You are on page 1of 10

ARTICLE IN PRESS

G Model
IMBIO-50677; No. of Pages 10

Immunobiology xxx (2011) xxx–xxx

Contents lists available at ScienceDirect

Immunobiology
journal homepage: www.elsevier.de/imbio

Review

Macrophages in skin injury and repair


Babak Mahdavian Delavary a,b,∗ , Willem M. van der Veer a,b ,
Marjolein van Egmond a,c , Frank B. Niessen b , Robert H.J. Beelen a
a
Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
b
Department of Plastic and Reconstructive Surgery, VU University Medical Center, Amsterdam, The Netherlands
c
Department of Surgery, VU University Medical Center, Amsterdam, The Netherlands

a r t i c l e i n f o a b s t r a c t

Article history: After recruitment to the wound bed, monocytes differentiate into macrophages. Macrophages play a
Received 14 December 2010 central role in all stages of wound healing and orchestrate the wound healing process. Their func-
Accepted 4 January 2011 tional phenotype is dependent on the wound microenvironment, which changes during healing, hereby
altering macrophage phenotype. During the early and short inflammatory phase macrophages exert
Keywords: pro-inflammatory functions like antigen-presenting, phagocytosis and the production of inflammatory
Inflammation
cytokines and growth factors that facilitate the wound healing process. As such, the phenotype of wound
Macrophages
macrophages in this phase is probably the classically activated or the so-called M1 phenotype. During
Phenotype
Remodelling
the proliferative phase, macrophages stimulate proliferation of connective, endothelial and epithelial
Skin tissue directly and indirectly. Especially fibroblasts, keratinocytes and endothelial cells are stimulated by
Wound healing macrophages during this phase to induce and complete ECM formation, reepithelialization and neovascu-
larization. Subsequently, macrophages can change the composition of the ECM both during angiogenesis
and in the remodelling phase by release of degrading enzymes and by synthesizing ECM molecules. This
suggests an important role for alternatively activated macrophages in this phase of wound healing. Patho-
logical functioning of macrophages in the wound healing process can result in derailed wound healing,
like the formation of ulcers, chronic wounds, hypertrophic scars and keloids. However, the exact role
of macrophages in these processes is still incompletely understood. For treating wound repair disorders
more should be elucidated on the role of macrophages in these conditions, especially their functional
phenotype, to find more therapeutic opportunities.
This review summarizes macrophage function in skin injury repair, thereby providing more insight in
macrophage function in wound healing and possible interventions in this process.
© 2011 Elsevier GmbH. All rights reserved.

Contents

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Phase I: Hemostasis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Phase II: Inflammation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Phase III: Proliferation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Phase IIIa: Angiogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Phase IIIb: Reepithelialization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Phase IV: Remodelling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Conclusion and perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

Introduction

∗ Corresponding author at: VU University Medical Center (VUmc), Department


The skin is our largest organ and its primary function is to
of Plastic, Reconstructive and Hand Surgery, Department of Molecular Cell Biology
and Immunology, Postbus 7057, Van der Boechorststraat 7, 1081 BT, Amsterdam,
serve as a protective barrier against infection and excessive water
The Netherlands. Tel.: +31 20 444 8055/20 4448083; fax: +31 20 444 8081. loss. It is also one of the most easily injured human organs. After
E-mail address: b.mahdavian@vumc.nl (B.M. Delavary). injury the skin needs to be repaired to maintain its function. The

0171-2985/$ – see front matter © 2011 Elsevier GmbH. All rights reserved.
doi:10.1016/j.imbio.2011.01.001

Please cite this article in press as: Mahdavian Delavary, B., et al., Macrophages in skin injury and repair. Immunobiology (2011),
doi:10.1016/j.imbio.2011.01.001
ARTICLE IN PRESS
G Model
IMBIO-50677; No. of Pages 10

2 B.M. Delavary et al. / Immunobiology xxx (2011) xxx–xxx

healing process of the injured site, which normally results in the through the vessel wall, they release enzymes that fragment ECM
formation of a scar, is an extremely complex process involving proteins, which creates space for monocytes to migrate into the
numerous cell types as well as growth factors, cytokines, and extra- wound bed. Subsequently, in reaction to the micro-environment,
cellular matrix (ECM) components (Breitkreutz et al. 2009). Wound monocytes differentiate into macrophages. Macrophage numbers
healing consists of dynamic processes including inflammation, increase during the phase of inflammation, peak during the phase
granulation tissue formation (proliferation), reepithelialization and of granulation tissue formation and decline during the maturation
remodelling. None of these phases corresponds to a precisely phase (Martin and Leibovich 2005).
defined period of time and all phases overlap (Eming et al. 2007). Macrophages in the wound bed can display different func-
When this complex process proceeds normally, a normotrophic tional phenotypes, which can roughly be divided into two
scar is formed. Derailment of wound healing however can cause groups: M1 (classically activated) and M2 (alternatively activated)
pathological conditions like a hypertrophic scar, keloid or chronic macrophages (Mantovani et al. 2002). These phenotypes are two
wounds and ulcers (Niessen et al. 1999; van der Veer et al. 2009). extremes of a continuum of macrophage function. There are sev-
To understand the etiology of these pathological conditions it is eral mediators that can stimulate macrophages to differentiate
important to understand skin composition and normal wound heal- into M1-macrophages, the most important being bacterial prod-
ing, and the role of the immune system in the wound healing ucts like lipopolysaccharide (LPS) and inflammatory cytokines like
process. interferon (IFN)-␥ (Mosser and Edwards 2008; Gordon 2003). M1
The skin can be divided into an epidermal and a dermal layer. macrophages exhibit antimicrobial properties by release of inflam-
The epidermis mainly contains keratinocytes, whereas the der- matory mediators inducing tumour necrosis factor (TNF)-␣, nitric
mal part consists of two compartments (Gurtner et al. 2008). The oxide (NO) and interleukin (IL)-6. Although this is important in
first is the cellular compartment, which is typically composed of host defence, the spectrum of expressed cytokines is also capa-
fibroblasts. The second is the acellular compartment, which mainly ble of inducing serious collateral tissue damage (Mosser 2003;
contains the extracellular matrix (ECM). The ECM can further be Wilson et al. 2005). On the other hand, macrophages activated by
functionally divided into a fibrillar fraction, which is characterized IL-4 and IL-13 develop into so-called alternatively (M2) activated
by fibrillar collagen bundles, elastic fibers, and microfibrils, and the macrophages (Gordon 2003), which suppress inflammatory reac-
non-fibrillar fraction that consists of glycosaminoglycans and pro- tions and adaptive immune responses. M2 macrophages have been
teoglycans. All these compartments and different components have reported to play an important role in wound healing, angiogenesis
their own specific function and work together to protect the body and the defence against parasitic infections (Gordon 2003). How-
against invading pathogens and maintenance of other functions like ever, despite their beneficial functions, M2 macrophages can also
thermoregulation and protection against water loss. be involved in different diseases, of which the most prominent are
Besides providing a structural barrier, the skin contains sev- allergy, asthma and fibrosis (Duffield 2003). These diseases are the
eral immune cells that can be activated by invading pathogens or result of a T-helper (Th)-2 response, which is predominated by IL-4
skin damage. One of the most important immune cells involved or IL-10 (Mosser and Edwards 2008).
in wound healing is the macrophage, which exhibits different The mentioned macrophage phenotypes will influence the
immunological functions in the skin, including phagocytosis and wound healing process in different ways, depending on the
antigen-presentation. Furthermore, macrophages produce many micro-environment in which they exert their function. As both
cytokines and chemokines that stimulate new capillary growth, macrophage phenotypes are pivotal for the developing wound, it
collagen synthesis and fibrosis (Mirza et al. 2009). This immune cell is likely that the balance between the two phenotypes is impor-
is thought to orchestrate the wound healing process throughout the tant in the different phases of wound healing. In the first, more
different phases (Mirza et al. 2009; Lucas et al. 2010; Leibovich and pro-inflammatory phase of the wound healing process, more M1
Ross 1975; Devalaraja et al. 2000). Leibovich et al. demonstrated macrophages are needed to scavenge debris and to kill possible
that mice with non-functioning macrophages display retarded invading pathogens. On the other hand, in a later phase in which
wound repair (Leibovich and Ross 1975). This retarded wound new tissue formation is more pronounced, the M2 macrophage
repair includes delayed reepithelialization, delayed neovascular- may have a more important role. In derailed healing processes the
ization and aberrant granulation tissue formation (Mirza et al. balance between the two phenotypes in the phases of repair is prob-
2009; Lucas et al. 2010). ably disturbed. As such, the role of macrophages in the different
Macrophages at the site of wound repair consist of two pop- phases of this process will be outlined in detail in this review.
ulations, which both have their origin in the bone marrow. The
first is the ‘resident’ tissue macrophage that is present in tissues
at all times. Under suitable stimuli it is capable of entering the Phase I: Hemostasis
mitotic cycle. Normal skin contains resident macrophages at a
low density of approximately 1–2 per mm2 (Dipietro et al. 1995). Skin injury causes cell damage and injury of blood vessels. To
MacDonald et al. have demonstrated a minor role for resident tis- prevent blood loss, blood vessels constrict within seconds after
sue macrophages in several processes including wound healing wounding, platelets aggregate and clotting and complement cas-
(Macdonald et al. 2010). They used a macrophage dependant mouse cades are activated. Together, these events are responsible for
wound model in which resident tissue macrophages were depleted the formation of a haemostatic blood clot, mainly composed of
using anti-CSF1R. In this model cellular reconstitution occurred cross-linked fibrin, fibronectin, vitronectin, trombospondin, ery-
within 7 days and neither the timing of the wound healing process throcytes and platelets (Midwood et al. 2004). The clot is a dynamic
nor the efficiency of tissue regeneration was affected by anti-CSF1R matrix of proteins and cells that contribute to hemostasis, serve
treatment (Macdonald et al. 2010). as a temporarily protective shield for the wound, act as a net-
The other major population is newly recruited from haematoge- work for incoming inflammatory cells and provide a reservoir of
nous precursor cells, called monocytes, which themselves are cytokines/growth factors. Platelets are one of the earliest sources
derived from a rapidly dividing pool of cells in the bone mar- of cytokines which mediate macrophage activation and chemo-
row, the promonocytes. The majority of the monocytes circulate taxis. Once trapped in the fibrin net, platelets release granules
in the bloodstream and the rest is stored in the spleen ready to that function as a reservoir for biologically active proteins, such
be deployed to injured or reactive tissue (Jia and Pamer 2009; as ‘Regulated on Activation Normal T cell Expressed and Secreted’
Swirski et al. 2009). Once newly recruited monocytes migrate (RANTES or CCL5) (Frank et al. 2000), thrombin, transforming

Please cite this article in press as: Mahdavian Delavary, B., et al., Macrophages in skin injury and repair. Immunobiology (2011),
doi:10.1016/j.imbio.2011.01.001
ARTICLE IN PRESS
G Model
IMBIO-50677; No. of Pages 10

B.M. Delavary et al. / Immunobiology xxx (2011) xxx–xxx 3

Fig. 1. Inflammatory phase: After migration, monocytes differentiate into macrophages. These macrophages probably exhibit a pro-inflammatory phenotype during this
phase and phagocytose debris and dead neutrophils.

growth factor (TGF)-␤, platelet derived growth factor (PDGF) and Phase II: Inflammation
vascular endothelial growth factor (VEGF). These early released
proteins act as chemoattractants for several inflammatory cells, Hemostasis and the release of chemoattractants that attract
including monocytes (Maraganore 1993). CCL5 is one of the most phagocytic immune cells can be marked as the initiation of the
important monocyte chemoattractants released by platelets after inflammatory phase (Fig. 1). Recruitment of immune cells is facil-
injury. Other important cytokines and chemokines that attract itated by chemokines, vasodilation and increase in blood vessel
monocytes to the wound bed are MCP-1 (CCL2), MIP-1␣ (CCL3), permeability. Initially blood vessels constrict to ensure hemostasis.
TGF-␣, fibronectin, elastin, C5a, C3a, nerve growth factor and ECM- In a later stage these vessels dilate, which peaks around 20 minutes
components as well. after wounding. The major factor responsible for vasodilation is
An important and early mediator of clotting released by platelets histamine. Histamine also causes blood vessel walls to become
is thrombin (He et al. 2010). Thrombin is a serine protease that porous, which subsequently leads to additional extravasation of
plays a central role in hemostasis after tissue injury by convert- inflammatory cells into the wound area. The initial and most
ing soluble plasma fibrinogen into an insoluble fibrin clot and by obvious function of inflammatory cells at the site of injury is to
promoting platelet aggregation (He et al. 2010). Thrombin not provide specific and non-specific defence against pathogens. The
only mediates clot formation, it also plays a role in inflamma- first cells that infiltrate the wound are polymorphonuclear cells,
tion. The pro-inflammatory effects of thrombin include stimulation or neutrophils, which remove foreign particles and bacteria from
of vasodilation responsible for plasma extravasation, edema, and the wound area. In the skin, neutrophils appear in the wound
an increased expression of endothelial adhesion molecules that bed within hours after injury. After exerting their function, neu-
cause monocyte adhesion and infiltration. Thrombin also stimu- trophils are extruded with the eschar (the crust containing dead
lates release of pro-inflammatory cytokines, like CCL2, IL-6 and IL-8 cells and degradative products of the wound) or phagocytosed by
by endothelial cells, which induce monocyte chemotaxis (Marin macrophages. It has been reported that depletion of neutrophils
et al. 2001). Thrombin furthermore induces release of inflammatory in mice or guinea pigs does not result in a decreased number of
cytokines by peripheral blood monocytes, including IL-6, IFN-␥, wound macrophages in normal wound healing (Dovi et al. 2003;
IL-1␤ and TNF-␣. These early produced cytokines are typically pro- Simpson and Ross 1972; Devalaraja et al. 2000). Thus recruit-
inflammatory cytokines, which likely result in differentiation of ment of monocytes to the wound in normal wound healing can
monocytes into M1 macrophages. also proceed in a normal manner when neutrophils are depleted.

Please cite this article in press as: Mahdavian Delavary, B., et al., Macrophages in skin injury and repair. Immunobiology (2011),
doi:10.1016/j.imbio.2011.01.001
ARTICLE IN PRESS
G Model
IMBIO-50677; No. of Pages 10

4 B.M. Delavary et al. / Immunobiology xxx (2011) xxx–xxx

Additionally, the role of neutrophils in normal wound healing out- Phase III: Proliferation
come is inconclusive in our opinion. Some studies suggest a positive
effect of neutrophils in wound healing and some studies suggest The proliferative phase of cutaneous wound healing, also called
a negative effect. Dovi et al. reported that neutrophil depletion granulation phase, is characterized by active fibroplasia, epider-
in the mouse results in accelerated reepithelialization and has mal regeneration, wound contraction and angiogenic sprouting
no effect on the wound disruption strength or collagen content (Fig. 2). It has been reported that upon phagocytosis of apoptotic
(Dovi et al. 2003). On the other hand, Devalaraja et al. reported cells M1 macrophages revert to that of M2 macrophages (Duffield
a delayed reepithelialization and decreased neovascularization in 2003; Gordon 2003). It has furthermore been suggested that M2
CXCR2 knockout mice (Devalaraja et al. 2000). Neutrophils are nev- macrophages derived from M1 macrophages contribute to the res-
ertheless a major source of several pro-inflammatory cytokines, olution of inflammation and the wound healing process (Goerdt
such as IL-1␣, IL-1␤, IL-6 and TNF-␣, which can stimulate newly and Orfanos 1999; Porcheray et al. 2005). Additionally, Lucas et al.
attracted monocytes to differentiate into M1 macrophages (Hubner recently demonstrated that depletion of macrophages during the
et al. 1996; Werner and Grose 2003). proliferative phase significantly disturbed the transition of the mid
Monocytes migrate through the vessel wall in response to stage to the late stage of repair response (Lucas et al. 2010). It can
chemotactic stimuli. Transendothelial monocyte migration starts be concluded from these findings that macrophages have a great
with the initial contact of monocytes with the vascular endothe- role in the resolution of the inflammatory phase and consequently
lium, mediated by adhesion molecules of the selectin family, their the transition of the inflammatory phase to the proliferative
carbohydrate ligands, or by ␣4-integrins (Imhof and Urrand-Lions phase.
2004). This contact results in a weak binding of monocytes to Within a few days of wound debridement by inflammatory
the endothelium and initiates the adhesion cascade. Subsequently, cells, the injured dermis starts to gain volume by the formation
selectins, which are cell-surface proteins, interact with glycopro- of granulation tissue. As recently reported by Lucas et al. the dom-
tein ligands on monocytes allowing them to bind to endothelial inating macrophage population in the wound at day 5 is the M2
cells (Imhof and Urrand-Lions 2004). The rolling monocyte is then macrophage population (Lucas et al. 2010). In the normal wound
stimulated by cytokines and chemokines to engage its surface the M2 macrophage probably dominates the wound very early
integrins that interact with other receptors, like inter-cellular adhe- in the healing process and the inflammatory phase is very short
sion molecule (ICAM) and vascular cell adhesion molecule (VCAM), due to absence of bacterial product or excess dead tissue material.
expressed by endothelial cells. This results in a firm adhesion to Furthermore, it has been reported that M2 macrophages suppress
the endothelial wall, followed by transmigration into the wound inflammatory responses and adaptive Th1-immunity, but scavenge
site. debris, and promote angiogenesis, tissue remodelling and repair.
Monocytes migrate along defined chemotactic gradients and Additionally, they promote a Th2 response, which has been linked
accumulate within avascular hypoxic areas of the wound bed. to fibrinogenesis (Wynn 2008). M2 macrophages are furthermore
With their integrin receptors and selectines, they bind to spe- a prominent source of TGF-␤, which promotes many aspects of
cific proteins, like CAMs of the extracellular matrix (Brown 1995). wound repair including inflammation, chemotaxis, wound contrac-
After migration to the ECM, monocytes are stimulated by several tion, angiogenesis and ECM deposition. TGF-␤ not only stimulates
cytokines like IL-4, IL-10, IFN-y, IL-13, bacterial product like LPS and chemotaxis of and cytokine production by macrophages, it is one
ECM-components to differentiate into macrophages, which peaks of the most important cytokines that influence fibroblast func-
approximately 42 hours after wounding. Monocytes immigrating tion, chemotaxis and ECM deposition. This multipotent protein
into the wound usually develop an inflammatory or debriding phe- promotes fibrosis by directly stimulating mesenchymal cells and
notype, depending on the signals generated in the surrounding fibroblasts (Martin and Leibovich 2005). TGF-␤ not only stimulates
tissue (Stout et al. 2009). It has recently been described by Daley collagen production, but also reduces degradation of the wound
and co-workers that early (day 1) infiltrating macrophages in the matrix by collagenase and through increased inhibition of MMPs
wound mainly (85%) display a M1 phenotype in a sponge mouse via increased expression of TIMPs. TGF-␤ has three isotypes (TGF-
wound model (Daley et al. 2010). ␤1, -␤2 and -␤3), which all stimulate infiltration of inflammatory
Within three to five days after injury, macrophages are the most cells and fibroblasts. However, at gestational ages associated with
prominent cells in the healing tissue. Lucas et al. demonstrated that scarless repair, low levels of TGF-␤1 and high levels of TGF-␤3 are
depletion of macrophages during the inflammatory phase resulted expressed. Furthermore, in fetal wound experiments, a high TGF-
in significant delay of wound repair in a mouse model (Lucas et al. ␤3/TGF-␤1 ratio was associated with scarless healing (Bullard et al.
2010). Macrophages are firstly important in clearance of senescent 2003), suggesting that the relative proportion of each subtype may
cells and debris within the wound. In case of pathogen spreading be crucial for scarring.
in the wound bed, macrophages phagocytose these pathogens and TGF-␤ signals through transmembrane receptors that trigger
present antigens to T-cells. Macrophage involved in clearance of intracellular regulatory proteins known as Smads. A key cyto-
cells or dead tissue undergo apoptosis. Macrophages that survive plasmic mediator of TGF-␤ signalling is Smad3. Recent studies
and do not undergo apoptosis, remain in the wound bed area and suggest that attenuation of the Smad3 signalling improves the
exert other functions that influence the wound healing process, like healing of wounds with an inhibition of fibrosis. Additionally,
stimulation of collagen production, angiogenesis and reepithelial- wounds in Smad3-null mice exposed to irradiation show decreased
ization (Baum and Arpey 2005). This change in functional activity wound width, enhanced epithelialization and reduced numbers of
appears to be initiated predominantly by phagocytosis of apop- myofibroblasts compared to wounds of irradiated wild-type mice
totic cells (Stout 2010). Moreover, it has recently been reported by (Ashcroft et al. 1999). These findings suggest an important role for
Khallou-Laschet et al. that macrophages can phenotypically con- TGF-␤ and Smad3 in wound healing and scar formation.
vert from a M1 to a M2 phenotype (Khallou-Laschet et al. 2010). The involved transcription factors regulate transcription of tar-
They clearly demonstrated a phenotype switch of bone-marrow get genes, including procollagen I and III (Roberts et al. 2003).
derived macrophages in an atherosclerosis model in ApoE KO mice Hypertrophic scar formation is mainly associated with overexpres-
due to environmental changes in cytokine expression. As such, sion of TGF-␤1 and -␤2 (Ishida et al. 2008; Shah et al. 1995; Wang
macrophages play a pivotal role in the transition of the inflamma- et al. 2000). Counteracting the effect of TGF-␤1 and -␤2 significantly
tory phase to the proliferative phase in which they coordinate and reduced scarring in several animal wound models (Choi et al. 1996;
sustain the wound healing events (Singer and Clark 1999). Huang et al. 2002; Shah et al. 1995).

Please cite this article in press as: Mahdavian Delavary, B., et al., Macrophages in skin injury and repair. Immunobiology (2011),
doi:10.1016/j.imbio.2011.01.001
ARTICLE IN PRESS
G Model
IMBIO-50677; No. of Pages 10

B.M. Delavary et al. / Immunobiology xxx (2011) xxx–xxx 5

Fig. 2. The proliferative phase (including reepithelialization): During this phase active fibroplasia, neovascularization and reepithelization occurs. Dependent on the macrophage
phenotype more or less ECM is produced via fibroblasts and myofibroblasts.

The role of fibroblasts in wound healing has been extensively and keratinocyte proliferation (Werner and Grose 2003). Ablation
studied over the past years. Fibroblasts and myofibroblasts mainly of macrophages in the wound consequently results in decreased
produce the new ECM necessary for supporting cells and blood expression of TGF-␤, reduced proliferation of fibroblasts and less
vessels, which provides nutrients and oxygen needed for cell ECM deposition (Mirza et al. 2009).
growth and proliferation. Some fibroblasts are already present Eventually, during the proliferative phase, the fibrin clot is trans-
in the wound area, scattered within the tissue without forming formed into connective tissue rich in blood vessels, underlying the
cell–cell contacts, while others are derived from nearby undiffer- pink granular appearance. This transformation requires an accurate
entiated mesenchymal cells (Ross et al. 1970). Both TGF-␤ and balance between matrix degradation and production.
PDGF can mediate the transition of these mesenchymal cells into
myofibroblasts (Kalluri and Neilson 2003; Werner and Grose 2003).
Furthermore, both cytokines stimulate these (myo)fibroblasts to Phase IIIa: Angiogenesis
produce collagen and other ECM components.
Myofibroblasts are characterized by ␣-smooth muscle actin The formation of new capillaries from pre-existing blood ves-
(␣-SMA) expression and are involved in wound contraction sels during wound healing mainly presents as angiogenic sprouting
(Martinez-Ferrer et al. 2010). Compared to normal dermal fibrob- and is an essential component of wound healing because of the
lasts, myofibroblasts produce higher amounts of ECM components increased nutrient demand (Fig. 3). This process proceeds through
(Huet et al. 2008). Collagen production by fibroblasts is initiated a series of steps. Macrophages play an important role in this pro-
between 3 and 5 days after injury and is stimulated by TGF-␤, PDGF, cess, which has been illustrated in for instance the cornea, the rabbit
Fibroblast Growth Factor 2 (FGF2) and Insulin Like Growth Factor ear (Thakral et al. 1979), and in vitro (Greenburg and Hunt 1978).
1 (IGF-1) (Ishida et al. 2008; Vogler et al. 2003; Werner and Grose Macrophage depletion leads to delayed and abnormal neovascular-
2003), which are mainly produced by macrophages. ization.
Simultaneously, fibroblasts and endothelial cells start to migrate Firstly, the ECM and basement membrane are degraded to facil-
over the provisional wound matrix into the wound space with the itate migrating endothelial cells. Degradation is mediated by MMPs
help of matrix metalloproteinases (MMPs). The migrated endothe- produced by several cells including macrophages, which are a
lial cells provide the formation of new vessels in the wound bed. rich source for MMPs and serine protease. Besides macrophages,
An important cytokine that promotes this migration is IL-1, which endothelial cells themselves also produce a number of proteases
stimulates the release of MMPs and synergistically induces collage- by which they locally degrade the basement membrane. Subse-
nase activity in conjunction with IFN-␥ and TNF-␣. Increased levels quently, endothelial cells migrate into the wound by adherence to
of IL-1 may consequently result in ECM degradation and slower integrin cell surface receptors on the ECM. Migration is stimulated
wound healing (Niessen et al. 2001). by among others VEGF, FGF, Angiopoietins and TGF-␤. The expres-
Macrophages provide an ongoing source of pro-inflammatory sion of integrin receptors on endothelial cells is also stimulated
cytokines, including IL-1␣, IL-1␤, IL-6 and TNF-␣, which are not by VEGF, FGF and TGF-␤. TGF-␤ not only stimulates endothelial
only responsible for the control of inflammatory cell adhesion and migration, differentiation and tubule formation, but also amplifies
migration, but are also important for the stimulation of fibroblast the latter mentioned processes.

Please cite this article in press as: Mahdavian Delavary, B., et al., Macrophages in skin injury and repair. Immunobiology (2011),
doi:10.1016/j.imbio.2011.01.001
ARTICLE IN PRESS
G Model
IMBIO-50677; No. of Pages 10

6 B.M. Delavary et al. / Immunobiology xxx (2011) xxx–xxx

Fig. 3. Angiogenesis: This is part of the proliferative phase. angiogenesis is predominantly regulated by macrophages, endothelial cells and keratinocytes. The most important
mediator during this phase is VEGF.

Simultaneously, these secretory proteins regulate capillary dominantly stimulated by M2 macrophages, especially through the
branching and neovascularization during ischemia reperfusion. formation of VEGF (Tammela et al. 2008). Previous studies mainly
Among them is plasminogen activator (PA), which can degrade ECM focussed on understanding how tip cells initiate vessel sprouting
molecules resulting in pro-angiogenic fragments like hyaluronic while very little is known about how these cells fuse with neigh-
acid and fibrin (van Hinsbergh et al. 1997). Inhibition of PA conse- bouring sprouts to form a perfused vessel (Schmidt and Carmeliet
quently leads to suppression of angiogenesis. 2010). Recently it has been reported by Fantin et al. that con-
Once recruited to the wound, macrophages induce angiogene- trary to expectation, tip cells lack the ability to recognize, find and
sis. This is predominantly exerted by releasing TNF-␣ and VEGF. fuse with other tip cells. Instead, macrophages serve as guidance
TNF-␣ may in turn induce VEGF expression by keratinocytes and bridge cells in this fusion process (Fantin et al. 2010). These
and fibroblasts (Frank et al. 1995). Depletion of macrophages in findings further support an important role for macrophages in
the wound results in reduced vascularization (Mirza et al. 2009) angiogenesis.
and also leads to severe hemorrhage, fibrin and serum exudates VEGF is one of the most important pro-angiogenic mediators
in macrophage depleted granulation tissue (Lucas et al. 2010). and stimulates multiple components of the angiogenic cascade. It
This has been reported to be mainly caused by withdrawal of is present in wound fluid in high concentrations (Howdieshell et al.
macrophage-derived TGF-␤1 and VEGF (Mirza et al. 2009; Lucas 1998), and it is secreted by different cell types in response to ker-
et al. 2010), supporting an important role for macrophages in pro- atinocyte growth factor (KGF), TGF-␤ (Ferrara et al. 1995; Ferrara
moting angiogenesis via their secretory products. Furthermore, 1995), hypoxia (Shweiki et al. 1992), bFGF (Stavri et al. 1995) and
there is an important relation between macrophages, monocytes PDGF (Frank et al. 1995). Platelets are one of the first cells to
and endothelial cells which originates from embryogenesis. Both appear in the wound site followed by neutrophils and macrophages.
monocytes and endothelial cells have the same precursor cell, the Platelets release VEGF after stimulation, particularly after thrombin
hemangioblast. Monocytes and macrophages have the ability to stimulation (Mohle et al. 1997), leading to stimulation of angiogen-
acquire endothelial properties when placed in angiogenic condi- esis in the early phase of wound healing.
tions. Moreover, cells with a hybrid phenotype, positive for both The endothelium of the microvasculature exhibits several
monocytic marker CD45 and endothelial V-cadherin, were isolated important responses to extracellular VEGF. The first is increased
from tumours (Conejo-Garcia et al. 2005). endothelial proliferation, which leads to a greater population size.
For induction of angiogenesis, macrophages first have to be acti- The second is increased permeability, exerted through an unknown
vated in a specific way. Several studies have reported that M2 change in endothelial cell structure and function (Dvorak et al.
macrophages promote angiogenesis. Under several stimuli, like 1995). This increased permeability results in a provisional extra-
low oxygen tension or high concentrations of lactate pyruvate or cellular matrix necessary for angiogenesis. Macrophages can also
hydrogen ions that have also been observed in the wound area, increase vascular permeability by releasing vaso-active substances
macrophages can become angiogenic (Knighton et al. 1983). such as vascular permeability factor (Berse et al. 1992), sub-
Another very important function has recently been suggested to stance P (Pascual and Bost 1990), platelet activating factor and
be conducted by (resident) tissue macrophages in angiogenesis, as prostaglandins (Middleton and Thatcher 1998). These findings sup-
these cells are located near vessel branches. Specialized endothe- port the ability of the monocyte/macrophage lineage, especially
lial cells at the forefront of the sprout, the so called tip cells, are the M2 macrophage (the major source of TGF-␤), to contribute to
thought to navigate to their target. The formation of tip cells is pre- angiogenesis during wound healing.

Please cite this article in press as: Mahdavian Delavary, B., et al., Macrophages in skin injury and repair. Immunobiology (2011),
doi:10.1016/j.imbio.2011.01.001
ARTICLE IN PRESS
G Model
IMBIO-50677; No. of Pages 10

B.M. Delavary et al. / Immunobiology xxx (2011) xxx–xxx 7

Phase IIIb: Reepithelialization age or when wound healing is complicated by infection or other
activity which activates the adaptive leukocyte system, T-cells play
Reepithelialization includes the re-establishment of an intact an important role (Tredget et al. 2006). Macrophages and T-cells
epidermis over the newly formed (scar) tissue (Fig. 2). In intact cross-talk via cytokines and co-stimulators, including cell surface
skin, the basal keratinocyte layer is attached to the basal lamina. molecules, which can result in a wide range of outcomes in function
After injury, keratinocyte organization is disturbed and has to be of these cells (Doherty 1995). Moreover, macrophages stimulate T-
restored. This restoration process initiates within hours after injury cell expansion and differentiation to Th1 and Th2 cells. In this case,
and is performed by keratinocytes that move across the granula- T-cells make wound healing more complex and may prolong or
tion tissue. This process, called contact guidance, continues until derail the wound healing process in several aspects. This may result
keratinocytes from the opposing sides of the wound re-establish in necrosis, fibrosis, ulcera formation and granuloma formation.
contact. Before migration, keratinocytes change shape, become flat- T-lymphocytes infiltrate the wound bed in the late inflamma-
ter and elongated, and extend cellular processes like lamellipodia tory phase of wound repair, suggesting that these cells are involved
and fillopodia (Santoro and Gaudino 2005). in either the late proliferative or remodelling phase. In skin wounds,
TGF-␤1 is one of the most important ligands for epithelial T-cell numbers peak at day 7, but remain in the scar for up to
cell migration during reepithelialization (O’Kane and Ferguson months after wounding (unpublished data). Studies in mice lack-
1997), as it stimulates expression of integrin subunits that pro- ing T-cells suggest that the primary function of T-cells in incision
mote keratinocyte migration on the provisional ECM. Mirza et al. wounds is downregulation of fibrous tissue growth. This suggests
have recently shown that depletion of macrophages in the wound that more Th1 cells populate the normal wound during the remod-
results in delayed reepithelialization (Mirza et al. 2009), sup- elling phase, because a Th2 response results in the production of
porting that macrophage-derived TGF-␤ plays a crucial role in ECM. Th1 cells mainly produce IFN-y which results in the differen-
reepithelialization. Several other growth factors are important for tiation of macrophages into M1-macrophages.
keratinocyte proliferation at the wound edge, including epidermal Macrophages also play an important role in modulating the
growth factor (EGF), TGF-␣, keratinocyte growth factor (KGF) and capacity of other cell types to secrete neutral proteinases that
heparin-binding epidermal growth factor (HB-EGF), all acting as degrade matrix macromolecules. ECM breakdown is fundamental
ligands for the EGF receptor. In order to create a path through the for wound healing and tissue remodelling. One of the most impor-
fibrin clot, the keratinocytes in the wound edge have to dissolve tant components of ECM is collagen which increases the strength
the fibrin barrier. The major fibrinolytic enzyme in this process of the wound. The important processes in the remodelling phase
is plasmin, which is derived from activated plasminogen out of are the rearrangement and cross-linking of initially deposited col-
the bloodstream. Furthermore, various MMPs play a crucial role lagen fibers and substitution of type III collagen by type I collagen,
in this process. These MMPs are upregulated by keratinocytes and which is a stronger collagen fibril. In vivo, collagen is primarily pro-
macrophages in the wound edge. Degradation of ECM components duced by (myo)fibroblasts. Increasing quantities of ECM signals the
by MMPs is required to remove and reorganize provisional matrices fibroblasts to decrease subsequent collagen production. Further-
and to allow migration (Ravanti and Kahari 2000). more, IFN-␥ and TNF-␣ stimulate fibroblasts to decrease collagen
The reepithelialization process is made easier by contraction of synthesis.
the underlying connective tissue, which brings the wound mar- MMPs and their natural inhibitors (TIMPs) are generally
gins towards each other. This contraction process is performed by accepted as being important mediators of proteolytic activity dur-
myofibroblasts, activated by TGF-␤ and PDGF (Frank et al. 1995). ing many physiological and pathological remodelling processes,
Once the wound surface is covered by a monolayer of keratinocytes, including tissue repair. The MMP family has the combined capacity
epidermal migration ceases and a new stratified epidermis with to degrade virtually all ECM components and basement mem-
underlying basal lamina is re-established from the margins of the brane macromolecules. Macrophages are a rich source of MMPs
wound inwards. At this moment, the defect is filled with gran- and serine proteases. They are especially involved in MMP-2, MMP-
ulation tissue and covered by a newly formed epidermal layer. 12 and MMP-19 expression, and they synthesize tissue inhibitors
Nevertheless, the wound healing process, particularly the remod- of metallo- and serine proteases. MMP-12, for example, is a
elling phase, can still go on for months. The remodelling process is macrophage-specific metallo-elastase (Shapiro et al. 1993), which
crucial for the scar strength and appearance. has been suggested to play a role in capillary regression in skin
wounds, resulting in a decrease of erythema (Madlener et al. 1998).
Furthermore, macrophages have been reported to be able to com-
Phase IV: Remodelling plete the hydrolysis of the matrix proteins into small molecules
(Jones and Scott-Burden 1979). Proteinases, including PA, can
The remodelling phase is the last and longest phase of the also mediate extracellular protein degradation by macrophages
wound healing process and continues for weeks to months (Fig. 4). (Gordon et al. 1974; Jones and Scott-Burden 1979). Besides pro-
During this phase cell proliferation slows down, protein synthesis moting the formation of ECM, macrophages also promote tissue
decreases and remodelling of collagen into larger, more organised integrity by producing type VIII collagen (Weitkamp et al. 1999)
fibrils occurs. As the nutrient demand within the tissue decreases, and some matrix proteins (Gratchev et al. 2001).
a regression of recently formed capillaries occurs, and the red- The remodelling phase is primarily dependent on tissue break-
ness of the scar will fade. Most endothelial cells, macrophages and down and ECM production and macrophages play a crucial role in
myofibroblasts undergo apoptosis, or exit the wound (Gurtner et al. both processes. The breakdown is directly regulated and the ECM
2008). production is mainly regulated via fibroblasts. As recently pub-
The wound remodelling process is a balance between ECM lished, in the last phase of normal wound healing, the number of
production, breakdown and remodelling. This balance is probably wound macrophages is decreased, which will result in less ECM
determined by the microenvironment of the scar by means of a Th2 production. Beside the changed macrophage numbers, different or
or Th1 dominated environment (Tredget et al. 2006) and is proba- reduced activity of macrophages in the last phase of wound healing
bly maintained by the T-cells that infiltrate the wound and signal could also result in less ECM production. These changes, together
macrophages and other immune cells. with contraction of the wound, will result in a decrease in wound
It has been reported that T-cells do not play a prominent role size and volume. In pathological conditions macrophage numbers
in wound healing in case of minor damage. In case of major dam- and function could be changed resulting in aberrant scarring.

Please cite this article in press as: Mahdavian Delavary, B., et al., Macrophages in skin injury and repair. Immunobiology (2011),
doi:10.1016/j.imbio.2011.01.001
ARTICLE IN PRESS
G Model
IMBIO-50677; No. of Pages 10

8 B.M. Delavary et al. / Immunobiology xxx (2011) xxx–xxx

Fig. 4. Remodelling phase: During this phase the balance between ECM-breakdown and -formation is important and determines the eventual scar result. The balance is
determined by among others the micro-environment, macrophage phenotype and T-cell response.

Conclusion and perspectives References

The phases of wound healing normally proceed in a predictable Ashcroft, G.S., Yang, X., Glick, A.B., Weinstein, M., Letterio, J.L., Mizel, D.E., Anzano,
M., Greenwell-Wild, T., Wahl, S.M., Deng, C., Roberts, A.B., 1999. Mice lacking
timely manner. In humans, problems with the wound healing can Smad3 show accelerated wound healing and an impaired local inflammatory
manifest as either delayed or excessive. Delayed wound healing response. Nat. Cell Biol. 1, 260–266.
can result in a chronic wound and excessive wound healing results Baum, C.L., Arpey, C.J., 2005. Normal cutaneous wound healing: clinical correlation
with cellular and molecular events. Dermatol. Surg. 31, 674–686.
in a hypertrophic scar or keloid scar formation. Excessive scar for- Berse, B., Brown, L.F., van de, W.L., Dvorak, H.F., Senger, D.R., 1992. Vascular
mation is characterized by an elevated ECM deposition and scar permeability factor (vascular endothelial growth factor) gene is expressed
vascularization. Excessive scars can cause disfigurement and func- differentially in normal tissues, macrophages, and tumors. Mol. Biol. Cell 3,
211–220.
tional impairment. In burns, for example, joint function can be Breitkreutz, D., Mirancea, N., Nischt, R., 2009. Basement membranes in skin: unique
severely impaired due to the excessive scar. Much research has matrix structures with diverse functions? Histochem. Cell Biol. 132, 1–10.
been conducted, primarily focussing on fibroblasts and TGF-␤, to Brown, E.J., 1995. Phagocytosis. Bioessays 17, 109–117.
Bullard, K.M., Longaker, M.T., Lorenz, H.P., 2003. Fetal wound healing: current biol-
elucidate the wound healing process, and to find novel treatment
ogy. World J. Surg. 27, 54–61.
possibilities. Unfortunately, studies on wound healing have not yet Choi, B.M., Kwak, H.J., Jun, C.D., Park, S.D., Kim, K.Y., Kim, H.R., Chung, H.T., 1996.
led to substantial advances in patient care. Control of scarring in adult wounds using antisense transforming growth factor-
Until now, macrophage phenotype has not been regarded to play beta 1 oligodeoxynucleotides. Immunol. Cell Biol. 74, 144–150.
Conejo-Garcia, J.R., Buckanovich, R.J., Benencia, F., Courreges, M.C., Rubin, S.C.,
a substantial role in the outcome of the wound healing process. Carroll, R.G., Coukos, G., 2005. Vascular leukocytes contribute to tumor vascu-
Since the role of macrophage phenotype has been highlighted in larization. Blood 105, 679–681.
fibrosis in several studies and some studies underline the impor- Daley, J.M., Brancato, S.K., Thomay, A.A., Reichner, J.S., Albina, J.E., 2010. The pheno-
type of murine wound macrophages. J. Leukoc. Biol. 87, 59–67.
tance of macrophages in wound healing, the interest in macrophage Devalaraja, R.M., Nanney, L.B., Du, J., Qian, Q., Yu, Y., Devalaraja, M.N., Richmond, A.,
function in wound repair is rising. 2000. Delayed wound healing in CXCR2 knockout mice. J. Invest. Dermatol. 115,
Based on the mentioned studies in this paper we hypothesize 234–244.
Dipietro, L.A., Polverini, P.J., Rahbe, S.M., Kovacs, E.J., 1995. Modulation of JE/MCP-1
that the inflammatory phase in excessive scarring is prolonged and expression in dermal wound repair. Am. J. Pathol. 146, 868–875.
therefore more macrophages are attracted to the wound bed. The Doherty, T.M., 1995. T-cell regulation of macrophage function. Curr. Opin. Immunol.
phenotype of newly attracted macrophages will first be more pro- 7, 400–404.
Dovi, J.V., He, L.K., Dipietro, L.A., 2003. Accelerated wound closure in neutrophil-
inflammatory (M1) and will switch to a more pro-fibrotic (M2) depleted mice. J. Leukoc. Biol. 73, 448–455.
phenotype. The M2 macrophages could furthermore be more active Duffield, J.S., 2003. The inflammatory macrophage: a story of Jekyll and Hyde. Clin.
in excessive scarring because of more intense stimuli. Sci. (Lond.) 104, 27–38.
Dvorak, H.F., Brown, L.F., Detmar, M., Dvorak, A.M., 1995. Vascular permeability fac-
As macrophages play an important role in wound healing,
tor/vascular endothelial growth factor, microvascular hyperpermeability, and
intervention in macrophage function in several pathological con- angiogenesis. Am. J. Pathol. 146, 1029–1039.
ditions may result in less ulceration and chronic wounding Eming, S.A., Krieg, T., Davidson, J.M., 2007. Inflammation in wound repair: molecular
and may prevent hypertrophic scar and keloid formation. For and cellular mechanisms. J. Invest. Dermatol. 127, 514–525.
Fantin, A., Vieira, J.M., Gestri, G., Denti, L., Schwarz, Q., Prykhozhij, S., Peri, F., Wilson,
intervention, more knowledge on the role of macrophages in S.W., Ruhrberg, C., 2010. Tissue macrophages act as cellular chaperones for vas-
would healing, especially their functional phenotype (M1/M2) in cular anastomosis downstream of VEGF-mediated endothelial tip cell induction.
the different phases and in different pathological conditions is Blood 116, 829–840.
Ferrara, N., 1995. The role of vascular endothelial growth factor in pathological
needed. angiogenesis. Breast Cancer Res. Treat. 36, 127–137.

Please cite this article in press as: Mahdavian Delavary, B., et al., Macrophages in skin injury and repair. Immunobiology (2011),
doi:10.1016/j.imbio.2011.01.001
ARTICLE IN PRESS
G Model
IMBIO-50677; No. of Pages 10

B.M. Delavary et al. / Immunobiology xxx (2011) xxx–xxx 9

Ferrara, N., Heinsohn, H., Walder, C.E., Bunting, S., Thomas, G.R., 1995. The regulation Martin, P., Leibovich, S.J., 2005. Inflammatory cells during wound repair: the good,
of blood vessel growth by vascular endothelial growth factor. Ann. N. Y. Acad. the bad and the ugly. Trends Cell Biol. 15, 599–607.
Sci. 752, 246–256. Martinez-Ferrer, M., Fshar-Sherif, A.R., Uwamariya, C., de, C.B., Davidson, J.M.,
Frank, S., Hubner, G., Breier, G., Longaker, M.T., Greenhalgh, D.G., Werner, S., 1995. Bhowmick, N.A., 2010. Dermal transforming growth factor-beta responsive-
Regulation of vascular endothelial growth factor expression in cultured ker- ness mediates wound contraction and epithelial closure. Am. J. Pathol. 176, 98–
atinocytes. Implications for normal and impaired wound healing. J. Biol. Chem. 107.
270, 12607–12613. Middleton, M., Thatcher, N., 1998. G- and GM-CSF. Int. J. Antimicrob. Agents 10,
Frank, S., Kampfer, H., Wetzler, C., Stallmeyer, B., Pfeilschifter, J., 2000. Large induc- 91–93.
tion of the chemotactic cytokine RANTES during cutaneous wound repair: a Midwood, K.S., Williams, L.V., Schwarzbauer, J.E., 2004. Tissue repair and the dynam-
regulatory role for nitric oxide in keratinocyte-derived RANTES expression. ics of the extracellular matrix. Int. J. Biochem. Cell Biol. 36, 1031–1037.
Biochem. J. 347 (Pt 1), 265–273. Mirza, R., Dipietro, L.A., Koh, T.J., 2009. Selective and specific macrophage ablation
Goerdt, S., Orfanos, C.E., 1999. Other functions, other genes: alternative activation is detrimental to wound healing in mice. Am. J. Pathol. 175, 2454–2462.
of antigen-presenting cells. Immunity 10, 137–142. Mohle, R., Green, D., Moore, M.A., Nachman, R.L., Rafii, S., 1997. Constitutive pro-
Gordon, S., 2003. Alternative activation of macrophages. Nat. Rev. Immunol. 3, duction and thrombin-induced release of vascular endothelial growth factor by
23–35. human megakaryocytes and platelets. Proc. Natl. Acad. Sci. U. S. A. 94, 663–
Gordon, S., Unkeless, J.C., Cohn, Z.A., 1974. Induction of macrophage plasminogen 668.
activator by endotoxin stimulation and phagocytosis: evidence for a two-stage Mosser, D.M., 2003. The many faces of macrophage activation. J. Leukoc. Biol. 73,
process. J. Exp. Med. 140, 995–1010. 209–212.
Gratchev, A., Guillot, P., Hakiy, N., Politz, O., Orfanos, C.E., Schledzewski, K., Goerdt, Mosser, D.M., Edwards, J.P., 2008. Exploring the full spectrum of macrophage acti-
S., 2001. Alternatively activated macrophages differentially express fibronectin vation. Nat. Rev. Immunol. 8, 958–969.
and its splice variants and the extracellular matrix protein betaIG-H3. Scand. J. Niessen, F.B., Andriessen, M.P., Schalkwijk, J., Visser, L., Timens, W., 2001.
Immunol. 53, 386–392. Keratinocyte-derived growth factors play a role in the formation of hypertrophic
Greenburg, G.B., Hunt, T.K., 1978. The proliferative response in vitro of vascular scars. J. Pathol. 194, 207–216.
endothelial and smooth muscle cells exposed to wound fluids and macrophages. Niessen, F.B., Spauwen, P.H., Schalkwijk, J., Kon, M., 1999. On the nature of hyper-
J. Cell Physiol. 97, 353–360. trophic scars and keloids: a review. Plast. Reconstr. Surg. 104, 1435–1458.
Gurtner, G.C., Werner, S., Barrandon, Y., Longaker, M.T., 2008. Wound repair and O’Kane, S., Ferguson, M.W., 1997. Transforming growth factor beta s and wound
regeneration. Nature 453, 314–321. healing. Int. J. Biochem. Cell Biol. 29, 63–78.
He, S., Blomback, M., Bark, N., Johnsson, H., Wallen, N.H., 2010. The direct thrombin Pascual, D.W., Bost, K.L., 1990. Substance P production by P388D1 macrophages: a
inhibitors (argatroban, bivalirudin and lepirudin) and the indirect Xa-inhibitor possible autocrine function for this neuropeptide. Immunology 71, 52–56.
(danaparoid) increase fibrin network porosity and thus facilitate fibrinolysis. Porcheray, F., Viaud, S., Rimaniol, A.C., Leone, C., Samah, B., reuddre-Bosquet, N.,
Thromb. Haemost. 103, 1076–1084. Dormont, D., Gras, G., 2005. Macrophage activation switching: an asset for the
Howdieshell, T.R., Riegner, C., Gupta, V., Callaway, D., Grembowicz, K., Sathya- resolution of inflammation. Clin. Exp. Immunol. 142, 481–489.
narayana, McNeil, P.L., 1998. Normoxic wound fluid contains high levels of Ravanti, L., Kahari, V.M., 2000. Matrix metalloproteinases in wound repair (review).
vascular endothelial growth factor. Ann. Surg. 228, 707–715. Int. J. Mol. Med. 6, 391–407.
Huang, J.S., Wang, Y.H., Ling, T.Y., Chuang, S.S., Johnson, F.E., Huang, S.S., 2002. Syn- Roberts, A.B., Russo, A., Felici, A., Flanders, K.C., 2003. Smad3: a key player in patho-
thetic TGF-beta antagonist accelerates wound healing and reduces scarring. genetic mechanisms dependent on TGF-beta. Ann. N. Y. Acad. Sci. 995, 1–10.
FASEB J. 16, 1269–1270. Ross, R., Everett, N.B., Tyler, R., 1970. Wound healing and collagen formation. VI.
Hubner, G., Brauchle, M., Smola, H., Madlener, M., Fassler, R., Werner, S., 1996. Dif- The origin of the wound fibroblast studied in parabiosis. J. Cell Biol. 44, 645–
ferential regulation of pro-inflammatory cytokines during wound healing in 654.
normal and glucocorticoid-treated mice. Cytokine 8, 548–556. Santoro, M.M., Gaudino, G., 2005. Cellular and molecular facets of keratinocyte reep-
Huet, E., Vallee, B., Szul, D., Verrecchia, F., Mourah, S., Jester, J.V., Hoang-Xuan, ithelization during wound healing. Exp. Cell Res. 304, 274–286.
T., Menashi, S., Gabison, E.E., 2008. Extracellular matrix metalloproteinase Schmidt, T., Carmeliet, P., 2010. Blood-vessel formation: bridges that guide and unite.
inducer/CD147 promotes myofibroblast differentiation by inducing alpha- Nature 465, 697–699.
smooth muscle actin expression and collagen gel contraction: implications in Shah, M., Foreman, D.M., Ferguson, M.W., 1995. Neutralisation of TGF-beta 1 and
tissue remodeling. FASEB J. 22, 1144–1154. TGF-beta 2 or exogenous addition of TGF-beta 3 to cutaneous rat wounds reduces
Imhof, B.A., Urrand-Lions, M., 2004. Adhesion mechanisms regulating the migration scarring. J. Cell Sci. 108 (Pt. 3), 985–1002.
of monocytes. Nat. Rev. Immunol. 4, 432–444. Shapiro, S.D., Kobayashi, D.K., Ley, T.J., 1993. Cloning and characterization of a unique
Ishida, Y., Gao, J.L., Murphy, P.M., 2008. Chemokine receptor CX3CR1 mediates skin elastolytic metalloproteinase produced by human alveolar macrophages. J. Biol.
wound healing by promoting macrophage and fibroblast accumulation and Chem. 268, 23824–23829.
function. J. Immunol. 180, 569–579. Shweiki, D., Itin, A., Soffer, D., Keshet, E., 1992. Vascular endothelial growth factor
Jia, T., Pamer, E.G., 2009. Immunology. Dispensable but not irrelevant. Science 325, induced by hypoxia may mediate hypoxia-initiated angiogenesis. Nature 359,
549–550. 843–845.
Jones, P.A., Scott-Burden, T., 1979. Activated macrophages digest the extracellular Simpson, D.M., Ross, R., 1972. The neutrophilic leukocyte in wound repair a study
matrix proteins produced by cultured cells. Biochem. Biophys. Res. Commun. with antineutrophil serum. J. Clin. Invest. 51, 2009–2023.
86, 71–77. Singer, A.J., Clark, R.A., 1999. Cutaneous wound healing. N. Engl. J. Med. 341, 738–746.
Kalluri, R., Neilson, E.G., 2003. Epithelial-mesenchymal transition and its implica- Stavri, G.T., Zachary, I.C., Baskerville, P.A., Martin, J.F., Erusalimsky, J.D., 1995. Basic
tions for fibrosis. J. Clin. Invest. 112, 1776–1784. fibroblast growth factor upregulates the expression of vascular endothelial
Khallou-Laschet, J., Varthaman, A., Fornasa, G., Compain, C., Gaston, A.T., Clement, growth factor in vascular smooth muscle cells. Synergistic interaction with
M., Dussiot, M., Levillain, O., Graff-Dubois, S., Nicoletti, A., Caligiuri, G., 2010. hypoxia. Circulation 92, 11–14.
Macrophage plasticity in experimental atherosclerosis. PLoS One 5, e8852. Stout, R.D., 2010. Editorial: macrophage functional phenotypes: no alternatives in
Knighton, D.R., Hunt, T.K., Scheuenstuhl, H., Halliday, B.J., Werb, Z., Banda, M.J., 1983. dermal wound healing? J. Leukoc. Biol. 87, 19–21.
Oxygen tension regulates the expression of angiogenesis factor by macrophages. Stout, R.D., Watkins, S.K., Suttles, J., 2009. Functional plasticity of macrophages:
Science 221, 1283–1285. in situ reprogramming of tumor-associated macrophages. J. Leukoc. Biol. 86,
Leibovich, S.J., Ross, R., 1975. The role of the macrophage in wound repair. A study 1105–1109.
with hydrocortisone and antimacrophage serum. Am. J. Pathol. 78, 71–100. Swirski, F.K., Nahrendorf, M., Etzrodt, M., Wildgruber, M., Cortez-Retamozo, V.,
Lucas, T., Waisman, A., Ranjan, R., Roes, J., Krieg, T., Muller, W., Roers, A., Eming, Panizzi, P., Figueiredo, J.L., Kohler, R.H., Chudnovskiy, A., Waterman, P., Aikawa,
S.A., 2010. Differential roles of macrophages in diverse phases of skin repair. J. E., Mempel, T.R., Libby, P., Weissleder, R., Pittet, M.J., 2009. Identification of
Immunol. 184, 3964–3977. splenic reservoir monocytes and their deployment to inflammatory sites. Sci-
Macdonald, K.P., Palmer, J.S., Cronau, S., Seppanen, E., Olver, S., Raffelt, N.C., Kuns, ence 325, 612–616.
R., Pettit, A.R., Clouston, A., Wainwright, B., Branstetter, D., Smith, J., Paxton, Tammela, T., Zarkada, G., Wallgard, E., Murtomaki, A., Suchting, S., Wirzenius,
R.J., Cerretti, D.P., Bonham, L., Hill, G.R., Hume, D.A., 2010. An antibody against M., Waltari, M., Hellstrom, M., Schomber, T., Peltonen, R., Freitas, C., Duarte,
the colony-stimulating factor 1 receptor (CSF1R) depletes the resident subset of A., Isoniemi, H., Laakkonen, P., Christofori, G., Yla-Herttuala, S., Shibuya, M.,
monocytes and tissue and tumor-associated macrophages but does not inhibit Pytowski, B., Eichmann, A., Betsholtz, C., Alitalo, K., 2008. Blocking VEGFR-3
inflammation. Blood. suppresses angiogenic sprouting and vascular network formation. Nature 454,
Madlener, M., Parks, W.C., Werner, S., 1998. Matrix metalloproteinases (MMPs) and 656–660.
their physiological inhibitors (TIMPs) are differentially expressed during exci- Thakral, K.K., Goodson III, W.H., Hunt, T.K., 1979. Stimulation of wound blood vessel
sional skin wound repair. Exp. Cell Res. 242, 201–210. growth by wound macrophages. J. Surg. Res. 26, 430–436.
Mantovani, A., Sozzani, S., Locati, M., Allavena, P., Sica, A., 2002. Macrophage Tredget, E.E., Yang, L., Delehanty, M., Shankowsky, H., Scott, P.G., 2006. Polarized
polarization: tumor-associated macrophages as a paradigm for polarized M2 Th2 cytokine production in patients with hypertrophic scar following thermal
mononuclear phagocytes. Trends Immunol. 23, 549–555. injury. J. Interferon Cytokine Res. 26, 179–189.
Maraganore, J.M., 1993. Thrombin, thrombin inhibitors, and the arterial thrombotic van der Veer, Bloemen, M.C., Ulrich, M.M., Molema, G., van Zuijlen, P.P., Middelkoop,
process. Thromb. Haemost. 70, 208–211. E., Niessen, F.B., 2009. Potential cellular and molecular causes of hypertrophic
Marin, V., Montero-Julian, F.A., Gres, S., Boulay, V., Bongrand, P., Farnarier, C., Kaplan- scar formation. Burns 35, 15–29.
ski, G., 2001. The IL-6-soluble IL-6Ralpha autocrine loop of endothelial activation van Hinsbergh, V.W., Koolwijk, P., Hanemaaijer, R., 1997. Role of fibrin and plasmino-
as an intermediate between acute and chronic inflammation: an experimental gen activators in repair-associated angiogenesis: in vitro studies with human
model involving thrombin. J. Immunol. 167, 3435–3442. endothelial cells. EXS 79, 391–411.

Please cite this article in press as: Mahdavian Delavary, B., et al., Macrophages in skin injury and repair. Immunobiology (2011),
doi:10.1016/j.imbio.2011.01.001
ARTICLE IN PRESS
G Model
IMBIO-50677; No. of Pages 10

10 B.M. Delavary et al. / Immunobiology xxx (2011) xxx–xxx

Vogler, R., Sauer, B., Kim, D.S., Schafer-Korting, M., Kleuser, B., 2003. Sphingosine- Werner, S., Grose, R., 2003. Regulation of wound healing by growth factors and
1-phosphate and its potentially paradoxical effects on critical parameters of cytokines. Physiol. Rev. 83, 835–870.
cutaneous wound healing. J. Invest. Dermatol. 120, 693–700. Wilson, H.M., Chettibi, S., Jobin, C., Walbaum, D., Rees, A.J., Kluth, D.C., 2005.
Wang, R., Ghahary, A., Shen, Q., Scott, P.G., Roy, K., Tredget, E.E., 2000. Hypertrophic Inhibition of macrophage nuclear factor-kappaB leads to a dominant anti-
scar tissues and fibroblasts produce more transforming growth factor-beta1 inflammatory phenotype that attenuates glomerular inflammation in vivo. Am.
mRNA and protein than normal skin and cells. Wound. Repair Regen. 8, 128–137. J. Pathol. 167, 27–37.
Weitkamp, B., Cullen, P., Plenz, G., Robenek, H., Rauterberg, J., 1999. Human Wynn, T.A., 2008. Cellular and molecular mechanisms of fibrosis. J. Pathol. 214,
macrophages synthesize type VIII collagen in vitro and in the atherosclerotic 199–210.
plaque. FASEB J. 13, 1445–1457.

Please cite this article in press as: Mahdavian Delavary, B., et al., Macrophages in skin injury and repair. Immunobiology (2011),
doi:10.1016/j.imbio.2011.01.001

You might also like