Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

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 de-
cades 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.
wound healing; regeneration; fibrosis; cancer; tumor stroma; myofibroblasts; tu-
mor- or cancer-associated fibroblasts; desmoplasia; epithelial-to-mesenchymal
transition

THE SHEER COMPLEXITY OF THE tumor stroma constitutes a re- also throughout development, inflammation, and fibrosis, as
markable discerning challenge for investigators. However, the well as during tumor progression. In EMT, epithelial cells
tumor niche resembles a site of chronic wound healing (16, 84, acquire a mesenchymal migratory phenotype, switching the
317). Out of the many similarities between wound healing and expression of known cell-cell adhesion molecules like E-
tumor progression, the mutual presence of myofibroblastic cadherin in favor of mesenchymal proteins such as N-cadherin
cells has emerged as a particularly common hallmark (56). and vimentin (362). In cancer, EMT facilitates metastasis by
Myofibroblasts are contractile spindle-shaped and stress fiber- promoting ECM proteolysis and enhancing malignant cell
forming mesenchymal cells that are known to express alpha motility, thus conferring an ability to invade (362). EMT also
smooth muscle actin (␣-SMA), contain excessive structural renders cancer cells a stem-like character, which is believed to
rough endoplasmic reticulum, and produce copious amounts of be responsible for therapeutic evasion, as well as a distinctive
extracellular matrix (ECM) (56, 90, 183). Myofibroblasts are prolonged latency/survival mechanism known as dormancy
also seen during fetal development (183) and are transiently (38). Myofibroblastic differentiation, also known as fibroblas-
present at sites of acute wound healing to repair injured ECM tic activation, describes the process by which the mesenchy-
(i.e., to renew and/or to crosslink ECM fibers) and facilitate mal/fibroblastic components of the stroma (i.e., resident fibro-
wound closure. Nonetheless, their constant presence in tumoral- blasts, recruited bone marrow-derived mesenchymal cells,
activated stroma (i.e., desmoplasia) is known to encourage pericytes, endothelial cells, and others) acquire a myofibroblas-
neoplastic progression (16, 56). Interestingly, similar persistent tic phenotype (90). EMT and myofibroblast differentiation are
myofibroblastic populations are also common occurrences dur- regulated by similar factors and signaling pathways (Supple-
ing chronic tissue fibrosis (56, 317, 390). Hence, we propose to mental Table S1).1 Moreover, some investigators believe that
review the triad “wound healing/chronic fibrosis/cancer” stromal myofibroblasts arise also from epithelial cells via
(WHFC) through the lens of wound healing while highlighting EMT-like processes (56, 123). Importantly, the healing process
their numerous common and few dissimilar traits. during acute injury involves four components: coagulation,
The wound healing response to injury is known to trigger inflammation, cellular proliferation, and ECM repair (212).
functional (i.e., proliferative) and phenotypic changes in fibro- Hence, wound healing (or regeneration) could be regarded as a
blasts, lymphocytes, and epithelial and endothelial cells (212). four-component process that constitutes the foundation for cell
For example, the changes in epithelial and stromal cells re- proliferation, EMT, and myofibroblast differentiation in the
spectively correspond to the epithelial-mesenchymal transition WHFC triad. Similarly to how Champollion deciphered hiero-
(EMT) and myofibroblastic differentiation processes (212). glyphs by understanding Greek and Coptic, wound healing
Both EMT and myofibroblastic differentiation are common constitutes “the Rosetta Stone” needed for a better understand-
occurrences during wound healing (or tissue regeneration) but ing of the WHFC triad (Fig. 1).
Chronic fibrosis and tumorigenesis can both result from a
* B. Rybinski and J. Franco-Barraza contributed equally to this work. prolonged and exacerbated healing response that could stem
Address for reprint requests and other correspondence: E. Cukierman,
Cancer Biology, Fox Chase cancer Center, 333 Cottman Ave. (W428), Phil-
1
adelphia, PA 19111 (e-mail: Edna.Cukierman@FCCC.edu). The online version of this article contains supplemental material.

1094-8341/14 Copyright © 2014 the American Physiological Society 223


Downloaded from journals.physiology.org/journal/physiolgenomics (180.242.184.036) on February 16, 2024.
Review
224 THE WHFC TRIAD

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.

from chronic injury (195, 317, 390), and as such the similari- fibrotic myofibroblasts and TAFs both remodel the ECM fiber
ties that tumors and their stroma have in common with chronic distribution by physically contracting and pulling on ECM
fibrosis are intriguing. In fact, it is well known that chronic proteins after engaging particular sets of integrins (140). Even
fibrosis, such as in liver cirrhosis induced by persistent viral though it remains unclear whether TAFs and fibrotic myofi-
infection (i.e., hepatitis), sustained alcohol consumption or broblasts possess unique traits that differentiate them, similar-
toxin-containing food, constitutes a direct cancer predisposi- ities, such as their contractile activity and the common expres-
tion (86, 91, 117). The epithelia from both fibrotic and tumor sion of ␣-SMA and others, suggest numerous phenotypic
tissues are characterized by the presence of hyperproliferating commonalities (56). Hence, it is only logical to assume that
cells (219, 269, 288); however, carcinoma cells carry transfor- effective targeting drugs that interfere with one (i.e., chronic
mative mutations (as well as epigenetic changes) presumed to fibrosis) will also stall the other (i.e., desmoplasia). Supple-
be absent in the fibrotic tissue. Also intriguing is the stromal mental Table S2 includes a list of drugs targeting both diseases.
element of both conditions, a dense myofibroblastic component While chronic fibrosis predisposes the affected tissues to
with an excessive deposition of ECM (56). The large popula- develop cancer (9, 30, 81, 91, 117, 232), a desmoplastic
tion of myofibroblasts at the tumor stroma is referred to as reaction that correlates with poor prognosis (56, 125, 317) is
cancer- or carcinoma-associated fibroblasts (i.e., known as also believed to enhance tumor progression. In turn, trans-
CAFs) and also, in this review, as tumor-associated fibroblasts formed tumor cells also promote TAF activation, thus support-
(TAFs). TAFs and fibrotic myofibroblasts drive tumor progres- ing the desmoplastic reaction (16, 20, 56). Describing the
sion and chronic fibrosis, respectively, through both paracrine relationship between cancer and fibrosis in these terms implies
effects on surrounding cells and exacerbated synthesis of ECM that fibrosis could be sparking tumorigenesis and tumor pro-
(16, 56, 286, 389). TAFs express a plethora of protumorigenic gression might be instigating chronic fibrosis. We suggest that
growth factors, inflammatory cytokines, proteolytic enzymes, the relationship between cancer and fibrosis (i.e., the WHFC
and ECM proteins such as collagen (Col)-I and -III as well as triad) constitutes both correlation and causation (Fig. 1). There-
a fibronectin spliced variant known as ED-A and more (56). fore, here we review the mechanisms by which the prolonged
TAFs are also known to contribute to the degradation of four component process of wound healing facilitates chronic
Col-IV, a protein associated with the epithelial ECM known as fibrosis and tumorigenesis through a common mechanistic link:
basement membrane (158, 235). Alterations to mesenchymal the WHFC triad.
ECM deposition, together with the hyperproliferation of the
stromal compartment in the vicinity of neoplastic lesions, are THE COAGULATION CASCADE FUELS THE WHFC TRIAD
known as “desmoplasia” or “desmoplastic reaction.” As such,
the desmoplastic reaction bears a striking resemblance to the Wound healing begins with the process of coagulation where
modified ECM typical of chronic fibrosis, which is also char- platelets from circulating blood accumulate at the damaged site
acterized by increased Col-I and -III deposition (390). Finally, and, together with increased levels of coagulation factors as

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


Downloaded from journals.physiology.org/journal/physiolgenomics (180.242.184.036) on February 16, 2024.
Review
THE WHFC TRIAD 225
well as with fibrinogen deposition and cleavage, result in the healing is eventually broken down by plasmin, a cleaved
formation of insoluble fibrin strands building up a clot (212). subproduct of plasminogen (111). Plasmin also plays a role in
The clot prevents blood loss and constitutes a short lived the ECM context due to its proteolytic activity in Col degra-
scaffold or provisional ECM that serves both for cell migra- dation, transforming growth factor beta (TGF-␤) activation
tion/invasion and as a growth factor reservoir (212). (see below) and release of other ECM-bound growth factors
In chronic fibrosis as in cancer, a process resembling clotting (111). Urokinase-type plasminogen activator (uPA) and tissue-
fuels disease progression. For example, fibrin bundles can type plasminogen activator (tPA) can each cleave plasminogen
accumulate at the tumor stroma as a result of a leaky tumor to obtain plasmin, and both enzymes are inactivated by plas-
vasculature (260), and similar fibrin deposition has also been minogen activator inhibitor-1 (PAI-1)(111). Interestingly, uPA
reported in a variety of fibrotic tissues (151, 308). Although the has been reported to inhibit fibrosis (216, 334, 335) but also to
role of fibrin in chronic fibrosis remains rather unclear, it is known promote tumor progression(335). Similarly, the role of tPA is
that fibrinogen exerts a mitogenic effect on fibroblasts (337). also unclear as it has been reported both as promoter as well as
During tumor progression, fibrin facilitates angiogenesis (93); inhibitor of chronic fibrosis and cancer (62, 144, 196, 303,
meanwhile, fibrinogen also promotes angiogenesis as well as 405). Finally, PAI-1 has also been shown to either promote or
oncogenic proliferation and antitumorigenic immune suppres- inhibit these two diseases (111, 155). The above-mentioned
sion (135). Moreover, elevated plasma fibrinogen correlates facts together provide rationale to suggest that the coagulation
with a poor cancer prognosis (110, 203, 225, 290, 296, 321, cascade constitutes a shared WHFC triad targetable pathway.
356). Intriguingly, fibrinogen may induce a prolonged inflam-
matory state in several types of cancer and fibrosis (68), CHRONIC INFLAMMATION IS A DRIVER OF THE WHFC
suggesting a possible link between chronic inflammation and TRIAD
persistent activity of the coagulation cascade.
Although overly simplified, it is generally agreed that, dur-
The cleavage of fibrinogen is considered the end result of the
ing homeostatic disturbances (i.e., during wound healing), M1
coagulation cascade signaling pathway (160). Thrombin di-
type macrophages drive an inflammatory response known to be
rectly cleaves fibrinogen into fibrin (95). Besides its role in the
tumor suppressive and antifibrotic, while M2 type macro-
coagulation cascade, thrombin promotes cancer progression
phages promote profibrotic and protumorigenic effects (96,
and fibrosis (47, 95). In cancer, thrombin induces cell prolif-
390). In response to macrophages and to injured epithelial
eration, tumor growth, angiogenesis, and metastasis (95). The
cells, naïve CD4⫹ T-cells can differentiate into largely antifi-
ability of thrombin to act as a mitogen is not limited to an
brotic and antitumorigenic Th1 cells or profibrotic and protu-
epithelial context. It also has been reported to enhance fibro-
morigenic Th2, Th17, and Treg cells, while CD8⫹ T-cells are
blast proliferation in a model of bleomycin-induced pulmonary
considered cytotoxic and constitute a positive neoplastic prog-
fibrosis (358). Furthermore, thrombin inhibition reduces the
nostic marker (390). During the acute wound healing response,
severity of both pulmonary (358) and liver fibrosis (83).
immunity is triggered to prevent infection and to induce quick
The ability of thrombin to promote cancer and fibrosis is
healing. Hence, a plethora of acute inflammatory secreted
believed to be mediated in part through its ability to activate
cytokines induce cellular proliferation, EMT, and myofibro-
the protease-activated receptor (PAR)-1 (47, 95). PAR-1 is
blastic differentiation (56, 390).
present in a variety of tumor types (89), and its activation
Importantly, even though EMT has been proved to be
promotes cancer cell proliferation and angiogenesis (95). In the
difficult to detect in many cancers, chronic inflammation asso-
clinic, PAR-1 signaling is suspected to facilitate melanoma
ciated with fibrosis (389) and tumor progression (103) could
metastasis (416), and it correlates with poor prognosis of
induce persistent EMT as well as myofibroblast differentiation,
gastric, prostate, and breast cancers (28, 100, 313). PAR-1 also
both of which are known to further fuel the WHFC triad. It has
facilitates pulmonary fibrosis (143), while PAR-1-deficient
been shown that inflammatory responses promote tumor de-
mice seem to resist bleomycin-induced pulmonary fibrosis
velopment and progression including metastasis, and this axis
(142). PAR-1 is one of four PAR isoforms (PAR-1 to -4), and,
has been thoroughly reviewed (120). In the following sections
while thrombin induces many of its effects via PAR-1, PAR-3
as well as in Supplemental Table S1, we highlight several
and -4 are also suitable to be activated by thrombin (47).
inflammatory factors that influence chronic fibrosis and tumor
However, PAR-2 can be activated by other molecules from the
progression because of their effects on cellular plasticity re-
coagulation cascade such as the tissue factor (TF) and activated
lated to the WHFC triad. Please note that many of these factors
factor X (FXA) (47). TF is expressed by many types of cancer
have also been reviewed elsewhere for their effects in regulat-
cells (169), and TF or FXA-induced PAR-2 signaling has been
ing the plasticity of mesenchymal stem cells (343, 397, 406).
implicated in both chronic fibrosis and tumor progression (33,
34, 151, 169, 318, 347). Interestingly, TF, FXA, thrombin, and TGF-␤
activation of PAR-1 or PAR-2 may possibly promote myofi-
broblastic differentiation (Supplemental Table S1). This type The TGF-␤ superfamily of cytokines plays a notorious role
of common occurrence opens up the possibility that other in the WHFC triad. TGF-␤ exerts a dual effect during cancer
enzymes from the coagulation cascade could also induce/ progression, modulating the behavior of both epithelial and
sustain myofibroblastic populations during chronic fibrosis and stromal cells imparting both pro- and antitumorigenic effects
tumor desmoplasia. (238). For example, it has been described that TGF-␤ plays an
To this end, the excess of fibrin deposition observed in antitumoral role on preneoplastic liver lesions by inhibiting
chronic fibrosis and desmoplastic stroma indicates a possible epithelial cells’ growth and driving proapoptotic signaling
enhancement in fibrin synthesis but also a likely decreased rate when induced by other cytokines such as interferon-␣ (70). On
of its degradation. The fibrin/platelet clot formed during wound the other hand, during more advanced tumorigenic stages,

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


Downloaded from journals.physiology.org/journal/physiolgenomics (180.242.184.036) on February 16, 2024.
Review
226 THE WHFC TRIAD

TGF-␤ has also been associated with increased invasion and role for IL-6-induced myofibroblast differentiation in fibrosis.
metastasis of malignant cells, correlating with enhanced activ- In addition, IL-6 induces epithelial cancer cell proliferation
ity and over expression of several integrins (238) as well as (315) while also activating the EMT program (145, 348) and
triggering EMT (242). Regarding its effects on stromal cells, contributing to a preference for bone metastasis (8, 327). It is
TGF-␤ can also induce both myofibroblastic differentiation well documented that IL-6 promotes colitis-associated cancer
(238) and altered ECM production (254). Of note and docu- via a STAT3-dependent mechanism (119). Finally, myofibro-
mented in several models of renal fibrosis (413), TGF-␤ can, in blasts from a variety of tissues are known to produce IL-6 (46,
addition, induce endothelial to mesenchymal transduction (16, 330, 361), suggesting that mesenchymal IL-6 may participate
412). Although it acts through different cells and mechanisms in the WHFC triad by facilitating tissue fibrosis, tumoral
and clearly participates as a tumor suppressor at early tumor- desmoplasia, epithelial growth, and metastasis.
igenic stages, it is nevertheless clear that TGF-␤ also promotes
fibrosis (238) as well as tumor progression and metastasis IL-17
(242). Certainly more information is needed in this regard as
IL-17 is a cytokine that is upregulated in response to Th17
knockout of the TGF-␤ receptor II was shown to have tumor-
effector cytokine IL-23 (121, 379). IL-17 upregulation has
promoting consequences (271), while collective cell migration been associated, together with other cytokines such as IL-6 and
was shown to improve under TGF-␤ signaling depletion (243). IL-22, as proinflammatory (394) as well as protumorigenic
Nonetheless, TGF-␤ is evidently the most dominant profibro- (379). IL-17 is known to act on myeloid and mesenchymal
genic factor known in all tissues, and anti-TGF-␤ agents (see cells to induce expression of IL-6 (and others) (394). In
Supplemental Table S2) are highly effective in a variety of addition, IL-17 is believed to induce expression of CXC
fibrosis models. Latent TGF-␤ is secreted by various cell types, chemokines known for their neurophil chemo-attractive capa-
such as lymphocytes, platelets, cancer cells, myofibroblasts, bilities (394). Interestingly, IL-17 has also been associated with
and others, during the WHFC triad. For example, latent TGF-␤ the expression of ECM remodeling enzymes such as matrix
can be secreted by cancer cells (242), inducing myofibroblastic metalloproteinases (MMPs) MMP1, MMP3, MMP9, and
differentiation of stromal cells, contributing to enrich an inva- MMP13, which are linked to both chronic inflammation and
sive TAF population (72, 211). Latent TGF-␤ is the inactive cancer (394). In particular, IL-17 has been shown to be acti-
form of TGF-␤ that is found linked to the ECM and is vated by local microbial factors that can particularly penetrate
sequestered by the latency-associated protein (LAP) and latent neoplastic but not normal tissues. For example, a deterioration
TGF-␤-binding protein 1 (5). Latent TGF-␤ can be activated of the colorectal cancer tumoral barrier is induced in early
by ECM damage and myofibroblastic engagement of LAP via lesions, allowing microbial factors to promote local IL-17-
cell-matrix receptors such as integrins ␣v␤1, ␣v␤3, ␣v␤5, dependent inflammation to further drive tumor growth (121).
␣v␤6, ␣v␤8, and others (257). TGF-␤ can be released and Interestingly, IL-17 has an ability to activate fibroblasts in
activated via ECM contraction induced by myofibroblastic autoimmunity, liver fibrosis, and cancer (246). Together these
cells (5). In fact, it has been suggested that changes in matrix facts emphasize an important role for IL-17 in the WHFC triad.
stiffness could regulate the equilibrium between storage and
release of matrix-bound factors such as TGF-␤ (383). In Chemokines
addition, the integrin-mediated binding of LAP may trigger a
conformational change in the molecule, releasing the bioactive For the success of a wound resolution, it is necessary to
form of this factor (5). It is noteworthy that myofibroblasts coordinate the recruitment of several types of cells (e.g.,
secrete and activate TGF-␤, establishing a self-sustained feed- inflammatory cells, macrophages, fibroblasts, and endothelial
back, contributing to expand their own population. In addition, and epithelial cells) to the insulted tissue. This recruitment is
TGF-␤ effects are known to induce an increase in Col-I-rich mediated by chemotactic cytokines (chemokines), which are
ECM production (254). Autocrine TGF-␤ signaling has been diffusible molecules that activate cell proliferation and direct
shown to drive myofibroblast differentiation during tumor cell migration toward the wound (21). The cytokines and
progression (185), suggesting a similar operational process in chemokines involved in wound healing and their influence in
chronic fibrosis. Interestingly, the activation of TGF-␤ and tumor microenvironment (370) have been listed in several
subsequent myofibroblast differentiation can be triggered in publications (21, 191, 411). In this review, we highlight some
vitro by a constant interstitial fluidic flow (265), suggesting the chemokines that are especially relevant to the WHFC triad.
possibility of a physical effect of augmented tissue pressure, During wound healing, the CXC chemokine family facili-
enhancing and expanding both myofibroblast and TAFs popu- tates angiogenesis as well as lymphocyte recruitment (411).
lations at the vicinity of the WHFC triad. To this end, it is not CXC receptors (CXCRs), in combination with chemokine
surprising then to recognize that all the WHFC triad diseases ligands such as CXCL8 (184), play a key role during various
present changes in interstitial fluid flow. cellular key responses (76, 253, 411). Therefore, it is not
surprising that deregulation of the CXCRs/CXCL8 axis leads
Interleukin-6 to fibrotic pathologies affecting lung (237, 340), kidney (351),
and liver (417). Moreover, CXCR receptors have been pro-
An important acute phase mediator, interleukin-6 (IL-6), is posed as therapeutic targets in some fibrotic conditions such as
also a proinflammatory factor that has a prominent role in asthma (48, 175). In cancer, CXCRs/CXCL8 seem to be
stromal activation. IL-6 null mice exhibit delayed wound responsible for driving cancer cell proliferation, survival, and
healing related to a diminished myofibroblast differentiation metastasis (104). Stromal CXCL8 potentiates the metastatic
(105). Moreover, IL-6 null mice exhibit an attenuated lung phenotype of cancer cells (104). More recently, CXCR2 has
fibrosis phenotype in response to radiation (311), suggesting a been implicated in sustaining a protumor inflammatory envi-

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


Downloaded from journals.physiology.org/journal/physiolgenomics (180.242.184.036) on February 16, 2024.
Review
THE WHFC TRIAD 227
ronment in colon cancer not only by recruiting inflammatory moral components (298). Moreover, in breast cancer cells in
cells but also by recruiting myeloid-derived suppressor cells vitro, CXCR4 signaling has been found to induce endocrine
into the tumor milieu and therefore obstructing antitumor therapy resistance (300).
immune responses (170). The broad spectra of CXCRs/CXCL8 Several inflammatory stimuli are responsible for the expres-
effects on cancer progression remains unclear, and yet CXCL8 sion of particular sets of chemotactic receptors in cancer cells
expression has been proposed as a chemotherapy effectiveness (371), and more recently we have started to learn about the
marker (104). cooperation between diverse chemotactic receptors driving the
Additional chemotactic axes relevant to the WHFC triad, metastatic phenotype as in the case of induction of CXCR2
which present contentious roles in fibrosis, are CXCR3 and expression via CXCR4 activation in breast cancer (336). The
their relevant ligand chemokines such as CXCL10 (411). complexity in the chemokines receptor/ligand networks calls
Activity of CXCL10/CXCR3 promotes fibroblastic maturation, for a better understanding of the role of inflammatory signals
and it also inhibits endothelial cell proliferation and migration modulating the evolution of the WHFC triad to develop better
into the wound, a crucial step for wound tissue remodeling and therapeutic strategies.
maturation (21). Since CXCR3 activity has been associated
with myofibroblastic differentiation and ECM maturation, it is Cyclo-oxygenase-2
expected that CXCR3-associated chemokines will be impli-
The enzyme cyclo-oxygenase-2 (COX-2) converts arachi-
cated in fibrosis. Indeed, in liver, CXCR3 is upregulated and is
donic acid to prostaglandin endoperoxide H2 (PGE2), and its
believed to play a profibrotic role in chronic hepatitis C (414).
expression triggers an inflammatory response (6). Neverthe-
In fact, serological levels of CXCR3-associated chemokines
less, the function of COX-2 and PGE2 seems to be contradic-
have been proposed to help monitor the progression and
tory and perhaps cell type specific during chronic fibrotic
complications of chronic liver diseases (352). In contrast,
conditions. For example, PGE2 inhibits hepatic fibrosis (23) as
studies in chronic pancreatitis have proposed an anti-fibrotic
well as pulmonary fibrosis where it has been shown to limit
role for CXCR3 and its associated chemokine CXCL9 via
fibroblast proliferation and myofibroblast differentiation (37,
downregulation of Col-1-␣1 and TGF-␤ (329). Additionally in
187). Also, although COX-2 is expressed briefly in response to
the pancreas, the blockade of CXCL10/CXCR3 promotes a
tissue injury (131), the use of COX-2 and prostaglandin inhib-
fibrotic condition in renal disease by upregulation of TGF-␤
itors appears only to impart a modest effect over wound
(262). Disparities in the CXCR3 activity on different organs
healing (131, 244). Even though these types of inhibitors block
during a fibrotic response suggest a more complicated interplay
inflammation, they exacerbate injury since they prevent wound
of immune cells for a balanced regulation between pro- and
healing especially in the intestine (354). Conversely, COX-2
antifibrotic cytokines/chemokines, which needs more investi-
inhibition reduces renal fibrosis (129, 302), suggesting that
gation.
PGE2 may contribute to fibrosis in the kidney. Anti-inflamma-
CXCL10/CXCR3 plays important roles in cancer as well.
tory COX-2 inhibitory therapy in combination with chemopre-
For example, in murine models of breast cancer, hypoxia
vention and as adjuvant to chemotherapy shows some thera-
inducible factor (HIF)-1␣ mediates the expression of CXCL10
peutic promise (178). The role of COX-2 has been particularly
and CXCR3 in mesenchymal stem cells and breast cancer cells,
well studied in colon cancer. However, COX-2 inhibition as a
respectively, promoting metastasis (50). Moreover, in mela-
therapy may require more investigation, as prolonged COX-2
noma murine models as well as in colon cancer patients,
inhibition promotes drug resistance, resulting in increased
CXCR3 promotes metastasis to lymph nodes (172, 173), and
intestinal fibrosis and tumor recurrence in mice (44, 69).
recent findings have demonstrated a synergism between
Intriguingly, the induced drug resistance is believed be medi-
CXCR3 and CXCR4 to promote metastasis to liver and lungs
ated by intestinal myofibroblasts, as chronic COX-2 inhibition
(258).
promotes TGF-␤-induced myofibroblastic differentiation and
Perhaps the best-known chemokine reported in the WHFC
augmented stromal PGE2 synthesis (44, 69). Moreover, TAFs
triad is CXCL12, also known as stromal-derived factor
from colon adenocarcinomas express a variety of factors that
(SDF)-1, which together with its main receptor CXCR4 in-
also drive tumor progression (188, 231). Hence, it is possible
duces migration of endothelial cells during wound-induced
that the persistent myofibroblastic population maintained by
angiogenesis (411). The chemotactic properties of CXCL12
chronic COX-2 inhibition is responsible for tumor recurrence
have also been documented during mobilization of fibrocytes
as well as its correlation with tumor progression (178).
from the bone marrow to the lungs (284), which in concert with
other chemokines contribute to aggravate pulmonary fibrosis Peroxisome Proliferator Activated Receptor Gamma
(230). Moreover, the SDF-1/CXCR4 axis has been shown to
trigger prostate myofibroblastic differentiation in vitro, sug- While the previously discussed proinflammatory molecules
gesting a role in fibrosis (109). seem to promote the WHFC triad, the role of the peroxisome
In cancer, the SDF-1/CXCR4 axis has been implicated in the proliferator activated receptor gamma (PPAR-␥), a transcrip-
progression of several types of malignancies, and its expres- tion factor involved in immunomodulation, is notable for its
sion is associated with poor prognosis (66, 132, 387, 393, 395). inhibitory effects over fibrosis and cancer. During the late
An important study almost a decade ago demonstrated that stages of wound healing, PPAR-␥ activation stalls fibroblast
SDF-1 secreted by TAFs (but not normal fibroblastic cells) proliferation and overthrows the myofibroblast phenotype
triggers both angiogenesis and tumor growth via CXCR4 (248), suggesting a mechanism for the termination of the
(275). SDF-1/CXCR4 activity is known to induce EMT and healing response. PPAR-␥ agonists are recognized for their
promote cell motility, invasion, and metastasis (299, 336), inhibitory effects on TGF-␤-induced myofibroblast differenti-
demonstrating a strong relationship between stromal and tu- ation (31, 41, 202, 239, 241, 252, 380). This antifibrotic effect

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


Downloaded from journals.physiology.org/journal/physiolgenomics (180.242.184.036) on February 16, 2024.
Review
228 THE WHFC TRIAD

has been proposed as a strategy to attenuate pulmonary and FGF-2


renal interstitial fibrosis (174, 194, 332). Then again, in cancer,
some investigators have proposed an oncogenic, while others FGF-2, also known as basic fibroblast growth factor, encom-
have stated a tumor-suppressive, role for PPAR-␥ (124). For passes a pivotal role during wound healing (220). FGF-2 exerts
example, PPAR-␥ agonists have been shown to impart anti- a robust mitogenic effect upon fibroblasts, and its participation
cancer effects (22), while a tumor-promoting effect is observed has been linked to multiple fibrotic disorders (26, 152, 162,
when PPAR-␥ is inhibited (236) (see Supplemental Table S2). 177, 344 –346). In addition, FGF-2 can be induced by TGF-␤
Nevertheless, PPAR-␥ signaling has also been shown to exert (see above), suggesting an important correlation between the
an inhibitory effect on TGF-␤-induced EMT in lung cancer two molecules (177, 345). Nonetheless, FGF-2 has also been
cells and subsequent metastasis (297). Even more, PPAR-␥ reported to inhibit TGF-␤-induced myofibroblast differentia-
ligands can also induce EMT upon intestinal epithelial cells tion (92, 150, 154, 234, 339). Interestingly, during nonpatho-
through a Rho-dependent activation of ERK1/2 (51), suggesting logical wound healing, FGF-2 inhibits excessive scarring by
a tight regulation of the PPAR-␥ signaling effects that could be triggering myofibroblastic apoptosis, yet quiescent (i.e., non-
associated with a particular cell type in a particular TGF-␤ activated) fibroblasts appear to be refractory to this molecule
signaling context. Interestingly, there is evidence to suggest an (1, 339). These observations suggest that, under physiological
intricate regulation of the tumor stroma in colon adenocarci- conditions, FGF-2 may play a key regulatory role during
nomas, where COX-2 and PPAR-␥ have been found coex- wound resolution, encouraging fibroblast proliferation to repair
pressed only in myofibroblasts at the tumor-associated but not damaged tissue but restricting the myofibroblastic population
the normal stroma (372). These findings suggest a PPAR-␥ to limit excessive scarring.
negative regulation of both fibrosis and cancer where its Nevertheless, FGF-2 effects have been implicated in the
presence at late stages of nonpathological wound healing development of several tumors by promoting proliferation
possibly contributes to prevent the WHFC triad occurrences. and survival of cancer cells and even supporting tumor
angiogenesis (368). It is not surprising that the effect of
CELL PROLIFERATION: A COMMON AVENUE USED BY THE FGF-2 and the FGF receptor family (FGFR) in cancer is
WHFC TRIAD considered “complex” as the pathway has been shown to
A hallmark of developmental growth but also of wound have both protumorigenic and tumor-suppressive roles
healing is a transient hyperproliferative wave affecting both (368). What is more, FGFR-induced activities can be ligand-
epithelial and stromal cells (183). This feature results from a independent (via genomic mutations of signaling molecules)
temporary burst of diverse mitogenic signals triggered by a or ligand dependent (368). Although the role of FGF-2 in
disturbance in the homeostasis of a given tissue (i.e., during desmoplasia remains rather obscure, its detection and im-
wound healing). Expectedly, a similar spectrum of mitogens plication at the stroma of prostate and mammary cancers as
can also be constitutively detected in fibrotic and tumor tissues well as other malignancies (223, 367, 403) suggests a fairly
(Fig. 1), promoting epithelial and mesenchymal cell prolifera- broad participation (267).
tion. Growth factors have a preponderate role among the large
variety of mitogenic signals at the WHFC triad (Supplemental Epidermal Growth Factor Receptor
Table S1). Here we emphasize some of them.
The epidermal growth factor receptor (EGFR) family, also
Platelet-derived Growth Factor known as the ErbB family, consists of pleiotropic membrane-
spanning cell surface receptors with intracellular tyrosine ki-
One of the most abundant molecules released in response to nase activities that are triggered by homo- or hetero-dimeriza-
tissue injury is the platelet-derived growth factor (PDGF) tion when engaged by growth factors. Some of the family
(227). PDGF is a key player in the persistence of chronic growth factor ligands are known to participate in wound
fibrosis, exerting a strong mitogenic signal upon fibroblasts and healing [i.e., epidermal growth factor (EGF), heparin-binding
myofibroblasts (32). Also, PDGF can stimulate the prolifera-
epidermal growth factor (HB-EGF), and TGF-␣] (281, 409).
tion of epithelial cancer cells in different types of tumors,
Under physiological conditions, EGFR signaling drives wound
exerting its influence through both paracrine and autocrine
healing, and controlled activity of EGFR by HB-EGF has been
mechanisms (133, 287). Hence, PDGF-induced cell prolifera-
tion could very well sustain a desmoplastic reaction while proposed as a therapeutic approach to prompt and accelerate
supporting tumor progression, thus fueling a protumorigenic cutaneous wounds healing (161, 363). Of note, aberrant EGFR
vicious cycle (20). Some studies suggest that PDGF could activation is known to promote fibrosis in tissues such as lung,
sustain or even initiate desmoplasia in breast cancer (148, 326), heart, and pancreas (128, 214, 229, 245). EGFR activity has
while others point to the fact that PDGF is a promigratory been implicated in the progression of malignancies such as
factor (67) as well as an EMT regulator (388). Furthermore, colon and lung cancer (94, 134, 314). Nevertheless, EGFR
PDGF expression can also affect other growth factors, as activity has also been implicated in tissue regeneration during
inhibition of stromal PDGF receptors in a murine model of acute and chronic colitis where EGFR activity seems to limit
cervical cancer was shown to alter the levels of expression of fibrosis induced tumorigenesis (80). Then again, EGFR signal-
the fibroblast growth factors (FGF)-2 and -7 (287). Moreover, ing activates EMT in malignant cells (130, 312). Doubly,
the inhibition of FGF-2 and FGF-7 in cervical cancer restricts EGFR activity affects the tumor stroma by inducing HB-EGF
their respective mitogenic and angiogenic effects, suggesting overexpression in TAFs, thus supporting tumor progression by
PDGFR as an attractive therapeutic target (287). direct and indirect effects (259).

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


Downloaded from journals.physiology.org/journal/physiolgenomics (180.242.184.036) on February 16, 2024.
Review
THE WHFC TRIAD 229
Additional Deregulated Developmental Signals Promoting ergistic interaction could constitute an interesting therapeutic
WHFC approach.
The healing of a wounded tissue is known to be influenced PATHOLOGICALLY ALTERED ECMS CONSTITUTE BOTH
by signaling molecules involved in embryonic development CAUSE AND CONSEQUENCE OF CHRONIC FIBROSIS AND
(183). Some of these signaling molecules are highly relevant TUMOR PROGRESSION
during wound resolution and, when deregulated, are major
players in the WHFC triad. To this end, Notch and Hedgehog Below we discuss the participation of physical ECM (i.e.,
(Hh) signaling cascades constitute two well-known examples architecture or stiffness) properties, as well as noteworthy
of developmental signaling implicated in these processes (see components of pathological ECM common to the WHFC triad.
Supplemental Table S1). Topographical/architectural Changes in ECM
Notch signaling is an evolutionary well-conserved pathway
known for regulating embryonic morphogenesis, tissue homeo- A pathological mesenchymal ECM differs from its normal
stasis, angiogenesis, and maintenance of the vascular system counterpart and is recognized by an altered and constitutive
(25). The role of Notch during wound healing has been dem- expression of its building molecules, as well as for their
onstrated in vitro as well as by genetic manipulation of murine characteristic architectural organization (294, 295), which to-
models where Notch-deficient animals present delayed wound gether are believed to impart distinct physical and biochemical
healing (54, 77, 228). Regarding its mechanisms of action, it is properties to the ECM (97, 410). Moreover, the altered archi-
known that Notch signaling drives EMT, providing a vast tectural composition of a pathological ECM has been associ-
source of fibroblastic cells necessary for wound repair (171). In ated with clinical outcomes in breast cancer (59). Functionally,
addition, Notch activity has also been associated with fibrosis it has been suggested that topographical ECM alterations can
promotion in a variety of tissues (such as skin, kidney, lung, impact normal stromal cells’ behaviors, thereby supporting a
and cardiac tissue), influencing cell proliferation, extensive phenotypic switch (i.e., myofibroblastic activation) (4, 139).
fibrilogenesis, TGF-␤ production, and ␣-SMA expression Concurrently, these newly activated cells continue synthesiz-
(171). In cancer (but not in regenerating epithelium during ing the above-mentioned altered ECM, feeding a vicious cycle
wound healing), Notch has also been shown to have an onco- affecting additional adjacent stromal (4, 16, 115, 137), as well
genic role, promoting protein biosynthesis, cell growth, apo- as epithelial cells (16, 45, 97, 116, 197, 201), in a continuously
ptosis resistance, and angiogenesis (305). Moreover, it has also expanding “field effect.”
been implicated in triggering EMT (224), fueling cancer inva- ECM Stiffness
sion and enriching the desmoplastic TAF population. Never-
theless, there is evidence to suggest that Notch, depending on Both fibrotic and tumor-associated (i.e., desmoplastic)
the cellular context, can exert a tumor suppressive role (224). ECMs are characterized by an increase in stiffness compared
Another important signaling pathway in the WHFC triad is with their normal stromal equivalent, which is known to be
the Hh cascade. The Hh signaling pathway is triggered by more flexible or compliant (116, 208, 277, 364). A less flexible
several types of Hh ligands (i.e., Sonic, Desert, and Indian). environment exerts a greater physical tension to the cell body,
The ligands bind to the receptor Patched, which in turn releases which is transduced into multiple intracellular signals, modi-
Smoothened to promote Gli transcription factors (Gli-1, Gli-2, fying cell behavior. For example, relatively stiff ECMs alter
and Gli-3) (58). Gli-1 appears to be the most abundant Hh- cell growth, modify gene expression, and alter the differenti-
activated transcription factor, while Sonic Hh (SHh) is the ated status of cells (292, 338, 378). Moreover, ECMs subjected
better characterized ligand in the signaling pathway (58). SHh to a higher physical tension induce structural modifications to
signaling is essential for wound healing as demonstrated by the ECM-associated molecules, modulating their bioactivity (i.e.,
inhibitory effect imparted by the topical application of a releasing active molecules of TGF-␤), which in turn, for
cyclopamine, a well-characterized inhibitor of smoothened, at instance, can trigger myofibroblastic differentiation (139) and
all stages of the wound healing process (200). However, SHh also further fuel the production of the altered (166) and stiffer
signaling has also been shown to promote fibrosis in several ECM. Hence, in chronic fibrosis, tissue stiffness fuels a posi-
organs such as lung, liver, and skin (157). Likewise, Hh has tive feedback enhancing the chronicity of fibrotic disease
been shown to participate in tumor progression (58) by pro- (218). Meanwhile in cancer, a rigid ECM induces tumor
moting stromal activation/desmoplasia (149, 359). For exam- progression (42, 208, 282, 410) but also an enrichment of
ple, in pancreatic as well as in prostate cancer, it has been activated/myofibroblastic cells (i.e., TAFs), which in turn sus-
found that SHh is released by cancer cells, affecting stromal tain a desmoplastic reaction and further promote tumorigenesis
cells, causing differentiation and proliferation of ␣-SMA-pos- (16, 20, 64, 116, 399). ECM rigidity regulates the effects of
itive myofibroblastic TAFs (15, 149, 199, 328, 376). Besides TGF-␤ upon tumor cells, as compliant matrices facilitate
its ability to activate the tumor stroma, SHh ligands also TGF-␤-induced apoptosis, while stiffer matrices promote
promote EMT in cancer cells (396). Interestingly, while SHh TGF-␤-induced EMT (206). Various ECM-related modifiers
signaling facilitates tumor progression through multiple mech- and a plethora of molecular components participate in the
anisms, it is possible that SHh ligands are not the only possible WHFC triad, and a few are listed below as well as in Supple-
triggers of SHh signaling, as it has been found that TGF-␤ can mental Table S1.
also induce the activity of Gli-1 (157). This signaling pathway Fibronectin
overlap strongly suggests a common participation in the
WHFC triad. Although the relationship between SHh signaling A native well-conserved constituent of the mesenchymal
and TGF-␤ activation merits additional investigation, the syn- ECM that plays a determinant role in the WHFC triad is

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


Downloaded from journals.physiology.org/journal/physiolgenomics (180.242.184.036) on February 16, 2024.
Review
230 THE WHFC TRIAD

fibronectin (FN). Plasma FN (307) is commonly found in the tic reaction (12). For example, it was reported that pancreatic
circulation, while cellular FN (400) is the glycoprotein that is cancer cells can trigger fibroblastic activation while promoting
expressed by fibroblastic cells and is incorporated into the increased deposition of Col-I and -III in a nude murine model
fibrillar ECM mesh. From a single human gene and as product (12). In breast cancer, Col-I cross-linking is increased, showing
of alternative mRNA splicing, several cellular FN variants can a reduction on its degradation rate, resulting in excessive
be expressed (384). Two spliced exons called “extra domains deposition (168). In sum, Col-I and -III changes are typically
A” and “B” (EDA and EDB) are particularly and abundantly described in diseases associated with the WHFC triad.
expressed during embryonic developmental stages, while an
important (i.e., normal) reduction is observed in adult tissues Lysyl Oxidase
(384). Nevertheless, during the physiological ECM restructur-
An important regulator of ECM rigidity is the activity of the
ing of wounded tissue, EDA-FN is upregulated by myofibro-
enzyme lysyl oxidase (LOX), which catalyzes the cross-linking
blastic cells (118). Moreover, in a positive feedback regulation,
of collagen (see above) and elastin bundles, modifying the
EDA-FN has been shown to exert a permissive action during
tensile strength of the ECM (165). LOX is essential for main-
myofibroblastic activation (82). EDA-FN overexpression has
taining ECM strength during normal development (233) and
been found to occur in response to increased ECM tension becomes highly expressed during the course of several fibrotic
(364), and it is influenced by high levels of TGF-␤ at the diseases (164, 392). Throughout cancer progression, LOX
specific microenvironment of acute injuries (324). The rela- facilitates Col-I cross-linking, promoting ECM stiffening and
tionship of EDA-FN with TGF-␤ appears to be a determinant therefore altering known integrin-regulated signaling pathways
for the progression of several fibrotic diseases (i.e., atheroscle- (208). In fact, increased LOX activity has been associated with
rosis and lung and liver fibrosis) as demonstrated in various in a poor prognosis in several carcinomas (392). LOX and LOX-
vivo FN mutant animal models (384). Researchers believe that like enzyme 2 (LOXL2) are downstream targets of HIF-1␣
the sustained TGF-␤-promoted myofibroblastic differentiation and, under hypoxic conditions, promote EMT by repressing
occurs via production and signaling of EDA-FN, supporting a E-cadherin (319). In a xenograft tumor model, the inhibition of
chronic fibrotic niche (384). EDA-FN also influences the tumor LOX2 decreased the number of ␣-SMA-positive TAFs (17).
microenvironment, affecting both stromal and tumor cells. At Furthermore, LOXL2 has been proposed as a prognostic bio-
the stromal compartment, EDA-FN seems to be intimately marker for squamous cell carcinomas (283). Together these
related to the bioavailability of active TGF-␤ for the subse- data support the correlation between increased cross-linked
quent sustainability of the reactive stroma (384). EDA-FN is Col, stiffer ECM, an enriched TAF population, and the exac-
known for promoting tumor growth, EMT, and tumor-induced erbation of tumor desmoplastic reaction. Interestingly, the
lymphangiogenesis (349, 391). EDB-FN has also been associ- same LOX inhibitory antibody hinders murine liver and pul-
ated with tumor progression, in particular with tumor angio- monary fibrosis (17), suggesting a compelling similarity as
genesis and is being tested as a potential tumor target in the proposed in the WHFC triad (Supplemental Table S2).
clinic (320). Hence, alternative spliced forms of FN seem to
play important roles in the WHFC triad. MMPs

Collagen Even though, intuitively, MMPs, are ECM-cleaving protein-


ases, they do not antagonize fibrosis, cancer, or fibrosis-
As the major ECM component in the connective tissue and induced cancer. Hence, these enzymes are regarded as ECM
the most abundant protein in mammals, collagen has been remodeling proteins as opposed to ECM-degrading proteins
described as a heterotrimeric triple helix glycoprotein that can (278). MMPs play a crucial role during inflammation, re-
be assembled into de novo fibrous matrices during healing, epithelialization, and ECM remodeling associated with regen-
playing a crucial role during the repair of tissue defects. erative wound healing (113). Specifically, MMPs mediate
Collagen deposition increments the ECM tension during the ECM remodeling during scar resolution, cleave and activate
restoration of the vessels network and the recovery of tissue ECM-associated growth factors, regulate chemokine activity,
architecture during wound healing (118). Among the collagen and also facilitate keratinocyte migration into the wound (113,
family, Col-I is the most abundant interstitial protein of the 176, 278). MMPs can be expressed by inflammatory cells,
ECM (107) often seen in fibrotic tissue and desmoplasia. Then epithelial cells, fibroblasts, and myofibroblasts. The sources
again, Col-IV is the main component of epithelial and endo- and functions of MMPs implicated in the context of cancer
thelial basal membrane (167). The activated/myofibroblastic (176) and fibrosis (278) are very diverse and beyond the scope
phenotype seen in the WHFC triad is commonly described as of this review. Nonetheless, we highlight two gelatinases,
containing abundant Col-I and -III, although reports showing MMP-2 and MMP-9 (19, 176), as these enzymes seem to
changes in Col-IV and -V are also evident (136). In addition, convey a recurrent presence in the WHFC triad.
Col-VI spiked some attention due to its selective expression in The participation of MMP-2 and MMP-9 in chronic fibrosis
some fibrotic conditions (i.e., kidney and myocardial fibrosis) appears to be contentious. For example, a clear upregulation of
as well as in hepatocellular carcinoma (136). In fact, its level these MMPs expressed by myofibroblasts is evident during the
of expression has been recently proposed to serve as a clinical development of idiopathic pulmonary fibrosis (322). Also,
prognostic marker (204, 205). Carcinomas that typically pres- MMP-2 has been found to be highly expressed in fibrotic liver
ent an aggressive progression (i.e., liver and pancreas) are myofibroblasts (293), which incidentally has been proven to
characterized by a large deposition of interstitial FN and constitute a noteworthy predisposition to developing hepatic
collagen. These occurrences are blamed for augmented tumor cell carcinoma (86). Nonetheless, a downregulation of MMP-2
cell proliferation that is accompanied by an intense desmoplas- has been reported during in vitro myofibroblastic differentia-

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


Downloaded from journals.physiology.org/journal/physiolgenomics (180.242.184.036) on February 16, 2024.
Review
THE WHFC TRIAD 231
tion of rat embryonic fibroblasts (141), while its inhibition has and larynx (355). Interestingly, epithelial HA accumulation in
been shown to accelerate murine renal fibrosis (268). colon cancer constitutes a bad prognostic occurrence (304),
In cancer, MMP-2 and MMP-9 are known to promote tumor while stromal HA buildups have been associated with an
progression by facilitating the release of stored latent growth unfavorable prognosis in ovarian, prostate, breast, and nons-
and other factors (i.e., TGF-␤) from the ECM, thus enhancing mall cell lung cancers (11, 217, 289, 382). In the case of liver,
respectively cell proliferation and migration (36). MMPs can the main organ in charge of processing HA (98), a large
also facilitate cancer cell migration (385) by interaction with synthesis of the glycosaminoglycan and increasing circulating
cell-matrix adhesion molecules, such as integrins, promoting levels of HA have been found to correlate with liver fibrosis
degradation of cancer cells from cell-cell and cell-matrix ad- (cirrhosis) (280). In addition, hepatic cirrhosis is known to
hesions (36). To further fuel the recurring cycle, EMT en- predispose for hepatocarcinoma, and HA deposition has also
hances the expression of MMP-2 and MMP-9 (36). Interest- been associated with cancer progression (285, 341). Most of
ingly, MMP cleaved ECM peptides (i.e., soluble ECMs) are the mechanisms of HA’s driven tumor progression deal with
known to induce an integrin-dependent TGF-␤-like signaling the promotion of cell proliferation, NF-␬B activity, and angio-
response (106). Also, CD147, a cell-surface glycoprotein up- genesis (333). HA also binds to the cellular receptor CD44,
regulated in tumor cells, can stimulate both stromal MMP which encourages motility and invasion (333). Intriguingly,
production and myofibroblast differentiation (147). In sum, HA expression is increased via growth factors such as EGF,
MMPs produced by cancer cells promote tumoral invasion as keratinocyte growth factor, and PDGF (355), suggesting a
well as myofibroblast differentiation, reinforcing a tumor- more complicated role for certain growth factors, which could
supportive stroma, which in turn further secretes and activates promote both cancer cell proliferation and HA accumulation.
additional MMPs to further fuel this vicious cycle. Interestingly, the antiproliferative and proapoptotic effect of
HA inhibition, affecting both the tumor and surrounding
Hyaluronan stroma (285), highlights the importance of designing antican-
cer strategies focused on attacking both the tumor and its
A particular component of the ECM, with no inflammatory associated fibrotic stroma.
activity but related to inflammation, is the glycosaminoglycan
hyaluronan (HA). This macropolysaccharide molecule con- Periostin
tains multiple unit repetitions of N-acetyl-glucosamine as well
as of glucuronic acid, and it binds and stores large amounts of Periostin is a matricellular protein that increases ECM stiff-
water molecules conforming a viscous gel (355). HA has been ness by direct binding to fibrillar ECM proteins such as Col-I
suggested to function as a pliable substrate for tissue remod- (270) and by interacting with the TGF-␤-related protein
eling necessary for wound healing (355). It also binds fibrin- BMP-1, which is needed to activate LOX (240). In addition,
ogen, supporting clotting, and acts as a mesh that traps inflam- periostin functions as a ligand for integrins ␣v␤3, ␣v␤5, and
matory cells (342). Large HA molecules of ⬃25,000 disaccha- ␣6␤4 (102, 306). In general, periostin expression seems to play
ride units (355) encourage cellular quiescence, while small HA a main role during wound healing and tissue regeneration
fragments ranging from 4 to 25 units encourage proliferation (reviewed in Ref. 192). For example, periostin-deficient mice
and angiogenesis (342). It has been proposed that HA break- present a reduction in the number of cutaneous wound healing/
down at the injury site renders fragments that participate in the repair myofibroblasts (88). Periostin has been associated with
initiation of the healing response (342). myofibroblastic differentiation in vivo and in vitro via an
HA accumulation is associated with exacerbation of fibrosis integrin-dependent mechanism as well as with ECM remodel-
(27, 159). The expression of HA synthesizing enzymes “hya- ing (60, 87). As a consequence, periostin increases are also
luronan synthase (HAS) 1–3” during chronic fibrosis has been associated with pathologies marked by a fibrotic condition such
associated with an increased inflammatory reaction (52, 213). as idiopathic pulmonary fibrosis, renal fibrosis, asthma, and
Knockdown of HAS2 reduces the effectiveness of TGF-␤- skin sclerosis in scleroderma (261, 323, 331, 401).
stimulated EMT (291), and elevated HAS2 expression is suf- Regarding cancer, periostin has been found to be expressed
ficient to induce EMT in normal epithelial cells (418). More- by cancer epithelial cells (255, 306), and recently it has been
over, inhibition of HA synthesis antagonizes TGF-␤-induced associated with increased esophageal cancer invasion of p53
myofibroblastic differentiation (381), and inhibition of HAS2 mutants via STAT1 induction (386). Moreover, periostin can
impairs myofibroblast accumulation in mouse models of pul- also be expressed by stromal cells and, as such, has been
monary fibrosis (213). Since elevated HA has been observed in identified in desmoplastic pancreatic cancer (102), colon can-
a variety of fibrotic diseases (210, 213, 357) and HAS2 cer, cholangiocarcinoma (179, 369), and other types of cancer
regulates TGF-␤-dependent functions (291), their accumula- (193, 222, 249, 255, 306). Consequently, periostin seems to
tion and increase in expression are respectively suggestive (i.e., facilitate both tumoral and stromal progressions. Periostin
markers) of fibrotic myofibroblast differentiation and EMT. integrin-dependent signaling activates focal adhesion kinase
With regards to cancer, increased HA is linked to poor (FAK), as well as the PI3K/AKT pathway, thus generally
cancer outcomes (355), while upregulation of HAS2 is a promoting malignant cell survival, proliferation, and angiogen-
common occurrence in highly metastatic breast cancer cells esis (255, 306). Moreover, activation of integrins by periostin
(273). Nevertheless, the role of HA in cancer seems to be tissue promotes EMT in cancer cells (255). Intriguingly, in bladder
specific. In general, accumulation of HA in tissues where it is cancer, periostin seems to function as a tumor suppressor by in
normally absent facilitates tumor growth (i.e., gastric cancers), vitro inhibition of invasion and reduction of in vivo metastasis
while reduced HA is associated with cancer in normally HA- (180). Moreover, in many cancer cell lines in vitro periostin
rich stratified epithelia containing tissues such as skin, mouth, seems to suppress anchorage-independent growth, while low

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


Downloaded from journals.physiology.org/journal/physiolgenomics (180.242.184.036) on February 16, 2024.
Review
232 THE WHFC TRIAD

periostin mRNA levels are observed in lung cancer in vivo sis has demonstrated that not all the cellular responses to
(408). SPARC signaling depend on TGF-␤ (114), and, although there
is a certain parallelism between them promoting neoplasia
Tenascin C (242), a balance of both SPARC and TGF-␤ activity could be
responsible for tumor progression (114). These facts suggest
Tenascin C is another ECM-related glycoprotein expressed
that an intricate regulation of the SPARC/TGF-␤ signaling axis
in response to tissue injury known to modulate the wound
also constitutes a common occurrence of the WHFC triad
healing response (251). Tenascin C levels decrease subse-
(Supplemental Table S1 and Fig. 1).
quently 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 mis- Proteoglycans
regulated and continuous expression of tenascin C is a common Comprising a diverse group of members, the proteoglycan
occurrence, which is believed to drive fibrosis and cancer via family includes both cell surface proteins, such as heparan
increased angiogenesis, ECM alterations, and immunomodula- sulfate proteoglycans, and ECM components, such as small
tion (250). For example, tenascin C-deficient mice are pro- leucine-rich proteoglycans (SLRPs), and large chondroitin sul-
tected from pulmonary and liver fibrosis (43, 85). In addition, fate proteoglycans (153). Proteoglycans participate in immu-
tenascin C has been shown to promote tumorigenic progression nity regulation (57, 99), ECM assembly (53), and cancer
by supporting cell motility, metastasis, and more (101, 127, modulation (153). However, as a complete description of
207, 274, 279). Tenascin C is synthesized by both tumoral and proteoglycans is beyond the scope of this review, we highlight
stromal cells (71, 276). Then again, tenascin C could also a selection of ECM proteoglycans most relevant to the WHFC
indirectly promote the WHFC triad by its effect on the down- triad.
regulation of tPA, which facilitates fibrin accumulation, thus Versican, a large chondroitin sulfate proteoglycan, is up-
triggering a mitogenic response (40). regulated during both wound repair and cancer progression
(63, 112, 301, 353). During normal conditions, myofibroblasts
Osteonectin/SPARC
from human granulation tissue express elevated levels of ver-
The secreted protein and rich in cysteine (SPARC), also sican (126). There is a lack of studies investigating the role of
known as osteonectin, is a matricellular protein that is known versican in the context of chronic fibrosis. However, the
to affect ECM remodeling/dynamics. SPARC is often highly expression of versican mRNA has been detected as increased
expressed during wound healing, cancer, and fibrosis (18, 55, in rat lungs after bleomycin-induced injury, and TGF-␤ sig-
365). For example, the upregulated expression of SPARC naling increases versican synthesis in fibrotic lung in vivo
seems to be a common event during several chronic fibrotic (373). On the other hand, versican expression correlates with a
conditions such as in the cases of heart, lungs, kidneys, liver, poor prognosis in many cancer types (301, 325). This pro-
dermis, intestine, and eyes (365). SPARC is produced by teoglycan interacts with multiple ECM and cell surface com-
fibroblasts in response to TGF-␤, but it can also promote Col-I ponents, promoting cell proliferation, cell motility, and metas-
and TGF-␤ synthesis (365). In a SPARC null murine model, a tasis (301, 310, 382). Intriguingly, in breast cancer, versican
decreased deposition of dermal collagen constitutes a predom- has been associated with chemotherapy resistance related to
inant trait accompanied by an enhanced contractile behavior of EGFR hypersignaling and enhanced cancer cell self-renewal
dermal fibroblasts (39). By affecting collagen deposition and (78, 79). Moreover, versican synthesis appears to be upregu-
reducing the number of myofibroblasts, decreased SPARC lated by TAFs in the ovarian cancer stroma (407) where TGF-␤
expression reduces liver fibrosis (10). Nonetheless, despite the signaling triggers versican expression, and together they con-
implication of SPARC in the control of wound healing, its tribute to promote ovarian cancer aggressiveness (407).
precise role remains rather unclear. For instance, it has been The SLRP biglycan expression is enhanced in myofibro-
reported that SPARC can both induce and inhibit the wound blasts from human granulation tissue (126). The role of bigly-
healing process (18, 39). can in fibrosis may be tissue specific since biglycan does not
Similarly, SPARC has also been implicated in tumor pro- affect TGF-␤-induced pulmonary fibrosis (186), and biglycan
gression where it is believed to sometimes promote, and at mRNA appears not to be increased in mercuric chloride-
other times prevent, this process apparently in a tissue- and induced renal tubulointerstitial fibrosis (350). However, bigly-
cell-specific manner (55). Among other functions, SPARC can expression and serological levels of its processed byprod-
participates both in carcinoma EMT and in TAFs’ myofibro- ucts correlate with the extent of liver fibrosis in murine models
blastic differentiation, thus promoting both tumorigenesis and (108, 204). Biglycan is also involved with progression and
desmoplasia (55). In prostate cancer, for example, the expres- poor prognosis in a variety of malignancies such as gastroin-
sion of SPARC by malignant cells can be predictive of metas- testinal and endometrial cancers (7, 122, 221, 377, 415). In
tasis, while in pancreatic and nonsmall cell lung cancers, contrast, biglycan expression correlates with a favorable prog-
elevated expression of SPARC at the tumor stroma indicates nosis in bladder cancer where it seems to have an inhibitory
poor prognostics (29, 74, 189). Regarding SPARC’s tumor- effect on tumor cell proliferation (266).
suppressing effects in breast, ovarian, colon, and bladder car- In addition to biglycan, the SLRP decorin also plays an
cinomas, SPARC has been associated with inhibition of pro- active role in the WHFC triad, although its participation in
liferation and induction of apoptosis (55, 309). Intriguingly, a chronic fibrosis and tumor progression seems to have an
positive feedback between SPARC and TGF-␤ has been ob- inhibitory effect. Animal models have shown that absence of
served, as both can induce the expression of each other (365). decorin exacerbates fibrosis (13, 247, 316). Moreover, inter-
Nevertheless, a comprehensive comparative proteomic analy- esting data suggest that decorin itself could be a beneficial

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


Downloaded from journals.physiology.org/journal/physiolgenomics (180.242.184.036) on February 16, 2024.
Review
THE WHFC TRIAD 233
antifibrotic therapeutic alternative (156, 186, 402). A suggested further fueling the WHFC triad’s fibrosis/tumorigenesis vi-
antifibrotic mechanism of decorin deals, at least partially, with cious cycle (see Fig. 1).
the inactivation of TGF-␤ signaling (14), impairing myofibro- To further complicate things, although SASP seems to fuel
blastic differentiation in liver fibrosis models (14). Another the WHFC triad, evidence also suggests that the sole escape of
mechanism described is the direct interaction with the cytokine fibroblasts from senescence can also support WHFC. For
known as connective tissue growth factor or CTGF, inhibiting example, during normal wound healing, the matricellular pro-
its fibroblastic activation role (374). In cancer, decorin gene tein CCN1 (also known as cysteine-rich protein 61 or CYR61)
therapy or delivery of decorin protein inhibits tumor growth, is expressed by myofibroblasts and tends to accumulate along
tumor progression, and metastasis (153). Besides the attenua- with the myofibroblastic cell population (198). High concen-
tion of TGF-␤ signaling by decorin described in fibrosis trations of CCN1 induce myofibroblast senescence (198) and
models (14, 402), another mechanism of action has been found promote downregulation of collagen and TGF-␤ as well as
in the tumor microenvironment where it could also be inhib- upregulation of MMPs (198). Together these changes induce a
iting EGFR signaling (153). Decorin binds to EGFR overlap- senescent matrix-degrading cell state (198). During wound
ping with the EGF binding domain and inducing internalization healing resolution, myofibroblast senescence serves to prevent
of EGFR via caveolar endocytosis, resulting in decreasing pathological scarring (198). Hence, senescence is generally
EGFR accessibility on the cell surface (153). Together, these recognized to inhibit fibrosis (163, 190), so it is not surprising
effects evoke a downregulation of EGFR signaling in tumor then that mice expressing defective CCN1 suffer from exacer-
xenografts and apoptosis induction (153). Decorin has also bated fibrosis in response to injury (198). Furthermore, recent
shown additional antitumor effects including the inhibition of findings have shown that restoring lost levels of CNN1 in a
angiogenesis and induction of p21 (153, 264). Although re- hepatic fibrosis murine model can induce myofibroblastic se-
duced decorin expression in the tumor stroma indicates poor nescence and reduce chronic tissue-damaging fibrosis (182).
prognosis in models of breast cancer (264), decorin can be Nevertheless, the role that CCN1 plays in both tumor-associ-
ated stroma (i.e., desmoplasia) and in tumorigenesis remains
found in both the tumor stroma and fibrotic tissue (14, 153).
rather obscure. CCN1 has been shown to present opposing
This could be explained as a protective response (153), al-
effects on tumor progression, as some data indicates that CCN1
though it has also been observed that collagen-bound decorin
inhibits tumor growth by inducing apoptosis and senescence,
found in the ECM could not be effectively protective (14).
while other reports suggest it promotes tumor growth via the
Future work may seek to clarify the influence of decorin on the
stimulation of cancer cell proliferation and angiogenesis (198).
myofibroblastic cells during chronic fibrosis and tumor desmo- In addition, it is known that CCN1 induces senescence by
plasia. activating the p16(INK4A)/pRb pathway (198). Interestingly,
TAFs are hyperproliferative cells that express diminished lev-
CHRONIC FIBROSIS AND CANCER DIFFER FROM els of p16(INK4A) (3). Also, p16(INK4A) downregulation in
TRANSIENT WOUND HEALING IN THAT THEY BOTH normal human breast stromal fibroblasts is accompanied by an
ENCOMPASS A SUSTAINED MYOFIBROBLASTIC POPULATION increase in ␣-SMA expression, suggesting that absence of
p16(INK4A) could promote myofibroblastic differentiation
While regenerative healing wounds, fibrosis, and tumor-
(3). Additionally, p16(INK4A)-deficient fibroblasts enhance
associated desmoplasia all contain metabolically active myo-
xenograft tumor growth compared with effects imparted by
fibroblasts and EMT cells, it is only under normal healing/
wild-type cells (3). Finally, fibroblasts from oral squamous cell
regenerative (i.e., nonpathological) conditions that the activity carcinoma with a p53 and p16(INK4A) inactive background
of these cells eventually ceases. This may result from activated have been shown to induce the upregulation of ␣-SMA expres-
cells’ (i.e., EMT or myofibroblasts) senescence and subsequent sion, which correlates with poor patient prognosis (215). All
elimination. This can occur through p53- and/or integrin- these data strongly suggest that inactive p16(INK4A) is a
regulated apoptosis or potentially by cell clearance via natural hallmark of TAFs and, therefore, of cancer progression. Re-
killer (NK) cells and/or by interferon gamma secretion (146, garding p53, it has recently been shown that stromal p53
181, 198, 209, 360, 398). From the one side, myofibroblastic decrease in chronic liver damage promotes tumor development
apoptosis could promote pathologic scarring (75). Then again, by a noncell autonomous mechanism that upregulates a tumor
the persistence of myofibroblasts during fibrosis and cancer- promoting M2 macrophage microenvironmental state (226).
associated desmoplasia suggests that these cells escape from Hence, there is strong possibility that at least two distinctive
their natural elimination process by alterations in the myofi- populations of TAFs that drive tumorigenesis coexist at the
broblastic cells-clearance mechanisms. Alternatively, persis- tumor stroma: a hyperproliferative p16 low population and a
tent activated cells may “sense” chronic injury or tumor- rather quiescent SASP positive and presumably p16 high
associated microenvironment (i.e., desmoplasia) and consistently population. This type of TAF heterogeneity reveals a high level
respond “normally.” In addition, fibroblastic cells naturally prog- of complexity at the tumor-stromal interaction niche, which
ress and become senescent in response to a variety of signals. requires a better understanding to improve the design of in-
Nevertheless, under pathological circumstances when clear- creasingly effective stroma targeting therapeutics.
ance mechanisms (i.e., wound resolution) are evaded, cells can If one considers the fact that cell death, for example during
develop what is known as the “senescence associated secretory injury, causes fibrosis, then another important fact of the
phenotype” or SASP, thus turning them into sustained and WHFC triad is that some types of fibrotic tissues are often
proinflammatory senescent cells (61). In response to SASP, characterized by regions of cellular death (219, 269, 288),
affected fibroblastic cells produce a variety of diffusible factors while, similarly, tumors contain noteworthy areas of necrotic
that in turn can drive both fibrosis and tumorigenesis (61), tissue (226). Cell death is well known to activate an inflam-

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


Downloaded from journals.physiology.org/journal/physiolgenomics (180.242.184.036) on February 16, 2024.
Review
234 THE WHFC TRIAD

matory response, which when sustained and prolonged, in turn, carcinomas, where studies have shown promising results tar-
would facilitate both fibrosis and tumorigenesis (35, 195, 389). geting and repressing both tumor and stroma (285). These may
Moreover, whether fibrotic myofibroblasts and TAFs are se- offer an advantage when used against cancer, while some
nescent or hyperproliferative, they also must evade stromal cell chemotherapies and targeted cancer treatments could also have
clearing mechanisms, such as apoptosis, which promote wound great impact on resolving fibrotic chronic diseases. For this we
resolution (Fig. 1). In hypertrophic scars, the presence of have put together Supplemental Table S2, which lists some of
apoptosis-resistant myofibroblasts high in antiapoptotic protein the current common WHFC triad treatments under investiga-
Bcl-2 has been reported (256). Although the mechanisms tion or in clinical use today.
remain unclear, TGF-␤ may induce myofibroblast resistance to
apoptosis (138). Intriguingly, the fact that tumors release a Reflecting Upon the WHFC Triad
variety of molecules that can interfere with the immune re-
sponse as well as with NK cell infiltration and activation (209) This review attempts to highlight the importance of under-
suggests the prevalence of an immunosuppressive microenvi- standing wound healing mechanisms, as a pathway to compre-
ronment that may protect fibrotic myofibroblasts and desmo- hend two subsequent conditions, derived from the persistence
plastic TAFs from active NK cell elimination. For example, it and exacerbation of the healing process. In the absence of
has been shown that TGF-␤ is a potent inhibitor of NK (366). wound resolution, continuous alterations to tissue homeostasis
Although there is not much information about this mechanism engage both stroma and epithelia for the onset of chronic
in cancer and specifically on the tumor stroma, research on the fibrosis and/or cancer. We propose the consolidation of com-
chronic fibrosis field has shown a relationship between the monalities from these three conditions under the concept of the
persistence of myofibroblasts with an immunosuppressed mi- WHFC triad. In other words, similar mechanisms operate in
croenvironment profile (i.e., myofibroblasts attract immune- this triad to sustain a vicious cycle where a fibrotic stroma
suppressive cells) (38, 181, 375). Hence, facilitating the NK could trigger a malignant behavior of epithelial cells, while the
cell-mediated elimination of myofibroblastic and tumoral cells progression of carcinomas also promotes a fibrotic/desmoplas-
is expected to be a feasible approach to counteract the WHFC tic stroma fueling the conditions for both (Fig. 1).
triad (38). The WHFC triad is assembled by common biological re-
sponses from the three described conditions. Several proteins
CONCLUDING REMARKS and signaling pathways encompass a remarkable degree of
overlap amongst wound healing, pathological chronic fibrosis,
Possible Approaches to Clinically Interfere With the WHFC
and cancer (317). The mentioned players can be clustered
Triad
based on their biological mechanisms, in a coagulation/fibri-
Since a convergence between wound healing, cancer, and nolysis-like response, inflammation signaling processes, as
fibrosis is compelling, its implications for fibrosis and cancer well as ECM remodeling mechanisms. These biological occur-
treatment are profound. To achieve effective drug delivery, it is rences are responsible for triggering hyperproliferative cell
crucial to comprehend the challenge that a fibrotic microenvi- behavior and phenotypic/activating modifications such as EMT
ronment represents. For example, due to a dense and altered and myofibroblastic differentiation (Fig. 1 and Supplemental
ECM deposition, the free access for drugs and other molecules, Table S1).
including glucose and oxygen, is severely impaired in desmo- Specifically for cancer, the WHFC triad constitutes a rather
plastic malignancies (65). In this context, it is necessary to fertile ground facilitating genetic mutations and epigenetic
develop novel drug delivery strategies to obtain better results hyperplastic occurrences, which eventually constitute the ma-
while targeting both fibrosis (to obtain stromal normalization) lignant cell population (91). Moreover, an additional difference
and cancer (see Supplemental Table S2). This is true in between chronic fibrosis and cancer is that, although both
particular with regards to carcinomas displaying an intense expand in an immediate field-like effect through the affected
desmoplastic reaction such as in pancreatic cancer (263). tissue, only cancer is known to metastasize to tissues localized
Importantly, it is troubling that current approaches aimed at at considerable distances from the primary lesion, while sec-
treating fibrosis and cancer also counteractively promote the ondary neoplastic niches are actively preconditioned to facili-
loss of homeostatic equilibrium, which in turn further promotes tate metastatic cancer cell adaptation (120). Importantly, cru-
the WHFC triad (Fig. 1). For example, surgical procedures and cial aspects of the WHFC triad that have already started to
radiation are effective in resecting and eliminating cancerous receive attention, such as genetic and epigenetic signatures as
and chronically fibrotic masses. Nevertheless, the same treat- well as microRNA profiles (24, 404), although in need of being
ments also create wounds that must be healed, hence possibly fully understood, are already being proposed to serve as pre-
further promoting fibrosis and hyperplastic growth (2, 49, 73). dictors of the outcome of some fibrosis-related malignancies.
Moreover, while surgery triggers a wound healing response, Hence, we conclude by stating that a better recognition and a
chemotherapy also activates the same mechanisms that induce deeper understanding of the commonalities and disparities
fibrosis (49, 272). Above we stated that fibrotic disease fuels within the WHFC triad will surely provide insights for a more
cancer progression and vice versa (Fig. 1). Since it is possible effective therapeutic targeting of these interconnected pathol-
that radiation and chemotherapy-induced fibrosis also stimulate ogies (Supplemental Table S2).
relapse or latent escape of secondary tumors, a more optimistic
perspective will rely on the use of antifibrotic therapies in ACKNOWLEDGMENTS
cancer treatments. This approach could have a great impact on The authors thank Drs. Kerry Campbell and Sergei Grivennikov from Fox
those malignancies where fibrosis constitutes a clear predispo- Chase Cancer Center (FCCC) and Dr. Rebecca Wells from the University of
sition, such as in the cases of pancreatic or hepatic cell Pennsylvania for insightful comments during the preparation of this document.

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


Downloaded from journals.physiology.org/journal/physiolgenomics (180.242.184.036) on February 16, 2024.
Review
THE WHFC TRIAD 235
Graphic design was by Perla Ovseiovich from Paralelo 19, Mexico City. 17. Barry-Hamilton V, Spangler R, Marshall D, McCauley S, Rodriguez
Proofreading was conducted with the help of Ellen Ragan. HM, Oyasu M, Mikels A, Vaysberg M, Ghermazien H, Wai C,
Current address for B. Rybinski: JVA Group, Dept. 4, Tumor Microenvi- Garcia CA, Velayo AC, Jorgensen B, Biermann D, Tsai D, Green J,
ronment, Avondale, PA. Zaffryar-Eilot S, Holzer A, Ogg S, Thai D, Neufeld G, Van Vlasse-
laer P, Smith V. Allosteric inhibition of lysyl oxidase-like-2 impedes the
GRANTS development of a pathologic microenvironment. Nat Med 16: 1009 –
1017, 2010.
Funding used for the preparation of this manuscript included finances from
18. Basu A, Kligman LH, Samulewicz SJ, Howe CC. Impaired wound
the Commonwealth of Pennsylvania, FCCC’s Keystone Program in Personal-
healing in mice deficient in a matricellular protein SPARC (osteonectin,
ized Kidney Cancer Therapy, a Nodal Temple-FCCC Award, The Bucks
BM-40). BMC Cell Biol 2: 15, 2001.
County Board of Associates, as well as National Cancer Institute Grants
19. Bauvois B. New facets of matrix metalloproteinases MMP-2 and MMP-9
CA-113451 and CA-06927.
as cell surface transducers: outside-in signaling and relationship to tumor
progression. Biochim Biophys Acta 1825: 29 –36, 2012.
DISCLOSURES
20. Beacham DA, Cukierman E. Stromagenesis: the changing face of
No conflicts of interest, financial or otherwise, are declared by the author(s). fibroblastic microenvironments during tumor progression. Sem Cancer
Biol 15: 329 –341, 2005.
AUTHOR CONTRIBUTIONS 21. Behm B, Babilas P, Landthaler M, Schreml S. Cytokines, chemokines
and growth factors in wound healing. J Eur Acad Dermatol Venereol 26:
Author contributions: B.R., J.F.-B., and E.C. drafted manuscript; J.F.-B. 812–820, 2012.
and E.C. conception and design of research; J.F.-B. and E.C. prepared figures; 22. Belfiore A, Genua M, Malaguarnera R. PPAR-gamma agonists and
J.F.-B. and E.C. edited and revised manuscript; E.C. approved final version of their effects on IGF-I receptor signaling: Implications for cancer. PPAR
manuscript.
Res 2009: 830501, 2009.
23. Beno DW, Davis BH. Prostaglandin E suppresses hepatic fibrosis:
REFERENCES
section I. The in vivo approach; section II. The in vitro approach. Am J
1. Abe M, Yokoyama Y, Ishikawa O. A possible mechanism of basic Ther 2: 687–705, 1995.
fibroblast growth factor-promoted scarless wound healing: the induction 24. Bian EB, Zhao B, Huang C, Wang H, Meng XM, Wu BM, Ma TT,
of myofibroblast apoptosis. Eur J Dermatol 22: 46 –53, 2012. Zhang L, Lv XW, Li J. New advances of DNA methylation in liver
2. Abratt RP, Morgan GW. Lung toxicity following chest irradiation in fibrosis, with special emphasis on the crosstalk between microRNAs and
patients with lung cancer. Lung Cancer 35: 103–109, 2002. DNA methylation machinery. Cell Signal 25: 1837–1844, 2013.
3. Al-Ansari MM, Hendrayani SF, Shehata AI, Aboussekhra A. 25. Bielefeld KA, Amini-Nik S, Alman BA. Cutaneous wound healing:
p16(INK4A) represses the paracrine tumor-promoting effects of breast recruiting developmental pathways for regeneration. Cell Mol Life Sci
stromal fibroblasts. Oncogene 32: 2356 –2364, 2013. 70: 2059 –2081, 2013.
4. Amatangelo MD, Bassi DE, Klein-Szanto AJ, Cukierman E. Stroma- 26. Bishen KA, Radhakrishnan R, Satyamoorthy K. The role of basic
derived three-dimensional matrices are necessary and sufficient to pro- fibroblast growth factor in oral submucous fibrosis pathogenesis. J Oral
mote desmoplastic differentiation of normal fibroblasts. Am J Pathol 167: Pathol Med 37: 402–411, 2008.
475–488, 2005. 27. Bjermer L, Lundgren R, Hallgren R. Hyaluronan and type III procol-
5. Annes JP, Munger JS, Rifkin DB. Making sense of latent TGFbeta lagen peptide concentrations in bronchoalveolar lavage fluid in idiopathic
activation. J Cell Sci 116: 217–224, 2003. pulmonary fibrosis. Thorax 44: 126 –131, 1989.
6. Aoki T, Narumiya S. Prostaglandins and chronic inflammation. Trends 28. Black PC, Mize GJ, Karlin P, Greenberg DL, Hawley SJ, True LD,
Pharmacol Sci 33: 304 –311, 2012. Vessella RL, Takayama TK. Overexpression of protease-activated
7. Aprile G, Avellini C, Reni M, Mazzer M, Foltran L, Rossi D, Cereda receptors-1,-2, and-4 (PAR-1, -2, and -4) in prostate cancer. Prostate 67:
S, Iaiza E, Fasola G, Piga A. Biglycan expression and clinical outcome 743–756, 2007.
in patients with pancreatic adenocarcinoma. Tumour Biol 34: 131–137, 29. Bloomston M, Ellison EC, Muscarella P, Al-Saif O, Martin EW,
2013. Melvin WS, Frankel WL. Stromal osteonectin overexpression is asso-
8. Ara T, Declerck YA. Interleukin-6 in bone metastasis and cancer ciated with poor outcome in patients with ampullary cancer. Ann Surg
progression. Eur J Cancer 46: 1223–1231, 2010. Oncol 14: 211–217, 2007.
9. Archontogeorgis K, Steiropoulos P, Tzouvelekis A, Nena E, Bouros 30. Bobba RK, Holly JS, Loy T, Perry MC. Scar carcinoma of the lung: a
D. Lung cancer and interstitial lung diseases: a systematic review. historical perspective. Clin Lung Cancer 12: 148 –154, 2011.
Pulmon Med 2012: 315918, 2012. 31. Bogatkevich GS, Highland KB, Akter T, Silver RM. The PPAR-
10. Atorrasagasti C, Peixoto E, Aquino JB, Kippes N, Malvicini M, gamma agonist rosiglitazone is antifibrotic for scleroderma lung fibro-
Alaniz L, Garcia M, Piccioni F, Fiore EJ, Bayo J, Bataller R, blasts: mechanisms of action and differential racial effects. Pulmon Med
Guruceaga E, Corrales F, Podhajcer O, Mazzolini G. Lack of the 2012: 545172, 2012.
matricellular protein SPARC (secreted protein, acidic and rich in cys- 32. Bonner JC. Regulation of PDGF and its receptors in fibrotic diseases.
teine) attenuates liver fibrogenesis in mice. PLoS One 8: e54962, 2013. Cytokine Growth Factor Rev 15: 255–273, 2004.
11. Auvinen P, Tammi R, Parkkinen J, Tammi M, Agren U, Johansson 33. Borensztajn K, Peppelenbosch MP, Spek CA. Factor Xa: at the
R, Hirvikoski P, Eskelinen M, Kosma VM. Hyaluronan in peritumoral crossroads between coagulation and signaling in physiology and disease.
stroma and malignant cells associates with breast cancer spreading and Trends Mol Med 14: 429 –440, 2008.
predicts survival. Am J Pathol 156: 529 –536, 2000. 34. Borensztajn K, von der Thusen JH, Peppelenbosch MP, Spek CA.
12. Bachem MG, Schunemann M, Ramadani M, Siech M, Beger H, Buck The coagulation factor Xa/protease activated receptor-2 axis in the
A, Zhou S, Schmid-Kotsas A, Adler G. Pancreatic carcinoma cells progression of liver fibrosis: a multifaceted paradigm. J Cell Mol Med 14:
induce fibrosis by stimulating proliferation and matrix synthesis of 143–153, 2010.
stellate cells. Gastroenterology 128: 907–921, 2005. 35. Borges FT, Melo SA, Ozdemir BC, Kato N, Revuelta I, Miller CA,
13. Baghy K, Dezso K, Laszlo V, Fullar A, Peterfia B, Paku S, Nagy P, Gattone VH 2nd, Lebleu VS, Kalluri R. TGF-beta1-containing exo-
Schaff Z, Iozzo RV, Kovalszky I. Ablation of the decorin gene enhances somes from injured epithelial cells activate fibroblasts to initiate tissue
experimental hepatic fibrosis and impairs hepatic healing in mice. Lab regenerative responses and fibrosis. J Am Soc Nephrol 24: 385–392,
Invest 91: 439 –451, 2011. 2013.
14. Baghy K, Iozzo RV, Kovalszky I. Decorin-TGFbeta axis in hepatic 36. Bourboulia D, Stetler-Stevenson WG. Matrix metalloproteinases
fibrosis and cirrhosis. J Histochem Cytochem 60: 262–268, 2012. (MMPs) and tissue inhibitors of metalloproteinases (TIMPs): positive
15. Bailey JM, Mohr AM, Hollingsworth MA. Sonic hedgehog paracrine and negative regulators in tumor cell adhesion. Sem Cancer Biol 20:
signaling regulates metastasis and lymphangiogenesis in pancreatic can- 161–168, 2010.
cer. Oncogene 28: 3513–3525, 2009. 37. Bozyk PD, Moore BB. Prostaglandin E2 and the pathogenesis of
16. Barcellos-Hoff MH, Lyden D, Wang TC. The evolution of the cancer pulmonary fibrosis. Am J Respir Cell Mol Biol 45: 445–452, 2011.
niche during multistage carcinogenesis. Nat Rev Cancer 13: 511–518, 38. Brabletz T. To differentiate or not–routes towards metastasis. Nat Rev
2013. Cancer 12: 425–436, 2012.

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


Downloaded from journals.physiology.org/journal/physiolgenomics (180.242.184.036) on February 16, 2024.
Review
236 THE WHFC TRIAD

39. Bradshaw AD, Reed MJ, Sage EH. SPARC-null mice exhibit acceler- 62. Corte MD, Verez P, Rodriguez JC, Roibas A, Dominguez ML,
ated cutaneous wound closure. J Histochem Cytochem 50: 1–10, 2002. Lamelas ML, Vazquez J, Garcia Muniz JL, Allende MT, Gonzalez
40. Brellier F, Hostettler K, Hotz HR, Ozcakir C, Cologlu SA, Togbe D, LO, Fueyo A, Vizoso F. Tissue-type plasminogen activator (tPA) in
Ryffel B, Roth M, Chiquet-Ehrismann R. Tenascin-C triggers fibrin breast cancer: relationship with clinicopathological parameters and prog-
accumulation by downregulation of tissue plasminogen activator. FEBS nostic significance. Breast Cancer Res Treat 90: 33–40, 2005.
Lett 585: 913–920, 2011. 63. Coulson-Thomas VJ, Coulson-Thomas YM, Gesteira TF, de Paula
41. Burgess HA, Daugherty LE, Thatcher TH, Lakatos HF, Ray DM, CA, Mader AM, Waisberg J, Pinhal MA, Friedl A, Toma L, Nader
Redonnet M, Phipps RP, Sime PJ. PPARgamma agonists inhibit HB. Colorectal cancer desmoplastic reaction up-regulates collagen syn-
TGF-beta induced pulmonary myofibroblast differentiation and collagen thesis and restricts cancer cell invasion. Cell Tiss Res 346: 223–236,
production: implications for therapy of lung fibrosis. Am J Physiol Lung 2011.
Cell Mol Physiol 288: L1146 –L1153, 2005. 64. Cukierman E, Bassi DE. Physico-mechanical aspects of extracellular
42. Butcher DT, Alliston T, Weaver VM. A tense situation: forcing tumour matrix influences on tumorigenic behaviors. Sem Cancer Biol 20: 139 –
progression. Nat Rev Cancer 9: 108 –122, 2009. 145, 2010.
43. Carey WA, Taylor GD, Dean WB, Bristow JD. Tenascin-C deficiency 65. Cukierman E, Khan DR. The benefits and challenges associated with
attenuates TGF-ss-mediated fibrosis following murine lung injury. Am J the use of drug delivery systems in cancer therapy. Biochem Pharmacol
Physiol Lung Cell Mol Physiol 299: L785–L793, 2010. 80: 762–770, 2010.
44. Carothers AM, Davids JS, Damas BC, Bertagnolli MM. Persistent 66. D’Alterio C, Avallone A, Tatangelo F, Delrio P, Pecori B, Cella L,
cyclooxygenase-2 inhibition downregulates NF-␬B, resulting in chronic Pelella A, D’Armiento FP, Carlomagno C, Bianco F, Silvestro L,
intestinal inflammation in the min/⫹ mouse model of colon tumorigen- Pacelli R, Napolitano M, Iaffaioli RV, Scala S. A prognostic model
esis. Cancer Res 70: 4433–4442, 2010. comprising pT stage, N status, and the chemokine receptors CXCR4 and
45. Castelló-Cros R, Khan DR, Simons J, Valianou M, Cukierman E. CXCR7 powerfully predicts outcome in neoadjuvant resistant rectal
Staged stromal extracellular 3D matrices differentially regulate breast cancer patients. Int J Cancer [Epub ahead of print].
cancer cell responses through PI3K and beta1-integrins. BMC Cancer 9: 67. Damm EW, Winklbauer R. PDGF-A controls mesoderm cell orienta-
94, 2009. tion and radial intercalation during Xenopus gastrulation. Development
46. Catarzi S, Favilli F, Romagnoli C, Marcucci T, Picariello L, Tonelli 138: 565–575, 2011.
F, Vincenzini MT, Iantomasi T. Oxidative state and IL-6 production in 68. Davalos D, Akassoglou K. Fibrinogen as a key regulator of inflamma-
intestinal myofibroblasts of Crohn’s disease patients. Inflamm Bowel Dis tion in disease. Sem Immunopathol 34: 43–62, 2012.
17: 1674 –1684, 2011. 69. Davids JS, Carothers AM, Damas BC, Bertagnolli MM. Chronic
47. Chambers RC, Laurent GJ. Coagulation cascade proteases and tissue cyclooxygenase-2 inhibition promotes myofibroblast-associated intesti-
fibrosis. Biochem Soc Transact 30: 194 –200, 2002. nal fibrosis. Cancer Prev Res 3: 348 –358, 2010.
48. Chapman RW, Phillips JE, Hipkin RW, Curran AK, Lundell D, Fine 70. de Lujan Alvarez M, Ronco MT, Ochoa JE, Monti JA, Carnovale
JS. CXCR2 antagonists for the treatment of pulmonary disease. Phar- CE, Pisani GB, Lugano MC, Carrillo MC. Interferon alpha-induced
apoptosis on rat preneoplastic liver is mediated by hepatocytic transform-
macol Therapeut 121: 55–68, 2009.
ing growth factor beta(1). Hepatology 40: 394 –402, 2004.
49. Charpidou AG, Gkiozos I, Tsimpoukis S, Apostolaki D, Dilana KD,
71. De Wever O, Nguyen QD, Van Hoorde L, Bracke M, Bruyneel E,
Karapanagiotou EM, Syrigos KN. Therapy-induced toxicity of the
Gespach C, Mareel M. Tenascin-C and SF/HGF produced by myofi-
lungs: an overview. Anticancer Res 29: 631–639, 2009.
broblasts in vitro provide convergent pro-invasive signals to human colon
50. Chaturvedi P, Gilkes DM, Wong CC, Kshitiz Luo W, Zhang H, Wei
cancer cells through RhoA and Rac. FASEB J 18: 1016 –1018, 2004.
H, Takano N, Schito L, Levchenko A, Semenza GL. Hypoxia-induc-
72. De Wever O, Westbroek W, Verloes A, Bloemen N, Bracke M,
ible factor-dependent breast cancer-mesenchymal stem cell bidirectional
Gespach C, Bruyneel E, Mareel M. Critical role of N-cadherin in
signaling promotes metastasis. J Clin Invest 123: 189 –205, 2013.
myofibroblast invasion and migration in vitro stimulated by colon-
51. Chen L, Necela BM, Su W, Yanagisawa M, Anastasiadis PZ, Fields
cancer-cell-derived TGF-beta or wounding. J Cell Sci 117: 4691–4703,
AP, Thompson EA. Peroxisome proliferator-activated receptor gamma 2004.
promotes epithelial to mesenchymal transformation by Rho GTPase- 73. Demicheli R, Retsky MW, Hrushesky WJ, Baum M, Gukas ID. The
dependent activation of ERK1/2. J Biol Chem 281: 24575–24587, 2006. effects of surgery on tumor growth: a century of investigations. Ann
52. Chen L, Neville RD, Michael DR, Martin J, Luo DD, Thomas DW, Oncol 19: 1821–1828, 2008.
Phillips AO, Bowen T. Identification and analysis of the human hyalu- 74. Derosa CA, Furusato B, Shaheduzzaman S, Srikantan V, Wang Z,
ronan synthase 1 gene promoter reveals Smad3- and Sp3-mediated Chen Y, Seifert M, Ravindranath L, Young D, Nau M, Dobi A,
transcriptional induction. Matrix Biol 31: 373–379, 2012. Werner T, McLeod DG, Vahey MT, Sesterhenn IA, Srivastava S,
53. Chen S, Birk DE. The regulatory roles of small leucine-rich proteogly- Petrovics G. Elevated osteonectin/SPARC expression in primary pros-
cans in extracellular matrix assembly. FEBS J 280: 2120 –2137, 2013. tate cancer predicts metastatic progression. Prost Cancer Prostat Dis 15:
54. Chigurupati S, Arumugam TV, Son TG, Lathia JD, Jameel S, 150 –156, 2012.
Mughal MR, Tang SC, Jo DG, Camandola S, Giunta M, Rakova I, 75. Desmouliere A, Redard M, Darby I, Gabbiani G. Apoptosis mediates
McDonnell N, Miele L, Mattson MP, Poosala S. Involvement of notch the decrease in cellularity during the transition between granulation tissue
signaling in wound healing. PLoS One 2: e1167, 2007. and scar. Am J Pathol 146: 56 –66, 1995.
55. Chlenski A, Cohn SL. Modulation of matrix remodeling by SPARC in 76. Devalaraja RM, Nanney LB, Du J, Qian Q, Yu Y, Devalaraja MN,
neoplastic progression. Sem Cell Dev Biol 21: 55–65, 2010. Richmond A. Delayed wound healing in CXCR2 knockout mice. J
56. Cirri P, Chiarugi P. Cancer associated fibroblasts: the dark side of the Invest Dermatol 115: 234 –244, 2000.
coin. Am J Cancer Res 1: 482–497, 2011. 77. Dishowitz MI, Mutyaba PL, Takacs JD, Barr AM, Engiles JB, Ahn
57. Clark SJ, Bishop PN, Day AJ. The proteoglycan glycomatrix: a sugar J, Hankenson KD. Systemic inhibition of canonical notch signaling
microenvironment essential for complement regulation. Front Immunol results in sustained callus inflammation and alters multiple phases of
4: 412, 2013. fracture healing. PLoS One 8: e68726, 2013.
58. Cohen DJ. Targeting the hedgehog pathway: role in cancer and clinical 78. Du WW, Fang L, Yang X, Sheng W, Yang BL, Seth A, Zhang Y,
implications of its inhibition. Hematol/oncol Clin No Am 26: 565–588, Yang BB, Yee AJ. The role of versican in modulating breast cancer cell
viii, 2012. self-renewal. Mol Cancer Res 11: 443–455, 2013.
59. Conklin MW, Eickhoff JC, Riching KM, Pehlke CA, Eliceiri KW, 79. Du WW, Yang BB, Yang BL, Deng Z, Fang L, Shan SW, Jeyapalan
Provenzano PP, Friedl A, Keely PJ. Aligned collagen is a prognostic Z, Zhang Y, Seth A, Yee AJ. Versican G3 domain modulates breast
signature for survival in human breast carcinoma. Am J Pathol 178: cancer cell apoptosis: a mechanism for breast cancer cell response to
1221–1232, 2011. chemotherapy and EGFR therapy. PLoS One 6: e26396, 2011.
60. Conway SJ, Molkentin JD. Periostin as a heterofunctional regulator of 80. Dube PE, Yan F, Punit S, Girish N, McElroy SJ, Washington MK,
cardiac development and disease. Curr Genomics 9: 548 –555, 2008. Polk DB. Epidermal growth factor receptor inhibits colitis-associated
61. Coppe JP, Desprez PY, Krtolica A, Campisi J. The senescence- cancer in mice. J Clin Invest 122: 2780 –2792, 2012.
associated secretory phenotype: the dark side of tumor suppression. Ann 81. Duell EJ, Lucenteforte E, Olson SH, Bracci PM, Li D, Risch HA,
Rev Pathol 5: 99 –118, 2010. Silverman DT, Ji BT, Gallinger S, Holly EA, Fontham EH, Maison-

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


Downloaded from journals.physiology.org/journal/physiolgenomics (180.242.184.036) on February 16, 2024.
Review
THE WHFC TRIAD 237
neuve P, Bueno-de-Mesquita HB, Ghadirian P, Kurtz RC, Ludwig E, 105. Gallucci RM, Lee EG, Tomasek JJ. IL-6 modulates alpha-smooth
Yu H, Lowenfels AB, Seminara D, Petersen GM, La Vecchia C, muscle actin expression in dermal fibroblasts from IL-6-deficient mice. J
Boffetta P. Pancreatitis and pancreatic cancer risk: a pooled analysis in Invest Dermatol 126: 561–568, 2006.
the International Pancreatic Cancer Case-Control Consortium (PanC4). 106. Garamszegi N, Garamszegi SP, Samavarchi-Tehrani P, Walford E,
Ann Oncol 23: 2964 –2970, 2012. Schneiderbauer MM, Wrana JL, Scully SP. Extracellular matrix-
82. Dugina V, Fontao L, Chaponnier C, Vasiliev J, Gabbiani G. Focal induced transforming growth factor-beta receptor signaling dynamics.
adhesion features during myofibroblastic differentiation are controlled by Oncogene 29: 2368 –2380, 2010.
intracellular and extracellular factors. J Cell Sci 114: 3285–3296, 2001. 107. Gelse K, Poschl E, Aigner T. Collagens–structure, function, biosynthe-
83. Duplantier JG, Dubuisson L, Senant N, Freyburger G, Laurendeau sis. Adv Drug Deliv Rev 55: 1531–1546, 2003.
I, Herbert JM, Desmouliere A, Rosenbaum J. A role for thrombin in 108. Genovese F, Barascuk N, Larsen L, Larsen MR, Nawrocki A, Li Y,
liver fibrosis. Gut 53: 1682–1687, 2004. Zheng Q, Wang J, Veidal SS, Leeming DJ, Karsdal MA. Biglycan
84. Dvorak HF. Tumors: wounds that do not heal. Similarities between fragmentation in pathologies associated with extracellular matrix remod-
tumor stroma generation and wound healing. N Engl J Med 315: 1650 – eling by matrix metalloproteinases. Fibrogenesis Tiss Repair 6: 9, 2013.
1659, 1986. 109. Gharaee-Kermani M, Kasina S, Moore BB, Thomas D, Mehra R,
85. El-Karef A, Yoshida T, Gabazza EC, Nishioka T, Inada H, Sakakura Macoska JA. CXC-type chemokines promote myofibroblast phenocon-
T, Imanaka-Yoshida K. Deficiency of tenascin-C attenuates liver fibro- version and prostatic fibrosis. PLoS One 7: e49278, 2012.
sis in immune-mediated chronic hepatitis in mice. J Pathol 211: 86 –94, 110. Ghezzi F, Cromi A, Siesto G, Giudici S, Serati M, Formenti G,
2007. Franchi M. Prognostic significance of preoperative plasma fibrinogen in
86. El-Serag HB. Epidemiology of viral hepatitis and hepatocellular carci- endometrial cancer. Gynecol Oncol 119: 309 –313, 2010.
noma. Gastroenterology 142: 1264 –1273.e1, 2012. 111. Ghosh AK, Vaughan DE. PAI-1 in tissue fibrosis. J Cell Physiol 227:
87. Elliott CG, Wang J, Guo X, Xu SW, Eastwood M, Guan J, Leask A, 493–507, 2012.
Conway SJ, Hamilton DW. Periostin modulates myofibroblast differ- 112. Ghosh S, Albitar L, LeBaron R, Welch WR, Samimi G, Birrer MJ,
entiation during full-thickness cutaneous wound repair. J Cell Sci 125: Berkowitz RS, Mok SC. Up-regulation of stromal versican expression in
121–132, 2012. advanced stage serous ovarian cancer. Gynecol Oncol 119: 114 –120,
88. Elliott CG, Wang J, Guo X, Xu SW, Eastwood M, Guan J, Leask A, 2010.
Conway SJ, Hamilton DW. Periostin modulates myofibroblast differ- 113. Gill SE, Parks WC. Metalloproteinases and their inhibitors: regulators
entiation during full-thickness cutaneous wound repair. J Cell Sci 125: of wound healing. Int J Biochem Cell Biol 40: 1334 –1347, 2008.
121–132, 2012. 114. Girotti MR, Fernandez M, Lopez JA, Camafeita E, Fernandez EA,
89. Elste AP, Petersen I. Expression of proteinase-activated receptor 1– 4 Albar JP, Benedetti LG, Valacco MP, Brekken RA, Podhajcer OL,
(PAR 1– 4) in human cancer. J Mol Histol 41: 89 –99, 2010. Llera AS. SPARC promotes cathepsin B-mediated melanoma invasive-
90. Eyden B, Banerjee SS, Shenjere P, Fisher C. The myofibroblast and its ness through a collagen I/alpha2beta1 integrin axis. J Invest Dermatol
tumours. J Clin Pathol 62: 236 –249, 2009. 131: 2438 –2447, 2011.
91. Farazi PA, DePinho RA. Hepatocellular carcinoma pathogenesis: from
115. Godbout C, Follonier Castella L, Smith EA, Talele N, Chow ML,
genes to environment. Nat Rev Cancer 6: 674 –687, 2006.
Garonna A, Hinz B. The mechanical environment modulates intracel-
92. Fedak PW, Bai L, Turnbull J, Ngu J, Narine K, Duff HJ. Cell therapy
lular calcium oscillation activities of myofibroblasts. PLoS One 8:
limits myofibroblast differentiation and structural cardiac remodeling:
e64560, 2013.
basic fibroblast growth factor-mediated paracrine mechanism. Circ Heart
116. Goetz JG, Minguet S, Navarro-Lérida I, Lazcano JJ, Samaniego R,
Fail 5: 349 –356, 2012.
Calvo E, Tello M, Osteso-Ibáñez T, Pellinen T, Echarri A, Cerezo A,
93. Fernandez PM, Patierno SR, Rickles FR. Tissue factor and fibrin in
Klein-Szanto AJ, Garcia R, Keely PJ, Sánchez-Mateos P, Cukierman
tumor angiogenesis. Sem Thrombosis Hemostasis 30: 31–44, 2004.
E, Del Pozo MA. Biomechanical remodeling of the microenvironment
94. Filosto S, Becker CR, Goldkorn T. Cigarette smoke induces aberrant
EGF receptor activation that mediates lung cancer development and by stromal caveolin-1 favors tumor invasion and metastasis. Cell 146:
resistance to tyrosine kinase inhibitors. Mol Cancer Therapeut 11: 148 –163, 2011.
795–804, 2012. 117. Goldacre MJ, Wotton CJ, Yeates D, Seagroatt V, Collier J. Liver
95. Franchini M, Mannucci PM. Thrombin and cancer: from molecular cirrhosis, other liver diseases, pancreatitis and subsequent cancer: record
basis to therapeutic implications. Sem Thrombosis Hemostasis 38: 95– linkage study. Eur J Gastroenterol Hepatol 20: 384 –392, 2008.
101, 2012. 118. Greaves NS, Ashcroft KJ, Baguneid M, Bayat A. Current understand-
96. Franciszkiewicz K, Boissonnas A, Boutet M, Combadiere C, Mami- ing of molecular and cellular mechanisms in fibroplasia and angiogenesis
Chouaib F. Role of chemokines and chemokine receptors in shaping the during acute wound healing. J Dermatol Sci 72: 206 –217, 2013.
effector phase of the antitumor immune response. Cancer Res 72: 119. Grivennikov S, Karin E, Terzic J, Mucida D, Yu GY, Vallabha-
6325–6332, 2012. purapu S, Scheller J, Rose-John S, Cheroutre H, Eckmann L, Karin
97. Frantz C, Stewart KM, Weaver VM. The extracellular matrix at a M. IL-6 and Stat3 are required for survival of intestinal epithelial cells
glance. J Cell Sci 123: 4195–4200, 2010. and development of colitis-associated cancer. Cancer Cell 15: 103–113,
98. Fraser JR, Laurent TC, Pertoft H, Baxter E. Plasma clearance, tissue 2009.
distribution and metabolism of hyaluronic acid injected intravenously in 120. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, can-
the rabbit. Biochem J 200: 415–424, 1981. cer. Cell 140: 883–899, 2010.
99. Frey H, Schroeder N, Manon-Jensen T, Iozzo RV, Schaefer L. 121. Grivennikov SI, Wang K, Mucida D, Stewart CA, Schnabl B, Jauch
Biological interplay between proteoglycans and their innate immune D, Taniguchi K, Yu GY, Osterreicher CH, Hung KE, Datz C, Feng
receptors in inflammation. FEBS J 280: 2165–2179, 2013. Y, Fearon ER, Oukka M, Tessarollo L, Coppola V, Yarovinsky F,
100. Fujimoto D, Hirono Y, Goi T, Katayama K, Yamaguchi A. Prognostic Cheroutre H, Eckmann L, Trinchieri G, Karin M. Adenoma-linked
value of protease-activated receptor-1 (PAR-1) and matrix metallopro- barrier defects and microbial products drive IL-23/IL-17-mediated tu-
teinase-1 (MMP-1) in gastric cancer. Anticancer Res 28: 847–854, 2008. mour growth. Nature 491: 254 –258, 2012.
101. Fukunaga-Kalabis M, Martinez G, Nguyen TK, Kim D, Santiago- 122. Gu X, Ma Y, Xiao J, Zheng H, Song C, Gong Y, Xing X. Up-regulated
Walker A, Roesch A, Herlyn M. Tenascin-C promotes melanoma biglycan expression correlates with the malignancy in human colorectal
progression by maintaining the ABCB5-positive side population. Onco- cancers. Clin Exp Med 12: 195–199, 2012.
gene 29: 6115–6124, 2010. 123. Guarino M, Tosoni A, Nebuloni M. Direct contribution of epithelium to
102. Fukushima N, Kikuchi Y, Nishiyama T, Kudo A, Fukayama M. organ fibrosis: epithelial-mesenchymal transition. Hum Pathol 40: 1365–
Periostin deposition in the stroma of invasive and intraductal neoplasms 1376, 2009.
of the pancreas. Mod Pathol 21: 1044 –1053, 2008. 124. Gupta RA, Dubois RN. Controversy: PPAR␥ as a target for treatment of
103. Fuxe J, Karlsson MCI. TGF-␤-induced epithelial-mesenchymal transi- colorectal cancer. Am J Physiol Gastrointest Liver Physiol 283: G266 –
tion: a link between cancer and inflammation. Sem Cancer Biol 22: G269, 2002.
455–461, 2012. 125. Gupta V, Bassi DE, Simons JD, Devarajan K, Al-Saleem T, Uzzo RG,
104. Gales D, Clark C, Manne U, Samuel T. The chemokine CXCL8 in Cukierman E. Elevated expression of stromal palladin predicts poor
carcinogenesis and drug response. ISRN Oncol 2013: 859154, 2013. clinical outcome in renal cell carcinoma. PLoS One 6: e21494, 2011.

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


Downloaded from journals.physiology.org/journal/physiolgenomics (180.242.184.036) on February 16, 2024.
Review
238 THE WHFC TRIAD

126. Hakkinen L, Westermarck J, Kahari VM, Larjava H. Human gran- 148. Hurst NJ Jr, Najy AJ, Ustach CV, Movilla L, Kim HR. Platelet-
ulation-tissue fibroblasts show enhanced proteoglycan gene expression derived growth factor-C (PDGF-C) activation by serine proteases: im-
and altered response to TGF-beta 1. J Dent Res 75: 1767–1778, 1996. plications for breast cancer progression. Biochem J 441: 909 –918, 2012.
127. Hancox RA, Allen MD, Holliday DL, Edwards DR, Pennington CJ, 149. Hwang RF, Moore TT, Hattersley MM, Scarpitti M, Yang B, De-
Guttery DS, Shaw JA, Walker RA, Pringle JH, Jones JL. Tumour- vereaux E, Ramachandran V, Arumugam T, Ji B, Logsdon CD,
associated tenascin-C isoforms promote breast cancer cell invasion and Brown JL, Godin R. Inhibition of the hedgehog pathway targets the
growth by matrix metalloproteinase-dependent and independent mecha- tumor-associated stroma in pancreatic cancer. Mol Cancer Res 10:
nisms. Breast Cancer Res 11: R24, 2009. 1147–1157, 2012.
128. Hardie WD, Davidson C, Ikegami M, Leikauf GD, Le Cras TD, 150. Ichiki Y, Smith EA, LeRoy EC, Trojanowska M. Basic fibroblast
Prestridge A, Whitsett JA, Korfhagen TR. EGF receptor tyrosine growth factor inhibits basal and transforming growth factor-beta induced
kinase inhibitors diminish transforming growth factor-alpha-induced pul- collagen alpha 2(I) gene expression in scleroderma and normal fibro-
monary fibrosis. Am J Physiol Lung Cell Mol Physiol 294: L1217– blasts. J Rheumatol 24: 90 –95, 1997.
L1225, 2008. 151. Imokawa S, Sato A, Hayakawa H, Kotani M, Urano T, Takada A.
129. Harding P. Do COX-2 inhibitors reduce renal fibrosis? J Hypertens 22: Tissue factor expression and fibrin deposition in the lungs of patients
43–45, 2004. with idiopathic pulmonary fibrosis and systemic sclerosis. Am J Respir
130. Hardy KM, Booth BW, Hendrix MJ, Salomon DS, Strizzi L. ErbB/ Crit Care Med 156: 631–636, 1997.
EGF signaling and EMT in mammary development and breast cancer. J 152. Inoue Y, King TE Jr, Barker E, Daniloff E, Newman LS. Basic
Mamm Gland Biol Neoplas 15: 191–199, 2010. fibroblast growth factor and its receptors in idiopathic pulmonary fibrosis
131. Hardy MM, Blomme EA, Lisowski A, Chinn KS, Jones A, Harmon and lymphangioleiomyomatosis. Am J Respir Crit Care Med 166: 765–
JM, Opsahl A, Ornberg RL, Tripp CS. Selective cyclooxygenase-2 773, 2002.
inhibition does not alter keratinocyte wound responses in the mouse 153. Iozzo RV, Sanderson RD. Proteoglycans in cancer biology, tumour
epidermis after abrasion. J Pharmacol Exp Therapeut 304: 959 –967, microenvironment and angiogenesis. J Cell Mol Med 15: 1013–1031,
2003. 2011.
132. Heinrich EL, Lee W, Lu J, Lowy AM, Kim J. Chemokine CXCL12 154. Ishiguro S, Akasaka Y, Kiguchi H, Suzuki T, Imaizumi R, Ishikawa
activates dual CXCR4 and CXCR7-mediated signaling pathways in Y, Ito K, Ishii T. Basic fibroblast growth factor induces down-regulation
pancreatic cancer cells. J Translat Med 10: 68, 2012. of alpha-smooth muscle actin and reduction of myofibroblast areas in
133. Heldin CH. Autocrine PDGF stimulation in malignancies. Ups J Med Sci open skin wounds. Wound Repair Regen 17: 617–625, 2009.
117: 83–91, 2012. 155. Iwaki T, Urano T, Umemura K. PAI-1, progress in understanding the
134. Herbst RS. Review of epidermal growth factor receptor biology. Int J clinical problem and its aetiology. Br J Haematol 157: 291–298, 2012.
Radiat Oncol Biol Physics 59: 21–26, 2004. 156. Jarvinen TA, Ruoslahti E. Target-seeking antifibrotic compound en-
135. Herrick S, Blanc-Brude O, Gray A, Laurent G. Fibrinogen. Int J hances wound healing and suppresses scar formation in mice. Proc Natl
Biochem Cell Biolo 31: 741–746, 1999. Acad Sci USA 107: 21671–21676, 2010.
157. Javelaud D, Pierrat MJ, Mauviel A. Crosstalk between TGF-beta and
136. Hinz B. Formation and function of the myofibroblast during tissue repair.
hedgehog signaling in cancer. FEBS Lett 586: 2016 –2025, 2012.
J Invest Dermatol 127: 526 –537, 2007.
158. Jedeszko C, Victor BC, Podgorski I, Sloane BF. Fibroblast hepatocyte
137. Hinz B. The myofibroblast: paradigm for a mechanically active cell. J
growth factor promotes invasion of human mammary ductal carcinoma in
Biomechan 43: 146 –155, 2010.
situ. Cancer Res 69: 9148 –9155, 2009.
138. Hinz B, Gabbiani G. Fibrosis: recent advances in myofibroblast biology
159. Jiang D, Liang J, Noble PW. Hyaluronan as an immune regulator in
and new therapeutic perspectives. F1000 Biol Rep 2: 78, 2010.
human diseases. Physiol Rev 91: 221–264, 2011.
139. Hinz B, Mastrangelo D, Iselin CE, Chaponnier C, Gabbiani G.
160. Johari V, Loke C. Brief overview of the coagulation cascade. Dis Mon
Mechanical tension controls granulation tissue contractile activity and
58: 421–423, 2012.
myofibroblast differentiation. Am J Pathol 159: 1009 –1020, 2001.
161. Johnson NR, Wang Y. Controlled delivery of heparin-binding EGF-like
140. Hinz B, Phan SH, Thannickal VJ, Prunotto M, Desmouliere A, growth factor yields fast and comprehensive wound healing. J Controlled
Varga J, De Wever O, Mareel M, Gabbiani G. Recent developments Release 166: 124 –129, 2013.
in myofibroblast biology: paradigms for connective tissue remodeling. 162. Ju W, Zhihong Y, Zhiyou Z, Qin H, Dingding W, Li S, Baowei Z,
Am J Pathol 180: 1340 –1355, 2012. Xing W, Ying H, An H. Inhibition of alpha-SMA by the ectodomain of
141. Howard EW, Crider BJ, Updike DL, Bullen EC, Parks EE, Haaksma FGFR2c attenuates lung fibrosis. Mol Med 18: 992–1002, 2012.
CJ, Sherry DM, Tomasek JJ. MMP-2 expression by fibroblasts is 163. Jun JI, Lau LF. The matricellular protein CCN1 induces fibroblast
suppressed by the myofibroblast phenotype. Exp Cell Res 318: 1542– senescence and restricts fibrosis in cutaneous wound healing. Nat Cell
1553, 2012. Biol 12: 676 –685, 2010.
142. Howell DC, Johns RH, Lasky JA, Shan B, Scotton CJ, Laurent GJ, 164. Kagan HM. Intra- and extracellular enzymes of collagen biosynthesis as
Chambers RC. Absence of proteinase-activated receptor-1 signaling biological and chemical targets in the control of fibrosis. Acta Trop 77:
affords protection from bleomycin-induced lung inflammation and fibro- 147–152, 2000.
sis. Am J Pathol 166: 1353–1365, 2005. 165. Kagan HM, Li W. Lysyl oxidase: properties, specificity, and biological
143. Howell DC, Laurent GJ, Chambers RC. Role of thrombin and its roles inside and outside of the cell. J Cell Biochem 88: 660 –672, 2003.
major cellular receptor, protease-activated receptor-1, in pulmonary fi- 166. Kakudo N, Kushida S, Suzuki K, Ogura T, Notodihardjo PV, Hara
brosis. Biochem Soc Transact 30: 211–216, 2002. T, Kusumoto K. Effects of transforming growth factor-beta1 on cell
144. Hsiao Y, Zou T, Ling CC, Hu H, Tao XM, Song HY. Disruption of motility, collagen gel contraction, myofibroblastic differentiation, and
tissue-type plasminogen activator gene in mice aggravated liver fibrosis. extracellular matrix expression of human adipose-derived stem cell. Hum
J Gastroenterol Hepatol 23: e258 –e264, 2008. Cell 25: 87–95, 2012.
145. Huang C, Yang G, Jiang T, Zhu G, Li H, Qiu Z. The effects and 167. Kalluri R. Basement membranes: structure, assembly and role in tumour
mechanisms of blockage of STAT3 signaling pathway on IL-6 inducing angiogenesis. Nat Rev Cancer 3: 422–433, 2003.
EMT in human pancreatic cancer cells in vitro. Neoplasma 58: 396 –405, 168. Kass L, Erler JT, Dembo M, Weaver VM. Mammary epithelial cell:
2011. influence of extracellular matrix composition and organization during
146. Hudspeth K, Pontarini E, Tentorio P, Cimino M, Donadon M, development and tumorigenesis. Int J Biochem Cell Biol 39: 1987–1994,
Torzilli G, Lugli E, Della Bella S, Gershwin ME, Mavilio D. The role 2007.
of natural killer cells in autoimmune liver disease: a comprehensive 169. Kasthuri RS, Taubman MB, Mackman N. Role of tissue factor in
review. J Autoimmun 46: 55–65, 2013. cancer. J Clin Oncol 27: 4834 –4838, 2009.
147. Huet E, Vallee B, Szul D, Verrecchia F, Mourah S, Jester JV, 170. Katoh H, Wang D, Daikoku T, Sun H, Dey SK, Dubois RN. CXCR2-
Hoang-Xuan T, Menashi S, Gabison EE. Extracellular matrix metal- expressing myeloid-derived suppressor cells are essential to promote
loproteinase inducer/CD147 promotes myofibroblast differentiation by colitis-associated tumorigenesis. Cancer Cell 24: 631–644, 2013.
inducing alpha-smooth muscle actin expression and collagen gel con- 171. Kavian N, Servettaz A, Weill B, Batteux F. New insights into the
traction: implications in tissue remodeling. FASEB J 22: 1144 –1154, mechanism of notch signalling in fibrosis. Open Rheumatol J 6: 96 –102,
2008. 2012.

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


Downloaded from journals.physiology.org/journal/physiolgenomics (180.242.184.036) on February 16, 2024.
Review
THE WHFC TRIAD 239
172. Kawada K, Hosogi H, Sonoshita M, Sakashita H, Manabe T, Shima- 192. Kudo A. Periostin in fibrillogenesis for tissue regeneration: periostin
hara Y, Sakai Y, Takabayashi A, Oshima M, Taketo MM. Chemo- actions inside and outside the cell. Cell Mol Life Sci 68: 3201–3207,
kine receptor CXCR3 promotes colon cancer metastasis to lymph nodes. 2011.
Oncogene 26: 4679 –4688, 2007. 193. Kudo Y, Iizuka S, Yoshida M, Nguyen PT, Siriwardena SB, Tsun-
173. Kawada K, Sonoshita M, Sakashita H, Takabayashi A, Yamaoka Y, ematsu T, Ohbayashi M, Ando T, Hatakeyama D, Shibata T, Koi-
Manabe T, Inaba K, Minato N, Oshima M, Taketo MM. Pivotal role zumi K, Maeda M, Ishimaru N, Ogawa I, Takata T. Periostin directly
of CXCR3 in melanoma cell metastasis to lymph nodes. Cancer Res 64: and indirectly promotes tumor lymphangiogenesis of head and neck
4010 –4017, 2004. cancer. PLoS One 7: e44488, 2012.
174. Kawai T, Masaki T, Doi S, Arakawa T, Yokoyama Y, Doi T, Kohno 194. Kulkarni AA, Thatcher TH, Olsen KC, Maggirwar SB, Phipps RP,
N, Yorioka N. PPAR-gamma agonist attenuates renal interstitial fibrosis Sime PJ. PPAR-gamma ligands repress TGFbeta-induced myofibroblast
and inflammation through reduction of TGF-beta. Lab Invest 89: 47–58, differentiation by targeting the PI3K/Akt pathway: implications for
2009. therapy of fibrosis. PLoS One 6: e15909, 2011.
175. Keglowich L, Roth M, Philippova M, Resink T, Tjin G, Oliver B, 195. Kuraishy A, Karin M, Grivennikov SI. Tumor promotion via injury-
Lardinois D, Dessus-Babus S, Gosens R, Hostettler Haack K, Tamm and death-induced inflammation. Immunity 35: 467–477, 2011.
M, Borger P. Bronchial smooth muscle cells of asthmatics promote 196. Kurata K, Maruyama S, Kato S, Sato W, Yamamoto J, Ozaki T,
angiogenesis through elevated secretion of CXC-chemokines (ENA-78, Nitta A, Nabeshima T, Morita Y, Mizuno M, Ito Y, Yuzawa Y,
GRO-alpha, and IL-8). PLoS One 8: e81494, 2013. Matsuo S. Tissue-type plasminogen activator deficiency attenuates peri-
176. Kessenbrock K, Plaks V, Werb Z. Matrix metalloproteinases: regula- toneal fibrosis in mice. Am J Physiol Renal Physiol 297: F1510 –F1517,
tors of the tumor microenvironment. Cell 141: 52–67, 2010. 2009.
177. Khalil N, Xu YD, O’Connor R, Duronio V. Proliferation of pulmonary 197. Kwon Y, Cukierman E, Godwin AK. Differential expression of adhe-
interstitial fibroblasts is mediated by transforming growth factor-beta1- sive molecules and proteases define mechanisms of ovarian tumor cell
induced release of extracellular fibroblast growth factor-2 and phosphor- matrix penetration/invasion. PLoS One 6: e18872, 2011.
ylation of p38 MAPK and JNK. J Biol Chem 280: 43000 –43009, 2005. 198. Lau LF. CCN1/CYR61: the very model of a modern matricellular
178. Khan Z, Khan N, Tiwari RP, Sah NK, Prasad GB, Bisen PS. Biology protein. Cell Mol Life Sci 68: 3149 –3163, 2011.
of Cox-2: an application in cancer therapeutics. Curr Drug Targets 12: 199. Lauth M, Toftgard R. Hedgehog signaling and pancreatic tumor devel-
1082–1093, 2011. opment. Adv Cancer Res 110: 1–17, 2011.
179. Kikuchi Y, Kashima TG, Nishiyama T, Shimazu K, Morishita Y, 200. Le H, Kleinerman R, Lerman OZ, Brown D, Galiano R, Gurtner
Shimazaki M, Kii I, Horie H, Nagai H, Kudo A, Fukayama M. GC, Warren SM, Levine JP, Saadeh PB. Hedgehog signaling is
Periostin is expressed in pericryptal fibroblasts and cancer-associated essential for normal wound healing. Wound Repair Regen 16: 768 –773,
fibroblasts in the colon. J Histochem Cytochem 56: 753–764, 2008. 2008.
180. Kim CJ, Yoshioka N, Tambe Y, Kushima R, Okada Y, Inoue H. 201. Lee HO, Mullins SR, Franco-Barraza J, Valianou M, Cukierman E,
Periostin is down-regulated in high grade human bladder cancers and Cheng JD. FAP-overexpressing fibroblasts produce an extracellular
suppresses in vitro cell invasiveness and in vivo metastasis of cancer matrix that enhances invasive velocity and directionality of pancreatic
cells. Int J Cancer 117: 51–58, 2005. cancer cells. BMC Cancer 11: 245, 2011.
181. Kim JH, Kim HY, Kim S, Chung JH, Park WS, Chung DH. Natural 202. Lee HM, Kang HJ, Park HH, Hong SC, Kim JK, Cho JH. Effect of
killer T (NKT) cells attenuate bleomycin-induced pulmonary fibrosis by peroxisome proliferator-activated receptor gamma agonists on myofibro-
producing interferon-gamma. Am J Pathol 167: 1231–1241, 2005. blast differentiation and collagen production in nasal polyp-derived
182. Kim KH, Chen CC, Monzon RI, Lau LF. Matricellular protein CCN1 fibroblasts. Ann Otol Rhinol Laryngol 118: 721–727, 2009.
promotes regression of liver fibrosis through induction of cellular senes- 203. Lee SE, Lee JH, Ryu KW, Nam BH, Cho SJ, Lee JY, Kim CG, Choi
cence in hepatic myofibroblasts. Mol Cell Biol 33: 2078 –2090, 2013. IJ, Kook MC, Park SR, Kim YW. Preoperative plasma fibrinogen level
183. Klingberg F, Hinz B, White ES. The myofibroblast matrix: implications is a useful predictor of adjacent organ involvement in patients with
for tissue repair and fibrosis. J Pathol 229: 298 –309, 2013. advanced gastric cancer. J Gastr Cancer 12: 81–87, 2012.
184. Koch AE, Polverini PJ, Kunkel SL, Harlow LA, DiPietro LA, Elner 204. Leeming DJ, Byrjalsen I, Jimenez W, Christiansen C, Karsdal MA.
VM, Elner SG, Strieter RM. Interleukin-8 as a macrophage-derived Protein fingerprinting of the extracellular matrix remodelling in a rat
mediator of angiogenesis. Science 258: 1798 –1801, 1992. model of liver fibrosis–a serological evaluation. Liver Int 33: 439 –447,
185. Kojima Y, Acar A, Eaton EN, Mellody KT, Scheel C, Ben-Porath I, 2013.
Onder TT, Wang ZC, Richardson AL, Weinberg RA, Orimo A. 205. Leeming DJ, Sand JM, Nielsen MJ, Genovese F, Martinez FJ,
Autocrine TGF-beta and stromal cell-derived factor-1 (SDF-1) signaling Hogaboam CM, Han MK, Klickstein LB, Karsdal MA. Serological
drives the evolution of tumor-promoting mammary stromal myofibro- investigation of the collagen degradation profile of patients with chronic
blasts. Proc Natl Acad Sci USA 107: 20009 –20014, 2010. obstructive pulmonary disease or idiopathic pulmonary fibrosis. Biomark
186. Kolb M, Margetts PJ, Sime PJ, Gauldie J. Proteoglycans decorin and Insights 7: 119 –126, 2012.
biglycan differentially modulate TGF-beta-mediated fibrotic responses in 206. Leight JL, Wozniak MA, Chen S, Lynch ML, Chen CS. Matrix
the lung. Am J Physiol Lung Cell Mol Physiol 280: L1327–L1334, 2001. rigidity regulates a switch between TGF-beta1-induced apoptosis and
187. Kolodsick JE, Peters-Golden M, Larios J, Toews GB, Thannickal epithelial-mesenchymal transition. Mol Biol Cell 23: 781–791, 2012.
VJ, Moore BB. Prostaglandin E2 inhibits fibroblast to myofibroblast 207. Leins A, Riva P, Lindstedt R, Davidoff MS, Mehraein P, Weis S.
transition via E. prostanoid receptor 2 signaling and cyclic adenosine Expression of tenascin-C in various human brain tumors and its relevance
monophosphate elevation Am J Resp Cell Mol Biol 29: 537–544, 2003. for survival in patients with astrocytoma. Cancer 98: 2430 –2439, 2003.
188. Konstantinopoulos PA, Vandoros GP, Karamouzis MV, Gkermpesi 208. Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, Fong
M, Sotiropoulou-Bonikou G, Papavassiliou AG. EGF-R is expressed SF, Csiszar K, Giaccia A, Weninger W, Yamauchi M, Gasser DL,
and AP-1 and NF-kappaB are activated in stromal myofibroblasts sur- Weaver VM. Matrix crosslinking forces tumor progression by enhancing
rounding colon adenocarcinomas paralleling expression of COX-2 and integrin signaling. Cell 139: 891–906, 2009.
VEGF. Cell Oncol 29: 477–482, 2007. 209. Levy EM, Roberti MP, Mordoh J. Natural killer cells in human cancer:
189. Koukourakis MI, Giatromanolaki A, Brekken RA, Sivridis E, Gatter from biological functions to clinical applications. J Biomed Biotechnol
KC, Harris AL, Sage EH. Enhanced expression of SPARC/osteonectin 2011: 676198, 2011.
in the tumor-associated stroma of non-small cell lung cancer is correlated 210. Lewis A, Steadman R, Manley P, Craig K, de la Motte C, Hascall V,
with markers of hypoxia/acidity and with poor prognosis of patients. Phillips AO. Diabetic nephropathy, inflammation, hyaluronan and inter-
Cancer Res 63: 5376 –5380, 2003. stitial fibrosis. Histol Histopathol 23: 731–739, 2008.
190. Krizhanovsky V, Yon M, Dickins RA, Hearn S, Simon J, Miething C, 211. Lewis MP, Lygoe KA, Nystrom ML, Anderson WP, Speight PM,
Yee H, Zender L, Lowe SW. Senescence of activated stellate cells limits Marshall JF, Thomas GJ. Tumour-derived TGF-beta1 modulates myo-
liver fibrosis. Cell 134: 657–667, 2008. fibroblast differentiation and promotes HGF/SF-dependent invasion of
191. Kubo H, Hayashi T, Ago K, Ago M, Kanekura T, Ogata M. Temporal squamous carcinoma cells. Br J Cancer 90: 822–832, 2004.
expression of wound healing-related genes in skin burn injury. Leg Med 212. Li J, Chen J, Kirsner R. Pathophysiology of acute wound healing. Clin
16: 8 –13, 2014. Dermatol 25: 9 –18, 2007.

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


Downloaded from journals.physiology.org/journal/physiolgenomics (180.242.184.036) on February 16, 2024.
Review
240 THE WHFC TRIAD

213. Li Y, Jiang D, Liang J, Meltzer EB, Gray A, Miura R, Wogensen L, 236. Maniati E, Bossard M, Cook N, Candido JB, Emami-Shahri N,
Yamaguchi Y, Noble PW. Severe lung fibrosis requires an invasive Nedospasov SA, Balkwill FR, Tuveson DA, Hagemann T. Crosstalk
fibroblast phenotype regulated by hyaluronan and CD44. J Exp Med 208: between the canonical NF-kappaB and Notch signaling pathways inhibits
1459 –1471, 2011. PPARgamma expression and promotes pancreatic cancer progression in
214. Lian H, Ma Y, Feng J, Dong W, Yang Q, Lu D, Zhang L. Heparin- mice. J Clin Invest 121: 4685–4699, 2011.
binding EGF-like growth factor induces heart interstitial fibrosis via an 237. Marcos V, Zhou Z, Yildirim AO, Bohla A, Hector A, Vitkov L,
Akt/mTor/p70s6k pathway. PLoS One 7: e44946, 2012. Wiedenbauer EM, Krautgartner WD, Stoiber W, Belohradsky BH,
215. Lim KP, Cirillo N, Hassona Y, Wei W, Thurlow JK, Cheong SC, Rieber N, Kormann M, Koller B, Roscher A, Roos D, Griese M,
Pitiyage G, Parkinson EK, Prime SS. Fibroblast gene expression Eickelberg O, Doring G, Mall MA, Hartl D. CXCR2 mediates NADPH
profile reflects the stage of tumour progression in oral squamous cell oxidase-independent neutrophil extracellular trap formation in cystic fibrosis
carcinoma. J Pathol 223: 459 –469, 2011. airway inflammation. Nat Med 16: 1018 –1023, 2010.
216. Lin Y, Xie WF, Chen YX, Zhang X, Zeng X, Qiang H, Chen WZ, 238. Margadant C, Sonnenberg A. Integrin-TGF-beta crosstalk in fibrosis,
Yang XJ, Han ZG, Zhang ZB. Treatment of experimental hepatic cancer and wound healing. EMBO Rep 11: 97–105, 2010.
fibrosis by combinational delivery of urokinase-type plasminogen acti- 239. Marra F, Efsen E, Romanelli RG, Caligiuri A, Pastacaldi S, Batig-
vator and hepatocyte growth factor genes. Liver Int 25: 796 –807, 2005. nani G, Bonacchi A, Caporale R, Laffi G, Pinzani M, Gentilini P.
217. Lipponen P, Aaltomaa S, Tammi R, Tammi M, Agren U, Kosma Ligands of peroxisome proliferator-activated receptor gamma modulate
VM. High stromal hyaluronan level is associated with poor differentia- profibrogenic and proinflammatory actions in hepatic stellate cells. Gas-
tion and metastasis in prostate cancer. Eur J Cancer 37: 849 –856, 2001. troenterology 119: 466 –478, 2000.
218. Liu F, Mih JD, Shea BS, Kho AT, Sharif AS, Tager AM, Tschump- 240. Maruhashi T, Kii I, Saito M, Kudo A. Interaction between periostin
erlin DJ. Feedback amplification of fibrosis through matrix stiffening and BMP-1 promotes proteolytic activation of lysyl oxidase. J Biol Chem
and COX-2 suppression. J Cell Biol 190: 693–706, 2010. 285: 13294 –13303, 2010.
219. Liu Y. Cellular and molecular mechanisms of renal fibrosis. Nat Rev 241. Masamune A, Kikuta K, Satoh M, Sakai Y, Satoh A, Shimosegawa T.
Nephrol 7: 684 –696, 2011. Ligands of peroxisome proliferator-activated receptor-gamma block ac-
220. Liu Y, Cai S, Shu XZ, Shelby J, Prestwich GD. Release of basic tivation of pancreatic stellate cells. J Biol Chem 277: 141–147, 2002.
fibroblast growth factor from a crosslinked glycosaminoglycan hydrogel 242. Massague J. TGFbeta in cancer. Cell 134: 215–230, 2008.
promotes wound healing. Wound Repair Regen 15: 245–251, 2007. 243. Matise LA, Palmer TD, Ashby WJ, Nashabi A, Chytil A, Aakre M,
221. Liu Y, Li W, Li X, Tai Y, Lu Q, Yang N, Jiang J. Expression and Pickup MW, Gorska AE, Zijlstra A, Moses HL. Lack of transforming
significance of biglycan in endometrial cancer. Arch Gynecol Obstet growth factor-beta signaling promotes collective cancer cell invasion
[Epub ahead of print]. through tumor-stromal crosstalk. Breast Cancer Res 14: R98, 2012.
222. Liu Y, Liu BA. Enhanced proliferation, invasion, and epithelial-mesen- 244. McGrath MH. The effect of prostaglandin inhibitors on wound contrac-
chymal transition of nicotine-promoted gastric cancer by periostin. World tion and the myofibroblast. Plast Reconstr Surg 69: 74 –85, 1982.
J Gastroenterol 17: 2674 –2680, 2011. 245. Means AL, Ray KC, Singh AB, Washington MK, Whitehead RH,
Harris RC Jr, Wright CV, Coffey RJ Jr, Leach SD. Overexpression
223. Liu Z, Hartman YE, Warram JM, Knowles JA, Sweeny L, Zhou T,
of heparin-binding EGF-like growth factor in mouse pancreas results in
Rosenthal EL. Fibroblast growth factor receptor mediates fibroblast-
fibrosis and epithelial metaplasia. Gastroenterology 124: 1020 –1036,
dependent growth in EMMPRIN-depleted head and neck cancer tumor
2003.
cells. Mol Cancer Res 9: 1008 –1017, 2011.
246. Meng F, Wang K, Aoyama T, Grivennikov SI, Paik Y, Scholten D,
224. Lobry C, Oh P, Aifantis I. Oncogenic and tumor suppressor functions
Cong M, Iwaisako K, Liu X, Zhang M, Osterreicher CH, Stickel F,
of Notch in cancer: it’s NOTCH what you think. J Exp Med 208:
Ley K, Brenner DA, Kisseleva T. Interleukin-17 signaling in inflam-
1931–1935, 2011.
matory, Kupffer cells, and hepatic stellate cells exacerbates liver fibrosis
225. Lu K, Zhu Y, Sheng L, Liu L, Shen L, Wei Q. Serum fibrinogen level
in mice. Gastroenterology 143: 765–776, 2012.
predicts the therapeutic response and prognosis in patients with locally
247. Merline R, Lazaroski S, Babelova A, Tsalastra-Greul W, Pfeilschifter J,
advanced rectal cancer. Hepatogastroenterology 58: 1507–1510, 2011. Schluter KD, Gunther A, Iozzo RV, Schaefer RM, Schaefer L. Decorin
226. Lujambio A, Akkari L, Simon J, Grace D, Tschaharganeh DF, deficiency in diabetic mice: aggravation of nephropathy due to overexpres-
Bolden JE, Zhao Z, Thapar V, Joyce JA, Krizhanovsky V, Lowe SW. sion of profibrotic factors, enhanced apoptosis and mononuclear cell infil-
Non-cell-autonomous tumor suppression by p53. Cell 153: 449 –460, tration. J Physiol Pharmacol 60, Suppl 4: 5–13, 2009.
2013. 248. Michalik L, Wahli W. Involvement of PPAR nuclear receptors in tissue
227. Lynch SE, Nixon JC, Colvin RB, Antoniades HN. Role of platelet- injury and wound repair. J Clin Invest 116: 598 –606, 2006.
derived growth factor in wound healing: synergistic effects with other 249. Michaylira CZ, Wong GS, Miller CG, Gutierrez CM, Nakagawa H,
growth factors. Proc Natl Acad Sci USA 84: 7696 –7700, 1987. Hammond R, Klein-Szanto AJ, Lee JS, Kim SB, Herlyn M, Diehl JA,
228. Ma A, Zhao B, Boulton M, Albon J. A role for Notch signaling in Gimotty P, Rustgi AK. Periostin, a cell adhesion molecule, facilitates
corneal wound healing. Wound Repair Regen 19: 98 –106, 2011. invasion in the tumor microenvironment and annotates a novel tumor-
229. Madala SK, Schmidt S, Davidson C, Ikegami M, Wert S, Hardie invasive signature in esophageal cancer. Cancer Res 70: 5281–5292,
WD. MEK-ERK pathway modulation ameliorates pulmonary fibrosis 2010.
associated with epidermal growth factor receptor activation. Am J Respir 250. Midwood KS, Hussenet T, Langlois B, Orend G. Advances in tenas-
Cell Mol Biol 46: 380 –388, 2012. cin-C biology. Cell Mol Life Sci 68: 3175–3199, 2011.
230. Maharaj S, Shimbori C, Kolb M. Fibrocytes in pulmonary fibrosis: a 251. Midwood KS, Orend G. The role of tenascin-C in tissue injury and
brief synopsis. Eur Respir Rev 22: 552–557, 2013. tumorigenesis. J Cell Commun Signal 3: 287–310, 2009.
231. Maihofner C, Charalambous MP, Bhambra U, Lightfoot T, Gei- 252. Milam JE, Keshamouni VG, Phan SH, Hu B, Gangireddy SR,
sslinger G, Gooderham NJ. Expression of cyclooxygenase-2 parallels Hogaboam CM, Standiford TJ, Thannickal VJ, Reddy RC. PPAR-
expression of interleukin-1beta, interleukin-6 and NF-kappaB in human gamma agonists inhibit profibrotic phenotypes in human lung fibroblasts
colorectal cancer. Carcinogenesis 24: 665–671, 2003. and bleomycin-induced pulmonary fibrosis. Am J Physiol Lung Cell Mol
232. Maisonneuve P, Marshall BC, Lowenfels AB. Risk of pancreatic Physiol 294: L891–L901, 2008.
cancer in patients with cystic fibrosis. Gut 56: 1327–1328, 2007. 253. Milatovic S, Nanney LB, Yu Y, White JR, Richmond A. Impaired
233. Maki JM, Sormunen R, Lippo S, Kaarteenaho-Wiik R, Soininen R, healing of nitrogen mustard wounds in CXCR2 null mice. Wound Repair
Myllyharju J. Lysyl oxidase is essential for normal development and Regen 11: 213–219, 2003.
function of the respiratory system and for the integrity of elastic and 254. Montesano R, Orci L. Transforming growth factor beta stimulates
collagen fibers in various tissues. Am J Pathol 167: 927–936, 2005. collagen-matrix contraction by fibroblasts: implications for wound heal-
234. Maltseva O, Folger P, Zekaria D, Petridou S, Masur SK. Fibroblast ing. Proc Natl Acad Sci USA 85: 4894 –4897, 1988.
growth factor reversal of the corneal myofibroblast phenotype. Invest 255. Morra L, Moch H. Periostin expression and epithelial-mesenchymal
Ophthalmol Vis Sci 42: 2490 –2495, 2001. transition in cancer: a review and an update. Virchows Arch 459:
235. Maneva-Radicheva L, Ebert U, Dimoudis N, Altankov G. Fibroblast 465–475, 2011.
remodeling of adsorbed collagen type IV is altered in contact with cancer 256. Moulin V, Larochelle S, Langlois C, Thibault I, Lopez-Valle CA, Roy
cells. Histol Histopathol 23: 833–842, 2008. M. Normal skin wound and hypertrophic scar myofibroblasts have

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


Downloaded from journals.physiology.org/journal/physiolgenomics (180.242.184.036) on February 16, 2024.
Review
THE WHFC TRIAD 241
differential responses to apoptotic inductors. J Cell Physiol 198: 350 – growth and angiogenesis through elevated SDF-1/CXCL12 secretion.
358, 2004. Cell 121: 335–348, 2005.
257. Munger JS, Sheppard D. Cross talk among TGF-beta signaling path- 276. Oskarsson T, Acharyya S, Zhang XH, Vanharanta S, Tavazoie SF,
ways, integrins, and the extracellular matrix. Cold Spring Harbor Per- Morris PG, Downey RJ, Manova-Todorova K, Brogi E, Massague J.
spect Biol 3: a005017, 2011. Breast cancer cells produce tenascin C as a metastatic niche component
258. Murakami T, Kawada K, Iwamoto M, Akagami M, Hida K, Nakani- to colonize the lungs. Nat Med 17: 867–874, 2011.
shi Y, Kanda K, Kawada M, Seno H, Taketo MM, Sakai Y. The role 277. Otranto M, Sarrazy V, Bonte F, Hinz B, Gabbiani G, Desmouliere A.
of CXCR3 and CXCR4 in colorectal cancer metastasis. Int J Cancer 132: The role of the myofibroblast in tumor stroma remodeling. Cell Adhes
276 –287, 2013. Migrat 6: 203–219, 2012.
259. Murata T, Mizushima H, Chinen I, Moribe H, Yagi S, Hoffman RM, 278. Page-McCaw A, Ewald AJ, Werb Z. Matrix metalloproteinases and the
Kimura T, Yoshino K, Ueda Y, Enomoto T, Mekada E. HB-EGF and regulation of tissue remodelling. Nat Rev Mol Cell Biol 8: 221–233,
PDGF mediate reciprocal interactions of carcinoma cells with cancer- 2007.
associated fibroblasts to support progression of uterine cervical cancers. 279. Paron I, Berchtold S, Voros J, Shamarla M, Erkan M, Hofler H,
Cancer Res 71: 6633–6642, 2011. Esposito I. Tenascin-C enhances pancreatic cancer cell growth and
260. Nagy JA, Brown LF, Senger DR, Lanir N, Van de Water L, Dvorak motility and affects cell adhesion through activation of the integrin
AM, Dvorak HF. Pathogenesis of tumor stroma generation: a critical pathway. PLoS One 6: e21684, 2011.
role for leaky blood vessels and fibrin deposition. Biochim Biophys Acta 280. Parsian H, Rahimipour A, Nouri M, Somi MH, Qujeq D, Fard MK,
948: 305–326, 1989. Agcheli K. Serum hyaluronic acid and laminin as biomarkers in liver
261. Naik PK, Bozyk PD, Bentley JK, Popova AP, Birch CM, Wilke CA, fibrosis. J Gastrointest Liver Dis 19: 169 –174, 2010.
Fry CD, White ES, Sisson TH, Tayob N, Carnemolla B, Orecchia P, 281. Pastore S, Mascia F, Mariani V, Girolomoni G. The epidermal growth
Flaherty KR, Hershenson MB, Murray S, Martinez FJ, Moore BB. factor receptor system in skin repair and inflammation. J Invest Dermatol
Periostin promotes fibrosis and predicts progression in patients with 128: 1365–1374, 2008.
idiopathic pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 303: 282. Paszek MJ, Zahir N, Johnson KR, Lakins JN, Rozenberg GI, Gefen
L1046 –L1056, 2012. A, Reinhart-King CA, Margulies SS, Dembo M, Boettiger D, Ham-
262. Nakaya I, Wada T, Furuichi K, Sakai N, Kitagawa K, Yokoyama H, mer DA, Weaver VM. Tensional homeostasis and the malignant phe-
Ishida Y, Kondo T, Sugaya T, Kawachi H, Shimizu F, Narumi S, notype. Cancer Cell 8: 241–254, 2005.
Haino M, Gerard C, Matsushima K, Kaneko S. Blockade of IP-10/ 283. Peinado H, Moreno-Bueno G, Hardisson D, Perez-Gomez E, Santos
CXCR3 promotes progressive renal fibrosis. Nephron Exp Nephrol 107: V, Mendiola M, de Diego JI, Nistal M, Quintanilla M, Portillo F,
e12–e21, 2007. Cano A. Lysyl oxidase-like 2 as a new poor prognosis marker of
263. Neesse A, Krug S, Gress TM, Tuveson DA, Michl P. Emerging squamous cell carcinomas. Cancer Res 68: 4541–4550, 2008.
concepts in pancreatic cancer medicine: targeting the tumor stroma. 284. Phillips RJ, Burdick MD, Hong K, Lutz MA, Murray LA, Xue YY,
OncoTargets Ther 7: 33–43, 2013. Belperio JA, Keane MP, Strieter RM. Circulating fibrocytes traffic to
264. Neill T, Schaefer L, Iozzo RV. Decorin: a guardian from the matrix. Am
the lungs in response to CXCL12 and mediate fibrosis. J Clin Invest 114:
J Pathol 181: 380 –387, 2012.
438 –446, 2004.
265. Ng CP, Hinz B, Swartz MA. Interstitial fluid flow induces myofibroblast
285. Piccioni F, Malvicini M, Garcia MG, Rodriguez A, Atorrasagasti C,
differentiation and collagen alignment in vitro. J Cell Sci 118: 4731–
Kippes N, Piedra Buena IT, Rizzo MM, Bayo J, Aquino J, Viola M,
4739, 2005.
Passi A, Alaniz L, Mazzolini G. Antitumor effects of hyaluronic acid
266. Niedworok C, Rock K, Kretschmer I, Freudenberger T, Nagy N,
inhibitor 4-methylumbelliferone in an orthotopic hepatocellular carci-
Szarvas T, Vom Dorp F, Reis H, Rubben H, Fischer JW. Inhibitory
noma model in mice. Glycobiology 22: 400 –410, 2012.
role of the small leucine-rich proteoglycan biglycan in bladder cancer.
286. Pietras K, Ostman A. Hallmarks of cancer: interactions with the tumor
PLoS One 8: e80084, 2013.
stroma. Exp Cell Res 316: 1324 –1331, 2010.
267. Nikitovic D, Berdiaki A, Banos A, Tsatsakis A, Karamanos NK,
Tzanakakis GN. Could growth factor-mediated extracellular matrix 287. Pietras K, Pahler J, Bergers G, Hanahan D. Functions of paracrine
deposition and degradation offer the ground for directed pharmacological PDGF signaling in the proangiogenic tumor stroma revealed by pharma-
targeting in fibrosarcoma? Curr Med Chem 20: 2868 –2880, 2013. cological targeting. PLoS Med 5: e19, 2008.
268. Nishida M, Okumura Y, Ozawa S, Shiraishi I, Itoi T, Hamaoka K. 288. Pinzani M, Rombouts K. Liver fibrosis: from the bench to clinical
MMP-2 inhibition reduces renal macrophage infiltration with increased targets. Dig Liver Dis 36: 231–242, 2004.
fibrosis in UUO. Biochem Biophys Res Commun 354: 133–139, 2007. 289. Pirinen R, Tammi R, Tammi M, Hirvikoski P, Parkkinen JJ, Jo-
269. Noble PW, Barkauskas CE, Jiang D. Pulmonary fibrosis: patterns and hansson R, Bohm J, Hollmen S, Kosma VM. Prognostic value of
perpetrators. J Clin Invest 122: 2756 –2762, 2012. hyaluronan expression in non-small-cell lung cancer: Increased stromal
270. Norris RA, Damon B, Mironov V, Kasyanov V, Ramamurthi A, expression indicates unfavorable outcome in patients with adenocarci-
Moreno-Rodriguez R, Trusk T, Potts JD, Goodwin RL, Davis J, noma. Int J Cancer 95: 12–17, 2001.
Hoffman S, Wen X, Sugi Y, Kern CB, Mjaatvedt CH, Turner DK, 290. Polterauer S, Grimm C, Seebacher V, Concin N, Marth C, Tomovski
Oka T, Conway SJ, Molkentin JD, Forgacs G, Markwald RR. C, Husslein H, Leipold H, Hefler-Frischmuth K, Tempfer C, Rein-
Periostin regulates collagen fibrillogenesis and the biomechanical prop- thaller A, Hefler L. Plasma fibrinogen levels and prognosis in patients
erties of connective tissues. J Cell Biochem 101: 695–711, 2007. with ovarian cancer: a multicenter study. Oncologist 14: 979 –985, 2009.
271. Novitskiy SV, Pickup MW, Gorska AE, Owens P, Chytil A, Aakre M, 291. Porsch H, Bernert B, Mehic M, Theocharis AD, Heldin CH, Heldin
Wu H, Shyr Y, Moses HL. TGF-beta receptor II loss promotes mam- P. Efficient TGFbeta-induced epithelial-mesenchymal transition depends
mary carcinoma progression by Th17 dependent mechanisms. Cancer on hyaluronan synthase HAS2. Oncogene 32: 4355–4365, 2013.
Discov 1: 430 –441, 2011. 292. Prajapati RT, Chavally-Mis B, Herbage D, Eastwood M, Brown RA.
272. Ohbayashi M, Suzuki M, Yashiro Y, Fukuwaka S, Yasuda M, Mechanical loading regulates protease production by fibroblasts in three-
Kohyama N, Kobayashi Y, Yamamoto T. Induction of pulmonary dimensional collagen substrates. Wound Repair Regen 8: 226 –237, 2000.
fibrosis by methotrexate treatment in mice lung in vivo and in vitro. J 293. Preaux AM, Mallat A, Nhieu JT, D’Ortho MP, Hembry RM, Mavier
Toxicol Sci 35: 653–661, 2010. P. Matrix metalloproteinase-2 activation in human hepatic fibrosis reg-
273. Okuda H, Kobayashi A, Xia B, Watabe M, Pai SK, Hirota S, Xing F, ulation by cell-matrix interactions. Hepatology 30: 944 –950, 1999.
Liu W, Pandey PR, Fukuda K, Modur V, Ghosh A, Wilber A, 294. Provenzano PP, Eliceiri KW, Campbell JM, Inman DR, White JG,
Watabe K. Hyaluronan synthase HAS2 promotes tumor progression in Keely PJ. Collagen reorganization at the tumor-stromal interface facil-
bone by stimulating the interaction of breast cancer stem-like cells with itates local invasion. BMC Med 4: 38, 2006.
macrophages and stromal cells. Cancer Res 72: 537–547, 2012. 295. Provenzano PP, Inman DR, Eliceiri KW, Knittel JG, Yan L, Rueden
274. Orend G, Chiquet-Ehrismann R. Tenascin-C induced signaling in CT, White JG, Keely PJ. Collagen density promotes mammary tumor
cancer. Cancer Lett 244: 143–163, 2006. initiation and progression. BMC Med 6: 11, 2008.
275. Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, 296. Qiu J, Yu Y, Fu Y, Ye F, Xie X, Lu W. Preoperative plasma fibrinogen,
Naeem R, Carey VJ, Richardson AL, Weinberg RA. Stromal fibro- platelet count and prognosis in epithelial ovarian cancer. J Obstet
blasts present in invasive human breast carcinomas promote tumor Gynaecol Res 38: 651–657, 2012.

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


Downloaded from journals.physiology.org/journal/physiolgenomics (180.242.184.036) on February 16, 2024.
Review
242 THE WHFC TRIAD

297. Reka AK, Kurapati H, Narala VR, Bommer G, Chen J, Standiford 316. Schaefer L, Macakova K, Raslik I, Micegova M, Grone HJ, Schon-
TJ, Keshamouni VG. Peroxisome proliferator-activated receptor- herr E, Robenek H, Echtermeyer FG, Grassel S, Bruckner P,
gamma activation inhibits tumor metastasis by antagonizing Smad3- Schaefer RM, Iozzo RV, Kresse H. Absence of decorin adversely
mediated epithelial-mesenchymal transition. Mol Cancer Therapeut 9: influences tubulointerstitial fibrosis of the obstructed kidney by enhanced
3221–3232, 2010. apoptosis and increased inflammatory reaction. Am J Pathol 160: 1181–
298. Rhodes LV, Antoon JW, Muir SE, Elliott S, Beckman BS, Burow 1191, 2002.
ME. Effects of human mesenchymal stem cells on ER-positive human 317. Schafer M, Werner S. Cancer as an overhealing wound: an old hypoth-
breast carcinoma cells mediated through ER-SDF-1/CXCR4 crosstalk. esis revisited. Nat Rev Mol Cell Biol 9: 628 –638, 2008.
Mol Cancer 9: 295, 2010. 318. Schaffner F, Ruf W. Tissue factor and PAR2 signaling in the tumor
299. Rhodes LV, Bratton MR, Zhu Y, Tilghman SL, Muir SE, Salvo VA, microenvironment. Arterioscler Thromb Vasc Biol 29: 1999 –2004, 2009.
Tate CR, Elliott S, Nephew KP, Collins-Burow BM, Burow ME. 319. Schietke R, Warnecke C, Wacker I, Schodel J, Mole DR, Campean
Effects of SDF-1-CXCR4 signaling on microRNA expression and tu- V, Amann K, Goppelt-Struebe M, Behrens J, Eckardt KU, Wiesener
morigenesis in estrogen receptor-alpha (ER-alpha)-positive breast cancer MS. The lysyl oxidases LOX and LOXL2 are necessary and sufficient to
cells. Exp Cell Res 317: 2573–2581, 2011. repress E-cadherin in hypoxia: insights into cellular transformation pro-
300. Rhodes LV, Short SP, Neel NF, Salvo VA, Zhu Y, Elliott S, Wei Y, cesses mediated by HIF-1. J Biol Chem 285: 6658 –6669, 2010.
Yu D, Sun M, Muir SE, Fonseca JP, Bratton MR, Segar C, Tilghman 320. Schliemann C, Neri D. Antibody-based targeting of the tumor vascula-
SL, Sobolik-Delmaire T, Horton LW, Zaja-Milatovic S, Collins- ture. Biochim Biophys Acta 1776: 175–192, 2007.
Burow BM, Wadsworth S, Beckman BS, Wood CE, Fuqua SA, 321. Seebacher V, Polterauer S, Grimm C, Husslein H, Leipold H, Hefler-
Nephew KP, Dent P, Worthylake RA, Curiel TJ, Hung MC, Rich- Frischmuth K, Tempfer C, Reinthaller A, Hefler L. The prognostic
mond A, Burow ME. Cytokine receptor CXCR4 mediates estrogen- value of plasma fibrinogen levels in patients with endometrial cancer: a
independent tumorigenesis, metastasis, and resistance to endocrine ther- multi-centre trial. Br J Cancer 102: 952–956, 2010.
apy in human breast cancer. Cancer Res 71: 603–613, 2011. 322. Selman M, Ruiz V, Cabrera S, Segura L, Ramirez R, Barrios R,
301. Ricciardelli C, Sakko AJ, Ween MP, Russell DL, Horsfall DJ. The Pardo A. TIMP-1, -2, -3, and -4 in idiopathic pulmonary fibrosis. A
biological role and regulation of versican levels in cancer. Cancer Metast prevailing nondegradative lung microenvironment? Am J Physiol Lung
Rev 28: 233–245, 2009. Cell Mol Physiol 279: L562–L574, 2000.
302. Richter CM, Godes M, Wagner C, Maser-Gluth C, Herzfeld S, Dorn 323. Sen K, Lindenmeyer MT, Gaspert A, Eichinger F, Neusser MA,
M, Priem F, Slowinski T, Bauer C, Schneider W, Neumayer HH, Kretzler M, Segerer S, Cohen CD. Periostin is induced in glomerular
Kurtz A, Hocher B. Chronic cyclooxygenase-2 inhibition does not alter injury and expressed de novo in interstitial renal fibrosis. Am J Pathol
blood pressure and kidney function in renovascular hypertensive rats. J 179: 1756 –1767, 2011.
Hypertens 22: 191–198, 2004. 324. Serini G, Bochaton-Piallat ML, Ropraz P, Geinoz A, Borsi L, Zardi
303. Roda O, Ortiz-Zapater E, Martinez-Bosch N, Gutierrez-Gallego R, L, Gabbiani G. The fibronectin domain ED-A is crucial for myofibro-
Vila-Perello M, Ampurdanes C, Gabius HJ, Andre S, Andreu D, blastic phenotype induction by transforming growth factor-beta1. J Cell
Real FX, Navarro P. Galectin-1 is a novel functional receptor for tissue Biol 142: 873–881, 1998.
plasminogen activator in pancreatic cancer. Gastroenterology 136: 325. Setoguchi T, Kikuchi H, Yamamoto M, Baba M, Ohta M, Kamiya K,
1379 –1390, 2009. Tanaka T, Baba S, Goto-Inoue N, Setou M, Sasaki T, Mori H,
304. Ropponen K, Tammi M, Parkkinen J, Eskelinen M, Tammi R, Sugimura H, Konno H. Microarray analysis identifies versican and CD9
Lipponen P, Agren U, Alhava E, Kosma VM. Tumor cell-associated as potent prognostic markers in gastric gastrointestinal stromal tumors.
hyaluronan as an unfavorable prognostic factor in colorectal cancer. Cancer Sci 102: 883–889, 2011.
Cancer Res 58: 342–347, 1998. 326. Shao ZM, Nguyen M, Barsky SH. Human breast carcinoma desmopla-
305. Roy M, Pear WS, Aster JC. The multifaceted role of Notch in cancer. sia is PDGF initiated. Oncogene 19: 4337–4345, 2000.
Curr Opin Genet Dev 17: 52–59, 2007. 327. Shariat SF, Kattan MW, Traxel E, Andrews B, Zhu K, Wheeler TM,
306. Ruan K, Bao S, Ouyang G. The multifaceted role of periostin in Slawin KM. Association of pre- and postoperative plasma levels of
tumorigenesis. Cell Mol Life Sci 66: 2219 –2230, 2009. transforming growth factor beta(1) and interleukin 6 and its soluble
307. Ruoslahti E, Vaheri A, Kuusela P, Linder E. Fibroblast surface receptor with prostate cancer progression. Clin Cancer Res 10: 1992–
antigen: a new serum protein. Biochim Biophys Acta 322: 352–358, 1973. 1999, 2004.
308. Ruppert C, Markart P, Wygrecka M, Preissner KT, Gunther A. Role 328. Shaw A, Gipp J, Bushman W. The Sonic Hedgehog pathway stimulates
of coagulation and fibrinolysis in lung and renal fibrosis. Hamostaseolo- prostate tumor growth by paracrine signaling and recapitulates embry-
gie 28: 30 –36, 2008. onic gene expression in tumor myofibroblasts. Oncogene 28: 4480 –4490,
309. Said N, Frierson HF, Sanchez-Carbayo M, Brekken RA, Theodor- 2009.
escu D. Loss of SPARC in bladder cancer enhances carcinogenesis and 329. Shen J, Gao J, Chen C, Lu H, Hu G, Shen J, Zhu S, Wu M, Wang
progression. J Clin Invest 123: 751–766, 2013. X, Qian L, Yu Y, Han W, Wan R, Wang X. Antifibrotic role of
310. Said N, Sanchez-Carbayo M, Smith SC, Theodorescu D. RhoGDI2 chemokine CXCL9 in experimental chronic pancreatitis induced by
suppresses lung metastasis in mice by reducing tumor versican expres- trinitrobenzene sulfonic acid in rats. Cytokine 64: 382–394, 2013.
sion and macrophage infiltration. J Clin Invest 122: 1503–1518, 2012. 330. Shimada M, Andoh A, Hata K, Tasaki K, Araki Y, Fujiyama Y,
311. Saito-Fujita T, Iwakawa M, Nakamura E, Nakawatari M, Fujita H, Bamba T. IL-6 secretion by human pancreatic periacinar myofibroblasts
Moritake T, Imai T. Attenuated lung fibrosis in interleukin 6 knock-out in response to inflammatory mediators. J Immunol 168: 861–868, 2002.
mice after C-ion irradiation to lung. J Radiat Res (Tokyo) 52: 270 –277, 331. Sidhu SS, Yuan S, Innes AL, Kerr S, Woodruff PG, Hou L, Muller
2011. SJ, Fahy JV. Roles of epithelial cell-derived periostin in TGF-beta
312. Sakuma K, Aoki M, Kannagi R. Transcription factors c-Myc and activation, collagen production, and collagen gel elasticity in asthma.
CDX2 mediate E-selectin ligand expression in colon cancer cells under- Proc Natl Acad Sci USA 107: 14170 –14175, 2010.
going EGF/bFGF-induced epithelial-mesenchymal transition. Proc Natl 332. Sime PJ. The antifibrogenic potential of PPARgamma ligands in pul-
Acad Sci USA 109: 7776 –7781, 2012. monary fibrosis. J Investig Med 56: 534 –538, 2008.
313. Salah Z, Uziely B, Jaber M, Maoz M, Cohen I, Hamburger T, Maly 333. Sironen RK, Tammi M, Tammi R, Auvinen PK, Anttila M, Kosma
B, Peretz T, Bar-Shavit R. Regulation of human protease-activated VM. Hyaluronan in human malignancies. Exp Cell Res 317: 383–391,
receptor 1 (hPar1) gene expression in breast cancer by estrogen. FASEB 2011.
J 26: 2031–2042, 2012. 334. Sisson TH, Hattori N, Xu Y, Simon RH. Treatment of bleomycin-
314. Sasaki T, Nakamura T, Rebhun RB, Cheng H, Hale KS, Tsan RZ, induced pulmonary fibrosis by transfer of urokinase-type plasminogen
Fidler IJ, Langley RR. Modification of the primary tumor microenvi- activator genes. Hum Gene Ther 10: 2315–2323, 1999.
ronment by transforming growth factor alpha-epidermal growth factor 335. Smith HW, Marshall CJ. Regulation of cell signalling by uPAR. Nat
receptor signaling promotes metastasis in an orthotopic colon cancer Rev Mol Cell Biol 11: 23–36, 2010.
model. Am J Pathol 173: 205–216, 2008. 336. Sobolik T, Su YJ, Wells S, Ayers GD, Cook RS, Richmond A. CXCR4
315. Sasser AK, Sullivan NJ, Studebaker AW, Hendey LF, Axel AE, Hall drives the metastatic phenotype in breast cancer through induction of
BM. Interleukin-6 is a potent growth factor for ER-alpha-positive human CXCR2, and activation of MEK and PI3K pathways. Mol Biol Cell
breast cancer. FASEB J 21: 3763–3770, 2007. [Epub ahead of print].

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


Downloaded from journals.physiology.org/journal/physiolgenomics (180.242.184.036) on February 16, 2024.
Review
THE WHFC TRIAD 243
337. Sorensen I, Susnik N, Inhester T, Degen JL, Melk A, Haller H, 359. Theunissen JW, de Sauvage FJ. Paracrine hedgehog signaling in
Schmitt R. Fibrinogen, acting as a mitogen for tubulointerstitial fibro- cancer. Cancer Res 69: 6007–6010, 2009.
blasts, promotes renal fibrosis. Kidney Int 80: 1035–1044, 2011. 360. Tian Z, Chen Y, Gao B. Natural killer cells in liver disease. Hepatology
338. Spencer VA, Xu R, Bissell MJ. Gene expression in the third dimension: 57: 1654 –1662, 2013.
the ECM-nucleus connection. J Mamm Gland Biol Neoplas 15: 65–71, 361. Tiggelman AM, Boers W, Linthorst C, Brand HS, Sala M, Chamu-
2010. leau RA. Interleukin-6 production by human liver (myo)fibroblasts in
339. Spyrou GE, Naylor IL. The effect of basic fibroblast growth factor on culture. Evidence for a regulatory role of LPS, IL-1 beta and TNF alpha.
scarring. Br J Plast Surg 55: 275–282, 2002. J Hepatol 23: 295–306, 1995.
340. Sterclova M, Vasakova M, Pavlicek J, Metlicka M, Krasna E, Striz 362. Tiwari N, Gheldof A, Tatari M, Christofori G. EMT as the ultimate
I. Chemokine receptors in a regulation of interstitial lung fibrosis and survival mechanism of cancer cells. Sem Cancer Biol 22: 194 –207, 2012.
inflammation. Exp Lung Res 35: 514 –523, 2009. 363. Tolino MA, Block ER, Klarlund JK. Brief treatment with heparin-
341. Stern R. Hyaluronidases in cancer biology. Sem Cancer Biol 18: 275– binding EGF-like growth factor, but not with EGF, is sufficient to
280, 2008. accelerate epithelial wound healing. Biochim Biophys Acta 1810: 875–
342. Stern R, Asari AA, Sugahara KN. Hyaluronan fragments: an informa- 878, 2011.
tion-rich system. Eur J Cell Biol 85: 699 –715, 2006. 364. Tomasek JJ, Gabbiani G, Hinz B, Chaponnier C, Brown RA. Myo-
343. Strieter RM, Keeley EC, Burdick MD, Mehrad B. The role of fibroblasts and mechano-regulation of connective tissue remodelling. Nat
circulating mesenchymal progenitor cells, fibrocytes, in promoting pul- Rev Mol Cell Biol 3: 349 –363, 2002.
monary fibrosis. Trans Am Clin Climatol Assoc 120: 49 –59, 2009. 365. Trombetta-Esilva J, Bradshaw AD. The function of SPARC as a
344. Strutz F, Zeisberg M, Hemmerlein B, Sattler B, Hummel K, Becker mediator of fibrosis. Open Rheumatol J 6: 146 –155, 2012.
V, Muller GA. Basic fibroblast growth factor expression is increased in 366. Trotta R, Dal Col J, Yu J, Ciarlariello D, Thomas B, Zhang X, Allard
human renal fibrogenesis and may mediate autocrine fibroblast prolifer- J 2nd, Wei M, Mao H, Byrd JC, Perrotti D, Caligiuri MA. TGF-beta
ation. Kidney Int 57: 1521–1538, 2000. utilizes SMAD3 to inhibit CD16-mediated IFN-gamma production and
345. Strutz F, Zeisberg M, Renziehausen A, Raschke B, Becker V, van antibody-dependent cellular cytotoxicity in human NK cells. J Immunol
Kooten C, Muller G. TGF-beta 1 induces proliferation in human renal 181: 3784 –3792, 2008.
fibroblasts via induction of basic fibroblast growth factor (FGF-2). 367. Tsunoda S, Sakurai H, Saito Y, Ueno Y, Koizumi K, Saiki I. Massive
Kidney Int 59: 579 –592, 2001. T-lymphocyte infiltration into the host stroma is essential for fibroblast
346. Strutz F, Zeisberg M, Ziyadeh FN, Yang CQ, Kalluri R, Muller GA, growth factor-2-promoted growth and metastasis of mammary tumors via
Neilson EG. Role of basic fibroblast growth factor-2 in epithelial- neovascular stability. Am J Pathol 174: 671–683, 2009.
mesenchymal transformation. Kidney Int 61: 1714 –1728, 2002. 368. Turner N, Grose R. Fibroblast growth factor signalling: from develop-
347. Su X, Matthay MA. Role of protease activated receptor 2 in experimen- ment to cancer. Nat Rev Cancer 10: 116 –129, 2010.
tal acute lung injury and lung fibrosis. Anatom Rec (Hoboken) 292: 369. Utispan K, Thuwajit P, Abiko Y, Charngkaew K, Paupairoj A,
580 –586, 2009. Chau-in S, Thuwajit C. Gene expression profiling of cholangiocarci-
348. Sullivan NJ, Sasser AK, Axel AE, Vesuna F, Raman V, Ramirez N, noma-derived fibroblast reveals alterations related to tumor progression
Oberyszyn TM, Hall BM. Interleukin-6 induces an epithelial-mesen- and indicates periostin as a poor prognostic marker. Mol Cancer 9: 13,
chymal transition phenotype in human breast cancer cells. Oncogene 28: 2010.
2940 –2947, 2009. 370. Valdivia-Silva J, Franco-Barraza J, Cukierman E, Garcia-Zepeda E.
349. Sun X, Fa P, Cui Z, Xia Y, Sun L, Li Z, Tang A, Gui Y, Cai Z. The Novel insights into the role of inflammation in promoting breast cancer
EDA-containing cellular fibronectin induces epithelial-mesenchymal development. In: Breast Cancer - Focusing Tumor Microenvironment,
transition in lung cancer cells through integrin ␣9␤1-mediated activation Stem Cells and Metastasis, edited by Gunduz M, and Gunduz E. InTech,
of PI3-K/Akt and Erk1/2. Carcinogenesis 35: 184 –191, 2014. 2011, p. 129 –164.
350. Suzuki K, Wang R, Kubota H, Shibuya H, Saegusa J, Sato T. Kinetics 371. Valdivia-Silva JE, Franco-Barraza J, Silva AL, Pont GD, Soldevila
of biglycan, decorin and thrombospondin-1 in mercuric chloride-induced G, Meza I, Garcia-Zepeda EA. Effect of pro-inflammatory cytokine
renal tubulointerstitial fibrosis. Exp Mol Pathol 79: 68 –73, 2005. stimulation on human breast cancer: implications of chemokine receptor
351. Svensson M, Yadav M, Holmqvist B, Lutay N, Svanborg C, Godaly expression in cancer metastasis. Cancer Lett 283: 176 –185, 2009.
G. Acute pyelonephritis and renal scarring are caused by dysfunctional 372. Vandoros GP, Konstantinopoulos PA, Sotiropoulou-Bonikou G,
innate immunity in mCxcr2 heterozygous mice. Kidney Int 80: 1064 – Kominea A, Papachristou GI, Karamouzis MV, Gkermpesi M, Vara-
1072, 2011. kis I, Papavassiliou AG. PPAR-gamma is expressed and NF-kB path-
352. Tacke F, Zimmermann HW, Berres ML, Trautwein C, Wasmuth way is activated and correlates positively with COX-2 expression in
HE. Serum chemokine receptor CXCR3 ligands are associated with stromal myofibroblasts surrounding colon adenocarcinomas. J Cancer
progression, organ dysfunction and complications of chronic liver dis- Res Clin Oncol 132: 76 –84, 2006.
eases. Liver Int 31: 840 –849, 2011. 373. Venkatesan N, Tsuchiya K, Kolb M, Farkas L, Bourhim M, Ouzzine
353. Takahashi Y, Kuwabara H, Yoneda M, Isogai Z, Tanigawa N, M, Ludwig MS. Glycosyltransferases and glycosaminoglycans in bleo-
Shibayama Y. Versican G1 and G3 domains are upregulated and latent mycin and transforming growth factor-␤1-induced pulmonary fibrosis.
transforming growth factor-beta binding protein-4 is downregulated in Am J Resp Cell Mol Biol [Epub ahead of print].
breast cancer stroma. Breast Cancer 19: 46 –53, 2012. 374. Vial C, Gutierrez J, Santander C, Cabrera D, Brandan E. Decorin
354. Takeuchi K, Tanaka A, Kato S, Amagase K, Satoh H. Roles of COX interacts with connective tissue growth factor (CTGF)/CCN2 by LRR12
inhibition in pathogenesis of NSAID-induced small intestinal damage. inhibiting its biological activity. J Biol Chem 286: 24242–24252, 2011.
Clinica Chimica Acta 411: 459 –466, 2010. 375. Wallach-Dayan SB, Golan-Gerstl R, Breuer R. Evasion of myofibro-
355. Tammi RH, Kultti A, Kosma VM, Pirinen R, Auvinen P, Tammi MI. blasts from immune surveillance: a mechanism for tissue fibrosis. Proc
Hyaluronan in human tumors: pathobiological and prognostic messages Natl Acad Sci USA 104: 20460 –20465, 2007.
from cell-associated and stromal hyaluronan. Sem Cancer Biol 18: 376. Walter K, Omura N, Hong SM, Griffith M, Vincent A, Borges M,
288 –295, 2008. Goggins M. Overexpression of smoothened activates the sonic hedgehog
356. Tang L, Liu K, Wang J, Wang C, Zhao P, Liu J. High preoperative signaling pathway in pancreatic cancer-associated fibroblasts. Clin Can-
plasma fibrinogen levels are associated with distant metastases and cer Res 16: 1781–1789, 2010.
impaired prognosis after curative resection in patients with colorectal 377. Wang B, Li GX, Zhang SG, Wang Q, Wen YG, Tang HM, Zhou CZ,
cancer. J Surg Oncol 102: 428 –432, 2010. Xing AY, Fan JW, Yan DW, Qiu GQ, Yu ZH, Peng ZH. Biglycan
357. Tangkijvanich P, Kongtawelert P, Pothacharoen P, Mahachai V, expression correlates with aggressiveness and poor prognosis of gastric
Suwangool P, Poovorawan Y. Serum hyaluronan: a marker of liver cancer. Exp Biol Med (Maywood) 236: 1247–1253, 2011.
fibrosis in patients with chronic liver disease. Asian Pac J Allergy 378. Wang H, Haeger SM, Kloxin AM, Leinwand LA, Anseth KS. Redi-
Immunol 21: 115–120, 2003. recting valvular myofibroblasts into dormant fibroblasts through light-
358. Tani K, Yasuoka S, Ogushi F, Asada K, Fujisawa K, Ozaki T, Sano mediated reduction in substrate modulus. PLoS One 7: e39969, 2012.
N, Ogura T. Thrombin enhances lung fibroblast proliferation in bleo- 379. Wang K, Grivennikov SI, Karin M. Implications of anti-cytokine
mycin-induced pulmonary fibrosis. Am J Respir Cell Mol Biol 5: 34 –40, therapy in colorectal cancer and autoimmune diseases. Ann Rheum Dis
1991. 72: ii100 –ii103, 2013.

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


Downloaded from journals.physiology.org/journal/physiolgenomics (180.242.184.036) on February 16, 2024.
Review
244 THE WHFC TRIAD

380. Wang W, Liu F, Chen N. Peroxisome proliferator-activated receptor- 400. Yamada KM, Olden K, Pastan I. Transformation-sensitive cell surface
gamma (PPAR-gamma) agonists attenuate the profibrotic response in- protein: isolation, characterization, and role in cellular morphology and
duced by TGF-beta1 in renal interstitial fibroblasts. Med Inf (Lond) 2007: adhesion. Ann NY Acad Sci 312: 256 –277, 1978.
62641, 2007. 401. Yamaguchi Y, Ono J, Masuoka M, Ohta S, Izuhara K, Ikezawa Z,
381. Webber J, Jenkins RH, Meran S, Phillips A, Steadman R. Modulation Aihara M, Takahashi K. Serum periostin levels are correlated with
of TGFbeta1-dependent myofibroblast differentiation by hyaluronan. Am progressive skin sclerosis in patients with systemic sclerosis. Brit J
J Pathol 175: 148 –160, 2009. Dermat 168: 717–725, 2013.
382. Ween MP, Oehler MK, Ricciardelli C. Role of versican, hyaluronan 402. Yan W, Wang P, Zhao CX, Tang J, Xiao X, Wang DW. Decorin gene
and CD44 in ovarian cancer metastasis. Int J Mol Sci 12: 1009 –1029, delivery inhibits cardiac fibrosis in spontaneously hypertensive rats by
2011. modulation of transforming growth factor-beta/Smad and p38 mitogen-
383. Wells RG, Discher DE. Matrix elasticity, cytoskeletal tension, and activated protein kinase signaling pathways. Hum Gene Ther 20: 1190 –
TGF-beta: the insoluble and soluble meet. Sci Signal 1: pe13, 2008. 1200, 2009.
384. White ES, Muro AF. Fibronectin splice variants: understanding their 403. Yang F, Strand DW, Rowley DR. Fibroblast growth factor-2 mediates
multiple roles in health and disease using engineered mouse models. transforming growth factor-beta action in prostate cancer reactive stroma.
IUBMB Life 63: 538 –546, 2011. Oncogene 27: 450 –459, 2008.
385. Wolf K, Mazo I, Leung H, Engelke K, von Andrian UH, Deryugina 404. Yang IV. Epigenomics of idiopathic pulmonary fibrosis. Epigenomics 4:
EI, Strongin AY, Brocker EB, Friedl P. Compensation mechanism in 195–203, 2012.
tumor cell migration: mesenchymal-amoeboid transition after blocking 405. Yang J, Shultz RW, Mars WM, Wegner RE, Li Y, Dai C, Nejak K,
of pericellular proteolysis. J Cell Biol 160: 267–277, 2003. Liu Y. Disruption of tissue-type plasminogen activator gene in mice
386. Wong GS, Lee JS, Park YY, Klein-Szanto AJ, Waldron TJ, Cuki- reduces renal interstitial fibrosis in obstructive nephropathy. J Clin Invest
erman E, Herlyn M, Gimotty P, Nakagawa H, Rustgi A K. Periostin 110: 1525–1538, 2002.
cooperates with mutant p53 to mediate invasion through the induction of 406. Yang X, Hou J, Han Z, Wang Y, Hao C, Wei L, Shi Y. One cell,
STAT1 signaling in the esophageal tumor microenvironment. Oncogen- multiple roles: contribution of mesenchymal stem cells to tumor devel-
esis 5: e59, 2013. opment in tumor microenvironment. Cell Biosci 3: 5, 2013.
387. Wu J, Wu X, Liang W, Chen C, Zheng L, An H. Clinicopathological 407. Yeung TL, Leung CS, Wong KK, Samimi G, Thompson MS, Liu J,
and prognostic significance of chemokine receptor CXCR4 overexpres- Zaid TM, Ghosh S, Birrer MJ, Mok SC. TGF-beta modulates ovarian
sion in patients with esophageal cancer: a meta-analysis. Tumour Biol cancer invasion by upregulating CAF-derived versican in the tumor
[Epub ahead of print]. microenvironment. Cancer Res 73: 5016 –5028, 2013.
388. Wu Q, Hou X, Xia J, Qian X, Miele L, Sarkar FH, Wang Z. Emerging 408. Yoshioka N, Fuji S, Shimakage M, Kodama K, Hakura A, Yutsudo
roles of PDGF-D in EMT progression during tumorigenesis. Cancer M, Inoue H, Nojima H. Suppression of anchorage-independent growth
Treat Rev 39: 640 –646, 2013. of human cancer cell lines by the TRIF52/Periostin/OSF-2 gene. Exp Cell
389. Wynn TA. Cellular and molecular mechanisms of fibrosis. J Pathol 214: Res 279: 91–99, 2002.
199 –210, 2008. 409. Yu FS, Yin J, Xu K, Huang J. Growth factors and corneal epithelial
390. Wynn TA, Ramalingam TR. Mechanisms of fibrosis: therapeutic trans- wound healing. Brain Res Bull 81: 229 –235, 2010.
lation for fibrotic disease. Nat Med 18: 1028 –1040, 2012. 410. Yu H, Mouw JK, Weaver VM. Forcing form and function: biomechani-
391. Xiang L, Xie G, Ou J, Wei X, Pan F, Liang H. The extra domain A of cal regulation of tumor evolution. Trends Cell Biol 21: 47–56, 2011.
fibronectin increases VEGF-C expression in colorectal carcinoma involv- 411. Zaja-Milatovic S, Richmond A. CXC chemokines and their receptors:
ing the PI3K/AKT signaling pathway. PLoS One 7: e35378, 2012. a case for a significant biological role in cutaneous wound healing. Histol
392. Xiao Q, Ge G. Lysyl oxidase, extracellular matrix remodeling and Histopathol 23: 1399 –1407, 2008.
cancer metastasis. Cancer Microenvir 5: 261–273, 2012. 412. Zeisberg EM, Potenta S, Xie L, Zeisberg M, Kalluri R. Discovery of
393. Xu CZ, Wang PH, Yan XJ, Wang T, Chen D, Zhang ZJ, Shi RJ. endothelial to mesenchymal transition as a source for carcinoma-associ-
Expression of CXCR4 is associated with progression and invasion in ated fibroblasts. Cancer Res 67: 10123–10128, 2007.
patients with nasal-surface basal cell carcinoma. ORL J Otorhinolaryngol 413. Zeisberg EM, Potenta SE, Sugimoto H, Zeisberg M, Kalluri R.
Relat Spec 75: 332–341, 2014. Fibroblasts in kidney fibrosis emerge via endothelial-to-mesenchymal
394. Xu S, Cao X. Interleukin-17 and its expanding biological functions. Cell transition. J Am Soc Nephrol 19: 2282–2287, 2008.
Mol Immunol 7: 164 –174, 2010. 414. Zeremski M, Petrovic LM, Chiriboga L, Brown QB, Yee HT,
395. Xu TP, Shen H, Liu LX, Shu YQ. The impact of chemokine receptor Kinkhabwala M, Jacobson IM, Dimova R, Markatou M, Talal AH.
CXCR4 on breast cancer prognosis: a meta-analysis. Cancer Epidemiol Intrahepatic levels of CXCR3-associated chemokines correlate with liver
37: 725–731, 2013. inflammation and fibrosis in chronic hepatitis C. Hepatology 48: 1440 –
396. Xu X, Zhou Y, Xie C, Wei SM, Gan H, He S, Wang F, Xu L, Lu J, 1450, 2008.
Dai W, He L, Chen P, Wang X, Guo C. Genome-wide screening 415. Zhu YH, Yang F, Zhang SS, Zeng TT, Xie X, Guan XY. High
reveals an EMT molecular network mediated by Sonic hedgehog-Gli1 expression of biglycan is associated with poor prognosis in patients with
signaling in pancreatic cancer cells. PLoS One 7: e43119, 2012. esophageal squamous cell carcinoma. Int J Clin Exp Pathol 6: 2497–
397. Xu X, Zhu F, Zhang M, Zeng D, Luo D, Liu G, Cui W, Wang S, Guo 2505, 2013.
W, Xing W, Liang H, Li L, Fu X, Jiang J, Huang H. Stromal 416. Zigler M, Kamiya T, Brantley EC, Villares GJ, Bar-Eli M. PAR-1
cell-derived factor-1 enhances wound healing through recruiting bone and thrombin: the ties that bind the microenvironment to melanoma
marrow-derived mesenchymal stem cells to the wound area and promot- metastasis. Cancer Res 71: 6561–6566, 2011.
ing neovascularization. Cells Tiss Organs 103–113, 2012. 417. Zimmermann HW, Seidler S, Gassler N, Nattermann J, Luedde T,
398. Xue W, Zender L, Miething C, Dickins RA, Hernando E, Krizha- Trautwein C, Tacke F. Interleukin-8 is activated in patients with
novsky V, Cordon-Cardo C, Lowe SW. Senescence and tumour clear- chronic liver diseases and associated with hepatic macrophage accumu-
ance is triggered by p53 restoration in murine liver carcinomas. Nature lation in human liver fibrosis. PLoS One 6: e21381, 2011.
445: 656 –660, 2007. 418. Zoltan-Jones A, Huang L, Ghatak S, Toole BP. Elevated hyaluronan
399. Yamada KM, Cukierman E. Modeling tissue morphogenesis and can- production induces mesenchymal and transformed properties in epithelial
cer in 3D. Cell 130: 601–610, 2007. cells. J Biol Chem 278: 45801–45810, 2003.

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


Downloaded from journals.physiology.org/journal/physiolgenomics (180.242.184.036) on February 16, 2024.

You might also like