Skip to main content

Antibacterial activity of medicinal plants and their role in wound healing

Abstract

Background

The study of plant-based medications, or phytomedicine, involves a wide spectrum of biological activities. Due to the existence of secondary metabolites, herbal medicine has been used and practiced throughout history for the treatment of both acute and chronic conditions. Over the past century or so, numerous novel compounds with medicinal potential have been derived from plants. In the age of growing super infections and the emergence of resistant strains, natural medicines are inspiring optimism.

Main body of the abstract

The review discusses the role of herbal medicine as antibacterial agents and their use in wound care and management of wounds and the critical role of secondary metabolites of herbal plants in fighting bacterial infections. Some medicinal plants such as St. John’s wort (SJW) (Hypericum perforatum), Rosemary (Rosmarinus officinalis), Ginger (Zingiber officinale), and nopal cactus (Opuntia ficusindica (L.)) also possess wide range of biological activities and can give a synergistic effect if combined with antibiotics. In addition, natural biopolymers play an important role in the management of wounds as well as the physiological processes of the skin (hemostasis, inflammation, proliferation, and remodelling).

Method

A narrative review of papers relevant to the use of phytomedicine in treating infections was conducted by using electronic databases PubMed, CrossREF, and Google Scholar.

Short conclusion

Phytomedicine is one of the top options for the treatment of chronic illnesses for millions of people around the world. To learn about the bioactive components of medicinal plants, their medical benefits, and their synergistic or additive effects to enhance the action of medications, substantial new studies are still needed.

Graphical abstract

Highlights

  • Phytomedicine involves a wide spectrum of biological activities.

  • Bioactive compounds extracted from plants are used for the treatment of both acute and chronic conditions.

  • Natural plant & secondary metabolites play a significant role in the treatment of bacterial infections.

  • Natural biopolymers are used in wound care and restoring physiological processes of the skin.

  • Substantial new studies are needed to learn about bioactive components medical benefits, and their synergistic or additive effects.

Background

An herbal remedy known as phytomedicine is utilized all over the world to treat or prevent physical and mental illnesses [1]. Herbal medicine or a phytopharmaceutical preparation is a type of medication that is made in a crude form solely from whole plants or specific plant parts [2]. Herbal medicine, which has a history spanning more than 3000 years and was enumerated in Sheng Nong's herbal book “The Devine Farmer's Classic of Herbalism” [3], is the foundation of traditional Chinese medicine (TCM). Herbal medicine is one of the most sought-after treatments by 3.5–4 billion people worldwide, mainly in Africa, India, and China, according to the World Health Organization (WHO) [4]. Products made from medicinal plants have greatly increased over the past 10 years, which has revolutionized and improved phytomedicine. There are about 35,000 plant species that are utilized as medicines, but only 20% of them go through the phytochemical analysis stage and only 10% make it to the biological screening stage, leaving the rest in need of further study [5]. As herbal medicine becomes more and more popular, it is important to maintain quality, safety, and to prevent potential toxicity [6,7,8]. In addition to long-term boiled extract and cold infusion of plants, plants can be extracted using alcoholic, vinegar, and hot water as well as other solvents, times, and temperatures [1]. Due to the existence of secondary metabolites, which are abundant in bioactive substances, herbs are used to treat both acute and chronic illnesses, including depression, cardiovascular disease, inflammation, and others [9, 10]. Plant constitutes are used directly as therapeutic agents or as models for pharmacologically active compounds or as starting materials for the synthesis of drugs (Table 1) such as morphine which was produced from opium extracted from Papaver somniferum, digoxin from Digitalis purpurea, antimalarials such as quinine from Cinchona bark, and over 60% of cancer therapeutics are based on natural products such as paclitaxel from the Pacific yew tree [11,12,13,14,15]. Due to inadequate research methodologies, time-consuming, and expensive isolation techniques, there is little information available regarding the composition of the majority of herbal medications. As a result, this page discusses the crucial role of medicinal plant extract in the battle against bacterial infections and the management of skin wounds and disorders such as atopic dermatitis (AD) and diabetic foot ulcers (DFUs). Additionally, extracellular matrix (ECM) and biopolymers derived from microorganisms, animals, and plants (cellulose, hyaluronic acid, collagen, alginate, and chitosan) all have bioactive qualities that make them useful in the treatment of wounds and the healing process.

Table 1 List of drugs derived from plant origin and their clinical use
Fig. 1
figure 1

Chemical structure of drugs derived from plant origin; Aescin, aesculetin, agrimophol, allyl isothiocyanate, anisodamine, artemisinin, aspirin, atropine, berberine, bromelain, camphor, camptothecin, catechin, cocaine, codeine, colchicine. Convallatoxin, curcumin, deslanoside, digoxin, emetine, ephedrine, etoposide, and Galantamine

Fig. 2
figure 2

Chemical structure of drugs derived from plant origin cont., glaucarubin, glaucine, glycyrrhizin, gossypol, hesperidin, hyoscyamine, irinotecan, L-Dopa (Levodopa), morphine, ouabain, paclitaxel, papain, papaverine, physostigmine, pilocarpine, pseudoephedrine, quinine, reserpine, and scopolamine

Fig. 3
figure 3

Chemical structure of drugs derived from plants cont. sennosides A, B, tetrahydrocannabinol (THC), theophylline, thymol, tubocurarine, and yohimbine

Method

We evaluated the scientific literature collected from PubMed, CrossREF, and Google Scholar databases, considering all the articles published between 1975 and 2023. The keywords used were “herbal plants”, “phytomedicine”, “secondary metabolites”, “synergistic”, “antibacterial”, and “wound healing”. Article titles and abstracts were manually screened, and studies not related to the topic were excluded.

Main text

Antibacterial activities of medicinal plants

Globally high rates of morbidity and mortality are mostly caused by infectious diseases. Millions of people per year die as a result of the advent of bacterial strains that are resistant [81]. Antibiotic resistance in bacteria evolves through intrinsic or acquired resistance, by chromosomal mutation, or by horizontal gene transfer (HGT) [82, 83]. Several mechanism can lead to the development of antibiotic resistance such as alteration in cell membrane permeability either by reducing antibiotic penetration or increasing its elimination by efflux pumps; moreover, bacteria can deactivate the antibiotic itself or modify the antibiotic targets, in addition to other alternative pathways which were described in literature and are illustrated in Fig. 4 [83,84,85,86,87,88]. The most harmful microorganisms for human health have been recognized by the World Health Organization (WHO) and are categorized into three priority groups: critical pathogens (AcinetobacterEnterobacteriaceaeand Pseudomonas), high-priority pathogens (Campylobacter, Enterococcus faeciumHelicobacter pyloriNisseria gonorrhoeae, Staphylococcus aureusSalmonella spp.), and medium-priority pathogens (Streptococcus pneumoniaeShigella spp.) [83, 89,90,91]. Pathogenic plant bacteria can cause diseases on susceptible plant hosts which starts usually with low numbers of pathogen cells and then colonize and multiply to large amounts in living plant tissue. This results in the alteration of plant’s developmental system which eventually leads to reduction of plant growth and yield. Disease severity depends on the host genetic constitute, environmental conditions, and the pathogen [92]. Herbal plants produce unlimited wide variety of secondary metabolites which are mostly aromatic and phenol derivatives that gives them the ability to safeguard plants against pathogens [9, 93, 94]. Plants use oxygen for their growth and development but in stress like pathogen attack; the usage of oxygen causes the production of reactive oxygen species (ROS) in the plant and results in photo-oxidative damage [95,96,97]. In stress conditions plants induce excessive biochemical changes to activate defence pathways such as changing cell wall composition, detoxification of several ROS species, induction of enzymatic and nonenzymatic components, and alteration of pathogen activates [98,99,100,101]. Numerous studies have demonstrated that various chemicals components of herbal plants possess antibacterial properties that can protect the human body against diseases without being damaging to cells [102]. Herbal or synthetic antimicrobial agents are both possible. The main negative effect of synthetic substances including antibiotics, metals, and metal oxide nanoparticles is the production of ROS, which is extremely hazardous and can lead to cancer [103]. On the other hand, herbal antimicrobial compounds including cinnamon, thyme, chamomile, eucalyptus, lemon balm, garlic, ginger, and others are free scavengers that can prevent ROS generation [102]. Antimicrobial phytochemicals can be divided into several categories such as phenolics and polyphenol (e.g. catechol and caffeic acids (Fig. 5)) [104]; terpenoids and essential oils (e.g. camphor, farnesol and artemisinin (Fig. 5)) [104, 105]; alkaloids (e.g. morphine (Fig. 5)) [104]; lectins and polypeptides (e.g. Thionins (Fig. 5)) [106,107,108]; polyacetylenes (e.g. 8S-heptadeca-2(Z),9(Z)-diene-4,6-diyne-1,8-diol (Fig. 5)) [109], and many others.

Fig. 4
figure 4

Structure of a gram-negative bacteria and its mechanism of resistance and b gram-positive bacteria and its resistance mechanism

Fig. 5
figure 5

Chemical structure of antimicrobial phytochemicals including catechol, caffeic acids, camphor, farnesol, artemisinin, morphine, thionins, 8S-heptadeca-2(Z),9(Z)-diene-4,6-diyne-1,8-diol, luteolin, quercetin, epicatechin, benzoic, cinnamic acids, tyrosol, hydroxytyrosol, sulphoraphane, proanthocyanidin (TPA), quercetin 3-O-glucoside, quercetin pentosides, gallic acid, naringenin, salicylic acid, kaempferol, astragalin, tellimagrandin I, II, rugosin D, and spiraeoside

Antibacterial phytochemicals

Phenolics and polyphenol

Polyphenols are a wide class of chemical substances found in plants that have a variety of biological functions [110]. Flavonoids (such as luteolin (Fig. 5), quercetin (Fig. 5), catechin (Fig. 2), and epicatechin (Fig. 5)) and nonflavonoids (such as benzoic, cinnamic acids, tyrosol, and hydroxytyrosol (Fig. 5)) are two categories of polyphenols that can be simple, large, or complex compounds[111]. Phenolic compounds have been used in traditional medicine, and although their mode of action as antibacterial agents is still not completely understood, it does appear to be distinct from that of antibiotics, reducing the risk of cross-resistance and making them a promising agent against resistant pathogens [112,113,114]. Inhibiting the growth of Helicobacter pylori is one of the potential benefits of polyphenols, which are powerful antioxidant and anti-inflammatory compounds found in foods like broccoli, garlic, liquorice, cranberries, and curcumin [115, 116]. Due to antibiotic resistance and issues with patient compliance, H. pylori eradication rates with triple therapy, which includes clarithromycin, metronidazole, or amoxicillin in combination with a proton pump inhibitor, are lower than 50–70%. The bismuth-containing quadruple therapy, as well as sequential and hybrid regimens, is now advised for treating H. pylori infection. There is not currently a therapy with > 90% eradication rates, though [117,118,119,120,121]. To determine the impact of polyphenol components (garlic, liquorice, cranberry, curcumin, and broccoli) on the elimination of H. pylori infection, Wang et al. [122] conducted a meta-analysis study. The study discovered that polyphenol compounds may have a positive impact on the elimination of H. pylori, since the total elimination rate of the bacteria was higher in the polyphenol compounds group than in the control group. The research backs the adjuvant use of polyphenolic substances in the treatment of H. pylori. The polyphenol content of garlic makes it a potent alternative to conventional H. pylori treatment, according to in vitro studies [123,124,125]. Additionally, liquorice has anti-H. pylori properties [126]. Studies conducted in vitro revealed that Glycyrrhiza glabra and its aqueous extract have anti-H. pylori activity. Possible mechanisms of action include blocking dihydrofolate reductase, DNA gyrase, protein synthesis, and H. pylori adherence to human stomach tissue. Additionally, the components of cranberry juice prevent the adherence of a number of pathogenic infections, including the influenza virus, E. coli, and H. pylori [127,128,129]. According to in vitro research, H. pylori was prevented from proliferating and was caused to produce a coccoid form by the polyphenols in cranberry extract [130,131,132]. Turmeric root contains the poly-phenolic compound curcumin (diferuloylmethane) (Fig. 5) [133]. Curcumin administration significantly reduced inflammation in gastric mucosa infected with H. pylori, according to animal studies [134]. Additionally, in vivo administration of sulphoraphane-rich fresh broccoli (Fig. 5) sprouts to mouse models reduced H. pylori colonization and prevented lipid peroxidation in the stomach mucosa [135, 136]. Żurek et al. [137] investigated the biological potential of polyphenolic substances isolated from walnut (Juglans regia L.). The edible kernels of walnuts, which also include polyunsaturated fatty acids (PUFA), tocopherols, phytosterols, and vitamins in addition to a number of other bioactive chemicals, are a rich source of polyphenols [138,139,140,141]. In addition, other parts of walnuts such as leaves are valuable products as well and their content was assessed spectrophotometrically by UPLC-PDA-MS/MS method. Several phenolic compounds were identified and expressed as total phenolic (TPC), proanthocyanidin (TPA) (Fig. 5), and total flavonoid contents (TFC) with quercetin 3-O-glucoside (Fig. 5) and quercetin pentosides (Fig. 5) dominating. Aqueous walnut leaf extracts antibacterial and antifungal activities were tested against Gram-negative bacteria (E. coli, Klebsiella pneumoniaePseudomonas aeruginosa), Gram-positive bacteria (Staphylococcus aureusEnterococcus faecalis, Streptococcus pyogenes), and fungus. (C. albicans). Result showed highest susceptibility (at 10 mg/ml concentration) against K. pneumoniae, and S. pyogenes, less bactericidal activity against gram-positive and without any antifungal effect. These findings may be explained by the fact that gram-negative bacteria's cell walls contain lipopolysaccharide (LPS), which creates a hydrophilic environment and shields it from hydrophobic molecules. The aqueous and ethanol extracts of walnut leaves may contain more hydrophilic compounds as a result, leading to higher inhibitory activity against gram-negative bacteria [137]. In similar context, Bouslamti et al. [142] tested antioxidant and antibacterial activity of Solanum elaeagnifolium leaf and fruit extracts against Gram-positive (Staphylococcus aureus and Bacillus subtilis), Gram-negative bacteria (Escherichia coli and Proteus mirabilis), and Candida albicans. HPLC and colorimetric methods were used to determine the chemical composition of S. elaeagnifolium fruits and leaf extracts which are quercetin (Fig. 5), luteolin (Fig. 5), gallic acid (Fig. 5), and naringenin (Fig. 5). Result showed that the extracts generated good antioxidant activity and potent antifungal activity. Nonetheless, further studies are needed to assess potential adverse effects. Meadowsweet plant (Filipendula ulmaria (L.) Maxim.) has been also used in traditional medicine due to its wide range of pharmacological effects [143, 144]. For the biological activity of meadowsweet, phenolic secondary metabolites are thought to be responsible, including phenolic acids and their derivatives (such as gallic acid and salicylic acid in Fig. 5), flavonoids and flavonoid glycosides (such as kaempferol and astragalin in Fig. 5), and tannins (such as tellimagrandin I, II, and rugosin D) [145, 146]. Meadowsweet extracts from different organs and parts (leaves, flowers, fruits, and roots) have been studied for their antioxidant and antibacterial properties by Savina et al. [147] Gram-negative bacteria P. aeruginosa and Gram-positive bacteria B. subtilis were used to investigate the antibacterial activity of flower and fruit extracts. All portions of the meadowsweet plant had a high overall phenol level, whereas the flowers had a high flavonoid concentration. The primary flavonoids in meadowsweet include luteolin, kaempferol derivatives, and spiraeoside (Fig. 5), which all have potent antibacterial properties. Meadowsweet plants were also found to have significant amounts of salicylic acid and its derivatives, which are thought to have anti-inflammatory effects. Furthermore, it was shown that meadowsweet roots had greater total catechin and proanthocyanidin contents than other sections, while the fruits had higher total tannin contents, particularly tellimagrandins I and II and rugosin D. All of the components had anti-inflammatory and antibacterial effects.

Terpenoids and essential oils (EOs)

More than 17,500 plant species can produce essential oils (EOs), which are volatile secondary metabolites with a particular flavour or scent [148]. The cytoplasm and plastid of plant cells produce EOs compounds, which are then stored in intricate secretory structures including glands and secretory cavities before being present as liquid drops in the flowers, leaves, stems, fruits, bark, and roots of plants. In addition to many other substances like fatty acids, oxides, and derivatives of sulphur, the primary constituents of EOs include terpenes, terpenoids, and phenylpropanoids [149, 150]. EOs are produced through mechanical cold pressing of plants, steam distillation, dry distillation, hydrodistillation, and more recent techniques including microwaves and supercritical fluid extraction. Depending on the method used, different chemical compositions of EO were obtained [150,151,152]. Due to their biological characteristics, including their antibacterial qualities, EOs have been utilized as perfumes, food spices, and in folk medicine [153]. Terpenes, which make up the majority of essential oils (EOs), are generated from the isoprenoid pathway and are made up of isoprene units (C5). On the basis of this, terpenes are classified as monoterpenes (C10), sesquiterpenes (C15), diterpenes (C20), triterpenes (C30), and carotenoids (C40). Terpenes have a variety of chemical properties, including alcohol (terpineol, menthol, carveol, linalool, and citronellol; Fig. 6), aldehyde (citral and citronellal; Fig. 6), ketone (carvone; Fig. 6 and camphor; Fig. 5), phenol (thymol; Fig. 3; and carvacrol; Fig. 6); ether (eucalyptol; Fig. 6), and hydrocarbon (pinene, and limonene; Fig. 6) groups [154, 155]. An antibacterial, antifungal, antioxidant, anti-inflammatory, analgesic, antimutagenic, and wound-healing compound, thymol is a phenolic monoterpene. Due to its strong antibacterial characteristics, it also enhances digestion, lessens respiratory issues, and is used in dentistry to treat infections of the oral cavity [156]. Majorana syriaca, sometimes known as thyme, is a widespread east Mediterranean aromatic species with a high content of essential oils that give the plants their distinct flavour and perfume as well as antibacterial and antifungal properties. Due to its high volatile oil content, M. syriaca is used in traditional medicine to cure the flu, colds, and cough. The primary essential oil in thyme is thymol, which is used in mouthwashes as an antibacterial. Additionally, thyme extracts are added to cough syrups to treat coughs and other respiratory issues like bronchial issues. Thyme essential oils' high phenol concentration is what gives them their potent antibacterial properties, allowing for usage as a potent disinfectant, in oral medicinal preparations, and as a flavouring ingredient in numerous food products [157,158,159,160,161]. Abu-Lafi et al. [162] used static headspace-gas chromatography/mass spectrometry (SD-GCMS) analysis to determine essential oils in thyme leaves and identified 29 monoterpenes (oxygenated and hydrocarbons) such as thymol, carvacrol, a-phellandrene, a-pinene, c-terpinene, o-cymene, p-cymene, and b-myrcene (Fig. 6). The primary components of oxygenated monoterpenes in thyme leaves were the phenolic substances thymol and its geometric isomer carvacrol. The investigation also revealed the presence of thymoquinone, which is known to play a protective function against oxidative damage brought on by chemicals that produce free radicals, such as carbon tetrachloride. Free terpenes present in EOs were investigated for their antibacterial activity by Guimares AC et al. [154]. Results showed that hydrocarbons such terpinene, camphene, R-(-)-limonene, and (+)-α-pinene have inferior antibacterial action to oxygenated terpenes like phenolics (Fig. 6), which is consistent with the findings of earlier studies [163,164,165,166,167]. Eugenol and terpineol showed rapid and excellent bactericidal action against Salmonella enterica and S. aureus strains, respectively. Moreover, carveol, citronellol, and geraniol exhibited rapid bactericidal effect against E. coli. Therefore, hydroxyl groups in phenolic and alcohol compounds resulted in higher antimicrobial activity than hydrocarbons [154]. The antibacterial properties of EOs from Cinnamomum cassia bark and Eucalyptus globulus leaves are well established. These EOs' main secondary metabolites, 1,8-cineole (Fig. 6) and trans-cinnamaldehyde (Fig. 6), are what provide these compounds their therapeutic properties. However, ethnobotanical physicians prefer the use of entire EOs over purified components to treat bacterial infections [168,169,170]. Therefore, a set of 6 g-positive and -negative bacteria were used by Nguyen HTT et al. [171] to evaluate the antibacterial activity of plant EOs to their separated main components. According to the findings, entire oils of eucalyptus and cinnamon with low concentrations of 1,8-cineole (61.2%) and trans-cinnamaldehyde (89.1%) have more favorable effects than the active components that have been refined to less than 99%. Additionally, CC crude extract had greater and stronger effects on both gram-positive and gram-negative bacteria compared to EG. The study's findings support the use of complete essential oils for bacterial infections in traditional medicine since they offer advantages over isolated constituents that might not have the same effects when used as medications. To combat foodborne infections, EOs can also stop bacterial growth [172]. Another study by Trinh et al. [173] looked at the antibacterial properties of trans-cinnamaldehyde, the major component of Cinnamomum cassia essential oil, in relation to the Listeria innocua strain. The accumulation of trans-cinnamaldehyde in the hydrophobic core of the cytoplasmic membrane of L. innocua, which results in membrane disruption, was found to be the cause of the antibacterial action of trans-cinnamaldehyde as well as other minor C. cassia essential oil. Although there was no evidence of large hole creation or cell lysis, viable but nonculturable (VBNC) cells did start to appear. To use C. cassia EO and trans-cinnamaldehyde rationally in food preservation, a better understanding of their modes of action is required. Kolozsváriné Nagy J et al. [174] tested EOs of cinnamon, eucalyptus, thyme, clove, tea tree, rosemary, lemon grass, lemon balm, and citronella grass against Xanthomonas arboricola pv. pruni (Xap) which is responsible for water-soaked spots on the surface of leaves, and fruits that may lead to severe infections and complete destruction of the plant. High-performance thin-layer chromatography (HPTLC)-Xap combined with solid-phase microextraction-gas chromatography/mass spectrometry (SPME-GC/MS) was used to identify active EOs’ components. All EOs inhibited bacterium isolates with superiority to cinnamon with MIC values of 31.25 µg/mL and 62.5 µg/mL, while the tea tree EO was the least effective with the highest MIC values. Compounds that are identified to have antibacterial activity by HPTLC zones are trans-cinnamaldehyde in cinnamon, thymol in thyme, eugenol in clove, terpinen-4-ol (Fig. 6) in tea tree, borneol in rosemary (Fig. 6), citral in lemon grass and lemon balm, and citronellal and nerol in citronella grass (Fig. 6). Biofilms produced by bacteria are one of the reasons for the resistance of bacteria and can act as reservoirs for spoilage bacteria in food such as Shewanella putrefaciens which is the chief spoilage bacteria in fish [175, 176]. Therefore, Xie et al. [177] studied antibacterial effectiveness of Ocimum gratissimum L. essential oil (OGEO) in vitro against Shewanella putrefaciens to be used as natural preservative. The main active ingredients of OGEO are eugenol and caryophyllene (Fig. 6) which have antibacterial activity according to previous studies. The study demonstrated that OGEO had a positive inhibitory effect on the growth of S. putrefaciens and act by disrupting the formation of biofilms and cell membranes of the bacteria with minimum inhibitory concentration and minimum bactericidal concentration of 0.1%. inhibiting S. putrefaciens by EOs provide a new method of inhibiting the spoilage of food. Ocimum basilicum (Great basil or Saint-Joseph’s wort) from Lamiaceae family contains many bioactive secondary metabolites such as polyphenols, flavonoids, and terpenes. Main EOs included in the plan are linalool, methyl estragole, methyl cinnamate, and methyl chavicol (Fig. 6) [178,179,180,181]. Ocimum basilicum has many pharmacological activities and has been used in traditional medicine as antioxidant, anticancer, antiviral, antiaging, and antimicrobial properties [182,183,184]. Studies showed that the biological activity of O. basilicum is related to cinnamic acid (Fig. 5) derivative of the polyphenoid rosmarinic acid (Fig. 6) [185]. Eid et al. [186] studied O. basilicum seeds essential oil biological activity and showed that it had a broad-spectrum antibacterial activity. The oil suppressed the development of all tested microbial strains (minimum inhibitory concentrations (MICs) between 1 and 2.3 µg/mL) and fungus strain C. albicans (MICs of 1.3 µg/ml for fungus). Antibacterial activity may refer to the presence of phenolic components in the essential oil which trigger intracellular ATP and potassium ion leakage and lead to cell death. Yaldiz et al. [187] also investigated antibacterial, antiquorum sensing, and antibiofilm capabilities of the ethanol extract and essential oil derived from O. basilicum. Results revealed that in addition to having antifungal effects on C. albicans, basil essential oil also demonstrated antibacterial activity and antiquorum sensing activity against some Gram-positive and -negative bacterial species.

Fig. 6
figure 6

Chemical structure of essential oils (EOs) and terpenoids including terpineol, menthol, carveol, linalool, citronellol, citral, carvone, carvacrol, eucalyptol, a-pinene,limonene, a-phellandrene, c-terpinene, o-cymene, p-cymene, b-myrcene , camphene, eugenol, 1,8-cineole, terpinen-4-ol, nerol, caryophyllene, methyl estragole, methyl cinnamate, methyl chavicol, and rosmarinic acid

Alkaloids

Alkaloids are a wide and diverse group of secondary metabolites found in 300 plant families and can be found as well in bacteria, fungi, and animals. Alkaloid name came from their basic nature and is characterized by the presence of a basic nitrogen atom in the form of a primary (RNH2), secondary (R2NH), or tertiary amine (R3N). Alkaloids can be monomers or oligomers and can be classified into three major categories: true-alkaloids with N-atom in the heterocycle, proto-alkaloids without N-atom, and pseudo-alkaloids with a basic carbon skeleton [112, 188,189,190,191,192]. Heterocyclic alkaloids can be divided into 14 subgroups that include, pyrrolizidines, pyrrolidines, indoles, isoquinolines, quinolizidines, purines, piperidines, tropanes, and imidazoles [193]. A rich source of bioactive compounds is Marine invertebrates which developed a defence chemical system to protect themselves from predation. Marine sponges are a rich source of secondary metabolites which are responsible for their numerous biological activities such as anti-inflammatory,anticancer, antiviral, antibacterial, and anticoagulant activities [194,195,196]. The sponge Luffariella variabilis exhibit antibacterial activity against gram-positive bacteria due to the presence of sesterterpenes manoalide, secomanoalide,and trans- and cis-neomanoalide (Fig. 7) [197]. Moreover, the sponges Poecillastra sp. and Jaspis sp., contain psammaplin A (Fig. 7), a bromotyrosine-derived natural product, while the sponge Siliquaria sp. contain the motualevic acids (A–F) (Fig. 7) a halogenated glycyl-lipid conjugates which all inhibit gram positive-bacteria only and are inactive against gram-negative bacteria [198, 199]. Antibacterial and antibiofilm activity of another group called the pyrrole-imidazole alkaloids was studied and tested against gram-positive and -negative bacteria. Bromoageliferin (Fig. 7) is a member from pyrrole-imidazole alkaloids family isolated from marine sponges with other members such as oroidin and sceptrin (Fig. 7). Reports documented the inhibition of Rhodospirillum salexigens SCRC 113 biofilms formation a marine bacterium by oroidin and bromoageliferin which resulted in using them as templates to develop numerous analogues and study their antibiofilm activity [200,201,202,203,204,205,206,207,208,209]. Melander et al. [210] studied pyrrole-imidazole alkaloids (monomeric and dimeric alkaloids) ability to inhibit biofilm formation and suppress antibiotic resistance against gram-negative Acinetobacter baumannii and gram-positive methicillin-resistant S. aureus (MRSA). Result showed that monomeric alkaloid oroidin exhibited modest activity against both strains, while stevensine (monomeric oroidin analogue) (Fig. 7) was inactive. On the other hand, the dimeric alkaloids Sceptrin was slightly more active against A. baumannii, while dibromosceptrin (Fig. 7) and bromoageliferein were both more active against MRSA with bromoageliferin as the most potent compound. Result indicated that both monomeric and dimeric alkaloids can inhibit phenotypic and genotypic bacterial resistance mechanisms; therefore, a high-throughput screening is needed to identify marine natural compounds to be used as adjuvants with antibiotics to restore FDA-approved antibiotics efficacy and to find alternative approaches to combating MDR bacteria. Tuberculosis (TB) is a widespread infectious disease, and due to the increase of multidrug-resistant (MDR) and extensively drug-resistant (XDR) strains new antiTB agents are needed. Arai et al. [211] isolated several compounds from marine sponge of Haliclona sp. such as tetracyclic alkylpiperidine alkaloid, 22-hydroxyhaliclonacyclamine B, and haliclonacyclamine A and B alkaloids (Fig. 7) to be used as antidormant mycobacterial compounds. Result showed strong antimycobacterial activity under both aerobic and hypoxic condition against Mycobacterium smegmatis and M. bovis Bacille de Calmette et Guérin (BCG) from both haliclonacyclamine. Haliclonacyclamine B exhibited bactericidal activity against M. bovis(BCG), while hydroxyhaliclonacyclamine B showed weaker antimicrobial activities against Mycobacterium bacilli and reduced antimycobacterial activity which may be due to the presence of the 22-hydroxy group. Wijaya et al. [212] studied Dicranostigma franchetianum plant from Papaveraceae family and isolated a wide spectrum of isoquinoline alkaloids (IAs) such as berberine, chelerythrine, protopine, and sanguinarine (Fig. 7). Alkaloids were tested against Mycobacterium tuberculosis H37Ra and four other mycobacterial strains. Most isolated alkaloids exhibited weak or no antimycobacterial activity; however, benzophenanthridine (6-ethoxydihydrochelerythrine) and bisbenzophenanthridine (bis-(6-(5,6-dihydrochelerythrinyl))ether) alkaloid derivatives (Fig. 7) showed moderate antimycobacterial activity against all tested strains. Moreover, semisynthetic berberine derivatives resulted in a significant increase in antimycobacterial activity against all tested strains. Further studies are needed to develop more potent berberine derivatives with low cytotoxic profile. On the other hand, Dong et al. [213] conducted a bioassay-guided phytochemical study on the semi-mangrove plant Myoporum bontioides A. Gray, which belong to Myoporaceae family and tested its activity against methicillin-resistant S. aureus (MRSA). New sesquiterpene alkaloids and furanosesquiterpenes were isolated from the plant, and result showed that sesquiterpene alkaloids displayed potent anti-MRSA activity. Alkaloids also can be used in food system to prevent or treat foodborne diseases such as oliveridine and pachypodanthine (Fig. 7) an aporphinoid alkaloids. Marco Di et al. [214] tested these alkaloids against Yersinia enterocolitica an important foodborne pathogen that cause a gastrointestinal disease in humans called yersiniosis. Result showed that both oliveridine and pachypodanthine inhibited the growth of Y. enterocolitica with superiority for oliveridine with lower MIC values which open an opportunity to develop potential antimicrobial agents to prevent or treat foodborne diseases.

Fig. 7
figure 7

Chemical structure of alkaloids compounds including secomanoalide, neomanoalide, psammaplin A, motualevic acids (AF), bromoageliferin, oroidin, sceptrin, stevensine, dibromosceptrin, 22-hydroxyhaliclonacyclamine B, haliclonacyclamine A and B, berberine, chelerythrine, protopine, sanguinarine, 6-ethoxydihydrochelerythrine, bis-(6-(5,6-dihydrochelerythrinyl))ether, oliveridine, and pachypodanthine

Lectins and polypeptides

Naturally antimicrobial proteins and peptides can be found in humans, animals, plants, and microorganisms. Plants include proteins called lectins that attach to certain carbohydrates on the surfaces of microbes, aiding in the defensive mechanisms against pathogens. Plant lectins are classified according to their affinity for monosaccharides and complex glycans, but they do not interact with endogenous cellular glycans. Instead, they have a strong affinity for the sugars on bacteria, fungi, and other organisms, which suggests that they serve as a plant defence molecule [215,216,217,218]. Lectins have different types including C-type lectins (e.g. endocytic lectins), S-type lectins (e.g. galectins), L-type lectins, P-type lectins, M-type lectins, Jacalin-related lectin (JRL),siglecs, Oscillatoria agardhii agglutinin homolog (OAAH), Cyanovirin-N homologs (CVNHs), Galanthus nivalis agglutinin-like (GNA-like) lectins, and others [219]. Lectins have been used in medicine as immunomodulators against tumour cells and microbial infections. Moreover, lectins showed to be able to disrupt quorum sensing (QS) signal transduction and interfere with nonessential functions for cell viability [220, 221]. Lectins purified from Moringa oleifera leaf (SLL-1, SLL-2, and SLL-3) showed variable antibacterial potency against E. coliShigella dysenteriae, and S. aureus, while water-soluble lectin purified from Moringa oleifera seeds significantly reduced S. aureus but not E. coli [222, 223]. On the other hand, mannose-glucose-binding lectin isolated from Calliandra surinamensis leaf did not kill S. aureus nor Staphylococcus saprophyticus but reduced their growth and their biofilm formation with no activity against E. coli [224]. In addition, a rich source of lectins is Marine species that include green algae (22%), red algae (61%), and cyanobacteria (17%) [225]. Few literature studies discussed the potential usefulness of algal or cyanobacterial lectins as antibacterial agents [226,227,228]. Purified lectins isolated from two red algal species, Eucheuma serra (ESA) and Galaxaura marginate (GMA), showed strong inhibitory action against marine gram-negative Vibrio vulnificus, while no action recorded for other two Vibrio species, V. peagius and V. neresis according to Liao et al. [229] study. Selectivity in the inhibition is referred to the differences in bacterial surface carbohydrates. Furthermore, Hung et al. [230] isolated lectins from Eucheuma denitculatum (EDA) red algae which also exhibited activity selectivity against V. alginolyticus, but not against V. parahaemolyticus or V. harveyi due to the binding of lectins to high-mannose N-glycans. Moreover, Holanda et al. [231] isolated lectins from Solieria filiformis red alga and tested its activity against gram-negative and -positive bacteria. Result showed that lectins inhibited the growth of the gram-negative species; Salmonella typhi, Serratia marcescens, Klebsiella pneumoniae, Pseudomonas aeruginosa, Enterobacter aerogenes, and Proteus sp., while it stimulated the growth of the gram-positive species Bacillus cereus. No activity was noticed against S. aureus, B. subtilis, E. coli, and Salmonella typhimurium. The interaction between the S. filiformis lectin and the gram-negative bacteria's cell surface receptors, which encouraged changes in the flow of nutrients, may be the cause of the activity against gram-negative bacteria. These findings raised the possibility of using marine lectins as organic substitutes for antibiotics in the treatment of gram-negative pathogens [232]. As mediators of innate immunity in all living things, antimicrobial peptides (AMPs) or host defence peptides are also recognized to play a significant role in biological processes. Because AMPs are cationic and amphipathic, they can more easily pass through microbial cytoplasmic membranes and cause cell lysis. The usage of AMPs is constrained for a variety of reasons, including their undesirable cell toxicity, restricted availability, high cost during synthesis, sensitivity to protease degradation, and others [233, 234]. Compared to other species, plants have more AMPs, which typically have 20–50 amino acid residues and are abundant in glycine, cysteine, and positively charged residues [234, 235]. Plant AMPs are divided into thionins, defensins, α-hairpinins, nonspecific lipid transfer proteins (nsLTPs), hevein- and knottin-like peptides based on their 3D structure and cysteine signature [236]. A study of cysteine-rich peptides (CRPs) revealed that they all had a structural component in common called the γ-core that is responsible for their antibacterial activity. This structural element has an antiparallel β-hairpin conformation [237]. Thionins are poisonous to yeast, fungus, and bacteria. Antifungal activity was defined as the development of pores or a specific contact with a particular lipid domain as a result of direct protein-membrane interactions between positively charged thionin and the negatively charged phospholipids in fungal membranes [238, 239]. The antibacterial, antifungal, proteinase, and insect amylase inhibitory properties of plant defensins also classify them as γ-thionins [240, 241]. Although the exact mechanism of action of defensins is yet unknown, and not all plant defensins work in the same way, they almost certainly use glucosylceramides as receptors for fungal cell membrane entry. Ion outflow and membrane rupture are caused by defensins’ positive charges repelling one another into the cell membrane [242]. Similar behaviour has been seen with nsLTPs, which interfere with the biological membrane permeability and integrity of pathogens [243]. Gram-positive bacteria (Bacillus megaterium and Sarcina lutea) and a number of fungi were able to halt the formation of powerful nsLTPs that were isolated from onion seeds [244, 245]. Sunflower (Helianthus annuus) seed-derived nsLTP also demonstrated antifungal activity [246]. The α-hairpinins are a different small family of AMPs that includes peptides with a wide range of biological activities, including antifungal and antibacterial activity [247]. The rubber tree (Hevea brasiliensis) (which produces latex that is similar to hevein) gave its name to the family of AMPs that resembles hevein. Hevein-like peptides are extremely stable, cysteine-rich substances that withstand heat and protease activity. They are active against a variety of phytopathogens, including bacteria and fungus, due to their capacity to interact with pathogens’ cell wall chitin, which is missing in plants [248]. Tk-AMP-K210-23, a knottin-like peptide isolated from T. kiharae seeds, was also discovered to have antibacterial action. According to the results of flow cytometry, there were less Cr. neoformans cells, which suggest that the peptide caused cell lysis by interfering with the integrity of the membrane [249,250,251]. More research and focus are required on peptides that are particularly effective against specific infections in order to create new antimicrobial agents.

Polyacetylenes

A wide variety of biomasses, including plants (particularly species of the Apiaceae family and carrot), marine organisms, fungi, insects, and people, include polyacetylenes, which are naturally occurring molecules characterized by the presence of two or more carbon–carbon triple bonds. There are just a few papers on the antibacterial potential of polyacetylenes, which are renowned for their anti-inflammatory and anticancer properties [252, 253]. A large number of natural polyacetylenes have been isolated such as 8S-heptadeca-2(Z),9(Z)-diene-4,6-diyne-1,8-diol (Fig. 5) from Bupleurum salicifolium Soland (Umbelliferae). This polyacetylene showed antibiotic activity against gram-positive bacteria S. aureus and Bacillus subtilis with no activity against gram-negative bacteria (E. coli, Salmonella sp., Pseuknas aeuriginosa) and the yeast Candida albicans [109]. Falcarinol-type polyynes (Fig. 8) showed to have antifungal activity that protects carrots from fungi as Botrytis cinerea Pers, while panaxydol and panaxytriol (Fig. 8) from Panax ginseng found to have cytotoxic effect against numerous cancer cell lines [254,255,256]. Exocarpos latifolius Kuntze stems were used to extract exocarpic acid derivatives (Fig. 8); some derivatives exhibited antimycobacterial activity against M. tuberculosis, while others exhibited no activity [253, 257]. Amadaldehyde, a C63 polyacetylenic aldehyde that was isolated from mango ginger (the rhizomes of Curcuma amada Roxb.), was shown to have antibacterial and antioxidant activity against microorganisms in addition to cytotoxicity and platelet aggregation inhibitory effect [258]. Several polyynes such as epoxide-ketone are found in one of the most important oriental medicinal plants P. ginseng C.A. Meyer. These compounds exhibited antimicrobial activity against several bacteria such as Bacillus subtilis, S. aureus, Cryptococcus neoformans, and Aspergillus fumigatus which conclude that P. ginseng releases antimicrobial polyacetylenes into the surrounding soil from roots to defend the plant[259]. Moreover, triyne carboxylic acid compound (octadeca-9,11,13-triynoic acid) (Fig. 8) isolated from the roots of Polyalthia cerasoides (Roxb.), showed antimalarial and antimycobacterial activity against Plasmodium falciparum and M. tuberculosis, respectively. Furthermore, 13,14-dihydrooropheic acid (Fig. 8) from the extract of Mitrephora celebica Scheff, and debilisones A–F (Fig. 8) from the Thai herbal plan Polyalthia debilis (Pierre) exhibited antibacterial activity against Mycobacterium smegmatis and M. tuberculosis, respectively [260,261,262].

Fig. 8
figure 8

Chemical structure of polyacetylenes compounds including falcarinol, panaxydol, panaxytriol, amadaldehyde, octadeca-9,11,13-triynoic acid, 13,14- dihydrooropheic acid, debilisones (AF)

Medicinal plants with antimicrobial activities

St. John’s wort (SJW) (Hypericum perforatum)

There are 500 species in the Hypericum (Hypericaceae) family, and they are all found worldwide. The most prevalent species is Hypericum perforatum, also known as St. John's wort (SJW), which is one of the most well-known and widely used herbs in the world. The herb has been widely used for therapeutic purposes across the globe [263, 264] and has been included into traditional medicine. The aerial parts or flowering tops make up the crude form of SJW medicine, which is employed in multi-ingredient formulations or as a monopreparation (either as is or as an extract) [265]. The upper stem leaves and flowering tops of H. perforatum are used in dietary supplements to treat mild to severe depression [266]. The flowering tops are typically prepared and used for its hypnotic and tonic properties or to speed wound healing. Due to varying extraction techniques, individual plant extracts do not include the entire group of phytoconstituents but only some of them, which causes a number of issues that affect the usage of H. perforatum in pharmaceutical formulations. Additionally, some of the active ingredients may experience problems with stability with exposure to time or light[267]. Due to the plant's synthesis of bioactive secondary metabolites like naphthodianthrones, flavonoids, bioflavonoids, phloroglucinols, xanthones, proanthocyanidins, acid phenols, and essential oils [263, 268, 269], it has been the subject of phytochemical studies.

Antibacterial activity of SJW

Sherif et al. [270] investigated the antibacterial activity of the Hypericum perforatum plant. The plant extract and its fractions were evaluated by Liquid Chromatography Electrospray Ionization Tandem Mass Spectrometric (LC–ESI–MS/MS) and tested against MRSA, Enterococcus faecalis, E. coli, and K. pneumonia MDR isolates. The results demonstrated that different extract from H. perforatum had a promising antibacterial activity against tested pathogens, particularly MDR-K. pneumoniae, with inhibition zones ranging from 17.9 to 27.9 mm and strong antioxidant effects, as opposed to sub-fractions extract, which demonstrated lesser inhibition zones and higher MIC values. Bacterial cell membranes are affected by extract of H. perforatum, which causes cell shrinkage and deformation that results in cell lysing. Additionally, the extract promoted cell elongation and thickness, which is similar to the actions of penicillin and the antibiotic cefotaxime, according to published research [271,272,273,274]. These findings indicate that these herbal extracts can be used to treat resistant bacteria, but further research is required to clarify why the total extract showed the most potent antibacterial activity over the subfraction? Is it an additive effect of specific compounds that act together with different mechanism to inhibit resistance bacteria or something else?. Using H. perforatum flower extracts, Okmen et al. [275] investigated the antibacterial activity of the bacteria that cause mastitis, a complicated condition characterized by inflammation of the parenchyma of the mammary glands and bacterial alterations in milk. Along with coagulase-negative staphylococci (CNS), Staphylococcus, Streptococcus, and coliform bacteria are the pathogens that cause the disease [276]. Although this condition is frequently treated with antibiotics, new antibiotics are still needed to combat germs because of the emergence of antibiotic resistance. The study's findings indicated that H. perforatum flower methanol extracts exhibit bactericidal action against gram-positive and gram-negative bacteria (S. aureus, Proteus vulgaris, P. aeruginosa, and E. coli), with the highest inhibition zone against Coagulase-negative Staphylococci. The plant extract also demonstrated antimutagenic qualities. However, additional tests are needed to know the extract constitutes and the bioactive compounds responsible for the biological activities. Polyphenolic substances with pharmacological characteristics are called xanthones. Strong free radical scavengers, xanthones have been shown to have activity against a variety of bacteria, including vancomycin-resistant enterococci, methicillin- and multidrug-resistant S. aureus, and M. tuberculosis [277,278,279,280,281,282,283]. When H. perforatum was exposed to Colletotrichum gloeosporioides cell wall extracts, xanthone (Fig. 9) buildup was seen as a defence mechanism [284]. After elicitation with Agrobacteriumtumefaciens, Franklin et al. [285] demonstrated that the antioxidant and antibacterial properties in H. perforatum cells had dramatically increased. The up-regulation of xanthone metabolism, particularly paxanthone (Fig. 9), which increased 12-fold within 24 h, was the cause of the enhanced activity. After 12 h of co-cultivation, the viability of A. tumefaciens was reported to have dramatically decreased. As a result of the buildup of xanthones, H. perforatum cell antibacterial activity rose ten times. Numerous infectious illnesses are brought on by S. aureus. Due to its development of resistance against practically all standard of care (SOC) antibiotics, Methicillin-resistant S. aureus (MRSA) has, regrettably, become a significant problem. Therefore, it is urgently necessary to create new tactics to combat MRSA [286,287,288]. Wang et al. [289] investigated the effect of hypericin (HYP) from H. perforatum on the susceptibility of β-lactam antibiotics (cefazolin, oxacillin, and nafcillin) and their synergistic effect with oxacillin in a murine bacteremia model. Result showed that HYP significantly reduced the minimum inhibitory concentrations (MICs) of β-lactam antibiotics and SarA (RNA-binding protein) which is a key regulator that bind on target promoters to control S. aureus virulence factors. Moreover, HYP enhanced the efficacy of oxacillin in MRSA bacteremia model; therefore, these results might be due to the synergistic effect of HYP with oxacillin.

Fig. 9
figure 9

Chemical structure of bioactive compounds found in St. John’s wort (SJW), rosemary, ginger which include, xanthone, paxanthone, hypericin, carnosol carnosic acid, p-cymene-7-ol, suberoylanilide hydroxamic acid, rosmanol, epirosmanol, isorosmanol, rosmaridiphenol, pyrogallol, ellagic acid, benzoic acid, butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), α-curcumene, α-farnesene, β-bisabolene, β-sesquiphellandrene, zingiberene, gingerol, paradol, shogaol, gingerenone-A, zingerone, 6-dehydrogingerdione, 10-gingerol, 12-gingerol, trans-anethole, and m-phenylphenol

Rosemary (Rosmarinus officinalis, L.)

In addition to its use in food preservation to stop oxidation and microbial contamination, Rosmarinus officinalis, L., a member of the Lamiaceae family that originated in the Mediterranean, is renowned for its antioxidant, hepatoprotective, antiangiogenic, and potential treatment for Alzheimer's disease properties [290,291,292]. Numerous polyphenolic substances, including rosmarinic acid (Fig. 6), hesperidin (Fig. 2), carnosol, and carnosic acid, are found in rosemary (Fig. 9). According to various studies [293,294,295], rosemary essential oil contains 1,8-cineole, camphor, and -pinene p-cymene-7-ol in addition to borneol (Fig. 9). Fresh or dried leaves, flowers, fruits, roots, stems, seeds, and bark can all be used to make plant extracts, while dried samples were shown to contain higher quantities of flavonoids. Air, microwave, oven, and freeze drying are all acceptable drying techniques [296,297,298].

Antibacterial activity of rosemary

Rosmarinus officinalis was found to have antibacterial properties in many research. In their research on the antibacterial properties of several natural extracts, including rosemary, Fernández-López et al. [299] examined how long the shelf life of veal meatballs could be stored for. The results showed that all of the studied microorganisms were susceptible to rosemary extracts (oil extract, water miscible extract, oil, and water-miscible extract), with oil extract having the strongest inhibitory impact. The most vulnerable bacteria were Brochothrix spp., which might be a reference to the antibacterial action of nonpolar phenolic compounds against gram-positive bacteria. Govaris et al. [300], Gomez-Estaca et al. [301], and both Camo et al. [302] and Quattara et al. [303] reported that rosemary EOs prevented food-spoileding bacteria from growing. Rosmarinus officinalis and Ocimum basilicum essential oils have been shown to have antibacterial action against multidrug-resistant clinical isolates of E. coli by Sienkiewicz et al. [293]. Since both EOs were effective against every clinical strain of E. coli, it can be inferred that they can be used to treat and prevent the emergence of resistance strains. Probuseenivasan et al. [304] and Mihajilov-Kristev et al. [305] found similar results, confirming rosemary essential oil's potent antibacterial action against E. coli. Carnosol, carnosic acid, rosmarinic acid, rosmanol, epirosmanol, isorosmanol, and rosmaridiphenol (Fig. 9) interact with the cell membrane and alter the production of nutrients, genetic material, and fatty acids to produce rosemary’s inhibitory effects. Additionally, they affect electron transport, result in cellular component leakage, interact with proteins in the membrane, and cause a loss of membrane functionality [306, 307]. To increase the effectiveness of antibiotics against multi-drug-resistant bacteria like MRSA, Ekambaram SP et al. [308] looked into the antibacterial activity and synergistic effect of rosmarinic acid (dimer of caffeic acid) with conventional antibiotics. The agar well diffusion method was used to assess the antibacterial activity of rosmarinic acid against microorganisms. In comparison to using an antibiotic alone, the results demonstrated that rosmarinic acid had a synergistic impact with the medications ofloxacin, amoxicillin, and vancomycin against S. aureus. However, only the vancomycin and rosmarinic acid combination was effective against MRSA. The activity of rosmarinic acid on surface proteins known as microbial surface components recognizing adhesive matrix molecules (MSCRAMM’s) present in S. aureus and MRSA was suggested to be the mechanism of action. Pomegranate, rosemary, and antibiotic were combined in a study by Abu El-Wafa et al. [309]. P. aeruginosa isolates with significant biofilm producers and antibiotic resistance were examined for the synergistic effects of the combination. Pomegranate and rosemary plant extracts were the most successful at inhibiting biofilm by lowering swimming and twitching motility, which in turn decreased bacterial cells adhering to surfaces and quorum-sensing (QS) signals. The presence of polyphenol molecules such catechol, pyrogallol (Fig. 9), gallic, ellagic, rosmarinic acid, and benzoic acid may be referred to as these activities. There are, however, limited reports of plant extracts’ antibacterial and antibiofilm activity [114, 310,311,312]. As a result, combining plant extracts with antibiotics may be able to prevent and get rid of microbial biofilms. Pomegranate and rosemary plant extracts combined with piperacillin, ceftazidime, imipenem, gentamycin, or levofloxacin had synergistic effects against a P. aeruginosa isolate and dramatically reduced biofilm mass after 24 h compared to the use of the plant extracts separately or together. To investigate the effects of medicinal plants and commercial antibiotics against bacterial pathogens, additional in vitro and in vivo investigations are required. In vitro tests using butylated hydroxytoluene (BHT) and butylated hydroxyanisole (BHA), two antioxidant food additives, and rosemary methanol extract were also conducted by Romano et al. [313] to examine the antioxidant and antibacterial properties of the extract (Fig. 9). The outcomes demonstrated that rosemary extracts (rosmarinic acid, carnosic acid, and carnosol) increased the antioxidant activity of BHT and BHA as well as the antibacterial activity of BHA.

Ginger (Zingiber officinale)

Zingiber officinale, a member of the Zingiberaceae family, has been used as a spice and a herbal remedy since ancient times [314]. Due to the presence of numerous bioactive components like terpene (e.g. α-curcumene, α-farnesene, β-bisabolene, β-sesquiphellandrene, and zingiberene) (Fig. 9), and phenolic compounds (e.g. gingerols, paradols, shogaols, gingerenone-A (Fig. 9), quercetin (Fig. 5), zingerone, and 6-dehydrogingerdione (Fig. 9)) [315,316,317,318,319], the ginger root is the most significant component that is used to cure a variety of illnesses including nausea, emesis, headaches, and colds. The majority of biological actions, including those that are antioxidant, anticancer, anti-inflammatory, and antibacterial, are caused by phenolic compounds [319,320,321,322]. Additionally, a number of studies [323,324,325,326,327,328] demonstrated that ginger can prevent and treat a wide range of illnesses, including neurodegenerative diseases, diabetes mellitus, obesity, cardiovascular diseases, respiratory problems, and nausea and emesis brought on by chemotherapy.

Antibacterial activity of ginger

Ginger essential oil is derived from the roots and has potential uses in a variety of industries, including pharmaceuticals, cosmetics, and food. Shewanella putrefaciens is a Gram-negative spoilage bacterium that can be found in aquatic products. Zhang et al. [329] studied the antibacterial activity of ginger essential oil against this bacterium. Result showed that ginger EO (Zingiberene, zingerone and α-curcumene) displayed significant antibacterial activity with MIC and MBC values of 2.0 and 4.0 μL/mL, respectively, against S. putrefaciens by disturbing cell membrane integrity. Additionally, ginger essential oil (EO) has the power to alter biofilm metabolism and kill it, supporting its usage as a natural food preservative. By Wang et al. [330], who extracted ginger essential oil using supercritical CO2 and steam distillation techniques, ginger was also demonstrated to be excellent for food preservation. The primary components were identified by GC–MS as zingiberene and α-curcumene, and their antibacterial efficacy was evaluated mostly against the pathogens that cause food deterioration, E. coli and S. aureus. Through bacterial cell membrane damage that resulting in protein and nucleic acid leaks, ginger EOs demonstrated remarkable bactericidal efficacy. This decreases bacterial metabolic activity, which ultimately leads to bacterial cell death. The expression of several genes encoding important enzymes, the tricarboxylic acid cycle, DNA metabolism, and proteins involved in cell membranes is all inhibited by ginger EOs. Similar mechanisms of action were also observed by Zhang et al. [331] who found that ginger extract damaged the Ralstonia solanacearum cell membrane’s permeability and integrity, leading to the leakage of several cell components, including nucleic acids, proteins, and others, as well as changes in the bacteria's shape. Additionally, Atai et al. [332] showed that ethanol ginger extract can be utilized to treat oral candidiasis and suppress the growth of Candida albicans. In the same context, Park et al. [333] assessed the ginger’s ability to suppress mouth microorganisms linked to periodontitis. Result revealed that ginger extracts including 10-gingerol and 12-gingerol (Fig. 9) exhibited antibacterial activities against anaerobic Gram-negative bacteria, Porphyromonas endodontalis, Porphyromonas gingivalis, and Prevotella intermedia which causes periodontal diseases. Chairgulprasert et al. [334] extracted ginger EOs by steam distillation of fresh rhizomes and identified its chemical constituents by GC–MS which found the presence of trans-anethole, m-phenylphenol, (Fig. 9) estragol, and camphor in the extract. Results showed that petroleum ether and dichloromethane extracts in addition to the EOS were able to inhibit different bacterial pathogens such as E. coli, S. aureus, Bacillus substilis, and Sarcina sp.; on the other hand, no activity was recorded against P. aeruginosa. Research investigations are focusing on the effectiveness of combining medicinal herbs with antibiotics for enhanced antibacterial action. Using antibiotics (Ceftazidime), Sagar PK et al. [335] assessed the antibacterial activity of crude methanol extracts of medicinal plants like eucalyptus, clove, and ginger against P. aeruginosa. Isolates. With ceftazidime, all plant extracts had a synergistic impact on P. aeruginosa. When ginger and eucalyptus extracts were mixed, their separate MICs were not reduced; however, when ginger and clove extracts were combined, a maximum twofold reduction in MIC was discovered. When ginger extract was used with aminoglycosides to treat vancomycin-resistant enterococci (VRE), MIC values were also lowered. By increasing membrane permeability and improving the influx of aminoglycosides into enterococcal cells, 10-gingerol has been shown by Nagoshi et al. [336] to be able to lower MICs of many aminoglycosides, including arbekacin, bacitracin, and polymyxin B. The effectiveness of 6-gingerol alone and in combination with amphotericin B against Leishmania in vivo murine models was evaluated by Alireza Keyhani et al. [337]. In addition to its capacity to create an apoptotic index, raise the expression of Th1-related cytokines, and decrease transcription factor levels, the combination demonstrated a strong antioxidant and extreme leishmanicidal activity. When combined with fluconazole, methanol ginger extract demonstrated antifungal activity against drug-resistant vulvovaginal candidiasis (VVC) in a mouse model, according to Khan et al. [338]. As compared to fluconazole or ginger extract used alone, which did not entirely cure VVC, in vitro results revealed better activity for the combination of fluconazole and ginger extract against C. albicans. Azole-resistant candidiasis may therefore be treated by administering this combination.

Cactus (Opuntia ficus-indica)

Opuntia ficus-indica (L.), sometimes known as the nopal cactus, is a tropical and subtropical plant found in South Africa, Mexico, Latin America, and Mediterranean nations [339]. All cactus portions contain high levels of polyphenols, which have anti-inflammatory and antioxidant qualities. The most significant source of polyphenols and flavonoids, including gallic acid, is found in flower parts. These compounds have cytotoxic and antioxidant properties that lessen DNA damage [340,341,342]. Cactus fruit, also known as cactus pear, also contains fibres, ascorbic acid, vitamin E, amino acids, and carotenoids, all of which have hypoglycemic and hypolipidemic effects [342,343,344]. Due to the abundance of fatty acids, it contains, including oleic acid, palmitic acid, linolenic acid, and linoleic acid (Fig. 10); cactus cladodes can have a hypocholesterolemic impact [345, 346]. Therefore, Opuntia ficus indica have been used in traditional medicine due to the abundance natural compounds and derivatives for treating burns, oedema, wounds obesity, and hyperlipidemia, in addition to its antiviral, anti-inflammatory, and hypoglycemic properties[347].

Fig. 10
figure 10

Chemical structure of bioactive compounds from nopal cactus including oleic acid, palmitic acid, linolenic acid, linoleic acid, octadecadienoic acid, dihydroauroglaucin, isotetrahydroauroglaucin, cristatumin B, quercetin5,4'-dimethyl ether, isorhamentin3-O-glucoside, isorhamnetin

Antibacterial activity of Cactus

The extensive use of cactus pears flowers in traditional medicine is a result of their abundance in natural bioactive substances. Protein, fibre, and minerals make up the chemical makeup of cactus hexane extracts, as demonstrated by Ennouri et al. [348]. In the plant extract, octadecadienoic acid and palmitic acid (Fig. 10) were the primary components and potassium was the leading mineral. Antibacterial activity against E. coli and S. aureus was highly effective which suggest using cactus as a food preservative. There are few studies available on the antibacterial activities of O. ficus-indica. The plant has proven to be effective in wound healing and skin conditions such as healing laser-induced skin burns. Khémiri et al. [349] demonstrated that cactus was able to inhibit cutaneous infections by showing antimicrobial effect against Enterobacter cloacae, antifungal activity against Penicillium, Aspergillus, and Fusarium (opportunistic cutaneous moulds), and antiyeast effect against Candida sake and Candida parapsilosis. Therefore, cactus-extracted oil is a good healing agent due to its antibacterial effect and the ability to reduce reepithelialization phase. Future investigations are needed to identify the active compounds in the oil extract and its mechanism of action involved in the healing process. Ammar et al. [350] also evaluated cactus flowers extracts (mucilaginous and methanol) antioxidant and antibacterial activities for enhancing wound healing in excision wound model in rats. A beneficial effect was noticed on cutaneous repair which assessed by acceleration in wound contraction and remodelling phases. Both extract showed antibacterial activities against tested gram-negative and gram-positive bacterial strain, E. coli, S. aureus, Bacillus subtilis, and Listeria monocytogenes, while no antibacterial activity was noticed against P.aeruginosa. The result supports the use of O. ficus-indica as therapeutic agent for dermal wound healing, but more research is needed to know the exact mechanism of action. Alqurashi et al. [351] explored biological activities of oil from Opuntia ficus-indica seed and showed that it possesses an inhibitory action against Saccharomyces cervisiae while no activity against Aspergillus niger. On the other hand, Elkady et al. [352] isolated endophytic Aspergillus niger fungus from cactus fruit peels and tested the effect of endophytic ethyl acetate extract and its isolated compounds (dihydroauroglaucin, isotetrahydroauroglaucin, and cristatumin B) (Fig. 10) on resistant bacterial strains. Result showed excellent activity against Gram-negative and gram-positive resistant bacteria. Another study done by Elkady et al. [353] tested methanol extract of cactus against pneumonial pathogens (Streptococcus pneumoniae, Klebsiella pneumoniae, P. aeruginosa, Legionella pneumophila, Moraxella catarrhalis, and Stenotrophomonas maltophilia). The isorhamentin3-O-glucoside and isorhamnetin (Fig. 10) compounds show moderate antibacterial activity against all tested microorganisms, while quercetin5,4'-dimethyl ether, which was isolated from the cactus ethyl acetate fraction, demonstrated the most antibacterial activity. According to these findings, O. ficus-indica and the components that were extracted from it can work as new all-natural antibacterial agents for the treatment of infectious disorders. Further studies in vivo are needed to confirm the antibacterial activity of the extracts. Sánchez et al. [353] studied antibacterial and antibiofilm activity of several methanolic plant extracts against nosocomial microorganisms. Methanolic plant extracts were tested against specific clinical bacterial isolates by using well diffusion method, and results showed that Prosopis laevigata extract was active against all the clinical isolates with highest inhibition diameter against S. aureus strain compared to Gutierrezia microcephala and O. ficus-indica that showed lesser inhibition diameter and no activity for Nothoscordum bivalve extract. E. coli was less susceptible, while K. pneumoniae and E. faecalis were more resistant to the extracts except for P. laevigata and O. ficus-indica. The major reduction on the specific biofilm formation index (SBF) in dose-dependent manner and on cytotoxic activity (using brine shrimp lethality test) was caused by O. ficus-indica. Unfortunately, there are no available studies and data on the synergistic effect of O. ficus-indica with antibiotics which highlights the need for researcher to focus on studying synergistic or additive effect of herbal plants, especially O. ficus-indica with antibiotic to discover new treatments.

Medicinal plants and wound healing

The epidermis, dermis, and hypodermis are the three layers of the skin, one of the biggest organs in the body. Additionally, it serves as the initial line of defence against aggressors like infections, chemicals, and physical contact. Due to the function of the epidermis, which blocks the entry or exit of water- or water-soluble substances, and the hypodermis, which blocks heat loss due to the poor thermal conductivity of fat, it also has the capacity to prevent water loss and preserve temperature [354,355,356,357]. In order to maintain skin hemostasis during inflammation, immune cells and nonimmune cells form a structure known as skin-associated lymphoid tissue (SALT). Additionally, the skin's microbiome, which includes bacteria, fungi, and viruses, is crucial for immune response, concluding that the skin serves as more than just a physical barrier [354, 358, 359]. Over the years, natural product ingredients such polyphenols, fatty acids, probiotics, polysaccharides, and others have demonstrated their efficacy as immune system modulators. The secret to controlling or curing skin inflammatory problems may lie in using natural products [360,361,362]. Skin can react to infectious agents through innate and adaptive immune processes, just as other tissues like mucosal surfaces. To isolate the damaged area, stop bleeding, and initiate the coagulation cascade, a clot must first form in order for the wound healing process to begin. Then comes the inflammatory phase, where immune cells begin to infiltrate and high levels of pro-inflammatory mediators are discovered to stop pathogen entry and more serious problems. The next stage is the proliferative phase, which is characterized by a significant growth of skin-resident cells including fibroblasts and high levels of angiogenesis. The remodelling phase, which may last for more than a year after the injury, is the longest and involves the skin regaining its natural structure. Any issue throughout these stages may hamper wound healing, which may then result in infections, excruciating pain, and occasionally neurological damage [363,364,365,366]. Several factors can cause impaired wound healing such as local factors that influence the characteristics of the wound like oxygenation and infections and systemic factors in which overall health or disease state affects the ability to heal like hormones and diabetes [367,368,369,370]. Impaired wound healing can result from a variety of factors, including local ones that alter the characteristics of the wound, such oxygenation and infections, as well as systemic ones, like hormones and diabetes, that have an impact on overall health or disease states and the capacity to heal. Numerous abnormalities, including fibrosis, scarring, and nonhealing wounds like persistent ulcers, can result from aberrant wound repair [371, 372]. New compounds with antioxidant, anti-inflammatory, and anticarcinogenic properties are being researched to prevent skin damage. Natural substances have been employed as antitumoural, analgesic, anti-inflammatory, and antioxidant agents [373, 374]. Table 2 lists the top plants for healing wounds.

Table 2 Most used medicinal plants in treating skin disorders
Fig. 11
figure 11

Chemical structure of active compounds found in medicinal plants used to treat skin disorders including, acemannan, L-Borneol, boswellic acids, brazilin, sappanchalcone, esculetin, quercetin-3-O-glucoside, celosin I, celosin II, asiaticoside, and madecassoside

Fig. 12
figure 12

Chemical structure of active compounds found in medicinal plants used to treat skin diseases cont. including cinnamaldehyde, furanoeudesma-1,3-diene, tannin, ginsenoside, emodin, polydatin, resveratrol, kushenol, sophoraflavanone B, kaurenoic acid, afzelin, hyperoside, and rutin

Biopolymers

Pathogenic microorganisms (bacteria, viruses, and unicellular and multicellular eukaryotes) can cause several series diseases which are a public health concern [440]. Different pathogens accumulate in acute and chronic wounds such as Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus pyogenes, Pseudomonas aeruginosa, and others which can affect wound healing process and increase life-threatening problems [441, 442]. Because they are naturally occurring biomolecules derived from bacteria, animals, and plants, biopolymers play a significant role in wound care. These bioactive features include antimicrobial, cell proliferative, angiogenic, and immune-modulatory polymers. These qualities, along with their biodegradability, renewability, and decreased antigenicity, rendered them more advantageous for the healing process than synthetic materials, which have problems with biocompatibility because of their harmful effects [443,444,445]. Natural biomaterials like cellulose, hyaluronic acid, collagen, alginate, and chitosan (Table 3) are frequently used in the wound care industry. With the advancement of technology, the characteristics of biopolymers can be improved to meet a variety of wound care needs, including tissue repair, scar-less healing, and integrity restoration of lost tissue [446, 447]. By preventing microbes from directly interacting with the bacterial cell wall, producing ROS to increase oxidative stress, and inducing the leaking of macromolecules like DNA and proteins from microorganisms, antimicrobial biomaterials can aid in the healing of wounds [442, 448].

Table 3 Natural biopolymers used in wound healing process

Extracellular vehicles (EVs)

Extracellular matrix (ECM), growth factors, and hormones are just a few of the biomaterials and composites that have recently undergone modifications to improve cell survival, motility, and proliferation. One of the most promising methods for wound healing is extracellular vehicles (EVs). EVs are released by a variety of cells and are crucial for the phases of wound healing (hemostasis, inflammation, proliferation, and remodelling) as well as intercellular communication that promotes regeneration. Additionally, by labelling EVs with certain surface proteins, they can be created to carry particular cargo and employed for targeted delivery [469,470,471,472,473]. Exosomes, which are created during endosomal sorting, microvesicles, which directly arise from plasma membranes, and apoptotic bodies, which are created following cell death, can all be classified as subpopulations of EVs [474,475,476]. The regulation of hemostasis and each stage of wound healing are considerably aided by EV. The most prevalent EVs in blood circulation are platelet-derived EVs (PEVs) [476,477,478]. The activated form of integrin IIb-3 mediates the role of PEVs from thrombin-stimulated platelets in the development of fibrin clots [479]. This form of integrin αIIbβ3 has a high affinity for fibrinogen. PEVs were also discovered to bind tissue factor (TF) and factor XII, as well as to stimulate the generation of thrombin, but only in the presence of factor VII and xii, indicating that they mediate both intrinsic and extrinsic coagulation pathways [480]. Neutrophil-derived EVs (NDEVs) have been shown to have pro- or anti-inflammatory effects by boosting the production of ROS and IL-8 [481]. EVs are also involved in inflammation. Additionally, EVs take part in the remodelling phase (starting fibroplast differentiation) and the proliferation phase (EVs from wound edge keratinocytes, or KCs-EVs) [482]. EVs can be made from plants, stem cells, or engineering [482, 483]. Membranous vesicles formed from plants are similar to mammalian exosomes but have different chemical compositions and include fewer proteins and no cholesterol in the lipid layer. EVs made from plants are less harmful, safer, and expensive. Wheat, broccoli, ginger, grapefruit, grape, lemon, and EVs were also discovered in the xylem and phloem of woody plants, according to recent investigations [484, 485]. Several plant have been known in pharmacognosy and phytochemistry to own antibleeding and hemostatic properties such as Rubia cordifolia,Alchornea cordifeliaAspilia africanaBaphia nitidaAgeratum conyzoides, Chromolaena odorata, Jathropha curcasLandolphia owariensis, Dalbergia sissooAloe spesiosaBeta vulgaris, Humulus lupulusSalix alba, and others [486,487,488]. Different studies demonstrated the effect of plant-derived EVs on wound healing; Perut et al. [489] isolated and purified plant-derived exosome-like nanovesicles (EPDENs) from strawberry juice of Fragaria x ananassa which is characterized of high anthocyanins, folic acid, flavonols, vitamin C, and short RNAs and miRNAs contents. The uptake of Fragaria-derived EPDENs by mesenchymal stromal cells (MSC) did not affect cell viability and prevented oxidative stress which may be due to the presence of vitamin C. Additionally, exosome-like nanovesicles isolated from Citrus limon L. (EXO-CLs) were investigated and examined in vitro on MSC by Baldini et al. [490]. The findings indicated that EXO-CLs contained short RNA sequences (20–30 bp), vitamin C, and citrate. EXO-CLs were taken up by MSC and had a significant antioxidant activity, according to in vitro tests. Ju et al. team also found that grape exosome-like nanoparticles (GELNs) have a protective effect against dextran sulphate sodium-induced colitis and facilitate intestinal tissue remodelling [491]. Similar outcomes were also observed with ginger-derived EVs, which promoted intestinal wound healing and are presently being investigated in clinical trials for inflammatory bowel disease and colon cancer [320, 482, 484]. Other plant-derived EVs, such as those from grapefruit and wheatgrass, improved wound healing by boosting cell viability and motility [485, 492]. The formation of hypertrophic scars and keloid lesions has also been demonstrated to be decreased by herbal extracts and active herbal compounds, including onion extract, epigallocatechin gallate from green tea, resveratrol obtained from peanuts, and others [493]. To retain EVs at the wound site and encourage longer, more effective results, they can be applied topically or encapsulated into scaffolds like hydrogels. Natural polymers like chitosan, alginate, and collagen are examples of hydrogels. Synthetic polymers like polyethene glycol, polyglycolic acid, and polyurethane are also possible [494,495,496,497]. A promising method to introduce EVs into the wound site with extended release involves encapsulating them in hydrogels [498]. To create naturally produced EVs for wound healing, further research on particular plant species is required.

Treatment of skin disorders

Diabetic foot ulcers (DFUs)

Patients with uncontrolled diabetes mellitus frequently develop diabetic foot ulcers (DFUs), which can be brought on by poor glycemic management, peripheral vascular disease, neuropathy, or inadequate foot care [499]. According to the International Diabetes Foundation [500], there are 40–60 million persons worldwide with DFUs. DFUs can be treated using several methods, including as gene therapy, stem cells, skin substitutes, and antibiotics. Due to the high cost of local debridement (removal of nonviable wound tissue) and negative pressure therapy, antibiotic resistance as a result of prolonged use, the ineffectiveness of growth factors to inhibit bacterial growth, and the existence of stem cell and gene therapies in the experimental stage, nonsurgical treatments for DFUs must be quick and inexpensive [501]. Due to their anti-inflammatory qualities, several herbal extracts have been utilized as traditional treatments to treat wounds. Liu et al. [501] designed an experiment to explore the effects of five herbal extracts on wound healing, Bauhinia purpurea (inhibit inflammation, and act as analgesic and antipyretic) [502], Paeoniae rubrae (ameliorating inflammation by inhibiting glycogen synthase kinase 3β (GSK3β)) [503], Angelica dahurica (accelerate wound healing by regulating inflammation) [504], Acorus calamus L (promote collagen maturation) [505], and Radix Angelicae biseratae (inhibit inflammation and regulate immune system in osteoarthritis) [506]. In order to determine the mechanisms of action for wound healing, the mixture of herbal plants was identified by Ultra-High-Performance Liquid Chromatography (UHPLC) and Quadrupole Exactive-Mass Spectrometer (QE-MS) and tested in vivo on a rat model with diabetic ulcer wound utilizing transcriptomics and proteomics. The mixture speeds up the healing of wounds by encouraging angiogenesis and the growth of M2 macrophages, according to the results. Specific miRNAs and proteins were found to be crucial for controlling wound healing by transcriptomics and proteomics. Consequently, the herbal combination may offer a potential method to quicken the healing of diabetic wounds [501].

Atopic dermatitis (AD)

Atopic dermatitis (AD), a chronic and relapsing inflammatory skin condition that affects children and is characterized by itchy, eczematous skin lesions, is another skin condition [507, 508]. Intense itching results in skin damage that compromises tissue repair and allows microorganisms to infiltrate the skin [509, 510]. The absence of precise disease processes is a challenge for the development of successful AD therapeutics. Flavonoids, a type of secondary metabolite found in plants, exhibit a variety of antiallergic properties, including antioxidant, anti-inflammatory, antiangiogenic, antibacterial, and antiviral effects [511, 512]. In foods ingested as part of a daily diet, quercetin (Fig. 5) is an illustration of a flavonoid [513]. Quercetin inhibits the release of histamine, proinflammatory cytokines, and interleukin (IL)-4 and -13, among other antiallergic characteristics. Despite this, there have only been a few research on quercetin's effects on AD [514, 515]. Therefore, Beken et al. [516] studied the effect of quercetin on AD model of human keratinocyte and treated it with IL- 4, -13, and tumour necrosis factor-α (TNF-α) to mimic AD in vitro. Result showed that quercetin accelerated wound healing by reducing AD-inducing agents IL-1β, IL-6, IL-8, thymic stromal lymphopoietin, phosphorylation of extracellular signal-regulated kinase 1/2/mitogen-activated protein kinase (ERK1/2 MAPK), and nuclear factor-kappa B (NF-κB), while it upregulated the expression of IL-10, and antioxidant enzymes; glutathione peroxidase (GPx), superoxide dismutase-1 (SOD1), SOD2, and catalase (CAT), in addition to mRNA expression of Twist and Snail. Therefore, quercetin may act as a potential therapy for AD symptoms.

Conclusions and future recommendations

Extensive and inappropriate uses of antibiotics resulted in the development of antimicrobial resistance and the rise of bacterial strains that were resistant to multiple drugs (MDR) and multiple drugs extensively (XDR), which made the most powerful medications useless. Antimicrobial resistance is very concerning and urgent issue, and scientists are aware that the shelf life of antibiotics is finite. Natural product research is receiving a lot of interest internationally. Today, millions of people around the world turn to phytomedicine as one of their top options for the treatment of chronic illnesses. Medicinal plant extracts are crucial in the fight against infectious diseases that pose a threat to public health globally. Alkaloids, phenolics, polyphenols, terpenoids, essential oils, lectins, polypeptides, and polyacetylenes are a few examples of antimicrobial phytochemicals that can be utilized as adjuvants or substitutes against bacterial infections. Medicinal plants including St. John’s wort (Hypericum perforatum), Rosemary (Rosmarinus officinalis), Ginger (Zingiber officinale), and nopal cactus (Opuntia ficus-indica (L.)) are attracting the interest of researchers due to their high phytochemical contents. Additionally, probiotics, polysaccharides, polyphenols, fatty acids, and other bioactive substances can modulate the immune system and treat inflammatory skin diseases such atopic dermatitis (AD) and diabetic foot ulcers (DFUs). Natural anticarcinogenic, anti-inflammatory, and antioxidant substances can be employed to stop skin deterioration. Biopolymers derived from microorganisms, animals, and plants (cellulose, hyaluronic acid, collagen, alginate, and chitosan) and extracellular matrix (ECM) have bioactive properties that make them promising approaches for wound healing. These properties include antimicrobial, immune-modulatory, cell proliferative, and angiogenic effects.

It is essential to create innovative plans and tactics to deal with the issue of rising AMR. Medicinal plants contain unlimited source of bioactive compounds which has been used in the treatment of many diseases, especially against bacterial infections. Despite that fact, natural compounds have not yet been thoroughly investigated and many are still unexplored. Therefore, researchers should make efforts to isolate and identify new bioactive compounds from plant source to face antibacterial resistance and find new effective treatments. Researchers should focus on using appropriate extraction methods to isolate compounds from bioactive extracts, study the mechanism of action, test the compounds in vivo in animal models, and apply structural modification to improve pharmacodynamics and pharmacokinetics. Moreover, further studies should be done on the synergistic or additive interactions between plant compounds itself and with antibiotics to enhance the action of medications.

Availability of data and materials

Not applicable.

Abbreviations

AMPs:

Antimicrobial peptides

AD:

Atopic dermatitis

BHT:

Butylated hydroxytoluene

BHA:

Butylated hydroxyanisole

CAT:

Catalase

COPD:

Chronic obstructive pulmonary disease

CNS:

Coagulase-negative staphylococci

CHF:

Congestive heart failure

CVNHs:

Cyanovirin-N homologs

COX-2:

Cyclooxygenase-2

CXCL:

Chemokine

CRPs:

Cysteine-rich peptides

DFUs:

Diabetic foot ulcers

DCM:

Dichloromethane

EAE:

Enzyme-assisted extraction

SFE:

Supercritical fluid extraction

EOs:

Essential oils

EDA:

Eucheuma denitculatum

ESA:

Eucheuma serra

EXO-CLs:

Exosome-like nanovesicles isolated from Citrus limon L.

ECM:

Extracellular matrix

ERK1/2 MAPK:

Extracellular signal-regulated kinase 1/2/mitogen-activated protein kinase

EVs:

Extracellular vehicles

GNA-like:

Galanthus nivalis agglutinin-like

GMA:

Galaxaura marginate

GC:

Gas chromatography

GPx:

Glutathione peroxidase

GSK3β:

Glycogen synthase kinase 3β

GELNs:

Grape exosome-like nanoparticles

SD-GCMS:

Headspace-gas chromatography/mass spectrometry

HTS:

High-throughput screening

HPLC:

High-performance liquid chromatography

HPTLC:

High-performance thin-layer chromatography

HA:

Hyaluronic acid

HPF:

Hyperforin

Hyp:

Hypericin

IR:

Infrared spectroscopy

IFN-γ:

Interferon

IL:

Interleukin

IAs:

Isoquinoline alkaloids

JRL:

Jacalin-related lectin

LC–ESI–MS/MS:

Liquid Chromatography Electrospray Ionization Tandem Mass Spectrometric

LPS:

Lipopolysaccharide

MS:

Mass spectrometry

MSC:

Mesenchymal stromal cells

MeOH:

Methanol

MRSA:

Methicillin-resistant S. aureus

MSCRAMM’s:

Microbial surface components recognizing adhesive matrix molecules

MICs:

Minimum inhibitory concentrations

MDR/XDR:

Multi- and extensively drug-resistant

NDEVs:

Neutrophil-derived EVs

NLRP3:

NOD-like receptor 3

NF-κB:

Nuclear factor-kappa B

NMR:

Nuclear magnetic resonance spectroscopy

OGEO:

Ocimum gratissimum L. essential oil

OAAH:

Oscillatoria agardhii agglutinin homolog

OA:

Osteoarthritis

PC:

Paper chromatography

PEVs:

Platelet-derived EVs

TPA:

Proanthocyanidin

PGE2:

Prostaglandin E2

QE-MS:

Quadrupole exactive-mass spectrometer

QS:

Quorum sensing

ROS:

Reactive oxygen species

STAT:

Signal transducer and activator of transcription

SALT:

Skin-associated lymphoid tissue

SPME-GC/MS:

Solid-phase microextraction-gas chromatography/mass spectrometry

SJW:

St. John’s wort

SOC:

Standard of care

SAHA:

Suberoylanilide hydroxamic acid

S-CO2:

Supercritical carbon dioxide

SF:

Supercritical fluid

SOD1:

Superoxide dismutase-1

THC:

Tetrahydrocannabinol

TLC:

Thin-layer chromatography

TF:

Tissue factor

TLR4:

Toll-like receptor 4

TFC:

Total flavonoid contents

TPC:

Total phenolic

TCM:

Traditional Chinese medicine

TB:

Tuberculosis

TNF-α:

Tumour necrosis factor-α

UHPLC:

Ultra-high-performance liquid chromatography

UAE:

Ultrasound-assisted extraction

UV:

Ultraviolet spectroscopy

VRE:

Vancomycin-resistant enterococci

VBNC:

Viable but nonculturable cells

VVC:

Vulvovaginal candidiasis

WHO:

World Health Organization

ARs:

α2-Adrenergic receptors

References

  1. Wachtel-Galor S, Benzie IFF (2011) Herbal medicine: an introduction to its history, usage, regulation, current trends, and research needs. In: Benzie IFF, Wachtel-Galor S (eds) Herbal Medicine: biomolecular and clinical aspects (2nd ed.). Boca Raton (FL)

  2. Obodozie OO (2012) Pharmacokinetics and drug interactions of herbal medicines: a missing critical step in the phytomedicine/drug development process. Read Adv Pharmacokinet: Theory Methods Appl. https://doi.org/10.5772/33699

    Article  Google Scholar 

  3. Xutian S, Zhang J, Louise W (2009) New exploration and understanding of traditional Chinese medicine. Am J Chin Med 37(3):411–426. https://doi.org/10.1142/S0192415X09006941

    Article  PubMed  Google Scholar 

  4. Pan SY, Zhou SF, Gao SH, Yu ZL, Zhang SF, Tang MK et al (2013) New perspectives on how to discover drugs from herbal medicines: CAMs outstanding contribution to modern therapeutics. Evid Based Complement Alternat Med 2013:627375. https://doi.org/10.1155/2013/627375

    Article  PubMed  PubMed Central  Google Scholar 

  5. Khan H (2015) Brilliant future of phytomedicines in the light of latest technological developments. J Phytopharmacol 4(1):58–60

    Article  Google Scholar 

  6. Ernst E, Schmidt K, Wider B (2005) CAM research in Britain: the last 10 years. Complement Ther Clin Pract 11(1):17–20. https://doi.org/10.1016/j.ctnm.2004.09.005

    Article  CAS  PubMed  Google Scholar 

  7. Ribnicky DM, Poulev A, Schmidt B, Cefalu WT, Raskin I (2008) Evaluation of botanicals for improving human health. Am J Clin Nutr 87(2):472S-475S. https://doi.org/10.1093/ajcn/87.2.472S

    Article  CAS  PubMed  Google Scholar 

  8. Kantor M (2009) The role of rigorous scientific evaluation in the use and practice of complementary and alternative medicine. J Am Coll Radiol 6(4):254–262. https://doi.org/10.1016/j.jacr.2008.09.012

    Article  PubMed  Google Scholar 

  9. Wink M (2015) Modes of action of herbal medicines and plant secondary metabolites. Medicines (Basel) 2(3):251–286. https://doi.org/10.3390/medicines2030251

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Srivastava A, Srivastava P, Pandey A, Khanna VK, Pant AB (2019). Phytomedicine, pp 625–655.https://doi.org/10.1016/b978-0-12-814619-4.00025-2

  11. Li JW, Vederas JC (2009) Drug discovery and natural products: end of an era or an endless frontier? Science 325(5937):161–165. https://doi.org/10.1126/science.1168243

    Article  CAS  PubMed  Google Scholar 

  12. Rousseaux CG, Schachter H (2003) Regulatory issues concerning the safety, efficacy and quality of herbal remedies. Birth Defects Res B Dev Reprod Toxicol 68(6):505–510. https://doi.org/10.1002/bdrb.10053

    Article  CAS  PubMed  Google Scholar 

  13. Hartmann T (2007) From waste products to ecochemicals: fifty years research of plant secondary metabolism. Phytochemistry 68(22–24):2831–2846. https://doi.org/10.1016/j.phytochem.2007.09.017

    Article  CAS  PubMed  Google Scholar 

  14. Rishton GM (2008) Natural products as a robust source of new drugs and drug leads: past successes and present day issues. Am J Cardiol 101(10A):43D-49D. https://doi.org/10.1016/j.amjcard.2008.02.007

    Article  CAS  PubMed  Google Scholar 

  15. Schmidt B, Ribnicky DM, Poulev A, Logendra S, Cefalu WT, Raskin I (2008) A natural history of botanical therapeutics. Metabolism 57(7 Suppl 1):S3-9. https://doi.org/10.1016/j.metabol.2008.03.001

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Raafat M, Kamel AA, Shehata AH, Ahmed AF, Bayoumi AMA, Moussa RA et al (2022) Aescin protects against experimental benign prostatic hyperplasia and preserves prostate histomorphology in rats via suppression of inflammatory cytokines and COX-2. Pharmaceuticals (Basel). https://doi.org/10.3390/ph15020130

    Article  PubMed  Google Scholar 

  17. Jeong NH, Yang EJ, Jin M, Lee JY, Choi YA, Park PH et al (2018) Esculetin from Fraxinus rhynchophylla attenuates atopic skin inflammation by inhibiting the expression of inflammatory cytokines. Int Immunopharmacol 59:209–216. https://doi.org/10.1016/j.intimp.2018.04.005

    Article  CAS  PubMed  Google Scholar 

  18. Wu J, Mu R, Sun M, Zhao N, Pan M, Li H et al (2019) Derivatives of natural product agrimophol as disruptors of intrabacterial pH homeostasis in Mycobacterium tuberculosis. ACS Infect Dis 5(7):1087–1104. https://doi.org/10.1021/acsinfecdis.8b00325

    Article  CAS  PubMed  Google Scholar 

  19. Zhao N, Sun M, Burns-Huang K, Jiang X, Ling Y, Darby C et al (2015) Identification of Rv3852 as an agrimophol-binding protein in Mycobacterium tuberculosis. PLoS ONE 10(5):e0126211. https://doi.org/10.1371/journal.pone.0126211

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Agrawal S, Yallatikar T, Gurjar P (2019) Brassica Nigra: ethopharmacological review of a routinely used condiment. Curr Drug Discov Technol 16(1):40–47. https://doi.org/10.2174/1570163815666180308143400

    Article  CAS  PubMed  Google Scholar 

  21. Sharma HK, Ingle S, Singh C, Sarkar BC, Upadhyay A (2012) Effect of various process treatment conditions on the allyl isothiocyanate extraction rate from mustard meal. J Food Sci Technol 49(3):368–372. https://doi.org/10.1007/s13197-011-0282-7

    Article  CAS  PubMed  Google Scholar 

  22. Poupko JM, Baskin SI, Moore E (2007) The pharmacological properties of anisodamine. J Appl Toxicol 27(2):116–121. https://doi.org/10.1002/jat.1154

    Article  CAS  PubMed  Google Scholar 

  23. Wang H, Lu Y, Chen HZ (2003) Differentiating effects of anisodamine on cognitive amelioration and peripheral muscarinic side effects induced by pilocarpine in mice. Neurosci Lett 344(3):173–176. https://doi.org/10.1016/s0304-3940(03)00444-0

    Article  CAS  PubMed  Google Scholar 

  24. Weathers PJ, Arsenault PR, Covello PS, McMickle A, Teoh KH, Reed DW (2011) Artemisinin production in Artemisia annua: studies in planta and results of a novel delivery method for treating malaria and other neglected diseases. Phytochem Rev 10(2):173–183. https://doi.org/10.1007/s11101-010-9166-0

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Liu N, Mathews A, Swanson J, Mhaskar R, Mathews A, Ayoubi N, Mirza AS (2020) Aspirin use for cardiovascular disease prevention in the uninsured population. SAGE Open Med 8:2050312120938224. https://doi.org/10.1177/2050312120938224

    Article  PubMed  PubMed Central  Google Scholar 

  26. Arif H, Aggarwal S (2023) Salicylic acid (Aspirin). In: StatPearls. Treasure Island (FL)

  27. McLendon K, Preuss CV (2023) Atropine. In: StatPearls. Treasure Island (FL)

  28. Neag MA, Mocan A, Echeverria J, Pop RM, Bocsan CI, Crisan G, Buzoianu AD (2018) Berberine: botanical occurrence, traditional uses, extraction methods, and relevance in cardiovascular, metabolic, hepatic, and renal disorders. Front Pharmacol 9:557. https://doi.org/10.3389/fphar.2018.00557

    Article  PubMed  PubMed Central  Google Scholar 

  29. Varilla C, Marcone M, Paiva L, Baptista J (2021) Bromelain, a group of pineapple proteolytic complex enzymes (Ananas comosus) and their possible therapeutic and clinical effects. A Summary. Foods. https://doi.org/10.3390/foods10102249

  30. Lee SH, Kim DS, Park SH, Park H (2022) Phytochemistry and applications of cinnamomum camphora essential oils. Molecules. https://doi.org/10.3390/molecules27092695

    Article  PubMed  PubMed Central  Google Scholar 

  31. Martino E, Della Volpe S, Terribile E, Benetti E, Sakaj M, Centamore A et al (2017) The long story of camptothecin: from traditional medicine to drugs. Bioorg Med Chem Lett 27(4):701–707. https://doi.org/10.1016/j.bmcl.2016.12.085

    Article  CAS  PubMed  Google Scholar 

  32. Bae J, Kim N, Shin Y, Kim S-Y, Kim Y-J (2020) Activity of catechins and their applications. Biomed Dermatol. https://doi.org/10.1186/s41702-020-0057-8

    Article  Google Scholar 

  33. Biondich AS, Joslin JD (2016) Coca: the history and medical significance of an ancient andean tradition. Emerg Med Int 2016:4048764. https://doi.org/10.1155/2016/4048764

    Article  PubMed  PubMed Central  Google Scholar 

  34. Richards JR, Laurin EG (2023) Cocaine. In StatPearls. Treasure Island (FL)

  35. Bhandari M, Bhandari A, Bhandari A (2011) Recent updates on codeine. Pharm. Methods 2(1):3–8. https://doi.org/10.4103/2229-4708.81082

    Article  Google Scholar 

  36. Dasgeb B, Kornreich D, McGuinn K, Okon L, Brownell I, Sackett DL (2018) Colchicine: an ancient drug with novel applications. Br J Dermatol 178(2):350–356. https://doi.org/10.1111/bjd.15896

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Chao PK, Chang HF, Ou LC, Chuang JY, Lee PT, Chang WT et al (2019) Convallatoxin enhance the ligand-induced mu-opioid receptor endocytosis and attenuate morphine antinociceptive tolerance in mice. Sci Rep 9(1):2405. https://doi.org/10.1038/s41598-019-39555-x

    Article  PubMed  PubMed Central  Google Scholar 

  38. Morimoto M, Tatsumi K, Yuui K, Terazawa I, Kudo R, Kasuda S (2021) Convallatoxin, the primary cardiac glycoside in lily of the valley (Convallaria majalis), induces tissue factor expression in endothelial cells. Vet Med Sci 7(6):2440–2444. https://doi.org/10.1002/vms3.614

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Sharifi-Rad J, Rayess YE, Rizk AA, Sadaka C, Zgheib R, Zam W et al (2020) Turmeric and its major compound curcumin on health: bioactive effects and safety profiles for food, pharmaceutical, biotechnological and medicinal applications. Front Pharmacol 11:01021. https://doi.org/10.3389/fphar.2020.01021

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Liu M, Huang Q, Li L, Li X, Zhang Z, Dong JT (2021) The cardiac glycoside deslanoside exerts anticancer activity in prostate cancer cells by modulating multiple signaling pathways. Cancers (Basel). https://doi.org/10.3390/cancers13225809

    Article  PubMed  PubMed Central  Google Scholar 

  41. Ershad M, Meredith A, Shah N, Khalid MM (2023) Cardioactive steroid toxicity. In: StatPearls. Treasure Island (FL)

  42. Cummings ED, Swoboda HD (2023) Digoxin toxicity. In: StatPearls. Treasure Island (FL)

  43. Lee MR (2008) Ipecacuanha: the South American vomiting root. J R Coll Physic Edinb 38(4):355–360

    CAS  Google Scholar 

  44. Tang S, Ren J, Kong L, Yan G, Liu C, Han Y et al (2023) Ephedrae herba: a review of its phytochemistry, pharmacology, clinical application, and alkaloid toxicity. Molecules. https://doi.org/10.3390/molecules28020663

    Article  PubMed  PubMed Central  Google Scholar 

  45. Wooltorton E, Sibbald B (2002) Ephedra/ephedrine: cardiovascular and CNS effects. CMAJ 166(5):633

    PubMed  PubMed Central  Google Scholar 

  46. Shah Z, Gohar UF, Jamshed I, Mushtaq A, Mukhtar H, Zia-Ui-Haq M et al (2021) Podophyllotoxin: history, recent advances and future prospects. Biomolecules 11(4):7. https://doi.org/10.3390/biom11040603

    Article  CAS  Google Scholar 

  47. Sinkule JA (1984) Etoposide: a semisynthetic epipodophyllotoxin. Chemistry, pharmacology, pharmacokinetics, adverse effects and use as an antineoplastic agent. Pharmacotherapy 4(2):61–73. https://doi.org/10.1002/j.1875-9114.1984.tb03318.x

    Article  CAS  PubMed  Google Scholar 

  48. Bogdanova Y, Stoeva T, Yanev S, Pandova B, Molle E, Burrus M, Stanilova M (2009) Influence of plant origin on propagation capacity and alkaloid biosynthesis during long-term in vitro cultivation of Leucojum aestivum L. In Vitro Cell Dev Biol Plant 45(4):458–465. https://doi.org/10.1007/s11627-008-9178-2

    Article  CAS  PubMed  Google Scholar 

  49. Kim JK, Park SU (2017) Pharmacological aspects of galantamine for the treatment of Alzheimer’s disease. EXCLI J 16:35–39. https://doi.org/10.17179/excli2016-820

    Article  PubMed  PubMed Central  Google Scholar 

  50. Yeo D, Huynh N, Beutler JA, Baldwin GS, He H, Nikfarjam M (2016) Glaucarubinone combined with gemcitabine improves pancreatic cancer survival in an immunocompetent orthotopic murine model. J Invest Surg 29(6):366–372. https://doi.org/10.3109/08941939.2016.1160167

    Article  PubMed  PubMed Central  Google Scholar 

  51. Meyer GM, Meyer MR, Wissenbach DK, Maurer HH (2013) Studies on the metabolism and toxicological detection of glaucine, an isoquinoline alkaloid from Glaucium flavum (Papaveraceae), in rat urine using GC-MS, LC-MS(n) and LC-high-resolution MS(n). J Mass Spectrom 48(1):24–41. https://doi.org/10.1002/jms.3112

    Article  CAS  PubMed  Google Scholar 

  52. Pastorino G, Cornara L, Soares S, Rodrigues F, Oliveira M (2018) Liquorice (Glycyrrhiza glabra): a phytochemical and pharmacological review. Phytother Res 32(12):2323–2339. https://doi.org/10.1002/ptr.6178

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Pal D, Sahu P, Sethi G, Wallace CE, Bishayee A (2022) Gossypol and its natural derivatives: multitargeted phytochemicals as potential drug candidates for oncologic diseases. Pharmaceutics. https://doi.org/10.3390/pharmaceutics14122624

    Article  PubMed  PubMed Central  Google Scholar 

  54. Pyrzynska K (2022) Hesperidin: a review on extraction methods, stability and biological activities. Nutrients. https://doi.org/10.3390/nu14122387

    Article  PubMed  PubMed Central  Google Scholar 

  55. Alizadeh A, Moshiri M, Alizadeh J, Balali-Mood M (2014) Black henbane and its toxicity: a descriptive review. Avicenna J Phytomed 4(5):297–311

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Kciuk M, Marciniak B, Kontek R (2020) Irinotecan-still an important player in cancer chemotherapy: a comprehensive overview. Int J Mol Sci. https://doi.org/10.3390/ijms21144919

    Article  PubMed  PubMed Central  Google Scholar 

  57. Bhattacharyya KB (2022) The Story of levodopa: a long and arduous journey. Ann Indian Acad Neurol 25(1):124–130. https://doi.org/10.4103/aian.aian_474_21

    Article  PubMed  PubMed Central  Google Scholar 

  58. Pathan H, Williams J (2012) Basic opioid pharmacology: an update. Br J Pain 6(1):11–16. https://doi.org/10.1177/2049463712438493

    Article  PubMed  PubMed Central  Google Scholar 

  59. Conti CR (2011) Intravenous morphine and chest pain. Clin Cardiol 34(8):464–465. https://doi.org/10.1002/clc.20927

    Article  PubMed  PubMed Central  Google Scholar 

  60. Du J, Jiang L, Chen F, Hu H, Zhou M (2021) Cardiac glycoside ouabain exerts anticancer activity via downregulation of STAT3. Front Oncol 11:684316. https://doi.org/10.3389/fonc.2021.684316

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Skubnik J, Svobodova Pavlickova V, Psotova J, Rimpelova S (2021) Cardiac glycosides as autophagy modulators. Cells. https://doi.org/10.3390/cells10123341

    Article  PubMed  PubMed Central  Google Scholar 

  62. Sharifi-Rad J, Quispe C, Patra JK, Singh YD, Panda MK, Das G et al (2021) Paclitaxel: application in modern oncology and nanomedicine-based cancer therapy. Oxid Med Cell Longev 2021:3687700. https://doi.org/10.1155/2021/3687700

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Kong YR, Jong YX, Balakrishnan M, Bok ZK, Weng JKK, Tay KC et al (2021) Beneficial role of Carica papaya extracts and phytochemicals on oxidative stress and related diseases: a mini review. Biology (Basel). https://doi.org/10.3390/biology10040287

    Article  PubMed  PubMed Central  Google Scholar 

  64. Kang YM, Kang HA, Cominguez DC, Kim SH, An HJ (2021) Papain ameliorates lipid accumulation and inflammation in high-fat diet-Induced obesity mice and 3T3-L1 adipocytes via AMPK activation. Int J Mol Sci. https://doi.org/10.3390/ijms22189885

    Article  PubMed  PubMed Central  Google Scholar 

  65. Ashrafi S, Alam S, Sultana A, Raj A, Emon NU, Richi FT et al (2023) Papaverine: a miraculous alkaloid from opium and its multimedicinal application. Molecules. https://doi.org/10.3390/molecules28073149

    Article  PubMed  PubMed Central  Google Scholar 

  66. Zhao B, Moochhala SM, Tham SY (2004) Biologically active components of Physostigma venenosum. J Chromatogr B Analyt Technol Biomed Life Sci 812(1–2):183–192. https://doi.org/10.1016/j.jchromb.2004.08.031

    Article  CAS  PubMed  Google Scholar 

  67. Andrade OA, Zafar Gondal A (2023). Physostigmine. In: StatPearls. Treasure Island (FL)

  68. Lima DF, de Lima LI, Rocha JA, de Andrade IM, Grazina LG, Villa C et al (2017) Seasonal change in main alkaloids of jaborandi (Pilocarpus microphyllus Stapf ex Wardleworth), an economically important species from the Brazilian flora. PLoS ONE 12(2):e017028. https://doi.org/10.1371/journal.pone.0170281

    Article  CAS  Google Scholar 

  69. Glowacka K, Wiela-Hojenska A (2021) Pseudoephedrine-benefits and risks. Int J Mol Sci. https://doi.org/10.3390/ijms22105146

    Article  PubMed  PubMed Central  Google Scholar 

  70. Achan J, Talisuna AO, Erhart A, Yeka A, Tibenderana JK, Baliraine FN et al (2011) Quinine, an old anti-malarial drug in a modern world: role in the treatment of malaria. Malar J 10:144. https://doi.org/10.1186/1475-2875-10-144

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Shamon SD, Perez MI (2016) Blood pressure-lowering efficacy of reserpine for primary hypertension. Cochrane Database Syst Rev 12(12):CD007655. https://doi.org/10.1002/14651858.CD007655.pub3

    Article  PubMed  Google Scholar 

  72. Spinks A, Wasiak J (2011) Scopolamine (hyoscine) for preventing and treating motion sickness. Cochrane Database Syst Rev 2011(6):CD002851. https://doi.org/10.1002/14651858.CD002851.pub4

    Article  PubMed  PubMed Central  Google Scholar 

  73. Rama Reddy NR, Mehta RH, Soni PH, Makasana J, Gajbhiye NA, Ponnuchamy M, Kumar J (2015) Next generation sequencing and transcriptome analysis predicts biosynthetic pathway of sennosides from Senna (Cassia angustifolia Vahl.), a non-model plant with potent laxative properties. PLoS ONE 10(6):e0129422. https://doi.org/10.1371/journal.pone.0129422

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Breijyeh Z, Jubeh B, Bufo SA, Karaman R, Scrano L (2021) Cannabis: a toxin-producing plant with potential therapeutic uses. Toxins (Basel). https://doi.org/10.3390/toxins13020117

    Article  PubMed  Google Scholar 

  75. Barnes PJ (2010) Theophylline. Pharmaceuticals (Basel) 3(3):725–747. https://doi.org/10.3390/ph3030725

    Article  CAS  PubMed  Google Scholar 

  76. Jean-Marie E, Bereau D, Robinson JC (2021) Benefits of polyphenols and methylxanthines from Cocoa beans on dietary metabolic disorders. Foods. https://doi.org/10.3390/foods10092049

  77. Kowalczyk A, Przychodna M, Sopata S, Bodalska A, Fecka I (2020) Thymol and thyme essential oil-new insights into selected therapeutic applications. Molecules. https://doi.org/10.3390/molecules25184125

    Article  PubMed  PubMed Central  Google Scholar 

  78. Bowman WC (2006) Neuromuscular block. Br J Pharmacol 147(1(Suppl 1)):S277-286. https://doi.org/10.1038/sj.bjp.0706404

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Patel S, Shukla J, Jain S, Paliwal V, Tripathi N, Paliwal S, Sharma S (2022) Repositioning of tubocurarine as analgesic and anti-inflammatory agent: Exploring beyond myorelaxant activity. Biochem Pharmacol 205:115248. https://doi.org/10.1016/j.bcp.2022.115248

    Article  CAS  PubMed  Google Scholar 

  80. Jabir NR, Firoz CK, Zughaibi TA, Alsaadi MA, Abuzenadah AM, Al-Asmari AI et al (2022) A literature perspective on the pharmacological applications of yohimbine. Ann Med 54(1):2861–2875. https://doi.org/10.1080/07853890.2022.2131330

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Antimicrobial Resistance C (2022) Global burden of bacterial antimicrobial resistance in 2019: a systematic analysis. Lancet 399(10325):629–655. https://doi.org/10.1016/S0140-6736(21)02724-0

    Article  Google Scholar 

  82. Sundin GW, Wang N (2018) Antibiotic resistance in plant-pathogenic bacteria. Annu Rev Phytopathol 56:161–180. https://doi.org/10.1146/annurev-phyto-080417-045946

    Article  CAS  PubMed  Google Scholar 

  83. Breijyeh Z, Jubeh B, Karaman R (2020) Resistance of gram-negative bacteria to current antibacterial agents and approaches to resolve it. Molecules. https://doi.org/10.3390/molecules25061340

    Article  PubMed  PubMed Central  Google Scholar 

  84. Verhaegen M, Bergot T, Liebana E, Stancanelli G, Streissl F, Mingeot-Leclercq MP et al (2023) On the use of antibiotics to control plant pathogenic bacteria: a genetic and genomic perspective. Front Microbiol 14:1221478. https://doi.org/10.3389/fmicb.2023.1221478

    Article  PubMed  PubMed Central  Google Scholar 

  85. Alekshun MN, Levy SB (2007) Molecular mechanisms of antibacterial multidrug resistance. Cell 128(6):1037–1050. https://doi.org/10.1016/j.cell.2007.03.004

    Article  CAS  PubMed  Google Scholar 

  86. van Hoek AH, Mevius D, Guerra B, Mullany P, Roberts AP, Aarts HJ (2011) Acquired antibiotic resistance genes: an overview. Front Microbiol 2:203. https://doi.org/10.3389/fmicb.2011.00203

    Article  PubMed  PubMed Central  Google Scholar 

  87. Blair JM, Webber MA, Baylay AJ, Ogbolu DO, Piddock LJ (2015) Molecular mechanisms of antibiotic resistance. Nat Rev Microbiol 13(1):42–51. https://doi.org/10.1038/nrmicro3380

    Article  CAS  PubMed  Google Scholar 

  88. Lin J, Nishino K, Roberts MC, Tolmasky M, Aminov RI, Zhang L (2015) Mechanisms of antibiotic resistance. Front Microbiol 6:34. https://doi.org/10.3389/fmicb.2015.00034

    Article  PubMed  PubMed Central  Google Scholar 

  89. Karaman R, Jubeh B, Breijyeh Z (2020) Resistance of gram-positive bacteria to current antibacterial agents and overcoming approaches. Molecules. https://doi.org/10.3390/molecules25122888

    Article  PubMed  PubMed Central  Google Scholar 

  90. Asokan GV, Ramadhan T, Ahmed E, Sanad H (2019) WHO global priority pathogens list: a bibliometric analysis of medline-pubmed for knowledge mobilization to infection prevention and control practices in bahrain. Oman Med J 34(3):184–193. https://doi.org/10.5001/omj.2019.37

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Breijyeh Z, Karaman R (2023) Design and synthesis of novel antimicrobial agents. Antibiotics (Basel). https://doi.org/10.3390/antibiotics12030628

    Article  PubMed  Google Scholar 

  92. Tampakaki AP, Hatziloukas E, Panopoulos NJ (2009) Encyclopedia of microbiology, Third edition. ed., Plant Pathogens, Bacterial., Academic Press, Oxford, San Diego

  93. Srivastava J, Chandra H, Nautiyal AR, Kalra SJ (2014) Antimicrobial resistance (AMR) and plant-derived antimicrobials (PDA(m)s) as an alternative drug line to control infections. 3 Biotech 4(5):451–460. https://doi.org/10.1007/s13205-013-0180-y

    Article  PubMed  Google Scholar 

  94. Chassagne F, Samarakoon T, Porras G, Lyles JT, Dettweiler M, Marquez L et al (2020) A systematic review of plants with antibacterial activities: a taxonomic and phylogenetic perspective. Front Pharmacol 11:586548. https://doi.org/10.3389/fphar.2020.586548

    Article  CAS  PubMed  Google Scholar 

  95. Singla P, Bhardwaj RD, Kaur S, Kaur J (2019) Antioxidant potential of barley genotypes inoculated with five different pathotypes of Puccinia striiformis f. sp. hordei. Physiol Mol Biol Plants 25(1):145–157. https://doi.org/10.1007/s12298-018-0614-4

    Article  CAS  PubMed  Google Scholar 

  96. Xie Y, Hou W, Song X, Yu Y, Huang J, Sun X et al (2016) Ferroptosis: process and function. Cell Death Differ 23(3):369–379. https://doi.org/10.1038/cdd.2015.158

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Mittler R (2017) ROS are good. Trends Plant Sci 22(1):11–19. https://doi.org/10.1016/j.tplants.2016.08.002

    Article  CAS  PubMed  Google Scholar 

  98. Debona D, Rodrigues FA, Rios JA, Nascimento KJ (2012) Biochemical changes in the leaves of wheat plants infected by Pyricularia oryzae. Phytopathology 102(12):1121–1129. https://doi.org/10.1094/PHYTO-06-12-0125-R

    Article  CAS  PubMed  Google Scholar 

  99. Akter S, Huang J, Waszczak C, Jacques S, Gevaert K, Van Breusegem F, Messens J (2015) Cysteines under ROS attack in plants: a proteomics view. J Exp Bot 66(10):2935–2944. https://doi.org/10.1093/jxb/erv044

    Article  CAS  PubMed  Google Scholar 

  100. Hussain SS, Ali M, Ahmad M, Siddique KH (2011) Polyamines: natural and engineered abiotic and biotic stress tolerance in plants. Biotechnol Adv 29(3):300–311. https://doi.org/10.1016/j.biotechadv.2011.01.003

    Article  CAS  PubMed  Google Scholar 

  101. Huang H, Ullah F, Zhou DX, Yi M, Zhao Y (2019) Mechanisms of ROS regulation of plant development and stress responses. Front Plant Sci 10:800. https://doi.org/10.3389/fpls.2019.00800

    Article  PubMed  PubMed Central  Google Scholar 

  102. Parham S, Kharazi AZ, Bakhsheshi-Rad HR, Nur H, Ismail AF, Sharif S et al (2020) Antioxidant, antimicrobial and antiviral properties of herbal materials. Antioxidants (Basel). https://doi.org/10.3390/antiox9121309

    Article  PubMed  Google Scholar 

  103. Yu Z, Li Q, Wang J, Yu Y, Wang Y, Zhou Q, Li P (2020) Reactive oxygen species-related nanoparticle toxicity in the biomedical field. Nanoscale Res Lett 15(1):115. https://doi.org/10.1186/s11671-020-03344-7

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Cowan MM (1999) Plant products as antimicrobial agents. Clin Microbiol Rev 12(4):564–582. https://doi.org/10.1128/CMR.12.4.564

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Vishwakarma R (1990) Stereoselective synthesis of α-arteether from artemisinin. J Nat Prod 53(1):216–217

    Article  CAS  Google Scholar 

  106. De Bolle MF, Osborn RW, Goderis IJ, Noe L, Acland D, Hart CA et al (1996) Antimicrobial peptides from Mirabilis jalapa and Amaranthus caudatus: expression, processing, localization and biological activity in transgenic tobacco. Plant Mol Biol 31(5):993–1008. https://doi.org/10.1007/BF00040718

    Article  PubMed  Google Scholar 

  107. Colilla FJ, Rocher A, Mendez E (1990) Gamma-Purothionins: amino acid sequence of two polypeptides of a new family of thionins from wheat endosperm. FEBS Lett 270(1–2):191–194. https://doi.org/10.1016/0014-5793(90)81265-p

    Article  CAS  PubMed  Google Scholar 

  108. Mendez E, Moreno A, Colilla F, Pelaez F, Limas GG, Mendez R et al (1990) Primary structure and inhibition of protein synthesis in eukaryotic cell-free system of a novel thionin, gamma-hordothionin, from barley endosperm. Eur J Biochem 194(2):533–539. https://doi.org/10.1111/j.1432-1033.1990.tb15649.x

    Article  CAS  PubMed  Google Scholar 

  109. Estevez-Braun A, Estevez-Reyes R, Moujir LM, Ravelo AG, Gonzalez AG (1994) Antibiotic activity and absolute configuation of 8S-heptadeca-2(Z),9(Z)-diene-4,6-diyne-1,8-diol from Bupleurum salicifolium. J Nat Prod 57(8):1178–1182. https://doi.org/10.1021/np50110a009

    Article  CAS  PubMed  Google Scholar 

  110. Vauzour D, Rodriguez-Mateos A, Corona G, Oruna-Concha MJ, Spencer JP (2010) Polyphenols and human health: prevention of disease and mechanisms of action. Nutrients 2(11):1106–1131. https://doi.org/10.3390/nu2111106

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Daglia M (2012) Polyphenols as antimicrobial agents. Curr Opin Biotechnol 23(2):174–181. https://doi.org/10.1016/j.copbio.2011.08.007

    Article  CAS  PubMed  Google Scholar 

  112. Cushnie TP, Lamb AJ (2005) Antimicrobial activity of flavonoids. Int J Antimicrob Agents 26(5):343–356. https://doi.org/10.1016/j.ijantimicag.2005.09.002

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Barbieri R, Coppo E, Marchese A, Daglia M, Sobarzo-Sanchez E, Nabavi SF, Nabavi SM (2017) Phytochemicals for human disease: an update on plant-derived compounds antibacterial activity. Microbiol Res 196:44–68. https://doi.org/10.1016/j.micres.2016.12.003

    Article  CAS  PubMed  Google Scholar 

  114. Cheesman MJ, Ilanko A, Blonk B, Cock IE (2017) Developing new antimicrobial therapies: are synergistic combinations of plant extracts/compounds with conventional antibiotics the solution? Pharmacogn Rev 11(22):57–72. https://doi.org/10.4103/phrev.phrev_21_17

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Sarkar A, De R, Mukhopadhyay AK (2016) Curcumin as a potential therapeutic candidate for Helicobacter pylori associated diseases. World J Gastroenterol 22(9):2736–2748. https://doi.org/10.3748/wjg.v22.i9.2736

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Guerra-Valle M, Orellana-Palma P, Petzold G (2022) Plant-based polyphenols: anti-Helicobacter pylori effect and improvement of gut microbiota. Antioxidants (Basel). https://doi.org/10.3390/antiox11010109

    Article  PubMed  Google Scholar 

  117. Fallone CA, Chiba N, van Zanten SV, Fischbach L, Gisbert JP, Hunt RH et al (2016) The toronto consensus for the treatment of Helicobacter pylori infection in adults. Gastroenterology 151(1):51-69e14. https://doi.org/10.1053/j.gastro.2016.04.006

    Article  PubMed  Google Scholar 

  118. Ranjbar R, Chehelgerdi M (2018) Genotyping and antibiotic resistance properties of Helicobacter pylori strains isolated from human and animal gastric biopsies. Infect Drug Resist 11:2545–2554. https://doi.org/10.2147/IDR.S187885

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Liu WZ, Xie Y, Lu H, Cheng H, Zeng ZR, Zhou LY et al (2018) Fifth Chinese national consensus report on the management of Helicobacter pylori infection. Helicobacter 23(2):e12475. https://doi.org/10.1111/hel.12475

    Article  PubMed  Google Scholar 

  120. Chey WD, Leontiadis GI, Howden CW, Moss SF (2018) Correction: ACG clinical guideline: treatment of Helicobacter pylori infection. Am J Gastroenterol 113(7):1102. https://doi.org/10.1038/s41395-018-0132-6

    Article  PubMed  Google Scholar 

  121. Li BZ, Threapleton DE, Wang JY, Xu JM, Yuan JQ, Zhang C et al (2015) Comparative effectiveness and tolerance of treatments for Helicobacter pylori: systematic review and network meta-analysis. BMJ 351:h4052. https://doi.org/10.1136/bmj.h4052

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Wang Q, Yao C, Li Y, Luo L, Xie F, Xiong Q, Feng P (2023) Effect of polyphenol compounds on Helicobacter pylori eradication: a systematic review with meta-analysis. BMJ Open 13(1):e062932. https://doi.org/10.1136/bmjopen-2022-062932

    Article  PubMed  PubMed Central  Google Scholar 

  123. O’Gara EA, Maslin DJ, Nevill AM, Hill DJ (2008) The effect of simulated gastric environments on the anti-Helicobacter activity of garlic oil. J Appl Microbiol 104(5):1324–1331. https://doi.org/10.1111/j.1365-2672.2007.03637.x

    Article  CAS  PubMed  Google Scholar 

  124. Canizares P, Gracia I, Gomez LA, Martin de Argila C, de Rafael L, Garcia A (2002) Optimization of Allium sativum solvent extraction for the inhibition of in vitro growth of Helicobacter pylori. Biotechnol Prog 18(6):1227–1232. https://doi.org/10.1021/bp025592z

    Article  CAS  PubMed  Google Scholar 

  125. Lawson LD, Wang ZJ, Papadimitriou D (2001) Allicin release under simulated gastrointestinal conditions from garlic powder tablets employed in clinical trials on serum cholesterol. Planta Med 67(1):13–18. https://doi.org/10.1055/s-2001-10624

    Article  CAS  PubMed  Google Scholar 

  126. Lee HA, Kim JY, Kim J, Nam B, Kim O (2020) Anti-Helicobacter pylori activity of acomplex mixture of Lactobacillus paracasei HP7 including the extract of Perilla frutescens var. acuta and Glycyrrhiza glabra. Lab Anim Res 36:40. https://doi.org/10.1186/s42826-020-00073-x

    Article  PubMed  PubMed Central  Google Scholar 

  127. Asha MK, Debraj D, Prashanth D, Edwin JR, Srikanth HS, Muruganantham N et al (2013) In vitro anti-Helicobacter pylori activity of a flavonoid rich extract of Glycyrrhiza glabra and its probable mechanisms of action. J Ethnopharmacol 145(2):581–586. https://doi.org/10.1016/j.jep.2012.11.033

    Article  CAS  PubMed  Google Scholar 

  128. Wittschier N, Faller G, Hensel A (2009) Aqueous extracts and polysaccharides from liquorice roots (Glycyrrhiza glabra L.) inhibit adhesion of Helicobacter pylori to human gastric mucosa. J Ethnopharmacol 125(2):218–223. https://doi.org/10.1016/j.jep.2009.07.009

    Article  CAS  PubMed  Google Scholar 

  129. Shmuely H, Yahav J, Samra Z, Chodick G, Koren R, Niv Y, Ofek I (2007) Effect of cranberry juice on eradication of Helicobacter pylori in patients treated with antibiotics and a proton pump inhibitor. Mol Nutr Food Res 51(6):746–751. https://doi.org/10.1002/mnfr.200600281

    Article  CAS  PubMed  Google Scholar 

  130. Zhang L, Ma J, Pan K, Go VL, Chen J, You WC (2005) Efficacy of cranberry juice on Helicobacter pylori infection: a double-blind, randomized placebo-controlled trial. Helicobacter 10(2):139–145. https://doi.org/10.1111/j.1523-5378.2005.00301.x

    Article  PubMed  Google Scholar 

  131. Li ZX, Ma JL, Guo Y, Liu WD, Li M, Zhang LF et al (2021) Suppression of Helicobacter pylori infection by daily cranberry intake: a double-blind, randomized, placebo-controlled trial. J Gastroenterol Hepatol 36(4):927–935. https://doi.org/10.1111/jgh.15212

    Article  CAS  PubMed  Google Scholar 

  132. Matsushima M, Suzuki T, Masui A, Kasai K, Kouchi T, Takagi A et al (2008) Growth inhibitory action of cranberry on Helicobacter pylori. J Gastroenterol Hepatol 23(Suppl 2):S175-180. https://doi.org/10.1111/j.1440-1746.2008.05409.x

    Article  PubMed  Google Scholar 

  133. Irving GR, Karmokar A, Berry DP, Brown K, Steward WP (2011) Curcumin: the potential for efficacy in gastrointestinal diseases. Best Pract Res Clin Gastroenterol 25(4–5):519–534. https://doi.org/10.1016/j.bpg.2011.09.005

    Article  CAS  PubMed  Google Scholar 

  134. Santos AM, Lopes T, Oleastro M, Gato IV, Floch P, Benejat L et al (2015) Curcumin inhibits gastric inflammation induced by Helicobacter pylori infection in a mouse model. Nutrients 7(1):306–320. https://doi.org/10.3390/nu7010306

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Yanaka A, Fahey JW, Fukumoto A, Nakayama M, Inoue S, Zhang S et al (2009) Dietary sulforaphane-rich broccoli sprouts reduce colonization and attenuate gastritis in Helicobacter pylori-infected mice and humans. Cancer Prev Res (Phila) 2(4):353–360. https://doi.org/10.1158/1940-6207.CAPR-08-0192

    Article  CAS  PubMed  Google Scholar 

  136. Chang YW, Jang JY, Kim YH, Kim JW, Shim JJ (2015) The effects of broccoli sprout extract containing sulforaphane on lipid peroxidation and helicobacter pylori infection in the gastric mucosa. Gut Liver 9(4):486–493. https://doi.org/10.5009/gnl14040

    Article  CAS  PubMed  Google Scholar 

  137. Zurek N, Pycia K, Pawlowska A, Potocki L, Kapusta IT (2023) Chemical profiling, bioactive properties, and anticancer and antimicrobial potential of Juglans regia L. leaves. Molecules. https://doi.org/10.3390/molecules28041989

    Article  PubMed  PubMed Central  Google Scholar 

  138. Pereira JA, Oliveira I, Sousa A, Ferreira IC, Bento A, Estevinho L (2008) Bioactive properties and chemical composition of six walnut (Juglans regia L.) cultivars. Food Chem Toxicol 46(6):2103–2111. https://doi.org/10.1016/j.fct.2008.02.002

    Article  CAS  PubMed  Google Scholar 

  139. Abdallah IB, Tlili N, Martinez-Force E, Rubio AG, Perez-Camino MC, Albouchi A, Boukhchina S (2015) Content of carotenoids, tocopherols, sterols, triterpenic and aliphatic alcohols, and volatile compounds in six walnuts (Juglans regia L.) varieties. Food Chem 173:972–978. https://doi.org/10.1016/j.foodchem.2014.10.095

    Article  CAS  PubMed  Google Scholar 

  140. Pycia K, Kapusta I, Jaworska G, Jankowska A (2019) Antioxidant properties, profile of polyphenolic compounds and tocopherol content in various walnut (Juglans regia L.) varieties. Eur Food Res Technol 245:607–616

    Article  CAS  Google Scholar 

  141. Sandu-Balan Tabacariu A, Ifrim IL, Patriciu OI, Stefanescu IA, Finaru AL (2024) Walnut by-products and elderberry extracts-sustainable alternatives for human and plant health. Molecules. https://doi.org/10.3390/molecules29020498

    Article  PubMed  PubMed Central  Google Scholar 

  142. Bouslamti M, Metouekel A, Chelouati T, El Moussaoui A, Barnossi AE, Chebaibi M et al (2022) Solanum elaeagnifolium Var. obtusifolium (Dunal) dunal: antioxidant, antibacterial, and antifungal activities of polyphenol-rich extracts chemically characterized by use of in vitro and in silico approaches. Molecules. https://doi.org/10.3390/molecules27248688

    Article  PubMed  PubMed Central  Google Scholar 

  143. Bespalov VG, Alexandrov VA, Semenov AL, Vysochina GI, Kostikova VA, Baranenko DA (2018) The inhibitory effect of Filipendula ulmaria (L.) Maxim. on colorectal carcinogenesis induced in rats by methylnitrosourea. J Ethnopharmacol 227:1–7. https://doi.org/10.1016/j.jep.2018.08.013

    Article  PubMed  Google Scholar 

  144. Bijttebier S, Van der Auwera A, Voorspoels S, Noten B, Hermans N, Pieters L, Apers S (2016) A first step in the quest for the active constituents in filipendula ulmaria (Meadowsweet): comprehensive phytochemical identification by liquid chromatography coupled to quadrupole-orbitrap mass spectrometry. Planta Med 82(6):559–572. https://doi.org/10.1055/s-0042-101943

    Article  CAS  PubMed  Google Scholar 

  145. Katanic J, Boroja T, Mihailovic V, Nikles S, Pan SP, Rosic G et al (2016) In vitro and in vivo assessment of meadowsweet (Filipendula ulmaria) as anti-inflammatory agent. J Ethnopharmacol 193:627–636. https://doi.org/10.1016/j.jep.2016.10.015

    Article  PubMed  Google Scholar 

  146. Pukalskiene M, Venskutonis PR, Pukalskas A (2015) Phytochemical characterization of Filipendula ulmaria by UPLC/Q-TOF-MS and evaluation of antioxidant activity. Rec Nat Prod 9(3):451

    CAS  Google Scholar 

  147. Savina T, Lisun V, Feduraev P, Skrypnik L (2023) Variation in phenolic compounds, antioxidant and antibacterial activities of extracts from different plant organs of meadowsweet (Filipendula ulmaria (L.) Maxim.). Molecules. https://doi.org/10.3390/molecules28083512

    Article  PubMed  PubMed Central  Google Scholar 

  148. Mérillon J-M, Riviere C (2018) Natural antimicrobial agents, vol 19. Springer

    Book  Google Scholar 

  149. Stringaro A, Colone M, Angiolella L (2018) Antioxidant, antifungal, antibiofilm, and cytotoxic activities of mentha spp. essential oils. Medicines (Basel). https://doi.org/10.3390/medicines5040112

    Article  PubMed  Google Scholar 

  150. Winska K, Maczka W, Lyczko J, Grabarczyk M, Czubaszek A, Szumny A (2019) Essential oils as antimicrobial agents-myth or real alternative? Molecules. https://doi.org/10.3390/molecules24112130

    Article  PubMed  PubMed Central  Google Scholar 

  151. Arora R, Singh B, Vig AP, Arora S (2016) Conventional and modified hydrodistillation method for the extraction of glucosinolate hydrolytic products: a comparative account. Springerplus 5:479. https://doi.org/10.1186/s40064-016-2021-z

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Baj T, Sieniawska E, Kowalski R, Wesolowskp M, Ulewicz-Magulska B (2015) Effectiveness of the deryng and clevenger-type apparatus in isolation of various types of components of essential oil from the Mutelina purpurea thell. Flowers Acta Pol Pharm 72(3):507–515

    CAS  PubMed  Google Scholar 

  153. Brnawi WI, Hettiarachchy NS, Horax R, Kumar-Phillips G, Ricke S (2019) Antimicrobial activity of leaf and bark cinnamon essential oils against Listeria monocytogenes and Salmonella typhimurium in broth system and on celery. J Food Process Preserv 43(3):e13888

    Article  Google Scholar 

  154. Guimaraes AC, Meireles LM, Lemos MF, Guimaraes MCC, Endringer DC, Fronza M, Scherer R (2019) Antibacterial activity of terpenes and terpenoids present in essential oils. Molecule. https://doi.org/10.3390/molecules24132471

    Article  Google Scholar 

  155. Iriti M, Colnaghi G, Chemat F, Smadja J, Faoro F, Visinoni FA (2006) Histo-cytochemistry and scanning electron microscopy of lavender glandular trichomes following conventional and microwave-assisted hydrodistillation of essential oils: a comparative study. Flavour Fragr J 21(4):704–712

    Article  CAS  Google Scholar 

  156. Nagoor Meeran MF, Javed H, Al Taee H, Azimullah S, Ojha SK (2017) Pharmacological properties and molecular mechanisms of thymol: prospects for its therapeutic potential and pharmaceutical development. Front Pharmacol 8:380. https://doi.org/10.3389/fphar.2017.00380

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Alizadeh A, Khosh-khui M, Javidnia K, Firuzi O, Jokar S (2011) Chemical composition of the essential oil, total phenolic content and antioxidant activity in Origanum majorana L. (Lamiaceae) cultivated in Iran. Adv Environ Biol 5(8):2326–2331

    CAS  Google Scholar 

  158. Farag SE, Aziz NH, Attia ES (1995) Effect of irradiation on the microbiological status and flavouring materials of selected spices. Z Lebensm Unters Forsch 201(3):283–288. https://doi.org/10.1007/BF01193005

    Article  CAS  PubMed  Google Scholar 

  159. Dimitrijević SI, Mihajlovski KR, Antonović DG, Milanović-Stevanović MR, Mijin DŽ (2007) A study of the synergistic antilisterial effects of a sub-lethal dose of lactic acid and essential oils from Thymus vulgaris L., Rosmarinus officinalis L. and Origanum vulgare L. Food Chem 104(2):774–782

    Article  Google Scholar 

  160. Penalver P, Huerta B, Borge C, Astorga R, Romero R, Perea A (2005) Antimicrobial activity of five essential oils against origin strains of the Enterobacteriaceae family. APMIS 113(1):1–6. https://doi.org/10.1111/j.1600-0463.2005.apm1130101.x

    Article  CAS  PubMed  Google Scholar 

  161. Ali-Shtayeh MS, Jamous RM, Al-Shafie JH, Elgharabah WA, Kherfan FA, Qarariah KH et al (2008) Traditional knowledge of wild edible plants used in Palestine (Northern West Bank): a comparative study. J Ethnobiol Ethnomed 4:13. https://doi.org/10.1186/1746-4269-4-13

    Article  PubMed  PubMed Central  Google Scholar 

  162. Abu-Lafi S, Odeh I, Dewik H, Qabajah M, Hanus LO, Dembitsky VM (2008) Thymol and carvacrol production from leaves of wild Palestinian Majorana syriaca. Bioresour Technol 99(9):3914–3918. https://doi.org/10.1016/j.biortech.2007.07.042

    Article  CAS  PubMed  Google Scholar 

  163. Dorman HJ, Deans SG (2000) Antimicrobial agents from plants: antibacterial activity of plant volatile oils. J Appl Microbiol 88(2):308–316. https://doi.org/10.1046/j.1365-2672.2000.00969.x

    Article  CAS  PubMed  Google Scholar 

  164. Guarda A, Rubilar JF, Miltz J, Galotto MJ (2011) The antimicrobial activity of microencapsulated thymol and carvacrol. Int J Food Microbiol 146(2):144–150. https://doi.org/10.1016/j.ijfoodmicro.2011.02.011

    Article  CAS  PubMed  Google Scholar 

  165. Wattanasatcha A, Rengpipat S, Wanichwecharungruang S (2012) Thymol nanospheres as an effective anti-bacterial agent. Int J Pharm 434(1–2):360–365. https://doi.org/10.1016/j.ijpharm.2012.06.017

    Article  CAS  PubMed  Google Scholar 

  166. Campos-Requena VH, Rivas BL, Pérez MA, Figueroa CR, Sanfuentes EA (2015) The synergistic antimicrobial effect of carvacrol and thymol in clay/polymer nanocomposite films over strawberry gray mold. LWT-Food Sci Technol 64(1):390–396

    Article  CAS  Google Scholar 

  167. Nostro A, Roccaro AS, Bisignano G, Marino A, Cannatelli MA, Pizzimenti FC et al (2007) Effects of oregano, carvacrol and thymol on Staphylococcus aureus and Staphylococcus epidermidis biofilms. J Med Microbiol 56(Pt 4):519–523. https://doi.org/10.1099/jmm.0.46804-0

    Article  CAS  PubMed  Google Scholar 

  168. Williamson EM (2001) Synergy and other interactions in phytomedicines. Phytomedicine 8(5):401–409. https://doi.org/10.1078/0944-7113-00060

    Article  CAS  PubMed  Google Scholar 

  169. Sebei K, Sakouhi F, Herchi W, Khouja ML, Boukhchina S (2015) Chemical composition and antibacterial activities of seven Eucalyptus species essential oils leaves. Biol Res 48(1):7. https://doi.org/10.1186/0717-6287-48-7

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Huang D, Xu J-G, Liu J-X, Zhang H, Hu Q (2014) Chemical constituents, antibacterial activity and mechanism of action of the essential oil from Cinnamomum cassia bark against four food-related bacteria. Microbiology 83:357–365

    Article  CAS  Google Scholar 

  171. Nguyen HTT, Miyamoto A, Nguyen HT, Pham HT, Hoang HT, Tong NTM et al (2023) Short communication: antibacterial effects of essential oils from Cinnamomum cassia bark and Eucalyptus globulus leaves-the involvements of major constituents. PLoS ONE 18(7):e0288787. https://doi.org/10.1371/journal.pone.0288787

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Zhang Y, Kong J, Xie Y, Guo Y, Cheng Y, Qian H, Yao W (2018) Essential oil components inhibit biofilm formation in Erwinia carotovora and Pseudomonas fluorescens via anti-quorum sensing activity. Lwt 92:133–139

    Article  CAS  Google Scholar 

  173. Trinh NT, Dumas E, Thanh ML, Degraeve P, Ben Amara C, Gharsallaoui A, Oulahal N (2015) Effect of a vietnamese Cinnamomum cassia essential oil and its major component trans-cinnamaldehyde on the cell viability, membrane integrity, membrane fluidity, and proton motive force of Listeria innocua. Can J Microbiol 61(4):263–271. https://doi.org/10.1139/cjm-2014-0481

    Article  CAS  PubMed  Google Scholar 

  174. Kolozsvarine Nagy J, Moricz AM, Boszormenyi A, Ambrus A, Schwarczinger I (2023) Antibacterial effect of essential oils and their components against Xanthomonas arboricola pv. pruni revealed by microdilution and direct bioautographic assays. Front Cell Infect Microbiol 13:1204027. https://doi.org/10.3389/fcimb.2023.1204027

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Bono G, Okpala COR, Vitale S, Ferrantelli V, Noto AD, Costa A et al (2017) Effects of different ozonized slurry-ice treatments and superchilling storage (− 1 degrees C) on microbial spoilage of two important pelagic fish species. Food Sci Nutr 5(6):1049–1056. https://doi.org/10.1002/fsn3.486

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Wang Y, Hong X, Liu J, Zhu J, Chen J (2020) Interactions between fish isolates Pseudomonas fluorescens and Staphylococcus aureus in dual-species biofilms and sensitivity to carvacrol. Food Microbiol 91:103506. https://doi.org/10.1016/j.fm.2020.103506

    Article  CAS  PubMed  Google Scholar 

  177. Xie Y, Zhang C, Mei J, Xie J (2023) Antimicrobial effect of Ocimum gratissimum L. essential oil on shewanella putrefaciens: insights based on the cell membrane and external structure. Int J Mol Sci. https://doi.org/10.3390/ijms241311066

    Article  PubMed  PubMed Central  Google Scholar 

  178. Miraj S, Kiani S (2016) Study of pharmacological effect of Ocimum basilicum: a review. Pharm Lett 8(9):276–280

    CAS  Google Scholar 

  179. Antora RA, Salleh RM (2017) Antihyperglycemic effect of Ocimum plants: a short review. Asian Pac J Trop Biomed 7(8):755–759

    Article  Google Scholar 

  180. Hashim M, Ahmad B, Drouet S, Hano C, Abbasi BH, Anjum S (2021) Comparative effects of different light sources on the production of key secondary metabolites in plants in vitro cultures. Plants (Basel). https://doi.org/10.3390/plants10081521

    Article  PubMed  PubMed Central  Google Scholar 

  181. Joshi RK (2014) Chemical composition and antimicrobial activity of the essential oil of Ocimum basilicum L. (sweet basil) from Western Ghats of North West Karnataka, India. Anc Sci Life 33(3):151–156. https://doi.org/10.4103/0257-7941.144618

    Article  PubMed  PubMed Central  Google Scholar 

  182. Ch M, Naz S, Sharif A, Akram M, Saeed M (2015) Biological and pharmacological properties of the sweet basil (Ocimum basilicum). Br J Pharm Res 7(5):330–339

    Article  Google Scholar 

  183. Rahayu S, Sirait LI, Aritonang T, Natzir R, Massi MN, Rauf S et al (2017) Ocimum basilicum as alternative natural cancer care. Int J Sci: Basic Appl Res (IJSBAR) 34(3):302–308

    Google Scholar 

  184. Güez CM, Souza ROD, Fischer P, Leão MFDM, Duarte JA, Boligon AA et al (2017) Evaluation of basil extract (Ocimum basilicum L.) on oxidative, anti-genotoxic and anti-inflammatory effects in human leukocytes cell cultures exposed to challenging agents. Braz J Pharm Sci. https://doi.org/10.1590/s2175-97902017000115098

    Article  Google Scholar 

  185. Touiss I, Ouahhoud S, Harnafi M, Khatib S, Bekkouch O, Amrani S, Harnafi H (2021) Toxicological evaluation and hepatoprotective efficacy of rosmarinic acid-rich extract from Ocimum basilicum L. Evid Based Complement Alternat Med 2021:6676998. https://doi.org/10.1155/2021/6676998

    Article  PubMed  PubMed Central  Google Scholar 

  186. Eid AM, Jaradat N, Shraim N, Hawash M, Issa L, Shakhsher M et al (2023) Assessment of anticancer, antimicrobial, antidiabetic, anti-obesity and antioxidant activity of Ocimum Basilicum seeds essential oil from Palestine. BMC Complement Med Ther 23(1):221. https://doi.org/10.1186/s12906-023-04058-w

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Yaldiz G, Camlica M, Erdonmez D (2023) Investigation of some basil genotypes in terms of their effect on bacterial communication system, and antimicrobial activity. Microb Pathog 182:106247. https://doi.org/10.1016/j.micpath.2023.106247

    Article  CAS  PubMed  Google Scholar 

  188. Debnath B, Singh WS, Das M, Goswami S, Singh MK, Maiti D, Manna K (2018) Role of plant alkaloids on human health: a review of biological activities. Mater Today Chem 9:56–72

    Article  CAS  Google Scholar 

  189. Robbers JE, Speedie MK, Tyler VE (1996) Pharmacognosy and pharmacobiotechnology. Williams & Wilkins, Philadelphia, pp 144–185

    Google Scholar 

  190. Amirkia V, Heinrich M (2014) Alkaloids as drug leads: a predictive structural and biodiversity-based analysis. Phytochem Lett 10:xlviii–liii

    Article  CAS  Google Scholar 

  191. Casciaro B, Mangiardi L, Cappiello F, Romeo I, Loffredo MR, Iazzetti A et al (2020) Naturally-occurring alkaloids of plant origin as potential antimicrobials against antibiotic-resistant infections. Molecules. https://doi.org/10.3390/molecules25163619

    Article  PubMed  PubMed Central  Google Scholar 

  192. Evans WC, Evans D, Trease GE (2009) Trease and evans pharmacognosy, 16th edn. Saunders/Elsevier, Edinburgh, New York

    Google Scholar 

  193. Othman L, Sleiman A, Abdel-Massih RM (2019) Antimicrobial activity of polyphenols and alkaloids in middle eastern plants. Front Microbiol 10:911. https://doi.org/10.3389/fmicb.2019.00911

    Article  PubMed  PubMed Central  Google Scholar 

  194. Kiuru P, D’Auria MV, Muller CD, Tammela P, Vuorela H, Yli-Kauhaluoma J (2014) Exploring marine resources for bioactive compounds. Planta Med 80(14):1234–1246. https://doi.org/10.1055/s-0034-1383001

    Article  CAS  PubMed  Google Scholar 

  195. Laport MS, Santos OC, Muricy G (2009) Marine sponges: potential sources of new antimicrobial drugs. Curr Pharm Biotechnol 10(1):86–105. https://doi.org/10.2174/138920109787048625

    Article  CAS  PubMed  Google Scholar 

  196. Blunt JW, Copp BR, Keyzers RA, Munro MH, Prinsep MR (2015) Marine natural products. Nat Prod Rep 32(2):116–211. https://doi.org/10.1039/c4np00144c

    Article  CAS  PubMed  Google Scholar 

  197. Ebada SS, Lin W, Proksch P (2010) Bioactive sesterterpenes and triterpenes from marine sponges: occurrence and pharmacological significance. Mar Drugs 8(2):313–346. https://doi.org/10.3390/md8020313

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Kim D, Lee IS, Jung JH, Yang SI (1999) Psammaplin A, a natural bromotyrosine derivative from a sponge, possesses the antibacterial activity against methicillin-resistant Staphylococcus aureus and the DNA gyrase-inhibitory activity. Arch Pharm Res 22(1):25–29. https://doi.org/10.1007/BF02976431

    Article  CAS  PubMed  Google Scholar 

  199. Keffer JL, Plaza A, Bewley CA (2009) Motualevic acids A-F, antimicrobial acids from the sponge Siliquariaspongia sp. Org Lett 11(5):1087–1090. https://doi.org/10.1021/ol802890b

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Yamada A, Kitamura H, Yamaguchi K, Fukuzawa S, Kamijima C, Yazawa K et al (1997) Development of chemical substances regulating biofilm formation. Bull Chem Soc Jpn 70(12):3061–3069

    Article  CAS  Google Scholar 

  201. Huigens RW 3rd, Richards JJ, Parise G, Ballard TE, Zeng W, Deora R, Melander C (2007) Inhibition of Pseudomonas aeruginosa biofilm formation with Bromoageliferin analogues. J Am Chem Soc 129(22):6966–6967. https://doi.org/10.1021/ja069017t

    Article  CAS  PubMed  Google Scholar 

  202. Ballard TE, Richards JJ, Wolfe AL, Melander C (2008) Synthesis and antibiofilm activity of a second-generation reverse-amide oroidin library: a structure-activity relationship study. Chemistry 14(34):10745–10761. https://doi.org/10.1002/chem.200801419

    Article  CAS  PubMed  Google Scholar 

  203. Huigens RW 3rd, Ma L, Gambino C, Moeller PD, Basso A, Cavanagh J et al (2008) Control of bacterial biofilms with marine alkaloid derivatives. Mol Biosyst 4(6):614–621. https://doi.org/10.1039/b719989a

    Article  CAS  PubMed  Google Scholar 

  204. Richards JJ, Ballard TE, Huigens RW 3rd, Melander C (2008) Synthesis and screening of an oroidin library against Pseudomonas aeruginosa biofilms. ChemBioChem 9(8):1267–1279. https://doi.org/10.1002/cbic.200700774

    Article  CAS  PubMed  Google Scholar 

  205. Richards JJ, Ballard TE, Melander C (2008) Inhibition and dispersion of Pseudomonas aeruginosa biofilms with reverse amide 2-aminoimidazole oroidin analogues. Org Biomol Chem 6(8):1356–1363. https://doi.org/10.1039/b719082d

    Article  CAS  PubMed  Google Scholar 

  206. Richards JJ, Huigens Iii RW, Ballard TE, Basso A, Cavanagh J, Melander C (2008) Inhibition and dispersion of proteobacterial biofilms. Chem Commun (Camb) 14:1698–1700. https://doi.org/10.1039/b719802g

    Article  CAS  Google Scholar 

  207. Huigens RW 3rd, Rogers SA, Steinhauer AT, Melander C (2009) Inhibition of Acinetobacter baumannii, Staphylococcus aureus and Pseudomonas aeruginosa biofilm formation with a class of TAGE-triazole conjugates. Org Biomol Chem 7(4):794–802. https://doi.org/10.1039/b817926c

    Article  CAS  PubMed  Google Scholar 

  208. Bunders CA, Richards JJ, Melander C (2010) Identification of aryl 2-aminoimidazoles as biofilm inhibitors in Gram-negative bacteria. Bioorg Med Chem Lett 20(12):3797–3800. https://doi.org/10.1016/j.bmcl.2010.04.042

    Article  CAS  PubMed  Google Scholar 

  209. Rogers SA, Bero JD, Melander C (2010) Chemical synthesis and biological screening of 2-aminoimidazole-based bacterial and fungal antibiofilm agents. ChemBioChem 11(3):396–410. https://doi.org/10.1002/cbic.200900617

    Article  CAS  PubMed  Google Scholar 

  210. Melander RJ, Liu HB, Stephens MD, Bewley CA, Melander C (2016) Marine sponge alkaloids as a source of anti-bacterial adjuvants. Bioorg Med Chem Lett 26(24):5863–5866. https://doi.org/10.1016/j.bmcl.2016.11.018

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Arai M, Ishida S, Setiawan A, Kobayashi M (2009) Haliclonacyclamines, tetracyclic alkylpiperidine alkaloids, as anti-dormant mycobacterial substances from a marine sponge of Haliclona sp. Chem Pharm Bull (Tokyo) 57(10):1136–1138. https://doi.org/10.1248/cpb.57.1136

    Article  CAS  PubMed  Google Scholar 

  212. Wijaya V, Jandourek O, Kroustkova J, Hradiska-Breiterova K, Korabecny J, Sobolova K et al (2022) Alkaloids of dicranostigma franchetianum (Papaveraceae) and Berberine derivatives as a new class of antimycobacterial agents. Biomolecules. https://doi.org/10.3390/biom12060844

    Article  PubMed  PubMed Central  Google Scholar 

  213. Dong LM, Huang LL, Dai H, Xu QL, Ouyang JK, Jia XC et al (2018) Anti-MRSA sesquiterpenes from the semi-mangrove plant myoporum bontioides A. Gray Mar Drugs. https://doi.org/10.3390/md16110438

    Article  PubMed  Google Scholar 

  214. Di Marco N, Lucero-Estrada C, Pungitore CR (2019) Aporphinoid alkaloids as antimicrobial agents against Yersinia enterocolitica. Lett Appl Microbiol 68(5):437–445. https://doi.org/10.1111/lam.13120

    Article  CAS  PubMed  Google Scholar 

  215. da Silva LC, Correia MT (2014) Plant lectins and toll-like receptors: implications for therapy of microbial infections. Front Microbiol 5:20. https://doi.org/10.3389/fmicb.2014.00020

    Article  PubMed  PubMed Central  Google Scholar 

  216. Nawrot R, Barylski J, Nowicki G, Broniarczyk J, Buchwald W, Gozdzicka-Jozefiak A (2014) Plant antimicrobial peptides. Folia Microbiol (Praha) 59(3):181–196. https://doi.org/10.1007/s12223-013-0280-4

    Article  CAS  PubMed  Google Scholar 

  217. Zhang LJ, Gallo RL (2016) Antimicrobial peptides. Curr Biol 26(1):R14-19. https://doi.org/10.1016/j.cub.2015.11.017

    Article  CAS  PubMed  Google Scholar 

  218. Konozy EHE, Osman MEM, Dirar AI, Ghartey-Kwansah G (2022) Plant lectins: a new antimicrobial frontier. Biomed Pharmacother 155:113735. https://doi.org/10.1016/j.biopha.2022.113735

    Article  CAS  PubMed  Google Scholar 

  219. Ghazarian H, Idoni B, Oppenheimer SB (2011) A glycobiology review: carbohydrates, lectins and implications in cancer therapeutics. Acta Histochem 113(3):236–247. https://doi.org/10.1016/j.acthis.2010.02.004

    Article  CAS  PubMed  Google Scholar 

  220. Jayanthi S, Ishwarya R, Anjugam M, Iswarya A, Karthikeyan S, Vaseeharan B (2017) Purification, characterization and functional analysis of the immune molecule lectin from the haemolymph of blue swimmer crab Portunus pelagicus and their antibiofilm properties. Fish Shellfish Immunol 62:227–237. https://doi.org/10.1016/j.fsi.2017.01.019

    Article  CAS  PubMed  Google Scholar 

  221. Klein RC, Fabres-Klein MH, de Oliveira LL, Feio RN, Malouin F, Ribon Ade O (2015) A C-type lectin from Bothrops jararacussu venom disrupts Staphylococcal biofilms. PLoS ONE 10(3):e0120514. https://doi.org/10.1371/journal.pone.0120514

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Khatun S, Khan M, Ashraduzzaman M, Pervin F, Bari L, Absar N (2009) Antibacterial activity and cytotoxicity of three lectins purified from drumstick (Moringa oleifera Lam.) leaves. J Bio-Sci 17:89–94

    Article  Google Scholar 

  223. Ferreira R, Napoleão TH, Santos AF, Sá R, Carneiro-da-Cunha M, Morais M et al (2011) Coagulant and antibacterial activities of the water-soluble seed lectin from Moringa oleifera. Lett Appl Microbiol 53(2):186–192

    Article  CAS  PubMed  Google Scholar 

  224. Procópio TF, de Siqueira Patriota LL, de Moura MC, da Silva PM, de Oliveira APS, do Nascimento Carvalho LV et al (2017) CasuL: a new lectin isolated from Calliandra surinamensis leaf pinnulae with cytotoxicity to cancer cells, antimicrobial activity and antibiofilm effect. Int J Biol Macromol 98:419–429

    Article  PubMed  Google Scholar 

  225. Singh RS, Thakur SR, Bansal P (2015) Algal lectins as promising biomolecules for biomedical research. Crit Rev Microbiol 41(1):77–88. https://doi.org/10.3109/1040841X.2013.798780

    Article  CAS  PubMed  Google Scholar 

  226. Singh RS, Walia AK, Khattar JS, Singh DP, Kennedy JF (2017) Cyanobacterial lectins characteristics and their role as antiviral agents. Int J Biol Macromol 102:475–496. https://doi.org/10.1016/j.ijbiomac.2017.04.041

    Article  CAS  PubMed  Google Scholar 

  227. Bhowmick S, Mazumdar A, Moulick A, Adam V (2020) Algal metabolites: an inevitable substitute for antibiotics. Biotechnol Adv 43:107571. https://doi.org/10.1016/j.biotechadv.2020.107571

    Article  CAS  PubMed  Google Scholar 

  228. Breitenbach Barroso Coelho LC, Marcelino Dos Santos Silva P, de Felix Oliveira W, de Moura MC, Viana Pontual E, Soares Gomes F et al (2018) Lectins as antimicrobial agents. J Appl Microbiol 125(5):1238–1252. https://doi.org/10.1111/jam.14055

    Article  CAS  PubMed  Google Scholar 

  229. Liao WR, Lin JY, Shieh WY, Jeng WL, Huang R (2003) Antibiotic activity of lectins from marine algae against marine vibrios. J Ind Microbiol Biotechnol 30(7):433–439. https://doi.org/10.1007/s10295-003-0068-7

    Article  CAS  PubMed  Google Scholar 

  230. Hung LD, Hirayama M, Ly BM, Hori K (2015) Purification, primary structure, and biological activity of the high-mannose N-glycan-specific lectin from cultivated Eucheuma denticulatum. J Appl Phycol 27(4):1657–1669. https://doi.org/10.1007/s10811-014-0441-0

    Article  CAS  PubMed  Google Scholar 

  231. Holanda ML, Melo VM, Silva LM, Amorim RC, Pereira MG, Benevides NM (2005) Differential activity of a lectin from Solieria filiformis against human pathogenic bacteria. Braz J Med Biol Res 38(12):1769–1773. https://doi.org/10.1590/s0100-879x2005001200005

    Article  CAS  PubMed  Google Scholar 

  232. Fernandez Romero JA, Paglini MG, Priano C, Koroch A, Rodriguez Y, Sailer J, Teleshova N (2021) Algal and cyanobacterial lectins and their antimicrobial properties. Mar Drugs. https://doi.org/10.3390/md19120687

    Article  PubMed  PubMed Central  Google Scholar 

  233. Jubeh B, Breijyeh Z, Karaman R (2020) Antibacterial prodrugs to overcome bacterial resistance. Molecules. https://doi.org/10.3390/molecules25071543

    Article  PubMed  PubMed Central  Google Scholar 

  234. Sarkar T, Chetia M, Chatterjee S (2021) Antimicrobial peptides and proteins: from nature’s reservoir to the laboratory and beyond. Front Chem 9:691532. https://doi.org/10.3389/fchem.2021.691532

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  235. Feurstein C, Meyer V, Jung S (2022) Structure-activity predictions from computational mining of protein databases to assist modular design of antimicrobial peptides. Front Microbiol 13:812903. https://doi.org/10.3389/fmicb.2022.812903

    Article  PubMed  PubMed Central  Google Scholar 

  236. Tam JP, Wang S, Wong KH, Tan WL (2015) Antimicrobial peptides from plants. Pharmaceuticals (Basel) 8(4):711–757. https://doi.org/10.3390/ph8040711

    Article  CAS  PubMed  Google Scholar 

  237. Yount NY, Yeaman MR (2004) Multidimensional signatures in antimicrobial peptides. Proc Natl Acad Sci U S A 101(19):7363–7368. https://doi.org/10.1073/pnas.0401567101

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  238. De Lucca AJ, Cleveland TE, Wedge DE (2005) Plant-derived antifungal proteins and peptides. Can J Microbiol 51(12):1001–1014. https://doi.org/10.1139/w05-063

    Article  PubMed  Google Scholar 

  239. Stec B (2006) Plant thionins: the structural perspective. Cell Mol Life Sci 63(12):1370–1385. https://doi.org/10.1007/s00018-005-5574-5

    Article  CAS  PubMed  Google Scholar 

  240. Wijaya R, Neumann GM, Condron R, Hughes AB, Polya GM (2000) Defense proteins from seed of Cassia fistula include a lipid transfer protein homologue and a protease inhibitory plant defensin. Plant Sci 159(2):243–255. https://doi.org/10.1016/s0168-9452(00)00348-4

    Article  CAS  PubMed  Google Scholar 

  241. Stotz HU, Thomson JG, Wang Y (2009) Plant defensins: defense, development and application. Plant Signal Behav 4(11):1010–1012. https://doi.org/10.4161/psb.4.11.9755

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  242. Pelegrini PB, Franco OL (2005) Plant gamma-thionins: novel insights on the mechanism of action of a multi-functional class of defense proteins. Int J Biochem Cell Biol 37(11):2239–2253. https://doi.org/10.1016/j.biocel.2005.06.011

    Article  CAS  PubMed  Google Scholar 

  243. Vieira Bard GC, Nascimento VV, Ribeiro SF, Rodrigues R, Perales J, Teixeira-Ferreira A et al (2015) Characterization of peptides from capsicum annuum hybrid seeds with inhibitory activity against alpha-amylase, Serine Proteinases and Fungi. Protein J 34(2):122–129. https://doi.org/10.1007/s10930-015-9604-3

    Article  CAS  PubMed  Google Scholar 

  244. Cammue BP, Thevissen K, Hendriks M, Eggermont K, Goderis IJ, Proost P et al (1995) A potent antimicrobial protein from onion seeds showing sequence homology to plant lipid transfer proteins. Plant Physiol 109(2):445–455. https://doi.org/10.1104/pp.109.2.445

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  245. Amador VC, Santos-Silva CAD, Vilela LMB, Oliveira-Lima M, de Santana Rego M, Roldan-Filho RS et al (2021) Lipid transfer proteins (LTPs)-structure, diversity and roles beyond antimicrobial activity. Antibiotics (Basel). https://doi.org/10.3390/antibiotics10111281

    Article  PubMed  Google Scholar 

  246. Regente MC, De La Canal L (2000) Purification, characterization and antifungal properties of a lipid-transfer protein from sunflower (Helianthus annuus) seeds. Physiol Plant 110(2):158–163

    Article  CAS  Google Scholar 

  247. Slavokhotova AA, Rogozhin EA (2020) Defense peptides from the alpha-hairpinin family are components of plant innate immunity. Front Plant Sci 11:465. https://doi.org/10.3389/fpls.2020.00465

    Article  PubMed  PubMed Central  Google Scholar 

  248. Slavokhotova AA, Shelenkov AA, Andreev YA, Odintsova TI (2017) Hevein-like antimicrobial peptides of plants. Biochemistry (Mosc) 82(13):1659–1674. https://doi.org/10.1134/S0006297917130065

    Article  CAS  PubMed  Google Scholar 

  249. Yeaman MR, Yount NY (2003) Mechanisms of antimicrobial peptide action and resistance. Pharmacol Rev 55(1):27–55. https://doi.org/10.1124/pr.55.1.2

    Article  CAS  PubMed  Google Scholar 

  250. Egorov TA, Odintsova TI, Pukhalsky VA, Grishin EV (2005) Diversity of wheat anti-microbial peptides. Peptides 26(11):2064–2073. https://doi.org/10.1016/j.peptides.2005.03.007

    Article  CAS  PubMed  Google Scholar 

  251. Slezina MP, Istomina EA, Kulakovskaya EV, Korostyleva TV, Odintsova TI (2022) The gamma-core motif peptides of AMPs from grasses display inhibitory activity against human and plant pathogens. Int J Mol Sci. https://doi.org/10.3390/ijms23158383

    Article  PubMed  PubMed Central  Google Scholar 

  252. Hinds L, Kenny O, Hossain MB, Walsh D, Sheehy E, Evans P et al (2017) Evaluating the antibacterial properties of polyacetylene and glucosinolate compounds with further identification of their presence within various Carrot (Daucus carota) and Broccoli (Brassica oleracea) Cultivars using high-performance liquid chromatography with a diode array detector and ultra performance liquid chromatography-tandem mass spectrometry analyses. J Agric Food Chem 65(33):7186–7191. https://doi.org/10.1021/acs.jafc.7b02029

    Article  CAS  PubMed  Google Scholar 

  253. Negri R (2015) Polyacetylenes from terrestrial plants and fungi: recent phytochemical and biological advances. Fitoterapia 106:92–109. https://doi.org/10.1016/j.fitote.2015.08.011

    Article  CAS  PubMed  Google Scholar 

  254. Harding VK, Heale JB (1981) The accumulation of inhibitory compounds in the induced resistance response of carrot root slices to Botrytis cinerea. Physiol PlantPathol 18(1):7–15. https://doi.org/10.1016/S0048-4059(81)80048-3

    Article  CAS  Google Scholar 

  255. Matsunaga H, Katano M, Yamamoto H, Mori M, Takata K (1989) Studies on the panaxytriol of Panax ginseng C. A. Meyer Isolation, determination and antitumor activity. Chem Pharm Bull (Tokyo) 37(5):1279–1281. https://doi.org/10.1248/cpb.37.1279

    Article  CAS  PubMed  Google Scholar 

  256. Matsunaga H, Katano M, Yamamoto H, Fujito H, Mori M, Takata K (1990) Cytotoxic activity of polyacetylene compounds in Panax ginseng C. A. Meyer. Chem Pharm Bull (Tokyo) 38(12):3480–3482. https://doi.org/10.1248/cpb.38.3480

    Article  CAS  PubMed  Google Scholar 

  257. Koch M, Bugni TS, Pond CD, Sondossi M, Dindi M, Piskaut P et al (2009) Antimycobacterial activity of Exocarpos latifolius is due to exocarpic acid. Planta Med 75(12):1326–1330. https://doi.org/10.1055/s-0029-1185687

    Article  CAS  PubMed  Google Scholar 

  258. Policegoudra RS, Rehna K, Rao LJ, Aradhya SM (2010) Antimicrobial, antioxidant, cytotoxicity and platelet aggregation inhibitory activity of a novel molecule isolated and characterized from mango ginger (Curcuma amada Roxb.) rhizome. J Biosci 35(2):231–240. https://doi.org/10.1007/s12038-010-0027-1

    Article  CAS  PubMed  Google Scholar 

  259. Fukuyama N, Shibuya M, Orihara Y (2012) Antimicrobial polyacetylenes from Panax ginseng hairy root culture. Chem Pharm Bull (Tokyo) 60(3):377–380. https://doi.org/10.1248/cpb.60.377

    Article  CAS  PubMed  Google Scholar 

  260. Zgoda JR, Freyer AJ, Killmer LB, Porter JR (2001) Polyacetylene carboxylic acids from Mitrephora celebica. J Nat Prod 64(10):1348–1349. https://doi.org/10.1021/np0102509

    Article  CAS  PubMed  Google Scholar 

  261. Panthama N, Kanokmedhakul S, Kanokmedhakul K (2010) Polyacetylenes from the roots of Polyalthia debilis. J Nat Prod 73(8):1366–1369. https://doi.org/10.1021/np1001913

    Article  CAS  PubMed  Google Scholar 

  262. Wang ML, Du J, Chen RY, Yu DQ (2000) Isolation and structure elucidation of novel gamma-lactones from Saccopetalum prolificum. J Asian Nat Prod Res 2(3):231–236. https://doi.org/10.1080/10286020008039916

    Article  CAS  PubMed  Google Scholar 

  263. Crockett SL, Robson NK (2011) Taxonomy and chemotaxonomy of the genus hypericum. Med Aromat Plant Sci Biotechnol 5(Special Issue 1):1–13

    PubMed  PubMed Central  Google Scholar 

  264. Robson NKB (2002) Studies in the genus Hypericum L. (Guttiferae) 4(2). Section 9. Hypericum sensu lato (part 2): subsection 1. Hypericum series 1. Hypericum. Bull Nat Hist Mus: Bot Ser 32(2):61–123. https://doi.org/10.1017/S096804460200004X

    Article  Google Scholar 

  265. Bruni R, Sacchetti G (2009) Factors affecting polyphenol biosynthesis in wild and field grown St. John’s Wort (Hypericum perforatum L. Hypericaceae/Guttiferae). Molecules 14(2):682–725. https://doi.org/10.3390/molecules14020682

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  266. Müller WE (2005) St. John's Wort and its active principles in depression and anxiety: Birkhäuser Basel

  267. Franchi GG, Nencini C, Collavoli E, Massarelli P (2011) Composition and antioxidant activity in vitro of different St. John’s Wort (Hypericum perforatum L.) extracts. J Med Plants Res 5(17):4349–4353

    CAS  Google Scholar 

  268. Avato P (2005) A survey of the Hypericum genus: secondary metabolites and bioactivity. Stud Nat Prod Chem 30:603–634. https://doi.org/10.1016/S1572-5995(05)80043-2

    Article  CAS  Google Scholar 

  269. Kakouri E, Trigas P, Daferera D, Skotti E, Tarantilis PA, Kanakis C (2023) Chemical characterization and antioxidant activity of nine hypericum species from greece. Antioxidants (Basel). https://doi.org/10.3390/antiox12040899

    Article  PubMed  Google Scholar 

  270. Sherif MM, Elshikh HH, Abdel-Aziz MM, Elaasser MM, Yosri M (2023) In vitro antibacterial and phytochemical screening of hypericum perforatum extract as potential antimicrobial agents against multi-drug-resistant (MDR) strains of clinical origin. Biomed Res Int 2023:6934398. https://doi.org/10.1155/2023/6934398

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  271. Supardy NA, Ibrahim D, Sulaiman SF, Zakaria NA (2012) Inhibition of Klebsiella pneumoniae ATCC 13883 cells by hexane extract of Halimeda discoidea (Decaisne) and the identification of its potential bioactive compounds. J Microbiol Biotechnol 22(6):872–881. https://doi.org/10.4014/jmb.1111.11053

    Article  PubMed  Google Scholar 

  272. Rajeshwari H, Nagveni S, Oli A, Parashar D, Chandrakanth KR (2009) Morphological changes of Klebsiella pneumoniae in response to Cefotaxime: a scanning electron microscope study. World J Microbiol Biotechnol 25(12):2263–2266. https://doi.org/10.1007/s11274-009-0126-z

    Article  Google Scholar 

  273. Satta G, Canepari P, Botta G, Fontana R (1980) Control of cell septation by lateral wall extension in a pH-conditional morphology mutant of Klebsiella pneumoniae. J Bacteriol 142(1):43–51. https://doi.org/10.1128/jb.142.1.43-51.1980

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  274. Spratt BG (1975) Distinct penicillin binding proteins involved in the division, elongation, and shape of Escherichia coli K12. Proc Natl Acad Sci U S A 72(8):2999–3003. https://doi.org/10.1073/pnas.72.8.2999

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  275. Okmen G, Balpinar N (2017) The biological activities of Hypericum Perforatum L. Afr J Tradit Complement Altern Med 14(1):213–218. https://doi.org/10.21010/ajtcam.v14i1.23

    Article  CAS  PubMed  Google Scholar 

  276. Taponen S, Simojoki H, Haveri M, Larsen HD, Pyorala S (2006) Clinical characteristics and persistence of bovine mastitis caused by different species of coagulase-negative staphylococci identified with API or AFLP. Vet Microbiol 115(1–3):199–207. https://doi.org/10.1016/j.vetmic.2006.02.001

    Article  CAS  PubMed  Google Scholar 

  277. Jiang DJ, Dai Z, Li YJ (2004) Pharmacological effects of xanthones as cardiovascular protective agents. Cardiovasc Drug Rev 22(2):91–102. https://doi.org/10.1111/j.1527-3466.2004.tb00133.x

    Article  CAS  PubMed  Google Scholar 

  278. Rukachaisirikul V, Kamkaew M, Sukavisit D, Phongpaichit S, Sawangchote P, Taylor WC (2003) Antibacterial xanthones from the leaves of Garcinia nigrolineata. J Nat Prod 66(12):1531–1535. https://doi.org/10.1021/np0303254

    Article  CAS  PubMed  Google Scholar 

  279. Rukachaisirikul V, Tadpetch K, Watthanaphanit A, Saengsanae N, Phongpaichit S (2005) Benzopyran, biphenyl, and tetraoxygenated xanthone derivatives from the twigs of Garcinia nigrolineata. J Nat Prod 68(8):1218–1221. https://doi.org/10.1021/np058050a

    Article  CAS  PubMed  Google Scholar 

  280. Sukpondma Y, Rukachaisirikul V, Phongpaichit S (2005) Xanthone and sesquiterpene derivatives from the fruits of Garcinia scortechinii. J Nat Prod 68(7):1010–1017. https://doi.org/10.1021/np0580098

    Article  CAS  PubMed  Google Scholar 

  281. Xiao ZY, Shiu WKP, Zeng YH, Mu Q, Gibbons S (2008) A naturally occurring inhibitory agent from Hypericum sampsonii with. Activity against multidrug-resistant Staphylococcus aureus. Pharm Biol 46(4):250–253. https://doi.org/10.1080/13880200701739405

    Article  CAS  Google Scholar 

  282. Sakagami Y, Iinuma M, Piyasena KG, Dharmaratne HR (2005) Antibacterial activity of alpha-mangostin against vancomycin resistant Enterococci (VRE) and synergism with antibiotics. Phytomedicine 12(3):203–208. https://doi.org/10.1016/j.phymed.2003.09.012

    Article  CAS  PubMed  Google Scholar 

  283. Suksamrarn S, Suwannapoch N, Phakhodee W, Thanuhiranlert J, Ratananukul P, Chimnoi N, Suksamrarn A (2003) Antimycobacterial activity of prenylated xanthones from the fruits of Garcinia mangostana. Chem Pharm Bull (Tokyo) 51(7):857–859. https://doi.org/10.1248/cpb.51.857

    Article  CAS  PubMed  Google Scholar 

  284. Conceicao LF, Ferreres F, Tavares RM, Dias AC (2006) Induction of phenolic compounds in Hypericum perforatum L. cells by Colletotrichum gloeosporioides elicitation. Phytochemistry 67(2):149–155. https://doi.org/10.1016/j.phytochem.2005.10.017

    Article  CAS  PubMed  Google Scholar 

  285. Franklin G, Conceicao LF, Kombrink E, Dias AC (2009) Xanthone biosynthesis in Hypericum perforatum cells provides antioxidant and antimicrobial protection upon biotic stress. Phytochemistry 70(1):60–68. https://doi.org/10.1016/j.phytochem.2008.10.016

    Article  CAS  PubMed  Google Scholar 

  286. Bayer MG, Heinrichs JH, Cheung AL (1996) The molecular architecture of the sar locus in Staphylococcus aureus. J Bacteriol 178(15):4563–4570. https://doi.org/10.1128/jb.178.15.4563-4570.1996

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  287. Wang L, Yang R, Yuan B, Liu Y, Liu C (2015) The antiviral and antimicrobial activities of licorice, a widely-used Chinese herb. Acta Pharm Sin B 5(4):310–315. https://doi.org/10.1016/j.apsb.2015.05.005

    Article  PubMed  PubMed Central  Google Scholar 

  288. Boucher H, Miller LG, Razonable RR (2010) Serious infections caused by methicillin-resistant Staphylococcus aureus. Clin Infect Dis 51(Suppl 2):S183-197. https://doi.org/10.1086/653519

    Article  CAS  PubMed  Google Scholar 

  289. Wang G, Li L, Wang X, Li X, Zhang Y, Yu J et al (2019) Hypericin enhances beta-lactam antibiotics activity by inhibiting sarA expression in methicillin-resistant Staphylococcus aureus. Acta Pharm Sin B 9(6):1174–1182. https://doi.org/10.1016/j.apsb.2019.05.002

    Article  PubMed  PubMed Central  Google Scholar 

  290. Tai J, Cheung S, Wu M, Hasman D (2012) Antiproliferation effect of Rosemary (Rosmarinus officinalis) on human ovarian cancer cells in vitro. Phytomedicine 19(5):436–443. https://doi.org/10.1016/j.phymed.2011.12.012

    Article  CAS  PubMed  Google Scholar 

  291. Bozin B, Mimica-Dukic N, Samojlik I, Jovin E (2007) Antimicrobial and antioxidant properties of rosemary and sage (Rosmarinus officinalis L. and Salvia officinalis L., Lamiaceae) essential oils. J Agric Food Chem 55(19):7879–7885. https://doi.org/10.1021/jf0715323

    Article  CAS  PubMed  Google Scholar 

  292. Houlihan CM, Ho CT, Chang SS (1984) Elucidation of the chemical structure of a novel antioxidant, rosmaridiphenol, isolated from rosemary. J Am Oil Chem Soc 61(6):1036–1039

    Article  CAS  Google Scholar 

  293. Sienkiewicz M, Lysakowska M, Pastuszka M, Bienias W, Kowalczyk E (2013) The potential of use basil and rosemary essential oils as effective antibacterial agents. Molecules 18(8):9334–9351. https://doi.org/10.3390/molecules18089334

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  294. Jiang Y, Wu N, Fu YJ, Wang W, Luo M, Zhao CJ et al (2011) Chemical composition and antimicrobial activity of the essential oil of Rosemary. Environ Toxicol Pharmacol 32(1):63–68. https://doi.org/10.1016/j.etap.2011.03.011

    Article  CAS  PubMed  Google Scholar 

  295. Bendeddouche MS, Benhassaini H, Hazem Z, Romane A (2011) Essential oil analysis and antibacterial activity of Rosmarinus tournefortii from Algeria. Nat Prod Commun 6(10):1511–1514

    CAS  PubMed  Google Scholar 

  296. Vongsak B, Sithisarn P, Mangmool S, Thongpraditchote S, Wongkrajang Y, Gritsanapan W (2013) Maximizing total phenolics, total flavonoids contents and antioxidant activity of Moringa oleifera leaf extract by the appropriate extraction method. Ind Crops Prod 44:566–571

    Article  CAS  Google Scholar 

  297. Azwanida N (2015) A review on the extraction methods use in medicinal plants, principle, strength and limitation. Med Aromat Plants 4:1–6

    Google Scholar 

  298. de Oliveira JR, Camargo SEA, de Oliveira LD (2019) Rosmarinus officinalis L. (rosemary) as therapeutic and prophylactic agent. J Biomed Sci 26(1):5. https://doi.org/10.1186/s12929-019-0499-8

    Article  PubMed  PubMed Central  Google Scholar 

  299. Fernandez-Lopez J, Zhi N, Aleson-Carbonell L, Perez-Alvarez JA, Kuri V (2005) Antioxidant and antibacterial activities of natural extracts: application in beef meatballs. Meat Sci 69(3):371–380. https://doi.org/10.1016/j.meatsci.2004.08.004

    Article  CAS  PubMed  Google Scholar 

  300. Govaris A, Florou-Paneri P, Botsoglou E, Giannenas I, Amvrosiadis I, Botsoglou N (2007) The inhibitory potential of feed supplementation with rosemary and/or α-tocopheryl acetate on microbial growth and lipid oxidation of turkey breast during refrigerated storage. LWT-Food Sci Technol 40(2):331–337

    Article  CAS  Google Scholar 

  301. Gomez-Estaca J, Lopez de Lacey A, Lopez-Caballero ME, Gomez-Guillen MC, Montero P (2010) Biodegradable gelatin-chitosan films incorporated with essential oils as antimicrobial agents for fish preservation. Food Microbiol 27(7):889–896. https://doi.org/10.1016/j.fm.2010.05.012

    Article  CAS  PubMed  Google Scholar 

  302. Camo J, Beltrán JA, Roncalés P (2008) Extension of the display life of lamb with an antioxidant active packaging. Meat Sci 80(4):1086–1091

    Article  CAS  PubMed  Google Scholar 

  303. Ouattara B, Sabato SF, Lacroix M (2001) Combined effect of antimicrobial coating and gamma irradiation on shelf life extension of pre-cooked shrimp (Penaeus spp.). Int J Food Microbiol 68(1–2):1–9. https://doi.org/10.1016/s0168-1605(01)00436-6

    Article  CAS  PubMed  Google Scholar 

  304. Prabuseenivasan S, Jayakumar M, Ignacimuthu S (2006) In vitro antibacterial activity of some plant essential oils. BMC Complement Altern Med 6:39. https://doi.org/10.1186/1472-6882-6-39

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  305. Mihajilov-Krstev T, Radnović D, Kitić D, Stojanović-Radić Z, Zlatković B (2010) Antimicrobial activity of Satureja hortensis L. essential oil against pathogenic microbial strains. Arch Biol Sci 62(1):159–166

    Article  Google Scholar 

  306. Bernardes WA, Lucarini R, Tozatti MG, Souza MG, Silva ML, Filho AA et al (2010) Antimicrobial activity of Rosmarinus officinalis against oral pathogens: relevance of carnosic acid and carnosol. Chem Biodivers 7(7):1835–1840. https://doi.org/10.1002/cbdv.200900301

    Article  CAS  PubMed  Google Scholar 

  307. Nieto G, Ros G, Castillo J (2018) Antioxidant and antimicrobial properties of rosemary (Rosmarinus officinalis, L.): a review. Medicines (Basel). https://doi.org/10.3390/medicines5030098

    Article  PubMed  Google Scholar 

  308. Ekambaram SP, Perumal SS, Balakrishnan A, Marappan N, Gajendran SS, Viswanathan V (2016) Antibacterial synergy between rosmarinic acid and antibiotics against methicillin-resistant Staphylococcus aureus. J Intercult Ethnopharmacol 5(4):358–363. https://doi.org/10.5455/jice.20160906035020

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  309. Abu El-Wafa WM, Ahmed RH, Ramadan MA (2020) Synergistic effects of pomegranate and rosemary extracts in combination with antibiotics against antibiotic resistance and biofilm formation of Pseudomonas aeruginosa. Braz J Microbiol 51(3):1079–1092. https://doi.org/10.1007/s42770-020-00284-3

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  310. Nasri H, Bahmani M, Shahinfard N, Moradi Nafchi A, Saberianpour S, Rafieian Kopaei M (2015) Medicinal plants for the treatment of acne vulgaris: a review of recent evidences. Jundishapur J Microbiol 8(11):e25580. https://doi.org/10.5812/jjm.25580

    Article  PubMed  PubMed Central  Google Scholar 

  311. Kali A, Bhuvaneshwar D, Charles PM, Seetha KS (2016) Antibacterial synergy of curcumin with antibiotics against biofilm producing clinical bacterial isolates. J Basic Clin Pharm 7(3):93–96. https://doi.org/10.4103/0976-0105.183265

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  312. Pramila D, Xavier R, Marimuthu K, Kathiresan S, Khoo M, Senthilkumar M et al (2012) Phytochemical analysis and antimicrobial potential of methanolic leaf extract of peppermint (Mentha piperita: Lamiaceae). J Med Plants Res 6(2):331–335

    Google Scholar 

  313. Romano CS, Abadi K, Repetto V, Vojnov AA, Moreno S (2009) Synergistic antioxidant and antibacterial activity of rosemary plus butylated derivatives. Food Chem 115(2):456–461

    Article  CAS  Google Scholar 

  314. Han YA, Song CW, Koh WS, Yon GH, Kim YS, Ryu SY et al (2013) Anti-inflammatory effects of the Zingiber officinale roscoe constituent 12-dehydrogingerdione in lipopolysaccharide-stimulated Raw 264.7 cells. Phytother Res 27(8):1200–1205. https://doi.org/10.1002/ptr.4847

    Article  CAS  PubMed  Google Scholar 

  315. Stoner GD (2013) Ginger: is it ready for prime time? Cancer Prev Res (Phila) 6(4):257–262. https://doi.org/10.1158/1940-6207.CAPR-13-0055

    Article  PubMed  Google Scholar 

  316. Prasad S, Tyagi AK (2015) Ginger and its constituents: role in prevention and treatment of gastrointestinal cancer. Gastroenterol Res Pract 2015:142979. https://doi.org/10.1155/2015/142979

    Article  PubMed  PubMed Central  Google Scholar 

  317. Ji K, Fang L, Zhao H, Li Q, Shi Y, Xu C et al (2017) Ginger oleoresin alleviated gamma-ray irradiation-induced reactive oxygen species via the Nrf2 protective response in human mesenchymal stem cells. Oxid Med Cell Longev 2017:1480294. https://doi.org/10.1155/2017/1480294

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  318. Schadich E, Hlavac J, Volna T, Varanasi L, Hajduch M, Dzubak P (2016) Effects of ginger phenylpropanoids and quercetin on Nrf2-ARE pathway in human BJ fibroblasts and HaCaT keratinocytes. Biomed Res Int 2016:2173275. https://doi.org/10.1155/2016/2173275

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  319. Yeh H-Y, Chuang C-H, Chen H-C, Wan C-J, Chen T-L, Lin L-Y (2014) Bioactive components analysis of two various gingers (Zingiber officinale Roscoe) and antioxidant effect of ginger extracts. LWT-Food Sci Technol 55(1):329–334

    Article  CAS  Google Scholar 

  320. Zhang M, Viennois E, Prasad M, Zhang Y, Wang L, Zhang Z et al (2016) Edible ginger-derived nanoparticles: a novel therapeutic approach for the prevention and treatment of inflammatory bowel disease and colitis-associated cancer. Biomaterials 101:321–340. https://doi.org/10.1016/j.biomaterials.2016.06.018

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  321. Kumar NV, Murthy PS, Manjunatha JR, Bettadaiah BK (2014) Synthesis and quorum sensing inhibitory activity of key phenolic compounds of ginger and their derivatives. Food Chem 159:451–457. https://doi.org/10.1016/j.foodchem.2014.03.039

    Article  CAS  PubMed  Google Scholar 

  322. Citronberg J, Bostick R, Ahearn T, Turgeon DK, Ruffin MT, Djuric Z et al (2013) Effects of ginger supplementation on cell-cycle biomarkers in the normal-appearing colonic mucosa of patients at increased risk for colorectal cancer: results from a pilot, randomized, and controlled trial. Cancer Prev Res (Phila) 6(4):271–281. https://doi.org/10.1158/1940-6207.CAPR-12-0327

    Article  PubMed  Google Scholar 

  323. Mao QQ, Xu XY, Cao SY, Gan RY, Corke H, Beta T, Li HB (2019) Bioactive compounds and bioactivities of ginger (Zingiber officinale Roscoe). Foods. https://doi.org/10.3390/foods8060185

  324. Ho SC, Chang KS, Lin CC (2013) Anti-neuroinflammatory capacity of fresh ginger is attributed mainly to 10-gingerol. Food Chem 141(3):3183–3191. https://doi.org/10.1016/j.foodchem.2013.06.010

    Article  CAS  PubMed  Google Scholar 

  325. Wei CK, Tsai YH, Korinek M, Hung PH, El-Shazly M, Cheng YB et al (2017) 6-paradol and 6-shogaol, the pungent compounds of ginger, promote glucose utilization in adipocytes and myotubes, and 6-paradol reduces blood glucose in high-fat diet-fed mice. Int J Mol Sci. https://doi.org/10.3390/ijms18010168

    Article  PubMed  PubMed Central  Google Scholar 

  326. Suk S, Kwon GT, Lee E, Jang WJ, Yang H, Kim JH et al (2017) Gingerenone A, a polyphenol present in ginger, suppresses obesity and adipose tissue inflammation in high-fat diet-fed mice. Mol Nutr Food Res. https://doi.org/10.1002/mnfr.201700139

    Article  PubMed  PubMed Central  Google Scholar 

  327. Akinyemi AJ, Thome GR, Morsch VM, Stefanello N, Goularte JF, Belló-Klein A et al (2015) Effect of dietary supplementation of ginger and turmeric rhizomes on angiotensin-1 converting enzyme (ACE) and arginase activities in L-NAME induced hypertensive rats. J Funct Foods 17:792–801

    Article  CAS  Google Scholar 

  328. Townsend EA, Siviski ME, Zhang Y, Xu C, Hoonjan B, Emala CW (2013) Effects of ginger and its constituents on airway smooth muscle relaxation and calcium regulation. Am J Respir Cell Mol Biol 48(2):157–163. https://doi.org/10.1165/rcmb.2012-0231OC

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  329. Zhang C, Xie Y, Qiu W, Mei J, Xie J (2023) Antibacterial and antibiofilm efficacy and mechanism of ginger (Zingiber officinale) essential oil against Shewanella putrefaciens. Plants (Basel). https://doi.org/10.3390/plants12081720

    Article  PubMed  PubMed Central  Google Scholar 

  330. Wang X, Shen Y, Thakur K, Han J, Zhang JG, Hu F, Wei ZJ (2020) Antibacterial activity and mechanism of ginger essential oil against Escherichia coli and Staphylococcus aureus. Molecules. https://doi.org/10.3390/molecules25173955

    Article  PubMed  PubMed Central  Google Scholar 

  331. Zhang L, Qin M, Yin J, Liu X, Zhou J, Zhu Y, Liu Y (2022) Antibacterial activity and mechanism of ginger extract against Ralstonia solanacearum. J Appl Microbiol 133(4):2642–2654. https://doi.org/10.1111/jam.15733

    Article  CAS  PubMed  Google Scholar 

  332. Atai Z, Atapour M, Mohseni M (2009) Inhibitory effect of ginger extract on Candida albicans. Am J Appl Sci 6(6):1067–1069

    Article  Google Scholar 

  333. Park M, Bae J, Lee DS (2008) Antibacterial activity of [10]-gingerol and [12]-gingerol isolated from ginger rhizome against periodontal bacteria. Phytother Res 22(11):1446–1449. https://doi.org/10.1002/ptr.2473

    Article  CAS  PubMed  Google Scholar 

  334. Chairgulprasert V, Prasertsongskun S, Wichaporn W (2005) Chemical constituents of the essential oil and antibacterial activity of Zingiber wrayi var. halabala. Songklanakarin J Sci Technol 27(4):813–818

    Google Scholar 

  335. Sagar PK, Sharma P, Singh R (2020) Antibacterial efficacy of different combinations of clove, eucalyptus, ginger, and selected antibiotics against clinical isolates of Pseudomonas aeruginosa. Ayu 41(2):123–129. https://doi.org/10.4103/ayu.AYU_101_19

    Article  PubMed  Google Scholar 

  336. Nagoshi C, Shiota S, Kuroda T, Hatano T, Yoshida T, Kariyama R, Tsuchiya T (2006) Synergistic effect of [10]-gingerol and aminoglycosides against vancomycin-resistant enterococci (VRE). Biol Pharm Bull 29(3):443–447. https://doi.org/10.1248/bpb.29.443

    Article  CAS  PubMed  Google Scholar 

  337. Keyhani A, Sharifi I, Salarkia E, Khosravi A, Tavakoli Oliaee R, Babaei Z et al (2021) In vitro and in vivo therapeutic potentials of 6-gingerol in combination with amphotericin B for treatment of Leishmania major infection: Powerful synergistic and multifunctional effects. Int Immunopharmacol 101(Pt B):108274. https://doi.org/10.1016/j.intimp.2021.108274

    Article  CAS  PubMed  Google Scholar 

  338. Khan A, Azam M, Allemailem KS, Alrumaihi F, Almatroudi A, Alhumaydhi FA et al (2021) Coadministration of ginger extract and fluconazole shows a synergistic effect in the treatment of drug-resistant vulvovaginal candidiasis. Infect Drug Resist 14:1585–1599. https://doi.org/10.2147/IDR.S305503

    Article  PubMed  PubMed Central  Google Scholar 

  339. Butera D, Tesoriere L, Di Gaudio F, Bongiorno A, Allegra M, Pintaudi AM et al (2002) Antioxidant activities of sicilian prickly pear (Opuntia ficus indica) fruit extracts and reducing properties of its betalains: betanin and indicaxanthin. J Agric Food Chem 50(23):6895–6901. https://doi.org/10.1021/jf025696p

    Article  CAS  PubMed  Google Scholar 

  340. Khan NS, Ahmad A, Hadi SM (2000) Anti-oxidant, pro-oxidant properties of tannic acid and its binding to DNA. Chem Biol Interact 125(3):177–189. https://doi.org/10.1016/s0009-2797(00)00143-5

    Article  CAS  PubMed  Google Scholar 

  341. You BR, Park WH (2010) Gallic acid-induced lung cancer cell death is related to glutathione depletion as well as reactive oxygen species increase. Toxicol In Vitro 24(5):1356–1362. https://doi.org/10.1016/j.tiv.2010.04.009

    Article  CAS  PubMed  Google Scholar 

  342. El-Mostafa K, El Kharrassi Y, Badreddine A, Andreoletti P, Vamecq J, El Kebbaj MS et al (2014) Nopal cactus (Opuntia ficus-indica) as a source of bioactive compounds for nutrition, health and disease. Molecules 19(9):14879–14901. https://doi.org/10.3390/molecules190914879

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  343. Schaffer S, Schmitt-Schillig S, Muller WE, Eckert GP (2005) Antioxidant properties of Mediterranean food plant extracts: geographical differences. J Physiol Pharmacol 56(Suppl 1):115–124

    PubMed  Google Scholar 

  344. Valente LM, Scheinvar LA, da Silva GC, Antunes AP, dos Santos FA, Oliveira TF et al (2007) Evaluation of the antitumor and trypanocidal activities and alkaloid profile in species of Brazilian Cactaceae. Pharmacogn Mag 3(11):167–172

    Google Scholar 

  345. Abidi S, Salem HB, Vasta V, Priolo A (2009) Supplementation with barley or spineless cactus (Opuntia ficus indica f. inermis) cladodes on digestion, growth and intramuscular fatty acid composition in sheep and goats receiving oaten hay. Small Ruminant Res 87(1–3):9–16

    Article  Google Scholar 

  346. Soel SM, Choi OS, Bang MH, Yoon Park JH, Kim WK (2007) Influence of conjugated linoleic acid isomers on the metastasis of colon cancer cells in vitro and in vivo. J Nutr Biochem 18(10):650–657. https://doi.org/10.1016/j.jnutbio.2006.10.011

    Article  CAS  PubMed  Google Scholar 

  347. Kaur M, Kaur A, Sharma R (2012) Pharmacological actions of Opuntia ficus indica: a review. J Appl Pharm Sci 2(7):15–18

    Google Scholar 

  348. Ennouri M, Ammar I, Khemakhem B, Attia H (2014) Chemical composition and antibacterial activity of Opuntia ficus-indica f. inermis (cactus pear) flowers. J Med Food 17(8):908–914. https://doi.org/10.1089/jmf.2013.0089

    Article  CAS  PubMed  Google Scholar 

  349. Khemiri I, Essghaier Hedi B, Sadfi Zouaoui N, Ben Gdara N, Bitri L (2019) The antimicrobial and wound healing potential of Opuntia ficus indica L. inermis extracted oil from Tunisia. Evid Based Complement Alternat Med 2019:9148782. https://doi.org/10.1155/2019/9148782

    Article  PubMed  PubMed Central  Google Scholar 

  350. Ammar I, Bardaa S, Mzid M, Sahnoun Z, Rebaii T, Attia H, Ennouri M (2015) Antioxidant, antibacterial and in vivo dermal wound healing effects of opuntia flower extracts. Int J Biol Macromol 81:483–490. https://doi.org/10.1016/j.ijbiomac.2015.08.039

    Article  CAS  PubMed  Google Scholar 

  351. Alqurashi AS, Al Masoudi LM, Hamdi H, Abu Zaid A (2022) Chemical composition and antioxidant, antiviral, antifungal, antibacterial and anticancer potentials of Opuntia ficus-indica seed oil. Molecules. https://doi.org/10.3390/molecules27175453

    Article  PubMed  PubMed Central  Google Scholar 

  352. Elkady WM, Raafat MM, Abdel-Aziz MM, Al-Huqail AA, Ashour ML, Fathallah N (2022) Endophytic fungus from Opuntia ficus-indica: a source of potential bioactive antimicrobial compounds against multidrug-resistant bacteria. Plants (Basel). https://doi.org/10.3390/plants11081070

    Article  PubMed  PubMed Central  Google Scholar 

  353. Elkady WM, Bishr MM, Abdel-Aziz MM, Salama OM (2020) Identification and isolation of anti-pneumonia bioactive compounds from Opuntia ficus-indica fruit waste peels. Food Funct 11(6):5275–5283. https://doi.org/10.1039/d0fo00817f

    Article  CAS  PubMed  Google Scholar 

  354. Kabashima K, Honda T, Ginhoux F, Egawa G (2019) The immunological anatomy of the skin. Nat Rev Immunol 19(1):19–30

    Article  CAS  PubMed  Google Scholar 

  355. Hsu Y-C, Li L, Fuchs E (2014) Emerging interactions between skin stem cells and their niches. Nat Med 20(8):847–856

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  356. Ovaere P, Lippens S, Vandenabeele P, Declercq W (2009) The emerging roles of serine protease cascades in the epidermis. Trends Biochem Sci 34(9):453–463

    Article  CAS  PubMed  Google Scholar 

  357. Wolf K, Müller R, Borgmann S, Brocker E-B, Friedl P (2003) Amoeboid shape change and contact guidance: T-lymphocyte crawling through fibrillar collagen is independent of matrix remodeling by MMPs and other proteases. Blood 102(9):3262–3269

    Article  CAS  PubMed  Google Scholar 

  358. Fernandes A, Rodrigues PM, Pintado M, Tavaria FK (2023) A systematic review of natural products for skin applications: targeting inflammation, wound healing, and photo-aging. Phytomedicine 115:154824. https://doi.org/10.1016/j.phymed.2023.154824

    Article  CAS  PubMed  Google Scholar 

  359. Varadé J, Magadán S, González-Fernández Á (2021) Human immunology and immunotherapy: main achievements and challenges. Cell Mol Immunol 18(4):805–828

    Article  PubMed  Google Scholar 

  360. Albanesi C, Madonna S, Gisondi P, Girolomoni G (2018) The interplay between keratinocytes and immune cells in the pathogenesis of psoriasis. Front Immunol 9:1549

    Article  PubMed  PubMed Central  Google Scholar 

  361. Chen L, Yu J (2016) Modulation of toll-like receptor signaling in innate immunity by natural products. Int Immunopharmacol 37:65–70

    Article  PubMed  PubMed Central  Google Scholar 

  362. Wang S, Li Z, Ma Y, Liu Y, Lin C-C, Li S et al (2021) Immunomodulatory effects of green tea polyphenols. Molecules 26(12):3755

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  363. Nguyen AV, Soulika AM (2019) The dynamics of the skin’s immune system. Int J Mol Sci 20(8):1811

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  364. Akita S (2019) Wound repair and regeneration: mechanisms, signaling, vol 20. MDPI, p 6328

    Google Scholar 

  365. Trinh X-T, Long N-V, Van Anh LT, Nga PT, Giang NN, Chien PN et al (2022) A comprehensive review of natural compounds for wound healing: targeting bioactivity perspective. Int J Mol Sci 23(17):9573

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  366. MacLeod AS, Mansbridge JN (2016) The innate immune system in acute and chronic wounds. Adv Wound Care 5(2):65–78

    Article  Google Scholar 

  367. Dunnill C, Patton T, Brennan J, Barrett J, Dryden M, Cooke J et al (2017) Reactive oxygen species (ROS) and wound healing: the functional role of ROS and emerging ROS-modulating technologies for augmentation of the healing process. Int Wound J 14(1):89–96. https://doi.org/10.1111/iwj.12557

    Article  PubMed  Google Scholar 

  368. Edwards R, Harding KG (2004) Bacteria and wound healing. Curr Opin Infect Dis 17(2):91–96. https://doi.org/10.1097/00001432-200404000-00004

    Article  PubMed  Google Scholar 

  369. Hardman MJ, Ashcroft GS (2008) Estrogen, not intrinsic aging, is the major regulator of delayed human wound healing in the elderly. Genome Biol 9(5):R80. https://doi.org/10.1186/gb-2008-9-5-r80

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  370. Patel S, Srivastava S, Singh MR, Singh D (2019) Mechanistic insight into diabetic wounds: pathogenesis, molecular targets and treatment strategies to pace wound healing. Biomed Pharmacother 112:108615. https://doi.org/10.1016/j.biopha.2019.108615

    Article  CAS  PubMed  Google Scholar 

  371. Guo S, Dipietro LA (2010) Factors affecting wound healing. J Dent Res 89(3):219–229. https://doi.org/10.1177/0022034509359125

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  372. Rodrigues M, Kosaric N, Bonham CA, Gurtner GC (2019) Wound healing: a cellular perspective. Physiol Rev 99(1):665–706. https://doi.org/10.1152/physrev.00067.2017

    Article  CAS  PubMed  Google Scholar 

  373. Ding S, Jiang H, Fang J (2018) Regulation of immune function by polyphenols. J Immunol Res 2018:1–8

    Article  Google Scholar 

  374. Shedoeva A, Leavesley D, Upton Z, Fan C (2019) Wound healing and the use of medicinal plants. Evid Based Complement Alternat Med 2019:2684108. https://doi.org/10.1155/2019/2684108

    Article  PubMed  PubMed Central  Google Scholar 

  375. Nemeth E, Bernath J (2008) Biological activities of yarrow species (Achillea spp.). Curr Pharm Des 14(29):3151–3167. https://doi.org/10.2174/138161208786404281

    Article  CAS  PubMed  Google Scholar 

  376. Bakkali F, Averbeck S, Averbeck D, Idaomar M (2008) Biological effects of essential oils: a review. Food Chem Toxicol 46(2):446–475. https://doi.org/10.1016/j.fct.2007.09.106

    Article  CAS  PubMed  Google Scholar 

  377. Nemeth E (2005) Essential oil composition of species in the genus Achillea. J Essent Oil Res 17(5):501–512

    Article  CAS  Google Scholar 

  378. Vitale S, Colanero S, Placidi M, Di Emidio G, Tatone C, Amicarelli F, D’Alessandro AM (2022) Phytochemistry and biological activity of medicinal plants in wound healing: an overview of current research. Molecule. https://doi.org/10.3390/molecules27113566

    Article  Google Scholar 

  379. Tadic V, Arsic I, Zvezdanovic J, Zugic A, Cvetkovic D, Pavkov S (2017) The estimation of the traditionally used yarrow (Achillea millefolium L. Asteraceae) oil extracts with anti-inflamatory potential in topical application. J Ethnopharmacol 199:138–148. https://doi.org/10.1016/j.jep.2017.02.002

    Article  CAS  PubMed  Google Scholar 

  380. Jettanacheawchankit S, Sasithanasate S, Sangvanich P, Banlunara W, Thunyakitpisal P (2009) Acemannan stimulates gingival fibroblast proliferation; expressions of keratinocyte growth factor-1, vascular endothelial growth factor, and type I collagen; and wound healing. J Pharmacol Sci 109(4):525–531. https://doi.org/10.1254/jphs.08204fp

    Article  CAS  PubMed  Google Scholar 

  381. Garcia-Orue I, Gainza G, Gutierrez FB, Aguirre JJ, Evora C, Pedraz JL et al (2017) Novel nanofibrous dressings containing rhEGF and Aloe vera for wound healing applications. Int J Pharm 523(2):556–566. https://doi.org/10.1016/j.ijpharm.2016.11.006

    Article  CAS  PubMed  Google Scholar 

  382. Xu D, Pan Y, Chen J (2019) Chemical constituents, pharmacologic properties, and clinical applications of Bletilla striata. Front Pharmacol 10:1168. https://doi.org/10.3389/fphar.2019.01168

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  383. He X, Wang X, Fang J, Zhao Z, Huang L, Guo H, Zheng X (2017) Bletilla striata: medicinal uses, phytochemistry and pharmacological activities. J Ethnopharmacol 195:20–38. https://doi.org/10.1016/j.jep.2016.11.026

    Article  CAS  PubMed  Google Scholar 

  384. Pang Y, Wang D, Hu X, Wang H, Fu W, Fan Z et al (2014) Effect of volatile oil from Blumea Balsamifera (L.) DC. leaves on wound healing in mice. J Tradit Chin Med 34(6):716–724. https://doi.org/10.1016/s0254-6272(15)30087-x

    Article  PubMed  Google Scholar 

  385. Chen M, Qin JJ, Fu JJ, Hu XJ, Liu XH, Zhang WD, Jin HZ (2010) Blumeaenes A-J, sesquiterpenoid esters from Blumea balsamifera with NO inhibitory activity. Planta Med 76(9):897–902. https://doi.org/10.1055/s-0029-1240800

    Article  CAS  PubMed  Google Scholar 

  386. Dozmorov MG, Yang Q, Wu W, Wren J, Suhail MM, Woolley CL et al (2014) Differential effects of selective frankincense (Ru Xiang) essential oil versus non-selective sandalwood (Tan Xiang) essential oil on cultured bladder cancer cells: a microarray and bioinformatics study. Chin Med 9:18. https://doi.org/10.1186/1749-8546-9-18

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  387. Zhao W, Entschladen F, Liu H, Niggemann B, Fang Q, Zaenker KS, Han R (2003) Boswellic acid acetate induces differentiation and apoptosis in highly metastatic melanoma and fibrosarcoma cells. Cancer Detect Prev 27(1):67–75. https://doi.org/10.1016/s0361-090x(02)00170-8

    Article  CAS  PubMed  Google Scholar 

  388. Jahandideh M, Hajimehdipoor H, Mortazavi SA, Dehpour A, Hassanzadeh G (2016) A wound healing formulation based on Iranian traditional medicine and Its HPTLC fingerprint. Iran J Pharm Res 15(Suppl):149–157

    PubMed  PubMed Central  Google Scholar 

  389. Banno N, Akihisa T, Yasukawa K, Tokuda H, Tabata K, Nakamura Y et al (2006) Anti-inflammatory activities of the triterpene acids from the resin of Boswellia carteri. J Ethnopharmacol 107(2):249–253. https://doi.org/10.1016/j.jep.2006.03.006

    Article  CAS  PubMed  Google Scholar 

  390. Tewtrakul S, Tungcharoen P, Sudsai T, Karalai C, Ponglimanont C, Yodsaoue O (2015) Antiinflammatory and wound healing effects of Caesalpinia sappan L. Phytother Res 29(6):850–856. https://doi.org/10.1002/ptr.5321

    Article  PubMed  Google Scholar 

  391. Zhao H, Wang X, Li W, Koike K, Bai H (2014) A new minor homoisoflavonoid from Caesalpinia sappan. Nat Prod Res 28(2):102–105. https://doi.org/10.1080/14786419.2013.847439

    Article  CAS  PubMed  Google Scholar 

  392. Dinda M, Dasgupta U, Singh N, Bhattacharyya D, Karmakar P (2015) PI3K-mediated proliferation of fibroblasts by Calendula officinalis tincture: implication in wound healing. Phytother Res 29(4):607–616. https://doi.org/10.1002/ptr.5293

    Article  CAS  PubMed  Google Scholar 

  393. Nicolaus C, Junghanns S, Hartmann A, Murillo R, Ganzera M, Merfort I (2017) In vitro studies to evaluate the wound healing properties of Calendula officinalis extracts. J Ethnopharmacol 196:94–103. https://doi.org/10.1016/j.jep.2016.12.006

    Article  PubMed  Google Scholar 

  394. Leach MJ (2008) Calendula officinalis and wound healing: a systematic review. Wounds 20(8):236–243

    PubMed  Google Scholar 

  395. Wu QB, Wang Y, Liang L, Jiang Q, Guo ML, Zhang JJ (2013) Novel triterpenoid saponins from the seeds of Celosia argentea L. Nat Prod Res 27(15):1353–1360. https://doi.org/10.1080/14786419.2012.740034

    Article  CAS  PubMed  Google Scholar 

  396. Priya KS, Arumugam G, Rathinam B, Wells A, Babu M (2004) Celosia argentea Linn. leaf extract improves wound healing in a rat burn wound model. Wound Repair Regen 12(6):618–625. https://doi.org/10.1111/j.1067-1927.2004.12603.x

    Article  PubMed  Google Scholar 

  397. Shukla A, Rasik AM, Jain GK, Shankar R, Kulshrestha DK, Dhawan BN (1999) In vitro and in vivo wound healing activity of asiaticoside isolated from Centella asiatica. J Ethnopharmacol 65(1):1–11. https://doi.org/10.1016/s0378-8741(98)00141-x

    Article  CAS  PubMed  Google Scholar 

  398. Liu M, Dai Y, Li Y, Luo Y, Huang F, Gong Z, Meng Q (2008) Madecassoside isolated from Centella asiatica herbs facilitates burn wound healing in mice. Planta Med 74(8):809–815. https://doi.org/10.1055/s-2008-1074533

    Article  CAS  PubMed  Google Scholar 

  399. Somboonwong J, Kankaisre M, Tantisira B, Tantisira MH (2012) Wound healing activities of different extracts of Centella asiatica in incision and burn wound models: an experimental animal study. BMC Complement Altern Med 12:103. https://doi.org/10.1186/1472-6882-12-103

    Article  PubMed  PubMed Central  Google Scholar 

  400. Yuan X, Han L, Fu P, Zeng H, Lv C, Chang W et al (2018) Cinnamaldehyde accelerates wound healing by promoting angiogenesis via up-regulation of PI3K and MAPK signaling pathways. Lab Invest 98(6):783–798. https://doi.org/10.1038/s41374-018-0025-8

    Article  CAS  PubMed  Google Scholar 

  401. Mansour G, Ouda S, Shaker A, Abdallah HM (2014) Clinical efficacy of new aloe vera- and myrrh-based oral mucoadhesive gels in the management of minor recurrent aphthous stomatitis: a randomized, double-blind, vehicle-controlled study. J Oral Pathol Med 43(6):405–409. https://doi.org/10.1111/jop.12130

    Article  PubMed  Google Scholar 

  402. Abdul-Ghani RA, Loutfy N, Hassan A (2009) Myrrh and trematodoses in Egypt: an overview of safety, efficacy and effectiveness profiles. Parasitol Int 58(3):210–214. https://doi.org/10.1016/j.parint.2009.04.006

    Article  PubMed  Google Scholar 

  403. Shen T, Li GH, Wang XN, Lou HX (2012) The genus Commiphora: a review of its traditional uses, phytochemistry and pharmacology. J Ethnopharmacol 142(2):319–330. https://doi.org/10.1016/j.jep.2012.05.025

    Article  CAS  PubMed  Google Scholar 

  404. Nomicos EY (2007) Myrrh: medical marvel or myth of the Magi? Holist Nurs Pract 21(6):308–323. https://doi.org/10.1097/01.HNP.0000298616.32846.34

    Article  PubMed  Google Scholar 

  405. Joe B, Vijaykumar M, Lokesh BR (2004) Biological properties of curcumin-cellular and molecular mechanisms of action. Crit Rev Food Sci Nutr 44(2):97–111. https://doi.org/10.1080/10408690490424702

    Article  CAS  PubMed  Google Scholar 

  406. Thangapazham RL, Sharad S, Maheshwari RK (2013) Skin regenerative potentials of curcumin. BioFactors 39(1):141–149. https://doi.org/10.1002/biof.1078

    Article  CAS  PubMed  Google Scholar 

  407. Su X, Liu X, Wang S, Li B, Pan T, Liu D et al (2017) Wound-healing promoting effect of total tannins from Entada phaseoloides (L.) Merr. in rats. Burns 43(4):830–838. https://doi.org/10.1016/j.burns.2016.10.010

    Article  PubMed  Google Scholar 

  408. Dong Y, Shi H, Yang H, Peng Y, Wang M, Li X (2012) Antioxidant phenolic compounds from the stems of Entada phaseoloides. Chem Biodivers 9(1):68–79. https://doi.org/10.1002/cbdv.201100002

    Article  CAS  PubMed  Google Scholar 

  409. Tie L, Yang HQ, An Y, Liu SQ, Han J, Xu Y et al (2012) Ganoderma lucidum polysaccharide accelerates refractory wound healing by inhibition of mitochondrial oxidative stress in type 1 diabetes. Cell Physiol Biochem 29(3–4):583–594. https://doi.org/10.1159/000338512

    Article  CAS  PubMed  Google Scholar 

  410. Sanodiya BS, Thakur GS, Baghel RK, Prasad GB, Bisen PS (2009) Ganoderma lucidum: a potent pharmacological macrofungus. Curr Pharm Biotechnol 10(8):717–742. https://doi.org/10.2174/138920109789978757

    Article  CAS  PubMed  Google Scholar 

  411. Hsieh TC, Wu JM (2013) Regulation of cell cycle transition and induction of apoptosis in HL-60 leukemia cells by the combination of Coriolus versicolor and Ganoderma lucidum. Int J Mol Med 32(1):251–257. https://doi.org/10.3892/ijmm.2013.1378

    Article  PubMed  Google Scholar 

  412. Li F, Zhang Y, Zhong Z (2011) Antihyperglycemic effect of ganoderma lucidum polysaccharides on streptozotocin-induced diabetic mice. Int J Mol Sci 12(9):6135–6145. https://doi.org/10.3390/ijms12096135

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  413. Gao Y, Tang W, Gao H, Chan E, Lan J, Zhou S (2004) Ganoderma lucidum polysaccharide fractions accelerate healing of acetic acid-induced ulcers in rats. J Med Food 7(4):417–421. https://doi.org/10.1089/jmf.2004.7.417

    Article  CAS  PubMed  Google Scholar 

  414. Ran X, Ma L, Peng C, Zhang H, Qin LP (2011) Ligusticum chuanxiong Hort: a review of chemistry and pharmacology. Pharm Biol 49(11):1180–1189. https://doi.org/10.3109/13880209.2011.576346

    Article  CAS  PubMed  Google Scholar 

  415. Xiao Y, Wang YC, Li LL, Jin YC, Sironi L, Wang Y, Wang Y (2014) Lactones from Ligusticum chuanxiong Hort. reduces atherosclerotic lesions in apoE-deficient mice via inhibiting over expression of NF-kB-dependent adhesion molecules. Fitoterapia 95:240–246. https://doi.org/10.1016/j.fitote.2014.02.012

    Article  CAS  PubMed  Google Scholar 

  416. Liu JL, Zheng SL, Fan QJ, Yuan JC, Yang SM, Kong FL (2015) Optimisation of high-pressure ultrasonic-assisted extraction and antioxidant capacity of polysaccharides from the rhizome of Ligusticum chuanxiong. Int J Biol Macromol 76:80–85. https://doi.org/10.1016/j.ijbiomac.2015.02.031

    Article  CAS  PubMed  Google Scholar 

  417. Lin YL, Wang GJ, Huang CL, Lee YC, Liao WC, Lai WL et al (2009) Ligusticum chuanxiong as a potential neuroprotectant for preventing serum deprivation-induced apoptosis in rat pheochromocytoma cells: functional roles of mitogen-activated protein kinases. J Ethnopharmacol 122(3):417–423. https://doi.org/10.1016/j.jep.2009.02.011

    Article  PubMed  Google Scholar 

  418. Liang MJ, He LC, Yang GD (2005) Screening, analysis and in vitro vasodilatation of effective components from Ligusticum chuanxiong. Life Sci 78(2):128–133. https://doi.org/10.1016/j.lfs.2005.04.038

    Article  CAS  PubMed  Google Scholar 

  419. Cho J-G, Lee M-K, Lee J-W, Park H-J, Lee D-Y, Lee Y-H et al (2010) Physicochemical characterization and NMR assignments of ginsenosides Rb 1, Rb 2, R c, and Rd isolated from Panax ginseng. J Ginseng Res 34(2):113–121

    Article  CAS  Google Scholar 

  420. Kim WK, Song SY, Oh WK, Kaewsuwan S, Tran TL, Kim WS, Sung JH (2013) Wound-healing effect of ginsenoside Rd from leaves of Panax ginseng via cyclic AMP-dependent protein kinase pathway. Eur J Pharmacol 702(1–3):285–293. https://doi.org/10.1016/j.ejphar.2013.01.048

    Article  CAS  PubMed  Google Scholar 

  421. Kim YS, Cho IH, Jeong MJ, Jeong SJ, Nah SY, Cho YS et al (2011) Therapeutic effect of total ginseng saponin on skin wound healing. J Ginseng Res 35(3):360–367. https://doi.org/10.5142/jgr.2011.35.3.360

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  422. Chen X, Wang M, Xu X, Liu J, Mei B, Fu P et al (2017) Panax ginseng total protein promotes proliferation and secretion of collagen in NIH/3T3 cells by activating extracellular signal-related kinase pathway. J Ginseng Res 41(3):411–418. https://doi.org/10.1016/j.jgr.2017.02.001

    Article  PubMed  PubMed Central  Google Scholar 

  423. Lee J, Jung E, Lee J, Huh S, Kim J, Park M et al (2007) Panax ginseng induces human Type I collagen synthesis through activation of Smad signaling. J Ethnopharmacol 109(1):29–34. https://doi.org/10.1016/j.jep.2006.06.008

    Article  CAS  PubMed  Google Scholar 

  424. Choi S (2002) Epidermis proliferative effect of the Panax ginseng ginsenoside Rb2. Arch Pharm Res 25(1):71–76. https://doi.org/10.1007/BF02975265

    Article  CAS  PubMed  Google Scholar 

  425. Lee MH, Kao L, Lin CC (2011) Comparison of the antioxidant and transmembrane permeative activities of the different Polygonum cuspidatum extracts in phospholipid-based microemulsions. J Agric Food Chem 59(17):9135–9141. https://doi.org/10.1021/jf201577f

    Article  CAS  PubMed  Google Scholar 

  426. Jiao Y, Wu Y, Du D (2018) Polydatin inhibits cell proliferation, invasion and migration, and induces cell apoptosis in hepatocellular carcinoma. Braz J Med Biol Res 51(4):e6867. https://doi.org/10.1590/1414-431X20176867

    Article  PubMed  PubMed Central  Google Scholar 

  427. Liu Z, Wei F, Chen LJ, Xiong HR, Liu YY, Luo F et al (2013) In vitro and in vivo studies of the inhibitory effects of emodin isolated from Polygonum cuspidatum on Coxsakievirus B(4). Molecules 18(10):11842–11858. https://doi.org/10.3390/molecules181011842

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  428. Peng W, Qin R, Li X, Zhou H (2013) Botany, phytochemistry, pharmacology, and potential application of Polygonum cuspidatum Sieb.et Zucc.: a review. J Ethnopharmacol 148(3):729–745. https://doi.org/10.1016/j.jep.2013.05.007

    Article  CAS  PubMed  Google Scholar 

  429. Tang T, Yin L, Yang J, Shan G (2007) Emodin, an anthraquinone derivative from Rheum officinale Baill, enhances cutaneous wound healing in rats. Eur J Pharmacol 567(3):177–185. https://doi.org/10.1016/j.ejphar.2007.02.033

    Article  CAS  PubMed  Google Scholar 

  430. Wang H, Song H, Yue J, Li J, Hou YB, Deng JL (2012) Rheum officinale (a traditional Chinese medicine) for chronic kidney disease. Cochrane Database Syst Rev 7:CD008000. https://doi.org/10.1002/14651858.CD008000.pub2

    Article  Google Scholar 

  431. Yanwen W, Wenyuan G, Xiaohe X, Yi L (2005) Calorimetric investigation of the effect of hydroxyanthraquinones in Rheum officinale Baill on Staphylococcus aureus growth. Thermochim Acta 429(2):167–170

    Article  Google Scholar 

  432. Zhang H, Chen J, Cen Y (2018) Burn wound healing potential of a polysaccharide from Sanguisorba officinalis L. in mice. Int J Biol Macromol 112:862–867. https://doi.org/10.1016/j.ijbiomac.2018.01.214

    Article  CAS  PubMed  Google Scholar 

  433. Yang JH, Hwang YH, Gu MJ, Cho WK, Ma JY (2015) Ethanol extracts of Sanguisorba officinalis L. suppress TNF-alpha/IFN-gamma-induced pro-inflammatory chemokine production in HaCaT cells. Phytomedicine 22(14):1262–1268. https://doi.org/10.1016/j.phymed.2015.09.006

    Article  CAS  PubMed  Google Scholar 

  434. Yu T, Lee YJ, Yang HM, Han S, Kim JH, Lee Y et al (2011) Inhibitory effect of Sanguisorba officinalis ethanol extract on NO and PGE(2) production is mediated by suppression of NF-kappaB and AP-1 activation signaling cascade. J Ethnopharmacol 134(1):11–17. https://doi.org/10.1016/j.jep.2010.08.060

    Article  CAS  PubMed  Google Scholar 

  435. He X, Fang J, Huang L, Wang J, Huang X (2015) Sophora flavescens Ait.: Traditional usage, phytochemistry and pharmacology of an important traditional Chinese medicine. J Ethnopharmacol 172:10–29. https://doi.org/10.1016/j.jep.2015.06.010

    Article  CAS  PubMed  Google Scholar 

  436. Balekar N, Nakpheng T, Katkam NG, Srichana T (2012) Wound healing activity of ent-kaura-9(11),16-dien-19-oic acid isolated from Wedelia trilobata (L.) leaves. Phytomedicine 19(13):1178–1184. https://doi.org/10.1016/j.phymed.2012.07.014

    Article  CAS  PubMed  Google Scholar 

  437. Govindappa M, Naga S, Poojashri M, Sadananda T, Chandrappa C (2011) Antimicrobial, antioxidant and in vitro anti-inflammatory activity of ethanol extract and active phytochemical screening of Wedelia trilobata (L.) Hitchc. J Pharmacogn Phytother 3(3):43–51

    Google Scholar 

  438. Li XQ, Kang R, Huo JC, Xie YH, Wang SW, Cao W (2017) Wound-healing activity of Zanthoxylum bungeanum maxim seed oil on experimentally burned rats. Pharmacogn Mag 13(51):363–371. https://doi.org/10.4103/pm.pm_211_16

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  439. Artaria C, Maramaldi G, Bonfigli A, Rigano L, Appendino G (2011) Lifting properties of the alkamide fraction from the fruit husks of Zanthoxylum bungeanum. Int J Cosmet Sci 33(4):328–333. https://doi.org/10.1111/j.1468-2494.2010.00629.x

    Article  CAS  PubMed  Google Scholar 

  440. Balloux F, van Dorp L (2017) Q&A: What are pathogens, and what have they done to and for us? BMC Biol 15(1):91. https://doi.org/10.1186/s12915-017-0433-z

    Article  PubMed  PubMed Central  Google Scholar 

  441. Negut I, Grumezescu V, Grumezescu AM (2018) Treatment strategies for infected wounds. Molecules. https://doi.org/10.3390/molecules23092392

    Article  PubMed  PubMed Central  Google Scholar 

  442. Miguel SP, Moreira AF, Correia IJ (2019) Chitosan based-asymmetric membranes for wound healing: a review. Int J Biol Macromol 127:460–475. https://doi.org/10.1016/j.ijbiomac.2019.01.072

    Article  CAS  PubMed  Google Scholar 

  443. Ali Demir S, Erdal C (2011) Biopolymers as wound healing materials: challenges and new strategies. Biomater Appl Nanomed. https://doi.org/10.5772/25177

    Article  Google Scholar 

  444. Rehm BH (2010) Bacterial polymers: biosynthesis, modifications and applications. Nat Rev Microbiol 8(8):578–592. https://doi.org/10.1038/nrmicro2354

    Article  CAS  PubMed  Google Scholar 

  445. Smith AM, Moxon S, Morris GA (2016) Biopolymers as wound healing materials. pp 261–287.https://doi.org/10.1016/b978-1-78242-456-7.00013-1

  446. Hamdan S, Pastar I, Drakulich S, Dikici E, Tomic-Canic M, Deo S, Daunert S (2017) Nanotechnology-driven therapeutic interventions in wound healing: potential uses and applications. ACS Cent Sci 3(3):163–175. https://doi.org/10.1021/acscentsci.6b00371

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  447. Sahana TG, Rekha PD (2018) Biopolymers: applications in wound healing and skin tissue engineering. Mol Biol Rep 45(6):2857–2867. https://doi.org/10.1007/s11033-018-4296-3

    Article  CAS  PubMed  Google Scholar 

  448. Kanikireddy V, Varaprasad K, Jayaramudu T, Karthikeyan C, Sadiku R (2020) Carboxymethyl cellulose-based materials for infection control and wound healing: a review. Int J Biol Macromol 164:963–975. https://doi.org/10.1016/j.ijbiomac.2020.07.160

    Article  CAS  PubMed  Google Scholar 

  449. Gaspar-Pintiliescu A, Stanciuc AM, Craciunescu O (2019) Natural composite dressings based on collagen, gelatin and plant bioactive compounds for wound healing: a review. Int J Biol Macromol 138:854–865. https://doi.org/10.1016/j.ijbiomac.2019.07.155

    Article  CAS  PubMed  Google Scholar 

  450. Felician FF, Yu RH, Li MZ, Li CJ, Chen HQ, Jiang Y et al (2019) The wound healing potential of collagen peptides derived from the jellyfish Rhopilema esculentum. Chin J Traumatol 22(1):12–20. https://doi.org/10.1016/j.cjtee.2018.10.004

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  451. Bolla SR, Mohammed Al-Subaie A, Yousuf Al-Jindan R, Papayya Balakrishna J, Kanchi Ravi P, Veeraraghavan VP et al (2019) In vitro wound healing potency of methanolic leaf extract of Aristolochia saccata is possibly mediated by its stimulatory effect on collagen-1 expression. Heliyon 5(5):e01648. https://doi.org/10.1016/j.heliyon.2019.e01648

    Article  PubMed  PubMed Central  Google Scholar 

  452. Chattopadhyay S, Raines RT (2014) Review collagen-based biomaterials for wound healing. Biopolymers 101(8):821–833. https://doi.org/10.1002/bip.22486

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  453. Naomi R, Bt Hj Idrus R, Fauzi MB (2020) Plant- versus bacterial-derived cellulose for wound healing: a review. Int J Environ Res Public Health 17(18):7. https://doi.org/10.3390/ijerph17186803

    Article  CAS  Google Scholar 

  454. Chan HC, Chia CH, Zakaria S, Ahmad I, Dufresne A (2012) Production and characterisation of cellulose and nano-crystalline cellulose from kenaf core wood. BioResources 8(1):785–794

    Article  Google Scholar 

  455. Kurczewska J, Pecyna P, Ratajczak M, Gajecka M, Schroeder G (2017) Halloysite nanotubes as carriers of vancomycin in alginate-based wound dressing. Saudi Pharm J 25(6):911–920. https://doi.org/10.1016/j.jsps.2017.02.007

    Article  PubMed  PubMed Central  Google Scholar 

  456. Dowling MB, Chaturvedi A, MacIntire IC, Javvaji V, Gustin J, Raghavan SR et al (2016) Determination of efficacy of a novel alginate dressing in a lethal arterial injury model in swine. Injury 47(10):2105–2109. https://doi.org/10.1016/j.injury.2016.05.003

    Article  PubMed  Google Scholar 

  457. Sun J, Tan H (2013) Alginate-based biomaterials for regenerative medicine applications. Materials (Basel) 6(4):1285–1309. https://doi.org/10.3390/ma6041285

    Article  CAS  PubMed  Google Scholar 

  458. Ozaki CK, Hamdan AD, Barshes NR, Wyers M, Hevelone ND, Belkin M, Nguyen LL (2015) Prospective, randomized, multi-institutional clinical trial of a silver alginate dressing to reduce lower extremity vascular surgery wound complications. J Vasc Surg 61(2):419-427e411. https://doi.org/10.1016/j.jvs.2014.07.034

    Article  PubMed  Google Scholar 

  459. Prasathkumar M, Sadhasivam S (2021) Chitosan/Hyaluronic acid/Alginate and an assorted polymers loaded with honey, plant, and marine compounds for progressive wound healing-know-how. Int J Biol Macromol 186:656–685. https://doi.org/10.1016/j.ijbiomac.2021.07.067

    Article  CAS  PubMed  Google Scholar 

  460. Zhai P, Peng X, Li B, Liu Y, Sun H, Li X (2020) The application of hyaluronic acid in bone regeneration. Int J Biol Macromol 151:1224–1239. https://doi.org/10.1016/j.ijbiomac.2019.10.169

    Article  CAS  PubMed  Google Scholar 

  461. Luo Z, Dai Y, Gao H (2019) Development and application of hyaluronic acid in tumor targeting drug delivery. Acta Pharm Sin B 9(6):1099–1112. https://doi.org/10.1016/j.apsb.2019.06.004

    Article  PubMed  PubMed Central  Google Scholar 

  462. Papakonstantinou E, Roth M, Karakiulakis G (2012) Hyaluronic acid: a key molecule in skin aging. Dermatoendocrinol 4(3):253–258. https://doi.org/10.4161/derm.21923

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  463. Lee EH, Lim SJ, Lee MK (2019) Chitosan-coated liposomes to stabilize and enhance transdermal delivery of indocyanine green for photodynamic therapy of melanoma. Carbohydr Polym 224:115143. https://doi.org/10.1016/j.carbpol.2019.115143

    Article  CAS  PubMed  Google Scholar 

  464. Zepon KM, Martins MM, Marques MS, Heckler JM, Dal Pont Morisso F, Moreira MG et al (2019) Smart wound dressing based on kappa-carrageenan/locust bean gum/cranberry extract for monitoring bacterial infections. Carbohydr Polym 206:362–370. https://doi.org/10.1016/j.carbpol.2018.11.014

    Article  CAS  PubMed  Google Scholar 

  465. Sheokand B, Vats M, Kumar A, Srivastava CM, Bahadur I, Pathak SR (2023) Natural polymers used in the dressing materials for wound healing: past, present and future. J Polym Sci. https://doi.org/10.1002/pol.20220734

    Article  Google Scholar 

  466. Arthe R, Arivuoli D, Ravi V (2020) Preparation and characterization of bioactive silk fibroin/paramylon blend films for chronic wound healing. Int J Biol Macromol 154:1324–1331. https://doi.org/10.1016/j.ijbiomac.2019.11.010

    Article  CAS  PubMed  Google Scholar 

  467. Pollini M, Paladini F (2020) Bioinspired Materials for wound healing application: the potential of silk fibroin. Materials (Basel). https://doi.org/10.3390/ma13153361

    Article  PubMed  Google Scholar 

  468. Patil PP, Reagan MR, Bohara RA (2020) Silk fibroin and silk-based biomaterial derivatives for ideal wound dressings. Int J Biol Macromol 164:4613–4627. https://doi.org/10.1016/j.ijbiomac.2020.08.041

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  469. Wei H, Chen Q, Lin L, Sha C, Li T, Liu Y et al (2021) Regulation of exosome production and cargo sorting. Int J Biol Sci 17(1):163–177. https://doi.org/10.7150/ijbs.53671

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  470. Chung IM, Rajakumar G, Venkidasamy B, Subramanian U, Thiruvengadam M (2020) Exosomes: current use and future applications. Clin Chim Acta 500:226–232. https://doi.org/10.1016/j.cca.2019.10.022

    Article  CAS  PubMed  Google Scholar 

  471. Tran PHL, Xiang D, Tran TTD, Yin W, Zhang Y, Kong L et al (2020) Exosomes and nanoengineering: a match made for precision therapeutics. Adv Mater 32(18):e1904040. https://doi.org/10.1002/adma.201904040

    Article  CAS  PubMed  Google Scholar 

  472. Rahmati S, Shojaei F, Shojaeian A, Rezakhani L, Dehkordi MB (2020) An overview of current knowledge in biological functions and potential theragnostic applications of exosomes. Chem Phys Lipids 226:104836. https://doi.org/10.1016/j.chemphyslip.2019.104836

    Article  CAS  PubMed  Google Scholar 

  473. Meldolesi J (2018) Exosomes and ectosomes in intercellular communication. Curr Biol 28(8):R435–R444. https://doi.org/10.1016/j.cub.2018.01.059

    Article  CAS  PubMed  Google Scholar 

  474. Doyle LM, Wang MZ (2019) Overview of extracellular vesicles, their origin, composition, purpose, and methods for exosome isolation and analysis. Cells. https://doi.org/10.3390/cells8070727

    Article  PubMed  PubMed Central  Google Scholar 

  475. Zaborowski MP, Balaj L, Breakefield XO, Lai CP (2015) Extracellular vesicles: composition, biological relevance, and methods of study. Bioscience 65(8):783–797. https://doi.org/10.1093/biosci/biv084

    Article  PubMed  PubMed Central  Google Scholar 

  476. Kerris EWJ, Hoptay C, Calderon T, Freishtat RJ (2020) Platelets and platelet extracellular vesicles in hemostasis and sepsis. J Investig Med 68(4):813–820. https://doi.org/10.1136/jim-2019-001195

    Article  PubMed  Google Scholar 

  477. Arraud N, Linares R, Tan S, Gounou C, Pasquet JM, Mornet S, Brisson AR (2014) Extracellular vesicles from blood plasma: determination of their morphology, size, phenotype and concentration. J Thromb Haemost 12(5):614–627. https://doi.org/10.1111/jth.12554

    Article  CAS  PubMed  Google Scholar 

  478. Lopez E, Srivastava AK, Burchfield J, Wang YW, Cardenas JC, Togarrati PP et al (2019) Platelet-derived- extracellular vesicles promote hemostasis and prevent the development of hemorrhagic shock. Sci Rep 9(1):17676. https://doi.org/10.1038/s41598-019-53724-y

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  479. Lee JH, Jung H, Song J, Choi ES, You G, Mok H (2020) Activated platelet-derived vesicles for efficient hemostatic activity. Macromol Biosci 20(3):e1900338. https://doi.org/10.1002/mabi.201900338

    Article  CAS  PubMed  Google Scholar 

  480. Wang Y, Zhang S, Luo L, Norstrom E, Braun OO, Morgelin M, Thorlacius H (2018) Platelet-derived microparticles regulates thrombin generation via phophatidylserine in abdominal sepsis. J Cell Physiol 233(2):1051–1060. https://doi.org/10.1002/jcp.25959

    Article  CAS  PubMed  Google Scholar 

  481. Kolonics F, Kajdacsi E, Farkas VJ, Veres DS, Khamari D, Kittel A et al (2021) Neutrophils produce proinflammatory or anti-inflammatory extracellular vesicles depending on the environmental conditions. J Leukoc Biol 109(4):793–806. https://doi.org/10.1002/JLB.3A0320-210R

    Article  CAS  PubMed  Google Scholar 

  482. Narauskaite D, Vydmantaite G, Rusteikaite J, Sampath R, Rudaityte A, Stasyte G et al (2021) Extracellular vesicles in skin wound healing. Pharmaceuticals (Basel). https://doi.org/10.3390/ph14080811

    Article  PubMed  Google Scholar 

  483. Shabbir A, Cox A, Rodriguez-Menocal L, Salgado M, Van Badiavas E (2015) Mesenchymal stem cell exosomes induce proliferation and migration of normal and chronic wound fibroblasts, and enhance angiogenesis in vitro. Stem Cells Dev 24(14):1635–1647. https://doi.org/10.1089/scd.2014.0316

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  484. Kocak P, Kala E, Gunes M, Unsal N, Yilmaz H, Metin B, Sahin F (2020) Edible plant-derived exosomes and their therapeutic applicatons. J Biomed Imag Bioeng 4:130–135

    Google Scholar 

  485. Sahin F, Kocak P, Gunes MY, Ozkan I, Yildirim E, Kala EY (2019) In vitro wound healing activity of wheat-derived nanovesicles. Appl Biochem Biotechnol 188(2):381–394. https://doi.org/10.1007/s12010-018-2913-1

    Article  CAS  PubMed  Google Scholar 

  486. Omar G, Abdallah L, Rahim A, Othman R, Barakat A (2017) Selected wild plants ethanol extracts bioactivity on the coagulation cascade. J Sci Res Reps 13(6):1–10

    Google Scholar 

  487. Nyeem MAB, Mannan MA (2018) Rubia cordifolia-phytochemical and Pharmacological evaluation of indigenous medicinal plant: a review. Int J Physiol Nutri Phys Edu 3(1):766–771

    Google Scholar 

  488. Chamara A, Thiripuranathar G (2020) Assessment of haemostatic activity of medicinal plants using in vitro methods: a concise review. IOSR J Pharm Biol Sci 15:26–34

    Google Scholar 

  489. Perut F, Roncuzzi L, Avnet S, Massa A, Zini N, Sabbadini S et al (2021) Strawberry-derived exosome-like nanoparticles prevent oxidative stress in human mesenchymal stromal cells. Biomolecules. https://doi.org/10.3390/biom11010087

    Article  PubMed  PubMed Central  Google Scholar 

  490. Baldini N, Torreggiani E, Roncuzzi L, Perut F, Zini N, Avnet S (2018) Exosome-like nanovesicles isolated from Citrus limon L. exert antioxidative effect. Curr Pharm Biotechnol 19(11):877–885. https://doi.org/10.2174/1389201019666181017115755

    Article  CAS  PubMed  Google Scholar 

  491. Ju S, Mu J, Dokland T, Zhuang X, Wang Q, Jiang H et al (2013) Grape exosome-like nanoparticles induce intestinal stem cells and protect mice from DSS-induced colitis. Mol Ther 21(7):1345–1357. https://doi.org/10.1038/mt.2013.64

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  492. Savci Y, Kirbas OK, Bozkurt BT, Abdik EA, Tasli PN, Sahin F, Abdik H (2021) Grapefruit-derived extracellular vesicles as a promising cell-free therapeutic tool for wound healing. Food Funct 12(11):5144–5156. https://doi.org/10.1039/d0fo02953j

    Article  CAS  PubMed  Google Scholar 

  493. Lingzhi Z, Meirong L, Xiaobing F (2020) Biological approaches for hypertrophic scars. Int Wound J 17(2):405–418. https://doi.org/10.1111/iwj.13286

    Article  PubMed  Google Scholar 

  494. Riau AK, Ong HS, Yam GHF, Mehta JS (2019) Sustained delivery system for stem cell-derived exosomes. Front Pharmacol 10:1368. https://doi.org/10.3389/fphar.2019.01368

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  495. Las Heras K, Igartua M, Santos-Vizcaino E, Hernandez RM (2020) Chronic wounds: current status, available strategies and emerging therapeutic solutions. J Control Releas 328:532–550. https://doi.org/10.1016/j.jconrel.2020.09.039

    Article  CAS  Google Scholar 

  496. Pan Z, Ye H, Wu D (2021) Recent advances on polymeric hydrogels as wound dressings. APL Bioeng 5(1):011504. https://doi.org/10.1063/5.0038364

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  497. Negut I, Dorcioman G, Grumezescu V (2020) Scaffolds for wound healing applications. Polymers (Basel). https://doi.org/10.3390/polym12092010

    Article  PubMed  Google Scholar 

  498. Huang CC, Kang M, Shirazi S, Lu Y, Cooper LF, Gajendrareddy P, Ravindran S (2021) 3D Encapsulation and tethering of functionally engineered extracellular vesicles to hydrogels. Acta Biomater 126:199–210. https://doi.org/10.1016/j.actbio.2021.03.030

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  499. Oliver TI, Mutluoglu M (2023) Diabetic foot ulcer. In: StatPearls. Treasure Island (FL) ineligible companies. Disclosure: Mesut Mutluoglu declares no relevant financial relationships with ineligible companies

  500. McDermott K, Fang M, Boulton AJM, Selvin E, Hicks CW (2023) Etiology, epidemiology, and disparities in the burden of diabetic foot ulcers. Diabetes Care 46(1):209–221. https://doi.org/10.2337/dci22-0043

    Article  PubMed  Google Scholar 

  501. Liu Y, Zhang X, Yang L, Zhou S, Li Y, Shen Y et al (2023) Proteomics and transcriptomics explore the effect of mixture of herbal extract on diabetic wound healing process. Phytomedicine 116:154892. https://doi.org/10.1016/j.phymed.2023.154892

    Article  CAS  PubMed  Google Scholar 

  502. Shreedhara CS, Vaidya VP, Vagdevi HM, Latha KP, Muralikrishna KS, Krupanidhi AM (2009) Screening of Bauhinia purpurea Linn. for analgesic and anti-inflammatory activities. Indian J Pharmacol 41(2):75–79. https://doi.org/10.4103/0253-7613.51345

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  503. Lu Z, Ye Y, Liu Y, Yang X, Ding Q, Wang Y et al (2021) Aqueous extract of paeoniae radix rubra prevents deep vein thrombosis by ameliorating inflammation through inhibiting GSK3beta activity. Phytomedicine 92:153767. https://doi.org/10.1016/j.phymed.2021.153767

    Article  CAS  PubMed  Google Scholar 

  504. Hu Y, Lei S, Yan Z, Hu Z, Guo J, Guo H et al (2021) Angelica dahurica regulated the polarization of macrophages and accelerated wound healing in diabetes: a network pharmacology study and in vivo experimental validation. Front Pharmacol 12:678713. https://doi.org/10.3389/fphar.2021.678713

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  505. Ponrasu T, Madhukumar KN, Ganeshkumar M, Iyappan K, Sangeethapriya V, Gayathri VS, Suguna L (2014) Efficacy of Acorus calamus on collagen maturation on full thickness cutaneous wounds in rats. Pharmacogn Mag 10(Suppl 2):S299-305. https://doi.org/10.4103/0973-1296.133283

    Article  PubMed  PubMed Central  Google Scholar 

  506. Xi Y, Zhao T, Shi M, Zhang X, Bao Y, Gao J et al (2023) Potential therapeutic mechanism of radix angelicae biseratae and dipsaci radix herb pair against osteoarthritis: based on network pharmacology and molecular docking. Evid Based Complement Alternat Med 2023:2140327. https://doi.org/10.1155/2023/2140327

    Article  PubMed  PubMed Central  Google Scholar 

  507. Weidinger S, Novak N (2016) Atopic dermatitis. Lancet 387(10023):1109–1122. https://doi.org/10.1016/S0140-6736(15)00149-X

    Article  PubMed  Google Scholar 

  508. Boccardi D, D’Auria E, Turati F, Sortino S, Riva E, Cerri A (2017) Disease severity and quality of life in children with atopic dermatitis: PO-SCORAD in clinical practice. Minerva Pediatr 69(5):373–380. https://doi.org/10.23736/S0026-4946.16.04294-8

    Article  PubMed  Google Scholar 

  509. Beltrani VS (1999) The clinical spectrum of atopic dermatitis. J Allergy Clin Immunol 104(3 Pt 2):S87-98. https://doi.org/10.1016/s0091-6749(99)70050-3

    Article  CAS  PubMed  Google Scholar 

  510. Bridgman AC, Block JK, Drucker AM (2018) The multidimensional burden of atopic dermatitis: an update. Ann Allergy Asthma Immunol 120(6):603–606. https://doi.org/10.1016/j.anai.2018.03.009

    Article  PubMed  Google Scholar 

  511. Erdogan S, Doganlar O, Doganlar ZB, Serttas R, Turkekul K, Dibirdik I, Bilir A (2016) The flavonoid apigenin reduces prostate cancer CD44(+) stem cell survival and migration through PI3K/Akt/NF-kappaB signaling. Life Sci 162:77–86. https://doi.org/10.1016/j.lfs.2016.08.019

    Article  CAS  PubMed  Google Scholar 

  512. Erdogan S, Doganlar O, Doganlar ZB, Turkekul K (2018) Naringin sensitizes human prostate cancer cells to paclitaxel therapy. Prostate Int 6(4):126–135. https://doi.org/10.1016/j.prnil.2017.11.001

    Article  PubMed  Google Scholar 

  513. Mlcek J, Jurikova T, Skrovankova S, Sochor J (2016) Quercetin and its anti-allergic immune response. Molecules. https://doi.org/10.3390/molecules21050623

    Article  PubMed  PubMed Central  Google Scholar 

  514. Weng Z, Zhang B, Asadi S, Sismanopoulos N, Butcher A, Fu X et al (2012) Quercetin is more effective than cromolyn in blocking human mast cell cytokine release and inhibits contact dermatitis and photosensitivity in humans. PLoS ONE 7(3):e33805. https://doi.org/10.1371/journal.pone.0033805

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  515. Park EJ, Kim JY, Jeong MS, Park KY, Park KH, Lee MW et al (2015) Effect of topical application of quercetin-3-O-(2’’-gallate)-alpha-l-rhamnopyranoside on atopic dermatitis in NC/Nga mice. J Dermatol Sci 77(3):166–172. https://doi.org/10.1016/j.jdermsci.2014.12.005

    Article  CAS  PubMed  Google Scholar 

  516. Beken B, Serttas R, Yazicioglu M, Turkekul K, Erdogan S (2020) Quercetin improves inflammation, oxidative stress, and impaired wound healing in atopic dermatitis model of human keratinocytes. Pediatr Allergy Immunol Pulmonol 33(2):69–79. https://doi.org/10.1089/ped.2019.1137

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

ZB helped in writing—original draft preparation, conceptualization, and methodology. RK contributed to supervision, reviewing and editing. All authors have read and approved the manuscript.

Corresponding author

Correspondence to Rafik Karaman.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Breijyeh, Z., Karaman, R. Antibacterial activity of medicinal plants and their role in wound healing. Futur J Pharm Sci 10, 68 (2024). https://doi.org/10.1186/s43094-024-00634-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43094-024-00634-0

Keywords