Next Article in Journal
Effect of Copolymer Properties on the Phase Behavior of Ibuprofen–PLA/PLGA Mixtures
Next Article in Special Issue
Natural Products as New Approaches for Treating Bladder Cancer: From Traditional Medicine to Novel Drug Discovery
Previous Article in Journal
Therapeutic Efficacy of a Formulation Prepared with Linum usitatissimum L., Plantago ovata Forssk., and Honey on Uncomplicated Pelvic Inflammatory Disease Analyzed with Machine Learning Techniques
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Advances in Using Natural Antibacterial Additives in Bioactive Wound Dressings

1
Department of Chemistry and Biotechnology, School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Hawthorn, VIC 3122, Australia
2
ARC Training Centre Training Centre in Surface Engineering for Advanced Materials (SEAM), School of Engineering, Swinburne University of Technology, Hawthorn, VIC 3122, Australia
*
Authors to whom correspondence should be addressed.
Pharmaceutics 2023, 15(2), 644; https://doi.org/10.3390/pharmaceutics15020644
Submission received: 10 January 2023 / Revised: 7 February 2023 / Accepted: 9 February 2023 / Published: 14 February 2023
(This article belongs to the Special Issue Biomedical Applications of Natural Plant Extract)

Abstract

:
Wound care is a global health issue with a financial burden of up to US $96.8 billion annually in the USA alone. Chronic non-healing wounds which show delayed and incomplete healing are especially problematic. Although there are more than 3000 dressing types in the wound management market, new developments in more efficient wound dressings will require innovative approaches such as embedding antibacterial additives into wound-dressing materials. The lack of novel antibacterial agents and the misuse of current antibiotics have caused an increase in antimicrobial resistance (AMR) which is estimated to cause 10 million deaths by 2050 worldwide. These ongoing challenges clearly indicate an urgent need for developing new antibacterial additives in wound dressings targeting microbial pathogens. Natural products and their derivatives have long been a significant source of pharmaceuticals against AMR. Scrutinising the data of newly approved drugs has identified plants as one of the biggest and most important sources in the development of novel antibacterial drugs. Some of the plant-based antibacterial additives, such as essential oils and plant extracts, have been previously used in wound dressings; however, there is another source of plant-derived antibacterial additives, i.e., those produced by symbiotic endophytic fungi, that show great potential in wound dressing applications. Endophytes represent a novel, natural, and sustainable source of bioactive compounds for therapeutic applications, including as efficient antibacterial additives for chronic wound dressings. This review examines and appraises recent developments in bioactive wound dressings that incorporate natural products as antibacterial agents as well as advances in endophyte research that show great potential in treating chronic wounds.

Graphical Abstract

1. Introduction

Wound care is a global health issue. A retrospective analysis of the Medicare dataset for 2014 in the USA indicated that 8.2 million Medicare beneficiaries had at least one type of wound or related infection [1], indicating that the financial burden equates to between US $28.1 billion to US $96.8 billion, including the cost of infection management [2].
Wound repair mechanisms consist of four main phases including haemostasis, inflammation, proliferation, and dermal remodelling. In the haemostasis phase, a blood clot is formed to prevent exsanguination from vascular damage. In this step, platelet receptors interact with extracellular matrix proteins to promote adherence to the blood vessel wall. The second phase of wound healing is inflammation which is the primary defence against pathogenic wound invasion, followed by proliferation as the third phase. In this phase, activation of keratinocytes, fibroblasts, macrophages, and endothelial cells will help the process of wound closure, matrix deposition, and angiogenesis. Finally, in the matrix remodelling phase, a fibrin clot is deposited leading to the formation of a scar. For more information, readers can refer to review papers by Wilkinson et al. [3] and Carr et al. [4], scrutinising various mechanisms of wound healing.
There are two classes of wounds—acute and chronic. Acute wounds are injuries to the skin which are healed by the normal process of wound repair [5]. Wounds that have not progressed through the normal repair process and remain unhealed for an extended period are referred to as chronic wounds. The latter type of wound is a burden to the healthcare system [6], to the extent that 2% of the national health expenditure in Australia is spent on these types of wounds, equivalent to more than AUD 3.5 billion annually [7].
The delayed and incomplete healing process of chronic non-healing wounds exposes patients to a high risk of infection. Thus, for severe and chronic wounds, more advanced treatments and wound dressings should be applied to assist with accelerating wound healing [8] and preventing infection [9].
Among the more than 3000 wound dressing types on the wound management market, different characteristics can be achieved based on the intrinsic properties of the polymers used in wound dressing preparation. These characteristics include their ability to absorb exudate, combat infection, relieve pain, promote autolytic debridement, or even provide and maintain a moist environment at the wound surface. However, there is no wound dressing that possesses all these properties. The type of wound dressing is selected based on the patient’s health status, wound type, location, depth, amount of exudate, wound adhesion, and economic considerations [10,11]. Hydrogels, foams, dermal patches, films, nanoparticles, hydrocolloids, nanofibers, and membranes are the main groups of dressings, and their description, characteristics, and polymers used to make them are summarised in Table 1 [10].
Studies show that the challenges of wound dressings linked to wound infection are significant. In most acute and chronic infections, a mixed population of both aerobic and anaerobic microorganisms is observed [62] and yet to be eliminated. This challenge emphasises the importance of strategies that target the most common bacteria on the wound surface. Data from recent studies on various wound infections (e.g., surgical incisions, burns, abscesses, and traumatic wounds) confirm the presence of Pseudomonas sp. and Staphylococcus aureus as the most common Gram-negative and Gram-positive bacteria, respectively, on wound surfaces with a share of 58.4% for Gram-negative and 41.6% for Gram-positive bacteria [63]. The percentage of each group of bacteria can be observed in detail in Figure 1.
There is a significant need for antibacterial wound dressings for controlling and reducing bacterial infections. One of the best approaches is wound dressings containing antibiotics as an additive. Numerous reports can be found on wound dressings containing antibiotic additives and their effects on wound dressings [64,65,66].
By using an efficient amount of antibiotics, suitable treatment of wound infections can be achieved. However, high amounts of antibiotics will cause systemic toxicity [67]. In order to overcome these detrimental effects, the antibacterial compounds and antibiotics are embedded in wound dressings for sustained and controlled drug release [10]. A lack of new antibiotics and antibacterial agents, as well as widespread distribution and misuse of these antibacterial compounds, has caused an increase in antimicrobial resistance (AMR). It has been proposed that AMR has the potential to kill ten million lives by 2050 worldwide, costing an estimated US $100 trillion [68]. This can result in the return to a pre-antibiotic era, with infections caused by multiple-resistant pathogens [31]. Thus, there is an urgent need for sustainable novel antibacterial additives to overcome this major clinical problem.
Looking at the newly approved drugs between 1981 and 2019, followed by the share of each source in previous researched studies, indicates that the share of natural or naturally inspired approved drugs has increased over time [69,70]. One of the most important sources of these novel pharmaceuticals is plants, which form a large portion of these newly approved drugs. Botanical-based natural antibacterial compounds have attracted great attention in recent years, indicating their great potential to be used as antibacterial additives in different medical applications including wound dressings.
There has always been a significant need for novel antibacterial additives to improve wound dressing characteristics. After investigating the importance of botanical-based antibacterial compounds as one of the most important sources of pharmaceuticals, the efficiency of the resultant developed dressings using these additives has been scrutinised in each of the main groups of polymer wound dressings. The focus of this review is on the dressings containing additives against targeted bacteria, including P. aeruginosa, S. aureus, Escherichia coli, and other wound-infecting bacteria.

2. Bioactive Wound Dressings (Polymer + Additives)

2.1. Hydrogels

Hydrogel wound dressings containing natural antibacterial bio-additives have been used extensively for wound treatments. These additives are able to optimise the antibacterial properties against the unfavourable increase of bacterial proliferation in hydrogel wound dressings [11,71].
Plant-based antibacterial additives have been used in hydrogels to increase their activity against bacteria in infected wounds [71]. One of the most important class of additives in this group are the essential oils. The presence of essential oils as hydrophobic compounds in the hydrogel texture leads to good mechanical properties, degradability, improvement of the porous structure, and antioxidant properties [72]. Altaf et al. used a solution casting method to produce a polyvinyl alcohol/starch hydrogel membrane containing various concentrations of clove essential oil. The products resulted in excellent antibacterial activity, with a minimum inhibition zone of 34 ± 0.42 mm against S. aureus and 31 mm against E. coli [71]. The synthesis scheme has been demonstrated in Figure 2. Other essential oils used in hydrogels are lavender and tea tree oil [73]. Using these two essential oils in gellan gum hydrogels at 25% w/w resulted in an efficient zone of inhibition of 20 mm against S. aureus and 30 mm against E. coli in standard disc diffusion assays [73]. Several studies have used different essential oils in hydrogels, such as basil oil [74], tea tree oil [75], sweet fennel oil [76], rosemary essential oil, orange essential oil [77], and Thymus daenesis oil [78], to improve their antibacterial activity.
In addition to essential oils, plant extracts have been used as additives in hydrogel wound dressings. In a study by Shukla et al., a bioactive hydrogel dressing containing an ethanolic extract of Morus alba leaves was used against diabetic wounds. The apigenin derived from the extract was tailored with gellan gum-poly ethylene glycol-chitosan hydrogels and screened in vivo for its effectiveness. The results indicated that the apigenin additives caused effective stimulation of wound contraction and increase in the collagen content in diabetic as well as normal wound tissues, which leads to an accelerated wound healing process [79]. The antibacterial activity of Morus alba extracts against S. aureus has been previously investigated, resulting in a minimum inhibitory concentration (MIC) of 250 µg/mL [80].

2.2. Hydrocolloids

Hydrocolloids have been previously used along with natural antibacterial additives to improve their characteristics against wound bacteria and reduce the unpleasant odour [81]. These additives are the extracts of some pre-approved antibacterial plants, such as Centella asiatica (CA) and Phellodendri amurensis (PA) [82,83], which have been used in different studies against several bacteria. After loading CA plant extracts in alginate hydrocolloids using a hot melting method, Jin et al. showed excellent swelling, drug release, and mechanical properties compared with similar commercial products. Enhanced healing process in excision, infection, and abrasion wounds were observed in a rat wound model, which suggests that this extract is a potential candidate for the treatment of various wounds [82]. The preparation technique has been demonstrated in Figure 3. Antibacterial activity tests of the CA extracts at 100 µg/mL against P. aeruginosa, S. aureus, and E. coli resulted in zones of inhibition between 28–30 mm [83].
Another application of hydrocolloids containing CA extracts is skin treatment. Kuo et al. produced an anti-acne patch with gelatin/chitosan (GC) bilayer hydrocolloid patches. This anti-acne bilayer patch was loaded with Cortex PA and CA extracts. The results indicated that CA could reduce scar formation and improve the wound healing process. Water retention rate, weight loss rate, antibacterial activity, and in vitro cytotoxicity were tested as well. The results indicated that skin fibroblast cell viability was accelerated and the water retention of the patches was improved, which contributed to the exudate absorption [84].

2.3. Foams

Foams are another group of polymer wound dressings that have been previously used with additives to accelerate wound recovery. There are some reports on using plant-derived extracts as antibacterial agents in foam-based dressings. Nantaporn et al. prepared polyurethane foam sheets containing silver and asiaticoside (AS) (an extract derived from Centella asiatica plant) for healing dermal wounds. AS in a foam formulation played an essential role to increase the healing rate. The MIC of the additives against P. aeruginosa, S. aureus, E. coli, and B. subtilis were in a range of 0.4–3.1 ppm. However, the foam dressing released 4–5 ppm of the additive. The clear zones from disc diffusion assays were statistically larger than other tested formulations [21]. AS has been proved to be efficiently mixed with other polymers in different studies. Phaechamud et al. developed an absorbent chitosan-based dressing containing silver and asiaticoside as an additive. This dressing showed a successful controlled drug release along with angiogenic activity, indicating the potential to be further utilised as absorbents in medical wound dressings [85]. In what follows, the scheme of the preparation technique has been demonstrated in Figure 4.
The other group of natural plant-based antibacterial additives used in foams is essential oils. The antibacterial activity of plant essential oils such as oregano and thyme has been proven previously, with MIC values of 0.0781 µL/mL [86] and 0.125 mg/mL [87], respectively. Adding these oils to a natural polymer such as sweet potato starch-based foam, along with their antibacterial activity, may lead to a lower degradation under the thermoforming temperature and higher mechanical resistance [88].

2.4. Films

Films have previously been used as bioactive wound dressings [10]. These types of wound dressings have been used with both plant extracts and essential oils. Some studies have shown the utilisation of different plants and plant extracts in film dressings. These plants are normally chosen based on their healing and antibacterial properties. Koga et al. developed an alginate film containing Aloe vera (Aloe barbadensis Miller) gel [89]. Aloe vera has already exhibited several pharmaceutical activities, such as the ability to promote the healing process as well as the ability to stimulate the proliferation of fibroblasts [90]. After characterising the different aspects of films containing Aloe vera, the results indicated adequate transparency, uniformity, mechanical tensile strength, and hydration capacity, which makes them an ideal candidate to be used as dressings. Furthermore, the films modulated the inflammatory phase, increased angiogenesis, and stimulated collagenesis, which leads to improved healing [89]. Figure 5 demonstrates the preparation process for these types of film.
The second group of additives used in film wound dressings are essential oils. Several types of essential oils have been used as an additive to optimise the antibacterial properties of film dressings. Clove, cinnamon, chamomile, thymol, lavender, tea tree, peppermint, Eucalyptus globulus juvenile, lemongrass, and lemon are some of the essential oils that have been used as antibacterial additives [91,92,93,94,95].
A combination of gelatin with clove essential oil (CEO) and hydrotalcite (HT) nanoparticles was prepared by Guilherme et al. as a wound dressing. In this study, CEO-containing films exhibited bactericidal activity against S. aureus and E. coli. HT was also hypothesised to relate positively to the antimicrobial performance of using films and enhance physical properties, which was lowered by the CEO [91].
One of the challenges in preparing films containing essential oils is choosing the proper oil to be used in the process. In this context, comparisons have been made between using each type of essential oil in a wound dressing environment. Liakos et al. used various types of essential oils such as lavender, tea tree, peppermint, Elicriso italic, cinnamon, Eucalyptus globulus, lemon, and lemongrass as an additive in sodium alginate matrixes. The produced films were tested for their antibacterial and anti-fungal properties. Among all the samples tested against E. coli, the cinnamon essential oils showed the largest inhibition zone of 12 mm, followed by lemongrass essential oil with an inhibition zone of 3 mm. The results of the antibacterial tests along with their stability indicates that films containing essential oils have the potential to be used as antibacterial wound-dressing materials [93].

2.5. Dermal Patches

The drugs used in these types of wound dressings should be penetrable to the skin, which makes most drugs unsuitable in this application. Solubility and diffusivity are two factors that determine the maximum skin penetration flux [96]. Some botanical-based additives have been used to improve the characteristics of dermal patches used in skin care and the prevention of mosquito bites [97]. In a study by Sroczyk et al., a polyimide patch was loaded with blackcurrant seed oil for atopic skin hydration studies. The application of these patches was against atopic dermatitis as a common disease among children. In this disease, gamma-linoleic acid is decreased, so the blackcurrant seed oil was used to restore the gamma-linoleic acid deficiencies. Based on the results, these patches adjust to skin movements, are stable with plant oils, and exchange air due to their high permeability, which makes them a good candidate to be used in skin care and treatment [97]. The process scheme has been demonstrated in Figure 6. There are different types of botanical-based oils with a high level of gamma-linoleic acid that can be used as additives instead of blackcurrant seed oil, such as Nigella sativa [98], borage [99], hempseed [100], and evening primrose [101].
As previously mentioned, another application of botanical-based skin patches is in the prevention of mosquito bites. In this case, essential oils as additives in patches act as insect repellents. Chattopadhyay et al. developed a patch from an optimised mixture of cinnamon, lemongrass, and eucalyptus essential oils embedded into ethylcellulose and polyvinylpyrrolidone polymer patches. These patches were shown to be safe and effective and to contain good physico-chemical properties at room temperature. The additives in this case are not only environmentally friendly but also make the patch more effective than the previous synthetic commercial products by providing complete protection for a longer time [102].

2.6. Fibers and Nanofibers-Based Electrospun Polymers

Bioactive agents added during nanofiber production have been shown to improve the wound healing process [10]. There are several strategies to tailor bioactive additives into the fibres, including emulsion electrospinning, blend electrospinning, co-axial electrospinning, and surface immobilization [103].
There are several studies indicating the use of natural botanical-based bio-additives such as plant extracts and essential oils in electrospun polymer wound dressings.
Plant extracts have been added to the polymer electrospun fibres based on the final properties required for the wound dressing. Numerous types of plant extracts have been used as an additive to nanofibers such as Azadirachta Indica [104], tumeric [105], Clerodendrum phlomidis [106], Gymnema sylvestre [107], Carica papaya [108], Aloe vera [109], Lawsonia inermis [110], Garcinia mangostana [111], mucilage [112], clove [113], Ataria multiflora [114], pomegranate [115], Achillea lyconica [116], corn [117], fenugreek [118], henna [119], and chamomile [120].
These extracts have been proved to be effective in diabetic wound dressings. In a study by Ranjbar-Mohammadi et al., curcumin extracted from turmeric was used as an antibacterial additive in polycaprolactone electrospun fibres. The experiments indicated that the wound dressing was active for the treatment of diabetic wounds. Exhibiting an MIC of 62.5 µg/mL against P. aeruginosa [121], curcumin showed a more accelerated wound healing process in comparison with the blank sample [105]. Another application of nanofibers containing plant extracts is skin tissue engineering. Henna leaf extract-loaded chitosan-based nanofibrous mats were used as a wound dressing by Yousefi et al. The final product displayed efficient antibacterial activity due to Lawsonia inermis (Henna) leaf extracts in mats (2 wt%), with zones of inhibition against S. aureus and E. coli of 18 mm and 25 mm, respectively. The presence of henna extract caused a reduction in the fibre diameter of the mats, which makes it favourable for wound healing applications due to increasing the surface area. Furthermore, the combined advantageous features including high biocompatibility, synergistic antibacterial activity, and acceleration of wound healing can be observed by using this additive in a mixture with polymer nanofibers [119].
The next group of botanical-based additives used in nanofiber polymer wound dressings is essential oils. Different types of essential oils have previously been used as additives in a mixture with polymer nanofibers targeting wound bacteria. These plants include lavender oil [122], thyme oil [123], cinnamon oil [124], and rosemary/oregano oil [125] that have shown antibacterial activity against the most common wound bacteria such as S. aureus, E. coli, and P. aeruginosa [122,123,124,125,126].
An improved wound healing device using encapsulation of cerium oxide (CeO2) and peppermint oil (PM oil) on polyethylene oxide/graphene oxide (PEO/GO) electrospun polymeric mats was shown by Suganya et al. This study involved testing against Gram-positive bacteria (S. aureus) and Gram-negative bacteria (E. coli) and evaluated in vitro cytotoxicity. The results indicated that the CeO2-PM oil-PEO/GO nanofibrous mats were less toxic to the L929 fibroblast cells. Furthermore, evaluations demonstrated that the incorporation of the plant-based bioactive agent and CeO2 in a nanofibrous mat accelerates re-epithelialization and collagen deposition, which makes the system an efficient potential candidate to be applied as wound dressings with skin infections [127]. The MIC values for peppermint essential oils are 3.1 µL/mL and 6.3 µL/mL against S. aureus and E. coli, respectively [128]. In what follows, the preparation technique of CeO2-PM oil-PEO/GO nanofibrous mats is demonstrated in Figure 7.

2.7. Membranes

Another group of wound dressings that have been used in combination with plant-based natural additives are membranes. Both essential oils and plant extracts have shown the ability to optimise the characteristics of the final dressings. Egri et al. developed Hypericum perforatum oil-loaded polycaprolactone membranes to be used in wound dressing applications. After investigating the mechanical strength and antibacterial activity, the product exhibited sufficient elasticity and activity against S. aureus and E. coli, with inhibition zones of 8–13 mm and 10–12.2 mm, respectively. Not having the risk of adhering to the wound surface, not having apoptotic/necrotic effects, being biocompatible, and having a proliferative effect on cells are some of the advantageous features of the Hyperium perforatum-loaded membranes [129]. The preparation scheme of this membrane is demonstrated in Figure 8.
Another type of essential oil used in membranes is Artemisia argyi. The efficiency of this essential oil has previously been investigated against wound bacteria such as S. aureus, P. aeruginosa and E. coli, with MIC values of 16 µg/mL, 64 µg/mL, and 32 µg/mL, respectively [130]. Ting-Ting et al. fabricated Artemisia argyi oil-microcapsule (AAO-MC)/PVC fibrous membrane wound dressings and showed that the production process was enhanced using emulsification-internal gelation. The results showed excellent stability and a slow release of the oil. Furthermore, the produced membrane showed good water vapor transmission and high hydrophilicity as well as an excellent antibacterial rate of 94.3%, which is calculated by the difference between the colony counts of the blank specimen and the colony counts of culture medium that has been cultured with a bacterial solution for a specified time divided by the colony counts of the blank specimen [131].
Based on the targeted bacteria and the final characteristics, other types of essential oils may be used as additives, such as cabreuva (Myrocarpus fastigiatus) [132] and oregano [133]. The MIC values of pure oregano essential oil have been determined to be 0.25 mg/mL, 0.64 mg/mL, and 0.16 mg/mL against E. coli, P. aeruginosa, and S. aureus, respectively [134,135].
The addition of cabreuva essential oil to poly (vinyl alcohol) membranes proves its effectiveness against S. aureus. Its capacity to produce cell regeneration along with no detectable toxicity makes it a suitable dressing for superficial burns or minor wounds [132]. Oregano essential oils have been used with poly (L-lactide-co-caprolactone)/silk fibroin membranes as shown by Khan et al., showing a highly active membrane against both Gram-negative (E. coli) and Gram-positive (S. aureus) bacteria. The results indicated an accelerated healing process, boosted granulation, and also re-epithelialization, which confirms its potential to be used as a wound dressing [133].

2.8. Polymer-Drug Conjugates

Linkers used for the conjugation of drugs to polymers function to control the drug release in a pH specific manner and in the presence of enzymes depending on the chemistry of the linker employed [136]. For improving the therapeutic advantages of this type of wound dressing, moiety and solubilising units are also incorporated into polymer–drug conjugates [137,138]. Several studies indicate the use of plant extracts and essential oils conjugated with polymers. Some of the essential oils that have previously been used in polymer nanocarriers are thyme [139,140], peppermint oil [141], green tea oil [141], etc.
In a study by Shetta et al., peppermint and green tea essential oils were encapsulated into chitosan nanoparticles using the emulsification/ionic gelation method. The final product was tested against S. aureus and E. coli, showing minimum bactericidal concentration (MBC) values of 1.11 mg/mL and >2.72 mg/mL for peppermint oil and 0.57 mg/mL and 1.15 mg/mL for green tea, respectively, demonstrating their potential to be used in wound dressing applications [141]. Figure 9 demonstrates the preparation steps of this product.
Another group of botanical-based antibacterial additives with the potential to be conjugated with polymers are plant extracts. Some of the utilised plant extracts conjugated with polymer wound dressings are polyphenolics and hydrolysable tannins from Hamamelis virginiana [142], seaweed extract [143], Mcrotyloma uniflorum [144], Aloe vera [145], and curcumin [146].
In a study by Yang et al., gallic acid was conjugated to a 2-hydroxy (ethyl methacrylate-co-2-diethylamino) methacrylate hydrogel. Gallic acid used in this study was extracted from an Indian plant called Terminalia bellinca, showing antioxidant and cytoprotective characteristics. The multifunctional hydrogel was used as a carrier for cell therapy and drug delivery applications. The results indicated that the product caused a faster recovery in affected tissues, which shows their significant potential to be used in medical applications [147].

2.9. Other Polymer Wound Dressings

Other types of polymer wound dressings including 3D-printed scaffolds, emulgels, and nanoemulgels have been used with various plant-based antibacterial additives previously. There are several studies indicating the use of essential oils and plant extracts in these types of wound dressings.
In a study by Ilhan et al., Satureja cuneifolia plant extracts were blended with sodium alginate and polyethylene 3D-printed scaffolds for treating diabetic ulcers. Disc diffusion testings against S. aureus demonstrated that the samples containing Satureja cuneifolia extracts (between 0.5 to 2 wt%) have an inhibition zone of 12–13 mm, which indicates their remarkable activity against Gram-positive bacteria. However, their activity against E. coli was reported to be in much higher concentrations (700 µg/mL) [148].
Emulgels and nanoemulgels have been used extensively with plant extracts and essential oils as an additive. Ocimum basilicum extracts [149], clove oil [150], rosemary oil [151], and piper betle oil [152] are some of these additives.
In a study by Razdan et al., clove oil-based nanoemulgels were used as a burn wound dressing. Levofloxacin nanoemulgels were combined with clove oil and were examined in vivo against P. aeruginosa biofilm-infected burn wounds. The product was tested against mice and the wound closure state was observed on the 1st, 3rd, 7th, 10th, and 15th day. The results indicated a faster reduction in wound size and a complete wound closure after 15 days in comparison with the samples without the additive, which were not completely closed in that period [150].
As mentioned before, one of the ways to improve wound dressing characteristics is to include bioactive additives. The role of natural antibacterial additives in polymer wound dressing groups were summarised before. In the following, different groups of plant-based natural products, as the source of novel antibacterial additives against the most common wound bacteria (S. aureus, E. coli, and P. aeruginosa), are discussed [69].

3. Plant-Based Bio-Additives as Novel Antibacterial and Antimicrobial Additives to the Polymer Wound Dressings

Two thirds of new antibacterial therapies [69], as well as several antibacterials currently in clinical trials, are natural products. The efficacy of these products is likely the result of their evolutionary process to be bioactive, providing organisms a selective advantage in the environment. [153].
Plant-based natural resources are promising antibacterial candidates for wound treatments. There are different types of plant-based antibacterial and antimicrobial agents including plant extracts, essential oils (EOs), and endophytes [31]. In what follows, a description and rationale of choosing each of these groups is discussed. Moreover, some examples in different types of each group along with their activity against targeted wound infection bacteria are demonstrated.

3.1. Plant Extracts

Plant extracts have been used against specific biological targets or related diseases. There are several extraction methods including cutting, chopping, macerating, and grinding raw or dried plant material followed by adding at least one solvent. Based on the requested final product, the ratio of the extracted material amount (kg) and the used volume (L) of the solvent may be different. Some of the other factors controlling the characteristics of the final products are solvent type (alcohols, oils, or water), the solvent temperature used in the process of extraction, and the time of extraction (between 1 h to 120 h). A pharmaceutically accepted excipient such as cellulose derivatives (as diluents), gelatin (as a binder), carbohydrates (as fillers), phosphate-buffered saline (as a buffering agent), polyvinylpyrrolidone (as a dispersion enhancer), and silica (as a lubricant) can be added to the embodiment to improve its formulation properties [31].
Indigenous plants are one of the most important sources of these antibacterial additives. The valuable information about their ethnobotanical use is gained from the local population’s knowledge. This knowledge can be utilised to transform these traditional medicines into clinical applications. There are some patents to protect these discoveries based on their specificity, habitat, and composition [31].
There are many plants that have been investigated for their antibacterial activity against pathogens. Most of these plants have already been used in local folk medicine for various applications. For example, Roja et al, investigated the potential inhibitory effect of methanol leaf extracts of Acalipha alinifolia (AA), Delonix elata (DE), Digera muricate (DM), Hygrophilia auriculate (HA), Jatropha gasipifed (JG), Maeua oblongifolia (MO), Pterocarpus santalinus (PS), Punica granatum (PG), Syzygium cumini (SC), Gyrocaspus americana (GA), and Euphorbia heterophilla (EH) on bacterial isolates of septic wound infections. Each one of these plants has been used in local folk medicine. The results indicated that PG and SC have potential antibacterial activity against the predominant isolates from septic wounds including P. aeruginosa, S. aureus, Klebsiella pneumoniae, and E. coli [154].
Azizah et al. studied the antibacterial activities of E. glabra against S. aureus and S. epidermidis. The bioactive compounds were extracted via solvent extraction and tested against selected bacteria via screening using agar diffusion methods. The results indicated activity against both bacteria, with MIC values between 32–512 µg/mL [155]. In another study, the antibacterial constituents from the indigenous Australian medicinal plant Eremophila duttonii F. Muel were investigated by Joshua et al. The bioactive compounds were extracted using solvent extraction with hexane, dichloromethane, and ethanol. All the compounds showed appreciable activity against Gram-positive organisms, including S. aureus, S. epidermidis, and Streptococcus pneumoniae [156].
In Table 2, the common medical uses of some of these plants and the chemical class of major compounds are shown for more insight into the rationale for the utilisation of these sources.

3.2. Essential Oils

EO fractions are the carrier of the fragrance of plants. These secondary metabolite oils include a large number of compounds based on an isoprene structure called terpenes. Having the chemical backbone of C10H16, they exist as diterpenes, triterpenes, tetraterpenes, and hemiterpenes as well as sesquiterpenes.
Terpenoids are terpenes that contain additional functional groups. Basically, essential oils are terpenoid compounds [221]. They are synthesised from acetate units and share their origins with fatty acids. Due to their extensive branching and cyclization, they differ from fatty acids [222]. Essential oils have been extensively studied due to their inhibitory activity against pathogens [31]. Terpenes or terpenoids are active against bacteria [223], fungi [224], viruses [225], and protozoa [226]. One of the examples of this activity is triterpenoid betulinic acid, which has been shown to inhibit HIV. The mechanism of action of terpenes is not fully understood to date but it has been speculated to involve membrane disruption caused by these lipophilic compounds [222]. The processing unit of essential oils have been demonstrated in Figure 10 [227].
A skin lotion was prepared using an antibacterial essential oil containing a mixture of Camellia japonica L. oil with simple volatile aromatic compounds extracted from Juniperus chinesis L. and Aquilaria agallocha Lam. The steam distillation process was performed at a temperature ranging from 65 to 75 °C for 45 to 50 h [228]. The use of antibacterial essential oils caused the removal of adolescent acne and prevented skin aging. After testing the effectiveness of various essential oils based on the aforementioned substances on a cohort of 100 people, the results indicated that the essential oils were reported as moderate or high in antibacterial, antioxidant, and skin moisturization characteristics as well as acne reduction [31].
Mixing different essential oils is one of the ways to optimise the characteristics of the final product. Essential oil mixtures are able to show activity against numerous strains of bacteria (such as E. coli, S. aureus, and Shigella, or Shiga toxin-producing E. coli), viable but not-culturable bacteria, bacterial spores, helminth, protozoan, fungus, or virus. [31].
Another technique that can be used for achieving more efficient antibacterial activity in essential oils is nanoemulsification. Lida et al. prepared five different nano-emulsions from Lavandula angustifolia, Rosmarinus officinalis, and Satureja khuzistanica essential oils (SKEO) as well as two EO constituents (carvacrol and 1,8-cineol). After characterisation, the formulations demonstrated long-term stability. The nanoemulsification of the essential oils caused a more efficient antibacterial activity against P. aeruginosa. The MIC for all the crude essential oils was 64 mg/mL. However, the nano-emulsion compound of Satureja khuzistanica and carvacrol showed a MIC of 8 mg/mL. The MIC reported for the Lavandula angustifolia, rosmarius officinalis, and 1,8-cineol nano-emulsions was 16 mg/mL [229].
As mentioned in the previous section, essential oils have been widely used in wound dressings against wound pathogens. Table 3 shows examples of different antimicrobial essential oils along with their reported properties in medical applications, their activity against wound-infected bacteria, and their chemical constituents.

3.3. Endophytes: A Novel Source of Bioactive Compounds

Endophytes are defined as the microbes colonising the internal tissues of plants, which cause no immediate negative effects [279]. They have extensive biodiversity and are considered as a sustainable source for novel pharmaceutical applications. The discovery of endophytes dates back to 1904. However, these groups of microorganisms did not receive much attention. With the discovery of paclitaxel (Taxol) from the endophytic fungus Taxomyces andreanea, which has been isolated from Taxus brevifolia as an important anti-cancer drug, the attention changed dramatically [280]. The isolation of penicillin from Penicillium notatum in the 1940s by Sir Howard Florey and his team alerted the world to the significance of fungi as a novel source for bioactive compounds. Plants actively combat pathogenic attack by producing antimicrobial compounds. Screening plants for endophytic isolation has led to novel and interesting compounds [281,282]. This has subsequently directed research to consider endophytes from ethno-pharmaceutically used plants as a source of new therapeutic compounds. Due to the success of some previous medicinal drugs from microbial origins, drug discovery has been more focused on microorganisms instead of plants. Thus, it has led to the consideration of endophytic fungi as a promising rich source of natural products in the search for new drug sources [283].
Endophytes can be considered as chemical synthesisers in plants; many are responsible for synthesising bioactive compounds used as a potential source of many pharmaceutical leads. These sources have been proven to show extensive potential to be used against multi-drug resistant (MDR) microorganisms [283]. They also have been proven to be useful in novel drug discovery by the chemical diversity of their secondary metabolites, to the extent that many of them are the source of production for novel antibacterial [284,285], antiviral [286,287], antifungal [288,289], anti-inflammatory [290,291], anti-tumour [292,293], and anti-malaria [294,295] compounds. These compounds are from different chemical classes, such as alkaloids [296], terpenoids [297], flavonoids [298], phenolic compounds [299], and steroid derivatives [300]. Table 4 lists these compounds, their host plants, and their bioactivity.
Endophytes are believed to provide resistance against pathogenic attack of the host plant by producing secondary metabolites [320]. They are a great source of natural products which exhibit an extensive array of bioactivity to the extent that many of the endophytic fungi are known to produce antibacterial and antimicrobial substances. Antimicrobial metabolites are defined as the low molecular weight organic natural substances, which have been made by active microorganisms at low concentrations against other microorganisms [321]. The crude extracts from the culture broths of endophytic fungi have shown activity against pathogenic fungi, bacteria, and yeasts, cytotoxic activity on human cell lines, anti-Herpes simplex virus type 1 (anti-HSV), and malaria parasites. Different antimicrobial activities by geographically different endophytes have been studied [322]. Different natural products have been produced from endophytic fungi, such as anti-cancerous, antioxidants, antiviral, anti-insecticidal, immunosuppressant, antimicrobial, anti-malarial, and anti-mycobacterial compounds [323,324,325].
It has been reported that medicinal plants can harbor endophytes [326], which protect the host plants from infectious agents and adapts them to environmental conditions. This mechanism is enhanced by contributing to the compounds produced by the host plant that protect against biotic and abiotic stress factors [327,328,329,330,331]. Some researchers have reported that in many cases, host plant tolerance to biotic stress is related to natural products produced by endophytic fungi [332].
Different factors are responsible for the rationale for choosing the proper plant among the numerous species available. Basically, reaching a particular microbial metabolite requires a particular biotope, at both environmental and organismal levels. Plants growing in an area with great biodiversity, in a unique habitat or containing novel strategies for survival are likely to be good candidates due to their unusual biology. Thus, they are considered important for researching unusual endophytic species [333]. The second group of selected plants are the ones which are asymptomatically infected with phytopathogens. These plants are likely to have endophytes with antimicrobial features [334]. Plants with an ethnobotanical history, which have been used by Indigenous people as traditional medicines, have great potential for the discovery of novel bioactive endophytes. Endophytic Streptomyces isolated from an Australian medicinal plant, snakevine (Kennadia nigriscans), is an example of this group [335].
Many Australian native plants have a long history of being used as medicinal and culinary herbs. Some of them are even considered to be equivalent to the Mediterranean herbs. Lots of Mediterranean herbs have been investigated and their therapeutic properties have been well-documented. However, there is limited information about the use of Australian native plants in medicine [336]. Some of these plants have been investigated for their antibacterial components, such as Eremophila glabra [155], Eremophila duttonii [156], and Eremophila alternifolia [337].
The biological activity depends on the natural products that endophytes produce in the host plant [338]. Thus, research regarding this important source of bioactive compounds has resulted in potential drug compounds as antibacterial additives. In what follows, some studies of different applications of endophytes are discussed.
In a study by Xing et al., endophytic fungi from two types of orchids called Dendrobium devonianum and Dendrobium thysiflorum were isolated and identified. The extracted compounds using ethanol as solvent were tested against six pathogenic microbes (Escherichia coli, Bacillus subtilis, Staphylococcus aureus, Candida albicans, Cryptococcus neoformans, and Aspergillus fumigatus). The antimicrobial activity of the extracts was tested using the agar diffusion method with a concentration of 100 µg/disk. The results indicated that 10 and 11 endophytic fungi extracts originating from Dendrobium devonianum and Dendrobium thysiflorum, respectively, showed antimicrobial activity against at least one of the pathogenic bacteria listed above. Out of the fungal endophytes of both plants, Phoma displayed strong inhibitory activity with an inhibition zone of more than 20 mm, and Epicoccum nigrum isolated from Dendrobium devonianum showed stronger antibacterial activity than ampicillin sodium [339].
In another study, Dang et al. isolated Trichoderma ovalisporum endophytic fungi from Panax notoginseng and tested their antibacterial activity. After growing the chosen isolate in potato dextrose agar medium, samples were filtered and extracted with ethyl acetate. Finally, the crude extracted compounds were tested against Staphylococcus aureus and Escherichia coli for their antibacterial activity using disc diffusion. The results indicated a bacteria-free zone diameter of 12 mm for both strains [340]. The process of growth and extraction in endophytic fungi can be observed in Figure 11 [340].
The antimicrobial and antibacterial activities of these bioactive compounds have already been scrutinised by researchers and the same classes of chemical compounds have been used extensively in wound dressing applications in several studies. The examples below describe antibacterial compounds that have been added to wound-dressing materials. As endophytes are known to produce these compounds, it demonstrates that endophytic fungi could be an alternative and sustainable source of these valuable products.
Soares et al. developed a chitosan-based hydrogel containing flavonoids isolated from Passiflora edulis Sims for wound healing purposes in a diabetic rat model. The results demonstrated effective wound healing ability. In addition, the formulation could stimulate the antioxidant defence system, which positively influenced the treatment of skin lesions in diabetic rats, representing their potential use as dressings in wound treatment [341].
In a study, Azzazy et al. developed chitosan-coated PLGA nanoparticles loaded with Peganum harmala alkaloids for wound dressing applications. In this study, the harmala alkaloid-rich fraction loaded into PLGA nanoparticles coated with chitosan in the emulsion-solvent evaporation method was used. The results indicated that the wound closure rate was superior in comparison with the blank sample. In addition, the developed formulation demonstrated synergistic antibacterial and wound healing properties, leading to efficient wound management [342].

4. Conclusions

Antibacterial agents derived from natural products have made a considerable impact in the development of novel materials for the treatment of wounds. Plant-based compounds, including saponins, tannins, alkaloids, alkenyl phenols, glycoalkaloids, flavonoids, sesquiterpenes, lactones, terpenoids, and phorbol esters, have contributed a large portion of these antibacterial agents. Plant extracts and essential oils are reported in numerous studies as two major sources of antibacterial additives in all types of wound dressings. In this paper, we introduced an alternative promising source of antibacterial compounds, namely endophytes, which are recognised as sources of compounds with useful pharmaceutical properties. The diverse bioactive compounds extracted from endophytic fungi with antibacterial activities should be the focus of future development as a sustainable source of chemicals for wound dressing applications. To our knowledge, there is no study showing the use of the antibacterial compounds sourced from endophytic fungi in wound dressing applications. However, the abovementioned features and characteristics of bioactive compounds existing in endophytic fungi, along with the proven antibacterial characteristics of the extracts from endophytic fungi, show the great potential of using endophytes as new antibacterial additives for wound dressing applications, leading to new and effective products to combat acute and chronic wound infections. However, there are some limitations in using endophytic fungi extracts as novel antibacterial agents, including the low concentration of the active compounds in the extraction method and the lack of adequate in vivo trials. Overcoming these limitations requires further research in developing the previous methods of extraction, designing a method for purifying active extracted compounds, and in vivo studies in order to examine the products in a practical environment, which will potentially be the future steps of scrutinising these novel antibacterial agents.

Author Contributions

Conceptualization, M.F., E.A.P., P.K. and B.Z.; investigation, M.F.; writing—original draft preparation, M.F.; writing—review and editing, M.F.; visualization, M.F.; supervision, E.A.P., P.K. and B.Z.; project administration, M.F. and B.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sen, C.K. Human Wounds and Its Burden: An Updated Compendium of Estimates. Adv. Wound Care 2019, 8, 39–48. [Google Scholar] [CrossRef]
  2. Nussbaum, S.R.; Carter, M.J.; Fife, C.E.; DaVanzo, J.; Haught, R.; Nusgart, M.; Cartwright, D. An Economic Evaluation of the Impact, Cost, and Medicare Policy Implications of Chronic Nonhealing Wounds. Value Health 2018, 21, 27–32. [Google Scholar] [CrossRef]
  3. Wilkinson, H.N.; Hardman, M.J. Wound healing: Cellular mechanisms and pathological outcomes. Open Biol. 2020, 10, 200223. [Google Scholar] [CrossRef]
  4. Carr, N.J. The pathology of healing and repair. Surgery 2022, 40, 13–19. [Google Scholar] [CrossRef]
  5. Jones, V.; Grey, J.; Harding, K. Wound dressings. BMJ 2006, 332, 777. [Google Scholar] [CrossRef]
  6. Sen, C.; Roy, S.; Gordillo, G. Wound Healing (Neligan Plastic Surgery: Volume One); Elsevier: Amsterdam, The Netherlands, 2017. [Google Scholar]
  7. McCosker, L.; Tulleners, R.; Cheng, Q.; Rohmer, S.; Pacella, T.; Graves, N.; Pacella, R. Chronic wounds in Australia: A systematic review of key epidemiological and clinical parameters. Int. Wound J. 2019, 16, 84–95. [Google Scholar] [CrossRef]
  8. Dart, A.; Bhave, M.; Kingshott, P. Antimicrobial Peptide-Based Electrospun Fibers for Wound Healing Applications. Macromol. Biosci. 2019, 19, e1800488. [Google Scholar] [CrossRef]
  9. Sood, A.; Granick, M.; Tomaselli, N. Wound Dressings and Comparative Effectiveness Data. Adv. Wound Care 2014, 3, 511–529. [Google Scholar] [CrossRef]
  10. Aderibigbe, B.A. Chapter 6—Efficacy of Polymer-Based Wound Dressings in Chronic Wounds. In Modeling and Control of Drug Delivery Systems; Azar, A.T., Ed.; Academic Press: Cambridge, MA, USA, 2021; pp. 79–110. [Google Scholar]
  11. Sikka, M.P.; Midha, V.K. 16—The role of biopolymers and biodegradable polymeric dressings in managing chronic wounds. In Advanced Textiles for Wound Care, 2nd ed.; Rajendran, S., Ed.; Woodhead Publishing: Cambridge, UK, 2019; pp. 463–488. [Google Scholar]
  12. Ahmed, E.M. Hydrogel: Preparation, characterization, and applications: A review. J. Adv. Res. 2015, 6, 105–121. [Google Scholar] [CrossRef]
  13. Kamoun, E.A.; Kenawy, E.-R.; Chen, X. A review on polymeric hydrogel membranes for wound dressing applications: PVA-based hydrogel dressings. J. Adv. Res. 2017, 8, 217–233. [Google Scholar] [CrossRef]
  14. Zeng, D.; Shen, S.; Fan, D. Molecular design, synthesis strategies and recent advances of hydrogels for wound dressing applications. Chin. J. Chem. Eng. 2021, 30, 308–320. [Google Scholar] [CrossRef]
  15. Agarwal, A.; McAnulty, J.F.; Schurr, M.J.; Murphy, C.J.; Abbott, N.L. 8—Polymeric materials for chronic wound and burn dressings. In Advanced Wound Repair Therapies; Farrar, D., Ed.; Woodhead Publishing: Cambridge, UK, 2011; pp. 186–208. [Google Scholar]
  16. Leveriza-Oh, M.; Phillips, T. Chapter 32—Dressings and postoperative carea. In Lower Extremity Soft Tissue & Cutaneous Plastic Surgery, 2nd ed.; Dockery, G.D., Crawford, M.E., Eds.; W.B. Saunders: Oxford, UK, 2012; pp. 471–488. [Google Scholar]
  17. Nielsen, J.; Fogh, K. Clinical utility of foam dressings in wound management: A review. Chronic Wound Care Manag. Res. 2015, 2, 31–38. [Google Scholar]
  18. Kirwan, H.; Pignataro, R. Chapter 2—The Skin and Wound Healing. In Pathology and Intervention in Musculoskeletal Rehabilitation, 2nd ed.; Magee, D.J., Zachazewski, J.E., Quillen, W.S., Manske, R.C., Eds.; W.B. Saunders: Oxford, UK, 2016; pp. 25–62. [Google Scholar]
  19. Weller, C. 4—Interactive dressings and their role in moist wound management. In Advanced Textiles for Wound Care; Rajendran, S., Ed.; Woodhead Publishing: Cambridge, UK, 2009; pp. 97–113. [Google Scholar]
  20. Chaganti, P.; Gordon, I.; Chao, J.H.; Zehtabchi, S. A systematic review of foam dressings for partial thickness burns. Am. J. Emerg. Med. 2019, 37, 1184–1190. [Google Scholar] [CrossRef]
  21. Namviriyachote, N.; Lipipun, V.; Akkhawattanangkul, Y.; Charoonrut, P.; Ritthidej, G.C. Development of polyurethane foam dressing containing silver and asiaticoside for healing of dermal wound. Asian J. Pharm. Sci. 2019, 14, 63–77. [Google Scholar] [CrossRef]
  22. Shi, C.; Wang, C.; Liu, H.; Li, Q.; Li, R.; Zhang, Y.; Liu, Y.; Shao, Y.; Wang, J. Selection of Appropriate Wound Dressing for Various Wounds. Front. Bioeng. Biotechnol. 2020, 8, 182. [Google Scholar] [CrossRef]
  23. Peles, Z.; Zilberman, M. Novel soy protein wound dressings with controlled antibiotic release: Mechanical and physical properties. Acta Biomater. 2012, 8, 209–217. [Google Scholar] [CrossRef]
  24. Srivastava, C.M.; Purwar, R.; Kannaujia, R.; Sharma, D. Flexible silk fibroin films for wound dressing. Fibers Polym. 2015, 16, 1020–1030. [Google Scholar] [CrossRef]
  25. Pastore, M.N.; Kalia, Y.N.; Horstmann, M.; Roberts, M.S. Transdermal patches: History, development and pharmacology. Br. J. Pharmacol. 2015, 172, 2179–2209. [Google Scholar] [CrossRef]
  26. Levin, A. The Clinical Epidemiology of Cardiovascular Diseases in Chronic Kidney Disease: Clinical Epidemiology of Cardiovascular Disease in Chronic Kidney Disease Prior to Dialysis. Semin. Dial. 2003, 16, 101–105. [Google Scholar] [CrossRef]
  27. Cilurzo, F.; Musazzi, U.M.; Franzé, S.; Fedele, G.; Minghetti, P. Design of in vitro skin permeation studies according to the EMA guideline on quality of transdermal patches. Eur. J. Pharm. Sci. 2018, 125, 86–92. [Google Scholar] [CrossRef]
  28. Auda, S.H.; Mahrous, G.M.; Ibrahim, M.A.; Shazly, G.A.; Salem-Bekhit, M.M. Novel chlorhexidine dermal patches, preparation characterization and antimicrobial evaluation. Polym. Bull. 2017, 74, 3995–4007. [Google Scholar] [CrossRef]
  29. Suhaeri, M.; Noh, M.; Moon, J.; Kim, I.; Oh, S.; Ha, S.; Lee, J.; Park, K. Novel skin patch combining human fibroblast-derived matrix and ciprofloxacin for infected wound healing. Theranostics 2018, 8, 5025–5038. [Google Scholar] [CrossRef]
  30. Nilani, P.; Pranavi, A.; Duraisamy, B.; Damodaran, P.; Subhashini, V.; Elango, K. Formulation and evaluation of wound healing dermal patch. Afr. J. Pharm. Pharmacol. 2011, 5, 1252–1257. [Google Scholar]
  31. Guglielmi, P.; Pontecorvi, V.; Rotondi, G. Natural compounds and extracts as novel antimicrobial agents. Expert Opin. Ther. Pat. 2020, 30, 949–962. [Google Scholar] [CrossRef]
  32. Khil, M.S.; Cha, D.I.; Kim, H.Y.; Kim, I.S.; Bhattarai, N. Electrospun Nanofibrous Polyurethane Membrane as Wound Dressing. J. Biomed. Mater. Res. Part B Appl. Biomater. 2003, 67, 675–679. [Google Scholar] [CrossRef]
  33. Rho, K.S.; Jeong, L.; Lee, G.; Seo, B.; Park, Y.J.; Hong, S.; Roh, S.; Cho, J.J.; Park, W.H.; Min, B. Electrospinning of collagen nanofibers: Effects on the behavior of normal human keratinocytes and early-stage wound healing. Biomaterials 2006, 27, 1452–1461. [Google Scholar] [CrossRef]
  34. Ju, H.W.; Lee, O.J.; Lee, J.M.; Moon, B.M.; Park, H.J.; Park, Y.R.; Lee, M.C.; Kim, S.H.; Chao, J.R.; Ki, C.S.; et al. Wound healing effect of electrospun silk fibroin nanomatrix in burn-model. Int. J. Biol. Macromol. 2016, 85, 29–39. [Google Scholar] [CrossRef]
  35. Lin, J.; Li, C.; Zhao, Y.; Hu, J.; Zhang, L. Co-electrospun Nanofibrous Membranes of Collagen and Zein for Wound Healing. ACS Appl. Mater. Interfaces 2012, 4, 1050–1057. [Google Scholar] [CrossRef]
  36. Venugopal, J.R.; Zhang, Y.; Ramakrishna, S. In Vitro Culture of Human Dermal Fibroblasts on Electrospun Polycaprolactone Collagen Nanofibrous Membrane. Artif. Organs 2006, 30, 440–446. [Google Scholar] [CrossRef]
  37. Coelho, D.S.; Veleirinho, B.; Alberti, T.; Maestri, A.; Yunes, R.; Dias, P.F.; Maraschin, M. Electrospinning technology: Designing nanofibers toward wound healing application. In Nanomaterials-Toxicity, Human Health and Environment; Intechopen: London, UK, 2018; pp. 1–19. [Google Scholar]
  38. Shi, X.; Zhou, W.; Ma, D.; Ma, Q.; Bridges, D.; Ma, Y.; Hu, A. Electrospinning of Nanofibers and Their Applications for Energy Devices. J. Nanomater. 2015, 2015, 140716. [Google Scholar] [CrossRef]
  39. Azimi, B.; Maleki, H.; Zavagna, L.; De la Ossa, J.G.; Linari, S.; Lazzeri, A.; Danti, S. Bio-Based Electrospun Fibers for Wound Healing. J. Funct. Biomater. 2020, 11, 67. [Google Scholar] [CrossRef]
  40. Abrigo, M.; Kingshott, P.; McArthur, S. Electrospun Polystyrene Fiber Diameter Influencing Bacterial Attachment, Proliferation, and Growth. ACS Appl. Mater. Interfaces 2015, 7, 7644–7652. [Google Scholar] [CrossRef]
  41. Genevro, G.M.; Gomes Neto, R.J.; de Paulo, L.A.; Lopes, P.S.; de Moraes, M.A.; Beppu, M.M. Glucomannan asymmetric membranes for wound dressing. J. Mater. Res. 2019, 34, 481–489. [Google Scholar] [CrossRef]
  42. Sahana, T.G.; Rekha, P. Biopolymers: Applications in wound healing and skin tissue engineering. Mol. Biol. Rep. 2018, 45, 2857–2867. [Google Scholar] [CrossRef]
  43. López-Mata, M.A.; Gastelum-Cabrera, M.; Valbuena-Gregorio, E.; Zamudio-Flores, P.B.; Burruel-Ibarra, S.E.; Morales-Figueroa, G.G.; Quihui-Cota, L.; Juárez-Onofre, J.E. Physicochemical properties of novel pectin/Aloe gel membranes. Iran. Polym. J. 2018, 27, 545–553. [Google Scholar] [CrossRef]
  44. Azad, A.K.; Sermsintham, N.; Chandrkrachang, S.; Stevens, W.F. Chitosan membrane as a wound-healing dressing: Characterization and clinical application. J. Biomed. Mater. Res. Part B Appl. Biomater. 2004, 69B, 216–222. [Google Scholar] [CrossRef]
  45. Chattopadhyay, S.; Raines, R. Collagen-based biomaterials for wound healing. Biopolymers 2014, 101, 821–833. [Google Scholar] [CrossRef]
  46. Pires, A.L.R.; de Azevedo Motta, L.; Dias, A.M.A.; de Sousa, H.C.; Moraes, Â.M.; Braga, M.E.M. Towards wound dressings with improved properties: Effects of poly(dimethylsiloxane) on chitosan-alginate films loaded with thymol and beta-carotene. Mater. Sci. Eng. C 2018, 93, 595–605. [Google Scholar] [CrossRef]
  47. Bueno, C.Z.; Moraes, A.J. Development of porous lamellar chitosan-alginate membranes: Effect of different surfactants on biomaterial properties. Appl. Polym. Sci. 2011, 122, 624. [Google Scholar] [CrossRef]
  48. Boateng, J.S.; Matthews, K.H.; Stevens, H.N.E.; Eccleston, G.M. Wound Healing Dressings and Drug Delivery Systems: A Review. J. Pharm. Sci. 2008, 97, 2892–2923. [Google Scholar] [CrossRef]
  49. Kimna, C.; Tamburaci, S.; Tihminlioglu, F. Novel zein-based multilayer wound dressing membranes with controlled release of gentamicin. J. Biomed. Mater. Res. Part B Appl. Biomater. 2019, 107, 2057–2070. [Google Scholar] [CrossRef]
  50. Popa, G.-M.L.; Truşcă, R.D.; Ilie, C.-I.; Țiplea, R.E.; Ficai, D.; Oprea, O.; Stoica-Guzun, A.; Ficai, A.; Dițu, L.-M. Antibacterial Activity of Bacterial Cellulose Loaded with Bacitracin and Amoxicillin: In Vitro Studies. Molecules 2020, 25, 4069. [Google Scholar]
  51. Cheng, C.-F.; Wu, K.; Chen, Y.; Hung, S. Bacterial adhesion to antibiotic-loaded guided tissue regeneration membranes—A scanning electron microscopy study. J. Formos. Med. Assoc. 2015, 114, 35–45. [Google Scholar] [CrossRef]
  52. Lei, J.; Sun, L.; Li, P.; Zhu, C.; Lin, Z.; Mackey, V.; Coy, D.H.; He, Q. The wound dressings and their applications in wound healing and management. Health Sci. J. 2019, 13, 1–8. [Google Scholar]
  53. Pasut, G.; Veronese, F. Polymer–drug conjugation, recent achievements and general strategies. Prog. Polym. Sci. 2007, 32, 933–961. [Google Scholar] [CrossRef]
  54. Rohini, N.A.; Joseph, A.; Mukerji, A. Polymeric prodrugs: Recent achievements and general strategies. J. Antivir. Antiretrovir. 2013, 2, S15. [Google Scholar]
  55. Abuchowski, A.; van Es, T.; Palczuk, N.C.; Davis, F.F. Alteration of immunological properties of bovine serum albumin by covalent attachment of polyethylene glycol. J. Biol. Chem. 1977, 252, 3578–3581. [Google Scholar] [CrossRef]
  56. Abuchowski, A.; McCoy, J.R.; Palczuk, N.C.; van Es, T.; Davis, F.F. Effect of covalent attachment of polyethylene glycol on immunogenicity and circulating life of bovine liver catalase. J. Biol. Chem. 1977, 252, 3582–3586. [Google Scholar] [CrossRef]
  57. Schoemaker, N.E.; van Kesteren, C.; Rosing, H.; Jansen, S.; Swart, M.; Lieverst, J.; Fraier, D.; Breda, M.; Pellizzoni, C.; Spinelli, R.; et al. A phase I and pharmacokinetic study of MAG-CPT, a water-soluble polymer conjugate of camptothecin. Br. J. Cancer 2002, 87, 608–614. [Google Scholar] [CrossRef]
  58. Kim, C.J.; Lee, Y.S.; Lee, K.H.; Jeong, B.; Kim, T.W.; Kang, T.H.; Kim, H.S.; Park, J. Effect of topical Paclitaxel using PEG/PLGA polymer on the animal model of cervical cancer. Korean J. Gynecol. Oncol. 2008, 19, 68–74. [Google Scholar] [CrossRef]
  59. Maeda, H. SMANCS and polymer-conjugated macromolecular drugs: Advantages in cancer chemotherapy. Adv. Drug Deliv. Rev. 2001, 46, 169–185. [Google Scholar] [CrossRef]
  60. Balasubramaniam, M.P.; Murugan, P.; Chenthamara, D.; Ramakrishnan, S.G.; Salim, A.; Lin, F.; Robert, B.; Subramaniam, S. Synthesis of chitosan-ferulic acid conjugated poly(vinyl alcohol) polymer film for an improved wound healing. Mater. Today Commun. 2020, 25, 101510. [Google Scholar] [CrossRef]
  61. Hardwicke, J.T.; Hart, J.; Bell, A.; Duncan, R.; Thomas, D.W.; Moseley, R. The effect of dextrin–rhEGF on the healing of full-thickness, excisional wounds in the (db/db) diabetic mouse. J. Control. Release 2011, 152, 411–417. [Google Scholar] [CrossRef]
  62. Bowler, P.G.; Duerden, B.I.; Armstrong, D.G. Wound microbiology and associated approaches to wound management. Clin. Microbiol. Rev. 2001, 14, 244–269. [Google Scholar] [CrossRef]
  63. Alam, M.M.; Islam, M.N.; Hossain Hawlader, M.D.; Ahmed, S.; Wahab, A.; Islam, M.; Uddin, K.M.R.; Hossain, A. Prevalence of multidrug resistance bacterial isolates from infected wound patients in Dhaka, Bangladesh: A cross-sectional study. Int. J. Surg. Open 2021, 28, 56–62. [Google Scholar] [CrossRef]
  64. Unalan, I.; Slavik, B.; Buettner, A.; Goldmann, W.H.; Frank, G.; Boccaccini, A.R. Physical and Antibacterial Properties of Peppermint Essential Oil Loaded Poly (ε-caprolactone) (PCL) Electrospun Fiber Mats for Wound Healing. Front. Bioeng. Biotechnol. 2019, 7, 346. [Google Scholar] [CrossRef]
  65. Javanbakht, S.; Nabi, M.; Shadi, M.; Amini, M.M.; Shaabani, A. Carboxymethyl cellulose/tetracycline@UiO-66 nanocomposite hydrogel films as a potential antibacterial wound dressing. Int. J. Biol. Macromol. 2021, 188, 811–819. [Google Scholar] [CrossRef]
  66. Zhong, Y.; Xiao, H.; Seidi, F.; Jin, Y. Natural Polymer-Based Antimicrobial Hydrogels without Synthetic Antibiotics as Wound Dressings. Biomacromolecules 2020, 21, 2983–3006. [Google Scholar] [CrossRef]
  67. Negut, I.; Grumezescu, V.; Grumezescu, A. Treatment Strategies for Infected Wounds. Molecules 2018, 23, 2392. [Google Scholar] [CrossRef]
  68. Jasovský, D.; Littmann, J.; Zorzet, A.; Cars, O. Antimicrobial resistance—A threat to the world’s sustainable development. Upsala J. Med. Sci. 2016, 121, 159–164. [Google Scholar] [CrossRef]
  69. Newman, D.J.; Cragg, G. Natural Products as Sources of New Drugs over the 30 Years from 1981 to 2010. J. Nat. Prod. 2012, 75, 311–335. [Google Scholar] [CrossRef]
  70. Newman, D.J.; Cragg, G. Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019. J. Nat. Prod. 2020, 83, 770–803. [Google Scholar] [CrossRef]
  71. Altaf, F.; Niazi, M.B.K.; Jahan, Z.; Ahmad, T.; Akram, M.A.; Safdar, A.; Butt, M.S.; Noor, T.; Sher, F. Synthesis and Characterization of PVA/Starch Hydrogel Membranes Incorporating Essential Oils Aimed to be Used in Wound Dressing Applications. J. Polym. Environ. 2021, 29, 156–174. [Google Scholar] [CrossRef]
  72. Syed, I.; Garg, S.; Sarkar, P. 5—Entrapment of essential oils in hydrogels for biomedical applications. In Polymeric Gels; Pal, K., Banerjee, I., Eds.; Woodhead Publishing: Cambridge, UK, 2018; pp. 125–141. [Google Scholar]
  73. Mahmood, H.; Khan, I.U.; Asif, M.; Khan, R.U.; Asghar, S.; Khalid, I.; Khalid, S.H.; Irfan, M.; Rehman, F.; Shahzad, Y.; et al. In vitro and in vivo evaluation of gellan gum hydrogel films: Assessing the co impact of therapeutic oils and ofloxacin on wound healing. Int. J. Biol. Macromol. 2021, 166, 483–495. [Google Scholar] [CrossRef]
  74. Chinnaiyan, S.K.; Pandiyan, R.; Natesan, S.; Chindam, S.; Gouti, A.K.; Sugumaran, A. Fabrication of basil oil Nanoemulsion loaded gellan gum hydrogel—Evaluation of its antibacterial and anti-biofilm potential. J. Drug Deliv. Sci. Technol. 2022, 68, 103129. [Google Scholar] [CrossRef]
  75. Ghosh, B.; Bhattacharya, D.; Mukhopadhyay, M. A hydrogel sheet mask with tea tree essential oil entrapment and targeted dose delivery capability. Mater. Today Proc. 2022, 57, 77–83. [Google Scholar] [CrossRef]
  76. Barradas, T.N.; Senna, J.P.; Cardoso, S.A.; Nicoli, S.; Padula, C.; Santi, P.; Rossi, F.; de Holanda e Silva, K.G.; Mansur, C.R.E. Hydrogel-thickened nanoemulsions based on essential oils for topical delivery of psoralen: Permeation and stability studies. Eur. J. Pharm. Biopharm. 2017, 116, 38–50. [Google Scholar] [CrossRef]
  77. Goudoulas, T.B.; Vanderhaeghen, S.; Germann, N. Micro-dispersed essential oils loaded gelatin hydrogels with antibacterial activity. LWT 2022, 154, 112797. [Google Scholar] [CrossRef]
  78. Shabkhiz, M.A.; Khalil Pirouzifard, M.; Pirsa, S.; Mahdavinia, G.R. Alginate hydrogel beads containing Thymus daenensis essential oils/Glycyrrhizic acid loaded in β-cyclodextrin. Investigation of structural, antioxidant/antimicrobial properties and release assessment. J. Mol. Liq. 2021, 344, 117738. [Google Scholar] [CrossRef]
  79. Shukla, R.; Kashaw, S.K.; Jain, A.P.; Lodhi, S. Fabrication of Apigenin loaded gellan gum–chitosan hydrogels (GGCH-HGs) for effective diabetic wound healing. Int. J. Biol. Macromol. 2016, 91, 1110–1119. [Google Scholar] [CrossRef]
  80. Aelenei, P.; Luca, S.V.; Horhogea, C.E.; Rimbu, C.M.; Dimitriu, G.; Macovei, I.; Silion, M.; Aprotosoaie, A.C.; Miron, A. Morus alba leaf extract: Metabolite profiling and interactions with antibiotics against Staphylococcus spp. including MRSA. Phytochem. Lett. 2019, 31, 217–224. [Google Scholar] [CrossRef]
  81. Akhmetova, A.; Saliev, T.; Allan, I.U.; Illsley, M.J.; Nurgozhin, T.; Mikhalovsky, S. A Comprehensive Review of Topical Odor-Controlling Treatment Options for Chronic Wounds. J. Wound Ostomy Cont. Nurs. 2016, 43, 598–609. [Google Scholar] [CrossRef]
  82. Jin, S.G.; Kim, K.S.; Yousaf, A.M.; Kim, D.W.; Jang, S.W.; Son, M.; Kim, Y.H.; Yong, C.S.; Kim, J.O.; Choi, H. Mechanical properties and in vivo healing evaluation of a novel Centella asiatica-loaded hydrocolloid wound dressing. Int. J. Pharm. 2015, 490, 240–247. [Google Scholar] [CrossRef]
  83. Thangavel, A.; Ayyanar, M.; Justin Koilpillai, Y.; Sekar, T. Phytochemical Screening and Antibacterial activity of leaf and callus extracts of Centella asiatica. Bangladesh J. Pharmacol. 2011, 6, 55–60. [Google Scholar]
  84. Kuo, C.-W.; Chiu, Y.-F.; Wu, M.-H.; Li, M.-H.; Wu, C.-N.; Chen, W.-S.; Huang, C.-H. Gelatin/Chitosan Bilayer Patches Loaded with Cortex Phellodendron amurense/Centella asiatica Extracts for Anti-Acne Application. Polymers 2021, 13, 579. [Google Scholar] [CrossRef]
  85. Phaechamud, T.; Yodkhum, K.; Charoenteeraboon, J.; Tabata, Y. Chitosan-aluminum monostearate composite sponge dressing containing asiaticoside for wound healing and angiogenesis promotion in chronic wound. Mater. Sci. Eng. C 2015, 50, 210–225. [Google Scholar] [CrossRef]
  86. Oh, J.; Kim, H.; Beuchat, L.R.; Ryu, J.-H. Inhibition of Staphylococcus aureus on a laboratory medium and black peppercorns by individual and combinations of essential oil vapors. Food Control 2022, 132, 108487. [Google Scholar] [CrossRef]
  87. He, Q.; Zhang, L.; Yang, Z.; Ding, T.; Ye, X.; Liu, D.; Guo, M. Antibacterial mechanisms of thyme essential oil nanoemulsions against Escherichia coli O157:H7 and Staphylococcus aureus: Alterations in membrane compositions and characteristics. Innov. Food Sci. Emerg. Technol. 2022, 75, 102902. [Google Scholar] [CrossRef]
  88. Cruz-Tirado, J.P.; Barros Ferreira, R.S.; Lizárraga, E.; Tapia-Blácido, D.R.; Silva, N.C.C.; Angelats-Silva, L.; Siche, R. Bioactive Andean sweet potato starch-based foam incorporated with oregano or thyme essential oil. Food Packag. Shelf Life 2020, 23, 100457. [Google Scholar] [CrossRef]
  89. Koga, A.Y.; Pereira, A.V.; Lipinski, L.C.; Oliveira, M.R.P. Evaluation of wound healing effect of alginate films containing Aloe vera (Aloe barbadensis Miller) gel. J. Biomater. Appl. 2018, 32, 1212–1221. [Google Scholar] [CrossRef]
  90. Atiba, A.; Nishimura, M.; Kakinuma, S.; Hiraoka, T.; Goryo, M.; Shimada, Y.; Ueno, H.; Uzuka, Y. Aloe vera oral administration accelerates acute radiation-delayed wound healing by stimulating transforming growth factor-β and fibroblast growth factor production. Am. J. Surg. 2011, 201, 809–818. [Google Scholar] [CrossRef]
  91. Guilherme, E.D.; de Souza, C.W.; Bernardo, M.P.; Zenke, M.; Mattoso, L.H.; Moreira, F.K. Antimicrobially active gelatin/[Mg-Al-CO3]-LDH composite films based on clove essential oil for skin wound healing. Mater. Today Commun. 2021, 27, 102169. [Google Scholar]
  92. Razavi, M.S.; Golmohammadi, A.; Nematollahzadeh, A.; Rovera, C.; Farris, S. Cinnamon Essential Oil Encapsulated into a Fish Gelatin-Bacterial Cellulose Nanocrystals Complex and Active Films Thereof. Food Biophys. 2021, 17, 38–46. [Google Scholar] [CrossRef]
  93. Liakos, I.; Rizzello, L.; Scurr, D.J.; Pompa, P.P.; Bayer, I.S.; Athanassiou, A. All-natural composite wound dressing films of essential oils encapsulated in sodium alginate with antimicrobial properties. Int. J. Pharm. 2014, 463, 137–145. [Google Scholar] [CrossRef]
  94. Kavoosi, G.; Dadfar, S.M.; Purfard, A.M. Mechanical, physical, antioxidant, and antimicrobial properties of gelatin films incorporated with thymol for potential use as nano wound dressing. J. Food Sci. 2013, 78, E244–E250. [Google Scholar] [CrossRef]
  95. Otoni, C.G.; Moura, M.R.; Aouada, F.A.; Camilloto, G.P.; Cruz, R.S.; Lorevice, M.V.; Soares, N.F.F.; Mattoso, L.H.C. Antimicrobial and physical-mechanical properties of pectin/papaya puree/cinnamaldehyde nanoemulsion edible composite films. Food Hydrocoll. 2014, 41, 188–194. [Google Scholar] [CrossRef]
  96. Roberts, M.S. Solute-Vehicle-Skin Interactions in Percutaneous Absorption: The Principles and the People. Ski. Pharmacol. Physiol. 2013, 26, 356–370. [Google Scholar] [CrossRef]
  97. Sroczyk, E.A.; Berniak, K.; Jaszczur, M.; Stachewicz, U. Topical electrospun patches loaded with oil for effective gamma linoleic acid transport and skin hydration towards atopic dermatitis skincare. Chem. Eng. J. 2022, 429, 132256. [Google Scholar] [CrossRef]
  98. Tuter, M.; Secundo, F.; Riva, S.; Aksoy, H.A.; Ustun, G. Partial purification of Nigella sativa L. Seed lipase and its application in transesterification reactions. J. Am. Oil Chem. Soc. 2003, 80, 43–48. [Google Scholar] [CrossRef]
  99. Foster, R.H.; Hardy, G.; Alany, R. Borage oil in the treatment of atopic dermatitis. Nutrition 2010, 26, 708–718. [Google Scholar] [CrossRef]
  100. Callaway, J.C. Hempseed as a nutritional resource: An overview. Euphytica 2004, 140, 65–72. [Google Scholar] [CrossRef]
  101. Yoon, S.; Lee, J.; Lee, S. The Therapeutic Effect of Evening Primrose Oil in Atopic Dermatitis Patients with Dry Scaly Skin Lesions Is Associated with the Normalization of Serum Gamma-Interferon Levels. Ski. Pharmacol. Physiol. 2002, 15, 20–25. [Google Scholar] [CrossRef]
  102. Chattopadhyay, P.; Dhiman, S.; Borah, S.; Rabha, B.; Chaurasia, A.K.; Veer, V. Essential oil based polymeric patch development and evaluating its repellent activity against mosquitoes. Acta Trop. 2015, 147, 45–53. [Google Scholar] [CrossRef]
  103. Shan, Y.-H.; Peng, L.-H.; Liu, X.; Chen, X.; Xiong, J.; Gao, J.-Q. Silk fibroin/gelatin electrospun nanofibrous dressing functionalized with astragaloside IV induces healing and anti-scar effects on burn wound. Int. J. Pharm. 2015, 479, 291–301. [Google Scholar] [CrossRef]
  104. Safaee-Ardakani, M.R.; Hatamian-Zarmi, A.; Sadat, S.M.; Mokhtari-Hosseini, Z.B.; Ebrahimi-Hosseinzadeh, B.; Rashidiani, J.; Kooshki, H. Electrospun Schizophyllan/polyvinyl alcohol blend nanofibrous scaffold as potential wound healing. Int. J. Biol. Macromol. 2019, 127, 27–38. [Google Scholar] [CrossRef]
  105. Ranjbar-Mohammadi, M.; Rabbani, S.; Bahrami, S.H.; Joghataei, M.; Moayer, F. Antibacterial performance and in vivo diabetic wound healing of curcumin loaded gum tragacanth/poly (ε-caprolactone) electrospun nanofibers. Mater. Sci. Eng. C 2016, 69, 1183–1191. [Google Scholar] [CrossRef]
  106. Faraji, S.; Nowroozi, N.; Nouralishahi, A.; Shayeh, J.S. Electrospun poly-caprolactone/graphene oxide/quercetin nanofibrous scaffold for wound dressing: Evaluation of biological and structural properties. Life Sci. 2020, 257, 118062. [Google Scholar] [CrossRef]
  107. Ramalingam, R.; Dhand, C.; Leung, C.M.; Ong, S.T.; Annamalai, S.K.; Kamruddin, M.; Verma, N.K.; Ramakrishna, S.; Lakshminarayanan, R.; Arunachalam, K.D. Antimicrobial properties and biocompatibility of electrospun poly-ε-caprolactone fibrous mats containing Gymnema sylvestre leaf extract. Mater. Sci. Eng. C 2019, 98, 503–514. [Google Scholar] [CrossRef]
  108. Amiri, N.; Ajami, S.; Shahroodi, A.; Jannatabadi, N.; Darban, S.A.; Bazzaz, B.S.F.; Pishavar, E.; Kalalinia, F.; Movaffagh, J. Teicoplanin-loaded chitosan-PEO nanofibers for local antibiotic delivery and wound healing. Int. J. Biol. Macromol. 2020, 162, 645–656. [Google Scholar] [CrossRef]
  109. Abdel-Mohsen, A.; Abdel-Rahman, R.; Kubena, I.; Kobera, L.; Spotz, Z.; Zboncak, M.; Prikryl, R.; Brus, J.; Jancar, J. Chitosan-glucan complex hollow fibers reinforced collagen wound dressing embedded with aloe vera. Part I: Preparation and characterization. Carbohydr. Polym. 2020, 230, 115708. [Google Scholar] [CrossRef]
  110. Dong, W.-H.; Liu, J.-X.; Mou, X.-J.; Liu, G.-S.; Huang, X.-W.; Yan, X.; Ning, X.; Russell, S.J.; Long, Y.-Z. Performance of polyvinyl pyrrolidone-isatis root antibacterial wound dressings produced in situ by handheld electrospinner. Colloids Surf. B Biointerfaces 2020, 188, 110766. [Google Scholar] [CrossRef]
  111. Ajmal, G.; Bonde, G.V.; Mittal, P.; Khan, G.; Pandey, V.K.; Bakade, B.V.; Mishra, B. Biomimetic PCL-gelatin based nanofibers loaded with ciprofloxacin hydrochloride and quercetin: A potential antibacterial and anti-oxidant dressing material for accelerated healing of a full thickness wound. Int. J. Pharm. 2019, 567, 118480. [Google Scholar] [CrossRef]
  112. Urena-Saborio, H.; Alfaro-Viquez, E.; Esquivel-Alvarado, D.; Madrigal-Carballo, S.; Gunasekaran, S. Electrospun plant mucilage nanofibers as biocompatible scaffolds for cell proliferation. Int. J. Biol. Macromol. 2018, 115, 1218–1224. [Google Scholar] [CrossRef]
  113. Jafari, A.; Amirsadeghi, A.; Hassanajili, S.; Azarpira, N. Bioactive antibacterial bilayer PCL/gelatin nanofibrous scaffold promotes full-thickness wound healing. Int. J. Pharm. 2020, 583, 119413. [Google Scholar] [CrossRef]
  114. Ardekani, N.T.; Khorram, M.; Zomorodian, K.; Yazdanpanah, S.; Veisi, H.; Veisi, H. Evaluation of electrospun poly (vinyl alcohol)-based nanofiber mats incorporated with Zataria multiflora essential oil as potential wound dressing. Int. J. Biol. Macromol. 2019, 125, 743–750. [Google Scholar] [CrossRef]
  115. Abou Zekry, S.S.; Abdellatif, A.; Azzazy, H.M. Fabrication of pomegranate/honey nanofibers for use as antibacterial wound dressings. Wound Med. 2020, 28, 100181. [Google Scholar] [CrossRef]
  116. Zhou, Y.; Yang, H.; Liu, X.; Mao, J.; Gu, S.; Xu, W. Electrospinning of carboxyethyl chitosan/poly(vinyl alcohol)/silk fibroin nanoparticles for wound dressings. Int. J. Biol. Macromol. 2013, 53, 88–92. [Google Scholar] [CrossRef]
  117. Varshney, N.; Sahi, A.K.; Poddar, S.; Mahto, S.K. Soy protein isolate supplemented silk fibroin nanofibers for skin tissue regeneration: Fabrication and characterization. Int. J. Biol. Macromol. 2020, 160, 112–127. [Google Scholar] [CrossRef]
  118. Selvaraj, S.; Fathima, N.N. Fenugreek Incorporated Silk Fibroin Nanofibers—A Potential Antioxidant Scaffold for Enhanced Wound Healing. ACS Appl. Mater. Interfaces 2017, 9, 5916–5926. [Google Scholar] [CrossRef]
  119. Yousefi, I.; Pakravan, M.; Rahimi, H.; Bahador, A.; Farshadzadeh, Z.; Haririan, I. An investigation of electrospun Henna leaves extract-loaded chitosan based nanofibrous mats for skin tissue engineering. Mater. Sci. Eng. C 2017, 75, 433–444. [Google Scholar] [CrossRef]
  120. Charernsriwilaiwat, N.; Rojanarata, T.; Ngawhirunpat, T.; Sukma, M.; Opanasopit, P. Electrospun chitosan-based nanofiber mats loaded with Garcinia mangostana extracts. Int. J. Pharm. 2013, 452, 333–343. [Google Scholar] [CrossRef]
  121. Adamczak, A.; Ożarowski, M.; Karpiński, T. Curcumin, a Natural Antimicrobial Agent with Strain-Specific Activity. Pharmaceuticals 2020, 13, 153. [Google Scholar] [CrossRef]
  122. Özlem, E. Production of lavender oil loaded antibacterial polymeric membranes. Cumhur. Sci. J. 2020, 41, 160–168. [Google Scholar]
  123. Chomachayi, M.D.; Solouk, A.; Akbari, S.; Sadeghi, D.; Mirahmadi, F.; Mirzadeh, H. Electrospun nanofibers comprising of silk fibroin/gelatin for drug delivery applications: Thyme essential oil and doxycycline monohydrate release study. J. Biomed. Mater. Res. Part A 2018, 106, 1092–1103. [Google Scholar] [CrossRef]
  124. Son, B.C.; Park, C.; Kim, C. Fabrication of antimicrobial nanofiber air filter using activated carbon and cinnamon essential oil. J. Nanosci. Nanotechnol. 2020, 20, 4376–4380. [Google Scholar] [CrossRef]
  125. Liakos, I.L.; Holban, A.M.; Carzino, R.; Lauciello, S.; Grumezescu, A.M. Electrospun fiber pads of cellulose acetate and essential oils with antimicrobial activity. Nanomaterials 2017, 7, 84. [Google Scholar] [CrossRef]
  126. Rieger, K.A.; Schiffman, J. Electrospinning an essential oil: Cinnamaldehyde enhances the antimicrobial efficacy of chitosan/poly(ethylene oxide) nanofibers. Carbohydr. Polym. 2014, 113, 561–568. [Google Scholar] [CrossRef]
  127. Bharathi, B.S.; Stalin, T. Cerium oxide and peppermint oil loaded polyethylene oxide/graphene oxide electrospun nanofibrous mats as antibacterial wound dressings. Mater. Today Commun. 2019, 21, 100664. [Google Scholar]
  128. Bazargani, M.M.; Rohloff, J. Antibiofilm activity of essential oils and plant extracts against Staphylococcus aureus and Escherichia coli biofilms. Food Control 2016, 61, 156–164. [Google Scholar] [CrossRef]
  129. Eğri, Ö.; Erdemir, N. Production of Hypericum perforatum oil-loaded membranes for wound dressing material and in vitro tests. Artif. Cells Nanomed. Biotechnol. 2019, 47, 1404–1415. [Google Scholar] [CrossRef]
  130. Xiang, F.; Bai, J.; Tan, X.; Chen, T.; Yang, W.; He, F. Antimicrobial activities and mechanism of the essential oil from Artemisia argyi Levl. et Van. var. argyi cv. Qiai. Ind. Crops Prod. 2018, 125, 582–587. [Google Scholar] [CrossRef]
  131. Li, T.-T.; Li, J.; Zhang, Y.; Huo, J.-L.; Liu, S.; Shiu, B.-C.; Lin, J.-H.; Lou, C.-W. A study on artemisia argyi oil/sodium alginate/PVA nanofibrous membranes: Micro-structure, breathability, moisture permeability, and antibacterial efficacy. J. Mater. Res. Technol. 2020, 9, 13450–13458. [Google Scholar] [CrossRef]
  132. Lamarra, J.; Bucci, P.; Giannuzzi, L.; Montanari, J.; Rivero, S.; Pinotti, A. Biomaterial-based dressings as vehicle for chitosan-encapsulated cabreuva essential oil: Cytotoxicity and regenerative activity. React. Funct. Polym. 2020, 156, 104728. [Google Scholar] [CrossRef]
  133. Khan, A.u.R.; Huang, K.; Jinzhong, Z.; Zhu, T.; Morsi, Y.; Aldalbahi, A.; El-Newehy, M.; Yan, X.; Mo, X. PLCL/Silk fibroin based antibacterial nano wound dressing encapsulating oregano essential oil: Fabrication, characterization and biological evaluation. Colloids Surf. B: Biointerfaces 2020, 196, 111352. [Google Scholar] [CrossRef]
  134. Özkalp, B.; Sevgi, F.; Özcan, M.; Özcan, M. The antibacterial activity of essential oil of oregano (Origanum vulgare L.). J. Food Agric. Environ. 2010, 8, 6–8. [Google Scholar]
  135. Lu, M.; Dai, T.; Murray, C.K.; Wu, M.X. Bactericidal Property of Oregano Oil Against Multidrug-Resistant Clinical Isolates. Front. Microbiol. 2018, 9, 2329. [Google Scholar] [CrossRef]
  136. Greco, F.; Vicent, M. Combination therapy: Opportunities and challenges for polymer–drug conjugates as anticancer nanomedicines. Adv. Drug Deliv. Rev. 2009, 61, 1203–1213. [Google Scholar] [CrossRef]
  137. Aderibigbe, B.A. Design and therapeutic efficacy of polymer based drug delivery systems for antimalarials. Polym. Sci. Res. Adv. Pract. Appl. Educ. Asp. 2016, 188–198. [Google Scholar]
  138. Sanchis, J.; Canal, F.; Lucas, R.; Vicent, M.J. Polymer–drug conjugates for novel molecular targets. Nanomedicine 2010, 5, 915–935. [Google Scholar] [CrossRef]
  139. Trifković, K.T.; Milašinović, N.Z.; Djordjević, V.B.; Krušić, M.T.K.; Knežević-Jugović, Z.D.; Nedović, V.A.; Bugarski, B.M. Chitosan microbeads for encapsulation of thyme (Thymus serpyllum L.) polyphenols. Carbohydr. Polym. 2014, 111, 901–907. [Google Scholar] [CrossRef]
  140. Sotelo-Boyás, M.; Correa-Pacheco, Z.; Bautista-Baños, S.; Gómez y Gómez, Y. Release study and inhibitory activity of thyme essential oil-loaded chitosan nanoparticles and nanocapsules against foodborne bacteria. Int. J. Biol. Macromol. 2017, 103, 409–414. [Google Scholar] [CrossRef]
  141. Shetta, A.; Kegere, J.; Mamdouh, W. Comparative study of encapsulated peppermint and green tea essential oils in chitosan nanoparticles: Encapsulation, thermal stability, in-vitro release, antioxidant and antibacterial activities. Int. J. Biol. Macromol. 2019, 126, 731–742. [Google Scholar] [CrossRef]
  142. Francesko, A.; Fernandes, M.; Rocasalbas, G.; Gautier, S.; Tzanov, T. Polymers in Wound Repair. In Advanced Polymers in Medicine; Springer: Cham, Switzerland, 2015; pp. 401–431. [Google Scholar]
  143. Croisier, F.; Jérôme, C. Chitosan-based biomaterials for tissue engineering. Eur. Polym. J. 2013, 49, 780–792. [Google Scholar] [CrossRef]
  144. Muthukumar, T.; Senthil, R.; Sastry, T. Synthesis and characterization of biosheet impregnated with Macrotyloma uniflorum extract for burn/wound dressings. Colloids Surf B Biointerfaces 2013, 102, 694–699. [Google Scholar] [CrossRef]
  145. Silva, S.S.; Caridade, S.G.; Mano, J.F.; Reis, R.L. Effect of crosslinking in chitosan/aloe vera-based membranes for biomedical applications. Carbohydr Polym 2013, 98, 581–588. [Google Scholar] [CrossRef]
  146. Agarwal, R.; Alam, S.; Gupta, B. Preparation of Curcumin Loaded Poly(Vinyl Alcohol)-Poly(Ethylene Oxide)-Carboxymethyl Cellulose Membranes for Wound Care Application. J. Biomater. Tissue Eng. 2013, 3, 273–283. [Google Scholar] [CrossRef]
  147. Yang, J.Y.; Singh, D.; Singh, D.; Lee, E.; Choi, S.; Han, S.S.; Park, S.J. Terminalia bellirica Extracts Loaded on Stimuli Responsive HEMA-DEA Hydrogel for Enhanced Growth and Proliferation of Mesenchymal Stem Cells. J. Biomater. Tissue Eng. 2014, 4, 37–45. [Google Scholar] [CrossRef]
  148. Ilhan, E.; Cesur, S.; Guler, E.; Topal, F.; Albayrak, D.; Guncu, M.M.; Cam, M.E.; Taskin, T.; Sasmazel, H.T.; Aksu, B.; et al. Development of Satureja cuneifolia-loaded sodium alginate/polyethylene glycol scaffolds produced by 3D-printing technology as a diabetic wound dressing material. Int. J. Biol. Macromol. 2020, 161, 1040–1054. [Google Scholar] [CrossRef]
  149. Ali Khan, B.; Ullah, S.; Khan, M.K.; Alshahrani, S.M.; Braga, V.A. Formulation and evaluation of Ocimum basilicum-based emulgel for wound healing using animal model. Saudi Pharm. J. 2020, 28, 1842–1850. [Google Scholar] [CrossRef]
  150. Razdan, K.; Kanta, S.; Chaudhary, E.; Kumari, S.; Rahi, D.K.; Yadav, A.K.; Sinha, V.R. Levofloxacin loaded clove oil nanoscale emulgel promotes wound healing in Pseudomonas aeruginosa biofilm infected burn wound in mice. Colloids Surf. B Biointerfaces 2023, 222, 113113. [Google Scholar] [CrossRef]
  151. Eid, A.M.; Jaradat, N.; Issa, L.; Abu-Hasan, A.; Salah, N.; Dalal, M.; Mousa, A.; Zarour, A. Evaluation of anticancer, antimicrobial, and antioxidant activities of rosemary (Rosmarinus Officinalis) essential oil and its Nanoemulgel. Eur. J. Integr. Med. 2022, 55, 102175. [Google Scholar] [CrossRef]
  152. Ting, T.C.; Amat Rahim, N.F.; Che Zaudin, N.A.; Abdullah, N.H.; Mohamad, M.; Shoparwe, N.F.; Mhd Ramle, S.F.; Aimi, Z.; Abdul Hamid, Z.A.; Yusof, A.H. Development and Characterization of Nanoemulgel Containing Piper betle Essential Oil as Active Ingredient. IOP Conf. Ser. Earth Environ. Sci. 2020, 596, 012032. [Google Scholar] [CrossRef]
  153. Barrett, J.F. Can biotech deliver new antibiotics? Curr. Opin. Microbiol. 2005, 8, 498–503. [Google Scholar] [CrossRef]
  154. Pallavali, R.R.; Avula, S.; Degati, V.L.; Penubala, M.; Damu, A.G.; Durbaka, V.R.P. Data of antibacterial activity of plant leaves crude extract on bacterial isolates of wound infections. Data Brief 2019, 24, 103896. [Google Scholar] [CrossRef]
  155. Algreiby, A.A.; Hammer, K.A.; Durmic, Z.; Vercoe, P.; Flematti, G.R. Antibacterial compounds from the Australian native plant Eremophila glabra. Fitoterapia 2018, 126, 45–52. [Google Scholar] [CrossRef]
  156. Smith, J.E.; Tucker, D.; Watson, K.; Jones, G. Identification of antibacterial constituents from the indigenous Australian medicinal plant Eremophila duttonii F. Muell. (Myoporaceae). J. Ethnopharmacol. 2007, 112, 386–393. [Google Scholar] [CrossRef]
  157. Revathi, P.; Parimelazhagan, T.; Manian, S. Quantification of phenolic compounds, in vitro antioxidant analysis and screening of chemical compounds using GC-MS in Acalypha alnifolia Klein ex willd.: A leafy vegetable. Int. J. Pharma Biosci. 2013, 4, 973–986. [Google Scholar]
  158. Noumedem, J.A.; Tamokou, J.D.; Teke, G.N.; Momo, R.C.; Kuete, V.; Kuiate, J.R. Phytochemical analysis, antimicrobial and radical-scavenging properties of Acalypha manniana leaves. SpringerPlus 2013, 2, 503. [Google Scholar] [CrossRef]
  159. Evanjelene, V.K.; Natarajan, D. Evaluation of antioxidant, phytochemical and antibacterial properties of Acalypha alnifolia Klein ex Willd. J. Chem. Pharm. Res. 2013, 5, 205–212. [Google Scholar]
  160. Duraipandiyan, V.; Ayyanar, M.; Ignacimuthu, S. Antimicrobial activity of some ethnomedicinal plants used by Paliyar tribe from Tamil Nadu, India. BMC Complement. Altern. Med. 2006, 6, 35. [Google Scholar] [CrossRef]
  161. Rao, R.V.; Rao, G.; Rao, B. Anti-inflammatory activity of the leaves and bark of delonix elata. Anc. Sci. Life 1997, 17, 141–143. [Google Scholar]
  162. Amabye, T.; Bezabh, A.; Mekonen, F. Phytochemical Constituents and Antioxidant Activity of Delonix elata L. Flower Extract. J. Anal. Pharm. Res. 2016, 2, 00006. [Google Scholar]
  163. Gopal, M.; Shamanna, M. Anti-arthritic and immune modifying potential of delonix elata bark extracts. Res. J. Pharm. Biol. Chem. Sci. 2013, 4, 1642–1648. [Google Scholar]
  164. Kumarappan, M.; Salwe, K.; Shetty, H. Evaluation of Protective Effect of Delonix Elata on Chronic Inflammation and Comparison of its Ulcerogenic Potential with Ibuprofen. J. Pharma Biosci. 2011, 2, 237–243. [Google Scholar]
  165. Vijayasanthi, M.; Kannan, V. Antimicrobial activities of Delonix elata (Bojer ex Hook.) Raf. and Spathodea campanulata P. Beauv. Afr. J. Microbiol. Res. 2014, 8, 697–701. [Google Scholar]
  166. Singh, S.; Kumar, S. A review: Introduction to genus Delonix. World J. Pharm. Pharm. Sci. 2014, 3, 2042–2055. [Google Scholar]
  167. Modi, A.; Mishra, V.; Bhatt, A.; Jain, A.; Mansoori, M.H.; Gurnany, E.; Kumar, V. Delonix regia: Historic perspectives and modern phytochemical and pharmacological researches. Chin. J. Nat. Med. 2016, 14, 31–39. [Google Scholar]
  168. Rahman, M.; Hasan, N.; Das, A.K.; Hossain, T.; Jahan, R.; Khatun, A.; Rahmatullah, M. Effect of Delonix regia leaf extract on glucose tolerance in glucose-induced hyperglycemic mice. Afr. J. Tradit. Complement. Altern. Med. 2011, 8, 34–36. [Google Scholar]
  169. Sama, K.; Xavier, V.; Raja, A. Preliminary phytochemical screening of root bark of Delonix regia. Int. J. Pharm. Life Sci. 2011, 2, 42–43. [Google Scholar]
  170. Kumar, A.R.; Shaik, R.; Yeshwanth, D. Phytochemical evaluation of Delonix regia, Samanea saman, Bauhinia variegatga. Int. J. Res. Pharm. Chem. 2013, 3, 768–772. [Google Scholar]
  171. Shiramane, R.S.; Biradar, K.V.; Chivde, B.V.; Shambhulingayya, H.; Goud, V. In-vivo antidiarrhoeal activity of ethanolic extract of Delonix regia flowers in experimental induced diarrhoea in wistar albino rats. Int. J. Res. Pharm. Chem. 2011, 1, 2231–2781. [Google Scholar]
  172. Shewale, V.D.; Deshmukh, T.A.; Patil, L.S.; Patil, V.R. Anti-inflammatory activity of delonix regia (boj. Ex. Hook). Adv. Pharmacol. Sci. 2011, 2012, 789713. [Google Scholar]
  173. Shabir, G.; Anwar, F.; Sultana, B.; Khalid, Z.M.; Afzal, M.; Khan, Q.M.; Ashrafuzzaman, M. Antioxidant and antimicrobial attributes and phenolics of different solvent extracts from leaves, flowers and bark of Gold Mohar [Delonix regia (Bojer ex Hook.) Raf.]. Molecules 2011, 16, 7302–7319. [Google Scholar] [CrossRef]
  174. Adjé, F.; Lozano, Y.F.; Lozano, P.; Adima, A.; Chemat, F.; Gaydou, E.M. Optimization of anthocyanin, flavonol and phenolic acid extractions from Delonix regia tree flowers using ultrasound-assisted water extraction. Ind. Crops Prod. 2010, 32, 439–444. [Google Scholar] [CrossRef]
  175. Vivek, M.N.; Sachidananda Swamy, H.C.; Manasa, M.; Pallavi, S.; Yashoda Kambar Asha, M.M.; Chaithra, M.; Prashith Kekuda, T.R.; Mallikarjun, N.; Onkarappa, R. Antimicrobial and antioxidant activity of leaf and flower extract of Caesalpinia pulcherrima, Delonix regia and Peltaphorum ferrugineum. J. Appl. Pharm. Sci. 2013, 3, 64. [Google Scholar]
  176. Gupta, R.; Chandra, S. Chemical investigation of Delonix regia Raf. flowers. Indian J. Pharm. 1971, 33, 75. [Google Scholar]
  177. Khan, M.A.; Saxena, A.; Fatima, F.T.; Sharma, G.; Goud, V.; Husain, A. Study of wound healing activity of Delonix regia flowers in experimental animal models. Am. J. PharmTech Res. 2012, 2, 380–390. [Google Scholar]
  178. Parekh, J.; Jadeja, D.; Chanda, S. Efficacy of aqueous and methanol extracts of some medicinal plants for potential antibacterial activity. Turk. J. Biol. 2006, 29, 203–210. [Google Scholar]
  179. Swamy, H.S.; Asha, M.M.; Chaithra, M.; Vivek, M.N.; Yoshoda Kambar, P.K.T.R. Antibacterial activity of flower extract of Caesalpinia pulcherrima, Delonix regia and Peltaphorum ferrugineum against urinary tract pathogens. Int. Res. J. Biol. Sci. 2014, 3, 80–83. [Google Scholar]
  180. Mathad, P.; Mety, S.S. Phytochemical and Antimicrobial Activity of Digera Muricata (L.) Mart. E-J. Chem. 2010, 7, 509254. [Google Scholar] [CrossRef]
  181. Ghaffar, A.; Tung, B.T.; Rahman, R.; Nadeem, F.; Idrees, M. Botanical Specifications, Chemical Composition and Pharmacological Applications of Tartara (Digera muricata L.)–A Review. Int. J. Chem. Biochem. Sci. 2019, 16, 17–22. [Google Scholar]
  182. Khan, M.R.; Ahmed, D. Protective effects of Digera muricata (L.) Mart. on testis against oxidative stress of carbon tetrachloride in rat. Food Chem. Toxicol. 2009, 47, 1393–1399. [Google Scholar] [CrossRef]
  183. Raj, V.P.; Chandrasekhar, R.H.; Vijayan, P.; Dhanaraj, S.A.; Rao, M.C.; Rao, V.J.; Nitesh, K. In vitro and in vivo hepatoprotective effects of the total alkaloid fraction of Hygrophila auriculata leaves. Indian J. Pharmacol. 2010, 42, 99–104. [Google Scholar]
  184. Ahmed, S.; Riaz, M.; Malik, A.; Shahid, M. Effect of seed extracts of Withania somnifera, Croton tiglium and Hygrophila auriculata on behavior and physiology of Odontotermes obesus (Isoptera, Termitidae). Biologia 2007, 62, 770–773. [Google Scholar] [CrossRef]
  185. Govindachari, T.; Nagarajan, K.; Pai, B. Isolation of lupeol from the root of Asteracantha longifolia Nees. Indian J. Sci. Res. B 1957, 16, 72p. [Google Scholar]
  186. Nabèrè, O.; Adama, H.; Samson, G.; Kiessoum, K.; Patrice, Z.; Roland, M.N.-T.; Moussa, C.; Martin, K.; Jea, M.F. Antibacterial and phytochemical studies of three Acanthaceae species used in Burkina Faso traditional medicine. J. Appl. Pharm. Sci. 2013, 3, 49. [Google Scholar]
  187. Godbole, N.; Gunde, B.; Srivastava, P. An investigation of oil from seed of Hygrophila spinosa. Oil Soap 1941, 18, 206–207. [Google Scholar] [CrossRef]
  188. Parashar, V.; Singh, H. Investigation of Astercantha longifolia Nees. Indian J. Pharmacol. 1965, 27, 109–113. [Google Scholar]
  189. Shanmugasundaram, P.; Venkataraman, S. Hepatoprotective and antioxidant effects of Hygrophila auriculata (K. Schum) Heine Acanthaceae root extract. J. Ethnopharmacol. 2006, 104, 124–128. [Google Scholar] [CrossRef]
  190. Vijayakumar, M.; Govindarajan, R.; Rao, G.M.; Rao, C.V.; Shirwaikar, A.; Mehrotra, S.; Pushpangadan, P. Action of Hygrophila auriculata against streptozotocin-induced oxidative stress. J. Ethnopharmacol. 2006, 104, 356–361. [Google Scholar] [CrossRef]
  191. Esther, V.C.J.; Saraswathi, R.; Dhanasekar, S. In vitro antibacterial and antifungal activities along with X-ray irradiation studies of medicinal plant Hygrophila Auriculata. Int. J. Pharm. Pharm. Sci. 2012, 4, 352–358. [Google Scholar]
  192. Vlietinck, A.; Van Hoof, L.; Totte, J.; Lasure, A.; Berghe, D.V.; Rwangabo, P.C.; Mvukiyumwami, J. Screening of hundred Rwandese medicinal plants for antimicrobial and antiviral properties. J. Ethnopharmacol. 1995, 46, 31–47. [Google Scholar] [CrossRef]
  193. Laxmichand, B.H.; Modi, D. A Comprehensive Review on Maerua Oblongifolia. Int. J. Res. Advent Technol. 2019, 7, 721–727. [Google Scholar]
  194. Abdel-Mogib, M. A lupane triterpenoid from Maerua oblongifolia. Phytochemistry 1999, 51, 445–448. [Google Scholar] [CrossRef]
  195. Arulanand Raj, N.; Gopal, V.; Dhivya, S.; Jayabalan, G. Evaluation of wound healing effect of Maerua Oblongifolia in albino rats. World J. Pharm. Res. 2018, 8, 1380–1385. [Google Scholar]
  196. Sasi Priya, S. Maerua oblongifolia–What do we really know? Overview, Progress and Perspectives. J. PeerScientist 2020, 2, e1000012. [Google Scholar]
  197. Manjunatha, B. Antibacterial activity of Pterocarpus santalinus. Indian J. Pharm. Sci. 2006, 68, 115. [Google Scholar] [CrossRef]
  198. Karthick, M.; Parthiban, K. Chemical characterization of Pterocarpus santalinus wood using GC-MS. J. Pharmacogn. Phytochem. 2019, 8, 380–382. [Google Scholar]
  199. Stella, J.; Krishnamoorthy, P.; Mohamed, A.J.; Anand, M. Free Radical Scavenging and Antibacterial Evaluation of Pterocarpus Santalinus Leaf In-Vitro Study. Int. J. Pharm. Sci. Res. 2011, 2, 1204. [Google Scholar]
  200. Chagas, V.T.; França, L.M.; Malik, S.; Paes, A.M.A. Syzygium cumini (L.) skeels: A prominent source of bioactive molecules against cardiometabolic diseases. Front. Pharmacol. 2015, 6, 259. [Google Scholar] [CrossRef]
  201. Bandiola, T.; Ignacio, G.B.; Yunson, E.G.; Bandiola, P.D. Syzygium cumini (L.) Skeels: A review of its phytochemical constituents, toxicity studies, and traditional and pharmacological uses. Int. J. Appl. Pharm. Biol. Res. 2017, 2, 15–23. [Google Scholar]
  202. Ayyanar, M.; Subash-Babu, P. Syzygium cumini (L.) Skeels: A review of its phytochemical constituents and traditional uses. Asian Pac. J. Trop. Biomed. 2012, 2, 240–246. [Google Scholar]
  203. Gowri, S.S.; Vasantha, K. Phytochemical screening and antibacterial activity of Syzygium cumini (L.)(Myrtaceae) leaves extracts. Int. J. Pharm. Tech. Res. 2010, 2, 1569–1573. [Google Scholar]
  204. McKenzie, A.; Price, J. The alkaloids of Gyrocarpus americanus Jacq. Aust. J. Chem. 1953, 6, 180–185. [Google Scholar] [CrossRef]
  205. Bradacs, G.; Maes, L.; Heilmann, J. In vitro cytotoxic, antiprotozoal and antimicrobial activities of medicinal plants from Vanuatu. Phytother. Res. 2010, 24, 800–809. [Google Scholar] [CrossRef]
  206. Steenkamp, V.; Fernandes, A.C.; van Rensburg, C.E.J. Antibacterial activity of Venda medicinal plants. Fitoterapia 2007, 78, 561–564. [Google Scholar] [CrossRef]
  207. Prakash, C.V.S.; Prakash, I. Bioactive chemical constituents from pomegranate (Punica granatum) juice, seed and peel—A review. Int. J. Res. Chem. Environ. 2011, 1, 1–18. [Google Scholar]
  208. Jurenka, J. Therapeutic applications of pomegranate (Punica granatum L.): A review. Altern. Med. Rev. 2008, 13, 128–144. [Google Scholar]
  209. Gil, M.I.; Tomás-Barberán, F.A.; Hess-Pierce, B.; Holcroft, D.M.; Kader, A.A. Antioxidant activity of pomegranate juice and its relationship with phenolic composition and processing. J. Agric. Food Chem. 2000, 48, 4581–4589. [Google Scholar] [CrossRef]
  210. Schubert, S.Y.; Lansky, E.P.; Neeman, I. Antioxidant and eicosanoid enzyme inhibition properties of pomegranate seed oil and fermented juice flavonoids. J. Ethnopharmacol. 1999, 66, 11–17. [Google Scholar] [CrossRef]
  211. Murthy, K.N.C.; Jayaprakasha, G.K.; Singh, R.P. Studies on Antioxidant Activity of Pomegranate (Punica granatum) Peel Extract Using in Vivo Models. J. Agric. Food Chem. 2002, 50, 4791–4795. [Google Scholar] [CrossRef]
  212. Prashanth, D.; Asha, M.; Amit, A. Antibacterial activity of Punica granatum. Fitoterapia 2001, 72, 171–173. [Google Scholar] [CrossRef]
  213. Álvarez-Martínez, F.J.; Rodríguez, J.C.; Borrás-Rocher, F.; Barrajón-Catalán, E.; Micol, V. The antimicrobial capacity of Cistus salviifolius and Punica granatum plant extracts against clinical pathogens is related to their polyphenolic composition. Sci. Rep. 2021, 11, 588. [Google Scholar] [CrossRef]
  214. Falodun, A.; Okunrobo, L.; Uzoamaka, N. Short Communication—Phytochemical screening and anti-inflammatory evaluation of methanolic and aqueous extracts of Euphorbia heterophylla Linn (Euphorbiaceae). Afr. J. Biotechnol. 2006, 5, 529–531. [Google Scholar]
  215. Onwuka, G.I. Food Analysis and Instrumentation: Theory and Practice; Napthali Prints: Lagos, Nigeria, 2005. [Google Scholar]
  216. Akinmutimi, A. Nutritive value of raw and processed jack fruit seeds (Artocarpus heterophyllus). Chem. Analysis. Agric. J. 2006, 1, 266–271. [Google Scholar]
  217. Chang, C.-C.; Yang, M.-H.; Wen, H.-M.; Chern, J.-C. Estimation of total flavonoid content in propolis by two complementary colorimetric methods. J. Food Drug Anal. 2002, 10, 178–182. [Google Scholar]
  218. James, O.; Friday, E.T. Phytochemical composition, bioactivity and wound healing potential of Euphorbia heterophylla (Euphorbiaceae) leaf extract. Int. J. Pharm. Biomed. Res. 2010, 1, 54–63. [Google Scholar]
  219. Ughachukwu, P.; Ezenyeaku, C.C.T.; Ochiogu, B.C.; Ezeagwuna, D.A.; Anahalu, I.C. Evaluation of antibacterial activities of Euphorbia heterophylla. IOSR J. Dent. Med. Sci. 2014, 13, 69–75. [Google Scholar]
  220. Falodun, A.; Agbakwuru, E.; Ukoh, G. Antibacterial Activity of Euphor-Bia Heterophylla Linn (Family-Euphorbiaceae). Biol. Sci. PJSIR 2003, 46, 471–472. [Google Scholar]
  221. Hosseinzadeh, M.H.; Ghalavand, A.; Boojar, M.M.-A.; Modarres-Sanavy, S.A.M.; Mokhtassi-Bidgoli, A. Application of manure and biofertilizer to improve soil properties and increase grain yield, essential oil and ω3 of purslane (Portulaca oleracea L.) under drought stress. Soil Tillage Res. 2021, 205, 104633. [Google Scholar] [CrossRef]
  222. Cowan, M.M. Plant products as antimicrobial agents. Clin. Microbiol. Rev. 1999, 12, 564–582. [Google Scholar] [CrossRef]
  223. Gozari, M.; Alborz, M.; El-Seedi, H.R.; Jassbi, A.R. Chemistry, biosynthesis and biological activity of terpenoids and meroterpenoids in bacteria and fungi isolated from different marine habitats. Eur. J. Med. Chem. 2021, 210, 112957. [Google Scholar] [CrossRef]
  224. Tripathi, N.; Asthana, A.; Dixit, S. Toxicity of some terpenoids against fungi infesting fruits and seeds of Capsicum annuum L. during storage. J. Phytopathol. 1984, 110, 328–335. [Google Scholar] [CrossRef]
  225. Jiang, Z.-Y.; Zhou, J.; Huang, C.-G.; Hu, Q.-F.; Huang, X.-Z.; Wang, W.; Zhang, L.-Z.; Li, G.-P.; Xia, F.-T. Two novel antiviral terpenoids from the cultured Perovskia atriplicifolia. Tetrahedron 2015, 71, 3844–3849. [Google Scholar] [CrossRef]
  226. 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; Elsevier: Amsterdam, The Netherlands, 2018; pp. 227–250. [Google Scholar]
  227. Aziz, Z.A.; Ahmad, A.; Setapar, S.H.; Karakucuk, A.; Azim, M.M.; Lokhat, D.; Rafatullah, M.; Ganash, M.; Kamal, M.A.; Ashraf, G.M. Essential Oils: Extraction Techniques, Pharmaceutical and Therapeutic Potential—A Review. Curr. Drug Metab. 2018, 19, 1100–1110. [Google Scholar] [CrossRef]
  228. Ge, L.; Lin, B.; Mo, J.; Chen, Q.; Su, L.; Li, Y.; Yang, K. Composition and antioxidant and antibacterial activities of essential oils from three yellow Camellia species. Trees 2019, 33, 205–212. [Google Scholar] [CrossRef]
  229. Ghaderi, L.; Aliahmadi, A.; Ebrahimi, S.N.; Rafati, H. Effective Inhibition and eradication of Pseudomonas aeruginosa biofilms by Satureja khuzistanica essential oil nanoemulsion. J. Drug Deliv. Sci. Technol. 2021, 61, 102260. [Google Scholar] [CrossRef]
  230. Zheng, G.-Q.; Kenney, P.M.; Lam, L.K.T. Sesquiterpenes from Clove (Eugenia caryophyllata) as Potential Anticarcinogenic Agents. J. Nat. Prod. 1992, 55, 999–1003. [Google Scholar] [CrossRef]
  231. Beuchat, L. Control of Foodborne Pathogens and Spoilage Microorganisms by Naturally Occurring Antimicrobials. In Microbial Food Contamination; CRC Press: Boca Raton, FL, USA, 2000. [Google Scholar]
  232. Anwer, M.K.; Jamil, S.; Ibnouf, E.O.; Shakeel, F. Enhanced antibacterial effects of clove essential oil by nanoemulsion. J. Oleo Sci. 2014, 63, 347–354. [Google Scholar] [CrossRef]
  233. Miyazawa, M.; Hisama, M. Suppression of chemical mutagen-induced SOS response by alkylphenols from clove (Syzygium aromaticum) in the Salmonella typhimurium TA1535/pSK1002 umu test. J. Agric. Food Chem. 2001, 49, 4019–4025. [Google Scholar] [CrossRef]
  234. Friedman, M.; Henika, P.R.; Mandrell, R.E. Bactericidal activities of plant essential oils and some of their isolated constituents against Campylobacter jejuni, Escherichia coli, Listeria monocytogenes, and Salmonella enterica. J. Food Prot. 2002, 65, 1545–1560. [Google Scholar] [CrossRef]
  235. Cressy, H.K.; Jerrett, A.R.; Osborne, C.M.; Bremer, P.J. A novel method for the reduction of numbers of Listeria monocytogenes cells by freezing in combination with an essential oil in bacteriological media. J. Food Prot. 2003, 66, 390–395. [Google Scholar] [CrossRef]
  236. Ojeda-Sana, A.M.; van Baren, C.M.; Elechosa, M.A.; Juárez, M.A.; Moreno, S. New insights into antibacterial and antioxidant activities of rosemary essential oils and their main components. Food Control 2013, 31, 189–195. [Google Scholar] [CrossRef]
  237. Sienkiewicz, M.; Łysakowska, M.; Pastuszka, M.; Bienias, W.; Kowalczyk, E. The potential of use basil and rosemary essential oils as effective antibacterial agents. Molecules 2013, 18, 9334–9351. [Google Scholar] [CrossRef]
  238. Amani, F.; Sami, M.; Rezaei, A. Characterization and Antibacterial Activity of Encapsulated Rosemary Essential Oil within Amylose Nanostructures as a Natural Antimicrobial in Food Applications. Starch Stärke 2021, 73, 2100021. [Google Scholar] [CrossRef]
  239. Diao, W.-R.; Hu, Q.-P.; Zhang, H.; Xu, J.-G. Chemical composition, antibacterial activity and mechanism of action of essential oil from seeds of fennel (Foeniculum vulgare Mill.). Food Control 2014, 35, 109–116. [Google Scholar] [CrossRef]
  240. Özbek, H.; Uğraş, S.; Dülger, H.; Bayram, I.; Tuncer, I.; Öztürk, G.; Öztürk, A. Hepatoprotective effect of Foeniculum vulgare essential oil. Fitoterapia 2003, 74, 317–319. [Google Scholar] [CrossRef]
  241. Singh, G.; Maurya, S.; De Lampasona, M.P.; Catalan, C. Chemical constituents, antifungal and antioxidative potential of Foeniculum vulgare volatile oil and its acetone extract. Food Control 2006, 17, 745–752. [Google Scholar] [CrossRef]
  242. Tognolini, M.; Ballabeni, V.; Bertoni, S.; Bruni, R.; Impicciatore, M.; Barocelli, E. Protective effect of Foeniculum vulgare essential oil and anethole in an experimental model of thrombosis. Pharmacol. Res. 2007, 56, 254–260. [Google Scholar] [CrossRef]
  243. Choi, E.-M.; Hwang, J.-K. Antiinflammatory, analgesic and antioxidant activities of the fruit of Foeniculum vulgare. Fitoterapia 2004, 75, 557–565. [Google Scholar] [CrossRef]
  244. Abou El-Soud, N.; El-Laithy, N.; El-Saeed, G.; Wahby, M.; Khalil, M.; Morsy, F.; Shaffie, N. Antidiabetic activities of Foeniculum vulgare Mill. essential oil in streptozotocin-induced diabetic rats. Maced. J. Med. Sci. 2011, 4, 139–146. [Google Scholar]
  245. Pradhan, M.; Sribhuwaneswari, S.; Karthikeyan, D.; Minz, S.; Sure, P.; Chandu, A.N.; Mishra, U.; Kamalakannan, K.; Saravanankumar, A.; Sivakumar, T. In-vitro cytoprotection activity of Foeniculum vulgare and Helicteres isora in cultured human blood lymphocytes and antitumour activity against B16F10 melanoma cell line. Res. J. Pharm. Technol. 2008, 1, 450–452. [Google Scholar]
  246. Lee, H.-S. Acaricidal activity of constituents identified in Foeniculum vulgare fruit oil against Dermatophagoides spp.(Acari: Pyroglyphidae). J. Agric. Food Chem. 2004, 52, 2887–2889. [Google Scholar] [CrossRef]
  247. Arweiler, N.B.; Donos, N.; Netuschil, L.; Reich, E.; Sculean, A. Clinical and antibacterial effect of tea tree oil—A pilot study. Clin. Oral Investig. 2000, 4, 70–73. [Google Scholar] [CrossRef]
  248. Lee, C.-J.; Chen, L.-W.; Chen, L.-G.; Chang, T.-L.; Huang, C.-W.; Huang, M.-C.; Wang, C.-C. Correlations of the components of tea tree oil with its antibacterial effects and skin irritation. J. Food Drug Anal. 2013, 21, 169–176. [Google Scholar] [CrossRef]
  249. Mondello, F.; De Bernardis, F.; Girolamo, A.; Cassone, A.; Salvatore, G. In vivo activity of terpinen-4-ol, the main bioactive component of Melaleuca alternifolia Cheel (tea tree) oil against azole-susceptible and-resistant human pathogenic Candida species. BMC Infect. Dis. 2006, 6, 158. [Google Scholar] [CrossRef]
  250. El Atki, Y.; Aouam, I.; El Kamari, F.; Taroq, A.; Nayme, K.; Timinouni, M.; Lyoussi, B.; Abdellaoui, A. Antibacterial activity of cinnamon essential oils and their synergistic potential with antibiotics. J. Adv. Pharm. Technol. Res. 2019, 10, 63. [Google Scholar] [CrossRef]
  251. Yang, K.; Liu, A.; Hu, A.; Li, J.; Zen, Z.; Liu, Y.; Tang, S.; Li, C. Preparation and characterization of cinnamon essential oil nanocapsules and comparison of volatile components and antibacterial ability of cinnamon essential oil before and after encapsulation. Food Control 2021, 123, 107783. [Google Scholar] [CrossRef]
  252. Seyed Ahmadi, S.G.; Farahpour, M.R.; Hamishehkar, H. Topical application of Cinnamon verum essential oil accelerates infected wound healing process by increasing tissue antioxidant capacity and keratin biosynthesis. Kaohsiung J. Med. Sci. 2019, 35, 686–694. [Google Scholar] [CrossRef]
  253. Radünz, M.; Mota Camargo, T.; Santos Hackbart, H.C.; Inchauspe Correa Alves, P.; Radünz, A.L.; Avila Gandra, E.; da Rosa Zavareze, E. Chemical composition and in vitro antioxidant and antihyperglycemic activities of clove, thyme, oregano, and sweet orange essential oils. LWT 2021, 138, 110632. [Google Scholar] [CrossRef]
  254. Liu, Z.; Lin, D.; Shen, R.; Zhang, R.; Liu, L.; Yang, X. Konjac glucomannan-based edible films loaded with thyme essential oil: Physical properties and antioxidant-antibacterial activities. Food Packag. Shelf Life 2021, 29, 100700. [Google Scholar] [CrossRef]
  255. Govaris, A.; Solomakos, N.; Pexara, A.; Chatzopoulou, P.S. The antimicrobial effect of oregano essential oil, nisin and their combination against Salmonella Enteritidis in minced sheep meat during refrigerated storage. Int. J. Food Microbiol. 2010, 137, 175–180. [Google Scholar] [CrossRef]
  256. Cattelan, M.G.; de Castilhos, M.B.M.; Sales, P.J.P.; Hoffmann, F.L. Antibacterial activity of oregano essential oil against foodborne pathogens. Nutr. Food Sci. 2013, 43, 169–174. [Google Scholar] [CrossRef]
  257. Cui, H.; Zhang, C.; Li, C.; Lin, L. Antibacterial mechanism of oregano essential oil. Ind. Crop. Prod. 2019, 139, 111498. [Google Scholar] [CrossRef]
  258. Beatovic, D.; Krstic-Milosevic, D.; Trifunovic, S.; Siljegovic, J.; Glamoclija, J.; Ristic, M.; Jelacic, S. Chemical composition, antioxidant and antimicrobial activities of the essential oils of twelve Ocimum basilicum L. cultivars grown in Serbia. Rec. Nat. Prod. 2015, 9, 62. [Google Scholar]
  259. Gaio, I.; Saggiorato, A.G.; Treichel, H.; Cichoski, A.J.; Astolfi, V.; Cardoso, R.I.; Toniazzo, G.; Valduga, E.; Paroul, N.; Cansian, R.L. Antibacterial activity of basil essential oil (Ocimum basilicum L.) in Italian-type sausage. J. Verbrauch. Lebensm. 2015, 10, 323–329. [Google Scholar] [CrossRef]
  260. Sharafati Chaleshtori, R.; Rokni, N.; Rafieian-Kopaei, M.; Deris, F.; Salehi, E. Antioxidant and antibacterial activity of basil (Ocimum basilicum L.) essential oil in beef burger. J. Agric. Sci. Technol. 2015, 17, 817–826. [Google Scholar]
  261. Van Dat, D.; Van Cuong, N.; Le, P.H.A.; Anh, T.T.L.; Viet, P.T.; Huong, N.T.L. Orange Peel Essential Oil Nanoemulsions Supported by Nanosilver for Antibacterial Application. Indones. J. Chem. 2020, 20, 430–439. [Google Scholar]
  262. Do Evangelho, J.A.; da Silva Dannenberg, G.; Biduski, B.; el Halal, S.L.M.; Kringel, D.H.; Gularte, M.A.; Fiorentini, A.M.; da Rosa Zavareze, E. Antibacterial activity, optical, mechanical, and barrier properties of corn starch films containing orange essential oil. Carbohydr. Polym. 2019, 222, 114981. [Google Scholar] [CrossRef]
  263. Kang, J.; Jin, W.; Wang, J.; Sun, Y.; Wu, X.; Liu, L. Antibacterial and anti-biofilm activities of peppermint essential oil against Staphylococcus aureus. LWT 2019, 101, 639–645. [Google Scholar] [CrossRef]
  264. Desam, N.R.; Al-Rajab, A.J.; Sharma, M.; Mylabathula, M.M.; Gowkanapalli, R.R.; Albratty, M. Chemical constituents, in vitro antibacterial and antifungal activity of Mentha×Piperita L. (peppermint) essential oils. J. King Saud Univ. Sci. 2019, 31, 528–533. [Google Scholar] [CrossRef]
  265. Afsharypuor, S.; Rahiminezhad, M.; Ghaemmaghami, L.; Soleimani, M.S.; Khanmohammadic, M.; Afsharipour, N. Essential Oil Constituents of Leaves of the Male and Female Shrubs of Juniperus chinensis L. from Isfahan. Iran. J. Pharm. Sci. 2007, 3, 177–180. [Google Scholar]
  266. Majumder, S.; Ghosh, A.; Bhattacharya, M. Natural anti-inflammatory terpenoids in Camellia japonica leaf and probable biosynthesis pathways of the metabolome. Bull. Natl. Res. Cent. 2020, 44, 141. [Google Scholar] [CrossRef]
  267. Feás, X.; Estevinho, L.M.; Salinero, C.; Vela, P.; Sainz, M.J.; Vázquez-Tato, M.P.; Seijas, J.A. Triacylglyceride, Antioxidant and Antimicrobial Features of Virgin Camellia oleifera, C. reticulata and C. sasanqua Oils. Molecules 2013, 18, 4573–4587. [Google Scholar] [CrossRef]
  268. Chung, I.-M.; Kim, M.Y.; Park, W.-H.; Moon, H.-I. Antiatherogenic activity of Dendropanax morbifera essential oil in rats. Die Pharm. Int. J. Pharm. Sci. 2009, 64, 547–549. [Google Scholar]
  269. Werka, J.S.; Boehme, A.K.; Setzer, W.N. Biological Activities of Essential Oils from Monteverde, Costa Rica. Nat. Prod. Commun. 2007, 2, 1934578X0700201204. [Google Scholar] [CrossRef]
  270. Chaleshtori, R.S.; Kopaei, M.R.; Salehi, E. Bioactivity of Apium petroselinum and Portulaca oleracea Essential Oils as Natural Preservatives. Jundishapur J. Microbiol. 2015, 8, e20128. [Google Scholar] [CrossRef]
  271. Pang, J.; Dong, W.; Li, Y.; Xia, X.; Liu, Z.; Hao, H.; Jiang, L.; Liu, Y. Purification of Houttuynia cordata Thunb. Essential Oil Using Macroporous Resin Followed by Microemulsion Encapsulation to Improve Its Safety and Antiviral Activity. Molecules 2017, 22, 293. [Google Scholar] [CrossRef]
  272. Lu, H.; Wu, X.; Liang, Y.; Zhang, J. Variation in Chemical Composition and Antibacterial Activities of Essential Oils from Two Species of Houttuynia Thunb. Chem. Pharm. Bull. 2006, 54, 936–940. [Google Scholar] [CrossRef]
  273. Ali, E.M. Phytochemical composition, antifungal, antiaflatoxigenic, antioxidant, and anticancer activities of Glycyrrhiza glabra L. and Matricaria chamomilla L. essential oils. J. Med. Plants Res. 2013, 7, 2197–2207. [Google Scholar]
  274. Bahrami, A.; Fattahi, R. Biodegradable carboxymethyl cellulose–polyvinyl alcohol composite incorporated with Glycyrrhiza Glabra L. essential oil: Physicochemical and antibacterial features. Food Sci. Nutr. 2021, 9, 4974–4985. [Google Scholar] [CrossRef]
  275. Yong, C.; Zhang, Y.-Q. Advances in the Studies of Chemical Constituents of Genus Ziziphus. Nat. Prod. Res. Dev. 2011, 23, 979–982. [Google Scholar]
  276. Al-Reza, S.M.; Rahman, A.; Lee, J.; Kang, S.C. Potential roles of essential oil and organic extracts of Zizyphus jujuba in inhibiting food-borne pathogens. Food Chem. 2010, 119, 981–986. [Google Scholar] [CrossRef]
  277. Kasuya, H.; Hata, E.; Satou, T.; Yoshikawa, M.; Hayashi, S.; Masuo, Y.; Koike, K. Effect on Emotional Behavior and Stress by Inhalation of the Essential oil from Chamaecyparis obtusa. Nat. Prod. Commun. 2013, 8, 1934578X1300800428. [Google Scholar] [CrossRef]
  278. Yang, J.-K.; Choi, M.-S.; Seo, W.-T.; Rinker, D.L.; Han, S.W.; Cheong, G.-W. Chemical composition and antimicrobial activity of Chamaecyparis obtusa leaf essential oil. Fitoterapia 2007, 78, 149–152. [Google Scholar] [CrossRef]
  279. Rana, K.L.; Kour, D.; Kaur, T.; Devi, R.; Yadav, A.N.; Yadav, N.; Dhaliwal, H.S.; Saxena, A.K. Endophytic microbes: Biodiversity, plant growth-promoting mechanisms and potential applications for agricultural sustainability. Antonie Leeuwenhoek 2020, 113, 1075–1107. [Google Scholar] [CrossRef]
  280. Nisa, H.; Kamili, A.N.; Nawchoo, I.A.; Shafi, S.; Shameem, N.; Bandh, S.A. Fungal endophytes as prolific source of phytochemicals and other bioactive natural products: A review. Microb. Pathog. 2015, 82, 50–59. [Google Scholar] [CrossRef]
  281. Caruso, D.J.; Palombo, E.A.; Moulton, S.E.; Zaferanloo, B. Exploring the Promise of Endophytic Fungi: A Review of Novel Antimicrobial Compounds. Microorganisms 2022, 10, 1990. [Google Scholar] [CrossRef]
  282. Zaferanloo, B.; Mahon, P.J.; Palombo, E.A. Endophytes from medicinal plants as novel sources of bioactive compounds. In Medicinal Plants: Diversity and Drugs; CRC Press: Boca Raton, FL, USA, 2012; Volume 355, p. 411. [Google Scholar]
  283. Pasrija, P.; Girdhar, M.; Kumar, M.; Arora, S.; Katyal, A. Endophytes: An unexplored treasure to combat Multidrug resistance. Phytomedicine Plus 2022, 2, 100249. [Google Scholar] [CrossRef]
  284. Deshmukh, S.K.; Dufossé, L.; Chhipa, H.; Saxena, S.; Mahajan, G.B.; Gupta, M.K. Fungal Endophytes: A Potential Source of Antibacterial Compounds. J. Fungi 2022, 8, 164. [Google Scholar] [CrossRef]
  285. Samadzadeh, S.; Farzaneh, M.; Shahsavari, Z.; Ebrahimi, S.N.; Asadollahi, M.; Mirjalili, M.H. Characterization and antimicrobial activity of fungal endophytes from Crocus caspius (Iridaceae). Biocatal. Agric. Biotechnol. 2022, 43, 102429. [Google Scholar] [CrossRef]
  286. Dos Santos Lacerda, Í.C.; Polonio, J.C.; Golias, H.C. ENDophytic Fungi as Sources of Antiviral Compounds—A Review. Chem. Biodivers. 2022, 19, e202100971. [Google Scholar]
  287. Macías-Rubalcava, M.L.; Hernández-Bautista, B.E.; Jiménez-Estrada, M.; González, M.C.; Glenn, A.E.; Hanlin, R.T.; Hernández-Ortega, S.; Saucedo-García, A.; Muria-González, J.M.; Anaya, A.L. Naphthoquinone spiroketal with allelochemical activity from the newly discovered endophytic fungus Edenia gomezpompae. Phytochemistry 2008, 69, 1185–1196. [Google Scholar] [CrossRef]
  288. Wu, S.H.; Huang, R.; Miao, C.P.; Chen, Y.W. Two new steroids from an endophytic fungus Phomopsis sp. Chem. Biodivers. 2013, 10, 1276–1283. [Google Scholar] [CrossRef]
  289. Rojas-Solís, D.; Zetter-Salmón, E.; Contreras-Pérez, M.; del Carmen Rocha-Granados, M.; Macías-Rodríguez, L.; Santoyo, G. Pseudomonas stutzeri E25 and Stenotrophomonas maltophilia CR71 endophytes produce antifungal volatile organic compounds and exhibit additive plant growth-promoting effects. Biocatal. Agric. Biotechnol. 2018, 13, 46–52. [Google Scholar] [CrossRef]
  290. Mishra, P.; Verekar, S.; Kulkarni-Almeida, A.; Roy, S.; Jain, S.; Balakrishnan, A.; Vishwakarma, R.; Deshmukh, S. Anti-inflammatory and anti-diabetic naphtha quinones from an endophytic fungus Dendryphion nanum (Nees) S. Hughes. Indian J. Chem. Sect. B 2013, 52B, 565–567. [Google Scholar]
  291. Toghueo, R.M.K. Anti-leishmanial and anti-inflammatory agents from endophytes: A review. Nat. Prod. Bioprospecting 2019, 9, 311–328. [Google Scholar] [CrossRef]
  292. Banyal, A.; Thakur, V.; Thakur, R.; Kumar, P. Endophytic microbial diversity: A new hope for the production of novel anti-tumor and anti-HIV agents as future therapeutics. Curr. Microbiol. 2021, 78, 1699–1717. [Google Scholar] [CrossRef]
  293. Zaferanloo, B.; Pepper, S.A.; Coulthard, S.A.; Redfern, C.P.F.; Palombo, E.A. Metabolites of endophytic fungi from Australian native plants as potential anticancer agents. FEMS Microbiol. Lett. 2018, 365, fny078. [Google Scholar] [CrossRef]
  294. Jung, C.; Arnold, A.E. The Effects of Endohyphal Bacteria on Anti-Cancer and Anti-Malaria Metabolites of Endophytic Fungi. Bachelor’s Thesis, The University of Arizona, Tucson, AZ, USA, 2012. [Google Scholar]
  295. Kyeremeh, K.; Owusu, K.B.; Ofosuhene, M.; Ohashi, M.; Agyapong, J.; Camas, A.S.; Camas, M. Anti-Proliferative and Anti-Plasmodia Activity of Quinolactacin A2, Citrinadin A and Butrecitrinadin co-isolated from a Ghanaian Mangrove Endophytic Fungus Cladosporium oxysporum strain BRS2A-AR2F. J. Chem. Appl. 2017, 3, 12. [Google Scholar]
  296. Qin, J.-C.; Zhang, Y.-M.; Gao, J.-M.; Bai, M.-S.; Yang, S.-X.; Laatsch, H.; Zhang, A.-L. Bioactive metabolites produced by Chaetomium globosum, an endophytic fungus isolated from Ginkgo biloba. Bioorganic Med. Chem. Lett. 2009, 19, 1572–1574. [Google Scholar] [CrossRef]
  297. Kim, S.; Shin, D.S.; Lee, T.; Oh, K.B. Periconicins, two new fusicoccane diterpenes produced by an endophytic fungus Periconia sp. with antibacterial activity. J. Nat. Prod. 2004, 67, 448–450. [Google Scholar] [CrossRef]
  298. Dai, J.; Krohn, K.; Flörke, U.; Draeger, S.; Schulz, B.; Kiss-Szikszai, A.; Antus, S.; Kurtán, T.; van Ree, T. Metabolites from the endophytic fungus Nodulisporium sp. from Juniperus cedre. Eur. J. Org. Chem. 2006, 2006, 3498–3506. [Google Scholar] [CrossRef]
  299. 299. Han, Z.; Mei, W.; Cui, H.; Zeng, Y.; Lin, H.; Hong, K.; Dai, H. Antibacterial constituents from the endophytic fungus Penicillium sp. of mangrove plant Cerbera manghas. Chem. J. Chin. Univ. 2008, 29, 752. [Google Scholar]
  300. Lu, H.; Zou, W.X.; Meng, J.C.; Hu, J.; Tan, R.X. New bioactive metabolites produced by Colletotrichum sp., an endophytic fungus in Artemisia annua. Plant Sci. 2000, 151, 67–73. [Google Scholar] [CrossRef]
  301. Dissanayake, R.K.; Ratnaweera, P.B.; Williams, D.E.; Wijayarathne, C.D.; Wijesundera, R.L.C.; Andersen, R.J.; de Silva, E.D. Antimicrobial activities of endophytic fungi of the Sri Lankan aquatic plant Nymphaea nouchali and chaetoglobosin A and C, produced by the endophytic fungus Chaetomium globosum. Mycology 2016, 7, 1–8. [Google Scholar] [CrossRef]
  302. Wang, F.W.; Jiao, R.H.; Cheng, A.B.; Tan, S.H.; Song, Y.C. Antimicrobial potentials of endophytic fungi residing in Quercus variabilis and brefeldin A obtained from Cladosporium sp. World J. Microbiol. Biotechnol. 2007, 23, 79–83. [Google Scholar] [CrossRef]
  303. Mousa, W.K.; Raizada, M.N. The diversity of anti-microbial secondary metabolites produced by fungal endophytes: An interdisciplinary perspective. Front. Microbiol. 2013, 4, 65. [Google Scholar] [CrossRef]
  304. Kim, I.H.; Takashima, S.; Hitotsuyanagi, Y.; Hasuda, T.; Takeya, K. New Quassinoids, Javanicolides C and D and Javanicosides B−F, from Seeds of Brucea javanica. J. Nat. Prod. 2004, 67, 863–868. [Google Scholar] [CrossRef]
  305. Singh, M.P.; Janso, J.E.; Luckman, S.W.; Brady, S.F.; Clardy, J.; Greenstein, M.; Maiese, W.M. Biological activity of guanacastepene, a novel diterpenoid antibiotic produced by an unidentified fungus CR115. J. Antibiot. 2000, 53, 256–261. [Google Scholar] [CrossRef]
  306. Huang, Z.; Cai, X.; Shao, C.; She, Z.; Xia, X.; Chen, Y.; Yang, J.; Zhou, S.; Lin, Y. Chemistry and weak antimicrobial activities of phomopsins produced by mangrove endophytic fungus Phomopsis sp. ZSU-H76. Phytochemistry 2008, 69, 1604–1608. [Google Scholar] [CrossRef]
  307. Wicklow, D.T.; Roth, S.; Deyrup, S.T.; Gloer, J.B. A protective endophyte of maize: Acremonium zeae antibiotics inhibitory to Aspergillus flavus and Fusarium verticillioides1 1Dedicated to John Webster on the occasion of his 80th birthday. Mycol. Res. 2005, 109, 610–618. [Google Scholar] [CrossRef]
  308. Wicklow, D.T.; Poling, S.M. Antimicrobial activity of pyrrocidines from Acremonium zeae against endophytes and pathogens of maize. Phytopathology 2009, 99, 109–115. [Google Scholar] [CrossRef]
  309. Rukachaisirikul, V.; Sommart, U.; Phongpaichit, S.; Sakayaroj, J.; Kirtikara, K. Metabolites from the endophytic fungus Phomopsis sp. PSU-D15. Phytochemistry 2008, 69, 783–787. [Google Scholar] [CrossRef]
  310. Phongpaichit, S.; Rungjindamai, N.; Rukachaisirikul, V.; Sakayaroj, J. Antimicrobial activity in cultures of endophytic fungi isolated from Garcinia species. FEMS Immunol. Med. Microbiol. 2006, 48, 367–372. [Google Scholar] [CrossRef]
  311. Noble, H.M.; Langley, D.; Sidebottom, P.J.; Lane, S.J.; Fisher, P.J. An echinocandin from an endophytic Cryptosporiopsis sp. and Pezicula sp. in Pinus sylvestris and Fagus sylvatica. Mycol. Res. 1991, 95, 1439–1440. [Google Scholar] [CrossRef]
  312. Schulz, B.; Sucker, J.; Aust, H.J.; Krohn, K.; Ludewig, K.; Jones, P.G.; Döring, D. Biologically active secondary metabolites of endophytic Pezicula species. Mycol. Res. 1995, 99, 1007–1015. [Google Scholar] [CrossRef]
  313. Tejesvi, M.; Segura, D.; Schnorr, K.; Sandvang, D.; Mattila, S.; Olsen, P.; Neve, S.; Kruse, T.; Kristensen, H.; Pirttilä, A.M. An antimicrobial peptide from endophytic Fusarium tricinctum of Rhododendron tomentosum Harmaja. Fungal Divers. 2013, 60, 153–159. [Google Scholar] [CrossRef]
  314. Cui, H.B.; Mei, W.L.; Miao, C.D.; Lin, H.P.; Hong, K.; Dai, H.F. Antibacterial constituents from the endophytic fungus Penicillium sp. 0935030 of mangrove plant Acrostichum aureurm. Chin. J. Antibiot. 2008, 33, 407–410. [Google Scholar]
  315. Kjer, J.; Wray, V.; Edrada-Ebel, R.; Ebel, R.; Pretsch, A.; Lin, W.; Proksch, P. Xanalteric acids I and II and related phenolic compounds from an endophytic Alternaria sp. isolated from the mangrove plant Sonneratia alba. J. Nat. Prod. 2009, 72, 2053–2057. [Google Scholar] [CrossRef]
  316. Aly, A.H.; Edrada-Ebel, R.; Wray, V.; Müller, W.E.; Kozytska, S.; Hentschel, U.; Proksch, P.; Ebel, R. Bioactive metabolites from the endophytic fungus Ampelomyces sp. isolated from the medicinal plant Urospermum picroides. Phytochemistry 2008, 69, 1716–1725. [Google Scholar] [CrossRef]
  317. Brady, S.F.; Bondi, S.M.; Clardy, J. The guanacastepenes: A highly diverse family of secondary metabolites produced by an endophytic fungus. J. Am. Chem. Soc. 2001, 123, 9900–9901. [Google Scholar] [CrossRef]
  318. Brady, S.F.; Singh, M.P.; Janso, J.E.; Clardy, J. Guanacastepene, a fungal-derived diterpene antibiotic with a new carbon skeleton. J. Am. Chem. Soc. 2000, 122, 2116–2117. [Google Scholar] [CrossRef]
  319. Souza, J.J.d.; Vieira, I.J.C.; Rodrigues-Filho, E.; Braz-Filho, R. Terpenoids from Endophytic Fungi. Molecules 2011, 16, 10604–10618. [Google Scholar] [CrossRef]
  320. Fadiji, A.E.; Babalola, O.O. Elucidating Mechanisms of Endophytes Used in Plant Protection and Other Bioactivities With Multifunctional Prospects. Front. Bioeng. Biotechnol. 2020, 8, 467. [Google Scholar] [CrossRef]
  321. Wani, M.C.; Taylor, H.L.; Wall, M.E.; Coggon, P.; McPhail, A.T. Plant antitumor agents. VI. Isolation and structure of taxol, a novel antileukemic and antitumor agent from Taxus brevifolia. J. Am. Chem. Soc. 1971, 93, 2325–2327. [Google Scholar] [CrossRef]
  322. Yang, X.; Strobel, G.; Stierle, A.; Hess, W.M.; Lee, J.; Clardy, J. A fungal endophyte-tree relationship: Phoma sp. in Taxus wallachiana. Plant Sci. 1994, 102, 1–9. [Google Scholar] [CrossRef]
  323. Strobel, G.; Daisy, B. Bioprospecting for Microbial Endophytes and Their Natural Products. Microbiol. Mol. Biol. Rev. 2003, 67, 491–502. [Google Scholar] [CrossRef]
  324. Schulz, B.; Boyle, C.; Draeger, S.; Römmert, A.-K.; Krohn, K. Endophytic fungi: A source of novel biologically active secondary metabolites* *Paper presented at the British Mycological Society symposium on Fungal Bioactive Compounds, held at the University of Wales Swansea on 22–27 April 2001. Mycol. Res. 2002, 106, 996–1004. [Google Scholar] [CrossRef]
  325. Wiyakrutta, S.; Sriubolmas, N.; Panphut, W.; Thongon, N.; Danwisetkanjana, K.; Ruangrungsi, N.; Meevootisom, V. Endophytic fungi with anti-microbial, anti-cancer and anti-malarial activities isolated from Thai medicinal plants. World J. Microbiol. Biotechnol. 2004, 20, 265–272. [Google Scholar] [CrossRef]
  326. Strobel, G.A. Microbial gifts from rain forests1. Can. J. Plant Pathol. 2002, 24, 14–20. [Google Scholar] [CrossRef]
  327. Redman, R.S.; Sheehan, K.B.; Stout, R.G.; Rodriguez, R.J.; Henson, J.M. Thermotolerance generated by plant/fungal symbiosis. Science 2002, 298, 1581. [Google Scholar] [CrossRef]
  328. Arnold, A.E.; Mejía, L.C.; Kyllo, D.; Rojas, E.I.; Maynard, Z.; Robbins, N.; Herre, E.A. Fungal endophytes limit pathogen damage in a tropical tree. Proc. Natl. Acad. Sci. USA 2003, 100, 15649–15654. [Google Scholar] [CrossRef]
  329. Bae, H.; Sicher, R.C.; Kim, M.S.; Kim, S.-H.; Strem, M.D.; Melnick, R.L.; Bailey, B.A. The beneficial endophyte Trichoderma hamatum isolate DIS 219b promotes growth and delays the onset of the drought response in Theobroma cacao. J. Exp. Bot. 2009, 60, 3279–3295. [Google Scholar] [CrossRef]
  330. Akello, J.; Dubois, T.; Gold, C.S.; Coyne, D.; Nakavuma, J.; Paparu, P. Beauveria bassiana (Balsamo) Vuillemin as an endophyte in tissue culture banana (Musa spp.). J. Invertebr. Pathol. 2007, 96, 34–42. [Google Scholar] [CrossRef]
  331. Giordano, L.; Gonthier, P.; Varese, G.C.; Miserere, L.; Nicolotti, G. Mycobiota inhabiting sapwood of healthy and declining Scots pine (Pinus sylvestris L.) trees in the Alps. Fungal Divers. 2009, 38, 69–83. [Google Scholar]
  332. Saikkonen, K.; Faeth, S.H.; Helander, M.; Sullivan, T.J. Fungal Endophytes: A Continuum of Interactions with Host Plants. Annu. Rev. Ecol. Syst. 1998, 29, 319–343. [Google Scholar] [CrossRef]
  333. Verma, S.K.; Lal, M.; Das, M.D. Structural Elucidation of Bioactive Secondary Metabolites from Endophytic Fungus. Asian J. Pharm. Clin. Res. 2017, 10, 395–400. [Google Scholar] [CrossRef]
  334. Tuntiwachwuttikul, P.; Taechowisan, T.; Wanbanjob, A.; Thadaniti, S.; Taylor, W.C. Lansai A–D, secondary metabolites from Streptomyces sp. SUC1. Tetrahedron 2008, 64, 7583–7586. [Google Scholar] [CrossRef]
  335. Castillo, U.F.; Strobel, G.A.; Ford, E.J.; Hess, W.M.; Porter, H.; Jensen, J.B.; Albert, H.; Robison, R.; Condron, M.A.M.; Teplow, D.B.; et al. Munumbicins, wide-spectrum antibiotics produced by Streptomyces NRRL 30562, endophytic on Kennedia nigriscans. Microbiology 2002, 148, 2675–2685. [Google Scholar] [CrossRef]
  336. Agatonovic-Kustrin, S.; Doyle, E.; Gegechkori, V.; Morton, D.W. High-performance thin-layer chromatography linked with (bio)assays and FTIR-ATR spectroscopy as a method for discovery and quantification of bioactive components in native Australian plants. J. Pharm. Biomed. Anal. 2020, 184, 113208. [Google Scholar] [CrossRef]
  337. Biva, I.J.; Ndi, C.P.; Griesser, H.J.; Semple, S.J. Antibacterial constituents of Eremophila alternifolia: An Australian aboriginal traditional medicinal plant. J. Ethnopharmacol. 2016, 182, 1–9. [Google Scholar] [CrossRef]
  338. Zhang, H.W.; Song, Y.C.; Tan, R.X. Biology and chemistry of endophytes. Nat. Prod. Rep. 2006, 23, 753–771. [Google Scholar] [CrossRef]
  339. Xing, Y.-M.; Chen, J.; Cui, J.-L.; Chen, X.-M.; Guo, S.-X. Antimicrobial Activity and Biodiversity of Endophytic Fungi in Dendrobiumdevonianum and Dendrobium thyrsiflorum from Vietman. Curr. Microbiol. 2011, 62, 1218–1224. [Google Scholar] [CrossRef]
  340. Dang, L.; Li, G.; Yang, Z.; Luo, S.; Zheng, X.; Zhang, K. Chemical constituents from the endophytic fungus Trichoderma ovalisporum isolated from Panax notoginseng. Ann. Microbiol. 2010, 60, 317–320. [Google Scholar] [CrossRef]
  341. Soares, R.D.F.; Campos, M.G.N.; Ribeiro, G.P.; Salles, B.C.C.; Cardoso, N.S.; Ribeiro, J.R.; Souza, R.M.; Leme, K.C.; Soares, C.B.; de Oliveira, C.M. Development of a chitosan hydrogel containing flavonoids extracted from Passiflora edulis leaves and the evaluation of its antioxidant and wound healing properties for the treatment of skin lesions in diabetic mice. J. Biomed. Mater. Res. Part A 2020, 108, 654–662. [Google Scholar] [CrossRef]
  342. Azzazy, H.M.; Fahmy, S.A.; Mahdy, N.K.; Meselhy, M.R.; Bakowsky, U. Chitosan-Coated PLGA Nanoparticles Loaded with Peganum harmala Alkaloids with Promising Antibacterial and Wound Healing Activities. Nanomaterials 2021, 11, 2438. [Google Scholar] [CrossRef]
Figure 1. Distribution of the wound infections by: (a) the Gram-negative bacteria and (b) the Gram-positive bacteria.
Figure 1. Distribution of the wound infections by: (a) the Gram-negative bacteria and (b) the Gram-positive bacteria.
Pharmaceutics 15 00644 g001
Figure 2. Synthesis scheme of polyvinyl alcohol/starch hydrogel membranes.
Figure 2. Synthesis scheme of polyvinyl alcohol/starch hydrogel membranes.
Pharmaceutics 15 00644 g002
Figure 3. Preparation technique of alginate hydrocolloids using hot melt coating.
Figure 3. Preparation technique of alginate hydrocolloids using hot melt coating.
Pharmaceutics 15 00644 g003
Figure 4. Preparation scheme of chitosan-based bioactive foams.
Figure 4. Preparation scheme of chitosan-based bioactive foams.
Pharmaceutics 15 00644 g004
Figure 5. Preparation process of Aloe vera-containing alginate films.
Figure 5. Preparation process of Aloe vera-containing alginate films.
Pharmaceutics 15 00644 g005
Figure 6. Process scheme of dermal patches containing natural additives.
Figure 6. Process scheme of dermal patches containing natural additives.
Pharmaceutics 15 00644 g006
Figure 7. Preparation technique of CeO2-PM oil-PEO/GO nanofibrous mats.
Figure 7. Preparation technique of CeO2-PM oil-PEO/GO nanofibrous mats.
Pharmaceutics 15 00644 g007
Figure 8. Preparation scheme of essential oil-based polycaprolactone membranes.
Figure 8. Preparation scheme of essential oil-based polycaprolactone membranes.
Pharmaceutics 15 00644 g008
Figure 9. Preparation steps of chitosan/essential oil nanoparticles.
Figure 9. Preparation steps of chitosan/essential oil nanoparticles.
Pharmaceutics 15 00644 g009
Figure 10. Essential oils processing units.
Figure 10. Essential oils processing units.
Pharmaceutics 15 00644 g010
Figure 11. Extraction process of antibacterial compounds from endophytes.
Figure 11. Extraction process of antibacterial compounds from endophytes.
Pharmaceutics 15 00644 g011
Table 1. Different types of wound dressings, their wound target, and polymer type.
Table 1. Different types of wound dressings, their wound target, and polymer type.
VarietyDescriptionAdvantagesDisadvantagesWound Type ApplicationPolymerRef.
HydrogelsWater-absorbent cross-linked polymeric networks resulting from the reaction of monomersEfficient flexibility, good ability in swelling and sustaining a significant amount of water, moisturizing, removal of necrotic tissue, good porosity, and monitoring the wound without removing the dressingInability to absorb enough exudates leading to bacterial proliferation, and low mechanical strengthChemotherapy peelsPolyethylene oxide, polyvinyl pyrrolidine, Polyvinyl alcohol[10,11,12,13,14]
Ulcers
Laser resurfacing
Average thickness wounds
Donor graft sites and artificial organ wounds
HydrocolloidsColloidal material (gel) constituted with elastomers and adhesives in the form of films or sheetsExcellent exudate absorption properties, transparency, enhanced angiogenesis, and formation of granulation tissueNot permeable to gas, vapor, water, and bacteria, their debriding capability, skin maceration, and producing a foul smellChronic ulcersPectin, carboxymethylcellulose, gelatin, and cellulose[10,13,15,16]
Burns
Average thickness wounds
Donor graft sites
FoamsA porous structure using capillary action as its mechanism to absorb fluidsExudate absorbance, preventing bacteria invasion, maintaining sufficient moisture at the wound surface, being removed easily, protecting the skin around the wound, maintaining an efficient temperature, mechanical protection, being nontoxic, being cost-effective with a long shelf lifeDrying out the wound in case of minimal or no exudate presence and maceration of the surrounding skin in case of exudate saturation in dressingChronic woundsPolyurethane, silicone, silk fibroin[13,17,18,19,20,21]
Burns
Mohs surgery and wounds
Laser resurfacing wounds
FilmsConsists of adhesives, porous, and thin transparent polymersThe possibility of having a high mechanical strength, high water transmission rate, protecting the wound against bacterial infectionThe possibility of having a low mechanical strengthSuperficial woundsSoy protein isolates, chitosan, polyvinyl alcohol[13,22,23,24]
Laser wounds
Surgery defect sites
Skin tears
Dermal patchesDressings consisted of a multilayered structure with an impermeable excipient-loaded film, drugs, and a release linerSuitable for skin adhesion, not having a liquid reservoir, controlling the drug delivery rateNeeding flux moderation in case of loading with highly soluble drugs, and decrease in drug release rate with wear time, not suitable for most of the drugsHypertensionPoly(vinyl pyrrolidones), poly(vinyl alcohol)[25,26,27,28,29,30]
Topical wounds
Fibers and nanofibersPolymeric fibers produced with electrospinning processExcellent mechanical properties, thermal stability, antimicrobial activity, biodegradability, control in water vapor transmission rate, oxygen permeability, fluid drainage ability, high porosity, and high surface areaHigher cost of production in some cases, hard to produce fibers with diameters less than 10 nmPartial thickness burnsPolyurethane, collagen, silk fibroin, polycaprolactone, poly (lactic-co-glycolic acid), polyethylene oxide, etc.[31,32,33,34,35,36,37,38,39,40]
Diabetic ulcers
Bone bleeding
Chronic infected wounds
Acute wounds
Venous ulcers
Pressure ulcers
Membranes A thin semi-permeable barrierPorous structure, transparency, excessive loss of water, the ability to contain an occlusive layer to impede microbial invasionCytotoxicity in some casesSuperficial woundsPectin, collagen, chitosan, chitin, alginate, zein, polycaprolactone, polyvinyl acetate, polyvinyl alcohol, polytetrafluoroethylene, cellulose, etc.[41,42,43,44,45,46,47,48,49,50,51,52]
Frictional wounds
Skin-scratching wounds
Skin donor sites
Skin with external contamination
Polymer-drug conjugatesPolymer-based water-soluble nanocarriers conjugated with bioactive agentsImproving the water solubility of the hydrophobic drugs, enhancing the pharmacokinetic profile of the conjugated drug, extending the volume of distribution, and protecting the conjugated drug against degradationLimitations to be applied on a large scale, low stability in vivo, short half-life, and immunogenicityDiabetic wounds such as venous leg and lower limb ulcersN-(2-hydroxypropyl) methacrylamide copolymer, polyglutamic acid, Poly(ethylene glycol), Polyamidoamine, hyaluronic acid, poly (vinyl ether-co-maleic anhydride), poly (vinyl pyrrolidone), etc.[53,54,55,56,57,58,59,60,61]
Table 2. Plant species and their activity in wound dressing.
Table 2. Plant species and their activity in wound dressing.
Scientific NameClasses of Chemical CompoundsCommon Medical UsesSolvents Used for ExtractionActivity against Infected Wound BacteriaReferences
Acalipha alinifolia/fruticosaPhenolic compounds, cardiac glycosides, tannins, flavonoids, and phytosterolsAnti-bacterial, antifungal, antioxidant, and anthelmintic propertiesAqueous, acetone, and methanolS. aureus[157,158,159,160]
asthma, pneumonia, scabies, and skin diseasesP. aeruginosa
Delonix elataSaponins, tannins, flavonoids, and steroidsanti-inflammatory, anti-arthritic, and antioxidantAqueous, ethanol, chloroform, acetone, petroleum ether, and methanolS. aureus[161,162,163,164,165]
E. coli
P. aeruginosa
Delonix regiaFlavonoids, alkaloids, terpenoids, steroids, and phenolic acidsAnti-diarrhoeal, anti-inflammatory, antidiabetic, antioxidant, hepatoprotective, antimicrobial, anthelmintic, wound healing, gastroprotectiveAqueous, MethanolE. coli[166,167,168,169,170,171,172,173,174,175,176,177,178,179]
P. aeruginosa
S. aureus
Klebsiella pneumoniae
Digera muricataPhenol, flavonoids, alkaloids, terpenes, sterols, tannins, glycosides, and ligninsAntibacterial, antifungal, diuretic, laxative, free radical scavenger activity, anthelminticPetroleum ether, chloroform, ethanol, distilled waterE. coli[180,181,182]
S. aureus
Hygrophilia auriculataAlkaloids, terpenoids, tannins, flavonoids, and fatty acidsMedicinal usage in Indian AyurvedaDistilled water, 50% aqueous ethanol, methanol, petroleum ether, chloroform, diethyl etherP. aeruginosa[183,184,185,186,187,188,189,190,191,192]
S. aureus
E. coli
K. pneumoniae
Maerua oblongifoliaAlkaloids, terpenoids carbohydrates, glycosides, phytosterols, saponins, proteins, and amino acidsWound healing activity, treating toothache, the roots of this plant possess alternative, tonic, and medicinal propertiesPetrol-Et2-MeOH (1:1:1), Dichloromethane/methanol, aqueousE. coli[193,194,195,196]
S. aureus
K. pneumoniae
Pterocarpus santalinusAlkaloids, phenols, saponins, glycosides, flavonoids, triterpenoids, sterols, and tanninsAntipyretic, anti-inflammatory, anthelmintic, tonic, haemorrhage, dysentery, aphrodisiac, anti-hyperglycaemic and diaphoretic70% methanolS. aureus[197,198,199]
P. aeruginosa
E. coli
Syzygium cuminiFlavonoids, glucoside derivatives, and phenolsDiabetes, sores and ulcers, leucorrhoea, and antidote in opium poisoningMethanol, aqueousP. aeruginosa[200,201,202,203]
E. coli
S. aureus
Gyrocarpus americanusAlkaloidsUnknown medicinal valuesEthanol, methanol, waterNA[204,205,206]
Punica granatumPolyphenols, sterols, triterpenoids, flavonoids, fatty acids, and tanninsAnti-inflammatory, anti-cancer, antioxidant, and antibacterial activityMethanol, petroleum ether, chloroform, aqueous, chloramphenicolS. aureus[207,208,209,210,211,212,213]
E. coli
K. pneumoniae
Euphorbia heterophillaFlavonoids, saponins, diterpenes, and phorbol estersWound healing activity, used for the treatment of constipation, bronchitis, and asthma, anti-inflammatory activityAqueous, petroleum ether, Butanol, ethanolS. aureus[214,215,216,217,218,219,220]
E. coli
K. pneumoniae
P. aeruginosa
Table 3. Application and activity of antimicrobial essential oils.
Table 3. Application and activity of antimicrobial essential oils.
PlantChemical ConstituentsActivity and UseActivity against Infected Wound BacteriaReferences
Clove (Syzygium aromaticum L.)Eugenol, acetyleugenol, thymol, cinnamaldehyde, etc.Anti-inflammatory, antibacterial, antifungal, anti-allergic, anti-carcinogenic, anti-mutagenicP. aeruginosa[230,231,232,233,234,235]
S. aureus
K. pneumoniae
Rosemary (Rosmarinus officinalis L.)A-pinene, myrcene, 1,8-cineole, borneol, and camphorAntioxidant, antimicrobialS. aureus[236,237,238]
K. pneumoniae
E. coli
Fennel (Foeniculum vulgare Mill.)Trans-anethole, estragoleHepatoprotective, antioxidant, anti-inflammatory, antidiabetic, antitumor, and acaricidalS. aureus[239,240,241,242,243,244,245,246]
E. coli
P. aeruginosa
Tea tree (Melaleuca alternifolia)Terpinen-4-ol, 1,8-Cineol, α-Pinene, α-Terpineol, Sabinene,Antibacterial, antifungal, used for skin treatment, airway treatment, oral treatment, and vaginal infectionsS. aureus[247,248,249]
E. coli
P. aeruginosa
Cinnamon (Cinnamomum cassia)Cinnamaldehyde, (−)-α-Pinene/Ylangene, terpenes, aldehydes, etc.Antibacterial, antioxidant, wound healing applicationsS. aureus[250,251,252]
E. coli
P. aeruginosa
Thyme (Thymus vulgaris L.)1R-α-pinene, o-cymol, 4-carene, β-linalool, Camphor, Thymol, CarvacrolAntioxidant, antibacterialS. aureus[253,254]
E. coli
OreganoLinalool, Thymol, Carvacrol, Ethyl caprate, etc.Antioxidant, antibacterial, anti-inflammatoryS. aureus[253,255,256,257]
E. coli
Basil (Ocimum basilicum L.)Linalool, 1,8-cineole, aromadendrene, and transcaryophylleneAntibacterial and antioxidantS. aureus[258,259,260]
K. pneumoniae
E. coli
Shigella flexneri
Orange (Citrus sinensis)Limonene, alcohol compounds, carvone, β-myrceneAntibacterialE. coli[261,262]
P. aeruginosa
S. aureus
Peppermint (Mentha × piperita L.)Menthol, Menthone, Limonene, β-pinene, α-pinene, Menthyl acetate etc.Antibacterial, antifungalS. aureus[263,264]
E. coli
Juniperus chinensis L. Bornyl acetate, sabinene, trans-sabinyl acetate, carotol, elemolAir cleaning effect and antibacterial activity (mainly against Propionibacterium acnes), used for skincare and cleansingE. coli[265]
P. aeruginosa
S. aureus
Camellia oilTriterpenes, Sesquiterpenes, tocopherols, phthalate esters, and cannabinoidSkin-moisturizing effect, skin-soothing effect; effective against atopic or allergic skin conditions (effects due to high amount of oleic acid); prevention of skin dryness and alleviating itching (due to the gamma-linolenic acid content)E. coli[266,267]
Bacillus cereus
C. albicans
Dendropanax speciesɤ-elemene, tetramethyltricyclo hydrocarbons, β-Selinene and β-ZingibereneAntioxidant and antimicrobial or antibacterial activitiesS. aureus[268,269]
Bacillus. cereus
Portulaca oleracea L. Phenolic compounds, α-linolenic acid (ω3)Alleviate skin irritation and allergic responses, antibacterial and anti-inflammatory effects/it is used mainly in acne care products and cosmetics for sensitive skinS. aureus[221,270]
Klebsiella oxytoca
Houttuynia cordata Thunb. 3-oxododecanal (Hou), 2- undecanone, pinene, camphene, myrcene, limoneneEffective in treating skin inflammation and atopic diseasesS. aureus[271,272]
Glycyrrhiza glabra L. α-pinene, β-pinene, octanol, γ-terpinene, stragole, isofenchon, β-caryophyllene, citronellyl acetate, caryophyllene oxide, and geranyl hexanolateDetoxifying and anti-inflammatory effects, effective in alleviating skin diseases such as acne, atopy, eczema, urticarialE. coli[273,274]
S. aureus
Ziziphus jujuba Mill. Triterpenoid acids, alkaloids, saponins, flavonoids, and their glycosidesEffective in moisturizing the skin and keeping the skin healthyE. coli[275,276]
S. aureus
P. aeruginosa
Chamaecyparis obtuse (Siebold & Zucc.) Endl. δ-cadinene, α-pinene, γ-cadinene, α-cedrol, α-muurolene, γ-eudesmol, γ-muurolene, α-elemene and α-copaeneSterilising effect (due to the high content of phytoncide)S. aureus[277,278]
Table 4. Bioactive compounds produced by antibacterial endophytic fungi.
Table 4. Bioactive compounds produced by antibacterial endophytic fungi.
Types of Endophytic CompoundsEndophyteHost PlantMain BioactivityActivity against Wound Infection BacteriaReferences
Aliphatic compoundsChaetomium globosumGinkgo bilobaAntifungal, antibacterialS. aureus[296,301]
Cladosporium sp.Quercus variabilisAntifungal, antimicrobialNA[302]
Fungal endophytesChinese herbsAntimicrobial, antibacterialKlebsiella pneumonia[300,303,304,305]
S. aureus
P. aeruginosa
Phomopsis sp.Excoecaria agallochaAntifungal, antimicrobialS. aureus[306]
E. coli
AlkaloidsAcremonium zeaeMaizeAntifungal, antibacterialPseudomonas fluorescens[307,308]
Enterobacter agglomerans
Phomopsis sp.Garcinia dulcisAntibacterialS. aureus[309,310]
FlavonoidsNodulisporium sp.Juniperus cedreAntifungal, AntibacterialNA[298]
PeptidesCryptosporiopsis sp., Pezicula sp.Pinus sylvestris and Fagus sylvaticaAntifungal, antibacterialE. coli[311,312]
Fusarium tricinctumRhododendron tomentosumAntimicrobialS. aureus[313]
Penicillium sp.Acrostichum aureurmAntifungal, AntibacterialS. aureus[314]
PhenolsAlternaria sp.Sonneratia albaAntibacterialS. aureus[315]
Phoma speciesSaurauia scaberrinaeAntibacterialS. aureus[299]
Penicillium sp.Cerbera manghasAntibacterialS. aureus[299]
QuinonesAmpelomyces sp.Urospermum picroidesAntibacterialS. aureus[316]
SteroidsColletotrichum sp.Artemisia annuaAntifungal, antimicrobialS. aureus[300]
P. aeruginosa
Nodulisporium sp.Juniperus cedreAntifungal, AntibacterialNA[298]
Fungal endophytesDaphnopsis americanaAntibacterialS. aureus[317,318]
Periconia sp.Taxus cuspidateAntibacterialS. aureus[297]
K. pneumoniae
Fungal endophytesNAAntimicrobialS. aureus[319]
P. aeruginosa
E. coli
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

Firoozbahr, M.; Kingshott, P.; Palombo, E.A.; Zaferanloo, B. Recent Advances in Using Natural Antibacterial Additives in Bioactive Wound Dressings. Pharmaceutics 2023, 15, 644. https://doi.org/10.3390/pharmaceutics15020644

AMA Style

Firoozbahr M, Kingshott P, Palombo EA, Zaferanloo B. Recent Advances in Using Natural Antibacterial Additives in Bioactive Wound Dressings. Pharmaceutics. 2023; 15(2):644. https://doi.org/10.3390/pharmaceutics15020644

Chicago/Turabian Style

Firoozbahr, Meysam, Peter Kingshott, Enzo A. Palombo, and Bita Zaferanloo. 2023. "Recent Advances in Using Natural Antibacterial Additives in Bioactive Wound Dressings" Pharmaceutics 15, no. 2: 644. https://doi.org/10.3390/pharmaceutics15020644

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