immune system wound healing

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 23

COMPREHENSIVE INVITED REVIEW

Immune Regulation of Skin Wound Healing:


Mechanisms and Novel Therapeutic Targets

Jacqueline Larouche,1,{ Sumit Sheoran,1,{ Kenta Maruyama,2


and Mikaël M. Martino1,*
1
European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute,
Monash University, Victoria, Australia.
2
WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.
{
These authors contributed equally.
Downloaded by Gothenburg University Library from online.liebertpub.com at 02/12/18. For personal use only.

Significance: The immune system plays a central role in orchestrating the


tissue healing process. Hence, controlling the immune system to promote
tissue repair and regeneration is an attractive approach when designing re-
generative strategies. This review discusses the pathophysiology of both acute
and chronic wounds and possible strategies to control the immune system to
accelerate chronic wound closure and promote skin regeneration (scar-less
healing) of acute wounds.
Recent Advances: Recent studies have revealed the key roles of various im-
mune cells and immune mediators in skin repair. Thus, immune components
Mikaël M. Martino, PhD
have been targeted to promote chronic wound repair or skin regeneration and
several growth factors, cytokines, and biomaterials have shown promising Submitted for publication September 20, 2017.
results in animal models. However, these novel strategies are often struggling Accepted in revised form December 12, 2017.
*Correspondence: European Molecular Biology
to meet efficacy standards in clinical trials, partly due to inadequate drug de- Laboratory Australia, Australian Regenerative
livery systems and safety concerns. Medicine Institute, 15 Innovation Walk, Building
Critical Issues: Excess inflammation is a major culprit in the dysregulation of 75, Level 1, Martino Lab, Victoria 3800, Australia
(e-mail: mikael.martino@monash.edu).
normal wound healing, and further limiting inflammation effectively reduces
scarring. However, current knowledge is insufficient to efficiently control in-
flammation and specific immune cells. This is further complicated by inadequate
drug delivery methods.
Future Directions: Improving our understanding of the molecular pathways
through which the immune system controls the wound healing process could
facilitate the design of novel regenerative therapies. Additionally, better delivery
systems may make current and future therapies more effective. To promote the
entry of current regenerative strategies into clinical trials, more evidence on
their safety, efficacy, and cost-effectiveness is also needed.

Keywords: immune system, chronic wounds, scarring, biomaterials,


therapeutics, immunomodulation

SCOPE AND SIGNIFICANCE treatment costs.1 With rising rates


Chronic skin wounds and exces- of obesity and diabetes, and an aging
sive scarring represent large and population, these numbers are pro-
growing challenges to healthcare jected to rise.1 Ineffectively treated
systems globally. For example, in chronic wounds can result in infec-
2009, chronic wounds affected 6.5 tion, sepsis, amputation, and, in ex-
million people in the United States, treme cases, death of the patient.
leading to US$25 billion in annual Scarring can also have considerable

ADVANCES IN WOUND CARE, VOLUME 00, NUMBER 00


Copyright ª 2018 by Mary Ann Liebert, Inc. DOI: 10.1089/wound.2017.0761
j 1
2 LAROUCHE ET AL.

functional and physiological consequences. In mune response to dermal wounding. However,


2004, an estimated 234 million surgeries were many of the mechanisms through which immune
performed globally, corresponding to about one cells act to regulate the wound healing response are
operation per 25 people.2 These incisions result in still unknown. Elucidating these pathways may aid
a scar, each of which is very difficult to treat and in the development of novel regenerative strategies
impossible to prevent. Nonetheless, $12 billion is for promoting wound resolution and preventing scar
spent annually in the United States to reduce formation.
scar size.1
The immune system is a key player throughout TRANSLATIONAL RELEVANCE
the wound healing process, actively participating
There has been a recent growing interest in the
to reestablish homeostasis following tissue injury
development of immune-based therapies for both
via multiple mechanisms.3 For example, the im-
chronic wounds and scar prevention, mostly fo-
mune response to tissue injury can mobilize tissue
cusing on the delivery of growth factors, cytokines,
resident stem/progenitor cells, promote cell differ-
and other immunomodulatory factors. However,
Downloaded by Gothenburg University Library from online.liebertpub.com at 02/12/18. For personal use only.

entiation, extracellular matrix (ECM) deposition,


translating these therapies for chronic wound res-
and stimulate growth factor secretion to promote
olution and scar prevention into the clinic has
neoangiogenesis. Since these processes require a
been particularly difficult, due to challenges re-
coordinated series of immune events, their dis-
lated to delivery methods and safety. Most thera-
ruption can lead to a variety of wound pathologies,
peutics currently rely on discrete, bolus doses, given
including chronic wounds and scar formation.
via injections or applied topically to the wound site.
Chronic wounds are defined as wounds that fail
Because of natural clearance, doses are given at
to resolve after 1 month, often having persistent
supra-physiological levels to sustain drug presence
infections and microbial films. Clinically, they are
throughout the wound healing process. This results
classified as either venous or arterial ulcers, pres-
in both safety concerns and poor long-term efficacy.
sure sores, or diabetic ulcers (Fig. 1).4 Alternatively,
Hence, developing new methods to effectively con-
dermal fibrosis results from increased deposition of
trol and sustain drug release over longer periods of
ECM and hyper-proliferation of keratinocytes at
time is critical to the success of immune therapies.
the wound site. Keloids and hypertrophic (HT)
scars both represent clinical pathologies of scar-
ring, though even normal scar formation repre- CLINICAL RELEVANCE
sents a failure of the dermal layer to regenerate Numerous immune-based therapies for both
postwounding (Fig. 2).4 Not surprisingly, chronic chronic wounds and scar prevention have entered
wounds and scarring represent nearly opposite clinical trials in the past decade. For example,
extremes of dysregulated inflammation and im- macrophage-activating lipopeptide-2 ( MALP-2)

Figure 1. Clinical presentations of various chronic wound pathologies. Impaired wound healing cascade will lead to the development of nonhealing wound
pathologies such as: (A) venous leg ulcer (B) arterial ulcer (C) diabetic foot ulcer (D) pressure sore. Reprinted with adaptation with permission from Ref.4
ªAAAS Publishing Group.
IMMUNE REGULATION OF SKIN WOUND HEALING 3

Figure 2. Clinical examples of scarring pathologies. (A) Hypertrophic scar (B) Keloid. Reprinted with adaptation with permission from Ref.4 ªAAAS
Publishing Group.

and platelet-rich plasma (PRP) gels have reached strategies to promote chronic wound resolution and
Downloaded by Gothenburg University Library from online.liebertpub.com at 02/12/18. For personal use only.

clinical trials to resolve chronic wounds.5,6 In the dermal regeneration (scar-less healing).
case of scar prevention, transforming growth factor
(TGF)-b3, interleukin (IL)-10, mannose-6-phosphate
(M6P), and nefopam have all reached clinical trials BACKGROUND
with varying levels of success. TGF-b3 significantly Role of the immune system in acute
decreased scar size in both phase I and II clini- skin wound healing
cal trials, though complete regeneration was never Following injury, an inflammatory response is
achieved and it failed to meet efficacy standards both normal and critical in restoring tissue ho-
in phase III.7,8 In the case of therapeutics for both meostasis. Wound healing is typically divided into
chronic wounds and scar prevention, it is believed four phases: hemostasis, inflammation, prolifera-
that improved delivery methods that permit sus- tion, and remodeling. These phases result from
tained release may facilitate clinical effectiveness. a cascade of events, largely mediated by immune
In this review, we describe the molecular patho- cells and signaling molecules (Fig. 3A).9 In re-
physiology of acute wounds and major etiologies that sponse to cues from both the damaged ECM and
alter the normal wound healing cascade, leading to tissue-resident cells, thrombocytes and immune
development of chronic wounds and scars. Then, we cells accumulate at the wound site in a stepwise
highlight the possible approaches to control the im- manner. Platelets are the first cells to arrive, at
mune system using biomaterial and biologic-based which point they help initiate the coagulation

Figure 3. Overview of the immune mechanisms in acute and chronic wound healing. (A) Acute wound healing results from a well-coordinated series of
events divided into four overlapping phases: hemostasis, inflammation, proliferation/matrix deposition, and tissue remodeling. Neutrophils and macrophages
are particularly important in mediating this process, though T cells and platelets also play key roles. (B) High numbers of inflammatory cells and the formation
of a biofilm preclude the restoration of tissue homeostasis in chronic wounds. Excess secretion of inflammatory mediators leads to growth factor and ECM
degradation and prevents macrophage phenotype conversion, which creates a feed-forward loop preventing resolution. Black arrows indicate differentiation,
blue arrows indicate inhibition and red arrows indicate induction. ECM, extracellular matrix.
4 LAROUCHE ET AL.

cascade to prevent further blood loss and provide a most tissues that can proliferate upon injury.9
provisional ECM for further cell infiltration. Also, While, it has been demonstrated in a murine
by secreting TGF-b1 and platelet-derived growth wound model that tissue-resident macrophages
factors (PDGFs), they play a critical role in the have little impact on the timing of the wound
activation of fibroblasts and mesenchymal cells healing process or tissue integrity following reso-
as well as in the recruitment and activation of lution,27 their role in skin wound healing is still
neutrophils and macrophages.10–12 Unsurprisingly, elusive.
platelet defects are correlated with impaired wound Macrophages undergo phenotypic changes
healing, and delivering autologous PRP improves throughout the healing process, which helps
wound resolution.13–16 transition the wound microenvironment from a
Neutrophils are the first immune cells recruited pro-inflammatory into a pro-resolution state.9 The
into wounded tissue, and remain for about 24 h most commonly studied phenotypes are the
before undergoing apoptosis.17 They play a cen- proinflammatory—commonly referred to as ‘‘classi-
tral role in both killing microbes and promoting cally activated’’ or M1—and anti-inflammatory—
Downloaded by Gothenburg University Library from online.liebertpub.com at 02/12/18. For personal use only.

wound healing.18 Neutrophils control invading referred to as ‘‘alternatively activated’’ or M2—


pathogens by secreting a variety of antimicrobial macrophages. Indeed, this M1 and M2 classification
substances—reactive oxygen species (ROS), anti- originated from in vitro characterization, but a
microbial peptides, and antimicrobial proteases— modified classification system has been recently
and by phagocytosing them with the help of proposed to link macrophage populations to activa-
neutrophil extracellular traps (NETs).19,20 Neu- tion pathways [e.g., M(IFN-c), M(IL-4), and so on].28
trophils also secrete various cytokines and growth Nevertheless, it is very difficult to know by which
factors, including IL-17 and vascular endothelial pathways macrophages have been activated in vivo.
growth factor (VEGF). Cytokines and growth fac- Hence, in this review, we refer to macrophage phe-
tors are both chemotactic for inflammatory cells, notypes based on their primary function in vivo
and promote the proliferation of fibroblasts, kera- (‘‘anti-inflammatory,’’ ‘‘proinflammatory,’’ ‘‘profi-
tinocytes, and endothelial cells.18,21 Demonstrat- brotic,’’ etc.).
ing the importance of neutrophils during wound Many studies have confirmed that macrophages
healing, perturbation of neutrophil recruitment are critical for proper wound healing.29–32 Upon
during the early phase of wound healing by initial infiltration, proinflammatory macrophages
knocking out C-X-C motif chemokine receptor 2 (the so-called M1) remove cellular debris, damaged
(CXCR2) in mice impairs wound healing.22 CXCR2 matrix, microbes, and neutrophils. They also secrete
is an essential mediator of neutrophil chemotaxis proinflammatory cytokines and growth factors (in-
via the release of numerous chemokine ligands, cluding IL-1, fibroblast growth factor [FGF]-2,
including C-X-C chemokine ligand (CXCL)-1, -5, PDGF, and VEGF), which mobilize more immune
and -8, by keratinocytes at the wound site.23 cells, and promote the proliferation of keratinocytes,
However, CXCR2 knock-out mice also exhibit al- fibroblasts, and epithelial cells. During the forma-
tered temporal patterns of monocyte infiltration, tion of new tissue, micro environmental cues trig-
decreased secretion of IL-1b, and reduced kerati- ger macrophages to transition into a functionally
nocyte migration and proliferation, so impaired and phenotypically anti-inflammatory state (the
wound healing may be the result of numerous so-called M2). At this point, they begin secreting
compounding factors.22 Another study found ac- anti-inflammatory cytokines, such as TGF-b1,
celerated wound closure and reepithelialization in which promotes ECM synthesis and wound con-
neutrophil-depleted mice, suggesting a relatively traction.33 In the final stage of wound healing, anti-
complex role of neutrophils during skin repair.24 inflammatory macrophages help reorganizing the
Cytokines released by neutrophils during apo- ECM along tension lines and phagocytose remain-
ptosis are chemotactic for monocytes, which start ing debris.17 Demonstrating the importance of these
to arrive 5 to 6 h postinjury. These monocytes dif- immune cells, Lucas et al. depleted macrophages
ferentiate into macrophages, which can remain from mice at each phase of tissue repair. Early-stage
for several weeks at the wound site.17 Monocyte- macrophage depletion significantly reduced granu-
derived macrophages have been widely studied lation tissue formation, impaired epithelialization,
in the context of wound healing, and are often and resulted in decreased scar formation. Mid-stage
considered to be the most important immune cell macrophage depletion resulted in severe hemor-
type in this process.25,26 In addition to macro- rhage, while late-stage depletion did not signifi-
phages deriving from mobilized monocytes, there cantly impact the outcome of the healing response.34
is a population of tissue-resident macrophages in Moreover, anti-inflammatory macrophages play an
IMMUNE REGULATION OF SKIN WOUND HEALING 5

important role in angiogenesis. They indirectly TGF-b, PDGF, FGF-2, IGF1, tumor necrosis factor
promote angiogenesis by degrading the ECM to (TNF)-a, and VEGF.9 TGF-b directly promotes ECM
create tunnels that guide endothelial proliferation synthesis by stimulating mesenchymal cells to dif-
and migration, and they release angiogenic factors, ferentiate into myofibroblasts. Furthermore, TGF-b
such as FGF and placental growth factor (PlGF).35 and PDGF trigger fibroblasts and myofibroblasts to
T lymphocytes (T cells) are also critical in tissue produce new ECM.9 TH2 cells and Tregs also con-
remodeling and the resolution of inflammation. tribute to matrix formation by secreting TGF-b1,
During the inflammatory phase of wound healing, IL-4, -5, -13, and -21, which play a distinct role
macrophages secrete chemokines such as interferon in fibrinogenesis, encouraging anti-inflammatory
gamma (IFN-c) that attract T cells to the wound macrophage polarization, and suppressing other
site.36,37 IFN-c also encourages CD4+ type 1 T helper inflammatory cell types.3,9,48 For example, studies
(TH1) polarization, which contributes to the initial have suggested that IL-4 is almost twice as potent as
proinflammatory wound microenvironment.38 Tis- TGF-b at stimulating collagen synthesis.49
sue resident cdT cells also play a role in regulating During the final phase of wound healing, anti-
Downloaded by Gothenburg University Library from online.liebertpub.com at 02/12/18. For personal use only.

skin wound healing. Indeed, cdT cell-deficient mice fibrotic macrophages release numerous matrix
show defects in skin development due to decreased metalloproteinases (MMPs) (including MMP-2, -
keratinocyte proliferation.39 These immune cells 12, and -19) and trigger the production of type VIII
most likely regulate keratinocyte proliferation and collagen critical for improving local tissue integri-
differentiation via their secretion of various growth ty.9 The result is a decrease in cell proliferation and
factors, such as FGF-7, FGF-10, and insulin-like protein synthesis, and an ECM that is remodeled
growth factor (IGF)-1.40 Importantly, there are into larger fibrils oriented along tension lines.
significant differences between mouse and human Eventually, most endothelial cells, macrophages,
cdT cells, though studies exist that suggest a similar and myofibroblasts either exit the wound or un-
role of human cdT cells in wound healing.41,42 For dergo apoptosis, and recently formed capillaries
example, cdT cells isolated from healthy human begin to regress, potentially due to reduced nutri-
skin produce a constituent level of IGF-1, which ent requirements.9 Depending on the size of the
increases threefold upon stimulation. Additionally, initial injury, this entire process usually results in
based on an in vitro skin organ culture model, T-cell a small scar.4
receptor (TCR)-specific activation of human cdT
cells accelerates the rate of early wound closure.
This increase in wound closure was dependent on DISCUSSION OF FINDINGS
IGF-1 secretion.42 Regulatory T cells (Tregs) also AND RELEVANT LITERATURE
play a central role in maintaining skin homeosta- Role of the immune system
sis.43 They secrete arginase and anti-inflammatory in chronic wounds
cytokines (including IL-10 and TGF-b1), which en- While the wound healing cascade is well coordi-
courages anti-inflammatory macrophage polariza- nated in acute wound healing, chronic wounds fail to
tion and suppresses the inflammatory response.44 A progress beyond the inflammatory phase (Table 1),
recent study using Foxp3-DTR transgenic mice which precludes proliferation, matrix deposition,
(where Tregs are depleted following diphtheria and ultimately, wound resolution.50,51 Indeed, many
toxin injection), showed that wound healing is studies have shown that there is an imbalance be-
slower in Treg depleted mice compared with wild- tween pro- and anti-inflammatory signals in chronic
type controls.45 This clearly shows the importance of wounds, which disturbs the microenvironment and
Tregs in cutaneous wound healing. Contrastingly, hampers the wound healing process.52,53
natural killer T cells have an inhibitory role on
wound healing. Indeed, mouse studies have shown Role of neutrophils in chronic wounds. While
that depleting natural killer T cells increases wound neutrophils play a role in reestablishing tissue ho-
closure rate.46,47 meostasis through pathogen phagocytosis and mac-
As inflammation is resolved, the wound pro- rophage recruitment, excessive neutrophil activity
gresses into the final two stages of the wound can contribute to the development of nonhealing
healing process: matrix formation with angiogen- wounds.10,21 For instance, excess neutrophils at the
esis and remodeling. This resolution is largely de- wound site lead to an overproduction of ROS, caus-
pendent upon macrophages, Tregs and TH2 cells.9 ing ECM and cell membrane damage, and resulting
Anti-inflammatory macrophages are a prominent in premature cell senescence (Fig. 3B).19,54 Not only
source of cytokines and growth factors that pro- do ROS directly damage the ECM, but they also
mote matrix deposition and angiogenesis, including activate proteases (MMPs and serine proteases) and
6 LAROUCHE ET AL.

Table 1. Comparison of the immune microenvironment contributing to the development of nonhealing


between acute and chronic wounds
wounds.69 Moreover, macrophage conversion from
Components Acute Wounds Chronic Wounds a proinflammatory to an anti-inflammatory phe-
Inflammation Controlled Increased
notype is critical to wound resolution.70 If this
Neutrophils Present only during early phase Prolonged presence phenotypic conversion does not occur or reach
Controlled NETosis Increased NETosis completion, it may lead to the development of
Macrophages Polarization transition from Impaired polarization chronic wounds (Fig. 3B). For example, using iron
pro- to anti- inflammatory
phenotype
to induce an incomplete macrophage phenotypic
Inflammatory cytokines Low level Increased level switch impairs healing in both humans and
Anti-inflammatory High level Low level mice.71 In these studies, 80% of macrophages at
cytokines the wound margins had a proinflammatory phe-
Granulation tissue Present Low amount
MMPs Low level High level
notype, contributing to the persistence of the
TIMPs Normal level Low level when wounds.71 Other studies in diabetic mouse mod-
compared to els have also shown that when macrophages do
Downloaded by Gothenburg University Library from online.liebertpub.com at 02/12/18. For personal use only.

increased MMPs not undergo the appropriate phenotypic conver-


Fibrosis Low level Increased
Growth factors/cytokines Normal degradation rate Increased degradation
sion, it leads to a reduction in key growth factors,
rate such as TGF-b1, VEGF, and IGF-1, which are
necessary for the progression into the prolifera-
MMP, matrix metalloproteinase; TIMP, tissue inhibitor of matrix
metalloproteinase. tion phase.72 Proinflammatory macrophages also
secrete inflammatory mediators, such as TNF-a,
IL-17 and IL-1b, ROS, and inducible nitric oxide
inactivate protease inhibitors.55–57 Unsurprisingly, synthase (iNOS), which have negative effects on
this results in increased proteolysis, further ex- the wound microenvironment at high concentra-
acerbating ECM degradation.58–61 In addition, tions.73 For instance, surplus TNF-a leads to in-
proteases released by neutrophils are capable of creased secretion of MMP-1 and MMP-3 and
degrading key wound healing growth factors, decreased tissue inhibitor of matrix metallopro-
including PDGF-BB and TGF-b1 (Fig. 3B).10,62 teinase (TIMP)-1 secretion, which directly contrib-
Finally, although the mechanism is not fully un- utes to wound chronicity through excess ECM
derstood, it has been shown that excess NET- proteolysis.74 Similarly, excess ROS disturb the ox-
osis impairs wound resolution in both mice and idant/antioxidant balance. Not only does this en-
humans.63–66 hance the signaling pathways that regulate the
Because neutrophils are recruited to wound sites secretion of proinflammatory cytokines/chemokines
in such large numbers and exacerbate the proin- (IL-1, IL-6, and TNF-a) and MMPs, but it is also
flammatory microenvironment through toxic com- implicated in premature fibroblast senescence.75,76
pounds and cytokine secretion, their efferocytosis This further contributes to wound chronicity as
is critical to inflammation resolution. In fact, dys- senescent fibroblasts produce elevated levels of
regulated neutrophil apoptosis is likely a key con- proteases (including MMPs 2, 3, and 9), and fewer
tributor to the nonhealing state of chronic wounds. protease inhibitors.4,76 Altogether, changes in
Chronic wounds in diabetic mice have increased macrophage polarization are highly regulated in
numbers of apoptotic cells, most of which are neu- acute wound healing, and disruptions contribute
trophils, resulting in a prolonged inflammatory to the development of wound chronicity.
state.67 Interestingly, neutrophils isolated from Beyond secreting cytokines and growth fac-
chronic diabetic mouse wounds undergo less apo- tors to mediate wound healing, macrophages
ptosis in response to S. aureus infection. This also play a critical role in phagocytosing apoptotic
results in sustained secretion of proinflammatory cells and other debris in the wound bed. As pre-
cytokines, including TNF-a.68 The discrepancy po- viously mentioned, efficient efferocytosis is es-
tentially results from an overall increase in neu- pecially critical in the context of chronic wounds
trophil recruitment to diabetic wounds or from the due to increased infiltration and apoptosis of
lack of bacterial infection in the second study. neutrophils. However, in chronic wounds, mac-
However, in both cases, dysregulated neutrophil rophages exhibit a reduced phagocytic capacity,
apoptosis contributed to the chronic wound state. exacerbating the apoptotic cell burden.67 In ad-
dition, unsuccessful efferocytosis results in a
Role of macrophages in chronic wounds. In- higher ratio of pro:anti-inflammatory cytokines,
creased macrophage infiltrate at the wound site can further compromising resolution of diabetic
also perturb the normal wound healing process, wounds.67
IMMUNE REGULATION OF SKIN WOUND HEALING 7

Role of adaptive immune cells in chronic wounds. lead to future infections and wound healing
Compared to our understanding of the role of the complications.81
innate immune system in exacerbating nonhealing
wounds, little is known about the role of the Regulation of skin scarring
adaptive immune system. In a human ex vivo by the immune system
model, it has been shown that a higher number of Scar formation begins in the third phase of wound
Langerhans cells (LCs) (a dendritic cell subtype healing, when fibroblasts, keratinocytes, and epi-
present in early-phase wound healing) is present in thelial cells are recruited into the site of injury. At
healing diabetic foot ulcers (DFUs) compared with this point, fibroblasts differentiate into myofibro-
nonhealing DFUs.77 One of the main roles of LCs blasts, which secrete and assemble new ECM.48
following tissue injury is activating and recruiting This new ECM is later remodeled to form scar tis-
T cells.77 Interestingly, there are also fewer T sue. Not only does the secretion of cytokines by
lymphocytes in chronic wounds, and those that are leukocytes play a direct role in the initial recruit-
present exhibit an unresponsive, functionally im- ment of fibroblasts, but the general inflammatory
Downloaded by Gothenburg University Library from online.liebertpub.com at 02/12/18. For personal use only.

paired state.39,42 Indeed, cd and ab T cells isolated response also plays a key role in mediating future
from human chronic wounds fail to secrete IGF-1 scar development (Fig. 4). This is most evident in the
and IL-2, even following stimulation with phorbol differences between fetal and adult wound healing.
myristate acetate and ionomycin.42 Nevertheless, In the first two trimesters of human development,
while chronic wounds have prolonged infiltration fetuses heal dermal wounds without a scar. This
and different ratios of CD4 and CD8 T cells com- phenomenon is likely dependent upon the absence
pared with acute wounds, depleting mice of CD4 of immune cells, since the transition into the third
and CD8 T cells does not appear to affect wound trimester is characterized by the development of
closure rates.69,78 myeloid lineages and significant scarring following
injury in both humans and mice.82 Wild-type mice
Bacterial colonization in chronic wounds. undergo a similar transition, evidenced by scarless
Bacterial overload is another major factor pre- wound healing in embryonic day 15 (E15) mice and
cluding the resolution of chronic wounds. In cer- scar formation in embryonic day 18 (E18) mice.78,82
tain microenvironments, including those in Early studies found similar scarless wound healing
chronic wounds, bacteria thrive as complex surface- in neonatal PU.1 null mice, which lack neutrophils,
attached communities enclosed in an ECM com- macrophages, and mast cells. They also have re-
posed of hydrated polymers and debris. This is duced mRNA levels of TGF-b1 and PDGF, which act
known as a biofilm. Interestingly, biofilms are not as important profibrotic mediators stimulating
only immune to destruction by the host’s immune myofibroblast differentiation and ECM synthesis.
system, but they also likely manipulate and depend Interestingly, Tcell-deficient athymic nude-nu mice
upon the inflammatory response as a source of also heal with minimal scarring.82–83
sustained nutrients.79,80 Bacterial pathogens in
the biofilm express a range of virulence factors that Role of neutrophils in skin scarring. Potentially
upregulate the levels of proinflammatory cyto- because they only participate in the earliest stages
kines, ROS and MMPs, while diminishing levels of of wound healing, few studies have looked at the
TIMPs and growth factors. This creates a steady role of neutrophils in dermal scarring and they are
state of hyper-inflammation that is outside of believed to have a negligible long-term impact.
the host’s control, contributing to the etiology of Nonetheless, it is interesting to note that neutro-
chronic wounds and making resolution challeng- phils are absent from scarless fetal wounds, there
ing. Biofilms further impair wound healing by is an 80% reduction of neutrophil circulation in Ac-
perturbing the tight junctions between epithelial omys mice (which heal full-thickness excisional
cells. Tight junctions create a fluid-impermeable skin wounds with complete regeneration of der-
barrier by joining the cytoskeletons of adjacent mal components and without scarring) compared
epithelial cells. Because of this, they are critical with Mus mice that heal with a scar, and there is a
to skin’s protective function. Biofilms interfere significant reduction in neutrophil recruitment to
with this barrier partly by causing the down- wounds in the oral mucosa, which also heals rap-
regulation of proteins responsible for creating idly and with minimal scarring.78,84
the tight junctions, including zona occludens-1
and zona occludens-2.81 Hence, even though the Role of macrophages in skin scarring. The role
biofilm-infected wound may eventually appear of macrophages in dermal scarring has been ex-
closed, functionally compromised, leaky skin can tensively studied, and shown to change throughout
Downloaded by Gothenburg University Library from online.liebertpub.com at 02/12/18. For personal use only. 8 LAROUCHE ET AL.

Figure 4. Biomaterial and molecular-based strategies to resolve chronic wounds principally focus on promoting reepithelialization, angiogenesis, progenitor
cell recruitment, directing macrophage polarization, and inhibiting the migration of excess inflammatory cells. Current strategies are very diverse, either
directly delivering cytokines and growth factors to the wound, or by using siRNAs, miRNAs, stem cells, and EVs to alter cytokine expression and production by
cells in the wound bed. Material-based strategies are also being explored, particularly for their potential to direct macrophage polarization. Blue arrows
indicate inhibition and red arrows indicate induction. EV, extracellular vesicle; siRNA, small interfering RNA.

the stages of wound healing.25 Monocytes are the It is possible that changes in macrophage po-
primary cells that secrete IL-1, which increases larization are critical in determining the vary-
collagen synthesis, and promotes fibroblast and ing role of macrophages in wound healing. Indeed,
keratinocyte proliferation.23 Ablating or depleting in muscle remodeling, increases in the anti-
macrophages during the acute and subacute inflammatory population and higher ratios of
phases of repair results in slower wound closure, anti- to proinflammatory macrophages within
epithelialization, and granulation tissue forma- the remodeling site at 14 days postinjury were
tion, but significantly reduced scarring.25,34,85 Si- associated with reduced scarring.89 During
milarly, reducing macrophage migration, early stage wound healing, there is a mixed pro/
adhesion, and their ability to produce TGF-b1 by anti-inflammatory activation phenotype. This
diminishing b-catenin levels results in slower transitions to an anti-inflammatory macrophage
wound repair but less scarring.86 If macrophage polarization in late stage wound healing charac-
inhibition or ablation is discontinued, wound clo- terized by an upregulation of IL-10.25 Since wound
sure rates are rescued by their infiltration in the associated macrophages are the main sustained
later stages of wound repair without affecting scar source of TGF-bs (1, 2, and 3) and extensively se-
formation or morphology.85 Alternatively, only crete MMPs to activate TGF-b, differences in TGF-b
depleting macrophages in the late stage wound and MMP secretion between classically and alter-
healing has no effect on scar morphology com- natively activated macrophages could account for
pared to control mice.25,34 Furthermore, a study the time-specific role of macrophages.85,90 For ex-
by Dardenne et al. showed that wounds treated ample, IL-4, a cytokine that promotes profibrotic
with high mobility group box-1 (HMGB-1), an macrophage polarization, was significantly down-
alarmin, have increased scarring. These wounds regulated in Acomys, which heal without a scar,
showed a significant increase in macrophages 48 h compared to Mus. Macrophages polarized via IL-4
postinjury and no differences in neutrophil infil- (M(IL-4)) are known to secrete and activate TGF-b1,
tration or mast cell numbers or degranulation.87 so their absence likely reduces collagen deposition.91
Contrarily, a study examining HT and normo- Broek et al. also found increased and prolonged ex-
trophic (NT) scars on human subjects found de- pression of anti-inflammatory macrophages in HT
layed and prolonged macrophage infiltration in scar compared with NT scar, and decreases in in-
HT scars compared with NT scars, suggesting an flammatory gene expression (including IL-10) over a
important role for initial macrophage infiltration 52-week period.88 Hence, it is possible that the
rather than later-stage macrophage presence in macrophages present were remaining in the profi-
reducing fibrosis.88 brotic polarization state rather than transitioning to
IMMUNE REGULATION OF SKIN WOUND HEALING 9

an antifibrotic M(IL-10)-like polarization, leading to TH1 cells primarily secrete IFN-c, IL-2, and TNF-a,
the HT scar development. while TH2 cells primarily secrete IL-4, 5, 6, 9, 10,
and 13, many of which are proinflammatory or
Role of mast cells in skin scarring. The final profibrotic.23 Additionally, epidermal cdT cells re-
immune cell subtype about which there exists sub- spond within hours to tissue damage, releasing
stantial literature is the mast cell, though the effect numerous growth factors to promote keratinocyte
of mast cells in scar formation remains somewhat proliferation, immune cell infiltration, and expedite
under debate.92 Mast cells are tissue resident and wound closure in mice.101 The effect of cdT cells on
become activated following tissue injury, releasing scar formation remains unknown, though data
granules into the extracellular space. They secrete summarized in previous paragraphs correlating
TGF-b1, as well as other proteases that promote fi- early macrophage infiltration with increased scar
brotic responses in fibroblasts and collagen fibril size suggest that cdT cells may increase scarring.101
formation.92 Mast cells also form gap junctions with Together, these data suggest T cells play an impor-
fibroblasts to stimulate myofibroblast differentia- tant role in wound repair and scar formation and
Downloaded by Gothenburg University Library from online.liebertpub.com at 02/12/18. For personal use only.

tion, proliferation, and ECM contraction.93–96 It has support the need for controlling the immune system.
been found that mice treated with disodium cro-
moglycate (DSCG) to inhibit mast cell activation Role of other immune cell subtypes in skin scar-
results in a reduced scar width, higher fibrillar ring. Research into the role of eosinophils, den-
density, and collagen fibers that were oriented more dritic cells, and B cells on cutaneous scar formation
similarly to normal tissue compared to controls. is also lacking. Eosinophils stimulate fibroblasts to
DSCG treated mice also showed decreased neutro- upregulate a-smooth muscle actin and collagen
phil recruitment and fewer proinflammatory cyto- secretion in bleomycin-induced models of fibrosis
kines.97 There are significantly less dermal mast and secrete growth factors that promote keratino-
cells in second trimester fetal wounds compared to cyte migration in vitro, suggesting a possible, but
third trimester wounds, and almost no degranula- unknown, role in dermal scar formation.78 Den-
tion. Interestingly, injecting mast cell lysates into dritic cells are known to play a role in the rate of
E15 wounds resulted in scarring, and wounds in reepithelialization, but they likely do not play an
E18 mast cell-deficient mice healed with signifi- important role in scarring.78 Finally, one study
cantly less scarring than wild-type littermates.98 showed that mice deficient in CD19, a critical pos-
There is also less mast cell degranulation in oral itive regulator of B cells, showed reduced recruit-
mucosal wounds compared with cutaneous wounds, ment of neutrophils and macrophages to the wound
and higher mast cell numbers in HT scars compared site, reduced proinflammatory and profibrotic cy-
with normal scars.82,92 Contrarily, a study by Will- tokines (including TGF-b), reduced granulation
enborg et al. showed genetic ablation of mast cells tissue formation, and slower reepithelialization.102
does not alter the amount of scar tissue, or the in- This suggests that depleting B cells may be an in-
filtration of leukocytes into the injury site.99 Fur- teresting therapeutic avenue for scar prevention,
ther research into the role of mast cells in scar though there is minimal supporting research.
formation is needed to resolve this discrepancy.

Role of T cells in skin scarring. Though they are FUTURE DIRECTIONS


known to play a role in immune cell recruitment and Therapeutic strategies targeting the immune
cytokine expression, the role of T cells on scar de- system for chronic wounds
velopment is unclear. Recently, Nosbaum et al. Since chronic wounds are often the result of ex-
found that mice depleted of Tregs have significantly cessive inflammation, controlling the immune re-
slower wound closure and increased granulation sponse is an attractive avenue for designing novel
tissue compared to wild-type mice partly due to their regenerative strategies. To this end, numerous
suppression of IFN-c, which reduces the recruit- material-based and molecular strategies have been
ment of proinflammatory macrophages.45 An earlier explored, including targeting the immune response
study showed that CD4-deficient mice had reduced using cytokines, protease inhibitors, miRNA, small
neutrophil recruitment to the wound site, whereas interfering RNA (siRNA) and extracellular vesicles
CD8-deficient mice had less neutrophils and mac- (EVs) (Fig. 5).
rophages. The changes in cytokine expression also
differ according to whether mice are CD4 deficient Biomaterial-based strategies to resolve chronic
or CD8 deficient.100 While there are many subsets of wounds. Various biomaterials have shown inter-
CD4 lymphocytes, two main types are TH1 and TH2. esting results for promoting wound resolution
Downloaded by Gothenburg University Library from online.liebertpub.com at 02/12/18. For personal use only. 10 LAROUCHE ET AL.

Figure 5. Molecular and cellular mechanisms involved in scar formation and dermal regeneration. (A) Immune cells act along numerous, redundant
pathways to promote excess ECM deposition and hyperproliferation of keratinocytes, leading to scar formation. Key immune cell types involved in pathologic
scar formation include macrophages, mast cells, neutrophils, eosinophils, and T cells. (B) Dermal regeneration is improved by generally decreasing innate and
adaptive immune cell recruitment and activation, and encouraging antifibrotic macrophage polarization (e.g. via IL-10). Pathways that induce antifibrotic
macrophage polarization in vivo are still very elusive, though research on other tissues suggests that Tregs may help promote antifibrotic macrophage
polarization and suppress proinflammatory immune responses. Black arrows indicate differentiation, blue arrows indicate inhibition and red arrows indicate
induction. IL, interleukin; Treg, regulatory T cells.

by modulating the immune system. Current mimic the natural ECM structure by imprinting
biomaterial-based strategies focus either on mac- small patterns on the surface, which improves cell
rophage adhesion and recruitment, or on directing adhesion, proliferation, and migration.111 These
their polarization to encourage inflammation res- patterns not only affect the function of epithelial
olution. One method of controlling macrophage cells, endothelial cells, and fibroblasts, but they
polarization is to modify the surface chemistry also promote anti-inflammatory macrophage po-
of the biomaterial. Indeed, macrophage adhesion is larization by encouraging a specific cell shape.111
enhanced on hydrophobic surfaces, and inhibited While the effect of incorporating complex matrix
on hydrophilic surfaces. However, even though topography onto micro-patterned gels has shown
fewer macrophages and foreign body giant cells enhanced skin regeneration and graft perfor-
(FBGCs) adhere to hydrophobic surfaces, those mance in animal models of chronic wounds, its
that do adhere secrete more cytokines than mac- effect on immune cells in this context remains
rophages/FBGCs adhered to hydrophilic/charged elusive.112
surfaces.103 This may result from the unique acti- The biomaterial origin also affects its immu-
vation of biomaterial-adherent macrophages and a nomodulatory properties. Synthetic biomaterials
phenotypic switch that occurs over time.82 Fur- can be advantageous because they usually avoid
thermore, it has been demonstrated that monocyte- an undesirable host immune response driven by
derived macrophages change their surface protein the antigens in naturally derived materials.17 For
expression depending upon the chemical composi- example, treatment of wounds in diabetic mice
tion of the biomaterial surface they are in contact with poly(methacrylic acid-co-methyl methacry-
with.104 These findings highlight the role of bio- late) (PMMA) beads accelerated chronic wound
material surface chemistry in altering macrophage healing, presumably by increasing sonic hedge-
response and activation. hog signaling (Shh).113 Shh is implicated in CD4
Another important factor to biomaterial design T cell activation and increased proliferation of
is the surface topography. Indeed, various studies hematopoietic stem cells (HSCs) and keratino-
have shown the importance of surface topography cytes.114,115 On the other hand, naturally derived
in altering macrophage response.105–110 For ex- biomaterials can more effectively replace the
ample, surface topography has been altered to lost components of the original ECM. Naturally
IMMUNE REGULATION OF SKIN WOUND HEALING 11

derived biomaterials that are commonly used ment of progenitor cells to the site of injury.129,130
for chronic wound resolution include hyaluronic Therefore, DDHAM appears to be an effective
acid (HA), chitosan, fibrin, and collagen.113 For option for designing regenerative strategies for
example, using chitosan-based biomaterial N- chronic wounds.
carboxymethyl chitosan, 5-methyl pyrrolidinone
chitosan (MPC) to deliver neurotensin to dia- Cytokine-based strategies to resolve chronic
betic wounds in mice induced rapid healing wounds. Cytokines are obviously critical during
(50% wound area reduction) by reducing levels of the time course of wound healing, making them
TNF-a, inflammatory cells, and MMP-9 at the site an attractive target for therapeutic development.
of injury.116 Several other studies have shown simi- For example, IL-1b is part of a proinflammatory
lar results using chitosan-based biomaterials for positive feedback loop that sustains a persistent
chronic wound repair.117–119 Additionally, a clinical proinflammatory wound macrophage phenotype,
trial delivering PRP in a HA scaffold accelerated contributing to impaired healing of diabetic
wound closure by 48% in 3 days, and lead to com- wounds.62,131 A study by Rita and colleagues showed
Downloaded by Gothenburg University Library from online.liebertpub.com at 02/12/18. For personal use only.

plete closure after two doses.5 PRP is a known a significant increase in wound closure in diabetic
source of numerous proresolution growth factors mice using an antibody to inhibit IL-1b. This down-
and cytokines, including PGDF, TGF-b1, TGF-a, regulated proinflammatory macrophages and upre-
granulocyte macrophage colony-stimulating factor gulated prohealing factors in the wound.131 Using
(GM-CSF, also known as colony-stimulating factor IL-1 receptor agonist (IL-1Ra) is another means of
2), and others.120 blocking IL-1 signaling pathways. Indeed, interac-
Among naturally derived biomaterials, decel- tion between IL-1b and IL-1Ra with IL-1R is critical
lularized ECM possesses immunomodulatory prop- for epithelial wound healing, and the IL-1Ra:IL-1
erties. Decellularized ECM is isolated from donor ratio is known to be higher in fluids from healing
tissues by removing the cellular components using wounds than those from chronic wounds (480:1 vs.
mechanical, chemical, or enzymatic methods.121 A 7:1).62,132 Not surprisingly, blocking IL-1 using IL-
common origin for ECM-based biomaterials is por- 1Ra has been shown to accelerate wound healing in
cine small intestinal submucosa, which has been diabetic mouse corneas. TNF-a is a second proin-
shown to modulate the wound healing microen- flammatory cytokine that contributes to a chronic
vironment in rats via macrophage polarization.122 wound state. Neutralizing TNF-a at the incision site
Interestingly, one mechanism by which decellular- significantly accelerated chronic wound healing
ized ECM modulates macrophage polarization is in mice by decreasing inflammatory cell infil-
through matrix-bound nanovesicles (MBVs), which trates and suppressing proinflammatory macro-
survive the chemical and enzymatic processes of phage activation.133
decellularization. These MBVs contain miRNA that SDF-1 is another promising chemokine to
polarizes macrophages to an anti-inflammatory promote wound healing. For example, delivering
phenotype.123 Decellularized and dehydrated hu- nanosized SDF-1 liposomes to diabetic mouse
man amniotic membrane (DDHAM) products have wounds promotes dermal cell proliferation, in-
also demonstrated potential for resolving chronic creases granulation tissue formation, and accel-
wounds and are clinically available. Indeed, nu- erates wound closure.134 Moreover, the liposomes
merous clinical trials have shown that DDHAM protect SDF-1 from degradation by proteases and
products successfully promote the resolution of serine exopeptidase. However, heparin sulfate
chronic wounds that are unresponsive to traditional present in the wound bed causes dimerization of
therapies.124–127 Moreover, in a clinical trial evalu- the SDF-1 liposomes, thereby reducing their bio-
ating DDHAM allograft safety, patients with vari- availability. A more effective delivery method
ous types of chronic wounds did not experience any could probably increase the effectiveness of SDF-1
product-related adverse effects.124 DDHAM pro- treatments.
motes wound resolution by stimulating the secre- GM-CSF, which facilitates differentiation of
tion of important growth factors, cytokines and HSCs to granulocytes and macrophages, also
proteases, including PDGFs, TGF-a, TGF-b1, FGF- plays key role in wound healing. GM-CSF has
2, epidermal growth factor, placental growth factor, been shown to accelerate wound closure in diabetic
granulocyte colony-stimulating factor (also known mice by increasing reepithelialization, recruiting
as colony-stimulating factor 3), IL-4, IL-10, and leukocytes, increasing angiogenesis, and upregu-
various TIMPs.128 DDHAM also increases the re- lating proinflammatory mediators, such as IL-6
lease of stromal cell-derived factor (SDF)-1, which is and macrophage chemoattractant protein (MCP)-
reduced in diabetic wounds and aids in the recruit- 1.135 Considering proinflammatory mediators are
12 LAROUCHE ET AL.

known to exacerbate chronic wounds when present for resolving inflammation during skin wound
for long periods of time, this study is somewhat healing, a similar strategy may be useful for
paradoxical. However, numerous other studies chronic wounds.148 However, no cytokine-based,
support GM-CSF effectiveness in resolving chronic chronic wound therapies have reached clinical
wounds.136–137 Clinical translation has still not trials; we need a more effective delivery method to
been achieved, though, because most of these facilitate clinical translation.
studies used intradermal or subcutaneous injec-
tions to deliver GM-CSF to the wound site, which Protease inhibitor-based strategies to resolve
raised concerns over injection pain, toxicity, side chronic wounds. The microenvironment of
effects, and uneven distribution.140 As an alterna- chronic wounds not only has reduced growth factor
tive, GM-CSF has been delivered using alginate. In levels, but also has an imbalance of MMPs and
clinical trials, this delivery method accelerated TIMPs. This can lead to the degradation of exoge-
wound healing and relieved pain.140 Altogether nously delivered growth factors, exacerbating the
GM-CSF is a potent cytokine for chronic wounds, problem.149 Thus, one way to overcome this issue is
Downloaded by Gothenburg University Library from online.liebertpub.com at 02/12/18. For personal use only.

but more efficient delivery methods are still re- to deliver protease inhibitors. For example, deliv-
quired for clinical translation. ering ND-336, which selectively inhibits MMP-2,
Delivery of other cytokines has also shown MMP-9, and MMP-14, accelerates wound closure
promise in resolving chronic wounds. Surprisingly, in diabetic mice by decreasing inflammation, en-
IL-22, a proinflammatory cytokine, helps resolve hancing reepithelialization, and increasing angio-
chronic diabetic wounds in mice by inducing kera- genesis (Table 2).150 Incorporating protease
tinocyte proliferation and signal transducer and inhibitors into wound dressings has also been ex-
activator of transcription 3 (STAT3) activation.141 plored. For example, combining collagen and oxi-
Additionally, anti-inflammatory cytokines have dized regenerated cellulose matrix into wound
been widely used for macrophage polarization. dressings improves wound repair by binding to
For example, recombinant IL-10 has been incor- and inactivating MMPs.151 Silver dressings and
porated into dextrin nanogel matrix to resolve hydrogels, which are antimicrobial have also
chronic wounds, though clinical trial results were shown promising results in porcine models and
not promising.142,144 Treatment of skin wounds in clinical trials.152,153 In addition, silver inactivates
diabetic mice using TGF-b1 also rescued wound proteases by displacing zinc from MMPs, which
healing and normalized macrophage polarization.72 promotes faster granulation tissue formation.152
Though not itself a cytokine, using MALP-2 in Finally, incorporating oleic acid/albumin formula-
diabetic mice promoted wound healing by increas- tions into topically applied cotton fiber dressings
ing macrophage infiltration and directing anti- targets neutrophil elastase in vitro, suggesting that
inflammatory polarization.144 MALP-2 stimulates it may help resolve chronic wounds.154
in vivo synthesis of macrophage inflammatory
protein (MIP)-1a, MIP-2, and MCP-1.145 Phase I miRNA- and siRNA-based strategies to resolve
clinical trial results using MALP-2 to treat diabetic chronic wounds. miRNAs are short, noncoding
wounds in 12 patients were also promising.6 RNA molecules that repress gene expression by
Since prolonged presence of proinflammatory binding to mRNA targets, either causing degrada-
cytokines may prevent resolution, and both pro- tion or inhibiting translation. Interestingly, miR-
and anti-inflammatory cytokines are necessary for NAs can modulate multiple genes, and studies in
acute wound healing, sequential delivery of pro- diabetic mouse models have shown that they play a
and anti-inflammatory cytokines could be an in- key role in orchestrating the inflammatory response
teresting strategy for improving chronic wound during wound healing. An imbalance of these sig-
healing. This concept has been explored in other nals may result in an improper inflammatory cas-
tissues with some success. For example, to improve cade, leading to the development of chronic
bone repair, decellularized bone was engineered to wounds.155 Indeed, many have been implicated in
sequentially release IFN-c and IL-4 and implanted the regulation of chronic wounds, including miR-21,
in mice at the site of injury. The sequential release -99, and -132.156–158 For example, miR-21 promotes
promoted macrophage polarization to switch from fibroblast recruitment by modulating the TGF-b
a pro- to an anti-inflammatory phenotype, result- pathway, which is significantly reduced in diabetic
ing in improved wound healing.146 Similar strate- wounds.156 miR-99 overexpression in diabetic
gies for releasing cytokines in a biphasic manner wounds increases the migration and proliferation
have been reviewed by Alvarez et al.147 Consider- of keratinocytes by regulating the PI3K/Akt path-
ing crosstalk between IFN-c and TGF-b is essential way, which is implicated in proinflammatory cyto-
IMMUNE REGULATION OF SKIN WOUND HEALING 13

Table 2. List of potential immunomodulatory therapeutics for chronic wounds

Pharmaceutical Effects Outcome Species References


113
PMMA Activates T cells Increases wound closure Diabetic mouse
Increases HSC and keratinocyte proliferation
62,131,132
IL-1Ra Downregulates inflammation triggered by IL-1R Increases wound closure Diabetic mouse
134
SDF-1 Increases dermal cell proliferation promotes granulation tissue formation Increases wound closure Diabetic mouse
135–140
GM-CSF Increases re-epithelialization and angiogenesis Increases wound closure Diabetic mouse
Promotes leukocyte recruitment
Increases collagen deposition
Increases proinflammatory cytokine production
141–143
IL-22 Increases keratinocyte proliferation Accelerates wound healing Diabetic mouse
72
TGF-b1 Promotes ECM deposition Improves diabetic wound healing Diabetic mouse
Promotes granulation tissue formation directs macrophage polarization
6,144,145
MALP-2 Macrophage infiltration and activation Promotes early wound closure Diabetic mouse
Phase I
150
ND-336 Inhibits MMPs Accelerates wound closure Diabetic mouse
Downloaded by Gothenburg University Library from online.liebertpub.com at 02/12/18. For personal use only.

Increases reepithelialization and angiogenesis Reduces inflammation


156
miR-21 Modulates TGF-b pathway Accelerates wound closure Diabetic mouse
157
miR-99 Regulates PI3K/AKT pathway Accelerates wound closure Diabetic mouse
158
miR-132 Decreases chemokine secretion reduces leukocyte recruitment Promotes transition into Diabetic mouse
proliferative phase
162
miR-126-3p Promotes reepithelialization and angiogenesis Accelerates wound closure Diabetic mouse
163–165
siRNA Regulates immune cell infiltration decreases protease activity Accelerates wound closure Diabetic mouse
167
Fibrocyte-derived Increases keratinocyte proliferation activates fibroblasts Accelerates wound closure Diabetic mouse
exosomes
5,120
PRP gels Source of cytokines and growth factors Improves healing in diabetic patients Phase I
124–130
DDHAM Increase cytokine/growth factor production Improves healing in diabetic patients Phase I
Increases progenitor cell recruitment
152,153
Silver dressings Antimicrobial Accelerates wound closure Clinically available
Inhibits MMPs
Promotes granulation tissue formation

DDHAM, dehydrated human amniotic membrane; ECM, extracellular matrix; GM-CSF, granulocyte macrophage colony-stimulating factor; HSC, hematopoietic
stem cell; IL, interleukin; IL-1Ra, IL-1 receptor agonist; MALP, macrophage-activating lipopeptide; PMMA, poly(methacrylic acid-co-methyl methacrylate); PRP,
platelet-rich plasma; SDF, stromal cell-derived factor; siRNA, small interfering RNA; TGF, transforming growth factor.

kine secretion and the inhibition of Tregs.157,159,160 and reepithelization of full-thickness skin defects
Finally, miR-132 promotes the transition from the in a diabetic rat model.162 However, these miRNA
inflammatory phase to the proliferation phase of delivery methods have yet to be translated into
wound healing by decreasing chemokine secretion the clinic and more efficient delivery strategies
and suppressing the NF-jB pathway, thereby re- are still needed.
ducing leukocytes recruitment.158 Altering miR- siRNA has been widely used for gene silencing
NA levels at the site of injury is therefore a purposes to improve wound healing (Table 2). For
promising strategy for designing chronic wound instance, siRNA against the kelch-like ECH asso-
therapies (Table 2). ciated protein 1 improved wound closure in a dia-
Unfortunately, despite the pivotal role of miR- betic mouse model by repressing NF-E2-related
NA in regulating chronic wounds, this path has factor 2. This siRNA was delivered in a lipopro-
not been extensively explored and miRNA-based teoplex nanoparticle based system and helped
therapeutic strategies are scarce due to the lack of maintain normal levels of inflammatory cells
optimal delivery systems. Current delivery sys- and ROS.163 MMP-9 siRNA delivered by b-CD-
tems are based on soluble injections and oligonu- (D3)7, a cationic, star-shaped polymer, to diabetic
cleotide conjugation, both of which are inefficient rat wounds also accelerated wound closure rate by
due to bio-fluid degradation. To overcome this decreasing MMP-9 expression.164 Though not yet
issue, Li et al. developed a peptide-based, self- tested in the context of chronic wounds, delivering
assembled, three-dimensional (3D), supramolec- MMP-9 siRNA using liposomes may be more ef-
ular hydrogel that facilitated miRNA delivery to fective as they have higher rates of interfer-
encapsulated cells, even in the presence of fetal ence.165 Overall, miRNAs and siRNAs have shown
bovine serum.161 In another study, miR-126-3p promising potential to modulate the immune system
was overexpressed in synovium mesenchymal and accelerate wound healing, though none have
stem cells (MSCs), which promoted angiogenesis reached clinical trials. Improvements in delivery
14 LAROUCHE ET AL.

methods are still required for their use as thera- Hence, immunomodulatory biomaterials that fa-
peutics in the clinic. cilitate scarless healing are very desirable. Many
recent studies have focused specifically on immu-
EV-based strategies to resolve chronic wounds. nomodulatory biomaterials that reduce scar for-
EVs are spherical particles enclosed by a phos- mation by delivering MSCs or adipose-derived
pholipid bilayer that range from 30 to 2,000 nm stem cells (ASCs) to the wound area (Table 3).
in diameter. They are very important in cell–cell MSCs have been successfully encapsulated in and
communication, facilitating the exchange of bio- delivered to wounds in animal models by gelatin
molecules such as cytokines, DNA, non-coding microspheres and microcryogels, or loaded into a
RNAs, MMPs, miRNAs, and mRNAs. These bio- 3D graphene foam.169 It has been shown that they
molecules modify recipient cell protein production, release prostaglandin E2 (PGE2), which sup-
gene expression, and behavior in response to local presses the release of proinflammatory cytokines
environmental factors.166 EVs are released by (TNF-a, IFN-c, IL-6, IL-8, and IL-12p70) and in-
every cell type in the ECM and are rapidly taken up creases the release of anti-inflammatory cytokines
Downloaded by Gothenburg University Library from online.liebertpub.com at 02/12/18. For personal use only.

by the targeted cells. Thus, because of these prop- (IL-10 and IL-12p40) and TGF-b1 by macro-
erties, EVs can potentially be used as immuno- phages.170,171 Additionally, PGE2 attenuates the
modulatory molecules to promote wound healing. proliferation of T cells in the wound and is a co-
For example, delivering human fibrocyte-derived factor in the transition from TH1 to TH2 cells,
exosomes has been shown to accelerate wound which favor inflammation resolution and, most
closure in diabetic mice by promoting angiogenesis, likely, tissue regeneration.170 The altered cyto-
increasing keratinocyte migration and prolifera- kine release profile inhibits further neutrophil
tion, and activating fibroblasts. This was a result of invasion and respiratory burst, which causes local
increased expression of heat shock protein-90a, tissue damage and likely increases scar size.171
STAT3, and certain miRNAs, including miR-124 Delivering ASCs in polyhydroxybutyrate-co-
(anti-inflammatory), miR-126 (proangiogenic), and hydroxyvalerate constructs achieves a similar out-
miR-21 (regulates collagen deposition).167 EVs can come in rats and may be more clinically appropriate
also promote wound healing by directing macro- since the yield following isolation is much higher
phage polarization. For example, exosomes from than MSCs.169,172
supernatant of lipopolysaccharide-preconditioned Aside from the delivery of stem cells, a few other
mesenchymal stromal cells promote wound closure immunomodulatory biomaterial strategies have
by regulating the TLR4/NF-jB/STAT3/AKT sig- been explored, some of which are also being ex-
naling pathway in diabetic rats.168 These studies plored to promote chronic wound resolution (Ta-
demonstrate the therapeutic potential of EVs for ble 3). Treatment of dermal wounds in children
promoting wound resolution by modulating the with neonatal foreskin tissue cultures leads to
immune system. However, like other therapeutic wound closure and tissue regeneration with mini-
strategies, EV-based therapies have yet to enter mal scarring.173 Since scarless fetal wound healing
clinical trials and more research is required to is dependent upon the reduced inflammatory re-
improve delivery methods. sponse, it is likely that treatment of skin wounds
with fetal tissue cultures also reduces scarring
Therapeutic strategies targeting the immune in adult wounds by attenuating the inflammatory
system for skin scar prevention and immune response.169 Alternatively, delivering
Since the immune system has been shown to play ECM proteins that are more abundant in fetal
such a critical role in modulating scar formation, wounds has shown potential for decreasing scar
recent research has explored both pharmaceuticals formation. For example, HA hydrogels minimize
and the development of biomaterials that target scarring by reducing TGF-b1 secretion in rabbit
immune cells to promote scarless wound healing. and rat models.169 Furthermore, incorporating
Leukocytes, namely neutrophils, macrophages, hyaluronan and type III collagen into collagen-
mast cells, and Tregs, are the most common targets heparin scaffolds in an attempt to recapitulate the
for many of these therapeutics (Fig. 6). fetal ECM may also aid in scar prevention.113
The chemo-physical properties are also an im-
Material-based strategies to reduce scar- portant parameter. Though not studied in relation
ring. Clinically, even when administering a to dermal scar formation, softer gels and materials
pharmaceutical to promote wound healing, large with 1–5 lm wide gratings reduce the inflammatory
dermal wounds require bandaging to protect ex- response and encourage anti-inflammatory mac-
posed tissue and prevent excessive fluid loss. rophage polarization.174 In addition to increasing
Downloaded by Gothenburg University Library from online.liebertpub.com at 02/12/18. For personal use only. IMMUNE REGULATION OF SKIN WOUND HEALING 15

Figure 6. Therapeutic strategies for cutaneous wound regeneration that are targeting the immune system. Therapeutics for scar prevention should, in
general, promote antifibrotic macrophage polarization, and prevent mast cell degranulation and inflammatory macrophage polarization/recruitment. Pending
future studies confirming the role of Tregs in skin regeneration, therapeutics should also promote their recruitment. This is currently being done via delivery of
cytokines, chemokines, other antifibrotic mediators, and stem cells, though delivering siRNA, miRNA, and EVs may also be promising. Biomaterials present a
promising means of delivering antifibrotic modulators, and their properties can also encourage macrophage polarization. Black arrows indicate differentiation,
blue arrows indicate inhibition, and red arrows indicate induction.

stiffness, cross-linking changes the ultrastructure, elicit detrimental immune responses to promote
composition and surface topology, and prevents scar development.155 Further elucidating the
material degradation and release of important bio- mechanisms by which these biomaterials reduce
active matricryptic peptides from naturally derived the inflammatory response, and developing novel
materials.155 Indeed, when using naturally derived materials that exploit numerous of these proper-
materials, chemical cross-linking triggers a chronic ties could lead to considerable improvements in
foreign body response. Finally, large amounts wound care for scar-free dermal regeneration.
of cellular material resulting from incomplete
decellularization of naturally derived materials Molecular-based strategies to reduce scarring.
Recently, many different molecular-based immu-
nomodulatory strategies for preventing scar for-
Table 3. Biomaterial-based immunomodulatory strategies mation have been explored with some success
to reduce scarring (Table 4). For example, Chemerin15 (C15), a reso-
Material Property Function References lution mediator, has been shown to reduce both
169–172 neutrophil and macrophage infiltration in mice by
Deliver stem cells Reduces neutrophil recruitment,
prevents respiratory burst around 70% and 40%, respectively, and restricted
Encourages antifibrotic their area of infiltration nearly 10-fold, reducing
macrophage polarization scar size.175 Mechanistically, C15 competitively
Attenuates T cell proliferation,
inhibits its proinflammatory precursor, full-length
encourages TH2 transition
Deliver neonatal cells Attenuates inflammatory response 169,173 chemerin. Both proteins interact with the ChemR23
HA-based Reduces TGF-b1 secretion 113,169
receptor upregulated on neutrophils, macrophages,
155
Stiffness Softer gels encourage anti-inflammatory and keratinocytes following wounding, which plays
polarization and reduced inflammation
174 a role in cell recruitment and adhesion to activated
Surface topography 1–5 lm gratings encourage anti-inflammatory
macrophage polarization and platelets.175 Similarly, injecting alpha-melanocyte-
reduce inflammation stimulating hormone (aMSH), a neuropeptide with
89
Chemical cross-linking Triggers chronic foreign body strong anti-inflammatory and immunomodulatory
response (increases scarring)
activity, into mice 30 min before wounding resulted
HA, hyaluronic acid. in significantly smaller scars and a reduction of
16 LAROUCHE ET AL.

Table 4. List of potential immunomodulatory therapeutics to reduce skin scarring

Pharmaceutical Target Function Species Reference


175
C15 Neutrophils Reduces recruitment, restricts area of infiltration Mouse
Macrophages
176,177
aMSH Leukocytes Suppresses TNF-a and IL-6 mRNA expression Mouse
Lymphocytes
Mast cells Induces IL-10 generation of Tregs
178
PLCe Leukocytes Diminishes IL-6, CXCL-1, CXCL-2, and CCL-20 Mouse
179
CXCR4 agonist Lymphocytes Reduces recruitment Mouse
Monocytes
180
PDRN Leukocytes Reduces recruitment Rat
Mast cells Inhibits degranulation
169
Curcumin Leukocytes Reduces IL-1b, IL-6, and IL-8 Rabbit
182
FM Cytokine profile Reduces TGF-b1 expression Mouse
7,8
TGF-b3 Lymphocytes Induces Tregs Phase III clinical trial
Macrophages Affects polarization
Downloaded by Gothenburg University Library from online.liebertpub.com at 02/12/18. For personal use only.

185
M6P Cytokine profile Inhibits TGF-b1 and TGF-b2 Phase II clinical trial
3,185
IL-10 Lymphocytes Induces Tregs Phase II clinical trial
Macrophages Encourages antifibrotic macrophage polarization
186
Nefopam Macrophages Reduces migration and adhesion Phase I clinical trial

aMSH, alpha-melanocyte-stimulating hormone; C15, Chemerin 15; CCL, C-C chemokine ligand; CXCL, C-X-C chemokine ligand; CXCR, C-X-C motif chemokine
receptor; FM, fibromodulin; M6P, mannose-6-phosphate; PDRN, polydeoxyribonucleotide; PLCe, phospholipase Ce; TNF, tumor necrosis factor; Treg, regulatory T cells.

leukocytes and mast cells. Though the mechanism wounding showed decreased amounts of proin-
by which aMSH reduced scarring was not specifi- flammatory cytokines, increased amounts of anti-
cally studied, previous research has shown aMSH inflammatory cytokines, and had smaller scars
suppresses IL-1, TNF-a, IFN-c, and IL-6 mRNA 40 days postwounding.181 Curcumin is a natural
expression, and induces Tregs via IL-10.176,177 Mast material that also reduces proinflammatory cyto-
cells can be specifically targeted to reduce scar for- kines, including IL-1b, IL-6, and IL-8 to reduce
mation by administering stabilizers to prevent de- scarring in rabbits.169 Finally, though its mecha-
granulation, such as cromoglicic acid and ketotifen, nism of action is still somewhat unclear, fi-
using tyrosine kinase inhibitors to reduce mast cell bromodulin, a proteoglycan that participates in
numbers following injury, or by neutralizing chy- the assembly of collagen fibers in the ECM, was
mase activity.92 Another potential therapeutic tar- recently found to restore scar-free wound healing
get for reducing leukocyte recruitment is in late-stage gestational murine wounds by de-
Phospholipase Ce (PLCe). PLCe knockout mice have creasing TGF-b1 expression.182
significantly reduced levels of proinflammatory cy- While the strategies above are still in the phases
tokines and chemokines, including IL-6, CXCL-1, of initial exploration and preclinical development,
CXCL-2, and C-C chemokine ligand (CCL)-20, and four molecular-based strategies for reducing scar
heal dermal wounds with less scarring.178 SDF-1, formation have reached clinical trials: TGF-b3
which is potently chemotactic for lymphocytes and (planned trade name Avotermin), M6P, IL-10 (plan-
monocytes, binds to the CXCR4 and CXCR7 recep- ned trade name Prevascar), and nefopam (planned
tors. Targeting that pathway using CXCR4 agonist trade mane ScarX). TGF-b3 is a cytokine whose
CTCE-9908 decreased cell recruitment and reduced presence is associated with scar-free wound heal-
scarring in mice.179 Similarly, administration of ing. While its role on the immune response has yet
polydeoxyribonucleotide (PDRN) to excisional skin to be carefully evaluated, there is evidence that it
wounds in rats significantly reduced scarring. induces Tregs and affects macrophage polariza-
PDRN inhibits mast cell degranulation, decreases tion.90,183,184 Renovo Ltd. translated numerous
inflammatory cell recruitment, and reduces the preclinical studies showing a decrease in scarring
amount of proinflammatory mediators (including following local administration of TGF-b3 before
TNF-a, IL-6, and HMGB-1) at the wound site.180 and after operation into phase I and II clinical tri-
Interestingly, exploiting oral tolerance to common als.8 Further evaluation was halted after Avo-
dietary proteins, specifically zein, which is found in termin failed to meet its efficacy endpoints during
corn, can also result in less prominent scarring in phase III, potentially because the dose was halved.
mice. Since reexposure to orally tolerated proteins However, since temporal variation critically af-
blocks nonspecific inflammation, mice that received fects healing, continuing studies are exploring the
a parenteral injection of zein immediately before efficacy of continuously delivering TGF-b3 in de-
IMMUNE REGULATION OF SKIN WOUND HEALING 17

creasing scarring.7 M6P is a potent inhibitor of SUMMARY


TGF-b1 and TGF-b2. While it was successful in Recent findings have greatly improved our un-
phase I clinical trials, in the phase II exploratory derstanding of the roles of the immune system
trial neither topical nor intradermal application in acute wound healing. The immune response is
showed statistical significance.185 A phase II trial either dysregulated in chronic wounds, or leads
injecting human recombinant IL-10 into surgical to undesirable scar development. Macrophages
wound margins showed a statistically significant clearly play an important role, especially through
decrease in scar development. IL-10 not only in- their phenotypic changes. Further clarifying ac-
duces Tregs, but also leads to an anti-inflammatory, tivation pathways could yield therapies that di-
alternatively activated macrophage polarization.3 rectly impact their polarization to improve wound
In a second phase II trial that extended for longer outcomes. Additionally, Tregs have been shown to
time points following treatment, there was actually have regenerative properties in other tissues, yet
more scarring following IL-10 treatment compared there is a lack of information explaining their role
with placebo controls.185 Finally, topical nefopam, in wound resolution and scar formation.3
Downloaded by Gothenburg University Library from online.liebertpub.com at 02/12/18. For personal use only.

was first developed as a non-narcotic analgesic Ultimately, translating current and future
drug and attenuates b-catenin levels, thereby af- immune-modulatory therapeutics hinges on the
fecting the migration and adhesion of macro- development of improved drug delivery methods.
phages. Phase I clinical trials for its effectiveness Many therapies that have reached clinical trials
in reducing dermal scarring have just begun, failed because current delivery methods rely on
though around 30 years of data exist to demon- supra-physiological bolus doses rather than con-
strate its systemic safety.186 trolled, sustained release. Because of natural drug
Finally, interesting directions for future de- clearance, meeting both safety and efficacy bench-
velopment of molecular-based scar prevention marks in this way is incredibly challenging. This is
strategies may focus on chemokine receptor especially evident in chronic wound therapies,
pathways. There exist considerable data sup- which have fallen short largely because of inef-
porting both positive and negative roles of differ- ficient delivery methods. Hence, novel delivery
ent chemokines and cytokines in wound healing systems are critical for successful translation of
and scar formation,179 yet few of these pathways therapeutics currently being explored and those
have been explored as therapeutics. Some che- explored in the future. Perhaps certain strategies
mokine receptor pathways that show promise for that failed clinical trials in the past may become
successful therapeutic developments for skin re- successful using novel delivery strategies.
generation include C-X3-C chemokine motif re-
ceptor (CX3CR)1, C-C chemokine receptor (CCR)2
and CXCR3.179,187,188 ACKNOWLEDGMENTS
As previously emphasized, a major letdown of AND FUNDING SOURCES
many current strategies is their reliance on very This work was supported in part by the research
high concentration (supra-physiological) doses, grant of Astellas Foundation for Research on Me-
which do not remain at the wound site for long tabolic Disorders and by the Australian Research
enough without performing multiple deliveries. Council (DE170100398) to M.M.M. and a Whitaker
Hence, a critical aspect for these strategies is to International Fellowship awarded to J.L.
develop appropriate delivery systems. For exam-
ple, various protein engineering strategies have AUTHOR DISCLOSURE AND GHOSTWRITING
been developed to confer a high affinity of thera- No competing financial interests exist. The
peutics to the endogenous ECM.189 These ap- authors listed expressly wrote the content of this ar-
proaches allow the retention of the therapeutic at ticle. No ghostwriters were used to write this article.
the delivery site without using biomaterials.
Perhaps, using a similar delivery strategy would
improve the clinical efficacy of protein-based ABOUT THE AUTHORS
therapeutics for scar prevention. Extensive re- Jacqueline Larouche, BS, received her degree
search has also been done developing controlled in Biomedical Engineering from the Georgia In-
release polymers, though many remain untested stitute of Technology. She is currently a Whitaker
in the context of wound healing and scar forma- Fellow at the Australian Regenerative Medicine
tion.190 Ongoing research in this area will likely Institute (ARMI) in Monash University, where
potentiate therapeutic effectiveness of the she focuses on growth factor engineering to en-
molecular-based targets discussed herein. hance regeneration. Sumit Sheoran, MSc, re-
18 LAROUCHE ET AL.

ceived his degree from the Manipal


TAKE-HOME MESSAGES
University School of Regenerative Med-
icine and is currently a research fellow at  Immune cells and immune mediators are intimately involved in every
ARMI. Kenta Maruyama, MD, PhD, stage of wound healing. Early studies showed improved healing in im-
obtained his degrees from Keio Uni- munodeficient mice and fetuses, sparking increased research into the
versity and Osaka University School of differential role of immune cells in wound repair.
Medicine. After clinical residency at the  Chronic wounds are trapped in a chronic inflammatory state, precluding
National Hospital Organization Tokyo restoration of the normal anatomical structure and function in sequential
Medical Center, he joined the Im- manner.
munology Frontier Research Center at  In chronic wounds, increased biofilm load leads to imbalance between
Osaka University as a research fellow inflammatory and anti-inflammatory mediators. This creates a steady
and became Assistant Professor in 2014. state of hyper-inflammation and the immune system is unable to over-
His research focuses on bone-biology, whelm the bacterial load.
pain-biology, molecular-immuno-physiol-  Hyper-inflammation causes overproduction of MMPs and ROS and
Downloaded by Gothenburg University Library from online.liebertpub.com at 02/12/18. For personal use only.

ogy, and geriatric medicine. Mikaël M. decreases in TIMPs, contributing to development of chronic wounds.
Martino, PhD, obtained his degree in  Macrophage phenotype conversion from proinflammatory to anti-
Biotechnology and Bioengineering at the inflammatory macrophage is critical in the process of normal wound
Ecole Polytechnique Fédérale de Lau- healing. This conversion is impaired in chronic wounds.
sanne. Then, he became a postdoctoral
 Macrophages, mast cells, neutrophils, and most T cells promote rapid
fellow and an Assistant Professor in the
wound closure and scar formation. Inducing Tregs and promoting
Immunology Frontier Research Centre at antifibrotic macrophage polarization may help attenuate the immune
Osaka University, focusing on how the response to promote scarless healing.
immune system influences tissue re-
 Scar therapies mostly focus on reducing immune cell recruitment, in-
generation. In 2016, he became a Eur-
ducing Tregs or interfering with the macrophage polarization and TGF-b
opean Molecular Biology Laboratory
pathways.
(EMBL) Australia group leader at ARMI.
His long-term goal is to work at the in-  Development of a novel drug delivery system that controls therapeutic
terface between bioengineering and im- release at the wound site is required to safely and effectively resolve
munology for designing efficient chronic wounds and promote scarless healing.
regenerative medicine strategies.

REFERENCES
1. Sen CK, Gordillo GM, Roy S, et al. Human skin 6. Niebuhr M, Muhlradt PF, Wittmann M, Kapp skin injury and repair. Immunobiology 2011;216:
wounds: a major and snowballing threat to A, Werfel T. Intracutaneous injection of the 753–762.
public health and the economy. Wound Repair macrophage-activating lipopeptide-2 (MALP-2)
10. Diegelmann RF, Evans MC. Wound healing: an
Regen 2010;17:763–771. which accelerates wound healing in mice—a
overview of acute, fibrotic and delayed healing.
2. Weiser TG, Regenbogen SE, Thompson KD, et al. phase I trial in 12 patients. Exp Dermatol 2008;
Front Biosci 2004;9:283–289.
An estimation of the global volume of surgery: a 17:1052–1056.
modelling strategy based on available data. 11. Szpaderska AM, Egozi EI, Gamelli RL, DiPietro
7. Samadikuchaksaraei A, Mehdipour A, Roudkenar
Lancet 2008;372:139–144. LA. The effect of thrombocytopenia on dermal
MH, et al. A dermal equivalent engineered with
wound healing. J Invest Dermatol 2003;120:
3. Julier Z, Park AJ, Briquez PS, Martino MM. TGF-b3 expressing bone marrow stromal cells
1130–1137.
Promoting tissue regeneration by modulating the and amniotic membrane: cosmetic healing of
immune system. Acta Biomater 2017;53:13–28. full-thickness skin wounds in rats. Artif Organs 12. Martin P, Leibovich SJ. Inflammatory cells during
2016;40:E266–E279. wound repair: the good, the bad and the ugly.
4. Eming SA, Martin P, Tomic-Canic M. Wound
Trends Cell Biol 2005;15:599–607.
repair and regeneration: mechanisms, signalling, 8. Ferguson MWJ, Duncan J, Bond J, et al. Pro-
and translation. Wound Healing 2014;6:265sr6. phylactic administration of avotermin for im- 13. Deppermann C, Cherpokova D, Nurden P, et al.
provement of skin scarring: three double-blind, Gray platelet syndrome and defective thrombo-
5. Ramos-Torrecillas J, Garcia-Martinez O, De
placebo-controlled, phase I/II studies. Lancet inflammation in Nbeal2-deficient mice. J Clin
Luna-Bertos E, Ocana-Peinado FM, Ruiz C. Ef-
2009;373:1264–1274. Invest 2013;123:3331–3342.
fectiveness of platelet-rich plasma and hya-
luronic acid for the treatment and care of 9. Delavary BM, van der Veer WM, van Egmond 14. Martinez-Zapata MJ, Marti-Carvajal AJ, Sola
pressure ulcers. Biol Res Nurs 2015;17:152–158. M, Niessen FB, Beelen RHJ. Macrophages in I, et al. Autologous platelet-rich plasma for
IMMUNE REGULATION OF SKIN WOUND HEALING 19

treating chronic wounds. Cochrane Database 31. Nagaoka T, Kaburagi Y, Hamaguchi Y, et al. 47. Schneider DF, Palmer JL, Tulley JM, et al. A
Syst Rev 2012;10:CD006899. Delayed wound healing in the absence of in- novel role for NKT cells in cutaneous wound
tercellular adhesion molecule-1 or L-selectin repair. J Surg Res 2011;168:325–333.e321.
15. Martinez-Zapata MJ, Marti-Carvajal AJ, Sola
expression. Am J Pathol 2000;157:237–247.
I, et al. Autologous platelet-rich plasma for 48. Wynn TA. Cellular and molecular mechanisms of
treating chronic wounds. Cochrane Database of 32. Mori R, Kondo T, Ohshima T, Ishida Y, Mukaida fibrosis. J Pathol 2008;214:199–210.
Systematic Reviews 2016:CD006899. N. Accelerated wound healing in tumor necro-
sis factor receptor p55-deficient mice with re- 49. Fertin C, Nicolas JF, Gillery P, Kalis B, Ban-
16. Suthar M, Gupta S, Bukhari S, Ponemone V. chereau J, Maquart FX. Interleukin-4 stimulates
duced leukocyte infiltration. FASEB J 2002;16:
Treatment of chronic non-healing ulcers using collagen synthesis by normal and scleroderma
963–974.
autologous platelet rich plasma: a case series. J fibroblasts in dermal equivalents. Cell Mol Biol
Biomed Sci 2017;24:16. 33. DiPietro LA, Polverini PJ. Role of the macro- 1991;37:823–829.
phage in the positive and negative regulation of
17. Temenoff JS, Mikos AG. Biomaterials: The In- wound neovascularization. Behring Inst Mitt 50. Eming SA, Hammerschmidt M, Krieg T, Roers
tersection of Biology and Materials Science, 1st 1993:238–247. A. Interrelation of immunity and tissue repair
ed. Upper Saddle River, NJ: Pearson Prentice or regeneration. Semin Cell Dev Biol 2009;20:
Hall, 2008. 34. Lucas T, Waisman A, Ranjan R, et al. Differential 517–527.
roles of macrophages in diverse phases of skin
18. Kolaczkowska E, Kubes P. Neutrophil recruitment repair. J Immunol 2010;184:3964–3977. 51. Stojadinovic A, Carlson JW, Schultz GS, Davis
Downloaded by Gothenburg University Library from online.liebertpub.com at 02/12/18. For personal use only.

and function in health and inflammation. Nat TA, Elster EA. Topical advances in wound care.
Rev Immunol 2013;13:159–175. 35. Jetten N, Verbruggen S, Gijbels MJ, Post MJ, Gynecol Oncolol 2008;111(2 Suppl):S70–S80.
De Winther MP, Donners MM. Anti-inflammatory
19. Weiss SJ. Tissue destruction by neutrophils. N M2, but not pro-inflammatory M1 macrophages 52. Eming SA, Koch M, Krieger A, et al. Differential
Engl J Med 1989;320:365–376. promote angiogenesis in vivo. Angiogenesis 2014; proteomic analysis distinguishes tissue repair
17:109–118. biomarker signatures in wound exudates ob-
20. Brinkmann V, Reichard U, Goosmann C, et al.
tained from normal healing and chronic wounds.
Neutrophil extracellular traps kill bacteria. Sci- 36. Sallusto F, Baggiolini M. Chemokines and leu- J Proteome Res 2010;9:4758–4766.
ence 2004;303:1532–1535. kocyte traffic. Nat Immunol 2008;9:949–952.
53. Schultz GS, Mast BA. Molecular analysis of the
21. Wilgus TA, Roy S, McDaniel JC. Neutrophils and 37. Baggiolini M. Chemokines and leukocyte traffic. environments of healing and chronic wounds:
wound repair: positive actions and negative re- Nature 1998;392:565–568. cytokines, proteases and growth factors. Prim
actions. Adv Wound Care (New Rochelle) 2013;
38. Farrar MA, Schreiber RD. The molecular cell bi- Intention 1999;7:7–15.
2:379–388.
ology of interferon-gamma and its receptor. 54. Ben-Porath I, Weinberg RA. The signals and
22. Devalaraja RM, Nanney LB, Du J, et al. Delayed Annu Rev Immunol 1993;11:571–611. pathways activating cellular senescence. Int
wound healing in CXCR2 knockout mice. J Invest
39. Xu P, Fu X, Xiao N, et al. Involvements of J Biochem Cell Biol 2005;37:961–976.
Dermatol 2000;115:234–244.
gammadeltaT lymphocytes in acute and chronic 55. Wysocki AB, Staiano-Coico L, Grinnell F. Wound
23. Zaja-Milatovic S, Richmond A. CXC chemokines skin wound repair. Inflammation 2017;40:1416– fluid from chronic leg ulcers contains elevated
and their receptors: a case for a significant bi- 1427. levels of metalloproteinases MMP-2 and MMP-
ological role in cutaneous wound healing. Histol
40. Jameson J, Ugarte K, Chen N, et al. A role for 9. J Invest Dermatol 1993;101:64–68.
Histopathol 2008;23:1399–1407.
skin gammadelta T cells in wound repair. Sci- 56. Weckroth M, Vaheri A, Lauharanta J, Sorsa T,
24. Dovi JV, He LK, DiPietro LA. Accelerated wound ence 2002;296:747–749. Konttinen YT. Matrix metalloproteinases, gela-
closure in neutrophil-depleted mice. J Leukoc
41. Pang DJ, Neves JF, Sumaria N, Pennington DJ. tinase and collagenase, in chronic leg ulcers.
Biol 2003;73:448–455.
Understanding the complexity of cd T-cell sub- J Invest Dermatol 1996;106:1119–1124.
25. Willenborg S, Eming SA. Macrophages—sen- sets in mouse and human. Immunology 2012; 57. Saarialho-Kere UK. Patterns of matrix metallo-
sors and effectors coordinating skin damage and 136:283–290. proteinase and TIMP expression in chronic ulcers.
repair. J Ger Soc Dermatol 2014;12:214–221.
42. Toulon A, Breton L, Taylor KR, et al. A role for Arch Dermatol Res 1998;290 Suppl:S47–S54.
26. Murray PJ, Wynn TA. Protective and pathogenic human skin-resident T cells in wound healing. J
58. Grinnell F, Zhu M. Fibronectin degradation in
functions of macrophage subsets. Nature 2011; Exp Med 2009;206:743–750.
chronic wounds depends on the relative levels
11:723–737.
43. Lei H, Schmidt-Bleek K, Dienelt A, Reinke P, Volk of elastase, alpha1-proteinase inhibitor, and
27. Macdonald KP, Palmer JS, Cronau S, et al. An H-D. Regulatory T cell-mediated anti-inflammatory alpha2-macroglobulin. J Invest Dermatol 1996;
antibody against the colony-stimulating factor 1 effects promote successful tissue repair in both 106:335–341.
receptor (CSF1R) depletes the resident subset indirect and direct manners. Front Pharmacol
59. Grinnell F, Ho CH, Wysocki A. Degradation of
of monocytes and tissue and tumor-associated 2015;6:184.
fibronectin and vitronectin in chronic wound
macrophages but does not inhibit inflammation.
44. Liu G, Ma H, Qiu L, et al. Phenotypic and func- fluid: analysis by cell blotting, immunoblotting,
Blood 2010;116:3955–3963.
tional switch of macrophages induced by regu- and cell adhesion assays. J Invest Dermatol
28. Murray PJ, Allen JE, Biswas SK, et al. Macro- latory CD4+CD25+ T cells in mice. Immunol Cell 1992;98:410–416.
phage activation and polarization: nomenclature Biol 2011;89:130–142.
60. Rao CN, Ladin DA, Liu YY, Chilukuri K, Hou
and experimental guidelines. Immunity 2014;41:
45. Nosbaum A, Prevel N, Truong HA, Mehta P, ZZ, Woodley DT. Alpha 1-antitrypsin is degraded
14–20.
Ettinger M, Scharschmidt TC, Ali NH, Pauli ML, and non-functional in chronic wounds but intact
29. Leibovich SJ, Ross R. The role of the macro- Abbas AK, Rosenblum MD. Cutting edge: regu- and functional in acute wounds: the inhibitor
phage in wound repair. A study with hydrocor- latory T cells facilitate cutaneous wound heal- protects fibronectin from degradation by chronic
tisone and antimacrophage serum. Am J Pathol ing. J Immunol 2016;196:2010–2014. wound fluid enzymes. J Invest Dermatol 1995;
1975;78:71–100. 105:572–578.
46. Brubaker AL, Schneider DF, Kovacs EJ. Neu-
30. Subramaniam M, Saffaripour S, Van De Water trophils and natural killer T cells as negative 61. Herrick S, Ashcroft G, Ireland G, Horan M,
L, et al. Role of endothelial selectins in wound regulators of wound healing. Expert Rev Dermatol McCollum C, Ferguson M. Up-regulation of elas-
repair. Am J Pathol 1997;150:1701–1709. 2011;6:5–8. tase in acute wounds of healthy aged humans
20 LAROUCHE ET AL.

and chronic venous leg ulcers are associated with 77. Stojadinovic O, Yin N, Lehmann J, Pastar I, 93. Au SR, Au K, Saggers GC, Kame N, Ehrlich HP.
matrix degradation. Lab Invest 1997;77:281–288. Kirsner RS, Tomic-Canic M. Increased number of Rat mast cells communicate with fibroblasts via
Langerhans cells in the epidermis of diabetic gap junction intercellular communications. J Cell
62. Zhao R, Liang H, Clarke E, Jackson C, Xue M.
foot ulcers correlates with healing outcome. Biochem 2007;100:1170–1177.
Inflammation in chronic wounds. Int J Mol Sci
Immunol Res 2013;57:222–228.
2016;17:pii: E2085. 94. Foley T, Saggers GC, Moyer K, Ehrlich HP. Rat
78. Cash J, P. Martin. Myeloid cells in cutaneous mast cells enhance fibroblast proliferation and
63. Wong SL, Demers M, Martinod K, et al. Dia-
wound repair. Microbiol Spectr 2016;4. doi: fibroblast-populated collagen lattice contrac-
betes primes neutrophils to undergo NETosis,
10.1128/microbiolspec.MCHD-0017-2015 tion through gap junctional intercellular com-
which impairs wound healing. Nat Med 2015;
munications. Plastic Reconstr Surg 2011;127:
21:815–819. 79. Costerton JW, Stewart PS, Greenberg EP. Bac-
1478–1486.
terial biofilms: a common cause of persistent
64. Roth Flach RJ, Czech MP. NETs and traps delay
infections. Science 1999;284:1318–1322. 95. Pistorio A, Ehrlich HP. Modulatory effects
wound healing in diabetes. Trends Endocrinol
of connexin-43 expression on gap junction
Metab 2015;26:451–452. 80. Wolcott RD, Rhoads DD, Dowd SE. Biofilms and
intercellular communications with mast cells
chronic wound inflammation. J Wound Care
65. Fadini GP, Menegazzo L, Rigato M, et al. NE- and fibroblasts. J Cell Biochem 2011;112:
2008;17:333–341.
Tosis delays diabetic wound healing in mice 1441–1449.
and humans. Diabetes 2016;65:1061–1071. 81. Roy S, Elgharably H, Sinha M, et al. Mixed-
96. Foley T, Ehrlich HP. Through gap junction com-
Downloaded by Gothenburg University Library from online.liebertpub.com at 02/12/18. For personal use only.

species biofilm compromises wound healing by


66. Papayannopoulos V. Sweet NETs, bitter wounds. munications, co-cultured mast cells and fibro-
disrupting epidermal barrier function. J Pathol
Immunity 2015;43:223–225. blasts generate fibroblast activities allied with
2014;233:331–343.
hypertrophic scarring. Plast Reconstr Surg 2013;
67. Khanna S, Biswas S, Shang Y, et al. Macrophage
82. Wilgus TA. Immune cells in the healing skin 131:1036–1044.
dysfunction impairs resolution of inflammation
wound: influential players at each stage of re-
in the wounds of diabetic mice. PLoS One 2010; 97. Chen L, Schrementi ME, Ranzer MJ, Wilgus TA,
pair. Pharmacol Res 2008;58:112–116.
5:e9539. DiPietro LA. Blockade of mast cell activation
83. Tan J, Wu J. Current progress in understanding reduces cutaneous scar formation. PLoS One
68. Hanses F, Park S, Rich J, Lee JC. Reduced
the molecular pathogenesis of burn scar con- 2014;9:e85226.
neutrophil apoptosis in diabetic mice during
tracture. Burns Trauma 2017;5:14.
staphylococcal infection leads to prolonged 98. Wulff BC, Parent AE, Meleski MA, DiPietro LA,
Tnfalpha production and reduced neutrophil 84. Brant JO, Lopez MC, Baker HV, Barbazuk WB, Schrementi ME, Wilgus TA. Mast cells contrib-
clearance. PLoS One 2011;6:e23633. Maden M. A comparative analysis of gene ex- ute to scar formation during fetal wound heal-
pression profiles during skin regeneration in Mus ing. J Invest Dermatol 2012;132:458–465.
69. Loots MA, Lamme EN, Zeegelaar J, Mekkes JR,
and Acomys. PLoS one 2015;10:e0142931.
Bos JD, Middelkoop E. Differences in cellular 99. Willenborg S, Eckes B, Brinckmann J, et al.
infiltrate and extracellular matrix of chronic di- 85. Zhu Z, Ding J, Ma Z, Iwashina T, Tredget EE. Sys- Genetic ablation of mast cells redefines the
abetic and venous ulcers versus acute wounds. J temic depletion of macrophages in the subacute role of mast cells in skin wound healing and
Invest Dermatol 1998;111:850–857. phase of wound healing reduces hypertrophic scar bleomycin-induced fibrosis. J Invest Dermatol
formation. Wound Repair Regen 2016;24:644–656. 2014;134:2005–2015.
70. Snyder RJ, Lantis J, Kirsner RS, Shah V, Moly-
neaux M, Carter MJ. Macrophages: a review of 86. Amini-Nik S, Cambridge E, Yu W, et al. ß- 100. Chen L, Mehta ND, Zhao Y, DiPietro LA. Absence
their role in wound healing and their therapeutic Catenin-regulated myeloid cell adhesion and of CD4 or CD8 lymphocytes changes infiltration
use. Wound Repair Regen 2016;24:613–629. migration determine wound healing. J Clin In- of inflammatory cells and profiles of cytokine
vest 2014;126:2599–2610. expression in skin wounds, but does not impair
71. Sindrilaru A, Peters T, Wieschalka S, et al. An
healing. Exp Dermatol 2014;23:189–194.
unrestrained proinflammatory M1 macrophage 87. Dardenne AD, Wulff BC, Wilgus TA. The alar-
population induced by iron impairs wound min HMGB-1 influences healing outcomes in 101. Havran WL, Jameson JM. Epidermal T cells and
healing in humans and mice. J Clin Invest 2011; fetal skin wounds. Wound Repair Regen 2013; wound healing. J Immunol 2010;184:5423–5428.
121:985–997. 21:282–291.
102. Iwata Y, Yoshizaki A, Komura K, et al. CD19, a
72. Okizaki S, Ito Y, Hosono K, et al. Suppressed 88. Broek LJ, Veer WM, Jong EH, Gibbs S, Niessen response regulator of B lymphocytes, regulates
recruitment of alternatively activated macro- F. Suppressed inflammatory gene expression wound healing through hyaluronan-induced TLR4
phages reduces TGF-beta1 and impairs wound during human hypertrophic scar compared to signaling. Am J Pathol 2009;175:649–660.
healing in streptozotocin-induced diabetic mice. normotrophic scar formation. Exp Dermatol 2015;
103. Jones JA, Chang DT, Meyerson H, et al. Pro-
Biomed Pharmacother 2015;70:317–325. 24:623–629.
teomic analysis and quantification of cytokines
73. Hesketh M, Sahin KB, West ZE, Murray RZ. 89. Brown BN, Londono R, Tottey S, et al. Macro- and chemokines from biomaterial surface-
Macrophage phenotypes regulate scar formation phage phenotype as a predictor of constructive adherent macrophages and foreign body giant
and chronic wound healing. Int J Mol Sci 2017; remodeling following the implantation of bio- cells. J Biomed Mater Res A 2007;83:585–596.
18:pii: E1545. logically derived surgical mesh materials. Acta
104. Dinnes DL, Marcal H, Mahler SM, Santerre JP,
Biomater 2012;8:978–987.
74. Subramaniam K, Pech CM, Stacey MC, Wallace Labow RS. Material surfaces affect the protein
HJ. Induction of MMP-1, MMP-3 and TIMP-1 in 90. Rodero MP, Khosrotehrani K. Skin wound heal- expression patterns of human macrophages: a
normal dermal fibroblasts by chronic venous leg ing modulation by macrophages. Int J Clin Exp proteomics approach. J Biomed Mater Res A
ulcer wound fluid. Int Wound J 2008;5:79–86. Pathol 2010;3:643–653. 2007;80:895–908.
75. Eming SA, Krieg T, Davidson JM. Inflammation 91. Murray LA, Chen Q, Kramer MS, et al. TGF-beta 105. Yim EK, Leong KW. Significance of synthetic
in wound repair: molecular and cellular mecha- driven lung fibrosis is macrophage dependent nanostructures in dictating cellular response.
nisms. J Invest Dermatol 2007;127:514–525. and blocked by Serum amyloid P. Int J Biochem Nanomedicine 2005;1:10–21.
Cell Biol 2011;43:154–162.
76. Mendez MV, Stanley A, Park HY, Shon K, Phillips 106. Fink J, Fuhrmann R, Scharnweber T, Franke RP.
T, Menzoian JO. Fibroblasts cultured from ve- 92. Wilgus TA, Wulff BC. The importance of mast Stimulation of monocytes and macrophages:
nous ulcers display cellular characteristics of cells in dermal scarring. Adv Wound Care 2014; possible influence of surface roughness. Clin
senescence. J Vasc Surg 1998;28:876–883. 3:356–365. Hemorheol Microcirc 2008;39:205–212.
IMMUNE REGULATION OF SKIN WOUND HEALING 21

107. Curtis A, Wilkinson C. Topographical control of biologic scaffolds and in vivo relevance. Methods upregulation of proinflammatory cytokines. Br J
cells. Biomaterials 1997;18:1573–1583. 2015;84:25–34. Dermatol 2010;162:478–486.
108. Wojciak-Stothard B, Curtis A, Monaghan W, 122. Badylak SF, Valentin JE, Ravindra AK, McCabe 136. Jaschke E, Zabernigg A, Gattringer C. Re-
MacDonald K, Wilkinson C. Guidance and acti- GP, Stewart-Akers AM. Macrophage phenotype combinant human granulocyte-macrophage colony-
vation of murine macrophages by nanometric as a determinant of biologic scaffold remodel- stimulating factor applied locally in low doses
scale topography. Exp Cell Res 1996;223:426–435. ing. Tissue Eng Part A 2008;14:1835–1842. enhances healing and prevents recurrence of
chronic venous ulcers. Int J Dermatol 1999;38:
109. Chen S, Jones JA, Xu Y, Low HY, Anderson JM, 123. Huleihel L, Hussey GS, Naranjo JD, et al. Matrix-
380–386.
Leong KW. Characterization of topographical ef- bound nanovesicles within ECM bioscaffolds. Sci
fects on macrophage behavior in a foreign body Adv 2016;2:e1600502. 137. Da Costa RM, Ribeiro Jesus FM, Aniceto C,
response model. Biomaterials 2010;31:3479–3491. Mendes M. Randomized, double-blind, placebo-
124. Smiell JM, Treadwell T, Hahn HD, Hermans
controlled, dose-ranging study of granulocyte-
110. Waterfield JD, Ali TA, Nahid F, Kusano K, MH. Real-world experience with a decellularized
macrophage colony stimulating factor in patients
Brunette DM. The effect of surface topography dehydrated human amniotic membrane allograft.
with chronic venous leg ulcers. Wound Repair
on early NFkappaB signaling in macrophages. J Wounds 2015;27:158–169.
Regen 1999;7:17–25.
Biomed Mater Res A 2010;95837–847.
125. Zelen C, Serena T, Fetterolf D. Dehydrated hu-
138. Tursen U, Api H, Kaya TI, Cinel L, Ikizoglu G.
111. McWhorter FY, Wang T, Nguyen P, Chung T, Liu man amnion/chorion membrane allografts in
Rapid healing of chronic leg ulcers during peri-
Downloaded by Gothenburg University Library from online.liebertpub.com at 02/12/18. For personal use only.

WF. Modulation of macrophage phenotype by patients with chronic diabetic foot ulcers: a long-
lesional injections of granulocyte-macrophage
cell shape. Proc Natl Acad Sci U S A 2013;110: term follow-up study. Wound Med 2014;4:1–4.
colony-stimulating factor therapy in a patient
17253–17258.
126. Forbes J, Fetterolf DE. Dehydrated amniotic with cutaneous polyarteritis nodosa. J Eur Acad
112. Clement AL, Moutinho TJ, Jr, Pins GD. Micro- membrane allografts for the treatment of chronic Dermatol Venereol 2006;20:1341–1343.
patterned dermal-epidermal regeneration matrices wounds: a case series. J Wound Care 2012;21:
139. Siddiqui FH, Mokhashi MH, Boathman A.
create functional niches that enhance epidermal 290, 292, 294–296.
Recombinant granulocyte-macrophage colony-
morphogenesis. Acta Biomater 2013;9:9474–9484.
127. Sheikh E, Sheikh E, Fetterolf D. Use of dehy- stimulating factor in the treatment of indolent
113. Hu MS, Maan ZN, Wu J-C, et al. Tissue engi- drated human amniotic membrane allografts to ulcers with Klippel-Trenaunay-Weber syndrome:
neering and regenerative repair in wound heal- promote healing in patients with refractory non a case report. J Pediatr Surg 2007;42:558–560.
ing. Ann Biomed Eng 2014;42:1494–1507. healing wounds. Int Wound J 2013;11:711–717.
140. Huang G, Sun T, Zhang L, et al. Combined
114. Stewart GA, Lowrey JA, Wakelin SJ, et al. Sonic 128. Koob TJ, Rennert R, Zabek N, et al. Biological application of alginate dressing and human
hedgehog signaling modulates activation of and properties of dehydrated human amnion/chorion granulocyte-macrophage colony stimulating fac-
cytokine production by human peripheral CD4+ T composite graft: implications for chronic wound tor promotes healing in refractory chronic skin
cells. J Immunol 2002;169:5451. healing. Int Wound J 2013;10:493–500. ulcers. Exp Ther Med 2014;7:1772–1776.
115. Fitzpatrick LE, Lisovsky A, Sefton MV. The ex- 129. Maan ZN, Rennert RC, Koob TJ, Januszyk M, Li 141. Avitabile S, Odorisio T, Madonna S, et al.
pression of sonic hedgehog in diabetic wounds WW, Gurtner GC. Cell recruitment by amnion Interleukin-22 promotes wound repair in diabe-
following treatment with poly(methacrylic acid- chorion grafts promotes neovascularization. J tes by improving keratinocyte pro-healing func-
co-methyl methacrylate) beads. Biomaterials Surg Res 2015;193:953–962. tions. J Invest Dermatol 2015;135:2862–2870.
2012;33:5297–5307.
130. Gallagher KA, Liu ZJ, Xiao M, et al. Diabetic 142. Carvalho V, Castanheira P, Faria TQ, et al. Bio-
116. Moura LI, Dias AM, Leal EC, Carvalho L, de impairments in NO-mediated endothelial progen- logical activity of heterologous murine
Sousa HC, Carvalho E. Chitosan-based dressings itor cell mobilization and homing are reversed by interleukin-10 and preliminary studies on the use
loaded with neurotensin—an efficient strategy hyperoxia and SDF-1 alpha. J Clin Invest 2007; of a dextrin nanogel as a delivery system. Int J
to improve early diabetic wound healing. Acta 117:1249–1259. Pharm 2010;400:234–242.
Biomater 2014;10:843–857.
131. Mirza RE, Fang MM, Ennis WJ, Koh TJ. Blocking 143. Braat H, Peppelenbosch MP, Hommes DW.
117. Sandri G, Bonferoni MC, Ferrari F, et al. interleukin-1beta induces a healing-associated Interleukin-10-based therapy for inflammatory
Montmorillonite-chitosan-silver sulfadiazine na- wound macrophage phenotype and improves bowel disease. Expert Opin Biol Ther 2003;3:
nocomposites for topical treatment of chronic healing in type 2 diabetes. Diabetes 2013;62: 725–731.
skin lesions: in vitro biocompatibility, antibacte- 2579–2587.
144. Deiters U, Barsig J, Tawil B, Muhlradt PF. The
rial efficacy and gap closure cell motility prop-
132. Yan C, Gao N, Sun H, et al. Targeting imbalance macrophage-activating lipopeptide-2 accelerates
erties. Carbohydr Polym 2014;102:970–977.
between IL-1beta and IL-1 receptor antagonist wound healing in diabetic mice. Exp Dermatol
118. Mori M, Rossi S, Ferrari F, et al. Sponge-like ameliorates delayed epithelium wound healing 2004;13:731–739.
dressings based on the association of chitosan in diabetic mouse corneas. Am J Pathol 2016;
145. Deiters U, Mühlradt PF. Mycoplasmal lipopeptide
and sericin for the treatment of chronic skin ul- 186:1466–1480.
MALP-2 induces the chemoattractant proteins
cers. I. Design of experiments-assisted develop-
133. Ashcroft GS, Jeong MJ, Ashworth JJ, et al. macrophage inflammatory protein 1alpha (MIP-
ment. J Pharm Sci 2016;105:1180–1187.
Tumor necrosis factor-alpha (TNF-alpha) is a 1alpha), monocyte chemoattractant protein 1,
119. Levengood SL, Erickson AE, Chang FC, Zhang M. therapeutic target for impaired cutaneous wound and MIP-2 and promotes leukocyte infiltration in
Chitosan-poly(caprolactone) nanofibers for skin healing. Wound Repair Regen 2012;20:38–49. mice. Infect Immun 1999;67:3390–3398.
repair. J Mater Chem B Mater Biol Med 2017;5:
134. Olekson MA, Faulknor R, Bandekar A, Semp- 146. Spiller KL, Nassiri S, Witherel CE, et al. Se-
1822–1833.
kowski M, Hsia HC, Berthiaume F. SDF-1 lipo- quential delivery of immunomodulatory cytokines
120. Rozman P, Bolta Z. Use of platelet growth fac- somes promote sustained cell proliferation in to facilitate the M1-to-M2 transition of macro-
tors in treating wounds and soft-tissue injuries. mouse diabetic wounds. Wound Repair Regen phages and enhance vascularization of bone
Acta Dermatovenerol Alp Pannonica Adriat 2007; 2015;23:711–723. scaffolds. Biomaterials 2015;37:194–207.
16:156–165.
135. Fang Y, Shen J, Yao M, Beagley KW, Hambly BD, 147. Alvarez MM, Liu JC, Trujillo-de Santiago G, et al.
121. Keane TJ, Swinehart IT, Badylak SF. Methods Bao S. Granulocyte-macrophage colony-stimulating Delivery strategies to control inflammatory re-
of tissue decellularization used for preparation of factor enhances wound healing in diabetes via sponse: modulating M1–M2 polarization in tis-
22 LAROUCHE ET AL.

sue engineering applications. J Control Release microRNA into the encapsulated cells. Chem 175. Cash JL, Bass MD, Campbell J, Barnes M, Kubes
2016;240:349–363. Commun (Camb) 2014;50:3722–3724. P, Martin P. Resolution mediator Chemerin15
reprograms the wound microenvironment to
148. Ishida Y, Kondo T, Takayasu T, Iwakura Y, 162. Tao SC, Guo SC, Li M, Ke QF, Guo YP, Zhang CQ.
promote repair and reduce scarring. Curr Biol
Mukaida N. The essential involvement of cross- Chitosan wound dressings incorporating exosomes
2014;24:1406–1414.
talk between IFN-gamma and TGF-beta in the derived from microRNA-126-overexpressing syno-
skin wound-healing process. J Immunol 2004; vium mesenchymal stem cells provide sustained 176. Soares de Souza K, Cantaurti TA, Azevedo GM,
172:1848–1855. release of exosomes and heal full-thickness skin et al. Improved cutaneous wound healing after
defects in a diabetic rat model. Stem Cells Transl intraperitoneal injection of alpha-melanocyte-
149. Yager DR, Chen SM, Ward SI, Olutoye OO,
Med 2016;6:736–747. stimulating hormone. Exp Dermatol 2015;24:
Diegelmann RF, Kelman Cohen I. Ability of
198–203.
chronic wound fluids to degrade peptide growth 163. Rabbani PS, Zhou A, Borab ZM, et al. Novel
factors is associated with increased levels of lipoproteoplex delivers Keap1 siRNA based gene 177. Taylor AW, Lee DJ. The alpha-melanocyte
elastase activity and diminished levels of pro- therapy to accelerate diabetic wound healing. stimulating hormone induces conversion of ef-
teinase inhibitors. Wound Repair Regen 1997;5: Biomaterials 2017;132:1–15. fector T cells into Treg cells. J Transplant 2011;
23–32. 2011:246856.
164. Li N, Luo HC, Ren M, et al. Efficiency and safety
150. Gao M, Nguyen TT, Suckow MA, et al. Accel- of beta-CD-(D3)7 as siRNA carrier for decreasing 178. Zhu X, Sun Y, Mu X, et al. Phospholipase CE defi-
eration of diabetic wound healing using a novel matrix metalloproteinase-9 expression and im- ciency delays the early stage of cutaneous wound
Downloaded by Gothenburg University Library from online.liebertpub.com at 02/12/18. For personal use only.

protease-anti-protease combination therapy. proving wound healing in diabetic rats. ACS Appl healing and attenuates scar formation in mice.
Proc Natl Acad Sci U S A 2015;112:15226– Mater Interfaces 2017;9:17417–17426. Biochem Biophys Res Commun 2017;484:144–151.
15231.
165. Luo H-C, Li N, Yan L, et al. Comparison of the 179. Ding J, Tredget EE. The role of chemokines in
151. Ovington LG. Advances in wound dressings. Clin cellular transport mechanism of cationic, star- fibrotic wound healing. Adv Wound Care 2015;
Dermatol 2007;25:33–38. shaped polymers and liposomes in HaCat cells. 4:673–686.
Int J Nanomedicine 2017;12:1085–1096.
152. Wright JB, Lam K, Buret AG, Olson ME, Burrell RE. 180. Jeong W, Yang CE, Roh TS, Kim JH, Lee JH,
Early healing events in a porcine model of con- 166. van der Pol E, Boing AN, Harrison P, Sturk A, Lee WJ. Scar prevention and enhanced wound
taminated wounds: effects of nanocrystalline silver Nieuwland R. Classification, functions, and clin- healing induced by polydeoxyribonucleotide in a
on matrix metalloproteinases, cell apoptosis, and ical relevance of extracellular vesicles. Pharma- rat incisional wound-healing model. Int J Mol
healing. Wound Repair Regen 2002;10:141–151. col Rev 2012;64:676–705. Sci 2017;18:pii: E1698.
153. Forlee M, Rossington A, Searle R. A prospective, 167. Geiger A, Walker A, Nissen E. Human fibrocyte- 181. Cantaruti TA, Costa RA, Soares de Souza K, Vaz
open, multicentre study to evaluate a new gel- derived exosomes accelerate wound healing in NM, Carvalho CR. Indirect effects of immuno-
ling fibre dressing containing silver in the man- genetically diabetic mice. Biochem Biophys Res logical tolerance to a regular dietary protein
agement of venous leg ulcers. Int Wound J 2014; Commun 2015;467:303–309. reduce cutaneous scar formation. Immunology
11:438–445. 2017;151:314–323.
168. Ti D, Hao H, Tong C, et al. LPS-preconditioned
154. Edwards JV, Howley P, Davis R, Mashchak mesenchymal stromal cells modify macrophage 182. Zheng Z, Zhang X, Dang C, et al. Fibromodulin
A, Goheen SC. Protease inhibition by oleic acid polarization for resolution of chronic inflamma- is essential for fetal-type scarless cutaneous
transfer from chronic wound dressings to albu- tion via exosome-shuttled let-7b. J Transl Med wound healing. Am J Pathol 2016;186:2824–
min. Int J Pharm 2007;340:42–51. 2015;13:308. 2832.
155. Roy S, Sen CK. MiRNA in innate immune re- 169. Rahimnejad M, Derakhshanfar S, Zhong W.
183. Hall BE, Wankhade UD, Konkel JE, et al.
sponses: novel players in wound inflammation. Biomaterials and tissue engineering for scar
Transforming growth factor-b3 (TGF-b3) knock-in
Physiol Genomics 2011;43:557–565. management in wound care. Burns Trauma
ameliorates inflammation due to TGF-b1 defi-
2017;5:4.
156. Madhyastha R, Madhyastha H, Nakajima Y, ciency while promoting glucose tolerance. J Biol
Omura S, Maruyama M. MicroRNA signature in 170. Maggini J, Mirkin G, Bognanni I, et al. Mouse Chem 2013;288:32074–32092.
diabetic wound healing: promotive role of miR- bone marrow-derived mesenchymal stromal cells
184. Yoshii A, Kitahara S, Ueta H, Matsuno K, Ezaki
21 in fibroblast migration. Int Wound J 2012;9: turn activated macrophages into a regulatory-like
T. Role of uterine contraction in regeneration of
355–361. profile. PLoS One 2010;5.
the murine postpartum endometrium. Biol Re-
157. Jin Y, Tymen SD, Chen D, et al. MicroRNA-99 171. Jackson WM, Nestl LJ, Tuan RS. Mesenchymal prod 2014;91:32.
family targets AKT/mTOR signaling pathway in stem cell therapy for attenuation of scar for-
mation during wound healing. Stem Cell Res 185. Tziotzios C, Profyris C, Sterling J. Cutaneous
dermal wound healing. PLoS One 2013;8:e64434. scarring: pathophysiology, molecular mecha-
Ther 2012;3:20–28.
158. Li D, Wang A, Liu X, et al. MicroRNA-132 en- nisms, and scar reduction therapeutics. J Am
hances transition from inflammation to prolifer- 172. Bertozzi N, Simonacci F, Grieco MP, Grignaffini E, Acad Dermatol 2012;66:13–24.
Raposio E. The biological and clinical basis for
ation during wound healing. J Clin Invest 2015;
the use of adipose-derived stem cells in the field 186. Poon R, Hong H, Wei X, Pan J, Alman BA. A
125:3008–3026.
of wound healing. Ann Med Surg 2017;20:41–48. high throughput screen identifies Nefopam as
159. Abu-Eid R, Samara RN, Ozbun L, et al. Selective targeting cell proliferation in b-catenin driven
173. De Buys Roessingh AS, Hohlfeld J, Scaletta C,
inhibition of regulatory T cells by targeting the neoplastic and reactive fibroproliferative disor-
et al. Development, characterization, and use
PI3K-Akt pathway. Cancer Immunol Res 2014;2: ders. PLoS One 2012;7:e37940.
of a fetal skin cell bank for tissue engineering
1080–1089.
in wound healing. Cell Transplant 2006;15: 187. Ishida Y, Gao JL, Murphy PM. Chemokine receptor
160. Xie S, Chen M, Yan B, He X, Chen X, Li D. 823–834. CXC3CR1 mediates skin wound healing by pro-
Identification of a role for the PI3K/AKT/mTOR moting macrophage and fibroblast accumulation
174. Sridharan R, Cameron AR, Kelly DJ, Kearney CJ,
signaling pathway in innate immune cells. PLoS and function. J Immunol 2008;180:569–579.
O’Brien FJ. Biomaterial based modulation of
One 2014;9:e94496.
macrophage polarization: a review and sug- 188. Huen AC, Wells A. The beginning of the end:
161. Li J, Kooger R, He M, Xiao X, Zheng L, Zhang Y. gested design principles. Mater Today 2015;18: CXCR3 signaling in late stage wound healing.
A supramolecular hydrogel as a carrier to deliver 313–325. Adv Wound Care 2012;1:244–248.
IMMUNE REGULATION OF SKIN WOUND HEALING 23

189. Martino MM, Briquez PS, Güç E, et al. Growth


EV ¼ extracellular vesicle NET ¼ neutrophil extracellular trap
factors engineered for super-affinity to the ex-
FBGC ¼ foreign body giant cell NT ¼ normotrophic
tracellular matrix enhancing tissue healing. Sci-
FGF ¼ fibroblast growth factor PDGF ¼ platelet-derived growth factor
ence 2014;343:885–888.
GM-CSF ¼ granulocyte macrophage PDRN ¼ polydeoxyribonucleotide
190. Kamaly N, Yameen B, Wu J, Farokhzad OC. colony-stimulating factor PGE2 ¼ prostaglandin E2
Degradable controlled-release polymers and HA ¼ hyaluronic acid PLCe ¼ phospholipase Ce
polymeric nanoparticles: mechanisms of con- HMGB ¼ high-motility group box PRP ¼ platelet-rich plasma
trolling drug release. Chem Rev 2016;116: HSC ¼ hematopoietic stem cell ROS ¼ reactive oxygen species
2602–2663. HT ¼ hypertrophic SDF ¼ stromal cell-derived factor
IFN ¼ interferon Shh ¼ sonic hedgehog signaling
IGF ¼ insulin-like growth factor siRNA ¼ small interfering RNA
Abbreviations and Acronyms IL ¼ interleukin STAT3 ¼ signal transducer and activator
aMSH ¼ alpha-melanocyte-stimulating hormone IL-1Ra ¼ IL-1 receptor agonist of transcription 3
ASC ¼ adipose-derived stem cell LC ¼ Langerhans cells TCR ¼ T cell receptor
C15 ¼ Chemerin 15 M6P ¼ mannose-6-phosphate TGF ¼ transforming growth factor
CXCL ¼ C-X-C chemokine ligand MALP ¼ macrophage-activating lipopeptide TH1 ¼ type 1 T helper
CXCR2 ¼ C-X-C motif chemokine receptor 2 MBV ¼ matrix-bound nanovesicle TIMP ¼ tissue inhibitor of matrix
Downloaded by Gothenburg University Library from online.liebertpub.com at 02/12/18. For personal use only.

DDHAM ¼ dehydrated human amniotic membrane MCP ¼ macrophage chemoattractant protein metalloproteinase
DFU ¼ diabetic foot ulcer MIP ¼ macrophage inflammatory protein TNF ¼ tumor necrosis factor
DSCG ¼ disodium cromoglicate MMP ¼ matrix metalloproteinase Treg ¼ regulatory T cells
ECM ¼ extracellular matrix MSC ¼ mesenchymal stem cell VEGF ¼ vascular endothelial growth factor

You might also like