Next Article in Journal
A Comparative Analysis on the Environmental Impact of Selected Methods for Determining the Profile of Fatty Acids in Cheese
Previous Article in Journal
An Anionic Porous Indium-Organic Framework with Nitrogen-Rich Linker for Efficient and Selective Removal of Trace Cationic Dyes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advances in Controllable Release Essential Oil Microcapsules and Their Promising Applications

School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, China
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(13), 4979; https://doi.org/10.3390/molecules28134979
Submission received: 7 April 2023 / Revised: 13 June 2023 / Accepted: 16 June 2023 / Published: 25 June 2023

Abstract

:
Essential oils (EOs) have emerged as natural and popular ingredients used in the preparation of safe and sustainable products because of their unique characteristics, such as antibacterial and antioxidant activity. However, due to their high volatility, poorly solubility in water, and susceptibility to degradation and oxidation, the application of EOs is greatly limited. One of the promising strategies for overcoming these restrictions is encapsulation, which involves in the entrapment of EOs inside biocompatible materials to utilize their controllable release and good bioavailability. In this review, the microencapsulation of the controllable release EOs and their applications are investigated. The focus is on the antimicrobial mechanism of various EOs on different bacteria and fungi, release mechanism of microencapsulated EOs, and preparation research progress of the controllable EOs microcapsules. In addition, their applications are introduced in relation to the food, textiles, agriculture, and medical fields.

1. Introduction

Essential oils (EOs) are secondary metabolites produced from plant tissues, such as flowers, buds, stems, leaves, roots, fruits, bark, resins, or wood, and they are mainly stored in secretory cells, epidermal cells, cavities, canals, or glandular trichomes of plants [1]. EOs are complex mixtures of odoriferous, volatile, and oily hydrophobic aromatic liquids. The main chemical composition includes monoterpenes, sesquiterpenes, diterpenes, and their oxygenated forms. Additionally, there are various phenols, aldehydes, ketones, esters, alcohols, and hydrocarbons present in EOs, such as eugenol [2], carvacrol [3], and limonene, [4] etc., which impart to them broad biological activity, such as antioxidant, antimicrobial, insecticidal, and therapeutic properties. Currently, approximately 3000 EOs have been established, 300 of which are especially important in the food, medicine, textile, cosmetics, and agricultural fields [5].
EOs are considered highly valuable rare substances owing to their low extraction yields (approximately 1%). Additionally, EOs are extremely unstable, and several external factors (e.g., oxygen, light, acidic or alkaline conditions, etc.) can lead to the degradation, transformation, or volatilization of EOs and reduce their biological activity [6]. Microencapsulation technology involves embedding active substances in wall materials, protecting core materials from environmental influence and denaturation, and achieving controllable release or stable storage of active substances [7]. Thus, the microencapsulation of EOs is very crucial to protecting them from the effects of adverse conditions and ensure their quality. Usually, microcapsules are formed by natural or synthetic polymer materials through various interactions during the preparation process, such as ion interaction, electrostatic interaction, hydrophilic hydrophobic interaction, covalent bonds, and hydrogen bonds. According to the preparation principles, the preparation methods of microcapsules can be divided into physical methods (e.g., interfacial polymerization, inclusion complexation, ionic gelation, coacervation, etc.) and chemical methods (e.g., spray-drying method, lyophilization, emulsification method, oil/water method, ultrasonic method, solvent evaporation method, phase dispersion coacervation method, layer-by-layer assembly method, layer-by-layer desolvation method, etc.) [8,9]. Generally, more than one of the above methods are often combined to prepare the expected microcapsules. Appropriate preparation methods can improve the encapsulation efficiency of EOs and reduce economic consumption. Many authors in previous investigations have confirmed the strong influences of encapsulation methods, total solid content, emulsions properties, and interactions between EOs and wall materials on encapsulation efficiency [10,11]. According to the research of Mehran et al. [12] and Jafari et al. [13], encapsulation efficiency increases with an increase in total content of wall materials. The results can be related to the increase of emulsion viscosity. In addition, the higher temperature of spray drying has a negative effect on encapsulation efficiency. This outcome can be attributed to the breakdown of the microcapsule wall at higher temperatures, consequently resulting in the release of EOs. In this review, we summarize the antimicrobial mechanism of various EOs on different bacteria and fungi as well as the release mechanism and preparation research progress of controllable EOs microcapsules with various natural and stimuli-responsive substances as wall materials. In addition, the applications of controllable release EOs microcapsules are introduced into the fields of food, textiles, agriculture, and medicine (Figure 1).

2. Antimicrobial Mechanism of EOs

Most EOs have a wide spectrum of antimicrobial activity against foodborne pathogenic and spoilage microorganisms, and the antimicrobial capacity is largely dependent on their chemical constituent and cell type. For example, trans-cinnamaldehyde is one of the major compounds of EOs and has the proficiency to inhibit the growth of E. coli by depleting intracellular ATP levels, reducing bacterial cell division protein FtsZ expression, GTP-dependent polymerization, and GTP hydrolysis [14]. Phenolic compounds, such as carvacrol, eugenol, and thymol, can effectively inhibit the growth of microorganisms by disrupting cell membranes, leading to alteration in electron flow, driving force of protons and active transport, and coagulation of cell contents [15,16]. Additionally, thymol is also able to inhibit the expression of thioredoxin-1 and is effective against several vital enzyme pathways and metabolites, such as the phosphotransferase system, the efflux system, and energy metabolism. Monoterpenes target membrane-embedded proteins, membrane fluidity–permeability mechanisms, and ion transport processes [17]. Carvone is another important EO constituent which can disrupt the outer membranes of the cells but does not affect cellular ATP pools [18]. In general, the major compounds are largely responsible for antibacterial activity of different EOs, and this activity is attributed to the free hydroxyl groups of these compounds. EOs containing aldehyde or phenolic groups, such as cinnamaldehyde, linanaldehyde, eugenol, and thymol, have shown the highest antibacterial activity, followed by EOs containing terpenes (e.g., β-myrcene, α-thujone, etc.), which exhibit weaker antibacterial effects [19]. Other EOs containing ketones or esters have much weaker activity, and EOs containing terpene hydrocarbons are usually inactive [20,21].
Owing to the different compositions and thicknesses of cell membranes, EOs display varying resistances against different types of microorganisms. Most components (90–95%) in the cell wall of gram-positive bacteria are peptidoglycan, which allows easy diffusion of hydrophobic components, such as EOs, into cells and can directly damage cell membranes, block enzyme systems, and increase ion permeability. In contrast, the peptidoglycan layer is thinner for the cell walls of gram-negative bacteria, which are covered by outer membranes that consisting of double-layer phospholipids. The hydrophilic transmembrane channels at the surface of the outer membrane leads to the easy penetration of hydrophilic substances, while the hydrophobic molecules, such as EOs, slowly traverse the outer membrane through porins due to the interaction between EOs and the lipopolysaccharide layer [22,23]. Therefore, the antibacterial effects of EOs against gram-positive bacteria are reported to be higher than against gram-negative bacteria. For example, cunila EOs display more pronounced antimicrobial activity against gram-positive bacteria than against gram-negative bacteria [24]. However, eugenol and isoeugenol are more active against gram-negative bacteria compared to gram-positive bacteria [25].
EOs can easily penetrate the lipid layer of the cell wall and disrupt the cell membrane’s structure, thereby exerting multiple antibacterial mechanisms through action on intra-cellular proteins, enzymes, DNA, metabolism, energy production, etc. In this review, the inhibitory mechanisms of different EOs on various bacteria (Table 1) and fungus (Table 2) are revealed from the aspects of the biomacromolecules, such as DNA, proteins, and lipids; the cell wall or cell membrane integrity; the metabolic pathway; and the expression of related virulence genes.

3. Preparation of Controllable-Release EOs Microcapsules

Currently, in terms of wall materials, EO microcapsules include single-layer microcapsules, multilayer microcapsules, and stimuli-responsive microcapsules. Generally speaking, the release of microcapsules can be divided into two stages, namely immediate triggered release, which is controlled by the start of the release system, and sustained release, which is mainly the continuous and one-time diffusion of EOs from the polymer shell before and after its rupture [50]. Different release mechanisms of the microencapsulated EOs have diffusion, fragmentation, polymer relaxation (shrinking/swelling), and erosion, which are substantially related to the type of wall materials and encapsulation method [51]. Additionally, it is very significant for the application of microcapsules. Table 3 summarizes the wall material types, microcapsule methods, release models, and release mechanisms of different EOs microcapsules.

3.1. Single-Layer Microcapsules

A wide range of substances, including polysaccharides (e.g., gum arabic, persian gum, maltodextrin, cyclodextrin, hydroxypropyl-β-cyclodextrin, inulin, modified starch, pectin, sodium alginate, and chitosan, etc.), proteins (e.g., gelatin, whey protein, mung bean isolate protein, rice protein, zein, etc.), lipids, poly (D, L-lactide-co-glycolide) (e.g., polyvinyl alcohol, polyurethane, etc.), and silica-biocompatible materials are some of the wall materials used in the food and medical fields. Especially for plant-derived proteins/polysaccharides, all exhibit good biocompatibility, biodegradability, bioadhesion, and non-toxicity and have been widely applied for food processing due to their advantages of lower cost, higher sustainability, and versatility [63,64]. Dima et al. [52] has prepared coriander EO microcapsules by spray-drying with chitosan, alginate, chitosan/alginate, or chitosan/inulin as wall materials. The release kinetics show that the swelling degree and release rate of chitosan microcapsules are the highest at pH 2.5, and that those of alginate microcapsules are the highest at pH 6.5 in the simulated human gastrointestinal digestive system. However, they are all resistant to pH and temperature variations and can ensure a slow release of coriander EO. Das et al. [65] and Hasheminejad et al. [66] have encapsulated Coriandrum sativum EO and clove EO with chitosan-based nanomatrix, which is able to improve the shelf life of stored rice and pomegranate arils and is obviously better than free EOs. However, owing to the different polarities of EOs and chitosan, the binding force is low and the chitosan-containing EOs rapidly lose their antimicrobial properties, resulting in insufficient efficacy [67]. Bajac et al. [54] encapsulates juniper berry EO by spray-drying with gum arabic, maltodextrin, sodium alginate, or whey protein concentrate as wall materials. The release kinetics indicate that the release time and speed of the microcapsules formed by sodium alginate (almost all release within 15 min) are faster than those of gum arabic (almost all release within 30 min). However, using a combination of maltodextrin and gum arabic as wall materials allows the slowest release from microparticles, and it further prolongs the complete release of EOs to about 45 min. An antimicrobial sachet releasing vapor of oregano EO is developed by spray-drying using polyvinyl alcohol as wall material, and release of oregano EO occurs under conditions of high humidity and room temperature [68]. Traditional single-layer encapsulation has the disadvantages of poor mechanical and barrier properties. Therefore, multilayer encapsulation has attracted considerable attention in recent years.

3.2. Multilayer Microcapsules

Multilayer encapsulation is a method of achieving sustained release of EOs using multiple wall materials, which prepares by “layer-by-layer” (LBL) self-assembly technology, involving electrostatic deposition between wall materials with opposite charges and spontaneous crosslinking to form a polyelectrolyte layer. Multilayer encapsulation uses EOs as the intermediate or inner layer, which can effectively improve slow release and retention rate of EOs [69]. LBL technology mainly focuses on the homogeneous emulsification of mixtures. The preparation process should be such that the instability caused by bridging, consumption, flocculation, or coalescence of the substrate is avoided. Simultaneously, the size distribution, shell composition, shell porosity, and diffusion of the condensed layer affect EO release [70]. Composite coacervation has become the most commonly used multilayer encapsulation method owing to its high stability and loading rate, which is a polymerization phenomenon initiated by two biopolymers with opposite charges—generally proteins (amino) and polysaccharides (sugar carboxyl)—through electrostatic forces, hydrogen bonds, or hydrophobic forces under strict control of process parameters such as pH, temperature, ionic strength, stirring rate, polymer ratio, concentration, and molecular weight [71].
Qiu et al. [64] prepares double-layer microcapsules containing mung bean isolated protein/apricot peel pectin/rose EO. The microcapsules release rose EO rapidly for the first 9 min and then release rose EO continuously over the next 26 min. β-Lactoglobulin/sodium alginate/black pepper EO microcapsules give similar results and are very stable in the in vitro digestion (oral and gastric) stage, in which 65.5% of EO is transported to intestine. Mehran et al. [12] used a spray-drying method to prepare inulin/gum arabic/peppermint EO microcapsules. The system explosively releases EO within 7 min of the start, and then the release rate decreases successively in 50% ethanol, 10% ethanol, 3% acetic acid, and distilled water. The Peppas–Sahlin model (Fickian diffusion) is the best kinetic model for describing the release of peppermint EO. Cyclodextrins have hydrophilic surfaces and hydrophobic cavities. Natural cyclodextrins, including α-, β-, and γ-cyclodextrins, are widely used because of their suitable cavity sizes, high retention rates, and low price. Compared to β-cyclodextrin, hydroxypropyl-β-cyclodextrin has higher water solubility and lower toxicity and has been approved by the FDA for use as food additives. Hou et al. [72] prepared a hydroxypropyl-β-cyclodextrin/cinnamon EO nanoemulsion which can protect EOs and enable sustained release to exert long-term antibacterial effects. An EO viscous core microcapsule is formulated by encapsulating EOs with intermediate non-crosslinked polymer shells and crosslinked polymer outer shells followed by dissolution of the intermediate shells with EOs to form viscous cores. The study found that the microcapsule has better heat resistance, extreme long-lasting release property, and viscoelasticity and that it is more adhesive to cotton fabrics, which is more consumer-friendly compared to melamine-formaldehyde microcapsules [58]. According to the research of Noghabi et al. [59], maltodextrin is used in combination with Persian gum to achieve proper the encapsulation of cinnamon EO by spray drying. Maltodextrin as secondary wall material protects cinnamon EO against oxidation appropriately and offers low viscosity at high solids concentration. Sun et al. [73] has used porous starch as a core material carrier to adsorb fennel EO and sodium alginate/chitosan as wall materials to successfully prepare sodium alginate/chitosan/porous starch–fennel EO microcapsules using a polyelectrolyte composite coagulation method. In the open and closed systems, the cumulative release rates of fennel EO in 16 days are 70.62% and 43.87%, respectively, with a good slow-release effect. Basil EO microcapsules are successfully produced with different wall materials, including gum arabic, maltodextrin, and whey protein isolate. Results show that the ternary combination has a high product yield, encapsulation efficiency, and eugenol release rate in ethanol [74]. A star anise EO microcapsule was produced from rice protein and depolymerized pectin using electrostatic complexation and exhibits a sustained release and good antibacterial effect [75]. Compared to single-layer encapsulation, multilayer microcapsules have higher stability and resistance to environmental strains, such as oxidation, thermal change, and ionic strength, which can effectively improve the physical and chemical stability of EOs and allow the control and triggering of the retention and release characteristics of EOs, achieving continuous, high-content, and uniform release [76].

3.3. Stimuli-Responsive Microcapsules

With the continuous development of microencapsulation technology, the function of embedding and protecting core materials of traditional microencapsuation can no longer meet the needs of industry. Recently, more and more scholars have paid attention to targeted, controllable, triggered, and other special functional releases. Stimuli-responsive microcapsules have drawn great attention in various fields and are involved in the permeability or rupture changes of wall materials under specific environmental conditions, such as temperature, humidity, pH, light, enzymes, magnetic fields, etc., which cause the EOs to be released. These environmental-stimuli-responsive microcapsules have the advantages of rapid response, large internal capacity, and stable capsule membrane [77]. EOs can maintain high activity for a given time and continuously produce activity where it is needed. Chitosan, poly (D, L-lactide-co-glycolide), protein fragment (gelatin), and silica biocompatible materials are some of the wall materials used in the stimuli-responsive microcapsules field [78]. Xu et al. [57] has established a triggering system of core-shell structure, oregano EO loaded microcapsules using chitosan-decorated TiO2 (a UV-light-sensitive shell material) as wall materials, which can be used to control the release by UV irradiation depending on specific needs and can be applied in the food industry. Compared with single-shell or single-stimulated microcapsules, the controllable release of core substances from double-shell microcapsules is more durable. The outer shell not only provides a good and stable environment for the inner layer, but the design of double-shell layer structure can also make the layers independent from each other, and the improvement of different layers can provide multiple functions to the microcapsules. Zhu et al. [55] prepared a bio-based, material-modified polylactic acid/octenyl succinic anhydride–chitosan/tea tree EO microcapsule with a pH-responsive function and antibacterial properties via the double emulsion method and the solvent evaporation method, which can accelerate the release at pH = 5 and maintain 100% antibacterial efficiency after 72 h. A new double-shell lignin microcapsule containing lemon EO was successfully fabricated using the interfacial polymerization and free radical polymerization methods. Lemon EO is further located at the core of the microcapsule and is released by thermal response [60]. As natural biocompatible polysaccharides, anionic sodium alginate and cationic chitosan can be widely used to prepare various drug delivery systems owing to their pH sensitivity and enzymatic degradability, which can achieve the selected release of EOs in a specific environment [79].

4. Application of Controllable Release EOs Microcapsules

4.1. Food Industry

Foodborne spoilage and pathogenic bacteria are major concerns contributing to food quality and safety, with foodborne diseases constituting a global health problem. Chemical antisepsis is an important method adopted by the food industry to control foodborne microorganisms and prolong food shelf life. However, in recent years, owing to food safety accidents caused by synthetic preservatives and higher requirements for food quality and flavor, the development of natural, environmental, and healthy preservatives has become a new trend. EOs derived from plants have become a promising natural antiseptic candidate owing to their powerful and broad-spectrum antibacterial activity. An appropriate amount of EOs does not affect the flavor of the product and increases the appetite of consumers to a certain extent. Additionally, EOs have been generally recognized as safe (GRAS), and some are approved by the U.S. Food and Drug Administration (FDA) to be used as food additives, such as lemon, basil, coriander, clove, thyme, and vanilla EO. Currently, approximately 3000 of EOs are known, and some of them are commercially important, being employed in the agronomic, cosmetic, chemical, perfume, fragrance, pharmaceutical, and food industries [80,81].
Research indicates that the interaction of polysaccharides and proteins could improve the stability of protein nanoparticles to the environment. Cai et al. [82] prepared a multilayer encapsulated gliadin/glycyrrhizic polysaccharide/tea tree EO nanoparticle. The inhibition rates of the EOs nanoparticles on Salmonella typhimurium on the surface of pork and chicken products were 98.52% and 97.86%, respectively. At the same time, the EO nanoparticles delayed lipid peroxidation of meat products, and the quality and color of meat products did not change significantly within the 5-day storage period. Recently, various microcapsules loaded with EOs have been used for active film/coating applications. For example, a microcapsule loaded with oregano EO via ionic gelation has been developed. Sodium alginate is used as the wall material, with calcium chloride as a crosslinking agent. Compared to soy protein concentrate films with free oregano EO, incorporating the oregano EO microcapsule significantly improves the composite film’s antimicrobial ability against E. coli and S. aureus [83]. Additionally, EOs can be used to develop biodegradable active packaging, which is an interesting substitute in the food packaging industry. To improve the slow-release effect of cinnamon EO, β-cyclodextrin/cinnamon EO microcapsules were firstly prepared by Shao et al. [84]. Continuously, the starch/cinnamon EO microcapsules were used as coating for the paper to prepare bioactive packaging materials, which had remarkable bacteriostatic effects on mesophilic, psychrophilic, pseudomonad, yeast, and mold counts. This can effectively prolong the storage life and ensure the safety of mushrooms. Modified atmosphere packaging is considered a good method to help maintain post-harvest quality and prolong the storage time of fruit. Li et al. [85] prepared the 3D printing of lemongrass EO/β-cyclodextrin/popping candy modified atmosphere packaging for strawberry preservation. Lemongrass EO was successfully encapsulated in β-cyclodextrin and had a strong bacteriostatic effect on Aspergillus niger and Botrytis cinerea. Simultaneously, the popping candy can release CO2 and extend the shelf life of strawberries. Tian et al. [86] firstly reported the preparation of sustained-release tea tree EO solid preservative based on polysaccharide. The hydrogen bond interaction of the starch–carboxymethyl cellulose network structure and the hydrophobic interaction of SiO2-adsorbing tea tree EO resulted in a compact structure and excellent sustained-release performance of the solid preservative, which improved sensory quality and reduced nutrient loss and microbial spoilage (molds and yeasts) in modified atmosphere packaging. Biodegradable and effective starch-based anti-bacterial film packagings have been successfully made by incorporating polyurethane-encapsulated EO microcapsules, including tree EO microcapsules, lavender EO microcapsules, and perilla leaf EO microcapsules as alternative synthetic preservatives for food preservation [87].

4.2. Textiles

Textiles are one of the most important necessities in daily life. With the increasing demand of consumers for disinfected and easy-care textiles, and the continuous improvement of people’s health awareness, the production of multifunctional medical natural textiles with antibacterial, antioxidant, insect-resistant, and sanitary properties has become a primary demand of customers. Owing to the adverse effects of many synthetic antibacterial agents and insect repellents on the environment and human health, EOs are being used as natural antibacterials and insect repellents due to their extensive biological activities in textile production [88]. Microencapsulation technology not only prevents EOs from leaking but also extends the antibacterial time and service life of the fabric coatings, and the release of the core material is controllable. A chitosan/gelatin/rosemary EO microcapsule has been utilized for the development of multifunctional and efficient linen fabric with a dip-rolling drying method. When the encapsulation efficiency of rosemary EO exceeds 70%, the multifunctional textile has long-lasting mosquito repellent ability (>90%), inhibition activity against E. coli (>93%) and S. aureus (>95%), significant anti-oxidant ability (>91%), and a pleasant aroma [56]. A gum arabic/citronella EO microcapsule prepared using a two-step method of emulsification and spray-drying has the advantages of prolonging the volatility and reducing the irritation of EOs, which can be dipped on non-woven fabrics for the preparation of cosmetic textiles [89]. A gelatin/gum arabic/citronella EO microcapsule is prepared through composite coagulation and spray-drying, and then they dip the microcapsules slurry into cotton fabric to prepare insect repellent textiles with the traditional dip-rolling drying method. The insect repellent effect within three weeks is more than 90%, which is a durable multifunctional textile [90]. Liu et al. [91] utilized chitosan–gelatin as a shell material to prepare microcapsules containing patchouli oil via the complex coacervation method, and the prepared microcapsules were grafted onto cotton fabric. The developed fabric displays 65% antibacterial activity for S. aureus and E. coli after 25 washes. Chen et al. [92] proposed a facile and environmentally friendly method for the preparation of aqueous multifunctional fabric coating using cellulose/silica hybrid microcapsules. They first synthesized lavender-fragrance-oil-loaded (LFO) cellulose/silica hybrid microcapsules containing EOs in one step through emulsion solvent diffusion and then added aqueous polysiloxane resins to prepare multifunctional fabric coating. The obtained fabric coating allows for pH-controllable stimulation of the release of LFO. The fabrics treated with the coating retained more than 30% of their fragrance after 90 days and exhibited excellent UV resistance. Exploring new combinations of biopolymers and EOs microcapsules for the development of multifunctional textiles is a broad prospect.

4.3. Agriculture Field

Chemical pesticides are severely restricted owing to serious negative consequences, such as soil and water contamination, high persistence in the environment and as residues in foods, and toxicity to people and animals. Therefore, plant pesticides—as effective, natural, and ecological insecticidal agents—can be developed and applied for integrated pest control. In general, EOs and their constituents can exhibit strong pesticide potential against insect pests, weeds, and fungi. Reports show that the neurotoxic effects of EOs can be prevented by blocking the octopamine receptors or by acetyl-cholinesterase inhibition and also through blocking the γ-aminobutyric acid receptor [93]. A gum arabic/maltodextrin/Schinus molle EO microcapsule has been employed to control the release of EO, which not only protects them from the external environment but improves insecticidal potential on Haematobia irritans. Compared to free EOs (96% mortality rate of flies within 2 h), the microcapsules have a slow-release effect (71% of EOs retention within 366 h) and a lasting insecticidal effect (32% and 73% mortality rate of flies within 2 h and 4 h, respectively) [94]. Similarly, Ahsaei et al. [95] prepared octenyl succinic anhydride–starch/Rosmarinus officinalis or Zataria multiflora EOs microcapsules using the spray-drying method, which displayed a more effective and long-term fight against Tribolium confusum than free EOs. Free EOs had no insecticidal activity after 15 days of storage, whereas the insecticidal rates of EOs microcapsules in the same period were 46.6% and 35.5%, respectively, indicating that microencapsulation increases the insecticidal persistence of EOs. Lv et al. [96] developed a novel, efficient, and safe antimildew for peanut kernel postharvest storage. The antimildew, cinnamon–Litsea cubeba compound essential oil (CLCEO) microcapsule was synthesized with CLCEO as the core material, and β-cyclodextrin was used as the wall material. CLCEO microcapsules can effectively reduce the total number of fungal colonies, the relative abundance of Aspergillus spp., and the atoxin B1 content of peanut kernels and have a positive effect on slowing down the increase in the acid value of peanut oil without causing any adverse effect on the viability and sensory properties during storage process. Thus, plant EOs have been explored as low-risk insecticides, some of which are classified under the Generally Recognized as Safe (GRAS) category and meet the criteria of reduced-risk pesticides [97].

4.4. Medical Field

EOs possess a number of beneficial biological properties, such as antioxidant, antimicrobial, anti-proliferative, antiulcer, anti-inflammatory, and anticancer activities. Reports show that the absorption of EOs mainly occurs in the distal small intestine rather than the stomach and proximal small intestine. Excessive and early release of EOs in the stomach has adverse side effects on human health, such as nausea, dizziness, and accelerated heartbeat. In this respect, microencapsulation may be an approach to achieving the protection and controllable release of EOs. Beeswax is a gastro-resistant material which is frequently used in the preparation of controllable release drug delivery systems. Beeswax microcapsules retard the release of water-insoluble drugs in the simulated gastric solution at pH 1.2 due to their highly crystalline structure and highly hydrophobic nature. A beeswax/carboxymethyl cellulose/gelatin holy basil EOs microcapsule was prepared using a simple coacervation method by Ngamekaue et al. [98]. It exposed the minimized holy basil EOs in simulated gastric fluids and delivered the EOs to the distal small intestine, displaying effective antioxidant and antibacterial activities. As natural biocompatible polysaccharides, anionic sodium alginate and cationic chitosan can be widely used to prepare various drug delivery systems owing to their pH sensitivity and enzymatic degradability, which can achieve the selected release of EOs based on the gastrointestinal microenvironment [79]. For example, a porous alginate/chitosan/basil oil microcapsule which retains the antioxidant and antimicrobial activities of basil oil under storage, in addition to a great tolerance to acids, bile, trypsin, and thermal conditions, was prepared [99]. However, the microencapsulation technique for EOs with effective and efficient delivery in animal’s bodies is a point to be further developed.

5. Conclusions and Prospect

EOs have been recognized as important natural sources with a widespread range of bioactivity. Considerable research reports the great prospects for EOs as bioactive ingredients in the food industry, pharmaceutical cosmetics, and botanical pesticide production. However, conventional formulation procedure will diminish EOs’ efficiency and limit their uses in practice owing to sensitivity. Thus, loading them in protective biopolymer materials will offer controllable release and delay their fast evaporation and degradation. In this review, we summarize the antimicrobial mechanism, the release mechanism of microencapsulated EOs, and the various natural and stimuli-responsive materials used for the fabrication of EOs microcapsules. In addition, the applications of controllable release EOs microcapsules are introduced in relation to the food, textile, agriculture, and medical fields.
However, the key technology now lies in manufacturing intelligent microcapsules with multiple reactions and compartments which can simultaneously respond to a wide range of stimuli, truly achieving intelligence. Additionally, attention should also be paid to enhancing the retention of active ingredients in microcapsules, especially when the microcapsules are stimulated by external environments. Future research may also use several innovative techniques to develop novel sustained EO release and retention enhancement strategies in microcapsules, such as low-temperature plasma, pulsed light, electrostatic self-assembly techniques, radiation, and electrospinning and electrospraying technologies. In addition, given the rising interest in EOs, consumer health concerns must be considered an urgent topic to be addressed. Although several organizations have collaborated with governments to implement the regulation of EOs worldwide, strong national and international regulations should be established before applying EOs microcapsules in various fields.

Author Contributions

The manuscript was written by Y.Z. Data statistics were completed by Y.W. Literature organization was provided by H.L. Critical review of the manuscript was performed by Z.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Scientific and Technological Innovation Programs of Higher Education Institutions in Shanxi, grant number 20200L0287.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Conflicts of Interest

There are no conflict of interest to declare.

References

  1. Bakkali, F.; Averbeck, S.; Averbeck, D.; Idaomar, M. Biological effects of essential oils—A review. Food Chem. Toxicol. 2008, 46, 446–475. [Google Scholar] [CrossRef]
  2. Bonilla, J.; Poloni, T.; Lourenço, R.V.; Sobral, P.J.A. Antioxidant potential of eugenol and ginger essential oils with gelatin/chitosan films. Food Biosci. 2018, 23, 107–114. [Google Scholar] [CrossRef]
  3. Pierini, G.D.; Bortolato, S.A.; Robledo, S.N.; Alcaraz, M.R.; Fernández, H.; Goicoechea, H.C.; Zon, M.A. Second-order electrochemical data generation to quantify carvacrol in oregano essential oils. Food Chem. 2022, 368, 130840. [Google Scholar] [CrossRef] [PubMed]
  4. Corrêa, A.N.R.; Weimer, P.; Rossi, R.C.; Hoffmann, J.F.; Koester, L.S.; Suyenaga, E.S.; Ferreira, C.D. Lime and orange essential oils and d-limonene as a potential COVID-19 inhibitor: Computational, in chemico, and cytotoxicity analysis. Food Biosci. 2023, 51, 102348. [Google Scholar] [CrossRef] [PubMed]
  5. Tariq, S.; Wani, S.; Rasool, W.; Shafi, K.; Bhat, M.A.; Prabhakar, A.; Shalla, A.H.; Rather, M.A. A comprehensive review of the antibacterial, antifungal and antiviral potential of essential oils and their chemical constituents against drug-resistant microbial pathogens. Microb. Pathog. 2019, 134, 103580. [Google Scholar] [CrossRef]
  6. Asbahani, A.E.; Miladi, K.; Badri, W.; Sala, M.; Addi, E.H.A.; Casaniana, H.; Mousadik, A.E.; Hartmann, D.; Jilale, A.; Renaud, F.N.R.; et al. Essential oils: From extraction to encapsulation. Int. J. Pharm. 2015, 483, 220–243. [Google Scholar] [CrossRef] [PubMed]
  7. Kouamé, K.J.E.; Bora, A.F.M.; Li, X.; Sun, Y.; Liu, L. Novel trends and opportunities for microencapsulation of flaxseed oil in foods: A review. J. Funct. Foods 2021, 87, 104812. [Google Scholar] [CrossRef]
  8. Meng, Q.; Zhong, S.; Gao, Y.; Cui, X. Advances in polysaccharide-based nano/microcapsules for biomedical applications: A review. Int. J. Biol. Macromol. 2022, 220, 878–891. [Google Scholar] [CrossRef] [PubMed]
  9. Wang, Y.; Ye, A.; Hou, Y.; Jin, Y.; Xu, X.; Han, J.; Liu, W. Microcapsule delivery systems of functional ingredients in infant formulae: Research progress, technology, and feasible application of liposomes. Trends Food Sci. Tech. 2022, 119, 36–44. [Google Scholar] [CrossRef]
  10. Álvarez-Henao, M.V.; Saavedra, N.; Medina, S.; Cartagena, C.J.; Alzate, L.M.; Londonõ-Londonõ, J. Microencapsulation of lutein by spray-drying: Characterization and stability analyses to promote its use as a functional ingredient. Food Chem. 2018, 256, 181–187. [Google Scholar] [CrossRef]
  11. Baysal, G.; Olcay, H.S.; Lumberjack, B.; Özpinar, H. The antioxidant and antibacterial properties of chitosan encapsulated with the bee pollen and the apple cider vinegar. J. Biomater. Sci. Polym. Ed. 2022, 33, 995–1011. [Google Scholar] [CrossRef]
  12. Mehran, M.; Masoum, S.; Memarzadeh, M. Microencapsulation of Mentha spicata essential oil by spray drying: Optimization, characterization, release kinetics of essential oil from microcapsules in food models. Ind. Crops Prod. 2020, 154, 112694. [Google Scholar] [CrossRef]
  13. Jafari, S.M.; Assadpoor, E.; Bhandari, B.; He, Y. Nano-particle encapsulation of fish oil by spray drying. Food Res. Int. 2008, 41, 172–183. [Google Scholar] [CrossRef]
  14. Domadia, P.; Swarup, S.; Bhunia, A.; Sivaraman, J.; Dasgupta, D. Inhibition of bacterial cell division protein FtsZ by cinnamaldehyde. Biochem. Pharmacol. 2007, 74, 831–840. [Google Scholar] [CrossRef] [PubMed]
  15. Bhavaniramya, S.; Vishnupriya, S.; Al-Aboody, M.S.; Vijayakumar, R.; Baskaran, D. Role of essential oils in food safety: Antimicrobial and antioxidant applications. Grain Oil Sci. Technol. 2019, 2, 49–55. [Google Scholar] [CrossRef]
  16. Lima, V.N.; Oliveira-Tintino, C.D.M.; Santos, E.S.; Morais, L.P.; Tintino, S.R.; Freitas, T.S.; Geraldo, Y.S.; Pereira, R.L.S.; Cruz, R.P.; Menezes, I.R.A. Antimicrobial and enhancement of the antibiotic activity by phenolic compounds: Gallic acid, caffeic acid and pyrogallol. Microb. Pathog. 2016, 99, 56–61. [Google Scholar] [CrossRef]
  17. Swain, S.S.; Paidesetty, S.K.; Padhy, R.N.; Hussain, T. Nano-technology platforms to increase the antibacterial drug suitability of essential oils: A drug prospective assessment. OpenNano 2023, 9, 100115. [Google Scholar] [CrossRef]
  18. Pauli, A. Antimicrobial properties of essential oil constituents. Int. J. Aromather. 2001, 11, 126–133. [Google Scholar] [CrossRef]
  19. Zhu, Y.; Li, C.; Cui, H.; Lin, L. Encapsulation strategies to enhance the antibacterial properties of essential oils in food system. Food Control 2021, 123, 107856. [Google Scholar] [CrossRef]
  20. Delaquis, P.J.; Stanich, K.; Girard, B.; Mazza, G. Antimicrobial activity of individual and mixed fractions of dill, cilantro, coriander and eucalyptus essential oils. Int. J. Food Microbiol. 2002, 74, 101–109. [Google Scholar] [CrossRef]
  21. Ju, J.; Chen, X.; Xie, Y.; Yu, H.; Guo, Y.; Cheng, Y.; Qian, H.; Yao, W. Application of essential oil as a sustained release preparation in food packaging. Trends Food Sci. Tech. 2019, 92, 22–32. [Google Scholar] [CrossRef]
  22. Semeniuc, C.A.; Pop, C.R.; Rotar, A.M. Antibacterial activity and interactions of plant essential oil combinations against Gram-positive and Gram-negative bacteria. J. Food Drug Anal. 2017, 25, 403–408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Saei-Dehkordi, S.S.; Tajik, H.; Moradi, M.; Khalighi-Sigaroodi, F. Chemical composition of essential oils in Zataria multiflora Boiss. from different parts of Iran and their radical scavenging and antimicrobial activity. Food Chem. Toxicol. 2010, 48, 1562–1567. [Google Scholar] [CrossRef]
  24. Sandri, G.; Zacaria, J.; Fracaro, F.; Delamare, A.P.L.; Echeverri-garay, S. Antimicrobial activity of the essential oils of Brazilian species of the genus Cunila against foodborne pathogens and spoiling bacteria. Food Chem. 2007, 103, 823. [Google Scholar] [CrossRef]
  25. Meenua, M.; Padhanb, B.; Patel, M.; Patel, R.; Xu, B. Antibacterial activity of essential oils from different parts of plants against Salmonella and Listeria spp. Food Chem. 2023, 404, 134723. [Google Scholar] [CrossRef] [PubMed]
  26. Li, J.; Li, C.; Shi, C.; Aliakbarlu, J.; Cui, H.; Lin, L. Antibacterial mechanisms of clove essential oil against Staphylococcus aureus and its application in pork. Int. J. Food Microbiol. 2022, 380, 109864. [Google Scholar] [CrossRef] [PubMed]
  27. Costa, W.K.; Oliveira, A.M.; Santos, I.B.S.; Silva, V.B.G.; Silva, E.K.C.; Alves, J.V.O.; Silva, A.P.S.; Lima, V.L.M.; Correia, M.T.S.; Silva, M.V. Antibacterial mechanism of Eugenia stipitata McVaugh essential oil and synergistic effect against Staphylococcus aureus. S. Afr. J. Bot. 2022, 147, 724–730. [Google Scholar] [CrossRef]
  28. He, Q.; Zhang, L.; Yang, Z.; Ding, T.; Ye, X.; Liu, D.; Guo, M. Antibacterial mechanisms of thyme essential oil nanoemulsions against Escherichia coli O157:H7 and Staphylococcus aureus: Alterations in membrane compositions and characteristics. Innov. Food Sci. Emerg. 2022, 75, 102902. [Google Scholar] [CrossRef]
  29. Cui, H.; Bai, M.; Sun, Y.; Abdel-Samie, M.A.; Lin, L. Antibacterial activity and mechanism of Chuzhou chrysanthemum essential oil. J. Funct. Foods 2018, 48, 159–166. [Google Scholar] [CrossRef]
  30. Wang, F.; Wei, F.; Song, C.; Jiang, B.; Tian, S.; Yi, J.; Yu, C.; Song, Z.; Sun, L.; Bao, Y.; et al. Dodartia orientalis L. essential oil exerts antibacterial activity by mechanisms of disrupting cell structure and resisting biofilm. Ind. Crops Prod. 2017, 109, 358–366. [Google Scholar] [CrossRef]
  31. Liu, Q.; Gao, Y.; Fu, X.; Chen, W.; Yang, J.; Chen, Z.; Wang, Z.; Zhuan, S.; Xiang, X.; Feng, J.; et al. Preparation of peppermint oil nanoemulsions: Investigation of stability, antibacterial mechanism and apoptosis effects. Colloids Surf. B 2021, 201, 111626. [Google Scholar] [CrossRef] [PubMed]
  32. Chen, F.; Miao, X.; Lin, Z.; Xiu, Y.; Shi, L.; Zhang, Q.; Liang, D.; Lin, S.; He, B. Disruption of metabolic function and redox homeostasis as antibacterial mechanism of Lindera glauca fruit essential oil against Shigella flexneri. Food Control 2021, 130, 108282. [Google Scholar] [CrossRef]
  33. Zhou, C.; Li, C.; Siva, S.; Cui, H.; Lin, L. Chemical composition, antibacterial activity and study of the interaction mechanisms of the main compounds present in the Alpinia galanga rhizomes essential oil. Ind. Crops Prod. 2021, 165, 113441. [Google Scholar] [CrossRef]
  34. Barbosa, L.N.; Alves, F.C.B.; Andrade, B.F.M.T.; Albano, M.; Rall, V.L.M.; Fernandes, A.A.H.; Buzalaf, M.A.R.; Leite, A.L.; Pontes, L.G.; Santos, L.D.; et al. Proteomic analysis and antibacterial resistance mechanisms of Salmonella Enteritidis submitted to the inhibitory effect of Origanum vulgare essential oil, thymol and carvacrol. J. Proteom. 2020, 214, 103625. [Google Scholar] [CrossRef]
  35. Yang, Z.; He, Q.; Ismail, B.; Hu, Y.; Guo, M. Ultrasonication induced nano-emulsification of thyme essential oil: Optimization and antibacterial mechanism against Escherichia coli. Food Control 2022, 133, 108609. [Google Scholar] [CrossRef]
  36. Dai, J.; Li, C.; Cui, H.; Lin, L. Unraveling the anti-bacterial mechanism of Litsea cubeba essential oil against E. coli O157:H7 and its application in vegetable juices. Int. J. Food Microbiol. 2021, 338, 108989. [Google Scholar] [CrossRef]
  37. Cui, H.; Zhang, C.; Li, C.; Lin, L. Antibacterial mechanism of oregano essential oil. Ind. Crops Prod. 2019, 139, 111498. [Google Scholar] [CrossRef]
  38. Hu, W.; Li, C.; Dai, J.; Cui, H.; Lin, L. Antibacterial activity and mechanism of Litsea cubeba essential oil against methicillin-resistant Staphylococcus aureus (MRSA). Ind. Crops Prod. 2019, 130, 34–41. [Google Scholar] [CrossRef]
  39. Huang, J.; Qian, C.; Xu, H.; Huang, Y. Antibacterial activity of Artemisia asiatica essential oil against some common respiratory infection causing bacterial strains and its mechanism of action in Haemophilus influenzae. Microb. Pathog. 2018, 114, 470–475. [Google Scholar] [CrossRef]
  40. Meng, X.; Li, D.; Zhou, D.; Wang, D.; Liu, Q.; Fang, S. Chemical composition, antibacterial activity and related mechanism of the essential oil from the leaves of Juniperus rigida Sieb. et Zucc against Klebsiella pneumoniae. J. Ethnopharmacol. 2016, 194, 698–705. [Google Scholar] [CrossRef]
  41. Wang, H.; Ma, Y.; Liu, L.; Liu, Y.; Niu, X. Incorporation of clove essential oil nanoemulsion in chitosan coating to control Burkholderia gladioli and improve postharvest quality of fresh Tremella fuciformis. LWT 2022, 1770, 114059. [Google Scholar] [CrossRef]
  42. Zhan, J.; He, F.; Cai, H.; Wu, M.; Xiao, Y.; Xiang, F.; Yang, Y.; Ye, C.; Wang, S.; Li, S. Composition and antifungal mechanism of essential oil from Chrysanthemum morifolium cv. Fubaiju. J. Funct. Foods 2021, 87, 104746. [Google Scholar] [CrossRef]
  43. Zhang, R.; Cui, Y.; Cheng, M.; Guo, Y.; Wang, X.; Wang, J. Antifungal activity and mechanism of cinnamon essential oil loaded into mesoporous silica nanoparticles. Ind. Crops Prod. 2021, 171, 113846. [Google Scholar] [CrossRef]
  44. Hu, Z.; Yuan, K.; Zhou, Q.; Lu, C.; Du, L.; Liu, F. Mechanism of antifungal activity of Perilla frutescens essential oil against Aspergillus flavus by transcriptomic analysis. Food Control 2021, 123, 107703. [Google Scholar] [CrossRef]
  45. Kumar, A.; Singh, P.P.; Prakash, B. Unravelling the antifungal and anti-aflatoxin B1 mechanism of chitosan nanocomposite incorporated with Foeniculum vulgare essential oil. Carbohyd. Polym. 2020, 236, 116050. [Google Scholar] [CrossRef] [PubMed]
  46. Krishnamoorthy, R.; Gassem, M.A.; Athinarayanan, J.; Periyasamy, V.S.; Prasad, S.; Alshatwi, A.A. Antifungal activity of nanoemulsion from Cleome viscosa essential oil against food-borne pathogenic Candida albicans. Saudi J. Biol. Sci. 2021, 28, 286–293. [Google Scholar] [CrossRef] [PubMed]
  47. Bhattacharya, R.; Sourirajan, A.; Sharma, P.; Kumar, A.; Upadhyay, N.K.; Shukla, R.K.; Dev, K.; Krishnakuma, B.; Singh, M.; Bose, D. Bioenhancer potential of Aegle marmelos (L.) Corrêa essential oil with antifungal drugs and its mode of action against Candida albicans. Biocatal. Agric. Biotechnol. 2023, 48, 102647. [Google Scholar] [CrossRef]
  48. Hendges, C.; Stangarlin, J.R.; Zamban, V.C.; Mascaro, M.H.N.; Carmelo, D.B. Antifungal activity and control of the early blight in tomato through tea tree essential oil. Crop Prot. 2021, 148, 105728. [Google Scholar] [CrossRef]
  49. Zhang, Y.; Dai, J.; Ma, X.; Jia, C.; Han, J.; Song, C.; Liu, Y.; Wei, D.; Xu, H.; Qin, J.; et al. Nano-emulsification essential oil of Monarda didyma L. to improve its preservation effect on postharvest blueberry. Food Chem. 2023, 417, 135880. [Google Scholar] [CrossRef]
  50. Chen, X.; Lu, L.X.; Qiu, X.L.; Tang, Y.L. Advances in Mechanism Research on Controlled Release Antimicrobial Food Packaging Films. J. Food Sci. Biotechnol. 2020, 39, 1–7. [Google Scholar]
  51. Khounvilay, K.; Estevinho, B.N.; Sittikijyothin, W. Citronella oil microencapsulated in carboxymethylated tamarind gum and its controlled release. Eng. J. 2019, 23, 217–227. [Google Scholar] [CrossRef]
  52. Dima, C.; Pătraşcu, L.; Cantaragiu, A.; Alexe, P.; Dima, Ş. The kinetics of the swelling process and the release mechanisms of Coriandrum sativum L. essential oil from chitosan/alginate/inulin microcapsules. Food Chem. 2016, 195, 39–48. [Google Scholar] [CrossRef]
  53. Campelo-Felix, P.H.; Souza, H.J.B.; Figueiredo, J.A.; Fernandes, R.V.B.; Botrel, D.A.; Oliveira, C.R.; Yoshida, M.I.; Borges, S.V. Prebiotic Carbohydrates: Effect on Reconstitution, Storage, Release, and Antioxidant Properties of Lime Essential Oil Microparticles. J. Agric. Food Chem. 2017, 65, 445–453. [Google Scholar] [CrossRef]
  54. Bajac, J.; Nikolovski, B.; Lončarević, I.; Petrović, J.; Bajac, B.; Đurović, S.; Petrović, L. Microencapsulation of juniper berry essential oil (Juniperus communis L.) by spray drying: Microcapsule characterization and release kinetics of the oil. Food Hydrocoll. 2022, 125, 107430. [Google Scholar] [CrossRef]
  55. Zhu, Z.; Hu, J.; Zhong, Z. Preparation and characterization of long-term antibacterial and pH-responsive Polylactic acid/Octenyl succinic anhydride-chitosan @ tea tree oil microcapsules. Int. J. Biol. Macromol. 2022, 220, 1318–1328. [Google Scholar] [CrossRef] [PubMed]
  56. Singh, N.; Sheikh, J. Novel Chitosan-Gelatin microcapsules containing rosemary essential oil for the preparation of bioactive and protective linen. Ind. Crops Prod. 2022, 178, 114549. [Google Scholar] [CrossRef]
  57. Xu, L.; Xu, X.; Xu, Y.; Huang, M.; Li, Y. Fabrication and immediate release characterization of UV responded oregano essential oil loaded microcapsules by chitosan-decorated titanium dioxide. Food Chem. 2023, 400, 133965. [Google Scholar] [CrossRef]
  58. Gao, M.; Ji, M.; He, Y.; Pan, X.; Wang, X.; Si, T.; Sun, Y. Construction of consumer-friendly essential oil microcapsules with viscous cores to provide extra long-lasting release. Powder Technol. 2023, 413, 118040. [Google Scholar] [CrossRef]
  59. Noghabi, M.S.; Molaveisi, M.; Dehnad, D. Development of Persian gum-based microcapsules to speed up the release of cinnamon essential oil in the simulated saliva conditions. LWT Food Sci. Technol. 2023, 183, 114802. [Google Scholar] [CrossRef]
  60. Zhao, B.; Ni, Y.; Chen, K.; Lin, Z.; Jia, Z.; Qiu, H. Double-shell lignin microcapsules were prepared by one-step method for fabric coatings with UV resistance and durable antibacterial activity. Prog. Org. Coat. 2023, 179, 107518. [Google Scholar] [CrossRef]
  61. Zhao, Y.; Li, H.; Wang, Y.; Zhang, Z.; Wang, Q. Preparation, characterization and release kinetics of a multilayer encapsulated Perilla frutescens L. essential oil hydrogel bead. Int. J. Biol. Macromol. 2023, 124776. [Google Scholar] [CrossRef]
  62. Peppas, N.A.; Sahlin, J.J.A. simple equation for the description of solute release. III. Coupling of diffusion and relaxation. Int. J. Pharm. 1989, 57, 169–172. [Google Scholar] [CrossRef]
  63. Heydari, M.K.; Assadpour, E.; Jafari, S.M.; Javadian, H. Encapsulation of rose essential oil using whey protein concentrate-pectin nanocomplexes: Optimization of the effective parameters. Food Chem. 2021, 356, 129731. [Google Scholar] [CrossRef]
  64. Qiu, L.; Zhang, M.; Adhikari, B.; Chang, L. Microencapsulation of rose essential oil in mung bean protein isolate-apricot peel pectin complex coacervates and characterization of microcapsules. Food Hydrocoll. 2022, 124, 107366. [Google Scholar] [CrossRef]
  65. Das, S.; Singh, V.K.; Dwivedy, A.K.; Chaudhari, A.K.; Upadhyay, N.; Singh, P.; Sharam, S.; Dubey, N.K. Encapsulation in chitosan-based nanomatrix as an efficient green technology to boost the antimicrobial, antioxidant and in situ efficacy of Coriandrum sativum essential oil. Int. J. Biol. Macromol. 2019, 133, 294–305. [Google Scholar] [CrossRef]
  66. Hasheminejad, N.; Khodaiyan, F. The effect of clove essential oil loaded chitosan nanoparticles on the shelf life and quality of pomegranate arils. Food Chem. 2020, 309, 125520. [Google Scholar] [CrossRef]
  67. Zhang, W.; Shu, C.; Chen, Q.; Cao, J.; Jiang, W. The multilayer film system improved the release and retention properties of cinnamon EOs and its application as coating in inhibition to penicillium expansion of apple fruit. Food Chem. 2019, 299, 125109. [Google Scholar] [CrossRef]
  68. Chang, Y.; Choi, I.; Cho, A.R.; Han, J. Reduction of Dickeya chrysanthemi on fresh-cut iceberg lettuce using antimicrobial sachet containing microencapsulated oregano essential oil. LWT Food Sci. Technol. 2017, 82, 361–368. [Google Scholar] [CrossRef]
  69. Copado, C.N.; Julio, L.M.; Diehl, B.W.K.; Ixtaina, V.Y.; Tomás, M.C. Multilayer microencapsulation of chia seed oil by spray-drying using electrostatic deposition technology. LWT 2021, 152, 112206. [Google Scholar] [CrossRef]
  70. Yang, L.; Guo, Z.; Li, W.; Gou, Q.; Han, L.; Yu, Q. The impact of lemon seeds oil microcapsules based on a bilayer macromolecule carrier on the storage of the beef jerky. Food Packag. Shelf Life 2022, 32, 100838. [Google Scholar] [CrossRef]
  71. Muhoza, B.; Xia, S.; Wang, X.; Zhang, X.; Li, Y.; Zhang, S. Microencapsulation of essential oils by complex coacervation method: Preparation, thermal stability, release properties and applications. Crit. Rev. Food Sci. Nutr. 2022, 62, 1363–1382. [Google Scholar] [CrossRef] [PubMed]
  72. Hou, K.; Xu, Y.; Cen, K.; Gao, C.; Feng, X.; Tang, X. Nanoemulsion of cinnamon essential oil Co-emulsi ed with hydroxypropyl-β-cyclodextrin and Tween-80: Antibacterial activity, stability and slow release performance. Food Biosci. 2021, 43, 101232. [Google Scholar] [CrossRef]
  73. Sun, Y.; Zhang, M.; Bhandari, B.; Bai, B. Fennel essential oil loaded porous starch-based microencapsulation as an efficient delivery system for the quality improvement of ground pork. Int. J. Biol. Macromol. 2021, 172, 464–474. [Google Scholar] [CrossRef]
  74. Ozdemir, N.; Bayrak, A.; Tat, T.; Altay, F.; Kiralan, M.; Kurt, A. Microencapsulation of basil essential oil: Utilization of gum arabic/whey protein isolate/maltodextrin combinations for encapsulation efficiency and in vitro release. J. Food Meas. Charact. 2021, 15, 1865–1876. [Google Scholar] [CrossRef]
  75. Zhang, L.; Zhang, M.; Ju, R.; Bhandari, B.; Liu, K. Antibacterial mechanisms of star anise essential oil microcapsules encapsulated by rice protein-depolymerized pectin electrostatic complexation and its application in crab meatballs. Int. J. Food Microbiol. 2023, 384, 109963. [Google Scholar] [CrossRef]
  76. Jiménez-Martín, E.; Gharsallaoui, A.; Pérez-Palacios, T.; Carrascal, J.R.; Rojas, T.A. Suitability of using monolayered and multilayered emulsions for microencapsulation of ω-3 fatty acids by spray drying: Effect of Storage at Different Temperatures. Food Bioprocess Technol. 2015, 8, 100–111. [Google Scholar] [CrossRef]
  77. Lu, L.X.; Xu, J.; Pan, L. Research progress on stimulus-responsive microcapsules. J. Food Sci. Biotechnol. 2021, 40, 8–17. [Google Scholar]
  78. Niu, Y.; Wu, J.; Kang, Y.; Zhao, Q.; Xiao, Z.; Zhao, D. Encapsulation technique and application progress of mechanical stimuli-responsive microcapsules. Prog. Org. Coat. 2023, 176, 107390. [Google Scholar] [CrossRef]
  79. Gao, Y.; Ma, Q.; Cao, J.; Shi, Y.; Wang, J.; Ma, H.; Sun, Y.; Song, Y. Bifunctional alginate/chitosan stabilized perfluorohexane nanodroplets as smart vehicles for ultrasound and pH responsive delivery of anticancer agents. Int. J. Biol. Macromol. 2021, 191, 1068–1078. [Google Scholar] [CrossRef]
  80. Coimbra, A.; Ferreira, S.; Duarte, A.P. Biological properties of Thymus zygis essential oil with emphasis on antimicrobial activity and food application. Food Chem. 2022, 393, 133370. [Google Scholar] [CrossRef] [PubMed]
  81. Zhang, Y.; Li, D.; Lv, J.; Li, Q.; Kong, C.; Luo, Y. Effect of cinnamon essential oil on bacterial diversity and shelf-life in vacuum-packaged common carp (Cyprinus carpio) during refrigerated storage. Int. J. Food Microbiol. 2017, 249, 1–8. [Google Scholar] [CrossRef] [PubMed]
  82. Cai, M.; Zhang, G.; Wang, J.; Li, C.; Cui, H.; Lin, L. Application of glycyrrhiza polysaccharide nanofibers loaded with tea tree essential oil/gliadin nanoparticles in meat preservation. Food Biosci. 2021, 43, 101270. [Google Scholar] [CrossRef]
  83. Zhang, W.; Jiang, H.; Rhim, J.; Cao, J.; Jiang, W. Effective strategies of sustained release and retention enhancement of EOs in active food packaging films/coatings. Food Chem. 2022, 367, 130671. [Google Scholar] [CrossRef] [PubMed]
  84. Shao, P.; Yu, J.; Chen, H.; Gao, H. Development of microcapsule bioactive paper loaded with cinnamon essential oil to improve the quality of edible fungi. Food Packag. Shelf Life 2021, 27, 100617. [Google Scholar] [CrossRef]
  85. Li, S.; Jiang, Y.; Wang, M.; Li, R.; Dai, J.; Yan, J.; Qin, W.; Liu, Y. 3D printing of essential oil/β-cyclodextrin/popping candy modified atmosphere packaging for strawberry preservation. Carbohyd. Polym. 2022, 297, 120037. [Google Scholar] [CrossRef]
  86. Tian, Y.; Zhou, L.; Liu, J.; Yu, K.; Yu, W.; Jiang, H.; Zhong, J.; Zou, L.; Liu, W. Effect of sustained-release tea tree essential oil solid preservative on fresh-cut pineapple storage quality in modified atmospheres packaging. Food Chem. 2023, 417, 135898. [Google Scholar] [CrossRef]
  87. Wang, W.; Zhang, W.; Li, L.; Deng, W.; Liu, M.; Hu, J. Biodegradable starch-based packaging films incorporated with polyurethane-encapsulated essential-oil microcapsules for sustained food preservation. Int. J. Biol. Macromol. 2023, 235, 123889. [Google Scholar] [CrossRef]
  88. Chatha, S.A.S.; Asgher, M.; Asgher, R.; Hussain, A.I.; Iqbal, Y.; Hussain, S.M.; Bilal, M.; Saleem, F.; Iqbal, H.M.N. Environmentally responsive and anti-bugs textile nishes-Recent trends, challenges, and future perspectives. Sci. Total Environ. 2019, 690, 667–682. [Google Scholar] [CrossRef]
  89. Yinggam, B.; Kacha, W.; Rungseevijitprapa, W.; Sudta, P.; Prasitpuriprecha, C.; Brantner, A. Response surface optimization of spray-dried citronella oil microcapsules with reduced volatility and irritation for cosmetic textile uses. Powder Technol. 2019, 355, 372–385. [Google Scholar] [CrossRef]
  90. Specos, M.M.M.; García, J.J.; Tornesello, J.; Marino, P.; Vecchia, M.D.; Tesoriero, M.V.D.; Hermida, L.G. Microencapsulated citronella oil for mosquito repellent finishing of cotton textiles. Trans. R. Soc. Trop. Med. Hyg. 2010, 104, 653–658. [Google Scholar] [CrossRef]
  91. Liu, J.; Liu, C.; Liu, Y.; Chen, M.; Hu, Y.; Yang, Z. Study on the grafting of chitosan–gelatin microcapsules onto cotton fabrics and its antibacterial effect. Colloids Surf. B 2013, 109, 103–108. [Google Scholar] [CrossRef]
  92. Chen, K.; Xu, C.; Zhou, J.; Zhao, R.; Gao, Q.; Wang, C. Multifunctional fabric coatings with slow-releasing fragrance and UV resistant properties from ethyl cellulose/silica hybrid microcapsules. Carbohydr. Polym. 2020, 232, 115821. [Google Scholar] [CrossRef]
  93. Hammoud, Z.; Abada, M.B.; Greige-Gerges, H.; Elaissari, A.; Jemâa, J.M.B. Insecticidal effects of natural products in free and encapsulated forms: An overview. J. Nat. Pestic. Res. 2022, 1, 100007. [Google Scholar] [CrossRef]
  94. López, A.; Castro, S.; Andina, M.J.; Ures, X.; Munguía, B.; Llabot, J.M.; Elder, H.; Dellacassa, E.; Palma, S.; Domínguez, L. Insecticidal activity of microencapsulated Schinus molle essential oil. Ind. Crops Prod. 2014, 53, 209–216. [Google Scholar] [CrossRef]
  95. Ahsaei, S.M.; Rodríguez-Rojo, S.; Salgado, M.; Cocero, M.J.; Talebi-Jahromi, K.; Amoabediny, G. Insecticidal activity of spray dried microencapsulated essential oils of Rosmarinus officinalis and Zataria multiflora against Tribolium confusum. Crop Prot. 2020, 128, 104996. [Google Scholar] [CrossRef]
  96. Lv, H.; Huo, S.; Zhao, L.; Zhang, H.; Liu, Y.; Liu, S.; Tani, A.; Wang, R. Preparation and application of cinnamon-Litsea cubeba compound essential oil microcapsules for peanut kernel postharvest storage. Food Chem. 2023, 415, 135734. [Google Scholar] [CrossRef]
  97. Kim, J.; Jang, M.; Shin, E.; Kim, J.; Lee, S.H.; Park, C.G. Fumigant and contact toxicity of 22 wooden essential oils and their major components against Drosophila suzukii (Diptera: Drosophilidae). Pestic. Biochem. Physiol. 2016, 33, 35–43. [Google Scholar] [CrossRef] [PubMed]
  98. Ngamekaue, N.; Chitprasert, P. Effects of beeswax-carboxymethyl cellulose composite coating on shelf-life stability and intestinal delivery of holy basil essential oil-loaded gelatin microcapsules. Int. J. Biol. Macromol. 2019, 135, 1088–1097. [Google Scholar] [CrossRef] [PubMed]
  99. Thuekeaw, S.; Angkanaporn, K.; Chirachanchai, S.; Nuengjamnong, C. Dual pH responsive via double-layered microencapsulation for controlled release of active ingredients in simulated gastrointestinal tract: A model case of chitosan-alginate microcapsules containing basil oil (Ocimum basilicum Linn.). Polym. Degrad. Stab. 2021, 191, 109660. [Google Scholar] [CrossRef]
Figure 1. Application of EOs microcapsules in food, textiles, agriculture, and medical fields.
Figure 1. Application of EOs microcapsules in food, textiles, agriculture, and medical fields.
Molecules 28 04979 g001
Table 1. The inhibition mechanism of different essential oils (EOs) against various bacteria.
Table 1. The inhibition mechanism of different essential oils (EOs) against various bacteria.
EOsBacteriaAntibacterial Mechanism
Clove EOs [26]Listeria monocytogenes
(1)
Biomacromolecules: change DNA structure [26];
(2)
Cell wall/membrane integrity: destroy the integrity of the cell membrane, leading to the efflux of biomacromolecules [26].
Clove EOs [26]
Eugenia stipitata
EOs [27]
Thyme EOs [28]
Chuzhou chrysanthemum EOs [29]
Dodartia orientalis L. EOs [30]
Peppermint EOs [31]
Staphylococcus aureus
(1)
Biomacromolecules: interact with DNA and affects the accessory gene regulator (Agr) system, resulting in decreased expression of virulence genes [26];
(2)
Cell wall/membrane integrity: Disruption of cell membrane structure leading to the extravasation of ions (K+), nucleic acids, and proteins [27,29,30]; involves membrane potential depolarization and fluidity reduction possibly related to changes in fatty acid composition [28]; causes the destruction of structures and the function of the cell wall [30];
(3)
Metabolic pathway: inhibits the respiratory metabolism by inhibiting the tricarboxylic acid cycle pathway [26]; affects Embden–Meyerhof–Parnas pathway and decreases key enzyme activity (PFK, HK and PK) [29]; improves the reactive oxygen species (ROS) and malondialdehyde (MDA) level [31].
Lindera glauca fruit EOs [32]Shigella
flexneri
(1)
Cell wall/membrane integrity: disruption of cell membrane structure leading to the extravasation of ions (K+), nucleic acids, and proteins [32];
(2)
Metabolic pathway: induces H2O2 accumulation and lipid peroxidation (ROS and MDA); inhibits respiratory metabolism and gives rise to a disturbance of redox homeostasis [32].
Chuzhou chrysanthemum EOs [29]
Dodartia orientalis L. EOs [30]
Peppermint EOs [31]
Alpinia galanga rhizomes EOs [33]
Thyme EOs [34,35]
Litsea cubeba EOs [36]
Escherichia
coli
(1)
Biomacromolecules: disrupt DNA replication [36];
(2)
Cell wall/membrane integrity: disruption of cell membrane structure leading to the extravasation of ions (K+), nucleic acids and proteins [28,30,33,35,36]; involving membrane potential depolarization and fluidity reduction possibly related to the changes of fatty acid composition [28]; causes the destruction of structure and function of cell wall [30];
(3)
Metabolic pathway: affects hexose monophophate (HMP) pathway and decreases key enzyme activity (G6PHD) [29]; improves reactive oxygen species (ROS) and malondialdehyde (MDA) levels [31]; inhibits respiratory metabolism [36];
(4)
Virulence gene expression: inhibits enzymatic activity (P-type ATPases) and down-regulates the expression of four virulence genes (stx1, stx2, ehxA, eae) [33,36].
Dodartia orientalis L. EOs [30]
Origanum vulgare EOs [34]
Salmonella Enteritidis
(1)
Cell wall/membrane integrity: disruption of cell membrane structure leading to the extravasation of ions (K+), nucleic acids, and proteins; causes the destruction of structure and function of cell wall [30];
(2)
Biomacromolecules/metabolic pathway: DNA inhibition; key protein expression changes; bacterial oxidative stress; cellular metabolic imbalance [34].
Oregano EOs [37]
Litsea cubeba EOs [38]
Methicillin-resistant Staphylococcus aureus
(1)
Biomacromolecules: disrupts DNA replication by reacting with DNA in the form of chimera [38];
(2)
Cell wall/membrane integrity: disruption of cell membrane structure leading to the extravasation of ions (K+), nucleic acids, and proteins [37,38];
(3)
Metabolic level: inhibits respiratory metabolism by inhibiting the tricarboxylic acid cycle pathway and key enzymes [37]; affects HMP pathway and decreases key enzyme activity (G6PHD) [38];
(4)
Gene expression: inhibition of relative expression of pvl gene [37,38].
Artemisia asiatica EOs [39]Haemophilus influenzae
(1)
Cell wall/membrane integrity: leads to cell wall damage, cell deformation, and cell shrinkage [39];
Juniperus rigida EOs [40]Klebsiella pneumoniae
(1)
Cell wall/membrane integrity: causes irreversible damage to the cell wall and membrane, leading to the leakage of proteins and DNA/RNA. [40];
Clove EOs [41]Burkholderia gladioli
(1)
Cell wall/membrane integrity: damages cell membranes, resulting in leakage of bacterial cytoplasm [41];
(2)
Toxin expression: reduces the expression of bongkrekic acid [41];
Table 2. The inhibition mechanism of different EOs against various fungus.
Table 2. The inhibition mechanism of different EOs against various fungus.
EOsFungusAntifungal Mechanism
Chrysanthemum morifolium cv. Fubaiju EOs [42]C. albicans;
C. glabrata;
C. tropicalis;
S. cerevisiae;
D. hansenii;
Z. parabailii
(1)
Biomacromolecules: mitochondrial and DNA damage [42];
(2)
Cell wall/membrane integrity: plasma membrane disruption [42].
Cinnamon EOs [43]Mucor sp. FJ09;
Mucor circinelloides CNRMA 03.0371
(1)
Spore/mycelial: destroys the mycelial morphology [43];
(2)
Cell wall/membrane integrity: increases membrane permeability, leading to the leakage of cellular components [43].
(3)
Metabolic level: increases reactive oxygen species content and alters soluble sugar and malondialdehyde content [43];
(4)
Gene expression: upregulation expression of nox1 and nox2 genes [43];
Perilla frutescens EOs [44]
Foeniculum vulgare EOs [45]
Aspergillus flavus
Aflatoxin B1
(1)
Spore/mycelial: affects spore development [44];
(2)
Cell wall/membrane integrity: changes membrane permeability, leading to the leakage of biomacromolecules and ions [44,45]; changes mitochondrial membrane potential [45];
(3)
Metabolic level: inhibits ATPase activity and increases the generation of ROS [44,45]; affects amino acid metabolism [44]; alters ergosterol content, C-sources utilization, and Nor-1 gene expression [45].
Cleome viscosa EOs [46]
Aegle marmelos L. Corrêa EOs [47]
Candida albicans
(1)
Cell wall/membrane integrity: reduces the chitin levels and interferes with cell wall biosynthesis [46]; binds to ergosterol in the membrane and results in ion permeability increase [47].
Tea tree EOs [48]
Monarda didyma L. EOs [49]
Alternaria solani
(1)
Spore/mycelial: affects mycelial growth [48,49].
(2)
Metabolic level: increases the peroxidase and phenylalanine ammonialyase activities [48].
Monarda didyma L. EOs [49]Colletotrichum sp.
(1)
Spore/mycelial: affects mycelial growth [49].
Table 3. The release models and release mechanism of EOs microcapsules.
Table 3. The release models and release mechanism of EOs microcapsules.
Core MaterialsWall
Materials
Microcapsule MethodRelease ModelRelease Mechanism References
Coriander EOsChitosan/Chitosan-Alginate/Spray-dryingMt/M = ktnAnomalous diffusion[52]
Coriander EOsAlginate/Chitosan-InulinSpray-dryingMt/M = ktnDiffusion swelling[52]
Lime EOswhey proteinSpray-dryingMt/M = ktnFickian diffusion[53]
Lime EOswhey protein-inulinSpray-dryingMt/M = ktnAnomalous diffusion[53]
Lime EOswhey protein-oligofructoseSpray-dryingMt/M = ktnAnomalous diffusion[53]
Spearmint EOsInulin-Gum ArabicSpray-dryingMt/M = k1tm + k2t2mFickian diffusion[12]
Juniper berry EOsGum arabic/Gum arabic-Maltodextrin/Sodium AlginateSpray-dryingMt/M = k1tm + k2t2mFickian diffusion[54]
Tea tree EOsPolylactic acid/Octenyl succinic anhydride chitosan Double emulsion and solvent evaporation methodMt/M = k1tm + k2t2mFick diffusion and skeletal dissolution[55]
Rosemary EOsChitosan-GelatinSpray dryingMt/M = ktnFickian diffusion[56]
Oregano EOsChitosan-decorated Titanium DioxideIon-exchange-mediated self-assembly techniqueMt/M = ktnFickian diffusion[57]
Caprylic/capric triglycerideMelamine-formaldehydeCrosslinked methodMt/M = ktBurst release[58]
Caprylic/capric triglyceridePolystyreneCrosslinked methodMt/M = ktnDiffusion[58]
Cinnamon EOsPersian gum-maltodextrinSpray-dryingMt/M = k1tm + k2t2mFickian diffusion[59]
Cinnamon EOsPersian gum-maltodextrinSpray-dryingMt/M = ktnFickian diffusion[59]
Lemon EOsPolyurethane-ligninA joint method of interfacial polymerization with free radical copolymerizationMt/M = kt1/2Diffusion[60]
Perilla frutescens L. EOsStarch sodium octylsuccinate/sodium alginate/chitosanCoacervation methodsMt/M = k1tm + k2t2mFick diffusion[61]
Note: In the first-order (Mt/M = kt) and Korsmeyer–Peppas (Mt/M = ktn) models, Mt/M is the fraction of drugs released, k is the kinetic constant, t is the release time, and n is the diffusional exponent for drug release. In the Peppas–Sahlin model (Mt/M = k1tm + k2t2m), where the first term of the right-hand side is the Fickian contribution, the second term is the Case-II relaxational contribution. The coefficient m is the purely Fickian diffusion exponent for a device of any geometrical shape which exhibits controllable release, and k1 and k2 are diffusion and erosion constants, respectively [62].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhao, Y.; Wang, Y.; Zhang, Z.; Li, H. Advances in Controllable Release Essential Oil Microcapsules and Their Promising Applications. Molecules 2023, 28, 4979. https://doi.org/10.3390/molecules28134979

AMA Style

Zhao Y, Wang Y, Zhang Z, Li H. Advances in Controllable Release Essential Oil Microcapsules and Their Promising Applications. Molecules. 2023; 28(13):4979. https://doi.org/10.3390/molecules28134979

Chicago/Turabian Style

Zhao, Yana, Yanbo Wang, Zhijun Zhang, and Huizhen Li. 2023. "Advances in Controllable Release Essential Oil Microcapsules and Their Promising Applications" Molecules 28, no. 13: 4979. https://doi.org/10.3390/molecules28134979

Article Metrics

Back to TopTop