You are on page 1of 18

PROGRESS REPORT

Nitric Oxide www.advhealthmat.de

Nitric Oxide Therapy for Diabetic Wound Healing


Maggie J. Malone-Povolny, Sara E. Maloney, and Mark H. Schoenfisch*

Nitric oxide (NO), the endogenous


Nitric oxide (NO) represents a potential wound therapeutic agent due to regulator of inflammation and an antibac-
its ability to regulate inflammation and eradicate bacterial infections. Two terial agent with no demonstrated resist-
broad strategies exist to utilize NO for wound healing; liberating NO from ance, has become an attractive candidate
for wound healing therapy.[7] The ability of
endogenous reservoirs, and supplementing NO from exogenous sources.
NO to serve as a single agent wound ther-
This progress report examines the efficacy of a variety of NO-based methods apeutic or in conjunction with established
to improve wound outcomes, with particular attention given to diabetes- wound therapy protocols (e.g., wound
associated chronic wounds. dressings, HBOT) underlines its versa-
tility and merits further research. Herein,
we review the recent developments of NO-
1. Introduction based therapies for wound healing, with an emphasis on the
role of these therapies regarding diabetic wound healing.
Chronic wounds are wounds that have entered a state of patho-
logical inflammation instead of following a healthy healing
timeline. While the typical progression of a healthy wound 1.1. Biology of Wound Healing
from injury to complete wound closure occurs within 30 d,
chronic wounds stall and can remain unresolved indefinitely.[1,2] Normal wound healing consists of four interconnected and
In total, chronic wounds (e.g., pressure ulcers, vascular ulcers, overlapping phases: hemostasis, inflammation, proliferation,
and diabetic ulcers) affect between 2.4 and 4.5 million people in and tissue remodeling, with each phase involving different cell
the United States causing a significant humanistic and finan- populations (Figure  1).[1,6] Acute wounds occur after a sudden
cial burden.[3] Such chronicity can have severe ramifications for insult to the skin, often in the form of a cut or burn. Imme-
diabetic patients, as chronic wounds are the leading cause of diately following injury, re-establishment of hemostasis to stop
diabetes-associated amputations. Due to the threat of chronic bleeding begins through the formation of a clot by local vascu-
wounds to diabetic health and the increasing prevalence of lature constriction, platelet degranulation, and fibrin activation.
diabetes, many researchers have focused on developing more Once bleeding is under control, the inflammatory stage com-
effective strategies for wound treatment. mences. Dead and damaged cells and invading microbes in
The difficulties associated with designing an appropriate the injured tissue are removed through matrix metalloprotease
treatment are potentiated by the systemic complications of dia- (MMP)-mediated phagocytosis and release of reactive oxygen
betes (e.g., tissue hypoxia, impaired inflammatory response, species (ROS) by infiltrating neutrophils, macrophages, and
and decreased collagen production).[4] The typical protocol for lymphocytes.[8] Upon clearance of pathogens and detritus, the
a chronic wound treatment includes debridement of necrotic proliferation phase of healing begins. Growth factors stimulate
tissue, use of topical antibiotics to control infection, and applica- the generation of new tissue, including the formation of blood
tion of a wound dressing (e.g., films, fibers, and hydrogels). Addi- vessels by angiogenesis, granulation tissue by fibroblasts, and
tional therapeutic intervention may take the form of negative pres- a provisional extracellular matrix (ECM) by MMPs. During this
sure therapy, hyperbaric oxygen therapy (HBOT), or application phase, the open surface area of the wound shrinks as myofi-
of growth factors.[3,5,6] A major cause of stalled wound healing broblasts at the wound edges undergo contraction.[9] The final
stems from unresolved infection.[2] Inflamed wounds provide a phase of wound healing is tissue remodeling; new tissue formed
suitable environment for bacterial colonization, with infection in the proliferation phase matures and the vascular density of
causing tissue damage and triggering further inflammation. The the wound decreases from high proliferation-phase levels.
increasing prevalence of antibiotic-resistant bacteria renders treat-
ment with antibiotics less effective, motivating the use of a topical
antimicrobial agent that does not foster resistance. 1.2. Impaired Wound Healing in Diabetes

M. J. Malone-Povolny, S. E. Maloney, Prof. M. H. Schoenfisch


Not all wounds proceed according to a normal healing timeline.
Department of Chemistry Some injuries result in the development of chronic wounds,
University of North Carolina at Chapel Hill wherein injured tissue enters a state of pathologic inflamma-
Chapel Hill, NC 27599, USA tion that results in protracted and incomplete healing. In acute
E-mail: schoenfisch@unc.edu wounds, the four stages of healing typically take place within
The ORCID identification number(s) for the author(s) of this article a month, and the healed tissue closely resembles prewound
can be found under https://doi.org/10.1002/adhm.201801210.
tissue.[10] However, the chronic wound healing timeline can
DOI: 10.1002/adhm.201801210 extend for months or even stall in the inflammation phase

Adv. Healthcare Mater. 2019, 8, 1801210 1801210  (1 of 18) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

indefinitely. As opposed to the sudden injury often responsible


for acute wounds, chronic wounds are more likely to be caused Maggie J. Malone-Povolny
by persistent stimuli (i.e., repeated tissue trauma, pressure is a graduate student in the
points, or ischemia). In these cases, cutaneous tissue breaks Department of Chemistry
down and the damaged tissue can become a medium for bacte- at the University of North
rial growth.[11] Infection and insufficient tissue oxygenation are Carolina at Chapel Hill, under
two major factors that contribute to chronic wound formation.[1] the direction of Prof. Mark H.
Incomplete microbial clearance elongates the inflammatory Schoenfisch. Her research con-
stage of healing. If inflammation is left unchecked, biofilms cerns the use of nitric oxide to
may form, resulting in the self-secretion of an exopolysaccha- extend the lifetime of contin-
ride matrix (EPS) that protects bacteria from the host immune uous glucose biosensors. She
response and antibiotic treatment.[1,2] Normoxic conditions are holds a B.S. in chemistry from
also crucial because oxygen is required for various processes in the University of St. Thomas.
wound healing, including epithelization, angiogenesis, and col-
lagen deposition.[12] Of note, hypoxia (<20 mm Hg) is a reliable
Sara E. Maloney received
indicator of chronic wounds.[13]
her B.S. in chemistry from
Therefore, systemic factors or diseases that induce tissue
Towson University. She is
hypoxia and/or leave a patient vulnerable to infection often
working toward her Ph.D. in
lead to chronic wounds. Conditions that correspond with poor
chemistry under the guidance
wound healing outcomes include malnutrition, senescence,
of Prof. Mark H. Schoenfisch
stress, obesity, alcoholism, cancer, immunodeficiency, and
at the University of North
ischemia. However, diabetes is the disease that contributes to
Carolina at Chapel Hill. Her
the greatest number of diagnosed chronic wounds. Collectively,
research interests include
diabetes is a group of metabolic diseases typified by dysregula-
developing nitric oxide-
tion of blood glucose levels that often leads to chronic wound
releasing biopolymers for
formation through complex pathophysiological mechanisms
wound healing applications.
(Figure  2).[1,14] First, frequent hyperglycemia (i.e., increased
blood glucose levels) results in increased levels of advanced
glycation end-products (AGEs) in the blood. Glycation end-prod- Mark H. Schoenfisch is
ucts directly contribute to the formation of high concentrations the Peter A. Ornstein
of ROS and reactive nitrogen species (RNS). When ROS/RNS Distinguished Professor of
levels exceed the antioxidant capacity of a given tissue, damage to Chemistry in the Department
the host may tissue occur.[15] In the inflammation stage of wound of Chemistry and Eshelman
healing, ROS/RNS species are essential to wound healing in that School of Pharmacy, Division
they accelerate the clearance of dead tissue and pathogens. At of Pharmacoengineering and
excessive concentrations, these reactive species can prevent tran- Molecular Pharmaceutics.
sition from the inflammatory to the proliferative stage and thus His research interests include
hinder the formation of new, healthy tissue.[9,13] analytical sensors, biomate-
The relative levels of signaling molecules (e.g., proinflam- rials, and nitric oxide-release
matory cytokines, proteases, and growth factors) in chronic therapeutics.
wounds are often imbalanced, contrary to the strict regulation
and coordination found in acute wounds. Chronic wounds
experience a dramatic increase in the concentration of proin- with coinfections because defective leukocyte chemotaxis and
flammatory cytokines directly after injury. Elevated cytokine phagocytosis lead to poorer bacterial load clearance and the
concentrations result in a prolonged inflammatory stage and formation of intractable biofilms.[20] Endotoxins from bacteria
overproduction of MMPs.[16] Upregulation of MMPs causes increase tissue damage, further impairing wound healing.
the continuous breakdown of ECM components and further A schematic of the differences between normal and diabetic
degradation of already undersupplied growth factors.[8,16,17] As wounds is summarized in Figure 3.[21]
such, dysregulated cytokine expression is highly detrimental to While diabetes is a systemic pressure that may cause any
regrowth of healthy tissue. wound a patient endures to become chronic, diabetics are
Lower levels of neuropeptides (e.g., substance P and nerve especially prone to develop a certain type of chronic wound
growth factor) attributed to diabetic neuropathy are also respon- known as a diabetic foot ulcer (DFU). With a lifetime incidence
sible for decreased cell chemotaxis and growth factor produc- in 25% of diabetic patients, DFUs account for almost half of
tion.[18] Decreased levels of growth factors, particularly vascular all diabetes-related hospitalizations and directly precede more
endothelial growth factor (VEGF), disrupt the homing and than 80% of diabetes-related lower leg amputations.[22] Like
mobilization of endothelial progenitor cells (EPCs), which are many chronic wounds, DFUs are often the result of repeated
responsible for restoration of the vasculature. Diminished angi- tissue trauma. This trauma is compounded by the oxidative
ogenesis results in hypoxia caused by insufficient tissue perfu- stress associated with chronic hyperglycemia, which serves to
sion.[19] Diabetic people are more likely to have chronic wounds damage the structural integrity of blood vessels, causing them

Adv. Healthcare Mater. 2019, 8, 1801210 1801210  (2 of 18) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Figure 1.  Time course of different cells populations in the wound during the healing process. Reproduced with permission.[153] Copyright 1997, Elsevier.

to become enlarged. These enlarged veins prevent blood flow As a result, many studies focus on DFUs as both a model for
upwards and out of lower extremities, leading to increased chronic wounds and a critical therapeutic target.
pressure and decreased circulation in the legs and feet.[23] The In summary, acute wounds have an environment with suit-
chronic wound is infiltrated by neutrophils and macrophages able mitogenic activity, low concentrations of inflammatory
in response to the release of inflammatory cytokines by cytokines and MMPs, high levels of growth factors, sufficient
damaged and infected tissue. Concentrations of the inflamma- bacterial clearance, and functioning fibroblasts. This envi-
tory cytokines tumor necrosis factor (TNF-α) and interleukin ronment promotes wounds that heal quickly and completely
have been reported to be up to 100-fold greater in DFUs than without the need for significant therapeutic intervention. In
in acute wounds.[8] A prolonged inflammatory phase effec- contrast, chronic wounds have low mitogenic activity, high
tively bypasses the release of growth factors that would signal levels of both inflammatory cytokines and MMPs, low levels
the healing cascade to begin in a healthy wound.[24,25] Dia- of growth factors, biofilm formation, and fibroblasts that are
betic patients also have an impaired ability to fight infection, senescent. These wounds experience elongated or even com-
resulting from an impaired immune response to invading pletely arrested healing timelines, which often result in severe
pathogens.[20] Wounds that occur in the lower extremities of patient morbidity.[8] The process of wound healing is best
diabetic patients thus result in hypoxic, infected tissue, which is understood as a sequential shift in predominant cell popula-
a potent combination that often compromises wound healing. tions as progression is made through the four phases.[26] As
the population and activity of a given cell
type changes through the different stages
of healing, any proposed therapeutic for a
wound must be effectively timed to changing
cell populations.[1]

1.3. Role of Endogenous Nitric Oxide in


Wound Healing

Nitric oxide is an endogenous gasotransmitter


that plays a central role in wound healing. It
is generated from the terminal guanidine
moiety of L-arginine through a five electron
Figure 2.  The potential effects of diabetes on wound healing. Adapted with permission.[1] Copy- oxidation process catalyzed by a group of
right 2010, SAGE Publications. three isozymes, called nitric oxide synthases

Adv. Healthcare Mater. 2019, 8, 1801210 1801210  (3 of 18) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Figure 3.  Mechanisms of normal versus diabetic wound healing. In a normal wound, macrophages combat wound bed pathogens, while fibroblasts
deliver VEGF to initiate regrowth of epithelial cells and close the open wound. In diabetic wounds, insufficient angiogenesis and macrophage func-
tion result in significant pathogen invasion, while lower fibroblast presence delays re-epithelization and wound closure. Adapted with permission.[154]
Copyright 2017, The Royal Society of Chemistry.

(NOS).[27] This group of isozymes includes endothelial (eNOS), processes, and nm–µm concentrations generated by iNOS respon-
neuronal (nNOS), and inducible nitric oxide synthase (iNOS). sible for proinflammatory and antimicrobial effects.[30,31] Thus,
Two of the isozymes, nNOS and eNOS, are constitutively iNOS is the most impactful isozyme in regulating pathogen
expressed and catalyze low level NO generation through cyclic clearance and acute inflammation in wound healing. Tempo-
guanosine monophosphate (cGMP) in vascular endothelial cells rally, NO is produced at higher levels by iNOS during the inflam-
and neurons, respectively.[28] The third isozyme, iNOS, is an matory phase; lower eNOS-derived NO levels become relevant
inducible form only produced in response to acute inflamma- during the proliferative and maturation phases (Figure 4).[32]
tory stimuli.[29] These stimuli include invading pathogens and Nitric oxide plays a central role in the regulation of three
proinflammatory cytokines (e.g., interferon, INF-α, and tumor major parts of the wound healing process: vascular homeo-
necrosis factor, TNF) released after wounding events by neutro- stasis, inflammation, and antimicrobial action. Nitric oxide
phils and macrophages. Once fully expressed, the concentration impacts vascular homeostasis, which is disrupted in the event
of iNOS greatly exceeds that of the two constitutively expressed of an injury. Upon tissue injury, a healthy healing process
forms (i.e., eNOS and nNOS).[30] Nitric oxide has concentration- depends upon platelet aggregation and blood clot formation
dependent characteristics, with pm–nm concentrations generated to stop further blood loss. Nitric oxide generated by eNOS
by eNOS and nNOS corresponding with anti-inflammatory serves to prevent platelet adhesion to the vessel walls. Reactive

Figure 4.  Temporal production of NO within the healing wound juxtaposed with the phases of healing and the cell populations that predominate each
phase. Reproduced with permission.[32] Copyright 2000, Wolters Kluwer Health, Lippincott Williams & Wilkins.

Adv. Healthcare Mater. 2019, 8, 1801210 1801210  (4 of 18) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

oxygen species released by the damaged vasculature react with 1.4. Nitric Oxide as a Wound Healing Therapeutic
and inactivate NO, with the resulting NO deficiency leading
to platelet aggregation and thrombus formation.[33,34] Nitric Since the discovery of the roles of endogenous NO in wound
oxide has been shown to regulate cytokines that initiate inflam- healing processes in the late 1990s, many have pointed to the
mation (e.g., interleukins, monocytes, and neutrophils) with potential utility of NO as a therapeutic agent for the treatment
downstream effects on the recruitment of keratinocytes.[35,36] A of chronic wounds.[32,50] As described above, levels of NO have
detailed summary of NO’s impact on a panel of inflammatory been implicated in the failure of the healing process. Insuf-
cytokines has previously been reported by Kobayashi et al.[35] ficient NO in diabetic wounds impedes migration and action
Nitric oxide has also been shown to upregulate MMP concen- of wound healing cell types, while excessive NO, associated
trations,[37] modulate the migration and attachment of neutro- with infected or highly inflamed wounds, results in further
phils to the endothelium,[36,38] and coordinate leukocyte recruit- tissue damage. Therefore, modulation of NO has been seen as
ment in the wound-healing timeline.[39] an attractive solution to impaired wound healing. The use of
Lastly, NO plays an essential role in antimicrobial mecha- NO as a wound healing therapeutic may be categorized by two
nisms that decrease bacterial loads at wound sites. Upon injury broad strategies: generating NO from endogenous reservoirs
to the skin, a portal is created for the invasion of the injured and dosing with NO from exogenous sources.
tissue by microbial pathogens.[40] The tissue damage caused Generation of NO from endogenous sources may be accom-
by microbe-released toxins can delay or prevent proper healing plished by two mechanisms: L-arginine supplementation as a
mechanisms. Nitric oxide has antimicrobial properties resulting substrate for NOS and inhibition of NOSs, specifically iNOS.
from a combination of nitrosative and oxidative mechanisms.[41] Dosing exogenous NO can be accomplished by a number of
Upon reaction with oxygen and superoxide, nitric oxide forms methods. Simple techniques include treating wounds using
dinitrogen trioxide and peroxynitrite, respectively. Dinitrogen formulations of nitrite under acidified conditions or gaseous
trioxide causes DNA deamination; peroxynitrite results in lipid NO. Further, small molecule NO-releasing donors can be
peroxidation and membrane damage.[42,43] While eukaryotic applied directly to wounds via their encapsulation in polymers,
cells have evolved mechanisms to scavenge these reactive spe- gels, and dressings. Nitric oxide donors can also be chemically
cies and thus mitigate their impact, bacteria have not and will attached to macromolecular scaffolds, providing potentially
be killed.[44] In addition, NO has been shown to exhibit dose- greater stability to labile NO donors and enabling targeted, tun-
dependent antibacterial activity against biofilms.[45] With addi- able delivery of NO. The goal of both endogenous and exoge-
tional protection afforded by an EPS, these communities of nous therapeutic NO dosage is to regulate inflammation and
bacteria are highly resistant to traditional antibiotics.[46] Though tissue remodeling in chronic wounds.
the exact mechanism of NO’s antibiofilm action remains
unclear, it is known that NO facilitates biofilm dispersal at
low NO concentrations (10−12–10−9 m)[47] and complete biofilm 2. Endogenous Nitric Oxide for Chronic Wound
eradication at higher NO concentrations (10−6–10−3 m).[45,48]
Therapy
Large concentrations of NO from macrophage-generated iNOS,
therefore, act as a broad-spectrum antimicrobial against both 2.1. L-Arginine Supplementation
planktonic and biofilm-based bacteria, reducing bacterial loads
to a level at which normal wound healing processes are able to L-arginine is the sole substrate for nitric oxide synthases and
proceed.[49] the immediate precursor to NO production. Nitric oxide is gen-
Abnormal production of NO, which is a characteristic of erated on the terminal guanidine residue of L-arginine, which
diabetes, has been linked to impaired wound healing and the also generates citrulline as an end product.[60] The body does
development of chronic wounds.[31] Current evidence suggests not provide sufficient amounts of arginine during the times of
that diabetic wound fluid has significantly lower levels of NO stress associated with a wounding event. Thus, supplementa-
than healthy wound fluid, due to downregulated eNOS.[50,51] tion of endogenous arginine stores has been proposed as a way
Macrophages, keratinocytes, and fibroblasts all express elevated to normalize or enhance wound healing. While arginine plays
levels of iNOS at a healthy wound site, but that expression is an important role in other physiological processes (e.g., regula-
suppressed in diabetes. In the absence of the recruiting effect tion of growth hormones, role in the urea cycle, and antioxidant
of NO, macrophages are found in lower densities in chronic generation), for the purposes of this progress report, attention
wounds, diminishing pathogen clearance.[52,53] Inhibited NO will be given to the explicit effect arginine has on NO levels in
production strongly reduces the number of keratinocytes wound healing.
during the process of re-epithelization and further antagonizes Dietary supplementation has been the primary route of argi-
keratinocyte proliferation and differentiation.[54–56] Fibroblasts nine dosage employed. A typical diet allows for up to 5 g of
in diabetic chronic wounds express lower levels of both iNOS arginine to be absorbed per day, with the rest passed through
and eNOS, become senescent, and do not produce collagen the excretory system.[61,62] Nearly 40% of absorbed arginine is
or form ECM. Thus, decreased NO in extracellular wound degraded in the intestines, with the remainder portioned into
fluid corresponds with decreased collagen content and weaker the circulatory system via the portal vein.[61,63] Circulating argi-
wound breaking strength.[57] Levels of NO may also be used as nine is available for NO production and is typically present
prognostic markers, with abnormal NO concentrations serving in concentrations of 75 × 10−6–100  × 10−6 m. Of note, protein-
as reliable predictors of poor healing outcomes in septic burn bound arginine is not available for oxidation by nitric oxide syn-
patients and chronic DFUs.[58,59] thases, as the isozymes only react with free arginine.[61] Given

Adv. Healthcare Mater. 2019, 8, 1801210 1801210  (5 of 18) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

the absorption mechanism of arginine, diet supplementation as the addition of arginine was speculated to impact vascular
would maximize available free arginine in circulation, and thus deficiency. A conclusion about the direct impact NO had on
increase NO production. wound characteristics from this study remains speculative, as
Supplemental arginine therapy has been studied clinically biomarkers for NO activity were not analyzed. While DFUs
since 1978, even before the establishment of arginine’s role as without concomitant impaired blood flow may not benefit
an NOS substrate. In an initial study, Seifter et al. compared from supplemental arginine, patients with impaired perfusion
wound characteristics in rats with an arginine-free diet to those may experience better healing outcome. This result points to a
in rats with an arginine-supplemented diet (i.e., additional mechanism wherein nutrient deficiencies associated with poor
1.8% arginine). The authors found that collagen precursor circulation may be overcome by dosing extra nutrients, such as
deposition and wound breaking strength increased in the argi- arginine, to promote wound healing.
nine-supplemented rats.[64] While this study motivated decades Dietary supplementation is not the only way that arginine
of subsequent dietary arginine research, it was entirely based has been used as a wound healing therapeutic. Administration
on rats with nonchronic wounds, leaving it unclear whether of arginine directly to the wound site has also been shown to
the arginine supplementation would really alter the healing improve wound healing outcomes. Shearer et al. reported that
processes of chronic wounds. The first study that considered systemic administration of arginine can lead wound-site macro­
diabetes-associated chronic wounds was published by Witte phages to release high levels of arginase, suppressing the
et al. in which diabetes was induced using streptozotocin prior amount NO produced through substrate limitation.[68] In this
to creating incisional wounds.[50] A subset of the diabetic rats manner, local administration of arginine to a wound site may
were fed with 1 g kg−1 arginine by gavage, whereas control rats more effectively and selectively modulate wound healing. This
were dosed with saline. Concentrations of nitrite and nitrate strategy was explored by Varedi et al., with a particular interest
were correlated to wound breaking strength. Wound fluid con- in how topical administration of arginine might impact the
centrations of NO and its stable byproducts were depressed in rate of wound closure.[69] Three and five days after creating
the diabetic rats but returned to normal in arginine-fed rats. excisional dorsum wounds in diabetic rats, L-arginine in saline
The wound breaking strength of arginine-supplemented dia- (200  × 10−6 or 400 × 10−6 m) was applied to the wound. The
betic rats was significantly improved compared to saline-dosed rate of wound closure following this treatment was profoundly
controls. This study was the first to suggest that arginine sup- accelerated, with less than 15% of arginine-supplemented
plementation restores NO-production pathways that are typi- wounds still open 12 d postwounding compared to more than
cally deficient in diabetes. Shi et al. reported subsequently that 40% of the control wounds. In order to understand the change
wound breaking strength increases corresponded to enhanced in wound closure rates, tissue morphology was monitored by
hydroxyproline deposition, a collagen precursor.[65] Diabetic rats post-terminal tissue explanation and staining. Arginine-sup-
that were fed a combination arginine/proline-supplemented plemented wounds contained basal keratinocytes that assumed
diet had improved angiogenesis and wound closure rates a flatter, more cuboidal morphology as well as a thinner epi-
compared to isonitrogenous control-fed rats. Macrophage- dermis. Together, these morphological changes suggest that the
expression of iNOS was also reduced in the treated rats, an proliferation and differentiation of keratinocytes is significantly
indication of reduced inflammation favoring expedited wound impacted by the local application of arginine.
closure. The study elucidated the daily progression of wound Therapeutic release of arginine from a traditional wound
healing over the first 5 d after wounding. At day 5, significant dressing has also been investigated as means for accelerating
improvements in angiogenesis and nitrogen balance begin to wound healing. While topical application of aqueous arginine
present.[66] An interesting result of this work was that epitheli- represents a simple method of local delivery, other research has
zation appeared to be unaffected by arginine supplementation focused on the controlled delivery of L-arginine from hydrocol-
after 5 d of treatment, suggesting that dietary supplementation loid-based wound dressings. Encapsulation of L-arginine in a
of arginine may have insufficient therapeutic power in this nanofibrous hydrocolloid was studied by Reesi et al. using a
important aspect of wound healing. rodent wound-healing model.[70] Lignin nanofibers were sur-
More recently, the impact of dietary supplementation of argi- face modified with arginine via electrostatic interactions, which
nine was examined in human trials. As with all human studies, resulted in sustained release of arginine from the dressing.
a large number of subjects and blinded experiments are essen- Nanofibers have become an attractive wound dressing material
tial for overcoming bias in sampling and clinical assessment. due to their high aspect ratio and 3D structure that stimulate
One such double-blind study assessed the effect of an arginine- cell proliferation and skin regeneration, promote hemostasis,
containing oral nutritional supplement on wound healing of and absorb wound exudate.[71] The authors reported con-
DFUs in 270 diabetic patients with chronic wounds.[67] A subset trolled release of arginine for >24 h after application. Arginine-
of the subjects (n  = 130) ingested a drink containing arginine releasing nanofibrous dressings were compared to both dress-
twice daily for 16 weeks. No significant overall difference was ings without arginine and to aqueous solutions of arginine. The
observed between the nutrient-supplemented and control direct comparison to aqueous arginine was used to elucidate
groups in terms of time to heal or wound closure. However, the impact of controlled release via the dressing. Therapeutics
an interesting subpopulation result was discovered. Diabetic applied to incisional wounds were analyzed based on apparent
patients treated with arginine who had decreased limb perfu- re-epithelialization, collagen deposition, vascular formation,
sion were 1.66 times more likely to heal than those with normal and wound closure rates. Arginine-releasing dressings were
limb perfusion. The authors attributed these data to the posi- characterized as having increased re-epithelization, more exten-
tive influence of NO on microvascular hemodynamic changes, sive collagen deposition, and a greater number of new vessels

Adv. Healthcare Mater. 2019, 8, 1801210 1801210  (6 of 18) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

than any of the control therapeutics. The most dramatic result mercan et al.[76] The authors of this study were especially inter-
came from the comparison of wound closure rates between ested about large NO levels produced by iNOS during sepsis,
arginine dressings, arginine solutions, and controls. While the whereby NO acts as an endotoxin. In order to reduce potential
control- and solution-treated wounds achieved closure levels of tissue damage at high NO concentrations, two iNOS inhibi-
only (57.3 ± 4.9) and (49.1 ± 4.2)%, respectively, treatment with tors N-nitro-L-arginine methyl ester (L-NAME) and N5-(1-
sustained arginine release resulted in (93.2 ± 3.7)% closure as imino-methyl)-L-ornithine (L-NIO) were employed to reverse
compared to day 0. This dramatic improvement in closure rates the impairment of wound healing by endotoxemia. Escherichia
constitutes compelling evidence for the benefits of arginine coli endotoxin was injected into the wounds, which were subse-
release from a polymer dressing into a wound. quently treated with 10 mg kg−1 L-NAME, L-NIO, or saline con-
In summary, the use of arginine represents a potential trol. The extent of collagen and wound breaking strength were
therapy for treating chronic wounds due to its ability to increase studied as a function of the specific treatment given. Endotoxin-
NO concentrations in the wound area. Therapeutic arginine can infected wounds treated with either inhibitor displayed much
be dosed through dietary supplementation, topical application, higher collagen content than wounds treated with saline control.
or controlled release from a wound dressing. As local, controlled Further, mean tensile strength of the wounds also improved
delivery of any therapeutic is desired to achieve selective and with the addition of the iNOS inhibitors, with the irreversible
effective treatment, wound dressings that controllably release action of L-NIO actually displaying tensile strength that was not
arginine are a promising future direction for the field. distinguishable from a noninfected healed wound, suggesting
that iNOS inhibition, especially by L-NIO, restores the produc-
tion of collagen in impaired wounds to normal levels.
2.2. NOS Isozymes Regulation However, Stallmeyer et al. reported an important caveat to the
Karamercan sepsis study.[56] They found that L-NIO treatment in
Nitric oxide synthase isozymes are responsible for NO pro- rats that were not septic experienced a severely impaired re-epi-
duction in the body. In this manner, direct regulation of these thelization. In particular, the epithelia at the edges of the wound
enzymes may prove useful for therapeutic purposes. Schwen- displayed an atrophied morphology, with lessened keratinocyte
tker et al. suggested that iNOS is the most efficacious choice proliferation. It was concluded that iNOS is essential to re-epi-
among the nitric oxide synthase isozymes for use as a wound thelization and therefore critical to healthy wound healing. With
healing therapeutic.[72] Both eNOS and iNOS play important respect to wound healing, underexpression of iNOS was deter-
roles in the wound healing process: eNOS is expressed consti- mined to be as detrimental as uninhibited iNOS.
tutively and iNOS synthesis is upregulated in times of tissue The ratio of eNOS to iNOS in a wound environment appears
stress. Though eNOS and iNOS are both necessary and active to be essential to the prognosis of wound healing. While inhi-
in wound healing mechanisms, the moderate levels of NO gen- bition of iNOS may improve healing outcomes in severely
erated by eNOS regulate inflammation whereas iNOS provides infected wounds, it is antagonistic to healing of noninfected
levels of NO that are large enough to eradicate acute infection- chronic wounds. NOS regulation as a therapeutic for wound
causing pathogens. Previous reports have described wound healing does not appear to be as broadly applicable as other
healing in iNOS-knockout mice and have found that cutaneous NO-based therapeutic strategies.
wound healing is severely impaired in the total absence of
iNOS.[73] Other studies have conversely found that inhibition of
nitric oxide synthase improves ischemia and reperfusion inju- 3. Exogenous Nitric Oxide for Chronic
ries.[74] These opposing results, which point to the concentra-
Wound Therapy
tion-dependent impact of NO, motivate the regulation of NOS
enzymes as a potential therapy. 3.1. Gaseous NO
The possibility of delivering NOS to the wound site using
a gene vector technique was reported by Yamasaki et al.[75] A straightforward way to dose NO to a chronic wound is in its
The severely compromised wound healing response in iNOS gaseous form. Applying high pressures of therapeutic gases,
knockout mice was restored following a single application of particularly oxygen, is commonly employed through hyper-
an adenovirus gene vector containing human iNOS cDNA. In baric therapy.[6] Hyperbaric oxygen therapy (HBOT) is a rec-
comparison to control wounds, wounds treated with the iNOS ommended therapy for treating chronic wounds because it
vector had increased nitrite concentrations and less time to counteracts tissue hypoxia. Nitric oxide can also be employed
complete wound closure (15 vs 25 d). The authors chose iNOS in hyperbaric therapy and has even been included in traditional
over its constitutive isoforms because high-level NO generation HBOT as a mediator of the toxic effects of high pressures of
catalyzed by iNOS allowed for application of a single topical oxygen.[6] As a gas, NO can be administered directly to a wound
dose rather than periodic reapplication. Despite the promising as the primary therapeutic agent without the need for any
results of this study, no additional reports have appeared fur- additional carrier species. The dose and duration of therapy is
thering the delivery of NOS genes for wound healing applica- controllable through manipulation of the concentration and/or
tions. In light of the considerable improvement in gene delivery flow rate of the therapeutic gas.
methods in those intervening years, this iNOS delivery strategy Miller et al. reported the use of gaseous NO to address a
merits careful reconsideration. chronic DFU that had persisted for more than 2 years.[77] For
The utility of regulating iNOS expression in response to 14 consecutive days, the foot of the diabetic patient was con-
infections present in chronic wounds was explored by Kara- tinuously exposed to 200 ppm of NO (balance air) using a

Adv. Healthcare Mater. 2019, 8, 1801210 1801210  (7 of 18) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

hyperbaric boot device. While the wound was initially badly for treatment of chronic wounds that are not simultaneously
infected and malodorous, steady and positive healing was infected.
quickly noted. At day 3, healthy granulation tissue had formed, The therapeutic potential of gaseous NO is often limited
and by day 14, the ulcer surface area was reduced by 70%. Six to a hospital setting due to the hazards associated with pres-
weeks after treatment, the wound was 90% healed, with 100% surized NO cylinders and need for continuous oversight.[49,83]
healing achieved at 26 weeks. While the healing timeline of this Additional concerns regarding gaseous NO delivery include
wound was still extended, the gaseous NO initiated healing of NO’s high reactivity, particularly with oxygen in the air, to form
the wound that heretofore had not advanced for 2 years. Impor- harmful byproducts, such as nitrogen dioxide (NO2).[84] This
tantly, side effects or discomfort from this treatment were not reactivity often necessitates that gaseous NO therapy occur in
reported. anoxic environments, limiting the potential for therapeutic use
This promising preliminary case study was supplemented outside of a hospital setting.[83] Both NO and NO2 pose serious
by the analysis of the efficacy of gaseous NO in more statisti- systemic toxicity concerns when inhaled, starting at concentra-
cally robust studies, at the cost of moving from a human sub- tions as low as 40 and 1.5 ppm, respectively.[85] Thus, signifi-
ject to an animal model. Due to NO’s role as an antimicrobial cant safety protocols and monitoring are required for gaseous
agent, gaseous NO administration may reduce the influence NO treatments of chronic wounds.
that bacterial infection has on impaired healing. In terms of
safety, Rezakhanlou et al. reported that gaseous NO could be
safely dosed for 8 h at 5, 25, 75, and 200 ppm using a mouse 3.2. Generation of Nitric Oxide by Acidified Nitrite
lymphocyte model.[78] None of the studied concentrations of
gaseous NO damaged the ECM and some even increased lym- The reaction of nitrite with acid also results in NO production
phocyte proliferation. Pressures greater than 200 ppm resulted exogenously through a three-step reaction. First, nitrite reacts
in decreased cell viability and immune cell proliferation. These with free hydrogen cations to form nitrous acid (HNO2). Nitrous
results imply that wound infections can be treated with NO at acid then decomposes to yield dinitrogen trioxide (N2O3), with
concentrations of 5–200 ppm. With particular interest in NO’s water as a byproduct. Subsequently, N2O3 dissociates in aqueous
antibacterial action, Ghaffari et al. tested 8-h exposures of environments to form equimolar amounts of nitrogen dioxide
200 ppm on rabbits with severe Staphylococcus aureus infected (NO2) and NO.[86] This facile generation of NO allows for local-
wounds.[79] While this treatment protocol did not impact re- ized treatment, with salts like sodium nitrite easily incorporated
epithelization or angiogenesis, it improved collagen deposition into creams and ointments. While nitrite applied by itself to a
and wound breaking strength. Most promisingly, the bacterial wound would interact with basal acidic species to produce NO,
loads of the wounds were reduced by one log after just three it is most effective to provide acidic coreactants to assure com-
daily rounds of treatment. The reduced infection corresponded plete conversion of nitrite to NO.[87] Two creams combined on
with a less severe immune response, an essential step in a the skin, one incorporating sodium nitrite and the other an
healthy wound timeline progression. This study serves as evi- acidic agent, usually citric or ascorbic acid, will immediately
dence for gaseous NO as a potent antibacterial agent and its generate NO. The NO payloads are tuned by varying the concen-
potential utility for treating chronic wounds. tration of sodium nitrite and acid in the respective creams.
Shekhter et al. employed higher concentrations of NO (up In two studies, Zhu et al. demonstrated the use of acidified
to 500 ppm) but for only 60 s once-daily for 6 d to mitigate nitrite solutions to treat full thickness burn wounds in rats and
toxicity concerns.[80] Even with an intermittent dosing mecha- mice.[88,89] In both cases, the wounds were nonchronic, but the
nism, rats with both infected and clean wounds were character- experiments proved useful in understanding the effects of NO on
ized as having reduced tissue hypoxia and microbial infection, general wound healing. For example, wound closure rates and re-
increased angiogenesis, significantly reduced recovery time, epithelization were assessed upon applying two-part hydroxyethyl-
and improved tissue morphology. Analysis of the wound fluid cellulose-based gel to burn wounds. The authors reported at least
suggested a mechanism for gaseous NO’s impact on wound a 50% increase in the wound closure rate for all nitrite-treated
healing. Specifically, the authors noted that exposure to gaseous wounds relative to controls. The concentration of NO in the wound
NO increased the levels of peroxynitrite in the wound tissue. fluid was measured electrochemically to couple results to thera-
Peroxynitrite, a cytotoxic free radical, initiates mobilization of peutic mechanism. Higher concentrations of NO (>5  × 10−3 m)
protective mechanisms, including NO production.[81,82] The resulted in more beneficial outcomes. Furthermore, suspension
authors established that treatment by gaseous NO is accompa- of the treatment after 3 d resulted in loss of healing improve-
nied by enhanced levels of NO in tissue, which regulates the ments. The authors claimed that NO-promoted growth factors
healing processes of impaired chronic wounds. (e.g., transforming growth factor (TGF-β1), and interleukin (IL-8))
Gaseous NO as a therapeutic for nonhealing wounds has also stimulated inflammatory cell infiltration, which is essential for the
been explored in terms of antimicrobial action. Many chronic removal of damaged tissue and pathogens. Together, the studies
wounds enter into an impaired healing state, in part, because underlined the importance of quantifying NO in the wound to
of concomitant infections. While NO has been shown to reduce ensure proper therapeutic timeline and doses.
the bacterial load in infected wounds, the direct impact of gas- Of importance, the physiology of healthy wounds and
eous NO on inflammatory processes requires further elucida- chronic wounds is different. Chronic diabetic wounds are often
tion. Currently, such impact has not been separated from the so impaired that they have stopped progressing along a healing
effect of lowered bacterial load. Understanding the role of gas- timeline. In these cases, any healing enhancement is consid-
eous NO therapy in the inflammatory response might allow ered a positive outcome. Diabetes-associated chronic wounds

Adv. Healthcare Mater. 2019, 8, 1801210 1801210  (8 of 18) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

in mice were used by Weller et al. to study the effects of an preclinical studies.[95,96,99,100] The biocidal action of the NO-np
acidified nitrite cream on wound closure.[90] Specifically, mice system was initially examined against MRSA- and Acinetobacter
treated daily with an acidified nitrite cream 18 d postwounding baumannii-infected wounds using mouse models.[95,96] In both
had greater than 90% closure of incisional wounds versus con- instances, accelerated wound closure rates were reported with
trols, which were only 60% healed (i.e., closed). The timing of NO treatment. Mihu et al. reported complete closure of full-
the treatment protocol was found to be critical to the outcome, thickness infected wounds within 7–8 d as compared to control
with cream applied on the day of wounding significantly inhi­ wounds, which healed only after 10–12 d.[96] Further analysis
biting wound closure rates when compared to a placebo cream determined that the bacterial levels in each set of wounds was
(51.9% vs 80.6%, respectively). Creams applied beginning the significantly reduced by the addition of NO-np. Increased col-
third day after wounding achieved full wound closure over the lagen content was also reported for mice treated with NO-np,
same timeline, indicating that supplemental NO in diabetic an early indicator of tissue remodeling (Figure 5).[95,96]
models in the early stages of healing, hemostasis and early To explore the behavior of fibroblasts in response to NO-np
inflammation, may not be as helpful as in later stages, when therapy, an in vitro scratch assay was performed using human-
NO deficiency can cause healing processes to stall. sourced dermal fibroblasts.[98] Fibroblast migration was
Numerous examples have appeared in the literature observed to accelerate when treated with NO-np relative to
describing the application of acidified nitrite creams to human controls (non-NO-releasing nanoparticles) or those wounds
wounds. The first such study commenced in 2004 and moni- not treated. More rapid migration of fibroblasts is expected
tored 37 patients with Buruli ulcers (chronic ulcers contami- to increase collagen formation, a primary function of this
nated with a severe mycobacterial infection).[91] Sodium nitrite/ cell type. This in vitro assay potentially points to a biological
citric acid gels at two doses (6 and 9 wt% in aqueous cream) mechanism for the increased collagen content observed for
were applied daily to the Buruli ulcers for 6 weeks. Patients NO-treated tissue. An additional study evaluated NO’s role in
treated with the nitrite-based cream had dramatic improvement wound healing by applying the NO-np to noninfected mouse
in healing outcomes. Ulcer size decreased by almost 56% when wounds.[100] Again, increased collagen content was noted in
treated with the acidified nitrite gel, while the placebo group the NO-np-treated mice. More macrophage-like cells were also
saw no change or even increases in ulcer size. This study pro- observed in response to NO treatment, supporting the theory
duced preliminary evidence that treatment with acidified nitrite that NO promotes infiltration of inflammatory cells to the
creams is both well tolerated and effective. Nevertheless, further
work was needed to broaden the applicability of the therapeutic
technique to other chronic wound types. Ormerod et al. used a
similar nitrite cream to treat clinical wounds contaminated with
methicillin-resistant S. aureus (MRSA).[92] Antibiotic-resistant
bacteria that colonize a wound represent a formidable obstacle
to wound healing, as traditional antibiotics fail to eradicate the
entrenched bacteria. Wounds infected with MRSA are exceed-
ingly unlikely to spontaneously heal or be impacted by existing
pharmaceutical intervention.[93] Weller et al. reports that S.
aureus is susceptible to the antimicrobial action of NO.[94] Addi-
tionally, initial NO resistance susceptibility testing indicates
that the likelihood for resistance is low.[7,95–97] Ormerod et al.
reported full eradiation of MRSA colonies with corresponding
improvement in wound closure rates for 15 wounds across eight
patients receiving daily applications of 4.5 wt% nitrite cream
over a 5 d period.[92] Though a lower concentration of nitrite was
used in this study relative to that described in the aforemen-
tioned Buruli ulcer study (i.e., 4.5 wt% vs 6 wt%), both found
that acidified nitrite creams were effective in treating infected,
nonhealing wounds in humans. Nevertheless, this discrepancy
suggests that the required NO dose using acidified nitrite may
vary depending on the condition of the chronic wound.
While acidified nitrite is often applied suspended in oint-
ment, Friedman and co-workers reported on a nitrite-containing
hydrogel/glass composite nanoparticle (NO-np) system.[98] The
particle backbone was composed of tetramethylorthosilicate,
polyethylene glycol (PEG), and chitosan. Nitric oxide, gener- Figure 5.  NO-nps decrease collagen degradation in skin lesions of BALB/c
ated in the particle pores from the thermal reduction of nitrite mice. Histological analysis of mice uninfected and untreated, uninfected
treated with nanoparticles without nitric oxide (np), uninfected treated
by glucose, is released through hydration-mediated uncapping
with NO-nps (NO), untreated MRSA-infected, np-treated MRSA-infected,
of the pores. The overall duration of release from the NO-np and MRSA-infected treated with NO, after 7 d of treatment. Mice were
was roughly 24 h.[98] The authors reported the efficacy of such infected with 107 bacterial cells. The blue stain indicates collagen. Repro-
particles as a wound healing platform through a number of duced with permission.[95] Copyright 2009, Elsevier.

Adv. Healthcare Mater. 2019, 8, 1801210 1801210  (9 of 18) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

wound site. Treatment with NO-np also enhanced wound vas- Often, instability may be circumvented by storing dry samples
cularization, resulting in greater tissue oxygenation, a factor in in dark, cold environments, though this obviously may reduce
promoting wound healing.[100] By increasing fibroblast/mac- the viability of RSNO-based dressings in a clinical setting.
rophage migration and angiogenesis, wounds treated with acid- A second class of NO donors, diazeniumdiolates (NONO-
ified nitrite should heal faster than untreated wounds. ates), are often exploited to address RSNO storage concerns.
These studies show that nitrite-derived NO treatment Diazeniumdiolate NO donors can form on either carbon or
is beneficial to both healthy and infected wounds. Further nitrogen atoms, but N-diazeniumdiolates are the far more
research determined whether these benefits also apply to a common system due to spontaneous release of NO under physi-
healing-impaired model using nonobese, diabetic, severe ological conditions.[97,107] Briefly, N-diazeniumdiolates form
combined immunodeficiency mice.[99] After 7 d of once daily on secondary amines through reaction with high pressures of
treatment, the NO-np materials led to wound healing. The NO- gaseous NO under basic conditions. Proton-initiated release of
treated wounds had ≈57% closure versus ≈15% for controls. NO occurs spontaneously in aqueous media at physiological
The authors concluded that treatment with the acidified nitrite- pH.[107,108] The NO-release kinetics of NONOates are also con-
based NO-releasing nanoparticles was beneficial for healing of trolled by environmental factors (e.g., pH, temperature) and
healthy, infected, and immunocompromised chronic wounds. the structure of the NO donor precursor (e.g., polyamines). For
Of note, these nanoparticles could also be used in combination instance, the presence of protonated primary amines on the NO
with traditional wound treatments (e.g., wound dressings) for donor stabilized the negative charge of the NONOate, and the
individuals struggling with nonhealing diabetic wounds. ensuing, more stable NONOate structure does not break down
Certain side effects must be considered when using acidified to NO as quickly, prolonging NO release.[108–110] Due to their
nitrite-based treatments, as both sodium nitrite and acids can NO-release mechanism, the storage of NONOates requires an
cause skin irritation. A dose-dependent relationship regarding anhydrous environment in order to preserve NO payloads.
adverse reactions (e.g., itching, pain) was reported by Ormerod While low molecular weight NO donors (<1 kDa) are capable
et al. for acidified nitrite treatments.[101] Careful consideration of tunable NO storage and release, certain limitations have
should be made when developing nitrite creams in order to slowed their clinical use. Nitric oxide release from low mole­
accelerate wound healing status without invoking undue skin cular weight donors is often not as well controlled (i.e., rapid
irritation. Identifying the lowest effective doses of nitrite and acid release of NO from precursor structure resulting in undesirable
will ensure maximal healing efficacy with minimal side effects. donor breakdown) and the precursors can be cytotoxic, particu-
Finally, an inability to tune the NO-release kinetics of acidi- larly in the case of the amine-containing donors for N-diaze-
fied nitrite systems undermines their therapeutic utility, as one niumdiolates.[111,112] These consequences may be mitigated
cannot control the rate of NO delivery to the wound. Neverthe- through the use of macromolecular scaffolds for NO delivery.
less, acidified nitrite, whether delivered through a two-part cream Low molecular weight donors may be encapsulated within a
or a nanoparticle system, represents a simple, potentially clini- scaffold (e.g., liposomes) or covalently bound to the scaffold
cally viable method to improve the healing of impaired wounds. structure (e.g., biopolymers, dendrimers, silica nanoparti-
cles).[112] Modifying the scaffold (e.g., molecular weight, size/
shape of nanoparticles) alters the NO-release properties (i.e.,
3.3. Low Molecular Weight and Macromolecular NO Donors NO payload, half-life, and release duration) as well as the thera-
peutic utility (e.g., antibacterial efficacy, cytotoxicity). Utilization
A multitude of NO donor systems have been developed to of macromolecular scaffolds thus allows for enhanced control
store and spontaneously release NO. The vast majority of cur- over NO-release kinetics, with promising utility for enhancing
rent wound treatment strategies employ one of two classes of wound healing. In addition to control over NO-release kinetics
donors: S-nitrosothiols and N-diazeniumdiolates. While both through the choice of NO donor or scaffold, encapsulation
types of donors release NO under physiological conditions, within a polymer matrix allows for precise control over the
each donor category has unique advantages with regards to the properties of the wound dressings. Factors such as hydropho-
therapeutic release of NO. bicity or porosity of the polymer may impact the kinetics of NO
S-nitrosothiols, referred to as RSNOs, are an endogenous release and achievable NO payloads, affecting both antibacterial
class of molecules and also easily formed exogenously via efficacy and wound healing. Balancing these factors, along with
nitrosation of primary thiol groups.[102] Nitric oxide stored on specific physical properties of the films (e.g., flexibility, wound
RSNOs is released by photothermal degradation via homolytic exudate absorption), allows for the fine-tuned development of
cleavage of the SN bond, which liberates one mole of NO wound dressings. To date, NO-based treatment strategies for
per thiol group. Additionally, copper(I) can cause the catalytic both diabetic and nondiabetic chronic wounds are being devel-
decomposition of RSNOs to release NO.[102,103] Due to low oped using low molecular weight donors (e.g., N-diazeniumdi-
endogenous copper levels, photothermal decomposition is the olates and S-nitrosothiols) and macromolecular scaffolds.
primary trigger for NO release in biological systems.[104–106]
These release triggers make it possible for release of NO under
physiological conditions. Diverse NO-release kinetics are pos- 3.3.1. S-Nitrosothiol NO Donors
sible depending on the parent structure of the RSNO and the
degree of exposure to external triggers.[102,103] Instability due to S-nitrosothiols are the endogenous sink of NO in the body.
heat and light exposure may result in premature NO release, The biocompatibility associated with this role motivates the
leading to concerns regarding storage of RSNO-based materials. use of RSNOs for therapeutic action. Li and Lee synthesized

Adv. Healthcare Mater. 2019, 8, 1801210 1801210  (10 of 18) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

poly(vinyl pyrrolidone) (PVP) and poly(vinyl methyl ether-co- illuminated NO-releasing SNAP-gelatin fibers are cytotoxic to
maleic anhydride) (PVMMA) copolymers that gelled from the fully ascertain their potential as wound dressings.
hydrogen bonding between the polymers. The PVMMA was Small molecule RSNOs have also been encapsulated within
directly modified with thiol-containing glutathione (GSH) or polymeric films. Kim et al. reported on NO-releasing chitosan
S-nitroso(γ-Glu-Cys)5-Gly (PC5), precursors for RSNOs.[113] This films doped with NO-modified glutathione (GSNO).[116] As
PVP-PVMMA complex released NO under ambient light condi- chitosan itself has been described as an antibacterial mate-
tions for 10 d (total NO payload of ≈45–80 nmol mg−1 polymer). rial,[117,118] NO-releasing chitosan may prove superior for wound
A diabetic rat model was used to assess the wound healing healing applications. In vitro analysis showed that treatment of
properties of the dry powder. Transparent wound dressings S. aureus and Pseudomonas aeruginosa colonies with the GSNO-
were used to cover the wound and provide a physical barrier. containing chitosan led to a one- and two-log reduction in bac-
Statistically significant improvements in wound closure rates terial load, respectively, versus chitosan control films.[116] A
were observed on days 4, 7, and 10 in rats treated with GSNO- full-thickness (i.e., damage inflicted through the epidermis into
PVMMA/PVP. Wound closure rates were less significant after subcutaneous tissue) rat wound model was used to evaluate the
day 10, coinciding with exhaustion of the NO supply and moti- wound healing potential of the novel dressing. While wound
vating the development of systems capable of extended NO size decreased with time with the control chitosan films, the use
release.[113] This approach is attractive because it is simple and of GSNO-doped chitosan films accelerated wound healing to
easily incorporated with traditional wound dressings. achieve nearly complete closure by day 15 (Figure  6).[116] Anal-
Covalent modification of a polymer with RSNO donors has ysis of fully healed wounds revealed an enhanced formation of
also proved useful in controlling NO-release kinetics. Vogt granulation tissue and increased collagen fiber content. In addi-
et al. found that S-nitroso-N-acetylpenicillamine (SNAP), a tion to bacterial inhibition, the results of this study indicate the
small molecule NO donor, released NO in an extended fashion benefits of NO-releasing chitosan films on tissue remodeling.
when covalently affixed to gelatin nanofibers.[114] As the pri- Hydrogel-based wound treatments provide many benefits for
mary mechanism of NO release from this RSNO was pho- wound healing, primarily in terms of wound hydration.[119] Due
tolysis, the NO release was tunable by light exposure. Nitric to this advantage, a number of small molecule RSNOs encap-
oxide totals were tuned from 24 to 59 nmol mg−1 fibers upon sulated in hydrogels have been developed and evaluated for
illumination with 16–54 candela light. In vitro antibacterial wound healing benefits. De Oliveira and coworkers prepared
assays with Staphylococcus aureus using the zone of inhibition Pluronic-F127 hydrogels that encapsulated two low molecular
assay indicated increased inhibition zones for SNAP-gelatin weight RSNOs: S-nitrosoglutathione (GSNO) and S-nitroso-
fibers relative to controls. Indeed, illumination of the SNAP- N-acetylcysteine.[120] Nitric oxide release through photothermal
gelatin fibers with 54 candela light facilitated complete bacterial decomposition was stabilized by the hydrogel matrix, resulting
inhibition. The light-dependent bacterial inhibition indicates in extended NO release and a half-life of greater than 3 h. Initial
that NO concentration is an essential consideration for NO- in vivo studies in which the hydrogels were applied to human
releasing wound dressings, as higher NO payloads demonstrate forearms confirmed that elevated dermal blood flow persisted
enhanced antibacterial efficacy. Of note, high NO payloads have for at least 3 h after hydrogel application.[121] In this manner,
been shown to be cytotoxic to healthy mammalian cells,[115] and topical application of NO was useful for increasing blood
thus additional studies are required to determine whether the flow near the wound site and providing a potential benefit

Figure 6.  Representative photographs of Sprague–Dawley rat wounds treated with gauze controls, chitosan (CS) films, and NO-releasing chitosan
(CS/NO) films for 15 d. Reproduced with permission.[116] Copyright 2015, Elsevier.

Adv. Healthcare Mater. 2019, 8, 1801210 1801210  (11 of 18) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

towards wound healing. As complications of diabetes include healing efficacy of low molecular weight donor NONOate-modi­
poor blood flow, the authors subsequently focused on diabetic fied diethylenetriamine (DETA/NO). As a powder, DETA/NO
wounds in streptozotocin-induced diabetic rats.[122] Two doses was applied to full-thickness wounds in streptozotocin-induced
of GSNO (23 × 10−3 and 230 × 10−3 m) loaded hydrogels were diabetic rats to monitor any effects over a 21 d period.[26]
applied to the skin of healthy and diabetic rats. The hydrogel Wound closure rates served as the primary measurement of
with greater GSNO concentration (230 × 10−3 m) doubled the wound healing. The application of DETA/NO led to accelerated
perfusion in both normal and diabetic rats. In contrast, the wound closure rates in the diabetic rats.[26] Increased concen-
hydrogel containing the lower GSNO dose (23 × 10−3 m) did trations of nitrate in the rat’s urine provided evidence, albeit
not impact blood flow, suggesting a concentration threshold indirect, that DETA/NO delivered NO to the wound site. Due
for NO to promote perfusion. Of importance, no systemic to the indirect nature of this method, however, the influence of
effects (e.g., changes in systolic blood pressure or heart rate) NO-release kinetics remains nebulous. Follow-on studies exam-
were reported, suggesting the safety and tolerability of this ining whether direct application of the NO donor is cytotoxic
treatment. Increased localized tissue perfusion with the larger to surrounding tissue remain necessary, as small molecule NO
NO dose suggests high potential utility, but further work is donors have been found to be cytotoxic due to their potential
required to determine appropriate dosing protocols. In another decomposition to carcinogenic nitrosamines.[83,111,125]
study, the NO-releasing wound dressings were applied to exci- Macromolecular scaffolds tend to be less toxic as the propen-
sional wounds using a healthy rat model.[123] The GSNO-loaded sity to be phagocytosed is lower. In addition, such materials facil-
hydrogels were applied over varying dosage schedules for 8 d itate additional chemical modifications that may promote tissue
following wounding. Wounded rats had the best healing out- integration. The cytotoxicity concerns for low molecular weight
comes when treated with NO-releasing hydrogels over a period NONOate-based therapeutics can be mitigated by covalently
corresponding with both the inflammatory and proliferative binding the N-diazeniumdiolate NO donors to the macro­
phases rather than during either of those phases alone. At day molecular scaffold. Hetrick et al. first compared the antibacterial
14, only 16.6% of the wounds treated during just one phase efficacy of low molecular weight NONOates (e.g., NO-releasing
were completely healed, whereas 50% of the wounds treated proline (PROLI/NO)) to macromolecular scaffolds (e.g., silica
over both phases healed fully. Application of NO through both nanoparticles).[111] The authors confirmed that at the respective
inflammatory and proliferative phases appears more likely to bactericidal doses, the NO-releasing silica nanoparticles were
benefit overall wound healing in terms of enhanced re-epitheli- significantly less toxic than PROLI/NO, indicating the benefits
alization, collagen fiber maturation, and tissue granulation. of macromolecular NO release for mitigating cytotoxicity.
More recently, de Oliveira and co-workers reported on the Hetrick et al. also reported on the role of NO-release kinetics
combined use of NO-releasing hydrogels and NO-releasing on bactericidal action by comparing N-methylaminopropyltri-
film dressings on mouse excisional wounds during the ini- methoxysilane (MAP3)- and N-(6-aminohexyl)aminopropyltri-
tial stages of healing.[124] Following their previous study, they methoxysilane (AHAP3)-modified silica nanoparticles against
applied either control or GSNO-doped Pluronic-F127 hydrogel bacteria biofilms common to wounds (i.e., P. aeruginosa, S.
to a wound bed. A polymer film containing RSNO-function- aureus, Escherichia coli, Staphylococcus epidermidis, and Can-
alized poly(vinyl alcohol) (PVA) was applied on top of the dida albicans). The NO-release half-lives of the two particle
hydrogel as a wound dressing. Using real-time NO measure- systems were ≈6 and 18 min, respectively. Faster NO-releasing
ments, the authors determined that the underlying hydrogel MAP3-modified particles were ≈1000 times more efficacious
served to modulate the NO release from the PVA film, less- than AHAP3-modified particles with respect to biocidal effi-
ening the burst release and concomitantly extending the overall cacy. With MAP3-modified silica nanoparticles, biofilms from
NO-release duration (>24 h). Together, the combination of the the five tested pathogens were reduced by at least 99%, with
hydrogel and the film outperformed either material on its own Gram-negative E. coli and P. aeruginosa reduced by 99.999%.[126]
in terms of enhanced wound contraction, increased collagen However, even with the use of a nontoxic macromolecular scaf-
deposition, and an accelerated inflammatory stage. The authors fold, considerable cytotoxicity toward L929 mouse fibroblasts
attributed the increased wound-healing activity to the controlled exposed to the NO-releasing nanoparticles was observed, with
manner in which the combined materials released NO. A slow, toxicity comparable with commercial antiseptics (i.e., povidone
prolonged NO-release profile appeared to be most advanta- iodine and chlorhexidine). These materials will only represent
geous for improving wound-healing outcomes. a viable chronic wound treatment option when their toxicity
towards healthy mammalian cells has been addressed.
Shabani et al. reported on an NO-releasing polymer whereby
3.3.2. N-Diazeniumdiolate NO Donors the NONOate group was incorporated within a polyethyl-
eneimine cellulose (PEIC/NO) backbone to demonstrate the
Relative to their S-nitrosothiol counterparts, N-diazeniumdi- utility of covalently binding the NONOates to macromolecular
olates (NONOates) are more often utilized for NO-based wound scaffolds.[127] Repeated application of the PEIC/NO to full-
healing treatments because of their ability to spontaneously thickness dermal models (rat model) enhanced wound clo-
release NO in aqueous media. Both low molecular weight sure rates relative to controls. The authors further analyzed
NONOates and NONOate-modified macromolecular scaffolds the impact of these donors on systemic blood pressure. A two
have been investigated through direct application and in con- NO donor comparison study of PEIC/NO and DETA/NO (half-
junction with current wound treatments, such as wound dress- lives of ≈16 and 20 h, respectively) demonstrated a more severe
ings. For example, Dashti et al. described the in vivo wound decrease in systolic blood pressure for DETA/NO compared

Adv. Healthcare Mater. 2019, 8, 1801210 1801210  (12 of 18) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

to PEIC/NO treatments. However, the mildly depressed sys- compared to controls.[131] The benefit of encapsulating a low
tolic pressure when using PEIC/NO returned to normal levels molecular weight NO donor within a polyurethane matrix lies
within ≈3 h. In contrast, the topical application of DETA/NO in the direct applicability to nearly any commercially available
led to a significant decrease in systolic pressure that lasted for wound dressing.
>6 h.[127] The authors attributed this prolonged hypotension to While polymeric films represent a diverse class of wound
the greater NO storage characteristic of DETA/NO, implying dressings, the use of fibers has additional advantages, par-
that the NO payload delivered to the wound plays a large role ticularly for wound healing applications. Polymeric fibers are
in the efficacy and impact of an NO-based wound healing treat- characterized by increased surface area per volume compared
ment. A follow-on study by the same group investigated the to films. Furthermore, the nanofibrous matrix of the fibers
effects of a similar polymer backbone, linear polyethyleneimine mimics the ECM, providing a 3D scaffold for the migration
(L-PEI/NO), on wound repair in aged rats with impaired wound of inflammatory cells.[132,133] An increase in wound dressing
healing.[128] Surprisingly, neither L-PEI nor L-PEI/NO promoted porosity has been shown to facilitate more oxygen and moisture
wound healing. In addition to L-PEI’s inherent toxicity at high permeation.[134] It has also been reported that the hydration
doses, the addition of NO-release led to clotting and impaired (i.e., water absorption) of electrospun fibers is 10–100 times
collagen organization. The prolonged, high concentrations of greater than analogue film dressings.[132,133,135] Drawbacks to
NO delivered from this system at the studied doses also led to using fibers as NO-releasing wound dressings include potential
adverse tissue responses,[128] reaffirming the need to tune NO undesirable leaching of the NO-modified materials, hastened
payload for a given application. NO-release kinetics due to increased water absorption, and a
Different NO-release kinetics from polyethyleneimine (PEI) lower volume capacity, leading to decreased NO payloads.[136]
were reported by Nurhasni et al. via poly(lactic-co-glycolic acid) The design of NO-releasing electrospun polymer fibers
(PLGA)-coated NONOate-functionalized PEI nanoparticles.[129] containing both low molecular weight NO donors and NO
Sustained NO release (>6 d) was observed and attributed to less donor-modified macromolecular scaffolds was first investi-
water-induced breakdown of the NONOate (to NO). The NO- gated by Coneski et al.[137] Briefly, PROLI/NO, a fast-releasing
releasing PLGA/PEI particles demonstrated potent antibacterial low molecular weight NO donor with a half-life of ≈2 s, was
action against both MRSA and P. aeruginosa (≈99.99% killing), doped into polyurethane and poly(vinyl chloride) fibers.
with low cytotoxicity to mammalian cells (>80% cell viability) The encapsulation of PROLI/NO within the polymer matrix
at particle concentrations as low as 5 mg mL−1. This wound slowed the rate of NO release, increasing the NO release half-
healing therapeutic was studied in vivo using a full-thickness lives by up to 200 times depending on fiber composition and
mouse wound model following application of a single treat- resulting hydrophilicity. Koh et al. synthesized amine-function-
ment 1 d postwounding. Compared to controls, the PLGA/ alized NO-releasing silica nanoparticles doped into electrospun
PEI particles significantly reduced wound area by day 7 (75% polyurethane fibers for application to wounds dressings. The
smaller), promoting skin structure resembling a healthy epi- resulting NO payloads for the silica-doped fibers ranged from
dermis. Together, the studies give evidence that sustained, 7.5 to 120 nmol mg−1.[136] While tunable NO-release kinetics
low doses of NO may be more effective at marshalling wound were achieved by varying NO donor type, polyurethane hydro-
healing than a high NO flux via burst release. phobicity, and dopant concentrations, the overall NO pay-
Though direct application of NO-releasing materials to a loads were low relative to fiber mass. Additionally, fibers con-
wound clearly affects wound healing, NO-based wound treat- taining AHAP3-modified particles leached high levels of silica,
ment strategies must translate to clinical scenarios in order to limiting the therapeutic utility of the NO-releasing fibers.
be relevant.[130] In this regard, both low molecular weight NO Worley et al. subsequently developed dual-action antibacterial
donors and NO-donor-modified macromolecular scaffolds have polyurethane fibers containing biocidal quaternary ammo-
been incorporated within polymeric wound dressings to exploit nium and NO-releasing poly(amidoamine) dendrimers. Using
the potential therapeutic utility of NO release. For example, two biocides improved the antibacterial efficacy and reduced
polymeric films doped with low molecular weight NO donors the risk of resistance by way of multiple mechanistic bacterial
were reported by Brisbois et al. in a study that evaluated the eradication pathways.[138] The authors reported a 4-log reduc-
wound healing potential of polyurethane films doped with tion in bacterial viability for both Gram-negative (E. coli and
dibutylhexanediamine (DBHD/N2O2) and PLGA.[131] The two- P. aeruginosa) and Gram-positive (S. aureus and methicillin-
layer arrangement consisted of an active polyurethane layer resistant S. aureus) bacteria. Minimal cytotoxicity (<20% reduc-
containing DBHD/N2O2 and PLGA, surrounded by a non-NO- tion in viability) to L929 mouse fibroblasts was observed, fur-
releasing topcoat prepared with the same polyurethane composi- ther supporting the therapeutic utility of these wound dress-
tion. Nitric oxide-release kinetics were dependent on the hydro- ings. Of note, none of these materials have been evaluated
phobicity of the polyurethane (i.e., water uptake of 6.2 ± 0.7 preclinically to date, and thus their therapeutic utility has not
vs 22.5 ± 1.1 wt% for polyurethanes SG-80A and SP-60D-20, yet been determined. Nevertheless, the promising nature of
respectively), as increasing hydrophilicity resulted in greater their controllable NO-release kinetics and the prior literature
water diffusion into the film to liberate NO. SG-80A polyure- demonstrating the benefits of NO suggest they could improve
thane, with an NO flux of 12.8–14.5 × 10−10 mol cm−2 min−1 healing outcomes, especially given their efficient NO-delivery
for ≈24 h, was chosen for in vivo testing. The infection model and minimal toxicity.
consisted of exposing mouse burn wounds to Acinetobacter Nitric oxide-releasing electrospun polymer fibers can also
baumannii. Infected wounds treated with the NO-releasing be prepared using acrylonitrile-based terpolymers covalently
polyurethane exhibited a ≈4 log reduction in bacterial counts modified with NONOates.[139] Lowe et al. reported a similar

Adv. Healthcare Mater. 2019, 8, 1801210 1801210  (13 of 18) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Figure 7.  Histological analyses of mouse wound beds with daily administration of NO-releasing dressings. a) Representative histology images of
smooth muscle actin immunohistochemical staining (capillaries stained red). b) Average capillary density for control and NO-treated wounds. Repro-
duced with permission.[139] Copyright 2015, Elsevier.

extended release duration (≈7–14 d) of similar NO payloads production. Additionally, granulation tissue was found to be
to that achieved by Koh et al. (i.e., 79 nmol mg−1 fibers).[136,139] thicker in NO-treated wounds, indicative of increased mechan-
While low NO loading is still prevalent in these fibers, covalent ical stability. As impaired diabetic wound healing is characterized
bonding of the NONOates to the polymer backbone mitigates by decreased fibroblast proliferation, the increase in collagen
the concerns associated with NO donor or macromolecular production and wound breaking strength upon NO treatment
scaffold leaching. These polymer fibers were evaluated using an suggest high clinical potential to improve wound healing.
in vivo excisional wound mouse model to monitor the effects
of NO on wound closure and regulation of endogenous NO-
inducible genes.[139] On day 14, the wounds treated with NO 3.3.3. Zeolites
displayed more significant wound closure relative to controls.
In addition, 60% of the NO-treated wounds displayed a greater While the vast majority of NO-donor-based wound treatment
capillary density than controls (Figure  7).[139] Upregulation of strategies employ the use of N-diazeniumdiolates or S-nitroso-
NO-induced gene expression was also observed for three genes thiols, alternative chemistries do exist. For example, Neidrauer
(i.e., JunB, cFOS, and eNOS) within 30 min of treatment. The et al. developed a topical ointment containing NO-loaded zeo-
analysis of wound healing by monitoring NO-induced gene lites.[141] Rather than relying on covalent binding of NO to a
expression and angiogenesis provides further quantitative evi- donor, zeolites are porous structures that retain gaseous NO
dence of the benefits NO release may have on wound healing through chemisorption. Displacement of the gas by water leads
outcomes. to the spontaneous release of NO in aqueous conditions. The
Beyond films and fibers, NONOate-based wound therapies antibacterial action of an NO-releasing zeolite-based ointment
have also employed hydrogels to create ointments and dress- was exceptional, yielding at least 5-log reductions of E. coli, A.
ings. Kang et al. described an NO-releasing ointment that com- baumannii, S. epidermidis, and MRSA and a 3-log reduction
bined NONOate-functionalized PEI with Pluronic-F127 in a against a fungal species C. albicans after an 8 h exposure period.
PEG hydrogel matrix.[140] While the NONOates released NO for Unfortunately, cytotoxicity studies indicated that only ≈33%
>4 h, the PEG matrix maintained a moist environment neces- of fibroblast cells were viable after treatment for 24 h. While
sary for wound healing. When applied daily to a full-thickness similar results are observed with commercial antiseptics (e.g.,
mouse wound model for 13 d, this NO-releasing ointment povidone and iodine), the cytotoxicity elicited by the zeolites
resulted in advanced re-epithelization, formation of granula- suggest poor utility for wound healing applications. Neverthe-
tion tissue, improved angiogenesis, and near complete wound less, the ointment was tested using obese rats with full-thick-
closure. By incorporating the NO-releasing polymer within ness wounds. Healing rates were determined to be faster for
the moisture-retaining hydrogel matrix, the authors were able NO-treated wounds compared to wounds treated with a control
to demonstrate accelerated wound-healing activity, especially ointment (15.1% and 11.7% d−1, respectively). Despite the ear-
during the proliferative phase of healing. lier noted cytotoxicity, adverse side-effects were not observed in
Masters et al. reported the benefits of covalently coupling the in vivo study.
an NO donor to a PVA hydrogels.[130] The authors modified
the PVA hydrogels with amines to facilitate subsequent NO
donor loading, with two formulations giving total NO pay- 4. Perspectives on NO Therapy for Wound
loads of 0.06 and 0.48 µmol g−1 hydrogel. In vitro cytotox-
Healing
icity studies using fibroblasts revealed that the hydrogels were
nontoxic (>90% cell viability). The utility of the dressings was 4.1. Kinetic Dependence of Nitric Oxide
then tested in vivo using a genetically induced diabetic mouse
model. The application of NO to the wounds led to a significant The field’s knowledge of the biological activity of NO related
increase in collagen; however, this study also noted a concentra- to wound healing is still growing. The healing result of any
tion dependence. The increased NO concentration provided by NO-based wound therapy appears to be highly dependent
the greater payload system facilitated an increase in collagen on NO payload and delivery duration stemming from the

Adv. Healthcare Mater. 2019, 8, 1801210 1801210  (14 of 18) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

concentration-dependent nature of NO on physiological pro- a given indication (i.e., wound healing). As our understanding
cesses. While micromolar concentrations of NO are associated of the biological role of NO in wound healing continues to
with pathogen clearance and proinflammatory signaling, pico- expand, more accurate quantitation of NO is absolutely neces-
molar concentrations of NO stimulate tissue regeneration and sary to fully harness it as a therapeutic.
the resolution of the healing process. The kinetic dependence
of NO’s antimicrobial action has been demonstrated in several
studies,[142,143] albeit this body of work is contradictory. Addi- 4.2. Choice of Chronic Wound Model
tionally, both increased[88,89,116] and decreased[123,144] inflamma-
tory cell counts have been reported for materials with similar In addition to careful monitoring and reporting of NO-release
NO-release properties. A more definitive evaluation of NO kinetics, the use of appropriate and established preclinical
flux on inflammatory cell count and other markers of wound models is essential for assessing the therapeutic to ensure
healing progression is needed to understand whether these translation to the desired host (i.e., humans). Wound healing
opposing results are a result NO release kinetics or other fac- is a complex process consisting of overlapping physiological
tors (e.g., NO donor precursor and/or the polymeric backbone). phases. The extent and nature of healing, therefore, cannot be
Accurate measurement and reporting of relevant NO kinetic properly evaluated by a single assay or test. Rather, a diverse
parameters is also essential for truly understanding both results set of assays should be performed using a given test material,
and how best to implement a given NO-based therapeutic. and then evaluated using an in vivo model. Nearly all of the
While measurement of NO is difficult, even in vitro, because it materials/strategies described in this review were tested using
is a highly reactive species that rapidly degrades to nitrite in oxy- only rodent models with acute wounds. Of course, the healing
genated media (i.e., half-life of NO is <2 s),[37] accurate methods processes between mice/rats and humans differ in significant
for measuring NO quickly do exist, including chemilumines- ways. While mice and rat wounds undergo healing by contrac-
cence, colorimetric assays, and electrochemistry.[145] Further ture of the wound edges, human wounds heal by re-epitheliza-
work is required to enable the use of these analytical tools in tion.[147] Such a difference calls into question the conclusions
more complex settings (i.e., in vivo models), where NO release made about wound healing efficacy for materials only assessed
could be measured alongside assessment of wound healing effi- in a rodent model. Swine represent the gold standard animal
cacy.[88,89] Methods that facilitate kinetic profiling of NO release, model for predicting human wound healing.[148] Like humans,
both in vitro and in vivo, would enable defining important pigs heal through re-epithelization and have similar responses
kinetic parameters, including normalized NO payload, half-life, to growth factors, distinct from small mammals (Figure 8).[148]
and total duration of release. Such ideas are not new; Hunter In a review of animal studies, Sullivan et al. reported the effi-
et al. reported the need for accurate and normalized NO meas- cacy of materials tested on pigs had a 78% percent concord-
urement in complex media.[146] Standard reporting of kinetic ance with human studies, while small mammal studies had
parameters would facilitate easier comparison of NO donor sys- only 53% concordance.[148] In vitro studies were characterized
tems/materials developed in different labs, but also more defin- by 57% concordance, which suggests, surprisingly, that studies
itive conclusions about the ideal NO-release kinetic profile for done in vitro have an equal, or even slightly greater, propensity

Figure 8.  Comparison of histological features from swine and human skin. H&E stained section taken through comparable portions of the dermis in
both tissues. Reproduced with permission.[148] Copyright 2001, Wiley-VCH.

Adv. Healthcare Mater. 2019, 8, 1801210 1801210  (15 of 18) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

to predict response in humans as in vivo work carried out in [6] D. Mathieu, Handbook on Hyperbaric Medicine, Springer,
rodent models. Indeed, no animal model will ever fully rep- Dordrecht, The Netherlands 2006.
licate human wound healing. Thus, it is important to select [7] A. W. Smith, Adv. Drug Delivery Rev. 2005, 57, 1539.
models that come the closest to doing so prior to clinical trials. [8] B. A. Mast, G. S. Schultz, Wound Repair Regener. 1996, 4, 411.
[9] A. C. L. Campos, A. K. Groth, A. B. Branco, Curr. Opin. Clin. Nutr.
Evidence suggests that a shift to pigs as the animal model of
Metab. Care 2008, 11, 281.
choice for wound healing studies would yield more convincing [10] G. Han, R. Ceilley, Adv. Ther. 2017, 34, 599.
and translatable results. [11] J. K. Stechmiller, B. Childress, L. Cowan, Nutr. Clin. Pract. 2005, 20, 52.
Upon selecting an appropriate animal model to evaluate [12] D. M. Castilla, Z.-J. Liu, O. C. Velazquez, Adv. Wound Care 2012, 1, 225.
a potential wound healing therapy, additional steps must be [13] A. A. Tandara, T. A. Mustoe, World J. Surg. 2004, 28, 294.
taken to replicate a chronic wound environment. As discussed [14] A. B. Olokoba, O. A. Obateru, L. B. Olokoba, Oman Med. J. 2012,
in this paper (Section 1.3), chronic and healthy wounds differ 27, 269.
in important ways. Indeed, chronic wounds have been char- [15] S. Bansal, M. Siddarth, D. Chawla, B. D. Banerjee, S. V. Madhu,
acterized as having lower levels of oxygen and NO, prolonged A. K. Tripathi, Mol. Cell. Biochem. 2012, 361, 289.
inflammatory response, fewer growth factors, and increased [16] M. K. Trengove, M. C. Stacey, S. McCauley, N. Bennett, J. Gibson,
F. Burslem, G. Murphy, G. Schultz, Wound Repair Regener. 1999, 7,
susceptibility to infection.[8] Testing a chronic wound thera-
442.
peutic in a healthy wound model fails to take into account these [17] A. B. Wysocki, L. Staiano-Coico, F. Grinnell, J. Invest. Dermatol.
vital differences. Many models have been developed to mimic 1993, 101, 64.
the conditions of a chronic wound in animals. Most commonly, [18] H. Galkowska, U. Wojewodzka, W. L. Olszewski, Wound Repair
type 1 and type 2 diabetes are genetically induced in rodents.[149] Regener. 2006, 14, 558.
Type 1 diabetes is also modeled in swine by dosing with alloxan [19] M. G. Tonnesen, X. Feng, R. A. F. Clark, J. Invest. Dermatol. Symp.
or streptozocin, resulting in pancreatic β-cell destruction.[150] Proc. 2000, 5, 40.
Additional casual factors for chronic wounds (e.g., obesity and [20] T. Hirsch, M. Spielmann, B. Zuhaili, T. Koehler, M. Fossum,
infection) can also be replicated in preclinical wound models, H. U. Steinau, F. Yao, L. Steinstraesser, A. B. Onderdonk,
depending on the therapeutic target of a given wound healing E. Eriksson, BMC Surg. 2008, 8, 1.
[21] H. Brem, M. Tomic-Canic, J. Clin. Invest. 2007, 117, 1219.
therapy.[151,152] Again, the complexity of a chronic wound is dif-
[22] G. E. Reiber, E. J. Boyko, D. G. Smith, Diabetes in America,
ficult to fully recapitulate in a single animal model, but careful US Government Printing Office, Bethesda, MD 1995, pp. 409–428.
consideration must be taken in the choice of model for a given [23] I. C. Valencia, A. Falabella, R. S. Kirsner, W. H. Eaglstein, J. Am.
wound healing therapeutic. Acad. Dermatol. 2001, 44, 401.
[24] R. A. Bryant, Acute and Chronic Wounds: Nursing Management,
Mosby, London 2000.
[25] T. J. Fahey, A. Sadaty, W. G. Jones, A. Barber, B. Smoller,
Acknowledgements G. T. Shires, J. Surg. Res. 1991, 50, 308.
Financial support was provided by the National Institutes of Health [26] N. Dashti, M. Ansari, M. Shabani, S. Vardasti, A. Mirsalehian,
(DK108318). M. H. Noori Mughehi, Z. N. Hatmi, Iran. J. Public Health 2003,
32, 59.
[27] D. S. Bredt, S. H. Snyder, Proc. Natl. Acad. Sci. USA 1990, 87, 682.
[28] R. G. Knowles, S. Moncada, Biochem. J. 1994, 298, 249.
Conflict of Interest [29] U. Förstermann, W. C. Sessa, Eur. Heart J. 2012, 33, 829.
The authors declare the following competing financial interest(s): M.H.S. [30] S. Donnini, M. Ziche, Antioxid. Redox Signaling 2002, 4, 817.
is a cofounder and maintains a financial interest in Novan, Inc. and Vast [31] C. F. Nathan, J. B. Hibbs, Curr. Opin. Immunol. 1991, 3, 65.
Therapeutics, Inc. Both companies commercialize macromolecular nitric [32] D. T. Efron, D. Most, A. Barbul, Curr. Opin. Clin. Nutr. Metab. Care
oxide storage and release vehicles for clinical indications. 2000, 3, 197.
[33] J. Loscalzo, J. Freedman, A. Inbal, J. F. Keaney, A. D. Michelson,
J. A. Vita, Trans. Am. Clin. Climatol. Assoc. 2000, 111, 158.
[34] J. E. Freedman, J. Loscalzo, J. Thromb. Haemostasis 2003, 1, 1183.
Keywords [35] Y. Kobayashi, J. Leukocyte Biol. 2010, 88, 1157.
chronic wounds, diabetes, nitric oxide, wound healing [36] M. N. Ajuebor, L. Virág, R. J. Flower, M. Perretti, C. Szabó,
Immunology 1998, 95, 625.
Received: September 27, 2018 [37] D. D. Thomas, L. A. Ridnour, J. S. Isenberg, W. Flores-Santana,
Revised: November 12, 2018 C. H. Switzer, S. Donzelli, P. Hussain, C. Vecoli, N. Paolocci,
Published online: January 15, 2019 S. Ambs, C. Colton, C. Harris, D. Roberts, D. Wink, Free Radic.
Biol. Med. 2008, 45, 18.
[38] S. Nolan, R. Dixon, K. Norman, P. Hellewell, V. Ridger, Am. J.
Pathol. 2008, 172, 265.
[1] S. Guo, L. A. DiPietro, J. Dent. Res. 2010, 89, 219. [39] G. Andonegui, P. Kubes, in Nitric Oxide Inflamm., Birkhauser
[2] T. Bjarnsholt, K. Kirketerp-Møller, P. Ø. Jensen, K. G. Madsen, Verlag, Basel, Switzerland 2001, pp. 117–118.
R. Phipps, K. Krogfelt, N. Høiby, M. Givskov, Wound Repair [40] A. I. Vinik, R. E. Maser, B. D. Mitchell, R. Freeman, Diabetes Care
Regener. 2008, 16, 2. 2003, 26, 1553.
[3] N. Richmond, A. Maderal, A. Vivas, Dermatol. Ther. 2013, 26, 187. [41] D. A. Wink, J. B. Mitchell, Free Radical Biol. Med. 1998, 25, 434.
[4] R. Blakytny, E. Jude, Diabetic Med. 2006, 23, 594. [42] F. C. Fang, Nat. Rev. Microbiol. 2004, 2, 820.
[5] J. G. Powers, C. Higham, K. Broussard, T. J. Phillips, J. Am. Acad. [43] M. N. Möller, Q. Li, J. R. Lancaster, A. Denicola, IUBMB Life 2007,
Dermatol. 2016, 74, 607. 59, 243.

Adv. Healthcare Mater. 2019, 8, 1801210 1801210  (16 of 18) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

[44] F. C. Fang, mBio 2011, 2, 1. [80] A. B. Shekhter, V. A. Serezhenkov, T. G. Rudenko, A. V. Pekshev,


[45] N. Barraud, M. Kelso, S. Rice, S. Kjelleberg, Curr. Pharm. Des. A. F. Vanin, Nitric Oxide 2005, 12, 210.
2014, 21, 31. [81] K. Laude, C. Thuillez, V. Richard, Am. J. Physiol.: Heart Circ. Physiol.
[46] U. Römling, C. Balsalobre, J. Intern. Med. 2012, 272, 541. 2002, 283, H1418.
[47] S. Hossain, L.-M. Nisbett, E. M. Boon, Acc. Chem. Res. 2017, 50, 1633. [82] P. García-Nogales, A. Almeida, J. P. Bolaños, J. Biol. Chem. 2003,
[48] B. H. Neufeld, M. M. Reynolds, Biointerphases 2016, 11, 031012. 278, 864.
[49] D. O. Schairer, J. S. Chouake, J. D. Nosanchuk, A. J. Friedman, [83] Y. Yang, P. K. Qi, Z. L. Yang, N. Huang, Biosurf. Biotribol. 2015, 1,
Virulence 2012, 3, 271. 177.
[50] M. B. Witte, T. Kiyama, A. Barbul, Br. J. Surg. 2002, 89, 1594. [84] O. I. Miller, D. S. Celermajer, J. E. Deanfield, D. J. Macrae,
[51] X. L. Du, D. Edelstein, S. Dimmeler, Q. Ju, C. Sui, M. Brownlee, Arch. Dis. Child. 1994, 70, F47.
J. Clin. Invest. 2001, 108, 1341. [85] B. Weinberger, D. L. Laskin, D. E. Heck, J. D. Laskin, Toxicol. Sci.
[52] K. Shimizu, S. Naito, Y. Urata, I. Sekine, T. Kondo, I. Katayama, 2001, 59, 5.
Br. J. Dermatol. 1998, 138, 769. [86] R. C. Brasted, Anal. Chem. 1952, 24, 1111.
[53] M. R. Kreuger, D. R. Tames, M. Mariano, Immunology 1998, 95, 278. [87] R. N. Dave, H. M. Joshi, V. P. Venugopalan, J. Mater. Sci.: Mater.
[54] I. Arany, M. M. Brysk, H. Brysk, S. K. Tyring, Biochem. Biophys. Res. Med. 2012, 23, 3097.
Commun. 1996, 220, 618. [88] H. Zhu, B. Ka, F. Murad, World J. Surg. 2007, 31, 624.
[55] V. Krischel, D. Bruch-Gerharz, C. Suschek, K. D. Kröncke, [89] H. Zhu, X. Wei, K. Bian, F. Murad, J. Burn Care Res. 2008, 29, 804.
T. Ruzicka, V. Kolb-Bachofen, J. Invest. Dermatol. 1998, 111, 286. [90] R. Weller, M. J. Finnen, Nitric Oxide 2006, 15, 395.
[56] B. Stallmeyer, H. Kämpfer, N. Kolb, J. Pfeilschifter, S. Frank, [91] R. Phillips, O. Adjei, S. Lucas, N. Benjamin, Society 2004, 48,
J. Invest. Dermatol. 1999, 113, 1090. 2866.
[57] M. R. Schäffer, U. Tantry, P. A. Efron, G. M. Ahrendt, F. J. Thornton, [92] A. D. Ormerod, A. A. J. Shah, H. Li, N. B. Benjamin,
A. Barbul, Surgery 1997, 121, 513. G. P. Ferguson, C. Leifert, BMC Res. Notes 2011, 4, 458.
[58] J. V. Boykin, J. Wound Ostomy Continence Nurs. 2010, 37, 25. [93] N. Tentolouris, G. Petrikkos, N. Vallianou, C. Zachos, G. L. Daikos,
[59] J.-C. Preiser, P. Reper, D. Vlasselaer, B. Vray, H. Zhang, G. Metz, P. Tsapogas, G. Markou, N. Katsilambros, Clin. Microbiol. Infect.
A. Vanderkelen, J.-L. Vincent, J. Trauma: Inj., Infect., Crit. Care 1996, 2006, 12, 186.
40, 368. [94] R. Weller, R. J. Price, A. D. Ormerod, N. Benjamin, J. Appl. Micro-
[60] J. B. Hibbs, R. R. Taintor, Z. Vavrin, Science 1987, 235, 473. biol. 2001, 90, 648.
[61] G. P. Zaloga, R. Siddiqui, C. Terry, P. E. Marik, Nutr. Clin. Pract. [95] L. R. Martinez, G. Han, M. Chacko, M. R. Mihu, M. Jacobson,
2004, 19, 201. P. Gialanella, A. J. Friedman, J. D. Nosanchuk, J. M. Friedman,
[62] C. Nieves, B. Langkamp-Henken, Biomed. Pharmacother. 2002, 56, J. Invest. Dermatol. 2009, 129, 2463.
471. [96] M. R. Mihu, U. Sandkovsky, G. Han, J. M. Friedman, J. D. Nosanchuk,
[63] H. S. Sitren, H. Fisher, Br. J. Nutr. 1977, 37, 195. L. R. Martinez, Virulence 2010, 1, 62.
[64] E. Seitfer, G. Rettura, A. Barbul, S. M. Levenson, Surgery 1978, 84, [97] L. Yang, E. S. Feura, M. J. R. Ahonen, M. H. Schoenfisch, Adv.
224. Healthcare Mater. 2018, 7, 1.
[65] H. P. Shi, R. S. Fishel, D. T. Efron, J. Z. Williams, M. H. Fishel, [98] A. J. Friedman, G. Han, M. S. Navati, M. Chacko, L. Gunther,
A. Barbul, J. Surg. Res. 2002, 106, 299. A. Alfieri, J. M. Friedman, Nitric Oxide 2008, 19, 12.
[66] A. Raynaud-Simon, L. Belabed, G. Le Naour, J. Marc, F. Capron, [99] K. Blecher, L. R. Martinez, C. Tuckman-Vernon, P. Nacharaju,
L. Cynober, S. Darquy, Am. J. Physiol.: Regul., Integr. Comp. Physiol. D. Schairer, J. Chouake, J. M. Friedman, A. Alfieri, C. Guha,
2012, 303, R1053. J. D. Nosanchuk, A. Friedman, Nanomedicine 2012, 8, 1364.
[67] D. G. Armstrong, J. R. Hanft, V. R. Driver, A. P. S. Smith, [100] G. Han, L. N. Nguyen, C. MacHerla, Y. Chi, J. M. Friedman,
J. L. Lazaro-Martinez, A. M. Reyzelman, G. J. Furst, D. J. Vayser, J. D. Nosanchuk, L. R. Martinez, Am. J. Pathol. 2012, 180, 1465.
H. L. Cervantes, R. J. Snyder, M. Moore, P. May, J. Nelson, [101] A. D. Ormerod, P. C. Van Voorst Vader, S. Majewski,
G. Baggs, A. Voss, Diabetic Med. 2014, 31, 1069. W. Vanscheidt, N. Benjamin, W. Van Der Meijden, JAMA Dermatol.
[68] J. D. Shearer, J. R. Richards, C. D. Mills, M. D. Caldwell, Am. J. 2015, 151, 854.
Physiol. 1997, 272, 181. [102] P. G. Wang, M. Xian, X. Tang, X. Wu, Z. Wen, T. Cai, A. J. Janczuk,
[69] M. Varedi, Z. Akbari, G. A. Dehghani, S. Z. Tabei, Iran. J. Basic Chem. Rev. 2002, 102, 1091.
Med. Sci. 2009, 12, 173. [103] R. J. Singh, N. Hogg, J. Joseph, B. Kalyanaraman, J. Biol. Chem.
[70] F. Reesi, M. Minaiyan, A. Taheri, Drug Delivery Transl. Res. 2018, 8, 1996, 271, 18596.
111. [104] D. L. H. Williams, Acc. Chem. Res. 1999, 32, 869.
[71] Y. Liu, S. Zhou, Y. Gao, Y. Zhai, Asian J. Pharm. Sci. 2018, 4, 1. [105] A. P. Dicks, D. L. H. Williams, Chem. Biol. 1996, 3, 655.
[72] A. Schwentker, Y. Vodovotz, R. Weller, T. R. Billiar, Nitric Oxide [106] S. Catalani, M. Paganelli, M. E. Gilberti, L. Rozzini, F. Lanfranchi,
2002, 7, 1. A. Padovani, P. Apostoli, J. Trace Elem. Med. Biol. 2018, 45, 176.
[73] T. Kitano, H. Yamada, M. Kida, Y. Okada, S. Saika, M. Yoshida, [107] J. A. Hrabie, L. K. Keefer, Chem. Rev. 2002, 102, 1135.
Dermatol. Res. Pract. 2017, 2017, 1. [108] L. K. Keefer, ACS Chem. Biol. 2011, 6, 1147.
[74] J. L. Viñas, A. Sola, M. Genescà, V. Alfaro, F. Pí, G. Hotter, [109] J. A. Hrabie, J. R. Klose, D. A. Wink, L. K. Keefer, J. Org. Chem.
Free Radical Biol. Med. 2006, 40, 992. 1993, 58, 1472.
[75] K. Yamasaki, H. D. J. Edington, C. McClosky, E. Tzeng, A. Lizonova, [110] M. J. R. Ahonen, D. J. Suchyta, H. Zhu, M. H. Schoenfisch,
I. Kovesdi, D. L. Steed, T. R. Billiar, J. Clin. Invest. 1998, 101, 967. Biomacromolecules 2018, 19, 1189.
[76] A. Karamercan, S. Ercan, S. Bozkurt, Curr. Ther. Res. 2006, 67, 378. [111] E. M. Hetrick, J. H. Shin, N. A. Stasko, C. B. Johnson, D. A. Wespe,
[77] C. C. Miller, M. K. Miller, A. Ghaffari, B. Kunimoto, J. Cutaneous E. Holmuhamedov, M. H. Schoenfisch, ACS Nano 2008, 2, 235.
Med. Surg. 2004, 8, 233. [112] D. A. Riccio, M. H. Schoenfisch, Chem. Soc. Rev. 2012, 41, 3731.
[78] A. Moeen Rezakhanlou, C. Miller, B. McMullin, A. Ghaffari, [113] Y. Li, P. I. Lee, Mol. Pharmaceutics 2010, 7, 254.
R. Garcia, A. Ghahary, Cell Biol. Int. 2011, 35, 407. [114] C. Vogt, Q. Xing, W. He, B. Li, M. C. Frost, F. Zhao,
[79] A. Ghaffari, R. Jalili, M. Ghaffari, C. C. Miller, A. Ghahary, Wound Biomacromolecules 2013, 14, 2521.
Repair Regener. 2007, 15, 368. [115] M. P. Murphy, Biochim. Biophys. Acta, Bioenerg. 1999, 1411, 401.

Adv. Healthcare Mater. 2019, 8, 1801210 1801210  (17 of 18) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

[116] J. H. J. O. Kim, J. K. Noh, R. K. Thapa, N. Hasan, M. Choi, [134] C. J. Doillon, J. Biomater. Appl. 1987, 2, 562.
J. H. J. O. Kim, J. H. Lee, S. K. Ku, J. W. Yoo, Int. J. Biol. Macromol. [135] P. Zahedi, I. Rezaeian, S. O. Ranaei-Siadat, S. H. Jafari,
2015, 79, 217. P. Supaphol, Adv. Powder Technol. 2010, 21, 77.
[117] N. R. Sudarshan, D. G. Hoover, D. Knorr, Food Biotechnol. 1992, 6, [136] A. Koh, A. W. Carpenter, D. L. Slomberg, M. H. Schoenfisch,
257. ACS Appl. Mater. Interfaces 2013, 5, 7956.
[118] R. Singh, K. Shitiz, A. Singh, Int. Wound J. 2017, 14, 1276. [137] P. N. Coneski, J. A. Nash, M. H. Schoenfisch, ACS Appl. Mater.
[119] J. Koehler, F. P. Brandl, A. M. Goepferich, Eur. Polym. J. 2018, 100, Interfaces 2011, 3, 426.
1. [138] B. V. Worley, R. J. Soto, P. C. Kinsley, M. H. Schoenfisch,
[120] S. M. Shishido, A. B. Seabra, W. Loh, M. G. De Oliveira, ACS Biomater. Sci. Eng. 2016, 2, 426.
Biomaterials 2003, 24, 3543. [139] A. Lowe, J. Bills, R. Verma, L. Lavery, K. Davis, K. J. Balkus, Acta
[121] A. B. Seabra, A. Fitzpatrick, J. Paul, M. G. De Oliveira, R. Weller, Biomater. 2015, 13, 121.
Br. J. Dermatol. 2004, 151, 977. [140] Y. Kang, J. Kim, Y. M. Lee, S. Im, H. Park, W. J. Kim, J. Controlled
[122] A. B. Seabra, E. Pankotai, M. Fehér, Á. Somlai, L. Kiss, L. Bíró, Release 2015, 220, 624.
C. Szabó, M. Kollai, M. G. De Oliveira, Z. Lacza, Br. J. Dermatol. [141] M. Neidrauer, U. K. Ercan, A. Bhattacharyya, J. Samuels, J. Sedlak,
2007, 156, 814. R. Trikha, K. A. Barbee, M. S. Weingarten, S. G. Joshi, J. Med.
[123] T. P. Amadeu, A. B. Seabra, M. G. de Oliveira, A. Monte-Alto-Costa, Microbiol. 2014, 63, 203.
J. Surg. Res. 2008, 149, 84. [142] Y. Lu, D. L. Slomberg, B. Sun, M. H. Schoenfisch, Small 2013, 9,
[124] F. S. Schanuel, K. S. Raggio Santos, A. Monte-Alto-Costa, 2189.
M. G. de Oliveira, Colloids Surf., B 2015, 130, 182. [143] C. J. Backlund, B. V. Worley, A. R. Sergesketter, M. H. Schoenfisch,
[125] G. M. Annich, J. P. Meinhardt, K. A. Mowery, B. A. Ashton, J. Dent. Res. 2015, 94, 1092.
S. I. Merz, R. B. Hirschl, M. E. Meyerhoff, R. H. Bartlett, Crit. Care [144] T. P. Amadeu, A. B. Seabra, M. G. de Oliveira, A. M. A. Costa,
Med. 2000, 28, 915. J. Eur. Acad. Dermatol. Venereol. 2007, 21, 629.
[126] E. M. Hetrick, J. H. Shin, H. S. Paul, M. H. Schoenfisch, [145] E. M. Hetrick, M. H. Schoenfisch, Annu. Rev. Anal. Chem. 2009, 2,
Biomaterials 2009, 30, 2782. 409.
[127] M. Shabani, S. K. Pulfer, J. P. Bulgrin, D. J. Smith, Wound Repair [146] R. A. Hunter, B. J. Privett, W. H. Henley, E. R. Breed,
Regener. 1996, 4, 353. Z. Liang, R. Mittal, B. P. Yoseph, J. E. McDunn, E. M. Burd,
[128] J. A. Bauer, W. Rao, D. J. Smith, Wound Repair Regener. 1998, 6, C. M. Coopersmith, J. M. Ramsey, M. H. Schoenfisch, Anal. Chem.
569. 2013, 85, 6066.
[129] H. Nurhasni, J. Cao, M. Choi, I. Kim, B. L. Lee, Y. Jung, J.-W. Yoo, [147] H. D. Zomer, A. G. Trentin, J. Dermatol. Sci. 2018, 90, 3.
Int. J. Nanomed. 2015, 10, 3065. [148] T. P. Sullivan, W. H. Eaglstein, S. C. Davis, P. Mertz, Wound Repair
[130] K. S. B. Masters, S. J. Leibovich, P. Belem, J. L. West, Regener. 2001, 9, 66.
L. A. Poole-Warren, Wound Repair Regener. 2002, 10, 286. [149] A. J. F. King, Br. J. Pharmacol. 2012, 166, 877.
[131] E. J. Brisbois, J. Bayliss, J. Wu, T. C. Major, C. Xi, S. C. Wang, [150] D. A. Bellinger, E. P. Merricks, T. C. Nichols, ILAR J. 2006, 47,
R. H. Bartlett, H. Handa, M. E. Meyerhoff, Acta Biomater. 2014, 10, 243.
4136. [151] M. E. Spurlock, N. K. Gabler, J. Nutr. 2008, 138, 397.
[132] M. Liu, X.-P. Duan, Y.-M. Li, D.-P. Yang, Y.-Z. Long, Mater. Sci. [152] J. Rich, J. C. Lee, Diabetes 2005, 54, 2904.
Eng., C 2017, 76, 1413. [153] M. B. Witte, A. Barbul, Surg. Clin. North Am. 1997, 77, 509.
[133] N. Bhardwaj, S. C. Kundu, Biotechnol. Adv. 2010, 28, 325. [154] L. N. Kasiewicz, K. A. Whitehead, Biomater. Sci. 2017, 5, 1962.

Adv. Healthcare Mater. 2019, 8, 1801210 1801210  (18 of 18) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim

You might also like