15
Role of the Urokinase-Fibrinolytic System in EpithelialeMesenchymal Transition during Lung Injury Q19 Amarnath Satheesh Marudamuthu,* Yashodhar Prabhakar Bhandary,* Shwetha Kumari Shetty,* Jian Fu, y Venkatachalem Sathish, z YS Prakash, z and Sreerama Shetty* Q1 Q2 From the Department of Medicine,* Texas Lung Injury Institute, University of Texas Health Science Center at Tyler, Tyler, Texas; the Center for Research on Environmental Disease and Toxicology, y College of Medicine, University of Kentucky, Lexington, Kentucky; and the Department of Anesthesiology, z Mayo Clinic, Rochester, Minnesota Accepted for publication August 28, 2014. Address correspondence to Sreerama Shetty Q4 , Ph.D., Texas Lung Injury Institute, Depart- ment of Medicine, University of Texas Health Science Center at Tyler, Tyler, TX 75708. E-mail: sreerama.shetty@uthct. edu. Alveolar type II epithelial (ATII) cell injury precedes development of pulmonary brosis. Mice lacking urokinase-type plasminogen activator (uPA) are highly susceptible, whereas those decient in plasminogen activator inhibitor (PAI-1) are resistant to lung injury and pulmonary brosis. Epithelialemesenchymal transition (EMT) has been considered, at least in part, as a source of myobroblast formation during brogenesis. However, the contribution of altered expression of major components of the uPA system on ATII cell EMT during lung injury is not well understood. To investigate whether changes in uPA and PAI-1 by ATII cells contribute to EMT, ATII cells from patients with idiopathic pulmonary brosis and chronic obstructive pulmonary disease, and mice with bleomycin-, transforming growth factor be, or passive cigarette smokeeinduced lung injury were analyzed for uPA, PAI-1, and EMT markers. We found reduced expression of E-cadherin and zona occludens-1, whereas collagen-I and a-smooth muscle actin were increased in ATII cells isolated from injured lungs. These changes were associated with a parallel increase in PAI-1 and reduced uPA expression. Further, inhibition of Src kinase activity using caveolin-1 scaf- folding domain peptide suppressed bleomycin-, transforming growth factor be, or passive cigarette smokeeinduced EMT and restored uPA expression while suppressing PAI-1. These studies show that induction of PAI-1 and inhibition of uPA during brosing lung injury lead to EMT in ATII cells. (Am J Pathol 2015, 185: 1e14; http://dx.doi.org/10.1016/j.ajpath.2014.08.027) Idiopathic Q5 pulmonary brosis (IPF) and other interstitial lung diseases are characterized by destruction of lung architecture due to excessive deposition of extracellular matrix proteins by activated broblasts or myobroblasts, leading to progressive dyspnea and loss of lung function. 1e3 The origins of myo- broblasts participating in the pathological remodeling of IPF lungs are not clear. Histopathological evaluation demonstrates that myobroblasts accumulate in broblastic foci. Emerging evidence suggests that polarized type II alveolar epithelial (ATII) cells undergo epithelialemesenchymal transitions (EMT) after lung injury. The ATII cells assume phenotypic changes such as increased migration, invasion, resistance to apoptosis, and production of elevated levels of extracellular matrix proteins 4,5 and therefore serve as a source of myo- broblasts. Understanding the possible mechanisms contrib- uting to EMT in ATII cells may help identify new targets to treat or at least limit brogenesis after lung injury. A number of molecular processes are involved in the initiation of EMT in ATII cells. 5 Components of the brino- lytic system such as urokinase-type plasminogen activator (uPA), uPA plasma membrane receptor (uPAR), and its major inhibitor, plasminogen activator inhibitor (PAI-1) are all elaborated by ATII cells. These proteins independently in- uence a broad range of biological processes germane to lung injury and its repair. 6 However, their role in brogenesis via EMT is unclear. Recent publications using bleomycin (BLM) 7 and a passive cigarette smoke (PCS) 8 or adenovirus expressing constitutively active transforming growth factor b Supported in part by Flight Q3 Attendant Medical Research Institute Clinical Innovator Award FAMRI-ID-082380 (S.S.), American Heart Association grant GRNT19020001 (S.S.), and NIH/National Heart, Lung, and Blood Institute grant HL093547 (S.S.). Disclosures: None declared. Copyright ª 2014 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved. http://dx.doi.org/10.1016/j.ajpath.2014.08.027 ajp.amjpathol.org The American Journal of Pathology, Vol. 185, No. 1, January 2015 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 FLA 5.2.0 DTD ĸ AJPA1868_proof ĸ 15 November 2014 ĸ 11:35 am ĸ EO: AJP14_0185

Role of the Urokinase-Fibrinolytic System in Epithelial–Mesenchymal Transition during Lung Injury

Embed Size (px)

Citation preview

Page 1: Role of the Urokinase-Fibrinolytic System in Epithelial–Mesenchymal Transition during Lung Injury

Q19

Q1Q2

Q4

Q5

The American Journal of Pathology, Vol. 185, No. 1, January 2015

1234567891011121314151617181920212223242526272829303132333435363738394041424344454647484950515253545556575859606162

ajp.amjpathol.org

6364656667686970717273747576777879

Role of the Urokinase-Fibrinolytic System inEpithelialeMesenchymal Transition during Lung InjuryAmarnath Satheesh Marudamuthu,* Yashodhar Prabhakar Bhandary,* Shwetha Kumari Shetty,* Jian Fu,y

Venkatachalem Sathish,z YS Prakash,z and Sreerama Shetty*

8081828384

From the Department of Medicine,* Texas Lung Injury Institute, University of Texas Health Science Center at Tyler, Tyler, Texas; the Center for Research onEnvironmental Disease and Toxicology,y College of Medicine, University of Kentucky, Lexington, Kentucky; and the Department of Anesthesiology,z MayoClinic, Rochester, Minnesota

858687

Accepted for publication

C

P

h

8889909192939495

August 28, 2014.

Address correspondence toSreerama Shetty, Ph.D., TexasLung Injury Institute, Depart-ment of Medicine, University ofTexas Health Science Center atTyler, Tyler, TX 75708.E-mail: [email protected].

opyright ª 2014 American Society for Inve

ublished by Elsevier Inc. All rights reserved

ttp://dx.doi.org/10.1016/j.ajpath.2014.08.027

96979899100101102103

FL

Alveolar type II epithelial (ATII) cell injury precedes development of pulmonary fibrosis. Mice lackingurokinase-type plasminogen activator (uPA) are highly susceptible, whereas those deficient in plasminogenactivator inhibitor (PAI-1) are resistant to lung injury and pulmonary fibrosis. Epithelialemesenchymaltransition (EMT) has been considered, at least in part, as a source of myofibroblast formation duringfibrogenesis. However, the contribution of altered expression of major components of the uPA system onATII cell EMT during lung injury is not well understood. To investigate whether changes in uPA and PAI-1by ATII cells contribute to EMT, ATII cells from patients with idiopathic pulmonary fibrosis and chronicobstructive pulmonary disease, and mice with bleomycin-, transforming growth factor be, or passivecigarette smokeeinduced lung injury were analyzed for uPA, PAI-1, and EMT markers. We found reducedexpression of E-cadherin and zona occludens-1, whereas collagen-I and a-smooth muscle actin wereincreased in ATII cells isolated from injured lungs. These changes were associated with a parallel increasein PAI-1 and reduced uPA expression. Further, inhibition of Src kinase activity using caveolin-1 scaf-folding domain peptide suppressed bleomycin-, transforming growth factor be, or passive cigarettesmokeeinduced EMT and restored uPA expression while suppressing PAI-1. These studies show thatinduction of PAI-1 and inhibition of uPA during fibrosing lung injury lead to EMT in ATII cells.(Am J Pathol 2015, 185: 1e14; http://dx.doi.org/10.1016/j.ajpath.2014.08.027)

Supported in part by Flight Q3Attendant Medical Research Institute ClinicalInnovator Award FAMRI-ID-082380 (S.S.), American Heart Associationgrant GRNT19020001 (S.S.), and NIH/National Heart, Lung, and BloodInstitute grant HL093547 (S.S.).

Disclosures: None declared.

104105106107108109110111112113114115116117118119120121122123

Idiopathic pulmonary fibrosis (IPF) and other interstitial lungdiseases are characterized by destruction of lung architecturedue to excessive deposition of extracellular matrix proteins byactivated fibroblasts or myofibroblasts, leading to progressivedyspnea and loss of lung function.1e3 The origins of myofi-broblasts participating in the pathological remodeling of IPFlungs are not clear. Histopathological evaluation demonstratesthat myofibroblasts accumulate in fibroblastic foci. Emergingevidence suggests that polarized type II alveolar epithelial(ATII) cells undergo epithelialemesenchymal transitions(EMT) after lung injury. The ATII cells assume phenotypicchanges such as increased migration, invasion, resistance toapoptosis, and production of elevated levels of extracellularmatrix proteins4,5 and therefore serve as a source of myofi-broblasts. Understanding the possible mechanisms contrib-uting to EMT in ATII cells may help identify new targets totreat or at least limit fibrogenesis after lung injury.

stigative Pathology.

.

A 5.2.0 DTD � AJPA1868_proof �

A number of molecular processes are involved in theinitiation of EMT in ATII cells.5 Components of the fibrino-lytic system such as urokinase-type plasminogen activator(uPA), uPA plasmamembrane receptor (uPAR), and its majorinhibitor, plasminogen activator inhibitor (PAI-1) are allelaborated by ATII cells. These proteins independently in-fluence a broad range of biological processes germane to lunginjury and its repair.6 However, their role in fibrogenesis viaEMT is unclear. Recent publications using bleomycin(BLM)7 and a passive cigarette smoke (PCS)8 or adenovirusexpressing constitutively active transforming growth factor b

124

15 November 2014 � 11:35 am � EO: AJP14_0185

Page 2: Role of the Urokinase-Fibrinolytic System in Epithelial–Mesenchymal Transition during Lung Injury

Marudamuthu et al

125126127128129130131132133134135136137138139140141142143144145146147148149150151152153154155156157158159160161162163164165166167168169170171172173174175176177178179180181182183184185186

187188189190191192193194195196197198199200201202203204205206207208209210211212213214215216217218219220221222223224225226227228229230231232233234235236237238239240241242243244245246247

(Ad-TGF-b)1,9 exposure model of lung injury indicate that acoordinate increase in PAI-1 and a decrement in uPA by ATIIcells promote lung injury and subsequent pulmonary fibrosis(PF). We also found that caveolin-1 scaffolding domainpeptide (CSP) acts as a competitor to caveolin-1, restoresexpression of uPA and uPAR, and inhibits PAI-1 in ATII cellsafter lung injury. These changes prevent development of PFafter lung injury.7 Recent literature suggests that up to 30% to50% of myofibroblasts may be derived via EMT duringfibrogenesis.10e12 However, an in vivo genetic lineage tracingstudy reported by Rock et al13 contradicts these findings. Ourobjective in the current study is to elucidate the role of alteredexpression of uPA, uPAR, and PAI-1 after lung injury inEMT, and further evaluate whether reinstatement of baselineexpression of uPA, uPAR, and PAI-1 by CSP interventionafter lung injury reduces EMT in ATII cells.

Materials and Methods

Isolation and Analysis of ATII Cells from IPF and COPDLung Tissues

IPF or chronic obstructive pulmonary disease (COPD), andcontrol (without overt IPF or COPD) donor lung sampleswere collected from patients undergoing thoracic surgery atSt. Mary’s Hospital (Rochester, MN). Protocols wereapproved by institutional review boards of Mayo Clinic(Rochester, MN) and The UT Health Science Center at Tyler(Tyler, TX). ATII cells were isolated from the IPF, COPD,and control lung tissues, and aliquots were stained for in-clusion bodies to assess the purity of cell preparation as wedescribed recently.8 The lysates from these cells wereimmunoblotted for expression of EMT markers. Lung sec-tions from patients with IPF and COPD, diffused alveolardamage, or healthy donors were provided by the Lung TissueResearch Consortium of the National Heart, Lung, and BloodInstitute or by the Department of Pathology at the UT HealthScience Center at Tyler. These sections were subjected toimmunohistochemical (IHC) analysis to assess changes inantigen levels for uPA, PAI-1, and EMT markers.

BLM- and PCS-Induced Lung Injury

Wild-type (WT) or uPA-, uPAR-, and PAI-1edeficient miceof C57BL/6 background were bred in our facilities or werepurchased from The Jackson Laboratory (Bar Harbor, ME).All animal experiments were performed according to approvedprotocols under the guidelines of the Animal Care and UseCommittee of The UT Health Science Center at Tyler. Forin vitro experiments, ATII cells were isolated from the lungs ofuninjured mice as we reported elsewhere.7,8 The purities ofATII cell preparations were approximately 90% to 95%, basedon lithium carbonate staining for inclusion bodies. These cellswere treated with 40 mg/mL BLM with or without 10 nmol/LCSP or control peptide (CP), or 2 ng/mL purified TGF-b1alone in Matrigel-coated culture dishes containing epithelial

2FLA 5.2.0 DTD � AJPA1868_proof �

cell growthesupplemented alveolar epithelial cell medium(AEpiCM) from ScienCell Research Laboratories (Carlsbad,CA) for 3 days.7,8,14e17 For in vivo studies, mice were exposedto 2 U BLM per kg body weight for 3 days or PCS for 20weeks with or without 18.75 mg of CSP or CP per kg bodyweight as described previously.7,8 ATII cells were isolated andused for Western blot analysis or real-time PCR analysis toassess changes in the expression of E-cadherin, ZO-1,collagen-I, and a-smooth muscle actin (a-SMA) protein andmRNA, respectively. Real-time PCR using primers for mouseE-cadherin, forward: 50-AATGGCGGCAATGCAATCCCA-AGA-30 and reverse: 50-TGCCACAGACCGATTGTGGA-GATA-30; mouse collagen-I, forward: 50-CCAAGGGTAAC-AGCGGTGAA-30 and reverse: 50-CCTCGTTTTCCTTC-TTCTCCG-30; mouse ZO-1, forward: 50-ATTCTGAAGA-AATGATGAGA-30 and reverse: 50-TCCTGATTGACCA-CTTTTAA-30; a-SMA, forward: 50-GGCTCTGGGCTCTG-TAAGG-30 and reverse: 50-CTCTTGCTCTGGGCTTCATC-30; and b-actin, forward: 50-CACCGCAGCTCGTAGCTCT-TCTCCAGGG-30 and reverse: 50-CCAGCCATGTACGT-TGCTATCCAG-30. Lung sections were separately assessedfor collagen-I, E-cadherin, a-SMA, and ZO-1 antigen levelsby IHC analysis. Lung homogenates and bronchoalveolarlavage fluids of mice exposed to BLMwith or without CSP orCP for 3 days were analyzed for active TGF-b byWestern blotanalysis. The findings were independently confirmed byenzyme-linked immunosorbent assay (ELISA) for activeTGF-b.

Effect of TGF-b Overexpression on ATII Cell EMT and PF

ATII cells isolated from WT mice were exposed to Ad-TGF-b in the presence or absence of 10 nmol/L CSP or CPas described above. ATII cells untreated or treated withempty adenovirus (Ad-EV) for 3 days were used as controls.For in vivo studies, WT mice were exposed to saline or Ad-EV or Ad-TGF-b (109 plaque forming units) throughintranasal instillation as described earlier.9 After 24 hours,mice exposed to Ad-TGF-b were treated with or without18.75 mg CSP or CP per kg body weight. ATII cells wereisolated 3 days after instillation of Ad-TGF-b and analyzedfor changes in EMT. Lung homogenates of WT mice wereanalyzed for total hydroxyproline contents 21 days aftertransduction with Ad-TGF-b to assess changes in PF as wedescribed earlier.7

Effect of p53-Binding uPA, uPAR, and PAI-1 mRNA 30-UTR Sequences on BLM-Induced ATII Cell EMT in Mice

The competitive inhibition of p53 from binding to endog-enous uPA, uPAR, and PAI-1 mRNAs in ATII cells byoverexpression of p53-binding chimeric uPA, uPAR, andPAI-1 30-untranslated region (UTR) sequences, concurrentlyrestores uPA and uPAR, and inhibits PAI-1 expressionwithout affecting BLM- or PCS-induced p53 in mice.8,18

Therefore, we exposed mice to an i.v. (through orbital

ajp.amjpathol.org - The American Journal of Pathology

248

15 November 2014 � 11:35 am � EO: AJP14_0185

Page 3: Role of the Urokinase-Fibrinolytic System in Epithelial–Mesenchymal Transition during Lung Injury

½F1�½F1�

½F2�½F2�

6

uPA-Fibrinolytic System in ATII Cell EMT

249250251252253254255256257258259260261262263264265266267268269270271272273274275276277278279280281282283284285286287288289290291292293294295296297298299300301302303304305306307308309310

311312313314315316317318319320321322323324325326327328329330331332333334335336337338339340341342343344345346347348349350351352353354355356357358359360361362363364365366367368369370371

plexus) injection of lentivirus vector containing SP-B pro-moter expressing p53-binding or nonbinding controlchimeric sequences of uPA, uPAR, and PAI-1 30-UTRmRNA, and the transduction efficiency was confirmed byexpression of luciferase in ATII cells as described previ-ously.18 Twenty-four hours after lentiviral transduction,these mice were exposed to BLM. ATII cells were isolated72 hours after initiation of BLM injury and analyzed forEMT markers by Western blot analysis.

Effect of Inhibition of uPA or PAI-1 Expression in PAI-1e and uPA-Deficient ATII Cells, Respectively, on EMT

ATII cells isolated from uninjured mice lacking PAI-1expression were treated with lentiviral vector expressinguPA shRNA to inhibit baseline uPA expression. PAI-1edeficient ATII cells exposed to control shRNA or naiveATII cells were used as controls. Similarly, ATII cells iso-lated from uPA-deficient mice were exposed to BLM in thepresence or absence of PAI-1 shRNA or control shRNA.The lysates from these mice were analyzed for uPA,E-cadherin, ZO-1, and a-SMA expression.

Generation of Retroviral Plasmid

Retrovirus vector (pLNCX) containing Src mutant Y416FcDNA was cotransfected with packaging plasmid pUMVCand auxiliary plasmid pCMV-VSV-G (Addgene, Cam-bridge, MA) using Lipofectamine 2000 (Life Technologies,Grand Island, NY) in 293T cells to obtain phage particles.The viral titers were measured using 293 T cells and latertransduced ATII cells isolated from mouse lungs.

Localization of Biotin-Labeled CSP in Mouse Lungs

To determine the distribution of CSP in the mouse lungs,mice were exposed to saline or BLM for 72 hours throughintranasal instillation. These mice were i.p. injected withbiotin-labeled CSP. After 24 hours, mice were euthanized,and the lung sections were analyzed for biotin-labeled CSPusing anti-biotin antibody.

Statistical Analysis

The statistical differences between various experimentalconditions were analyzed by t-test and one way analysis ofvariance. P < 0.05 was considered significant.

Results

ATII Cell EMT Is Associated with Increased Expressionof PAI-1 and Concurrent Inhibition of uPA in IPF andCOPD Lungs

ATII cell damage precedes development of PF, and restora-tion of uPA and inhibition of PAI-1 expression prevents

The American Journal of Pathology - ajp.amjpathol.orgFLA 5.2.0 DTD � AJPA1868_proof �

development of PF after fibrosing lung injury in animalmodels. Multiple recent studies indicate that EMT in ATIIcells promotes fibrogenesis after lung injury. Therefore, weisolated ATII cells from the human lung tissues and stainedfor inclusion bodies to assess the purity of ATII cell prepa-ration using lithium carbonate, which indicated purityapproximately 90% to 95% (Figure 1A). Next, we tested theATII cell lysates for changes in uPA and PAI-1 expression.ATII cells isolated from human IPF lungs showed a markedincrease in PAI-1 and reduction in uPA expression comparedto those extracted from histologically normal lungs(Figure 1B). ATII cells from IPF lungs showed suppressionof epithelial cell markers (E-cadherin and ZO-1) expression,whereas mesenchymal cell markers (collagen-I and a-SMA)were increased compared to their baseline expressions foundin control lungs. We also found similar changes in theexpression of uPA, PAI-1, and markers of EMT in ATII cellsfrom the lungs of patients with COPD. Consistent with theresults of Western blot analysis of isolated ATII cells, IHCanalyses of IPF and COPD lung sections showed increasedcollagen-I and inhibition of ZO-1 staining compared to theirexpression in control subjects (Figure 1C). Similar changeswere also observed in lung sections of patients with diffusedalveolar damage. This indicates a causal link between alteredATII cell collagen-I and ZO-1 expression, and EMT as aresult of chronic lung injury.

Effect of CSP on BLM-Induced EMT in ATII Cells

Because ATII cells from human IPF lungs showed increasedEMT and PAI-1 with reduction in uPA expression, we isolatedATII cells frommouse lungs and exposed them to BLM for 72hours and analyzed changes in the expression of E-cadherin,ZO-1, collagen-I, and a-SMA to assess EMT in vitro. Expo-sure of ATII cells to BLM suppressed expression of E-cad-herin and ZO-1, while increasing collagen-I and a-SMA(Figure 2A). ATII cells treated with the profibrogenic cyto-kine, TGF-b, also revealed a significant increase in EMTcompared to phosphate-buffered saline (PBS)-treated controlATII cells. Exposure of ATII cells to BLM with CSP restoredE-cadherin and ZO-1 expression while significantly reducingthe expression of collagen-I and a-SMA. However, thescrambled CP failed to affect BLM-induced EMT in murineATII cells. Because treatment of mouse ATII cells with puri-fied TGF-b protein increased EMT, we next transduced ATIIcells with Ad-TGF-b in the presence or absence of CSP or CPand tested for changes in EMT markers. Ad-TGF-b increasedEMT in ATII cells in comparison with those transduced withAd-EV (Figure 2B). Further, treatment of ATII cells with CSP,but not CP, Qsuppressed TGF-beinduced EMT. To determinewhether in vitro culture condition alters phenotypes of ATIIcells during the course of our experiments, we maintainedcontrol ATII cells isolated from uninjured mice in growthfactoresupplemented AEpiCM medium for 0 to 3 days andanalyzed for changes in surfactant proteins by Western blotanalysis. ATII cells cultured in AEpiCM retained baseline

3

372

15 November 2014 � 11:35 am � EO: AJP14_0185

Page 4: Role of the Urokinase-Fibrinolytic System in Epithelial–Mesenchymal Transition during Lung Injury

print&web4C=FPO

Figure 1 Q18Altered expressions of plasminogen activator inhibitor (PAI-1) and urokinase-type plasminogen activator (uPA) are associated with alveolar typeII (ATII) cell epithelialemesenchymal transition in idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary disease (COPD) lungs. A: ATII cellsisolated from lung explants were stained for inclusion bodies using lithium carbonate to assess the purity of the cell preparation. B: ATII cells isolated frompatients (n Z 3) with IPF (gray bars) or COPD (white bars) and from healthy donors. The lysates were analyzed for changes in the expression of uPA, PAI-1,collagen-I, a-SMA, E-cadherin, ZO-1, and b-actin. The fold changes in densities of individual bands are presented as a bar graph after normalization againstthe corresponding densities of b-actin antigens present in each sample. C: Paraffin-embedded sections from lung tissues of patients with IPF or COPD ordiffused alveolar damage (DAD) and healthy donors were subjected to IHC analysis for changes in collagen-I and ZO-1 antigen levels in situ. Representativephotomicrograph (from n Z 5) is shown. ****P < 0.0001.

Marudamuthu et al

373374375376377378379380381382383384385386387388389390391392393394395396397398399400401402403404405406407408409410411412413414415416417418419420421422423424425426427428429430431432433434

435436437438439440441442443444445446447448449450451452453454455456457458459460461462463464465466467468469470471472473474475476477478479480481482483484485486487488489490491492493494495

SP-A, SP-B, and SP-C expression at least for 3 days, indi-cating preservation of cellular phenotypes (Figure 2C).

Next, we exposed the mice to BLM through intranasalinstillation, and some mice were exposed to CSP or CP 24hours after initiation of BLM injury. Saline-treated micewere used as controls. The lung sections from these micewere subjected to IHC analysis for changes in EMT markersin situ. Following BLM-induced injury, lung sections ofmice showed a marked inhibition of ZO-1 antigen expres-sion, whereas collagen-I antigen levels were markedlyincreased in the lung parenchyma (Supplemental Figure S1).Further, treatment of mice with CSP markedly reducedBLM-induced EMT, which was confirmed by the changesin ZO-1 or collagen-I expression, whereas the response ofCP to BLM-induced EMT was negative. Because PCSexposure causes lung injury, albeit to a lesser extent thanBLM-induced lung injury in mice,7,8 and ATII cells isolatedfrom patients with COPD also showed increased EMT(Figure 1, B and C), we exposed mice to PCS and analyzedthe lung sections for changes in markers for EMT. PCSexposure increased collagen-I with concurrent suppressionof ZO-1 expression (Supplemental Figure S2). However,treatment with CSP markedly reversed PCS-inducedchanges in the EMT markers.

4FLA 5.2.0 DTD � AJPA1868_proof �

To further show these changes for alveolar epithelium, weisolated ATII cells from mice exposed to BLM (Figure 2D)or PCS (Figure 2E) and analyzed for changes in expressionof EMT markers. ATII cells collected from mice exposed tointranasal BLM or PCS showed increased expression ofcollagen-I and a-SMA, whereas epithelial cell markers suchas E-cadherin and ZO-1 were significantly suppressedcompared to baseline levels in mice without lung injury(Figure 2, D and E). Further, treatment of mice with CSPafter BLM or PCS exposure injury significantly preventedEMT. We next evaluated changes in E-cadherin, ZO-1,collagen-I, and a-SMA expression at the mRNA level.Consistent with protein expression and mRNA analysis byquantitative real-time PCR, the results indicated that BLMinjury increased EMT (Figure 2F). In addition, treatment ofmice with CSP attenuated BLM-mediated induction ofcollagen-I and a-SMA mRNA, and restored the expressionof E-cadherin and ZO-1 mRNA in ATII cells. There were nosignificant changes in the expression of EMT markers inBLM-injured mice treated with CP. Because TGF-b inducesEMT in ATII cells in vitro (Figure 2B), and is a majormediator of fibrosis in diverse organs, including lungs,19,20

we next tested whether CSP reverses ATII cell EMT inmice exposed to Ad-TGF-b. We found increased EMT in

ajp.amjpathol.org - The American Journal of Pathology

496

15 November 2014 � 11:35 am � EO: AJP14_0185

Page 5: Role of the Urokinase-Fibrinolytic System in Epithelial–Mesenchymal Transition during Lung Injury

Q7

Figure 2 Inhibition of bleomycin (BLM)-induced epithelialemesenchymal transitions (EMT) in ATII cells by caveolin-1 scaffolding domain peptide (CSP).A: ATII cells isolated from WT mice were treated with PBS or 40 mg/mL BLM alone or BLM with 10 nmol/L CSP or control peptide (CP) for 72 hours in culturedishes at 37�C. The lysates were immunoblotted for changes in the expression of collagen-I, a-SMA, E-cadherin, ZO-1, and b-actin antigens. The lysates fromATII cells treated with 2 ng/mL transforming growth factor b (TGF-b) were used as a positive control for comparison. The densities of individual bands werenormalized against the corresponding densities of b-actin. The fold changes in proteins are presented as a bar graph. Differences between PBS control and BLMor BLM and BLMþCSP groups are statistically significant. B: ATII cells isolated from WT mice were exposed to PBS, Ad-EV, or Ad-TGF-b with or without CSP orCP in culture dishes. After 72 hours, the lysates were analyzed for changes in EMT markers by Western blot analysis. C: ATII cells isolated from WT mice werecultured in Matrigel-coated plastic dishes for 0 to 3 days at 37�C. The lysates were later analyzed for changes in SP-A, SP-B, and SP-C expression by Westernblot analysis. D: WT mice were exposed to saline or 2 U/kg body weight BLM through intranasal instillation. After 24 hours, mice exposed to BLM were i.p.injected with or without 18.75 mg/kg body weight of CSP or CP. Three days after BLM injury, ATII cells were isolated and the lysates were immunoblotted forchanges in the expression of collagen-I, a-SMA, E-cadherin, ZO-1, and b-actin. The densities of individual bands were measured and normalized against thecorresponding densities of b-actin. The fold changes in proteins are presented as a bar graph. E: WT mice were exposed to ambient air or PCS from 40cigarettes (approximately 90 mg/m3 total solid particulates) using a mechanical smoking chamber over a 2-hour period for 5 days per week. After 4 weeks ofPCS exposure, mice exposed to PCS were i.p. injected with or without 18.75 mg/kg body weight of CSP or CP once every week for 4 more weeks. After 20 weeksof PCS exposure, ATII cells were isolated, and the lysates were tested for changes in the expression of collagen-I, a-SMA, E-cadherin, ZO-1 and b-actin byWestern blot analysis. F: Total RNA isolated from ATII cells of mice treated with saline, BLM, BLMþCSP, or CP were analyzed for changes in the expression ofE-cadherin, collagen-I, ZO-1, and a-SMA mRNAs by quantitative real-time PCR. Changes in their expression levels were normalized to the corresponding levelsof b-actin transcripts. The data were presented relative to that of saline-treated control groups. G: WT mice were exposed to saline, Ad-EV, or Ad-TGF-b byintranasal instillation. Twenty-four hours later, mice transduced with Ad-TGF-b were treated with or without CSP or CP. ATII cells were isolated from these mice72 hours after exposure to TGF-b and analyzed for changes in EMT markers by Western blot analysis. H: WT mice treated with Ad-EV or Ad-TGF-b alone or Ad-TGF-b with CSP or CP as described in G were euthanized 21 days after initial transduction with Ad-TGF-b. The lung homogenates were analyzed for totalhydroxylproline contents to assess changes in lung fibrosis. I: Mice were i.v. (via orbital plexus) injected with or without lentivirus expressing p53-binding ornonep53-binding control chimeric 30-UTR sequences of uPA/uPAR/PAI-1 mRNA 30-UTR as described elsewhere.14 Twenty-four hours later, the mice wereexposed to BLM, and ATII cells were isolated 72 hours after inception of BLM injury. ATII cells isolated from mice exposed to saline were used as control forcomparison. The lysates were tested for changes in the expression of collagen-I, a-SMA, E-cadherin, ZO-1, and b-actin. ****P < 0.0001 versus control.

uPA-Fibrinolytic System in ATII Cell EMT

497498499500501502503504505506507508509510511512513514515516517518519520521522523524525526527528529530531532533534535536537538539540541542543544545546547548549550551552553554555556557558

559560561562563564565566567568569570571572573574575576577578579580581582583584585586587588589590591592593594595596597598599600601602603604605606607608609610611612613614615616617618619

ATII cells from mice transduced with Ad-TGF-b comparedto those exposed to Ad-EV (Figure 2G). Consistent withBLM-induced injury, treatment of mice exposed to Ad-TGF-b with CSP, but not CP, suppressed EMT in ATIIcells. Further analysis of lung tissues for total hydroxypro-line contents confirmed increased PF in mice exposed toAd-TGF-b 21 days later (Figure 2H). This was significantly

The American Journal of Pathology - ajp.amjpathol.orgFLA 5.2.0 DTD � AJPA1868_proof �

suppressed in mice exposed to Ad-TGF-b and later treatedwith CSP.

ATII cells isolated from IPF and COPD showed increasedEMT associated with a parallel increase in PAI-1 and inhibi-tion of uPA expression (Figure 1). p53 concurrently inhibitsuPA and uPARwhile inducing PAI-1 expression in ATII cellsduring BLM or PCS exposure injury by binding unique

5

620

15 November 2014 � 11:35 am � EO: AJP14_0185

Page 6: Role of the Urokinase-Fibrinolytic System in Epithelial–Mesenchymal Transition during Lung Injury

½F3�½F3�

Q8

½F4�½F4�

½F5�½F5�

Marudamuthu et al

621622623624625626627628629630631632633634635636637638639640641642643644645646647648649650651652653654655656657658659660661662663664665666667668669670671672673674675676677678679680681682

683684685686687688689690691692693694695696697698699700701702703704705706707708709710711712713714715716717718719720721722723724725726727728729730731732733734735736737738739740741742743

30-UTR sequences.7,8,18 CSP inhibits BLM- or PCS-inducedATII cell injury and prevents BLM-induced development ofPF. The process involves inhibition of p53 expression andreversal of p53-mediated downstream changes in uPA, uPAR,and PAI-1. To independently confirm whether p53-mediatedchanges in uPA, uPAR, and PAI-1 expression in ATII cellscontribute to EMT after BLM lung injury, we transducedmouse lung ATII cells with lentivirus expressing p53-bindingsequences under the control of SP-B promoter in vivo. Thesemice were later exposed to BLM, and ATII cells were isolatedand tested for changes in EMT. Overexpression of p53-binding 30-UTR sequences significantly suppressed BLM-induced EMT in ATII cells (Figure 2I). However, miceexposed to control sequence showed increased EMT in ATIIcells after exposure to BLM.

Role of uPA in CSP-Mediated Inhibition ofBLM-Induced ATII Cell EMT

ATII cells from COPD and IPF lungs showed reduction inuPA expression (Figure 1). Studies suggests that mice lack-ing uPA expression are highly susceptible to BLM-inducedPF. We further found that treatment of uPA-deficient micewith CSP after BLM injury failed to prevent the developmentof PF.7 To determine whether CSP-mediated inhibition ofATII cell EMT in WT mice required restoration of uPAexpression, we isolated ATII cells from uninjured uPA-deficient mice. The purity of ATII cell preparation wasconfirmed by lithium carbonate staining for inclusion bodies(Supplemental Figure S3A). These cells were treated withBLM in the presence or absence of CSP or CP in vitro.Treatment of uPA-deficient mouse ATII cells with eitherBLM or TGF-b in vitro, significantly induced EMT(Figure 3A). Interestingly, unlike WT ATII cells, ATII cellslacking uPA expression with BLM injury failed to respond toCSP treatment. Our results suggest that restoration of uPAexpression after BLM injury is critical in the control of EMT.

We then exposed uPA-deficient mice to BLM with orwithout CSP or CP, and analyzed the lung tissues for EMT insitu by IHC. uPA-deficient mice exposed to BLM showedincreased expression of collagen-I and a-SMA, whereas ZO-1antigen levels were significantly reduced after BLM injury(Supplemental Figure S4). Interestingly, unlike WT mice,uPA-deficient mice with BLM-induced lung injury failed torespond to CSP treatment. Analysis of isolated ATII cells fromthese mice, for changes in the expression of EMT markers byWestern blot analysis, indicated an increased EMT comparedto their baseline levels in uPA-deficient mice without lunginjury (Figure 3B). Treatment of uPA-deficient mice witheither CSP or CP after BLM injury failed to reverse ATII cellEMT that was due to BLM injury. mRNA analyses forchanges in the expression of EMT markers in uPA-deficientmice with BLM injury further confirmed that CSP failed torestore their expression (Figure 3C). These data indicate theimportance of uPA expression by ATII cells to prevent EMTduring BLM-induced lung injury. The literature21,22 suggests

6FLA 5.2.0 DTD � AJPA1868_proof �

that uPA through plasmin generation can activate extracellularmatrixetrapped TGF-b, whereas ATII cells lacking uPAexpression resist CSP-mediated suppression of EMT afterBLM injury. Therefore, we analyzed lung homogenates andbronchoalveolar lavage fluids from WT and uPA-deficientmice for active TGF-b by Western blot analysis (Figure 3D)and ELISA (Figure 3E). Our results confirmed that BLM lunginjury increased active TGF-b levels inWT and uPA-deficientmice, which was significantly suppressed in WT mice withBLM injury exposed to CSP.

Role of uPAR in ATII Cell EMT

Because most of the cellular functions of uPA are dependenton its binding to uPAR, we also isolated ATII cells fromuPAR-deficient mice, confirmed the purity by lithium car-bonate staining for inclusion bodies (SupplementalFigure S3B), and used these cells to study the role ofuPAR in EMT. Consistent with the responses of WT anduPA-deficient ATII cells, ATII cells lacking uPAR showedincreased EMT following exposure to BLM. Treatment witheither CSP or CP failed to significantly attenuate EMTinduced in ATII cells because of BLM (Figure 4A). Tofurther confirm the above findings in vivo, we exposeduPAR-deficient mice to BLM and treated them with CSP orCP. ATII cells were isolated and analyzed for changes inEMT markers. Reduced E-cadherin and ZO-1 with increasedcollagen-I and a-SMA expression indicated EMT in ATIIcells isolated from mice after BLM injury (Figure 4B).Further, uPAR-deficient mice exposed to BLM resisted CSPtreatment and showed increased ATII cell EMT. The ana-lyses of ATII cells for changes in E-cadherin, ZO-1,collagen-I, and a-SMA mRNAs further proved that ATIIcell-specific uPAR expression is also required for CSP toinhibit BLM-induced EMT (Figure 4C).

Role of PAI-1 Expression in ATII Cell EMT

PAI-1 expression is increased during BLM- or PCS-inducedlung injury, and mice that lack PAI-1 expression resist ATIIcell injury and subsequent development of PF.7,8 Weinvestigated whether PAI-1 expression contributes to EMTin ATII cells. ATII cells lacking PAI-1 when exposed toBLM in vitro, resisted inhibition of E-cadherin and ZO-1(Figure 5A). Interestingly, baseline expression levels ofcollagen-I and a-SMA in PAI-1edeficient ATII cellswithout BLM injury were highly elevated and comparablewith WT ATII cells with BLM injury (Figure 2A). How-ever, treatment of PAI-1edeficient ATII cells with BLMalone, or BLMþCSP, or BLMþCP failed to significantlyalter the baseline levels of E-cadherin, ZO-1, collagen-I, anda-SMA expression.Analysis of ATII cells isolated from the lungs of PAI-

1edeficient mice exposed to BLM showed little inhibitionof basal E-cadherin and ZO-1 expression in vivo(Figure 5B). Similarly, in mice lacking PAI-1 expression,

ajp.amjpathol.org - The American Journal of Pathology

744

15 November 2014 � 11:35 am � EO: AJP14_0185

Page 7: Role of the Urokinase-Fibrinolytic System in Epithelial–Mesenchymal Transition during Lung Injury

Q9

10

A B C

ββ-actin

D E

TGF-

CP

BLM

CSP

- + + +- - + -- - - +

WT

- + + +- - + -- - - +

uPA–/–

0

100

200

300

400

500

600

Saline BLM BLM+CSP****

*

*

BLM+CP

WT uPA–/–

******** ****

****

TGF-β

in B

ALF

(pg/

mL)

0

1

2

3

4

BLMBLM+CSPBLM+CP

****

Collagen-I

Fol

d C

hang

e in

mR

NA

vs s

alin

e co

ntro

l

****

α-SMA

**** **** **** ****

**** ******** **** ****

****

0.0

1.1

2.2

3.3

BLMBLM+CSPBLM+CP

****

E-CadherinCollagen-I ZO-1

Fold

cha

nge

in p

rote

invs

PB

S co

ntro

l

0.0

1.1

2.2

3.3

BLMBLM+CSPBLM+CP

****

E-CadherinCollagen-I ZO-1

Fold

cha

nge

in p

rote

invs

sal

ine

cont

rol

E-Cadherin

ZO-1

Collagen-Iα-SMA

α-SMA

β-actin

α-SMA

α-SMA

β-actin

β

BLM

CSP

CP

TGF-

βTGF-

- + + + -- - + - -- - - + -- - - - +

E-CadherinZO-1

Collagen-I

CP

BLM

CSP

- + + +- - + -- - - +

************ ********

******************** ****************

******** ******** **************** ****************

β

Figure 3 Role of urokinase-type plasminogen activator (uPA) in ATII cell epithelialemesenchymal transitions after bleomycin (BLM) injury. A: ATII cellsisolated from mice deficient in uPA expression were treated with PBS, BLM, BLMþCSP, or BLMþCP in vitro. The lysates were tested for the expression ofcollagen-I, a-SMA, E-cadherin, ZO-1, and b-actin antigens by Western blot analysis. The lysates from uPA-deficient ATII cells treated with TGF-b were used as acontrol for comparison. The individual bands were quantitated and normalized to the corresponding values of b-actin loading controls and the fold changes inproteins are presented as a bar graph. Differences between treatments versus control are statistically significant. B: Mice deficient in uPA expression wereexposed to saline or BLM as described in Figure 2D. Twenty-four hours after initial exposure to BLM, mice were i.p. injected with or without caveolin-1scaffolding domain peptide (CSP) or control peptide (CP). Seventy-two hours after initiation of BLM injury, ATII cells were isolated and the lysates wereevaluated for changes in the expression of collagen-I, a-SMA, E-cadherin, ZO-1, and b-actin by Western blot analysis. The densities of individual bands werenormalized against the corresponding densities of b-actin protein, and the fold changes in proteins are presented as a bar graph. C: Total RNA isolated fromATII cells of uPA-deficient mice exposed to saline, BLM, BLMþCSP, or CP were analyzed for changes in the expression of E-cadherin, collagen-I, ZO-1, anda-SMA mRNA levels by real-time PCR. The data were normalized with corresponding levels of b-actin mRNA. The fold changes of mRNA are presented as a bargraph. D: The lung homogenates of WT and uPA-deficient mice exposed to saline, BLM, BLMþCSP, or BLMþCP were tested for active TGF-b by Western blotanalysis. E: The bronchoalveolar lavage fluids (BALF) of WT and uPA-deficient mice exposed to saline, BLM, BLMþCSP, or BLMþCP were analyzed for activeTGF-b byELISA. ****P < 0.0001 versus control (A, D, E, and F).

uPA-Fibrinolytic System in ATII Cell EMT

745746747748749750751752753754755756757758759760761762763764765766767768769770771772773774775776777778779780781782783784785786787788789790791792793794795796797798799800801802803804805806

807808809810811812813814815816817818819820821822823824825826827828829830831832833834835836837838839840841842843844845846847848849850851852853854855856857858859860861862863864865866867

BLM failed to augment baseline levels of collagen-I and a-SMA, which were unusually high in ATII cells. Treatmentof PAI-1edeficient mice, exposed to BLM with either CSPor CP, failed to affect baseline expression of E-cadherin,ZO-1, collagen-I, and a-SMA further. Analysis of ATII cellmRNA for EMT markers (Figure 5C) likewise demonstratedresistance of PAI-1edeficient mice to BLM.

Mice deficient in uPA expression exhibit increased EMTand develop PF after BLM injury, whereas those lackingPAI-1 resist both EMT and PF. Therefore, we next inhibiteduPA expression in ATII cells obtained from PAI-1edeficientmice to determine whether increased uPA expression, as aresult of a lack of PAI-1, provided resistance against BLM-induced EMT. Inhibition of uPA expression by at least 75% to80% using shRNA failed to alter E-cadherin, ZO-1, collagen-I,

The American Journal of Pathology - ajp.amjpathol.orgFLA 5.2.0 DTD � AJPA1868_proof �

and a-SMA expression in PAI-1edeficient ATII cells(Figure 5D). On the contrary, inhibition of PAI-1 expressionusing shRNA in uPA-deficient ATII cells resisted BLM-induced EMT, supporting the pivotal role played byincreased PAI-1 expression in the induction of ATII cell EMT.

Role of Activation of Src Kinase in BLM-Induced ATIICell EMT

BLM and TGF-b lung injuries increase Src kinase activationand induction of PAI-1 expression is further dependent onactivation of Src kinases Q.23 To investigate the involvement ofSrc kinase activation in ATII cell EMT, we tested ATII celllysates for changes in the phosphorylation of tyrosine residues418 (Y418) and 527 (Y527). We found that ATII cells isolated

7

868

15 November 2014 � 11:35 am � EO: AJP14_0185

Page 8: Role of the Urokinase-Fibrinolytic System in Epithelial–Mesenchymal Transition during Lung Injury

½F6�½F6�

C

A

0.0

1.2

2.4

3.6BLMBLM+CSPBLM+CPTGF-ββ

Collagen-I

Collagen-I

ZO-1α-SMA

α-SMA

Fold

Cha

nge

in p

rote

invs

PB

S co

ntro

l

0.0

1.2

2.4

3.6 BLMBLM+CSPBLM+CP

ZO-1

Fold

Cha

nge

in p

rote

invs

sal

ine

cont

rol

0

1

2

3

4

BLMBLM+CSPBLM+CP

E-CadherinCollagen-I ZO-1α-SMA

Fold

cha

nge

in m

RN

Avs

sal

ine

cont

rol

B

BLM

CSP

CP

TGF-ββ

E-Cadherin

ZO-1

Collagen-Iα-SMA

α-SMA

β-actin

β-actin

- + + + -- - + - -- - - + -- - - - +

E-CadherinZO-1

Collagen-I

BLM

CSP

CP

- + + +- - + -- - - +

E-Cadherin

E-Cadherin

**** **** **** ****

**** **** ****

**** **** **** ****

**** **** ****

**** **** ******** **** ****

**** **** ******** **** ****

******** ****

****

**** ****

**** **** ********

**** **** **** ****Figure 4 Effect of uPA plasma membranereceptor (uPAR) expression on bleomycin (BLM)-induced ATII cell epithelialemesenchymal transi-tions. A: ATII cells isolated from mice lackinguPAR expression were exposed to PBS, BLM,BLMþCSP, or BLMþCP in vitro. The lysates wereimmunoblotted for the expression of collagen-I,a-SMA, E-cadherin, ZO-1, and b-actin antigens.The lysates from uPAR-deficient ATII cells treatedwith transforming growth factor b (TGF-b) wereused as a control. The densities of individualbands were compiled and normalized againstcorresponding values of b-actin loading controls.The fold changes in proteins are presented as a bargraph, and the differences between treatmentsversus control were statistically significant. B:uPAR-deficient mice were treated with saline orBLM and 24 hours after the initial exposure toBLM, mice with BLM injury were i.p. injected withor without caveolin-1 scaffolding domain peptide(CSP) or control peptide (CP). Three days after theinitiation of BLM injury, ATII cells were isolatedfrom these mice, and the cell lysates were immu-noblotted for altered expression of collagen-I,a-SMA, E-cadherin, ZO-1, and b-actin. The foldchanges of proteins are presented as a bar graph. C:ATII cell RNA from uPAR-deficient mice exposed tosaline, BLM, BLMþCSP, or CP were tested forchanges in the expression of E-cadherin, collagen-I,ZO-1, and a-SMA mRNAs by real-time PCR. The foldchanges of mRNA are presented as a bar graph.****P < 0.0001 versus control.

Marudamuthu et al

869870871872873874875876877878879880881882883884885886887888889890891892893894895896897898899900901902903904905906907908909910911912913914915916917918919920921922923924925926927928929930

931932933934935936937938939940941942943944945946947948949950951952953954955956957958959960961962963964965966967968969970971972973974975976977978979980981982983984985986987988989990991

fromWT (Figure 6A), uPA-deficient (Figure 6B), and uPAR-deficient (Figure 6C) mice exposed to BLM show increasedY418 phosphorylation, indicating activation of Src kinase. Thiswas reversed after treatment of WT mice with CSP throughinhibitory phosphorylation (Y527) of Src kinase. However,treatment of uPA- or uPAR-deficient mice with CSP failed toinhibit BLM-induced activation of Src kinase (Figure 6, B andC) or EMT (Figures 3 and 4). Interestingly, BLM failed toincrease phosphorylation of Y418 and inhibit phosphorylationof Y527 Src kinases from baseline levels observed in controlmice lacking PAI-1 expression (Figure 6D). Our data thereforesuggest that induction of Src activation in ATII cells by BLMcontributes to EMT. This is reversed by CSP-mediatedinhibitory (Y527) phosphorylation of Src kinases. The pro-cess involves inhibition of BLM-induced PAI-1 expressionand restoration of baseline levels of uPA and uPARexpression.

Microscopic examination of lung sections of mice i.p.injected with biotin-labeled CSP revealed that the peptideCSP was predominantly distributed in the alveolar andairway epithelium of the BLM-injured lungs compared withuninjured epithelium of the saline-treated lungs. This in-dicates that the lung injury facilitates CSP adsorption(Supplemental Figure S5).

To further confirm that BLM-induced activation of Src ki-nase contributes to ATII cell EMT, we treated ATII cells with

8FLA 5.2.0 DTD � AJPA1868_proof �

BLM in the presence of Src kinase inhibitor PP2,24 and thecontrol cells received PP3. Inhibition of Src activationsignificantly suppressed BLM-induced EMT in ATII cells(Figure 6E). These changes were associated with parallel in-hibition of BLM-induced PAI-1 expression and restoration ofuPA expression. Because BLM induces Src activation throughY418, we transduced ATII cells with dominant-negative Y418Fmutant Src kinase and exposed these cells to BLM. The lysateswere tested for changes in EMTmarkers. Our data indicate thattransduction of ATII cells with Y418F mutant Src kinase failedto induce EMT after exposure to BLM (Figure 6F). Becauseactivated Src kinase phosphorylates caveolin-1 during renalepithelial cell EMT,25,26 and CSP inhibits activation of Srckinase, we analyzed ATII cells for changes in total and Y14

phosphorylated caveolin-1. BLM injury increased both totaland Y14 phosphorylated forms of caveolin-1 in ATII cells ofWT and uPA-deficient mice (Figure 6G). However in WTmice, CSP suppressed Y14 phosphorylation of caveolin-1probably through inhibition of Src kinase activation withoutaffecting total caveolin-1, which is otherwise increased afterBLM lung injury. Interestingly, CSP failed to suppress Srcactivation or Y14 phosphorylation of caveolin-1 in uPA- oruPAR-deficient mice, indicating the inevitability of CSP-mediated uPA and uPAR expression in inactivation of Srckinase through Y527 phosphorylation7 and downstream Y14

phosphorylation of caveolin-1. Consistent with Src activation,

ajp.amjpathol.org - The American Journal of Pathology

992

15 November 2014 � 11:35 am � EO: AJP14_0185

Page 9: Role of the Urokinase-Fibrinolytic System in Epithelial–Mesenchymal Transition during Lung Injury

A

B

C

0.0

0.4

0.8

1.2BLM BLM+CSP BLM+CP TGF-ββ

E-CadherinCollagen-I ZO-1α-SMA

Fold

cha

nge

in p

rote

invs

PB

S co

ntro

l

0.0

0.4

0.8

1.2 BLM BLM+CSP BLM+CP

E-Cadherin 1-OZCollagen-I α-SM A

Fold

cha

nge

in p

rote

invs

sal

ine

cont

rol

0.0

0.4

0.8

1.2BLM BLM+CSP BLM+CP

E-Cadherin 1-OZCollagen-I α-SMA

Fold

cha

nge

in m

RN

Avs

sal

ine

cont

rol

E-Cadherin

BLM

CSP

CP

TGF-ββ

ZO-1

Collagen-I

α-SMA

α-SMA

β-actin

β-actin

α-SMA

β-actin

α-SMA

β-actin

- + + + -- - + - -- - - + -- - - - +

BLM

CSP

CP

E-Cadherin

ZO-1

Collagen-I

- + + +- - + -- - - +

Control ShRNA

Collagen-I

E-CadherinZO-1

uPA

PAI-1 AT II cells

uPA ShRNA

- + -- - +

D

E-Cadherin

ZO-1

PAI-1

uPA AT II cells

BLM

Control ShRNA

PAI-1 ShRNA

- + + +- - + -- - - +

Figure 5 The contribution of plasminogen activator inhibitor (PAI-1) in bleomycin (BLM)-induced ATII cell epithelialemesenchymal transitions (EMT). A:ATII cells isolated from PAI-1edeficient mice were treated with PBS, BLM, BLMþCSP, or BLMþCP in vitro. The lysates were immunoblotted for changes incollagen-I, a-SMA, E-cadherin, ZO-1, and b-actin expression. The lysates from ATII cells lacking PAI-1 expression that were treated with transforming growthfactor b (TGF-b) were used as a control. The individual densities of bands were quantitated and normalized to the corresponding values of b-actin. The foldchanges in protein expression are presented as a bar graph. Differences between treatments versus control are not statistically significant. B: Mice lacking PAI-1expression were treated with saline or BLM, and 24 hours later, mice exposed to BLM were i.p. injected with or without caveolin-1 scaffolding domainpeptide (CSP) or control peptide (CP). Three days after initiation of BLM injury, ATII cells were isolated from saline, BLM-, BLMþCSP-, or BLMþCP-treatedmice. The lysates were examined for the expression of collagen-I, a-SMA, E-cadherin, ZO-1, and b-actin by Western blot analysis. The densities of bandswere normalized against the corresponding levels of b-actin. The bar graph represents the fold change of proteins. C: Total RNA isolated from ATII cells ofPAI-1edeficient mice exposed to saline, BLM, BLMþCSP, or CP were analyzed for changes in the expression of E-cadherin, ZO-1, collagen-I, and a-SMAmRNA levels by real-time PCR. The data were normalized with corresponding levels of b-actin mRNA. The fold changes of RNA are presented as a bar graph.D: ATII cells isolated from PAI-1edeficient or uPA-deficient mice were treated with lentivirus expressing uPA or PAI-1 shRNA. ATII cells obtained from bothuPA-deficient and PAI-1edeficient mice exposed to nonspecific shRNA or naive ATII cells were used as controls for comparison. Because BLM injury causesEMT in uPA-deficient mice, whereas those lacking PAI-1 expression resist BLM-induced EMT, ATII cells from uPA-deficient mice treated with or withoutPAI-1 shRNA or control shRNA were later exposed to BLM for 72 hours. The lysates from PAI-1edeficient and uPA-deficient ATII cells were analyzed forchanges in EMT markers by Western blot analysis.

uPA-Fibrinolytic System in ATII Cell EMT

9939949959969979989991000100110021003100410051006100710081009101010111012101310141015101610171018101910201021102210231024102510261027102810291030103110321033103410351036103710381039104010411042104310441045104610471048104910501051105210531054

1055105610571058105910601061106210631064106510661067106810691070107110721073107410751076107710781079108010811082108310841085108610871088108910901091109210931094109510961097109810991100110111021103110411051106110711081109111011111112111311141115

BLM or BLMþCSP failed to alter the baseline expression ofthe total or Y14 phosphorylated caveolin-1 level in PAI-1edeficient mice.

The expression of TGF-b, a profibrotic cytokine that in-duces EMT in ATII cells, was significantly increased after

The American Journal of Pathology - ajp.amjpathol.orgFLA 5.2.0 DTD � AJPA1868_proof �

BLM injury and is a major contributor to the developmentof PF in multiple organs, including lungs. We thereforeexposed the ATII cells to TGF-b alone or TGF-b in thepresence of TGF-b receptor kinase inhibitor (SB-431542)27

and found that TGF-b increased EMT, indicated by the

9

1116

15 November 2014 � 11:35 am � EO: AJP14_0185

Page 10: Role of the Urokinase-Fibrinolytic System in Epithelial–Mesenchymal Transition during Lung Injury

11

E

B DC

- + + + -- - + - -- - - + -- - - - +

- + + + -- - + - -- - - + -- - - - +

SB-431542 - - + - + +

PAI-1uPA

E-CadherinZO-1

Collagen-I

P-Y418-SRCP-Y527SRC

SRC

- + + + -- - + - -- - - + -- - - - +

A

BLM

CSPCP

- + + + -- - + - -- - - + -- - - - +TGF-ββ

TGF-ββ

SRC

β-actin

P-Y418SRCP-Y527SRC

F

PAI-1uPA

E-CadherinZO-1

Collagen-Iα-SMA

α-SMA

α-SMA

β-actin

β-actin

β-actinβ-actin-

P-Y418-SRC

PP2

BLM

PP3

- + + +

- - - + - - + -

SRC

E-CadherinZO-1

P-Y418-SRCSRC

- + - ++ + - -- - + +

BLM

EVSrcY418F

BLM

PAI-1–/–

G H

P-Cav-1-

Salin

e

Non

e

CSP

CP

Salin

e

Non

e

CP

BLM BLMWT uPA

–/–

CSP

Cav-1-

Salin

e

Non

e

CP

BLM

uPAR–/–

CSP

Salin

e

Non

e

CP

CSP

Figure 6 Effect of tyrosine phosphorylation of Src kinases on bleomycin (BLM)-induced ATII cell epithelialemesenchymal transitions (EMT) in WT, urokinase-type plasminogen activator (uPA)-, uPA plasma membrane receptor (uPAR)-, and plasminogen activator inhibitor (PAI-1)edeficient mice. AeD: ATII cellsisolated from WT (A), uPA-deficient (B), uPAR-deficient (C) and PAI-1edeficient (D) mice were treated with saline or BLM with or without caveolin-1 scaffoldingdomain peptide (CSP) or control peptide (CP) in culture dishes. The lysates were tested for changes in the phosphorylation of tyrosine (Y418 and Y527) residues ofSrc kinases by Western blot analysis using phospho-specific antibodies. The samemembrane was stripped and tested for total Src kinase and b-actin proteins. ATIIcell lysates treated with TGF-b were also used as controls. E: ATII cells were treated with PBS, BLM, BLM in the presence of 10 mmol/L Src kinase inhibitor PP2 orcontrol PP3 in culture dishes for 72 hours. The lysates were tested for changes in tyrosine Y418 phosphorylation of Src kinases, PAI-1, uPA, collagen-I, a-SMA,E-cadherin, ZO-1, and b-actin by Western blot analysis. F: ATII cells isolated from WT mice were transduced with retrovirus expressing dominant-negative Y418Fmutant Src kinase. ATII cells from WT mice exposed to empty vector (EV) were used as controls for comparison. After 24 hours, these cells were treated with BLM,and the lysates were analyzed for changes in EMT markers 72 hours after BLM injury. WT ATII cells treated with PBS or BLM alone were used as controls. The lysateswere analyzed for changes in EMT markers. G: ATII cells isolated fromWT and uPA-deficient mice were exposed to saline or BLM or BLM with CSP or CP for 72 hours,and the lysates were immunoblotted for total and phosphorylated caveolin-1 (P-Cav-1), and b-actin using specific antibodies. H: ATII cells were treated with PBS,TGF-b, or TGF-b in the presence of 10 mmol/L inhibitor of TGF-b receptor kinase (SB-431542) in culture dishes for 72 hours. The lysates were immunoblotted forchanges in tyrosine phosphorylation of Src kinases, PAI-1, uPA, collagen-I, a-SMA, E-cadherin, ZO-1, and b-actin by Western blot analysis.

Marudamuthu et al

11171118111911201121112211231124112511261127112811291130113111321133113411351136113711381139114011411142114311441145114611471148114911501151115211531154115511561157115811591160116111621163116411651166116711681169117011711172117311741175117611771178

1179118011811182118311841185118611871188118911901191119211931194119511961197119811991200120112021203120412051206120712081209121012111212121312141215121612171218121912201221122212231224122512261227122812291230123112321233123412351236123712381239

changes in the markers as well as Src activation (Figure 6H).These changes were associated with parallel induction ofPAI-1 and inhibition of uPA. However, inhibition of TGF-breceptor kinase significantly suppressed TGF-beinducedEMT, expression of PAI-1, and activation of Src kinase withrestoration of baseline uPA expression.

10FLA 5.2.0 DTD � AJPA1868_proof �

Discussion

uPA is mainly involved in extravascular proteolysis and tissueremodeling.28e30 uPAR, a glycosylphosphatidylinositol Q-linked receptor, is implicated in multiple uPA-mediatedcellular functions. Bronchoalveolar lavage fluids normally

ajp.amjpathol.org - The American Journal of Pathology

1240

15 November 2014 � 11:35 am � EO: AJP14_0185

Page 11: Role of the Urokinase-Fibrinolytic System in Epithelial–Mesenchymal Transition during Lung Injury

12

Figure 7 Regulation of ATII cell epithelialemesenchymal transitions(EMT) and pulmonary fibrosis (PF) through p53euPAefibrinolytic systemcross talk. Increased expression and phosphorylation of caveolin-1 byactivated Src kinases augments p53 expression in ATII cells during fibrosinglung injury. p53 in turn binds to urokinase-type plasminogen activator(uPA), uPA plasma membrane receptor (uPAR), and plasminogen activatorinhibitor (PAI-1) mRNAs leading to suppression of uPA and uPAR, andincreased PAI-1 expression.49e51 This results in enhanced ATII cell EMT anddevelopment of PF. Inhibition of p53 interaction with endogenous uPA,uPAR, and PAI-1 mRNA in ATII cells either by inhibiting p53 expressionusing CSP or competitive inhibition through overexpression of p53-binding30-UTR sequences of uPA, uPA, and PAI-1 mRNA restores uPA, uPAR, andPAI-1 expression and mitigates ATII cell EMT and development of PF afterfibrosing lung injury.

uPA-Fibrinolytic System in ATII Cell EMT

12411242124312441245124612471248124912501251125212531254125512561257125812591260126112621263126412651266126712681269127012711272127312741275127612771278127912801281128212831284128512861287128812891290129112921293129412951296129712981299130013011302

1303130413051306130713081309131013111312131313141315131613171318131913201321132213231324132513261327132813291330133113321333133413351336133713381339134013411342134313441345134613471348134913501351135213531354135513561357135813591360136113621363

contain high levels of uPA activity, but it is markedly reducedin patients with IPF, COPD, sarcoidosis, and adult respiratorydistress syndrome,2,31,32 as well as in mice with BLM- andPCS-induced lung injury because of increased local expressionof PAI-1.7,33 Proinflammatory stimuli such as TNF-a andTGF-b augment uPA, uPAR, and PAI-1 expression, andPAI-1 inhibits uPA activity and promotes cycling of uPAeuPARePAI-1.30 Lung epithelial cells are often targets of acuteand chronic lung injuries and a key driver of normal repairbecause of their constant contact with the outside environmentand their ability to suppress fibroblast growth.34

IPF is one of the most common forms of interstitial lungdiseases, characterized by apoptosis of ATII cells, alveolarfibrin deposition, depressed fibrinolysis, inflammation,myofibroblast accumulation, and progressive loss of lungfunction as a result of accumulation of extracellular matrixproteins.2,35,36 Myofibroblasts play a vital role in fibro-genesis after lung injury. The literature suggests that myo-fibroblasts arise from activation of resident lung fibroblasts,from differentiation of bone-marrowederived stromal cellsand also from ATII cells via EMT.1,37,38 Other reportssuggest increased expression of mesenchymal marker pro-teins and mRNAs by hyperplastic ATII cells in human IPFlungs4 and that approximately 30% to 50% of fibroblasts arealso derived via EMT in BLM-induced lung injury.11 Thissupports the importance of transformation of ATII cells intomyofibroblasts via EMT in the pathogenesis of PF12 inaddition to apoptosis of ATII cells.7,8 Most recent in vivogenetic lineage tracing studies using single-dose BLM inmice13 suggest that epithelial cells do not generate myofi-broblasts, thus undermining the importance of EMT in thecontext of PF. However, others studies using a mouse modelof either a single dose or repeated doses of BLM-inducedlung injury indicate that EMT promotes fibro-genesis.11,39,40 The discrepancies between studies could betechnical, which is beyond the scope of the present study.

Irrespective of whether or not reversible phenotypicchanges, such as EMT in ATII cells during lung injury,contribute to fibrosis, ATII cells in IPF and COPD lungsshow EMT with elevated PAI-1 and concurrent suppressionof uPA. Recently, Wang et al41 showed association ofepithelial overexpression of uPAR with EMT in COPDlungs. Although CS is a risk factor for lung fibrosis,42,43 it isa major contributor for lung carcinogenesis, and miceexposed to CS do not develop lung fibrosis despite theincreased EMT. In addition, increased senescence andapoptosis, rather than excess proliferation, of lung epithelialcells are hallmarks of COPD. Further, uPA and uPARprovides proproliferative and prosurvival signals in diversecell types, including lung epithelial cells,44,45 and micelacking uPA or uPAR resist tumor growth,46 whereas uPAand uPAR deficiency, rather than overexpression, contrib-utes to multiple organ fibrosis, including PF. This is furthersupported by the fact that lung injuries, including BLM- orCS-induced lung injury, are often associated with reduceduPA and uPAR expression because of excess expression of

The American Journal of Pathology - ajp.amjpathol.orgFLA 5.2.0 DTD � AJPA1868_proof �

PAI-1 and PAI-1emediated turnover of the uPAeuPARcomplex.18,47 Besides, DNA damage caused by BLM or CSor other injuries augments p53 in airway epithelial cells Q. p53in turn concurrently inhibits uPA and uPAR while inducingPAI-1 expression both at the transcriptional48,49 and post-transcriptional levels.50e52 Therefore, EMT associated withincreased uPAR in COPD lungs may be more related tocarcinogenesis than fibrosis of lungs or other organs.41

We further found that BLM-, TGF-be, and PCS-inducedlung injury drives ATII cells to EMT and, later, BLM- andTGF-beinduced PF in WT and uPA-deficient mice,whereas PAI-1edeficient mice resist EMT and PF. Wefound that tissues, as well as isolated ATII cells from thelungs of WT mice with BLM- or PCS-exposure injury,show induction of PAI-1, and reciprocal suppression of uPAand uPAR expression and apoptosis.7,8,18 Treatment of WTmice with CSP inhibited BLM-induced PAI-1 while aug-menting uPA and uPAR, and protected the mice againstATII cell apoptosis and development of PF.7 However, inuPA- and uPAR-deficient mice, CSP failed to mitigatePAI-1 expression or apoptosis in ATII cells 3 days afterBLM injury, and they exhibited severe PF when tested 21

11

1364

15 November 2014 � 11:35 am � EO: AJP14_0185

Page 12: Role of the Urokinase-Fibrinolytic System in Epithelial–Mesenchymal Transition during Lung Injury

Q13

14

½F7�½F7�

Marudamuthu et al

13651366136713681369137013711372137313741375137613771378137913801381138213831384138513861387138813891390139113921393139413951396139713981399140014011402140314041405140614071408140914101411141214131414141514161417141814191420142114221423142414251426

1427142814291430143114321433143414351436143714381439144014411442144314441445144614471448144914501451145214531454145514561457145814591460146114621463146414651466146714681469147014711472147314741475147614771478147914801481148214831484148514861487

days later.7 This is consistent with increased PAI-1, andinhibition of uPA and uPAR appears to promote ATII cellapoptosis, EMT, and fibrosis after lung injury in mice.

Our findings suggest that CSP, an intervention that targetsthis pathway, concurrently protects the lung epithelium fromapoptosis7,8 as well as EMT, and prevents PF after BLM-induced lung injury via uPA-mediated inhibition of PAI-1.The paradigms that drive ATII cells to these two oppositefates during lung injury are still unclear. However, we spec-ulate that EMT is part of a naturally evolving adaptive safe-guard response to protect ATII cells against apoptosis duringchronic lung injury. This postulate is supported by the fact thatsimultaneous apoptosis and EMT in ATII cells as a result ofendoplasmic reticulum stress contribute to the development ofPF.18,53 In addition, BLM and TGF-b concurrently promoteapoptotic death54 and EMT55e57 in lung epithelial cells, andapoptosis alone could contribute to the development of PF.7,58

PAI-1, a downstream product of TGF-b, simultaneouslycauses apoptosis,7,8,40 senescence,59,60 and EMT61 in diversecell types, including ATII cells.

It has been previously reported62 that macrophage surfaceactivation of TGF-b is dependent on uPAeuPAR-mediatedplasmin generation. However, we found that BLM lunginjury increased active TGF-b in WT and uPA-deficientmice, which is consistent with findings of silica-inducedfibrosing injury reported earlier.21,63 In addition, despitehaving increased plasminogen activator activity, micelacking PAI-1 expression resist EMT and organfibrosis.18,64,65 On the contrary, those deficient in uPA anduPAR expression exhibit accelerated fibrosis7,18,65,66 andlung-specific expression of uPA protecting them againstdevelopment of PF after BLM injury.67 PAI-1 expression isdisproportionately increased in injured lungs33 and alsoinduced by TGF-b,23 which in turn irreversibly suppressesboth uPA activity and its steady-state level by PAI-1emediated turnover of the uPAeuPAR complex. Theprotective effects of increased uPA and uPAR may involvesuppression of PAI-1 through increased uPA- and uPAR-mediated turnover of PAI-1 protein,47 or inhibition of p53expression and reversal of p53-mediated induction of PAI-1expression by increased uPA and uPAR,7,44,51,52 or both.

We found that dysregulation of uPA and PAI-1 expres-sion in ATII cells promotes both apoptosis and EMT afterBLM-, TGF-be, or PCS-induced lung injury, and reversalof these changes by CSP intervention prevents BLM- andTGF-beinduced EMT and PF.7 Therefore, inhibition ofEMT by CSP intervention may be associated with resolutionof lung injury. In addition, CSP requires uPA expression toinhibit BLM-induced PAI-1 and apoptosis7 or EMT. Thus,dysregulated alveolar fibrinolysis, as a result of increasedexpression of PAI-1 or inhibition of uPA and uPAR as aconsequence of lung injury, contributes to ATII cellapoptosis, EMT, and development of PF. uPA can induceitself or its cell surface receptor, uPAR,44,68 and inhibitPAI-1 expression,69 which requires uPA interaction withuPAR. Further, uPA- and uPAR-deficient mice resist CSP

12FLA 5.2.0 DTD � AJPA1868_proof �

treatment after BLM injury, whereas CSP is effective in WTmice. We therefore believe that protection against BLM-induced EMT by CSP requires both uPA and uPARexpression. On the basis of our recent work,7 we also sus-pect that the inability of CSP to mitigate ATII cell EMT, inboth uPA- or uPAR-deficient mice after BLM injury, is dueto a lack of uPA-mediated inhibition of PAI-1 expression.Interestingly, the basal expression of collagen-I and a-SMAis highly elevated in PAI-1edeficient ATII cells, which iscomparable to or even higher than that of WT ATII cellsexposed to BLM. However, PAI-1edeficient mice resistspontaneous or inducible ATII cell apoptosis and develop-ment of PF. Further, protection against development of PFin mice by transplantation of healthy ATII cells into lungs ofmice with BLM- or silica-induced injury57,63 underscoresthe importance of initial ATII cell apoptosis for subsequentPF. On the basis of these findings and a recent lineagestudy13 using a single-hit BLM model, we believe ATII cellapoptosis, rather than EMT, mediated by increased PAI-1probably plays a dominant role in the subsequent develop-ment of PF. EMT may be a reversible safeguard mechanismevolved to provide transitory protection against apoptosisand ATII cell damage through undergoing phenotypicchanges without significant impact on PF per se.Although several pathways could concurrently operate,

our studies indicate a direct role for uPA and PAI-1 in theprocess. We provide evidence that supports the role ofactivated Src kinase in facilitating BLM- or TGF-beinduced EMT in ATII cells. PP2 and Y418F mutant Srckinase attenuated BLM- and TGF-beinduced phosphory-lation of b-cateninY654 and EMT, and inhibited PAI-1 Q,24,70e72 while restoring ATII cell uPA expression.BLM-induced activation of Src kinase and phosphorylationof caveolin-1 was inhibited by CSP through phosphoryla-tion of Y527.7 These results suggest a role for increased Srckinase activation in EMT via induction of PAI-1 and inhi-bition of uPA and uPAR expression (Figure 7). This sup-ports the contention that apoptosis and EMT, as a result ofincreased PAI-1 by ATII cells, contributes to fibrogenesisafter BLM injury.

Acknowledgments

We thank Dr. Rui-Ming Liu (Department of EnvironmentalScience School of Public Health, University of Alabama,Birmingham, AL) for sharing the AdTGF-b1 virus.A.S.M., Y.P.B., and S.K.S. performed experiments,

analyzed data, and participated in the presentation of themanuscript; S.S. designed the study, interpreted the results,and drafted the manuscript; J.F. provided critical reagentsand cDNA constructs. V.S. and Y.S.P. classified patientsand provided critical patient samples and participated in theinterpretation of results, writing, and critical revision of themanuscript; and all of the authors reviewed and approvedthe final content of the manuscript.

ajp.amjpathol.org - The American Journal of Pathology

1488

15 November 2014 � 11:35 am � EO: AJP14_0185

Page 13: Role of the Urokinase-Fibrinolytic System in Epithelial–Mesenchymal Transition during Lung Injury

uPA-Fibrinolytic System in ATII Cell EMT

14891490149114921493149414951496149714981499150015011502150315041505150615071508150915101511151215131514151515161517151815191520152115221523152415251526152715281529153015311532153315341535153615371538153915401541154215431544154515461547154815491550

1551155215531554155515561557155815591560156115621563156415651566156715681569157015711572157315741575157615771578157915801581158215831584158515861587158815891590159115921593159415951596159715981599160016011602160316041605160616071608160916101611

Supplemental Data

Supplemental material for this article can be found athttp://dx.doi.org/10.1016/j.ajpath.2014.08.027.

References

1. Kim KK, Kugler MC, Wolters PJ, Robillard L, Galvez MG,Brumwell AN, Sheppard D, Chapman HA: Alveolar epithelial cellmesenchymal transition develops in vivo during pulmonary fibrosisand is regulated by the extracellular matrix. Proc Natl Acad Sci U S A2006, 103:13180e13185

2. Chapman HA: Disorders of lung matrix remodeling. J Clin Invest2004, 113:148e157

3. Kalluri R, Neilson EG: Epithelial-mesenchymal transition and itsimplications for fibrosis. J Clin Invest 2003, 112:1776e1784

4. Konigshoff M, Kramer M, Balsara N, Wilhelm J, Amarie OV,Jahn A, Rose F, Fink L, Seeger W, Schaefer L, Günther A,Eickelberg O: WNT1-inducible signaling protein-1 mediates pulmo-nary fibrosis in mice and is upregulated in humans with idiopathicpulmonary fibrosis. J Clin Invest 2009, 119:772e787

5. Zhong Q, Zhou B, Ann DK, Minoo P, Liu Y, Banfalvi A,Krishnaveni MS, Dubourd M, Demaio L, Willis BC, Kim KJ,duBois RM, Crandall ED, Beers MF, Borok Z: Role of endoplasmicreticulum stress in epithelial-mesenchymal transition of alveolarepithelial cells: effects of misfolded surfactant protein. Am J RespirCell Mol Biol 2011, 45:498e509

6. Shetty S, Bdeir K, Cines DB, Idell S: Induction of plasminogenactivator inhibitor-1 by urokinase in lung epithelial cells. J Biol Chem2003, 278:18124e18131

7. Bhandary YP, Shetty SK, Marudamuthu AS, Gyetko MR, Idell S,Gharaee-Kermani M, Shetty RS, Starcher BC, Shetty S: Regulation ofalveolar epithelial cell apoptosis and pulmonary fibrosis by coordinateexpression of components of the fibrinolytic system. Am J PhysiolLung Cell Mol Physiol 2012, 302:L463eL473

8. Shetty SK, Bhandary YP, Marudamuthu AS, Abernathy D,Velusamy T, Starcher B, Shetty S: Regulation of airway and alveolarepithelial cell apoptosis by p53-induced plasminogen activatorinhibitor-1 during cigarette smoke exposure injury. Am J Respir CellMol Biol 2012, 47:474e483

9. Huang WT, Vayalil PK, Miyata T, Hagood J, Liu RM: Thera-peutic value of small molecule inhibitor to plasminogen activatorinhibitor-1 for lung fibrosis. Am J Respir Cell Mol Biol 2012, 46:87e95

10. Degryse AL, Tanjore H, Xu XC, Polosukhin VV, Jones BR,McMahon FB, Gleaves LA, Blackwell TS, Lawson WE: Repetitiveintratracheal bleomycin models several features of idiopathic pul-monary fibrosis. Am J Physiol Lung Cell Mol Physiol 2010, 299:L442eL452

11. Tanjore H, Xu XC, Polosukhin VV, Degryse AL, Li B, Han W,Sherrill TP, Plieth D, Neilson EG, Blackwell TS, Lawson WE:Contribution of epithelial-derived fibroblasts to bleomycin-inducedlung fibrosis. Am J Respir Crit Care Med 2009, 180:657e665

12. Radisky DC, Kenny PA, Bissell MJ: Fibrosis and cancer: do myo-fibroblasts come also from epithelial cells via EMT? J Cell Biochem2007, 101:830e839

13. Rock JR, Barkauskas CE, Cronce MJ, Xue Y, Harris JR, Liang J,Noble PW, Hogan BL: Multiple stromal populations contribute topulmonary fibrosis without evidence for epithelial to mesenchymaltransition. Proc Natl Acad Sci U S A 2011, 108:E1475eE1483

14. Corti M1, Brody AR, Harrison JH: Isolation and primary culture ofmurine alveolar type II cells. Am J Respir Cell Mol Biol 1996, 14:309e315

15. Dobbs LG: Isolation and culture of alveolar type II cells. Am JPhysiol 1990, 258:L134eL147

The American Journal of Pathology - ajp.amjpathol.orgFLA 5.2.0 DTD � AJPA1868_proof �

16. Rice WR, Conkright JJ, Na CL, Ikegami M, Shannon JM,Weaver TE: Maintenance of the mouse type II cell phenotype in vitro.Am J Physiol Lung Cell Mol Physiol 2002, 283:L256eL264

17. Meng X, Ezzati P, Wilkins JA: Requirement of podocalyxin in TGF-beta induced epithelial mesenchymal transition. PLoS One 2011, 6:e18715

18. BhandaryYP, Shetty SK,MarudamuthuAS, Ji HL, Neuenschwander PF,BoggaramV,Morris GF, Fu J, Idell S, Shetty S: Regulation of lung injuryand fibrosis by p53-mediated changes in urokinase and plasminogenactivator inhibitor-1. Am J Pathol 2013, 183:131e143

19. Kim KK, Wei Y, Szekeres C, Kugler MC, Wolters PJ, Hill ML,Frank JA, Brumwell AN, Wheeler SE, Kreidberg JA, Chapman HA:Epithelial cell alpha3beta1 integrin links beta-catenin and Smadsignaling to promote myofibroblast formation and pulmonary fibrosis.J Clin Invest 2009, 119:213e224

20. Miyoshi K, Yanagi S, Kawahara K, Nishio M, Tsubouchi H,Imazu Y, Koshida R, Matsumoto N, Taguchi A, Yamashita S,Suzuki A, Nakazato M: Epithelial Pten controls acute lung injury andfibrosis by regulating alveolar epithelial cell integrity. Am J RespirCrit Care Med 2013, 187:262e275

21. Matrat M, Lardot C, Huaux F, Broeckaert F, Lison D: Role of uro-kinase in the activation of macrophage-associated TGF-beta in silica-induced lung fibrosis. J Toxicol Environ Health A 1998, 55:359e371

22. Munger JS, Harpel JG, Gleizes PE, Mazzieri R, Nunes I, Rifkin DB:Latent transforming growth factor-beta: structural features andmechanisms of activation. Kidney Int 1997, 51:1376e1382

23. Samarakoon R, Higgins CE, Higgins SP, Kutz SM, Higgins PJ:Plasminogen activator inhibitor type-1 gene expression and inducedmigration in TGF-beta1-stimulated smooth muscle cells is pp60 (c-src)/MEK-dependent. J Cell Physiol 2005, 204:236e246

24. Kong L, Deng Z, Shen H, Zhang Y: Src family kinase inhibitor PP2efficiently inhibits cervical cancer cell proliferation through down-regulating phospho-Src-Y416 and phospho-EGFR-Y1173. Mol CellBiochem 2011, 348:11e19

25. Bailey KM, Liu J: Caveolin-1 up-regulation during epithelial tomesenchymal transition is mediated by focal adhesion kinase. J BiolChem 2008, 283:13714e13724

26. Chen J, Chen JK, Harris RC: Angiotensin II induces epithelial-to-mesenchymal transition in renal epithelial cells through reactive ox-ygen species/Src/caveolin-mediated activation of an epidermalgrowth factor receptor-extracellular signal-regulated kinase signalingpathway. Mol Cell Biol 2012, 32:981e991

27. Inman GJ, Nicolas FJ, Callahan JF, Harling JD, Gaster LM,Reith AD, Laping NJ, Hill CS: SB-431542 is a potent and specificinhibitor of transforming growth factor-beta superfamily type I activinreceptor-like kinase (ALK) receptors ALK4, ALK5, and ALK7. MolPharmacol 2002, 62:65e74

28. Irigoyen JP, Munoz-Canoves P, Montero L, Koziczak M,Nagamine Y: The plasminogen activator system: biology and regu-lation. Cell Mol Life Sci 1999, 56:104e132

29. Andreasen PA, Kjoller L, Christensen L, Duffy MJ: The urokinase-type plasminogen activator system in cancer metastasis: a review.Int J Cancer 1997, 72:1e22

30. Andreasen PA, Egelund R, Petersen HH: The plasminogen activationsystem in tumor growth, invasion, and metastasis. Cell Mol Life Sci2000, 57:25e40

31. Gharaee-Kermani M, Hu B, Phan SH, Gyetko MR: The role ofurokinase in idiopathic pulmonary fibrosis and implication for ther-apy. Expert Opin Investig Drugs 2008, 17:905e916

32. Bertozzi P, Astedt B, Zenzius L, Lynch K, LeMaire F, Zapol W,Chapman HA Jr: Depressed bronchoalveolar urokinase activity inpatients with adult respiratory distress syndrome. N Engl J Med 1990,322:890e897

33. Olman MA, Mackman N, Gladson CL, Moser KM, Loskutoff DJ:Changes in procoagulant and fibrinolytic gene expression duringbleomycin-induced lung injury in the mouse. J Clin Invest 1995, 96:1621e1630

13

1612

15 November 2014 � 11:35 am � EO: AJP14_0185

Page 14: Role of the Urokinase-Fibrinolytic System in Epithelial–Mesenchymal Transition during Lung Injury

17

Marudamuthu et al

16131614161516161617161816191620162116221623162416251626162716281629163016311632163316341635163616371638163916401641164216431644164516461647164816491650165116521653165416551656165716581659166016611662166316641665166616671668166916701671167216731674

1675167616771678167916801681168216831684168516861687168816891690169116921693169416951696169716981699170017011702170317041705170617071708170917101711171217131714171517161717171817191720172117221723172417251726172717281729173017311732173317341735

34. Caniggia I, Tseu I, Rolland G, Edelson J, Tanswell AK, Post M:Inhibition of fibroblast growth by epithelial cells in fetal rat lung. AmJ Respir Cell Mol Biol 1995, 13:91e98

35. Thannickal VJ, Toews GB, White ES, Lynch JP III, Martinez FJ:Mechanisms of pulmonary fibrosis. Annu Rev Med 2004, 55:395e417

36. Selman M, Pardo A: Role of epithelial cells in idiopathic pulmonaryfibrosis: from innocent targets to serial killers. Proc Am Thorac Soc2006, 3:364e372

37. Aghajanova L, Horcajadas JA, Esteban FJ, Giudice LC: The bonemarrow-derived human mesenchymal stem cell: potential progenitor ofthe endometrial stromal fibroblast. Biol Reprod 2010, 82:1076e1087

38. Wynn TA: Integrating mechanisms of pulmonary fibrosis. J Exp Med2011, 208:1339e1350

39. Tanjore H, Cheng DS, Degryse AL, Zoz DF, Abdolrasulnia R,Lawson WE, Blackwell TS: Alveolar epithelial cells undergoepithelial-to-mesenchymal transition in response to endoplasmic re-ticulum stress. J Biol Chem 2011, 286:30972e30980

40. Willis BC, duBois RM, Borok Z: Epithelial origin of myofibroblastsduring fibrosis in the lung. Proc Am Thorac Soc 2006, 3:377e382

41. Wang Q, Wang Y, Zhang Y, Zhang Y, Xiao W: The role of uPAR inepithelial-mesenchymal transition in small airway epithelium of patientswith chronic obstructive pulmonary disease. Respir Res 2013, 14:67

42. Baumgartner KB, Samet JM, Stidley CA, Colby TV, Waldron JA:Cigarette smoking: a risk factor for idiopathic pulmonary fibrosis. AmJ Respir Crit Care Med 1997, 155:242e248

43. Zhang H, Liu H, Borok Z, Davies KJ, Ursini F, Forman HJ: Cigarettesmoke extract stimulates epithelial-mesenchymal transition throughSrc activation. Free Radic Biol Med 2012, 52:1437e1442

44. Shetty S, Idell S: Urokinase induces expression of its own receptor inBeas2B lung epithelial cells. J Biol Chem 2001, 276:24549e24556

45. Tkachuk N, Kiyan J, Tkachuk S, Kiyan R, Shushakova N, Haller H,Dumler I: Urokinase induces survival or pro-apoptotic signals inhuman mesangial cells depending on the apoptotic stimulus. BiochemJ 2008, 415:265e273

46. Gutierrez LS, Schulman A, Brito-Robinson T, Noria F, Ploplis VA,Castellino FJ: Tumor development is retarded in mice lacking thegene for urokinase-type plasminogen activator or its inhibitor, plas-minogen activator inhibitor-1. Cancer Res 2000, 60:5839e5847

47. Shetty S, Kumar A, Johnson A, Pueblitz S, Idell S: Urokinase re-ceptor in human malignant mesothelioma cells: role in tumor cellmitogenesis and proteolysis. Am J Physiol 1995, 268:L972eL982

48. Kunz C, Pebler S, Otte J,, von der Ahe D: Differential regulation ofplasminogen activator and inhibitor gene transcription by the tumorsuppressor p53. Nucleic Acids Res 1995, 23:3710e3717

49. Parra M, Jardí M, Koziczak M, Nagamine Y, Muñoz-Cánoves P: p53phosphorylation at serine 15 is required for transcriptional inductionof the plasminogen activator inhibitor-1 (PAI-1) gene by the alky-lating agent N-methyl-N’-nitro-N-nitrosoguanidine. J Biol Chem2001, 276:36303e36310

50. Shetty S, Velusamy T, Idell S, Shetty P, Mazar AP, Bhandary YP,Shetty RS: Regulation of urokinase receptor expression by p53: novel rolein stabilization of uPAR mRNA. Mol Cell Biol 2007, 27:5607e5618

51. Shetty S, Shetty P, Idell S, Velusamy T, Bhandary YP, Shetty RS:Regulation of plasminogen activator inhibitor-1 expression by tumorsuppressor protein p53. J Biol Chem 2008, 283:19570e19580

52. Shetty P, Velusamy T, Bhandary YP, Shetty RS, Liu MC, Shetty S:Urokinase expression by tumor suppressor protein p53: a novel rolein mRNA turnover. Am J Respir Cell Mol Biol 2008, 39:364e372

53. Thomas AQ, Lane K, Phillips J III, Prince M, Markin C, Speer M,Schwartz DA, Gaddipati R, Marney A, Johnson J, Roberts R,Haines J, Stahlman M, Loyd JE: Heterozygosity for a surfactantprotein C gene mutation associated with usual interstitial pneumonitisand cellular nonspecific interstitial pneumonitis in one kindred. Am JRespir Crit Care Med 2002, 165:1322e1328

54. Hagimoto N, Kuwano K, Inoshima I, Yoshimi M, Nakamura N,Fujita M, Maeyama T, Hara N: TGF-beta 1 as an enhancer of Fas-

14FLA 5.2.0 DTD � AJPA1868_proof �

mediated apoptosis of lung epithelial cells. J Immunol 2002, 168:6470e6478

55. Willis BC, Borok Z: TGF-beta-induced EMT: mechanisms and im-plications for fibrotic lung disease. Am J Physiol Lung Cell MolPhysiol 2007, 293:L525eL534

56. Alipio ZA, Jones N, Liao W, Yang J, Kulkarni S, Sree KK, Hauer-Jensen M, Ward DC, Ma Y, Fink LM: Epithelial to mesenchymaltransition (EMT) induced by bleomycin or TGF-b1/EGF in murineinduced pluripotent stem cell-derived alveolar Type II-like cells.Differentiation 2011, 82:89e98

57. Kasai H, Allen JT, Mason RM, Kamimura T, Zhang Z: TGF-beta1induces human alveolar epithelial to mesenchymal cell transition(EMT). Respir Res 2005, 6:56

58. Serrano-Mollar A, Nacher M, Gay-Jordi G, Closa D, Xaubet A,Bulbena O: Intratracheal transplantation of alveolar type II cells re-verses bleomycin-induced lung fibrosis. Am J Respir Crit Care Med2007, 176:1261e1268

59. Elzi DJ, Lai Y, Song M, Hakala K, Weintraub ST, Shiio Y: Plas-minogen activator inhibitor 1-insulin-like growth factor bindingprotein 3 cascade regulates stress-induced senescence. Proc Natl AcadSci U S A 2012, 109:12052e12057

60. Kortlever RM, Higgins PJ, Bernards R: Plasminogen activatorinhibitor-1 is a critical downstream target of p53 in the induction ofreplicative senescence. Nat Cell Biol 2006, 8:877e884

61. Jeong SS, Chun MK: Inhibition of plasminogen activator inhibitor-1expression in smoke exposed alveolar type II cells attenuates epi-thelialemesenchymal transition. Tuberc Respir Dis 2011, 70:462e473

62. Venkatraman L, Chia SM, Narmada BC, White JK, Bhowmick SS,Forbes Dewey C Jr, So PT, Tucker-Kellogg L, Yu H: Plasmin trig-gers a switch-like decrease in thrombospondin-dependent activationof TGF-b1. Biophys J 2012, 103:1060e1068

63. Spitalieri P, Quitadamo MC, Orlandi A, Guerra L, Giardina E,Casavola V, Novelli G, Saltini C, Sangiuolo F: Rescue of murinesilica-induced lung injury and fibrosis by human embryonic stemcells. Eur Respir J 2012, 39:446e457

64. Eitzman DT, McCoy RD, Zheng X, Fay WP, Shen T, Ginsburg D,Simon RH: Bleomycin-induced pulmonary fibrosis in transgenic micethat either lack or overexpress the murine plasminogen activatorinhibitor-1 gene. J Clin Invest 1996, 97:232e237

65. Zhang G, Kim H, Cai X, López-Guisa JM, Alpers CE, Liu Y,Carmeliet P, Eddy AA: Urokinase receptor deficiency acceleratesrenal fibrosis in obstructive nephropathy. J Am Soc Nephrol 2003,14:1254e1271

66. Mondino A, Blasi F: uPA and uPAR in fibrinolysis, immunity andpathology. Trends Immunol 2004, 25:450e455

67. Sisson TH, Hanson KE, Subbotina N, Patwardhan A, Hattori N,Simon RH: Inducible lung-specific urokinase expression reducesfibrosis and mortality after lung injury in mice. Am J Physiol LungCell Mol Physiol 2002, 283:L1023eL1032

68. Shetty S, Pendurthi UR, Halady PK, Azghani AO, Idell S: Urokinaseinduces its own expression in Beas2B lung epithelial cells. Am JPhysiol Lung Cell Mol Physiol 2002, 283:L319eL328

69. Shetty S, Gyetko MR, Mazar AP: Induction of p53 by urokinase inlung epithelial cells. J Biol Chem 2005, 280:28133e28141

70. Ulsamer A, Wei Y, Kim KK, Tan K, Wheeler S, Xi Y, Thies RS,Chapman HA: Axin pathway activity regulates in vivo pY654-ß-catenin accumulation and pulmonary fibrosis. J Biol Chem 2012, 287:5164e5172

71. Samarakoon R, Chitnis SS, Higgins SP, Higgins CE, Krepinsky JC,Higgins PJ: Redox-induced Src kinase and caveolin-1 signaling inTGF-b1-initiated SMAD2/3 activation and PAI-1 expression. PLoSOne 2011, 6:e22896

72. Hu M, Che P, Han X, Cai GQ, Liu G, Antony V, Luckhardt T,Siegal GP, Zhou Y, Liu RM, Desai LP, O’Reilly PJ, Thannickal VJ,Ding Q: Therapeutic targeting of Src kinase in myofibroblast differ-entiation and pulmonary fibrosis. J Pharmacol Exp Ther 2014, 351:87e95 Q

ajp.amjpathol.org - The American Journal of Pathology

1736

15 November 2014 � 11:35 am � EO: AJP14_0185

Page 15: Role of the Urokinase-Fibrinolytic System in Epithelial–Mesenchymal Transition during Lung Injury

Q15 Q16

173717381739174017411742174317441745174617471748174917501751175217531754175517561757175817591760

176117621763176417651766176717681769177017711772177317741775177617771778177917801781178217831784

Supplemental Figure S1 Inhibition of bleomycin (BLM)-induced epithelialemesenchymal transitions in ATII cells by caveolin-1 scaffolding domainpeptide (CSP). Wild-type mice were exposed to saline or BLM (2 U/kg body weight) through intranasal instillation. After 24 hours, mice exposed BLM were i.p.injected with or without 18.75 mg/kg body weight of CSP or control peptide (CP). Three days after BLM treatment, the lungs were inflated and the lungsections subjected to immunohistochemical analysis using antiecollagen-I and antieZO-1 antibodies.

Supplemental Figure S2 Inhibition of passive cigarette smoke (PCS)-induced epithelialemesenchymal transitions in ATII cells by caveolin-1 scaffoldingdomain peptide (CSP). Wild-type mice were exposed to ambient air or PCS from 40 cigarettes (approximately 90 mg/m3 total solid particulates) using amechanical smoking chamber over a 2-hour period for 5 days per week. After 4 weeks of PCS exposure, mice exposed to PCS were i.p. injected with or without18.75 mg/kg body weight of CSP or control peptide (CP) once every week for 4 more weeks. After 20 weeks of PCS exposure, the lungs were inflated and thelung sections subjected to immunohistochemical analysis using antiecollagen-I and antieZO-1 antibodies.

Supplemental Figure S3 Isolation of ATII cells lacking urokinase-type plasminogen activator (uPA) or uPA plasma membrane receptor (uPAR)expression. ATII cells isolated from uPA-deficient (A) or uPAR-deficient (B) mice were stained with lithium carbonate for inclusion bodies to assess the purityof cell preparation.

Supplemental Figure S4 Role of urokinase-type plasminogen activator (uPA) in ATII cell epithelialemesenchymal transitions after bleomycin (BLM)injury. Mice deficient in uPA expression were exposed to saline or BLM (2 U/kg body weight) through intranasal instillation. Twenty-four hours after initialexposure to BLM, mice were i.p. injected with or without caveolin-1 scaffolding domain peptide (CSP) or control peptide (CP). Seventy-two hours afterinitiation of BLM injury, the lung sections were subjected to immunohistochemical analysis using antieZO-1, collagen-I, and a-SMA antibodies.

Supplemental Figure S5 Distribution of biotin-labeled caveolin-1 scaffolding domain peptide (CSP) in the lungs of mice. Mice exposed to saline orbleomycin (BLM) for 3 days were i.p. injected with vehicle or biotin-labeled CSP; 24 hours after biotin-CSP, lung sections were analyzed for biotin-CSP usinganti-biotin antibody. Lung sections from the vehicle-treated mice were used as controls. Representative fields from one of the six sections are shown (n Z2 per group). Arrows indicate alveolar epithelial reactivity. Original magnification: �200.

FLA 5.2.0 DTD � AJPA1868_proof � 15 November 2014 � 11:35 am � EO: AJP14_0185