Next Article in Journal
Correction: Fernandes et al. Stress Granule Assembly Can Facilitate but Is Not Required for TDP-43 Cytoplasmic Aggregation. Biomolecules 2020, 10, 1367
Next Article in Special Issue
Macrophages in Skin Wounds: Functions and Therapeutic Potential
Previous Article in Journal
Anthelmintic Activity and Cytotoxic Effects of Compounds Isolated from the Fruits of Ozoroa insignis Del. (Anacardiaceae)
Previous Article in Special Issue
Pinocembrin Ameliorates Skin Fibrosis via Inhibiting TGF-β1 Signaling Pathway
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bioactive Antimicrobial Peptides as Therapeutic Agents for Infected Diabetic Foot Ulcers

by
Jessica Da Silva
1,2,3,
Ermelindo C. Leal
1,3,* and
Eugénia Carvalho
1,3,*
1
Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal
2
PhD Programme in Experimental Biology and Biomedicine, University of Coimbra, 3004-504 Coimbra, Portugal
3
Institute of Interdisciplinary Research, University of Coimbra, Casa Costa Alemão, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal
*
Authors to whom correspondence should be addressed.
Biomolecules 2021, 11(12), 1894; https://doi.org/10.3390/biom11121894
Submission received: 10 November 2021 / Revised: 10 December 2021 / Accepted: 14 December 2021 / Published: 17 December 2021
(This article belongs to the Special Issue Cellular and Molecular Mechanisms of Wound Healing)

Abstract

:
Diabetic foot ulcer (DFU) is a devastating complication, affecting around 15% of diabetic patients and representing a leading cause of non-traumatic amputations. Notably, the risk of mixed bacterial–fungal infection is elevated and highly associated with wound necrosis and poor clinical outcomes. However, it is often underestimated in the literature. Therefore, polymicrobial infection control must be considered for effective management of DFU. It is noteworthy that antimicrobial resistance is constantly rising overtime, therefore increasing the need for new alternatives to antibiotics and antifungals. Antimicrobial peptides (AMPs) are endogenous peptides that are naturally abundant in several organisms, such as bacteria, amphibians and mammals, particularly in the skin. These molecules have shown broad-spectrum antimicrobial activity and some of them even have wound-healing activity, establishing themselves as ideal candidates for treating multi-kingdom infected wounds. Furthermore, the role of AMPs with antifungal activity in wound management is poorly described and deserves further investigation in association with antibacterial agents, such as antibiotics and AMPs with antibacterial activity, or alternatively the application of broad-spectrum antimicrobial agents that target both aerobic and anaerobic bacteria, as well as fungi. Accordingly, the aim of this review is to unravel the molecular mechanisms by which AMPs achieve their dual antimicrobial and wound-healing properties, and to discuss how these are currently being applied as promising therapies against polymicrobial-infected chronic wounds such as DFUs.

1. Introduction

Diabetes mellitus (DM) is a chronic disease, with a continuously increasing worldwide prevalence, that affected 463 million adults globally, in 2019 [1,2,3]. In Europe alone, the DM prevalence was about 59 million adults in 2019, and it is estimated to rise to over 68 million by 2045, representing an increase of 15% [1,3]. Similarly, the DM-associated complications are also expected to increase [1,2,3,4]. Indeed, diabetic foot and lower limb complications affect between 40 to 60 million people globally, representing an important source of morbidity in people with DM [1,3]. About 15% of patients with DM will develop foot ulcers in their life time, requiring prolonged hospitalizations and amputations in 85% of the cases [1,2,3,4,5,6].
A diabetic foot ulcer (DFU) is a devastating and costly complication of diabetes, consisting of deep tissue lesions associated with both peripheral neuropathy and peripheral vascular disease [7,8]. DFU represents a severe public health problem with an urgent need for new effective treatments, which are crucial to reduce the associated high morbidity and mortality rates, as well as to reduce the economic and social burden [2,6,8]. The persistent hyperglycemia, chronic inflammation, hypoxia, peripheral neuropathy, impaired angiogenesis, and difficulty to fight infections in diabetes are factors that impair the wound healing progress. Importantly, around 60% of DFUs become infected, predominantly with bacterial colonies of S. aureus and C. striatum, and fungal colonies of C. albicans [9,10,11,12,13,14,15,16]. Moreover, anaerobic bacteria of the Bacteroidales order, namely Bacteroides spp. and Prevotella spp., have also associated with non-healing DFUs, whereas fungal pathogens have been highly associated with wound necrosis and poor clinical outcomes [17,18,19,20]. Still, the literature is almost exclusively focused on Gram-positive and Gram-negative bacteria, and few studies have considered the prevalence of anaerobic bacteria and fungi in DFUs. Therefore, multi-kingdom microbiome infection control is imperative for the management of this kind of infected wounds. Indeed, wound microbiota and microbial biofilms are thought to contribute to the failure of chronic wound to heal; hence, the control of pathogen infection is a good therapeutic solution, since it can improve the microenvironment and promote sustained healing over time.
Antimicrobial peptides (AMPs) are endogenous peptides found in different organisms, particularly in the skin, that act as a first line of defense against infection [21,22]. Furthermore, these molecules not only play key roles in fighting infection through broad-spectrum antimicrobial activity against Gram-positive and Gram-negative bacteria, viruses, and fungi, but they also play important roles in wound healing [23,24,25,26]. Nonetheless, endogenous AMP expression and/or activity can be dysfunctional under certain conditions, such as diabetes, making them not able to promote adequate healing and fight infection at the wound site. Accordingly, suitable therapeutic strategies for the management of polymicrobial-infected wounds should rely on the performance of chemical modifications and/or the use of novel delivery systems for exogenous AMPs, to increase their stability, reduce their toxicity, enhance their dual antimicrobial and wound-healing activities, and improve their targeting and prolonged delivery at the wound site.
Therefore, this review aims to describe the molecular mechanisms by which AMPs achieve their dual multi-kingdom antimicrobial and wound-healing properties. In addition, it will describe how these AMPs are currently being applied as promising therapies to combat polymicrobial infection in DFUs.

2. Diabetic Foot Infection

Wound healing comprises a complex and dynamic series of cellular and biochemical events which consists of the following four overlapping phases: hemostasis, inflammation, proliferation, and remodeling [5,27,28,29]. The hemostasis phase begins with constriction of the injured blood vessels and activation of platelets to form a fibrin clot to stop the bleeding [27,30]. Subsequently, the inflammatory phase initiates with the recruitment of neutrophils to the clot as a first line of defense against pathogens to remove debris, in order to provide a propitious environment for wound healing [27,28,30]. Neutrophils reach their peak population between 24 and 48 h after injury, after which they reduce greatly in number, and macrophages, in turn, arrive at the wound site and continue clearing debris [27,28]. Macrophages secrete growth factors and proteins that attract adaptive immune system cells to the wound site, such as Langerhans cells, dermal dendritic cells and T cells, which are involved either in the clearance of cellular debris or in the combat of infection [27,28]. Once the wound has been cleaned out and the inflammation decrease, the proliferative phase occurs with the following three different stages: filling of the wound with granulation tissue, contraction of the wound margins, and covering of the wound with epithelial cells, also called re-epithelization [27,28,30]. Finally, the remodeling phase takes place with collagen fiber reorganization, tissue remodeling and maturation, and an overall increase in the tensile strength can be observed [27,28,30].
However, besides its complexity, the healing process is also susceptible to interruption or delay, due to impairment of local and systemic factors that are important in the healing process. Chronic non-healing wounds often develop in people with diabetes. If wounds do not heal within 12 weeks, they are defined as chronic wounds according to the Food and Drug Administration [29,31,32]. Furthermore, in the presence of conditions such as hyperglycemia, chronic inflammation, hypoxia, peripheral neuropathy, impaired angiogenesis, and infection the wound healing progress in diabetes can become stalled [31,32].
Diabetic foot infections (DFIs) are defined by invasion and multiplication of microorganisms in diabetic non-healing wounds and are associated with tissue destruction and/or alterations in the host’s inflammatory response [9,33,34]. DFIs are among the most serious and frequent complications in people with diabetes. They are estimated to develop in about 60% of all DFU cases and represent an important source of morbidity in these patients [9,35,36,37]. Several aspects of the wound microbiology influence the development of DFI, including the microbial load, the microbe diversity, the existence of pathogenic microorganisms, and the synergistic association amongst microbial species [9,33,35,38]. Among the most predominantly identified bacteria in DFUs are not only Gram-positive bacteria, such as S. aureus (MSSA—methicillin-susceptible Staphylococcus aureus, and MRSA—methicillin-resistant Staphylococcus aureus), Streptococcus β-hemolytic and C. striatum, but also Gram-negative bacteria, such as P. aeruginosa, E. coli, A. baumannii, Proteus spp., Enterobacter spp., and Citrobacter spp., in addition to some anaerobes deeper in the wound bed, such as Bacteroides spp., Prevotella spp., Clostridium spp., and Peptostreptococcus spp. (Table 1).
Furthermore, DFUs have a polymicrobial basis, and the risk for the diabetic foot syndrome development is mostly associated with mycotic infections [11,12,13,14,33]. However, few studies have considered the prevalence of fungal colonies in DFUs. Indeed, more than a quarter of DFUs undergo fungal infection, but remain undetected or undiagnosed by regular and standard microbiology laboratory protocols in the DFU clinics, in most cases, as it also happens with anaerobic bacteria [14,20,39,40]. It has also been demonstrated that patients with higher systemic glycosylated hemoglobin levels, such as diabetic patients, have significantly more fungal infections, which contribute to delayed wound healing [14]. Importantly, the mycobiome represents a scaffold for bacterial attachment and provides additional protection from external threats, promoting the formation of multi-kingdom biofilms [19,20]. Moreover, increased fungal pathogens in DFUs have been highly associated with wound necrosis and poor clinical outcomes [19,20]. The fungi most commonly isolated are Candida spp., Trichophyton spp., Aspergillus spp., Trichosporon spp., and Cladosporium herbarum (Table 1).
The formation of microbial biofilm in DFUs, defined as a structured arrangement of microorganisms in a self-produced polysaccharide matrix with transformed phenotype and growth patterns, has been related to wound chronicity and infection [9,35,38,41]. Biofilms may be explained by the organization of these microorganisms into functionally equivalent pathogroups (FEP) in DFUs, where pathogenic and commensal microorganisms co-aggregate symbiotically in a pathogenic biofilm for more efficient nutrient cycling and enhanced protection from external threats, further promoting chronic infection [17,35,42,43]. Additionally, it is noteworthy that biofilm-forming microbial colonies are 10 to 1000 times more resistant to antimicrobials, including both antibiotics and antiseptics, in comparison with planktonic ones, which consists of free-floating microorganisms. Therefore, it is urgent to find effective treatments for chronic infected DFUs with a polymicrobial basis. The combination of multidisciplinary treatment approaches should help to overcome some of the DFI-related hurdles [17,35,38]. As a result, the role of AMPs with antifungal activity in wound management needs to be considered and further investigated, in association with antibacterial agents, such as antibiotics and AMPs with antibacterial activity, or alternatively the application of a broad-spectrum antimicrobial agent that targets both bacteria and fungi.

3. Antimicrobial Peptides

Antimicrobial peptides (AMPs), also known as endogenous host defense peptides, are naturally abundant peptides found in bacteria, plants, insects, amphibians, reptiles, and mammals. These peptides play essential roles in the innate immune response and contribute to the first line of defense against infection [21,22,26,45]. Upon injury and infection, the innate immune system is activated and leads to the production of these small molecules by different resident cells of the skin such as keratinocytes, the predominant cell type of the epidermis [26,41,42,43]. Indeed, pathogen-associated molecular patterns (PAMPs), such as lipoarabinomannan, lipopolysaccharides and proinflammatory cytokines, are recognized by the innate immune system, leading to the up-regulation and overexpression of AMPs to promote a fast and effective response to injury and infection [23,29,45,46].
AMPs are composed of 15 to 50 amino acids, are generally positively charged, form amphipathic structures, and are classified into different categories according to their primary structures and topologies, including human endogenous β-defensins (hBDs) 1–3, cathelicidin antimicrobial peptide (LL-37) and dermcidins [41,45,46,47,48,49]. The two most predominant types of AMPs in human skin include hBDs and cathelicidins, particularly hBDs 1–3 and LL-37, with their primary, secondary and tertiary structures, and their related physicochemical properties presented in Table 2 [30,46,49,50]. These physicochemical properties, including length, molecular weight (MW), isoelectric point (pI), net charge, and hydrophobicity, are important to predict their antimicrobial potential for further clinical application.
The hBDs 1–3 and LL-37 have a peptide length below 50 amino acids (aa) and a relatively similar MW, thereby being referred to as AMPs, but also as small peptides (Table 2). The small length and low MW of these peptides can promote their insertion of the peptide into the microbial membrane, contributing to their higher antimicrobial activity [51]. Moreover, all four of these endogenous AMPs exhibit a high positive net charge, ranging from +4 to +11, and a relatively similar isoelectric point, ranging from 8.55 to 11.15 (Table 2). This net positive charge is a requirement for their antimicrobial potential, in order to permeabilize the negatively charged membranes of microbes [22,46,47,52]. In addition, the hydrophobicity properties are also crucial for partial or total insertion of AMPs into the membrane’s hydrophobic core [51]. This AMP membrane insertion will enable the destabilization of the bilayer and/or promote the cell depolarization, denoting the importance of a high AMP hydrophobicity for antimicrobial potential [51]. All four peptides (hBDs 1–3 and LL-37) exhibit a high hydrophobicity value ranging from +28.98 to +45.26 kcal/mol, another important property highlighting their antimicrobial potential (Table 2). Furthermore, the secondary and tertiary structure properties are another key feature influencing the biological function of these small peptides [51]. Regarding the secondary structure, hBDs 1–3 present a mixed α-helix + β-strand conformation, whereas LL-37 exhibits only an α-helix arrangement (Table 2) [30,49], which are the most common conformations in AMPs [53]. In regard to their tertiary structure, hBDs 1–3 present a relatively similar “defensin-like” topology, i.e., a core consisting of three antiparallel β-sheets interconnected with three intramolecular disulfide bridges flanked by an α-helix segment, all together stabilized by a disulfide bridge, making them members of the defensin family. On the other hand, LL-37 presents a predominant α-helical conformation, making it the only human member of the cathelicidin family (Table 2) [30,52,54]. As a result, these physicochemical properties greatly influence the activity and the potential of AMPs, therefore highlighting the need for the inclusion of such parameters when evaluating AMPs and selecting them for further clinical application.
Besides their well-known broad-spectrum antimicrobial activity against Gram-positive and Gram-negative bacteria, viruses, and fungi, some AMPs also play key roles in wound healing by promoting cell migration and proliferation, angiogenesis, chemokine and cytokine production, and wound closure (Figure 1) [23,24,25,26,50]. Therefore, these aforementioned AMPs are usually referred to as peptides with dual antimicrobial and wound-healing properties [46].
AMPs can achieve direct eradication of microbes by disrupting microbial membranes through pore formation and by interacting with intracellular targets, such as hBD-2, hBD-3 and LL-37 (Figure 1) [22,47,49]. This antimicrobial mechanism of action by the disruption of microbial membranes is based on the permeabilization of negatively charged membranes of the microbes followed by microbial lysis, due to the positive charge of AMPs [22,46,47]. On the other hand, AMPs can also modulate the host immune system by the recruitment and activation of immune cells through the induction of chemokine and cytokine production, and, therefore, enhancing indirect pathogen killing and clearance and controlling inflammation, namely hBD-2, hBD-3, LL-37 and dermcidin-1L (Figure 1) [25,30,47,49].
Importantly, some AMPs are also able to promote re-epithelization and wound closure through activation of receptor-signaling mechanisms responsible for cell proliferation and migration, such as hBD-2, hBD-3 and LL-37 (Figure 1) [54]. In addition, they can also support angiogenesis by the induction of endothelial cell tube formation and up-regulation of angiogenic proteins, namely LL-37 (Figure 1) [54]. Furthermore, they can enhance extracellular matrix synthesis, promote the contraction capacity of fibroblasts by inducing fibroblast-to-myofibroblast differentiation, and enhance wound healing by increasing α-smooth muscle actin expression by fibroblasts (Figure 1) [23,47,49,55].
These small peptides, both from natural sources or synthetically produced, have been investigated in depth in the last years, and some of them are even in clinical trials. Indeed, LL-37 (ropocamptide) from Promore Pharma AB is currently under phase IIb for treating venous leg ulcers [55], and pexiganan (Locilex®) from Dipexium Pharmaceuticals, Inc., an analogue of peptide magainin II from frog skin of Xenopus laevis, was under phase III for treating mild infected DFUs [52]. Unfortunately, Locilex® was discontinued for not meeting greater primary or secondary clinical endpoints versus the vehicle plus standard of care [52]. Moreover, FirstString Research, Inc. also developed a Granexin® Gel that contains the synthetic aCT1 peptide for treating DFUs [56]. This formulation was under phase III until May 2020, and has since been terminated without safety or efficacy concerns, with final data to be published [56].
Therefore, AMPs need to be further investigated as promising alternatives to conventional antibiotics to overcome the emergence of multidrug-resistant (MDR) microorganisms and as an attractive strategy for polymicrobial-infected DFUs, due to their dual antimicrobial and wound-healing properties [21,25,47,57].

4. Changes of Endogenous AMPs in DFUs

Wound healing and infection control are efficiently carried out in the skin by AMPs and other molecules, such as growth factors. Important endogenous AMPs participating in these events include hBDs, LL-37, and dermcidins, which are naturally abundant in different organisms, particularly in the skin [23,53,58]. However, their expression levels and/or activity may be altered under certain conditions, including diabetes, leading to inadequate infection control, and contributing to impaired wound healing.
Lan et al. have shown that when human keratinocytes isolated from normal adult foreskin are cultured in vitro under a high glucose environment for 7 days, hBD-2 expression is reduced through the downregulation of signal transducer and activator of transcription 1 (STAT-1) signaling [59]. Indeed, STAT-1 is a transcription factor that is involved in the upregulation of many genes, due to a signal by either type I, II or III interferons, suggesting that functional STAT-1 signaling is required to achieve optimal hBD-2 transcription. In addition, the skin of streptozotocin (STZ)-induced diabetic rats showed inadequate β-defensin expression after wounding compared with skin from control rats, contributing to poor diabetic wound healing [59]. Moreover, Gonzalez-Curiel et al. have determined that patients with type 2 diabetes express lower levels of CAMP (LL-37) and DEFB4 (hBD-2) genes in peripheral blood cells, which could explain the higher susceptibility to infectious diseases [23]. Moreover, Galkowska et al. have revealed that chronic wounds, grade 2–4 DFUs according to the Wagner’s classification and venous calf ulcers, present underexpression of hBD-2 in comparison to normal skin, which may point to the involvement of this peptide in the chronicity of ulcers [60]. Conversely, Rivas-Santiago et al. have demonstrated that hBDs were overexpressed in biopsies from grade 3 DFUs according to the Wagner’s classification, whereas LL-37 is under expressed or absent in comparison with biopsies from healthy skin donors [50]. Although Rivas-Santiago et al. found that hBDs are expressed in DFUs, their activity seems to be inefficient to fight infection and promote proper wound healing [50].
All together, these results suggest that though some endogenous AMPs are expressed in DFU, their expression level and activity is not appropriate, highlighting the need to restore the expression level and enhance the activity of these peptides at the wound site. When doing this, one needs to bear in mind factors that weaken their function, such as those found in the diabetic microenvironment, protease degradation and serum inactivation.
Nonetheless, neither the increase of PAMPs to induce up-regulation and overexpression of AMPs nor the increase of AMPs itself should be used as therapeutic approaches, due to undesirable side effects in patients that PAMPs may induce and due to the potential toxicity of free AMPs, respectively [23,48]. Therefore, a suitable strategy may rely on the performance of chemical modifications and/or the use of delivery systems, in order to increase the stability of these peptides in the DFU microenvironment, reduce their toxicity, enhance their dual antimicrobial and wound-healing activities, and improve their targeting and prolonged delivery, including deeper in the wound bed [41,48].

5. Endogenous and Synthetic AMPs as Promising Therapeutic Agents for Infected Wounds

Several AMPs are being studied as promising therapies to combat infected non-healing wounds, and some of them are even in clinical trials as formerly referred, either free or loaded onto a delivery system. Table 3 and Table 4 summarize the AMPs being explored as promising therapies for chronic non-healing wounds and their respective roles in antimicrobial and wound-healing activities, either free or loaded on a delivery system, respectively. All AMPs are presented according to the following criteria: (1) free vs. chemically modified or loaded on a delivery system vs. chemically modified and loaded on a delivery system; (2) endogenous vs. synthetic; and (3) alphabetic order.

5.1. Free AMPs

In recent years, bare AMPs have been broadly investigated to unravel their dual antimicrobial and wound-healing properties to be used as therapeutic approaches for infected chronic wounds (Table 3). One example is the study by Gonzalez-Curiel et al., who have demonstrated in vitro, that free endogenous hBD-2, a 41-amino acid peptide, has antimicrobial activity against E. coli and promotes keratinocyte migration in primary cultures developed from skin biopsies of patients with DFUs, at a concentration of 0.8 µg/mL [23]. Similar results were also obtained with LL-37, a peptide consisting of 37 amino acids, at a concentration of 0.1 µg/mL [23]. In addition, Liu et al. have studied CW49, a short peptide with an 11-amino acid sequence, identified in frog skin of Odorrana graham. This peptide was shown to have strong angiogenic ability and a significant anti-inflammatory effect, but it had little effect on re-epithelialization, being applied twice a day for 12 days at a concentration of 200 µg/mL in full-thickness dermal wounds, in both normal and diabetic animals [61]. Moreover, Steinstraesser et al. demonstrated the lack of cytotoxicity towards immortalized human keratinocyte HaCaT cells and primary human fibroblasts of synthetic innate defense regulator-1018 (IDR-1018), a 12-amino acid peptide, compared to LL-37, at a concentration of up to 200 µg/mL (p < 0.0001) [62]. IDR-1018 also exhibited enhanced wound-healing and antimicrobial potential in S. aureus-infected porcine wounds, as well as in non-diabetic murine wounds at a concentration of 200 µg/mL, applied every 48 h for 10 or 14 days [62]. However, this wound-healing effect was not demonstrated in the diabetic murine wounds, suggesting that one or more signaling pathways by which IDR-1018 acts may be impaired in the diabetic animals [62]. Additionally, Marin-Luevano et al. have also unraveled the properties of IDR-1018 in a high glucose environment [57]. This peptide was shown to have angiogenic ability and anti-inflammatory effect at both concentrations of 25 or 50 µg/mL, as well as promote the migration of endothelial cells under conditions of hypoxia at a concentration of 25 µg/mL, while epithelial cells migration increased only under conditions of normoxia [57]. Furthermore, pexiganan is another peptide, with a 22-amino acid sequence that has been studied, particularly for its broad-spectrum antimicrobial activity. According to Flamm et al., pexiganan has demonstrated antimicrobial activity against several Gram-positive and Gram-negative bacteria, including E. coli, E. cloacae, Citrobacter species, P. vulgaris, M. morganii, K. pneumoniae, S. marcescens, P. aeruginosa, A. baumannii (resistant to ≥4 antimicrobials), S. agalactiae, S. pyogenes, E. faecium, and even MSSA and MRSA, with an overall minimum inhibitory concentration (MIC) of MIC50 = 16 µg/mL, excepted for M. morganii, P. vulgaris, and S. marcescens that presented MIC50 > 512 µg/mL [58].
Despite several studies unravelling the properties of these promising peptides to be used as therapeutic approaches for chronic infected wounds, as alternatives to antibiotics, free AMPs still present limited activity due to their susceptibility to the microenvironment found in non-healing wounds, and their inherent toxicity [41,63]. A way to overcome some of these hurdles has been to perform chemical modifications of the peptides (Table 3). Among the main chemical modifications that have been investigated and have shown specific advantages to improve specific AMP characteristics are: lipidation (covalent binding of a lipid group to a peptide); glycosylation (covalent attachment of a glycan, or also termed carbohydrate, to a peptide); guanidination (conversion of some or all of the lysine residues to homoarginine residues); hydrazidation (binding of a hydrazide to a peptide); and small molecule conjugation (incorporation of a small molecule in AMPs, such as antibiotics, ionic liquids, or even small peptides, among others) [64,65]. Lipidation and glycosylation have mainly been explored as chemical modifications for solubility and activity improvement and stability enhancement against protease degradation, whereas the other chemical modifications have also been applied for antimicrobial activity enhancement [64,65]. Gomes et al. have synthesized 3.1-PP4, a 16-amino acid peptide, by developing hybrid peptide constructs combining the wide spectrum antimicrobial peptide 3.1 and the collagen-inducing peptide PP4, one important example of small molecule conjugation of two peptides [66]. This hybrid peptide had low toxicity towards HFF-1 human fibroblasts (half maximal inhibitory concentration—IC50 = 134 ± 10 µg/mL) and antimicrobial potential against E. coli (MIC = 2 µg/mL), P. aeruginosa (MIC = 4.2 µg/mL), and even against MDR isolates of K. pneumoniae, E. coli, and P. aeruginosa (1 < MIC < 8.2 µg/mL) [66]. The peptide 3.1-PP4 also interfered with the formation of K. pneumoniae biofilms of resistant clinical isolates [66]. More recently, they have also synthesized its N-methyl imidazole derivative MeIm-3.1-PP4, a chemical modification that improved its solubility and enzymatic stability towards tyrosinases. In addition, PP4-3.1 and its N-methyl imidazole derivative MeIm-PP4-3.1 were also synthesized via another chemical modification that could broaden its spectrum activity, including against fungal pathogens, relative to its reversed isomer 3.1-PP4 [67]. Interestingly, PP4-3.1 showed the highest activity against Gram-positive and Gram-negative bacteria, including MDR isolates (0.8 ≤ MIC ≤ 5.7 µM), either in planktonic or biofilm form, as well as against relevant Candida spp. [67]. It is noteworthy that MeIm-PP4-3.1 was almost twice more cytotoxic than PP4-3.1 in HaCaT cells (IC50 = 5.7 ± 1.0 or 13.0 ± 1.0 µM, respectively), highlighting the higher potential of PP4-3.1 [67]. Furthermore, Mi et al. have produced A-hBD-2, a 41-amino acid peptide, through the replacement of the GIGDP unit on the N-terminal of hBD-2 by APKAM [26]. This modification improved the structural stability of hBD-2 and led to no cytotoxicity in HaCaT cells, at a concentration of up to 100 µg/mL after 24–72 h of incubation, while also improving its antimicrobial activity against S. aureus, at concentrations of 50, 70, and 100 µg/mL, when compared to hBD-2 [26]. Moreover, these authors demonstrated the potential of A-hBD-2 to promote migration and proliferation of keratinocytes at a concentration of 20 µg/mL, via the phosphorylation of epidermal growth factor receptor (EGFR) and the signal transducer and activator of transcription 3 (STAT3). In addition, A-hBD-2 (20 µg/mL) decreased the terminal differentiation of keratinocytes, enhanced the mobilization of intracellular calcium ions (Ca2+), and promoted wound healing of full-thickness wounds in a rat model, suggesting that A-hBD-2 may be a promising candidate therapy for chronic wounds [26]. Similarly, Mouritzen et al. have assessed the potential of bovine lactoferricin (LFcinB), a 25-amino acid peptide derived from acidic hydrolysis of bovine lactoferrin (bLf), in diabetic wound healing [68]. LFcinB promoted keratinocyte migration in vitro and ex vivo at a concentration of 25 µg/mL, and enhanced wound healing in a type 1 diabetic mouse model at both concentrations of 12.5 or 25 µg/wound, applied topically over 10 consecutive days (twice the first 2 days and then once daily) [68]. Moreover, LFcinB had antimicrobial activity against B. pumilus and S. aureus, and increased S. xylosus prevalence, a commensal bacterium of the skin, in the type 1 diabetic mouse model [68]. It was also shown to induce angiogenesis and collagen deposition, while decreasing oxidative stress and the M1/M2 macrophage ratio, suggesting a reduction of inflammation in wounds of this diabetic mouse model [68]. In turn, Kim et al. have synthesized SHAP1, a 19-amino acid peptide, through the addition of the two capping motifs APKAM and LQKKGI into the N- and C-terminal ends, respectively, to ensure structural stability of the secondary structure of the entire peptide, irrespective of surrounding salt concentration [69]. They also showed that this peptide had no cytotoxicity towards human erythrocytes and HaCaT cells up to a concentration of 200 µM, and it proved to have greater stability to protease exposure in the wound fluid, such as human neutrophil elastase and S. aureus V8 proteinase [69]. Moreover, they revealed that the SHAP1 peptide had stronger wound closure activity compared to LL-37 in vitro by inducing HaCaT cell migration, at a concentration of 1 µM, and accelerated healing of full-thickness excisional wounds in mice at a concentration of 1 µM/wound, applied one time a day for 2 days post-injury. In addition, it had potent antimicrobial activity against S. aureus, and enhanced wound healing in S. aureus-infected murine wounds at a concentration of 1 µM/wound, applied one time a day for 2 days post-injury [69]. Finally, Tomioka et al. have developed a synthetic stable and short peptide, SR-0379, with 20 amino acids, including a lysine residue that has been converted to D-lysine to improve its stability [47]. This peptide (10 µg/mL) promoted the proliferation of normal human dermal fibroblast cells via the PI3 kinase-Akt-mTOR pathway through integrin-mediated interactions. SR-0379 also revealed antimicrobial activity not only against bacteria (E. coli, P. aeruginosa, and S. aureus), including drug-resistant bacteria (MRSA and A. baumannii MDR), but also against fungi (C. krusei, T. mentagrophytes, and T. rubrum) [47]. Additionally, they also demonstrated that SR-0379 induced wound healing in vivo in the following two different wound-healing models in rats: full-thickness wounds under a diabetic conditions (at a concentration of 200 µg/mL, applied at each time point—days 0, 6, 13 and 20) and acutely infected full-thickness wounds with S. aureus (at a concentration of 1000 µg/mL, applied at each time point—days 8 and 15) [47].

5.2. Loaded AMPs on Delivery Systems

Nonetheless, chemical modifications may not be enough to fully improve the properties of bare AMPs. Therefore, other appropriate strategies have been made available, namely the encapsulation in delivery systems. This will not only increase the stability of these peptides in the DFU microenvironment, but it may also reduce their inherent toxicity, and enhance their dual antimicrobial and wound-healing capacity [41,48,63]. In addition, it will improve their targeting and prolonged their delivery, optimizing their effectiveness for treating non-healing infected wounds [41,48,63]. Different approaches have been developed in this way (Table 4). Bolatchiev et al. have developed a niosomal gel made of silicon to encapsulate separately the human neutrophil peptide-1 (HNP-1, or α-defensin-1), a 75-amino acid peptide, or the human β-defensin-1 (hBD-1), a 47-amino acid peptide. They have demonstrated their antimicrobial activity against MRSA-infected wounds in rats, and against MSSA and MRSA isolated from patients with DFIs, at peptide concentrations of 2 or 1 µg/mL, respectively (MIC = 1 µg/mL for MSSA and MIC = 0.5 µg/mL for MRSA) [70]. Importantly, these authors have demonstrated that there was no in vitro synergistic action based on the calculated fractional inhibitory concentration index of these two peptides in combination with cefotaxime, a third-generation broad-spectrum bactericidal cephalosporin antibiotic, against MSSA, as well as against MRSA, since this strain of staphylococci has natural resistance to cefotaxime [70]. Another delivery system that has been tested by Santos et al. is a guar gum gel used for the topical delivery of nisin, a 34-amino acid peptide that belongs to class I bacteriocins and is produced by the bacteria Lactococcus lactis [25,71]. In 2016, this guar gum gel, used as a nisin delivery system, was shown to exhibit antimicrobial activity against S. aureus biofilm-producing isolates collected from DFU patients, including multidrug-resistant clinical isolates (overall MIC = 180.8 53.9 µg/mL) [25]. A few years later, it was shown to have an even higher inhibitory efficacy against S. aureus biofilm formation from DFI patients, when combined with chlorhexidine, an antiseptic agent, at a peptide concentration of 22.5 µg/mL and at an antiseptic agent concentration of 6 µg/mL, suggesting a synergistic action between the peptide and the antiseptic agent, contrary to Bolatchiev et al. [71]. However, no significant differences were shown between the efficacy of this combination and the conventional antibiotic-based protocols regarding biofilm eradication [71]. Furthermore, Grek et al. have developed a hydroxyethyl cellulose gel, called Granexin®, for the topical delivery of aCT1, a 25-amino acid synthetic peptide mimetic of the C-terminus of connexin43 (Cx43), which is known to have roles in dermal wound healing [56]. This formulation, applied topically with a peptide concentration of 100 µM on days 0 and 3, and then weekly from weeks 1–12, was shown to decrease ulcer areas, to promote ulcer re-epithelialization, and to decrease time-to-complete-ulcer closure in DFU patients within a randomized, investigator-blinded, multi-center clinical trial [56]. It is noteworthy that aCT1-containing Granexin® was under a phase III clinical trial until May 2020, and has since been terminated without safety or efficacy concerns, with final data to be published [56].
Similarly, Bayramov et al. have assessed the efficacy of the following three different types of formulations: a gel containing 1.5% peptide, a Stratex® dressing coated with ASP-1 gel to obtain 0.74 mg/cm2 peptide, and a hydrophilic polyurethane (PU)-based dressing containing 0.66 mg/cm2 peptide, for the topical delivery of the following two synthetic peptides separately: ASP-1, a 24-amino acid peptide and ASP-2, a 12-amino acid peptide [72]. All the three delivery systems loaded either with ASP-1 or ASP-2 induced in vitro eradication of mono- and polymicrobial biofilms of MDR pathogens, including S. aureus, A. baumannii, K. pneumoniae, P. aeruginosa, and MRSA, and presented a higher biocompatibility index (BI) when compared to free ASP-1 or free ASP-2, with a more favorable BI for ASP-2, in primary human epidermal fibroblast cells [72]. In turn, Zhao et al. have developed a glucose oxidase (GOx)-loaded hydrogel formed by the self-assembly of an heptapeptide, IKYLSVN, known for its antimicrobial properties, with a peptide concentration of 10 mg/mL [73]. This formulation was shown to have antimicrobial activity against S. aureus in vitro cultures and to reduce blood glucose concentration of diabetic patients [73]. Additionally, Comune et al. have developed a gold-nanoscale formulation (gold nanoparticles—Au NPs) to carry synthetic LL-37, a modified form of the endogenous LL-37 with a C-terminal cysteine [48]. This gold-nanoscale LL-37 delivery system, with a peptide concentration of about 2.34 µg/mL, increased the phosphorylation of EGFR and extracellular signal-regulated protein kinase 1/2 (ERK1/2), and it promoted the migratory properties of keratinocytes in vitro [48]. They also observed higher wound-healing activity and higher expression of collagen, interleukin 6 (IL6), and vascular endothelial growth factor (VEGF) after intradermal administration of LL-37-Au NPs at several sites around the wound, when compared to free LL-37 or empty Au NPs in an in vivo mouse model of full-thickness excisional wounds [48]. Furthermore, Lipsky et al. have produced a cream for topical delivery of pexiganan and tested its efficacy in a randomized, controlled, double-blinded, multicenter clinical trial for treating diabetic patients with a mildly infected DFU, when compared to oral ofloxacin antibiotic [74]. These authors demonstrated similar results between pexiganan 0.8% topical cream and oral ofloxacin treatments applied twice daily for 14 days along with standard local wound care, regarding clinical improvement, overall microbiological eradication against S. aureus, E. coli, E. cloacae and S. marcescens, P. aeruginosa, Enterococcus species, MSSA and MRSA, and wound healing improvement [74]. However, bacterial resistance to ofloxacin emerged in some of the patients, but not against pexiganan, suggesting that topical pexiganan could still be an encouraging alternative to oral antibiotic therapy in treating patients with mildly infected DFU [74].
By combining chemical modifications and the use of delivery systems, Song et al. used the synthetic Cys-KR12 peptide, originated from LL-37, and immobilized it onto a silk-fibroin (SF) nanofiber membrane, with peptide concentrations of 50, 100, 200, and 500 µg/mL [75]. This Cys-KR12-immobilized SF nanofiber membrane, containing 200 or 500 µg/mL of peptide, exhibited antimicrobial activity against the following four pathogenic bacterial strains: S. aureus, S. epidermidis, E. coli and P. aeruginosa without biofilm formation [75]. Moreover, these authors have demonstrated that this system promoted proliferation of keratinocytes and fibroblasts in vitro, enhanced differentiation of keratinocytes, and repressed lipopolysaccharides (LPS)-induced tumor necrosis factor alpha (TNF-α) expression of monocytes [75]. Furthermore, Sultan et al. have developed a novel peptide-based bioadhesive hydrogel formulation [76]. They synthesized K11R-K17R peptide, a 24-amino acid peptide, derivative of Histatin-5 (Hst-5), through substitution of lysine residues at positions 11 and 17 with arginine residues, to produce a stable variant peptide that is resistant to proteolytic degradation [76]. In addition, this peptide was used with the FDA-approved hydroxypropyl methylcellulose (HPMC)-based bioadhesive hydrogel as a delivery system to evaluated its efficacy in vitro [76]. This formulation, with a peptide concentration of 2 mg/mL, was shown to have antimicrobial activity against fungal C. albicans strains, resistant to traditional antifungals, in addition to promoting cell proliferation and migration of human oral keratinocytes [76].
Moreover, Gawande et al. have formulated a wound gel combining DispersinB®, an antibiofilm enzyme, with Pluronic F-127, a gelling agent, and KSL-W, a synthetic 10-amino acid peptide, representing an analogue of KSL that is known to have antimicrobial activity [77]. This formulation, with a peptide concentration of 125 or 250 µg/mL, had in vitro antibiofilm and antimicrobial activity against chronic wound infection associated biofilm-embedded bacteria, including MRSA (250 µg/mL), S. epidermidis (250 µg/mL), Coagulase-negative Staphylococci (CoNS) (250 µg/mL), and A. baumannii (125 µg/mL) [77]. Furthermore, Riool et al. have developed an HPMC gel to deliver TC19, a synthetic 15-amino acid peptide, derived from the human thrombocidin (TC)-1-derived peptide L3 [78]. This peptide had reduced cytotoxicity in normal human dermal fibroblasts, at a peptide concentration of up to 80 µM, after 1 h and 4 h of incubation. It also had efficient and rapid antimicrobial activity against several bacterial species of the ESKAPE panel, including E. faecium (MDR), MRSA, K. pneumoniae (MDR), A. baumannii (MDR), P. aeruginosa (PDR—pandrug-resistant, meaning non-susceptibility to all agents in all antimicrobial categories) and E. cloacae (MDR) [78]. Additionally, TC19 reduced bacterial resistance in vitro, and reduced pro-inflammatory activity of bacterial cell envelope components [78]. Then, the 2% TC19-containing HPMC gel, was tested and it was shown to increase antimicrobial activity against MRSA and A. baumannii (MDR) in a murine superficial wound infection model 4 h after its topical application [78]. Finally, Lin et al. have produced an alginate/hyaluronic acid/collagen (Alg/HA/Cil) wound dressing to immobilize Tet213, a synthetic 10-amino acid peptide, consisting on the cysteinylated form of HHC36 peptide, known to have high antimicrobial activity [79]. This Tet213-loaded dressing, with a peptide concentration of 500 µg/mL, was shown to exhibit in vitro antimicrobial activity against E. coli, S. aureus and MRSA, as well as to promote proliferation of NIH 3T3 fibroblast cells [79]. Besides, Tet213-loaded Alg/HA/Col-dressing, applied one day after bacterial challenge and changed every 3 days for up to 14 days, induced wound healing, re-epithelialization, collagen deposition, and angiogenesis in an in vivo rat model of partial-thickness wounds with mixed-bacterial infection (E. coli and S. aureus) [79].
Numerous authors have been developing and applying delivery systems to enhance the properties of AMPs, which have proven to be encouraging approaches for treating non-healing infected DFUs. In fact, these systems protect AMPs from host diabetic microenvironment, protease degradation and serum inactivation, reduce their inherent toxicity and improve their targeting and prolonged delivery. Nonetheless, the use of delivery systems themselves can also interfere with wound healing, and consequently needs to be considered when assessing the efficacy of a given formulation. It is noteworthy that some delivery systems themselves also have antimicrobial properties and can moisten the wound microenvironment, which will facilitate the wound-healing process. Finally, these formulations will also prevent the emergence of bacterial or fungal resistance, therefore becoming attractive alternatives to the use of antibiotics and antifungals.

6. Conclusions and Future Perspectives

Despite a wealth of research about AMPs and their respective application as potential therapy for non-healing infected wounds, this area needs further investigation. There is evidence that the performance of chemical modifications and the use of delivery systems can greatly improve the characteristics of AMPs to be applied as alternatives to antibiotics and antifungals. AMP-based approaches could be a solution for the emergence of antimicrobial resistance or could be applied in association with antibiotics or antifungals to promote a synergistic action for treating chronic wounds. However, few have been developed to treat polymicrobial infections that include anaerobic bacteria, fungi, and biofilms, and consequently to improve the treatment of infected DFUs. Only Gomes et al., Tomioka et al., and Sultan et al. have evaluated the action of PP4-3.1, SR-0379 and K11R-K17R against fungi, respectively, without any study considering the action of AMPs against anaerobic bacteria present in the DFU microenvironment. Therefore, further studies will need to include more models of infection with anaerobic bacteria, fungi, and biofilms, since infected DFUs tend to have a multi-kingdom basis. Indeed, non-healing DFUs have been highly associated with fungal pathogens and anaerobic bacteria [11,12,13,14,17,18]. It is noteworthy that the infection models used in the different studies presented herein include microorganisms that are more pathogenic and predominant in DFUs, such as S. aureus (MSSA and MRSA), P. aeruginosa, E. coli, and A. baumannii, as well as some Candida spp. However, these infection models only include one or two of these microbes, and do not consider the complexity of polymicrobial infections and biofilms in human-infected chronic wounds. Furthermore, more accurate models of infected DFUs need to be included in future research to prove the efficacy of novel AMP delivery systems as therapeutic approaches for treating chronic infected wounds. Indeed, better wound models also need to be implemented to better mimic the human condition, including full-thickness infected wound models. Together, these future improvements could conduct to a greater translation into the clinical practice and consequently to a reduction of clinical trial failure rates, leading to effective management and treatment approaches for multi-kingdom infected DFUs, to enhance the health and the quality of life of these patients.

Author Contributions

Conceptualization, J.D.S., E.C.L. and E.C.; investigation (literature review), J.D.S.; writing—original draft preparation, J.D.S.; writing—review and editing, J.D.S., E.C.L., and E.C.; supervision, E.C.L. and E.C.; funding acquisition, J.D.S., E.C.L. and E.C. All authors have read and agreed to the published final version of the manuscript.

Funding

This work was financed by the European Regional Development Fund (ERDF) through the Centro 2020 Regional Operational Programme under project CENTRO-01-0145-FEDER-000012 (HealthyAging2020) and through the COMPETE 2020—Operational Programme for Competitiveness and Internationalisation and Portuguese national funds via FCT—Fundação para a Ciência e a Tecnologia, under projects POCI-01-0145-FEDER-007440, DL57/2016/CP1448/CT0024 (E.C.L.), PhD Scholarship 2020.04990.BD (J.D.S.), UIDB/04539/2020 and UIDP/04539/2020.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The Figure was created in the web application BioRender.com (Available online: Biorender.com, accessed on 9 November 2021).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. International Diabetes Federation. IDF Diabetes Atlas, 9th ed.; International Diabetes Federation: Brussels, Belgium, 2019; ISBN 978-2-930229-87-4. [Google Scholar]
  2. Vibha, S.P.; Kulkarni, M.M.; Kirthinath Ballala, A.B.; Kamath, A.; Maiya, G.A. Community based study to assess the prevalence of diabetic foot syndrome and associated risk factors among people with diabetes mellitus. BMC Endocr. Disord. 2018, 18, 43. [Google Scholar] [CrossRef] [PubMed]
  3. Saeedi, P.; Petersohn, I.; Salpea, P.; Malanda, B.; Karuranga, S.; Unwin, N.; Colagiuri, S.; Guariguata, L.; Motala, A.A.; Ogurtsova, K.; 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, 157, 107843. [Google Scholar] [CrossRef] [Green Version]
  4. Brocco, E.; Ninkovic, S.; Marin, M.; Whisstock, C.; Bruseghin, M.; Boschetti, G.; Viti, R.; Forlini, W.; Volpe, A. Diabetic foot management: Multidisciplinary approach for advanced lesion rescue. J. Cardiovasc. Surg. 2018, 59, 670–684. [Google Scholar] [CrossRef]
  5. Singer, A.J.; Clark, R.A.F. Cutaneous Wound Healing. N. Engl. J. Med. 1999, 341, 738–746. [Google Scholar] [CrossRef] [PubMed]
  6. Reardon, R.; Simring, D.; Kim, B.; Mortensen, J.; Williams, D.; Leslie, A. The diabetic foot ulcer. Aust. J. Gen. Pract. 2020, 49, 250–255. [Google Scholar] [CrossRef]
  7. Apelqvist, J. Diagnostics and treatment of the diabetic foot. Endocrine 2012, 41, 384–397. [Google Scholar] [CrossRef] [PubMed]
  8. Uivaraseanu, B.; Bungau, S.; Tit, D.M.; Fratila, O.; Rus, M.; Maghiar, T.A.; Maghiar, O.; Pantis, C.; Vesa, C.M.; Zaha, D.C. Clinical, Pathological and Microbiological Evaluation of Diabetic Foot Syndrome. Medicina 2020, 56, 380. [Google Scholar] [CrossRef]
  9. Ramirez-Acuña, J.M.; Cardenas-Cadena, S.A.; Marquez-Salas, P.A.; Garza-Veloz, I.; Perez-Favila, A.; Cid-Baez, M.A.; Flores-Morales, V.; Martinez-Fierro, M.L. Diabetic foot ulcers: Current advances in antimicrobial therapies and emerging treatments. Antibiotics 2019, 8, 193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Kalan, L.R.; Meisel, J.S.; Loesche, M.A.; Horwinski, J.; Soaita, I.; Chen, X.; Uberoi, A.; Gardner, S.E.; Grice, E.A. Strain- and Species-Level Variation in the Microbiome of Diabetic Wounds Is Associated with Clinical Outcomes and Therapeutic Efficacy. Cell Host Microbe 2019, 25, 641–655. [Google Scholar] [CrossRef]
  11. Kareliya, H.; Bichile, L.; Bal, A.; Varaiya, A.; Bhalekar, P. Fungal Infection in Diabetic Foot a Clinicomicrobiological Study. Acta Sci. Mcrobiology 2019, 2, 49–55. [Google Scholar] [CrossRef]
  12. Kalshetti, V.T.; Wadile, R.; Bothikar, S.; Ambade, V.; Bhate, V. Study of fungal infections in diabetic foot Ulcer. Indian J Microbiol Res 2017, 4, 87–89. [Google Scholar] [CrossRef]
  13. Raiesi, O.; Shabandoust, H.; Dehghan, P.; Shamsaei, S.; Soleimani, A. Fungal infection in foot diabetic patients. J. Basic Res. Med. Sci. 2018, 5, 47–51. [Google Scholar] [CrossRef] [Green Version]
  14. Chellan, G.; Shivaprakash, S.; Ramaiyar, S.K.; Varma, A.K.; Varma, N.; Sukumaran, M.T.; Vasukutty, J.R.; Bal, A.; Kumar, H. Spectrum and prevalence of fungi infecting deep tissues of lower-limb wounds in patients with type 2 diabetes. J. Clin. Microbiol. 2010, 48, 2097–2102. [Google Scholar] [CrossRef] [Green Version]
  15. Barwell, N.D.; Devers, M.C.; Kennon, B.; Hopkinson, H.E.; McDougall, C.; Young, M.J.; Robertson, H.M.A.; Stang, D.; Dancer, S.J.; Seaton, A.; et al. Diabetic foot infection: Antibiotic therapy and good practice recommendations. Int. J. Clin. Pract. 2017, 71, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Hobizal, K.B.; Wukich, D.K. Diabetic foot infections: Current concept review. Diabet. Foot Ankle 2012, 3, 18409. [Google Scholar] [CrossRef]
  17. Percival, S.L.; Malone, M.; Mayer, D.; Salisbury, A.M.; Schultz, G. Role of anaerobes in polymicrobial communities and biofilms complicating diabetic foot ulcers. Int. Wound J. 2018, 15, 776–782. [Google Scholar] [CrossRef]
  18. MacDonald, A.; Brodell, J.D.; Daiss, J.L.; Schwarz, E.M.; Oh, I. Evidence of differential microbiomes in healing versus non-healing diabetic foot ulcers prior to and following foot salvage therapy. J. Orthop. Res. 2019, 37, 1596–1603. [Google Scholar] [CrossRef]
  19. Kalan, L.; Loesche, M.; Hodkinson, B.P.; Heilmann, K.; Ruthel, G.; Gardner, S.E.; Grice, E.A. Redefining the chronic-wound microbiome: Fungal communities are prevalent, dynamic, and associated with delayed healing. MBio 2016, 7, e01058-16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Kalan, L.; Grice, E.A. Fungi in the wound microbiome. Adv. Wound Care 2018, 7, 247–255. [Google Scholar] [CrossRef] [PubMed]
  21. Korting, H.C.; Schlölmann, C.; Stauss-Grabo, M.; Schäfer-Korting, M. Antimicrobial peptides and skin: A paradigm of translational medicine. Skin Pharmacol. Physiol. 2012, 25, 323–334. [Google Scholar] [CrossRef] [Green Version]
  22. da Costa, J.P.; Cova, M.; Ferreira, R.; Vitorino, R. Antimicrobial peptides: An alternative for innovative medicines? Appl. Microbiol. Biotechnol. 2015, 99, 2023–2040. [Google Scholar] [CrossRef]
  23. Gonzalez-Curiel, I.; Trujillo, V.; Montoya-Rosales, A.; Rincon, K.; Rivas-Calderon, B.; De Haro-Acosta, J.; Marin-Luevano, P.; Lozano-Lopez, D.; Enciso-Moreno, J.A.; Rivas-Santiago, B. 1,25-dihydroxyvitamin D3 induces LL-37 and HBD-2 production in keratinocytes from diabetic foot ulcers promoting wound healing: An in vitro model. PLoS ONE 2014, 9, e111355. [Google Scholar] [CrossRef]
  24. Meng, L.; Guo, X.; Yang, X.; Liu, H.; Yu, M.; Wu, Y.; Zhu, Z. Human α defensins promote the expression of the inflammatory cytokine interleukin-8 under high-glucose conditions: Novel insights into the poor healing of diabetic foot ulcers. J. Biochem. Mol. Toxicol. 2019, 33, e22351. [Google Scholar] [CrossRef]
  25. Santos, R.; Gomes, D.; Macedo, H.; Barros, D.; Tibério, C.; Veiga, A.S.; Tavares, L.; Castanho, M.; Oliveira, M. Guar gum as a new antimicrobial peptide delivery system against diabetic foot ulcers Staphylococcus aureus isolates. J. Med. Microbiol. 2016, 65, 1092–1099. [Google Scholar] [CrossRef] [PubMed]
  26. Mi, B.; Liu, J.; Liu, Y.; Hu, L.; Liu, Y.; Panayi, A.C.; Zhou, W.; Liu, G. The designer antimicrobial peptide A-hBD-2 facilitates skin wound healing by stimulating keratinocyte migration and proliferation. Cell. Physiol. Biochem. 2018, 51, 647–663. [Google Scholar] [CrossRef] [PubMed]
  27. Rodrigues, M.; Kosaric, N.; Bonham, C.A.; Gurtner, G.C. Wound healing: A cellular perspective. Physiol. Rev. 2019, 99, 665–706. [Google Scholar] [CrossRef] [PubMed]
  28. Childs, D.R.; Murthy, A.S. Overview of Wound Healing and Management. Surg. Clin. North Am. 2017, 97, 189–207. [Google Scholar] [CrossRef] [PubMed]
  29. Petkovic, M.; Sørensen, A.E.; Leal, E.C.; Carvalho, E.; Dalgaard, L.T. Mechanistic Actions of microRNAs in Diabetic Wound Healing. Cells 2020, 9, 2228. [Google Scholar] [CrossRef]
  30. Petkovic, M.; Vangmouritzen, M.; Mojsoska, B.; Jenssen, H. Immunomodulatory properties of host defence peptides in skin wound healing. Biomolecules 2021, 11, 952. [Google Scholar] [CrossRef]
  31. Hurlow, J.J.; Humphreys, G.J.; Bowling, F.L.; McBain, A.J. Diabetic foot infection: A critical complication. Int. Wound J. 2018, 15, 814–821. [Google Scholar] [CrossRef]
  32. Pouget, C.; Dunyach-Remy, C.; Pantel, A.; Schuldiner, S.; Sotto, A.; Lavigne, J.P. Biofilms in diabetic foot ulcers: Significance and clinical relevance. Microorganisms 2020, 8, 1580. [Google Scholar] [CrossRef]
  33. Han, S.H.; Lee, J.S.; Song, K.H.; Choe, Y.B.; Ahn, K.J.; Lee, Y.W. Differences in foot skin microbiomes between patients with type 2 diabetes and healthy individuals. Mycoses 2020, 63, 314–322. [Google Scholar] [CrossRef] [PubMed]
  34. Pereira, S.G.; Moura, J.; Carvalho, E.; Empadinhas, N. Microbiota of chronic diabetic wounds: Ecology, impact, and potential for innovative treatment strategies. Front. Microbiol. 2017, 8, 1791. [Google Scholar] [CrossRef]
  35. Mottola, C.; Mendes, J.J.; Cristino, J.M.; Cavaco-Silva, P.; Tavares, L.; Oliveira, M. Polymicrobial biofilms by diabetic foot clinical isolates. Folia Microbiol. 2016, 61, 35–43. [Google Scholar] [CrossRef] [PubMed]
  36. MacLeod, A.S. Bad “Staph” in the Wound Environment of Diabetic Foot Ulcers. Cell Host Microbe 2019, 25, 638–640. [Google Scholar] [CrossRef] [PubMed]
  37. Dörr, S.; Freier, F.; Schlecht, M.; Lobmann, R. Bacterial diversity and inflammatory response at first-time visit in younger and older individuals with diabetic foot infection (DFI). Acta Diabetol. 2021, 58, 181–189. [Google Scholar] [CrossRef]
  38. Murali, T.S.; Kavitha, S.; Spoorthi, J.; Bhat, D.V.; Prasad, A.S.B.; Upton, Z.; Ramachandra, L.; Acharya, R.V.; Satyamoorthy, K. Characteristics of microbial drug resistance and its correlates in chronic diabetic foot ulcer infections. J. Med. Microbiol. 2014, 63, 1377–1385. [Google Scholar] [CrossRef]
  39. Dowd, S.E.; Delton Hanson, J.; Rees, E.; Wolcott, R.D.; Zischau, A.M.; Sun, Y.; White, J.; Smith, D.M.; Kennedy, J.; Jones, C.E. Survey of fungi and yeast in polymicrobial infections in chronic wounds. J. Wound Care 2011, 20, 40–47. [Google Scholar] [CrossRef]
  40. Mehra, B.K.; Singh, A.K.; Gupta, D.; Narang, R.; Patil, R. A Clinicomicrobiological Study on Incidence of Mycotic Infections in Diabetic Foot Ulcers. Int. J. Sci. Study 2017, 50, 12. [Google Scholar] [CrossRef]
  41. Thapa, R.K.; Diep, D.B.; Tønnesen, H.H. Topical antimicrobial peptide formulations for wound healing: Current developments and future prospects. Acta Biomater. 2020, 103, 52–67. [Google Scholar] [CrossRef]
  42. Ong, P.Y.; Ohtake, T.; Brandt, C.; Strickland, I.; Boguniewicz, M.; Ganz, T.; Gallo, R.L.; Leung, D.Y.M. Endogenous Antimicrobial Peptides and Skin Infections in Atopic Dermatitis. N. Engl. J. Med. 2002, 347, 1151–1160. [Google Scholar] [CrossRef] [Green Version]
  43. Pfalzgraff, A.; Brandenburg, K.; Weindl, G. Antimicrobial peptides and their therapeutic potential for bacterial skin infections and wounds. Front. Pharmacol. 2018, 9, 281. [Google Scholar] [CrossRef]
  44. Vatan, A.; Saltoglu, N.; Yemisen, M.; Balkan, I.I.; Surme, S.; Demiray, T.; Mete, B.; Tabak, F. Association between biofilm and multi/extensive drug resistance in diabetic foot infection. Int. J. Clin. Pract. 2018, 72, e13060. [Google Scholar] [CrossRef]
  45. Duplantier, A.J.; van Hoek, M.L. The human cathelicidin antimicrobial peptide LL-37 as a potential treatment for polymicrobial infected wounds. Front. Immunol. 2013, 4, 143. [Google Scholar] [CrossRef] [Green Version]
  46. Gomes, A.; Teixeira, C.; Ferraz, R.; Prudencio, C.; Gomes, P. Wound-healing peptides for treatment of chronic diabetic foot ulcers and other infected skin injuries. Molecules 2017, 22, 1743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Tomioka, H.; Nakagami, H.; Tenma, A.; Saito, Y.; Kaga, T.; Kanamori, T.; Tamura, N.; Tomono, K.; Kaneda, Y.; Morishita, R. Novel anti-microbial peptide SR-0379 accelerates wound healing via the PI3 kinase/Akt/mTOR pathway. PLoS ONE 2014, 9, e92597. [Google Scholar] [CrossRef]
  48. Comune, M.; Rai, A.; Chereddy, K.K.; Pinto, S.; Aday, S.; Ferreira, A.F.; Zonari, A.; Blersch, J.; Cunha, R.; Rodrigues, R.; et al. Antimicrobial peptide-gold nanoscale therapeutic formulation with high skin regenerative potential. J. Control. Release 2017, 262, 58–71. [Google Scholar] [CrossRef]
  49. Pazgier, M.; Hoover, D.M.; Yang, D.; Lu, W.; Lubkowski, J. Human β-defensins. Cell. Mol. Life Sci. 2006, 63, 1294–1313. [Google Scholar] [CrossRef]
  50. Rivas-Santiago, B.; Trujillo, V.; Montoya, A.; Gonzalez-Curiel, I.; Castañeda-Delgado, J.; Cardenas, A.; Rincon, K.; Hernandez, M.L.; Hernández-Pando, R. Expression of antimicrobial peptides in diabetic foot ulcer. J. Dermatol. Sci. 2012, 65, 19–26. [Google Scholar] [CrossRef]
  51. Torrent, M.; Andreu, D.; Nogués, V.M.; Boix, E. Connecting peptide physicochemical and antimicrobial properties by a rational prediction model. PLoS ONE 2011, 6, e16968. [Google Scholar] [CrossRef]
  52. NIHR Horizon Scanning Research and Intelligence Centre. Pexiganan (Locilex) for Mild Foot Infection in Diabetic Foot Ulcers, University of Birmingham: Birmingham, UK, 2015.
  53. Zhang, M.; Zhao, J.; Zheng, J. Molecular understanding of a potential functional link between antimicrobial and amyloid peptides. Soft Matter 2014, 10, 7425–7451. [Google Scholar] [CrossRef]
  54. Herman, A.; Herman, A.P. Antimicrobial peptides activity in the skin. Ski. Res. Technol. 2019, 25, 111–117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Grönberg, A.; Mahlapuu, M.; Ståhle, M.; Whately-Smith, C.; Rollman, O. Treatment with LL-37 is safe and effective in enhancing healing of hard-to-heal venous leg ulcers: A randomized, placebo-controlled clinical trial. Wound Repair Regen. 2014, 22, 613–621. [Google Scholar] [CrossRef]
  56. Grek, C.L.; Prasad, G.M.; Viswanathan, V.; Armstrong, D.G.; Gourdie, R.G.; Ghatnekar, G.S. Topical administration of a connexin43-based peptide augments healing of chronic neuropathic diabetic foot ulcers: A multicenter, randomized trial. Wound Repair Regen. 2015, 23, 203–212. [Google Scholar] [CrossRef] [PubMed]
  57. Marin-Luevano, P.; Trujillo, V.; Rodriguez-Carlos, A.; González-Curiel, I.; Enciso-Moreno, J.A.; Hancock, R.E.W.; Rivas-Santiago, B. Induction by innate defence regulator peptide 1018 of pro-angiogenic molecules and endothelial cell migration in a high glucose environment. Peptides 2018, 101, 135–144. [Google Scholar] [CrossRef]
  58. Flamm, R.K.; Rhomberg, P.R.; Simpson, K.M.; Farrell, D.J.; Sader, H.S.; Jones, R.N. In vitro spectrum of pexiganan activity when tested against pathogens from diabetic foot infections and with selected resistance mechanisms. Antimicrob. Agents Chemother. 2015, 59, 1751–1754. [Google Scholar] [CrossRef] [Green Version]
  59. Lan, C.C.E.; Wu, C.S.; Huang, S.M.; Kuo, H.Y.; Wu, I.H.; Liang, C.W.; Chen, G.S. High-glucose environment reduces human β-defensin-2 expression in human keratinocytes: Implications for poor diabetic wound healing. Br. J. Dermatol. 2012, 166, 1221–1229. [Google Scholar] [CrossRef] [PubMed]
  60. Galkowska, H.; Olszewski, W.L.; Wojewodzka, U. Expression of natural antimicrobial peptide β-defensin-2 and Langerhans cell accumulation in epidermis from human non-healing leg ulcers. Folia Histochem. Cytobiol. 2005, 43, 133–136. [Google Scholar] [CrossRef]
  61. Liu, H.; Duan, Z.; Tang, J.; Lv, Q.; Rong, M.; Lai, R. A short peptide from frog skin accelerates diabetic wound healing. FEBS J. 2014, 281, 4633–4643. [Google Scholar] [CrossRef] [Green Version]
  62. Steinstraesser, L.; Hirsch, T.; Schulte, M.; Kueckelhaus, M.; Jacobsen, F.; Mersch, E.A.; Stricker, I.; Afacan, N.; Jenssen, H.; Hancock, R.E.W.; et al. Innate defense regulator peptide 1018 in wound healing and wound infection. PLoS ONE 2012, 7, e39373. [Google Scholar] [CrossRef] [Green Version]
  63. Magana, M.; Pushpanathan, M.; Santos, A.L.; Leanse, L.; Fernandez, M.; Ioannidis, A.; Giulianotti, M.A.; Apidianakis, Y.; Bradfute, S.; Ferguson, A.L.; et al. The value of antimicrobial peptides in the age of resistance. Lancet Infect. Dis. 2020, 20, e216–e230. [Google Scholar] [CrossRef]
  64. Gomes, A.; Ferraz, R.; Teixeira, C. The Emerging Role of Ionic Liquid-Based Approaches for Enhanced Skin Permeation of Bioactive Molecules: A Snapshot of the Past Couple of Years. Int. J. Mol. Sci. 2021, 22, 11991. [Google Scholar] [CrossRef]
  65. Li, W.; Separovic, F.; O’Brien-Simpson, N.M.; Wade, J.D. Chemically modified and conjugated antimicrobial peptides against superbugs. Chem. Soc. Rev. 2021, 50, 4932–4973. [Google Scholar] [CrossRef] [PubMed]
  66. Gomes, A.; Bessa, L.J.; Fernandes, I.; Ferraz, R.; Mateus, N.; Gameiro, P.; Teixeira, C.; Gomes, P. Turning a collagenesis-inducing peptide into a potent antibacterial and antibiofilm agent against multidrug-resistant gram-negative bacteria. Front. Microbiol. 2019, 10, 1915. [Google Scholar] [CrossRef] [Green Version]
  67. Gomes, A.; Bessa, L.J.; Fernandes, I.; Ferraz, R.; Martins, M.C.L.; Mateus, N.; Gameiro, P.; Teixeira, C.; Gomes, P. Disclosure of a Promising Lead to Tackle Complicated Skin and Skin Structure Infections: Antimicrobial and Antibiofilm Actions of Peptide PP4-3. Pharmaceutics 2021, 13, 1962. [Google Scholar] [CrossRef]
  68. Mouritzen, M.V.; Petkovic, M.; Qvist, K.; Poulsen, S.S.; Alarico, S.; Leal, E.C.; Dalgaard, L.T.; Empadinhas, N.; Carvalho, E.; Jenssen, H. Improved diabetic wound healing by LFcinB is associated with relevant changes in the skin immune response and microbiota. Mol. Ther. Methods Clin. Dev. 2021, 20, 726–739. [Google Scholar] [CrossRef]
  69. Jung Kim, D.; Lee, Y.W.; Park, M.K.; Shin, J.R.; Lim, K.J.; Cho, J.H.; Kim, S.C. Efficacy of the designer antimicrobial peptide SHAP1 in wound healing and wound infection. Amino Acids 2014, 46, 2333–2343. [Google Scholar] [CrossRef] [PubMed]
  70. Bolatchiev, A.; Baturin, V.; Bazikov, I.; Maltsev, A.; Kunitsina, E. Effect of antimicrobial peptides HNP-1 and hBD-1 on Staphylococcus aureus strains in vitro and in vivo. Fundam. Clin. Pharmacol. 2020, 34, 102–108. [Google Scholar] [CrossRef] [PubMed]
  71. Santos, R.; Ruza, D.; Cunha, E.; Tavares, L.; Oliveira, M. Diabetic foot infections: Application of a nisin-biogel to complement the activity of conventional antibiotics and antiseptics against Staphylococcus aureus biofilms. PLoS ONE 2019, 14, e0220000. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Bayramov, D.; Li, Z.; Patel, E.; Izadjoo, M.; Kim, H.; Neff, J. A Novel Peptide-Based Antimicrobial Wound Treatment is Effective Against Biofilms of Multi-Drug Resistant Wound Pathogens. Mil. Med. 2018, 183, 481–486. [Google Scholar] [CrossRef] [Green Version]
  73. Zhao, Y.; Du, X.; Jiang, L.; Luo, H.; Wang, F.; Wang, J.; Qiu, L.; Liu, L.; Liu, X.; Wang, X.; et al. Glucose Oxidase-Loaded Antimicrobial Peptide Hydrogels: Potential Dressings for Diabetic Wound. J. Nanosci. Nanotechnol. 2019, 20, 2087–2094. [Google Scholar] [CrossRef]
  74. Lipsky, B.A.; Holroyd, K.J.; Zasloff, M. Topical versus systemic antimicrobial therapy for treating mildly infected diabetic foot ulcers: A randomized, controlled, double-blinded, multicenter trial of pexiganan cream. Clin. Infect. Dis. 2008, 47, 1537–1545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Song, D.W.; Kim, S.H.; Kim, H.H.; Lee, K.H.; Ki, C.S.; Park, Y.H. Multi-biofunction of antimicrobial peptide-immobilized silk fibroin nanofiber membrane: Implications for wound healing. Acta Biomater. 2016, 39, 146–155. [Google Scholar] [CrossRef] [PubMed]
  76. Sultan, A.S.; Vila, T.; Hefni, E.; Karlsson, A.J.; Jabra-Rizk, M.A. Evaluation of the antifungal and wound-healing properties of a novel peptide-based bioadhesive hydrogel formulation. Antimicrob. Agents Chemother. 2019, 63, e00888-19. [Google Scholar] [CrossRef] [PubMed]
  77. Gawande, P.V.; Leung, K.P.; Madhyastha, S. Antibiofilm and Antimicrobial Efficacy of DispersinB®-KSL-W Peptide-Based Wound Gel Against Chronic Wound Infection Associated Bacteria. Curr. Microbiol. 2014, 68, 635–641. [Google Scholar] [CrossRef]
  78. Riool, M.; de Breij, A.; Kwakman, P.H.S.; Schonkeren-Ravensbergen, E.; de Boer, L.; Cordfunke, R.A.; Malanovic, N.; Drijfhout, J.W.; Nibbering, P.H.; Zaat, S.A.J. Thrombocidin-1-derived antimicrobial peptide TC19 combats superficial multi-drug resistant bacterial wound infections. Biochim. Biophys. Acta Biomembr. 2020, 1862, 183282. [Google Scholar] [CrossRef]
  79. Lin, Z.; Wu, T.; Wang, W.; Li, B.; Wang, M.; Chen, L.; Xia, H.; Zhang, T. Biofunctions of antimicrobial peptide-conjugated alginate/hyaluronic acid/collagen wound dressings promote wound healing of a mixed-bacteria-infected wound. Int. J. Biol. Macromol. 2019, 140, 330–342. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Mechanisms of action of AMPs supporting their therapeutic application for treating infected non-healing wounds—unraveled antimicrobial and wound-healing properties: (a) direct killing of microbes; (b) modulation of the host’s immune system; (c) promotion of cell migration and proliferation; (d) induction of angiogenesis; and (e) enhancement of extracellular matrix synthesis. Figure created in BioRender.com (accessed on 9 November 2021).
Figure 1. Mechanisms of action of AMPs supporting their therapeutic application for treating infected non-healing wounds—unraveled antimicrobial and wound-healing properties: (a) direct killing of microbes; (b) modulation of the host’s immune system; (c) promotion of cell migration and proliferation; (d) induction of angiogenesis; and (e) enhancement of extracellular matrix synthesis. Figure created in BioRender.com (accessed on 9 November 2021).
Biomolecules 11 01894 g001
Table 1. Most predominantly identified microorganisms in DFUs, comprising both Gram-positive and Gram-negative bacteria, as well as anaerobic bacteria and fungi. All microorganisms are presented in order of the greatest abundance in DFUs.
Table 1. Most predominantly identified microorganisms in DFUs, comprising both Gram-positive and Gram-negative bacteria, as well as anaerobic bacteria and fungi. All microorganisms are presented in order of the greatest abundance in DFUs.
Gram-PositiveBACTERIA
Gram-Negative
Anaerobes
S. aureus (MSSA and MRSA) [8,9,10,14,15,17,32,34,35,36,37,38]
C. striatum [10,32,34,43]
Streptococcus β-hemolytic [8,9,17,18,32]
P. aeruginosa [8,9,10,14,17,35,36,37,38,44]
Proteus spp. [8,9,17,37]
Enterobacter spp. [8,17,37]
Citrobacter spp. [8,17,42]
E. coli [8,17,37]
A. baumannii [8,17,38,43]
Bacteroides spp. [9,17,18]
Prevotella spp. [9,10,17]
Peptostreptococcus spp. [9]
Clostridium spp. [9]
FUNGI
C. albicans [11,12,13,14,19,20,39,40]C. tropicalis [12,14,20,39,40]C. glabrata [12,39,40]
C. parapsilosis [14,19,20,39,40]T. rubrum [12,13,44]T. mentagrophytes [12,13,40]
A. fumigatus [12,14,20]T. asahii [14,19,20]C. herbarum [19,20,40]
Table 2. Main endogenous AMP primary, secondary, and tertiary structures, and their related physicochemical properties, including length, molecular weight (MW), isoelectric point (pI), net charge, and hydrophobicity. PBD codes were obtained from the Protein Data Bank: www.rcsb.org (accessed on 1 December 2021). The physicochemical properties were obtained from www.pepdraw.com (accessed on 5 December 2021) and confirmed in other similar software, whereas the secondary and tertiary structures were obtained from www.compbio.dundee.ac.uk/jpred4/index.html (accessed on 7 December 2021) and www.rcsb.org/structure/ (accessed on 1 December 2021), respectively.
Table 2. Main endogenous AMP primary, secondary, and tertiary structures, and their related physicochemical properties, including length, molecular weight (MW), isoelectric point (pI), net charge, and hydrophobicity. PBD codes were obtained from the Protein Data Bank: www.rcsb.org (accessed on 1 December 2021). The physicochemical properties were obtained from www.pepdraw.com (accessed on 5 December 2021) and confirmed in other similar software, whereas the secondary and tertiary structures were obtained from www.compbio.dundee.ac.uk/jpred4/index.html (accessed on 7 December 2021) and www.rcsb.org/structure/ (accessed on 1 December 2021), respectively.
AMPPrimary
Structure
Length
(aa)
PDB
Code
Secondary StructureTertiary
Structure
MW
(Da)
pINet
Charge
Hydrophobicity (kcal/mol)
hBD-1DHYNCVSSGGQCLYSACPIFTKIQGTCYRGKAKCCK361IJUα-helix + β-strandthree antiparallel β-sheets stabilized by three disulfide bridges and flanked by an α-helix segment, together stabilized by a disulfide bridge3931.778.55+4+28.98
hBD-2GIGDPVTCLKSGAICHPVFCPRRYKQIGTCGLPGTKCCKKP411FD44331.179.26+6+32.25
hBD-3GIINTLQKYYCRVRGGRCAVLSCLPKEEQIGKCSTRGRKCCRRKK45Not found5157.7010.47+11+45.26
LL-37LLGDFFRKSKEKIGKEFKRIVQRIKDFLRNLVPRTES372K6Oα-helixone α-helical conformation4490.5711.15+6+41.03
Table 3. Free AMPs being applied as promising therapies for infected chronic wounds and their respective roles in antimicrobial and wound-healing activities. All AMPs are presented according to the following criteria: (1) free vs. chemically modified; (2) endogenous vs. synthetic; and (3) alphabetic order. AMP sequences are presented using the one-letter amino acid code, as per the IUPAC-IUBMB Joint Commission on Biochemical Nomenclature rules. ↑—increase; ↓—decrease. 1 AMPs that were tested against fungi.
Table 3. Free AMPs being applied as promising therapies for infected chronic wounds and their respective roles in antimicrobial and wound-healing activities. All AMPs are presented according to the following criteria: (1) free vs. chemically modified; (2) endogenous vs. synthetic; and (3) alphabetic order. AMP sequences are presented using the one-letter amino acid code, as per the IUPAC-IUBMB Joint Commission on Biochemical Nomenclature rules. ↑—increase; ↓—decrease. 1 AMPs that were tested against fungi.
AMPSequenceSourceDelivery MethodRole in Antimicrobial and
Wound-Healing Activities
Reference
hBD-2


LL-37
GIGDPVTCLKSGAICHPV
FCPRRYKQIGTCGLPGTK
CCKKP
LLGDFFRKSKEKIGKEFK
RIVQRIKDFLRNLVPRTES
Endogenous

(human)
Endogenous
(human)
Free


Free
↑ antimicrobial activity (E. coli)
↑ keratinocyte migration
[23]
CW49APFRMGICTTNSynthetic
(frog skin)
Free↑ angiogenic ability
↑ anti-inflammatory effect
little effect on re-epithelialization
[61]
IDR-1018VRLIVAVRIWRR-NH2SyntheticFree↓ in vitro toxicity compared to LL-37
↑ wound healing in S. aureus infected porcine and non-diabetic but not in diabetic murine wounds
[62]
IDR-1018VRLIVAVRIWRR-NH2SyntheticFree↑ angiogenic ability
↑ anti-inflammatory effect
↑ migration of endothelial cells
[57]
PexigananGIGKFLKKAKKFGKAFV
KILKK
Synthetic
(analogue of magainin II—frog skin)
Free↑ antimicrobial activity (E. coli, E. cloacae, Citrobacter spp., P. vulgaris, M. morganii, K. pneumoniae, S. marcescens, P. aeruginosa, A. baumannii, S. agalactiae, S. pyogenes, E. faecium, MSSA and MRSA)[58]
3.1-PP4KKLLKWLLKLLKTTKSSyntheticFree
(chemically modified)
↓ toxicity to HFF-1 human fibroblasts
↑ antimicrobial activity (E. coli, P. aeruginosa, and K. pneumoniae, including MDR isolates)
↓ formation of K. pneumoniae biofilms
[66]
PP4-3.11KTTKSKKLLKWLLKLLSyntheticFree
(chemically modified)
↑ antimicrobial activity (Gram-positive and Gram-negative bacteria, including MDR isolates, as well as against relevant Candida spp.)[67]
A-hBD-2APKAMVTCLKSGAICHP
VFCPRRYKQIGTCGLPGT
KCCKKP
SyntheticFree
(chemically modified)
↑ structural stability
↓ toxicity to keratinocytes
↑ antimicrobial activity (S. aureus)
↑ migration and proliferation of keratinocytes
↓ terminal differentiation of keratinocytes
↑ mobilization of intracellular Ca2+
↑ wound healing in vivo
[26]
LFcinBFKCRRWQWRMKKLGAP
SITCVRRAF
Synthetic
(derived from bLF)
Free
(chemically modified)
↑ keratinocyte migration in vitro and ex vivo
↑ wound healing
↑ antimicrobial activity (B. pumilus and S. aureus)
↑ angiogenesis and collagen deposition
↓ inflammation
[68]
SHAP1APKAMKLLKKLLKLQK
KGI
SyntheticFree
(chemically modified)
↓ toxicity to human erythrocytes and keratinocytes
↑ stability to proteases exposure
↑ wound closure compared to LL- 37 in vitro
↑ healing in vivo full-thickness excisional wounds
↑ antimicrobial activity (S. aureus)
↑ healing in S. aureus-infected murine wounds
[69]
SR-03791MLKLIFLHRLKRMRKRL
DLysRK
SyntheticFree
(chemically modified)
↑ proliferation of human dermal fibroblasts
↑ antimicrobial activity (bacteria, including drug-resistant, and also fungi, namely: E. coli, P. aeruginosa, S. aureus, C. krusei, T. mentagrophytes, T. rubrum, MRSA and A. baumannii (MDR))
↑ accelerated wound healing in two different wound-healing rat models
[47]
Table 4. AMPs loaded on delivery systems being applied as promising therapies for infected chronic wounds and their respective roles in antimicrobial and wound-healing activities. All AMPs are presented according to the following criteria: (1) loaded on a delivery system vs. chemically modified and loaded on a delivery system; (2) endogenous vs. synthetic; and (3) alphabetic order. AMP sequences are presented using the one-letter amino acid code, as per the IUPAC-IUBMB Joint Commission on Biochemical Nomenclature rules. ↑—increase; ↓—decrease. 1 AMPs that were tested against fungi; 2 AMPs that were/are under clinical trials.
Table 4. AMPs loaded on delivery systems being applied as promising therapies for infected chronic wounds and their respective roles in antimicrobial and wound-healing activities. All AMPs are presented according to the following criteria: (1) loaded on a delivery system vs. chemically modified and loaded on a delivery system; (2) endogenous vs. synthetic; and (3) alphabetic order. AMP sequences are presented using the one-letter amino acid code, as per the IUPAC-IUBMB Joint Commission on Biochemical Nomenclature rules. ↑—increase; ↓—decrease. 1 AMPs that were tested against fungi; 2 AMPs that were/are under clinical trials.
AMPSequenceSourceDelivery MethodRole in Antimicrobial and
Wound-Healing Activities
Reference
hBD-1



HNP-1
GNFLTGLGHRSDHYNCV
SSGGQCLYSACPIFTKIQ
GTCYRGKAKCCK

EPLQARADEVAAAPEQIAADIPEVVVSLAWDESL
APKHPGSRKNMACYCRI
PACIAGERRYGTCIYQGR
LWAFCC
Endogenous (human)


Endogenous (human)
Niosomal gel



Niosomal gel
↑ antimicrobial activity (MRSA-infected wound in rats and MSSA and MRSA isolated from patients with DFIs)[70]
NisinITSISLCTPGCKTGALMGCNMKTATCH(or N)CSIHVSKEndogenous
(bacteria)
Guar gum gel↑ antimicrobial activity against S. aureus DFU biofilm-producing isolates, including some multidrug-resistant clinical isolates[25]
NisinITSISLCTPGCKTGALMGCNMKTATCH(or N)CSIHVSKEndogenous
(bacteria)
Guar gum gel↑ antibacterial activity against biofilms formed by DFI S. aureus[71]
aCT1 2RQPKIWFPNRRKPWKKRPRPDDLEI-acidSynthetic
(analogue of Cx43)
Hydroxyethyl cellulose gel↓ ulcer area in DFU patients
↑ ulcer re-epithelialization in DFU patients
↓ time-to-complete-ulcer closure in DFU patients
[56]
ASP-1


ASP-2
RRWVRRVRRWVRRVVRVVRRWVRR

RWWRWWRRWWRR
SyntheticGel, Stratex or PU-based dressings↑ eradication of mono- and polymicrobial biofilms of MDR pathogens: S. aureus, A. baumannii, K. pneumoniae, P. aeruginosa, and MRSA
↑ BI compared to free ASP-1 and ASP-2
[72]
IKYLSVNIKYLSVNSyntheticGOx-loaded hydrogel↑ antimicrobial activity (S. aureus)
↓ blood glucose concentration of diabetic patients
[73]
LL-37LLGDFFRKSKEKIGKEFK
RIVQRIKDFLRNLVPRTE
SC
SyntheticGold-nanoscale formulation↑ phosphorylation of EGFR and ERK1/2
↑ migratory properties of keratinocytes
↑ wound-healing activity in vivo
↑ expression of collagen, IL6 and VEGF
[48]
Pexiganan 2GIGKFLKKAKKFGKAFV
KILKK
Synthetic
(analogue of magainin II—frog skin)
Cream=clinical outcome, microbiological eradication (S. aureus, E. coli, E. cloacae, S. marcescens, P. aeruginosa, Enterococcus spp., MSSA and MRSA), and wound healing as ofloxacin
↓ bacterial resistance in vivo
[74]
Cys-KR12CKRIVKRIKKWLRSynthetic
(originated from LL37)
SF nanofiber membrane
(chemically modified)
↑ antimicrobial activity (S. aureus, S. epidermidis, E. coli, and P. aeruginosa)
↑ proliferation of keratinocytes and fibroblasts
↑ differentiation of keratinocytes
↓ LPS-induced TNF-α expression of monocytes
[75]
K11R-K17R1DSHAKRHHGYRRKFHERHHSHRGYSynthetic (analogue of Hst-5 peptide)HPMC-based bioadhesive hydrogel
(chemically modified)
↑ antimicrobial activity (C. albicans strains resistant to traditional antifungals)
↑ cell proliferation and migration in human oral keratinocytes
[76]
KSL-WKKVVFWVKFKSynthetic
(analogue of KSL peptide)
Pluronic
F-127 gel
(chemically modified)
↑ antibiofilm and antimicrobial activity (chronic wound infection biofilm-embedded bacteria, including MRSA, S. epidermidis, CoNS, and A. baumannii)[77]
TC19LRCMCIKWWSGKHPKSynthetic
(derived from human TC-1-derived
peptide L3)
HPMC gel
(chemically modified)
↓ toxicity to human fibroblasts
↑ antimicrobial activity (ESKAPE panel in vitro, and MRSA and A. baumannii (MDR) in a murine superficial wound infection model)
↓ bacterial resistance inflammation in vitro
[78]
Tet213KRWWKWWRRCSynthetic
(cysteinylated HHC36
peptide)
Alg/HA/Col dressing
(chemically modified)
↑ antimicrobial activity (E. coli, S. aureus, MRSA)
↑ proliferation of NIH 3T3 fibroblast cells
↑ wound healing, re-epithelialization, collagen deposition, and angiogenesis in vivo rat model of partial-thickness mixed-bacterial infected wounds
[79]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Da Silva, J.; Leal, E.C.; Carvalho, E. Bioactive Antimicrobial Peptides as Therapeutic Agents for Infected Diabetic Foot Ulcers. Biomolecules 2021, 11, 1894. https://doi.org/10.3390/biom11121894

AMA Style

Da Silva J, Leal EC, Carvalho E. Bioactive Antimicrobial Peptides as Therapeutic Agents for Infected Diabetic Foot Ulcers. Biomolecules. 2021; 11(12):1894. https://doi.org/10.3390/biom11121894

Chicago/Turabian Style

Da Silva, Jessica, Ermelindo C. Leal, and Eugénia Carvalho. 2021. "Bioactive Antimicrobial Peptides as Therapeutic Agents for Infected Diabetic Foot Ulcers" Biomolecules 11, no. 12: 1894. https://doi.org/10.3390/biom11121894

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop