Outstaying their Welcome: The Persistent Myofibroblast in IPF

Editorial

Austin J Pulm Respir Med 2014;1(1): 1002.

Outstaying their Welcome: The Persistent Myofibroblast in IPF

Steven K. Huang and Jeffrey C. Horowitz*

Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan Medical Center, USA

*Corresponding author: Jeffrey C. Horowitz, Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical Center, 6301 MSRB III, 1150 W. Medical Center Drive, Ann Arbor MI 48109, USA

Received: January 17, 2014; Accepted: January 22, 2014; Published: January 25, 2014

Pulmonary fibrosis is the final common end point of a variety of acute and chronic causes of lung injury. Regardless of the cause, it represents an aberrant wound-repair response characterized by the excessive accumulation of extracellular matrix (ECM) and the replacement of normal lung parenchyma with scar tissue. Fibroblasts are mesenchymal cells that serve a critical role in both normal and fibrotic repair processes. These cells exist along a spectrum of activation states spanning from relatively undifferentiated quiescence during times of homeostasis to differentiated, highly secretory and contractile smooth muscle-like cells with organized α–smooth muscle actin–containing stress fibers (i. e. myofibroblasts) following tissue injury [1]. In their fully activated state, myofibroblasts are the primary cells responsible for the synthesis, secretion, and remodeling of the ECM. While the resolution of normal wound repair coincides with substantial myofibroblast apoptosis, fibrotic repair is associated with the continued accumulation of these activated cells [2,3].

Idiopathic pulmonary fibrosis (IPF) is the most common and clinically refractory of the idiopathic interstitial lung diseases. Our understanding of IPF pathogenesis has evolved from one in which fibrosis was driven by excessive inflammation to one in which fibrosis results from dysfunctional interactions between an injured epithelium, reparative fibroblasts and select inflammatory cells [4,5]. Specifically, a role for alternatively activated (M2) macrophages and/ or fibrocytes, has been recognized [6,7]. Although this understanding continues to evolve, specific pathologic lesions called “fibroblastic foci” have been consistently associated with IPF and provide a window into the pathobiology of IPF [8]. These foci are comprised of activated myofibroblasts in close approximation to an injured alveolar epithelium. While not unique to IPF, these foci are thought to represent the site of “active” fibrosis within IPF lungs. Clinically, the number of fibroblastic foci correlates with mortality in IPF [9]. Biologically, these foci are characterized by an “apoptosis paradox” wherein there is prominent epithelial cell apoptosis but insufficient mesenchymal cell apoptosis [3,10]. The aberrant interactions and dysfunctional cross–talk between fibroblasts and epithelial cells may promote a self–perpetuating cycle that maintains this apoptosis paradox, even if the initial stimulus of epithelial injury has abated [8].

As noted, the unrestrained accumulation of myofibroblasts is a key feature that differentiates fibrotic from physiologic repair. The accrual of these cells represents the combined effects of cell trafficking, proliferation and death. Of these, proliferation has received considerable attention; indeed, fibroblastic foci were initially defined as “small aggregates of actively proliferating fibroblasts and myofibroblasts” [11,12]. Certainly, studies suggest that fibroblast proliferation is important, especially in the early phases of wound repair and fibrosis [13]. Moreover, soluble mediators implicated in fibrosis have been shown to induce fibroblast proliferation in vitro [14–16]. Comparisons of IPF and normal lung fibroblasts, however, have shown variable results, and some studies suggest that IPF fibroblasts actually have a decreased rate of proliferation [17,18]. Moreover, studies of IPF tissue have demonstrated that while there is substantial epithelial cell proliferation, there is little to suggest robust fibroblast proliferation within fibroblastic foci [19,20].

Decreased fibroblast apoptosis within the fibroblastic foci represents another mechanism by which these cells accumulate. In support of this, studies of IPF lung biopsies have consistently shown a lack of apoptotic cells within the myofibroblast niche [19–24], and a growing body of literature supports the hypothesis that the acquisition of an apoptosis–resistant phenotype contributes to the fibroblast accumulation in IPF [22,23,25–29]. The mechanisms underlying apoptosis resistance are likely multifactorial and may contribute to the clinical heterogeneity observed in patients with IPF. Soluble mediators strongly associated with fibrosis, most notably transforming growth factor beta–1 (TGF–β1) and endothelin–1 (ET–1), promote fibroblast resistance to death receptor–mediated apoptosis via activation of focal adhesion kinase (FAK) and the PI3K⁄AKT signaling pathways [30–33]. Each of these pro–survival protein kinases has been found to be expressed at increased levels in IPF fibroblasts and⁄or fibroblastic foci, and inhibition of these kinases has been shown to attenuate lung fibrogenesis in animal models [34–39]. TGF–β1 and ET–1 also utilize FAK and AKT to induce expression of endogenous inhibitors of apoptosis, including X–linked inhibitor of apoptosis (XIAP) and survivin, which are expressed at increased levels in fibroticlung fibroblasts and in the fibroblastic foci of IPF tissue [22,26,33]. Pharmacologic blockade or gene–silencing of these and other endogenous inhibitors of apoptosis enhances fibroblast sensitivity to apoptotic stimuli [26,27,29,32,33]. In contrast, antifibrotic mediators, such as prostaglandin E2 (PGE2), induce fibroblast apoptosis through decreased AKT signaling and suppression of XIAP and survivin [22,40]. Diminished PGE2 production and⁄or responsiveness may, therefore, also contribute to fibroblast apoptosis resistance [41]. Several studies have also shown decreased expression of Fas⁄CD95, a receptor which mediates apoptosis induced by extrinsic signals such as Fas ligand [23,42]. Loss of Fas⁄CD95 expression is mediated, inpart, through epigenetic mechanisms [43]. Stimuli that increase Fas⁄ CD95 expression on fibroblasts lead to their increased susceptibility to apoptosis [44].

Beyond the realm of soluble mediators, fibroblast interactions with the ECM are critical in the regulation of fibroblast survival and apoptosis. Disruption of biochemical and biomechanical signals generated by cell–matrix interactions can enhance sensitivity to apoptosis [45–47]. Our studies have shown that plasminogen activation, which is strongly linked with protection from fibrosis in murine models, induces fibroblast apoptosis in conjunction with fibronectin proteolysis [45]. In addition, recent studies show that alterations in ECM stiffness is sufficient to modulate fibroblast apoptosis and that fibroblasts have increased rates of apoptosis (and decreased proliferation) when exposed to substrates with a compliance similar to that of normal lung. As substrate stiffness increases toward that of fibrotic lung tissue, there is a decline in fibroblast apoptosis coupled with an increase in fibroblast proliferation [46]. These and other studies [48] suggest that the fibrotic lung matrix itself might be sufficient to support persistent myofibroblast survival and accumulation, even in the absence of ongoing lung injury or continued stimulation by soluble mediators.

IPF is a heterogeneous disease with dismal outcomes and inadequate treatment options [4,25]. The pathogenesis involves complex interactions between multiple cell types, multiple soluble mediators and dynamic cell–matrix interactions that result in the accumulation and persistence of fibroblasts and myofibroblasts. Further elucidation of the mechanisms regulating fibroblast proliferation and survival may lead to novel therapeutic interventions in IPF [25].

Acknowledgement

This work has been supported by research grants from the NIH⁄ NHLBI: R01HL105489

References

  1. Tomasek JJ, Gabbiani G, Hinz B, Chaponnier C, Brown RA. Myoibroblasts and mechano-regulation of connective tissue remodelling. Nat Rev Mol Cell Biol. 2002; 3: 349-363.
  2. Hinz B, Phan SH, Thannickal VJ, Galli A, Bochaton-Piallat ML, Gabbiani G. The myoibroblast: one function, multiple origins. Am J Pathol. 2007; 170: 1807-1816.
  3. Thannickal VJ, Horowitz JC. Evolving concepts of apoptosis in idiopathic pulmonary ibrosis. Proc Am Thorac Soc. 2006; 3: 350-356.
  4. Noble PW, Barkauskas CE, Jiang D. Pulmonary ibrosis: patterns and perpetrators. J Clin Invest. 2012; 122: 2756-2762.
  5. Wynn TA. Integrating mechanisms of pulmonary ibrosis. J Exp Med. 2011; 208: 1339-1350.
  6. Homer RJ, Elias JA, Lee CG, Herzog E. Modern concepts on the role of inlammation in pulmonary ibrosis. Arch Pathol Lab Med. 2011; 135: 780- 788.
  7. Osterholzer JJ, Christensen PJ, Lama V, Horowitz JC, Hattori N, Subbotina N, et al. PAI-1 promotes the accumulation of exudate macrophages and worsens pulmonary ibrosis following type II alveolar epithelial cell injury. J Pathol. 2012; 228: 170-180.
  8. Horowitz JC, Thannickal VJ. Epithelial-mesenchymal interactions in pulmonary ibrosis. Semin Respir Crit Care Med. 2006; 27: 600-612.
  9. King TE Jr, Schwarz MI, Brown K, Tooze JA, Colby TV, Waldron JA Jr, et al. Idiopathic pulmonary ibrosis: relationship between histopathologic features and mortality. Am J Respir Crit Care Med. 2001; 164: 1025-1032.
  10. Horowitz JC, Peters-Golden M. Prostaglandin E2’s new trick: “decider” of differential alveolar cell life and death. Am J Respir Crit Care Med. 2010; 182: 2-3.
  11. Katzenstein AL, Myers JL. Idiopathic pulmonary ibrosis: clinical relevance of pathologic classiication. Am J Respir Crit Care Med. 1998; 157: 1301-1315.
  12. Vancheri C. Idiopathic pulmonary ibrosis: an altered ibroblast proliferation linked to cancer biology. Proc Am Thorac Soc. 2012; 9: 153-157.
  13. Raghu G, Chen YY, Rusch V, Rabinovitch PS. Differential proliferation of ibroblasts cultured from normal and ibrotic human lungs. Am Rev Respir Dis. 1988; 138: 703-708.
  14. Khalil N, Xu YD, O’Connor R, Duronio V. Proliferation of pulmonary interstitial ibroblasts is mediated by transforming growth factor-beta1-induced release of extracellular ibroblast growth factor-2 and phosphorylation of p38 MAPK and JNK. J Biol Chem. 2005; 280: 43000-43009.
  15. Shahar I, Fireman E, Topilsky M, Grief J, Schwarz Y, Kivity S, et al. Effect of endothelin-1 on alpha-smooth muscle actin expression and on alveolar ibroblasts proliferation in interstitial lung diseases. Int J Immunopharmacol. 1999; 21: 759-775.
  16. Xiao L, Du Y, Shen Y, He Y, Zhao H, Li Z. TGF-beta 1 induced ibroblast proliferation is mediated by the FGF-2/ERK pathway. Front Biosci (Landmark Ed). 2012; 17: 2667-2674.
  17. Mio T, Nagai S, Kitaichi M, Kawatani A, Izumi T. Proliferative characteristics of ibroblast lines derived from open lung biopsy specimens of patients with IPF (UIP). Chest. 1992; 102: 832-837.
  18. Ramos C, Montaño M, García-Alvarez J, Ruiz V, Uhal BD, Selman M, et al. Fibroblasts from idiopathic pulmonary ibrosis and normal lungs differ in growth rate, apoptosis, and tissue inhibitor of metalloproteinases expression. Am J Respir Cell Mol Biol. 2001; 24: 591-598.
  19. Cha SI, Groshong SD, Frankel SK, Edelman BL, Cosgrove GP, Terry-Powers JL, et al. Compartmentalized expression of c-FLIP in lung tissues of patients with idiopathic pulmonary ibrosis. Am J Respir Cell Mol Biol. 2010; 42: 140- 148.
  20. Leppäranta O, Pulkkinen V, Koli K, Vähätalo R, Salmenkivi K, Kinnula VL, et al. Transcription factor GATA-6 is expressed in quiescent myoibroblasts in idiopathic pulmonary ibrosis. Am J Respir Cell Mol Biol. 2010; 42: 626-632.
  21. Kuwano K, Kunitake R, Kawasaki M, Nomoto Y, Hagimoto N, Nakanishi Y, et al. P21Waf1/Cip1/Sdi1 and p53 expression in association with DNA strand breaks in idiopathic pulmonary ibrosis. Am J Respir Crit Care Med. 1996; 154: 477-483.
  22. Maher TM, Evans IC, Bottoms SE, Mercer PF, Thorley AJ, Nicholson AG, et al. Diminished prostaglandin E2 contributes to the apoptosis paradox in idiopathic pulmonary ibrosis. Am J Respir Crit Care Med. 2010; 182: 73-82.
  23. Nho RS, Peterson M, Hergert P, Henke CA. FoxO3a (Forkhead Box O3a) deiciency protects Idiopathic Pulmonary Fibrosis (IPF) ibroblasts from type I polymerized collagen matrix-induced apoptosis via caveolin-1 (cav-1) and Fas. PLoS One. 2013; 8: e61017.
  24. Korfei M, Ruppert C, Mahavadi P, Henneke I, Markart P, Koch M, et al. Epithelial endoplasmic reticulum stress and apoptosis in sporadic idiopathic pulmonary ibrosis. Am J Respir Crit Care Med. 2008; 178: 838-846.
  25. Blackwell TS, Tager AM, Borok Z, Moore BB, Schwartz DA, Anstrom KJ, et al. Future Directions in Idiopathic Pulmonary Fibrosis Research. An NHLBI Workshop Report. Am J Respir Crit Care Med. 2014; 189: 214-222.
  26. Ajayi IO, Sisson TH, Higgins PD, Booth AJ, Sagana RL, Huang SK, et al. X-linked inhibitor of apoptosis regulates lung ibroblast resistance to Fas- mediated apoptosis. Am J Respir Cell Mol Biol. 2013; 49: 86-95.
  27. Golan-Gerstl R, Wallach-Dayan SB, Zisman P, Cardoso WV, Goldstein RH, Breuer R. Cellular FLICE-like inhibitory protein deviates myoibroblast fas- induced apoptosis toward proliferation during lung ibrosis. Am J Respir Cell Mol Biol. 2012; 47: 271-279.
  28. 28. Horowitz JC, Cui Z, Moore TA, Meier TR, Reddy RC, Toews GB, et al. Constitutive activation of prosurvival signaling in alveolar mesenchymal cells isolated from patients with nonresolving acute respiratory distress syndrome. Am J Physiol Lung Cell Mol Physiol. 2006; 290: L415-425.
  29. 29. Sisson TH, Maher TM, Ajayi IO, King JE, Higgins PD, Booth AJ, et al. Increased survivin expression contributes to apoptosis-resistance in IPF ibroblasts. Adv Biosci Biotechnol. 2012; 3: 657-664.
  30. Horowitz JC, Lee DY, Waghray M, Keshamouni VG, Thomas PE, Zhang H, et al. Activation of the pro-survival phosphatidylinositol 3-kinase/AKT pathway by transforming growth factor-beta1 in mesenchymal cells is mediated by p38 MAPK-dependent induction of an autocrine growth factor. J Biol Chem. 2004; 279: 1359-1367.
  31. Horowitz JC, Rogers DS, Sharma V, Vittal R, White ES, Cui Z, et al. Combinatorial activation of FAK and AKT by transforming growth factor-beta1 confers an anoikis-resistant phenotype to myoibroblasts. Cell Signal. 2007; 19: 761-771.
  32. Kulasekaran P, Scavone CA, Rogers DS, Arenberg DA, Thannickal VJ, Horowitz JC. Endothelin-1 and transforming growth factor-beta1 independently induce ibroblast resistance to apoptosis via AKT activation. Am J Respir Cell Mol Biol. 2009; 41: 484-493.
  33. Horowitz JC, Ajayi IO, Kulasekaran P, Rogers DS, White JB, Townsend SK, et al. Survivin expression induced by endothelin-1 promotes myoibroblast resistance to apoptosis. Int J Biochem Cell Biol. 2012; 44: 158-169.
  34. Ding Q, Cai GQ, Hu M, Yang Y, Zheng A, Tang Q, et al. FAK-related nonkinase is a multifunctional negative regulator of pulmonary ibrosis. Am J Pathol. 2013; 182: 1572-1584.
  35. Kang HR, Lee CG, Homer RJ, Elias JA. Semaphorin 7A plays a critical role in TGF-beta1-induced pulmonary ibrosis. J Exp Med. 2007; 204: 1083-1093.
  36. Korfhagen TR, Le Cras TD, Davidson CR, Schmidt SM, Ikegami M, Whitsett JA, et al. Rapamycin prevents transforming growth factor-alpha-induced pulmonary ibrosis. Am J Respir Cell Mol Biol. 2009; 41: 562-572.
  37. Lagares D, Busnadiego O, García-Fernández RA, Kapoor M, Liu S, Carter DE, et al. Inhibition of focal adhesion kinase prevents experimental lung ibrosis and myoibroblast formation. Arthritis Rheum. 2012; 64: 1653-1664.
  38. Lagares D, Busnadiego O, García-Fernández RA, Lamas S, Rodríguez- Pascual F. Adenoviral gene transfer of endothelin-1 in the lung induces pulmonary ibrosis through the activation of focal adhesion kinase. Am J Respir Cell Mol Biol. 2012; 47: 834-842.
  39. Vittal R, Horowitz JC, Moore BB, Zhang H, Martinez FJ, Toews GB, et al. Modulation of prosurvival signaling in ibroblasts by a protein kinase inhibitor protects against ibrotic tissue injury. Am J Pathol. 2005; 166: 367-375.
  40. Huang SK, White ES, Wettlaufer SH, Grifka H, Hogaboam CM, Thannickal VJ, et al. Prostaglandin E(2) induces ibroblast apoptosis by modulating multiple survival pathways. FASEB J. 2009; 23: 4317-4326.
  41. Huang SK, Wettlaufer SH, Chung J, Peters-Golden M. Prostaglandin E2 inhibits speciic lung ibroblast functions via selective actions of PKA and Epac-1. Am J Respir Cell Mol Biol. 2008; 39: 482-489.
  42. Bühling F, Wille A, Röcken C, Wiesner O, Baier A, Meinecke I, et al. Altered expression of membrane-bound and soluble CD95/Fas contributes to the resistance of ibrotic lung ibroblasts to FasL induced apoptosis. Respir Res. 2005; 6: 37.
  43. Huang SK, Scruggs AM, Donaghy J, Horowitz JC, Zaslona Z, Przybranowski S, et al. Histone modiications are responsible for decreased Fas expression and apoptosis resistance in ibrotic lung ibroblasts. Cell Death Dis. 2013; 4: e621.
  44. Wynes MW, Edelman BL, Kostyk AG, Edwards MG, Coldren C, Groshong SD, et al. Increased cell surface Fas expression is necessary and suficient to sensitize lung ibroblasts to Fas ligation-induced apoptosis: implications for ibroblast accumulation in idiopathic pulmonary ibrosis. J Immunol. 2011; 187: 527-537.
  45. Horowitz JC, Rogers DS, Simon RH, Sisson TH, Thannickal VJ. Plasminogen activation induced pericellular ibronectin proteolysis promotes ibroblast apoptosis. Am J Respir Cell Mol Biol. 2008; 38: 78-87.
  46. Liu F, Mih JD, Shea BS, Kho AT, Sharif AS, Tager AM, et al. Feedback ampliication of ibrosis through matrix stiffening and COX-2 suppression. J Cell Biol. 2010; 190: 693-706.
  47. Xia H, Nho RS, Kahm J, Kleidon J, Henke CA. Focal adhesion kinase is upstream of phosphatidylinositol 3-kinase/Akt in regulating ibroblast survival in response to contraction of type I collagen matrices via a beta 1 integrin viability signaling pathway. J Biol Chem. 2004; 279: 33024-33034.
  48. Booth AJ, Hadley R, Cornett AM, Dreffs AA, Matthes SA, Tsui JL, et al. Acellular normal and ibrotic human lung matrices as a culture system for in vitro investigation. Am J Respir Crit Care Med. 2012; 186: 866-876.

Download PDF

Citation: Huang SK, Horowitz JC. Outstaying their Welcome: The Persistent Myofibroblast in IPF. Austin J Pulm Respir Med 2014;1(1): 1002. ISSN:2381-9022

Home
Journal Scope
Online First
Current Issue
Editorial Board
Instruction for Authors
Submit Your Article
Contact Us