Next Article in Journal
Epidemiological Evaluation of Events Allegedly Attributable to COVID-19 Vaccination: A Cross-Sectional Study in the Brazilian Amazon
Next Article in Special Issue
Terpenes from Cecropia Species and Their Pharmacological Potential
Previous Article in Journal
Production and Immunogenicity Assessment of LTp50: An Escherichia coli-Made Chimeric Antigen Targeting S1- and S2-Epitopes from the SARS-CoV-2/BA.5 Spike Protein
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Use of Medicinal Plants in the Process of Wound Healing: A Literature Review

by
Mayra Cedillo-Cortezano
,
Luis Ruben Martinez-Cuevas
,
Jesús A. Márquez López
,
Ingrid L. Barrera López
,
Samantha Escutia-Perez
and
Vera L. Petricevich
*
Facultad de Medicina, Universidad Autónoma del Estado de Morelos (UAEM), Cuernavaca 62350, Mexico
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2024, 17(3), 303; https://doi.org/10.3390/ph17030303
Submission received: 30 November 2023 / Revised: 9 January 2024 / Accepted: 10 January 2024 / Published: 27 February 2024

Abstract

:
The literature on the use of medicinal plants in wound healing was comprehensively searched to obtain and assess the data. The data were procured via clinical studies that utilized medicinal plants and their compounds in vitro and in vivo for wound healing. This review collected data from electronic databases, including Google Scholar, PubMed, Science Direct, Web of Science, SciFinder, Thesis, and Scopus, using the search terms “natural products”, “wound healing”, and “natural compounds”, along with the keywords “plants”, “extracts”, and “phytochemicals”. Results from the last decade reveal a total of 62 families and 109 genera of medicinal plants, and their compounds have been studied experimentally both in vivo and in vitro and clinically found to effectively promote healing. This activity is related to the presence of secondary metabolites such as flavonoids, alkaloids, saponins, tannins, terpenoids, and phenolic compounds, which act at different stages through different mechanisms to exert anti-inflammatory, antimicrobial, and antioxidant effects, confirming that the use of medicinal plants could be an adequate alternative to current conventional practices for treating wounds.

Graphical Abstract

1. Introduction

Statistics derived by the World Health Organization (WHO) have shown that around 80% of the world’s population uses traditional medicine for primary healthcare, and 85% of this group utilizes plants. One of the great challenges of modern medicine concerns the healing and treatment of wounds. Studies by the WHO show that around 5 million people die annually because of imperfect wound healing. The use of natural products has shown promise in preventing and treating wounds. This review aims to elucidate the modes of preparation of herbal treatments, their phytochemical contents, and their use in formulations for wounds. Plants show a broad spectrum of bioactive phytochemicals, categorizable into the families of alkaloids, carotenoids, phenolic compounds, steroids, flavonoids, saponins, tannins, and terpenoids. These compounds act at different phases of the healing process through different mechanisms and show anti-inflammatory, antimicrobial, and antioxidant effects, whilst promoting collagen synthesis, cell proliferation, and angiogenesis. The application of natural compounds via new systems can contribute to enhancements in wound treatment.

2. Methods

2.1. Information Sources, Searching, and Selection of Studies

For this review, the period considered was January 2013 to October 2023, and the electronic databases used include Google Scholar, PubMed, Scopus, Science Direct, Web of Science, SciFinder, and Theses. The terms used for the search were “wound healing” (sought in the titles and abstracts) and the keywords “plant”, “extract”, “natural compounds”, and “phytochemicals”. We also manually searched for references to bioactive phytochemicals that act at different phases of the healing process through different mechanisms and have anti-inflammatory, antimicrobial, antioxidant, and cell proliferation-promoting effects. The full documents were read to verify that they met the inclusion criteria.

2.2. Eligibility Criteria

Inclusion criteria: Studies published in English, including theses, articles, and proceedings from January 2013 to 01 October 2023, with “wound healing” in the title or abstract, addressing experimental or clinical studies.
Exclusion criteria: Newspapers and reviews.

2.3. Results

Following the initial screening, we identified approximately 5000 articles in different databases, citing about 480 different genera of plants with healing activities. Only 22% of these were included in this study, in accordance with the eligibility criteria. This is the first study in the last ten years to address plants and major and/or new compounds with regard to their activity in wound healing. The results obtained from eligible studies reveal a total of 62 families and 109 genera of medicinal plants used for wound treatment that have been discussed in studies from the last 10 years. Their effects, attributed to flavonoids, alkaloids, saponins, and phenolic compounds, which act at different stages through different mechanisms, include anti-inflammatory, antimicrobial, and antioxidant effects.

3. Classification of Wounds

The Healing Society defines wounds as physical lesions resulting from an opening or breaking of the skin that causes disturbances within the anatomy and normal functioning of the skin [1,2]. Wound healing can be a complex process because it entails a series of interdependent and overlapping stages: inflammation (exudative phase), reconstruction (proliferative phase), epithelization (regenerative phase), and maturation [3].
Several factors may affect the healing process, including (a) the presence of a contaminated surface contacting the wound; (b) delays due to the consumption of infected nutrients as a source of energy by white blood cells; (c) associated illnesses, such as diabetes and morbid obesity, which cause hyperglycemia and thus impact the defense mechanisms of the body, impairing the capacities of white blood cells in general, and especially neutrophils; and (d) treatment with radio-chemotherapy, NSAIDs and immunosuppressive drugs [4,5,6]. Wounds can be classified in various ways based on their etiology, their position, the kind of injury, the associated changes in bodily function, the wound depth, tissue loss, or clinical appearance. Table 1 describes the classification of a wound.

3.1. Healing Process

Wounds can change the physiology of the skin, particularly those that affect the dermal layer. Therefore, tissue lesions can modify the anatomical structure of the skin, and the degree of damage to the tissue is highly dependent upon the healing mechanism. The wound-healing process entails a cascade of cellular and molecular events aimed at restoring the injured area [7].
The healing cascade is an organized sequence of events, and classifications have been applied to it to facilitate our understanding of the dynamic processes it involves that closely determine healing [8,9]. Different authors divide up the healing process in different ways; some consider the initial step to involve inflammation, followed by proliferation and ending with repair in the remodeling stage [7,8,9] (Figure 1).

3.2. Inflammation

Inflammation is a defensive reaction to harmful agents, including microorganisms and damaged cells produced by the body, and promotes biological processes such as vascular responses and systemic reactions intended to reestablish the equilibrium of tissue homeostasis. In the absence of the inflammatory process, infections would develop in an uncontrolled manner; thus, the destructive processes unfolding in organs would continue until a total loss of function. The inflammatory process can be assessed clinically using five classical signs, called the “Cardinal Signals”: swelling, heat, redness, pain, and loss of function [10,11].
The major function of the inflammatory response is the conduction of leukocytes to the affected region, which play an important role in defense by phagocytizing or producing substances that destroy microorganisms and necrotic tissues; they can inactivate or degrade antigens. Circulating cells such as neutrophils, monocytes, lymphocytes, and eosinophils reach the site of aggression through the bloodstream, crossing the vessel wall and migrating toward the site of aggression in significant quantities during the first 24 h of injury. The circulating cells are attracted by the notable inflammatory cytokine effects produced by activated platelets, endothelial cells, and the degraded products of the pathogens present in the lesion [12,13].
These chemical mediators can cause the dilation of the arterioles and increase the permeability of capillaries and venules, allowing a greater flow of blood to the damaged area, as well as the exudation (extravasation) of liquids, proteins, and defense cells into the interstitial space. Fluid exudation results in inflammatory edema, whereby the blood gradually becomes more viscous due to the increased density of red blood cells, and the circulation of the small vessels gradually slows, ultimately culminating in blood stasis (small, dilated vessels filled with red blood cells). At the same time, leukocytes migrate through the vascular wall into the interstitial space (called transmigration, diapedesis, or leukocyte emigration); this constitutes the initial (acute) phase of any type of inflammatory response, also referred to as the vascular phenomena [12,14,15].
The recruitment and activation of inflammatory cells, in either acute or chronic inflammation, are consequences of physical changes occurring at the wound site. The first of these changes is the liberation of substances following platelet degranulation. These include thrombin, which prompts the release of distinct growth factors such as platelet-derived growth factors (PDGFs), transforming growth factor-β (TGF-β), epidermal growth factor (EGF), transforming growth factor-α (TGF-α), and endothelial cell growth factor (VEGF). The mediators mentioned beforehand include adhesive glycoproteins such as fibronectin and thrombospondin—important constituents of the extracellular matrix [16].
The inflammatory phase of wound healing includes the activation of vasoactive substances such as serotonin, bradykinin, prostaglandins, and histamine. These can increase the permeability of the endothelium at the lesion site and enhance interstitial fluid perfusion in this area. The increase in permeability facilitates infiltration by immune and repair cells that facilitate the previously described events, while the increase in circulation leads to greater oxygen distribution in the tissue; consequently, the temperature increases at the site of injury. The warm and humid microenvironment thus produced within the wound is essential for the ensuing healing phase. At the end of the inflammatory stage of wound healing, macrophages synthesize distinct growth factors, such as PDGF, TGF-β, fibroblast growth factor (FGF), and VEGF, which stand out as the major cytokines required to stimulate the formation of granulation tissue and thus generate the environment required for the next phases of cell proliferation and repair [1,17].

3.3. Proliferation

The closure of the lesion occurs in the proliferation phase. Angiogenesis occurs as a result of the formation of granulation tissue and is responsible for filling the injured tissue. The new extracellular matrix that will be involved in cell growth and the new blood vessels that will convey oxygen and nutrients indispensable for local cellular metabolism are produced with the aid of fibroblasts. With the progression of the proliferative phase, the provisional matrix changes as a result of the newly formed granulation tissue. Wound epithelialization represents the final stage of the proliferative phase [18,19].
The formation of the extracellular matrix provides a substrate for cell adhesion and regulates the growth, movement, and differentiation of cells within it. The extracellular matrix consists of structural proteins, including collagen and elastin, along with an interstitial matrix composed of adhesive glycoproteins, proteoglycan, and glycosaminoglycan [20,21].
The increase in microvascular permeability, characteristic of the inflammatory process, represents the first stage of this proliferative process; here, cellular elements, along with cytokines, are released, and we also see the formation of the provisional extracellular matrix necessary for the migration and proliferation of endothelial cells [22].
Angiogenesis is an exceedingly important stage in the healing process, during which new blood vessels are formed from preexisting vessels. The new vessels participate in the formation of provisional granulation and the supply of nutrients and oxygen to the growing tissue. On the other hand, vasculogenesis refers to the early stages of vascular development, during which vascular endothelial precursor cells enact the mobilization of endothelial progenitors derived from bone marrow [23].
Fibroblast migration is induced by the PDGF and TGF-β released by macrophages. When fibroblasts reach the wound bed, they proliferate and produce matrix proteins such as fibronectin, collagen, and proteoglycans. These components help build the new extracellular matrix, which supports the further growth of cells essential for the repair process. A crucial interaction takes place between the fibroblasts and extracellular matrix, which regulates the additional synthesis of the components along with tissue remodeling [24].
The process of re-epithelialization in the injured tissue is accelerated by the contraction of the underlying connective tissue, which is responsible for the approximation of the wound’s margins. This contraction is induced by myofibroblasts, activated by TGF-β and PDGF; thus, these myofibroblasts play an important role in wound healing, especially for open lesions. When present in open wounds, these cells produce larger amounts of extracellular matrix components. However, if abnormalities arise in the physiological process, such as delays, this may cause cicatricial defects due to alterations to the differentiation of fibroblasts in myofibroblasts [25].

3.4. Remodeling

The final stage of wound healing involves the remodeling or maturation of granulation tissue into mature connective tissue or scar tissue. The wound-healing process is most potent during this phase. Wound maturation begins during the third week after the wounding and is characterized by an increase in resistance and a controlled decrease in the amount of collagen. This mechanism is characterized by a balance in the production and destruction of collagen fibers, resulting from the action of an enzyme called collagenase. An imbalance in this relationship can favor the emergence of hypertrophic and keloid scars [1,2].
Finally, the remodeling process consists of the proper deposit of elements previously mentioned, mainly including collagen fibers. This stage involves a change in the type of collagen present and its disposition. Type III collagen, which is initially more abundant in the wound than type I, is more actively degraded over time; in contrast, the production of collagen I by fibroblasts increases, and this causes an increase in tension force and the reduction in the quantity of collagen [3,4].
Numerous factors can aggravate the wound-healing process, specifically as regards the biological events that comprise it. Factors such as advanced age, the patient’s nutritional status, and vascular changes can directly alter the healing process. However, diabetes mellitus also drastically alters the process of tissue recovery, interfering at all stages and thus causing serious complications for the patient.
The process of wound healing is complicated when the patient is diabetic. In patients with diabetes, wounds show less revascularization and lower expression of growth factors compared to injuries in non-diabetics, thus impairing healing. These complications can evolve to produce severe consequences, such as a stagnant repair mechanism leading to a loss of tissue function [26,27,28].
Impaired healing in diabetic patients is characterized by acute inflammation and abnormalities in angiogenesis, entailing difficulties in forming new blood. The proper healing of a wound requires a regulated inflammatory response; however, diabetic wounds show prolonged inflammatory responses. Wounds in DM1 exhibit increased expression of inflammatory cytokines, including tumor necrosis factor alpha (TNF-α) and interleukins IL-1 and IL-6, and decreased IL-10, leading to injury following a prolonged inflammatory phase. This deregulated and prolonged inflammation leads to the wound becoming chronic and unable to be completely healed [29]. These chronic injuries, such as foot ulcers (diabetic foot), lead to high morbidity and increased treatment costs. In addition, foot ulcers substantiate more than 50% of the cases of amputation among diabetics. Increased oxidative stress is one of the leading causes of wound complications in diabetics, causing late scarring. Reducing persistent inflammation and the excretion of free radicals by incorporating an anti-inflammatory and antioxidant agent into wound treatment has become an important strategy for improving the healing of diabetic wounds [30].
Factors associated with both angiogenesis and the vasculogenesis process are vital for wound healing, as they play a vital role in the delivery of oxygen, nutrients, and other mediators to the wound site. Thus, they have become therapeutic targets that can improve the healing of damaged wounds in diabetes patients when activated, thus restoring the neovasculogenesis mechanism [31].

4. Medicinal Plants Used for Wound Healing

Preparations made using medical plants (such as extracts) and the active compounds present in some of these plants have been used to accelerate wound healing. The ethnopharmacological approach to investigating medicinal plants consists of combining information acquired from users of medicinal flora (traditional communities) with the results of chemical and pharmacological studies [32].
The application of medicinal plants has always been a part of the evolution of humanity; these plants represent one of the first therapeutic resources to be used by humans, and they still hold great importance for the maintenance of human health. According to the World Health Organization (WHO, 2002) [33], approximately 80% of the population in developing countries use traditional medicine as their primary healthcare, most of which entails using plant extracts or their active compounds. According to the statistics provided by the WHO, medicinal plants, herbal preparations, or derived products are conventionally used in primary care in various countries. The WHO classifies a medicinal plant as a plant species that, when administered to humans, exerts a pharmacological action. The findings of ethnopharmacology, in terms of the therapeutic properties of plants and popular knowledge regarding their usage, have been presented as source material for developing technical scientific knowledge. The accumulation of information regarding the use of natural assets by traditional populations has provided researchers with models for the sustainable use of these resources while also providing directions for the exploitation of the pharmacological properties of certain species. Over the centuries, products of plant origin have been commonly used as the basis of treatments for different diseases by virtue of knowledge transmitted down through generations, and certain plant species can be understood as sources of active molecules [34,35,36].
In the context of wound healing, the utilization of plants and plant extracts dates to the prehistoric era [37]. Records describe the use of plants and extracts in the form of poultices to stop hemorrhages and to facilitate cicatrization. Other uses have been described in relation to the ingestion of certain plants, which act systemically [36]. Thus, the data collected through the years confirm that the development of modern medicine has only been possible via the inheritance of ancient healing methods and the empirical knowledge pertaining to such practices [37]. Every year, approximately 100 million patients around the world acquire scars resulting from surgical interventions, burns, or tissue ruptures due to accidents of various kinds, which require effective and rapid treatment. These statistics indicate that wound healing is a modern therapeutic challenge [38]. Multiple studies have sought to improve the treatment of wounds by promoting the healing process; nevertheless, the most effective organic and inorganic substances in this regard remain a scientific mystery to this day [39]. Healing involves several complex processes in which different cellular structures are involved. The process begins with an amplified immune response that prevents wound complications, enacted via chemoattraction, which facilitates the development of other mediators necessary to subsequent phases, such as inflammation, cell proliferation, and re-epithelization, which eventually lead to wound closure [40].
Medicinal plants are significant sources of novel chemical substances with valuable therapeutic effects. Table 2 displays the families and genera of plants utilized for wound healing. A total of 62 families and 109 genera were documented with applicability in wound healing and treatment based on traditional medicine (Table 2). Most of the wound-healing information was collected from recent literature from the last 10 years. The Euphorbiaceae family was the least represented (five members), followed by the Asteraceae family (six members) and the Fabaceae family (eight members). The most commonly used plant parts were cited as leaves (37%), followed by fruits (9%), seeds (8%), roots (8%), aerial parts (7%), flowers (6%), the whole plant (6%) bark (5%), saponins (3%), rhizome (2%) and others. These data also show that medicinal plants are used to treat wounds in many different parts of the world. Different families and genera have been analyzed in this work as regards their components. In 36% of the genera, the major phytochemical compounds found were alkaloids, steroids, flavonoids, saponins, tannins, and terpenes.

5. Bioactive Phytocompounds with Wound-Healing Properties

In the relevant literature, a variety of studies have addressed different plants with wound-healing properties. These studies have described the pharmacological activities of plants employed in wound healing and their molecular mechanisms to validate their traditional use and development into safe and effective herbal treatments for wounds. Due to the plants’ metabolism, secondary metabolites can be considered as bioactive molecules with therapeutic potential of great value in the pharmaceutical, cosmetic, and food industries, as concerns the design and formulation of medicines for different illnesses with less severe side effects [148,149,150,151,152]. The bioactive phytochemical compounds found include secondary metabolites such as alkaloids, essential oils, flavonoids, tannins, terpenoids, saponins, and phenolic compounds [153,154,155,156] (Figure 2).
The allocation of these active compounds into different plant parts, as has been widely described, involves the use of different selective solvents to derive complex mixtures of groups of metabolites (Figure 3).
Phenolic acids are the bioactive compounds most widely found in legumes, cereals, vegetables, and fruits. They are also responsible for certain characteristics of foods, such as aroma and astringency, as well as color and flavor [153,155,156,157] (Figure 4).
These compounds also play a role in plants’ ability to protect themselves against different insults, such as ultraviolet radiation and pathogens [153,156,157]. Importantly, the amounts of phenolic compounds produced by plants can vary according to environmental conditions, genetic factors, and degree of maturation [158]. In the literature, it has been described how phenolic compounds act as anti-inflammatory and antiproliferative agents, antioxidants, transduction modulators, stimulants of collagen production, and antimicrobials, in addition to carrying out other functions [154,156,157]. These compounds can be categorized into hydroxybenzoic acids, such as gallic and vanillic acids, as well as hydroxycinnamic acids such as ferulic and caffeic acids. They have also been shown to have immunomodulatory, antioxidant, hepatoprotective, and anti-inflammatory actions [159] (Figure 5).
Potent antioxidant agents, such as flavonoids, act as reducing agents and protect against radiation [160,161]. These protective effects mean that they can modulate pro-inflammatory molecules, such as those involved in the healing process [160,162]. The effects of flavonoids in the inflammatory process extend to the treatment of diseases linked to inflammation and processes of which inflammation is a part, such as the healing process and the inhibition of invasion, angiogenesis, and metastasis mechanisms [160].
Tannins are the most complex of the phenolic compounds, categorizable as condensates or hydrolysable. Their role is to protect plants from pathogens through protein complexation, and via their antimutagenic activity, they promote healing through the modulation of different cellular mechanisms and growth factors [153,156,157] (Figure 6).

6. Activity of Bioactive Phytochemicals in Wound Healing

Impaired vascular function, ischemia, superficial debris, and necrosis are the main factors that cause poor immune responses and, consequently, contribute to the development of continued chronic wounds. Excessive bacterial growth and the formation of a biofilm lead to a chronic and self-perpetuating inflammatory state via the modification of aspects of the wound microenvironment, such as its humidity, pH, metalloproteinases, and reactive oxygen species. As many of these microenvironment-related factors as possible must be taken into account to develop beneficial therapeutic strategies [163]. Nature, as described in the literature, is a rich source of therapeutic possibilities. Secondary metabolites can promote the wound-healing process through their pharmacological effects on the body. These compounds include phenolics, alkaloids, and fatty acids, as well as glycosylates and polysaccharides. Such compounds have also been confirmed to have beneficial effects related to their anti-inflammatory, antioxidant, and antibacterial properties, and they promote collagen synthesis and facilitate protective cell regeneration [164,165,166]. In addition, these active compounds present low toxicity and good absorption by the skin barrier [164]. The improved efficiency of treatments using natural extracts is related to the establishment of synergy, which enhances the effects of products of natural origin as well as current therapeutic approaches. Various studies have demonstrated that such synergistic interaction is a result of these substances’ antibacterial, antioxidant, and anti-inflammatory properties [167]. Active research in this area is currently focused on developing wound treatments able to prevent microorganisms from entering wounds with a bactericidal effect. Recent studies have shown that the use of vegetal extracts and their secondary metabolites has been integrated into diverse treatment modalities, and this has been proven to be effective against both Gram-positive and Gram-negative bacteria [168]. Some have already been selected for use in clinical trials or incorporated into nanoparticles [169]. Studies have shown that natural metabolites can represent beneficial candidates for use in wound healing. One obstacle in developing their clinical use is their poor oral or topical bioavailability.

6.1. Essential Oils

Research has shown that volatile essential oils present a variety of beneficial properties, such as antioxidant, antiviral, anticancer, insecticidal, anti-inflammatory, antiallergic, and antimicrobial effects [168]. These mixtures of lipophilic components are considered safe and biocompatible, although due to their low water solubility, bioavailability, and stability, their therapeutic uses can be limited [169].

6.2. Polyphenols

Polyphenols are considered multifaceted agents due to their beneficial activities, such as antibacterial, anticancer, anti-inflammatory, and antioxidant effects, in addition to their complex wound-healing properties [170]. However, the main problems include their hydrophobicity and poor water solubility, permeability, and bioavailability.

6.3. Flavonoids

As an exemplary flavonoid, quercetin has been harnessed for its antibacterial, anti-inflammatory, and antioxidant activities. When converted into quercetin nanofibers, it provides a large porous surface area that can carry many active compounds that facilitate penetration into the skin. Trials conducted with quercetin patches have shown them to have antibacterial activity that combats acne [171]. In other trials, film structures of N-carboxybutyl chitosan (CBC) and agarose were analyzed for their potential utilization in topical membranous wound treatment. Other research has demonstrated the use of polymeric biomaterials loaded with quercetin and thymol. These have been utilized both individually and in the form of mixtures of these two substances, which have anti-inflammatory and anesthetic properties. The incorporation of quercetin into semisolid bases such as creams and acid carbomer gels has been proposed to investigate the effects of additives such as propylene glycol and polyethylene glycol on its release and skin retention. With respect to quercetin and chrysin, or quercetin within chitosan nanoparticles, propylene glycol is an absorption accelerator that can also prolong the antioxidant activity [172,173]. Another study has demonstrated that polymeric nanoparticles can enhance antiradical activity, along with chelating quercetin and catechin [174]. Other studies have demonstrated the additional benefits offered by apigenin to the skin via the stimulation of epidermal differentiation, the synthesis and secretion of lipids, and cutaneous antimicrobial production. In vitro studies have demonstrated that hesperidin and naringin obtained from citrus fruits can be used to synthesize stabilized nanoparticles in a green manner [174].
Phytochemicals have been described to enhance the effects of antibiotics due to their low toxicity and anti-infective, anti-inflammatory, and antioxidant properties [175]. They can act as efflux pump inhibitors, preventing biofilm formation or targeting specific bacterial virulence factors [175]. Research confirms that plants from different families can facilitate the healing process and attenuate inflammation [176,177,178]. Certain compounds, including the flavonoid baicalein and the monoterpene phenol thymol, have an inhibitory effect on inflammation that has been demonstrated in mixtures of ethanol and can act synergistically, suggesting their use as an alternative treatment to antibiotics [176,177,178].

7. Mechanisms of Effects of Phytochemicals on Wound-Healing Agents

7.1. Antioxidant Activities of Wound-Healing Agents

Large amounts of energy must be produced for normal cellular activities, which is achieved through mechanisms such as oxidation, resulting in the generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) [156]. These reactive species possess unpaired electrons in their valence shell and are unstable [154]. Radical species include hydroxyl radicals (OH), nitric oxide radicals (NO), singlet oxygen (1O2), and superoxide radicals (O2). These are produced naturally in the body, but adverse factors such as stress and pollution can increase their levels, causing them to damage molecules such as proteins and DNA, leading to the disintegration of cell membranes. Oxidative stress is strongly linked to the development of chronic diseases and aging [154,157,160].
The use of antioxidants to control the levels of reactive species in the body is recommended. Antioxidants are defined as substances with the capacity to control the oxidation of biomolecules and act in the sequestration of reactive species such as ROS or RNS; some can also chelate metal ions and modulate enzymes related to oxidative stress [154,156]. Such enzymes include catalase (CAT), which catalyzes the degradation of hydrogen peroxide (H2O2), and glutathione peroxidase (GPx), which removes hydroperoxides [154].
Non-enzymatic processes involve transferrin, reduced glutathione (GSH), ubiquinol, and melatonin [154,156]. The antioxidant effects of these compounds are related to the presence of phenolic compounds, amino acids, sterols, ascorbic acid, peptides, and phospholipids in their composition [179]. Several studies have shown that antioxidants have anti-inflammatory, vasodilatory, antitumor, antiallergic, antiviral, and cardioprotective activities, among other properties [156,157,179].
During the healing process, excess free radicals are produced at the site of injury. This can be limited by the presence of antioxidants, which prevent some of the damage caused to cells [180]. The antioxidant action of medicinal plants is strongly related to the quantities of bioactive compounds they contain, such as flavonoids, which act as antioxidants and also directly participate in the inflammatory phase, limiting cellular damage due to their effects on prostaglandins and macrophages [181]. Flavonoids are also capable of increasing the resistance of collagen fibers, thus facilitating the process of the contraction and re-epithelization of wounds [180,182].
The healing process restores tissue integrity when an injury occurs [183]. It can be impeded by factors such as diabetes, which causes it to be slower and less efficient, thus potentially causing chronicity [184,185]. Poor healing can lead to the loss of tissue function, the chronification of injuries, and amputation, and it can also produce physical, psychological, social, and economic damage [184]. The treatments used to promote healing include the use of natural products and their derivatives. Some of the medications currently available are not completely effective in treating chronic wounds. For this reason, it is essential to continue research into new substances with more effective healing properties. Flavonoids and tannins have shown antiproliferative properties and are capable of regulating the production of free radicals; they are also involved in limiting inflammatory mechanisms [180]. Further investigations in the pharmaceutical, food, and cosmetic sectors will be essential in addressing the sources of antioxidants and substances that can be used to treat certain conditions, such as chronic wounds and cancer. Following the formation of ROS, the wound-healing process is significantly delayed; however, their formation is limited by the presence of flavonoids, which are responsible for increasing the levels of common antioxidant enzymes. The use of flavonoids in the clinical setting is very limited due to their low bioavailability. An important property of flavonoids obtained from plants is their lipophilicity against Gram-positive bacteria, which is a product of their involvement in the damage done to the respiratory chain and other aspects [186].

7.2. Anti-Inflammatory Properties of Wound-Healing Agents

Flavonoids have also been suggested as a candidate for use in the treatment of a variety of skin lesions, with minimal side effects when administered by topical application due to their lipophilic nature [187]. The many properties exhibited by flavonoids, such as their anti-inflammatory, antimicrobial, and antifibrotic effects, can be understood as a result of their polyhydroxy structure. Among all the structurally different flavonoids, twenty-four have demonstrated the ability to accelerate the healing process, and the most studied are quercetin, epigallocatechin gallate, and naringenin [55] (Figure 7).
Numerous studies have also shown that flavonoids are capable of decreasing the levels of inflammatory mediators, such as prostaglandins and leukotriene, and pro-inflammatory cytokines, such as IL-1β, TNF-α, IL-6, and IFN-γ. They can also increase the production of anti-inflammatory mediators, such as interleukin 10 (IL-10), negatively regulate the expression of nuclear factor kappa B (NF-κB), and block cyclooxygenase activity.
Prenylated flavonoids are found in plants’ roots, bark, seeds, and buds. These are part of a subclass of modified flavonoids with at least one lipophilic side chain of variable length, and they possess favorable biological activities, such as antimicrobial, antifungal, larvicidal, estrogenic, osteogenic, immunosuppressive, anticancer, anti-inflammatory, antioxidant, antiallergic and cytotoxic effects [188]. The group of prenylated flavonoids includes C-prenylated chalcones/dihydrochalcones, flavanones, flavones, flavonols, isoflavones, and, less frequently, O-prenylated forms (Figure 8).
These structures can be replaced, following oxidation, reduction, dehydration, and/or cycling, with 3,3-dimethylallyl, 1,1-dimethylallyl, geranyl, lavandulyl, and farnesyl side chains [188]. Studies have shown the advantages offered by prenyl compared to flavonoids. Prenylated flavonoids have a greater affinity with the cellular membrane and P-glycoprotein inhibitors [189] and show antibacterial and inhibitory or enzyme-enhancing actions, while prenylation causes an increase in lipophilicity and the affinity for biological membranes [188,189].
Diplacone, with its 6-geranyl-30,40,5,7-tetrahydroxyflavanone structure, has shown anti-inflammatory properties both in vitro and in vivo, with different mechanisms of action. It can cause reductions in TNF-α and MCP-1 expression and regulates the expression of zinc-finger protein 36, which increases cytokine degradation [190]. Another compound, isobavachalcone, suppresses the production of nitric oxide and negatively regulates inflammation-related enzymes such as iNOS and 15-LOX [191,192]. Licochalcone A is a 5-(2-methylbut-3-en-2-yl) chalcone obtained from licorice roots, and it has been traditionally used to treat inflammatory diseases. It inhibits the activation of transcription factors such as NF-κB and AP-1; it also suppresses pro-inflammatory cytokines and NO and PGE2 production [193,194]. The main function of Sophoraflavanone G is the inhibition of eicosanoid-forming enzymes [195]. It can also disrupt NF-κB and MAPK signaling pathways [195,196]. Another prenylated chalcone is xanthohumol, which is found in Humulus lupulus L. hops and has antistaphylococcal activity [197,198]. Its anti-inflammatory effect is enacted through the inhibition of NO levels due to the suppression of inducible NO synthase, and it inhibits both the activation of NF-κB [199,200] and the production of the cytokines MCP-1, TNF-α, and IL-12, as well as oxidative stress [201,202,203].

7.3. Antimicrobial Effects of Wound-Healing Agents

In the previous sections, we mention that flavonoids are widely used as effective therapeutic agents and that numerous in vitro and in vivo studies have confirmed them to have important functions, mainly defensive and regulatory [203]. Regarding their functions as protective agents against microorganisms, flavonoids act directly on bacterial cells, as well as suppressing virulence and the formation of biofilms. They can also act synergistically with antibiotics [204]. These properties have enabled the production and use of semisynthetic or synthetic flavonoids to combat microorganisms [205].
The antibacterial activities of flavonoids and prenylated flavonoids are due to the structure of 2-phenyl-1,4-benzopyrone, which has been suggested to be capable of influencing different cellular processes [204]. Apigenin and quercetin present the ability to inhibit bacterial cell walls by inhibiting D-alanine–D-alanine (D-Ala–D-Ala) ligase, which is crucial to the completion of peptidoglycan precursors [206]. Researchers have indicated that several flavonoids can modify membrane permeability and damage membrane functions. On the other hand, flavanols, flavolans, and green tea catechins have been shown to disturb bacterial cytoplasmic membranes through hydrogen peroxide [207,208]. Another flavone, Artocarpin, obtained from the Moraceae family, with prenyl in position 3 and a (1E)-3-methylbut-1-enyl moiety in position 6, presented remarkable antibacterial activity [197,209,210,211].

8. Conclusions

Plants are excellent wound healers, and when used in the context of different wound models, they can be employed as part of proper measures to treat wounds and control the healing process. Thus, herbal medicines have gained popularity in several countries. The factors that must be considered in the healing of a lesion are the wound closure rate, epithelialization, tensile strength, histopathology, and granuloma weight. This study discusses how traditional medicines could play important roles in wound healing. Modern knowledge of these bioactive principles can provide alternatives to improve or accelerate wound healing with minimal toxicity. The preliminary evidence and results in the current literature suggest that this is an active area of study. In future studies, factors such as the potential toxicity to human cells, kinetics and speed of healing, wound types, chronicity, timing of application, and dose of therapeutic agent must be considered. The preparation of formulations that include medicinal plants as part of their release and distribution systems for their anti-inflammatory, antioxidant, and wound-healing properties requires further investigation. These proposed studies on natural or synthetic formulations can be achieved by acquiring certain quantities of pure compounds and their extracts for standardization.

Author Contributions

Conceptualization, V.L.P. and L.R.M.-C.; methodology, M.C.-C.; software (GraphPadPrism 8.0.1), S.E.-P.; formal analysis, V.L.P. and M.C.-C.; investigation, J.A.M.L. and I.L.B.L.; resources, M.C.-C.; writing—original draft preparation, V.L.P. and S.E.-P.; writing—review and editing, V.L.P.; visualization, M.C.-C. and S.E.-P.; supervision, V.L.P.; project administration, V.L.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Secretaria de Educación Pública (México) (SEP-PROMEP:UAEMMOR-CA-07) and Consejo Nacional de Ciencia y Tecnología (CONACyT-México ON.551-6/18-7513).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing is not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Strodtbeck, F. Physiology of wound healing. Newborn Infant. Nurs. Rev. 2001, 1, 43–52. [Google Scholar] [CrossRef]
  2. Young, A.; McNaught, C.E. The physiology of wound healing. Surgery 2011, 29, 475–479. [Google Scholar] [CrossRef]
  3. Singer, A.J.; Clarck, R.A.F. Cutaneous wound healing. N. Engl. J. Med. 1999, 341, 738–746. [Google Scholar] [CrossRef] [PubMed]
  4. Broughton, G.; Janis, J.E.; Attinger, C.E. The basic science of wound healing. Plast. Reconstr. Surg. 2006, 117 (Suppl. 7), S12–S34. [Google Scholar] [CrossRef] [PubMed]
  5. Andrade, M.G.L. Alterações do processo de cicatrização de queimaduras em indivíduos diabéticos. Rev. Bras. Queimaduras 2013, 12, 42–48. [Google Scholar]
  6. Brem, H.; Tomic-Canic, M. Cellular and molecular basis of wound healing in diabetes. J. Clin. Investig. 2007, 117, 1219–1222. [Google Scholar] [CrossRef] [PubMed]
  7. Isaac, C. Processo de cura das feridas: Cicatrização fisiológica. Rev. Med. 2010, 89, 125–131. [Google Scholar] [CrossRef]
  8. Mandelbaum, S.H.; Di Santis, E.P.; Mandelbaum, M.H.S.A. Cicatrization: Current concepts and auxiliary resources—Parte I. An. Bras. Dermatol. 2003, 72, 393–410. [Google Scholar] [CrossRef]
  9. Mallefet, P.; Dweck, A.C. Mechanism of Wound Healing Examined. Personal. Care 2008, 9, 75–83. [Google Scholar]
  10. Pober, J.S.; Sessa, W.C. Evolving functions of endothelial cells in inflammation. Nat. Rev. Immunol. 2007, 7, 803–815. [Google Scholar] [CrossRef]
  11. Kumar, V.; Abbas, A.K.; Aster, J.C. Robbins Patologia Básica, 9th ed.; Elsevier: Rio de Janeiro, Brazil, 2013; p. 928. [Google Scholar]
  12. Lamano, T.L.C. Patologia Geral. Inflamação; Universidade de São Paulo: Ribeirão Preto, Brazil, 2008. [Google Scholar]
  13. Rodero, M.P.; Khosrotehrani, K. Skin wound healing modulation by macrophages. Int. J. Clin. Exp. Pathol. 2010, 3, 643–653. [Google Scholar]
  14. Ross, R.; Odland, G. Human wound repair. II. Inflammatory cells, epithelial-mesenchymal interrelations, and fibrogenesis. J. Cell Biol. 1968, 39, 152–168. [Google Scholar] [CrossRef] [PubMed]
  15. Kim, M.H.; Liu, W.; Borjesson, D.L.; Curry, F.-R.E.; Miller, L.S.; Cheung, A.L.; Liu, F.-T.; Isseroff, R.R.; Simon, S.I. Dynamics of neutrophil infiltration during cutaneous wound healing and infection using fluorescence imaging. J. Investig. Dermatol. 2008, 128, 1812–1820. [Google Scholar] [CrossRef] [PubMed]
  16. Deppermann, C.; Cherpokova, D.; Nurden, P.; Schulz, J.-N.; Thielmann, I.; Kraft, P.; Vögtle, T.; Kleinschnitz, C.; Dütting, S.; Krohne, G.; et al. Gray platelet syndrome and defective thrombo-inflammation in Nbeal2-deficient mice. J. Clin. Investig. 2013, 123, 3331–3342. [Google Scholar] [CrossRef] [PubMed]
  17. Mendonça, R.J.; Coutinho-Neto, J. Aspectos celulares da cicatrização. An. Bras. Dermatol. 2009, 84, 257–262. [Google Scholar] [CrossRef]
  18. Waldrop, J.; Doughty, D. Wound-healing Physiology. In Acute and Chronic Wounds: Nursing Management; Mosby Inc.: London, UK, 2000; pp. 17–39. [Google Scholar]
  19. Enoch, S.; Leaper, D. Basic science of wound healing. Surgery 2007, 26, 31–37. [Google Scholar]
  20. Eckes, B.; Nischt, R.; Krieg, T. Cell-matrix interactions in dermal repair and scarring. Fibrogenesis Tissue Repair. 2010, 3, 4. [Google Scholar] [CrossRef]
  21. Reinke, J.M.; Sorg, H. Wound Repair and Regeneration. Eur. Surg. Res. 2012, 49, 35–43. [Google Scholar] [CrossRef]
  22. Waldrop, M.A.; Kolb, S.J. Current treatment options in neurology-SMA therapeutics. Curr. Treat Options Neurol. 2019, 21, 25. [Google Scholar] [CrossRef]
  23. Eming, S.A.; Brachvogel, B.; Odorisio, B.; Koch, M. Regulation of angiogenesis: Wound healing as a model. Prog. Histochem. Cytochem. 2007, 42, 115–170. [Google Scholar] [CrossRef]
  24. Beldon, P. Basic science of wound healing. Surgery 2010, 28, 409–412. [Google Scholar] [CrossRef]
  25. Delavary, B.M.; van der Veer, W.M.; van Egmond, M.; Niessen, F.B.; Beelen, R.H. Macrophages in skin injury and repair. Immunobiology 2011, 216, 753–762. [Google Scholar] [CrossRef] [PubMed]
  26. Galiano, R.D.; Michaelis, J.; Dobryansky, M.; Levine, J.P.; Gurtner, G.C. Quantitative and reproducible murine model of excisional wound healing. Wound Rep. Regen. 2004, 12, 485–492. [Google Scholar] [CrossRef]
  27. Galkowska, H.; Wojewodzka, U.; Olszewski, W.L. Chemokines, cytokines, and growth factors in keratinocytes and dermal endothelial cells in the margin of chronic diabetic foot ulcers. Wound Rep. Regen. 2006, 14, 558–565. [Google Scholar] [CrossRef]
  28. Eo, H.; Lee, H.J.; Lim, Y. Ameliorative Effect of Dietary Genistein on Diabetes Induced Hyper-Inflammation and Oxidative Stress During Early Stage of Wound Healing in Alloxan Induced Diabetic Mice. Biochem. Biophys. Rev. Commun. 2016, 478, 1021–1027. [Google Scholar] [CrossRef] [PubMed]
  29. Park, J.H.; Kim, S.; Hong, H.S.; Son, Y. Substance P promotes diabetic wound healing by modulating inflammation and restoring cellular activity of mesenchymal stem cells. Wound Rep. Reg. 2016, 24, 337–348. [Google Scholar] [CrossRef] [PubMed]
  30. Romana-Souza, A.P.; Nascimento, A.P.; Monte-Alto-Costa, A. Propranolol improves cutaneous wound healing in streptozotocin-induced diabetic rats. Eur. J. Pharmacol. 2009, 611, 77–84. [Google Scholar] [CrossRef]
  31. Kant, V.; Gopal, A.; Pathak, N.N.; Kumar, P.; Tandan, S.K.; Kumar, D. Antioxidant and anti-inflammatory potential of curcumin accelerated the cutaneous wound healing in streptozotocin-induced diabetic rats. Int. Immunopharmacol. 2014, 20, 322–330. [Google Scholar] [CrossRef] [PubMed]
  32. Kant, V.; Gopal, A.; Kumar, D.; Pathak, N.N.; Ram, M.; Jangir, B.L.; Tandan, S.K.; Kumar, D. Curcumin-induced angiogenesis hastens wound healing in diabetic rats. J. Surg. Res. 2015, 193, 978–988. [Google Scholar] [CrossRef]
  33. WHO Implementation of WHO Traditional Medicine Strategy 2014–2023; Bolletin; WHO: Geneva, Switzerland, 2013; ISBN 9789241506090.
  34. Alvim, N.A.T.; Ferreira, M.D.A.; Cabral, I.E.; Almeida Filho, A.J.D. O uso de plantas medicinais como recurso terapêutico: Das influências da formação profissional às implicações éticas e legais de sua aplicabilidade como extensão da prática de cuidar realizada pela enfermeira. Rev. Lat.-Am. Enferm. 2006, 14, 316–323. [Google Scholar] [CrossRef] [PubMed]
  35. Carvalho, A.C.B.; Silveira, D. Drogas vegetais: Uma antiga nova forma de utilização de plantas medicinais. Brasília Méd. 2010, 48, 219–237. [Google Scholar]
  36. Silva, D.M.; Mocelin, K.R. O cuidado de enfermagem ao cliente portador de feridas sob a ótica do cuidado transcultural. Nursing 2007, 9, 8188. [Google Scholar]
  37. Aquino, D.; Silva, R.B.L.D.; Gomes, V.F.; Araújo, E.C.D. Nível de conhecimento sobre riscos e benefícios do uso de plantas medicinais e fitoterápicos de uma comunidade do Recife—PE. Rev. Enferm. UFPE Online 2007, 1, 107–110. [Google Scholar] [CrossRef]
  38. Sund, B. New Developments in Wound Care; Clínica Report; PJB Publications: New York, NY, USA, 2000; pp. 1–255. [Google Scholar]
  39. Wanda, A.; Dorsette, M. Rat models of skin wound healing: A review. Wound Rep. Regen. 2004, 12, 591–599. [Google Scholar]
  40. Jurjus, A.; Atiyeh, B.; Abdallah, I.; Jurjus, R.; Hayek, S.; Jaoude, M.; Gerges, A.; Tohme, R. Pharmacological modulation of wound healing in experimental burns. Burns 2007, 3, 892–907. [Google Scholar] [CrossRef]
  41. Agyare, C.; Bempah, S.B.; Boakye, Y.D.; Ayande, P.G.; Adarkwa-Yiadom, M.; Mensah, K.B. Evaluation of antimicrobial and wound healing potential of Justicia flava and Lannea welwitschii. Evid. Based Complement. Altern. Med. 2013, 2013, 632927. [Google Scholar] [CrossRef]
  42. Al-Bayaty, F.H.; Abdulla, M.A.; Hassan, M.I.A.; Ali, H.M. Effect of Andrographis paniculata leaf extract on wound healing in rats. Nat. Prod. Res. 2012, 26, 423–429. [Google Scholar] [CrossRef]
  43. Mengie, T.; Mequanente, S.; Nigussie, D.; Legesse, B.; Makonnen, E. Investigation of wound healing and anti-inflammatory activities of 80% methanol leaf extract of Achyranthes aspera L. (Amaranthaceae) in rats. J. Inflamm. Res. 2021, 14, 1775–1787. [Google Scholar] [CrossRef]
  44. Muniandy, K.; Gothai, S.; Sean Tan, W.; Suresh Kumar, S.; Mohd Esa, N.; Chandramohan, G.; Al-Numair, K.; Arulselvan, P. In Vitro Wound Healing Potential of Stem Extract of Alternanthera sessilis. Evid. Based Complement. Altern. Med. 2018, 2018, 3142073. [Google Scholar] [CrossRef]
  45. Kallappa, M.H.; Ganjihal, S.S.; Chavadi, D.V. Alternanthera triandra seed oil: A moderate source of ricinoleic acid and its possible industrial utilization. Ind. Crops Prod. 2004, 2, 133–136. [Google Scholar]
  46. Ofusori, A.E.; Raharjo, Y.; Ofusori, D.A.; Adekunle, V.O. A comparative study of dichloromethane and ethyl acetate root extracts of Celoisa trigyna: Phytochemical and wound healing effects analyses. J. Wound Manag. Res. 2023, 19, 87–95. [Google Scholar] [CrossRef]
  47. Pattnaik, A.; Sarkar, R.; Sharma, A.; Yadav, K.K.; Kumar, A.; Roy, P.; Sen, T. Pharmacological studies on Buchanania lanzan Spreng.-A focus on wound healing with particular reference to anti-biofilm properties. Asian Pac. J. Trop. Biomed. 2013, 3, 967–974. [Google Scholar] [CrossRef] [PubMed]
  48. Hsiao, C.Y.; Hung, C.-Y.; Tsai, T.-H.; Chak, K.-F. A Study of the wound healing mechanism of a traditional Chinese medicine, Angelica sinensis, using a proteomic approach. Evid.-Based Complement. Altern. Med. 2012, 2012, 467531. [Google Scholar] [CrossRef] [PubMed]
  49. Tanga, B.M.; Bang, S.; Fang, X.; Seo, C.; de Zoysa, M.; Saadeldin, M.I.M.; Lee, S.; Park, S.; Chung, S.-O.; Lee, G.-J.; et al. Centella asiatica extract in carboxymethyl cellulose at its optimal concentration improved wound healing in mice model. Heliyon 2022, 8, e12031. [Google Scholar] [CrossRef] [PubMed]
  50. Gohari, A.R.; Saeidnia, S.A. Review on Phytochemistry of Cuminum cyminum seeds and its standards from field to market. Pharmacogn. J. 2011, 3, 1–5. [Google Scholar] [CrossRef]
  51. Wu, J.-G.; Wei, Y.-J.; Ran, X.; Zhang, H.; Nian, H.; Qin, L.-P. Inhibitory effects of essential oil from rhizomes of Ligusticum chuanxiong on hypertrophic scarring in the rabbit ear model. Pharm. Biol. 2011, 49, 764–769. [Google Scholar] [CrossRef] [PubMed]
  52. Kabesh, K.; Senthulkumar, P.; Ragunathan, R.; Kumar, R. Phytochemical análisis of Catharantus roseus plan extract and its anti-microbial acitivity. Int. J. Pure App. Biosci. 2015, 3, 162–172. [Google Scholar]
  53. Agyare, C.; Dwobeng, A.S.; Agyepong, N.; Boakye, Y.D.; Mensah, K.B.; Ayande, P.G.; Adarkwa-Yiradon, M. Antimicrobial, antioxidant, and wound healing properties of Kigelia africana (Lam.) Beneth. and Strophanthus hispidus DC. Adv. Pharmacol. Sci. 2013, 2013, 692613. [Google Scholar] [CrossRef] [PubMed]
  54. Meenu, N.C.; Manokari, S.L.; Yogeswari, G.; Duraisami, R. Analysis of phytochemical constituents and antibacterial activity of Wrightia tinctoria: Traditional medicinal plant of India for application on wound dressing materials. Indian J. Trad. Knowl. 2022, 21, 48–54. [Google Scholar]
  55. Omale, J.; Victoria, I.A. Excision and incision wound healing potential of Saba florida (Benth) leaf extract in Rattus novergicus. Int. J. Pharm. Biomed. Res. 2010, 1, 101–107. [Google Scholar]
  56. Namgoong, S.; Lee, H.; Han, S.-K.; Lee, H.-W.; Jeong, S.-H.; Dhong, E.-S. Effect of Panax ginseng extract on the activity of diabetic fibroblasts in vitro. Int. Wound J. 2019, 16, 737–745. [Google Scholar] [CrossRef]
  57. Lu, T.; Gao, Y.; Duan, Y.; Cui, C.; Zhang, L.; Mingning, S. Panax notoginseng saponins improves healing of high glucose-induced wound through the GSK-3β/β-catenin pathway. Environ. Toxicol. 2022, 37, 1867–1877. [Google Scholar]
  58. Ali-Seyed, M.; Ayesha, S. Calotropis A multi-potential plant to human kind: Special focus on its wound healing efficacy. Biocatal. Agric. Biotechnol. 2020, 28, 101725. [Google Scholar] [CrossRef]
  59. Mali, R.P.; Raq, P.S.; Vikle, D.N. Wound healing activity of Calotropis procera root bark on diabetic rats. J. Drug Deliv. Ther. 2020, 10, 86–89. [Google Scholar] [CrossRef]
  60. Garcia-Orue, I.; Gainza, G.; Gutierrez, F.B.; Aguirre, J.J.; Evora, C.; Pedraz, J.L.; Hernandez, R.M.; Delgado, A.; Igartua, M. Novel nanofibrous dressings containing rhEGF and Aloe vera for wound healing applications. Int. J. Pharm. 2016, 523, 556–566. [Google Scholar] [CrossRef] [PubMed]
  61. Tadić, V.; Arsić, I.; Zvezdanović, J.; Zugić, A.; Cvetković, D.; Pavkov, S. The Estimation of the Traditionally Used Yarrow (Achillea millefolium L. Asteraceae) Oil Extracts with Anti-Inflamatory Potential in Topical Application. J. Ethnopharmacol. 2017, 199, 138–148. [Google Scholar] [CrossRef] [PubMed]
  62. Nascimento, B.A.C.; Gardinassi, L.G.; Silveira, I.M.G.; Galucci, M.G.; Tomre, M.A.; Oliveira, J.F.D.; Moreira, M.R.A.; Meirelles, A.F.G.; Faccioli, L.H.; Tefé-Silva, C.; et al. Arctium lappa extracts suppresses inflammation and inhibits melanoma progression. Medines 2019, 6, 81. [Google Scholar] [CrossRef]
  63. Pang, Y.; Wang, D.; Hu, X.; Wang, H.; Fu, W.; Fan, Z.; Chen, X.; Yu, F. Effect of volatile oil from Blumea balsamifera (L.) DC. leaves on wound healing in mice. J. Tradit. Chin. Med. 2014, 34, 716–724. [Google Scholar] [CrossRef] [PubMed]
  64. Dinda, M.; Mazumdar, S.; Das, S.; Ganguly, D.; Dasgupta, U.B.; Dutta, A.; Jana, K.; Karmakar, P. The water fraction of Calendula officinalis hydroethanol extract stimulates in vitro and in vivo proliferation of dermal fibroblasts in wound healing. Phytother. Res. 2016, 30, 1696–1707. [Google Scholar] [CrossRef] [PubMed]
  65. Parente, L.M.; Lino Júnior, R.d.S.; Faustino Tresvenzol, L.M.; Vinaud, M.C.; de Paula, J.R.; Paulo, N.M. Wound healing and anti-inflammatory effect in animal models of Calendula officinalis L. growing in Brazil. Evid.-Based Complement. Altern. Med. 2012, 2012, 375671. [Google Scholar] [CrossRef]
  66. Gao, S.-Q.; Chang, C.; Niu, X.-Q.; Li, L.-J.; Zhang, Y.; Gao, J.-Q. Topical application of Hydroxysafflor yellow A accelerates the wound healing in streptozotocin induced T1DM rats. Eur. J. Pharmacol. 2018, 823, 72–78. [Google Scholar] [CrossRef]
  67. Balekar, N.; Nakpheng, T.; Katkam, N.G.; Srichana, T. Wedelia trilobata L. Phytochemical and Pharmacological Reviews. Chiang Mai J. Sci. 2014, 41, 590–605. [Google Scholar]
  68. Agyare, G.; Koffuor, A.; Boamah, V.E.; Adu, F.; Mensah, K.B.; Adu-Amoah, L. Antimicrobial and anti-inflammatory activities of Pterygota macrocarpa and Cola gigantea (Sterculiaceae). Evid.-Based Complement. Altern. Med. 2012, 2012, 902394. [Google Scholar] [CrossRef]
  69. Kuklkarni, M.; Singhai, R.G.; Bhise, K.; Tambe, R. Phytochemical screening, HPTLC studies screening of antioxidants activity of extracts of leaves of Spanthodea campanulate. J. Pharmacogn. Phytochem. 2014, 3, 8–13. [Google Scholar]
  70. Elshamy, A.I.; Ammar, N.M.; Hassan, H.A.; El-Kashak, W.A.; Al-Rejaie, S.S.; Abd-ElGawad, A.M.; Farrag, A.-R.H. Topical Wound Healing Activity of Myricetin Isolated from Tecomaria capensis v. aurea. Molecules 2020, 25, 4870. [Google Scholar] [CrossRef]
  71. Nakuntwalai, W.; Thungmungmee, S.; Khobjai, W. Factor promoting wound healing: Radical scavenging and anti-inflammatory activity and growth factor promotion of Heliotropium indicum. Int. J. Appl. Pharm. 2019, 11, 44–48. [Google Scholar]
  72. Kojoma, M. Cultivation study of Lithospermum erythrorhizon to obtain “Shikon” as a purple dye and traditional medicine–root growth and shikonin derivatives content. In Proceedings of the ISHS Acta Horticulturae 1361: XXXI International Horticultural Congress (IHC2022): International Symposium on Natural Colorants from Plants, Angers, France, 14–20 August 2022; Räisänen, R., de la Sayette, A., Eds.; 2022. [Google Scholar]
  73. Kavousi, A.; Nikkhah, E.; Tayarani-Najaran, Z.; Javadi, B. Wound Healing Effects and Related Mechanims of Action of Methanol Extracts of Boswellia sacra and Commiphora myrrha Oleo-Gum Resins on Adult Human Dermal Fibroblasts (HDFa). Available online: https://ssrn.com/id3969804 (accessed on 9 January 2024).
  74. Yuan, X.; Han, L.; Fu, P.; Zeng, H.; Lv, C.; Chang, W.; Runyon, R.S.; Ishii, M.; Han, L.; Liu, K.; et al. Cinnamaldehyde accelerates wound healing by promoting angiogenesis via up-regulation of PI3K and MAPK signaling pathways. Lab. Investig. 2018, 98, 783–798. [Google Scholar] [CrossRef]
  75. Koshak, A.; Algandaby, M.N.; Mujallid, M.I.; Abdel-Naim, A.B.; Alhakamy, N.A.; Fahmy, U.A.; Alfarsi, A.; Badr-Eldin, S.M.; Neamatallah, T.; Nasrullah, M.Z.; et al. Wound healing activity of Opuntia ficus-indica fixed oil formulated in a self-nanoemulsifying formulation. Int. J. Nanomed. 2021, 16, 3889–3905. [Google Scholar] [CrossRef] [PubMed]
  76. Fanani Hakim, R.; Dinni, F. Effect of Carica papaya extract toward incised wound healing process in mice (Mus musculus) clinically and histologically. Evid.-Bases Complemment. Altern. Med. 2019, 2019, 8306519. [Google Scholar] [CrossRef]
  77. Quillay Davila, M.A.; Arana Arias, Y.A.; Jaramillo Jaramillo, C.G.; Buelle, S.C.; Rojas de Astudillo, L.L.; Jaramillo Alcívar, V. Contenido de saponinas y actividad cicatrizante de Cecropia peltata y Parthenium hysterophorus. Rev. Cuba. Farm. 2018, 51. Available online: http://www.revfarmacia.sld.cu/index.php/far/article/view/250/147 (accessed on 9 January 2024).
  78. Agyare, C.; Ansah, A.O.; Ossei, P.P.S.; Apenteng, J.A.; Boakye, Y.D. Wound healing and anti-infective properties of Myrianthus arboreus and Alchornea cordifolia. Med. Chem. 2014, 4, 533–539. [Google Scholar] [CrossRef]
  79. Chen, W.C.; Liou, S.-S.; Tzeng, T.-F.; Lee, S.-L.; Liu, I.-M. Wound repair and anti-inflammatory potential of Lonicera japonica in excision wound-induced rats. BMC Complement. Altern. Med. 2012, 12, 226. [Google Scholar] [CrossRef] [PubMed]
  80. Kisseih, E.; Lechtenber, M.; Petereit, F.; Sendker, J.; Brandt, S.; Agyare, C.; Hensel, A. Phytochemical characterization and in vitro wound healing activity of leaf extracts from Combretum mucronatum Schum. & Thonn.: Oligomeric procyanidins as strong inductors of cellular differentiation. J. Ethnopharmacol. 2015, 174, 628–636. [Google Scholar]
  81. Jokar, A.; Masoomi, F.; Sadeghpour, O.; Nassiri-Toosi, M.; Hamedi, S. Potential therapeutic applications for Terminalia chebula in Iranian traditional medicine. J. Trd. Chin. Med. 2016, 36, 250–254. [Google Scholar]
  82. Khan, M.U.; Khalilullah, H.; Akhtar, J.; Elhasan, G.O. Terminalia chebula: An ephemeral glance. Int. J. Pharm. Phram. 2015, 7, 40–43. [Google Scholar]
  83. Rodrigues Dantas Araujo, E.; Costa da Silva, V.; Figueiredo de Lima, J.B.; Schlamb, J.; Freitas Fernandes-Pedrosa, M.; Silva Junior, A.A.; Rathinasabapathy, T.; Moncada, M.; Esposito, D.; Coelho Bernardo Guerra, G.; et al. Gel formulated with Bryophyllum pinnatum leaf extract promotes skin wound healing in vivo by increasing VEGF expresión: A novel potential active ingredient for pharmaceuticals. Front. Pharmacol. 2023, 13, 1104705. [Google Scholar] [CrossRef] [PubMed]
  84. Ullah Arshad, M.; Hassan, A. Medical treatment of various diseases through Nagarmotha (Cyperus rotundus) plant. Eur. J. Biol. Med. Sci. Res. 2022, 10, 26–43. [Google Scholar] [CrossRef]
  85. Bigonya, P.; Agrawal, S.; Verma, N.K. Potential wound healing activity of Euphoria hirta Linn total flavonoids fractions. Int. J. Pharm. Sci. Rev. Res. 2013, 22, 149–156. [Google Scholar]
  86. Nwala, C.O.; Akaninwor, J.O.; Monanu, M.O. Phytochemical screening and wound healing activities of extracts of Jatropha curcas leaf formulated in a simple ointment base. Int. J. Eng. Sci. Invent. 2013, 2, 72–75. [Google Scholar]
  87. Kabran, F.A.; Okpekon, T.A.; Roblot, F.; Seon-Meniel, T.; Leblanc, K.; Bories, C.; Champy, P.; Yolou, S.F.; Loiseau, P.M.; Djakoure, L.A.; et al. Bioactive phloroglucinols from Mallotus oppositifolius. Fitoterapia 2015, 107, 100–104. [Google Scholar] [CrossRef]
  88. Ajirni, N.A.; Nazaruddin, A.; Sutriana, D.; Masyitha Isa, M. The effect of ethanol of Malacca leaves (Phyllanthus emblica) on the number of fibroblast cells in White rats (Tattus norvegicus) burns wound. Int. J. Trop. Vet. Biomed. Res. 2020, 5, 7–12. [Google Scholar] [CrossRef]
  89. Boakye, Y.D.; Agyare, C.; Ayande, P.G.; Asiamah, E.A.; Titloye, N.A. Wound healing activity of geraniin and aqueous leaf extract of Phyllanthus muellerianus (Kuntze) Excel (Euphorbiaceae). Planta Med. 2014, 80, WS20. [Google Scholar] [CrossRef]
  90. Jeong, S.Y.; Moon, M.Y.; Ryu, E.K.; Lee, J.S.; Chan, J. Identification of the phytochemical compounds and their type I procollagen induction in Astragalus membranaceus Sprouts grown under different light conditions. J. Appl. Pharmacetical. Sci. 2018, 8, 1–7. [Google Scholar]
  91. Tewtrakul, S.; Tungcharoen, P.; Sudsai, T.; Karalai, C.; Ponglimanont, C.; Yodsaoue, O. Antiinflammatory and wound healing effects of Caesalpinia sappan L. Phytother. Res. 2015, 29, 850–856. [Google Scholar] [CrossRef]
  92. Su, X.; Liu, X.; Wang, S.; Li, B.; Pan, T.; Liu, D.; Wang, F.; Diao, Y.; Li, K. Wound-healing promoting effect of total tannins from Entada phaseoloides (L.) Merr. In rats. Burns 2017, 43, 830–838. [Google Scholar] [CrossRef] [PubMed]
  93. Malekzadeh, M.; Sandoughdaran, S.; Shandiz, F.H.; Honary, S. The efficacy of Licorice root (Glycyrrhiza glabra) and yarrow (Achillea millefolium) in preventing radiation dermatitis in patients with breast cancer, a randomized, double-blinded, placebo-controlled clinical trial. Asian Pac. J. Cancer Care 2016, 1, 9. [Google Scholar] [CrossRef]
  94. Saini, S.; Dhiman, A.; Nanda, S. Traditional Indian Medicinal plants with potential wound healing activity: A review. Int. J. Pharm. Sci. Res. 2009, 7, 1809–1891. [Google Scholar]
  95. Kannan, S.; Vijay Jesuraj, S.A.; Jeeva Kumar, E.S.; Saminathan, K.; Suthakaran, R.; Saminathan, K.; Suthakaran, R.; Ravi Kumar, M.; Parimala Devi, B. Wound healing activity of Mimosa pudica Linn formulation. Int. J. PharmTech. Res. 2009, 1, 1554–1558. [Google Scholar]
  96. Saini, P.; Kumar Verma, P. Evaluation of the wound healing properties of Mimosa pudica Linn. In streptozocin-induced diabetes mellitus in rats. Int. J. Pharm. Sci. Res. 2019, 10, 661–665. [Google Scholar]
  97. Xu, X.; Li, X.; Zhang, L.; Liu, Z.; Pan, Y.; Chen, D.; Bin, D.; Deng, Q.; Sun, Y.; Hoffman, R.M.; et al. Enhancement of wound healing by the Chinese medicine herbal mixture Sophora flavescens in a rat model of perianal ulceration. Vivo 2017, 31, 543–549. [Google Scholar]
  98. Lodhi, S.; Jain, A.; Jain, A.P.; Pawar, R.S.; Singhai, A.K. Effects of flavonoids from Martynia annua and Tephora purpurea on cutaneous wound healing. Avicenna J. Phytomed. 2016, 6, 578–591. [Google Scholar]
  99. Chokpaisam, J.; Chusri, S.; Ammuaikit, T.; Udomuksorn, W.; Voravuthikunchai, S.P. Potential wound healing activity of Cuercus infectoria formulation in diabetic rats. PeerJ 2017, 5, e3608. [Google Scholar] [CrossRef]
  100. Cheng, P.G.; Phan, C.-W.; Sabaratnam, V.; Abdullah, N.; Abdulla, M.A.; Kuppusamy, U.R. Polysaccharides-rich extract of Ganoderma lucidum (M.A. Curtis:Fr.) P. Karst accelerates wound healing in streptozotocin-induced diabetic rats. Evid.-Based Complement. Altern. Med. 2013, 2013. [Google Scholar] [CrossRef]
  101. Ngwoke, K.G.; Akwaqbulam, A.G.; Erhirhie, E.O.; Ajaghaku, D.L.; Chiedu Okoye, F.B.; Okechukwu Esimone, C. Antioxidant, Anti-inflammatory, Analgesic Properties, and Phytochemical Characterization of Stem Bark Extract and Fractions of Anthocleista nobilis. Pharmacogn. Res. 2018, 10, 81–87. [Google Scholar] [CrossRef]
  102. Bardaa, S.; Makni, K.; Boudaouara, O.; Bardaa, T.; Ktari, N.; Hachira, S.; Salah, R.B.; Kallel, R.; Sahnound, Z.; Boufi, S. Development and evaluation of the wound healing effect of a novel topical cream formula based on Ginkgo biloba extract on wounds in diabetic rats. BioMed Res. Int. 2021, 2021, 6474706. [Google Scholar] [CrossRef] [PubMed]
  103. Hariharapura, R.; Srinivasan, R.; Ashok, G.; Dongra, S.H.; Jagani, H.V.; Vijayan, P. Investigation of the antioxidant and hepatoprotective potential Hypericum mysorense. Antioxidants 2014, 3, 526–543. [Google Scholar] [CrossRef] [PubMed]
  104. Gigliobianco, M.R.; Cortese, M.; Vargas Peregrina, D.V.; Villa, C.; Lupidi, G.; Pruccoli, L.; Angeloni, C.; Tarozzi, A. Development of new extracts of Crocus sativus L. By products from two different Italian regions as new potential active ingredient in cosmetic formulations. Cosmetics 2021, 8, 51. [Google Scholar] [CrossRef]
  105. Jayapal, V.; Subha, V.; Pradeep, J.; Janardan Salwe, K.; Manimekalai, R.; Tahinamala, R.; Kumar, B.; Rayvathy, B.; Ravi Kumar, S. Evaluation of wound healing potential of the essential oil of Ocimum sanctum L. (Thulasi/basil) containing ointment in female Wistar albino rats. J. Pharmacogn. Phytochem. 2023, 12, 189–193. [Google Scholar] [CrossRef]
  106. Rašković, A.; Milanović, I.; Pavlović, N.; Ćebović, T.; Vukmirović, S.; Mikov, M. Antioxidant Activity of Rosemary (Rosmarinus officinalis L.) Essential Oil and Its Hepatoprotective Potential. BMC Complement. Altern. Med. 2014, 14, 225. [Google Scholar] [CrossRef] [PubMed]
  107. Karimzadeh, S.; Farahpour, M.R. Topical application of Salvia officinalis hydroethanolic leaf extract improves wound healing process. Indian J. Exp. Biol. 2017, 55, 98–106. [Google Scholar]
  108. Guevara-Vazquez, A.M.; Marin-Tello, C.L. Wound healing activity of Allium cepa L. bulbs in a second-degree burn wound model in Holtzman rats. Vitae 2021, 28, 345737. [Google Scholar] [CrossRef]
  109. Park, J.-Y.; Hyck, K.; Lim, D.-W.; Kim, J.-E.; Park, W.-H.; Park, S.-D. Ethanol Extract of Lycopodium serratum Thunb. Attenuates Lipopolysaccharide-Induced C6 Glioma Cells Migration via Matrix Metalloproteinase-9 Expression. Chin. J. Integr. Med. 2018, 24, 860–866. [Google Scholar] [CrossRef]
  110. Dutta, S.; Pattnak, A.K.; Bersa, S.E. Wound healing potential of methanolic extract and its fraction of Lawsonia alba Lam leaves formulated a topical gel. World J. Pharm. Res. 2016, 5, 1091–1109. [Google Scholar]
  111. El Massoudi, S.; Zinedine, A.; Rocha, J.M.; Benidir, M.; Najjari, I.; El Ghadraoui, L.; Benjelloun, M.; Errachidi, F. Phenolic composition and wound healing potential assessment of Moroccan Henna (Lawsonia inermis) Aqueous extract. Cosmetics 2023, 10, 92. [Google Scholar] [CrossRef]
  112. Lukiswanto, B.S.; Miranti, A.; Sudjarwo, S.A.; Primarizky, H.; Yuniati, W.M. Evaluation of wound healing potential of pomegranate (Punica granatum) whole fruit extract on skin burn wound in rats (Rattus norvegicus). J. Adv. Vet. Anim. Res. 2019, 6, 202–207. [Google Scholar] [CrossRef] [PubMed]
  113. Balla, R.; Kaur, R.; Kaur, B.; Kaur, P. Hibiscus rosa sinensis Linn. A phytochemical and pharmacological review. Int. J. Health Sci. 2022, 6, 5165–5193. [Google Scholar]
  114. Almasian, A.; Najafi, F.; Eftekhari, M.; Shams Ardekani, M.R.; Sharifzadeh, M.; Khanavi, M. Polyurethane/carboxymethylcellulose nanofibers containing Malva sylvestris extract for healing diabetic wounds: Preparation, characterization, in vitro and in vivo studies. Mat. Sci. Eng. 2020, 114, 111039. [Google Scholar] [CrossRef] [PubMed]
  115. Asif, A.H.; Mulla, S.M.; Ashariff, A.; Sreeharsha, N.; Meravanige, G.; Shiroorkar, P.N.; Basheeruddin Asdaq, S.M.; Khalid Answer, M.D.; Roopashree, T.S.; Karnatti, R.K. Exploring the topical gel of Thespesia populnea leaf extract for in vivo wound healing efficacy. Pharmacogn. Mag. 2022, 18, 519–523. [Google Scholar]
  116. Maan, P.; Singh Yadav, K.; Yadav, N.P. Wound healing activity of Azadirachta indica A. Juss stem bark in mice. Pharmacogn. Mag. 2017, 13 (Suppl. S2), S316–S320. [Google Scholar]
  117. Silva, D.F.; Lima, K.T.; Bastos, G.T.N.; Oliveira, J.A.R.; do Nascimento, L.A.S.; Costa, C.E.F.; Filho, N.R.; Concha, V.O.; Passos, M.F. PCL/Andiroba oil (Carapa guianensis Aubl.) hybrid film for wound healing applications. Polymers 2021, 13, 1591. [Google Scholar] [CrossRef]
  118. Yadav, E.; Singh, D.; Yadav, P.; Vrma, A. Antioxidant and anti-inflammatory properties of Prosopis cineraria based phenolic rich ointment in wound healing. Biomed. Pharmacother. 2018, 108, 1572–1583. [Google Scholar] [CrossRef]
  119. Gupta, R.; Gupta, J. Ointment of methanolic extract Ficus religiosa: Traditional approach in wound healing in rats. Int. J. Pharm. Sci. Res. 2016, 7, 5006–5011. [Google Scholar]
  120. Susanto, A.; Muhaimina, R.K.; Amalliya, A.; Sutjiatmo, A.B. The effectiveness of ethanolic of Moringa oleífera Lam. Gel on the wound healing process of the rat’s palate. J. Int. Dent. Med. Res. 2019, 12, 504–509. [Google Scholar]
  121. Kumar, M.; Kumar Gautan, M.; Singh, A.; Kumar Goel, R. Healing effect of Musa sapientum var. Paradisiaca in diabetic rats with co-occurring gastric ulcer: Cytokines and growth factor by PCR amplification. BMC Complement. Altern. Med. 2013, 13, 305. [Google Scholar] [CrossRef]
  122. Shrimali, H.; Kumar Mandal, U.; Nivsarkar, M.; Shrivastawa, N. Fabrication and evaluation of a medicated hydrogel film with embelin from Embelia ribes for wound healing activity. Future J. Pharm. Sci. 2019, 5, 12. [Google Scholar] [CrossRef]
  123. Moreira, P.; Sousa, F.J.; Matos, P.; Sousa Brites, G.; Gonçalves, M.J.; Cavaleiro, C.; Figueirinha, A.; Salgueiro, L.; Batista, M.T.; Costa Branco, P.; et al. Chemical composition and effect against skin alterations of bioactive extracts obtained by the hydrodistillation of Eucalyptus globulus leaves. Pharmaceutics 2022, 14, 561. [Google Scholar] [CrossRef]
  124. Khuanekkaphan, M.; Noysang, C.; Khobjai, W. Anti-aging potential and phytochemicals of Centella asiática, nelumbo nucifera and Hibiscus sabdariffa extracts. J. Adv. Pharm. Technol. Res. 2020, 11, 174–178. [Google Scholar] [CrossRef] [PubMed]
  125. Mittal, A.; Satish, S.S.; Anima, P. Evaluation of wound healing, antioxidant and antimicrobial efficacy of Jasminum auriculatum Vahl. leaves. Avicenna J. Phytomed. 2015, 6, 295–304. [Google Scholar]
  126. Chaturvedi, A.P.; Kumar, M.; Tripathi, Y.B. Efficacy of Jasminum grandiflorum L. leaf extract on dermal wound healing in rats. Int. Wound J. 2013, 10, 675–682. [Google Scholar] [CrossRef] [PubMed]
  127. Zhang, C.; He, Y.; Chen, Z.; Shi, J.; Qu, Y.; Zhang, J. Effect of Polysaccharides from Bletilla striata on the Healing of Dermal Wounds in Mice. Evid.-Based Complement. Altern. Med. 2019, 2019, 9212314. [Google Scholar] [CrossRef]
  128. Ekiert, H.; Klimek-Szczykutowicz, M.; Szopa, A. Paeonia suffruticosa (Mountan Peony)—A review of the chemical composition, traditional and profesional use in medicine, position in cosmetics industries and biotechnological studies. Plants 2022, 11, 3379. [Google Scholar] [CrossRef]
  129. Ayele, T.M.; Chekol Abebe, E.; Tilahum Muche, Z.; Mekonnen Agidew, M.; Shumet Yimer, Y.; Testaw Addis, G.; Dagnaw Baye, N.; Bogale Kassie, A.; Adela Alemu, M.; Gobezie Yblet, T.; et al. Evaluation of in vivo wound-healing and anti-inflammatory activities of solvent fractions of fruits of Argemone mexicana L. (Papaveraceae). Evid.-Based Complement. Altern. Med. 2022, 2022, 6154560. [Google Scholar] [CrossRef]
  130. Heba, E.; El Aty, A.; Zaazaa, A.M.; Mohamed, S.H.; El Dayem, S.A.; Foda, F. Promising Therapeutic Efficacy of Trigonella-foenum graecum and Bone Marrow-Derived Mesenchymal Stem Cells on Skeletal Muscle Atrophy in Experimental Rat Model. Biointerrface Res. Appl. Chem. 2023, 13, 133. [Google Scholar] [CrossRef]
  131. Somwanshi Sachim, B.; Hiremath Shivanand, N. In vivo evaluation of the wound healing activity of the Sesamum indicum L. seed extract in novel ethosomal vesicular system. J. Drug Del. Therap. 2018, 8, 411–420. [Google Scholar] [CrossRef]
  132. Ghanadian, M.; Soltani, R.; Homayouni, A.; Khorvash, F.; Jouabadi, S.M.; Abdollahzadeh, M. The Effect of Plantago Major Hydroalcoholic Extract on the Healing of Diabetic Foot and Pressure Ulcers: A Randomized Open-Label Controlled Clinical Trial. Int. J. Low Extrem. Wounds 2022. [Google Scholar] [CrossRef]
  133. Yang, W.-T.; Ke, C.-Y.; Wu, W.-T.; Harn, H.-J.; Teng, Y.-H.; Lee, R.-P. Effects of Angelica dahurica and Rheum officinale extracts on excisional wound healing in rats. Evid.-Based Complement. Altern. Med. 2017, 2017, 1583031. [Google Scholar] [CrossRef] [PubMed]
  134. Budiawan, A.; Purwanto, A.; Puradewa, L.; Dwi Cahyani, E.; Endang Purwaningsh, C. Wound healing activity and flavonoid contentes of pursale (Portulaca grandiflora) of various varieties. RSC Adv. 2023, 13, 9871. [Google Scholar] [CrossRef]
  135. Mahomoodally, M.F.; Jugreet, S.; Sinan, K.I.; Zengin, G.; Gunes, A.K.; Ramazan, C.; Josef, J.; Zoltan, C.; Angelin, P.; Angeles Flores, G.; et al. Pharmacological potential and chemical characrization of Bridelia ferruginea Benth.—A native tropical African Medicinal Plants. Antibiotics 2021, 10, 223. [Google Scholar] [CrossRef]
  136. Zhang, H.; Chen, J.; Cen, Y. Burn wound healing potential of a polysaccharide from Sanguisorba officinalis L. in mice. Int. J. Biol. Macromol. 2018, 112, 862–867. [Google Scholar] [CrossRef] [PubMed]
  137. Ly, H.T.; Pham Nguyen, M.T.; Oanh Nguyen, T.K.; Quynh Bui, T.P.; Ke, X.; Le, V.M. Phytochemical analysis and wound healing activity of Noni (Morinda citrifolia) leaf extract. J. Herbs. Spices Med. Plants 2020, 26, 379–393. [Google Scholar] [CrossRef]
  138. Wen, M.; Chen, Q.; Chen, W.; Yang, J.; Zhou, X.; Zhang, C.; Wu, A.; Lai, J.; Chen, J.; Mei, Q.; et al. A comprehensive review of Rubia cordifolia L.: Traditional uses, phytochemistry, pharmacological activities, and clinical applications. Front. Pharmacol. 2022, 13, 965390. [Google Scholar] [CrossRef]
  139. Azmil, L.; Shukla, I.; Goutam, A.; Allauddin Rao, C.H.V.; Jawaid, T.; Kamal, M.; Awaad, A.S.; Alqasoumi, S.I. In vitro wound healing activity of 1-hydroxy-5,7-dimethoxy-2-naphthalene-carboxaldehyde (HDNC) and other isolates of Aegle marmelos L.: Enhances keratinocytes motility via Wnt/β-catenin and RAS-ERK pathways. Saudi Pharm. J. 2019, 27, 532–539. [Google Scholar] [CrossRef] [PubMed]
  140. Zhang, M.; Wang, J.; Zhu, L.; Li, T.; Jiang, W.; Peng, W.; Wu, C. Zanthoxylum bungeanum Maxim. (Rutacea): A systematic Review of traditional uses, botany, phytochemistry, pharmacology, pharmacokinetics, and toxicology. Int. J. Mol. Sci. 2017, 18, 2172. [Google Scholar] [CrossRef] [PubMed]
  141. Albano, M.N.; da Silveira, M.R.; Danielski, L.G.; Florentino, D.; Petronilho, F.; Piovezan, A.P. Anti-Inflammatory and Antioxidant Properties of Hydroalcoholic Crude Extract from Casearia sylvestris Sw. (Salicaceae). J. Ethnopharmacol. 2013, 147, 612–617. [Google Scholar] [CrossRef]
  142. Xu, L.; Zhang, W.; Zeng, L.; Jin, J.-O. Rehmannia glutinosa polysaccharide induced an anti-cancer effect by activating natural killer cells. Int. J. Biol. Macromol. 2017, 105, 680–685. [Google Scholar] [CrossRef]
  143. Kil, Y.-S.; Park, J.; Han, A.-R.; Woo, H.; Seo, E.-K. A new 9,10-dihydrophenanthrene and cell proliferative 3,4-δ-dehydrotocopherols from Stemona tuberosa. Molecules 2015, 20, 5965–5974. [Google Scholar] [CrossRef] [PubMed]
  144. Hajiaghaalipour, F.; Kanthimathi, M.S.; Abdulla, M.A.; Sanusi, J. The effect of Camellia sinensis on wound healing potential in an animal model. Evid.-Based Complement. Altern. Med. 2013, 2013. [Google Scholar] [CrossRef] [PubMed]
  145. Chen, Y.; Guo, J.; Tang, Y.; Wu, L.; Tao, W.; Qian, Y.; Duan, J.-A. Pharmacokinetic profile and metabolite identification of yuanhuapine, a bioactive component in Daphne genka by ultra-high performance liquid chromatography coupled with tandem mass spectrometry. J. Pharm. Biomed. Anal. 2015, 112, 60–69. [Google Scholar] [CrossRef] [PubMed]
  146. Liang, J.H.; Lin, H.R.; Yang, C.S.; Liaw, C.C.; Wang, I.C.; Chen, J.J. Bioactive components from Ampelopsis japonica with antioxidant, glucosidase, and antiacetylcholinesterase activities. Antioxidants 2022, 11, 1228. [Google Scholar] [CrossRef]
  147. Irham, W.H.; Hardiyanti, R. Wound healing bioactivity of Curcuma longa Linn. Rasayan J. Chem. 2021, 14, 2386–2391. [Google Scholar] [CrossRef]
  148. Singh, S.; Gupta, A.; Singh, B.B. Effect of foliage supplementation to Heteropogon contortus based diets on nutrients digestibility, gas and metabolites production in sheep and goat inoculums. Anim. Nutr. Feed Technol. 2016, 16, 439–450. [Google Scholar] [CrossRef]
  149. Chandika, P.; Ko, S.-C.; Jung, W.-K. Marine-derived biological macromolecule-based biomaterials for wound healing and skin tissue regeneration. Macromolecules 2015, 77, 24–25. [Google Scholar] [CrossRef] [PubMed]
  150. Ghitescu, R.E.; Volf, I.; Carausu, C.; Bühlmann, A.M.; Gilca, J.A.; Popa, V.I. Optimization of ultrasound-assisted extraction of polyphenols from spruce wood bark. Ultrason. Sonochem. 2015, 22, 535–541. [Google Scholar] [CrossRef] [PubMed]
  151. Arruda, H.S.; Pereira, G.A.; Pastore, G.M. Optimization of extraction parameters of total phenolics from Annona crassiflora Mart. (araticum) fruits using response surface methodology. Food Anal. Methods 2017, 10, 100–110. [Google Scholar] [CrossRef]
  152. George, E.; Kasipandi, M.; Vekataramana, M.; Kumar, K.N.; Allen, J.A.; Parimelazhagan, T.; Gopalan, N. In vitro anti-oxidant and cytotoxic analysis of Pogostemon mollis Benth. Bangladesh J. Pharmacol. 2016, 11, 148–158. [Google Scholar] [CrossRef]
  153. Velderrain-Rodríguez, G.; Palafox-Carlos, H.; Wall-Medrano, A.; Ayala-Zavala, J.F.; Chen, C.-Y.O.; Robles-Sánchez, M.; Astiazaran-García, H.; Alvarez-Parrilla, E.; González-Aguilar, G.A. Phenolic compounds: Their journey after intake. Food Funct. 2014, 5, 189–197. [Google Scholar] [CrossRef] [PubMed]
  154. Arruda, H.S.; Silva, E.K.; Pereira, E.K.; Angolini, C.F.F.; Eberlin, M.N.; Meireles, M.A.A.; Pastore, G.M. Effects of high-intensity ultrasound process parameters on the phenolic compound recovery from araticum peel. Ultrason. Sonochem. 2019, 50, 82–95. [Google Scholar] [CrossRef] [PubMed]
  155. Arruda, H.S.; Pereira, G.A.; Pastore, G.M. Brazilian Cerrado fruit araticum (Annona crassiflora Mart.) as a potential source of natural antioxidant compounds. Int. Food Res. J. 2018, 25, 2005–2012. Available online: http://www.ifrj.upm.edu.my/25(05)2018/(33).pdf (accessed on 9 January 2024).
  156. Arruda, H.S.; Fernandes, R.V.B.; Botrel, D.A.; Almeida, M.E.F. Frutos do Cerrado: Conhecimento e aceitação de Annona crassiflora Mart. (araticum) e Eugenia dysenterica Mart. (Caigata) por crianças utilizando o paladar e a visão. J. Health Biol. Sci. 2015, 3, 224–230. [Google Scholar] [CrossRef]
  157. Chen, M.-X.; Huo, J.-M.; Hu, J.; Xu, Z.-P.; Zhang, X. Amaryllidaceae alkaloids from Crinum latifolium with cytotoxic, antimicrobial, antioxidant, and anti-inflammatory activities. Fitoterapia 2018, 130, 48–53. [Google Scholar] [CrossRef]
  158. Formagio, A.S.N.; Vieira, M.C.; Volobuff, C.R.F.; Silva, M.S.; Matos, A.I.; Cardoso, C.A.L.; Carvalho, J.E. In vitro biological screening of the anticholinesterase and antiproliferative activities of medicinal plants belonging to Annonaceae. Braz. J. Med. Biol. Res. 2015, 48, 308–315. [Google Scholar] [CrossRef]
  159. Liu, Y.; Xiao-Hong, G.; Tang, J.; Liu, K. Antioxidant Activities of Hops (Humulus Lupulus) and Their Products. J. Am. Soc. Brew. Chem. 2018, 65, 116–121. [Google Scholar] [CrossRef]
  160. Zhang, I.; Ravipati, A.S.; Koyyalamudi, S.R.; Jeong, S.C.; Reddy, N.; Smith, P.T.; Bartlet, J.; Shanmugam, K.; Munch, G.; Wu, M.J. Antioxidant and anti-inflammatory activities of selected medicinal plants containing phenolic and flavonoid compounds. J Agric. Food Chem. 2011, 59, 12361–12367. [Google Scholar] [CrossRef]
  161. Fattahi, M.; Rahimi, R. Optimization of extraction parameters of phenolic antioxidants from leaves of Capparis spinosa using response surface methodology. Food Anal. Method 2016, 9, 2321–2334. [Google Scholar] [CrossRef]
  162. Formagio, A.S.N.; Masetto, T.E.; da Baldivia, D.S.; Vieira, M.C.; Zarate, N.A.H.; Pereira, Z.V. Potencial alelopático de cinco espécies da família Annonaceae. Braz. J. Bioscien. 2010, 8, 349–354. [Google Scholar]
  163. Scalise, A.; Bianchi, A.; Tartaglione, C.; Bolletta, E.; Pierangeli, M.; Torresetti, M.; Marazzi, M.; Di Benedetto, G. Microenvironment and Microbiology of Skin Wounds: The Role of Bacterial Biofilms and Related Factors. Semin. Vasc. Surg. 2015, 28, 151–159. [Google Scholar] [CrossRef] [PubMed]
  164. Ibrahim, N.; Wong, S.K.; Mohamed, I.N.; Mohamed, N.; Chin, K.Y.; Ima-Nirwana, S.; Shuid, A.N. Wound Healing Properties of Selected Natural Products. Int. J. Environ. Res. Public Health 2018, 15, 2360. [Google Scholar] [CrossRef] [PubMed]
  165. Tsala, D.E.; Amadou, D.; Habtemariam, S. Natural Wound Healing and Bioactive Natural Products. Phytopharmacology 2013, 4, 532–560. [Google Scholar]
  166. Bittner Fialová, S.; Rendeková, K.; Mucaji, P.; Nagy, M.; Slobodníková, L. Antibacterial Activity of Medicinal Plants and Their Constituents in the Context of Skin and Wound Infections, Considering European Legislation and Folk Medicine A Review. Int. J. Mol. Sci. 2021, 22, 10746. [Google Scholar] [CrossRef] [PubMed]
  167. Amparo, T.R.; Seibert, J.B.; de Abreu Vieira, P.M.; Teixeira, F.F.M.; dos Santos, O.D.F.; de Souza, G.H.B. Herbal Medicines to the Treatment of Skin and Soft Tissue Infections: Advantages of the Multi-Targets Action. Phyther. Res. 2020, 34, 94–103. [Google Scholar] [CrossRef]
  168. Simões, D.; Miguel, S.P.; Ribeiro, M.P.; Coutinho, P.; Mendonça, A.G.; Correia, J.J. Recent Advances on Antimicrobial Wound Dressing: A Review. Eur. J. Pharm. Biopharm. 2018, 127, 130–141. [Google Scholar] [CrossRef]
  169. Andreu, V.; Mendoza, G.; Arruebo, M.; Irusta, S. Smart Dressings Based on Nanostructured Fibers Containing Natural Origin Antimicrobial, Anti-Inflammatory, and Regenerative Compounds. Materials 2015, 8, 5154–5193. [Google Scholar] [CrossRef]
  170. Maheshwari, R.K.; Singh, A.K.; Gaddipati, J.; Srimal, R.C. Multiple Biological Activities of Curcumin: A Short Review. Life Sci. 2006, 78, 2081–2087. [Google Scholar] [CrossRef] [PubMed]
  171. Amer, S.S.; Mamdouh, W.; Nasr, M.; ElShaer, A.; Polycarpou, E.; Abdel-Aziz, R.T.A.; Sammour, O.A. Quercetin Loaded CosmNutraceutical Electrospun Composite Nanofibers for Acne Alleviation: Preparation, Characterization and Experimental Clinical Appraisal. Int. J. Pharm. 2022, 612, 121309. [Google Scholar] [CrossRef] [PubMed]
  172. Dyja, R.; Jankowski, A. The Effect of Additives on Release and in Vitro Skin Retention of Flavonoids from Emulsion and Gel Semisolid Formulations. Int. J. Cosmet. Sci. 2017, 39, 442–449. [Google Scholar] [CrossRef] [PubMed]
  173. Roy, P.; Parveen, S.; Ghosh, P.; Ghatak, K.; Dasgupta, S. Flavonoid Loaded Nanoparticles as an Effective Measure to Combat Oxidative Stress in Ribonuclease A. Biochimie 2019, 162, 185–197. [Google Scholar] [CrossRef]
  174. Anwar, A.; Masri, A.; Rao, K.; Rajendran, K.; Khan, N.A.; Shah, M.R.; Siddiqui, R. Antimicrobial Activities of Green Synthesized Gums-Stabilized Nanoparticles Loaded with Flavonoids. Sci. Rep. 2019, 9, 3122. [Google Scholar] [CrossRef]
  175. Gomes, F.; Henriques, M. Control of Bovine Mastitis: Old and Recent Therapeutic Approaches. Curr. Microbiol. 2016, 72, 377–382. [Google Scholar] [CrossRef]
  176. Mala, L.; Lalouckova, K.; Skrivanova, E. Bacterial Skin Infections in Livestock and Plant-Based Alternativ es to Their Antibiotic Treatment. Animals 2021, 11, 2473. [Google Scholar] [CrossRef]
  177. Cheng, W.N.; Han, S.G. Bovine Mastitis: Risk Factors, Therapeutic Strategies, and Alternative Treatments—A Review. AsianAustralas. J. Anim. Sci. 2020, 33, 1699–1713. [Google Scholar] [CrossRef]
  178. Mushtaq, S.; Shah, A.M.A.; Lone, S.A.; Hussain, A.; Hassan, Q.P.; Ali, M.N. Bovine Mastitis: An Appraisal of Its Alternative Herbal Cure. Microb. Pathog. 2018, 114, 357–361. [Google Scholar] [CrossRef]
  179. Tohma, H.; Gulcin, I.; Bursal, E.; Goren, A.; Alwasel, S.H.; Koksal, I. Antioxidant activity and phenolic compounds of ginger (Zingiber officinale Rosc.) determined by HPLC-MS/MS. Food Meas. 2017, 11, 556–566. [Google Scholar] [CrossRef]
  180. Moghadam, M.; Salami, M.; Mohammadian, M.; Khodadadi, M.; Emam-Djomeh, Z. Development of antioxidant edible films based on mung bean protein enriched with pomegranate peel. Food Hydrocoll. 2020, 104, 105735. [Google Scholar] [CrossRef]
  181. Krishnaiah, D.; Sarbatly, R.; Nithyanandam, R. A review of the antioxidant potential of medicinal plant species. Food Biproducts Process. 2011, 89, 217–233. [Google Scholar] [CrossRef]
  182. Ambika, P.P.S.; Chauhan, S.S.M. Activity-guided isolation of antioxidants from the leaves of Terminalia arjuna. Former. Nat. Prod. Lett. 2014, 28, 760–763. [Google Scholar] [CrossRef]
  183. Odeh, D.; Orsolic, N.; Berendika, M.; Dikic, D.; Drozdek, S.D.; Balbino, S.; Repajic, M.; Dragovic-Uzelac, V.; Jurcevic, I.L. Antioxidant and anti-atherogenic activities of essential oils from Myrtus communis L. and Laurus nobilis L. in rat. Nutrients 2022, 14, 1465. [Google Scholar] [CrossRef] [PubMed]
  184. Machado, A.; Ferreira, S.; da Silva Medeiros, F.; Fujiwara, R.; de Souza Filho, J.; Pimenta, L. Nematicidal activity of Annona crassiflora leaf extract on Caenorhabditis elegans. Parasites Vectors 2015, 8, 113. [Google Scholar] [CrossRef] [PubMed]
  185. Quílez, A.M.; Fernández-Arche, M.A.; García-Giménez, M.D.; De la Puerta, R. Potential therapeutic applications of the genus Annona: Local and traditional uses and pharmacology. J. Ethnopharm. 2018, 225, 244–270. [Google Scholar] [CrossRef] [PubMed]
  186. Yuan, G.; Guan, Y.; Yi, H.; Lai, S.; Sun, Y.; Cao, S. Antibacterial Activity and Mechanism of Plant Flavonoids to Gram-Positive Bacteria Predicted from Their Lipophilicities. Sci. Rep. 2021, 11, 10471. [Google Scholar] [CrossRef]
  187. Carvalho, M.T.B.; Araújo-Filho, H.G.; Barreto, A.S.; Quintans-Júnior, L.J.; Quintans, J.S.S.; Barreto, R.S.S. Wound Healing Properties of Flavonoids: A Systematic Review Highlighting the Mechanisms of Action. Phytomedicine 2021, 90, 153636. [Google Scholar] [CrossRef]
  188. Yang, X.; Jiang, Y.; Yang, J.; He, J.; Sun, J.; Chen, F.; Zhang, M.; Yang, B. Prenylated Flavonoids, Promising Nutraceuticals with Impressive Biological Activities. Trends Food Sci. Technol. 2015, 44, 93–104. [Google Scholar] [CrossRef]
  189. Mukai, R. Prenylation Enhances the Biological Activity of Dietary Flavonoids by Altering Their Bioavailability. Biosci. Biotechnol. Biochem. 2018, 82, 207–215. [Google Scholar] [CrossRef]
  190. Hošek, J.; Závalová, V.; Šmejkal, K.; Bartoš, M. Effect of Diplacone on Lps-Induced Inflammatory Gene Expression in Macrophages. Folia Biol. 2010, 56, 124–130. [Google Scholar]
  191. Shin, H.J.; Shon, D.H.; Youn, H.S. Isobavachalcone Suppresses Expression of Inducible Nitric Oxide Synthase Induced by Toll-like Receptor Agonists. Int. Immunopharmacol. 2013, 15, 38–41. [Google Scholar] [CrossRef]
  192. Dzoyem, J.P.; Nkuete, A.H.L.; Ngameni, B.; Eloff, J.N. Anti-Inflammatory and Anticholinesterase Activity of Six Flavonoids Isolated from Polygonum and Dorstenia Species. Arch. Pharm. Res. 2017, 40, 1129–1134. [Google Scholar] [CrossRef] [PubMed]
  193. Hu, J.; Liu, J. Licochalcone Attenuates Lipopolysaccharide-Induced Acute Kidney Injury by Inhibiting NF-KB Activation. Inflammation 2016, 39, 569–574. [Google Scholar] [CrossRef]
  194. Jia, T.; Qiao, J.; Guan, D.; Chen, T. Anti-Inflammatory Effects of Licochalcone A on IL-1β-Stimulated Human Osteoarthritis Chondrocytes. Inflammation 2017, 40, 1894–1902. [Google Scholar] [CrossRef]
  195. Cha, S.M.; Cha, J.D.; Jang, E.J.; Kim, G.U.; Lee, K.Y. Sophoraflavanone G Prevents Streptococcus mutans Surface Antigen I/IIInduced Production of NO and PGE2 by Inhibiting MAPK-Mediated Pathways in RAW 264.7 Macrophages. Arch. Oral Biol. 2016, 68, 97–104. [Google Scholar] [CrossRef]
  196. Wun, Z.Y.; Lin, C.F.; Huang, W.C.; Huang, Y.L.; Xu, P.Y.; Chang, W.T.; Wu, S.J.; Liou, C.J. Anti-Inflammatory Effect of Sophoraflavanone G Isolated from Sophora flavescens in Lipopolysaccharide-Stimulated Mouse Macrophages. Food Chem. Toxicol. 2013, 62, 253–361. [Google Scholar] [CrossRef]
  197. Song, M.; Liu, Y.; Li, T.; Liu, X.; Hao, Z.; Ding, S.; Panichayupakaranant, P.; Zhu, K.; Shen, J. Plant Natural Flavonoids against Multidrug Resistant Pathogens. Adv. Sci. 2021, 8, 2100749. [Google Scholar] [CrossRef]
  198. Bogdanova, K.; Röderova, M.; Kolar, M.; Langova, K.; Dusek, M.; Jost, P.; Kubelkova, K.; Bostik, P.; Olsovska, J. Antibiofilm Activity of Bioactive Hop Compounds Humulone, Lupulone and Xanthohumol toward Susceptible and Resistant Staphylococci. Res. Microbiol. 2018, 169, 127–134. [Google Scholar] [CrossRef]
  199. Hartkorn, A.; Hoffmann, F.; Ajamieh, H.; Vogel, S.; Heilmann, J.; Gerbes, A.L.; Vollmar, A.M.; Zahler, S. Antioxidant Effects of Xanthohumol and Functional Impact on Hepatic Ischemia-Reperfusion Injury. J. Nat. Prod. 2009, 72, 1741–1747. [Google Scholar] [CrossRef]
  200. Cho, Y.C.; Kim, H.J.; Kim, Y.J.; Lee, K.Y.; Choi, H.J.; Lee, I.S.; Kang, B.Y. Differential Anti-Inflammatory Pathway by Xanthohumol in IFN-γ and LPS-Activated Macrophages. Int. Immunopharmacol. 2008, 8, 567–573. [Google Scholar] [CrossRef]
  201. Lupinacci, E.; Meijerink, J.; Vincken, J.P.; Gabriele, B.; Gruppen, H.; Witkamp, R.F. Xanthohumol from Hop (Humulus lupulus L.) Is an Efficient Inhibitor of Monocyte Chemoattractant Protein-1 and Tumor Necrosis Factor-α Release in LPS-Stimulated RAW 264.7 Mouse Macrophages and U937 Human Monocytes. J. Agric. Food Chem. 2009, 57, 7274–7281. [Google Scholar] [CrossRef]
  202. Cho, Y.C.; You, S.K.; Kim, H.J.; Cho, C.W.; Lee, I.S.; Kang, B.Y. Xanthohumol Inhibits IL-12 Production and Reduces Chronic Allergic Contact Dermatitis. Int. Immunopharmacol. 2010, 10, 556–561. [Google Scholar] [CrossRef] [PubMed]
  203. Negrão, R.; Costa, R.; Duarte, D.; Gomes, T.T.; Coelho, P.; Guimarães, J.T.; Guardão, L.; Azevedo, I.; Soares, R. XanthohumolSupplemented Beer Modulates Angiogenesis and Inflammation in a Skin Wound Healing Model. Involvement of Local Adipocytes. J. Cell. Biochem. 2012, 113, 100–109. [Google Scholar] [CrossRef] [PubMed]
  204. Górniak, I.; Bartoszewski, R.; Króliczewski, J. Comprehensive Review of Antimicrobial Activities of Plant Flavonoids. Phytochem. Rev. 2019, 18, 241–272. [Google Scholar] [CrossRef]
  205. Sarbu, L.G.; Bahrin, L.G.; Babii, C.; Stefan, M.; Birsa, M.L. Synthetic Flavonoids with Antimicrobial Activity: A Review. J. Appl. Microbiol. 2019, 127, 1282–1290. [Google Scholar] [CrossRef] [PubMed]
  206. Wu, D.; Kong, Y.; Han, C.; Chen, J.; Hu, L.; Jiang, H.; Shen, X. D-Alanine:D-Alanine Ligase as a New Target for the Flavonoids Quercetin and Apigenin. Int. J. Antimicrob. Agents 2008, 32, 421–426. [Google Scholar] [CrossRef] [PubMed]
  207. Sirk, T.W.; Brown, E.F.; Sum, A.K.; Friedman, M. Molecular Dynamics Study on the Biophysical Interactions of Seven Green Tea Catechins with Lipid Bilayers of Cell Membranes. J. Agric. Food Chem. 2008, 56, 7750–7758. [Google Scholar] [CrossRef]
  208. Kusuda, M.; Inada, K.; Ogawa, T.O.; Yoshida, T.; Shiota, S.; Tsuchiya, T.; Hatano, T. Polyphenolic Constituent Structures of Zanthoxylum piperitum Fruit and the Antibacterial Effects of Its Polymeric Procyanidin on Methicillin-Resistant Staphylococcus aureus. Biosci. Biotechnol. Biochem. 2006, 70, 1423–1431. [Google Scholar] [CrossRef] [PubMed]
  209. Chan, E.W.C.; Wong, S.K.; Tangah, J.; Chan, H.T. Chemistry and Pharmacology of Artocarpin: An Isoprenyl Flavone from Artocarpus Species. Syst. Rev. Pharm. 2018, 9, 58–63. [Google Scholar] [CrossRef]
  210. Dej-Adisai, S.; Meechai, I.; Puripattanavong, J.; Kummee, S. Antityrosinase and Antimicrobial Activities from Thai Medicinal Plants. Arch. Pharm. Res. 2014, 37, 473–483. [Google Scholar] [CrossRef] [PubMed]
  211. Septama, A.W.; Panichayupakaranant, P. Synergistic Effect of Artocarpin on Antibacterial Activity of Some Antibiotics against Methicillin-Resistant Staphylococcus aureus, Pseudomonas aeruginosa, and Escherichia coli. Pharm. Biol. 2016, 54, 686–691. [Google Scholar] [CrossRef]
Figure 1. Wound-healing stages.
Figure 1. Wound-healing stages.
Pharmaceuticals 17 00303 g001
Figure 2. Secondary metabolites.
Figure 2. Secondary metabolites.
Pharmaceuticals 17 00303 g002
Figure 3. Selective solvents and groups of metabolites.
Figure 3. Selective solvents and groups of metabolites.
Pharmaceuticals 17 00303 g003
Figure 4. General structure of phenolic acids.
Figure 4. General structure of phenolic acids.
Pharmaceuticals 17 00303 g004
Figure 5. Other phenolic compounds.
Figure 5. Other phenolic compounds.
Pharmaceuticals 17 00303 g005
Figure 6. Hydrolysable and condensed tannins.
Figure 6. Hydrolysable and condensed tannins.
Pharmaceuticals 17 00303 g006
Figure 7. Most widely studied flavonoids.
Figure 7. Most widely studied flavonoids.
Pharmaceuticals 17 00303 g007
Figure 8. Prenylated flavonoids.
Figure 8. Prenylated flavonoids.
Pharmaceuticals 17 00303 g008
Table 1. Classification of a wound.
Table 1. Classification of a wound.
ClassificationType
CausePathological: resulting from a pathology (pressure ulcer, neoplasia).
Surgical or traumatic: resulting from surgery or trauma.
Iatrogenic: resulting from procedures or treatment with radiotherapy.
EvolutionAcute: wounds of easy resolution, rupture of vascularization, and mediate triggering of homeostasis (cuts, scoring, burns).
Chronic: long-lasting wounds (deviation from the physiological cicatricial process).
Presence of infectionClean: free of microorganisms.
Clean-contaminated: lesions less than 6 h between trauma and initial care.
Infected: presence of local infectious agent.
Contaminated: wounds serviced more than 6 h after trauma.
Regarding tissue
impairment
Stage I: skin integrates with signs of hyperemia, discoloration, or hardening.
Stage II: the epidermis and dermis are ruptured, with subcutaneous tissue showing hyperemia, blisters, and a shallow crater.
Stage III: total loss of cutaneous tissue, necrosis of the subcutaneous tissue to the muscular fascia.
Stage IV: great tissue destruction with necrosis reaching muscles, tendons, and bones.
Degree of opennessOpen: wounds in which the edges of the skin do not touch.
Closed: wounds where the edges of the skin are juxtaposed.
Table 2. Families and Genus with wound healing activity.
Table 2. Families and Genus with wound healing activity.
FamilyGenusPart Used/Type ExtractionCompoundsRef.
AcanthaceaeJusticia flavaLeaf/MethanolAlkaloids, Flavonoids, Glycosides, Tannins[41]
A. paniculataLeaf/10% aqueous extract Diterpenoids[42]
Amaranthaceae Achyranthes aspera Leaf/EthanolFlavonoids, Saponins, Tepernoids[43]
A. sessilis Stem and Leaf/Methanol 2,4-dihydroxy-2,5-dimethyl-3(2H)-furan-3-one, hexadecanoic acid, 2-1,2,4-trioxolane,3-phenyl-, palmitate-ethyl-, L-glutamic acid. [44]
A.triandra Air seed/Petroleum etherOil ricinoleic acid[45]
Celosia argentea Root/Dichloromethane and ethyl acetateTerpenoids[46]
Anacardiaceae Buchanania lanzan S. Root/Petroleum etherAlkaloids, Flavonoids, Polyphenols, Steroids[47]
Lannea welwitschii Hiern Leaf/MethanolAlkaloids, Flavonoids, Glycosides, Steroids, Tannins[41]
Apiaceae Angelica sinensis Leaf/Ethanoln-buthylidenephthalide and proteins[48]
Centella asiatica Leaf/MethanolAsiaticoside, Madecassic acid, Madecassoside asiatic acid, Triterpenes,[49]
Cuminum cyminum SeedsEssential oils[50]
L. striatum RhizomaEssential oils[51]
Apocyanaceae Catharanthus roseus Leaf/Aqueous and MethanolAlkaloids, Phenols, Proteins, Saponins, Tannins[52]
S. hispidus Leaf and RootAlkaloids, Flavonoids, Saponins, Tannins, [53]
Wrightia tinctoria Leaf/AqueousAlkaloids, Flavonoids, Phenolics, Saponins, Tannins[54]
Saba florida Leaf/Methanol 99.9%Total extract[55]
AraliaceaePanax ginsengPanax ginseng saponins (PGS)Ginsenoside Rb1 (G-Rb1)[56]
Panax notoginsengPanax notoginseng saponins (PNS)High-glucose (HG-30Mn)[57]
AsclepiadaceaeCalotropis gigantheaRoot BarkTaraxasteroryl isovalerate, Gigantin, Giganteol, Isogiganteol, α-amyrin-3-amyrin, Taraxasterol [58]
Calotropis proceraRoot bark/Ethanol Alkaloids, Flavonoids, Steroids, Tannins[59]
AsphodelaceaeAloe veraLeaf/Acetonic extractPolymers[60]
Asteraceae Achillea millefolium Aerial partsYarrow Oil[61]
Arctium lappa Ground bark/Ethanol Alkaloids, Flavonoids, Lignans, Phenolic acid, Tannins, Terpenoids[62]
Blumea balsamifera Leaf/Methanol 95%Flavonoids, Nonvalatile constituents[63]
Calendula officinalis Flowers/HydroethanolRutin, Quercetin-3-O-glucoside[64,65]
Carthamus tinctorius SaflowersHydroxysaflow yellow A (HSYA)[66]
Wedelia trilobata Leaves/Ethylacetate,
Chloroform:Methanol
Kaura-9(11),16-dien-19-oic acid[67]
Bignoniaceae Kigelia africana Leaves/Roots/MethanolFlavonoids, Carbohydrates, Sapogenetic glycosides, Saponins, Steroids[68]
S. campanulata Leaf/MethnolFlavonoids, Phenols, Saponins, Steroids[69]
Tecoma capensis Shoots/Hydroalcoholic Myrecetin[70]
Boraginaceae H. indicum Leaf/Ethanol Crude extract[71]
L. erythrorhizon RootPurification of Shikonin[72]
Burseracea Boswelia sacra Leaf/MethanolOil[73]
C. myrrha Leaf/MethanolOleo-gum-resins[73,74]
CactaceaeO. ficus-indicaSeed/Oil extractionOFI-SNEDDSs[75]
CaricaceaeCarica papayaPapaya fruit extractionCrude extract[76]
Cecropiaceae Cecropia peltata LeafSaponins[77]
Myrianthus arboreus Leaves/EthanolAlkaloids, Flavonoids, Glycosides, Sterols, Tannins, Terpenoids[78]
CaprifoliaceaLocinera japonicaFlowers/EthanolChlorogenic acid[79]
Combretaceae C. mucronatum Leaf/EthanolProcyanidin B2[80]
Terminalia chebula Fruit extractionAnthraquinone, Flavonoids, Sapogenins, Saponins, Steroids, Tannins[81]
Terminalia arjuna Fruit extraction/MethanolAnthraquinones, Carbohydrates, Flavonol, Glucose sorbitol, Hydrolyzable Tannins[82]
Crassulaceae Bryophylum pinnatum Lam Leaf/AqueousPatulitin-O-deoxy-hexoside-O-hexoside, Quercetin-O-hexoside, Quercetin-O-deoxy-hexoside-O-pentoside[83]
CyperaceaCyperus rotundus LAerial part/MethanolAlkaloids, Phenols[84]
Euphorbiacea Alchornea cordifolia (Schum & Thonn) Leaf/EthanolQuercetin, Hyperin, Guaijaverin[78]
Euphoria hirta Whole plant/MethanolAlkaloids, Flavonoids, Glycosides, Proteins, Saponins, Tannins[85]
Jatropha curcas L. Flowers/MethanolAlkaloids, Flavonoids, Glycoside, Saponins, Tannins[86]
Mallotus oppositifolius (Geiseler) Leaf/EthanolAspinidiol B, methylene bis-aspidinol, α-tocoferol[87]
P. emblica L. Leaves/EthanolFlavonoids, Saponins, Tannins[88]
P. muellerianus (Kuntze) Leaf/AqueousGeranin[89]
Fabaceae Astragalus membranaceus Sprants Seeds/EthanolTryptophan, Linoleic acid, Adenine[90]
Caesalpinia sappan L. WoodSappachalcone[91]
Entada phaseoloides Total Tannins[92]
Glycyrrhiza glabra L. Root/EthanolGlycyrrhiza cream[93]
Indigofera enneaphylla L. Whole plant/Petroleum ether, Ethyl Acetate, EthanolFlavonoids, Saponins, Tannins[94]
Mimosa pudica L. Seeds/Ethyl Acetate
Root/Petroleum ether
Alkaloids, Glycosides, Phytosterol [95,96]
Sophora flavescens Compound, Sophora flavescen lotion[97]
Tephrosia purpurea Aerial plants/EthanolFlavonoids, TPF-A 7 peaks[98]
FagaceaeQuercus infectoria OliverNutgails/EthanolPharmaceutical formulations[99]
GanodermataceaeGanoderma lucidumFruting bodies/Hot waterPolysaccharides 25.1% Ganodermic acid A[100]
Gentianaceae Anthocleista nobilis G. DonStem bark/Ethanol
Ethyl Acetate
Buthanol
n-Hexane
Isovitexin and Isovitexin-2”-O-xyl
Isovitexin
Apigenin monoglycoside
p-Hydroxybenzoic acid, Sarasinside
[101]
Ginkgoaceae Ginkgo biloba L. Leaf/AqueousMyricerin, Quercetin, Kaempferol, Isorhamnitin, Terpenes lactones, Ginkgolic acid[102]
Hypericaceae Hypericu mysorense Parts plant/Methanol Flavonoids, Saponins, Tannins[103]
IridaceaeCrocus sativus L.Stigmas/Glycerin/water/EthanolFlavonoids, Anthocyanins[104]
Lamiaceae Occimum sanctum L. Leaf/WaterEssential Oil[105]
Rosmarinus officinalis Aerial parts/HydrodistillationEssential Oil[106]
Salvia miltiorrhiza Leaf/HydroethanolicFlavonoids, Total Phenols[107]
Lauraceae Cinnamomum cassia Cinnamon Oils[74]
Liliaceae Allium cepa L. Onion/Ethanol 95%Alkaloids, Flavonoids, Phenols, Tannins[108]
Lycopodiaceae Lycopodium serratum Aerial parts/EthanolCrude etanol extract[109]
Lythraceae Lawsonia alba Leaf/MethanolCoumarin, Flavonoid, Steroid, Tannin, Terpenoid [110]
Lawsonia inermis L. Leaf/AqueousTotal Phenols, Total Flavonoids, Total Tannins, Saponins[111]
Punica granatum L. Fruit wholePomegranate are Tannins, Flavonoids, Punicic acid, Phytoestrogen[112]
Malvaceae Hibiscus rosa sinensis L. Flowers/MethanolPhenolic compounds, Flavonoids, Essential Oils, Anthocyanins[113]
Malva sylvestris Flowers/Ethanol:Water (80:20)Total phenolic, Flavonoids, Anthocyanin[114]
Thespesia populnea L. Fruit/AqueousGlycosides, Flavonoids, Alkaloids, Phytosterol, Quercetin, Rutin, Lupeal[115]
Martyniaccae Martynia annua Leaf/EthanolGlycosides, Phenols, Flavonoids, Tannins, Anthocyanins MAF-C 7 peaks[98]
Meliaceae A. indica A. Juss Steam bark/Water:EthanolCrude[116]
Carapa guianensis Aubl Andiroba seed oilLauric axid, Myristic, Palmitic acid, Stearic acid, Oleic acid, Linoleica cid, Lignoceric acid, Palmitoleic acid, Heptadecanoic acid, Arachidic acid, Behenic acid[117]
Mimosaceae Prosopis cineraria Leaves/Petroleum etherProtocatechuic acid, Caffeic acid, Chlorogenic acid, Ferrulic acid[118]
Moraceae Ficus religiosa L. Leaves/MethanolGlycosides, Alkaloids, Tannins, Terpenoids[119]
Moringaceae Moringa oleífera Lam. Leaves/EthanolFlavonoids, Phenolic acids[120]
Musaceae Musa sapientum L. Fruits/EthanolSaponins, Flavonoids, Glycosides, Steroids, Alkaloids[121]
Myrsinaceae Embelia ribes Burn. Fruits/Petroleum etherEmbelin[122]
Myrtaceae Eucalyptus globulus Leaves/Hydrodistillation1,8-cineole content 72.3%, α-pinone 9.4%[123]
Nymphaeaceae Nelumbo nucifera Aerial part/Ethanol30 peaks Ethanol,2-(-Octadecinyloxy, γ-sitosterol, Hexadecanoic acid[124]
Oleaceae Jasminum auriculatum Vahl.Leaves/Petroleum etherAlkaloids, Carbohydrates, Flavonoids, Phenolic compounds, Saponins, Steroids, Tannins, Tepernoids[125]
Jasminum grandiflorum L.Leaves/MethanolCrude[126]
Orchidaceae Bletilla striata Root/Boiled water Polysaccharide content (65.3%)[127]
Paeoniaceae Paeonia suffruticosa Bark root/Alcohol Flavonoids, Phenolic acid, Polysaccharide, Saponins[128]
Papaveraceae Argemone mexicana L. Fruits/MethanolAlkaloids, Flavonoids, Glycosides, Saponins, Steroids, Tannins, Terpenoids[129]
Papilionaceae Trigonella foenum-graecum Aerial part/MethanolFlavonoids[130]
Pedaliaceae Sesamum indicum Seed/EthanolSesame Oil[131]
Plantaginaceae Plantago Leaves/Distilled waterPolyphenolic compounds[132]
Polygonaceae Rheum officinale Powders/EthanolTMC extracts[133]
Potulacaceae Portulaca grandiflora Total plant/EthanolAlkaloids, Flavonoids, Saponins, Terpenoids[134]
Phyllanthaceae Bridelia ferruginea Benth. Leaves/Methanol
Stem barks/Ethyl Acetate
High phenolic content
High flavonoids content
[135]
Rosaceae Sanguisorba officinalis Polysaccharide[136]
Rubiaceae Morinda citrifolia L. LeafAlkaloids, Coumarins, Flavonoids, Saponins, Tannins, Triterpenes[137]
Rubia cordifolia L. 100 Compounds bicyclic peptides, terpenes, polysaccharides, Flavonoids, Quinones[138]
Rutaceae Aegle marmelos L. Flower/Ethanol 60%Aegelin, Cineol, Cuminaldheyde, Luvangetin, Eugenol[139]
Zanthoxylum bungeanum Maxim 140 constituents of this plantAlkaloids, Fatty acids, Flavonoids, Tepernoids, Flavonoids[140]
Salicaceae Casearia sylvestris Leaves/HydroalcoholicCrude extract[141]
Scrophulariaceae Rehmannia glutinosa Polysaccharides[142]
Stemonaceae Stemona tuberosa 9,10-dihydro-5-methoxy-8-methyl-2,7-phenanthrenediol[143]
Theaceae Camellia sinensis Tea leaves/Methanol [144]
Thymelaeaceae Daphne genkwa Sie. Diterpenoids/yuanhuapine[145]
Vitaceae Ampelopsis japonica Root/MethanolCatechin, Gallic acid, Kaempferol, Euscaphic acid, Resveratrol, Epicatechin[146]
Zingiberaceae Curcuma longa Linn Extracts Alkaloids, Flavonoids, Phenolic, Saponins, Terpenoids, Steroids[147]
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

Cedillo-Cortezano, M.; Martinez-Cuevas, L.R.; López, J.A.M.; Barrera López, I.L.; Escutia-Perez, S.; Petricevich, V.L. Use of Medicinal Plants in the Process of Wound Healing: A Literature Review. Pharmaceuticals 2024, 17, 303. https://doi.org/10.3390/ph17030303

AMA Style

Cedillo-Cortezano M, Martinez-Cuevas LR, López JAM, Barrera López IL, Escutia-Perez S, Petricevich VL. Use of Medicinal Plants in the Process of Wound Healing: A Literature Review. Pharmaceuticals. 2024; 17(3):303. https://doi.org/10.3390/ph17030303

Chicago/Turabian Style

Cedillo-Cortezano, Mayra, Luis Ruben Martinez-Cuevas, Jesús A. Márquez López, Ingrid L. Barrera López, Samantha Escutia-Perez, and Vera L. Petricevich. 2024. "Use of Medicinal Plants in the Process of Wound Healing: A Literature Review" Pharmaceuticals 17, no. 3: 303. https://doi.org/10.3390/ph17030303

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