11
The importance of matrix metalloproteinase-3 in respiratory disorders Expert Rev. Respir. Med. 8(4), 411–421 (2014) Cory M Yamashita 1 , Derek C Radisky 2 , Yael Aschner 3,4 and Gregory P Downey* 3–5 1 Department of Medicine, University of Western Ontario, London, ON, Canada 2 Mayo Clinic Cancer Center, Jacksonville, FL, USA 3 Departments of Medicine and Pediatrics, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO, USA 4 Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver, Aurora, CO, USA 5 Department of Immunology, University of Colorado Denver, Aurora, CO, USA *Author for correspondence: Tel.: +1 303 398 1436 Fax: +1 303 270 2243 [email protected] The matrix metalloproteinases (MMPs) encompass a family of zinc-dependent endopeptidases that are secreted into the extracellular environment or remain bound to the cell surface. While MMPs were initially identified based on their ability to degrade collagen and other components of the extracellular matrix, recent studies indicate that their non-degradative functions are physiologically paramount. In particular, MMPs are now known to participate in diverse physiological processes that control key aspects of inflammatory and immune responses and neoplasia, in part by selective triggering of cellular signaling pathways via limited proteolytic processing of extracellular and membrane-associated proteins, including cytokines and cell surface receptors. Herein, we focus on the unique roles of MMP-3 (stromelysin-1) in acute lung injury and repair, pulmonary fibrosis, and lung cancer. KEYWORDS: acute lung injury • acute respiratory distress syndrome • epithelial mesenchymal transformation • extracellular matrix • lung cancer • proteinase • proteolytic processing • pulmonary fibrosis • signal transduction The matrix metalloproteinases (MMPs) include a family of more than 20 structurally related zinc-dependent endopeptidases that were ini- tially recognized for their ability to degrade col- lagen and other components of the extracellular matrix (ECM), processes that play a critical physiological role in tissue remodeling in wound repair. More recent evidence, however, indicates that their nondegradative functions may be physiologically dominant [1,2]. MMPs have been subdivided based on their substrate specificity and structural properties into sub- groups that include gelatinases (MMP-2 and 9); stromelysins (MMP–3, 10 and 11); collagenases (MMP-1, 8 and 13); matrilysins (MMP-7 and 26) and membrane-type MMPs (MMP-14, 15, 16, 17, 24 and 25) [3]. It should be noted, how- ever, that classifying MMPs according to sub- strate specificity is largely based on in vitro observations and may not reflect in vivo func- tion. As mentioned above, in addition to their ability to remodel the ECM, MMPs are able to posttranslationally modify soluble and mem- brane-associated proteins by a process termed limited proteolytic processing, which can result in activation or inactivation of cytokines, growth factors and cell surface receptors [4]. By virtue of their multiple biological activities, MMPs are believed to participate in a variety of physiological processes such as repair of tissues after injury, inflammation and immune responses [5] and are key contributors to a diverse range of pathological processes including rheumatoid arthritis and fibrosis of the liver, kidneys, heart and lungs [610]. Notably, MMP-3 (stromelysin- 1) has been directly implicated in epithelial- mesenchymal transition (EMT), a phenotypic alteration in which epithelial cells detach from neighboring cells and the underlying basement membrane and become more motile and inva- sive [11,12]. Importantly, EMT may contribute to the pathogenesis of neoplasia [13] and pulmonary fibrosis [14], although the latter remains controver- sial [1517]. There is mounting evidence that MMPs have pleiotropic effects in the context of tissue remodeling and repair that extend beyond their roles in matrix remodeling. In this regard, an emerging literature suggests that MMPs, including MMP-3, can regulate tissue repair by altering the activity of other nonma- trix proteins including cytokines and mem- brane receptors [18,19], thus facilitating cellular signaling through limited proteolytic process- ing and triggering activation of pathways lead- ing to EMT [13]. This phenomenon, whereby informahealthcare.com 10.1586/17476348.2014.909288 Ó 2014 Informa UK Ltd ISSN 1747-6348 411 Review Expert Review of Respiratory Medicine Downloaded from informahealthcare.com by Rona Williamson on 07/16/14 For personal use only.

The importance of matrix metalloproteinase-3 in respiratory disorders

Embed Size (px)

Citation preview

The importance of matrixmetalloproteinase-3 inrespiratory disordersExpert Rev. Respir. Med. 8(4), 411–421 (2014)

Cory M Yamashita1,Derek C Radisky2,Yael Aschner3,4 andGregory PDowney*3–5

1Department of Medicine, University of

Western Ontario, London, ON, Canada2Mayo Clinic Cancer Center,

Jacksonville, FL, USA3Departments of Medicine and

Pediatrics, Division of Pulmonary,

Critical Care, and Sleep Medicine,

National Jewish Health, Denver, CO,

USA4Department of Medicine, Division of

Pulmonary Sciences and Critical Care

Medicine, University of Colorado

Denver, Aurora, CO, USA5Department of Immunology, University

of Colorado Denver, Aurora, CO, USA

*Author for correspondence:

Tel.: +1 303 398 1436

Fax: +1 303 270 2243

[email protected]

The matrix metalloproteinases (MMPs) encompass a family of zinc-dependent endopeptidasesthat are secreted into the extracellular environment or remain bound to the cell surface. WhileMMPs were initially identified based on their ability to degrade collagen and other componentsof the extracellular matrix, recent studies indicate that their non-degradative functions arephysiologically paramount. In particular, MMPs are now known to participate in diversephysiological processes that control key aspects of inflammatory and immune responses andneoplasia, in part by selective triggering of cellular signaling pathways via limited proteolyticprocessing of extracellular and membrane-associated proteins, including cytokines and cellsurface receptors. Herein, we focus on the unique roles of MMP-3 (stromelysin-1) in acute lunginjury and repair, pulmonary fibrosis, and lung cancer.

KEYWORDS: acute lung injury • acute respiratory distress syndrome • epithelial mesenchymal transformation

• extracellular matrix • lung cancer • proteinase • proteolytic processing • pulmonary fibrosis • signal transduction

The matrix metalloproteinases (MMPs) includea family of more than 20 structurally relatedzinc-dependent endopeptidases that were ini-tially recognized for their ability to degrade col-lagen and other components of the extracellularmatrix (ECM), processes that play a criticalphysiological role in tissue remodeling inwound repair. More recent evidence, however,indicates that their nondegradative functionsmay be physiologically dominant [1,2]. MMPshave been subdivided based on their substratespecificity and structural properties into sub-groups that include gelatinases (MMP-2 and 9);stromelysins (MMP–3, 10 and 11); collagenases(MMP-1, 8 and 13); matrilysins (MMP-7 and26) and membrane-type MMPs (MMP-14, 15,16, 17, 24 and 25) [3]. It should be noted, how-ever, that classifying MMPs according to sub-strate specificity is largely based on in vitroobservations and may not reflect in vivo func-tion. As mentioned above, in addition to theirability to remodel the ECM, MMPs are able toposttranslationally modify soluble and mem-brane-associated proteins by a process termedlimited proteolytic processing, which can resultin activation or inactivation of cytokines,growth factors and cell surface receptors [4]. Byvirtue of their multiple biological activities,

MMPs are believed to participate in a variety ofphysiological processes such as repair of tissuesafter injury, inflammation and immune responses[5] and are key contributors to a diverse range ofpathological processes including rheumatoidarthritis and fibrosis of the liver, kidneys, heartand lungs [6–10]. Notably, MMP-3 (stromelysin-1) has been directly implicated in epithelial-mesenchymal transition (EMT), a phenotypicalteration in which epithelial cells detach fromneighboring cells and the underlying basementmembrane and become more motile and inva-sive [11,12]. Importantly, EMT may contribute tothe pathogenesis of neoplasia [13] and pulmonaryfibrosis [14], although the latter remains controver-sial [15–17].

There is mounting evidence that MMPshave pleiotropic effects in the context of tissueremodeling and repair that extend beyondtheir roles in matrix remodeling. In thisregard, an emerging literature suggests thatMMPs, including MMP-3, can regulate tissuerepair by altering the activity of other nonma-trix proteins including cytokines and mem-brane receptors [18,19], thus facilitating cellularsignaling through limited proteolytic process-ing and triggering activation of pathways lead-ing to EMT [13]. This phenomenon, whereby

informahealthcare.com 10.1586/17476348.2014.909288 � 2014 Informa UK Ltd ISSN 1747-6348 411

Review

Exp

ert R

evie

w o

f R

espi

rato

ry M

edic

ine

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Ron

a W

illia

mso

n on

07/

16/1

4Fo

r pe

rson

al u

se o

nly.

a class of tissue degrading enzymes can paradoxically promotethe deposition of excess tissue matrix (e.g., collagen), may beattributable in part to these nondegradative signaling functions.The current manuscript will review the involvement of MMPs,specifically MMP-3, in three pathological processes involvingthe lungs: acute lung injury (ALI) and repair, pulmonary fibro-sis and lung cancer. Although the mechanisms supporting therole of MMP-3 in the pathobiology of lung injury, repair andfibrosis may be more apparent, several lines of evidence pointtoward shared mechanisms underlying the development oftumor growth and cancer biology that will be highlighted inthe last section of this review.

Role of MMP-3 in acute lung injury & repairALI and the acute respiratory distress syndrome (ARDS) repre-sent a spectrum of diseases characterized by the development ofnoncardiogenic pulmonary edema, accompanied by an intenseneutrophilic pulmonary inflammatory response resulting from abroad spectrum of both direct (i.e., gastric aspiration, severepneumonia) and indirect lung insults (e.g., sepsis, multitrauma,transfusion of blood products). ARDS represents a large burdenof disease in the intensive care unit (ICU) population, withover 200,000 cases annually and a high mortality of approxi-mately 40% [20]. Several multicenter randomized clinical trialshave demonstrated improved outcomes in ARDS by the use ofpositive end-expiratory pressure (PEEP) [21], low tidal volumeventilation [22] and conservative fluid management strategies [23].Notably, while the use of high levels of PEEP has not beenshown to improve overall survival in ARDS when compared tolow levels of PEEP, the use of high levels of PEEP was associ-ated with decreased mortality in the ICU in the subgroup ofpatients with most severe lung injury (‘ARDS’) and improvedoxygenation status [24,25].

Recently, the definition of ARDS has been revised to reflectdifferences in blood oxygenation that allows greater predictivevalue in terms of morbidity, while the term ‘ALI’ has beenremoved from clinical diagnostic criteria [26]. Nonetheless, theterm ALI continues to be used in animal studies that areintended to model human ARDS, and for the purpose of thisreview, ALI/ARDS will be utilized interchangeably to reflecttheir common pathophysiology. The pathogenesis of ARDS/ALI is complex and involves neutrophil-dependent lung endo-thelial and epithelial cell injury, resulting in accumulation ofprotein-rich pulmonary edema as well as impaired removal ofedema fluid and resolution of inflammation [20,27].

The pleiotropic effects of MMPs have made them an attrac-tive area of focus in the context of ARDS. In the clinical realm,elevated levels of several MMPs have been identified in bron-choalveolar lavage fluid from patients with ARDS [28–30] and animbalance between MMPs and their endogenous inhibitors, thetissue inhibitors of MMPs (TIMPs), has been hypothesized toplay a role in disease pathogenesis [31]. Elevated levels ofMMP-1 and/or MMP-3 were found to be predictive of mortal-ity in patients with an established diagnosis of ARDS [28]. Stud-ies utilizing animal models have also provided evidence of the

importance of MMP-3 in the pathogenesis of ALI. For example,mice genetically deficient in MMP-3 or MMP-9 were protectedfrom lung injury triggered by intratracheal administration ofIgG immune complexes or macrophage inflammatory protein-2,a potent neutrophil chemotactic factor [32,33]. The MMP-3-defi-cient animals exhibited reduced neutrophil recruitment into thelungs, a finding that was not recapitulated in MMP-9 knockoutmice, suggesting that MMP-3 is specifically required for neutro-phil recruitment into the lungs and highlighting the concept thatthe mechanisms of involvement in ALI differ between individualMMPs. Coupled with the evidence that MMP-3-deficient ani-mals are protected from bleomycin-induced pulmonary fibro-sis [34], these observations support the hypothesis thatMMP-3 plays an important role in both lung injury and repair.Further studies in this area, in particular, those employing geneti-cally altered species of animals such as MMP-3-deficient mice areneeded to delineate how compensatory changes in other MMPsand/or endogenous inhibitors such as TIMPs may be impacted.Although the TIMPs have also been implicated in lung injury [35]

and repair [36], specific interactions between MMP-3 and endoge-nous TIMPs in the setting of ALI represents an area ofongoing investigation.

Although the mechanisms by which MMP-3 contributes tothe pathogenesis of ALI are incompletely understood, a numberof possibilities can be hypothesized from studies investigatingits role in disease models of other organs (FIGURE 1). MMP-3 isable to induce neutrophil chemoattractant activity in interleu-kin-1b-stimulated intestinal epithelial cells (Caco-3 cells) by amechanism involving proteolytic cleavage of chemotacticallyinert platelet basic protein yielding the active neutrophilchemokine (C-X-C motif) ligand-7 (neutrophil activating pep-tide 2) [37]. Additional studies have shown that chondrocyte-derived MMP-3 was capable of generating a macrophage che-motactic factor implicated in disc degeneration in a model ofdisc herniation resorption [38]. The specific mechanisms bywhich MMP-3 itself could promote neutrophil migration, ashas been observed in MMP-3-deficient models of ALI, areunknown and further studies characterizing the role ofMMP-3 in generating neutrophil-specific chemoattractants arerequired [39].

In addition to the intense neutrophilic and proinflammatoryresponses implicated in the pathogenesis of ALI/ARDS, breachof the alveolar capillary barrier facilitates the movement ofprotein-rich plasma into the alveoli and represents a hallmarkfeature of ARDS (FIGURE 1). Prior studies in mouse mammaryepithelial cells revealed that treatment with MMP-3 resulted inloss of intact E-cadherin, a key transmembrane protein in theformation of interepithelial cell (adherens) junctions [13]. Whilethis mechanism has been implicated in malignant transforma-tion of mammary epithelial cells in breast cancer, it remainsuncertain if MMP-3-mediated cleavage of E-cadherin contrib-utes to lung epithelial cell injury and thus to the pathogenesisof ALI.

A recent study further highlights the complexity of the localeffects of MMP-3 within the airspaces of the distal lung. In

Review Yamashita, Radisky, Aschner & Downey

412 Expert Rev. Respir. Med. 8(4), (2014)

Exp

ert R

evie

w o

f R

espi

rato

ry M

edic

ine

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Ron

a W

illia

mso

n on

07/

16/1

4Fo

r pe

rson

al u

se o

nly.

this study, shedding of the receptor foradvanced glycation end products (RAGE)from cultured rat alveolar epithelial cellsvia MMP-3- and MMP-13-mediated pro-teolysis yielded bioactive soluble products(sRAGE) [40]. Furthermore, a significantcorrelation was found between levels ofMMP-3 and MMP-13 and sRAGE inhuman ALI/ARDS bronchoalveolar lavagesamples [40]. Although elevated levels ofboth alveolar and circulating sRAGE cor-relate with adverse outcomes in the settingof ALI/ARDS, the possibility has beenraised that both MMP-3 and MMP-13could limit proinflammatory gene expres-sion in the lungs through decoy receptoractivities of sRAGE, thus providing a neg-ative feedback mechanism [41]. Furtherstudies will be required to delineate theprecise proteolytic processing activities ofMMP-3 with alveolar epithelial basedRAGE and how this interaction couldimpact pro- or anti-inflammatory responsesin the lungs.

Recently, the contribution of injuriousmechanical ventilation to lung injury inARDS has received considerable attention[42–44]. Evidence suggests that excessivemechanical stress can induce both macro-and microscopic injury to the lungs,resulting in proinflammatory responsesthat contribute to adverse patient out-comes. With this in mind, the involve-ment of MMPs in the pathogenesis oflung injury resulting from the effects ofmechanical stress has been an area ofrecent interest, with several recent studiesproviding an interesting perspective onthe role of MMPs in ventilator-inducedlung injury. These studies highlight a potentially complex inter-action between stress- and mechanical stretch-mediated releaseof MMPs that may individually promote acute inflammatoryresponses [45] or, conversely, protect against neutrophilic inflam-mation via modulation of airspace cytokine responses [46].Although these studies suggest a mechanism by whichMMP-3 could modulate the development of ALI in responseto ventilator-induced lung injury, specific studies addressingthis aspect of ALI are lacking and this remains an area requir-ing additional investigation.

In summary, multiple observations suggest the role ofMMP-3 in the pathogenesis of ALI via mechanisms such asneutrophil recruitment, epithelial cell injury and promotion ofacute inflammatory responses. MMP-3 may also play a role inresolution of ALI. However, it remains unclear whether theseenzymes represent nonspecific markers of tissue injury or

biomarkers of disease severity, or whether they play a moredirect role in disease initiation and/or progression and thusmight represent a relevant therapeutic target. Further investiga-tion and validation, including in models of direct and indirectALI, will be critical in determining the exact mechanisms bywhich MMP-3 promotes lung injury.

Role of MMP-3 in pulmonary fibrosis

Fibrotic lung diseases represent a diverse group of progressiveand ultimately fatal disorders collectively termed idiopathicinterstitial pneumonias, of which idiopathic pulmonary fibrosis(IPF) is the most common [47–50]. Clinically, IPF is character-ized by progressive dyspnea and a restrictive physiologicaldefect that is attributable to excessive and disorganized deposi-tion of ECM including collagen and fibronectin within thelung parenchyma [51]. Despite an improved understanding of

Edemafluid

Alveolus

Neutrophil chemotaxis

Tight andadherensjunctions

VE-Cadherin

E-Cadherin

ATI cell

Basementmembrane

MMP3

ATII cell

sRAGE

↑ Endothelialpermeability

↑ Endothelialpermeability Platelets CXCL7

Neutrophil

Pulmonarycapillary

Pulmonaryendothelium

Figure 1. The role of matrix metalloproteinase-3 in acute lung injury/acuterespiratory distress syndrome. Elevated levels of MMP-3 observed in the setting ofacute lung injury/acute respiratory distress syndrome have been demonstrated to provide achemotactic gradient for neutrophil recruitment to the lung interstitium and alveolar space,disrupt junctional proteins along vascular endothelial cells and degrade basementmembrane and cleave adherens junctions (i.e., E-cadherin) between alveolar epithelial celljunctions.ATI cell: Alveolar type I epithelial cell; ATII: Alveolar type II epithelial cell;CXCL-7: Chemokine (C-X-C motif) ligand 7; MMP-3: Matrix metalloproteinase 3;sRAGE: Soluble receptor for advanced glycation end products.

The importance of matrix metalloproteinase-3 in respiratory disorders Review

informahealthcare.com 413

Exp

ert R

evie

w o

f R

espi

rato

ry M

edic

ine

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Ron

a W

illia

mso

n on

07/

16/1

4Fo

r pe

rson

al u

se o

nly.

specific biological mechanisms that propagate this uncontrolledfibrotic process, limited progress has been made in the treat-ment of this devastating condition [47,50,52]. Both genetic andenvironmental factors appear to contribute to the pathogenesisof IPF [51,53–58]. The incidence of IPF increases with age [59]

possibly attributable to cumulative age-related genetic modifica-tions, which impair the ability of the lung to repair itself afterrepeated injury. This leads to release of profibrotic cytokinesand growth factors such as TGF-b and CTGF that inducefibroblast and myofibroblast accumulation, excessive depositionof ECM and progressive widespread lung fibrosis [50,51,60].

While it is evident that MMPs could participate in the deg-radation of the ECM during the injury phase, it is less intuitivethat a class of tissue degrading enzymes could also participate inexcessive deposition of ECM. Consistent with this concept,recent evidence from our group and others indicates thatMMPs can modify the composition of the ECM both directly

and via modulation of the activity of sol-uble or matrix-bound proteins such ascytokines and growth factors of mem-brane receptors, and through the genera-tion of signaling mediators derivedthrough these processes (FIGURE 2). Previousstudies from our laboratory demonstratedthat human fibroblasts selectively secreteMMP-3 (compared to other MMPs)when stimulated with the profibroticmediator IL-1b under conditions wherefocal adhesions are formed [61]. Ourgroup has also provided strong evidencefor the pathophysiological importance ofMMP-3 in the development of pulmo-nary fibrosis [34]. In this regard, wereported (FIGURE 3) that explanted lung tis-sues from patients with IPF expressedgreater levels of MMP-3 compared tocontrols [34]. Further in vivo studies dem-onstrated that the intratracheal administra-tion of recombinant MMP-3 viaadenoviral-mediated gene delivery eliciteda robust profibrotic response in rats, andconversely mice genetically deficient inMMP-3 were resistant to the developmentof bleomycin-induced pulmonary fibrosis(FIGURE 3) [34].

Excessive fibroblast accumulation(FIGURE 2), a hallmark feature of pulmonaryfibrosis, has been proposed to occurthrough a variety of mechanisms includ-ing proliferation of local pulmonaryfibroblasts; recruitment of circulatingfibrocytes and conversion of epithelialcells to a fibroblast phenotype, termedEMT. With respect to the latter process,Konigshoff and colleagues demonstrated

that activation of the canonical Wnt-b-catenin signaling path-way is a key regulator of alveolar type II cell hyperplasia andplasticity leading to EMT [62,63]. Additional studies from ourlaboratory revealed that exogenous MMP-3-induced activationof the Wnt-1 inducible signaling pathway protein, a targetgene of the Wnt-b-catenin pathway, in primary lung epithelialcells [34]. Furthermore, we demonstrated that bleomycin-exposed MMP-3-deficient mice exhibited reduced expression ofcyclin D1 (a target gene of the Wnt-b-catenin pathway)compared to wild-type control animals. Taken together, thesefindings support the role of MMP-3 in the development ofpulmonary fibrosis via mechanisms involving mesenchymaltransition of lung epithelial cells. It is noteworthy, however,that the importance of EMT as a source for the excess fibro-blasts in murine models of pulmonary fibrosis is controver-sial [16] and additional studies are needed to reconcile theseapparently discrepant observations.

Pulmonarycapillary

Myofibroblast

MMP3

↑ Collagen and fibronectin

ECMdegradation

Fibroblast

ECM

Alveolus

ATI cell

ATII cell

E-cadherin degradation

? EMT

Macrophage

Macrophage

Neutrophil

Pro-fibroticgrowth factors

NeutrophilFibrocyte

Monocyte

Figure 2. The role of matrix metalloproteinase-3 in pulmonary fibrosis. Elevatedlevels of MMP-3 observed in the setting of progressive diffuse pulmonary fibrosis such asidiopathic pulmonary fibrosis have been demonstrated to induce mesenchymal transitionof lung epithelial cells, induce myofibroblast differentiation of fibroblasts, degradecomponents of the basement membrane and thus induce profibrotic responsiveness offibroblasts and cleave adherens junctions (i.e., E-cadherin) between alveolar epithelialcell junctions.ATI cell: Alveolar type I epithelial cell; ATII: Alveolar type II epithelial cell; ECM: Extracellularmatrix; EMT: Epithelial-mesenchymal transition; MMP-3: Matrix metalloproteinase 3.

Review Yamashita, Radisky, Aschner & Downey

414 Expert Rev. Respir. Med. 8(4), (2014)

Exp

ert R

evie

w o

f R

espi

rato

ry M

edic

ine

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Ron

a W

illia

mso

n on

07/

16/1

4Fo

r pe

rson

al u

se o

nly.

More recently, Rajshankar and colleagues have provided furtherinsight into the potential mechanisms by which MMP-3 mayenhance a profibrotic milieu in the context of pulmonary fibro-sis [64]. In this study, proteolysis of the fibronectin matrix byMMP-3 yielded degradation products that promoted IL-1b-mediated cell-dependent anchorage and adhesion. Althoughexperiments conducted in this study were performed primarilyusing human gingival fibroblasts, such findings support thenotion that in addition to MMP-3-mediated triggering of

b-catenin-dependent signaling pathways, MMP-3-mediated alter-ations in the ECM can lead to profibrotic changes in the pheno-typic behavior of fibroblasts by modifying cell–matrix interactionspresumably by signaling through integrins and associated signal-ing molecules in the context of focal adhesions.

In summary, strong evidence from in vitro animal and humanstudies supports the importance of MMP-3 in the pathogenesisof pulmonary fibrosis. Nonetheless, many questions remain to beanswered such as whether enhanced expression of MMP-3 is

Ave

rag

e ex

pre

ssio

n IP

F

Average expression controlsR

elat

ive

MM

P3

exp

ress

ion

0

MMP9MMP7

MMP1

MMP11

MMP10

MMP13

MMP1

1

2

3

4

5

Control

0WT MMP-3

Null

NS BLM

WT MMP-3Null

Co

llag

en m

g/m

l

20

40

60

IPF

Con

trol

IPF

Alveoli Airway

4X PS stain 40X PS stain 40X H and E stain

WT

mou

seM

MP

3 nu

ll m

ouse

100

μm

100

μm10

0 μm

100

μm

A B

D E

C

Figure 3. Matrix metalloproteinase-3 is a critical mediator of idiopathic pulmonary fibrosis. (A) Gene expression array analysisof human IPF/UIP lung tissue samples demonstrates a significant increase in MMP-3 mRNA expression (q = 0) compared with non-IPF con-trol samples. Relative expression of other MMPs (i.e., MMP-1, -7, -9, -10, -11 and -13) are shown for comparison. (B) Quantitative RT-PCR analysis of explanted surgical lung specimens demonstrates a greater than fourfold increase in MMP-3 mRNA levels in IPF versusnon-IPF lung specimens (*p < 0.05 IPF vs control). (C) Immunohistochemical analysis of human control (top) and IPF (bottom) lung sec-tions to demonstrate cellular expression of MMP-3 (original magnification, 40). Representative sections from control lung demonstratethat MMP-3 staining (brown) is largely confined to alveolar macrophages (solid arrow). By contrast, in sections from IPF lung, there isstaining of alveolar macrophages (solid arrowheads, bottom left), alveolar epithelial cells (open arrowheads, bottom left and right), airwayepithelial cells (solid arrowheads, bottom right) and intravascular leukocytes (solid arrow, bottom right). Scale bars: 100 mm. (D) Lungsections (top) from wild-type (WT) and MMP-3-null mice treated with bleomycin (original magnification, 4 and 40 as indicated), stainedwith Picrosirius Red (PS) or with H&E demonstrating an increase in collagen (red staining) deposition in WT compared with MMP-3-nullmice. (E) Sircol assay of lung collagen content illustrating that bleomycin-treated MMP-3-null mice have significantly less collagen thanbleomycin-treated WT controls (*p < 0.05 WT BLM vs MMP-3-null BLM).BLM: Bleomycin; IPF: Idiopathic pulmonary fibrosis; MMP-3: Matrix metalloproteinase 3; NS: Normal saline.Data reprinted with permission from [34].

The importance of matrix metalloproteinase-3 in respiratory disorders Review

informahealthcare.com 415

Exp

ert R

evie

w o

f R

espi

rato

ry M

edic

ine

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Ron

a W

illia

mso

n on

07/

16/1

4Fo

r pe

rson

al u

se o

nly.

sufficient to induce clinical disease in humans in addition toidentifying the specific cellular sources of MMP-3 involved inthese fibrogenic processes. Until the discrete origins of diseasepathogenesis become more clearly defined and the precise mecha-nisms that result in the release of MMP-3 become evident, itremains uncertain whether therapeutic targeting of MMP-3 is aviable option to prevent pathological fibrosis.

Role of MMP-3-induced EMT in lung cancer progression

Lung cancer represents a major healthcare burden for bothdeveloped and developing nations and it is one that isincreasing in prevalence [65]. Lung cancers are the secondleading cause of morbidity and mortality globally, and amongthese cancers non-small-cell lung cancer represents the vastmajority (>80%) [65–67]. The principal environmental risk fac-tor for lung cancer is cigarette smoke exposure, althoughother environmental and genetic factors also pose significantrisk [65]. Increased expression of MMP-3 has been linked totumor growth and metastasis in human breast, colon, cervicaland lung cancers [68–73], but the mechanism by whichMMP-3 acts in human cancer development remains uncer-tain (FIGURE 4). In cell culture models, MMPs have been identi-fied as key inducers of EMT: MMP-3, MMP-7 andMMP-28 have all been shown to induce EMT in A549 lungadenocarcinoma cells [74–76]. Furthermore, MMP-3 has beenshown to induce EMT in human and mouse mammary epi-thelial cells [13,77,78] and in normal murine lung epithelialcells [34]. The role of MMP-3-induced EMT in cigarette

smoke-associated lung cancer development has recently beenidentified [79].

Insight into how MMPs could stimulate EMT was providedby transgenic mouse models with inducible expression ofMMP-3 in lung alveolar cells. Induction of MMP-3 in thesemice led to increased expression of Rac1b, an alternativelyspliced isoform of Rac1, that has previously been shown to reg-ulate actin cytoskeleton organization, membrane trafficking,proliferation and gene expression [80–83]. Rac1b results frominclusion of the 57 nucleotide exon 3b, which leads to a19-amino acid insertion in the protein sequence [84,85]. Theresulting Rac1b isoform accelerates GDP/GTP exchange andimpaired GTP hydrolysis [86–88]. Rac1b has been identified inmalignant colorectal [84], papillary thyroid [89] and breast can-cers [85,90]. Additional studies using cultured cells demonstratedthat Rac1b could stimulate oncogenic activities through theinduction of reactive oxygen species production and consequentpromotion of EMT [13,91,92]. Further studies implicated theinduction of Rac1b by MMP-3 as essential for MMP-3-in-duced EMT and acquisition of malignant characteristics in cul-tured lung epithelial cells [79].

These results identified MMP-3-induced Rac1b as a poten-tial pathway for lung cancer development. In support of thispossibility, investigation of human biopsy tissue samplesrevealed that Rac1b was upregulated both in stages 1 and 2 oflung tumor progression and in response to cigarette smokeexposure (FIGURE 5). Additionally, analyses of transcriptional pro-files induced in lung epithelial cells exposed to MMP-3 showedsignificant similarities to lung tissue from smokers [79], impli-cating a possible MMP-Rac1b signaling axis in cigarettesmoke-associated lung cancer development. These observationsindicate a novel point of therapeutic intervention: while inhibi-tors of MMPs have not been successful anticancer agents [93],due in part to the requirement of these proteins in normalphysiological functions, Rac1b has been identified uniquely intumors and tumor-derived cell lines and has no known physio-logical functions. Thus, targeted inhibition of Rac1b functionis a potential therapeutic approach to attenuate MMP-3-in-duced EMT. Such a strategy would have multiple potentialbenefits as the cellular changes associated with EMT-like transi-tions alter EGFR-dependent signaling pathways and have beenlinked to lung tumor cell, xenograft and patient insensitivity toselective EGFR tyrosine kinase inhibitors [94–96]. BlockingMMP-3/Rac1b-induced EMT could therefore potentially sensi-tize tumor cells and enhance responses to existing therapieswhile independently reducing EMT-associated tumor progres-sion and metastasis of lung cancer.

Concluding remarksMMPs are key participants in diverse physiological and path-ophysiological processes that control key aspects of inflamma-tory and immune responses and neoplasia. In addition todegradation and remodeling of ECM proteins, MMPs caninitiate cellular signaling pathways via limited proteolyticprocessing of extracellular and membrane-associated proteins

MMP-3

GF;ROSTumor

formation

Fibrosis-,angiogenesis-

inducing factors

A

B

CD

E

Figure 4. The role of matrix metalloproteinase-3 in tumorformation. (A) MMP-3 triggers alveolar epithelial cells toincrease expression of Rac1b and consequently (B) undergoepithelial-mesenchymal transition, producing an activated cellthat can (C) generate GF and ROS that further disrupt normallung tissue structure, (D) fibrosis- and angiogenesis-inducing fac-tors that support developing tumors and also (E) directly lead totumor formation.GF: Growth factor; ROS: Reactive oxygen species; MMP-3: Matrixmetalloproteinase 3.

Review Yamashita, Radisky, Aschner & Downey

416 Expert Rev. Respir. Med. 8(4), (2014)

Exp

ert R

evie

w o

f R

espi

rato

ry M

edic

ine

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Ron

a W

illia

mso

n on

07/

16/1

4Fo

r pe

rson

al u

se o

nly.

including cytokines and cell surface receptors. In the currentmanuscript, we have discussed the importance of MMPs, spe-cifically MMP-3, in ALI and repair, pulmonary fibrosis andlung cancer.

Expert commentaryMMPs, including MMP-3, while initially identified based ontheir ability to degrade collagen and other components of theECM, have recently been shown to play a role in more diverse

A

D G

I

E F

B C

MMP-3

LAC

Rac1b

*** ***

0

2

1

ntg MMP-3 ntg MMP-3

3

MMP-3 Rac1b

0

2

Rel

ativ

ein

ten

sity

Rel

ativ

ein

ten

sity

MM

P-3

ntg

*

***

***

N 1 20.0000

0.0005

0.0010

Rac

1b/1

8S

Normal

IHC: Rac1b

Never

Curre

nt0.000000

0.000025

0.000050

0.000075

0.000100

[MMP-3][MMP-3]

Rac

1b/1

8S

*

Vim

enti

n/

GA

PD

H

0.0

0.4

0.8

1.2

1.6

Rac

1b/

GA

PD

H

0

1

2

3

siRac1bMMP-3

* **

siRac1b+ --MMP-3- +

++-

+ --- +

++-

H

J

0

1

2

3

4

5

6

0 U/m

l

25 U

/ml

50 U

/ml

100

U/ml

0 U/m

l

25 U

/ml

50 U

/ml

100

U/ml

Rac

1b/G

AP

DH

0

0.5

1

1.5

2

2.5

Vim

enti

n/G

AP

DH

Figure 5. MMP-3 induces tumorigenic Rac1b in lung cancer. (A) Immunohistochemistry showing increased expression of Rac1b(brown) in lung adenocarcinoma compared to normal adjacent lung. Scale bar: 200 mm. (B) Normalized Rac1b expression comparingbiopsies of stage 1 and 2 lung adenocarcinoma with adjacent noninvolved tissue (N). (C) Normalized Rac1b expression comparingpatients identified as current smokers to never smokers in lung homogenates. (D) Immunohistochemistry showing increased expressionof MMP-3 and Rac1b staining (brown) in adenomas from transgenic MMP-3-expressing mice (MMP-3) and nontransgenic (ntg) controlmice. Scale bar: 100 mm. (E and F) Histograms showing increased stain intensity of MMP-3 (E) and Rac1b (F) in MMP-3 transgenic mice ascompared to ntg. (G and H) Rac1b (G) and vimentin (H) expression comparing H661 cell homogenates treated for 2 d with 0, 25, 50 or100 U/ml of recombinant human MMP-3 by qPCR. (I and J) Rac1b (I) or vimentin (J) expression comparing H661 cell homogenates thatwere untreated with cells transfected with siRNA reagents selectively targeting Rac1b (siRac1b) either in the presence or absence of recombi-nant human MMP-3 by qPCR. Whisker plots indicating data range (whiskers) and interquartile range (boxes) (B,C) or averages ± SEM(E–J) are shown; *p < 0.05; ***p < 0.001.IHC: Immunohistochemistry; LAC: Lung adenocarcinoma; MMP-3: Matrix metalloproteinase 3.Data adapted from [74].

The importance of matrix metalloproteinase-3 in respiratory disorders Review

informahealthcare.com 417

Exp

ert R

evie

w o

f R

espi

rato

ry M

edic

ine

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Ron

a W

illia

mso

n on

07/

16/1

4Fo

r pe

rson

al u

se o

nly.

physiological processes through triggering of multiple cellularsignaling pathways via limited proteolytic processing of extra-cellular and membrane-associated proteins, including cytokines,growth factors and cell surface receptors. These pleiotropiceffects make MMP-3 a fascinating potential mediator in pul-monary pathological processes, including ALI/ARDS, fibrosisand cancer, which together make up a significant burden ofrespiratory disease. The ability of MMP-3 to produce activemediators of signaling pathways that regulate functions asdiverse as neutrophil recruitment, epithelial-to-mesenchymaltransition and inflammation, among others, may underliethe pathogenesis of these various disease states. In addition,recent work has shown that MMPs have the ability to alterthe underlying matrix, which may further enhance theproinflammatory and profibrotic milieu lead to dysfunc-tional repair resulting in fibrosis or neoplasia. Further workwill be required to more clearly define the role of MMPs,MMP-3 in particular, in respiratory disease. However, theseenzymes represent attractive potential therapeutic targetsthat are much needed in the treatment of these commonand often fatal lung diseases.

Five-year viewThe next 5 years will bring further mechanistic investigationinto the role of MMPs in human respiratory disease as well asin the pathogenesis of diseases of other organ systems. We willlikely see additional animal model studies of ALI that further

clarify how MMP-3 participates in neutrophil chemotaxis andbreakdown of the alveolar epithelial barrier. Work in the fieldof pulmonary fibrosis will need to focus on the contributionsof MMPs to the phenomenon of EMT as well as to the altera-tions in the matrix that are induced by proteolytic cleavage bythese enzymes and the way that these changes influence down-stream signaling cascades. Studies of lung cancer biology mayfocus on the potential for therapeutic blocking of EMTinduced by MMP-3-mediated formation of Rac1b. It will beuseful to determine whether MMPs represent direct players indisease initiation and progression, or whether they are morenonspecific markers of tissue injury. While human trials ofinhibitors of MMPs in the treatment of respiratory disease arelikely beyond the scope of the next 5 years, it may be possibleto see animal studies of these compounds initiated withinthis timeframe.

Financial & competing interests disclosure

C Yamashita has received grant funding from the Canadian Institute of

Health. D Radisky, R Zemans and G Downey have received grant fund-

ing from the National Institute of Health. The authors have no other rele-

vant affiliations or financial involvement with any organization or entity

with a financial interest in or financial conflict with the subject matter or

materials discussed in the manuscript. This includes employment, consul-

tancies, honoraria, stock ownership or options, expert testimony, grants or

patents received or pending or royalties.

No writing assistance was utilized in the production of this manuscript.

Key issues

• The matrix metalloproteinases (MMPs) are a family of enzymes with the ability to degrade components of the extracellular matrix

(ECM). They play critical roles in tissue remodeling and wound repair.

• MMPs also participate in limited proteolytic processing, resulting in activation and inactivation of soluble and membrane-associated

proteins, such as cytokines, growth factors and cell surface receptors. These functions may allow MMPs to paradoxically promote depo-

sition of excess ECM.

• MMP-3 has been implicated in a range of pathological processes, including acute lung injury (ALI), pulmonary fibrosis and lung cancer.

• MMP-3 levels have been shown to be elevated in clinical specimens of patients with ALI, and in animal studies and mice

genetically deficient in MMP-3 are protected in ALI models. This may be due to impaired neutrophil recruitment in

MMP-3 knockout mice.

• MMP-3 may also promote breakdown of alveolar epithelial barriers and acute inflammatory responses, particularly in the setting of

ventilator-induced lung injury: both mechanisms could contribute to the development of ALI.

• Genetic deletion of MMP-3 in mice confers protection from bleomycin-induced fibrosis, while transient overexpression of MMP-3 results

in profibrotic responses in rat lungs. This likely occurs by induction of the Wnt-b-catenin pathway by MMP-3, resulting in epithelial-

mesenchymal transition of lung alveolar epithelial cells.

• MMP-3-mediated degradation of the ECM enhances a profibrotic environment, which may affect the phenotype of fibroblasts and

promote further deposition of ECM and fibrosis.

• MMP-3 may promote progression and metastasis of lung cancer by increasing expression of Rac1b, an isoform of Rac1, which

has been implicated in various cancers through induction of reactive oxygen species and promotion of epithelial-mesenchymal

transition.

• Targeting of MMP-3 may be a potential therapeutic strategy for the treatment of multiple respiratory disease processes, including ALI,

fibrosis and lung cancer.

Review Yamashita, Radisky, Aschner & Downey

418 Expert Rev. Respir. Med. 8(4), (2014)

Exp

ert R

evie

w o

f R

espi

rato

ry M

edic

ine

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Ron

a W

illia

mso

n on

07/

16/1

4Fo

r pe

rson

al u

se o

nly.

References

Papers of special note have been highlighted as:

• of interest

•• of considerable interest

1. Page-McCaw A, Ewald AJ, Werb Z. Matrix

metalloproteinases and the regulation of

tissue remodelling. Nat Rev Mol Cell Biol

2007;8(3):221-33

2. Hadler-Olsen E, Fadnes B, Sylte I, et al.

Regulation of matrix metalloproteinase

activity in health and disease. FEBS J 2011;

278(1):28-45

3. Chakraborti S, Mandal M, Das S, et al.

Regulation of matrix metalloproteinases:

an overview. Mol Cell Biochem 2003;

253(1-2):269-85

4. Sternlicht MD, Werb Z. How matrix

metalloproteinases regulate cell behavior.

Annu Rev Cell Dev Biol 2001;17:463-516

5. Khokha R, Murthy A, Weiss A.

Metalloproteinases and their natural

inhibitors in inflammation and immunity.

Nat Rev Immunol 2013;13(9):649-65

6. Green MJ, Gough AK, Devlin J, et al.

Serum MMP-3 and MMP-1 and

progression of joint damage in early

rheumatoid arthritis. Rheumatology

(Oxford) 2003;42(1):83-8

7. Heymans S, Lupu F, Terclavers S, et al.

Loss or inhibition of uPA or

MMP-9 attenuates LV remodeling and

dysfunction after acute pressure overload in

mice. Am J Pathol 2005;166(1):15-25

8. Rosas IO, Richards TJ, Konishi K, et al.

MMP1 and MMP7 as potential peripheral

blood biomarkers in idiopathic pulmonary

fibrosis. PLoS Med 2008;5(4):e93

9. Uchinami H, Seki E, Brenner DA,

D’Armiento J. Loss of MMP 13 attenuates

murine hepatic injury and fibrosis during

cholestasis. Hepatology 2006;44(2):420-9

10. Zuo F, Kaminski N, Eugui E, et al. Gene

expression analysis reveals matrilysin as a

key regulator of pulmonary fibrosis in mice

and humans. Proc Natl Acad Sci USA

2002;99(9):6292-7

11. Radisky DC. Epithelial-mesenchymal

transition. J Cell Sci 2005;118(Pt 19):

4325-6

12. Weinberg RA. Twisted epithelial-

mesenchymal transition blocks senescence.

Nat Cell Biol 2008;10(9):1021-3

13. Radisky DC, Levy DD, et al. Rac1b and

reactive oxygen species mediate

MMP-3-induced EMT and genomic

instability. Nature 2005;436(7047):123-7

14. Selman M, Pardo A, Kaminski N.

Idiopathic pulmonary fibrosis: aberrant

recapitulation of developmental programs?

PLoS Med 2008;5(3):e62

15. Bartis D, Mise N, Mahida RY, et al.

Epithelial-mesenchymal transition in lung

development and disease: does it exist and is

it important? Thorax 2013; doi:10.1136/

thoraxjnl-2013-204608

16. Rock JR, Barkauskas CE, Cronce MJ, et al.

Multiple stromal populations contribute to

pulmonary fibrosis without evidence for

epithelial to mesenchymal transition. Proc

Natl Acad Sci USA 2011;108(52):E1475-83

17. Yamada M, Kuwano K, Maeyama T, et al.

Dual-immunohistochemistry provides little

evidence for epithelial-mesenchymal

transition in pulmonary fibrosis. Histochem

Cell Biol 2008;129(4):453-62

18. Maeda S, Dean DD, Gomez R, et al. The

first stage of transforming growth factor

beta1 activation is release of the large latent

complex from the extracellular matrix of

growth plate chondrocytes by matrix vesicle

stromelysin-1 (MMP-3). Calcif Tissue Int

2002;70(1):54-65

19. Lochter A, Galosy S, Muschler J, et al.

Matrix metalloproteinase

stromelysin-1 triggers a cascade of molecular

alterations that leads to stable

epithelial-to-mesenchymal conversion and a

premalignant phenotype in mammary

epithelial cells. J Cell Biol 1997;139(7):

1861-72

20. Matthay MA, Zemans RL. The acute

respiratory distress syndrome: pathogenesis

and treatment. Annu Rev Pathol 2011;6:

147-63

21. Mercat A, Richard JC, Vielle B, et al.

Positive end-expiratory pressure setting in

adults with acute lung injury and acute

respiratory distress syndrome: a randomized

controlled trial. JAMA 2008;299(6):646-55

22. Ventilation with lower tidal volumes as

compared with traditional tidal volumes for

acute lung injury and the acute respiratory

distress syndrome. The Acute Respiratory

Distress Syndrome Network. N Engl J Med

2000;342(18):1301-8

23. Wiedemann HP, Wheeler AP, Bernard GR,

et al. Comparison of two fluid-management

strategies in acute lung injury. N Engl J

Med 2006;354(24):2564-75

24. Briel M, Meade M, Mercat A, et al. Higher

vs lower positive end-expiratory pressure in

patients with acute lung injury and acute

respiratory distress syndrome: systematic

review and meta-analysis. JAMA 2010;

303(9):865-73

25. Santa Cruz R, Rojas JI, Nervi R, et al.

High versus low positive end-expiratory

pressure (PEEP) levels for mechanically

ventilated adult patients with acute lung

injury and acute respiratory distress

syndrome. Cochrane Database Syst Rev

2013;6:CD009098

26. ARDS Definition Task Force. Ranieri VM,

Rubenfeld GD, Thompson BT, et al. Acute

respiratory distress syndrome: the Berlin

Definition. JAMA 2012;307(23):2526-33

27. Matthay MA, Ware LB, Zimmerman GA.

The acute respiratory distress syndrome.

J Clin Invest 2012;122(8):2731-40

28. Fligiel SE, Standiford T, Fligiel HM, et al.

Matrix metalloproteinases and matrix

metalloproteinase inhibitors in acute lung

injury. Hum Pathol 2006;37(4):422-30

29. O’Kane CM, McKeown SW, Perkins GD,

et al. Salbutamol up-regulates matrix

metalloproteinase-9 in the alveolar space in

the acute respiratory distress syndrome. Crit

Care Med 2009;37(7):2242-9

30. Ricou B, Nicod L, Lacraz S, et al. Matrix

metalloproteinases and TIMP in acute

respiratory distress syndrome. Am J Respir

Crit Care Med 1996;154(2 Pt 1):346-52

31. Lanchou J, Corbel M, Tanguy M, et al.

Imbalance between matrix

metalloproteinases (MMP-9 and MMP-2)

and tissue inhibitors of metalloproteinases

(TIMP-1 and TIMP-2) in acute respiratory

distress syndrome patients. Crit Care Med

2003;31(2):536-42

32. Warner RL, Beltran L, Younkin EM, et al.

Role of stromelysin 1 and gelatinase B in

experimental acute lung injury. Am J Respir

Cell Mol Biol 2001;24(5):537-44

• An animal model study showed that

compared to wild type, mice genetically

deficient in MMP-9 and MMP-3 were

protected from the development of acute

lung injury in an experimental model of

acute lung injury. MMP-3-deficient mice

also showed impaired neutrophil

recruitment in response to injury.

33. Nerusu KC, Warner RL, Bhagavathula N,

et al. Matrix metalloproteinase-3

(stromelysin-1) in acute inflammatory tissue

injury. Exp Mol Pathol 2007;83(2):169-76

34. Yamashita CM, Dolgonos L, Zemans RL,

et al. Matrix metalloproteinase 3 is a

mediator of pulmonary fibrosis. Am J

Pathol 2011;179(4):1733-45

•• Expression of MMP-3 is enhanced in the

lungs of patients with idiopathic

pulmonary fibrosis. Animal model

experiments revealed increased fibrosis in

the setting of transient adenoviral

vector-mediated expression of

recombinant MMP-3, while genetic

The importance of matrix metalloproteinase-3 in respiratory disorders Review

informahealthcare.com 419

Exp

ert R

evie

w o

f R

espi

rato

ry M

edic

ine

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Ron

a W

illia

mso

n on

07/

16/1

4Fo

r pe

rson

al u

se o

nly.

deletion of MMP-3 in mice protected

against bleomycin-induced pulmonary

fibrosis. In vitro experiments suggest that

MMP-3 induces epithelial-mesenchymal

transition via cleavage of E-cadherin and

subsequent activation of b-catenin

signaling pathways.

35. Martin EL, McCaig LA, Moyer BZ, et al.

Differential response of TIMP-3 null mice

to the lung insults of sepsis, mechanical

ventilation, and hyperoxia. Am J Physiol

Lung Cell Mol Physiol 2005;289(2):

L244-51

36. Gill SE, Huizar I, Bench EM, et al. Tissue

inhibitor of metalloproteinases 3 regulates

resolution of inflammation following acute

lung injury. Am J Pathol 2010;176(1):

64-73

37. Kruidenier L, MacDonald TT, Collins JE,

et al. Myofibroblast matrix

metalloproteinases activate the neutrophil

chemoattractant CXCL7 from intestinal

epithelial cells. Gastroenterology 2006;

130(1):127-36

38. Haro H, Crawford HC, Fingleton B, et al.

Matrix metalloproteinase-7-dependent

release of tumor necrosis factor-alpha in a

model of herniated disc resorption. J Clin

Invest 2000;105(2):143-50

39. Van Lint P, Libert C. Chemokine and

cytokine processing by matrix

metalloproteinases and its effect on

leukocyte migration and inflammation.

J Leukoc Biol 2007;82(6):1375-81

40. Yamakawa N, Uchida T, Matthay MA,

Makita K. Proteolytic release of the receptor

for advanced glycation end products from

in vitro and in situ alveolar epithelial cells.

Am J Physiol Lung Cell Mol Physiol 2011;

300(4):L516-25

• In vitro experiments showed that the

release of soluble receptor for advanced

glycation end products (RAGE) from

cultured alveolar epithelial cells is

mediated by proteolysis by MMP-3 and

MMP-13 following LPS treatment.

Soluble RAGE expression in rat

bronchoalveolar lavage was increased by

MMP-3 and MMP-13 proteolysis and in

humans antigen levels of these enzymes

were positively correlated with RAGE

levels in pulmonary edema fluid.

41. Hergrueter AH, Nguyen K, Owen CA.

Matrix metalloproteinases: all the RAGE in

the acute respiratory distress syndrome. Am

J Physiol Lung Cell Mol Physiol 2011;

300(4):L512-15

42. Uhlig S, Ranieri M, Slutsky AS. Biotrauma

hypothesis of ventilator-induced lung injury.

Am J Respir Crit Care Med 2004;169(2):

314-15; author reply 315

43. Tremblay LN, Slutsky AS.

Ventilator-induced lung injury: from the

bench to the bedside. Intensive Care Med

2006;32(1):24-33

44. Fan E, Villar J, Slutsky AS. Novel

approaches to minimize ventilator-induced

lung injury. BMC Med 2013;11:85

45. Albaiceta GM, Gutierrez-Fernandez A,

Garcıa-Prieto E, et al. Absence or inhibition

of matrix metalloproteinase-8 decreases

ventilator-induced lung injury. Am J Respir

Cell Mol Biol 2010;43(5):555-63

46. Albaiceta GM, Gutierrez-Fernandez A,

Parra D, et al. Lack of matrix

metalloproteinase-9 worsens

ventilator-induced lung injury. Am J Physiol

Lung Cell Mol Physiol 2008;294(3):

L535-43

47. Raghu G, Collard HR, Egan JJ, et al. An

official ATS/ERS/JRS/ALAT statement:

idiopathic pulmonary fibrosis:

evidence-based guidelines for diagnosis and

management. Am J Respir Crit Care Med

2011;183(6):788-824

48. Maher TM. Idiopathic pulmonary fibrosis:

pathobiology of novel approaches to

treatment. Clin Chest Med 2012;33(1):

69-83

49. Nathan SD, Shlobin OA, Weir N, et al.

Long-term course and prognosis of

idiopathic pulmonary fibrosis in the new

millennium. Chest 2011;140(1):221-9

50. King TE Jr, Pardo A, Selman M. Idiopathic

pulmonary fibrosis. Lancet 2011;378(9807):

1949-61

51. Wynn TA. Integrating mechanisms of

pulmonary fibrosis. J Exp Med 2011;

208(7):1339-50

52. Downey GP. Resolving the scar of

pulmonary fibrosis. N Engl J Med 2011;

365(12):1140-1

53. Xaubet A, Marin-Arguedas A, Lario S, et al.

Transforming growth factor-beta1 gene

polymorphisms are associated with disease

progression in idiopathic pulmonary

fibrosis. Am J Respir Crit Care Med 2003;

168(4):431-5

54. Seibold MA, Wise AL, Speer MC, et al.

A common MUC5B promoter

polymorphism and pulmonary fibrosis. N

Eng J Med 2011;364(16):1503-12

55. Armanios MY, Chen JJ, Cogan JD, et al.

Telomerase mutations in families with

idiopathic pulmonary fibrosis. N Engl J

Med 2007;356(13):1317-26

56. Alder JK, Cogan JD, Brown AF, et al.

Ancestral mutation in telomerase causes

defects in repeat addition processivity and

manifests as familial pulmonary fibrosis.

PLoS Genet 2011;7(3):e1001352

57. Tsakiri KD, Cronkhite JT, Kuan PJ, et al.

Adult-onset pulmonary fibrosis caused by

mutations in telomerase. Proc Natl Acad Sci

USA 2007;104(18):7552-7

58. Fingerlin TE, Murphy E, Zhang W, et al.

Genome-wide association study identifies

multiple susceptibility loci for pulmonary

fibrosis. Nat Genet 2013;45(6):613-20

59. Raghu G, Weycker D, Edelsberg J, et al.

Incidence and prevalence of idiopathic

pulmonary fibrosis. Am J Respir Crit Care

Med 2006;174(7):810-16

60. Sisson TH, Mendez M, Choi K, et al.

Targeted injury of type II alveolar epithelial

cells induces pulmonary fibrosis. Am J

Respir Crit Care Med 2010;181(3):254-63

61. Wang Q, Rajshankar D, Laschinger C,

et al. Importance of protein-tyrosine

phosphatase-alpha catalytic domains for

interactions with SHP-2 and

interleukin-1-induced matrix

metalloproteinase-3 expression. J Biol Chem

2010;285(29):22308-17

• In vitro experiments showed that the

release of soluble receptor for advanced

glycation end products (RAGE) from

cultured alveolar epithelial cells is

mediated by proteolysis by MMP-3 and

MMP-13 following LPS treatment.

Soluble RAGE expression in rat

bronchoalveolar lavage was increased by

MMP-3 and MMP-13 proteolysis and in

humans antigen levels of these enzymes

were positively correlated with RAGE

levels in pulmonary edema fluid.

62. Konigshoff M, Kramer M, Balsara N, et al.

WNT1-inducible signaling

protein-1 mediates pulmonary fibrosis in

mice and is upregulated in humans with

idiopathic pulmonary fibrosis. J Clin Invest

2009;119(4):772-87

63. Konigshoff M, Balsara N, Pfaff EM, et al.

Functional Wnt signaling is increased in

idiopathic pulmonary fibrosis. PLoS One

2008;3(5):e2142

64. Rajshankar D, Downey GP, McCulloch CA.

IL-1beta enhances cell adhesion to degraded

fibronectin. FASEB J 2012;26(11):4429-44

•• In vitro experiments demonstrated that

IL-1b-mediated cell adhesion is promoted

by proteolytic degradation of the matrix

by MMP-3.

Review Yamashita, Radisky, Aschner & Downey

420 Expert Rev. Respir. Med. 8(4), (2014)

Exp

ert R

evie

w o

f R

espi

rato

ry M

edic

ine

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Ron

a W

illia

mso

n on

07/

16/1

4Fo

r pe

rson

al u

se o

nly.

65. Alberg AJ, Brock MV, Ford JG, et al.

Epidemiology of lung cancer: diagnosis and

management of lung cancer, 3rd ed:

American College of Chest Physicians

evidence-based clinical practice guidelines.

Chest 2013;143(5 Suppl):e1S-29S

66. Molina JR, Yang P, Cassivi SD, et al.

Non-small cell lung cancer: epidemiology,

risk factors, treatment, and survivorship.

Mayo Clin Proc 2008;83(5):584-94

67. Reck M, Heigener DF, Mok T, et al.

Management of non-small-cell lung cancer:

recent developments. Lancet 2013;

382(9893):709-19

68. Lievre A, Milet J, Carayol J. Genetic

polymorphisms of MMP1, MMP3 and

MMP7 gene promoter and risk of colorectal

adenoma. BMC Cancer 2006;6:270

69. Hagemann T, Bozanovic T, Hooper S,

et al. Molecular profiling of cervical cancer

progression. Br J Cancer 2007;96(2):321-8

70. Fang S, Jin X, Wang R, et al.

Polymorphisms in the MMP1 and

MMP3 promoter and non-small cell lung

carcinoma in North China. Carcinogenesis

2005;26(2):481-6

71. Liu D, Guo H, Li Y, et al. Association

between polymorphisms in the promoter

regions of matrix metalloproteinases

(MMPs) and risk of cancer metastasis:

a meta-analysis. PLoS One 2012;7(2):

e31251

72. Mendes O, Kim HT, Stoica G. Expression

of MMP2, MMP9 and MMP3 in breast

cancer brain metastasis in a rat model. Clin

Exp Metastasis 2005;22(3):237-46

73. Sun T, Gao Y, Tan W, et al. Haplotypes in

matrix metalloproteinase gene cluster on

chromosome 11q22 contribute to the risk of

lung cancer development and progression.

Clin Cancer Res 2006;12(23):7009-17

74. Radisky DC, Przybylo JA. Matrix

metalloproteinase-induced fibrosis and

malignancy in breast and lung. Proc Am

Thorac Soc 2008;5(3):316-22

• Transgenic expression of MMP-3 in mice

results in fibrosis and tumor formation,

which is due to the induction of EMT, a

process which is dependent on the

sgeneration of reactive oxygen species.

75. Illman SA, Lehti K, Keski-Oja J, Lohi J.

Epilysin (MMP-28) induces TGF-beta

mediated epithelial to mesenchymal

transition in lung carcinoma cells. J Cell Sci

2006;119(Pt 18):3856-65

76. McGuire JK, Li Q, Parks WC. Matrilysin

(matrix metalloproteinase-7) mediates

E-cadherin ectodomain shedding in injured

lung epithelium. Am J Pathol 2003;162(6):

1831-43

77. Lochter A, Sternlicht MD, Werb Z, et al.

The significance of matrix

metalloproteinases during early stages of

tumor progression. Ann N Y Acad Sci

1998;857:180-93

78. Noel A, Boulay A, Kebers F, et al.

Demonstration in vivo that

stromelysin-3 functions through its

proteolytic activity. Oncogene 2000;19(12):

1605-12

79. Stallings-Mann ML, Waldmann J, Zhang Y,

et al. Matrix metalloproteinase induction of

Rac1b, a key effector of lung cancer

progression. Sci Transl Med 2012;4(142):

142ra95

•• Transgenic expression of Rac1b in mouse

lung epithelial cells stimulated EMT and

spontaneous tumor development by

MMP-induced mechanisms. Rac1b was

also found to be expressed in early-stage

human lung adenocarcinomas.

80. Bar-Sagi D, Hall A. Ras and Rho GTPases:

a family reunion. Cell 2000;103(2):227-38

81. Boettner B, Van Aelst L. The role of Rho

GTPases in disease development. Gene

2002;286(2):155-74

82. Price LS, Collard JG. Regulation of the

cytoskeleton by Rho-family GTPases:

implications for tumour cell invasion. Semin

Cancer Biol 2001;11(2):167-73

83. Sahai E, Marshall CJ. RHO-GTPases and

cancer. Nat Rev Cancer 2002;2(2):133-42

84. Jordan P, Brazao R, Boavida MG, et al.

Cloning of a novel human Rac1b splice

variant with increased expression in

colorectal tumors. Oncogene 1999;18(48):

6835-9

85. Schnelzer A, Prechtel D, Knaus U, et al.

Rac1 in human breast cancer:

overexpression, mutation analysis, and

characterization of a new isoform, Rac1b.

Oncogene 2000;19(26):3013-20

86. Fiegen D, Haeusler LC, Blumenstein L,

et al. Alternative splicing of Rac1 generates

Rac1b, a self-activating GTPase. J Biol

Chem 2004;279(6):4743-9

87. Matos P, Collard JG, Jordan P.

Tumor-related alternatively spliced Rac1b is

not regulated by Rho-GDP dissociation

inhibitors and exhibits selective downstream

signaling. J Biol Chem 2003;278(50):

50442-8

88. Orlichenko L, Geyer R, Yanagisawa M,

et al. The 19-amino acid insertion in the

tumor-associated splice isoform Rac1b

confers specific binding to p120 catenin.

J Biol Chem 2010;285(25):19153-61

89. Silva AL, Carmo F, Bugalho MJ. RAC1b

overexpression in papillary thyroid

carcinoma: a role to unravel. Eur J

Endocrinol 2013;168(6):795-804

90. Pelisch F, Khauv D, Risso G, et al.

Involvement of hnRNP A1 in the matrix

metalloprotease-3-dependent regulation of

Rac1 pre-mRNA splicing. J Cell Biochem

2012;113(7):2319-29

91. Nelson CM, Khauv D, Bissell MJ,

Radisky DC. Change in cell shape is

required for matrix

metalloproteinase-induced epithelial-

mesenchymal transition of mammary

epithelial cells. J Cell Biochem 2008;105(1):

25-33

92. Lee K, Chen QK, Lui C, et al. Matrix

compliance regulates Rac1b localization,

NADPH oxidase assembly, and

epithelial-mesenchymal transition. Mol Biol

Cell 2012;23(20):4097-108

93. Coussens LM, Fingleton B, Matrisian LM.

Matrix metalloproteinase inhibitors and

cancer: trials and tribulations. Science 2002;

295(5564):2387-92

94. Thomson S, Buck E, Petti F, et al.

Epithelial to mesenchymal transition is a

determinant of sensitivity of non-small-cell

lung carcinoma cell lines and xenografts to

epidermal growth factor receptor inhibition.

Cancer Res 2005;65(20):9455-62

95. Witta SE, Gemmill RM, Hirsch FR, et al.

Restoring E-cadherin expression increases

sensitivity to epidermal growth factor

receptor inhibitors in lung cancer cell lines.

Cancer Res 2006;66(2):944-50

96. Yauch RL, Januario T, Eberhard DA, et al.

Epithelial versus mesenchymal phenotype

determines in vitro sensitivity and predicts

clinical activity of erlotinib in lung cancer

patients. Clin Cancer Res 2005;11(24 Pt 1):

8686-98

The importance of matrix metalloproteinase-3 in respiratory disorders Review

informahealthcare.com 421

Exp

ert R

evie

w o

f R

espi

rato

ry M

edic

ine

Dow

nloa

ded

from

info

rmah

ealth

care

.com

by

Ron

a W

illia

mso

n on

07/

16/1

4Fo

r pe

rson

al u

se o

nly.