Reff 4

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Chemokine Involvement in Fetal

and Adult Wound Healing

Swathi Balaji,1,{ Carey L. Watson,1,{ Rajeev Ranjan,1 Alice King,1


Paul L. Bollyky,2 and Sundeep G. Keswani1,*
1
Laboratory for Regenerative Wound Healing, Division of Pediatric, General, Thoracic and Fetal Surgery, Cincinnati
Children’s Hospital and the University of Cincinnati, College of Medicine, Cincinnati, Ohio.
2
Division of Infectious Diseases, Department of Medicine, Stanford School of Medicine, Palo Alto, California.

Significance: Fetal wounds heal with a regenerative phenotype that is indis-


tinguishable from surrounding skin with restored skin integrity. Compared to
this benchmark, all postnatal wound healing is impaired and characterized by
scar formation. The biologic basis of the fetal regenerative phenotype can serve
as a roadmap to recapitulating regenerative repair in adult wounds. Reduced
leukocyte infiltration, likely mediated, in part, through changes in the che-
mokine milieu, is a fundamental feature of fetal wound healing.
Sundeep G. Keswani, MD
Recent Advances: The contributions of chemokines to wound healing are a
Submitted for publication September 26, 2014.
topic of active investigation. Recent discoveries have opened the possibility of
Accepted in revised form September 29, 2014. targeting chemokines therapeutically to treat disease processes and improve
*Correspondence: Laboratory for Regenerative healing capability, including the possibility of achieving a scarless phenotype
Wound Healing, Division of Pediatric, General,
Thoracic and Fetal Surgery, Cincinnati Children’s
in postnatal wounds.
Hospital Medical Center, 3333 Burnet Avenue, Critical Issues: Successful wound healing is a complex process, in which there
MLC 11025, Cincinnati, OH 45229-3039 is a significant interplay between multiple cell types, signaling molecules,
(e-mail: sundeep.keswani@cchmc.org)
growth factors, and extracellular matrix. Chemokines play a crucial role in
this interplay and have been shown to have different effects in various stages
of the healing process. Understanding how these chemokines are locally pro-
duced and regulated during wound healing and how the chemokine milieu
differs in fetal versus postnatal wounds may help us identify ways in which we
can target chemokine pathways.
Future Directions: Further studies on the role of chemokines and their role in
the healing process will greatly advance the potential for using these mole-
cules as therapeutic targets.

SCOPE AND SIGNIFICANCE understanding of these mechanisms


Fetal tissue is unique since it has will lead to more successful therapeu-
the ability to heal without scarring, tic interventions in the future. Che-
with total regeneration of epidermal mokines, a class of signaling molecules
and dermal layers, including dermal known for leukocyte recruitment, are
appendages such as hair follicles and pivotal in regulating the wound heal-
sweat glands.1 There are key differ- ing process. However, new and more
ences between the fetal scarless complex roles for chemokines have
wound healing phenotype and adult been discovered in wound healing, in-
wound healing, and although the ex- cluding the regulation of resident
act mechanisms are unknown, a better cell migration, neovascularization,

{
These two authors contributed equally.

660 j ADVANCES IN WOUND CARE, VOLUME 4, NUMBER 11


Copyright ª 2015 by Mary Ann Liebert, Inc. DOI: 10.1089/wound.2014.0564
CHEMOKINES IN FETAL AND ADULT WOUND HEALING 661

inflammation, and tissue repair.2 This review will DISCUSSION OF FINDINGS


begin with a brief introduction to chemokines and AND RELEVANT LITERATURE
then discuss their roles in the wound healing process. Chemokine classification and function
Furthermore, we will discuss some key differences Chemokines are a diverse family of chemotactic
between chemokine expression and function in the cytokines. They are small (8–10 kDa) positively
fetal versus the adult wound environment that may charged secreted proteins that facilitate cell–cell
contribute to the characteristic differences in the communication through both autocrine and para-
wound healing phenotype. crine mechanisms with the specific endpoint of cell
trafficking. Chemokines have been the subject of
TRANSLATIONAL RELEVANCE much research since the 1970s and 1980s, when
Chemokines are intimately involved in the or- they were first discovered,6–8 and a great deal has
chestration of wound healing. They are the impor- been learned about their role in diverse processes,
tant regulators of inflammation, neovascularization, including angiogenesis, inflammation, leukocyte
reepithelialization, and repair processes during trafficking and differentiation, and the response to
wound healing. If we can identify how chemokine infection and autoimmune disease.
expression patterns are different in fetal versus Chemokines share certain structural charac-
adult physiological wound healing, these regula- teristics. They share 20–50% homology in amino
tory molecules and the G-protein–coupled recep- acid sequence,2,9 including the presence of cysteine
tors they signal through could be targeted residues in conserved locations that determine
therapeutically to recreate a more fetal-like their shape.10 They are classified into four families
wound milieu and stimulate regenerative wound based on the number and positioning of the cyste-
healing. Moreover, coadministration of chemo- ine residues: C, CC, CXC, and the CX3C (Fig. 1).
kines (or chemokine pathway inhibitors) might Members of the C chemokine group, such as XCL1
complement the local application of growth fac- (lymphotacin) and XCL2 (SCM-1b), have only two
tors to improve healing through promoting the cysteines, one N-terminal and one downstream. CC
migration of resident and inflammatory cells. chemokines have two adjacent cysteines near their
Despite the great promise of chemokine-targeted N-terminal end. An example of a CC chemokine is
therapy, much work still needs to be done to de-
termine whether control of this signaling system
will prove to be fruitful from bench to bedside.

CLINICAL RELEVANCE
In the developed world, more than 100 million
patients acquire scars each year, many of which
cause significant morbidity. There are an esti-
mated 11 million keloid scars and 4 million burn
scars.3,4 Although global figures are unknown,
they are likely much higher. Furthermore, al-
though there is a paucity of data that adequately
describes the negative physical impact of scars on a
patient’s quality of life, scars can also cause psy-
chological impact with anxiety and negative impact
on self-consciousness.5 By understanding the un-
derlying molecular mechanisms in the fetal re-
generative response to injury, we may be able to
Figure 1. Chemokine family structure. Chemokines contain cysteines
develop therapies based on this knowledge to re- (usually four) in conserved positions. The spacing between the first two
capitulate the fetal phenotype in postnatal tissues. cysteines determines the type of chemokine. The C subfamily contains only
Patients would benefit from improved healing, and one of the proximal n-terminal cysteines. In the CC subfamily, the first two
cysteines are adjacent to each other, in the CXC family, there is one amino
care providers would be able to have more influence
acid between the first two cysteines, and in the CX3C family, the two
over the healing process after invasive procedures. cysteines are separated by three amino acids. The first cysteine (C) in the
Chemokines undoubtedly play a pivotal role in the sequence forms a covalent bond with the third, the second and the fourth
healing process and further investigations into cysteines also form a disulfide bond to create the tertiary structure char-
acteristic of chemokines. To see this illustration in color, the reader is
their mechanisms of action may allow us to im-
referred to the web version of this article at www.liebertpub.com/wound
prove outcomes in our patients.
662 BALAJI ET AL.

CCL-2, also known as the monocyte chemoat- cell surface, chemokines are bound to extracellular
tractant protein-1 (MCP-1), which induces trans- matrix (ECM) proteoglycans, particularly those
endothelial migration of monocytes into sites of containing the heparin sulfate glycosaminoglycan.
tissue remodeling to become macrophages.11 The resulting sequestration and diffusion of che-
Members of the CXC group share two conserved mokines create gradients within tissues that gov-
cysteines separated by an amino acid.9 The CXC ern cell chemotaxis.15 All chemokines signal
family is further divided into two groups based on through the G-protein-linked transmembrane re-
the presence of glutamic acid (E), leucine (L), and ceptors. As with the chemokines themselves, the
arginine (R) immediately before the first cysteine. chemokine receptors are likewise divided into four
This ELR + sequence is important for receptor se- families with distinct chemokine-binding proper-
lectivity and binding. An example of an ELR + CXC ties: XCR1 binds XC chemokines, CCR1-10 recep-
chemokine is interleukin (IL)-18, which is a che- tors bind CC chemokines, CXCR1-7 receptors bind
moattractant for neutrophils.12 An example of an CXC chemokines, and CX3CR1 binds CX3CL1. Al-
ELR - chemokine is CXCL13, which induces lym- though some chemokine–chemokine receptor in-
phocyte egress into surrounding tissues.13 Of note, teractions are selective, many chemokine receptors
ELR + CXC chemokines are said to be angiogenic, bind multiple chemokines, engendering both re-
whereas chemokines lacking this sequence (ELR - dundancy and plasticity in chemotactic responses.16
CXC chemokines) are said to be angiostatic.14 The On a functional basis, chemokines can be catego-
CX3C family contains two cysteines separated by rized as either being homeostatic, in which case they
three amino acids. The only CX3C chemokine are constitutively expressed, or inflammatory, in
identified to date is CX3CL1 (fractalkine). Over 45 which case they are present only in the setting of an
chemokines have been reported to date and many inflammatory response. These distinctions are likely
of them impact the behavior of cell types that in- to be highly relevant to chemotaxis during wound
fluence cutaneous wound healing (Table 1). healing responses, where an inflammatory influx
Along with the composition of the local chemo- occurs into previously homeostatic tissues.9 How-
kine milieu, chemokine diffusion and clearance are ever, in debilitating ischemic or chronic wounds that
also tightly regulated. Within tissues and at the are associated with poor wound healing outcome,

Table 1. Chemokines and chemokine receptors in wound healing

Chemokines

Systemic name Human ligand Mouse ligand Chemokine receptors Target cells in wound healing

CCL1 I-309/TCA-3 TCA-3 CCR8 Monocytes, macrophages


CCL2 MCP-1 MCP-1 CCR2 Monocytes, T lymphocytes, mast cells, keratinocytes, endothelial cells, fibroblasts
CCL3 MIP-1a MIP-1a CCR4, CCR5 Monocytes, macrophages
CCL4 MIP-1b MIP-1b CCR1, CCR5 Monocytes, macrophages
CCL5 RANTES RANTES CCR1, CCR3, CCR4, CCR5 Monocytes, macrophages
CCL7 MCP-3 MARC CCR1, CCR2, CCR3, CCR5 Neutrophils
CCL8 MCP-2 MCP-2 CCR1, CCR2, CCR3, CCR5 Mast cells, monocytes, T lymphocytes
CCL22 MDC ABCD-1 CCR4 T lymphocytes
CCL23 MPIF-1 CCL6, C10 CCR1 Neutrophils, monocytes, T lymphocytes
CCL27 CTACK/ILC CTACK/ILC CCR10 T lymphocytes
CXCL1 GROa/MGSAa GRO/KC CXCR2 Neutrophils, keratinocytes, endothelial cells, fibroblasts
CXCL2 GROb/MGSAb GRO/KC CXCR2 Endothelial cells
CXCL3 GROg/MGSAg GRO/KC CXCR2 Endothelial cells
CXCL4 PF4 PF4 CXCR3b Fibroblasts
CXCL5 ENA-78 GCP-2/LIX CXCR2 Neutrophils
CXCL6 GCP-2 GCP-2/LIX CXCR1, CXCR2 Neutrophilic granulocytes
CXCL7 NAP-2 NAP-2 CXCR2 Neutrophils, leucocytes, macrophages, keratinocytes, endothelial cells, fibroblasts
CXCL8 IL-8 MIP-2 CXCR1, CXCR2 Neutrophils, leucocytes, macrophages, keratinocytes, endothelial cells, fibroblasts
CXCL9 MIG MIG CXCR3 T lymphocytes, endothelial cells, fibroblasts
CXCL10 IP-10 IP-10 CXCR3 T lymphocytes, endothelial cells, fibroblasts
CXCL11 I-TAC I-TAC CXCR3 T lymphocytes
CXCL12 SDF-1a/b SDF-1a/b CXCR4 T lymphocytes, keratinocytes, endothelial cells, endothelial progenitor cells
CXCL13 BLC/BCA-1 BLC/BCA-1 CXCR5 B cells, T lymphocytes
CX3CL1 Fractalkine Fractalkine CX3CR1 Fibroblasts, NK cells, T cells, endothelial cells, epithelial cells, macrophages,
and vascular smooth muscle cells

IL, interleukin; MCP, monocyte chemoattractant protein; MIP-1a or b, macrophage inflammatory protein-1 (MIP-1a = CCL3; MIP-1b = CCL4); RANTES,
regulated on activation, normal T expressed and secreted (CCL5); SDF-1, stromal cell-derived factor-1.
CHEMOKINES IN FETAL AND ADULT WOUND HEALING 663

such as in diabetes, the distinctions between basal the inflammatory phase. Chemokine CXCL4 par-
and inflammatory chemokine levels blur, with im- ticipates in the initial coagulation process and
portant consequences for immune regulation at prevents the premature development of blood ves-
these sites.17 Along with these contextual associa- sels. Platelet degranulation releases a plethora of
tions, chemokines have particular tropisms in terms cytokines that activate the tissue resident cells,
of the cell types they impact, reflecting the distri- including the macrophages, keratinocytes, and fi-
bution of chemokine receptors (Table 1). broblasts. These activated cells in turn produce
inflammatory mediators such as the proin-
Chemokine expression/function during
flammatory chemokines CXCL8 and CCL2, which
the different phases of wound healing
promote the directional migration of inflammatory
Normal wound healing occurs through a series of
cells and endothelial cells to the wounded area,
sequential overlapping stages (Fig. 2). During
further regulating the inflammation and angio-
these stages, chemokines control the trafficking of
genesis at the tissue repair site.
specialized cell types to local sites of injury in a
There are some suggestions that chemokine ex-
time- and context-dependent manner. In this
pression in fetal wounds during hemostasis is dif-
study, we will address the fundamental biology of
ferent from that seen in postnatal wounds. Porcine
chemokines in wound healing, organized according
fetal platelets may release less cytokines (and
to the stages of a normal wound response.
presumably chemokines).18 It may also be the case
The hemostasis stage of wound healing that fetal tissues inhibit platelet-mediated chemo-
The initial stage of a wound response is hemo- taxis. Olutoye et al.19 had demonstrated that hya-
stasis, occurring within minutes to hours of injury. luronan (HA), the predominant glycosaminoglycan
Bleeding as a consequence of tissue injury results GAG in the fetal dermal matrix, inhibits platelet
in the release of platelets, plasma fibronectin, and aggregation and cytokine release. The authors
prothrombin and in the formation of a clot that concluded that the HA-induced inhibition of
serves as a provisional matrix for the initiation of platelet aggregation and resultant attenuation of

Figure 2. Chemokines and their roles in the various phases of wound healing. Normal wound healing progresses through a series of sequential overlapping
stages. The initial stage of a wound response is hemostasis, occurring within minutes to hours of injury. Clot formation occurs that sets the stage for the
inflammatory phase of healing, which begins with neutrophils coming in first followed by macrophages. This phase is followed by proliferative and maturation
phases. Reepithelialization and granulation tissue formation in which the keratinocytes migrate to cover the wound, and the wound tissue begins its repair by cell
proliferation, ECM production, and blood vessel development. Finally, during the remodeling, many of the extracellular elements are removed by apoptosis, and the
ECM is remodeled. During these stages, chemokines control the trafficking of specialized cell types to local sites of injury in a time- and context-dependent
manner. ECM, extracellular matrix. To see this illustration in color, the reader is referred to the web version of this article at www.liebertpub.com/wound
664 BALAJI ET AL.

inflammatory response may explain, in part, the expression pattern and function of neutrophils in
minimal inflammation and modest cellular infil- fetal versus adult wound healing29 that may dis-
trates, characteristic of fetal wounds. However, tinguish whether a wound heals regeneratively or
fetal platelets may also produce factors that ac- with a scar.30 Adult neutrophils are characterized
tively promote tissue regeneration, in part, by a much higher expression of cell adhesion mol-
through the recruitment of mesenchymal stem ecules than the fetal neutrophils,31 which may in-
cells (MSCs).20 Further elucidation of the chemo- duce increased neutrophil–endothelial interaction
kine profile from fetal tissues during homeostatic and thereby increased migration of neutrophils into
conditions and from the midgestation scarless the wound bed that then can contribute to scarring
wound healing, late-gestation transitional wound phenotype of the adult wounds.32 In comparison,
healing, and postnatal stages is required to un- fetal wounds have in fact been demonstrated to have
derstand the factors leading to reduced inflamma- a smaller fraction of neutrophils. In addition, the
tory infiltrates in fetal wounds. expression levels of proinflammatory chemokine
IL-8 (CXCL8) are higher during adult wound repair
The inflammatory stage of wound healing compared to a much lower expression in the mid-
Hemostasis sets the stage for the inflammatory gestational fetal wounds that heal scarlessly.33,29
stage of wound healing, occurring in the first 72 h of Since CXCL8 is the main neutrophil chemoat-
a wound response. The inflammatory stage is tractant, this could explain the difference between
characterized by an influx of inflammatory cell the number and activation of neutrophils in fetal
types and the increase in proinflammatory media- versus adult wounds.
tors is highly regulated by chemokines. However, Following the initial influx of neutrophils to kill
in fetal wound healing, numbers of inflammatory potential pathogens and decontaminate the
cells are reduced, they are less activated, and have wound, a switch in the leukocyte recruitment oc-
more short-lived responses.21 curs that favors the recruitment of cells of mono-
The rapid influx of inflammatory cell types be- cyte/macrophage lineage to the wound.22 The
gins acutely after injury with the release of plate- differential release of their respective chemoat-
lets and neutrophils directly from circulation into tractants in the wound microenvironment, which
the wound. Activated platelets release a multitude is, in part, orchestrated by IL-6 and its soluble re-
of inflammatory mediators, which promote the di- ceptor,34 facilities the temporal switch in the leu-
rectional migration of leukocytes to the wounded kocyte recruitment from neutrophils to monocytes.
area. Neutrophils dominate the initial influx of Neutrophils themselves are thought to mediate
leukocytes, and at day 1 postinjury, neutrophils this switch by releasing soluble factors into the
have been shown to constitute nearly 50% of wound early inflammatory wound microenvironment that
cells.22 Neutrophils express CXC chemokine re- then initiates monocyte recruitment.35 MCP-1,
ceptor 2 and are recruited in response to CXCL8 also referred to as CCL-2, is a predominant che-
(IL-8), a potent proinflammatory chemokine that is mokine implicated in monocyte recruitment.
involved in this process initially.2 In humans, Monocytes express chemokine receptor 2 that plays
CXCL8 is a potent activator and chemoattractant an important role in their response to MCP-1. In
of neutrophils and as neutrophils arrive at the new addition to infiltrating neutrophils, MCP-1 is also
wound, they also secrete CXCL8, further aug- known to be predominantly produced in situ by
menting the effect.23 It is suggested that CXCL8 tissue-resident macrophages at the site of wound
also plays a role in the inhibition of neutrophil healing.36 Monocyte chemotaxis is also partly reg-
apoptosis, thus potentiating the inflammatory ulated by other CC chemokine family members
process.24 CXCL8 also stimulates endothelial per- such as CCL5 (regulated on activation, normal T
meability, thereby facilitating leukocyte migration expressed and secreted [RANTES]), CCL3 (mac-
into injured tissues,25,26 and has been shown to rophage inflammatory protein [MIP]-1a), CCL4
have increased expression levels in chronic (MIP-1b), CCL1 (I-309), and CCL-7 (MCP-3).23
wounds.9 Other inflammation-related chemokines Monocytes differentiate into macrophages once
that may play a role after injury are CXCL1, 4, 5, 6, within the wound and play an essential role in the
and 7. Each of these chemokines, along with removal of debris and dead cells, as antigen-pre-
CXCL8, forms gradients through interactions with senting cells, and as secretors of additional growth
proteoglycans, rendering directional movement of factors. A study by Kaesler et al.37 demonstrated
leukocytes into wounded areas.23,27,28 that injury induced the expression of chemokine
Neutrophils are also present in fetal wound CCL-6 (C10), which is a potent chemoattractant
healing, but there are important differences in the for macrophages and may contribute to strong
CHEMOKINES IN FETAL AND ADULT WOUND HEALING 665

macrophage influx observed in cutaneous wound resulted in the augmented healing response, char-
healing. There are differences between monocytes acterized by improved collagen fiber organization
and macrophages within the fetal and adult and reduction in wound scar width, without nega-
wounds, with a decrease in the number and per- tive effects on wound breaking strength. These data
sistence of these cell types, as well as macrophages demonstrate that chemokine regulation can offer a
in their active and mature forms in fetal wounds.38 potential therapeutic benefit for wound healing.
It is unclear whether these differences are attrib- Lymphocytes are the final inflammatory cell
utable to differences in chemokine secretion. type to be recruited to the wound, where they serve
Another cell type, mast cells, is not only resident as the immunological effector cells and producers
in the skin but also recruited to wounds and is one of of additional growth factors. CCL3, CCL4, and
the initial cell types to respond to trauma with the CCL5 are also chemoattractants of lymphocytes
release of inflammatory mediators. There is growing and are produced in granulation tissue.2 Specifi-
evidence to demonstrate that chemokines and their cally, B lymphocytes produce antibody responses to
receptors regulate mast cell tissue localization and antigens present in the wound, and T lymphocytes
function. Human mast cells of different origin ex- produce cytokines and promote the cytolytic ac-
press several chemokine receptors (CXCR1, CXCR2, tivity. There are also differences in lymphocyte
CXCR3, CXCR4, CX3CR1, CCR1, CCR3, CCR4, and responses between fetal and adult wounds. While B
CCR5). Seven chemokines (CXCL1, CXCL5, lymphocytes were seen in adult mouse wounds,
CXCL8, CXCL14, CX3CL1, CCL5, and CCL11) have they were not present in fetal wounds.38
been shown to act on some of these receptors and to In addition to the specific roles for specific cell
induce mast cell migration.39 The chemokine MCP-1 types in wound healing, there are data implicating
has been shown to be a potent attractant for mast specific chemokines. CX3CL1 and its receptor
cells, which are increasingly considered to be im- CX3CR1 is particularly highly expressed in murine
portant mediators in wound healing.40 Mast cells models of skin wound healing.45 CX3CL1 is ex-
degranulate after injury releasing multiple proin- pressed by endothelial cells, epithelial cells, macro-
flammatory mediators and vasoactive amines41 and phages, and vascular smooth muscle cells. CX3CR1
have also shown to be abundant in both keloids and is expressed by monocytes/macrophages, fibroblasts,
hypertrophic scars.42 Mast cells also release che- NK cells, a subpopulation of T cells, and smooth
mokines such as IL-8, MIP-1a, and MIP-1b inde- muscle cells. Loss of CX3CR1 function delayed
pendent of degranulation, when stimulated through wound closure in CX3CR1 knockout mice, charac-
the activation of the CD30 pathway.43 Mast cells terized by reduced macrophages and macrophage
synthesize IL-4 and stimulate fibroblast prolifera- products, such as transforming growth factor-b
tion.40 Mast cells can have both stimulatory and (TGF-b)1 and vascular endothelial growth factor,
inhibitory functions in skin inflammation that are reduced myofibroblasts and collagen deposition, and
regulated by chemokines (reviewed by Harvima reduced neovascularization. The transfer of normal
et al.44). In fetal skin that heals with the scarless bone marrow for wild-type mice rescued the pheno-
phenotype (embryonic day 15 [E15] in murine type in these mice, suggesting an important profi-
model], mast cells differ in size (smaller), number brotic and angiogenic role for the CX3CL1/CX3CR1
(decreased), granularity (less), and maturity when axis in cutaneous wound healing. Since the CX3CL1/
compared with mast cells in more mature skin (E18) CX3CR1 axis is associated with several cell types
and they do not degranulate as effectively. Con- and signaling factors (including TGF-b1, myofibro-
sistent with this, Wulff et al. demonstrated that in- blasts, and collagen deposition) that distinguish the
jection of mast cell lysates into E15 wounds fetal wound healing phenotype from adult pheno-
disrupted the scarless healing phenotype and that type, it can be hypothesized that targeted manipu-
mice deficient in mast cells when injured at E18, lations of this axis may facilitate recapitulating
healed with significantly smaller scars, suggesting fetal-like wound healing in postnatal wounds.
that mast cells may influence the scarless healing Along with differences in cell types, fetal and
phenotype.42 Mast cells are also increased in the adult wounds also differ in terms of their ECM
skin in chronic skin inflammatory conditions, for milieu in ways that likely influence wound healing
example, psoriasis, basal cell carcinoma, and and inflammatory responses. The fetal wound
chronic ulcers. Disodium cromoglycate, a pharma- ECM is significantly different from the adult
cologic inhibitor of mast cell activation and degran- wound environment. The ECM of fetal scarless
ulation, has been used to decrease the amount of wounds is characterized by the increased levels of
inflammatory cytokines such as IL-1b and chemo- type-III to type-I collagen ratio and high and per-
kine CXCL1 (Murine IL-8)41 in the wounds, which sistent levels of high-molecular-weight HA. This
666 BALAJI ET AL.

distinct ECM serves as structural scaffolding that important both for remodeling and granulation
facilitates increased cellular migration. On the tissue formation. They are also known to regulate
other hand, in adult wounds, HA levels are only the inflammatory process by the release of inflam-
transiently elevated and low-molecular-weight matory mediators.49 They are one of the more po-
(LMW) HA predominates. The LMW form of HA tent effector cells that manipulate the chemokine
( < 200 kDa) is known to accumulate at sites of in- expression using autocrine and paracrine mecha-
flammation, influencing various activities, includ- nisms. Fibroblasts also manipulate chemokine
ing monocyte activation, leukocyte adhesion to signaling by being target cells for chemokines.
endothelium, and smooth muscle cell migration, For example, MCP-1 can induce the expression of
after wound healing. In addition, it has also been TGF-b and collagen synthesis by rat fibroblasts.50
shown to induce chemokines such as MCP-1, MCP-1 has also been shown to enhance MMP-1 and
RANTES, and IL-8, which can alter the wound TIMP-1 gene expression in primary human dermal
healing phenotype. HA is also shown to suppress fibroblasts.51 This MCP-1-mediated fibroblast sig-
platelet aggregation and the release of growth naling can further influence the chemokine milieu.
factors in a dose-dependent manner. Matrix turn- CXCL11 is important in dermal–epidermal inter-
over also regulates the wound repair outcome. actions and in maturation of the healing tissue.
Dang et al.46 demonstrated that the E16 mid- Feugate et al.52,53 demonstrated that the chicken
gestation murine wounds that completely re- chemotactic and angiogenic factor, a CXC chemo-
epithelialize within 72 h and heal without a scar kine ortholog of human IL-8, stimulates fibroblasts
have greater expression of matrix metalloprotei- to produce early granulation tissue and ECM
nases (MMPs) relative to tissue inhibitors of me- components, including tenacin, fibronectin, and
talloproteinases (TIMP), which favors ECM collagen, as well as stimulates the differentiation of
turnover, faster migration of cells, and promotes fibroblasts to myofibroblasts.
scarless repair compared to the late-gestation Previous reports have described functional dif-
scarring wounds at E19. A study by Bullard et al.47 ferences between adult and fetal fibroblasts, in-
demonstrated that the fetal skin has greater cluding more efficient migration. Fetal fibroblasts
amounts of MMPs (interstitial collagenase, stimulated by lipopolysaccharide or platelet-derived
stomelysin-1, and gelatinase-A) than adult skin, growth factor have an attenuated IL-6 and IL-8 re-
with the expression pattern localized to dermal sponse compared to adult fibroblasts, suggesting
cells, keratinocytes, and around vascular struc- that even when stimulated to release proin-
tures, implicating a significant role in wound flammatory cytokines, the response of the fetal fi-
healing and neovascularization. Chemokine sig- broblast is blunted.33,54 It is suggested that some
naling can be significantly influenced by MMPs in functional differences observed between adult and
several ways, including the release of chemokines fetal fibroblasts may be a direct result of the unique
bound to the cell surface or ECM, which can impact HA-rich ECM observed in the fetus. Fibroblasts
chemokine gradients, inactivation of the chemokine, have also been shown to have differences in gene
and/or generation of more powerful chemotactic or expression between murine (E15) and (E18) stages,
antagonistic derivatives (reviewed by Van Lint and revealing upregulation of several inflammatory
Libert48). These findings reiterate the association genes, for example, cyclooxygenase-1 (COX-1) in the
between inflammation, ECM deposition, ECM E18 fibroblasts versus E15.49,55 Although not much
turnover, and remodeling, and suggest that chemo- is known about the specific role of chemokines in the
kines may be the major regulators of the interde- fetal scarless healing phenotype, differences be-
pendence between these wound healing processes. tween all the inflammatory cell subtypes and fibro-
blasts suggest that chemokine differences are likely
The proliferation and maturation
and may serve as potential targets for therapeutic
stage of wound healing
intervention in the healing process.
Inflammation sets the stage for the proliferative
stage, occurring in the first 2–3 weeks after
Reepithelialization
wounding. The proliferation phase is characterized
Reepithelialization of the wound during the
by granulation tissue formation, reepithelializa-
proliferative stage is an important process, which
tion, and neovascularization.
occurs through the proliferation and migration of
Granulation tissue formation keratinocytes from wound edges to cover the
The chemokine milieu regulates the movement wound. Factors released by platelets such as epi-
of resident and infiltrating wound cells as well as dermal growth factor and TGF-b stimulate the
influences tissue remodeling. Fibroblasts are keratinocytes near the wound edge to proliferate,
CHEMOKINES IN FETAL AND ADULT WOUND HEALING 667

and other factors released by other cell types in the sue by means of endothelial cell proliferation and
wound, including CXCL1 (growth-related onco- sprouting from preexisting microvasculature in a
gene alpha [Gro-a]), CXCL8 (IL-8), fibroblast process called angiogenesis. Multiple chemokines
growth factor, and keratinocyte growth factor, fur- and cytokines produced by a variety of wound cells
ther stimulate the proliferation and migration of influence the angiogenesis process.65 The tissue-
keratinocytes.2 CXCL8, specifically, is expressed resident cells, including macrophages, keratino-
highly along the denuded wound surface and both cytes, and fibroblasts, produce angiogenic growth
CXCL8 and CXCL1 bind to CXCR2, expressed on factors such as VEGF, while in parallel, release of
keratinocytes.23,56 In addition, CXCL1 and CXCR2 the proteolytic enzymes allows ECM degradation
are expressed in normal human epidermis and they and endothelial cell migration.
have been shown to be induced during the wound When the wound defect is filled, angiogenesis
healing phases of inflammation, reepithelialization, has to cease. A temporal change in the balance
and neovascularization of human burn wounds.57 between proangiogenic and angiostatic factors
Mice lacking CXCR2 receptors were observed to regulated by chemokines plays an important role in
have severely impaired reepithelialization in vivo.58 the orchestration of this cessation. Endothelial
Conversely, topical application of CXCL8 or CXCL1 cells differentially express several chemokines in
appears to have favorable effects on wound healing response to the wound microenvironmental stim-
in mouse models.59,60 uli.66 These include MCP-1 and RANTES, as well
Keratinocytes migrate through the new ECM as IL-8, Gro-a, Gro-b (CXCL2), and Gro-c (CXCL3).
through integrin interaction with receptors on the CTAP-III, b thromboglobulin, and NAP-2 have also
cells, and reepithelialization is therefore depen- been described to induce endothelial cell prolifer-
dent upon the presence of proteases such as plas- ation in vitro and angiogenesis in vivo. ELR + che-
min and MMPs for breakdown of ECM during mokines IL-8 and Gro-a are significantly induced at
migration.2,61 Another chemokine, CXCL11, is the neovascularization sites in the wound granu-
produced by basal keratinocytes and promotes the lation tissue immediately after wounding and then
migration of undifferentiated keratinocytes into markedly decline after day 4.22 The CXCR2 che-
the wound and therefore playing an active role in mokine receptor, which binds to all ELR + chemo-
reepithelialization.62 Interestingly, another che- kines, is also believed to play an important role in
mokine, CCL27, has been reported to regulate the the mediation of neovascularization (reviewed by
recruitment of bone marrow-derived keratinocyte Gillitzer and Goebeler23). Furthermore, chemo-
stem cells to wounds through its receptor CCR10, kines indirectly influence neovascularization by
suggesting chemokine-mediated recruitment of facilitating the recruitment of support cells that
keratinocytes from outside of the wound.63 are essential for tissue revascularization. DiPietro
Undoubtedly, chemokines play a vital role in the et al.67 reported that depletion of MIP-1a signifi-
reepithelialization process. Little is known about cantly reduced the angiogenic activity of murine
how chemokines impact keratinocytes in fetal wounds. It was suggested that MIP-1a promotes
scarless wound healing. Studies have shown that recruitment of macrophages to wound sites, which
activated epidermal keratinocytes near the wound in turn act as a source of angiogenic cytokines.
edge in fetal wounds have an upregulation of epi- Several ELR - CXC chemokines, including in-
dermal integrin receptor, specific for fibronectin, terferon-c-inducible cytokines IP-10, CXCL-9
tenascin, collagen, laminin, and other proteins in (Mig), CXCL4, 9, 10, and 11 or I-TAC exhibit
the provisional ECM, allowing rapid proliferation growth inhibitory functions, including inhibition of
and migration of keratinocytes in fetal wounds.30 angiogenesis. IP-10 and Mig have also been shown
Another study, using allogenic peripheral blood to be highly expressed after day 4 during the later
mononuclear cell proliferation tests, demonstrated stages of normal wound healing,22 with an inhibi-
that fetal keratinocytes and fibroblasts possessed tory action on fibroblast motility and recruitment.
immunosuppressive properties, in part, mediated While there is clearly a role for chemokines in the
by IL-8, vascular endothelial growth factor (VEGF), regulation of wound neovascularization and mat-
and granulocyte macrophage colony stimulating uration, the exact differences between the fetal and
factor (GM-CSF),64 suggesting that these cells po- adult phenotypes are not completely understood.
larize the cytokine/chemokine milieu.
Remodeling
Neovascularization During wound maturation and remodeling, the
During granulation tissue formation, new blood ECM turnover occurs with disorganized collagen
vessels are formed to revascularize the wound tis- fibers being rearranged and crosslinked, along
668 BALAJI ET AL.

with a decrease in cellularity and regression of the injuries,76 where they engraft and contribute to
neovasculature. Fibroblasts and myofibroblasts tissue recovery.76 Interestingly, recent reports
synthesize mature connective tissue as well as demonstrated that mechanical stretching can up-
produce metalloproteinases that remove the na- regulate stromal cell-derived factor-1 alpha (SDF-
scent matrix and facilitate matrix maturation. In 1a) and hypoxia-inducible factor-1 alpha in skin
postnatal tissues, this results in scar formation, and recruit circulating MSCs and other stem cells
while fetal wound healing is associated with re- through the SDF-1a/CXCR4 pathway.77 The sig-
generation of specialized dermal structures. Che- naling between SDF-1 and its receptor CXCR4 is
mokines CXCL9, CXCL10, and CXCL11 are critical for the homing and recruitment of circu-
important in the process of epidermal and dermal lating progenitor cells after tissue injury and is-
maturation and regression of neovasculature. The chemia. SDF-1 is a potent chemoattractant for
wound repair and regeneration is thought to be EPCs. The other major chemokines and respective
further influenced by stem cell recruitment and receptors that regulate EPC activation and homing
mechanical forces that impact the wound bed, both are IL-8 and CXCR2, growth-regulated oncogene-a
of which are governed, in part, by chemokines. and CXCR1, CCL5, CCR5, and C–C chemokine and
chemokine (C–C motif) receptors 2 and 5.78 Upon
Stem cell recruitment. Tissue repair and re- interaction with tissue-specific chemokines, EPCs
generation involve selective recruitment of circu- become activated and initiate integrin-mediated
lating or resident stem cell populations (Fig. 3). adhesion to endothelial vascular cells and, conse-
Transition from regenerative to scar forming cu- quently, transendothelial migration into sites of
taneous repair coincides most evidently with the vascular and tissue remodeling. CXCL8 stimulates
development and maturation of the hair follicle. vascular permeability, which is important for both
The role of chemokines in the folliculogenesis is not angiogenesis and vasculogenesis. EPC invasion
elucidated. In response to wound healing, stem into the vascular injury site depends on the
cells from the basal layer of the epidermis and from breakdown and remodeling of the vessel basement
the bulge area of the hair follicles get activated and membrane and the interstitial wound space medi-
migrate into the wound to contribute to the re- ated by extracellular proteases. As discussed pre-
epithelialization and wound healing processes.68,69 viously, chemokines play a major role in the
Chemokines and chemoattractant signals also regulation of the ECM remodeling and may there-
regulate the mobilization and homing of bone by influence wound neovascularization mediated
marrow-derived as well as circulating stem cell through both angiogenesis and stem cell-depen-
populations to the tissue repair sites, including dent vasculogenesis mechanisms.
MSCs and endothelial progenitor cells (EPCs). Amniotic stem cells are known to enhance fetal
Under the guidance of chemokine signaling, MSCs wound healing responses in animal models.79 Fur-
can selectively migrate to injured sites, including thermore, Roubelakis et al.20 recently reported that
skin wound healing,63,70,71 bone fractures,72,73 platelet-rich plasma promotes regenerative wound
myocardial infarctions,74,75 and ischemic cerebral healing through recruitment of MSCs. Together

Figure 3. Chemokine gradient. Establishment of a chemokine concentration gradient after injury and hypoxia, which facilitates directional migration and
recruitment of bone marrow-derived as well as circulating stem and progenitor cells, inflammatory cells, as well as tissue-resident cells to the sites of tissue
repair. To see this illustration in color, the reader is referred to the web version of this article at www.liebertpub.com/wound
CHEMOKINES IN FETAL AND ADULT WOUND HEALING 669

these data suggest that there may be a role


TAKE HOME MESSAGES
for circulating stem and progenitor cells in
Basic science advances
the fetal response that may similarly be, in
Chemokines are a family of small cytokines that could be considered as one
part, regulated by chemokines. This is an
of the major regulators of wound healing. They play a significant role as soluble
exciting area for future research.
mediators and adhesion factors involved in the recruitment and trafficking of
inflammatory as well as tissue-resident cells. Chemokine expression and
Mechanotransduction. Not all mid- function vary in a temporal and spatial manner during the different phases of
gestation fetal wounds heal without scars. wound healing. They are both mechanosensitive and mechanomodulatory. They
In fact, larger excisional wounds in the fetus regulate the sequential orchestration of wound healing by governing angio-
heal with a characteristic postnatal scar genesis, granulation tissue production, and matrix catabolism that together
formation at gestational age that should mediate the remodeling of the wound tissue.
heal without scar. The larger scarring fetal The midgestation fetus is capable of regenerative wound healing and is
wound may have increased shear forces and characterized by an anti-inflammatory milieu with decreased levels of IL-8 and
mechanical stress, suggesting that biome- IL-6, decreased neovascularization, and significantly reduced mechanical
chanical forces may be, in part, an under- stresses, suggesting a preeminent role for chemokines in regulating the fetal
lying mechanism of the fetal regenerative scarless phenotype. Further understanding of this can lead to the development
phenotype. In fact it has been well estab- of targeted therapeutics to recapitulate fetal-like scarless wound healing in
lished that during the remodeling phase of postnatal wounds with multiple etiologies.
wound healing, myofibroblasts undergo ap-
Clinical science advance
optosis, which is thought to result from a Chemokine activation of G-protein receptors makes them very amenable to
release of mechanical tension after their the development of small molecules that serve as agonists or antagonists of
contraction of the nascent matrix.80–82 In- function for therapeutic wound healing purposes. In addition, neutralizing an-
creased mechanical force induces local pro- tibodies directed to either the chemokine or its receptor can be added to the
duction of profibrotic growth factors, such as wounds to promote wound healing. For example, CCR5 and CXCR4 antagonists
an increase in TGF-b1 and TGF-b2. Several are being tested in several preclinical animal models and in clinical trials to
studies have demonstrated that mechanical improve healing outcomes.
stretch can change the expression pattern of More studies on the role of chemokines and their receptors need to be
chemokines. IL-1b, tumor necrosis factor-a, performed to use them effectively in the clinic.
IL-6, IL-8, COX-2, prostaglandin E2, pre-B-
cell colony-enhancing factor, monocyte Relevance to clinical care
The therapeutic applications of chemokines may be beneficial to any pa-
chemotactic and activating factor/MCP-1,
thology that is characterized by unresolved wound healing and excess fibrosis,
granulocyte colony-stimulating factor, GM-
including cancer, chronic wounds, pulmonary fibrosis, renal fibrosis, hypertro-
CSF, and macrophage colony-stimulating
phic scarring, and keloids. Chronic inflammation and alteration in angiogenesis
factor are now well recognized as stretch- can potentially be reduced or eliminated by interfering with proinflammatory or
responsive genes.83,84 Interestingly, several angiogenic chemokines and/or their receptors. Similarly, neutralization of pro-
studies have demonstrated in both in vitro fibrotic chemokine activity can greatly reduce fibrosis. However, one chemokine
and in vivo models that mechanical stretch may have different downstream effects on different cell types in the wound, and
increases the production of LMW HA as targeted inhibition/or activation of chemokines may alter multiple wound out-
well as an increase in IL-8 production.85 comes. Thus, further detailed study of the mechanisms and effects of chemokine
The implications of decreasing trans- manipulation is necessary before targeting these pathways and can become a
duction of profibrogenic mechanical stress clinical reality.
are readily translated to postnatal wounds
in the effort to ameliorate scar formation.
Mechanomodulatory devices designed to offload influenced by regulatory molecules such as the che-
wound stress have demonstrated a significant de- mokines. Chemokines play multiple roles in injury
crease in scarring with reduced area of fibrosis in repair, not confined to their well-characterized roles
large animal studies.86 However, the exact regu- in leukocyte chemotaxis and angiogenesis. It is
lation of the chemokine expression by these devices becoming clear that chemokines also play an inte-
has to be elucidated. gral part in reepithelialization, granulation tissue
formation, and remodeling of the healing wounds.
Alteration in chemokine expression, longevity, and
FUTURE DIRECTIONS localization can result in pathological healing
The studies reviewed here show that normal states. In a recent murine model, overproduction of
wound healing requires the sequential stimulation CXCL10, very early after wounding, resulted in
and resolution of multiple phases, which are abnormal initiation and resolution of inflammation
670 BALAJI ET AL.

and impaired healing, presumably because the (S.G.K.), the Wound Healing Society Foundation
normal function of CXCL10 is to attract lympho- 3M Award (S.G.K.), the NIH/NIDDK award
cytes to the wound tissue in the later stages of 1R01DK096087 (P.L.B.), and the NIH/NHLBI
healing to finalize the process. It has been sug- award 1R01HL113294-01A1 (P.L.B.).
gested that the change in the expression of this
chemokine resulted in the presence of lymphocytes
AUTHOR DISCLOSURE
at the same time as neutrophils and macrophages,
AND GHOSTWRITING
leading to confusion in the orchestrated progres-
sion of wound healing, ultimately resulting in There are no competing financial interests. The
impaired healing. Chemokines may also play a content of this article is expressly written by the
critical role in the pathogenesis of chronic wounds, authors listed. No ghostwriters were used to write
not only by altering the inflammatory and angio- this article.
genesis pathways, but also through their role in
combating with biofilm forming multicellular or- ABOUT THE AUTHORS
ganisms. In addition, chemokine overexpression Swathi Balaji, PhD, is a postdoctoral research
has been demonstrated to be associated with fi- fellow in the laboratory for regenerative wound
brosis in multiple organs. CXCL1, 2 and their re- healing at CCHMC specializing in mechanisms
ceptors CXCR1, 2, respectively, have been shown to underlying the fetal regenerative wound healing.
be increased in burn wounds and keloids, which Carey L. Watson, MD, is a University of Cincin-
may contribute to the excessive scarring seen in nati General Surgery Resident who is completing a
these conditions. Overexpression of CXCL8 and two-year postdoctoral research fellowship at Cin-
CCL3 in the lavage fluid of lungs is characteristic of cinnati Children’s Hospital Medical Center
progressive idiopathic pulmonary fibrosis in pa- (CCHMC). Rajeev Ranjan, PhD, is a postdoctoral
tients.87 CXCL10 and CXCL9 have been shown to research fellow in the laboratory for regenerative
be upregulated in bleomycin-induced lung fibrosis. wound healing at CCHMC. Alice King, MD, is a
CXCL1, 5, and 8 are overexpressed in fibrotic University of Cincinnati General Surgery Resident
pancreatitis.88 Targeted manipulation of the che- who has completed a two-year postdoctoral re-
mokine activity can be a novel therapeutic strategy search fellowship in the laboratory for regenerative
to reduce the fibrosis in these diseases. In this wound healing at CCHMC. Paul L. Bollyky, MD,
context, understanding the differences in the che- DPhil, is an immunologist and infectious disease
mokine profile in fetal versus adult wounds can specialist at Stanford University whose laboratory
take us one step closer to using the fetal regener- has a special interest in the relationship of in-
ative wound healing as a roadmap to recapitulate flammation and the extracellular matrix in wound
scarless healing in postnatal wounds and, in fact, healing. Sundeep G. Keswani, MD, is a pediatric
in any process characterized by excessive fibrosis. and fetal surgeon at CCHMC. He is the Principal
Investigator of the laboratory for regenerative
ACKNOWLEDGMENTS wound healing at CCHMC focused on elucidating
AND FUNDING SOURCES the mechanisms underlying the fetal regenerative
This work is supported by the NIH/NIGMS phenomenon and is also the director of Pediatric
awards K08 GM098831-03 and 1R01GM111808-01 Wound Care Center at CCHMC.

REFERENCES
1. Leung A, Crombleholme TM, Keswani SG. Fetal 4. Sund B. New Developments in Wound Care. transformed Chinese hamster and human cells.
wound healing: implications for minimal scar London, United Kingdom: PJB Publications, Proc Natl Acad Sci U S A 1987;84:7188–7192.
formation. Curr Opin Pediat 2012;24:371–378. 2000:1–255.
7. Deuel TF, Keim PS, Farmer M, Heinrikson RL.
2. Martins-Green M, Petreaca M, Wang L. Chemo- 5. Brown BC, McKenna SP, Siddhi K, McGrouther Amino acid sequence of human platelet factor 4.
kines and their receptors are key players in the DA, Bayat A. The hidden cost of skin scars: quality Proc Natl Acad Sci U S A 1977;74:2256–2258.
orchestra that regulates wound healing. Adv of life after skin scarring. J Plast Reconstr Aesthet
8. Sugano S, Stoeckle MY, Hanafusa H. Transfor-
Wound Care 2013;2:327–347. Surg 2008;61:1049–1058.
mation by Rous sarcoma virus induces a novel
3. Bayat A, McGrouther DA, Ferguson MW. Skin 6. Anisowicz A, Bardwell L, Sager R. Constitutive gene with homology to a mitogenic platelet pro-
scarring. BMJ 2003;326:88–92. overexpression of a growth-regulated gene in tein. Cell 1987;49:321–328.
CHEMOKINES IN FETAL AND ADULT WOUND HEALING 671

9. Fivenson DP, Faria DT, Nickoloff BJ, et al. Che- 25. Petreaca ML, Yao M, Liu Y, Defea K, Martins-Green skin wound healing: expression of monocyte
mokine and inflammatory cytokine changes during M. Transactivation of vascular endothelial growth chemoattractant protein-1 is correlated with re-
chronic wound healing. Wound Repair Regen factor receptor-2 by interleukin-8 (IL-8/CXCL8) is cruitment of mast cells which synthesize inter-
1997;5:310–322. required for IL-8/CXCL8-induced endothelial per- leukin-4 in vivo. J Pathol 2000;190:100–106.
meability. Mol Biol Cell 2007;18:5014–5023.
10. Vinader V, Afarinkia K. A beginner’s guide to 41. Chen L, Schrementi ME, Ranzer MJ, Wilgus TA,
chemokines. Future Med Chem 2012;4:845–852. 26. Schraufstatter IU, Chung J, Burger M. IL-8 acti- DiPietro LA. Blockade of mast cell activation re-
vates endothelial cell CXCR1 and CXCR2 through duces cutaneous scar formation. PLoS One
11. Deshmane SL, Kremlev S, Amini S, Sawaya BE.
Rho and Rac signaling pathways. Am J Physiol 2014;9:e85226.
Monocyte chemoattractant protein-1 (MCP-1): an
Lung Cell Mol Physiol 2001;280:L1094–L1103.
overview. J Interferon Cytokine Res 2009;29:313– 42. Wulff BC, Parent AE, Meleski MA, DiPietro LA,
326. 27. Patel DD, Koopmann W, Imai T, Whichard LP, Schrementi ME, Wilgus TA. Mast cells contribute
Yoshie O, Krangel MS. Chemokines have diverse to scar formation during fetal wound healing. J
12. Lin F, Nguyen CM, Wang SJ, Saadi W, Gross SP,
abilities to form solid phase gradients. Clin Im- Invest Dermatol 2012;132:458–465.
Jeon NL. Effective neutrophil chemotaxis is
munol 2001;99:43–52.
strongly influenced by mean IL-8 concentration. 43. Fischer M, Harvima IT, Carvalho RF, et al. Mast
Biochem Biophys Res Commun 2004;319:576–581. 28. Proudfoot AE, Handel TM, Johnson Z, et al. Gly- cell CD30 ligand is upregulated in cutaneous in-
cosaminoglycan binding and oligomerization are flammation and mediates degranulation-indepen-
13. Ebisuno Y, Tanaka T, Kanemitsu N, et al. Cutting
essential for the in vivo activity of certain che- dent chemokine secretion. J Clin Invest
edge: the B cell chemokine CXC chemokine ligand
mokines. Proc Natl Acad Sci U S A 2003;100: 2006;116:2748–2756.
13/B lymphocyte chemoattractant is expressed in
1885–1890.
the high endothelial venules of lymph nodes and 44. Harvima IT, Nilsson G. Mast cells as regulators of
Peyer’s patches and affects B cell trafficking 29. Satish L, Kathju S. Cellular and molecular char- skin inflammation and immunity. Acta Derm Ve-
across high endothelial venules. J Immunol acteristics of scarless versus fibrotic wound nereol 2011;91:644–650.
2003;171:1642–1646. healing. Dermatol Res Pract 2010;2010:790234.
45. Ishida Y, Gao JL, Murphy PM. Chemokine receptor
14. Mehrad B, Keane MP, Strieter RM. Chemokines 30. Lo DD, Zimmermann AS, Nauta A, Longaker MT, CX3CR1 mediates skin wound healing by pro-
as mediators of angiogenesis. Thromb Haemost Lorenz HP. Scarless fetal skin wound healing moting macrophage and fibroblast accumulation
2007;97:755–762. update. Birth Defects Res 2012;96:237–247. and function. J Immunol 2008;180:569–579.
15. Kunkel EJ, Butcher EC. Chemokines and the tis- 31. Naik-Mathuria B, Gay AN, Zhu X, Yu L, Cass DL, 46. Dang CM, Beanes SR, Lee H, Zhang X, Soo C, Ting
sue-specific migration of lymphocytes. Immunity Olutoye OO. Age-dependent recruitment of neu- K. Scarless fetal wounds are associated with an
2002;16:1–4. trophils by fetal endothelial cells: implications in increased matrix metalloproteinase-to-tissue-de-
scarless wound healing. J Pediatr Surg rived inhibitor of metalloproteinase ratio. Plast
16. Allen SJ, Crown SE, Handel TM. Chemokine: re-
2007;42:166–171. Reconstr Surg 2003;111:2273–2285.
ceptor structure, interactions, and antagonism.
Annu Rev Immunol 2007;25:787–820. 32. Olutoye OO, Zhu X, Cass DL, Smith CW. Neu- 47. Bullard KM, Cass DL, Banda MJ, Adzick NS.
trophil recruitment by fetal porcine endothelial Transforming growth factor beta-1 decreases in-
17. Hjelmstrom P, Fjell J, Nakagawa T, Sacca R, Cuff
cells: implications in scarless fetal wound healing. terstitial collagenase in healing human fetal skin.
CA, Ruddle NH. Lymphoid tissue homing chemo-
Pediatr Res 2005;58:1290–1294. J Pediatr Surg 1997;32:1023–1027.
kines are expressed in chronic inflammation. Am J
Pathol 2000;156:1133–1138. 33. Liechty KW, Adzick NS, Crombleholme TM. Di- 48. Van Lint P, Libert C. Chemokine and cytokine
minished interleukin 6 (IL-6) production during processing by matrix metalloproteinases and its
18. Olutoye OO, Yager DR, Cohen IK, Diegelmann RF.
scarless human fetal wound repair. Cytokine effect on leukocyte migration and inflammation. J
Lower cytokine release by fetal porcine platelets: a
2000;12:671–676. Leukoc Biol 2007;82:1375–1381.
possible explanation for reduced inflammation after
fetal wounding. J Pediatr Surg 1996;31:91–95. 34. Hurst SM, Wilkinson TS, McLoughlin RM, et al. Il-6 49. Wulff BC, Yu L, Parent AE, Wilgus TA. Novel
and its soluble receptor orchestrate a temporal switch differences in the expression of inflammation-as-
19. Olutoye OO, Barone EJ, Yager DR, Uchida T, Co-
in the pattern of leukocyte recruitment seen during sociated genes between mid- and late-gestational
hen IK, Diegelmann RF. Hyaluronic acid inhibits
acute inflammation. Immunity 2001;14:705–714. dermal fibroblasts. Wound Repair Regen 2013;21:
fetal platelet function: implications in scarless
103–112.
healing. J Pediatr Surg 1997;32:1037–1040. 35. Ryan GB, Majno G. Acute inflammation. A review.
Am J Pathol 1977;86:183–276. 50. Gharaee-Kermani M, Denholm EM, Phan SH.
20. Roubelakis MG, Trohatou O, Roubelakis A, et al.
Costimulation of fibroblast collagen and trans-
Platelet-rich plasma (PRP) promotes fetal mesen- 36. Henderson RB, Hobbs JA, Mathies M, Hogg N.
forming growth factor beta1 gene expression by
chymal stem/stromal cell migration and wound Rapid recruitment of inflammatory monocytes is
monocyte chemoattractant protein-1 via specific
healing process. Stem Cell Rev 2014;10:417–428. independent of neutrophil migration. Blood
receptors. J Biol Chem 1996;271:17779–17784.
2003;102:328–335.
21. Rolfe KJ, Grobbelaar AO. A review of fetal scar-
51. Yamamoto T, Eckes B, Mauch C, Hartmann K, Krieg
less healing. ISRN Dermatol 2012;2012:698034. 37. Kaesler S, Regenbogen J, Durka S, Goppelt A,
T. Monocyte chemoattractant protein-1 enhances
Werner S. The healing skin wound: a novel site of
22. Engelhardt E, Toksoy A, Goebeler M, Debus S, gene expression and synthesis of matrix metallo-
action of the chemokine C10. Cytokine
Brocker EB, Gillitzer R. Chemokines IL-8, GROal- proteinase-1 in human fibroblasts by an autocrine
2002;17:157–163.
pha, MCP-1, IP-10, and Mig are sequentially and IL-1 alpha loop. J Immunol 2000;164:6174–6179.
differentially expressed during phase-specific in- 38. Cowin AJ, Brosnan MP, Holmes TM, Ferguson
52. Feugate JE, Wong L, Li QJ, Martins-Green M. The
filtration of leukocyte subsets in human wound MW. Endogenous inflammatory response to der-
CXC chemokine cCAF stimulates precocious de-
healing. Am J Pathol 1998;153:1849–1860. mal wound healing in the fetal and adult mouse.
position of ECM molecules by wound fibroblasts,
Dev Dyn 1998;212:385–393.
23. Gillitzer R, Goebeler M. Chemokines in cutaneous accelerating development of granulation tissue.
wound healing. J Leukoc Biol 2001;69:513–521. 39. Juremalm M, Nilsson G. Chemokine receptor ex- BMC Cell Biol 2002;3:13.
pression by mast cells. Chem Immunol Allergy
24. Conus S, Perozzo R, Reinheckel T, et al. Caspase-8 53. Feugate JE, Li Q, Wong L, Martins-Green M. The
2005;87:130–144.
is activated by cathepsin D initiating neutrophil cxc chemokine cCAF stimulates differentiation of
apoptosis during the resolution of inflammation. J 40. Trautmann A, Toksoy A, Engelhardt E, Brocker EB, fibroblasts into myofibroblasts and accelerates
Exp Med 2008;205:685–698. Gillitzer R. Mast cell involvement in normal human wound closure. J Cell Biol 2002;156:161–172.
672 BALAJI ET AL.

54. Liechty KW, Crombleholme TM, Cass DL, Martin 68. Ito M, Liu Y, Yang Z, et al. Stem cells in the hair 81. Bride J, Viennet C, Lucarz-Bietry A, Humbert P. In-
B, Adzick NS. Diminished interleukin-8 (IL-8) pro- follicle bulge contribute to wound repair but not dication of fibroblast apoptosis during the matura-
duction in the fetal wound healing response. J to homeostasis of the epidermis. Nat Med tion of disc-shaped mechanically stressed collagen
Surg Res 1998;77:80–84. 2005;11:1351–1354. lattices. Arch Dermatol Res 2004;295:312–317.
55. Hu MS, Januszyk M, Hong WX, et al. Gene ex- 69. Blanpain C, Fuchs E. Epidermal homeostasis: a 82. Carlson MA, Longaker MT, Thompson JS. Wound
pression in fetal murine keratinocytes and fibro- balancing act of stem cells in the skin. Nat Rev splinting regulates granulation tissue survival. J
blasts. J Surg Res 2014;190:344–357. Mol Cell Biol 2009;10:207–217. Surg Res 2003;110:304–309.
56. Zaja-Milatovic S, Richmond A. CXC chemokines 70. Sasaki M, Abe R, Fujita Y, Ando S, Inokuma D, 83. Kendal-Wright CE. Stretching, mechanotransduc-
and their receptors: a case for a significant bio- and Shimizu H: Mesenchymal stem cells are re- tion, and proinflammatory cytokines in the fetal
logical role in cutaneous wound healing. Histol cruited into wounded skin and contribute to membranes. Reprod Sci 2007;14:35–41.
Histopathol 2008;23:1399–1407. wound repair by transdifferentiation into multiple
84. Okada M, Matsumori A, Ono K, et al. Cyclic stretch
skin cell type. J Immunol 2008;180:2581.
57. Nanney LB, Mueller SG, Bueno R, Peiper SC, upregulates production of interleukin-8 and monocyte
Richmond A. Distributions of melanoma growth 71. Ceradini DJ, Kulkarni AR, Callaghan MJ, et al. chemotactic and activating factor/monocyte che-
stimulatory activity of growth-regulated gene and Progenitor cell trafficking is regulated by hypoxic moattractant protein-1 in human endothelial cells.
the interleukin-8 receptor B in human wound re- gradients through HIF-1 induction of SDF-1. Nat Arterioscler Thromb Vasc Biol 1998;18:894–901.
pair. Am J Pathol 1995;147:1248–1260. Med 2004;10:858–864.
85. Mascarenhas MM, Day RM, Ochoa CD, et al. Low
58. Devalaraja RM, Nanney LB, Du J, et al. Delayed 72. Kitaori T, Ito H, Schwarz EM, et al. Stromal cell- molecular weight hyaluronan from stretched lung
wound healing in CXCR2 knockout mice. J Invest derived factor 1/CXCR4 signaling is critical for the enhances interleukin-8 expression. Am J Respir
Dermatol 2000;115:234–244. recruitment of mesenchymal stem cells to the Cell Mol Biol 2004;30:51–60.
fracture site during skeletal repair in a mouse
59. Rennekampff HO, Hansbrough JF, Kiessig V, Dore 86. Gurtner GC, Dauskardt RH, Wong VW, et al. Im-
model. Arthritis Rheum 2009;60:813–823.
C, Sticherling M, Schroder JM. Bioactive inter- proving cutaneous scar formation by controlling
leukin-8 is expressed in wounds and enhances 73. Ito H. Chemokines in mesenchymal stem cell the mechanical environment: large animal and
wound healing. J Surg Res 2000;93:41–54. therapy for bone repair: a novel concept of re- phase I studies. Ann Surg 2011;254:217–225.
cruiting mesenchymal stem cells and the pos-
60. Rennekampff HO, Hansbrough JF, Woods V, Jr., 87. Antoniou KM, Tzanakis N, Tzortzaki EG, et al. Dif-
sible cell sources. Mod Rheumatol 2011;21:
Dore C, Kiessig V, Schroder JM. Role of mela- ferent angiogenic CXC chemokine levels in
113–121.
noma growth stimulatory activity (MGSA/gro) on bronchoalveolar lavage fluid after interferon gam-
keratinocyte function in wound healing. Arch 74. Abbott JD, Huang Y, Liu D, Hickey R, Krause DS, ma-1b therapy in idiopathic pulmonary fibrosis pa-
Dermatol Res 1997;289:204–212. Giordano FJ. Stromal cell-derived factor-1alpha tients. Pulm Pharmacol Ther 2008;21:840–844.
plays a critical role in stem cell recruitment to the
61. Romer J, Bugge TH, Pyke C, et al. Impaired wound 88. Saurer L, Reber P, Schaffner T, et al. Differential
heart after myocardial infarction but is not suffi-
healing in mice with a disrupted plasminogen expression of chemokines in normal pancreas and
cient to induce homing in the absence of injury.
gene. Nat Med 1996;2:287–292. in chronic pancreatitis. Gastroenterology 2000;118:
Circulation 2004;110:3300–3305.
356–367.
62. Satish L, Blair HC, Glading A, Wells A. Interferon-
75. Kollar K, Cook MM, Atkinson K, Brooke G. Mo-
inducible protein 9 (CXCL11)-induced cell motility
lecular mechanisms involved in mesenchymal
in keratinocytes requires calcium flux-dependent Abbreviations and Acronyms
stem cell migration to the site of acute myocardial
activation of mu-calpain. Mol Cell Biol 2005;25:
infarction. Int J Cell Biol 2009;2009:904682. COX ¼ cyclooxygenase
1922–1941.
E15 ¼ embryonic day 15
63. Inokuma D, Abe R, Fujita Y, et al. CTACK/CCL27 76. Backly RM, Cancedda R. Bone marrow stem cells
ECM ¼ extracellular matrix
in clinical application: harnessing paracrine roles
accelerates skin regeneration via accumulation of EPC ¼ endothelial progenitor cell
bone marrow-derived keratinocytes. Stem Cells and niche mechanisms. Adv Biochem Eng/Bio-
Gro-a ¼ growth-related oncogene alpha
2006;24:2810–2816. technol 2010;123:265–292.
HA ¼ hyaluronan
77. Zhou SB, Wang J, Chiang CA, Sheng LL, Li QF. IL ¼ interleukin
64. Zuliani T, Saiagh S, Knol AC, Esbelin J, Dreno B.
Mechanical stretch upregulates SDF-1alpha in LMW ¼ low molecular weight
Fetal fibroblasts and keratinocytes with immuno-
skin tissue and induces migration of circulating MCP ¼ monocyte chemoattractant protein
suppressive properties for allogeneic cell-based
bone marrow-derived stem cells into the ex- MIP-1a or b ¼ macrophage inflammatory
wound therapy. PLoS One 2013;8:e70408.
panded skin. Stem Cells 2013;31:2703–2713. protein-1 (MIP-1a ¼ CCL3;
65. Carmeliet P. Angiogenesis in health and disease. MIP-1b ¼ CCL4)
Nat Med 2003;9:653–660. 78. Ishida Y, Kimura A, Kuninaka Y, et al. Pivotal role MMP ¼ matrix metalloproteinase
of the CCL5/CCR5 interaction for recruitment of MSC ¼ mesenchymal stem cell
66. Goebeler M, Yoshimura T, Toksoy A, Ritter U, endothelial progenitor cells in mouse wound RANTES ¼ regulated on activation, normal
Brocker EB, Gillitzer R. The chemokine repertoire healing. J Clin Invest 2012;122:711–721. T expressed and secreted
of human dermal microvascular endothelial cells
79. Klein JD, Turner CG, Steigman SA, et al. Amniotic (CCL5)
and its regulation by inflammatory cytokines. J
mesenchymal stem cells enhance normal fetal SDF-1a ¼ stromal cell-derived factor-1 alpha
Invest Dermatol 1997;108:445–451.
wound healing. Stem Cells Dev 2011;20:969–976. TGFb ¼ transforming growth factor beta
67. DiPietro LA, Burdick M, Low QE, Kunkel SL, Stri- TIMP ¼ tissue inhibitors of
eter RM. MIP-1alpha as a critical macrophage 80. Desmouliere A, Chaponnier C, Gabbiani G. Tissue metalloproteinases
chemoattractant in murine wound repair. J Clin repair, contraction, and the myofibroblast. Wound VEGF ¼ vascular endothelial growth factor
Invest 1998;101:1693–1698. Repair Regen 2005;13:7–12.

You might also like