmonary fibrosis. J Clin Invest. 2013; 123:1096–108.
Article
67. Griffith DE, Eagle G, Thomson R, Aksamit TR, Hasegawa N, Morimoto K, et al. Amikacin liposome inhalation suspension for treatment-refractory lung disease caused by Mycobacterium avium complex (CONVERT): a prospective, open-label, randomized study. Am J Respir Crit Care Med. 2018; 198:1559–69.
Article
68. Yoshida K, Kuwano K, Hagimoto N, Watanabe K, Matsuba T, Fujita M, et al. MAP kinase activation and apoptosis in lung tissues from patients with idiopathic pulmonary fibrosis. J Pathol. 2002; 198:388–96.
Article
69. Alcorn JF, van der Velden J, Brown AL, McElhinney B, Irvin CG, Janssen-Heininger YM. c-Jun N-terminal kinase 1 is required for the development of pulmonary fibrosis. Am J Respir Cell Mol Biol. 2009; 40:422–32.
Article
70. Van der Velden JL, Alcorn JF, Chapman DG, Lundblad LK, Irvin CG, Davis RJ, et al. Airway epithelial specific deletion of Jun-N-terminal kinase 1 attenuates pulmonary fibrosis in two independent mouse models. PLoS One. 2020; 15:e0226904.
Article
71. Bennett B, Blease K, Ye Y, Azaryan A, Ramirez-Valle F, Ceres R, et al. CC-90001, a second generation Jun N-terminal kinase (JNK) inhibitor for the treatment of idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2017; 195:A5409.
72. Van der Velden JL, Ye Y, Nolin JD, Hoffman SM, Chapman DG, Lahue KG, et al. JNK inhibition reduces lung remodeling and pulmonary fibrotic systemic markers. Clin Transl Med. 2016; 5:36.
73. Clement DL, Mally S, Stock C, Lethan M, Satir P, Schwab A, et al. PDGFRα signaling in the primary cilium regulates NHE1-dependent fibroblast migration via coordinated differential activity of MEK1/2-ERK1/2-p90RSK and AKT signaling pathways. J Cell Sci. 2013; 126(Pt 4):953–65.
Article
74. Delehedde M, Seve M, Sergeant N, Wartelle I, Lyon M, Rudland PS, et al. Fibroblast growth factor-2 stimulation of p42/44MAPK phosphorylation and IkappaB degradation is regulated by heparan sulfate/heparin in rat mammary fibroblasts. J Biol Chem. 2000; 275:33905–10.
75. Seko Y, Takahashi N, Tobe K, Ueki K, Kadowaki T, Yazaki Y. Vascular endothelial growth factor (VEGF) activates Raf-1, mitogen-activated protein (MAP) kinases, and S6 kinase (p90rsk) in cultured rat cardiac myocytes. J Cell Physiol. 1998; 175:239–46.
Article
76. Rovida E, Navari N, Caligiuri A, Dello Sbarba P, Marra F. ERK5 differentially regulates PDGF-induced proliferation and migration of hepatic stellate cells. J Hepatol. 2008; 48:107–15.
Article
77. Roberts OL, Holmes K, Muller J, Cross DA, Cross MJ. ERK5 and the regulation of endothelial cell function. Biochem Soc Trans. 2009; 37(Pt 6):1254–9.
Article
78. Kimura TE, Jin J, Zi M, Prehar S, Liu W, Oceandy D, et al. Targeted deletion of the extracellular signal-regulated protein kinase 5 attenuates hypertrophic response and promotes pressure overload-induced apoptosis in the heart. Circ Res. 2010; 106:961–70.
Article
79. Urushihara M, Takamatsu M, Shimizu M, Kondo S, Kinoshita Y, Suga K, et al. ERK5 activation enhances mesangial cell viability and collagen matrix accumulation in rat progressive glomerulonephritis. Am J Physiol Renal Physiol. 2010; 298:F167–76.
Article
80. Kim S, Lim JH, Woo CH. ERK5 inhibition ameliorates pulmonary fibrosis via regulating Smad3 acetylation. Am J Pathol. 2013; 183:1758–68.
Article
81. Frodin M, Gammeltoft S. Role and regulation of 90 kDa ribosomal S6 kinase (RSK) in signal transduction. Mol Cell Endocrinol. 1999; 151:65–77.
Article
82. Morales-Ibanez O, Affo S, Rodrigo-Torres D, Blaya D, Millan C, Coll M, et al. Kinase analysis in alcoholic hepatitis identifies p90RSK as a potential mediator of liver fibrogenesis. Gut. 2016; 65:840–51.
Article
83. Jo E, Park SJ, Choi YS, Jeon WK, Kim BC. Kaempferol suppresses transforming growth factor-β1-induced epithelial-to-mesenchymal transition and migration of A549 lung cancer cells by inhibiting akt1-mediated phosphorylation of Smad3 at threonine-179. Neoplasia. 2015; 17:525–37.
Article
84. Kim S, Han JH, Kim S, Lee H, Kim JR, Lim JH, et al. p90RSK inhibition ameliorates TGF-β1 signaling and pulmonary fibrosis by inhibiting Smad3 transcriptional activity. Cell Physiol Biochem. 2020; 54:195–210.
Article
Full Text Links
  • YUJM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr