Вы находитесь на странице: 1из 32

14

Wound healing
Chandan K. Sen and Sashwati Roy

SYNOPSIS

There are three general techniques of wound treatment: primary


intention, secondary intention, and tertiary intention
The following overlapping phases drive the overall healing
response: hemostasis, inflammation, proliferative, and
remodeling
Successful hemostasis or blood coagulation results in prevention
of blood loss by plugging the wound within seconds through
vasoconstriction and formation of a hemostatic blood clot
consisting of platelets and fibrin. The process is divided into
initiation and amplification. Initiation is caused by an extrinsic
pathway, whereas amplification is executed by an intrinsic
pathway
The inflammatory response during normal healing is characterized
by spatially and temporally changing patterns of specific leukocyte
subsets. Dysregulated inflammation complicates wound healing.
Complete resolution of an acute inflammatory response is the ideal
outcome following an insult
Infection is a common problem in chronic wounds, frequently
resulting in nonhealing wounds and significant patient morbidity
and mortality. Microorganisms do not always live as pure cultures
of dispersed single cells but instead accumulate at interfaces to
form polymicrobial aggregates such as films, mats, flocs, sludge,
or biofilms
In an open wound that has undergone contraction, restoration of
an intact epidermal barrier is enabled through wound
epithelialization, also known as re-epithelialization. During the
proliferative phase of wound healing, the granulation tissue is light
red or dark pink in color because of perfusion by new capillary
loops. It is soft to the touch, moist, and granular in appearance.
The granulation tissue serves as a bed for tissue repair

2013, Elsevier Inc. All rights reserved.

Wound vascularization may be achieved by angiogenesis or


vasculogenesis
A wound is generally considered chronic if it has not healed in 4
weeks. Chronic wounds can be broadly classified into three major
categories: venous and arterial ulcers, diabetic ulcers, and
pressure ulcers
Vascular complications commonly associated with problematic
wounds are primarily responsible for wound ischemia. Limitations
in the ability of the vasculature to deliver O2-rich blood to the
wound tissue leads to, among other consequences, hypoxia. Three
major factors may contribute to wound tissue hypoxia: (1)
peripheral vascular diseases garroting O2 supply; (2) increased O2
demand of the healing tissue; and (3) generation of reactive
oxygen species (ROS) by way of respiratory burst and for redox
signaling
Small RNAs are a new class of regulators of eukaryotic biology.
Alongside other small interfering RNAs (siRNAs), miRNAs execute
posttranscriptional gene silencing through mRNA destabilization as
well as translational repression. miRNAs are emerging as a key
regulatory of the overall wound-healing process
The regenerative potential of injured adult tissue suggests the
physiological existence of cells capable of participating in the
reparative process. Bone marrow-derived mesenchymal stem cells
(BM-MSCs) have been shown to promote the healing of diabetic
wounds, implying a profound therapeutic potential for skin defects
such as chronic wounds and burns
Scars (also called cicatrices) are macroscopic fibrous tissue that
visibly replaces normal skin after injury. There is a wide spectrum
of skin scarring postwounding, including scarless fetal wound
healing, fine-line or normal scars, stretched scars, atrophic
(depressed) scars, scar contractures, hypertrophic scars, and
keloids.

Acute wounds

Introduction
Physical trauma represents one of the most primitive
challenges that threatened survival. In other words,
injury eliminated the unfit. A Sumerian clay tablet (c.
2150 BC) described early wound care that included
washing the wound in beer and hot water, using poultices from substances such as wine dregs and lizard
dung, and bandaging the wound. Ancient scriptures
depicting the science of life or Ayurveda date from the
sixth to seventh century BC, and represent the beginning
of planned physical injury with the intent to cure.1,2
Hippocrates (c. 400 BC) detailed the importance of draining pus from the wound, and Galen (c. 130200 AD)
described the principle of first- and second-intention
healing.3 Wound healing advanced slowly over the centuries, with major advances in the 19th century in the
importance of controlling infection, hemostasis, and
necrotic tissue.4 Today, surgical trauma, taken together
with injury caused during accidents and secondary to
other clinical conditions, e.g., diabetes, represents a substantial cost to society.5 Any solution to wound-healing
problems will require a multifaceted comprehensive
approach. First and foremost, the wound environment
will have to be made receptive to therapies. Second, the
appropriate therapeutic regimen needs to be identified
and provided while managing systemic limitations that
could secondarily limit the healing response. This
chapter aims to present an overall outline of the cutaneous wound-healing process.

Acute wounds
Any violation of live tissue integrity may be regarded
as a wound. Skin is the largest organ of the human body,
covering about 3000 square inches (7620cm2) in an
average adult. The most important role of the skin for
terrestrial animals is to protect the water-rich internal
organs from the dry external environment. As a primary
line of defense against external threats, maintenance of
integrity of the skin is a key prerequisite for healthy
survival. Thus, healthy skin can regenerate and repair
itself under most common conditions. Skin wounds
may be open when manifested as a tear, cut, or puncture. Blunt force trauma may cause closed wounds or

Laceration

241

Puncture wound

Fig. 14.1 Laceration and puncture wounds.

contusion where the skin appears to be intact but suffers


from damage caused to underlying tissues.
Open wounds may be generally categorized as:
Lacerations ragged tears and cuts; masses of torn
tissue underneath; caused by dull knife, bomb
fragments and machinery and may include crushing
of tissues; frequently contaminated (Fig. 14.1)
Puncture e.g., sharp penetrations caused by nails,
needles, wire, or bullets (Fig. 14.1). These are of
great concern in patients with diabetes, as many of
these patients have polyneuropathy and have
insensate feet, leading to occult injury. Many times
these patients will step on thumb tacks, safety pins,
or other sharp household objects and not even
know it: this, coupled with compromised vascular
status, leads to chronic wound infection
Abrasions the superficial layer of the skin is
removed; skinned knee or elbows and rope burns
are examples; an abrasion lends itself to infection
Avulsions sections of skin torn off either in part
(attached to body) or totally (detached from body);
heavy bleeding is common
Amputations traumatic amputation results in
nonsurgical removal of limb from the body and
accompanies heavy bleeding.
There are
treatment:

three

general

techniques

of

wound

1. primary intention, in which all tissues, including


the skin, are closed with suture material after
completion of the operation

242

14 Wound healing

2. secondary intention, in which the wound is left


open and closes naturally

Scab

Clot

Neutrophils

24 hours

3. tertiary intention, in which the wound is left open


for a number of days and then closed if it is found
to be clean.

The wound-healing process


The entire wound-healing process may be viewed as a
cascade that is governed by numerous feedback and
feedforward regulatory loops driven by signals from the
wound tissue itself, wound microenvironment, as well
as interventions under conditions where the wound is
subjected to therapy. For simplicity of understanding
the several interdigitated biological processes that drive
the overall healing response, wound healing is commonly discussed as the following overlapping phases:
hemostasis and inflammation, proliferative (granulation, vascularization, and wound closure; closure may
be discussed as wound contraction and epithelialization) and remodeling (can continue from weeks to years
and encompasses scarring, tensile strength, and turnover of extracellular matrix (ECM) components). These
stages, taken as a whole, are also referred to as the
wound-healing cascade (Fig. 14.2).

Hemostasis
For bleeding wounds, the highest priority is to stop
bleeding and this is achieved by hemostasis. Hemostasis
is thus a protective physiological response to vascular
injury that results in exposure of blood components to
the subendothelial layers of the vessel wall. Through
successful hemostasis blood loss is prevented by plugging the wound within seconds through vasoconstriction and formation of a hemostatic blood clot consisting
of platelets and fibrin. Hemostasis requires both platelets and the blood coagulation system. The process of
blood coagulation may be subdivided into initiation
and amplification. Initiation is caused by an extrinsic
pathway, whereas amplification is executed by an intrinsic pathway. The intrinsic pathway consists of plasma
factor XI (FXI), IX, and VIII (Fig. 14.3). Tissue factor
(TF) generates a thrombin burst, a process by which

Re-epithelialization
Fibroblast

New
capillary

Granulation
tissue Macrophage

3-7 days

Weeks

Wound contraction

Fig. 14.2 (AC) Phases of cutaneous wound healing: a simplified representation.

Hemostasis

243

Tissue damage
Vessel injury
Healing

Collagen
FXII

Kallikrein

Prekallikrein
WBC

FXIIa

TF

FXIII Platelet

RBC

HK

Polymerization
and
stabilization

FXIIIa

Intrinsic
pathway

FVII

FXI

FVIIa

FXIa
Ca2+
Phospholipids

Phospholipids

Platelet Extrinsic
pathway

Ca2+

Protein-C
Protein-S

Fibrinogen

TM

TFPI
FIX

FVIII

FIXa

PF3

Antithombin
III

FV
FX

FVIIIa

FVa

Thrombin
Common
pathway

FX

FXa

Prothrombin

Fig. 14.3 The blood clotting cascade. FXII, factor XII,


HK, high-molecular-weight kininogen; TF, tissue factor;
WBC, white blood cell; RBC, red blood cell; TM,
thrombomodulin; PF3, platelet factor 3; TFPI, tissue factor
pathway inhibitor.

thrombin is released instantaneously. Thrombin is a key


driver of the overall coagulation cascade.
The extrinsic pathway responsible for the initiation of
blood coagulation consists of the transmembrane receptor TF and plasma FVII/VIIa. On the other hand, the
intrinsic pathway consists of plasma FXI, FIX, and FVIII.

Under physiological conditions, TF is constitutively


expressed by adventitial cells surrounding blood vessels
and initiates clotting. Examples of such adventitial
cells include vascular smooth-muscle cells, pericytes,
and adventitial fibroblasts.6,7 TF may also contribute to
amplification of blood coagulation through its so-called

244

14 Wound healing

blood-borne form, which is present as cell-derived


microparticles as well as through TF expressed within
platelets.68
Levels of FVIIa, a key player of the extrinsic pathway,
in the circulating blood are higher than any other activated coagulation factor. Following injury to the blood
vessel wall, FVII comes into contact with TF expressed
on TF-bearing cells (e.g., white blood cells) and forms
an activated complex (TFFVIIa). TFFVIIa activates
FIX and FX, resulting in FIXa and FXa, respectively. FVII
is activated by thrombin, FXIa, FXII, and FXa. The activation of FXa by TFFVIIa is almost immediately inhibited by the TF pathway inhibitor. FXa and its cofactor
FVa form the prothrombinase complex, which activates
prothrombin to thrombin. Thrombin is a serine protease
that plays a central role in hemostasis after tissue injury
by converting soluble plasma fibrinogen into an insoluble fibrin clot and by promoting platelet aggregation.
Thrombin activates other components of the coagulation cascade, including FV and FVIII, which in turn
activates FXI and cascades to the activation of FIX.
Thrombin also activates and releases FVIII from being
bound to von Willebrand factor. FVIIIa is the cofactor of
FIXa, and together they form the tenase complex,
which activates FX. In this way the cycle continues.
The intrinsic pathway begins with formation of the
primary complex on collagen by high-molecular-weight
kininogen, prekallikrein, and FXII (Hageman factor).
Prekallikrein is converted to kallikrein, and FXII becomes
FXIIa. FXIIa converts FXI into FXIa. FXIa activates
FIX, which together with its cofactor FVIIIa forms the
tenase complex. The tenase complex activates FX to FXa
(Fig. 14.3).
The blood clot physically helps plug the wound,
minimizing blood loss. It is primarily made up of
cross-linked fibrin, cells such as erythrocytes and platelets, as well as other ECM proteins such as fibronectin,
vitronectin, and thrombospondin. Current understanding portrays the clot as a dynamic structural matrix
containing functionally active proteins and cells. In
addition to containment of blood loss, the clot serves as
a first aid against microbial invasion. The clot also serves
as a provisional matrix for the homing of blood-borne
cells, including inflammatory as well as stem or progenitor cells. The provisional matrix is enriched in
cytokines and growth factors which then regulate the
function of the homing cells.9,10

The formation of blood clot is initiated by the proteolytic cleavage of fibrinogen by thrombin. As a result,
fibrin is produced and forms cross-links with each other.
Cross-linked fibrin entraps platelets, and together they
adhere to the subendothelium through adhesion molecules called integrins. Clot fibrin plays a key role in
mounting the inflammatory process as well as in facilitating wound angiogenesis and stromal cell proliferation. Fibrin binds to integrin CD11b/CD18 on infiltrating
monocytes and neutrophils. It also binds to fibroblast
growth factor-2 (FGF-2) and vascular endothelial growth
factor (VEGF) that help the wound tissue vascularize.
Fibrin also binds to insulin-like growth factor-I and promotes stromal cell proliferation.11,12
Blood clot represents the seat of wound chemotaxis.
Thrombin, released by platelets at the wound site, is an
early mediator of clot development.13 Thrombin is a
serine protease that converts soluble plasma fibrinogen
into an insoluble fibrin clot. In addition, it promotes
platelet aggregation. Thrombin function may be viewed
as an interface between the hemostasis phase of wound
healing and the ensuing inflammatory phase as it plays
a potent role in mounting wound inflammation. The
proinflammatory effects of thrombin include stimulation of vasodilation responsible for plasma extravasation, edema, and an increased expression of endothelial
cell adhesion molecules that helps monocytes and others
cells extravasate and infiltrate the wound site. Thrombin
also induces the release of proinflammatory cytokines
like CCL2, interleukin-6 (IL-6), and IL-8 by endothelial
cells. These cytokines induce monocyte chemotaxis.14
Furthermore, thrombin induces the release of inflammatory cytokines by monocytes, including IL-6, interferon-,
IL-1, and tumor necrosis factor- (TNF-). These earlyphase cytokines are typically proinflammatory, which
may govern the differentiation of blood-derived monocytes into M1 wound macrophages.15 Wound chemotaxis is also driven by the degradation of fibrin and
subsequent activation of the complement system. As
part of this process several chemotactic agents and
cytokines are released, which in turn launch the inflammatory phase of wound healing through chemotactic
recruitment of blood-borne immune cells.16 Platelets are
one of the earliest sources of cytokines which execute
immune cell chemotaxis as well as macrophage activation. Once trapped in the fibrin net, platelets release
granules that function as a reservoir for biologically

Inflammation

active proteins, such as RANTES (regulated on activation, normal T cell expressed, and secreted or CCL5),
thrombin, transforming growth factor- (TGF-),
platelet-derived growth factor (PDGF), and VEGF.
CCL5 is one of the most potent monocyte chemoattractants released by platelets after injury. Other cytokines
and chemokines that attract monocytes to the wound
bed include monocyte chemoattractant protein-1
(MCP-1) (CCL2), MIP-1 (CCL3), TGF-, fibronectin,
elastin, C5a, C3a, nerve growth factor, and ECM
components.17,18

Inflammation
Tissue injury triggers an acute-phase inflammation
(Latin, inflammare, to set on fire) response that is meant
to prepare the wound site for subsequent wound closure
(Fig. 14.4). Inflammation encompasses a series of
responses of vascularized tissues of the body to injury.
Local chemical mediators that are biosynthesized during
acute inflammation give rise to the macroscopic events
characterized by Celsus in the first century, namely,
rubor (redness), tumor (swelling), calor (heat), and dolor
(pain). At cellular and molecular levels, inflammation
results from the coordination of manifold systems of
receptors and sensors that affect transcriptional and
posttranslational programs necessary for host defense
and resolution of infection. During normal healing, the
inflammatory response is characterized by spatially and
temporally changing patterns of specific leukocyte
subsets.

Platelets
Formation of the clot or thrombus is dependent on
platelet activation. The platelet-rich blood clot also
entraps polymorphonuclear leukocytes (neutrophils).
This helps amplify blood coagulation and lays the foundation for the subsequent acute-phase inflammatory
response. In a matter of hours after injury, large numbers
of neutrophils extravasate by transmigrating across the
endothelial cell wall of blood capillaries to the wound
site. To enable this, local blood vessels are activated
by proinflammatory cytokines such as IL-1, TNF-,
and interferon-. These cytokines induce the expression
of adhesion molecules necessary for adhesion of

Initiate
tissue
repair

Cytokines
signaling
path to
injury site

245

Neutrophil

6
3

Platelets

Mast cell

Macrophage
1
1 Bacteria and other pathogens enter wound
2 Platelets from blood release blood-clotting proteins at wound site.
3 Mast cells secrete factors that mediate vasodilation and vascular constriction.
Delivery of blood, plasma and cells to injured area
4 Neutrophils and macrophages remove pathogens by phagocytosis
5 Macrophages secrete hormones called cytokines that attract immune system cells
to the site and activate cells involved in tissue repair
6 Inflammatory response continues until the foreign material is eliminated and the
wound is repaired

Fig. 14.4 The wound inflammatory response.

leukocytes and diapedesis (Fig. 14.5). Adhesion molecules such as integrins as well as P-selectin and E-selectin
play a central role in enabling diapedesis of neutrophils
(Fig. 14.6). These adhesion molecules bind with integrins
expressed on the cell surface of neutrophils, such as
CD11a/CD18 (LFA), CD 11b/CD18 (MAC-1), CD11c/
CD18 (gp150, 95), and CD11d/CD18. Alongside
cytokines, chemokines play a major role in mounting
acute-phase inflammation after injury. Chemokines
include IL-8, MCP-1, and growth-related oncogene-. In
the case of an infected wound, bacterial products such
as lipopolysaccharide and formyl-methionyl peptides
can enhance neutrophil recruitment to the wound site
(Fig. 14.7).

14 Wound healing

246

Normal permeability of capillary

Small amount of fluid

Rolling

Shedding of
L-selectin

Adhesion

Diapedesis

Neutrophil

Capillary wall

Monocyte

Increased permeability of capillary


during inflammation

More fluid and


antimicrobal chemicals

Activating substances
released by
bacteria
and
damaged
tissues

C3a C5a,
chemokines,
histamine,
prostaglandins
and leukotrienes

Lipopolysaccharides,
interleukin-1, and
tumor necrosis factor

L-selectin
Sialyl-Lewis

Interstitial
spaces

Monocyte squeezing
through interstitial space

Integrin
E-selectin

Fig. 14.6 Extravasation of neutrophils in response to tissue injury. Neutrophils


move along the capillaries in a rolling motion which is facilitated by the binding
and release of L-selectin on the neutrophil surface to sialyl-Lewis, a carbohydrate
ligand expressed on the inner wall of capillaries by endothelial cells. Upon injury
and/or infection the release of lipopolysaccharides, tumor necrosis factor-alpha,
and interleukin-1 results in the shedding of L-selectin by the neutrophils which
then strongly adhere to the inner wall of the capillary by the binding of integrin
on the neutrophils to E-selectins on endothelial cells. After such adhesion, the
process of diapedesis begins, allowing the neutrophils to extravasate to the
wound site.

Fig. 14.5 Diapedesis. In healthy permeable capillaries the endothelium lining


prevents blood cells from leaving the circulation. In response to injury, blood
vessels around the wound site undergo vasodilation, increasing the permeability of
capillaries. Such change enables inflammatory cells to extravasate through the
capillary wall and migrate to the site of injury. This process includes release of fluid
from the vessels to the extracellular space, resulting in the edematous response
commonly noted during inflammation. Blood vessels possess a built-in pathway for
such diapedesis to occur in response to tissue injury.

Neutrophils
Neutrophils traverse postcapillary venules at sites of
inflammation, degrade pathogens within phagolysosomes, and undergo apoptosis. Neutrophils serve a
wide range of functions, ranging from phagocytosis of
infectious agents to cleansing of devitalized tissue.
When coated with opsonins (generally complement
and/or antibody), microorganisms bind to specific
receptors on the surface of the phagocyte and invagination of the cell membrane occurs with the incorporation
of the microorganism into an intracellular phagosome.
There follows a burst of oxygen consumption, and
much, if not all, of the extra oxygen consumed is converted to highly reactive oxygen species. This is called

Fibrinplatelet clot

Collagen

Elastin

Red blood
cell

Fibroblast

Cytokines &
growth factors

Fig. 14.7 Diapedesis and migration of leukocytes to the wound site.

Inflammation

respiratory burst. In addition, the cytoplasmic granules


discharge their contents into the phagosome, and death
of the ingested microorganism soon follows. Among the
antimicrobial systems formed in the phagosome is one
consisting of myeloperoxidase (MPO), released into the
phagosome during the degranulation process, hydrogen peroxide (H2O2), formed by the respiratory burst
and a halide, particularly chloride. The initial product
of the MPO-H2O2-chloride system is hypochlorous acid,
and subsequent formation of chlorine, chloramines,
hydroxyl radicals, singlet oxygen, and ozone has been
proposed. These same toxic agents can be released to
the outside of the cell, where they may attack normal
tissue and thus contribute to the pathogenesis of
disease.19 Other products delivered by neutrophils to
the wound site include antimicrobials such as cationic
peptides and eicosanoids as well as proteases such as
elastase, cathepsin G, proteinase 3, and urokinase-type
plasminogen activator. As it relates to the overall inflammatory process elicited in response to injury, neutrophils
are major players because they can modify macrophage
function and therefore regulate innate immune response
during wound healing.20 In the absence of neutrophils,
wound site macrophages seem to lack guidance in conducting the healing process.21
In a healing wound, neutrophil infiltration ceases
after a few days of injury. Expended neutrophils are
programmed to die and dying neutrophils are recognized by wound site macrophages and phagocytosed.
The wound site contains a small portion of macrophages
that are resident. Most macrophages at the wound
site are recruited from the peripheral circulation.
Extravasation of peripheral blood monocytes is enabled
by the interaction between endothelial vascular cell
adhesion molecule-1 and monocyte very late antigen-4
(41 integrin). Factors that guide the extravasated
monocyte to the wound site include growth factors,
chemotactic proteins, proinflammatory cytokines, and
chemokines such as macrophage inflammatory protein
1, MCP-1, and RANTES. The source of these chemoattractants includes clot-associated platelets, wound edge
hyperproliferative keratinocytes, wound tissue fibroblasts and subsets of leukocyte already at the wound
site. Once the monocyte leaves the blood vessel to transmigrate into the wound site through the ECM micro
environment, the process of monocyte differentiation
to macrophages has started.

247

Gram-negative
bacteria

Lipopolysaccharide

Toll-like
receptor

Cell membrane

Nucleus
Cytoplasm

Fig. 14.8 Toll-like receptors: responding to infectious agents and the wound
microenvironment.

Mediators present in the microenvironment that the


monocyte traverses to reach the wound site interact
with receptors on the monocyte cell surface, bringing
forth major changes in the transcriptomic as well as
proteomic make of the cell. Major examples of such
receptors present on the monocyte surface include Tolllike receptors (TLRs: Fig. 14.8), complement receptors,
and Fc receptors. At the wound site, macrophages
function as antigen-presenting cells and phagocytes
scavenging dead cells and debris. In addition, they
deliver a wide range of growth factors that are known
for their abilities to execute the wound-healing process.
Such growth factors include TGF-, TGF-, basic FGF
(bFGF), VEGF, and PDGF. These growth factors enable
wound healing by causing cell proliferation and synthesis of ECM and inducing angiogenesis. Macrophages
play a crucial role in enabling wound healing.
Macrophage depletion is known to impair wound
closure markedly.4,22

Mast cells
Mast cells are best known for their central role in mediating allergic responses. Beyond that function, it is now

248

14 Wound healing

known that mast cells are physiologically significant


in recognizing pathogens and in regulating immune
response.23 Mast cells may instantly release several
proinflammatory mediators from intracellular stores. In
addition, they are localized in the hostenvironment
interface. These properties make mast cells key players
in finetuning immune responses during infection.
Recent studies using mast cell activators as effective
vaccine adjuvants show the potential of harnessing
these cells to confer protective immunity against microbial pathogens.24 Mast cell activation helps initiate the
inflammatory phase of wound healing. In response to
injury, mast cells at the wound site degranulate within
a matter of hours and therefore become histologically
silent at the wound tissue. After about 48 hours of injury,
mast cells are again seen in the wound tissue and their
number increases as healing progresses.25 On one hand,
impaired wound healing has been reported in mast celldeficient mice.26 On the other hand, mast cells have been
implicated in skin wound fibrosis.27 With the aid of a
wide array of newly formed or preformed mediators
released by degranulation, the activated mast cell controls the key events of the healing phases: triggering and
modulation of the inflammatory stage, proliferation of
connective cellular elements, and final remodeling of
the newly formed connective tissue matrix. The importance of the mast cell in regulating healing processes is
also demonstrated by the fact that a surplus or deficit
of degranulated biological mediators causes impaired
repair, with the formation of exuberant granulation
tissue (e.g., keloids and hypertrophic scars), delayed
closure (dehiscence), and chronicity of the inflam
matory stage.28

Macrophages
Macrophages represent the predominant cell type in a
healing wound 35 days following injury. The primary
acute function of wound macrophages, which arrive at
an injury site hours later than neutrophils, is to operate
as voracious phagocytes cleansing the wound of all
matrix and cell debris, including fibrin and apoptotic
neutrophils. Macrophages also produce a range of
cytokines, growth and angiogenic factors that drive
fibroblast proliferation and angiogenesis.4,2932 In a
classic study, Leibovich and Ross33 demonstrated that
antimacrophage serum combined with hydrocortisone

diminished the accumulation of macrophages in healing


skin wounds of adult guinea pigs. Such depletion
resulted in impaired disposal of damaged tissue and
provisional matrix, compromised fibroblast count, and
delayed healing. Today, macrophages have emerged to
be a pivotal driver of efficient skin repair.34,35 Macrophages
are plastic and heterogeneous cells broadly categorized
into two groups: classically activated or type I macrophages, which are proinflammatory effectors, and alternatively activated or type II macrophages.36 In the
inflamed tissue, it is unclear whether the type II macrophages that appear during the healing phase originate
from newly attracted monocytes or from a switch in the
activation state of previously proinflammatory macrophages. The macrophage population first taking part in
inflammation may change its phenotype and assume
the role to resolve inflammation.37,38 Macrophages from
diabetic wounds display dysfunctional inflammatory
responses.39 A persistent inflammatory state of diabetic
wound macrophages is caused by impairment in the
ability of these cells to phagocytose apoptotic cells at the
wound site which in turn prevents the switch from M1
to M2 phenotype.39

Resolution of inflammation
Inflammatory responses elicited by injury are only
helpful to the healing process if they are timely and
transient. Dysregulated inflammation complicates
wound healing.39 Complete resolution of an acute
inflammatory response is the ideal outcome following
an insult. For resolution to ensue, further leukocyte
recruitment must be halted and accompanied by
removal of leukocytes from inflammatory sites. Reso
lution of inflammation is executed by a number of key
factors. At the wound site successful phagocytosis of
dead neutrophils by macrophages is a key factor.
Impairment in macrophage function at the wound site
derails the resolution of inflammation.39 Lipid mediators, such as the lipoxins, resolvins, protectins, and
newly identified maresins, have emerged as a novel
genus of potent and stereoselective players that
counterregulate excessive acute inflammation and
stimulate molecular and cellular events that define
resolution.40 Successful resolution paves the path for
the healing process to progress towards successful
wound closure. Prolonged inflammation may not only

Infection

compromise wound closure but may also worsen scar


outcomes.41,42

Infection
Infection is a common problem in chronic wounds, frequently resulting in nonhealing and significant patient
morbidity and mortality.43 Wound infection and the subsequent release of proinflammatory modulators result
in pain and delayed healing. The pain, in turn, compromises the immune response to infection.44 All wounds
become contaminated by bacteria from the surrounding
skin, the local environment, and autologous patient
sources. The local environment is particularly relevant
for hospitalized patients. Colonization is defined as the
presence of proliferating bacteria without a noticeable
host response. Colonization of the wound may enhance
or impede wound healing, depending upon the bacterial load. Bacterial loads in excess of 105 organisms/
gram of tissue are a threat to wound healing, although
this threshold may be altered by the status of the host
immune system and the number and types of bacterial
species present. The concept of critical colonization
is controversial and not universally accepted. Critical
colonization is characterized by increased bacterial
burden or covert infection, and the wound at this stage
may enter a nonhealing, chronic inflammatory state.
Substantial colonization may not cause the obvious
signs of inflammation but will likely affect wound
healing with failure to heal or slowing of progression.
Signs of critical colonization are atrophy or deterioration of granulation tissue, discoloration of granulation
tissue to deep red or gray, increased wound friability,
and increased drainage. The transition to infection
occurs when bacterial proliferation overcomes the hosts
immune response and host injury occurs. Several factors
determine transition from colonization to infection: the
bioburden itself, the virulence of the organisms, the synergistic action of different bacterial species, and the
ability of the host to mount an immune response.43
During the past decade, there has been rapid progress
in the understanding of innate immune recognition
of microbial components and its critical role in host
defense against infection. The early concept of innate
immunity was that it nonspecifically recognized
microbes; however, the discovery of TLRs (Fig. 14.8) in

249

the mid-1990s showed that pathogen recognition by the


innate immune system is instead actually specific,
relying on germline-encoded pattern recognition receptors (PRRs) that have evolved to detect components of
foreign pathogens, referred to as pathogen-associated
molecular patterns (PAMPs).45 TLRs regulate innate and
adaptive immune responses and are important modulators of inflammation during wound-healing responses.
The finding that there is activation of TLR signaling
during tissue damage in several disease situations in the
absence of infection suggests that endogenous molecules serve as TLR agonists, although it is unclear
whether this response is biologically important for
maintenance of homeostasis, such as tissue repair, or
whether this recognition is simply accidental. It is noteworthy that microbial infection triggers the production
of modified endogenous molecules (such as highmobility group protein B1, oxidized phospholipids,
-defensin 2, and nucleic acids) that are recognized by
TLRs or other cytosolic PRRs. This may suggest that
these endogenous molecules, along with PAMPs, act as
adjuvants to activate innate immune programs via TLRs
and/or other PRRs, and have key roles in facilitating
adaptive immunity against infecting microbes.
Chronic wounds have a complex colonizing flora that
changes over time. Staphylococcus aureus and coagulasenegative staphylococci are the most commonly isolated
organisms. Chronic wounds are colonized by multiple
bacterial species and many persist in the wound once
they are established. In chronic venous leg ulcers the
most common bacteria noted, in order of abundance,
were S. aureus, Enterococcus faecalis, Pseudomonas aeruginosa, coagulase-negative staphylococci, Proteus spp.,
and anaerobic bacteria. Resident (colonizing) bacterial
species are commonly present in ulcers. The longer an
ulcer remains unhealed, the more likely it will acquire
multiple aerobic organisms and a significant anaerobic
population. Chronic wounds are commonly complicated by underlying ischemia. Thus, they tend to have
a low tissue oxygen level. This facilitates the growth of
anaerobes in ischemic wounds. Adequate delivery of
oxygen to the wound tissue is vital for optimal healing
and resistance to infection.46 Hospitalization, surgical
procedures, and prolonged or broad-spectrum anti
biotic therapy may predispose patients to colonization or infection, or both, with resistant organisms,
including S. aureus (methicillin-resistant S. MRSA) or

250

14 Wound healing

vancomycin-resistant enterococci.43 Because inflammatory responses to microbial invasion may be diminished


in persons with diabetes, clinical signs of infection are
often absent in persons with diabetic foot ulcers when
infection is limited to localized tissue.47

drug and other physiologic changes that could impair


drug effectiveness.43

Biofilm

Wounds larger than can be closed by diffusion of oxygen


from neighboring intact blood vessels or wounds complicated by underlying ischemia largely rely on wound
vascularization for their closure. Wound vascularization
may be achieved by angiogenesis or vasculogenesis.
Angiogenesis represents sprouting of capillaries from
existing blood vessels in the wound edge tissue.
Vasculogenesis relies on the formation of new blood
vessels by mobilization of bone marrow-derived
endothelial stem cells.
Wound vascularization is regulated by all phases in
wound healing hemostasis, inflammation, tissue formation, as well as tissue remodeling. The hemostatic
plug provides a bed for blood-borne cells to home. Once
cells entangle in this plug, the ECM environment modifies cell function towards successful healing. Platelets in
the clot serve as a source of growth factors and cytokines
which recruit several cell types, including endothelial
cells, to the wound site. During the inflammatory phase,
leukocytes at the wound site serve as a major source of
proangiogenic factors such as VEGF-A and IL-8 that lay
the early foundation for successful wound tissue vascularization. As neutrophils are expended and undergo
cell death, the number of macrophages at the wound
site substantially increases. Macrophage-derived TGF, TGF-, bFGF, PDGF, and VEGF play a key role in
driving skin wound angiogenesis. Growing evidence
demonstrates that no single angiogenic factor is singularly effective in significantly influencing wound outcomes. Wound vascularization is a sophisticated process
requiring dynamic, temporally and spatially regulated
interaction between cells, angiogenic factors, and the
ECM.
Key processes in tissue vascularization are depicted
in Figure 14.9. Angiogenic cues are elicited by micro
environmental signals such as hypoxia and are amplified by angiogenic factors such as VEGF expressed by
and released from cells at the wound site. VEGF was
originally identified as an endothelial cell-specific
growth factor-stimulating angiogenesis and vascular
permeability. Some family members, VEGF C and D, are

Microorganisms do not always live as pure cultures of


dispersed single cells but instead accumulate at interfaces to form polymicrobial aggregates such as films,
mats, flocs, sludge, or biofilms. The biofilm state of
microorganisms may lead to an increase in virulence
and propensity to cause infection. In most biofilms, the
microorganisms account for less than 10% of the dry
mass, whereas the matrix can account for over 90%. The
matrix is the extracellular material, mostly produced by
the organisms themselves, in which the biofilm cells are
embedded. It consists of a conglomeration of different
types of biopolymers known as extracellular polymeric substances (EPS) that forms the scaffold for the
three-dimensional architecture of the biofilm and is
responsible for adhesion to surfaces and for cohesion in
the biofilm. The formation of a biofilm allows a lifestyle
that is entirely different from the planktonic state.
Although the precise and molecular interactions of the
various secreted biofilm matrix polymers have not been
defined, and the contributions of these components to
matrix integrity are poorly understood at a molecular
level, several functions of EPS have been determined,
demonstrating a wide range of advantages for the
biofilm mode of life.
The architecture of biofilms is influenced by many
factors, including hydrodynamic conditions, concentration of nutrients, bacterial motility, and intercellular
communication, as well as exopolysaccharides and proteins.48 Chronic wounds offer attractive conditions for
biofilm production because proteins (collagen, fibronectin) and damaged tissues are present, which allow
attachment. The biofilm impedes healing of chronic
wounds. Most of the chronic wound pathogens, such as
MRSA and Pseudomonas spp., are typical biofilm producers. Compared to bacteria in the unattached freeliving planktonic form, bacteria that reside within
mature biofilms are highly resistant to traditional antibiotic therapies. Bacteria in biofilms grow more slowly,
and slower growth may lead to decreased uptake of the

Vascularization

Vascularization

Angiogenic
factor (VEGF)
production
Release
Angiogenic
factors bind
to endothelial
cell receptors
3

Endothelial cell
activation

Loop
formation

Vascular
stabilization

7
ECM
remodeling
6

5
Endothelial cell
proliferation

Directional
migration

10
Pericyte

Tie-2

y3

251

8
Tube
formation

Fig. 14.9 Neoangiogenesis: the formation of new


blood vessels. VEGF, vascular endothelial growth
factor; ECM, extracellular matrix.

specifically involved in lymphangiogenesis. Ligation of


these angiogenic factors with their corresponding receptors elicits a multitude of cell-signaling processes that
activate microvascular endothelial cells. For example,
VEGF and their endothelial tyrosine kinase receptors
are central regulators of tissue vascularization. VEGF
signaling through VEGFR-2 is the major angiogenic
pathway. VEGFR-3 has also been shown to be important
for angiogenesis, acting together with VEGF/VEGFR-2
and Dll4/Notch signaling to control angiogenic sprouting.49 The biological significance of other angiogenic
factors such as EGF and bFGF is mediated by their corresponding receptors, which also belong to the family
of tyrosine kinase receptors EGFR, FGFR-1, FGFR-2,
FGFR-3, and FGFR-4.
Other tyrosine kinase receptors of outstanding significance in this regard are Tie-1 and Tie-2. Along with
the VEGF receptor, these are the only known endothelial
cell-specific receptor tyrosine kinases. Tie-2 is induced
on the endothelium of neovessels in skin wounds and
downregulated as newly formed vessels regress. As an
indicator of Tie-2 activation, Tie-2 phosphorylation is
detected in skin wounds at all stages of the healing
process.50 Activated microvascular endothelial cells
respond by proliferating, which is followed by directional migration of these cells. Migration is a complex

process in which cells move in a given direction either


in response to changes in the extracellular environment
or as a consequence of an intrinsic propensity for directional movement. ECM remodeling by proteases promotes cell migration, a critical event in the formation of
new vessels. Temporal and spatial regulation of ECM
remodeling events allows for local changes in net matrix
deposition or degradation, which in turn contributes to
control of cell growth, migration, and differentiation
during different stages of angiogenesis. Matrix-bound
growth factors released by proteases and/or by angiogenic factors promote angiogenesis by enhancing
endothelial migration and growth. Matrix molecules
promote endothelial cell growth and morphogenesis,
and/or stabilize nascent blood vessels. Hence, ECM
molecules and ECM remodeling events play a key role
in regulating angiogenesis.51
The formation of the capillary-like tubes is specific
to endothelial cells and integral to the process of angiogenesis. The basement membrane represents a biologically functional highly specialized ECM on which the
basal nonluminal surface of endothelial cells rests. This
matrix forms a continuous sleeve around the endothelial cells, and maintains the tube-like structures of the
blood vessels. More than 20 years ago Kubota etal.
observed that endothelial cells plated on a reconstituted

252

14 Wound healing

basement membrane matrix, rapidly attached, aligned,


and formed capillary-like tubules. The cells did not proliferate. The vessels that are thus formed contain a
lumen and tight cellcell contacts. The cells are polarized with the nuclei basally located towards the basement membrane matrix. Furthermore, the capillary-like
structures take up acetylated low-density lipoprotein,
which is a marker of differentiation for these cells.52
Angiogenesis not only depends on endothelial cell invasion and proliferation, but also requires pericyte coverage of vascular sprouts for vessel stabilization. These
processes are coordinated by VEGF and PDGF through
their cognate receptors on endothelial cells and vascular
smooth-muscle cells, respectively.53 Structural support
to blood vessels is provided to pericytes and vascular
smooth-muscle cells. Normal pericytes are embedded
within the basement membrane of capillaries, either as
solitary cells or a single-cell layer, where they coordinate
intercellular signaling with endothelial cells and other
components of the blood vessel wall to prevent leakage.
In contrast, vascular smooth-muscle cells form single or
multiple layers around arteries and veins to mediate
vascular tone and contraction. Pericyte coverage is
required for the stabilization of immature endothelial
tubes.54 Pericytes have function beyond angiogenesis
that are relevant to wound healing. Skin pericytes may
act as mesenchymal stem cells (MSCs), exhibiting the
capacity to differentiate into bone, fat, and cartilage lineages. Thus, pericytes represent a potent stem cell population in the skin that is capable of modifying the ECM
microenvironment and promoting epidermal tissue
renewal from nonstem cells.55

Wound closure
Wound contraction and re-epithelialization contribute
to closure of wounds that heal by secondary intention.
Wound contraction represents an early response to
injury that is aimed at juxtaposing the edges of an open
wound.56 This early phase of wound closure appears to
be mediated by a contractile purse-string force produced by a circumferentially arranged band of fusiformshaped epidermal cells situated in the wound margin.57
Fibroblasts at the wound edge tissue are recognized to
play a key role in enabling wound contraction.58 During
the inflammatory phase of wound healing, fibroblasts

acquire smooth-muscle cell characteristics and differentiate into contractile myofibroblasts. The first phenotypic transition of wound edge fibroblasts into so-called
protomyofibroblasts is characterized by neoformation
of contractile -cytoplasmic actin stress fibers and occurs
in response to profibrotic cytokines and to altered properties of the ECM. In the presence of TGF-1 in a
mechanically restrained environment, these cells express
-smooth-muscle actin de novo, which significantly
increases their contractile activity and is a hallmark of
the differentiated myofibroblast.59 Wound contraction
may significantly contribute to wound closure, although
this contribution is much larger in loose-skinned rodents
than in humans.
In an open wound that has undergone contraction,
restoration of an intact epidermal barrier is enabled
through wound epithelialization, also known as
re-epithelialization.60,61 A wound that is not epithelialized is not considered healed, no matter how
perfectly restored the underlying dermal structures
may be. Thus, wound epithelialization, also called
re-epithelialization, is a critical and defining feature of
wound repair. Re-epithelialization of the wound can be
conceptually viewed as the result of three overlapping
keratinocyte functions: migration, proliferation, and differentiation. The sequence of events by which keratinocytes accomplish the task of re-epithelialization is
generally believed to begin with dissolution of cellcell
and cellsubstratum contacts. This is followed by the
polarization and initiation of directional migration in
basal and a subset of suprabasilar keratinocytes over the
provisional wound matrix. A subset of keratinocytes
immediately adjacent to, but not within, the wound bed
then undergoes mitosis. Finally, there is multilayering
of the newly formed epidermis and induction of differentiation specific gene products to restore the functionality of the epidermis. The most limiting factor in
wound re-epithelialization is migration, since defects in
this function, but not in proliferation or differentiation,
are associated with the clinical phenotype of chronic
nonhealing wounds.62 The process of epithelialization
continues until the barrier is re-established and the
wound is covered. The process of re-epithelialization is
accelerated by a moist environment63,64 and is facilitated
by the enzyme matrix metalloproteinase 1, a collagenase
which lessens the affinity of the collagenintegrin
contacts.65

Chronic wounds

Proliferative phase
The proliferative phase starts around 2 days after injury
and normally lasts up to 3 weeks in a healing cutaneous
wound. This phase overlaps with the inflammatory
phase and supports re-epithelialization, the formation
of new blood vessels, and the influx of fibroblasts and
laying down of the ECM. By the time this phase begins,
the degradation of the fibrin clot by the macrophages
has begun and invading endothelial cells and fibroblasts
rapidly fill that space. Migrating fibroblasts produce
the cytokines that induce keratinocytes to migrate and
proliferate.66 Activated macrophages produce several
cytokines, such as PDGF and TNF-, which also induce
fibroblasts to produce keratinocyte growth factor which
in turn induces wound re-epithelialization.60,61

Granulation tissue
The fibrin clot formed during hemostasis participates in
the early inflammatory phase and is replaced by a perfused, fibrous connective tissue that grows from the
base of a wound and is able to fill wounds of almost any
size. During the proliferative phase of wound healing,
this granulation tissue is light red or dark pink in color
because of perfusion by new capillary loops. It is soft to
the touch, moist, and granular in appearance. The granulation tissue serves as a bed for tissue repair. The ECM
of granulation tissue is created and modified by fibroblasts. Initially, it consists of a network of type III collagen, a weaker form of the structural protein that can be
produced rapidly. This is later replaced by the stronger,
long-stranded type I collagen, as evidenced in scar
tissue. Formation and contraction of the granulation
tissue represent integral aspects of the healing wound.
In ischemic wounds, contraction of the granulation
tissue is impaired because of faulty myofibroblast
function, cells responsible for granulation tissue
contraction.67

Chronic wounds
A wound is generally considered chronic if it has not
healed in 4 weeks. Chronic wounds have been also
defined as wounds that have not shown a 2040% reduction in area after 24 weeks of optimal therapy. Standard

253

surgical textbooks define chronic wounds as wounds


which have not healed in 3 months. Chronic wounds
can be broadly classified into three major categories:
venous and arterial ulcers, diabetic ulcers, and pressure
ulcers.

Venous ulcers
Venous ulcers (stasis ulcer or varicose ulcers) are wounds
that are thought to occur due to improper functioning
of venous valves, usually of the legs. They are the major
cause of chronic wounds, occurring in 5070% of chronic
wound cases.68 Venous ulcers develop mostly along the
medial distal leg, and can be very painful. According to
the revised clinical, etiology, anatomy, and pathophysiology (CEAP) classification of chronic venous disease
published in 2004, a venous ulcer is defined as a fullthickness defect of skin, most frequently in the ankle
region, that fails to heal spontaneously and is sustained
by chronic venous disease.69 Systematically, venous
ulcer may be defined as a defect in the skin with surrounding pigmentation and dermatitis, located in the
lower leg (usually in the gaiter region) that has been
present for greater than 30 days, characterized by
persistent venous hypertension and abnormal venous
function (result of venous reflux and/or obstruction
confirmed by hemodynamic and/or physiologic assessment), without a primary or associated arterial, immunologic, endocrine, or systemic cause. It is recognized
that ulcers can be caused purely by venous pathology
such as venous reflux or obstruction. When these abnormalities are combined with additional pathologic conditions, they contribute to the causation and perpetuation
of the ulcer. The latter situation includes comorbid conditions such as arterial ischemia, swelling and lymphedema, trauma, autoimmune disorders, neurotrophic
conditions, and diabetic vascular disease. These ulcers
are categorized as of mixed origin, in which the venous
component may or may not play a dominant role.
Successful treatment of such ulcers includes not only the
venous component but also concomitant management
of the comorbid condition.

Arterial ulcers
Because both arterial and venous ulcers typically occur
on the lower leg, differentiating between them can be

254

14 Wound healing

challenging for wound care practitioners. However,


they have very different pathophysiologies and management pathways. The most common cause of arterial
ulcers is atherosclerosis. Risk factors for the development of atherosclerosis include age, smoking, diabetes
mellitus, hypertension, dyslipidemia, family history,
obesity, and sedentary lifestyle.70 Ischemia and necrosis
are common consequences. Both acute and chronic arterial insufficiency can lead to the formation of lowerextremity ulcers. Arterial insufficiency can occur at any
level, from large arteries to arterioles and capillaries.
Tissue ischemia that leads to leg ulcers tends to occur
more in the setting of large-vessel or mixed disease.
Vascular claudication, with exercise, at night, or while
one is resting, is often the most distinguishing characteristic of arterial ulcers. Determining the ankle brachial
index give an indication of a patients ability to heal.
However, diabetic patients may have falsely elevated
ankle brachial index results secondary to vessel calcification. Patients with arterial ulcers must have increased/
adequate blood supply to heal and benefit most from
revascularization procedures. It should be noted that
arterial insufficiency might act in concert with other
pathological mechanisms, leading to tissue necrosis and
ulceration. Diabetic foot ulcers, for example, may result
from the combination of neuropathy, trauma, and arterial insufficiency.

Healthy foot

Diabetic foot

Blood vessel damage in the feet may cause tissue damage


such as sores, lesions and poor circulation that can lead to amputation

Fig. 14.10 The ischemic diabetic foot.

Diabetic ulcers
Diabetic ulcers are the most common foot injuries
leading to lower extremity amputation (Figs 14.10 and
14.11). In diabetics, the effects of peripheral neuropathy,
peripheral vascular disease, and infection often combine
to facilitate the development of diabetic ulcers that
can lead to gangrene and amputation. Diabetic persons,
like people who are not diabetic, may develop atherosclerotic disease of large- and medium-sized arteries,
such as aortoiliac and femoropopliteal atherosclerosis.
However, significant atherosclerotic disease of the infrapopliteal segments is particularly common in the
diabetic population. Underlying digital artery disease,
when compounded by an infected ulcer in close proximity, may result in complete loss of digital collaterals and
precipitate gangrene. The reason for the prevalence of
this form of arterial disease in diabetic persons is thought
to result from a number of metabolic abnormalities,

Diabetic foot ulcer

Fig. 14.11 Diabetic foot ulcer.

Chronic wounds

255

motor nerves can lead to clawing of the toes because of


a resulting imbalance of intrinsic and extrinsic muscles.
The long-term morbidity of nerve injuries of the foot is
predominantly related to sensory nerve injury, except
when a tibial nerve injury causes intrinsic muscle function loss. The two main problems associated with injuries to these nerves are the lack of sensation in the distal
distribution of the nerve (Fig. 14.12) and the formation
of a painful neuroma. When the nerve injury occurs in
a weight-bearing area of the foot, the presence of a
painful neuroma often leads to complex regional pain
syndrome type I.74

Saphenous nerve
Deep peroneal nerve
Superficial peroneal
nerve
Medial plantar nerve
Lateral plantar nerve
Calcaneal branch
(tibial nerve)
Sural nerve

Pressure ulcers
Dorsal surface

Plantar surface

Fig. 14.12 Cutaneous innervations of the foot.

including high low-density lipoprotein and verylow-density lipoprotein levels, elevated plasma von
Willebrand factor, inhibition of prostacyclin synthesis,
elevated plasma fibrinogen levels, and increased platelet adhesiveness.71 When peripheral arterial insufficiency complicates neuropathy there is a 10-fold risk of
ulceration progressing to infection, gangrene, and
amputation.72
Peripheral sensory neuropathy affects 50% of diabetic
patients and is attributed to chronic hyperglycemia. It
is a major cause of foot ulceration and lower limb amputation. In addition to poor glucose control, traditional
cardiovascular risk factors for macrovascular disease
are independent-risk factors for incident peripheral
neuropathy. In addition, data from the EURODIAB
cohort suggest that female sex may be an independentrisk factor. This makes it difficult to identify specific
diabetes components of neuropathy.73 Nerve injuries of
the foot can also be caused by penetrating wounds. The
sequelae of such injuries depend on the nerve injured
and the level of the injury. In the foot and ankle, the
main function of the nerves is to provide sensation.
Generally, the tibial nerve and its branches (i.e., the
medial and lateral plantar nerves) innervate the intrinsic musculature, although the deep peroneal nerve
innervates the extensor digitorum brevis and extensor
hallucis brevis muscles (Fig. 14.12). Denervation of these

A pressure ulcer is a localized injury to the skin or


underlying tissue, usually over a bony prominence, as
a result of unrelieved pressure. Pressure ulcers or pressure sores represent a common health problem, particularly among the physically limited or bedridden elderly.
Pressure ulcers on the buttocks affect nearly all wheelchair users. Pressure ulcer is a general term covering a
number of different tissue injuries, from superficial heel
sores to deep pressure sores under the buttocks. Because
compressive forces, shearing forces, and/or friction are
the major underlying causes some call them decubitus
ulcer.75 Pressure ulcers occur in approximately 515% of
patients in home care, healthcare facilities, and hospitals. Pressure ulcers are painful, decrease the quality of
life, increase susceptibility to infections, risk of death
and nursing workload, and create significant costs.76
Predisposing factors are classified as intrinsic (e.g.,
limited mobility, poor nutrition, comorbidities, aging
skin) or extrinsic (e.g., pressure, friction, shear, moisture). When an ulcer occurs, documentation of each
ulcer (i.e., size, location, eschar and granulation tissue,
exudate, odor, sinus tracts, undermining, and infection)
and appropriate staging (I through IV) are essential to
the wound assessment.77
While there is little doubt that pressure ulcers are
related to the mechanical insult of soft tissue, several
hypotheses have been developed pertaining to the link
between mechanical loading and tissue necrosis. The
two major hypotheses deal with tissue deformation and
ischemia.78 Pressure-induced soft-tissue deformation
causes cell necrosis. There is a time/strain relationship,
meaning that time does indeed play a role. In other

256

14 Wound healing

words, external tissue loading for a long time or on a


chronic basis results in ulcer formation. Prevention
includes identifying at-risk persons and implementing
specific prevention measures such as following a patientrepositioning schedule, keeping the head of the bed at
the lowest safe elevation to prevent shear, and using
pressure-reducing surfaces. Ischemia is a state caused
by lack of blood supply to any given tissue. The hypothesis proposes that a mechanical loading of the tissue
impinges on the arterial blood vessels, thereby causing
local ischemia. Since cells depend on oxygen, heat, and
nutrients transported by the blood, they will become
hypoxic and subsequently necrotic.

ATP
O2
NO

Protein
synthesis

Mature collagen
Superoxide
anion radical

Neutrophil
Macrophage

Endothelial cells
Infectious
pathogens

H2O2

Ischemia and tissue oxygenation


Vascular complications commonly associated with
problematic wounds are primarily responsible for
wound ischemia. Limitations in the ability of the vasculature to deliver O2-rich blood to the wound tissue lead
to, among other consequences, hypoxia. Hypoxia is a
reduction in oxygen delivery below tissue demand,
whereas ischemia is a lack of perfusion, characterized
not only by hypoxia but also by insufficient nutrient
supply.79 Hypoxia, by definition, is a relative term. It is
defined by a lower tissue partial pressure of oxygen
(pO2) compared to the pO2 to which the specific tissue
element in question is adjusted under healthy conditions in vivo. Depending on the magnitude, cells confronting hypoxic challenge either induce an adaptive
response that includes increasing the rates of glycolysis
and conserve energy or undergo cell death. Generally,
acute mild to moderate hypoxia supports adaptation
and survival. In contrast, chronic extreme hypoxia leads
to tissue loss.
While the tumor tissue is metabolically designed to
thrive under conditions of hypoxia, hypoxia of the
wound primarily caused by vascular limitations is
intensified by coincident conditions (e.g., infection,
pain, anxiety and hyperthermia) and leads to poor
healing outcomes. Oxygen and its reactive derivatives
(Fig. 14.13) are required for oxidative metabolismderived energy synthesis, protein synthesis, and the
maturation (hydroxylation) of extracellular matrices
such as collagen. Molecular oxygen is also required for
nitric oxide (NO) synthesis which in turn plays a key

Red blood
cells

Redox
signals
HOCl

Fig. 14.13 Molecular oxygen and its derivatives in wound healing. ATP, adenosine
triphosphate; NO, nitric oxide.

role in the regulation of vascular tone as well as in angiogenesis. In a wound setting, large amounts of molecular oxygen are partially reduced to form reactive oxygen
species (ROS). ROS includes oxygen free radicals such
as superoxide anion as well as its nonradical derivative,
hydrogen peroxide (H2O2). Superoxide anion radical is
the one-electron reduction product of oxygen. NADPH
oxidases represent one major source of superoxide anion
radicals at the wound site. NADPH oxidases in phagocytic cells help fight infection. Superoxide anion also
drives endothelial cell signaling such as required during
angiogenesis. In biological tissues, superoxide anion
radical rapidly dismutates to hydrogen peroxide, either
spontaneously or facilitated by enzymes called superoxide dismutases. Endogenous hydrogen peroxide
drives redox signaling, a molecular network of signal
propagation that supports key aspects of wound healing
such as cell migration, proliferation, and angiogenesis.
Neutrophil-derived hydrogen peroxide may be utilized
by MPO to mediate peroxidation of chloride ions, resulting in the formation of hypochlorous acid (HOCl), a
potent disinfectant (Fig. 14.13).
Three major factors may contribute to wound tissue
hypoxia: (1) peripheral vascular diseases garroting O2
supply; (2) increased O2 demand of the healing tissue;

Ischemia and tissue oxygenation

Stratum corneum
Stratum
granulosum
Keratinocyte in
epidermis

257

Bacteria

Basal layer
Myofibroblast
Endothelial
progenitor cell

Endothelial
cell
Dermis

Pocket of oxygenation
Pocket of hypoxia
Near-anoxic necrotic core

and (3) generation of ROS by way of respiratory burst


and for redox signaling.80 Other related factors, such as
arterial hypoxia (e.g., pulmonary fibrosis or pneumonia,
sympathetic response to pain, hypothermia, anemia
caused by major blood loss, cyanotic heart disease, high
altitude), may contribute to wound hypoxia as well.
Depending on factors such as these, it is important to
recognize that wound hypoxia may range anywhere
from near anoxia to mild to modest hypoxia. In this
context it is also important to appreciate that point
measurements performed in the wound tissue may not
provide a complete picture of the wound tissue biology
because it is likely that the magnitude of wound hypoxia
is not uniformly distributed throughout the affected
tissue, especially in large wounds. This is most likely
the case in chronic wounds presented clinically as
opposed to experimental wounds which are more controlled and homogeneous in nature. In any single
problem wound presented in the clinic, it is likely that
there are pockets of near anoxia as well as that of different grades of hypoxia (Fig. 14.14). As the weakest link
in the chain, tissue at the near-anoxic pockets will be
vulnerable to necrosis which in turn may propagate
secondary tissue damage and infection. Pockets of
extreme hypoxia may be flooded with hypoxia-inducible
angiogenic factors but would fail to vascularize functionally because of insufficient O2 that is necessary to
fuel the repair process. Indeed, uncontrolled expression

Neutrophil

Macrophage

Fig. 14.14 Heterogeneous distribution of oxygen in the


wound tissue: pockets of graded levels of hypoxia. Shade
of blue represents graded hypoxia. Shade of red or pink
represents oxygenated tissue. Tissue around each blood
vessel is dark pink in shade, representing regions that are
well oxygenated (oxygen-rich pockets). Bacteria and
bacterial infection are presented by shades of green on the
surface of the open wound.

of VEGF and its receptors leads to insufficient skin angiogenesis.81 Whether cells in the pockets of extreme
hypoxia are O2-responsive is another concern. Even if
such cells may have passed the point of no return in the
survival curve, correction of tissue oxygenation is likely
to help clean up the dead or dying tissue and replace
the void with proliferating neighboring cells. Pockets of
moderate or mild hypoxia are likely to be the point of
origin of successful angiogenic response as long as other
barriers such as infection and epigenetic alterations are
kept to a minimum.

Limited supply and high demand:


the oxygen imbalance
Peripheral vascular disease can affect the arteries and
the veins as well as the lymph vessels. The most common
and important type of peripheral vascular disease is
peripheral arterial disease, or PAD, which affects about
8 million Americans. The ankle brachial pressure index
represents a simple noninvasive method to detect arterial insufficiency within a limb. Arterial diseases, especially those associated with diabetes, represent a major
complicating factor in wound healing. PAD is the only
identifiable etiology in approximately 10% of leg ulcers.
In an ischemic limb, peripheral tissues are deprived of
blood supply as PAD progresses, causing tissue loss,
ulcers, and gangrene.

258

14 Wound healing

Venous insufficiency, on the other hand, is the root


cause of most leg ulcers. Chronic venous insufficiency,
characterized by the retrograde flow of blood in the
lower extremity, is associated with changes in the venous
wall and valves generally caused by inflammatory disorders induced by venous hypertension and associated
fluid shear stress. Factors causing arterial hypoxemia
may also limit O2 supply to the wound tissue.
Compromised pulmonary health, loss of hepatic function, hemodialysis, anemia, altitude hypoxemia, nitroglycerin therapy, nasal packing, critical illness, pain,
and hypothermia are examples of conditions associated
with arterial hypoxemia. Vasoconstricting drugs may
contribute to tissue hypoxia as well.79
Increased energy demand of the healing tissue leads
to a hypermetabolic state wherein additional energy is
generated from oxidative metabolism, increasing the O2
demand of the healing tissue. Adenosine triphosphate
(ATP) thus generated powers tissue repair. At the injury
site, extracellular ATP may be contributed by platelets
and other disintegrating cells. Extracellular ATP liberated during hypoxia or inflammation can either signal
directly to purinergic receptors or, after phosphohydrolytic metabolism, can activate surface adenosine receptors. Purinergic signaling may influence numerous
aspects of wound biology, including immune response,
inflammation, vascular as well as epithelial biology.
ATP may be immunostimulatory or vice versa, depending on extracellular concentrations as well as on expression patterns of purinergic receptors and ectoenzymes.
Extracellular ATP induces receptor activation in epithelial cells. ATP, released upon epithelial injury, acts as
an early signal to trigger cell responses, including an
increase in heparin-binding epidermal growth factor
(EGF)-like growth factor shedding, subsequent transactivation of the EGF receptor and its downstream signaling, resulting in wound healing. ATP released from the
injured epithelial cells is now known also to turn on
NADPH oxidases, the activity of which is critically
required to produce the redox signals required for
wound healing.80 Human endothelial cells are rich in
purinergic receptors and therefore responsive to extracellular ATP as well. ATP induces endotheliumdependent vasodilation. Both ATP as well as adenosine
regulate smooth muscle and endothelial cell proliferation. Recognizing that hypoxia limits ATP synthesis in
the ischemic wound tissue, therapeutic ATP delivery

systems have been studied for their effect on wound


healing. While these approaches may compensate for
the deficiency of ATP per se in the ischemic wound
tissue, they will fail to address the other essential functions of O2 and its derivatives in wound healing, as
discussed below.
Absolute requirements for O2 arise in several points
along the angiogenic sequence. For instance, all vessels
require a net or sheath of ECM, mainly collagen and
proteoglycans, to guide tube formation and resist the
pressures of blood flow. Conditions for collagen deposition and polymerization can be created only if molecular O2 is available to be incorporated into the structure
of nascent collagen by prolyl- and lysyl hydroxylases.
Without the obligatory extracellular hydroxylated collagen, new capillary tubes assemble poorly and remain
fragile.8284 This has a convincing clinical correlate in
scurvy, i.e., ascorbate deficiency. Scurvy results from
insufficient intake of ascorbate which is required for
correct collagen synthesis in humans. Ascorbate is
required for the posttranslational hydroxylation of collagen that enables the matured collagen molecules to
escape to the extracellular space and provide the necessary tensile strength. In scurvy, the collagenous sheath
cannot form because, under ascorbate-deficient conditions, collagen cannot be hydroxylated. Consequently,
new vessels fail to mature. Older vessels weaken and
break, and wounds fail to heal. Thus, while hypoxia is
a proved instigator of molecular signals for angiogenesis, it is also a proven enemy of vessel growth itself in
nontumor tissues. Collagen deposition proceeds in
direct proportion to pO2 across the entire physiologic
range, from zero to hundreds of mmHg. The Km for
O2 for this reaction is approximately 25 and the Vmax
is approximately 250mmHg, suggesting that new
vessels cannot even approach their greatest possible
rate of growth unless the wound tissue pO2 is high.85
Angiogenesis is directly proportional to pO2 in injured
tissues.83 Hypoxic wounds deposit collagen poorly and
become infected easily, both of which are problems of
considerable clinical significance.79

Redox signaling
Additional high demand for oxygen is placed by a
family of enzymes known as NADPH oxidases, which
are known to be highly active at the wound site.86 Recent

MicroRNAs

work has identified that oxygen is not only required to


disinfect wounds and fuel healing but that oxygendependent redox-sensitive signaling processes represent an integral component of the healing cascade.80 The
widely held notion that biological free radicals are
necessarily agents of destruction is now facing serious
challenge.87 Over a decade ago it was proposed that in
biological systems oxidants are not necessarily always
the triggers for oxidative damage and that oxidants
such as H2O2 could actually serve as signaling messengers and drive several aspects of cellular signaling.88
Today, that concept is much more developed and mature.
Evidence supporting the role of oxidants such as H2O2
as signaling messenger is compelling.8999

Nitric oxide
At the wound site, NO is generated by an oxygendependent biosynthetic process. In the late 1970s,
research was unfolding that implicated NO involvement in the process of vasodilation. By 1986, research
culminated in the identification of NO as the endotheliumderived relaxing factor responsible for the maintenance
of vascular tone, thus implicating NO as a potential
wound-healing agent.100 Maximal NO synthase activity
is noted early in cutaneous wound healing, with sustained production up to 10 days after wounding. Wound
macrophages represent a major source of NO production in the early phase of wound healing.101 Inhibition
of wound NO synthesis lowered wound collagen accumulation and wound-breaking strength, suggesting that
NO synthesis is critical to wound collagen accumulation
and acquisition of mechanical strength. Later it was
demonstrated that wound fibroblasts are phenotypically altered during the healing process to synthesize
NO, which, in turn, regulates their collagen synthetic
and contractile activities.102 The blockade of NO synthesis impairs cutaneous wound healing, acting in early
and late phases of wound repair.103 Interestingly,
impaired diabetic wound healing is associated with
decreased wound NO synthesis.104

MicroRNAs
Wound healing is largely dependent on injury-inducible
protein-coding genes as they serve as drivers of an

259

inherent tissue repair program that seeks to restore the


injured tissue both structurally as well as functionally.
There are two key steps that separate a protein-coding
gene from its corresponding protein. First, the DNA
hosting the gene must transcribe to mRNA. Finally, the
mRNA must be translated to protein. Work emerging
during recent years demonstrates that both of these
critical steps are subject to robust and redundant regulation by microRNAs (miRNAs: 1922 nucleotides long),
which are noncoding RNAs found in all eukaryotic
cells. Work during the past decade recognizes small
RNAs as a new class of regulators of eukaryotic biology.
Alongside other small interfering RNAs (siRNAs),
miRNAs execute posttranscriptional gene silencing
through mRNA destabilization as well as translational
repression. In simple words, whether a gene would
code a protein or not is decided upon by miRNAs for
which the gene is a target. miRNAs form basepairs with
specific sequences in protein-coding mRNAs. Nearperfect pairing induces cleavage of the target mRNA,
whereas partial pairing results in translational repression and mRNA decay through deadenylation pathways.105 According to the miRbase database, the human
genome encodes 1048 miRNAs. This count is rapidly
growing. These miRNAs may regulate more than a third
of all protein-coding genes and virtually all biological
processes. Mammalian cells express cell type-specific
miRNAs which silence unique subsets of target genes
within the cell. While miRNAs are mostly known for
being functional in the cytoplasm, nuclear miRNAs may
also participate in gene regulation. Initially considered
an oddity, miRNA-dependent control of gene expression is now accepted as being integral to the normal
function of cells and organisms. miRNAs are emerging
as key regulators of the overall wound-healing process.106

Inflammation
Disruption of miRNA biogenesis has a major impact on
the overall immune system. Emerging studies indicate
that miRNAs, especially miR-21, miR-146a/b, and miR155, play a key role in regulating several hubs that
orchestrate the inflammatory process.107 miRNAs have
been directly implicated in the pathogenesis of inflammatory diseases such as osteoarthritis and rheumatoid
arthritis. Resolvin-regulated specific miRNAs target
genes involved in resolution of inflammation establish

260

14 Wound healing

a novel resolution circuit involving RvD1 receptordependent regulation of specific miRNAs.108 Fur
thermore, the brain-specific microRNA-124 can tame
inflammation by turning off activated microglial cells
and macrophages.109 Of relevance to tissue repair is also
the regulatory loop where cytokines, including those
elicited following injury, are regulated by miRNAs as
well as regulate miRNA expression.110,111

Angiogenesis
In 20052008, the first series of observations establishing
key significance of miRNAs in the regulation of mammalian vascular biology came from experimental studies
involved in arresting miRNA biogenesis to deplete
the miRNA pools of vascular tissues and cells.112 Dicerdependent biogenesis of miRNA is required for blood
vessel development during embryogenesis. Mice with
endothelial cell-specific deletion of Dicer, a key enzyme
supporting biogenesis of miRNAs, display defective
postnatal angiogenesis. NADPH oxidase-derived ROS
drive wound angiogenesis. Endothelial NADPH oxidase
is subject to control by miRNAs.113 Hypoxia is widely
recognized as a cue that drives angiogenesis as part of
an adaptive response to vascularize the oxygen-deficient
host tissue. Hypoxia-sensitive miR-200b is involved in
such induction of angiogenesis via directly targeting
Ets-1.114 Various aspects of angiogenesis, such as proliferation, migration, and morphogenesis of endothelial
cells, can be regulated by specific miRNAs in an
endothelial-specific manner. miRNAs known to regulate angiogenesis in vivo are referred to as angiomiRs.115
miRNA-126 is specific to endothelial cells and regulates
vascular integrity and developmental angiogenesis.
Manipulating angiomiRs in the setting of tissue repair
represents a new therapeutic approach that could be
effective in promoting wound angiogenesis.

Hypoxia response
Tissue injury is often associated with disruption of vascular supply to the injury site. Thus, the injured tissue
often suffers from insufficient oxygen supply or hypoxia.
Under conditions of additional underlying ischemia,
hypoxia is severe and seriously limits wound healing.79
Hypoxia induces specific miRNAs, collectively referred
to as hypoxamirs.116 miRNA-210 is a classical

hypoxamir. Expression of hypoxia-inducible factor 1


(HIF-1) is also controlled by specific miRNAs. In turn,
HIF-1 controls the expression of hypoxamirs, which
are induced in the injured tissue.117 Hypoxamirs are
also induced by HIF-independent pathways. Although
hypoxamirs generally favor angiogenesis, their metabolic and cell cycle arrest functions are in conflict with
wound healing, especially in an ischemic setting.
Silencing specific hypoxamirs may therefore represent a
prudent approach to facilitate tissue repair. miRNA-210
represses mitochondrial respiration116 and exaggerates
production of undesired mitochondrial ROS.118 These
outcomes are not compatible with the higher energy
demands associated with tissue repair. miR-210 also
silences signaling via FGF,119 a key contributor to wound
healing. The injured tissue is highly rich in ROS.86 In
addition, at the site of injury transition metal ions are
released from a protein-bound state. Conditions such as
these cause DNA damage which opposes tissue repair.
DNA repair systems are therefore of key significance in
enabling tissue repair. miR-210 antagonizes DNA
repair.120 This is another hypoxamir function that is in
conflict with wound healing. Compatible with the
common observation that ischemic wounds are refractory to healing response, elevated miRNA-210 in
ischemic wounds attenuated keratinocyte proliferation
and impaired wound closure.106,121

Stem cells
Endogenous miRNA-binding sites have been identified
in murine embryonic stem cells (ESCs). miRNAs govern
ESCs function by serving as control hubs managing
regulatory networks. A central importance of such governance is highlighted by the observation that ESCs
lacking miRNAs lose their stemness. ESCs with deficient miRNA biogenesis systems switch to a mode of
ongoing cell division. They do not differentiate on
demand because of failure to turn off the pluripotency
regulatory program.122 miRNAs conduct the orchestra
of critical gene regulatory networks controlled by
pluripotency factors within stem cells. Individual
miRNA-dependent pathways that promote the reprogramming of somatic cells into induced pluripotent
stem (iPS) cells have been now identified. Manipulation
of specific cellular miRNAs helps enhance reprogramming of somatic cells to an ESC-like phenotype, helping

Stem cells

to generate iPS cells.123 Expression of miRNAs is also


subject to control by epigenetic factors.124 Such control
influences the balance between proliferation and differentiation of stem cells. In executing such control, the
miRNA element of epigenetics cross-talks with changes
in chromatin structure as well as with changes in DNA
methylation. Collectively, this provides for a mechanism by which the tissue injury microenvironment can
influence miRNA-dependent reparative and regenerative processes.

Stem cells

261

Totipotent cells

Pluripotent stem cell

Blood
stem cells

Other committed
stem cells

Erythrocytes

Platelets

White blood cells

Specialized cells

Fig. 14.15 Stem cell renewal.

on
ati
nti
re
tio
n

iffe
-d

tia
en
er
iff
-d
De

on
n

io

iat

nt

Pluripotent
stem cell

re

tro

ati

ffe

Re

nti

re

iffe

-d

Re

Totipotent
cells

Di

The regenerative potential of injured adult tissue suggests the physiological existence of cells capable of participating in the reparative process. The epithelium of
the skin, the epidermis, is in a continuous equilibrium
of growth and differentiation and has the remarkable
capacity to self-renew completely, which relies on reservoirs of stem cells. In mammals, there are two broad
types of stem cells: ESCs that are isolated from the inner
cell mass of blastocysts, and adult stem cells that are
found in various tissues. In adult organisms, stem cells
and progenitor cells act as a repair system for the body,
replenished in adult tissues. Stem cells have the property of self-renewal without differentiation and have the
potential to differentiate into any type of cell. Totipotent
stem cells have the ability to give rise to a whole organism due to their ability to differentiate into embryonic
and extraembryonic cells. Pluripotent stem cells are
derived from totipotent cells and can differentiate into
all cell types but cannot give rise to a new organism (Fig.
14.15). ESCs are derived from the developing embryo,
usually from the inner cell mass of a blastocyst or earlier
morula stages. Lost or damaged cells can be replaced by
differentiation, dedifferentiation, or transdifferentiation
(Fig. 14.16). Recent advances have shown that the addition of a group of genes can not only restore pluripotency in a fully differentiated cell state (differentiation)
but can also induce the cell to proliferate (dedifferentiation) or even switch to another cell type (transdifferentiation). Dedifferentiation is represented by a terminally
differentiated cell reverting back to a less-differentiated
stage from within its own lineage. This process allows
the cell to proliferate again before redifferentiating,
leading to the replacement of those cells that have been

Progenitor cells
Blood
stem
cells
Other
tissues

Red blood
cells

White blood
cells

Muscle

Nerve

Bone

Trans-differentiation

Fig. 14.16 Stem cell differentiation, dedifferentiation, and transdifferentiation.

lost. Transdifferentiation is another naturally occurring


mechanism that takes dedifferentiation a step further
and sees cells regressing to a point where they can
switch lineages, allowing them to differentiate into
another cell type. Furthermore, reprogramming aims to
revert differentiated cells to pluripotency. From here,
they can differentiate into almost any cell type. Although
reprogramming occurs naturally during fertilization to
produce totipotent cells that can differentiate into any

14 Wound healing

262

Hair shaft

Skeletal
muscle

Liver

Sebaceous gland
Outer root sheath

Bulge stem cells

Inner root sheath

Basement
membrane

Bone marrow
stromal cell
Epithelial
cell
Blood cells

Matrix stem cells


Dermal papilla

Neuron

TA cell

Glial cell
Blood
vessel

Basement membrane

CNS
stem cells
Fat cell

Paneth cell
Stem cell

Cardiac muscle
B

Mesenchymal cell

Fig. 14.17 Bone marrow stem cells. CNS, central nervous system.

cell type, it has not yet formally been shown to be a


genuine regenerative response. Furthermore, reprogramming sidesteps the necessity of using embryos
for regenerative therapies by using differentiated cells
taken from a patient. From a clinical perspective, this
comes with the additional bonus of circumventing
the immunological problems such as transplant rejection and graft-versus-host disease associated with
engraftment.125
The bone marrow (Fig. 14.17), home of stem and progenitor cells, contributes a significant proportion of cells
in the skin. The normal skin has long been known to
contain bone marrow-derived cells that are involved in
host defense and inflammatory processes, including
wound healing. However, recent studies demonstrate
that the bone marrow contributes not only inflammatory cells, but also keratinocytes and fibroblast-shaped
cells to the skin. Similar to leukocytes in trafficking,

Fig. 14.18 Stem cells in the epidermis. Hair follicles act as reservoirs of stem
cells in the skin. (A) Cross-sectional view of a hair follicle. The matrix stem cells
differentiate into various parts of the hair while the long-term stem cells are present
in the bulge region. The stem cells from the bulge maintain the sebaceous gland
and epidermal stem cells. (B) A mammalian gut crypt. Stem cells are located at the
basal region with the Paneth cells. The transit amplifying (TA) cells are stem cell
progeny which move upwards and differentiate.

stem/progenitor cells derived from the bone marrow


could home to injured tissues and participate in the
repair/regeneration. Moreover, culture-expanded bone
marrow-derived MSCs have been shown to promote the
healing of diabetic wounds, implying a profound therapeutic potential for skin defects such as chronic wounds
and burns.126
During the inflammatory phase, leukocytes migrating to the wound site are hematopoietic cells derived
from the bone marrow. In the skin the bulge of the hair
follicle (Fig. 14.18) plays a role as a reservoir of stem
cells. In mice it has been shown that the bulge region
contains hematopoietic cells that are identical to the

Stem cells

bone marrow-derived cells and also to those found in


fetal circulation.127 Moreover this region also serves as a
reservoir for precursors of mast cells. Discovery of these
epidermal stem cells in the hair follicle bulge led to the
hypothesis that these cells are necessary for both epidermal renewal as well as wound healing.128 It was shown
that cells from the bulge do not contribute to epidermal
regeneration; however upon injury cells from the bulge
are recruited into the epidermis and migrate in a linear
manner toward the center of the wound.129 These cells
were noted to be transient, living only for a few weeks,
thus representing an acute response to injury.
There are two main branches of stem cells in the bone
marrow, including hematopoietic stem cells (HSCs) and
MSCs. Adult bone marrow-derived HSCs have long
been recognized as a precursor to all blood cell lineages,
including erythrocytes, platelets, and white blood cells.
Additionally, HSCs may also give rise to fibrocytes and
endothelial progenitor cells. Circulating endothelial
precursor cells play a role in neoangiogenesis which is
essential for healing.130,131 Bone marrow-derived stem
cells also contribute to the deposition of collagen III in
the wound132 and differentiate into fibroblasts,133 keratinocytes,134 and fibrocytes.135 Bone marrow-derived
MSCs, which are also referred to as bone marrow mesenchymal stromal cells or marrow stromal cells, are selfrenewing and expandable stem cells. Although present
as a rare cell population in the bone marrow, representing about 0.0010.01% of the nucleated cells, about
10-fold less abundant than HSCs, MSCs are expandable
in culture and multipotent, capable of differentiating
into several cell types.
Because of their properties of regeneration and differentiation the use of stem cells to heal problem wounds
has long been of interest. Indeed, autologous bone
marrow aspirates and cultured cells were helpful in
healing chronic wounds.136 In the case of burn wounds,
bone marrow-derived stem cell treatment shows
promise as well.137 Another major source of adult stem
cells is adipose tissue.138 The capability of adiposederived adult stem cells to differentiate into bone,
muscle, fat, or cartilage, or into cells of mesenchymal
lineage, makes them a prime target for therapeutic use.
It has been shown that adult stem cells enhanced wound
healing in a murine model.139,140
Hair follicles are an integral part of mammalian skin
where they help the epidermis to maintain the bodys

263

protective barrier against its external environment. With


rare exceptions (such as palms and eyelids), hair follicles are present all over the skin and play an important
role in physiological tissue renewal and regeneration
after injury. Hair follicles represent an autonomous miniorgan, which provides an excellent model system for
studying the biology of adult stem cells.141 There are
obvious differences between human and mouse skin,
yet the knowledge gained from lineage-tracing experiments in mice has greatly expanded our understanding
of the cellular behavior of the different keratinocyte
populations during physiological and injury-induced
tissue regeneration. As in most of the bodys organs, the
skin experiences constant renewal. The upper keratinized layer of the interfollicular epidermis, which consists of terminally differentiated cells, is shed and
replaced by cells originating from the actively proliferating layer beneath. In contrast, hair follicles undergo
cycles of growth (anagen) and rest (telogen). The potential for proliferation to sustain the lifelong replenishment of normal cell loss and to repair occasional tissue
damage lies within the population of epidermal stem
cells. The hair follicle bulge harbors stem cells that may
help in renewal and repair of the skin. The term bulge,
originally called der Wulst, was introduced in 1903 by
a German morphologist, P. Sthr, to describe an eminent
structure at the site of attachment of the arrector pili
muscle in human hair follicles.141 Similar to many other
somatic stem cells, bulge cells are slow-cycling in nature.
This feature has permitted their initial identification and
isolation as label-retaining cells that can retain a pulse
of nucleotide label following a long chase period and
is frequently regarded as a defining characteristic of
the hair follicle stem cell. In addition, the availability
of several immunohistochemical markers, including
keratin-15 and CD34, that specifically label murine follicular stem cells has given researchers the ability to
examine carefully the signals required for adult stem
cell activation and renewal. We now know that the
bulge serves as a repository of long-lived multipotent
stem cells, imparted with the capacity to differentiate
into all cell types that constitute the lower cyclic portion
of the hair follicle, as well as the interfollicular epidermis during wound repair.142,143
Although research into the use of stem cells for regenerative medicine is on a steep upward slope, clinical
success has not been as forthcoming. This has been

264

14 Wound healing

primarily attributed to a lack of information on the basic


biology of stem cells, which remains insufficient to
justify clinical studies. Since most clinical protocols use
intravenous application of MSCs, it has become important to understand their trafficking in the blood stream.
Moreover, since relatively little is known where the
transplanted MSCs might locate, a better understanding
of involved homing mechanisms will likely shed light
on how MSCs exert their therapeutic effects. For
instance, it is unclear whether mechanisms used at
injured sites are location-specific or whether this recruitment can be modulated for therapeutic purposes. In
addition, it has recently been suggested that platelets
may play an important role in stem cell recruitment to
sites of injury. A better understanding of the mechanisms used by stem cells during tissue homing would
allow us to develop strategies to improve recruitment
of these rare cells.144

Induced pluripotent stem cells


Pluripotent stem cells possess the unique property of
differentiating into all other cell types of the human
body. The discovery of iPS cells in 2006 has opened up
new avenues in clinical medicine.145,146 Recent breakthrough studies using a combination of four factors to
reprogram human somatic cells into pluripotent stem
cells without using embryos or eggs have led to an
important revolution in stem cell research. It is now
possible to convert somatic cells, such as skin fibroblasts
and B lymphocytes, into pluripotent stem cells that
closely resemble ESCs. Recently, functional neurons,
cardiomyocytes, pancreatic islet cells, hepatocytes, and
retinal cells have been derived from human iPSCs, thus
reconfirming the pluripotency and differentiation capacity of these cells. These findings further open up the
possibility of using iPSCs in cell replacement therapy
for various disorders, including chronic wounds.

Scar
Scars (also called cicatrices) are macroscopic fibrous
tissue that visibly replaces normal skin after injury.
There is a wide spectrum of skin scarring postwounding, including scarless fetal wound healing, fine-line
(normal) scars, stretched (widespread) scars, atrophic

(depressed) scars, scar contractures, hypertrophic scars,


and keloids. Postsurgical scar assessment is fundamental for a complete functional evaluation and as an
outcome measure. The Vancouver Scar Scale is the most
widely used rating scale for scars but the Patient and
Observer Scar Assessment Scale is recognized as the
most comprehensive tool, taking into account the important aspect of the patients perspective. Recently, a new
scale, called the Stony Brook Scar Evaluation Scale, has
been proposed.147 Although scar remodeling occurs for
months to years after the initial injury, complete restoration of the normal ECM architecture is never achieved.
Mature scars restore only 70% of the tensile strength of
normal skin, and prescar function is never completely
recovered.148
Scarring is an integral component of the healing
process and an outcome of the remodeling stage of
wound repair which begins 23 weeks after injury and
lasts for a year or more.149 During this stage, all of the
processes activated after injury wind down and cease.
Most of the endothelial cells, macrophages, and myofibroblasts undergo apoptosis or exit from the wound,
leaving a mass that contains few cells and consists
mostly of collagen and other ECM proteins.149,150
Epithelialmesenchymal interactions probably con
tinuously regulate skin integrity and homeostasis. In
addition, over 612 months, the acellular matrix is
actively remodeled from a mainly type III collagen
backbone to one predominantly composed of type I collagen.151 This process is carried out by matrix metalloproteinases that are secreted by epidermal cells,
endothelial cells, fibroblasts, and the macrophages
remaining in the scar, and it strengthens the repaired
tissue. However, the tissue never regains the properties
of uninjured skin.
Fibroblasts at the wound site originating from surrounding tissues as well as supplied in the form of
marrow-derived blood-borne cells are recognized as the
primary drivers of scar formation. Platelets, macrophages, T lymphocytes, mast cells, Langerhans cells,
and keratinocytes are directly and indirectly involved
in the activation of fibroblasts, which in turn produce
excess ECM. Dermal scarring can result in loss of function, movement restriction, and disfigurement. The
mechanism of scar formation involves inflammation,
fibroplasia, formation of granulation tissue, and scar
maturation. In response to tissue injury, the acute

Scar

265

inflammatory response is followed by the proliferation


of fibroblasts, which are cells responsible for synthesizing various tissue components, including collagen
and fibrin. During the acute inflammatory phase, circulating progenitor cells migrate to injured tissue. Rapid
cellular proliferation occurs, which ultimately results in
the formation of new blood vessels and epithelium.
Fibroblasts then differentiate into myofibroblasts, which
are the cells responsible for collagen deposition and
wound contraction. Scar formation ultimately results
from excess accumulation of an unorganized ECM.148,152
Sensory nerves, including nonmyelinated C fibers
and delta fibers, traverse all cutaneous layers, including
the epidermis. These fibers run parallel to capillaries
coursing around follicular complexes. A higher density
of nerve fiber is associated with scar tissue. Unlike
diabetic tissues, hypertrophic scar has excessive
neuropeptide activity. In addition, proinflammatory
substance P concentration is greater in the hypertrophic
scar samples when compared with normal uninjured
skin. High substance P and low neutral endopeptidase activity in scar tissue is believed to induce an
exuberant neuroinflammatory response contributing to
scar formation.153

Hypertrophic scar

Keloid

Regenerative fetal healing

Keloid scarring, also known as keloid disease, is a locally


aggressive benign fibroproliferative scar that continually grows beyond the confines of the original wound
and invades into surrounding healthy skin. Unlike
hypertrophic scars which stay within the boundaries of
the original wounds and usually regress spontaneously,
keloids grow beyond the boundaries of the original
wounds and rarely regress. These pathological scars are
not only aesthetically displeasing, but can also be both
painful and functionally disabling, causing patients
both physical and psychological distress. There is a
strong genetic predisposition to keloid disease. First,
keloids are more common in ethnicities with darkerpigmented skins and have been reported to be 515
times more prevalent in blacks than in whites. Familial
heritability and prevalence in twins also support the
concept of the genetic susceptibility to keloid scarring.
The major pathways involved in keloid disease include
apoptosis, mitogen-activated protein kinase, TGF-,
IL-6, and plasminogen activator inhibitor-1.154

In contrast to adult wound healing, early-gestation fetal


skin wound healing occurs rapidly, in a regenerative
fashion, and without scar formation. The accelerated
rate of healing, relative lack of an acute inflammatory
response, and an absence of neovascularization distinguish fetal from adult wound healing.156 Scarless wound
healing has been observed in the fetuses of mice, rats,
pigs, monkeys, and humans. Fetal skin heals scarlessly
before a certain gestational age, after which point typical
scar formation occurs. In humans, scarring of wounds
begins at approximately 24 weeks of gestation, whereas
in mice scarring of wounds begins on embryonic day
18.5 (average gestation period for mice is 20 days). This
transition point, however, is modulated by wound size.
For example, as wound size increases in fetal lambs, the
ability to heal scarlessly is lost earlier during gestation.148 In response to tissue injury, the fetal dermis has
the ability to regenerate a nondisrupted collagen matrix
that is identical to that of the original tissue. In addition,
dermal structures, such as sebaceous glands and hair

The pathophysiology of hypertrophic scar formation


involves a constitutively active proliferative phase of
wound healing.155 The scar tissue has a unique structural makeup that is highly vascular, with inflammatory
cells and fibroblasts contributing to an abundant and
disorganized matrix structure. The net result is that the
original skin defect is replaced by a nonfunctional mass
of tissue. Beyond these observations, investigations into
the pathophysiology of the disease have been limited by
the absence of a practical animal model and have relied
upon the use of human pathological specimens. These
studies are problematic in that such specimens represent the terminal stages of the scarring process and may
not contain the initiating factors that originally led to
the development of the disease. While animal models
have provided some insight into the genetics and pathogenesis of cutaneous fibrosis, it is unclear how closely
the process of hypertrophic scarring in these models
resembles that seen in humans. Specifically, it is
unknown whether the same factors that initiate hypertrophic scarring in these species are involved in human
disease.155

266

14 Wound healing

follicles, form normally after fetal injury. Although the


exact mechanisms of scarless fetal wound healing are
still unknown, they are thought to be due to differences
between the ECM, inflammatory response, cellular
mediators, differential gene expression, and stem cell
function in fetal and postnatal wounds.148

Acknowledgment
Supported by NIH RO1 grants GM069589, GM077185,
NS42617, DK076566, and HL 073087. The authors thank
Sabyasachi Biswas PhD, Rashmet Reen PhD, and Viren
Patel MD for support in writing this article.

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


30. Martin P. Wound healing aiming for perfect skin
regeneration. Science. 1997;276:7581.
The healing of an adult skin wound is a complex process
requiring the collaborative efforts of many different tissues
and cell lineages. This review discusses the key signals and
processes that regulate the normal adult cutaneous wound
repair.
31. Martin P, Leibovich SJ. Inflammatory cells during
wound repair: the good, the bad and the ugly. Trends
Cell Biol. 2005;15:599607.
39. Khanna S, Biswas S, Shang Y, et al. Macrophage
dysfunction impairs resolution of inflammation in the
wounds of diabetic mice. PLoS One. 2010;5:e9539.
53. Carmeliet P. Angiogenesis in life, disease and
medicine. Nature. 2005;438:932936.
79. Sen CK. Wound healing essentials: let there be oxygen.
Wound Repair Regen. 2009;17:118.
86. Roy S, Khanna S, Nallu K, et al. Dermal wound
healing is subject to redox control. Mol Ther. 2006;13:
211220.
H2O2 has been shown to support wound healing by inducing
VEGF expression in human keratinocytes. This work
presents the first in vivo evidence indicating that strategies
to influence the redox environment of the wound site may
have a bearing on healing outcomes.
98. Sen CK. The general case for redox control of wound
repair. Wound Repair Regen. 2003;11:431438.
At very low concentrations, reactive oxygen species may
regulate cellular signaling pathways by redox-dependent

mechanisms. Redox-based strategies may serve as effective


adjuncts to jump-start healing of chronic wounds. The
review focuses on the understanding of wound site
redox biology and novel insights into the fundamental
mechanisms that would help to optimize conditions for
oxygen therapy.
106. Banerjee J, Chan YC, Sen CK. MicroRNAs in skin and
wound healing. Physiol Genomics. 2010.
MicroRNAs (MiRNAs) are small endogenous RNA
molecules about 22 nucleotides in length that are capable
of posttranscriptional gene regulation by binding to their
target messenger RNAs (mRNAs). This review focuses on
the role of miRNAs in cutaneous biology, the various
methods of microRNA modulation and the therapeutic
opportunities in treatment of skin diseases and wound
healing.
129. Ito M, Liu Y, Yang Z, et al. Stem cells in the hair
follicle bulge contribute to wound repair but not to
homeostasis of the epidermis. Nat Med. 2005;11:
13511354.
The discovery of long-lived epithelial stem cells in the bulge
region of the hair follicle led to the hypothesis that epidermal
renewal and epidermal repair after wounding both depend
on these cells. This paper discusses the implications of
epithelial stem cells for both gene therapy and developing
treatments for wounds.
149. Gurtner GC, Werner S, Barrandon Y, et al. Wound
repair and regeneration. Nature. 2008;453:
314321.

References

References
1. Mehra R. Historical survey of wound healing. Bull
Indian Inst Hist Med Hyderabad. 2002;32:159175.
2. Wangensteen OH. Surgeons and wound management:
historical aspects. Conn Med. 1975;39:568574.
3. Broughton 2nd. G, Janis JE, Attinger CE. A brief
history of wound care. Plast Reconstr Surg. 2006;117:
6S-11S.
4. Eming SA, Krieg T, Davidson JM. Inflammation in
wound repair: molecular and cellular mechanisms.
J Invest Dermatol. 2007;127:514525.
5. Sen CK, Gordillo GM, Roy S, et al. Human skin
wounds: a major and snowballing threat to public
health and the economy. Wound Repair Regen. 2009;17:
763771.
6. Mackman N, Tilley RE, Key NS. Role of the extrinsic
pathway of blood coagulation in hemostasis and
thrombosis. Arterioscler Thromb Vasc Biol. 2007;27:
16871693.
7. Schecter AD, Spirn B, Rossikhina M, et al. Release of
active tissue factor by human arterial smooth muscle
cells. Circ Res. 2000;87:126132.
8. Smyth SS, McEver RP, Weyrich AS, et al. Platelet
functions beyond hemostasis. J Thromb Haemost.
2009;7:17591766.
9. Huntington JA. Molecular recognition mechanisms of
thrombin. J Thromb Haemost. 2005;3:18611872.
10. Midwood KS, Williams LV, Schwarzbauer JE. Tissue
repair and the dynamics of the extracellular matrix.
Int J Biochem Cell Biol. 2004;36:10311037.
11. Sahni A, Odrljin T, Francis CW. Binding of basic
fibroblast growth factor to fibrinogen and fibrin. J Biol
Chem. 1998;273:75547559.
12. Tuan TL, Wu H, Huang EY, et al. Increased
plasminogen activator inhibitor-1 in keloid fibroblasts
may account for their elevated collagen accumulation
in fibrin gel cultures. Am J Pathol. 2003;162:15791589.
13. He S, Blomback M, Bark N, et al. The direct thrombin
inhibitors (argatroban, bivalirudin and lepirudin) and
the indirect Xa-inhibitor (danaparoid) increase fibrin
network porosity and thus facilitate fibrinolysis.
Thromb Haemost. 2010;103:10761084.
14. Marin V, Montero-Julian FA, Gres S, et al. The IL-6soluble IL-6Ralpha autocrine loop of endothelial
activation as an intermediate between acute and
chronic inflammation: an experimental model
involving thrombin. J Immunol. 2001;167:34353442.
15. Delavary BM, van der Veer WM, van Egmond M, et al.
Macrophages in skin injury and repair. Immunobiology.
2011.
16. Baum CL, Arpey CJ. Normal cutaneous wound
healing: clinical correlation with cellular and

17.

18.

19.
20.

21.

22.

23.
24.

25.

26.

27.

28.
29.

30.

266.e1

molecular events. Dermatol Surg. 2005;31:674686;


discussion 686.
Frank S, Kampfer H, Wetzler C, et al. Large induction
of the chemotactic cytokine RANTES during cutaneous
wound repair: a regulatory role for nitric oxide in
keratinocyte-derived RANTES expression. Biochem J.
2000;347(Pt 1):265273.
Maraganore JM. Thrombin, thrombin inhibitors, and
the arterial thrombotic process. Thromb Haemost.
1993;70:208211.
Klebanoff SJ. Myeloperoxidase: friend and foe. J Leukoc
Biol. 2005;77:598625.
Daley JM, Reichner JS, Mahoney EJ, et al. Modulation
of macrophage phenotype by soluble product(s)
released from neutrophils. J Immunol. 2005;174:
22652272.
Peters T, Sindrilaru A, Hinz B, et al. Wound-healing
defect of CD18(/) mice due to a decrease in TGFbeta1 and myofibroblast differentiation. EMBO J.
2005;24:34003410.
DiPietro LA. Wound healing: the role of the
macrophage and other immune cells. Shock.
1995;4:233240.
de Vries VC, Noelle RJ. Mast cell mediators in
tolerance. Curr Opin Immunol. 2010;22:643648.
Abraham SN, St John AL. Mast cell-orchestrated
immunity to pathogens. Nat Rev Immunol. 2010;10:
440452.
Trautmann A, Toksoy A, Engelhardt E, et al. Mast cell
involvement in normal human skin wound healing:
expression of monocyte chemoattractant protein-1
is correlated with recruitment of mast cells which
synthesize interleukin-4 in vivo. J Pathol. 2000;190:
100106.
Shiota N, Nishikori Y, Kakizoe E, et al.
Pathophysiological role of skin mast cells in wound
healing after scald injury: study with mast celldeficient W/W(V) mice. Int Arch Allergy Immunol.
2010;151:8088.
Gallant-Behm CL, Hildebrand KA, Hart DA. The
mast cell stabilizer ketotifen prevents development
of excessive skin wound contraction and fibrosis
in red Duroc pigs. Wound Repair Regen. 2008;16:
226233.
Noli C, Miolo A. The mast cell in wound healing.
Vet Dermatol. 2001;12:303313.
Eming SA, Werner S, Bugnon P, et al. Accelerated
wound closure in mice deficient for interleukin-10.
Am J Pathol. 2007;170:188202.
Martin P. Wound healing aiming for perfect skin
regeneration. Science. 1997;276:7581.
The healing of an adult skin wound is a complex process
requiring the collaborative efforts of many different tissues
and cell lineages. This review discusses the key signals and

266.e2

31.

32.

33.

34.

35.

36.

37.

38.

39.

40.

41.

42.

43.
44.
45.

14 Wound healing

processes that regulate the normal adult cutaneous wound


repair.
Martin P, Leibovich SJ. Inflammatory cells during
wound repair: the good, the bad and the ugly. Trends
Cell Biol. 2005;15:599607.
Rappolee DA, Mark D, Banda MJ, et al. Wound
macrophages express TGF-alpha and other growth
factors in vivo: analysis by mRNA phenotyping.
Science. 1988;241:708712.
Leibovich SJ, Ross R. The role of the macrophage in
wound repair. A study with hydrocortisone and
antimacrophage serum. Am J Pathol. 1975;78:71100.
Goren I, Allmann N, Yogev N, et al. A transgenic
mouse model of inducible macrophage depletion:
effects of diphtheria toxin-driven lysozyme M-specific
cell lineage ablation on wound inflammatory,
angiogenic, and contractive processes. Am J Pathol.
2009;175:132147.
Mirza R, DiPietro LA, Koh TJ. Selective and specific
macrophage ablation is detrimental to wound healing
in mice. Am J Pathol. 2009;175:24542462.
Martinez FO, Helming L, Gordon S. Alternative
activation of macrophages: an immunologic functional
perspective. Annu Rev Immunol. 2009;27:451483.
Grinberg S, Hasko G, Wu D, et al. Suppression of
PLCbeta2 by endotoxin plays a role in the adenosine
A(2A) receptor-mediated switch of macrophages from
an inflammatory to an angiogenic phenotype. Am J
Pathol. 2009;175:24392453.
Porcheray F, Viaud S, Rimaniol AC, et al. Macrophage
activation switching: an asset for the resolution of
inflammation. Clin Exp Immunol. 2005;142:481489.
Khanna S, Biswas S, Shang Y, et al. Macrophage
dysfunction impairs resolution of inflammation in the
wounds of diabetic mice. PLoS One. 2010;5:e9539.
Spite M, Serhan CN. Novel lipid mediators promote
resolution of acute inflammation: impact of aspirin and
statins. Circ Res. 2010;107:11701184.
Gordon A, Kozin ED, Keswani SG, et al. Permissive
environment in postnatal wounds induced by
adenoviral-mediated overexpression of the antiinflammatory cytokine interleukin-10 prevents scar
formation. Wound Repair Regen. 2008;16:7079.
Li P, Liu P, Xiong RP, et al. Ski, a modulator of wound
healing and scar formation in the rat skin and rabbit
ear. J Pathol. 2011;223:659671.
Siddiqui AR, Bernstein JM. Chronic wound infection:
facts and controversies. Clin Dermatol. 2010;28:519526.
White RJ. Wound infection-associated pain. J Wound
Care. 2009;18:245249.
Kawai T, Akira S. The role of pattern-recognition
receptors in innate immunity: update on Toll-like
receptors. Nat Immunol. 2010;11:373384.

46. Gottrup F. Oxygen in wound healing and infection.


World J Surg. 2004;28:312315.
47. Gardner SE, Frantz RA. Wound bioburden and
infection-related complications in diabetic foot ulcers.
Biol Res Nurs. 2008;10:4453.
48. Flemming HC, Wingender J. The biofilm matrix. Nat
Rev Microbiol. 2010;8:623633.
49. Lohela M, Bry M, Tammela T, et al. VEGFs
and receptors involved in angiogenesis versus
lymphangiogenesis. Curr Opin Cell Biol. 2009;21:
154165.
50. Peters KG, Kontos CD, Lin PC, et al. Functional
significance of Tie2 signaling in the adult vasculature.
Recent Prog Horm Res. 2004;59:5171.
51. Sottile J. Regulation of angiogenesis by extracellular
matrix. Biochim Biophys Acta. 2004;1654:1322.
52. Kubota Y, Kleinman HK, Martin GR, et al. Role of
laminin and basement membrane in the morphological
differentiation of human endothelial cells into
capillary-like structures. J Cell Biol. 1988;107:15891598.
53. Carmeliet P. Angiogenesis in life, disease and
medicine. Nature. 2005;438:932936.
54. Raza A, Franklin MJ, Dudek AZ. Pericytes and vessel
maturation during tumor angiogenesis and metastasis.
Am J Hematol. 2010;85:593598.
55. Paquet-Fifield S, Schluter H, Li A, et al. A role for
pericytes as microenvironmental regulators of human
skin tissue regeneration. J Clin Invest. 2009;119:
27952806.
56. Regan MC, Kirk SJ, Wasserkrug HL, et al. The wound
environment as a regulator of fibroblast phenotype.
J Surg Res. 1991;50:442448.
57. Baur Jr. PS, Parks DH, Hudson JD. Epithelial mediated
wound contraction in experimental wounds the
purse-string effect. J Trauma. 1984;24:713720.
58. Sen CK, Roy S. Oxygenation state as a driver of
myofibroblast differentiation and wound contraction:
hypoxia impairs wound closure. J Invest Dermatol.
2010;130:27012703.
59. Follonier L, Schaub S, Meister JJ, et al. Myofibroblast
communication is controlled by intercellular
mechanical coupling. J Cell Sci. 2008;121:33053316.
60. Brauchle M, Angermeyer K, Hubner G, et al. Large
induction of keratinocyte growth factor expression by
serum growth factors and pro-inflammatory cytokines
in cultured fibroblasts. Oncogene. 1994;9:31993204.
61. Takehara K. Growth regulation of skin fibroblasts.
J Dermatol Sci. 2000;24(Suppl 1):S70S77.
62. Sivamani RK, Garcia MS, Isseroff RR. Wound
re-epithelialization: modulating keratinocyte migration
in wound healing. Front Biosci. 2007;12:28492868.
63. Field CK, Kerstein MD. Overview of wound-healing in
a moist environment. Am J Surg. 1994;167:S2S6.

References

64. Svensjo T, Pomahac B, Yao F, et al. Accelerated healing


of full-thickness skin wounds in a wet environment.
Plast Reconstr Surg. 2000;106:602612.
65. Pilcher BK, Dumin JA, Sudbeck BD, et al. The activity
of collagenase-1 is required for keratinocyte migration
on a type I collagen matrix. J Cell Biol. 1997;137:
14451457.
66. Werner S, Krieg T, Smola H. Keratinocyte-fibroblast
interactions in wound healing. J Invest Dermatol.
2007;127:9981008.
67. Alizadeh N, Pepper MS, Modarressi A, et al. Persistent
ischemia impairs myofibroblast development in
wound granulation tissue: a new model of delayed
wound healing. Wound Repair Regen. 2007;15:809816.
68. Gillespie DL, Kistner B, Glass C, et al. Venous ulcer
diagnosis, treatment, and prevention of recurrences.
J Vasc Surg. 2010;52:8S14S.
69. Eklof B, Rutherford RB, Bergan JJ, et al. Revision of the
CEAP classification for chronic venous disorders:
consensus statement. J Vasc Surg. 2004;40:12481252.
70. Hess CT. Arterial ulcer checklist. Adv Skin Wound Care.
2010;23:432.
71. Stillman RM. Diabetic ulcers. In: eMedicine: WebMD,
2010. Available online at: http://emedicine.
medscape.com/article/460282-overview.
72. Simms M. Surgical treatment of the neuroischemic
foot. J Cardiovasc Surg (Torino). 2009;50:293311.
73. Karvestedt L, Martensson E, Grill V, et al. Peripheral
sensory neuropathy associates with micro- or
macroangiopathy: results from a population-based
study of type 2 diabetic patients in Sweden. Diabetes
Care. 2009;32:317322.
74. Thordarson DB, Shean CJ. Nerve and tendon
lacerations about the foot and ankle. J Am Acad Orthop
Surg. 2005;13:186196.
75. Kottner J, Balzer K, Dassen T, et al. Pressure ulcers: a
critical review of definitions and classifications. Ostomy
Wound Manage. 2009;55:2229.
76. Soppi E. [Pressure ulcer occurrence, pathophysiology
and prevention.] Duodecim. 2010;126:261268.
77. Bluestein D, Javaheri A. Pressure ulcers: prevention,
evaluation, and management. Am Fam Physician.
2008;78:11861194.
78. Olesen CG, de Zee M, Rasmussen J. Missing links in
pressure ulcer research an interdisciplinary overview.
J Appl Physiol. 2010;108:14581464.
79. Sen CK. Wound healing essentials: let there be oxygen.
Wound Repair Regen. 2009;17:118.
80. Sen CK, Roy S. Redox signals in wound healing.
Biochim Biophys Acta. 2008;1780:13481361.
81. Distler O, Distler JH, Scheid A, et al. Uncontrolled
expression of vascular endothelial growth factor and
its receptors leads to insufficient skin angiogenesis in

82.

83.
84.

85.

86.

87.

88.
89.

90.

91.

92.
93.
94.
95.

96.

97.

266.e3

patients with systemic sclerosis. Circ Res. 2004;95:


109116.
Berthod F, Germain L, Tremblay N, et al. Extracellular
matrix deposition by fibroblasts is necessary to
promote capillary-like tube formation in vitro. J Cell
Physiol. 2006;207:491498.
Hopf HW, Gibson JJ, Angeles AP, et al. Hyperoxia and
angiogenesis. Wound Repair Regen. 2005;13:558564.
Hunt TK, Aslam RS, Beckert S, et al. Aerobically
derived lactate stimulates revascularization and tissue
repair via redox mechanisms. Antioxid Redox Signal.
2007;9:11151124.
Myllyla R, Tuderman L, Kivirikko K. Mechanism of the
prolyl hydroxlase reaction. 2. Kinetic analysis of the
reaction sequence. Eur J Biochem. 1977;80:349357.
Roy S, Khanna S, Nallu K, et al. Dermal wound
healing is subject to redox control. Mol Ther. 2006;13:
211220.
H2O2 has been shown to support wound healing by inducing
VEGF expression in human keratinocytes. This work
presents the first in vivo evidence indicating that strategies
to influence the redox environment of the wound site may
have a bearing on healing outcomes.
Linnane AW, Kios M, Vitetta L. The essential
requirement for superoxide radical and nitric oxide
formation for normal physiological function and
healthy aging. Mitochondrion. 2007;7:15.
Sen CK, Packer L. Antioxidant and redox regulation of
gene transcription. Faseb J. 1996;10:709720.
Cheng Y, Song C. Hydrogen peroxide homeostasis
and signaling in plant cells. Sci China C Life Sci.
2006;49:111.
Gechev TS, Hille J. Hydrogen peroxide as a signal
controlling plant programmed cell death. J Cell Biol.
2005;168:1720.
Jones RD, Morice AH. Hydrogen peroxide an
intracellular signal in the pulmonary circulation:
involvement in hypoxic pulmonary vasoconstriction.
Pharmacol Ther. 2000;88:153161.
Neill S, Desikan R, Hancock J. Hydrogen peroxide
signalling. Curr Opin Plant Biol. 2002;5:388395.
Reth M. Hydrogen peroxide as second messenger in
lymphocyte activation. Nat Immunol. 2002;3:11291134.
Rhee SG. Redox signaling: hydrogen peroxide as
intracellular messenger. Exp Mol Med. 1999;31:5359.
Rhee SG, Bae YS, Lee SR, et al. Hydrogen peroxide: a
key messenger that modulates protein phosphorylation
through cysteine oxidation. Sci STKE. 2000;2000:PE1.
Sen CK. Antioxidant and redox regulation of cellular
signaling: introduction. Med Sci Sports Exerc.
2001;33:368370.
Sen CK. Cellular thiols and redox-regulated signal
transduction. Curr Top Cell Regul. 2000;36:130.

266.e4

14 Wound healing

98. Sen CK. The general case for redox control of wound
repair. Wound Repair Regen. 2003;11:431438.
At very low concentrations, reactive oxygen species may
regulate cellular signaling pathways by redox-dependent
mechanisms. Redox-based strategies may serve as effective
adjuncts to jump-start healing of chronic wounds. The
review focuses on the understanding of wound site
redox biology and novel insights into the fundamental
mechanisms that would help to optimize conditions for
oxygen therapy.
99. Stone JR, Yang S. Hydrogen peroxide: a signaling
messenger. Antioxid Redox Signal. 2006;8:243270.
100. Schaffer MR, Tantry U, Gross SS, et al. Nitric oxide
regulates wound healing. J Surg Res. 1996;63:237240.
101. Lee RH, Efron D, Tantry U, et al. Nitric oxide in the
healing wound: a time-course study. J Surg Res.
2001;101:104108.
102. Schaffer MR, Efron PA, Thornton FJ, et al. Nitric oxide,
an autocrine regulator of wound fibroblast synthetic
function. J Immunol. 1997;158:23752381.
103. Amadeu TP, Costa AM. Nitric oxide synthesis
inhibition alters rat cutaneous wound healing. J Cutan
Pathol. 2006;33:465473.
104. Schaffer MR, Tantry U, Efron PA, et al. Diabetesimpaired healing and reduced wound nitric oxide
synthesis: a possible pathophysiologic correlation.
Surgery. 1997;121:513519.
105. Pasquinelli AE. Molecular biology. Paring miRNAs
through pairing. Science. 2010;328:14941495.
106. Banerjee J, Chan YC, Sen CK. MicroRNAs in skin and
wound healing. Physiol Genomics. 2010.
MicroRNAs (MiRNAs) are small endogenous RNA
molecules about 22 nucleotides in length that are capable of
posttranscriptional gene regulation by binding to their
target messenger RNAs (mRNAs). This review focuses on
the role of miRNAs in cutaneous biology, the various
methods of microRNA modulation and the therapeutic
opportunities in treatment of skin diseases and wound
healing.
107. Roy S, Sen CK. miRNA in innate immune responses:
novel players in wound inflammation. Physiol
Genomics. 2011;43:557565.
108. Recchiuti A, Krishnamoorthy S, Fredman G, et al.
MicroRNAs in resolution of acute inflammation:
identification of novel resolvin D1-miRNA circuits.
FASEB J. 2011;25:544560.
109. Ponomarev ED, Veremeyko T, Barteneva N, et al.
MicroRNA-124 promotes microglia quiescence and
suppresses EAE by deactivating macrophages via the
C/EBP-alpha-PU.1 pathway. Nat Med. 2011;17:6470.
110. Asirvatham AJ, Magner WJ, Tomasi TB. miRNA
regulation of cytokine genes. Cytokine. 2009;45:5869.
111. Hata A, Davis BN. Control of microRNA biogenesis by
TGFbeta signaling pathway-A novel role of Smads in

112.

113.

114.

115.
116.

117.

118.

119.

120.

121.

122.

123.

124.

125.

126.

the nucleus. Cytokine Growth Factor Rev. 2009;20:


517521.
Sen CK, Gordillo GM, Khanna S, et al. Micromanaging
vascular biology: tiny microRNAs play big band. J Vasc
Res. 2009;46:527540.
Shilo S, Roy S, Khanna S, et al. Evidence for the
involvement of miRNA in redox regulated angiogenic
response of human microvascular endothelial cells.
Arterioscler Thromb Vasc Biol. 2008;28:471477.
Chan YC, Khanna S, Roy S, et al. miR-200b targets
Ets-1 and is down-regulated by hypoxia to induce
angiogenic response of endothelial cells. J Biol Chem.
2011;286:20472056.
Wang S, Olson EN. AngiomiRs key regulators of
angiogenesis. Curr Opin Genet Dev. 2009;19:205211.
Chan SY, Zhang YY, Hemann C, et al. MicroRNA-210
controls mitochondrial metabolism during hypoxia by
repressing the iron-sulfur cluster assembly proteins
ISCU1/2. Cell Metab. 2009;10:273284.
Loscalzo J. The cellular response to hypoxia: tuning
the system with microRNAs. J Clin Invest. 2010;120:
38153817.
Favaro E, Ramachandran A, McCormick R, et al.
MicroRNA-210 regulates mitochondrial free radical
response to hypoxia and Krebs cycle in cancer cells by
targeting iron sulfur cluster protein ISCU. PLoS One.
2010;5:e10345.
Tsuchiya S, Fujiwara T, Sato F, et al. MicroRNA-210
regulates cancer cell proliferation through targeting
fibroblast growth factor receptor-like 1 (FGFRL1). J Biol
Chem. 2011;286:420428.
Crosby ME, Kulshreshtha R, Ivan M, et al. MicroRNA
regulation of DNA repair gene expression in hypoxic
stress. Cancer Res. 2009;69:12211229.
Biswas S, Roy S, Banerjee J, et al. Hypoxia inducible
microRNA 210 attenuates keratinocyte proliferation
and impairs closure in a murine model of ischemic
wounds. Proc Natl Acad Sci USA. 2010;107:69766981.
Tiscornia G, Izpisua Belmonte JC. MicroRNAs in
embryonic stem cell function and fate. Genes Dev.
2010;24:27322741.
Li Z, Yang CS, Nakashima K, et al. Small RNAmediated regulation of iPS cell generation. EMBO J.
2011;30:828834.
Liu C, Teng ZQ, Santistevan NJ, et al. Epigenetic
regulation of miR-184 by MBD1 governs neural stem
cell proliferation and differentiation. Cell Stem Cell.
2010;6:433444.
Jopling C, Boue S, Izpisua Belmonte JC.
Dedifferentiation, transdifferentiation and
reprogramming: three routes to regeneration. Nat Rev
Mol Cell Biol. 2011;12:7989.
Wu Y, Zhao RC, Tredget EE. Concise review: bone
marrow-derived stem/progenitor cells in cutaneous

References

127.

128.

129.

130.

131.

132.

133.

134.

135.

136.

137.

138.

repair and regeneration. Stem Cells. 2010;28:


905915.
Kumamoto T, Shalhevet D, Matsue H, et al. Hair
follicles serve as local reservoirs of skin mast cell
precursors. Blood. 2003;102:16541660.
Cotsarelis G, Sun TT, Lavker RM. Label-retaining cells
reside in the bulge area of pilosebaceous unit:
implications for follicular stem cells, hair cycle, and
skin carcinogenesis. Cell. 1990;61:13291337.
Ito M, Liu Y, Yang Z, et al. Stem cells in the hair
follicle bulge contribute to wound repair but not to
homeostasis of the epidermis. Nat Med. 2005;11:
13511354.
The discovery of long-lived epithelial stem cells in the bulge
region of the hair follicle led to the hypothesis that epidermal
renewal and epidermal repair after wounding both depend
on these cells. This paper discusses the implications of
epithelial stem cells for both gene therapy and developing
treatments for wounds.
Kopp HG, Ramos CA, Rafii S. Contribution of
endothelial progenitors and proangiogenic
hematopoietic cells to vascularization of tumor and
ischemic tissue. Curr Opin Hematol. 2006;13:175181.
Morris LM, Klanke CA, Lang SA, et al.
Characterization of endothelial progenitor cells
mobilization following cutaneous wounding. Wound
Repair Regen. 2010;18:383390.
Fathke C, Wilson L, Hutter J, et al. Contribution of
bone marrow-derived cells to skin: collagen deposition
and wound repair. Stem Cells. 2004;22:812822.
Ishii GH, Sangai T, Sugiyama K, et al. In vivo
characterization of bone marrow-derived fibroblasts
recruited into fibrotic lesions. Stem Cells. 2005;23:
699706.
Deng WM, Han Q, Liao LM, et al. Engrafted bone
marrow-derived Flk-1(+) mesenchymal stem cells
regenerate skin tissue. Tissue Engineering. 2005;11:
110119.
Mori L, Bellini A, Stacey MA, et al. Fibrocytes
contribute to the myofibroblast population in wounded
skin and originate from the bone marrow. Exp Cell Res.
2005;304:8190.
Badiavas EV, Falanga V. Treatment of chronic wounds
with bone marrow-derived cells. Arch Dermatol.
2003;139:510516.
Rasulov MF, Vasilchenkov AV, Onishchenko NA,
et al. First experience in the use of bone marrow
mesenchymal stem cells for the treatment of a
patient with deep skin burns. Bull Exp Biol Med.
2005;139:141144.
Zuk PA, Zhu M, Ashjian P, et al. Human adipose tissue
is a source of multipotent stem cells. Mol Biol Cell.
2002;13:42794295.

266.e5

139. Ebrahimian TG, Pouzoulet F, Squiban C, et al. Cell


therapy based on adipose tissue-derived stromal cells
promotes physiological and pathological wound
healing. Arterioscler Thromb Vasc Biol. 2009;29:
503510.
140. Kim WS, Park BS, Sung JH, et al. Wound healing
effect of adipose-derived stem cells: a critical role
of secretory factors on human dermal fibroblasts.
J Dermatol Sci. 2007;48:1524.
141. Jaks V, Kasper M, Toftgard R. The hair follicle a stem
cell zoo. Exp Cell Res. 2010;316:14221428.
142. Myung P, Andl T, Ito M. Defining the hair follicle stem
cell (Part I). J Cutan Pathol. 2009;36:10311034.
143. Myung P, Andl T, Ito M. Defining the hair follicle stem
cell (Part II). J Cutan Pathol. 2009;36:11341137.
144. Kavanagh DP, Kalia N. Hematopoietic stem cell
homing to injured tissues. Stem Cell Rev. 2011;7:
672682.
145. Yamanaka S. Induction of pluripotent stem cells from
mouse fibroblasts by four transcription factors. Cell
Prolif. 2008;41(Suppl 1):5156.
146. Yoshida Y, Yamanaka S. iPS cells: a source of cardiac
regeneration. J Mol Cell Cardiol. 2011;50:327332.
147. Vercelli S, Ferriero G, Sartorio F, et al. How to assess
postsurgical scars: a review of outcome measures.
Disabil Rehabil. 2009;31:20552063.
148. Larson BJ, Longaker MT, Lorenz HP. Scarless fetal
wound healing: a basic science review Plast Reconstr
Surg. 2010;126:11721180.
149. Gurtner GC, Werner S, Barrandon Y, et al. Wound
repair and regeneration. Nature. 2008;453:314321.
150. Singer AJ, Clark RA. Cutaneous wound healing.
N Engl J Med. 1999;341:738746.
151. Lovvorn 3rd. HN, Cheung DT, Nimni ME,
et al. Relative distribution and crosslinking
of collagen distinguish fetal from adult sheep
wound repair. J Pediatr Surg. 1999;34:
218223.
152. Ferreira LM, Gragnani A, Furtado F, et al. Control
of the skin scarring response. An Acad Bras Cienc.
2009;81:623629.
153. Scott JR, Muangman P, Gibran NS. Making sense of
hypertrophic scar: a role for nerves. Wound Repair
Regen. 2007;15(Suppl 1):S27S31.
154. Shih B, Bayat A. Genetics of keloid scarring. Arch
Dermatol Res. 2010;302:319339.
155. Aarabi S, Longaker MT, Gurtner GC. Hypertrophic
scar formation following burns and trauma: new
approaches to treatment. PLoS Med. 2007;4:e234.
156. Stelnicki EJ, Chin GS, Gittes GK, et al. Fetal wound
repair: where do we go from here? Semin Pediatr Surg.
1999;8:124130.

Вам также может понравиться