Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Oct 1.
Published in final edited form as: Semin Cancer Biol. 2012 Jul 31;22(5-6):471–483. doi: 10.1016/j.semcancer.2012.07.003

A comparison of epithelial-to-mesenchymal transition and re-epithelialization

Philip L Leopold a,*, Jan Vincent b, Hongjun Wang a
PMCID: PMC3595494  NIHMSID: NIHMS404929  PMID: 22863788

Abstract

Wound healing and cancer metastasis share a common starting point, namely, a change in the phenotype of some cells from stationary to motile. The term, epithelial-to-mesenchymal transition (EMT) describes the changes in molecular biology and cellular physiology that allow a cell to transition from a sedentary cell to a motile cell, a process that is relevant not only for cancer and regeneration, but also for normal development of multicellular organisms. The present review compares the similarities and differences in cellular response at the molecular level as tumor cells enter EMT or as keratinocytes begin the process of re-epithelialization of a wound. Looking toward clinical interventions that might modulate these processes, the mechanisms and outcomes of current and potential therapies are reviewed for both anti-cancer and pro-wound healing treatments related to the pathways that are central to EMT. Taken together, the comparison of re-epithelialization and tumor EMT serves as a starting point for the development of therapies that can selectively modulate different forms of EMT.

Keywords: Epithelial-to-mesenchymal transition, Keratinocyte, Dermal wound healing, Metastasis, Metastasis

1. Introduction

Epithelial-to-mesenchymal transition, or EMT, describes a series of cascades that lead to a change in cell phenotype from a stationary, differentiated phenotype characteristic of healthy epithelial tissue to a migratory, de-differentiated phenotype characteristic of a fibroblast or metastasizing tumor cell [1,2]. The idea of comparing EMT to normal physiological processes such as re-epithelialization and cell migration in development is not a new idea. Grunert et al. [3] compared EMT to a number of different types of changes in cells resulting from normal physiology, pathophysiology, and transformation in an attempt to define EMT as a process rather than any one observation. More recently, three distinct manifestations of EMT have been proposed [4,5] based on stages of development and associated biomarkers such that the EMT commonly associated with cancer metastasis (Type III) can be clearly differentiated from the involvement of EMT in either embryogenesis (Type I) or regeneration/fibrosis in mature tissue (Type II). The use of only three categories of EMT may ultimately fail to reflect the complexity of cell plasticity of cells as they respond to changing environments. For example, Acloque et al. [6] have differentiated three stages of Type I EMT during embryogenesis. Despite the focus on the differentiators of various manifestations of EMT, it is most useful to identify the similarities and common mechanisms that give remarkable plasticity to our cells, both tumor and non-tumor. This approach is especially valuable in light of the potential for cross-fertilization in understanding the mechanisms of both EMT and its converse process, mesenchymal-to-epithelial transition (MET) that accompanies both physiological and pathological forms of EMT.

This review seeks to identify and update similarities and differences between traditional EMT mechanisms as they are currently understood in tumor progression and the specific case of re-epithelialization which represents, perhaps, the most minimal and reversible form of regenerative EMT in normal tissues [7]. With respect to re-epithelialization, keratinocytes are taken as model for comparison with tumor EMT although it is clear that similar re-epithelialization occurs in epithelial cells of the airway, digestive tract, kidney, and cornea, just to name a few sites. Furthermore, the focus in this review will be on early events that transform a mature, differentiated keratinocyte into a migrating keratinocyte and how these events compare to the initial transformation of a primary tumor cell into a metastatic tumor cell. In addition to interesting comparisons and contrasts in the biology of these processes, there is a compelling clinical need to establish a functional differentiation between tumor EMT and normal tissue regeneration due to the fact that normal wound healing must continue, even in the context of anti-metastatic therapies. Therefore, anti-metastatic therapies should be developed with an understanding of potential impacts on wound healing just as wound healing therapies must avoid induction of metastasis in the event that a malignancy is present.

2. A comparison of re-epithelialization and epithelial-to-mesenchymal transition

The epithelial layer of keratinocytes is maintained in mature skin by differentiation of progenitor cells that occupy the basal layer of the skin [8]. Under normal conditions, keratinocytes go through a process of terminal differentiation during which the cells convert from a cuboidal epithelium to a squamous epithelium with concomitant cell death such that the outer layer of the epidermis is composed of the keratin cytoskeleton and associated lipids providing a mechanical and hydration barrier to protect the underlying tissue. The barrier function of keratinocytes is enhanced by multiple cell–cell and cell–substrate interactions including cadherin-associated desmosomes and adherens junctions, integrin-associated hemi-desmosomes, focal adhesions, and tight junctions [913]. Keratinocytes also maintain connexin-associated gap junctions that facilitate intercellular communication among keratinocytes [14]. Upon wounding, the cells of the skin proceed through a characteristic response leading to re-establishment of the mechanical and hydration barrier. Mechanical strength is provided by the extracellular matrix of the dermis while the hydration barrier is re-established through re-epithelialization. Upon experiencing a cutaneous wound, four phases of response occur. These include an inflammatory response, a granulation phase during which connective tissue and extracellular matrix components are rebuilt, epithelialization to cover the wound and re-establish the important barrier functions of the skin, and finally scar formation [15]. All too often, necrosis accompanies wound healing, and debridement of the necrotic tissue is required to enable keratinocyte access to the dermis so that re-epithelialization can occur [16].

The process of EMT describes the conversion of cells from sedentary, proliferative cells into migratory cells, a process that is essential to accomplish re-epithelialization during wound healing. In both wounded skin and tumors, cells undergoing EMT lose contact with each other, change their mode of association with the extracellular matrix, and become motile. In both cases, the cells must be able to re-model the extracellular matrix around them and, thus, secrete proteases to assist in the process. The changes that accompany wound healing are desirable and the triggers that initiate the change in phenotype are clearly definable. In contrast, the changes that occur in a tumor as it transitions from quiescent to metastatic are deleterious and the triggers are a conundrum. With an eye toward understanding the similarities and differences between tumor EMT and reepithelialization of a wound, the principal growth factors, signaling pathways, transcriptions controls, and target genes are compared in Table 1.

Table 1.

Keratinocyte responses during re-epithelialization vs. markers of EMT.a

Hallmarks of keratinocytes
in wound healing
Refs. Hallmarks of tumor cells
in EMT
Refs.
Growth factor responses

      EGF/TGF-α ↑ [1820] EGF ↑ [3234]
      TGF-β ↑ [21, 22] TGF-β ↑ [35, 36]
      KGF ↑ [26] KGF ↑ [53]
      IGF1 ↑ [23] IGF1 ↑ [4447]
      HGF ↑ [24, 25] HGF ↑ [43]
      PDGF ↑ [27, 28] PDGF ↑ [3742]
      TNFα ↑ [18] TNFα ↑ [4852]

      CCN2, 4 ↑; CCN 3, 5 ↓ [30, 31] CCN1, 4, 6 ↑; CCN 2, 3, 5 ↓ [5458]

Signaling pathways

      ERK1,2/JNK/p38 MAPK ↑ [59] ERK1,2/JNK/p38 MAPK ↑ [67, 68]
      ERK5 ↑ [7] ERK5 ↑ [7173]
      PI3K/Akt/mTOR ↑ [61] PI3K/Akt/mTOR ↑ [69, 70]
      RhoA ↑ [6466] RhoA ↑ [74]
      GSK3β ↓ [63] GSK3β ↓ [75, 76, 78]

Transcriptional/post-transcriptional regulation

      c-Fos ↑ [59, 109, 110] c-Fos ↑ [82, 111]
      β-catenin ↑ [80, 81] β-catenin ↑ [82, 83]

      Snail (Snail1) ↔ [85] Snail (Snail1) ↑ [9094]

      Slug (Snail2) ↑ [7, 84, 8789] Slug (Snail2) ↑ [86]
      Smad3/4 ↑ [21, 97, 98] Smad 3/4 [32, 94, 99, 100]
      Twist ↑ [108] Twist ↑ [101, 102]
      Lef-1 ↑ [19] Lef-1 ↑ [82, 103]

      Not reported Ets-1 ↑ [104, 105]
      Not reported Zeb-1 ↑ [91, 106]
      Not reported FOXC2 ↑ [107]

      MicroRNA [112, 113] MicroRNA [114116]

      Not reported miR-200/Zeb-1 feedback loop [157]

Cellular/extracellular matrix interactions

      Gap junctions
      Connexins (Cx26, Cx43) ↓ [117] Connexins (Cx26, Cx43) ↓ [118]
      Desmosomes

      Desmoglein 1 ↓ [119] Desmoplakin ↓ [86, 120, 121]
      Desmoglein 2 ↑ Desmoglein ↓
      Desmoglein 1 ↑ Plakoglobin ↓
      Desmoglein 3 ↓

      Hemidesomsomes

      α6β4 integrin ↔ [122, 123] α6β4 integrin ↓ [124]

      Adherens junctions
      E-cadherin ↓ [119] E-cadherin ↓ [74, 82, 83, 101]
      Tight junction

      ZO-1 ↔ [131] ZO-1 ↓ [126]
      Occludins ↔ Occludins [133]
      Claudin 1 ↔ Claudin 1 ↓ [134, 135]
      Coxsackie-adenovirus receptor ↔ or ↑ [132] Coxsackie-adenovirus receptor ↓ [94]

      Enhanced focal adhesions (increased interaction with fibronectin and vitron ectin)
      Integrin ↑: αVβ6, α2β1, α3β1, α5β1 [136, 137] Integrin ↑: αVβ6, α5β1, β4 [124, 138]
      Integrin ↓: αVβ5 [136]
      Other
      Not expressed N-cadherin ↑ [129, 130, 158]

      Scribble ↔ [156] Not reported

      Matrix metalloproteinases
      MMP-1 (collagenase 1), −3 (stromelysin 1) ↑ [151153] MMP-1 (collagenase 1), −3 (stromelysin 1) ↔/↑ [104, 147, 154]
      MMP-2 (gelatinase A), −9 (gelatinase B), −13 (collagenase 3), −14 (MT1-MMP) ↑ [139144] MMP-2 (gelatinase A), −9 (gelatinase B), −13 (collagenase 3), −14 (MT1-MMP) ↑ [92, 104, 145150]
a

Light gray shading indicates actual of potential differential responses between keratinocyte wound healing and tumor EMT.

2.1. Growth factors

Rapid responses and ongoing development of the wound healing response is coordinated by a series of growth factors [15,17]. Upon wounding of epidermal cells, blood clot formation induces the release of a host of growth factors from platelets. These growth factors play an important role in attracting macrophages and leucocytes. Together, the four cells types, keratinocytes, platelets, macrophages and leucocytes, release a host of additional growth factors including epidermal growth factor (EGF), transforming growth factor α (TGFα), and transforming growth factor β1 and 2 (TGFβ1, TGFβ2) [1822]. Later in wound healing, other growth factors act on keratinocytes including TGFβ3 from macrophages, insulin-like growth factor (IGF) from fibroblasts and epidermal cells, and hepatocyte growth factor (HGF) and keratinocyte growth factor (KGF) from fibroblasts [2327]. These growth factors play complementary roles in inducing keratinocyte proliferation and migration (see targets of signaling pathways discussed below). In addition, several other growth factors are actively involved in wound healing. For example, platelet derived growth factor (PDGF) is initially released from platelets at the site of the blood clot, later participates in an autocrine signaling loop in epithelial cells [27,28]. TNFα can be released from macrophages or directly from epithelial cells, and leads to synthesis and secretion of a host of pro-inflammatory cytokines [18]. Finally, a new layer of regulation recently came to light through our understanding of an exciting new group of molecules designated as the “CCN” family (based on the names of the first three members: Cysteine-rich Angiogenic Inducer 61; Connective Tissue Growth Factor; Nephroblastoma Overexpressed) [29,30]. The CCN protein family regulates many aspects of growth, development, and regeneration by interacting with integrins, triggering release of growth factors, cytokines, and matrix metalloproteinases, specific binding interactions with multiple extracellular matrix proteins, and by affecting bioavailability of growth-promoting molecules. CCN2 and CCN4 are specifically up-regulated during wound healing while CCN3 and CCN5 are down-regulated [2931].

The process of EMT for cancer cells aligns well with wound healing in terms of the growth factors that drive the process. Like wound healing, EMT is induced by increased levels of EGF, TGFβ1 [3236], and PDGF [3742]. In addition, hepatocyte growth factor (HGF) activation of the c-MET pathway is often part of the EMT milieu [43]. IGF1 and its receptor, IGFR have been implicated in EMT in breast cancer [4447]. TNFα is also a major signaling molecule involved in EMT in a variety of carcinomas [4850] and likely plays a key role in linking inflammation and EMT [51,52]. KGF has not been identified as a major player in EMT, but at least one study noted its contribution to producing an EMT phenotype in cultured carcinoma cells under specified growth conditions [53]. HGF (also known as scatter factor) was one of the key growth factors that led to the identification of paracrine regulation of EMT [43]. In summary, EGF, TGFβ1, PDGF, IGF, TNFα, and HGF show similar regulation in both re-epithelialization and EMT. Although KGF expression and signaling are consistent with proliferation of tumor cells, relatively little is known about KGF in EMT compare to the significant role played by KGF during re-epithelialization.

In both wound healing and tumor EMT, CCN3 and CCN5 are down-regulated while CCN4 is upregulated [5457]. CCN1 and CCN6 have also been characterized to have tumor promoting activity [55,57,58]. CCN2 expression is more complex. Although CCN2 expression occurs during wound healing, it is associated with fibrotic rather than epidermal healing [31] while CCN2 expression is low in many different tumors [29]. The precise spectrum of CCN proteins that show evidence of up- or down-regulation may be a critical point of distinction between wound healing and EMT.

2.2. Intracellular signaling

During wound healing, the growth factors that act on keratinocytes stimulate a distinct subset of the intracellular signaling pathways within these epidermal cells. The pathways include increased signaling through p21Ras, extracellular signal-regulated kinase (ERK) isoforms 1, 2, and 5, c-Jun NH2 terminal kinase (JNK), and p38 mitogen activated protein kinase (MAPK) pathways [7,59,60]. In addition, the IGF1-stimulated phosphoinositol-3-kinase/protein kinase B/mammalian target of rapamycin pathway, better known as the PI3K/Akt/mTOR axis, is also activated [61]. GSK3β plays an important role in integrating signals downstream of the EGF-stimulated MAPK pathway during keratinocyte migration in wound healing [62,63], and GSK3β must be suppressed to permit migration [63]. RhoGTPases are a group of Ras-family enzymes that act downstream of IGF receptor activation, and the increase in Rho activity serves as an intermediary in re-organization of the cytoskeleton during early responses to wound healing [6466].

EMT utilizes the same signaling pathways as wound healing by increasing the activity of both the Ras/ERK/MAPK pathway [67,68] and the PI3K/Akt/mTOR axis [69,70] during EMT. Even ERK5, which is not directly involved in the classical ERK/JNK/MAPK pathway, shows elevated activity in both wound healing and EMT [7173]. Likewise, RhoA activity is elevated in both settings [74], and GSK3β activity is reduced during EMT just as it is during wound healing [7578]. The similarity between the intracellular signaling pathways used by wound healing and EMT pathways is most likely a consequence of the similarity of the growth factors and receptors used to stimulate the process.

2.3. Transcription factors and post-transcriptional control

β-Catenin is part of the TGFβ signaling pathway and has a unique biology in that it resides at cell junctions in quiescent cells, but translocates to the nucleus to activate the Tcf–Lef-1 transcription factors upon activation [79]. β-Catenin is instrumental in the wound healing response [80] and migrates to the nucleus during the wounding response of keratinocytes [81]. β-Catenin translocation to the nucleus has long been a hallmark of EMT [82,83] and is often used as a diagnostic test to determine whether typical EMT changes are occurring [6].

Snail family transcription factors including Snail1 (Snail) and Snail2 (Slug) mediate gene expression changes downstream of the TGFβ and EGF receptors [84,85]. As transcription factors, Snail family members repress the expression of cell junction proteins (see below), another hallmark of EMT. While Slug activity is upregulated in both wounded epithelium and in tumor cells undergoing EMT [7,84,8689], Snail is only involved in EMT and has not been shown to be a major player in keratinocytes during wound healing [85,9094].

The Smad family of transcription factors has also been implicated in both wound healing and EMT. Smads, in a similar manner to Snail and Slug, act downstream of TGFβ signaling [95,96]. Smad3 and Smad4 expression are both required for normal wound healing, although Smad4 appears to have its primary activity through a paracrine mechanism while loss of Smad3 changes keratinocyte properties directly [21,97,98]. Smad-mediated signaling is fundamental to TGFβ-induced EMT [32,99,100], and Smad3 and 4 form complexes with Snail as part of TGFβ-induced EMT [94].

A host of other transcription factors have been implicated in EMT. Some prominent ones include Twist, Lef-1, Zeb-1, Ets-1, and FoxC2 [82,91,102107]. Among these factors, Twist and Lef-1 have demonstrated roles in keratinocyte wound healing [46,108], but Zeb-1, Ets-1, and FoxC2 remain to be characterized in an epidermal wound healing setting. The expression levels of the transcription factors themselves can be modified during epithelial wound response. For example, the c-Fos gene is among the earliest genes to show elevated expression in epithelial cells following a wound [59,109,110]. Induced nuclear localization of c-Fos and induced expression of c-Fos have both been used as models of EMT [82,111].

Post-transcriptional regulation has become an active area of research, and characterization of microRNA involvement in both wound healing and EMT is advancing rapidly. This topic has been the subject of several recent comprehensive reviews for both epidermal wound healing [112,113] and for EMT [114116]. There is significant, but not complete, overlap in the groups of microRNA that have been implicated in the two processes. MicroRNAs miR-21, -29a, -31, -34, -125b, -155, and -203 are involved in both processes. Of interest, a prominent feedback loop involving the miR-200 family and Zeb-1 has not yet been reported in epidermal wound healing [157].

2.4. Cell junctions

The change from a sedentary cell to a motile cell must ultimately involve a change in the way that cells interact with their environment. Strong, stable interactions such as most cell–cell junctions cannot be maintained during motility. At the same time, a new, strong but flexible interaction with the extracellular matrix is required to accomplish movement. As a result, upand down-regulation of the proteins involved in these junctions, quite naturally, represents the end target for many of the ligand/receptor/signal/transcription factor cascades described above. Gap junctions allow communication between adjacent cells, but such junctions become dispensible in both keratinocytes and tumor cells undergoing EMT, so connexin proteins, including Cx26 and Cx43, are downregulated [117,118]. Hemidesmosomes and desmosomes provide tissue integrity through the strength of the intercellular protein–protein interactions. Although there is a dearth of quantitative data on the subject, desmosome components (e.g., desmoglein and desmocollin) in keratinocytes appear to have a complex reaction to wounding during which the levels of different family members may be elevated or reduced [119]. In contrast, the desmosome disruption and resulting re-distribution of components has been clearly documented in EMT tumor cells [86,120,121]. The principal protein in hemidesmosomes, α6β4 integrin, releases from hemidesmosomes as they dissemble, but goes on to play an important role in fibronectin binding during cell migration. While α6β4 integrin persists in wounded keratinocytes [122,123], its expression level is reduced in EMT tumor cells [124]. E-cadherin presence at junctions is clearly reduced in both wounded keratinocytes [119] as well as in EMT tumor cells [74,82,83,101,104,125128] where loss of E-cadherin from adherens and tight junctions is a common feature of EMT. Interestingly, while E-cadherin expression is reduced, a non-epithelial cadherin, N-cadherin, often shows elevated expression in migrating tumor cells and is also a hallmark of EMT [129,130,158]. Tight junction proteins including zonula occludens 1, occludin, claudin 1, and coxsackie-adenovirus receptor show a differential response comparing wound healing to EMT. During wound healing, these proteins maintain their expression levels [131,132] while they are uniformly reduced in EMT cells [94,126,133135].

The cell matrix interactions of both wounded keratinocytes and EMT tumors require expression of integrins capable of binding fibronectin, laminin 5, and collagen I, all of which are observed in both cell types [124,136138]. Both migrating keratinocytes and EMT tumor cells continuously re-model the extracellular matrix around them. The two types of cells make copious use of matrix metalloproteinase (MMP) enzymes with elevated expression of MMP-2 (gelatinase A), MMP-9 (gelatinase B), MMP-13 (collagenase 3), and MMP-14 (MT1-MMP) in a consistent manner in skin [139144] and tumors [92,104,145150]. MMP-1 and -3 are also involved in wound healing [151153] and, to a lesser extent, in EMT [104,147,154].

Finally, Scribble is an actin-associated protein that helps to confer polarity in epithelial cell monolayers and has been identified as a component of the basolateral epithelial junction in Drosophila [155]. Scribble maintains its expression and function in migrating keratinocytes during wound healing where it confers polarity to the migration of keratinocytes in a wound [156]. It will be interesting to see what happens to Scribble in tumor cells and whether there is a polarized migration of tumor cells during EMT.

3. Clinical implications of the comparison of EMT and re-epithelialization

A number of anti-cancer drugs have been developed based on the receptors and/or signaling pathways that give rise to EMT. Given the prevalence of shared signaling and mechanisms between EMT and re-epithelialization, it stands to reason that inhibition of EMT may also result in the inhibition of wound healing at the level of epithelialization. It is not surprising to find a conflict between cancer therapy and wound healing when discussing anticancer drugs that target angiogenesis such as the anti-vascular endothelial growth factor (VEGF) monoclonal antibody, Bevacizumab (Avastin™) [159,160]. However, the focus here remains on mechanisms directly related to the transition of cell phenotypes at the onset of metastasis or wound healing. Table 2 lists data on adverse events of current anti-EMT and pro-wound healing therapies as well as potential therapeutic targets.

Table 2.

Therapeutic implications of EMT/re-epithelialization comparison.

Target Anti-EMT therapy Effect on re-epithelialization
EGF receptor (Her-1) Gefintinib (Iressa™) [238245] Rash in 53% of patients [246]; also, inhibition of corneal epithelialization [164]; case reports differ in opinion on potential significance for patients [247249]
EGF receptor (Her-1) Erlotinib (Tarceva™) [239241,243,250257] Rash in 79% of patients [246]; also delays tympanic membrane healing [165167]; case reports of epithelialization defect in the cornea [258] and scarring alopecia [259]
EGF receptor (Her-1) Cetuximab (Erbitux™ antibody) [240,260262] Acneiform skin rash in 90% of patients [162]; high incidence of skin toxicity during combined radiotherapy [263]; a related monoclonal antibody against EGFR has similar outcomes [162]
EGF receptor (Her-1) and Her-2 Lapatinib [264] Extensive cutaneous effects [265]
TGFβ-receptor 1, TGFβ-receptor 2 LY2109761 [266271] No reports, but Flechsig [272] report that LY2109761 has anti-fibrotic activity in lung suggesting a potential effect on wound healing
TGFβ-receptor 1 SM16 [273,274] No reports on epithelialization
PDGF receptor (also targets c-KIT and BCR-ABL tyrosine kinases) Imatinib mesylate (Gleevec™) [275,276] 30–45% of patients experience skin eruptions [169]; in an experimental study, imatinib delayed wound healing; direct effect on epithelialization implied by Figure 2 although direct effect on epithelialization vs. indirect effect through impaired fibrosis was not clear [170]; other multikinase inhibitors including sorafenib and sunitinib have similar problems [162]
Endothelin A receptor ABT-627 or ZD4054 [170172] No reports of effects on epithelialization
Hepatocyte growth factor receptor (c-MET) Tivantinib (ARQ 147) [173] No cutaneous adverse events in a phase I dose escalation study [174]

Target Pro-epithelialization therapy Effect on EMT

PDGF receptor Becaplermin (Regranex™, rhPDGF-BB) [277280] Becaplermin produces EMT in tumor cells [38,42,182]; although there was no report of increased incidence of cancer in several clinical trials with topical application [175181], a post-marketing study concluded that there was a dose-dependent increased risk of death from cancer that has now been incorporated into the product label [183]; more recently, a follow up study concluded that there was no statistically significant link between Becaplermin therapy and either cancer incidence or severity [184]
EGF receptor/insulin-like growth factor receptor I Cathelicidin/LL-37 [185190] Induces metastatic phenotype in breast cancer [189]; causes ovarian tumor progression [190]

Potential targets Re-epithelialization/EMT relationship

Tissue transglutaminase Activated during corneal re-epithelialization [200]; proposed target for anti-EMT drug [203]
β-1,6-N-acetylglucosaminyltransferase V (Mgat5) Upregulated in carcinomas [206]; increased expression enhances wound healing [108]
Sphingosine-1-PO4 receptor Anti-proliferative, anti-motility effects in keratinocytes that express Smad3 [207209]; proposed target for EMT drugs [212,213,237]
Complex protease mixtures Proteases have been used to debride wounds [229,231]; however, proteases also modulate the wound healing response at many levels [228] and may have a synergistic ability to improve epithelialization [230,231]
Prostaglandin E2 Inhibits EMT in transformed kidney epithelial (MDCK) cells [214]; little or no effect on wound healing [215,216]; however, enhances EMT in colorectal cancer (Caco2) cells [217] and favors intestinal tumor growth [218]
Hypoxia inducible factors (HIFs) Mixed data on effects of anti-HIF treatments and hyperbaric oxygen on tumor growth and re-epithelialization [219224]
AMP-activated protein kinase/Glut1 glucose transporter Metformin (hepatic gluconeogenesis inhibitor) reverses EMT-like characteristics of transformed cells [232,233]; curiously, metformin was also reported to speed healing of ulcerations including epithelialization [234]; these findings must be considered within the context of the conflicting reports that high glucose inhibits proliferation and induces differentiation of keratinocytes [235] while Li et al. [236] provide evidence that hyperglycemia elevates metastatic risk in the pancreas
EGF receptor via the P2Y receptor ATP, UTP, and diadenosine polyphosphates have been reported to have enhancing and inhibitory properties with respect to keratinocyte spreading and migration, in part due to use of multiple receptor and G proteins [281290]; P2Y receptors are elevated in some cancers [291], enhance interaction of tumor cells with lymphatic endothelium [292], and, most recently, have been implicated in Ca2+ signaling during EGF-induced EMT [293]

3.1. Effect of anti-metastasis drugs on wound healing

To review the potential for adverse events relating to wound healing in the context of anti-EMT therapies, several EMT-directed therapies were evaluated with regard to adverse effects on wound healing. This topic has already been the subject of significant concern [161163]. Four therapies that target the EGF receptor were examined including Gefintinib (Iressa™), Erlotinib (Tarceva™), the monoclonal antibody, Cetuximab (Erbitux™ antibody), and Lapatinib. All four therapies target EGF receptor 1 (Her-1/Erb1) while Lapatinib also inhibits Her-2 (Erb2). All four therapies are associated with significant rash formation as described in reviews of the clinical literature as well as case reports. In the cases of Gefintinib and Erlotinib, experimental models suggest that the therapies can cause direct inhibition of re-epithelialization [164167].

Several multikinase drugs have been developed with Imatinib (Gleevec™) being the most prominent. Imatinib acts on the PDGF receptor as well as the tyrosine kinases such as c-KIT and the Bcr-Abl gene product [168]. Sorafenib and Sunitinib are two other multikinase inhibitors that, along with Imatinib, cause rashes [162,169]. Imatinib, as a PDGF inhibitor, has been reported to inhibit fibrosis, but PDGF also has a role in epithelialization and a careful examination of the data from Rajkumar [170] shows that epithelial cell proliferation in healing skin is diminished by imatinib treatment.

Some therapies may have less dermal involvement. TGFβ receptor 1 and TGFβ receptor 2 have also been used as targets for anti-EMT therapies. LY2109761 and SM16 directly against these targets have not been examined in depth with respect to their effects on re-epithelialization. However, should the experience with EGF receptors hold, cutaneous side effects can be anticipated. Two other classes of drugs targeted at the endothelin receptor (ABT-627 or ZD4054) and the HGF receptor (Tivantinib) show anti-EMT activity [171173]. Detrimental effects on wound healing have not been reported in the case of these drugs, and there was a distinct lack of skin reactions in a phase I trial of Tivantinib [174]. These results either indicate that certain pathways for blocking EMT are inherently less toxic to the process of cutaneous wound healing or that dose-limiting toxicity was not yet reached and that further investigation is warranted.

3.2. Effect of wound-healing drugs on metastasis

While there is a wealth of data on the effects of chemotherapy drugs on wound healing, there are relatively few drugs that have been approved to enhance wound epithelialization, and thus less opportunity to evaluate a connection between drugs that enhance epithelialization and cancer metastasis.

Becaplermin (Regranex™), a form of recombinant human PDGF based on a homodimer of the PDGF-B isoform, is an FDA-approved growth factor for wound care. Although extensive experience in clinical trials has not suggested problems related to the advancement of cancer [175181], laboratory experiments have demonstrated the concept that PDGF can induce the EMT phenotype [38,42,182]. A phase IV post-marketing study of Becaplermin noted an apparent dose-dependent increase in mortality due to cancer although no overall increased incidence of cancer was noted, leading to a modified label of the product [183]. This finding was particularly concerning since exactly this type of accelerated cancer as a by-product of growth factor therapy is a common concern with growth-promoting biologics. Later, a larger study failed to indicate a higher cancer incidence or mortality rate in a treated population [184].

Another potential wound healing biologic that has direct effects on keratinocytes is the cathelicidin-derived peptide, LL-37. Although originally identified for anti-microbial activities, LL-37 can interact directly with keratinocytes causing epidermal cell migration [185]. The surprising biological activity of LL-37 was later traced to its ability to induce cell signaling via binding to the EGF receptor and insulin-like growth factor receptor type I [186188]. The potential for cross-talk between re-epithelialization and EMT is particularly clear with LL-37 due to its ability to promote metastatic phenotypes in breast and ovarian cancer models [189,190]. Not surprisingly, high levels of LL-37 have been found in breast tumors [191].

Other FDA approved treatments for wound healing include therapeutics based on fibrin and non-physiological polymers. Fibrin glues have also been approved by the FDA for wound healing (Evicel™, Tisseel™, Artiss™), but these products are not examined in detail herein due to the fact that keratinocytes do not bind to fibrin due to a lack of αVβ3 integrin expression [192,193]. However, the use of fibrin does echo the issue of biologics and EMT since melanoma cells, upon a shift toward a more aggressive phenotype begin expressing αVβ3 integrin [194]. Likewise, cyanoacrylate polymers have long been used in wound healing. However, due to their chemical rather than biologic nature, a discussion of cyanoacrylates is beyond the scope of this review. Fortunately, the use of cyanoacrylates, including 2-octylcyanacrylate (Dermabond™), has not been associated with EMT or metastasis [195].

3.3. Potential for identifying new targets for wound healing or anti-EMT therapies

The value in identifying similarities and differences between wound healing therapeutics and EMT therapeutics lies in the comparison of drug targets and the potential development of new strategies for intervention. A review of the literature suggests several potential targets for anti-EMT drugs/pro-wound healing drugs based on patterns of effects regarding EMT and epithelialization that are similar to several of the drugs under development or approved for use as described in Sections 3.1 and 3.2 (Table 2).

Potential anti-EMT/pro-wound healing targets have consistent actions when comparing their activities in epithelial wound healing and EMT. Transglutaminase 2, also known as tissue transglutaminase, is a widely expressed enzyme involved in formation of covalent crosslinks between the primary amine on glutamine and a primary amino group on another molecule, e.g., a lysine on another protein [196]. The modifications that transglutaminases carry out are typically considered to be important for stabilization of proteins for the purpose of increased mechanical strength and resistance to degradation, e.g., as needed in hair and skin. While skin and hair follicle keratinocytes express a unique form transglutaminase as part of their normal function [197,198], tissue transglutaminase activity is upregulated during epithelial wound healing in skin as well as cornea [199,200]. Mehta, Verma and colleagues have made an argument that tissue transglutaminase should be good target for inhibition of cancer progression [201203] based on the fact that tissue transglutaminase activity is elevated in metastatic cancers (reviewed by [203]) and that inhibition of transglutaminase activity reduced cancer progression [204,205].

While tissue transglutaminase has been long recognized for its importance in the skin and in wound healing and only recently was suggested as an anti-cancer target, the converse is true for β-1,6-N-acetylglucosaminyltransferase V (Mgat5). Mgat5, an enzyme that controls crosslinking of carbohydrate groups at the cell surface, can crosslink growth factor receptors including the EGF receptor and TGFβ receptor, thus delaying their removal from the cell surface [206]. In addition, Mgat5 is involved with positive feedback in the EMT pathway since it is also upregulated by TGFβ signaling [206]. Only recently was there a suggestion that the inherent growth potentiation observed with Mgat5 might also contribute to re-epithelialization [108]. Certainly, transient upregulation of Mgat5 might be very helpful in wound healing in contrast to constitutive upregulation that promotes metastasis.

Sphingosine-1-PO4 (S1P), like tissue transglutaminase and Mgat5, induces epithelial migration, but does so without increasing proliferation [207209]. Thus, S1P would appear to be an anti-EMT candidate molecule. An earlier characterization of S1P as inducing differentiation stood out as contrary to the complementary effects that S1P shares with TGFβ; however, these finding can now be reconciled since the elevated transglutaminase levels observed by Vogler et al. were likely an increase in tissue transglutaminase as opposed to keratinocyte transglutaminase, and thus could be understood to be a marker of EMT [203,207]. Along these same lines of reasoning, S1P has been linked to both positive and negative regulation of metastasis in melanoma cells [210] and elevated expression levels in colon carcinoma [211]. It also explains why inhibitors of this pathway are of interest in anti-cancer drug development [212,213]. In summary, sphingosine-1-phosphate and its receptor play context-dependent roles in wound healing and EMT.

Some interventions that appear to have simple, straightforward effects suffer from multiple, complex interactions in in vivo systems due to the presence of multiple target cells and competing pathways. Prostaglandin E2, like LL-37 discussed above, has multiple targets and, thus, defies a simple relationship to cancer progression and wound healing. Prostaglandins are well known for their antiinflammatory effects and, thus, would be expected to limit both wound healing and EMT which depend on inflammatory signaling. A report that prostaglandin E2 inhibited EMT was consistent with our understanding of the mechanism of EMT [214]. Similarly, prostaglandin E2 failed to show significant effects on wound healing [215,216]. However, using alternative experimental systems, Tanaka et al. showed that prostaglandin E2 favored EMT-like changes in Caco-2 cells, with the notable exception that ZO-1 was not removed from tight junctions [217]. In fact, ZO-1 retention is recognized as one of the differentiating factors between wound healing and tumor EMT as highlighted in Table 1. Interestingly, a separate report shows that prostaglandin E2 accelerates tumor growth of intestinal tumors via a novel mechanism involving DNA methylation [218] and highlights the difficulty in accomplishing manipulation of cell physiology with surgical precision in vivo. The hypoxia response, like that of prostaglandins, is complex, touching on angiogenic growth factors produced in response to low tissue oxygenation, changes in levels of reactive oxygen species, and changes in metabolism. As a result, hypoxia and responses to hyperbaric oxygen do not follow a simple formula when comparing in vitro and in vivo data [219224].

The elevated levels of MMPs characterized during epidermal wound healing suggest that the extracellular milieu of a wound is highly proteolytic. This environment might have wound healing significance over and above the need for re-modeling of the extracellular matrix. Prior work on inflammatory responses has demonstrated that neutrophil proteases including elastase and cathepsin G cause proteolysis of extracellular matrix proteins, as expected, but also cleave insulin-like growth factor 1 as well as its binding proteins [225]. Extrapolating to a wound, the highly proteolytic environment might be changing signaling pathways in addition to structure. MT1-MMP has been recognized to cause changes in the migration of prostate cancer cells by a mechanism involving cleavage of the extracellular matrix protein, laminin-5 [226]. Upstream of MMPs and signaling molecules, there exists a complex protease-based activation system that modulates both cancer progression and wound healing through the use of enzyme-activating convertases [227,228]. Therefore, one might imagine that wholesale intervention at the level of proteolysis might have a beneficial effect on wound healing. Various enzymatic combinations have been used for debridement of wounds [229]. A preparation of enzymes from North Atlantic krill has been effective at debridement, but has also shown promise in improving epithelialization [230,231].

The final potential target to be addressed here is the cellular metabolism, particularly as controlled by glucose levels. The drug, metformin, is known to inhibit hepatic gluconeogenesis and to lower blood glucose levels in vivo. Recently, metformin was reported to reverse cellular changes associated with EMT [232,233] using in vitro models. As a result, one would expect metformin to inhibit wound healing. However, metformin sped overall wound healing and epithelialization, in particular, in ulcers [234]. The apparently disparate results may reflect an underlying complexity in how metabolism affects tumors vs. normal tissue. Sparvchikov et al. [235] reported that high glucose inhibited proliferation of keratinocytes and, instead, favored keratinocyte differentiation. In contrast, Li et al. [236], studying hyperglycemia, found that hyperglycemia elevated metastatic risk, at least for pancreatic cancer.

4. Conclusions

As noted above, EMT is now broadly recognized as a normal physiological process that occurs in development (Type I EMT) and in tissue regeneration (Type II EMT) as well as in pathological conditions such as metastasis (Type III EMT). Therefore, it is not surprising that these processes share so many biochemical mechanisms. Future development of therapies that enhance wound healing without promoting tumor growth and fight metastasis without compromising wound healing will require precision targeting of pathways that are differentially utilized by these processes. This review has only started the process of a full accounting of shared and distinct pathways between tumor EMT and wound healing. Even in the cross section of mechanisms discussed here, there appear to be opportunities for differential treatment including potential targets such as CCN2, Snail transcription, and, possibly tight junction proteins that play roles in cytosolic sequestration of transcription factors. Other potential targets may require further research for validation, but Ets-1, Zeb-1, FoxC2, and miR200 are likely candidates for selective manipulation of Type III EMT. In addition to the value in identifying differences between the Type II and III pathways, there is also potential value in understanding the similarities. Insofar as some drug targets are likely to be shared between the two pathways, validation of a target in one pathway may serve as a starting point for drug development in the other pathway. Control of drug delivery may be the key to the use of therapies that have potential to stimulate or inhibit both healing and metastasis.

In part, because of all of the individual relationships that are coming to light through wound healing and cancer research, the most significant challenge going forward appears to be one of bioinformatics. Our understanding of the individual molecular events is excellent and growing by the day. However, our ability to integrate all of the molecular controls and feedback into a meaningful picture of wound healing or metastasis is limiting. At this point, understanding how entire families of chemokines, receptors, signal transducers, transcription factors, micro RNAs, and target proteins are coordinately controlled is not a trivial problem. Improved computational models that can incorporate all of the knowledge about the pathways in a quantitative manner will likely represent the next major step toward developing therapies in this arena.

Acknowledgements

This work was supported by the BioInnovation Program at the Stevens Institute of Technology (PL, HW), a grant from the National Institutes of Health NIAMS 1R21 AR056416 (HW), and Arcimboldo, AB (JV).

Footnotes

Conflict of interest statement

JV declares the receipt of salary and other compensation due to his position as Director of Research and Development for Arcimboldo, AB.

References

  • 1.Savagner P. The epithelial–mesenchymal transition (EMT) phenomenon. Annals of Oncology. 2010;21(Suppl. 7):vii89–vii92. doi: 10.1093/annonc/mdq292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Chaffer CL, Weinberg RA. A perspective on cancer cell metastasis. Science. 2011;331:1559–1564. doi: 10.1126/science.1203543. [DOI] [PubMed] [Google Scholar]
  • 3.Grunert S, Jechlinger M, Beug H. Diverse cellular and molecular mechanisms contribute to epithelial plasticity and metastasis. Nature Reviews Molecular Cell Biology. 2003;4:657–665. doi: 10.1038/nrm1175. [DOI] [PubMed] [Google Scholar]
  • 4.Kalluri R, Weinberg RA. The basics of epithelial–mesenchymal transition. The Journal of Clinical Investigation. 2009;119:1420–1428. doi: 10.1172/JCI39104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Zeisberg M, Neilson EG. Biomarkers for epithelial–mesenchymal transitions. The Journal of Clinical Investigation. 2009;119:1429–1437. doi: 10.1172/JCI36183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Acloque H, Adams MS, Fishwick K, Bronner-Fraser M, Nieto MA. Epithelial–mesenchymal transitions: the importance of changing cell state in development and disease. The Journal of Clinical Investigation. 2009;119:1438–1449. doi: 10.1172/JCI38019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Arnoux V, Nassour M, L’Helgoualćh A, Hipskind RA, Savagner P. Erk5 controls Slug expression and keratinocyte activation during wound healing. Molecular Biology of the Cell. 2008;19:4738–4749. doi: 10.1091/mbc.E07-10-1078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Clayton E, Doupe DP, Klein AM, Winton DJ, Simons BD, Jones PH. A single type of progenitor cell maintains normal epidermis. Nature. 2007;446:185–189. doi: 10.1038/nature05574. [DOI] [PubMed] [Google Scholar]
  • 9.Pummi K, Malminen M, Aho H, Karvonen SL, Peltonen J, Peltonen S. Epidermal tight junctions: ZO-1 and occludin are expressed in mature, developing, and affected skin and in vitro differentiating keratinocytes. The Journal of Investigative Dermatology. 2001;117:1050–1058. doi: 10.1046/j.0022-202x.2001.01493.x. [DOI] [PubMed] [Google Scholar]
  • 10.Watt FM. Role of integrins in regulating epidermal adhesion, growth and differentiation. EMBO Journal. 2002;21:3919–3926. doi: 10.1093/emboj/cdf399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Litjens SH, de Pereda JM, Sonnenberg A. Current insights into the formation and breakdown of hemidesmosomes. Trends in Cell Biology. 2006;16:376–383. doi: 10.1016/j.tcb.2006.05.004. [DOI] [PubMed] [Google Scholar]
  • 12.Green KJ, Simpson CL. Desmosomes: new perspectives on a classic. Journal of Investigative Dermatology. 2007;127:2499–2515. doi: 10.1038/sj.jid.5701015. [DOI] [PubMed] [Google Scholar]
  • 13.Niessen CM, Gottardi CJ. Molecular components of the adherens junction. Biochimica et Biophysica Acta. 2008;1778:562–571. doi: 10.1016/j.bbamem.2007.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Mese G, Richard G, White TW. Gap junctions: basic structure and function. Journal of Investigative Dermatology. 2007;127:2516–2524. doi: 10.1038/sj.jid.5700770. [DOI] [PubMed] [Google Scholar]
  • 15.Singer AJ, Clark RA. Cutaneous wound healing. New England Journal of Medicine. 1999;341:738–746. doi: 10.1056/NEJM199909023411006. [DOI] [PubMed] [Google Scholar]
  • 16.Hellgren L, Vincent J. Debridement: an essential step in wound healing. In: Westerhof W, editor. Leg ulcers: diagnosis and treatment. Amsterdam, London, New York, Tokyo: Elseviser Scienve Publication; 1993. pp. 305–312. [Google Scholar]
  • 17.Barrientos S, Stojadinovic O, Golinko MS, Brem H, Tomic-Canic M. Growth factors and cytokines in wound healing. Wound Repair and Regeneration. 2008;16:585–601. doi: 10.1111/j.1524-475X.2008.00410.x. [DOI] [PubMed] [Google Scholar]
  • 18.Tomic-Canic M, Komine M, Freedberg IM, Blumenberg M. Epidermal signal transduction and transcription factor activation in activated keratinocytes. Journal of Dermatological Science. 1998;17:167–181. doi: 10.1016/s0923-1811(98)00016-4. [DOI] [PubMed] [Google Scholar]
  • 19.Kim I, Mogford JE, Chao JD, Mustoe TA. Wound epithelialization deficits in the transforming growth factor-alpha knockout mouse. Wound Repair and Regeneration. 2001;9:386–390. doi: 10.1046/j.1524-475x.2001.00386.x. [DOI] [PubMed] [Google Scholar]
  • 20.Hardwicke J, Schmaljohann D, Boyce D, Thomas D. Epidermal growth factor therapy and wound healing—past, present and future perspectives. Surgeon. 2008;6:172–177. doi: 10.1016/s1479-666x(08)80114-x. [DOI] [PubMed] [Google Scholar]
  • 21.Roberts AB, Russo A, Felici A, Flanders KC. Smad3: a key player in pathogenetic mechanisms dependent on TGF-beta. Annals of the New York Academy of Sciences. 2003;995:1–10. doi: 10.1111/j.1749-6632.2003.tb03205.x. [DOI] [PubMed] [Google Scholar]
  • 22.Klass BR, Grobbelaar AO, Rolfe KJ. Transforming growth factor beta1 signalling, wound healing and repair: a multifunctional cytokine with clinical implications for wound repair, a delicate balance. Postgraduate Medical Journal. 2009;85:9–14. doi: 10.1136/pgmj.2008.069831. [DOI] [PubMed] [Google Scholar]
  • 23.Brown DL, Kane CD, Chernausek SD, Greenhalgh DG. Differential expression and localization of insulin-like growth factors I and II in cutaneous wounds of diabetic and nondiabetic mice. American Journal of Pathology. 1997;151:715–724. [PMC free article] [PubMed] [Google Scholar]
  • 24.Cowin AJ, Kallincos N, Hatzirodos N, Robertson JG, Pickering KJ, Couper J, et al. Hepatocyte growth factor and macrophage-stimulating protein are upregulated during excisional wound repair in rats. Cell and Tissue Research. 2001;306:239–250. doi: 10.1007/s004410100443. [DOI] [PubMed] [Google Scholar]
  • 25.Chmielowiec J, Borowiak M, Morkel M, Stradal T, Munz B, Werner S, et al. c-Met is essential for wound healing in the skin. Journal of Cell Biology. 2007;177:151–162. doi: 10.1083/jcb.200701086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Werner S, Krieg T, Smola H. Keratinocyte–fibroblast interactions in wound healing. Journal of Investigative Dermatology. 2007;127:998–1008. doi: 10.1038/sj.jid.5700786. [DOI] [PubMed] [Google Scholar]
  • 27.Antoniades HN, Galanopoulos T, Neville-Golden J, Kiritsy CP, Lynch SE. Injury induces in vivo expression of platelet-derived growth factor (PDGF) and PDGF receptor mRNAs in skin epithelial cells and PDGF mRNA in connective tissue fibroblasts. Proceedings of the National Academy of Sciences of the United States of America. 1991;88:565–569. doi: 10.1073/pnas.88.2.565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Deuel TF, Kawahara RS, Mustoe TA, Pierce AF. Growth factors and wound healing: platelet-derived growth factor as a model cytokine. Annual Review of Medicine. 1991;42:567–584. doi: 10.1146/annurev.me.42.020191.003031. [DOI] [PubMed] [Google Scholar]
  • 29.Dhar A, Ray A. The CCN family proteins in carcinogenesis. Experimental Oncology. 2010;32:2–9. [PubMed] [Google Scholar]
  • 30.Jun JI, Lau LF. Taking aim at the extracellular matrix: CCN proteins as emerging therapeutic targets. Nature Reviews Drug Discovery. 2011;10:945–963. doi: 10.1038/nrd3599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Rittie L, Perbal B, Castellot JJ, Jr, Orringer JS, Voorhees JJ, Fisher GJ. Spatial–temporal modulation of CCN proteins during wound healing in human skin in vivo. Journal of Cell Communication and Signaling. 2011;5:69–80. doi: 10.1007/s12079-010-0114-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Zavadil J, Narasimhan M, Blumenberg M, Schneider RJ. Transforming growth factor-beta and microRNA:mRNA regulatory networks in epithelial plasticity. Cells Tissues Organs. 2007;185:157–161. doi: 10.1159/000101316. [DOI] [PubMed] [Google Scholar]
  • 33.Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A, Farshid G, et al. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nature Cell Biology. 2008;10:593–601. doi: 10.1038/ncb1722. [DOI] [PubMed] [Google Scholar]
  • 34.Park SM, Gaur AB, Lengyel E, Peter ME. The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes and Development. 2008;22:894–907. doi: 10.1101/gad.1640608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Fuxe J, Karlsson MC. TGF-β-induced epithelial–mesenchymal transition: a link between cancer and inflammation. Seminars in Cancer Biology. 2012 May; doi: 10.1016/j.semcancer.2012.05.004. http://dx.doi.org/10.1016/j.semcancer.2012.05.004 [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
  • 36.Wendt MK, Tian M, Schiemann WP. Deconstructing the mechanisms and consequences of TGF-beta-induced EMT during cancer progression. Cell and Tissue Research. 2012;347:85–101. doi: 10.1007/s00441-011-1199-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Gotzmann J, Fischer AN, Zojer M, Mikula M, Proell V, Huber H, et al. A crucial function of PDGF in TGF-beta-mediated cancer progression of hepatocytes. Oncogene. 2006;25:3170–3185. doi: 10.1038/sj.onc.1209083. [DOI] [PubMed] [Google Scholar]
  • 38.Jechlinger M, Sommer A, Moriggl R, Seither P, Kraut N, Capodiecci P, et al. Autocrine PDGFR signaling promotes mammary cancer metastasis. Journal of Clinical Investigation. 2006;116:1561–1570. doi: 10.1172/JCI24652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Yang L, Lin C, Liu ZR. P68 RNA helicase mediates PDGF-induced epithelial mesenchymal transition by displacing Axin from beta-catenin. Cell. 2006;127:139–155. doi: 10.1016/j.cell.2006.08.036. [DOI] [PubMed] [Google Scholar]
  • 40.Kong D, Wang Z, Sarkar SH, Li Y, Banerjee S, Saliganan A, et al. Platelet-derived growth factor-D overexpression contributes to epithelial–mesenchymal transition of PC3 prostate cancer cells. Stem Cells. 2008;26:1425–1435. doi: 10.1634/stemcells.2007-1076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Kong D, Li Y, Wang Z, Banerjee S, Ahmad A, Kim HR, et al. miR-200 regulates PDGF-D-mediated epithelial–mesenchymal transition, adhesion, and invasion of prostate cancer cells. Stem Cells. 2009;27:1712–1721. doi: 10.1002/stem.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Patel P, West-Mays J, Kolb M, Rodrigues JC, Hoff CM, Margetts PJ. Platelet derived growth factor B and epithelial mesenchymal transition of peritoneal mesothelial cells. Matrix Biology. 2010;29:97–106. doi: 10.1016/j.matbio.2009.10.004. [DOI] [PubMed] [Google Scholar]
  • 43.Li Y, Bhargava MM, Joseph A, Jin L, Rosen EM, Goldberg ID. Effect of hepatocyte growth factor/scatter factor and other growth factors on motility and morphology of non-tumorigenic and tumor cells. In Vitro Cellular and Developmental Biology Animal. 1994;30A:105–110. doi: 10.1007/BF02631401. [DOI] [PubMed] [Google Scholar]
  • 44.Playford MP, Bicknell D, Bodmer WF, Macaulay VM. Insulin-like growth factor 1 regulates the location, stability, and transcriptional activity of beta-catenin. Proceedings of the National Academy of Sciences of the United States of America. 2000;97:12103–12108. doi: 10.1073/pnas.210394297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Morali OG, Delmas V, Moore R, Jeanney C, Thiery JP, Larue L. IGF-II induces rapid beta-catenin relocation to the nucleus during epithelium to mesenchyme transition. Oncogene. 2001;20:4942–4950. doi: 10.1038/sj.onc.1204660. [DOI] [PubMed] [Google Scholar]
  • 46.Kim HJ, Litzenburger BC, Cui X, Delgado DA, Grabiner BC, Lin X, et al. Constitutively active type I insulin-like growth factor receptor causes transformation and xenograft growth of immortalized mammary epithelial cells and is accompanied by an epithelial-to-mesenchymal transition mediated by NF-kappaB and snail. Molecular and Cellular Biology. 2007;27:3165–3175. doi: 10.1128/MCB.01315-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Sivakumar R, Koga H, Selvendiran K, Maeyama M, Ueno T, Sata M. Autocrine loop for IGF-I receptor signaling in SLUG-mediated epithelial–mesenchymal transition. International Journal of Oncology. 2009;34:329–338. [PubMed] [Google Scholar]
  • 48.Bates RC, Mercurio AM. Tumor necrosis factor-alpha stimulates the epithelial-to-mesenchymal transition of human colonic organoids. Molecular Biology of the Cell. 2003;14:1790–1800. doi: 10.1091/mbc.E02-09-0583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Dong R, Wang Q, He XL, Chu YK, Lu JG, Ma QJ. Role of nuclear factor kappa B and reactive oxygen species in the tumor necrosis factor-alpha-induced epithelial–mesenchymal transition of MCF-7 cells. Brazilian Journal of Medical and Biological Research. 2007;40:1071–1078. doi: 10.1590/s0100-879x2007000800007. [DOI] [PubMed] [Google Scholar]
  • 50.Chuang MJ, Sun KH, Tang SJ, Deng MW, Wu YH, Sung JS, et al. Tumor-derived tumor necrosis factor-alpha promotes progression and epithelial–mesenchymal transition in renal cell carcinoma cells. Cancer Science. 2008;99:905–913. doi: 10.1111/j.1349-7006.2008.00756.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Grund EM, Kagan D, Tran CA, Zeitvogel A, Starzinski-Powitz A, Nataraja S, et al. Tumor necrosis factor-alpha regulates inflammatory and mesenchymal responses via mitogen-activated protein kinase kinase, p38, and nuclear factor kappaB in human endometriotic epithelial cells. Molecular Pharmacology. 2008;73:1394–1404. doi: 10.1124/mol.107.042176. [DOI] [PubMed] [Google Scholar]
  • 52.Wu Y, Deng J, Rychahou PG, Qiu S, Evers BM, Zhou BP. Stabilization of snail by NF-kappaB is required for inflammation-induced cell migration and invasion. Cancer Cell. 2009;15:416–428. doi: 10.1016/j.ccr.2009.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Savagner P, Valles AM, Jouanneau J, Yamada KM, Thiery JP. Alternative splicing in fibroblast growth factor receptor 2 is associated with induced epithelial–mesenchymal transition in rat bladder carcinoma cells. Molecular Biology of the Cell. 1994;5:851–862. doi: 10.1091/mbc.5.8.851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Jiang WG, Watkins G, Fodstad O, Douglas-Jones A, Mokbel K, Mansel RE. Differential expression of the CCN family members Cyr61, CTGF and Nov in human breast cancer. Endocrine Related Cancer. 2004;11:781–791. doi: 10.1677/erc.1.00825. [DOI] [PubMed] [Google Scholar]
  • 55.Zhang Y, Pan Q, Zhong H, Merajver SD, Kleer CG. Inhibition of CCN6 (WISP3) expression promotes neoplastic progression and enhances the effects of insulin-like growth factor-1 on breast epithelial cells. Breast Cancer Research. 2005;7:R1080–R1089. doi: 10.1186/bcr1351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Dhar G, Mehta S, Banerjee S, Gardner A, McCarty BM, Mathur SC, et al. Loss of WISP-2/CCN5 signaling in human pancreatic cancer: a potential mechanism for epithelial–mesenchymal-transition. Cancer Letters. 2007;254:63–70. doi: 10.1016/j.canlet.2007.02.012. [DOI] [PubMed] [Google Scholar]
  • 57.Davies SR, Davies ML, Sanders A, Parr C, Torkington J, Jiang WG. Differential expression of the CCN family member WISP-1, WISP-2 and WISP-3 in human colorectal cancer and the prognostic implications. International Journal of Oncology. 2010;36:1129–1136. doi: 10.3892/ijo_00000595. [DOI] [PubMed] [Google Scholar]
  • 58.Haque I, Mehta S, Majumder M, Dhar K, De A, McGregor D, et al. Cyr61/CCN1 signaling is critical for epithelial–mesenchymal transition and stemness and promotes pancreatic carcinogenesis. Molecular Cancer. 2011;10:8. doi: 10.1186/1476-4598-10-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Turchi L, Chassot AA, Rezzonico R, Yeow K, Loubat A, Ferrua B, et al. Dynamic characterization of the molecular events during in vitro epidermal wound healing. Journal of Investigative Dermatology. 2002;119:56–63. doi: 10.1046/j.1523-1747.2002.01805.x. [DOI] [PubMed] [Google Scholar]
  • 60.Tscharntke M, Pofahl R, Krieg T, Haase I. Ras-induced spreading and wound closure in human epidermal keratinocytes. FASEB Journal. 2005;19:1836–1838. doi: 10.1096/fj.04-3327fje. [DOI] [PubMed] [Google Scholar]
  • 61.Squarize CH, Castilho RM, Bugge TH, Gutkind JS. Accelerated wound healing by mTOR activation in genetically defined mouse models. PLoS One. 2010;5:e10643. doi: 10.1371/journal.pone.0010643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Koivisto L, Alavian K, Hakkinen L, Pelech S, McCulloch CA, Larjava H. Glycogen synthase kinase-3 regulates formation of long lamellipodia in human keratinocytes. Journal of Cell Science. 2003;116:3749–3760. doi: 10.1242/jcs.00693. [DOI] [PubMed] [Google Scholar]
  • 63.Koivisto L, Jiang G, Hakkinen L, Chan B, Larjava H. HaCaT keratinocyte migration is dependent on epidermal growth factor receptor signaling and glycogen synthase kinase-3alpha. Experimental Cell Research. 2006;312:2791–2805. doi: 10.1016/j.yexcr.2006.05.009. [DOI] [PubMed] [Google Scholar]
  • 64.Santos MF, McCormack SA, Guo Z, Okolicany J, Zheng Y, Johnson LR, et al. Rho proteins play a critical role in cell migration during the early phase of mucosal restitution. Journal of Clinical Investigation. 1997;100:216–225. doi: 10.1172/JCI119515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Haase I, Evans R, Pofahl R, Watt FM. Regulation of keratinocyte shape, migration and wound epithelialization by IGF-1- and EGF-dependent signalling pathways. Journal of Cell Science. 2003;116:3227–3238. doi: 10.1242/jcs.00610. [DOI] [PubMed] [Google Scholar]
  • 66.Jackson B, Peyrollier K, Pedersen E, Basse A, Karlsson R, Wang Z, et al. RhoA is dispensable for skin development, but crucial for contraction and directed migration of keratinocytes. Molecular Biology of the Cell. 2011;22:593–605. doi: 10.1091/mbc.E09-10-0859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Janda E, Lehmann K, Killisch I, Jechlinger M, Herzig M, Downward J, et al. Ras and TGF[beta] cooperatively regulate epithelial cell plasticity and metastasis: dissection of Ras signaling pathways. Journal of Cell Biology. 2002;156:299–313. doi: 10.1083/jcb.200109037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Janda E, Litos G, Grunert S, Downward J, Beug H. Oncogenic Ras/Her-2 mediate hyperproliferation of polarized epithelial cells in 3D cultures and rapid tumor growth via the PI3K pathway. Oncogene. 2002;21:5148–5159. doi: 10.1038/sj.onc.1205661. [DOI] [PubMed] [Google Scholar]
  • 69.Keniry M, Parsons R. The role of PTEN signaling perturbations in cancer and in targeted therapy. Oncogene. 2008;27:5477–5485. doi: 10.1038/onc.2008.248. [DOI] [PubMed] [Google Scholar]
  • 70.Wu T, Mohan C. The AKT axis as a therapeutic target in autoimmune diseases. Endocrine, Metabolic & Immune Disorders Drug Targets. 2009;9:145–150. doi: 10.2174/187153009788452417. [DOI] [PubMed] [Google Scholar]
  • 71.Mehta PB, Jenkins BL, McCarthy L, Thilak L, Robson CN, Neal DE, et al. MEK5 overexpression is associated with metastatic prostate cancer, and stimulates proliferation, MMP-9 expression and invasion. Oncogene. 2003;22:1381–1389. doi: 10.1038/sj.onc.1206154. [DOI] [PubMed] [Google Scholar]
  • 72.Castro NE, Lange CA. Breast tumor kinase and extracellular signal-regulated kinase 5 mediate Met receptor signaling to cell migration in breast cancer cells. Breast Cancer Research. 2010;12:R60. doi: 10.1186/bcr2622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Ramsay AK, McCracken SR, Soofi M, Fleming J, Yu AX, Ahmad I, et al. ERK5 signalling in prostate cancer promotes an invasive phenotype. British Journal of Cancer. 2011;104:664–672. doi: 10.1038/sj.bjc.6606062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Bhowmick NA, Ghiassi M, Bakin A, Aakre M, Lundquist CA, Engel ME, et al. Transforming growth factor-beta1 mediates epithelial to mesenchymal trans-differentiation through a RhoA-dependent mechanism. Molecular Biology of the Cell. 2001;12:27–36. doi: 10.1091/mbc.12.1.27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Bachelder RE, Yoon SO, Franci C, de Herreros AG, Mercurio AM. Glycogen synthase kinase-3 is an endogenous inhibitor of Snail transcription: implications for the epithelial–mesenchymal transition. Journal of Cell Biology. 2005;168:29–33. doi: 10.1083/jcb.200409067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Ma C, Wang J, Gao Y, Gao TW, Chen G, Bower KA, et al. The role of glycogen synthase kinase 3beta in the transformation of epidermal cells. Cancer Research. 2007;67:7756–7764. doi: 10.1158/0008-5472.CAN-06-4665. [DOI] [PubMed] [Google Scholar]
  • 77.Ko H, Kim HS, Kim NH, Lee SH, Kim KH, Hong SH, et al. Nuclear localization signals of the E-cadherin transcriptional repressor Snail. Cells Tissues Organs. 2007;185:66–72. doi: 10.1159/000101305. [DOI] [PubMed] [Google Scholar]
  • 78.Vincent T, Kukalev A, Andang M, Pettersson R, Percipalle P. The glycogen synthase kinase (GSK) 3beta represses RNA polymerase I transcription. Oncogene. 2008;27:5254–5259. doi: 10.1038/onc.2008.152. [DOI] [PubMed] [Google Scholar]
  • 79.Nelson WJ, Nusse R. Convergence of Wnt, beta-catenin, and cadherin pathways. Science. 2004;303:1483–1487. doi: 10.1126/science.1094291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Cheon SS, Wei Q, Gurung A, Youn A, Bright T, Poon R, et al. Beta-catenin regulates wound size and mediates the effect of TGF-beta in cutaneous healing. FASEB Journal. 2006;20:692–701. doi: 10.1096/fj.05-4759com. [DOI] [PubMed] [Google Scholar]
  • 81.Stoll SW, Rittie L, Johnson JL, Elder JT. Heparin-binding EGF-like growth factor promotes epithelial–mesenchymal transition in human keratinocytes. Journal of Investigative Dermatology. 2012 doi: 10.1038/jid.2012.78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Eger A, Stockinger A, Schaffhauser B, Beug H, Foisner R. Epithelial mesenchymal transition by c-Fos estrogen receptor activation involves nuclear translocation of beta-catenin and upregulation of beta-catenin/lymphoid enhancer binding factor-1 transcriptional activity. Journal of Cell Biology. 2000;148:173–188. doi: 10.1083/jcb.148.1.173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Kim HJ, Roux A, Sonenshein AL. Direct and indirect roles of CcpA in regulation of Bacillus subtilis Krebs cycle genes. Molecular Microbiology. 2002;45:179–190. doi: 10.1046/j.1365-2958.2002.03003.x. [DOI] [PubMed] [Google Scholar]
  • 84.Shirley SH, Hudson LG, He J, Kusewitt DF. The skinny on Slug. Molecular Carcinogenesis. 2010;49:851–861. doi: 10.1002/mc.20674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Sou PW, Delic NC, Halliday GM, Lyons JG. Snail transcription factors in keratinocytes: enough to make your skin crawl. The International Journal of Biochemistry & Cell Biology. 2010;42:1940–1944. doi: 10.1016/j.biocel.2010.08.021. [DOI] [PubMed] [Google Scholar]
  • 86.Savagner P, Yamada KM, Thiery JP. The zinc-finger protein slug causes desmosome dissociation, an initial and necessary step for growth factor-induced epithelial–mesenchymal transition. Journal of Cell Biology. 1997;137:1403–1419. doi: 10.1083/jcb.137.6.1403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Savagner P, Kusewitt DF, Carver EA, Magnino F, Choi C, Gridley T, et al. Developmental transcription factor slug is required for effective re-epithelialization by adult keratinocytes. Journal of Cellular Physiology. 2005;202:858–866. doi: 10.1002/jcp.20188. [DOI] [PubMed] [Google Scholar]
  • 88.Kusewitt DF, Choi C, Newkirk KM, Leroy P, Li Y, Chavez MG, et al. Slug/Snai2 is a downstream mediator of epidermal growth factor receptor-stimulated reepithelialization. Journal of Investigative Dermatology. 2009;129:491–495. doi: 10.1038/jid.2008.222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Hudson LG, Newkirk KM, Chandler HL, Choi C, Fossey SL, Parent AE, et al. Cutaneous wound reepithelialization is compromised in mice lacking functional Slug (Snai2) Journal of Dermatological Science. 2009;56:19–26. doi: 10.1016/j.jdermsci.2009.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Cano A, Perez-Moreno MA, Rodrigo I, Locascio A, Blanco MJ, del Barrio MG, et al. The transcription factor snail controls epithelial–mesenchymal transitions by repressing E-cadherin expression. Nature Cell Biology. 2000;2:76–83. doi: 10.1038/35000025. [DOI] [PubMed] [Google Scholar]
  • 91.Guaita S, Puig I, Franci C, Garrido M, Dominguez D, Batlle E, et al. Snail induction of epithelial to mesenchymal transition in tumor cells is accompanied by MUC1 repression and ZEB1 expression. Journal of Biological Chemistry. 2002;277:39209–39216. doi: 10.1074/jbc.M206400200. [DOI] [PubMed] [Google Scholar]
  • 92.Olmeda D, Jorda M, Peinado H, Fabra A, Cano A. Snail silencing effectively suppresses tumour growth and invasiveness. Oncogene. 2007;26:1862–1874. doi: 10.1038/sj.onc.1209997. [DOI] [PubMed] [Google Scholar]
  • 93.Medici D, Hay ED, Olsen BR. Snail and Slug promote epithelial–mesenchymal transition through beta-catenin-T-cell factor-4-dependent expression of transforming growth factor-beta3. Molecular Biology of the Cell. 2008;19:4875–4887. doi: 10.1091/mbc.E08-05-0506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Vincent T, Neve EP, Johnson JR, Kukalev A, Rojo F, Albanell J, et al. A SNAIL1-SMAD3/4 transcriptional repressor complex promotes TGF-beta mediated epithelial–mesenchymal transition. Nature Cell Biology. 2009;11:943–950. doi: 10.1038/ncb1905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Massague J, Seoane J, Wotton D. Smad transcription factors. Genes and Development. 2005;19:2783–2810. doi: 10.1101/gad.1350705. [DOI] [PubMed] [Google Scholar]
  • 96.Heldin CH, Moustakas A. Role of Smads in TGFbeta signaling. Cell and Tissue Research. 2012;347:21–36. doi: 10.1007/s00441-011-1190-x. [DOI] [PubMed] [Google Scholar]
  • 97.Ashcroft GS, Roberts AB. Loss of Smad3 modulates wound healing. Cytokine and Growth Factor Reviews. 2000;11:125–131. doi: 10.1016/s1359-6101(99)00036-2. [DOI] [PubMed] [Google Scholar]
  • 98.Owens P, Engelking E, Han G, Haeger SM, Wang XJ. Epidermal Smad4 deletion results in aberrant wound healing. American Journal of Pathology. 2010;176:122–133. doi: 10.2353/ajpath.2010.090081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Piek E, Moustakas A, Kurisaki A, Heldin CH, ten Dijke P. TGF-(beta) type I receptor/ALK-5 and Smad proteins mediate epithelial to mesenchymal transdifferentiation in NMuMG breast epithelial cells. Journal of Cell Science. 1999;112:4557–4568. doi: 10.1242/jcs.112.24.4557. [DOI] [PubMed] [Google Scholar]
  • 100.Eger A, Stockinger A, Park J, Langkopf E, Mikula M, Gotzmann J, et al. beta-Catenin and TGFbeta signalling cooperate to maintain a mesenchymal phenotype after FosER-induced epithelial to mesenchymal transition. Oncogene. 2004;23:2672–2680. doi: 10.1038/sj.onc.1207416. [DOI] [PubMed] [Google Scholar]
  • 101.Rosivatz E, Becker I, Specht K, Fricke E, Luber B, Busch R, et al. Differential expression of the epithelial–mesenchymal transition regulators snail, SIP1, and twist in gastric cancer. American Journal of Pathology. 2002 Nov;161:1881–1891. doi: 10.1016/S0002-9440(10)64464-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Yang J, Mani SA, Donaher JL, Ramaswamy S, Itzykson RA, Come C, et al. Twist a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell. 2004;117:927–939. doi: 10.1016/j.cell.2004.06.006. [DOI] [PubMed] [Google Scholar]
  • 103.Reichert M, Müller T, Hunziker W. The PDZ domains of zonula occludens-1 induce an epithelial to mesenchymal transition of Madin-Darby canine kidney I cells. Evidence for a role of beta-catenin/Tcf/Lef signaling. The Journal of Biological Chemistry. 2000;275:9492–9500. doi: 10.1074/jbc.275.13.9492. [DOI] [PubMed] [Google Scholar]
  • 104.Gilles C, Polette M, Birembaut P, Brunner N, Thompson EW. Expression of c-ets-1 mRNA is associated with an invasive, EMT-derived phenotype in breast carcinoma cell lines. Clinical and Experimental Metastasis. 1997;15:519–526. doi: 10.1023/a:1018427027270. [DOI] [PubMed] [Google Scholar]
  • 105.Taki M, Verschueren K, Yokoyama K, Nagayama M, Kamata N. Involvement of Ets-1 transcription factor in inducing matrix metalloproteinase-2 expression by epithelial–mesenchymal transition in human squamous carcinoma cells. International Journal of Oncology. 2006;28:487–496. [PubMed] [Google Scholar]
  • 106.Chua HL, Bhat-Nakshatri P, Clare SE, Morimiya A, Badve S, Nakshatri H. NF-kappaB represses E-cadherin expression and enhances epithelial to mesenchymal transition of mammary epithelial cells: potential involvement of ZEB-1 and ZEB-2. Oncogene. 2007;26(5):711–724. doi: 10.1038/sj.onc.1209808. [DOI] [PubMed] [Google Scholar]
  • 107.Mani SA, Yang J, Brooks M, Schwaninger G, Zhou A, Miura N, et al. Mesenchyme Forkhead 1 (FOXC2) plays a key role in metastasis and is associated with aggressive basal-like breast cancers. Proceedings of the National Academy of Sciences of the United States of America. 2007;104:10069–10074. doi: 10.1073/pnas.0703900104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Terao M, Ishikawa A, Nakahara S, Kimura A, Kato A, Moriwaki K, et al. Enhanced epithelial–mesenchymal transition-like phenotype in N-acetylglucosaminyltransferase V transgenic mouse skin promotes wound healing. Journal of Biological Chemistry. 2011;286:28303–28311. doi: 10.1074/jbc.M111.220376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Beuerman RW, Thompson HW. Molecular and cellular responses of the corneal epithelium to wound healing. Acta Ophthalmologica Supplement. 1992:7–12. doi: 10.1111/j.1755-3768.1992.tb02161.x. [DOI] [PubMed] [Google Scholar]
  • 110.Martin P, Nobes CD. An early molecular component of the wound healing response in rat embryos—induction of c-fos protein in cells at the epidermal wound margin. Mechanisms of Development. 1992;38:209–215. doi: 10.1016/0925-4773(92)90054-n. [DOI] [PubMed] [Google Scholar]
  • 111.Mejlvang J, Kriajevska M, Berditchevski F, Bronstein I, Lukanidin EM, Pringle JH, et al. Characterization of E-cadherin-dependent and -independent events in a new model of c-Fos-mediated epithelial–mesenchymal transition. Experimental Cell Research. 2007;313:380–393. doi: 10.1016/j.yexcr.2006.10.017. [DOI] [PubMed] [Google Scholar]
  • 112.Bavan L, Midwood K, Nanchahal J. MicroRNA epigenetics: a new avenue for wound healing research. BioDrugs. 2011;25:27–41. doi: 10.2165/11585010-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 113.Schneider MR. MicroRNAs as novel players in skin development, homeostasis and disease. British Journal of Dermatology. 2012;166:22–28. doi: 10.1111/j.1365-2133.2011.10568.x. [DOI] [PubMed] [Google Scholar]
  • 114.Dykxhoorn DM. MicroRNAs and metastasis: little RNAs go a long way. Cancer Research. 2010;70:6401–6406. doi: 10.1158/0008-5472.CAN-10-1346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Wang Z, Li Y, Ahmad A, Azmi AS, Kong D, Banerjee S, et al. Targeting miRNAs involved in cancer stem cell and EMT regulation: an emerging concept in overcoming drug resistance. Drug Resistance Updates. 2010;13:109–118. doi: 10.1016/j.drup.2010.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Bullock MD, Sayan AE, Packham GK, Mirnezami AH. MicroRNAs: critical regulators of epithelial to mesenchymal (EMT) and mesenchymal to epithelial transition (MET) in cancer progression. Biologie Cellulaire. 2012;104:3–12. doi: 10.1111/boc.201100115. [DOI] [PubMed] [Google Scholar]
  • 117.Goliger JA, Paul DL. Wounding alters epidermal connexin expression and gap junction-mediated intercellular communication. Molecular Biology of the Cell. 1995;6:1491–1501. doi: 10.1091/mbc.6.11.1491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.McLachlan E, Shao Q, Wang HL, Langlois S, Laird DW. Connexins act as tumor suppressors in three-dimensional mammary cell organoids by regulating differentiation and angiogenesis. Cancer Research. 2006;66:9886–9894. doi: 10.1158/0008-5472.CAN-05-4302. [DOI] [PubMed] [Google Scholar]
  • 119.Moll I, Houdek P, Schafer S, Nuber U, Moll R. Diversity of desmosomal proteins in regenerating epidermis: immunohistochemical study using a human skin organ culture model. Archives for Dermatological Research. 1999;291:437–446. doi: 10.1007/s004030050435. [DOI] [PubMed] [Google Scholar]
  • 120.Boyer B, Tucker GC, Valles AM, Franke WW, Thiery JP. Rearrangements of desmosomal and cytoskeletal proteins during the transition from epithelial to fibroblastoid organization in cultured rat bladder carcinoma cells. Journal of Cell Biology. 1989;109:1495–1509. doi: 10.1083/jcb.109.4.1495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Baumgart E, Cohen MS, Silva Neto B, Jacobs MA, Wotkowicz C, Rieger-Christ KM, et al. Identification and prognostic significance of an epithelial–mesenchymal transition expression profile in human bladder tumors. Clinical Cancer Research. 2007;13:1685–1694. doi: 10.1158/1078-0432.CCR-06-2330. [DOI] [PubMed] [Google Scholar]
  • 122.Mainiero F, Pepe A, Yeon M, Ren Y, Giancotti FG. The intracellular functions of alpha6beta4 integrin are regulated by EGF. Journal of Cell Biology. 1996;134:241–253. doi: 10.1083/jcb.134.1.241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Margadant C, Frijns E, Wilhelmsen K, Sonnenberg A. Regulation of hemidesmosome disassembly by growth factor receptors. Current Opinion in Cell Biology. 2008;20:589–596. doi: 10.1016/j.ceb.2008.05.001. [DOI] [PubMed] [Google Scholar]
  • 124.Maschler S, Wirl G, Spring H, Bredow DV, Sordat I, Beug H, et al. Tumor cell invasiveness correlates with changes in integrin expression and localization. Oncogene. 2005;24:2032–2041. doi: 10.1038/sj.onc.1208423. [DOI] [PubMed] [Google Scholar]
  • 125.Hirohashi S. Inactivation of the E-cadherin-mediated cell adhesion system in human cancers. American Journal of Pathology. 1998;153:333–339. doi: 10.1016/S0002-9440(10)65575-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Bakin AV, Tomlinson AK, Bhowmick NA, Moses HL, Arteaga CL. Phosphatidylinositol 3-kinase function is required for transforming growth factor beta-mediated epithelial to mesenchymal transition and cell migration. Journal of Biological Chemistry. 2000;275:36803–36810. doi: 10.1074/jbc.M005912200. [DOI] [PubMed] [Google Scholar]
  • 127.Hajra KM, Chen DY, Fearon ER. The SLUG zinc-finger protein represses E-cadherin in breast cancer. Cancer Research. 2002;62:1613–1618. [PubMed] [Google Scholar]
  • 128.Blanco MJ, Moreno-Bueno G, Sarrio D, Locascio A, Cano A, Palacios J, et al. Correlation of Snail expression with histological grade and lymph node status in breast carcinomas. Oncogene. 2002;21:3241–3246. doi: 10.1038/sj.onc.1205416. [DOI] [PubMed] [Google Scholar]
  • 129.Islam S, Carey TE, Wolf GT, Wheelock MJ, Johnson KR. Expression of N-cadherin by human squamous carcinoma cells induces a scattered fibroblastic phenotype with disrupted cell–cell adhesion. Journal of Cell Biology. 1996;135:1643–1654. doi: 10.1083/jcb.135.6.1643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Yang Z, Zhang X, Gang H, Li X, Li Z, Wang T, et al. Up-regulation of gastric cancer cell invasion by Twist is accompanied by N-cadherin and fibronectin expression. Biochemical and Biophysical Research Communications. 2007;358:925–930. doi: 10.1016/j.bbrc.2007.05.023. [DOI] [PubMed] [Google Scholar]
  • 131.Brandner JM, Kief S, Grund C, Rendl M, Houdek P, Kuhn C, et al. Organization and formation of the tight junction system in human epidermis and cultured keratinocytes. European Journal of Cell Biology. 2002;81:253–263. doi: 10.1078/0171-9335-00244. [DOI] [PubMed] [Google Scholar]
  • 132.Witherden DA, Verdino P, Rieder SE, Garijo O, Mills RE, Teyton L, et al. The junctional adhesion molecule JAML is a costimulatory receptor for epithelial gammadelta T cell activation. Science. 2010;329:1205–1210. doi: 10.1126/science.1192698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Dhawan P, Singh AB, Deane NG, No Y, Shiou SR, Schmidt C, et al. Claudin-1 regulates cellular transformation and metastatic behavior in colon cancer. Journal of Clinical Investigation. 2005;115:1765–1776. doi: 10.1172/JCI24543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Martinez-Estrada OM, Culleres A, Soriano FX, Peinado H, Bolos V, Martinez FO, et al. The transcription factors Slug and Snail act as repressors of Claudin-1 expression in epithelial cells. Biochemical Journal. 2006;394:449–457. doi: 10.1042/BJ20050591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Medici D, Hay ED, Goodenough DA. Cooperation between snail and LEF-1 transcription factors is essential for TGF-beta1-induced epithelial–mesenchymal transition. Molecular Biology of the Cell. 2006;17:1871–1879. doi: 10.1091/mbc.E05-08-0767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Clark RA, Ashcroft GS, Spencer MJ, Larjava H, Ferguson MW. Reepithelialization of normal human excisional wounds is associated with a switch from alpha v beta 5 to alpha v beta 6 integrins. British Journal of Dermatology. 1996;135:46–51. [PubMed] [Google Scholar]
  • 137.Haapasalmi K, Zhang K, Tonnesen M, Olerud J, Sheppard D, Salo T, et al. Keratinocytes in human wounds express alpha v beta 6 integrin. Journal of Investigative Dermatology. 1996;106:42–48. doi: 10.1111/1523-1747.ep12327199. [DOI] [PubMed] [Google Scholar]
  • 138.Tse JC, Kalluri R. Mechanisms of metastasis: epithelial-to-mesenchymal transition and contribution of tumor microenvironment. Journal of Cellular Biochemistry. 2007;101:816–829. doi: 10.1002/jcb.21215. [DOI] [PubMed] [Google Scholar]
  • 139.Okada A, Tomasetto C, Lutz Y, Bellocq JP, Rio MC, Basset P. Expression of matrix metalloproteinases during rat skin wound healing: evidence that membrane type-1 matrix metalloproteinase is a stromal activator of pro-gelatinase A. Journal of Cell Biology. 1997;137:67–77. doi: 10.1083/jcb.137.1.67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Madlener M, Parks WC, Werner S. Matrix metalloproteinases (MMPs) and their physiological inhibitors (TIMPs) are differentially expressed during excisional skin wound repair. Experimental Cell Research. 1998;242:201–210. doi: 10.1006/excr.1998.4049. [DOI] [PubMed] [Google Scholar]
  • 141.Mohan R, Chintala SK, Jung JC, Villar WV, McCabe F, Russo LA, et al. Matrix metalloproteinase gelatinase B (MMP-9) coordinates and effects epithelial regeneration. Journal of Biological Chemistry. 2002;277:2065–2072. doi: 10.1074/jbc.M107611200. [DOI] [PubMed] [Google Scholar]
  • 142.Seomun Y, Kim JT, Joo CK. MMP-14 mediated MMP-9 expression is involved in TGF-beta1-induced keratinocyte migration. Journal of Cellular Biochemistry. 2008;104:934–941. doi: 10.1002/jcb.21675. [DOI] [PubMed] [Google Scholar]
  • 143.Kyriakides TR, Wulsin D, Skokos EA, Fleckman P, Pirrone A, Shipley JM, et al. Mice that lack matrix metalloproteinase-9 display delayed wound healing associated with delayed reepithelization and disordered collagen fibrillogenesis. Matrix Biology. 2009;28:65–73. doi: 10.1016/j.matbio.2009.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Hattori N, Mochizuki S, Kishi K, Nakajima T, Takaishi H, D’Armiento J, et al. MMP-13 plays a role in keratinocyte migration, angiogenesis, and contraction in mouse skin wound healing. American Journal of Pathology. 2009;175:533–546. doi: 10.2353/ajpath.2009.081080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Gilles C, Polette M, Piette J, Birembaut P, Foidart JM. Epithelial-to-mesenchymal transition in HPV-33-transfected cervical keratinocytes is associated with increased invasiveness and expression of gelatinase A. International Journal of Cancer. 1994;59:661–666. doi: 10.1002/ijc.2910590514. [DOI] [PubMed] [Google Scholar]
  • 146.Pulyaeva H, Bueno J, Polette M, Birembaut P, Sato H, Seiki M, et al. MT1-MMP correlates with MMP-2 activation potential seen after epithelial to mesenchymal transition in human breast carcinoma cells. Clinical & Experimental Metastasis. 1997;15:111–120. doi: 10.1023/a:1018444609098. [DOI] [PubMed] [Google Scholar]
  • 147.Gilles C, Polette M, Seiki M, Birembaut P, Thompson EW. Implication of collagen type I-induced membrane-type 1-matrix metalloproteinase expression and matrix metalloproteinase-2 activation in the metastatic progression of breast carcinoma. Laboratory Investigation. 1997;76:651–660. [PubMed] [Google Scholar]
  • 148.Martorana AM, Zheng G, Crowe TC, O’Grady RL, Lyons JG. Epithelial cells up-regulate matrix metalloproteinases in cells within the same mammary carcinoma that have undergone an epithelial–mesenchymal transition. Cancer Research. 1998;58:4970–4979. [PubMed] [Google Scholar]
  • 149.Tester AM, Ruangpanit N, Anderson RL, Thompson EW. MMP-9 secretion and MMP-2 activation distinguish invasive and metastatic sublines of a mouse mammary carcinoma system showing epithelial–mesenchymal transition traits. Clinical and Experimental Metastasis. 2000;18:553–560. doi: 10.1023/a:1011953118186. [DOI] [PubMed] [Google Scholar]
  • 150.Billottet C, Tuefferd M, Gentien D, Rapinat A, Thiery JP, Broet P, et al. Modulation of several waves of gene expression during FGF-1 induced epithelial–mesenchymal transition of carcinoma cells. Journal of Cellular Biochemistry. 2008;104:826–839. doi: 10.1002/jcb.21667. [DOI] [PubMed] [Google Scholar]
  • 151.Pilcher BK, Dumin JA, Sudbeck BD, Krane SM, Welgus HG, Parks WC. The activity of collagenase-1 is required for keratinocyte migration on a type I collagen matrix. Journal of Cell Biology. 1997;137:1445–1457. doi: 10.1083/jcb.137.6.1445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Giannelli G, Falk-Marzillier J, Schiraldi O, Stetler-Stevenson WG, Quaranta V. Induction of cell migration by matrix metalloprotease-2 cleavage of laminin-5. Science. 1997;277:225–228. doi: 10.1126/science.277.5323.225. [DOI] [PubMed] [Google Scholar]
  • 153.Bullard KM, Lund L, Mudgett JS, Mellin TN, Hunt TK, Murphy B, et al. Impaired wound contraction in stromelysin-1-deficient mice. Annals of Surgery. 1999;230:260–265. doi: 10.1097/00000658-199908000-00017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Wilkins-Port CE, Ye Q, Mazurkiewicz JE, Higgins PJ. TGF-beta1 + EGF-initiated invasive potential in transformed human keratinocytes is coupled to a plasmin/MMP-10/MMP-1-dependent collagen remodeling axis: role for PAI-1. Cancer Research. 2009;69:4081–4091. doi: 10.1158/0008-5472.CAN-09-0043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Zhao M, Szafranski P, Hall CA, Goode S. Basolateral junctions utilize warts signaling to control epithelial–mesenchymal transition and proliferation crucial for migration and invasion of Drosophila ovarian epithelial cells. Genetics. 2008;178:1947–1971. doi: 10.1534/genetics.108.086983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Dow LE, Kauffman JS, Caddy J, Zarbalis K, Peterson AS, Jane SM, et al. The tumour-suppressor Scribble dictates cell polarity during directed epithelial migration: regulation of Rho GTPase recruitment to the leading edge. Oncogene. 2007;26:2272–2282. doi: 10.1038/sj.onc.1210016. [DOI] [PubMed] [Google Scholar]
  • 157.Brabletz S, Brabletz T. The ZEB/miR-200 feedback loop—a motor of cellular plasticity in development and cancer. EMBO Reports. 2010;11:670–677. doi: 10.1038/embor.2010.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Tomita K, van Bokhoven A, van Leenders GJ, Ruijter ET, Jansen CF, Bussemakers MJ, et al. Cadherin switching in human prostate cancer progression. Cancer Research. 2000;60:3650–3654. [PubMed] [Google Scholar]
  • 159.Derleth C, Mayer IA. Antiangiogenic therapies in early-stage breast cancer. Clinical Breast Cancer. 2010;10(Suppl. 1):E23–E31. doi: 10.3816/CBC.2010.s.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Hompes D, Ruers T. Review: incidence and clinical significance of Bevacizumab-related non-surgical and surgical serious adverse events in metastatic colorectal cancer. European Journal of Surgical Oncology. 2011;37:737–746. doi: 10.1016/j.ejso.2011.06.004. [DOI] [PubMed] [Google Scholar]
  • 161.Myskowski PL, Halpern AC. Cutaneous adverse reactions to therapeutic monoclonal antibodies for cancer. Current Allergy and Asthma Reports. 2008;8:63–68. doi: 10.1007/s11882-008-0012-z. [DOI] [PubMed] [Google Scholar]
  • 162.Myskowski PL, Halpern AC. Skin reactions to the new biologic anticancer drugs. Current Opinion in Supportive and Palliative Care. 2009;3:294–299. doi: 10.1097/SPC.0b013e328332d397. [DOI] [PubMed] [Google Scholar]
  • 163.Burtness B, Anadkat M, Basti S, Hughes M, Lacouture ME, McClure JS, et al. NCCN task force report: management of dermatologic and other toxicities associated with EGFR inhibition in patients with cancer. Journal of the National Comprehensive Cancer Network. 2009;7(Suppl. 1):S5–S21. doi: 10.6004/jnccn.2009.0074. quiz S22–24. [DOI] [PubMed] [Google Scholar]
  • 164.Nakamura Y, Sotozono C, Kinoshita S. The epidermal growth factor receptor (EGFR): role in corneal wound healing and homeostasis. Experimental Eye Research. 2001;72:511–517. doi: 10.1006/exer.2000.0979. [DOI] [PubMed] [Google Scholar]
  • 165.Kaftan H, Vogelgesang S, Lempas K, Hosemann W, Herzog M. Inhibition of epidermal growth factor receptor by erlotinib: wound healing of experimental tympanic membrane perforations. Otology and Neurotology. 2007;28:245–249. doi: 10.1097/01.mao.0000244366.24449.db. [DOI] [PubMed] [Google Scholar]
  • 166.Kaftan H, Reuther L, Miehe B, Hosemann W, Herzog M. Delay of tympanic membrane wound healing in rats with topical application of a tyrosine kinase inhibitor. Wound Repair and Regeneration. 2008;16:364–369. doi: 10.1111/j.1524-475X.2008.00375.x. [DOI] [PubMed] [Google Scholar]
  • 167.Kaftan H, Reuther L, Miehe B, Hosemann W, Herzog M. The influence of inhibition of the epidermal growth factor receptor on tympanic membrane wound healing in rats. Growth Factors. 2010;28:286–292. doi: 10.3109/08977191003620238. [DOI] [PubMed] [Google Scholar]
  • 168.Quintás-Cardama A, Cortes J. Chronic myeloid leukemia in the tyrosine kinase inhibitor era: what is the best therapy? Current Oncology Reports. 2009;11:337–345. doi: 10.1007/s11912-009-0046-y. [DOI] [PubMed] [Google Scholar]
  • 169.Robert C, Soria JC, Spatz A, Le Cesne A, Malka D, Pautier P, et al. Cutaneous side-effects of kinase inhibitors and blocking antibodies. Lancet Oncology. 2005;6:491–500. doi: 10.1016/S1470-2045(05)70243-6. [DOI] [PubMed] [Google Scholar]
  • 170.Rajkumar VS, Shiwen X, Bostrom M, Leoni P, Muddle J, Ivarsson M, et al. Platelet-derived growth factor-beta receptor activation is essential for fibroblast and pericyte recruitment during cutaneous wound healing. American Journal of Pathology. 2006;169:2254–2265. doi: 10.2353/ajpath.2006.060196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Rosano L, Spinella F, Di Castro V, Nicotra MR, Dedhar S, de Herreros AG, et al. Endothelin-1 promotes epithelial-to-mesenchymal transition in human ovarian cancer cells. Cancer Research. 2005;65:11649–11657. doi: 10.1158/0008-5472.CAN-05-2123. [DOI] [PubMed] [Google Scholar]
  • 172.Rosano L, Cianfrocca R, Spinella F, Di Castro V, Nicotra MR, Lucidi A, et al. Acquisition of chemoresistance and EMT phenotype is linked with activation of the endothelin A receptor pathway in ovarian carcinoma cells. Clinical Cancer Research. 2011;17:2350–2360. doi: 10.1158/1078-0432.CCR-10-2325. [DOI] [PubMed] [Google Scholar]
  • 173.Adjei AA, Schwartz B, Garmey E. Early clinical development of ARQ 197, a selective, non-ATP-competitive inhibitor targeting MET tyrosine kinase for the treatment of advanced cancers. Oncologist. 2011;16:788–799. doi: 10.1634/theoncologist.2010-0380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Rosen LS, Senzer N, Mekhail T, Ganapathi R, Chai F, Savage RE, et al. A phase I dose-escalation study of Tivantinib (ARQ 197) in adult patients with metastatic solid tumors. Clinical Cancer Research. 2011;17:7754–7764. doi: 10.1158/1078-0432.CCR-11-1002. [DOI] [PubMed] [Google Scholar]
  • 175.Wieman TJ, Smiell JM, Su Y. Efficacy and safety of a topical gel formulation of recombinant human platelet-derived growth factor-BB (becaplermin) in patients with chronic neuropathic diabetic ulcers. A phase III randomized placebo-controlled double-blind study. Diabetes Care. 1998;21:822–827. doi: 10.2337/diacare.21.5.822. [DOI] [PubMed] [Google Scholar]
  • 176.Smiell JM. Clinical safety of becaplermin (rhPDGF-BB) gel. Becaplermin Studies Group. American Journal of Surgery. 1998;176:68S–73S. doi: 10.1016/s0002-9610(98)00174-3. [DOI] [PubMed] [Google Scholar]
  • 177.Rees RS, Robson MC, Smiell JM, Perry BH. Becaplermin gel in the treatment of pressure ulcers: a phase II randomized, double-blind, placebo-controlled study. Wound Repair and Regeneration. 1999;7:141–147. doi: 10.1046/j.1524-475x.1999.00141.x. [DOI] [PubMed] [Google Scholar]
  • 178.Guzman-Gardearzabal E, Leyva-Bohorquez G, Salas-Colin S, Paz-Janeiro JL, Alvarado-Ruiz R, Garcia-Salazar R. Treatment of chronic ulcers in the lower extremities with topical becaplermin gel.01%: a multicenter open-label study. Advances in Therapy. 2000;17:184–189. doi: 10.1007/BF02850294. [DOI] [PubMed] [Google Scholar]
  • 179.Nevins M, Giannobile WV, McGuire MK, Kao RT, Mellonig JT, Hinrichs JE, et al. Platelet-derived growth factor stimulates bone fill and rate of attachment level gain: results of a large multicenter randomized controlled trial. Journal of Periodontology. 2005;76:2205–2215. doi: 10.1902/jop.2005.76.12.2205. [DOI] [PubMed] [Google Scholar]
  • 180.Jayakumar A, Rajababu P, Rohini S, Butchibabu K, Naveen A, Reddy PK, et al. Multi-centre, randomized clinical trial on the efficacy and safety of recombinant human platelet-derived growth factor with beta-tricalcium phosphate in human intra-osseous periodontal defects. Journal of Clinical Periodontology. 2011;38:163–172. doi: 10.1111/j.1600-051X.2010.01639.x. [DOI] [PubMed] [Google Scholar]
  • 181.Digiovanni CW, Baumhauer J, Lin SS, Berberian WS, Flemister AS, Enna MJ, et al. Prospective, randomized, multi-center feasibility trial of rhPDGF-BB versus autologous bone graft in a foot and ankle fusion model. Foot and Ankle International. 2011;32:344–354. doi: 10.3113/FAI.2011.0344. [DOI] [PubMed] [Google Scholar]
  • 182.Fischer AN, Fuchs E, Mikula M, Huber H, Beug H, Mikulits W. PDGF essentially links TGF-beta signaling to nuclear beta-catenin accumulation in hepatocellular carcinoma progression. Oncogene. 2007;26:3395–3405. doi: 10.1038/sj.onc.1210121. [DOI] [PubMed] [Google Scholar]
  • 183.Update of safety review: follow-up to the March 27, 2008, Communication about the ongoing safety review of regranex (becaplermin) 2008 Jun 6; Page last updated: 01.07.2010; page accessed 7.18.2012, http://www.fda.gov/Drugs/DrugSafety/PostmarketDrugSafetyInformationforPatientsandProviders/DrugSafetyInformationforHeathcareProfessionals/ucm072148.htm.
  • 184.Ziyadeh N, Fife D, Walker AM, Wilkinson GS, Seeger JD. A matched cohort study of the risk of cancer in users of becaplermin. Advances in Skin & Wound Care. 2011;24:31–39. doi: 10.1097/01.ASW.0000392922.30229.b3. [DOI] [PubMed] [Google Scholar]
  • 185.Heilborn JD, Nilsson MF, Kratz G, Weber G, Sorensen O, Borregaard N, et al. The cathelicidin anti-microbial peptide LL-37 is involved in re-epithelialization of human skin wounds and is lacking in chronic ulcer epithelium. Journal of Investigative Dermatology. 2003;120:379–389. doi: 10.1046/j.1523-1747.2003.12069.x. [DOI] [PubMed] [Google Scholar]
  • 186.Tokumaru S, Sayama K, Shirakata Y, Komatsuzawa H, Ouhara K, Hanakawa Y, et al. Induction of keratinocyte migration via transactivation of the epidermal growth factor receptor by the antimicrobial peptide LL-37. Journal of Immunology. 2005;175:4662–4668. doi: 10.4049/jimmunol.175.7.4662. [DOI] [PubMed] [Google Scholar]
  • 187.Yin J, Yu FS. LL-37 via EGFR transactivation to promote high glucose-attenuated epithelial wound healing in organ-cultured corneas. Investigative Ophthalmology and Visual Science. 2010;51:1891–1897. doi: 10.1167/iovs.09-3904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Girnita A, Zheng H, Gronberg A, Girnita L, Stahle M. Identification of the cathelicidin peptide LL-37 as agonist for the type I insulin-like growth factor receptor. Oncogene. 2012;31:352–365. doi: 10.1038/onc.2011.239. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 189.Weber G, Chamorro CI, Granath F, Liljegren A, Zreika S, Saidak Z, et al. Human antimicrobial protein hCAP18/LL-37 promotes a metastatic phenotype in breast cancer. Breast Cancer Research. 2009;11:R6. doi: 10.1186/bcr2221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Coffelt SB, Marini FC, Watson K, Zwezdaryk KJ, Dembinski JL, LaMarca HL, et al. The pro-inflammatory peptide LL-37 promotes ovarian tumor progression through recruitment of multipotent mesenchymal stromal cells. Proceedings of the National Academy of Sciences of the United States of America. 2009;106:3806–3811. doi: 10.1073/pnas.0900244106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Heilborn JD, Nilsson MF, Jimenez CI, Sandstedt B, Borregaard N, Tham E, et al. Antimicrobial protein hCAP18/LL-37 is highly expressed in breast cancer and is a putative growth factor for epithelial cells. International Journal of Cancer. 2005;114:713–719. doi: 10.1002/ijc.20795. [DOI] [PubMed] [Google Scholar]
  • 192.Kim JP, Zhang K, Chen JD, Kramer RH, Woodley DT. Vitronectin-driven human keratinocyte locomotion is mediated by the alpha v beta 5 integrin receptor. Journal of Biological Chemistry. 1994;269:26926–26932. [PubMed] [Google Scholar]
  • 193.Kubo M, Van de Water L, Plantefaber LC, Mosesson MW, Simon M, Tonnesen MG, et al. Fibrinogen and fibrin are anti-adhesive for keratinocytes: a mechanism for fibrin eschar slough during wound repair. Journal of Investigative Dermatology. 2001;117:1369–1381. doi: 10.1046/j.0022-202x.2001.01551.x. [DOI] [PubMed] [Google Scholar]
  • 194.Johnson JP. Cell adhesion molecules in the development and progression of malignant melanoma. Cancer and Metastasis Reviews. 1999;18:345–357. doi: 10.1023/a:1006304806799. [DOI] [PubMed] [Google Scholar]
  • 195.Eaglstein WH, Sullivan T. Cyanoacrylates for skin closure. Dermatologic Clinics. 2005;23:193–198. doi: 10.1016/j.det.2004.09.003. [DOI] [PubMed] [Google Scholar]
  • 196.Greenberg CS, Birckbichler PJ, Rice RH. Transglutaminases: multifunctional cross-linking enzymes that stabilize tissues. FASEB Journal. 1991;5:3071–3077. doi: 10.1096/fasebj.5.15.1683845. [DOI] [PubMed] [Google Scholar]
  • 197.Chung SI, Folk JE. Transglutaminase from hair follicle of guinea pig (crosslinking-fibrin-glutamyllysine-isoenzymes-purified enzyme) Proceedings of the National Academy of Sciences of the United States of America. 1972;69:303–307. doi: 10.1073/pnas.69.2.303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Buxman MM, Wuepper KD. Keratin cross-linking and epidermal transglutaminase. A review with observations on the histochemical and immunochemical localization of the enzyme. Journal of Investigative Dermatology. 1975;65:107–112. doi: 10.1111/1523-1747.ep12598072. [DOI] [PubMed] [Google Scholar]
  • 199.Lichti U, Ben T, Yuspa SH. Retinoic acid-induced transglutaminase in mouse epidermal cells is distinct from epidermal transglutaminase. Journal of Biological Chemistry. 1985;260:1422–1426. [PubMed] [Google Scholar]
  • 200.Zhang W, Shiraishi A, Suzuki A, Zheng X, Kodama T, Ohashi Y. Expression and distribution of tissue transglutaminase in normal and injured rat cornea. Current Eye Research. 2004;28:37–45. doi: 10.1076/ceyr.28.1.37.23493. [DOI] [PubMed] [Google Scholar]
  • 201.Verma A, Mehta K. Tissue transglutaminase-mediated chemoresistance in cancer cells. Drug Resistance Updates. 2007;10:144–151. doi: 10.1016/j.drup.2007.06.002. [DOI] [PubMed] [Google Scholar]
  • 202.Mangala LS, Fok JY, Zorrilla-Calancha IR, Verma A, Mehta K. Tissue transglutaminase expression promotes cell attachment, invasion and survival in breast cancer cells. Oncogene. 2007;26:2459–2470. doi: 10.1038/sj.onc.1210035. [DOI] [PubMed] [Google Scholar]
  • 203.Mehta K, Kumar A, Kim HI. Transglutaminase 2: a multi-tasking protein in the complex circuitry of inflammation and cancer. Biochemical Pharmacology. 2010;80:1921–1929. doi: 10.1016/j.bcp.2010.06.029. [DOI] [PubMed] [Google Scholar]
  • 204.Xu L, Begum S, Hearn JD, Hynes RO. GPR56, an atypical G protein-coupled receptor, binds tissue transglutaminase, TG2, and inhibits melanoma tumor growth and metastasis. Proceedings of the National Academy of Sciences of the United States of America. 2006;103:9023–9028. doi: 10.1073/pnas.0602681103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Yuan L, Siegel M, Choi K, Khosla C, Miller CR, Jackson EN, et al. Transglutaminase-2 inhibitor, KCC009, disrupts fibronectin assembly in the extracellular matrix and sensitizes orthotopic glioblastomas to chemotherapy. Oncogene. 2007;26:2563–2573. doi: 10.1038/sj.onc.1210048. [DOI] [PubMed] [Google Scholar]
  • 206.Partridge EA, Le Roy C, Di Guglielmo GM, Pawling J, Cheung P, Granovsky M, et al. Regulation of cytokine receptors by Golgi N-glycan processing and endocytosis. Science. 2004;306:120–124. doi: 10.1126/science.1102109. [DOI] [PubMed] [Google Scholar]
  • 207.Vogler R, Sauer B, Kim DS, Schafer-Korting M, Kleuser B. Sphingosine-1-phosphate and its potentially paradoxical effects on critical parameters of cutaneous wound healing. Journal of Investigative Dermatology. 2003;120:693–700. doi: 10.1046/j.1523-1747.2003.12096.x. [DOI] [PubMed] [Google Scholar]
  • 208.Sauer B, Vogler R, von Wenckstern H, Fujii M, Anzano MB, Glick AB, et al. Involvement of Smad signaling in sphingosine 1-phosphate-mediated biological responses of keratinocytes. Journal of Biological Chemistry. 2004;279:38471–38479. doi: 10.1074/jbc.M313557200. [DOI] [PubMed] [Google Scholar]
  • 209.Kim DS, Kim SY, Kleuser B, Schafer-Korting M, Kim KH, Park KC. Sphingosine-1-phosphate inhibits human keratinocyte proliferation via Akt/protein kinase B inactivation. Cellular Signalling. 2004;16:89–95. doi: 10.1016/s0898-6568(03)00114-1. [DOI] [PubMed] [Google Scholar]
  • 210.Yamaguchi H, Kitayama J, Takuwa N, Arikawa K, Inoki I, Takehara K, et al. Sphingosine-1-phosphate receptor subtype-specific positive and negative regulation of Rac and haematogenous metastasis of melanoma cells. Biochemical Journal. 2003;374:715–722. doi: 10.1042/BJ20030381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Kawamori T, Osta W, Johnson KR, Pettus BJ, Bielawski J, Tanaka T, et al. Sphingosine kinase 1 is up-regulated in colon carcinogenesis. FASEB Journal. 2006;20:386–388. doi: 10.1096/fj.05-4331fje. [DOI] [PubMed] [Google Scholar]
  • 212.French KJ, Schrecengost RS, Lee BD, Zhuang Y, Smith SN, Eberly JL, et al. Discovery and evaluation of inhibitors of human sphingosine kinase. Cancer Research. 2003;63:5962–5969. [PubMed] [Google Scholar]
  • 213.Visentin B, Vekich JA, Sibbald BJ, Cavalli AL, Moreno KM, Matteo RG, et al. Validation of an anti-sphingosine-1-phosphate antibody as a potential therapeutic in reducing growth, invasion, and angiogenesis in multiple tumor lineages. Cancer Cell. 2006;9:225–238. doi: 10.1016/j.ccr.2006.02.023. [DOI] [PubMed] [Google Scholar]
  • 214.Zhang A, Wang MH, Dong Z, Yang T. Prostaglandin E2 is a potent inhibitor of epithelial-to-mesenchymal transition: interaction with hepatocyte growth factor. American Journal of Physiology Renal Physiology. 2006;291:F1323–F1331. doi: 10.1152/ajprenal.00480.2005. [DOI] [PubMed] [Google Scholar]
  • 215.Talwar M, Moyana TN, Bharadwaj B, Tan LK. The effect of a synthetic analogue of prostaglandin E2 on wound healing in rats. Annals of Clinical and Laboratory Science. 1996;26:451–457. [PubMed] [Google Scholar]
  • 216.Muller-Decker K, Hirschner W, Marks F, Furstenberger G. The effects of cyclooxygenase isozyme inhibition on incisional wound healing in mouse skin. Journal of Investigative Dermatology. 2002;119:1189–1195. doi: 10.1046/j.1523-1747.2002.19501.x. [DOI] [PubMed] [Google Scholar]
  • 217.Tanaka MN, Diaz BL, de Souza W, Morgado-Diaz JA. Prostaglandin E2-EP1 and EP2 receptor signaling promotes apical junctional complex disassembly of Caco-2 human colorectal cancer cells. BMC Cell Biology. 2008;9:63. doi: 10.1186/1471-2121-9-63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Xia D, Wang D, Kim SH, Katoh H, DuBois RN. Prostaglandin E2 promotes intestinal tumor growth via DNA methylation. Nature Medicine. 2012;18:224–226. doi: 10.1038/nm.2608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Dimitrijevich SD, Paranjape S, Wilson JR, Gracy RW, Mills JG. Effect of hyperbaric oxygen on human skin cells in culture and in human dermal and skin equivalents. Wound Repair and Regeneration. 1999;7:53–64. doi: 10.1046/j.1524-475x.1999.00053.x. [DOI] [PubMed] [Google Scholar]
  • 220.Feldmeier J, Carl U, Hartmann K, Sminia P. Hyperbaric oxygen: does it promote growth or recurrence of malignancy. Undersea and Hyperbaric Medicine. 2003;30:1–18. [PubMed] [Google Scholar]
  • 221.Kairuz E, Upton Z, Dawson RA, Malda J. Hyperbaric oxygen stimulates epidermal reconstruction in human skin equivalents. Wound Repair and Regeneration. 2007;15:266–274. doi: 10.1111/j.1524-475X.2007.00215.x. [DOI] [PubMed] [Google Scholar]
  • 222.Thom SR. Hyperbaric oxygen: its mechanisms and efficacy. Plastic and Reconstructive Surgery. 2011;127(Suppl. 1):131S–141S. doi: 10.1097/PRS.0b013e3181fbe2bf. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Jiang J, Tang YL, Liang XH. EMT: a new vision of hypoxia promoting cancer progression. Cancer Biology and Therapy. 2011;11:714–723. doi: 10.4161/cbt.11.8.15274. [DOI] [PubMed] [Google Scholar]
  • 224.Jing SW, Wang YD, Chen LQ, Sang MX, Zheng MM, Sun GG, et al. Hypoxia suppresses E-cadherin and enhances matrix metalloproteinase-2 expression favoring esophageal carcinoma migration and invasion via hypoxia inducible factor-1 alpha activation. Diseases of the Esophagus. 2012 doi: 10.1111/j.1442-2050.2011.01321.x. http://dx.doi.org/10.1111/j.1442-2050.2011.01321.x. [DOI] [PubMed] [Google Scholar]
  • 225.Gibson TL, Cohen P. Inflammation-related neutrophil proteases, cathepsin G and elastase, function as insulin-like growth factor binding protein proteases. Growth Hormone & IGF Research. 1999;9:241–253. doi: 10.1054/ghir.1999.0115. [DOI] [PubMed] [Google Scholar]
  • 226.Udayakumar TS, Chen ML, Bair EL, Von Bredow DC, Cress AE, Nagle RB, et al. Membrane type-1-matrix metalloproteinase expressed by prostate carcinoma cells cleaves human laminin-5 beta3 chain and induces cell migration. Cancer Research. 2003;63:2292–2299. [PubMed] [Google Scholar]
  • 227.López de Cicco R, Bassi DE, Zucker S, Seidah NG, Klein-Szanto AJ. Human carcinoma cell growth and invasiveness is impaired by the propeptide of the ubiquitous proprotein convertase furin. Cancer Research. 2005;65:4162–4171. doi: 10.1158/0008-5472.CAN-04-2820. [DOI] [PubMed] [Google Scholar]
  • 228.Ra HJ, Parks WC. Control of matrix metalloproteinase catalytic activity. Matrix Biology. 2007;26:587–596. doi: 10.1016/j.matbio.2007.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Mekkes JR, Le Poole IC, Das PK, Kammeyer A, Westerhof W. In vitro tissue-digesting properties of krill enzymes compared with fibrinolysin/DNAse, papain and placebo. The International Journal of Biochemistry & Cell Biology. 1997;29:703–706. doi: 10.1016/s1357-2725(96)00168-9. [DOI] [PubMed] [Google Scholar]
  • 230.Sangwan VS, Akpek EK, Voo I, Zhao T, Pinar V, Yang J, et al. Krill protease effects on wound healing after corneal alkali burn. Cornea. 1999;18:707–711. doi: 10.1097/00003226-199911000-00014. [DOI] [PubMed] [Google Scholar]
  • 231.Vanscheidt W, Vincent J. Krill enzymes in the therapy of chronical wounds. Zeitschrift fur Vulnerologie. 2007;1:10–15. [Google Scholar]
  • 232.Cufi S, Vazquez-Martin A, Oliveras-Ferraros C, Martin-Castillo B, Joven J, Menendez JA. Metformin against TGFbeta-induced epithelial-to-mesenchymal transition (EMT): from cancer stem cells to aging-associated fibrosis. Cell Cycle. 2010;9:4461–4468. doi: 10.4161/cc.9.22.14048. [DOI] [PubMed] [Google Scholar]
  • 233.Vazquez-Martin A, Oliveras-Ferraros C, Cufi S, Del Barco S, Martin-Castillo B, Menendez JA. Metformin regulates breast cancer stem cell ontogeny by transcriptional regulation of the epithelial–mesenchymal transition (EMT) status. Cell Cycle. 2010;9:3807–3814. [PubMed] [Google Scholar]
  • 234.Baraka AM, Deif MM. Role of activation of 5′-adenosine monophosphate-activated protein kinase in gastric ulcer healing in diabetic rats. Pharmacology. 2011;88:275–283. doi: 10.1159/000331879. [DOI] [PubMed] [Google Scholar]
  • 235.Spravchikov N, Sizyakov G, Gartsbein M, Accili D, Tennenbaum T, Wertheimer E. Glucose effects on skin keratinocytes: implications for diabetes skin complications. Diabetes. 2001;50:1627–1635. doi: 10.2337/diabetes.50.7.1627. [DOI] [PubMed] [Google Scholar]
  • 236.Li W, Ma Q, Liu J, Han L, Ma G, Liu H, et al. Hyperglycemia as a mechanism of pancreatic cancer metastasis. Frontiers in Bioscience. 2012;17:1761–1774. doi: 10.2741/4017. [DOI] [PubMed] [Google Scholar]
  • 237.Herzinger T, Kleuser B, Schäfer-Korting M, Korting HC. Sphingosine-1-phosphate signaling and the skin. American Journal of Clinical Dermatology. 2007;8:329–336. doi: 10.2165/00128071-200708060-00002. [DOI] [PubMed] [Google Scholar]
  • 238.Shrader M, Pino MS, Brown G, Black P, Adam L, Bar-Eli M, et al. Molecular correlates of gefitinib responsiveness in human bladder cancer cells. Molecular Cancer Therapeutics. 2007;6:277–285. doi: 10.1158/1535-7163.MCT-06-0513. [DOI] [PubMed] [Google Scholar]
  • 239.Frederick BA, Helfrich BA, Coldren CD, Zheng D, Chan D, Bunn PA, Jr, et al. Epithelial to mesenchymal transition predicts gefitinib resistance in cell lines of head and neck squamous cell carcinoma and non-small cell lung carcinoma. Molecular Cancer Therapeutics. 2007;6:1683–1691. doi: 10.1158/1535-7163.MCT-07-0138. [DOI] [PubMed] [Google Scholar]
  • 240.Fuchs BC, Fujii T, Dorfman JD, Goodwin JM, Zhu AX, Lanuti M, et al. Epithelialto-mesenchymal transition and integrin-linked kinase mediate sensitivity to epidermal growth factor receptor inhibition in human hepatoma cells. Cancer Research. 2008;6:2391–2399. doi: 10.1158/0008-5472.CAN-07-2460. [DOI] [PubMed] [Google Scholar]
  • 241.Rho JK, Choi YJ, Lee JK, Ryoo BY, Na II, Yang SH, et al. Epithelial to mesenchymal transition derived from repeated exposure to gefitinib determines the sensitivity to EGFR inhibitors in A549, a non-small cell lung cancer cell line. Lung Cancer. 2009;63:219–226. doi: 10.1016/j.lungcan.2008.05.017. [DOI] [PubMed] [Google Scholar]
  • 242.Chang TH, Tsai MF, Su KY, Wu SG, Huang CP, Yu SL, et al. Slug confers resistance to the epidermal growth factor receptor tyrosine kinase inhibitor. American Journal of Respiratory and Critical Care Medicine. 2011;183:1071–1079. doi: 10.1164/rccm.201009-1440OC. [DOI] [PubMed] [Google Scholar]
  • 243.Chung JH, Rho JK, Xu X, Lee JS, Yoon HI, Lee CT, et al. Clinical and molecular evidences of epithelial to mesenchymal transition in acquired resistance to EGFR-TKIs. Lung Cancer. 2011;73:176–182. doi: 10.1016/j.lungcan.2010.11.011. [DOI] [PubMed] [Google Scholar]
  • 244.Maseki S, Ijichi K, Tanaka H, Fujii M, Hasegawa Y, Ogawa T, et al. Acquisition of EMT phenotype in the gefitinib-resistant cells of a head and neck squamous cell carcinoma cell line through Akt/GSK-3beta/snail signalling pathway. British Journal of Cancer. 2012;106(6):1196–1204. doi: 10.1038/bjc.2012.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Pang LY, Bergkvist GT, Cervantes-Arias A, Yool DA, Muirhead R, Argyle DJ. Identification of tumour initiating cells in feline head and neck squamous cell carcinoma and evidence for gefitinib induced epithelial to mesenchymal transition. Veterinary Journal. 2012;193:46–52. doi: 10.1016/j.tvjl.2012.01.009. [DOI] [PubMed] [Google Scholar]
  • 246.Zahorowska B, Crowe PJ, Yang JL. Combined therapies for cancer: a review of EGFR-targeted monotherapy and combination treatment with other drugs. Journal of Cancer Research and Clinical Oncology. 2009;135:1137–1148. doi: 10.1007/s00432-009-0622-4. [DOI] [PubMed] [Google Scholar]
  • 247.Govindan R, Behnken D, Read W, McLeod H. Wound healing is not impaired by the epidermal growth factor receptor-tyrosine kinase inhibitor gefitinib. Annals of Oncology. 2003;14:1330–1331. doi: 10.1093/annonc/mdg352. [DOI] [PubMed] [Google Scholar]
  • 248.Donovan JC, Ghazarian DM, Shaw JC. Scarring alopecia associated with use of the epidermal growth factor receptor inhibitor gefitinib. Archives of Dermatology. 2008;144:1524–1525. doi: 10.1001/archderm.144.11.1524. [DOI] [PubMed] [Google Scholar]
  • 249.Muraoka T, Tsukuda K, Toyooka S, Kagawa S, Naomoto Y, Takemoto M, et al. Ileal perforation induced by acute radiation injury under gefitinib treatment. International Journal of Clinical Oncology. 2011;16:774–777. doi: 10.1007/s10147-011-0249-8. [DOI] [PubMed] [Google Scholar]
  • 250.Thomson S, Buck E, Petti F, Griffin G, Brown E, Ramnarine N, 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 Research. 2005;65:9455–9462. doi: 10.1158/0008-5472.CAN-05-1058. [DOI] [PubMed] [Google Scholar]
  • 251.Yauch RL, Januario T, Eberhard DA, Cavet G, Zhu W, Fu L, et al. Epithelial versus mesenchymal phenotype determines in vitro sensitivity and predicts clinical activity of erlotinib in lung cancer patients. Clinical Cancer Research. 2005;11:8686–8698. doi: 10.1158/1078-0432.CCR-05-1492. [DOI] [PubMed] [Google Scholar]
  • 252.Buck E, Eyzaguirre A, Barr S, Thompson S, Sennello R, Young D, et al. Loss of homotypic cell adhesion by epithelial–mesenchymal transition or mutation limits sensitivity to epidermal growth factor receptor inhibition. Molecular Cancer Therapeutics. 2007;6:532–541. doi: 10.1158/1535-7163.MCT-06-0462. [DOI] [PubMed] [Google Scholar]
  • 253.Jacobs MA, Wotkowicz C, Baumgart ED, Neto BS, Rieger-Christ KM, Bernier T, et al. Epidermal growth factor receptor status and the response of bladder carcinoma cells to erlotinib. Journal of Urology. 2007;178:1510–1514. doi: 10.1016/j.juro.2007.05.113. [DOI] [PubMed] [Google Scholar]
  • 254.Haddad Y, Choi W, McConkey DJ. Delta-crystallin enhancer binding factor-1 controls the epithelial to mesenchymal transition phenotype and resistance to the epidermal growth factor receptor inhibitor erlotinib in human head and neck squamous cell carcinoma lines. Clinical Cancer Research. 2009;15:532–542. doi: 10.1158/1078-0432.CCR-08-1733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Bryant JL, Britson J, Balko JM, Willian M, Timmons R, Frolov A, et al. A microRNA gene expression signature predicts response to erlotinib in epithelial cancer cell lines and targets EMT. British Journal of Cancer. 2012;106:148–156. doi: 10.1038/bjc.2011.465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Dennis M, Wang G, Luo J, Lin Y, Dohadwala M, Abemayor E, et al. Snail controls the mesenchymal phenotype and drives erlotinib resistance in oral epithelial and head and neck squamous cell carcinoma cells. Otolaryngology: Head and Neck Surgery. 2012 doi: 10.1177/0194599812446407. http://dx.doi.org/10.1177/0194599812446407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Nie F, Yang J, Wen S, An YL, Ding J, Ju SH, et al. Involvement of epidermal growth factor receptor overexpression in the promotion of breast cancer brain metastasis. Cancer. 2012 doi: 10.1002/cncr.27553. http://dx.doi.org/10.1002/cncr.27553. [DOI] [PubMed] [Google Scholar]
  • 258.Johnson KS, Levin F, Chu DS. Persistent corneal epithelial defect associated with erlotinib treatment. Cornea. 2009;28:706–707. doi: 10.1097/ICO.0b013e31818fdbc6. [DOI] [PubMed] [Google Scholar]
  • 259.Hepper DM, Wu P, Anadkat MJ. Scarring alopecia associated with the epidermal growth factor receptor inhibitor erlotinib. Journal of the American Academy of Dermatology. 2011;64:996–998. doi: 10.1016/j.jaad.2009.08.024. [DOI] [PubMed] [Google Scholar]
  • 260.Skvortsova I, Skvortsov S, Raju U, Stasyk T, Riesterer O, Schottdorf EM, et al. Epithelial-to-mesenchymal transition and c-myc expression are the determinants of cetuximab-induced enhancement of squamous cell carcinoma radioresponse. Radiotherapy and Oncology. 2010;96:108–115. doi: 10.1016/j.radonc.2010.04.017. [DOI] [PubMed] [Google Scholar]
  • 261.Oliveras-Ferraros C, Vazquez-Martin A, Cufi S, Queralt B, Baez L, Guardeno R, et al. Stem cell property epithelial-to-mesenchymal transition is a core transcriptional network for predicting cetuximab (Erbitux) efficacy in KRAS wild-type tumor cells. Journal of Cellular Biochemistry. 2011;112:10–29. doi: 10.1002/jcb.22952. [DOI] [PubMed] [Google Scholar]
  • 262.Holz C, Niehr F, Boyko M, Hristozova T, Distel L, Budach V, et al. Epithelial–mesenchymal-transition induced by EGFR activation interferes with cell migration and response to irradiation and cetuximab in head and neck cancer cells. Radiotherapy and Oncology. 2011;101:158–164. doi: 10.1016/j.radonc.2011.05.042. [DOI] [PubMed] [Google Scholar]
  • 263.Tejwani A, Wu S, Jia Y, Agulnik M, Millender L, Lacouture ME. Increased risk of high-grade dermatologic toxicities with radiation plus epidermal growth factor receptor inhibitor therapy. Cancer. 2009;115:1286–1299. doi: 10.1002/cncr.24120. [DOI] [PubMed] [Google Scholar]
  • 264.Konecny GE, Venkatesan N, Yang G, Dering J, Ginther C, Finn R, et al. Activity of lapatinib a novel HER2 and EGFR dual kinase inhibitor in human endometrial cancer cells. British Journal of Cancer. 2008;98:1076–1084. doi: 10.1038/sj.bjc.6604278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Lacouture ME, Laabs SM, Koehler M, Sweetman RW, Preston AJ, Di Leo A, et al. Analysis of dermatologic events in patients with cancer treated with lapatinib. Breast Cancer Research and Treatment. 2009;114:485–493. doi: 10.1007/s10549-008-0020-7. [DOI] [PubMed] [Google Scholar]
  • 266.Melisi D, Ishiyama S, Sclabas GM, Fleming JB, Xia Q, Tortora G, et al. LY2109761, a novel transforming growth factor beta receptor type I and type II dual inhibitor, as a therapeutic approach to suppressing pancreatic cancer metastasis. Molecular Cancer Therapeutics. 2008;7:829–840. doi: 10.1158/1535-7163.MCT-07-0337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Fransvea E, Angelotti U, Antonaci S, Giannelli G. Blocking transforming growth factor-beta up-regulates E-cadherin and reduces migration and invasion of hepatocellular carcinoma cells. Hepatology. 2008;47:1557–1566. doi: 10.1002/hep.22201. [DOI] [PubMed] [Google Scholar]
  • 268.Fransvea E, Mazzocca A, Antonaci S, Giannelli G. Targeting transforming growth factor (TGF)-betaRI inhibits activation of beta1 integrin and blocks vascular invasion in hepatocellular carcinoma. Hepatology. 2009;49:839–850. doi: 10.1002/hep.22731. [DOI] [PubMed] [Google Scholar]
  • 269.Mazzocca A, Fransvea E, Dituri F, Lupo L, Antonaci S, Giannelli G. Down-regulation of connective tissue growth factor by inhibition of transforming growth factor beta blocks the tumor-stroma cross-talk and tumor progression in hepatocellular carcinoma. Hepatology. 2010;51:523–534. doi: 10.1002/hep.23285. [DOI] [PubMed] [Google Scholar]
  • 270.Zhang B, Halder SK, Kashikar ND, Cho YJ, Datta A, Gorden DL, et al. Antimetastatic role of Smad4 signaling in colorectal cancer. Gastroenterology. 2010;138:969–980. doi: 10.1053/j.gastro.2009.11.004. e961–963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Ganapathy V, Ge R, Grazioli A, Xie W, Banach-Petrosky W, Kang Y, et al. Targeting the transforming growth factor-beta pathway inhibits human basal-like breast cancer metastasis. Molecular Cancer. 2010;9:122. doi: 10.1186/1476-4598-9-122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Flechsig P, Dadrich M, Bickelhaupt S, Jenne J, Hauser K, Timke C, et al. LY2109761 attenuates radiation-induced pulmonary murine fibrosis via reversal of TGF-beta and BMP associated proinflammatory and proangiogenic signals. Clinical Cancer Research. 2012 doi: 10.1158/1078-0432.CCR-11-2855. [DOI] [PubMed] [Google Scholar]
  • 273.Rausch MP, Hahn T, Ramanathapuram L, Bradley-Dunlop D, Mahadevan D, Mercado-Pimentel ME, et al. An orally active small molecule TGF-beta receptor I antagonist inhibits the growth of metastatic murine breast cancer. Anticancer Research. 2009;29:2099–2109. [PMC free article] [PubMed] [Google Scholar]
  • 274.Garrison K, Hahn T, Lee WC, Ling LE, Weinberg AD, Akporiaye ET. The small molecule TGF-beta signaling inhibitor SM16 synergizes with agonistic OX40 antibody to suppress established mammary tumors and reduce spontaneous metastasis. Cancer Immunology, Immunotherapy. 2012;61:511–521. doi: 10.1007/s00262-011-1119-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Vittal R, Zhang H, Han MK, Moore BB, Horowitz JC, Thannickal VJ. Effects of the protein kinase inhibitor, imatinib mesylate, on epithelial/mesenchymal phenotypes: implications for treatment of fibrotic diseases. The Journal of Pharmacology and Experimental Therapeutics. 2007;321:35–44. doi: 10.1124/jpet.106.113407. [DOI] [PubMed] [Google Scholar]
  • 276.Puissant A, Dufies M, Fenouille N, Ben Sahra I, Jacquel A, Robert G, et al. Imatinib triggers mesenchymal-like conversion of CML cells associated with increased aggressiveness. Journal of Molecular and Cellular Biology. 2012;4:207–220. doi: 10.1093/jmcb/mjs010. [DOI] [PubMed] [Google Scholar]
  • 277.Tarnawski A, Szabo IL, Husain SS, Soreghan B. Regeneration of gastric mucosa during ulcer healing is triggered by growth factors and signal transduction pathways. Journal of Physiology, Paris. 2001;95:337–344. doi: 10.1016/s0928-4257(01)00046-8. [DOI] [PubMed] [Google Scholar]
  • 278.Ehrlich HP, Freedman BM. Topical platelet-derived growth factor in patients enhances wound closure in the absence of wound contraction. Cytokines, Cellular and Molecular Therapy. 2002;7:85–90. doi: 10.1080/13684730310001643. [DOI] [PubMed] [Google Scholar]
  • 279.Ermolov AS, Smirnov SV, Khvatov VB, Istranov LP, Koniushko OI, Kolokolchikova EG, et al. The use of bioactive wound dressing, stimulating epithelial regeneration of IIIa-degree burn wounds. Bulletin of Experimental Biology and Medicine. 2008;146:153–157. doi: 10.1007/s10517-008-0225-0. [DOI] [PubMed] [Google Scholar]
  • 280.Alexaki VI, Simantiraki D, Panayiotopoulou M, Rasouli O, Venihaki M, Castana O, et al. Adipose tissue-derived mesenchymal cells support skin reepithelialization though secretion of KGF-1 and PDGF-BB: comparison with dermal fibroblasts. Cell Transplantation. 2012 doi: 10.3727/096368912X637064. [DOI] [PubMed] [Google Scholar]
  • 281.Pintor J, Bautista A, Carracedo G, Peral A. UTP and diadenosine tetraphosphate accelerate wound healing in the rabbit cornea. Ophthalmic and Physiological Optics. 2004;24:186–193. doi: 10.1111/j.1475-1313.2004.00182.x. [DOI] [PubMed] [Google Scholar]
  • 282.Klepeis VE, Weinger I, Kaczmarek E, Trinkaus-Randall V. P2Y receptors play a critical role in epithelial cell communication and migration. Journal of Cellular Biochemistry. 2004;93:1115–1133. doi: 10.1002/jcb.20258. [DOI] [PubMed] [Google Scholar]
  • 283.Weinger I, Klepeis VE, Trinkaus-Randall V. Tri-nucleotide receptors play a critical role in epithelial cell wound repair. Purinergic Signal. 2005;1:281–292. doi: 10.1007/s11302-005-8132-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284.Mediero A, Peral A, Pintor J. Dual roles of diadenosine polyphosphates in corneal epithelial cell migration. Investigative Ophthalmology and Visual Science. 2006;47:4500–4506. doi: 10.1167/iovs.06-0209. [DOI] [PubMed] [Google Scholar]
  • 285.Taboubi S, Milanini J, Delamarre E, Parat F, Garrouste F, Pommier G, et al. G alpha(q/11)-coupled P2Y2 nucleotide receptor inhibits human keratinocyte spreading and migration. FASEB Journal. 2007;21:4047–4058. doi: 10.1096/fj.06-7476com. [DOI] [PubMed] [Google Scholar]
  • 286.Taboubi S, Garrouste F, Parat F, Pommier G, Faure E, Monferran S, et al. Gq-coupled purinergic receptors inhibit insulin-like growth factor-I/phosphoinositide 3-kinase pathway-dependent keratinocyte migration. Molecular Biology of the Cell. 2010;21:946–955. doi: 10.1091/mbc.E09-06-0497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Boucher I, Rich C, Lee A, Marcincin M, Trinkaus-Randall V. The P2Y2 receptor mediates the epithelial injury response and cell migration. American Journal of Physiology Cell Physiology. 2010;299:C411–C421. doi: 10.1152/ajpcell.00100.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288.Boucher I, Kehasse A, Marcincin M, Rich C, Rahimi N, Trinkaus-Randall V. Distinct activation of epidermal growth factor receptor by UTP contributes to epithelial cell wound repair. The American Journal of Pathology. 2011;178:1092–1105. doi: 10.1016/j.ajpath.2010.11.060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289.Gendaszewska-Darmach E, Kucharska M. Nucleotide receptors as targets in the pharmacological enhancement of dermal wound healing. Purinergic Signalling. 2011;7:193–206. doi: 10.1007/s11302-011-9233-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290.Degagné E, Degrandmaison J, Grbic DM, Vinette V, Arguin G, Gendron FP. P2Y(2) receptor promotes intestinal epithelial cell migration through a Go protein and integrin α(v) pathway allowing microtubule stabilization and mucosal re-epithelization in experimental colitis. Journal of Cellular Physiology. 2012 May; doi: 10.1002/jcp.24109. http://dx.doi.org/10.1002/jcp.24109 [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
  • 291.Kawai Y, Kaidoh M, Ohhashi T. MDA-MB-231 produces ATP-mediated ICAM-1-dependent facilitation of the attachment of carcinoma cells to human lymphatic endothelial cells. American Journal of Physiology Cell Physiology. 2008;295:C1123–C1132. doi: 10.1152/ajpcell.00247.2008. [DOI] [PubMed] [Google Scholar]; Li S, Huang S, Peng SB. Overexpression of G protein-coupled receptors in cancer cells: involvement in tumor progression. International Journal of Oncology. 2005;27:1329–1339. [PubMed] [Google Scholar]
  • 292.Davis FM, Kenny PA, Soo ET, van Denderen BJ, Thompson EW, Cabot PJ, et al. Remodeling of purinergic receptor-mediated Ca2+ signaling as a consequence of EGF-induced epithelial–mesenchymal transition in breast cancer cells. PLoS One. 2011;6:e23464. doi: 10.1371/journal.pone.0023464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293.Davis FM, Kenny PA, Soo ET, van Denderen BJ, Thompson EW, Cabot PJ, Parat MO, Roberts-Thomson SJ, Monteith GR. Remodeling of purinergic receptor-mediated Ca2+ signaling as a consequence of EGF-induced epithelial-mesenchymal transition in breast cancer cells. PLoS One. 2011;6:e23464. doi: 10.1371/journal.pone.0023464. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES