You are on page 1of 12

Clin Plastic Surg 30 (2003) 1 – 12

Acute wound healing


An overview
JoAn L. Monaco, MS, MD, W. Thomas Lawrence, MPH, MD, FACS*
Section of Plastic Surgery, Sutherland Institute, University of Kansas Medical Center, 3901 Rainbow Boulevard,
Kansas City, KS 66160, USA

The past 2 decades have produced more advances Phases of wound healing
in wound care than have the previous 2000 years as
a result of rapid expansion in the knowledge of the Healing of an acute wound follows a predictable
healing process at the molecular level. The coordi- chain of events. This chain of events occurs in a
nated interplay of technology and expanding scien- carefully regulated fashion that is reproducible from
tific knowledge has provided wound-care methods wound to wound. The phases of wound healing are
that have greatly improved the ability to heal overlapping, but are described in a linear fashion for
wounds with few complications. These advances the purpose of clarity. The five phases that characterize
serve as a prelude to the impetus that is likely to wound healing include (1) hemostasis, (2) inflam-
come in the future. mation, (3) cellular migration and proliferation, (4)
Two broad categories exist for the classification of protein synthesis and wound contraction, and (5)
wounds: chronic and acute. Acute wounds undergo a remodeling (Fig. 1).
complex interactive process involving a variety of
cell types that leads to a healed wound. Conversely, Hemostasis
chronic wounds have proceeded through portions of
the repair process without establishing a functional All significant trauma creates a vascular injury and
anatomic result [1]. The present discussion focuses thereby initiates the molecular and cellular responses
on the healing of acute wounds. that establish hemostasis. The healing process cannot
Although the wound-healing process varies among proceed until hemostasis is accomplished. Primary
different tissue types, there are more similarities than contributors to hemostasis include vasoconstriction,
differences between them. In this discussion, skin is platelet aggregation, and fibrin deposition resulting
considered as a representative tissue type. There are from the coagulation cascades. The end product of the
also different types of acute skin wounds, including hemostatic process is clot formation. Clots are pri-
incisional wounds, partial thickness injuries, and marily composed of fibrin mesh and aggregated plate-
wounds involving significant tissue loss. Different lets along with embedded blood cells [2]. The
types of wounds involve different phases of the healing importance of clot formation is profound. This process
process to varying degrees, although the phases them- prevents further fluid and electrolyte loss from the
selves remain the same. wound site and limits contamination from the outside
environment. Fibrin is also the mesh material in the
provisional wound matrix onto which fibroblasts and
other cells migrate as the healing process proceeds.

Vasoconstriction
* Corresponding author. Vasoconstriction is initiated by the release of vaso-
E-mail address: tlawrence@kumc.edu (W.T. Lawrence). active amines, which occurs when the dermis is

0094-1298/03/$ – see front matter D 2003, Elsevier Science (USA). All rights reserved.
PII: S 0 0 9 4 - 1 2 9 8 ( 0 2 ) 0 0 0 7 0 - 6
2 J.L. Monaco, W.T. Lawrence / Clin Plastic Surg 30 (2003) 1–12

Fig. 1. The acute wound-healing cascade. The progression of acute wound healing from hemostasis to the final phases of
remodeling is dependent on a complex interplay of varied acute wound-healing events. Cytokines play a central role in wound
healing and serve as a central signal for various cell types and healing events.

penetrated. Epinephrine is released into the peripheral growth factor-2 (FGF-2), and platelet-derived epi-
circulation, whereas stimulation of the sympathetic dermal growth factors (EGFs), and endothelial cell
nervous system results in local norepinephrine release. growth factors [6]. Of these factors, PDGF, TGF-b,
Injured cells secrete prostaglandins, such as thrombox- and FGF-2 are the most important.
ane, that contribute further to vasoconstriction. Dense bodies contain necessary fuel-providing
compounds that contribute to the healing process.
Platelet aggregation These compounds include calcium, serotonin, ADP,
Platelet aggregation is stimulated by exposure to and ATP. Some of these compounds are also involved
tissue factors released by damaged cells. Platelets in initiation of the coagulation cascades.
adhere to the vascular subendothelium and to each
other in a process involving fibrinogen and von Fibrin and the coagulation cascades
Willebrand factor [3]. Laminin, thrombospondin, and The coagulation cascades are composed of intrinsic
vitronectin may also be involved. As platelets aggre- and extrinsic components that are individually trig-
gate and adhere, they release the contents of alpha gered. The intrinsic cascade is not required for normal
granules, dense bodies, and lysosomes within their healing, whereas the extrinsic cascade is essential [7].
cytoplasm [4]. The intrinsic coagulation cascade is initiated by activa-
Alpha granules contain a variety of immunomodu- tion of factor XII, which occurs when blood is exposed
latory and proteinaceous factors that are involved in to foreign surfaces. The more critical extrinsic coagu-
both the early and late phases of healing. Specifically, lation cascade is initiated by exposure to a ‘‘tissue
these factors include albumin, fibrinogen, fibronectin factor’’ that binds factor VII or factor VIIa. Tissue
[5], IgG, and coagulation factors V and VIII, as well as factor is found on extravascular cell surfaces and, in
platelet-derived growth factor (PDGF), transforming particular, on adventitial fibroblasts. The actions of the
growth factors a and b (TGF-a and TGF-b), fibroblast intrinsic and extrinsic pathways result in the produc-
J.L. Monaco, W.T. Lawrence / Clin Plastic Surg 30 (2003) 1–12 3

tion of thrombin, which catalyzes the conversion of products and mast cell-derived factors such as leuko-
fibrinogen to fibrin. trienes, prostaglandins, and, in particular, histamine
Thrombin itself stimulates increased vascular per- contribute to vasodilation [17,18]. Kinins, released by
meability in addition to facilitating the extravascular protein-binding molecules in the plasma via activa-
migration of inflammatory cells [8]. Fibrin forms the tion of kallikrein, also contribute to this process.
meshwork that stabilizes the platelet plug. It also Capillary leak is primarily mediated by the release
becomes a key component of the provisional matrix of histamine, kinins, and prostaglandins. There is an
that develops in the wound soon after injury. Fibrin additional influence from thrombin and the com-
becomes coated with vitronectin derived from serum plement system. Complement factors C3a and C5a
and aggregating platelets. This action facilitates the are significant contributors to capillary leak and
binding of fibronectins, which are produced by fibro- also act as chemoattractants for neutrophils and
blasts and epithelial cells [5]. Fibronectins are the monocytes [19]. Their chemotactic function is depen-
second key component of the early provisional wound dent on the release of TGF-b and formyl-methionyl
matrix. The fibronectin molecule has nearly a dozen peptides [20].
binding sites for cellular attachment [9]. These attach- Leukocyte migration into the wounded area is
ment sites are essential for cellular migration along the stimulated by collagen, elastin breakdown products,
matrix. The fibrin – fibronectin matrix also traps cir- complement factors, and immunomodulatory factors
culating cytokines for use in the ensuing stages of including TGF-b, tumor necrosis factor-a (TNF-a),
healing [10]. interleukin-1 (IL-1), PDGF, leukotriene B4, and plate-
Inadequate fibrin formation is associated with let factor IV. Leukocytes adhere to endothelial cells
impaired wound healing. This is observed in factor lining the capillaries in the wounded area through an
XIII (fibrin-stabilizing factor) deficiency, which is interaction of intracellular adhesion molecules on the
thought to impair cellular adhesion and chemotaxis endothelial cell membranes and integrins expressed on
[11]. Any process that removes fibrin from the wound the cell surface of the leukocytes. This process is
will disrupt the formation of extracellular matrix and known as margination. Platelet factor IV and platelet-
also, consequently, will delay wound healing [12,13]. activating factor then increase the expression of CD11/
CD18, an integrin on the neutrophil surface that
Inflammation facilitates transmigration of leukocytes through the
endothelium [21] in a process known as diapedesis.
Inflammation is characterized by the erythema, Migrating monocytes transform into macrophages
edema, heat, and pain as first described by Hunter in as they migrate into the extravascular space in a
1794. At the tissue level, increased vascular perme- process that is stimulated by chemotactic factors such
ability and the sequential migration of leukocytes into as fibronectin, elastin derived from damaged matrix,
the extravascular space characterize inflammation [1]. complement components, enzymatically active
One of the primary functions of inflammation is to thrombin, TGF-b, and serum factors [11]. All leuko-
bring inflammatory cells to the injured area [14]. cytes require activation before they can perform their
These cells then destroy bacteria and eliminate debris vital functions in the wound environment [22]. IL-2
from dying cells and damaged matrix so that the and INF-s derived from T lymphocytes are involved
repair processes can proceed [15]. in macrophage activation [23].
Although inflammation is often thought of as the After activation, macrophages and neutrophils
second phase of wound healing, signs of inflam- initiate cellular wound debridement by phagocytosing
mation, including erythema and heat, develop soon bacteria and foreign material [24]. Both cell types
after injury as a result of vasodilatation. Vasodilata- have surface receptors that allow them to recognize,
tion follows the initial vasoconstriction that reverses bind, and engulf foreign material [25]. Macrophage
10 to 15 minutes after injury. Simultaneously, the binding receptors include the immunoglobulin-type
endothelial cells lining the capillaries in the vicinity adhesion molecule CD14 that binds lipopolysaccha-
of the wound develop gaps between them, which ride [26] and the CD11b-CD18 ab integrin. After
permit the leakage of plasma from the intravascular binding, bacteria and debris are engulfed and digested
space to the extravascular compartment [16]. The by oxygen radicals and hydrolytic enzymes within
migration of fluid into the injured area generates the inflammatory cells. In addition to phagocytosing
edema, which contributes to the sensation of pain debris, macrophages also contribute to its breakdown
that characterizes inflammation. extracellularly by releasing matrix metalloproteinases
The transition from vasoconstriction to vasodila- (MMPs) such as collagenase and elastase into the
tion is mediated by a variety of factors. Endothelial wounded area [27].
4 J.L. Monaco, W.T. Lawrence / Clin Plastic Surg 30 (2003) 1–12

In addition to contributing to phagocytosis, mac- ing of the epidermal growth factor receptor. Further-
rophages are also a primary source of cytokines that more, macrophages also release nitric oxide, which
mediate later aspects of the healing process. Some may serve an antimicrobial function as well as other
macrophages primarily phagocytose foreign materials functions during the healing process [28]. The
and produce MMPs, whereas others primarily pro- inhibition of nitric oxide release has been found to
duce cytokines. Unlike polymorphonuclear leuko- impair wound healing in an in vivo mouse model
cytes, the removal of macrophages from the healing [31]. The complete role of nitric oxide in wound
milieu will significantly alter and impede the healing healing has yet to be fully delineated.
process [14]. The presence or absence of polymor- T lymphocytes play a crucial role in the wound-
phonuclear leukocytes will only alter the rate of healing process as well, and the removal of circulat-
wound infection [28]. The added function of cytokine ing T lymphocytes inhibits the healing cascade [32].
production differentiates the activities of the two cell Seventy to eighty percent of normal peripheral blood
types and makes macrophages more essential. lymphocytes are mature T lymphocytes. B lympho-
The role of the macrophage is complex in that cytes contribute the remaining 10% to 15% and have
this multipurpose cell is involved in many aspects of not been found to play any role in wound healing
healing through the cytokines and immunomodula- [33]. Typically, both CD4-positive and CD8-positive
tory factors it produces (Fig. 2). Macrophage-pro- T lymphocytes are present in maximal concentrations
duced cytokines are involved in angiogenesis, 5 to 7 days after injury under the influence of IL-2
fibroblast migration and proliferation, collagen pro- and various other immunomodulatory factors [34,35].
duction [29], and possibly wound contraction. TGF- T lymphocytes—especially CD4-positive T lympho-
b, IL-1, insulin-like growth factor-1 (IGF-1), FGF-2, cytes—are important sources of cytokines including
and PDGF are several of the more critical macro- IL-1, IL-2, TNF-a, fibroblast activating factor, EGF,
phage-derived cytokines. TGF-b regulates its own and TGF-b [36 – 38]. Among other functions, these
production by macrophages in an autocrine manner factors regulate the process of T-cell proliferation and
[30]. It also stimulates macrophages to secrete differentiation in an autocrine fashion. T cells are also
PDGF, FGF-2, TNF-a, and IL-1 [20] through bind- the primary effectors of cell-mediated immunity and

Fig. 2. Cell interactions. Acute wound healing is dependent on a complex interplay of various inflammatory markers and cell
types. Macrophages are crucial to the various phases of acute wound healing and serve as a central stimulator for several different
cell types involved in wound healing.
J.L. Monaco, W.T. Lawrence / Clin Plastic Surg 30 (2003) 1–12 5

subsets of T cells mature into cytotoxic cells capable multiple different activities by single cytokines [46].
of lysing virus-infected and foreign cells [22]. This redundancy is most likely important in that
Other inflammatory cell types include eosinophils different factors may play greater or lesser roles at
and basophils. These leukocytes are nonspecific different time points or at different sites.
amplifiers and effectors of specific immune re- Many cytokines can also stimulate several differ-
sponses [39]. Both eosinophils and basophils in ent cellular functions in a single cell, which are often
unchecked accumulation can lead to host tissue dependent on the concentration of the cytokine. This
damage, as seen in eosinophil-mediated systemic further allows individual cytokines to have varying
necrotizing vasculitis [33]. These cell types reach effects at different points during the healing process.
their highest concentrations in wounds 24 to 48 hours TGF-b, for example, is a macrophage chemoattractant
after injury, and are capable of producing TGF-a when circulating in the fentomolar range, but cannot
[32]. Their complete role in inflammation remains to stimulate collagen synthesis by fibroblasts until found
be delineated. in the nanomolar range [16]. In addition, PDGF
As the healing process proceeds, inflammatory facilitates chemotaxis for fibroblasts, but only at a
cells trapped within clots are sloughed [40]. Neutro- 100-fold greater concentration gradient than is neces-
phils remaining within the wound become senescent sary to stimulate fibroblast proliferation [47,48].
and undergo apoptosis [41]. Apoptosis is character- Cellular activity is also regulated by the balance of
ized by the activation of endogenous calcium- cytokines and cytokine isoforms with conflicting
dependent endonucleases. The activation of these activities. Cytokines such as PDGF, TGF-b, and
endonucleases results in cleavage of chromatin into FGF-2 accelerate collagen synthesis scar formation
oligonucleosome DNA fragments [42], and is indic- [49]. Other cytokines slow collagen synthesis, includ-
ative of irreversible cell death. The stimuli that lead to ing the b3 isoform of TGF-b and INF-a and INF-a2b
inflammatory cell apoptosis during tissue repair and [41]. Cellular activity is, therefore, modulated by the
scar formation have yet to be determined. Neutrophils balance of cytokines with competing functions. Loss
are the first of the inflammatory cells to become of network balance has been implicated in chronic
apoptotic. They are then phagocytosed by macro- wound healing pathways [2].
phages [24]. Macrophages and lymphocytes remain
in the wound for approximately 7 days and then Cellular migration and proliferation
gradually diminish in number unless a noxious stimu-
lant of further inflammation persists. Inflammatory The cellular milieu in wounds changes dramat-
cell apoptosis influences antigen presentation and, ically in the first week post acute injury. The initial
probably more importantly, contributes to modulation fibrin – fibronectin matrix is heavily populated by
of cytokine concentrations [16]. inflammatory cells, whereas fibroblasts and endothe-
lial cells will predominate as healing progresses. Re-
Cytokines establishment of the epithelial surface is also initiated
within the first several days after injury, as is revas-
The wound-healing process is, in large part, cularization of the damaged area. Cytokine networks
regulated by the ordered production of cytokines that continue to be a part of the process as cytokine
control gene activation responsible for cellular migra- release contributes to fibroplasia, epithelialization,
tion and proliferation and synthetic activities. As and angiogenesis [44]. Although much is known
mentioned, platelets and macrophages are key cyto- about the signals that stimulate the predominant
kine sources, although many other cells produce activities during this phase of healing, less is known
them. Control of the release of cytokines is, in part, about the signals that bring these activities to a
regulated by other cytokines and, in part, regulated controlled end. Negative feedback mechanisms that
by characteristics of the tissue milieu [43,44]. An deactivate cells after they have completed their work
often underreported signal for cytokine release is are also essential for normal healing.
tissue hypoxia. Hypoxia has been shown to stimulate Additional fibroblasts are required in the healing
the release of TNF-a, TGF-b, vascular endothelial wound, in that native cells are lost or damaged in any
growth factor (VEGF), and IL-8 (IL-8) from fibro- injury. Repopulation of the wounded area with fibro-
blasts, endothelial cells, and macrophages [2,36, blasts occurs as a result of fibroblast migration from
37,45]. adjacent tissues and proliferation of cells in the wound.
‘‘Cytokine networks’’ exist where multiple cell In addition, undifferentiated cells in the vicinity of the
types involved in the healing process have receptors wound may transform into fibroblasts under the influ-
for the same cytokine. This permits stimulation of ence of cytokines in the wound milieu [50].
6 J.L. Monaco, W.T. Lawrence / Clin Plastic Surg 30 (2003) 1–12

Factors that stimulate fibroblast migration include During angiogenesis, endothelial sprouts derive
PDGF [48], TGF-b [50], EGF [51], and fibronectin from intact capillaries at the wound periphery [62].
[18]. Upregulation of cell membrane integrin recep- The sprouts grow through cellular migration and
tors that bind fibronectin and fibrin in the provisional proliferation. The endothelial cells develop a cur-
wound matrix is required for fibroblasts to migrate vature and begin to produce a lumen as the chain of
[52,53], and PDGF [54,55] and TGF-b [50] both endothelial cells elongates. Eventually, the endothe-
contribute to the migratory process in this manner. lial sprout comes into contact with a sprout derived
Integrin expression is, therefore, vital to the migra- from a different capillary, and they interconnect
tion of fibroblasts and other cell types. There are over generating a new capillary.
20 different integrin molecules, and most have a and b Endothelial migration during angiogenesis in-
subunits [21]. Different cells express different integ- volves binding integrin domains within the provi-
rins, and individual cells can often express more than sional fibrin – fibronectin matrix in a similar manner
one integrin, often under the influence of varied to fibroblasts. Upregulation of ab3 integrins is spe-
cytokines. Cellular migration requires cell mem- cifically associated with angiogenesis. Endothelial
brane-bound integrins to be bound to fibronectin in cell migration is also facilitated by the cell’s ability
the extracellular matrix [56]. A migrating cell then to produce MMPs that break down collagen and
develops lamellopodia that extend outward until plasminogen activator, which facilitates movement
another binding site is detected in the matrix [57]. through the matrix.
By releasing the primary binding site and pulling itself As mentioned, the angiogenic process is regulated
toward the second site, the cell migrates, using the new by a variety of cytokines. The two most important
site as an anchor [17]. Fibronectin fragments stimulate cytokines that contribute to angiogenesis are FGF-2
this migratory process [18]. The orientation of fibers in [63]—which has also been known as basic fibroblast
the matrix also influences cellular migration, in that growth factor—and VEGF [64]. Heparin is a neces-
cells tend to migrate along fibers and not across them. sary cofactor for FGF-2 [65]. Cytokine concentra-
The ability of fibroblasts to migrate may be tions diminish as the wounded area becomes
impeded by residual debris in the wound envi- revascularized, and this flux in angiogenic cytokines
ronment. To facilitate migration through such debris, may facilitate maturation of the vascular system.
fibroblasts secrete several proteolytic enzymes
including MMP-1, gelatinase (MMP-2), and strome- Epithelialization
lysin (MMP-3) [58,59]. TGF-b stimulates fibroblasts Following acute injury, reconstruction of injured
to secrete these enzymes [30,60,61]. epithelium is crucial for re-establishment of the barrier
The proliferation of residual fibroblasts in the functions of the skin. Reconstruction of injured epi-
wounded area, as well as fibroblasts that have migrated thelium begins almost immediately after wounding.
to it, is regulated by a variety of cytokines. PDGF and Incisional skin injuries, with a minimal epithelial gap,
TGF-b are two of the most important cytokines are typically re-epithelialized within 24 to 48 hours
involved in this process. PDGF often works in concert after initial injury [2,66], although larger wounds can
with IGF to facilitate fibroblast proliferation [48]. take much longer to regenerate a neoepithelium.
PDGF primarily stimulates cells to progress through During the first 24 hours after injury, basal cells
the early G0 and G1 phases of the cell cycle [47]. IGF, present at the wound edge elongate and begin to
which is derived primarily from hepatocytes and migrate across the denuded wound surface. If the
fibroblasts [37], then facilitates progression through initial injury does not destroy epithelial appendages
the subsequent S1, G2, and M phases of the cell cycle. such as hair follicles and sweat glands, these struc-
tures also contribute migratory epithelial cells to the
Angiogenesis healing process. These cells migrate across the
The angiogenic process becomes active from day 2 wounded area essentially as a monolayer. Approx-
after wounding [39]. Factors in the wound milieu that imately 24 hours after the initiation of cellular migra-
contribute to angiogenesis include high lactate levels, tion, basal cells at the wound edge and in the
acidic pH, and, in particular, decreased oxygen tension appendages, if present, begin to proliferate, contrib-
[35]. The severe degree of hypoxia in granulation uting additional cells to the healing monolayer. The
tissue most likely results from both disruption of the migration of epithelial cells continues until overlap is
native vasculature and increased oxygen consumption achieved with other epithelial cells migrating from
by cells in the wound environment [40]. Proliferating different directions. At that point, ‘‘contact inhibi-
cells are known to consume oxygen three to five times tion’’ results in cessation of cellular migration. The
faster than do cells in resting phases of the cell cycle. processes of cellular migration and proliferation
J.L. Monaco, W.T. Lawrence / Clin Plastic Surg 30 (2003) 1–12 7

occur under the control of various cytokines includ- synthesis continues at a maximal rate for 2 to 4 weeks
ing EGF [66,67], TGF-a, platelet-derived EGF, and and subsequently begins to slow.
keratinocyte growth factor (KGF, also known as Healing aberrations are often the result of aberra-
FGF-7) [68]. Some derive from inflammatory cells tions in collagen deposition, although the underlying
and others derive from the epithelial cells themselves. causes may vary. In diabetes, impaired activation of
Cellular migration may also require the secretion of inflammatory cells coupled with deficiencies in other
MMPs to penetrate eschar or scab [69]. aspects of the healing process result in limited col-
Epithelial cell migration requires the development lagen deposition and impaired healing [77]. Con-
of actin filaments within the cytoplasm of migratory versely, keloid formation results from excessive
cells and the disappearance of desmosomes and collagen synthesis for which a preventative measure
hemidesmosomes that link them to one another and has yet to be found [78].
to the basement membrane, respectively. At least As mentioned, the initial wound matrix is com-
some of these processes are dependent on changes posed primarily of fibrin and fibronectin. As protein
in integrins expressed on the cell membranes [70]. It synthesis accelerates, the nature of the wound matrix
is thought that decreased calcium or increased mag- changes. Collagen and other proteins such as proteo-
nesium concentrations stimulate the downregulation glycans gradually replace fibrin as primary matrix
of the critical integrins, although the precise signal is constituents. Proteoglycans are a key component of
not yet known [56]. mature matrix and are actively synthesized during
If the epidermal basement membrane is intact, this phase of healing. Additional proteins such as
cells simply migrate over it. In wounds in which it thrombospondin I and SPARC (secreted protein
has been destroyed, the cells initially begin to mi- acidic rich in cysteine)—which support cellular
grate over the fibrin – fibronectin provisional matrix recruitment and stimulate wound remodeling—are
[12,13]. As they migrate across the matrix, however, also produced and are found in the mature wound
epithelial cells regenerate a new basement membrane. matrix as well [79].
Re-establishment of a basement membrane under the Collagen makes up 25% of protein in the body
migrating cells involves the secretion of tenasin, and more than 50% of protein in scar tissue [74].
vitronectin, and type I and V collagens [71]. The concentration of collagen subtypes varies among
When contact inhibition is achieved, hemides- tissues. Type I collagen predominates and makes up
mosomes re-form between the cells and basement 80% to 90% of the collagen seen in intact dermis.
membrane, and tenasin and vitronectin secretion The remaining 10% to 20% is type III collagen. In
diminishes. The cells become more basaloid [72], contrast, granulation tissue that forms soon after
and further cellular proliferation generates a multi- injury contains 30% type III collagen. Accelerated
laminated neoepidermis covered by keratin. The neo- type III collagen synthesis is correlated with fibro-
epidermis is similar to the native epidermis, although nectin secretion after injury [75]. Type II collagen is
it is slightly thinner, the basement membrane is seen almost exclusively in cartilage, whereas type IV
flatter, and rete pegs that normally penetrate the collagen is found in basement membranes. Type V
dermis are absent. collagen is found in blood vessels, whereas type VII
collagen forms the anchoring fibrils of epidermal
Protein synthesis and wound contraction basement membrane [80].
Type I collagen consists of a triple helix involving
Synthesis and deposition of proteins and wound three polypeptide chains that are synthesized sepa-
contraction are the wound-healing events that begin rately within the fibroblast. The polypeptide chains
to predominate 4 to 5 days after wounding. The consist of a repeating glycine-X-Y pattern, in which
quality and quantity of matrix deposited during this the X position is often proline and the Y position is
phase of healing significantly influence the strength often hydroxyproline. The interaction of chains ini-
of a scar [73]. Collagen constitutes more than 50% of tiates the formation of the triple helix, which is
the protein in scar tissue, and its production is secreted as ‘‘procollagen’’ into the extracellular envi-
essential to the healing process [74]. Fibroblasts are ronment [81].
responsible for the synthesis of collagen and other Collagen undergoes eight posttranslational steps
proteins regenerated during the repair process. Col- intracellularly prior to its extracellular secretion in
lagen synthesis is stimulated by TGF-b, PDGF, and the form of procollagen [29]. A critical step involves
EGF [75]. Collagen synthesis is also affected by hydroxylation of proline and lysine moieties within
characteristics of the patient and the wound including the polypeptide chains. Hydroxylation of proline and
age, tension, pressure, and stress [76]. Collagen lysine requires specific enzymes and the cofactors
8 J.L. Monaco, W.T. Lawrence / Clin Plastic Surg 30 (2003) 1–12

oxygen, vitamin C, ferrous iron, and a ketoglutarate. between anatomic locations, but averages approxi-
Hydroxyproline is an important marker of the quan- mately 0.6 to 0.7 mm per day. The rate of contraction
tity of collagen within tissues in that it is almost can often be predicted by the degree of skin laxity at
exclusively found in collagen. Hydroxylysine is an the wound site. A wound on the scalp or pretibial area
essential element for cross-link formation both within will contract significantly more slowly than a buttock
and between collagen molecules [81]. Deficiencies in wound. Wound shape also affects the rate of contrac-
vitamin C (scurvy) or suppression of enzymatic tion, with square wounds contracting more quickly
activity by corticosteroids can lead to underhydroxy- than circular wounds. Circular stomas are therefore
lated collagen that is incapable of generating strong less likely to have compromised patency secondary
cross-links. to contraction.
Procollagen-C proteinases and procollagen-N pro- Wound contraction is characterized by a predom-
teinases cleave the propeptide ends of the procollagen inance of myofibroblasts at the wound periphery.
molecules after they have been secreted into the Myofibroblasts are modified fibroblasts that were
extracellular space. This decreases the solubility of initially described by Gabbiani et al in 1971 [83].
the molecules and initiates the formation of fibrils. The defining characteristics of myofibroblasts include
During fibril formation, collagen molecules are ini- actin-rich microfilaments in the cytoplasm, a multi-
tially linked together by electrostatic bonds. Sub- lobulated nucleus, and abundant rough endoplasmic
sequent to this, free amino groups on lysine and reticulum that can only be discerned by electron
hydroxylysine residues within the collagen molecules microscopy. The time frame in which myofibroblasts
are transformed to aldehyde residues by the enzyme are present within the wound does not correspond
lysyl oxidase. The aldehyde residues interact with perfectly to the course of wound contraction, although
nontransformed lysine or hydroxlysine residues on it is fairly close. Myofibroblasts appear 4 to 6 days
adjacent molecules, resulting in the formation of after initial injury and are commonly seen in the
stable covalent cross-links between the molecules wound during the ensuing 2 to 3 weeks. Their dis-
[35]. These cross-links stabilize the conjoined col- appearance is suspected to be via apoptosis. Although
lagen molecules as fibrils and fibers. Gabbiani et al [83] postulated that these cells were the
As mentioned, proteoglycans are also normal der- ‘‘motor’’ that contracted a wound, more recent work
mal matrix proteins that are synthesized by fibroblasts with collagen lattices has suggested that fibroblasts in
after injury. Their concentration in injured tissue the central portion of the wound may be more critical
gradually increases with time in a manner paralleling to the contraction process [42]. It is clear, however,
collagen. Proteoglycans consist of a protein core that the process of wound contraction is cell mediated
covalently linked to one or more glycosaminoglycans and does not require collagen synthesis. TGF-b and
[82]. Proteoglycans bind proteins and alter their ori- possibly other cytokines are involved in the wound
entation in a manner that influences their activity. contraction process [84].
Dermatan sulfate is a proteoglycan that orients col- Wound contraction is sometimes not a desirable
lagen molecules in a manner that facilitates fibril healing event. Wound contraction across joints can
formation. Hyaluronan, another proteoglycan, contrib- produce contractures that significantly limit function.
utes to skin’s viscoelastic properties and acts as a In cases in which contraction inhibition is preferred,
potent modulator of cellular migration [82]. skin grafting, especially with thicker grafts, is used to
Elastin is another component of wound matrix that limit contraction. Splints can also limit undesirable
provides elasticity to normal skin. It is not synthesized contraction in certain anatomic locations if utilized
in response to injury and is subsequently not found in for prolonged periods.
scar. This lack of elastin in scar tissue contributes to the
increased stiffness and decreased elasticity of scar as Remodeling
compared with normal dermis.
Wound contraction begins 4 to 5 days after initial Scar remodeling begins to predominate as the
injury and actively continues for approximately primary wound-healing activity approximately
2 weeks. The process continues for a longer period 21 days after injury. The rate of collagen synthesis
of time in wounds that remain open at the end of the diminishes and reaches coincidence with the rate of
2-week interval. In an open wound, the results of collagen breakdown. The downregulation of collagen
wound contraction are evident because wound edges synthesis is mediated by g-interferon [85], TNF-a
draw closer to each other. In an incisional wound, [86], and collagen matrix itself [29].
wound contraction simply results in scar shortening Matrix metalloproteinases (MMPs) are intimately
and is less apparent. The rate of contraction varies involved with the breakdown of collagen molecules
J.L. Monaco, W.T. Lawrence / Clin Plastic Surg 30 (2003) 1–12 9

that occurs actively during the remodeling process. have the ability to regenerate differentiated structures.
The MMPs have been alluded to previously and are Technology and increased scientific knowledge have
involved in many aspects of the healing process. established a coordinated interplay that has improved
MMPs represent a family of at least 25 enzymes that the ability to manage wounds in a logical manner, and,
break down different extracellular matrices [58 – 60]. on occasion, to accelerate the healing process. Insight
They are produced by a variety of cell types, and into the complex chain of events leading to the
different cells generally synthesize different enzymes. formation of scar is a necessity for every individual
The MMP activity within tissues is modulated by who attempts wound management.
tissue inhibitors of metalloproteinases (TIMPs) [87].
Four isoforms of TIMPs have been described [87].
The balance of MMPs and TIMPs within tissues is References
critical to enzyme activity and is regulated by cyto-
kines including TGF-b, PDGF, and IL-1 [43,61,88]. [1] Cohen IK, Diegelman RF, Dome RY, et al. Wound care
All of the functions provided by MMPs during and wound healing. In: Schwartz SI, Shires GT, Spencer
wound healing have not been clearly delineated. FC, et al, editors. Principles of surgery. seventh edition.
Elevated concentrations of MMPs are seen in chronic New York: McGraw-Hill; 1999. p. 263 – 95.
ulcers [77], yet deletion of MMP-3 in mice causes [2] Lawrence WT. Physiology of the acute wound. Clin
failure of wound contraction with a significant delay Plas Surg 1998;25(3):321 – 40.
[3] Cines DB, Pollack ES, Buck CA, et al. Endothelial
in healing [73], thereby implicating a complex regu-
cells in physiology and in the pathophysiology of vas-
latory pathway. cular disorders. Blood 1998;91:3527 – 61.
The nature of the wound matrix changes with scar [4] White JD, Gerrard JM. Ultrastructural features of abnor-
remodeling. Immature scar contains a disorganized mal platelets: a review. Am J Pathol 1976;83:589 – 632.
array of fine collagen fibers, which is gradually [5] Hynes RO. Fibronectins. Sci Am 1986;254:42 – 61.
replaced by thicker fibers arranged in an orientation [6] Deuel TF, Senior RM, Huang JS, et al. Chemotaxis of
paralleling skin stresses. In addition, the number of monocytes and neutrophils to platelet-derived growth
cross-links both within and between molecules gradu- factor. J Clin Invest 1982;69:1046 – 9.
ally increases. As the nature of the collagen matrix [7] Dahlback B. Blood coagulation. Lancet 2000;355:
changes, it becomes less cellular through apoptosis of 1627 – 32.
[8] Stiernbery J, Redin WR, Warner WS, et al. The role of
cells involved in the healing process. As mentioned,
thrombin and thrombin receptor activating peptide
the ratio of type I to type III collagen changes, and the (TRAP-508) in initiation of tissue repair. Thromb Hae-
quantity of water and proteoglycans diminish. Nor- most 1993;70:158 – 62.
mal skin shows a basketlike weave pattern that is [9] Grinnell F. Fibronectin and wound healing. J Cell Bio-
never completely reproduced with scar remodeling. chem 1984;26:107 – 16.
Although seemingly not as complex as other [10] Blatti SP, Foster DN, Ranganathan G, et al. Induction of
aspects of the healing process, remodeling is essential fibronectin gene transcription and mRNA is a primary
to the formation of a strong wound. The remodeling response to growth-factor stimulation of AKR-2B cells.
process is associated with a substantial increase in Proc Natl Acad Sci USA 1988;85:1119 – 23.
wound-breaking strength. Wound strength 1 week [11] Fukai F, Suzuki H, Suzuki K, et al. Rat plasma fibro-
nectin contains two distinct chemotactic domains for
after injury is 3% of normal dermis. After 3 weeks,
fibroblastic cells. J Biol Chem 1991;266:8807 – 13.
when the remodeling phase begins to predominate, the [12] Clark RA, Lanigan JM, DellaPelle P, et al. Fibronectin
wound will have only approximately 20% the strength and fibrin provide a provisional matrix for epidermal
of normal dermis. At 3 months, however, the wound cell migration during wound re-epithelialization.
will have 80% the strength of normal dermis, with the J Invest Dermatol 1982;79:264 – 9.
significant increase in strength resulting from the [13] Wysocki AB, Grinell F. Fibronectin profiles in nor-
contribution of remodeling. Remodeling will continue mal and chronic wound fluid. Lab Invest 1990;63:
for up to 12 months after a wound is created, although 825 – 31.
scars never regain the strength of normal dermis. [14] Kloth LC, McCulloch JM, Feedar JA. Wound healing.
In: Kloth LC, McCulloch JM, Feedar JA, editors.
Wound healing: alternatives in management. Philadel-
phia: FA Davis Company; 1990. p. 3 – 13.
Summary [15] Majno G, Shea SM, Leventhal M. Endothelial contrac-
tion induced by histamine type mediators: an electron
The ability to heal an injury is a biologic necessity microscopic study. J Cell Biol 1969;42:647 – 72.
for all organisms, with mammals lagging in pro- [16] Cohen IK, Diegelman RF, Dorne RY, et al. Wound heal-
ficiency when compared with lower life forms that ing. In: Greenfield LJ, Mulholland MW, Oldham KT,
10 J.L. Monaco, W.T. Lawrence / Clin Plastic Surg 30 (2003) 1–12

et al, editors. Surgery: scientific principles and practice. modulation of fibroblast proliferation. J Immunol
third edition. Philadelphia: Lippincott-Raven; 1993. 1982;128:1484 – 6.
p. 86. [35] Witte MB, Barbul A. Wound healing: general principles
[17] Carter SB. Cell movement and cell spreading: a pas- of wound healing. Surg Clin N Am 1997;77:509 – 28.
sive or an active process? Nature 1970;255:858 – 9. [36] Minchenko A, Salceda S, Bauer T, Caro J. Hypoxia
[18] Postlethwaite AE, Keski-Oja J, Ballan G, et al. Induc- regulatory elements of the human vascular endothelial
tion of fibroblast chemotaxis by fibronectin. J Exp Med growth factor gene. Cell Mol Biol Res 1994;40:35 – 9.
1981;153:494 – 9. [37] Patel B, Khaliq A, Jarvis-Evans J, et al. Oxygen regu-
[19] Glat PM, Longaker MT. Wound healing. In: Aston SJ, lation of TGF-beta 1 mRNA in human hepatoma (Hep
Besley RW, Thorn CH, editors. Grabb and Smith’s G2) cells. Biochem Mol Biol Int 1994;34:639 – 44.
plastic surgery. fifth edition. Philadelphia: Lippincott- [38] Schreiber AB, Winkler ME, Derynck R. Transforming
Raven; 1997. p. 3. growth factor type alpha: a more potent angiogenic
[20] Wahl SM, Hunt DA, Wakefield LM, et al. Transform- mediator than epidermal growth factor. Science 1986;
ing growth factor type beta induces monocyte chemo- 232:1250 – 3.
taxis and growth factor production. Proc Natl Acad Sci [39] Grotendorst GR, Pencev D, Martin GR, et al. Molec-
USA 1987;84:5788 – 92. ular mediators of tissue repair. In: Hunt TK, Heppen-
[21] Hynes RO. Integrins: a family of cell surface receptors. stall RB, Pines E, et al, editors. Soft and hard tissue
Cell 1987;48:549 – 54. repair: biological and clinical aspects. New York:
[22] Fauci AS, Haynes BF. Cellular and molecular basis of Praeger; 1984. p. 20.
immunity. In: Issclacher K, Braunwald E, Wilson J, [40] Gailit J, Clark RA. Wound repair in the context of ex-
et al, editors. Harrison’s principles of internal medicine. tracellular matrix [review]. Curr Opin Cell Biol 1994;
13th edition. New York: McGraw-Hill; 1994. p. 1552. 6:717 – 25.
[23] Ono I. Roles of cytokines in wound healing processes. [41] Simpson DM, Ross R. The neutrophilic leukocyte in
Nippon Geka Gakki Zasshi 1999;100:522 – 8. wound repair: a study with antineutrophil serum. J Clin
[24] Newman SL, Henson JE, Henson PM. Phagocytosis of Invest 1972;51:2009 – 23.
senescent neutrophils by human monocyte derived [42] Erlich HP. The role of connective tissue matrix in wound
macrophages and rabbit inflammatory macrophages. healing. In: Hunt TK, editor. Growth factors and other
J Exp Med 1982;156:430 – 42. aspects of wound healing: biological and clinical impli-
[25] Ezekowitz RA, Williams DJ, Koziel H, et al. Uptake of cations. Alan R Liss, Inc.; 1988. p. 243 – 58.
Pneumocystis carnii mediated by the macrophage [43] Circolo A, Welgus HG, Pierce GF, et al. Differential
mannose receptor. Nature 1991;351:155 – 8. regulation of the expression of proteinases/antiprotei-
[26] Wright SD, Ramos RA, Tobias PS, Ulevitch RJ, nases in fibroblasts: effects of interleukin-1 and plate-
Matchison JC. CD14, a receptor for complexes of lip- let-derived growth factor. J Biol Chem 1991;266:
opolysaccharide (LPS) and LPS-binding protein. Sci- 12283 – 8.
ence 1990;249:1431 – 3. [44] Fauci AS. Resistance to HIV-1 infection: its in the
[27] Werb Z, Banda MJ, Jones PA. Degradation of connec- genes. Nat Med 1996;2:966 – 7.
tive tissue matrices by macrophages: proteolysis of elas- [45] Scannell G, Waxman K, Kaml GJ, et al. Hypoxia in-
tin, glycoproteins and collagen by proteinases isolated duces a human macrophage cell line to release tumor
from macrophages. J Exp Med 1980;152:1340 – 52. necrosis factor-alpha and its soluble receptors in vitro.
[28] Malawista SE, Montgomery RR, van Blaricom G. Evi- J Surg Res 1993;54(4):281 – 5.
dence for reactive nitrogen intermediates in killing of [46] Gharaee-Kermani M. Role of cytokines and cytokine
staphylococci by human neutrophil cytoplasts. A new therapy in wound healing and fibrotic disease. Curr
microbicidal pathway for polymorphonuclear leuko- Pharm Des 2001;7(11):1083 – 103.
cytes. J Clin Invest 1992;90:631 – 6. [47] Ross R. Platelet-derived growth factor. Ann Rev Med
[29] Madden JW, Peacock Jr EE. Studies on the biology of 1987;38:71 – 9.
collagen during wound healing: rate of collagen syn- [48] Seppä H, Grotendorst GR, Seppä S, et al. Platelet-de-
thesis and deposition in cutaneous wounds of the rat. rived growth factor in chemotactic for fibroblasts. J Cell
Surgery 1968;64:288 – 94. Biol 1982;92:584 – 8.
[30] McCartney-Francis N, Mizel D, Wong H, et al. Trans- [49] Roberts AB, Anzano MA, Wakefield LM, et al. Type
forming growth factor-beta (TGF-b) as an immunore- beta transforming growth factor: a bifunctional regula-
gulatory molecule [abstract]. FASEB J 1988;2:A875. tor of cellular growth. Proc Natl Acad Sci USA 1985;
[31] Schaffer MR, Tandry U, Gross SS, et al. Nitric oxide 82:119 – 23.
regulates wound healing. J Surg Res 1996;63:237 – 40. [50] Postlethwaite AE, Keski-Oja L, Moses HL, et al. Stim-
[32] Peterson JM, Barbul A, Breslin RJ, et al. Significance ulation of the chemotactic migration of human fibro-
of T-lymphocytes in wound healing. Surgery 1987; blasts by transforming growth factor-b. J Exp Med
102:300 – 5. 1987;165:251 – 6.
[33] Martin CW, Muir IF. The role of lymphocytes in [51] Westermark B, Blomquist W. Stimulation of fibroblast
wound healing. Br J Plast Surg 1990;43:655 – 62. migration by epidermal growth factor. Cell Biol Int
[34] Nielson EG, Phillips SM, Jimeney S. Lymphokine Rep 1980;4:649 – 54.
J.L. Monaco, W.T. Lawrence / Clin Plastic Surg 30 (2003) 1–12 11

[52] Hynes RO. Integrins: versatility, modulation and sig- duction of keratinocyte growth factor expression in the
naling in cell adhesion. Cell 1992;69:11. dermis during wound healing. Proc Natl Acad Sci USA
[53] Singer II, Scott S, Kawka DW, et al. Cell surface 1992;89:6896 – 900.
distribution of fibronectin and vibronectin recep- [69] Vu TH, Werb Z. Matrix metalloproteinases: effectors
tors depends on substrate composition and extrac- of development and normal physiology. Genes Dev
ellular matrix accumulation. J Cell Biol 1988;106: 2000;14:2123 – 33.
2171 – 82. [70] Cavani A, Zambruno G, Marconi A, et al. Distinctive
[54] Ahlen K, Rubin K. Platelet-derived growth factor-BB integrin expression in the newly formed epidermis dur-
stimulates synthesis of the integrin alpha 2-subunit ing wound healing in humans. J Invest Dermatol 1993;
in human diploid fibroblasts. Exp Cell Res 1994; 101:600 – 4.
215:347. [71] Mackie EJ, Halfter W, Liverani D. Induction of tenas-
[55] Gailit J, Bueller H, Clark R. Platelet-derived growth cin in healing wounds. J Cell Biol 1988;107:2757 – 67.
factor and inflammatory cytokines have differential ef- [72] Odland G, Ross R. Human growth repair: epidermal
fects on the expression of integrins alpha1b1 and alpha regeneration. J Cell Biol 1968;39:135 – 51.
5b1 by human dermal fibroblasts. J Cell Physiol 1996; [73] Bullard KM, Lund L, Mudgett JS, et al. Impaired
169:281 – 9. wound contraction in stromelysin-1 deficient mice.
[56] Grzesiak HH, Piershbacher MD. Shifts in concentra- Ann Surg 1999;230:260 – 5.
tions of magnesium and calcium in early porcine and [74] Nimmi ME. Collagen: its structure and function in
rat wound fluids activate the cell migratory response. normal and pathological connective tissues. Semin Ar-
J Clin Invest 1995;95:227 – 33. thritis Rheum 1974;4:95 – 150.
[57] Reed MJ, Puolakkainen P, Lane TF, et al. Differ- [75] Ignotz RA, Massaugue J. Transforming growth factor-
ential expression of SPARC and thrombospondin 1 beta stimulates the expression of fibronectin and colla-
in wound repair: immunolocalization and in situ gen and their incorporation into the extracellular ma-
hybridization. J Histochem Cytochem 1993;41: trix. J Biol Chem 1986;261:4337 – 45.
1467 – 77. [76] Caterson B, Lowther DA. Changes in the metabolism
[58] Parks WC. Matrix metalloproteinases in repair. Wound of the proteoglycans from sheep articular cartilage in
Repair Reg 1999;7:423 – 33. response to mechanical stress. Biochem Biophys Acta
[59] Parsons SL, Watson SA, Brown PD, et al. Matrix met- 1978;540:412 – 22.
alloproteinases. Br J Surg 1997;84:160 – 6. [77] Fahey III TJ, Sadaty A, Jones II WG, et al. Diabetes
[60] Assoian RK, Sporn MB. Type beta transforming impairs the late inflammatory response to wound heal-
growth factor in human platelets: release during degra- ing. J Surg Res 1991;50:308 – 13.
nulation and action on vascular smooth muscle cells. [78] Ketchum LD, Smith J, Robinson DW, et al. Treatment
J Cell Biol 1986;102:1217 – 23. of hypertrophic scars, colloids and scar contractor by
[61] Werb Z, Tremble P, Damsky CH. Regulation of extrac- triamcinolone acetonide. Plast Reconstr Surg 1966;
ellular matrix degradation by cell – extracellular matrix 38:209 – 18.
interactions. Cell Differ Dev 1990;32:299 – 306. [79] Reed MJ, Puolakkainen P, Lane TF, et al. Differ-
[62] Folkman J, Klagsbrun M. Angiogenic factors. Science ential expression of SPARC and thrombospondin I
1987;235:442 – 7. in wound repair: immunolocalization and in situ
[63] Gospodarowicz D, Neufeld G, Schweigerer L. Fibro- hybridization. J Histochem Cytochem 1993;41:
blast growth factor: structural and biologic properties. 1467 – 77.
J Cell Physiol 1987;5(Suppl 15):15 – 26. [80] Hering TM, Marchant RE, Anderson JM. Type V col-
[64] Gospodarowicz D, Abraham J, Schilling J. Isolation lagen during granulation tissue development. Exp Mol
and characterization of a vascular endothelial cell Pathol 1983;39:219 – 29.
mitogen produced by pituitary-derived follicular [81] Veis A, Averey J. Modes of intermolecular crosslinking
stellate cells. Proc Natl Acad Sci USA 1989;86: in mature insoluble collagen. J Biol Chem 1965;
7311 – 5. 240:3899 – 908.
[65] Shing Y, Folkmann J, Sullivan R, et al. Heparin [82] Hassel JR, Kimura JH, Hascall VC. Proteoglycan core
affinity: purification of a tumor-derived capillary en- protein families. Ann Rev Biochem 1986;55:539 – 67.
dothelial cell growth factor. Science 1984;223: [83] Gabbianni G, Ryan GB, Majno G. Presence of modi-
1296 – 9. fied fibroblasts in granulation tissue and their possi-
[66] Nanney LB, Magid M, Stoscheck CM, et al. Com- ble role in wound contraction. Experientia 1971;27:
parison of epidermal growth factor binding and re- 549 – 50.
ceptor distribution in normal human epidermis and [84] Clark RA, Nielsen LD, Welch MP, McPherson JM. Col-
epidermal appendages. J Invest Dermatol 1984;83: lagen matrices attenuate the collagen synthetic response
385 – 93. of cultured fibroblasts to TGF-b. J Cell Sci 1995;
[67] Niall M, Ryan GB, O’Brien BM. The effect of epider- 108:1251 – 61.
mal growth factor on wound healing in mice. J Surg [85] Granstein RD, Murphy GF, Margolis RJ, et al. Gamma
Res 1982;33:164 – 9. interferon inhibits collagen synthesis in vivo in the
[68] Werner S, Peters KG, Longaker MT, et al. Large in- mouse. J Clin Invest 1987;79:1254 – 8.
12 J.L. Monaco, W.T. Lawrence / Clin Plastic Surg 30 (2003) 1–12

[86] Buck M, Houglum K, Chojkier M. Tumor necrosis tors of metalloproteinases: evolution, structure and
factor-alpha inhibits collagen alpha a (I) gene expres- function. Biochim Biophys Acta 2000;1477:267 – 83.
sion and wound healing in a murine model of cachexia. [88] Haroon ZA, Raleigh JA, Greenberg CS, et al. Early
Am J Pathol 1996;149:195 – 204. wound healing exhibits cytokine surge without evi-
[87] Brew K, Kinakarpandian J, Nagase H. Tissue inhibi- dence of hypoxia. Ann Surg 2000;231:137 – 47.

You might also like