You are on page 1of 21

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

net/publication/260197003

Dynamics of Transforming Growth Factor Beta Signaling in Wound Healing and


Scarring

Article  in  Advances in Wound Care · June 2013


DOI: 10.1089/wound.2013.0429 · Source: PubMed

CITATIONS READS
184 19,961

4 authors, including:

Kenneth W Finnson Sarah Mclean


McGill University The University of Western Ontario
53 PUBLICATIONS   1,735 CITATIONS    16 PUBLICATIONS   567 CITATIONS   

SEE PROFILE SEE PROFILE

Anie Philip
McGill University
116 PUBLICATIONS   2,450 CITATIONS   

SEE PROFILE

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

CD109 in cancer View project

Regulation of the blood-epididymis barrier View project

All content following this page was uploaded by Kenneth W Finnson on 16 July 2014.

The user has requested enhancement of the downloaded file.


COMPREHENSIVE INVITED REVIEW

Dynamics of Transforming Growth Factor Beta


Signaling in Wound Healing and Scarring

Kenneth W. Finnson,1 Sarah McLean,2


Gianni M. Di Guglielmo,2 and Anie Philip1,*
1
Division of Plastic Surgery, Department of Surgery, Montreal General Hospital, McGill University, Montreal, Canada.
2
Department of Physiology and Pharmacology, Western University, London, Canada.

Significance: Wound healing is an intricate biological process in which the


skin, or any other tissue, repairs itself after injury. Normal wound healing
relies on the appropriate levels of cytokines and growth factors to ensure that
cellular responses are mediated in a coordinated manner. Among the many
growth factors studied in the context of wound healing, transforming growth
factor beta (TGF-b) is thought to have the broadest spectrum of effects. Anie Philip, PhD
Recent Advances: Many of the molecular mechanisms underlying the TGF-b/
Smad signaling pathway have been elucidated, and the role of TGF-b in wound Submitted for publication March 22, 2013.
*Correspondence: Division of Plastic Surgery,
healing has been well characterized. Targeting the TGF-b signaling pathway Montreal General Hospital, 1650 Cedar Avenue,
using therapeutic agents to improve wound healing and/or reduce scarring has Room C9-158, Montreal, QC H3G 1A4, Canada
been successful in pre-clinical studies. (e-mail: anie.philip@mcgill.ca).

Critical Issues: Although TGF-b isoforms (b1, b2, b3) signal through the same
cell surface receptors, they display distinct functions during wound healing
Abbreviations
in vivo through mechanisms that have not been fully elucidated. The challenge and Acronyms
of translating preclinical studies targeting the TGF-b signaling pathway to a
ALK = activin-like receptor
clinical setting may require more extensive preclinical research using animal
kinase
models that more closely mimic wound healing and scarring in humans, and
AMH = anti-müllerian hormone
taking into account the spatial, temporal, and cell-type–specific aspects of
BMP = bone morphogenetic
TGF-b isoform expression and function. protein
Future Directions: Understanding the differences in TGF-b isoform signaling CM = conditioned medium
at the molecular level and identification of novel components of the TGF-b ECM = extracellular matrix
signaling pathway that critically regulate wound healing may lead to the EMT = epithelial-to-mesenchymal
discovery of potential therapeutic targets for treatment of impaired wound transition
healing and pathological scarring. GDF = growth and differentiation
factor
GS = glycine/serine
SCOPE AND SIGNIFICANCE differentiation, extracellular matrix HTS = hypertrophic scar
Wound healing is a complex (ECM) production, and immune IFN = interferon
physiological response to injury and modulation.1 The present review fo- I-Smad = inhibitory Smad
involves three main overlapping cuses on the current state of knowl- LAP = latency-associated
phases: inflammation, proliferation, edge on the TGF-b signaling peptide
and maturation.1 Among the many pathway and the approaches that LLC = large latent complex
cytokines and growth factors in- have been used to manipulate this LOX = lysyl oxidase
volved in wound healing, transform- pathway to improve wound healing LTBP = latent TGF-b binding
ing growth factor beta (TGF-b) has and reduce scarring. protein
the broadest spectrum of effects.1 MH = Mad homology
TRANSLATIONAL RELEVANCE MIS = müllerian inhibiting
TGF-b plays an essential role in
substance
wound healing through its pleiotro- Research on understanding the
(continued)
pic effects on cell proliferation and molecular mechanisms by which

ADVANCES IN WOUND CARE, VOLUME 2, NUMBER 5


Copyright ª 2013 by Mary Ann Liebert, Inc. DOI: 10.1089/wound.2013.0429
j 195
196 FINNSON ET AL.

TGF-b signaling regulates wound motes wound closure and resolution


Abbreviations healing have led to the development through the production of ECM pro-
and Acronyms (continued) and use of therapeutic agents that teins and the inhibition of matrix
modulate TGF-b signaling. Ther- metalloproteinases (MMPs). How-
MMP = matrix metalloproteinase
apeutic agents tested in wound ever, in fibrotic diseases, excessive
P144 = peptide 144
rSmad = receptor regulated
healing and scarring models include TGF-b production and signaling pro-
Smads small-molecule inhibitors of the type motes extensive tissue fibrosis, which
SLC = small latent complex I TGF-b receptor (activin-like recep- can compromise normal tissue func-
Smurf = Smad ubiquitination tor kinase 5 [ALK-5]), anti–TGF-b tion.3 Understanding the molecular
regulatory factor neutralizing antibodies, and re- mechanisms involved in regulating
SSc = systemic sclerosis combinant TGF-b3 protein. These TGF-b signaling during wound
sTbRII = soluble TbRII molecules have shown promise in healing and scarring may provide
TbRI, TbRII = TGF-b receptor pre-clinical studies, but none have important insights into how its dys-
types I and II yet been approved by the U.S. Food & regulation may contribute to im-
TIMP = tissue inhibitor of Drug Administration for clinical use. paired wound healing or abnormal
MMPs scarring.
TGF-b = transforming growth
factor beta CLINICAL RELEVANCE TGF-b superfamily members. The
TSP-1 = thrombospondin 1 TGF-b superfamily consists of struc-
TGF-b regulates almost all aspects
WT = wild-type turally and functionally related cy-
of wound healing, and aberrant
TGF-b signaling has been implicated tokines that signal through a pair of
in pathological skin disorders, in- transmembrane serine-threonine ki-
cluding chronic wounds and exces- nase receptors known as the type I
sive scarring. Targeting the TGF-b and type II receptors (TbRI [also
signaling pathway represents a via- known as ALK] and TbRII, respec-
ble strategy for the development of tively), which, in turn, activate in-
novel therapeutic agents that im- tracellular Smad transcription
prove wound healing and reduce factors to mediate downstream sig-
pathological scarring. naling events. The TGF-b superfam-
ily contains > 30 ligands, which are
structurally characterized by the
DISCUSSION OF FINDINGS presence of a cysteine-knot motif.4
AND RELEVANT LITERATURE This superfamily is divided into two
Overview of TGF-b signaling subfamilies, the bone morphogenetic
The TGF-b signaling pathway is protein (BMP)/growth and differ-
essential for numerous cell functions entiation factor (GDF)/müllerian-
and was thought to arise with the inhibiting substance (MIS) subfamily
development of metazoans. In devel- and the TGF-b/activin/Nodal sub-
opment, TGF-b plays numerous roles, family (Fig. 1).5 These two sub-
including induction of epithelial-to- families are classified partially on the
mesenchymal transition (EMT) in basis of which ALK they bind as well
endocardial cells, which is necessary as the subset of Smad transcription
for normal heart development.1,2 factors they activate. The BMP/GDF/
TGF-b also has several roles in nor- MIS subfamily generally binds to
mal tissue homeostasis, regulating ALK1, -2, -3, or -6, and activates the
diverse functions such as cellular dif- receptor-regulated Smads (rSmads)
ferentiation, apoptosis, cell-cycle ar- Smad-1, -5, and -8; whereas the TGF-
rest, ECM production, and cellular b/activin/Nodal subfamily binds to
migration. Partly owing to its pleio- ALK4, -5, and -7, and activates the
tropic effects in numerous cell types, rSmads Smad-2 and -3.6 In addition
TGF-b has also been implicated in to the rSmads, another class of
several pathologies, including fibro- Smads known as the inhibitory
sis. In wound healing, TGF-b pro- Smads (I-Smads), Smad-6 and -7 act
TGF-b IN WOUND HEALING 197

Figure 1. Members of the TGF-b superfamily and their signaling components. TGF-b superfamily members include TGF-b (-b1, -b2, and -b3), activin, nodal,
BMPs (-2, -4, and -7), AMH/MIS, and GDF-5. TGF-b superfamily members signal through a unique pair of transmembrane serine-threonine kinases known as the
type I and type II receptors to mediate intracellular Smad signaling. The TGF-b/activin/Nodal subfamily binds to ALK 4, 5, and 7 and activates Smads 2 and 3;
whereas the BMP/GDF/MIS subfamily generally binds to ALK 1, 2, 3, or 6 and activates Smads 1, 5, and 8. Activated Smad2/3 and Smad1/5/8 form a complex
with Smad4 and enter the nucleus, where they regulate target gene expression. Accessory or co-receptors (betaglycan, endoglin, CD109, and cripto) are
potent modulators of signaling by TGF-b superfamily members. TGF-b, transforming growth factor beta; BMP, bone morphogenetic protein; AMH, anti-müllerian
hormone; MIS, Müllerian inhibiting substance; GDF, growth and differentiation factor; ALK, activin-like receptor kinase. To see this illustration in color, the
reader is referred to the web version of this article at www.liebertpub.com/wound

as adaptor proteins that bind Smad ubiquitin reg- as a signal to provide precursor cells during muscle
ulatory factor (Smurfs)-1 and Smurf-2, which ubi- repair.
quitinate the receptor complex, targeting it for GDFs are also known for their roles in tissue
degradation via proteosomal and lysosomal path- development. For example, GDF-8 (also known as
ways. myostatin) is a negative regulator of skeletal
The BMP/GDF/MIS subfamily is best known for muscle growth and has been implicated in the
its role in bone development and regeneration and maintenance of whole body homeostasis.10 Finally
consists of *20 cytokines.4 BMPs are so called due MIS, also known as anti-müllerian hormone, in-
to their ability to induce osteoblast differentiation hibits development of the müllerian ducts during
and promote bone formation.4 While the majority of embryogenesis and appears to have a negative
BMP ligand gene knockout mice are non-viable and regulatory role in follicular development in the
die during embryogenesis, mice lacking BMP type I adult ovary.11
receptor A (BMPR-IA) are viable and have appen- The TGF-b/activin/Nodal subfamily has been
dicular skeletal defects.7 Although BMPs are studied in-depth due to its roles in tissue morpho-
known to inhibit myogenic (muscle cell) differen- genesis, cancer, and wound healing. It consists of
tiation and promote osteogenic differentiation five activins/inhibins, Nodal, and three TGF-bs
during embryonic development, emerging evidence (TGF-b1, TGF-b2, and TGF-b3).6 While the acti-
indicates that BMPs have pro-proliferative effects vins were initially discovered due to their ability to
on muscle progenitor (satellite) cells during adult induce follicle stimulating hormone release, it has
skeletal muscle regeneration after injury.8 For ex- now been appreciated that activins, particularly
ample, administering BMP4 to mice that have activin A, play a role in wound healing.12 Activin is
undergone muscle injury reduces differentiation of up-regulated during cutaneous wound healing in
satellite cells and sustains their proliferative po- humans.13 Keratinocytes, fibroblasts, and inflam-
tential.9 Therefore, it seems that BMPs are not matory cells appear to be sources of activin in the
only implicated in fracture healing, but can also act wound microenvironment.12,13 Further, transgenic
198 FINNSON ET AL.

mice overexpressing activin A in the epidermis ligands have different effects during wound heal-
display enhanced wound healing with increased ing and scarring. For example, TGF-b1 is found at
granulation tissue formation as compared with very high levels in the wound microenvironment
wild-type (WT) littermates;14 whereas epidermal and promotes myofibroblast differentiation, pro-
overexpression of the activin antagonist follistatin duction of ECM components, and fibroblast che-
is associated with impaired wound healing and motaxis (reviewed by Ferguson and O’Kane).26
reduced granulation tissue formation.15 A critical Overall, TGF-b1 promotes the formation of a scar
role for endogenous activin in wound healing was during adult wound healing. On the other hand,
also shown in another study where blocking follis- the embryonic wound microenvironment contains
tatin expression in the epidermis led to enhanced high levels of TGF-b3 and low levels of TGF-b1.26
re-epithelialization without affecting the quality of Further, adding exogenous TGF-b3 to an adult
the healed wound.16 wound promotes scar-free healing in rats,27 and
Similar to other TGF-b superfamily members, injuries obtained in utero heal scar free possibly
Nodal is critical for proper early development and due to the relatively high levels of TGF-b3 com-
is best known for its role in inducing mesoderm and pared with TGF-b1.26
endoderm formation.17 Nodal is also essential for
maintaining human embryonic stem cell plur- Activation of latent TGF-b. TGF-b is synthe-
ipotency, which may explain its frequent over- sized as a homo-dimeric proprotein (pro-TGF-b)
expression in a number of human cancers.18 and undergoes proteolytic cleavage in the trans-
However, Nodal is generally not found in adult Golgi network by furin-like enzymes, giving rise to
tissues, and its potential role in wound repair is not a C-terminal mature TGF-b dimer and N-terminal
known.18 pro-peptide known as latency-associated peptide
While the role of other TGF-b superfamily (LAP). LAP remains non-covalently associated
members in wound healing is still being investi- with mature TGF-b, rendering TGF-b inactive in a
gated, the TGF-b subfamily, consisting of TGF-b1, so-called small latent complex (SLC) (Fig. 2). In
2, and 3, has long been appreciated as having most cases, the SLC forms a complex with another
critical roles in wound healing, and will be the focus protein called latent TGF-b binding protein (LTBP)
of the remainder of this review. via intermolecular disulfide bonds, giving rise to
The TGF-b isoforms (-b1, -b2, and -b3) have been the large latent complex (LLC), the most abundant
conserved throughout evolution, and orthologs secreted form. The LLC can associate with the
to human TGF-b can be found in Drosophila ECM by covalent cross-linking of LTBP with ECM
melanogaster and Xenopus laevis (reviewed by Wu proteins (Fig. 2).28,29 Latent TGF-b can be acti-
and Hill).2 The TGF-b ligands share a significant vated in vivo by molecules such as thrombospondin
sequence homology. Together, they have > 76% 1 (TSP-1), integrins, MMPs, and plasmin, and
identity in their active domains.19,20 Despite in vitro by acidic or alkali conditions, heat dena-
structural similarities, TGF-b ligands have distinct turation, or shear stress.30–33 All three TGF-b iso-
affinities for TGF-b receptors. The three TGF-b li- forms exist in latent complexes34,35 and, in their
gands are produced by a number of different cell active forms, exist as homodimers that are stabi-
types, and the production of all three occurs during lized by disulfide bridges and hydrophobic inter-
development, although TGF-b1 is the predominant actions.
type in adults.21,22 Each TGF-b ligand has rela- It has been suggested that activation of latent
tively specific, non-overlapping functions in vivo. TGF-b occurs at two time points during wound
Mice containing deletions of these genes illustrate healing: immediately after wounding, and during
that these ligands have distinct functional roles. re-epithelialization.32 Platelets are thought to be
For example, Tgfb1 - / - mice develop significant the primary source of activated TGF-b immediately
problems in utero, including vasculogenic and after injury. Of the total TGF-b that is released by
hematopoietic defects.23 Mice that survive gesta- platelets, only a small amount is activated, and it is
tion develop a severe wasting inflammatory thought that the rest of it may remain in its SLC
syndrome.23 Tgfb2 - / - mice have a myriad of de- form.32 Platelet-derived latent TGF-b can be acti-
velopmental defects, including skeletal, cardio- vated by TSP-1, which is also contained in the
vascular, pulmonary, and visual problems.24 platelet secretory granules,36 as well as by the fi-
Interestingly, Tgfb3 - / - null mice have the least brinolytic enzyme plasmin during dissolution of
defects and die after birth due to an inability to the blood clot.37 The activated TGF-b then acts as a
suckle caused by cleft palate.25 Similar to their potent chemoattractant for inflammatory cells that
non-redundant roles in development, the TGF-b invade the wound microenvironment, leading to
TGF-b IN WOUND HEALING 199

latent TGF-b activation may represent an impor-


tant regulatory mechanism that controls TGF-b
bioavailability and action during wound healing.

TGF-b/Smad signaling pathway. As mentioned


earlier, TGF-b signaling is mediated by a pair of
transmembrane serine-threonine kinase receptors
known as TbRI (or ALK) and TbRII. TbRII is a
62 kDa protein containing a short cysteine-rich,
N-glycosylated extracellular domain, a single trans-
membrane domain, and a serine-threonine kinase
intracellular domain.39 The cytoplasmic domain of
TbRII is also serine-threonine rich, which is lack-
ing in TbRI.40 At the cell surface, TbRII exists as a
homodimer in the absence and presence of ligand.41
TbRII binds TGF-b1 and TGF-b3 with a relatively
high affinity,42,43 but is unable to bind TGF-b2,
which requires the presence of a TGF-b coreceptor
known as betaglycan (TbRIII).39 In the absence of
ligands, TbRII undergoes autophosphorylation on
serine residues Ser549, Ser551, Ser223, Ser226, and
Ser227.43,44 In response to TGF-b binding, TbRII
forms a heterotetrameric complex comprising two
pairs of TbRII and TbRI. TbRII then phosphory-
lates TbRI at serine-threonine residues in its gly-
cine/serine (GS) domain, leading to the activation
of TbRI, which, in turn, phosphorylates and acti-
vates intracellular Smad2 and Smad3 proteins,
which are central mediators of TGF-b signaling.45
Activated Smad-2 and -3 then interact with the co-
Smad (Smad4) and enter the nucleus to regulate
gene transcription (Fig. 3).
TbRI and TbRII are structurally similar, al-
though TbRI contains a shorter extracellular do-
Figure 2. Latent and active forms of TGF-b ligand. TGF-b is synthesized as
main and cannot bind ligands in the absence of
a homo-dimeric proprotein (pro–TGF-b) and undergoes proteolytic cleavage TbRII.46 Akin to TbRII, TbRI also exists as a
in the trans-Golgi network by furin-like enzymes, giving rise to the mature homodimer at the cell surface and contains a
TGF-b dimer and its pro-peptide, also known as LAP. TGF-b is secreted serine/threonine kinase intracellular domain.39
either as an SLC, which comprises the mature TGF-b dimer in association
with LAP, or as an LLC in which the LAP portion of SLC is covalently linked
However, TbRI contains a unique intracellular
to a protein known as latent TGF-b binding protein (LTBP). (A) SLC: The GS-rich region that is phosphorylated by TbRII.19
mature TGF-b dimer (red) is non-covalently associated with its LAP (green). Once phosphorylated, the GS domain of TbRI acts
*Asterisks indicate the regions that have undergone proteolytic processing as a docking platform for the so-called receptor-
by furin-like enzymes in the trans-Golgi before secretion. (B) LLC: The SLC
is covalently linked to latent TGF-b binding protein (LTBP, yellow) by disulfide
regulated Smad proteins, Smad-2 and -3 (Fig. 3).47
bonds to form the LLC. (C) Mature TGF-b dimer: The mature TGF-b dimer is Mutations of the GS domain have highlighted the
released from the latent complex by different mechanisms, giving rise to the importance of this region to TGF-b signal trans-
active form of TGF-b that can bind its receptors and elicit biological re- duction; mutations of two or more glycine or serine
sponses. LAP, latency associated peptide; SLC, small latent complex; LLC,
large latent complex. To see this illustration in color, the reader is referred to
residues in the GS domain impair TGF-b signaling
the web version of this article at www.liebertpub.com/wound activity.48 Mutation of threonine 204 to aspartic
acid increases TGF-b signal transduction in the
absence of ligands, as it generates a constitutively
further activation of latent TGF-b during the early active TbRI.48 These mutational studies confirm
re-epithelialization phase. Macrophages recruited that TbRI is the key player in Smad signal trans-
to the wound microenvironment secrete plasmin- duction.
ogen activators, which can also activate latent Smad proteins typically consist of two domains
TGF-b.38 Overall, the spatiotemporal control of that are separated by a variable linker region. The
200 FINNSON ET AL.

Figure 3. TGF-b/Smad signal transduction pathway. TGF-b signaling is initiated when the TGF-b ligand binds to the extracellular domain of TbRII. TGF-b1 and
TGF-b3 bind TbRII with a high affinity, whereas TGF-b2 requires the TGF-b co-receptor betaglycan (TbRIII) to ‘‘present’’ it to TbRII. TGF-b-associated TbRII then
recruits TbRI, resulting in the formation of a heterotetrameric receptor signaling complex comprising one TGF-b ligand, one homo-dimeric TbRII, and one homo-
dimeric TbRI. TbRII is a constitutively active kinase that phosphorylates TbRI, resulting in activation of TbRI kinase activity. TbRI then phosphorylates intracellular
Smad2 and Smad3 proteins, which, in turn, form a complex with Smad4 and enter the nucleus to regulate gene transcription. TbRII, type II TGF-b receptor. To see
this illustration in color, the reader is referred to the web version of this article at www.liebertpub.com/wound

amino Mad homology (MH) 1 domain has DNA TGF-b coreceptors. In addition to type I and
binding capabilities in some Smad sub-types, while type II TGF-b signaling receptors, there are three
the carboxy MH2 domain has been shown to me- TGF-b coreceptors: betaglycan (TbRIII), endoglin,
diate interactions with a variety of proteins.49 The and the recently discovered CD109. They play im-
activated GS domain of TbRI serves as a docking portant roles in modulating TGF-b signaling and
site for Smad2 and Smad3 via their MH1 do- are considered accessory receptors, as they have no
mains.47 TbRI phosphorylates rSmads on the con- signaling or enzymatic activity. Endoglin and be-
served SSXS motif located at the C-termini of taglycan are structurally related, with large,
Smads 2 and 3 (e.g., serine residues 465 and 467 in heavily glycosylated extracellular domains, and a
the MH2 domain of Smad2).50–52 The phosphory- short cytoplasmic region with high sequence simi-
lated serine residues of Smad2/3 serve as a docking larity.55–57 Both receptors can be phosphorylated
site for Smad4, promoting the dissociation of on serine/threonine residues in their cytoplasmic
Smad2/3 from TbRI and the formation of a het- domain.58–60 At the cell surface, endoglin and be-
eromeric complex with Smad4.50,53 Smad2/3 are taglycan form homodimers,61,62 as well as form
generally located cytoplasmically in the absence of complexes with TbRI and TbRII.59,63 Though
ligands, but on ligand stimulation, they translocate structurally similar, these co-receptors differ in
to the nucleus through its interaction with Smad4,5 their ligand-binding ability and expression. Beta-
which is able to bind with nucleoporins; the inter- glycan can bind all three TGF-b isoforms with a
action of Smad4 with the nucleoporin importin-1a high affinity,62 whereas endoglin binds TGF-b1
is thought to mediate the translocation of the Smad and TGF-b3 in the presence of TbRII, but does not
heteromeric complex to the nucleus.54 In the nu- bind TGF-b2.58,59 Betaglycan is widely expressed
cleus, the heteromeric Smad complex binds to in adult and fetal tissues,39 whereas endoglin is
promoters or enhancers of TGF-b target genes,such thought to be primarily expressed in proliferating
as the Smad binding element via its MH1 domain, endothelial cells,64 although recent studies show
and interacts with transcriptional co-activators its expression in other cell types such as chon-
and co-repressors in order to induce cell-specific drocytes65–67 and skin fibroblasts.68,69 Betaglycan
transcriptional programs.5,19 is thought to facilitate TGF-b signaling by
TGF-b IN WOUND HEALING 201

‘‘presenting’’ the TGF-b ligand to TbRII, whereas docytosis and membrane-raft dependent endocy-
endoglin inhibits TGF-b/ALK5/Smad signaling, tosis.77 Current evidence indicates that distinct
although other mechanisms also exist.61 endocytic pathways regulate TGF-b receptor sig-
Another TGF-b coreceptor, CD109, has been naling and turnover. At the cell surface, TGF-b
shown to bind TGF-b ligand and inhibit TGF-b receptor complexes can access both clathrin-coated
signal transduction.70–74 CD109 is a glycopho- pits and membrane rafts (Fig. 4).78 Inhibition of
sphatidylinositol-anchored protein that binds clathrin-coated pit internalization through the use
TGF-b1 with a high affinity and also forms a com- of a dominant-negative epidermal growth factor
plex with TbRI, TbRII, and betaglycan.70,74 Work substrate 15 (Eps15) mutant shifts receptors into
by Bizet et al. demonstrated that the association of membrane raft fractions; similarly, inhibition of
CD109 with the TGF-b receptor complex increases membrane raft formation through cholesterol de-
the internalization of the receptors via caveolae pletion shifts receptors back into non-membrane
and enhances receptor degradation.75 In a follow- raft fractions.78 TGF-b receptors internalized via
up article, Bizet et al. illustrated that CD109 clathrin-mediated endocytosis access the early en-
promotes TbRI degradation in a Smad7/Smurf2- dosome, a signaling endosome, which propagates
dependent manner.76 TGF-b signal transduction through the recruit-
ment of Smad2 and Smad3.78,79 Membrane raft
Endocytosis and TGF-b signaling. Endocytosis endocytosis of TGF-b receptors, however, results in
refers to the process in which cell-surface associ- receptors being targeted to the caveolin-1 positive
ated molecules enter the cell without passing vesicle.80 Unlike the early endosome, the caveolin-
through the plasma membrane. Essentially, the 1 positive vesicle promotes association of Smad7,
plasma membrane invaginates, budding off and not Smad2/3, with the receptor complex,78 with
forming a vesicle containing the internalized cargo. Smad7 acting as an adaptor that binds Smurf-2,
There are several methods of endocytosis of cell- which ubiquitinates the receptor complex while
surface receptors, including clathrin-mediated en- targeting it for degradation78,81,82 (Fig. 4).

Figure 4. Regulation of TGF-b signaling by clathrin-dependent and -independent endocytosis. TGF-b receptors can be internalized by clathrin-dependent and
clathrin-independent, membrane-raft dependent mechanisms. TGF-b receptors internalized via clathrin-coated pit mediated endocytosis traffic to Smad anchor
for receptor activation (SARA)-containing early endosome and propagate signal transduction. TGF-b receptors in the early endosome can be recycled back to
the plasma membrane in a Rab11-dependent manner. TGF-b receptors internalized by membrane-raft dependent endocytosis traffic to caveolin-1 positive
vesicles, where they are targeted for Smad7/Smurf2-mediated ubiquitination and proteosomal/lysosomal degradation. Other potential intracellular trafficking
pathways for TGF-b receptors, including bi-directional trafficking between the early endosome and caveolin-1 positive vesicles (intermediate pathways) or
direct trafficking of TGF-b receptors from the early endosome to proteosomal/lysosomal degradation pathways, are current topics of investigation. Smurf,
Smad ubiquitination regulatory factor. To see this illustration in color, the reader is referred to the web version of this article at www.liebertpub.com/wound
202 FINNSON ET AL.

TGF-b and wound healing the wound site in response to TGF-b, differentiate
Role of TGF-b in cutaneous wound healing: an into activated macrophages (also in response to
overview. Wound healing is a complex and dy- TGF-b) that engulf and digest foreign particles and
namic physiological process that involves numer- necrotic debris, and release TGF-b and other
ous secreted cytokines and growth factors and the growth factors to stimulate capillary growth and
interaction of a variety of different cell types.1,83 initiate granulation tissue formation. Once im-
The three phases of wound healing are known as mune cells become activated, they are susceptible
inflammation, tissue formation (proliferation), and to TGF-b1-mediated suppression to reverse the
maturation (tissue remodeling), which overlap in inflammatory process.87 Thus, TGF-b plays a dual
time. TGF-b plays a critical role in regulating role in the inflammation phase of wound healing by
multiple cellular responses that occur in all three exerting pro-inflammatory effects during the early
phases of wound healing (Fig. 5). Hemostasis may stages and later contributing to the resolution of
be defined as the stoppage of bleeding after an in- inflammation.
jury and involves vasoconstriction, platelet aggre- The second phase of wound healing is known as
gation, and blood coagulation.84 Inflammation ‘‘tissue formation’’ or the proliferative phase. Dur-
ensues shortly thereafter and lasts for about 2–4 ing this phase, TGF-b orchestrates many cellular
days. Inflammation is characterized by the re- responses, including re-epithelialization as well as
cruitment of immune cells such as neutrophils and formation of new blood vessels (angiogenesis), fi-
macrophages to the injured site in response to broblast proliferation, and production of ECM
chemotactic cytokines such as TGF-b.85 Neu- components, leading to granulation tissue forma-
trophils cleanse the wounded area of foreign par- tion and wound contraction.88 TGF-b regulates
ticles, secrete chemicals to kill bacteria, and are wound angiogenesis by stimulating endothelial cell
then extruded with the eschar (scab) or phagocy- migration, differentiation, and capillary tubule
tosed by macrophages.86 Monocytes also infiltrate formation.89 TGF-b also promotes fibroblast trans-

Figure 5. Schematic diagram showing the role of TGF-b in regulating all three phases of the wound healing process. Inflammation: TGF-b acts as a potent
chemoattractant for immune cells (neutrophils and macrophages) during the early stages of inflammation, regulates immune cell function, and contributes to
resolution of inflammation. Proliferation: TGF-b promotes angiogenesis by stimulating endothelial cell migration, differentiation, and capillary tubule formation.
TGF-b also stimulates fibroblast proliferation, promotes fibroblast trans-differentiation into myofibroblasts, and stimulates ECM production. In addition, TGF-b
inhibits keratinocyte proliferation and enhances keratinocyte migration, promoting re-epithelialization. Maturation: TGF-b regulates the balance of ECM
synthesis and degradation by tightly controlling the production of ECM components and regulating their rate of degradation by modulating synthesis of MMPs
and production of protease inhibitors such as TIMPs. TGF-b also regulates ECM remodeling by stimulating production of LOXs, which play an important role in
collagen cross-linking. ECM, extracellular matrix; MMPs, matrix metalloproteinases; TIMPs, tissue inhibitors of matrix metalloproteinases; LOXs, lysyl oxidases.
TGF-b IN WOUND HEALING 203

Figure 6. Myofibroblasts originate from different cell types and play a critical role in wound healing and scarring. Myofibroblasts are specialized cells that
express alpha smooth muscle actin (a-SMA, red) and display a contractile phenotype. During wound healing, resident fibroblasts trans-differentiate into
myofibroblasts in response to TGF-b and promote wound contraction. Fibrocytes are circulating bone marrow-derived cells that can enter tissues and
differentiate into myofibroblasts in response to TGF-b. Myofibroblasts also originate from other cell types such as epithelial cells through epithelial-to-
mesenchymal transition (EMT) and perivascular cells (pericytes) by trans-differentiation, and these processes have been implicated in the pathogenesis of
hypertrophic scarring.

differentiation into myofibroblasts (Fig. 6), which collagen production, which is tightly regulated by
play a critical role in wound contraction.90 TGF-b1 TGF-b, the transition from the proliferative phase
has been shown to increase the expression ECM to maturation phase is dependent on the ongoing
components, including fibronectin, the fibronectin synthesis and degradation of collagen at a low
receptor, and collagens, and to reduce their deg- rate.86 Collagen degradation is mediated by MMPs
radation by down-regulating the expression and and other serine proteases that are also regulated
activity of matrix-degrading enzymes such as by TGF-b.93 Further, collagen degradation by
MMPs and increasing the expression of protease MMPs is counterbalanced by the presence of en-
inhibitors such as tissue inhibitors of MMPs dogenous TIMPs whose expression is also regu-
(TIMPs).91 Although TGF-b is a potent inhibitor of lated by TGF-b.94 Although the rate of collagen
epithelial (keratinocyte) cell proliferation in vitro, production and degradation are balanced during
TGF-b may have both pro- and antiproliferative the maturation phase, there continues to be an
effects in keratinocytes in vivo, depending on the extensive remodeling process in which disorga-
differentiation state and/or other factors such as nized collagen fibers are re-organized, cross-linked,
TGF-b concentration or the timing of its adminis- and aligned along tension lines.95 Cross-linking of
tration.91 In addition, TGF-b stimulates keratino- collagen is mediated by enzymes known as lysyl
cyte migration in vitro, possibly by regulating oxidases (LOXs), which have been shown to in-
integrin expression,92 which is thought to be im- crease collagen cross-link dependent contraction
portant for the migratory component of re-epithe- in vitro96 and enhance the wound’s tensile strength
lialization.87 Thus, TGF-b affects multiple cell in vivo.97,98 TGF-b has been shown to increase LOX
types during the proliferative phase of wound expression in many different cell types, including
healing, contributing to the formation of granula- skin fibroblasts in vitro,99,100 but its role in regu-
tion tissue consisting of newly formed blood ves- lating LOX expression during wound healing
sels, proliferating fibroblasts, and ECM in vivo remains to be explored.
components, which sets the stage for normal scar
formation. Analysis of the functional role of TGF-b in wound
The final stage of wound healing, known as healing using animal models. Much of what has
‘‘maturation’’ or the remodeling phase, can last for been learned about the role of TGF-b in wound
several years. Although the proliferation phase of healing has come from animal studies. These
wound healing is characterized by an increase in studies have used different approaches, including
204 FINNSON ET AL.

incisional and excisional wounds generated in exogenous application of recombinant TGF-b3


various animal species followed by measurement of protein or reducing TGF-b1 and TGF-b2 levels
TGF-b signaling pathway components, and treat- using neutralizing antibodies (see below).
ing wounds with recombinant TGF-b (-b1, -b2, and The potential for TGF-b to regulate wound
-b3) proteins or anti–TGF-b neutralizing anti- healing was first implicated in a study showing
bodies and measuring various wound healing and/ that a subcutaneous injection of TGF-b into normal
or scarring parameters. Another approach used (non-wounded) skin of new born mice caused
was to perform wound healing studies on mice with granulation tissue formation with the induction of
genetic modifications of specific components of the angiogenesis and collagen production.104 A subse-
TGF-b signaling pathway. This section highlights quent study showed that TGF-b administered to
the current knowledge on the role of TGF-b sig- rat incisional wounds accelerated wound healing
naling in wound healing in non–genetically modi- and increased wound strength.105 These studies
fied animal models. For information on the role of used TGF-b purified from human platelets and,
TGF-b signaling in wound healing obtained using therefore, could not distinguish which TGF-b iso-
mice with genetic modification in the TGF-b sig- form(s) was responsible for these effects. Sub-
naling pathway, the reader is referred to the ‘‘Cri- sequent studies showed that neutralization of
tical Review’’ by Finnson et al.,101 published in the TGF-b1 and TGF-b2 using anti-TGF-b1/2 neutral-
current issue of Advances in Wound Care. izing antibody, or exogenous addition of TGF-b3,
The relationship between endogenous TGF-b reduces cutaneous scarring in rat incisional
and wound healing first became evident in studies wounds without reducing the wound tensile
that measured TGF-b isoform levels in experi- strength.27,106,107 These studies were important in
mental wound healing models. For example, an establishing the pro-scarring effects of TGF-b1 and
early study investigating TGF-b1 expression and TGF-b2 and the antiscarring potential of TGF-b3.
activity in vivo using two different models of cuta- The role of TGF-b in wound healing and scarring
neous injury (human suction blister and partial was further examined by Thomas Mustoe’s group,
thickness excisional wounds in porcine skin) who investigated the effects of an anti-TGF-b1, 2,
showed that TGF-b1 expression/activity was in- and 3 monoclonal antibody on wound healing and
creased immediately (within 5 min) after injury hypertrophic scar (HTS) formation in rabbit ear
and progressed outward from the site of injury, wounds.108 They found that early treatment of the
particularly at the leading edge of the migrating wounds (days 0, 2, and 4 post-wounding) with
epithelial sheet.102 Another study showed that neutralizing antibody impaired wound healing
TGF-b isoforms (-b1, -b2, and -b3) display marked without decreasing scar hypertrophy, whereas as
differences in spatiotemporal expression during middle (days 7, 9, and 11) and late (days 11, 12, and
excisional wound repair in pigs, with TGF-b2 and 13) treatment of wounds significantly reduced scar
TGF-b3 expression becoming prevalent 24 h after hypertrophy.108 Although this study could not de-
wounding, particularly in the migrating epidermis, termine which TGF-b isoform(s) are involved, a
and TGF-b1 expression increasing later at 5 days follow-up study investigating the temporal ex-
post-wounding, when re-epithelialization was pression of TGF-b isoforms in the rabbit ear model
complete.103 All three TGF-b isoforms were de- showed that elevated levels of TGF-b1, and possi-
tected in the mesenchymal cells (dermis) and basal bly TGF-b2, are associated with HTS formation.109
lamina, suggesting their involvement in dermal– Another study using the rabbit ear model showed
epidermal interaction during wound healing.103 An that viral delivery of a dominant negative (trun-
intriguing aspect of TGF-b isoform expression cated) mutant of TbRII (to block endogenous TGF-b
comes from studies comparing their expression signaling) 8–12 days post-wounding decreased
profiles in embryonic (scar-free) and adult (scar hypertrophic scarring.110 The rabbit ear model has
forming) wounds. Embryonic wounds have been also provided insight on the role of TGF-b signaling
shown to express high levels of TGF-b3, produced during the earlier wound healing events. For ex-
mainly by skin cells (keratinocytes and fibroblasts), ample, one study showed that an injection of ade-
and low levels of TGF-b1 and TGF-b2; whereas novirus-containing Smad3 48 h before wound
adults wounds contain mostly TGF-b1 and TGF-b2 healing led to enhanced re-epithelialization and
derived from degranulating platelets and immune granulation tissue formation in rabbit ear wounds.111
cells.26 These findings prompted the hypothesis In addition, using the rabbit ear model under is-
that the ratio of TGF-b3 to TGF-b1 (or TGF-b2) is chemic conditions, it was shown that adenoviral
an important factor in scar-free wound healing and delivery of Smad3 enhanced re-epithelialization
led to studies aimed at increasing TGF-b3 levels by but that granulation tissue parameters were not
TGF-b IN WOUND HEALING 205

affected by Smad3 under ischemic conditions.112 cutaneous wound healing is beneficial for improv-
Further studies using TGF-b isoform–specific ing scarring outcome.
neutralizing antibodies and adenoviral delivery
of Smad2, Smad3, and/or Smad4 at the early (days Aberrant TGF-b signaling in pathological
0–4) and late (after day 7) stages of wound healing scarring: HTSs and keloids
in the rabbit ear model are possible avenues of Hypertrophic scarring. Hypertrophic scarring
future research to further elucidate the role of often occurs after deep burn injury or trauma and
TGF-b/Smad signaling in this valuable animal is characterized by excessive ECM deposition.115
model. HTSs appear as elevated red scar tissue that re-
Further evidence that decreasing TGF-b sig- mains within the boundary of the original injury
naling improves scarring outcome without com- (Fig. 7A).115 Although the pathophysiological
promising wound healing is provided by a recent mechanism(s) involved in hypertrophic scarring
study using transgenic mice overexpressing are not fully understood, many studies indicate
CD109, a TGF-b co-receptor and a potent TGF-b that aberrant TGF-b signaling plays a key role in
antagonist, in the skin. These mice display de- its etiology. For example, Scott and colleagues
creased inflammation and granulation tissue for- showed that TGF-b1 protein was present in HTSs,
mation and improved collagen architecture particularly in the deep dermis, as well as in the
without a compromise in wound tensile strength as dermis of mature ‘‘normal’’ scars but was not de-
compared with WT littermates, in a manner con- tected in normal (non-scarred) dermis.116 In addi-
sistent with inhibition of TGF-b signaling.113 Fur- tion, Wang and colleagues showed that TGF-b1
thermore, these mice also show a diminished mRNA levels are higher in HTS tissue and in cul-
fibrotic response in a bleomycin-induced skin fi- tured HTS fibroblasts as compared with normal
brosis model.114 Together, these studies support skin tissue and cells, and that cultured HTS fibro-
the notion that dampening TGF-b signaling during blasts secrete more TGF-b1 protein than normal

Figure 7. Characteristics of human hypertrophic scars and keloids. (A) Hypertrophic scar: This appears as a red, raised scar that does not extend beyond the
boundaries of the original injury. They have nodular collagen deposits containing a-SMA producing myofibroblasts that are involved in scar contracture.
Hypertrophic scars can regress with time. The main findings from studies on the role of TGF-b signaling and hypertrophic scarring are indicated. (B) Keloid:
This appears as a shiny and smooth protuberance ranging from pink to purple in color and extends beyond the boundaries of the original wound. Unlike
hypertrophic scars, keloids do not have nodular collagen deposits, a-SMA-producing myofibroblast, do not undergo scar contracture, and do not regress with
time. The main findings from studies on the role of TGF-b signaling and keloid formation are indicated. Images were obtained with permission from the DermNet
NZ Web site (www.dermnetnz.org). To see this illustration in color, the reader is referred to the web version of this article at www.liebertpub.com/wound
206 FINNSON ET AL.

skin fibroblasts.117 HTS skin tissue has been vascular space128 suggests that non-resident fi-
shown to display persistent ALK5 and TbRII ex- broblasts may play a more important role in
pression compared with normal wound tissue, scarring that previously thought.
where ALK5 and TbRII expression declines during Although TGF-b1 serum levels have been shown
the tissue remodeling phase.118 In addition, to be increased in patients with severe HTS re-
downstream TGF-b/Smad signaling appears to be sulting from burn injury, these findings have not
activated in HTS fibroblasts, as evidenced by a been universally reproduced. For example, a recent
predominantly nuclear localization of Smad2 in study on post-burn scarring in children showed
HTS skin fibroblasts as compared with normal fi- that although plasma TGF-b1 levels significantly
broblasts.119 The pleiotropic effects of TGF-b1 in increased during the first 2 weeks post-injury and
fibroblasts, including the induction of their differ- then declined in patients who healed with good
entiation into myofibroblasts, increasing ECM quality post-burn scars, the early increase in
production,90 and stimulating synthesis of TIMPs plasma TGF-b1 levels was not detected in patients
that inhibit ECM degradation by MMPs,120 con- who developed HTS.129 Further longitudinal
tribute to the formation of HTS. studies characterizing TGF-b1 serum levels in a
In addition to the effects of local TGF-b produc- larger cohort of burn patients will be needed to
tion by resident fibroblasts, alterations in systemic determine whether TGF-b1 serum levels can be
TGF-b levels have also been implicated in the used as an indicator for predicting clinical scar
pathogenesis of HTS. For example, Tredget et al. outcomes or whether they might serve as a clinical
showed that patients with severe HTS display in- tool for selecting patient groups to be targeted for
creased TGF-b1 serum levels after thermal injury anti-scarring therapy.
and that treatment with interferon (IFN)-a 2b
significantly reduced TGF-b1 serum levels and Keloids. Keloids are another type of patholog-
enhanced resolution of HTS.121 IFN-a 2b has been ical scar for which aberrant TGF-b signaling is
shown to inhibit TGF-b1 protein production by thought to play a pathophysiological role. Unlike
HTS fibroblasts,122 and other studies have shown HTS, keloids often appear as shiny rounded pro-
that IFNs increase expression of the I-Smad7,123 tuberances with colors ranging from pink to purple,
which has been shown to inhibit TGF-b signaling and scarring extends beyond the boundaries of the
by different mechanisms (see the section En- original injury (Fig. 7B).115 Studies using cultured
docytosis and TGF-b signaling). In addition, IFN-a keloid fibroblasts have shown that these cells pro-
2b has also been shown to inhibit TGF-b-induced duce higher amounts of ‘‘pro-scarring’’ TGF-b1 and
alpha smooth muscle actin expression in a unique TGF-b2 as compared with normal fibroblasts.130,131
subpopulation of leukocytes known as fibro- Another study demonstrated that keloid fibro-
cytes,124 which are circulating bone marrow de- blasts exhibit increased expression of ALK5 and
rived cells that can leave the blood, enter tissues, TbRII as well as increased phosphorylation of
and differentiate into myofibroblasts in response to Smad3 relative to normal fibroblasts.132 Interest-
TGF-b.125 Interestingly, dermal fibroblasts treated ingly, increased TGF-b/Smad3 signaling has been
with conditioned medium (CM) from burn patient implicated in keloid pathogenesis via epithelial–
fibrocytes (but not by CM from normal fibrocytes) mesenchymal interactions, where keloid keratino-
showed an increase in proliferation, migration, and cytes act through a paracrine mechanism to
contractility, which was abrogated by application increase ALK5 and TbRII expression and Smad3
of a TGF-b1 neutralizing antibody.126 These find- signaling in keloid fibroblasts.133 Genetic studies
ings suggest that fibrocytes in burn patients may have not revealed an association between keloid
act as a source of systemic TGF-b1 as well as a disease and the occurrence of common polymor-
target for TGF-b1-induced myofibroblast differen- phisms or mutations in genes encoding the three
tiation action in tissues, thereby creating a vicious TGF-b isoforms (-b1, -b2, and -b3) in Cauca-
cycle of TGF-b1 production and action in patho- sians134–136 or Smads (-3, -6, and -7) in a Jamaican
genesis of HTS. In addition, myofibroblasts origi- population.137 However, a recent study has re-
nating from other cell types, such as epithelial cells vealed an association of TGF-b1 and Smad4 vari-
by EMT and perivascular cells (pericytes) via ants in the etiology of keloid scar in the Malay
trans-differentiation, have also been implicated in population.138 Further genetic studies investigat-
pathological scarring (Fig. 6).127 A recent study ing the potential link between keloid disease and
showing that a major proportion of collagen-over- polymorphisms/mutations in components of the
producing cells generated by scarring are derived TGF-b signaling pathway using larger cohorts of
from profibrotic progenitors residing in the peri- patients, particularly individuals with darker skin
TGF-b IN WOUND HEALING 207

pigmentation who are more susceptible to keloid In addition to TGF-b neutralizing antibodies,
formation, are warranted. TGF-b ligand traps represent another class of
molecules in development for neutralizing excess
Strategies used for targeting the TGF-b TGF-b produced in pathological conditions. One
signaling pathway to improve wound example is the soluble TbRII (sTbRII) containing
healing outcome the extracellular domain of TbRII fused to the Fc
The TGF-b signaling pathway is considered a region of IgG1, which was developed as a TGF-b–
promising target for the treatment of many path- sequestering agent and has shown efficacy in var-
ological skin conditions, ranging from chronic ious animal models of human disease.142 A recent
(non-healing) wounds to hypertrophic scarring study has shown that adenoviral delivery of sTbRII
and keloid formation. Manipulation of the TGF-b accelerates lymphatic regeneration and decreases
signaling pathway is also thought to be a suitable inflammation and fibrosis in a mouse tail model of
strategy for improving the cosmetic appearance of lymphedema,143 suggesting that sTbRII may be
non-pathological ‘‘normal’’ scars that result from applicable to other models of wound healing, scar-
surgery or minor injury. This section highlights ring, and fibrosis. LAP is another molecule that has
some of the therapeutic avenues used to target the potential for development as a TGF-b trap for the
TGF-b signaling pathway for improving clinical treatment of TGF-b-driven pathologies. As an ex-
scar outcomes. ample, one study has shown that treatment with
human recombinant LAP (TGF-b1) prevents skin
Neutralizing antibodies and ligand traps. The fibrosis in a mouse model of scleroderma (murine
concept of blocking TGF-b signaling using anti- sclerodermatous graft-versus-host disease).144
TGF-b neutralizing antibodies has been around Other molecules with the potential to act as TGF-b
since the early 1990s with the pioneering work of ligand traps include decorin and fibromodulin,
Ferguson and colleagues, who showed that neu- which are members of the small leucine-rich pro-
tralizing antibodies to TGF-b1/b2 reduces cutane- tein (SLRP) family. Decorin is thought to inhibit all
ous scarring in adult rodents.27,106 This work was TGF-b isoforms,145 which may pose some limita-
expanded on by Mustoe’s group, who investigated tions for its potential as an antiscarring agent, as
the effects of an anti-TGF-b1, 2, and 3 monoclonal it would be expected to block not only the pro-
antibody on wound healing and HTS formation in scarring effects of TGF-b1 and TGF-b2, but also the
rabbit ear wounds.108 They found that early anti-scarring effects of TGF-b3. A recent study
treatment of the wounds (days 0, 2, and 4 post- demonstrated that a recombinant decorin protein
wounding) with a neutralizing antibody impaired fused to a wound-homing peptide (CARSKNKDC;
wound healing without decreasing scar hypertro- CAR peptide) displays enhanced TGF-b1 and TGF-
phy; whereas middle (days 7, 9, and 11) and late b2 neutralization activity and reduced TGF-b3
(days 11, 12, and 13) treatment of wounds signifi- neutralization activity as compared with non-
cantly reduced scar hypertrophy.108 CAT-192 is targeted decorin in vitro, and also enhances wound
a human monoclonal antibody that neutralizes healing and suppresses scar formation in mice at
TGF-b1 and was shown to improve corneal wound doses where non-targeted decorin is inactive.146
healing in bovine organ cultures by promoting Decorin has also been shown to block the activity of
re-epithelialization.139 connective tissue growth factor (CCN2), a down-
Scleroderma, or systemic sclerosis (SSc), is a stream mediator of pro-fibrotic TGF-b action,147
rare connective tissue disease that is characterized suggesting that its antiscarring activity may ex-
by autoimmunity, vasculopathy, and fibrosis tend beyond its effects on TGF-b neutralization.
(scarring) of the skin and internal organs.140 Due to Fibromodulin has also been shown to promote
the potentially critical role of TGF-b in the patho- wound healing and to reduce scarring in animal
genesis of SSc, it was thought that CAT-192 might models148,149 and may represent a therapeutic
provide therapeutic benefit to SSc patients. Al- target for the treatment of HTS.150,151
though a Phase I/II clinical trial showed that CAT- TGF-b co-receptors represent a unique class of
192 was safe and well tolerated across all dose molecules that are starting to receive attention for
levels, no conclusions regarding its efficacy in SSc their potential as TGF-b ligand neutralizing agents.
could be made.141 Future preclinical studies The ability of TGF-b co-receptors, particularly beta-
showing that CAT-192 reduces cutaneous scarring glycan and CD109, to bind TGF-b isoforms with high
would be expected to translate well to a clinical affinity and specificity make them attractive tar-
setting, as safety and tolerability studies on CAT- gets for the development of TGF-b isoform-specific
192 have already been performed.141 traps. As an example, peptide 144 (P144) is a 14-mer
208 FINNSON ET AL.

peptide from human betaglycan that was designed full-thickness incisional wounds in the rat skin.158
as a TGF-b1 inhibitor and has shown efficacy in re- Topical application of a novel ALK5 inhibitor (CP-
ducing fibrosis in different animal models.152,153 639180) was shown to reduce collagen deposition in
P144 is currently being tested in a Phase II clinical a rat dermal incision wound healing model.159
trial for the treatment of skin fibrosis in SSc.154,155 The latter two studies demonstrate the feasibility
of using topically applied small-molecule ALK5
ALK5 kinase inhibitors. Since excessive TGF-b kinase inhibitors to improve cutaneous scarring
has been shown to lead to fibrotic scarring, blocking after wound healing. A recent study reported the
TGF-b action by inhibiting TGF-b receptor kinase discovery of a series of small molecules known as
activity using small-molecule inhibitors is expected 2-(1H-pyrazol-1-yl)pyridines that act as potent
to be beneficial in promoting an antiscarring effect. ALK5 kinase inhibitors and have demonstrated
Several small molecule inhibitors that block ALK5 their potential utility in the prevention of dermal
kinase activity have been tested for their efficacy to scarring.160 Topical application of one of these
improve scarring in preclinical models. The small compounds (PF-03671148) in a rat incisional
molecule SB431542, which was developed as a po- wound repair model led to a reduction in fibrotic
tent inhibitor of ALK5,156 has been shown to re- gene expression without altering the normal
duce scar formation in the eye after glaucoma wound healing process.160 Although these studies
filtration surgery in rabbits as evidenced by a de- are promising, it remains to be seen whether any of
crease in collagen deposition in the subconjunctival these ALK5 inhibitors will be of clinical use, as
space in the experimental groups.157 SB431542 none of them have yet been screened in Phase I
used in combination with recombinant human clinical trials for safety and tolerability.
granulocyte colony-stimulating factor (G-CSF) and
macrophage colony-stimulating factor (M-CSF) TGF-b3. As mentioned earlier, research on
was shown to improve wound breaking strength in the expression and function of TGF-b isoforms

Figure 8. Therapeutic strategies that have been used to target the TGF-b signaling pathway to reduce scarring. (1) Neutralizing TGF-b antibodies and (2)
ligand traps bind TGF-b and neutralize TGF-b activity by preventing its binding to the TGF-b signaling receptors (TbRII and TbRI). (3) Small molecules that
inhibit ALK5 kinase activity, including CP-639180 (depicted), prevent ALK5-induced phosphorylation of Smad2 and Smad3 and downstream signaling events. (4)
Recombinant TGF-b3 protein (avotermin) binds to TGF-b signaling receptors and elicits Smad2/3-dependent signaling. Unlike TGF-b1 and TGF-b2 isoforms that
have pro-scarring effects, TGF-b3 has anti-scarring properties. The molecular mechanisms underlying the different responses of the TGF-b isoforms have not
been elucidated. (5) Antisense oligonucleotides are single-stranded DNA or RNA sequences that are complementary to a specific mRNA sequence. They bind
to their target mRNA sequence and silence gene expression by blocking protein translation or promoting degradation of the mRNA transcript. To see this
illustration in color, the reader is referred to the web version of this article at www.liebertpub.com/wound
TGF-b IN WOUND HEALING 209

(-b1, -b2, and -b3) in scar-free (embryonic)


TAKE-HOME MESSAGES
and scar-forming (adult) wound healing
models has shown that the three TGF-b Basic science advances
 TGF-b plays important roles in all three phases (inflammation, prolifer-
isoforms play very different roles in
ation, and maturation) of wound healing.
scarring, with TGF-b1 and -b2 showing
pro-scarring effects and TGF-b3 display-  TGF-b isoforms (-b1, -b2, and -b3) play distinct roles in wound healing
ing antiscarring effects.161 The discovery with TGF-b1/2 having predominantly pro-scarring roles and TGF-b3
of the antiscarring properties of TGF-b3 having mainly anti-scarring effects.
led to a clinical development program for  Recent advances in our understanding of TGF-b signaling, including TGF-
evaluating recombinant human TGF-b3 b synthesis and activation, TGF-b receptor activity, Smad pathways, and
(avotermin) as a therapeutic intervention modulation by co-receptors, provide new opportunities to delineate the
(prophylatic) to reduce scarring in human mechanisms by which TGF-b signaling regulates specific wound healing
surgical wounds. Prophylactic adminis- events.
tration of avotermin was successful for Clinical science advances
improvement of skin scarring in several  Several therapeutic agents, including anti–TGF-b neutralizing antibodies,
double-blind, placebo-controlled, phase I/ TGF-b ligand traps, small-molecule inhibitors of TGF-b signaling recep-
II studies.162–165 Avotermin was shown to tors, and TGF-b3, have shown promising results in improving scarring in
significantly improve the visual appear- preclinical studies. TGF-b3 was successful in reducing scarring in Phase
ance of scars,162–165 decrease scar surface I/II clinical trials but failed to show efficacy in a Phase III clinical trial.
area, and promote a collagen organization  The potential beneficial effects of TGF-b isoforms during the early stages
that more closely resembled normal skin of wound healing and the different effects of TGF-b1/b2 (pro-scarring)
in 14 of 19 cases.162 Unfortunately, avo- and TGF-b3 (anti-scarring) at the later stages of wound healing suggest
termin failed to show efficacy in Phase III that modulation of TGF-b signaling to promote wound healing and/or
trials, possibly due to the use of a different reduce scarring may require agents that modulate TGF-b signaling in a
TGF-b3 standard, which led to a twofold temporal and isoform-specific manner.
overestimation of TGF-b3 concentration,
Relevance to clinical care
and, therefore, a 50% lower dose of TGF-b3
 The development of therapeutic agents that target the TGF-b signaling
used in the Phase III clinical trial as com- pathway to reduce scarring is relevant to clinicians and surgeons who
pared with the Phase I/II clinical trials.166 treat conditions such as burn injury or other trauma that lead to hy-
pertrophic scarring, keloid formation, and/or tissue fibrosis.
Antisense oligonucleotides. Antisense
oligonucleotides are single-stranded DNA
or RNA that are complementary to a specific mRNA Future studies targeting other components of
sequence which blocks target gene expression either the TGF-b signaling pathway to modulate wound
by inhibiting RNA translation or by promoting en- healing and scarring outcome are eagerly awaited.
zymatic degradation of the mRNA target.167 Several Figure 8 depicts strategies for targeting the TGF-b
studies demonstrate that antisense oligonucleotides pathway to improve wound healing outcome as
targeting components of the TGF-b signaling path- described earlier.
way have the potential to modulate wound healing
and scarring outcomes. In one study, topical appli-
cation of antisense TGF-b1 oligonucleotides reduced LIMITATIONS AND FUTURE DIRECTIONS
scarring of incisional wounds in mice as compared Although the TGF-b signaling pathway is con-
with sense control oligonucleotides.168 In addition, sidered a promising therapeutic target for the
TGF-b1 antisense oligonucleotides reduced scarring treatment of impaired wound healing and exces-
and improved surgical outcome in animal models in sive scarring (fibrosis), currently, no therapies are
which surgical procedures performed resemble available that target the TGF-b signaling pathway
those done in glaucoma patients.169 In addition, to improve wound healing outcome. One potential
Smad3 antisense oligonucleotides accelerated explanation for the lack of ‘‘translatability’’ of
wound healing and reduced scarring in a mouse TGF-b research is that the animal models used in
excisional wound model.170 More advanced methods preclinical research may not mimic the wound heal-
of gene silencing such as RNA interference have also ing and scarring responses in humans. Accordingly,
been successfully employed in wound healing and there has been recent progress in the development
scarring studies. Accordingly, transcutaneous de- of animal models such as the nude mouse model of
livery of Smad3 siRNA decreases radiation-induced hypertrophic scarring, which displays many char-
skin fibrosis as compared with control siRNA.171 acteristics of human hypertrophic scarring172,173
210 FINNSON ET AL.

and chemically induced animal models of systemic with the figure preparation. The work presented in
sclerosis (scleroderma) that reproduce the entire this article was supported by a Canadian Institute
spectrum of the human disease, including inflam- of Health Research Grant to A.P.
mation, autoimmunity, vasculopathy, and fibro-
sis.174 Such models provide powerful tools for
future preclinical studies, exploring the utility of AUTHOR DISCLOSURE AND GHOSTWRITING
agents that target the TGF-b signaling pathway to No competing financial interests exist. The con-
improve wound healing or scarring outcomes. In tent of this article was expressly written by the
addition, identifying the cellular source of TGF-b authors listed. No ghostwriters were used to write
that causes scarring and fibrosis and devising this article.
strategies using specific modes of delivery such as
topical treatment and intralesional delivery or
ABOUT THE AUTHORS
systemic administration may be important. For
example, emerging evidence indicates that fibro- Dr. Kenneth Finnson is a Research Associate
blasts from the deeper dermal layers produce more with Dr. Anie Philip in the Department of Surgery,
of the pro-fibrotic TGF-b1 and less anti-fibrotic McGill University, Montreal, Quebec, Canada. Dr.
decorin, fibromodulin, and TGF-b3 than fibroblasts Gianni Di Guglielmo is an Associate Professor in
from superficial layers and contribute to the de- the Department of Physiology and Pharmacology,
velopment of HTS after injuries involving the deep Western University, London, Ontario, Canada. Dr.
dermis.150,151,175 The latter findings have impor- Di Guglielmo’s research focuses on understanding
tant implications for selecting an appropriate the role of TGF-b receptor endocytosis in the reg-
strategy for delivering an anti-TGF-b therapy ulation of TGF-b signaling. Dr. Sarah McLean is
specifically to the deep dermis to ameliorate fibro- a recent PhD graduate from Dr. Di Guglielmo’s lab.
sis in HTS patients. Dr. McLean’s PhD work focused on the role of the
TGF-b co-receptor betaglycan in regulating TGF-b
signaling. Dr. Anie Philip is a Professor in
ACKNOWLEDGMENTS the Department of Surgery, McGill University.
AND FUNDING SOURCE Dr. Philip’s research program focuses on under-
The authors thank Phililp lab members Anshu- standing the molecular mechanisms of TGF-b
man Saksena and Christopher Chiavatti for help signaling in wound healing, scarring, and fibrosis.

REFERENCES
1. Penn JW, Grobbelaar AO, and Rolfe KJ: The role 8. Ruschke K, Hiepen C, Becker J, and Knaus P: 14. Munz B, Smola H, Engelhardt F, Bleuel K,
of the TGF-b family in wound healing, burns and BMPs are mediators in tissue crosstalk of the Brauchle M, Lein I, Evans LW, Huylebroeck D,
scarring: a review. Int J Burns Trauma 2012; regenerating musculoskeletal system. Cell Tis- Balling R, and Werner S: Overexpression of ac-
2: 18. sue Res 2012; 347: 521. tivin A in the skin of transgenic mice reveals
new activities of activin in epidermal morpho-
2. Wu MY and Hill CS: TGF-b superfamily signaling 9. Ono Y, Calhabeu F, Morgan JE, Katagiri T, genesis, dermal fibrosis and wound repair.
in embryonic development and homeostasis. Dev Amthor H, and Zammit PS: BMP signalling per- EMBO J 1999; 18: 5205.
Cell 2009; 16: 329. mits population expansion by preventing pre-
mature myogenic differentiation in muscle 15. Wankell M, Munz B, Hübner G, Hans W, Wolf E,
3. Biernacka A, Dobaczewski M, and Frangogiannis
NG: TGF-b signaling in fibrosis. Growth Factors satellite cells. Cell Death Differ 2011; 18: 222. Goppelt A, and Werner S: Impaired wound
2011; 29: 196. 10. Elliott B, Renshaw D, Getting S, and Mackenzie healing in transgenic mice overexpressing the
R: The central role of myostatin in skeletal activin antagonist follistatin in the epidermis.
4. Rider CC and Mulloy B: Bone morphogenetic EMBO J 2001; 20: 5361.
protein and growth differentiation factor cyto- muscle and whole body homeostasis. Acta Phy-
kine families and their protein antagonists. siol (Oxf) 2012; 205: 324.
16. Antsiferova M, Klatte JE, Bodó E, Paus R, Jor-
Biochem J 2010; 429: 1. 11. Knight PG and Glister C: TGF-b superfamily cano JL, Matzuk MM, Werner S, and Kögel H:
5. Schmierer B and Hill CS: TGF-b/Smad signal members and ovarian follicle development. Re- Keratinocyte-derived follistatin regulates epi-
transduction: molecular specificity and functional production 2006; 132: 191. dermal homeostasis and wound repair. Lab In-
flexibility. Nat Rev Mol Cell Biol 2007; 8: 970. vest 2009; 89: 131.
12. Werner S and Alzheimer C: Roles of activin in
6. Horbelt D, Denkis A, and Knaus P: A portrait of tissue repair, fibrosis, and inflammatory disease. 17. Schier AF: Nodal morphogens. Cold Spring Harb
transforming growth factor-b superfamily sig- Cytokine Growth Factor Rev 2006; 17: 157. Perspect Biol 2009; 1: a003459.
nalling: background matters. Int J Biochem Cell
13. Hubner G, Hu Q, Smola H, and Werner S: Strong 18. Strizzi L, Hardy KM, Kirschmann DA, Ahrlund-
Biol 2012; 44: 469.
induction of activin expression after injury sug- Richter L, and Hendrix MJ: Nodal expression and
7. Chen D, Zhao M, and Mundy GR: Bone morpho- gests an important role of activin in wound re- detection in cancer: experience and challenges.
genetic proteins. Growth Factors 2004; 22: 233. pair. Dev Biol 1996; 173: 490. Cancer Res 2012; 72: 1915.
TGF-b IN WOUND HEALING 211

19. Shi Y and Massague J: Mechanisms of TGF-b 33. Yu Q and Stamenkovic I: Cell surface-localized 48. Wieser R, Wrana JL, and Massague J: GS do-
signaling from cell membrane to the nucleus. matrix metalloproteinase-9 proteolytically acti- main mutations that constitutively activate TbR-I,
Cell 2003; 113: 685. vates TGF-b and promotes tumor invasion and the downstream signaling component in the
angiogenesis. Genes Dev 2000; 14: 163. TGF-b receptor complex. EMBO J 1995; 14: 2199.
20. Laverty HG, Wakefield LM, Occleston NL, O’Kane
S, and Ferguson MW: TGF-b3 and cancer: a 34. Annes JP, Rifkin DB, and Munger JS: The in- 49. Attisano L, and Lee-Hoeflich ST: The Smads.
review. Cytokine Growth Factor Rev 2009; 20: tegrin aVb6 binds and activates latent TGFb3. Genome Biol 2001; 2: REVIEWS3010.
305. FEBS Lett 2002; 511: 65.
50. Nakao A, Imamura T, Souchelnytskyi S, Kawa-
21. Schlingensiepen KH, Schlingensiepen R, Stein- 35. Dallas SL, Zhao S, Cramer SD, Chen Z, Peehl bata M, Ishisaki A, Oeda E, Tamaki K, Hanai J,
brecher A, Hau P, Bogdahn U, Fischer-Blass B, DM, and Bonewald LF: Preferential production of Heldin CH, Miyazono K, and ten Dijke P: TGF-b
and Jachimczak P: Targeted tumor therapy with latent transforming growth factor b-2 by primary receptor-mediated signalling through Smad2,
the TGF-b2 antisense compound AP 12009. Cy- prostatic epithelial cells and its activation by Smad3 and Smad4. EMBO J 1997; 16: 5353.
tokine Growth Factor Rev 2006; 17: 129. prostate-specific antigen. J Cell Physiol 2005;
51. Nakao A, Röijer E, Imamura T, Souchelnytskyi S,
202: 361.
22. Dunker N and Krieglstein K: Targeted mutations Stenman G, Heldin CH, and ten Dijke P: Identi-
of transforming growth factor-b genes reveal 36. Ahamed J, Janczak CA, Wittkowski KM, and fication of Smad2, a human Mad-related protein
important roles in mouse development and adult Coller BS: In vitro and in vivo evidence that in the transforming growth factor-b signaling
homeostasis. Eur J Biochem 2000; 267: 6982. thrombospondin-1 (TSP-1) contributes to stirring- pathway. J Biol Chem 1997; 272: 2896.
23. Shull MM, Ormsby I, Kier AB, Pawlowski S, and shear-dependent activation of platelet-de-
52. Macı́as-Silva M, Abdollah S, Hoodless PA, Pir-
Diebold RJ, Yin M, Allen R, Sidman C, Proetzel rived TGF-b1. PLoS One 2009; 4: e6608.
one R, Attisano L, and Wrana JL: MADR2 is a
G, Calvin D, Annunziata N, and Doetschman T: 37. Grainger DJ, Wakefield L, Bethell HW, Farndale substrate of the TGFb receptor and its phos-
Targeted disruption of the mouse transforming RW, and Metcalfe JC: Release and activation of phorylation is required for nuclear accumulation
growth factor-b1 gene results in multifocal in- platelet latent TGF-b in blood clots during dis- and signaling. Cell 1996; 87: 1215.
flammatory disease. Nature 1992; 359: 693. solution with plasmin. Nat Med 1995; 1: 932.
53. Massague J and Gomis RR: The logic of TGF-b
24. Sanford LP, Ormsby I, Gittenberger-de Groot AC, 38. Nunes I, Shapiro RL, and Rifkin DB: Character- signaling. FEBS Lett 2006; 580: 2811.
Sariola H, Friedman R, Boivin GP, Cardell EL, and ization of latent TGF-b activation by murine
Doetschman T: TGFb2 knockout mice have mul- 54. Xiao Z, Latek R, and Lodish HF: An extended
peritoneal macrophages. J Immunol 1995; 155:
tiple developmental defects that are non-over- bipartite nuclear localization signal in Smad4 is
1450.
lapping with other TGFb knockout phenotypes. required for its nuclear import and transcrip-
Development 1997; 124: 2659. 39. Derynck R and Feng XH: TGF-b receptor signal- tional activity. Oncogene 2003; 22: 1057.
ing. Biochim Biophys Acta 1997; 1333: F105.
25. Proetzel G, Pawlowski SA, Wiles MV, Yin M, 55. López-Casillas F, Cheifetz S, Doody J, Andres JL,
Boivin GP, Howles PN, Ding J, Ferguson MW, 40. Attisano L and Wrana JL: Signal transduction by Lane WS, and Massagué J: Structure and ex-
and Doetschman T: Transforming growth factor- members of the transforming growth factor-b pression of the membrane proteoglycan beta-
b3 is required for secondary palate fusion. Nat superfamily. Cytokine Growth Factor Rev 1996; glycan, a component of the TGF-beta receptor
Genet 1995; 11: 409. 7: 327. system. Cell 1991; 67: 785.

26. Ferguson MW and O’Kane S: Scar-free healing: 41. Henis YI, Moustakas A, Lin HY, and Lodish HF: 56. Moren A, Ichijo H, and Miyazono K: Molecular
from embryonic mechanisms to adult therapeutic The types II and III transforming growth factor-b cloning and characterization of the human and
intervention. Philos Trans R Soc Lond B Biol Sci receptors form homo-oligomers. J Cell Biol 1994; porcine transforming growth factor-b type III
2004; 359: 839. 126: 139. receptors. Biochem Biophys Res Commun 1992;
189: 356.
27. Shah M, Foreman DM, and Ferguson MW: 42. Lin HY, Moustakas A, Knaus P, Wells RG, Henis
Neutralisation of TGF-b1 and TGF-b2 or exoge- YI, and Lodish HF: The soluble exoplasmic do- 57. Gougos A and Letarte M: Primary structure of en-
nous addition of TGF-b3 to cutaneous rat main of the type II transforming growth factor doglin, an RGD-containing glycoprotein of human
wounds reduces scarring. J Cell Sci 1995; 108 (TGF)-b receptor. A heterogeneously glycosy- endothelial cells. J Biol Chem 1990; 265: 8361.
(Pt 3): 985. lated protein with high affinity and selectivity for
58. Yamashita H, Ichijo H, Grimsby S, Morén A, ten
TGF-b ligands. J Biol Chem 1995; 270: 2747.
28. Horiguchi M, Ota M, and Rifkin DB: Matrix Dijke P, and Miyazono K: Endoglin forms a het-
control of transforming growth factor-b function. 43. Lin HY, Wang XF, Ng-Eaton E, Weinberg RA, and eromeric complex with the signaling receptors
J Biochem 2012; 152: 321. Lodish HF: Expression cloning of the TGF-b type for transforming growth factor-b. J Biol Chem
II receptor, a functional transmembrane serine/ 1994; 269: 1995.
29. Annes JP, Munger JS, and Rifkin DB: Making threonine kinase. Cell 1992; 68: 775.
sense of latent TGF-b activation. J Cell Sci 2003; 59. Guerrero-Esteo M, Sanchez-Elsner T, Letamendia
116: 217. 44. Luo K, Zhou P, and Lodish HF: The specificity of A, and Bernabeu C: Extracellular and cytoplasmic
the transforming growth factor b receptor ki- domains of endoglin interact with the trans-
30. Sato Y and Rifkin DB: Inhibition of endothelial nases determined by a spatially addressable forming growth factor-b receptors I and II. J Biol
cell movement by pericytes and smooth muscle peptide library. Proc Natl Acad Sci USA 1995; Chem 2002; 277: 29197.
cells: activation of a latent transforming growth 92: 11761.
factor-b1-like molecule by plasmin during co- 60. Chen W, Kirkbride KC, How T, Nelson CD, Mo J,
culture. J Cell Biol 1989; 109: 309. 45. Wrana JL, Attisano L, Wieser R, Ventura F, and Frederick JP, Wang XF, Lefkowitz RJ, and Blobe
Massague J: Mechanism of activation of the GC: b-Arrestin 2 mediates endocytosis of type III
31. Pircher R, Jullien P, and Lawrence DA: Beta-
TGF-b receptor. Nature 1994; 370: 341. TGF-b receptor and down-regulation of its sig-
transforming growth factor is stored in human
naling. Science 2003; 301: 1394.
blood platelets as a latent high molecular weight 46. Bierie B, and Moses HL: Tumour microenviron-
complex. Biochem Biophys Res Commun 1986; ment: TGF-b: the molecular Jekyll and Hyde of 61. Bernabeu C, Lopez-Novoa JM, and Quintanilla
136: 30. cancer. Nat Rev Cancer 2006; 6: 506. M: The emerging role of TGF-b superfamily co-
receptors in cancer. Biochim Biophys Acta 2009;
32. Brunner G and Blakytny R: Extracellular regula- 47. Huse M, Muir TW, Xu L, Chen YG, Kuriyan J,
1792: 954.
tion of TGF-b activity in wound repair: growth Massagué J: The TGF-b receptor activation
factor latency as a sensor mechanism for injury. process: an inhibitor- to substrate-binding 62. Wong SH, Hamel L, Chevalier S, and Philip A:
Thromb Haemost 2004; 92: 253. switch. Mol Cell 2001; 8: 671. Endoglin expression on human microvascular
212 FINNSON ET AL.

endothelial cells: association with betaglycan CD109 as part of the TGF-b receptor system in 89. Li J, Zhang YP, and Kirsner RS: Angiogenesis in
and formation of higher order complexes with human keratinocytes. FASEB J 2006; 20: 1525. wound repair: angiogenic growth factors and
TGF-b signalling receptors. Eur J Biochem 2000; the extracellular matrix. Microsc Res Tech 2003;
75. Bizet AA, Liu K, Tran-Khanh N, Saksena A,
267: 5550. 60: 107.
Vorstenbosch J, Finnson KW, Buschmann MD,
63. McLean S and Di Guglielmo GM: TbRIII directs and Philip A: The TGF-b co-receptor, CD109, 90. Hinz B, Phan SH, Thannickal VJ, Prunotto M,
clathrin-mediated endocytosis of TGFb type I promotes internalization and degradation of TGF- Desmoulière A, Varga J, De Wever O, Mareel M,
and II receptors. Biochem J 2010; 429: 137. b receptors. Biochim Biophys Acta: Molecular and Gabbiani G: Recent developments in myofi-
Cell Research 2011; 1813: 742. broblast biology: paradigms for connective tissue
64. ten Dijke P, Goumans MJ, and Pardali E: En-
remodeling. Am J Pathol 2012; 180: 1340.
doglin in angiogenesis and vascular diseases. 76. Bizet AA, Tran-Khanh N, Saksena A, Liu K,
Angiogenesis 2008; 11: 79. Buschmann MD, and Philip A: CD109-mediated 91. Barrientos S, Stojadinovic O, Golinko MS, Brem
degradation of the TGF-b receptors and inhibi- H, and Tomic-Canic M: Growth factors and cy-
65. Finnson KW, Parker WL, Chi Y, Hoemann CD,
tion of TGF-b responses involve regulation of tokines in wound healing. Wound Repair Regen
Goldring MB, Antoniou J, and Philip A: Endoglin
Smad7 and Smurf2 localization and function. J 2008; 16: 585.
differentially regulates TGF-b-induced Smad2/3
Cell Biochem 2012; 113: 238.
and Smad1/5 signalling and its expression cor- 92. Gailit J, Welch MP, and Clark RA: TGF-b1
relates with extracellular matrix production and 77. Doherty GJ and McMahon HT: Mechanisms of stimulates expression of keratinocyte integrins
cellular differentiation state in human chon- endocytosis. Annu Rev Biochem 2009; 78: 857. during re-epithelialization of cutaneous wounds.
drocytes. Osteoarthritis Cartilage 2010; 18: 1518. J Invest Dermatol 1994; 103: 221.
78. Di Guglielmo GM, Le Roy C, Goodfellow AF, and
66. Finnson KW, Parker WL, ten Dijke P, Thorikay M, Wrana JL. Distinct endocytic pathways regulate 93. Xue M, Le NT, and Jackson CJ: Targeting matrix
and Philip A: ALK1 opposes ALK5/Smad3 sig- TGF-b receptor signalling and turnover. Nat Cell metalloproteases to improve cutaneous wound
naling and expression of extracellular matrix Biol 2003; 5: 410. healing. Expert Opin Ther Targets 2006; 10: 143.
components in human chondrocytes. J Bone
79. Mitchell H, Choudhury A, Pagano RE, and Leof 94. Brew K, Dinakarpandian D, and Nagase H: Tis-
Miner Res 2008; 23: 896.
EB: Ligand-dependent and -independent trans- sue inhibitors of metalloproteinases: evolution,
67. Parker WL, Goldring MB, and Philip A: Endoglin forming growth factor-b receptor recycling reg- structure and function. Biochim Biophys Acta
is expressed on human chondrocytes and forms ulated by clathrin-mediated endocytosis and 2000; 1477: 267.
a heteromeric complex with betaglycan in a li- Rab11. Mol Biol Cell 2004; 15: 4166.
gand and type II TGFb receptor independent 95. Lorenz H, and Longaker M: Wounds: Biology,
80. Razani B, Zhang XL, Bitzer M, von Gersdorff G, Pathology and Management. New York: Springer,
manner. J Bone Miner Res 2003; 18: 289.
Böttinger EP, and Lisanti MP: Caveolin-1 regu- 2003, pp. 77–88 (2003).
68. Morris E, Chrobak I, Bujor A, Hant F, Mummery lates transforming growth factor (TGF)-b/Smad
C, Ten Dijke P, and Trojanowska M: Endoglin signaling through an interaction with the TGF-b 96. Redden RA, and Doolin EJ: Collagen crosslinking
promotes TGF-b/Smad1 signaling in scleroderma type I receptor. J Biol Chem 2001; 276: 6727. and cell density have distinct effects on fibro-
fibroblasts. J Cell Physiol 2011; 226: 3340. blast-mediated contraction of collagen gels. Skin
81. Kavsak P, Rasmussen RK, Causing CG, Bonni S, Res Technol 2003; 9: 290.
69. Leask A, Abraham DJ, Finlay DR, Holmes A, Zhu H, Thomsen GH, and Wrana JL: Smad7
Pennington D, Shi-Wen X, Chen Y, Venstrom K, binds to Smurf2 to form an E3 ubiquitin ligase 97. Udupa SL: Inhibition of lysyl oxidase by isoniazid
Dou X, Ponticos M, Black C, Bernabeu C, Jack- that targets the TGF-b receptor for degradation. and its effect on wound healing. Indian J Exp
man JK, Findell PR, and Connolly MK: Dysregu- Mol Cell 2000; 6: 1365. Biol 1995; 33: 278.
lation of transforming growth factor b signaling 98. Lau YK, Gobin AM, and West JL: Overexpression
82. Ogunjimi AA, Briant DJ, Pece-Barbara N, Le Roy
in scleroderma: overexpression of endoglin in of lysyl oxidase to increase matrix crosslinking
C, Di Guglielmo GM, Kavsak P, Rasmussen RK,
cutaneous scleroderma fibroblasts. Arthritis and improve tissue strength in dermal wound
Seet BT, Sicheri F, and Wrana JL: Regulation of
Rheum 2002; 46: 1857. healing. Ann Biomed Eng 2006; 34: 1239.
Smurf2 ubiquitin ligase activity by anchoring the
70. Tam B, Germain L, and Philip A: TGF-b receptor E2 to the HECT domain. Mol Cells 2005; 19: 297. 99. Colwell AS, Krummel TM, Longaker MT, and
expression on human keratinocytes: a 150 kDa Lorenz HP: Early-gestation fetal scarless wounds
83. Schultz GS, Davidson JM, Kirsner RS, Bornstein
GPI-anchored TGF-b1 binding protein forms a have less lysyl oxidase expression. Plast Reconstr
P, and Herman IM: Dynamic reciprocity in the
heteromeric complex with type I and type II re- Surg 2006; 118: 1125; discussion 1130–1131.
wound microenvironment. Wound Repair Regen
ceptors. J Cell Biochem 1998; 70: 573.
2011; 19: 134. 100. Szauter KM, Cao T, Boyd CD, and Csiszar K: Lysyl
71. Tam B and Philip A: Transforming growth factor- oxidase in development, aging and pathologies
84. Clemetson KJ: Platelets and primary haemos-
b receptor expression on human skin fibroblasts: of the skin. Pathol Biol (Paris) 2005; 53: 448.
tasis. Thromb Res 2012; 129: 220.
dimeric complex formation of type I and type II
receptors and identification of glycosyl phos- 85. Wang XJ, Han G, Owens P, Siddiqui Y, and Li 101. Finnson KW, Arany P, and Philip A: Transforming
phatidylinositol-anchored transforming growth AG: Role of TGF b-mediated inflammation in growth factor-beta signaling in cutaneous wound
factor-b binding proteins. J Cell Physiol 1998; cutaneous wound healing. J Investig Dermatol healing: lessons learned from animal studies.
176: 553. Symp Proc 2006; 11: 112. Adv Wound Care 2013; 2: XXX.
72. Tam B, Larouche D, Germain L, Hooper N, and 86. Singer AJ and Clark RA: Cutaneous wound 102. Kane CJ, Hebda PA, Mansbridge JN, and Ha-
Philip A: Characterization of a 150 kDa accessory healing. N Engl J Med 1999; 341: 738. nawalt PC: Direct evidence for spatial and tem-
receptor for TGF-b1 on keratinocytes: direct ev- poral regulation of transforming growth factor
87. Wahl S: Cytokines in wound healing. R&D Sys-
idence for a GPI anchor and ligand binding of the b1 expression during cutaneous wound healing.
tems, Inc., 2002; available online at http://www
released form. J Cell Biochem 2001; 83: 494. J Cell Physiol 1991; 148: 157.
.rndsystems.com/mini_review_detail_objectname
73. Tam BYY, Finnson KW, and Philip A: Glycosyl- _MR02_CytokineWoundHealing.aspx (accessed 103. Levine JH, Moses HL, Gold LI, and Nanney LB:
phosphatidylinositol-anchored proteins regulate May 7, 2013). Spatial and temporal patterns of immunoreactive
transforming growth factor-b signaling in human transforming growth factor b1, b2, and b3 during
88. Midwood KS, Williams LV, and Schwarzbauer
keratinocytes. J Biol Chem 2003; 278: 49610. excisional wound repair. Am J Pathol 1993; 143: 368.
JE: Tissue repair and the dynamics of the ex-
74. Finnson KW, Tam BY, Liu K, Marcoux A, Lepage tracellular matrix. Int J Biochem Cell Biol 2004; 104. Roberts AB, Sporn MB, Assoian RK, Smith JM,
P, Roy S, Bizet AA, and Philip A: Identification of 36: 1031. Roche NS, Wakefield LM, Heine UI, Liotta LA,
TGF-b IN WOUND HEALING 213

Falanga V, and Kehrl JH: Transforming growth factor fibroblasts produce more transforming growth growth factor b 1, 2, and 3 proteins in keloids.
type b: rapid induction of fibrosis and angiogenesis factor-b1 mRNA and protein than normal skin Ann Plast Surg 1999; 43: 179.
in vivo and stimulation of collagen formation in vitro. and cells. Wound Repair Regen 2000; 8: 128.
131. Xia W, Phan TT, Lim IJ, Longaker MT, and Yang
Proc Natl Acad Sci USA 1986; 83: 4167.
118. Schmid P, Itin P, Cherry G, Bi C, and Cox DA: GP: Complex epithelial-mesenchymal interac-
105. Mustoe TA, Pierce GF, Thomason A, Gramates P, Enhanced expression of transforming growth tions modulate transforming growth factor-b
Sporn MB, and Deuel TF: Accelerated healing of factor-b type I and type II receptors in wound expression in keloid-derived cells. Wound Repair
incisional wounds in rats induced by transform- granulation tissue and hypertrophic scar. Am J Regen 2004; 12: 546.
ing growth factor-b. Science 1987; 237: 1333. Pathol 1998; 152: 485.
132. Chin GS, Liu W, Peled Z, Lee TY, Steinbrech DS,
106. Shah M, Foreman DM, and Ferguson MW: 119. Xie JL, Qi SH, Pan S, Xu YB, Li TZ, Liu XS, and Liu Hsu M, and Longaker MT: Differential expression
Neutralising antibody to TGF-b 1, 2 reduces P: Expression of Smad protein by normal skin of transforming growth factor-b receptors I and
cutaneous scarring in adult rodents. J Cell Sci fibroblasts and hypertrophic scar fibroblasts in II and activation of Smad 3 in keloid fibroblasts.
1994; 107 (Pt 5): 1137. response to TGF-b1. Dermatol Surg 2008; 34: Plast Reconstr Surg 2001; 108: 423.
1216; discussion 1224–1225.
107. Shah M, Foreman DM, and Ferguson MW: 133. Phan TT, Lim IJ, Aalami O, Lorget F, Khoo A, Tan
Control of scarring in adult wounds by neu- 120. Eickelberg O, Köhler E, Reichenberger F, Bert- EK, Mukhopadhyay A, and Longaker MT: Smad3
tralising antibody to transforming growth factor schin S, Woodtli T, Erne P, Perruchoud AP, and signalling plays an important role in keloid
b. Lancet 1992; 339: 213. Roth M: Extracellular matrix deposition by pri- pathogenesis via epithelial-mesenchymal inter-
mary human lung fibroblasts in response to TGF- actions. J Pathol 2005; 207: 232.
108. Lu L, Saulis AS, Liu WR, Roy NK, Chao JD,
b1 and TGF-b3. Am J Physiol 1999; 276: L814.
Ledbetter S, and Mustoe TA: The temporal ef- 134. Bayat A, Bock O, Mrowietz U, Ollier WE, and
fects of anti-TGF-b1, 2, and 3 monoclonal anti- 121. Tredget EE, Shankowsky HA, Pannu R, Nedelec Ferguson MW: Genetic susceptibility to keloid
body on wound healing and hypertrophic scar B, Iwashina T, Ghahary A, Taerum TV, and Scott disease and transforming growth factor b2
formation. J Am Coll Surg 2005; 201: 391. PG: Transforming growth factor-b in thermally polymorphisms. Br J Plast Surg 2002; 55: 283.
injured patients with hypertrophic scars: effects
109. Kryger ZB, Sisco M, Roy NK, Lu L, Rosenberg D, 135. Bayat A, Bock O, Mrowietz U, Ollier WE, and
of interferon alpha-2b. Plast Reconstr Surg 1998;
and Mustoe TA: Temporal expression of the Ferguson MW: Genetic susceptibility to keloid
102: 1317; discussion 1329–1330.
transforming growth factor-b pathway in the disease and hypertrophic scarring: transforming
rabbit ear model of wound healing and scarring. 122. Tredget EE, Wang R, Shen Q, Scott PG, and growth factor b1 common polymorphisms and
J Am Coll Surg 2007; 205: 78. Ghahary A: Transforming growth factor-b mRNA plasma levels. Plast Reconstr Surg 2003; 111:
and protein in hypertrophic scar tissues and fibro- 535; discussion 544–546.
110. Reid RR, Roy N, Mogford JE, Zimmerman H, Lee
blasts: antagonism by IFN-a and IFN-c in vitro and
C, and Mustoe TA: Reduction of hypertrophic 136. Bayat A, Walter JM, Bock O, Mrowietz U, Ollier
in vivo. J Interferon Cytokine Res 2000; 20: 143.
scar via retroviral delivery of a dominant nega- WE, and Ferguson MW: Genetic susceptibility to
tive TGF-b receptor II. J Plast Reconstr Aesthet 123. Ulloa L, Doody J, and Massague J: Inhibition of keloid disease: mutation screening of the TGFb3
Surg 2007; 60: 64; discussion 73–74. transforming growth factor-b/Smad signalling by gene. Br J Plast Surg 2005; 58: 914.
the interferon-c/STAT pathway. Nature 1999;
111. Sumiyoshi K, Nakao A, Setoguchi Y, Okumura K, 137. Brown JJ, Ollier W, Arscott G, Ke X, Lamb J, Day
397: 710.
and Ogawa H: Exogenous Smad3 accelerates P, and Bayat A: Genetic susceptibility to keloid
wound healing in a rabbit dermal ulcer model. J 124. Wang J, Jiao H, Stewart TL, Shankowsky HA, scarring: Smad gene SNP frequencies in Afro-
Invest Dermatol 2004; 123: 229. Scott PG, Tredget EE: Improvement in postburn Caribbeans. Exp Dermatol 2008; 17: 610.
hypertrophic scar after treatment with IFN-al-
112. Kloeters O, Jia SX, Roy N, Schultz GS, Leinfellner 138. Emami A, Halim AS, Salahshourifar I, Yussof SJ,
pha2b is associated with decreased fibrocytes. J
G, and Mustoe TA: Alteration of Smad3 signaling Khoo TL, and Kannan TP: Association of TGF-b1
Interferon Cytokine Res 2007; 27: 921.
in ischemic rabbit dermal ulcer wounds. Wound and Smad4 variants in the etiology of keloid scar
Repair Regen 2007; 15: 341. 125. Grieb G, Steffens G, Pallua N, Bernhagen J, and in the Malay population. Arch Dermatol Res
Bucala R: Circulating fibrocytes: biology and 2012; 304: 541.
113. Vorstenbosch J, Gallant-Behm C, Trzeciak A, Roy
mechanisms in wound healing and scar forma-
S, Mustoe T, and Philip A: Transgenic mice 139. Carrington LM, Albon J, Anderson I, Kamma C,
tion. Int Rev Cell Mol Biol 2011; 291: 1.
overexpressing CD109 in the epidermis display and Boulton M: Differential regulation of key
decreased inflammation and granulation tissue 126. Wang JF, Jiao H, Stewart TL, Shankowsky HA, stages in early corneal wound healing by TGF-b
and improved collagen architecture during wound Scott PG, and Tredget EE: Fibrocytes from burn isoforms and their inhibitors. Invest Ophthalmol
healing. Wound Repair Regen 2013; 21: 235. patients regulate the activities of fibroblasts. Vis Sci 2006; 47: 1886.
Wound Repair Regen 2007; 15: 113.
114. Vorstenbosch J, Al-Ajmi H, Winocour S, Trzeciak
140. Leask A: Emerging targets for the treatment of
A, Lessard L, and Philip A: CD109 overexpression 127. Sarrazy V, Billet F, Micallef L, Coulomb B, and scleroderma. Expert Opin Emerg Drugs 2012; 17: 173.
ameliorates skin fibrosis in a bleomycin-induced Desmouliere A: Mechanisms of pathological
mouse model of scleroderma. Arthritis Rheum; scarring: role of myofibroblasts and current de- 141. Denton CP, Merkel PA, Furst DE, Khanna D,
2013; 65: 1378. velopments. Wound Repair Regen 2011; 19 Emery P, Hsu VM, Silliman N, Streisand J,
Suppl 1: s10. Powell J, Akesson A, Coppock J, Hoogen FV,
115. Gauglitz GGM, Korting HCM, Pavicic TM, Ruzicka
Herrick A, Mayes MD, Veale D, Haas J, Led-
TM, and Jeschke MGMP: Hypertrophic scarring and 128. Dulauroy S, Di Carlo SE, Langa F, Eberl G, and
better S, Korn JH, Black CM, Seibold JR; Cat-192
keloids: pathomechanisms, current and emerging Peduto L: Lineage tracing and genetic ablation of
Study Group; Scleroderma Clinical Trials Con-
treatment strategies. Mol Med 2011; 17: 113. ADAM12( + ) perivascular cells identify a major
sortium: Recombinant human anti-transforming
source of profibrotic cells during acute tissue
116. Scott PG, Dodd CM, Tredget EE, Ghahary A, and growth factor b1 antibody therapy in systemic
injury. Nat Med 2012; 18: 1262.
Rahemtulla F: Immunohistochemical localization sclerosis: a multicenter, randomized, placebo-
of the proteoglycans decorin, biglycan and ver- 129. Rorison P, Thomlinson A, Hassan Z, Roberts SA, controlled phase I/II trial of CAT-192. Arthritis
sican and transforming growth factor-b in human Ferguson MW, and Shah M: Longitudinal changes Rheum 2007; 56: 323.
post-burn hypertrophic and mature scars. Histo- in plasma transforming growth factor b-1 and
142. Russo LM, Brown D, and Lin HY: The soluble
pathology 1995; 26: 423. post-burn scarring in children. Burns 2010; 36: 89.
transforming growth factor-beta receptor: ad-
117. Wang R, Ghahary A, Shen Q, Scott PG, Roy K, 130. Lee TY, Chin GS, Kim WJ, Chau D, Gittes GK, vantages and applications. Int J Biochem Cell
and Tredget EE: Hypertrophic scar tissues and and Longaker MT: Expression of transforming Biol 2009; 41: 472.
214 FINNSON ET AL.

143. Avraham T, Daluvoy S, Zampell J, Yan A, Haviv cebo controlled clinical trial, to evaluate effi- 164. Bush J, Duncan JA, Bond JS, Durani P, So K,
YS, Rockson SG, and Mehrara BJ: Blockade of cacy and safety of P144 topical administration Mason T, O’Kane S, and Ferguson MW: Scar-
transforming growth factor-b1 accelerates lym- for skin fibrosis in patients with systemic scle- improving efficacy of avotermin administered
phatic regeneration during wound repair. Am J rosis. ClinicalTrials.gov [Identifier: NCT00574613] into the wound margins of skin incisions as
Pathol 2010; 77: 3202. 2013 Feb 8; available online at http://clinical evaluated by a randomized, double-blind, place-
trials.gov/ct2/show/NCT00574613 (accessed bo-controlled, phase II clinical trial. Plast Re-
144. Zhang Y, McCormick LL, and Gilliam AC: Latency-
May 7, 2013). constr Surg 2010; 126: 1604.
associated peptide prevents skin fibrosis in mu-
rine sclerodermatous graft-versus-host disease, 155. Matucci M, Krieg T, Müller-Ladner U, Czirják L, 165. Ferguson MW, Duncan J, Bond J, Bush J, Durani
a model for human scleroderma. J Invest Der- Denton C, and Pablos JL: Open label extension P, So K, Taylor L, Chantrey J, Mason T, James G,
matol 2003; 121: 713. (OLE) for the patients treated in the ISD002- Laverty H, Occleston NL, Sattar A, Ludlow A,
P144-07 study with P144 topical adminsitration O’Kane S: Prophylactic administration of avo-
145. Hildebrand A, Romarı́s M, Rasmussen LM, Hei-
for skin fibrosis in patients with systemic scle- termin for improvement of skin scarring: three
negård D, Twardzik DR, Border WA, and Ruo-
rosis. ClinicalTrials.gov [Identifier: NCT00781053] double-blind, placebo-controlled, phase I/II
slahti E: Interaction of the small interstitial
2013 Feb 8; available online at http://clinical studies. Lancet 2009; 373: 1264.
proteoglycans biglycan, decorin and fibromodulin
trials.gov/show/NCT00781053 (accessed May 7,
with transforming growth factor b. Biochem J 166. Little JA, Murdy R, Cossar N, Getliffe KM, Hanak J,
2013).
1994; 302 (Pt 2): 527. and Ferguson MW: TGF b3 immunoassay stan-
156. Inman GJ, Nicolás FJ, Callahan JF, Harling JD, dardization: comparison of NIBSC reference prep-
146. Jarvinen TA and Ruoslahti E: Target-seeking
Gaster LM, Reith AD, Laping NJ, and Hill CS: SB- aration code 98/608 with avotermin lot 205-0505-
antifibrotic compound enhances wound healing
431542 is a potent and specific inhibitor of 005. J Immunoassay Immunochem 2012; 33: 66.
and suppresses scar formation in mice. Proc Natl
transforming growth factor-b superfamily type I
Acad Sci USA 2010; 107: 21671. 167. Deleavey GF and Damha MJ: Designing chemi-
activin receptor-like kinase (ALK) receptors ALK4,
147. Neill T, Schaefer L, and Iozzo RV: Decorin: a ALK5, and ALK7. Mol Pharmacol 2002; 62: 65. cally modified oligonucleotides for targeted gene
guardian from the matrix. Am J Pathol 2012; silencing. Chem Biol 2012; 19: 937.
157. Xiao YQ, Liu K, Shen JF, Xu GT, and Ye W: SB-
181: 380. 168. Choi BM, Kwak HJ, Jun CD, Park SD, Kim KY,
431542 inhibition of scar formation after filtra-
148. Stoff A, Rivera AA, Mathis JM, Moore ST, tion surgery and its potential mechanism. Invest Kim HR, and Chung HT: Control of scarring in
Banerjee NS, Everts M, Espinosa-de-los-Monteros Ophthalmol Vis Sci 2009; 50: 1698. adult wounds using antisense transforming
A, Novak Z, Vasconez LO, Broker TR, Richter DF, growth factor-b1 oligodeoxynucleotides. Im-
158. Sugiyama K, Ishii G, Ochiai A, and Esumi H: munol Cell Biol 1996; 74: 144.
Feldman D, Siegal GP, Stoff-Khalili MA, and Curiel
Improvement of the breaking strength of wound
DT: Effect of adenoviral mediated overexpression
by combined treatment with recombinant human 169. Cordeiro MF, Mead A, Ali RR, Alexander RA,
of fibromodulin on human dermal fibroblasts and
G-CSF, recombinant human M-CSF, and a TGF- Murray S, Chen C, York-Defalco C, Dean NM,
scar formation in full-thickness incisional wounds.
b1 receptor kinase inhibitor in rat skin. Cancer Schultz GS, and Khaw PT: Novel antisense oli-
J Mol Med (Berl) 2007; 85: 481.
Sci 2008; 99: 1021. gonucleotides targeting TGF-b inhibit in vivo
149. Zheng Z, Nguyen C, Zhang X, Khorasani H, Wang scarring and improve surgical outcome. Gene
159. Bian F, Render J, Ren XD, Chio C, Chan K, Boys
JZ, Zara JN, Chu F, Yin W, Pang S, Le A, Ting K, Ther 2003; 10: 59.
M, Lala DS, and Pocalyko D: An activin receptor-
and Soo C: Delayed wound closure in fi-
like kinase 5 inhibitor reduces collagen deposi- 170. Hong HJ, Jin SE, Park JS, Ahn WS, and Kim CK:
bromodulin-deficient mice is associated with
tion in a rat dermal incision wound healing Accelerated wound healing by Smad3 antisense
increased TGF-b3 signaling. J Invest Dermatol
model. Plast Reconstr Surg 2011; 128: 451e. oligonucleotides-impregnated chitosan/alginate
2010; 131: 769.
polyelectrolyte complex. Biomaterials 2008; 29:
160. Boys ML, Bian F, Kramer JB, Chio CL, Ren XD,
150. Honardoust D, Varkey M, Hori K, Ding J, Shan- 4831.
Chen H, Barrett SD, Sexton KE, Iula DM, Filzen
kowsky HA, and Tredget EE: Small leucine-rich
GF, Nguyen MN, Angell P, Downs VL, Wang Z, 171. Lee JW, Tutela JP, Zoumalan RA, Thanik VD,
proteoglycans, decorin and fibromodulin, are re-
Raheja N, Ellsworth EL, Fakhoury S, Bratton LD, Nguyen PD, Varjabedian L, Warren SM, and
duced in postburn hypertrophic scar. Wound
Keller PR, Gowan R, Drummond EM, Maiti SN, Saadeh PB: Inhibition of Smad3 expression in
Repair Regen 2011; 19: 368.
Hena MA, Lu L, McConnell P, Knafels JD, Tha- radiation-induced fibrosis using a novel method
151. Honardoust D, Varkey M, Marcoux Y, Shan- nabal V, Sun F, Alessi D, McCarthy A, Zhang E, for topical transcutaneous gene therapy. Arch
kowsky HA, and Tredget EE: Reduced decorin, Finzel BC, Patel S, Ciotti SM, Eisma R, Payne NA, Otolaryngol Head Neck Surg 2010; 136: 714.
fibromodulin, and transforming growth factor-b3 Gilbertsen RB, Kostlan CR, Pocalyko DJ, and Lala
in deep dermis leads to hypertrophic scarring. J DS: Discovery of a series of 2-(1H-pyrazol-1- 172. Momtazi M, Kwan P, Ding J, Anderson CC,
Burn Care Res 2012; 33: 218. yl)pyridines as ALK5 inhibitors with potential Honardoust D, Goekjian S, and Tredget EE: A
utility in the prevention of dermal scarring. nude mouse model of hypertrophic scar shows
152. Ezquerro IJ, Lasarte JJ, Dotor J, Castilla-Cortázar morphologic and histologic characteristics of
Bioorg Med Chem Lett 2012; 22: 3392.
I, Bustos M, Peñuelas I, Blanco G, Rodrı́guez C, human hypertrophic scar. Wound Repair Regen
Lechuga Mdel C, Greenwel P, Rojkind M, Prieto 161. Durani P, Occleston N, O’Kane S, and Ferguson 2013; 21: 77.
J, and Borrás-Cuesta F: A synthetic peptide from MW: Avotermin: a novel antiscarring agent. Int J
transforming growth factor-b type III receptor Low Extrem Wounds 2008; 7: 160. 173. Wang J, Ding J, Jiao H, Honardoust D, Momtazi M,
inhibits liver fibrogenesis in rats with carbon Shankowsky HA, and Tredget EE: Human hyper-
162. So K, McGrouther DA, Bush JA, Durani P, Taylor L, trophic scar-like nude mouse model: characteriza-
tetrachloride liver injury. Cytokine 2003; 22: 12.
Skotny G, Mason T, Metcalfe A, O’Kane S, and tion of the molecular and cellular biology of the
153. Santiago B, Gutierrez-Cañas I, Dotor J, Palao G, Ferguson MW: Avotermin for scar improvement scar process. Wound Repair Regen 2011; 19: 274.
Lasarte JJ, Ruiz J, Prieto J, Borrás-Cuesta F, and following scar revision surgery: a randomized, dou-
Pablos JL: Topical application of a peptide in- ble-blind, within-patient, placebo-controlled, phase 174. Batteux F, Kavian N, and Servettaz A: New in-
hibitor of transforming growth factor-b1 ame- II clinical trial. Plast Reconstr Surg 2011; 128: 163. sights on chemically induced animal models of
liorates bleomycin-induced skin fibrosis. J Invest systemic sclerosis. Curr Opin Rheumatol 2011;
163. McCollum PT, Bush JA, James G, Mason T, 23: 511.
Dermatol 2005; 125: 450.
O’Kane S, McCollum C, Krievins D, Shiralkar S,
154. Matucci M, Krieg T, Müller-Ladner U, Czirják L, and Ferguson MW: Randomized phase II clinical 175. Wang J, Dodd C, Shankowsky HA, Scott PG, and
Denton C, and Pablos JL: Phase II, multicenter, trial of avotermin versus placebo for scar im- Tredget EE: Deep dermal fibroblasts contribute to
randomized, double-blind, intraindividually pla- provement. Br J Surg 2011; 98: 925. hypertrophic scarring. Lab Invest 2008; 88: 1278.

View publication stats

You might also like