Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/325783543

Cancer: the Dark Side of Wound Healing

Article  in  FEBS Journal · June 2018


DOI: 10.1111/febs.14586

CITATIONS READS

10 311

3 authors:

Gopinath Meenakshisundaram Shan Quah

38 PUBLICATIONS   409 CITATIONS   
University of Oxford
16 PUBLICATIONS   124 CITATIONS   
SEE PROFILE
SEE PROFILE

Prabha Sampath
Institute of Medical Biology
50 PUBLICATIONS   1,105 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Glioma stem cells View project

MicroRNAs in Atopic Dermatitis View project

All content following this page was uploaded by Prabha Sampath on 14 July 2018.

The user has requested enhancement of the downloaded file.


STATE-OF-THE-ART REVIEW

Cancer: the dark side of wound healing


Gopinath M. Sundaram1, Shan Quah1 and Prabha Sampath1,2,3
1 Institute of Medical Biology, Agency for Science Technology & Research (A*STAR), Singapore City, Singapore
2 Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
3 Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore City, Singapore

Keywords Complex multicellular organisms have evolved sophisticated mechanisms to


angiogenesis; chronic wounds; growth rapidly resolve epithelial injuries. Epithelial integrity is critical to maintain-
factors; invasion; metastasis; squamous cell
ing internal homeostasis. An epithelial breach represents the potential for
carcinoma; wound healing
pathogen ingress and fluid loss, both of which may have severe conse-
Correspondence quences if not limited. The mammalian wound healing response involves a
P. Sampath, Institute of Medical Biology, finely tuned, self-limiting series of cellular and molecular events orches-
Agency for Science Technology & Research trated by the transient activation of specific signalling pathways. Accurate
(A*STAR), 8A Biomedical Grove, #05-41 regulation of these events is essential; failure to initiate key steps at the
Immunos, Singapore City, Singapore right time delays healing and leads to chronic wounds, while aberrant initi-
138648
ation of wound healing processes may produce cell behaviours that pro-
Fax: +65 6464 2049
mote cancer progression. In this review, we discuss how wound healing
Tel: +65 6407 0171
E-mail: prabha.sampath@imb.a-star.edu.sg pathways co-opted in cancer lose their stringent regulation and become
compromised in their reversibility. We hypothesize on how the comman-
(Received 27 February 2018, revised 17 deering of wound healing ‘master regulators’ is involved in this process,
May 2018, accepted 13 June 2018) and also highlight the implications of these findings in the treatment of
both chronic wounds and cancer.
doi:10.1111/febs.14586

Introduction
Cancer is a leading cause of death worldwide, with 8.8 One way in which this might be achieved is through
million attributable fatalities in 2015 alone (Source: the study of epithelial wound healing. The physiology
World Health Organization). Over 90% of cancers underlying wound healing is highly similar to that
originate in epithelial tissues, where the primary involved in cancer progression, especially in the case
tumour itself is rarely a significant cause of mortality. of epithelial cancers. While this idea is not new, the
Instead, the process of metastasis, whereby cells leave hypothesis of ‘cancer as an overhealing wound’ was
the primary tumour and colonize distant organs, is brought forward almost three decades ago, it has been
responsible for most cancer-related deaths. Under- gaining traction in recent years [1–3]. Many signalling
standing tumorigenesis and the metastatic process is pathways and molecular mechanisms which are critical
essential to the development of anticancer therapies. for wound healing have been implicated either in

Abbreviations
BCC, basal cell carcinoma; BMP, bone morphogenetic protein; CIS, carcinoma in situ; CSC, cancer stem cell; ECM, extracellular matrix;
EGF, epidermal growth factor; EMT, epithelial–mesenchymal transition; FAK, focal adhesion kinase; FGF, fibroblast growth factor; FSTL1,
follistatin like-1; HFSC, hair follicular stem cell; HGF, hepatocyte growth factor; HIF, hypoxia inducible factor; IFESC, interfollicular epidermal
stem cell; IFNc, interferon c; IL, interleukin; lncRNA, long noncoding RNA; miRNA, microRNA; MMP, matrix metalloproteinase; NET,
neutrophil extracellular traps; PDGF, platelet-derived growth factor; RTK, receptor tyrosine kinase; SCC, squamous cell carcinoma; SMAD,
mothers against decapentaplegic homologue; TAM, tumour-associated macrophage; TAN, tumour-associated neutrophil; TERT, telomerase
reverse transcriptase; TGF, transforming growth factor; TIMP, tissue inhibitor of metalloproteinase; TNF, tumour necrosis factor;
VEGF, vascular endothelial growth factor; ZEB, zing finger E-box binding homeobox.

The FEBS Journal (2018) ª 2018 Federation of European Biochemical Societies 1


Parallels between wound healing and cancer G. M. Sundaram et al.

cancer cell proliferation or in metastasis [3,4]. There- markers associated with hyperproliferation and migration
fore, elucidating the link between wound healing and as early as 6 h postinjury [14]. The time taken to achieve
metastatic cancer progression may aid in the formula- wound closure depends on the severity of the injury.
tion of better strategies to combat cancer. Superficial wounds confined to epidermal tissue heal
within days without significant scarring. Full thickness
wounds involving dermal or subcutaneous tissue take
Wound healing
longer to heal and are often associated with significant
The epithelium forms a protective barrier around the scarring in adult skin [15]. While restoration of barrier
organism, guarding it from external insults. The integ- function through the formation of neoepidermis occurs
rity of epithelial tissues, such as skin, is critical to quickly, reconstitution of dermal tissue may take several
maintaining a sterile internal environment. Multicellu- months [16]. In this remodelling phase, dermal fibrob-
lar organisms have therefore evolved sophisticated lasts/myofibroblasts actively reshape the dermal matrix
means by which epithelial wounds may be rapidly by secreting collagen fibres and metalloproteinases to
repaired, incorporating highly orchestrated pro- restore it to pre-injury conditions.
grammes involving multiple cell and tissue types [5]. Cell behaviours involved in re-epithelialization are
Wound healing occurs at the cellular, tissue and very similar to those that occur during cancer initia-
organismal levels. As organismal complexity increases, tion and metastasis. One critical distinction between
there is a correlated increase in the number of compo- wound repair and cancer progression is that the
nents involved in the wound healing process [6]. This promigratory, hyperproliferative behaviour demon-
is generally accompanied by a reduction in the organ- strated by keratinocytes during wound healing is self-
ism’s regenerative capabilities. Morphologically simple limiting; keratinocytes return to a quiescent state when
organisms such as planarians are capable of regenerat- epithelial repair is complete. In contrast, loss of con-
ing a large proportion of their bodies following ampu- trol over tumour cell proliferation and migration is a
tation [7]. Vertebrate regeneration potential is hallmark of malignant cancer.
restricted to limbs and specific organs in amphibians,
while mammals have difficulty repairing all but the
Molecular similarities between wound
smallest epithelial lesions without scarring [8,9].
healing and cancer
The seminal link between wound healing and cancer
Phases of wound healing
was first proposed nearly two centuries ago [17]. Over
The cutaneous wound healing response is classically the years, multiple lines of evidence have emerged in
subdivided into overlapping phases with distinct func- support of the notion that wound healing and tumori-
tional objectives and biochemical events [10,11]. Hae- genesis are two sides of the same coin. All the major
mostasis is initiated immediately following injury. A pathways implicated in wound healing are also active
fibrin clot is formed at the wound site to minimize in cancers [1,3]. Inflammation, an essential event in the
blood loss. This fibrin plug serves as a provisional wound healing response, is a key driver for cancer
matrix for the migration of various cell types such as [18,19]. As mentioned above, enhanced proliferation
keratinocytes, macrophages and neutrophils, and a and migration of cancer cells is similar to the beha-
trap for various growth factors in the wound milieu vioural profile of wounded epithelial cells [3,20]. Extra-
[12]. The inflammatory phase is initiated with the infil- cellular matrix remodelling during later stages of
tration of circulating neutrophils at the wound site. wound healing also has its parallels in tumours, where
Resident immune cells such as Langerhans, natural cancerous tissue is capable of altering the surrounding
killer (NK) and cd T cells are activated. These release matrix to support its own growth and invasion [21].
a plethora of cytokines and chemokines which serve as The wound microenvironment is conducive to cancer
alarm signals and actively recruit systemic immune progression, as gleaned from a study performed on mice
cells to combat infection at the wound site [13]. by Stuelten et al. [22]. In this report, mice with a local
Re-epithelialization represents the most critical phase subcutaneous wound created adjacent to the site where
of wound healing, whereby keratinocytes adjacent to the breast cancer cells were injected had higher incidences of
wound edge initiate hyperproliferation and migration on tumours and developed larger tumours relative to mice
the wound bed to repair the wound. These cell behaviours with distally created wounds, implying that local injury
necessitate reciprocal interaction between keratinocytes, response may play a role in tumorigenesis. In a separate
dermal cells such as fibroblasts and immune cells. Re- study, biopsies performed on mouse mammary tumours
epithelialization is initiated rapidly; keratinocytes express induced significant lung metastatic colonization. It was

2 The FEBS Journal (2018) ª 2018 Federation of European Biochemical Societies


G. M. Sundaram et al. Parallels between wound healing and cancer

proposed that inflammation associated with the local epidermal growth factor (EGF), fibroblast growth fac-
wound healing response at biopsy sites contributed to tor (FGF), transforming growth factor-b families
metastasis [23]. This effect was abrogated in mice lack- (TGF-b) and hepatocyte growth factor (HGF/c-MET).
ing the proinflammatory cytokine IL-6. Injury-induced
tumour growth also occurs in nonmammalian verte-
Epidermal growth factor family
brates. In a RasG12V -driven melanoma model of zebra-
fish, repeated wounding led to increased incidence of The epidermal growth factor (EGF) family is one of
melanoma formation compared to unwounded controls. the best-characterized growth factor signalling path-
This process was mediated by inflammatory neutrophils ways involved in keratinocyte proliferation and migra-
[24,25]. tion [35]. This family of growth factors includes EGF,
A relationship between wound healing and cancer heparin binding EGF (HB-EGF), transforming growth
progression may also be inferred from studies on factor-a (TGF-a), heregulin and amphiregulin [36] that
chronic wounds in diabetics. In 2015, diabetes afflicted work in both autocrine and paracrine manners. EGFR
400 million adults worldwide. About 15% of diabetic (otherwise known as ErbB11 or HER1) is a 170 kDa
patients develop nonhealing foot ulcers [26,27]. Met- transmembrane glycoprotein which belongs to the
formin, an FDA-approved drug widely used in the receptor associated tyrosine kinase (RTK) family [36].
treatment of diabetes mellitus, significantly improves EGF can also induce heterodimerization across the
the outcome of these wounds. Metformin also has a RTK family and activation of the mitogen-associated
negative effect on cancer progression [28,29]. However, protein kinase (MAPK) pathway, which promotes cell
the relationship between chronic diabetic ulcers and proliferation and migration. EGFR is predominantly
cancer is far from straightforward. Platelet derived expressed in the basal layer of the human epidermis.
growth factor (PDGF-BB), marketed as Becaplermin, Its expression is upregulated in wound edge ker-
is clinically approved for the treatment of chronic atinocytes after injury [37].
ulcers. Diabetic patients treated with Becaplermin are
at increased risk of developing cancer, implying that
The EGF pathway in wound healing,
certain drugs which accelerate wound healing may
tumour progression and metastasis
inadvertently induce cancer-promoting pathways [30].
Although wound healing and cancer are highly compli- Cell proliferation and migration, the two vital processes
cated processes, both result from coordinated regulation required for wound healing and cancer progression, are
over two relatively simple cell behaviours – migration activated by the EGFR pathway. This pathway is
and proliferation. These behaviours are shaped by inter- essential for wound healing. EGFR!/! mouse skin
actions between various cell types, including epithelial, grafted onto nude mice displayed wound healing defects
endothelial, mesenchymal and immune cells, through after full thickness injury [38]. EGFR ligands such as
growth factor/cytokine signalling networks. An example TGF-a, HB-EGF and amphiregulin are upregulated in
of such coordination may be seen during re-epithelializa- keratinocytes upon wounding [39,40]. In chronic ulcers,
tion. Keratinocyte migration over the wound bed is EGFR signalling is reduced due to a number of factors
guided by intracellular mechanisms such as cytoskeletal including cytoplasmic EGFR retention, as well as
rearrangement, as well as extracellular events such as decreased EGFR levels resulting from the protease-rich
cell–cell and cell–matrix interactions, basement mem- milieu in the wound environment [41] (Fig. 1).
brane remodelling and proteinase-mediated matrix alter- In cancer, one of the initial steps in metastasis is the
ation. Many of these events also operate during cancer dissociation of cells from the primary tumour and
progression [31–33]. A number of promigratory proteins stromal invasion. This process is accompanied by
required for keratinocyte migration are also abundantly hemidesmosome disassembly, which results from the
expressed in epithelial cancers [34]. Here we provide a dis- disruption of a6b4 integrins in response to EGFR sig-
course into the commonalities between wound healing nalling [42]. Notably, this EGF-induced disassembly
and epithelial cancers, with a focus on invasive and meta- process, which occurs during invasion and metastasis
static squamous cell carcinomas (SCCs). of SCC cells in vitro and in vivo [43], also occurs dur-
ing normal epithelial cell migration. Thus, EGF-
mediated hemidesmosome disassembly represents a
Growth factor networks in wound
function present in healthy epithelia which has been
healing and cancer metastasis
co-opted by epithelial cancer cells for dissociation from
A large number of growth factors influence wound their site of origin. Nevertheless, cell dissociation from
healing and metastasis, including the members of the the primary tumour alone is insufficient for stromal

The FEBS Journal (2018) ª 2018 Federation of European Biochemical Societies 3


Parallels between wound healing and cancer G. M. Sundaram et al.

Fig. 1. Schematic representing common growth factor signalling pathways that regulate key cellular processes required for wound healing and
cancer. The effect of each growth factor on various physiological processes in a healing wound is listed above. The bottom panel elucidates the
role of growth factors in processes involved in cancer growth and metastasis. Note that TGF-b decreases the proliferation of epithelial cells in
both wound healing and cancer, while promotes keratinocyte migration in wound healing and tumour-promoting processes in cancer.

invasion. In order for metastasis to progress, cancer and head and neck SCCs (cSCC & HNSCC) [49,50].
cells must degrade the basement membrane delineating In cancer tissues, EGFR expression status is positively
the epithelial compartment from the stroma. Basement correlated with lymph node involvement, tumour grade
membrane degradation requires dissolution of the and stage [21,51,52]. Recently, EGF has been shown to
extracellular matrix (ECM), involving a remodelling induce EMT and confer stem cell-like properties to
process highly similar to that which occurs during oral cancer cells through the ‘Warburg effect’ [53].
wound healing. During re-epithelialization, wound EGFR activation may also activate multiple other
edge keratinocytes form an epithelial tongue that pathways such as PI3-AKT, JAK-STAT and nuclear
migrates across the wound bed while constantly modi- factor kappa B (NF-kB) [54]. Moderate success has
fying the wound ECM [33]. In both wound healing been attained using targeted therapies against EGFR
and stromal invasion, ECM remodelling results from in the treatment of metastatic disease in multiple can-
the activities of matrix metalloproteases (MMPs). cer types including HNSCC, oesophageal, gastric, col-
MMPs are transcriptional targets of the EGFR path- orectal and lung SCC [55–59].
way [44] which degrade ECM components. Tissue inhi-
bitors of MMPs (TIMPs) exert spatiotemporal control
TGF-b signalling pathway
over MMP activity, and affect the healing status of an
acute or chronic wound [33]. The MMP2 and MMP9 TGF-b possesses three isoforms, all of which stimulate
proteins are present in wound fluids but virtually the same intracellular signalling SMADs (SMAD2/3)
absent from normal skin [45]. Increased MMP9 while performing different functions in development
expression is associated with poor healing in chronic and tissue homeostasis [60]. In the canonical TGF-b
wounds, and correlates with poor overall survival sta- pathway, TGF-b ligands bind to TGF-b receptor I
tus in HNSCC [46,47]. Stromal invasion of cutaneous (TGFBRI, otherwise known as ALK-5) and cause it
SCC is higher in patients with increased MMP2 to heterodimerize with TGF-b receptor II (TGFBRII).
expression [48]. These studies indicate that while EGF- This stimulates phosphorylation of the receptor regu-
induced MMPs are a crucial requirement for wound lated SMAD2/3 (R-SMADs) complex. Phosphorylated
healing, uncontrolled expression of these proteases can SMAD2/3 binds to SMAD4 (co-SMAD) and enters
have pathological consequences including chronic the nucleus, where it transactivates transcriptional tar-
wounds and metastasis of SCC. gets. In cutaneous wound healing, TGF-b is released
EGFR amplification, overexpression and gain-of- initially by platelets trapped in the fibrin clot, and at
function mutations are highly prevalent in cutaneous later stages by keratinocytes [61–63].

4 The FEBS Journal (2018) ª 2018 Federation of European Biochemical Societies


G. M. Sundaram et al. Parallels between wound healing and cancer

TGF-b in wound healing and cancer wound healing and cancer cell invasion [79] (Fig. 1).
Downregulation of E-cadherin in epithelial cells, a
The role of TGF-b in wound healing is complex, and classical marker of EMT, also occurs during ker-
likely to vary temporally as well as by its distinct atinocyte migration in wound healing [80]. Transcrip-
effects on different cell types. TGF-b1 is secreted by tional targets of TGF-b1 are often involved in cell
macrophages, stromal fibroblasts and keratinocytes migration, invasion and EMT. TGF-b1 activates sev-
and its expression is stringently regulated during eral EMT-associated master transcription factors
wound healing [64,65]. Mice with a TGFBRII deletion which are Snail 1, Snail 2 (Slug), and zinc finger E-box
in dermal fibroblasts display severe defects in full binding homeobox (ZEB) 1, ZEB2 and TWIST [81].
thickness wound healing and delayed re-epithelializa- During EMT, TGF-b1-mediated Snail activation leads
tion [66]. However, epidermal overexpression of TGF- to direct suppression E-cadherin gene expression, via
b1 also delays full thickness wound healing due to HDAC recruitment to the E-cadherin promoter, or
excessive inflammation [67]. This is likely due to the through activating ZEB-1 [82,83].
differential ability of TGF-b to orchestrate conflicting
pathways in wound healing. It is well known that
TGF-b exerts a negative effect on keratinocyte prolif-
Fibroblast growth factor family in
eration, but promotes both in vitro and in vivo ker-
wound healing and cancer
atinocyte migration [68]. Several studies have reported
defective TGF-b signalling in chronic wounds due to The FGF family includes over 22 structurally related
lack of TGFBR1, TGFBR2 or TGF-b ligand expres- FGF ligands, most of which interact with four FGF
sion [69–71]. In such wounds, TGF-b treatment was receptors (FGFR1, FGFR2, FGFR3 and FGFR4).
met with limited success, perhaps due to the lack of FGFs function primarily as mitogens controlling pro-
TGFBR expression and the protease-rich environment liferation and differentiation of various tissues, and
[72]. also regulate wound healing and angiogenesis [84].
TGF-b has gained considerable attention in past Several FGF ligands such as FGF7, FGF10 and
years for its puzzling role in cancer progression and FGF22 are transcriptionally induced upon injury in
metastasis. Some authors argue that TGF-b1 and its mice, although only FGFR1 is upregulated in similar
receptors are downregulated in SCC [73,74], while conditions [85]. FGF2 stimulates keratinocyte migra-
others presenting evidence for aberrant TGF-b1 over- tion in vitro through Rac activation and lamellipodia
expression in head and neck tumours [75]. It is possi- formation, and its loss causes delayed wound healing
ble that TGF-b initially exerts growth suppression on in mice [86]. FGF7, otherwise known as keratinocyte
epithelial cells, while also promoting EMT at later growth factor, is critical to keratinocyte proliferation.
stages. In transgenic mice, induction of TGF-b1 over- Inhibiting FGF7 signalling in mice causes epidermal
expression at papilloma stage induced rapid metastasis, atrophy and substantial delays in re-epithelialization
while induction of TGF-b1 at earlier stages led to [87]. In addition to its effects on proliferation, FGF7
tumour suppression [76]. Similarly, mice expressing is a more potent stimulator of keratinocyte migration
dominant-negative TGFBRII (DbRII) in the epidermis than FGF2 [88].
exhibit faster tumour formation and malignant pro- Fibroblast growth factor pathways utilized in wound
gression, but reduced EMT. This suggests that TGF- healing also feature in cancer progression. FGFR1
b1 has distinct effects on keratinocyte proliferation expression correlates with the epithelial–mesenchymal
and metastasis, and that normal TGFBRII exerts a transition (EMT) status of cell lines in vitro. Treatment
tumour-suppressive effect in the epidermis. In support of high EMT cell lines with the FGFR inhibitor
of these findings, Han et al. showed increasing TGF- PD173074 induces mesenchymal–epithelial transition,
b1 expression in stage wise SCC progression from acti- suggesting a potent role of FGF signalling in cancer
nic keratosis (epidermal hyperplasia), carcinoma metastasis [89] (Fig. 1). FGFR1 was recently shown to
in situ (CIS) towards metastatic SCC, while TGFBRII be a part of a molecular signature network with b3-
showed an opposite trend [77]. Thus, TGF-b signalling integrin and focal adhesion kinase (FAK) which drives
is activated in a cancer stage specific manner. This EMT and metastasis of breast cancer [90]. Oncogenic
may explain why TGF-b and TGFBR were not identi- driver mutations and copy number changes in FGFR
fied as hot spots for SCC risk, unlike EGF/EGFR and ligands occur in a subset of HNSCC patients [91].
[78]. Aberrant FGF signalling may promote both angiogen-
The dissociation of cell–cell junctions to facilitate esis and the acquisition of chemoresistance against
keratinocyte migration is a prerequisite for both tyrosine kinase inhibitors [92].

The FEBS Journal (2018) ª 2018 Federation of European Biochemical Societies 5


Parallels between wound healing and cancer G. M. Sundaram et al.

Hepatocyte growth factor/c-MET keratinocytes. Both macrophages and neutrophils pre-


receptor signalling in wound healing dominate the tumour mass and play pivotal roles in
and cancer injury and cancer. Metastatic tumours often show infil-
tration of tumour-associated macrophages and neu-
Hepatocyte growth factor (HGF), also known as scat- trophils (TAMs and TANs respectively) which provide
ter factor (SF), is another growth factor involved in an abundant supply of proangiogenic factors in the
wound healing and cancer. It is mostly secreted by tumour microenvironment [105]. Adaptive immune
mesenchymal cells, although injury can upregulate cells such as T- and B-lymphocytes may also have
HGF and its receptor in murine epidermal ker- tumour-promoting properties [105].
atinocytes [93]. The HGF receptor c-MET controls
proliferation and also promotes keratinocyte migration
by coordinating the formation and orientation of focal Neutrophils in wound healing and
adhesions [93]. In diabetic rats, exogenous HGF cancer
administration accelerates wound healing by inducing Neutrophils contribute to the innate immune response
cell adhesion molecules b1-integrin and integrin-linked against infection following a barrier breach (Fig. 2).
kinase both in vivo and in vitro [94]. Platelet degranulation provides chemo-attractants
An association of c-MET with migration, invasion which recruit circulating neutrophils to the injury site
and angiogenesis is frequently observed in cancers. [106], where they perform antimicrobial functions.
While genetic mutations or amplifications are rare at Neutrophil infiltration is also seen in epithelial
the c-MET locus in cancer, an activating point muta- tumours. Although tumour-associated neutrophils
tion, Y1253D, in the MET gene is tightly linked to (TANs) are expected to be antitumorigenic, TAN-asso-
distant metastasis in HNSCC patients [95]. c-MET ciated tumours show poor clinical outcome and
overexpression is seen in HNSCC, cSCC and cuta- increased metastasis frequency. This is likely due to
neous melanoma patients, with elevated serum HGF inflammation associated with neutrophil infiltration,
levels also observed in oral SCC [96,97]. In conjunc- which promotes tumour growth [107]. In addition to
tion with its promigratory role, overexpression of killing foreign pathogens through engulfing and the
c-MET in oral SCC disrupts cadherin junctions and use of antimicrobial peptides, neutrophils form web-
significantly augments invasion [98]. Interestingly, like structures known as neutrophil extracellular traps
c-MET is also secreted in exosomes. This increases (NETs). NETs consist of chromatin fibres and antimi-
metastasis frequency in melanoma, perhaps by precon- crobial proteins and form a mesh-like structure used
ditioning possible metastasis niches such as bone mar- to trap pathogens. In mouse breast cancer, NET for-
row for cancer cells to thrive after colonization [99]. A mation occurs in the absence of infection and enhances
role for HGF/c-MET signalling in conveying stem cell lung metastasis. This clearly demonstrates the capacity
properties and drug resistance has been recently for cancers to seize neutrophil-mediated innate immu-
demonstrated [100,101]. The HGF/c-MET pathway is nity to promote cancer progression [108].
currently an attractive therapeutic target in multiple
cancers [102].
Macrophages in wound healing and
cancer
Inflammation and the immune system
Following neutrophil recruitment, circulating mono-
in tumour metastasis and wound
cytes transmigrate through endothelial cells, enter the
healing
injury site and differentiate into macrophages. Macro-
Inflammation is the body’s natural response to infec- phages perform several key tasks in regulating inflam-
tion, wounds and foreign body infiltration, and is mation during both wound healing and cancer
mainly mediated by immune cells via cytokines and metastasis, and are subdivided based on their cytokine
chemokines (Fig. 2). Inflammation is prevalent in both expression profiles [109]. M1 macrophages are proin-
healing wounds and tumours [19]. Several studies have flammatory, activated by IFN-c, and secrete IL-1b,
reported a low but significantly reduced risk of SCC in TNF-a and MMP9. M2 macrophages, activated by
people taking nonsteroidal anti-inflammatory drugs IL-4, are anti-inflammatory and secrete higher levels of
such as aspirin or ibuprofen, suggesting a role for TGF-b and IL-10 [110,111]. The presence of M1
inflammation in cancer [103,104]. Multiple cell types macrophages is associated with a chronic wound phe-
contribute to inflammation including platelets, mast notype in mouse models. Antagonizing IL-1b with a
cells, neutrophils, macrophages, dendritic cells and neutralizing antibody causes proinflammatory, chronic

6 The FEBS Journal (2018) ª 2018 Federation of European Biochemical Societies


G. M. Sundaram et al. Parallels between wound healing and cancer

Fig. 2. Illustration showing the three important cell types that play an integral role in both wound healing and cancer. Depending on the
cytokine milieu in the micro environment, macrophages differentiate into either the M1 or M2 subtype with opposing functions in regulating
inflammation in both wound and cancer niches. Macrophages also regulate angiogenesis and extracellular matrix composition in tumour
stroma making the tumour environment conducive for tumour growth. Even though the main function of neutrophils in the wound bed to
promote inflammation and defence, tumour-associated neutrophils (TANs) perform functions akin to macrophages. Both interfollicular
epidermal (IFESC) and hair follicular (HFSC) stem cells contribute to re-epithelialization. Recent evidence indicates their active role in
conferring drug resistance and EMT properties via acting as cancer stem cells.

wounds to adopt a healing phenotype containing M2 and migration into the surrounding ECM (reviewed in
macrophages [110]. In cancer, M1 macrophages antag- [114]).
onize tumour growth, while the M2 phenotype is con- Hypoxia is a key trigger for angiogenesis in the
ducive to tumour progression. M2 macrophages are wound environment and in tumours [115]. Reduced
also known as tumour-associated macrophages oxygen tension is detected by hypoxia inducible factor,
(TAMs) [112]. Hypoxia, which is common in tumour HIF1a, which stimulates production of the proangio-
microenvironments, is a major inducer of intratumoral genetic factor VEGF [116,117]. VEGF and HIF1a
macrophage recruitment through chemo-attractants mRNAs are absent in unwounded skin, upregulated
such as EMAPII, endothelin and VEGF-A. Macro- upon wounding and restored to basal levels after re-
phages in turn modulate the tumour microenvironment epithelialization [117]. Neovascularization of the
to favour invasion by secreting proteases and growth wound bed permits adequate supply of nutrients, oxy-
factors [113]. gen and immune cells for proper wound healing [118];
vascular insufficiency is a common causative factor in
chronic wounds, which also exhibit reduced levels of
Angiogenesis in wound healing and
VEGF secretion [116,119]. Angiogenesis in the wound
cancer
environment, like keratinocyte migration during re-
Both wound repair and metastasis involve the coordi- epithelialization, is self-limiting and terminates upon
nation of cell movement and proliferation. These pro- healing. Stringent regulation of angiogenesis during
cesses require an energy supply, which is obtained pre- and post-wound healing is orchestrated through
from the vasculature. Angiogenesis, referring to the controlled expression of negative (CXCL4 and Throm-
growth of new blood vessels, is essential to the pro- bospondin-1) and positive (FGF-2, VEGF and PDGF-
gression of cancer and wound healing. Wound angio- BB) regulators of angiogenesis [120]. During solid
genesis involves multiple steps with striking parallels tumour progression, hypoxia develops at the centre of
to metastasis, including endothelial cell activation, pro- the tumour mass. Both HIF1a and VEGF mRNAs
liferation, invasion through the basement membrane, increase in abundance from simple hyperplasia to

The FEBS Journal (2018) ª 2018 Federation of European Biochemical Societies 7


Parallels between wound healing and cancer G. M. Sundaram et al.

invasive SCC in multistage carcinogenesis mouse mod- healing time in rats [133]. Composite skin grafts con-
els [117]. Notably, the first report showing the ability taining stem cells derived from IFE or HF has been suc-
of endothelial cells to form capillary tubes in vitro cessfully used to treat chronic wounds [134].
demonstrated the dependency of this process on Both IFESCs and HFSCs contribute to skin cancers
tumour conditioned media, suggesting that cancer cells [135]. LGR6 is a marker associated with stem/progeni-
can induce angiogenesis [121]. Tumour cells secreted tor cells within the IFE or HF niche [136]. Unsurpris-
FGFs stimulate migration of vascular endothelial cells ingly, LGR6 was also found to be a specific CSC
towards the tumour mass and work in synergy with marker in mouse SCC models [137]. The contribution
VEGF [122]. VEGF directly stimulates the secretion of of injury-induced HFSC migration to cancer is demon-
matrix metalloproteinases (MMP-1 and MMP-2) by strated in two contiguous articles published in 2011 on
endothelial cells [123]. MMPs degrade the basement transgenic BCC mouse models. In these models, injury
membrane and allow for endothelial cell invasion. increased the frequency of BCC lesions by recruiting
Basement membrane degradation also allows tumour HFSCs to tumours, implicating a link between wound-
cells to escape the epithelial compartment boundary, ing, stem cells and cancer [138,139]. Cancer stem cells
thereby facilitating metastasis. (CSCs) may be derived from tissue-resident stem cells,
Angiogenesis in epithelial wounds and carcinoma is or from cells which acquire the capability to self-
also stimulated by inflammation. Epidermal injury trig- renew. They contribute to drug resistance and closely
gers rapid recruitment of leucocytes and initiates the associate with tumour recurrence [140].
inflammatory response. Neutrophils, macrophages, The contribution of epithelial stem cells to cancer
endothelial cells and keratinocytes in acute wounds metastasis is currently an active area of investigation. Sev-
secrete VEGF-A and promote angiogenesis [124–126]. eral studies have highlighted that the EMT and stemness
Although tumour angiogenesis does not guarantee properties are probably linked. For example, TWIST is a
metastasis, it is permissive for malignancy. The initia- classical EMT marker in epithelial cancers. Conditional
tion of angiogenesis is cited as a pivotal point in tumour ablation of TWIST at different stages of carcinogenesis
progression (the ‘angiogenic switch’), following which revealed its role in CSC maintenance independent of its
tumours can more readily expand in size as well as allow EMT related functions. [141]. Forced expression of other
for intravasation of cancer cells into the bloodstream. EMT drivers such as Snail led to the development of stem
Angiogenesis at secondary sites also facilitates the cell-like traits in mammary epithelium. Further suggest-
growth of metastases. The importance of angiogenesis ing EMT transition and CSC emergence may be coupled
to cancer progression has made it the subject of much in cancers [142]. Counterintuitively, EMT transition is a
research into anticancer drugs. Insights into candidate reversible process in both wound healing and cancers.
antiangiogenics have been gleaned from the understand- EMT is only transiently activated during the re-epithelia-
ing of angiogenesis in wound healing, and strategies are lization phase of wound healing. [143]. In cancers, EMT
being developed to either target endothelial cells is an early event and once cells reach the metastatic
directly, or to block key growth factors and proangio- organs, a mesenchymal–epithelial transition (MET)
genic pathways involved in cancer progression [127]. appears to be imperative for successful colonization [144].
Indeed, in mouse tumours derived from bladder and
prostate cancer cell lines, the tumour initiating cells
Role of stem cells in wound healing
(TICs) isolated from metastatic sites had highly enriched
and cancer metastasis
epithelial gene programme, while overexpression of Snail
Stemness, the ability of a cell to perform self-renewal, is in these cells led to reduced TIC attributes and less meta-
important for the provision of material for wound clo- static potential [145]. Taken together, the link between
sure, and is also essential to tumour formation and the cancer stem cells and metastasis is highly plastic. Multiple
repopulation of tumours following treatment. Stem cells factors such as tumour microenvironment and contextual
are present in several reservoirs within the skin, includ- dependences decide its nature [146].
ing the interfollicular epidermis (IFESC) and hair folli-
cles (HFSC) [128]. Stem cells in both niches participate
Noncoding RNAs in wound healing
in wound healing and in skin homeostasis, though their
and cancer
relative contributions may differ based on the wound
niche [129–131]. In mice, the absence of hair follicles Noncoding RNAs (ncRNA) have received remarkable
results in delayed re-epithelialization, followed by com- attention in recent years due to their pivotal and pleo-
plete wound closure [132]. The addition of purified tropic role in the regulation of proteome output of the
CD34+ HFSCs to full thickness wounds accelerates cell. They can be broadly classified based on their size,

8 The FEBS Journal (2018) ª 2018 Federation of European Biochemical Societies


G. M. Sundaram et al. Parallels between wound healing and cancer

namely, small; microRNAs (miRNAs), piRNAs and healing. Ectopic miR-483-3p expression suppresses the
endosiRNAs, medium sized; snRNA, snoRNA, tRNA in vitro and in vivo growth of HNSCC cells [164].
and large ncRNAs; intergenic/intragenic long noncod- miR-132, cited as a potential therapeutic for chronic
ing RNA (lncRNAs) and circular RNAs (cRNAs) [147]. wounds, is expressed at significantly lower levels in
A number of microRNAs help to regulate the intri- diabetic ulcers relative to normal wounds. Diabetic
cate wound healing cascade in mammals, some of mice also have reduced wound expression of miR-132
which also have well-defined roles in cancer progres- relative to wild-type mice; topical replenishment of
sion [148]. During cutaneous injury, miR-21 is upregu- miR-132 in these mice stimulates keratinocyte prolifer-
lated in both the epithelial and mesenchymal ation and accelerates wound closure [165]. However,
compartments of skin, where it actively promotes the clinical use of miR-132 in chronic wound manage-
wound healing. AntagomiR inhibition of miR-21 in ment should be approached with caution as it has
wounds reduces wound contraction and re-epitheliali- known oncogenic functions in laryngeal SCC [166].
zation [149,150]. miR-21 is a direct target of the Unlike miRNAs, lncRNAs and the roles they play
TGF-b signalling pathway, and is essential for TGF- in wound healing and epithelial cancers (excluding
b-driven keratinocyte migration [150]. miR-21 is also melanoma) are less well understood [167]. Both wound
considered to be an oncomiR in multiple cancers and healing and cancer progression are associated with
a prognostic biomarker in HNSCC, where its expression dedifferentiation of epidermal cells and their concomi-
correlates negatively with patient survival [151,152]. tant re-entry into cell cycle [94]. Differentiation antag-
Specific upregulation of miR-21 occurs during the pro- onizing noncoding RNA (DANCR) and terminal
gression from carcinoma in situ to invasive SCC, differentiation induced noncoding RNA (TINCR)
implying that its promigratory role in wound healing have opposing effects on epidermal differentiation
also occurs in cancer [153]. miR-31 is upregulated in [168,169]. TINCR has prodifferentiation functions and
wound edge keratinocytes, where it stimulates migra- may act as a tumour suppressor in cSCC, where it is
tion and proliferation in wound healing [154]. It is also suppressed [168]. Lee et al. identified eight novel
upregulated in oral SCC and has oncogenic roles in lncRNAs dysregulated in SCC – SCC misregulated
oral epithelial carcinogenesis [155,156]. miR-424 is transcripts (SMRT). The reciprocal link between neo-
induced in response to hypoxia in wounds, where it plasia and differentiation status is apparent in the case
stimulates angiogenesis [157]. Hypoxia-driven induc- of SMRT-2, which is significantly downregulated in
tion of miR-424 also promotes chemoresistance in can- SCC tissues. SMRT-2 is also essential for terminal ker-
cer by inhibiting apoptosis [158]. In pancreatic cancer, atinocyte differentiation in healthy skin. [170]. H19, a
miR-424-5p enhances proliferation and invasion in maternally imprinted lncRNA, has dual functions. It
addition to its antiapoptotic effects [159]. MicroRNAs acts either as a reservoir for the controlled release of
which delay wound healing also inhibit cancer progres- miR-675, or as a sponge for let-7 family microRNAs
sion. miR-203 is an antiproliferative miRNA with piv- [171,172]. H19 is implicated in several aspects of can-
otal roles in epidermal homeostasis. It is specifically cer progression such as genome instability, cell sur-
downregulated in the hyperproliferative compartment vival, hypoxia response, proliferation and EMT [173].
of the migrating tongue in healing wounds [160]. In H19 also promotes angiogenesis during wound healing;
the context of cancer, miR-203 exerts tumour-suppres- its downregulation in diabetic patients is correlated
sive characteristics at multiple stages of epithelial with impaired angiogenesis. Targeted delivery of H19
tumour progression; by targeting c-Myc leading to dif- lncRNA accelerates wound healing in diabetic rat
ferentiation, suppressing tumour initiating cells and by models [174].
promoting mesenchymal–epithelial transition (MET) in Cancer cells adopt multiple signalling pathways,
SCC. miR-200c suppresses keratinocyte migration and growth factors and molecular processes, ordinarily
delays re-epithelialization in human ex vivo wound used in wound healing, for their own benefit. We spec-
models [161]. In epithelial cancers, miR-200c exerts ulate that this is more likely to be achieved through
tumour-suppressive effects by targeting key mediators the hijacking of specific ‘master switches’ at the top of
of EMT and angiogenesis such as ZEB1, ZEB2, each pathway, rather than co-option of individual
MMP2 and VEGF [162,163]. The restoration of epi- pathway components. One clear example by which a
dermal homeostasis after wound closure, involving wound healing master regulator has been comman-
reversion of cells to a nonmigratory state, is also under deered can be seen in miR-198, a microRNA derived
miRNA regulation. miR-483-3p restricts keratinocyte from the 30 UTR of the follistatin-like-1 (FSTL1) tran-
proliferation by inducing cell cycle arrest, and is specif- script (Fig. 3). FSTL1 is capable of switching func-
ically upregulated during later stages of wound tions under different physiological contexts, acting

The FEBS Journal (2018) ª 2018 Federation of European Biochemical Societies 9


Parallels between wound healing and cancer G. M. Sundaram et al.

Fig. 3. A wound healing process hijacked in


cancer. Graphical representation of a dual
state molecular switch in wound healing
and cancer. MicroRNA-198 and FSTL1
protein are derived from the same primary
transcript. In physiological wound healing,
TGF-b triggers the switch from expression
of antimigratory miR-198 to FSTL1 to
initiate keratinocyte migration. In chronic
wounds, due to lack of TGF-b signalling, a
defective switch leads to persistent
expression miR-198 coupled with lack of
FSTL1 suppresses migration. On the other
hand, SCCs hijack this molecular switch
using EGF signalling as a surrogate for TGF-
b to turn off tumour-suppressive miR-198
and gain FSTL1 to promote cancer invasion
and metastasis.

either as a noncoding RNA (pri-miR-198), or a mes- demonstrated in several other cancers such as pancre-
senger RNA encoding the FSTL1 protein [63]. This atic, colorectal, liver and lung cancer [144,176–178].
change in function is reversible and forms the basis of FSTL1 promotes cell survival, invasion and metastasis
a bistable molecular switch controlling production of in glioma, liver, oesophageal SCC and lung cancer
either miR-198 or FSTL1. Crucially, the state of the [179–183]. Given the wide range of miR-198 targets
switch governs the migratory status of epidermal ker- being distinct in different tissues and the modulation
atinocytes. In healthy epidermis, exclusive expression of various signalling pathways by FSTL1 in different
of miR-198 suppresses promigratory targets, blocking cancers, miR-198/FSTL1 molecular switch may be a
keratinocyte migration. Upon injury, TGF-b signalling master controller in physiological wound healing and
flips the switch, causing wound edge keratinocytes shut cancer. It is also likely that other master switches exist
down miR-198 synthesis in favour of FSTL1 produc- and sit atop the various wound healing pathways and
tion. In addition to the derepression of promigratory processes which are seized for the progression of can-
targets caused by the absence of miR-198, FSTL1 also cer.
stimulates keratinocyte migration. Thus, a single
molecular switch regulates the expression of whole
Conclusion and future perspectives
suite of migration associated genes. In epithelial can-
cers, EGF signalling hijacks the switch to promote The need to demarcate the self from the surroundings
oncogenesis. Processing of the tumour-suppressive through maintenance of an intact barrier between the
miR-198 is abnormally suppressed, leading to sus- internal and external environments is universal to all
tained FSTL1 expression. The loss of miR-198 allows cellular life. It is therefore unsurprising that wound
for expression of its downstream promigratory targets, healing exists in one form or another, even in the sim-
while FSTL1 is a promigratory, proinflammatory plest animals. These basally branching metazoans
secreted protein which also stimulates EGF signalling. often possess immense regenerative potential such that
This creates a feed-forward loop which locks the much of the body can be regrown after injury. In ver-
switch in its defective position [175], creating a cellular tebrates, retention of a limited capacity for limb regen-
state resembling a prolonged wound healing phase. In eration is observed in some amphibian species.
this state, uncontrolled cell migration is highly con- Mammals have very restricted capabilities for tissue
ducive to metastasis [34]. The effects of this faulty replacement; most limbs and organs do not regenerate,
switch may not be limited to epithelial cancers. The and even epithelial wound repair is accompanied by
tumour-suppressive role of miR-198 has been some degree of scarring [9]. The evolutionary origin of

10 The FEBS Journal (2018) ª 2018 Federation of European Biochemical Societies


G. M. Sundaram et al. Parallels between wound healing and cancer

tumours is likely to be linked to that of wound heal- undergoing colorectal and pancreatic cancer resection
ing; spontaneous tumour formation is known to occur [196]. It is likely that multiple factors affect the rela-
in Hydra (Cnidaria), at the base of the animal evolu- tionship between anticancer treatment and wound
tionary tree. It is therefore possible that co-option of healing. Thus, careful consideration of conditions such
wound repair pathways in tumourigenesis was present as wound aetiology, drug dosage and timing of treat-
at the dawn of animal life, and that the two processes ment relative to surgery may help to minimize wound
remained linked till the present day [184]. healing complications [190].
In mammals, the literature is replete with studies on Humanity’s understanding of the interrelationship
the occurrence of common molecular ground between between epithelial wound healing and cancer has a
wound healing and tumour progression. Mice with long and extensive history, just as the two processes
basal keratinocyte-specific telomerase (TERT) overex- themselves have evolved and intertwined over phyloge-
pression show increased tumour incidence as well as netic time. While we are clear on the broad parallels
accelerated wound healing [185]. Growth-promoting between both events, the literature is far less certain
molecular pathways, such as mTOR activation, are on their distinctions. What makes cancer different
associated with both improved wound healing and from wound healing? What molecular processes govern
increased cancer incidence [186,187]. The use of BRAF self-limitation at each step in the intricate series of
inhibitors in melanoma represents an interesting events in the mammalian wound healing cascade?
demonstration of the integral link between cancer and How, over the course of cancer progression, do
wound healing. BRAF inhibitors enhance patient sur- tumour cells gradually erode away these molecular
vival, but also stimulate metastasis through ERK/ controls and commandeer the relevant pathways for
MAPK activation in RAS mutant melanoma cells their own benefit? To understand how cancer differs
[188]. This same drug-induced MAPK activation from wound healing in healthy epithelia is to gain a
improves wound healing in mouse models by stimulat- glimpse into the restorative processes required to limit
ing keratinocyte hyperproliferation [189]. the progression of cancer.
Given this degree of overlap between cellular pro-
cesses and signalling pathways used in wound healing
and cancer, and the effects of chronic wound therapy Acknowledgements
on cancer risk noted at the start of this review, the This work was supported by National Medical
possibility that anticancer treatment could interfere Research Council (NMRC) Individual Research Grant
with wound healing should not be discounted. This is (IRG) to PS and Biomedical Research Council of Sin-
particularly relevant in the management of surgical gapore, A*STAR.
wounds incurred during tumour removal or resection
procedures. Therapeutic strategies targeted at rapidly
proliferating tumour cells tend to have side effects on Conflict of interest
wound healing. Radiotherapy, for example, delays
The authors declare no competing financial interests.
wound healing due to cytotoxic effects on skin, con-
nective tissue and surrounding vasculature [190]. The
effects of chemotherapy have mainly been studied in References
animal models; in humans, cutaneous side effects
1 Dvorak HF (2015) Tumors: wounds that do not heal–
including alopecia and pigment changes are well
Redux. Cancer Immunol Res 3, 1–11.
known but wound healing studies tend to yield con-
2 Dvorak HF (1986) Tumors: wounds that do not heal.
flicting results. Alkylating agents such as cisplatin sig- Similarities between tumor stroma generation and
nificantly affect rat wound healing, but human studies wound healing. N Engl J Med 315, 1650–1659.
using therapeutic doses of cisplatin have not produced 3 Sch€afer M & Werner S (2008) Cancer as an
conclusive findings [191]. Gefitinib and cetuximab, overhealing wound: an old hypothesis revisited. Nat
both EGFR inhibitors, do not compromise surgical Rev Mol Cell Biol 9, 628–638.
wound healing in HNSCC patients [192,193]. Antian- 4 Rybinski B, Franco-Barraza J & Cukierman E
giogenics such as the systemic VEGF inhibitor Beva- (2014) The wound healing, chronic fibrosis, and
cizumab, on the other hand, may have adverse effects cancer progression triad. Physiol Genomics 46, 223–
including wound dehiscence, subcutaneous bleeding 244.
and infection [194,195]. Again, data on this are incon- 5 Eming SA, Martin P & Tomic-Canic M (2014) Wound
clusive; other studies report negligible effects of Beva- repair and regeneration: mechanisms, signaling, and
cizumab on surgical wound healing in patients translation. Sci Transl Med 6, 265sr6.

The FEBS Journal (2018) ª 2018 Federation of European Biochemical Societies 11


Parallels between wound healing and cancer G. M. Sundaram et al.

6 Sonnemann KJ & Bement WM (2011) Wound repair: 22 Stuelten CH, Barbul A, Busch JI, Sutton E, Katz R,
toward understanding and integration of single-cell Sato M, Wakefield LM, Roberts AB & Niederhuber
and multicellular wound responses. Annu Rev Cell Dev JE (2008) Acute wounds accelerate tumorigenesis by a
Biol 27, 237–263. T cell-dependent mechanism. Cancer Res 68, 7278–
7 S"
anchez Alvarado A (2006) Planarian regeneration: its 7282.
end is its beginning. Cell 124, 241–245. 23 Hobson J, Gummadidala P, Silverstrim B, Grier D,
8 Martin P (2004) Parallels between tissue repair and Bunn J, James T & Rincon M (2013) Acute
embryo morphogenesis. Development 131, 3021–3034. inflammation induced by the biopsy of mouse
9 Erickson JR & Echeverri K (2018) Learning from mammary tumors promotes the development of
regeneration research organisms: the circuitous road to metastasis. Breast Cancer Res Treat 139, 391–401.
scar free wound healing. Dev Biol 433, 144–154. 24 Antonio N, Bonnelykke-Behrndtz ML, Ward LC,
10 Takeo M, Lee W & Ito M (2015) Wound healing and Collin J, Christensen IJ, Steiniche T, Schmidt H, Feng
skin regeneration. Cold Spring Harb Perspect Med 5, Y & Martin P (2015) The wound inflammatory
1–12. response exacerbates growth of pre-neoplastic cells and
11 Shaw TJ & Martin P (2009) Wound repair at a glance. progression to cancer. EMBO J 34, 2219–2236.
J Cell Sci 122, 3209–3213. 25 Rosowski EE & Huttenlocher A (2015) Neutrophils,
12 Nurden AT, Nurden P, Sanchez M, Andia I & wounds, and cancer progression. Dev Cell 34,
Eduardo A (2008) Platelets and wound healing. Front 134–136.
Biosci 2, 3532–3548. 26 Noor S, Zubair M & Ahmad J (2015) Diabetic foot
13 Strbo N, Yin N & Stojadinovic O (2014) Innate and ulcer – a review on pathophysiology, classification and
adaptive immune responses in wound epithelialization. microbial etiology. Diabetes Metab Syndr Clin Res Rev
Adv Wound Care 3, 492–501. 9, 192–199.
14 Paladini RD, Takahashi K, Bravo NS & Coulombe 27 Brem H & Tomic-Canic M (2007) Cellular and
PA (1996) Onset of re-epithelialization after skin injury molecular basis of wound healing in diabetes. J Clin
correlates with a reorganization of keratin filaments in Invest 117, 1219–1222.
wound edge keratinocytes: defining a potential role for 28 Pernicova I & Korbonits M (2014) Metformin-Mode
keratin 16. J Cell Biol 132, 381–397. of action and clinical implications for diabetes and
15 Walmsley GG, Maan ZN, Wong VW, Duscher D, Hu cancer. Nat Rev Endocrinol 10, 143–156.
MS, Zielins ER, Wearda T, Muhonen E, McArdle A, 29 Morales DR & Morris AD (2015) Metformin in
Tevlin R et al. (2015) Scarless wound healing. Plast cancer treatment and prevention. Annu Rev Med 66,
Reconstr Surg 135, 907–917. 17–29.
16 Yates CC, Hebda P & Wells A (2012) Skin wound 30 Barrientos S, Brem H, Stojadinovic O & Tomic-Canic
healing and scarring: fetal wounds and regenerative M (2014) Clinical application of growth factors and
restitution. Birth Defects Res Part C – Embryo Today cytokines in wound healing. Wound Repair Regen 22,
Rev 96, 325–333. 569–578.
17 Virchow R & Virchow R (1863) Aetiologie der 31 Abreu-Blanco MT, Watts JJ, Verboon JM &
neoplastischen Geschwulste/Pathogenie der Parkhurst SM (2012) Cytoskeleton responses in wound
neoplastischen Geschwulste. Verlag von August repair. Cell Mol Life Sci 69, 2469–2483.
Hirschwald, Berlin. 32 Hopkinson SB, Hamill KJ, Wu Y, Eisenberg JL,
18 Antonio N, Ward LC, Collin J, Steiniche T, Schmidt Hiroyasu S & Jones JCR (2014) Focal contact and
H, Feng Y, Martin P, Bonnelykke-Behrndtz ML, hemidesmosomal proteins in keratinocyte migration
Ward LC, Collin J et al. (2015) The wound and wound repair. Adv Wound Care 3, 247–263.
inflammatory response exacerbates growth of pre- 33 Caley MP, Martins VLC & O’Toole EA (2015)
neoplastic cells and progression to cancer. EMBO J Metalloproteinases and wound healing. Adv Wound
34, 1–18. Care 4, 225–234.
19 Balkwill F & Mantovani A (2001) Inflammation and 34 Sundaram GM, Ismail HM, Bashir M, Muhuri M,
cancer: Back to Virchow? Lancet 357, 539–545. Vaz C, Nama S, Ow GS, Vladimirovna IA,
20 Colotta F, Allavena P, Sica A, Garlanda C & Ramalingam R, Burke B et al. (2017) EGF hijacks
Mantovani A (2009) Cancer-related inflammation, the miR-198/FSTL1 wound-healing switch and steers a
seventh hallmark of cancer: links to genetic instability. two-pronged pathway toward metastasis. J Exp Med
Carcinogenesis 30, 1073–1081. 214, 2889–2900.
21 Lu Y, Jingyan G, Baorong S, Peng J, Xu Y & Cai S 35 Hardwicke J, Schmaljohann D, Boyce D & Thomas D
(2012) Expression of EGFR, Her2 predict lymph node (2008) Epidermal growth factor therapy and wound
metastasis (LNM)-associated metastasis in colorectal healing — past, present and future perspectives.
cancer. Cancer Biomarkers 11, 219–226. Surgeon 6, 172–177.

12 The FEBS Journal (2018) ª 2018 Federation of European Biochemical Societies


G. M. Sundaram et al. Parallels between wound healing and cancer

36 Wieduwilt MJ & Moasser MM (2008) The epidermal 49 Uribe P & Gonzalez S (2011) Epidermal growth factor
growth factor receptor family: biology driving targeted receptor (EGFR) and squamous cell carcinoma of the
therapeutics. Cell Mol Life Sci 65, 1566–1584. skin: molecular bases for EGFR-targeted therapy.
37 Schelfhout VR, Coene ED, Delaey B, Waeytens AA, Pathol – Res Pract 207, 337–342.
De Rycke L, Deleu M & De Potter CR (2002) The 50 Vogelstein B, Papadopoulos N, Velculescu VE, Zhou
role of heregulin-alpha as a motility factor and S, Diaz LA & Kinzler KW (2013) Cancer genome
amphiregulin as a growth factor in wound healing. landscapes. Science (80-). 340, 1546–1558.
J Pathol 198, 523–533. 51 Huang S-F, Chien H-T, Cheng S-D, Chuang W-Y,
38 Repertinger SK, Campagnaro E, Fuhrman J, El- Liao C-T & Wang H-M (2017) EGFR copy number
Abaseri T, Yuspa SH & Hansen LA (2004) EGFR alterations in primary tumors, metastatic lymph nodes,
enhances early healing after cutaneous incisional and recurrent and multiple primary tumors in oral
wounding. J Invest Dermatol 123, 982–989. cavity squamous cell carcinoma. BMC Cancer 17, 592.
39 Pastore S, Mascia F, Mariani V & Girolomoni G 52 Ohashi Y, Kumagai K, Miyata Y, Matsubara R,
(2008) The epidermal growth factor receptor system in Kitaura K, Suzuki S, Hamada Y & Suzuki R (2016)
skin repair and inflammation. J Invest Dermatol 128, Overexpression of ErbB4 is an independent marker for
1365–1374. lymph node metastasis in Japanese patients with oral
40 Shirakata Y (2005) Heparin-binding EGF-like growth squamous cell carcinoma. Oral Surg Oral Med Oral
factor accelerates keratinocyte migration and skin Pathol Oral Radiol 122, 313–321.
wound healing. J Cell Sci 118, 2363–2370. 53 Xu Q, Zhang Q, Ishida Y, Hajjar S, Tang X, Shi H,
41 Pastar I, Stojadinovic O, Yin NC, Ramirez H, Dang CV & Le AD (2016) EGF induces epithelial-
Nusbaum AG, Sawaya A, Patel SB, Khalid L, Isseroff mesenchymal transition and cancer stem-like cell
RR & Tomic-Canic M (2014) Epithelialization in properties in human oral cancer cells via promoting
wound healing: a comprehensive review. Adv Wound warburg effect. Oncotarget 8, 9557–9571.
Care 3, 445–464. 54 Pastore S, Lulli D & Girolomoni G (2014) Epidermal
42 Mainiero F, Pepe A, Yeon M, Ren Y & Giancotti FG growth factor receptor signalling in keratinocyte
(1996) The intracellular functions of a6b4 integrin are biology: implications for skin toxicity of tyrosine
regulated by EGF. J Cell Biol 134, 241–253. kinase inhibitors. Arch Toxicol 88, 1189–1203.
43 Mariotti A, Kedeshian PA, Dans M, Curatola AM, 55 Chan DLH, Segelov E, Wong RSH, Smith A,
Gagnoux-Palacios L & Giancotti FG (2001) EGF-R Herbertson RA, Li BT, Tebbutt N, Price T & Pavlakis
signaling through Fyn kinase disrupts the function of N (2017) Epidermal growth factor receptor (EGFR)
integrin a6b4 at hemidesmosomes: role in epithelial inhibitors for metastatic colorectal cancer. Cochrane
cell migration and carcinoma invasion. J Cell Biol 155, Database Syst, Rev.
447–458. 56 Cassell A & Grandis JR (2010) Investigational EGFR-
44 Hudson LG, Moss NM & Stack MS (2009) EGF- targeted therapy in head and neck squamous cell
receptor regulation of matrix metalloproteinases in carcinoma. Expert Opin Investig Drugs 19, 709–722.
epithelial ovarian carcinoma. Futur Oncol 5, 323–338. 57 Echarri MJ, Lopez-Martin A & Hitt R (2016)
45 Salo T, Makela M, Kylmaniemi M, Autio-Harmainen Targeted therapy in locally advanced and recurrent/
H & Larjava H (1994) Expression of matrix metastatic head and neck squamous cell carcinoma
metalloproteinase-2 and -9 during human wound (LA-R/M HNSCC). Cancers (Basel) 8, 27.
healing. Lab Invest 70, 176–182. 58 Bartholomew C, Eastlake L, Dunn P & Yiannakis D
46 Ruokolainen H, P€a€akk€ o P & Turpeenniemi-Hujanen (2017) EGFR targeted therapy in lung cancer; an
T (2004) Expression of matrix metalloproteinase-9 in evolving story. Respir Med Case Reports 20, 137–140.
head and neck squamous cell carcinoma: a potential 59 Ayyappan S, Prabhakar D & Sharma N (2013)
marker for prognosis. Clin Cancer Res 10, Epidermal growth factor receptor (EGFR)-targeted
3110–3116. therapies in esophagogastric cancer. Anticancer Res 33,
47 Liu Y, Min D, Bolton T, Nub"e V, Twigg SM, Yue 4139–4155.
DK & McLennan SV (2009) Increased matrix 60 Roberts AB & Sporn MB (1992) Differential
metalloproteinase-9 predicts poor wound healing in expression of the TGF-b isoforms in embryogenesis
diabetic foot ulcers. Diabetes Care 32, 117–119. suggests specific roles in developing and adult tissues.
48 O’Grady A, Dunne C, O’Kelly P, Murphy GM, Mol Reprod Dev 32, 91–98.
Leader M & Kay E (2007) Differential expression of 61 Nagaraj NS & Datta PK (2010) Targeting the
matrix metalloproteinase (MMP)-2, MMP-9 and tissue transforming growth factor-beta signaling pathway in
inhibitor of metalloproteinase (TIMP)-1 and TIMP-2 human cancer. Expert Opin Investig Drugs 19, 77–91.
in non-melanoma skin cancer: implications for tumour 62 Calleja LR, Jacques C, Lamoureux F, Baud’Huin M,
progression. Histopathology 51, 793–804. Gabriel MT, Quillard T, Sahay D, Perrot P, Amiaud

The FEBS Journal (2018) ª 2018 Federation of European Biochemical Societies 13


Parallels between wound healing and cancer G. M. Sundaram et al.

J, Charrier C et al. (2016) DNp63a silences a miRNA differentiation in human oral squamous cell
program to aberrantly initiate a wound-healing carcinomas. Int J Oncol 32, 323–331.
program that promotes TGFb-induced metastasis. 74 Meng W, Xia Q, Wu L, Chen S, He X, Zhang L, Gao
Cancer Res 76, 3236–3251. Q & Zhou H (2011) Downregulation of TGF-beta
63 Sundaram GM, Common JEA, Gopal FE, Srikanta S, receptor types II and III in oral squamous cell
Lakshman K, Lunny DP, Lim TC, Tanavde V, Lane carcinoma and oral carcinoma-associated fibroblasts.
EB & Sampath P (2013) ‘See-saw’ expression of BMC Cancer 11, 1–13.
microRNA-198 and FSTL1 from a single transcript in 75 Lu SL, Reh D, Li AG, Woods J, Corless CL, Kulesz-
wound healing. Nature 495, 103–106. Martin M & Wang XJ (2004) Overexpression of
64 O’Kane S & Ferguson MW (1997) Transforming transforming growth factor b1 in head and neck
growth factor beta s and wound healing. Int J Biochem epithelia results in inflammation, angiogenesis, and
Cell Biol 29, 63–78. epithelial hyperproliferation. Cancer Res 64, 4405–4410.
65 Yang L, Chan T, Demare J, Iwashina T, Ghahary A, 76 Weeks BH, He W, Olson KL & Wang XJ (2001)
Scott PG & Tredget EE (2001) Healing of burn Inducible expression of transforming growth factor
wounds in transgenic mice overexpressing transforming beta1 in papillomas causes rapid metastasis. Cancer
growth factor-beta 1 in the epidermis. Am J Pathol Res 61, 7435–7443.
159, 2147–2157. 77 Han G, Lu SL, Li AG, He W, Corless CL, Kulesz-
66 Denton CP, Khan K, Hoyles RK, Shiwen X, Leoni Martin M & Wang XJ (2005) Distinct mechanisms of
P, Chen Y, Eastwood M & Abraham DJ (2009) TGF-b1-mediated epithelial-to- mesenchymal
Inducible lineage-specific deletion of TbetaRII in transition and metastasis during skin carcinogenesis.
fibroblasts defines a pivotal regulatory role during J Clin Invest 115, 1714–1723.
adult skin wound healing. J Invest Dermatol 129, 78 Nerlich AG, Sauer U, Ruoss I & Hagedorn HG (2003)
194–204. High frequency of TGF-beta-receptor-II mutations in
67 Wang X-J, Han G, Owens P, Siddiqui Y & Li AG microdissected tissue samples from laryngeal squamous
(2006) Role of TGF beta-mediated inflammation in cell carcinomas. Lab Invest 83, 1241–1251.
cutaneous wound healing. J Investig Dermatol Symp 79 Weber CE, Li NY, Wai PY & Kuo PC (2012)
Proc 11, 112–117. Epithelial-mesenchymal transition, TGF-b, and
68 D"ecline F, Okamoto O, Mallein-Gerin F, Helbert B, osteopontin in wound healing and tissue remodeling
Bernaud J, Rigal D & Rousselle P (2003) Keratinocyte after injury. J Burn Care Res 33, 311–318.
motility induced by TGF-b1 is accompanied by 80 Kuwahara M, Hatoko M, Tada H & Tanaka A (2001)
dramatic changes in cellular interactions with laminin E-cadherin expression in wound healing of mouse skin.
5. Cell Motil Cytoskeleton 54, 64–80. J Cutan Pathol 28, 191–199.
69 Pastar I, Stojadinovic O, Krzyzanowska A, Barrientos 81 Krisanaprakornkit S & Iamaroon A (2012) Epithelial-
S, Stuelten C, Zimmerman K, Blumenberg M, Brem H mesenchymal transition in oral squamous cell
& Tomic-Canic M (2010) Attenuation of the carcinoma. ISRN Oncol 2012, 1–10.
transforming growth factor beta-signaling pathway in 82 Yokoyama K, Kamata N, Hayashi E, Hoteiya T,
chronic venous ulcers. Mol Med 16, 92–101. Ueda N, Fujimoto R & Nagayama M (2001) Reverse
70 Schmid P, Cox D, Bilbe G, McMaster G, Morrison C, correlation of E-cadherin and snail expression in oral
Stahelin H, Luscher N & Seiler W (1993) TGF-beta s squamous cell carcinoma cells in vitro. Oral Oncol 37,
and TGF-beta type II receptor in human epidermis: 65–71.
differential expression in acute and chronic skin 83 vonBurstin J, Eser S, Paul MC, Seidler B, Brandl M,
wounds. J Pathol 171, 191–197. Messer M, vonWerder A, Schmidt A, Mages J, Pagel
71 Jude EB, Blakytny R, Bulmer J, Boulton AJM & P et al. (2009) E-cadherin regulates metastasis of
Ferguson MWJ (2002) Transforming growth factor- pancreatic cancer in vivo and is suppressed by a
beta 1, 2, 3 and receptor type I and II in diabetic foot SNAIL/HDAC1/HDAC2 repressor complex.
ulcers. Diabet Med 19, 440–447. Gastroenterology 137, 361–371.
72 Nunan R, Harding KG & Martin P (2014) Clinical 84 Ornitz DM & Itoh N (2015) The fibroblast growth
challenges of chronic wounds: searching for an optimal factor signaling pathway. Wiley Interdiscip Rev Dev
animal model to recapitulate their complexity. Dis Biol 4, 215–266.
Model Mech 7, 1205–1213. 85 Komi-Kuramochi A, Kawano M, Oda Y, Asada M,
73 Mincione G, Di Marcantonio MC, Artese L, Vianale Suzuki M, Oki J & Imamura T (2005) Expression of
G, Piccirelli A, Piccirilli M, Perrotti V, Rubini C, fibroblast growth factors and their receptors during
Piattelli A & Muraro R (2008) Loss of expression of full-thickness skin wound healing in young and aged
TGF-beta1, TbetaRI, and TbetaRII correlates with mice. J Endocrinol 186, 273–289.

14 The FEBS Journal (2018) ª 2018 Federation of European Biochemical Societies


G. M. Sundaram et al. Parallels between wound healing and cancer

86 Sogabe Y, Abe M, Yokoyama Y & Ishikawa O (2006) and neck squamous cell cancer. Clin Cancer Res 15,
Basic fibroblast growth factor stimulates human 3740–3750.
keratinocyte motility by Rac activation. Wound Repair 97 Lee YJ, Kim DH, Lee SH, Kim DW, Nam HS & Cho
Regen 14, 457–462. MK (2011) Expression of the c-met proteins in
87 Werner S, Smola H, Liao X, Longaker MT, Krieg T, malignant skin cancers. Ann Dermatol 23, 33–38.
Hofschneider PH & Williams LT (1994) The function 98 Murai M, Shen X, Huang L, Carpenter WM, Lin CS,
of KGF in morphogenesis of epithelium and Silverman S, Regezi J & Kramer RH (2004)
reepithelialization of wounds. Science 266, 819–822. Overexpression of c-met in oral SCC promotes
88 Tsuboi R, Sato C, Kurita Y, Ron D, Rubin JS & hepatocyte growth factor-induced disruption of cadherin
Ogawa H (1993) Keratinocyte growth factor (FGF-7) junctions and invasion. Int J Oncol 25, 831–840.
stimulates migration and plasminogen activator 99 Peinado H, Ale#ckovi"c M, Lavotshkin S, Matei I,
activity of normal human keratinocytes. J Invest Costa-Silva B, Moreno-Bueno G, Hergueta-Redondo
Dermatol 101, 49–53. M, Williams C, Garc"ıa-Santos G, Ghajar CM et al.
89 Nguyen PT, Tsunematsu T, Yanagisawa S, Kudo Y, (2012) Melanoma exosomes educate bone marrow
Miyauchi M, Kamata N & Takata T (2013) The progenitor cells toward a pro-metastatic phenotype
FGFR1 inhibitor PD173074 induces mesenchymal- through MET. Nat Med 18, 883–891.
epithelial transition through the transcription factor 100 Sun S & Wang Z (2011) Head neck squamous cell
AP-1. Br J Cancer 109, 2248–2258. carcinoma c-Met(+) cells display cancer stem cell
90 Brown WS, Tan L, Smith A, Gray NS, Wendt properties and are responsible for cisplatin-resistance
MK, Wendt M, Taylor M, Schiemann B, Sossey- and metastasis. Int J Cancer 129, 2337–2348.
Alaoui K, Schiemann W et al. (2016) Covalent 101 Lim YC, Kang HJ & Moon JH (2014) C-Met
targeting of fibroblast growth factor receptor pathway promotes self-renewal and tumorigenecity of
inhibits metastatic breast cancer. Mol Cancer Ther head and neck squamous cell carcinoma stem-like cell.
15, R24–R2106. Oral Oncol 50, 633–639.
91 Tillman BN, Yanik M, Birkeland AC, Liu CJ, 102 Matsumoto K, Umitsu M, De Silva DM, Roy A &
Hovelson DH, Cani AK, Palanisamy N, Carskadon S, Bottaro DP (2017) Hepatocyte growth factor/MET in
Carey TE, Bradford CR et al. (2016) Fibroblast cancer progression and biomarker discovery. Cancer
growth factor family aberrations as a putative driver Sci 108, 296–307.
of head and neck squamous cell carcinoma in an 103 Reinau D, Surber C, Jick SS & Meier CR (2015)
epidemiologically low-risk patient as defined by Nonsteroidal anti-inflammatory drugs and the risk of
targeted sequencing. Head Neck 38, E1646–E1652. nonmelanoma skin cancer. Int J Cancer 137, 144–153.
92 Brands RC, Knierim LM, De Donno F, Steinacker V, 104 Muranushi C, Olsen CM, Pandeya N & Green AC
Hartmann S, Seher A, K€ ubler AC & M€ uller-Richter (2015) Aspirin and nonsteroidal anti-inflammatory
UDA (2017) Targeting VEGFR and FGFR in head drugs can prevent cutaneous squamous cell carcinoma:
and neck squamous cell carcinoma in vitro. Oncol Rep a systematic review and meta-analysis. J Invest
38, 1877–1885. Dermatol 135, 975–983.
93 Chmielowiec J, Borowiak M, Morkel M, Stradal T, 105 Smith HA & Kang Y (2013) The metastasis-promoting
Munz B, Werner S, Wehland J, Birchmeier C & roles of tumor-associated immune cells. J Mol Med 91,
Birchmeier W (2007) c-Met is essential for wound 411–429.
healing in the skin. J Cell Biol 177, 151–162. 106 Koh TJ & DiPietro LA (2011) Inflammation and
94 Li JF, Duan HF, Wu CT, Zhang DJ, Deng Y, Yin wound healing: the role of the macrophage. Expert
HL, Han B, Gong HC, Wang HW & Wang YL (2013) Rev Mol Med 13, e23.
HGF accelerates wound healing by promoting the 107 Uribe-Querol E & Rosales C (2015) Neutrophils in
dedifferentiation of epidermal cells through b 1 - cancer: two sides of the same coin. J Immunol Res
Integrin/ILK pathway. Biomed Res Int 2013, 1–9. 2015, 983698.
95 Ghadjar P, Blank-Liss W, Simcock M, Hegyi I, Beer 108 Park J, Wysocki RW, Amoozgar Z, Maiorino L, Fein
KT, Moch H, Aebersold DM & Zimmer Y (2009) MR, Jorns J, Schott AF, Kinugasa-Katayama Y, Lee
MET Y1253D-activating point mutation and Y, Won NH et al. (2016) Cancer cells induce
development of distant metastasis in advanced head metastasis-supporting neutrophil extracellular DNA
and neck cancers. Clin Exp Metastasis 26, traps. Sci Transl Med 8, 138.
809–815. 109 Condeelis J & Pollard JW (2006) Macrophages:
96 Knowles LM, Stabile LP, Egloff AM, Rothstein ME, Obligate partners for tumor cell migration, invasion,
Thomas SM, Gubish CT, Lerner EC, Seethala RR, and metastasis. Cell 124, 263–266.
Suzuki S, Quesnelle KM et al. (2009) HGF and c-Met 110 Mirza RE, Fang MM, Ennis WJ & Kohl TJ (2013)
participate in paracrine tumorigenic pathways in head Blocking interleukin-1b induces a healing-associated

The FEBS Journal (2018) ª 2018 Federation of European Biochemical Societies 15


Parallels between wound healing and cancer G. M. Sundaram et al.

wound macrophage phenotype and improves healing production and regulated angiogenesis. J Immunol 170,
in type 2 diabetes. Diabetes 62, 2579–2587. 3369–3376.
111 Martinez FO & Gordon S (2014) The M1 and M2 126 Ohki Y, Heissig B, Sato Y, Akiyama H, Zhu Z,
paradigm of macrophage activation: time for Hicklin DJ, Shimada K, Ogawa H, Daida H, Hattori
reassessment. F1000Prime Rep. 6, 13. K et al. (2005) Granulocyte colony-stimulating factor
112 Yang L & Zhang Y (2017) Tumor-associated promotes neovascularization by releasing vascular
macrophages: from basic research to clinical endothelial growth factor from neutrophils. FASEB J
application. J Hematol Oncol 10, 58. 19, 2005–2007.
113 Chanmee T, Ontong P, Konno K & Itano N (2014) 127 Bielenberg DR & Zetter BR (2015) The contribution
Tumor-associated macrophages as major players in of angiogenesis to the process of metastasis. Cancer J
the tumor microenvironment. Cancers (Basel) 6, (United States) 21, 267–273.
1670–1690. 128 Plikus MV, Gay DL, Treffeisen E, Wang A,
114 Ucuzian AA, Gassman AA, East AT & Greisler HP Supapannachart RJ & Cotsarelis G (2012) Epithelial
(2010) Molecular mediators of angiogenesis. J Burn stem cells and implications for wound repair. Semin
Care Res 31, 158–175. Cell Dev Biol 23, 946–953.
115 Winkler F (2017) Hostile takeover: how tumours 129 Ito M, Yang Z, Andl T, Cui C, Kim N, Millar SE &
hijack pre-existing vascular environments to thrive. Cotsarelis G (2007) Wnt-dependent de novo hair
J Pathol 242, 267–272. follicle regeneration in adult mouse skin after
116 Bao P, Kodra A, Tomic-Canic M, Golinko MS, wounding. Nature 447, 316–320.
Ehrlich HP, Brem H & Ph D (2009) The role of 130 Vagnozzi AN, Reiter JF & Wong SY (2015) Hair
vascular endothelial growth factor in wound healing. follicle and interfollicular epidermal stem cells make
J Surg Res 153, 347–358. varying contributions to wound regeneration. Cell
117 Elson DA, Ryan HE, Snow JW, Johnson R & Cycle 14, 3408–3417.
Arbeit JM (2000) Coordinate up-regulation of 131 Mascr"e G, Dekoninck S, Drogat B, Youssef KK,
hypoxia inducible factor (HIF)-1a and HIF-1 Broh"ee S, Sotiropoulou PA, Simons BD & Blanpain
target genes during multi-stage epidermal C (2012) Distinct contribution of stem and
carcinogenesis and wound healing. Cancer Res 60, progenitor cells to epidermal maintenance. Nature
6189–6195. 489, 257–262.
118 Tonnesen MG, Feng X & Clark RAF (2000) 132 Langton AK, Herrick SE & Headon DJ (2008) An
Angiogenesis in wound healing. J Investig extended epidermal response heals cutaneous wounds
Dermatology Symp Proc 5, 40–46. in the absence of a hair follicle stem cell contribution.
119 Zhou K, Ma Y & Brogan MS (2015) Chronic and J Invest Dermatol 128, 1311–1318.
non-healing wounds: the story of vascular 133 Heidari F, Yari A, Rasoolijazi H, Soleimani M,
endothelial growth factor. Med Hypotheses 85, 399– Dehpoor A, Sajedi N, Joulai Veijouye S & Nobakht
404. M (2016) Bulge hair follicle stem cells accelerate
120 Kareva I, Abou-Slaybi A, Dodd O, Dashevsky O & cutaneous wound healing in rats. Wounds a Compend
Klement GL (2016) Normal wound healing and tumor Clin Res Pract 28, 132–141.
angiogenesis as a game of competitive inhibition. 134 Ojeh N, Pastar I, Tomic-Canic M & Stojadinovic O
PLoS One 11, e0166655. (2015) Stem cells in skin regeneration, wound healing,
121 Folkman J & Haudenschild C (1980) Angiogenesis and their clinical applications. Int J Mol Sci 16,
in vitro. Nature 288, 551–556. 25476–25501.
122 Korc M & Friesel RE (2009) The role of fibroblast 135 Lapouge G, Youssef KK, Vokaer B, Achouri Y,
growth factors in tumor growth. Curr Cancer Drug Michaux C, Sotiropoulou PA & Blanpain C (2011)
Targets 9, 639–651. Identifying the cellular origin of squamous skin
123 Unemori EN, Ferrara N, Bauer EA & Amento EP tumors. Proc Natl Acad Sci USA 108, 7431–7436.
(1992) Vascular endothelial growth factor induces 136 F€ullgrabe A, Joost S, Are A, Jacob T, Sivan U,
interstitial collagenase expression in human endothelial Haegebarth A, Linnarsson S, Simons BD, Clevers H,
cells. J Cell Physiol 153, 557–562. Toftg$ ard R et al. (2015) Dynamics of
124 Eming SA, Brachvogel B, Odorisio T & Koch M Lgr6 + progenitor cells in the hair follicle, sebaceous
(2007) Regulation of angiogenesis: wound healing as a gland, and interfollicular epidermis. Stem Cell Reports
model. Prog Histochem Cytochem 42, 115–170. 5, 843–855.
125 Li A, Dubey S, Varney ML, Dave BJ & Singh RK 137 Huang PY, Kandyba E, Jabouille A, Sjolund J,
(2003) IL-8 directly enhanced endothelial cell survival, Kumar A, Halliwill K, McCreery M, DelRosario R,
proliferation, and matrix metalloproteinases Kang HC, Wong CE et al. (2017) Lgr6 is a stem cell

16 The FEBS Journal (2018) ª 2018 Federation of European Biochemical Societies


G. M. Sundaram et al. Parallels between wound healing and cancer

marker in mouse skin squamous cell carcinoma. Nat 151 Cho WCS (2007) OncomiRs: The discovery and
Genet 49, 1624–1634. progress of microRNAs in cancers. Mol Cancer 6, 60.
138 Kasper M, Jaks V, Are A, Bergstrom A, Schwager A, 152 Arantes LMRB, Laus AC, Melendez ME, de
Svard J, Teglund S, Barker N & Toftgard R (2011) Carvalho AC, Sorroche BP, De Marchi PRM,
Wounding enhances epidermal tumorigenesis by Evangelista AF, Scapulatempo-Neto C, de Souza
recruiting hair follicle keratinocytes. Proc Natl Acad Viana L & Carvalho AL (2017) MiR-21 as prognostic
Sci USA 108, 4099–4104. biomarker in head and neck squamous cell carcinoma
139 Wong SY & Reiter JF (2011) Wounding mobilizes patients undergoing an organ preservation protocol.
hair follicle stem cells to form tumors. Proc Natl Acad Oncotarget 8, 9911–9921.
Sci USA 108, 4093–4098. 153 Stojadinovic O, Ramirez H, Pastar I, Gordon KA,
140 Jian Z, Strait A, Jimeno A & Wang X-J (2017) Cancer Stone R, Choudhary S, Badiavas E, Nouri K &
stem cells in squamous cell carcinoma. J Invest Tomic-Canic M (2017) MiR-21 and miR-205 are
Dermatol 137, 31–37. induced in invasive cutaneous squamous cell
141 Beck B, Lapouge G, Rorive S, Drogat B, carcinomas. Arch Dermatol Res 309, 133–139.
Desaedelaere K, Delafaille S, Dubois C, Salmon I, 154 Li D, Li X, Wang A, Meisgen F, Pivarcsi A, Sonkoly
Willekens K, Marine JC et al. (2015) Different levels E, St$
ahle M & Land"en NX (2015) MicroRNA-31
of Twist1 regulate skin tumor initiation, stemness, and promotes skin wound healing by enhancing
progression. Cell Stem Cell 16, 67–79. keratinocyte proliferation and migration. J Invest
142 Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Dermatol 135, 1676–1685.
Zhou AY, Brooks M, Reinhard F, Zhang CC, 155 Zhang T, Wang Q, Zhao D, Cui Y, Cao B, Guo L &
Shipitsin M et al. (2008) The epithelial-mesenchymal Lu SH (2011) The oncogenetic role of microRNA-31
transition generates cells with properties of stem cells. as a potential biomarker in oesophageal squamous cell
Cell 133, 704–715. carcinoma. Clin Sci 121, 437–447.
143 Haensel D & Dai X (2018) Epithelial-to-mesenchymal 156 Hung PS, Tu HF, Kao SY, Yang CC, Liu CJ, Huang
transition in cutaneous wound healing: where we are TY, Chang KW & Lin SC (2014) miR-31 is
and where we are heading. Dev Dyn 247, 473–480. upregulated in oral premalignant epithelium and
144 Nieto MA, Huang RYYJ, Jackson RAA & Thiery contributes to the immortalization of normal oral
JPP (2016) EMT: 2016. Cell 166, 21–45. keratinocytes. Carcinogenesis 35, 1162–1171.
145 Celi%
a-Terrassa T, Meca-Cort"es O, " Mateo F, De Paz 157 Ghosh G, Subramanian IV, Adhikari N, Zhang X,
AM, Rubio N, Arnal-Estap"e A, Ell BJ, Bermudo R, Joshi HP, Basi D, Chandrashekhar YS, Hall JL, Roy
D"ıaz A, Guerra-Rebollo M et al. (2012) Epithelial- S, Zeng Y et al. (2010) Hypoxia-induced microRNA-
mesenchymal transition can suppress major attributes 424 expression in human endothelial cells regulates
of human epithelial tumor-initiating cells. J Clin Invest HIF-a isoforms and promotes angiogenesis. J Clin
122, 1849–1868. Invest 120, 4141–4154.
146 Beerling E, Seinstra D, de Wit E, Kester L, van der 158 Zhang D, Shi Z, Li M & Mi J (2014) Hypoxia-
Velden D, Maynard C, Sch€afer R, van Diest P, Voest E, induced miR-424 decreases tumor sensitivity to
van Oudenaarden A et al. (2016) Plasticity between chemotherapy by inhibiting apoptosis. Cell Death Dis
epithelial and mesenchymal states unlinks EMT from 5, e1301.
metastasis-enhancing stem cell capacity. Cell Rep 14, 159 Wu K, Hu G, He X, Zhou P, Li J, He B & Sun W
2281–2288. (2013) MicroRNA-424-5p suppresses the expression of
147 Hombach S & Kretz M (2016) Non-coding RNAs: socs6 in pancreatic cancer. Pathol Oncol Res 19, 739–
classification, biology and functioning. Adv Exp Med 748.
Biol 937, 3–17. 160 Viticchi%e G, Lena AM, Cianfarani F, Odorisio T,
148 Luan A, Hu MS, Leavitt T, Brett EA, Wang KC, Annicchiarico-Petruzzelli M, Melino G & Candi E
Longaker MT & Wan D (2018) Noncoding RNAs in (2012) MicroRNA-203 contributes to skin re-
wound healing: a new and vast frontier. Adv Wound epithelialization. Cell Death Dis 3, e435.
Care 7, 19–27. 161 Aunin E, Broadley D, Ahmed MI, Mardaryev AN &
149 Wang T, Feng Y, Sun H, Zhang L, Hao L, Shi C, Wang Botchkareva NV (2017) Exploring a Role for
J, Li R, Ran X, Su Y et al. (2012) MiR-21 regulates Regulatory miRNAs in Wound Healing during
skin wound healing by targeting multiple aspects of the Ageing: Involvement of miR-200c in wound repair, p.
healing process. Am J Pathol 181, 1911–1920. 7. Sci, Rep.
150 Yang X, Wang J, Guo SL, Fan KJ, Li J, Wang YL, 162 Tamagawa S, Beder LB, Hotomi M, Gunduz M, Yata
Teng Y & Yang X (2011) miR-21 promotes K, Grenman R & Yamanaka N (2014) Role of miR-
keratinocyte migration and re-epithelialization during 200c/miR-141 in the regulation of epithelial-
wound healing. Int J Biol Sci 7, 685–690. mesenchymal transition and migration in head and

The FEBS Journal (2018) ª 2018 Federation of European Biochemical Societies 17


Parallels between wound healing and cancer G. M. Sundaram et al.

neck squamous cell carcinoma. Int J Mol Med 33, endogenous RNA for the treatment of diabetic
879–886. wounds. Drug Deliv 25, 241–255.
163 Sulaiman SA, Ab Mutalib N-S & Jamal R (2016) 175 Ellisen LW (2017) A wound-healing program is
miR-200c regulation of metastases in ovarian cancer: hijacked to promote cancer metastasis. J Exp Med
potential role in epithelial and mesenchymal transition. 214, 2813–2815.
Front Pharmacol 7, 271. 176 Wu S, Zhang G, Li P, Chen S, Zhang F, Li J, Jiang
164 Bertero T, Bourget-Ponzio I, Puissant A, Loubat A, C, Chen X, Wang Y, Du Y et al. (2016) miR-198
Mari B, Meneguzzi G, Auberger P, Barbry P, Ponzio targets SHMT1 to inhibit cell proliferation and
G & Rezzonico R (2013) Tumor suppressor function enhance cell apoptosis in lung adenocarcinoma. Tumor
of miR-483-3p on squamous cell carcinomas due to its Biol 37, 5193–5202.
pro-apoptotic properties. Cell Cycle 12, 2183–2193. 177 Marin-Muller C, Li D, Bharadwaj U, Li M, Chen C,
165 Li X, Li D, Wang A, Chu T, Lohcharoenkal W, Hodges SE, Fisher WE, Mo Q, Hung M-C & Yao Q
Zheng X, Gr€ unler J, Narayanan S, Eliasson S, Herter (2013) A tumorigenic factor interactome connected
EK et al. (2017) MicroRNA-132 with therapeutic through tumor suppressor MicroRNA-198 in human
potential in chronic wounds. J Invest Dermatol 137, pancreatic cancer. Clin Cancer Res 19, 5901–5913.
2630–2638. 178 Wang M, Wang J, Kong X, Chen H, Wang Y, Qin
166 Lian R, Lu B, Jiao L, Li S, Wang H, Miao W & Yu M, Lin Y, Chen H, Xu J, Hong J et al. (2014) MiR-
W (2016) MiR-132 plays an oncogenic role in 198 represses tumor growth and metastasis in
laryngeal squamous cell carcinoma by targeting colorectal cancer by targeting fucosyl transferase 8. Sci
FOXO1 and activating the PI3K/AKT pathway. Eur J Rep 4, 6145.
Pharmacol 792, 1–6. 179 Kudo-Saito C, Fuwa T, Murakami K & Kawakami Y
167 Kaushik SB & Kaushik N (2016) Non-coding RNAs (2013) Targeting FSTL1 prevents tumor bone
in skin cancers: an update. Non-coding RNA Res 1, metastasis and consequent immune dysfunction.
83–86. Cancer Res 73, 6185–6193.
168 Kretz M, Siprashvili Z, Chu C, Webster DE, Zehnder 180 Bae W-J, Lee S-H, Rho Y, Koo B-S & Lim Y-C
A, Qu K, Lee CS, Flockhart RJ, Groff AF, Chow J (2016) Transforming growth factor b1 enhances
et al. (2013) Control of somatic tissue differentiation stemness of head and neck squamous cell carcinoma
by the long non-coding RNA TINCR. Nature 493, cells through activation of Wnt signaling. Oncol Lett
231–235. 12, 5315–5320.
169 Kretz M, Webster DE, Flockhart RJ, Lee CS, 181 Yang W, Wu Y, Wang C, Liu Z, Xu M & Zheng X
Zehnder A, Lopez-Pajares V, Qu K, Zheng GXY, (2017) FSTL1 contributes to tumor progression via
Chow J, Kim GE et al. (2012) Suppression of attenuating apoptosis in a AKT/GSK-3 b - dependent
progenitor differentiation requires the long noncoding manner in hepatocellular carcinoma. Cancer
RNA ANCR. Genes Dev 26, 338–343. Biomarkers 20, 75–85.
170 Lee CS, Mah A, Aros CJ, Lopez-Pajares V, Bhaduri 182 Jin X, Nie E, Zhou X, Zeng A, Yu T, Zhi T, Jiang K,
A, Webster DE, Kretz M & Khavari PA (2018) Wang Y, Zhang J & You Y (2017) Fstl1 promotes
Cancer-associated long noncoding RNA SMRT-2 glioma growth through the BMP4/Smad1/5/8 signaling
controls epidermal differentiation. J Invest Dermatol pathway. Cell Physiol Biochem 44, 1616–1628.
138, 1445–1449. 183 Chi-Chung Lau M, Ng KY, Wong TL, Tong M, Lee
171 Keniry A, Oxley D, Monnier P, Kyba M, Dandolo L, TK, Ming XY, Law S, Lee NP, Cheung AL, Qin YR
Smits G & Reik W (2012) The H19 lincRNA is a et al. (2017) FSTL1 promotes metastasis and
developmental reservoir of miR-675 that suppresses chemoresistance in esophageal squamous cell
growth and Igf1r. Nat Cell Biol 14, 659–665. carcinoma through NFjB-BMP signaling cross-talk.
172 Kallen AN, Zhou XB, Xu J, Qiao C, Ma J, Yan L, Cancer Res 77, 5886–5899.
Lu L, Liu C, Yi JS, Zhang H et al. (2013) The 184 Domazet-Lo#so T, Klimovich A, Anokhin B, Anton-
imprinted H19 LncRNA antagonizes Let-7 Erxleben F, Hamm MJ, Lange C & Bosch TCG
MicroRNAs. Mol Cell 52, 101–112. (2014) Naturally occurring tumours in the basal
173 Raveh E, Matouk IJ, Gilon M & Hochberg A (2015) metazoan Hydra. Nat Commun 5, 4222.
The H19 Long non-coding RNA in cancer initiation, 185 Gonz" arez E, Samper E, Ram"ırez A, Flores JM,
alez-Su"
progression and metastasis – a proposed unifying Mart"ın-Caballero J, Jorcano JL & Blasco MA (2001)
theory. Mol Cancer 14, 184. Increased epidermal tumors and increased skin wound
174 Tao S-C, Rui B-Y, Wang Q-Y, Zhou D, Zhang Y & healing in transgenic mice overexpressing the catalytic
Guo S-C (2018) Extracellular vesicle-mimetic subunit of telomerase, mTERT, in basal keratinocytes.
nanovesicles transport LncRNA-H19 as competing EMBO J 20, 2619–2630.

18 The FEBS Journal (2018) ª 2018 Federation of European Biochemical Societies


G. M. Sundaram et al. Parallels between wound healing and cancer

186 Squarize CH, Castilho RM, Bugge TH & Gutkind JS 192 Govindan R, Behnken D, Read W & McLeod H
(2010) Accelerated wound healing by mTOR (2003) Wound healing is not impaired by the
activation in genetically defined mouse models. PLoS epidermal growth factor receptor-tyrosine kinase
One 5, e10643. inhibitor gefitinib [4]. Ann Oncol 14, 1330–1331.
187 Blagosklonny MV (2011) Molecular damage in cancer: 193 Dean NR, Sweeny L, Harari PM, Bonner JA, Jones
an argument for mtor-driven aging. Aging (Albany, V, Clemons L, Geye H & Rosenthal EL (2011)
NY) 3, 1130–1141. Wound healing following combined radiation and
188 Sanchez-Laorden B, Viros A, Girotti MR, Pedersen cetuximab therapy in head and neck cancer patients.
M, Saturno G, Zambon A, Niculescu-Duvaz D, J Wound Care 20, 166–170.
Turajlic S, Hayes A, Gore M et al. (2014) BRAF 194 Bodnar RJ (2014) Anti-angiogenic drugs: involvement
inhibitors induce metastasis in RAS mutant or in cutaneous side effects and wound-healing
inhibitor-resistant melanoma cells by reactivating complication. Adv Wound Care 3, 635–646.
MEK and ERK signaling. Sci Signal 7, ra30. 195 Gordon CR, Rojavin Y, Patel M, Zins JE, Grana G,
189 Escuin-Ordinas H, Li S, Xie MW, Sun L, Hugo W, Kann B, Simons R & Atabek U (2009) A review on
Huang RR, Jiao J, de-Faria FM, Realegeno S, bevacizumab and surgical wound healing: an
Krystofinski P et al. (2016) Cutaneous wound healing important warning to all surgeons. Ann Plast Surg 62,
through paradoxical MAPK activation by BRAF 707–709.
inhibitors. Nat Commun 7, 12348. 196 Cheon EC, Small W, Strouch MJ, Krantz SB,
190 Payne WG, Naidu DK, Wheeler CK, Barkoe D, Rademaker A, Mulcahy MF, Benson AB, Bentrem DJ
Mentis M, Salas RE, Smith DJ & Robson MC (2008) & Talamonti MS (2010) The effects of bevacizumab
Wound healing in patients with cancer. Eplasty 8, e9. on postoperative complications in patients undergoing
191 Kamil N, Kamil S, Ahmed SP, Ashraf R, Khurram M colorectal and pancreatic cancer resection. J Surg
& Ali MO (2010) Toxic effects of multiple anticancer Oncol 102, 539–542.
drugs on skin. Pak J Pharm Sci 23, 7–14.

The FEBS Journal (2018) ª 2018 Federation of European Biochemical Societies 19

View publication stats

You might also like