Skip to main content
American Journal of Respiratory Cell and Molecular Biology logoLink to American Journal of Respiratory Cell and Molecular Biology
. 2014 May;50(5):853–856. doi: 10.1165/rcmb.2014-0089PS

Alveolar Epithelium: Beyond the Barrier

Zea Borok 1,
PMCID: PMC4068954  PMID: 24783957

Abstract

I am deeply honored to have been awarded an American Thoracic Society Recognition Award for Scientific Accomplishment for 2014. Over the last 20 years, it has become clear that the alveolar epithelium, my area of research focus, is not simply a gas exchange surface and barrier to leakage of fluid and protein into the alveoli, but is an active participant in the pathogenesis of a number of lung diseases, including pulmonary fibrosis. Recognition by this Award stimulates a review of the awardee’s contributions to the field, as summarized in this perspective.

Why Alveolar Epithelium?

My entry into the study of alveolar epithelial cell (AEC) biology was in many ways serendipitous when I joined the laboratory of Dr. Edward Crandall as a junior faculty member. A specific research focus was less important to me at the time than my desire that the area be relevant to human disease. The laboratory’s focus on AEC biology and regulation of alveolar epithelial barrier properties appealed to me because of its direct relevance to my clinical interests in acute lung injury and the acute respiratory distress syndrome. As the field evolved over the years, it has become clear that the alveolar epithelium is far more than “just a barrier,” and is increasingly appreciated to play an active and important role in the pathogenesis of human disease (1, 2).

Regulation of Alveolar Epithelial Barrier Properties

My early work focused on characterization of ion transport molecules expressed by AECs, and elucidation of their transcriptional regulation to understand mechanisms regulating alveolar fluid clearance and alveolar barrier properties (36). The complexity of the distal lung necessitated the use of simplified models to characterize these properties. Many of our studies were conducted using a model of AECs in primary culture on semipermeable supports, which recapitulates many of the properties of alveolar epithelium in vivo (7, 8), and in which alveolar type II (AT2) cells in primary culture undergo transdifferentiation to a type I (AT1) cell–like phenotype (9). Using this model, we established serum-free conditions for AEC culture (8) and identified factors (e.g., epidermal growth factor and keratinocyte growth factor [KGF]) that up-regulate transport properties through effects on both Na pumps and channels (35, 10), and members of the claudin family of tight junction proteins, suggesting that these factors might be useful for enhancing fluid clearance after lung injury.

Type I–Like AEC Monolayers versus Type I Cells

Our in vitro model of AT2 cells in primary culture used for studies of AEC physiology initially encountered skepticism, because it was thought to represent dedifferentiation of AT2 cells rather than differentiation to another interrelated phenotype. With the advent of new AT1 cell phenotypic markers, it became clear that AT2 cells in primary culture acquire many characteristics of AT1 cells over time (9, 11, 12), recapitulating the process in vivo where AT2 cells serve as progenitors of AT1 cells (13). This led to the concept that AT2 cells in primary culture undergo transdifferentiation to an “AT1 cell–like” phenotype. This concept was somewhat controversial, and I recall having to insert a caveat into grant proposals indicating that “while AT1-like cells acquire many but not all the properties of AT1 cells, they nevertheless constitute a useful model of alveolar epithelium, studies of which provide significant mechanistic insights into AEC function and biology.” This begged the question of how closely AT1-like cells resembled AT1 cells in vivo, and led us to develop methods for isolation of AT1 cells from rat lung (14). Although our studies of AT1-like cells had suggested a role for AT1 cells in ion transport in distal lung, a direct contribution of AT1 cells to alveolar homeostasis had not yet been demonstrated. To address this question, we and others developed techniques for isolation of highly purified (∼95% purity) populations of AT1 cells from rat lung (14, 15). We demonstrated that freshly isolated AT1 cells indeed express subunits of Na pumps and Na channels, confirming observations in AT1-like cells and implicating AT1 cells in ion transport and alveolar fluid homeostasis. Successful isolation of these highly purified populations of AT1 cells formed the basis for subsequent phenotypic characterization of these important lung cells using one of the early microarray platforms (which could analyze expression of only 1,000 genes!) and further studies of the contributions of AT1 cells to alveolar and lung biology (16). Many of our initial observations in AT1-like cells were subsequently corroborated in isolated AT1 cells or in vivo, emphasizing the utility of this now generally accepted model for functional studies of AT1 cells.

Plasticity of AEC Phenotype

Using this in vitro model of AEC transdifferentiation, we made the important observation that the phenotype of AECs in primary culture could be significantly modulated by exogenous soluble factors or interactions with substratum (1720). It had been believed that, over time, AT2 cells in primary culture invariably default to an AT1 cell phenotype. Whether or not this transition could be modulated was unknown. Using a number of AT1 cell phenotypic markers, we demonstrated that AECs that had already acquired AT1 cell characteristics in culture could be induced to revert to an AT2 cell phenotype through exposure to KGF or changes in cell shape, indicating that AEC phenotype is actively regulated. Effects of KGF were subsequently shown to be mediated by c-Jun N-terminal kinase signaling (21). The demonstration of reversible AEC phenotypic transitions challenges the widely held belief that AT1 cells are terminally differentiated. Although still to be demonstrated by lineage tagging in vivo using mice that we have developed with AT1 cell–specific reporter expression (22), the notion that AT1 cells may not be terminally differentiated is supported by observations of others that isolated AT1 cells can proliferate in culture (23, 24). It also suggests greater plasticity of AEC phenotype than previously appreciated, a concept that is becoming more widely accepted with recent demonstrations of stem cell plasticity/reprogramming.

To elucidate mechanisms regulating AEC phenotype and further characterize interrelationships between AT2 and AT1 cells, we characterized the promoter of the water channel, aquaporin-5, which, within distal lung epithelium, is selectively expressed in AT1 cells (25, 26). We used Aqp5 as a prototype AT1 cell gene with which to elucidate molecular mechanisms that regulate phenotype transitions between AT2 and AT1 cells by transcriptional activators (e.g., GATA-binding factor 6 (27) and repressors (e.g., Forkhead box p2 and A1 Foxp2 and FoxA1 (28, 29) that are also implicated in lung development through their interactions with other lung-enriched transcription factors (TFs). Extending our studies of single genes and TFs, we recently undertook a genome-wide analysis of transcriptomic and epigenetic changes accompanying AEC differentiation in vitro. Integration of transcriptional changes with surrounding chromatin modifications enabled characterization of molecular signaling events that were activated or repressed during adult AEC differentiation and identification of putative novel regulators (e.g., hepatocyte nuclear factor 4a) and signaling pathways (e.g., retinoid X receptor pathway) involved in this process (30). The demonstration that AEC phenotype is actively regulated and elucidation of underlying regulatory pathways are of particular relevance when considering strategies to modulate AEC differentiation to enhance alveolar epithelial repair.

Central Role of Alveolar Epithelium in Pulmonary Fibrosis: Apoptosis, Epithelial–Mesenchymal Transition, and Epithelial–Fibroblast Cross-Talk

Our observations of AEC plasticity raised the question of whether AECs could also give rise to other (nonepithelial) cell types. We showed that AECs exposed to transforming growth factor (TGF)-β undergo epithelial–mesenchymal transition (EMT) in vitro, as evidenced by loss of epithelial and acquisition of mesenchymal markers (31, 32). We further demonstrated colocalization of epithelial and mesenchymal markers in up to 80% of hyperplastic AECs in lung biopsies of patients with idiopathic pulmonary fibrosis (IPF), suggesting that EMT may contribute to the pathogenesis of fibrosis in vivo. Subsequent demonstration that endoplasmic reticulum (ER) stress (either chemically induced or in response to accumulation of misfolded mutant surfactant protein) induces EMT in AECs in a dose-dependent manner (with lower levels of ER stress inducing EMT and higher levels inducing apoptosis) suggests that expression of mesenchymal markers may represent an adaptive response of AECs to injury, which, when overwhelmed, leads to cell death (33, 34). Lineage tracing studies have since raised questions about the precise role of EMT in fibrogenesis (because the number of EMT-derived fibroblasts has been variable among different studies, and sometimes low to absent) (3538). Nevertheless, a number of studies in addition to our own have shown that AECs in IPF are abnormal and express mesenchymal markers (37, 39, 40), leading to a major paradigm shift that suggests a central role of alveolar epithelium in IPF pathogenesis (2, 41). The demonstration that deletion of the TGF-β type II receptor specifically in lung epithelium protects mice from bleomycin-induced pulmonary fibrosis (42, 43) further supports a central role for alveolar epithelium in fibrogenesis, even if indirectly, as a result of aberrant epithelial–mesenchymal interactions (44). Evidence of AEC abnormalities (including morphologic changes, apoptosis [45], EMT, up-regulation of TGF-β [46] and other cytokines [47], and evidence of ER stress [33, 48]) in IPF lung, and of fibrosis in association with genetic abnormalities (e.g., surfactant and telomerase mutations) that affect the alveolar epithelium, strongly support the evolving notion that epithelial abnormalities as a result of injury (as yet of unknown cause) and aberrant repair contribute to IPF pathogenesis (41). Demonstration that the injured alveolar epithelium is abnormal or “reprogrammed” in IPF has identified new areas of investigation aimed at understanding how the abnormal epithelium contributes to fibrosis and elucidating genetic and other mechanisms underlying epithelial injury.

Translational Aspects

Aberrant wngless-related MMTV integration site/β-catenin signaling in alveolar epithelium has been implicated in the pathogenesis of IPF as a result of epithelial hyperplasia, altered AEC differentiation, and altered epithelial–mesenchymal cross-talk (49, 50). Interactions of β-catenin with the homologous transcriptional coactivators, cAMP response element–binding protein–binding protein (CBP) and p300, have been shown to differentially regulate subsets of target genes, leading to different functional outcomes (51). We have shown that interactions between β-catenin and TGF-β pathways are implicated in EMT, and are dependent on specific interactions of β-catenin with the transcriptional coactivator, CBP (52). Importantly, treatment with ICG-001, a selective small-molecule inhibitor of β-catenin–CBP interactions, both prevents and reverses bleomycin-induced fibrosis while preserving epithelial integrity, suggesting a potential novel therapeutic approach to pulmonary fibrosis (53). We also recently demonstrated that the peroxisome proliferator–activated receptor γ agonist troglitazone ameliorates TGF-β–induced EMT in a peroxisome proliferator–activated receptor γ–independent manner in AECs, likely through inhibition of β-catenin–dependent signaling downstream of TGF-β. Understanding the contribution(s) of alveolar epithelium to fibrogenesis may offer unique opportunities for development of new therapeutic interventions in IPF.

Beyond the Barrier

Research in AEC biology, as with other fields, has, to a large extent, been driven by technological advances and feasibility. In my case, what began as physiologic measurements in simplified cell culture models (3, 4, 54), and subsequently highly purified populations of isolated AECs (14), evolved to elucidation of the molecular bases of AEC function and phenotype. From initial analyses of expression of selected genes and TFs of interest at baseline and after injury (5, 6, 25), and subsequent generation of mice with the capacity for modulation of genes of interest in AT1 cells in vivo (22), our studies have culminated recently in genome-wide analyses leading to insights into transcriptomic and epigenetic changes that accompany AEC differentiation (30). We have come full circle, with the field now being poised for the study of cellular heterogeneity with advances in technology for single-cell RNA-Seq and generation of differentiated AT2 cells through direct cellular reprogramming.

Going Forward

When starting my research career, I remember being worried that I would run out of questions to ask. After more than 20 years, I know now that the opposite is true: the questions are infinite, and each result leads to more questions. It has been exciting to be working in such a rapidly advancing field, where the importance of alveolar epithelium in both acute and chronic lung disease has become much better appreciated. Rather than a passive bystander, the epithelium is actively involved in human disease pathogenesis. Building on our initial in vitro studies, we plan to further investigate differential contributions of AT2 and AT1 cells to alveolar function, fibrogenesis, and repair in vivo and, applying genome-wide approaches to our in vitro model and purified AEC populations, elucidate signaling pathways that are aberrantly activated in AECs in the fibrotic lung. We hope to apply knowledge gained from elucidation of pathways regulating normal and aberrant AEC differentiation to enhance epithelial repair mechanisms, with the ultimate goal of ameliorating fibrotic lung disease.

Footnotes

This work was supported by National Institutes of Health research grants R37HL062569, R01HL112638, and R01HL114094.

Author disclosures are available with the text of this article at www.atsjournals.org.

References

  • 1.Bhattacharya J, Matthay MA. Regulation and repair of the alveolar–capillary barrier in acute lung injury. Annu Rev Physiol. 2013;75:593–615. doi: 10.1146/annurev-physiol-030212-183756. [DOI] [PubMed] [Google Scholar]
  • 2.Selman M, Pardo A. Role of epithelial cells in idiopathic pulmonary fibrosis: from innocent targets to serial killers. Proc Am Thorac Soc. 2006;3:364–372. doi: 10.1513/pats.200601-003TK. [DOI] [PubMed] [Google Scholar]
  • 3.Borok Z, Danto SI, Dimen LL, Zhang XL, Lubman RL. Na(+)-K(+)-ATPase expression in alveolar epithelial cells: upregulation of active ion transport by KGF. Am J Physiol. 1998;274:L149–L158. doi: 10.1152/ajplung.1998.274.1.L149. [DOI] [PubMed] [Google Scholar]
  • 4.Borok Z, Hami A, Danto SI, Lubman RL, Kim KJ, Crandall ED. Effects of EGF on alveolar epithelial junctional permeability and active sodium transport. Am J Physiol. 1996;270:L559–L565. doi: 10.1152/ajplung.1996.270.4.L559. [DOI] [PubMed] [Google Scholar]
  • 5.Danto SI, Borok Z, Zhang XL, Lopez MZ, Patel P, Crandall ED, Lubman RL. Mechanisms of EGF-induced stimulation of sodium reabsorption by alveolar epithelial cells. Am J Physiol. 1998;275:C82–C92. doi: 10.1152/ajpcell.1998.275.1.C82. [DOI] [PubMed] [Google Scholar]
  • 6.Borok Z, Mihyu S, Fernandes VF, Zhang XL, Kim KJ, Lubman RL. KGF prevents hyperoxia-induced reduction of active ion transport in alveolar epithelial cells. Am J Physiol. 1999;276:C1352–C1360. doi: 10.1152/ajpcell.1999.276.6.C1352. [DOI] [PubMed] [Google Scholar]
  • 7.Cheek JM, Kim KJ, Crandall ED. Tight monolayers of rat alveolar epithelial cells: bioelectric properties and active sodium transport. Am J Physiol. 1989;256:C688–C693. doi: 10.1152/ajpcell.1989.256.3.C688. [DOI] [PubMed] [Google Scholar]
  • 8.Kim KJ, Borok Z, Crandall ED. A useful in vitro model for transport studies of alveolar epithelial barrier. Pharm Res. 2001;18:253–255. doi: 10.1023/a:1011040824988. [DOI] [PubMed] [Google Scholar]
  • 9.Cheek JM, Evans MJ, Crandall ED. Type I cell–like morphology in tight alveolar epithelial monolayers. Exp Cell Res. 1989;184:375–387. doi: 10.1016/0014-4827(89)90337-6. [DOI] [PubMed] [Google Scholar]
  • 10.Kemp PJ, Borok Z, Kim KJ, Lubman RL, Danto SI, Crandall ED. Epidermal growth factor regulation in adult rat alveolar type II cells of amiloride-sensitive cation channels. Am J Physiol. 1999;277:C1058–C1065. doi: 10.1152/ajpcell.1999.277.6.C1058. [DOI] [PubMed] [Google Scholar]
  • 11.Dobbs LG, Williams MC, Gonzalez R. Monoclonal antibodies specific to apical surfaces of rat alveolar type I cells bind to surfaces of cultured, but not freshly isolated, type II cells. Biochim Biophys Acta. 1988;970:146–156. doi: 10.1016/0167-4889(88)90173-5. [DOI] [PubMed] [Google Scholar]
  • 12.Danto SI, Zabski SM, Crandall ED. Reactivity of alveolar epithelial cells in primary culture with type I cell monoclonal antibodies. Am J Respir Cell Mol Biol. 1992;6:296–306. doi: 10.1165/ajrcmb/6.3.296. [DOI] [PubMed] [Google Scholar]
  • 13.Adamson IY, Bowden DH. The type 2 cell as progenitor of alveolar epithelial regeneration: a cytodynamic study in mice after exposure to oxygen. Lab Invest. 1974;30:35–42. [PubMed] [Google Scholar]
  • 14.Borok Z, Liebler JM, Lubman RL, Foster MJ, Zhou B, Li X, Zabski SM, Kim KJ, Crandall ED. Na transport proteins are expressed by rat alveolar epithelial type I cells. Am J Physiol Lung Cell Mol Physiol. 2002;282:L599–L608. doi: 10.1152/ajplung.00130.2000. [DOI] [PubMed] [Google Scholar]
  • 15.Johnson MD, Widdicombe JH, Allen L, Barbry P, Dobbs LG. Alveolar epithelial type I cells contain transport proteins and transport sodium, supporting an active role for type I cells in regulation of lung liquid homeostasis. Proc Natl Acad Sci USA. 2002;99:1966–1971. doi: 10.1073/pnas.042689399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Qiao R, Zhou B, Liebler JM, Li X, Crandall ED, Borok Z. Identification of three genes of known function expressed by alveolar epithelial type I cells. Am J Respir Cell Mol Biol. 2003;29:98–105. doi: 10.1165/rcmb.2002-0196OC. [DOI] [PubMed] [Google Scholar]
  • 17.Borok Z, Danto SI, Lubman RL, Cao Y, Williams MC, Crandall ED. Modulation of t1alpha expression with alveolar epithelial cell phenotype in vitro. Am J Physiol. 1998;275:L155–L164. doi: 10.1152/ajplung.1998.275.1.L155. [DOI] [PubMed] [Google Scholar]
  • 18.Borok Z, Lubman RL, Danto SI, Zhang XL, Zabski SM, King LS, Lee DM, Agre P, Crandall ED. Keratinocyte growth factor modulates alveolar epithelial cell phenotype in vitro: expression of aquaporin 5. Am J Respir Cell Mol Biol. 1998;18:554–561. doi: 10.1165/ajrcmb.18.4.2838. [DOI] [PubMed] [Google Scholar]
  • 19.Danto SI, Shannon JM, Borok Z, Zabski SM, Crandall ED. Reversible transdifferentiation of alveolar epithelial cells. Am J Respir Cell Mol Biol. 1995;12:497–502. doi: 10.1165/ajrcmb.12.5.7742013. [DOI] [PubMed] [Google Scholar]
  • 20.Borok Z, Hami A, Danto SI, Zabski SM, Crandall ED. Rat serum inhibits progression of alveolar epithelial cells toward the type I cell phenotype in vitro. Am J Respir Cell Mol Biol. 1995;12:50–55. doi: 10.1165/ajrcmb.12.1.7811470. [DOI] [PubMed] [Google Scholar]
  • 21.Qiao R, Yan W, Clavijo C, Mehrian-Shai R, Zhong Q, Kim KJ, Ann D, Crandall ED, Borok Z. Effects of KGF on alveolar epithelial cell transdifferentiation are mediated by JNK signaling. Am J Respir Cell Mol Biol. 2008;38:239–246. doi: 10.1165/rcmb.2007-0172OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Flodby P, Borok Z, Banfalvi A, Zhou B, Gao D, Minoo P, Ann DK, Morrisey EE, Crandall ED. Directed expression of Cre in alveolar epithelial type 1 cells. Am J Respir Cell Mol Biol. 2010;43:173–178. doi: 10.1165/rcmb.2009-0226OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Gonzalez RF, Allen L, Dobbs LG. Rat alveolar type I cells proliferate, express OCT-4, and exhibit phenotypic plasticity in vitro. Am J Physiol Lung Cell Mol Physiol. 2009;297:L1045–L1055. doi: 10.1152/ajplung.90389.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Wang S, Hubmayr RD. Type I alveolar epithelial phenotype in primary culture. Am J Respir Cell Mol Biol. 2011;44:692–699. doi: 10.1165/rcmb.2009-0359OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Borok Z, Li X, Fernandes VF, Zhou B, Ann DK, Crandall ED. Differential regulation of rat aquaporin-5 promoter/enhancer activities in lung and salivary epithelial cells. J Biol Chem. 2000;275:26507–26514. doi: 10.1074/jbc.M910007199. [DOI] [PubMed] [Google Scholar]
  • 26.Zhou B, Ann DK, Flodby P, Minoo P, Liebler JM, Crandall ED, Borok Z. Rat aquaporin-5 4.3-kb 5′-flanking region differentially regulates expression in salivary gland and lung in vivo. Am J Physiol Cell Physiol. 2008;295:C111–C120. doi: 10.1152/ajpcell.90620.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Zhou B, Francis TA, Yang H, Tseng W, Zhong Q, Frenkel B, Morrisey EE, Ann DK, Minoo P, Crandall ED, et al. GATA-6 mediates transcriptional activation of aquaporin-5 through interactions with Sp1. Am J Physiol Cell Physiol. 2008;295:C1141–C1150. doi: 10.1152/ajpcell.00120.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Zhou B, Zhong Q, Minoo P, Li C, Ann DK, Frenkel B, Morrisey EE, Crandall ED, Borok Z. Foxp2 inhibits Nkx2.1-mediated transcription of SP-C via interactions with the Nkx2.1 homeodomain. Am J Respir Cell Mol Biol. 2008;38:750–758. doi: 10.1165/rcmb.2007-0350OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Minoo P, Hu L, Xing Y, Zhu NL, Chen H, Li M, Borok Z, Li C. Physical and functional interactions between homeodomain NKX2.1 and winged helix/forkhead FOXA1 in lung epithelial cells. Mol Cell Biol. 2007;27:2155–2165. doi: 10.1128/MCB.01133-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Marconett CN, Zhou B, Rieger ME, Selamat SA, Dubourd M, Fang X, Lynch SK, Stueve TR, Siegmund KD, Berman BP, et al. Integrated transcriptomic and epigenomic analysis of primary human lung epithelial cell differentiation. PLoS Genet. 2013;9:e1003513. doi: 10.1371/journal.pgen.1003513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Willis BC, Liebler JM, Luby-Phelps K, Nicholson AG, Crandall ED, du Bois RM, Borok Z. Induction of epithelial–mesenchymal transition in alveolar epithelial cells by transforming growth factor-beta1: potential role in idiopathic pulmonary fibrosis. Am J Pathol. 2005;166:1321–1332. doi: 10.1016/s0002-9440(10)62351-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Willis BC, duBois RM, Borok Z. Epithelial origin of myofibroblasts during fibrosis in the lung. Proc Am Thorac Soc. 2006;3:377–382. doi: 10.1513/pats.200601-004TK. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Zhong Q, Zhou B, Ann DK, Minoo P, Liu Y, Banfalvi A, Krishnaveni MS, Dubourd M, Demaio L, Willis BC, et al. Role of endoplasmic reticulum stress in epithelial-mesenchymal transition of alveolar epithelial cells: effects of misfolded surfactant protein. Am J Respir Cell Mol Biol. 2011;45:498–509. doi: 10.1165/rcmb.2010-0347OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Kage H, Borok Z. EMT and interstitial lung disease: a mysterious relationship. Curr Opin Pulm Med. 2012;18:517–523. doi: 10.1097/MCP.0b013e3283566721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.DeMaio L, Buckley ST, Krishnaveni MS, Flodby P, Dubourd M, Banfalvi A, Xing Y, Ehrhardt C, Minoo P, Zhou B, et al. Ligand-independent transforming growth factor-β type I receptor signalling mediates type I collagen–induced epithelial–mesenchymal transition. J Pathol. 2012;226:633–644. doi: 10.1002/path.3016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Rock JR, Barkauskas CE, Cronce MJ, Xue Y, Harris JR, Liang J, Noble PW, Hogan BL. Multiple stromal populations contribute to pulmonary fibrosis without evidence for epithelial to mesenchymal transition. Proc Natl Acad Sci USA. 2011;108:E1475–E1483. doi: 10.1073/pnas.1117988108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Kim KK, Kugler MC, Wolters PJ, Robillard L, Galvez MG, Brumwell AN, Sheppard D, Chapman HA. Alveolar epithelial cell mesenchymal transition develops in vivo during pulmonary fibrosis and is regulated by the extracellular matrix. Proc Natl Acad Sci USA. 2006;103:13180–13185. doi: 10.1073/pnas.0605669103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Tanjore H, Xu XC, Polosukhin VV, Degryse AL, Li B, Han W, Sherrill TP, Plieth D, Neilson EG, Blackwell TS, et al. Contribution of epithelial-derived fibroblasts to bleomycin-induced lung fibrosis. Am J Respir Crit Care Med. 2009;180:657–665. doi: 10.1164/rccm.200903-0322OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Larsson O, Diebold D, Fan D, Peterson M, Nho RS, Bitterman PB, Henke CA. Fibrotic myofibroblasts manifest genome-wide derangements of translational control. PLoS ONE. 2008;3:e3220. doi: 10.1371/journal.pone.0003220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Jayachandran A, Königshoff M, Yu H, Rupniewska E, Hecker M, Klepetko W, Seeger W, Eickelberg O. SNAI transcription factors mediate epithelial–mesenchymal transition in lung fibrosis. Thorax. 2009;64:1053–1061. doi: 10.1136/thx.2009.121798. [DOI] [PubMed] [Google Scholar]
  • 41.Blackwell TS, Tager AM, Borok Z, Moore BB, Schwartz DA, Anstrom KJ, Bar-Joseph Z, Bitterman P, Blackburn MR, Bradford W, et al. Future directions in idiopathic pulmonary fibrosis research: an NHLBI workshop report. Am J Respir Crit Care Med. 2014;189:214–222. doi: 10.1164/rccm.201306-1141WS. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Li M, Krishnaveni MS, Li C, Zhou B, Xing Y, Banfalvi A, Li A, Lombardi V, Akbari O, Borok Z, et al. Epithelium-specific deletion of TGF-β receptor type II protects mice from bleomycin-induced pulmonary fibrosis. J Clin Invest. 2011;121:277–287. doi: 10.1172/JCI42090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Degryse AL, Tanjore H, Xu XC, Polosukhin VV, Jones BR, Boomershine CS, Ortiz C, Sherrill TP, McMahon FB, Gleaves LA, et al. TGFβ signaling in lung epithelium regulates bleomycin-induced alveolar injury and fibroblast recruitment. Am J Physiol Lung Cell Mol Physiol. 2011;300:L887–L897. doi: 10.1152/ajplung.00397.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Sakai N, Tager AM. Fibrosis of two: epithelial cell–fibroblast interactions in pulmonary fibrosis. Biochim Biophys Acta. 2013;1832:911–921. doi: 10.1016/j.bbadis.2013.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Uhal BD. Apoptosis in lung fibrosis and repair. Chest. 2002;122(6 Suppl):293S–298S. doi: 10.1378/chest.122.6_suppl.293s. [DOI] [PubMed] [Google Scholar]
  • 46.Khalil N, O’Connor RN, Flanders KC, Unruh H. TGF-beta 1, but not TGF-beta 2 or TGF-beta 3, is differentially present in epithelial cells of advanced pulmonary fibrosis: an immunohistochemical study. Am J Respir Cell Mol Biol. 1996;14:131–138. doi: 10.1165/ajrcmb.14.2.8630262. [DOI] [PubMed] [Google Scholar]
  • 47.Aumiller V, Balsara N, Wilhelm J, Günther A, Königshoff M. WNT/β-catenin signaling induces IL-1β expression by alveolar epithelial cells in pulmonary fibrosis. Am J Respir Cell Mol Biol. 2013;49:96–104. doi: 10.1165/rcmb.2012-0524OC. [DOI] [PubMed] [Google Scholar]
  • 48.Lawson WE, Cheng DS, Degryse AL, Tanjore H, Polosukhin VV, Xu XC, Newcomb DC, Jones BR, Roldan J, Lane KB, et al. Endoplasmic reticulum stress enhances fibrotic remodeling in the lungs. Proc Natl Acad Sci USA. 2011;108:10562–10567. doi: 10.1073/pnas.1107559108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Morrisey EE. Wnt signaling and pulmonary fibrosis. Am J Pathol. 2003;162:1393–1397. doi: 10.1016/S0002-9440(10)64271-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Königshoff M, Balsara N, Pfaff EM, Kramer M, Chrobak I, Seeger W, Eickelberg O. Functional Wnt signaling is increased in idiopathic pulmonary fibrosis. PLoS ONE. 2008;3:e2142. doi: 10.1371/journal.pone.0002142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Ma H, Nguyen C, Lee KS, Kahn M. Differential roles for the coactivators CBP and p300 on TCF/beta-catenin–mediated survivin gene expression. Oncogene. 2005;24:3619–3631. doi: 10.1038/sj.onc.1208433. [DOI] [PubMed] [Google Scholar]
  • 52.Zhou B, Liu Y, Kahn M, Ann DK, Han A, Wang H, Nguyen C, Flodby P, Zhong Q, Krishnaveni MS, et al. Interactions between β-catenin and transforming growth factor-β signaling pathways mediate epithelial–mesenchymal transition and are dependent on the transcriptional co-activator cAMP-response element–binding protein (CREB)–binding protein (CBP) J Biol Chem. 2012;287:7026–7038. doi: 10.1074/jbc.M111.276311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Henderson WR, Jr, Chi EY, Ye X, Nguyen C, Tien YT, Zhou B, Borok Z, Knight DA, Kahn M. Inhibition of Wnt/beta-catenin/CREB binding protein (CBP) signaling reverses pulmonary fibrosis. Proc Natl Acad Sci USA. 2010;107:14309–14314. doi: 10.1073/pnas.1001520107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Borok Z, Danto SI, Zabski SM, Crandall ED. Defined medium for primary culture de novo of adult rat alveolar epithelial cells. In Vitro Cell Dev Biol Anim. 1994;30A:99–104. doi: 10.1007/BF02631400. [DOI] [PubMed] [Google Scholar]

Articles from American Journal of Respiratory Cell and Molecular Biology are provided here courtesy of American Thoracic Society

RESOURCES