Next Article in Journal
Stimulated Parotid Saliva Is a Better Method for Depression Prediction
Next Article in Special Issue
A Comprehensive Review on Collagen Type I Development of Biomaterials for Tissue Engineering: From Biosynthesis to Bioscaffold
Previous Article in Journal
Comparing Intraperitoneal and Intravenous Personalized ErbB2CAR-T for the Treatment of Epithelial Ovarian Cancer
Previous Article in Special Issue
Ultra-Fine Polyethylene Hernia Meshes Improve Biocompatibility and Reduce Intraperitoneal Adhesions in IPOM Position in Animal Models
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Combination Strategies of Different Antimicrobials: An Efficient and Alternative Tool for Pathogen Inactivation

by
Nagaraj Basavegowda
and
Kwang-Hyun Baek
*
Department of Biotechnology, Yeungnam University, Gyeongsan 38451, Korea
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(9), 2219; https://doi.org/10.3390/biomedicines10092219
Submission received: 1 August 2022 / Revised: 2 September 2022 / Accepted: 4 September 2022 / Published: 7 September 2022
(This article belongs to the Special Issue Biomaterial Modifications and Improvement of Their Biocompatibility)

Abstract

:
Despite the discovery and development of an array of antimicrobial agents, multidrug resistance poses a major threat to public health and progressively increases mortality. Recently, several studies have focused on developing promising solutions to overcome these problems. This has led to the development of effective alternative methods of controlling antibiotic-resistant pathogens. The use of antimicrobial agents in combination can produce synergistic effects if each drug invades a different target or signaling pathway with a different mechanism of action. Therefore, drug combinations can achieve a higher probability and selectivity of therapeutic responses than single drugs. In this systematic review, we discuss the combined effects of different antimicrobial agents, such as plant extracts, essential oils, and nanomaterials. Furthermore, we review their synergistic interactions and antimicrobial activities with the mechanism of action, toxicity, and future directions of different antimicrobial agents in combination. Upon combination at an optimum synergistic ratio, two or more drugs can have a significantly enhanced therapeutic effect at lower concentrations. Hence, using drug combinations could be a new, simple, and effective alternative to solve the problem of antibiotic resistance and reduce susceptibility.

1. Introduction

The rapid emergence and spread of multidrug-resistant (MDR) bacteria has become a serious global public health threat [1]. Long-term exposure and increased use and abuse of antibiotics could result in bacterial tolerance, which renders them less effective or even ineffective, and the mechanism includes changing the targets of antibiotics [2]. MDR species are not only restricted to hospitals or healthcare environments; they are also found in humans, animals, plants, food, water, soil, and air. Moreover, they can be passed from person to person and between animals and persons. Antibiotic resistance is observed in various extracellular, intracellular, pathogenic, and nonpathogenic bacterial species. Among Gram-positive MDR species, Staphylococcus aureus, Streptococcus pneumoniae, Enterococcus faecium, and Enterococcus faecalis are the most common. Among Gram-negative strains, Pseudomonas aeruginosa, Escherichia coli, Klebsiella pneumoniae, and Acinetobacter baumannii are the most common MDR species [3]. However, methicillin-resistant Staphylococcus aureus causes pneumonia, bacteremia, soft tissue infections, and other fatal diseases [4]. Similarly, multiple antibiotic-resistant Acinetobacter spp. and Klebsiella spp. are the most commonly reported.
Recent studies suggest that biofilm-associated infections account for more than 65% of all infections, and antibiotics lack effectiveness against biofilm-associated bacteria [5]. Biofilms can shield bacteria from host defenses, disinfectants, antibiotics, and many antimicrobial agents. This leads to a reduced bacterial growth rate, decreased metabolic activity, and promotion of tolerance to antibiotics [6]. Moreover, the excessive use of antibiotics is often not tolerated by the host organism, whereas lower doses are ineffective. In addition, conventional antibiotics support antibiotic resistance in viable bacteria [7]. Pathogens growing in biofilms exhibit both adaptive resistance to all antimicrobial agents and the host immune system by 10- to 1000-fold compared to their free-living, planktonic counterparts [8]. Hence, there is urgent need to search for alternative, novel, efficient antimicrobial agents and more targeted treatment strategies to overcome antibiotic resistance. An alternative strategy currently in practice or under trials includes using different antimicrobial agents in combination to produce synergistic effects [9]. Combination therapy is an attractive and optional treatment because it represents potential adjuvant targets of non-overlapping signaling pathways and decreases the risk of developing cross-resistance [10].
Many plants have been used as sources of natural products to maintain good health, especially antimicrobial compounds [11]. Plants have evolved many alternative strategies against pathogens, which involve various phytochemicals, secondary metabolites, and other chemical compounds [12]. Bioactive compounds derived from plants, such as alkaloids, phenols, flavonoids, tannins, peptides, and other medicinally important compounds, are responsible for their antimicrobial ability against MDR pathogens [13]. Combining two or more plant extracts or their phytochemical components produces mutual antimicrobial enhancement, an unlimited pool of compounds, and the expansion or strengthening of their effects when combined as a multidrug [14]. Combinations of different drugs elicit several advantages over their use as individual moieties, including enhancing the effectiveness of other antimicrobial agents, reduction in dosage, fewer side effects, better synergistic effect, attack of multiple target sites, reduced risk, and exhibition of potent and rapid antibacterial effects against MDR pathogens [15]. The pharmacological effects of these combinations could be initiated by multiple mechanisms of action of herbal–herbal interactions.
Similarly, combining plant extracts or active phytochemicals with antibiotics improves their efficacy against resistant bacterial pathogens [16]. Synergism due to this combination helps minimize the minimum inhibitory concentrations (MICs) of these agents and reduces the economic cost and sensory impact [17]. Another strategic approach to combat MDR bacteria involves using essential oils (EOs) combined with conventional antibiotics or plant-derived phytochemicals. EOs have been widely used for their unique flavors; fragrances; and antibacterial, antioxidant, antifungal, anti-inflammatory, and anticarcinogenic properties [18]. Combining two or more EOs or their components or interactions between EOs and their components with antibiotics is a promising alternative strategy to increase their additive and synergistic antimicrobial effects. EOs and antibiotics, in combination, produce stronger bacterial inhibition compared to when they are individually administered because they target different pathways to create multifaceted effects against powerful bacterial defenses, consequently needing a decreased dose of each component [19]. The synergism between EOs and antibiotics may be attributed partly to the EO-induced permeabilization of the cell membrane, resulting in the immediate transport of antibiotics into the interior of the cell [20].
Antimicrobial nanomaterials represent another strategic approach to fighting MDR bacteria in clinical practice. Metal and metal oxide-based nanoparticles (NPs) have been widely investigated over the last decade, owing to their favorable chemical, physical, magnetic, electrical, thermal, optical, and biological properties [21]. Consequently, nanomaterials have emerged as new tools to combat deadly bacterial infections due to their specific features, such as size, shape, morphology, stability, and surface charge [22]. A combination of EOs and nanomaterials might establish functional materials with modified surfaces, improved inhibitory effects, and the ability to bind target microorganisms to achieve maximum synergistic performance [23]. Thus, combining nanomaterials with either EOs or plant extracts may improve their interaction with the bacterial cell membrane, thereby inducing the disruption, damage, and killing of bacteria [22]. This review highlights the effects of different antimicrobial agents and the synergistic effects of combinations of plant extracts, EOs, and nanomaterials. Furthermore, we discuss their antimicrobial activities, mechanisms of action, and future perspectives on using different combinations of antimicrobial agents.

2. Antibacterial Activities of Plant-Derived Compounds

Although different kinds of synthetic antimicrobial agents have been introduced to the market in many countries, natural medicine from plants might effectively treat certain diseases such as diarrhea, cold, labor pain, and dental diseases. Globally, approximately 60,000 plant species are used for medicinal purposes, of which approximately 28,000 are well-documented, and only 3000 are estimated to be traded internationally [24]. As a result, the search for herbal medicines with relevant biological activity has gained additional value as they are associated with fewer side effects and are much cheaper and affordable [25]. Plants usually produce two types of metabolites, primary and secondary, that can be found in extracts of their flowers, roots, leaves, bulbs, seeds, and bark (Figure 1). Primary metabolites are crucial for plant growth and development, whereas secondary metabolites are involved in plant defense, physiology, and environmental communication [26]. Secondary metabolites include many specialized and active compounds derived from primary metabolites. These compounds show promising results in controlling the development of resistance against bacterial pathogens, including MDR bacteria, and combating other bacterial infections. Plant secondary metabolites are classified into three categories on the basis of their biosynthetic origins: terpenoids, phenolics, and alkaloids [27].

2.1. Terpenoids

Terpenes are an extensive and diverse group of naturally occurring, highly enriched compounds of secondary plant metabolites. On the basis of the number of their isoprene structures or units, they are classified as monoterpenes, diterpenes, triterpenes, tetraterpenes, or sesquiterpenes. Terpenes are also called isoprenoids, and their derivatives that contain additional elements, such as oxygen, are usually termed terpenoids. Monoterpenes are the smallest terpenes, comprising two isoprene units. Monoterpenes contain volatile compounds found in EOs extracted from different flowers, fruits, and leaves and are commonly used in fragrances and aromatherapy. The antimicrobial properties of these compounds have been studied for two decades, and several studies have reported that thymol, carvacrol, eugenol, and menthol exhibit significant activity against many pathogens [28]. Geraniol and thymol have shown the most activity against Enterobacter species and S. aureus and E. coli, respectively [29,30]. Diterpenes are naturally occurring chemical compounds that contain active groups such as vitamin A. Phytol is an acyclic diterpene alcohol that acts as an antitumor, cytotoxic, and anti-inflammatory agent. Diterpenes also inhibit the growth of Staphylococcus aureus, Pseudomonas aeruginosa, Vibrio cholerae, and Candida spp. [31]. Triterpenes contain six isoprene units derived from mevalonic acid and have been shown to inhibit the growth of Mycobacterium tuberculosis. The combination of rifampicin and oleanolic acid has shown synergistic antibacterial effects against some pathogens [32]. Tetraterpenes are also known as carotenoids because beta-carotene is a yellow pigment in carrots. Similarly, yellow, orange, and red organic pigments are produced by plants, and these substances have effective antifungal and antibacterial properties [33]. Sesquiterpenes are the most diverse group of terpenoids, consisting of three units of isoprene with a lower vapor pressure than monoterpenes because of their high molecular weight. Farnesol, a natural sesquiterpene, demonstrated antibacterial activity against S. aureus and S. epidermidis [34].

2.2. Phenolics

Phenols are the simplest bioactive phytochemicals. They are monomeric components of polyphenols and acids with a single substituted phenolic ring and are typically found in plant tissues such as melanin and lignin. The components catechol, orcinol, tarragon, pyrogallol, phloroglucinol, pyrocatechol, resorcinol, and thyme are effective against viruses, bacteria, and fungi [35]. Both catechol and pyrogallol are hydroxylated phenolic compounds that are toxic against microorganisms; catechol has two hydroxyl groups, while pyrogallol has three. The microbial toxicity of phenolic compounds depends mainly on the number and position of their hydroxyl groups, as hydroxylation increases toxicity [36]. The presence of two or more hydroxyl groups located at ortho, para, or meta positions to each other is the key factor for their antimicrobial activity. The presence of a hydroxyl group at the meta position of thymol makes it a more effective antibacterial agent than carvacrol, which has a similar structure, whose hydroxyl group is in the ortho position.

2.3. Alkaloids

Alkaloids are cyclic-nitrogen-containing organic compounds that have various chemical structures. More than 18,000 alkaloids have been discovered and studied phytochemically from different sources. Alkaloids are grouped into several classes, as natural, semi-synthetic, or synthetic, on the basis of their heterocyclic ring systems and biosynthetic precursors [37]. Alkaloids have various pharmacological activities, including antitumor, antihyperglycemic, anti-allergic, antidiabetic, antihyperlipidemic, and antibacterial. Piperine, berberine, quinolone, reserpine, sanguinarine, tomatidine, chanoclavine, conessine, and squalamine are the most important alkaloids with potent antibacterial activity. Piperine isolated from Piper nigrum and Piper longum inhibited the growth of mutant S. aureus when co-administered with ciprofloxacin [38]. A list of plants whose parts were reported to have antimicrobial activity against various pathogens, as well as their corresponding mechanisms of action, are summarized in Table 1.

3. Antimicrobial Efficacy of EOs

EOs are aromatic, lipophilic, and complex mixtures of volatile secondary metabolites that are mainly obtained from different parts of plants, such as leaves, herbs, flowers, buds, fruits, twigs, wood, bark, roots, and seeds [73]. EOs are extracted using hydrodistillation or steam distillation. EOs are lighter than water, with a strong flavor and odor reminiscent of their plant origin. The chemical composition of EOs is highly complex, with the main components being flavonoids, flavones, flavonols, phenols, polyphenols, tannins, alkaloids, quinones, coumarins, terpenoids, polypeptides, and lectins [22]. These compounds show potential pharmacological activities such as hepatoprotective, anti-inflammatory, antioxidant, anticancer, antiseptic, insecticidal, anti-parasitic, anti-allergic, antiviral, and antimicrobial properties [74]. Essential-oil-based products are in high demand in aromatherapy; as flavor-enhancers in food, beverages, cosmetics, perfumes, soaps, plastics, and resins; and in the pharmaceutical industries [75]. EOs have more than 50 components; however, only two or three of them are the major components present in high proportions. The other minor components are present in low amounts.
The amount of the different components of EOs varies with the parts and species of plants, as they are chemically derived from compounds and their derivatives [76]. The major constituents of EOs are terpenes and terpenoids, while other important compounds include aromatic and aliphatic constituents. The volatile components of EOs include a variety of chemicals such as alcohols (such as menthol, borneol, nerol, and linalool), acids (such as geranic acid and benzoic acid), aldehydes (such as citral), esters (such as linalyl acetate, citronellyl acetate, and menthyl), ketones (such as carvone, camphor, and pulegone), hydrocarbons (such as α-pinene, α-terpinene, myrcene, camphene, and p-cimene), ketones (such as camphor, pulegone, and carvone), phenols (such as carvacrol and thymol), lactones (such as bergapten), and peroxides (such as ascaridole), all of which play major roles in the composition of EOs (Figure 2) [77]. EOs show strong antibacterial activity against various pathogenic bacteria, including MDR pathogens, by penetrating the membrane of bacterial cells and disrupting their cellular structure. The antibacterial effectiveness of EOs differs across plant species and target bacteria depending on their cell wall structure (Gram-positive or Gram-negative). The association of some major constituents of EOs, such as eugenol, thymol, carvacrol, carvone, p-cymene, terpinene-4-ol, and cinnamic aldehyde, which easily penetrate and split in the lipid membrane, could disrupt the cell membrane; prevent cellular respiration; and lead to the loss of cell membrane integrity, removal of cellular contents, and finally cell death [78].
EOs of Thymus serrulatus and Thymus schimperi were shown to possess strong antibacterial activity against Lactobacillus and S. mutans, with higher contents of thymol and carvacrol compounds reported to be the cause of this inhibition [79]. Similarly, EOs have been isolated from various parts of Eugenia caryophylata, such as the buds, leaves, and stems, with the main components being eugenol, β-caryophyllene, and eugenyl acetate. These EOs are effective against S. aureus, E. coli, B. subtilis, and S. typhimurium [80]. Likewise, tea tree EO has been reported to cause changes in the membrane permeability and mycelial morphology of Monilinia fructicola [81]. Furthermore, a recent study of lavender EO against A. hydrophila, A. caviae, A. dhakensis, C. freundii, P. mirabilis, and S. enterica showed the presence of the major compounds linalool and linalyl acetate [82]. In another study, winter savory EO exhibited the strongest inhibitory effect against clinical oral isolates of Candida spp., with thymol as the major compound [83]. It has been reported that among the commercially available EOs, such as anise, cinnamon, clove, cumin, laurel, Mexican lime, and Mexican oregano, oregano EO has the highest antibacterial activity against S. typhimurium and E. coli, with thymol as its major compound [84]. Some of the most important and active EOs, their major constituents, mechanisms of action, and antimicrobial potential against pathogenic microbes are summarized in Table 2.

4. Antimicrobial Nanomaterials

With the emergence of bacterial resistance and biofilm-associated infections, clinical research is needed to develop novel, effective, long-term antibacterial and biofilm-preventative agents. Metals have been extensively studied among the most promising novel antimicrobial agents [111]. Recently, metal-based nanomaterials have become the most extensively and rapidly emerging materials in the field of medicine. Different types of metallic NPs have demonstrated strong antibacterial activity in many recent studies [112]. Generally, NPs have fascinating characteristics, such as a high surface area-to-volume ratio, size, shape, and surface activity, and exhibit superior electrical, catalytic, and optical properties. Due to their unique properties, NPs have a more well-developed surface than their microscale counterparts, affecting their antimicrobial efficiency and effectiveness [113]. Similar to antibiotics, metals can selectively inhibit metabolic pathways by interacting with bactericidal activity and ultimately kill MDR bacteria [112]; however, cells deviate from metal transport systems and metalloproteins [114]. Hence, NPs showed noticeable antimicrobial activity against both Gram-negative and Gram-positive pathogens such as E. faecalis, B. subtilis, S. epidermidis, multidrug-resistant S. aureus, and E. coli strains.
Metal NPs such as Ag, Au, Cu, Zn, Ti, Ga, Al, and Pt [115] and metal oxide NPs such as CuO, MgO, ZnO, TiO2, NiO2, SiO2, and Fe3O4 are known to display various antimicrobial properties, which have been known and applied for decades [116]. In addition, graphene oxide (GO) and carbon nanotubes (CNTs), such as single-walled carbon nanotubes (SWCNTs) and multi-walled carbon nanotubes (MWCNTs), are also excellent candidates due to their antimicrobial activities (Figure 3). Recently, metal–organic frameworks (MOF) and metal sulfide nanomaterials, such as FeS-, Ags-, ZnS-, and CdS-MOFs and Zn-, Cu-, and Mn-based MOFs, have also been proven to have antibacterial activities [117]. Multimetallic NPs, particularly NPs formed by at least two metals, such as bimetallic, trimetallic, and quadrametallic NPs, display rich optical, electronic, and magnetic properties. The properties of multimetallic NPs, including size, shape, surface area, and zeta potential, enhance their interaction with bacterial cell membranes. They could disrupt cell membranes, produce reactive oxygen species (ROS), damage the DNA, induce protein dysfunction, and may be potentiated by the host immune system [23].
Metallic biopolymer-based nanocomposite systems are well-known candidates as antimicrobial nanomaterials. In particular, cationic chitosan-based NPs bind to anionic cell membranes, resulting in alterations to the cell membrane, leakage of intracellular compounds, and eventually cell death [118]. Antimicrobial peptides (AMPs) have attracted great interest because of their high biocompatibility and low probability of inducing bacterial resistance. AMP-conjugated nanomaterials can hinder the growth of pathogens and kill bacteria on the basis of the inherent action of typical combination strategies [119].
AgNPs are considered the most common antimicrobial agents that can destroy a wide range of Gram-negative and Gram-positive bacteria. Ag ions combine with disulfide or sulfhydryl groups of enzymes, disrupt normal metabolic processes, and ultimately lead to cell death [120]. The bactericidal efficacy of Au NPs might have a greater chance of penetrating the bacterial cell wall by generating holes, leading to increased permeability and higher oxidative stress within the cytoplasm. Similarly, ZnO NPs displayed vigorous antimicrobial activity by releasing Zn2+ ions and generating ROS, owing to their electrostatic interaction and internalization. In contrast, smaller ZnO NPs increased the interaction and abrasiveness of the bacterial cell wall [121]. Cu and CuO NPs showed excellent antimicrobial activity against different strains of bacteria by releasing Cu2+ ions and stimulating ROS production [122]. Various studies have revealed the visible-light-induced antibacterial properties of Fe-, Cu-, Ni-, and Ag-doped TiO2 NPs against E. coli and S. aureus [123,124]; however, TiO2 NPs adversely affect human cells and tissues, so their use remains limited. SiO2 NPs, especially mesoporous NPs, have attracted considerable attention because their properties, such as size, matrix, and surface functions, which can be tuned to improve their interaction with and penetration of biofilm-producing bacteria [125].
Compared to monometallic NPs, multimetallic NPs, such as bi-, tri-, and quadrametallic NPs, have gained great importance and interest due to their unique physical, chemical, electrical, optical, and catalytic properties and applications in different fields [126]. Multimetallic NPs can be altered or tuned by controlling their structure, morphology, and chemical composition to achieve strong synergistic interactions and performance [127]. When at least two metals are formed as NPs, combinatorial approaches, such as structural changes, deduction of the lattice parameters, and total electronic charge shift improvements, are expected [128]. Recently, bimetallic Ag/Cu and Cu/Zn [129], and trimetallic Cu/Cr/Ni [130] and CuO/NiO/ZnO [131] NPs have exhibited remarkably improved antimicrobial performance compared to monometallic NPs. Recent studies on the antimicrobial activities of metal and metal oxides, including mono-, bi-, and trimetallic NPs, against various bacterial strains and their respective mechanisms of action are shown in Table 3.
Core–shell quantum dots (CSQDs) are a new type of fluorescent antibacterial nanomaterial with unique physical and chemical properties. Owing to their high electron transfer, CSQDs produce a large number of free electrons and holes that accumulate ROS inside the cell, inhibiting their respiration and replication [167]. CSQDs exert antimicrobial effects by destroying cell walls, binding with genetic material, and inhibiting energy production. The antimicrobial activities of some CSQDs and their mechanisms of action are listed in Table 4.
Additionally, the antibacterial properties of graphene involve both chemical and physical modes of action. The chemical action is associated with oxidative stress generated by charge transfer and ROS, while the physical action is induced by the direct contact of graphene with bacterial membranes [175]. Similarly, CNTs are more effective and cost-efficient, exhibiting strong antimicrobial properties owing to their remarkable structure. This mechanism is based on the interaction of CNTs with microorganisms and the disruption of their metabolic processes, cellular membranes, and morphology. Table 5 summarizes the various reported antimicrobial activities of graphene and CNTs.
Dendrimers are macromolecules with highly branched tree-like dendritic structures, narrow sizes, relatively large molecular masses, and well-defined globular structures [183]. Dendrimers peripherally cationic and highly water soluble due to numerous peripheral hydrophilic groups compatible with water [184]. Dendrimers can incorporate biologically active agents in the interior or periphery; therefore, they serve as carriers of biologically active agents [185]. Antimicrobial polymers or their composites can prevent or suppress the growth of microbes on their surfaces or in the environment. Positively charged polymer surface groups are attracted to negatively charged cell membranes, leading to cell membrane damage and cell death [186]. A few recent publications on the antimicrobial activities of dendrimers and polymer nanocomposites are summarized in Table 6.

5. Synergistic Antimicrobial Activity of Plant Extracts, EOs, and Nanomaterials

A synergistic effect is a process in which chemical substances or biological structures interact or combine to create an effect greater than the sum of the effects of the individual components. Synergy is the concept wherein the performance of two or more antimicrobial agents is combined and the effects of such mixtures are greater than those of the separate or individual components, enhancing solubility. In recent years, the synergistic combination of different antimicrobials and plant extracts has been considered a unique strategy to increase the spectrum of antimicrobial activity of these substances and prevent the development of resistant strains [194]. Plant extracts, EOs, and nanomaterials are commonly used as antimicrobial agents for the treatment of many infectious diseases. However, these antimicrobials are not very effective against acute infections because they lack a standardized and clinically applicable pharmaceutical form. Consequently, various antibiotics have been discovered as synthetic antimicrobials; however, these drugs are highly toxic and have poor tolerability, so bacteria develop resistance against them. Hence, a possible approach to improve and enhance antibacterial activity is to use combinations of different antimicrobials. These combinatorial approaches can be used alone or with other antimicrobials against a wide range of pathogens [195]. Compared to the individual substances or components, multicomponent antimicrobials display increased antimicrobial activity; therefore, other molecules present in the antimicrobial agents could control the function of the main components and improve their synergistic effects. Moreover, combining different antimicrobials offers many advantages, including a reduction in dosage, fewer side effects, decreased toxicity, extensive antibacterial action, and the ability to attack multiple target sites with increased efficacy [196]. Some combinations of antimicrobial agents, such as plant extracts, EOs, antibiotics, and NPs, are summarized in Table 7.
Combinations of antimicrobial agents, such as EOs/EOs, plant extract/plant extract, and NPs/NPs, already have confirmed antimicrobial activities [203]. Ncube et al. evaluated the bulb and leaf extracts of three medicinal plants, independently and in combination, against S. aureus. Their results showed the strongest synergistic effect compared with the effects observed with individual extracts [12]. Similarly, a combination of Bulbine frutescens and Vernonia lasiopus plant extracts showed improved antimicrobial activity against E. coli [204]. Obuekwe et al. found the largest zones of inhibition against S. aureus using a combination of Ocimum gratissimum and Ficus exasperate, and Bryophyllum pinnatum and Ocimum gratissimum against E. coli [205]. Recently, EO–EO associations showed a synergistic effect against vancomycin-resistant enterococci (VRE), methicillin-resistant S. aureus (MRSA), and extended-spectrum β-lactamase (ESBL)-producing Escherichia coli [195]. A mixture of R. abyssinicus and D. penninervium EOs showed strong synergistic effects against MRSA and P. aeruginosa [206].
The synergistic antibacterial activities of cumin, cardamom, and dill weed EOs against C. coli and C. jejuni have been reported [91]. In a previous study, a combination of cinnamon and clove EOs showed synergistic antibacterial activity against foodborne S. aureus, L. monocytogenes, S. typhimurium, and P. aeruginosa [207]. Garza-Cervantes et al. examined the synergistic antimicrobial activities of silver in combination with other transition metals (Zn, Co, Cd, Ni, and Cu). Their results exhibited synergism since the antimicrobial effects of the combinations against E. coli and B. subtilis increased up to eightfold when compared to the individual metals [208]. Similarly, β-lactam is the most common bactericidal agent recommended for the treatment of several infectious diseases. However, the increasing emergence of β-lactam resistance due to β-lactamase enzyme production is one of the most serious public health threats. Hence, current clinical trials suggest the use of proper combinations of β-lactam and β-lactamase inhibitors [209]. β-Lactam inhibitors are associated with β-lactam antibiotics because they are hydrolyzed by β-lactamases, and their main objective is to protect the associated antibiotics. β-Lactam inhibitors prevent the hydrolytic action of β-lactam antibiotics by binding to the active site of β-lactamase enzymes [210].

6. Currently Available Conventional Antibiotics

Currently, there are 32 antibacterial agents in clinical development phases 1–3 targeting WHO priority pathogens, 12 of which have activity against Gram-negative pathogens. Since 2018, several new products have entered phase 1 trials, and two new recombinant topoisomerase inhibitors, zoliflodacin and gepotidacin, have moved from phase 2 to phase 3 trials. Similarly, lefamulin and relebactum moved from phase 3 trials to FDA approval, and omadacycline and eravacycline have moved from NDA submission to gaining FDA approval. An additional product of β-lactam (cefideocol) is more stable against a variety of β-lactamases and has activity against all three critical priority pathogens [211]. β-Lactams are well-established and widely used antibiotics, including penicillins, cephalosporins, carbapenems, and monobactams. β-Lactams interrupt cell wall formation and subsequently disrupt peptidoglycan biosynthesis. However, the emergence of bacteria that produce β-lactamase enzymes that hydrolyze β-lactam antibiotics has rendered many antimicrobial agents ineffective. β-Lactamases are a diverse class of enzymes produced by bacteria that break the β-lactam ring open, inactivating the antibiotic. Table 8 summarizes several mechanisms of resistance to different target drugs with different modes of action.

7. Antibacterial Mechanisms of Plant Extracts, EOs, and Nanomaterials

The antimicrobial mechanism of plant extracts is more strongly correlated with the levels of their constituent phenolic compounds, especially flavonoids and their derivatives. The interaction of polyphenols with lipid bilayers can trigger and disrupt plasma membrane function, change its permeability, and form small pores. These could lead to the leakage of cell components, altering the surface electrical charge potential and bacterial polarity, modifying membrane fluidity, delocalizing membrane lipids and proteins, as well as other phenomena responsible for antibacterial activity. These alterations can cause severe damage to the bacteria by partitioning their membrane and cell wall, interrupting DNA and RNA synthesis and function, disrupting normal cell communication, and preventing biofilm formation [217]. Some studies have shown that secondary metabolites of plant extracts, such as alkaloids, terpenoids, and phenolic compounds, interfere with enzymes and proteins of the microbial cell membrane and inhibit enzymes necessary for amino acid biosynthesis. Other studies have ascribed the inhibitory effect of these plant extracts to their hydrophobicity since they can react with proteins and mitochondria, disturbing their structures and altering their permeability [218].
The antibacterial mechanism of EOs does not comprise a single action; however, various biochemical and structural mechanisms are simultaneously engaged at multiple sites in the bacterial cell membrane and cytoplasm. The primary antimicrobial effects of EOs are correlated with an increase in membrane permeability and plasma membrane disruption. The bioactive components found in EOs, such as thymol, eugenol, and carvacrol, might attach to the cell surface and penetrate the target region, especially the phospholipid bilayer of the cell membrane [76]. It has been shown that EO accumulation can disrupt membrane integrity and membrane proteins, increase membrane permeability, induce the leakage of cellular contents, and reduce the intracellular ATP pool. This consequently leads to cytoplasmic coagulation and the denaturation of enzymes, inhibiting the synthesis of DNA and proteins required for bacterial growth. Furthermore, the sustained loss of metabolites and ions due to EO administration can further disturb bacterial metabolic processes, leading to cell death [219].
The bactericidal mechanism of nanomaterials mainly depends on the type of NPs used, such as metals, metal oxides, and other nanocomposites. NPs bind to the bacterial cell wall, form membrane-penetrating pores, and release metal ions due to deposition. The adhesion of nanomaterials and microbial cells can be achieved through electrostatic attractions, hydrophobic interactions, Van der Waals forces, and receptor–ligand interactions, leading to cell wall destruction [220]. Furthermore, the positively charged surfaces of nanomaterials could promote the attachment of negatively charged bacterial surfaces, which may exert and strengthen their bactericidal effect. In addition, the generation of free radicals and ROS can destroy the cell membrane, disrupting the antioxidant defense system and causing mechanical damage to the cell membrane. Thereafter, nanomaterials interact with important cellular organelles such as DNA, enzymes, ribosomes, and lysosomes, resulting in oxidative stress, changes in membrane permeability, heterogeneous alterations, electrolyte balance disorders, changes in gene expression, and protein deactivation [220]. Possible modes of action when combining plant extracts, EOs, and nanomaterials are illustrated in Figure 4.

8. Concluding Remarks and Prospects for Future Research

Although the pharmaceutical industry has introduced many new antibiotics, the increasing prevalence of serious clinical complications related to MDR pathogens is a great challenge for researchers, clinicians, and the pharmacological industries. Therefore, searching for the most promising novel antibacterial agents with alternative strategies to combat bacterial infections is an ideal solution to treat infections that threaten human health. The ultimate goal is to offer appropriate and effective antimicrobial drugs to infected patients. The use of combination therapies is an effective way to improve the treatment of many health conditions, prevent the development of MDR pathogens, and reduce the treatment duration. Combination therapies can target and interact in multiple pathways and exhibit greater therapeutic efficacy than single antimicrobial-agent-based therapies. Moreover, they have recently been regarded as promising, cost-effective, and potentially able to mitigate side effects to the body with lower drug concentrations. There is plenty of evidence to support the effectiveness of medicinal plants in the treatment of infectious diseases. However, very few studies have reported the synergistic effects of plant extracts and phytochemical combinations of herbal remedies.
Combinations of different plant species or mixtures of different phytochemicals have been shown to exert potential antimicrobial activity against several human pathogens with diverse mechanisms of action. The curative effects of plant extract combinations showed both intrinsic and antibiotic-resistance-modifying activities. Some plant extracts are not effective as antibiotic agents alone; however, when combined with other antibacterial plant extracts, their bioavailability and antibacterial activity are enhanced. Similarly, there is much evidence suggesting the antimicrobial effects of individual EOs against different pathogens in vitro, but very few studies have reported the effects of EO combinations. Compared to a single EO or its chemical constituents, combining more than two EOs can increase and improve their antimicrobial activities due to an increased diversity of components and multiple sites of action. Furthermore, many studies have reported the potential antimicrobial activities of different nanomaterials against MDR pathogens. However, only a small percentage of studies have discussed the synergistic effects of multimetallic NPs, such as bi-, tri-, and quadrametallic and metal oxide nanocomposites. The synergistic effects of these multimetallic NPs have attracted considerable attention, owing to their diverse and tunable physicochemical properties and favorable catalytic properties compared with monometallic NPs.
The present review describes the synergistic effects of plant extracts/plant extracts, EOs/EOs, and nanomaterials/nanomaterials as efficient alternative strategies for pathogen inactivation or infectious diseases. However, further research is needed to assess the synergistic effects of combinations of plant extracts/EOs, plant extracts/nanomaterials, and nanomaterials/EOs to achieve better antimicrobial results. In addition, more effort is required to investigate the synergistic effects of combinations of plant extracts/EOs/nanomaterials. Consequently, β-lactam/β-lactamase inhibitor combinations are more effective on the different bacterial species. Combinations comprising three different antimicrobial agents might have enhanced synergistic antimicrobial activity compared to the effects of a combination of only two antimicrobial agents. Furthermore, the concentrations of the plant extracts, EOs, nanomaterial types, proper dosage, and choice of materials are essential to maximizing their therapeutic benefit. It is also important to consider environmental issues, health and safety concerns, risk assessments, potential toxicity, and hazards before considering them novel antimicrobial agents. These new, modern, and creative therapeutic strategies may exert a critical synergistic effect and serve as alternatives to conventional antibiotics for controlling the spread of pathogens. Finally, we believe this review provides necessary information about the combination of different antimicrobial agents to produce synergistic effects for the control and treatment of a wide range of pathogenic infections and may also play an essential role in many medical applications.

Author Contributions

Data collection, N.B. and K.-H.B.; writing—original draft preparation, N.B.; writing—review and editing, K.-H.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by PJ015726, Rural Development Administration, Republic of Korea.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This work was carried out with the support of the Cooperative Research Pro-gram for Agriculture Science and Technology Development (Project No. PJ015726), RDA, Republic of Korea.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Pang, X.; Xiao, Q.; Cheng, Y.; Ren, E.; Lian, L.; Zhang, Y.; Gao, H.; Wang, X.; Leung, W.; Chen, X. Bacteria-responsive nanoliposomes as smart sonotheranostics for multidrug resistant bacterial infections. ACS Nano 2019, 13, 2427–2438. [Google Scholar] [CrossRef] [PubMed]
  2. Baym, M.; Stone, L.K.; Kishony, R. Multidrug evolutionary strategies to reverse antibiotic resistance. Science 2016, 351, aad3292. [Google Scholar] [CrossRef]
  3. Sun, Y.; Ye, J.; Hou, Y.; Chen, H.; Cao, J.; Zhou, T. Predation efficacy of Bdellovibrio bacteriovorus on multidrug-resistant clinical pathogens and their corresponding biofilms. Jpn. J. Infect. Dis. 2017, 70, 485–489. [Google Scholar] [CrossRef] [PubMed]
  4. Dong, P.; Mohammad, H.; Hui, J.; Leanse, L.G.; Li, J.; Liang, L.; Dai, T.; Seleem, M.N.; Cheng, J. Photolysis of Staphyloxanthin in methicillin-resistant Staphylococcus aureus potentiates killing by reactive oxygen species. Adv. Sci. 2019, 6, 1900030. [Google Scholar] [CrossRef]
  5. Jacqueline, C.; Caillon, J. Impact of bacterial biofilm on the treatment of prosthetic joint infections. J. Antimicrob. Chemother. 2014, 69, i37–i40. [Google Scholar] [CrossRef]
  6. Dincer, S.; Uslu, F.M.; Delik, A. Antibiotic resistance in biofilm. In Bacterial Biofilms; IntechOpen: London, UK, 2020; Available online: https://www.intechopen.com/chapters/72109 (accessed on 30 July 2022). [CrossRef]
  7. Beyth, N.; Houri-Haddad, Y.; Domb, A.; Khan, W.; Hazan, R. Alternative Antimicrobial Approach: Nano-Antimicrobial Materials. Evid.-Based Complement. Altern. Med. 2015, 2015, 246012. [Google Scholar] [CrossRef]
  8. Dostert, M.; Belanger, C.R.; Hancock, R.E.W. Design and assessment of anti-biofilm peptides: Steps toward clinical application. J. Innate Immun. 2019, 11, 193–204. [Google Scholar] [CrossRef] [PubMed]
  9. Kaur, I. Novel strategies to combat antimicrobial resistance. J. Infect. Dis. Ther. 2016, 4, 292. [Google Scholar] [CrossRef]
  10. Bozic, I.; Reiter, J.G.; Allen, B.; Antal, T.; Chatterjee, K.; Shah, P.; Moon, Y.S.; Yaqubie, A.; Kelly, N.; Le, D.T. Evolutionary dynamics of cancer in response to targeted combination therapy. Elife 2013, 2, e00747. [Google Scholar] [CrossRef] [PubMed]
  11. Nascimento, G.G.F.; Locatelli, J.; Freitas, P.C.; Silva, G.L. Antibacterial activity of plant extracts and phytochemicals on antibiotic-resistant bacteria. Braz. J. Microbiol. 2000, 31, 247–256. [Google Scholar] [CrossRef]
  12. Ncube, B.; Finnie, J.F.; Van Staden, J. In vitro antimicrobial synergism within plant extract combinations from three South African medicinal bulbs. J. Ethnopharmacol. 2012, 139, 81–89. [Google Scholar] [CrossRef] [PubMed]
  13. Aggarwal, B.; Lamba, H.S.; Ajeet, P.S. Various pharmacological aspects of Cocos nucifera—A review. Am. J. Pharmacol. Sci. 2017, 5, 25–30. [Google Scholar]
  14. Olajuyigbe, O.O.; Afolayan, A.J. Evaluation of combination effects of ethanolic extract of Ziziphus mucronata Willd. subsp. mucronata Willd. and antibiotics against clinically important bacteria. Sci. World J. 2013, 2013, 769594. [Google Scholar] [CrossRef]
  15. León-Buitimea, A.; Garza-Cárdenas, C.R.; Garza-Cervantes, J.A.; Lerma-Escalera, J.A.; Morones-Ramírez, J.R. The demand for new antibiotics: Antimicrobial peptides, nanoparticles, and combinatorial therapies as future strategies in antibacterial agent design. Front. Microbiol. 2020, 11, 1669. [Google Scholar] [CrossRef]
  16. Cheesman, M.J.; Ilanko, A.; Blonk, B.; Cock, I.E. Developing new antimicrobial therapies: Are synergistic combinations of plant extracts/compounds with conventional antibiotics the solution? Pharmacogn. Rev. 2017, 11, 57. [Google Scholar]
  17. Reda, F.M.; El-Zawahry, Y.A.; Omar, A.R. Synergistic effect of combined antibiotic and methanol extract of Eucalyptus camaldulensis leaf against Staphylococcus aureus and Pseudomonas aeruginosa. Int. J. Appl. Sci. Biotechnol. 2017, 5, 486–497. [Google Scholar] [CrossRef]
  18. Basavegowda, N.; Baek, K.-H. Synergistic Antioxidant and Antibacterial Advantages of Essential Oils for Food Packaging Applications. Biomolecules 2021, 11, 1267. [Google Scholar] [CrossRef]
  19. Horváth, G.; Bencsik, T.; Ács, K.; Kocsis, B. Sensitivity of ESBL-Producing Gram-Negative Bacteria to Essential Oils, Plant Extracts, and Their Isolated Compounds; Academic Press: Amsterdam, The Netherlands, 2016; pp. 239–269. [Google Scholar]
  20. Aleksic, V.; Mimica-Dukic, N.; Simin, N.; Nedeljkovic, N.S.; Knezevic, P. Synergistic effect of Myrtus communis L. essential oils and conventional antibiotics against multi-drug resistant Acinetobacter baumannii wound isolates. Phytomedicine 2014, 21, 1666–1674. [Google Scholar] [CrossRef]
  21. Dikshit, P.K.; Kumar, J.; Das, A.K.; Sadhu, S.; Sharma, S.; Singh, S.; Gupta, P.K.; Kim, B.S. Green synthesis of metallic nanoparticles: Applications and limitations. Catalysts 2021, 11, 902. [Google Scholar] [CrossRef]
  22. Basavegowda, N.; Patra, J.K.; Baek, K.-H. Essential oils and mono/Bi/tri-metallic nanocomposites as alternative sources of antimicrobial agents to combat multidrug-resistant pathogenic microorganisms: An overview. Molecules 2020, 25, 1058. [Google Scholar] [CrossRef]
  23. Basavegowda, N.; Baek, K.-H. Multimetallic nanoparticles as alternative antimicrobial agents: Challenges and perspectives. Molecules 2021, 26, 912. [Google Scholar] [CrossRef]
  24. Jenkins, M.; Timoshyna, A.; Cornthwaite, M. Wild at Home: Exploring the global harvest, trade and use of wild plant ingredients. Retrieved Jan. 2018, 26, 2019. [Google Scholar]
  25. Manzo, L.M.; Moussa, I.; Ikhiri, K.; Yu, L. Toxicity studies of Acacia nilotica (L.): A review of the published scientific literature. J. Herbmed Pharmacol. 2019, 8, 163–172. [Google Scholar] [CrossRef]
  26. Pott, D.M.; Osorio, S.; Vallarino, J.G. From central to specialized metabolism: An overview of some secondary compounds derived from the primary metabolism for their role in conferring nutritional and organoleptic characteristics to fruit. Front. Plant Sci. 2019, 10, 835. [Google Scholar] [CrossRef]
  27. Eljounaidi, K.; Lichman, B.R. Nature’s chemists: The discovery and engineering of phytochemical biosynthesis. Front. Chem. 2020, 8, 1041. [Google Scholar] [CrossRef]
  28. Kusakizako, T.; Miyauchi, H.; Ishitani, R.; Nureki, O. Structural biology of the multidrug and toxic compound extrusion superfamily transporters. Biochim. Biophys. Acta BBA-Biomembr. 2020, 1862, 183154. [Google Scholar] [CrossRef]
  29. Dreier, J.; Ruggerone, P. Interaction of antibacterial compounds with RND efflux pumps in Pseudomonas aeruginosa. Front. Microbiol. 2015, 6, 660. [Google Scholar] [CrossRef] [PubMed]
  30. Zacchino, S.A.; Butassi, E.; Cordisco, E.; Svetaz, L.A. Hybrid combinations containing natural products and antimicrobial drugs that interfere with bacterial and fungal biofilms. Phytomedicine 2017, 37, 14–26. [Google Scholar] [CrossRef]
  31. Batista, O.; Duarte, A.; Nascimento, J.; Simões, M.F.; de la Torre, M.C.; Rodríguez, B. Structure and antimicrobial activity of diterpenes from the roots of Plectranthus hereroensis. J. Nat. Prod. 1994, 57, 858–861. [Google Scholar] [CrossRef]
  32. Cox-Georgian, D.; Ramadoss, N.; Dona, C.; Basu, C. Therapeutic and medicinal uses of terpenes. In Medicinal Plants; Springer: Berlin/Heidelberg, Germany, 2019; pp. 333–359. [Google Scholar]
  33. Pan, N.; Sun, G. Functional Textiles for Improved Performance, Protection and Health; Elsevier: Amsterdam, The Netherlands, 2011; ISBN 0857092871. [Google Scholar]
  34. Gomes, F.I.A.; Teixeira, P.; Azeredo, J.; Oliveira, R. Effect of farnesol on planktonic and biofilm cells of Staphylococcus epidermidis. Curr. Microbiol. 2009, 59, 118–122. [Google Scholar] [CrossRef]
  35. Gyawali, R.; Ibrahim, S.A. Natural products as antimicrobial agents. Food Control 2014, 46, 412–429. [Google Scholar] [CrossRef]
  36. Cowan, M.M. Plant products as antimicrobial agents. Clin. Microbiol. Rev. 1999, 12, 564–582. [Google Scholar] [CrossRef] [Green Version]
  37. Thawabteh, A.; Juma, S.; Bader, M.; Karaman, D.; Scrano, L.; Bufo, S.A.; Karaman, R. The biological activity of natural alkaloids against herbivores, cancerous cells and pathogens. Toxins 2019, 11, 656. [Google Scholar] [CrossRef]
  38. Khan, I.A.; Mirza, Z.M.; Kumar, A.; Verma, V.; Qazi, G.N. Piperine, a phytochemical potentiator of ciprofloxacin against Staphylococcus aureus. Antimicrob. Agents Chemother. 2006, 50, 810–812. [Google Scholar] [CrossRef] [PubMed]
  39. Lagha, R.; Ben Abdallah, F.; Al-Sarhan, B.O.; Al-Sodany, Y. Antibacterial and biofilm inhibitory activity of medicinal plant essential oils against Escherichia coli isolated from UTI patients. Molecules 2019, 24, 1161. [Google Scholar] [CrossRef]
  40. Gomes, F.; Dias, M.I.; Lima, Â.; Barros, L.; Rodrigues, M.E.; Ferreira, I.C.F.R.; Henriques, M. Satureja montana L. and Origanum majorana L. decoctions: Antimicrobial activity, mode of action and phenolic characterization. Antibiotics 2020, 9, 294. [Google Scholar] [CrossRef]
  41. Sampath Kumar, N.S.; Sarbon, N.M.; Rana, S.S.; Chintagunta, A.D.; Prathibha, S.; Ingilala, S.K.; Jeevan Kumar, S.P.; Sai Anvesh, B.; Dirisala, V.R. Extraction of bioactive compounds from Psidium guajava leaves and its utilization in preparation of jellies. AMB Express 2021, 11, 1–9. [Google Scholar] [CrossRef] [PubMed]
  42. Agyare, C.; Bempah, S.B.; Boakye, Y.D.; Ayande, P.G.; Adarkwa-Yiadom, M.; Mensah, K.B. Evaluation of antimicrobial and wound healing potential of Justicia flava and Lannea welwitschii. Evid.-Based Complement. Altern. Med. 2013, 2013, 632927. [Google Scholar] [CrossRef] [PubMed]
  43. Ismail, R.M.; Saleh, A.H.A.; Ali, K.S. GC-MS analysis and antibacterial activity of garlic extract with antibiotic. J. Med. Plants Stud 2020, 8, 26–30. [Google Scholar]
  44. Sabo, V.A.; Knezevic, P. Antimicrobial activity of Eucalyptus camaldulensis Dehn. plant extracts and essential oils: A review. Ind. Crops Prod. 2019, 132, 413–429. [Google Scholar] [CrossRef]
  45. Mahdavi, B.; Ghorat, F.; Nasrollahzadeh, M.S.; Hosseyni-Tabar, M.; Rezaei-Seresht, H. Chemical composition, antioxidant, antibacterial, cytotoxicity, and hemolyses activity of essential oils from flower of Matricaria chamomilla var. chamomilla. Anti-Infect. Agents 2020, 18, 224–232. [Google Scholar] [CrossRef]
  46. Antolak, H.; Czyżowska, A.; Kręgiel, D. Activity of Mentha piperita L. Ethanol Extract against Acetic Acid Bacteria Asaia spp. Foods 2018, 7, 171. [Google Scholar] [CrossRef]
  47. Al-Hadid, K.J. Quantitative analysis of antimicrobial activity of’Foeniculum vulgare’: A review. Plant Omics 2017, 10, 28–36. [Google Scholar] [CrossRef]
  48. Abdel-Naime, W.A.; Fahim, J.R.; Fouad, M.A.; Kamel, M.S. Antibacterial, antifungal, and GC–MS studies of Melissa officinalis. S. Afr. J. Bot. 2019, 124, 228–234. [Google Scholar] [CrossRef]
  49. Petkova, N.; Hambarlyiska, I.; Tumbarski, Y.; Vrancheva, R.; Raeva, M.; Ivanov, I. Phytochemical Composition and Antimicrobial Properties of Burdock (Arctium lappa L.) Roots Extracts. Biointerface Res. Appl. Chem. 2021, 12, 2826–2842. [Google Scholar]
  50. Fathi, M.; Ghane, M.; Pishkar, L. Phytochemical Composition, Antibacterial, and Antibiofilm Activity of Malva sylvestris Against Human Pathogenic Bacteria. Jundishapur J. Nat. Pharm. Prod. 2022, 17, e114164. [Google Scholar] [CrossRef]
  51. Nadia, Z.; Rachid, M. Antioxidant and antibacterial activities of Thymus vulgaris L. Med. Aromat. Plant Res. J. 2013, 1, 5–11. [Google Scholar]
  52. Mejía-Argueta, E.L.; Santillán-Benítez, J.G.; Canales-Martinez, M.M.; Mendoza-Medellín, A. Antimicrobial activity of Syzygium aromaticum L. essential oil on extended-spectrum beta-lactamases-producing Escherichia coli. Bull. Natl. Res. Cent. 2020, 44, 1–7. [Google Scholar] [CrossRef]
  53. Tian, C.; Chang, Y.; Zhang, Z.; Wang, H.; Xiao, S.; Cui, C.; Liu, M. Extraction technology, component analysis, antioxidant, antibacterial, analgesic and anti-inflammatory activities of flavonoids fraction from Tribulus terrestris L. leaves. Heliyon 2019, 5, e02234. [Google Scholar] [CrossRef]
  54. Salma, U.; Saha, S.K.; Sultana, S.; Ahmed, S.M.; Haque, S.D.; Mostaqim, S. The Antibacterial Activity of Ethanolic Extract of Cinnamon (Cinnamomum zeylanicum) against two Food Borne Pathogens: Staphylococcus aureus and Escherichia coli. Mymensingh Med. J. MMJ 2019, 28, 767–772. [Google Scholar] [PubMed]
  55. Wang, X.; Shen, Y.; Thakur, K.; Han, J.; Zhang, J.-G.; Hu, F.; Wei, Z.-J. Antibacterial activity and mechanism of ginger essential oil against Escherichia coli and Staphylococcus aureus. Molecules 2020, 25, 3955. [Google Scholar] [CrossRef]
  56. Czernicka, L.; Grzegorczyk, A.; Marzec, Z.; Antosiewicz, B.; Malm, A.; Kukula-Koch, W. Antimicrobial potential of single metabolites of Curcuma longa assessed in the total extract by thin-layer chromatography-based bioautography and image analysis. Int. J. Mol. Sci. 2019, 20, 898. [Google Scholar] [CrossRef]
  57. Lingaraju, D.P.; Sudarshana, M.S.; Mahendra, C.; Rao, K.P. Phytochemical screening and antimicrobial activity of leaf extracts of Eryngium foetidum L.(Apiaceae). Indo Am. J. Pharm. Res. 2016, 6, 4339–4344. [Google Scholar]
  58. Ojah, E.O.; Oladele, E.O.; Chukwuemeka, P. Phytochemical and antibacterial properties of root extracts from Portulaca oleracea Linn. (Purslane) utilised in the management of diseases in Nigeria. J. Med. Plants Econ. Dev. 2021, 5, 103. [Google Scholar] [CrossRef]
  59. Ahmed, Z.; Noor, A.A. Antibacterial activity of Momordica charantia L. and Citrus limon L. on gram positive and gram negative bacteria. Pure Appl. Biol. 2020, 9, 207–218. [Google Scholar] [CrossRef]
  60. Nigussie, D.; Davey, G.; Legesse, B.A.; Fekadu, A.; Makonnen, E. Antibacterial activity of methanol extracts of the leaves of three medicinal plants against selected bacteria isolated from wounds of lymphoedema patients. BMC Complement. Med. Ther. 2021, 21, 1–10. [Google Scholar] [CrossRef] [PubMed]
  61. Srikacha, N.; Ratananikom, K. Antibacterial activity of plant extracts in different solvents against pathogenic bacteria: An in vitro experiment. J. Acute Dis. 2020, 9, 223. [Google Scholar]
  62. Ribeiro, J.; Silva, V.; Aires, A.; Carvalho, R.; Igrejas, G.; Poeta, P. Antimicrobial activity of phenolic compounds extracted from Platanus hybrida: Exploring alternative therapies for a post-antibiotic era. Proceedings 2020, 66, 18. [Google Scholar] [CrossRef]
  63. Álvarez-Martínez, F.J.; Rodríguez, J.C.; Borrás-Rocher, F.; Barrajón-Catalán, E.; Micol, V. The antimicrobial capacity of Cistus salviifolius and Punica granatum plant extracts against clinical pathogens is related to their polyphenolic composition. Sci. Rep. 2021, 11, 1–12. [Google Scholar] [CrossRef] [PubMed]
  64. Lubis, R.R.; Marlisa, D.D.W. Antibacterial activity of betle leaf (Piper betle L.) extract on inhibiting Staphylococcus aureus in conjunctivitis patient. Am. J. Clin. Exp. Immunol. 2020, 9, 1. [Google Scholar] [PubMed]
  65. El-Beltagi, H.S.; Mohamed, H.I.; Abdelazeem, A.S.; Youssef, R.; Safwat, G. GC-MS Analysis, Antioxidant, Antimicrobial and Anticancer Activities of Extracts from Ficus Sycomorus Fruits and Leaves; Academic Press: Cambridge, MA, USA, 2019. [Google Scholar]
  66. Aabed, K.; Mohammed, A.E.; Benabdelkamel, H.; Masood, A.; Alfadda, A.A.; Alanazi, I.O.; Alnehmi, E.A. Antimicrobial Mechanism and Identification of the Proteins Mediated by Extracts from Asphaltum punjabianum and Myrtus communis. ACS Omega 2020, 5, 31019–31035. [Google Scholar] [CrossRef]
  67. Dib, K.; Cherrah, Y.; Rida, S.; Filali-Maltouf, A.; Ennibi, O. In Vitro Antibacterial Activity of Myrtus communis L. and Marrubium vulgare L. Leaves against Aggregatibacter actinomycetemcomitans and Eikenella corrodens. Evid.-Based Complement. Altern. Med. 2021, 2021, 8351332. [Google Scholar] [CrossRef]
  68. Fahmy, A.H.; Alam, I.P.; Salim, H.M. Bactericidal effects of Extract Bacil Leaves in in-vitro study of Pesudomonas aeruginosa. In Proceedings of the Proceeding Surabaya International Health Conference 2019, Surabaya, Indonesia, 15–30 May 2019; Volume 1, pp. 24–28. [Google Scholar]
  69. Setiawan, T.; Lay, B.W. Clitoria Ternatea ethanolic extract prevents dental caries via inhibiting streptococcus mutans growth and quorum sensing. Food Res. 2021, 5, 492–497. [Google Scholar]
  70. Souissi, M.; Azelmat, J.; Chaieb, K.; Grenier, D. Antibacterial and anti-inflammatory activities of cardamom (Elettaria cardamomum) extracts: Potential therapeutic benefits for periodontal infections. Anaerobe 2020, 61, 102089. [Google Scholar] [CrossRef] [PubMed]
  71. Antika, L.D.; Triana, D.; Ernawati, T. Antimicrobial activity of quinine derivatives against human pathogenic bacteria. In Proceedings of the IOP Conference Series: Earth and Environmental Science, Tangerang, Indonesia, 23–24 October 2019; IOP Publishing: Bristol, UK, 2020; Volume 462, p. 12006. [Google Scholar] [CrossRef]
  72. Na, S.; Kim, J.-H.; Rhee, Y.K.; Oh, S.-W. Enhancing the antimicrobial activity of ginseng against Bacillus cereus and Staphylococcus aureus by heat treatment. Food Sci. Biotechnol. 2018, 27, 203–210. [Google Scholar] [CrossRef]
  73. Stephane, F.F.Y.; Jules, B.K.J. Terpenoids as important bioactive constituents of essential oils, essential oils. In Essential Oils-Bioactive Compounds, New Perspectives and Applications; IntechOpen: London, UK, 2020. [Google Scholar]
  74. Korinek, M.; Handoussa, H.; Tsai, Y.-H.; Chen, Y.-Y.; Chen, M.-H.; Chiou, Z.-W.; Fang, Y.; Chang, F.-R.; Yen, C.-H.; Hsieh, C.-F. Anti-inflammatory and antimicrobial volatile oils: Fennel and cumin inhibit neutrophilic inflammation via regulating calcium and MAPKs. Front. Pharmacol. 2021, 12, 674095. [Google Scholar] [CrossRef] [PubMed]
  75. Mahanta, B.P.; Bora, P.K.; Kemprai, P.; Borah, G.; Lal, M.; Haldar, S. Thermolabile essential oils, aromas and flavours: Degradation pathways, effect of thermal processing and alteration of sensory quality. Food Res. Int. 2021, 145, 110404. [Google Scholar] [CrossRef]
  76. Chouhan, S.; Sharma, K.; Guleria, S. Antimicrobial activity of some essential oils—Present status and future perspectives. Medicines 2017, 4, 58. [Google Scholar] [CrossRef] [PubMed]
  77. Swamy, M.K.; Akhtar, M.S.; Sinniah, U.R. Antimicrobial properties of plant essential oils against human pathogens and their mode of action: An updated review. Evid.-Based Complement. Altern. Med. 2016, 2016, 3012462. [Google Scholar] [CrossRef]
  78. Teixeira, B.; Marques, A.; Ramos, C.; Neng, N.R.; Nogueira, J.M.F.; Saraiva, J.A.; Nunes, M.L. Chemical composition and antibacterial and antioxidant properties of commercial essential oils. Ind. Crops Prod. 2013, 43, 587–595. [Google Scholar] [CrossRef]
  79. Damtie, D.; Mekonnen, Y. Antibacterial activity of essential oils from Ethiopian thyme (Thymus serrulatus and Thymus schimperi) against tooth decay bacteria. PLoS ONE 2020, 15, e0239775. [Google Scholar] [CrossRef] [PubMed]
  80. Sohilait, H.J.; Kainama, H.; Nindatu, M. Chemical composition and antibacterial activity of the essential oils from different parts of Eugenia caryophylata, Thunb grown in Amboina Island. Int. J. Org. Chem. 2018, 8, 229–239. [Google Scholar] [CrossRef]
  81. Xu, Y.; Wei, J.; Wei, Y.; Han, P.; Dai, K.; Zou, X.; Jiang, S.; Xu, F.; Wang, H.; Sun, J. Tea tree oil controls brown rot in peaches by damaging the cell membrane of Monilinia fructicola. Postharvest Biol. Technol. 2021, 175, 111474. [Google Scholar] [CrossRef]
  82. Hossain, S.; Heo, H.; De Silva, B.C.J.; Wimalasena, S.; Pathirana, H.; Heo, G.-J. Antibacterial activity of essential oil from lavender (Lavandula angustifolia) against pet turtle-borne pathogenic bacteria. Lab. Anim. Res. 2017, 33, 195–201. [Google Scholar] [CrossRef]
  83. Nikolić, M.; Jovanović, K.K.; Marković, T.; Marković, D.; Gligorijević, N.; Radulović, S.; Soković, M. Chemical composition, antimicrobial, and cytotoxic properties of five Lamiaceae essential oils. Ind. Crops Prod. 2014, 61, 225–232. [Google Scholar] [CrossRef]
  84. Andrade-Ochoa, S.; Chacón-Vargas, K.F.; Sánchez-Torres, L.E.; Rivera-Chavira, B.E.; Nogueda-Torres, B.; Nevárez-Moorillón, G.V. Differential antimicrobial effect of essential oils and their main components: Insights based on the cell membrane and external structure. Membranes 2021, 11, 405. [Google Scholar] [CrossRef] [PubMed]
  85. Helal, I.M.; El-Bessoumy, A.; Al-Bataineh, E.; Joseph, M.R.P.; Rajagopalan, P.; Chandramoorthy, H.C.; Ben Hadj Ahmed, S. Antimicrobial efficiency of essential oils from traditional medicinal plants of Asir region, Saudi Arabia, over drug resistant isolates. Biomed Res. Int. 2019, 2019, 8928306. [Google Scholar] [CrossRef] [Green Version]
  86. Mahboubi, M.; Heidarytabar, R.; Mahdizadeh, E.; Hosseini, H. Antimicrobial activity and chemical composition of Thymus species and Zataria multiflora essential oils. Agric. Nat. Resour. 2017, 51, 395–401. [Google Scholar] [CrossRef]
  87. Radünz, M.; da Trindade, M.L.M.; Camargo, T.M.; Radünz, A.L.; Borges, C.D.; Gandra, E.A.; Helbig, E. Antimicrobial and antioxidant activity of unencapsulated and encapsulated clove (Syzygium aromaticum, L.) essential oil. Food Chem. 2019, 276, 180–186. [Google Scholar] [CrossRef]
  88. Alizadeh Behbahani, B.; Falah, F.; Lavi Arab, F.; Vasiee, M.; Tabatabaee Yazdi, F. Chemical composition and antioxidant, antimicrobial, and antiproliferative activities of Cinnamomum zeylanicum bark essential oil. Evid.-Based Complement. Altern. Med. 2020, 2020, 5190603. [Google Scholar] [CrossRef]
  89. Brun, P.; Bernabè, G.; Filippini, R.; Piovan, A. In vitro antimicrobial activities of commercially available tea tree (Melaleuca alternifolia) essential oils. Curr. Microbiol. 2019, 76, 108–116. [Google Scholar] [CrossRef]
  90. Lorenzo-Leal, A.C.; Palou, E.; López-Malo, A.; Bach, H. Antimicrobial, cytotoxic, and anti-inflammatory activities of Pimenta dioica and Rosmarinus officinalis essential oils. Biomed Res. Int. 2019, 2019, 1639726. [Google Scholar] [CrossRef]
  91. Mutlu-Ingok, A.; Tasir, S.; Seven, A.; Akgun, N.; Karbancioglu-Guler, F. Evaluation of the single and combined antibacterial efficiency of essential oils for controlling Campylobacter coli, Campylobacter jejuni, Escherichia coli, Staphylococcus aureus, and mixed cultures. Flavour Fragr. J. 2019, 34, 280–287. [Google Scholar] [CrossRef]
  92. Gishen, N.Z.; Taddese, S.; Zenebe, T.; Dires, K.; Tedla, A.; Mengiste, B.; Shenkute, D.; Tesema, A.; Shiferaw, Y.; Lulekal, E. In vitro antimicrobial activity of six Ethiopian medicinal plants against Staphylococcus aureus, Escherichia coli and Candida albicans. Eur. J. Integr. Med. 2020, 36, 101121. [Google Scholar] [CrossRef]
  93. Tu, X.-F.; Hu, F.; Thakur, K.; Li, X.-L.; Zhang, Y.-S.; Wei, Z.-J. Comparison of antibacterial effects and fumigant toxicity of essential oils extracted from different plants. Ind. Crops Prod. 2018, 124, 192–200. [Google Scholar] [CrossRef]
  94. Zhang, J.; Ye, K.-P.; Zhang, X.; Pan, D.-D.; Sun, Y.-Y.; Cao, J.-X. Antibacterial activity and mechanism of action of black pepper essential oil on meat-borne Escherichia coli. Front. Microbiol. 2017, 7, 2094. [Google Scholar] [CrossRef]
  95. Moumni, S.; Elaissi, A.; Trabelsi, A.; Merghni, A.; Chraief, I.; Jelassi, B.; Chemli, R.; Ferchichi, S. Correlation between chemical composition and antibacterial activity of some Lamiaceae species essential oils from Tunisia. BMC Complement. Med. Ther. 2020, 20, 1–15. [Google Scholar] [CrossRef] [Green Version]
  96. Reyes-Jurado, F.; Cervantes-Rincón, T.; Bach, H.; López-Malo, A.; Palou, E. Antimicrobial activity of Mexican oregano (Lippia berlandieri), thyme (Thymus vulgaris), and mustard (Brassica nigra) essential oils in gaseous phase. Ind. Crops Prod. 2019, 131, 90–95. [Google Scholar] [CrossRef]
  97. Li, Z.-H.; Cai, M.; Liu, Y.-S.; Sun, P.-L.; Luo, S.-L. Antibacterial activity and mechanisms of essential oil from Citrus medica L. var. sarcodactylis. Molecules 2019, 24, 1577. [Google Scholar] [CrossRef]
  98. Clavijo-Romero, A.; Quintanilla-Carvajal, M.X.; Ruiz, Y. Stability and antimicrobial activity of eucalyptus essential oil emulsions. Food Sci. Technol. Int. 2019, 25, 24–37. [Google Scholar] [CrossRef]
  99. Ghasemian, A.; Al-Marzoqi, A.-H.; Mostafavi, S.K.S.; Alghanimi, Y.K.; Teimouri, M. Chemical composition and antimicrobial and cytotoxic activities of Foeniculum vulgare Mill essential oils. J. Gastrointest. Cancer 2020, 51, 260–266. [Google Scholar] [CrossRef] [PubMed]
  100. Marinkovic, J.; Markovic, T.; Nikolic, B.; Soldatovic, I.; Ivanov, M.; Ciric, A.; Sokovic, M.; Markovic, D. Antibacterial and Antibiofilm Potential of Leptospermum petersonii FM Bailey, Eucalyptus citriodora Hook., Pelargonium graveolens L’Hér. and Pelargonium roseum (Andrews) DC. Essential Oils Against Selected Dental Isolates. J. Essent. Oil Bear. Plants 2021, 24, 304–316. [Google Scholar] [CrossRef]
  101. Khalil, N.; Ashour, M.; Fikry, S.; Singab, A.N.; Salama, O. Chemical composition and antimicrobial activity of the essential oils of selected Apiaceous fruits. Futur. J. Pharm. Sci. 2018, 4, 88–92. [Google Scholar] [CrossRef]
  102. Kebede, B.H.; Forsido, S.F.; Tola, Y.B.; Astatkie, T. Free radical scavenging capacity, antibacterial activity and essential oil composition of turmeric (Curcuma domestica) varieties grown in Ethiopia. Heliyon 2021, 7, e06239. [Google Scholar] [CrossRef]
  103. Santamarta, S.; Aldavero, A.C.; Rojo, M. Antibacterial Properties of Cymbopogon martinii essential Oil against Bacillus subtillis food industry pathogen. Proceedings 2020, 66, 1. [Google Scholar] [CrossRef]
  104. Hojjati, M. Chemical constituents and antibacterial activity of dill (Anethum graveolens) essential oil. In Proceedings of the 15th ASEAN Conference on Food Science and Technology, Singapore, 14–16 November 2017; pp. 14–17. [Google Scholar]
  105. Amor, G.; Caputo, L.; La Storia, A.; De Feo, V.; Mauriello, G.; Fechtali, T. Chemical composition and antimicrobial activity of Artemisia herba-alba and Origanum majorana essential oils from Morocco. Molecules 2019, 24, 4021. [Google Scholar] [CrossRef] [PubMed]
  106. Fidan, H.; Stefanova, G.; Kostova, I.; Stankov, S.; Damyanova, S.; Stoyanova, A.; Zheljazkov, V.D. Chemical composition and antimicrobial activity of Laurus nobilis L. essential oils from Bulgaria. Molecules 2019, 24, 804. [Google Scholar] [CrossRef] [Green Version]
  107. Bączek, K.B.; Kosakowska, O.; Przybył, J.L.; Pióro-Jabrucka, E.; Costa, R.; Mondello, L.; Gniewosz, M.; Synowiec, A.; Węglarz, Z. Antibacterial and antioxidant activity of essential oils and extracts from costmary (Tanacetum balsamita L.) and tansy (Tanacetum vulgare L.). Ind. Crops Prod. 2017, 102, 154–163. [Google Scholar] [CrossRef]
  108. Soliman, F.M.; Fathy, M.M.; Salama, M.M.; Saber, F.R. Comparative study of the volatile oil content and antimicrobial activity of Psidium guajava L. and Psidium cattleianum Sabine leaves. Bull. Fac. Pharm. Cairo Univ. 2016, 54, 219–225. [Google Scholar] [CrossRef]
  109. Erdogan, A.; Ozkan, A. Investigatıon of antioxıdative, cytotoxic, membrane-damaging and membrane-protective effects of the essentıal oil of Origanum majorana and its oxygenated monoterpene component linalool in human-derived Hep G2 cell line. Iran. J. Pharm. Res. IJPR 2017, 16, 24. [Google Scholar] [PubMed]
  110. Simirgiotis, M.J.; Burton, D.; Parra, F.; López, J.; Muñoz, P.; Escobar, H.; Parra, C. Antioxidant and antibacterial capacities of Origanum vulgare L. essential oil from the arid Andean Region of Chile and its chemical characterization by GC-MS. Metabolites 2020, 10, 414. [Google Scholar] [CrossRef]
  111. Middaugh, J.; Hamel, R.; Jean-Baptiste, G.; Beriault, R.; Chenier, D.; Appanna, V.D. Aluminum triggers decreased aconitase activity via Fe-S cluster disruption and the overexpression of isocitrate dehydrogenase and isocitrate lyase: A metabolic network mediating cellular survival. J. Biol. Chem. 2005, 280, 3159–3165. [Google Scholar] [CrossRef]
  112. Macomber, L.; Elsey, S.P.; Hausinger, R.P. Fructose-1, 6-bisphosphate aldolase (class II) is the primary site of nickel toxicity in Escherichia coli. Mol. Microbiol. 2011, 82, 1291–1300. [Google Scholar] [CrossRef]
  113. Seil, J.T.; Webster, T.J. Antimicrobial applications of nanotechnology: Methods and literature. Int. J. Nanomed. 2012, 7, 2767. [Google Scholar]
  114. Lemire, J.A.; Harrison, J.J.; Turner, R.J. Antimicrobial activity of metals: Mechanisms, molecular targets and applications. Nat. Rev. Microbiol. 2013, 11, 371–384. [Google Scholar] [CrossRef]
  115. Malarkodi, C.; Rajeshkumar, S.; Paulkumar, K.; Vanaja, M.; Gnanajobitha, G.; Annadurai, G. Biosynthesis and antimicrobial activity of semiconductor nanoparticles against oral pathogens. Bioinorg. Chem. Appl. 2014, 2014, 347167. [Google Scholar] [CrossRef] [PubMed]
  116. Biswal, A.K.; Misra, P.K. Biosynthesis and characterization of silver nanoparticles for prospective application in food packaging and biomedical fields. Mater. Chem. Phys. 2020, 250, 123014. [Google Scholar] [CrossRef]
  117. Yaqoob, A.A.; Ahmad, H.; Parveen, T.; Ahmad, A.; Oves, M.; Ismail, I.M.I.; Qari, H.A.; Umar, K.; Mohamad Ibrahim, M.N. Recent Advances in Metal Decorated Nanomaterials and Their Various Biological Applications: A Review. Front. Chem. 2020, 8, 1–23. [Google Scholar] [CrossRef] [PubMed]
  118. Díez-Pascual, A.M. Antibacterial activity of nanomaterials. Nanomaterials 2018, 8, 359. [Google Scholar] [CrossRef]
  119. Pardhi, D.M.; Karaman, D.Ş.; Timonen, J.; Wu, W.; Zhang, Q.; Satija, S.; Mehta, M.; Charbe, N.; McCarron, P.A.; Tambuwala, M.M. Anti-bacterial activity of inorganic nanomaterials and their antimicrobial peptide conjugates against resistant and non-resistant pathogens. Int. J. Pharm. 2020, 586, 119531. [Google Scholar] [CrossRef]
  120. Egger, S.; Lehmann, R.P.; Height, M.J.; Loessner, M.J.; Schuppler, M. Antimicrobial properties of a novel silver-silica nanocomposite material. Appl. Environ. Microbiol. 2009, 75, 2973–2976. [Google Scholar] [CrossRef]
  121. Yuan, P.; Ding, X.; Yang, Y.Y.; Xu, Q. Metal nanoparticles for diagnosis and therapy of bacterial infection. Adv. Healthc. Mater. 2018, 7, 1701392. [Google Scholar] [CrossRef]
  122. Hsueh, Y.-H.; Tsai, P.-H.; Lin, K.-S. Ph-dependent antimicrobial properties of copper oxide nanoparticles in staphylococcus aureus. Int. J. Mol. Sci. 2017, 18, 793. [Google Scholar] [CrossRef] [PubMed]
  123. Yadav, H.M.; Kolekar, T.V.; Pawar, S.H.; Kim, J.-S. Enhanced photocatalytic inactivation of bacteria on Fe-containing TiO2 nanoparticles under fluorescent light. J. Mater. Sci. Mater. Med. 2016, 27, 1–9. [Google Scholar] [CrossRef]
  124. Moongraksathum, B.; Chen, Y.-W. Anatase TiO2 co-doped with silver and ceria for antibacterial application. Catal. Today 2018, 310, 68–74. [Google Scholar] [CrossRef]
  125. Şen Karaman, D.; Manner, S.; Rosenholm, J.M. Mesoporous silica nanoparticles as diagnostic and therapeutic tools: How can they combat bacterial infection? Ther. Deliv. 2018, 9, 241–244. [Google Scholar] [CrossRef]
  126. Mishra, K.; Basavegowda, N.; Lee, Y.R. Biosynthesis of Fe, Pd, and Fe-Pd bimetallic nanoparticles and their application as recyclable catalysts for [3 + 2] cycloaddition reaction: A comparative approach. Catal. Sci. Technol. 2015, 5, 2612–2621. [Google Scholar] [CrossRef]
  127. Zhang, J.; Ma, J.; Fan, X.; Peng, W.; Zhang, G.; Zhang, F.; Li, Y. Graphene supported Au-Pd-Fe3O4 alloy trimetallic nanoparticles with peroxidase-like activities as mimic enzyme. Catal. Commun. 2017, 89, 148–151. [Google Scholar] [CrossRef]
  128. Deepak, F.L.; Mayoral, A.; Arenal, R. Advanced Transmission Electron Microscopy: Applications to Nanomaterials; Springer: Berlin/Heidelberg, Germany, 2015; ISBN 3319151770. [Google Scholar]
  129. Merugu, R.; Gothalwal, R.; Deshpande, P.K.; De Mandal, S.; Padala, G.; Chitturi, K.L. Synthesis of Ag/Cu and Cu/Zn bimetallic nanoparticles using toddy palm: Investigations of their antitumor, antioxidant and antibacterial activities. Mater. Today Proc. 2021, 44, 99–105. [Google Scholar] [CrossRef]
  130. Vaseghi, Z.; Tavakoli, O.; Nematollahzadeh, A. Rapid biosynthesis of novel Cu/Cr/Ni trimetallic oxide nanoparticles with antimicrobial activity. J. Environ. Chem. Eng. 2018, 6, 1898–1911. [Google Scholar] [CrossRef]
  131. Paul, D.; Mangla, S.; Neogi, S. Antibacterial study of CuO-NiO-ZnO trimetallic oxide nanoparticle. Mater. Lett. 2020, 271, 127740. [Google Scholar] [CrossRef]
  132. Dong, Y.; Zhu, H.; Shen, Y.; Zhang, W.; Zhang, L. Antibacterial activity of silver nanoparticles of different particle size against Vibrio Natriegens. PLoS ONE 2019, 14, e0222322. [Google Scholar] [CrossRef] [PubMed]
  133. Manikandan, V.; Velmurugan, P.; Park, J.-H.; Chang, W.-S.; Park, Y.-J.; Jayanthi, P.; Cho, M.; Oh, B.-T. Green synthesis of silver oxide nanoparticles and its antibacterial activity against dental pathogens. 3 Biotech 2017, 7, 1–9. [Google Scholar] [CrossRef] [PubMed]
  134. Liu, S.; Wang, C.; Hou, J.; Wang, P.; Miao, L.; Li, T. Effects of silver sulfide nanoparticles on the microbial community structure and biological activity of freshwater biofilms. Environ. Sci. Nano 2018, 5, 2899–2908. [Google Scholar] [CrossRef]
  135. Zhang, S.S.; Wang, X.; Su, H.F.; Feng, L.; Wang, Z.; Ding, W.Q.; Blatov, V.A.; Kurmoo, M.; Tung, C.H.; Sun, D.; et al. A Water-Stable Cl@Ag14 Cluster Based Metal-Organic Open Framework for Dichromate Trapping and Bacterial Inhibition. Inorg. Chem. 2017, 56, 11891–11899. [Google Scholar] [CrossRef]
  136. Suryavanshi, P.; Pandit, R.; Gade, A.; Derita, M.; Zachino, S.; Rai, M. Colletotrichum sp.-mediated synthesis of sulphur and aluminium oxide nanoparticles and its in vitro activity against selected food-borne pathogens. LWT-Food Sci. Technol. 2017, 81, 188–194. [Google Scholar] [CrossRef]
  137. Ortiz-Benítez, E.A.; Velázquez-Guadarrama, N.; Durán Figueroa, N.V.; Quezada, H.; Olivares-Trejo, J.d.J. Antibacterial mechanism of gold nanoparticles on Streptococcus pneumoniae. Metallomics 2019, 11, 1265–1276. [Google Scholar] [CrossRef]
  138. Rieznichenko, L.S.; Gruzina, T.G.; Dybkova, S.M.; Ushkalov, V.O.; Ulberg, Z.R. Investigation of bismuth nanoparticles antimicrobial activity against high pathogen microorganisms. Am. J. Bioterror. Biosecur. Biodef 2015, 2, 1004. [Google Scholar]
  139. Keihan, A.H.; Veisi, H.; Veasi, H. Green synthesis and characterization of spherical copper nanoparticles as organometallic antibacterial agent. Appl. Organomet. Chem. 2017, 31, e3642. [Google Scholar] [CrossRef]
  140. Marquis, G.; Ramasamy, B.; Banwarilal, S.; Munusamy, A.P. Evaluation of antibacterial activity of plant mediated CaO nanoparticles using Cissus quadrangularis extract. J. Photochem. Photobiol. B Biol. 2016, 155, 28–33. [Google Scholar] [CrossRef]
  141. Qamar, H.; Rehman, S.; Chauhan, D.K.; Tiwari, A.K.; Upmanyu, V. Green synthesis, characterization and antimicrobial activity of copper oxide nanomaterial derived from Momordica charantia. Int. J. Nanomed. 2020, 15, 2541. [Google Scholar] [CrossRef] [PubMed]
  142. Pop, O.L.; Mesaros, A.; Vodnar, D.C.; Suharoschi, R.; Tăbăran, F.; Magerușan, L.; Tódor, I.S.; Diaconeasa, Z.; Balint, A.; Ciontea, L. Cerium oxide nanoparticles and their efficient antibacterial application in vitro against gram-positive and gram-negative pathogens. Nanomaterials 2020, 10, 1614. [Google Scholar] [CrossRef] [PubMed]
  143. Katata-Seru, L.; Moremedi, T.; Aremu, O.S.; Bahadur, I. Green synthesis of iron nanoparticles using Moringa oleifera extracts and their applications: Removal of nitrate from water and antibacterial activity against Escherichia coli. J. Mol. Liq. 2018, 256, 296–304. [Google Scholar] [CrossRef]
  144. Saqib, S.; Munis, M.F.H.; Zaman, W.; Ullah, F.; Shah, S.N.; Ayaz, A.; Farooq, M.; Bahadur, S. Synthesis, characterization and use of iron oxide nano particles for antibacterial activity. Microsc. Res. Tech. 2019, 82, 415–420. [Google Scholar] [CrossRef] [PubMed]
  145. Argueta-Figueroa, L.; Torres-Gómez, N.; García-Contreras, R.; Vilchis-Nestor, A.R.; Martínez-Alvarez, O.; Acosta-Torres, L.S.; Arenas-Arrocena, M.C. Hydrothermal synthesis of pyrrhotite (Fex-1S) nanoplates and their antibacterial, cytotoxic activity study. Prog. Nat. Sci. Mater. Int. 2018, 28, 447–455. [Google Scholar] [CrossRef]
  146. Narayanasamy, P.; Switzer, B.L.; Britigan, B.E. Prolonged-acting, multi-targeting gallium nanoparticles potently inhibit growth of both HIV and mycobacteria in co-infected human macrophages. Sci. Rep. 2015, 5, 1–7. [Google Scholar] [CrossRef]
  147. Kamran, U.; Bhatti, H.N.; Iqbal, M.; Jamil, S.; Zahid, M. Biogenic synthesis, characterization and investigation of photocatalytic and antimicrobial activity of manganese nanoparticles synthesized from Cinnamomum verum bark extract. J. Mol. Struct. 2019, 1179, 532–539. [Google Scholar] [CrossRef]
  148. Maji, J.; Pandey, S.; Basu, S. Synthesis and evaluation of antibacterial properties of magnesium oxide nanoparticles. Bull. Mater. Sci. 2020, 43, 1–10. [Google Scholar] [CrossRef]
  149. Kumar, G.S.; Venkataramana, B.; Reddy, S.A.; Maseed, H.; Nagireddy, R.R. Hydrothermal synthesis of Mn3O4 nanoparticles by evaluation of pH effect on particle size formation and its antibacterial activity. Adv. Nat. Sci. Nanosci. Nanotechnol. 2020, 11, 35006. [Google Scholar] [CrossRef]
  150. André, V.; da Silva, A.R.F.; Fernandes, A.; Frade, R.; Garcia, C.; Rijo, P.; Antunes, A.M.M.; Rocha, J.; Duarte, M.T. Mg-and Mn-MOFs boost the antibiotic activity of nalidixic acid. ACS Appl. Bio Mater. 2019, 2, 2347–2354. [Google Scholar] [CrossRef]
  151. Din, M.I.; Nabi, A.G.; Rani, A.; Aihetasham, A.; Mukhtar, M. Single step green synthesis of stable nickel and nickel oxide nanoparticles from Calotropis gigantea: Catalytic and antimicrobial potentials. Environ. Nanotechnol. Monit. Manag. 2018, 9, 29–36. [Google Scholar] [CrossRef]
  152. Behera, N.; Arakha, M.; Priyadarshinee, M.; Pattanayak, B.S.; Soren, S.; Jha, S.; Mallick, B.C. Oxidative stress generated at nickel oxide nanoparticle interface results in bacterial membrane damage leading to cell death. RSC Adv. 2019, 9, 24888–24894. [Google Scholar] [CrossRef] [PubMed]
  153. Mohana, S.; Sumathi, S. Multi-functional biological effects of palladium nanoparticles synthesized using Agaricus bisporus. J. Clust. Sci. 2020, 31, 391–400. [Google Scholar] [CrossRef]
  154. Ahmed, K.B.A.; Raman, T.; Anbazhagan, V. Platinum nanoparticles inhibit bacteria proliferation and rescue zebrafish from bacterial infection. Rsc. Adv. 2016, 6, 44415–44424. [Google Scholar] [CrossRef]
  155. Geoffrion, L.D.; Hesabizadeh, T.; Medina-Cruz, D.; Kusper, M.; Taylor, P.; Vernet-Crua, A.; Chen, J.; Ajo, A.; Webster, T.J.; Guisbiers, G. Naked selenium nanoparticles for antibacterial and anticancer treatments. ACS Omega 2020, 5, 2660–2669. [Google Scholar] [CrossRef]
  156. Smirnov, N.A.; Kudryashov, S.I.; Nastulyavichus, A.A.; Rudenko, A.A.; Saraeva, I.N.; Tolordava, E.R.; Gonchukov, S.A.; Romanova, Y.M.; Ionin, A.A.; Zayarny, D.A. Antibacterial properties of silicon nanoparticles. Laser Phys. Lett. 2018, 15, 105602. [Google Scholar] [CrossRef]
  157. Eisa, N.E.; Almansour, S.; Alnaim, I.A.; Ali, A.M.; Algrafy, E.; Ortashi, K.M.; Awad, M.A.; Virk, P.; Hendi, A.A.; Eissa, F.Z. Eco-synthesis and characterization of titanium nanoparticles: Testing its cytotoxicity and antibacterial effects. Green Process. Synth. 2020, 9, 462–468. [Google Scholar] [CrossRef]
  158. Tiwari, V.; Mishra, N.; Gadani, K.; Solanki, P.S.; Shah, N.A.; Tiwari, M. Mechanism of anti-bacterial activity of zinc oxide nanoparticle against carbapenem-resistant Acinetobacter baumannii. Front. Microbiol. 2018, 9, 1218. [Google Scholar] [CrossRef]
  159. Khan, M.; Shaik, M.R.; Khan, S.T.; Adil, S.F.; Kuniyil, M.; Khan, M.; Al-Warthan, A.A.; Siddiqui, M.R.H.; Nawaz Tahir, M. Enhanced antimicrobial activity of biofunctionalized zirconia nanoparticles. ACS Omega 2020, 5, 1987–1996. [Google Scholar] [CrossRef]
  160. Pezeshkpour, V.; Khosravani, S.A.; Ghaedi, M.; Dashtian, K.; Zare, F.; Sharifi, A.; Jannesar, R.; Zoladl, M. Ultrasound assisted extraction of phenolic acids from broccoli vegetable and using sonochemistry for preparation of MOF-5 nanocubes: Comparative study based on micro-dilution broth and plate count method for synergism antibacterial effect. Ultrason. Sonochem. 2018, 40, 1031–1038. [Google Scholar] [CrossRef]
  161. Andrade, G.R.S.; Nascimento, C.C.; Lima, Z.M.; Teixeira-Neto, E.; Costa, L.P.; Gimenez, I.F. Star-shaped ZnO/Ag hybrid nanostructures for enhanced photocatalysis and antibacterial activity. Appl. Surf. Sci. 2017, 399, 573–582. [Google Scholar] [CrossRef]
  162. Addae, E.; Dong, X.; McCoy, E.; Yang, C.; Chen, W.; Yang, L. Investigation of antimicrobial activity of photothermal therapeutic gold/copper sulfide core/shell nanoparticles to bacterial spores and cells. J. Biol. Eng. 2014, 8, 1–11. [Google Scholar] [CrossRef]
  163. Lozhkomoev, A.S.; Bakina, O.V.; Pervikov, A.V.; Kazantsev, S.O.; Glazkova, E.A. Synthesis of CuO–ZnO composite nanoparticles by electrical explosion of wires and their antibacterial activities. J. Mater. Sci. Mater. Electron. 2019, 30, 13209–13216. [Google Scholar] [CrossRef]
  164. Singh, S.; Barick, K.C.; Bahadur, D. Inactivation of bacterial pathogens under magnetic hyperthermia using Fe3O4–ZnO nanocomposite. Powder Technol. 2015, 269, 513–519. [Google Scholar] [CrossRef]
  165. Yadav, N.; Jaiswal, A.K.; Dey, K.K.; Yadav, V.B.; Nath, G.; Srivastava, A.K.; Yadav, R.R. Trimetallic Au/Pt/Ag based nanofluid for enhanced antibacterial response. Mater. Chem. Phys. 2018, 218, 10–17. [Google Scholar] [CrossRef]
  166. Alzahrani, K.E.; Niazy, A.A.; Alswieleh, A.M.; Wahab, R.; El-Toni, A.M.; Alghamdi, H.S. Antibacterial activity of trimetal (CuZnFe) oxide nanoparticles. Int. J. Nanomed. 2018, 13, 77. [Google Scholar] [CrossRef] [PubMed]
  167. Rajendiran, K.; Zhao, Z.; Pei, D.-S.; Fu, A. Antimicrobial activity and mechanism of functionalized quantum dots. Polymers 2019, 11, 1670. [Google Scholar] [CrossRef]
  168. Kumari, A.; Thakur, N.; Vashishtt, J.; Singh, R.R. Structural, luminescent and antimicrobial properties of ZnS and CdSe/ZnS quantum dot structures originated by precursors. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2020, 229, 117962. [Google Scholar] [CrossRef] [PubMed]
  169. Sabah, A.; Tasleem, S.; Murtaza, M.; Nazir, M.; Rashid, F. Effect of polymer capping on photonic multi-core–shell quantum dots CdSe/CdS/ZnS: Impact of sunlight and antibacterial activity. J. Phys. Chem. C 2020, 124, 9009–9020. [Google Scholar] [CrossRef]
  170. Shariati, M.R.; Samadi-Maybodi, A.; Colagar, A.H. Dual cocatalyst loaded reverse type-I core/shell quantum dots for photocatalytic antibacterial applications. J. Mater. Chem. A 2018, 6, 20433–20443. [Google Scholar] [CrossRef]
  171. Mir, I.A.; Alam, H.; Priyadarshini, E.; Meena, R.; Rawat, K.; Rajamani, P.; Rizvi, M.S.; Bohidar, H.B. Antimicrobial and biocompatibility of highly fluorescent ZnSe core and ZnSe@ ZnS core-shell quantum dots. J. Nanoparticle Res. 2018, 20, 1–11. [Google Scholar] [CrossRef]
  172. Li, W.; Song, P.; Xin, Y.; Kuang, Z.; Liu, Q.; Ge, F.; Zhu, L.; Zhang, X.; Tao, Y. The effects of luminescent CdSe quantum dot-functionalized antimicrobial peptides nanoparticles on antibacterial activity and molecular mechanism. Int. J. Nanomed. 2021, 16, 1849. [Google Scholar] [CrossRef] [PubMed]
  173. Chai, S.; Zhou, L.; Pei, S.; Zhu, Z.; Chen, B. P-Doped Carbon Quantum Dots with Antibacterial Activity. Micromachines 2021, 12, 1116. [Google Scholar] [CrossRef] [PubMed]
  174. Elyamny, S.; Eltarahony, M.; Abu-Serie, M.; Nabil, M.M.; Kashyout, A.E.-H.B. One-pot fabrication of Ag@ Ag2O core–shell nanostructures for biosafe antimicrobial and antibiofilm applications. Sci. Rep. 2021, 11, 1–13. [Google Scholar] [CrossRef] [PubMed]
  175. Radhi, A.; Mohamad, D.; Rahman, F.S.A.; Abdullah, A.M.; Hasan, H. Mechanism and factors influence of graphene-based nanomaterials antimicrobial activities and application in dentistry. J. Mater. Res. Technol. 2021, 11, 1290–1307. [Google Scholar] [CrossRef]
  176. Raja, A.; Selvakumar, K.; Rajasekaran, P.; Arunpandian, M.; Ashokkumar, S.; Kaviyarasu, K.; Bahadur, S.A.; Swaminathan, M. Visible active reduced graphene oxide loaded titania for photodecomposition of ciprofloxacin and its antibacterial activity. Colloids Surf. A Physicochem. Eng. Asp. 2019, 564, 23–30. [Google Scholar] [CrossRef]
  177. Trinh, L.T.; Quynh, L.A.B.; Hieu, N.H. Synthesis of zinc oxide/graphene oxide nanocomposite material for antibacterial application. Int. J. Nanotechnol. 2018, 15, 108–117. [Google Scholar] [CrossRef]
  178. Yang, Z.; Hao, X.; Chen, S.; Ma, Z.; Wang, W.; Wang, C.; Yue, L.; Sun, H.; Shao, Q.; Murugadoss, V. Long-term antibacterial stable reduced graphene oxide nanocomposites loaded with cuprous oxide nanoparticles. J. Colloid Interface Sci. 2019, 533, 13–23. [Google Scholar] [CrossRef]
  179. Tu, Q.; Zhang, Q.; Wang, Y.; Jiao, Y.; Xiao, J.; Peng, T.; Wang, J. Antibacterial properties of poly (dimethylsiloxane) surfaces modified with graphene oxide-catechol composite. Prog. Org. Coat. 2019, 129, 247–253. [Google Scholar] [CrossRef]
  180. Hassani, M.; Tahghighi, A.; Rohani, M.; Hekmati, M.; Ahmadian, M.; Ahmadvand, H. Robust antibacterial activity of functionalized carbon nanotube-levofloxacine conjugate based on in vitro and in vivo studies. Sci. Rep. 2022, 12, 1–19. [Google Scholar] [CrossRef] [PubMed]
  181. Abo-Neima, S.E.; Motaweh, H.A.; Elsehly, E.M. Antimicrobial activity of functionalised carbon nanotubes against pathogenic microorganisms. IET Nanobiotechnol. 2020, 14, 457–464. [Google Scholar] [CrossRef]
  182. Mohammed, M.K.A.; Ahmed, D.S.; Mohammad, M.R. Studying antimicrobial activity of carbon nanotubes decorated with metal-doped ZnO hybrid materials. Mater. Res. Express 2019, 6, 55404. [Google Scholar] [CrossRef]
  183. Alfei, S.; Schito, A.M.; Zuccari, G. Considerable Improvement of Ursolic Acid Water Solubility by its Encapsulation in Dendrimer Nanoparticles: Design, Synthesis and Physicochemical Characterization. Nanomaterials 2021, 11, 2196. [Google Scholar] [CrossRef] [PubMed]
  184. Alfei, S.; Brullo, C.; Caviglia, D.; Zuccari, G. Preparation and Physicochemical Characterization of Water-Soluble Pyrazole-Based Nanoparticles by Dendrimer Encapsulation of an Insoluble Bioactive Pyrazole Derivative. Nanomaterials 2021, 11, 2662. [Google Scholar] [CrossRef]
  185. Jiang, G.; Liu, S.; Yu, T.; Wu, R.; Ren, Y.; van der Mei, H.C.; Liu, J.; Busscher, H.J. PAMAM dendrimers with dual-conjugated vancomycin and Ag-nanoparticles do not induce bacterial resistance and kill vancomycin-resistant Staphylococci. Acta Biomater. 2021, 123, 230–243. [Google Scholar] [CrossRef] [PubMed]
  186. Alfei, S.; Schito, A.M. Positively charged polymers as promising devices against multidrug resistant gram-negative bacteria: A Review. Polymers 2020, 12, 1195. [Google Scholar] [CrossRef]
  187. Mazumder, A.; Davis, J.; Rangari, V.; Curry, M. Synthesis, characterization, and applications of dendrimer-encapsulated zero-valent Ni nanoparticles as antimicrobial agents. Int. Sch. Res. Not. 2013, 2013, 843709. [Google Scholar] [CrossRef]
  188. Gholami, M.; Mohammadi, R.; Arzanlou, M.; Akbari Dourbash, F.; Kouhsari, E.; Majidi, G.; Mohseni, S.M.; Nazari, S. In vitro antibacterial activity of poly (amidoamine)-G7 dendrimer. BMC Infect. Dis. 2017, 17, 1–11. [Google Scholar] [CrossRef] [PubMed]
  189. Schito, A.M.; Alfei, S. Antibacterial activity of non-cytotoxic, amino acid-modified polycationic dendrimers against Pseudomonas aeruginosa and other non-fermenting gram-negative bacteria. Polymers 2020, 12, 1818. [Google Scholar] [CrossRef]
  190. Tauanov, Z.; Zakiruly, O.; Baimenova, Z.; Baimenov, A.; Akimbekov, N.S.; Berillo, D. Antimicrobial Properties of the Triclosan-Loaded Polymeric Composite Based on Unsaturated Polyester Resin: Synthesis, Characterization and Activity. Polymers 2022, 14, 676. [Google Scholar] [CrossRef]
  191. Jaramillo, A.F.; Riquelme, S.A.; Sánchez-Sanhueza, G.; Medina, C.; Solís-Pomar, F.; Rojas, D.; Montalba, C.; Melendrez, M.F.; Pérez-Tijerina, E. Comparative study of the antimicrobial effect of nanocomposites and composite based on poly (butylene adipate-co-terephthalate) using Cu and Cu/Cu2O nanoparticles and CuSO4. Nanoscale Res. Lett. 2019, 14, 1–17. [Google Scholar] [CrossRef]
  192. Jalageri, M.D.; Puttaiahgowda, Y.M. Design and antimicrobial activity of piperazine polymer nanocomposite. Mater. Today Proc. 2019, 15, 262–267. [Google Scholar] [CrossRef]
  193. Gautam, S.; Sharma, S.; Sharma, B.; Jain, P. Antibacterial efficacy of poly (vinyl alcohol) nanocomposites reinforced with graphene oxide and silver nanoparticles for packaging applications. Polym. Compos. 2021, 42, 2829–2837. [Google Scholar] [CrossRef]
  194. Aiyegoro, O.A.; Afolayan, A.J.; Okoh, A.I. Synergistic interaction of Helichrysum pedunculatum leaf extracts with antibiotics against wound infection associated bacteria. Biol. Res. 2009, 42, 327–338. [Google Scholar] [CrossRef]
  195. Iseppi, R.; Mariani, M.; Condò, C.; Sabia, C.; Messi, P. Essential oils: A natural weapon against antibiotic-resistant bacteria responsible for nosocomial infections. Antibiotics 2021, 10, 417. [Google Scholar] [CrossRef]
  196. Bassolé, I.H.N.; Lamien-Meda, A.; Bayala, B.; Tirogo, S.; Franz, C.; Novak, J.; Nebié, R.C.; Dicko, M.H. Composition and antimicrobial activities of Lippia multiflora Moldenke, Mentha x piperita L. and Ocimum basilicum L. essential oils and their major monoterpene alcohols alone and in combination. Molecules 2010, 15, 7825–7839. [Google Scholar] [CrossRef]
  197. Soltanzadeh, M.; Peighambardoust, S.H.; Ghanbarzadeh, B.; Mohammadi, M.; Lorenzo, J.M. Chitosan nanoparticles encapsulating lemongrass (Cymbopogon commutatus) essential oil: Physicochemical, structural, antimicrobial and in-vitro release properties. Int. J. Biol. Macromol. 2021, 192, 1084–1097. [Google Scholar] [CrossRef]
  198. Saquib, S.A.; AlQahtani, N.A.; Ahmad, I.; Kader, M.A.; Al Shahrani, S.S.; Asiri, E.A. Evaluation and comparison of antibacterial efficacy of herbal extracts in combination with antibiotics on periodontal pathobionts: An in vitro microbiological study. Antibiotics 2019, 8, 89. [Google Scholar] [CrossRef]
  199. Bahmani, M.; Taherikalani, M.; Khaksarian, M.; Rafieian-Kopaei, M.; Ashrafi, B.; Nazer, M.; Soroush, S.; Abbasi, N.; Rashidipour, M. The synergistic effect of hydroalcoholic extracts of Origanum vulgare, Hypericum perforatum and their active components carvacrol and hypericin against Staphylococcus aureus. Futur. Sci. OA 2019, 5, FSO371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  200. Musimun, C.; Papiernik, D.; Permpoonpattana, P.; Chumkaew, P.; Srisawat, T. Synergy of green-synthesized silver nanoparticles and Vatica diospyroides fruit extract in inhibiting Gram-positive bacteria by inducing membrane and intracellular disruption. J. Exp. Nanosci. 2022, 17, 420–438. [Google Scholar] [CrossRef]
  201. Aabed, K.; Mohammed, A.E. Synergistic and antagonistic effects of biogenic silver nanoparticles in combination with antibiotics against some pathogenic microbes. Front. Bioeng. Biotechnol. 2021, 9, 652362. [Google Scholar] [CrossRef]
  202. Lee, N.L.S.; Yuen, K.Y.; Kumana, C.R. β-Lactam antibiotic and β-lactamase inhibitor combinations. JAMA 2001, 285, 386–388. [Google Scholar] [CrossRef] [PubMed]
  203. Rakholiya, K.D.; Kaneria, M.J.; Chanda, S.V. Medicinal plants as alternative sources of therapeutics against multidrug-resistant pathogenic microorganisms based on their antimicrobial potential and synergistic properties. In Fighting Multidrug Resistance with Herbal Extracts, essential Oils and Their Components; Academic Press: Cambridge, MA, USA, 2013; pp. 165–179. [Google Scholar] [CrossRef]
  204. Rachuonyo, H.; Ogola, P.; Arika, W.; Wambani, J.; Gatheri, G. Combined effect of crude leaf extracts of selected medicinal plants against selected enteric bacterial pathogens and Candida albicans. J. Antimicrob Agents 2016, 2, 1212–2472. [Google Scholar]
  205. Obuekwe, I.S.; Okoyomo, E.P.; Anka, U.S. Effect of Plant Extract Combinations on Some Bacterial Pathogens. J. Appl. Sci. Environ. Manag. 2020, 24, 627–632. [Google Scholar] [CrossRef]
  206. Gadisa, E.; Usman, H. Evaluation of Antibacterial Activity of Essential Oils and Their Combination against Multidrug-Resistant Bacteria Isolated from Skin Ulcer. Int. J. Microbiol. 2021, 2021, 6680668. [Google Scholar] [CrossRef]
  207. Purkait, S.; Bhattacharya, A.; Bag, A.; Chattopadhyay, R.R. Synergistic antibacterial, antifungal and antioxidant efficacy of cinnamon and clove essential oils in combination. Arch. Microbiol. 2020, 202, 1439–1448. [Google Scholar] [CrossRef] [PubMed]
  208. Garza-Cervantes, J.A.; Chávez-Reyes, A.; Castillo, E.C.; García-Rivas, G.; Antonio Ortega-Rivera, O.; Salinas, E.; Ortiz-Martínez, M.; Gómez-Flores, S.L.; Peña-Martínez, J.A.; Pepi-Molina, A. Synergistic antimicrobial effects of silver/transition-metal combinatorial treatments. Sci. Rep. 2017, 7, 1–16. [Google Scholar] [CrossRef]
  209. Alfei, S.; Schito, A.M. β-Lactam Antibiotics and β-Lactamase Enzymes Inhibitors, Part 2: Our Limited Resources. Pharmaceuticals 2022, 15, 476. [Google Scholar] [CrossRef] [PubMed]
  210. Alfei, S.; Zuccari, G. Recommendations to Synthetize Old and New β-Lactamases Inhibitors: A Review to Encourage Further Production. Pharmaceuticals 2022, 15, 384. [Google Scholar] [CrossRef] [PubMed]
  211. World Health Organization. 2019 Antibacterial Agents in Clinical Development: An Analysis of the Antibacterial Clinical Development Pipeline; World Health Organization: Geneva, Switzerland, 2019. [Google Scholar]
  212. Das, B.; Verma, J.; Kumar, P.; Ghosh, A.; Ramamurthy, T. Antibiotic resistance in Vibrio cholerae: Understanding the ecology of resistance genes and mechanisms. Vaccine 2020, 38, A83–A92. [Google Scholar] [CrossRef]
  213. Peterson, E.; Kaur, P. Antibiotic resistance mechanisms in bacteria: Relationships between resistance determinants of antibiotic producers, environmental bacteria, and clinical pathogens. Front. Microbiol. 2018, 9, 2928. [Google Scholar] [CrossRef]
  214. Sultan, I.; Rahman, S.; Jan, A.T.; Siddiqui, M.T.; Mondal, A.H.; Haq, Q.M.R. Antibiotics, resistome and resistance mechanisms: A bacterial perspective. Front. Microbiol. 2018, 9, 2066. [Google Scholar] [CrossRef]
  215. Kapoor, G.; Saigal, S.; Elongavan, A. Action and resistance mechanisms of antibiotics: A guide for clinicians. J. Anaesthesiol. Clin. Pharmacol. 2017, 33, 300. [Google Scholar] [CrossRef] [PubMed]
  216. Reygaert, W.C. An overview of the antimicrobial resistance mechanisms of bacteria. AIMS Microbiol. 2018, 4, 482. [Google Scholar] [CrossRef] [PubMed]
  217. Álvarez-Martínez, F.J.; Barrajón-Catalán, E.; Herranz-López, M.; Micol, V. Antibacterial plant compounds, extracts and essential oils: An updated review on their effects and putative mechanisms of action. Phytomedicine 2021, 90, 153626. [Google Scholar] [CrossRef]
  218. Mostafa, A.A.; Al-Askar, A.A.; Almaary, K.S.; Dawoud, T.M.; Sholkamy, E.N.; Bakri, M.M. Antimicrobial activity of some plant extracts against bacterial strains causing food poisoning diseases. Saudi J. Biol. Sci. 2018, 25, 361–366. [Google Scholar] [CrossRef] [PubMed]
  219. Nazzaro, F.; Fratianni, F.; De Martino, L.; Coppola, R.; De Feo, V. Effect of essential oils on pathogenic bacteria. Pharmaceuticals 2013, 6, 1451–1474. [Google Scholar] [CrossRef]
  220. Wang, L.; Hu, C.; Shao, L. The antimicrobial activity of nanoparticles: Present situation and prospects for the future. Int. J. Nanomed. 2017, 12, 1227. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The extracts of plant organs, namely, the root, bark, bulbs, leaf, flower, and seed, may encompass distinctive phytochemicals with antimicrobial properties.
Figure 1. The extracts of plant organs, namely, the root, bark, bulbs, leaf, flower, and seed, may encompass distinctive phytochemicals with antimicrobial properties.
Biomedicines 10 02219 g001
Figure 2. Schematic representation of some important EOs containing various components that have been screened for their antimicrobial properties.
Figure 2. Schematic representation of some important EOs containing various components that have been screened for their antimicrobial properties.
Biomedicines 10 02219 g002
Figure 3. Schematic representation of different nanomaterials that possess antimicrobial activity.
Figure 3. Schematic representation of different nanomaterials that possess antimicrobial activity.
Biomedicines 10 02219 g003
Figure 4. Proposed antibacterial mechanisms of combinations of plant extracts, EOs, and nanomaterials.
Figure 4. Proposed antibacterial mechanisms of combinations of plant extracts, EOs, and nanomaterials.
Biomedicines 10 02219 g004
Table 1. A list of plants whose parts have been reported to have antimicrobial activity against various pathogens, as well as their corresponding mechanisms of action.
Table 1. A list of plants whose parts have been reported to have antimicrobial activity against various pathogens, as well as their corresponding mechanisms of action.
PlantsPartsPathogensMechanismRef.
Alchornea cordifoliaflowerE. colidamage of cell wall[39]
Origanum majoranaleavesS. aureus, K. pneumoniaemembrane damage[40]
Psidium guajavaleavesB. subtilis, S. aureuscell wall damage[41]
Justicia flavaleavesE. coli, P. aeruginosachanges in internal pH[42]
Allium sativumbulbsP. aeruginosa, S. aureuscell membrane integrity[43]
Lannea welwitschiileavesE. coli, P. aeruginosacell wall integrity[42]
Eucalyptus camaldulensisleaves, barkS. aureus, B. subtilisleakage of cell constituents[44]
Matricaria chamomillaflowersS. aureus, P. aeruginosacell wall degradation[45]
Mentha piperitaleavesS. aureus, B. subtilisdamage of cytoplasmic membranes[46]
Foeniculum vulgareseedsA. flavus, C. albicanscellular DNA damages[47]
Melissa officinalisleavesS. aureus, P. aeruginosadisrupt the membrane structure[48]
Arctium lapparootsP. aeruginosa, S. aureusdamage by oxidative stress[49]
Malva sylvestrisflower, leavesS. aureus, E. faecalisdamaging the membrane[50]
Thymus vulgarisleavesE. coli, S. aureuschemical affinity for membrane lipids[51]
Syzygium aromaticumbudsE. colimembrane damage and intracellular content leakage[52]
Tribulus terrestrisleavesEscherichia coli, Salmonellamembrane damage and leakage of cellular materials[53]
Cinnamomum zeylanicumbarkS. aureus, E. coliinhibiting of various cellular enzymes[54]
Zingiber officinalerhizomeE. coli, S. aureusdamage to cell membrane[55]
Curcuma longarhizomeS. aureus, B. subtilisloss of membrane integrity[56]
Eryngium foetidumleavesP. aeruginosa, C. albicansdisruption of the cell membrane[57]
Portulaca oleracearootsE. cloacae, B. subtilisinhibiting the efflux pumps[58]
Momordica charantiapeelsS. aureus, B. cereusdisintegrates the membrane[59]
Lawsonia inermisleavesS. aureus, E. coliinactivating microbial adhesions[60]
Azadirachta indicaleavesS. pyogenesinactivating microbial enzymes[60]
Achyranthes asperaleavesS. pyogenesinhibiting energy metabolism[60]
Acacia niloticaseedsS. aureuscell membrane permeability[61]
Platanus hybridafruitsE. faecalis, E. faeciumInhibiting the biofilm production[62]
Cistus salviifoliusaerial partsS. aureuscell wall alterations[63]
Punica granatumpeelsS. aureuscell wall alterations[63]
Piper betleleavesS. aureusdestruction of the bacteria cell wall[64]
Ficus sycomorusleaves, fruitsE. coli, S. aureuspermeability of the cell membranes[65]
Myrtus communisleavesE. coliproteins in the outer membrane specifically involved[66]
Asphaltum punjabianummineral resinE. coliproteins involved specifically in the outer membrane[66]
Marrubium vulgareleavesA. actinomycetemcomitans, E. corrodensaffect cytoplasmic membrane[67]
Ocimum
basilicum
leavesP. aeruginosabacterial cells will lose cations and macromolecules[68]
Clitoria ternateaflowersStreptococcus mutansquorum sensing inhibition[69]
Elettaria cardamomumSeedsP. gingivaliscell membrane disrupted[70]
Cinchona officinalisbarkE. coli, P. aeruginosastructural damage of bacterial cells[71]
Panax ginsengrootsB. cereus, S. aureuschanges in the membrane potential[72]
Table 2. Antimicrobial properties of various EOs with their respective plant sources and mechanisms of action.
Table 2. Antimicrobial properties of various EOs with their respective plant sources and mechanisms of action.
Essential OilsPlant SourceMajor ComponentsPathogensModes of ActionRef.
BasilOcimum basilicumlinaloolS. aureusdisrupt the permeability barrier[85]
ThymeThymus vulgaristhymolP. aeruginosa,
A. niger
interferes with membrane functions[86]
CloveSyzygium aromaticumeugenolS. aureus,
S. Typhimurium
sensitivity to eugenol[87]
CinnamonCinnamomum zeylanicumcinnamaldehydeE. coli,
L. innocua
facilitate intracellular compounds leakage[88]
Tea treeMelaleuca alternifoliaterpinen-4-olP. aeruginosa,
C. glabrata
alterations of the biological membrane[89]
RosemaryRosmarinus officinalisα-pineneC. albicansrupture of the membranes and cell wall[90]
DillAnethum graveolenscarvoneS. aureus, E. colilesion in the plasma membrane[91]
CuminCuminum cyminump-mentha-1,3-dien-7-alS. aureus, E. colideformation of the cell membrane[91]
CardamomElettaria cardamomumα-terpinly acetateE. coli, S. aureusdamage the cell membrane[91]
PeppermintMentha piperitamentholE. coli, S. aureuslysis and loss of membrane integrity[92]
AnisePimpinella anisumanetholeS. aureus, B. subtilisalter the cell membrane permeability[93]
Black pepperPiper nigrumα-pineneE. colileakage, disorder, and death by breaking cell membrane[94]
SageSalvia officinalisα-thujoneP. aeruginosachanged the cell membrane permeability[95]
LavenderLavandula angustifolialinaloolS. aureus, E. coli, C. albicansdamaging the cell wall and membrane[92]
MustardBrassica nigraallyl isothiocyanateA. fumigatus, A. nomiusdisrupt the cell wall thus causing cell lysis[96]
CitronCitrus medicalimoneneS. aureus, E. colidestruction of the cell membrane[97]
EucalyptusEucalyptus globulus1,8-cineoleE. coli, S. aureuspenetrate the membrane and damage cell organelles[98]
FennelFoeniculum vulgaretrans-anetholeS. aureus, E. colicell deformation and integrity of cell membranes[99]
Rose geraniumPelargonium roseumcitronellolS. salivariusinteraction with nitrogen in
proteins and nucleic acids
[100]
CarawayCarum carvicarvoneE. coli, B. bronchisepticaalteration in the structure of cell wall[101]
CorianderCoriandrum sativumlinaloolS. tyhimurium, E. colicell wall damage by over expression of genes[101]
TurmericCurcuma longaα-turmeroneS. aureusinducing leakage of ions and important cell contents[102]
PalmarosaCymbopogon martiniigeraniolB. subtillisalteration in cytoplasm and swelling[103]
DillAnethum graveolensα-phellandreneS. aureusdisrupt the permeability barrier[104]
ArmoiseArtemisia herba-albathujoneS. aureus, S. Typhimuriumchanging the membrane potential[105]
LaurelLaurus nobilis1,8-cineoleS. aureus. P. aeruginosadisrupt cellular membranes and increase membrane permeability[106]
GingerZingiber officinalezingibereneS. aureus, E. colidestroy membrane structure, increase cell membrane permeability[55]
CostmaryTanacetum balsamitaβ-thujoneL. monocytogenes, S. sonneidamage to the cellular membranes[107]
GuavaPsidium cattleianum Sabineα-pineneS. aureus, N. gonorrhoeaepropagate through cell membranes and cause the death[108]
MarjoramOriganum majoranaterpinen-4-olS. aureus, K. clocaeexhibited membrane
and DNA damaging effects
[109]
OreganoOriganum vulgarethymolS. aureus, S. entericaalteration of the bacterial plasma membrane[110]
Table 3. Antimicrobial activities of different metal and metal oxide nanomaterials against various pathogens and their respective mechanisms of action.
Table 3. Antimicrobial activities of different metal and metal oxide nanomaterials against various pathogens and their respective mechanisms of action.
NPsSize (nm)BacteriaModes of ActionRef.
Ag10V. natriegensrupture of cell membrane and DNA damage[132]
Ag2O10L. acidophilus, S. mutansprevents the growth of pathogen[133]
Ag2S65Phormidium spp.cell membrane inhibition[134]
Ag-MOF-S. aureusstable in water and the existence of Ag+ ions[135]
Al2O330S. typhi, F. oxysporumdisintegration of outer membrane by ROS[136]
Au20S. pneumoniaecellular disruption[137]
Bi40M. arginini, E. coliinhibits protein synthesis[138]
Cu15B. subtilis, S. aureussynergistic effects of functional groups[139]
CaO58S. aureus, E. colidestruction of the cell membrane[140]
CuO60B. cereusdamage of several biochemical processes[141]
CeO25B. cereus, E. colioxidative stress induced by the pro-oxidants[142]
CdS25S. aureus, Lactobacillus sp.CdS NPs impregnated and surrounded by the bacterial cell[115]
Fe474E. colistrong affinity between positively charged NPs and negatively charged cell membrane[143]
Fe3O425E. coli, S. aureusplasma membrane disruption[144]
FeS35E. coli, S. aureusinternalization of nanomaterials on cell membrane[145]
Ga305M. tuberculosisreduction of mycobacterium growth rate[146]
Mn50E. coli, S. aureusprotein inactivation and membrane permeability decreases[147]
MgO27E. coli, Bacillus sp.loss of membrane integrity and leakage of intracellular molecules[148]
Mn3O4130P. aeruginosa, K. pneumoniadisrupting bacterial cell membrane[149]
Mg-MOF-E. coli, S. aureuspeptide–nalidixic acid conjugation formed[150]
Mn-MOF-E. faecalis, P. aeruginosapeptide–nalidixic acid conjugation formed[150]
Ni60P. aeruginosadestruction of cell membrane[151]
NiO40E. coli, B. subtilisoxidative stress generated at the NPs interface resulted in membrane damage[152]
Pd13S. pyrogens, B. subtiliscell membrane damage and apoptosis[153]
Pt2A. hydrophila, E. coligeneration of ROS and decrease cell viability[154]
Se85S. aureus, E. coliROS causing cell membrane damage[155]
Si90P. aeruginosa, S. aureusdirect mechanical damage to the cell membrane[156]
TiO29.2E. coliouter cell membrane damaged by attacking hydroxyl radicals and ROS[157]
ZnO30A. baumanniiproduction of ROS increases[158]
ZrO22.5S. mitis, S. mutans, R. dentocariosaNPs enhance the interaction with bacterial constituents[159]
Zn-MOF-P. aeruginosacausing cell damage by interaction with hydroxyl group of peptidoglycan[160]
Ag/ZnO43P. aeruginosa, S. aureusleaching of silver as Ag+[161]
Au/CuS2B. anthraciscell membrane damage[162]
CuO/ZnO50 and 82S. aureus, E. colimembrane depolarization caused due to lectrostatic interaction of NPs[163]
Fe3O4/ZnO200E. coli, S. aureusplasm membrane disruption includes oxidative stress[164]
Au/Pt/Ag20E. faecalis, E. coliROS production[165]
Cu/Zn/Fe42E. coli, E. faecaliscell disruption by released ions[166]
Table 4. Antimicrobial activities of core–shell quantum dots against various pathogens with their respective mechanisms of action.
Table 4. Antimicrobial activities of core–shell quantum dots against various pathogens with their respective mechanisms of action.
NPsSize (nm)BacteriaModes of ActionRef.
ZnS and CdSe/ZnS quantum dot1.9E. coli, B. subtilistoxic composition of CdSe QDs demonstrating antimicrobial behavior[168]
CdSe/CdS/ZnS multi-core–shell quantum dots12–38K. pneumoniae, P. aeruginosarupturing of the membrane wall and cause of the decay of bacteria[169]
Ag-PdS/ZnS/CdS core–shell quantum dots8S. saprophyticus, E. coliestablishment of the catalyst–microorganism complex and a catalyst-related ROS[170]
ZnSe@ZnS core–shell quantum dots3.6 and 4.8E. coli, S. aureushigh affinity towards the thiol groups of bacterial cell surface proteins[171]
Peptide-loaded CdSe quantum dot9 and 14E. coli, S. aureusAP loaded on CdSe NPs had a higher water solubility and bioavailability[172]
P-doped carbon quantum dots2.75–4.25E. coli, S. aureuscell walls wrinkled and broken[173]
Ag@Ag2O core–shell19–60P. aeruginosa, S. aureusblockage of DNA replication and repair processes[174]
Table 5. Antimicrobial activities of graphene and CNTs against various pathogens and their corresponding mechanisms of action.
Table 5. Antimicrobial activities of graphene and CNTs against various pathogens and their corresponding mechanisms of action.
NPsSize (nm)BacteriaModes of ActionRef.
rGO-TiO232E. coli, S. aureusimprove the contact between TiO2 surface and bacteria[176]
GO-ZnO14–26E. coliinduces ROS to kill the bacteria[177]
GO-Cu2O30E. coli, S. aureuscopper ions react with cytoplasmic constituents[178]
DMS-GO-DMA--E. coli, S. aureusGO induces membrane stress on contact by disrupting and
damaging cell membranes
[179]
MWCNT-LVX--S. aureus, P. aeruginosainhibition of bacterial DNA replication[180]
F-MWNTs--E. coli, S. aureussmaller diameter of MWNTs can endorse damage to cell membrane through the cell–surface interaction[181]
Ag-doped ZnO on SWCNTs12–15E. coli, S. aureusproduction of ROS on the interaction samples with bacterial membrane[182]
Au-doped ZnO on MWCNTs12–18E. coli, S. aureusthe toxicity of carbon nanotube is mainly affected by diameter, length, and surface functional group[182]
Table 6. Antimicrobial activities of dendrimers and polymer composites against various pathogens with their mechanisms of action.
Table 6. Antimicrobial activities of dendrimers and polymer composites against various pathogens with their mechanisms of action.
NPsSize (nm)BacteriaModes of ActionRef.
Van-PAMAM-AgNP dendrimers--S. aureusheterofunctionalized Van-PAMAM-AgNP dendrimers for intra-cellular
entry through the cell wall and bacterial killing
[185]
G4-PAMAM dendrimer10E. coli, B. subtilisdisrupting of the cell membrane function and inhibiting cell wall synthesis, nucleic acid synthesis, and protein synthesis[187]
PAMAM-G7 dendrimer20P. mirabilis, S. aureusdendrimers are mediated by disrupting the bacterial outer and inner membrane by terminal amine
groups
[188]
Amino-acid-modified polycationic dendrimers--P. aeruginosaloss of membrane potential, inhibition of biosynthetic pathways, and free radical production[189]
Triclosan-loaded polymeric composite--S. aureus, K. pneumoniaeat high concentrations, triclosan destroys the bacterial membrane, leading to its death[190]
PBAT/Cu-NPs100–200A. baumannii, E. faecalispolymer and metal nanocomposites increase the number of ions released from the nanoparticles into the polymer matrix[191]
Piperazine polymer nanocomposite559.7E. coli, S. aureusnanoparticles are distributed within the suitable polymer matrix[192]
PVA/GO/Ag nanocomposites--E. coli, S. aureusphysical interactions of the bacterial cell with the nanoparticle[193]
Table 7. Antimicrobial activity of combinations of plant extract, EOs, antibiotics, and NPs against different pathogens.
Table 7. Antimicrobial activity of combinations of plant extract, EOs, antibiotics, and NPs against different pathogens.
Antimicrobial AgentsCombinationsPathogensRef.
EOs/EOsMelaleuca alternifolia/Cupressus sempervirensE. coli[195]
EOs/antibioticsEucalyptus globulus/oxacillinS. aureus[195]
EOs/NPsLemongrass/chitosan NPE. coli, S. aureus[197]
Plant extract/antibioticsSalvadora persica/amoxicillinP. gingivalis, T. forsythia[198]
Plant extract/EOsOriganum vulgare/carvacrolS. aureus[199]
Plant extract/NPsVatica diospyroides/Ag NPsS. aureus, B. subtilis[200]
NPs/antibioticsAgNPs/fluconazoleS. aureus, E. coli[201]
β-Lactam/β-lactamase inhibitoramoxicillin/potassium clavulanateS. aureus[202]
Table 8. Mode of action of different classes of antibiotics with their resistance profiles and target bacteria.
Table 8. Mode of action of different classes of antibiotics with their resistance profiles and target bacteria.
AntibioticsSpecific DrugModes of ActionResistance ProfilesTarget BacteriaRef.
β-LactamsPenicillin G, amoxicillin, cephalosporin CCell wall synthesis inhibitionHydrolysis, efflux, altered target, reduced permeabilityS. aureus, P. aeruginosa[212]
AminoglycosidesStreptomycin, gentamicinInhibition of translation and cell membrane synthesisModifying enzyme inactivation by phosphorylationP. aeruginosa, V. cholerae[213]
TetracyclinesMinocycline, doxycycline30S ribosomal subunitMonooxygenation, ribosomal modificationStaphylococci, Streptococci[214]
GlycopeptidesVancomycin, teicoplaninPeptidoglycan biosynthesisAltered targetS. haemolyticus, E. faecium[214]
MacrolidesErythromycin, azithromycinInhibition of protein synthesisGlycosylation, efflux, methylationStreptococci, Staphylococci[215]
PhenicolsChloramphenicolInhibition of protein synthesisAcetylation by chloramphenicol acetyltransferaseB. subtilis, S. pneumoniae[216]
RifamycinRifampinInhibition of nucleic acid synthesisADP-ribosylation, effluxV. cholerae, E. coli[213]
QuinoloneCiprofloxacin, levofloxacinInhibitors of DNA synthesisAltered DNA gyraseS. aureus, P. aeruginosa[212]
Cationic peptidesPolymyxin B, colistinDisrupt membranesAltered target, effluxE. coli, S. typhimurium[213]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Basavegowda, N.; Baek, K.-H. Combination Strategies of Different Antimicrobials: An Efficient and Alternative Tool for Pathogen Inactivation. Biomedicines 2022, 10, 2219. https://doi.org/10.3390/biomedicines10092219

AMA Style

Basavegowda N, Baek K-H. Combination Strategies of Different Antimicrobials: An Efficient and Alternative Tool for Pathogen Inactivation. Biomedicines. 2022; 10(9):2219. https://doi.org/10.3390/biomedicines10092219

Chicago/Turabian Style

Basavegowda, Nagaraj, and Kwang-Hyun Baek. 2022. "Combination Strategies of Different Antimicrobials: An Efficient and Alternative Tool for Pathogen Inactivation" Biomedicines 10, no. 9: 2219. https://doi.org/10.3390/biomedicines10092219

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

Article Metrics

Back to TopTop