Skip to main content
Physiological Genomics logoLink to Physiological Genomics
. 2016 Oct 7;48(12):889–896. doi: 10.1152/physiolgenomics.00066.2016

Reactive oxygen species and bacterial biofilms in diabetic wound healing

Aksone Nouvong 1,2, Aaron M Ambrus 3,4, Ellen R Zhang 3,4, Lucas Hultman 2, Hilary A Coller 3,4,
PMCID: PMC5206388  PMID: 27764766

Abstract

Chronic wounds are a common and debilitating complication for the diabetic population. It is challenging to study the development of chronic wounds in human patients; by the time it is clear that a wound is chronic, the early phases of wound healing have passed and can no longer be studied. Because of this limitation, mouse models have been employed to better understand the early phases of chronic wound formation. In the past few years, a series of reports have highlighted the importance of reactive oxygen species and bacterial biofilms in the development of chronic wounds in diabetics. We review these recent findings and discuss mouse models that are being utilized to enhance our understanding of these potentially important contributors to chronic wound formation in diabetic patients.

Keywords: wounds, diabetes, reactive oxygen species, biofilm

Chronic Wounds Are a Major Public Health Concern

Chronic wounds are wounds that do not follow the normal healing progression and persist, usually for more than 3 mo. Chronic wounds are a burden for both affected patients and the healthcare system. Most chronic wounds can be assigned to one of three categories: diabetic foot ulcers, ulcers resulting from arterial or venous insufficiency, or pressure ulcers (56). We focus here on diabetic foot ulcers, which are the most common cause of nontraumatic major amputation around the world and the most costly type of wound (25, 61, 67, 84). An estimated 67% of amputations occur in diabetic patients (73), and diabetic foot ulcers precede an estimated 80% of diabetic lower extremity amputations (32). Today, 382 million people have been diagnosed with diabetes, and 25% of these patients will develop a foot ulcer in their lifetime (32, 69). One-quarter of these ulcers will not heal, and up to 28% will result in an amputation (32, 69). It is estimated that an amputation due to diabetes occurs every 30 s worldwide (26). If a chronic wound leads to amputation, the prognosis for the patient is poor. Five-year mortality rates after the development of a foot ulcer are between 43 and 55%, which is higher than some common cancers like melanoma, lymphoma, and cancer of the testis, breast, prostate, uterus, larynx, and cervix (65, 70). Furthermore, the numbers of patients with diabetic foot ulcers are increasing. By 2025, more than 500 million people are expected to have diabetes, 125 million diabetics will develop foot ulcers, and 20 million diabetics will undergo amputations (18).

Patients with lower limb chronic wounds can experience impaired mobility, time lost from work, and adverse effects on finances. In a study assessing the psychological implications of leg ulcers, 68% of affected patients reported a negative emotional impact on their lives, including feelings of fear, social isolation, anger, depression, and negative self-image (64). In addition to the effects on individual patients, diabetic foot ulcers are also a significant drain on healthcare funds. In the US alone, the cost for the treatment of foot diseases in patients with diabetes is at least $6 billion annually (32). For all of these reasons, chronic diabetic wounds are an important public health problem.

Normal Wound Healing

On a cellular level, chronic wounds occur when the normal wound healing process is derailed. A complex series of events involving many cell types (neutrophils, monocytes, endothelial cells, keratinocytes, and fibroblasts) is activated by the presence of an acute wound (30, 77). The stages of wound healing are hemostasis, inflammation, proliferation, and remodeling. The first stage is to re-establish hemostasis at the wound site by formation of a network of fibrin fibrils generated via thrombin-mediated cleavage of fibrinogen (78). In the next stage, neutrophils are recruited to the wound site to destroy pathogenic organisms. Macrophages arrive at the wound site and engulf debris and dead cells. This inflammatory phase ends when inflammatory cells are actively removed, likely via apoptosis. In the following phase, keratinocytes proliferate to reform the epidermis. Fibroblasts migrate to the wound site and are stimulated to proliferate by platelet-derived growth factor (PDGF) and other growth factors (16, 33, 83, 92). During this phase, fibroblasts and their derived myofibroblasts break down the fibrin clots by secreting plasminogen activators that convert plasminogen to plasmin (49, 96), which cleaves fibrin to dissolve the clot. Fibroblasts and myofibroblasts also secrete collagen and other extracellular matrix proteins that form granulation tissue. The formation of granulation tissue is accompanied by the development of new blood vessels, re-epithelialization of the wound, and contraction of the granulation tissue to bring the edges of the wound closer together (2, 30, 34, 74, 92). In a final phase, fibroblasts, macrophages and endothelial cells secrete extracellular matrix proteins and matrix metalloproteinases that remodel the granulation tissue, replacing the collagen type III matrix with a stronger, more organized collagen type I matrix (19).

Chronic Wounds Fail to Progress through Timely Repair

While normal wound healing proceeds via a well-orchestrated series of events moving through the four sequential, temporary, and overlapping phases of hemostasis, inflammation, proliferation, and remodeling, chronic wounds stall before reaching the later phases. It has not been straightforward to define chronic wounds in humans or mice. In 1994, Lazarus et al. (43) defined a chronic wound as one that fails to progress through a timely sequence of repair. Chronic wounds have also been defined as wounds that exhibit defective regulation of one or more steps in the wound healing process (46, 56). In particular, it has been noted that chronic wounds often stall in the inflammatory phase (46, 56). Other definitions of chronic wounds focus on the amount of time they require to heal. In humans, wounds are considered chronic if they have not healed for 3 mo (46). In mice, some argue that if a wound fails to heal by 26 days, it should be considered chronic (5, 18).

Challenges in Studying Chronic Wounds

When chronic wounds are the subject of investigation, researchers face significant challenges. Different types of wounds in diabetics may have different etiologies. Determining the root cause of a wound is complex because the patients often have multiple different comorbidities. Another major challenge in wound healing research is the difficulty of obtaining samples from the early stages of healing in a chronic wound in humans. When a patient presents for treatment of a chronic wound, the early phases of its wound healing have passed and can no longer be studied.

For these reasons, some researchers have turned to mouse models to complement studies on wound healing in humans. Use of genetically inbred mice can provide a large number of subjects with a consistent genotype for studying wound healing that is unachievable in humans. The ability to manipulate the mouse genome provides opportunities to study the effects of a single gene on wound healing. Furthermore, with mice, the early events after a wound is introduced can be studied under precise temporal control, with sufficient numbers of mice to draw statistically significant conclusions.

It is important to note that mouse models also have significant limitations. Mouse models do not accurately simulate many aspects of the condition in diabetic wounds of humans. In general, animal models are limited in their ability to simulate processes that are associated with aging, which is a major factor in chronic wounds in humans. For chronic wounds, characteristics of aging such as neuropathy, peripheral arterial disease, or venous insufficiency (54, 56) are likely to play an important role and are not well simulated in current mouse models. In addition, the wound healing process is different in mice than humans. Mouse skin has a thin muscle layer, the panniculus carnosus layer, under the dermis. In humans, and other higher primates, there are only remnants of panniculus carnosus muscle, primarily in the neck where it is called the platysma muscle. After injury in a mouse, this muscle layer produces rapid wound contraction that promotes healing (100). In humans, wounds heal through re-epithelialization and granulation tissue formation without the benefit of contraction (54). In humans, contraction is slow and incomplete. For these reasons, mouse models of wound healing have limitations in their ability to provide insight into the human condition. We describe here recent studies using both human and mouse models designed to better understand the distinction between chronic and acute wounds.

Chronic Wounds Are Stalled in the Inflammatory Phase

Chronic wounds have been characterized as being stalled in the inflammatory phase of wound healing, which is based on the observation of higher levels of certain types of immune cells. In one study, patients with both venous insufficiency and diabetic chronic wounds had a lower ratio of CD4+ to CD8+ T cells due to low levels of CD4+ T cells, higher numbers of macrophages, and more B cells and plasma cells than patients with properly healing acute wounds (45). Chronic wounds were characterized by prolonged and persistent T cell and macrophage infiltration (45). In another study of nonhealing venous leg ulcers, inflammatory cells, B cells, and macrophages were present in high numbers (98). In yet another study, Rosner and colleagues (72) found a statistically significant increase in the mean numbers of macrophages and neutrophils from the distant area toward the center of the ulcer in venous leg ulcers. At the center and the edge of the ulcer, macrophages were abundant, comprising 63% (center) and 53% (edge) of the cells. In the distant area, T cells were predominant and were largely CD8+ (72). B cells were rare (72). One limitation of this study was that wounds healing normally were not included. Moore and colleagues (52), in another study, found that at the wound margin of chronic wounds, CD45+ leucocytes were largely lymphocytes, and T lymphocytes were more prevalent than B lymphocytes. These investigators also found CD68+ macrophages present in all layers of the dermis at the wound margin, but found that these macrophages lacked activation markers CD16 (Fc γ III receptor) and CD35 (C3b receptor) (52). In this study as well, no normally healing wounds were included for comparison. The reason for the increased infiltration of immune cells into chronic wounds is not clear, but chronic wounds have also been found to have higher levels of proinflammatory growth factors and cytokines (3, 24, 27, 82, 89) that may recruit these inflammatory cells. These inflammatory factors and cytokines have been hypothesized to reflect complications from metabolic dysfunction in diabetic patients (20).

Chronic Wounds Have Higher Levels of Proteinases

Chronic wounds often contain an accumulation of devitalized tissue (7), which may reflect higher levels of proteases such as matrix metalloproteinases and gelatinases that degrade extracellular matrix proteins (101), as well as lower levels of the molecules that inhibit these enzymes (11, 24, 44, 59, 101). Elevated levels of matrix metalloproteinase activity can contribute to the degradation of the extracellular matrix, growth factors, and their receptors (7, 42, 90, 97).

Chronic Wounds Contain Senescent Fibroblasts

Another characteristic of chronic wounds is poor infiltration of fibroblasts and blood vessels into the wound area (19). The fibroblasts that are present in venous ulcers have been reported to display the morphological characteristics of senescent cells (19, 24, 50, 51, 56). The granulation tissue in chronic wounds usually contains fewer fibroblasts and less collagen than in wounds that heal well (41, 48, 86). Extracellular matrix deposition and remodeling are often compromised (19). Fibroblasts isolated from chronic wounds have been shown to have diminished migratory capacity (7), reduced proliferative capacity in response to stimulation with PDGF (1), diminished responsiveness to transforming growth factor (TGF)-β (60), and altered production of extracellular matrix proteins (23). Thus, the fitness and function of fibroblasts in chronic wounds may be jeopardized, or the wound environment may affect their altered ability to coordinate wound healing.

Treatments for Chronic Wounds

There are several options for treatment of diabetic foot ulcers. Standard of care includes the debridement of necrotic tissue [a process in which necrotic and senescent tissue as well as foreign and infected material are removed from the wound, and healthy tissue is cut to stimulate the healing process (7)], mechanical off-loading of pressure areas (66, 95), improving the regulation of blood glucose, infection control, wound dressings that promote a moist environment to assist healing (95), and revascularization. When wounds do not respond to standard care, advanced wound care modalities can be incorporated into the treatment plans. Some of these treatments involve reintroducing peptide growth factors that promote wound healing. Recombinant PDGF (79, 94), EGF, VEGF, and TGF-β have been reported to promote wound healing (87). Other treatment products are designed to reduce the high levels of matrix metalloproteinases that are thought to inhibit healing (40, 76, 85). Biological skin equivalents are also available, and these can take the form of an acellular scaffold that provides a matrix for cells, a combination of cells and a scaffold, or skin equivalents that are created from single-layered or bilayered cultured skin constructs, or membranes from other parts of the body such as the amnion (21, 47, 68, 88, 103). Despite the availability of multiple different wound healing treatments, many patients do not heal, making improvements in our management of wounds a critical public health need.

Elevated Reactive Oxygen Species in Diabetic Wounds

In the last decade, there has been increased attention to the possibility that reactive oxygen species (ROS) are part of the mechanism through which some wounds exhibit a delay in healing. In this context, ROS include radical and nonradical molecules that can be formed from oxygen, such as superoxide anion, hydroxyl radicals, singlet oxygen, and hydrogen peroxide. A hypothesis that excessive ROS can delay wound healing was proposed in 2005 for venous leg ulcers (98). The argument was based on the importance of neutrophils for defense against bacteria. In the initial response to a fresh wound, neutrophils release an oxidative burst of ROS generated by the plasma membrane-localized NADPH oxidase complex. Wlaschek and Scharffetter-Kochanek (98) reasoned that patients with venous ulcers exhibit an extended inflammatory phase with more neutrophils in the region for longer periods of time than individuals with rapidly healing wounds. They hypothesized that excess neutrophils and macrophages present in chronic wounds would result in excess levels of ROS. The result, they argued, would be an imbalance between the pro-oxidants and antioxidants in the wound, which leads to oxidative stress (98). Some studies have provided data consistent with this hypothesis. Superoxide released by neutrophils in chronic wounds has been reported to be increased 170% compared with acute wounds (98). Elevated ROS in the wound environment has been hypothesized to lead to perpetuation of inflammation, activation of proteolytic pathways, and tissue damage (98).

Because ROS are difficult to measure, some researchers have measured the byproducts of the reaction of ROS with more stable molecules to assess ROS levels in chronic wounds. Levels of allantoin and uric acid, stable oxidation products, were fivefold higher in fluid from chronic wounds than fluid from acute wounds (36). Measurement of an oxidized prostaglandin, 8-isoprostane, revealed higher levels of 8-isoprostane in fluid from chronic wounds than acute wound fluid (102). Patients with chronic wounds have also been shown to have low levels of the antioxidant molecules that are important for detoxifying ROS. In particular, chronic wounds have been reported to have lower levels of natural antioxidants vitamins A and E (71) and glutathione (53). These data support the notion that there is a high level of ROS in diabetic chronic wounds.

Physiological Effects of High ROS

Elevated levels of ROS in chronic wounds may have one of many deleterious effects. ROS have been reported to damage endothelial cells, thus preventing angiogenesis necessary for proper wound healing (28). ROS have also been reported to induce upregulation of ICAM-1, which results in extravasation of activated leukocytes (28). Elevated levels of ROS have also been reported to inhibit keratinocyte migration and thus delay re-epithelialization of wounds (57). In addition, ROS can inactivate the tissue inhibitors of metalloproteinases and thereby promote a highly proteolytic microenvironment in chronic wounds (81). In fibroblasts, matrix metalloproteinase expression is induced after exposure to singlet oxygen (99), hydrogen peroxide (8, 91), or hydroxyl radicals (9).

Chronic Wounds Are Commonly Infected with Bacteria

Further contributing to the altered microenvironment in chronic wounds is the fact that they are frequently colonized by bacterial species that impair wound healing (6, 35). Some of these species can form biofilms, which are complex microenvironments consisting of bacteria and polymers the bacteria secrete such as polysaccharides, proteins, lipids, and nucleic acids (22). The bacteria within biofilms are notoriously resistant to antibiotics (22, 80, 104). Studies in human patients have shown that chronic wounds are more likely to contain biofilms than acute wounds. In one study, James and colleagues (35) found that 60% of the chronic wounds studied contained biofilm-forming bacteria, compared with just 6% of acute wounds. Bacterial biofilms in the chronic wounds were often found to be characterized by diverse microbial communities including anaerobic bacteria. In another study, Bjarnsholt and colleagues (4) proposed a similar hypothesis that chronic wounds result from a failure to eradicate pathogens. They analyzed chronic wounds and found microcolonies with the capacity to become biofilms. They hypothesized that formation of biofilms protects these bacteria from being killed by neutrophils in chronic wounds. Subsequent studies have continued to find that wound debridement samples contain high levels of bacteria, with Staphylococcus aureus being the most common type (58). The exact role of biofilms in chronic wounds is not yet clear. One study found that biofilm-forming bacteria and soluble products from these bacteria reduce keratinocyte viability in vitro and decrease the ability of keratinocytes to close an in vitro wound (39), suggesting one possible mechanism for their association with chronic wounds.

db/db Mouse as a Model for Wound Healing in Diabetics

Challenges associated with obtaining early chronic wound samples from human diabetic patients have led to the creation of mouse models as a tool to better understand the early process of chronic wound formation. One frequently used model is the db/db mouse. Db/db mice have an inactivating mutation in the gene encoding the leptin receptor (13). These mice develop insulin resistance, obesity, and diabetes with hyperglycemia (13, 14).

Wound healing in db/db mice is delayed compared with wild-type controls (93). Normal healing in wild-type mice is associated with an increase and a subsequent decrease in the levels of the neutrophil chemoattractant macrophage inflammatory protein (MIP)-2 and the macrophage chemoattractant monocyte chemoattractant protein (MCP)-1. In contrast, db/db mice have a large and more sustained increase in the levels of both MIP-2 and MCP-1, which results in elevated numbers of polymorphonuclear cells and macrophages in the wound tissue (93). This sustained cytokine release may contribute to the delayed wound healing observed in these animals (93).

It should be noted that the db/db mouse has significant drawbacks as a model to simulate human diabetes. Human diabetics do not typically have a mutation in the leptin receptor, and therefore the genetic basis for the diabetic phenotype in these mice may be different from that in humans. In addition, the db/db mice are obese. If wounds are introduced without being splinted, an inconsistent expansion of the wounds can occur due to the altered skin tension. A rabbit model for diabetes also exists in which islet cells die in response to treatment with streptozotocin, simulating Type 1 diabetes. However, since Type 2 diabetes is much more common in human patients, findings in a rabbit model of Type 1 diabetes may not be relevant for the much larger human population of Type 2 diabetic patients.

High Levels of ROS and Biofilms in the db/db Mouse Model

The db/db mouse model has been used to study in more depth the role of ROS and biofilms in delayed wound healing in diabetics. Mudge and colleagues (53) found that wounds introduced into a db/db mouse have low levels of the antioxidant glutathione compared with wounds introduced into control mice. Furthermore, treating wounds in db/db mice with glutathione resulted in significantly expedited wound healing, demonstrating the critical role of glutathione and antioxidants in determining wound healing rates in mice with simulated diabetes (53).

Dhall et al. (18) of the Martins-Green laboratory found that superoxide dismutase (SOD) activity and levels of hydrogen peroxide (the product of SOD) are higher in db/db mice compared with controls. The activities of catalase and glutathione peroxidase, antioxidant enzymes that convert hydrogen peroxide to water, were lower in db/db mice than controls (18). The results are consistent with the presence of oxidative stress in db/db mice.

The Martins-Green laboratory (18) went on to inhibit catalase and glutathione peroxidase and thereby create high levels of ROS in the wounds. This treatment caused the wounds in db/db mice to form exudate and become chronic, in some cases lasting 100 days. db/db mice treated with inhibitors of catalase and glutathione peroxidase were more likely to develop spontaneous microbial infections. Microbial communities in wounds in db/db mice evolved from containing species that do not produce biofilms to containing biofilm-producing species (18). Treating the wounds with antioxidants, α-tocopherol and N-acetylcysteine, improved wound healing dramatically (18). When the wounds were treated with antioxidants, the bacteria in the wounds were less likely to generate biofilms and were more sensitive to antibiotic treatment. Granulation tissue was formed with thicker collagen fibers (18). The bacteria in the db/db mice treated with antioxidant inhibitors develop complex communities of bacteria including bacteria that are similar to those in human skin such as Staphylococcus epidermidis and other coagulase-negative staphylococci (29). The results support an important role for a combination of bacteria that create biofilms and ROS in promoting wounds that fail to heal in diabetic mice.

ROS and Biofilms in LIGHT−/− Mice

The laboratory of Dr. Martins-Green also reached a similar conclusion based on studies in a different model system of delayed wound healing. Petreaca et al. (63) of the Martins-Green laboratory discovered that VEGF induces macrophage apoptosis via a tumor necrosis factor superfamily member called TNFSF14 or LIGHT (Homologous to Lymphotoxins, exhibits Inducible expression, and competes with herpes simplex virus Glycoprotein D for Herpes virus entry mediator, a receptor expressed by T lymphocytes). Inhibition of LIGHT with an anti-LIGHT neutralizing antibody prevented VEGF-induced apoptotic death of cultured macrophages. The authors hypothesized that LIGHT promotes macrophage apoptosis during wound healing. They further hypothesized that mice with genetic inactivation of LIGHT would not be able to execute the series of events that normally occur during wound healing and would experience chronic wounds. To test their hypotheses, the authors generated genetically engineered mice with inactivation of TNFSF14/LIGHT (62). Wounds introduced into LIGHT−/− mice do exhibit several characteristics that are similar to those of chronic wounds in human patients. The LIGHT−/− mice have higher production of cytokines than control mice, in particular CXCL8, MCP-1/CCL2, and IP-10/CXCL10, which are expected to recruit neutrophils, macrophages and T-lymphocytes. In wounds in LIGHT−/− mice, neutrophils appeared within the first few hours after wounding and the number of neutrophils was significantly elevated compared with wounds introduced into wild-type mice. Neutrophils remained within the wound until day 9 in wounds introduced into LIGHT−/− mice, while in wounds introduced in control mice, the levels of neutrophils had declined by day 3. Macrophages were also more abundant in wounds in LIGHT−/− mice, but only on day 7. T cells were consistently more abundant in the wounds from the LIGHT−/− mice. A subset of the wounds in the LIGHT−/− mice failed to heal even after a prolonged period of time. These wounds showed signs of infection; they became crusty, oozed fluids, and opened and closed repeatedly. Taken together, the data indicate that the LIGHT−/− model shares some similarities to the situation in chronic wounds in diabetic patients.

With this new mouse model in hand, Dhall et al. (17) of the Martin-Greens laboratory found that wounds developed in LIGHT−/− mice have higher levels of hydrogen peroxide than wounds created in wild-type mice. These higher levels of reactive species in wounds generated in LIGHT−/− mice were associated with increased levels of oxidized macromolecules such as lipid peroxides, and increased apoptosis and necrosis in the LIGHT−/− mice compared with wounds in wild-type controls.

The authors tested the importance of ROS for wound healing by inhibiting antioxidant enzymes and introducing biofilm-forming bacteria (17). Wounds were reproducibly converted to a chronic state in LIGHT−/− mice by a combination of inhibition of the antioxidant enzymes catalase and glutathione peroxidase immediately postwounding, combined with subsequent application of the biofilm forming bacterium S. epidermidis. If any one of these three components was eliminated, the wounds did not consistently convert to a chronic state. Thus, the LIGHT−/− mouse model, like the db/db model, supports a role for a combination of high ROS and the presence of biofilm-producing bacteria in the development of chronic wounds.

Limitations of the Mouse Models

The availability of this new mouse model raises the question whether the model is likely to be informative about the wound healing process in diabetic patients. It is important to note that all mouse models are limited in their ability to model the human condition. In this case, the mice have a genetic abnormality in LIGHT rather than diabetes and, therefore, do not have some of the important characteristics of diabetic patients. Many diabetic patients are overweight. Because the LIGHT−/− mice are normal weight, they will not simulate some important aspects of diabetes. Diabetic patients also have high levels of glucose in their blood, and the LIGHT−/− mice do not share this property. This can be a critically important difference because hyperglycemia itself can lead to abnormal leucocyte function, such as decreased phagocytosis (37). Additionally, while LIGHT expression was inhibited in this mouse model, recent studies have shown that plasma levels of LIGHT are actually increased in Type 2 diabetes and obesity (15, 31). Also of importance, the wounds that are generated in the db/db and LIGHT−/− mice are on the dorsum, a position in which they will not experience pressure. These wounds will therefore not recapitulate the situation for most diabetic ulcers that are largely on the patients' legs and feet and will consequently bear weight. Whether a wound is on a portion of the body that bears weight is likely an important determinant of healing rates, as relieving the pressure from chronic wounds is an effective approach to therapy (66, 95).

ROS and Biofilms in Diabetic Mice: an Alternative Model of Low Levels of ROS

In addition to the db/db and LIGHT−/− models, other models for diabetic wound healing are also being put forward. The TallyHo mouse has been developed to study diabetes in mice. The TallyHo mouse has polygenic mutations that cause hyperglycemia, hyperinsulinemia, weight gain, and impaired glucose tolerance (38). Because diabetes in human patients is mostly polygenic, the TallyHo mouse model is considered more similar to human Type 2 diabetes than models of streptozotocin-induced diabetes or diabetes induced by a single mutation such as db/db (38). The TallyHo mouse has been demonstrated to exhibit delayed wound healing for incisional wounds, excisional wounds, and ischemia-reperfusion injury (10).

Nguyen and colleagues (55) studied the Tallyho model for mouse diabetes. They introduced S. aureus into wounds in C57BL/6 control mice and TallyHo mice and found the TallyHo mice had higher levels of bacteria than the controls. The TallyHo wounds into which S. aureus had been introduced, but not wounds in control mice with introduced S. aureus, became purulent and inflamed. The presence of S. aureus delayed wound healing for both diabetic and nondiabetic mice, but the effect was especially strong in the diabetic mice. The authors monitored myeloperoxidase activity as an indicator of the neutrophil oxidative burst. They found that sterile wounds from diabetic and wild-type mice showed modest myeloperoxidase activity. Inoculated wounds in wild-type mice had increased myeloperoxidase activity compared with noninoculated controls, while the induction in inoculated, diabetic mice was lower than in the control mice. The lack of neutrophil oxidative burst may be an important contributor to the increased bacterial burden in the diabetic wounds. The authors conclude, in contrast to the studies from the Martins-Green laboratory described above, that an impaired ability to respond to bacteria with an oxidative burst represents a potential mechanism underlying chronic wounds in diabetics.

Outstanding Questions for the Field

These studies raise numerous important questions for future research. One future question will be whether the LIGHT−/− model can be valuable for understanding chronic wounds in diabetic patients, given that the LIGHT gene is not directly related to diabetes and LIGHT levels are actually high in diabetics. Furthermore, the most important downstream regulators of the phenotype in the LIGHT−/− mice are not known, as in addition to a reported effect on macrophage apoptosis, according to the authors, LIGHT−/− mice also have increased levels of forkhead box protein A1 (FOXA1), cytochrome p450 2E1 (CYP2E1), and Toll-like receptor 6 (TLR6) (17). More generally, can mouse models that result in immune dysregulation and a stall in the inflammatory phase of wound healing be a valuable tool for understanding the processes that occur in diabetic patients?

Another important outstanding question for the field will be to determine which model of ROS in diabetic foot ulcers is more relevant. Is there an increase in harmful ROS in the wounded diabetic foot as a result of the higher levels of immune cells with the potential to generate ROS? Or alternatively, do high levels of glucose in diabetic patients inhibit the ability of immune cells in diabetic chronic wounds to become activated and perform a ROS burst, thus leading to lower levels of ROS and a failure to clear pathogens? Or, do each of these models occur in different subsets of patients?

Additional open questions for the field involve the development of bacterial biofilms. For instance, do these biofilms inhibit wound healing and if so, how? Are there strategies to limit biofilm production that can benefit patients with biofilm-containing chronic wounds? Would lowering ROS levels with antioxidants protect against biofilm formation in human patients? Or would lowering ROS allow bacteria to flourish and impede healing?

Finally, it will be important for the diabetes field to determine how we can best use mouse models to understand the course of wounds in diabetic patients. For instance, can mouse models be developed that allow scientists to rigorously investigate the role of weight, diet, or exercise in wound healing? The availability of additional mouse models that allow scientists to dissect the effects of different diabetic complications on wound healing could potentially develop into a valuable tool for researchers.

Given that up to 40% of diabetic patients die within 1 yr of amputation (12, 75), effective treatment and prevention modalities for patients with diabetic foot ulcers are desperately needed for an aging population. Improving our knowledge of the underlying biology of chronic wounds could result in evolved standards of care, potentially reducing the incidence of chronic wounds and their impact on patients and healthcare systems.

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the author(s).

AUTHOR CONTRIBUTIONS

A.N., A.M.A., E.R.Z., L.H., and H.A.C. edited and revised manuscript; A.N., A.M.A., E.R.Z., L.H., and H.A.C. approved final version of manuscript; H.A.C. drafted manuscript.

ACKNOWLEDGMENTS

H. A. Coller is a member of the UCLA Stem Cell Center, the Jonsson Comprehensive Cancer Center, the UCLA Molecular Biology Institute, and the UCLA Bioinformatics Interdepartmental Program.

REFERENCES

  • 1.Agren MS, Steenfos HH, Dabelsteen S, Hansen JB, Dabelsteen E. Proliferation and mitogenic response to PDGF-BB of fibroblasts isolated from chronic venous leg ulcers is ulcer-age dependent. J Invest Dermatol 112: 463–469, 1999. [DOI] [PubMed] [Google Scholar]
  • 2.Bainbridge P. Wound healing and the role of fibroblasts. J Wound Care 22: 407–408, 2013. [DOI] [PubMed] [Google Scholar]
  • 3.Beidler SK, Douillet CD, Berndt DF, Keagy BA, Rich PB, Marston WA. Inflammatory cytokine levels in chronic venous insufficiency ulcer tissue before and after compression therapy. J Vasc Surg 49: 1013–1020, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Bjarnsholt T, Kirketerp-Moller K, Jensen PO, Madsen KG, Phipps R, Krogfelt K, Hoiby N, Givskov M. Why chronic wounds will not heal: a novel hypothesis. Wound Repair Regen 16: 2–10, 2008. [DOI] [PubMed] [Google Scholar]
  • 5.Bonomo SR, Davidson JD, Tyrone JW, Lin X, Mustoe TA. Enhancement of wound healing by hyperbaric oxygen and transforming growth factor beta3 in a new chronic wound model in aged rabbits. Arch Surg 135: 1148–1153, 2000. [DOI] [PubMed] [Google Scholar]
  • 6.Bowler PG, Duerden BI, Armstrong DG. Wound microbiology and associated approaches to wound management. Clin Microbiol Rev 14: 244–269, 2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Brem H, Stojadinovic O, Diegelmann RF, Entero H, Lee B, Pastar I, Golinko M, Rosenberg H, Tomic-Canic M. Molecular markers in patients with chronic wounds to guide surgical debridement. Mol Med 13: 30–39, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Brenneisen P, Briviba K, Wlaschek M, Wenk J, Scharffetter-Kochanek K. Hydrogen peroxide (H2O2) increases the steady-state mRNA levels of collagenase/MMP-1 in human dermal fibroblasts. Free Radic Biol Med 22: 515–524, 1997. [DOI] [PubMed] [Google Scholar]
  • 9.Brenneisen P, Wenk J, Klotz LO, Wlaschek M, Briviba K, Krieg T, Sies H, Scharffetter-Kochanek K. Central role of ferrous/ferric iron in the ultraviolet B irradiation-mediated signaling pathway leading to increased interstitial collagenase [matrix-degrading metalloprotease (MMP)-1] and stromelysin-1 (MMP-3) mRNA levels in cultured human dermal fibroblasts. J Biol Chem 273: 5279–5287, 1998. [DOI] [PubMed] [Google Scholar]
  • 10.Buck DW 2nd, Jin DP, Geringer M, Hong SJ, Galiano RD, Mustoe TA. The TallyHo polygenic mouse model of diabetes: implications in wound healing. Plast Reconstr Surg 128: 427e–437e, 2011. [DOI] [PubMed] [Google Scholar]
  • 11.Bullen EC, Longaker MT, Updike DL, Benton R, Ladin D, Hou Z, Howard EW. Tissue inhibitor of metalloproteinases-1 is decreased and activated gelatinases are increased in chronic wounds. J Invest Dermatol 104: 236–240, 1995. [DOI] [PubMed] [Google Scholar]
  • 12.Cereceres S, Touchet T, Browning MB, Smith C, Rivera J, Hook M, Whitfield-Cargile C, Russell B, Cosgriff-Hernandez E. Chronic wound dressings based on collagen-mimetic proteins. Adv Wound Care 4: 444–456, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Chen H, Charlat O, Tartaglia LA, Woolf EA, Weng X, Ellis SJ, Lakey ND, Culpepper J, Moore KJ, Breitbart RE, Duyk GM, Tepper RI, Morgenstern JP. Evidence that the diabetes gene encodes the leptin receptor: identification of a mutation in the leptin receptor gene in db/db mice. Cell 84: 491–495, 1996. [DOI] [PubMed] [Google Scholar]
  • 14.Coleman DL. Diabetes-obesity syndromes in mice. Diabetes 31: 1–6, 1982. [DOI] [PubMed] [Google Scholar]
  • 15.Dandona P, Ghanim H, Monte SV, Caruana JA, Green K, Abuaysheh S, Lohano T, Schentag J, Dhindsa S, Chaudhuri A. Increase in the mediators of asthma in obesity and obesity with type 2 diabetes: reduction with weight loss. Obesity (Silver Spring) 22: 356–362, 2014. [DOI] [PubMed] [Google Scholar]
  • 16.De Donatis A, Ranaldi F, Cirri P. Reciprocal control of cell proliferation and migration. Cell Commun Signal 8: 20, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Dhall S, Do D, Garcia M, Wijesinghe DS, Brandon A, Kim J, Sanchez A, Lyubovitsky J, Gallagher S, Nothnagel EA, Chalfant CE, Patel RP, Schiller N, Martins-Green M. A novel model of chronic wounds: importance of redox imbalance and biofilm-forming bacteria for establishment of chronicity. PLos One 9: e109848, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Dhall S, Do DC, Garcia M, Kim J, Mirebrahim SH, Lyubovitsky J, Lonardi S, Nothnagel EA, Schiller N, Martins-Green M. Generating and reversing chronic wounds in diabetic mice by manipulating wound redox parameters. J Diabetes Res 2014: 562625, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Eming SA, Krieg T, Davidson JM. Inflammation in wound repair: molecular and cellular mechanisms. J Invest Dermatol 127: 514–525, 2007. [DOI] [PubMed] [Google Scholar]
  • 20.Falanga V. Wound healing and its impairment in the diabetic foot. Lancet 366: 1736–1743, 2005. [DOI] [PubMed] [Google Scholar]
  • 21.Fetterolf DE, Istwan NB, Stanziano GJ. An evaluation of healing metrics associated with commonly used advanced wound care products for the treatment of chronic diabetic foot ulcers. Manag Care 23: 31–38, 2014. [PubMed] [Google Scholar]
  • 22.Flemming HC, Wingender J. The biofilm matrix. Nat Rev Microbiol 8: 623–633, 2010. [DOI] [PubMed] [Google Scholar]
  • 23.Foy V, Li J, Kirsner R, Eaglstein W. Analysis of fibroblast defects in extracellular matrix production in chronic wounds. J Am Acad Dermatol 50: 168, 2004. [Google Scholar]
  • 24.Frykberg RG, Banks J. Challenges in the treatment of chronic wounds. Adv Wound Care 4: 560–582, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Frykberg RG, Habershaw GM, Chrzan JS. Epidemiology of the diabetic foot: ulcerations and amputations. In: Clinical Management of Diabetic Neuropathy, edited by Veves A. Humana, 1998, p. 273–290. [Google Scholar]
  • 26.Game FL, Hinchliffe RJ, Apelqvist J, Armstrong DG, Bakker K, Hartemann A, Londahl M, Price PE, Jeffcoate WJ. A systematic review of interventions to enhance the healing of chronic ulcers of the foot in diabetes. Diabetes Metab Res Rev 28, Suppl 1: 119–141, 2012. [DOI] [PubMed] [Google Scholar]
  • 27.Gohel MS, Windhaber RA, Tarlton JF, Whyman MR, Poskitt KR. The relationship between cytokine concentrations and wound healing in chronic venous ulceration. J Vasc Surg 48: 1272–1277, 2008. [DOI] [PubMed] [Google Scholar]
  • 28.Grether-Beck S, Olaizola-Horn S, Schmitt H, Grewe M, Jahnke A, Johnson JP, Briviba K, Sies H, Krutmann J. Activation of transcription factor AP-2 mediates UVA radiation- and singlet oxygen-induced expression of the human intercellular adhesion molecule 1 gene. Proc Natl Acad Sci USA 93: 14586–14591, 1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Grice EA, Segre JA. The skin microbiome. Nat Rev Microbiol 9: 244–253, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Gurtner GC, Werner S, Barrandon Y, Longaker MT. Wound repair and regeneration. Nature 453: 314–321, 2008. [DOI] [PubMed] [Google Scholar]
  • 31.Halvorsen B, Santilli F, Scholz H, Sahraoui A, Gulseth HL, Wium C, Lattanzio S, Formoso G, Di Fulvio P, Otterdal K, Retterstøl K, Holven KB, Gregersen I, Stavik B, Bjerkeli V, Michelsen AE, Ueland T, Liani R, Davi G, Aukrust P. LIGHT/TNFSF14 is increased in patients with type 2 diabetes mellitus and promotes islet cell dysfunction and endothelial cell inflammation in vitro. Diabetologia 59: 2134–2144, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Hingorani A, LaMuraglia GM, Henke P, Meissner MH, Loretz L, Zinszer KM, Driver VR, Frykberg R, Carman TL, Marston W, Mills JL Sr., Murad MH. The management of diabetic foot: a clinical practice guideline by the Society for Vascular Surgery in collaboration with the American Podiatric Medical Association and the Society for Vascular Medicine. J Vasc Surg 63: 3S–21S, 2016. [DOI] [PubMed] [Google Scholar]
  • 33.Hinz B, Phan SH, Thannickal VJ, Galli A, Bochaton-Piallat ML, Gabbiani G. The myofibroblast: one function, multiple origins. Am J Pathol 170: 1807–1816, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Iyer VR, Eisen MB, Ross DT, Schuler G, Moore T, Lee JC, Trent JM, Staudt LM, Hudson J Jr, Boguski MS, Lashkari D, Shalon D, Botstein D, Brown PO. The transcriptional program in the response of human fibroblasts to serum. Science 283: 83–87, 1999. [DOI] [PubMed] [Google Scholar]
  • 35.James GA, Swogger E, Wolcott R, Pulcini E, Secor P, Sestrich J, Costerton JW, Stewart PS. Biofilms in chronic wounds. Wound Repair Regen 16: 37–44, 2008. [DOI] [PubMed] [Google Scholar]
  • 36.James TJ, Hughes MA, Cherry GW, Taylor RP. Evidence of oxidative stress in chronic venous ulcers. Wound Repair Regen 11: 172–176, 2003. [DOI] [PubMed] [Google Scholar]
  • 37.Jeffcoate WJ, Game FL. Evidence for the use of biological therapies in ulcers of the foot in diabetes. BioDrugs 28: 1–6, 2014. [DOI] [PubMed] [Google Scholar]
  • 38.Kim JH, Stewart TP, Soltani-Bejnood M, Wang L, Fortuna JM, Mostafa OA, Moustaid-Moussa N, Shoieb AM, McEntee MF, Wang Y, Bechtel L, Naggert JK. Phenotypic characterization of polygenic type 2 diabetes in TALLYHO/JngJ mice. J Endocrinol 191: 437–446, 2006. [DOI] [PubMed] [Google Scholar]
  • 39.Kirker KR, Secor PR, James GA, Fleckman P, Olerud JE, Stewart PS. Loss of viability and induction of apoptosis in human keratinocytes exposed to Staphylococcus aureus biofilms in vitro. Wound Repair Regen 17: 690–699, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kloeters O, Unglaub F, de Laat E, van Abeelen M, Ulrich D. Prospective and randomised evaluation of the protease-modulating effect of oxidised regenerated cellulose/collagen matrix treatment in pressure sore ulcers. Int Wound J 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Kobrin KL, Thompson PJ, van de Scheur M, Kwak TH, Kim S, Falanga V. Evaluation of dermal pericapillary fibrin cuffs in venous ulceration using confocal microscopy. Wound Repair Regen 16: 503–506, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Lauer G, Sollberg S, Cole M, Flamme I, Sturzebecher J, Mann K, Krieg T, Eming SA. Expression and proteolysis of vascular endothelial growth factor is increased in chronic wounds. J Invest Dermatol 115: 12–18, 2000. [DOI] [PubMed] [Google Scholar]
  • 43.Lazarus GS, Cooper DM, Knighton DR, Percoraro RE, Rodeheaver G, Robson MC. Definitions and guidelines for assessment of wounds and evaluation of healing. Wound Repair Regen 2: 165–170, 1994. [DOI] [PubMed] [Google Scholar]
  • 44.Lim CS, Shalhoub J, Gohel MS, Shepherd AC, Davies AH. Matrix metalloproteinases in vascular disease–a potential therapeutic target? Curr Vasc Pharmacol 8: 75–85, 2010. [DOI] [PubMed] [Google Scholar]
  • 45.Loots MA, Lamme EN, Zeegelaar J, Mekkes JR, Bos JD, Middelkoop E. Differences in cellular infiltrate and extracellular matrix of chronic diabetic and venous ulcers versus acute wounds. J Invest Dermatol 111: 850–857, 1998. [DOI] [PubMed] [Google Scholar]
  • 46.MacLeod AS, Mansbridge JN. The innate immune system in acute and chronic wounds. Adv Wound Care 5: 65–78, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Marston WA, Hanft J, Norwood P, Pollak R, Dermagraft Diabetic Foot Ulcer Study Group. The efficacy and safety of Dermagraft in improving the healing of chronic diabetic foot ulcers: results of a prospective randomized trial. Diabetes Care 26: 1701–1705, 2003. [DOI] [PubMed] [Google Scholar]
  • 48.Martin P, Nunan R. Cellular and molecular mechanisms of repair in acute and chronic wound healing. Br J Dermatol 173: 370–378, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Medcalf RL, Hamilton JA. Human synovial fibroblasts produce urokinase-type plasminogen activator. Arthritis Rheum 29: 1397–1401, 1986. [DOI] [PubMed] [Google Scholar]
  • 50.Mendez MV, Stanley A, Park HY, Shon K, Phillips T, Menzoian JO. Fibroblasts cultured from venous ulcers display cellular characteristics of senescence. J Vasc Surg 28: 876–883, 1998. [DOI] [PubMed] [Google Scholar]
  • 51.Mendez MV, Stanley A, Phillips T, Murphy M, Menzoian JO, Park HY. Fibroblasts cultured from distal lower extremities in patients with venous reflux display cellular characteristics of senescence. J Vasc Surg 28: 1040–1050, 1998. [DOI] [PubMed] [Google Scholar]
  • 52.Moore K, Ruge F, Harding KG. T lymphocytes and the lack of activated macrophages in wound margin biopsies from chronic leg ulcers. Br J Dermatol 137: 188–194, 1997. [DOI] [PubMed] [Google Scholar]
  • 53.Mudge BP, Harris C, Gilmont RR, Adamson BS, Rees RS. Role of glutathione redox dysfunction in diabetic wounds. Wound Repair Regen 10: 52–58, 2002. [DOI] [PubMed] [Google Scholar]
  • 54.Mustoe T. Understanding chronic wounds: a unifying hypothesis on their pathogenesis and implications for therapy. Am J Surg 187: 65S–70S, 2004. [DOI] [PubMed] [Google Scholar]
  • 55.Nguyen KT, Seth AK, Hong SJ, Geringer MR, Xie P, Leung KP, Mustoe TA, Galiano RD. Deficient cytokine expression and neutrophil oxidative burst contribute to impaired cutaneous wound healing in diabetic, biofilm-containing chronic wounds. Wound Repair Regen 21: 833–841, 2013. [DOI] [PubMed] [Google Scholar]
  • 56.Nunan R, Harding KG, Martin P. Clinical challenges of chronic wounds: searching for an optimal animal model to recapitulate their complexity. Dis Model Mech 7: 1205–1213, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.O'Toole EA, Goel M, Woodley DT. Hydrogen peroxide inhibits human keratinocyte migration. Dermatol Surg 22: 525–529, 1996. [DOI] [PubMed] [Google Scholar]
  • 58.Oates A, Bowling FL, Boulton AJ, McBain AJ. Molecular and culture-based assessment of the microbial diversity of diabetic chronic foot wounds and contralateral skin sites. J Clin Microbiol 50: 2263–2271, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Palolahti M, Lauharanta J, Stephens RW, Kuusela P, Vaheri A. Proteolytic activity in leg ulcer exudate. Exp Dermatol 2: 29–37, 1993. [DOI] [PubMed] [Google Scholar]
  • 60.Pastar I, Stojadinovic O, Krzyzanowska A, Barrientos S, Stuelten C, Zimmerman K, Blumenberg M, Brem H, Tomic-Canic M. Attenuation of the transforming growth factor beta-signaling pathway in chronic venous ulcers. Mol Med 16: 92–101, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Pecoraro RE, Reiber GE, Burgess EM. Pathways to diabetic limb amputation. Basis for prevention. Diabetes Care 13: 513–521, 1990. [DOI] [PubMed] [Google Scholar]
  • 62.Petreaca ML, Do D, Dhall S, McLelland D, Serafino A, Lyubovitsky J, Schiller N, Martins-Green MM. Deletion of a tumor necrosis superfamily gene in mice leads to impaired healing that mimics chronic wounds in humans. Wound Repair Regen 20: 353–366, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Petreaca ML, Yao M, Ware C, Martins-Green MM. Vascular endothelial growth factor promotes macrophage apoptosis through stimulation of tumor necrosis factor superfamily member 14 (TNFSF14/LIGHT). Wound Repair Regen 16: 602–614, 2008. [DOI] [PubMed] [Google Scholar]
  • 64.Phillips T, Stanton B, Provan A, Lew R. A study of the impact of leg ulcers on quality of life: financial, social, and psychologic implications. J Am Acad Dermatol 31: 49–53, 1994. [DOI] [PubMed] [Google Scholar]
  • 65.Quaresma M, Coleman MP, Rachet B. 40-year trends in an index of survival for all cancers combined and survival adjusted for age and sex for each cancer in England and Wales, 1971–2011: a population-based study. Lancet 385: 1206–1218, 2015. [DOI] [PubMed] [Google Scholar]
  • 66.Rathur HM, Boulton AJ. The diabetic foot. Clin Dermatol 25: 109–120, 2007. [DOI] [PubMed] [Google Scholar]
  • 67.Reiber GE, Boyko EJ, Smith DG. Lower extremity foot ulcers and amputations in diabetes. In: National Diabetes Data Group ( NIH Publication No. 95-1468), edited by Harris MI, Cowie CC, Stern MP. Bethesda, MD: National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, 1995. [Google Scholar]
  • 68.Rennert RC, Rodrigues M, Wong VW, Duscher D, Hu M, Maan Z, Sorkin M, Gurtner GC, Longaker MT. Biological therapies for the treatment of cutaneous wounds: phase III and launched therapies. Expert Opin Biol Ther 13: 1523–1541, 2013. [DOI] [PubMed] [Google Scholar]
  • 69.Rice JB, Desai U, Ristovska L, Cummings AK, Birnbaum HG, Skornicki M, Margolis DJ, Parsons NB. Economic outcomes among Medicare patients receiving bioengineered cellular technologies for treatment of diabetic foot ulcers. J Med Econ 18: 586–595, 2015. [DOI] [PubMed] [Google Scholar]
  • 70.Robbins JM, Dillon J. Evidence-based approach to advanced wound care products. J Am Podiatr Med Assoc 105: 456–467, 2015. [DOI] [PubMed] [Google Scholar]
  • 71.Rojas AI, Phillips TJ. Patients with chronic leg ulcers show diminished levels of vitamins A and E, carotenes, and zinc. Dermatol Surg 25: 601–604, 1999. [DOI] [PubMed] [Google Scholar]
  • 72.Rosner K, Ross C, Karlsmark T, Petersen AA, Gottrup F, Vejlsgaard GL. Immunohistochemical characterization of the cutaneous cellular infiltrate in different areas of chronic leg ulcers. APMIS 103: 293–299, 1995. [DOI] [PubMed] [Google Scholar]
  • 73.Sen CK, Gordillo GM, Roy S, Kirsner R, Lambert L, Hunt TK, Gottrup F, Gurtner GC, Longaker MT. Human skin wounds: a major and snowballing threat to public health and the economy. Wound Repair Regen 17: 763–771, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Singer AJ, Clark RA. Cutaneous wound healing. N Engl J Med 341: 738–746, 1999. [DOI] [PubMed] [Google Scholar]
  • 75.Singh N, Armstrong DG, Lipsky BA. Preventing foot ulcers in patients with diabetes. JAMA 293: 217–228, 2005. [DOI] [PubMed] [Google Scholar]
  • 76.Smeets R, Ulrich D, Unglaub F, Woltje M, Pallua N. Effect of oxidised regenerated cellulose/collagen matrix on proteases in wound exudate of patients with chronic venous ulceration. Int Wound J 5: 195–203, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Sonnemann KJ, Bement WM. Wound repair: toward understanding and integration of single-cell and multicellular wound responses. Annu Rev Cell Dev Biol 27: 237–263, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Spotnitz WD. Commercial fibrin sealants in surgical care. Am J Surg 182: 8S–14S, 2001. [DOI] [PubMed] [Google Scholar]
  • 79.Steed DL. Clinical evaluation of recombinant human platelet-derived growth factor for the treatment of lower extremity diabetic ulcers. Diabetic Ulcer Study Group. J Vasc Surg 21: 71–78, 1995. [DOI] [PubMed] [Google Scholar]
  • 80.Stewart PS, Costerton JW. Antibiotic resistance of bacteria in biofilms. Lancet 358: 135–138, 2001. [DOI] [PubMed] [Google Scholar]
  • 81.Stricklin GP, Hoidal JR. Oxidant-mediated inactivation of TIMP. Matrix Suppl 1: 325, 1992. [PubMed] [Google Scholar]
  • 82.Trengove NJ, Bielefeldt-Ohmann H, Stacey MC. Mitogenic activity and cytokine levels in non-healing and healing chronic leg ulcers. Wound Repair Regen 8: 13–25, 2000. [DOI] [PubMed] [Google Scholar]
  • 83.Tschumperlin DJ. Fibroblasts and the ground they walk on. Physiology 28: 380–390, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Uccioli L, Izzo V, Meloni M, Vainieri E, Ruotolo V, Giurato L. Non-healing foot ulcers in diabetic patients: General and local interfering conditions and management options with advanced wound dressings. Proc Int Surg Wound Forum 24: 36–42, 2015. [DOI] [PubMed] [Google Scholar]
  • 85.Ulrich D, Smeets R, Unglaub F, Woltje M, Pallua N. Effect of oxidized regenerated cellulose/collagen matrix on proteases in wound exudate of patients with diabetic foot ulcers. J Wound Ostomy Continence Nurs 38: 522–528, 2011. [DOI] [PubMed] [Google Scholar]
  • 86.Van de Scheur M, Falanga V. Pericapillary fibrin cuffs in venous disease. A reappraisal. Dermatol Surg 23: 955–959, 1997. [DOI] [PubMed] [Google Scholar]
  • 87.Vranckx JJ, Hoeller D, Velander PE, Theopold CF, Petrie N, Takedo A, Eriksson E, Yao F. Cell suspension cultures of allogenic keratinocytes are efficient carriers for ex vivo gene transfer and accelerate the healing of full-thickness skin wounds by overexpression of human epidermal growth factor. Wound Repair Regen 15: 657–664, 2007. [DOI] [PubMed] [Google Scholar]
  • 88.Wainwright D, Madden M, Luterman A, Hunt J, Monafo W, Heimbach D, Kagan R, Sittig K, Dimick A, Herndon D. Clinical evaluation of an acellular allograft dermal matrix in full-thickness burns. J Burn Care Rehabil 17: 124–136, 1996. [DOI] [PubMed] [Google Scholar]
  • 89.Wallace HJ, Stacey MC. Levels of tumor necrosis factor-alpha (TNF-alpha) and soluble TNF receptors in chronic venous leg ulcers–correlations to healing status. J Invest Dermatol 110: 292–296, 1998. [DOI] [PubMed] [Google Scholar]
  • 90.Weckroth M, Vaheri A, Lauharanta J, Sorsa T, Konttinen YT. Matrix metalloproteinases, gelatinase and collagenase, in chronic leg ulcers. J Invest Dermatol 106: 1119–1124, 1996. [DOI] [PubMed] [Google Scholar]
  • 91.Wenk J, Brenneisen P, Wlaschek M, Poswig A, Briviba K, Oberley TD, Scharffetter-Kochanek K. Stable overexpression of manganese superoxide dismutase in mitochondria identifies hydrogen peroxide as a major oxidant in the AP-1-mediated induction of matrix-degrading metalloprotease-1. J Biol Chem 274: 25869–25876, 1999. [DOI] [PubMed] [Google Scholar]
  • 92.Werner S, Grose R. Regulation of wound healing by growth factors and cytokines. Physiol Rev 83: 835–870, 2003. [DOI] [PubMed] [Google Scholar]
  • 93.Wetzler C, Kampfer H, Stallmeyer B, Pfeilschifter J, Frank S. Large and sustained induction of chemokines during impaired wound healing in the genetically diabetic mouse: prolonged persistence of neutrophils and macrophages during the late phase of repair. J Invest Dermatol 115: 245–253, 2000. [DOI] [PubMed] [Google Scholar]
  • 94.Wieman TJ, Smiell JM, Su Y. Efficacy and safety of a topical gel formulation of recombinant human platelet-derived growth factor-BB (becaplermin) in patients with chronic neuropathic diabetic ulcers. A phase III randomized placebo-controlled double-blind study. Diabetes Care 21: 822–827, 1998. [DOI] [PubMed] [Google Scholar]
  • 95.Williams DT, Harding KG. New treatments for diabetic neuropathic foot ulceration: views from a wound healing unit. Curr Diab Rep 3: 468–474, 2003. [DOI] [PubMed] [Google Scholar]
  • 96.Wilson EL, Becker ML, Hoal EG, Dowdle EB. Molecular species of plasminogen activators secreted by normal and neoplastic human cells. Cancer Res 40: 933–938, 1980. [PubMed] [Google Scholar]
  • 97.Wlaschek M, Peus D, Achterberg V, Meyer-Ingold W, Scharffetter-Kochanek K. Protease inhibitors protect growth factor activity in chronic wounds. Br J Dermatol 137: 646, 1997. [DOI] [PubMed] [Google Scholar]
  • 98.Wlaschek M, Scharffetter-Kochanek K. Oxidative stress in chronic venous leg ulcers. Wound Repair Regen 13: 452–461, 2005. [DOI] [PubMed] [Google Scholar]
  • 99.Wlaschek M, Wenk J, Brenneisen P, Briviba K, Schwarz A, Sies H, Scharffetter-Kochanek K. Singlet oxygen is an early intermediate in cytokine-dependent ultraviolet-A induction of interstitial collagenase in human dermal fibroblasts in vitro. FEBS Lett 413: 239–242, 1997. [DOI] [PubMed] [Google Scholar]
  • 100.Wong VW, Sorkin M, Glotzbach JP, Longaker MT, Gurtner GC. Surgical approaches to create murine models of human wound healing. J Biomed Biotechnol 2011: 969618, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Wysocki AB, Staiano-Coico L, Grinnell F. Wound fluid from chronic leg ulcers contains elevated levels of metalloproteinases MMP-2 and MMP-9. J Invest Dermatol 101: 64–68, 1993. [DOI] [PubMed] [Google Scholar]
  • 102.Yeoh-Ellerton S, Stacey MC. Iron and 8-isoprostane levels in acute and chronic wounds. J Invest Dermatol 121: 918–925, 2003. [DOI] [PubMed] [Google Scholar]
  • 103.Zelen CM, Serena TE, Denoziere G, Fetterolf DE. A prospective randomised comparative parallel study of amniotic membrane wound graft in the management of diabetic foot ulcers. Int Wound J 10: 502–507, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Zhao G, Usui ML, Lippman SI, James GA, Stewart PS, Fleckman P, Olerud JE. Biofilms and inflammation in chronic wounds. Adv Wound Care 2: 389–399, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Physiological Genomics are provided here courtesy of American Physiological Society

RESOURCES