Sie sind auf Seite 1von 38

15  

Skin wound healing: Repair biology, wound,


and scar treatment
Ursula Mirastschijski, Andreas Jokuszies, and Peter M. Vogt

processes in the wound-healing scenario only arise


SYNOPSIS
when wound repair is disturbed.
 Fast and uneventful wound repair guarantees the integrity of Fast and uneventful wound closure restoring the
our body and is essential for survival of all living organisms.
integrity of the outer envelope is a criterion for survival
Overlapping wound healing phases reflect adult repair resulting in a
scar in contrast to scar-free fetal skin regeneration. for all living organisms. From prehistoric times up until
 Wound healing disorders extend between two extremes: chronic today’s highly developed medicine, the aims of wound
nonhealing ulcers on one side and excessive healing with repair have not changed: restoring of the outer surface
hypertrophic scarring on the other. to protect the individual from infection and dehy­
 Multifactorial causes lead to and sustain chronic wounds, while
dration. Egyptian papyri document the treatment of
excessive scarring is found predominantly after deep dermal burns.
In both cases, biomolecular processes are still not well delineated wounds and recommend the use of different dressings
and focus of intensive research. depending on wound quality.1 The use of honey as an
 Clinical wound management starts with an assessment of wound antiseptic has an astonishing history dating back more
type for choosing the appropriate dressing and ends with an than 4000 years and has been rediscovered in recent
interdisciplinary treatment of the wound and accompanying years for the treatment of chronic wounds.2 Furthermore,
co-morbidities.
 Long-term scar treatment is characterized by physiotherapy,
different resins, myrrh, frankincense, and cinnamon
pressure garments and surgical contracture release. were used for wound treatment. South American tribes
 Innovative therapies for aberrant wound repair are still insufficiently applied antiseptic resins such as copaiba, Tolu or
evaluated and mainly experimental. Clinical phase III studies are Peru balm on wounds.3 The oldest wound suture was
lacking for proof of efficacy and wide-spread clinical use. detected on an Egyptian mummy dating from the 21st
dynasty, around 1100 BC.1
Four thousand years later, fast and uneventful repair
is still the maxim of skin wound healing, albeit aesthetic
Wound repair biology issues around tiny and invisible scars play an increasing
role. Acute wounds that heal within 3 months are dis-
A human being has approximately 3000 skin injuries tinguished from chronic nonhealing wounds. The term
during a lifetime. Small wounds usually heal unevent- “acute” comprises burn wounds, traumatic lesions, or
fully. Questions about normal and pathological surgical incisional wounds.

©
2013, Elsevier Inc. All rights reserved.
268 • 15 • Skin wound healing: Repair biology, wound, and scar treatment

Major Clot formation Growth factor Collagen deposition Collagen cross-linking


event Haemostasis elaboration
Inflammatory
Repair
Proliferation
phase
Remodeling
Fibroblasts
Cellular Lymphocytes
influx Macrophages
Neutrophils
Vascular
Vasodilation
response
Vasoconstriction Fig. 15.1  The temporal patterns of repair phase, cellular
influx, and vascular response during wound repair. The time
Injury 3d 7d 3 weeks 1-2 years points are approximate, and overlap occurs during these
Time repair events.7

Adult wound repair Epidermis

The repair cascade consists of inflammatory, prolifera-


tive, and remodeling phases (Fig. 15.1). These overlap-
ping phases act in highly coordinated relationships to Blood vessel
heal skin defects. During the inflammatory phase,
hemostasis occurs and an acute inflammatory infiltrate Exposure Dermis
of thromboplastic
ensues. The proliferative phase is characterized by fibro- tissue elements
plasia, granulation, contraction, and epithelialization.
The final phase is remodeling, which is commonly Resident
described as scar maturation. moncyte

Platelet

Inflammatory phase Fibroblast


SQ fat
Red blood cell
Inflammation is the first stage of wound healing and
comprises cellular and vascular responses, including
hemostasis. At the injury site, lacerated vessels immedi-
Fig. 15.2  Hemostasis is stimulated by platelet degranulation and exposure of
ately constrict. Thromboplastic tissue products, pre- tissue thromboplastic agents immediately after tissue injury.7 SQ, subcutaneous.
dominantly from the subendothelium, are exposed.
Platelets adhere, aggregate, and form the initial hemo-
static plug.
The coagulation and complement cascades are initi-
ated. The intrinsic and extrinsic coagulation pathways surface.4,5 Interestingly, under thrombocytopenic condi-
lead to activation of prothrombin to thrombin, which tions, macrophages and T cells at the wound site com-
converts fibrinogen to fibrin, subsequently polymerized pensated for lack of platelet-derived growth factors
into a stable clot. As thrombus is formed, hemostasis in (PDGFs) and initiation of the inflammatory phase.6
the wound is achieved (Fig. 15.2). The aggregated plate- Immediately, the repair processes are initiated. After
lets degranulate, releasing potent chemoattractants for hemostasis, local vessels dilate secondary to the effects
inflammatory cells, activation factors for local fibrob- of the coagulation and complement cascades. Bradykinin
lasts and endothelial cells, and vasoconstrictors. The is a potent vasodilator and vascular permeability factor
adhesiveness of platelets is mediated by activated that is generated by activation of Hageman factor
integrin receptors such as GpIIb-IIIa (αIIbβ3) on their in the coagulation cascade.8 The complement cascade
Adult wound repair 269

growth factors. These growth factors activate and attract


local endothelial cells, fibroblasts, and keratinocytes to
begin their respective repair functions. More than 20
Keratinocytes
different cytokines and growth factors are known to be
secreted by macrophages (Table 15.1).11
Neutrophil
Depletion of monocytes and macrophages in combi-
nation with steroid therapy resulted in severe wound-
healing deficiency with poor debridement, delayed
fibroblast proliferation, and inadequate angiogenesis.12
In contradiction to the dogma that inflammation is
Fibrin matrix essential for skin wound healing, mice deficient in the
hematopoietic ETS family transcription factor PU.1
Fibroblast
showed normal wound repair13 despite immunoincom-
petent leukocytes and nonfunctional macrophages.14
Although the growth factor and cytokine pattern was
altered, incisional wounds healed uneventfully and
scar-free with phagocytic fibroblasts clearing the wound
debris. Obviously, attenuated inflammation is beneficial
for wound healing.9
Fig. 15.3  A neutrophil influx into the wound occurs within 24 hours. The
neutrophils scavenge debris and bacteria and secrete cytokines for monocyte and
lymphocyte attraction and activation. Keratinocytes begin migration when a
provisional matrix is present.7
Proliferation phase

Extracellular matrix formation


generates the C3a and C5a anaphylatoxins, which During fibroplasia, fibroblasts synthesize and deposit
directly increase blood vessel permeability and attract replacement extracellular matrix (ECM) at the wound
neutrophils and monocytes to the wound. These com- site. The proliferative phase begins with degradation of
plement components also stimulate the release of hista- the initial fibrin–platelet provisional matrix. Three major
mine and leukotrienes C4 and D4 from mast cells. The classes of proteases are involved in wound repair.
local endothelial cells then break cell–cell contact and Proteases of the serine, cysteine, and matrix metallopro-
increase permeability, which enhances the margination teinase (MMP) family are secreted to facilitate cellular
of inflammatory cells into the wound site.8 migration through fibrin clot and provisional matrix.
The initial influx of white blood cells in the wound Both tissue and urokinase plasminogen activator are
is composed of neutrophils (Fig. 15.3). This early neu- expressed to dissolve the blood clot.15 MMPs comprise
trophil infiltrate scavenges cellular debris, foreign endopeptidases such as collagenases, gelatinases, and
bodies, and bacteria. Activated complement fragments stomelysins that can degrade virtually all matrix pro-
aid in bacterial killing through opsonization. The neu- teins, activate growth factors, and bind on cell sur-
trophil infiltrate is decreased in clean surgical wounds faces.16,17 Cysteine proteases are present in epithelia
compared with contaminated or infected wounds. during wound repair and are involved in signaling and
Within 2–3 days, the inflammatory cell population basement membrane protein degradation.18
begins to shift to one of monocyte predominance. Macrophages, mast cells, and the adjacent ECM
Circulating monocytes and mast cells are attracted and release growth factors that stimulate fibroblast activa-
infiltrate the wound site.9,10 These monocytes differenti- tion.9 Local fibroblasts become activated and increase
ate into macrophages and, in conjunction with the resi- protein synthesis in preparation for cell division. As
dent tissue macrophages, orchestrate the repair process. fibroblasts proliferate, they become the predominant
Macrophages not only continue to phagocytose tissue cell type by 3–5 days in clean, noninfected wounds (Fig.
and bacterial debris but also secrete multiple peptide 15.4). After cell division and proliferation, fibroblasts
270 • 15 • Skin wound healing: Repair biology, wound, and scar treatment

Table 15.1  A partial list of growth factors present at the wound site*
Growth factor Cellular source Target cells Biologic activity

Activin Fibroblasts, keratinocytes Stromal cells Granulation tissue formation,


scarring
TGF-β1 and TGF-β2 Macrophages, platelets, fibroblasts, Inflammatory cells, keratinocytes, Chemotaxis, proliferation, matrix
keratinocytes fibroblasts production (fibrosis)
TGF-β3 Macrophages Fibroblasts Antiscarring?
TGF-α Macrophages, platelets, keratinocytes Keratinocytes, fibroblasts, Proliferation
endothelial cells
TNF-α Neutrophils, mast cells Macrophages, keratinocytes, Activation of growth factor
fibroblasts expression
PDGF Macrophages, platelets, keratinocytes, Neutrophils, macrophages, Chemotaxis, proliferation, matrix
fibroblasts, endothelial cells, fibroblasts, endothelial cells, production
vascular smooth-muscle cells vascular smooth-muscle cells
FGF-1, FGF-2, FGF-4 Macrophages, fibroblasts, endothelial Keratinocytes, fibroblasts, Angiogenesis, proliferation,
cells endothelial cells, chondrocytes chemotaxis
FGF-7 (KGF-1), FGF-10 Fibroblasts Keratinocytes Proliferation, chemotaxis
(KGF-2)
EGF Platelets, macrophages, keratinocytes Keratinocytes, fibroblasts, Proliferation, chemotaxis
endothelial cells
HB-EGF Macrophages, keratinocytes Keratinocytes, fibroblasts Proliferation, epithelial migration,
synergistic with IGF
IGF-1/Sm-C Fibroblasts, macrophages, platelets Fibroblasts, endothelial cells Proliferation, collagen synthesis
IL-1α and IL-1β Macrophages, neutrophils Macrophages, fibroblasts, Proliferation, collagenase
keratinocytes synthesis, chemotaxis
CTGF/CCN2 Fibroblasts, endothelial cells Fibroblasts Downstream of TGF-β1
VEGF Macrophages, keratinocytes, Endothelial cells Angiogenesis
fibroblasts
*Redundant biologic effects occur through both autocrine and paracrine mechanisms.
TGF-α, transforming growth factor-α; TGF-β, transforming growth factor-β; TNF-α, tumor necrosis factor-α; PDGF, platelet-derived growth factor; FGF, fibroblast growth fac-
tor; KGF, keratinocyte growth factor; EGF, epidermal growth factor; HB-EGF, heparin-binding EGF, IGF-1, insulin-like growth factor 1; Sm-C, somatostatin C; IL-1, interleukin-1;
CTGF, connective tissue growth factor; VEGF, vascular endothelial cell growth factor.
(Reproduced from Lorenz HP, Longaker MT. Wounds: biology, pathology, and management. New York: Springer-Verlag; 2000.)

begin the synthesis and secretion of ECM products. The phosphorylation occurs in the cytoplasmic domain of
initial fibrin matrix is replaced by a provisional matrix the integrin receptor, which starts a signal transduction
of fibronectin and hyaluronan, which facilitates fibro­ cascade that ultimately changes gene expression, and
blast migration. The control of ECM deposition by new cellular function ensues.
fibroblasts is complex and partially regulated by growth Fibronectin and the glycosaminoglycan hyaluronic
factors and interactions of fibroblast cell membrane acid compose the initial wound matrix.10 Hyaluronic
receptors with the ECM. Integrins are regulators of cel- acid provides a matrix that enhances cell migration
lular function during repair. They are transmembrane because of its large water content. Adhesion glycopro-
receptors with extracellular, membrane, and intracellu- teins, including fibronectin, laminin, and tenascin, are
lar protein domains. Integrins are heterodimeric and present throughout the early matrix and facilitate cell
composed of α and β subunits that interact to form the attachment and migration. Integrin receptors on
active protein receptor. Ligands to integrins include cell surfaces bind to the matrix glycosaminoglycans
growth factors and ECM structural components such as and glycoproteins. As fibroblasts enter and populate
collagen, elastin, and other cells.19,20 After ligands bind, the wound, they secrete hyaluronidase to digest
Adult wound repair 271

Scab
through a complex intracellular and extracellular
process. Coordinated transcription of genes on different
chromosomes (2, 6, 7, 12, 13, 17, and 21) occurs. In addi-
Keratinocyte
tion, several intracellular and extracellular modifica-
migration
tions are required to form the new collagen fiber.
Collagens contain a high fraction of proline amino acid
residues, and this is the basis of its triple helix structure.
With regard to collagens I and III, tropocollagen is
Myofibroblast
formed in the ECM.24 Tropocollagen molecules aggre-
Fibroblast gate laterally and are covalently cross-linked by the
Neutrophil enzyme lysyl oxidase to form collagen fibrils.25 The
Macrophage
fibrils interact with other fibril types, which then aggre-
gate into fibers. Fibers then aggregate into bundles and
form a collagen-rich scar.

Granulation tissue formation and angiogenesis


Granulation tissue is a dense population of blood
vessels, macrophages, and fibroblasts embedded within
Fig. 15.4  Fibroblasts are activated and present at the wound by 3–5 days after
injury. These cells secrete matrix components and growth factors that continue to
a loose provisional matrix of fibronectin, hyaluronic
stimulate healing. Keratinocytes migrate over the new matrix. Migration starts from acid, and collagen. Granulation tissue is clinically char-
the wound edges as well as from epidermal cell nests at sweat glands and hair acterized by its beefy-red appearance (i.e., “proud
follicles in the center of the wound.7
flesh”) and is present in open wounds. It is a conse-
quence of the rich bed of new capillary networks (neoan-
the provisional hyaluronic acid-rich matrix, and larger, giogenesis) that form by endothelial cell division and
sulfated glycosaminoglycans are subsequently depos- migration. The directed growth of vascular endothelial
ited. Concomitantly, new collagen is deposited by cells is stimulated by tissue hypoxia, and platelet
fibroblasts on to the fibronectin and glycosaminoglycan and activated macrophage and fibroblast products.26
scaffold in a disorganized manner, resulting in scar for- Examples are basic fibroblast and vascular endothelial
mation. The major fibrillar collagens composing the growth factor (VEGF), which induce migration and pro-
ECM in skin and scar are collagen types I and III. The liferation of endothelial cells.27 A prerequisite for
ratio of collagen type I to type III is 4 : 1 in both skin and endothelial sprouting is the expression of the integrin
wound scar. Although type III collagen is initially avβ3 at the leading capillary tip28 and the secretion of
deposited in relatively greater amounts in wounds, proteases for matrix degradation. Granulation tissue is
its amount is always less than type I collagen in a clinical indicator that an open wound is amenable to
mature scar.21 skin graft treatment. Wounds that benefit from skin
At least 28 different types of collagen are currently grafts are of sufficient size such that the healing time
known.22 Most collagen types of the ECM are synthe- would be decreased after grafting. Because granulation
sized by fibroblasts; however, some types are synthe- tissue has a high level of vascularity due to the abun-
sized by keratinocytes.23 The collagens share common dance of new capillary formation, it readily accepts and
characteristics: the basic structural unit is a right-handed supports skin grafts.
triple helix. Unique structural properties that distin-
guish the different collagen types include segments that
Wound contraction
interrupt the triple helix and fold it into other structures
with unique properties.24 Contraction is the process in which the surrounding
The major structural component of wound scar is skin is pulled circumferentially toward an open wound
collagen. Fibroblasts synthesize and secrete collagens (Fig. 15.5). This phenomenon does not occur with closed
272 • 15 • Skin wound healing: Repair biology, wound, and scar treatment

A B C D E

Fig. 15.5  Wound contraction is the process in which the surrounding tissue is pulled radially toward the wound. The wound size is decreased, which shortens the healing
time. Full-thickness, rat skin wounds on postoperative days 0 (A), 4 (B), 8 (C), 12 (D), and 14 (E) after wounding. Note completely healed wound on postoperative day 14.

surgical incisions. Open wounds after trauma, burns, forces by focal adhesion contacts that link the intracel-
and previously closed wounds secondarily opened by lular cytoskeleton to the ECM. Interestingly, intercellu-
infection are associated with contraction. Wound con- lar myofibroblast contacts via connexin-containing
traction decreases the size of the wound dramatically gap junctions are important for contractility because
without new tissue formation. This repair process connexin-43-depleted fibroblasts were incapable to
speeds wound closure compared with epithelialization contract collagen matrices.32 Furthermore, the role of
and scar formation alone. In addition, the area of insen- keratinocytes in wound contraction should not be
sate scar is smaller. Animals have a much greater ca­­ neglected. In vitro experiments showed higher contrac-
pacity for wound contraction than humans do. Most tile forces of keratinocytes compared with fibroblasts.33
mammalian animals (e.g., rodents, cats, dogs, sheep, Future research will shed further light on the biology of
and rabbits) have a panniculus carnosus, which is a skin wound contraction.
myofascial layer between the subcutaneous fat and Wound contraction must be distinguished from con-
musculoskeletal layers. This anatomy results in a plane tracture. Clinically, contracture is defined as tissue
of low resistance between two fascial layers, which shortening or distortion that causes decreased joint
allows enhanced skin mobility and therefore contrac- mobility and function. Scar contracture commonly
tion. The amount of contraction is related to both the refers to decreased function in the area, whereas scar
size of the wound and mobility of the skin. In humans, contraction refers to shortening of the scar length com-
contraction is greatest in the trunk and perineum, least pared with the original wound.
on the extremities, and intermediate on the head and
neck. Eighty percent to 90% of wound closure can be
Epithelial resurfacing
due to contraction in the trunk and perineum.29 These
regional differences in contraction are probably due to Morphologic changes in keratinocytes at the wound
relative differences in skin laxity. margin are evident within hours after injury. Marginal
It has become accepted that myofibroblasts play a basal cells dissolve intercellular contacts, e.g., desmo-
key role in wound contraction. After wounding, dermal somes and adherence junctions, and cell–matrix con-
fibroblasts at wound margins are activated by growth tacts, e.g., hemidesmosomes. Flattened migratory cells
factors released into the wound. Stimulated by mechan- express cytoplasmatic actin filaments, promigratory
ical tension and PDGF, they turn into stress fiber- integrins, e.g., α2β1, α5β1, αvβ5, αvβ6, and secrete pro-
expressing protomyofibroblasts. Protomyofibroblasts teases to enable movement.34 MMP-3 is localized to
are found in early granulation tissue and in normal wound margin keratinocytes and cleaves the intercel-
connective tissue with high mechanical load. lular contact protein E-cadherin.35 MMP-9 cleaves base-
Approximately 4 days after wounding, myofibroblasts ment proteins, e.g., collagen type IV and VII releasing
appear in the wound.30 Mechanical tension, activated keratinocytes from their substratum.36 MMP-1 degrades
transforming growth factor-β (TGF-β1)16 and the splice native collagen type I and III, smoothing the way for
variant ED-A fibro­­nectin trigger protofibroblast differ- newly formed matrix. Furthermore, MMP-1 binds to
entiation into alpha-smooth-muscle actin-expressing α2β1 integrin on cell membranes, presumably acting as
myofibroblasts.31 Myofibroblasts exert their contractile a migration promoter.37 Blocking MMP activity with
Regulation 273

synthetic broad-spectrum inhibitors abolished epithelial fill the wound site. The ultimate pattern of collagen in
migration in vitro and in vivo.38,39 scar is one of densely packed fibers and not the reticular
In contrast to keratinocytes are fibroblasts able to pattern found in unwounded dermis.
adopt an ameboid phenotype and squeeze through the Wound scar remodeling occurs during months to
matrix network in the presence of MMP inhibitors.40 years to form a “mature” scar. The early scar appearance
Recently, cysteine proteases, e.g., cathepsin-B, C, K and is red due to its dense capillary network induced at the
L, entered the field of skin wound healing, acting as injury site. When closure is complete, capillaries regress
endopeptidases involved in the degradation of ECM until relatively few remain. As the scar redness dissipates
proteins.18 during a period of months, the true scar pigmentation
After the re-establishment of the epithelial layer, becomes evident. Scars are usually hypopigmented after
keratinocytes and fibroblasts secrete laminin and type full maturation. However, scars can become hyperpig-
IV collagen to form the basement membrane.41 The mented in darker-pigmented patients and in those
keratinocytes become columnar and divide to restore lighter-pigmented patients whose scars receive excess
the layering of the epidermis and reform a barrier to sun exposure. For this reason, sun protection measures
further contamination and moisture loss. Keratinocytes are recommended for patients with early scars on sun-
can respond to foreign-body stimulation with migration exposed areas such as the scalp, face, and neck. During
as well. Sutures in skin wounds provide tracks along remodeling, wounds gradually become stronger with
which these cells can migrate. Fibrotic reactions, cysts, time. Wound tensile strength increases rapidly from 1 to
and sterile abscesses centered on the suture can occur. 8 weeks after wounding and correlates with collagen
These are treated by removal of the inciting suture and cross-linking by lysyl oxidase. However, the tensile
epithelial cell sinus track or cyst. strength of wounded skin at best reaches only appro­
ximately 80% that of unwounded skin45 but can be
Remodeling phase increased by synthetic MMP inhibitors.46 In addition,
scar is brittle and less elastic than normal skin. It is
The scaffold that supports cells in both the unwounded readily visible because of color, contour, and texture dif-
and wounded states is the ECM, which is the structural ferences compared with unwounded skin. Although
component of skin that must be repaired after injury. scars can be hidden well with proper surgical planning
The ECM is dynamic and is constantly undergoing and uneventful healing, they may have aesthetically unac-
remodeling during repair, which can be conceptualized ceptable appearances in nonelective wounds after trauma
as the balance between synthesis, deposition, and deg- and burns and in wounds with healing problems.
radation. Lysyl oxidase is the major intermolecular col-
lagen cross-linking enzyme.25 Collagen cross-linking
decreases its degradation and improves wound tensile Regulation
strength. The balance of collagen deposition and degra-
dation is in part determined by the regulation of MMP Growth factors in skin wound healing
activity.42 MMPs are inactivated by tissue inhibitors of
MMPs, α-macroglobulin, and degradation by other pro- Growth factors are the focal regulatory points of the
teases.17,43 Except for reduced wound contraction in repair process. They are polypeptides that are released
MMP-3-deficient mice,44 other MMP knockout mice by a variety of activated cells at the wound site (Table
have little or no wound-healing disturbances.36 15.1). They act in either a paracrine or autocrine fashion
Scar formation is the ultimate outcome of wound to stimulate or to inhibit gene expression by their target
repair in children and adults. The scar has no epidermal cells in the wound. In general, they stimulate cellular
appendages (hair follicles and sebaceous glands), and it proliferation and chemoattract new cells to the wound.
has a collagen pattern that is distinctly different from Myriad growth factors are present in wounds and many
unwounded skin. New collagen fibers secreted by have overlapping biologic functions. Most growth
fibroblasts are present as early as 3 days after wound- factors exist in several isoforms with several receptor
ing. As the collagen matrix forms, densely packed fibers types present in wounds, which increases the
274 • 15 • Skin wound healing: Repair biology, wound, and scar treatment

complexity of growth factor function. In addition to myofibroblast differentiation, and wound contraction.
growth factor ligands, their signaling receptors are TGF-β accelerates wound repair when it is applied
another locus for regulation of repair. Growth factors do experimentally to wounds that have no deficiency in
not have an effect on target cells without a functional repair. However, the increase in repair rate is at the
signaling receptor present on the cell surface. The level expense of increased fibrosis, which could be a detri-
of regulation is complex in that the growth factors have ment during normal skin healing. In addition, increased
multiple different receptor types to which they can bind TGF-β activity is associated with pathologic fibrosis in
and induce cell signaling. In recent years, the develop- multiple different organ systems, including heart, lung,
ment of transgenic and knockout mouse models includ- brain, liver, and kidney. TGF-β stimulates ECM synthe-
ing inducible and cre-lox technologies has generated sis by increasing collagen, elastin, and glycosamino­
new knowledge in the function of many growth factors glycan synthesis. It increases integrin expression, which
in wound healing. Clinical trials applying various enhances cell–matrix interactions. TGF-β increases
growth factors to chronic nonhealing wounds showed ECM accumulation by decreasing MMP and increasing
beneficial effects,47 although treatment efficacy should tissue inhibitor of MMP expression. Through these
be confirmed by larger, randomized, double-blinded mechanisms, exogenous TGF-β augments fibrosis at
studies.48 the wound site.51 Interestingly, studies in mice with
a lack of endogenous TGF-β activity demonstrate
accelerated healing with an impaired inflammatory
Platelet-derived growth factor
response.52 In chronic wounds, however, significantly
PDGF is released from platelet alpha granules immedi- reduced TGF-β was found leading to clinical trials with
ately after injury. PDGF attracts neutrophils, macro- TGF-β treatment that failed showing beneficial effects.53
phages, and fibroblasts to the wound and serves as a Activins and BMP are promigratory proteins secreted
powerful mitogen. Macrophages, endothelial cells, and by keratinocytes and fibroblasts during wound repair
fibroblasts also synthesize and secrete PDGF in the and promote keratinocyte differentiation.53 These find-
wound. PDGF stimulates fibroblasts to synthesize new ings underscore the complex effects of TGF-β and other
ECM, predominantly the noncollagenous components growth factors during the repair process.
such as glycosaminoglycans and adhesion proteins and
to contract these matrices.49 PDGF also increases the
Fibroblast growth factors
amount of collagenase secreted by fibroblasts, indicat-
ing a role for this growth factor in tissue remodeling. The fibroblast growth factors (FGFs) are a group of
PDGF strongly induces granulation tissue production heparin-binding growth factors that are secreted into
and was the first growth factor to be approved for the the ECM, where they remain dormant until activated by
treatment of diabetic nonhealing wounds.50 tissue injury. They are bound by heparin and the gly-
cosaminoglycan heparan sulfate. They have a broad
range of biologic functions, specific to each isoform.
Transforming growth factor-β FGF-1 (acidic FGF) and FGF-2 (basic FGF) stimulate
The TGF-β family comprises TGF-β1–3, bone morpho- angiogenesis.26 Endothelial cells, fibroblasts, and macro-
genic proteins (BMP), and activins. TGF-β1 predomi- phages produce FGF-1 and FGF-2. Basement membrane
nates in adult wound healing and is a promigratory serves as a storage depot for FGF-2, which is released
and profibrotic growth factor that directly stimulates on degradation of the heparin components of the base-
collagen synthesis and decreases ECM degradation by ment membrane at sites of injury. FGF-1 and FGF-2
fibroblasts. It is released from all cells at the wound stimulate endothelial cells to divide and form new capil-
site, including platelets, macrophages, fibroblasts, laries. They also chemoattract endothelial cells and
and keratinocytes. TGF-β acts in an autocrine fashion fibroblasts. FGF-7 (keratinocyte growth factor 1 (KGF-
to stimulate its own synthesis and secretion. TGF-β 1)), FGF-10 (KGF-2), and epidermal growth factor
also chemoattracts fibroblasts and macrophages to (EGF) stimulate epithelialization. KGF-1 and KGF-2
the wound, promotes granulation tissue formation, are expressed in wound fibroblasts and promote
Regulation 275

keratinocyte proliferation and migration in a paracrine the wound site and contribute to tissue remodeling and
fashion. Decreased expression of KGF-1 occurs in dia- angiogenesis.51 IL-6 is crucial for initiation of the wound-
betic and reduced levels of FGF-2 in aged mouse wounds healing response68 whereas overexpression leads to
with decreased angiogenic response.54,55 In an ischemic cutaneous scarring. TNF-α and interferon-γ have
rabbit wound model, exogenous KGF-2 treatment accel- been shown to downregulate ECM protein synthesis.
erates epithelialization without an increase in scar for- Granulocyte–macrophage colony-stimulating factor
mation.32 Exogenous KGF-2 also increases wound tensile (GM-CSF) is a pleiotropic cytokine stimulating migra-
strength, collagen content, and epidermal thickness in tion and proliferation during wound repair. Experimental
animal models of normal healing.56 Hitherto, only one treatment of diabetic and chronic wounds showed
clinical trial has been conducted using recombinant promising results with faster healing rates.69,70
human KGF-2 for treatment of chronic venous ulcers Although further studies are needed to determine the
and showed accelerated wound healing.57 precise growth factor combination that is optimal for
specific wound types, therapeutic growth factor appli-
cation has become a reality. The clinical use of PDGF-BB
Vascular endothelial growth factor
was approved by the Food and Drug Administration for
VEGF is also a potent angiogenic stimulus.58 It acts in a chronic ulcers. Further promising candidates are VEGF/
paracrine manner to stimulate vascular permeability PLGF, FGF-2, and GM-CSF.
and proliferation by endothelial cells after release from
platelets, endothelial cells, neutrophils, macrophages,
fibroblasts, and keratinocytes.59–61 Its expression is
Growth factor interactions with
increased in hypoxic conditions, such as those found at
the extracellular matrix
the wound site.62 Intramuscular gene transfer of VEGF
to patients with ischemic ulcers resulted in sprouting In normal, unwounded conditions, the ECM is a deposi-
of collateral vessels and limb salvage.63 Despite these tory of growth factors in latent forms. With injury and
promising results, careful use of VEGF is recommended matrix destruction, growth factors are released from the
because exogenous VEGF administration yielded vas- ECM in active form and thereby assist in initiating and
cular leakage and malformation.64 Placental growth regulating the repair process. For example, TGF-β is
factor (PLGF) is also proangiogenic and acts synergisti- bound in the ECM to the proteoglycan decorin and is
cally to VEGF.65 PLGF is upregulated in migrating kerat- inactive when bound. At sites of injury, TGF-β forms
inocytes and promotes granulation tissue formation and complexes with its binding protein, latency-associated
migration. Unlike VEGF, PLGF does not interfere with protein (LAP), and is released. Under acidic conditions,
lymphatic vessel formation66 and accelerates wound such as at sites of hypoxia and tissue injury, LAP disas-
healing in diabetic mice.67 sociates and active TGF-β is formed. LAP can also be
proteolytically cleaved and released by MMPs and other
proteases at the wound site. Active TGF-β immediately
Other growth factors and cytokines
binds to its receptors (RI and RII), which are present on
Multiple other growth factors affect wound repair. fibroblasts, macrophages, and endothelial cells.71 The
Epithelialization is also directly stimulated by members TGF-β RI and RII receptors form heterodimeric com-
of the EGF family, e.g., EGF, heparin-binding EGF (HB- plexes with each other, and TGF-β biologic activity is
EGF), and TGF-α. These mitogens are released by kerat- initiated in the target cell through the Smad pathway.72
inocytes to act in an autocrine fashion. Insulin-like The FGFs are another example of growth factors
growth factor 1 (IGF-1) stimulates cell proliferation and bound by the ECM. PDGF and HB-EGF have also been
collagen synthesis by fibroblasts and interacts synergis- shown to bind to ECM proteins. Through sequestration
tically with HB-EGF, PDGF, and FGF-2 to facilitate of growth factors with their subsequent release during
fibroblast proliferation.51 Various cytokines comprising injury, the ECM plays a fundamental role in wound
interleukins (IL), chemokines, and tumor necrosis repair by presenting growth factors that regulate the
factor-α (TNF-α) mediate inflammatory cell functions at repair processes.
276 • 15 • Skin wound healing: Repair biology, wound, and scar treatment

intrauterine environment, and fetal serum factors are


Fetal wound repair biology not required for scarless healing. Scarless repair appears
to be inherent to the fetal tissue and probably depends
The therapeutic solution for the reduction and possible on factors associated with skin development.75 Notably,
elimination of scar formation may be found in the mech- fetal scarless repair in skin is organ-specific with scar-
anisms responsible for scarless wound healing in the ring of intestinal wounds at the same gestational age.77
fetus. The early-gestation fetus can heal skin wounds The fetal environment alone cannot induce scarless
with regenerative-type repair and without scar forma- healing in adult skin. When adult skin is transplanted
tion.74,75 In scarless fetal wounds, the epidermis and on to a fetus and later wounded at a point in gestation
dermis are restored to a normal architecture. The col- when fetal skin heals scarlessly, the adult skin grafts still
lagen dermal matrix pattern is reticular and unchanged heal with scar formation.78 The transformation of adult
from unwounded dermis. The wound hair follicle and healing to scarless repair cannot be accomplished simply
sweat gland patterns are normal as well (Fig. 15.6) . The by perfusion of adult skin with fetal serum or by immer-
wound is not evident grossly unless a wound edge sion in amniotic fluid. Thus, induction of scarless healing
contour change is present, and this will cast a light in adult skin will require more than recreation of the
shadow under appropriate circumstances.73 Scarless fetal environment. The adult repair processes will likely
healing by the fetus does not depend on the fetal envi- have to be modified to recapitulate skin development.
ronment. Fetal wounds in marsupials76 or skin graft An understanding of the biology of scarless fetal
wounds heal without scar after transplantation to a wound repair will help surgeons develop therapeutic
postnatal environment. Thus, amniotic fluid, the strategies to minimize scar and fibrosis.

A B

C D

Fig. 15.6  Fetal rat skin wound made at gestational age 16.5 days (term = 21days), stained with hematoxylin and eosin. The hair follicle pattern is normal and there is
no dermal collagen scar formation. Black arrows indicate India ink tattoo made at the time of wounding to locate the scarless wound. (A,C) Healed wounds harvested at
72 hours (×100). The epidermal appendage (developing hair follicles) pattern shows numerous appendages in the healed wound. (B,D) Magnified views of the same
wounds show epidermal appendages (open arrows) within the wound site (×200). No inflammatory infiltrate is present. (Modified from Beanes SR, Hu FY, Soo C, et al.
Confocal microscopic analysis of scarless repair in the fetal rat: defining the transition. Plast Reconstr Surg 2002; 109:160–170.)
Fetal wound repair biology 277

Transition from scar-free healing transition wound has a normal reticular collagen and
to scar formation connective tissue matrix pattern but without restoration
of epidermal appendages.73 Thus, it has features of
Both gestational age and wound size determine whether both scar (no appendages) and scarless (normal ECM)
the fetus will heal a wound without scar.79 As gestation healing.
progresses, a transition from scarless healing to healing
with scar formation occurs before birth. In large-animal
models, such as the fetal lamb and monkey (Fig. 15.7), Differences between fetal and adult repair
this transition occurs during the early part of the third
trimester for incisional, closed wounds. In addition, Cellular differences
wound size affects the temporal occurrence of this tran-
Because fetal fibroblasts deposit matrix in a scar-free
sition. Open, excisional wounds must be made earlier
pattern, they are crucial for scarless repair. A number of
in gestation than closed, incisional wounds for scarless
studies have defined the functional differences between
healing to occur. Also, larger open wounds must be
fetal and postnatal dermal fibroblasts. First, fetal fibrob-
made earlier in gestation than smaller open wounds for
lasts synthesize more collagen type III and IV with
scarless healing.79 The exact reasons underlying these
simultaneous proliferation, in contrast to their postnatal
observations remain unknown but probably relate to
counterparts in vitro.80 Fetal fibroblasts failed to respond
whether the wound has healed before a certain thresh-
to TGF-β stimulation with collagen synthesis compared
old in development. The shift from scarless healing
to postnatal fibroblasts in vitro.81 Furthermore, higher
to scar formation is not abrupt but instead occurs
gene expression of MMP-1, -2, -3, -9, and -14 is found
gradually with an intermediate repair outcome that is
during fetal wound healing with virtually absent myofi-
neither regeneration nor scar: the transition wound. The
broblasts during early gestation.82,83 Fetal fibro­blasts
also migrate at a faster rate than postnatal fibro­blasts.84
Increased migration velocity during repair is likely to
affect collagen deposition and crosslinking.
In postnatal wounds, epidermal continuity is restored
by proliferating stem cells derived from the epidermal
basement membrane and the hair follicle bulge.85
Because immature fetal skin does not have hair bulges,
the stem cell origin is likely to be different to adult
wound repair. Indeed, a new stem cell was recently
discovered and named dot cell because of its extremely
small size.86 Dot cells are found in basement membranes
and in the blood during fetal development and have a
strong affinity to wounded tissue. After transplantation
of dot cells into adult murine wounds, scarless healing
was noted. Despite their small size, these cells seem to
have a great future in wound repair.

Differences in wound repair


Several parameters of repair are different in fetal wounds
Fig. 15.7  Fetal monkey full-thickness wedge excision lip wound made at the compared with postnatal wounds. The rate of repair for
beginning of the third trimester and harvested 2 weeks later. The cutaneous wound wounds of equal size is faster in the fetus. The collagen
is identified by the sutures. No scar was present histologically, and the orbicularis
oris muscle reformed across the defect.73 Interestingly, the naris was deformed synthesis rate is greater in fetal wounds. In addition, the
compared with the contralateral side. rate of epithelialization is faster in the fetal wound.80
278 • 15 • Skin wound healing: Repair biology, wound, and scar treatment

Fetal wound repair may be more rapid because there is wounds in a similar fashion, and this results in scar
little or no period of activation for the fetal fibroblast to formation.
synthesize ECM at the wound site.
A key difference between fetal and postnatal tissue
Differences in gene expression
repair is a reduced inflammatory cellular infiltrate in
fetal wound repair.87 Inflammation plays a prominent Because fetal skin is growing and differentiating, genes
role in postnatal repair, but it is not present in signifi- associated with development are likely to have an
cant amounts during scarless fetal healing. This may important role during scarless healing. Their expression
be due to the immature fetal immune system with may not occur during adult healing because of inactiva-
decreased platelet degranulation, PDGF and TGF-β tion at the end of development. Homeobox genes are
production.87 However a marked inflammatory cellular transcription factors that govern gene expression during
infiltrate occurs later in gestation after the transition embryogenesis. PRX2 is a homeobox gene that has
when fetal wounds form scar. The amount of inflam- increased expression during scarless fetal wound
mation correlates strongly with the amount of scar healing and only weak expression during adult skin
formation. healing.93 New data are available for Wnt proteins
involved in skin maturation, fetal and adult wound
repair.94,95 Wnt signaling is increased in postnatal but not
Growth factor expression in fetal wound repair in fetal murine wound healing with a TGF-β1-like profi-
brotic influence on postnatal but not on fetal fibrob-
Few growth factors associated with adult repair have lasts.96 Differential signaling in fetal and adult cells in
increased expression in scarless fetal wounds. This is response to wounding seems to direct the way to either
likely due to inherent major differences in the regulation tissue regeneration or scarring.
between the two repair systems. Growth factors that
are upregulated in scarless wounds are increased more
rapidly than in postnatal wounds. One example is
Differences in matrix composition
VEGF, which is expressed rapidly and threefold higher Fetal skin and wound matrix are composed of more
after injury in scarless wounds but is delayed in post- hyaluronic acid than are postnatal skin and scar.
natal wounds.88 The expression of TGF-β isoforms, Hyaluronic acid stimulates fibroblast migration and
receptors, and bioactivity modulators has been ana- probably affects the ECM deposition pattern.84 Elastin,
lyzed in fetal wounds. Compared with scarring wounds, present in adult skin, is absent in fetal dermis, although
scarless fetal wounds have more TGF-β3, receptor type no differences were found in basement membrane pro-
II, and fibromodulin expression.89 In contrast, increased teins or vessel formation.97 Fetal skin and wound ECM
TGF-β1, receptor type I, and decorin expression is asso- have a relatively greater amount of collagen type III,
ciated with the transition to scar formation. Thus, the but how this affects scar formation is not known. The
expression profile of isoforms, receptors, and modula- collagen fiber thickness is increased in scarring fetal
tors for a growth factor can be different in scarless and wounds compared with unwounded fetal skin as meas-
scarring wounds. By understanding the scarless repair ured by confocal microscopy. This distance between
expression profile for a growth factor, information is collagen fibers is also greater in scarring fetal wounds,
obtained that can permit recapitulation of this profile but the mechanisms causing these earliest changes in
during postnatal repair to reduce scar formation. healing associated with scar formation remain to be
Exogenous addition of several cytokines to fetal wounds elucidated.74 Scarless fetal skin repair is the blueprint
has also been performed and in every case has resulted for ideal repair. With regard to TGF-β3, an antiscarring
in accelerated scarring. The cytokines tested include product was developed98 but failed to prove efficacy
TGF-β1, TGF-β2, PDGF, and BMP-2, each of which has in clinical phase III trials. However, the mechanisms
increased expression during scar formation in postnatal responsible for scarless repair still remain to be fully
wounds.90–92 Thus, growth factors with increased expres- determined and when delineated will likely lead to
sion in scarring wounds probably modulate scarless innovative antiscarring treatment strategies.
Adult wound pathology 279

rotation and an air mattress or another type of low-


Adult wound pathology pressure bed. Wheelchair seat cushions commonly need
to be changed in patients with ischial pressure sore.
Nonhealing wounds Behavior, such as prolonged sitting in wheelchairs
Chronic or nonhealing wounds are open wounds that without equal weight distribution, must frequently
fail to epithelialize and close in a reasonable amount of be modified. Lower extremity contractures that cause
time, usually defined as 30 days. These wounds typi- excessive hip, knee, and ankle pressure may need
cally are clinically stagnant and unable to form robust release. Tight casts should be removed and replaced.
granulation tissue. Many factors contribute to inhibit Improperly fitting lower extremity prostheses can lead
healing in these patients, but no unifying theory can to stump wounds and need modification. Other contrib-
explain the etiopathogenesis of each individual non- uting factors, such as malnutrition, infection, and dia-
healing wound (Table 15.2). Medical conditions such as betes mellitus, should be identified and treated. Most
diabetes, arterial insufficiency, venous disease, lymph- pressure sores will heal with avoidance of pressure over
edema, steroid use, connective tissue disease, and radia- the involved area. However, they heal with scar, which
tion injury inhibit wound healing. Nonhealing wounds is less resistant to trauma than is normal skin. Thus, a
can also be due to pressure necrosis, infection (espe- higher incidence of recurrence exists after spontaneous
cially osteomyelitis), skin cancer, malnutrition, chronic closure of these wounds than if they are closed surgi-
dermatologic disease, and other metabolic conditions. cally with flaps of normal skin and muscle over the
In each case, treatment begins with debridement of any bony prominence.102,103
necrotic tissue present.99 However, despite optimal
treatment for each clinical problem, these wounds fre- Lower extremity wounds
quently still do not heal and surgical intervention is Leg wounds generally arise from either one of two dif-
required. ferent vascular diseases: arterial or venous insufficiency.
Most (80–90%) result from venous valvular disease
Pressure sores (venous insufficiency).104 The role of impaired lymphatic
drainage is discussed controversially in being a direct
Wounds that develop over a bone prominence, usually
cause of nonhealing wounds or aggravating venous
in the immobile patient, are termed pressure sores. They
insufficiency.104a
are also called decubitus ulcers or bed sores. The sacrum,
ischium, and greater trochanter are the most common Venous
locations affected.100 However, the metatarsal heads,
ankles, heels, knees, and occiput are susceptible under Increased venous pressure in the dependent lower
certain conditions. Another problem is malleolar skin extremity with valvular incompetence leads to localized
pressure necrosis due to constricting cast placement. edema with plasma extravasation. Fibrinogen leakage
The amount of tissue pressure necrosis is determined by results in formation of a fibrin layer around the capil-
both the degree and duration of the pressure. When the laries that impairs oxygen and nutrient diffusion.105
tissue pressure is greater than 25–30 mmHg and capil- Leukocytes may be trapped and activated in obstructed
lary perfusion pressure is blocked, microcirculation is capillaries. Oxygen radicals and proteases may then be
compromised. Necrosis can occur with as little as 2 released, which causes tissue necrosis. Postcapillary
hours of sustained pressure at this level.101 Skin is more obstruction leads to increased perfusion pressure and
resistant to pressure necrosis than are underlying fat hypoxia, with further necrosis. Without treatment, the
and muscle, which explains the common finding of a wound size can easily continue to grow.
small area of skin ulceration overlying a large cavitary
Arterial
volume of subcutaneous fat and muscle necrosis.
Treatment begins with identification and control of the Wounds require adequate oxygen delivery to heal.
factors that lead to increased pressure and subsequent Ischemic wounds heal poorly and have a much greater
wound formation. Paralyzed patients require periodic risk of infection.106 Minor trauma, resulting from
280 • 15 • Skin wound healing: Repair biology, wound, and scar treatment

Table 15.2  Factors that may impair normal healing and lead to chronic nonhealing wounds

Etiopathology Examples

Vascular Arterial Arteriosclerosis, arterial aneurysm, fat embolism with arterial obstruction, hypertension (Martorell
ulcer)
Lymphatic Lymphatic edema, lymphangiodysplasia
Venous Chronic venous insufficiency, necrotizing thrombophlebitis
Mixed Combined arteriosclerosis with venous insufficiency, arteriovenous malformations/dysplasia;
arteriovenous steal phenomenon (e.g., arteriovenous shunts, vascular compression/obstruction (due to
tumors, enlarged lymphatic nodes, etc.)
Vasculitis Wegener granulomatosis, Churg–Strauss vasculitis, Henoch–Schönlein purpura, Sneddon
syndrome, systemic lupus erythematosus, rheumatoid arthritis, Felty syndrome, Takayasu
arteriitis, polyarteritis nodosa, Kawasaki syndrome, pyoderma gangrenosum, necrobiosis
lipoidica diabeticorum, thrombangiitis obliterans (Buerger syndrome), allergic reactions
Vasculopathic Raynaud syndrome, systemic scleroderma, CREST, Klippel–Trenaunay syndrome, proteus
syndromes syndrome145,146, CLOVE syndrome,147 Kasabach–Merritt syndrome
Physical, chemical, Pressure Immobility, intra- and postoperative bedding, tight shoes and casts, compression therapy
and biological
causes
Trauma Lacerations, any type of soft-tissue and bone injury, vascular rupture
Thermal Burns / frostbite, electrical injury (electrical current/high voltage/lightning)
Radiation Radiation therapy
Chemical-toxic Extravasation, chemical burns (acids/bases), sclerotherapy
Infections Erysipelas, necrotizing fasciitis, septic cutaneous embolism, osteomyelitis, complications after
cutaneous infection
Herpes simplex, cytomegalovirus, human immunodeficiency virus, syphilis, leprosy, tuberculosis
Tropical ulcers, parasitic and vermicular infections
Neuropathic Posttraumatic Spinal lesions with palsy, peripheral nerve injury
Congenital Spina bifida, syringomyelia, multiple sclerosis, neurological syndromes
Systemic Diabetes mellitus, etyltoxic neuropathy, degenerative central and peripheral neuropathies
neuropathic Poliomyelitis, leprosy, tabes dorsalis
diseases
Hemopathological Systemic diseases Polycythemia vera, sickle-cell anemia, other anemias, thalassemia, thrombocythemia vera,
thrombocytopenic purpura, increased blood viscosity (paraneoplastic, paraproteinemia,
hyperglobulinemia, leukemia), complication after blood transfusion
Disturbed Factor V Leiden syndrome, antiphospholipid syndrome, disturbed fibrinolysis, factor XIII
hemostasiology deficiency syndrome, antithrombin III deficiency, protein C/S deficiency, Marcumar necrosis,
disseminated intravascular coagulation, necrosis due to vitamin K antagonist therapy
Neoplastic Cutaneous tumors Basal and squamous cell carcinoma, melanoma, Bowen syndrome, Marjolin ulcer (scar
diseases carcinoma),148 tumors with cutaneous metastasis or penetration (e.g., Paget syndrome)
Therapeutic Steroids, vaccination ulcer (BCG), cytostatic drugs, NSAIDs, extravasation of various drugs
modalities
Systemic diseases Hepatic and/or renal insufficiency, immunosuppression, sarcoidosis, homocysteinemia,
hemochromatosis
Other causes Alcoholism, obesity, gout, smoking, advanced age, malnutrition (e.g., vitamin, protein, and
micronutrient deficiency, scurvy); psychiatric diseases with self-harming, neglect, intravenous
drug abuse; foreign bodies/ projectiles with fistulas
CREST, calcinosis, Raynaud’s syndrome; esophageal dysmotility, sclerodactyly, and telangiectasia; CLOVE, congenital lipomatous overgrowth, vascular malformations, and
epidermal nevi; BCG, bacillus Calmette-Guérin; NSAIDs, nonsteroidal anti-inflammatory drugs.
Adult wound pathology 281

scratches and abrasions that would otherwise heal which suggests that many of these impairment mecha-
quickly in a well-vascularized extremity, can progress nisms are partially reversible in diabetic wounds.
to large wounds in ischemic limbs. Necrotizing infec-
tion that is not only limb- but also life-threatening can Radiation injury
develop. A reliable clinical sign of adequate arterial
inflow for healing is the presence of an arterial pulse. External-beam radiation through skin to treat deeper
The ankle brachial pressure index (ABPI) represents a disease has both acute and chronic effects on skin.
simple noninvasive method to detect arterial insuffi- Acutely, a self-limited erythema may develop that spon-
ciency within a limb. ABPI is calculated by dividing the taneously resolves. The late effect of radiation is a more
systolic blood pressure measured at the level of the significant injury to fibroblasts, keratinocytes, and
ankle by the systolic blood pressure measured in the endothelial cells. DNA damage to these cells propagates
brachial artery.107 Normal ranges of ABPI were defined and impairs the ability of these cells to divide success-
as being 0.91–1.3, whereas ratios of ≤0.4 imply clinically fully. Irradiated tissue usually has some degree of resid-
critical limb ischemia.108 Transcutaneous oxygen meas- ual endothelial cell injury and progressive endarteritis,
urement is a noninvasive method with 83% accuracy in which results in atrophy, fibrosis, and poor tissue
predictability of wound closure and 68% in predicting repair.122 Ultimately, the affected skin may spontane-
failure in an ischemic extremity.109,110 To standardize ously break down, but usually after repeated mild
transcutaneous oxygen measurements, tissue hypoxia trauma.122 When a surgical incision is placed through
was defined as <40 mmHg in healthy patients and irradiated skin on the trunk or extremities, it is not likely
<30 mmHg in patients with limb ischemia110 with likely to heal. Although improvement in chronic wound repair
wound-healing disturbances at rates lower than can be achieved with hyperbaric oxygen therapy,123 sur-
20 mmHg.99,111 A nonhealing wound in an ischemic gical intervention with resection of the wound to normal,
extremity is an indication for revascularization.112 nonirradiated tissue and coverage with a vascularized
flap are frequently necessary.

Diabetic
Infection
Wound healing is impaired in diabetic patients by
several mechanisms. Neuropathy is frequently present, Wound infection is an imbalance between host resist-
which leads to decreased sensation and biomechanical ance and bacterial growth.124 Bacterial infection impairs
joint instability. Arterial insufficiency is present in 30– healing through several mechanisms. At the wound site,
60% of cases. Studies have implicated an altered expres- acute and chronic inflammatory infiltrates slow fibro­
sion profile of growth factors in diabetic wounds.113 blast proliferation and thus slow ECM synthesis and
Decreased expression of GM-CSF receptor, PDGFs and deposition. Although the exact mechanisms are not
their receptors, VEGF and its type II receptor, EGF recep- known, sepsis causes systemic effects that also impede
tor, IGF-1, and nitric oxide synthase-2 has been demon- repair. A threshold number of bacteria in the wound is
strated in human diabetic foot ulcers.114 Sustained necessary to overcome host resistance and cause clinical
inflammation is marked by an increase in proinflamma- wound infection. Bacterial contamination results in clin-
tory cytokines115 and cells, e.g., macrophages, B cells, ical infection and delays healing if more than 105 organ-
and plasma cells, but decreased number of T cells in the isms per gram of tissue are present in the wound.125 Skin
wound margin.116 MMP production is deregulated with grafts on open wounds are likely to fail if quantitative
dislocation of proinflammatory MMP-9 to the ulcer culture shows more than 105 organisms per gram of
bed.117,118 Advanced glycation end-products and inflam- tissue, which provides further evidence that bacterial
matory mediators commit fibroblasts and vascular cells load has an impact on repair.126 Similarly, well-
to apoptosis and impair granulation tissue formation.119 vascularized muscle flaps heal open wounds success-
Diabetic fibroblasts and keratinocytes have reduced fully if bacterial loads are not greater than 105 organisms
proliferation rates and collagen production.120 Wound per gram of tissue.127 These studies demonstrate that
healing is enhanced if glucose levels are well controlled,121 high levels of bacteria inhibit the normal healing
282 • 15 • Skin wound healing: Repair biology, wound, and scar treatment

processes. Treatment of the closed infected wound conclusively in human studies, most surgeons adminis-
depends on whether fluid or necrotic tissue is present. ter vitamin A postoperatively to their patients receiving
If no fluid is draining or accumulating, the cellulitis can steroid therapy. Vitamin A is fat-soluble and can be
be successfully treated with appropriate antibiotics. The taken in toxic doses, so careful administration is essen-
wound should be opened and sutures removed; the tial. The oral dose is 25 000 IU/day.134
wound is irrigated and débrided if pus or necrotic tissue Vitamin B6 (pyridoxine) deficiency impairs collagen
is present. Administration of appropriate antibiotics cross-linking.137 Vitamin B1 (thiamine) and vitamin B2
after wound culture treats surrounding cellulitis. Signs (riboflavin) deficiencies cause syndromes associated
of wound infection include fever, tenderness, erythema, with poor wound repair. Supplementation with these
edema, and drainage. vitamins does not improve healing unless a pre-existing
deficiency is present. Deficiencies of trace metals such
as zinc and copper have been implicated in poor wound
Malnutrition and obesity
repair because these divalent cations are cofactors in
Wound healing is an anabolic event that requires addi- many important enzymatic reactions.134 Zinc deficiency
tional calorie intake.128 However, the precise calorie is associated with poor epithelialization and chronic,
requirements for optimal wound healing have to be cal- nonhealing wounds.138 Trace metal deficiency is now
culated for each patient individually.129 Large injuries extremely rare in both enterally and parenterally fed
such as burns greatly increase metabolic rate and nutri- patients. Excess administration of vitamins and miner-
tional requirements.130 Severely malnourished and als can be detrimental and cause toxic effects, especially
catabolic patients tend to impaired healing131 because by the fat-soluble vitamins. Adequate amounts are
nutrients such as proteins, vitamins, and minerals that present in today’s enteral feeding solutions and as
are important for effective wound repair are not substi- supplemental additives to parenteral solutions.
tuted. These nutrients are involved in collagen synthesis Supplemental administration is necessary only in defi-
and cross-linking and re-epithelialisation and promote ciency states and certain unique clinical situations, as
the inflammatory response in early wound healing. described before.
Wound dehiscence risk is increased in humans with Obesity interferes with repair independently of
hypoproteinemia132 and in protein-depleted rats, but diabetes.139 Obese patients with diabetes have
this can be reversed with protein repletion immediately impaired wound healing independently of glucose
after wounding.133 control and insulin therapy. Poor wound perfusion
Vitamin C (ascorbic acid) deficiency results in and necrotic adipose tissue probably contribute to
scurvy. In these patients, wound healing is arrested impaired healing in both diabetic and nondiabetic
during fibroplasia. Normal quantities of fibroblasts are obese patients.
present in the wound, but they produce an inadequate
amount of collagen.134 Vitamin C is necessary for
Medical treatment
hydroxylation of proline and lysine residues,135 and
without hydroxyproline, newly synthesized collagen Both topically applied steroids and pharmacologic
is not transported out of cells and collagen fibrils are steroid use impair healing, especially during the first 3
not cross-linked. Both mechanisms decrease wound days after wounding.140 Steroids reduce wound inflam-
tensile strength. mation, collagen synthesis, and contraction.134 The exact
Vitamin A (retinoic acid) is involved in multiple facets mechanisms by which steroids impair healing are not
of repair: fibroplasia, collagen synthesis and cross- fully understood. Glucocorticoids decrease PDGF and
linking, and epithelialization.134 Because vitamin A KGF expression in experimental wounds.141,142 Steroids
requirements increase after injury, severely injured stabilize lysosomal membranes and thereby decrease
patients require supplemental vitamin A to maintain the release of lysosomes at the repair site, which may
normal serum levels. Animal studies show that vitamin slow repair processes. Because steroids decrease inflam-
A also reverses the impaired healing that occurs with mation, they may decrease host bacterial resistance and
chronic steroid treatment.136 Although it is not proved thus increase wound infection complications. The entire
Adult wound pathology 283

Table 15.3  Diagnostic criteria for distinction between most institutions, chemotherapeutic agents are not
hypertrophic scars and keloids157,182 administered until at least 5–7 days postoperatively to
Hypertrophic scar Keloid
prevent impairment of the initial healing events.

Clinical Remain within the Grow beyond the wound


symptoms boundaries of the borders Excessive healing
wound
Rarely more than 1 cm Size variations; growth Normal wounds have “stop” signals that halt the repair
in thickness or width may be widespread, process when the dermal defect is closed and epitheli-
vertical, or both
alization is complete. When these signals are absent or
Form scar contractures No contractures
Less pruritic and painful Pruritic and painful ineffective, the repair process may continue unabated
Occur after injury only: Spontaneous and cause excessive scar formation. The underlying
disruption of skin appearance without molecular mechanisms leading to excessive repair are
continuity, burns skin injury possible still a topic of intensive research. Profibrotic cytokine
Generally arise within 4 Appear within several
overexpression and reduction of collagenase activity
weeks, grow months after initial
intensely for several scar, then gradually were found in skin tissue of burn patients.149 In cell
months, then proliferate indefinitely co-cultures, keratinocytes from hypertrophic scar tissue
regress often within or keloids induced increased proliferation and collagen
1 year
production in dermal fibroblasts.150,151 A lack of pro-
Occur on points with Occur often on the
excessive tensile chest, shoulders, grammed cell death, apoptosis, at the conclusion of
forces: across joint trunk, back of the repair with the continued presence of activated fibrob-
surfaces, sternum, neck and earlobes, lasts secreting ECM components has also been impli-
neck, palm, and rarely on the palms cated.152 Notwithstanding the molecular regulation of
soles after injury or soles
excessive scar formation, there are clinical factors that
Regress spontaneously Do not regress
spontaneously affect scar formation. To minimize visible scar on skin,
Histological Fine and thin collagen Large and thick collagen
elective incisions are least noticeable when they are
features fibers oriented fibers; closely placed parallel to the natural lines of skin tension
parallel to the packed collagen (Langer’s lines). This placement has two advantages:
epidermis fibrils; increased ratio the scar is parallel or within a natural skin crease, which
of type I to type III
camouflages the scar, and this location places the least
collagen
Nodules with increased Increased fibroblast amount of tension on the wound. Wound tension widens
fibroblast density density in enlarging the scar. Sharply defined and well-aligned wound edges
borders, acellular in that are approximated without tension heal with the
keloid center least amount of scar. Infection or separation of the
Presence of α-smooth- Lack of α-smooth-
muscle actin- muscle actin-
wound edges with subsequent secondary intention
expressing expressing healing also results in more scar formation. Hyper­
myofibroblasts is myofibroblasts pigmentation and hypo­pigmentation of the scar increase
typical its contrast with the surrounding skin, making the scar
more visible. Sun protection of all wounds is recom-
repair process is slowed, and risk of dehiscence and mended to prevent scar hyperpigmentation.
infection is increased. Vitamin A administration can
reverse this effect.136,143
Hypertrophic scar
Both radiation and chemotherapeutic agents have
their greatest effects on dividing cells. During the pro- Hypertrophic scars and keloids are unique to humans
liferative phase of repair, numerous cell types are and do not occur in animals for unknown reasons. These
dividing at the wound site. Antiproliferative chemo- pathologic scar types are distinguished on the basis of
therapeutic agents act to slow this process and thus their clinical characteristics. Hypertrophic scars are
retard healing.144 After oncologic surgical procedures, in defined as scars that have not overgrown the original
284 • 15 • Skin wound healing: Repair biology, wound, and scar treatment

wound boundaries but are instead raised. They usually stimulation. CTGF stimulates chemotaxis, prolifera-
form secondary to excessive tensile forces across the tion, MMP expression and ECM production.162 The
wound and are most common in wounds across joint proteoglycan decorin binds TGF-β and regulates colla-
surfaces on the extremities but also commonly occur on gen fibrillogenesis by downregulating TGF-β pro­
the sternum and neck. Physical therapy with range- duction.163,164 In contrast to normal dermal fibroblasts,
of-motion exercises is helpful in minimizing hyper- hypertrophic scar-derived fibroblasts secrete less
trophic scar as well as joint contracture in the extremities. decorin and probably contribute thereby to sustained
Hypertrophic scar is a self-limited type of overhealing TGF-β activity.165 A distinct histological difference
that can regress with time. These scars generally fade as between keloids and hypertrophic scars is the predomi-
well as flatten to the surrounding skin level. No clear nance of myofibroblasts in hypertrophic scar tissue.
histologic difference between hypertrophic scar and Controversial findings exist concerning the persistence
keloid has been demonstrated. Because of similar histo- of fibroblasts after accomplished healing. Programmed
logic findings in both hypertrophic scar and keloid,153 cell death is thought to underlie myofibroblast disap-
they are more easily differentiated by their clinical char- pearance after wound closure. Whether fibroblast resist-
acteristics (Table 15.3).154 ance to apoptosis leads to excessive scarring is still
Hypertrophic scars and keloids are both fibroprolif- unknown.157
erative disorders of wound repair with excess healing.155
However, because there are no animal models of either
Keloid
condition, there are few biochemical and molecular
data that distinguish the two entities in direct compari- Keloids are scars that overgrow the original wound
son. In addition, most studies analyze either one or the edges. This clinical characteristic distinguishes keloid
other, but not both. In general, both keloid and hyper- from hypertrophic scar. True keloid scar is not common
trophic scar fibroblasts have an upregulation of colla- and occurs mainly in darkly pigmented individuals
gen synthesis, deposition, and accumulation.154 It may with an incidence of 4.5–16% in African and Asian pop-
be that keloid fibroblasts respond to a greater degree ulations compared to less than 1% in Caucasians.166,167
than do hypertrophic scar fibroblasts to the signals Currently, it is proposed that keloids form in areas of
stimulating scar formation. For example, keloid fibrob- high skin tension,168 although this does not explain why
lasts respond to exogenous TGF-β with a much greater palms or soles are unaffected and why there is a high
increase in collagen production than do hypertrophic occurrence rate on tension-free earlobes. Another
scar fibroblasts.156 hypothesis postulates an immune response to the pilose-
Mast cells and fibrocytes, circulating bone marrow- baceous unit after dermal injury and subsequent
derived progenitor cells that differentiate into fibro­ cytokine release with fibroblast activation. This is sup-
blasts, are proposed to promote tissue fibrosis, ported by the fact that keloids preferentially occur on
especially in burn patients.157,158 Both cell types are anatomical sites with high concentrations of sebaceous
present in hypertrophic scar tissue and secrete profi- glands, such as the chest wall, neck, and pubic area.
brotic cytokines. Mast cell histamine enhances collagen Patients with keloids have a positive skin reaction to
production in fibroblasts159 and is elevated in the sebum antigen and benefit from desensitization with
plasma of patients developing hypertrophic scar.160 The sebum vaccine.169 A genetic predisposition is the point
keratinocyte’s regulatory role in this scenario has of discussion with autosomal-dominant features.170
gained more interest in the past years. By secreting Certain syndromes are associated with keloid forma-
various factors, keratinocytes stimulate migration, pro- tion, e.g., Rubinstein–Taybi syndrome,171 scleroderma,172
liferation, and matrix remodeling in a paracrine and and Touraine–Solente–Golé syndrome.173 Interestingly,
autocrine way.161 Increased collagen production was altered TGF-β regulation of the pro-opiomelanocortin
found in normal fibroblasts co-cultured with hyper- (POMC) gene expression in keloid-derived fibroblasts
trophic scar-derived keratinocytes.151 was found.174 A regulatory role of POMC in keloid for-
Hypertrophic scar fibroblasts produce more con­ mation is conceivable considering the fact that hitherto
nective tissue growth factor (CTGF/CCN2) after TGF-β no human albinos with keloids have been reported.
Clinical wound management 285

The keloid scar continues to enlarge past the original under tension. For this reason, most surgeons do not
wound boundaries and behaves like a benign skin close the subcutaneous fat layer, even in morbidly obese
tumor with continued slow growth. However, complete patients. Dead space here is better obliterated with a
excision with primary closure of the defect results in short course of closed suction drainage postoperatively,
recurrence in the majority of cases. Keloid patients may which can prevent seroma formation and possible infec-
have excessive start signals or lack the appropriate stop tion. The amount of tissue injury and degree of contami-
signals for healing. In the latter instance, the lack of stop nation influence the length and quality of healing. Small,
signals results in continued and unchecked repair. A clean closed wounds heal quickly with less scar forma-
possible mechanism of keloid formation may be the tion, whereas large, open dirty wounds heal slowly with
presence of persistent signals pushing fibroblasts to significant scar. To decrease scar formation and risk of
keep “healing” the wound site despite complete cover- infection, meticulous hemostasis should be performed.
age of the original wound. Studies have looked at apop- This limits the amount of hematoma to be cleared and
tosis in normal scar, hypertrophic scar, and keloid. thus decreases the inflammatory phase and probably
Similar numbers of apoptotic cells at the advancing decreases scar formation. Because hematoma is a culture
wound edge in both normal scar and hypertrophic scar medium for bacteria, less bleeding also decreases the
have been found.175 However, proapoptotic gene expres- risk of infection. By limiting the inflammation with
sion is decreased in keloids, suggesting persistence of sterile technique and tight hemostatic control, repair by
activated fibroblasts.176 activated fibroblasts can begin earlier and shorten the
Keloids consist mainly of collagen and are relatively healing period.
acellular in their central portions with fibroblasts present Smaller surgical scars are achieved with meticulous
along their enlarging borders. They do not contain a approximation of corresponding wound edges, no skin
significant excess number of fibroblasts. Keloid fibrob- edge trauma, and less resultant inflammation. Forceps
lasts respond differently than normal wound fibroblasts crush injury of the epidermis and dermis should be
to growth factors found at the repair site.177 Keloid- avoided by the use of fine forceps and skin hooks to
derived fibroblasts express increased levels of CTGF,178 retract and assist in dermal closure. This decreases the
TGF-β,179 and VEGF180 compared to normal skin fibrob- amount of necrotic tissue at the wound edge and thereby
lasts and show increased proliferation and collagen pro- reduces inflammation. Because suture material is a
duction compared to normal fibroblasts.181 foreign body, it generates an immune response and is
susceptible to infection. Some surgeons therefore close
the epidermis with Steri-Strips. There is no suture to
Clinical wound management leave a “railroad track” scar or serve as an infection
locus in the skin. Fibrin and other biologic glues and
Management is based on the classification of the wound
sealants are also used especially for superficial wounds
type. Surgically closed incisions obviously require a dif-
in infants without need for anesthesia. Depending on
ferent treatment from that of nonhealing open wounds.
the wound localization, different suture techniques and
Hypertrophic scars and keloids with their excessive
materials are recommended (Table 15.4).183
scarring require other different therapeutic approaches.
Closed wounds require less care than open wounds.
The treatment options for various wound types are
Closed wounds should be kept sterile for 24–48 hours
described below.
until epithelialization is complete. At this point, water
Incisional wounds barrier function has been restored and patients can be
allowed to shower or wash. This has a psychological
Incisional wounds closed in layers along tissue planes benefit during the postoperative recovery period. In
heal with primary intention. Deep sutures are placed in addition, gentle cleansing removes old serum and blood,
collagen-rich layers such as fascia and dermis. These which reduces potential bacterial accumulation and
strength layers can hold sutures under a high degree of infection risk. Tensile strength of a closed incisional
tension. Fatty tissue layers, such as subcutaneous fat, do wound is only 20% that of normal skin at 3 weeks when
not have significant collagen and cannot hold sutures collagen cross-linking is becoming significant. At 6
286 • 15 • Skin wound healing: Repair biology, wound, and scar treatment

Table 15.4  Recommended suture materials and techniques for closure of full-thickness skin wounds at various body sites183
Wound localization Suture material Closure technique Suture removal (days)*

Scalp 3-0 or 4-0 nonabsorbable, Simple, narrow stitches 7–12


monofilament
Face 4-0 or 5-0 synthetic absorbable In layers; in case of full-thickness wound: running or 3–5
or 6-0 nonabsorbable, single sutures
monofilament
Eyelid 5-0 or 6-0 nonabsorbable, Single-row stitches or running intracutaneous suture 5
monofilament (secured with Steri-Strip for 10 days)
Lip, oral cavity 4-0 or 5-0 absorbable for In layer: muscle, mucosa, skin; single stitches 3–5
mucosa, muscle, dermis;
6-0 monofilament
nonabsorbable for skin
Neck 4-0 absorbable, 5-0 Double-layered for better cosmetic result; 4–6
nonabsorbable, intracutaneous, running sutures
monofilament
Abdomen, back 3-0 or 4-0 absorbable, 3-0 or Single or multilayered sutures; intracutaneous, 6–12
4-0 nonabsorbable, running sutures
monofilament
Limbs 3-0 or 4-0 absorbable,† 3-0 or Single or multilayered stitches (dermis); running 6–14‡
4-0 nonabsorbable, intracutaneous sutures; splints for joint-crossing
monofilament wounds or wounds with high mechanical tension
Hands and feet 4-0 or 5-0 nonabsorbable, Single layer with simple or Allgöwer / Donati stitches; 7–12
monofil splints for joint-crossing wounds or wounds with
high mechanical tension
Nail bed 6-0 or 7-0 absorbable Meticulous approximation with single stitches Resorption
Use of magnifying glasses is recommended
Nail bed splinting with original or artificial nail to avoid
adhesions

*If nonabsorbable suture material was used.



Absorbable, monofilament sutures (e.g., polydioxanone) are more suitable for wounds with high mechanical tension, providing higher wound-breaking strength during
collagen bundle cross-linking.

Nonabsorbable sutures on legs remain for up to 21 days depending on limb perfusion status and skin quality.

weeks, wounds are at 70% of the tensile strength of closed wounds. The major difference is that each
normal skin, which is nearly the maximal tensile strength sequence is much longer, especially the proliferative
achieved by scar (75–80% of normal). Therefore, if phase. There is much more granulation tissue formation
absorbable suture is used to close deep structures that and contraction. This type of healing process is referred
are under significant tension, such as abdominal fascia, to as secondary intention.
the suture should retain significant tensile strength for Open wound edges are not approximated but are in­­
at least 6 weeks before absorption severely weakens the stead separated, which necessitates epithelial cell migra-
suture. In addition, heavy activity should be limited for tion across a longer distance. Before re-epithelialization
a minimum of 6 weeks when healing of abdominal can occur, a provisional matrix must be present.
fascial layers is necessary. Granulation tissue must form. There are variable
amounts of bacteria, tissue debris, and inflammation
Excisional wounds present, depending on wound location and etiology.
Infection, with high protein exudative losses and acute
Open wounds heal with the same basic processes of and chronic inflammation, can dysregulate repair
inflammation, proliferation, and remodeling as do and transform the healing wound into a clinically
Clinical wound management 287

nonhealing wound. The exact molecular mechanisms podiatrist, internist, endocrinologist, and specialist in
causing this shift from healing to a nonhealing wound microbiology. The team leader, usually a plastic surgeon,
with infection remain unknown. diagnoses the cause of the wound, coordinates appro-
During the proliferative phase of an uncomplicated priate referrals, and directs overall wound care.
course of secondary-intention healing, a bed of granula- Prosthetists, physical therapists, enterostomal thera-
tion tissue will be present. If no infection is present and pists, and clinical nurse specialists complete the team.
the area is of sufficient size that healing will not be Because the etiopathogenesis of chronic wounds is mul-
complete for at least 2–3 weeks, placement of a partial- tifactorial, coordinated care by multiple specialists is
or full-thickness skin graft should be considered. Grafts required for optimal results.
readily adhere to granulation tissue and will quickly To achieve standardized wound management, the
speed the repair process. Partial-thickness skin graft International Wound Bed Preparation Advisory Board
donor sites can heal in as little as 2 weeks, depending and the Canadian Chronic Wound Advisory Board have
on graft thickness. When an open wound heals, which proposed components relevant for wound bed prepara-
is generally defined as complete epithelialization, the tion: tissue debridement, infection/inflammation, and
dermal defect has been filled with collagen that is moisture balance. Even when these factors have been
covered by epithelium. This scar has less tensile strength corrected, some wounds do not heal and have an abrupt
and is more susceptible to trauma than normal skin. or steep epidermal edge, with no migration of epider-
Thus, these scars break down more easily from local mal cells across the wound surface. These four com­
trauma such as pressure. ponents of wound bed preparation can easily be
remembered using the mnemonic TIME – tissue,
infection/inflammation, moisture, and edge effect.99
Wound treatment Wound bed preparation is the management of a
Necrotic material should be removed from open wounds wound in order to accelerate endogenous healing or
on initial presentation and subsequently as it accumu- to facilitate the effectiveness of other therapeutic
lates. Necrotic tissue serves only as a culture source for measures. Treatment begins with debridement of
bacteria and does not aid healing. The only exception to necrotic tissue, which removes a potential source of
immediate debridement is a dry, chronic, arterial- bacterial infection. Depending on the bacterial load,
insufficiency eschar without evidence of infection. These systemic antimicrobial treatment adjusted to bacterial
types of wound may be best treated by revasculariza- drug resistance is recommended. To provide optimal
tion before debridement. healing conditions, wound moisture imbalances
Open wounds heal optimally in a moist, sterile envi- should be corrected with adequate dressing and com-
ronment. Numerous experimental and clinical studies pression therapy.186 Active medical comorbidities are
have demonstrated that a moist environment speeds aggressively treated. The wound characteristics (size,
healing.184,185 This is thought to occur by preventing des- depth, infection, necrotic tissue component, edema,
iccation at the base of the wound. Desiccation causes drainage) are documented, and local wound care is
necrosis at the base of the wound until an eschar forms, initiated. Care is continued until the wound is clean
which may take several days. During this time, the and ready for reconstruction or heals by secondary
wound is enlarging and initiation of the healing process intention.
is delayed. When the wound is kept covered and moist
without infection, desiccation necrosis and healing Wound dressings
delay are prevented.
A plethora of wound dressings are available for all types
of wounds. High levels of evidence on clinical efficacy
Chronic open wounds
could not be demonstrated for any type.187 However,
A team approach for the treatment of chronic wounds substantial improvement in both convenience and
is employed in wound centers. Members include a comfort has been gained compared with saline gauze
plastic surgeon, vascular surgeon, orthopedic surgeon, dressings. A new class of engineered skin replacements
288 • 15 • Skin wound healing: Repair biology, wound, and scar treatment

that show great promise in chronic wound care has been ulcers.189,190 An international expert working group
developed and is changing chronic wound care. The has defined recommendations for the use of vacuum-
optimal open wound dressing maintains a moist, clean assisted closure treatment for diabetic foot ulcers,
environment that prevents pressure and mechanical complex leg ulcers, pressure ulcers, dehiscent sternal
trauma, reduces edema, stimulates repair, and is inex- wounds, open abdominal wounds, and traumatic
pensive. Less frequent dressing changes and prevention wounds.191 Treatment by vacuum-assisted closure has
of skin irritation are also beneficial. induced granulation tissue formation over exposed
Different classifications of wound dressings are used bone and tendon. The beneficial mechanism is hypoth-
depending on their function in the wound (debride- esized to be due to its reduction of surrounding tissue
ment, antibacterial, occlusive, absorbent, adherence), edema, decrease of bacterial wound colonization, and
type of material used to produce the dressing (e.g., increase in local blood supply, granulation, and wound
hydrocolloid, alginate, collagen) and the physical form cell proliferation. Presumably mechanical transduction,
of the dressing (ointment, film, foam, gel).48 The latter the conversion of mechanical stimuli to biochemical
classifications were used in Table 15.5 to give an over- signals, is responsible for the effects seen with vacuum-
view of currently available wound dressings. assisted wound closure.192 Mechanical forces deform
The films are gas-permeable, maintain a moist envi- tissues; this leads to cellular deformation, which is
ronment, and are useful for partial-thickness dermal followed by stimulation of growth factor pathways,
wounds such as skin graft donor sites. For highly exu- resulting in increased mitosis and production of new
dative wounds, the absorptive dressings are useful to tissue.193
create a moist environment without excess fluid and Recently, the traditional negative-pressure therapy
proteinaceous material accumulation. Hydrocolloid combined with intermittent liquid instillation was intro-
dressings are useful for locations where adhesion is nec- duced for the treatment of problematic acute wounds.
essary, such as the extremities and over bone promi- Typical instillation solutions include normal saline, anti-
nences. They are relatively thick and can stay in place biotics, antifungals, antiseptics, and local anesthetics to
for 2–3 days. As their absorptive capacity is reached, reduce microbial infection or pain, respectively.194
they lose adhesiveness, and gentle atraumatic removal
is facilitated. Hydrogels are similar to hydrocolloid
Compression therapy
dressings except that they have little adhesiveness and
are especially useful for dressing tendons, ligaments or Edema in the lower extremity – whether it is due to
facial wounds. Alginates can absorb 15–20 times their venous insufficiency, lymphedema, cardiovascular
weight of fluid, making them suitable for highly exuding disease, or metabolic derangement – must be aggres-
wounds. However, they should not be used on wounds sively treated if a wound is present. Elevation and com-
with little or no exudate as they will adhere to the pression therapy must be applied. Four-layer dressing
healing wound surface, causing pain and damaging wraps of the lower extremity consisting of gauze, cast
healthy tissue on removal.188 padding, Coban, and elastic wraps are commonly used.
After a venous stasis wound has healed, 30–40-mmHg
pressure stockings must be worn continuously to
Subatmospheric pressure
prevent recurrence, which can be as low as 17% during
A subatmospheric dressing device (VAC, KCI, Texas, the next 3 years.195,196 Caution should be exercised in
US) is available for vacuum-assisted closure. The patients with peripheral neuropathy and arteriosclero-
apparatus consists of a sponge to fill the wound cavity, sis. Lymph edema associated with chronic leg ulcers
a vacuum pump, and a transparent film. It can be used that are refractory to conventional compression therapy
in both inpatient and outpatient settings. Vacuum- can be reduced with intermittent pneumatic compres-
assisted closure is useful for decreasing the size of large sion as an adjunct treatment. Through an electrically
wounds on the extremities and trunk in preparation for inflatable boot, compression within a range of 20–
reconstruction. The device can aid the healing of pres- 120 mmHg at preset intervals is provided, improving
sure sores, skin graft donor sites, and venous stasis venous and lymphatic flow.196
Table 15.5  Classes of wound dressings and engineered skin replacements*
Class Composition Characteristics and function Commercial examples

Gauze Woven cotton fibers Permeable with desiccation; debridement; Curity, Mepilex, Mepitel
painful removal
Tulles Open-weave cloth soaked in soft Low adherent, suitable for flat, shallow wounds Adaptic, Grassolind, Jelonet,
paraffin or chlorhexidine, with low exudates Tullegras, Urgotul
textiles, or multilayered or
perforated plastic films
Film Plastic (polyurethane); Allows water vapor permeation; adhesive; OpSite, Tegaderm
semipermeable impermeable for liquids and bacteria
Foam Hydrophilic (wound side) and Highly absorbent; for necrotic and exudative Lyofoam, Allevyn, Tielle
hydrophobic (outer side) wounds
polyurethane or silicone
foams; semipermeable
Hydrogel Water (96%) and polymer Aqueous environment; requires secondary Aquaform, Intrasite, Purilon,
(polyethylene oxide) dressing; no adherence; not recommended Vigilon, Aquasorb
if infection is present; semipermeable
Hydrocolloid Hydrophilic colloidal particles and Absorbs fluid; necrotic tissue autolysis; little Comfeel, DuoDERM, IntraSite,
adhesive adherence; occlusive Tegasorb
Absorptive powder, Starch copolymers, hydrocolloid Absorbs exudate; used as a filler; good for Aquacel, Geliperm, GranuGel
paste and fiber particles deep wounds paste, DuoDERM granules
Alginate Calcium and sodium salts of Absorbs exudate and forms a hydrophilic gel Algisite, Algosteril, Kaltostat
alginic acid found in brown after ion exchange with wound fluid; not SeaSorb, Sorbsan,
seaweed suited for dry wounds Suprasorb
Antimicrobial 1. Silver (in ionic or nanocrystalline Suited for colonized or infected wounds: 1. Acticoat, Actisorb, Silver 200,
dressings form) 1. Absorptive: cadexomer iodine (caution: thyroid Aquacel Ag
2. Iodine diseases) 2. Iodosorb
(a) as povidone-iodine 2. Control of odor caused by anaerobic bacteria, 3. Metrotop Gel
(b) as cadexomer iodine used for fungating malignant wounds 4. Octinidine/Lavanid Gel
3. Metronidazole gel 3. Used for burns
4. Octinidine gel
Silicone Silicone sheets Sheet induces a localized electromagnetic Sil-K, Mepitel, Mepilex
field and increased skin temperature;
decreases scar formation?
Subatmospheric Vacuum pump, sponge, plastic Sponge conforms to wound and vacuum VAC device
pressure film removes edema fluid and bacteria;
stimulation of granulation, vascularization,
and wound cell proliferation
Dermal collagen Fine-mesh fabric (silicone, nylon) Nonadherent; semipermeable Biobrane
replacement with dermal porcine collagens
Dermal matrix Acellular matrix Permeable; increased stimulation of repair? AlloDerm (human, dermis), SIS
replacement (porcine, small-bowel
submucosa), Integra
(bovine collagen, shark
cartilage, and silicone
sheet)
Dermal living Absorbable matrix populated with Permeable; increased stimulation of repair? Dermagraft (bioabsorbable
replacement allogenic human fibroblasts scaffold), TransCyte (nylon
mesh coated with porcine
collagen)
Epidermal living Autologous keratinocytes on Permeable; increased stimulation of repair? Epicel
replacement murine feeder cells
Skin living Bovine collagen matrix populated Impermeable; increased stimulation of repair? Apligraf, OrCel
replacement with neonatal human
fibroblasts with an outer layer
of human keratinocytes
*Multiple brands within each class are available, and a partial list is given. No particular brands are recommended.
(Modified from Lorenz et al.7 and Enoch et al.196)
290 • 15 • Skin wound healing: Repair biology, wound, and scar treatment

Pharmacologic treatment de-epidermized substratum compared to adult split


thickness skin.200 Presumably, artificial skin products
Collagenases, e.g., streptodornase, streptokinase (Vari­ accelerate wound healing by growth factor delivery and
dase), are useful for the treatment of wounds requiring paracrine stimulation of endogenous wound margin
fine debridement of necrotic tissue that is not amenable keratinocytes.201
to surgical debridement. This could be a thin, superficial Although the unique biology of engineered skin
layer of adherent exudate or small amounts of necrotic replacement dressings can aid in the treatment of
tissue remaining after a bedside wound debridement. impaired wounds, their efficacy compared with autog-
Theoretically, collagenase would be detrimental to a enous skin grafts has not been determined. For many
clean wound in the proliferative phase when ECM syn- biological products, no randomized controlled trials
thesis and deposition favor accumulation rather than exist and current evidence is based on nonrandomized
degradation. studies, reviews, and case reports. Products of human
Because of highly sensitizing potential and induction origin expose the patient to the danger of disease trans-
of bacterial resistance, antibiotic wound dressings and mission and bovine, porcine, or human constituents can
ointments have largely been abandoned. Instead, anti- cause immunological side-effects or have religious and
microbial products have become standard therapy for ethical implications.
contaminated or infected wounds. In burns, silver sul- Cultured epidermal autografts (Epicel) are used for
fadiazine cream has largely been replaced by octinidine second- and third-degree burns and require a culture
gel due to better dressing removal and burn wound time of 2–3 weeks until application to the wound. The
assessment qualities of the gel. patient’s keratinocytes are harvested and cultured on
irradiated murine feeder keratinocytes. Apart from a
long culture time, these products are difficult to handle
Engineered skin replacements
and lead to blistering, resulting in contractures and
With the biotechnology revolution, several dermal and scarring.202
skin replacements are now available through tissue-
engineering technology (Table 15.5). These products
Excessive scar treatment
have the additional potential benefit of accelerating or
augmenting repair because of their biocomponents.
Growth factors are present in products with acellular
Scar classification
dermal matrices. Some products contain living fibro­ Before antiscarring therapy is instituted, an accurate
blast and keratinocyte layers that secrete matrix com­ description and classification of the scar type are useful.
ponents, proteases, and active growth factors. These This assists with assessment of the outcome after late
products are typically placed on open wounds similarly follow-up. A clinically based scar classification scheme
to autologous skin grafts under sterile conditions with has been proposed by the International Advisory Panel
bolster support.197 Their major advantages are the lack on Scar Management, which is composed of plastic sur-
of a donor site wound and scar and placement in an geons, burn surgeons, and dermatologists (Table 15.6).203
office setting. Disadvantages are limited shelf-life and Consistent and accurate diagnosis of scar types is essen-
high cost, especially if they are applied repeatedly to tial for optimal management and useful literature
the same wound, yet models predict that they can be interpretation.
less expensive than nonsurgical therapy.198 The bilay-
ered human keratinocyte–fibroblast construct on a
Therapies
bovine collagen matrix, Apligraf ™, has been shown to
improve healing of venous stasis, diabetic, and arterial- Many treatment modalities have been developed for the
insufficiency wounds.199 However, these studies com­ prevention and management of hypertrophic scarring
pare its use against local wound care. No comparison and keloids. Many of the techniques have well-
has been made against autogenous skin graft treat- documented and time-proven clinical use, but few have
ment. In vitro, reduced keratinocyte migration on a been supported by randomized, prospective studies.
Clinical wound management 291

Table 15.6  Clinical classification of scars203 are mandatory.203 Sun protection to reduce scar hyper-
Scar type Characteristics
pigmentation is essential. Patients who are at increased
risk of excessive scarring benefit from preventive tech-
Mature scar A light-colored, flat scar niques, which include silicone gel sheeting or ointments,
Immature scar A red, sometimes itchy or painful, and slightly hypoallergenic microporous tape, and concurrent intral-
elevated scar in the process of esional steroid injection.203
remodeling. Many of these will mature
normally over time, become flat, and
Silicone gel sheeting is widely used for hypertrophic
assume a pigmentation that is similar to scar and keloid treatment. Silicone gel sheeting has a
the surrounding skin, although they can be 20-plus-year history with several randomized, control-
paler or slightly darker led trials that support its safe and effective use.204,205 It
Linear A red, raised, sometimes itchy scar confined is painless and thus useful for children. Proposed mech-
hypertrophic to the border of the original surgical anisms of action for scar reduction include improved
(e.g., surgical incision. This usually occurs within weeks
or traumatic) of surgery. These scars may increase in
hydration and occlusion,206 increased temperature ele-
scar size rapidly for 3–6 months and then, after vation of 1°C (or less) that can affect collagenase kinet-
a static phase, begin to regress. They ics, and change in the adhesion molecule expression of
generally mature to have an elevated, the lymphocytic infiltrate.207 When treatment with sili-
slightly rope-like appearance with
cone gel sheeting is not feasible (e.g., scar location on
increased width, which is variable. The full
maturation process may take up to 2 years the face, scalp, or neck), silicone oil-based creams are an
alternative.208 For example, silicone-based creams are
Widespread A widespread red, raised, sometimes itchy
hypertrophic scar confined to the border of the burn frequently used on cleft lip repair scars, hypertrophic
(e.g., burn) injury scars after burns, or hand injury without allergic or
scar other untoward reaction to date.
Minor keloid A focally raised, itchy scar extending over Microporous hypoallergenic tape can relieve tension
normal tissue. This may develop up to across wounds and minimize the excessive scar risk
1 year after injury and does not regress on
from shearing. Although there are no prospective, con-
its own. Simple surgical excision is often
followed by recurrence. There may be a trolled studies to support its use, tape is routinely used
genetic abnormality involved in keloid and recommended by many authors.203 The tape is
scarring. Typical sites include earlobes applied for a few weeks after surgery. In patients who
Major keloid A large, raised (>0.5 cm) scar, possibly are at extremely high risk, such as after excision of
painful or pruritic and extending over keloid or hypertrophic scar, concurrent intralesional
normal tissue. This often results after minor
steroid injections can be given prophylactically, fol-
trauma and can continue to spread for
years lowed by monthly injections as necessary. Success rates,
measured by no recurrence, are reported up to 92% for
keloids and 95% for hypertrophic scars at a mean
This inherent problem makes choosing the best treat- follow-up of 30.5 months.209
ment modality difficult on the basis of objective scien-
tific evidence. Therefore, most modalities are applied on Treatment algorithm
the basis of the treating physician’s personal biases and
experiences. Scar therapies, with their respective advan- Immature hypertrophic scars (red)
tages and disadvantages, are listed in Table 15.7.
Once excessive scarring is identified, the International
Advisory Panel on Scar Management consensus initial
Prevention
management is silicone gel sheeting, steroid injection,
The first step toward treatment of excessive scarring is and localized pressure therapy (Fig. 15.8).203 It can be
early recognition and institution of therapy after surgery difficult to predict whether immature hypertrophic
or trauma. Meticulous tissue handling, suturing, and scars (red, slightly raised) will regress or progress. When
wound management with efforts to prevent infection erythema persists for more than 1 month, the risk of
292 • 15 • Skin wound healing: Repair biology, wound, and scar treatment

Table 15.7  Scar prevention and reduction therapies203


Therapy Advantages Disadvantages

Surgical excision Excess scar removed High recurrence without adjuvant therapy, cost
Surgical lengthening (Z-plasty, W-plasty) Increased mobility and range of motion Some excess scar persists, occasionally worse
cosmesis
Steroid intralesional injection Cost, ease Multiple treatments; telangiectasia,
hypopigmentation
Silicone gel sheeting Cost, ease of use, noninvasive Difficult application on head, neck, across joints
Pressure therapy Noninvasive, some proven efficacy Cumbersome garment, cost high if custom-
made, constant use for months to years
Radiation after surgery Some proven efficacy Risk of carcinogenesis, cost
Laser Pulsed-dye 585- or 595-nm laser best for Costs, multiple treatments
decreasing red color; carbon dioxide, Dye laser: reduced efficacy in darker-skinned
Nd : YAG, pulsed Erb : YAG lasers with patients and thick (>1 cm) hypertrophic scars
some efficacy; selective photothermolysis 50% recurrence rate with carbon dioxide laser
by erbium-doped fiber lasers (1550 nm)
Cryotherapy Some proven efficacy in keloid reduction Hypopigmentation, pain, skin atrophy
Microporous tape Ease, low cost No proven benefit, except uncontrolled reports
Popular treatments (vitamin E, onion Ease, low cost No proven benefit
extracts, and other plant creams)
Physical therapy treatments: ultrasound, Patient participation; increased joint range of No quantitative proven efficacy, cost
pulsed electrical stimulation, motion; can decrease scar pain, pruritus
hydrotherapy, massage
Recombinant human TGF-β3 (avotermin), Clinical-phase trials Experimental, high costs, lack of efficacy
rh Il-10 (ilodecakin), TGF-β1 inhibitors
Anti-inflammatory/proliferative medication Fluorouracil: repeated application necessary, Fluorouracil: single administration with recurrence
injections (5-fluorouracil, interferons, no systemic effects Interferon: high recurrence rate, adverse
retinoids, histamine antagonists) Retinoids: still experimental, reduce scar systemic effects
tissue and sebum production Retinoids: adverse effects with photosensitivity
and skin irritation
Calcium channel blockers (verapamil) Clinical trials ongoing, good efficacy Still experimental
Experimental therapies (bleomycin, Experimental, some efficacy No proven benefit, systemic side-effects?
imiquimod, cyclosporine)
Medical needling Clinical phase trials using the Medical Pretreatment with retinoids, general anesthesia
Roll-CIT, low cost, ease needed for treatment of larger surfaces, short
follow-up, recurrence

progression to linear hypertrophic scar increases, and dehiscence. If the original wound was closed following
appropriate therapy should be started. the basic tenets described and healed otherwise unevent-
fully, re-excision with primary closure is not likely to
result in an improved scar. Recurrence of hypertrophic
Linear hypertrophic scars (red, slightly raised)
scar is high in these circumstances, and therefore most
Treatment options include the application of pressure plastic surgeons do not treat hypertrophic scar with exci-
garments or topical silicone sheets, 585-nm pulsed-dye sion and primary closure unless they plan adjuvant
laser therapy, and re-excision. The last option is most therapy. Silicone gel has proven benefit from rand-
useful in cases of excess scar due to wound infection or omized, controlled trials and is a recommended first-line
Clinical wound management 293

Scar

Classification

Immature hypertrophic Linear hypertrophic Minor keloid Major keloid Widespread burn
(red, slightly raised) (red/raised, itchy) (red, raised) -high risk- hypertrophic scar
(dark/raised) (red, raised)

If patient concerned or Silicone gel sheeting (2 months)


high risk for excessive
scar: tape or silicone
Initial gel sheeting/ointment Steroid injections 2.5–20 mg/mL (face); 20–40 mg/mL (body); Burn
management repeat monthly as needed specialty
Progress to treatment as unit
for hypertrophic scar
if erythema persists Localized pressure therapy if possible
more than 1 month Duration 3–12 months

Pressure
therapy Specific wavelength laser therapy Pressure
garments and/or
silicone gel
Secondary Surgery with adjunctive silicone gel sheeting sheeting
management (duration 2 months) (6–12 months)

Unit specializing in scar therapy

Combination/monotherapy—
Primarily: Steroids, silicones, pressure therapy, surgery/grafting
Occasionally: Cryotherapy, radiotherapy, laser, other therapies

Fig. 15.8  Scar management algorithm. (Reproduced from Mustoe TA, Cooter RD, Gold MH, et al. International clinical recommendations on scar management.
Plast Reconstr Surg 2002; 110: 560–571, with permission.)

therapy.203 Concurrent steroid injections are helpful for


Widespread burn hypertrophic scars (red, raised)
pruritic or resistant scars. Pressure therapy can be added
when feasible. Pulsed-dye laser (585 and 595 nm) treat- Extensive surface-area burn hypertrophic scars may
ment of hypertrophic scars is another alternative. Light best be treated at burn centers when feasible.
energy from the pulsed-dye laser is absorbed by intra- Multimodality therapies are generally used; these
vasal hemoglobin leading to coagulation, vessel obstruc- include silicone gel sheeting, custom-fitted pressure
tion, and tissue hypoxia with necrosis. As a consequence, garments, and physical therapy alone or with massage,
new collagen synthesis, collagen fiber realignment, and electrical stimulation, or ultrasound. Steroid injection of
remodeling are the molecular bases for the effects seen especially difficult areas is sometimes necessary. Laser
after pulsed-dye laser treatment.210 treatment can be useful.211 Surgical treatment with
294 • 15 • Skin wound healing: Repair biology, wound, and scar treatment

Z-plasty, excision and grafting, and flap reconstruction when excessive, lead to clinical disease. For example,
is frequently required.203 fibrosis of joint capsules leads to contracture, which can
be debilitating. Anastomotic fibrosis of free jejunal flaps
Minor keloids (red, raised) to the esophagus or pharynx leads to lumen obstruction.
Breast implant capsules are the normal fibrotic response
No uniformly successful treatment for keloid scar exists.
to injury, which can be excessive and develop into cap-
Excision and primary closure invariably result in recur-
sular contracture. Nerve repair sites with excess fibrosis
rence. Therefore, additional therapy is necessary, and its
commonly develop neuromas and functional repair
efficacy depends on the timing of the patient’s presenta-
failure. Scarring at tendon repair sites restricts motion.
tion. Steroid injection directly into the keloid has the
Dupuytren disease is a fibroproliferative disorder that
most benefit early in the keloid course.212 Steroid has
involves the palmar fascia and the deep dermis. Each
been shown to decrease collagen gene expression.213
of these excessive scarring conditions leads to morbid-
Mixed with 2% plain lidocaine in a 50 : 50 ratio, triamci-
ity of the patient, which can require further surgical
nolone acetonide 10 mg/mL is commonly used initially;
treatment.
if no response occurs, 40 mg/mL concentration is
attempted. Silicone gel sheeting should be used concur-
rently. Patients presenting with mature keloid lesions of Emerging scar therapies
months to years in duration that are slowly changing
A great deal of research is focused on the development
respond poorly to steroid injection and silicone sheet-
of treatment strategies to reduce or prevent scarring.
ing. Surgical excision with adjuvant therapy including
Prompted by fetal wound-healing observations, inves-
intralesional steroids, silicone sheeting, and pressure
tigators initially analyzed the antiscarring effect of anti-
therapy is a reasonable treatment alternative.210 Careful
TGF-β strategies. Treatment with neutralizing antibody
follow-up is necessary to prevent recurrence. A short
to TGF-β decreased the inflammatory response and
course of low-dose radiation therapy to the keloid exci-
reduced scar formation in experimental postnatal rodent
sion site immediately after excision has been shown to
wounds.215 More evidence that TGF-β-related strategies
reduce the rate of recurrence.214
may be successful is provided by a study showing that
exogenous addition to wounds of fibromodulin, a TGF-β
Major keloids (dark, raised)
modulator, reduces scar.89 Clearly, more studies are
Major keloids are difficult to treat effectively, and many needed, and because of the redundancy of action among
are resistant to any treatment. Surgical therapy with all growth factors, TGF-β is likely not to be the only growth
adjunctive therapies described before may still fail and factor targeted to reduce human scar and fibrosis.
result in recurrence. Radiation therapy is generally used Newer treatment modalities, such as intralesional
in this group, provided the patient is not young and injection of bleomycin, 5-fluorouracil, and, imiquimod
accepts the possible risk of late cancer formation. Before may be useful in the future. These act by decreasing
surgery is performed with postoperative adjuvant inflammation and collagen synthesis; however, their
therapy, patients should be counseled about the high mechanisms of action remain under investigation.
rate of recurrence with the risk that the next keloid will Interferon therapy was left because of adverse side-
be larger and more difficult to control. As recommended effects and high recurrence rates.210,216 Other novel strat-
by the International Advisory Panel on Scar egies include the application of antifibrotic human
Management, these patients may best be treated by recombinant growth factors and cytokines (e.g., rh
clinicians with a special interest in keloid treatment.203 IL-10), anti-inflammatory substances, protease inhibi-
tors, and molecules that interfere with profibrotic
Impact of scar on plastic surgery cytokine function (e.g., TGF-β) and collagen synthesis
at the wound site.217
Besides hypertrophic scarring and keloid formation in Clinical trials are currently performed using two dif-
the skin, scar affects every aspect of plastic surgery. ferent novel techniques for scar reduction. Selective
Scar and fibrosis are the outcome after tissue repair and, photothermolysis is induced by erbium-doped fiber
Future perspectives 295

lasers operating at a wavelength of 1550 nm, targeting Gene therapy


water as a chromophore. This creates dermal microle-
sions with controlled width, depth, and densities fol- Gene therapy to enhance wound repair through intro-
lowed by neocollagenesis.218 These lasers are used for duction of a growth factor gene, or other therapeutic
the treatment of hyperpigmentation and acne scars as gene, into repair cells is actively undergoing investiga-
well as surgical and traumatic scars. Percutaneous col- tion. With this approach, the gene must be delivered to
lagen induction therapy is currently used in our depart- the target cell and its expression sustained at a thera-
ment for reduction of hypertrophic scars, striae distensae, peutic level; the level and timing of its expression must
wrinkles, and skin laxity. After pretreatment with be reversibly controlled so that it can be stopped when
vitamin A and C cream, patients are treated under it is no longer needed.224 Several delivery vehicles are
anesthesia. Needles of the Medical Roll-CIT create being developed, each with unique advantages and dis-
dermal microwounds and separate collagen structures advantages. Replication-deficient adenovirus, herpes
connecting the scar with the upper dermis. Through the simplex virus type 1, and retrovirus vectors have been
following inflammatory reaction, collagen remodeling successfully used to transfect epidermal and dermal
is induced with consequent scar flattening.219 cells and tissues.224,225 Other delivery approaches elimi-
nate the virus and its possible risk of untoward side-
effects due to antigenicity, potential recombination with
Future perspectives wild-type viruses, and cellular damage from persistent
viral exposure. Nonviral methods are categorized into
Growth factor and chemical and physical methods of gene transfer.
protease-scavenging therapy Liposome-mediated gene transfer is a chemical method
in which the gene is bound to a synthetic liposome
The best-studied growth factors with the most promise vesicle and carried into the target cell by endocytosis.
to improve healing are PDGF, TGF-β, and members of FGF,226 IGF-1,224 KGF,227 and VEGF228 genes have been
the FGF family (Table 15.1). However, before wide- successfully transferred into experimental wounds.
spread use, several obstacles must be overcome, includ- Particle-mediated gene transfer (gene gun) directly
ing efficacious application, vehicle development, and injects DNA into the cells and has been used for TGF-
cost. For example, nonhealing human wound fluid and β1,229 EGF,230 and PDGF231 gene transfer in experimental
tissue have increased protease activity, which probably wounds. Gene therapy is likely to become an increas-
degrades rapidly exogenously applied peptide growth ingly important tool for the treatment of nonhealing
factors.118,220 Products targeting excessive activity could wounds as its methods advance. In a phase I clinical
be protease-scavenging matrices (e.g., Promogran),196 trial the safety, feasibility, and biologic plausibility of
selective inhibitors,221 or specific antibodies. adenovirus-mediated rhPDGF-BB gene therapy to treat
Experimental and clinical trials have reported some venous leg ulcer disease were demonstrated.232
efficacy of growth factors for treatment of chronic or Extreme redundancy exists in the function of growth
burn wounds. VEGF gene transfer was effective in factors at the wound site. In addition, multiple simulta-
ischemic leg ulcers with formation of collateral vessels.222 neous processes are occurring during repair. Thus, it is
Topical recombinant bovine FGF, human KGF (repifer- likely that multiple growth factors may need to be
min), and GM-CSF increased healing of chronic pres- added on impaired and even otherwise normal wounds
sure and venous leg ulcers, respectively. Topical to effect a clinically significant improvement in repair
recombinant human EGF had a positive effect on healing quality and rate. Different growth factor combinations
of second-degree burn wounds whereas topical recom- may be needed to treat impaired wounds due to
binant human PDGF-BB (becaplermin), TGF-β2 and different underlying diseases, such as diabetes versus
granulocyte colony-stimulating factor were effective on arterial insufficiency. To complicate matters further,
diabetic foot ulcers. However, for chronic wound neutralization of certain growth factors may be neces-
therapy, so far only PDGF-BB has been licensed for com- sary as well as augmentation of other factors in the
mercial use in the US and Europe.196,223 same wound.
296 • 15 • Skin wound healing: Repair biology, wound, and scar treatment

Stem cell therapy muscle, bone, tendons, cartilage, and fat, or to


“cross-differentiate” into nonmesodermal cells under
Multipotent adult stem cells have come into focus as appropriate conditions.235 Promising results were
cellular replacement and growth factor delivery source reported treating chronic wounds with bone marrow-
for skin tissue defects and nonhealing wounds because derived MSC.236 Because of easier accessibility and
of abundance and easy accessibility in each patient. tissue abundance, adipose tissue is an important
Furthermore, autologous grafting avoids the risk of source of mesenchymal stem cells. Harvesting and
rejection and ethical or moral considerations concerning labeling techniques have been refined and automa-
allogenic or embryonic material. Two major types of tized for better clinical availability.237,238 Pilot studies
adult stem cells are available: epidermal and mesenchy- using multipotent adipose tissue-derived stem cells
mal stem cells (MSCs). Epidermal stem cells are located (adMSC) for soft-tissue replacement and coverage
to the basal layer and hair follicles, and contribute to of nonhealing wounds are currently ongoing in our
re-epithelialization after wounding.233 Takahashi et al. department.
reported another promising access to epidermal cells Our understanding of the biomolecular regulation of
by reprogramming dermal fibroblasts into pluripotent repair and handling of autologous cells and tissues is
epidermal stem cells.234 rapidly expanding while modern biotechnology keeps
MSCs are found in various mesoderm-derived pace with scientific progress. Pilot studies will prove
tissues. MSCs have the potential either to transdiffer- whether it is worthwhile continuing these futuristic
entiate into tissues of mesodermal origin, e.g., skeletal approaches with controlled clinical trials.

Access the complete reference list online at http://www.expertconsult.com


10. Singer AJ, Clark RA. Cutaneous wound healing. 51. Werner S, Grose R. Regulation of wound healing by
N Engl J Med. 1999;341:738–746. growth factors and cytokines. Physiol Rev. 2003;83:
17. Sternlicht MD, Werb Z. How matrix metalloproteinases 835–870.
regulate cell behavior. Annu Rev Cell Dev Biol. 75. Buchanan EP, Longaker MT, Lorenz HP. Fetal skin
2001;17:463–516. wound healing. Adv Clin Chem. 2009;48:137–161.
26. Risau W. Mechanisms of angiogenesis. Nature. 1997; 86. Kong W, Li S, Longaker MT, et al. Blood-derived small
386:671–674. Dot cells reduce scar in wound healing. Exp Cell Res.
30. Tomasek JJ, Gabbiani G, Hinz B, et al. Myofibroblasts 2008;314:1529–1539.
and mechano-regulation of connective tissue 138. Lansdown AB, Mirastschijski U, Stubbs N, et al. Zinc
remodelling. Nat Rev Mol Cell Biol. 2002;3: in wound healing: theoretical, experimental, and
349–363. clinical aspects. Wound Repair Regen. 2007;15:2–16.
42. Toriseva M, Kahari VM. Proteinases in cutaneous 161. Werner S, Krieg T, Smola H. Keratinocyte-fibroblast
wound healing. Cell Mol Life Sci. 2009;66: interactions in wound healing. J Invest Dermatol.
203–224. 2007;127:998–1008.
References 296.e1

19. Heino J. The collagen receptor integrins have distinct


References ligand recognition and signaling functions. Matrix Biol.
2000;19:319–323.
1. Majno G. The healing hand: Man and wound in the ancient 20. White DJ, Puranen S, Johnson MS, et al. The collagen
world. Cambridge, Massachusetts: Harvard University receptor subfamily of the integrins. Int J Biochem Cell
Press; 1975. Biol. 2004;36:1405–1410.
2. Evans J, Flavin S. Honey: a guide for healthcare 21. Miller EJ, Gay S. Collagen structure and function.
professionals. Br J Nurs. 2008;17(15):S24, S26, S28–S30. Philadelphia: WB Saunders; 1992.
3. Vogel VJ. American Indian Medicine. New York: 22. Myers JC, Amenta PS, Dion AS, et al. The molecular
University of Oklahoma Press; 1970. structure of human tissue type XV presents a unique
4. Bennett JS, Berger BW, Billings PC. The structure and conformation among the collagens. Biochem J.
function of platelet integrins. J Thromb Haemost. 2007;404:535–544.
2009;7(Suppl 1):200–205. 23. Ryynanen J, Sollberg S, Olsen DR, et al. Transforming
5. Ginsberg MH, Du X, Plow EF. Inside-out integrin growth factor-beta up-regulates type VII collagen gene
signalling. Curr Opin Cell Biol. 1992;4:766–771. expression in normal and transformed epidermal
6. Szpaderska AM, Egozi EI, Gamelli RL, et al. The effect keratinocytes in culture. Biochem Biophys Res Commun.
of thrombocytopenia on dermal wound healing. 1991;180:673–680.
J Invest Dermatol. 2003;120:1130–1137. 24. Ricard-Blum S, Ruggiero F. The collagen superfamily:
7. Lorenz HP, Longaker MT. Wounds: biology, pathology, from the extracellular matrix to the cell membrane.
and management. New York: Springer-Verlag; 2000. Pathol Biol (Paris). 2005;53:430–442.
8. Roberts HR, Tabares AH. Overview of the coagulation 25. Kobayashi H, Ishii M, Chanoki M, et al.
reactions. New York: Marcel Dekker; 1995. Immunohistochemical localization of lysyl oxidase in
normal human skin. Br J Dermatol. 1994;131:325–330.
9. Martin P, Leibovich SJ. Inflammatory cells during
wound repair: the good, the bad and the ugly. Trends 26. Risau W. Mechanisms of angiogenesis. Nature.
Cell Biol. 2005;15:599–607. 1997;386:671–674.
10. Singer AJ, Clark RA. Cutaneous wound healing. 27. Dvorak HF. VPF/VEGF and the angiogenic response.
N Engl J Med. 1999;341:738–746. Semin Perinatol. 2000;24:75–78.
11. Riches DWH. Macrophage involvement in wound repair, 28. Brooks PC, Clark RA, Cheresh DA. Requirement of
remodeling, and fibrosis. New York: Plenum; 1996. vascular integrin alpha v beta 3 for angiogenesis.
Science. 1994;264:569–571.
12. Leibovich SJ, Ross R. The role of the macrophage in
wound repair. A study with hydrocortisone and 29. Berry DP, Harding KG, Stanton MR, et al. Human
antimacrophage serum. Am J Pathol. 1975;78:71–100. wound contraction: collagen organization, fibroblasts,
and myofibroblasts. Plast Reconstr Surg. 1998;102:
13. Martin P, D’Souza D, Martin J, et al. Wound healing in
124–131; discussion 132–4.
the PU.1 null mouse–tissue repair is not dependent on
inflammatory cells. Curr Biol. 2003;13:1122–1128. 30. Tomasek JJ, Gabbiani G, Hinz B, et al. Myofibroblasts
and mechano-regulation of connective tissue
14. McKercher SR, Torbett BE, Anderson KL, et al.
remodelling. Nat Rev Mol Cell Biol. 2002;3:349–363.
Targeted disruption of the PU.1 gene results in
multiple hematopoietic abnormalities. Embo J. 31. Hinz B, Mastrangelo D, Iselin CE, et al. Mechanical
1996;15:5647–5658. tension controls granulation tissue contractile activity
and myofibroblast differentiation. Am J Pathol.
15. Lund LR, Rømer J, Bugge TH, et al. Functional overlap
2001;159:1009–1020.
between two classes of matrix-degrading proteases in
wound healing. Embo J. 1999;18:4645–4656. 32. Ehrlich HP, Gabbiani G, Meda P. Cell coupling
modulates the contraction of fibroblast-populated
16. Mirastschijski U, Schnabel R, Claes J, et al. Matrix
collagen lattices. J Cell Physiol. 2000;184:86–92.
metalloproteinase inhibition delays wound healing and
blocks latent transforming growth factor-β1 promoted 33. Ralston DR, Layton C, Dalley AJ, et al. Keratinocytes
myofibroblast formation and function. Wound Repair contract human dermal extracellular matrix and reduce
Regen. 2010;18:223–224. soluble fibronectin production by fibroblasts in a skin
composite model. Br J Plast Surg. 1997;50:408–415.
17. Sternlicht MD, Werb Z. How matrix metalloproteinases
regulate cell behavior. Annu Rev Cell Dev Biol. 34. Martin P. Wound healing–aiming for perfect skin
2001;17:463–516. regeneration. Science. 1997;276:75–81.
18. Brix K, Dunkhorst A, Mayer K, et al. Cysteine 35. Noe V, Fingleton B, Jacobs K, et al. Release of an
cathepsins: cellular roadmap to different functions. invasion promoter E-cadherin fragment by matrilysin
Biochimie. 2008;90:194–207. and stromelysin-1. J Cell Sci. 2001;114:111–118.
296.e2 • 15 • Skin wound healing: Repair biology, wound, and scar treatment

36. Parks WC. Matrix metalloproteinases in repair. Wound 52. Ashcroft GS, Yang X, Glick AB, et al. Mice lacking
Repair Regen. 1999;7:423–432. Smad3 show accelerated wound healing and an
37. Dumin JA, Dickeson SK, Stricker TP, et al. Pro- impaired local inflammatory response. Nat Cell Biol.
collagenase-1 (matrix metalloproteinase-1) binds the 1999;1:260–266.
alpha(2)beta(1) integrin upon release from 53. Barrientos S, Stojadinovic O, Golinko MS, et al. Growth
keratinocytes migrating on type I collagen. J Biol Chem. factors and cytokines in wound healing. Wound Repair
2001;276:29368–29374. Regen. 2008;16:585–601.
38. Mirastschijski U, Haaksma CJ, Tomasek JJ, et al. Matrix 54. Swift ME, Kleinman HK, DiPietro LA. Impaired
metalloproteinase inhibitor GM 6001 attenuates wound repair and delayed angiogenesis in aged mice.
keratinocyte migration, contraction and myofibroblast Lab Invest. 1999;79:1479–1487.
formation in skin wounds. Exp Cell Res. 55. Werner S, Breeden M, Hubner G, et al. Induction of
2004;299:465–475. keratinocyte growth factor expression is reduced and
39. Mirastschijski U, Impola U, Karsdal MA, et al. Matrix delayed during wound healing in the genetically
metalloproteinase inhibitor BB-3103 unlike the serine diabetic mouse. J Invest Dermatol. 1994;103:469–473.
proteinase inhibitor aprotinin abrogates epidermal 56. Jimenez PA, Rampy MA. Keratinocyte growth factor-2
healing of human skin wounds ex vivo. J Invest accelerates wound healing in incisional wounds. J Surg
Dermatol. 2002;118:55–64. Res. 1999;81:238–242.
40. Friedl P. Dynamic imaging of cellular interactions with 57. Robson MC, Phillips TJ, Falanga V, et al. Randomized
extracellular matrix. Histochem Cell Biol. trial of topically applied repifermin (recombinant
2004;122:183–190. human keratinocyte growth factor-2) to accelerate
41. Marinkovich MP, Keene DR, Rimberg CS, et al. wound healing in venous ulcers. Wound Repair Regen.
Cellular origin of the dermal-epidermal basement 2001;9:347–352.
membrane. Dev Dyn. 1993;197:255–267. 58. Nissen NN, Polverini PJ, Koch AE, et al. Vascular
42. Toriseva M, Kahari VM. Proteinases in cutaneous endothelial growth factor mediates angiogenic activity
wound healing. Cell Mol Life Sci. 2009;66:203–224. during the proliferative phase of wound healing. Am J
Pathol. 1998;152:1445–1452.
43. Lambert E, Dasse E, Haye B, et al. TIMPs as
multifacial proteins. Crit Rev Oncol Hematol. 59. Banks RE, Forbes MA, Kinsey SE, et al. Release of the
2004;49:187–198. angiogenic cytokine vascular endothelial growth factor
(VEGF) from platelets: significance for VEGF
44. Bullard KM, Lund L, Mudgett JS, et al. Impaired
measurements and cancer biology. Br J Cancer.
wound contraction in stromelysin-1-deficient mice.
1998;77:956–964.
Ann Surg. 1999;230:260–265.
60. Gaudry M, Bregerie O, Andrieu V, et al. Intracellular
45. Levenson SM, Geever EF, Crowley LV, et al. The
pool of vascular endothelial growth factor in human
Healing Of Rat Skin Wounds. Ann Surg.
neutrophils. Blood. 1997;90:4153–4161.
1965;161:293–308.
61. Berse B, Brown LF, Van de Water L, et al. Vascular
46. Witte MB, Thornton FJ, Kiyama T, et al. permeability factor (vascular endothelial growth
Metalloproteinase inhibitors and wound healing: a factor) gene is expressed differentially in normal
novel enhancer of wound strength. Surgery. tissues, macrophages, and tumors. Mol Biol Cell.
1998;124:464–470. 1992;3:211–220.
47. Khan MN, Davies CG. Advances in the management 62. Frank S, Hubner G, Breier G, et al. Regulation of
of leg ulcers–the potential role of growth factors. Int vascular endothelial growth factor expression in
Wound J. 2006;3:113–120. cultured keratinocytes. Implications for normal and
48. Boateng JS, Matthews KH, Stevens HN, et al. Wound impaired wound healing. J Biol Chem.
healing dressings and drug delivery systems: a review. 1995;270:12607–12613.
J Pharm Sci. 2008;97:2892–2923. 63. Baumgartner I, Pieczek A, Manor O, et al. Constitutive
49. Heldin CH, Westermark B. Mechanism of action and in expression of phVEGF165 after intramuscular gene
vivo role of platelet-derived growth factor. Physiol Rev. transfer promotes collateral vessel development in
1999;79:1283–1316. patients with critical limb ischemia. Circulation.
50. Nagai MK, Embil JM. Becaplermin: recombinant 1998;97:1114–1123.
platelet derived growth factor, a new treatment for 64. Nagy JA, Dvorak AM, Dvorak HF. VEGF-A and the
healing diabetic foot ulcers. Expert Opin Biol Ther. induction of pathological angiogenesis. Annu Rev
2002;2:211–218. Pathol. 2007;2:251–275.
51. Werner S, Grose R. Regulation of wound healing by 65. Carmeliet P, Moons L, Luttun A, et al. Synergism
growth factors and cytokines. Physiol Rev. between vascular endothelial growth factor and
2003;83:835–870. placental growth factor contributes to angiogenesis
References 296.e3

and plasma extravasation in pathological conditions. 80. Whitby DJ, Ferguson MW. The extracellular matrix of
Nat Med. 2001;7:575–583. lip wounds in fetal, neonatal and adult mice.
66. Nagy JA, Dvorak AM, Dvorak HF. VEGF-A(164/165) Development. 1991;112:651–668.
and PlGF: roles in angiogenesis and arteriogenesis. 81. Rolfe KJ, Richardson J, Vigor C, et al. A role for
Trends Cardiovasc Med. 2003;13:169–175. TGF-beta1-induced cellular responses during wound
67. Cianfarani F, Zambruno G, Brogelli L, et al. Placenta healing of the non-scarring early human fetus? J Invest
growth factor in diabetic wound healing: altered Dermatol. 2007;127:2656–2667.
expression and therapeutic potential. Am J Pathol. 82. Dang CM, Beanes SR, Lee H, et al. Scarless fetal
2006;169:1167–1182. wounds are associated with an increased matrix
68. Gallucci RM, Simeonova PP, Matheson JM, et al. metalloproteinase-to-tissue-derived inhibitor of
Impaired cutaneous wound healing in interleukin- metalloproteinase ratio. Plast Reconstr Surg.
6-deficient and immunosuppressed mice. Faseb J. 2003;111:2273–2285.
2000;14:2525–2531. 83. Estes JM, Vande Berg JS, Adzick NS, et al. Phenotypic
69. Gough A, Clapperton M, Rolando N, et al. and functional features of myofibroblasts in sheep fetal
Randomised placebo-controlled trial of granulocyte- wounds. Differentiation. 1994;56:173–181.
colony stimulating factor in diabetic foot infection. 84. Ellis I, Banyard J, Schor SL. Differential response of
Lancet. 1997;350:855–859. fetal and adult fibroblasts to cytokines: cell migration
70. Cianfarani F, Tommasi R, Failla CM, et al. and hyaluronan synthesis. Development.
Granulocyte/macrophage colony-stimulating factor 1997;124:1593–1600.
treatment of human chronic ulcers promotes 85. Tumbar T, Guasch G, Greco V, et al. Defining the
angiogenesis associated with de novo vascular epithelial stem cell niche in skin. Science.
endothelial growth factor transcription in the ulcer 2004;303:359–363.
bed. Br J Dermatol. 2006;154:34–41. 86. Kong W, Li S, Longaker MT, et al. Blood-derived small
71. Gold LI, Sung JJ, Siebert JW, et al. Type I (RI) and type Dot cells reduce scar in wound healing. Exp Cell Res.
II (RII) receptors for transforming growth factor-beta 2008;314:1529–1539.
isoforms are expressed subsequent to transforming 87. Olutoye OO, Yager DR, Cohen IK, et al. Lower
growth factor-beta ligands during excisional wound cytokine release by fetal porcine platelets: a possible
repair. Am J Pathol. 1997;150:209–222. explanation for reduced inflammation after fetal
72. Ashcroft GS, Roberts AB. Loss of Smad3 modulates wounding. J Pediatr Surg. 1996;31:91–95.
wound healing. Cytokine Growth Factor Rev. 88. Colwell AS, Beanes SR, Soo C, et al. Increased
2000;11:125–131. angiogenesis and expression of vascular endothelial
73. Lorenz HP, Whitby DJ, Longaker MT, et al. Fetal growth factor during scarless repair. Plast Reconstr
wound healing. The ontogeny of scar formation Surg. 2005;115:204–212.
in the non-human primate. Ann Surg. 1993;217: 89. Soo C, Hu FY, Zhang X, et al. Differential expression of
391–396. fibromodulin, a transforming growth factor-beta
74. Beanes SR, Hu FY, Soo C, et al. Confocal microscopic modulator, in fetal skin development and scarless
analysis of scarless repair in the fetal rat: defining the repair. Am J Pathol. 2000;157:423–433.
transition. Plast Reconstr Surg. 2002;109:160–170. 90. Belford DA. The mechanism of excisional fetal wound
75. Buchanan EP, Longaker MT, Lorenz HP. Fetal skin repair in vitro is responsive to growth factors.
wound healing. Adv Clin Chem. 2009;48:137–161. Endocrinology. 1997;138:3987–3996.
76. Armstrong JR, Ferguson MW. Ontogeny of the skin 91. Lanning DA, Nwomeh BC, Montante SJ, et al. TGF-
and the transition from scar-free to scarring phenotype beta1 alters the healing of cutaneous fetal excisional
during wound healing in the pouch young of a wounds. J Pediatr Surg. 1999;34:695–700.
marsupial, Monodelphis domestica. Dev Biol. 92. Stelnicki EJ, Longaker MT, Holmes D, et al. Bone
1995;169:242–260. morphogenetic protein-2 induces scar formation and
77. Meuli M, Lorenz HP, Hedrick MH, et al. Scar skin maturation in the second trimester fetus. Plast
formation in the fetal alimentary tract. J Pediatr Surg. Reconstr Surg. 1998;101:12–19.
1995;30:392–395. 93. Stelnicki EJ, Arbeit J, Cass DL, et al. Modulation of
78. Longaker MT, Whitby DJ, Ferguson MW, et al. Adult the human homeobox genes PRX-2 and HOXB13 in
skin wounds in the fetal environment heal with scar scarless fetal wounds. J Invest Dermatol. 1998;111:
formation. Ann Surg. 1994;219:65–72. 57–63.
79. Cass DL, Bullard KM, Sylvester KG, et al. Wound size 94. Reddy S, Andl T, Bagasra A, et al. Characterization of
and gestational age modulate scar formation in fetal Wnt gene expression in developing and postnatal hair
wound repair. J Pediatr Surg. 1997;32:411–415. follicles and identification of Wnt5a as a target of Sonic
296.e4 • 15 • Skin wound healing: Repair biology, wound, and scar treatment

hedgehog in hair follicle morphogenesis. Mech Dev. 109. Padberg FT, Back TL, Thompson PN, et al.
2001;107:69–82. Transcutaneous oxygen (TcPO2) estimates probability
95. Colwell AS, Krummel TM, Longaker MT, et al. Wnt-4 of healing in the ischemic extremity. J Surg Res.
expression is increased in fibroblasts after TGF-beta1 1996;60:365–369.
stimulation and during fetal and postnatal wound 110. Fife CE, Smart DR, Sheffield PJ, et al. Transcutaneous
repair. Plast Reconstr Surg. 2006;117:2297–2301. oximetry in clinical practice: consensus statements
96. Carre AL, James AW, MacLeod L, et al. Interaction of from an expert panel based on evidence. Undersea
wingless protein (Wnt), transforming growth factor- Hyperb Med. 2009;36:43–53.
beta1, and hyaluronan production in fetal and 111. Mathieu D, Mani R. A review of the clinical
postnatal fibroblasts. Plast Reconstr Surg. significance of tissue hypoxia measurements in lower
2010;125:74–88. extremity wound management. Int J Low Extrem
97. Coolen NA, Schouten KC, Middelkoop E, et al. Wounds. 2007;6:273–283.
Comparison between human fetal and adult skin. Arch 112. Slovut DP, Sullivan TM. Critical limb ischemia:
Dermatol Res. 2010;302:47–55. medical and surgical management. Vasc Med.
98. Ferguson MW, Duncan J, Bond J, et al. Prophylactic 2008;13:281–291.
administration of avotermin for improvement of skin 113. Blakytny R, Jude EB. Altered molecular mechanisms of
scarring: three double-blind, placebo-controlled, phase diabetic foot ulcers. Int J Low Extrem Wounds.
I/II studies. Lancet. 2009;373:1264–1274. 2009;8:95–104.
99. Sibbald RG, Orsted H, Schultz GS, et al. Preparing the 114. Galkowska H, Wojewodzka U, Olszewski WL.
wound bed 2003: focus on infection and inflammation. Chemokines, cytokines, and growth factors in
Ostomy Wound Manage. 2003;49:23–51. keratinocytes and dermal endothelial cells in the
100. Bauer J, Phillips LG. MOC-PSSM CME article: Pressure margin of chronic diabetic foot ulcers. Wound Repair
sores. Plast Reconstr Surg. 2008;121(Suppl):1–10. Regen. 2006;14:558–565.
101. Schubert V, Heraud J. The effects of pressure and shear 115. Mishima Y, Kuyama A, Tada A, et al. Relationship
on skin microcirculation in elderly stroke patients between serum tumor necrosis factor-alpha and insulin
lying in supine or semi-recumbent positions. Age resistance in obese men with Type 2 diabetes mellitus.
Ageing. 1994;23:405–410. Diabetes Res Clin Pract. 2001;52:119–123.
102. Rennert R, Golinko M, Yan A, et al. Developing and 116. Loots MA, Lamme EN, Zeegelaar J, et al. Differences
evaluating outcomes of an evidence-based protocol for in cellular infiltrate and extracellular matrix of chronic
the treatment of osteomyelitis in Stage IV pressure diabetic and venous ulcers versus acute wounds.
ulcers: a literature and wound electronic medical J Invest Dermatol. 1998;111:850–857.
record database review. Ostomy Wound Manage. 117. Nwomeh BC, Liang HX, Cohen IK, et al. MMP-8 is the
2009;55:42–53. predominant collagenase in healing wounds and
103. Relander M, Palmer B. Recurrence of surgically treated nonhealing ulcers. J Surg Res. 1999;81:189–195.
pressure sores. Scand J Plast Reconstr Surg Hand Surg. 118. Mirastschijski U, Impola U, Jahkola T, et al. Ectopic
1988;22:89–92. localization of matrix metalloproteinase-9 in chronic
104. Raju S, Neglen P. Clinical practice. Chronic venous cutaneous wounds. Hum Pathol. 2002;33:355–364.
insufficiency and varicose veins. N Engl J Med. 119. Acosta JB, del Barco DG, Vera DC, et al. The pro-
2009;360:2319–2327. inflammatory environment in recalcitrant diabetic foot
104a.  Mortimer PS. Evaluation of lymphatic function: wounds. Int Wound J. 2008;5:530–539.
abnormal lymph drainage in venous disease. Int 120. Darby IA, Bisucci T, Hewitson TD, et al. Apoptosis is
Angiol. 1995;14(Suppl 1):32–35. increased in a model of diabetes-impaired wound
105. Valencia IC, Falabella A, Kirsner RS, et al. Chronic healing in genetically diabetic mice. Int J Biochem Cell
venous insufficiency and venous leg ulceration. J Am Biol. 1997;29:191–200.
Acad Dermatol. 2001;44:401–421; quiz 422–4. 121. Goodson 3rd WH, Hunt TK. Wound healing and the
106. Sen CK. Wound healing essentials: let there be oxygen. diabetic patient. Surg Gynecol Obstet. 1979;149:
Wound Repair Regen. 2009;17:1–18. 600–608.
107. Al-Qaisi M, Nott DM, King DH, et al. Ankle brachial 122. Bernstein EF, Harisiadis L, Salomon GD, et al. Healing
pressure index (ABPI): An update for practitioners. impairment of open wounds by skin irradiation.
Vasc Health Risk Manag. 2009;5:833–841. J Dermatol Surg Oncol. 1994;20:757–760.
108. Clark N. American Diabetes Association. Peripheral 123. Niinikoski J, Bakker D, Cronje F, et al. ECHM-ETRS
arterial disease in people with diabetes. Diabetes Care. joint conference on oxygen and tissue repair, Ravenna,
2003;26:3333–3341. Italy, October 27–28, 2006: recommendations by the
References 296.e5

international jury. Int J Low Extrem Wounds. glucocorticoids in vitro and during wound healing.
2007;6:139–142. J Invest Dermatol. 1995;105:579–584.
124. Robson MC. Wound infection. A failure of wound 143. Ehrlich HP, Tarver H, Hunt TK. Effects of vitamin A
healing caused by an imbalance of bacteria. Surg Clin and glucocorticoids upon inflammation and collagen
North Am. 1997;77:637–650. synthesis. Ann Surg. 1973;177:222–227.
125. Robson MC, Stenberg BD, Heggers JP. Wound healing 144. Drake DB, Oishi SN. Wound healing considerations in
alterations caused by infection. Clin Plast Surg. chemotherapy and radiation therapy. Clin Plast Surg.
1990;17:485–492. 1995;22:31–37.
126. Robson MC, Krizek TJ. Predicting skin graft survival. 145. Biesecker LG, Happle R, Mulliken JB, et al. Proteus
J Trauma. 1973;13:213–217. syndrome: diagnostic criteria, differential diagnosis,
127. Murphy RC, Robson MC, Heggers JP, et al. The effect and patient evaluation. Am J Med Genet.
of microbial contamination on musculocutaneous and 1999;84:389–395.
random flaps. J Surg Res. 1986;41:75–80. 146. Mirastschijski U, Altmann S, Lenz-Scharf O, et al.
128. Demling RH. Nutrition, anabolism, and the wound Syndromes with focal overgrowth in infancy:
healing process: an overview. Eplasty. 2009;9:e9. Diagnostic approach and surgical treatment. Scand J
129. Posthauer ME. The role of nutrition in wound care. Plast Reconstr Surg Hand Surg. 2009:1–4.
Adv Skin Wound Care. 2006;19:43–52; quiz 53–4. 147. Sapp JC, Turner JT, van de Kamp JM, et al. Newly
130. Barbul A, Purtill WA. Nutrition in wound healing. delineated syndrome of congenital lipomatous
Clin Dermatol. 1994;12:133–140. overgrowth, vascular malformations, and epidermal
131. Mathus-Vliegen EM. Old age, malnutrition, and nevi (CLOVE syndrome) in seven patients. Am J Med
pressure sores: an ill-fated alliance. J Gerontol A Biol Sci Genet A. 2007;143:2944–2958.
Med Sci. 2004;59:355–360. 148. Copcu E. Marjolin’s ulcer: a preventable complication
132. Col C, Soran A, Col M. Can postoperative abdominal of burns? Plast Reconstr Surg. 2009;124:156e–164e.
wound dehiscence be predicted? Tokai J Exp Clin Med. 149. Armour A, Scott PG, Tredget EE. Cellular and
1998;23:123–127. molecular pathology of HTS: basis for treatment.
133. Modolin M, Bevilacqua RG, Margarido NF, et al. Wound Repair Regen. 2007;15(Suppl 1):S6–17.
Effects of protein depletion and repletion on 150. Funayama E, Chodon T, Oyama A, et al. Keratinocytes
experimental open wound contraction. Ann Plast Surg. promote proliferation and inhibit apoptosis of the
1985;15:123–126. underlying fibroblasts: an important role in the
134. Levenson SM, Demetriou AA. Metabolic factors. pathogenesis of keloid. J Invest Dermatol.
Philadelphia: Saunders; 1992. 2003;121:1326–1331.
135. Alcain FJ, Buron MI. Ascorbate on cell growth and 151. Bellemare J, Roberge CJ, Bergeron D, et al. Epidermis
differentiation. J Bioenerg Biomembr. 1994;26:393–398. promotes dermal fibrosis: role in the pathogenesis of
136. Ehrlich HP, Hunt TK. Effects of cortisone and vitamin hypertrophic scars. J Pathol. 2005;206:1–8.
A on wound healing. Ann Surg. 1968;167:324–328. 152. Wassermann RJ, Polo M, Smith P, et al. Differential
137. Masse PG, Pritzker KP, Mendes MG, et al. Vitamin B6 production of apoptosis-modulating proteins in
deficiency experimentally-induced bone and joint patients with hypertrophic burn scar. J Surg Res.
disorder: microscopic, radiographic and biochemical 1998;75:74–80.
evidence. Br J Nutr. 1994;71:919–932. 153. Verhaegen PD, van Zuijlen PP, Pennings NM, et al.
138. Lansdown AB, Mirastschijski U, Stubbs N, et al. Zinc Differences in collagen architecture between keloid,
in wound healing: theoretical, experimental, and hypertrophic scar, normotrophic scar, and normal skin:
clinical aspects. Wound Repair Regen. 2007;15:2–16. An objective histopathological analysis. Wound Repair
139. Goodson 3rd WH, Hunt TK. Deficient collagen Regen. 2009;17:649–656.
formation by obese mice in a standard wound model. 154. Seifert O, Mrowietz U. Keloid scarring: bench and
Am J Surg. 1979;138:692–694. bedside. Arch Dermatol Res. 2009;301:259–272.
140. Marks Jr JG, Cano C, Leitzel K, et al. Inhibition of 155. Tredget EE, Nedelec B, Scott PG, et al. Hypertrophic
wound healing by topical steroids. J Dermatol Surg scars, keloids, and contractures. The cellular and
Oncol. 1983;9:819–821. molecular basis for therapy. Surg Clin North Am.
141. Beer HD, Longaker MT, Werner S. Reduced expression 1997;77:701–730.
of PDGF and PDGF receptors during impaired wound 156. Younai S, Nichter LS, Wellisz T, et al. Modulation of
healing. J Invest Dermatol. 1997;109:132–138. collagen synthesis by transforming growth factor-beta
142. Brauchle M, Fassler R, Werner S. Suppression of in keloid and hypertrophic scar fibroblasts. Ann Plast
keratinocyte growth factor expression by Surg. 1994;33:148–151.
296.e6 • 15 • Skin wound healing: Repair biology, wound, and scar treatment

157. van der Veer WM, Bloemen MC, Ulrich MM, et al. 172. Akintewe TA, Alabi GO. Scleroderma presenting with
Potential cellular and molecular causes of hypertrophic multiple keloids. Br Med J (Clin Res Ed).
scar formation. Burns. 2009;35:15–29. 1985;291:448–449.
158. Bellini A, Mattoli S. The role of the fibrocyte, 173. Hambrick Jr GW, Carter DM. Pachydermoperiostosis.
a bone marrow-derived mesenchymal progenitor, Touraine-Solente-Gole syndrome. Arch Dermatol.
in reactive and reparative fibroses. Lab Invest. 1966;94:594–607.
2007;87:858–870. 174. Teofoli P, Motoki K, Lotti TM, et al.
159. Kendall JC, Li XH, Galli SJ, et al. Promotion of mouse Propiomelanocortin (POMC) gene expression by
fibroblast proliferation by IgE-dependent activation of normal skin and keloid fibroblasts in culture:
mouse mast cells: role for mast cell tumor necrosis modulation by cytokines. Exp Dermatol. 1997;6:111–115.
factor-alpha and transforming growth factor-beta 1. 175. Appleton I, Brown NJ, Willoughby DA. Apoptosis,
J Allergy Clin Immunol. 1997;99:113–123. necrosis, and proliferation: possible implications in the
160. Tredget EE, Iwashina T, Scott PG, et al. Determination etiology of keloids. Am J Pathol. 1996;149:1441–1447.
of plasma Ntau-methylhistamine in vivo by isotope 176. Messadi DV, Doung HS, Zhang Q, et al. Activation of
dilution using benchtop gas chromatography-mass NFkappaB signal pathways in keloid fibroblasts.
spectrometry. J Chromatogr B Biomed Sci Appl. Arch Dermatol Res. 2004;296:125–133.
1997;694:1–9. 177. Bettinger DA, Yager DR, Diegelmann RF, et al. The
161. Werner S, Krieg T, Smola H. Keratinocyte-fibroblast effect of TGF-beta on keloid fibroblast proliferation
interactions in wound healing. J Invest Dermatol. and collagen synthesis. Plast Reconstr Surg.
2007;127:998–1008. 1996;98:827–833.
162. Brigstock DR. Regulation of angiogenesis and 178. Khoo YT, Ong CT, Mukhopadhyay A, et al.
Upregulation of secretory connective tissue growth
endothelial cell function by connective tissue growth
factor (CTGF) in keratinocyte-fibroblast coculture
factor (CTGF) and cysteine-rich 61 (CYR61).
contributes to keloid pathogenesis. J Cell Physiol.
Angiogenesis. 2002;5:153–165.
2006;208:336–343.
163. Yamaguchi Y, Mann DM, Ruoslahti E. Negative
179. Xia W, Phan TT, Lim IJ, et al. Complex epithelial-
regulation of transforming growth factor-beta by the mesenchymal interactions modulate transforming
proteoglycan decorin. Nature. 1990;346:281–284. growth factor-beta expression in keloid-derived cells.
164. Zhang Z, Li XJ, Liu Y, et al. Recombinant human Wound Repair Regen. 2004;12:546–556.
decorin inhibits cell proliferation and downregulates 180. Ong CT, Khoo YT, Tan EK, et al. Epithelial-
TGF-beta1 production in hypertrophic scar fibroblasts. mesenchymal interactions in keloid pathogenesis
Burns. 2007;33:634–641. modulate vascular endothelial growth factor
165. Scott PG, Dodd CM, Ghahary A, et al. Fibroblasts from expression and secretion. J Pathol. 2007;211:95–108.
post-burn hypertrophic scar tissue synthesize less 181. Lim IJ, Phan TT, Bay BH, et al. Fibroblasts cocultured
decorin than normal dermal fibroblasts. Clin Sci (Lond). with keloid keratinocytes: normal fibroblasts secrete
1998;94:541–547. collagen in a keloidlike manner. Am J Physiol Cell
166. Bloom D. Heredity of keloids; review of the literature Physiol. 2002;283:C212–C222.
and report of a family with multiple keloids in five 182. Ehrlich HP, Desmouliere A, Diegelmann RF, et al.
generations. N Y State J Med. 1956;56:511–519. Morphological and immunochemical differences
167. Oluwasanmi JO. Keloids in the African. Clin Plast Surg. between keloid and hypertrophic scar. Am J Pathol.
1974;1:179–195. 1994;145:105–113.
168. Brissett AE, Sherris DA. Scar contractures, 183. Vogt PM, Altintas MA, Radtke C, et al. [Bases and
hypertrophic scars, and keloids. Facial Plast Surg. methods of suturing]. Chirurg. 2009;80:437–444, 446–7.
2001;17:263–272. 184. Vogt PM, Andree C, Breuing K, et al. Dry, moist, and
169. Yagi KI, Dafalla AA, Osman AA. Does an immune wet skin wound repair. Ann Plast Surg. 1995;34:493–
reaction to sebum in wounds cause keloid scars? 499; discussion 499–500.
Beneficial effect of desensitisation. Br J Plast Surg. 185. Svensjo T, Pomahac B, Yao F, et al. Accelerated healing
1979;32:223–225. of full-thickness skin wounds in a wet environment.
Plast Reconstr Surg. 2000;106:602–612; discussion
170. Marneros AG, Norris JE, Olsen BR, et al. Clinical
613–4.
genetics of familial keloids. Arch Dermatol.
2001;137:1429–1434. 186. Schultz GS, Sibbald RG, Falanga V, et al. Wound bed
preparation: a systematic approach to wound
171. Hendrix Jr JD, Greer KE. Rubinstein–Taybi syndrome management. Wound Repair Regen. 2003;11
with multiple flamboyant keloids. Cutis. (Suppl 1):S1–28.
1996;57:346–348.
References 296.e7

187. Chaby G, Senet P, Vaneau M, et al. Dressings for acute 203. Mustoe TA, Cooter RD, Gold MH, et al. International
and chronic wounds: a systematic review. Arch clinical recommendations on scar management. Plast
Dermatol. 2007;143:1297–1304. Reconstr Surg. 2002;110:560–571.
188. Jones V, Grey JE, Harding KG. Wound dressings. Bmj. 204. Poston J. The use of silicone gel sheeting in the
2006;332:777–780. management of hypertrophic and keloid scars. J Wound
189. Argenta LC, Morykwas MJ. Vacuum-assisted closure: a Care. 2000;9:10–16.
new method for wound control and treatment: clinical 205. Wong TW, Chiu HC, Chang CH, et al. Silicone
experience. Ann Plast Surg. 1997;38:563–576; discussion cream occlusive dressing–a novel noninvasive regimen
577. in the treatment of keloid. Dermatology. 1996;192:
190. Genecov DG, Schneider AM, Morykwas MJ, et al. A 329–333.
controlled subatmospheric pressure dressing increases 206. Sawada Y, Sone K. Hydration and occlusion treatment
the rate of skin graft donor site reepithelialization. Ann for hypertrophic scars and keloids. Br J Plast Surg.
Plast Surg. 1998;40:219–225. 1992;45:599–603.
191. Apelqvist J, Armstrong DG, Augustin M, et al. Vacuum 207. Borgognoni L. Biological effects of silicone gel
assisted closure: recommendations for use. A sheeting. Wound Repair Regen. 2002;10:118–121.
consensus document. Int Wound J. 2008;5(Suppl 4):
208. Sawada Y, Sone K. Treatment of scars and keloids with
iii–19.
a cream containing silicone oil. Br J Plast Surg.
192. Morykwas MJ, Simpson J, Punger K, et al. Vacuum- 1990;43:683–688.
assisted closure: state of basic research and physiologic
209. Chowdri NA, Masarat M, Mattoo A, et al. Keloids and
foundation. Plast Reconstr Surg.
hypertrophic scars: results with intraoperative and
2006;117(Suppl):121S–126S.
serial postoperative corticosteroid injection therapy.
193. Silver FH, Siperko LM, Seehra GP. Mechanobiology of Aust N Z J Surg. 1999;69:655–659.
force transduction in dermal tissue. Skin Res Technol.
2003;9:3–23. 210. Parrett BM, Donelan MB. Pulsed dye laser in burn
scars: Current concepts and future directions.
194. Venturi ML, Attinger CE, Mesbahi AN, et al.
Burns. 2009;36:443–449.
Mechanisms and clinical applications of the vacuum-
assisted closure (VAC) Device: a review. Am J Clin 211. Liew SH, Murison M, Dickson WA. Prophylactic
Dermatol. 2005;6:185–194. treatment of deep dermal burn scar to prevent
hypertrophic scarring using the pulsed dye laser:
195. Falanga V. Care of venous leg ulcers. Ostomy Wound
a preliminary study. Ann Plast Surg. 2002;49:
Manage. 1999;45(1A Suppl):33S–43S; quiz 44S-45S.
472–475.
196. Enoch S, Grey JE, Harding KG. ABC of wound healing.
Non-surgical and drug treatments. Bmj. 212. Al-Attar A, Mess S, Thomassen JM, et al. Keloid
2006;332:900–903. pathogenesis and treatment. Plast Reconstr Surg.
2006;117:286–300.
197. Falanga V, Sabolinski M. A bilayered living skin
construct (APLIGRAF) accelerates complete closure of 213. Kauh YC, Rouda S, Mondragon G, et al. Major
hard-to-heal venous ulcers. Wound Repair Regen. suppression of pro-alpha1(I) type I collagen gene
1999;7:201–207. expression in the dermis after keloid excision and
immediate intrawound injection of triamcinolone
198. Schonfeld WH, Villa KF, Fastenau JM, et al. An
acetonide. J Am Acad Dermatol. 1997;37:586–589.
economic assessment of Apligraf (Graftskin) for the
treatment of hard-to-heal venous leg ulcers. Wound 214. Klumpar DI, Murray JC, Anscher M. Keloids treated
Repair Regen. 2000;8:251–257. with excision followed by radiation therapy. J Am Acad
Dermatol. 1994;31:225–231.
199. Ehrenreich M, Ruszczak Z. Update on tissue-
engineered biological dressings. Tissue Eng. 215. O’Kane S, Ferguson MW. Transforming growth factor
2006;12:2407–2424. beta s and wound healing. Int J Biochem Cell Biol.
200. Mirastschijski U, Bugdahl R, Rollman O, et al. 1997;29:63–78.
Epithelial regeneration from bioengineered skin 216. Davison SP, Mess S, Kauffman LC, et al. Ineffective
explants in culture. Br J Dermatol. 2006;154:42–49. treatment of keloids with interferon alpha-2b. Plast
201. Falanga V, Isaacs C, Paquette D, et al. Wounding of Reconstr Surg. 2006;117:247–252.
bioengineered skin: cellular and molecular aspects 217. Occleston NL, O’Kane S, Goldspink N, et al. New
after injury. J Invest Dermatol. 2002;119:653–660. therapeutics for the prevention and reduction of
202. Macri L, Clark RA. Tissue engineering for cutaneous scarring. Drug Discov Today. 2008;13:973–981.
wounds: selecting the proper time and space for 218. Jih MH, Kimyai-Asadi A. Fractional photothermolysis:
growth factors, cells and the extracellular matrix. Skin a review and update. Semin Cutan Med Surg.
Pharmacol Physiol. 2009;22:83–93. 2008;27:63–71.
296.e8 • 15 • Skin wound healing: Repair biology, wound, and scar treatment

219. Aust MC, Fernandes D, Kolokythas P, et al. 228. Taub PJ, Marmur JD, Zhang WX, et al. Locally
Percutaneous collagen induction therapy: an administered vascular endothelial growth factor cDNA
alternative treatment for scars, wrinkles, and skin increases survival of ischemic experimental skin flaps.
laxity. Plast Reconstr Surg. 2008;121:1421–1429. Plast Reconstr Surg. 1998;102:2033–2039.
220. Yager DR, Nwomeh BC. The proteolytic environment 229. Benn SI, Whitsitt JS, Broadley KN, et al. Particle-
of chronic wounds. Wound Repair Regen. mediated gene transfer with transforming growth
1999;7:433–441. factor-beta1 cDNAs enhances wound repair in rat skin.
221. Fray MJ, Dickinson RP, Huggins JP, et al. A potent, J Clin Invest. 1996;98:2894–2902.
selective inhibitor of matrix metalloproteinase-3 for the 230. Andree C, Swain WF, Page CP, et al. In vivo transfer
topical treatment of chronic dermal ulcers. J Med Chem. and expression of a human epidermal growth factor
2003;46:3514–3525. gene accelerates wound repair. Proc Natl Acad Sci U S
222. Makinen K, Manninen H, Hedman M, et al. Increased A. 1994;91:12188–12192.
vascularity detected by digital subtraction angiography 231. Eming SA, Whitsitt JS, He L, et al. Particle-mediated
after VEGF gene transfer to human lower limb artery: gene transfer of PDGF isoforms promotes wound
a randomized, placebo-controlled, double-blinded repair. J Invest Dermatol. 1999;112:297–302.
phase II study. Mol Ther. 2002;6:127–133. 232. Margolis DJ, Morris LM, Papadopoulos M, et al. Phase
223. Steed DL. Clinical evaluation of recombinant human I study of H5.020CMV.PDGF-beta to treat venous leg
platelet-derived growth factor for the treatment of ulcer disease. Mol Ther. 2009;17:1822–1829.
lower extremity diabetic ulcers. Diabetic Ulcer Study 233. Charruyer A, Ghadially R. Stem cells and tissue-
Group. J Vasc Surg. 1995;21:71–78; discussion engineered skin. Skin Pharmacol Physiol. 2009;22:55–62.
79–81. 234. Takahashi K, Tanabe K, Ohnuki M, et al. Induction of
224. Hirsch T, Spielmann M, Yao F, et al. Gene therapy in pluripotent stem cells from adult human fibroblasts by
cutaneous wound healing. Front Biosci. defined factors. Cell. 2007;131:861–872.
2007;12:2507–2518. 235. Schaffler A, Buchler C. Concise review: adipose
225. Vogt PM, Thompson S, Andree C, et al. Genetically tissue-derived stromal cells–basic and clinical
modified keratinocytes transplanted to wounds implications for novel cell-based therapies. Stem Cells.
reconstitute the epidermis. Proc Natl Acad Sci U S A. 2007;25:818–827.
1994;91:9307–9311. 236. Badiavas EV, Falanga V. Treatment of chronic wounds
226. Sun L, Xu L, Chang H, et al. Transfection with aFGF with bone marrow-derived cells. Arch Dermatol.
cDNA improves wound healing. J Invest Dermatol. 2003;139:510–516.
1997;108:313–318. 237. Bunnell BA, Flaat M, Gagliardi C, et al. Adipose-
227. Jeschke MG, Richter G, Hofstadter F, et al. Non-viral derived stem cells: isolation, expansion and
liposomal keratinocyte growth factor (KGF) cDNA differentiation. Methods. 2008;45:115–120.
gene transfer improves dermal and epidermal 238. Duckers HJ, Pinkernell K, Milstein AM, et al. The
regeneration through stimulation of epithelial Bedside Celution system for isolation of adipose
and mesenchymal factors. Gene Ther. 2002;9: derived regenerative cells. EuroIntervention.
1065–1074. 2006;2:395–398.

Das könnte Ihnen auch gefallen