Sei sulla pagina 1di 32

14  

Wound healing
Chandan K. Sen and Sashwati Roy

 Wound vascularization may be achieved by angiogenesis or


SYNOPSIS
vasculogenesis
 There are three general techniques of wound treatment: primary  A wound is generally considered chronic if it has not healed in 4

intention, secondary intention, and tertiary intention weeks. Chronic wounds can be broadly classified into three major
 The following overlapping phases drive the overall healing categories: venous and arterial ulcers, diabetic ulcers, and
response: hemostasis, inflammation, proliferative, and   pressure ulcers
remodeling  Vascular complications commonly associated with problematic

 Successful hemostasis or blood coagulation results in prevention wounds are primarily responsible for wound ischemia. Limitations
of blood loss by plugging the wound within seconds through in the ability of the vasculature to deliver O2-rich blood to the
vasoconstriction and formation of a hemostatic blood clot wound tissue leads to, among other consequences, hypoxia. Three
consisting of platelets and fibrin. The process is divided into major factors may contribute to wound tissue hypoxia: (1)
initiation and amplification. Initiation is caused by an extrinsic peripheral vascular diseases garroting O2 supply; (2) increased O2
pathway, whereas amplification is executed by an intrinsic   demand of the healing tissue; and (3) generation of reactive
pathway oxygen species (ROS) by way of respiratory burst and for redox
 The inflammatory response during normal healing is characterized signaling
by spatially and temporally changing patterns of specific leukocyte  Small RNAs are a new class of regulators of eukaryotic biology.

subsets. Dysregulated inflammation complicates wound healing. Alongside other small interfering RNAs (siRNAs), miRNAs execute
Complete resolution of an acute inflammatory response is the ideal posttranscriptional gene silencing through mRNA destabilization as
outcome following an insult well as translational repression. miRNAs are emerging as a key
 Infection is a common problem in chronic wounds, frequently regulatory of the overall wound-healing process
resulting in nonhealing wounds and significant patient morbidity  The regenerative potential of injured adult tissue suggests the

and mortality. Microorganisms do not always live as pure cultures physiological existence of cells capable of participating in the
of dispersed single cells but instead accumulate at interfaces to reparative process. Bone marrow-derived mesenchymal stem cells
form polymicrobial aggregates such as films, mats, flocs, sludge, (BM-MSCs) have been shown to promote the healing of diabetic
or biofilms wounds, implying a profound therapeutic potential for skin defects
 In an open wound that has undergone contraction, restoration of such as chronic wounds and burns
an intact epidermal barrier is enabled through wound  Scars (also called cicatrices) are macroscopic fibrous tissue that

epithelialization, also known as re-epithelialization. During the visibly replaces normal skin after injury. There is a wide spectrum
proliferative phase of wound healing, the granulation tissue is light of skin scarring postwounding, including scarless fetal wound
red or dark pink in color because of perfusion by new capillary healing, fine-line or normal scars, stretched scars, atrophic
loops. It is soft to the touch, moist, and granular in appearance. (depressed) scars, scar contractures, hypertrophic scars, and
The granulation tissue serves as a bed for tissue repair keloids.

©
2013, Elsevier Inc. All rights reserved.
Acute wounds 241

Laceration Puncture wound


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 poul-
tices 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 drain- Fig. 14.1  Laceration and puncture wounds.
ing pus from the wound, and Galen (c. 130–200 AD)
described the principle of first- and second-intention
healing.3 Wound healing advanced slowly over the cen- contusion where the skin appears to be intact but suffers
turies, with major advances in the 19th century in the from damage caused to underlying tissues.
importance of controlling infection, hemostasis, and Open wounds may be generally categorized as:
necrotic tissue.4 Today, surgical trauma, taken together
• Lacerations – ragged tears and cuts; masses of torn
with injury caused during accidents and secondary to
tissue underneath; caused by dull knife, bomb
other clinical conditions, e.g., diabetes, represents a sub-
fragments and machinery and may include crushing
stantial cost to society.5 Any solution to wound-healing
of tissues; frequently contaminated (Fig. 14.1)
problems will require a multifaceted comprehensive
approach. First and foremost, the wound environment • Puncture – e.g., sharp penetrations caused by nails,
will have to be made receptive to therapies. Second, the needles, wire, or bullets (Fig. 14.1). These are of
appropriate therapeutic regimen needs to be identified great concern in patients with diabetes, as many of
and provided while managing systemic limitations that these patients have polyneuropathy and have
could secondarily limit the healing response. This insensate feet, leading to occult injury. Many times
chapter aims to present an overall outline of the cutane- these patients will step on thumb tacks, safety pins,
ous wound-healing process. 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
Acute wounds removed; skinned knee or elbows and rope burns
are examples; an abrasion lends itself to infection
Any violation of live tissue integrity may be regarded
• Avulsions – sections of skin torn off either in part
as a wound. Skin is the largest organ of the human body,
(attached to body) or totally (detached from body);
covering about 3000 square inches (7620 cm2) in an
heavy bleeding is common
average adult. The most important role of the skin for
• Amputations – traumatic amputation results in
terrestrial animals is to protect the water-rich internal
nonsurgical removal of limb from the body and
organs from the dry external environment. As a primary
accompanies heavy bleeding.
line of defense against external threats, maintenance of
integrity of the skin is a key prerequisite for healthy There are three general techniques of wound
survival. Thus, healthy skin can regenerate and repair treatment:
itself under most common conditions. Skin wounds 1. primary intention, in which all tissues, including
may be open when manifested as a tear, cut, or punc- the skin, are closed with suture material after
ture. Blunt force trauma may cause closed wounds or completion of the operation
242 • 14 • Wound healing

2. secondary intention, in which the wound is left Scab Clot Neutrophils


open and closes naturally 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 A
as interventions under conditions where the wound is
subjected to therapy. For simplicity of understanding
New
the several interdigitated biological processes that drive Re-epithelialization capillary Granulation
the overall healing response, wound healing is com- Fibroblast tissue Macrophage
monly discussed as the following overlapping phases: 3-7 days

hemostasis and inflammation, proliferative (granula-


tion, vascularization, and wound closure; closure may
be discussed as wound contraction and epithelializa-
tion) and remodeling (can continue from weeks to years
and encompasses scarring, tensile strength, and turno-
ver of extracellular matrix (ECM) components). These
stages, taken as a whole, are also referred to as the
wound-healing cascade (Fig. 14.2).

Hemostasis B

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 Weeks Wound contraction
injury that results in exposure of blood components to
the subendothelial layers of the vessel wall. Through
successful hemostasis blood loss is prevented by plug-
ging the wound within seconds through vasoconstric-
tion and formation of a hemostatic blood clot consisting
of platelets and fibrin. Hemostasis requires both plate-
lets 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 intrin-
sic pathway. The intrinsic pathway consists of plasma
factor XI (FXI), IX, and VIII (Fig. 14.3). Tissue factor
C
(TF) generates a “thrombin burst,” a process by which
Fig. 14.2  (A–C) Phases of cutaneous wound healing: a simplified representation.
Hemostasis 243

Tissue damage
Vessel injury

Healing

Collagen

FXII Kallikrein Prekallikrein

FXIIa WBC FXIII Platelet Polymerization


TF RBC and
stabilization
HK
FXIIIa
Intrinsic
pathway FVII FVIIa

FXI FXIa
Ca2+

Phospholipids
Protein-C Fibrinogen
Platelet Extrinsic
Phospholipids Ca2+ Protein-S TM
pathway

TFPI
Antithombin
FIX FIXa PF3 FV III

Thrombin
FX FVa
FVIII FVIIIa
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 Under physiological conditions, TF is constitutively


driver of the overall coagulation cascade. expressed by adventitial cells surrounding blood vessels
The extrinsic pathway responsible for the initiation of and initiates clotting. Examples of such adventitial
blood coagulation consists of the transmembrane recep- cells include vascular smooth-muscle cells, pericytes,
tor TF and plasma FVII/VIIa. On the other hand, the and adventitial fibroblasts.6,7 TF may also contribute to
intrinsic pathway consists of plasma FXI, FIX, and FVIII. amplification of blood coagulation through its so-called
244 • 14 • Wound healing

blood-borne form, which is present as cell-derived The formation of blood clot is initiated by the proteo-
microparticles as well as through TF expressed within lytic cleavage of fibrinogen by thrombin. As a result,
platelets.6–8 fibrin is produced and forms cross-links with each other.
Levels of FVIIa, a key player of the extrinsic pathway, Cross-linked fibrin entraps platelets, and together they
in the circulating blood are higher than any other acti- adhere to the subendothelium through adhesion mole-
vated coagulation factor. Following injury to the blood cules called integrins. Clot fibrin plays a key role in
vessel wall, FVII comes into contact with TF expressed mounting the inflammatory process as well as in facili-
on TF-bearing cells (e.g., white blood cells) and forms tating wound angiogenesis and stromal cell prolifera-
an activated complex (TF–FVIIa). TF–FVIIa activates tion. Fibrin binds to integrin CD11b/CD18 on infiltrating
FIX and FX, resulting in FIXa and FXa, respectively. FVII monocytes and neutrophils. It also binds to fibroblast
is activated by thrombin, FXIa, FXII, and FXa. The acti- growth factor-2 (FGF-2) and vascular endothelial growth
vation of FXa by TF–FVIIa is almost immediately inhib- factor (VEGF) that help the wound tissue vascularize.
ited by the TF pathway inhibitor. FXa and its cofactor Fibrin also binds to insulin-like growth factor-I and pro-
FVa form the prothrombinase complex, which activates motes stromal cell proliferation.11,12
prothrombin to thrombin. Thrombin is a serine protease Blood clot represents the seat of wound chemotaxis.
that plays a central role in hemostasis after tissue injury Thrombin, released by platelets at the wound site, is an
by converting soluble plasma fibrinogen into an insolu- early mediator of clot development.13 Thrombin is a
ble fibrin clot and by promoting platelet aggregation. serine protease that converts soluble plasma fibrinogen
Thrombin activates other components of the coagula- into an insoluble fibrin clot. In addition, it promotes
tion cascade, including FV and FVIII, which in turn platelet aggregation. Thrombin function may be viewed
activates FXI and cascades to the activation of FIX. as an interface between the hemostasis phase of wound
Thrombin also activates and releases FVIII from being healing and the ensuing inflammatory phase as it plays
bound to von Willebrand factor. FVIIIa is the cofactor of a potent role in mounting wound inflammation. The
FIXa, and together they form the “tenase” complex, proinflammatory effects of thrombin include stimula-
which activates FX. In this way the cycle continues. tion of vasodilation responsible for plasma extravasa-
The intrinsic pathway begins with formation of the tion, edema, and an increased expression of endothelial
primary complex on collagen by high-molecular-weight cell adhesion molecules that helps monocytes and others
kininogen, prekallikrein, and FXII (Hageman factor). cells extravasate and infiltrate the wound site. Thrombin
Prekallikrein is converted to kallikrein, and FXII becomes also induces the release of proinflammatory cytokines
FXIIa. FXIIa converts FXI into FXIa. FXIa activates like CCL2, interleukin-6 (IL-6), and IL-8 by endothelial
FIX, which together with its cofactor FVIIIa forms the cells. These cytokines induce monocyte chemotaxis.14
tenase complex. The tenase complex activates FX to FXa Furthermore, thrombin induces the release of inflamma-
(Fig. 14.3). tory cytokines by monocytes, including IL-6, interferon-γ,
The blood clot physically helps plug the wound, IL-1β, and tumor necrosis factor-α (TNF-α). These early-
minimizing blood loss. It is primarily made up of phase cytokines are typically proinflammatory, which
cross-linked fibrin, cells such as erythrocytes and plate- may govern the differentiation of blood-derived mono-
lets, as well as other ECM proteins such as fibronectin, cytes into M1 wound macrophages.15 Wound chemo-
vitronectin, and thrombospondin. Current understand- taxis is also driven by the degradation of fibrin and
ing portrays the clot as a dynamic structural matrix subsequent activation of the complement system. As
containing functionally active proteins and cells. In part of this process several chemotactic agents and
addition to containment of blood loss, the clot serves as cytokines are released, which in turn launch the inflam-
a first aid against microbial invasion. The clot also serves matory phase of wound healing through chemotactic
as a provisional matrix for the homing of blood-borne recruitment of blood-borne immune cells.16 Platelets are
cells, including inflammatory as well as stem or pro- one of the earliest sources of cytokines which execute
genitor cells. The provisional matrix is enriched in immune cell chemotaxis as well as macrophage activa-
cytokines and growth factors which then regulate the tion. Once trapped in the fibrin net, platelets release
function of the homing cells.9,10 granules that function as a reservoir for biologically
Inflammation 245

active proteins, such as RANTES (regulated on activa-


tion, normal T cell expressed, and secreted or CCL5),
thrombin, transforming growth factor-β (TGF-β),
platelet-derived growth factor (PDGF), and VEGF.
CCL5 is one of the most potent monocyte chemoattract-
ants 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-1α (CCL3), TGF-α, fibronectin,
Cytokines
elastin, C5a, C3a, nerve growth factor, and ECM signaling
components.17,18 path to
Initiate injury site
tissue Neutrophil
repair 6

Inflammation 3 Platelets

Tissue injury triggers an acute-phase inflammation 4


(Latin, inflammare, to set on fire) response that is meant 5 Mast cell 2

to prepare the wound site for subsequent wound closure


Macrophage
(Fig. 14.4). Inflammation encompasses a series of
responses of vascularized tissues of the body to injury. 1
Local chemical mediators that are biosynthesized during 1 Bacteria and other pathogens enter wound
acute inflammation give rise to the macroscopic events
2 Platelets from blood release blood-clotting proteins at wound site.
characterized by Celsus in the first century, namely,
3 Mast cells secrete factors that mediate vasodilation and vascular constriction.
rubor (redness), tumor (swelling), calor (heat), and dolor Delivery of blood, plasma and cells to injured area
(pain). At cellular and molecular levels, inflammation 4 Neutrophils and macrophages remove pathogens by phagocytosis
results from the coordination of manifold systems of
5 Macrophages secrete hormones called cytokines that attract immune system cells
receptors and sensors that affect transcriptional and to the site and activate cells involved in tissue repair
posttranslational programs necessary for host defense 6 Inflammatory response continues until the foreign material is eliminated and the
and resolution of infection. During normal healing, the wound is repaired
inflammatory response is characterized by spatially and
Fig. 14.4  The wound inflammatory response.
temporally changing patterns of specific leukocyte
subsets.

Platelets leukocytes and diapedesis (Fig. 14.5). Adhesion mole-


cules such as integrins as well as P-selectin and E-selectin
Formation of the clot or thrombus is dependent on play a central role in enabling diapedesis of neutrophils
platelet activation. The platelet-rich blood clot also (Fig. 14.6). These adhesion molecules bind with integrins
entraps polymorphonuclear leukocytes (neutrophils). expressed on the cell surface of neutrophils, such as
This helps amplify blood coagulation and lays the foun- CD11a/CD18 (LFA), CD 11b/CD18 (MAC-1), CD11c/
dation for the subsequent acute-phase inflammatory CD18 (gp150, 95), and CD11d/CD18. Alongside
response. In a matter of hours after injury, large numbers cytokines, chemokines play a major role in mounting
of neutrophils extravasate by transmigrating across the acute-phase inflammation after injury. Chemokines
endothelial cell wall of blood capillaries to the wound include IL-8, MCP-1, and growth-related oncogene-α. In
site. To enable this, local blood vessels are activated the case of an infected wound, bacterial products such
by proinflammatory cytokines such as IL-1β, TNF-α, as lipopolysaccharide and formyl-methionyl peptides
and interferon-γ. These cytokines induce the expression can enhance neutrophil recruitment to the wound site
of adhesion molecules necessary for adhesion of (Fig. 14.7).
246 • 14 • Wound healing

Normal permeability of capillary


Small amount of fluid Shedding of
Rolling
L-selectin Adhesion Diapedesis

Neutrophil

Activating substances Lipopolysaccharides, C3a C5a,


Capillary wall Monocyte released by chemokines,
interleukin-1, and
bacteria tumor necrosis factor α histamine,
and prostaglandins
Increased permeability of capillary damaged and leukotrienes
during inflammation More fluid and tissues
antimicrobal chemicals
L-selectin Integrin
Sialyl-Lewis 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
Interstitial Monocyte squeezing wound site.
spaces through interstitial space

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 phagolyso-
somes, 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 invagina-
Fibrin-
tion of the cell membrane occurs with the incorporation platelet clot Collagen Elastin
of the microorganism into an intracellular phagosome.
Red blood Fibroblast Cytokines &
There follows a burst of oxygen consumption, and cell growth factors
much, if not all, of the extra oxygen consumed is con-
verted to highly reactive oxygen species. This is called Fig. 14.7  Diapedesis and migration of leukocytes to the wound site.
Inflammation 247

respiratory burst. In addition, the cytoplasmic granules


discharge their contents into the phagosome, and death Gram-negative
of the ingested microorganism soon follows. Among the bacteria
antimicrobial systems formed in the phagosome is one
consisting of myeloperoxidase (MPO), released into the
phagosome during the degranulation process, hydro-
gen peroxide (H2O2), formed by the respiratory burst
and a halide, particularly chloride. The initial product
of the MPO-H2O2-chloride system is hypochlorous acid, Toll-like Lipopolysaccharide
receptor
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 Cell membrane
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 Nucleus
elastase, cathepsin G, proteinase 3, and urokinase-type Cytoplasm
plasminogen activator. As it relates to the overall inflam-
matory process elicited in response to injury, neutrophils
are major players because they can modify macrophage
Fig. 14.8  Toll-like receptors: responding to infectious agents and the wound
function and therefore regulate innate immune response microenvironment.
during wound healing.20 In the absence of neutrophils,
wound site macrophages seem to lack guidance in con- Mediators present in the microenvironment that the
ducting the healing process.21 monocyte traverses to reach the wound site interact
In a healing wound, neutrophil infiltration ceases with receptors on the monocyte cell surface, bringing
after a few days of injury. Expended neutrophils are forth major changes in the transcriptomic as well as
programmed to die and dying neutrophils are recog- proteomic make of the cell. Major examples of such
nized by wound site macrophages and phagocytosed. receptors present on the monocyte surface include Toll-
The wound site contains a small portion of macrophages like receptors (TLRs: Fig. 14.8), complement receptors,
that are resident. Most macrophages at the wound and Fc receptors. At the wound site, macrophages
site are recruited from the peripheral circulation. function as antigen-presenting cells and phagocytes
Extravasation of peripheral blood monocytes is enabled scavenging dead cells and debris. In addition, they
by the interaction between endothelial vascular cell deliver a wide range of growth factors that are known
adhesion molecule-1 and monocyte very late antigen-4 for their abilities to execute the wound-healing process.
(α4β1 integrin). Factors that guide the extravasated Such growth factors include TGF-β, TGF-α, basic FGF
monocyte to the wound site include growth factors, (bFGF), VEGF, and PDGF. These growth factors enable
chemotactic proteins, proinflammatory cytokines, and wound healing by causing cell proliferation and synthe-
chemokines such as macrophage inflammatory protein sis of ECM and inducing angiogenesis. Macrophages
1α, MCP-1, and RANTES. The source of these chemoat- play a crucial role in enabling wound healing.
tractants includes clot-associated platelets, wound edge Macrophage depletion is known to impair wound
hyperproliferative keratinocytes, wound tissue fibrob- closure markedly.4,22
lasts and subsets of leukocyte already at the wound
site. Once the monocyte leaves the blood vessel to trans- Mast cells
migrate into the wound site through the ECM micro­
environment, the process of monocyte differentiation Mast cells are best known for their central role in medi-
to macrophages has started. ating allergic responses. Beyond that function, it is now
248 • 14 • Wound healing

known that mast cells are physiologically significant diminished the accumulation of macrophages in healing
in recognizing pathogens and in regulating immune skin wounds of adult guinea pigs. Such depletion
response.23 Mast cells may instantly release several resulted in impaired disposal of damaged tissue and
proinflammatory mediators from intracellular stores. In provisional matrix, compromised fibroblast count, and
addition, they are localized in the host–environment delayed healing. Today, macrophages have emerged to
interface. These properties make mast cells key players be a pivotal driver of efficient skin repair.34,35 Macrophages
in finetuning immune responses during infection. are plastic and heterogeneous cells broadly categorized
Recent studies using mast cell activators as effective into two groups: classically activated or type I macro-
vaccine adjuvants show the potential of harnessing phages, which are proinflammatory effectors, and alter-
these cells to confer protective immunity against micro- natively activated or type II macrophages.36 In the
bial pathogens.24 Mast cell activation helps initiate the inflamed tissue, it is unclear whether the type II macro-
inflammatory phase of wound healing. In response to phages that appear during the healing phase originate
injury, mast cells at the wound site degranulate within from newly attracted monocytes or from a switch in the
a matter of hours and therefore become histologically activation state of previously proinflammatory macro-
silent at the wound tissue. After about 48 hours of injury, phages. The macrophage population first taking part in
mast cells are again seen in the wound tissue and their inflammation may change its phenotype and assume
number increases as healing progresses.25 On one hand, the role to resolve inflammation.37,38 Macrophages from
impaired wound healing has been reported in mast cell- diabetic wounds display dysfunctional inflammatory
deficient mice.26 On the other hand, mast cells have been responses.39 A persistent inflammatory state of diabetic
implicated in skin wound fibrosis.27 With the aid of a wound macrophages is caused by impairment in the
wide array of newly formed or preformed mediators ability of these cells to phagocytose apoptotic cells at the
released by degranulation, the activated mast cell con- wound site which in turn prevents the switch from M1
trols the key events of the healing phases: triggering and to M2 phenotype.39
modulation of the inflammatory stage, proliferation of
connective cellular elements, and final remodeling of Resolution of inflammation
the newly formed connective tissue matrix. The impor-
tance of the mast cell in regulating healing processes is Inflammatory responses elicited by injury are only
also demonstrated by the fact that a surplus or deficit helpful to the healing process if they are timely and
of degranulated biological mediators causes impaired transient. Dysregulated inflammation complicates
repair, with the formation of exuberant granulation wound healing.39 Complete resolution of an acute
tissue (e.g., keloids and hypertrophic scars), delayed inflammatory response is the ideal outcome following
closure (dehiscence), and chronicity of the inflam­ an insult. For resolution to ensue, further leukocyte
matory stage.28 recruitment must be halted and accompanied by
removal of leukocytes from inflammatory sites. Reso­
Macrophages lution of inflammation is executed by a number of key
factors. At the wound site successful phagocytosis of
Macrophages represent the predominant cell type in a dead neutrophils by macrophages is a key factor.
healing wound 3–5 days following injury. The primary Impairment in macrophage function at the wound site
acute function of wound macrophages, which arrive at derails the resolution of inflammation.39 Lipid media-
an injury site hours later than neutrophils, is to operate tors, such as the lipoxins, resolvins, protectins, and
as voracious phagocytes cleansing the wound of all newly identified maresins, have emerged as a novel
matrix and cell debris, including fibrin and apoptotic genus of potent and stereoselective players that
neutrophils. Macrophages also produce a range of counterregulate excessive acute inflammation and
cytokines, growth and angiogenic factors that drive stimulate molecular and cellular events that define
fibroblast proliferation and angiogenesis.4,29–32 In a resolution.40 Successful resolution paves the path for
classic study, Leibovich and Ross33 demonstrated that the healing process to progress towards successful
antimacrophage serum combined with hydrocortisone wound closure. Prolonged inflammation may not only
Infection 249

compromise wound closure but may also worsen scar the mid-1990s showed that pathogen recognition by the
outcomes.41,42 innate immune system is instead actually specific,
relying on germline-encoded pattern recognition recep-
tors (PRRs) that have evolved to detect components of
Infection foreign pathogens, referred to as pathogen-associated
molecular patterns (PAMPs).45 TLRs regulate innate and
Infection is a common problem in chronic wounds, fre- adaptive immune responses and are important modula-
quently resulting in nonhealing and significant patient tors of inflammation during wound-healing responses.
morbidity and mortality.43 Wound infection and the sub- The finding that there is activation of TLR signaling
sequent release of proinflammatory modulators result during tissue damage in several disease situations in the
in pain and delayed healing. The pain, in turn, compro- absence of infection suggests that endogenous mole-
mises the immune response to infection.44 All wounds cules serve as TLR agonists, although it is unclear
become contaminated by bacteria from the surrounding whether this response is biologically important for
skin, the local environment, and autologous patient maintenance of homeostasis, such as tissue repair, or
sources. The local environment is particularly relevant whether this recognition is simply accidental. It is note-
for hospitalized patients. Colonization is defined as the worthy that microbial infection triggers the production
presence of proliferating bacteria without a noticeable of modified endogenous molecules (such as high-
host response. Colonization of the wound may enhance mobility group protein B1, oxidized phospholipids,
or impede wound healing, depending upon the bacte- β-defensin 2, and nucleic acids) that are recognized by
rial load. Bacterial loads in excess of 105 organisms/ TLRs or other cytosolic PRRs. This may suggest that
gram of tissue are a threat to wound healing, although these endogenous molecules, along with PAMPs, act as
this threshold may be altered by the status of the host adjuvants to activate innate immune programs via TLRs
immune system and the number and types of bacterial and/or other PRRs, and have key roles in facilitating
species present. The concept of critical colonization adaptive immunity against infecting microbes.
is controversial and not universally accepted. Critical Chronic wounds have a complex colonizing flora that
colonization is characterized by increased bacterial changes over time. Staphylococcus aureus and coagulase-
burden or covert infection, and the wound at this stage negative staphylococci are the most commonly isolated
may enter a nonhealing, chronic inflammatory state. organisms. Chronic wounds are colonized by multiple
Substantial colonization may not cause the obvious bacterial species and many persist in the wound once
signs of inflammation but will likely affect wound they are established. In chronic venous leg ulcers the
healing with failure to heal or slowing of progression. most common bacteria noted, in order of abundance,
Signs of critical colonization are atrophy or deteriora- were S. aureus, Enterococcus faecalis, Pseudomonas aerugi-
tion of granulation tissue, discoloration of granulation nosa, coagulase-negative staphylococci, Proteus spp.,
tissue to deep red or gray, increased wound friability, and anaerobic bacteria. Resident (colonizing) bacterial
and increased drainage. The transition to infection species are commonly present in ulcers. The longer an
occurs when bacterial proliferation overcomes the host’s ulcer remains unhealed, the more likely it will acquire
immune response and host injury occurs. Several factors multiple aerobic organisms and a significant anaerobic
determine transition from colonization to infection: the population. Chronic wounds are commonly compli-
bioburden itself, the virulence of the organisms, the syn- cated by underlying ischemia. Thus, they tend to have
ergistic action of different bacterial species, and the a low tissue oxygen level. This facilitates the growth of
ability of the host to mount an immune response.43 anaerobes in ischemic wounds. Adequate delivery of
During the past decade, there has been rapid progress oxygen to the wound tissue is vital for optimal healing
in the understanding of innate immune recognition and resistance to infection.46 Hospitalization, surgical
of microbial components and its critical role in host procedures, and prolonged or broad-spectrum anti­
defense against infection. The early concept of innate biotic therapy may predispose patients to coloniza-
immunity was that it nonspecifically recognized tion or infection, or both, with resistant organisms,
microbes; however, the discovery of TLRs (Fig. 14.8) in including S. aureus (methicillin-resistant S. – MRSA) or
250 • 14 • Wound healing

vancomycin-resistant enterococci.43 Because inflamma- drug and other physiologic changes that could impair
tory responses to microbial invasion may be diminished drug effectiveness.43
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 Vascularization
Biofilm Wounds larger than can be closed by diffusion of oxygen
from neighboring intact blood vessels or wounds com-
Microorganisms do not always live as pure cultures of plicated by underlying ischemia largely rely on wound
dispersed single cells but instead accumulate at inter- vascularization for their closure. Wound vascularization
faces to form polymicrobial aggregates such as films, may be achieved by angiogenesis or vasculogenesis.
mats, flocs, sludge, or biofilms. The biofilm state of Angiogenesis represents sprouting of capillaries from
microorganisms may lead to an increase in virulence existing blood vessels in the wound edge tissue.
and propensity to cause infection. In most biofilms, the Vasculogenesis relies on the formation of new blood
microorganisms account for less than 10% of the dry vessels by mobilization of bone marrow-derived
mass, whereas the matrix can account for over 90%. The endothelial stem cells.
matrix is the extracellular material, mostly produced by Wound vascularization is regulated by all phases in
the organisms themselves, in which the biofilm cells are wound healing – hemostasis, inflammation, tissue for-
embedded. It consists of a conglomeration of different mation, as well as tissue remodeling. The hemostatic
types of biopolymers – known as extracellular poly- plug provides a bed for blood-borne cells to home. Once
meric substances (EPS) – that forms the scaffold for the cells entangle in this plug, the ECM environment modi-
three-dimensional architecture of the biofilm and is fies cell function towards successful healing. Platelets in
responsible for adhesion to surfaces and for cohesion in the clot serve as a source of growth factors and cytokines
the biofilm. The formation of a biofilm allows a lifestyle which recruit several cell types, including endothelial
that is entirely different from the planktonic state. cells, to the wound site. During the inflammatory phase,
Although the precise and molecular interactions of the leukocytes at the wound site serve as a major source of
various secreted biofilm matrix polymers have not been proangiogenic factors such as VEGF-A and IL-8 that lay
defined, and the contributions of these components to the early foundation for successful wound tissue vascu-
matrix integrity are poorly understood at a molecular larization. As neutrophils are expended and undergo
level, several functions of EPS have been determined, cell death, the number of macrophages at the wound
demonstrating a wide range of advantages for the site substantially increases. Macrophage-derived TGF-
biofilm mode of life. β, TGF-α, bFGF, PDGF, and VEGF play a key role in
The architecture of biofilms is influenced by many driving skin wound angiogenesis. Growing evidence
factors, including hydrodynamic conditions, concentra- demonstrates that no single angiogenic factor is singu-
tion of nutrients, bacterial motility, and intercellular larly effective in significantly influencing wound out-
communication, as well as exopolysaccharides and pro- comes. Wound vascularization is a sophisticated process
teins.48 Chronic wounds offer attractive conditions for requiring dynamic, temporally and spatially regulated
biofilm production because proteins (collagen, fibronec- interaction between cells, angiogenic factors, and the
tin) and damaged tissues are present, which allow ECM.
attachment. The biofilm impedes healing of chronic Key processes in tissue vascularization are depicted
wounds. Most of the chronic wound pathogens, such as in Figure 14.9. Angiogenic cues are elicited by micro­
MRSA and Pseudomonas spp., are typical biofilm pro- environmental signals such as hypoxia and are ampli-
ducers. Compared to bacteria in the unattached free- fied by angiogenic factors such as VEGF expressed by
living planktonic form, bacteria that reside within and released from cells at the wound site. VEGF was
mature biofilms are highly resistant to traditional anti- originally identified as an endothelial cell-specific
biotic therapies. Bacteria in biofilms grow more slowly, growth factor-stimulating angiogenesis and vascular
and slower growth may lead to decreased uptake of the permeability. Some family members, VEGF C and D, are
Vascularization 251

1 9
Angiogenic Loop
factor (VEGF) formation
production

2 7 Vascular
Release stabilization
ECM
remodeling 10
Angiogenic
factors bind 6 Pericyte
to endothelial Tie-2
Directional
cell receptors 5 8
migration
3 Endothelial cell
4 Tube
proliferation
Endothelial cell αyβ3 formation
activation

Fig. 14.9  Neoangiogenesis: the formation of new


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

specifically involved in lymphangiogenesis. Ligation of process in which cells move in a given direction either
these angiogenic factors with their corresponding recep- in response to changes in the extracellular environment
tors elicits a multitude of cell-signaling processes that or as a consequence of an intrinsic propensity for direc-
activate microvascular endothelial cells. For example, tional movement. ECM remodeling by proteases pro-
VEGF and their endothelial tyrosine kinase receptors motes cell migration, a critical event in the formation of
are central regulators of tissue vascularization. VEGF new vessels. Temporal and spatial regulation of ECM
signaling through VEGFR-2 is the major angiogenic remodeling events allows for local changes in net matrix
pathway. VEGFR-3 has also been shown to be important deposition or degradation, which in turn contributes to
for angiogenesis, acting together with VEGF/VEGFR-2 control of cell growth, migration, and differentiation
and Dll4/Notch signaling to control angiogenic sprout- during different stages of angiogenesis. Matrix-bound
ing.49 The biological significance of other angiogenic growth factors released by proteases and/or by ang-
factors such as EGF and bFGF is mediated by their cor- iogenic factors promote angiogenesis by enhancing
responding receptors, which also belong to the family endothelial migration and growth. Matrix molecules
of tyrosine kinase receptors EGFR, FGFR-1, FGFR-2, promote endothelial cell growth and morphogenesis,
FGFR-3, and FGFR-4. and/or stabilize nascent blood vessels. Hence, ECM
Other tyrosine kinase receptors of outstanding sig- molecules and ECM remodeling events play a key role
nificance in this regard are Tie-1 and Tie-2. Along with in regulating angiogenesis.51
the VEGF receptor, these are the only known endothelial The formation of the capillary-like tubes is specific
cell-specific receptor tyrosine kinases. Tie-2 is induced to endothelial cells and integral to the process of angio-
on the endothelium of neovessels in skin wounds and genesis. The basement membrane represents a biologi-
downregulated as newly formed vessels regress. As an cally functional highly specialized ECM on which the
indicator of Tie-2 activation, Tie-2 phosphorylation is basal nonluminal surface of endothelial cells rests. This
detected in skin wounds at all stages of the healing matrix forms a continuous sleeve around the endothe-
process.50 Activated microvascular endothelial cells lial cells, and maintains the tube-like structures of the
respond by proliferating, which is followed by direc- blood vessels. More than 20 years ago Kubota et al.
tional migration of these cells. Migration is a complex observed that endothelial cells plated on a reconstituted
252 • 14 • Wound healing

basement membrane matrix, rapidly attached, aligned, acquire smooth-muscle cell characteristics and differen-
and formed capillary-like tubules. The cells did not pro- tiate into contractile myofibroblasts. The first pheno-
liferate. The vessels that are thus formed contain a typic transition of wound edge fibroblasts into so-called
lumen and tight cell–cell contacts. The cells are polar- protomyofibroblasts is characterized by neoformation
ized with the nuclei basally located towards the base- of contractile β-cytoplasmic actin stress fibers and occurs
ment membrane matrix. Furthermore, the capillary-like in response to profibrotic cytokines and to altered prop-
structures take up acetylated low-density lipoprotein, erties of the ECM. In the presence of TGF-β1 in a
which is a marker of differentiation for these cells.52 mechanically restrained environment, these cells express
Angiogenesis not only depends on endothelial cell inva- α-smooth-muscle actin de novo, which significantly
sion and proliferation, but also requires pericyte cover- increases their contractile activity and is a hallmark of
age of vascular sprouts for vessel stabilization. These the differentiated myofibroblast.59 Wound contraction
processes are coordinated by VEGF and PDGF through may significantly contribute to wound closure, although
their cognate receptors on endothelial cells and vascular this contribution is much larger in loose-skinned rodents
smooth-muscle cells, respectively.53 Structural support than in humans.
to blood vessels is provided to pericytes and vascular In an open wound that has undergone contraction,
smooth-muscle cells. Normal pericytes are embedded restoration of an intact epidermal barrier is enabled
within the basement membrane of capillaries, either as through wound epithelialization, also known as
solitary cells or a single-cell layer, where they coordinate re-epithelialization.60,61 A wound that is not epithelial-
intercellular signaling with endothelial cells and other ized is not considered “healed,” no matter how
components of the blood vessel wall to prevent leakage. perfectly restored the underlying dermal structures
In contrast, vascular smooth-muscle cells form single or may be. Thus, wound epithelialization, also called
multiple layers around arteries and veins to mediate re-epithelialization, is a critical and defining feature of
vascular tone and contraction. Pericyte coverage is wound repair. Re-epithelialization of the wound can be
required for the stabilization of immature endothelial conceptually viewed as the result of three overlapping
tubes.54 Pericytes have function beyond angiogenesis keratinocyte functions: migration, proliferation, and dif-
that are relevant to wound healing. Skin pericytes may ferentiation. The sequence of events by which keratino-
act as mesenchymal stem cells (MSCs), exhibiting the cytes accomplish the task of re-epithelialization is
capacity to differentiate into bone, fat, and cartilage line- generally believed to begin with dissolution of cell–cell
ages. Thus, pericytes represent a potent stem cell popu- and cell–substratum contacts. This is followed by the
lation in the skin that is capable of modifying the ECM polarization and initiation of directional migration in
microenvironment and promoting epidermal tissue basal and a subset of suprabasilar keratinocytes over the
renewal from nonstem cells.55 provisional wound matrix. A subset of keratinocytes
immediately adjacent to, but not within, the wound bed
then undergoes mitosis. Finally, there is multilayering
Wound closure of the newly formed epidermis and induction of dif-
ferentiation specific gene products to restore the func-
Wound contraction and re-epithelialization contribute tionality of the epidermis. The most limiting factor in
to closure of wounds that heal by secondary intention. wound re-epithelialization is migration, since defects in
Wound contraction represents an early response to this function, but not in proliferation or differentiation,
injury that is aimed at juxtaposing the edges of an open are associated with the clinical phenotype of chronic
wound.56 This early phase of wound closure appears to nonhealing wounds.62 The process of epithelialization
be mediated by a contractile “purse-string” force pro- continues until the barrier is re-established and the
duced by a circumferentially arranged band of fusiform- wound is covered. The process of re-epithelialization is
shaped epidermal cells situated in the wound margin.57 accelerated by a moist environment63,64 and is facilitated
Fibroblasts at the wound edge tissue are recognized to by the enzyme matrix metalloproteinase 1, a collagenase
play a key role in enabling wound contraction.58 During which lessens the affinity of the collagen–integrin
the inflammatory phase of wound healing, fibroblasts contacts.65
Chronic wounds 253

Proliferative phase surgical textbooks define chronic wounds as wounds


which have not healed in 3 months. Chronic wounds
The proliferative phase starts around 2 days after injury can be broadly classified into three major categories:
and normally lasts up to 3 weeks in a healing cutaneous venous and arterial ulcers, diabetic ulcers, and pressure
wound. This phase overlaps with the inflammatory ulcers.
phase and supports re-epithelialization, the formation
of new blood vessels, and the influx of fibroblasts and Venous ulcers
laying down of the ECM. By the time this phase begins,
the degradation of the fibrin clot by the macrophages Venous ulcers (stasis ulcer or varicose ulcers) are wounds
has begun and invading endothelial cells and fibroblasts that are thought to occur due to improper functioning
rapidly fill that space. Migrating fibroblasts produce of venous valves, usually of the legs. They are the major
the cytokines that induce keratinocytes to migrate and cause of chronic wounds, occurring in 50–70% of chronic
proliferate.66 Activated macrophages produce several wound cases.68 Venous ulcers develop mostly along the
cytokines, such as PDGF and TNF-α, which also induce medial distal leg, and can be very painful. According to
fibroblasts to produce keratinocyte growth factor which the revised clinical, etiology, anatomy, and pathophysi-
in turn induces wound re-epithelialization.60,61 ology (CEAP) classification of chronic venous disease
published in 2004, a venous ulcer is defined as a full-
thickness defect of skin, most frequently in the ankle
Granulation tissue
region, that fails to heal spontaneously and is sustained
The fibrin clot formed during hemostasis participates in by chronic venous disease.69 Systematically, venous
the early inflammatory phase and is replaced by a per- ulcer may be defined as a defect in the skin with sur-
fused, fibrous connective tissue that grows from the rounding pigmentation and dermatitis, located in the
base of a wound and is able to fill wounds of almost any lower leg (usually in the gaiter region) that has been
size. During the proliferative phase of wound healing, present for greater than 30 days, characterized by
this granulation tissue is light red or dark pink in color persistent venous hypertension and abnormal venous
because of perfusion by new capillary loops. It is soft to function (result of venous reflux and/or obstruction
the touch, moist, and granular in appearance. The gran- confirmed by hemodynamic and/or physiologic assess-
ulation tissue serves as a bed for tissue repair. The ECM ment), without a primary or associated arterial, immu-
of granulation tissue is created and modified by fibrob- nologic, endocrine, or systemic cause. It is recognized
lasts. Initially, it consists of a network of type III colla- that ulcers can be caused purely by venous pathology
gen, a weaker form of the structural protein that can be such as venous reflux or obstruction. When these abnor-
produced rapidly. This is later replaced by the stronger, malities are combined with additional pathologic condi-
long-stranded type I collagen, as evidenced in scar tions, they contribute to the causation and perpetuation
tissue. Formation and contraction of the granulation of the ulcer. The latter situation includes comorbid con-
tissue represent integral aspects of the healing wound. ditions such as arterial ischemia, swelling and lymph-
In ischemic wounds, contraction of the granulation edema, trauma, autoimmune disorders, neurotrophic
tissue is impaired because of faulty myofibroblast conditions, and diabetic vascular disease. These ulcers
function, cells responsible for granulation tissue are categorized as of mixed origin, in which the venous
contraction.67 component may or may not play a dominant role.
Successful treatment of such ulcers includes not only the
venous component but also concomitant management
Chronic wounds of the comorbid condition.

A wound is generally considered chronic if it has not Arterial ulcers


healed in 4 weeks. Chronic wounds have been also
defined as wounds that have not shown a 20–40% reduc- Because both arterial and venous ulcers typically occur
tion in area after 2–4 weeks of optimal therapy. Standard 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 man-
agement pathways. The most common cause of arterial
ulcers is atherosclerosis. Risk factors for the develop-
ment 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 arte-
rial insufficiency can lead to the formation of lower-
extremity 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 charac-
teristic of arterial ulcers. Determining the ankle brachial
index give an indication of a patient’s ability to heal.
However, diabetic patients may have falsely elevated
ankle brachial index results secondary to vessel calcifi- Healthy foot Diabetic foot
cation. Patients with arterial ulcers must have increased/
Blood vessel damage in the feet may cause tissue damage
adequate blood supply to heal and benefit most from such as sores, lesions and poor circulation that can lead to amputation
revascularization procedures. It should be noted that
Fig. 14.10  The ischemic diabetic foot.
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 arte-
rial insufficiency.

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 athero-
sclerotic disease of large- and medium-sized arteries,
such as aortoiliac and femoropopliteal atherosclerosis.
However, significant atherosclerotic disease of the infra-
popliteal segments is particularly common in the
diabetic population. Underlying digital artery disease,
when compounded by an infected ulcer in close proxim-
ity, may result in complete loss of digital collaterals and
precipitate gangrene. The reason for the prevalence of Diabetic foot ulcer
this form of arterial disease in diabetic persons is thought
to result from a number of metabolic abnormalities, Fig. 14.11  Diabetic foot ulcer.
Chronic wounds 255

motor nerves can lead to clawing of the toes because of


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

Pressure ulcers
A pressure ulcer is a localized injury to the skin or
underlying tissue, usually over a bony prominence, as
Dorsal surface Plantar surface
a result of unrelieved pressure. Pressure ulcers or pres-
Fig. 14.12  Cutaneous innervations of the foot. sure sores represent a common health problem, particu-
larly among the physically limited or bedridden elderly.
including high low-density lipoprotein and very- Pressure ulcers on the buttocks affect nearly all wheel-
low-density lipoprotein levels, elevated plasma von chair users. Pressure ulcer is a general term covering a
Willebrand factor, inhibition of prostacyclin synthesis, number of different tissue injuries, from superficial heel
elevated plasma fibrinogen levels, and increased plate- sores to deep pressure sores under the buttocks. Because
let adhesiveness.71 When peripheral arterial insuffi- compressive forces, shearing forces, and/or friction are
ciency complicates neuropathy there is a 10-fold risk of the major underlying causes some call them decubitus
ulceration progressing to infection, gangrene, and ulcer.75 Pressure ulcers occur in approximately 5–15% of
amputation.72 patients in home care, healthcare facilities, and hospi-
Peripheral sensory neuropathy affects 50% of diabetic tals. Pressure ulcers are painful, decrease the quality of
patients and is attributed to chronic hyperglycemia. It life, increase susceptibility to infections, risk of death
is a major cause of foot ulceration and lower limb ampu- and nursing workload, and create significant costs.76
tation. In addition to poor glucose control, traditional Predisposing factors are classified as intrinsic (e.g.,
cardiovascular risk factors for macrovascular disease limited mobility, poor nutrition, comorbidities, aging
are independent-risk factors for incident peripheral skin) or extrinsic (e.g., pressure, friction, shear, mois-
neuropathy. In addition, data from the EURODIAB ture). When an ulcer occurs, documentation of each
cohort suggest that female sex may be an independent- ulcer (i.e., size, location, eschar and granulation tissue,
risk factor. This makes it difficult to identify specific exudate, odor, sinus tracts, undermining, and infection)
diabetes components of neuropathy.73 Nerve injuries of and appropriate staging (I through IV) are essential to
the foot can also be caused by penetrating wounds. The the wound assessment.77
sequelae of such injuries depend on the nerve injured While there is little doubt that pressure ulcers are
and the level of the injury. In the foot and ankle, the related to the mechanical insult of soft tissue, several
main function of the nerves is to provide sensation. hypotheses have been developed pertaining to the link
Generally, the tibial nerve and its branches (i.e., the between mechanical loading and tissue necrosis. The
medial and lateral plantar nerves) innervate the intrin- two major hypotheses deal with tissue deformation and
sic musculature, although the deep peroneal nerve ischemia.78 Pressure-induced soft-tissue deformation
innervates the extensor digitorum brevis and extensor causes cell necrosis. There is a time/strain relationship,
hallucis brevis muscles (Fig. 14.12). Denervation of these 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


ATP
chronic basis results in ulcer formation. Prevention
includes identifying at-risk persons and implementing O2 Red blood
cells
specific prevention measures such as following a patient-
NO
repositioning 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 Protein
by lack of blood supply to any given tissue. The hypoth- synthesis Mature collagen

esis proposes that a mechanical loading of the tissue Superoxide


Neutrophil
anion radical
impinges on the arterial blood vessels, thereby causing
local ischemia. Since cells depend on oxygen, heat, and Macrophage
nutrients transported by the blood, they will become Endothelial cells
hypoxic and subsequently necrotic.
Infectious
pathogens H2O2

Ischemia and tissue oxygenation Redox


signals

Vascular complications commonly associated with HOCl


problematic wounds are primarily responsible for Fig. 14.13  Molecular oxygen and its derivatives in wound healing. ATP, adenosine
wound ischemia. Limitations in the ability of the vascu- triphosphate; NO, nitric oxide.
lature 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, role in the regulation of vascular tone as well as in ang-
whereas ischemia is a lack of perfusion, characterized iogenesis. In a wound setting, large amounts of molecu-
not only by hypoxia but also by insufficient nutrient lar oxygen are partially reduced to form reactive oxygen
supply.79 Hypoxia, by definition, is a relative term. It is species (ROS). ROS includes oxygen free radicals such
defined by a lower tissue partial pressure of oxygen as superoxide anion as well as its nonradical derivative,
(pO2) compared to the pO2 to which the specific tissue hydrogen peroxide (H2O2). Superoxide anion radical is
element in question is adjusted under healthy condi- the one-electron reduction product of oxygen. NADPH
tions in vivo. Depending on the magnitude, cells con- oxidases represent one major source of superoxide anion
fronting hypoxic challenge either induce an adaptive radicals at the wound site. NADPH oxidases in phago-
response that includes increasing the rates of glycolysis cytic cells help fight infection. Superoxide anion also
and conserve energy or undergo cell death. Generally, drives endothelial cell signaling such as required during
acute mild to moderate hypoxia supports adaptation angiogenesis. In biological tissues, superoxide anion
and survival. In contrast, chronic extreme hypoxia leads radical rapidly dismutates to hydrogen peroxide, either
to tissue loss. spontaneously or facilitated by enzymes called super-
While the tumor tissue is metabolically designed to oxide dismutases. Endogenous hydrogen peroxide
thrive under conditions of hypoxia, hypoxia of the drives redox signaling, a molecular network of signal
wound primarily caused by vascular limitations is propagation that supports key aspects of wound healing
intensified by coincident conditions (e.g., infection, such as cell migration, proliferation, and angiogenesis.
pain, anxiety and hyperthermia) and leads to poor Neutrophil-derived hydrogen peroxide may be utilized
healing outcomes. Oxygen and its reactive derivatives by MPO to mediate peroxidation of chloride ions, result-
(Fig. 14.13) are required for oxidative metabolism- ing in the formation of hypochlorous acid (HOCl), a
derived energy synthesis, protein synthesis, and the potent disinfectant (Fig. 14.13).
maturation (hydroxylation) of extracellular matrices Three major factors may contribute to wound tissue
such as collagen. Molecular oxygen is also required for hypoxia: (1) peripheral vascular diseases garroting O2
nitric oxide (NO) synthesis which in turn plays a key supply; (2) increased O2 demand of the healing tissue;
Ischemia and tissue oxygenation 257

Bacteria
Stratum corneum
Stratum
granulosum
Keratinocyte in
epidermis
Basal layer

Myofibroblast
Neutrophil
Endothelial
progenitor cell

Endothelial Macrophage
cell
Dermis 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
Pocket of oxygenation represents oxygenated tissue. Tissue around each blood
vessel is dark pink in shade, representing regions that are
Pocket of hypoxia well oxygenated (oxygen-rich pockets). Bacteria and
Near-anoxic necrotic core bacterial infection are presented by shades of green on the
surface of the open wound.

and (3) generation of ROS by way of respiratory burst of VEGF and its receptors leads to insufficient skin ang-
and for redox signaling.80 Other related factors, such as iogenesis.81 Whether cells in the pockets of extreme
arterial hypoxia (e.g., pulmonary fibrosis or pneumonia, hypoxia are O2-responsive is another concern. Even if
sympathetic response to pain, hypothermia, anemia such cells may have passed the point of no return in the
caused by major blood loss, cyanotic heart disease, high survival curve, correction of tissue oxygenation is likely
altitude), may contribute to wound hypoxia as well. to help clean up the dead or dying tissue and replace
Depending on factors such as these, it is important to the void with proliferating neighboring cells. Pockets of
recognize that wound hypoxia may range anywhere moderate or mild hypoxia are likely to be the point of
from near anoxia to mild to modest hypoxia. In this origin of successful angiogenic response as long as other
context it is also important to appreciate that point barriers such as infection and epigenetic alterations are
measurements performed in the wound tissue may not kept to a minimum.
provide a complete picture of the wound tissue biology
because it is likely that the magnitude of wound hypoxia Limited supply and high demand:
is not uniformly distributed throughout the affected the oxygen imbalance
tissue, especially in large wounds. This is most likely
the case in chronic wounds presented clinically as Peripheral vascular disease can affect the arteries and
opposed to experimental wounds which are more con- the veins as well as the lymph vessels. The most common
trolled and homogeneous in nature. In any single and important type of peripheral vascular disease is
problem wound presented in the clinic, it is likely that peripheral arterial disease, or PAD, which affects about
there are pockets of near anoxia as well as that of differ- 8 million Americans. The ankle brachial pressure index
ent grades of hypoxia (Fig. 14.14). As the weakest link represents a simple noninvasive method to detect arte-
in the chain, tissue at the near-anoxic pockets will be rial insufficiency within a limb. Arterial diseases, espe-
vulnerable to necrosis which in turn may propagate cially those associated with diabetes, represent a major
secondary tissue damage and infection. Pockets of complicating factor in wound healing. PAD is the only
extreme hypoxia may be flooded with hypoxia-inducible identifiable etiology in approximately 10% of leg ulcers.
angiogenic factors but would fail to vascularize func- In an ischemic limb, peripheral tissues are deprived of
tionally because of insufficient O2 that is necessary to blood supply as PAD progresses, causing tissue loss,
fuel the repair process. Indeed, uncontrolled expression ulcers, and gangrene.
258 • 14 • Wound healing

Venous insufficiency, on the other hand, is the root systems have been studied for their effect on wound
cause of most leg ulcers. Chronic venous insufficiency, healing. While these approaches may compensate for
characterized by the retrograde flow of blood in the the deficiency of ATP per se in the ischemic wound
lower extremity, is associated with changes in the venous tissue, they will fail to address the other essential func-
wall and valves generally caused by inflammatory dis- tions of O2 and its derivatives in wound healing, as
orders induced by venous hypertension and associated discussed below.
fluid shear stress. Factors causing arterial hypoxemia Absolute requirements for O2 arise in several points
may also limit O2 supply to the wound tissue. along the angiogenic sequence. For instance, all vessels
Compromised pulmonary health, loss of hepatic func- require a net or sheath of ECM, mainly collagen and
tion, hemodialysis, anemia, altitude hypoxemia, nitro- proteoglycans, to guide tube formation and resist the
glycerin therapy, nasal packing, critical illness, pain, pressures of blood flow. Conditions for collagen deposi-
and hypothermia are examples of conditions associated tion and polymerization can be created only if molecu-
with arterial hypoxemia. Vasoconstricting drugs may lar O2 is available to be incorporated into the structure
contribute to tissue hypoxia as well.79 of nascent collagen by prolyl- and lysyl hydroxylases.
Increased energy demand of the healing tissue leads Without the obligatory extracellular hydroxylated col-
to a hypermetabolic state wherein additional energy is lagen, new capillary tubes assemble poorly and remain
generated from oxidative metabolism, increasing the O2 fragile.82–84 This has a convincing clinical correlate in
demand of the healing tissue. Adenosine triphosphate scurvy, i.e., ascorbate deficiency. Scurvy results from
(ATP) thus generated powers tissue repair. At the injury insufficient intake of ascorbate which is required for
site, extracellular ATP may be contributed by platelets correct collagen synthesis in humans. Ascorbate is
and other disintegrating cells. Extracellular ATP liber- required for the posttranslational hydroxylation of col-
ated during hypoxia or inflammation can either signal lagen that enables the matured collagen molecules to
directly to purinergic receptors or, after phosphohydro- escape to the extracellular space and provide the neces-
lytic metabolism, can activate surface adenosine recep- sary tensile strength. In scurvy, the collagenous sheath
tors. Purinergic signaling may influence numerous cannot form because, under ascorbate-deficient condi-
aspects of wound biology, including immune response, tions, collagen cannot be hydroxylated. Consequently,
inflammation, vascular as well as epithelial biology. new vessels fail to mature. Older vessels weaken and
ATP may be immunostimulatory or vice versa, depend- break, and wounds fail to heal. Thus, while hypoxia is
ing on extracellular concentrations as well as on expres- a proved instigator of molecular signals for angiogen-
sion patterns of purinergic receptors and ectoenzymes. esis, it is also a proven enemy of vessel growth itself in
Extracellular ATP induces receptor activation in epithe- nontumor tissues. Collagen deposition proceeds in
lial cells. ATP, released upon epithelial injury, acts as direct proportion to pO2 across the entire physiologic
an early signal to trigger cell responses, including an range, from zero to hundreds of mmHg. The Km for
increase in heparin-binding epidermal growth factor O2 for this reaction is approximately 25 and the Vmax
(EGF)-like growth factor shedding, subsequent transac- is approximately 250 mmHg, suggesting that new
tivation of the EGF receptor and its downstream signal- vessels cannot even approach their greatest possible
ing, resulting in wound healing. ATP released from the rate of growth unless the wound tissue pO2 is high.85
injured epithelial cells is now known also to turn on Angiogenesis is directly proportional to pO2 in injured
NADPH oxidases, the activity of which is critically tissues.83 Hypoxic wounds deposit collagen poorly and
required to produce the redox signals required for become infected easily, both of which are problems of
wound healing.80 Human endothelial cells are rich in considerable clinical significance.79
purinergic receptors and therefore responsive to extra-
cellular ATP as well. ATP induces endothelium- Redox signaling
dependent vasodilation. Both ATP as well as adenosine
regulate smooth muscle and endothelial cell prolifera- Additional high demand for oxygen is placed by a
tion. Recognizing that hypoxia limits ATP synthesis in family of enzymes known as NADPH oxidases, which
the ischemic wound tissue, therapeutic ATP delivery are known to be highly active at the wound site.86 Recent
MicroRNAs 259

work has identified that oxygen is not only required to inherent tissue repair program that seeks to restore the
disinfect wounds and fuel healing but that oxygen- injured tissue both structurally as well as functionally.
dependent redox-sensitive signaling processes repre- There are two key steps that separate a protein-coding
sent an integral component of the healing cascade.80 The gene from its corresponding protein. First, the DNA
widely held notion that biological free radicals are hosting the gene must transcribe to mRNA. Finally, the
necessarily agents of destruction is now facing serious mRNA must be translated to protein. Work emerging
challenge.87 Over a decade ago it was proposed that in during recent years demonstrates that both of these
biological systems oxidants are not necessarily always critical steps are subject to robust and redundant regula-
the triggers for oxidative damage and that oxidants tion by microRNAs (miRNAs: 19–22 nucleotides long),
such as H2O2 could actually serve as signaling messen- which are noncoding RNAs found in all eukaryotic
gers and drive several aspects of cellular signaling.88 cells. Work during the past decade recognizes small
Today, that concept is much more developed and mature. RNAs as a new class of regulators of eukaryotic biology.
Evidence supporting the role of oxidants such as H2O2 Alongside other small interfering RNAs (siRNAs),
as signaling messenger is compelling.89–99 miRNAs execute posttranscriptional gene silencing
through mRNA destabilization as well as translational
Nitric oxide repression. In simple words, whether a gene would
code a protein or not is decided upon by miRNAs for
At the wound site, NO is generated by an oxygen- which the gene is a target. miRNAs form basepairs with
dependent biosynthetic process. In the late 1970s, specific sequences in protein-coding mRNAs. Near-
research was unfolding that implicated NO involve- perfect pairing induces cleavage of the target mRNA,
ment in the process of vasodilation. By 1986, research whereas partial pairing results in translational repres-
culminated in the identification of NO as the endothelium- sion and mRNA decay through deadenylation path-
derived relaxing factor responsible for the maintenance ways.105 According to the miRbase database, the human
of vascular tone, thus implicating NO as a potential genome encodes 1048 miRNAs. This count is rapidly
wound-healing agent.100 Maximal NO synthase activity growing. These miRNAs may regulate more than a third
is noted early in cutaneous wound healing, with sus- of all protein-coding genes and virtually all biological
tained production up to 10 days after wounding. Wound processes. Mammalian cells express cell type-specific
macrophages represent a major source of NO produc- miRNAs which silence unique subsets of target genes
tion in the early phase of wound healing.101 Inhibition within the cell. While miRNAs are mostly known for
of wound NO synthesis lowered wound collagen accu- being functional in the cytoplasm, nuclear miRNAs may
mulation and wound-breaking strength, suggesting that also participate in gene regulation. Initially considered
NO synthesis is critical to wound collagen accumulation an oddity, miRNA-dependent control of gene expres-
and acquisition of mechanical strength. Later it was sion is now accepted as being integral to the normal
demonstrated that wound fibroblasts are phenotypi- function of cells and organisms. miRNAs are emerging
cally altered during the healing process to synthesize as key regulators of the overall wound-healing process.106
NO, which, in turn, regulates their collagen synthetic
and contractile activities.102 The blockade of NO synthe- Inflammation
sis impairs cutaneous wound healing, acting in early
and late phases of wound repair.103 Interestingly, Disruption of miRNA biogenesis has a major impact on
impaired diabetic wound healing is associated with the overall immune system. Emerging studies indicate
decreased wound NO synthesis.104 that miRNAs, especially miR-21, miR-146a/b, and miR-
155, play a key role in regulating several hubs that
orchestrate the inflammatory process.107 miRNAs have
MicroRNAs been directly implicated in the pathogenesis of inflam-
matory diseases such as osteoarthritis and rheumatoid
Wound healing is largely dependent on injury-inducible arthritis. Resolvin-regulated specific miRNAs target
protein-coding genes as they serve as drivers of an genes involved in resolution of inflammation establish
260 • 14 • Wound healing

a novel resolution circuit involving RvD1 receptor- hypoxamir. Expression of hypoxia-inducible factor 1
dependent regulation of specific miRNAs.108 Fur­ (HIF-1) is also controlled by specific miRNAs. In turn,
thermore, the brain-specific microRNA-124 can tame HIF-1 controls the expression of hypoxamirs, which
inflammation by turning off activated microglial cells are induced in the injured tissue.117 Hypoxamirs are
and macrophages.109 Of relevance to tissue repair is also also induced by HIF-independent pathways. Although
the regulatory loop where cytokines, including those hypoxamirs generally favor angiogenesis, their meta-
elicited following injury, are regulated by miRNAs as bolic and cell cycle arrest functions are in conflict with
well as regulate miRNA expression.110,111 wound healing, especially in an ischemic setting.
Silencing specific hypoxamirs may therefore represent a
Angiogenesis prudent approach to facilitate tissue repair. miRNA-210
represses mitochondrial respiration116 and exaggerates
In 2005–2008, the first series of observations establishing production of undesired mitochondrial ROS.118 These
key significance of miRNAs in the regulation of mam- outcomes are not compatible with the higher energy
malian vascular biology came from experimental studies demands associated with tissue repair. miR-210 also
involved in arresting miRNA biogenesis to deplete silences signaling via FGF,119 a key contributor to wound
the miRNA pools of vascular tissues and cells.112 Dicer- healing. The injured tissue is highly rich in ROS.86 In
dependent biogenesis of miRNA is required for blood addition, at the site of injury transition metal ions are
vessel development during embryogenesis. Mice with released from a protein-bound state. Conditions such as
endothelial cell-specific deletion of Dicer, a key enzyme these cause DNA damage which opposes tissue repair.
supporting biogenesis of miRNAs, display defective DNA repair systems are therefore of key significance in
postnatal angiogenesis. NADPH oxidase-derived ROS enabling tissue repair. miR-210 antagonizes DNA
drive wound angiogenesis. Endothelial NADPH oxidase repair.120 This is another hypoxamir function that is in
is subject to control by miRNAs.113 Hypoxia is widely conflict with wound healing. Compatible with the
recognized as a cue that drives angiogenesis as part of common observation that ischemic wounds are refrac-
an adaptive response to vascularize the oxygen-deficient tory to healing response, elevated miRNA-210 in
host tissue. Hypoxia-sensitive miR-200b is involved in ischemic wounds attenuated keratinocyte proliferation
such induction of angiogenesis via directly targeting and impaired wound closure.106,121
Ets-1.114 Various aspects of angiogenesis, such as prolif-
eration, migration, and morphogenesis of endothelial Stem cells
cells, can be regulated by specific miRNAs in an
endothelial-specific manner. miRNAs known to regu- Endogenous miRNA-binding sites have been identified
late angiogenesis in vivo are referred to as angiomiRs.115 in murine embryonic stem cells (ESCs). miRNAs govern
miRNA-126 is specific to endothelial cells and regulates ESCs function by serving as control hubs managing
vascular integrity and developmental angiogenesis. regulatory networks. A central importance of such gov-
Manipulating angiomiRs in the setting of tissue repair ernance is highlighted by the observation that ESCs
represents a new therapeutic approach that could be lacking miRNAs lose their “stemness.” ESCs with defi-
effective in promoting wound angiogenesis. cient miRNA biogenesis systems switch to a mode of
ongoing cell division. They do not differentiate on
Hypoxia response demand because of failure to turn off the pluripotency
regulatory program.122 miRNAs conduct the orchestra
Tissue injury is often associated with disruption of vas- of critical gene regulatory networks controlled by
cular supply to the injury site. Thus, the injured tissue pluripotency factors within stem cells. Individual
often suffers from insufficient oxygen supply or hypoxia. miRNA-dependent pathways that promote the repro-
Under conditions of additional underlying ischemia, gramming of somatic cells into induced pluripotent
hypoxia is severe and seriously limits wound healing.79 stem (iPS) cells have been now identified. Manipulation
Hypoxia induces specific miRNAs, collectively referred of specific cellular miRNAs helps enhance reprogram-
to as hypoxamirs.116 miRNA-210 is a classical ming of somatic cells to an ESC-like phenotype, helping
Stem cells 261

to generate iPS cells.123 Expression of miRNAs is also


Totipotent cells
subject to control by epigenetic factors.124 Such control
influences the balance between proliferation and dif-
ferentiation of stem cells. In executing such control, the
miRNA element of epigenetics cross-talks with changes Pluripotent stem cell

in chromatin structure as well as with changes in DNA


methylation. Collectively, this provides for a mecha- Blood Other committed
nism by which the tissue injury microenvironment can stem cells stem cells
influence miRNA-dependent reparative and regenera-
tive processes.

Erythrocytes Platelets White blood cells


Stem cells
Specialized cells
The regenerative potential of injured adult tissue sug-
Fig. 14.15  Stem cell renewal.
gests the physiological existence of cells capable of par-
ticipating in the reparative process. The epithelium of
the skin, the epidermis, is in a continuous equilibrium
of growth and differentiation and has the remarkable Totipotent

on

Re
ati
capacity to self-renew completely, which relies on reser-

-d
cells

nti

iffe
re

re
iffe
voirs of stem cells. In mammals, there are two broad

nti
n

Di
tio
-d

ati
ffe
tro

tia

on
types of stem cells: ESCs that are isolated from the inner

re
Re

en
Pluripotent

nt
er

iat
cell mass of blastocysts, and adult stem cells that are iff stem cell

io
-d

n
De

found in various tissues. In adult organisms, stem cells


and progenitor cells act as a repair system for the body, Progenitor cells
replenished in adult tissues. Stem cells have the prop- Blood
erty of self-renewal without differentiation and have the stem
cells
potential to differentiate into any type of cell. Totipotent
stem cells have the ability to give rise to a whole organ- Other
ism due to their ability to differentiate into embryonic tissues
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. Red blood White blood Muscle Nerve Bone
14.15). ESCs are derived from the developing embryo, cells cells

usually from the inner cell mass of a blastocyst or earlier


Trans-differentiation
morula stages. Lost or damaged cells can be replaced by
differentiation, dedifferentiation, or transdifferentiation Fig. 14.16  Stem cell differentiation, dedifferentiation, and transdifferentiation.
(Fig. 14.16). Recent advances have shown that the addi-
tion of a group of genes can not only restore pluripo- lost. Transdifferentiation is another naturally occurring
tency in a fully differentiated cell state (differentiation) mechanism that takes dedifferentiation a step further
but can also induce the cell to proliferate (dedifferentia- and sees cells regressing to a point where they can
tion) or even switch to another cell type (transdifferen- switch lineages, allowing them to differentiate into
tiation). Dedifferentiation is represented by a terminally another cell type. Furthermore, reprogramming aims to
differentiated cell reverting back to a less-differentiated revert differentiated cells to pluripotency. From here,
stage from within its own lineage. This process allows they can differentiate into almost any cell type. Although
the cell to proliferate again before redifferentiating, reprogramming occurs naturally during fertilization to
leading to the replacement of those cells that have been produce totipotent cells that can differentiate into any
262 • 14 • Wound healing

Hair shaft

Sebaceous gland
Skeletal
Liver muscle Outer root sheath
“Bulge’ stem cells
Inner root sheath

Bone marrow Basement


stromal cell membrane Matrix stem cells
Dermal papilla
Epithelial A
cell

Blood cells Neuron

Glial cell TA cell


Blood
Basement membrane
vessel
CNS
stem cells

Paneth cell
Fat cell Cardiac muscle Stem cell
Mesenchymal cell
B
Fig. 14.17  Bone marrow stem cells. CNS, central nervous system.
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
cell type, it has not yet formally been shown to be a differentiate into various parts of the hair while the long-term stem cells are present
genuine regenerative response. Furthermore, repro- 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
gramming sidesteps the necessity of using embryos basal region with the Paneth cells. The transit amplifying (TA) cells are stem cell
for regenerative therapies by using differentiated cells progeny which move upwards and differentiate.
taken from a patient. From a clinical perspective, this
comes with the additional bonus of circumventing stem/progenitor cells derived from the bone marrow
the immunological problems such as transplant rejec- could home to injured tissues and participate in the
tion and graft-versus-host disease associated with repair/regeneration. Moreover, culture-expanded bone
engraftment.125 marrow-derived MSCs have been shown to promote the
The bone marrow (Fig. 14.17), home of stem and pro- healing of diabetic wounds, implying a profound thera-
genitor cells, contributes a significant proportion of cells peutic potential for skin defects such as chronic wounds
in the skin. The normal skin has long been known to and burns.126
contain bone marrow-derived cells that are involved in During the inflammatory phase, leukocytes migrat-
host defense and inflammatory processes, including ing to the wound site are hematopoietic cells derived
wound healing. However, recent studies demonstrate from the bone marrow. In the skin the bulge of the hair
that the bone marrow contributes not only inflamma- follicle (Fig. 14.18) plays a role as a reservoir of stem
tory cells, but also keratinocytes and fibroblast-shaped cells. In mice it has been shown that the bulge region
cells to the skin. Similar to leukocytes in trafficking, contains hematopoietic cells that are identical to the
Stem cells 263

bone marrow-derived cells and also to those found in protective barrier against its external environment. With
fetal circulation.127 Moreover this region also serves as a rare exceptions (such as palms and eyelids), hair folli-
reservoir for precursors of mast cells. Discovery of these cles are present all over the skin and play an important
epidermal stem cells in the hair follicle bulge led to the role in physiological tissue renewal and regeneration
hypothesis that these cells are necessary for both epider- after injury. Hair follicles represent an autonomous min-
mal renewal as well as wound healing.128 It was shown iorgan, which provides an excellent model system for
that cells from the bulge do not contribute to epidermal studying the biology of adult stem cells.141 There are
regeneration; however upon injury cells from the bulge obvious differences between human and mouse skin,
are recruited into the epidermis and migrate in a linear yet the knowledge gained from lineage-tracing experi-
manner toward the center of the wound.129 These cells ments in mice has greatly expanded our understanding
were noted to be transient, living only for a few weeks, of the cellular behavior of the different keratinocyte
thus representing an acute response to injury. populations during physiological and injury-induced
There are two main branches of stem cells in the bone tissue regeneration. As in most of the body’s organs, the
marrow, including hematopoietic stem cells (HSCs) and skin experiences constant renewal. The upper kerati-
MSCs. Adult bone marrow-derived HSCs have long nized layer of the interfollicular epidermis, which con-
been recognized as a precursor to all blood cell lineages, sists of terminally differentiated cells, is shed and
including erythrocytes, platelets, and white blood cells. replaced by cells originating from the actively prolifer-
Additionally, HSCs may also give rise to fibrocytes and ating layer beneath. In contrast, hair follicles undergo
endothelial progenitor cells. Circulating endothelial cycles of growth (anagen) and rest (telogen). The poten-
precursor cells play a role in neoangiogenesis which is tial for proliferation to sustain the lifelong replenish-
essential for healing.130,131 Bone marrow-derived stem ment of normal cell loss and to repair occasional tissue
cells also contribute to the deposition of collagen III in damage lies within the population of epidermal stem
the wound132 and differentiate into fibroblasts,133 kerati- cells. The hair follicle bulge harbors stem cells that may
nocytes,134 and fibrocytes.135 Bone marrow-derived help in renewal and repair of the skin. The term “bulge,”
MSCs, which are also referred to as bone marrow mes- originally called “der Wulst,” was introduced in 1903 by
enchymal stromal cells or marrow stromal cells, are self- a German morphologist, P. Stöhr, to describe an eminent
renewing and expandable stem cells. Although present structure at the site of attachment of the arrector pili
as a rare cell population in the bone marrow, represent- muscle in human hair follicles.141 Similar to many other
ing about 0.001–0.01% of the nucleated cells, about somatic stem cells, bulge cells are slow-cycling in nature.
10-fold less abundant than HSCs, MSCs are expandable This feature has permitted their initial identification and
in culture and multipotent, capable of differentiating isolation as label-retaining cells that can retain a pulse
into several cell types. of nucleotide label following a long chase period and
Because of their properties of regeneration and dif- is frequently regarded as a defining characteristic of
ferentiation the use of stem cells to heal problem wounds the hair follicle stem cell. In addition, the availability
has long been of interest. Indeed, autologous bone of several immunohistochemical markers, including
marrow aspirates and cultured cells were helpful in keratin-15 and CD34, that specifically label murine fol-
healing chronic wounds.136 In the case of burn wounds, licular stem cells has given researchers the ability to
bone marrow-derived stem cell treatment shows examine carefully the signals required for adult stem
promise as well.137 Another major source of adult stem cell activation and renewal. We now know that the
cells is adipose tissue.138 The capability of adipose- bulge serves as a repository of long-lived multipotent
derived adult stem cells to differentiate into bone, stem cells, imparted with the capacity to differentiate
muscle, fat, or cartilage, or into cells of mesenchymal into all cell types that constitute the lower cyclic portion
lineage, makes them a prime target for therapeutic use. of the hair follicle, as well as the interfollicular epider-
It has been shown that adult stem cells enhanced wound mis during wound repair.142,143
healing in a murine model.139,140 Although research into the use of stem cells for regen-
Hair follicles are an integral part of mammalian skin erative medicine is on a steep upward slope, clinical
where they help the epidermis to maintain the body’s success has not been as forthcoming. This has been
264 • 14 • Wound healing

primarily attributed to a lack of information on the basic (depressed) scars, scar contractures, hypertrophic scars,
biology of stem cells, which remains insufficient to and keloids. Postsurgical scar assessment is fundamen-
justify clinical studies. Since most clinical protocols use tal for a complete functional evaluation and as an
intravenous application of MSCs, it has become impor- outcome measure. The Vancouver Scar Scale is the most
tant to understand their trafficking in the blood stream. widely used rating scale for scars but the Patient and
Moreover, since relatively little is known where the Observer Scar Assessment Scale is recognized as the
transplanted MSCs might locate, a better understanding most comprehensive tool, taking into account the impor-
of involved homing mechanisms will likely shed light tant aspect of the patient’s perspective. Recently, a new
on how MSCs exert their therapeutic effects. For scale, called the Stony Brook Scar Evaluation Scale, has
instance, it is unclear whether mechanisms used at been proposed.147 Although scar remodeling occurs for
injured sites are location-specific or whether this recruit- months to years after the initial injury, complete restora-
ment can be modulated for therapeutic purposes. In tion of the normal ECM architecture is never achieved.
addition, it has recently been suggested that platelets Mature scars restore only 70% of the tensile strength of
may play an important role in stem cell recruitment to normal skin, and prescar function is never completely
sites of injury. A better understanding of the mecha- recovered.148
nisms used by stem cells during tissue homing would Scarring is an integral component of the healing
allow us to develop strategies to improve recruitment process and an outcome of the remodeling stage of
of these rare cells.144 wound repair which begins 2–3 weeks after injury and
lasts for a year or more.149 During this stage, all of the
Induced pluripotent stem cells processes activated after injury wind down and cease.
Most of the endothelial cells, macrophages, and myofi-
Pluripotent stem cells possess the unique property of broblasts undergo apoptosis or exit from the wound,
differentiating into all other cell types of the human leaving a mass that contains few cells and consists
body. The discovery of iPS cells in 2006 has opened up mostly of collagen and other ECM proteins.149,150
new avenues in clinical medicine.145,146 Recent break- Epithelial–mesenchymal interactions probably con­
through studies using a combination of four factors to tinuously regulate skin integrity and homeostasis. In
reprogram human somatic cells into pluripotent stem addition, over 6–12 months, the acellular matrix is
cells without using embryos or eggs have led to an actively remodeled from a mainly type III collagen
important revolution in stem cell research. It is now backbone to one predominantly composed of type I col-
possible to convert somatic cells, such as skin fibroblasts lagen.151 This process is carried out by matrix metallo-
and B lymphocytes, into pluripotent stem cells that proteinases that are secreted by epidermal cells,
closely resemble ESCs. Recently, functional neurons, endothelial cells, fibroblasts, and the macrophages
cardiomyocytes, pancreatic islet cells, hepatocytes, and remaining in the scar, and it strengthens the repaired
retinal cells have been derived from human iPSCs, thus tissue. However, the tissue never regains the properties
reconfirming the pluripotency and differentiation capac- of uninjured skin.
ity of these cells. These findings further open up the Fibroblasts at the wound site originating from sur-
possibility of using iPSCs in cell replacement therapy rounding tissues as well as supplied in the form of
for various disorders, including chronic wounds. marrow-derived blood-borne cells are recognized as the
primary drivers of scar formation. Platelets, macro-
phages, T lymphocytes, mast cells, Langerhans cells,
Scar and keratinocytes are directly and indirectly involved
in the activation of fibroblasts, which in turn produce
Scars (also called cicatrices) are macroscopic fibrous excess ECM. Dermal scarring can result in loss of func-
tissue that visibly replaces normal skin after injury. tion, movement restriction, and disfigurement. The
There is a wide spectrum of skin scarring postwound- mechanism of scar formation involves inflammation,
ing, including scarless fetal wound healing, fine-line fibroplasia, formation of granulation tissue, and scar
(normal) scars, stretched (widespread) scars, atrophic maturation. In response to tissue injury, the acute
Scar 265

inflammatory response is followed by the proliferation Hypertrophic scar


of fibroblasts, which are cells responsible for synthesiz-
ing various tissue components, including collagen The pathophysiology of hypertrophic scar formation
and fibrin. During the acute inflammatory phase, circu- involves a constitutively active proliferative phase of
lating progenitor cells migrate to injured tissue. Rapid wound healing.155 The scar tissue has a unique struc-
cellular proliferation occurs, which ultimately results in tural makeup that is highly vascular, with inflammatory
the formation of new blood vessels and epithelium. cells and fibroblasts contributing to an abundant and
Fibroblasts then differentiate into myofibroblasts, which disorganized matrix structure. The net result is that the
are the cells responsible for collagen deposition and original skin defect is replaced by a nonfunctional mass
wound contraction. Scar formation ultimately results of tissue. Beyond these observations, investigations into
from excess accumulation of an unorganized ECM.148,152 the pathophysiology of the disease have been limited by
Sensory nerves, including nonmyelinated C fibers the absence of a practical animal model and have relied
and delta fibers, traverse all cutaneous layers, including upon the use of human pathological specimens. These
the epidermis. These fibers run parallel to capillaries studies are problematic in that such specimens repre-
coursing around follicular complexes. A higher density sent the terminal stages of the scarring process and may
of nerve fiber is associated with scar tissue. Unlike not contain the initiating factors that originally led to
diabetic tissues, hypertrophic scar has excessive the development of the disease. While animal models
neuropeptide activity. In addition, proinflammatory have provided some insight into the genetics and patho-
substance P concentration is greater in the hypertrophic genesis of cutaneous fibrosis, it is unclear how closely
scar samples when compared with normal uninjured the process of hypertrophic scarring in these models
skin. High substance P and low neutral endopepti- resembles that seen in humans. Specifically, it is
dase activity in scar tissue is believed to induce an unknown whether the same factors that initiate hyper-
exuberant neuroinflammatory response contributing to trophic scarring in these species are involved in human
scar formation.153 disease.155

Keloid Regenerative fetal healing


Keloid scarring, also known as keloid disease, is a locally In contrast to adult wound healing, early-gestation fetal
aggressive benign fibroproliferative scar that continu- skin wound healing occurs rapidly, in a regenerative
ally grows beyond the confines of the original wound fashion, and without scar formation. The accelerated
and invades into surrounding healthy skin. Unlike rate of healing, relative lack of an acute inflammatory
hypertrophic scars which stay within the boundaries of response, and an absence of neovascularization distin-
the original wounds and usually regress spontaneously, guish fetal from adult wound healing.156 Scarless wound
keloids grow beyond the boundaries of the original healing has been observed in the fetuses of mice, rats,
wounds and rarely regress. These pathological scars are pigs, monkeys, and humans. Fetal skin heals scarlessly
not only aesthetically displeasing, but can also be both before a certain gestational age, after which point typical
painful and functionally disabling, causing patients scar formation occurs. In humans, scarring of wounds
both physical and psychological distress. There is a begins at approximately 24 weeks of gestation, whereas
strong genetic predisposition to keloid disease. First, in mice scarring of wounds begins on embryonic day
keloids are more common in ethnicities with darker- 18.5 (average gestation period for mice is 20 days). This
pigmented skins and have been reported to be 5–15 transition point, however, is modulated by wound size.
times more prevalent in blacks than in whites. Familial For example, as wound size increases in fetal lambs, the
heritability and prevalence in twins also support the ability to heal scarlessly is lost earlier during gesta-
concept of the genetic susceptibility to keloid scarring. tion.148 In response to tissue injury, the fetal dermis has
The major pathways involved in keloid disease include the ability to regenerate a nondisrupted collagen matrix
apoptosis, mitogen-activated protein kinase, TGF-β, that is identical to that of the original tissue. In addition,
IL-6, and plasminogen activator inhibitor-1.154 dermal structures, such as sebaceous glands and hair
266 • 14 • Wound healing

follicles, form normally after fetal injury. Although the Acknowledgment


exact mechanisms of scarless fetal wound healing are
still unknown, they are thought to be due to differences Supported by NIH RO1 grants GM069589, GM077185,
between the ECM, inflammatory response, cellular NS42617, DK076566, and HL 073087. The authors thank
mediators, differential gene expression, and stem cell Sabyasachi Biswas PhD, Rashmet Reen PhD, and Viren
function in fetal and postnatal wounds.148 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 mechanisms. Redox-based strategies may serve as effective
regeneration. Science. 1997;276:75–81. adjuncts to jump-start healing of chronic wounds. The
The healing of an adult skin wound is a complex process review focuses on the understanding of wound site
requiring the collaborative efforts of many different tissues redox biology and novel insights into the fundamental
and cell lineages. This review discusses the key signals and mechanisms that would help to optimize conditions for
processes that regulate the normal adult cutaneous wound oxygen therapy.
repair. 106. Banerjee J, Chan YC, Sen CK. MicroRNAs in skin and
31. Martin P, Leibovich SJ. Inflammatory cells during wound healing. Physiol Genomics. 2010.
wound repair: the good, the bad and the ugly. Trends MicroRNAs (MiRNAs) are small endogenous RNA
Cell Biol. 2005;15:599–607. molecules about 22 nucleotides in length that are capable
39. Khanna S, Biswas S, Shang Y, et al. Macrophage of posttranscriptional gene regulation by binding to their
dysfunction impairs resolution of inflammation in the target messenger RNAs (mRNAs). This review focuses on
wounds of diabetic mice. PLoS One. 2010;5:e9539. the role of miRNAs in cutaneous biology, the various
methods of microRNA modulation and the therapeutic
53. Carmeliet P. Angiogenesis in life, disease and
opportunities in treatment of skin diseases and wound
medicine. Nature. 2005;438:932–936.
healing.
79. Sen CK. Wound healing essentials: let there be oxygen.
129. Ito M, Liu Y, Yang Z, et al. Stem cells in the hair
Wound Repair Regen. 2009;17:1–18.
follicle bulge contribute to wound repair but not to
86. Roy S, Khanna S, Nallu K, et al. Dermal wound homeostasis of the epidermis. Nat Med. 2005;11:
healing is subject to redox control. Mol Ther. 2006;13: 1351–1354.
211–220.
The discovery of long-lived epithelial stem cells in the bulge
H2O2 has been shown to support wound healing by inducing region of the hair follicle led to the hypothesis that epidermal
VEGF expression in human keratinocytes. This work renewal and epidermal repair after wounding both depend
presents the first in vivo evidence indicating that strategies on these cells. This paper discusses the implications of
to influence the redox environment of the wound site may epithelial stem cells for both gene therapy and developing
have a bearing on healing outcomes. treatments for wounds.
98. Sen CK. The general case for redox control of wound 149. Gurtner GC, Werner S, Barrandon Y, et al. Wound
repair. Wound Repair Regen. 2003;11:431–438. repair and regeneration. Nature. 2008;453:
At very low concentrations, reactive oxygen species may 314–321.
regulate cellular signaling pathways by redox-dependent
References 266.e1

molecular events. Dermatol Surg. 2005;31:674–686;


References discussion 686.
17. Frank S, Kampfer H, Wetzler C, et al. Large induction
1. Mehra R. Historical survey of wound healing. Bull of the chemotactic cytokine RANTES during cutaneous
Indian Inst Hist Med Hyderabad. 2002;32:159–175. wound repair: a regulatory role for nitric oxide in
2. Wangensteen OH. Surgeons and wound management: keratinocyte-derived RANTES expression. Biochem J.
historical aspects. Conn Med. 1975;39:568–574. 2000;347(Pt 1):265–273.
3. Broughton 2nd. G, Janis JE, Attinger CE. A brief 18. Maraganore JM. Thrombin, thrombin inhibitors, and
history of wound care. Plast Reconstr Surg. 2006;117: the arterial thrombotic process. Thromb Haemost.
6S-11S. 1993;70:208–211.
4. Eming SA, Krieg T, Davidson JM. Inflammation in 19. Klebanoff SJ. Myeloperoxidase: friend and foe. J Leukoc
wound repair: molecular and cellular mechanisms. Biol. 2005;77:598–625.
J Invest Dermatol. 2007;127:514–525. 20. Daley JM, Reichner JS, Mahoney EJ, et al. Modulation
5. Sen CK, Gordillo GM, Roy S, et al. Human skin of macrophage phenotype by soluble product(s)
wounds: a major and snowballing threat to public released from neutrophils. J Immunol. 2005;174:
health and the economy. Wound Repair Regen. 2009;17: 2265–2272.
763–771. 21. Peters T, Sindrilaru A, Hinz B, et al. Wound-healing
6. Mackman N, Tilley RE, Key NS. Role of the extrinsic defect of CD18(−/−) mice due to a decrease in TGF-
pathway of blood coagulation in hemostasis and beta1 and myofibroblast differentiation. EMBO J.
thrombosis. Arterioscler Thromb Vasc Biol. 2007;27: 2005;24:3400–3410.
1687–1693. 22. DiPietro LA. Wound healing: the role of the
7. Schecter AD, Spirn B, Rossikhina M, et al. Release of macrophage and other immune cells. Shock.
active tissue factor by human arterial smooth muscle 1995;4:233–240.
cells. Circ Res. 2000;87:126–132. 23. de Vries VC, Noelle RJ. Mast cell mediators in
8. Smyth SS, McEver RP, Weyrich AS, et al. Platelet tolerance. Curr Opin Immunol. 2010;22:643–648.
functions beyond hemostasis. J Thromb Haemost. 24. Abraham SN, St John AL. Mast cell-orchestrated
2009;7:1759–1766. immunity to pathogens. Nat Rev Immunol. 2010;10:
9. Huntington JA. Molecular recognition mechanisms of 440–452.
thrombin. J Thromb Haemost. 2005;3:1861–1872. 25. Trautmann A, Toksoy A, Engelhardt E, et al. Mast cell
10. Midwood KS, Williams LV, Schwarzbauer JE. Tissue involvement in normal human skin wound healing:
repair and the dynamics of the extracellular matrix. expression of monocyte chemoattractant protein-1
Int J Biochem Cell Biol. 2004;36:1031–1037. is correlated with recruitment of mast cells which
11. Sahni A, Odrljin T, Francis CW. Binding of basic synthesize interleukin-4 in vivo. J Pathol. 2000;190:
fibroblast growth factor to fibrinogen and fibrin. J Biol 100–106.
Chem. 1998;273:7554–7559. 26. Shiota N, Nishikori Y, Kakizoe E, et al.
12. Tuan TL, Wu H, Huang EY, et al. Increased Pathophysiological role of skin mast cells in wound
plasminogen activator inhibitor-1 in keloid fibroblasts healing after scald injury: study with mast cell-
may account for their elevated collagen accumulation deficient W/W(V) mice. Int Arch Allergy Immunol.
in fibrin gel cultures. Am J Pathol. 2003;162:1579–1589. 2010;151:80–88.
13. He S, Blomback M, Bark N, et al. The direct thrombin 27. Gallant-Behm CL, Hildebrand KA, Hart DA. The
inhibitors (argatroban, bivalirudin and lepirudin) and mast cell stabilizer ketotifen prevents development
the indirect Xa-inhibitor (danaparoid) increase fibrin of excessive skin wound contraction and fibrosis
network porosity and thus facilitate fibrinolysis. in red Duroc pigs. Wound Repair Regen. 2008;16:
Thromb Haemost. 2010;103:1076–1084. 226–233.
14. Marin V, Montero-Julian FA, Gres S, et al. The IL-6- 28. Noli C, Miolo A. The mast cell in wound healing.
soluble IL-6Ralpha autocrine loop of endothelial Vet Dermatol. 2001;12:303–313.
activation as an intermediate between acute and 29. Eming SA, Werner S, Bugnon P, et al. Accelerated
chronic inflammation: an experimental model wound closure in mice deficient for interleukin-10.
involving thrombin. J Immunol. 2001;167:3435–3442. Am J Pathol. 2007;170:188–202.
15. Delavary BM, van der Veer WM, van Egmond M, et al. 30. Martin P. Wound healing – aiming for perfect skin
Macrophages in skin injury and repair. Immunobiology. regeneration. Science. 1997;276:75–81.
2011. The healing of an adult skin wound is a complex process
16. Baum CL, Arpey CJ. Normal cutaneous wound requiring the collaborative efforts of many different tissues
healing: clinical correlation with cellular and and cell lineages. This review discusses the key signals and
266.e2 • 14 • Wound healing

processes that regulate the normal adult cutaneous wound 46. Gottrup F. Oxygen in wound healing and infection.
repair. World J Surg. 2004;28:312–315.
31. Martin P, Leibovich SJ. Inflammatory cells during 47. Gardner SE, Frantz RA. Wound bioburden and
wound repair: the good, the bad and the ugly. Trends infection-related complications in diabetic foot ulcers.
Cell Biol. 2005;15:599–607. Biol Res Nurs. 2008;10:44–53.
32. Rappolee DA, Mark D, Banda MJ, et al. Wound 48. Flemming HC, Wingender J. The biofilm matrix. Nat
macrophages express TGF-alpha and other growth Rev Microbiol. 2010;8:623–633.
factors in vivo: analysis by mRNA phenotyping. 49. Lohela M, Bry M, Tammela T, et al. VEGFs
Science. 1988;241:708–712. and receptors involved in angiogenesis versus
33. Leibovich SJ, Ross R. The role of the macrophage in lymphangiogenesis. Curr Opin Cell Biol. 2009;21:
wound repair. A study with hydrocortisone and 154–165.
antimacrophage serum. Am J Pathol. 1975;78:71–100. 50. Peters KG, Kontos CD, Lin PC, et al. Functional
34. Goren I, Allmann N, Yogev N, et al. A transgenic significance of Tie2 signaling in the adult vasculature.
mouse model of inducible macrophage depletion: Recent Prog Horm Res. 2004;59:51–71.
effects of diphtheria toxin-driven lysozyme M-specific 51. Sottile J. Regulation of angiogenesis by extracellular
cell lineage ablation on wound inflammatory, matrix. Biochim Biophys Acta. 2004;1654:13–22.
angiogenic, and contractive processes. Am J Pathol. 52. Kubota Y, Kleinman HK, Martin GR, et al. Role of
2009;175:132–147. laminin and basement membrane in the morphological
35. Mirza R, DiPietro LA, Koh TJ. Selective and specific differentiation of human endothelial cells into
macrophage ablation is detrimental to wound healing capillary-like structures. J Cell Biol. 1988;107:1589–1598.
in mice. Am J Pathol. 2009;175:2454–2462. 53. Carmeliet P. Angiogenesis in life, disease and
36. Martinez FO, Helming L, Gordon S. Alternative medicine. Nature. 2005;438:932–936.
activation of macrophages: an immunologic functional 54. Raza A, Franklin MJ, Dudek AZ. Pericytes and vessel
perspective. Annu Rev Immunol. 2009;27:451–483. maturation during tumor angiogenesis and metastasis.
37. Grinberg S, Hasko G, Wu D, et al. Suppression of Am J Hematol. 2010;85:593–598.
PLCbeta2 by endotoxin plays a role in the adenosine 55. Paquet-Fifield S, Schluter H, Li A, et al. A role for
A(2A) receptor-mediated switch of macrophages from pericytes as microenvironmental regulators of human
an inflammatory to an angiogenic phenotype. Am J skin tissue regeneration. J Clin Invest. 2009;119:
Pathol. 2009;175:2439–2453. 2795–2806.
38. Porcheray F, Viaud S, Rimaniol AC, et al. Macrophage 56. Regan MC, Kirk SJ, Wasserkrug HL, et al. The wound
activation switching: an asset for the resolution of environment as a regulator of fibroblast phenotype.
inflammation. Clin Exp Immunol. 2005;142:481–489. J Surg Res. 1991;50:442–448.
39. Khanna S, Biswas S, Shang Y, et al. Macrophage 57. Baur Jr. PS, Parks DH, Hudson JD. Epithelial mediated
dysfunction impairs resolution of inflammation in the wound contraction in experimental wounds – the
wounds of diabetic mice. PLoS One. 2010;5:e9539. purse-string effect. J Trauma. 1984;24:713–720.
40. Spite M, Serhan CN. Novel lipid mediators promote 58. Sen CK, Roy S. Oxygenation state as a driver of
resolution of acute inflammation: impact of aspirin and myofibroblast differentiation and wound contraction:
statins. Circ Res. 2010;107:1170–1184. hypoxia impairs wound closure. J Invest Dermatol.
41. Gordon A, Kozin ED, Keswani SG, et al. Permissive 2010;130:2701–2703.
environment in postnatal wounds induced by 59. Follonier L, Schaub S, Meister JJ, et al. Myofibroblast
adenoviral-mediated overexpression of the anti- communication is controlled by intercellular
inflammatory cytokine interleukin-10 prevents scar mechanical coupling. J Cell Sci. 2008;121:3305–3316.
formation. Wound Repair Regen. 2008;16:70–79. 60. Brauchle M, Angermeyer K, Hubner G, et al. Large
42. Li P, Liu P, Xiong RP, et al. Ski, a modulator of wound induction of keratinocyte growth factor expression by
healing and scar formation in the rat skin and rabbit serum growth factors and pro-inflammatory cytokines
ear. J Pathol. 2011;223:659–671. in cultured fibroblasts. Oncogene. 1994;9:3199–3204.
43. Siddiqui AR, Bernstein JM. Chronic wound infection: 61. Takehara K. Growth regulation of skin fibroblasts.
facts and controversies. Clin Dermatol. 2010;28:519–526. J Dermatol Sci. 2000;24(Suppl 1):S70–S77.
44. White RJ. Wound infection-associated pain. J Wound 62. Sivamani RK, Garcia MS, Isseroff RR. Wound
Care. 2009;18:245–249. re-epithelialization: modulating keratinocyte migration
in wound healing. Front Biosci. 2007;12:2849–2868.
45. Kawai T, Akira S. The role of pattern-recognition
receptors in innate immunity: update on Toll-like 63. Field CK, Kerstein MD. Overview of wound-healing in
receptors. Nat Immunol. 2010;11:373–384. a moist environment. Am J Surg. 1994;167:S2–S6.
References 266.e3

64. Svensjo T, Pomahac B, Yao F, et al. Accelerated healing patients with systemic sclerosis. Circ Res. 2004;95:
of full-thickness skin wounds in a wet environment. 109–116.
Plast Reconstr Surg. 2000;106:602–612. 82. Berthod F, Germain L, Tremblay N, et al. Extracellular
65. Pilcher BK, Dumin JA, Sudbeck BD, et al. The activity matrix deposition by fibroblasts is necessary to
of collagenase-1 is required for keratinocyte migration promote capillary-like tube formation in vitro. J Cell
on a type I collagen matrix. J Cell Biol. 1997;137: Physiol. 2006;207:491–498.
1445–1457. 83. Hopf HW, Gibson JJ, Angeles AP, et al. Hyperoxia and
66. Werner S, Krieg T, Smola H. Keratinocyte-fibroblast angiogenesis. Wound Repair Regen. 2005;13:558–564.
interactions in wound healing. J Invest Dermatol. 84. Hunt TK, Aslam RS, Beckert S, et al. Aerobically
2007;127:998–1008. derived lactate stimulates revascularization and tissue
67. Alizadeh N, Pepper MS, Modarressi A, et al. Persistent repair via redox mechanisms. Antioxid Redox Signal.
ischemia impairs myofibroblast development in 2007;9:1115–1124.
wound granulation tissue: a new model of delayed 85. Myllyla R, Tuderman L, Kivirikko K. Mechanism of the
wound healing. Wound Repair Regen. 2007;15:809–816. prolyl hydroxlase reaction. 2. Kinetic analysis of the
68. Gillespie DL, Kistner B, Glass C, et al. Venous ulcer reaction sequence. Eur J Biochem. 1977;80:349–357.
diagnosis, treatment, and prevention of recurrences. 86. Roy S, Khanna S, Nallu K, et al. Dermal wound
J Vasc Surg. 2010;52:8S–14S. healing is subject to redox control. Mol Ther. 2006;13:
69. Eklof B, Rutherford RB, Bergan JJ, et al. Revision of the 211–220.
CEAP classification for chronic venous disorders: H2O2 has been shown to support wound healing by inducing
consensus statement. J Vasc Surg. 2004;40:1248–1252. VEGF expression in human keratinocytes. This work
70. Hess CT. Arterial ulcer checklist. Adv Skin Wound Care. presents the first in vivo evidence indicating that strategies
2010;23:432. to influence the redox environment of the wound site may
71. Stillman RM. Diabetic ulcers. In: eMedicine: WebMD, have a bearing on healing outcomes.
2010. Available online at: http://emedicine. 87. Linnane AW, Kios M, Vitetta L. The essential
medscape.com/article/460282-overview. requirement for superoxide radical and nitric oxide
72. Simms M. Surgical treatment of the neuroischemic formation for normal physiological function and
foot. J Cardiovasc Surg (Torino). 2009;50:293–311. healthy aging. Mitochondrion. 2007;7:1–5.
73. Karvestedt L, Martensson E, Grill V, et al. Peripheral 88. Sen CK, Packer L. Antioxidant and redox regulation of
sensory neuropathy associates with micro- or gene transcription. Faseb J. 1996;10:709–720.
macroangiopathy: results from a population-based 89. Cheng Y, Song C. Hydrogen peroxide homeostasis
study of type 2 diabetic patients in Sweden. Diabetes and signaling in plant cells. Sci China C Life Sci.
Care. 2009;32:317–322. 2006;49:1–11.
74. Thordarson DB, Shean CJ. Nerve and tendon 90. Gechev TS, Hille J. Hydrogen peroxide as a signal
lacerations about the foot and ankle. J Am Acad Orthop controlling plant programmed cell death. J Cell Biol.
Surg. 2005;13:186–196. 2005;168:17–20.
75. Kottner J, Balzer K, Dassen T, et al. Pressure ulcers: a 91. Jones RD, Morice AH. Hydrogen peroxide – an
critical review of definitions and classifications. Ostomy intracellular signal in the pulmonary circulation:
Wound Manage. 2009;55:22–29. involvement in hypoxic pulmonary vasoconstriction.
76. Soppi E. [Pressure ulcer – occurrence, pathophysiology Pharmacol Ther. 2000;88:153–161.
and prevention.] Duodecim. 2010;126:261–268. 92. Neill S, Desikan R, Hancock J. Hydrogen peroxide
77. Bluestein D, Javaheri A. Pressure ulcers: prevention, signalling. Curr Opin Plant Biol. 2002;5:388–395.
evaluation, and management. Am Fam Physician. 93. Reth M. Hydrogen peroxide as second messenger in
2008;78:1186–1194. lymphocyte activation. Nat Immunol. 2002;3:1129–1134.
78. Olesen CG, de Zee M, Rasmussen J. Missing links in 94. Rhee SG. Redox signaling: hydrogen peroxide as
pressure ulcer research – an interdisciplinary overview. intracellular messenger. Exp Mol Med. 1999;31:53–59.
J Appl Physiol. 2010;108:1458–1464. 95. Rhee SG, Bae YS, Lee SR, et al. Hydrogen peroxide: a
79. Sen CK. Wound healing essentials: let there be oxygen. key messenger that modulates protein phosphorylation
Wound Repair Regen. 2009;17:1–18. through cysteine oxidation. Sci STKE. 2000;2000:PE1.
80. Sen CK, Roy S. Redox signals in wound healing. 96. Sen CK. Antioxidant and redox regulation of cellular
Biochim Biophys Acta. 2008;1780:1348–1361. signaling: introduction. Med Sci Sports Exerc.
81. Distler O, Distler JH, Scheid A, et al. Uncontrolled 2001;33:368–370.
expression of vascular endothelial growth factor and 97. Sen CK. Cellular thiols and redox-regulated signal
its receptors leads to insufficient skin angiogenesis in transduction. Curr Top Cell Regul. 2000;36:1–30.
266.e4 • 14 • Wound healing

98. Sen CK. The general case for redox control of wound the nucleus. Cytokine Growth Factor Rev. 2009;20:
repair. Wound Repair Regen. 2003;11:431–438. 517–521.
At very low concentrations, reactive oxygen species may 112. Sen CK, Gordillo GM, Khanna S, et al. Micromanaging
regulate cellular signaling pathways by redox-dependent vascular biology: tiny microRNAs play big band. J Vasc
mechanisms. Redox-based strategies may serve as effective Res. 2009;46:527–540.
adjuncts to jump-start healing of chronic wounds. The 113. Shilo S, Roy S, Khanna S, et al. Evidence for the
review focuses on the understanding of wound site involvement of miRNA in redox regulated angiogenic
redox biology and novel insights into the fundamental response of human microvascular endothelial cells.
mechanisms that would help to optimize conditions for Arterioscler Thromb Vasc Biol. 2008;28:471–477.
oxygen therapy. 114. Chan YC, Khanna S, Roy S, et al. miR-200b targets
99. Stone JR, Yang S. Hydrogen peroxide: a signaling Ets-1 and is down-regulated by hypoxia to induce
messenger. Antioxid Redox Signal. 2006;8:243–270. angiogenic response of endothelial cells. J Biol Chem.
100. Schaffer MR, Tantry U, Gross SS, et al. Nitric oxide 2011;286:2047–2056.
regulates wound healing. J Surg Res. 1996;63:237–240. 115. Wang S, Olson EN. AngiomiRs – key regulators of
101. Lee RH, Efron D, Tantry U, et al. Nitric oxide in the angiogenesis. Curr Opin Genet Dev. 2009;19:205–211.
healing wound: a time-course study. J Surg Res. 116. Chan SY, Zhang YY, Hemann C, et al. MicroRNA-210
2001;101:104–108. controls mitochondrial metabolism during hypoxia by
102. Schaffer MR, Efron PA, Thornton FJ, et al. Nitric oxide, repressing the iron-sulfur cluster assembly proteins
an autocrine regulator of wound fibroblast synthetic ISCU1/2. Cell Metab. 2009;10:273–284.
function. J Immunol. 1997;158:2375–2381. 117. Loscalzo J. The cellular response to hypoxia: tuning
103. Amadeu TP, Costa AM. Nitric oxide synthesis the system with microRNAs. J Clin Invest. 2010;120:
inhibition alters rat cutaneous wound healing. J Cutan 3815–3817.
Pathol. 2006;33:465–473. 118. Favaro E, Ramachandran A, McCormick R, et al.
104. Schaffer MR, Tantry U, Efron PA, et al. Diabetes- MicroRNA-210 regulates mitochondrial free radical
impaired healing and reduced wound nitric oxide response to hypoxia and Krebs cycle in cancer cells by
synthesis: a possible pathophysiologic correlation. targeting iron sulfur cluster protein ISCU. PLoS One.
Surgery. 1997;121:513–519. 2010;5:e10345.
105. Pasquinelli AE. Molecular biology. Paring miRNAs 119. Tsuchiya S, Fujiwara T, Sato F, et al. MicroRNA-210
through pairing. Science. 2010;328:1494–1495. regulates cancer cell proliferation through targeting
106. Banerjee J, Chan YC, Sen CK. MicroRNAs in skin and fibroblast growth factor receptor-like 1 (FGFRL1). J Biol
wound healing. Physiol Genomics. 2010. Chem. 2011;286:420–428.
MicroRNAs (MiRNAs) are small endogenous RNA 120. Crosby ME, Kulshreshtha R, Ivan M, et al. MicroRNA
molecules about 22 nucleotides in length that are capable of regulation of DNA repair gene expression in hypoxic
posttranscriptional gene regulation by binding to their stress. Cancer Res. 2009;69:1221–1229.
target messenger RNAs (mRNAs). This review focuses on 121. Biswas S, Roy S, Banerjee J, et al. Hypoxia inducible
the role of miRNAs in cutaneous biology, the various microRNA 210 attenuates keratinocyte proliferation
methods of microRNA modulation and the therapeutic and impairs closure in a murine model of ischemic
opportunities in treatment of skin diseases and wound wounds. Proc Natl Acad Sci USA. 2010;107:6976–6981.
healing. 122. Tiscornia G, Izpisua Belmonte JC. MicroRNAs in
107. Roy S, Sen CK. miRNA in innate immune responses: embryonic stem cell function and fate. Genes Dev.
novel players in wound inflammation. Physiol 2010;24:2732–2741.
Genomics. 2011;43:557–565. 123. Li Z, Yang CS, Nakashima K, et al. Small RNA-
108. Recchiuti A, Krishnamoorthy S, Fredman G, et al. mediated regulation of iPS cell generation. EMBO J.
MicroRNAs in resolution of acute inflammation: 2011;30:828–834.
identification of novel resolvin D1-miRNA circuits. 124. Liu C, Teng ZQ, Santistevan NJ, et al. Epigenetic
FASEB J. 2011;25:544–560. regulation of miR-184 by MBD1 governs neural stem
109. Ponomarev ED, Veremeyko T, Barteneva N, et al. cell proliferation and differentiation. Cell Stem Cell.
MicroRNA-124 promotes microglia quiescence and 2010;6:433–444.
suppresses EAE by deactivating macrophages via the 125. Jopling C, Boue S, Izpisua Belmonte JC.
C/EBP-alpha-PU.1 pathway. Nat Med. 2011;17:64–70. Dedifferentiation, transdifferentiation and
110. Asirvatham AJ, Magner WJ, Tomasi TB. miRNA reprogramming: three routes to regeneration. Nat Rev
regulation of cytokine genes. Cytokine. 2009;45:58–69. Mol Cell Biol. 2011;12:79–89.
111. Hata A, Davis BN. Control of microRNA biogenesis by 126. Wu Y, Zhao RC, Tredget EE. Concise review: bone
TGFbeta signaling pathway-A novel role of Smads in marrow-derived stem/progenitor cells in cutaneous
References 266.e5

repair and regeneration. Stem Cells. 2010;28: 139. Ebrahimian TG, Pouzoulet F, Squiban C, et al. Cell
905–915. therapy based on adipose tissue-derived stromal cells
127. Kumamoto T, Shalhevet D, Matsue H, et al. Hair promotes physiological and pathological wound
follicles serve as local reservoirs of skin mast cell healing. Arterioscler Thromb Vasc Biol. 2009;29:
precursors. Blood. 2003;102:1654–1660. 503–510.
128. Cotsarelis G, Sun TT, Lavker RM. Label-retaining cells 140. Kim WS, Park BS, Sung JH, et al. Wound healing
reside in the bulge area of pilosebaceous unit: effect of adipose-derived stem cells: a critical role
implications for follicular stem cells, hair cycle, and of secretory factors on human dermal fibroblasts.
skin carcinogenesis. Cell. 1990;61:1329–1337. J Dermatol Sci. 2007;48:15–24.
129. Ito M, Liu Y, Yang Z, et al. Stem cells in the hair 141. Jaks V, Kasper M, Toftgard R. The hair follicle – a stem
follicle bulge contribute to wound repair but not to cell zoo. Exp Cell Res. 2010;316:1422–1428.
homeostasis of the epidermis. Nat Med. 2005;11: 142. Myung P, Andl T, Ito M. Defining the hair follicle stem
1351–1354. cell (Part I). J Cutan Pathol. 2009;36:1031–1034.
The discovery of long-lived epithelial stem cells in the bulge 143. Myung P, Andl T, Ito M. Defining the hair follicle stem
region of the hair follicle led to the hypothesis that epidermal cell (Part II). J Cutan Pathol. 2009;36:1134–1137.
renewal and epidermal repair after wounding both depend 144. Kavanagh DP, Kalia N. Hematopoietic stem cell
on these cells. This paper discusses the implications of homing to injured tissues. Stem Cell Rev. 2011;7:
epithelial stem cells for both gene therapy and developing 672–682.
treatments for wounds. 145. Yamanaka S. Induction of pluripotent stem cells from
130. Kopp HG, Ramos CA, Rafii S. Contribution of mouse fibroblasts by four transcription factors. Cell
endothelial progenitors and proangiogenic Prolif. 2008;41(Suppl 1):51–56.
hematopoietic cells to vascularization of tumor and 146. Yoshida Y, Yamanaka S. iPS cells: a source of cardiac
ischemic tissue. Curr Opin Hematol. 2006;13:175–181. regeneration. J Mol Cell Cardiol. 2011;50:327–332.
131. Morris LM, Klanke CA, Lang SA, et al. 147. Vercelli S, Ferriero G, Sartorio F, et al. How to assess
Characterization of endothelial progenitor cells postsurgical scars: a review of outcome measures.
mobilization following cutaneous wounding. Wound Disabil Rehabil. 2009;31:2055–2063.
Repair Regen. 2010;18:383–390.
148. Larson BJ, Longaker MT, Lorenz HP. Scarless fetal
132. Fathke C, Wilson L, Hutter J, et al. Contribution of wound healing: a basic science review Plast Reconstr
bone marrow-derived cells to skin: collagen deposition Surg. 2010;126:1172–1180.
and wound repair. Stem Cells. 2004;22:812–822.
149. Gurtner GC, Werner S, Barrandon Y, et al. Wound
133. Ishii GH, Sangai T, Sugiyama K, et al. In vivo repair and regeneration. Nature. 2008;453:314–321.
characterization of bone marrow-derived fibroblasts
recruited into fibrotic lesions. Stem Cells. 2005;23: 150. Singer AJ, Clark RA. Cutaneous wound healing.
699–706. N Engl J Med. 1999;341:738–746.
134. Deng WM, Han Q, Liao LM, et al. Engrafted bone 151. Lovvorn 3rd. HN, Cheung DT, Nimni ME,
marrow-derived Flk-1(+) mesenchymal stem cells et al. Relative distribution and crosslinking
regenerate skin tissue. Tissue Engineering. 2005;11: of collagen distinguish fetal from adult sheep
110–119. wound repair. J Pediatr Surg. 1999;34:
218–223.
135. Mori L, Bellini A, Stacey MA, et al. Fibrocytes
contribute to the myofibroblast population in wounded 152. Ferreira LM, Gragnani A, Furtado F, et al. Control
skin and originate from the bone marrow. Exp Cell Res. of the skin scarring response. An Acad Bras Cienc.
2005;304:81–90. 2009;81:623–629.
136. Badiavas EV, Falanga V. Treatment of chronic wounds 153. Scott JR, Muangman P, Gibran NS. Making sense of
with bone marrow-derived cells. Arch Dermatol. hypertrophic scar: a role for nerves. Wound Repair
2003;139:510–516. Regen. 2007;15(Suppl 1):S27–S31.
137. Rasulov MF, Vasil’chenkov AV, Onishchenko NA, 154. Shih B, Bayat A. Genetics of keloid scarring. Arch
et al. First experience in the use of bone marrow Dermatol Res. 2010;302:319–339.
mesenchymal stem cells for the treatment of a 155. Aarabi S, Longaker MT, Gurtner GC. Hypertrophic
patient with deep skin burns. Bull Exp Biol Med. scar formation following burns and trauma: new
2005;139:141–144. approaches to treatment. PLoS Med. 2007;4:e234.
138. Zuk PA, Zhu M, Ashjian P, et al. Human adipose tissue 156. Stelnicki EJ, Chin GS, Gittes GK, et al. Fetal wound
is a source of multipotent stem cells. Mol Biol Cell. repair: where do we go from here? Semin Pediatr Surg.
2002;13:4279–4295. 1999;8:124–130.

Potrebbero piacerti anche