Next Article in Journal
Aqueous Two-Phase Systems Based on Ionic Liquids and Deep Eutectic Solvents as a Tool for the Recovery of Non-Protein Bioactive Compounds—A Review
Next Article in Special Issue
Development of a SPE-HPLC-PDA Method for the Quantification of Phthalates in Bottled Water and Their Gene Expression Modulation in a Human Intestinal Cell Model
Previous Article in Journal
The Effect of Low Positive Temperatures on the Formation of Secondary Metabolites in Rhodiola quadrifida (Pall.) Fisch. et C.A. Mey. In Vitro Cultures
Previous Article in Special Issue
Influence of the Drying Method on the Volatile Component Profile of Hypericum perforatum Herb: A HS-SPME-GC/MS Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Microbial Natural Products with Wound-Healing Properties

Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Republic of Korea
*
Author to whom correspondence should be addressed.
Processes 2023, 11(1), 30; https://doi.org/10.3390/pr11010030
Submission received: 23 November 2022 / Revised: 16 December 2022 / Accepted: 20 December 2022 / Published: 23 December 2022

Abstract

:
Wound healing continues to pose a challenge in clinical settings. Moreover, wound management must be performed properly and efficiently. Acute wound healing involves multiple cell divisions, a new extracellular matrix, and the process of formation, such as growth factors and cytokines, which are released at the site of the wound to regulate the process. Any changes that disrupt the healing process could cause tissue damage and prolong the healing process. Various factors, such as microbial infection, oxidation, and inflammation, can delay wound healing. In order to counter these problems, utilizing natural products with wound-healing effects has been reported to promote this process. Several natural products have been associated with wound healing, most of which are from medicinal plants. However, secondary microbial metabolites have not been extensively studied for their wound-healing properties. Further, investigations on the wound-healing control of natural microbial products are required due to a lack of studies. This review discussed the in vivo and in vitro research on the wound healing activities of natural microbial products, which may assist in the development of better wound treatments in the future.

1. Introduction

The skin can be broken or opened by wounds caused by chemical, thermal, or physical injuries [1,2]. The skin is an organ that covers the external surface of the body, and it protects the bones, muscles, internal organs, and ligaments below it with multiple layers of ectodermal tissues. The skin also protects against heat, injury, light, and microbes [3,4,5]. Several regulatory molecules and cells must work together in order for wound re-epithelialization to occur. Within the skin layers, embedded cellular and molecular substances initiate healing at the designated phases when the cutaneous layer is damaged [6,7].
Despite a systematic process, wound healing in the human body is one of the most convoluted biological actions because the following phases overlap: inflammation, the cellular phase (granulation), contraction of the wound area (wound contraction), collagen deposition (collagen formation), epithelialization (epithelialization), and remodeling of scars (cicatrization) (Scheme 1) [3,6]. Upon successful completion of all these events, the disruption of the anatomical and the skin’s functional state is restored [3]. There are several factors that can interfere with these healing processes, resulting in impaired wound healing. In the case of impaired healing, such as delayed acute and chronic wounds, the healing process generally has not progressed through the normal healing stages. The majority of chronic wounds are caused by ulcers associated with ischemia, venous causes or infection, and diabetes mellitus [2].
A natural product with medicinal properties, which contribute to alleviating these factors, can facilitate the wound-healing process and be developed as a future drug. Over the last few years, numerous research has investigated the wound-healing properties of natural products that contain antioxidant, anti-inflammatory, collagen promotion, and antibacterial properties [5]. Various phytochemicals, including alkaloids, tannins, flavonoids, terpenoids, phenolic, essential oils, and saponin compounds, may contribute to the medicinal effects. Natural products, including phytochemicals, play an important role in wound healing due to these properties [3,7,8]. Among natural wound-healing materials, hyaluronic acid has been used in the preparation of wound bandages due to its collagen deposition enhancement, epithelialization, and wound vascularization. At present, there are many wound dressings that contain hyaluronic acid available on the market, such as Hyalo Regen from Fidia Pharma, USA, NJ, as well as Hyalofill® and Hyalosafe® from Anika Therapeutics, Bedford, MA [9].
Recently, natural microbial products have been reported to be an important source of drug discovery [10,11] due to their diverse chemical scaffolds [12]. However, the discovery of natural microbial products possessing wound-healing properties is still not widely investigated. The objective of this review is to highlight the use of selected microbial natural products in promoting wound-healing processes.

2. Biosurfactant

The ability of biosurfactants to interact with modifying surfaces makes them surface-active compounds (SACs). In nature and biotechnology, these biomolecules serve different functions due to their physiological roles and physicochemical properties due to their amphiphilic nature and their production by different microorganisms. The benefits of SACs over synthetic surfactants include low toxicity, increased biodegradability, low critical micelle concentrations, and environmental acceptability [13,14,15].
In addition, these compounds exhibit antifungal [16], antibacterial [17], antiviral [18], and antitumor [19] properties. Moreover, their antiadhesive activity and antibiofilm properties contribute to the inhibition of adhesion and colonization by pathogenic microorganisms and biofilm removal [13,20]. In chronic wounds, biofilms contribute to chronic wounds and infection, thereby causing a delay in the healing process. The most commonly found pathogens in chronic wounds are Pseudomonas aeruginosa and Staphylococcus aureus. It is difficult to identify biofilms embedded in deeper layers, such as P. aeruginosa biofilms, with wound smear cultures. Further, antibiotic resistance within the biofilms is a critical problem in chronic wound management [21,22,23]. Due to these reasons, the scientific community and physicians consider wound treatment and biofilm prevention as major priorities in the healing process. As a result, SACs have recently emerged as promising wound-healing agents that cause minimal irritation and are highly compatible with human skin [13,20]. Moreover, these bioproducts speed up the re-epithelialization process and the deposition of collagen, resulting in faster wound healing [24,25].
Surfactin A (1, Figure 1) isolated from Bacillus subtilis was reported to promote wound healing and the inhibition effects of scars. Amid the healing process, 1 upregulated expression of hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor. Moreover, it accelerated keratinocyte migration via the mitogen-activated protein kinase and factor nuclear-κB (NF-κB) signaling pathways, followed by the regulation of pro-inflammatory cytokine secretion and macrophage phenotypic exchange. In addition, 1 could heal the wound due to its antioxidant properties, with an IC50 = 0.55 mg/mL [25,26]. It also prevented scar tissue formation by inhibiting the expression of α-smooth muscle actin (α-SMA) and transforming growth factor (TGF-β).
An in vitro study of a biosurfactant (BS) of a glycolipid nature (2, Figure 1), isolated from Bacillus licheniformis SV1, exhibited adequate cytocompatibility and increased 3T3/NIH fibroblast proliferation. An approach using a BS ointment in vivo stimulated re-epithelialization, fibroblast proliferation, and the faster deposition of collagen in skin excision wounds in rats, thereby demonstrating its potential to improve wound healing through transdermal delivery [27].
In a previous study, mice were treated with an ointment containing rhamnolipid (3, Figure 1) after creating severe wounds on their backs. The results of histopathological studies showed that 3 improved wound closure and collagenases, and it reduced inflammation by reducing the level of TNF-α without causing any negative skin reactions. There is evidence that dirhamnolipid therapy can alleviate scarring on the skin, as it has shown effects in rabbit ear hypertrophic scar models, with a depletion in α-SMA expression, type I collagen fibers, and scar elevation index scores [28].
By substituting human skin with an in vitro model of human dermal fibroblasts, a cell culture model was utilized to demonstrate the wound-healing capacity of sophorolipid (4, Figure 1), revealing that 4 affected the human skin fibroblast expression of elastase inhibition collagen I mRNA, with IC50 = 38.5 μg/mL. Additionally, an in vitro wound-healing evaluation in the human colorectal adenocarcinoma (HT-29) cell line showed a significant increase in collagenase-1 expression in HT-29 colorectal adenocarcinoma cells after treatment for 48 h with 4 [29,30].

3. Flavonoids

The flavonoid class of compounds possesses various biological functions, making them important sources of new pharmaceuticals, including those for treating skin wounds. The structure–activity relationship is one of the main factors that contributes to the pharmaceutical properties of flavonoids [31,32,33]. Their anti-inflammatory [34], antibacterial [35], antioxidant [36], and antifibrotic [37] properties depend largely on the presence of hydroxyl groups due to high hydroxylation.
It has already been demonstrated that flavonoids exhibit significant pharmacological activity, such as increasing epithelialization rates, modulating inflammatory cytokinases, reducing mononuclear cells in the proliferative phase, accelerating wound contraction rates, and promoting vasculogenesis and angiogenesis. Therefore, there is potential for flavonoids to be used as a treatment for various chronic diseases that lead to cutaneous lesions, including diabetes mellitus, which causes many amputations worldwide [38,39,40,41].
Baicalin (5, Figure 2) is a flavone glycoside with anti-inflammatory, antiviral, and photoprotective properties [42]. Compound 5 blocks the pathological keratinocyte changes in psoriatic patients [43]. An antioxidant and anti-inflammatory effect may be responsible for the wound-healing properties of the baicalin nanohydrogel preparation. Moreover, 5 inhibits nitric oxide and tumor necrosis factor-α (TNF-α), which are both important elements in the inflammatory process [44]. Finally, 5 was found to exist in the endophytic fungus Spiropes sp. and played a major role in the pharmacological action of Scutellaria baicalensis [42,45].
Kaempferol (6, Figure 2) is known to inhibit cancer growth; reduce inflammation; promote antioxidant activity; and protect the heart, liver, and brain [46]. With a 1% w/w ointment concentration, 6 exhibited wound-healing activity in diabetic and non-diabetic rats. This wound-healing activity was studied utilizing incision and excision wound models [47]. According to both models, kaempferol showed crucial wound-healing activity, as indicated by the increase in the granulation tissue weight and hydroxyproline content in the incision wound model, as well as a reduction in the wound area, faster epithelialization, increased dry weight of the tissue, and increased hydroxyproline content in the excision wound model [48]. Moreover, 6 was found in many edible vegetables and the extract of the endophytic fungi Fusarium chlamydosporum from Tylophora indica [49,50,51].
Bioflavonoids such as quercetin (7, Figure 2) are known for their anti-atherosclerotic, anti-hypercholesterolemic, anti-hypertensive, anti-inflammatory, and anti-obesity properties [52]. Preclinical research has shown 7 to possess wound-healing properties [53,54]. A study by Jangde et al. evaluated quercetin’s wound-healing properties in vitro and in vivo. The results indicated that wound healing was significantly accelerated, and the wound termination time significantly decreased compared to the regular dosage forms. These results suggested that connectivity tissue disorders can be treated effectively as wound-healing approaches [54]. Quercetin was found in more than 20 plants [42]. Moreover, in terms of obtaining quercetin from bacteria, it was isolated for the first time from a cyanobacterium, Anabaena aequalis Borge [55].

4. Quinones

Biochemical pigments called quinones are established in several living organisms, including fungi, bacteria, a few animals, and higher plants. There are many forms of quinones in nature, such as naphthoquinones, benzoquinones, polycyclic quinones, and anthraquinones. Quinones are formed when polynuclear hydrocarbons, aromatic amines, and polyhydric phenols are oxidized [56]. Quinones and their derivatives are known to be potential antioxidants. In particular, hydroxyquinones are powerful antioxidants due to their dihydroxy groups located in the ortho position. Several hydroxyquinones, including pseudopurpurin, purpurin, and alizarin, are highly effective as antioxidants. In contrast, emodin, aloe-emodin, chrysophanol, and rhein have shown lower efficacy due to the absence of the ortho-dihydroxyl groups. This indicates that the ortho-dihydroxy structure of quinone and its derivatives significantly influences its radical scavenging properties [57].
A new antibiotic, MT81 (8, Figure 3), was isolated and purified from Penicillium nigricans culture media. It was found that 8 was a polyhydroxy anthraquinone, which exhibited antibacterial properties, along with an antiprotozoal effect toward Leishmania donovani promastigotes in vitro. Moreover, 8 was investigated for its wound-healing effect in mice. The result exhibited the healing effects of an 8 ointment on mice infected with pathogenic organisms, and it was comparable to the positive control, nitrofurazone. This indicates that the antimicrobial effect of 8 helps to heal infected wounds by inhibiting microorganisms [58].
Emodin (9, Figure 3) is a natural anthraquinone derivative found in a wide variety of higher plants. Emodin obtained from microorganisms was first described as frangula-emodin, which was isolated from the fungus Dermocybe sanguinea Wulf [59,60,61]. In addition, 9 was identified as one of the pigmented products in the culture extracts of Penicillium brunneum Udagawa, Cladosporium fulvum Cooke, Penicillium avellaneum, Penicilliopsis clavariaeformis, Penicillium islandicum Sopp, Aspergillus wentii Wehmer, Aspergillus ochraceus Wilhelm, and Aspergillus cristatus [62,63,64,65,66,67,68,69]. Neuroprotective, anti-inflammatory, anticancer, antibacterial, anti-osteoporotic, hepatoprotective, antiviral, anti-allergic, and immunosuppressive properties are known to be associated with 9 [70]. Furthermore, the wound-healing activity of 9 using the excisional wound model in rats has been reported at dose levels of 100, 200, and 400 μg/mL [71].
Hennotannic acid (10, Figure 3), also known as lawsone, is an orange-red dye extracted from the leaves of Lawsonia inermis L., often known as the henna tree. However, Sarang et al. reported the production of 10 from an endophytic fungus, Gibberella moniliformis, isolated from the leaf tissues of Lawsonia inermis [72]. Moreover, 10 exhibited antibacterial, antifungal, antiparasitic, antitumor, and antiviral properties [73]. By using an excision and incision model, Mandawgade and Patil investigated the wound-healing effects of 10 at dose levels of 50 mg/kg (per oral) and 0.1 mg/kg (topical/ointment). Based on the result, 10 exhibited significant wound-healing activity in both models [74]. Another study on 10 also demonstrated crucial wound-healing activity in rodents [75].

5. Phenolic Acids

Polyphenol is usually found in plants and marine organisms. Since polyphenols possess antimicrobial, regenerative, and antioxidant properties, they have significant potential for application in wound treatment [76,77,78]. Besides their antimicrobial and antioxidant activity, polyphenols are also considered to be highly bioactive agents in wound dressings for acute wound treatment, thereby playing an important role in wound healing [76,79].
Chlorogenic acid (11, Figure 4) is an ester of caffeic acid with phenolic acid (quinic acid), established in twenty medicinal plants and twenty-nine fungal taxa, including Phomopsis, Colleterotrichum, Phoma, Alternaria, and Xylariales [42,45]. Moreover, 11 showed significant wound-healing activity in both in vivo and in vitro investigations [80,81]. A recent study demonstrated that 11 promotes wound closure and capillary tube formation, as well as enhanced wound closure with keratinocytes in vitro [80]. In addition, 11 has been shown to enhance collagen synthesis by upregulating TNF-α and TGF-β during wound healing. It may promote wound healing in excision wounds due to its antioxidative properties [81].
Gallic acid (12, Figure 4) is a phenolic compound that can be found in plants, as well as nineteen endophytic fungal taxa [45]; 12 exhibits strong anti-inflammatory and antioxidant properties. During in vitro experiments, 12 accelerated keratinocyte and fibroblast migration and wound healing [82]. In addition, 12 enhanced the expression of TGF-β and inhibited the nuclear factor κB (NF-κB), along with the proliferation and maturation phase of wound healing [83].

6. Peptides

Several bioactive peptides have been explored for their prospective therapeutic use in wound healing [84]. Traditional wound-healing therapies, including cytokines, growth factors, immunomodulatory factors, and plant-derived chemicals, are particularly difficult to translate into clinical practice for acute wound healing [85]. Further, research on bioactive peptides with excessive specificity, stability, and activity has attracted considerable attention compared to drugs with high costs, low activity, and delivery and safety problems [84,86,87].
Acremonamide (13, Figure 5), a new cyclic depsipeptide obtained from Acremonium sp., was isolated in an ongoing effort to discover new marine-derived natural products with wound-healing effects [88]. It is known that species of the genus Acremonium produce various secondary metabolites, such as hydroquinones, diterpenes, isocoumarins, sesquiterpenes, peptides, benzophenones, and butanolides [89]. These fungal strains also produce cyclic depsipeptides, an interesting bioactive natural product [89,90,91,92]. In in vivo experiments using the human wound healing RT2 Profiler PCR array, adjustments in the wound-healing genes’ expression were screened to determine the mechanisms behind the wound-healing properties of 13. The result demonstrated, for the first time, that 13 increased keratinocyte and fibroblast motility, as well as COL1A2 and ACTC1 expression, thereby enhancing the wound-healing process [88].
Chondramide A (14, Figure 5) and its analogs isolated from a terrestrial myxobacterium, Chondramyces crocatus, were examined for the growth-inhibiting activity of actin cytoskeletons [93]. The actin cytoskeleton plays a significant role in wound healing—it allows actomyosin to contract, recruits repair machinery, and migrates cells [94]. By using highly purified recombinant actin from T. gondii, in vitro polymerization assays confirmed that both synthetic and natural products target the actin cytoskeleton, with EC50 values ranging from 0.3 to 1.3 μM. The results indicate that the chondramide treatment can prevent parasitic invasion and generate faster results than standard therapeutic agents such as pyrimethamine [95].
Other studies reported the wound-healing activity of jasplakinolide (15, Figure 5) and apratyramide (16, Figure 5) [96,97]. By binding to F-actin, 15 stabilized the actin filaments in vivo, leading to actin lumps and polynucleation, which are important in wound healing [96]. Moreover, 16, a natural product isolated from a cyanobacterium, has been described to exert wound-healing effects by inducing vascular endothelial growth factor A [97].

7. Triterpenoids

Terpenoids are plant-derived phytochemicals with a large variety of chemical structures derived from isoprene and usually have polycyclic structures. They are classified into monoterpenes, diterpenes, triterpenes, tetraterpenes, sesquiterpenes, and hemiterpenes, depending on the number of isoprene units in their structures. Triterpenes either have a tetracyclic or pentacyclic structure [98,99,100]. Among pentacyclic triterpenes, oleanane, lupane, and ursane derivatives exhibit anti-inflammatory, anticancer, antioxidant, antiviral, and cardioprotective activities. In contrast, tetracyclic triterpenes have mostly been studied for their cytotoxic and anticancer biological properties [101,102,103,104]. In recent years, systematic studies have examined the efficacy of triterpenes as wound healers. Based on the results of these studies, these phytocompounds have been displayed to promote wound healing by accelerating epithelialization and collagen formation and deposition, regardless of the wound type. Furthermore, their integration into various medicinal formulations is an effective option for wound management through their long-term delivery of active compounds. In conclusion, triterpenoids have been identified as an emerging class of wound care therapies [98,105].
Asiaticoside (17, Figure 6) is a triterpenoid saponin, which is found in Centella asiatica (L.), as well as in an endophytic fungus, Colletotrichum gloeosporioides, obtained from Centella asiatica (L.) [106,107]. Based on the ability of 17 to induce the development of granulation tissue and collagenase-induced epithelialization in rabbits, it was proven that 17 exhibits wound-healing properties [108]. In addition, 17 also induces type I collagen synthesis in human dermal fibroblast cells [109].

8. Others

In addition to their skin-rejuvenating properties, probiotics have also been described to ease atopic eczema, atopic dermatitis, wound healing, and the innate immunity of the skin. It has been demonstrated that non-viable cultures of lactic acid bacteria can exert antimicrobial and immunomodulatory effects. Several studies have shown that lactic acid bacteria compounds, such as cell wall fragments and metabolites, can enhance the skin barrier function and elicit particular immune responses [110,111,112].
Lipoteichoic acid (18, Figure 7) is an immune-stimulating systemic component in the cell walls of both pathogenic and non-pathogenic Gram-positive bacteria. It plays a crucial role in the growth and physiology of bacteria [113]. Previous research has revealed that 18 could distribute as a crucial pathogen-associated molecular motif, resulting in nitric oxide (NO) production and the stimulation of NF-ĸB, along with the construction of pro-inflammatory mediators and cytokines [114,115]. In addition, lipoteichoic acid from beneficial probiotics has been reported to induce tolerance by protecting against the production of the pro-inflammatory cytokines related to TNF-α sepsis and increasing the resistance to microbial infection among dermal cells [110]. Furthermore, upon the topical application of 18, it was found to promote skin protection against microbial infections by triggering toll-like receptors and displayed skin-wound-healing capabilities by activating human β-defensin mechanisms, in addition to its antimicrobial properties [116,117].
In chemical synthesis or microbial fermentation, lactic acid (19, Figure 7) is constructed as one of the α-hydroxy acids, which appear in the alpha position of the acid and comprise one hydroxyl group attached to the alpha position [118,119]. The use of lactic acid has been widespread for many years in skin-care products and cosmetic regimens, and it is known to show antibacterial properties that are effective against most pathogenic microbes [120,121]. In addition to its antimicrobial properties, 19 is considered a preventative remedy for acne vulgaris [122]. Further, because of its ability to improve the function of the stratum corneum barrier, 19 has shown potential for numerous skin applications, and it improved ceramide production by keratinocytes [123].
In heterofermentative lactic acid bacteria, acetic acid (20, Figure 7) can be produced from the hexose monophosphate or pentose pathway [124]. In particular, 20 has been used in treating microbial infections and superficial infections, as well as burns, on various occasions [125]. It has been shown to display antibacterial activity against many microbes, including P. aeruginosa and S. aureus [110]. When various antibiotic-resistant strains cause inflammation, and therapeutic options are inadequate, acetic acid has been recommended as the greatest treatment option [125].
Diacetyl (21, Figure 7), also known as 2,3-butanedione, can be produced by certain strains of Streptococcus, Leuconostoc, Pediococcus, and Lactobacillus, and 21 has shown possible antimicrobial dermal properties, with the highest sensitivity against Gram-negative bacteria and fungi rather than Gram-positive bacteria [121]. In addition, at a very low concentration of 100 ppm, 21 has been established to exert bactericidal activity against S. aureus and Escherichia coli. Although the antimicrobial activity of 21 has been proven thoroughly, there is little research on its topical function, and extensive investigation is required to determine how it affects the skin and other tissues [41,126,127,128].

9. Conclusions

This review highlighted several microbial natural products with antibacterial, anti-inflammatory, antioxidant, and actin cytoskeleton growth-inhibitory activities that are important in promoting wound-healing activities. Successful wound-healing treatment is crucial for the better quality of life of patients. Identifying the active compounds responsible for the wound-healing properties and their mechanism is currently challenging. There is a high demand for wound-healing agents; therefore, the upscaling of their production needs to be investigated. Several reviews have reported the wound-healing properties of plants and bioactive compounds derived from plants [3,129,130,131,132]. In addition, the antioxidant, antimicrobial, anticancer, and anti-inflammatory properties of microbial compounds have also been widely reviewed [11,133,134,135,136]. However, research on the wound-healing properties of microbial compounds is still lacking; therefore, further investigations on the wound-healing control of natural microbial products are required. This information may help to develop better treatments for wound healing.
Based on the reported results, compounds 1, 12, 13, 14, 15, 16, and 19 exert their wound-healing properties by accelerating keratinocyte migration to enhance wound closure. Moreover, compounds 8, 9, 10, 18, 19, 20, and 21 show antibacterial activity; the bioactive minimum inhibitory concentration (MIC) values of 9 were 28.9 µm for Bacillus subtilis and 14.4 µm for Staphylococcus aureus. However, 9 was not active against two Gram-negative bacteria (Klebsiella pneumoniae and Escherichia coli) at the highest concentration (1851.9 µm) tested [137]. Similarly, 9 has been reported to exhibit antimycobacterial and broad-spectrum antibacterial activity, particularly against M. tuberculosis (lowest MIC = 0.9 µm) and Gram-positive bacteria (lowest MIC < 14.8 µm) [69]. A broad range of MIC values of 10 was reported for Fusarium oxysporum (574 mmol/L), Aspergillus niger (861 mmol/L), A. flavus (287 mmol/L), and Candida albicans (2.933 mmol/L) [138]. In addition, compounds 1 and 3 prevented scar tissue formation, with a reduction in α-SMA expression. Compounds 3, 5, 11, and 18 reduced inflammation by decreasing the level of TNF-α, while compounds 1, 11, and 12 led to enhanced TGF-β expression. In the case of compound 11, it has been shown to enhance collagen synthesis by upregulating TNF-α and TGF-β during wound healing. Furthermore, compound 2 from Bacillus licheniformis increased 3T3/NIH fibroblast proliferation, compound 4 showed a significant increase in collagenase-1 expression in HT-29 colorectal adenocarcinoma cells, and compounds 6 and 7 are well known for their anti-inflammatory activity, which can significantly accelerate the increase in the granulation tissue weight. These results prove that all compounds can be used effectively as wound-healing treatments.

Author Contributions

Conceptualization, P.F.H. and S.-J.N.; investigation, P.F.H. and C.L.; writing—original draft preparation, P.F.H. and C.L.; writing—review and editing, P.F.H. and C.L.; supervision, S.-J.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Korean Government (Ministry of Science and ICT) grant no. 2022R1A2C1011848 to S.-J.N.

Acknowledgments

This research was supported by the National Research Foundation of Korea grant funded by the Korean Government (Ministry of Science and ICT; no. 2022R1A2C1011848 to S.-J.N.).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Barreto, R.S.; Albuquerque-Júnior, R.L.; Araújo, A.A.; Almeida, J.R.; Santos, M.R.; Barreto, A.S.; DeSantana, J.M.; Siqueira-Lima, P.S.; Quintans, J.S.; Quintans-Júnior, L.J. A systematic review of the wound-healing effects of monoterpenes and iridoid derivatives. Molecules 2014, 19, 846–862. [Google Scholar] [CrossRef] [PubMed]
  2. Guo, S.; Dipietro, L.A. Factors affecting wound healing. J. Dent. Res. 2010, 89, 219–229. [Google Scholar] [CrossRef] [PubMed]
  3. Ibrahim, N.I.; Wong, S.K.; Mohamed, I.N.; Mohamed, N.; Chin, K.-Y.; Ima-Nirwana, S.; Shuid, A.N. Wound Healing Properties of Selected Natural Products. Int J. Environ. Res. Public Health 2018, 15, 2360–2382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Kumar, B.; Vijayakumar, M.; Govindarajan, R.; Pushpangadan, P. Ethnopharmacological approaches to wound healing—Exploring medicinal plants of India. J. Ethnopharmacol. 2007, 114, 103–113. [Google Scholar] [CrossRef] [PubMed]
  5. Trinh, X.-T.; Long, N.-V.; Van Anh, L.T.; Nga, P.T.; Giang, N.N.; Chien, P.N.; Nam, S.-Y.; Heo, C.-Y. A Comprehensive Review of Natural Compounds for Wound Healing: Targeting Bioactivity Perspective. Int. J. Mol. Sci. 2022, 23, 9573. [Google Scholar] [CrossRef] [PubMed]
  6. Maheswary, T.; Nurul, A.A.; Fauzi, M.B. The Insights of Microbes’ Roles in Wound Healing: A Comprehensive Review. Pharmaceutics 2021, 13, 981–1002. [Google Scholar] [CrossRef] [PubMed]
  7. Thakur, R.; Jain, N.; Pathak, R.; Sandhu, S.S. Practices in wound healing studies of plants. Evid. Based Complement. Altern. Med. 2011, 2011, 438056. [Google Scholar] [CrossRef] [Green Version]
  8. Biswas, T.K.; Mukherjee, B. Plant medicines of Indian origin for wound healing activity: A review. Int. J. Low Extrem. Wounds 2003, 2, 25–39. [Google Scholar] [CrossRef]
  9. Laurano, R.; Boffito, M.; Ciardelli, G.; Chiono, V. Wound dressing products: A translational investigation from the bench to the market. Eng. Regen. 2022, 3, 182–200. [Google Scholar] [CrossRef]
  10. Fenical, W.; Jensen, P. Developing a new resource for drug discovery: Marine actinomycete bacteria. Nat. Chem. Biol. 2006, 2, 666–673. [Google Scholar] [CrossRef]
  11. Abdel-Razek, A.S.; El-Naggar, M.E.; Allam, A.; Morsy, O.M.; Othman, S.I. Microbial Natural Products in Drug Discovery. Processes 2020, 8, 470. [Google Scholar] [CrossRef] [Green Version]
  12. Kong, D.-X.; Jiang, Y.-Y.; Zhang, H.-Y. Marine natural products as sources of novel scaffolds: Achievement and concern. Drug Discov. Today 2010, 15, 884–886. [Google Scholar] [CrossRef] [PubMed]
  13. Araujo, J.; Monteiro, J.; Silva, D.; Alencar, A.; Silva, K.; Coelho, L.; Pacheco, W.; Silva, D.; Silva, M.; Silva, L.; et al. Surface-Active Compounds Produced by Microorganisms: Promising Molecules for the Development of Antimicrobial, Anti-Inflammatory, and Healing Agents. Antibiotics 2022, 11, 1106–1123. [Google Scholar] [CrossRef] [PubMed]
  14. Uzoigwe, C.; Burgess, J.G.; Ennis, C.J.; Rahman, P.K.S.M. Bioemulsifiers Are Not Biosurfactants and Require Different Screening Approaches. Front. Microbiol. 2015, 6, 245–250. [Google Scholar] [CrossRef] [Green Version]
  15. Sharma, D.; Misba, L.; Khan, A.U. Antibiotics versus Biofilm: An Emerging Battleground in Microbial Communities. Antimicrob. Resist. Infect. Control. 2019, 8, 76–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Kumari, A.; Kumari, S.; Prasad, G.S.; Pinnaka, A.K. Production of Sophorolipid Biosurfactant by Insect Derived Novel Yeast Metschnikowia churdharensis f.a., sp. nov., and Its Antifungal Activity against Plant and Human Pathogens. Front. Microbiol. 2021, 12, 678668–678680. [Google Scholar] [CrossRef] [PubMed]
  17. Ceresa, C.; Tessarolo, F.; Maniglio, D.; Tambone, E.; Carmagnola, I.; Fedeli, E.; Caola, I.; Nollo, G.; Chiono, V.; Allegrone, G.; et al. Medical-Grade Silicone Coated with Rhamnolipid R89 Is Effective against Staphylococcus spp. Biofilms. Molecules 2019, 24, 3843–3860. [Google Scholar] [CrossRef] [Green Version]
  18. Giugliano, R.; Buonocore, C.; Zannella, C.; Chianese, A.; Palma Esposito, F.; Tedesco, P.; De Filippis, A.; Galdiero, M.; Franci, G.; de Pascale, D. Antiviral Activity of the Rhamnolipids Mixture from the Antarctic Bacterium Pseudomonas gessardii M15 against Herpes simplex Viruses and Coronaviruses. Pharmaceutics 2021, 13, 2121–2138. [Google Scholar] [CrossRef]
  19. Kim, H.-Y.; Jung, H.; Kim, H.-M.; Jeong, H.-J. Surfactin Exerts an Anti-Cancer Effect through Inducing Allergic Reactions in Melanoma Skin Cancer. Int. Immunopharmacol. 2021, 99, 107934–107942. [Google Scholar] [CrossRef]
  20. Ceresa, C.; Fracchia, L.; Fedeli, E.; Porta, C.; Banat, I.M. Recent Advances in Biomedical, Therapeutic and Pharmaceutical Applications of Microbial Surfactants. Pharmaceutics 2021, 13, 466–491. [Google Scholar] [CrossRef]
  21. Rahim, K.; Saleha, S.; Zhu, X.; Huo, L.; Basit, A.; Franco, O.L. Bacterial Contribution in Chronicity of Wounds. Microb. Ecol. 2017, 73, 710–721. [Google Scholar] [CrossRef] [PubMed]
  22. Pastar, I.; Nusbaum, A.G.; Gil, J.; Patel, S.B.; Chen, J.; Valdes, J.; Stojadinovic, O.; Plano, L.R.; Tomic-Canic, M.; Davis, S.C. Interactions of Methicillin Resistant Staphylococcus aureus USA300 and Pseudomonas aeruginosa in Polymicrobial Wound Infection. PLoS ONE 2013, 8, e56846–e56857. [Google Scholar] [CrossRef] [PubMed]
  23. Banu, A.; Noorul Hassan, M.M.; Rajkumar, J.; Srinivasa, S. Spectrum of Bacteria Associated with Diabetic Foot Ulcer and Biofilm Formation: A Prospective Study. Australas. Med. J. 2015, 8, 280–285. [Google Scholar] [CrossRef] [PubMed]
  24. Ohadi, M.; Forootanfar, H.; Rahimi, H.R.; Jafari, E.; Shakibaie, M.; Eslaminejad, T.; Dehghannoudeh, G. Antioxidant Potential and Wound Healing Activity of Biosurfactant Produced by Acinetobacter junii B6. Curr. Pharm. Biotechnol. 2017, 18, 900–908. [Google Scholar] [CrossRef]
  25. Zouari, R.; Moalla-Rekik, D.; Sahnoun, Z.; Rebai, T.; Ellouze-Chaabouni, S.; Ghribi-Aydi, D. Evaluation of Dermal Wound Healing and in vitro Antioxidant Efficiency of Bacillus Subtilis SPB1 Biosurfactant. Biomed. Pharmacother. 2016, 84, 878–891. [Google Scholar] [CrossRef]
  26. Yan, L.; Liu, G.; Zhao, B.; Pang, B.; Wu, W.; Ai, C.; Zhao, X.; Wang, X.; Jiang, C.; Shao, D.; et al. Novel Biomedical Functions of Surfactin A from Bacillus subtilis in Wound Healing Promotion and Scar Inhibition. J. Agric. Food Chem. 2020, 68, 6987–6997. [Google Scholar] [CrossRef]
  27. Gupta, S.; Raghuwanshi, N.; Varshney, R.; Banat, I.M.; Srivastava, A.K.; Pruthi, P.A.; Pruthi, V. Accelerated in Vivo Wound Healing Evaluation of Microbial Glycolipid Containing Ointment as a Transdermal Substitute. Biomed. Pharmacother. 2017, 94, 1186–1196. [Google Scholar] [CrossRef] [PubMed]
  28. Shen, C.; Jiang, L.; Shao, H.; You, C.; Zhang, G.; Ding, S.; Bian, T.; Han, C.; Meng, Q. Targeted Killing of Myofibroblasts by Biosurfactant di-Rhamnolipid Suggests a Therapy against Scar Formation. Sci. Rep. 2016, 6, 37553–37563. [Google Scholar] [CrossRef] [Green Version]
  29. Maeng, Y.; Kim, K.T.; Zhou, X.; Jin, L.; Kim, K.S.; Kim, Y.H.; Lee, S.; Park, J.H.; Chen, X.; Kong, M.; et al. A Novel Microbial Technique for Producing High-Quality Sophorolipids from Horse Oil Suitable for Cosmetic Applications. Microb. Biotechnol. 2018, 11, 917–929. [Google Scholar] [CrossRef]
  30. Kwak, M.-J.; Park, M.-Y.; Kim, J.; Lee, H.; Whang, K.-Y. Curative Effects of Sophorolipid on Physical Wounds: In Vitro and in Vivo Studies. Vet. Med. Sci. 2021, 7, 1400–1408. [Google Scholar] [CrossRef]
  31. Carvalho, M.T.B.; Araújo-Filho, H.G.; Barreto, A.S.; Quintans-Júnior, L.J.; Quintans, J.S.S.; Barreto, R.S.S. Wound healing properties of flavonoids: A systematic review highlighting the mechanisms of action. Phytomedicine 2021, 90, 153636–153651. [Google Scholar] [CrossRef] [PubMed]
  32. Menezes, P.D.P.; Frank, L.A.; Lima, B.S.S.; de Carvalho, Y.M.B.G.; Serafini, M.R.; Quintans-Júnior, L.J.; Pohlmann, A.R.; Guterres, S.S.; Araújo, A.A.d.S. Hesperetin-loaded lipid-core nanocapsules in polyamide: A new textile formulation for topical drug delivery. Int. J. Nanomed. 2017, 12, 2069–2079. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Sak, K. Intake of individual flavonoids and risk of carcinogenesis: Overview of epidemiological evidence. Nutr. Cancer 2017, 69, 1119–1150. [Google Scholar] [CrossRef]
  34. Pushpavalli, G.; Kalaiarasi, P.; Veeramani, C.; Pugalendi, K.V. Effect of chrysin on hepatoprotective and antioxidant status in D-galactosamine-induced hepatitis in rats. Eur. J. Pharmacol. 2010, 631, 36–41. [Google Scholar] [CrossRef] [PubMed]
  35. Bagher, Z.; Ehterami, A.; Safdel, M.H.; Khastar, H.; Semiari, H.; Asefnejad, A.; Davachi, S.M.; Mirzaii, M.; Salehi, M. Wound healing with alginate/chitosan hydrogel containing hesperidin in rat model. J. Drug Deliv. Sci. Technol. 2020, 55, 101379. [Google Scholar] [CrossRef]
  36. Feng, Y.; Sanders, A.J.; Morgan, L.D.; Harding, K.G.; Jiang, W.G. Potential roles of suppressor of cytokine signaling in wound healing. Regen. Med. 2016, 11, 193–209. [Google Scholar] [CrossRef]
  37. Chen, R.F.; Chang, C.H.; Wang, C.T.; Yang, M.Y.; Wang, C.J.; Kuo, Y.R. Modulation of vascular endothelial growth factor and mitogen-activated protein kinase-related pathway involved in extracorporeal shockwave therapy accelerate diabetic wound healing. Wound Repair Regen. 2019, 27, 69–79. [Google Scholar] [CrossRef] [Green Version]
  38. Alexandru, V.; Balan, M.; Gaspar, A.; Craciunescu, O.; Moldovan, L. Studies on the antioxidant activity, phenol and flavonoid contents of some selected Romanian medicinal plants used for wound healing. Rom. Biotechnol. Lett. 2007, 12, 3467–3472. [Google Scholar]
  39. Haddadi, G.; Abbaszadeh, A.; Mosleh-Shirazi, M.A.; Okhovat, M.A.; Salajeghe, A.; Ghorbani, Z. Evaluation of the effect of hesperidin on vascular endothelial growth factor gene expression in rat skin animal models following cobalt-60 gamma irradiation. J. Cancer Res. Ther. 2018, 14, S1098–S1104. [Google Scholar]
  40. Kandhare, A.D.; Alam, J.; Patil, M.V.K.; Sinha, A.; Bodhankar, S.L. Wound healing potential of naringin ointment formulation via regulating the expression of inflammatory, apoptotic and growth mediators in experimental rats. Pharm. Biol. 2016, 54, 419–432. [Google Scholar] [CrossRef]
  41. Singh, W.R.; Devil, H.S.; Kumawat, S.; Sadam, A.; Appukuttan, A.; Patel, M.R.; Lingaraju, M.C.; Singh, T.U.; Kumar, D. Angiogenic and MMPs modulatory effects of icariin improved cutaneous wound healing in rats. Eur. J. Pharmacol. 2019, 858, 172466. [Google Scholar] [CrossRef] [PubMed]
  42. Parasuraman, S.; Perumal, P. Wound Healing Agents from Natural Sources. In Wound Healing Research, 1st ed.; Kumar, P., Kothari, V., Eds.; Springer: Singapore, 2021; pp. 95–148. [Google Scholar]
  43. Bonesi, M.; Loizzo, M.R.; Menichini, F.; Tundis, R. Flavonoids in treating psoriasis. In Immunity and Inflammation in Health and Disease, 1st ed.; Chatterjee, S., Jungraithmayr, W., Bagchi, D., Eds.; Academic Press: Cambridge, MA, USA, 2018; pp. 281–294. [Google Scholar]
  44. Manconi, M.; Manca, M.L.; Caddeo, C.; Cencetti, C.; di Meo, C.; Zoratto, N.; Nacher, A.; Fadda, A.M.; Matricardi, P. Preparation of gellan-cholesterol nanohydrogels embedding baicalin and evaluation of their wound healing activity. Eur. J. Pharm. Biopharm. 2018, 127, 244–249. [Google Scholar] [CrossRef] [PubMed]
  45. Huang, W.Y.; Cai, Y.Z.; Hyde, K.D.; Corke, H.; Sun, M. Biodiversity od endophytic fungi associated with 29 traditional Chinese medicinal plants. Fungal Divers. 2008, 33, 61–75. [Google Scholar]
  46. Ren, J.; Lu, Y.; Qian, Y.; Chen, B.; Wu, T.; Ji, G. Recent progress regarding kaempferol for the treatment of various diseases. Exp. Med. 2019, 18, 2759–2776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Özay, Y.; Güzel, S.; Yumrutaş, Ö.; Pehlivanoğlu, B.; Erdoğdu, İ.H.; Yildirim, Z.; Türk, B.A.; Darcan, S. Wound healing effect of kaempferol in diabetic and nondiabetic rats. J. Surg. Res. 2019, 233, 284–296. [Google Scholar] [CrossRef] [PubMed]
  48. Ambiga, S.; Narayanan, R.; Gowri, D.; Sukumar, D.; Madhavan, S. Evaluation of wound healing activity of flavonoids from Ipomoea carnea Jacq. Anc. Sci. Life 2007, 26, 45–51. [Google Scholar] [PubMed]
  49. Calderón-Montaño, J.M.; Burgos-Morón, E.; Pérez-Guerrero, C.; López-Lázaro, M. A review on the dietary flavonoid kaempferol. Mini Rev. Med. Chem. 2011, 11, 298–344. [Google Scholar] [CrossRef]
  50. Liu, R.H. Health-promoting components of fruits and vegetables in the diet. Adv. Nutr. 2013, 4, 384S–392S. [Google Scholar] [CrossRef] [Green Version]
  51. Pratibha, C.; Gajbhiye, S.; Roy, S.; Dudhale, R.; Chowdhary, A. Determination of Kaempferol in extracts of Fusarium chlamydosporum, an endophyticfungi of Tylophora indica (Asclepeadaceae) and its anti-microbial activity. Int. J. Pharm. Biol. Sci. 2014, 9, 51–55. [Google Scholar]
  52. David, A.V.A.; Arulmoli, R.; Parasuraman, S. Overviews of biological importance of quercetin: A bioactive flavonoid. Pharmacogn. Rev. 2016, 10, 84–89. [Google Scholar]
  53. Gomathi, K.; Gopinath, D.; Ahmed, M.R.; Jayakumar, R. Quercetin incorporated collagen matrices for dermal wound healing processes in rat. Biomaterials 2003, 24, 2767–2772. [Google Scholar] [CrossRef] [PubMed]
  54. Jangde, R.; Srivastava, S.; Singh, M.R.; Singh, D. In vitro and in vivo characterization of quercetin loaded multiphase hydrogel for wound healing application. Int. J. Biol. Macromol. 2018, 115, 1211–1217. [Google Scholar] [CrossRef] [PubMed]
  55. Abdel-Raouf, N.; Ibraheem, I.B.M.; Abdel-Tawab, S.; Naser, Y.A.G. Antimicrobial and antihyperlipidemic activities of isolated quercetin from anabaena aequalis(1). J. Phycol. 2011, 47, 955–962. [Google Scholar] [CrossRef] [PubMed]
  56. Devi, S.S.; Mehendale, H.M. Quinone. In Encyclopedia of Toxicology, 3rd ed.; Wxler, P., Ed.; Elsevier Inc.: Amsterdam, The Netherlands, 2014; pp. 26–28. [Google Scholar]
  57. Desam, N.R.; Al-Rajab, A.J. Herbal biomolecules: Anticancer agents. In Herbal Biomolecules in Health-Care Application, 1st ed.; Mandal, S.C., Dhara, A.K., Nayak, A.K., Eds.; Elsevier: Amsterdam, The Netherlands, 2022; pp. 435–474. [Google Scholar]
  58. Chatterjee, T.K.; Chakravorty, A. Wound healing properties of the new antibiotic (MT81) in mice. Indian Drugs 1993, 30, 450–452. [Google Scholar]
  59. Ismaiel, A.A.; Rabie, G.H.; Abd El-Aal, M.A. Antimicrobial and morphogenic effects of emodin produced by Aspergillus awamori WAIR120. Biologia 2016, 71, 464–474. [Google Scholar] [CrossRef]
  60. Izhaki, I. Emodin-a secondary metabolite with multiple ecological functions in higher plants. New Phytol. 2002, 155, 205–217. [Google Scholar] [CrossRef]
  61. Kögl, F.; Postowsky, J.J. Untersuchungen über Pilzfarbstoffe. II. Über die Farbstoffe des blutroten Hautkopfes (Dermocybe sanguinea Wulf.). Justus Liebigs Ann. Chem. 1925, 444, 1–7. [Google Scholar] [CrossRef]
  62. Agosti, G.; Birkinshaw, J.H.; Chaplen, P. Studies in the biochemistry of micro-organisms. 112. Anthraquinone pigments of strains of Cladosporium fulvum Cooke. Biochem. J. 1962, 85, 528–530. [Google Scholar] [CrossRef] [Green Version]
  63. Shibata, S.; Shoji, J.; Ohta, A.; Watanable, M. Metabolic products of fungi. XI. Some observations on the occurrence of skyrin and rugulosin in mold metabolites with reference to structural relationships between penicilliopsin and skyrin. Chem. Pharm. Bull. 1957, 5, 380–383. [Google Scholar] [CrossRef] [Green Version]
  64. Shibata, S.; Udagawa, S. Metabolic products of fungi. XIX. Isolation of rugulosin from Penicillium brunneum Udagawa. Chem. Pharm. Bull. 1963, 11, 402–403. [Google Scholar] [CrossRef] [Green Version]
  65. Natori, S.; Sato, F.; Udagawa, S. Anthraquinone metabolites of Talaromyces avellanens (Thom et Turreson), C.R. Benjamin and Preussia multispora (Saito et Minoura) Cain. Chem. Pharm. Bull. 1965, 13, 385–389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Ghosh, A.C.; Manmade, A.; Demain, A.L. Toxins from Penicillium islandicum Sopp. [in food, feed]. In Mycotoxins in Human and Animal Health; Rodricks, J.V., Hesseltine, C.W., Mehlman, M.A., Eds.; Pathotox: Chicago, IL, USA, 1977; pp. 625–638. [Google Scholar]
  67. Yamazaki, M.; Maebayashi, Y.; Miyaki, K. The isolation of secalonic acid A from Aspergillus ochraceus cultured on rice. Chem. Pharm. Bull. 1971, 19, 199–201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Wells, J.M.; Cole, R.J.; Kirksey, J.W. Emodin, a toxic metabolite of Aspergillus wentii isolated from weevil-damaged chestnuts. Appl. Microbiol. 1975, 30, 26–28. [Google Scholar] [CrossRef]
  69. Anke, H.; Kolthoum, I.; Laatsch, H. Metabolic products of microorganisms. 192. The anthraquinones of the Aspergillus glaucus group. II. Biological activity. Arch. Microbiol. 1980, 126, 231–236. [Google Scholar] [CrossRef] [PubMed]
  70. Dong, X.; Fu, J.; Yin, X.; Cao, S.; Li, X.; Lin, L.; Huyiligeqi, N.J. Emodin: A review of its pharmacology, toxicity and pharmacokinetics. Phytother. Res. 2016, 30, 1207–1218. [Google Scholar] [CrossRef] [PubMed]
  71. Tang, T.; Yin, L.; Yang, J.; Shan, G. Emodin, an anthraquinone derivative from Rheum officinale Baill, enhances cutaneous wound healing in rats. Eur. J. Pharm. 2007, 567, 177–185. [Google Scholar] [CrossRef]
  72. Sarang, H.; Rajani, P.; Vasanthakumari, M.M.; Kumara, P.M.; Siva, R.; Ravikanth, G.; Shaanker, R.U. An endophytic fungus, Gibberella moniliformis from Lawsonia inermis L. produces lawsone, an orange-red pigment. Antonie Van Leeuwenhoek 2017, 110, 853–862. [Google Scholar] [CrossRef]
  73. López López, L.I.; Nery Flores, S.D.; Silva Belmares, S.Y.; Sáenz, G.A. Naphthoquinones: Biological properties and synthesis of lawsone and derivatives-a structured review. Vitae 2014, 21, 248–258. [Google Scholar] [CrossRef]
  74. Mandawgade, S.D.; Patil, K.S. Wound healing potential of some active principles of Lawsonia alba Lam. leaves. Indian J. Pharm. Sci. 2003, 65, 390–394. [Google Scholar]
  75. Bascha, J.; Murthy, B.R.; Likhitha, P.R.; Ganesh, Y.; Bai, B.G.; Rani, R.J.; Prakash, P.; Shanmugham, V.; Kirthi, A. In vitro and in vivo assessment of lawsone microsphere loaded chitosan scaffolds. Int. J. Phytopharm. 2016, 6, 74–84. [Google Scholar]
  76. Guimaraes, I.; Baptista-Silva, S.; Pintado, M.; Oliveira, A.L. Poplyphenols: A promising Avenue in Therapeutic Solutions for Wound Care. Appl. Sci. 2021, 11, 1230. [Google Scholar] [CrossRef]
  77. Działo, M.A.; Mierziak, J.; Korzun, U.; Preisner, M.; Szopa, J.; Kulma, A. The potential of plant phenolics in prevention and therapy of skin disorders. Int. J. Mol. Sci. 2016, 17, 160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Ghuman, S.; Ncube, B.; Finnie, J.; McGaw, L.; Njoya, E.M.; Coopoosamy, R.; Van Staden, J. Antioxidant, anti-inflammatory and wound healing properties of medicinal plant extracts used to treat wounds and dermatological disorders. S. Afr. J. Bot. 2019, 126, 232–240. [Google Scholar] [CrossRef]
  79. Krausz, A.E.; Adler, B.L.; Cabral, V.; Navati, M.; Doerner, J.; Charafeddine, R.A.; Chandra, D.; Liang, H.; Gunther, L.; Clendaniel, A.; et al. Curcumin-encapsulated nanoparticles as innovative antimicrobial and wound healing agent. Nanomed. Nanotechnol. Biol. Med. 2015, 11, 195–206. [Google Scholar] [CrossRef] [Green Version]
  80. Moghadam, S.E.; Ebrahimi, S.N.; Salehi, P.; Moridi Farimani, M.; Hamburger, M.; Jabbarzadeh, E. Wound healing potential of chlorogenic acid and myricetin-3-O-β-rhamnoside isolated from Parrotia persica. Molecules 2017, 22, 1501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Chen, W.C.; Liou, S.S.; Tzeng, T.F.; Lee, S.L.; Liu, I.M. Effect of topical application of chlorogenic acid on excision wound healing in rats. Planta Med. 2013, 79, 616–621. [Google Scholar] [CrossRef] [PubMed]
  82. Yang, D.J.; Moh, S.H.; Son, D.H.; You, S.; Kinyua, A.W.; Ko, C.M.; Song, M.; Yeo, J.; Choi, Y.H.; Kim, K.W. Gallic acid promotes wound healing in normal and hyperglucidic conditions. Molecules 2016, 21, 899. [Google Scholar] [CrossRef] [Green Version]
  83. Karatas, O.; Gevrek, F. Gallic acid liposome and powder gels improved wound healing in wistar rats. Ann. Med. Res. 2019, 26, 2720–2727. [Google Scholar] [CrossRef]
  84. Song, Y.; Wu, C.; Zhang, X.; Bian, W.; Liu, N.; Yin, S.; Yang, M.; Luo, M.; Tang, J.; Yang, X. A short peptide potentially promotes the healing of skin wound. Biosci. Rep. 2019, 39, BSR20181734. [Google Scholar] [CrossRef]
  85. Larouche, J.; Sheoran, S.; Maruyama, K.; Martino, M.M. Immune regulation of skin wound healing: Mechanisms and novel therapeutic targets. Adv. Wound Care 2018, 7, 209–231. [Google Scholar] [CrossRef]
  86. Li, X.; Wang, Y.; Zou, Z.; Yang, M.; Wu, C.; Su, Y.; Tang, J.; Yang, X. OM-LV20, a novel peptide from odorous frog skin, accelerates wound healing in vitro and in vivo. Chem. Biol. Drug Des. 2018, 91, 126–136. [Google Scholar] [CrossRef] [PubMed]
  87. Hardwicke, J.; Schmaljohann, D.; Boyce, D.; Thomas, D. Epidermal growth factor therapy and wound healing–past, present and future perspectives. Surgeon 2008, 6, 172–177. [Google Scholar] [CrossRef] [PubMed]
  88. Kim, S.; Lee, C.W.; Park, S.-Y.; Asolkar, R.N.; Kim, H.; Kim, G.J.; Oh, S.J.; Kim, Y.; Lee, E.-Y.; Oh, D.-C.; et al. Acremonamide, a Cyclic Pentadepsipeptide with Wound-Healing Properties Isolated from a Marine-Derived Fungus of the Genus Acremonium. J. Nat. Prod. 2021, 84, 2249–2255. [Google Scholar] [CrossRef] [PubMed]
  89. Wang, X.; Gong, X.; Li, P.; Lai, D.; Zhou, L. Structural Diversity and Biological Activities of Cyclic Depsipeptides from Fungi. Molecules 2018, 23, 169–218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Ratnayake, R.; Fremlin, L.J.; Lacey, E.; Gill, J.H.; Capon, R.J. Acremolides A−D, Lipodepsipeptides from an Australian Marine-Derived Fungus, Acremonium sp. J. Nat. Prod. 2008, 71, 403–408. [Google Scholar] [CrossRef]
  91. Sun, P.; Maloney, K.N.; Nam, S.-J.; Haste, N.M.; Raju, R.; Aalbersberg, W.; Jensen, P.R.; Nizet, V.; Hensler, M.E.; Fenical, W. Fijimycins A–C, three antibacterial etamycin-class depsipeptides from a marine-derived Streptomyces sp. Bioorg. Med. Chem. 2011, 19, 6557–6562. [Google Scholar] [CrossRef] [Green Version]
  92. Amagata, T.; Morinaka, B.I.; Amagata, A.; Tenney, K.; Valeriote, F.A.; Lobkovsky, E.; Clardy, J.; Crews, P. A Chemical Study of Cyclic Depsipeptides Produced by a Sponge-Derived Fungus. J. Nat. Prod. 2006, 69, 1560–1565. [Google Scholar] [CrossRef] [Green Version]
  93. Kunze, B.; Jansen, R.; Sasse, F.; Hofle, G.; Reichenbach, H. Chondramides A~D, New Antifungal and Cytostatic Depsipeptides from Chondromyces crocatus (Myxobacteria). J. Antibiot. Res. 1995, 48, 1262–1266. [Google Scholar] [CrossRef]
  94. Abreu-Blanco, M.T.; Watts, J.J.; Verboon, J.M.; Parkhurst, S.M. Cytoskeleton Response in Wound Repair. Cell Mol. Life Sci. 2012, 69, 2469–2483. [Google Scholar] [CrossRef] [Green Version]
  95. Ma, C.I.; Diraviyam, K.; Maier, M.E.; Sept, D.; Sibley, L.D. Synthetic chondramide A analogues stabilize filamentous actin and block invasion by Toxoplasma gondii. J. Nat. Prod. 2013, 76, 1565–1572. [Google Scholar] [CrossRef]
  96. Tannert, R.; Milroy, L.G.; Ellinger, B.; Hu, T.S.; Arndt, H.D.; Waldmann, H. Synthesis and structure-activity correlation of natural-product inspired cyclodepsipeptides stabilizing F-actin. J. Am. Chem. Soc. 2010, 132, 3063–3077. [Google Scholar] [CrossRef]
  97. Cai, W.; Salvador-Reyes, L.A.; Zhang, W.; Chen, Q.-Y.; Matthew, S.; Ratnayake, R.; Seo, S.J.; Dolles, S.; Gibson, D.J.; Paul, V.J.; et al. Apratyramide, a Marine-Derived Peptidic Stimulator of VEGF-A and Other Growth Factors with Potential Application in Wound Healing. ACS Chem. Biol. 2018, 13, 91–99. [Google Scholar] [CrossRef] [PubMed]
  98. Ghiulai, R.; Roşca, O.J.; Antal, D.S.; Mioc, M.; Mioc, A.; Racoviceanu, R.; Macaşoi, I.; Olariu, T.; Dehelean, C.; Creţu, O.M.; et al. Tetracyclic and Pentacyclic Triterpenes with High Therapeutic Efficiency in Wound Healing Approaches. Molecules 2020, 25, 5557. [Google Scholar] [CrossRef] [PubMed]
  99. Isah, M.B.; Tajuddeen, N.; Umar, M.I.; AlHafiz, Z.A.; Mohammed, A.; Ibrahim, M.A. Terpenoids as emerging therapeutic agents: Cellular targets and mechanisms of action against protozoan parasites. In Studies in Natural Products Chemistry, 1st ed.; Atta-ur-Rahman, Ed.; Elsevier: Amsterdam, The Netherlands, 2018; Volume 59, pp. 227–250. [Google Scholar]
  100. Furtado, N.A.J.C.; Pirson, L.; Edelberg, H.; Miranda, L.M.; Loira-Pastoriza, C.; Préat, V.; Larondelle, Y.; Andre, C.M. Pentacyclic triterpene bioavailability: An overview of in vitro and in vivo studies. Molecules 2017, 22, 400. [Google Scholar] [CrossRef] [Green Version]
  101. Paduch, R.; Kandefer-Szerszen, M. Antitumor and antiviral activity of pentacyclic triterpenes. Mini-Rev. Org. Chem. 2014, 11, 262–268. [Google Scholar] [CrossRef]
  102. Chudzik, M.; Korzonek-Szlacheta, I.; Król, W. Triterpenes as potentially cytotoxic compounds. Molecules 2015, 20, 1610–1625. [Google Scholar] [CrossRef] [Green Version]
  103. Battineni, J.K.; Koneti, P.K.; Bakshi, V.; Boggula, N. Triterpenoids: A review. Int. J. Pharm. Pharm. Sci. 2018, 3, 91–96. [Google Scholar]
  104. Hill, R.A.; Connolly, J.D. Triterpenoids. Nat. Prod. Rep. 2017, 34, 90–122. [Google Scholar] [CrossRef] [Green Version]
  105. Agra, L.C.; Ferro, J.N.S.; Barbosa, F.T.; Barreto, E. Triterpenes with healing activity: A systematic review. J. Dermatol. Treat. 2015, 26, 465–470. [Google Scholar] [CrossRef]
  106. Sh Ahmed, A.; Taher, M.; Mandal, U.K.; Jaffri, J.M.; Susanti, D.; Mahmood, S.; Zakaira, Z.A. Pharmacological properties of Centella asiatica hydrogel in accelerating wound healing in rabbits. BMC Complement. Altern. Med. 2019, 19, 213. [Google Scholar] [CrossRef] [Green Version]
  107. Gupta, S.; Bhatt, P.; Chaturvedi, P. Determination and quantification of asiaticoside in endophytic fungus from Centella asiatica (L.) Urban. World J. Microbiol. Biotechnol. 2018, 34, 111. [Google Scholar] [CrossRef] [PubMed]
  108. Ozdemir, O.; Ozkan, K.; Hatipoglu, F.; Uyaroglu, A.; Arican, M. Effect of asiaticoside, collagenase, and alpha-chymotrypsin on wound healing in rabbits. Wounds 2016, 28, 279–286. [Google Scholar] [PubMed]
  109. Mukherjee, P.K.; Bahadur, S.; Chaudhary, S.K.; Harwansh, R.K.; Nema, N.K. Validation of medicinal herbs for skin aging. In Evidence-Based Validation of Berbal Medicine, 1st ed.; Mukherjee, P.K., Ed.; Elsevier: Amsterdam, The Netherlands, 2015; pp. 119–147. [Google Scholar]
  110. Lew, L.C.; Liong, M.T. Bioactives from probiotics for dermal health: Functions and benefits. J. Appl. Microbiol. 2013, 114, 1241–1253. [Google Scholar] [CrossRef] [PubMed]
  111. Arck, P.; Handjiski, B.; Hagen, E.; Pincus, M.; Bruenahl, C.; Bienenstock, J.; Paus, R. Is there a ‘gut-brainskin axis’? Exp. Derm. 2010, 19, 401–405. [Google Scholar] [CrossRef] [PubMed]
  112. Iordache, F.; Iordache, C.; Chifiriuc, M.C.; Bleotu, C.; Pavel, M.; Smarandache, D.; Sasarman, E.; Laza, V.; Bucu, M.; Dracea, O.; et al. Antimicrobial and immunomodulatory activity of some probiotic fractions with potential clinical application. Arch. Zootc. 2008, 11, 41–51. [Google Scholar]
  113. Villéger, R.; Saad, N.; Grenier, K.; Falourd, X.; Foucat, L.; Urdaci, M.C.; Bressollier, P.; Ouk, T.S. Characterization of lipoteichoic acid structures from three probiotic Bacillus strains: Involvement of D-alanine in their biological activity. Antonie Van Leeuwenhoek 2014, 106, 693–706. [Google Scholar] [CrossRef] [Green Version]
  114. Kao, S.J.; Lei, H.C.; Kuo, C.T.; Chang, M.S.; Chen, B.C.; Chang, Y.C.; Chiu, W.T.; Lin, C.H. Lipoteichoic acid induces nuclear factor-κB activation and nitric oxide synthase expression via phosphatidylinositol 3-kinase, Akt, and p38 MAPK in RAW 264.7 macrophages. Immunology 2005, 115, 366–374. [Google Scholar] [CrossRef]
  115. Lebeer, S.; Claes, I.J.; Vanderleyden, J. Anti-inflammatory potential of probiotics: Lipoteichoic acid makes a difference. Trends Microbiol. 2012, 20, 5–10. [Google Scholar] [CrossRef]
  116. Sumikawa, Y.; Asada, H.; Hoshino, K.; Azukizawa, H.; Katayama, I.; Akira, S.; Itami, S. Induction of β-defensin 3 in keratinocytes stimulated by bacterial lipopeptides through toll-like receptor 2. Microbes Infect. 2006, 8, 1513–1521. [Google Scholar] [CrossRef]
  117. Diamond, G.; Beckloff, N.; Weinberg, A.; Kisich, K.O. The roles of antimicrobial peptides in innate host defense. Curr. Pharm. Des. 2009, 15, 2377–2392. [Google Scholar] [CrossRef]
  118. Wee, Y.J.; Kim, J.N.; Ryu, H.W. Biotechnological production of lactic acid and its recent applications. Food Technol. Biotechnol. 2006, 44, 163–172. [Google Scholar]
  119. Tang, S.C.; Yang, J.H. Dual effects of alpha-hydroxy acids on the skin. Molecules 2018, 23, 863. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Lew, L.C.; Gan, C.Y.; Liong, M.T. Dermal bioactives from Lactobacilli and Bifidobacteria. Ann. Microbiol. 2013, 63, 1047–1055. [Google Scholar] [CrossRef]
  121. Smith, W.P. Epidermal and dermal effects of topical lactic acid. J. Am. Acad. Dermatol. 1996, 35, 388–391. [Google Scholar] [CrossRef]
  122. Pasricha, A.; Bhalla, P.; Sharma, K.B. Evaluation of lactic acid as an antibacterial agent. Indian J. Dermatol. Venereol. Leprol. 1979, 45, 159–161. [Google Scholar]
  123. Yamamoto, Y.; Uede, K.; Yonei, N.; Kishioka, A.; Ohtani, T.; Furukawa, F. Effects of alphahydroxy acids on the human skin of Japanese subjects: The rationale for chemical peeling. J. Dermatol. 2006, 33, 16–22. [Google Scholar] [CrossRef]
  124. Yeo, S.K.; Liong, M.T. Effect of prebiotics on viability and growth characteristics of probiotics in soymilk. J. Sci. Food Agric. 2010, 90, 267–275. [Google Scholar] [CrossRef]
  125. Nagoba, B.; Wadher, B.; Kulkarni, P.; Kolhe, S. Acetic acid treatment of pseudomonal wound infections. Eur. J. Gen. Med. 2008, 5, 104–106. [Google Scholar] [CrossRef]
  126. Lanciotti, R.; Patrignani, F.; Bagnolini, F.; Guerzoni, M.E.; Gardini, F. Evaluation of diacetyl antimicrobial activity against Escherichia coli, Listeria monocytogenes and Staphylococcus aureus. Food Microbiol. 2003, 20, 537–543. [Google Scholar] [CrossRef]
  127. Miller, L.S.; Cho, J.S. Immunity against Staphylococcus aureus cutaneous infections. Nat. Rev. Immunol. 2011, 11, 505–518. [Google Scholar] [CrossRef] [Green Version]
  128. Doern, G.V.; Jones, R.N.; Pfaller, M.A.; Kugler, K.C.; Beach, M.L. Bacterial pathogens isolated from patients with skin and soft tissue infections: Frequency of occurrence and antimicrobial susceptibility patterns from the SENTRY Antimicrobial Surveillance Program (United States and Canada, 1997). SENTRY Study Group (North America). Diagn. Microbiol. Infect. Dis. 1999, 34, 65–72. [Google Scholar] [PubMed]
  129. Salama, M.; Elgadir, M.A.; Adam, A. Anti-oxidant, antimicrobial and antiinflammatory effects of selected plants on wound healing. World J. Pharm. Pharm. Sci. 2014, 3, 1–11. [Google Scholar]
  130. Shafie, N.M.; Shah, R.N.I.R.S.; Krishnan, P.; Haleem, N.A.; Tan, T.Y.C. Scoping Review: Evaluation of Moringa oleifera (Lam.) for Potential Wound Healing in In Vivo Studies. Molecules 2022, 27, 5541. [Google Scholar] [CrossRef] [PubMed]
  131. Nowak, A.; Zielonka-Brzezicka, J.; Perużyńska, M.; Klimowicz, A. Epilobium angustifolium L. as a Potential Herbal Component of Topical Products for Skin Care and Treatment-A Review. Molecules 2022, 27, 3536. [Google Scholar] [CrossRef] [PubMed]
  132. Ryall, C.; Duarah, S.; Chen, S.; Yu, H.; Wen, J. Advancements in Skin Delivery of Natural Bioactive Products for Wound Management: A Brief Review of Two Decades. Pharmaceutics 2022, 14, 1072. [Google Scholar] [CrossRef] [PubMed]
  133. Hang, S.; Chen, H.; Wu, W.; Wang, S.; Fang, Y.; Sheng, R.; Tu, Q.; Guo, R. Progress in Isoindolone Alkaloid Derivatives from Marine Microorganism: Pharmacology, Preparation, and Mechanism. Mar. Drugs. 2022, 20, 405. [Google Scholar] [CrossRef] [PubMed]
  134. De Sá, J.D.M.; Kumla, D.; Dethoup, T.; Kijjoa, A. Bioactive Compounds from Terrestrial and Marine-Derived Fungi of the Genus Neosartorya. Molecules 2022, 27, 2351. [Google Scholar] [CrossRef]
  135. Wibowo, J.T.; Ahmadi, P.; Rahmawati, S.I.; Bayu, A.; Putra, M.Y.; Kijjoa, A. Marine-Derived Indole Alkaloids and Their Biological and Pharmacological Activities. Mar. Drugs 2021, 20, 3. [Google Scholar] [CrossRef]
  136. Zhang, D.; Li, S.; Fan, M.; Zhao, C. The Novel Compounds with Biological Activity Derived from Soil Fungi in the Past Decade. Drug Des. Devel. Ther. 2022, 16, 3493–3555. [Google Scholar] [CrossRef]
  137. Chukwujekwu, J.C.; Coombes, P.H.; Mulholland, D.A.; van Staden, J. Emodin, an antibacterial anthraquinone from the roots of Cassia occidentalis. S. Afr. J. Bot. 2006, 72, 295–297. [Google Scholar] [CrossRef] [Green Version]
  138. Xavier, M.R.; Santos, M.M.S.; Queiroz, M.G.; de Lima Silva, M.S.; Goes, A.J.S.; De Morais, M.A., Jr. Lawsone, a 2-hydroxy-1,4-naphthoquinone from Lawsonia inermis (henna), produces mitochondrial dysfunctions and triggers mitophagy in Saccharomyces cerevisiae. Mol. Biol. Rep. 2020, 47, 1173–1185. [Google Scholar] [CrossRef] [PubMed]
Scheme 1. Various stages of wound healing.
Scheme 1. Various stages of wound healing.
Processes 11 00030 sch001
Figure 1. Chemical structures of surfactin A (1), BS glycolipid (2), rhamnolipid (3), and sophorolipid (4).
Figure 1. Chemical structures of surfactin A (1), BS glycolipid (2), rhamnolipid (3), and sophorolipid (4).
Processes 11 00030 g001
Figure 2. Chemical structures of baicalin (5), kaempferol (6), and quercetin (7).
Figure 2. Chemical structures of baicalin (5), kaempferol (6), and quercetin (7).
Processes 11 00030 g002
Figure 3. Chemical structures of MT81 (8), emodin (9), and hennotanic acid (10).
Figure 3. Chemical structures of MT81 (8), emodin (9), and hennotanic acid (10).
Processes 11 00030 g003
Figure 4. Chemical structures of chlorogenic acid (11) and gallic acid (12).
Figure 4. Chemical structures of chlorogenic acid (11) and gallic acid (12).
Processes 11 00030 g004
Figure 5. Chemical structures of acremonamide (13), chondramide A (14), jasplakinolide (15), and apratyramide (16).
Figure 5. Chemical structures of acremonamide (13), chondramide A (14), jasplakinolide (15), and apratyramide (16).
Processes 11 00030 g005
Figure 6. Chemical structures of asiaticoside (17).
Figure 6. Chemical structures of asiaticoside (17).
Processes 11 00030 g006
Figure 7. Chemical structures of lipoteichoic acid (18), lactic acid (19), acetic acid (20), and diacetyl (21).
Figure 7. Chemical structures of lipoteichoic acid (18), lactic acid (19), acetic acid (20), and diacetyl (21).
Processes 11 00030 g007
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hillman, P.F.; Lee, C.; Nam, S.-J. Microbial Natural Products with Wound-Healing Properties. Processes 2023, 11, 30. https://doi.org/10.3390/pr11010030

AMA Style

Hillman PF, Lee C, Nam S-J. Microbial Natural Products with Wound-Healing Properties. Processes. 2023; 11(1):30. https://doi.org/10.3390/pr11010030

Chicago/Turabian Style

Hillman, Prima F., Chaeyoung Lee, and Sang-Jip Nam. 2023. "Microbial Natural Products with Wound-Healing Properties" Processes 11, no. 1: 30. https://doi.org/10.3390/pr11010030

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