You are on page 1of 10

COMPREHENSIVE INVITED REVIEW

Keratinocyte Migration and a Hypothetical


New Role for Extracellular Heat Shock Protein
90 Alpha in Orchestrating Skin Wound Healing

David T. Woodley,* Ashley Wysong,


Brittany DeClerck, Mei Chen, and Wei Li
Department of Dermatology, USC Laboratories for Investigative Dermatology, USC/Norris Cancer Center, The Keck
Medical Center and the Los Angeles Greater VA Healthcare System, University of Southern California, Los Angeles,
California.

Significance: The treatment and care of patients with skin wounds are a major
healthcare expenditure. Burn wounds, iatrogenic surgical wounds, venous
stasis dermatitis ulcers, diabetic lower limb ulcers, pressure ulcers, and skin
wounds from peripheral neuropathies are largely treated with only supportive
care. Despite a great deal of research into using growth factors as therapeutic
agents, to date, the field has been disappointing. The only biologic agent that is
Federal Drug Administration (FDA) approved for promoting skin wound
David T. Woodley, MD
healing is recombinant platelet-derived growth factor (PDGF-BB), but its
modest efficacy and expense limit its use clinically. Submitted for publication June 6, 2014. Ac-
Recent Advances: Acute hypoxia induced by the clotting of dermal blood ves- cepted in revised form August 9, 2014.
sels during the wounding of skin is a major stress factor that leads to the *Correspondence: USC/Norris Cancer Center
Topping Tower #3405, 1441 Eastlake Avenue,
re-programming of basal keratinocytes to initiate re-epithelialization. The Los Angeles, CA 90033
laterally migrating keratinocytes secrete extracellular heat shock protein 90 (e-mail: david.woodley@med.usc.edu).
alpha. Heat shock protein 90 alpha (hsp90a) engages low-density lipoprotein
receptor-related protein-1 (LRP-1) cellular receptors and works as an auto-
crine factor to stimulate keratinocyte migration (re-epithelialization) and as a
paracrine factor to stimulate the migration of dermal fibroblasts (fibroplasia)
and microvascular endothelial cells (neo-vascularization). Hypoxia-triggered
extracellular heat shock protein 90 alpha acts as the master regulator of initial
skin wound healing.
Critical Issues: It is not yet known how the engagement of hsp90a with the
LRP-1 receptor leads to increased motility of keratinocytes, fibroblasts, or
microvascular endothelial cells. Understanding the sequence of how an acute
skin wound via hypoxic stress leads to cellular events that ultimately induce
accelerated wound closure provides numerous targets for new wound-healing
therapeutic agents.
Future Directions: Developing data for an investigational new drug (IND)
application to the FDA for a Phase I study using hsp90a in human skin
wounds. Identifying the cellular signaling mechanisms by which hsp90a en-
hances skin cell migration, leading to accelerated wound closure.

SCOPE AND SIGNIFICANCE cess of re-epithelialization, which


This review focuses on the im- closes the wound. Dermal fibroblasts
portance of cell migration in the main (DFs) migration into the wound bed
processes of skin wound healing. is linked to the process of fibroplasia
Epidermal keratinocytes migration and the re-building of a neo-dermis.
is linked to the wound-healing pro- Peri-wound endothelial cells migrat-

ADVANCES IN WOUND CARE, VOLUME 4, NUMBER 4


Copyright ª 2015 by Mary Ann Liebert, Inc. DOI: 10.1089/wound.2014.0566
j 203
204 WOODLEY ET AL.

ing into the wound bed are linked to the process of the migration of DFs (‘‘fibroplasia’’), and the migra-
neoangiogenesis and vascularization of the neo- tion of HDMECs (‘‘angiogenesis/neo-vasculariza-
dermis. tion’’).

TRANSLATIONAL RELEVANCE DISCUSSION


Naturally occurring agents such as heat shock Acute low oxygen and stress re-programs
protein 90 alpha (hsp90a) can jump start the cell basal keratinocytes
migration of three major cells in human skin— Keratinocytes in the unwounded state differen-
keratinocytes, DFs, and microvascular endothelial tiate from the basal keratinocyte layer (Stratum
cells—and when added to full-thickness wounds basale) in which the cells are juxtaposed to the
made in animals (mice and pigs), induces acceler- basement membrane zone (BMZ) located at the
ated wound closure. Therefore, this agent could be dermal–epidermal junction (DEJ). These basal
developed as a wound-healing agent for humans keratinocytes differentiate and migrate upward
who have difficulty in healing skin wounds such as toward the surface of the skin where they eventu-
diabetic lower limb ulcers, pressure ulcers, and ally form the Stratum corneum, the most outer
lower limb ulcers due to venous stasis dermatitis. horny layer of the epidermis and the main protec-
tive barrier of the skin. In this process, the basal
keratinocytes, which are capable of proliferating,
CLINICAL RELEVANCE lose this ability and become nonproliferating
These concepts have great clinical relevance, Stratum spinulosum cells (the squamous cell lay-
because there is a great paucity of consistently effi- er), then Stratum granulosum cells, and, finally,
cacious biologic agents that can be added to non- Stratum corneum cells. This process is called ‘‘ter-
healing wounds and jump start wound closure. minal differentiation,’’ as the cells lose their nuclei
The problem of nonhealing wounds in human beings and become dead bags of cross-linked keratin fibers
is an enormous clinical problem with a very high complexed with filaggrin. When wounded, how-
burden on the healthcare costs of the United States. ever, basal keratinocytes become re-programmed
from cells destined toward terminal differentiation
BACKGROUND into cells that are capable of lateral migration and
continued proliferative potential. What causes this
The healing of human skin wounds is a complex
dramatic switch from a differentiation program to
and highly coordinated biological process.1,2 Of
a migratory re-epithelialization program? We be-
these processes, re-epithelialization, fibroplasia,
lieve that a critical switch is the acute change in
and neo-angiogenesis are three critical processes
oxygen tension that the basal cell keratinocytes
for healing the wound. What they have in common
experience when the skin is wounded. That is,
is the requirement for cell migration. ‘‘Re-epitheli-
when the skin is wounded and the dermal blood
alization’’ is the lateral migration of basal kerati-
vessels are clotted and no longer able to deliver
nocytes within the epidermis across the wound bed
oxygenated hemoglobin via the red blood cells to
and, when successful, closes the wound. The wound
the skin, the basal keratinocytes experience the
bed is initially an amorphous mass of clotted serum
stress of acute hypoxia.
within the rent in the skin. DFs from around
Varghese et al. directly measured the oxygen
the wound should migrate into the wound bed and
tension in human skin wounds under wound
synthesize and secrete new extracellular matrix
dressings and found that it was so low as to be al-
(ECM) molecules, especially type I and type III
most immeasurable.3 O’Toole et al.,4 using in vitro
collagen, to begin building a new dermis. This
keratinocyte migration assays, demonstrated that
process has been called ‘‘fibroplasia.’’
under hypoxic conditions, the keratinocytes ex-
Likewise, the neodermis should become vascu-
hibited increased cellular migration compared
larized if the wound is going to heal and transform
with cells migrating under normoxic conditions.4
into functional human skin. Similar to the peri-
wound fibroblasts, peri-wound human dermal mi- Hypoxia-driven keratinocyte migration
crovascular endothelial cells (HDMECs) should mi- is mediated by hypoxia-inducible factor-1
grate into the wound bed and establish new blood and extracellular heat shock protein alpha
vessels, a process called ‘‘angiogenesis’’ or ‘‘neo-vas- Woodley et al.5 showed that hypoxia-induced ker-
cularization.’’ For these three core wound healing atinocyte migration was mediated by hypoxia-induc-
processes to be accomplished, there needs to be the ible factor-1 (HIF-1), the master regulator of cellular
migration of keratinocytes (‘‘re-epithelialization’’), responses to environmental hypoxia. In vitro experi-
ROLE OF HSP90 ALPHA IN SKIN WOUNDS 205

ments showed that HIF-1 up-regulation led to the membranes of keratinocytes and have the same
extracellular secretion of hsp90a and increased cel- affinity.13 Why TGFa increases keratinocyte mi-
lular migration of keratinocytes, DFs and HDMECs. gration to a greater degree than EGF is not known.
In vivo experiments with murine and porcine wound- Recent evidence in our lab suggests that when
healing models showed that topically applied hsp90a TGFa is added to keratinocyte cultures, it induces
dramatically accelerated wound closure by promoting the secretion of keratinocyte-derived hsp90a,
re-epithelialization.6,7 When human keratinocytes whereas EGF does not (unpublished observation).
(HKs) are stimulated to migrate by hypoxia, they This may be an explanation of why TGFa is a su-
exhibit enhanced lamellipodia-building proteins (ez- perior motogen compared with EGF. To date, TGFa
rin, moesin, and radixin) and increased matrix me- has not been tested in wounds in human beings.
talloproteinases (MMP1, MMP2, and MMP9).4 EGF, however, has been used in one trial in which
These observations fit nicely into the earlier electron acute split thickness wounds were made in 12 in-
microscopy studies of Odland and Ross,8 who dividuals. EGF or the vehicle control was topically
showed that when basal keratinocytes migrate over applied to two identical wounds in the same pa-
a wound bed, their morphology is transformed from tient. In all 12 individuals, wound closure was ad-
cuboidal tombstone-like cells into flattened cells vanced in the wound receiving EGF compared with
with lamellipodia and filopodia at their advancing vehicle.14 This study was the first ‘‘proof of princi-
plasma membrane edge. ple’’ study that a growth factor could enhance the
The finding that migrating HKs express in- closure of a human wound. The closure of human
creased MMPs fits nicely into the concept that ker- wounds is largely mediated by re-epithelialization.
atinocyte migration is a ‘‘ratchet-like’’ process. That Therefore, since EGF enhances keratinocyte mi-
is, migrating keratinocytes secrete ECM macro- gration, it is likely that the mechanism by which
molecules at their advancing plasma membrane exogenously administered EGF demonstrated en-
edge, attach to the deposited ECM, and then secrete hanced closure of human wounds in this study was
MMPs to detach from this ECM and continue mi- by its ability to drive keratinocyte migration in vivo
grating via repeated steps of matrix attachment and and promote re-epithelialization. Nevertheless, it
detachment.9 For many years, the dogma was that is possible that besides enhancing re-epithelial-
only fibroblasts, and not keratinocytes, could syn- ization, topical EGF could have had influences on
thesize and secrete collagenase. It was then shown other processes which are necessary for wound
that HKs synthesized and secreted MMP1, MMP2, healing, but this was not addressed in the paper.
and MMP9. This was missed in earlier studies using When an acute wound is made, the skin at the
functional assays, because the keratinocytes also wound site suddenly experiences being bathed in
make large quantities of tissue inhibitor of me- serum rather than a filtrate of plasma. This sudden
talloproteinases (TIMPs) that nullified the collage- exposure to serum, similar to hypoxia, may present
nases and made them undetectable in functional another signal that the basal keratinocytes should
collagenase assays.10,11 Petersen et al.,12 in fact, abandon the mode to differentiate and embrace a
demonstrated that when HKs are stimulated to migratory mode. We noted in our keratinocyte
migrate, they increase their synthesis and secretion migration assays that the presence of serum, but
of MMPs, again in accordance with the ratchet not plasma, would induce increased keratinocyte
theory of keratinocyte motility.12 migration.15 Serum, compared with plasma, has
much higher levels of TGFa, which is the major
Growth factors can be mitogens or ‘‘motogens’’ ingredient in human serum that is responsible for
(i.e., agents that make cells migrate) enhancing keratinocyte migration.16 The notion
Other biological processes, in addition to the that fresh serum with its high concentration of
acute hypoxia signal, also influence HK migration. TGFa can promote keratinocyte migration and re-
Certain growth factors are not only ‘‘mitogens’’ that epithelialization may explain a clinical maneuver
drive cell division but also ‘‘motogens’’ which drive which has been used for years in dermatology to
cell migration by a different mechanism. Both ‘‘jump start’’ a nonhealing skin wound—namely to
epidermal growth factor (EGF) and transforming excise the wound, create a new fresh wound, and
growth factor alpha (TGFa) enhance keratinocyte flood the area with fresh serum.
migration. These two factors share the same re-
ceptor on the keratinocyte, the EGF receptor, but Transforming growth factor beta
TGFa is a more powerful keratinocyte ‘‘motogen’’ is a double-edged sword in wound healing
than EGF despite the fact that by Scatchard plots, Transforming growth factor beta (TGFb) is rich
EGF and TGFa bind equally well to the plasma in the wound bed of a healing skin wound. It is
206 WOODLEY ET AL.

known that TGFb induces local fibroblasts to wound might inhibit re-epithelialization at some
increase their synthesis and secretion of ECM level via its impact on keratinocyte proliferation.
molecules such as collagen I and III, while it also Once keratinocytes begin to migrate horizontally
decreases the fibroblast’s expression of MMPs. over the wound bed (within hours of wounding), the
Together, the TGFb influences are pro-wound keratinocytes behind the migratory cells lose con-
matrix and are thought to help the immature tact inhibition and begin to proliferate and subse-
wound bed transform into a neo-dermis. Sarret quently add to the migratory leading edge of
et al. showed that the presence of TGFb markedly keratinocytes. Nevertheless, the proliferative cells
inhibits the proliferative potential of HKs, but that are behind the migratory leading edge cells and
keratinocyte migration is not inhibited by TGFb.17 may not even be in contact geographically with
Among the three mammalian TGFb family TGFb in the wound bed.
members, TGFb3 appears to play a critical role in
Connective tissue macromolecules influence
the initial phase of wound closure, by coordinating
basal keratinocyte migration
the time of epidermal and dermal cell migration.18
Other major influences on HK migration are the
This finding is summarized in Figure 1, in which
ECM molecules to which the cells are juxtaposed.
TGFb3 plays a ‘‘traffic control’’ role to halt DF and
For example, a substratum of type I dermal colla-
endothelial cell migration until keratinocyte mi-
gen enhances keratinocyte migration, while a
gration occurs and the re-epithelialization process
substratum of laminin 1 (aka laminin 111), a major
is complete. How these observations translate
matrix glycoprotein in the BMZ within the DEJ,
in vivo into a healing skin wound is not clear and
inhibits keratinocyte migration.19 The two most
requires further study. Re-epithelialization is
predominant laminin isoforms within the DEJ
thought to be accomplished by both keratinocyte
of human skin are laminin 5 (aka laminin 332)
migration and keratinocyte proliferation, so in
and laminin 10 (aka 511). In the setting of wound
theory, the presence of high levels of TGFb in the
healing, secreted laminin 332 is proteolytically
processed into a pro-motility smaller molecule.
This processing may expose a cryptic site that
promotes cell migration or it may release an EGF-
like fragment which can promote both cell prolif-
eration and migration.20,21
Keratinocytes engage ECM molecules via in-
tegrin receptors. When juxtaposed on type I colla-
gen, their collagen-driven migration is mediated by
the a2b1 integrin.22 When juxtaposed to a matrix of
fibronectin, their migration is mediated by the a5b1
integrin.23 When there is migration on vitronectin,
an ECM expressed early in healing skin wounds,
the keratinocytes use avb5 integrin.24
Migrating keratinocytes regardless of the sub-
strate to which they are apposed build and dismantle
sequentially focal adhesions along their journey.
When they engage their ECM via the appropriate
integrin, focal adhesion kinase (FAK125) is phos-
Figure 1. A schematic representation of how plasma/serum/plasma phorylated and activated, which allows disassembly
transitions coordinate the orderly skin cell migration during wound healing.
Three major types of skin cells—HKs, DFs, and HDMECs—are shown here, as of focal adhesions to enable the ratchet like traction
indicated by different colors. The dermal cells express higher levels of TbRII of the migrating cells to continue.25 Taken together,
(symbolized by Y) than the epidermal cells. Therefore, the dermal cells are the ECM-driven haptotaxis and the serum factor-
sensitive to the anti-promotility effect of TGFb3 (red stars), whose concen- driven chemotaxis determine the initial and optimal
tration increases after the transition from plasma to serum in the wound bed.
Contributions by other cell types and matrix components to skin wound healing migration of keratinocytes in early wound healing.
are omitted for the sake of simplicity. The relative numbers and proportions of
the various types of cells do not quantitatively reflect those in real human skin.
Extracellular heat shock protein 90 alpha
BM, basement membrane; DF, dermal fibroblast; HDMECs, human dermal plays a central role in skin wound healing
microvascular endothelial cells; HK, human keratinocyte; TGFb3, transforming As previously mentioned, within the last several
growth factor beta 3. (Taken from Brandyopadhyay et al., JCB, 172:1093–1105, years, our laboratory has discovered another factor
2006 with permission.) To see this illustration in color, the reader is referred to
the web version of this article at www.liebertpub.com/wound
that un-expectedly drives keratinocyte migration as
an autocrine factor secreted from the basal kerati-
ROLE OF HSP90 ALPHA IN SKIN WOUNDS 207

nocytes in response to the stress of acute hypoxia


induced by wounding the skin. We noted in our ker-
atinocyte migration assays that the cells exhibited
increased migration when the oxygen tension in the
cell culture incubator was decreased. We then ex-
amined the conditioned medium under normoxic and
hypoxic conditions and noted four new and different
protein bands in the conditioned medium of hypoxic
cells. Using mass spectrometry, we identified one of
these bands as the secreted, extracellular form of
heat shock protein 90 alpha (xhsp90a).
Figure 2. A model for hypoxia-driven keratinocyte migration and re-epi-
The intracellular form of this protein has been thelialization. Hypoxia drives hsp90a secretion. This is mediated by HIF-1a.
known for many years as a chaperone protein with Secreted hsp90a then binds to the LRP-1 receptor and promotes migration
more than 100 client proteins that are shepherded of HKs. Likewise, it can also bind to nearby, peri-wound DFs and HDMECs,
through the cytosol, endoplasmic reticulum, and which also have the LRP-1 receptor on their cell surface and induce cell
motility and fibroplasia and neo-vascularization, respectively. HIF-1a, hyp-
Golgi apparatus. In fact, a great deal of research has oxia-inducible factor-1 alpha; hsp90a, heat shock protein 90 alpha; LRP-1,
been done on the intracellular form of hsp90a, a low-density lipoprotein receptor-related protein-1. (Taken from Woodley
molecule considered a reasonable target for anti- et al., JCS, 122:1495–1498, 2009 with permission.)
cancer therapy, as it regulates both cell proliferation
and inflammation. Adding the xhsp90a to kerati-
nocyte migration assays under normoxic conditions naling pathway or the network of pathways induced
induced the same increased keratinocyte migration by xhsp90a could be the same or completely different
observed in assays done under hypoxic conditions. in HKs, DFs, and HDMECs.
Further studies showed that hsp90a secretion was Perhaps even more interesting than xhsp90a’s
mediated by HIF-1a, which becomes rapidly up- role as an ‘‘autocrine motogen’’ for HKs is that it
regulated in HKs stressed by hypoxia (Fig. 1).26 also likely plays a role in both wound-healing fi-
Under the conditions of skin wounding, all of the broplasia and neo-angiogenesis by its ability also to
dermal blood vessels are clotted and the basal promote the cell migration of DFs and HDMECs
keratinocytes in the avascular epidermis experi- once it is secreted into the extracellular compart-
ence acute hypoxia. This stress invokes an up- ment. In the extracellular space, the keratinocyte-
regulation of HIF-1a and the subsequent secretion derived xhsp90a also acts as a ‘‘paracrine motogen’’
of xhsp90a by the exosomal protein trafficking for DFs and HDMECs in the peri-wound environ-
pathway rather than by conventional endoplasmic ment. Similar to keratinocytes, fibroblasts and
reticulum/Golgi pathway.5,26–28 Once secreted by HDMECs also have the same LRP-1 receptor on
the HK in response to hypoxia, xhsp90a acts as an their cell surfaces. Within the wound bed, kerati-
‘‘autocrine motogen’’ to stimulate migration in the nocyte-derived xhsp90a binds to the LRP-1 recep-
cell that secreted it, namely the keratinocyte. It tors on both fibroblasts and HDMECs and induces
does this by binding to the lipoprotein receptor low- cellular motility. The increased motility of peri-
density lipoprotein receptor-related protein-1 lesional DFs likely leads to the ingress of these cells
(LRP-1) on the cell surface of the keratinocyte.5 into the wound clot/wound bed, where they can
This mechanism is schematically shown in Figure 2. then synthesize and secrete new dermal matrices,
LRP-1 receptors are on many different types of particularly type III collagen and type I collagen.
cells, including HKs, DFs, and HDMECs. When it Likewise, the xhsp90a-induced increased migra-
is secreted by hypoxic, migrating keratinocytes tion of peri-lesional HDMECs likely leads to the
involved in the process of re-epithelialization into ingress of these cells in to the wound clot/wound
the extracellular spaces of the wound bed, it can bed, where they can form new dermal vascular
engage keratinocytes, DFs, and HDMECs via the tubes that eventually vascularize the neo-dermis.29
same receptor.5 What is not known is how the en- Growth factors, such as platelet-derived growth
gagement of this receptor by xhsp90a induces cel- factor (PDGF), EGF, and TGFa, tend to be rela-
lular signaling that leads to enhanced cellular tively small molecules. When exogenously added to
motility of HKs, HFs, and HDMECs. In addition, skin wounds, these small molecules face a wound
there are at least four distinct subdomains of the bed that is rich in proteolytic enzymes and capable
LRP-1 receptor and various ligands have affinity for of degrading them. That is likely one reason why
different LRP-1 subdomains, leading to different bi- PDGF-BB (Becaplermin), the only Federal Drug
ologic behaviors of the cells. Hypothetically, the sig- Administration (FDA)-approved growth factor in-
208 WOODLEY ET AL.

dicated for human skin wounds, needs to be added nonmigratory (Step 1). Within hours after skin in-
to the wounds every day in order to have efficacy. In jury, basal keratinocytes begin to migrate laterally
addition to proteolytic enzymes in the wound bed, across the wound bed, which is basically a serum
the wound bed is also rich in TGFb isoforms. TGFb clot. This initial keratinocyte migration is likely in-
isoforms serve to induce ECM in the neodermis by duced by hypoxia-driven hsp90a autocrine signaling
regulating fibroblast synthesis and secretion of plus serum-derived TGFa. As the keratinocytes
collagens and glycoproteins, as well as MMPs and migrate over the wound bed and become engaged
TIMPs. Nevertheless, these TGFb isoforms also in the process of re-epithelialization, they secrete
nullify the activities of growth factors in the wound hsp90a into the extracellular space of the wound
bed.18 Interestingly, xhsp90a in the wound bed is not bed. During this initial early postwound period,
nullified or even inhibited by the presence of TGFb.7 human dermal fibroblasts and HDMECs at the
Therefore, it has the ability to continue to be biolog- wound edge are unable to immediately move into
ically active and stimulate keratinocyte migration the wound bed due to the presence of TGFb3 (Step
(re-epithelialization), DF migration (fibroplasia), 2). Once the secreted hsp90a reaches a threshold
and HDMEC migration (neo-vascularization). These concentration of 10–30 lM, it triggers the DFs and
three salient wound-healing skin cells under the HDMECs to migrate into the wound bed from the
regulation of xhsp90a secreted by the hypoxic, mi- surrounding wound edge even in the presence of
grating keratinocytes is a clear instance in which TGFb3 (Step 3) and initiate the processes of fi-
there are coordinated epidermal–mesenchymal in- broplasia and neo-vascularization. When the mi-
teractions leading to the complex process of closing grating keratinocytes have completely re-surfaced
skin wounds. the wound, the process of re-epithelialization is
A summary of the actions of the secreted form of finished. The peri-wound fibroblasts that have mi-
hsp90a action in wound healing is shown in Figure grated into the wound bed begin to lay down new
3. Briefly, before injury, the keratinocytes, fibro- ECM and create a neo-dermis and re-model the
blasts, and HMECs in intact, un-wounded skin are wound. Peri-wound HDMECs that have migrated

Figure 3. Secreted hsp90, but not conventional growth factors, promotes re-epithelialization and recruits DFs and HDMECs into the wound bed. Step 1 is un-wounded,
intact skin with relatively low levels of TGFb and minimal cell migration. In this state, the basal keratinocytes are programmed to move upward and differentiate. Step 2 is
during initial wounding of the skin, which releases TGFb from several sources and the presence of high levels of TGFb in the wound bed inhibits growth factor function
while promoting extracellular matrix deposition into the wound bed. Step 3 is during early wound healing when basal keratinocytes migrate over the wound bed and
secrete hsp90a. The secreted hsp90a reaches a threshold concentration in the wound bed, which initiates the inward migration of DFs and HDMECs into the wound bed
and begins development of a neodermis. Step 4 is when the migrating HKs have completed the re-epithelialization process and the DFs and HDMECs, which are now
resident cells of the neodermis, remodel the wound and build new blood vessels, respectively. HDF, human dermal fibroblast. (Taken from Cheng et al., JCI, 121:4348–4361,
2011 with permission.) To see this illustration in color, the reader is referred to the web version of this article at www.liebertpub.com/wound
ROLE OF HSP90 ALPHA IN SKIN WOUNDS 209

into the wound bed/neo-dermis begin to re-form diabetic mice, pigs, or diabetic pigs, accelerates the
dermal blood vessels (Step 4). Implicit in the sce- closure of the wounds when compared with vehicle
nario just described is that the initiating event controls via enhanced re-epithelialization7 (Fig. 5).
which jump starts the wound healing program is Unlike canonical growth factors that are functionally
acute hypoxia, Mother Nature’s signal to the skin inhibited by elevated glucose, xhsp90a maintains its
cells that the programs of homeostasis and terminal biological activity in the presence of elevated glucose.
differentiation should be turned off and a new Diabetic skin wounds are often slow to heal, but there
wound-healing program with elements of a reca- are also nondiabetic slow-to-heal skin wounds such
pitulation of gestation should be immediately initi- as decubitus pressure ulcers and chronic stasis der-
ated. We believe that injury-induced secretion of matitis lower limb ulcers. Due to a paucity of animal
hsp90a is the predominant factor and driving force models that reflect nondiabetic chronic wounds, we
which leads this new hypoxia-driven wound-healing do not yet have data showing that xhsp90a or its F-5
program rather than conventional growth factors. fragment would enhance the closure of chronic
After initial wound closure, the dermal remodeling wounds due to these etiologies.
and neovascularization processes take many The exact timing of the various processes and
months to complete. Many other factors, including events leading to the successful healing of a skin
conventional growth factors, may play roles in the wound is not clear and often appears overlapping.
later events of wound healing, when the TGFb levels Wound contraction contributes as well to the
decrease healing of skin wounds, particularly in loose skin
The sub-domain of xhsp90a responsible for the animals such as mice and rats. Humans, similar to
enhanced motility of keratinocytes, fibroblasts, and pigs, are tight skin animals, and human skin
HDMECs is called the F-5 fragment. As shown in wounds also exhibit wound contraction, but to a
Figure 4, the F-5 fragment is distinct from the sub- lesser degree than rodents. We do not have data
domain within the molecule that binds to ATP and showing that xhsp90a has an influence on wound
causes hydrolysis of ATP and is involved in the in- contraction. We do have data showing that xhsp90a
tracellular chaperone functions of hsp90a. The sub- promotes the migration of keratinocytes, DFs, and
domain of xhsp90a that acts as an autocrine and
paracrine factor within the wound bed to enhance
skin cell motility resides within a small 115 amino
acid distinct F-5 fragment of xhsp90a. Moreover,
xhsp90a or F-5 fragment, when added in vivo to
standardized skin wounds made on normal mice,

Figure 4. F-5 fragment carries out the extracellular functions of hsp90a. A Figure 5. Topical hsp90a promotes wound closure. Topical recombinant
schematic representation of seven human hsp90a protein/peptides (wild hsp90a (0.45 mM) accelerated skin wound closure in diabetic (db/db) mice
type and mutants). The 115-amino-acid fragment, called F-5, is sufficient for from 35 to 18 days with a single application. Becaplermin (Regranex) also
the extracellular functions of hsp90a, specifically its function to drive the accelerated wound closure from 35 to 30 days with one application. (Taken
migration of HKs, DFs, and HDMECs. (Taken from Cheng et al., JCI, 121:4348– with permission from Cheng et al., JCI, 121:4348–4361, 2011.) To see this
4361, 2011 with permission.) To see this illustration in color, the reader is illustration in color, the reader is referred to the web version of this article
referred to the web version of this article at www.liebertpub.com/wound at www.liebertpub.com/wound
210 WOODLEY ET AL.

HDMECs using in vitro cellular motility assays,


TAKE HOME MESSAGE
and we have in vivo data showing that in our
Keratinocyte-derived extracellular heat shock protein 90
animal wound models the presence of exogenous
alpha promotes the cellular migration of keratinocytes as an
excess of promotes re-epithelialization. We do autocrine ‘‘motogen’’ and dermal fibroblasts and micro-
know that when an acute skin wound is made, vascular endothelial cells as a paracrine ‘‘motogen’’ in the
there is a delay of several hours before basal wound bed under the conditions of acute hypoxia from
keratocytes begin to move laterally across the wounding, and it drives three essential skin wound healing
wound bed. It is possible that the dual influences processes–namely, re-epithelialization, fibroplasia and neo-
of flooding the keratinocytes with serum TGFa vascularization.
plus the acute stress of hypoxia jump start this
process. Then, as the migrating neo-epithelium
begins to march across the wound bed, it deposits ACKNOWLEDGMENTS
xhsp90a into the wound bed, which, in turns, AND FUNDING SOURCES
initiates fibroplasia and neoangiogenesis by pro-
This work was supported by NIH RO1 AR47981
moting the migration of fibroblasts and HDMECs.
to Mei Chen; RC4AR060535 and RO1 AR33625 to
Although speculative, this scenario would, in
Mei Chen and David T. Woodley; RO1 GM066193
broad strokes, account for the fact that re-epi-
and RO1 GM067100 to Wei Li; and VA Merit
thelialization appears to begin slightly before fi-
Award to David T. Woodley.
broplasia and angiogenesis. There should be some
built-in inefficiency in this process, however,
possibly due to the inflammatory response and AUTHOR DISCLOSURE
engulfment of the wound bed with proteolytic AND GHOSTWRITING
enzymes, because we can dramatically accelerate No competing financial interests exist. The con-
the closure of skin wounds in animals by provid- tent of this article was expressly written by the
ing an excessive amount of xhsp90a to healing authors listed. No ghostwriters were used to write
wounds with just one application. Our laboratory this article.
is now attempting to define the optimal doses and
application frequency of xhsp90a with the hope
that we can bring this novel wound-healing agent ABOUT THE AUTHORS
from the laboratory bench to the bedside of pa- David T. Woodley, MD, completed his under-
tients with nonhealing skin wounds. graduate degree in English Literature at Wa-
shington University in St. Louis and his medical
school education at the University of Missouri in
SUMMARY Columbia, Missouri. He completed his dermatology
For skin wounds to heal, the migration of the residency training at the University of North Car-
three main types of skin cells—keratinocytes, fi- olina in Chapel Hill, North Carolina. He then
broblasts, and endothelial cells—should occur. completed a Dermatology Research Fellowship in
Without the migration of these cells, the the laboratory of Dr. Michel Prunieras at the
main processes of wound healing—re-epithelial- Rothschild Foundation and University of Paris in
ization, fibroplasia, and neovascularization—will Paris, France. He served as an Expert Investigator
not occur and the wound will not heal. While ca- at the National Institutes of Health for 3 years and
nonical growth factors from the serum and cells in then returned to Chapel Hill as an Assistant Pro-
the wound bed play roles in wound healing, the fessor of Dermatology. He left Chapel Hill in 1989
master regulator of the critical processes of cell to be Professor and Associate Chair of the Depart-
migration is likely xhsp90a, which acts as an au- ment of Dermatology at Stanford University. In
tocrine migration factor for keratinocytes and a 1992, he was appointed the Walter Hamlin Pro-
paracrine migration factor for DFs and microvas- fessor and Chair of Dermatology at Northwestern
cular endothelial cells. Adding excessive xhsp90a University. In 1999, he joined the medical faculty
to healing wounds in normal mice, diabetic mice, at the Keck School of Medicine of the University of
and pigs dramatically accelerates wound closure, Southern California (USC) and in 2004, he as-
suggesting that this agent could be developed for sumed the position as the Founding Chair of the
clinical use in human beings for patients with USC Department of Dermatology. His current title
difficult-to-heal skin wounds such as diabetic is Professor and Emeritus Founding Chair of the
lower limb ulcers, stasis dermatitis ulcers, and USC Department of Dermatology. Dr. Woodley is
pressure ulcers. the author of more than 200 original articles and is
ROLE OF HSP90 ALPHA IN SKIN WOUNDS 211

a clinician-investigator with continuous NIH fund- NIH RO1 grants, one Challenge Grant, and one VA
ing since 1982. He is the Co-Editor of a book titled Merit Review Grant support the USC Laboratory
The Biology of Skin with Dr. Ruth Freinkel and for Investigative Dermatology.
serves as an Associate Editor of The Journal of the Brittany DeClerck, MD, is an Assistant Pro-
American Academy of Dermatology, The Archives of fessor in the USC Department of Dermatology
Dermatology, and Clinical and Experimental Der- and in the USC Department of Pathology. She is
matology and Dermatology. Dr. Woodley’s scientific a board-certified dermatologist and a board-
interests include type VII collagen, keratinocyte certified dermatopathologist. She is interested in
motility, wound healing, keratinocyte-derived col- autoimmune bullous diseases and skin wound
lagenases, autoimmune bullous diseases, and he- healing.
reditary dystrophic epidermolysis bullosa. He has Ashley Wysong, MD, is an Assistant Professor
served on numerous American Academy of Derma- in the USC Department of Dermatology and is
tology committees, the Board of Directors of the Director of the Cutaneous Surgery Division and
Society for Investigative Dermatology, the Board of Mohs Micrographic Skin Cancer Program at USC.
Directors of the California Dermatology Society, She is a board-certified dermatologist and a fel-
and the Board of Directors of the LA Metropolitan lowship-trained Mohs micrographic skin cancer
Dermatology Society. He is the current past Pre- surgeon. She is interested in cutaneous surgery
sident of the LA Metropolitan Dermatology Society and skin wound healing.
and the President-Elect of the California Derma- Wei Li, PhD, is a tenured Professor in the USC
tology Society. He has been elected to the American Department of Dermatology. He is a NIH-funded
Society of Clinical Investigation (ASCI), the Amer- Principal Investigator on several grants pertaining
ican Dermatological Association (ADA), and the to wound healing, diabetes and extracellular
Association of American Physicians (AAP). Four hsp90a.

REFERENCES
1. Singer AJ, Clark RAF. Cutaneous wound healing. 8. Odland G, Ross R. Human wound repair. I. Epi- epidermal growth factor. N Engl J Med 1989;
N Engl J Med 1999;341:738–746. dermal regeneration. J Cell Biol 1968;39:135–151. 32:76–79.
2. Schreml S, Szeimies RM, Prantl L, Landthaler M, 9. Woodley DT, O’Keefe EJ, Prunieras M. Wound 15. Henry G, Li W, Fan JH, Kasahara N, Woodley DT.
Babilas P. Wound healing in the 21st century. J healing: a model for cell-matrix interactions. J Am Human serum (but not plasma) promotes human
Am Acad Dermatol 2010;63:866–881. Acad Dermatol 1985;12:420–433. keratinocyte migration, a critical process for
wound re-epithelialization. Lancet 2003;361:574–
3. Varghese MC, Balin AK, Carter DM, Caldwell D. 10. Woodley DT, Kelebec T, Banes AJ, Link W, Pru-
576.
Local environment of chronic wounds under syn- nieras M, Liotta LA. Adult human keratinocytes
thetic dressings. Arch Dermatol 1986;122:52–57. migrating over nonviable dermal collagen produce 16. Li Y, Chen M, Li W, Woodley DT. Transforming
collagenolytic enzymes that degrade type I and growth factor alpha elevated in human serum is
4. O’Toole EA, Marinkovich MP, Peavy C, et al. Hy-
type IV collagen. J Invest Dermatol 1986;86: the major pro-motility factor for human keratino-
poxia increases human keratinocyte motility on
418–423. cytes. J Invest Dermatol 2006;126:2096–2105.
connective tissue. J Clin Invest 1997;100:2881–
2891. 11. Petersen MJ, Woodley DT, Stricklin GP, O’Keefe 17. Sarret Y, Woodley DT, Grigsby, Wynn K, O’Keefe
EJ. Production of collagenase by cultured human EJ. Human keratinocyte locomotion: the effect of
5. Woodley DT, Li Y, Fan JF, Cheng F, Chen M, Li W.
keratinocytes. J Biol Chem 1987;262:835–840. selected cytokines. J Invest Dermatol 1992;98:
Hypoxia triggers an hsp90a–CD91 autocrine loop
12–16.
to enhance cell motility under serum factor pov- 12. Petersen MJ, Woodley DT, Stricklin GP, O’Keefe
erty. J Cell Sci 2009;122:1495–1498). EJ. Enhanced synthesis of collagenase by human 18. Brandyopadhyay B, Fan J, Guan S, et al. A ‘‘traffic
keratinocytes cultured on type I or type IV colla- control’’ roles for TGFb3:orchestrating dermal and
6. Li W, Li Y, Guan SX, et al. Extracellular hsp90a:
gen, J Invest Dermatol 1990;94:341–346. epidermal cell motility during wound healing. J
linking hypoxia signaling to promote human skin
Cell Biol 2006;172:1093–1105.
cell migration in vitro and wound healing in vivo. 13. Cha D, O’Brian P, Woodley DT, Hudson L. En-
EMBO J 2007;26:1221–1233. hanced modulation of keratinocyte motility by 19. Woodley DT, Bachmann PM, O’Keefe EJ. Laminin
transforming growth factor-alpha (TFG-alpha) rel- inhibits human keratinocyte migration. J Cell
7. Cheng CF, Sahu D, Tsen F, et al. A fragment of
ative to epidermal growth factor (EGF). J Invest Physiol 1988;136:140–146.
secreted hsp90a carries properties that enable
Dermatol 1996;106:590–597.
it to accelerate effectively both acute and dia- 20. Sugawara K, Tsuruta D, Ishii M, Jones JCR, Ko-
betic wounds in mice. J Clin Invest 2011;121: 14. Brown GL, Nanney LB, Griffen J, et al. Enhance- bayashi H. Laminin 332 and - 511 in skin. Exp
4348–4361. ment of wound healing by topical treatment with Dermatol 2008;17:473–480.
212 WOODLEY ET AL.

21. Sehgal BU, DeBiase PJ, Matzno S, et al. increased in human keratinocytes induced to mi-
Integrin beta 4 regulates migratory behavior grate by extracellular matrices. J Cell Physiol Abbreviations and Acronyms
of keratinocytes by determining laminin 332 2001;188:24–32. BMZ ¼ basement membrane zone
organization. J Biol Chem 2006;281:35487– ECM ¼ extracellular matrix
26. Li Y, Guan SX, Fan JF, et al. Extracellular
35498. DEJ ¼ dermal–epidermal junction
hsp90a mediates hypoxia signaling to pro-
DF ¼ dermal fibroblast
22. Kim JP, Zhang K, Kramer RH, Schall TJ, Woodley mote human skin cell migration in vitro and
EGF ¼ epidermal growth factor
DT. Integrin receptors and RGD sequences in wound healing in vivo. EMBO J 2007;26:
FAK ¼ focal adhesion kinase
human keratinocyte migration: unique anti-mi- 1221–1233.
FDA ¼ Federal Drug Administration
gratory function of alpha 3 beta 1. J Invest Der- 27. Cheng CF, Fedesco M, Guan SG, et al. TGF (alpha)- HDF ¼ human dermal fibroblast
matol 1992;98:764–770. stimulated secretion of HSP 90 (alpha): Using HDMECs ¼ human dermal microvascular
LRP-1/CD91 receptor to promote human skin endothelial cells
23. Kim JP, Chen JD, Woodley DT. Mechanisms of
cell migration against TGF (beta)-rich environment HIF-1a ¼ hypoxia-inducible factor-1 alpha
human keratinocyte migration of fibronectin: un-
in wound healing. Mol Cell Biol 2008;28:334– HK ¼ human keratinocyte
ique roles of RGD site and integrins. J Cell Physiol
3358. hsp90a ¼ heat shock protein 90 alpha
1992;151:443–450.
IND ¼ investigational new drug
28. Li W, Li Y, Guan S, et al. Extracellular heat shock
24. Kim JP, Zhang K, Chen JD, Kramer RH, Woodley LRP-1 ¼ low density lipoprotein receptor-
protein-90 alpha mediates hypoxia stress-induced
DT. Vitronectin-driven human keratinocyte lo- related protein-1
motility of primary humnan dermal fibroblasts.
comotion is mediated by the alpha v beta 5 MMP ¼ matrix metalloproteinase
EMBO J 2007;26:1221–1233.
integrin receptor. J Biol Chem 1994;269:26926– PDGF ¼ platelet-derived growth factor
26932. 29. Li W, Sahu D, Tsen F. Secreted heat shock TGFa ¼ transforming growth factor alpha
protein 90 (hsp90) in wound healing and TGFb ¼ transforming growth factor beta
25. Yurko MA, O’Toole EA, Woodley DT. Phosphor- cancer. Biochem Biophys Acta 2012;1823:730– TIMP ¼ tissue inhibitor of metalloproteinase
ylation of focal adhesion kinase [pp125(FAK)] is 741. xhsp90a ¼ extracellular heat shock protein
90 alpha

You might also like