The wound healing, chronic fibrosis, and cancer progression triad Brad Rybinski, Janusz Franco-Barraza and Edna Cukierman Physiol. Genomics 46:223-244, 2014. First published 11 February 2014; doi:10.1152/physiolgenomics.00158.2013 You might find this additional info useful... Supplemental material for this article can be found at: /content/suppl/2014/03/09/physiolgenomics.00158.2013.DC1.html This article cites 411 articles, 96 of which can be accessed free at: /content/46/7/223.full.html#ref-list-1 This article has been cited by 1 other HighWire hosted articles The Runt-related transcription factor 1 in prostate cancer-associated fibroblasts Hilary A. Coller PNAS, November 18, 2014; 111 (46): 16238-16239. [Full Text] [PDF]

Additional material and information about Physiological Genomics can be found at: http://www.the-aps.org/publications/pg

This information is current as of November 30, 2014.

Physiological Genomics publishes results of a wide variety of studies from human and from informative model systems with techniques linking genes and pathways to physiology, from prokaryotes to eukaryotes. It is published quarterly in January, April, July, and October by the American Physiological Society, 9650 Rockville Pike, Bethesda MD 20814-3991. Copyright © 2014 by the American Physiological Society. ISSN: 1094-8341, ESSN: 1531-2267. Visit our website at http://www.the-aps.org/.

Downloaded from on November 30, 2014

Updated information and services including high resolution figures, can be found at: /content/46/7/223.full.html

Physiol Genomics 46: 223–244, 2014. First published February 11, 2014; doi:10.1152/physiolgenomics.00158.2013.

Review

The wound healing, chronic fibrosis, and cancer progression triad Brad Rybinski,* Janusz Franco-Barraza,* and Edna Cukierman Cancer Biology Program, Fox Chase Cancer Center/Temple Health, Philadelphia, Pennsylvania Submitted 8 October 2013; accepted in final form 4 February 2014

Rybinski B, Franco-Barraza J, Cukierman E. The wound healing, chronic fibrosis, and cancer progression triad. Physiol Genomics 46: 223–244, 2014. First published February 11, 2014; doi:10.1152/physiolgenomics.00158.2013.—For decades tumors have been recognized as “wounds that do not heal.” Besides the commonalities that tumors and wounded tissues share, the process of wound healing also portrays similar characteristics with chronic fibrosis. In this review, we suggest a tight interrelationship, which is governed as a concurrence of cellular and microenvironmental reactivity among wound healing, chronic fibrosis, and cancer development/progression (i.e., the WHFC triad). It is clear that the same cell types, as well as soluble and matrix elements that drive wound healing (including regeneration) via distinct signaling pathways, also fuel chronic fibrosis and tumor progression. Hence, here we review the relationship between fibrosis and cancer through the lens of wound healing.

tumor stroma constitutes a remarkable discerning challenge for investigators. However, the tumor niche resembles a site of chronic wound healing (16, 84, 317). Out of the many similarities between wound healing and tumor progression, the mutual presence of myofibroblastic cells has emerged as a particularly common hallmark (56). Myofibroblasts are contractile spindle-shaped and stress fiberforming mesenchymal cells that are known to express alpha smooth muscle actin (␣-SMA), contain excessive structural rough endoplasmic reticulum, and produce copious amounts of extracellular matrix (ECM) (56, 90, 183). Myofibroblasts are also seen during fetal development (183) and are transiently present at sites of acute wound healing to repair injured ECM (i.e., to renew and/or to crosslink ECM fibers) and facilitate wound closure. Nonetheless, their constant presence in tumoralactivated stroma (i.e., desmoplasia) is known to encourage neoplastic progression (16, 56). Interestingly, similar persistent myofibroblastic populations are also common occurrences during chronic tissue fibrosis (56, 317, 390). Hence, we propose to review the triad “wound healing/chronic fibrosis/cancer” (WHFC) through the lens of wound healing while highlighting their numerous common and few dissimilar traits. The wound healing response to injury is known to trigger functional (i.e., proliferative) and phenotypic changes in fibroblasts, lymphocytes, and epithelial and endothelial cells (212). For example, the changes in epithelial and stromal cells respectively correspond to the epithelial-mesenchymal transition (EMT) and myofibroblastic differentiation processes (212). Both EMT and myofibroblastic differentiation are common occurrences during wound healing (or tissue regeneration) but

THE SHEER COMPLEXITY OF THE

* B. Rybinski and J. Franco-Barraza contributed equally to this work. Address for reprint requests and other correspondence: E. Cukierman, Cancer Biology, Fox Chase cancer Center, 333 Cottman Ave. (W428), Philadelphia, PA 19111 (e-mail: [email protected]).

also throughout development, inflammation, and fibrosis, as well as during tumor progression. In EMT, epithelial cells acquire a mesenchymal migratory phenotype, switching the expression of known cell-cell adhesion molecules like Ecadherin in favor of mesenchymal proteins such as N-cadherin and vimentin (362). In cancer, EMT facilitates metastasis by promoting ECM proteolysis and enhancing malignant cell motility, thus conferring an ability to invade (362). EMT also renders cancer cells a stem-like character, which is believed to be responsible for therapeutic evasion, as well as a distinctive prolonged latency/survival mechanism known as dormancy (38). Myofibroblastic differentiation, also known as fibroblastic activation, describes the process by which the mesenchymal/fibroblastic components of the stroma (i.e., resident fibroblasts, recruited bone marrow-derived mesenchymal cells, pericytes, endothelial cells, and others) acquire a myofibroblastic phenotype (90). EMT and myofibroblast differentiation are regulated by similar factors and signaling pathways (Supplemental Table S1).1 Moreover, some investigators believe that stromal myofibroblasts arise also from epithelial cells via EMT-like processes (56, 123). Importantly, the healing process during acute injury involves four components: coagulation, inflammation, cellular proliferation, and ECM repair (212). Hence, wound healing (or regeneration) could be regarded as a four-component process that constitutes the foundation for cell proliferation, EMT, and myofibroblast differentiation in the WHFC triad. Similarly to how Champollion deciphered hieroglyphs by understanding Greek and Coptic, wound healing constitutes “the Rosetta Stone” needed for a better understanding of the WHFC triad (Fig. 1). Chronic fibrosis and tumorigenesis can both result from a prolonged and exacerbated healing response that could stem 1

The online version of this article contains supplemental material.

1094-8341/14 Copyright © 2014 the American Physiological Society

223

Downloaded from on November 30, 2014

wound healing; regeneration; fibrosis; cancer; tumor stroma; myofibroblasts; tumor- or cancer-associated fibroblasts; desmoplasia; epithelial-to-mesenchymal transition

Review 224

THE WHFC TRIAD

from chronic injury (195, 317, 390), and as such the similarities that tumors and their stroma have in common with chronic fibrosis are intriguing. In fact, it is well known that chronic fibrosis, such as in liver cirrhosis induced by persistent viral infection (i.e., hepatitis), sustained alcohol consumption or toxin-containing food, constitutes a direct cancer predisposition (86, 91, 117). The epithelia from both fibrotic and tumor tissues are characterized by the presence of hyperproliferating cells (219, 269, 288); however, carcinoma cells carry transformative mutations (as well as epigenetic changes) presumed to be absent in the fibrotic tissue. Also intriguing is the stromal element of both conditions, a dense myofibroblastic component with an excessive deposition of ECM (56). The large population of myofibroblasts at the tumor stroma is referred to as cancer- or carcinoma-associated fibroblasts (i.e., known as CAFs) and also, in this review, as tumor-associated fibroblasts (TAFs). TAFs and fibrotic myofibroblasts drive tumor progression and chronic fibrosis, respectively, through both paracrine effects on surrounding cells and exacerbated synthesis of ECM (16, 56, 286, 389). TAFs express a plethora of protumorigenic growth factors, inflammatory cytokines, proteolytic enzymes, and ECM proteins such as collagen (Col)-I and -III as well as a fibronectin spliced variant known as ED-A and more (56). TAFs are also known to contribute to the degradation of Col-IV, a protein associated with the epithelial ECM known as basement membrane (158, 235). Alterations to mesenchymal ECM deposition, together with the hyperproliferation of the stromal compartment in the vicinity of neoplastic lesions, are known as “desmoplasia” or “desmoplastic reaction.” As such, the desmoplastic reaction bears a striking resemblance to the modified ECM typical of chronic fibrosis, which is also characterized by increased Col-I and -III deposition (390). Finally,

fibrotic myofibroblasts and TAFs both remodel the ECM fiber distribution by physically contracting and pulling on ECM proteins after engaging particular sets of integrins (140). Even though it remains unclear whether TAFs and fibrotic myofibroblasts possess unique traits that differentiate them, similarities, such as their contractile activity and the common expression of ␣-SMA and others, suggest numerous phenotypic commonalities (56). Hence, it is only logical to assume that effective targeting drugs that interfere with one (i.e., chronic fibrosis) will also stall the other (i.e., desmoplasia). Supplemental Table S2 includes a list of drugs targeting both diseases. While chronic fibrosis predisposes the affected tissues to develop cancer (9, 30, 81, 91, 117, 232), a desmoplastic reaction that correlates with poor prognosis (56, 125, 317) is also believed to enhance tumor progression. In turn, transformed tumor cells also promote TAF activation, thus supporting the desmoplastic reaction (16, 20, 56). Describing the relationship between cancer and fibrosis in these terms implies that fibrosis could be sparking tumorigenesis and tumor progression might be instigating chronic fibrosis. We suggest that the relationship between cancer and fibrosis (i.e., the WHFC triad) constitutes both correlation and causation (Fig. 1). Therefore, here we review the mechanisms by which the prolonged four component process of wound healing facilitates chronic fibrosis and tumorigenesis through a common mechanistic link: the WHFC triad. THE COAGULATION CASCADE FUELS THE WHFC TRIAD

Wound healing begins with the process of coagulation where platelets from circulating blood accumulate at the damaged site and, together with increased levels of coagulation factors as

Physiol Genomics • doi:10.1152/physiolgenomics.00158.2013 • www.physiolgenomics.org

Downloaded from on November 30, 2014

Fig. 1. The wound healing, chronic fibrosis, and cancer (WHFC) triad. Model depicting the WHFC triad commonalities based on wound healing. The cartoon highlights the fact that if wound resolution (as in acute wound healing) is not attained then chronic wound healing facilitates a vicious cycle between fibrosis and cancer. EMT, epithelial-mesenchymal transition.

Review THE WHFC TRIAD

healing is eventually broken down by plasmin, a cleaved subproduct of plasminogen (111). Plasmin also plays a role in the ECM context due to its proteolytic activity in Col degradation, transforming growth factor beta (TGF-␤) activation (see below) and release of other ECM-bound growth factors (111). Urokinase-type plasminogen activator (uPA) and tissuetype plasminogen activator (tPA) can each cleave plasminogen to obtain plasmin, and both enzymes are inactivated by plasminogen activator inhibitor-1 (PAI-1)(111). Interestingly, uPA has been reported to inhibit fibrosis (216, 334, 335) but also to promote tumor progression(335). Similarly, the role of tPA is also unclear as it has been reported both as promoter as well as inhibitor of chronic fibrosis and cancer (62, 144, 196, 303, 405). Finally, PAI-1 has also been shown to either promote or inhibit these two diseases (111, 155). The above-mentioned facts together provide rationale to suggest that the coagulation cascade constitutes a shared WHFC triad targetable pathway. CHRONIC INFLAMMATION IS A DRIVER OF THE WHFC TRIAD

Although overly simplified, it is generally agreed that, during homeostatic disturbances (i.e., during wound healing), M1 type macrophages drive an inflammatory response known to be tumor suppressive and antifibrotic, while M2 type macrophages promote profibrotic and protumorigenic effects (96, 390). In response to macrophages and to injured epithelial cells, naïve CD4⫹ T-cells can differentiate into largely antifibrotic and antitumorigenic Th1 cells or profibrotic and protumorigenic Th2, Th17, and Treg cells, while CD8⫹ T-cells are considered cytotoxic and constitute a positive neoplastic prognostic marker (390). During the acute wound healing response, immunity is triggered to prevent infection and to induce quick healing. Hence, a plethora of acute inflammatory secreted cytokines induce cellular proliferation, EMT, and myofibroblastic differentiation (56, 390). Importantly, even though EMT has been proved to be difficult to detect in many cancers, chronic inflammation associated with fibrosis (389) and tumor progression (103) could induce persistent EMT as well as myofibroblast differentiation, both of which are known to further fuel the WHFC triad. It has been shown that inflammatory responses promote tumor development and progression including metastasis, and this axis has been thoroughly reviewed (120). In the following sections as well as in Supplemental Table S1, we highlight several inflammatory factors that influence chronic fibrosis and tumor progression because of their effects on cellular plasticity related to the WHFC triad. Please note that many of these factors have also been reviewed elsewhere for their effects in regulating the plasticity of mesenchymal stem cells (343, 397, 406). TGF-␤ The TGF-␤ superfamily of cytokines plays a notorious role in the WHFC triad. TGF-␤ exerts a dual effect during cancer progression, modulating the behavior of both epithelial and stromal cells imparting both pro- and antitumorigenic effects (238). For example, it has been described that TGF-␤ plays an antitumoral role on preneoplastic liver lesions by inhibiting epithelial cells’ growth and driving proapoptotic signaling when induced by other cytokines such as interferon-␣ (70). On the other hand, during more advanced tumorigenic stages,

Physiol Genomics • doi:10.1152/physiolgenomics.00158.2013 • www.physiolgenomics.org

Downloaded from on November 30, 2014

well as with fibrinogen deposition and cleavage, result in the formation of insoluble fibrin strands building up a clot (212). The clot prevents blood loss and constitutes a short lived scaffold or provisional ECM that serves both for cell migration/invasion and as a growth factor reservoir (212). In chronic fibrosis as in cancer, a process resembling clotting fuels disease progression. For example, fibrin bundles can accumulate at the tumor stroma as a result of a leaky tumor vasculature (260), and similar fibrin deposition has also been reported in a variety of fibrotic tissues (151, 308). Although the role of fibrin in chronic fibrosis remains rather unclear, it is known that fibrinogen exerts a mitogenic effect on fibroblasts (337). During tumor progression, fibrin facilitates angiogenesis (93); meanwhile, fibrinogen also promotes angiogenesis as well as oncogenic proliferation and antitumorigenic immune suppression (135). Moreover, elevated plasma fibrinogen correlates with a poor cancer prognosis (110, 203, 225, 290, 296, 321, 356). Intriguingly, fibrinogen may induce a prolonged inflammatory state in several types of cancer and fibrosis (68), suggesting a possible link between chronic inflammation and persistent activity of the coagulation cascade. The cleavage of fibrinogen is considered the end result of the coagulation cascade signaling pathway (160). Thrombin directly cleaves fibrinogen into fibrin (95). Besides its role in the coagulation cascade, thrombin promotes cancer progression and fibrosis (47, 95). In cancer, thrombin induces cell proliferation, tumor growth, angiogenesis, and metastasis (95). The ability of thrombin to act as a mitogen is not limited to an epithelial context. It also has been reported to enhance fibroblast proliferation in a model of bleomycin-induced pulmonary fibrosis (358). Furthermore, thrombin inhibition reduces the severity of both pulmonary (358) and liver fibrosis (83). The ability of thrombin to promote cancer and fibrosis is believed to be mediated in part through its ability to activate the protease-activated receptor (PAR)-1 (47, 95). PAR-1 is present in a variety of tumor types (89), and its activation promotes cancer cell proliferation and angiogenesis (95). In the clinic, PAR-1 signaling is suspected to facilitate melanoma metastasis (416), and it correlates with poor prognosis of gastric, prostate, and breast cancers (28, 100, 313). PAR-1 also facilitates pulmonary fibrosis (143), while PAR-1-deficient mice seem to resist bleomycin-induced pulmonary fibrosis (142). PAR-1 is one of four PAR isoforms (PAR-1 to -4), and, while thrombin induces many of its effects via PAR-1, PAR-3 and -4 are also suitable to be activated by thrombin (47). However, PAR-2 can be activated by other molecules from the coagulation cascade such as the tissue factor (TF) and activated factor X (FXA) (47). TF is expressed by many types of cancer cells (169), and TF or FXA-induced PAR-2 signaling has been implicated in both chronic fibrosis and tumor progression (33, 34, 151, 169, 318, 347). Interestingly, TF, FXA, thrombin, and activation of PAR-1 or PAR-2 may possibly promote myofibroblastic differentiation (Supplemental Table S1). This type of common occurrence opens up the possibility that other enzymes from the coagulation cascade could also induce/ sustain myofibroblastic populations during chronic fibrosis and tumor desmoplasia. To this end, the excess of fibrin deposition observed in chronic fibrosis and desmoplastic stroma indicates a possible enhancement in fibrin synthesis but also a likely decreased rate of its degradation. The fibrin/platelet clot formed during wound

225

Review 226

THE WHFC TRIAD

Interleukin-6 An important acute phase mediator, interleukin-6 (IL-6), is also a proinflammatory factor that has a prominent role in stromal activation. IL-6 null mice exhibit delayed wound healing related to a diminished myofibroblast differentiation (105). Moreover, IL-6 null mice exhibit an attenuated lung fibrosis phenotype in response to radiation (311), suggesting a

role for IL-6-induced myofibroblast differentiation in fibrosis. In addition, IL-6 induces epithelial cancer cell proliferation (315) while also activating the EMT program (145, 348) and contributing to a preference for bone metastasis (8, 327). It is well documented that IL-6 promotes colitis-associated cancer via a STAT3-dependent mechanism (119). Finally, myofibroblasts from a variety of tissues are known to produce IL-6 (46, 330, 361), suggesting that mesenchymal IL-6 may participate in the WHFC triad by facilitating tissue fibrosis, tumoral desmoplasia, epithelial growth, and metastasis. IL-17 IL-17 is a cytokine that is upregulated in response to Th17 effector cytokine IL-23 (121, 379). IL-17 upregulation has been associated, together with other cytokines such as IL-6 and IL-22, as proinflammatory (394) as well as protumorigenic (379). IL-17 is known to act on myeloid and mesenchymal cells to induce expression of IL-6 (and others) (394). In addition, IL-17 is believed to induce expression of CXC chemokines known for their neurophil chemo-attractive capabilities (394). Interestingly, IL-17 has also been associated with the expression of ECM remodeling enzymes such as matrix metalloproteinases (MMPs) MMP1, MMP3, MMP9, and MMP13, which are linked to both chronic inflammation and cancer (394). In particular, IL-17 has been shown to be activated by local microbial factors that can particularly penetrate neoplastic but not normal tissues. For example, a deterioration of the colorectal cancer tumoral barrier is induced in early lesions, allowing microbial factors to promote local IL-17dependent inflammation to further drive tumor growth (121). Interestingly, IL-17 has an ability to activate fibroblasts in autoimmunity, liver fibrosis, and cancer (246). Together these facts emphasize an important role for IL-17 in the WHFC triad. Chemokines For the success of a wound resolution, it is necessary to coordinate the recruitment of several types of cells (e.g., inflammatory cells, macrophages, fibroblasts, and endothelial and epithelial cells) to the insulted tissue. This recruitment is mediated by chemotactic cytokines (chemokines), which are diffusible molecules that activate cell proliferation and direct cell migration toward the wound (21). The cytokines and chemokines involved in wound healing and their influence in tumor microenvironment (370) have been listed in several publications (21, 191, 411). In this review, we highlight some chemokines that are especially relevant to the WHFC triad. During wound healing, the CXC chemokine family facilitates angiogenesis as well as lymphocyte recruitment (411). CXC receptors (CXCRs), in combination with chemokine ligands such as CXCL8 (184), play a key role during various cellular key responses (76, 253, 411). Therefore, it is not surprising that deregulation of the CXCRs/CXCL8 axis leads to fibrotic pathologies affecting lung (237, 340), kidney (351), and liver (417). Moreover, CXCR receptors have been proposed as therapeutic targets in some fibrotic conditions such as asthma (48, 175). In cancer, CXCRs/CXCL8 seem to be responsible for driving cancer cell proliferation, survival, and metastasis (104). Stromal CXCL8 potentiates the metastatic phenotype of cancer cells (104). More recently, CXCR2 has been implicated in sustaining a protumor inflammatory envi-

Physiol Genomics • doi:10.1152/physiolgenomics.00158.2013 • www.physiolgenomics.org

Downloaded from on November 30, 2014

TGF-␤ has also been associated with increased invasion and metastasis of malignant cells, correlating with enhanced activity and over expression of several integrins (238) as well as triggering EMT (242). Regarding its effects on stromal cells, TGF-␤ can also induce both myofibroblastic differentiation (238) and altered ECM production (254). Of note and documented in several models of renal fibrosis (413), TGF-␤ can, in addition, induce endothelial to mesenchymal transduction (16, 412). Although it acts through different cells and mechanisms and clearly participates as a tumor suppressor at early tumorigenic stages, it is nevertheless clear that TGF-␤ also promotes fibrosis (238) as well as tumor progression and metastasis (242). Certainly more information is needed in this regard as knockout of the TGF-␤ receptor II was shown to have tumorpromoting consequences (271), while collective cell migration was shown to improve under TGF-␤ signaling depletion (243). Nonetheless, TGF-␤ is evidently the most dominant profibrogenic factor known in all tissues, and anti-TGF-␤ agents (see Supplemental Table S2) are highly effective in a variety of fibrosis models. Latent TGF-␤ is secreted by various cell types, such as lymphocytes, platelets, cancer cells, myofibroblasts, and others, during the WHFC triad. For example, latent TGF-␤ can be secreted by cancer cells (242), inducing myofibroblastic differentiation of stromal cells, contributing to enrich an invasive TAF population (72, 211). Latent TGF-␤ is the inactive form of TGF-␤ that is found linked to the ECM and is sequestered by the latency-associated protein (LAP) and latent TGF-␤-binding protein 1 (5). Latent TGF-␤ can be activated by ECM damage and myofibroblastic engagement of LAP via cell-matrix receptors such as integrins ␣v␤1, ␣v␤3, ␣v␤5, ␣v␤6, ␣v␤8, and others (257). TGF-␤ can be released and activated via ECM contraction induced by myofibroblastic cells (5). In fact, it has been suggested that changes in matrix stiffness could regulate the equilibrium between storage and release of matrix-bound factors such as TGF-␤ (383). In addition, the integrin-mediated binding of LAP may trigger a conformational change in the molecule, releasing the bioactive form of this factor (5). It is noteworthy that myofibroblasts secrete and activate TGF-␤, establishing a self-sustained feedback, contributing to expand their own population. In addition, TGF-␤ effects are known to induce an increase in Col-I-rich ECM production (254). Autocrine TGF-␤ signaling has been shown to drive myofibroblast differentiation during tumor progression (185), suggesting a similar operational process in chronic fibrosis. Interestingly, the activation of TGF-␤ and subsequent myofibroblast differentiation can be triggered in vitro by a constant interstitial fluidic flow (265), suggesting the possibility of a physical effect of augmented tissue pressure, enhancing and expanding both myofibroblast and TAFs populations at the vicinity of the WHFC triad. To this end, it is not surprising then to recognize that all the WHFC triad diseases present changes in interstitial fluid flow.

Review THE WHFC TRIAD

moral components (298). Moreover, in breast cancer cells in vitro, CXCR4 signaling has been found to induce endocrine therapy resistance (300). Several inflammatory stimuli are responsible for the expression of particular sets of chemotactic receptors in cancer cells (371), and more recently we have started to learn about the cooperation between diverse chemotactic receptors driving the metastatic phenotype as in the case of induction of CXCR2 expression via CXCR4 activation in breast cancer (336). The complexity in the chemokines receptor/ligand networks calls for a better understanding of the role of inflammatory signals modulating the evolution of the WHFC triad to develop better therapeutic strategies. Cyclo-oxygenase-2 The enzyme cyclo-oxygenase-2 (COX-2) converts arachidonic acid to prostaglandin endoperoxide H2 (PGE2), and its expression triggers an inflammatory response (6). Nevertheless, the function of COX-2 and PGE2 seems to be contradictory and perhaps cell type specific during chronic fibrotic conditions. For example, PGE2 inhibits hepatic fibrosis (23) as well as pulmonary fibrosis where it has been shown to limit fibroblast proliferation and myofibroblast differentiation (37, 187). Also, although COX-2 is expressed briefly in response to tissue injury (131), the use of COX-2 and prostaglandin inhibitors appears only to impart a modest effect over wound healing (131, 244). Even though these types of inhibitors block inflammation, they exacerbate injury since they prevent wound healing especially in the intestine (354). Conversely, COX-2 inhibition reduces renal fibrosis (129, 302), suggesting that PGE2 may contribute to fibrosis in the kidney. Anti-inflammatory COX-2 inhibitory therapy in combination with chemoprevention and as adjuvant to chemotherapy shows some therapeutic promise (178). The role of COX-2 has been particularly well studied in colon cancer. However, COX-2 inhibition as a therapy may require more investigation, as prolonged COX-2 inhibition promotes drug resistance, resulting in increased intestinal fibrosis and tumor recurrence in mice (44, 69). Intriguingly, the induced drug resistance is believed be mediated by intestinal myofibroblasts, as chronic COX-2 inhibition promotes TGF-␤-induced myofibroblastic differentiation and augmented stromal PGE2 synthesis (44, 69). Moreover, TAFs from colon adenocarcinomas express a variety of factors that also drive tumor progression (188, 231). Hence, it is possible that the persistent myofibroblastic population maintained by chronic COX-2 inhibition is responsible for tumor recurrence as well as its correlation with tumor progression (178). Peroxisome Proliferator Activated Receptor Gamma While the previously discussed proinflammatory molecules seem to promote the WHFC triad, the role of the peroxisome proliferator activated receptor gamma (PPAR-␥), a transcription factor involved in immunomodulation, is notable for its inhibitory effects over fibrosis and cancer. During the late stages of wound healing, PPAR-␥ activation stalls fibroblast proliferation and overthrows the myofibroblast phenotype (248), suggesting a mechanism for the termination of the healing response. PPAR-␥ agonists are recognized for their inhibitory effects on TGF-␤-induced myofibroblast differentiation (31, 41, 202, 239, 241, 252, 380). This antifibrotic effect

Physiol Genomics • doi:10.1152/physiolgenomics.00158.2013 • www.physiolgenomics.org

Downloaded from on November 30, 2014

ronment in colon cancer not only by recruiting inflammatory cells but also by recruiting myeloid-derived suppressor cells into the tumor milieu and therefore obstructing antitumor immune responses (170). The broad spectra of CXCRs/CXCL8 effects on cancer progression remains unclear, and yet CXCL8 expression has been proposed as a chemotherapy effectiveness marker (104). Additional chemotactic axes relevant to the WHFC triad, which present contentious roles in fibrosis, are CXCR3 and their relevant ligand chemokines such as CXCL10 (411). Activity of CXCL10/CXCR3 promotes fibroblastic maturation, and it also inhibits endothelial cell proliferation and migration into the wound, a crucial step for wound tissue remodeling and maturation (21). Since CXCR3 activity has been associated with myofibroblastic differentiation and ECM maturation, it is expected that CXCR3-associated chemokines will be implicated in fibrosis. Indeed, in liver, CXCR3 is upregulated and is believed to play a profibrotic role in chronic hepatitis C (414). In fact, serological levels of CXCR3-associated chemokines have been proposed to help monitor the progression and complications of chronic liver diseases (352). In contrast, studies in chronic pancreatitis have proposed an anti-fibrotic role for CXCR3 and its associated chemokine CXCL9 via downregulation of Col-1-␣1 and TGF-␤ (329). Additionally in the pancreas, the blockade of CXCL10/CXCR3 promotes a fibrotic condition in renal disease by upregulation of TGF-␤ (262). Disparities in the CXCR3 activity on different organs during a fibrotic response suggest a more complicated interplay of immune cells for a balanced regulation between pro- and antifibrotic cytokines/chemokines, which needs more investigation. CXCL10/CXCR3 plays important roles in cancer as well. For example, in murine models of breast cancer, hypoxia inducible factor (HIF)-1␣ mediates the expression of CXCL10 and CXCR3 in mesenchymal stem cells and breast cancer cells, respectively, promoting metastasis (50). Moreover, in melanoma murine models as well as in colon cancer patients, CXCR3 promotes metastasis to lymph nodes (172, 173), and recent findings have demonstrated a synergism between CXCR3 and CXCR4 to promote metastasis to liver and lungs (258). Perhaps the best-known chemokine reported in the WHFC triad is CXCL12, also known as stromal-derived factor (SDF)-1, which together with its main receptor CXCR4 induces migration of endothelial cells during wound-induced angiogenesis (411). The chemotactic properties of CXCL12 have also been documented during mobilization of fibrocytes from the bone marrow to the lungs (284), which in concert with other chemokines contribute to aggravate pulmonary fibrosis (230). Moreover, the SDF-1/CXCR4 axis has been shown to trigger prostate myofibroblastic differentiation in vitro, suggesting a role in fibrosis (109). In cancer, the SDF-1/CXCR4 axis has been implicated in the progression of several types of malignancies, and its expression is associated with poor prognosis (66, 132, 387, 393, 395). An important study almost a decade ago demonstrated that SDF-1 secreted by TAFs (but not normal fibroblastic cells) triggers both angiogenesis and tumor growth via CXCR4 (275). SDF-1/CXCR4 activity is known to induce EMT and promote cell motility, invasion, and metastasis (299, 336), demonstrating a strong relationship between stromal and tu-

227

Review 228

THE WHFC TRIAD

CELL PROLIFERATION: A COMMON AVENUE USED BY THE WHFC TRIAD

A hallmark of developmental growth but also of wound healing is a transient hyperproliferative wave affecting both epithelial and stromal cells (183). This feature results from a temporary burst of diverse mitogenic signals triggered by a disturbance in the homeostasis of a given tissue (i.e., during wound healing). Expectedly, a similar spectrum of mitogens can also be constitutively detected in fibrotic and tumor tissues (Fig. 1), promoting epithelial and mesenchymal cell proliferation. Growth factors have a preponderate role among the large variety of mitogenic signals at the WHFC triad (Supplemental Table S1). Here we emphasize some of them. Platelet-derived Growth Factor One of the most abundant molecules released in response to tissue injury is the platelet-derived growth factor (PDGF) (227). PDGF is a key player in the persistence of chronic fibrosis, exerting a strong mitogenic signal upon fibroblasts and myofibroblasts (32). Also, PDGF can stimulate the proliferation of epithelial cancer cells in different types of tumors, exerting its influence through both paracrine and autocrine mechanisms (133, 287). Hence, PDGF-induced cell proliferation could very well sustain a desmoplastic reaction while supporting tumor progression, thus fueling a protumorigenic vicious cycle (20). Some studies suggest that PDGF could sustain or even initiate desmoplasia in breast cancer (148, 326), while others point to the fact that PDGF is a promigratory factor (67) as well as an EMT regulator (388). Furthermore, PDGF expression can also affect other growth factors, as inhibition of stromal PDGF receptors in a murine model of cervical cancer was shown to alter the levels of expression of the fibroblast growth factors (FGF)-2 and -7 (287). Moreover, the inhibition of FGF-2 and FGF-7 in cervical cancer restricts their respective mitogenic and angiogenic effects, suggesting PDGFR as an attractive therapeutic target (287).

FGF-2 FGF-2, also known as basic fibroblast growth factor, encompasses a pivotal role during wound healing (220). FGF-2 exerts a robust mitogenic effect upon fibroblasts, and its participation has been linked to multiple fibrotic disorders (26, 152, 162, 177, 344 –346). In addition, FGF-2 can be induced by TGF-␤ (see above), suggesting an important correlation between the two molecules (177, 345). Nonetheless, FGF-2 has also been reported to inhibit TGF-␤-induced myofibroblast differentiation (92, 150, 154, 234, 339). Interestingly, during nonpathological wound healing, FGF-2 inhibits excessive scarring by triggering myofibroblastic apoptosis, yet quiescent (i.e., nonactivated) fibroblasts appear to be refractory to this molecule (1, 339). These observations suggest that, under physiological conditions, FGF-2 may play a key regulatory role during wound resolution, encouraging fibroblast proliferation to repair damaged tissue but restricting the myofibroblastic population to limit excessive scarring. Nevertheless, FGF-2 effects have been implicated in the development of several tumors by promoting proliferation and survival of cancer cells and even supporting tumor angiogenesis (368). It is not surprising that the effect of FGF-2 and the FGF receptor family (FGFR) in cancer is considered “complex” as the pathway has been shown to have both protumorigenic and tumor-suppressive roles (368). What is more, FGFR-induced activities can be ligandindependent (via genomic mutations of signaling molecules) or ligand dependent (368). Although the role of FGF-2 in desmoplasia remains rather obscure, its detection and implication at the stroma of prostate and mammary cancers as well as other malignancies (223, 367, 403) suggests a fairly broad participation (267). Epidermal Growth Factor Receptor The epidermal growth factor receptor (EGFR) family, also known as the ErbB family, consists of pleiotropic membranespanning cell surface receptors with intracellular tyrosine kinase activities that are triggered by homo- or hetero-dimerization when engaged by growth factors. Some of the family growth factor ligands are known to participate in wound healing [i.e., epidermal growth factor (EGF), heparin-binding epidermal growth factor (HB-EGF), and TGF-␣] (281, 409). Under physiological conditions, EGFR signaling drives wound healing, and controlled activity of EGFR by HB-EGF has been proposed as a therapeutic approach to prompt and accelerate cutaneous wounds healing (161, 363). Of note, aberrant EGFR activation is known to promote fibrosis in tissues such as lung, heart, and pancreas (128, 214, 229, 245). EGFR activity has been implicated in the progression of malignancies such as colon and lung cancer (94, 134, 314). Nevertheless, EGFR activity has also been implicated in tissue regeneration during acute and chronic colitis where EGFR activity seems to limit fibrosis induced tumorigenesis (80). Then again, EGFR signaling activates EMT in malignant cells (130, 312). Doubly, EGFR activity affects the tumor stroma by inducing HB-EGF overexpression in TAFs, thus supporting tumor progression by direct and indirect effects (259).

Physiol Genomics • doi:10.1152/physiolgenomics.00158.2013 • www.physiolgenomics.org

Downloaded from on November 30, 2014

has been proposed as a strategy to attenuate pulmonary and renal interstitial fibrosis (174, 194, 332). Then again, in cancer, some investigators have proposed an oncogenic, while others have stated a tumor-suppressive, role for PPAR-␥ (124). For example, PPAR-␥ agonists have been shown to impart anticancer effects (22), while a tumor-promoting effect is observed when PPAR-␥ is inhibited (236) (see Supplemental Table S2). Nevertheless, PPAR-␥ signaling has also been shown to exert an inhibitory effect on TGF-␤-induced EMT in lung cancer cells and subsequent metastasis (297). Even more, PPAR-␥ ligands can also induce EMT upon intestinal epithelial cells through a Rho-dependent activation of ERK1/2 (51), suggesting a tight regulation of the PPAR-␥ signaling effects that could be associated with a particular cell type in a particular TGF-␤ signaling context. Interestingly, there is evidence to suggest an intricate regulation of the tumor stroma in colon adenocarcinomas, where COX-2 and PPAR-␥ have been found coexpressed only in myofibroblasts at the tumor-associated but not the normal stroma (372). These findings suggest a PPAR-␥ negative regulation of both fibrosis and cancer where its presence at late stages of nonpathological wound healing possibly contributes to prevent the WHFC triad occurrences.

Review THE WHFC TRIAD

229

Additional Deregulated Developmental Signals Promoting WHFC

ergistic interaction could constitute an interesting therapeutic approach.

The healing of a wounded tissue is known to be influenced by signaling molecules involved in embryonic development (183). Some of these signaling molecules are highly relevant during wound resolution and, when deregulated, are major players in the WHFC triad. To this end, Notch and Hedgehog (Hh) signaling cascades constitute two well-known examples of developmental signaling implicated in these processes (see Supplemental Table S1). Notch signaling is an evolutionary well-conserved pathway known for regulating embryonic morphogenesis, tissue homeostasis, angiogenesis, and maintenance of the vascular system (25). The role of Notch during wound healing has been demonstrated in vitro as well as by genetic manipulation of murine models where Notch-deficient animals present delayed wound healing (54, 77, 228). Regarding its mechanisms of action, it is known that Notch signaling drives EMT, providing a vast source of fibroblastic cells necessary for wound repair (171). In addition, Notch activity has also been associated with fibrosis promotion in a variety of tissues (such as skin, kidney, lung, and cardiac tissue), influencing cell proliferation, extensive fibrilogenesis, TGF-␤ production, and ␣-SMA expression (171). In cancer (but not in regenerating epithelium during wound healing), Notch has also been shown to have an oncogenic role, promoting protein biosynthesis, cell growth, apoptosis resistance, and angiogenesis (305). Moreover, it has also been implicated in triggering EMT (224), fueling cancer invasion and enriching the desmoplastic TAF population. Nevertheless, there is evidence to suggest that Notch, depending on the cellular context, can exert a tumor suppressive role (224). Another important signaling pathway in the WHFC triad is the Hh cascade. The Hh signaling pathway is triggered by several types of Hh ligands (i.e., Sonic, Desert, and Indian). The ligands bind to the receptor Patched, which in turn releases Smoothened to promote Gli transcription factors (Gli-1, Gli-2, and Gli-3) (58). Gli-1 appears to be the most abundant Hhactivated transcription factor, while Sonic Hh (SHh) is the better characterized ligand in the signaling pathway (58). SHh signaling is essential for wound healing as demonstrated by the inhibitory effect imparted by the topical application of a cyclopamine, a well-characterized inhibitor of smoothened, at all stages of the wound healing process (200). However, SHh signaling has also been shown to promote fibrosis in several organs such as lung, liver, and skin (157). Likewise, Hh has been shown to participate in tumor progression (58) by promoting stromal activation/desmoplasia (149, 359). For example, in pancreatic as well as in prostate cancer, it has been found that SHh is released by cancer cells, affecting stromal cells, causing differentiation and proliferation of ␣-SMA-positive myofibroblastic TAFs (15, 149, 199, 328, 376). Besides its ability to activate the tumor stroma, SHh ligands also promote EMT in cancer cells (396). Interestingly, while SHh signaling facilitates tumor progression through multiple mechanisms, it is possible that SHh ligands are not the only possible triggers of SHh signaling, as it has been found that TGF-␤ can also induce the activity of Gli-1 (157). This signaling pathway overlap strongly suggests a common participation in the WHFC triad. Although the relationship between SHh signaling and TGF-␤ activation merits additional investigation, the syn-

PATHOLOGICALLY ALTERED ECMS CONSTITUTE BOTH CAUSE AND CONSEQUENCE OF CHRONIC FIBROSIS AND TUMOR PROGRESSION

Below we discuss the participation of physical ECM (i.e., architecture or stiffness) properties, as well as noteworthy components of pathological ECM common to the WHFC triad. Topographical/architectural Changes in ECM

ECM Stiffness Both fibrotic and tumor-associated (i.e., desmoplastic) ECMs are characterized by an increase in stiffness compared with their normal stromal equivalent, which is known to be more flexible or compliant (116, 208, 277, 364). A less flexible environment exerts a greater physical tension to the cell body, which is transduced into multiple intracellular signals, modifying cell behavior. For example, relatively stiff ECMs alter cell growth, modify gene expression, and alter the differentiated status of cells (292, 338, 378). Moreover, ECMs subjected to a higher physical tension induce structural modifications to ECM-associated molecules, modulating their bioactivity (i.e., releasing active molecules of TGF-␤), which in turn, for instance, can trigger myofibroblastic differentiation (139) and also further fuel the production of the altered (166) and stiffer ECM. Hence, in chronic fibrosis, tissue stiffness fuels a positive feedback enhancing the chronicity of fibrotic disease (218). Meanwhile in cancer, a rigid ECM induces tumor progression (42, 208, 282, 410) but also an enrichment of activated/myofibroblastic cells (i.e., TAFs), which in turn sustain a desmoplastic reaction and further promote tumorigenesis (16, 20, 64, 116, 399). ECM rigidity regulates the effects of TGF-␤ upon tumor cells, as compliant matrices facilitate TGF-␤-induced apoptosis, while stiffer matrices promote TGF-␤-induced EMT (206). Various ECM-related modifiers and a plethora of molecular components participate in the WHFC triad, and a few are listed below as well as in Supplemental Table S1. Fibronectin A native well-conserved constituent of the mesenchymal ECM that plays a determinant role in the WHFC triad is

Physiol Genomics • doi:10.1152/physiolgenomics.00158.2013 • www.physiolgenomics.org

Downloaded from on November 30, 2014

A pathological mesenchymal ECM differs from its normal counterpart and is recognized by an altered and constitutive expression of its building molecules, as well as for their characteristic architectural organization (294, 295), which together are believed to impart distinct physical and biochemical properties to the ECM (97, 410). Moreover, the altered architectural composition of a pathological ECM has been associated with clinical outcomes in breast cancer (59). Functionally, it has been suggested that topographical ECM alterations can impact normal stromal cells’ behaviors, thereby supporting a phenotypic switch (i.e., myofibroblastic activation) (4, 139). Concurrently, these newly activated cells continue synthesizing the above-mentioned altered ECM, feeding a vicious cycle affecting additional adjacent stromal (4, 16, 115, 137), as well as epithelial cells (16, 45, 97, 116, 197, 201), in a continuously expanding “field effect.”

Review 230

THE WHFC TRIAD

fibronectin (FN). Plasma FN (307) is commonly found in the circulation, while cellular FN (400) is the glycoprotein that is expressed by fibroblastic cells and is incorporated into the fibrillar ECM mesh. From a single human gene and as product of alternative mRNA splicing, several cellular FN variants can be expressed (384). Two spliced exons called “extra domains A” and “B” (EDA and EDB) are particularly and abundantly expressed during embryonic developmental stages, while an important (i.e., normal) reduction is observed in adult tissues (384). Nevertheless, during the physiological ECM restructuring of wounded tissue, EDA-FN is upregulated by myofibroblastic cells (118). Moreover, in a positive feedback regulation, EDA-FN has been shown to exert a permissive action during myofibroblastic activation (82). EDA-FN overexpression has been found to occur in response to increased ECM tension (364), and it is influenced by high levels of TGF-␤ at the specific microenvironment of acute injuries (324). The relationship of EDA-FN with TGF-␤ appears to be a determinant for the progression of several fibrotic diseases (i.e., atherosclerosis and lung and liver fibrosis) as demonstrated in various in vivo FN mutant animal models (384). Researchers believe that the sustained TGF-␤-promoted myofibroblastic differentiation occurs via production and signaling of EDA-FN, supporting a chronic fibrotic niche (384). EDA-FN also influences the tumor microenvironment, affecting both stromal and tumor cells. At the stromal compartment, EDA-FN seems to be intimately related to the bioavailability of active TGF-␤ for the subsequent sustainability of the reactive stroma (384). EDA-FN is known for promoting tumor growth, EMT, and tumor-induced lymphangiogenesis (349, 391). EDB-FN has also been associated with tumor progression, in particular with tumor angiogenesis and is being tested as a potential tumor target in the clinic (320). Hence, alternative spliced forms of FN seem to play important roles in the WHFC triad.

tic reaction (12). For example, it was reported that pancreatic cancer cells can trigger fibroblastic activation while promoting increased deposition of Col-I and -III in a nude murine model (12). In breast cancer, Col-I cross-linking is increased, showing a reduction on its degradation rate, resulting in excessive deposition (168). In sum, Col-I and -III changes are typically described in diseases associated with the WHFC triad.

Collagen

Even though, intuitively, MMPs, are ECM-cleaving proteinases, they do not antagonize fibrosis, cancer, or fibrosisinduced cancer. Hence, these enzymes are regarded as ECM remodeling proteins as opposed to ECM-degrading proteins (278). MMPs play a crucial role during inflammation, reepithelialization, and ECM remodeling associated with regenerative wound healing (113). Specifically, MMPs mediate ECM remodeling during scar resolution, cleave and activate ECM-associated growth factors, regulate chemokine activity, and also facilitate keratinocyte migration into the wound (113, 176, 278). MMPs can be expressed by inflammatory cells, epithelial cells, fibroblasts, and myofibroblasts. The sources and functions of MMPs implicated in the context of cancer (176) and fibrosis (278) are very diverse and beyond the scope of this review. Nonetheless, we highlight two gelatinases, MMP-2 and MMP-9 (19, 176), as these enzymes seem to convey a recurrent presence in the WHFC triad. The participation of MMP-2 and MMP-9 in chronic fibrosis appears to be contentious. For example, a clear upregulation of these MMPs expressed by myofibroblasts is evident during the development of idiopathic pulmonary fibrosis (322). Also, MMP-2 has been found to be highly expressed in fibrotic liver myofibroblasts (293), which incidentally has been proven to constitute a noteworthy predisposition to developing hepatic cell carcinoma (86). Nonetheless, a downregulation of MMP-2 has been reported during in vitro myofibroblastic differentia-

An important regulator of ECM rigidity is the activity of the enzyme lysyl oxidase (LOX), which catalyzes the cross-linking of collagen (see above) and elastin bundles, modifying the tensile strength of the ECM (165). LOX is essential for maintaining ECM strength during normal development (233) and becomes highly expressed during the course of several fibrotic diseases (164, 392). Throughout cancer progression, LOX facilitates Col-I cross-linking, promoting ECM stiffening and therefore altering known integrin-regulated signaling pathways (208). In fact, increased LOX activity has been associated with a poor prognosis in several carcinomas (392). LOX and LOXlike enzyme 2 (LOXL2) are downstream targets of HIF-1␣ and, under hypoxic conditions, promote EMT by repressing E-cadherin (319). In a xenograft tumor model, the inhibition of LOX2 decreased the number of ␣-SMA-positive TAFs (17). Furthermore, LOXL2 has been proposed as a prognostic biomarker for squamous cell carcinomas (283). Together these data support the correlation between increased cross-linked Col, stiffer ECM, an enriched TAF population, and the exacerbation of tumor desmoplastic reaction. Interestingly, the same LOX inhibitory antibody hinders murine liver and pulmonary fibrosis (17), suggesting a compelling similarity as proposed in the WHFC triad (Supplemental Table S2). MMPs

Physiol Genomics • doi:10.1152/physiolgenomics.00158.2013 • www.physiolgenomics.org

Downloaded from on November 30, 2014

As the major ECM component in the connective tissue and the most abundant protein in mammals, collagen has been described as a heterotrimeric triple helix glycoprotein that can be assembled into de novo fibrous matrices during healing, playing a crucial role during the repair of tissue defects. Collagen deposition increments the ECM tension during the restoration of the vessels network and the recovery of tissue architecture during wound healing (118). Among the collagen family, Col-I is the most abundant interstitial protein of the ECM (107) often seen in fibrotic tissue and desmoplasia. Then again, Col-IV is the main component of epithelial and endothelial basal membrane (167). The activated/myofibroblastic phenotype seen in the WHFC triad is commonly described as containing abundant Col-I and -III, although reports showing changes in Col-IV and -V are also evident (136). In addition, Col-VI spiked some attention due to its selective expression in some fibrotic conditions (i.e., kidney and myocardial fibrosis) as well as in hepatocellular carcinoma (136). In fact, its level of expression has been recently proposed to serve as a clinical prognostic marker (204, 205). Carcinomas that typically present an aggressive progression (i.e., liver and pancreas) are characterized by a large deposition of interstitial FN and collagen. These occurrences are blamed for augmented tumor cell proliferation that is accompanied by an intense desmoplas-

Lysyl Oxidase

Review THE WHFC TRIAD

tion of rat embryonic fibroblasts (141), while its inhibition has been shown to accelerate murine renal fibrosis (268). In cancer, MMP-2 and MMP-9 are known to promote tumor progression by facilitating the release of stored latent growth and other factors (i.e., TGF-␤) from the ECM, thus enhancing respectively cell proliferation and migration (36). MMPs can also facilitate cancer cell migration (385) by interaction with cell-matrix adhesion molecules, such as integrins, promoting degradation of cancer cells from cell-cell and cell-matrix adhesions (36). To further fuel the recurring cycle, EMT enhances the expression of MMP-2 and MMP-9 (36). Interestingly, MMP cleaved ECM peptides (i.e., soluble ECMs) are known to induce an integrin-dependent TGF-␤-like signaling response (106). Also, CD147, a cell-surface glycoprotein upregulated in tumor cells, can stimulate both stromal MMP production and myofibroblast differentiation (147). In sum, MMPs produced by cancer cells promote tumoral invasion as well as myofibroblast differentiation, reinforcing a tumorsupportive stroma, which in turn further secretes and activates additional MMPs to further fuel this vicious cycle.

A particular component of the ECM, with no inflammatory activity but related to inflammation, is the glycosaminoglycan hyaluronan (HA). This macropolysaccharide molecule contains multiple unit repetitions of N-acetyl-glucosamine as well as of glucuronic acid, and it binds and stores large amounts of water molecules conforming a viscous gel (355). HA has been suggested to function as a pliable substrate for tissue remodeling necessary for wound healing (355). It also binds fibrinogen, supporting clotting, and acts as a mesh that traps inflammatory cells (342). Large HA molecules of ⬃25,000 disaccharide units (355) encourage cellular quiescence, while small HA fragments ranging from 4 to 25 units encourage proliferation and angiogenesis (342). It has been proposed that HA breakdown at the injury site renders fragments that participate in the initiation of the healing response (342). HA accumulation is associated with exacerbation of fibrosis (27, 159). The expression of HA synthesizing enzymes “hyaluronan synthase (HAS) 1–3” during chronic fibrosis has been associated with an increased inflammatory reaction (52, 213). Knockdown of HAS2 reduces the effectiveness of TGF-␤stimulated EMT (291), and elevated HAS2 expression is sufficient to induce EMT in normal epithelial cells (418). Moreover, inhibition of HA synthesis antagonizes TGF-␤-induced myofibroblastic differentiation (381), and inhibition of HAS2 impairs myofibroblast accumulation in mouse models of pulmonary fibrosis (213). Since elevated HA has been observed in a variety of fibrotic diseases (210, 213, 357) and HAS2 regulates TGF-␤-dependent functions (291), their accumulation and increase in expression are respectively suggestive (i.e., markers) of fibrotic myofibroblast differentiation and EMT. With regards to cancer, increased HA is linked to poor cancer outcomes (355), while upregulation of HAS2 is a common occurrence in highly metastatic breast cancer cells (273). Nevertheless, the role of HA in cancer seems to be tissue specific. In general, accumulation of HA in tissues where it is normally absent facilitates tumor growth (i.e., gastric cancers), while reduced HA is associated with cancer in normally HArich stratified epithelia containing tissues such as skin, mouth,

and larynx (355). Interestingly, epithelial HA accumulation in colon cancer constitutes a bad prognostic occurrence (304), while stromal HA buildups have been associated with an unfavorable prognosis in ovarian, prostate, breast, and nonsmall cell lung cancers (11, 217, 289, 382). In the case of liver, the main organ in charge of processing HA (98), a large synthesis of the glycosaminoglycan and increasing circulating levels of HA have been found to correlate with liver fibrosis (cirrhosis) (280). In addition, hepatic cirrhosis is known to predispose for hepatocarcinoma, and HA deposition has also been associated with cancer progression (285, 341). Most of the mechanisms of HA’s driven tumor progression deal with the promotion of cell proliferation, NF-␬B activity, and angiogenesis (333). HA also binds to the cellular receptor CD44, which encourages motility and invasion (333). Intriguingly, HA expression is increased via growth factors such as EGF, keratinocyte growth factor, and PDGF (355), suggesting a more complicated role for certain growth factors, which could promote both cancer cell proliferation and HA accumulation. Interestingly, the antiproliferative and proapoptotic effect of HA inhibition, affecting both the tumor and surrounding stroma (285), highlights the importance of designing anticancer strategies focused on attacking both the tumor and its associated fibrotic stroma. Periostin Periostin is a matricellular protein that increases ECM stiffness by direct binding to fibrillar ECM proteins such as Col-I (270) and by interacting with the TGF-␤-related protein BMP-1, which is needed to activate LOX (240). In addition, periostin functions as a ligand for integrins ␣v␤3, ␣v␤5, and ␣6␤4 (102, 306). In general, periostin expression seems to play a main role during wound healing and tissue regeneration (reviewed in Ref. 192). For example, periostin-deficient mice present a reduction in the number of cutaneous wound healing/ repair myofibroblasts (88). Periostin has been associated with myofibroblastic differentiation in vivo and in vitro via an integrin-dependent mechanism as well as with ECM remodeling (60, 87). As a consequence, periostin increases are also associated with pathologies marked by a fibrotic condition such as idiopathic pulmonary fibrosis, renal fibrosis, asthma, and skin sclerosis in scleroderma (261, 323, 331, 401). Regarding cancer, periostin has been found to be expressed by cancer epithelial cells (255, 306), and recently it has been associated with increased esophageal cancer invasion of p53 mutants via STAT1 induction (386). Moreover, periostin can also be expressed by stromal cells and, as such, has been identified in desmoplastic pancreatic cancer (102), colon cancer, cholangiocarcinoma (179, 369), and other types of cancer (193, 222, 249, 255, 306). Consequently, periostin seems to facilitate both tumoral and stromal progressions. Periostin integrin-dependent signaling activates focal adhesion kinase (FAK), as well as the PI3K/AKT pathway, thus generally promoting malignant cell survival, proliferation, and angiogenesis (255, 306). Moreover, activation of integrins by periostin promotes EMT in cancer cells (255). Intriguingly, in bladder cancer, periostin seems to function as a tumor suppressor by in vitro inhibition of invasion and reduction of in vivo metastasis (180). Moreover, in many cancer cell lines in vitro periostin seems to suppress anchorage-independent growth, while low

Physiol Genomics • doi:10.1152/physiolgenomics.00158.2013 • www.physiolgenomics.org

Downloaded from on November 30, 2014

Hyaluronan

231

Review 232

THE WHFC TRIAD

periostin mRNA levels are observed in lung cancer in vivo (408). Tenascin C

Osteonectin/SPARC The secreted protein and rich in cysteine (SPARC), also known as osteonectin, is a matricellular protein that is known to affect ECM remodeling/dynamics. SPARC is often highly expressed during wound healing, cancer, and fibrosis (18, 55, 365). For example, the upregulated expression of SPARC seems to be a common event during several chronic fibrotic conditions such as in the cases of heart, lungs, kidneys, liver, dermis, intestine, and eyes (365). SPARC is produced by fibroblasts in response to TGF-␤, but it can also promote Col-I and TGF-␤ synthesis (365). In a SPARC null murine model, a decreased deposition of dermal collagen constitutes a predominant trait accompanied by an enhanced contractile behavior of dermal fibroblasts (39). By affecting collagen deposition and reducing the number of myofibroblasts, decreased SPARC expression reduces liver fibrosis (10). Nonetheless, despite the implication of SPARC in the control of wound healing, its precise role remains rather unclear. For instance, it has been reported that SPARC can both induce and inhibit the wound healing process (18, 39). Similarly, SPARC has also been implicated in tumor progression where it is believed to sometimes promote, and at other times prevent, this process apparently in a tissue- and cell-specific manner (55). Among other functions, SPARC participates both in carcinoma EMT and in TAFs’ myofibroblastic differentiation, thus promoting both tumorigenesis and desmoplasia (55). In prostate cancer, for example, the expression of SPARC by malignant cells can be predictive of metastasis, while in pancreatic and nonsmall cell lung cancers, elevated expression of SPARC at the tumor stroma indicates poor prognostics (29, 74, 189). Regarding SPARC’s tumorsuppressing effects in breast, ovarian, colon, and bladder carcinomas, SPARC has been associated with inhibition of proliferation and induction of apoptosis (55, 309). Intriguingly, a positive feedback between SPARC and TGF-␤ has been observed, as both can induce the expression of each other (365). Nevertheless, a comprehensive comparative proteomic analy-

Proteoglycans Comprising a diverse group of members, the proteoglycan family includes both cell surface proteins, such as heparan sulfate proteoglycans, and ECM components, such as small leucine-rich proteoglycans (SLRPs), and large chondroitin sulfate proteoglycans (153). Proteoglycans participate in immunity regulation (57, 99), ECM assembly (53), and cancer modulation (153). However, as a complete description of proteoglycans is beyond the scope of this review, we highlight a selection of ECM proteoglycans most relevant to the WHFC triad. Versican, a large chondroitin sulfate proteoglycan, is upregulated during both wound repair and cancer progression (63, 112, 301, 353). During normal conditions, myofibroblasts from human granulation tissue express elevated levels of versican (126). There is a lack of studies investigating the role of versican in the context of chronic fibrosis. However, the expression of versican mRNA has been detected as increased in rat lungs after bleomycin-induced injury, and TGF-␤ signaling increases versican synthesis in fibrotic lung in vivo (373). On the other hand, versican expression correlates with a poor prognosis in many cancer types (301, 325). This proteoglycan interacts with multiple ECM and cell surface components, promoting cell proliferation, cell motility, and metastasis (301, 310, 382). Intriguingly, in breast cancer, versican has been associated with chemotherapy resistance related to EGFR hypersignaling and enhanced cancer cell self-renewal (78, 79). Moreover, versican synthesis appears to be upregulated by TAFs in the ovarian cancer stroma (407) where TGF-␤ signaling triggers versican expression, and together they contribute to promote ovarian cancer aggressiveness (407). The SLRP biglycan expression is enhanced in myofibroblasts from human granulation tissue (126). The role of biglycan in fibrosis may be tissue specific since biglycan does not affect TGF-␤-induced pulmonary fibrosis (186), and biglycan mRNA appears not to be increased in mercuric chlorideinduced renal tubulointerstitial fibrosis (350). However, biglycan expression and serological levels of its processed byproducts correlate with the extent of liver fibrosis in murine models (108, 204). Biglycan is also involved with progression and poor prognosis in a variety of malignancies such as gastrointestinal and endometrial cancers (7, 122, 221, 377, 415). In contrast, biglycan expression correlates with a favorable prognosis in bladder cancer where it seems to have an inhibitory effect on tumor cell proliferation (266). In addition to biglycan, the SLRP decorin also plays an active role in the WHFC triad, although its participation in chronic fibrosis and tumor progression seems to have an inhibitory effect. Animal models have shown that absence of decorin exacerbates fibrosis (13, 247, 316). Moreover, interesting data suggest that decorin itself could be a beneficial

Physiol Genomics • doi:10.1152/physiolgenomics.00158.2013 • www.physiolgenomics.org

Downloaded from on November 30, 2014

Tenascin C is another ECM-related glycoprotein expressed in response to tissue injury known to modulate the wound healing response (251). Tenascin C levels decrease subsequently to wound healing resolution (251). This decrease in tenascin C is also associated with a natural reduction in myofibroblasts (251). Hence, it is not surprising that a misregulated and continuous expression of tenascin C is a common occurrence, which is believed to drive fibrosis and cancer via increased angiogenesis, ECM alterations, and immunomodulation (250). For example, tenascin C-deficient mice are protected from pulmonary and liver fibrosis (43, 85). In addition, tenascin C has been shown to promote tumorigenic progression by supporting cell motility, metastasis, and more (101, 127, 207, 274, 279). Tenascin C is synthesized by both tumoral and stromal cells (71, 276). Then again, tenascin C could also indirectly promote the WHFC triad by its effect on the downregulation of tPA, which facilitates fibrin accumulation, thus triggering a mitogenic response (40).

sis has demonstrated that not all the cellular responses to SPARC signaling depend on TGF-␤ (114), and, although there is a certain parallelism between them promoting neoplasia (242), a balance of both SPARC and TGF-␤ activity could be responsible for tumor progression (114). These facts suggest that an intricate regulation of the SPARC/TGF-␤ signaling axis also constitutes a common occurrence of the WHFC triad (Supplemental Table S1 and Fig. 1).

Review THE WHFC TRIAD

CHRONIC FIBROSIS AND CANCER DIFFER FROM TRANSIENT WOUND HEALING IN THAT THEY BOTH ENCOMPASS A SUSTAINED MYOFIBROBLASTIC POPULATION

While regenerative healing wounds, fibrosis, and tumorassociated desmoplasia all contain metabolically active myofibroblasts and EMT cells, it is only under normal healing/ regenerative (i.e., nonpathological) conditions that the activity of these cells eventually ceases. This may result from activated cells’ (i.e., EMT or myofibroblasts) senescence and subsequent elimination. This can occur through p53- and/or integrinregulated apoptosis or potentially by cell clearance via natural killer (NK) cells and/or by interferon gamma secretion (146, 181, 198, 209, 360, 398). From the one side, myofibroblastic apoptosis could promote pathologic scarring (75). Then again, the persistence of myofibroblasts during fibrosis and cancerassociated desmoplasia suggests that these cells escape from their natural elimination process by alterations in the myofibroblastic cells-clearance mechanisms. Alternatively, persistent activated cells may “sense” chronic injury or tumorassociated microenvironment (i.e., desmoplasia) and consistently respond “normally.” In addition, fibroblastic cells naturally progress and become senescent in response to a variety of signals. Nevertheless, under pathological circumstances when clearance mechanisms (i.e., wound resolution) are evaded, cells can develop what is known as the “senescence associated secretory phenotype” or SASP, thus turning them into sustained and proinflammatory senescent cells (61). In response to SASP, affected fibroblastic cells produce a variety of diffusible factors that in turn can drive both fibrosis and tumorigenesis (61),

further fueling the WHFC triad’s fibrosis/tumorigenesis vicious cycle (see Fig. 1). To further complicate things, although SASP seems to fuel the WHFC triad, evidence also suggests that the sole escape of fibroblasts from senescence can also support WHFC. For example, during normal wound healing, the matricellular protein CCN1 (also known as cysteine-rich protein 61 or CYR61) is expressed by myofibroblasts and tends to accumulate along with the myofibroblastic cell population (198). High concentrations of CCN1 induce myofibroblast senescence (198) and promote downregulation of collagen and TGF-␤ as well as upregulation of MMPs (198). Together these changes induce a senescent matrix-degrading cell state (198). During wound healing resolution, myofibroblast senescence serves to prevent pathological scarring (198). Hence, senescence is generally recognized to inhibit fibrosis (163, 190), so it is not surprising then that mice expressing defective CCN1 suffer from exacerbated fibrosis in response to injury (198). Furthermore, recent findings have shown that restoring lost levels of CNN1 in a hepatic fibrosis murine model can induce myofibroblastic senescence and reduce chronic tissue-damaging fibrosis (182). Nevertheless, the role that CCN1 plays in both tumor-associated stroma (i.e., desmoplasia) and in tumorigenesis remains rather obscure. CCN1 has been shown to present opposing effects on tumor progression, as some data indicates that CCN1 inhibits tumor growth by inducing apoptosis and senescence, while other reports suggest it promotes tumor growth via the stimulation of cancer cell proliferation and angiogenesis (198). In addition, it is known that CCN1 induces senescence by activating the p16(INK4A)/pRb pathway (198). Interestingly, TAFs are hyperproliferative cells that express diminished levels of p16(INK4A) (3). Also, p16(INK4A) downregulation in normal human breast stromal fibroblasts is accompanied by an increase in ␣-SMA expression, suggesting that absence of p16(INK4A) could promote myofibroblastic differentiation (3). Additionally, p16(INK4A)-deficient fibroblasts enhance xenograft tumor growth compared with effects imparted by wild-type cells (3). Finally, fibroblasts from oral squamous cell carcinoma with a p53 and p16(INK4A) inactive background have been shown to induce the upregulation of ␣-SMA expression, which correlates with poor patient prognosis (215). All these data strongly suggest that inactive p16(INK4A) is a hallmark of TAFs and, therefore, of cancer progression. Regarding p53, it has recently been shown that stromal p53 decrease in chronic liver damage promotes tumor development by a noncell autonomous mechanism that upregulates a tumor promoting M2 macrophage microenvironmental state (226). Hence, there is strong possibility that at least two distinctive populations of TAFs that drive tumorigenesis coexist at the tumor stroma: a hyperproliferative p16 low population and a rather quiescent SASP positive and presumably p16 high population. This type of TAF heterogeneity reveals a high level of complexity at the tumor-stromal interaction niche, which requires a better understanding to improve the design of increasingly effective stroma targeting therapeutics. If one considers the fact that cell death, for example during injury, causes fibrosis, then another important fact of the WHFC triad is that some types of fibrotic tissues are often characterized by regions of cellular death (219, 269, 288), while, similarly, tumors contain noteworthy areas of necrotic tissue (226). Cell death is well known to activate an inflam-

Physiol Genomics • doi:10.1152/physiolgenomics.00158.2013 • www.physiolgenomics.org

Downloaded from on November 30, 2014

antifibrotic therapeutic alternative (156, 186, 402). A suggested antifibrotic mechanism of decorin deals, at least partially, with the inactivation of TGF-␤ signaling (14), impairing myofibroblastic differentiation in liver fibrosis models (14). Another mechanism described is the direct interaction with the cytokine known as connective tissue growth factor or CTGF, inhibiting its fibroblastic activation role (374). In cancer, decorin gene therapy or delivery of decorin protein inhibits tumor growth, tumor progression, and metastasis (153). Besides the attenuation of TGF-␤ signaling by decorin described in fibrosis models (14, 402), another mechanism of action has been found in the tumor microenvironment where it could also be inhibiting EGFR signaling (153). Decorin binds to EGFR overlapping with the EGF binding domain and inducing internalization of EGFR via caveolar endocytosis, resulting in decreasing EGFR accessibility on the cell surface (153). Together, these effects evoke a downregulation of EGFR signaling in tumor xenografts and apoptosis induction (153). Decorin has also shown additional antitumor effects including the inhibition of angiogenesis and induction of p21 (153, 264). Although reduced decorin expression in the tumor stroma indicates poor prognosis in models of breast cancer (264), decorin can be found in both the tumor stroma and fibrotic tissue (14, 153). This could be explained as a protective response (153), although it has also been observed that collagen-bound decorin found in the ECM could not be effectively protective (14). Future work may seek to clarify the influence of decorin on the myofibroblastic cells during chronic fibrosis and tumor desmoplasia.

233

Review 234

THE WHFC TRIAD

CONCLUDING REMARKS

Possible Approaches to Clinically Interfere With the WHFC Triad Since a convergence between wound healing, cancer, and fibrosis is compelling, its implications for fibrosis and cancer treatment are profound. To achieve effective drug delivery, it is crucial to comprehend the challenge that a fibrotic microenvironment represents. For example, due to a dense and altered ECM deposition, the free access for drugs and other molecules, including glucose and oxygen, is severely impaired in desmoplastic malignancies (65). In this context, it is necessary to develop novel drug delivery strategies to obtain better results while targeting both fibrosis (to obtain stromal normalization) and cancer (see Supplemental Table S2). This is true in particular with regards to carcinomas displaying an intense desmoplastic reaction such as in pancreatic cancer (263). Importantly, it is troubling that current approaches aimed at treating fibrosis and cancer also counteractively promote the loss of homeostatic equilibrium, which in turn further promotes the WHFC triad (Fig. 1). For example, surgical procedures and radiation are effective in resecting and eliminating cancerous and chronically fibrotic masses. Nevertheless, the same treatments also create wounds that must be healed, hence possibly further promoting fibrosis and hyperplastic growth (2, 49, 73). Moreover, while surgery triggers a wound healing response, chemotherapy also activates the same mechanisms that induce fibrosis (49, 272). Above we stated that fibrotic disease fuels cancer progression and vice versa (Fig. 1). Since it is possible that radiation and chemotherapy-induced fibrosis also stimulate relapse or latent escape of secondary tumors, a more optimistic perspective will rely on the use of antifibrotic therapies in cancer treatments. This approach could have a great impact on those malignancies where fibrosis constitutes a clear predisposition, such as in the cases of pancreatic or hepatic cell

carcinomas, where studies have shown promising results targeting and repressing both tumor and stroma (285). These may offer an advantage when used against cancer, while some chemotherapies and targeted cancer treatments could also have great impact on resolving fibrotic chronic diseases. For this we have put together Supplemental Table S2, which lists some of the current common WHFC triad treatments under investigation or in clinical use today. Reflecting Upon the WHFC Triad This review attempts to highlight the importance of understanding wound healing mechanisms, as a pathway to comprehend two subsequent conditions, derived from the persistence and exacerbation of the healing process. In the absence of wound resolution, continuous alterations to tissue homeostasis engage both stroma and epithelia for the onset of chronic fibrosis and/or cancer. We propose the consolidation of commonalities from these three conditions under the concept of the WHFC triad. In other words, similar mechanisms operate in this triad to sustain a vicious cycle where a fibrotic stroma could trigger a malignant behavior of epithelial cells, while the progression of carcinomas also promotes a fibrotic/desmoplastic stroma fueling the conditions for both (Fig. 1). The WHFC triad is assembled by common biological responses from the three described conditions. Several proteins and signaling pathways encompass a remarkable degree of overlap amongst wound healing, pathological chronic fibrosis, and cancer (317). The mentioned players can be clustered based on their biological mechanisms, in a coagulation/fibrinolysis-like response, inflammation signaling processes, as well as ECM remodeling mechanisms. These biological occurrences are responsible for triggering hyperproliferative cell behavior and phenotypic/activating modifications such as EMT and myofibroblastic differentiation (Fig. 1 and Supplemental Table S1). Specifically for cancer, the WHFC triad constitutes a rather fertile ground facilitating genetic mutations and epigenetic hyperplastic occurrences, which eventually constitute the malignant cell population (91). Moreover, an additional difference between chronic fibrosis and cancer is that, although both expand in an immediate field-like effect through the affected tissue, only cancer is known to metastasize to tissues localized at considerable distances from the primary lesion, while secondary neoplastic niches are actively preconditioned to facilitate metastatic cancer cell adaptation (120). Importantly, crucial aspects of the WHFC triad that have already started to receive attention, such as genetic and epigenetic signatures as well as microRNA profiles (24, 404), although in need of being fully understood, are already being proposed to serve as predictors of the outcome of some fibrosis-related malignancies. Hence, we conclude by stating that a better recognition and a deeper understanding of the commonalities and disparities within the WHFC triad will surely provide insights for a more effective therapeutic targeting of these interconnected pathologies (Supplemental Table S2). ACKNOWLEDGMENTS The authors thank Drs. Kerry Campbell and Sergei Grivennikov from Fox Chase Cancer Center (FCCC) and Dr. Rebecca Wells from the University of Pennsylvania for insightful comments during the preparation of this document.

Physiol Genomics • doi:10.1152/physiolgenomics.00158.2013 • www.physiolgenomics.org

Downloaded from on November 30, 2014

matory response, which when sustained and prolonged, in turn, would facilitate both fibrosis and tumorigenesis (35, 195, 389). Moreover, whether fibrotic myofibroblasts and TAFs are senescent or hyperproliferative, they also must evade stromal cell clearing mechanisms, such as apoptosis, which promote wound resolution (Fig. 1). In hypertrophic scars, the presence of apoptosis-resistant myofibroblasts high in antiapoptotic protein Bcl-2 has been reported (256). Although the mechanisms remain unclear, TGF-␤ may induce myofibroblast resistance to apoptosis (138). Intriguingly, the fact that tumors release a variety of molecules that can interfere with the immune response as well as with NK cell infiltration and activation (209) suggests the prevalence of an immunosuppressive microenvironment that may protect fibrotic myofibroblasts and desmoplastic TAFs from active NK cell elimination. For example, it has been shown that TGF-␤ is a potent inhibitor of NK (366). Although there is not much information about this mechanism in cancer and specifically on the tumor stroma, research on the chronic fibrosis field has shown a relationship between the persistence of myofibroblasts with an immunosuppressed microenvironment profile (i.e., myofibroblasts attract immunesuppressive cells) (38, 181, 375). Hence, facilitating the NK cell-mediated elimination of myofibroblastic and tumoral cells is expected to be a feasible approach to counteract the WHFC triad (38).

Review THE WHFC TRIAD Graphic design was by Perla Ovseiovich from Paralelo 19, Mexico City. Proofreading was conducted with the help of Ellen Ragan. Current address for B. Rybinski: JVA Group, Dept. 4, Tumor Microenvironment, Avondale, PA. GRANTS Funding used for the preparation of this manuscript included finances from the Commonwealth of Pennsylvania, FCCC’s Keystone Program in Personalized Kidney Cancer Therapy, a Nodal Temple-FCCC Award, The Bucks County Board of Associates, as well as National Cancer Institute Grants CA-113451 and CA-06927. DISCLOSURES No conflicts of interest, financial or otherwise, are declared by the author(s). AUTHOR CONTRIBUTIONS Author contributions: B.R., J.F.-B., and E.C. drafted manuscript; J.F.-B. and E.C. conception and design of research; J.F.-B. and E.C. prepared figures; J.F.-B. and E.C. edited and revised manuscript; E.C. approved final version of manuscript. REFERENCES

17. Barry-Hamilton V, Spangler R, Marshall D, McCauley S, Rodriguez HM, Oyasu M, Mikels A, Vaysberg M, Ghermazien H, Wai C, Garcia CA, Velayo AC, Jorgensen B, Biermann D, Tsai D, Green J, Zaffryar-Eilot S, Holzer A, Ogg S, Thai D, Neufeld G, Van Vlasselaer P, Smith V. Allosteric inhibition of lysyl oxidase-like-2 impedes the development of a pathologic microenvironment. Nat Med 16: 1009 – 1017, 2010. 18. Basu A, Kligman LH, Samulewicz SJ, Howe CC. Impaired wound healing in mice deficient in a matricellular protein SPARC (osteonectin, BM-40). BMC Cell Biol 2: 15, 2001. 19. Bauvois B. New facets of matrix metalloproteinases MMP-2 and MMP-9 as cell surface transducers: outside-in signaling and relationship to tumor progression. Biochim Biophys Acta 1825: 29 –36, 2012. 20. Beacham DA, Cukierman E. Stromagenesis: the changing face of fibroblastic microenvironments during tumor progression. Sem Cancer Biol 15: 329 –341, 2005. 21. Behm B, Babilas P, Landthaler M, Schreml S. Cytokines, chemokines and growth factors in wound healing. J Eur Acad Dermatol Venereol 26: 812–820, 2012. 22. Belfiore A, Genua M, Malaguarnera R. PPAR-gamma agonists and their effects on IGF-I receptor signaling: Implications for cancer. PPAR Res 2009: 830501, 2009. 23. Beno DW, Davis BH. Prostaglandin E suppresses hepatic fibrosis: section I. The in vivo approach; section II. The in vitro approach. Am J Ther 2: 687–705, 1995. 24. Bian EB, Zhao B, Huang C, Wang H, Meng XM, Wu BM, Ma TT, Zhang L, Lv XW, Li J. New advances of DNA methylation in liver fibrosis, with special emphasis on the crosstalk between microRNAs and DNA methylation machinery. Cell Signal 25: 1837–1844, 2013. 25. Bielefeld KA, Amini-Nik S, Alman BA. Cutaneous wound healing: recruiting developmental pathways for regeneration. Cell Mol Life Sci 70: 2059 –2081, 2013. 26. Bishen KA, Radhakrishnan R, Satyamoorthy K. The role of basic fibroblast growth factor in oral submucous fibrosis pathogenesis. J Oral Pathol Med 37: 402–411, 2008. 27. Bjermer L, Lundgren R, Hallgren R. Hyaluronan and type III procollagen peptide concentrations in bronchoalveolar lavage fluid in idiopathic pulmonary fibrosis. Thorax 44: 126 –131, 1989. 28. Black PC, Mize GJ, Karlin P, Greenberg DL, Hawley SJ, True LD, Vessella RL, Takayama TK. Overexpression of protease-activated receptors-1,-2, and-4 (PAR-1, -2, and -4) in prostate cancer. Prostate 67: 743–756, 2007. 29. Bloomston M, Ellison EC, Muscarella P, Al-Saif O, Martin EW, Melvin WS, Frankel WL. Stromal osteonectin overexpression is associated with poor outcome in patients with ampullary cancer. Ann Surg Oncol 14: 211–217, 2007. 30. Bobba RK, Holly JS, Loy T, Perry MC. Scar carcinoma of the lung: a historical perspective. Clin Lung Cancer 12: 148 –154, 2011. 31. Bogatkevich GS, Highland KB, Akter T, Silver RM. The PPARgamma agonist rosiglitazone is antifibrotic for scleroderma lung fibroblasts: mechanisms of action and differential racial effects. Pulmon Med 2012: 545172, 2012. 32. Bonner JC. Regulation of PDGF and its receptors in fibrotic diseases. Cytokine Growth Factor Rev 15: 255–273, 2004. 33. Borensztajn K, Peppelenbosch MP, Spek CA. Factor Xa: at the crossroads between coagulation and signaling in physiology and disease. Trends Mol Med 14: 429 –440, 2008. 34. Borensztajn K, von der Thusen JH, Peppelenbosch MP, Spek CA. The coagulation factor Xa/protease activated receptor-2 axis in the progression of liver fibrosis: a multifaceted paradigm. J Cell Mol Med 14: 143–153, 2010. 35. Borges FT, Melo SA, Ozdemir BC, Kato N, Revuelta I, Miller CA, Gattone VH 2nd, Lebleu VS, Kalluri R. TGF-beta1-containing exosomes from injured epithelial cells activate fibroblasts to initiate tissue regenerative responses and fibrosis. J Am Soc Nephrol 24: 385–392, 2013. 36. Bourboulia D, Stetler-Stevenson WG. Matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs): positive and negative regulators in tumor cell adhesion. Sem Cancer Biol 20: 161–168, 2010. 37. Bozyk PD, Moore BB. Prostaglandin E2 and the pathogenesis of pulmonary fibrosis. Am J Respir Cell Mol Biol 45: 445–452, 2011. 38. Brabletz T. To differentiate or not–routes towards metastasis. Nat Rev Cancer 12: 425–436, 2012.

Physiol Genomics • doi:10.1152/physiolgenomics.00158.2013 • www.physiolgenomics.org

Downloaded from on November 30, 2014

1. Abe M, Yokoyama Y, Ishikawa O. A possible mechanism of basic fibroblast growth factor-promoted scarless wound healing: the induction of myofibroblast apoptosis. Eur J Dermatol 22: 46 –53, 2012. 2. Abratt RP, Morgan GW. Lung toxicity following chest irradiation in patients with lung cancer. Lung Cancer 35: 103–109, 2002. 3. Al-Ansari MM, Hendrayani SF, Shehata AI, Aboussekhra A. p16(INK4A) represses the paracrine tumor-promoting effects of breast stromal fibroblasts. Oncogene 32: 2356 –2364, 2013. 4. Amatangelo MD, Bassi DE, Klein-Szanto AJ, Cukierman E. Stromaderived three-dimensional matrices are necessary and sufficient to promote desmoplastic differentiation of normal fibroblasts. Am J Pathol 167: 475–488, 2005. 5. Annes JP, Munger JS, Rifkin DB. Making sense of latent TGFbeta activation. J Cell Sci 116: 217–224, 2003. 6. Aoki T, Narumiya S. Prostaglandins and chronic inflammation. Trends Pharmacol Sci 33: 304 –311, 2012. 7. Aprile G, Avellini C, Reni M, Mazzer M, Foltran L, Rossi D, Cereda S, Iaiza E, Fasola G, Piga A. Biglycan expression and clinical outcome in patients with pancreatic adenocarcinoma. Tumour Biol 34: 131–137, 2013. 8. Ara T, Declerck YA. Interleukin-6 in bone metastasis and cancer progression. Eur J Cancer 46: 1223–1231, 2010. 9. Archontogeorgis K, Steiropoulos P, Tzouvelekis A, Nena E, Bouros D. Lung cancer and interstitial lung diseases: a systematic review. Pulmon Med 2012: 315918, 2012. 10. Atorrasagasti C, Peixoto E, Aquino JB, Kippes N, Malvicini M, Alaniz L, Garcia M, Piccioni F, Fiore EJ, Bayo J, Bataller R, Guruceaga E, Corrales F, Podhajcer O, Mazzolini G. Lack of the matricellular protein SPARC (secreted protein, acidic and rich in cysteine) attenuates liver fibrogenesis in mice. PLoS One 8: e54962, 2013. 11. Auvinen P, Tammi R, Parkkinen J, Tammi M, Agren U, Johansson R, Hirvikoski P, Eskelinen M, Kosma VM. Hyaluronan in peritumoral stroma and malignant cells associates with breast cancer spreading and predicts survival. Am J Pathol 156: 529 –536, 2000. 12. Bachem MG, Schunemann M, Ramadani M, Siech M, Beger H, Buck A, Zhou S, Schmid-Kotsas A, Adler G. Pancreatic carcinoma cells induce fibrosis by stimulating proliferation and matrix synthesis of stellate cells. Gastroenterology 128: 907–921, 2005. 13. Baghy K, Dezso K, Laszlo V, Fullar A, Peterfia B, Paku S, Nagy P, Schaff Z, Iozzo RV, Kovalszky I. Ablation of the decorin gene enhances experimental hepatic fibrosis and impairs hepatic healing in mice. Lab Invest 91: 439 –451, 2011. 14. Baghy K, Iozzo RV, Kovalszky I. Decorin-TGFbeta axis in hepatic fibrosis and cirrhosis. J Histochem Cytochem 60: 262–268, 2012. 15. Bailey JM, Mohr AM, Hollingsworth MA. Sonic hedgehog paracrine signaling regulates metastasis and lymphangiogenesis in pancreatic cancer. Oncogene 28: 3513–3525, 2009. 16. Barcellos-Hoff MH, Lyden D, Wang TC. The evolution of the cancer niche during multistage carcinogenesis. Nat Rev Cancer 13: 511–518, 2013.

235

Review 236

THE WHFC TRIAD 62. Corte MD, Verez P, Rodriguez JC, Roibas A, Dominguez ML, Lamelas ML, Vazquez J, Garcia Muniz JL, Allende MT, Gonzalez LO, Fueyo A, Vizoso F. Tissue-type plasminogen activator (tPA) in breast cancer: relationship with clinicopathological parameters and prognostic significance. Breast Cancer Res Treat 90: 33–40, 2005. 63. Coulson-Thomas VJ, Coulson-Thomas YM, Gesteira TF, de Paula CA, Mader AM, Waisberg J, Pinhal MA, Friedl A, Toma L, Nader HB. Colorectal cancer desmoplastic reaction up-regulates collagen synthesis and restricts cancer cell invasion. Cell Tiss Res 346: 223–236, 2011. 64. Cukierman E, Bassi DE. Physico-mechanical aspects of extracellular matrix influences on tumorigenic behaviors. Sem Cancer Biol 20: 139 – 145, 2010. 65. Cukierman E, Khan DR. The benefits and challenges associated with the use of drug delivery systems in cancer therapy. Biochem Pharmacol 80: 762–770, 2010. 66. D’Alterio C, Avallone A, Tatangelo F, Delrio P, Pecori B, Cella L, Pelella A, D’Armiento FP, Carlomagno C, Bianco F, Silvestro L, Pacelli R, Napolitano M, Iaffaioli RV, Scala S. A prognostic model comprising pT stage, N status, and the chemokine receptors CXCR4 and CXCR7 powerfully predicts outcome in neoadjuvant resistant rectal cancer patients. Int J Cancer [Epub ahead of print]. 67. Damm EW, Winklbauer R. PDGF-A controls mesoderm cell orientation and radial intercalation during Xenopus gastrulation. Development 138: 565–575, 2011. 68. Davalos D, Akassoglou K. Fibrinogen as a key regulator of inflammation in disease. Sem Immunopathol 34: 43–62, 2012. 69. Davids JS, Carothers AM, Damas BC, Bertagnolli MM. Chronic cyclooxygenase-2 inhibition promotes myofibroblast-associated intestinal fibrosis. Cancer Prev Res 3: 348 –358, 2010. 70. de Lujan Alvarez M, Ronco MT, Ochoa JE, Monti JA, Carnovale CE, Pisani GB, Lugano MC, Carrillo MC. Interferon alpha-induced apoptosis on rat preneoplastic liver is mediated by hepatocytic transforming growth factor beta(1). Hepatology 40: 394 –402, 2004. 71. De Wever O, Nguyen QD, Van Hoorde L, Bracke M, Bruyneel E, Gespach C, Mareel M. Tenascin-C and SF/HGF produced by myofibroblasts in vitro provide convergent pro-invasive signals to human colon cancer cells through RhoA and Rac. FASEB J 18: 1016 –1018, 2004. 72. De Wever O, Westbroek W, Verloes A, Bloemen N, Bracke M, Gespach C, Bruyneel E, Mareel M. Critical role of N-cadherin in myofibroblast invasion and migration in vitro stimulated by coloncancer-cell-derived TGF-beta or wounding. J Cell Sci 117: 4691–4703, 2004. 73. Demicheli R, Retsky MW, Hrushesky WJ, Baum M, Gukas ID. The effects of surgery on tumor growth: a century of investigations. Ann Oncol 19: 1821–1828, 2008. 74. Derosa CA, Furusato B, Shaheduzzaman S, Srikantan V, Wang Z, Chen Y, Seifert M, Ravindranath L, Young D, Nau M, Dobi A, Werner T, McLeod DG, Vahey MT, Sesterhenn IA, Srivastava S, Petrovics G. Elevated osteonectin/SPARC expression in primary prostate cancer predicts metastatic progression. Prost Cancer Prostat Dis 15: 150 –156, 2012. 75. Desmouliere A, Redard M, Darby I, Gabbiani G. Apoptosis mediates the decrease in cellularity during the transition between granulation tissue and scar. Am J Pathol 146: 56 –66, 1995. 76. Devalaraja RM, Nanney LB, Du J, Qian Q, Yu Y, Devalaraja MN, Richmond A. Delayed wound healing in CXCR2 knockout mice. J Invest Dermatol 115: 234 –244, 2000. 77. Dishowitz MI, Mutyaba PL, Takacs JD, Barr AM, Engiles JB, Ahn J, Hankenson KD. Systemic inhibition of canonical notch signaling results in sustained callus inflammation and alters multiple phases of fracture healing. PLoS One 8: e68726, 2013. 78. Du WW, Fang L, Yang X, Sheng W, Yang BL, Seth A, Zhang Y, Yang BB, Yee AJ. The role of versican in modulating breast cancer cell self-renewal. Mol Cancer Res 11: 443–455, 2013. 79. Du WW, Yang BB, Yang BL, Deng Z, Fang L, Shan SW, Jeyapalan Z, Zhang Y, Seth A, Yee AJ. Versican G3 domain modulates breast cancer cell apoptosis: a mechanism for breast cancer cell response to chemotherapy and EGFR therapy. PLoS One 6: e26396, 2011. 80. Dube PE, Yan F, Punit S, Girish N, McElroy SJ, Washington MK, Polk DB. Epidermal growth factor receptor inhibits colitis-associated cancer in mice. J Clin Invest 122: 2780 –2792, 2012. 81. Duell EJ, Lucenteforte E, Olson SH, Bracci PM, Li D, Risch HA, Silverman DT, Ji BT, Gallinger S, Holly EA, Fontham EH, Maison-

Physiol Genomics • doi:10.1152/physiolgenomics.00158.2013 • www.physiolgenomics.org

Downloaded from on November 30, 2014

39. Bradshaw AD, Reed MJ, Sage EH. SPARC-null mice exhibit accelerated cutaneous wound closure. J Histochem Cytochem 50: 1–10, 2002. 40. Brellier F, Hostettler K, Hotz HR, Ozcakir C, Cologlu SA, Togbe D, Ryffel B, Roth M, Chiquet-Ehrismann R. Tenascin-C triggers fibrin accumulation by downregulation of tissue plasminogen activator. FEBS Lett 585: 913–920, 2011. 41. Burgess HA, Daugherty LE, Thatcher TH, Lakatos HF, Ray DM, Redonnet M, Phipps RP, Sime PJ. PPARgamma agonists inhibit TGF-beta induced pulmonary myofibroblast differentiation and collagen production: implications for therapy of lung fibrosis. Am J Physiol Lung Cell Mol Physiol 288: L1146 –L1153, 2005. 42. Butcher DT, Alliston T, Weaver VM. A tense situation: forcing tumour progression. Nat Rev Cancer 9: 108 –122, 2009. 43. Carey WA, Taylor GD, Dean WB, Bristow JD. Tenascin-C deficiency attenuates TGF-ss-mediated fibrosis following murine lung injury. Am J Physiol Lung Cell Mol Physiol 299: L785–L793, 2010. 44. Carothers AM, Davids JS, Damas BC, Bertagnolli MM. Persistent cyclooxygenase-2 inhibition downregulates NF-␬B, resulting in chronic intestinal inflammation in the min/⫹ mouse model of colon tumorigenesis. Cancer Res 70: 4433–4442, 2010. 45. Castelló-Cros R, Khan DR, Simons J, Valianou M, Cukierman E. Staged stromal extracellular 3D matrices differentially regulate breast cancer cell responses through PI3K and beta1-integrins. BMC Cancer 9: 94, 2009. 46. Catarzi S, Favilli F, Romagnoli C, Marcucci T, Picariello L, Tonelli F, Vincenzini MT, Iantomasi T. Oxidative state and IL-6 production in intestinal myofibroblasts of Crohn’s disease patients. Inflamm Bowel Dis 17: 1674 –1684, 2011. 47. Chambers RC, Laurent GJ. Coagulation cascade proteases and tissue fibrosis. Biochem Soc Transact 30: 194 –200, 2002. 48. Chapman RW, Phillips JE, Hipkin RW, Curran AK, Lundell D, Fine JS. CXCR2 antagonists for the treatment of pulmonary disease. Pharmacol Therapeut 121: 55–68, 2009. 49. Charpidou AG, Gkiozos I, Tsimpoukis S, Apostolaki D, Dilana KD, Karapanagiotou EM, Syrigos KN. Therapy-induced toxicity of the lungs: an overview. Anticancer Res 29: 631–639, 2009. 50. Chaturvedi P, Gilkes DM, Wong CC, Kshitiz Luo W, Zhang H, Wei H, Takano N, Schito L, Levchenko A, Semenza GL. Hypoxia-inducible factor-dependent breast cancer-mesenchymal stem cell bidirectional signaling promotes metastasis. J Clin Invest 123: 189 –205, 2013. 51. Chen L, Necela BM, Su W, Yanagisawa M, Anastasiadis PZ, Fields AP, Thompson EA. Peroxisome proliferator-activated receptor gamma promotes epithelial to mesenchymal transformation by Rho GTPasedependent activation of ERK1/2. J Biol Chem 281: 24575–24587, 2006. 52. Chen L, Neville RD, Michael DR, Martin J, Luo DD, Thomas DW, Phillips AO, Bowen T. Identification and analysis of the human hyaluronan synthase 1 gene promoter reveals Smad3- and Sp3-mediated transcriptional induction. Matrix Biol 31: 373–379, 2012. 53. Chen S, Birk DE. The regulatory roles of small leucine-rich proteoglycans in extracellular matrix assembly. FEBS J 280: 2120 –2137, 2013. 54. Chigurupati S, Arumugam TV, Son TG, Lathia JD, Jameel S, Mughal MR, Tang SC, Jo DG, Camandola S, Giunta M, Rakova I, McDonnell N, Miele L, Mattson MP, Poosala S. Involvement of notch signaling in wound healing. PLoS One 2: e1167, 2007. 55. Chlenski A, Cohn SL. Modulation of matrix remodeling by SPARC in neoplastic progression. Sem Cell Dev Biol 21: 55–65, 2010. 56. Cirri P, Chiarugi P. Cancer associated fibroblasts: the dark side of the coin. Am J Cancer Res 1: 482–497, 2011. 57. Clark SJ, Bishop PN, Day AJ. The proteoglycan glycomatrix: a sugar microenvironment essential for complement regulation. Front Immunol 4: 412, 2013. 58. Cohen DJ. Targeting the hedgehog pathway: role in cancer and clinical implications of its inhibition. Hematol/oncol Clin No Am 26: 565–588, viii, 2012. 59. Conklin MW, Eickhoff JC, Riching KM, Pehlke CA, Eliceiri KW, Provenzano PP, Friedl A, Keely PJ. Aligned collagen is a prognostic signature for survival in human breast carcinoma. Am J Pathol 178: 1221–1232, 2011. 60. Conway SJ, Molkentin JD. Periostin as a heterofunctional regulator of cardiac development and disease. Curr Genomics 9: 548 –555, 2008. 61. Coppe JP, Desprez PY, Krtolica A, Campisi J. The senescenceassociated secretory phenotype: the dark side of tumor suppression. Ann Rev Pathol 5: 99 –118, 2010.

Review THE WHFC TRIAD

82. 83. 84. 85.

86. 87.

88.

90. 91. 92.

93. 94.

95. 96.

97. 98. 99. 100. 101.

102. 103. 104.

105. Gallucci RM, Lee EG, Tomasek JJ. IL-6 modulates alpha-smooth muscle actin expression in dermal fibroblasts from IL-6-deficient mice. J Invest Dermatol 126: 561–568, 2006. 106. Garamszegi N, Garamszegi SP, Samavarchi-Tehrani P, Walford E, Schneiderbauer MM, Wrana JL, Scully SP. Extracellular matrixinduced transforming growth factor-beta receptor signaling dynamics. Oncogene 29: 2368 –2380, 2010. 107. Gelse K, Poschl E, Aigner T. Collagens–structure, function, biosynthesis. Adv Drug Deliv Rev 55: 1531–1546, 2003. 108. Genovese F, Barascuk N, Larsen L, Larsen MR, Nawrocki A, Li Y, Zheng Q, Wang J, Veidal SS, Leeming DJ, Karsdal MA. Biglycan fragmentation in pathologies associated with extracellular matrix remodeling by matrix metalloproteinases. Fibrogenesis Tiss Repair 6: 9, 2013. 109. Gharaee-Kermani M, Kasina S, Moore BB, Thomas D, Mehra R, Macoska JA. CXC-type chemokines promote myofibroblast phenoconversion and prostatic fibrosis. PLoS One 7: e49278, 2012. 110. Ghezzi F, Cromi A, Siesto G, Giudici S, Serati M, Formenti G, Franchi M. Prognostic significance of preoperative plasma fibrinogen in endometrial cancer. Gynecol Oncol 119: 309 –313, 2010. 111. Ghosh AK, Vaughan DE. PAI-1 in tissue fibrosis. J Cell Physiol 227: 493–507, 2012. 112. Ghosh S, Albitar L, LeBaron R, Welch WR, Samimi G, Birrer MJ, Berkowitz RS, Mok SC. Up-regulation of stromal versican expression in advanced stage serous ovarian cancer. Gynecol Oncol 119: 114 –120, 2010. 113. Gill SE, Parks WC. Metalloproteinases and their inhibitors: regulators of wound healing. Int J Biochem Cell Biol 40: 1334 –1347, 2008. 114. Girotti MR, Fernandez M, Lopez JA, Camafeita E, Fernandez EA, Albar JP, Benedetti LG, Valacco MP, Brekken RA, Podhajcer OL, Llera AS. SPARC promotes cathepsin B-mediated melanoma invasiveness through a collagen I/alpha2beta1 integrin axis. J Invest Dermatol 131: 2438 –2447, 2011. 115. Godbout C, Follonier Castella L, Smith EA, Talele N, Chow ML, Garonna A, Hinz B. The mechanical environment modulates intracellular calcium oscillation activities of myofibroblasts. PLoS One 8: e64560, 2013. 116. Goetz JG, Minguet S, Navarro-Lérida I, Lazcano JJ, Samaniego R, Calvo E, Tello M, Osteso-Ibáñez T, Pellinen T, Echarri A, Cerezo A, Klein-Szanto AJ, Garcia R, Keely PJ, Sánchez-Mateos P, Cukierman E, Del Pozo MA. Biomechanical remodeling of the microenvironment by stromal caveolin-1 favors tumor invasion and metastasis. Cell 146: 148 –163, 2011. 117. Goldacre MJ, Wotton CJ, Yeates D, Seagroatt V, Collier J. Liver cirrhosis, other liver diseases, pancreatitis and subsequent cancer: record linkage study. Eur J Gastroenterol Hepatol 20: 384 –392, 2008. 118. Greaves NS, Ashcroft KJ, Baguneid M, Bayat A. Current understanding of molecular and cellular mechanisms in fibroplasia and angiogenesis during acute wound healing. J Dermatol Sci 72: 206 –217, 2013. 119. Grivennikov S, Karin E, Terzic J, Mucida D, Yu GY, Vallabhapurapu S, Scheller J, Rose-John S, Cheroutre H, Eckmann L, Karin M. IL-6 and Stat3 are required for survival of intestinal epithelial cells and development of colitis-associated cancer. Cancer Cell 15: 103–113, 2009. 120. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, cancer. Cell 140: 883–899, 2010. 121. Grivennikov SI, Wang K, Mucida D, Stewart CA, Schnabl B, Jauch D, Taniguchi K, Yu GY, Osterreicher CH, Hung KE, Datz C, Feng Y, Fearon ER, Oukka M, Tessarollo L, Coppola V, Yarovinsky F, Cheroutre H, Eckmann L, Trinchieri G, Karin M. Adenoma-linked barrier defects and microbial products drive IL-23/IL-17-mediated tumour growth. Nature 491: 254 –258, 2012. 122. Gu X, Ma Y, Xiao J, Zheng H, Song C, Gong Y, Xing X. Up-regulated biglycan expression correlates with the malignancy in human colorectal cancers. Clin Exp Med 12: 195–199, 2012. 123. Guarino M, Tosoni A, Nebuloni M. Direct contribution of epithelium to organ fibrosis: epithelial-mesenchymal transition. Hum Pathol 40: 1365– 1376, 2009. 124. Gupta RA, Dubois RN. Controversy: PPAR␥ as a target for treatment of colorectal cancer. Am J Physiol Gastrointest Liver Physiol 283: G266 – G269, 2002. 125. Gupta V, Bassi DE, Simons JD, Devarajan K, Al-Saleem T, Uzzo RG, Cukierman E. Elevated expression of stromal palladin predicts poor clinical outcome in renal cell carcinoma. PLoS One 6: e21494, 2011.

Physiol Genomics • doi:10.1152/physiolgenomics.00158.2013 • www.physiolgenomics.org

Downloaded from on November 30, 2014

89.

neuve P, Bueno-de-Mesquita HB, Ghadirian P, Kurtz RC, Ludwig E, Yu H, Lowenfels AB, Seminara D, Petersen GM, La Vecchia C, Boffetta P. Pancreatitis and pancreatic cancer risk: a pooled analysis in the International Pancreatic Cancer Case-Control Consortium (PanC4). Ann Oncol 23: 2964 –2970, 2012. Dugina V, Fontao L, Chaponnier C, Vasiliev J, Gabbiani G. Focal adhesion features during myofibroblastic differentiation are controlled by intracellular and extracellular factors. J Cell Sci 114: 3285–3296, 2001. Duplantier JG, Dubuisson L, Senant N, Freyburger G, Laurendeau I, Herbert JM, Desmouliere A, Rosenbaum J. A role for thrombin in liver fibrosis. Gut 53: 1682–1687, 2004. Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med 315: 1650 – 1659, 1986. El-Karef A, Yoshida T, Gabazza EC, Nishioka T, Inada H, Sakakura T, Imanaka-Yoshida K. Deficiency of tenascin-C attenuates liver fibrosis in immune-mediated chronic hepatitis in mice. J Pathol 211: 86 –94, 2007. El-Serag HB. Epidemiology of viral hepatitis and hepatocellular carcinoma. Gastroenterology 142: 1264 –1273.e1, 2012. Elliott CG, Wang J, Guo X, Xu SW, Eastwood M, Guan J, Leask A, Conway SJ, Hamilton DW. Periostin modulates myofibroblast differentiation during full-thickness cutaneous wound repair. J Cell Sci 125: 121–132, 2012. Elliott CG, Wang J, Guo X, Xu SW, Eastwood M, Guan J, Leask A, Conway SJ, Hamilton DW. Periostin modulates myofibroblast differentiation during full-thickness cutaneous wound repair. J Cell Sci 125: 121–132, 2012. Elste AP, Petersen I. Expression of proteinase-activated receptor 1– 4 (PAR 1– 4) in human cancer. J Mol Histol 41: 89 –99, 2010. Eyden B, Banerjee SS, Shenjere P, Fisher C. The myofibroblast and its tumours. J Clin Pathol 62: 236 –249, 2009. Farazi PA, DePinho RA. Hepatocellular carcinoma pathogenesis: from genes to environment. Nat Rev Cancer 6: 674 –687, 2006. Fedak PW, Bai L, Turnbull J, Ngu J, Narine K, Duff HJ. Cell therapy limits myofibroblast differentiation and structural cardiac remodeling: basic fibroblast growth factor-mediated paracrine mechanism. Circ Heart Fail 5: 349 –356, 2012. Fernandez PM, Patierno SR, Rickles FR. Tissue factor and fibrin in tumor angiogenesis. Sem Thrombosis Hemostasis 30: 31–44, 2004. Filosto S, Becker CR, Goldkorn T. Cigarette smoke induces aberrant EGF receptor activation that mediates lung cancer development and resistance to tyrosine kinase inhibitors. Mol Cancer Therapeut 11: 795–804, 2012. Franchini M, Mannucci PM. Thrombin and cancer: from molecular basis to therapeutic implications. Sem Thrombosis Hemostasis 38: 95– 101, 2012. Franciszkiewicz K, Boissonnas A, Boutet M, Combadiere C, MamiChouaib F. Role of chemokines and chemokine receptors in shaping the effector phase of the antitumor immune response. Cancer Res 72: 6325–6332, 2012. Frantz C, Stewart KM, Weaver VM. The extracellular matrix at a glance. J Cell Sci 123: 4195–4200, 2010. Fraser JR, Laurent TC, Pertoft H, Baxter E. Plasma clearance, tissue distribution and metabolism of hyaluronic acid injected intravenously in the rabbit. Biochem J 200: 415–424, 1981. Frey H, Schroeder N, Manon-Jensen T, Iozzo RV, Schaefer L. Biological interplay between proteoglycans and their innate immune receptors in inflammation. FEBS J 280: 2165–2179, 2013. Fujimoto D, Hirono Y, Goi T, Katayama K, Yamaguchi A. Prognostic value of protease-activated receptor-1 (PAR-1) and matrix metalloproteinase-1 (MMP-1) in gastric cancer. Anticancer Res 28: 847–854, 2008. Fukunaga-Kalabis M, Martinez G, Nguyen TK, Kim D, SantiagoWalker A, Roesch A, Herlyn M. Tenascin-C promotes melanoma progression by maintaining the ABCB5-positive side population. Oncogene 29: 6115–6124, 2010. Fukushima N, Kikuchi Y, Nishiyama T, Kudo A, Fukayama M. Periostin deposition in the stroma of invasive and intraductal neoplasms of the pancreas. Mod Pathol 21: 1044 –1053, 2008. Fuxe J, Karlsson MCI. TGF-␤-induced epithelial-mesenchymal transition: a link between cancer and inflammation. Sem Cancer Biol 22: 455–461, 2012. Gales D, Clark C, Manne U, Samuel T. The chemokine CXCL8 in carcinogenesis and drug response. ISRN Oncol 2013: 859154, 2013.

237

Review 238

THE WHFC TRIAD 148. Hurst NJ Jr, Najy AJ, Ustach CV, Movilla L, Kim HR. Plateletderived growth factor-C (PDGF-C) activation by serine proteases: implications for breast cancer progression. Biochem J 441: 909 –918, 2012. 149. Hwang RF, Moore TT, Hattersley MM, Scarpitti M, Yang B, Devereaux E, Ramachandran V, Arumugam T, Ji B, Logsdon CD, Brown JL, Godin R. Inhibition of the hedgehog pathway targets the tumor-associated stroma in pancreatic cancer. Mol Cancer Res 10: 1147–1157, 2012. 150. Ichiki Y, Smith EA, LeRoy EC, Trojanowska M. Basic fibroblast growth factor inhibits basal and transforming growth factor-beta induced collagen alpha 2(I) gene expression in scleroderma and normal fibroblasts. J Rheumatol 24: 90 –95, 1997. 151. Imokawa S, Sato A, Hayakawa H, Kotani M, Urano T, Takada A. Tissue factor expression and fibrin deposition in the lungs of patients with idiopathic pulmonary fibrosis and systemic sclerosis. Am J Respir Crit Care Med 156: 631–636, 1997. 152. Inoue Y, King TE Jr, Barker E, Daniloff E, Newman LS. Basic fibroblast growth factor and its receptors in idiopathic pulmonary fibrosis and lymphangioleiomyomatosis. Am J Respir Crit Care Med 166: 765– 773, 2002. 153. Iozzo RV, Sanderson RD. Proteoglycans in cancer biology, tumour microenvironment and angiogenesis. J Cell Mol Med 15: 1013–1031, 2011. 154. Ishiguro S, Akasaka Y, Kiguchi H, Suzuki T, Imaizumi R, Ishikawa Y, Ito K, Ishii T. Basic fibroblast growth factor induces down-regulation of alpha-smooth muscle actin and reduction of myofibroblast areas in open skin wounds. Wound Repair Regen 17: 617–625, 2009. 155. Iwaki T, Urano T, Umemura K. PAI-1, progress in understanding the clinical problem and its aetiology. Br J Haematol 157: 291–298, 2012. 156. Jarvinen TA, Ruoslahti E. Target-seeking antifibrotic compound enhances wound healing and suppresses scar formation in mice. Proc Natl Acad Sci USA 107: 21671–21676, 2010. 157. Javelaud D, Pierrat MJ, Mauviel A. Crosstalk between TGF-beta and hedgehog signaling in cancer. FEBS Lett 586: 2016 –2025, 2012. 158. Jedeszko C, Victor BC, Podgorski I, Sloane BF. Fibroblast hepatocyte growth factor promotes invasion of human mammary ductal carcinoma in situ. Cancer Res 69: 9148 –9155, 2009. 159. Jiang D, Liang J, Noble PW. Hyaluronan as an immune regulator in human diseases. Physiol Rev 91: 221–264, 2011. 160. Johari V, Loke C. Brief overview of the coagulation cascade. Dis Mon 58: 421–423, 2012. 161. Johnson NR, Wang Y. Controlled delivery of heparin-binding EGF-like growth factor yields fast and comprehensive wound healing. J Controlled Release 166: 124 –129, 2013. 162. Ju W, Zhihong Y, Zhiyou Z, Qin H, Dingding W, Li S, Baowei Z, Xing W, Ying H, An H. Inhibition of alpha-SMA by the ectodomain of FGFR2c attenuates lung fibrosis. Mol Med 18: 992–1002, 2012. 163. Jun JI, Lau LF. The matricellular protein CCN1 induces fibroblast senescence and restricts fibrosis in cutaneous wound healing. Nat Cell Biol 12: 676 –685, 2010. 164. Kagan HM. Intra- and extracellular enzymes of collagen biosynthesis as biological and chemical targets in the control of fibrosis. Acta Trop 77: 147–152, 2000. 165. Kagan HM, Li W. Lysyl oxidase: properties, specificity, and biological roles inside and outside of the cell. J Cell Biochem 88: 660 –672, 2003. 166. Kakudo N, Kushida S, Suzuki K, Ogura T, Notodihardjo PV, Hara T, Kusumoto K. Effects of transforming growth factor-beta1 on cell motility, collagen gel contraction, myofibroblastic differentiation, and extracellular matrix expression of human adipose-derived stem cell. Hum Cell 25: 87–95, 2012. 167. Kalluri R. Basement membranes: structure, assembly and role in tumour angiogenesis. Nat Rev Cancer 3: 422–433, 2003. 168. Kass L, Erler JT, Dembo M, Weaver VM. Mammary epithelial cell: influence of extracellular matrix composition and organization during development and tumorigenesis. Int J Biochem Cell Biol 39: 1987–1994, 2007. 169. Kasthuri RS, Taubman MB, Mackman N. Role of tissue factor in cancer. J Clin Oncol 27: 4834 –4838, 2009. 170. Katoh H, Wang D, Daikoku T, Sun H, Dey SK, Dubois RN. CXCR2expressing myeloid-derived suppressor cells are essential to promote colitis-associated tumorigenesis. Cancer Cell 24: 631–644, 2013. 171. Kavian N, Servettaz A, Weill B, Batteux F. New insights into the mechanism of notch signalling in fibrosis. Open Rheumatol J 6: 96 –102, 2012.

Physiol Genomics • doi:10.1152/physiolgenomics.00158.2013 • www.physiolgenomics.org

Downloaded from on November 30, 2014

126. Hakkinen L, Westermarck J, Kahari VM, Larjava H. Human granulation-tissue fibroblasts show enhanced proteoglycan gene expression and altered response to TGF-beta 1. J Dent Res 75: 1767–1778, 1996. 127. Hancox RA, Allen MD, Holliday DL, Edwards DR, Pennington CJ, Guttery DS, Shaw JA, Walker RA, Pringle JH, Jones JL. Tumourassociated tenascin-C isoforms promote breast cancer cell invasion and growth by matrix metalloproteinase-dependent and independent mechanisms. Breast Cancer Res 11: R24, 2009. 128. Hardie WD, Davidson C, Ikegami M, Leikauf GD, Le Cras TD, Prestridge A, Whitsett JA, Korfhagen TR. EGF receptor tyrosine kinase inhibitors diminish transforming growth factor-alpha-induced pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 294: L1217– L1225, 2008. 129. Harding P. Do COX-2 inhibitors reduce renal fibrosis? J Hypertens 22: 43–45, 2004. 130. Hardy KM, Booth BW, Hendrix MJ, Salomon DS, Strizzi L. ErbB/ EGF signaling and EMT in mammary development and breast cancer. J Mamm Gland Biol Neoplas 15: 191–199, 2010. 131. Hardy MM, Blomme EA, Lisowski A, Chinn KS, Jones A, Harmon JM, Opsahl A, Ornberg RL, Tripp CS. Selective cyclooxygenase-2 inhibition does not alter keratinocyte wound responses in the mouse epidermis after abrasion. J Pharmacol Exp Therapeut 304: 959 –967, 2003. 132. Heinrich EL, Lee W, Lu J, Lowy AM, Kim J. Chemokine CXCL12 activates dual CXCR4 and CXCR7-mediated signaling pathways in pancreatic cancer cells. J Translat Med 10: 68, 2012. 133. Heldin CH. Autocrine PDGF stimulation in malignancies. Ups J Med Sci 117: 83–91, 2012. 134. Herbst RS. Review of epidermal growth factor receptor biology. Int J Radiat Oncol Biol Physics 59: 21–26, 2004. 135. Herrick S, Blanc-Brude O, Gray A, Laurent G. Fibrinogen. Int J Biochem Cell Biolo 31: 741–746, 1999. 136. Hinz B. Formation and function of the myofibroblast during tissue repair. J Invest Dermatol 127: 526 –537, 2007. 137. Hinz B. The myofibroblast: paradigm for a mechanically active cell. J Biomechan 43: 146 –155, 2010. 138. Hinz B, Gabbiani G. Fibrosis: recent advances in myofibroblast biology and new therapeutic perspectives. F1000 Biol Rep 2: 78, 2010. 139. Hinz B, Mastrangelo D, Iselin CE, Chaponnier C, Gabbiani G. Mechanical tension controls granulation tissue contractile activity and myofibroblast differentiation. Am J Pathol 159: 1009 –1020, 2001. 140. Hinz B, Phan SH, Thannickal VJ, Prunotto M, Desmouliere A, Varga J, De Wever O, Mareel M, Gabbiani G. Recent developments in myofibroblast biology: paradigms for connective tissue remodeling. Am J Pathol 180: 1340 –1355, 2012. 141. Howard EW, Crider BJ, Updike DL, Bullen EC, Parks EE, Haaksma CJ, Sherry DM, Tomasek JJ. MMP-2 expression by fibroblasts is suppressed by the myofibroblast phenotype. Exp Cell Res 318: 1542– 1553, 2012. 142. Howell DC, Johns RH, Lasky JA, Shan B, Scotton CJ, Laurent GJ, Chambers RC. Absence of proteinase-activated receptor-1 signaling affords protection from bleomycin-induced lung inflammation and fibrosis. Am J Pathol 166: 1353–1365, 2005. 143. Howell DC, Laurent GJ, Chambers RC. Role of thrombin and its major cellular receptor, protease-activated receptor-1, in pulmonary fibrosis. Biochem Soc Transact 30: 211–216, 2002. 144. Hsiao Y, Zou T, Ling CC, Hu H, Tao XM, Song HY. Disruption of tissue-type plasminogen activator gene in mice aggravated liver fibrosis. J Gastroenterol Hepatol 23: e258 –e264, 2008. 145. Huang C, Yang G, Jiang T, Zhu G, Li H, Qiu Z. The effects and mechanisms of blockage of STAT3 signaling pathway on IL-6 inducing EMT in human pancreatic cancer cells in vitro. Neoplasma 58: 396 –405, 2011. 146. Hudspeth K, Pontarini E, Tentorio P, Cimino M, Donadon M, Torzilli G, Lugli E, Della Bella S, Gershwin ME, Mavilio D. The role of natural killer cells in autoimmune liver disease: a comprehensive review. J Autoimmun 46: 55–65, 2013. 147. Huet E, Vallee B, Szul D, Verrecchia F, Mourah S, Jester JV, Hoang-Xuan T, Menashi S, Gabison EE. Extracellular matrix metalloproteinase inducer/CD147 promotes myofibroblast differentiation by inducing alpha-smooth muscle actin expression and collagen gel contraction: implications in tissue remodeling. FASEB J 22: 1144 –1154, 2008.

Review THE WHFC TRIAD

192. Kudo A. Periostin in fibrillogenesis for tissue regeneration: periostin actions inside and outside the cell. Cell Mol Life Sci 68: 3201–3207, 2011. 193. Kudo Y, Iizuka S, Yoshida M, Nguyen PT, Siriwardena SB, Tsunematsu T, Ohbayashi M, Ando T, Hatakeyama D, Shibata T, Koizumi K, Maeda M, Ishimaru N, Ogawa I, Takata T. Periostin directly and indirectly promotes tumor lymphangiogenesis of head and neck cancer. PLoS One 7: e44488, 2012. 194. Kulkarni AA, Thatcher TH, Olsen KC, Maggirwar SB, Phipps RP, Sime PJ. PPAR-gamma ligands repress TGFbeta-induced myofibroblast differentiation by targeting the PI3K/Akt pathway: implications for therapy of fibrosis. PLoS One 6: e15909, 2011. 195. Kuraishy A, Karin M, Grivennikov SI. Tumor promotion via injuryand death-induced inflammation. Immunity 35: 467–477, 2011. 196. Kurata K, Maruyama S, Kato S, Sato W, Yamamoto J, Ozaki T, Nitta A, Nabeshima T, Morita Y, Mizuno M, Ito Y, Yuzawa Y, Matsuo S. Tissue-type plasminogen activator deficiency attenuates peritoneal fibrosis in mice. Am J Physiol Renal Physiol 297: F1510 –F1517, 2009. 197. Kwon Y, Cukierman E, Godwin AK. Differential expression of adhesive molecules and proteases define mechanisms of ovarian tumor cell matrix penetration/invasion. PLoS One 6: e18872, 2011. 198. Lau LF. CCN1/CYR61: the very model of a modern matricellular protein. Cell Mol Life Sci 68: 3149 –3163, 2011. 199. Lauth M, Toftgard R. Hedgehog signaling and pancreatic tumor development. Adv Cancer Res 110: 1–17, 2011. 200. Le H, Kleinerman R, Lerman OZ, Brown D, Galiano R, Gurtner GC, Warren SM, Levine JP, Saadeh PB. Hedgehog signaling is essential for normal wound healing. Wound Repair Regen 16: 768 –773, 2008. 201. Lee HO, Mullins SR, Franco-Barraza J, Valianou M, Cukierman E, Cheng JD. FAP-overexpressing fibroblasts produce an extracellular matrix that enhances invasive velocity and directionality of pancreatic cancer cells. BMC Cancer 11: 245, 2011. 202. Lee HM, Kang HJ, Park HH, Hong SC, Kim JK, Cho JH. Effect of peroxisome proliferator-activated receptor gamma agonists on myofibroblast differentiation and collagen production in nasal polyp-derived fibroblasts. Ann Otol Rhinol Laryngol 118: 721–727, 2009. 203. Lee SE, Lee JH, Ryu KW, Nam BH, Cho SJ, Lee JY, Kim CG, Choi IJ, Kook MC, Park SR, Kim YW. Preoperative plasma fibrinogen level is a useful predictor of adjacent organ involvement in patients with advanced gastric cancer. J Gastr Cancer 12: 81–87, 2012. 204. Leeming DJ, Byrjalsen I, Jimenez W, Christiansen C, Karsdal MA. Protein fingerprinting of the extracellular matrix remodelling in a rat model of liver fibrosis–a serological evaluation. Liver Int 33: 439 –447, 2013. 205. Leeming DJ, Sand JM, Nielsen MJ, Genovese F, Martinez FJ, Hogaboam CM, Han MK, Klickstein LB, Karsdal MA. Serological investigation of the collagen degradation profile of patients with chronic obstructive pulmonary disease or idiopathic pulmonary fibrosis. Biomark Insights 7: 119 –126, 2012. 206. Leight JL, Wozniak MA, Chen S, Lynch ML, Chen CS. Matrix rigidity regulates a switch between TGF-beta1-induced apoptosis and epithelial-mesenchymal transition. Mol Biol Cell 23: 781–791, 2012. 207. Leins A, Riva P, Lindstedt R, Davidoff MS, Mehraein P, Weis S. Expression of tenascin-C in various human brain tumors and its relevance for survival in patients with astrocytoma. Cancer 98: 2430 –2439, 2003. 208. Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, Fong SF, Csiszar K, Giaccia A, Weninger W, Yamauchi M, Gasser DL, Weaver VM. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell 139: 891–906, 2009. 209. Levy EM, Roberti MP, Mordoh J. Natural killer cells in human cancer: from biological functions to clinical applications. J Biomed Biotechnol 2011: 676198, 2011. 210. Lewis A, Steadman R, Manley P, Craig K, de la Motte C, Hascall V, Phillips AO. Diabetic nephropathy, inflammation, hyaluronan and interstitial fibrosis. Histol Histopathol 23: 731–739, 2008. 211. Lewis MP, Lygoe KA, Nystrom ML, Anderson WP, Speight PM, Marshall JF, Thomas GJ. Tumour-derived TGF-beta1 modulates myofibroblast differentiation and promotes HGF/SF-dependent invasion of squamous carcinoma cells. Br J Cancer 90: 822–832, 2004. 212. Li J, Chen J, Kirsner R. Pathophysiology of acute wound healing. Clin Dermatol 25: 9 –18, 2007.

Physiol Genomics • doi:10.1152/physiolgenomics.00158.2013 • www.physiolgenomics.org

Downloaded from on November 30, 2014

172. Kawada K, Hosogi H, Sonoshita M, Sakashita H, Manabe T, Shimahara Y, Sakai Y, Takabayashi A, Oshima M, Taketo MM. Chemokine receptor CXCR3 promotes colon cancer metastasis to lymph nodes. Oncogene 26: 4679 –4688, 2007. 173. Kawada K, Sonoshita M, Sakashita H, Takabayashi A, Yamaoka Y, Manabe T, Inaba K, Minato N, Oshima M, Taketo MM. Pivotal role of CXCR3 in melanoma cell metastasis to lymph nodes. Cancer Res 64: 4010 –4017, 2004. 174. Kawai T, Masaki T, Doi S, Arakawa T, Yokoyama Y, Doi T, Kohno N, Yorioka N. PPAR-gamma agonist attenuates renal interstitial fibrosis and inflammation through reduction of TGF-beta. Lab Invest 89: 47–58, 2009. 175. Keglowich L, Roth M, Philippova M, Resink T, Tjin G, Oliver B, Lardinois D, Dessus-Babus S, Gosens R, Hostettler Haack K, Tamm M, Borger P. Bronchial smooth muscle cells of asthmatics promote angiogenesis through elevated secretion of CXC-chemokines (ENA-78, GRO-alpha, and IL-8). PLoS One 8: e81494, 2013. 176. Kessenbrock K, Plaks V, Werb Z. Matrix metalloproteinases: regulators of the tumor microenvironment. Cell 141: 52–67, 2010. 177. Khalil N, Xu YD, O’Connor R, Duronio V. Proliferation of pulmonary interstitial fibroblasts is mediated by transforming growth factor-beta1induced release of extracellular fibroblast growth factor-2 and phosphorylation of p38 MAPK and JNK. J Biol Chem 280: 43000 –43009, 2005. 178. Khan Z, Khan N, Tiwari RP, Sah NK, Prasad GB, Bisen PS. Biology of Cox-2: an application in cancer therapeutics. Curr Drug Targets 12: 1082–1093, 2011. 179. Kikuchi Y, Kashima TG, Nishiyama T, Shimazu K, Morishita Y, Shimazaki M, Kii I, Horie H, Nagai H, Kudo A, Fukayama M. Periostin is expressed in pericryptal fibroblasts and cancer-associated fibroblasts in the colon. J Histochem Cytochem 56: 753–764, 2008. 180. Kim CJ, Yoshioka N, Tambe Y, Kushima R, Okada Y, Inoue H. Periostin is down-regulated in high grade human bladder cancers and suppresses in vitro cell invasiveness and in vivo metastasis of cancer cells. Int J Cancer 117: 51–58, 2005. 181. Kim JH, Kim HY, Kim S, Chung JH, Park WS, Chung DH. Natural killer T (NKT) cells attenuate bleomycin-induced pulmonary fibrosis by producing interferon-gamma. Am J Pathol 167: 1231–1241, 2005. 182. Kim KH, Chen CC, Monzon RI, Lau LF. Matricellular protein CCN1 promotes regression of liver fibrosis through induction of cellular senescence in hepatic myofibroblasts. Mol Cell Biol 33: 2078 –2090, 2013. 183. Klingberg F, Hinz B, White ES. The myofibroblast matrix: implications for tissue repair and fibrosis. J Pathol 229: 298 –309, 2013. 184. Koch AE, Polverini PJ, Kunkel SL, Harlow LA, DiPietro LA, Elner VM, Elner SG, Strieter RM. Interleukin-8 as a macrophage-derived mediator of angiogenesis. Science 258: 1798 –1801, 1992. 185. Kojima Y, Acar A, Eaton EN, Mellody KT, Scheel C, Ben-Porath I, Onder TT, Wang ZC, Richardson AL, Weinberg RA, Orimo A. Autocrine TGF-beta and stromal cell-derived factor-1 (SDF-1) signaling drives the evolution of tumor-promoting mammary stromal myofibroblasts. Proc Natl Acad Sci USA 107: 20009 –20014, 2010. 186. Kolb M, Margetts PJ, Sime PJ, Gauldie J. Proteoglycans decorin and biglycan differentially modulate TGF-beta-mediated fibrotic responses in the lung. Am J Physiol Lung Cell Mol Physiol 280: L1327–L1334, 2001. 187. Kolodsick JE, Peters-Golden M, Larios J, Toews GB, Thannickal VJ, Moore BB. Prostaglandin E2 inhibits fibroblast to myofibroblast transition via E. prostanoid receptor 2 signaling and cyclic adenosine monophosphate elevation Am J Resp Cell Mol Biol 29: 537–544, 2003. 188. Konstantinopoulos PA, Vandoros GP, Karamouzis MV, Gkermpesi M, Sotiropoulou-Bonikou G, Papavassiliou AG. EGF-R is expressed and AP-1 and NF-kappaB are activated in stromal myofibroblasts surrounding colon adenocarcinomas paralleling expression of COX-2 and VEGF. Cell Oncol 29: 477–482, 2007. 189. Koukourakis MI, Giatromanolaki A, Brekken RA, Sivridis E, Gatter KC, Harris AL, Sage EH. Enhanced expression of SPARC/osteonectin in the tumor-associated stroma of non-small cell lung cancer is correlated with markers of hypoxia/acidity and with poor prognosis of patients. Cancer Res 63: 5376 –5380, 2003. 190. Krizhanovsky V, Yon M, Dickins RA, Hearn S, Simon J, Miething C, Yee H, Zender L, Lowe SW. Senescence of activated stellate cells limits liver fibrosis. Cell 134: 657–667, 2008. 191. Kubo H, Hayashi T, Ago K, Ago M, Kanekura T, Ogata M. Temporal expression of wound healing-related genes in skin burn injury. Leg Med 16: 8 –13, 2014.

239

Review 240

THE WHFC TRIAD 236. Maniati E, Bossard M, Cook N, Candido JB, Emami-Shahri N, Nedospasov SA, Balkwill FR, Tuveson DA, Hagemann T. Crosstalk between the canonical NF-kappaB and Notch signaling pathways inhibits PPARgamma expression and promotes pancreatic cancer progression in mice. J Clin Invest 121: 4685–4699, 2011. 237. Marcos V, Zhou Z, Yildirim AO, Bohla A, Hector A, Vitkov L, Wiedenbauer EM, Krautgartner WD, Stoiber W, Belohradsky BH, Rieber N, Kormann M, Koller B, Roscher A, Roos D, Griese M, Eickelberg O, Doring G, Mall MA, Hartl D. CXCR2 mediates NADPH oxidase-independent neutrophil extracellular trap formation in cystic fibrosis airway inflammation. Nat Med 16: 1018 –1023, 2010. 238. Margadant C, Sonnenberg A. Integrin-TGF-beta crosstalk in fibrosis, cancer and wound healing. EMBO Rep 11: 97–105, 2010. 239. Marra F, Efsen E, Romanelli RG, Caligiuri A, Pastacaldi S, Batignani G, Bonacchi A, Caporale R, Laffi G, Pinzani M, Gentilini P. Ligands of peroxisome proliferator-activated receptor gamma modulate profibrogenic and proinflammatory actions in hepatic stellate cells. Gastroenterology 119: 466 –478, 2000. 240. Maruhashi T, Kii I, Saito M, Kudo A. Interaction between periostin and BMP-1 promotes proteolytic activation of lysyl oxidase. J Biol Chem 285: 13294 –13303, 2010. 241. Masamune A, Kikuta K, Satoh M, Sakai Y, Satoh A, Shimosegawa T. Ligands of peroxisome proliferator-activated receptor-gamma block activation of pancreatic stellate cells. J Biol Chem 277: 141–147, 2002. 242. Massague J. TGFbeta in cancer. Cell 134: 215–230, 2008. 243. Matise LA, Palmer TD, Ashby WJ, Nashabi A, Chytil A, Aakre M, Pickup MW, Gorska AE, Zijlstra A, Moses HL. Lack of transforming growth factor-beta signaling promotes collective cancer cell invasion through tumor-stromal crosstalk. Breast Cancer Res 14: R98, 2012. 244. McGrath MH. The effect of prostaglandin inhibitors on wound contraction and the myofibroblast. Plast Reconstr Surg 69: 74 –85, 1982. 245. Means AL, Ray KC, Singh AB, Washington MK, Whitehead RH, Harris RC Jr, Wright CV, Coffey RJ Jr, Leach SD. Overexpression of heparin-binding EGF-like growth factor in mouse pancreas results in fibrosis and epithelial metaplasia. Gastroenterology 124: 1020 –1036, 2003. 246. Meng F, Wang K, Aoyama T, Grivennikov SI, Paik Y, Scholten D, Cong M, Iwaisako K, Liu X, Zhang M, Osterreicher CH, Stickel F, Ley K, Brenner DA, Kisseleva T. Interleukin-17 signaling in inflammatory, Kupffer cells, and hepatic stellate cells exacerbates liver fibrosis in mice. Gastroenterology 143: 765–776, 2012. 247. Merline R, Lazaroski S, Babelova A, Tsalastra-Greul W, Pfeilschifter J, Schluter KD, Gunther A, Iozzo RV, Schaefer RM, Schaefer L. Decorin deficiency in diabetic mice: aggravation of nephropathy due to overexpression of profibrotic factors, enhanced apoptosis and mononuclear cell infiltration. J Physiol Pharmacol 60, Suppl 4: 5–13, 2009. 248. Michalik L, Wahli W. Involvement of PPAR nuclear receptors in tissue injury and wound repair. J Clin Invest 116: 598 –606, 2006. 249. Michaylira CZ, Wong GS, Miller CG, Gutierrez CM, Nakagawa H, Hammond R, Klein-Szanto AJ, Lee JS, Kim SB, Herlyn M, Diehl JA, Gimotty P, Rustgi AK. Periostin, a cell adhesion molecule, facilitates invasion in the tumor microenvironment and annotates a novel tumorinvasive signature in esophageal cancer. Cancer Res 70: 5281–5292, 2010. 250. Midwood KS, Hussenet T, Langlois B, Orend G. Advances in tenascin-C biology. Cell Mol Life Sci 68: 3175–3199, 2011. 251. Midwood KS, Orend G. The role of tenascin-C in tissue injury and tumorigenesis. J Cell Commun Signal 3: 287–310, 2009. 252. Milam JE, Keshamouni VG, Phan SH, Hu B, Gangireddy SR, Hogaboam CM, Standiford TJ, Thannickal VJ, Reddy RC. PPARgamma agonists inhibit profibrotic phenotypes in human lung fibroblasts and bleomycin-induced pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 294: L891–L901, 2008. 253. Milatovic S, Nanney LB, Yu Y, White JR, Richmond A. Impaired healing of nitrogen mustard wounds in CXCR2 null mice. Wound Repair Regen 11: 213–219, 2003. 254. Montesano R, Orci L. Transforming growth factor beta stimulates collagen-matrix contraction by fibroblasts: implications for wound healing. Proc Natl Acad Sci USA 85: 4894 –4897, 1988. 255. Morra L, Moch H. Periostin expression and epithelial-mesenchymal transition in cancer: a review and an update. Virchows Arch 459: 465–475, 2011. 256. Moulin V, Larochelle S, Langlois C, Thibault I, Lopez-Valle CA, Roy M. Normal skin wound and hypertrophic scar myofibroblasts have

Physiol Genomics • doi:10.1152/physiolgenomics.00158.2013 • www.physiolgenomics.org

Downloaded from on November 30, 2014

213. Li Y, Jiang D, Liang J, Meltzer EB, Gray A, Miura R, Wogensen L, Yamaguchi Y, Noble PW. Severe lung fibrosis requires an invasive fibroblast phenotype regulated by hyaluronan and CD44. J Exp Med 208: 1459 –1471, 2011. 214. Lian H, Ma Y, Feng J, Dong W, Yang Q, Lu D, Zhang L. Heparinbinding EGF-like growth factor induces heart interstitial fibrosis via an Akt/mTor/p70s6k pathway. PLoS One 7: e44946, 2012. 215. Lim KP, Cirillo N, Hassona Y, Wei W, Thurlow JK, Cheong SC, Pitiyage G, Parkinson EK, Prime SS. Fibroblast gene expression profile reflects the stage of tumour progression in oral squamous cell carcinoma. J Pathol 223: 459 –469, 2011. 216. Lin Y, Xie WF, Chen YX, Zhang X, Zeng X, Qiang H, Chen WZ, Yang XJ, Han ZG, Zhang ZB. Treatment of experimental hepatic fibrosis by combinational delivery of urokinase-type plasminogen activator and hepatocyte growth factor genes. Liver Int 25: 796 –807, 2005. 217. Lipponen P, Aaltomaa S, Tammi R, Tammi M, Agren U, Kosma VM. High stromal hyaluronan level is associated with poor differentiation and metastasis in prostate cancer. Eur J Cancer 37: 849 –856, 2001. 218. Liu F, Mih JD, Shea BS, Kho AT, Sharif AS, Tager AM, Tschumperlin DJ. Feedback amplification of fibrosis through matrix stiffening and COX-2 suppression. J Cell Biol 190: 693–706, 2010. 219. Liu Y. Cellular and molecular mechanisms of renal fibrosis. Nat Rev Nephrol 7: 684 –696, 2011. 220. Liu Y, Cai S, Shu XZ, Shelby J, Prestwich GD. Release of basic fibroblast growth factor from a crosslinked glycosaminoglycan hydrogel promotes wound healing. Wound Repair Regen 15: 245–251, 2007. 221. Liu Y, Li W, Li X, Tai Y, Lu Q, Yang N, Jiang J. Expression and significance of biglycan in endometrial cancer. Arch Gynecol Obstet [Epub ahead of print]. 222. Liu Y, Liu BA. Enhanced proliferation, invasion, and epithelial-mesenchymal transition of nicotine-promoted gastric cancer by periostin. World J Gastroenterol 17: 2674 –2680, 2011. 223. Liu Z, Hartman YE, Warram JM, Knowles JA, Sweeny L, Zhou T, Rosenthal EL. Fibroblast growth factor receptor mediates fibroblastdependent growth in EMMPRIN-depleted head and neck cancer tumor cells. Mol Cancer Res 9: 1008 –1017, 2011. 224. Lobry C, Oh P, Aifantis I. Oncogenic and tumor suppressor functions of Notch in cancer: it’s NOTCH what you think. J Exp Med 208: 1931–1935, 2011. 225. Lu K, Zhu Y, Sheng L, Liu L, Shen L, Wei Q. Serum fibrinogen level predicts the therapeutic response and prognosis in patients with locally advanced rectal cancer. Hepatogastroenterology 58: 1507–1510, 2011. 226. Lujambio A, Akkari L, Simon J, Grace D, Tschaharganeh DF, Bolden JE, Zhao Z, Thapar V, Joyce JA, Krizhanovsky V, Lowe SW. Non-cell-autonomous tumor suppression by p53. Cell 153: 449 –460, 2013. 227. Lynch SE, Nixon JC, Colvin RB, Antoniades HN. Role of plateletderived growth factor in wound healing: synergistic effects with other growth factors. Proc Natl Acad Sci USA 84: 7696 –7700, 1987. 228. Ma A, Zhao B, Boulton M, Albon J. A role for Notch signaling in corneal wound healing. Wound Repair Regen 19: 98 –106, 2011. 229. Madala SK, Schmidt S, Davidson C, Ikegami M, Wert S, Hardie WD. MEK-ERK pathway modulation ameliorates pulmonary fibrosis associated with epidermal growth factor receptor activation. Am J Respir Cell Mol Biol 46: 380 –388, 2012. 230. Maharaj S, Shimbori C, Kolb M. Fibrocytes in pulmonary fibrosis: a brief synopsis. Eur Respir Rev 22: 552–557, 2013. 231. Maihofner C, Charalambous MP, Bhambra U, Lightfoot T, Geisslinger G, Gooderham NJ. Expression of cyclooxygenase-2 parallels expression of interleukin-1beta, interleukin-6 and NF-kappaB in human colorectal cancer. Carcinogenesis 24: 665–671, 2003. 232. Maisonneuve P, Marshall BC, Lowenfels AB. Risk of pancreatic cancer in patients with cystic fibrosis. Gut 56: 1327–1328, 2007. 233. Maki JM, Sormunen R, Lippo S, Kaarteenaho-Wiik R, Soininen R, Myllyharju J. Lysyl oxidase is essential for normal development and function of the respiratory system and for the integrity of elastic and collagen fibers in various tissues. Am J Pathol 167: 927–936, 2005. 234. Maltseva O, Folger P, Zekaria D, Petridou S, Masur SK. Fibroblast growth factor reversal of the corneal myofibroblast phenotype. Invest Ophthalmol Vis Sci 42: 2490 –2495, 2001. 235. Maneva-Radicheva L, Ebert U, Dimoudis N, Altankov G. Fibroblast remodeling of adsorbed collagen type IV is altered in contact with cancer cells. Histol Histopathol 23: 833–842, 2008.

Review THE WHFC TRIAD

257. 258.

259.

260.

261.

262.

264. 265. 266.

267.

268. 269. 270.

271.

272.

273.

274. 275.

276.

277.

278.

279.

280.

281.

282.

283.

284.

285.

286. 287.

288. 289.

290.

291.

292.

293.

294.

295.

296.

growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell 121: 335–348, 2005. Oskarsson T, Acharyya S, Zhang XH, Vanharanta S, Tavazoie SF, Morris PG, Downey RJ, Manova-Todorova K, Brogi E, Massague J. Breast cancer cells produce tenascin C as a metastatic niche component to colonize the lungs. Nat Med 17: 867–874, 2011. Otranto M, Sarrazy V, Bonte F, Hinz B, Gabbiani G, Desmouliere A. The role of the myofibroblast in tumor stroma remodeling. Cell Adhes Migrat 6: 203–219, 2012. Page-McCaw A, Ewald AJ, Werb Z. Matrix metalloproteinases and the regulation of tissue remodelling. Nat Rev Mol Cell Biol 8: 221–233, 2007. Paron I, Berchtold S, Voros J, Shamarla M, Erkan M, Hofler H, Esposito I. Tenascin-C enhances pancreatic cancer cell growth and motility and affects cell adhesion through activation of the integrin pathway. PLoS One 6: e21684, 2011. Parsian H, Rahimipour A, Nouri M, Somi MH, Qujeq D, Fard MK, Agcheli K. Serum hyaluronic acid and laminin as biomarkers in liver fibrosis. J Gastrointest Liver Dis 19: 169 –174, 2010. Pastore S, Mascia F, Mariani V, Girolomoni G. The epidermal growth factor receptor system in skin repair and inflammation. J Invest Dermatol 128: 1365–1374, 2008. Paszek MJ, Zahir N, Johnson KR, Lakins JN, Rozenberg GI, Gefen A, Reinhart-King CA, Margulies SS, Dembo M, Boettiger D, Hammer DA, Weaver VM. Tensional homeostasis and the malignant phenotype. Cancer Cell 8: 241–254, 2005. Peinado H, Moreno-Bueno G, Hardisson D, Perez-Gomez E, Santos V, Mendiola M, de Diego JI, Nistal M, Quintanilla M, Portillo F, Cano A. Lysyl oxidase-like 2 as a new poor prognosis marker of squamous cell carcinomas. Cancer Res 68: 4541–4550, 2008. Phillips RJ, Burdick MD, Hong K, Lutz MA, Murray LA, Xue YY, Belperio JA, Keane MP, Strieter RM. Circulating fibrocytes traffic to the lungs in response to CXCL12 and mediate fibrosis. J Clin Invest 114: 438 –446, 2004. Piccioni F, Malvicini M, Garcia MG, Rodriguez A, Atorrasagasti C, Kippes N, Piedra Buena IT, Rizzo MM, Bayo J, Aquino J, Viola M, Passi A, Alaniz L, Mazzolini G. Antitumor effects of hyaluronic acid inhibitor 4-methylumbelliferone in an orthotopic hepatocellular carcinoma model in mice. Glycobiology 22: 400 –410, 2012. Pietras K, Ostman A. Hallmarks of cancer: interactions with the tumor stroma. Exp Cell Res 316: 1324 –1331, 2010. Pietras K, Pahler J, Bergers G, Hanahan D. Functions of paracrine PDGF signaling in the proangiogenic tumor stroma revealed by pharmacological targeting. PLoS Med 5: e19, 2008. Pinzani M, Rombouts K. Liver fibrosis: from the bench to clinical targets. Dig Liver Dis 36: 231–242, 2004. Pirinen R, Tammi R, Tammi M, Hirvikoski P, Parkkinen JJ, Johansson R, Bohm J, Hollmen S, Kosma VM. Prognostic value of hyaluronan expression in non-small-cell lung cancer: Increased stromal expression indicates unfavorable outcome in patients with adenocarcinoma. Int J Cancer 95: 12–17, 2001. Polterauer S, Grimm C, Seebacher V, Concin N, Marth C, Tomovski C, Husslein H, Leipold H, Hefler-Frischmuth K, Tempfer C, Reinthaller A, Hefler L. Plasma fibrinogen levels and prognosis in patients with ovarian cancer: a multicenter study. Oncologist 14: 979 –985, 2009. Porsch H, Bernert B, Mehic M, Theocharis AD, Heldin CH, Heldin P. Efficient TGFbeta-induced epithelial-mesenchymal transition depends on hyaluronan synthase HAS2. Oncogene 32: 4355–4365, 2013. Prajapati RT, Chavally-Mis B, Herbage D, Eastwood M, Brown RA. Mechanical loading regulates protease production by fibroblasts in threedimensional collagen substrates. Wound Repair Regen 8: 226 –237, 2000. Preaux AM, Mallat A, Nhieu JT, D’Ortho MP, Hembry RM, Mavier P. Matrix metalloproteinase-2 activation in human hepatic fibrosis regulation by cell-matrix interactions. Hepatology 30: 944 –950, 1999. Provenzano PP, Eliceiri KW, Campbell JM, Inman DR, White JG, Keely PJ. Collagen reorganization at the tumor-stromal interface facilitates local invasion. BMC Med 4: 38, 2006. Provenzano PP, Inman DR, Eliceiri KW, Knittel JG, Yan L, Rueden CT, White JG, Keely PJ. Collagen density promotes mammary tumor initiation and progression. BMC Med 6: 11, 2008. Qiu J, Yu Y, Fu Y, Ye F, Xie X, Lu W. Preoperative plasma fibrinogen, platelet count and prognosis in epithelial ovarian cancer. J Obstet Gynaecol Res 38: 651–657, 2012.

Physiol Genomics • doi:10.1152/physiolgenomics.00158.2013 • www.physiolgenomics.org

Downloaded from on November 30, 2014

263.

differential responses to apoptotic inductors. J Cell Physiol 198: 350 – 358, 2004. Munger JS, Sheppard D. Cross talk among TGF-beta signaling pathways, integrins, and the extracellular matrix. Cold Spring Harbor Perspect Biol 3: a005017, 2011. Murakami T, Kawada K, Iwamoto M, Akagami M, Hida K, Nakanishi Y, Kanda K, Kawada M, Seno H, Taketo MM, Sakai Y. The role of CXCR3 and CXCR4 in colorectal cancer metastasis. Int J Cancer 132: 276 –287, 2013. Murata T, Mizushima H, Chinen I, Moribe H, Yagi S, Hoffman RM, Kimura T, Yoshino K, Ueda Y, Enomoto T, Mekada E. HB-EGF and PDGF mediate reciprocal interactions of carcinoma cells with cancerassociated fibroblasts to support progression of uterine cervical cancers. Cancer Res 71: 6633–6642, 2011. Nagy JA, Brown LF, Senger DR, Lanir N, Van de Water L, Dvorak AM, Dvorak HF. Pathogenesis of tumor stroma generation: a critical role for leaky blood vessels and fibrin deposition. Biochim Biophys Acta 948: 305–326, 1989. Naik PK, Bozyk PD, Bentley JK, Popova AP, Birch CM, Wilke CA, Fry CD, White ES, Sisson TH, Tayob N, Carnemolla B, Orecchia P, Flaherty KR, Hershenson MB, Murray S, Martinez FJ, Moore BB. Periostin promotes fibrosis and predicts progression in patients with idiopathic pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 303: L1046 –L1056, 2012. Nakaya I, Wada T, Furuichi K, Sakai N, Kitagawa K, Yokoyama H, Ishida Y, Kondo T, Sugaya T, Kawachi H, Shimizu F, Narumi S, Haino M, Gerard C, Matsushima K, Kaneko S. Blockade of IP-10/ CXCR3 promotes progressive renal fibrosis. Nephron Exp Nephrol 107: e12–e21, 2007. Neesse A, Krug S, Gress TM, Tuveson DA, Michl P. Emerging concepts in pancreatic cancer medicine: targeting the tumor stroma. OncoTargets Ther 7: 33–43, 2013. Neill T, Schaefer L, Iozzo RV. Decorin: a guardian from the matrix. Am J Pathol 181: 380 –387, 2012. Ng CP, Hinz B, Swartz MA. Interstitial fluid flow induces myofibroblast differentiation and collagen alignment in vitro. J Cell Sci 118: 4731– 4739, 2005. Niedworok C, Rock K, Kretschmer I, Freudenberger T, Nagy N, Szarvas T, Vom Dorp F, Reis H, Rubben H, Fischer JW. Inhibitory role of the small leucine-rich proteoglycan biglycan in bladder cancer. PLoS One 8: e80084, 2013. Nikitovic D, Berdiaki A, Banos A, Tsatsakis A, Karamanos NK, Tzanakakis GN. Could growth factor-mediated extracellular matrix deposition and degradation offer the ground for directed pharmacological targeting in fibrosarcoma? Curr Med Chem 20: 2868 –2880, 2013. Nishida M, Okumura Y, Ozawa S, Shiraishi I, Itoi T, Hamaoka K. MMP-2 inhibition reduces renal macrophage infiltration with increased fibrosis in UUO. Biochem Biophys Res Commun 354: 133–139, 2007. Noble PW, Barkauskas CE, Jiang D. Pulmonary fibrosis: patterns and perpetrators. J Clin Invest 122: 2756 –2762, 2012. Norris RA, Damon B, Mironov V, Kasyanov V, Ramamurthi A, Moreno-Rodriguez R, Trusk T, Potts JD, Goodwin RL, Davis J, Hoffman S, Wen X, Sugi Y, Kern CB, Mjaatvedt CH, Turner DK, Oka T, Conway SJ, Molkentin JD, Forgacs G, Markwald RR. Periostin regulates collagen fibrillogenesis and the biomechanical properties of connective tissues. J Cell Biochem 101: 695–711, 2007. Novitskiy SV, Pickup MW, Gorska AE, Owens P, Chytil A, Aakre M, Wu H, Shyr Y, Moses HL. TGF-beta receptor II loss promotes mammary carcinoma progression by Th17 dependent mechanisms. Cancer Discov 1: 430 –441, 2011. Ohbayashi M, Suzuki M, Yashiro Y, Fukuwaka S, Yasuda M, Kohyama N, Kobayashi Y, Yamamoto T. Induction of pulmonary fibrosis by methotrexate treatment in mice lung in vivo and in vitro. J Toxicol Sci 35: 653–661, 2010. Okuda H, Kobayashi A, Xia B, Watabe M, Pai SK, Hirota S, Xing F, Liu W, Pandey PR, Fukuda K, Modur V, Ghosh A, Wilber A, Watabe K. Hyaluronan synthase HAS2 promotes tumor progression in bone by stimulating the interaction of breast cancer stem-like cells with macrophages and stromal cells. Cancer Res 72: 537–547, 2012. Orend G, Chiquet-Ehrismann R. Tenascin-C induced signaling in cancer. Cancer Lett 244: 143–163, 2006. Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, Carey VJ, Richardson AL, Weinberg RA. Stromal fibroblasts present in invasive human breast carcinomas promote tumor

241

Review 242

THE WHFC TRIAD 316. Schaefer L, Macakova K, Raslik I, Micegova M, Grone HJ, Schonherr E, Robenek H, Echtermeyer FG, Grassel S, Bruckner P, Schaefer RM, Iozzo RV, Kresse H. Absence of decorin adversely influences tubulointerstitial fibrosis of the obstructed kidney by enhanced apoptosis and increased inflammatory reaction. Am J Pathol 160: 1181– 1191, 2002. 317. Schafer M, Werner S. Cancer as an overhealing wound: an old hypothesis revisited. Nat Rev Mol Cell Biol 9: 628 –638, 2008. 318. Schaffner F, Ruf W. Tissue factor and PAR2 signaling in the tumor microenvironment. Arterioscler Thromb Vasc Biol 29: 1999 –2004, 2009. 319. Schietke R, Warnecke C, Wacker I, Schodel J, Mole DR, Campean V, Amann K, Goppelt-Struebe M, Behrens J, Eckardt KU, Wiesener MS. The lysyl oxidases LOX and LOXL2 are necessary and sufficient to repress E-cadherin in hypoxia: insights into cellular transformation processes mediated by HIF-1. J Biol Chem 285: 6658 –6669, 2010. 320. Schliemann C, Neri D. Antibody-based targeting of the tumor vasculature. Biochim Biophys Acta 1776: 175–192, 2007. 321. Seebacher V, Polterauer S, Grimm C, Husslein H, Leipold H, HeflerFrischmuth K, Tempfer C, Reinthaller A, Hefler L. The prognostic value of plasma fibrinogen levels in patients with endometrial cancer: a multi-centre trial. Br J Cancer 102: 952–956, 2010. 322. Selman M, Ruiz V, Cabrera S, Segura L, Ramirez R, Barrios R, Pardo A. TIMP-1, -2, -3, and -4 in idiopathic pulmonary fibrosis. A prevailing nondegradative lung microenvironment? Am J Physiol Lung Cell Mol Physiol 279: L562–L574, 2000. 323. Sen K, Lindenmeyer MT, Gaspert A, Eichinger F, Neusser MA, Kretzler M, Segerer S, Cohen CD. Periostin is induced in glomerular injury and expressed de novo in interstitial renal fibrosis. Am J Pathol 179: 1756 –1767, 2011. 324. Serini G, Bochaton-Piallat ML, Ropraz P, Geinoz A, Borsi L, Zardi L, Gabbiani G. The fibronectin domain ED-A is crucial for myofibroblastic phenotype induction by transforming growth factor-beta1. J Cell Biol 142: 873–881, 1998. 325. Setoguchi T, Kikuchi H, Yamamoto M, Baba M, Ohta M, Kamiya K, Tanaka T, Baba S, Goto-Inoue N, Setou M, Sasaki T, Mori H, Sugimura H, Konno H. Microarray analysis identifies versican and CD9 as potent prognostic markers in gastric gastrointestinal stromal tumors. Cancer Sci 102: 883–889, 2011. 326. Shao ZM, Nguyen M, Barsky SH. Human breast carcinoma desmoplasia is PDGF initiated. Oncogene 19: 4337–4345, 2000. 327. Shariat SF, Kattan MW, Traxel E, Andrews B, Zhu K, Wheeler TM, Slawin KM. Association of pre- and postoperative plasma levels of transforming growth factor beta(1) and interleukin 6 and its soluble receptor with prostate cancer progression. Clin Cancer Res 10: 1992– 1999, 2004. 328. Shaw A, Gipp J, Bushman W. The Sonic Hedgehog pathway stimulates prostate tumor growth by paracrine signaling and recapitulates embryonic gene expression in tumor myofibroblasts. Oncogene 28: 4480 –4490, 2009. 329. Shen J, Gao J, Chen C, Lu H, Hu G, Shen J, Zhu S, Wu M, Wang X, Qian L, Yu Y, Han W, Wan R, Wang X. Antifibrotic role of chemokine CXCL9 in experimental chronic pancreatitis induced by trinitrobenzene sulfonic acid in rats. Cytokine 64: 382–394, 2013. 330. Shimada M, Andoh A, Hata K, Tasaki K, Araki Y, Fujiyama Y, Bamba T. IL-6 secretion by human pancreatic periacinar myofibroblasts in response to inflammatory mediators. J Immunol 168: 861–868, 2002. 331. Sidhu SS, Yuan S, Innes AL, Kerr S, Woodruff PG, Hou L, Muller SJ, Fahy JV. Roles of epithelial cell-derived periostin in TGF-beta activation, collagen production, and collagen gel elasticity in asthma. Proc Natl Acad Sci USA 107: 14170 –14175, 2010. 332. Sime PJ. The antifibrogenic potential of PPARgamma ligands in pulmonary fibrosis. J Investig Med 56: 534 –538, 2008. 333. Sironen RK, Tammi M, Tammi R, Auvinen PK, Anttila M, Kosma VM. Hyaluronan in human malignancies. Exp Cell Res 317: 383–391, 2011. 334. Sisson TH, Hattori N, Xu Y, Simon RH. Treatment of bleomycininduced pulmonary fibrosis by transfer of urokinase-type plasminogen activator genes. Hum Gene Ther 10: 2315–2323, 1999. 335. Smith HW, Marshall CJ. Regulation of cell signalling by uPAR. Nat Rev Mol Cell Biol 11: 23–36, 2010. 336. Sobolik T, Su YJ, Wells S, Ayers GD, Cook RS, Richmond A. CXCR4 drives the metastatic phenotype in breast cancer through induction of CXCR2, and activation of MEK and PI3K pathways. Mol Biol Cell [Epub ahead of print].

Physiol Genomics • doi:10.1152/physiolgenomics.00158.2013 • www.physiolgenomics.org

Downloaded from on November 30, 2014

297. Reka AK, Kurapati H, Narala VR, Bommer G, Chen J, Standiford TJ, Keshamouni VG. Peroxisome proliferator-activated receptorgamma activation inhibits tumor metastasis by antagonizing Smad3mediated epithelial-mesenchymal transition. Mol Cancer Therapeut 9: 3221–3232, 2010. 298. Rhodes LV, Antoon JW, Muir SE, Elliott S, Beckman BS, Burow ME. Effects of human mesenchymal stem cells on ER-positive human breast carcinoma cells mediated through ER-SDF-1/CXCR4 crosstalk. Mol Cancer 9: 295, 2010. 299. Rhodes LV, Bratton MR, Zhu Y, Tilghman SL, Muir SE, Salvo VA, Tate CR, Elliott S, Nephew KP, Collins-Burow BM, Burow ME. Effects of SDF-1-CXCR4 signaling on microRNA expression and tumorigenesis in estrogen receptor-alpha (ER-alpha)-positive breast cancer cells. Exp Cell Res 317: 2573–2581, 2011. 300. Rhodes LV, Short SP, Neel NF, Salvo VA, Zhu Y, Elliott S, Wei Y, Yu D, Sun M, Muir SE, Fonseca JP, Bratton MR, Segar C, Tilghman SL, Sobolik-Delmaire T, Horton LW, Zaja-Milatovic S, CollinsBurow BM, Wadsworth S, Beckman BS, Wood CE, Fuqua SA, Nephew KP, Dent P, Worthylake RA, Curiel TJ, Hung MC, Richmond A, Burow ME. Cytokine receptor CXCR4 mediates estrogenindependent tumorigenesis, metastasis, and resistance to endocrine therapy in human breast cancer. Cancer Res 71: 603–613, 2011. 301. Ricciardelli C, Sakko AJ, Ween MP, Russell DL, Horsfall DJ. The biological role and regulation of versican levels in cancer. Cancer Metast Rev 28: 233–245, 2009. 302. Richter CM, Godes M, Wagner C, Maser-Gluth C, Herzfeld S, Dorn M, Priem F, Slowinski T, Bauer C, Schneider W, Neumayer HH, Kurtz A, Hocher B. Chronic cyclooxygenase-2 inhibition does not alter blood pressure and kidney function in renovascular hypertensive rats. J Hypertens 22: 191–198, 2004. 303. Roda O, Ortiz-Zapater E, Martinez-Bosch N, Gutierrez-Gallego R, Vila-Perello M, Ampurdanes C, Gabius HJ, Andre S, Andreu D, Real FX, Navarro P. Galectin-1 is a novel functional receptor for tissue plasminogen activator in pancreatic cancer. Gastroenterology 136: 1379 –1390, 2009. 304. Ropponen K, Tammi M, Parkkinen J, Eskelinen M, Tammi R, Lipponen P, Agren U, Alhava E, Kosma VM. Tumor cell-associated hyaluronan as an unfavorable prognostic factor in colorectal cancer. Cancer Res 58: 342–347, 1998. 305. Roy M, Pear WS, Aster JC. The multifaceted role of Notch in cancer. Curr Opin Genet Dev 17: 52–59, 2007. 306. Ruan K, Bao S, Ouyang G. The multifaceted role of periostin in tumorigenesis. Cell Mol Life Sci 66: 2219 –2230, 2009. 307. Ruoslahti E, Vaheri A, Kuusela P, Linder E. Fibroblast surface antigen: a new serum protein. Biochim Biophys Acta 322: 352–358, 1973. 308. Ruppert C, Markart P, Wygrecka M, Preissner KT, Gunther A. Role of coagulation and fibrinolysis in lung and renal fibrosis. Hamostaseologie 28: 30 –36, 2008. 309. Said N, Frierson HF, Sanchez-Carbayo M, Brekken RA, Theodorescu D. Loss of SPARC in bladder cancer enhances carcinogenesis and progression. J Clin Invest 123: 751–766, 2013. 310. Said N, Sanchez-Carbayo M, Smith SC, Theodorescu D. RhoGDI2 suppresses lung metastasis in mice by reducing tumor versican expression and macrophage infiltration. J Clin Invest 122: 1503–1518, 2012. 311. Saito-Fujita T, Iwakawa M, Nakamura E, Nakawatari M, Fujita H, Moritake T, Imai T. Attenuated lung fibrosis in interleukin 6 knock-out mice after C-ion irradiation to lung. J Radiat Res (Tokyo) 52: 270 –277, 2011. 312. Sakuma K, Aoki M, Kannagi R. Transcription factors c-Myc and CDX2 mediate E-selectin ligand expression in colon cancer cells undergoing EGF/bFGF-induced epithelial-mesenchymal transition. Proc Natl Acad Sci USA 109: 7776 –7781, 2012. 313. Salah Z, Uziely B, Jaber M, Maoz M, Cohen I, Hamburger T, Maly B, Peretz T, Bar-Shavit R. Regulation of human protease-activated receptor 1 (hPar1) gene expression in breast cancer by estrogen. FASEB J 26: 2031–2042, 2012. 314. Sasaki T, Nakamura T, Rebhun RB, Cheng H, Hale KS, Tsan RZ, Fidler IJ, Langley RR. Modification of the primary tumor microenvironment by transforming growth factor alpha-epidermal growth factor receptor signaling promotes metastasis in an orthotopic colon cancer model. Am J Pathol 173: 205–216, 2008. 315. Sasser AK, Sullivan NJ, Studebaker AW, Hendey LF, Axel AE, Hall BM. Interleukin-6 is a potent growth factor for ER-alpha-positive human breast cancer. FASEB J 21: 3763–3770, 2007.

Review THE WHFC TRIAD

359. Theunissen JW, de Sauvage FJ. Paracrine hedgehog signaling in cancer. Cancer Res 69: 6007–6010, 2009. 360. Tian Z, Chen Y, Gao B. Natural killer cells in liver disease. Hepatology 57: 1654 –1662, 2013. 361. Tiggelman AM, Boers W, Linthorst C, Brand HS, Sala M, Chamuleau RA. Interleukin-6 production by human liver (myo)fibroblasts in culture. Evidence for a regulatory role of LPS, IL-1 beta and TNF alpha. J Hepatol 23: 295–306, 1995. 362. Tiwari N, Gheldof A, Tatari M, Christofori G. EMT as the ultimate survival mechanism of cancer cells. Sem Cancer Biol 22: 194 –207, 2012. 363. Tolino MA, Block ER, Klarlund JK. Brief treatment with heparinbinding EGF-like growth factor, but not with EGF, is sufficient to accelerate epithelial wound healing. Biochim Biophys Acta 1810: 875– 878, 2011. 364. Tomasek JJ, Gabbiani G, Hinz B, Chaponnier C, Brown RA. Myofibroblasts and mechano-regulation of connective tissue remodelling. Nat Rev Mol Cell Biol 3: 349 –363, 2002. 365. Trombetta-Esilva J, Bradshaw AD. The function of SPARC as a mediator of fibrosis. Open Rheumatol J 6: 146 –155, 2012. 366. Trotta R, Dal Col J, Yu J, Ciarlariello D, Thomas B, Zhang X, Allard J 2nd, Wei M, Mao H, Byrd JC, Perrotti D, Caligiuri MA. TGF-beta utilizes SMAD3 to inhibit CD16-mediated IFN-gamma production and antibody-dependent cellular cytotoxicity in human NK cells. J Immunol 181: 3784 –3792, 2008. 367. Tsunoda S, Sakurai H, Saito Y, Ueno Y, Koizumi K, Saiki I. Massive T-lymphocyte infiltration into the host stroma is essential for fibroblast growth factor-2-promoted growth and metastasis of mammary tumors via neovascular stability. Am J Pathol 174: 671–683, 2009. 368. Turner N, Grose R. Fibroblast growth factor signalling: from development to cancer. Nat Rev Cancer 10: 116 –129, 2010. 369. Utispan K, Thuwajit P, Abiko Y, Charngkaew K, Paupairoj A, Chau-in S, Thuwajit C. Gene expression profiling of cholangiocarcinoma-derived fibroblast reveals alterations related to tumor progression and indicates periostin as a poor prognostic marker. Mol Cancer 9: 13, 2010. 370. Valdivia-Silva J, Franco-Barraza J, Cukierman E, Garcia-Zepeda E. Novel insights into the role of inflammation in promoting breast cancer development. In: Breast Cancer - Focusing Tumor Microenvironment, Stem Cells and Metastasis, edited by Gunduz M, and Gunduz E. InTech, 2011, p. 129 –164. 371. Valdivia-Silva JE, Franco-Barraza J, Silva AL, Pont GD, Soldevila G, Meza I, Garcia-Zepeda EA. Effect of pro-inflammatory cytokine stimulation on human breast cancer: implications of chemokine receptor expression in cancer metastasis. Cancer Lett 283: 176 –185, 2009. 372. Vandoros GP, Konstantinopoulos PA, Sotiropoulou-Bonikou G, Kominea A, Papachristou GI, Karamouzis MV, Gkermpesi M, Varakis I, Papavassiliou AG. PPAR-gamma is expressed and NF-kB pathway is activated and correlates positively with COX-2 expression in stromal myofibroblasts surrounding colon adenocarcinomas. J Cancer Res Clin Oncol 132: 76 –84, 2006. 373. Venkatesan N, Tsuchiya K, Kolb M, Farkas L, Bourhim M, Ouzzine M, Ludwig MS. Glycosyltransferases and glycosaminoglycans in bleomycin and transforming growth factor-␤1-induced pulmonary fibrosis. Am J Resp Cell Mol Biol [Epub ahead of print]. 374. Vial C, Gutierrez J, Santander C, Cabrera D, Brandan E. Decorin interacts with connective tissue growth factor (CTGF)/CCN2 by LRR12 inhibiting its biological activity. J Biol Chem 286: 24242–24252, 2011. 375. Wallach-Dayan SB, Golan-Gerstl R, Breuer R. Evasion of myofibroblasts from immune surveillance: a mechanism for tissue fibrosis. Proc Natl Acad Sci USA 104: 20460 –20465, 2007. 376. Walter K, Omura N, Hong SM, Griffith M, Vincent A, Borges M, Goggins M. Overexpression of smoothened activates the sonic hedgehog signaling pathway in pancreatic cancer-associated fibroblasts. Clin Cancer Res 16: 1781–1789, 2010. 377. Wang B, Li GX, Zhang SG, Wang Q, Wen YG, Tang HM, Zhou CZ, Xing AY, Fan JW, Yan DW, Qiu GQ, Yu ZH, Peng ZH. Biglycan expression correlates with aggressiveness and poor prognosis of gastric cancer. Exp Biol Med (Maywood) 236: 1247–1253, 2011. 378. Wang H, Haeger SM, Kloxin AM, Leinwand LA, Anseth KS. Redirecting valvular myofibroblasts into dormant fibroblasts through lightmediated reduction in substrate modulus. PLoS One 7: e39969, 2012. 379. Wang K, Grivennikov SI, Karin M. Implications of anti-cytokine therapy in colorectal cancer and autoimmune diseases. Ann Rheum Dis 72: ii100 –ii103, 2013.

Physiol Genomics • doi:10.1152/physiolgenomics.00158.2013 • www.physiolgenomics.org

Downloaded from on November 30, 2014

337. Sorensen I, Susnik N, Inhester T, Degen JL, Melk A, Haller H, Schmitt R. Fibrinogen, acting as a mitogen for tubulointerstitial fibroblasts, promotes renal fibrosis. Kidney Int 80: 1035–1044, 2011. 338. Spencer VA, Xu R, Bissell MJ. Gene expression in the third dimension: the ECM-nucleus connection. J Mamm Gland Biol Neoplas 15: 65–71, 2010. 339. Spyrou GE, Naylor IL. The effect of basic fibroblast growth factor on scarring. Br J Plast Surg 55: 275–282, 2002. 340. Sterclova M, Vasakova M, Pavlicek J, Metlicka M, Krasna E, Striz I. Chemokine receptors in a regulation of interstitial lung fibrosis and inflammation. Exp Lung Res 35: 514 –523, 2009. 341. Stern R. Hyaluronidases in cancer biology. Sem Cancer Biol 18: 275– 280, 2008. 342. Stern R, Asari AA, Sugahara KN. Hyaluronan fragments: an information-rich system. Eur J Cell Biol 85: 699 –715, 2006. 343. Strieter RM, Keeley EC, Burdick MD, Mehrad B. The role of circulating mesenchymal progenitor cells, fibrocytes, in promoting pulmonary fibrosis. Trans Am Clin Climatol Assoc 120: 49 –59, 2009. 344. Strutz F, Zeisberg M, Hemmerlein B, Sattler B, Hummel K, Becker V, Muller GA. Basic fibroblast growth factor expression is increased in human renal fibrogenesis and may mediate autocrine fibroblast proliferation. Kidney Int 57: 1521–1538, 2000. 345. Strutz F, Zeisberg M, Renziehausen A, Raschke B, Becker V, van Kooten C, Muller G. TGF-beta 1 induces proliferation in human renal fibroblasts via induction of basic fibroblast growth factor (FGF-2). Kidney Int 59: 579 –592, 2001. 346. Strutz F, Zeisberg M, Ziyadeh FN, Yang CQ, Kalluri R, Muller GA, Neilson EG. Role of basic fibroblast growth factor-2 in epithelialmesenchymal transformation. Kidney Int 61: 1714 –1728, 2002. 347. Su X, Matthay MA. Role of protease activated receptor 2 in experimental acute lung injury and lung fibrosis. Anatom Rec (Hoboken) 292: 580 –586, 2009. 348. Sullivan NJ, Sasser AK, Axel AE, Vesuna F, Raman V, Ramirez N, Oberyszyn TM, Hall BM. Interleukin-6 induces an epithelial-mesenchymal transition phenotype in human breast cancer cells. Oncogene 28: 2940 –2947, 2009. 349. Sun X, Fa P, Cui Z, Xia Y, Sun L, Li Z, Tang A, Gui Y, Cai Z. The EDA-containing cellular fibronectin induces epithelial-mesenchymal transition in lung cancer cells through integrin ␣9␤1-mediated activation of PI3-K/Akt and Erk1/2. Carcinogenesis 35: 184 –191, 2014. 350. Suzuki K, Wang R, Kubota H, Shibuya H, Saegusa J, Sato T. Kinetics of biglycan, decorin and thrombospondin-1 in mercuric chloride-induced renal tubulointerstitial fibrosis. Exp Mol Pathol 79: 68 –73, 2005. 351. Svensson M, Yadav M, Holmqvist B, Lutay N, Svanborg C, Godaly G. Acute pyelonephritis and renal scarring are caused by dysfunctional innate immunity in mCxcr2 heterozygous mice. Kidney Int 80: 1064 – 1072, 2011. 352. Tacke F, Zimmermann HW, Berres ML, Trautwein C, Wasmuth HE. Serum chemokine receptor CXCR3 ligands are associated with progression, organ dysfunction and complications of chronic liver diseases. Liver Int 31: 840 –849, 2011. 353. Takahashi Y, Kuwabara H, Yoneda M, Isogai Z, Tanigawa N, Shibayama Y. Versican G1 and G3 domains are upregulated and latent transforming growth factor-beta binding protein-4 is downregulated in breast cancer stroma. Breast Cancer 19: 46 –53, 2012. 354. Takeuchi K, Tanaka A, Kato S, Amagase K, Satoh H. Roles of COX inhibition in pathogenesis of NSAID-induced small intestinal damage. Clinica Chimica Acta 411: 459 –466, 2010. 355. Tammi RH, Kultti A, Kosma VM, Pirinen R, Auvinen P, Tammi MI. Hyaluronan in human tumors: pathobiological and prognostic messages from cell-associated and stromal hyaluronan. Sem Cancer Biol 18: 288 –295, 2008. 356. Tang L, Liu K, Wang J, Wang C, Zhao P, Liu J. High preoperative plasma fibrinogen levels are associated with distant metastases and impaired prognosis after curative resection in patients with colorectal cancer. J Surg Oncol 102: 428 –432, 2010. 357. Tangkijvanich P, Kongtawelert P, Pothacharoen P, Mahachai V, Suwangool P, Poovorawan Y. Serum hyaluronan: a marker of liver fibrosis in patients with chronic liver disease. Asian Pac J Allergy Immunol 21: 115–120, 2003. 358. Tani K, Yasuoka S, Ogushi F, Asada K, Fujisawa K, Ozaki T, Sano N, Ogura T. Thrombin enhances lung fibroblast proliferation in bleomycin-induced pulmonary fibrosis. Am J Respir Cell Mol Biol 5: 34 –40, 1991.

243

Review 244

THE WHFC TRIAD 400. Yamada KM, Olden K, Pastan I. Transformation-sensitive cell surface protein: isolation, characterization, and role in cellular morphology and adhesion. Ann NY Acad Sci 312: 256 –277, 1978. 401. Yamaguchi Y, Ono J, Masuoka M, Ohta S, Izuhara K, Ikezawa Z, Aihara M, Takahashi K. Serum periostin levels are correlated with progressive skin sclerosis in patients with systemic sclerosis. Brit J Dermat 168: 717–725, 2013. 402. Yan W, Wang P, Zhao CX, Tang J, Xiao X, Wang DW. Decorin gene delivery inhibits cardiac fibrosis in spontaneously hypertensive rats by modulation of transforming growth factor-beta/Smad and p38 mitogenactivated protein kinase signaling pathways. Hum Gene Ther 20: 1190 – 1200, 2009. 403. Yang F, Strand DW, Rowley DR. Fibroblast growth factor-2 mediates transforming growth factor-beta action in prostate cancer reactive stroma. Oncogene 27: 450 –459, 2008. 404. Yang IV. Epigenomics of idiopathic pulmonary fibrosis. Epigenomics 4: 195–203, 2012. 405. Yang J, Shultz RW, Mars WM, Wegner RE, Li Y, Dai C, Nejak K, Liu Y. Disruption of tissue-type plasminogen activator gene in mice reduces renal interstitial fibrosis in obstructive nephropathy. J Clin Invest 110: 1525–1538, 2002. 406. Yang X, Hou J, Han Z, Wang Y, Hao C, Wei L, Shi Y. One cell, multiple roles: contribution of mesenchymal stem cells to tumor development in tumor microenvironment. Cell Biosci 3: 5, 2013. 407. Yeung TL, Leung CS, Wong KK, Samimi G, Thompson MS, Liu J, Zaid TM, Ghosh S, Birrer MJ, Mok SC. TGF-beta modulates ovarian cancer invasion by upregulating CAF-derived versican in the tumor microenvironment. Cancer Res 73: 5016 –5028, 2013. 408. Yoshioka N, Fuji S, Shimakage M, Kodama K, Hakura A, Yutsudo M, Inoue H, Nojima H. Suppression of anchorage-independent growth of human cancer cell lines by the TRIF52/Periostin/OSF-2 gene. Exp Cell Res 279: 91–99, 2002. 409. Yu FS, Yin J, Xu K, Huang J. Growth factors and corneal epithelial wound healing. Brain Res Bull 81: 229 –235, 2010. 410. Yu H, Mouw JK, Weaver VM. Forcing form and function: biomechanical regulation of tumor evolution. Trends Cell Biol 21: 47–56, 2011. 411. Zaja-Milatovic S, Richmond A. CXC chemokines and their receptors: a case for a significant biological role in cutaneous wound healing. Histol Histopathol 23: 1399 –1407, 2008. 412. Zeisberg EM, Potenta S, Xie L, Zeisberg M, Kalluri R. Discovery of endothelial to mesenchymal transition as a source for carcinoma-associated fibroblasts. Cancer Res 67: 10123–10128, 2007. 413. Zeisberg EM, Potenta SE, Sugimoto H, Zeisberg M, Kalluri R. Fibroblasts in kidney fibrosis emerge via endothelial-to-mesenchymal transition. J Am Soc Nephrol 19: 2282–2287, 2008. 414. Zeremski M, Petrovic LM, Chiriboga L, Brown QB, Yee HT, Kinkhabwala M, Jacobson IM, Dimova R, Markatou M, Talal AH. Intrahepatic levels of CXCR3-associated chemokines correlate with liver inflammation and fibrosis in chronic hepatitis C. Hepatology 48: 1440 – 1450, 2008. 415. Zhu YH, Yang F, Zhang SS, Zeng TT, Xie X, Guan XY. High expression of biglycan is associated with poor prognosis in patients with esophageal squamous cell carcinoma. Int J Clin Exp Pathol 6: 2497– 2505, 2013. 416. Zigler M, Kamiya T, Brantley EC, Villares GJ, Bar-Eli M. PAR-1 and thrombin: the ties that bind the microenvironment to melanoma metastasis. Cancer Res 71: 6561–6566, 2011. 417. Zimmermann HW, Seidler S, Gassler N, Nattermann J, Luedde T, Trautwein C, Tacke F. Interleukin-8 is activated in patients with chronic liver diseases and associated with hepatic macrophage accumulation in human liver fibrosis. PLoS One 6: e21381, 2011. 418. Zoltan-Jones A, Huang L, Ghatak S, Toole BP. Elevated hyaluronan production induces mesenchymal and transformed properties in epithelial cells. J Biol Chem 278: 45801–45810, 2003.

Physiol Genomics • doi:10.1152/physiolgenomics.00158.2013 • www.physiolgenomics.org

Downloaded from on November 30, 2014

380. Wang W, Liu F, Chen N. Peroxisome proliferator-activated receptorgamma (PPAR-gamma) agonists attenuate the profibrotic response induced by TGF-beta1 in renal interstitial fibroblasts. Med Inf (Lond) 2007: 62641, 2007. 381. Webber J, Jenkins RH, Meran S, Phillips A, Steadman R. Modulation of TGFbeta1-dependent myofibroblast differentiation by hyaluronan. Am J Pathol 175: 148 –160, 2009. 382. Ween MP, Oehler MK, Ricciardelli C. Role of versican, hyaluronan and CD44 in ovarian cancer metastasis. Int J Mol Sci 12: 1009 –1029, 2011. 383. Wells RG, Discher DE. Matrix elasticity, cytoskeletal tension, and TGF-beta: the insoluble and soluble meet. Sci Signal 1: pe13, 2008. 384. White ES, Muro AF. Fibronectin splice variants: understanding their multiple roles in health and disease using engineered mouse models. IUBMB Life 63: 538 –546, 2011. 385. Wolf K, Mazo I, Leung H, Engelke K, von Andrian UH, Deryugina EI, Strongin AY, Brocker EB, Friedl P. Compensation mechanism in tumor cell migration: mesenchymal-amoeboid transition after blocking of pericellular proteolysis. J Cell Biol 160: 267–277, 2003. 386. Wong GS, Lee JS, Park YY, Klein-Szanto AJ, Waldron TJ, Cukierman E, Herlyn M, Gimotty P, Nakagawa H, Rustgi A K. Periostin cooperates with mutant p53 to mediate invasion through the induction of STAT1 signaling in the esophageal tumor microenvironment. Oncogenesis 5: e59, 2013. 387. Wu J, Wu X, Liang W, Chen C, Zheng L, An H. Clinicopathological and prognostic significance of chemokine receptor CXCR4 overexpression in patients with esophageal cancer: a meta-analysis. Tumour Biol [Epub ahead of print]. 388. Wu Q, Hou X, Xia J, Qian X, Miele L, Sarkar FH, Wang Z. Emerging roles of PDGF-D in EMT progression during tumorigenesis. Cancer Treat Rev 39: 640 –646, 2013. 389. Wynn TA. Cellular and molecular mechanisms of fibrosis. J Pathol 214: 199 –210, 2008. 390. Wynn TA, Ramalingam TR. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat Med 18: 1028 –1040, 2012. 391. Xiang L, Xie G, Ou J, Wei X, Pan F, Liang H. The extra domain A of fibronectin increases VEGF-C expression in colorectal carcinoma involving the PI3K/AKT signaling pathway. PLoS One 7: e35378, 2012. 392. Xiao Q, Ge G. Lysyl oxidase, extracellular matrix remodeling and cancer metastasis. Cancer Microenvir 5: 261–273, 2012. 393. Xu CZ, Wang PH, Yan XJ, Wang T, Chen D, Zhang ZJ, Shi RJ. Expression of CXCR4 is associated with progression and invasion in patients with nasal-surface basal cell carcinoma. ORL J Otorhinolaryngol Relat Spec 75: 332–341, 2014. 394. Xu S, Cao X. Interleukin-17 and its expanding biological functions. Cell Mol Immunol 7: 164 –174, 2010. 395. Xu TP, Shen H, Liu LX, Shu YQ. The impact of chemokine receptor CXCR4 on breast cancer prognosis: a meta-analysis. Cancer Epidemiol 37: 725–731, 2013. 396. Xu X, Zhou Y, Xie C, Wei SM, Gan H, He S, Wang F, Xu L, Lu J, Dai W, He L, Chen P, Wang X, Guo C. Genome-wide screening reveals an EMT molecular network mediated by Sonic hedgehog-Gli1 signaling in pancreatic cancer cells. PLoS One 7: e43119, 2012. 397. Xu X, Zhu F, Zhang M, Zeng D, Luo D, Liu G, Cui W, Wang S, Guo W, Xing W, Liang H, Li L, Fu X, Jiang J, Huang H. Stromal cell-derived factor-1 enhances wound healing through recruiting bone marrow-derived mesenchymal stem cells to the wound area and promoting neovascularization. Cells Tiss Organs 103–113, 2012. 398. Xue W, Zender L, Miething C, Dickins RA, Hernando E, Krizhanovsky V, Cordon-Cardo C, Lowe SW. Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature 445: 656 –660, 2007. 399. Yamada KM, Cukierman E. Modeling tissue morphogenesis and cancer in 3D. Cell 130: 601–610, 2007.

The wound healing, chronic fibrosis, and cancer progression triad.

For decades tumors have been recognized as "wounds that do not heal." Besides the commonalities that tumors and wounded tissues share, the process of ...
263KB Sizes 2 Downloads 0 Views