Skip to main content
International Wound Journal logoLink to International Wound Journal
. 2022 Dec 23;20(6):2346–2359. doi: 10.1111/iwj.14048

The importance of inflammation control for the treatment of chronic diabetic wounds

Anna L Worsley 1,2, Dennis H Lui 2, Winnie Ntow‐Boahene 1,2, Wenhui Song 2, Liam Good 1, Janice Tsui 2,
PMCID: PMC10333011  PMID: 36564054

Abstract

Diabetic chronic wounds cause massive levels of patient suffering and economic problems worldwide. The state of chronic inflammation arises in response to a complex combination of diabetes mellitus‐related pathophysiologies. Advanced treatment options are available; however, many wounds still fail to heal, exacerbating morbidity and mortality. This review describes the chronic inflammation pathophysiologies in diabetic ulcers and treatment options that may help address this dysfunction either directly or indirectly. We suggest that treatments to reduce inflammation within these complex wounds may help trigger healing.

Keywords: chronic wounds, diabetes, diabetic ulcers, inflammation, wound healing

1. INTRODUCTION

Diabetes mellitus is a chronic metabolic condition of insulin resistance, reduced insulin production, and chronically elevated blood glucose levels. The global prevalence of diabetes has trebled in the last two decades, particularly in countries with developing economies, and is predicted to affect 1 in 10 people worldwide by 2045. 1 Diabetic foot ulceration is one of the major complications of diabetes. It is a serious, highly morbid condition, which has been shown to be independently associated with increased mortality. 2 Patients with diabetes mellitus are especially prone to developing foot ulceration due to peripheral neuropathy, which leads to biomechanical changes to the foot and loss of protective reflexes and sensation to injury. 3 , 4 , 5 The presence of peripheral vascular disease and a predilection to polymicrobial infection are also contributing factors. As many as one in three patients with diabetes mellitus will develop a diabetic foot ulcer during their lifetime. 3 , 4 , 6 , 7

The prognosis for patients suffering from diabetic foot ulceration is bleak. Thirty three percent of diabetic ulcers do not heal and remain as chronic wounds. 8 Of those ulcers that do achieve ‘healing’, 65% will experience re‐ulceration at 3 years, exemplifying the chronic, relapsing, and remitting nature of this condition. 3 Approximately 20% of moderate to severe diabetic foot ulcers lead to some form of amputation, and patients with diabetes are up to 25 times more likely to lose their leg than those not suffering from the condition. 3 In 2005, the International Diabetes Federation estimated that one lower limb was lost every 30 seconds due to diabetes worldwide. 9 Five‐year survival after diabetes‐related major amputation was estimated to be 47%, dropping to 17% for patients on dialysis. 10 These survival rates are comparable to, or worse than, other severe diseases, including heart failure, myocardial infarction, stroke, and some cancers. 11 , 12 A large, population‐based study found that over a follow‐up period of 10 years, patients with a history of diabetic foot ulceration were twice as likely to die than those without diabetes, consistent with results found by other studies. 12 , 13 , 14 The presence of foot ulceration in diabetic patients is associated with poor quality of life, low physical functioning, increased risk of depression, and anxiety, and is an independent risk factor for mortality. 2 , 15 The multitude of knock‐on effects an ulcer may have on a patient's health must be considered by health care professionals and carers. (Figure 1).

FIGURE 1.

FIGURE 1

Progression, associated risk factors, and subsequent health risks of diabetic foot ulceration. Created with BioRender.com

The burden of foot ulceration on health care systems, carers, as well as to society in terms of health economics cannot be overstated. Where diabetes‐related care was estimated to cost 176 billion USD annually, up to one third of this was attributed to lower extremity care. 3 , 15 The presence of diabetic foot ulceration was associated with a seven‐fold increase in requiring hospital admission, as infection and gangrene are common complications once an ulcer has formed. 16 The costs of hospital care, outpatient clinic care, informal care, sickness absence, and care after amputation amount to tens of billions worldwide. 11 , 17 , 18 , 19 , 20 , 21 Worryingly, these costs are set to continue to increase as both the prevalence of diabetes and life expectancy with diabetes are rising.

The mammalian body has evolved a carefully orchestrated series of defences to skin wounding, which include innate defences and acquired responses. These defences always include initial inflammation, cell proliferation, differentiation and migration, angiogenesis, extracellular matrix proliferation, and remodelling. Chronic inflammation is a key feature of both diabetes mellitus and wound chronicity. Though other inflammatory conditions can lead to wound chronicity (i.e., scleroderma), diabetic patients are at particular risk due to repeated injury due to peripheral neuropathy and the loss of protective reflexes and the polymicrobial bioburden in their wounds, particularly in foot ulcers. This chronic inflammatory state in diabetes mellitus disrupts the normal responses to wounding at a systemic and local level, leading to wounds that are unable to progress through the normal wound healing stages (Figure 2). 8 Moreover, patients with a chronic inflammatory state are less able to mount an appropriate systemic and local response to the microbial bioburden often present in wounds. This ‘burnt‐out’ innate immunity of diabetic patients, combined with the propensity for poly‐microbial wound infection in diabetic foot ulceration, are the factors, which drive the manifestation of overwhelming bacterial sepsis from diabetic foot infections, which can be life‐threatening. It is important to understand this inflammatory dysfunction in order to develop strategies to target chronic inflammation and improve wound healing without further jeopardising innate immunity to invading pathogens. This review summarises current knowledge concerning the role of chronic inflammation in chronic diabetic wounds, including diabetic foot ulcers, and the treatment options that may help address this dysfunction either directly or indirectly.

FIGURE 2.

FIGURE 2

An overview of the principal stages of normal wound healing 127 , 128 , 129 , 130 , 131 , 132

2. PATHOPHYSIOLOGY

2.1. Reduced initial inflammatory response

Relative to acute wounds, diabetic chronic wounds display weak early‐stage inflammatory responses (Figure 3), which may underlie the development of the chronic inflammatory phenotype of diabetic ulcers. At wounding, IL‐6, IL‐8, their receptors, the C‐C chemokine receptor type 2 (CCR2) for macrophage chemoattractant protein‐1 and prepro‐NPY, the precursor of neuropeptide Y (NPY), are present at much lower levels compared with non‐diabetic environments . 22 , 23 , 24 This is also mirrored in single‐cell transcriptomic and pathways analysis showing that systemic NK and T cells in diabetic patients exhibit inhibition of IL‐6, IL‐8, and CD28 signalling pathways. 25 There is also an increase of neutral endopeptidase in the skin around patients with diabetic chronic wounds. Neutral endopeptidase degrades substance P as part of its regulation, suggesting a reduction in substance P. 26 The neuropeptide substance P stimulates keratinocyte, fibroblast, and endothelial cell pro‐inflammatory responses. 26 These differences in the diabetic wound environment suggest a reduced initial inflammatory response compared with non‐diabetic acute wounds. 22 , 23 , 24 , 26

FIGURE 3.

FIGURE 3

Pathophysiology of diabetic chronic wounds. The condition illustrated arises through two stages. In the first stage there is a weak initial inflammatory response which is then followed by the chronic inflammatory response. During the weak initial inflammatory response, low levels of cytokines (IL‐6, IL‐8) CCR2 and prepro‐NPY are released within the wound environment. The levels of neutral endopeptidase, an enzyme which degrades substance P, also increases in the skin. In the second stage, there is reduced keratinocyte, fibroblast and endothelial cell stimulation. In contrast, the chronic inflammatory response is characterised by an accumulation of leukocytes (macrophages, neutrophils, basophils and T cells) due to an overexpression of the complement system, STAT4, OSM and chemoattractants MCP‐2 and MCP‐1. In addition, the dysregulation of SELPLG also reduces the clearance of these leukocytes from the wound site

2.2. The chronic inflammatory phenotype

Immune cell infiltration, accumulation, and pro‐inflammatory cell polarisations are promoted in the diabetic chronic wound environment, contributing to the chronic inflammatory phenotype (Figure 3). 27 The expression levels of the complement system, signal transducer and activator of transcription factor (STAT) 4, oncostatin M (OSM), OSM receptor subunit β (OSMRβ), macrophage inflammatory protein‐2, and macrophage chemoattractant protein‐1 expression have all been found to be increased in the diabetic ulcer environment. 24 , 28 This contributes to the accumulation of polymorphonuclear neutrophils and macrophages. Macrophages become hyperpolarised and are predominantly granulocytic (Gr) ‐1+, CD11b+, and CD14+ macrophages. 29 Their clearance is reduced in association with the dysregulation of the cell membrane protein selectin P ligand (SELPLG), causing a build‐up of their population and associated secreted pro‐inflammatory molecules. 30 α‐defensins are also upregulated in hyperglycaemic conditions, promoting IL‐8 expression and enhancing the recruitment of neutrophils, basophils, and T cells. 31 , 32 This influx and accumulation of immune system cells is followed by the release of associated pro‐inflammatory cytokines and chemokines, worsening the chronic inflammatory phenotype. Infection worsens the situation by promoting immune responses as well as complicating ischaemia and neuropathy. 33 Excretions from S. aureus biofilms, the dominating bacteria species in diabetic chronic wounds, were also found to directly contribute to the chronic inflammatory phenotype by causing pro‐inflammatory gene expression in epithelial keratinocytes. 34

2.2.1. Senescence‐associated secretory phenotype

Cellular senescence describes the process in which cells cease dividing and undergo phenotypic changes. As with many tissues in patients with diabetes, diabetic ulcers have an increased number of senescent cells displaying a pro‐inflammatory senescence‐associated secretory phenotype (SASP). This phenotype develops as a consequence of increased oxidative stress‐related RNA damage, DNA damage, and telomere shortening. 35 , 36 Experimental models of diabetic mice revealed a rapid accumulation of senescence cells of various cell types in wounded skin. 37 The majority of SASP cells are macrophages, which are hypothesised to be stalled between the transition from pro‐inflammatory M1 and pro‐resolution M2 phenotypes. Fibroblasts also show a strong senescent phenotype, adding to poor healing outcomes. 38 This is potentially in response to hyperglycaemia and increased local inflammation. 35 , 39 A SASP increases the release of pro‐inflammatory cytokines and chemokines, promoting a chronic inflammatory environment. CXC chemokine receptor 2 (CXCR2), the IL‐8 receptor, as well as chemokines chemokine (C‐X‐C motif) ligand 1 (CXCL) 1 and CXCL2 are highly expressed in senescent cells, potentially playing a role in the chronic inflammatory pathophysiology through immune cell recruitment. 31 , 32 , 38 Inhibition of CXCR2 has been shown to protect CXCL2 activity, dampen neutrophil infiltration, and reduce cellular senescence, promoting wound closure in diabetic ulcers. This suggests CXCR2 and IL‐8 play important roles in diabetic chronic wound pathophysiology. 38 SASPs may also contribute to diabetic ulcer formation as part of metaflammation. Monocyte chemoattractant protein‐1 (MCP‐1) is a pro‐inflammatory cytokine expressed in SASP cells. MCP‐1 is upregulated in diabetic rat models and has been found to be associated with patient susceptibility to diabetic ulcer formation. 40

2.2.2. Neutrophils

In the diabetic environment, neutrophils display increased superoxide production and protein kinase C activity. 41 They also have elevated basal calcium levels, overproduce peptidyl arginine deiminase type IV (PAD4), a calcium‐dependent enzyme, and overproduce neutrophil extracellular traps (NETs), impeding wound healing via increased NETosis. 42 Despite this increased NETosis, it was found that in diabetic mouse wounds infected with S. aureus neutrophil apoptosis was reduced, neutrophil clearance was reduced and neutrophil TNF‐α production increased. 43 Elevated saturated free fatty acids also promote neutrophil survival and reduce macrophage phagocytosis in association with prostaglandin production. 44 These processes cause the accumulation of neutrophils in diabetic ulcers, adding to the increased inflammation and reduced wound healing progression.

2.2.3. Macrophages

The persistence of pro‐inflammatory M1 macrophages within wounds has been hypothesised to be a key contributor to diabetic chronic wound pathology. However, newer transcriptomic data has identified that, at least in the wound, the M1 phenotype may also be associated with improved diabetic foot ulcer healing. 25 Driven by hyperglycaemia and hypoxia, the pro‐inflammatory phenotype is characterised by an increased release of inflammatory cytokines such as TNF‐α and IL‐1. 45 TNF‐α stimulates the histone acetyltransferase Males absent on the first (MOF) in macrophages and that this is increased in the diabetic environment. MOF adds to the activation of TNF‐α signalling and promotes NFκB–mediated gene transcription via H4K16 acetylation in wound macrophages, impeding wound healing processes. 45 In an obese mouse model, inhibiting TNF‐α signalling using neutralising monoclonal antibodies inactivated macrophages, reduced circulating monocyte populations and reduced inflammatory cytokine levels. This induced wound closure, suggesting TNF‐α signalling is another major contributing factor to diabetic ulcer development. 46

Sustained elevated expression of IL‐1β by the nodulation (NOD)‐, Leucine‐rich repeat (LRR)‐, and pyrin domain‐containing protein 3 (NLRP3) inflammasome further impedes macrophage transition to the pro‐healing M2 phenotype by downregulating peroxisome proliferator‐activated receptor (PPAR) ‐γ, a regulator of glucose and lipid metabolism. 47 Insulin treatment may reduce these effects. In vivo studies indicated that insulin promotes anti‐inflammatory phenotype transitions by upregulating PPAR‐γ. Furthermore, insulin activated Akt‐Rac‐1 (activated cell division control protein 42 kinase‐ras‐related C3 botulinum toxin substrate‐1) signalling, a regulatory pathway for glucose uptake that is downregulated in diabetic ulcers, inhibiting hyperglycaemia‐induced p38, NF‐κB, and STAT1 transcriptional activity activation. 48 The neuropeptide neurotensin has also been found to be involved in stimulating the migratory and inflammatory response of macrophages in hyperglycaemic environments, indicating the role of the neuroendocrine system on diabetic wound healing. 49

As mentioned above, the phagocytic and efferocytotic activity of macrophages is reduced in diabetes, increasing the population of dysfunctional cells in the diabetic chronic wound environment. The phagocytic activity of macrophages for apoptotic neutrophils has been found to be significantly reduced in diabetic ulcers. 50 Phagocytosis‐related genes for CD36 and Class B scavenger type 1 receptors are downregulated, pro‐apoptotic factors are downregulated, and pro‐apoptotic factors are upregulated in the pro‐inflammatory phenotype. 50 Macrophage efferocytosis impairment increases the burden of apoptotic cells at the wound site, causing the stimulation of pro‐inflammatory and the attenuation of anti‐inflammatory cytokine responses. 51

2.2.4. γδ T lymphocytes

Diabetic chronic wounds have lower numbers of skin‐resident γδ T lymphocytes, of which have reduced expression levels of FGF‐7, FGF‐10, and insulin‐like growth factor (IGF) ‐1 compared with acute healing models. 52 These growth factors are more pro‐healing than pro‐inflammatory, and in general, T lymphocytes have a more anti‐inflammatory phenotype. 53 , 54 , 55 Reduced population and activity of skin‐resident γδ T lymphocytes further promotes inflammation.

2.2.5. Other dysfunctional signalling molecules and receptors

Multiple areas of dysfunction cause the pro‐inflammatory phenotype found in diabetic ulcers. The dysregulation of upstream receptors that initiate inflammatory cascades, such as the toll‐like receptor (TLR) family, plays an important role. TLRs 2, 4, 7, and 9 are significantly upregulated in diabetic chronic wounds. 6 , 8 , 56 , 57 , 58 , 59 Antagonising TLR‐4 systemically in vivo or using knockout TLR‐4 in vivo models has been found to improve diabetic wound healing, and reduce pro‐inflammatory phenotypes. 8 , 59 In contrast, clinical studies of diabetic foot ulcers in patients with Indian heritage found reduced levels of TLR‐4 signalling in diabetic foot ulcers with specific TLR‐4 SNP genotypes, displaying potential variation in how TLR‐4 is dysfunctional. 60 The increase in TLR expression in diabetic ulcers reduces healing, increases MyD88 signalling and increases the expression and activation of NF‐κB, interferon and inflammatory cytokines and chemokines, such as IL‐6, TNF‐α, S100A8, Il‐8, and Il‐1β. 6 , 8 , 56 , 57 , 58 Many of these signalling molecules go on to promote further pro‐inflammatory cytokine release as part of positive feedback loops, worsening the inflammatory phenotype.

Many other mechanisms further downstream contribute to the dysfunctional healing seen in diabetic ulcers. For example, unregulated iron levels lead to reactive oxygen species (ROS) production, increased oxidative stress, and macrophage polarisation. 38 , 61 , 62 Granzyme B is a serine protease, which positively regulates apoptosis in normal wound healing. Expressed on immune system cells, it accumulates in the extracellular matrix (ECM) in the diabetic ulcer environment. It cleaves essential wound healing proteins such as fibronectin, preventing wound healing. 63 Sirtuin 6 is a sirtuin family protein involved in regulating many pathophysiological processes, including inflammation, glycolysis and DNA repair. 64 Sirtuin 6 deficiency in diabetic chronic wounds further exacerbates the pro‐inflammatory phenotype of diabetic chronic wounds by increasing NF‐κB activation, oxidative stress, and decreasing angiogenesis. 64

Chronic hyperglycaemia has been shown to upregulate the signalling molecule suppressor of cytokine signalling 3 (SOCS3), a protein usually involved in the suppression of inflammation. 65 Surprisingly, in the diabetic chronic wound environment, this exacerbates wound inflammation. This is in association with increased expression of chemokine macrophage inflammatory protein 2 (MIP‐2), increased expression of inflammatory enzymes cyclooxygenase (COX)‐2, inducible nitric oxide synthase (iNOS), and increased levels of TGF‐ β in epithelial cells. 66 COX‐1 and ‐2 expression and activity have also been found to be dysregulated using diabetic obese mouse wound models, with COX‐1 coupled prostaglandin directly contributing to impaired diabetic wound healing. 67

Endothelial overexpressed lipopolysaccharide‐associated factor 1 (EOLA1) is a recently discovered regulator of inflammation, which is downregulated in diabetic chronic wounds. 68 EOLA1 is expressed in leukocytes and endothelial cells. It has been found to be involved in cell growth promotion, apoptosis inhibition, and the downregulation of inflammatory cytokine secretion, including IL‐6 and intercellular adhesion molecule‐1 (ICAM‐1). 68 Therefore, its downregulation may play an important role in the chronic inflammatory phenotype of diabetic ulcers. In contrast, it is upregulated after LPS stimulation, suggesting a potential mechanism involved in reducing the diabetic ulcer's ability to fight infection.

3. CURRENT TREATMENTS THAT REDUCE INFLAMMATION

3.1. Commonly used treatments and management techniques

Many of the management principles and treatment options used to treat diabetic foot ulceration have indirect anti‐inflammatory effects. Diabetic chronic wounds remain open for extended periods of time, often months, due to dysfunctional immune responses. When considering that open diabetic wounds also encourage polymicrobial colonisation, unsurprisingly the rate of wound infection and deeper infection (including osteomyelitis) is high, with approximately 58% of cases being infected. 69 , 70 , 71 , 72 Infection, where present, must be controlled as a first priority. Localised infection increases inflammation, exacerbates the dysfunctional immune response, increases pain, and promotes morbidity. 3 , 51 , 73 Treatment for infection in these wounds is achieved with (1) prompt surgical debridement and drainage of any collections, if necessary, and (2) empirical antibiotics followed by sensitivity‐guided antibiotics after identification of bacterial strains is complete from tissue sampling. 74 Surgical debridement is performed to clean the wound, removing necrotic, non‐viable, or infected tissue as well as building up immune cells and cytokines in the exudate and slough, thereby reducing their pro‐inflammatory output. 75 Dressings can also contain antiseptic agents, such as silver, iodine, and polyhexamethylene biguanide (PHMB) to help stabilise the current infection and prevent further infection. 76 , 77 As well as benefiting wound healing, incorporating antimicrobials into dressings can improve pain, and reduce the risk of further complications, such as sepsis. 70 Routine wound care, including regular irrigation and prompt removal of excessively soiled dressings, may also reduce excessive inflammation.

Negative pressure wound therapy (NPWT) is a treatment, which may be applied after debridement and infection control to help promote wound approximation and healing. 74 It achieves this effect by removing excess wound exudate, inducing wound contraction, and promoting angiogenesis as well as wound granulation through mechanical stimulation. 78 NPWT may also reduce inflammation, however, reports are mixed. Wang et al reported that negative pressure therapy suppresses inflammation via down‐regulating the MAPK‐JNK signalling pathway in diabetic ulcers. 79 Ludwig‐Slomczynska et al found that NPWT for diabetic ulcers caused epigenetic changes that lead to the inhibition of complement system activation. 80 In contrast, Pawar et al recorded increased CD68 cell densities in response to NPWT for periprosthetic tissue treatment, indicating increased inflammation. 81 Furthermore, Norbury et al found that NPWT helped overcome immunoparalysis in the swine model of ischemia/reperfusion injury coupled with sepsis, specifically by increasing lymphocyte populations and increasing macrophage reactive oxygen species production. 82 The International Working Group on the Diabetic Foot (IWGDF) performed a systematic review of literature on the use of interventions to enhance healing in chronic diabetic foot ulcers, including the use of NPWT. 83 The working group recommended that NPWT should be considered to reduce wound size in post‐surgical wounds, and that it should be avoided in non‐surgical wounds. Their published recommendations take into account the variability of studies, the lack of blinding and the controls in studies, and potential for bias in the current literature.

3.2. Less common and future treatments

3.2.1. Hyperbaric oxygen therapy

Hyperbaric oxygen (HBO2) as a therapy has been studied in animal models and as a treatment for human injury and wound healing for more than four decades. 84 , 85 The treatment involves controlled exposure of the patient to high atmospheric pressure (between 2 and 3 atm), with 100% oxygen content in a compression chamber for a set amount of time (1–2 hours). The net effect of this therapy is to temporarily increase oxygen tension in the wound, followed by a decrease back to hypoxic conditions, and cycling between these two states appears to improve wound healing. Hyperbaric oxygen therapy reduces expression of inflammatory cytokines (including Interleukin‐1 and Interleukin‐2), increases angiogenesis, improves collagen formation, promotes fibroblast migration, reduces the metalloproteinase expression, and promotes both antibiotic and leucocyte function against microbes. 85 , 86 , 87 , 88 Studies in various animal models have demonstrated that wounds exposed to hyperbaric oxygen show increased granulation and accelerated wound contraction, as well as ameliorating inflammatory processes in other conditions. 86 , 89 However, the treatment does have risks, including barotrauma to the ears, seizures from acute central nervous system oxygen toxicity, and reversible myopia. A Cochrane review and meta‐analysis of HBO2 therapy for chronic wounds found moderate evidence that the treatment improved the chance of healing at 1 year in diabetic foot ulcers, but did not reduce the risk of amputation. 84 The provision of hyperbaric oxygen therapy requires specialist facilities, which are relatively more available in North America, but less common elsewhere in the world, limiting widespread adoption. Current guidance from the National Institute of Health and Care Excellence (NICE) does not recommend the use of hyperbaric oxygen in chronic diabetic foot wounds except when involved in a clinical trial. 74

3.2.2. Skin replacement and grafting, and amniotic membrane treatment

Replacement of epithelial tissue with autograft, allograft, xenograft, or bioengineered tissues has been studied as a treatment for chronic diabetic foot wounds. These skin substitutes are categorised by whether they are cellular or acellular scaffolds made from skin tissues or biomaterials and are further categorised as dermal or epidermal substitutes (or both). When applied to infection‐free and vascularised wounds, these tissues may integrate with the wound and act to improve extracellular matrix deposition and composition, and increase secretion of healing cytokines and growth factors, and ultimately increase re‐epithelialisation. 90 However, the exact effect of these treatments on chronic wound inflammation is not yet fully characterised. Numerous bioengineered skin substitutes containing fresh or cryopreserved human keratinocytes or fibroblasts have been applied to patients with diabetic foot ulceration, with positive preliminary results. 91 , 92 , 93 , 94 Unfortunately, the significant heterogeneity of skin substitute products available, heterogenous study design without blinding investigators, low study numbers, industry involvement in the majority of studies, and potential for bias all mean that at present, it is difficult to draw any firm conclusions about this potentially promising treatment. 90 , 91

Application of human amniotic membrane is another therapy, which may reduce inflammation in diabetic ulcers. Dehydrated or cryopreserved allogeneic human amniotic membrane is now commercially available, and its application in diabetic foot ulceration has been studied. Amniotic membrane tissue is rich in cytokines (IL‐4, IL‐6, IL‐8, and IL‐10), growth factors (including platelet derived growth factors, fibroblast growth factors, transforming growth factor α, epidermal growth factor, and granulocyte colony‐stimulating factor), native stem cells, and tissue inhibitors of metalloproteinases (TIMPs). 95 , 96 , 97 , 98 Amniotic membrane has been shown to reduce Th1 and Th2 cell cytokine synthesis and also lacks human leukocyte antigen (HLA) antigens, which make it less immunogenic compared with other skin substitutes. 97 , 99 Clinical studies examining human amniotic membrane allograft in chronic diabetic foot ulcers have shown improved wound healing in diabetic ulcers. 90 , 97 , 98 , 100 However, these trials were small, non‐blinded, and had industry involvement. Use of these products has been limited partly due to the prohibitive cost of each treatment cycle and the lack of clear long‐term evidence. At present, NICE guidelines suggest skin replacements can be considered only in diabetic foot ulcers, which have failed to progress, at the recommendation of a multidisciplinary foot care service. 74 Further well‐designed studies are required to examine the physiological effects and clinical effects, including long‐term effects, of skin substitutes and grafting in chronic diabetic wounds.

3.2.3. Growth factors

Delivering growth factors to the wound bed may help reduce inflammation as well as general wound healing promotion. Growth factors can be delivered directly via injection or by incorporation into biomaterials, or indirectly via stem cells, exosomes, platelet‐rich‐plasma, or allographs. 75 , 101 Growth factors used directly for diabetic foot ulcer treatments described in the literature include TGF‐β, FGF, VEGF, PDGF, and EGF, the latter two being the only ones to reach clinical evaluation. 102 PDGF and EGF have anti‐inflammatory properties when used in skin disorders. 103 , 104 In pre‐clinical and clinical studies, EGF has been shown to improve healing with the promotion of granulation and fibroblast proliferation. 101 , 105 , 106 , 107 Regranex is a gel containing PDGF; it is the only growth factor treatment approved by the United States food and drug administration, however, it is not recommended by The National Institute for Health and Care Excellence (NICE). 74 This is mainly in response to the formulation, dosing, safety, and efficacy issues. 53 One example raising safety concerns includes a retrospective observational study connecting Regranex to an increased risk of malignancies. Despite this study being unpublished and requiring further evidence, its use in high risk cancer patients is restricted in the United Kingdom as a consequence. 108 The clinical evidence of efficacy for Regranex has been seen as weak, and the product is not widely used. 109

3.2.4. Stem cell therapy

Stem cell therapy can be used to deliver active molecules and complexes to the wound bed. Its recommendation is not directly mentioned in NICE guidelines, however, it is sometimes used as a last attempt to avoid amputation in patients with no other revascularisation options. 110 Depending on the stem cells used, they can differentiate into various cell types that help promote healing and reduce inflammation. Stem cells secrete pro‐healing cytokines, chemokines, and growth factors, including VEGFs and IGF‐1. This aids immunomodulation, cell recruitment, ECM remodeling, angiogenesis, and neuroregeneration. 110 , 111 , 112 The most common stem cells used in the literature are bone marrow‐derived mesenchymal stem cells, consisting of 50% and 53% of pre‐clinical and clinical studies, respectively. Others include human umbilical cord mesenchymal stem cells and adipose tissue‐derived mesenchymal stem cells. Induced pluripotent stem cells (iPS) have not yet entered the literature for this use. 110 Though, recently, Gorecka et al described the use of smooth muscle cells derived from iPS cells, showing the promotion of angiogenesis and accelerated healing in vivo when incorporated into collagen scaffolds. 113 Preclinical and clinical evidence suggests that stem cells offer an effective treatment route for diabetic ulcers. 110 Stem cell administration techniques are varied; they include local injection, the most common method, and topically as part of a free liquid, hydrogel, or solid scaffold. More research is needed to distinguish the optimal type and delivery technique, as well as the eligibility of patients and their wounds. 110 , 112

The use of stem cell‐derived exosomes entered the literature more recently, offering a potential route for supplying the benefits of stem cells with reduced risks and costs. In preclinical studies, stem cell‐derived exosomes promoted anti‐inflammatory cell phenotypes, cell migration, and angiogenesis. 114 , 115 , 116 , 117 This was improved when the exosomes were from LPS‐preconditioned mesenchymal stem cells compared with non‐preconditioned cells. 118 The associated ablation of inflammation and induced wound healing was found to be through lethal‐7b miRNA shuttling associated with reduced TLR‐4 signalling, promoting M2 macrophage phenotype transitions. This adds to the evidence that TLR‐4 signalling dysfunction might play a major role in upstream diabetic wound healing prevention.

3.2.5. Receptor targets

Targeting cell signalling pathways via receptor targets offers a relatively new approach in the attempt to induce healing in diabetic ulcers by reducing inflammation directly. There are many receptors identified in the literature, which have dysfunctional inflammatory signalling that could act as potential therapeutic targets for diabetic ulcers in future treatments. For example, the antagonism of members of the TLR family, such as TLR‐4, TLR‐2, TLR‐7, and TLR‐9, as well as CXCR2 and OSMRβ, some of which have been tested in vivo already. 8 , 56 , 57 , 58 , 59 , 119 Naltrexone is one such drug, repurposed from use as an opiate antagonist for alcohol and opioid dependence. Recently, naltrexone has been shown to promote diabetic ulcer healing in vivo by 13% to 30% via upstream receptor targeting, offering novel future treatment pathways. 119 This may work through the inhibition of pro‐inflammatory signalling induced by TLR‐4, TLR‐7, and TLR‐9. 120 , 121

3.2.6. Non‐specific anti‐inflammatory compounds

Numerous substances display non‐specific anti‐inflammatory activity with varying potencies. However, most are still in the preclinical stages of evaluation. Insulin, applied topically, has been found to accelerate diabetic wound healing by promoting anti‐inflammatory macrophage phenotypes. 76 , 122 Hyaluronic acid provides anti‐inflammatory activity in a molecular weight dependent manner with promising wound healing ability in vivo. 123 , 124 More naturally sourced substances, for example, honey, curcumin, oregano extract, and hydroxytyrosol, can also provide anti‐inflammatory activity. 125 , 126 Many are non‐specific and are still in research for their effectiveness in vivo. They are sometimes incorporated into more complex biomaterials with promising preclinical results or into simple wound dressings for a more natural approach.

4. CONCLUDING REMARKS

This review highlights the pivotal role of disordered inflammation in diabetic chronic wounds. Future treatments that target and break the cycle of disordered inflammation may improve treatments. Current clinical guidelines do not include specific anti‐inflammatory therapies. However, there is growing evidence that inflammation control may help overcome healing deficiencies associated with current approaches. Many potential anti‐inflammatory therapeutic options and novel research pathways are available; for example, targeting upstream receptors and the use of less‐specific anti‐inflammatory peptides. These approaches could be used alongside current treatments, providing a multi‐faceted control, which seems appropriate for a multi‐faceted condition, and a combined approach may improve patient outcomes. Further investigation of pharmacological interventions on the TLR family, systemic anti‐inflammatories including blockade of the IL‐1 axis (ie, monoclonal antibodies to IL‐1 receptor or IL‐1β), application of autologous PBMC‐derived products to wounds, and improved dressing technologies which allow optimal wound healing conditions whilst delivering topical agents, are all practical areas worthy of further investigation as future therapies.

AUTHOR CONTRIBUTIONS

Anna Worsley, Liam Good, Wenhui Song and Janice Tsui conceptualised the manuscript. Anna Worsley and Dennis Lui wrote the manuscript. All authors read, discussed, commented and revised the manuscript.

CONFLICT OF INTEREST

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the content reported in this review article.

ACKNOWLEDGEMENTS

The authors acknowledge financial support by the Biotechnology and Biological Sciences Research Council (BBSRC LIDo DTP studentship, 1764829). W.S. also thank financial support by the Engineering and Physical Sciences Research Council in the United Kingdom [EP/L020904/1 and EP/R02961X/1]. Figures were created with BioRender.com.

Worsley AL, Lui DH, Ntow‐Boahene W, Song W, Good L, Tsui J. The importance of inflammation control for the treatment of chronic diabetic wounds. Int Wound J. 2023;20(6):2346‐2359. doi: 10.1111/iwj.14048

Work carried out in the above institutions (1, 2).

DATA AVAILABILITY STATEMENT

Data sharing not applicable to this article as no datasets were generated or analysed during the current study.

REFERENCES

  • 1. Saeedi P, Petersohn I, Salpea P, et al. Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: results from the international diabetes federation diabetes atlas, 9th edition. Diabetes Res Clin Pract. 2019. Nov;157:107843. [DOI] [PubMed] [Google Scholar]
  • 2. Martins‐Mendes D, Monteiro‐Soares M, Boyko EJ, et al. The independent contribution of diabetic foot ulcer on lower extremity amputation and mortality risk. J Diabetes Complications. 2014. Sep;28(5):632‐638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Armstrong DG, Boulton AJM, Bus SA. Diabetic foot ulcers and their recurrence. Ingelfinger JR, editor. N Engl J Med. 2017. Jun 15;376(24):2367‐2375. [DOI] [PubMed] [Google Scholar]
  • 4. Boulton AJM, Armstrong DG, Albert SF, et al. Comprehensive foot examination and risk assessment: a report of the task force of the foot care interest Group of the American Diabetes Association, with endorsement by the American Association of Clinical Endocrinologists. Diabetes Care. 2008. Aug 1;31(8):1679‐1685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Boulton A, Armstrong D, Hardman M, et al. Diagnosis and Management of Diabetic Foot Infections [internet]. Am Diabetes Assoc. 2020;2020(1). [cited 2021 Nov 24]. https://professional.diabetes.org/monographs [PubMed] [Google Scholar]
  • 6. Davis FM, Kimball A, Boniakowski A, Gallagher K. Dysfunctional wound healing in diabetic foot ulcers: new crossroads. Curr Diab Rep. 2018. Jan 23;18(1):2. [DOI] [PubMed] [Google Scholar]
  • 7. Boulton AJ, Vileikyte L, Ragnarson‐Tennvall G, Apelqvist J. The global burden of diabetic foot disease. Lancet. 2005. Nov;366(9498):1719‐1724. [DOI] [PubMed] [Google Scholar]
  • 8. Portou MJ, Yu R, Baker D, Xu S, Abraham D, Tsui J. Hyperglycaemia and Ischaemia impair wound healing via toll‐like receptor 4 pathway activation in vitro and in an experimental murine model. Eur J Vasc Endovasc Surg. 2020. Jan;59(1):117‐127. [DOI] [PubMed] [Google Scholar]
  • 9. Rayman G. Diabetes and foot care: time to act. Diabet FOOT. 2005;8(3):120‐121. [Google Scholar]
  • 10. Lavery LA, Hunt NA, Ndip A, Lavery DC, Van Houtum W, Boulton AJM. Impact of chronic kidney disease on survival after amputation in individuals with diabetes. Diabetes Care. 2010. Nov 1;33(11):2365‐2369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Armstrong DG, Swerdlow MA, Armstrong AA, Conte MS, Padula WV, Bus SA. Five year mortality and direct costs of care for people with diabetic foot complications are comparable to cancer. J Foot Ankle Res. 2020. Dec;13(1):16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Iversen MM, Tell GS, Riise T, et al. History of foot ulcer increases mortality among individuals with diabetes: ten‐year follow‐up of the Nord‐Trondelag Health Study, Norway. Diabetes Care. 2009. Dec 1;32(12):2193‐2199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Boyko EJ, Ahroni JH, Smith DG, Davignon D. Increased mortality associated with diabetic foot ulcer. Diabet Med. 1996. Nov;13(11):967‐972. [DOI] [PubMed] [Google Scholar]
  • 14. Apelqvist J, Larsson J, Agardh CD. Long‐term prognosis for diabetic patients with foot ulcers. J Intern Med. 1993. Jun;233(6):485‐491. [DOI] [PubMed] [Google Scholar]
  • 15. Polikandrioti M, Vasilopoulos G, Koutelekos I, et al. Quality of life in diabetic foot ulcer: associated factors and the impact of anxiety/depression and adherence to self‐care. Int J Low Extrem Wounds. 2020. Jun;19(2):165‐179. [DOI] [PubMed] [Google Scholar]
  • 16. Skrepnek GH, Mills JL, Lavery LA, Armstrong DG. Health care service and outcomes among an estimated 6.7 million ambulatory care diabetic foot cases in the U.S. Diabetes Care. 2017. Jul;40(7):936‐942. [DOI] [PubMed] [Google Scholar]
  • 17. Driver VR, Fabbi M, Lavery LA, Gibbons G. The costs of diabetic foot: the economic case for the limb salvage team. J Vasc Surg. 2010. Sep;52(3):17 S‐22 S. [DOI] [PubMed] [Google Scholar]
  • 18. Hex N, Bartlett C, Wright D, Taylor M, Varley D. Estimating the current and future costs of type 1 and type 2 diabetes in the UK, including direct health costs and indirect societal and productivity costs: estimating current and future costs of type 1 and type 2 diabetes in the UK. Diabet Med. 2012. Jul;29(7):855‐862. [DOI] [PubMed] [Google Scholar]
  • 19. Hicks CW, Selvarajah S, Mathioudakis N, et al. Burden of infected diabetic foot ulcers on hospital admissions and costs. Ann Vasc Surg. 2016. May;33:149‐158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Kerr M, Barron E, Chadwick P, et al. The cost of diabetic foot ulcers and amputations to the National Health Service in England. Diabet Med. 2019. Aug;36(8):995‐1002. [DOI] [PubMed] [Google Scholar]
  • 21. Ray JA, Valentine WJ, Secnik K, et al. Review of the cost of diabetes complications in Australia, Canada, France, Germany, Italy and Spain. Curr Med Res Opin. 2005. Oct;21(10):1617‐1629. [DOI] [PubMed] [Google Scholar]
  • 22. Shen TNY, Kanazawa S, Kado M, et al. Interleukin‐6 stimulates Akt and p38 MAPK phosphorylation and fibroblast migration in non‐diabetic but not diabetic mice. PLoS One. 2017;12(5):e0178232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Pradhan L, Cai X, Wu S, et al. Gene expression of pro‐inflammatory cytokines and neuropeptides in diabetic wound healing. J Surg Res. 2011;167:336‐342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Wetzler C, Kämpfer 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. 2000. Aug;115(2):245‐253. [DOI] [PubMed] [Google Scholar]
  • 25. Theocharidis G, Thomas BE, Sarkar D, et al. Single cell transcriptomic landscape of diabetic foot ulcers [internet]. Genomics. 2022:10;13(1):181. [cited 2021 Nov 24]. doi: 10.1101/2021.03.11.434413 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Antezana MA, Sullivan SR, Usui ML, et al. Neutral endopeptidase activity is increased in the skin of subjects with diabetic ulcers. J Invest Dermatol. 2002;119:1400‐1404. [DOI] [PubMed] [Google Scholar]
  • 27. Wicks K, Torbica T, Mace KA. Myeloid cell dysfunction and the pathogenesis of the diabetic chronic wound. Semin Immunol. 2014;26:341‐353. [DOI] [PubMed] [Google Scholar]
  • 28. Goren I, Kämpfer H, Müller E, Schiefelbein D, Pfeilschifter J, Frank S. Oncostatin M expression is functionally connected to neutrophils in the early inflammatory phase of skin repair: implications for Normal and diabetes‐impaired wounds. J Invest Dermatol. 2006;123:628‐637. [DOI] [PubMed] [Google Scholar]
  • 29. Bannon P, Wood S, Restivo T, Campbell L, Hardman MJ, Mace KA. Diabetes induces stable intrinsic changes to myeloid cells that contribute to chronic inflammation during wound healing in mice. Vol. 6, Disease Models& Mechanisms. 2013. p. 1434–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Torbica T, Wicks K, Umehara T, et al. Chronic inflammation in response to injury: retention of myeloid cells in injured tissue is driven by myeloid cell intrinsic factors. J Invest Dermatol. 2019;139:1583‐1592. [DOI] [PubMed] [Google Scholar]
  • 31. Jiang WG, Sanders AJ, Ruge F, Harding KG. Influence of interleukin‐8 (IL‐8) and IL‐8 receptors on the migration of human keratinocytes, the role of PLC‐γ and potential clinical implications. Exp Ther Med. 2011;3:231‐236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Bickel M. The role of interleukin‐8 in inflammation and mechanisms of regulation. J Periodontol. 1993;64:456‐460. [PubMed] [Google Scholar]
  • 33. Edmonds ME. 10 ‐ the diabetic foot: pathophysiology and treatment. Clin Endocrinol Metab. 1986;15:889‐916. [DOI] [PubMed] [Google Scholar]
  • 34. Daeschlein G, Hinz P, Kiefer T, Jünger M. Rolle des Mikrobioms bei chronischen Wunden. Der Hautarzt, 2019;70:422‐431. [DOI] [PubMed] [Google Scholar]
  • 35. Prattichizzo F, Nigris VD, Mancuso E, et al. Short‐term sustained hyperglycaemia fosters an archetypal senescence‐associated secretory phenotype in endothelial cells and macrophages. Redox Biol. 2018;15:170‐181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Kirkland JL, Tchkonia T. Cellular senescence: a translational perspective. Lancet. 2017;21:21‐28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Tomic‐Canic M, DiPietro LA. Cellular senescence in diabetic wounds: when too many retirees stress the system. J Invest Dermatol. 2019;139(5):997‐999. [DOI] [PubMed] [Google Scholar]
  • 38. Wilkinson HN, Clowes C, Banyard KL, Matteuci P, Mace KA, Hardman MJ. Elevated local senescence in diabetic wound healing is linked to pathological repair via CXCR2. J Invest Dermatol. 2019;139:1171‐1181. [DOI] [PubMed] [Google Scholar]
  • 39. Prattichizzo F, Nigris VD, Sala LL, Procopio AD, Olivieri F, Ceriello A. ‘Inflammaging’ as a druggable target: a senescence‐associated secretory phenotype—centered view of type 2 diabetes. Oxid Med Cell Longev. 2016;2016:1‐10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Su N, Zhao N, Wang G, et al. Association of MCP‐1 rs1024611 polymorphism with diabetic foot ulcers. Medicine (Baltimore). 2018;97:e11232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Karima M, Kantarci A, Ohira T, et al. Enhanced superoxide release and elevated protein kinase C activity in neutrophils from diabetic patients: association with periodontitis. Pathophysiology. 2005;78:862‐870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Wong SL, Demers M, Martinod K, et al. Diabetes primes neutrophils to undergo NETosis, which impairs wound healing. Nat Med. 2015;21:815‐819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Hanses F, Park S, Rich J, Lee JC. Reduced neutrophil apoptosis in diabetic mice during staphylococcal infection leads to prolonged Tnfα production and reduced neutrophil clearance. PLoS One. 2011;6:e23633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Hellmann J, Zhang MJ, Tang Y, Rane M, Bhatnagar A, Spite M. Increased saturated fatty acids in obesity alter resolution of inflammation in part by stimulating prostaglandin production. J Immunol. 2013;191:1383‐1392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. den Dekker AD, Davis FM, Joshi AD, et al. TNF‐α Regulates Diabetic Macrophage Function Through the Histone Acetyltransferase MOF. JCI Insight; 2020;5(5):e132306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Goren I, Müller E, Schiefelbein D, et al. Systemic anti‐TNFα treatment restores diabetes‐impaired skin repair in ob/ob mice by inactivation of macrophages. J Invest Dermatol. 2007;127:2259‐2267. [DOI] [PubMed] [Google Scholar]
  • 47. Mirza RE, Fang MM, Novak ML, et al. Macrophage PPARγ and impaired wound healing in type 2 diabetes. J Pathol. 2015;236:433‐444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Yu T, Gao M, Yang P, et al. Insulin promotes macrophage phenotype transition through PI3K/Akt and PPAR‐γ signaling during diabetic wound healing. J Cell Physiol. 2018;234:4217‐4231. [DOI] [PubMed] [Google Scholar]
  • 49. Moura LIF, Silva L, Leal EC, Tellechea A, Cruz MT, Carvalho E. Neurotensin modulates the migratory and inflammatory response of macrophages under hyperglycemic conditions. Biomed Res Int. 2013;2013:1‐13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Morey M, O'Gaora P, Pandit A, Hélary C. Hyperglycemia acts in synergy with hypoxia to maintain the pro‐inflammatory phenotype of macrophages. PLoS One. 2019;14:e0220577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Khanna S, Biswas S, Shang Y, et al. Macrophage dysfunction impairs resolution of inflammation in the wounds of diabetic mice. PLoS One. 2010;5:e9539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Xu P, Fu X, Xiao N, et al. Involvements of γδT lymphocytes in acute and chronic skin wound repair. Inflammation. 2017;40:1416‐1427. [DOI] [PubMed] [Google Scholar]
  • 53. Traversa B, Sussman G. The role of growth factors, cytokines and proteases in wound management. Prim Intent Aus J Wound Manag. 2001;9:161‐167. [Google Scholar]
  • 54. Wrigley S, Arafa D, Tropea D. Insulin‐like growth factor 1: At the crossroads of brain development and aging. Front Cell Neurosci. 2017;11:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Yun YR, Won JE, Jeon E, et al. Fibroblast growth factors: biology, function, and application for tissue regeneration. J Tissue Eng. 2010;1:218142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Singh K, Agrawal NK, Gupta SK, Mohan G, Chaturvedi S, Singh K. Genetic and epigenetic alterations in toll like receptor 2 and wound healing impairment in type 2 diabetes patients. J Diabetes Complications. 2015. Mar;29(2):222‐229. [DOI] [PubMed] [Google Scholar]
  • 57. Singh K, Agrawal NK, Gupta SK, Sinha P, Singha K. Increased Expression of TLR9 Associated with pro‐Inflammatory S100A8 and IL‐8 in Diabetic Wounds Could Lead to Unresolved Inflammation in Type 2 Diabetes Mellitus (T2DM) Cases with Impaired Wound Healing, 2016;30(1):99‐108. [DOI] [PubMed] [Google Scholar]
  • 58. Dasu MR, Thangappan RK, Bourgette A, DiPietro LA, Isseroff R, Jialal I. TLR2 expression and signaling‐dependent inflammation impair wound healing in diabetic mice. Lab Invest. 2010. Nov;90(11):1628‐1636. [DOI] [PubMed] [Google Scholar]
  • 59. Davis FM, den Dekker A, Kimball A, et al. Epigenetic regulation of TLR4 in diabetic macrophages modulates immunometabolism and wound repair. J Immunol. 2020;204:2503‐2513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Singh K, Singh VK, Agrawal NK, Gupta SK, Singh K. Genetic alterations in toll‐like receptor 4 signaling pathway and impairment of wound healing in patients with type 2 diabetes. Int J Lower Extrem Wounds. 2014;13:162‐163. [DOI] [PubMed] [Google Scholar]
  • 61. Wlaschek M, Singh K, Sindrilaru A, Crisan D, Scharffetter‐Kochanek K. Iron and iron‐dependent reactive oxygen species in the regulation of macrophages and fibroblasts in non‐healing chronic wounds. Free Rad Biol Med. 2019;133:262‐275. [DOI] [PubMed] [Google Scholar]
  • 62. Soares MP, Hamza I. Macrophages and iron metabolism. Immunity. 2016;44:492‐504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Hsu I, Parkinson LG, Shen Y, et al. Serpina3n accelerates tissue repair in a diabetic mouse model of delayed wound healing. Cell Death Dis. 2014;5:e1458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Thandavarayan RA, Garikipati VNS, Joladarashi D, et al. Sirtuin‐6 deficiency exacerbates diabetes‐induced impairment of wound healing. Exp Dermatol. 2015;24:773‐778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Carow B, Rottenberg ME. SOCS3, a major regulator of infection and inflammation. Front Immunol. 2014;5:58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Linke A, Goren I, Bösl MR, Pfeilschifter J, Frank S. Epithelial overexpression of SOCS‐3 in transgenic mice exacerbates wound inflammation in the presence of elevated TGF‐β1. J Invest Dermatol. 2010;130:866‐875. [DOI] [PubMed] [Google Scholar]
  • 67. Kämpfer H, Schmidt R, Geisslinger G, Pfeilschifter J, Frank S. Wound inflammation in diabetic ob/ob mice. Diabetes. 2005;54:1543‐1551. [DOI] [PubMed] [Google Scholar]
  • 68. Wu M, Leng W, Pan H, et al. The reduced expression of EOLA1 may Be related to refractory diabetic foot ulcer. Mediators Inflamm. 2019;2019:1‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Prompers L, Huijberts M, Apelqvist J, et al. High prevalence of ischaemia, infection and serious comorbidity in patients with diabetic foot disease in Europe. Baseline results from the Eurodiale study. Diabetologia. 2007. Jan;50(1):18‐25. [DOI] [PubMed] [Google Scholar]
  • 70. Koh GCKW, Peacock SJ, Poll T van der, Wiersinga WJ. The impact of diabetes on the pathogenesis of sepsis. Vol. 31, Eur J Clin Microbiol Infect Dis 2012. p. 379–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Dowd SE, Wolcott RD, Sun Y, McKeehan T, Smith E, Rhoads D. Polymicrobial nature of chronic diabetic foot ulcer biofilm infections determined using bacterial tag encoded FLX amplicon pyrosequencing (bTEFAP). Egles C, PLoS One. 2008;3(10):e3326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Benwan KA, Mulla AA, Rotimi VO. A study of the microbiology of diabetic foot infections in a teaching hospital in Kuwait. J Infect Public Health. 2012. Feb;5(1):1‐8. [DOI] [PubMed] [Google Scholar]
  • 73. Wadachi R, Hargreaves KM. Trigeminal nociceptors express TLR‐4 and CD14: a mechanism for pain due to infection. J Dent Res. 2006;85:49‐53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. NICE . Diabetic foot problems: prevention and management [Internet]. 2019. https://www.nice.org.uk/guidance/ng19/chapter/Recommendations
  • 75. Aldana PC, Khachemoune A. Diabetic foot ulcers: appraising standard of care and reviewing new trends in management. Am J Clin Dermatol. 2020;21:255‐264. [DOI] [PubMed] [Google Scholar]
  • 76. Worsley A, Vassileva K, Tsui J, Song W, Good L. Polyhexamethylene biguanide:polyurethane blend nanofibrous membranes for wound infect control. Polymers. 2019;11(5):915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Shi C, Wang C, Liu H, et al. Selection of appropriate wound dressing for various wounds. Front Bioeng Biotechnol. 2020;8:182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Wynn M, Freeman S. The efficacy of negative pressure wound therapy for diabetic foot ulcers: a systematised review. J Tissue Viability. 2019;28:152‐160. [DOI] [PubMed] [Google Scholar]
  • 79. Wang T, Li X, Fan L, et al. Negative pressure wound therapy promoted wound healing by suppressing inflammation via down‐regulating MAPK‐JNK signaling pathway in diabetic foot patients. Diabetes Res Clin Pract. 2019;150:81‐89. [DOI] [PubMed] [Google Scholar]
  • 80. Ludwig‐Slomczynska AH, Borys S, Seweryn MT, et al. DNA methylation analysis of negative pressure therapy effect in diabetic foot ulcers. Endocr Connect. 2019;8:1474‐1482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Pawar DRL, Jeyapalina S, Hafer K, Bachus KN. Influence of negative pressure wound therapy on peri‐prosthetic tissue vascularization and inflammation around porous titanium percutaneous devices. J Biomed Mater Res. 2019;107:2091‐2101. [DOI] [PubMed] [Google Scholar]
  • 82. Norbury KC, Moyer MP. Effect of negative pressure therapy on the inflammatory response of the intestinal microenvironment in a porcine septic model. Mediators Inflamm. 2015;2015:1‐12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Rayman G, Vas P, Dhatariya K, et al. Guidelines on use of interventions to enhance healing of chronic foot ulcers in diabetes (IWGDF 2019 update). Diabetes Metab Res Rev [Internet]. 2020. [cited 2021 Nov 24];36(S1):e3283. doi: 10.1002/dmrr.3283 [DOI] [PubMed] [Google Scholar]
  • 84. Kranke P, Bennett MH, Martyn‐St James M, Schnabel A, Debus SE, Weibel S. Hyperbaric oxygen therapy for chronic wounds. Cochrane Wounds Group, editor. Cochrane Database Syst Rev. 2015;2015(6):CD004123. [cited 2021 Nov 24]. doi: 10.1002/14651858.CD004123.pub4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Roth RN, Weiss LD. Hyperbaric oxygen and wound healing. Clin Dermatol 1994. Jan 1;12(1):141–56. [DOI] [PubMed] [Google Scholar]
  • 86. Zhao LL. Effect of hyperbaric oxygen and growth factors on rabbit ear ischemic ulcers. Arch Surg. 1994;129(10):1043‐1049. [DOI] [PubMed] [Google Scholar]
  • 87. Thom SR. Hyperbaric oxygen: its mechanisms and efficacy. Plast Reconstr Surg. 2011;127:131 S‐141 S. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Brouwer RJ, Lalieu RC, Hoencamp R, van Hulst RA, Ubbink DT. A systematic review and meta‐analysis of hyperbaric oxygen therapy for diabetic foot ulcers with arterial insufficiency. J Vasc Surg. 2020. Feb;71(2):682‐692.e1. [DOI] [PubMed] [Google Scholar]
  • 89. Al‐Waili NS, Butler GJ. Effects of hyperbaric oxygen on inflammatory response to wound and trauma: possible mechanism of action. Sci World J. 2006;6:425‐441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Santema TB, Poyck PP, Ubbink DT. Skin grafting and tissue replacement for treating foot ulcers in people with diabetes. Cochrane wounds group, editor. Cochrane Database Syst Rev. 2016;2(2):CD011255. [cited 2021 Nov 24]. doi: 10.1002/14651858.CD011255.pub2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Greer N, Foman NA, MacDonald R, et al. Advanced wound care therapies for nonhealing diabetic, venous, and arterial ulcers. Ann Intern Med. 2013 Oct 15;159(8):532–542. [DOI] [PubMed] [Google Scholar]
  • 92. Vyas K, Vasconez H. Wound healing: biologics, skin substitutes. Biomembr Scaffolds Healthc. 2014;2(3):356‐400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Eaglstein WH, Iriondo M, Laszlo K. A composite skin substitute (Graftskin) for surgical wounds. Dermatol Surg. 1995;21(10):839‐843. [DOI] [PubMed] [Google Scholar]
  • 94. Veves A, Falanga V, Armstrong DG, Sabolinski ML. Graftskin, a human skin equivalent, is effective in the management of noninfected neuropathic diabetic foot ulcers: a prospective randomized multicenter clinical trial. Diabetes Care. 2001. Feb 1;24(2):290‐295. [DOI] [PubMed] [Google Scholar]
  • 95. Koob TJ, Lim JJ, Massee M, et al. Angiogenic properties of dehydrated human amnion/chorion allografts: therapeutic potential for soft tissue repair and regeneration. Vasc Cell. 2014;6(1):10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Koob TJ, Rennert R, Zabek N, et al. Biological properties of dehydrated human amnion/chorion composite graft: implications for chronic wound healing. Int Wound J. 2013. Oct;10(5):493‐500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Haugh AM, Witt JG, Hauch A, et al. Amnion membrane in diabetic foot wounds: a meta‐analysis. Plast Reconstr Surg Glob Open. 2017. Apr;5(4):e1302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Loeffelbein DJ, Rohleder NH, Eddicks M, et al. Evaluation of human amniotic membrane as a wound dressing for Split‐thickness skin‐graft donor sites. Biomed Res Int. 2014;2014:1‐12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Ueta M, Kweon MN, Sano Y, et al. Immunosuppressive properties of human amniotic membrane for mixed lymphocyte reaction: amniotic membrane suppresses MLR. Clin Exp Immunol. 2002. Sep;129(3):464‐470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. 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. 2013. Oct;10(5):502‐507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Sridharan K, Sivaramakrishnan G. Growth factors for diabetic foot ulcers: mixed treatment comparison analysis of randomized clinical trials. Br J Clin Pharmacol. 2017;84:434‐444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Perez‐Favila A, Martinez‐Fierro ML, Rodriguez‐Lazalde JG, et al. Current therapeutic strategies in diabetic foot ulcers. Medicina. 2019;55:714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Choi SY, Lee YJ, Kim JM, Kang HJ, Cho SH, Chang SE. Epidermal growth factor relieves inflammatory signals in Staphylococcus aureus‐treated human epidermal keratinocytes and atopic dermatitis‐like skin lesions in Nc/Nga mice. Biomed Res Int. 2018;2018:1‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Pierce GF, Mustoe TA, Altrock BW, Deuel TF, Thomason A. Role of platelet‐derived growth factor in wound healing. J Cell Biochem. 1991;45:319‐326. [DOI] [PubMed] [Google Scholar]
  • 105. Dumantepe M, Fazliogullari O, Seren M, Uyar I, Basar F. Efficacy of intralesional recombinant human epidermal growth factor in chronic diabetic foot ulcers. Growth Factors. 2015;33:128‐132. [DOI] [PubMed] [Google Scholar]
  • 106. Garcia‐Orue I, Gainza G, BorjaGutierrez F, et al. Novel nanofibrous dressings containing rhEGF and Aloe vera for wound healing applications. Int J Pharm. 2017;523:556‐566. [DOI] [PubMed] [Google Scholar]
  • 107. Park KH, Han SH, Hong JP, et al. Topical epidermal growth factor spray for the treatment of chronic diabetic foot ulcers: a phase III multicenter, double‐blind, randomized, placebo‐controlled trial. Diabetes Res Clin Pract. 2018;142:335‐344. [DOI] [PubMed] [Google Scholar]
  • 108. Agency M, Regulatory H. Products. Becaplermim (Regranex) for Diabetic Ulcers. Medicines and Healthcare products Regulatory Agency; 2014. https://www.gov.uk/drug-safety-update/becaplermin-regranex-for-diabetic-ulcers [Google Scholar]
  • 109. Papanas N, Maltezos E. Becaplermin gel in the treatment of diabetic neuropathic foot ulcers. Clin Interv Aging. 2008;3:233‐240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Lopes L, Setia O, Aurshina A, et al. Stem cell therapy for diabetic foot ulcers: a review of preclinical and clinical research. Stem Cell Res Ther. 2018;9(1):188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Hsiao STF, Asgari A, Lokmic Z, et al. Comparative analysis of paracrine factor expression in human adult mesenchymal stem cells derived from bone marrow, adipose, and dermal tissue. Stem Cells Dev. 2011;21:2189‐2203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Domaszewska‐Szostek A, Krzyzanowska M, Siemionow M. Cell‐based therapies for chronic wounds tested in clinical studies: review. Ann Plast Surg. 2019;83:e96‐e109. [DOI] [PubMed] [Google Scholar]
  • 113. Gorecka J, Gao X, Fereydooni A, et al. Induced pluripotent stem cell‐derived smooth muscle cells increase angiogenesis and accelerate diabetic wound healing. Regen Med. 2020;15:1277‐1293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Li M, Wang T, Tian H, Wei G, Zhao L, Shi Y. Macrophage‐derived exosomes accelerate wound healing through their anti‐inflammation effects in a diabetic rat model. Artif Cells Nanomed Biotechnol. 2019;47:3793‐3803. [DOI] [PubMed] [Google Scholar]
  • 115. Geiger A, Walker A, Nissen E. Human fibrocyte‐derived exosomes accelerate wound healing in genetically diabetic mice. Biochem Biophys Res Commun. 2015;467:303‐309. [DOI] [PubMed] [Google Scholar]
  • 116. Zhang J, Chen C, Hu B, et al. Exosomes derived from human endothelial progenitor cells accelerate cutaneous wound healing by promoting angiogenesis through Erk1/2 signaling. Int J Biol Sci. 2016;12:1472‐1487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Dalirfardouei R, Jamialahmadi K, Jafarian AH, Mahdipour E. Promising effects of exosomes isolated from menstrual blood‐derived mesenchymal stem cell on wound‐healing process in diabetic mouse model. Tissue Eng Regen Med. 2019;13:555‐568. [DOI] [PubMed] [Google Scholar]
  • 118. Ti D, Hao H, Tong C, et al. LPS‐preconditioned mesenchymal stromal cells modify macrophage polarization for resolution of chronic inflammation via exosome‐shuttled let‐7b. J Transl Med. 2015;13:308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Immonen JA, Zagon IS, McLaughlin PJ. Topical naltrexone as treatment for type 2 diabetic cutaneous wounds. Adv Wound Care. 2014;3:419‐427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Zhang X, Cui F, Chen H, et al. Dissecting the innate immune recognition of opioid inactive isomer (+)‐naltrexone derived toll‐like receptor 4 (TLR4) antagonists. J Chem Inf Model. 2018;58:816‐825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Cant R, Dalgleish AG, Allen RL. Naltrexone inhibits IL‐6 and TNFα production in human immune cell subsets following stimulation with ligands for intracellular toll‐like receptors. Front Immunol. 2017;8:809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Yang P, Wang X, Wang D, et al. Topical insulin application accelerates diabetic wound healing by promoting anti‐inflammatory macrophage polarization. J Cell Sci. 2020;133:jcs235838. [DOI] [PubMed] [Google Scholar]
  • 123. Rayahin JE, Buhrman JS, Gemeinhart RA, et al. High and low molecular weight hyaluronic acid differentially influence macrophage activation. ACS Biomater Sci Eng. 2015;1:481‐493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Mendes C, Haupenthal DP, dos S, et al. Effects of the association between Photobiomodulation and hyaluronic acid linked gold nanoparticles in wound healing. ACS Biomater Sci Eng. 2020;6:5132‐5144. [DOI] [PubMed] [Google Scholar]
  • 125. Minden‐Birkenmaier BA, Meadows MB, Cherukuri K, et al. Manuka honey modulates the release profile of a dHL‐60 neutrophil model under anti‐inflammatory stimulation. J Tissue Viability. 2020;29:91‐99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Malkoc M, Yaman SO, Imamoglu Y, et al. Anti‐inflammatory, antioxidant and wound‐healing effects of mad honey in streptozotocin‐induced diabetic rats. J Apicult Res. 2020;59:426‐436. [Google Scholar]
  • 127. Wallace HA, Basehore BM, Zito PM. Wound Healing Phases. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2020. https://www.ncbi.nlm.nih.gov/books/NBK470443/ [PubMed] [Google Scholar]
  • 128. Palta S, Saroa R, Palta A. Overview of the coagulation system. Indian J Anaesth. 2014;58:515‐523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Simon PE, Meyers AD. Skin Wound Healing. New York: eMedicine; 2020. https://emedicine.medscape.com/article/884594-overview#a1 [Google Scholar]
  • 130. Stadelmann WK, Digenis AG, Tobin GR. Physiology and healing dynamics of chronic cutaneous wounds. Am J Surg. 1998;176:26 S‐38 S. [DOI] [PubMed] [Google Scholar]
  • 131. Portou MJ, Baker D, Abraham D, Tsui J. The innate immune system, toll‐like receptors and dermal wound healing: a review. Vascul Pharmacol. 2015. Aug;71:31‐36. [DOI] [PubMed] [Google Scholar]
  • 132. Gomes A, Teixeira C, Ferraz R, Prudêncio C, Gomes P. Wound‐healing peptides for treatment of chronic diabetic foot ulcers and other infected skin injuries. Molecules. 2017;22:1743. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data sharing not applicable to this article as no datasets were generated or analysed during the current study.


Articles from International Wound Journal are provided here courtesy of Wiley

RESOURCES